PT J AU Gao, X Honn, VK AF Gao, Xiang Honn, V Kenneth TI Recessive oncogenes: current status SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE oncogenes; cell cycle; heterozygosity; mutation ID oncogenes; cell cycle; heterozygosity; mutation AB Cell growth is under the control of a variety of positive and negative signals. An imbalance of such signals results in deregulation of cell behavior. Recessive oncogenes or tumor suppressor genes, opposite to dominant oncogenes, encode important cellular proteins which could function as negative regulators of the cell cycle, i.e., cell cycle brakes. Inactivation of recessive oncogenes, by allelic deletion, loss of expression, mutation, or functional inactivation by interacting with oncogene products of DNA tumor viruses or with amplified cellular binding proteins, will lead to uncontrolled cell growth or tumor formation. Besides the classic suppressor genes such as the p53 and RB, a growing number of novel tumor suppressor genes have been identified in recent years. While some tumor suppressor genes have been found to be important for the development of a large number of human malignancies (e.g., the p53 gene), others are more tumor type-specific (e.g., the NF-1 gene). Many human cancer types showed abnormalities of multiple tumor suppressor genes, offering strong support to the concept that tumorigenesis and progression result from an accumulation of multiple genetic alterations. In this review, we will begin with an overview (gene, transcript, protein and mechanisms of action) of the tumor suppressor genes (the RB, p53, DCC, APC, MCC, WT1, VHL, MST1, and BRCA1 genes) identified to date and then discuss the specific involvement of tumor suppressor genes in human malignancies including prostate cancer. Various chromosomal regions which potentially may contain tumor suppressor genes also will be reviewed. C1 [Gao, Xiang] Wayne State University, Department of Radiation Oncology, 432 Chemistry Building, MI48202 Detroit, USA. [Honn, V Kenneth] Wayne State University, Department of Radiation Oncology, 432 Chemistry Building, MI48202 Detroit, USA. RP Honn, VK (reprint author), Wayne State University, Department of Radiation Oncology, MI48202 Detroit, USA. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 7 EP 22 PG 16 ER PT J AU Nagy, P AF Nagy, Peter TI The Facultative Stem Cell: A New Star in Liver Pathology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE liver; stem cell; oval cell carcinogenesis ID liver; stem cell; oval cell carcinogenesis AB Although the unlimited capacity of hepatocytes to divide has been recently proven, more and more evidences support the existence of a primitive stem cell compartment in the liver. These cells probably do not participate in the usual maintenance of the liver mass, but they are activated in case of extensive hepatocyte injury. In vivo the oval cells show deep similarity to the primitive cells of the embryonic liver and seem to be the amplification compartment of the hepatic stem cells. A primitive epithelial cell population can be isolated from the normal liver and maintained in vitro. Studies of these two experimental systems provide most of the data about liver stem cells, which may become important for the clinical practice if we understand how their growth is regulated. C1 [Nagy, Peter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., 1085 Budapest, Hungary. RP Nagy, P (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 23 EP 26 PG 4 ER PT J AU Denton, G Price, RM AF Denton, Graeme Price, R Michael TI Immune Responses to the MUC1 Mucin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE MUC1 mucin; immunogenicity; peptide epitopes ID MUC1 mucin; immunogenicity; peptide epitopes AB MUC1 mucins are highly glycosylated glycoproteins expressed on the luminal surfaces of glandular epithelia. In breast and ovarian carcinomas, their expression is frequently upregulated and they may be secreted into the circulation of cancer patients. Early studies aimed at the production of anti-MUC1 monoclonal antibodies revealed that MUC1 was a potent immunogen in mice with many monoclonal antibodies raised defining epitopes within the protein core of MUC1. The immunogenicity of MUC1 has now been extended to human studies and it is apparent that patients with breast and ovarian malignant disease are able to mount immune responses against MUC1. These findings provide information on the mechanisms involved in the recognition of MUC1 expressing tumours. The utilisation of MUC1 related immunogens to stimulate immune responses to tumours could lead to the improved management of patients and the development of new immunotherapeutic strategies aimed at the eradication of MUC1 mucin expressing cancers. C1 [Denton, Graeme] University of Nottingham, School of Pharmaceutical Sciences, Cancer Research Laboratories, NG7 2RD Nottingham, UK. [Price, R Michael] University of Nottingham, School of Pharmaceutical Sciences, Cancer Research Laboratories, NG7 2RD Nottingham, UK. RP Denton, G (reprint author), University of Nottingham, School of Pharmaceutical Sciences, Cancer Research Laboratories, NG7 2RD Nottingham, UK. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 27 EP 31 PG 5 ER PT J AU Wang, YM Fodstad, O Wolf-Dieter, L Bengtsson, M Totterman, Th Funderud, S Martin, H Kvalheim, G AF Wang, Y Meng Fodstad, Oystein Wolf-Dieter, Ludwig Bengtsson, Mats Totterman, Thomas Funderud, Steinar Martin, Hans Kvalheim, Gunnar TI An Effective, Direct Immunomagnetic Procedure for Purging Acute Lymphoblastic Leukemia Cells from Human Bone Marrow SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bone marrow; leukemia; transplantation; purging ID bone marrow; leukemia; transplantation; purging AB The AB4 monoclonal antibody, which recognizes an HLA-DR epitope, was found to bind to a high percentage of malignant blast cells in samples obtained from 27 patients with ALL. These included 11 of 11 cases with c-ALL, 3 of 7 with pre-pre-B, and 8 of 9 cases with pre-B ALL. AB4 was used together with anti CD10 and anti CD19 antibodies and super-paramagnetic particles for developing a direct immunomagnetic procedure for purging human bone marrow of leukemic cells. In model experiments with KM3 cells admixed to mononuclear bone marrow cells, the individual antibodies each removed 2.8-3.1 logs and 3.6-4.1 logs of tumor cells with one and two purging cycles, respectively. In comparison, the efficacy of a mixture of the three antibodies was 4.4 logs with one treatment cycle, and > 5 logs with repeated treatments. Whereas the use of a commercially available anti-HLA-DR antibody resulted in a 90% reduction in the survival of CFU-GMs and normal blast colonies, AB4 had only a moderate effect on the progenitor cells (46% and 30% reduction). In conjunction with autologous transplantation, bone marrow from a patient was purged with the antibody mixture and 50% of the CFU-GMs and 47% of the CD34+ cells remained after treatment. The patient showed a normal engraftment, reaching a level of 0.5 x 109/l neutrophils by day 20 and 20 x 109/l platelets by day 30. It is concluded that the antibody cocktail may safely and effectively be used for rapid autograft purging in patients with c-ALL, and also in phenotypically selected cases with other subtypes of ALL. C1 [Wang, Y Meng] The Norwegian Radium Hospital, Department of Tumor Biology, N-0310 Oslo, Montebello, Norway. [Fodstad, Oystein] The Norwegian Radium Hospital, Department of Tumor Biology, N-0310 Oslo, Montebello, Norway. [Wolf-Dieter, Ludwig] Freie Universitat, Immunologisches ZellmarkerlaborBerlin, Germany. [Bengtsson, Mats] University Hospital, Department of ImmunologyUppsala, Sweden. [Totterman, Thomas] University Hospital, Department of ImmunologyUppsala, Sweden. [Funderud, Steinar] Norwegian Radium Hospital, Department of ImmunologyOslo, Montebello, Norway. [Martin, Hans] University Hospital of FrankfurtFrankfurt, Germany. [Kvalheim, Gunnar] The Norwegian Radium Hospital, Department of Medical Oncology and RadiotherapyOslo, Montebello, Norway. RP Wang, YM (reprint author), The Norwegian Radium Hospital, Department of Tumor Biology, N-0310 Oslo, Norway. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 32 EP 37 PG 6 ER PT J AU Szende, B Tyihak, E Szokan, Gy Katay, Gy AF Szende, Bela Tyihak, Erno Szokan, Gyula Katay, Gyorgy TI Possible Role of Formaldehyde in the Apoptotic and Mitotic Effect of 1-Methyl-Ascorbigen SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE formaldehyde; apoptosis; methylascorbigen ID formaldehyde; apoptosis; methylascorbigen AB PC-3 human prostate carcinoma cells were treated with 100 mg/ml 1-methyl-ascorbigen (Me-Asc). This treatment resulted in a significant decrease in tumor cell number in parallel with an increase in apoptotic cells. The formaldehyde (HCHO) level in the culture medium was also increased. Dimedone (Di), a known capture molecule forming formal-demethone with HCHO, applied simultaneously with Me-Asc in 10 mg/ml doses diminished the apoptosis-inducing effect of Me-Asc. The possible role of in situ generated HCHO in the induction of apoptosis is discussed. C1 [Szende, Bela] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., 1085 Budapest, Hungary. [Tyihak, Erno] Hungarian Academy of Sciences, Plant Protection InstituteBudapest, Hungary. [Szokan, Gyula] Eotvos Lorand University, Faculty of Science, Institute of Chemistry, Department of Organic ChemistryBudapest, Hungary. [Katay, Gyorgy] Eotvos Lorand University, Faculty of Science, Institute of Chemistry, Department of Organic ChemistryBudapest, Hungary. RP Szende, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 38 EP 42 PG 5 ER PT J AU Sameni, M Elliott, E Ziegler, G Fortgens, HP Dennison, C Sloane, FB AF Sameni, Mansoureh Elliott, Edith Ziegler, Grace Fortgens, H Philip Dennison, Clive Sloane, F Bonnie TI Cathepsin B and D are Localized at the Surface of Human Breast Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE aspartic proteases; breast cancer; cathepsins; cysteine proteases; oncogenic ras ID aspartic proteases; breast cancer; cathepsins; cysteine proteases; oncogenic ras AB Alterations in trafficking of cathepsins B and D have been reported in human and animal tumors. In MCF10 human breast epithelial cells, altered trafficking of cathepsin B occurs during their progression from a preneoplastic to neoplastic state. We now show that this is also the case for altered trafficking of cathepsin D. Nevertheless, the two cathepsins are not necessarily trafficked to the same vesicles. Perinuclear vesicles of immortal MCF10A cells label for both cathepsins B and D, yet the peripheral vesicles found in ras-transfected MCF10AneoT cells label for cathepsin B, cathepsin D or both enzymes. Studies at the electron microscopic level confirm these findings and show in addition surface labeling for both enzymes in the transfected cells. By immunofluorescence staining, cathepsin B can be localized on the outer surface of the cells. Similar patterns of peripheral intracellular and surface staining for cathepsin B are seen in the human breast carcinoma lines MCF7 and BT20. We suggest that the altered trafficking of cathepsins B and D may be of functional significance in malignant progression of human breast epithelial cells. Translocation of vesicles containing cathepsins B and D toward the cell periphery occurs in human breast epithelial cells that are at the point of transition between the pre-neoplastic and neoplastic state and remains part of the malignant phenotype of breast carcinoma cells. C1 [Sameni, Mansoureh] Wayne State University, Department of Pharmacology, 540 E Canfield, 48201 Detroit, Michigan, USA. [Elliott, Edith] University of Natal, Department of BiochemistryPietermaritzburg, South Africa. [Ziegler, Grace] Wayne State University, Department of Pharmacology, 540 E Canfield, 48201 Detroit, Michigan, USA. [Fortgens, H Philip] University of Natal, Department of BiochemistryPietermaritzburg, South Africa. [Dennison, Clive] University of Natal, Department of BiochemistryPietermaritzburg, South Africa. [Sloane, F Bonnie] Wayne State University, Department of Pharmacology, 540 E Canfield, 48201 Detroit, Michigan, USA. RP Sloane, FB (reprint author), Wayne State University, Department of Pharmacology, 48201 Detroit, USA. EM bsloane@med.wayne.edu NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 43 EP 53 PG 11 ER PT J AU Schmidt, B Valay, M Nahajevszky, S Pitlik, E Fust, Gy AF Schmidt, Bela Valay, Marta Nahajevszky, Sarolta Pitlik, Ervin Fust, Gyorgy TI Complement Synthesis Influencing Factors Produced by Acute Myeloid Leukemia Blast Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE AML; complement factor B; C1-INH; C3; C4; IL-1b; IL-2; IL-6; TNF ID AML; complement factor B; C1-INH; C3; C4; IL-1b; IL-2; IL-6; TNF AB In a previous study, we found hypercomplementaemia in the sera of acute myeloid leukemia patients. In this study we show that the supernatants of mononuclear cells, derived from peripheral blood taken in the blastic phase, from patients with acute myeloid leukemia (CM-AML) increased the in vitro complement protein synthesis of HepG2 hepatocellular carcinoma cells. This effect of CM-AML was mediated by heat labile soluble factors and involved the synthesis of mRNA and protein. Inhibition experiments with anti-cytokine antibodies and immunoaffinity chromatography revealed that this effect of CM-AML is mostly mediated by IL-1 and IL-6. C1 [Schmidt, Bela] National Institute of Haematology, Blood Transfusion and Immunology, Daroczi ut 24., H-1502 Budapest, Hungary. [Valay, Marta] National Institute of Haematology, Blood Transfusion and Immunology, Daroczi ut 24., H-1502 Budapest, Hungary. [Nahajevszky, Sarolta] National Institute of Haematology, Blood Transfusion and Immunology, Daroczi ut 24., H-1502 Budapest, Hungary. [Pitlik, Ervin] National Institute of Haematology, Blood Transfusion and Immunology, Daroczi ut 24., H-1502 Budapest, Hungary. [Fust, Gyorgy] National Institute of Haematology, Blood Transfusion and Immunology, Daroczi ut 24., H-1502 Budapest, Hungary. RP Fust, Gy (reprint author), National Institute of Haematology, Blood Transfusion and Immunology, H-1502 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 54 EP 59 PG 6 ER PT J AU Divald, A Gyorgydeak, Z Timar, F Zalatnai, A Bognar, R Horvath, G Lapis, K Jeney, A AF Divald, Andras Gyorgydeak, Zoltan Timar, Ferenc Zalatnai, Attila Bognar, Rezso Horvath, Gabor Lapis, Karoly Jeney, Andras TI Modification of Acute and Chronic Liver Damage by Thiazolidine Compounds SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE thiazolidine; liver damage; cirrhosis; hepatoprotection ID thiazolidine; liver damage; cirrhosis; hepatoprotection AB As high sulfhydril levels were shown to reduce the action of agents causing tissueinjury, increasing glutathion concentrations may have cytoprotective potential. In this study the hepatoprotective effects of several derivatives of 4carboxy5,5dimethyl thiazolidine, a modulator of glutathion metabolism were studied in rat liver damaged with CCl4. It was found that 4(S) carboxy 5,5dimethyl2 (5'nitro2furyl) thiazolidine (dimethylthiazolidinenitrofuran: DTNF) had the most significant hepatoprotective action; therefore it was subjected to detailed investigation in various models for acute and chronic liver injury. This compound was shown to ameliorate allylalcohol induced liver injury in rats, galactosamine induced hepatitis of mice and CCl4 induced chronic liver damage in rats. Our study on protein synthesis in primary hepatocyte suspension culture showed that cell injury induced by CCl4 could be reduced in the presence of this thiazolidine compound. C1 [Divald, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Gyorgydeak, Zoltan] Kossuth Lajos University, Institute of Organic ChemistryDebrecen, Hungary. [Timar, Ferenc] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Bognar, Rezso] Kossuth Lajos University, Institute of Organic ChemistryDebrecen, Hungary. [Horvath, Gabor] Kossuth Lajos University, Institute of Organic ChemistryDebrecen, Hungary. [Lapis, Karoly] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Jeney, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Jeney, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 60 EP 63 PG 4 ER PT J AU Aszalos, A AF Aszalos, Adorjan TI Modulation of Multidrug Resistance in Cancer by Immunosuppresive Agents (Preclinical Studies) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE multidrug; resistance; cancer; immunosuppression ID multidrug; resistance; cancer; immunosuppression AB This is a brief summary of the status of known immunosuppressive drugs describing their potential and mode of action to reverse the function of the MDR1 gene product, the P glycoprotein. Different aspects of these immunosuppressors have been reviewed in the recent literature. This summary will focus only on those studies which relate to the effect of these drugs on the P-glycoprotein. In addition, studies which may explain the mode of action, but do not deal directly with P-glycoprotein, are also summarized. C1 [Aszalos, Adorjan] Food and Dung Administration, Department of Health, Education and Welfare, 200 C Street SW, 20204 Washington, USA. RP Aszalos, A (reprint author), Food and Dung Administration, Department of Health, Education and Welfare, 20204 Washington, USA. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 64 EP 70 PG 7 ER PT J AU Sapi, Z Szapanidisz, J Toth, B Bodrogi, I AF Sapi, Zoltan Szapanidisz, Jorgosz Toth, Bernadette Bodrogi, Istvan TI DNA Ploidy Pattern in Pure Seminomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DNA ploidy; image cytometry; seminoma ID DNA ploidy; image cytometry; seminoma AB Nuclear extract and image cytometry was used to determine the DNA ploidy pattern of 31 pure seminomas. At least 5-year but usually 10-year follow-up was available to compare the clinical outcome to the DNA ploidy pattern. In 24 cases (77.4%) the DNA indexes (DI) showed tetraploid (1,8 < DI < 2,2) pattern confirming the recent cytogenetic and flow cytometric DNA studies of others. However, in 7 cases (22,6%) the tumors were aneuploid. Out of these aneuploid cases 2 had two subpopulations and 2 had less than 1,8 DI. These latter cases (providing the loss of chromosomal DNA) had elevated AFP levels in serum that raises the question of nonseminomatous transformation without any morphological evidence. Usually, the aneuploid cases had worse prognoses but there was no significant difference. Because of the small number of aneuploid cases wider clinicopathologic studies are required to confirm our results. C1 [Sapi, Zoltan] St John's Hospital, Department of Pathology, Diosarok u. 1., H-1122 Budapest, Hungary. [Szapanidisz, Jorgosz] St. John's Hospital, Department of UrologyBudapest, Hungary. [Toth, Bernadette] St John's Hospital, Department of Pathology, Diosarok u. 1., H-1122 Budapest, Hungary. [Bodrogi, Istvan] National Institute of Oncology, Department of Chemotherapy and ClinicopharmacologyBudapest, Hungary. RP Sapi, Z (reprint author), St John's Hospital, Department of Pathology, H-1122 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 71 EP 74 PG 4 ER PT J AU Orosz, Zs Besznyak, I AF Orosz, Zsolt Besznyak, Istvan TI Diffuse Inflammatory Pseudotumor of the Testis, the Epididymis and the Spermatic Cord SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE inflammatory pseudotumor; plasma cell granuloma; testis; myofibroblast ID inflammatory pseudotumor; plasma cell granuloma; testis; myofibroblast AB Inflammatory pseudotumors have been recognized in many parts of the body. A case of a diffuse variant which involved the testis, the epididymis and the spermatic cord is described. The patient had enlarged left testis for several months. Clinically, the lesion mimicred cancer. Histologically, the lesion contained hyalinized fibrous tissue with spindle cells, plasma cells and lymphocytes. Gradual involvement of vascular channels by the cellular elements of inflammatory pseudotumor was observed. Results of immunohistochemical studies showed a myofibroblast differentiation in the majority of spindle cells: intense antibody staining for smooth muscle actin, muscle specific actin, and vimentin. The ultrastructural findings, intracytoplasmic filaments with dense bodies, were also consistent with the myofibroblastic nature of these cells. The histiocyte differentiation of spindle cells is questionable in our case, because only scattered histiocyte-like cells showed positivity with the KP-1 (CD-3) antibody. C1 [Orosz, Zsolt] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Besznyak, Istvan] National Institute of Oncology, Department of SurgeryBudapest, Hungary. RP Orosz, Zs (reprint author), National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 75 EP 79 PG 5 ER PT J AU Balogh, K Ferencz, T Csikos, A Timar, J AF Balogh, Karoly Ferencz, Tamas Csikos, Andras Timar, Jozsef TI Tumoral Calcinosis in Infancy (A light and electron microscopic study with X-ray microanalysis) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE tumoral calcinosis; infancy; microanalysis ID tumoral calcinosis; infancy; microanalysis AB The most frequent form of idiopathic calcinosis is tumoral calcinosis (TC) which rarely occurs at young ages. We describe here a TC case of a young boy with its light microscopy completed with electron microscopic examinations. X-ray microanalysis revealed in the intracellular crystals CaCl2 besides the previously described hydroxyapatite. The significance of this finding is unknown at the moment. C1 [Balogh, Karoly] Harvard Medical School, Beth Israel Deaconess Medical Center, Department of Pathology and Laboratory Medicine, 185 Pilgrim Rd., 02215 Boston, MA, USA. [Ferencz, Tamas] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Csikos, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Timar, Jozsef] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Balogh, K (reprint author), Harvard Medical School, Beth Israel Deaconess Medical Center, Department of Pathology and Laboratory Medicine, 02215 Boston, USA. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 80 EP 84 PG 5 ER PT J AU Timar, J Jeney, A Kovalszky, I Kopper, L AF Timar, Jozsef Jeney, Andras Kovalszky, Ilona Kopper, Laszlo TI Role of Proteoglycans in Tumor Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE proteoglycan; metastasis; glycosaminoglycan; invasion ID proteoglycan; metastasis; glycosaminoglycan; invasion AB Data is now starting to accumulate on the differential expression of PGs in tumor cells of various invasive/metastatic potential. This is not so surprising if one considers the key functions that PGs play in the regulation of cell proliferation, adhesion and motility. However, characterization of PG expression in individual tumor types still awaits further detailed studies. Data on melanomas clearly indicate that PG phenotype is both specific and also promiscuous in a sense that ectopic expression of certain tissue specific PGs can occur in various tumors. Expression of a metastatic phenotype-specific splice variants of CD44 provides an example for the possible marker-function of PG. This also raises the hope that some PGs could be used as diagnostic/prognostic tools in pathology or even as a therapeutic targets against tumor dissemination. On the other hand, specific glycanation inhibitors may also be used for the modulation of tumor PG exist and the invasive phenotype. C1 [Timar, Jozsef] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Jeney, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1995 VL 1 IS 1 BP 85 EP 93 PG 9 ER PT J AU Isaacson, GP AF Isaacson, G Peter TI Primary Gastric Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE MALT; gastric lymphoma ID MALT; gastric lymphoma AB The pathogenesis of gastric MALT lymphoma starts with accumulation of MALT following infection of the stomach by H. pylori. Rarely this lymphoid infiltrate contains cells with a growth advantage possibly due to a genetic change (trisomy 3?). The result is a monoclonal lymphoproliferative lesion which is responsive to H. pylori driven T-cell help. Because its growth is dependent on the presence of local antigen, gastric MALT lymphoma remains localized for long periods and it is during this phase that the lymphoma can be treated by eradication of H. pylori. Further genetic changes, as yet uncharacterized may lead to escape from T-cell dependency and ultimately high grade transformation. C1 [Isaacson, G Peter] University College London Medical School, Department of Histopathology, University Street, WC1 E 6JJ London, UK. RP Isaacson, GP (reprint author), University College London Medical School, Department of Histopathology, WC1 E 6JJ London, UK. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 1996 VL 2 IS 1 BP 5 EP 10 PG 6 ER PT J AU Strausz, J AF Strausz, Janos TI Lung Cancer: A Bronchoscopic Approach SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE lung cancer; bronchoscopy; diagnosis; therapy ID lung cancer; bronchoscopy; diagnosis; therapy AB The unfavorable epidemiological data of lung cancer has not been changed during the past ten years. The only possibility to cure this malignancy is surgical resection. The five year survival rate after surgery is highly dependent on early discovery of the tumor. Today, bronchoscopy plays a central role in the diagnosis, staging and therapy of lung cancer. The main indications of diagnostic bronchoscopy are the identification of the tumor and the determination of its extent. The aim of therapeutic bronchoscopy - laser photocoagulation, high dose rate afterloading irradiation and stent implantation - is to provide an acceptable quality of life and to manage symptoms such as bleeding, cough and dyspnea. C1 [Strausz, Janos] National Koranyi Institute for TB and Pulmonology, Department of Pulmonology, Piheno u. 1., H-1529 Budapest, Hungary. RP Strausz, J (reprint author), National Koranyi Institute for TB and Pulmonology, Department of Pulmonology, H-1529 Budapest, Hungary. EM str12196@helka.iif.hu NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 1996 VL 2 IS 1 BP 11 EP 15 PG 5 ER PT J AU Rot, A AF Rot, Antal TI Inflammatory and Physiological Roles of Chemokines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE IL-8; MCP-1; RANTES; inflammation; sweat; milk; leukocytes ID IL-8; MCP-1; RANTES; inflammation; sweat; milk; leukocytes AB Chemokines, members of the family of chemotactic peptides, have a well documented function in different inflammatory diseases where they induce leukocyte emigration into lesions. Several recent observations indicate that, in addition to pathological states, chemokines are also produced and secreted under physiological conditions by various exocrine glands in amounts sufficient for their full biological effect. The glands involved in chemokine production and secretion include eccrine sweat glands, lactating mammary glands, lacrimal and salivary glands. It is suggested that analogous to their role in inflammatory diseases, chemokines produced by the exocrine glands are responsible for the induction of homeostatic leukocyte migration into mucosal epithelia and skin and also, mammary glands and milk. In addition, the mechanism by which chemokines induce leukocyte homing under physiological circumstances is discussed. C1 [Rot, Antal] Sandoz Research Institute, Department of Dermatology, Experimental Molecular Pathology, Brunner Str. 59., A-1235 Vienna, Austria. RP Rot, A (reprint author), Sandoz Research Institute, Department of Dermatology, Experimental Molecular Pathology, A-1235 Vienna, Austria. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 1996 VL 2 IS 1 BP 16 EP 20 PG 5 ER PT J AU Tang, GD Diglio, AC Honn, K AF Tang, G Dean Diglio, A Clement Honn, Kenneth TI Tyrosine Phosphorylation of a ~30 kD Protein Precedes avb3 Integrin-signaled Endothelial Cell Spreading and Motility on Matrix Proteins SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE integrin; motility; spreading; tyrosin phosphorylation; extracellular matrix ID integrin; motility; spreading; tyrosin phosphorylation; extracellular matrix AB A microvascular endothelial cell line (CD clone 4) isolated from murine lung adheres to and spreads well on fibronectin, vitronectin, and fibrinogen, but poorly on collagen type IV and laminin. Ligating cell surface av, b3, a4, a5, or b1 integrin receptors with monospecific antibodies promoted a dramatic cell spreading and motility on vitronectin or collagen IV. Antibodies directed to other adhesion molecules, including aIIb, PECAM-1, and P-selectin were ineffective. Ligation with monoclonal anti-av or -b3, but not -a4, -a5, or -b1 antibodies, induced a rapid, and dose-dependent tyrosine phosphorylation of a ~30 kD protein, which preceded CD clone 4 endothelial cell spreading and motility and was partially inhibited by genistein and completely inhibited by BAPTA. All other antibodies tested did not induce the tyrosine phosphorylation of the 30 kD protein as well as cell spreading and motility. The present results suggest that b1 and b3 integrins employ different biochemical mechanisms in signaling endothelial cell spreading and motility and that the tyrosine phosphorylation of the 30 kD protein (and probably other proteins) may play an important role in signaling b3 integrin-mediated endothelial cell interaction with other cells (e.g., tumor cells) and extracellular matrix. C1 [Tang, G Dean] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Diglio, A Clement] Wayne State University, Department of PathologyDetroit, USA. [Honn, Kenneth] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. RP Honn, K (reprint author), Wayne State University, Department of Radiation Oncology, 48202 Detroit, USA. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 1996 VL 2 IS 1 BP 21 EP 29 PG 9 ER PT J AU McDonald, SJ Wilson, MK Gartside, P Sonke, LR Pavelic, L Okum, E Neanen, J Gluckman, LJ Pavelic, PZ AF McDonald, S John Wilson, M Keith Gartside, Peter Sonke, L Robert Pavelic, Ljiljana Okum, Eric Neanen, Julie Gluckman, L Jack Pavelic, P Zlatko TI Immunohistochemical Expression of N-ras Oncogene is a Late Event in Head and Neck Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE head and neck cancer; immunohistochemistry; N-ras ID head and neck cancer; immunohistochemistry; N-ras AB This study investigated the expression of the N-ras oncogene in routinely processed tissue sections from 133 patients with squamous cell carcinoma of the head and neck (SCCHN) by immunohistochemistry using anti-N-ras monoclonal antibody. N-ras expression was present in 67 of 133 (49.6%) cases. There was a highly significant correlation between N-ras expression and clinical stage of disease (P=0.003). This study confirmed that overexpression of the N-ras oncogene is common in SCCHN and that it may be an important event in the late stage of disease. C1 [McDonald, S John] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45167-0528 Cincinnati, Ohio, USA. [Wilson, M Keith] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45167-0528 Cincinnati, Ohio, USA. [Gartside, Peter] University of Cincinnati College of Medicine, Department of Environmental HealthCincinnati, Ohio, USA. [Sonke, L Robert] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45167-0528 Cincinnati, Ohio, USA. [Pavelic, Ljiljana] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45167-0528 Cincinnati, Ohio, USA. [Okum, Eric] Ohio State University, Medical SchoolColumbus, Ohio, USA. [Neanen, Julie] Ohio State University, Medical SchoolColumbus, Ohio, USA. [Gluckman, L Jack] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45167-0528 Cincinnati, Ohio, USA. [Pavelic, P Zlatko] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45167-0528 Cincinnati, Ohio, USA. RP Pavelic, PZ (reprint author), University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45167-0528 Cincinnati, USA. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 1996 VL 2 IS 1 BP 30 EP 33 PG 4 ER PT J AU McDonald, SJ Gartside, SP Pavelic, JL Gluckman, LJ Pavelic, PZ AF McDonald, S John Gartside, S Peter Pavelic, J Ljiljana Gluckman, L Jack Pavelic, P Zlatko TI nm23-H1 Expression in Squamous Cell Carcinoma of the Head and Neck SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE head and neck cancer; nm23-H1 gene expression; immmunohistochemistry; prognosis ID head and neck cancer; nm23-H1 gene expression; immmunohistochemistry; prognosis AB Archival material from 47 patients with primary squamous cell carcinoma of the head and neck (SCCHN) was studied immunohistochemically for the presence of nm23-H1 protein. Our data indicate that nm23-H1 protein expression is a common event in SCCHN and that there is a trend toward correlation of increased expression of nm23-H1 with increasing tumor size (p = 0.072). The results also show that when adjusting for age and cause of death, there tended to be an inverse relationship between overall survival and the expression of nm23-H1 gene in the primary tumor (p = 0.088). C1 [McDonald, S John] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45267-0528 Cincinnati, Ohio, USA. [Gartside, S Peter] University of Cincinnati College of Medicine, Department of Environmental HealthCincinnati, Ohio, USA. [Pavelic, J Ljiljana] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45267-0528 Cincinnati, Ohio, USA. [Gluckman, L Jack] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45267-0528 Cincinnati, Ohio, USA. [Pavelic, P Zlatko] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45267-0528 Cincinnati, Ohio, USA. RP Pavelic, PZ (reprint author), University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45267-0528 Cincinnati, USA. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 1996 VL 2 IS 1 BP 34 EP 36 PG 3 ER PT J AU Piffko, J Bankfalvi, Ofner, D Totsch, M Berens, A Joos, U Bocker, W Schmid, WK AF Piffko, Jozsef Bankfalvi, Agnes Ofner, Dietmar Totsch, Martin Berens, Axel Joos, Ulrich Bocker, Werner Schmid, Werner Kurt TI Proliferative (MIB1, mdm2) Versus Anti-Proliferative (p53) Markers in Head and Neck Cancer. An Immunohistochemical Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p53; mdm2; MIB1; immunohistochemistry; wet autoclave pretreatment; head and neck cancer ID p53; mdm2; MIB1; immunohistochemistry; wet autoclave pretreatment; head and neck cancer AB Formalin fixed and paraffin embedded samples from 36 squamous cell carcinomas of the larynx and the oral cavity (pT2N0M0, R0) surrounded by non-tumorous mucosa were studied immunohistochemically using a panel of four different anti-p53 antibodies (CM1, PAb1801, D07, PAb240), a monoclonal anti-mdm2 antibody and MIB1, following wet autoclave antigen retrieval. P53 immunoreactivity was detected in 11/14 laryngeal and in 9/22 oral carcinomas. All p53 positive oral, and all but one laryngeal tumors revealed mdm2 positivity as well, whereas in p53 negative tumors 4/12 and 1/3 mdm2 immunopositive cases were demonstrated, respectively. MIB1 labeling indices of the tumors ranged between 18% - 64% in p53 positive cases, and 10% - 53% in p53 negative ones. The difference was not statistically significant. Close spatial coexpression of p53, mdm2 and MIB1 immunoreactivity was observed at the invasive front of the carcinomas and in the basal and suprabasal layers of the non-tumorous epithelium in all p53 positive cases. However, the MIB1 expression was similarly increased at the invasive margins in carcinomas lacking immunohistochemically detectable p53 alterations. Our results strongly suggest that p53 overexpression does not necessarily correspond to increased rate of proliferation, but rather to mdm2 overexpression and is largely dependent on the anatomical site in case of small and localized squamous cell carcinomas of the head and neck region. C1 [Piffko, Jozsef] University of Munster, Department of Cranio-Maxillofacial SurgeryMunster, Germany. [Bankfalvi, Agnes] University of Munster, Department of Pathology, Domagkstrasse 17, D-48149 Munster, Germany. [Ofner, Dietmar] University Hospital, Department of Surgery IInnsbruck, Austria. [Totsch, Martin] Medical University of Innsbruck, Department of PathologyInnsbruck, Austria. [Berens, Axel] University of Munster, Department of Cranio-Maxillofacial SurgeryMunster, Germany. [Joos, Ulrich] University of Munster, Department of Cranio-Maxillofacial SurgeryMunster, Germany. [Bocker, Werner] University of Munster, Department of Pathology, Domagkstrasse 17, D-48149 Munster, Germany. [Schmid, Werner Kurt] University of Munster, Department of Pathology, Domagkstrasse 17, D-48149 Munster, Germany. RP Schmid, WK (reprint author), University of Munster, Department of Pathology, D-48149 Munster, Germany. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 1996 VL 2 IS 1 BP 37 EP 42 PG 6 ER PT J AU Repassy, G Czigner, J Ribari, O AF Repassy, Gabor Czigner, Jeno Ribari, Otto TI Glottic Cancer of the Free Margin and Ventricular Surface of Vocal Cord SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE glottic cancer; intralaryngeal spread; histopathology ID glottic cancer; intralaryngeal spread; histopathology AB The authors differentiate cancer types of the glottis setting out from the free margin or the ventricular surface of the true vocal cord. The latter is considered to be a reliant clinicopathological unit starting from the dividing line of the stratified-ciliated epithelium margins, whereas the so called junctional tumor differs in its histogenesis and invasivity. They give a detailed description of intralaryngeal extension of these tumours on the basis of histopathological investigations. C1 [Repassy, Gabor] Medical University, Department of Oto-Rhino-Laryngology, Nagyerdei krt 98., H-4012 Debrecen, Hungary. [Czigner, Jeno] Szegedi Tudomanyegyetem, Ful-Orr-Gegeszeti KlinikaSzeged, Hungary. [Ribari, Otto] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. RP Repassy, G (reprint author), Medical University, Department of Oto-Rhino-Laryngology, H-4012 Debrecen, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 1996 VL 2 IS 1 BP 43 EP 47 PG 5 ER PT J AU Sztan, M Besznyak, I Kovacs, T Toth, J Szamel, I Olah, E AF Sztan, Marianna Besznyak, Istvan Kovacs, Tibor Toth, Jozsef Szamel, Iren Olah, Edit TI Lack of Correlation Between Survival and Allele Loss on Chromosome 7q31-32 in Primary Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chromosome 7q; tumor suppressor gene; loss of heterozygosity; primary breast carcinoma ID chromosome 7q; tumor suppressor gene; loss of heterozygosity; primary breast carcinoma AB High incidence of loss of heterozygosity (LOH), affecting the 7q31-32 chromosome region in sporadic primary human breast carcinomas suggests the presence of a tumor suppressor gene in this region which seems relevant to the development of breast cancer. To further determine the possible role of this region in the pathogenesis of human primary breast cancer and association with survival, LOH analysis was performed on 52 primary breast cancer patients using a set of highly polymorphic microsatellite markers. Our panel contained twenty biopsy cases of unknown survival, nineteen cases with more than five years survival and fourteen cases with less than two years survival. Corresponding normal and tumor DNAs were analyzed by polymerase chain reaction (PCR). The data presented here demonstrate that all patients were informative at least at one locus and 20 (38%) out of 53 cases showed LOH at one or more loci on chromosome 7q31-32. Relatively high incidence of LOH (34%) was detected at the D7S522 microsatellite marker located near to the cMet proto-oncogene while lower frequencies were observed at D7S523 (19%) and D7S495 (17%) loci, supporting the existence of a putative tumor suppressor gene at the chromosome 7q31.1 region. Our results suggest that allelic imbalance on 7q may occur at an early stage of breast carcinogenesis, as no correlation was observed between allelic loss and clinico-pathological data. C1 [Sztan, Marianna] National Institute of Oncology, Department of Biochemistry, Rath Gyorgy u. 7., H-1525 Budapest, Hungary. [Besznyak, Istvan] National Institute of Oncology, Department of SurgeryBudapest, Hungary. [Kovacs, Tibor] National Institute of Oncology, Department of SurgeryBudapest, Hungary. [Toth, Jozsef] National Institute of Oncology, Department of Diagnostic PathologyBudapest, Hungary. [Szamel, Iren] National Institute of Oncology, Department of Clinical ResearchBudapest, Hungary. [Olah, Edit] National Institute of Oncology, Department of Biochemistry, Rath Gyorgy u. 7., H-1525 Budapest, Hungary. RP Olah, E (reprint author), National Institute of Oncology, Department of Biochemistry, H-1525 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 1996 VL 2 IS 1 BP 48 EP 51 PG 4 ER PT J AU Kozma, L Papp, L Varga, Gomba, Sz AF Kozma, Laszlo Papp, Lajos Varga, Eva Gomba, Szabolcs TI Accumulation of Hg(II) Ions in Mouse Adrenal Gland SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hg(II); acute intoxication; adrenal gland; atomic absorption spectroscopy; autometallography ID Hg(II); acute intoxication; adrenal gland; atomic absorption spectroscopy; autometallography AB Female BALBc mice were administered HgCl2 at a single dose of 4 mg/kg i.p. The acute intoxication with Hg(II) salts (2 hr) caused accumulation of Hg(II) ions in the adrenal gland in general, and in the medulla, in particular. Based on data obtained with atomic absorption spectroscopy and quantitative cytochemistry, we determined the amount of mercury (II) in the adrenal glands and found it to be 14.2 ng Hg(II) (3.5 mg/kg wet weight of the adrenals). An uneven distribution of Hg(II) was found within the adrenal gland, not only between the medulla and cortex, but also within the cortex. The applied autometallographic method revealed that the cortex was negative except the zona glomerulosa, whereas the medulla showed a strong reaction localised to the chromaffin granules of the secretory cells. Both adrenaline and noradrenaline producing cells reacted. The comparison of the density of silver grains by scanning densitometry in the medulla and cortex revealed a significantly higher Hg(II) concentration in the medulla compared to the cortex (10 mg/kg vs 2 mg/kg, respectively). The results presented here suggest that there may be a connection between the symptoms of acute Hg(II) intoxication and its adrenal accumulation. C1 [Kozma, Laszlo] University of Debrecen, Faculty of Medicine, Department of Pathology, H-4012 Debrecen, Hungary. [Papp, Lajos] Lajos Kossuth University, Department of Inorganic and Analytical ChemistryDebrecen, Hungary. [Varga, Eva] Hungarian-Japanese Electron Microscopic CenterDebrecen, Hungary. [Gomba, Szabolcs] University of Debrecen, Faculty of Medicine, Department of Pathology, H-4012 Debrecen, Hungary. RP Kozma, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, H-4012 Debrecen, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 1996 VL 2 IS 1 BP 52 EP 55 PG 4 ER PT J AU Kovalszky, I Schaff, Zs Lapis, K Jeney, A AF Kovalszky, Ilona Schaff, Zsuzsa Lapis, Karoly Jeney, Andras TI Marker Enzymes of Rat Chemical Hepatocarcinogenesis in Human Liver Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE liver tumor; GGT; G-6-Pase; cytochrome P-450 ID liver tumor; GGT; G-6-Pase; cytochrome P-450 AB Reduced glucose-6-phosphatase, increased GGT activity and reduction of cytochrome P-450 content are considered to be markers of chemical hepatocarcinogenesis in rats. The significance of these changes were studied in certain human liver lesions; adenoma, focal nodular hyperplasia and hepatocellular carcinoma all developed in non-cirrhotic livers. Enzymes showed normal values in 4 out of 5 adenomas, in 2/13 FNH and in 4/18 HCC samples. The decreased cP-450 content in HCC proved to be the most consistent alteration (12/18). Only 3 HCC samples possessed changes off all enzymes. These data suggest that at least those enzymes which are used as markers in rat chemical hepatocarcinogenesis have little or no biological significance in human liver tumors, primarily due to the intertumoral heterogeneity of enzyme activity. Such heterogeneity was observed in the peritumoral ''normal'' liver tissue, too. C1 [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Lapis, Karoly] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Jeney, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Kovalszky, I (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM koval@korb1.sote.hu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 56 EP 58 PG 3 ER PT J AU Horvath, G Stotz, Gy Tolvaj, Gy Osztrogonacz, H David, K AF Horvath, Gabor Stotz, Gyula Tolvaj, Gyula Osztrogonacz, Henrik David, Karoly TI The Effect Of Long Term and High Dose Interferon Treatment In Chronic Hepatitis C SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chronic hepatitis C; interferon treatment ID chronic hepatitis C; interferon treatment AB The results of 43 interferon treatments of 35 patients (23 male, 12 female) are reported. The duration of the treatment was 6-18 months, the dose of interferon was 3x3-5 MU weekly. Complete response (HCV RNA became negative) was found in 11, relapse was observed in 3 patients. Partial response (transaminase levels became normal, or less than twice normal value, but patients remained HCV RNA positive) occurred in 23 cases, relapse was obeserved in 16. The therapy had no effect in 9 cases. The higher dose and longer term interferon therapy resulted in a higher rate of response to the treatment and a reduction in the number of relapses. C1 [Horvath, Gabor] Central Hospital of the Ministry of the Interior, 1st Department of Medicine, Budakeszi ut 48/b, 1121 Budapest, Hungary. [Stotz, Gyula] Central Hospital of the Ministry of the Interior, Department of PathologyBudapest, Hungary. [Tolvaj, Gyula] Central Hospital of the Ministry of the Interior, 1st Department of Medicine, Budakeszi ut 48/b, 1121 Budapest, Hungary. [Osztrogonacz, Henrik] Central Hospital of the Ministry of the Interior, 1st Department of Medicine, Budakeszi ut 48/b, 1121 Budapest, Hungary. [David, Karoly] Central Hospital of the Ministry of the Interior, 1st Department of Medicine, Budakeszi ut 48/b, 1121 Budapest, Hungary. RP Horvath, G (reprint author), Central Hospital of the Ministry of the Interior, 1st Department of Medicine, 1121 Budapest, Hungary. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 59 EP 62 PG 4 ER PT J AU Benfares, J Le Tourneau, A Audouin, J Szekeres, Gy AF Benfares, Jamal Le Tourneau, Agnes Audouin, Josee Szekeres, Gyorgy TI Effect of Ribonuclease A and Deoxyribonuclease I on Immunostaining of Ki-67 in Cultured Melanoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ki-67; immunocytochemistry; enzymatic pre-digestion; proliferating cells ID Ki-67; immunocytochemistry; enzymatic pre-digestion; proliferating cells AB Immunostaining of the cell cycle-associated Ki-67 antigen was studied, using the Ki-67-specific MIB-1 monoclonal antibody on slides prepared by cytocentrifugation of cultured A375 melanoma cells. Immunomorphological analysis of the Ki-67 immunostaining pattern of both nuclear and nucleolar locations was carried out following pre-treatment of the slides including ribonuclease and deoxyribonuclease pre-digestion of the cells. Immunostaining of nucleolar Ki-67 was reduced by ribonuclease pre-digestion, but was not altered by deoxyribonuclease pre-treatment. Ribonuclease did not reduce the staining intensity of Ki-67 in the nuclear matrix, but the intensity decreased after deoxyribonuclease pre-digestion. We suggest that the Ki-67 molecule may play an important role in ensuring contact between nuclear DNA and nucleolar RNA during transcriptional processes in cell proliferation. C1 [Benfares, Jamal] Immunotech, Antibody Department, R&D LaboratoryMarseille, France. [Le Tourneau, Agnes] Hotel-Dieu, Laboratory of PathologyParis, France. [Audouin, Josee] Hotel-Dieu, Laboratory of PathologyParis, France. [Szekeres, Gyorgy] Hisztopatologia KFT, Akac u. 8., H-7632 Pecs, Hungary. RP Szekeres, Gy (reprint author), Hisztopatologia KFT, H-7632 Pecs, Hungary. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 63 EP 65 PG 3 ER PT J AU Hrzenjak, MT Roguljic, A Efenberger-Marinculic, P Popovic, M Pisl, Z AF Hrzenjak, M Terezija Roguljic, Ante Efenberger-Marinculic, Palma Popovic, Maja Pisl, Zoran TI Total IgA and IgG in Sera of Patients With Different Primary Malignancies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE IgA; IgG; malignant tumors ID IgA; IgG; malignant tumors AB The concentrations of total serum IgA and IgG of 267 patients with different primary malignant tumors were measured by ELISA. Total serum IgA increased by 30% to 40% in patients with malignancies associated with mucous membranes (nasopharyngeal, gastrointestinal and bronchial carcinomas), while the change in total serum IgG was negligible. Although, the changes in Ig level could be influenced by many host factors, these data call attention to the potential indicative role of total serum IgA levels. Further studies are required to establish links between serum IgA levels and stages of tumor growth or tumor progression in order to use these values as prognostic factors. C1 [Hrzenjak, M Terezija] Faculty of Veterinary Medicine, Department of Biology, Heinzelova 55., 10000 Zagreb, Croatia. [Roguljic, Ante] University Hospital for TumorsZagreb, Croatia. [Efenberger-Marinculic, Palma] Institute for Medical Research and Occupational HealthZagreb, Croatia. [Popovic, Maja] Faculty of Veterinary Medicine, Department of Biology, Heinzelova 55., 10000 Zagreb, Croatia. [Pisl, Zoran] Institute for Medical Research and Occupational HealthZagreb, Croatia. RP Hrzenjak, MT (reprint author), Faculty of Veterinary Medicine, Department of Biology, 10000 Zagreb, Croatia. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 66 EP 68 PG 3 ER PT J AU Griciute, L Domkiene, V AF Griciute, Laima Domkiene, Vida TI Experimental Study on the Carcinogenicity of the Cytostatic Drug Ftorafur (Tegafur) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ftorafur; Tegafur; carcinogenicity; mouse; rat ID Ftorafur; Tegafur; carcinogenicity; mouse; rat AB Long-term carcinogenicity of Ftorafur (Tegafur) was studied in rodents. Rats and mice were treated for one year per os with 40 (mice) and 60 (rat) mg/kg Ftorafur twice a week and were followed for their entire life. Analysis of the data provide no evidence for the carcinogenicity of Ftorafur in rodents. These findings are similar to other antimetabolite studies and contrasts with the carcinogenic alkylating agents. C1 [Griciute, Laima] Lithuanian Oncology Center, Santariskiy 1., 2600 Vilnius, Lithuania. [Domkiene, Vida] Lithuanian Oncology Center, Santariskiy 1., 2600 Vilnius, Lithuania. RP Griciute, L (reprint author), Lithuanian Oncology Center, 2600 Vilnius, Lithuania. EM onko.centras@loc.lt NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 69 EP 70 PG 2 ER PT J AU Bankfalvi, Piffko, J Ofner, D Dreier, R Bocker, W Schmid, WK AF Bankfalvi, Agnes Piffko, Jozsef Ofner, Dietmar Dreier, Rita Bocker, Werner Schmid, Werner Kurt TI Significance of Wet Autoclave Pretreatment in Immunohistochemistry SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE Wet autoclave pretreatment; Immunohistochemistry; AgNOR ID Wet autoclave pretreatment; Immunohistochemistry; AgNOR AB Until recently the only way to rescue masked epitopes in routinely processed surgical pathological material was enzymatic digestion. The use of heat for antigen retrieval, first by microwave irradiation, represents an important breakthrough in immunohistochemistry. With the acceptance of microwave oven pretreatment, various modified techniques and alternative heating methods have also been proposed. Wet autoclave pretreatment for tissue proteolysis is a highly reliable alternative to the microwave antigen retrieval technique. It provides uniform heating of the slides, hence an even enhancement of staining intensity in a variety of formalin-sensitive antigens, and it also offers consistent interlaboratory results. The method has been introduced in routine diagnostic immunohistochemistry for the detection of estrogen- and progesterone receptors, L26-, Ki-67- and bcl-2 antigens and variable types of cytokeratins (1/5/10/11, 8, 13, 19). Experimentally, wet autoclaving can be used very successfully for the immunophenotyping of p53 and mdm2 expression, for the detection of adhesion molecules (CD44, integrins) and some anti-inflammatory molecules (annexins), among others. It has produced a substantial improvement in the visualisation of silver-stained nucleolar organizer regions- associated proteins (AgNORs) in routine paraffin sections and along with modified silver staining and standardized AgNOR parameters assessed by image analysis. Wet autoclaving-based AgNOR staining has been proposed by a European multicentric study group as the standardized method for AgNOR analysis in archival material. C1 [Bankfalvi, Agnes] University of Munster, Department of Pathology, Domagkstrasse 17, D-48149 Munster, Germany. [Piffko, Jozsef] University of Munster, Department of Cranio-Maxillofacial SurgeryMunster, Germany. [Ofner, Dietmar] University Hospital, Department of Surgery IInnsbruck, Austria. [Dreier, Rita] University of Munster, Department of Pathology, Domagkstrasse 17, D-48149 Munster, Germany. [Bocker, Werner] University of Munster, Department of Pathology, Domagkstrasse 17, D-48149 Munster, Germany. [Schmid, Werner Kurt] University of Munster, Department of Pathology, Domagkstrasse 17, D-48149 Munster, Germany. RP Schmid, WK (reprint author), University of Munster, Department of Pathology, D-48149 Munster, Germany. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 71 EP 77 PG 7 ER PT J AU Mihalik, R Uher, F Pocsik, Berczi, L Benczur, M Kopper, L AF Mihalik, Rudolf Uher, Ferenc Pocsik, Eva Berczi, Lajos Benczur, Miklos Kopper, Laszlo TI Detection of Drug-induced Apoptosis by Flow Cytometry after Alkaline Extraction of Ethanol Fixed Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE apoptosis; cell cycle; drug-induced; DNA extraction; flow cytometry ID apoptosis; cell cycle; drug-induced; DNA extraction; flow cytometry AB A new flow cytometric method was developed to detect apoptotic cells with fragmented DNA and to determine cell cycle distribution of viable cells, in the same sample, by propidium iodide staining. Apoptosis, in HT58 human B lymphoma cells, was induced by etoposide and/or by staurosporine. Using appropriate alkaline solutions (between 1-10 mN NaOH in 150 mM saline) followed by neutralization with buffer solution, the fragmented DNA can be extracted quantitatively from ethanol fixed cells. Further, good resolution of the cell cycle distribution can be obtained in unimpaired cells without RNase treatment. Furthermore, unlike the widely used hypotonic-detergent extraction of unfixed cells, the suggested extraction method can prevent drug-induced disintegration of dead cells when karyorrhexis occurs. C1 [Mihalik, Rudolf] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Uher, Ferenc] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Pocsik, Eva] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Berczi, Lajos] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Benczur, Miklos] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 78 EP 83 PG 6 ER PT J AU Orosz, Zs Toth, E Viski, A AF Orosz, Zsolt Toth, Erika Viski, Anna TI Osteoclastoma-like Giant Cell Tumor of the Lung SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE osteoclast-like giant cells; giant cell tumor; lung; p53; PCNA ID osteoclast-like giant cells; giant cell tumor; lung; p53; PCNA AB The main components of an unusual form of lung tumor were osteoclast-like multinucleated giant cells and mononuclear stromal cells. Besides, scattered islands of moderately differentiated squamous cells also appeared. Both the mononuclear and the osteoclast-like giant cells reacted with antibodies against CD68 and vimentin, but did not react with antibodies against cytokeratin, EMA and CEA, or lysozyme and a-1-antitrypsin. The p53 and PCNA antigens were positive only in mononuclear cells and not the osteoclast-like giant cells, suggesting that mononuclear cells represent proliferating elements with histiocytic differentiation while osteoclast-like giant cells are stromal, presumably reactive components of the tumor. C1 [Orosz, Zsolt] National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, Rath Gyorgy u. 7-9., 1122 Budapest, Hungary. [Toth, Erika] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Viski, Anna] Kaposi Mor Hospital, Department of PathologyKaposvar, Hungary. RP Orosz, Zs (reprint author), National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, 1122 Budapest, Hungary. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 84 EP 88 PG 5 ER PT J AU Toth, J Kerenyi, Suveges, I Futo, G AF Toth, Jeannette Kerenyi, Agnes Suveges, Ildiko Futo, Gabor TI Leiomyoma of The Ciliary Body and Hemangiopericytoma of the Choroid SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE intraocular non-melanocytic tumors; leiomyoma; hemangiopericytoma ID intraocular non-melanocytic tumors; leiomyoma; hemangiopericytoma AB Two unusual uveal tumors occurring in eyes enucleated for presumed malignant melanoma are discussed. One was a leiomyoma of the ciliary body, affecting a 22-year-old female, the other a hemangiopericytoma of the choroid in an 84-year-old male patient. The latter case is the fourth intraocular hemangiopericytoma reported in the literature to date. The histopathologic diagnosis was confirmed by immunohistochemistry and electron microscopy. C1 [Toth, Jeannette] Semmelweis University, Department of Ophthalmology, Tomo u. 25-29., 1083 Budapest, Hungary. [Kerenyi, Agnes] Semmelweis University, Department of Ophthalmology, Tomo u. 25-29., 1083 Budapest, Hungary. [Suveges, Ildiko] Semmelweis University, Department of Ophthalmology, Tomo u. 25-29., 1083 Budapest, Hungary. [Futo, Gabor] Szent Borbala Hospital, Department of OphthalmologyTatabanya, Hungary. RP Toth, J (reprint author), Semmelweis University, Department of Ophthalmology, 1083 Budapest, Hungary. EM tj@szem1.sote.hu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 89 EP 93 PG 5 ER PT J AU Patonai, A Csikos, A Deak, Gy AF Patonai, Attila Csikos, Andras Deak, Gyorgy TI Visceral Aluminum Deposition In Chronic Renal Insufficiency (Light Microscopy and X-ray Microanalysis) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE aluminum; renal insufficency; myocardium; lung; kidney ID aluminum; renal insufficency; myocardium; lung; kidney AB Aluminum is a common element in our environment, but has been proved to be toxic, mainly in chronic renal insufficiency. Most cases of ALU intoxication occur during hemodialysis due to treatment of aluminum-containing drugs. In the present case, we describe visceral manifestations of aluminum deposition in a middle aged, multidialysed, male patient. Light and polarization microscopy examinations and X-ray microanalysis revealed amorph, extracellular aluminum deposits in various parenchymal organs causing failure of heart, lung and kidney functions. There were no anamnestic data concerning aluminum-containing drugs or occupational exposure. C1 [Patonai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Csikos, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Deak, Gyorgy] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. RP Patonai, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM patonai@korb1.sote.hu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 94 EP 97 PG 4 ER PT J AU Honn, VK Aref, A Chen, QY Cher, LM Crissman, DJ Forman, DJ Gao, X Grignon, D Hussain, M Porter, TA Pontes, E Powell, I Redman, B Sakr, W Severson, R Tang, GD Wood, PD AF Honn, V Kenneth Aref, Amer Chen, Q Yong Cher, L Michael Crissman, D John Forman, D Jeffrey Gao, Xiang Grignon, David Hussain, Maha Porter, T Arthur Pontes, J. Edson Powell, Isaac Redman, Bruce Sakr, Wael Severson, Richard Tang, G Dean Wood, P David TI Prostate Cancer, Old Problems and New Approaches Part I. SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE prostate cancer; epidemiology; incidence; genetics; progression ID prostate cancer; epidemiology; incidence; genetics; progression AB Rates of prostate cancer (PCa) have increased so dramatically over the last decade that the age adjusted incidence rate for PCa is now greater than that any other cancer among men in the United States. This review, published as a three part series, provides a state-of-art assessment of the PCa problem in its divergent aspects. Part 1 covers epidemiology, incidence and progression. Several epidemiological studies have demostrated that first degree male relatives of men with PCa are at increased risk of developing the disease. Familial and genetic factors as well as medical, anthropometric, dietary, hormonal and occupational factors involved in PCa are discussed. Postmortem examination of the prostate in men without evidence of PCa documented a high frequency of adenocarcinoma. Latent disease occurred as early as the second decade of life. Although there is no significant difference in incidence between Caucasian and African-American males, high grade prostatic intraepithelial neoplasia (HGPIN) is higher in the latter group. While dietary fat, androgens and certain environmental factors may be determinants for PCa, the exact mechanism of tumorigenesis is still relatively unknown. The current thinking of the role of genomic instability, chromosomal alterations, tumor suppressor genes and the androgen receptor are explored. C1 [Honn, V Kenneth] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48201 Detroit, Michigan, USA. [Aref, Amer] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48201 Detroit, Michigan, USA. [Chen, Q Yong] Wayne State University, Department of PathologyDetroit, USA. [Cher, L Michael] Wayne State University, Department of UrologyDetroit, USA. [Crissman, D John] Wayne State University, Department of PathologyDetroit, USA. [Forman, D Jeffrey] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48201 Detroit, Michigan, USA. [Gao, Xiang] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48201 Detroit, Michigan, USA. [Grignon, David] Wayne State University, Department of PathologyDetroit, USA. [Hussain, Maha] Wayne State University, Department of MedicineDetroit, USA. [Porter, T Arthur] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48201 Detroit, Michigan, USA. [Pontes, J. Edson] Wayne State University, Department of UrologyDetroit, USA. [Powell, Isaac] Wayne State University, Department of UrologyDetroit, USA. [Redman, Bruce] Wayne State University, Department of MedicineDetroit, USA. [Sakr, Wael] Wayne State University, Department of PathologyDetroit, USA. [Severson, Richard] Wayne State University, Department of Family MedicineDetroit, USA. [Tang, G Dean] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48201 Detroit, Michigan, USA. [Wood, P David] Wayne State University, Department of UrologyDetroit, USA. RP Honn, VK (reprint author), Wayne State University, Department of Radiation Oncology, 48201 Detroit, USA. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1996 VL 2 IS 2 BP 98 EP 109 PG 12 ER PT J AU Tang, GD Porter, TA AF Tang, G Dean Porter, T Arthur TI Apoptosis: A Current Molecular Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE apoptosis; genes; review ID apoptosis; genes; review AB Apoptosis is a cell suicide program characterized by distinct morphological (cell shrinkage, membrane blebbing, pyknosis, chromatin margination, denser cytoplasmic images) and biochemical (e.g., DNA fragmentation into distinct ladders; degradation of apoptotic markers such as PARP and nuclear lamins) features. It is involved in multiple physiological processes examplified by involution of mammary tissues, embryonic development, homeostatic maintenance of tissues and organs, and maturation of the immune system, as well as in many pathological conditions represented by neurologic degeneration (Alzeimer's disease), autoimmune and inflammatory diseases, etiology of atherosclerosis, AIDS, and oncogenesis and tumor progression. Numerous molecular entities have been shown to regulate the apoptotic process. This review provides a concise summary of the recent data on the role of oncogenes/tumor suppressor genes, cytokines and growth factors/growth factor receptors, intracellular signal transducers, cell cycle regulators, reactive oxygen species or other free radicals, extracellular matrix regulators/cell adhesion molecules, and specific endonucleases and cytoplasmic proteases (the ICE family proteins) in regulating cell survival and apoptosis. Elucidation of the molecular mechanisms regulating apoptosis bears tremendous impact on enhancing our understanding of many diseases inflicting the human beings and undoubtedly brings us hope for the cure of these diseases. C1 [Tang, G Dean] Wayne State University, Department of Radiation Oncology, 407 Life Science Bldg., 48202 Detroit, MI, USA. [Porter, T Arthur] Wayne State University, Department of Radiation Oncology, 407 Life Science Bldg., 48202 Detroit, MI, USA. RP Tang, GD (reprint author), Wayne State University, Department of Radiation Oncology, 48202 Detroit, USA. EM dtang@cms.cc.wayne.edu NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 117 EP 131 PG 15 ER PT J AU Schaff, Zs Lotz, G Schulte-Herman, R AF Schaff, Zsuzsa Lotz, Gabor Schulte-Herman, Rolf TI Pathomorphological Characteristics and Pathogenesis of Viral Hepatitis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE pathomorphology; pathogenesis; viral; hepatitis ID pathomorphology; pathogenesis; viral; hepatitis AB Viral hepatitis (VH) is an inflammatory reaction of the liver to hepatotropic viruses. Acute VH can be classified according to the virus and type of necrosis. Chronic hepatitis (CH) might be active, persistent or lobular based on previous classification. More recently the grade (necroinflammatory activity) and stage (fibrosis and architectural distorsion) of CH have been distinguished and scored. Apoptosis and necrosis probably coexist in VH and contribute to hepatocyte death. Several ''death factors'', such as transforming growth factor b, Apo1/Fas and tumor necrosis factor play a role in the execution of cell death. Injury of hepatocytes during viral infection can occur as a direct effect of the virus or as a result of the host immune response. Expression of different viral antigens can be detected during VH and might be visualized. Phenotyping of the portal inflammatory cell infiltrate in CH has shown a T-cell zone comprised of CD4+ helper T cells and CD8+ supressor/cytotoxic T cells at the periphery of the lobules. The pathogenetic mechanisms responsible for the final outcome of viral infection depend on viral factors (such as genotype, mutation etc.), virus-host interaction, expression of viral protein, several cytokines etc. which finally lead to the well known histological alterations of viral hepatitis. C1 [Schaff, Zsuzsa] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Lotz, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Schulte-Herman, Rolf] Medical University of Vienna, Department of Experimental Pathology and Laboratory Animal PathologyVienna, Austria. RP Schaff, Zs (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 132 EP 143 PG 12 ER PT J AU Yamashima, T AF Yamashima, Tetsumori TI On Arachnoid Villi and Meningiomas: Functional Implication of Ultrastructure, Cell Adhesion Mechanisms, and Extracellular Matrix Composition SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE arachnoid villus; meningioma; E-cadherin; prostaglandin D2 synthesis; multilamellar phospholipids ID arachnoid villus; meningioma; E-cadherin; prostaglandin D2 synthesis; multilamellar phospholipids AB Arachnoid villi or granulations are small projections of the arachnoid barrier layer into the venous sinus and its major tributaries. They are closely related to the absorption of cerebrospinal fluid, and are widely accepted to be the origin of human meningiomas. Arachnoid villi and meningiomas show a number of similarities in ultrastructure, cell adhesion mechanisms, and extracellular matrix composition. Ultrastructurally, both arachnoid and meningioma cells are characterized by interdigitations connected with junctional complexes, and extracellular cisterns related to the fluid transport. Extracellular cisterns and the intercellular space reveal abundant membrane-derived multilamellar phospholipids when a conventional ultrastructural fixative supplemented with tannic acid is used. Both arachnoid and meningioma cells are connected by Ca2+-dependent adhesion molecules: epithelial-cadherins which are concentrated at the adherens junctions. Membrane-cytoskeleton interactions by means of merlin and a-catenin molecules are thought to be crucial in signal transduction resulting in contact inhibition of cell growth in normal arachnoid cells. Impairment of these molecules might be related to meningioma-genesis. Glutathione-independent prostaglandin D2 synthase [EC 5.3.99.2] responsible for the biosynthesis of prostaglandin D2 in the central nervous system is also consistently expressed in human arachnoid villi and meningiomas. The multilamellar phospholipids are conceivably related to this arachidonate metabolism. C1 [Yamashima, Tetsumori] Kanazawa University School of Medicine, Department of Neurosurgery, Takaramachi 13-1, 920 Kanazawa, Japan. RP Yamashima, T (reprint author), Kanazawa University School of Medicine, Department of Neurosurgery, 920 Kanazawa, Japan. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 144 EP 149 PG 6 ER PT J AU Gatenby, AR Seftor, AE Hendrix, JM AF Gatenby, A Robert Seftor, A Elisabeth Hendrix, JC Mary TI Fibroblast Enhancement of Tumor Invasion in a Tumor-Host Interface Recapitulated in-vitro SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE invasion; cartilage; pannus; matrix metalloproteinases; cytokines ID invasion; cartilage; pannus; matrix metalloproteinases; cytokines AB Tumor cells and fibroblasts were isolated from the tumor-host interface of a colon 4047 tumor growing subcutaneously in a Fischer 344 rat. The populations were co-cultured to recapitulate the tumor-host interface in vitro. The co-cultured populations grew in a predictable pattern with tumor cells forming nodules surrounded by fibroblasts. Population dynamic experiments demonstrated the fibroblasts enhanced the growth of the tumor cells but tumor inhibited and ultimately destroyed the fibroblasts. Video microscopic examination of the fibroblasts demonstrated intense membrane ruffling adjacent to the tumor nodules followed by membrane fragmentation and detachment. Immunohistochemical staining for gelatinase A was markedly positive within the fibroblasts surrounding the tumor nodules; but negative within the tumor and in fibroblasts when tumor was absent. This technique recapitulates many aspects of the tumor-host interface in vitro and may be a useful model for evaluating several aspects of tumor-host interaction. C1 [Gatenby, A Robert] Temple University Hospital, Department of Diagnostic Imaging, Broad and Ontario Streets, 19140 Philadelphia, PA, USA. [Seftor, A Elisabeth] University of Iowa, Department of AnatomyIowa City, USA. [Hendrix, JC Mary] University of Iowa, Department of AnatomyIowa City, USA. RP Gatenby, AR (reprint author), Temple University Hospital, Department of Diagnostic Imaging, 19140 Philadelphia, USA. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 151 EP 156 PG 6 ER PT J AU Frye, AC Yocum, ED Tuan, R Suyana, E Seftor, AE Seftor, ER Khalkhali-Ellis, Z Moore, LT Hendrix, JM AF Frye, A Catherine Yocum, E David Tuan, Rocky Suyana, Eiko Seftor, A Elisabeth Seftor, EB Richard Khalkhali-Ellis, Zhila Moore, L Terry Hendrix, JC Mary TI An in vitro Model for Studying Mechanisms Underlying Synoviocyte-Mediated Cartilage Invasion in Rheumatoid Arthritis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE invasion; cartilage; pannus; matrix metalloproteinases; cytokines ID invasion; cartilage; pannus; matrix metalloproteinases; cytokines AB Rheumatoid arthritis (RA) is a chronic inflammatory disease of joints involving the pathological development of an invasive and destructive pannus tissue which contributes to the loss of cartilage and bone. To further analyze the process of cartilage degradation and invasion, we have developed an in vitro model composed of cartilage matrix and synoviocytes (isolated from RA pannus tissue, as well as normal synovial membrane). The matrix is derived from pig articular cartilage and contains collagen type II and proteoglycans and is similar in composition to human cartilage. Data generated from this model reveal that synoviocytes isolated from RA pannus tissue invaded cartilage matrix in a manner which directly correlated with the severity of the disease. Analysis of mechanisms associated with the invasive process demonstrate that highly invasive RA synoviocytes maintain a round morphology during attachment and spreading on cartilage matrix, compared with their normal counterparts. Furthermore, the level of secretion of matrix metalloproteinase (MMP) activity was shown to correlate with the RA phenotype, which could be modulated with a novel MMP inhibitor. Normal synoviocytes could be ''converted'' to an RA phenotype by specific inflammatory cytokines, such that invasion of cartilage matrix was augmented by culturing these cells in the presence of 5 U/ml IL-1b or 18 U/ml TGFb. Invasion was inhibited by 150 U/ml TNFa, and unaffected by 100 ng/ml PDGF. In addition, synovial fluid from RA patients induced invasion of normal synoviocytes, in a concentration dependent manner, from 150% to 460%; however, synovial fluid from another inflammatory arthritidy (Crohn's) did not augment invasion to the same degree. Moreover, this ''conversion effect'' appears to be specific for synoviocytes, since similar effects could not be achieved with human skin fibroblasts. This in vitro model of synoviocyte-mediated cartilage invasion allows for further molecular characterization of the invasive properties of the synoviocyte which contribute to RA. C1 [Frye, A Catherine] University of Arizona, Department of Microbiology and ImmunologyTucson, USA. [Yocum, E David] University of Arizona, Department of RheumatologyTucson, USA. [Tuan, Rocky] Thomas Jefferson University, Department of Orthopaedic SurgeryPhiladelphia, USA. [Suyana, Eiko] Thomas Jefferson University, Department of Orthopaedic SurgeryPhiladelphia, USA. [Seftor, A Elisabeth] Saint Louis University, School of Medicine, Departments of Pediatrics and Internal MedicineSt. Louis, USA. [Seftor, EB Richard] Saint Louis University, School of Medicine, Departments of Pediatrics and Internal MedicineSt. Louis, USA. [Khalkhali-Ellis, Zhila] Saint Louis University, School of Medicine, Departments of Pediatrics and Internal MedicineSt. Louis, USA. [Moore, L Terry] Saint Louis University, School of Medicine, Departments of Pediatrics and Internal MedicineSt. Louis, USA. [Hendrix, JC Mary] Saint Louis University, School of Medicine, Departments of Pediatrics and Internal MedicineSt. Louis, USA. RP Hendrix, JM (reprint author), Saint Louis University, School of Medicine, Departments of Pediatrics and Internal Medicine, St. Louis, USA. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 157 EP 166 PG 10 ER PT J AU Gergely, P Vertesi, Cs AF Gergely, Peter Vertesi, Csaba TI Urinary Excretion of Thioamine Compounds in Patients with Malignant Tumors and Inflammation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE urinary thioamine; tumor; inflammation ID urinary thioamine; tumor; inflammation AB The Kokonov's reaction measures thioamine-like compounds excreted in the urine. We compared the results obtained by this reaction in patients with malignant tumors and inflammatory diseases. The majority of patients with advanced (stage III-IV) cancer displayed a positive test. The chance of obtaining a positive test was related to the tumor mass. Lymphoma patients in complete remission were negative. Patients suffering from bacterial infections, active autoimmune diseases, and acute pancreatitis or myocardial infarction also had positive tests. The increased excretion of thioamines, found in tumor-bearing patients can be explained by the activation of the immune system, probably by the inflammation around or within the tumor tissue. The Kokonov's reaction is not suitable to discriminate between tumor-bearing patients and healthy ones. It rather seems to be appropriate to assess inflammatory processes in infections and/or autoimmune diseases. C1 [Gergely, Peter] Semmelweis University, Department of Laboratory Medicine, Ulloi ut 26., H-1085 Budapest, Hungary. [Vertesi, Csaba] Chinoin Pharmacological and Chemical Works Co. Ltd.Budapest, Hungary. RP Gergely, P (reprint author), Semmelweis University, Department of Laboratory Medicine, H-1085 Budapest, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 167 EP 170 PG 4 ER PT J AU Patyanik, M Mayer, Ungar, L Polgar, I AF Patyanik, Mihaly Mayer, Arpad Ungar, Laszlo Polgar, Istvan TI Early Experiences with Combined Treatment (Surgery and Brachytherapy) of Gynecological Recurrences Infiltrating the Pelvic Wall SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE pelvic side wall relapses; combined operative and radiotherapeutic treatment ID pelvic side wall relapses; combined operative and radiotherapeutic treatment AB Neither the surgical nor the radiotherapeutic treatment of gynecological recurrences infiltrating the pelvic wall can be curative alone. The treatment of this group of patients is possible with the CORT (Combined Operative and Radiotherapeutic Treatment) method. As maximal as possible resection of the malignancy is done for patients having no distant metastases and the brachytherapy guiding tubes are implanted into the tumour bed on the pelvic wall. The brachytherapy is carried out postoperatively. An accurate analysis cannot be done because of the short follow up and the few cases. The authors do believe that the survival results of this poor prognostic group may improve by means of this method after proper selection of patients. C1 [Patyanik, Mihaly] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai Kozpont, Uzsoki u. 29., H-1145 Budapest, Hungary. [Mayer, Arpad] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai Kozpont, Uzsoki u. 29., H-1145 Budapest, Hungary. [Ungar, Laszlo] Szent Istvan Hospital, Department of Obstetrics and GynecologyBudapest, Hungary. [Polgar, Istvan] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai Kozpont, Uzsoki u. 29., H-1145 Budapest, Hungary. RP Patyanik, M (reprint author), Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai Kozpont, H-1145 Budapest, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 171 EP 173 PG 3 ER PT J AU Totth, Schnur, J Ladanyi, A Kopper, L AF Totth, Arpad Schnur, Janos Ladanyi, Andrea Kopper, Laszlo TI Intracerebral Human Lymphoma - An Experimental Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lymphoma; CNS; tumor-model ID lymphoma; CNS; tumor-model AB Primary central nervous system lymphomas are rather rare, however, their frequency seems to be increasing in both high risk and immunocompetent patients with very poor prognosis. Here we describe a model intracerebrally xenotransplating human non Hodgkin lymphoma cells of B cell origin (HT 58). This offers a unique possibility to study the theraputic response, especially on systemic treatment. Briefly, one million lymphoma cells grew as meningeal tumor mass, infiltrating the brain directly as well as via the perivascular space. The lymphoma cells preserved all the phenotypic characteristics of the source tumor. C1 [Totth, Arpad] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Schnur, Janos] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Ladanyi, Andrea] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM kopper@korb1.sote.hu NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 174 EP 176 PG 3 ER PT J AU Bergter, W Fetzer, IC Sattler, B Ramadori, G AF Bergter, Wolfgang Fetzer, Ingrid-Corina Sattler, Burkhardt Ramadori, Giuliano TI Granulomatous Hepatitis preceding Hodgkin's Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE granuloma; granulomatous hepatitis; Hodgkin's disease ID granuloma; granulomatous hepatitis; Hodgkin's disease AB Granulomas are a frequent finding in various organs with a wide range of causes. Hepatic granulomas most often occur in the course of sarcoidosis or tuberculosis. However, they also may develop during Hodgkin's disease or sometimes even precede lymphoma as shown in the case of a patient presented here. Early diagnosis of Hodgkin's disease is essential, and in the case of unexplained granulomatous hepatitis it can be achieved by taking the patients history into consideration. C1 [Bergter, Wolfgang] Abteilung Gastroenterologie und Endokrinologie, Zentrum Innere Medizin, Robert Koch Str 40, 37075 Gottingen, Germany. [Fetzer, Ingrid-Corina] Abteilung Gastroenterologie und Endokrinologie, Zentrum Innere Medizin, Robert Koch Str 40, 37075 Gottingen, Germany. [Sattler, Burkhardt] Georg-August-University, Department of PathologyGottingen, Germany. [Ramadori, Giuliano] Abteilung Gastroenterologie und Endokrinologie, Zentrum Innere Medizin, Robert Koch Str 40, 37075 Gottingen, Germany. RP Ramadori, G (reprint author), Abteilung Gastroenterologie und Endokrinologie, Zentrum Innere Medizin, 37075 Gottingen, Germany. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 177 EP 180 PG 4 ER PT J AU Kovacs, J Poka, R AF Kovacs, Judit Poka, Robert TI Lipoma of the Uterus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE uterine neoplasm; postmenopausal woman; lipoma; immunohistochemistry ID uterine neoplasm; postmenopausal woman; lipoma; immunohistochemistry AB Lipoma of the uterus is a rare condition usually developing in postmenopausal women. Clinical symptoms and physical signs are similar to those found in leiomyomas. The histiogenesis of these lesions is still unclear. We report a case of a pure intramural lipoma of the uterus illustrating characteristic morphological and histological findings. Additional immunohistochemical studies underline the lipomatous but not leiomyomatous nature of the tumor. C1 [Kovacs, Judit] University of Debrecen, Faculty of Medicine, Department of Pathology, H-4012 Debrecen, Hungary. [Poka, Robert] University Medical School of Debrecen, Department of Gynecology and ObstetricsDebrecen, Hungary. RP Kovacs, J (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, H-4012 Debrecen, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 181 EP 183 PG 3 ER PT J AU Kohalmi, F Strausz, J Egervary, M Szekeres, Gy Timar, J AF Kohalmi, Ferenc Strausz, Janos Egervary, Marta Szekeres, Gyorgy Timar, Jozsef TI Differential Expression of Markers in Extensive and Restricted Langerhans Cell Histiocytosis (LCH) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Langerhans cell histiocytosis; restricted and extensive variants; markers; immunohistochemistry ID Langerhans cell histiocytosis; restricted and extensive variants; markers; immunohistochemistry AB Langerhans cell histiocytosis (LCH) represents a poorly defined pathologic entity characterized by diverse clinical appearence and falling into two major categories namely a restricted and an extensive disease. Since the outcome and the course of the disease is variable, we postulated that this might be reflected by the phenotype of the Langerhans cells. We have selected 11 adult restricted cases and 10 extensive childhood cases and compared the phenotype of LCH cells by immunohistochemistry on paraffin sections. Morphometric analysis indicated a significantly higher expression of histiocytic (CD68, S-100, lysozyme) markers in the adult restricted cases compared to the extensive form of the disease. Both groups were equally positive for LCH marker CD1a and negative for T cell marker CD4. On the other hand, HLA-DR expression was significantly higher in LCH cells of the extensive childhood cases suggesting higher activation. These data suggest that LCH cells have a different phenotype in the extensive childhood and restricted adult LCH where the latter is characterized by a more differentiated histiocytic phenotype. C1 [Kohalmi, Ferenc] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Strausz, Janos] National Koranyi Institute of PulmonologyBudapest, Hungary. [Egervary, Marta] National Koranyi Institute of PulmonologyBudapest, Hungary. [Szekeres, Gyorgy] Laboratory of HistopathologyPecs-Cserkut, Hungary. [Timar, Jozsef] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 184 EP 187 PG 4 ER PT J AU Honn, VK Aref, A Chen, QY Cher, M Crissman, DJ Forman, DJ Gao, X Grignon, D Hussain, M Porter, TA Pontes, E Powell, I Redman, B Sakr, W Severson, R Tang, GD Wood, PD AF Honn, V Kenneth Aref, Amer Chen, Q Yong Cher, Michael Crissman, D John Forman, D Jeffrey Gao, Xiang Grignon, David Hussain, Maha Porter, T Arthur Pontes, J. Edson Powell, Isaac Redman, Bruce Sakr, Wael Severson, Richard Tang, G Dean Wood, P David TI Prostate Cancer - Old Problems and New Approaches Part II. SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE prostate cancer; diagnosis; prognosis; pathology; biology ID prostate cancer; diagnosis; prognosis; pathology; biology AB Diagnostic and prognostic markers for prostatic cancer (PCa) include conventional protein markers (e.g., PAP, PSA, PSMA, PIP, OA-519, Ki-67, PCNA, TF, collagenase, and TIMP 1), angiogenesis indicator (e.g., factor VIII), neuroendocrine differentiation status, adhesion molecules (E-cadherin, integrin), bone matrix degrading products (e.g., ICPT), as well as molecular markers (e.g., PSA, PSMA, p53, 12-LOX, and MSI). Currently, only PSA is used clinically for early diagnosis and monitoring of PCa. The histological differential diagnosis of prostatic adenocarcinoma includes normal tissues such as Cowper's gland, paraganglion tissue and seminal vesicle or ejaculatory duct as well as pathological conditions such as atypical adenomatous hyperplasia, atrophy, basal cell hyperplasia and sclerosing adenosis. A common PCa is characterized by a remarkable heterogeneity in terms of its differentiation, microscopic growth patterns and biological aggressiveness. Most PCa are multifocal with signi ficant variations in tumor grade between anatomically separated tumor foci. The Gleason grading system which recognizes five major grades defined by patterns of neoplastic growth has gained almost uniform acceptance. In predicting the biologic behavior of PCa clinical and pathological stages are used as the major prognostic indicators. Among the cell proliferation and death regulators androgens are critical survival factors for normal prostate epithelial cells as well as for the androgen-dependent human prostatic cancer cells. The androgen ablation has been shown to increase the apoptotic index in prostatic cancer patients and castration also promotes apoptotic death of human prostate carcinoma grown in mice. The progression of PCa, similarly to other malignancies, is a multistep process, accompanied by genetic and epigenetic changes, involving phenomenons as adhesion, invasion and angiogenesis (without prostate specific features). C1 [Honn, V Kenneth] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Aref, Amer] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Chen, Q Yong] Wayne State University, Department of PathologyDetroit, USA. [Cher, Michael] Wayne State University, Department of UrologyDetroit, USA. [Crissman, D John] Wayne State University, Department of PathologyDetroit, USA. [Forman, D Jeffrey] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Gao, Xiang] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Grignon, David] Wayne State University, Department of PathologyDetroit, USA. [Hussain, Maha] Wayne State University, Department of MedicineDetroit, USA. [Porter, T Arthur] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Pontes, J. Edson] Wayne State University, Department of UrologyDetroit, USA. [Powell, Isaac] Wayne State University, Department of UrologyDetroit, USA. [Redman, Bruce] Wayne State University, Department of MedicineDetroit, USA. [Sakr, Wael] Wayne State University, Department of PathologyDetroit, USA. [Severson, Richard] Wayne State University, Department of Family MedicineDetroit, USA. [Tang, G Dean] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Wood, P David] Wayne State University, Department of UrologyDetroit, USA. RP Honn, VK (reprint author), Wayne State University, Department of Radiation Oncology, 48202 Detroit, USA. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1996 VL 2 IS 3 BP 191 EP 211 PG 21 ER PT J AU Buzas, IE Mikecz, K Glant, TT AF Buzas, I Edit Mikecz, Katalin Glant, T Tibor TI Aggrecan: A Target Molecule of Autoimmune Reactions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE proteoglycan; aggrecan; arthritis; spondylitis; autoimmune; cartilage ID proteoglycan; aggrecan; arthritis; spondylitis; autoimmune; cartilage AB Aggrecan in cartilage forms aggregates with hyaluronan and link protein, embedded in a collagen network. It accounts for the compressive stiffness and resilience of the hyaline cartilage. Many forms of inflammatory arthritis were shown to be accompanied with aggrecan degradation and loss from the cartilage. The loss of this major component of cartilage renders the tissue more vulnerable when exposed to abrasive forces. Therefore, aggrecan degradation may significantly contribute to cartilage destruction in arthritis. Furthermore, fragments of degraded aggrecan are released during joint inflammation. Thus, molecules of an avascular, immune-privileged tissue (hyaline cartilage) may become accessible to the cells of the immune system. Similarly, there is a ''leakage'' of aggrecan fragments from cartilage during aging and after joint injury, which may also lead to autosensibilisation. Autoimmune reactivity to aggrecan can be detected in human joint diseases, as well as in animal models of arthritis. The epitopes involved in these processes are currently being identified. Recent data from work with mice suggest a strong immune response focused to the N-terminal G1 domain of aggrecan that leads to arthritis and spondylitis. C1 [Buzas, I Edit] University of Medicine, The Institute of Anatomy, Histology and Embryology, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. [Mikecz, Katalin] Rush Medical University at Rush-Presbyterian-St. Luke's Medical Center, Department of BiochemistryChicago, USA. [Glant, T Tibor] Rush Medical University at Rush-Presbyterian-St. Luke's Medical Center, Department of BiochemistryChicago, USA. RP Buzas, IE (reprint author), University of Medicine, The Institute of Anatomy, Histology and Embryology, H-4012 Debrecen, Hungary. EM edit@chondron.anat.dote.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1996 VL 2 IS 4 BP 219 EP 228 PG 10 ER PT J AU Folberg, R Fleck, M Mehaffey, GM Meyer, M Bentler, ES Woolson, FR Pe'er, J AF Folberg, Robert Fleck, Margaret Mehaffey, G Mary Meyer, Margaret Bentler, E Suzanne Woolson, F Robert Pe'er, Jacob TI Mapping the Location of Prognostically Significant Microcirculatory Patterns in Ciliary Body and Choroidal Melanomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE melanoma; metastasis; prognosis; angiogenesis; vascularity; image analysis ID melanoma; metastasis; prognosis; angiogenesis; vascularity; image analysis AB The microcirculation of choroidal and ciliary body melanomas is remodeled into architecturally distinctive patterns. The presence of two histologic microvascular patterns, networks and parallel vessels with cross-linking, is strongly associated with metastasis. This study was designed to test the hypothesis that networks and parallel vessels with cross-linking patterns are not distributed evenly throughout the tumor. From a set of 234 eyes removed for ciliary body or choroidal melanoma, 152 tumors contained at least one focus of either vascular networks or parallel vessels with cross-linking. Histological cross-sections were digitized and foci of tumor containing these patterns were pseudocolorized so that their location within the periphery or central tumor zone could be mapped. Ciliary body and choroidal melanomas vary widely in size and shape and it is not appropriate to describe the periphery of a tumor as a fixed value because in a small tumor, the periphery thus defined would occupy a larger percent area than in a larger tumor. In this study, the peripheral and central zones of each tumor were described by a function that was constant from tumor to tumor, allowing the width of the peripheral and central zones to vary proportionally with tumor size. Observed counts of vascular patterns per zone were compared statistically with expected counts based upon the percent area occupied by the peripheral and central zones. Discrete foci of networks and parallel with cross-linking vessels are over-represented in the tumor periphery (p < 0.0001). C1 [Folberg, Robert] University of Iowa, Department of Ophthalmology, 100 Medical Research Center, Room 233, 52242-1182 Iowa City, IA, USA. [Fleck, Margaret] University of Iowa, Department of Computer ScienceIowa City, USA. [Mehaffey, G Mary] University of Iowa, Department of Ophthalmology, 100 Medical Research Center, Room 233, 52242-1182 Iowa City, IA, USA. [Meyer, Margaret] University of Iowa, Department of Ophthalmology, 100 Medical Research Center, Room 233, 52242-1182 Iowa City, IA, USA. [Bentler, E Suzanne] University of Iowa, Department of Preventive MedicineIowa City, USA. [Woolson, F Robert] University of Iowa, Department of Preventive MedicineIowa City, USA. [Pe'er, Jacob] Hadassah-Hebrew University, Department of OphthalmologyJerusalem, Israel. RP Folberg, R (reprint author), University of Iowa, Department of Ophthalmology, 52242-1182 Iowa City, USA. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1996 VL 2 IS 4 BP 229 EP 236 PG 8 ER PT J AU Berczi, L Tamassy, K Fekete, B Kopper, L AF Berczi, Lajos Tamassy, Klara Fekete, Bela Kopper, Laszlo TI Eosinophils and Mast Cells in Helicobacter Pylori Infected Gastric Mucosa SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE eosinophilia; gastritis; Helicobacter pylori ID eosinophilia; gastritis; Helicobacter pylori AB Although Helicobacter pylori (HP) is frequently associated with chronic active gastritis and peptic ulcers, its exact pathogenic role or the pathomechanism is still unclear. Here, we describe a striking, statistically significant increase of eosinophils in HP infected gastric mucosa compared to HP negative gastritis with similar activity. In both cases, the mean number of the mast cells in the mucosa was comparable, although the individual values showed wide distribution. The source and role of eosinophilia in HP infected mucosa, the potential link between the degree of eosinophilia and the clinical progression, as well as between eosinophils and mast cells require further study. C1 [Berczi, Lajos] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Tamassy, Klara] Semmelweis University of Medicine, Teaching HospitalBudapest, Hungary. [Fekete, Bela] Semmelweis University of Medicine, Teaching HospitalBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Berczi, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM berczi@korb1.sote.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1996 VL 2 IS 4 BP 237 EP 238 PG 2 ER PT J AU Szende, B Lubben, Th Romics, I Vass, L AF Szende, Bela Lubben, Thomas Romics, Imre Vass, Laszlo TI Apoptosis in Untreated and Hormone-Treated Prostate Cancer of Various Histological Types SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apoptosis; prostate cancer; hormone therapy ID apoptosis; prostate cancer; hormone therapy AB A total of 102 (66 untreated and 36 hormone-treated) prostate cancers were examined histologically in order to determine their histological grade and the percentage of apoptotic tumor cells. The less differentiated the tumors were, the higher the spontaneous apoptotic activity was. Hormone therapy increased the apoptotic index in the prostate cancers. The increase was of greater significance in grade I than in grade II and grade III tumors. The therapeutic consequences of these findings and the possibility of different oncogene-expressions in various histological types of prostate cancer are discussed. C1 [Szende, Bela] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Lubben, Thomas] National Institute for Rheumatology and PhysiotherapyBudapest, Hungary. [Romics, Imre] National Institute for Rheumatology and PhysiotherapyBudapest, Hungary. [Vass, Laszlo] Flor Ferenc University Hospital of Pest CountyKistarcsa, Hungary. RP Szende, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM bszende@korb1.sote.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1996 VL 2 IS 4 BP 239 EP 241 PG 3 ER PT J AU Horvath, JA Szabo, A Gaspar, T Bognar, L Tekeres, M AF Horvath, J Attila Szabo, Anita Gaspar, Tunde Bognar, Lajos Tekeres, Miklos TI M-ESLON (retard release morphine sulphate capsules) for Pain Control in Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer pain; morphine sulphate; controlled release; side effects ID cancer pain; morphine sulphate; controlled release; side effects AB The pain of patients who were in the terminal phase of advanced malignant tumors was successfully relieved by M-ESLON capsules of controlled release morphine sulphate (10-30-60-100 mg). The most frequent side effects (sickness, vomiting, obstipation) were effectively controlled. The concentration of the drug in the plasma was stable, therefore the lasting pain relief was ensured. C1 [Horvath, J Attila] University of Pecs, Medical Faculty, Department of Anesthesia and Intensive Therapy, 13 Ifjusag Rd., H-7643 Pecs, Hungary. [Szabo, Anita] University of Pecs, Medical Faculty, Department of Anesthesia and Intensive Therapy, 13 Ifjusag Rd., H-7643 Pecs, Hungary. [Gaspar, Tunde] University of Pecs, Medical Faculty, Department of Anesthesia and Intensive Therapy, 13 Ifjusag Rd., H-7643 Pecs, Hungary. [Bognar, Lajos] University of Pecs, Medical Faculty, Department of Anesthesia and Intensive Therapy, 13 Ifjusag Rd., H-7643 Pecs, Hungary. [Tekeres, Miklos] University of Pecs, Medical Faculty, Department of Anesthesia and Intensive Therapy, 13 Ifjusag Rd., H-7643 Pecs, Hungary. RP Horvath, JA (reprint author), University of Pecs, Medical Faculty, Department of Anesthesia and Intensive Therapy, H-7643 Pecs, Hungary. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1996 VL 2 IS 4 BP 242 EP 243 PG 2 ER PT J AU Orosz, Zs Kelemen, J Szentirmay, Z AF Orosz, Zsolt Kelemen, Janos Szentirmay, Zoltan TI Granular Cell Variant of Atypical Fibroxanthoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE fibrohistiocytic tumors; granular cells; atypical fibroxanthoma ID fibrohistiocytic tumors; granular cells; atypical fibroxanthoma AB We report a case of atypical fibroxanthoma of the ear in which the dominant part of the tumor has granular cell appearance. Areas identical to conventional atypical fibroxanthoma were present only at the lateral infiltrating borders. Histologically the granular cells resembled those of the classical granular cell tumors but exhibited significant pleomorphism and a high mitotic rate. Immunostains for vimentin, CD68 and NK1/C3 were positive but for S-100, HMB-45, myogenic and epithelial markers were negative. The predominance of the granular cells in an atypical fibroxanthoma supports the concept that a small subset of tumors with granular cell phenotype are of nonneural origin. C1 [Orosz, Zsolt] National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Kelemen, Janos] National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Szentirmay, Zoltan] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. RP Orosz, Zs (reprint author), National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, H-1122 Budapest, Hungary. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1996 VL 2 IS 4 BP 244 EP 247 PG 4 ER PT J AU Sinkovics, GJ AF Sinkovics, G Joseph TI Contradictory Concepts in the Etiology and Regression of Kaposi's Sarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Growth factor cascade; human herpesvirus 8; apoptosis; protooncogenes-oncogenes; retro-lentiviruses ID Growth factor cascade; human herpesvirus 8; apoptosis; protooncogenes-oncogenes; retro-lentiviruses AB The Introduction is an overview of 3 decades of works performed by Professor Ferenc Gyorkey? (in many cases in collaboration with the author) and aimed at the elucidation of viral participation in the etiology of arteriosclerosis, SLE, hairy cell leukemia, HD, AIDS and KS. Controversial issues surrounding the etiology, treatment and regression of KS are discussed in terms of paracrine and autocrine loops of growth factors; protooncogene-oncogene activations, immunosuppression and retro- and/or herpesviral etiology. In regressing KS lesions the roles played by Fas, Bcl-2, Bax, TNFß; apoptotic-antiapoptotic events; and antiangiogenesis agents especially that of Hu-r-IFNa are elaborated on. C1 [Sinkovics, G Joseph] St. Joseph's Hospital, Cancer Institute, 3001 W. MLK Jr. Blvd., 33607 Tampa, Florida, USA. RP Sinkovics, GJ (reprint author), St. Joseph's Hospital, Cancer Institute, 33607 Tampa, USA. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1996 VL 2 IS 4 BP 249 EP 267 PG 19 ER PT J AU Nagy, K Barabas, Varkonyi, V Horvath, A AF Nagy, Karoly Barabas, Eva Varkonyi, Viktoria Horvath, Attila TI Determination of HIV-1 Subtypes in Hungary by Synthetic Peptides Representing the V3 Loop of env SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE HIV-1; subtypes; Hungary ID HIV-1; subtypes; Hungary AB For the determination of HIV-1 diversity and serotyping of HIV-1 subtypes, an enzyme immunoassay was developed based on synthetic peptides representing immunodominant epitopes of the V3 loop of HIV-1 subtypes A, B, C and E, respectively. Sera from 53 asymptomatic HIV-1 infected individuals were tested for their pattern of binding reactivity to the synthetic peptides. 45/52 (85%) of the sera reacted exclusively to V3 peptide representing HIV-1 B subtypes, 4/52 (7.6%) of the sera showed cross reacticity to A/B peptides and 1/52 (1.9%) of the sera reacted with both A and C peptides. No single reactivity with subtype A or E peptides have been observed. Results together with nucleotide sequence analysis of the V3 region of clinical isolates suggest that HIV-1 infection in Hungary has been induced predominantly by strains belonging to HIV-1 subtype B. C1 [Nagy, Karoly] National Institute for Dermato-Venereology, Maria u. 41., H-1085 Budapest, Hungary. [Barabas, Eva] National Institute for Dermato-Venereology, Maria u. 41., H-1085 Budapest, Hungary. [Varkonyi, Viktoria] National Institute for Dermato-Venereology, Maria u. 41., H-1085 Budapest, Hungary. [Horvath, Attila] National Institute for Dermato-Venereology, Maria u. 41., H-1085 Budapest, Hungary. RP Nagy, K (reprint author), National Institute for Dermato-Venereology, H-1085 Budapest, Hungary. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1996 VL 2 IS 4 BP 268 EP 271 PG 4 ER PT J AU Fife, K Bower, M AF Fife, Kathryn Bower, Mark TI Current Management of AIDS Related Non Hodgkin's Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE AIDS; Non-Hodgkin's lymphoma ID AIDS; Non-Hodgkin's lymphoma AB Non Hodgkin's lymphoma is the AIDS defining illness in 3-3.5% of patients and is increasing in incidence as the survival of HIV infected people improves. The incidence of these intermediate/high grade B cell malignancies is sixty times higher than in the general population. The most important prognostic factors are a CD4 positive lymphocyte count of <100 cells/mm3, a prior AIDS defining diagnosis, an ECOG performance status >2 and primary cerebral origin. Patients with any of these factors are most likely to benefit from palliative rather than radical treatment. Good prognosis patients have a 30-40% chance of cure from their lymphoma with carefully administered intensive chemotherapy. C1 [Fife, Kathryn] Charing Cross Hospital, Medical Oncology Unit, Fulham Palace Road, W6 8RF London, UK. [Bower, Mark] Charing Cross Hospital, Medical Oncology Unit, Fulham Palace Road, W6 8RF London, UK. RP Fife, K (reprint author), Charing Cross Hospital, Medical Oncology Unit, W6 8RF London, UK. EM m.bower@cxwms.ac.uk NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1996 VL 2 IS 4 BP 272 EP 275 PG 4 ER PT J AU Honn, VK Aref, A Chen, QY Cher, LM Crissman, DJ Forman, DJ Gao, X Grignon, D Hussain, M Porter, TA Pontes, E Powell, I Redman, B Sakr, W Severson, R Tang, GD Wood, PD AF Honn, V Kenneth Aref, Amer Chen, Q Yong Cher, L Michael Crissman, D John Forman, D Jeffrey Gao, Xiang Grignon, David Hussain, Maha Porter, T Arthur Pontes, J. Edson Powell, Isaac Redman, Bruce Sakr, Wael Severson, Richard Tang, G Dean Wood, P David TI Prostate Cancer Old Problems and New Approaches Part III. SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE prostate cancer; prevention; radiotherapy; chemotherapy ID prostate cancer; prevention; radiotherapy; chemotherapy AB In Part Three of this review, we begin with an analysis of prevention strategies for prostate cancer followed by a discussion of the clinical use of molecular techniques for the evaluation and treatment of patients with clinically localized prostate cancer. New developments in neutron and photon therapy of prostate cancer are addressed as well as the use of systemic radiotherapy for the treatment of bone metastases. Finally, we conclude with the role of hormonal therapy in the treatment of prostate cancer and the current status of development of chemo therapeutic regimens for the treatment of prostate cancer. C1 [Honn, V Kenneth] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Aref, Amer] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Chen, Q Yong] Wayne State University, Department of PathologyDetroit, USA. [Cher, L Michael] Wayne State University, Department of UrologyDetroit, USA. [Crissman, D John] Wayne State University, Department of PathologyDetroit, USA. [Forman, D Jeffrey] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Gao, Xiang] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Grignon, David] Wayne State University, Department of PathologyDetroit, USA. [Hussain, Maha] Wayne State University, Department of MedicineDetroit, USA. [Porter, T Arthur] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Pontes, J. Edson] Wayne State University, Department of UrologyDetroit, USA. [Powell, Isaac] Wayne State University, Department of UrologyDetroit, USA. [Redman, Bruce] Wayne State University, Department of MedicineDetroit, USA. [Sakr, Wael] Wayne State University, Department of PathologyDetroit, USA. [Severson, Richard] Wayne State University, Department of Family MedicineDetroit, USA. [Tang, G Dean] Wayne State University, Department of Radiation Oncology, 431 Chemistry, 48202 Detroit, MI, USA. [Wood, P David] Wayne State University, Department of UrologyDetroit, USA. RP Honn, VK (reprint author), Wayne State University, Department of Radiation Oncology, 48202 Detroit, USA. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1996 VL 2 IS 4 BP 276 EP 292 PG 17 ER PT J AU Schwab, M AF Schwab, Manfred TI MYCN Amplification in Neuroblastoma: a Paradigm for the Clinical Use of an Oncogene SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE neuroblastoma; N-Myc; prediction; therapy ID neuroblastoma; N-Myc; prediction; therapy AB Increase of the dosage of cellular oncogenes by DNA amplification is a frequent genetic alteration of cancer cells. The presence of amplified cellular oncogenes is usually signalled by conspicuous chromosomal abnormalities, ''double minutes'' (DMs) or ''homogeneously staining chromsomal regions'' (HSRs). Some human cancers carry a specific amplified oncogene at high incidence. In neuroblastomas the amplification of MYCN has been found associated with aggressively growing cancers and is an indicator for poor prognosis. MYCN amplification is of predictive value for identifying neuroblastoma patiens that require specific therapeutic regimens and for identifying patients that will not benefit from chemotherapy. C1 [Schwab, Manfred] German Cancer Research Center, Division of Cytogenetics, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany. RP Schwab, M (reprint author), German Cancer Research Center, Division of Cytogenetics, D-69120 Heidelberg, Germany. EM m.schwab@dkfz-heidelberg.de NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 3 EP 7 PG 5 ER PT J AU Uusitalo, M Kivela, T AF Uusitalo, Marita Kivela, Tero TI The HNK-1 Carbohydrate Epitope and the Human Eye in Health and Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Cell adhesion; Ciliary body; Inner connective tissue layer; Natural killer cell; Retina; Subepithelial matrix cell ID Cell adhesion; Ciliary body; Inner connective tissue layer; Natural killer cell; Retina; Subepithelial matrix cell AB The HNK-1 carbohydrate epitope is part of many cell membrane and extracellular matrix molecules, several of which have been implicated in cell adhesion. It is a versatile tool in eye research. In the human eye this epitope is present in the retina, the optic and ciliary nerves, the ciliary and iris epithelia, the zonular lamella, and the sclera. It is phylogenetically conserved, but the positive cell types vary from species to species. In addition to revealing interspecies differences in the vertebrate retina, the HNK-1 epitope has been used to identify a novel cell type in the eye: the subepithelial matrix cells that reside in the inner connective tissue layer (ICTL) of the ciliary body. Although these cells resemble fibroblasts in ultrastructure, they form a distinct cell population that differs in antigenic profile from fibroblasts in other tissues. The HNK-1 epitope is also associated with the elastic fiber system of the ICTL, which may be produced by the subepithelial matrix cells. It may help to structurally stabilize the ciliary body and the retina. The HNK-1 epitope is also involved in many important eye diseases. The subepithelial matrix cells seem to be susceptible to irrreversible atrophy as a result of glaucoma, thermal injury, and tissue compression. On the other hand, the HNK-1 epitope is found in the extracellular matrix of secondary cataracts and may contribute to its pathogenesis. Finally, this epitope has proved to be useful in identifying deposits of exfoliation material, and in tracing neuroepithelial derivatives in developmental anomalies and tumors of the eye. C1 [Uusitalo, Marita] Helsinki University Central Hospital, Department of Ophthalmology, Haartmaninkatu 4C, FIN-00290 Helsinki, Finland. [Kivela, Tero] Helsinki University Central Hospital, Department of Ophthalmology, Haartmaninkatu 4C, FIN-00290 Helsinki, Finland. RP Uusitalo, M (reprint author), Helsinki University Central Hospital, Department of Ophthalmology, FIN-00290 Helsinki, Finland. EM msuusita@cc.helsinki.fi NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 8 EP 14 PG 7 ER PT J AU Papai, Zs Feja, NCh Eid, H Sztan, M Olah, E Szendroi, M AF Papai, Zsuzsa Feja, N Christina Eid, Hanna Sztan, Mariann Olah, Edit Szendroi, Miklos TI P53 Overexpression as an Indicator of Overall Survival and Response to Treatment in Osteosarcomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE osteosarcoma; p53 overexpression ID osteosarcoma; p53 overexpression AB The p53 gene located at chromosome 17p13 is found to be altered (allelic loss or other mutation) in multiple human cancers, including osteosarcomas. The mutated gene produces a protein with a prolonged half-life thus rendering it detectable by conventional immunohistochemistry. We examined the correlation between p53 expression and clinical prognosis as well as response to therapy. Twenty-one patients with previously untreated and histologically verified highly malignant osteosarcoma were used for this study. Biopsy material taken both prior to the start of COSS 91 protocol and at the time of surgery (ten weeks later) was examined for alterations in p53 protein expression and drug resistance. Two patients who had strong (+++) p53 protein expression and three others who became positive during the chemotherapy had significantly worse prognosis (all of them died within one year) than those who showed no p53 expression both at biopsy and after chemotherapy (all 11 patients are alive, average follow-up time: 3.5 years). All patients who showed any kind of positive p53 protein expression on initial biopsy were non-responders to chemotherapy. In contrast, 69% (9 out of 13) of those who exhibited no p53 expression on initial biopsy were responders or intermediate responders to chemotherapy. We concluded that p53 expression may be a useful prognostic factor in osteosarcomas. The direct correlation between p53 positive expression and resistance to therapy can help in identifying patients who are in need of a more vigorous or different chemotherapeutical protocol. C1 [Papai, Zsuzsa] National Institute of Oncology, Rath Gyorgy ut 7., H-1525 Budapest, Hungary. [Feja, N Christina] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Eid, Hanna] National Institute of Oncology, Rath Gyorgy ut 7., H-1525 Budapest, Hungary. [Sztan, Mariann] National Institute of Oncology, Rath Gyorgy ut 7., H-1525 Budapest, Hungary. [Olah, Edit] National Institute of Oncology, Rath Gyorgy ut 7., H-1525 Budapest, Hungary. [Szendroi, Miklos] Semmelweis University, Department of OrthopedicsBudapest, Hungary. RP Papai, Zs (reprint author), National Institute of Oncology, H-1525 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 15 EP 19 PG 5 ER PT J AU Benedeczky, I Nemcsok, J AF Benedeczky, Istvan Nemcsok, Janos TI Giant Mitochondria as Possible Bioindicators of Environmental Injuries in Fish Liver SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE giant mitochondria; fish liver; herbicide (paraquat); electron microscopic cytopathology ID giant mitochondria; fish liver; herbicide (paraquat); electron microscopic cytopathology AB The effect of hypoxia (80 pHg) and simultaneously applied paraquat (1,1'-dimethyl-4,4-bipyridynum dichloride) was investigated on carp liver using electron microscopic methods. The appearance of giant mitochondria was the most conspicuous alteration in the liver cells. Most of the giant mitochondria were elongated and rod-shaped, often arranged side by side forming clusters beside the nucleus. Crook-like and irregular forms also occured among giant mitochondria. The lenght of the giant mitochondria often was greater than the diameter of nucleus: namely 5-10 µm. The outer membrane of the giant mitochondria was well preserved, but inner membranes (cristae) were usually absent, and a high density matrix filled in the inner space of mitochondria. High power magnification often revealed a regular, filamentous paracristal arrangement in the dense material of the matrix. Swollen giant mitochondria with light matrix and tubular elements also occured in low number. Although fine structural characteristics of carp liver giant mitochondria are not specific for inducing agents (hypoxia + paraquat treatment) the appearance of altered giant mitochondria may be a useful signal for monitoring cell damaging enviromental xenobiotics. C1 [Benedeczky, Istvan] Attila Jozsef University, Department of BiochemistrySzeged, Hungary. [Nemcsok, Janos] Attila Jozsef University, Department of BiochemistrySzeged, Hungary. RP Benedeczky, I (reprint author), Attila Jozsef University, Department of Biochemistry, Szeged, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 20 EP 25 PG 6 ER PT J AU Polanec, J Patzer, J Grzybowski, J Strukelj, M Pavelic, PZ AF Polanec, Janja Patzer, Jan Grzybowski, Jacek Strukelj, Milan Pavelic, P Zlatko TI Amount and Avidity of IgG Antibodies to Pseudomonas Aeruginosa Exotoxin A Antigen in Cystic Fibrosis Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ELISA; avidity; cystic fibrosis; exotoxin A; Pseudomonas aeruginosa ID ELISA; avidity; cystic fibrosis; exotoxin A; Pseudomonas aeruginosa AB Amount and avidity of serum IgG antibodies to Pseudomonas aeruginosa exotoxin A in sera of 31 patients with cystic fibrosis (CF) was studied. Eight patients had P. aeruginosa isolated from the sputum on multiple occasions, while from 23 patients no P. aeruginosa was isolated. Amount of IgG antibodies to P. aeruginosa exotoxin A were significantly increased in the serum of patients with P. aeruginosa pulmonary colonization (p<0.0001). On the contrary, serum IgG avidity in the colonized and in the non-colonized CF patients was low (<10) and was statistically different when compared to the 30 age-matched healthy controls (p<0.0001). There was no change in IgG avidity in six chronically infected CF patients from whom we obtained serum samples after half a year period (p=0,55). C1 [Polanec, Janja] Institute of MicrobiologyLjubljana, Slovenia. [Patzer, Jan] Children's Memorial HospitalWarsaw, Poland. [Grzybowski, Jacek] Military Institute of MedicineWarsaw, Poland. [Strukelj, Milan] Pediatric ClinicLjubljana, Slovenia. [Pavelic, P Zlatko] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45267-0528 Cincinnati, Ohio, USA. RP Pavelic, PZ (reprint author), University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck Surgery, 45267-0528 Cincinnati, USA. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 26 EP 29 PG 4 ER PT J AU Polanec, J Pavelic, PZ Krizman, I Osredkar, J AF Polanec, Janja Pavelic, P Zlatko Krizman, Igor Osredkar, Joe TI Low Serum Pancreatitis-Associated Protein Does not Exclude Complications in Mild Acute Pancreatitis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE pancreatitis-associated protein; ELISA; acute pancreatitis ID pancreatitis-associated protein; ELISA; acute pancreatitis AB Normal serum PAP levels on admission to the hospital in patiens with acute pancreatitis has been proposed to help select the patients who are not going to develop complications. The aims of this study were, first, to assess the specificity of serum pancreatitis - associated protein (PAP) serology test and second, to evalute the usefulness of the test for prediciting complications in acute pancreatitis on admission to the hospital. The sensitivity of the PAP ELISA in patiens with acute pancreatitis on admission to the hospital was 70% and the serum PAP levels significantly higher than in healthy controls (p < 0.0001). However, the serum PAP levels in patients with acute pancreatitis were not significantly different from values in patients with various abdominal diseases (p < 0.58). Serum PAP levels gave good correlation to APACHE II (p = 0.02) and CRP (p = 0.01). Two patients with local complications (necrotizing pancreatitis, pancreatic fluid collection) had elevated serum PAP levels on admission to the hospital (> 100 ng/ml). The diagnostic specififity of PAP ELISA is low. Patients, who develop local complications in acute pancreatitis can not be excluded by normal serum PAP levels on admission to the hospital. C1 [Polanec, Janja] Institute for Clinical Chemistry and BiochemistryLjubljana, Slovenia. [Pavelic, P Zlatko] University of Cincinnati College of Medicine, Department of Otolaryngology-Head and Neck SurgeryCincinnati, USA. [Krizman, Igor] Gastroenterology Clinic, Japljeva 2, 1000 Ljubljana, Slovenia. [Osredkar, Joe] Institute for Clinical Chemistry and BiochemistryLjubljana, Slovenia. RP Krizman, I (reprint author), Gastroenterology Clinic, 1000 Ljubljana, Slovenia. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 30 EP 33 PG 4 ER PT J AU Grinevich, AJ Beres, J Bendyug, DG AF Grinevich, Akimovich Jurij Beres, Jozsef Bendyug, Dmitrijevna Galina TI A Trace Element Preparation Increases Antitumor Activity in Mice SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE trace elements; antimetastatic; immunomodulation ID trace elements; antimetastatic; immunomodulation AB A trace element preparation (Beres Drops Plus, BDP) produced immunomodulatory effects in previous in vitro and in vivo experiments. Here, C57B1/6 inbred mice were transplanted with either Lewis lung tumor or with B16 melanoma. BDP was given intraperitoneally a. before transplantation; b. after transplantation or c. after the removal of the primary tumor. As a result, BDP pretreatment could slow down the tumor progression by decreasing the number and the volume of metastases as well as the proportion of mice with metastases without influencing the growth of the primary tumors. Furthermore, BDP treatment improved the immunological activity of the tumor-bearing host, too. These preliminary data suggest that the parenteral administration of the practically non-toxic BDP could help to control tumor progression in experimental models. C1 [Grinevich, Akimovich Jurij] Ministry of Health of Ukraine, Ukrainian Research Institute of Oncology and Radiology, 33/43 Lomonosov St, 252022 Kiev, Ukraine. [Beres, Jozsef] Beres Co. Ltd., Department of Research and DevelopmentBudapest, Hungary. [Bendyug, Dmitrijevna Galina] Ministry of Health of Ukraine, Ukrainian Research Institute of Oncology and Radiology, 33/43 Lomonosov St, 252022 Kiev, Ukraine. RP Grinevich, AJ (reprint author), Ministry of Health of Ukraine, Ukrainian Research Institute of Oncology and Radiology, 252022 Kiev, Ukraine. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 34 EP 37 PG 4 ER PT J AU Uleckiene, S Giciute, L AF Uleckiene, Saule Giciute, Laima TI Carcinogenicity of Sulfuric Acid in Rats and Mice SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE sulfuric acid; carcinogenicity; rats; mice ID sulfuric acid; carcinogenicity; rats; mice AB An International Agency for Research on Cancer (IARC) committee recognized aerosol of sulphuric acid as a human carcinogen on the basis of epidemiological studies. No experimental studies on the carcinogenicity, either of sulfuric acid aerosol or of sulfuric acid itself was available. Our aim was to determine whether sulfuric acid is a causal or modifying factor in carcinogenesis, especially in the respiratory tract. We used two species of laboratory animals (both sexes) - 315 Wistar rats and 219 CBAxC57Bl mice in a long term experimental study. The rats were treated with sulfuric acid (maximal tolerated doses, by chronic intratracheal instillations or by gastric intubations) and/or benzo(a)pyrene (by intratracheal instillations). The mice were treated with sulfuric acid (by chronic gastric intubations) and/or urethane (by intraperitoneal injections). We observed the animals throughout their lives and performed gross and microscopic examination of all organs. The results of the first year of study did not provide clear evidence either for sulfuric acid carcinogenicity or for co-carcinogenicity. However, in the second year tumors appeared in those organs where sulfuric acid acted directly. A modifying (stimulating) effect of sulfuric acid on carcinogenesis induced with benzo(a)pyren was observed in rats. Sulfuric acid did not influence lung carcinogenesis induced with urethane in mice. C1 [Uleckiene, Saule] Lithuanian Oncology Center, Santariskiu 1., 2600 Vilnius, Lithuania. [Giciute, Laima] Lithuanian Oncology Center, Santariskiu 1., 2600 Vilnius, Lithuania. RP Uleckiene, S (reprint author), Lithuanian Oncology Center, 2600 Vilnius, Lithuania. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 38 EP 43 PG 6 ER PT J AU Kovesi, Gy Paloczi, K Onody, K Fekete, B AF Kovesi, Gyorgy Paloczi, Katalin Onody, Klara Fekete, Bela TI Immunologic Profile of Patients Suffering from Herpes Simplex Virus (HSV) -Associated Oral Lesions Treated with Natural Human Interferon Alpha (Egiferon) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE interferon alpha; herpes virus; lymphocyte subpopulations ID interferon alpha; herpes virus; lymphocyte subpopulations AB 10 consecutive patients with HSV-associated chronic oral lesions were treated with Egiferon for ten days. There were a statistically significant increase in the Large Granule Lymphocyte (LGL) counts and the number of spontaneous E rosette forming cells by the end of the treatment period. Interferon alpha brought about a preferential expression of CD8, CD11b, CD14, CD25 and CD45RO cell surface molecules without any effect on the expression of CD2, CD3, CD4, CD20 and HLA-DR. C1 [Kovesi, Gyorgy] Semmelweis University, Department of Oral and Maxillofacial SurgeryBudapest, Hungary. [Paloczi, Katalin] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Onody, Klara] EGIS Pharmaceuticals PLCBudapest, Hungary. [Fekete, Bela] Semmelweis University of Medicine, Teaching Hospital, Kutvolgyi ut 4., 1125 Budapest, Hungary. RP Fekete, B (reprint author), Semmelweis University of Medicine, Teaching Hospital, 1125 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 44 EP 46 PG 3 ER PT J AU Kiss, F Salamon, F Rozsahegyi, J AF Kiss, Ferenc Salamon, Ferenc Rozsahegyi, Jozsef TI Changing Pattern of Bladder Cancer Cytology (Haynal Imre University Experience) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE urinary cytology; sensitivity; transitional cell neoplasms ID urinary cytology; sensitivity; transitional cell neoplasms AB Urinary cytology reports of 151 patients with histologically verified tumors from the periods 1981-1985 and 1991-1995 were analyzed. No significant change in the overall sensitivity of tumor detection (76% and 76.8%, respectively) was found. In the group of well-differentiated (G0-G1) tumors, however, 60% of the more recent cases were cytologically positive or suspicious, 23% more than ten years ago. A decrease in the detection rate of G2 tumors in the last period (72% versus 89%) was probably caused by false negative reports due to frequent inflammatory changes in the specimens. Poorly differentiated (G3-G4) transitional cell tumors resulted in a high rate of positive cytological diagnoses (93% in both periods). In cases with negative cytology at clinical suspicion of tumor, repeated sampling increased the detection rate of G0-G1 lesions from 53% up to 60%. Optimal sampling and preparation technique, cytopathologists training and improved follow-up of patients are preconditions of sensitive and specific urinary cytology. C1 [Kiss, Ferenc] Haynal Imre University of Health Sciences, Institute of Pathology, 2nd Institute of Pathology, Ulloi ut 93.Budapest, Hungary. [Salamon, Ferenc] Haynal Imre University of Health Sciences, Institute of Pathology, 2nd Institute of Pathology, Ulloi ut 93.Budapest, Hungary. [Rozsahegyi, Jozsef] Haynal Imre University of Health Sciences, Department of UrologyBudapest, Hungary. RP Kiss, F (reprint author), Haynal Imre University of Health Sciences, Institute of Pathology, Budapest, Hungary. EM kf@korb2.sote.hu NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 47 EP 50 PG 4 ER PT J AU Kovalszky, I Nagy, OJ Gallai, M Sebestyen, A Schaff, Zs Paku, S Jeney, A Iozzo, VR AF Kovalszky, Ilona Nagy, O Julia Gallai, Monika Sebestyen, Anna Schaff, Zsuzsa Paku, Sandor Jeney, Andras Iozzo, V Renato TI Altered Proteoglycan Gene Expression in Human Biliary Cirrhosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE proteoglycan; biliary cirrhosis; decorin; perlecan; syndecan; GAG ID proteoglycan; biliary cirrhosis; decorin; perlecan; syndecan; GAG AB Proteoglycans play key roles in the physiological assembly of extracellular matrices and in the modulation of growth factor activities. During liver regeneration there is a profound remodelling of the connective tissue network with a concurrent alteration in proteoglycan gene expression. In the present study we have analyzed in detail the biochemical and molecular properties of the proteoglycans associated with biliary cirrhosis. The three major proteoglycans of human liver, namely decorin, syndecan and perlecan, were markedly elevated in the cirrhotic parenchyma as compared to normal liver tissue. Particularly elevated (eight fold) was the perlecan. This proteoglycan had not only heparan sulfate but also chondroitin and dermatan sulfate. Reverse transcriptase PCR revealed a marked enhancement of decorin and syndecan expression and detectable message for perlecan was found only in the cirrhotic liver. These results indicate that significant proteoglycan alterations are associated with the development of biliary cirrhosis and provide basis for future studies aimed at the characterization of the molecular events involved in the regulation of extracellular matrix deposition in this common human disease. C1 [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Nagy, O Julia] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Gallai, Monika] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Paku, Sandor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Jeney, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Iozzo, V Renato] Thomas Jefferson UniversityPhiladelphia, USA. RP Kovalszky, I (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 51 EP 58 PG 8 ER PT J AU Bakos, N Krasznai, G Begany, AF Bakos, Noemi Krasznai, Geza Begany, Agnes TI Erythema Gyratum Repens an Immunological Paraneoplastic Dermatosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Erythema gyratum repens; Paraneoplastic dermatosis; Immunfluorescence ID Erythema gyratum repens; Paraneoplastic dermatosis; Immunfluorescence AB The authors present a patient with erythema gyratum repens who had a bronchogenic carcinoma. Autoantibodies and complement at the basement membrane zone of the skin was found which suggest that erythema gyratum repens may have an immunological pathogenesis but the nature of the antigen should be further characterised. C1 [Bakos, Noemi] Hetenyi Geza Hospital, Department of Dermatology, Toszegi u. 21., 5000 Szolnok, Hungary. [Krasznai, Geza] Hetenyi Geza County Hospital, Department of PathologySzolnok, Hungary. [Begany, Agnes] University of Debrecen, Department of DermatologyDebrecen, Hungary. RP Bakos, N (reprint author), Hetenyi Geza Hospital, Department of Dermatology, 5000 Szolnok, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 59 EP 61 PG 3 ER PT J AU Jassoy, Ch Heinkelein, M Sopper, S AF Jassoy, Christian Heinkelein, Martin Sopper, Sieghart TI The Role of the Envelope Glycoprotein in the Depletion of T Helper Cells in Human Immunodeficiency Virus Infection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE HIV; AIDS; envelope; T helper lymphocyte; pathogenesis; apoptosis ID HIV; AIDS; envelope; T helper lymphocyte; pathogenesis; apoptosis AB Infection with the human immunodeficiency virus (HIV) causes gradual depletion of CD4+ T helper lymphocytes and destruction of the lymphoid tissue, which ultimately leads to a fatal defect of the cellular immune system. Paramount to the understanding of the pathogenesis of HIV infection is to elucidate the mechanism which underlies the loss of T helper cells. Various ideas have been proposed in order to explain this issue. Several hypotheses have focused on the role of the envelope glycoprotein in this process. This review summarizes the data obtained and concepts proposed regarding the involvement of the HIV glycoprotein in the pathology of CD4+ T cell depletion. C1 [Jassoy, Christian] Julius-Maximilians University, Institute for Virology and Immunobiology, Versbacher Strasse 7, D-97078 Wurzburg, Germany. [Heinkelein, Martin] Julius-Maximilians University, Institute for Virology and Immunobiology, Versbacher Strasse 7, D-97078 Wurzburg, Germany. [Sopper, Sieghart] Julius-Maximilians University, Institute for Virology and Immunobiology, Versbacher Strasse 7, D-97078 Wurzburg, Germany. RP Jassoy, Ch (reprint author), Julius-Maximilians University, Institute for Virology and Immunobiology, D-97078 Wurzburg, Germany. EM viro023@rzbox.uni-wuerzburg.de NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 62 EP 67 PG 6 ER PT J AU Barabas, Falus, A Nagy, K Varkonyi, V Temesvari, E Horvath, A AF Barabas, Eva Falus, Andras Nagy, Karoly Varkonyi, Viktoria Temesvari, Erzsebet Horvath, Attila TI The Dominant T-Helper Lymphocyte Function of HIV Infected Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE HIV; Th1; Th2; RT-PCR ID HIV; Th1; Th2; RT-PCR AB In HIV infection, the decrease in the number and functional activity of lymphocytes is accompanied by atopia and an increased level of total IgE and some specific IgE antibodies. This could be explained by the Th2 dominance induced by HIV replication and so a Th1-Th2 switch could have prognostic value. We investigated the characteristic T-helper phenotype dominance and its relationship to cytokine expression and IgE immune response in the early stage of asymptomatic HIV infection. In the separated lymphocytes of i. asymptomatic HIV positive persons; ii. HIV negative homosexuals; iii. atopic patients; and iv. healthy controls, expression of mRNA for IFNg (Th1) and IL-10 (Th2) were determined by semiquantitative RT-PCR. The serum level of antibodies for HIV 1/2 and total/specific IgE were also determined. Transcription of mRNA of IFNg and IL-10 were more pronounced in HIV positive and atopic groups than in the healthy control, without lymphocyte phenotype dominance. In HIV negative persons, however, a significant Th2 dominance was detected. There was no significant difference in the IgE level between the 4 investigated groups. In the HIV positive cases, IL-10 expression and total serum IgE do not support a switch to Th2 dominance. In the atopic group, aside from the total IgE level, down regulation of IFNg was not observed. These results suggest a general activation of the immune system in the early stage of HIV infection. C1 [Barabas, Eva] National Institute for Dermato-Venereology, Maria str. 41, H-1085 Budapest, Hungary. [Falus, Andras] Semmelweis University of Medicine, Biological InstituteBudapest, Hungary. [Nagy, Karoly] National Institute for Dermato-Venereology, Maria str. 41, H-1085 Budapest, Hungary. [Varkonyi, Viktoria] National Institute for Dermato-Venereology, Maria str. 41, H-1085 Budapest, Hungary. [Temesvari, Erzsebet] National Institute for Dermato-Venereology, Maria str. 41, H-1085 Budapest, Hungary. [Horvath, Attila] National Institute for Dermato-Venereology, Maria str. 41, H-1085 Budapest, Hungary. RP Horvath, A (reprint author), National Institute for Dermato-Venereology, H-1085 Budapest, Hungary. EM aurelius@bor.sote.hu NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 68 EP 73 PG 6 ER PT J AU Casabona, J Gambus, G Vall, M Rodes, A AF Casabona, Jordi Gambus, Gemma Vall, Marti Rodes, Anna TI Risk Factors for AIDS Associated Kaposi's Sarcoma An European Multicentre Case Control Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE AIDS; Kaposi's sarcoma; risk factors; case-control study ID AIDS; Kaposi's sarcoma; risk factors; case-control study AB EURO-SHAKS, European study on HIV associated Kaposi's sarcoma, is a BIOMED 1 project financed by the European Union (DG XII). The Spanish side of the project has been financed by FISS (Fondo de Investigaciones Sanitarias). The aims of this study are to identify possible genetic, behavioural, biological and environmental risk factors for HIV associated Kaposi's sarcoma through a multicentre case-control study. An extensive personal questionnaire, a clinical data form and blood sample is required from all participants. In addition, a cutaneous biopsy is request from KS patients. The presence of several European groups in this project implies a large and diverse sample size and will allow to correlate the behaviour, clinical and biological data in different geographical areas, and therefore study the possible transmission routes as well as the natural history of the putative causal agent of KS. One of the main objectives of EURO-SHAKS is to create a European Bank of AIDS biological samples for possible future investigations. C1 [Casabona, Jordi] Hopital Universitari Germans Trias i Pujol. Crta Canyet, 08916 Badalona, Spain. [Gambus, Gemma] Hopital Universitari Germans Trias i Pujol. Crta Canyet, 08916 Badalona, Spain. [Vall, Marti] Hopital Universitari Germans Trias i Pujol. Crta Canyet, 08916 Badalona, Spain. [Rodes, Anna] Hopital Universitari Germans Trias i Pujol. Crta Canyet, 08916 Badalona, Spain. RP Casabona, J (reprint author), Hopital Universitari Germans Trias i Pujol. Crta Canyet, 08916 Badalona, Spain. EM jcasabona@ceescat.hugtip.scs.es NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 1997 VL 3 IS 1 BP 74 EP 77 PG 4 ER PT J AU Hagmann, W AF Hagmann, Wolfgang TI 12-Lipoxygenase in Human Tumor Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE epidermal growth factor; 12-lipoxygenase; metastasis; nuclear translocation; tumor cells ID epidermal growth factor; 12-lipoxygenase; metastasis; nuclear translocation; tumor cells AB Tumor cell proliferation and metastasis proceed via a network of interdependent molecular events with a vast array of molecular players and signal transduction mechanisms differing in various types of human tumors. In the sequence of events necessary for carcinogenesis, arachidonate metabolites have been documented to play a significant role at several steps. Arachidonate metabolism in human cells occurs via several enzymatic pathways, including enzymes such as cyclo-oxygenases and lipoxygenases. This review pays particular attention to one member of the lipoxygenase family of enzymes, namely 12-lipoxygenase, since an arachidonate metabolite generated via 12-lipoxygenase action, 12(S)-HETE, has been shown to elicit various prometastatic effects of tumor cells in vivo and in vitro. We focus especially on mechanisms of activation and modulation of 12-lipoxygenase expression in human tumor cells, since various tumor cells express 12-lipoxygenase or are responsive to metabolites derived from 12-lipoxygenase action, thus offering a potential for successful therapeutic intervention against such tumors. C1 [Hagmann, Wolfgang] Deutsches Krebsforschungszentrum, Division of Tumor Biochemistry, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany. RP Hagmann, W (reprint author), Deutsches Krebsforschungszentrum, Division of Tumor Biochemistry, D-69120 Heidelberg, Germany. EM W.Hagmann@DKFZ-Heidelberg.de NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 83 EP 88 PG 6 ER PT J AU Lah, TT Kos, J Blejec, A Frkovic-Georgio, S Golouh, R Vrhovec, I Turk, V AF Lah, T Tamara Kos, Janko Blejec, Andrej Frkovic-Georgio, Snezana Golouh, Rastko Vrhovec, Ivan Turk, Vito TI The Expression of Lysosomal Proteinases and Their Inhibitors in Breast Cancer: Possible Relationship to Prognosis of the Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; cathepsin B; cathepsin D; cathepsin L; lysosomal enzymes; metastasis; prognostic factors; stefins ID breast cancer; cathepsin B; cathepsin D; cathepsin L; lysosomal enzymes; metastasis; prognostic factors; stefins AB Proteolytic enzymes have been proposed as new biological prognostic indicators to facilitate decisions about treatment of breast cancer patients following surgery. We reported earlier that the activities of cysteine proteinases (CP), cathepsin (Cat) B and cathepsin (Cat) L and the expression of stefin A might be associated with breast tumor progression and prognosis. Here, the protein concentrations of Cats D, B and L and stefin A have been measured in a series of 60 matched pairs of breast tumours and control adjacent tissues, using ELISAs developed in our laboratory. Median tumor concentrations of Cat D (47 pm/mg), Cat B (222 ng/mg) and Cat L (88 ng/mg) were significantly (p<0.0005) increased by 7 fold, 27 fold and 6 fold, respectively. Much greater increases in the activities of Cat B (63 fold) and of Cat L (274 fold) were found, indicating enhanced activation of cysteine proteinases in tumors, due either to proteolytic activation of proCat B and proCat L and/or to a decrease in specific endogenous cystatins. However, the 1.6-fold decreased (p<0.0001) levels of inhibition by cystatins could not be entirely responsible for more than 100-fold increased ratio of CP:cystatins activity. Moreover, stefin A was either increased or decreased in tumor samples, resulting in a 1.4-fold median increase in tumors. Comparing the biological parameters with the established histo-pathological prognosticators, we found that the increased protein concentration of Cat B was associated with lymph node involvement (p<0.009) and higher stage (p<0.003), and both Cat B and Cat L activities were more increased in high grade tumours (p<0.05). Survival analysis revealed that stefin A was the most significant prognostic factor for disease-free (p<0.008) and overall survival (p<0.02), followed by increased Cat B activity and protein concentration. Cat L was of borderline significance while Cat D was not significant for prognosis. We conclude that enhanced activation of CP, due partially to an imbalance between cysteine proteinases and inhibitors is linked to the progression of breast cancer. Larger sample size is needed to confirm the prognostic significance of stefin A, Cat B and Cat L. C1 [Lah, T Tamara] National Institute of Biology, Laboratory of Molecular Biology and Biochemistry, Vecna pot 111, 1000 Ljubljana, Slovenia. [Kos, Janko] National Institute of Biology, Laboratory of Molecular Biology and BiochemistryLjubljana, Slovenia. [Blejec, Andrej] National Institute of Biology, Laboratory of Molecular Biology and Biochemistry, Vecna pot 111, 1000 Ljubljana, Slovenia. [Frkovic-Georgio, Snezana] Institute of Oncology, Department of PathologyLjubljana, Slovenia. [Golouh, Rastko] Institute of Oncology, Department of PathologyLjubljana, Slovenia. [Vrhovec, Ivan] Institute of Oncology, Department of PathologyLjubljana, Slovenia. [Turk, Vito] National Institute of Biology, Laboratory of Molecular Biology and BiochemistryLjubljana, Slovenia. RP Lah, TT (reprint author), National Institute of Biology, Laboratory of Molecular Biology and Biochemistry, 1000 Ljubljana, Slovenia. EM tamara.lah@ijs.si NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 89 EP 99 PG 11 ER PT J AU Mihalik, R Uher, F Petak, I Sebestyen, A Kopper, L AF Mihalik, Rudolf Uher, Ferenc Petak, Istvan Sebestyen, Anna Kopper, Laszlo TI Regulation of Differentiation, Proliferation and Drug-Induced Apoptosis in HT58 Lymphoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apoptosis; lymphoma; differentiation; etoposide; staurosporine; PMA ID apoptosis; lymphoma; differentiation; etoposide; staurosporine; PMA AB Recently, it has been suggested, that differentiated cells are more resistant to the apoptotic effect of DNA damaging agents possibly due to the decreased activity of ''damage detecting / apoptosis triggering'' mechanism. Previously, we have shown, that PMA pretreatment reduced etoposide- (ETO) but enhanced staurosporine- (STA) -induced apoptosis in HT58 cells. Data presented here show that the HT58 human, ''mature'' B-lymphoma cells exposed to PMA secrete more IgM into the supernatant indicating commitment of cells to perform differentiated function. The sensitivity of HT58 cells to ETO- or STA-induced apoptosis is influenced diversely with PMA pre- or posttreatment. Interestingly, the DNA damage (gamma radiation, bleomycin, ETO) or okadaic acic (30 nM) reduced the [PMA+STA] - induced apoptosis. C1 [Mihalik, Rudolf] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Uher, Ferenc] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Petak, Istvan] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM kopper@korb1.sote.hu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 100 EP 105 PG 6 ER PT J AU Kapustin, IS Popova, IT Lyschov, AA Togo, VA Abdulkadyrov, MK Blinov, NM AF Kapustin, I Sergey Popova, I Tamara Lyschov, A Anton Togo, V Alexander Abdulkadyrov, M Kudrat Blinov, N Michail TI HLA-DR2 Frequency Increase in Severe Aplastic Anemia Patients is Mainly Attributed to the Prevalence of DR15 Subtype SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE aplastic anemia; HLA-DR antigens; DNA extraction ID aplastic anemia; HLA-DR antigens; DNA extraction AB The association between severe aplastic anemia (AA) and DR2 antigen seems to be well established. However, since discrimination between two DR2-associated splits, namely DR15 and DR16, rarely was performed, it remains unclear whether one or both of these subvariants are responsible for AA susceptibility. In this study, we have analyzed the HLA-DR allelic distribution in a group of 37 AA patients of slavic origin from North-Western Russia. The experimental design included PCR-based amplification of DRB-specific sequences, followed by reverse dot-blot hybridization of the biotinylated PCR-product with the set of sequence-specific oligonucleotide probes. HLA-DRB alleles were identified by non-radioactive enzymatic reaction, then standard serological specificities of HLA-DR antigen were estimated according to the WHO nomenclature. Whereas DR15 subtype occurred more often in the patients (23.0% vs. 13.3%, p< 0.05), DR16 split did not show the same tendency. The results, show the overall predominance of HLA-DR2 specificity (DR15+DR16) did not reach statistical significance (24.4% vs.17.5%, p<0.2). Thus, we conclude that repeatedly reported DR2 frequency increase in AA patients is mainly attributed to the prevalence of DR15 subtype. C1 [Kapustin, I Sergey] Institute of Haematology and Transfusiology, Laboratory of Biochemistry, 2nd Sovietskaya st. 16, 193024 Saint-Petersburg, Russian Federation. [Popova, I Tamara] Institute of Haematology and Transfusiology, Laboratory of Biochemistry, 2nd Sovietskaya st. 16, 193024 Saint-Petersburg, Russian Federation. [Lyschov, A Anton] NN Petrov Institute of Oncology, Department of Molecular OncologySaint-Petersburg, Russian Federation. [Togo, V Alexander] NN Petrov Institute of Oncology, Department of Molecular OncologySaint-Petersburg, Russian Federation. [Abdulkadyrov, M Kudrat] Institute of Haematology and Transfusiology, Laboratory of Biochemistry, 2nd Sovietskaya st. 16, 193024 Saint-Petersburg, Russian Federation. [Blinov, N Michail] Institute of Haematology and Transfusiology, Laboratory of Biochemistry, 2nd Sovietskaya st. 16, 193024 Saint-Petersburg, Russian Federation. RP Blinov, NM (reprint author), Institute of Haematology and Transfusiology, Laboratory of Biochemistry, 193024 Saint-Petersburg, Russian Federation. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 106 EP 108 PG 3 ER PT J AU Cserni, G AF Cserni, Gabor TI Changes in Benign to Malignant Ratio of Surgically Treated Breast Diseases in a District Hospital SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE benign to malignant ratio; breast; fine-needle aspiration cytology; mammography ID benign to malignant ratio; breast; fine-needle aspiration cytology; mammography AB The benign to malignant ratio (BMR) of open surgical biopsies is often used to monitor the efficacy of diagnostic workup of breast lesions. Avoiding the unnecessary removal of benign lesions is of recognized importance. Histopathology archives of the Department of Pathology of the Bacs-Kiskun County Hospital were retrieved for breast lesions and the BMR of surgical specimens was determined for each year between the period of 1965-1996. The introduction of mammography and especially fine-needle aspiration cytology was paralleled by a reduction in the benign to malignant ratio from 1.7 to 0.7. Only the introduction of breast aspiration cytology seemed to have a significant effect on the BMR, but the more adequate diagnostic approach to breast lesions (mostly palpable in their nature) was in part masqueraded by the late shift in attitude of both surgeons and patients towards breast lumps. This is why the BMR can give a basic information on preoperative diagnostic workup of breast lesions, but in itself it is not able to monitor them. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., H-6000 Kecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 109 EP 114 PG 6 ER PT J AU Sztan, M Papai, Zs Szendroi, M Van Der Looij, M Olah, E AF Sztan, Marianna Papai, Zsuzsa Szendroi, Miklos Van Der Looij, Marco Olah, Edit TI Allelic Losses from Chromosome 17 in Human Osteosarcomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chromosome 17; TP53; tumor suppressor gene; loss of heterozygosity; osteosarcoma ID chromosome 17; TP53; tumor suppressor gene; loss of heterozygosity; osteosarcoma AB Genetic alterations of chromosome 17 have been reported to occur frequently both in human sporadic and familial malignancies. The present study was undertaken to explore the possible involvement of chromosome 17 genes including TP53 and the breast cancer susceptibility BRCA1 tumor suppressor genes in the development of sporadic osteogenic sarcoma. Fifteen patients were screened by polymerase chain reaction (PCR) for loss of heterozygosity (LOH) using four highly polymorphic markers. Loss of heterozygosity at the TP53 locus was detected in 40% (6/15) of informative cases while it was 14% (2/14) at the locus of thyroid hormone receptor alpha (THRA1), 21% (3/14) at the D17S855 locus intragenic to BRCA1 and 27% (4/15) at the D17S579 locus. In 53% of the cases studied at least one locus on chromosome 17 was affected by LOH. In our panel, the overall LOH frequency on 17p and 17q was observed to be 40% (6/15) and 27% (4/15), respectively. Comparison of LOH frequencies with clinical and prognostic features revealed significant correlation only with tumor recurrence. Our results confirm that the role of the TP53 tumor suppressor gene is important in the pathogenesis of sporadic osteosarcoma and suggest that 17q12-21 region abnormalities may be involved in the development and/or progression of this tumor. C1 [Sztan, Marianna] National Institute of Oncology, Department of Biochemistry, Rath Gyorgy u. 7-9., H-1525 Budapest, Hungary. [Papai, Zsuzsa] National Institute of Oncology, Department of Chemotherapy and ClinicopharmacologyBudapest, Hungary. [Szendroi, Miklos] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Van Der Looij, Marco] National Institute of Oncology, Department of Biochemistry, Rath Gyorgy u. 7-9., H-1525 Budapest, Hungary. [Olah, Edit] National Institute of Oncology, Department of Biochemistry, Rath Gyorgy u. 7-9., H-1525 Budapest, Hungary. RP Olah, E (reprint author), National Institute of Oncology, Department of Biochemistry, H-1525 Budapest, Hungary. EM e.olah@oncol.hu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 115 EP 120 PG 6 ER PT J AU Tsyplakov, ED Petrov, VS Kulagin, NR AF Tsyplakov, E Dmitry Petrov, V Semion Kulagin, N Roman TI Lymph Node Reaction to Cancer (Immunohistochemical and Ultrastructural Study) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lymph node; cancer; immunohistochemistry ID lymph node; cancer; immunohistochemistry AB A total of 153 regional lymph nodes obtained from 50 patients, operated for gastric, lung, breast, colonic and cervical cancers, were studied. Immunohistochemical methods were used to detect different markers and enzymes (CD1, CD2, CD3, CD4, CD8, CD20, CD30, CD35, CD45, l light Ig chain, lysozyme (muramidase), a-1-antichymotrypsin, protein S100 and FVIIIR). Results indicate that failure of local immunity is explained by the followings: 1. decrease in the total number of T-cells (suppressors as well as helpers); 2. high number of B-cells, plasmoblasts and antibody-forming plasmocytes, know to be able to block the cytotoxic T cells; 3. decrease in the number of incoming free phagocytes of monocytic origin and reduction in the phagocytic activity of fixed macrophages (sinus histiocytes); 4. high functional activity of dendritic reticulum cells; 5. non-handled stimulation of T cell response by the paracortical interdigitating reticulum cells; 6. reduction in area of postcapillary venules and impairment of lymphocyte recirculation through them. C1 [Tsyplakov, E Dmitry] Medical University, Department of Pathology, Butlerov str 49, 420012 Kazan, Russian Federation. [Petrov, V Semion] Kazan Cancer CenterKazan, Russian Federation. [Kulagin, N Roman] Medical University, Department of Pathology, Butlerov str 49, 420012 Kazan, Russian Federation. RP Tsyplakov, ED (reprint author), Medical University, Department of Pathology, 420012 Kazan, Russian Federation. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 121 EP 125 PG 5 ER PT J AU Jablonska, E Piotrowski, L Grabowska, Z AF Jablonska, Ewa Piotrowski, Leszek Grabowska, Zyta TI Serum Levels of IL-1b, IL-6, TNF-a, sTNF-RI and CRP in Patients with Oral Cavity Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE oral cancer; IL-1b; IL-6; TNF-a; sTNF-RI; CRP; SCC ID oral cancer; IL-1b; IL-6; TNF-a; sTNF-RI; CRP; SCC AB Pro-inflammatory cytokines, such as interleukin-1b (IL-1b), interleukin-6 (IL-6) and tumor necrosis factor- (TNF-a) play an essential role in the regulation of immune response to, and may have prognostic significance in, cancer. The aim of this study was to examine the relationship between the serum levels of IL-1b, IL-6 and TNF-a as well as the concentrations of soluble TNF receptor I (sTNF-RI) and C-reactive protein (CRP) in patients with squamous cell carcinoma of oral cavity. Results obtained were confronted with squamous cell carcinoma antigen (SCC) concentrations. IL-1b IL-6 and TNF-a serum levels as well as sTNF-RI and CRP concentrations were higher in patients than in controls. The increased serum levels appeared to be related to the clinical stage of disease. There was a correlation between IL-1b and sTNF-RI. IL-6 and IL-1b correlated with CRP levels. The mean concentrations of SCC were also elevated. IL-6 and sTNF-RI seemed to be the most sensitive parameters in early stages and may be used as additional markers in oral cancer. C1 [Jablonska, Ewa] Medical Academy, Department of Immmunopathology, Kilinski 1., 15-230 Bialystok, Poland. [Piotrowski, Leszek] Medical Academy, Deparment of Oral and Maxillofacial SurgeryBialystok, Poland. [Grabowska, Zyta] Medical Academy, Department of Immmunopathology, Kilinski 1., 15-230 Bialystok, Poland. RP Jablonska, E (reprint author), Medical Academy, Department of Immmunopathology, 15-230 Bialystok, Poland. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 126 EP 129 PG 4 ER PT J AU Ibanez-Manlapaz, GI McCoy, D Vincent, IV Ule, JU AF Ibanez-Manlapaz, G Irene McCoy, David Vincent, III Vincent Ule, Jo Ulla TI Malignant Mixed Mullerian Tumor of the Extra-genital Coelomic Epithelium: Report of Two Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE extra-genital mullerian tumor; carcinosarcoma; poor prognosis; female peritoneum ID extra-genital mullerian tumor; carcinosarcoma; poor prognosis; female peritoneum AB Malignant mixed mullerian tumors of the extra-genital coelomic epithelium (female peritoneum) are rare. Since the first case report in 1955, only nineteen have been described. In our Case 1 a 58 year-old G3P3 (gravidity = 3; parity = 3) white female with mixed mullerian tumor, homologous type, involving the abdominal peritoneum was treated with surgery and chemotherapy (doxorubicin hydrochloride and cis-platinum). She died of the disease 20 months after initial surgery. Case 2 is a 75 year-old G0P0 (gravidity = 0; parity = 0) white female with mixed mullerian tumor containing heterologous elements involving the pelvic peritoneum who was treated with surgery and chemotherapy (ifosfamide with mesna). She relapsed 6 months after surgery and refused any further treatment. She died 2 weeks later. These cases support the fact that malignant mixed mullerian tumor of the female peritoneum is rare and usually affects elderly females. It has poor prognosis and among the 15 reported cases with follow-up indicating time and presence of disease at death, only 5 survived more than 12 months after initial surgery. C1 [Ibanez-Manlapaz, G Irene] St. Frances Cabrini Hospital, Department of Pathology, 3330 Masonic Drive, 71301 Alexandria, Alexandria, USA. [McCoy, David] St. Frances Cabrini Hospital, Department of SurgeryAlexandria, Alexandria, USA. [Vincent, III Vincent] St. Frances Cabrini Hospital, Department of Obstetrics & GynecologyAlexandria, Alexandria, USA. [Ule, Jo Ulla] St. Frances Cabrini Hospital, Department of MedicineAlexandria, Alexandria, USA. RP Ibanez-Manlapaz, GI (reprint author), St. Frances Cabrini Hospital, Department of Pathology, 71301 Alexandria, USA. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 130 EP 134 PG 5 ER PT J AU Varkonyi, J Jakab, L Zalatnay, A Nagy, P Vamos, R Szombathy, T AF Varkonyi, Judit Jakab, Lajos Zalatnay, Attila Nagy, Peter Vamos, Rita Szombathy, Tamas TI Polychondritis Terminating in Eosinophilic Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE relapsing polychondritis; vitiligo; myelodysplasia; eosinophilia; eosinophil leukemia ID relapsing polychondritis; vitiligo; myelodysplasia; eosinophilia; eosinophil leukemia AB We report here on a patient presenting with Relapsing Polychondritis (RP) two years before the diagnosis of Myelodysplasia (MDS) terminating in Eosinophilic Leukemia (EoL). The evolution of RP several years prior to the presentation of MDS does not support a paraneoplastic etiology of RP in this patient. The terminal development of EoL in our case is assumed to represent clonal evolution caused by a second mutagenic event. The existence of autoimmune skin disorders in both the patient and his offspring (vitiligo and subacute cutaneous lupus erythematosus, respectively) implies that the coexistence of MDS and RP may have been caused by a functional disturbance of the immune system. C1 [Varkonyi, Judit] Semmelweis University, 3rd Department of Internal Medicine, 12 Eotvos ut, H-1121 Budapest, Hungary. [Jakab, Lajos] Semmelweis University, 3rd Department of Internal Medicine, 12 Eotvos ut, H-1121 Budapest, Hungary. [Zalatnay, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Nagy, Peter] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Vamos, Rita] Semmelweis University, Department of OphthalmologyBudapest, Hungary. [Szombathy, Tamas] Semmelweis University, 3rd Department of Internal Medicine, 12 Eotvos ut, H-1121 Budapest, Hungary. RP Varkonyi, J (reprint author), Semmelweis University, 3rd Department of Internal Medicine, H-1121 Budapest, Hungary. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 135 EP 138 PG 4 ER PT J AU Vadasz, G Sapi, Z Erdei, M Lovey, Gy Bodo, M AF Vadasz, Gizella Sapi, Zoltan Erdei, Mihaly Lovey, Gyorgy Bodo, Miklos TI Spontaneously curing anaplastic carcinoma in the lymph node SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE anaplastic carcinoma; spontaneous regression ID anaplastic carcinoma; spontaneous regression AB A well documented case of a spontaneously curing anaplastic carcinoma in lymph node is presented with a 16 year follow up. Reevaluation and detailed immunohistochemical examination confirmed the original diagnosis of anaplastic carcinoma. This is the first report of a spontaneously curing anaplastic carcinoma which raises the following questions: Was the tumor in the axillary lymph node a metastasis or a primary tumor? Does the anaplastic carcinoma demonstrate the same spontaneous regression characteristics as for example the neuroblastoma? C1 [Vadasz, Gizella] St John's Hospital, Department of Pathology, Diosarok u. 1., H-1125 Budapest, Hungary. [Sapi, Zoltan] St John's Hospital, Department of Pathology, Diosarok u. 1., H-1125 Budapest, Hungary. [Erdei, Mihaly] General PractitionerBudapest, Hungary. [Lovey, Gyorgy] St. Francis Hospital, Department of SurgeryBudapest, Hungary. [Bodo, Miklos] St John's Hospital, Department of Pathology, Diosarok u. 1., H-1125 Budapest, Hungary. RP Vadasz, G (reprint author), St John's Hospital, Department of Pathology, H-1125 Budapest, Hungary. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 139 EP 141 PG 3 ER PT J AU Tozser, J AF Tozser, Jozsef TI Specificity of Retroviral Proteinases Based on Substrates Containing Tyrosine and Proline at the Site of Cleavage SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE proteinases; retroviral; HIV; cleavage; specificity ID proteinases; retroviral; HIV; cleavage; specificity AB The retroviral proteinase (PR) plays crucial roles in the viral life cycle, therefore it is a target for chemotherapy. However, resistance rapidly develops due to frequent mutations. Studies to determine the common features of the specificity of different retroviral PRs may help to design broad spectrum inhibitors and reduce the possibility of viable mutants. We have studied the specificity of various retroviral proteinases including those the PR of HIV-1, HIV-2, equine infectious anemia virus and avian myeloblastosis virus using oligopeptide substrates. A series of oligopeptides containing substitutions in a sequence Val-Ser-Gln-Asn-Tyr*Pro-Ile-Val-Gln (asterisk indicates the site of cleavage) representing a naturally occurring cleavage site in HIV-1 was used to characterize the seven substrate binding subsites of the enzymes. The unsubstituted substrate is a typical class 1 cleavage site substrate containing an aromatic amino acid and a proline residue at the site of cleavage. The largest differences in kinetics of substrate hydrolysis were obtained with peptides containing substitutions of the Ser and Asn residues. Detailed analysis of the results by molecular modeling and comparison with previously reported data revealed the common characteristics of the specificity of the PRs as well as its strong dependence on the sequence context of the substrate. However, molecular modeling in many cases provided explanation for the sequence context dependence. Also, comparison of the specificity of the enzymes suggests that the specificity of HIV-1 and -2 PRs is rather exceptional preferring hydrophilic residues at the most discriminative positions while other PRs prefer hydrophobic residues. C1 [Tozser, Jozsef] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular Biology, Nagyerdei krt. 98., H-4012 Debrecen, Hungary. RP Tozser, J (reprint author), University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular Biology, H-4012 Debrecen, Hungary. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 142 EP 146 PG 5 ER PT J AU Aszalos, A Eckhardt, S AF Aszalos, Adorjan Eckhardt, Sandor TI Molecular Events as Targets of Anticancer Drug Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE cancer; molecular targets; chemotherapy ID cancer; molecular targets; chemotherapy AB The aim of this review is to introduce some molecular targets for cancer chemotherapy, with comments on their mode of action, preclinical and clinical results. The representatives of the following groups are covered: phosphorylation inhibitors, protein kinase modulators, receptor antagonists, immunomodulators, differentiating agents, multidrug resistance modulation, telomerase inhibitors, and bioreductive agents. C1 [Aszalos, Adorjan] Food and Drug Administration, 200 C Street, 20204 Washington, SW, USA. [Eckhardt, Sandor] National Institute of OncologyBudapest, Hungary. RP Aszalos, A (reprint author), Food and Drug Administration, 20204 Washington, USA. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 1997 VL 3 IS 2 BP 147 EP 158 PG 12 ER PT J AU Schweighoffer, T AF Schweighoffer, Tamas TI Molecular Cancer Vaccines: Tumor Therapy Using Antigen-Specific Immunizations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE vaccines; molecular; immunotherapy; tumor ID vaccines; molecular; immunotherapy; tumor AB Vaccination against tumors promises selective destruction of malignant cells by the host's immune system. Molecular cancer vaccines rely on recently identified tumor antigens as immunogens. Tumor antigens can be applied in many forms, as genes in recombinant vectors, as proteins or peptides representing T cell epitopes. Analysis of various aspects indicates some advantage for peptide-based vaccines over the other modalities. Further refinements and extensively monitored clinical trials are necessary to advance molecular cancer vaccines from concepts into powerful therapy. C1 [Schweighoffer, Tamas] Boehringer Ingelheim Research and Development, Department Cell Biology, Dr. Boehringer-Gasse 5, A-1120 Vienna, Austria. RP Schweighoffer, T (reprint author), Boehringer Ingelheim Research and Development, Department Cell Biology, A-1120 Vienna, Austria. EM schweig@bender.co.at NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 164 EP 176 PG 13 ER PT J AU Lewin, JK AF Lewin, J Klaus TI Post-transplant Lymphoproliferative Disorders SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE post-transplant; lymphoproliferation ID post-transplant; lymphoproliferation AB Post-transplant lymphoproliferative disorder (PTLD) is one of the most dreaded complications of orthotopic transplantation. It consists of a heterogeneous group of lymphoproliferative disorders of varying clonal composition, occurring in immunosuppressed organ allograft recipients and is frequently due to EBV infection. It is most common in heart/lung transplants followed by heart, liver, and kidney and rarely in bone marrow transplants. Clinically, PTLD can present in a number of ways ranging from features resembling infectious mononucleosis, lymphoproliferative masses involving both nodal and extranodal locations, to a fulminant form characterized by a combination of peripheral lymphadenopathy, severe metabolic acidosis, organ failure or allograft dysfunction. Pathologically PTLD is characterized by a dense inflammatory infiltrate with a spectrum ranging from that found in infectious mononucleosis to a polymorphous B-cell hyperplasia to that of a monomorphous lymphoma. Analysis of EBV is especially useful for the diagnosis of early cases of PTLD. In addition, immunophenotyping to determine the lymphocyte type (B or T cell type) and monoclonality are most helpful in determining the prognosis. C1 [Lewin, J Klaus] University of Southern California, Keck Medical Center, Department of Pathology, 10833 Le Conte Ave, 90095-1732 Los Angeles, CA, USA. RP Lewin, JK (reprint author), University of Southern California, Keck Medical Center, Department of Pathology, 90095-1732 Los Angeles, USA. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 177 EP 182 PG 6 ER PT J AU Kopper, L Sebestyen, A Gallai, M Kovalszky, I AF Kopper, Laszlo Sebestyen, Anna Gallai, Monika Kovalszky, Ilona TI Syndecan-1 - A New Piece in B-cell Puzzle SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE syndecan-1; heparan sulfate; proteoglycan; hematopoietic cells; B-cells ID syndecan-1; heparan sulfate; proteoglycan; hematopoietic cells; B-cells AB Syndecans are transmembrane proteoglycans, with core proteins mainly decorated with heparan sulfate chains. Syndecan-1 is expressed in a tissue-, cell- and differentiation-specific manner. Its extracellular domain can bind via HS chains to matrix elements, to growth factors (especially ''heparin-binding'' proteins) and to certain biological agents. The ectodomain released by proteolysis can also be functionally active. The cytoplasmic domain can take part in signaling processes as well as in modifying cell shape. In hematopoietic cells syndecan-1 is expressed in normal pre-B-cells and plasma cells, as well as in plasmocytoid and lymphoplasmocytoid malignancies. According to our study syndecan-1 is expressed in B-CLL cells both in tissue environment and in circulation. C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Gallai, Monika] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM kopper@korb1.sote.hu NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 183 EP 191 PG 9 ER PT J AU Kovacs, GG Majtenyi, K Laszlo, L AF Kovacs, G Gabor Majtenyi, Katalin Laszlo, Lajos TI Prion Protein Immunohistochemistry in Creutzfeldt-Jakob Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Creutzfeldt-Jakob disease; prion protein; immunohistochemistry ID Creutzfeldt-Jakob disease; prion protein; immunohistochemistry AB Creutzfeldt-Jakob disease is a transmissible spongiform encephalopathy characterized clinically by dementia, myoclonus and, in some cases, periodic triphasic EEG-patterns. Neuropathologically the main features are spongiform change, astrocytosis, neuronal cell loss and, in a small percent of cases, amyloid plaques. Prion protein immunohistochemistry is used for definitive diagnosis of these diseases. In our study we present different immunostaining patterns in light microscopy using anti prion protein, and with immunogold labelling for ultrastructural localization of prion protein. Our results demonstrate the clinicopathological heterogeneity of Creutzfeldt-Jakob disease and reveal the role of the endosomal-lysosomal system in the pathogenesis. C1 [Kovacs, G Gabor] Semmelweis University of Medicine, Department of NeurologyBudapest, Hungary. [Majtenyi, Katalin] National Institute of Psychiatry and Neurology, Department of NeuropathologyBudapest, Hungary. [Laszlo, Lajos] Eotvos University, Department of General Zoology, H-1445 Budapest, Hungary. RP Laszlo, L (reprint author), Eotvos University, Department of General Zoology, H-1445 Budapest, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 193 EP 197 PG 5 ER PT J AU Shan, L Kakudo, K Nakamura, M Nakamura, Y Yokoi, T Ishimoto, J Kawahara, K Takami, H AF Shan, Liang Kakudo, Kennichi Nakamura, Misa Nakamura, Yasushi Yokoi, Toyoharu Ishimoto, Junya Kawahara, Katsuhiko Takami, Hiroshi TI Clonality of the Parathyroid Nodules with Uremic Parathyroid Hyperplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE parathyroid adenoma; uremic parathyroid hyperplasia; clonality; PGK gene ID parathyroid adenoma; uremic parathyroid hyperplasia; clonality; PGK gene AB Clonal assessment suggests that most parathyroid adenomas and a subset of uremic parathyroid hyperplasia are monoclonal. A weakness that remains in the prior clonal studies is assessing the clonal status of the tissue fragments containing multiple nodules rather than a single nodule emerging in the uremic parathyroid hyperplasia. We applied the X chromosome-linked phosphoglycerate kinase (PGK) gene inactivation assay method for clonality to study individual nodules. Materials were obtained from 31 cases with parathyroid adenoma and 16 with uremic parathyroid hyperplasia. 17 cases were heterozygous in the PGK-1 locus. We were able to assess the clonality of 10 parathyroid adenomas and 7 hyperplastic glands. Monoclonality was demonstrable in 9 of the 10 parathyroid adenomas and in 4 of the 7 hyperplastic glands. Further analysis of 11 individual nodules microdissected from 3 monoclonal and 1 polyclonal hyperplastic glands revealed that 6 nodules were monoclonal and 5 were polyclonal. Nodules arising in a hyperplastic gland could be of monoclonal or polyclonal origin. Polyclonal and monoclonal nodules coexisted within single glands. Our findings indicate a progression from generalized hyperplasia to a monoclonal tumor in uremic parathyroid hyperplasia. Comparing clonality with the parathyroid hormone (PTH) immunoreactivity and histological features, we found that monoclonal nodules showed a homogeneous immunoreactivity against PTH antibody, whereas most of the polyclonal nodules showed a heterogeneous staining. Classic morphological criteria alone was inadequate to distinguish a monoclonal from a polyclonal nodule. C1 [Shan, Liang] Wakayama Medical College, Second Department of Pathology, 27 Kyubancho, 640 Wakayama, Wakayama, Japan. [Kakudo, Kennichi] Wakayama Medical College, Second Department of Pathology, 27 Kyubancho, 640 Wakayama, Wakayama, Japan. [Nakamura, Misa] Wakayama Medical College, Second Department of Pathology, 27 Kyubancho, 640 Wakayama, Wakayama, Japan. [Nakamura, Yasushi] Wakayama Medical College, Second Department of Pathology, 27 Kyubancho, 640 Wakayama, Wakayama, Japan. [Yokoi, Toyoharu] Wakayama Medical College, Second Department of Pathology, 27 Kyubancho, 640 Wakayama, Wakayama, Japan. [Ishimoto, Junya] Ishimoto HospitalWakayama, Japan. [Kawahara, Katsuhiko] Kyoritsu HospitalNagoya, Japan. [Takami, Hiroshi] Teikyo University, Medical School, Department of SurgeryTeikyo, Japan. RP Shan, L (reprint author), Wakayama Medical College, Second Department of Pathology, 640 Wakayama, Japan. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 198 EP 203 PG 6 ER PT J AU Csuka, O Remenar, Koronczay, K Doleschall, Z Nemeth, Gy AF Csuka, Orsolya Remenar, Eva Koronczay, Krisztina Doleschall, Zoltan Nemeth, Gyorgy TI Predictive Value of p53, Bcl2 and Bax in the Radiotherapy of Head and Neck Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE radiosensitivity; head and neck cancer; p53; Bcl2; Bax ID radiosensitivity; head and neck cancer; p53; Bcl2; Bax AB Radiation is known to induce DNA damage resulting in the onset of apoptosis. The apoptosis is modulated by p53, Bcl2 and Bax proteins. High level of wild type p53 is required for radiation induced apoptosis. The p53 status, therefore, may be a crucial determinant of radiosensitivity of tumor cells. Overexpression of Bcl2, however, inhibits apoptosis via hetero- and homodimeric interaction. Bax might function as a cell death effector molecule that is neutralized by Bcl2. The aim of the present study is to investigate the correlation between p53, Bcl2, Bax and c-myc levels and the clinical response of head and neck cancer patients to radiation. The base line and 30 GY gamma radiation induced values of p53, Bcl2, Bax and c-myc were estimated by Western blot in 40 biopsies of head and neck cancers. We found that the radiosensitivity of head and neck cancer patients depends on the ratio of p53, Bcl2 and Bax protein levels. High Bcl2 levels resulted in radioresistance of cancer patients. Overexpression of Bax and c-myc may ensure the radiosensitivity of head and neck cancer patients. Our studies indicate that prediction of radiation sensitivity of tumors could be based on the simultaneous evaluation of p53, Bax and Bcl2 levels. C1 [Csuka, Orsolya] National Institute of Oncology, Rath gYorgy u. 7-9., H-1122 Budapest, Hungary. [Remenar, Eva] National Institute of Oncology, Rath gYorgy u. 7-9., H-1122 Budapest, Hungary. [Koronczay, Krisztina] National Institute of Oncology, Rath gYorgy u. 7-9., H-1122 Budapest, Hungary. [Doleschall, Zoltan] National Institute of Oncology, Rath gYorgy u. 7-9., H-1122 Budapest, Hungary. [Nemeth, Gyorgy] National Institute of Oncology, Rath gYorgy u. 7-9., H-1122 Budapest, Hungary. RP Csuka, O (reprint author), National Institute of Oncology, H-1122 Budapest, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 204 EP 210 PG 7 ER PT J AU Sergi, C Willig, F Thomsen, M Herwart, FO Krempien, B AF Sergi, Consolato Willig, Friedrich Thomsen, Marc Herwart, F Otto Krempien, Burkhard TI Bronchopneumonia Disguising Lung Metastases of a Painless Central Chondrosarcoma of Pubis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE lung metastasis; primary chondrosarcoma ID lung metastasis; primary chondrosarcoma AB Chondrosarcoma is a generally locally malignant chondroid-forming bone tumor with a low potential for distant metastases. A small and completely painless central chondrosarcoma of pubis metastasizing to the lungs in a 63-year-old woman with bronchopneumonia is reported. Here we emphasize the mimicry and low growth of the chondrosarcoma and the easiness with which the diagnosis in completely asymptomatic patients can be missed. Although painless chondrosarcoma metastasizing to lung is rather rare, this tumor should be always included in the differential diagnosis of malignancies in this age category. C1 [Sergi, Consolato] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220, D-69120 Heidelberg, Germany. [Willig, Friedrich] University of Heidelberg, Speyererhof LehrkrankenhausHeidelberg, Germany. [Thomsen, Marc] University of Heidelberg, Department of Orthopedic SurgeryHeidelberg, Germany. [Herwart, F Otto] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220, D-69120 Heidelberg, Germany. [Krempien, Burkhard] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220, D-69120 Heidelberg, Germany. RP Sergi, C (reprint author), University of Heidelberg, Department of Pathology, D-69120 Heidelberg, Germany. EM Consolato_Sergi@krzmail.krz.uni-heidelberg.de NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 211 EP 214 PG 4 ER PT J AU Lotz, G Koltai, P Schaff, Zs AF Lotz, Gabor Koltai, Pal Schaff, Zsuzsa TI Giant Cell Hepatitis in Adults SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE giant cell; hepatitis; adult ID giant cell; hepatitis; adult AB Giant cell hepatitis is a frequent reaction of the liver to different injuries in newborns and in childhood, but rare in adults. This form of hepatitis is often accompanied by cholestasis and shows fast progression to cirrhosis. In most cases autoimmune, metabolic, toxic or viral origin can be found, but sometimes the etiology remains hidden. This paper introduces two adult giant cell hepatitis cases. Hepatitis C virus infection was the possible origin in the first case and autoimmune disease in the other one. C1 [Lotz, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Koltai, Pal] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 215 EP 218 PG 4 ER PT J AU Toth, J Elek, G AF Toth, Jozsef Elek, Gabor TI Histiocytic and T-cell Rich B-Cell Lymphoma (TCRBCL) of the Stomach SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE B-cell lymphoma; gastric lymphoma; extranodal lymphoma; T-cell rich B-cell lymphoma; histiocyte-rich lymphoma; gastroscopy ID B-cell lymphoma; gastric lymphoma; extranodal lymphoma; T-cell rich B-cell lymphoma; histiocyte-rich lymphoma; gastroscopy AB Although stomach is a frequent site of extranodal lymphomas, histiocyte-rich TCRBCL has not yet been described there. Even histology of repeated gastrobiopsies of this uncommon, diffuse, large B-cell lymphoma may be inconclusive and partial gastrectomy cannot be avoided. It is only immunohistology (CD20, CD43, CD68) of the paraffin blocks from the resection specimen that can lead to the final diagnosis of intermediate grade malignant lymphoma. C1 [Toth, Jozsef] National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, Rath Gy. U. 5-7., H-1122 Budapest, Hungary. [Elek, Gabor] Central Railway Hospital and Polyclinic, Department of PathologyBudapest, Hungary. RP Toth, J (reprint author), National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, H-1122 Budapest, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 219 EP 223 PG 5 ER PT J AU Tarjan, V Ujhelyi, E Szabo, J Kellner, R Krall, G Gyuris, Mihaly, I Fust, G AF Tarjan, Veronika Ujhelyi, Eszter Szabo, Janos Kellner, Robert Krall, Geza Gyuris, Agnes Mihaly, Ilona Fust, George TI Three Cases of HIV-1 Seroreversion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE HIV; transient seropositivity ID HIV; transient seropositivity AB Three patients were enrolled, two as hemophiliacs, and one with acute EBV infection. Serial serum samples of each patient were tested with at least 3 different HIV antibody EIA tests, an immunofluorescent test and two western blots (WB). In the third case, PCR and reverse transcriptase enzyme activity measurement were also done. One of the regularly checked serum samples of hemophiliac patients was reactive with different HIV screening and confirmatory assays. Their next blood samples, two weeks and one month later, respectively, were negative with the same tests. In Case 3. two and a half years after the first examination, the EIA tests results changed to negative, but the WB was still indeterminate. In the case of the two hemophiliac patients, the patients may have been exposed to HIV containing blood products (before 1985), but were not infected. Regular treatment with factor VIII concentrate, in which HIV antigens may be present, can boost the immune response and results in transient seropositivity. In the case of the EBV infected patient, the transient HIV seropositivity may be the consequence of EBV induced proliferation of anti-HIV-antibody producing B cell clones. During our ten year HIV confirmatory practice we tested more than 40000 samples, from which transient seropositivity were observed only in the three cases summarized in this paper. C1 [Tarjan, Veronika] National Institute of Haematology, Blood Transfusion and Immunology, H-1518 Budapest, Hungary. [Ujhelyi, Eszter] National Institute of Haematology, Blood Transfusion and Immunology, H-1518 Budapest, Hungary. [Szabo, Janos] National Institute of Haematology, Blood Transfusion and Immunology, H-1518 Budapest, Hungary. [Kellner, Robert] Blood Transfusion CenterBaja, Hungary. [Krall, Geza] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Gyuris, Agnes] Orszagos Egeszsegbiztositasi PenztarBudapest, Hungary. [Mihaly, Ilona] St. Laszlo HospitalBudapest, Hungary. [Fust, George] Semmelweis University, 3rd Department of Internal MedicineBudapest, Hungary. RP Tarjan, V (reprint author), National Institute of Haematology, Blood Transfusion and Immunology, H-1518 Budapest, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 224 EP 228 PG 5 ER PT J AU Silletti, S Paku, S Raz, A AF Silletti, Steve Paku, Sandor Raz, Avraham TI Tumor Cell Motility and Metastasis. Autocrine Motility Factor as an Example of Ecto/Exoenzyme Cytokines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE migration; autocrine motility factor; neuroleukin; phosphohexose isomerase; metastasis ID migration; autocrine motility factor; neuroleukin; phosphohexose isomerase; metastasis AB Cellular locomotion plays a critical role in such normal processes as embryonic development, tissue segregation, as well as the infiltration of fibroblasts and vascular cells during wound repair and the inflammatory responses of the adult immune system. During tumor invasion and metastasis the processes of cell migration achieve dire significance. Disruption of normal homeostatic mechanisms to benefit the survival of the individual tumor cell is a common theme discovered during the characterization of factors once thought to be tumor-specific. One such molecule, tumor cell autocrine motility factor, was so described and has only recently been identified as a normal protein involved in intracellular glycolysis as well as implicated as an extracellular effector of normal cell functions including survival of certain populations of neurons. This molecule represents a member of the newly emerging family of intracellular enzymes whose disparate functions as extracellular mediators of cellular responses defines a new class of ecto/exoenzymes which play a role in normal cellular processes and are inappropriately utilized by tumor cells to elicit new survival strategies. C1 [Silletti, Steve] The Scripps Research Institute, Departments of Immunology and Vascular BiologyLa Jolla, USA. [Paku, Sandor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Raz, Avraham] Detroit Medical Center, Barbara Ann Karmanos Cancer Institute, 110 E. Warren Avenue, 48201 Detroit, Michigan, USA. RP Raz, A (reprint author), Detroit Medical Center, Barbara Ann Karmanos Cancer Institute, 48201 Detroit, USA. EM raza@kci.wayne.edu NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 1997 VL 3 IS 3 BP 230 EP 254 PG 25 ER PT J AU Degott, C AF Degott, Claude TI Drug-Induced Liver Injury Cholestatic Injury, Acute and Chronic SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE cholestasis; drug-induced ID cholestasis; drug-induced AB Cholestasis is the failure of bile to reach duodenum due to three different mechanisms: a. alteration of bile secretion by hepatocytes into the canaliculus with or without liver cell damage; b. obstruction of the intrahepatic bile ducts caused by diseases of ductules or small/medium bile ducts; c. obstruction of extrahepatic bile ducts. This short review focuses on drugs which may induce cholestasis by any of these mechanisms. C1 [Degott, Claude] Service D'Anatomie et de Cytologie Pathologique, 100 bd du Gal-Leclerc, 92118 Clichy, Cedex, France. RP Degott, C (reprint author), Service D'Anatomie et de Cytologie Pathologique, 92118 Clichy, France. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1997 VL 3 IS 4 BP 260 EP 263 PG 4 ER PT J AU Luo, J Sharma, N Seftor, AE De Larco, J Heidger, MP Hendrix, JM Lubaroff, MD AF Luo, Jun Sharma, Navesh Seftor, A Elisabeth De Larco, Joseph Heidger, M Paul Hendrix, JC Mary Lubaroff, M David TI Heterogeneous Expression of Invasive and Metastatic Properties in a Prostate Tumor Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prostate cancer; invasion; metastasis; E-cadherin; heterogeneity ID prostate cancer; invasion; metastasis; E-cadherin; heterogeneity AB Cellular heterogeneity of neoplasia is well demonstrated in the Dunning R-3327 rat prostate adenocarcinoma. In this study, we measured the differential expression of invasive and metastatic properties of this prostate model by cloning from a heterogeneous parental cell line. Four cell clones were derived and characterized by morphological studies, E-cadherin expression, and invasive and metastatic potential. Three of the clones (clones 5A, 5C, and 5D) demonstrated a fibroblastic morphology and were anchored to the substrate by loose microvillous processes. The fourth clone (clone 5B) grew in tight clusters and displayed many closely spaced microvilli, long overlapping cytoplasmic regions with well-defined junctional complexes. The parental line (R3327-5) demonstrated a combination of both these growth patterns. E-cadherin expression was absent in clones 5A, 5C, and 5D and very prominent in clone 5B, when compared to the parental line. The absence of E-cadherin expression correlated with increased invasiveness, as measured in an in vitro invasion assay. Subcutaneous injections of clones 5A, 5C, and 5D yielded lung metastases and no primary tumors at the site of inoculation while clone 5B was tumorigenic and produced fewer lung metastases in vivo. These clones, therefore, provide a potential for studying a variety of molecules involved in prostate cancer invasion and metastasis, especially for the direct testing of the significance of E-cadherin expresssion in prostate cancer progression. C1 [Luo, Jun] University of Iowa, Department of Anatomy, 1-101 Bowen Science Building, 51 Newton Road, 52242-1109 Iowa City, IA, USA. [Sharma, Navesh] University of Iowa, Department of Anatomy, 1-101 Bowen Science Building, 51 Newton Road, 52242-1109 Iowa City, IA, USA. [Seftor, A Elisabeth] University of Iowa, Department of Anatomy, 1-101 Bowen Science Building, 51 Newton Road, 52242-1109 Iowa City, IA, USA. [De Larco, Joseph] University of Iowa, Department of Anatomy, 1-101 Bowen Science Building, 51 Newton Road, 52242-1109 Iowa City, IA, USA. [Heidger, M Paul] University of Iowa, Department of Anatomy, 1-101 Bowen Science Building, 51 Newton Road, 52242-1109 Iowa City, IA, USA. [Hendrix, JC Mary] University of Iowa, Department of Anatomy, 1-101 Bowen Science Building, 51 Newton Road, 52242-1109 Iowa City, IA, USA. [Lubaroff, M David] The University of Iowa, Departments of Urology and Microbiology and the Veterans Affairs Medical CenterIowa City, USA. RP Hendrix, JM (reprint author), University of Iowa, Department of Anatomy, 52242-1109 Iowa City, USA. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1997 VL 3 IS 4 BP 264 EP 271 PG 8 ER PT J AU Kovacs, J Gomba, Sz Zilahy, M AF Kovacs, Judit Gomba, Szabolcs Zilahy, Monika TI Comparison of the morphology of renal cysts and cystic renal tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE renal cysts; renal tumors; tubular markers; cell proliferation; immunohistochemistry ID renal cysts; renal tumors; tubular markers; cell proliferation; immunohistochemistry AB Renal tumors appear uncommonly with cystic changes. They may develop due to necrosis though well-formed real cysts are also known. Such lesions may present problems in distinguishing them from benign renal cysts. Conditions leading to cyst formation are not known, however cell proliferation, altered extracellular matrix production and oncoprotein expression have been reported in cystic renal disorders. In the present study, we analysed the morphological features of 23 cystic renal tumors in comparison with 16 benign cysts using immunohistochemical and lectin binding methods. By our knowledge there has not been any piblication on such studies. The cystic renal tumors were represented predominantly in males and the size of the cysts was slightly larger than that of benign cysts. Tumorous cysts shared similar morphological appearance to solitary and multilocular cysts. They all showed strong epithelial membrane antigen reactivity on the luminal surface of the cells indicating distal tubular origin. Cell proliferation and p53 expression proved to be low excluding their role in the formation of the cysts. The amount of extracellular matrix and basement membrane was increased with an elevated type IV collagen and reduced fibronectin content. Polycystic kidney disease is different from tumorous cysts as cell proliferation, p53 oncoprotein expression and the composiition of extracellular matrix proved to be the opposite. As renal cell tumors arise from proximal tubules, neoplastic or metaplastic differentiation toward distal tubular direction seems to be the key even in cyst formation. Altered cell-matrix or cell-cell contact can modulate this transformation providing a basis for further results. C1 [Kovacs, Judit] University of Debrecen, Faculty of Medicine, Department of Pathology, H-4012 Debrecen, Hungary. [Gomba, Szabolcs] University of Debrecen, Faculty of Medicine, Department of Pathology, H-4012 Debrecen, Hungary. [Zilahy, Monika] University of Debrecen, Faculty of Medicine, Department of Pathology, H-4012 Debrecen, Hungary. RP Kovacs, J (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, H-4012 Debrecen, Hungary. EM kovacsj@lib.dote.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1997 VL 3 IS 4 BP 272 EP 277 PG 6 ER PT J AU Thomas, AP Oykutlu, D Pou, B Tyler, D Oberley, WL Robinson, AR Lenel, CJ AF Thomas, A Patricia Oykutlu, Dilek Pou, Bel Tyler, Denise Oberley, W Larry Robinson, A Robert Lenel, C Julia TI Immunohistochemical Characterization of Antioxidant Enzymes in Human Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; antioxidant enzymes; immunohistochemistry ID Breast cancer; antioxidant enzymes; immunohistochemistry AB Intrinsic antioxidant enzymes (AE) are essential for protection against potential cellular damage by free radicals (FRs), which affect a variety of biological processes. The levels or activities of AEs can be abnormal in human malignancies in general, and FR production is a possible mechanism of estrogen related carcinogenesis specifically. However, the role of AEs in breast cancer ramains unclear. Immunodetectable AEs were characterized in 95 node negative cancers using rabbit polyclonal antibodies. Results were correlated with established and experimental biomarkers of breast cancer. AEs were greater than benign differentiated epithelium in more than 40% and lower in 10-14% of tumors. Patterns of staining were enzyme and tumor pattern specific. Increased immunodetectable AE was associated with large, poorly differentiated tumors, and younger age. Catalase correlated with nuclear grade and disease related death (p< 0.05), and highlighted tumor microvasculature. Additional work in this area may further elucidate the role of AEs in breast cancer growth and progression. C1 [Thomas, A Patricia] University of Iowa, Department of Pathology, 5239 RCP;200 Hawkins Drive, 42242 Iowa City, Iowa, USA. [Oykutlu, Dilek] University of Iowa, Department of Pathology, 5239 RCP;200 Hawkins Drive, 42242 Iowa City, Iowa, USA. [Pou, Bel] University of Iowa, Department of Pathology, 5239 RCP;200 Hawkins Drive, 42242 Iowa City, Iowa, USA. [Tyler, Denise] University of Iowa, Department of Pathology, 5239 RCP;200 Hawkins Drive, 42242 Iowa City, Iowa, USA. [Oberley, W Larry] University of Iowa, Department of Pathology, 5239 RCP;200 Hawkins Drive, 42242 Iowa City, Iowa, USA. [Robinson, A Robert] University of Iowa, Department of Pathology, 5239 RCP;200 Hawkins Drive, 42242 Iowa City, Iowa, USA. [Lenel, C Julia] University of Iowa, Department of Pathology, 5239 RCP;200 Hawkins Drive, 42242 Iowa City, Iowa, USA. RP Thomas, AP (reprint author), University of Iowa, Department of Pathology, 42242 Iowa City, USA. EM patricia-thomas@uiowa.edu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1997 VL 3 IS 4 BP 278 EP 286 PG 9 ER PT J AU Cserni, G Viragh, Sz AF Cserni, Gabor Viragh, Szabolcs TI Immunohistochemical and Ultrastructural Analysis of a Mammary Cystic Hypersecretory Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE breast; cystic hypersecretory carcinoma; electronmicroscopy; immunohistochemistry ID breast; cystic hypersecretory carcinoma; electronmicroscopy; immunohistochemistry AB Cystic hypersecretory carcinoma (CHC) is a rare variant of intraductal carcinoma. A CHC in a 50-year-old woman was excised and processed for light and electron microscopy and immunohistochemistry. The tumor had a marked cystic appearance. The walls of the cysts consisted of epithelial and myoepithelial cells and a well-developed basement membrane. The epithelial cells contained well-developed rough-surfaced endoplasmatic reticulum and Golgi apparatus. Secretory granules were not detected, with the exception of a few mucus-producing cells. The secretion was predominantly homogenous, reminiscent of thyroid colloid, and demonstrated distinct PAS positivity. The cells displayed a strong labeling with epithelial membrane antigen (EMA) and EMA-positive structures were observed within the intraluminal secretion, too. Some of these were stained by alcian blue. In addition, the colloid-like material was admixed with mucus showing a filamentous internal structure and lipid droplets resulting in some heterogenity of the secretion. Intraductal micropapillary proliferation in some of the cysts and adjacent nondistended ducts was a further defining feature of the tumor. Steroid hormone receptor and Ki-67 proliferation marker immuno his Tochemistry showed scattered positivity among the tumor cells. These results are in agreement with previous observations and further clarify the nature of this low-grade in situ cancer. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., H-6000 Kecskemet, Hungary. [Viragh, Szabolcs] Haynal Imre University of Health Sciences, Institute of PathologyBudapest, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.c3.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1997 VL 3 IS 4 BP 287 EP 292 PG 6 ER PT J AU Ciardi, A Pecorella, I Trombetta, G Memeo, L De Quarto, A Di Tondo, U AF Ciardi, Antonio Pecorella, Irene Trombetta, Giorgio Memeo, Lorenzo De Quarto, Angeloluca Di Tondo, Ugo TI An Unusual Case of Neurofibroma of the Thyroid Capsule SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Visceral neurofibroma; peripheral nerve sheath tumors; thyroid capsule ID Visceral neurofibroma; peripheral nerve sheath tumors; thyroid capsule AB A solitary neurofibroma arising from the thyroid capsule in a 64 year-old woman is reported. The tumor was adherent to the lower margin of the thyroid gland and extended into the anterior mediastinum. The unusual nature of the lesion, both its relationship to the thyroid gland and to the occurrence of a neural tumor in the anterior mediastinum is discussed. C1 [Ciardi, Antonio] University of Rome, Campus Bio-Medico, Department of Pathology, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy. [Pecorella, Irene] University of Rome, Campus Bio-Medico, Department of Pathology, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy. [Trombetta, Giorgio] University of Rome, Campus Bio-Medico, Department of Pathology, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy. [Memeo, Lorenzo] University of Rome, Campus Bio-Medico, Department of Pathology, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy. [De Quarto, Angeloluca] University of Rome, Campus Bio-Medico, Department of Pathology, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy. [Di Tondo, Ugo] University of Rome, Campus Bio-Medico, Department of Pathology, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy. RP Ciardi, A (reprint author), University of Rome, Campus Bio-Medico, Department of Pathology, 00161 Rome, Italy. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1997 VL 3 IS 4 BP 293 EP 295 PG 3 ER PT J AU Prohaszka, Z Toth, DF Banhegyi, D Fust, G AF Prohaszka, Zoltan Toth, D Ferenc Banhegyi, Denes Fust, George TI Role of Complement and Antibodies in the Control and Facilitation of HIV Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE HIV; complement; gp41; gp120; mannan-binding lectin; enhancing antibodies; autoantibodies; heat-shock protein 60; C1q ID HIV; complement; gp41; gp120; mannan-binding lectin; enhancing antibodies; autoantibodies; heat-shock protein 60; C1q AB In humans the HIV infection results in a chronic disease with a permanent fight between factors controlling HIV and the escape of the virus. Fromthese control mechanisms the present review summarizes the role betwen complement and autoantibodies; the competition of complement and anti-HIV antibodies for binding sites, the role of mannan-binding lectin in the susceptibility to and in the survival after HIV infection, the contribution of complement-dependent enhancing type antibodies to the clinical progression of HIV disease as well as the changing pattern of some autoantibodies (mimicking MHC class II molecules, anti-heat shock protein 60 antibodies and anti-C1q antibodies) which were found to correlate to immunological and clinical parameters. C1 [Prohaszka, Zoltan] Semmelweis University, 3rd Department of Internal Medicine, Eotvos ut 12., H-1121 Budapest, Hungary. [Toth, D Ferenc] University of Debrecen, Faculty of Medicine, Department of Medical MicrobiologyDebrecen, Hungary. [Banhegyi, Denes] St. Istvan and St Laszlo Hospital, Immunology LaboratoryBudapest, Hungary. [Fust, George] Semmelweis University, 3rd Department of Internal Medicine, Eotvos ut 12., H-1121 Budapest, Hungary. RP Prohaszka, Z (reprint author), Semmelweis University, 3rd Department of Internal Medicine, H-1121 Budapest, Hungary. EM prohoz@bkt.sote.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1997 VL 3 IS 4 BP 296 EP 302 PG 7 ER PT J AU Gyuris, Segesdi, J Mezei, M Balog, K Jelenik, Zs Takacs, M Berencsi, Gy Foldes, I Majtenyi, K Kollar, K Minarovits, J AF Gyuris, Agnes Segesdi, Judit Mezei, Maria Balog, Katalin Jelenik, Zsuzsanna Takacs, Maria Berencsi, Gyorgy Foldes, Istvan Majtenyi, Katalin Kollar, Katalin Minarovits, Janos TI Virological, Neurological and Histological Investigations of a Child Born to a Mother with AIDS SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE HIV; mother to child transmission; central nervous system development; delayed myelinization ID HIV; mother to child transmission; central nervous system development; delayed myelinization AB HIV-1 was isolated from a child at 6 and 9 months of age, proving the vertical transmission of infection from the mother with AIDS. The p24 antigen test of the plasma at 9 months of age was positive as well. A positive PCR reaction was detected in J34 cells, infected with the supernatant of the peripheral blood lymphocytes of the child. According to phenotypic characterization, the virus proved to be a SI (syncytium inducing) isolate, growing in PBL, MT2, J34 and other T and monocytic cell lines. The isolate was AZT sensitive. Two methods were applied for genotypic characterization: 1. Heteroduplex mobility assay (HMA), 2. Sequence analysis of a part of the env gene. On the basis of both of these methods, this virus belongs to the B subtype of HIV-1, which is prevalent mainly in Europe and in the USA. The neurological status of the child was followed regularly. At autopsy the presence of p24 antigen was detected in glial cells of the frontal cortex, proving the presence of the virus in the brain. A retardation of the development of the central nervous system could be observed as well. C1 [Gyuris, Agnes] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. [Segesdi, Judit] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. [Mezei, Maria] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. [Balog, Katalin] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. [Jelenik, Zsuzsanna] St. Laszlo HospitalBudapest, Hungary. [Takacs, Maria] Bela Johan National Institute of Hygiene, Department of VirologyBudapest, Hungary. [Berencsi, Gyorgy] Bela Johan National Institute of Hygiene, Department of VirologyBudapest, Hungary. [Foldes, Istvan] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. [Majtenyi, Katalin] Orszagos Pszichiatriai es Neurologiai Intezet, Alkohologiai OsztalyBudapest, Hungary. [Kollar, Katalin] St. Panthaleon HospitalDunaujvaros, Hungary. [Minarovits, Janos] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. RP Gyuris, (reprint author), Bela Johan National Institute of Hygiene, Microbiological Research Group, H-1529 Budapest, Hungary. EM gyuris@microbi.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1997 VL 3 IS 4 BP 303 EP 308 PG 6 ER PT J AU Knox, JR Connors, AT AF Knox, J Richard Connors, A Tom TI Prodrugs in Cancer Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE antibody targeting; drug targeting; gene therapy; antitumor agents ID antibody targeting; drug targeting; gene therapy; antitumor agents AB At present, chemotherapy is not very effective against common solid cancers especially once they have metastasised. However, laboratory experiments and studies on dose intensification in humans have indicated that some anti-cancer agents might be curative but only if the dose given was very much higher than that presently obtainable clinically. Prodrugs, activated by enzymes expressed at raised level in tumors, can deliver at least 50-fold the normal dose and can cure animals with tumors normally resistant to chemotherapy. This approach has not yet proved to be practicable clinically because of the rarity of human tumors expressing a high level of an activating enzyme. However, new therapies have been proposed overcome this limitation of prodrug therapy. Enzymes that activate prodrugs can be directed to human tumor xenografts by conjugating them to tumor associated antibodies. After allowing for the conjugate to clear from the blood a prodrug is administered which is normally inert but which is activated by the enzyme delivered to the tumor. This procedure is referred to as ADEPT (antibody-directed enzyme prodrug therapy). Early clinical trials are promising and indicate that ADEPT may become an effective treatment for all solid cancers for which tumor associated or tumor specific antibodies are known. Tumors have also been targeted with the genes encoding for a prodrug activating enzymes. This approach has been called gene-directed enzyme prodrug therapy (GDEPT) or VDEPT (virus-directed enzyme prodrug therapy) and has shown good results in animal models. These new therapies may finally realise the potential of prodrugs in cancer chemotherapy. C1 [Knox, J Richard] Imperial College School of Medicine, Department of Medical Oncology, Charing Cross Campus, The Reynolds Building, St. Dunstan's Road, W68RP London, UK. [Connors, A Tom] The School of Pharmacy, Centre for Polymer TherapeuticsLondon, UK. RP Knox, JR (reprint author), Imperial College School of Medicine, Department of Medical Oncology, W68RP London, UK. EM r.knox@cxwms.ac.uk NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 1997 VL 3 IS 4 BP 309 EP 324 PG 16 ER PT J AU Klein, E AF Klein, Eva TI The Complexity of the Epstein-Barr Virus Infection in Humans SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE EBV; infections mononucleosis; immune response ID EBV; infections mononucleosis; immune response AB The Epstein-Barr virus (EBV) was isolated 40 years ago from cultures of Burkitt lymphoma cells (BL). The tumor was encountered in Africa and exhibited characteristical geographical, clinical and pathological features. Serological studies revealed that the virus is ubiquitous in humans. The primary infection is often accompanied by the syndrome of acute infectious mononucleosis (IM). It can induce malignant proliferation of B lymphocytes in conditions of immunodeficiency. EBV can immortalize B lymphocytes in culture. These cells carry the virus as episomes and express 9 virally encoded proteins. Their immunological recognition constitutes the surveillance which is responsible for the healthy virus carrier state. The main virus reservoir is represented by a low number of resting B lymphocyte which contain the viral genome but do not express its transformation proteins. The viral genom is detectable in all African BLs, in variable proportions of nasopharyngeal carcinoma, Hodgkin's disease, T cell lymphoma, lymphoepithelial like carcinoma, gastric carcinoma and leiomyosarcoma cases. The role of EBV in the genesis of these tumors is unknown. C1 [Klein, Eva] Karolinska Institutet, Microbiology and Tumor Biology Center (MTC), S-17177 Stockholm, Sweden. RP Klein, E (reprint author), Karolinska Institutet, Microbiology and Tumor Biology Center (MTC), S-17177 Stockholm, Sweden. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 3 EP 7 PG 5 ER PT J AU Moldvay, J Strausz, J Egervary, M Agocs, L Bocsi, J Schaff, Zs AF Moldvay, Judit Strausz, Janos Egervary, Marta Agocs, Laszlo Bocsi, Jozsef Schaff, Zsuzsa TI P53 Expression in Stage I Squamous Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p53; immunohistochemistry; lung cancer; flow cytometry ID p53; immunohistochemistry; lung cancer; flow cytometry AB P53 expression was studied using immunohistochemistry in patients (n=94) with pathologic stage I squamous cell lung cancer treated surgically between 1991-1992. The overall p53 positivity ratio was 48/94. 83 of the cases proved to be suitable for follow-up analysis carried out in November, 1995. 46/83 were p53 positive, and 25/46 patients were alive at the time of analysis. The patients who died (21/46) had a mean survival time of 17.5 months. In p53 negative cases (37/83), however, 29/37 patients were still alive at the time of follow-up, and 8/37 had died with a mean survival time of 23.1 months. A significant correlation could be found between p53 immunopositivity and reduced survival time (p=0.0125). Interestingly, out of 83 cases analyzed histologic evidence of tuberculous scar tissue was present in 9 tumors with a p53 positivity ratio of only 1/9. When flow cytometry was used to examine tumor samples from all subgroups mentioned above (n=32), no correlation was found between the p53 immunopositivity or the prognosis and the DNA content of tumor tissues. Our results suggest that in the early stage of squamous cell lung cancer the p53 positivity may be an indicator of a more aggressive tumor behavior and a shortened survival time. C1 [Moldvay, Judit] National Koranyi Institute of Pulmonology, Department of Bronchology, Piheno u. 1., H-1529 Budapest, Hungary. [Strausz, Janos] National Koranyi Institute of Pulmonology, Department of Bronchology, Piheno u. 1., H-1529 Budapest, Hungary. [Egervary, Marta] National Koranyi Institute of Pulmonology, Department of Bronchology, Piheno u. 1., H-1529 Budapest, Hungary. [Agocs, Laszlo] Semmelweis Egyetem-Orszagos Onkologiai Intezet, Mellkassebeszeti KlinikaBudapest, Hungary. [Bocsi, Jozsef] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Moldvay, J (reprint author), National Koranyi Institute of Pulmonology, Department of Bronchology, H-1529 Budapest, Hungary. EM moldvay@toledo.koranyi.hu NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 8 EP 13 PG 6 ER PT J AU Repassy, G Forster-Horvath, Cs Juhasz, A Adany, R Tamassy, A Timar, J AF Repassy, Gabor Forster-Horvath, Csaba Juhasz, Attila Adany, Roza Tamassy, Anna Timar, Jozsef TI Expression of Invasion Markers CD44v6/v3, NM23 and MMP2 in Laryngeal and Hypopharyngeal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE laryngeal carcinoma; CD44v3; nm23; MMP2; immunohistochemistry ID laryngeal carcinoma; CD44v3; nm23; MMP2; immunohistochemistry AB Twelve laryngeal squamous cell carcinoma cases (7 laryngeal and 5 hypopharyngeal cancer; 15 samples) were analysed by immunohistochemistry for the expression of invasion markers CD44v6/v3, NM23 and matrix metalloproteinase, MMP2. The laryngeal epithelium showed CD44v6+/v3+/NM23-/MMP2- phenotype. When tumors were grouped into TNM categories the phenotype of the T2 and T3 tumors was similar, characterised by decreased CD44v3+ and lack of MMP2 expressions. Meanwhile the NM23 expression was more frequent in T3 tumors. In T4 stage the frequency of NM23 and MMP2 positive cases increased (5/6 and 4/6, respectively) but there was no correlation with the appearence of lymph node metastasis. Comparison of the phenotype of laryngeal and hypopharyngeal tumors, irrespective of the TNM stages, revealed characteristic differences: T2 stage laryngeal tumors showed decreased CD44v3 and occasional NM23 and MMP2 positivity, while in T3 stage these tumors were characterised by increased frequency of NM23 positivity. The phenotype of the hypopharyngeal tumors was significantly different with a high frequency of MMP2 positive cases (5/6) and NM23+/low CD44v3+ phenotype. The sharp differences in the phenotypes of laryngeal and hypopharyngeal carcinomas were connected to the differences in their invasive capacity unlike to the size of the tumors, since the T4 stage hypopharyngeal tumors had a significantly smaller size than laryngeal ones, even at lower stages. C1 [Repassy, Gabor] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Forster-Horvath, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Juhasz, Attila] Medical University, Department of Oto-Rhino-LaryngologyDebrecen, Hungary. [Adany, Roza] Debrecen Medical University, Department of Hygieny and EpidemiologyDebrecen, Hungary. [Tamassy, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 14 EP 21 PG 8 ER PT J AU Kolesnikova, IA Kubasova, T Konoplyannikov, GA Koteles, JGy AF Kolesnikova, I Antonina Kubasova, Tamara Konoplyannikov, G Anatolij Koteles, J Gyorgy TI Cellular Alterations Upon IR-Laser (890 nm) Exposures, In Vivo SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE IR-laser; gamma irradiation; abscopal cellular effects; cell membranes; cell proliferation ID IR-laser; gamma irradiation; abscopal cellular effects; cell membranes; cell proliferation AB Exposure of cultured cells and small animals to ionizing radiation as well as irradiation of cultured cells with He-Ne laser can cause changes in the functional condition of plasma membranes. The ionizing radiation-induced cell membrane alterations have been determined after either partial or local exposures. The aim of the present study was to reveal whether the local laser treatments cause a general, distant, so called ''abscopal'' effect measured at cellular level, when the laser treatment is intended as a stimulatory procedure. The biological effect of infrared laser (mean power of 5 Watts, 150 Hz frequency, 890 nm wavelength) was demonstrated through 3H-concanavalin A binding by blood cells of daily irradiated (altogether 10 exposures) oncological and non-oncological patients as well as by changes in the proliferation of bone marrow cells of whole body gamma-irradiated (4 Gy) rats, partially laser-treated. The lectin binding of lymphocytes of oncological, as well as ischaemic heart disease patients was increased immediately after the first laser treatment. However, it was decreased after completion of the full course. In cases of inflammatory diseases the test parameters were either unchanged or decreased as compared to their self-control values. The platelets and erythrocytes did not react in any group. Gamma irradiation caused a deep inhibition of proliferation of rat bone marrow cells. The number of fibroblast colony-forming units (CFU-F) could be increased again if the animals were partially exposed to laser. Laser irradiation of one of the femurs led to some recovery of CFU-F values in the exposed as well as unexposed femur. Thus, local infrared laser treatment induces abscopal effects on the cell membrane and cell proliferation characteristics. C1 [Kolesnikova, I Antonina] Russian Academy of Science, Medical Radiological Research Center, 249020 Obninsk, Russian Federation. [Kubasova, Tamara] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Frederic Joliot-Curie Sugarbiologiai es Sugaregeszsegugyi Kutato Intezet, H-1775 Budapest, Hungary. [Konoplyannikov, G Anatolij] Russian Academy of Science, Medical Radiological Research Center, 249020 Obninsk, Russian Federation. [Koteles, J Gyorgy] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Frederic Joliot-Curie Sugarbiologiai es Sugaregeszsegugyi Kutato Intezet, H-1775 Budapest, Hungary. RP Kubasova, T (reprint author), Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Frederic Joliot-Curie Sugarbiologiai es Sugaregeszsegugyi Kutato Intezet, H-1775 Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 22 EP 26 PG 5 ER PT J AU Zalgeviciene, V Zukiene, J Grazeliene, G Sinkeviciute, G Didziapetriene, J AF Zalgeviciene, Violeta Zukiene, Janina Grazeliene, Grazina Sinkeviciute, Giedre Didziapetriene, Janina TI Embryotoxicity and Teratogenicity of Some Derivatives of Chloroethylaminophenylacetic Acid SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE embryotoxiciy; terotogenicity; antitumor agents ID embryotoxiciy; terotogenicity; antitumor agents AB Embryotoxic and teratogenic properties of Lophenal, Phenalon, Pharanox and Pharanoxi selenate were investigated experimentally. All examined antitumour agents showed embryotoxic effects. Lophenal, Phenalon and Pharanox had teratogenic effects. By modifying the structure of Pharanox with selenium a reduction in teratogenic effect was achieved. C1 [Zalgeviciene, Violeta] Faculty of Medicine of Vilnius University, Department of the Experimental Therapy of Lithuanian Oncology Center, Santariskiu 1, 2600 Vilnius, Lithuania. [Zukiene, Janina] Faculty of Medicine of Vilnius University, Department of the Experimental Therapy of Lithuanian Oncology Center, Santariskiu 1, 2600 Vilnius, Lithuania. [Grazeliene, Grazina] Faculty of Medicine of Vilnius University, Department of the Experimental Therapy of Lithuanian Oncology Center, Santariskiu 1, 2600 Vilnius, Lithuania. [Sinkeviciute, Giedre] Faculty of Medicine of Vilnius University, Department of the Experimental Therapy of Lithuanian Oncology Center, Santariskiu 1, 2600 Vilnius, Lithuania. [Didziapetriene, Janina] Faculty of Medicine of Vilnius University, Department of the Experimental Therapy of Lithuanian Oncology Center, Santariskiu 1, 2600 Vilnius, Lithuania. RP Didziapetriene, J (reprint author), Faculty of Medicine of Vilnius University, Department of the Experimental Therapy of Lithuanian Oncology Center, 2600 Vilnius, Lithuania. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 27 EP 29 PG 3 ER PT J AU Sava, G Gagliardi, R Cocchietto, M Clerici, K Capozzi, I Marrella, M Alessio, E Mestroni, G Milanino, R AF Sava, Gianni Gagliardi, Renato Cocchietto, Moreno Clerici, Katiuscia Capozzi, Ilaria Marrella, Mauro Alessio, Enzo Mestroni, Giovanni Milanino, Roberto TI Comparison of the Effects of the Antimetastatic Compound ImH[trans-RuCl4(DMSO)Im] (NAMI-A) on the Arthritic Rat and on MCa Mammary Carcinoma in Mice SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ruthenium; inflammation; tumor; metastasis; treatment ID ruthenium; inflammation; tumor; metastasis; treatment AB The effects of the new molecule ImH[trans-RuCl4(DMSO)Im] (NAMI-A), administered orally or intraperitoneally to adjuvant-arthritic rats or orally to mice bearing s.c. or i.m. implants of MCa mammary carcinoma, were studied. NAMI-A was not able to modify the progression of chronic inflammation in the complete Freund-adjuvant injected animals. Histology indicated a significant worsening of the inflammatory process, characterised by an increased infiltration of inflammatory cells, as well as by a remarkable deposition of connective tissue fibres around the blood vessels and alveolar walls. NAMI-A had no effect on primary i.m. implanted MCa mammary carcinoma growth and its lung metastasis formation, but significantly interfered with the cell cycle of primary tumor cells following bolus oral administration. On the contrary, NAMI-A caused a significant inhibition of lung metastasis accompanied by a dramatic deposition of connective tissue fibres around the primary tumor mass, when given as medicated food to mice implanted s.c. with MCa tumor. These data indicated that NAMI-A is well absorbed after oral administration although there is no connection between lung concentration and the antimetastatic activity. Conversely, the marked deposition of connective tissues in NAMI-A treated animals is in agreement with the reported effects of the compund on extracellular matrix and tumor blood vessels. C1 [Sava, Gianni] Callerio Foundation, Institutes of Biological Research, via A. Fleming, 34127 Trieste, Italy. [Gagliardi, Renato] Callerio Foundation, Institutes of Biological Research, via A. Fleming, 34127 Trieste, Italy. [Cocchietto, Moreno] Callerio Foundation, Institutes of Biological Research, via A. Fleming, 34127 Trieste, Italy. [Clerici, Katiuscia] Callerio Foundation, Institutes of Biological Research, via A. Fleming, 34127 Trieste, Italy. [Capozzi, Ilaria] Callerio Foundation, Institutes of Biological Research, via A. Fleming, 34127 Trieste, Italy. [Marrella, Mauro] University of Verona, Institute of PharmacologyVerona, Italy. [Alessio, Enzo] University of Trieste, Department of Chemical SciencesTrieste, Italy. [Mestroni, Giovanni] University of Trieste, Department of Chemical SciencesTrieste, Italy. [Milanino, Roberto] University of Verona, Institute of PharmacologyVerona, Italy. RP Sava, G (reprint author), Callerio Foundation, Institutes of Biological Research, 34127 Trieste, Italy. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 30 EP 36 PG 7 ER PT J AU Hermanns, B Handt, S Kindler, J Fuzesi, L AF Hermanns, Benita Handt, Stefan Kindler, Joachim Fuzesi, Laszlo TI Coronary Vasculopathy in Polycythemia Vera SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE polycythaemia vera; myocardial infarction; coronary vasculary disease; intima proliferation ID polycythaemia vera; myocardial infarction; coronary vasculary disease; intima proliferation AB Thrombosis is a common complication in polycythemia often causing death. In coronary artery occlusion, thrombosis due to hyperviscosity and thrombocytosis is mostly discussed as the origin of the infarction. We discuss the case of a 30-year-old male patient, with polycythemia, who died of myocardial infarction. On autopsy the vessels showed neither ateriosclerotic changes nor thrombotic occlusions. Instead, a marked intima proliferation was found leading to multiple occlusions whereas media and adventitia were unchanged. This pattern of a coronary vasculopathy has not been described before, and can be interpreted as an alternative mechanism for vascular occlusion in polycythemia. Similar histopathological changes have already been found in skin lesions in erythromelalgia, a common symptom in polycythemia. C1 [Hermanns, Benita] Medical Faculty of the Technical University of Aachen, Department of Pathology, Pauwelsstr. 30, D-52057 Aachen, Germany. [Handt, Stefan] Medical Faculty of the Technical University of Aachen, Department of Pathology, Pauwelsstr. 30, D-52057 Aachen, Germany. [Kindler, Joachim] Hospital Marienhohe, Department of Internal MedicineWurselen, Germany. [Fuzesi, Laszlo] Medical Faculty of the Technical University of Aachen, Department of Pathology, Pauwelsstr. 30, D-52057 Aachen, Germany. RP Fuzesi, L (reprint author), Medical Faculty of the Technical University of Aachen, Department of Pathology, D-52057 Aachen, Germany. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 37 EP 39 PG 3 ER PT J AU Blaker, H Dyckhoff, G Weidauer, H Herwart, FO AF Blaker, Hendrik Dyckhoff, Gerhard Weidauer, Hagen Herwart, F Otto TI Carcinoid Tumor of the Middle Ear in a 28-Year-Old Patient SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE carcinoid; tumor; pathology; diagnosis; middle-ear ID carcinoid; tumor; pathology; diagnosis; middle-ear AB Carcinoid tumor of the middle ear is an extremely rare condition. The origin of the tumor cells is still speculative and the closeness of relationship to adenomas of the middle ear has been a matter of discussion since the first description of this tumor entity in 1980. In this study we report a case of a 28-year-old male patient with a carcinoid tumor of the middle ear. We present the results of histomorphological, immunohistochemical and electron microscopic examinations and compare our findings to those of previously published cases. C1 [Blaker, Hendrik] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220, 69120 Heidelberg, Germany. [Dyckhoff, Gerhard] University of Heidelberg, Department of OtorhinolaryngologyHeidelberg, Germany. [Weidauer, Hagen] University of Heidelberg, Department of OtorhinolaryngologyHeidelberg, Germany. [Herwart, F Otto] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220, 69120 Heidelberg, Germany. RP Blaker, H (reprint author), University of Heidelberg, Department of Pathology, 69120 Heidelberg, Germany. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 40 EP 43 PG 4 ER PT J AU Ala Andrade, L Torresan, ZR Sales, FJ Vicentini, R De Souza, AG AF Ala Andrade, Liliana Torresan, Z Renato Sales, FS Jose Vicentini, Regina De Souza, A Gustavo TI Intravenous Leiomyomatosis of the Uterus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE uterine tumor; intravenous leiomyomatosis ID uterine tumor; intravenous leiomyomatosis AB Three cases of intravenous leiomyomatosis (IVL) of the uterus, a rare benign smooth-muscle tumor, are described. A preoperative diagnosis of IVL was not made in any of the patients, all of which presented with a pelvic mass with the presumptive diagnosis of leiomyoma. Surgical exploration confirmed the presence of uterine mass and two of the three cases showed extra-uterine extension into the ovarian or uterine veins. Histological examination demonstrated a fascicular pattern of bland spindle-shaped smooth-muscle cells, which extended to veins inside the myometrium or to extrauterine veins. This was confirmed by immunohistochemical stain for desmin and factor VIII. Despite their histological benignity, these lesions have a tendency to metastasize and are closely related to the conditions called ''benign metastasizing leiomyoma'' and ''intracaval mass and cardiac extension''. The primary treatment of IVL is hysterectomy and excision of any extrauterine tumor, when technically feasible. Anti-estrogenic therapy has been suggested as potentially useful in controlling of unresectable tumor. According to the literature, the follow-up must be long and periodic postoperative ultrasonic or magnetic nuclear resonance imaging studies may be useful in detecting growth of residual intravascular tumor. C1 [Ala Andrade, Liliana] UNICAMP - University of Campinas, School of Medicine, Department of Pathology, 13083-970 Campinas, SP, Brazil. [Torresan, Z Renato] Faculdade de Ciencias Medicas, Universidade Estadual de Campinas, UNICAMP, Department of GynecologyCampinas, Brazil. [Sales, FS Jose] Faculdade de Ciencias Medicas, Universidade Estadual de Campinas, UNICAMP, Department of GynecologyCampinas, Brazil. [Vicentini, Regina] Faculdade de Ciencias Medicas, Universidade Estadual de Campinas, UNICAMP, Department of GynecologyCampinas, Brazil. [De Souza, A Gustavo] Faculdade de Ciencias Medicas, Universidade Estadual de Campinas, UNICAMP, Department of GynecologyCampinas, Brazil. RP Ala Andrade, L (reprint author), UNICAMP - University of Campinas, School of Medicine, Department of Pathology, 13083-970 Campinas, Brazil. EM anatpat@turing.unicamp.br NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 44 EP 47 PG 4 ER PT J AU Toth, Z Sipos, J AF Toth, Zoltan Sipos, Jozsef TI Biclonal primary plasma cell leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE primary plasma cell leukemia; biclonality; immunohistology; image cytometry ID primary plasma cell leukemia; biclonality; immunohistology; image cytometry AB Authors present a multiparameter pathological study in a case of rapid biclonal primary plasma cell leukemia. The immunohistochemical data revealed aberrant phenotypes (monocyte, epithelial and T-cell) probably in connection with microenvironmental influences. Biclonality can be attributed to class switching during malignant transformation. Static image cytometry showed aneuploidy. The blasts of this process are active immunoregulatory cells. C1 [Toth, Zoltan] County Hospital of Zala, Department of Pathology, Zrinyi ut 1., H-8901 Zalaegerszeg, Hungary. [Sipos, Jozsef] County Hospital of Zala, Department of Pathology, Zrinyi ut 1., H-8901 Zalaegerszeg, Hungary. RP Toth, Z (reprint author), County Hospital of Zala, Department of Pathology, H-8901 Zalaegerszeg, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 48 EP 51 PG 4 ER PT J AU Segesdi, J Gyuris, Banhegyi, D Vodros, D Bakos, Minarovits, J AF Segesdi, Judit Gyuris, Agnes Banhegyi, Denes Vodros, Dalma Bakos, Agnes Minarovits, Janos TI Plasma HIV-1 Load and Disease Progression in HIV-Infected Patients in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE plasma HIV-1 RNA copy number; NASBA; AIDS; AIDS Related Complex ID plasma HIV-1 RNA copy number; NASBA; AIDS; AIDS Related Complex AB Nucleic Acid Sequence Based Amplification (NASBA) is a suitable method for the quantification of HIV-1 RNA in plasma and serum samples. Since determination of the viral load appears to be a valuable marker for the prediction of disease progression and for monitoring the efficiency of antiretroviral therapy, the National AIDS Committee initiated the introduction of NASBA in Hungary at the end of 1996. We obtained plasma samples from patients with ARC and AIDS of the Szt. Laszlo Hospital, Budapest. We found an increased viral burden in untreated AIDS (CDC group C) patients compared to untreated ARC (CDC group B) patients. In plasma samples of clinically stable ARC and AIDS patients treated with antiretroviral drugs we detected relatively low HIV-1 RNA copy levels while similarly treated ARC and AIDS patients with progressive disease had high HIV-1 RNA copy numbers. The CD4+ T-cell count was lower in AIDS patients compared to ARC patients, as expected. In general, there was an inverse correlation (r = -0.487, P < 0.0001) between CD4+ T-cell counts and HIV-1 RNA levels. We concluded that measurement of HIV-1 RNA plasma level has an important role in assessing prognosis and effects of antiretroviral therapy in HIV-infected patients. C1 [Segesdi, Judit] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. [Gyuris, Agnes] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. [Banhegyi, Denes] St. Istvan and St Laszlo Hospital, Immunology LaboratoryBudapest, Hungary. [Vodros, Dalma] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. [Bakos, Agnes] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. [Minarovits, Janos] Bela Johan National Institute of Hygiene, Microbiological Research Group, Piheno ut 1., H-1529 Budapest, Hungary. RP Segesdi, J (reprint author), Bela Johan National Institute of Hygiene, Microbiological Research Group, H-1529 Budapest, Hungary. EM sgesdi@microbi.hu NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 52 EP 55 PG 4 ER PT J AU Marodi, LCs Csiszar, A Sierra-Vazquez, B Di Luca, D Barabas, Nagy, K Ongradi, J AF Marodi, Laszlo Csaba Csiszar, Anna Sierra-Vazquez, Beatriz Di Luca, Dario Barabas, Eva Nagy, Karoly Ongradi, Joseph TI Studies on the Antibodies to Human Herpesvirus Type 6 Among Hungarian Patients with Asymptomatic HIV Infection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE HHV-6; seroepidemiology; IgM; IgG avidity; asymptomatic HIV-1 infection ID HHV-6; seroepidemiology; IgM; IgG avidity; asymptomatic HIV-1 infection AB The occurrence and the possible role in promoting HIV infection by human herpesvirus type 6 (HHV-6) have not yet been revealed in Hungary. In different groups of patients, serum titre of IgM and IgG antibodies, as well as avidity of IgG were quantitated by indirect immunofluorescence and an enzyme-linked immunosorbent assay, using isolate U1102 of HHV-6 variant A as antigen. In 60% of HIV-seronegative adult controls, high avidity IgG antibodies were found in low titre suggesting childhood infection. In HIV-seronegative persons with high risk behaviour for HIV-infection, both IgM and low avidity IgG were frequently found in higher titre, representing either primary or frequent reinfections, or reactivation of latent HHV-6. In asymptomatic HIV-seropositive patients, high titre of high avidity IgG antibodies was predominant, proving virus infection in the near past. These results indicate the contribution of HHV-6 to immunosuppression prior to AIDS, predisposing the organism to HIV infection. C1 [Marodi, Laszlo Csaba] Semmelweis University, Department of Medical MicrobiologyBudapest, Hungary. [Csiszar, Anna] Semmelweis University, Department of Medical MicrobiologyBudapest, Hungary. [Sierra-Vazquez, Beatriz] Semmelweis University, Department of Medical MicrobiologyBudapest, Hungary. [Di Luca, Dario] University of Ferrara, Institute of MicrobiologyFerrara, Italy. [Barabas, Eva] National Institute for Dermato-Venereology, Maria u. 41., H-1085 Budapest, Hungary. [Nagy, Karoly] National Institute for Dermato-Venereology, Maria u. 41., H-1085 Budapest, Hungary. [Ongradi, Joseph] Semmelweis University, Department of Medical MicrobiologyBudapest, Hungary. RP Ongradi, J (reprint author), Semmelweis University, Department of Medical Microbiology, Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 56 EP 61 PG 6 ER PT J AU Paku, S AF Paku, Sandor TI Current Concepts of Tumor-Induced Angiogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE tumor; metastasis; angiogenesis; morphology ID tumor; metastasis; angiogenesis; morphology AB Tumor induced angiogenesis is responsible for the nutrition of the growing tumor and can also increase the probability of hematogenous tumor dissemination. Data obtained from morphological analysis of tumor angiogenesis can contribute to the development of new anti-angiogenic therapies. Based on in vitro and in vivo observations several models of angiogenesis were introduced, explaining the mechanism of lumen formation and the timing of basement membrane depositon. (1) Lumen is formed either by cell body curving or by fusion of intracellular vacuoles of nonpolarized endothelial cells. New basement membrane is deposited after lumen formation. (2) Slit-like lumen is immediately formed by migrating polarized endothelial cells. Basement membrane is continuously deposited during endothelial cell migration, only cellular processes of the endothelial cell migrating on the tip of the growing capillary are free of deposited basement membrane material. (3) Development of transluminal bridges in larger vessels - a process called intussusceptive growth - leads to the division of the vessels. These models, however, describe angiogenesis in tissues rich in connective tissue. Different processes of angiogenesis take place in organs - such as liver, lungs, adrenals, which are the most frequent sites of metastasis - having high vessel density without sufficient space for capillary sprouting. In the case of liver metastases of Lewis lung carcinoma the proliferation of endothelial cells was elicited only by direct contact between tumor and endothelial cells, leading to the development of large convoluted vessels inside the metastases. These vessels were continuous with the sinusoidal system, suggesting that these metastases have dual blood supply. This observation, among others, is in contrast to the generally accepted view that liver tumors have arterial blood supply. The increasing number of data demonstrating the dual or venous blood supply of liver metastases should be taken into consideration in the therapy of liver metastasis. C1 [Paku, Sandor] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, Ulloi ut 26., H-1085 Budapest, Hungary. RP Paku, S (reprint author), Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, H-1085 Budapest, Hungary. EM paku@korb1.sote.hu NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1998 VL 4 IS 1 BP 62 EP 75 PG 14 ER PT J AU Ross, JP Rao, Sh Cunningham, D AF Ross, J Paul Rao, Sheela Cunningham, David TI Chemotherapy of Oesophago-Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE oesophagus; gastric; chemotherapy; chemoradiation ID oesophagus; gastric; chemotherapy; chemoradiation AB Oesophageal and gastric cancers are common tumors that represent a number of challenges for oncologists, gastroenterologists and surgeons. The prognosis remains poor with the majority of patients presenting with advanced disease. Combined chemotherapy and radiotherapy has demonstrated a survival benefit in patients with loco-regional oesophageal cancer compared to radiotherapy alone. In an interim analysis we have observed a 62% response rate using a chemoradiation regimen based on protracted venous infusion of 5-fluorouracil and cisplatin combined with radiotherapy in patients with inoperable oesophageal cancer. Improved outcomes with loco-regional disease has rekindled interest in preoperative therapy. In a trial comparing preoperative chemoradiation to surgery alone in patients with operable oesophageal adenocarcinoma, survival was improved with multimodality treatment. In addition, a study including both adeno- and squamous carcinomas demonstrated a trend towards improved survival. A complete pathological response to chemoradiation was associated with significantly improved survival. Gastric cancer is one of the most chemosensitive solid tumors of the gastrointestinal tract with the majority of patients being suitable for palliative chemotherapy. The ECF (epirubicin, cisplatin, protracted venous infusion 5-fluorouracil) regimen was developed in the Gastrointestinal unit of the Royal Marsden Hospital and first reported in 1991. In a prospective randomised trial including 274 patients ECF has been compared with the standard combination of 5-fluorouracil, adriamycin and methotrexate (FAMTX) in patients with previously untreated gastric cancer. Overall response rate, failure-free and overall survival were significantly improved with ECE, ECF also demonstrated improved quality of life and cost effectiveness when compared to the FAMTX regimen. ECF should now be regarded as the standard treatment for advanced oesophago-gastric cancer against which new therapies should be compared. In addition the Medical Research Council are conducting a trial randomising patients between surgery alone and perioperative chemotherapy using the ECF regimen in operable gastric cancer. C1 [Ross, J Paul] The Royal Marsden Hospital, Department of Medicine and the Gastrointestinal Unit, Downs Road, SM2 5PT Sutton, Surrey, UK. [Rao, Sheela] The Royal Marsden Hospital, Department of Medicine and the Gastrointestinal Unit, Downs Road, SM2 5PT Sutton, Surrey, UK. [Cunningham, David] The Royal Marsden Hospital, Department of Medicine and the Gastrointestinal Unit, Downs Road, SM2 5PT Sutton, Surrey, UK. RP Cunningham, D (reprint author), The Royal Marsden Hospital, Department of Medicine and the Gastrointestinal Unit, SM2 5PT Sutton, UK. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 87 EP 95 PG 9 ER PT J AU Petty, DR Evans, TA Duncan, DI Kurbacher, MCh Cree, AI AF Petty, D Russel Evans, T Alan Duncan, D Iain Kurbacher, M Christian Cree, A Ian TI Drug Resistance in Ovarian Cancer - the Role of p53 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ovary; adenocarcinoma; p53; immunostaining; bcl-2; chemotherapy ID ovary; adenocarcinoma; p53; immunostaining; bcl-2; chemotherapy AB The aims were to determine the importance of p53 and bcl-2 expression on the response to chemotherapy with alkylating agents in patients with ovarian cancer. We have followed the response to chemotherapy in a series of 59 patients with ovarian adenocarcinoma designated as p53 and bcl-2 positive or negative by immunocytochemistry. Of these cases, 50 received either cisplatin + treosulfan or treosulfan alone. Immunocytochemistry for p53 was positive in 28/59 tumors. Patients were grouped according to their response to chemotherapy (stable or progressive disease) assessed at 6, 12, and 18 months. There was increasing divergence of p53+ and p53- tumors over time. Of those which were p53+, 25% showed progression at 6 months, 80% at 12 months and 89% progression at 18 months. In contrast, 23%, 50%, and 67% of p53- tumors showed progression at 6 , 12 and 18 months respectively. For bcl-2, in 23/55 positive tumors there was progression in 35%, 78% and 94% compared with 25%, 57% and 59% in bcl-2 negative tumors at 6, 12 and 18 months respectively. Those tumors which were bcl-2 and p53 negative were most likely to progress, while those which were bcl-2 and p53 positive had the best prognosis. These differences did not translate into increased overall survival with minimum follow-up of 12 months. This data lends support to our suggestion that despite initially increased susceptibility to alkylating agents, enhanced genomic instability due to p53 inactivation may render tumors more likely to develop resistance to chemotherapy over time. This effect may be altered by bcl-2 function, lack of which will lead to a good response to chemotherapy as the tumor's ability to undergo apoptosis will not be compromised. C1 [Petty, D Russel] Ninewells Hospital and Medical School, Department of Obstetrics and GynaecologyDundee, Scotland, UK. [Evans, T Alan] Ninewells Hospital and Medical School, Department of PathologyDundee, Scotland, UK. [Duncan, D Iain] Ninewells Hospital and Medical School, Department of Obstetrics and GynaecologyDundee, Scotland, UK. [Kurbacher, M Christian] University of Cologne Medical Center, Labor fur Chemosensitivitatestungen Universitats & FrauenklinikCologne, Germany. [Cree, A Ian] University College London, Institute of Ophthalmology, Department of Pathology, Bath Street, EC1V9EL London, UK. RP Cree, AI (reprint author), University College London, Institute of Ophthalmology, Department of Pathology, EC1V9EL London, UK. EM i.cree@ucl.ac.uk NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 97 EP 102 PG 6 ER PT J AU Athanassiadou, P Sakellariou, V Petrakakou, E Athanassiades, P Zerva, Ch Liossi, A Michalas, S AF Athanassiadou, Pauline Sakellariou, Vasiliki Petrakakou, Efthalia Athanassiades, Peter Zerva, Cherry Liossi, Anna Michalas, Stylianos TI Cathepsin-D Immunoreactivity in Ovarian Cancer: Correlation with Prognostic Factors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ovarian carcinoma; cathepsin D; immunocytochemistry; prognostic factors ID ovarian carcinoma; cathepsin D; immunocytochemistry; prognostic factors AB In view of the somewhat inconclusive nature of the reports of the role of Cathepsin D (Cath D) in ovarian carcinoma and its relationship with various other parameters of malignancy the present study was performed to aid in the further clarification of this role. One hundred freshly resected primary ovarian carcinomas of various histological types were studied for ER, PR and Cath D status and the results examined with respect to menopausal status, histology, size and lymph node invasion. In our series Cath D positivity was more frequent in serous than in other types of ovarian cancer but this Cath D positivity was not related to the frequency of lymph node invasion regardless of the size of the tumor. Nor was any association observed between Cath D positivity and ER or PR status of the tumors or the menopausal state of the patients. The reported prognostic value of Cath D, ER and PR is discussed as well as the distinction between tumor invasion by lymphatic channels and direct interstitial infiltration. It was concluded that Cath D may not play a role in the former mode but, as might be expected from its proteolytic properties, in local spread by means of tissue destruction. C1 [Athanassiadou, Pauline] Alexandra Hospital Medical School, Athens University, Department of Cytology, 75 Micras Asias Str, GR-11527 Athens, Greece. [Sakellariou, Vasiliki] Alexandra Hospital Medical School, Athens University, 1st Obstetrics ClinicAthens, Greece. [Petrakakou, Efthalia] Alexandra Hospital Medical School, Athens University, Department of Cytology, 75 Micras Asias Str, GR-11527 Athens, Greece. [Athanassiades, Peter] University of Athens, Alexandra Hospital, Clinical TherapeuticsAthens, Greece. [Zerva, Cherry] University of Athens, Alexandra Hospital, Clinical TherapeuticsAthens, Greece. [Liossi, Anna] Alexandra Hospital Medical School, Athens University, Department of Cytology, 75 Micras Asias Str, GR-11527 Athens, Greece. [Michalas, Stylianos] Alexandra Hospital Medical School, Athens University, 1st Obstetrics ClinicAthens, Greece. RP Athanassiadou, P (reprint author), Alexandra Hospital Medical School, Athens University, Department of Cytology, GR-11527 Athens, Greece. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 103 EP 107 PG 5 ER PT J AU Ladanyi, A Nagy, OJ Jeney, A Timar, J AF Ladanyi, Andrea Nagy, O Julianna Jeney, Andras Timar, Jozsef TI Cytokine Sensitivity of Metastatic Human Melanoma Cell Lines - Simultaneous Inhibition of Proliferation and Enhancement of Gelatinase Activity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE melanoma; cytokines; proliferation; metalloproteinases ID melanoma; cytokines; proliferation; metalloproteinases AB The effect of a panel of cytokines on the proliferation and type IV collagenase production was studied in four melanoma cell lines of different origin, tumorigenicity and metastatic capacity. TGF-?, TNF-? and to a lesser extent, IL-1? exhibited antiproliferative effect on the cell lines, with some lines showing varying degree of resistance. The sensitivity did not correlate directly with the origin or the biological behavior of the tumor lines, suggesting that cytokine resistance of advanced stage melanoma cells may be relative. IL-2, IL-10 and IL-12 displayed little or no effect on proliferation. The effect of cytokines on metalloproteinase production showed a cell line dependent pattern. Interestingly, those cytokines that exhibited the most pronounced antiproliferative activity, also proved most effective in stimulating collagenase secretion, often simultaneously, in the same line. The results indicate that pleiotropic cytokines can have positive and negative effects simultaneously on various steps of tumor progression. C1 [Ladanyi, Andrea] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, Ulloi ut 26., H-1085 Budapest, Hungary. [Nagy, O Julianna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Jeney, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Timar, Jozsef] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Ladanyi, A (reprint author), Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, H-1085 Budapest, Hungary. EM ladanyi@korb1.sote.hu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 108 EP 114 PG 7 ER PT J AU Grover-McKay, M Walsh, AS Seftor, AE Thomas, AP Hendrix, JM AF Grover-McKay, Maleah Walsh, A Susan Seftor, A Elisabeth Thomas, A Patricia Hendrix, JC Mary TI Role for Glucose Transporter 1 Protein in Human Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; invasion; GLUT expression ID breast cancer; invasion; GLUT expression AB Glycolysis is increased in cancer cells compared with normal cells. It has been shown that glucose enters cells via a family of five functional glucose transporters (GLUT). However, GLUT expression appears to be altered in human breast cancer, which may serve as a selective advantage and facilitate the metastatic potential of these cells. The relationship of GLUT isoform expression and breast cancer cell invasiveness has not been adequately addressed. Thus, the purpose of this study was to investigate whether an association exists between GLUT expression and human breast cancer cell invasiveness. Invasiveness of the human breast cancer lines MCF-7, MDA-MB-435 and MDA-MB-231 was measured using an in vitro assay and compared with cellular GLUT isoform expression, assessed by Western blot analysis and verified by immunohistochemistry in a poorly differentiated human ductal breast cancer. Cell surface GLUT-1 expression was associated with the invasive ability of MCF-7 (2.0 + 0.02%), MDA-MB-435 (6.4 + 0.4%), and MDA-MB-231 (19.3 + 2.0%). However, GLUT-2 and GLUT-5 were inversely associated with invasiveness; GLUT-3 expression was variable; and GLUT-4 was undetected. In a poorly differentiated human ductal breast cancer, in situ GLUT-1 staining was intense. GLUT-1 expression was associated with the in vitro invasive ability of human breast cancer cells which was validated in situ. If this relationship is found to exist in a larger number of human breast cancer tissues, it may be possible to develop diagnostic and therapeutic strategies based on targeted GLUT isoform expression. C1 [Grover-McKay, Maleah] University of Iowa College of Medicine and University of Iowa Cancer Center, Departments of Radiology, Internal Medicine, IA52242 Iowa City, Iowa, USA. [Walsh, A Susan] University of Iowa College of Medicine and University of Iowa Cancer Center, Departments of Radiology, Internal Medicine, IA52242 Iowa City, Iowa, USA. [Seftor, A Elisabeth] University of Iowa, Department of AnatomyIowa City, USA. [Thomas, A Patricia] University of Kansas Medical School, Department of PathologyKansas City, USA. [Hendrix, JC Mary] University of Iowa, Department of AnatomyIowa City, USA. RP Grover-McKay, M (reprint author), University of Iowa College of Medicine and University of Iowa Cancer Center, Departments of Radiology, Internal Medicine, IA52242 Iowa City, USA. EM maleah-grover@uiowa.edu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 115 EP 120 PG 6 ER PT J AU Bencsath, M Paloczi, K Szalai, Cs Szenthe, A Szeberenyi, J Falus, A AF Bencsath, Marta Paloczi, Katalin Szalai, Csaba Szenthe, Andrea Szeberenyi, Julia Falus, Andras TI Histidine Decarboxylase in Peripheral Lymphocytes of Healthy Individuals and Chronic Lymphoid Leukemia Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE histamine; lymphocyte; cell proliferation; leukemia ID histamine; lymphocyte; cell proliferation; leukemia AB Histidine decarboxylase (HDC), the only enzyme capable of synthetizing histamine, has been found in many proliferating cells and tissues suggesting a role of histamine in cellular proliferation. In this study expression of HDC and the significance of histamine in the proliferation of peripheral lymphocytes of five healthy persons and six patients with chronic lymphoid leukemia (CLL) was examined. Expression of HDC mRNA and the protein was proved by reverse transcriptase polymerase chain reaction and by immunoblot, respectively. The role of histamine was studied in proliferation assays by the action of irreversible HDC inhibitor (alpha-fluoromethylhistidine--aFMH) and also by competing for the intracellular binding sites of histamine using N,N-diethyl-2,4-phenylmethyl-phenoxy-ethanamine-HCl (DPPE). Inhibiting the HDC enzyme activity by aFMH and blocking the intracellular action of histamine by DPPE, a significant decrease in cell proliferation was observed in mitogen stimulated lymphocytes of healthy donors. In CLL patients the proliferation of leukemic lymphocytes was inhibited significantly by DPPE. aFMH inhibited only the de novo histamine formation but did not the proliferation. The observations suggest that HDC has functional relevance in lymphocytes, since mitogen induced lymphocyte proliferation of healthy donors is mainly enhanced by de novo synthesis and subsequent action of intracellular histamine. Alternatively, in constitutively proliferating chronic lymphoid leukemia cells we suggest that the preformed pool but not the de novo synthesized intracellular histamine interferes with cellular proliferation. C1 [Bencsath, Marta] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4., H-1089 Budapest, Hungary. [Paloczi, Katalin] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Szalai, Csaba] Heim Pal Children's HospitalBudapest, Hungary. [Szenthe, Andrea] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4., H-1089 Budapest, Hungary. [Szeberenyi, Julia] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4., H-1089 Budapest, Hungary. [Falus, Andras] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4., H-1089 Budapest, Hungary. RP Falus, A (reprint author), Semmelweis University, Department of Genetics, Cell and Immunobiology, H-1089 Budapest, Hungary. EM faland@net.sote.hu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 121 EP 124 PG 4 ER PT J AU Toth, E Lazar, I Selmeczi, D Marx, G AF Toth, Esther Lazar, Istvan Selmeczi, David Marx, George TI Lower Cancer Risk in Medium High Radon SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer risk; radon ID cancer risk; radon AB Radon exposure was shown to be carcinogenic and suggested as a possible causative factor for lung cancer in man. A hypothesis is introduced that medium high radon (between 110 and 165 Bq/m3) causes lower cancer risk among women younger than 61 years, independent of the type of cancer. The presented results verify this statement with a probability of not less than 98%. C1 [Toth, Esther] RAD Lauder Laboratory, Budakeszi ut 48., H-1121 Budapest, Hungary. [Lazar, Istvan] RAD Lauder Laboratory, Budakeszi ut 48., H-1121 Budapest, Hungary. [Selmeczi, David] RAD Lauder Laboratory, Budakeszi ut 48., H-1121 Budapest, Hungary. [Marx, George] Eotvos University, Atomic Physics DepartmentBudapest, Hungary. RP Toth, E (reprint author), RAD Lauder Laboratory, H-1121 Budapest, Hungary. EM et@rad.lauder.hu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 125 EP 129 PG 5 ER PT J AU Cserny, G AF Cserny, Gabor TI Proliferative Epithelial Changes in Ectopic Gastric Mucosa of Meckel's Diverticula SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Meckel's diverticulum; ectopic gastric mucosa; reflux gastritis; dysplasia; hyperplasia; p53; Ki-67 ID Meckel's diverticulum; ectopic gastric mucosa; reflux gastritis; dysplasia; hyperplasia; p53; Ki-67 AB Twenty-one Meckel's diverticula containing an adequate amount of assessable heterotopic gastric mucosa were investigated for epithelial changes. Marked or moderate foveolar hyperplasia was present in 52% and 29% of the cases, respectively. Four cases displayed an excessive epithelial proliferation indefinite for dysplasia. It is pointed out that reflux type gastritis or gastropathy, which is the most common lesion in the ectopic gastric mucosa of Meckel's diverticulum, can be associated with the same confusing epithelial proliferation as reflux gastritis in the stomach, but these lesions are best regarded as representing atypia of repair. Distinguishing features from dysplasia are maturation towards the surface, lack of hyperchromatism and abscence of atypical mitoses. Negative p53 immunostaining and localization of the Ki-67 positivity to the expanded neck region could be additive clues that can help to classify lesions indefinite for dysplasia as negative for dysplasia. On the basis of the similarities of the ectopic and ortotopic gastric mucosa, it is suggested that these additive clues previously used in other parts of the digestive tract could also apply for the stomach. C1 [Cserny, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., H-6000 Kecskemet, Hungary. RP Cserny, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.c3.hu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 130 EP 134 PG 5 ER PT J AU Rice, LR Tang, GD Taylor, DJ AF Rice, L Renee Tang, G Dean Taylor, D John TI Actin Cleavage in Various Tumor Cells is not a Critical requirement for executing apoptosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE actin cleavage; apoptosis; caspase ID actin cleavage; apoptosis; caspase AB Actin is a major cytoskeletal protein which is involved in many physiological cellular functions such as motility, cell shape, and adhesion. Recently, actin has also been reported to be cleaved by apoptotic proteases (i.e., caspases) and this cleavage is thought to contribute to the apoptotic process. However, conflicting data also exists as to whether actin represents a true caspase substrate during apoptosis induction in vivo (i.e., inside the cells). In this study, we critically examined the actin cleavage patterns during apoptosis of several tumor cell lines derived from three different species (i.e., mouse, rat, and human). Our findings demonstrate that: 1) actin cleavage in vivo is not a common phenomenon since apoptosis caused by multiple inducers in most cell types examined occurs without evidence of actin degradation; and 2) in certain cell types (e.g., U937), spontaneous, actin cleavage is observed which is not prevented by various specific chemical/peptide inhibitors of proteases such as caspases or serine proteases although apoptosis per se is retarded by some of these inhibitors. Our results conclude that actin is not a critical substrate for apoptotic proteases in vivo during apoptosis. C1 [Rice, L Renee] Wayne State University, Biological Sciences Department, 2105 Biological Science Bldg., 48202 Detroit, MI, USA. [Tang, G Dean] Wayne State University, Department of Radiation OncologyDetroit, USA. [Taylor, D John] Wayne State University, Biological Sciences Department, 2105 Biological Science Bldg., 48202 Detroit, MI, USA. RP Taylor, DJ (reprint author), Wayne State University, Biological Sciences Department, 48202 Detroit, USA. EM cheyenne@sun.science.wayne.edu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 135 EP 145 PG 11 ER PT J AU Zalatnai, A Zala, J Sandor, G AF Zalatnai, Attila Zala, Judit Sandor, Gabor TI Coccidioidomycosis in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE coccidioidomycosis; skin; pathology; human ID coccidioidomycosis; skin; pathology; human AB A case of an isolated subcutaneous coccidioidomycosis in a 61-year-old man is presented. The patient has lived and worked in Arizona for 3 years previously but developed no apparent clinical signs of the disease. The painless, cavitating, tumor-like mass was surgically excised and the diagnosis was established by histological demonstration of the fungi and confirmed by serum counterimmunoelectrophoresis. This represents the first imported case of coccidioidomycosis in Hungary. C1 [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Zala, Judit] Bela Johan National Center for Epidemiology, Mycological DepartmentBudapest, Hungary. [Sandor, Gabor] Central European UniversityBudapest, Hungary. RP Zalatnai, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM zalatnai@korb1.sote.hu NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 147 EP 151 PG 6 ER PT J AU Schmitz, G Aslanidis, Ch Lackner, JK AF Schmitz, Gerd Aslanidis, Charalampos Lackner, J Karl TI Recent Advances in Molecular Genetics of Cardiovascular Disorders - Implications for Atherosclerosis and Diseases of Cellular Lipid Metabolism SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE cardiovascular disease; risk assessment; atherosclerosis; thrombosis; hemostasis; hypertension; obesity; lipoproteins; molecular genetics ID cardiovascular disease; risk assessment; atherosclerosis; thrombosis; hemostasis; hypertension; obesity; lipoproteins; molecular genetics AB Two developments in molecular genetics will profoundly influence our understanding and the diagnosis of cardiovascular disorders. First, the identification of genes responsible for monogenic and polygenic traits by analysis of e.g. large pedigrees and affected sib pairs provides invaluable data regarding the role of specific genes in common diseases like arteriosclerosis, hypertension, diabetes, thrombosis/hemostasis and obesity. Besides the insights into the underlying pathophysiology, this knowledge will permit to identify persons at high risk for disease development. These patients can then obtain a targeted intervention. The second development is related to the availability of new analytical tools for molecular biology. New methods such as sequencing by hybridisation (SBH), DNA-array technology or matrix assisted laser desorption/ionisation-time of flight mass spectroscopy (MALDI-TOF) permit sequence analysis of complete genes within hours. Automated PCR-technologies with homogenous amplicon detection formats simplify PCR and permit its use in the routine laboratory setting. Considering cardiovascular diseases there is a number of genes involved in lipid metabolism (apolipoproteins, lipoprotein receptors, lipolytic enzymes), thrombosis/hemostasis (platelet receptors, pro- and anticoagulant proteins, fibrinogen, PAI's), hypertension (angiotensin converting enzyme, angiotensinogen) glucose metabolism (glucose transporters, enzymes) and obesity (hormones, receptors), that are interesting candidates for sophisticated genetic risk assessment. Furthermore, there are also gene candidates involved in processes of early atherogenesis and chronic inflammation such as complement proteins, cell adhesion molecules, and cellular receptors and enzymes. Most of these gene candidates were derived from pathophysiologic knowledge and subsequent epidemiological studies. However, it is foreseeable that in the coming years genes will be identified which were not known so far to be involved in cardiovascular diseases. Genetic studies will be of prime importance in this area, as is exemplified by animal models. In the long term, analysis of these candidate genes before the implementation of therapy will permit a targeted intervention approach towards high risk patients. This will reduce the overall costs of health care without reducing the quality. C1 [Schmitz, Gerd] University of Regensburg, Institute for Clinical Chemistry and Laboratory Medicine, D-93042 Regensburg, Germany. [Aslanidis, Charalampos] University of Regensburg, Institute for Clinical Chemistry and Laboratory Medicine, D-93042 Regensburg, Germany. [Lackner, J Karl] University of Regensburg, Institute for Clinical Chemistry and Laboratory Medicine, D-93042 Regensburg, Germany. RP Schmitz, G (reprint author), University of Regensburg, Institute for Clinical Chemistry and Laboratory Medicine, D-93042 Regensburg, Germany. EM Gerd.Schmitz@klinik.uni-regensburg.de NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1998 VL 4 IS 2 BP 152 EP 160 PG 9 ER PT J AU Canal, P Gamelin, E Vassal, G Robert, J AF Canal, Pierre Gamelin, Erick Vassal, Gilles Robert, Jacques TI Benefits of Pharmacological Knowledge in the Design and Monitoring of Cancer Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE anticancer drugs; pharmacokinetics; pharmacodynamics; dose adaptation; chemotherapy ID anticancer drugs; pharmacokinetics; pharmacodynamics; dose adaptation; chemotherapy AB Prescribing chemotherapy is a difficult task, because of drug resistance, which prevents all tumors to respond to a given protocol and because of drug toxicity, which is generally unavoidable but which must be limited to acceptable levels. The therapeutic window of anticancer drugs is very narrow and clinicians have to try to optimize the individual doses and schedules of the drugs to be administered. They can rely upon simple anthropometric features, such as body weight or surface area; they can also take into account the physiological status of the patient : age, liver and kidney function, genetic characteristics of drug metabolism, etc. The best way for dose adaptation lies in the establishment of pharmacokinetic/pharmacodynamic relationships, i.e., between the behavior of a drug in the body and its efficacy and toxicity. When it is established that the optimal effect of a drug is related to a given parameter, such as the area under the curve plotting plasma concentration vs. time (AUC), it becomes possible to administer the drug with the dose allowing to obtain the target parameter value. Individual dose adaptation can be achieved thanks to the study of the pharmacokinetics of a test dose preceding that of the therapeutic dose, or by the measure of drug plasma levels, either at steady state during a protracted infusion, or from cycle to cycle during repetitive protocols. Population analysis now allows the adaptation of anticancer drug dosing from a minimum knowledge of individual pharmacokinetic features, together with other characteristics of the patients such as age, gender or physiological functions. C1 [Canal, Pierre] Centre Claudius-Regaud, 20 rue du Pont-Saint-Pierre, 31052 Toulouse, France. [Gamelin, Erick] Centre Paul-Papin, 2 rue Moll, 49033 Angers, France. [Vassal, Gilles] Institute Gustave Roussy, rue Camille-Desmoulins, 94805 Villejuif, France. [Robert, Jacques] Institut Bergonie, Laboratoire de Biochimie et Pharmacologie, 180 rue de Saint-Genes, 33076 Bordeaux, France. RP Robert, J (reprint author), Institut Bergonie, Laboratoire de Biochimie et Pharmacologie, 33076 Bordeaux, France. EM robert@bergonie.org NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1998 VL 4 IS 3 BP 171 EP 178 PG 8 ER PT J AU Tang, GD Li, L Zhu, Z Joshi, B Johnson, RC Marnett, JL Honn, VK Crissman, DJ Krajewski, S Reed, CJ Timar, J Porter, TA AF Tang, G Dean Li, Li Zhu, Zhenyu Joshi, Bindu Johnson, R Carl Marnett, J Lawrence Honn, V Kenneth Crissman, D John Krajewski, Stanislaw Reed, C John Timar, Jozsef Porter, T Arthur TI BMD188, A Novel Hydroxamic Acid Compound, Demonstrates Potent Anti-Prostate Cancer Effects in vitro and in vivo by Inducing Apoptosis: Requirements for Mitochondria, Reactive Oxygen Species, and Proteases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE hydroxamic acid; prostate cancer; apoptosis; free radical; mitochondria; chemotherapy ID hydroxamic acid; prostate cancer; apoptosis; free radical; mitochondria; chemotherapy AB A newly synthesized cyclic hydroxamic acid compound, BMD188 [cis-1-hydroxy-4-(1-naphthyl)-6-octylpiperidine-2-one], was found to induce the apoptotic death of cultured prostate cancer cells by activating caspase-3. Orally administered BMD188 significantly inhibited the primary growth of prostate cancer cells (Du145) orthotopically implanted into SCID mice. Mechanistic studies indicated that BMD188 did not alter the protein levels of several Bcl-2 family members. In contrast, the BMD188 effect required three essential factors: reactive oxygen species (ROS), the mitochondrial respiratory chain function, and proteases. First, the apoptosis-inducing effect of BMD188 could be blocked by ROS scavengers such as Desferal. Second, both BMD188-induced PARP cleavage as well as PC3 cell apoptosis could be dramatically inhibited by several complex-specific mitochondrial respiration blockers. The involvement of mitochondria was also supported by the observations that BMD188 dramatically altered the mitochondrial distribution and morphology without affecting the cellular ATP levels. Finally, the apoptosis-inducing effect of BMD188 in PC3 cells could be significantly inhibited by serine protease inhibitors (TPCK and TLCK) as well as by caspase inhibitors (zVAD-fmk and DEVD-CHO). Collectively, the present study suggests that BMD188 and its analogs may find clinical applications in the treatment of prostate cancer patients by inducing apoptotic death of prostate cancer cells. C1 [Tang, G Dean] Wayne State University, Biomide LaboratoriesDetroit, USA. [Li, Li] Wayne State University, Biomide LaboratoriesDetroit, USA. [Zhu, Zhenyu] Wayne State University, Biomide LaboratoriesDetroit, USA. [Joshi, Bindu] Wayne State University, Biomide LaboratoriesDetroit, USA. [Johnson, R Carl] Wayne State University, Department of ChemistryDetroit, USA. [Marnett, J Lawrence] Vanderbilt University Medical Center, Department of BiochemistryNashville, USA. [Honn, V Kenneth] Wayne State University, Department of Radiation Oncology, 407 Life Science Bldg., 48202 Detroit, MI, USA. [Crissman, D John] Detroit Medical Center, Barbara Ann Karmanos Cancer InstituteDetroit, USA. [Krajewski, Stanislaw] The Burham Institute, Apoptosis & Cell Death ProgramLa Jolla, USA. [Reed, C John] The Burham Institute, Apoptosis & Cell Death ProgramLa Jolla, USA. [Timar, Jozsef] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Porter, T Arthur] Wayne State University, Department of Radiation Oncology, 407 Life Science Bldg., 48202 Detroit, MI, USA. RP Tang, GD (reprint author), Wayne State University, Biomide Laboratories, Detroit, USA. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1998 VL 4 IS 3 BP 179 EP 190 PG 12 ER PT J AU Ongradi, J Specter, S Horvath, A Friedman, H AF Ongradi, Joseph Specter, Steven Horvath, Attila Friedman, Herman TI Combined In Vitro Effect of Marijuana and Retrovirus on the Activity of Mouse Natural Killer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NK activity; retrovirus; marijuana; addition; mouse strains ID NK activity; retrovirus; marijuana; addition; mouse strains AB Both marijuana and retroviruses impair natural killer (NK) cell functions. No data on their simulataneous effects are available. Similarities to human AIDS induced early by Friend leukemia complex (FLC) and its replication competent helper Rowson-Parr virus (RPV) provides a mouse model to study drug-virus action. Leukemia suscep-tible BALB/c and resistant C57BL/6 mice were infected, then at time intervals their nylon wool-separated splenocytes were exposed to tetrahydrocannabinol (THC) for 3h.Natural killer (NK) cell activity against Yac-1 cells was assayed by 51Cr-release for 4 and 18h. Recovery of splenocytes was found to be suppressed by FLC, but in BALB/c only by RPV. After a transient enhancement in C57BL/6 by FLC, NK cell activity of both mice became suppressed early (2 to 4 days), normalized subsequently and enhanced late (11 to 14 days) postinfection. A moderate increase in BALB/c, no change in C57BL/6 were induced by low (1-2.5 µg/ml) THC doses. NK cell activity of BALB/c became suppressed exponentially by higher (5-10 µg/ ml) THC doses in 18h as compared to 4h assays, while its proportional and moderate impairment was seen in C57BL/6. The magnitude of NK cell activity of infected mice was determined by THC: enhancement or impairment followed those of untreated, infected counterparts, but on the level of THC-treated cells. Low doses hardly, high doses additively influenced NK cells of infected BALB/c. THC hardly affected very early and late enhancement in NK cell activiy of FLC infected C57BL/6, but augmented RPV induced suppression late in 18h assays. Genetic factors similar to endotoxin resistance, altered cytokine profile might determine these effects. Similar phenomena in humans might result in earlier manifestation of AIDS. C1 [Ongradi, Joseph] The University of South Florida College of Medicine, Departments of Medicine and Medical Microbiology-ImmunologyTampa, FL, USA. [Specter, Steven] The University of South Florida College of Medicine, Departments of Medicine and Medical Microbiology-ImmunologyTampa, FL, USA. [Horvath, Attila] National Institute for Dermato-Venereology, Maria u. 41., H-1085 Budapest, Hungary. [Friedman, Herman] The University of South Florida College of Medicine, Departments of Medicine and Medical Microbiology-ImmunologyTampa, FL, USA. RP Ongradi, J (reprint author), The University of South Florida College of Medicine, Departments of Medicine and Medical Microbiology-Immunology, Tampa, USA. EM ongjos@bor.sote.hu NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1998 VL 4 IS 3 BP 191 EP 199 PG 9 ER PT J AU Tornoczky, T Kelenyi, G Pajor, L AF Tornoczky, Tamas Kelenyi, Gabor Pajor, Laszlo TI EBER Oligonucleotide RNA in situ Hybridization in EBV Associated Neoplasms SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EBV; EBER; microwave; LMP-1; lymphoma; Warthin's tumor; nasopharyngeal carcinoma ID EBV; EBER; microwave; LMP-1; lymphoma; Warthin's tumor; nasopharyngeal carcinoma AB In virus associated diseases identification of viruses in cells can contribute to the understanding of the pathogenesis and may also help to establish the diagnosis. In the present communication, the effects of the microwave pretreatment (MWP) and that of the proteinase-K enzymatic predigestion (PKD) on EBER RNA oligonucleotide in situ hybridization (EBER-RNA-ISH) (EBER: Epstein-Barr-Encoded-(Early)-RNA) were studied. The efficacy of two EBV detecting methods, latent membrane protein-1 (LMP-1) immunohistochemistry and EBER-RNA-ISH were also compared. Our results show that microwave pretreatment enhances the intensity of the ISH signals and preserves significantly better the structure of the tissues compared with enzymatic predigestion. EBER-RNA-ISH, mainly in the nasopharyngeal carcinoma cases, showed a more frequent positivity than the immunohistochemical reaction for LMP-1, however in case of the Warthin?s tumor only the LMP-1 protein was expressed. C1 [Tornoczky, Tamas] University of Pecs, Department of Pathology, Szigeti ut 12., H-7643 Pecs, Hungary. [Kelenyi, Gabor] University of Pecs, Department of Pathology, Szigeti ut 12., H-7643 Pecs, Hungary. [Pajor, Laszlo] University of Pecs, Department of Pathology, Szigeti ut 12., H-7643 Pecs, Hungary. RP Tornoczky, T (reprint author), University of Pecs, Department of Pathology, H-7643 Pecs, Hungary. EM yst@pathology.pote.hu NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1998 VL 4 IS 3 BP 201 EP 205 PG 5 ER PT J AU Hrzenjak, MT Popovic, M Tiska-Rudman, L AF Hrzenjak, M Terezija Popovic, Maja Tiska-Rudman, Ljerka TI Fibrinolytic Activity of Earthworms Extract (G-90) on Lysis of Fibrin Clots Originated from the Venous Blood of Patients with Malignant Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE fibrinolytic activity; earthworms extract; fibrin clot; malignant tumors ID fibrinolytic activity; earthworms extract; fibrin clot; malignant tumors AB u-PA is secreted by the most malignant tumors. As a response to u-PA synthesis surrounding cells synthetize inhibitors of plasminogen activators for tissue protection. Plasminogen activators were found also in earthworm tissue. From the tissue homogenate of earthworm Eisenia foetida the glycolipoprotein mixture named G-90 was isolated. It contains two serine proteases (P I, P II) with fibrinolytic and anticoagulative activities. The fibrinolytic activity of G-90, P I and P II was tested in an in vitro euglobulinic test applied to fibrin clot from blood plasma of patients suffered from malignant tumors. G-90 and above-mentioned proteases applied in this study showed euglobulinic time proportionally with the concentrations of added substances. The influence of G-90 on the fibrinolysis rate does not depend only on its concentration, but depends too on histological type of tissue (organ) where the malignant tumors are located. Enzyme P I and P II do not show this activity. C1 [Hrzenjak, M Terezija] Faculty of Veterinary Medicine, Department of Biology, Heinzelova 55, 10000 Zagreb, Croatia. [Popovic, Maja] Faculty of Veterinary Medicine, Department of Biology, Heinzelova 55, 10000 Zagreb, Croatia. [Tiska-Rudman, Ljerka] University Hospital for TumorsZagreb, Croatia. RP Hrzenjak, MT (reprint author), Faculty of Veterinary Medicine, Department of Biology, 10000 Zagreb, Croatia. EM mthrzenj@rudjer.irb.hr NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1998 VL 4 IS 3 BP 206 EP 211 PG 6 ER PT J AU Leel-Ossy, L Gati, I AF Leel-Ossy, Lorant Gati, Istvan TI Corpus Amylaceum (Polyglucosan Body) in the Peripheral Olfactory System SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE corpus amylaceum; olfactory tract ID corpus amylaceum; olfactory tract AB Peripheral part of the olfactory system (bulb and tract) was investigated for the occurrence of corpus amylaceum (CA) (polyglucosan body) in 296 (281 pathological and 15 control cases) autopsied human brains. No significant differences were found in the incidence between the various age groups above 40 years or between different disease groups and the controls. The predominance of CA in the olfactory tract and its loose correlation with age at this localization over 40 years of age could be resulted by various factors, including the extremely rich astrocytic and capillary network in the intermediate zone, and the proximity of the olfactory tract to the external environment, which may result in the protective role of CA. The role of stress was proved by the HSP-60 positivity of CA. C1 [Leel-Ossy, Lorant] St. Borbala University Hospital, Department of Pathology, Neuropathological Laboratory, H-2800 Tatabanya, Hungary. [Gati, Istvan] University of Pecs, Department of NeurologyPecs, Hungary. RP Leel-Ossy, L (reprint author), St. Borbala University Hospital, Department of Pathology, Neuropathological Laboratory, H-2800 Tatabanya, Hungary. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1998 VL 4 IS 3 BP 212 EP 216 PG 5 ER PT J AU Szegedi, Zs Amin, F Szende, B Bowen, DI AF Szegedi, Zsolt Amin, Farhana Szende, Bela Bowen, Delme Ivor TI A New Method to Localize Acid Phosphatase Using the Confocal Laser-Scanning Microscope SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE acid phosphatase activity; cytometry; cell image analysis; confocal laser-scanning microscope ID acid phosphatase activity; cytometry; cell image analysis; confocal laser-scanning microscope AB The aim of the study was to work out a technique for the detection of acid phosphatase enzyme activity by confocal laser-scanning microscope using the histochemical acid phosphatase detection method (after Barka and Anderson 1962, modified by Bowen and Lewis 1985) routinely used for light microscopy. The density and the distribution of enzyme reaction product is dependent on the incubation time, as shown by different confocal images or ELISA reader. The inhibition of the enzyme activity with metal ions shows the same profile known from the literature. This staining method seems to be useful to demonstrate subcellular distribution of the enzyme in the lysosomes and in the Golgi apparatus. C1 [Szegedi, Zsolt] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, Ulloi ut 26., 1085 Budapest, Hungary. [Amin, Farhana] University of Cardiff, Department of Pure and Applied BiologyCardiff, Wales, UK. [Szende, Bela] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, Ulloi ut 26., 1085 Budapest, Hungary. [Bowen, Delme Ivor] University of Cardiff, Department of Pure and Applied BiologyCardiff, Wales, UK. RP Szegedi, Zs (reprint author), Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, 1085 Budapest, Hungary. EM szezso@korb1.sote.hu NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1998 VL 4 IS 3 BP 217 EP 223 PG 7 ER PT J AU Maldonado, JC Palazzolo, K Taylor, DJ AF Maldonado, J Carlos Palazzolo, Katherine Taylor, D John TI A Novel In Vitro System to Study Extravasated Tumor Cell-Induced Angiogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE blood vessels; collagen gel; extravasation; invasion; in vitro; metastasis; secondary angiogenesis ID blood vessels; collagen gel; extravasation; invasion; in vitro; metastasis; secondary angiogenesis AB Angiogenesis, the formation of new blood vessels from preexisting ones, is a fundamental stage in the metastatic pathway. For the primary tumor, this neovascularization provides nutrients and oxygen as well as a route by which metastatic tumor cells gain access to the circulatory system. Among these metastatic tumor cells, there are subgroups of cells that express an angiogenesis-inducing cells phenotype (AICs) as well as others that do not. Tumor cells not expressing the angiogenesis-inducing cells phenotype (non-AICs) invade new tissues and remain as dormant micrometastases unless they accompany AICs. Thus, either alone or with non-AICs, angiogenesis-inducing cells form rapidly growing, clinically detectable metastases. Much of the current research in this area is concentrated on the vascularization of primary tumors, but the regulation of angiogenesis by extravasating or invading tumor cells has not being extensively studied. We have developed a working model, which demonstrates that human metastatic prostate cancer cells (PC-3) appear to induce human vascular endothelial cells (HUVECs) to translocate across a Matrigel-coated 8 ?m membrane. The parameters of this model (i.e. pore size, seeding-cell density, seeding times) were established using highly invasive murine melanoma cells (B16F10) seeded on murine microvascular endothelial cells (CD3). We have further modified our model in order to include a host compartment made of collagen gel, in order to mimic the in vivo site of metastases-induced angiogenesis. C1 [Maldonado, J Carlos] Wayne State University, Biological Sciences Department, 5047 Gullen Mall, 48202 Detroit, MI, USA. [Palazzolo, Katherine] Wayne State University, Biological Sciences Department, 5047 Gullen Mall, 48202 Detroit, MI, USA. [Taylor, D John] Wayne State University, Biological Sciences Department, 5047 Gullen Mall, 48202 Detroit, MI, USA. RP Taylor, DJ (reprint author), Wayne State University, Biological Sciences Department, 48202 Detroit, USA. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1998 VL 4 IS 3 BP 225 EP 229 PG 5 ER PT J AU Albini, A AF Albini, Adriana TI Tumor and Endothelial Cell Invasion of Basement Membranes The Matrigel Chemoinvasion Assay As A Tool For Dissecting Molecular Mechanisms SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE invasion; metastasis; angiogenesis; basement membranes; matrigel; Boyden chamber; chemoinvasion ID invasion; metastasis; angiogenesis; basement membranes; matrigel; Boyden chamber; chemoinvasion AB The spread of cancer cells from a primary tumor to distant organs is the major cause of death of cancer patients. Metastatic lesions are often resistent to cancer therapy because of the progressive phenotypic changes that they have undergone. Several genetic and epigenetic factors, both in the cell and in the host, contribute to the development of tumor progression towards metastases. In this review we will analyze the steps involved in tumor metastases, which can be potential targets for anti-metastatic therapy. One of the most critical events in cancer metastasis is the invasion of basement membranes. An assay which we developed over ten years ago, the matrigel ''chemoinvasion'' assay, has been a useful tool for studying the mechanisms involved in tumor and endothelial cell invasion of basement membranes and for the screening of anti-invasive agents. Here we will describe the assay and review some of the major results obtained with it. C1 [Albini, Adriana] National Institute for Research on Cancer, Advanced Biotechnology Center, Largo Rosanna Benzi 10, 16132 Genova, Italy. RP Albini, A (reprint author), National Institute for Research on Cancer, Advanced Biotechnology Center, 16132 Genova, Italy. EM albini@sirio.cba.unige.it NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1998 VL 4 IS 3 BP 230 EP 241 PG 12 ER PT J AU Szakacs, G Jakab, K Antal, F Sarkadi, B AF Szakacs, Gergely Jakab, Katalin Antal, Ferenc Sarkadi, Balazs TI Diagnostics of Multidrug Resistance in Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE P-glycoprotein; MDR1; MRP; multidrug transporters; diagnostic methods ID P-glycoprotein; MDR1; MRP; multidrug transporters; diagnostic methods AB Multidrug resistance (MDR), caused by the overexpression of two membrane proteins, MDR1-Pgp and/or MRP, is a major obstacle in the chemotherapy of cancer. The proper laboratory diagnosis of clinical multidrug resistance is still an unresolved question, and this uncertainty, in a vicious cycle, does not allow the correct evaluation of the clinical relevance of the MDR phenomenon. Moreover, inefficient MDR diagnostics hinders the development of effective resistance-modulation strategies. In this review, after describing the basic features of the MDR drug pump proteins, the currently employed diagnostic methods are discussed. We suggest that a quantitative, functional method developed in our laboratory may provide a major help in the laboratory assessment of cancer MDR. C1 [Szakacs, Gergely] National Institute of Haematology, Blood Transfusion and Immunology, Daroczi u. 24., H-1113 Budapest, Hungary. [Jakab, Katalin] National Institute of Haematology, Blood Transfusion and Immunology, Daroczi u. 24., H-1113 Budapest, Hungary. [Antal, Ferenc] National Institute of Haematology, Blood Transfusion and Immunology, Daroczi u. 24., H-1113 Budapest, Hungary. [Sarkadi, Balazs] National Institute of Haematology, Blood Transfusion and Immunology, Daroczi u. 24., H-1113 Budapest, Hungary. RP Sarkadi, B (reprint author), National Institute of Haematology, Blood Transfusion and Immunology, H-1113 Budapest, Hungary. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 251 EP 257 PG 7 ER PT J AU Croce, VM Colussi, GA De Bravo, G Price, RM Segal-Eiras, A AF Croce, V Maria Colussi, G Andrea De Bravo, M. Garcia Price, R Michel Segal-Eiras, Amanda TI Immunohistopathological Characterization of Spontaneous Metastases in a Human Lung Mucoepidermoid Adenocarcinoma (HLMC) Xenograft SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistopathology; metastases; lung adenocarcinoma xenograft ID immunohistopathology; metastases; lung adenocarcinoma xenograft AB The most common clinical form of lung cancer is a disseminated disease with distant metastases; several years of cancer progression precede presentation, and this ultimately limits the efficacy of curative therapy. In this immunohistochemical study, we examined a mucinous adenocarcinoma cell line, maintained by xenogeneic transplantation, and a spontaneous metastatic variant which produces distant tumors (in liver, spleen and kidney). The aim was to investigate possible parameters which characterize the metastatic process. Histopathological comparison between the two subcutaneous transplanted tumor lines showed that both lines presented a similar cellular morphology, a different pattern of cellular growth and an increased vascularization in the metastatic line with respect to its parent. All the tumor sections expressed differential immune reactivity with monoclonal antibodies against Lewis y (MAb C14), sialyl-Lewis x (MAb SNH3) and Lewis x (MAb FH2) determinants. Neither expressed MUC 1 mucins detectable with monoclonal antibodies reactive with the mucin protein core (MAbs C595 and SM3) nor was carcinoembryonic antigen (MAb C365) expressed. Neoplastic cells were reactive with an anti-pan cytokeratin monoclonal antibody confirming their epithelial histogenesis. Our findings have been evaluated with respect to defining metastatic phenotypes in lung cancer by examination of distinct histopathological and immunological parameters. C1 [Croce, V Maria] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA)La Plata, Argentina. [Colussi, G Andrea] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA)La Plata, Argentina. [De Bravo, M. Garcia] National University of La Plata, IMBIOLPLa Plata, Argentina. [Price, R Michel] University of Nottingham, School of Pharmaceutical Sciences, Cancer Research LaboratoriesNottingham, UK. [Segal-Eiras, Amanda] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA)La Plata, Argentina. RP Segal-Eiras, A (reprint author), Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), La Plata, Argentina. EM aeiras@satlink.com NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 259 EP 266 PG 8 ER PT J AU Sipos, L Szegedi, Zs Fedorcsak, I Afra, D Szende, B AF Sipos, Laszlo Szegedi, Zsolt Fedorcsak, Imre Afra, Denes Szende, Bela TI Apoptosis and p53 Expression in Human Gliomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apoptosis; p53 expression; gliomas; grade ID apoptosis; p53 expression; gliomas; grade AB Twenty-five human gliomas of different histological grade and type were studied for p53 expression by immunohistochemistry and for apoptosis using ApopTagTM method. p53 expression (percentage of positive cells) was highest in anaplastic astrocytomas, followed by low grade astrocytomas and surprisingly in glioblastomas. Granular cytoplasmic p53 positivity appeared in 4/5 low grade oligodendroglioma and in 2/5 low grade mixed oligoastrocytomas. The means of apoptosis index in the different tumor types ranged between 0.8 and 11.5 with the highest values in anaplastic astrocytoma and glioblastomas. Although the number of cases per group were relatively low and the individual vales showed differences it seems that p53 expression is related to the biological aggressiveness of gliomas. It is also suggested that high level of apoptosis in malignant glioma could represent a p53 independent pathway. C1 [Sipos, Laszlo] National Institute of NeurosurgeryBudapest, Hungary. [Szegedi, Zsolt] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Fedorcsak, Imre] National Institute of NeurosurgeryBudapest, Hungary. [Afra, Denes] National Institute of NeurosurgeryBudapest, Hungary. [Szende, Bela] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Szende, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM bszende@korb1.sote.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 267 EP 270 PG 4 ER PT J AU Epelbaum, R Shalitin, Ch Segal, R Valansi, C Arselan, I Faraggi, D Leviov, M Ben-Shahar, M Haim, N AF Epelbaum, Ron Shalitin, Channa Segal, Ruth Valansi, Clari Arselan, Ida Faraggi, David Leviov, Michelle Ben-Shahar, Menahem Haim, Nissim TI Haptoglobin-related Protein as a Serum Marker in Malignant Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Haptoglobin-related protein; 21-kDa protein; ELISA; lymphoma ID Haptoglobin-related protein; 21-kDa protein; ELISA; lymphoma AB A novel serum 21 kDa haptoglobin-related protein (Hpr) was investigated in patients with malignant lymphoma, to evaluate its correlation with clinical and histologic features at presentation and its possible role as a tumor marker for patient outcome. One hundred fifty eight serum samples were taken from 88 patients with non-Hodgkin's lymphoma (n=58) and Hodgkin's disease (n=30) at presentation and in the course of follow-up. Sera from 61 healthy volunteers served as normal controls. Serum Hpr levels in the lymphoma patients (median 430x103 u/ml, range 0-4000x103) were significantly higher than in the control group (median 68x103 u/ml, range 0-180x103) (p=0.0001). Higher median Hpr values were detected in patients with advanced disease (p=0.013), ''B'' symptoms (p=0.029) and in males (p=0.053). There was also a significant correlation between Hpr and erythrocyte sedimentation rate (p=0.028). Serial determinations showed a significant decrease of the initial Hpr values obtained after treatment in 41 patients, 38 of whom achieved complete remission. In the follow-up period additional Hpr measurements were taken from 17 patients. Three of them eventually relapsed, and showed increased Hpr levels at the time of relapse. Hpr levels remained low in 11 of 14 patients who maintained complete remission, and increased in three. In conclusion, serum Hpr is a new serum tumor marker of potential use in the clinical setting of lymphoma. C1 [Epelbaum, Ron] Rambam Medical Center, Department of Oncology, 31096 Haifa, Israel. [Shalitin, Channa] Technion-Israel Intitute of Technology, Department of BiologyHaifa, Israel. [Segal, Ruth] Rambam Medical Center, Department of Oncology, 31096 Haifa, Israel. [Valansi, Clari] Technion-Israel Intitute of Technology, Department of BiologyHaifa, Israel. [Arselan, Ida] University of Haifa, Department of StatisticsHaifa, Israel. [Faraggi, David] University of Haifa, Department of StatisticsHaifa, Israel. [Leviov, Michelle] Rambam Medical Center, Department of Oncology, 31096 Haifa, Israel. [Ben-Shahar, Menahem] Rambam Medical Center, Department of Oncology, 31096 Haifa, Israel. [Haim, Nissim] Rambam Medical Center, Department of Oncology, 31096 Haifa, Israel. RP Epelbaum, R (reprint author), Rambam Medical Center, Department of Oncology, 31096 Haifa, Israel. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 271 EP 276 PG 6 ER PT J AU Szendroi, M Arato, G Ezzati, A Huttl, K Szavcsur, P AF Szendroi, Miklos Arato, Gabriella Ezzati, Ali Huttl, Kalman Szavcsur, Peter TI Aneurysmal Bone Cyst: its Pathogenesis Based on Angiographic, Immunohistochemical and Electron Microscopic Studies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE aneurysmal bone cyst; pathogenesis; angiography; histology ID aneurysmal bone cyst; pathogenesis; angiography; histology AB Based on angiographic, immunohistochemical as well as electron microscopic findings, authors outline a hypothesis for the etiopathogenesis of aneurysmal bone cysts. No changes were found at the arterial site in 16 studied aneurysmal bone cysts, with no signs of an arteriovenous shunt. In certain cases, however, dilated and tortous efferent veins became visible in the late venous phase. Due to the impedance of venous flow, the intracystic pressure increases and the small veins become dilated causing formation of aneurysmal slits. This is supported by the immunohistochemical finding that S-actin shows concentric arrangement around the aneurysmal cavities. Endothelial lining and basal membrane remnants were detectable in places, though the aneurysmal slits were devoid of continuous endothelial lining and basal membrane. We suggested that the aneurysmal bone cyst corresponds to a hemodynamic disturbance and is due to primary or secondary venous malformation of the bones. C1 [Szendroi, Miklos] Semmelweis University, Department of Orthopedics, Karolina ut 27, H-1113 Budapest, Hungary. [Arato, Gabriella] Haynal Imre University of Health Sciences, Institute of PathologyBudapest, Hungary. [Ezzati, Ali] Semmelweis University, Department of Orthopedics, Karolina ut 27, H-1113 Budapest, Hungary. [Huttl, Kalman] Semmelweis University, Department of Cardiovascular SurgeryBudapest, Hungary. [Szavcsur, Peter] National Institute of Oncology, Center of RadiologyBudapest, Hungary. RP Szendroi, M (reprint author), Semmelweis University, Department of Orthopedics, H-1113 Budapest, Hungary. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 277 EP 281 PG 5 ER PT J AU Nishimura, Y Sameni, M Sloane, FB AF Nishimura, Yukio Sameni, Mansoureh Sloane, F Bonnie TI Malignant Transformation Alters Intracellular Trafficking of Lysosomal Cathepsin D in Human Breast Epithelial Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cathepsin D; aspartic proteinase; lamp; microtubule; trafficking; breast cancer; Ras ID cathepsin D; aspartic proteinase; lamp; microtubule; trafficking; breast cancer; Ras AB Increased expression and alteration of intracellular trafficking of lysosomal cathepsins have been reported in malignant tumors, or in cells transformed by the transfection with the ras oncogene. In the present study, immortal MCF-10A human breast epithelial cells were transformed with the mutated ras oncogene. Both cell lines were investigated for changes in the intracellular localization of lysosomal cathepsin D and lamp-1 (lysosome-associated membrane protein) employing specific antibodies and confocal immunofluorescence microscopy. The results revealed that staining for cathepsin D along with for lamp-1 was mostly localized in the perinuclear region of MCF-10A cells. In contrast, the staining for these proteins was found to be widely distributed throughout the cytoplasm and at the cell periphery in MCF-10AneoT cells. The organization of microtubules, but not actin, appeared to differ between MCF-10A cells and their oncogenic ras transfectants. When the microtubules were depolymerized by treatment of MCF-10A cells with nocodazole, vesicles containing the lysosomal cathepsin D were dispersed in the cytoplasm and translocation of these vesicles to the cell periphery was observed. The intracellular localization of cathepsin D in the nocodazole-treated MCF-10A cells seemed to be similar to that observed in the oncogenic ras transfectants of these cells. When taxol, which inhibits microtubule depolymerization, was added to the culture medium of neoT cells, a polymerized microtubule network was observed, and the reclustering of cathepsin D and lamp-1 occurred in a unidirectional manner towards the perinuclear region. These findings support a model in which cytoskeletal microtubule organization is closely related to the trafficking of lysosomes/endodomes, and in which oncogenic ras interferes with such organization in human breast epithelial cells. C1 [Nishimura, Yukio] Kyushu University, Faculty of Pharmaceutical Sciences, 3-1-1 Maidashi, Higashi-ku, 812-0054 Fukuoka, Japan. [Sameni, Mansoureh] Wayne State University, Department of PharmacologyDetroit, USA. [Sloane, F Bonnie] Wayne State University, Department of PharmacologyDetroit, USA. RP Nishimura, Y (reprint author), Kyushu University, Faculty of Pharmaceutical Sciences, 812-0054 Fukuoka, Japan. EM nishimur@bioc.phar.kyushu-u.ac.jp NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 283 EP 296 PG 14 ER PT J AU Bajtai, A Hidvegi, J AF Bajtai, Attila Hidvegi, Judit TI The Role of Gastric Mucosal Dysplasia in the Development of Gastric Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gastric dysplasia; gastric cancer ID gastric dysplasia; gastric cancer AB It is widely acknowledged that most stomach carcinomas developed from dysplastic lesions of gastric mucosa. They can be found in known precancerous conditions as chronic gastritis, gastric adenoma, giant rugal hypertrophy, chronic peptic ulcer, gastric stump after partial resection and pernicious anemia. Several grading systems of gastric dysplasia have been suggested. Nagayo's or the ISGGC classification was applied to 367 biopsy specimens of 258 patients between 1979-1995. The frequency of moderate and severe dysplasia was 0,84% regarding all gastric endoscopies in the same period of time. Follow-up studies were performed in 56 cases in a period of 1-7 ys. In this group cancer developed during 14 patients. The authors' recommendation is to follow up the patients gastric dysplasia for least 10 ys after with diagnosis. C1 [Bajtai, Attila] Fovarosi Uzsoki utcai Korhaz, Pathologia, Uzsoki u. 29., H-1145 Budapest, Hungary. [Hidvegi, Judit] Fovarosi Uzsoki utcai Korhaz, Pathologia, Uzsoki u. 29., H-1145 Budapest, Hungary. RP Bajtai, A (reprint author), Fovarosi Uzsoki utcai Korhaz, Pathologia, H-1145 Budapest, Hungary. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 297 EP 300 PG 4 ER PT J AU Cserni, G AF Cserni, Gabor TI The Impact of Axillary Lymphadenopathy on Further Treatment in Breast Cancer? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; axillary metastasis; clinical staging; sampling; sentinel lymph node biopsy ID breast cancer; axillary metastasis; clinical staging; sampling; sentinel lymph node biopsy AB Clinical assessment is an important part of the breast cancer patients' work-up, but it has low sensitivity and specificity. In a retrospective study, histological slides of axillary clearance specimens were used to model palpability of the axillary lymph nodes. Obvious nodes (enlarged and involving considerable amount of lymphatic and/or metastatic tissue) and nodes equal to or larger than 1 cm or 1.5 cm were counted and the slides were subsequently reviewed. The false positive and negative rates expected on the basis of the model ranged from 24 to 72% and from 10 to 38%, respectively. This model (also valid for intraoperative assessment of nodal status by palpation) documents the lack of specificity of clinical staging of the axilla. These results question the practice of excluding patients with palpable axillary lymph node enlargement from less radical staging procedures such as axillary sampling or sentinel node biopsy C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., H-6000 Kecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.c3.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 301 EP 303 PG 3 ER PT J AU Soda, G Baiocchini, A Bosco, D Nardoni, S Melis, M AF Soda, Giuseppe Baiocchini, Andrea Bosco, Daniela Nardoni, Stefano Melis, Marco TI Oral Focal Mucinosis of the Tongue SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE focal mucinosis; oral ID focal mucinosis; oral AB Oral focal mucinosis (OFM) is an uncommon clinicopathological entity which is considered to be the oral counterpart of cutaneous focal mucinosis and cutaneous myxoid cyst. It is comprised of a clinically elevated mass with a histological feature of localised areas of myxomatous connective tissue. The present study adds a rare case of OFM of the tongue to the literature, and we present a review of the most characteristic oral myxomatous lesions. C1 [Soda, Giuseppe] University of Rome, Campus Bio-Medico, Department of Pathology, V. Le Regina Elena 324, 00161 Rome, Italy. [Baiocchini, Andrea] University of Rome, Campus Bio-Medico, Department of Pathology, V. Le Regina Elena 324, 00161 Rome, Italy. [Bosco, Daniela] University of Rome, Campus Bio-Medico, Department of Pathology, V. Le Regina Elena 324, 00161 Rome, Italy. [Nardoni, Stefano] University of Rome, Campus Bio-Medico, Department of Pathology, V. Le Regina Elena 324, 00161 Rome, Italy. [Melis, Marco] University of Rome, Campus Bio-Medico, Department of Pathology, V. Le Regina Elena 324, 00161 Rome, Italy. RP Melis, M (reprint author), University of Rome, Campus Bio-Medico, Department of Pathology, 00161 Rome, Italy. EM mmelis@axrma.uniromal.it NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 304 EP 307 PG 4 ER PT J AU Luttwak, Z Koren, R Veltman, V Halpern, M Manes, A Lask, D Siegal, A AF Luttwak, Zvi Koren, Rumelia Veltman, Vladimir Halpern, Marisa Manes, Aaron Lask, Dov Siegal, Annette TI Leiomyoma of the Urinary Bladder SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE bladder; leiomyoma ID bladder; leiomyoma AB Leiomyoma of the urinary bladder is a rarity but should be considered in the differential diagnosis of an intramural neoplasm. We report a case illustrating clinical and pathological features in particular the immunohistochemistry. Etiology and differential diagnosis are discussed. C1 [Luttwak, Zvi] Hasharon Hospital, Rabin Medical Center, Petah-Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Department of UrologyTel Aviv, Israel. [Koren, Rumelia] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Veltman, Vladimir] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Halpern, Marisa] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Manes, Aaron] Hasharon Hospital, Rabin Medical Center, Petah-Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Department of UrologyTel Aviv, Israel. [Lask, Dov] Hasharon Hospital, Rabin Medical Center, Petah-Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Department of UrologyTel Aviv, Israel. [Siegal, Annette] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. RP Koren, R (reprint author), A, Rabin Medical Center (Golda Campus), Department of Pathology, Petach Tikvah, Israel. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 308 EP 309 PG 2 ER PT J AU Naidoo, R Chetty, R AF Naidoo, Richard Chetty, Runjan TI The Application of Microsatellites in Molecular Pathology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE microsatellites; microsatellite analysis; microsatellite instability; allelic imbalance; genetic linkage; automated DNA sequencing ID microsatellites; microsatellite analysis; microsatellite instability; allelic imbalance; genetic linkage; automated DNA sequencing AB Present within the genome are large numbers of seemingly unimportant DNA segments arranged in repetitive units. Furthermore, these stretches of DNA contain variations or polymorphisms that are characteristic for an individual and results in a unique DNA fingerprint. Approximately 30% of the DNA repeat sequences are arranged as short tandem repeat sequences, which are called microsatellites. Microsatellites may consist of 1, 2 or 3 nucleotides; dinucleotides being the commonest. Microsatellites are characterised by being: stably inherited and hence highly conserved from one generation to the next, and unique to an individual and the same in different cells from the same individual. As a result of the above features, microsatellites can be used for personal identification, population genetic analysis and construction of evolutionary trees. In addition, they are located in several important gene loci and this allows microsatellites to be used as markers of disease and to provide information about individual gene status, especially in tumors. This can be accomplished by assessing allelic imbalance or loss of heterozygosity of a particular gene by analysing microsatellites located at specific loci in the gene. Recently, mutations within microsatellites have been described as a result of defective DNA repair mechanisms, resulting in the phenomenon of microsatellite instability. This has been implicated in the aetiopathogenesis of several hereditary and non-hereditary conditions. There are several ways of analysing microsatellites, the popular using radioactively-labelled primers and autoradiography. This method has several drawbacks, especially the use of radioactivity and interpretative/technical problems. The use of fluorescently-labelled primers, automated DNA sequencing coupled with a computer software package obviates these problems. This technique has the added advantage of analysing several microsatellites in large numbers of cases, simultaneously. Thus, microsatellite analysis has become an important investigative tool for the molecular biologist and has provided new information in many diseases. C1 [Naidoo, Richard] University of Natal School of Medicine, Molecular Biology Research Facility and Department of Pathology, Private Bag 7, 4013 Congella, Durban, South Africa. [Chetty, Runjan] University of Natal School of Medicine, Molecular Biology Research Facility and Department of Pathology, Private Bag 7, 4013 Congella, Durban, South Africa. RP Naidoo, R (reprint author), University of Natal School of Medicine, Molecular Biology Research Facility and Department of Pathology, 4013 Congella, South Africa. EM naidoor@med.und.ac.za NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 310 EP 315 PG 6 ER PT J AU Soti, Cs Csermely, P AF Soti, Csaba Csermely, Peter TI Molecular Chaperones in the Etiology and Therapy of Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE chaperone; heat shock protein; stress protein; metastasis; immunotherapy; drug resistance ID chaperone; heat shock protein; stress protein; metastasis; immunotherapy; drug resistance AB Molecular chaperones are ubiquitous, well-conserved proteins that account for 2-5 % of all cellular proteins in most cells. The present review summarizes our current knowledge about their involvement in the etiology and therapy of cancer with special emphasis on the expression of chaperones in malignant cells, their role in folding of (proto)oncogene products, cell cycle regulation, cell differentiation and apoptosis, development of metastasis, and their participation in the recognition of malignant cells. We also overview the importance of chaperones in hyperthermia, drug resistance, and recent approaches in chaperone-immunotherapy. C1 [Soti, Csaba] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, H-1444 Budapest, Hungary. [Csermely, Peter] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, H-1444 Budapest, Hungary. RP Csermely, P (reprint author), Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, H-1444 Budapest, Hungary. EM csermely@puskin.sote.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 1998 VL 4 IS 4 BP 316 EP 321 PG 6 ER PT J AU Clay, MT Custer, CM Spiess, JP Nishimura, IM AF Clay, M Timothy Custer, C Mary Spiess, J Paul Nishimura, I Michael TI Potential Use of T Cell Receptor Genes to Modify Hematopoietic Stem Cells for the Gene Therapy of Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE adoptive immunotherapy; gene therapy; stem cell; T cell receptor; tumor antigen ID adoptive immunotherapy; gene therapy; stem cell; T cell receptor; tumor antigen AB The purpose of this review is to illustrate some of the technical and biological hurdles that need to be addressed when developing new gene therapy based clinical trials. Gene transfer approaches can be used to ''mark” cells to monitor their persistence in vivo in patients, to protect cells from toxic chemotherapeutic agents, correct a genetic defect within the target cell, or to confer a novel function on the target cell. Selection of the most suitable vector for gene transfer depends upon a number of factors such as the target cell itself and whether gene expression needs to be sustained or transient. The TCR gene transfer approach described here represents one innovative strategy being pursued as a potential therapy for metastatic melanoma. Tumor reactive T cells can be isolated from the tumor infiltrating lymphocytes (TIL) of melanoma patients. A retroviral vector has been constructed containing the T cell receptor (TCR) &agr; and &bgr; chain genes from a MART-1(27-35)-specific T cell clone (TIL 5). Jurkat cells transduced with this virus specifically release cytokine in response to MART-1(27-35) peptide pulsed T2 cells, showing that the virus can mediate expression of a functional TCR. HLA-A2 transgenic mice are being used to examine whether transduced bone marrow progenitor cells will differentiate in vivo into mature CD8+ T cells expressing the MART-1(27-35)-specific TCR. Expression of the human TCR &agr; and &bgr; chain genes has been detected by RT-PCR in the peripheral blood of HLA-A2 transgenic mice reconstituted with transduced mouse bone marrow. Expression of the TIL 5 TCR genes in the peripheral blood of these mice was maintained for greater than 40 weeks after bone marrow reconstitution. TIL 5 TCR gene expression was also maintained following transfer of bone marrow from mice previously reconstituted with transduced bone marrow to secondary mouse recipients, suggesting that a pluripotent progenitor or lymphocyte progenitor cell has been transduced. C1 [Clay, M Timothy] National Institutes of Health, National Cancer Institute, Surgery Branch, Building 10, Room 2B04, 10 Center Drive, 20892 Bethesda, MD, USA. [Custer, C Mary] National Institutes of Health, National Cancer Institute, Surgery Branch, Building 10, Room 2B04, 10 Center Drive, 20892 Bethesda, MD, USA. [Spiess, J Paul] National Institutes of Health, National Cancer Institute, Surgery Branch, Building 10, Room 2B04, 10 Center Drive, 20892 Bethesda, MD, USA. [Nishimura, I Michael] National Institutes of Health, National Cancer Institute, Surgery Branch, Building 10, Room 2B04, 10 Center Drive, 20892 Bethesda, MD, USA. RP Clay, MT (reprint author), National Institutes of Health, National Cancer Institute, Surgery Branch, 20892 Bethesda, USA. EM Tim_Clay@nih.gov.usa CR Rosenberg SA, Abersold P, Cornetta K, et al: Gene transfer into humans: immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. N Engl J Med 323:570-578, 1990. Blaese RM, Culver KW, Miller AD, et al: T lymphocyte-directed gene therapy for ADA-SCID: initial trial results after 4 years. Science 270:475-480, 1995. Partridge TA, Davies KE: Myoblast-based gene therapies. Br Med Bull 51:123-137, 1995. Alton EW, Middleton PG, Caplen NJ, et al: Non-invasive liposome- mediated gene delivery can correct the ion transport defect in cystic fibrosis mutant mice. Nat Genet 5:135-142, 1993. Favrot M, Coll JL, Louis N, et al: Cell death and cancer: replacement of apoptotic genes and inactivation of death suppressor genes in therapy. Gene Ther 5:728-739, 1998. Vandendriessche T, Chuah MK, Chiang L, et al: Inhibition of clinical human immunodeficiency virus, HIV, type I isolates in primary CD4+ T lymphocytes by retroviral vectors expressing anti-HIV genes. J Virol 69:4045-4052, 1995. Pardoll D: Immunotherapy with cytokine gene-transduced tumor cells: the next wave in gene therapy for cancer. Curr Opin Oncol 4:1124-1129, 1992. Licht T, Herrmann F, Gottesman MM, et al: In vivo drug-selectable genes: a new concept in gene therapy. Stem Cells 15:104- 111, 1997. Greenberg PD, Finch RJ, Gavin MA, et al: Genetic modification of T-cell clones for therapy of human viral and malignant diseases. The Cancer J from Scientific American 4, suppl. 1): S100-S105, 1998. Pogulis RJ, Hansen MJ, Pease LR: Retroviral-mediated expression of an MHC class I-restricted T cell receptor in the CD8 T cell compartment of bone marrow-reconstituted mice. Human Gene Ther 9:2285-2297, 1998. Brenner MK: The contribution of marker gene studies to hemopoietic stem cell therapies. Stem Cells 13:453-461, 1995. Kessler PD, Podasakoff GM, Chen X, et al: Gene delivery to skeletal muscle results in sustained expression and systemic delivery of a therapeutic protein. Proc Natl Acad Sci USA 93:14082-14087, 1996. Fisher KJ, Jooss K, Alston J, et al: Recombinant adeno-associated virus for muscle directed gene therapy. Nat Med 3:306- 312, 1997. Monahan PE, Samulski RJ, Tazelaar J, et al: Direct intramuscular injection with recombinant AAV vectors results in sustained expression in a dog model of hemophilia. Gene Ther 5:40-49, 1998. Yang Y, Li Q, Ertl HC, et al: Cellular and humoral immune responses to viral antigens create barriers to lung-directed gene therapy with recombinant adenoviruses. J Virol 69:2004-2015, 1995. Miller AD, Rosman GJ: Improved retroviral vectors for gene transfer and expression. Biotechniques 7:980-990, 1989. Treisman J, Hwu P, Minamoto S, et al: Interleukin-2-transduced lymphocytes grow in an autocrine fashion and remain responsive to antigen. Blood 85:139-145, 1995. Markowitz D, Goff S, Bank A: A safe packaging line for gene transfer: separating viral genes on two different plasmids. J Virol 62:1120-1124, 1988. Miller AD, Garcia JV, von Suhr N, et al: Construction and properties of retrovirus packaging cells based on gibbon ape leukemia virus. J Virol 65:2220-2224, 1991. Miller AD, Buttimore C: Redesign of retrovirus packaging cell lines to avoid recombination leading to helper virus production. Molec Cell Biol 6:2895-2902, 1986. Pockaj BA, Sherry RM, Wei JP, et al: Localization of 111Indiumlabelled tumor infiltrating lymphocytes to tumor in patients receiving adoptive immunotherapy. Augmentation with cyclophosphamide and correlation with response. Cancer 15:1731- 1737, 1994. Kast WM, Offringa R, Peters PJ, et al: Eradication of adenovirus E1-induced tumors by E1A-specific cytotoxic T lymphocytes. Cell 59:603-614, 1989. Rodolfo M, Bassi C, Salvi C, et al: Therapeutic use of a longterm T cell line recognizing a common tumor associated antigen: the pattern of in vitro reactivity predicts the in vivo effect on different tumors. Cancer Immunol Immunother 34:53-63 1991. Feltkamp MC, Vreugenhil GR, Vierboom MPM, et al: Cytotoxic T lymphocytes raised against a subdominant epitope offered as a synthetic peptide eradicate human papillomavirus type 16-induced tumors. Eur J Immunol 25:2638-2642, 1995. Burger UL, Chang MP, Nagoshi M, et al: Improved in vivo efficiency of tumor-infiltrating lymphocytes after restimulation with irradiated tumor cells in vitro. Ann Surg Oncol 3:580-587, 1996. Shilyansky J, Yang JC, Custer MC, et al: Identification of a Tcell receptor from a therapeutic murine T-cell clone. J Immunother Emphasis Tumor Immunol 20:247-255, 1997. Rosenberg SA, Yannelli JR, Yang JC, et al: Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. J Natl Cancer Inst 86:1159- 1166, 1994. Kawakami Y, Eliyahu S, Sakaguchi K, et al: Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes. J Exp Med 180:347-352, 1994. Kawakami Y, Eliyahu S, Delgado CH, et al: Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor. Proc Natl Acad Sci USA 91:3515-3519, 1994. Cole DJ, Weil J, Shilyansky J, et al: Characterization of the functional specificity of a cloned T-cell receptor heterodimer recognizing the MART-1 melanoma antigen. Cancer Res 55:748-752, 1995. Kawasaki ES: In: PCR Protocols, eds. Innis, MA, Gelfand, DH, Sninsky, JJ and White, TJ, 146-152, Academic Press, San Diego, CA, 1990). Orkin SH, Motulsky AG: Report and recommendations of the panel to assess the NIH investment in research on gene therapy. NIH panel report. 1996. Blaese RM, Anderson WF: The ADA human gene therapy clinical protocol. Human Gene Ther 1:327-362, 1990. Blaese RM, Anderson WF, Culver K: The ADA human gene therapy clinical protocol. Human Gene Ther 4:521-527, 1990. Rosenberg SA, Kasid A, Anderson WF, et al: TNF/TIL human gene therapy clinical protocol. Human Gene Ther 1:441-480, 1990. Rosenberg SA, Anderson WF, Blaese MR, et al: Immunization of cancer patients using autologous cancer cells modified by insertion of the gene for interleukin-2. Human Gene Ther 3:75- 91, 1992. Cassileth PA, Podack E, Sridhar K, et al: Phase I study of transfected cancer cells expressing the interleukin-2 gene product in limited stage small cell lung cancer. Human Gene Ther 6:369- 383, 1995. Das Gupta TK, Cohen EP and Richards JM: Phase I evaluation of interleukin-2-transfected irradiated allogeneic melanoma for the treatment of metastatic melanoma, Human Gene Ther 8:1713-1726, 1997. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, June 9-10, 1994. Human Gene Ther 6:249-252, 1995. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, June 9-10, 1994. Human Gene Ther 6:255, 1995. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, September 12-13, 1994. Human Gene Ther 6:498-499, 1995. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, March 6-7, 1995. Human Gene Ther 6:1634-1636, 1995. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, June 8-9, 1995. Human Gene Ther 7:432-434, 1996. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, June 8-9, 1995. Human Gene Ther 7:432, 1996. Rosenberg SA, Anderson WF, Asher AL, et al: Immunization of cancer patients using autologous cancer cells modified by insertion of the gene for tumor necrosis factor. Human Gene Ther 3:57-73, 1992. Berns AJM, Clift S, Cohen LK, et al: Phase I study of non-replicating autologous tumor cell injections using cells prepared with or without GM-CSF gene transduction in patients with metastaic renal cell carcinoma. Human Gene Ther 6:347-368, 1995. Seigler HF, Darrow TL, Abdel-Wahab Z, et al: A phase I trial of human gamma interferon transduced autologous tumor cells in patients with disseminated malignant melanoma. Human Gene Ther 5:761-773, 1994. Dranoff G, Soiffer R, Lynch T, et al: A phase I study of vaccination with autologous, irradiated melanoma cells engineered to secrete human granulocyte-macrophage colony stimulating factor. Human Gene Ther 8:111-123, 1997. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, March 6-7, 1995. Human Gene Ther 6:1631-1634, 1995. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, September 11-12. Human Gene Ther 7:1033, 1996. Lotze MT, Rubin JT, Carty S, et al: Gene therapy of cancer: a pilot study of IL-4-gene-modified fibroblasts admixed with autologous tumor to elicit an immune response. Human Gene Ther 5:41-55, 1994. Sobol RE, Royston I, Fakhrai H, et al: Injection of colon carcinoma patients with autologous irradiated tumor cells and fibroblasts genetically modified to secrete interleukin-2, IL-2): a phase I study. Human Gene Ther 6:195-204, 1995. Tahara H, Lotze MT, Robbins PD, et al: IL-12 gene therapy using direct injection of tumors with genetically engineered autologous fibroblasts. Human Gene Ther 6:1607-1624, 1995. Deisseroth AB, Kavanagh J, Champlin R: Use of safety-modified retroviruses to introduce chemotherapy resistance sequences into normal hematopoietic cells for chemoprotection during the therapy of ovarian cancer: a pilot trial. Human Gene Ther 5:1507-1522, 1994. Hesdorffer C, Antman K, Bank A, et al: Human MDR gene transfer in patients with advanced cancer. Human Gene Ther 5:1151-1160, 1994. OShaughnessy JA, Cowan KH, Nienhuis AW, et al: Retroviral mediated transfer of the human multidrug resistance gene, MDR-1, into hematopoietic stem cells during autologous transplantation after intensive chemotherapy for metastatic breast cancer. Human Gene Ther. 5:891-911, 1994. Deisseroth AB, Holmes F, Hortobagyi G, et al: Use of safetymodified retroviruses to introduce chemotherapy resistance sequences into normal hematopoietic cells for chemoprotection during the therapy of breast cancer; a pilot trial. Human Gene Ther 7:401-416, 1996. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, June 7-8, 1993. Human Gene Ther 5:537- 539, 1994. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, September 11-12, 1995. Human Gene Ther 5:1036- 1038, 1996. Holt JA, Arteaga CB, Robertson D, et al: Gene therapy for the treatment of metastatic breast cancer by in vivo transduction with breast-targeted retroviral vector expressing antisense c-fos RNA. Human Gene Ther 7:1369-1380, 1996. Roth JA: Modification of tumor supressor gene expression in non-small cell lung cancer, NSCLC, with a retroviral vector expressing wildtype, normal, p53. Human Gene Ther 7:861- 874, 1996. Roth JA: Modification of tumor supressor gene expression and induction of apoptosis in non-small cell lung cancer, NSCLC, with an adenovirus vector expressing wildtype p53 and cisplatin. Human Gene Ther 7:1013-1030, 1996. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, June 12-13, 1997. Human Gene Ther 9:394-402, 1998. Culver KW, van Gilder J, Link CJ, et al: Gene therapy for the treatment of malignant brain tumors with in vivo tumor transduction with the Herpes Simplex thymidine kinase gene/ganciclovir system. Human Gene Ther 5:343-379, 1994. Raffel C, Culver KW, Kohn D, et al: Gene therapy for the treatment of recurrent pediatric malignant astrocytomas with in vivo tumor transduction with the Herpes Simplex thymidine kinase gene/ganciclovir system. Human Gene Ther 5:863-890, 1994. Galpin JE, Casciato DA, Richards SB: A phase I clinical trial to evaluate the safety and biological activity of HIV-IT(TAF)(HIV- 1IIIBenv-transduced, autologous fibroblasts, in asymptomatic HIV-1 infected subjects. Human Gene Ther 5:997-1017, 1994. Nabel GJ, Fox BA, Post L, et al: A molecular genetic intervention for AIDS – effects of a transdominant negative form of Rev. Human Gene Ther 5:79-92, 1994. Haubrich R, McCutchan JA: An open label, phase I/II clinical trial to evaluate the safety and biological activity of HIVIT( V)(HIV-1IIIB env/env retroviral vector, in HIV-1-infected subjects. Human Gene Ther 6:941-959, 1995. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, September 9-10, 1993. Human Gene Ther 5:655-657, 1994. Morgan R and Walker, R: Gene therapy for AIDS using retroviral mediated gene transfer to deliver HIV-1 antisense TAR and transdominant Rev protein genes to syngeneic lymphocytes in HIV-1 infected identical twins. Human Gene Ther 7:1281-1306, 1996. Crystal RG, Jaffe A, Brody S, et al: A phase I study, in cystic fibrosis patients, of the safety, toxicity, and biological efficacy of a single administration of a replication deficient, recombinant adenovirus carrying the cDNA of the normal cystic fibrosis transmembrane conductance regulator gene in the lung. Human Gene Ther 6:643-666, 1995. Wilson JM, Engelhardt JF, Grossman M, et al: Gene therapy of cystic fibrosis lung disease using E1 deleted adenoviruses: a phase I trial. Human Gene Ther 5:501-519, 1994. Welsh MJ, Smith AE, Zabner J, et al: Cystic fibrosis gene therapy using an adenovirus vector: in vivo safety and efficacy in nasal epithelium. Human Gene Ther 5:209-219, 1994. Wilmott RW, Whitsett JA, Trapnell B, et al: Gene therapy for cystic fibrosis utilizing a replication deficient recombinant adenovirus vector to deliver the human cystic fibrosis transmembrane conductance regulator cDNA to the airways. A phase I study. Human Gene Ther 5:1019-1057, 1994. Boucher RC, Knowles MR, Johnson LG, et al: Gene therapy for cystic fibrosis using E1-deleted adenovirus: a phase I trial in the nasal cavity, the University of North Carolina. Human Gene Ther 5:615-639, 1994. Sorcher EJ, Logan JJ, Frizzell RA, et al: Gene therapy for cystic fibrosis using cationic liposome mediated gene transfer: a phase I trial of safety and efficacy in the nasal airway. Human Gene Ther 5:1261-1279, 1994. Welsh MJ, Zabner J, Graham SM, et al: Adenovirus-mediated gene transfer for cystic fibrosis: Part A. Safety of dose and repeat administration in the nasal epithelium. Part B. Clinical efficacy in the maxillary sinus. Human Gene Ther. 6:205-218, 1995. Flotte TR, Carter B, Conrad C, et al: A phase I study of an adeno-associated virus-CFTR gene vector in adult CF patients with mild lung disease. Human Gene Ther 7:1145-1159, 1996. Crystal RG, Mastrangeli A, Sanders A, et al: Evaluation of repeat administration of a replication deficient, recombinant adenovirus containing the normal cystic fibrosis transmembrane conductance regulator cDNA to the airways of individuals with cystic fibrosis. Human Gene Ther 6:667-703, 1995. Dunbar C, Kohn D, Karlsson S, et al: Retroviral mediated transfer of the cDNA for human glucocerebrosidase into hematopoietic stem cells of patients with Gaucher disease. A phase I study. Human Gene Ther 7:231-253, 1996. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, December 2-3, 1993. Human Gene Ther 5:1178-1180, 1994. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, June 9-10, 1994. Human Gene Ther 6:246-247, 1995. Department of Health and Human Services, National Institutes of Health Recombinant DNA Advisory Committee, minutes of meeting, September 12, 1997. Human Gene Ther 9, 914, 1998. Whitley CB, McIvor RS, Aronovich EL, et al: Retroviral-mediated transfer of the Iduronate-2-sulfatase gene into lymphocytes for treatment of mild Hunter syndrome, Mucopolysaccharidosis Type II). Human Gene Ther 7:537-549, 1996. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 3 EP 15 PG 13 ER PT J AU Pillay, P Chetty, R Reddy, R AF Pillay, Preedashnie Chetty, Runjan Reddy, Roshilla TI Bcl-2 and p53 Immunoprofile in Kaposi's Sarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bcl-2; p53; Kaposi's sarcoma ID bcl-2; p53; Kaposi's sarcoma AB Seventy three cases of Kaposi's sarcoma (KS) from the 3 histological subtypes (patch, plaque and nodular) were assessed for bcl-2 and p53 protein expression. The aim was determine the level of expression of these proteins in KS and in the different subtypes. Commercially available antibodies to bcl-2 and p53 were applied after both microwave and pressure cooking antigen retrieval. Bcl-2 immunoexpression increased from the patch stage (36%) to the plaque stage (45%) to the nodular stage (70.83%). Better immunostaining for bcl-2 was obtained after pressure cooking. p53 on the other hand, was not expressed in the patch or plaque stages, but 54.16% of cases in the nodular stage were immunopositive. These results show a progression of immunoexpression of both bcl-2 and p53 from the early histological stages to the late tumor stage, implying that these proteins are upregulated late in the evolution of KS. C1 [Pillay, Preedashnie] University of Natal Medical School, Private Bag 7, Congella, 4013 Durban, South Africa. [Chetty, Runjan] University of Natal Medical School, Private Bag 7, Congella, 4013 Durban, South Africa. [Reddy, Roshilla] University of Natal Medical School, Private Bag 7, Congella, 4013 Durban, South Africa. RP Pillay, P (reprint author), University of Natal Medical School, 4013 Durban, South Africa. EM pillayp11@med.und.ac.za CR Bellamy COC: p53 and apoptosis. Brit Med Bull 53:522, 1996. Brown R: The bcl-2 family of proteins. Brit Med Bull 53:466, 1996. Dei Tos AP, Doglioni C, Laurino L, et al: p53 protein expression in non-neoplastic lesions and benign and malignant lesions of soft tissue. Histopathology 22:45-50 1993. Li JL, Huang Yau-Q, Cockerell CJ, et al: Expression and mutation of the tumor suppressor gene p53 in AIDS-associated Kaposi’s sarcoma. Am J Dermatopathol 19:373, 1997. Dada MA, Chetty R, Biddolph SC, et al: The immunoexpression of bcl-2 and p53 in Kaposi’s sarcoma. Histopathology 29:159- 163, 1996. Morris CB, Gendelman R, Marrogi AJ, et al: Immunohistochemical detection of bcl-2 in AIDS-associated and classical Kaposi’s sarcoma. Am J Pathol 148:1055-1063 1996. Chor PJ, Cruz DJ: Kaposi’s sarcoma – A clinicopathologic review and differential diagnosis. J Cutan Pathol 19:6-20, 1992. Bergman R, Ramon M, Kilim S, et al: An immunohistochemical study of p53 protein in classical Kaposi’s sarcoma. Am J Dermatopathol 18:367-370 1996. Noel JC , Simonart TF: p53 protein and Kaposi’s sarcoma, letter). Am J Dermatopathol 20:322, 1998. Noel JC, De Thier F, Simonart TF: p53 overexpression is a common but late event in the pathogenesis in Kaposi’s sarcoma. Arch Dermatol Res 289:660-661, 1997. Schulz TF, Chang Y, Moore PS: Kaposi’s Sarcoma-Associated Herpesvirus, Human Herpesvirus 8). In: Human Tumor Viruses, Ed McCance DJ), American Society for Microbiology, Washington DC, pp. 87-134, 1998. Cheng EH, Nicholas J, Bellows DS, et al: A BCL-2 homolog encoded by Kaposi’s sarcoma-associated virus, human herpesvirus 8, inhibits apoptosis but does not heterodimerise with Bax or Bak. Proc Natl Acad Sci. 94:690-694, 1997. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 17 EP 20 PG 4 ER PT J AU Rathore, A Kamarajan, P Mathur, M Sinha, S Sarkar, Ch AF Rathore, Annapurna Kamarajan, Pacchapam Mathur, Meera Sinha, Subrata Sarkar, Chitra TI Simultaneous Alterations of Retinoblastoma and p53 Protein Expression in Astrocytic Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE astrocytic tumors; p53; pRb; immunochemistry; survival ID astrocytic tumors; p53; pRb; immunochemistry; survival AB The genetic alterations frequently involved in glial malignancies are in the tumor suppressor genes, Rb and p53. An altered Rb expression or p53 overexpressions is thought to indicate defective tumor supression and subsequently more aggressive tumors. Therefore, to assess the alterations in the conjoint expression of Rb and p53 proteins in formalin fixed paraffin embedded sections, 64 astrolytic tumors were studied (16 astrocytomas, 7 gemistocytic astrocytomas, 19 anaplastic astrocytomas and 22 glioblastomas) using the avidin biotin immunoperoxidase technique. Fifty two cases (81.25%) were found to be positive for p53 protein. Seventeen of these showed aberrant heterogenous staining for pRb, of which 7 were glioblastomas. Only one case of astrocytoma showed aberrant expression of both p53 and Rb. Thus, of the 64 tumors, simultaneous aberrant expression of both p53 and Rb was seen in 21.9% of cases. This was more commonly observed among glioblastoma cases (7/22). No statistical difference was found between the survival rate of heterogenous pRb and p53 positivity in different grades of tumors. In glioblastomas, the survival rate appeared to be less in patients expressing heterogenous pRb, but this was not statistically significant. These results lead us to suspect that p53 and pRb pathways are inactivated, either throught mutation or as part of the neoplastic process in astrocytic tumors. C1 [Rathore, Annapurna] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Kamarajan, Pacchapam] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Mathur, Meera] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Sinha, Subrata] All India Institute of Medical Sciences, Department of BiochemistryNew Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. RP Sarkar, Ch (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM Sarkarch@medinst.ernet.in CR Aka K, Barner JM, Bondy ML, et al: Detection of p53 alterations in human astrocytomas using frozen tissue sections for the polymerase chain reactions. J Neurooncol 16:125-133, 1993. Bourne JA: Handbook of immunoperoxidase staining methods. Dako corporation, USA, 1983, p17. Buckovich K, Duffy LA, Harlow D: The retinoblastoma protein is phosphorylated during specific phases of the cell cycle. Cell 58:1097-1105, 1989. Cavenee WK, Dryja TP, Philips RA, et al: Expression of recessive allele by chromosomal mechanisms in retinoblastoma. Nature 305:779-784, 1983. Collins VP, James CD: Gene and chromosomal alterations associated with the development of human gliomas. FASEB J 7:926- 930, 1993. Cordon-Cardo C: Mutation of cell regulators. Biological and clinical implications for human neoplasia Am J Pathol 147:545- 560, 1995. Cordon-Cardo C, Wartinger D. Petrylak D, et al: Altered expression of the retinoblastoma gene product: Prognostic indicator in bladder cancer. J Natl Cancer Inst 84:1251-1256, 1992. Ellison DW, Gatter KC, Steart PV, et al: Expression of the p53 protein in a spectrum of astrocytic tumors. J Pathol 168:383- 386, 1992. Fults D, Brockmeyer D, Tullous MW, et al: p53 mutation and loss of heterozygosity on chromosome 17 and 10 during human astrocytoma progression. Cancer Res 52:674-679, 1992. Haapasalo H, Isola J, Sa P, et al: Aberrant p53 expression in astrocytic neoplasma of the brain. Association with proliferation. Am J Pathol 142:1347-1351, 1993. Hamel W, Westphal M, Shepard HM: Loss in expression of the retinoblastoma gene product in human gliomas is associated with advanced disease. J Neuro oncology 16:159-165, 1993. Hansen MF, Koufos A, Gallie BL, et al: Osteosarcoma and retinoblastoma: a shared chromosomal mechanism revealing recessive predisposition. Proc Natl Acad Sci USA 82:6216-6220, 1985. He J, Jeffrey JO, James CD: Lack of p16 INK4 or Retinoblastoma protein, pRb, or amplification associated over expression of cdk4 is observed in distinct subsets of malignant glial tumors and cell lines. Cancer Res 55:4833-4836, 1995. Henson JW, Schnitker BL, Lorrea KM, et al: The retinoblastoma gene is involved in malignant progression of Astrocytomas. Ann Neurol 36:714-721, 1994. Hesketh R: The oncogene handbook. Academic Press, California, 1994, p526. Ichimura K, Schmidt EE, Loike HM, et al: Human glioblastomas with an alteration of the CDKN2A, p16 INK4A, MTS1, and CDK4 gene have frequent mutations of the retinoblastoma gene. Oncogene 13:1065-1072, 1996. Ishikawa J, Xu H.J, Hu S-X, et al: Inactivation of the retinoblastoma gene in human bladder and renal cell carcinomas. Cancer Res 51:5736-5743, 1991. Iuzzolino P, Ghimenton C, Nicolato A, et al: p53 protein in lowgrade astrocytomas: a study with long-term follow-up. Br J Cancer 69:586-591, 1994. Jahnson S, Risberg B, Korlsson MG, et al: p53 and Rb immunostaining is locally advanced bladder cancer: Relation to prognostic variables and predictive value for the local response to radical radiotherapy. Eur Uro 28:135-142, 1995. Karpeh MS, Brennass MF, Canee WG, et al: Altered patterns of retinoblastoma gene product expression in adult soft tissue sarcomas. Br J Cancer 72:986-991, 1995. Kleihues P, Burger PC, Scheithauer BW: Histological typing of tumors of the central nervous system. International classification of tumors No. 21, 2nd edn. World Health Organization, Geneva, 1993. Lee WH, Shew JY, Hong FD: The retinoblastoma susceptibility gene encodes a nuclear phosphoprotein associated with DNA binding activity. Nature 329:642-645, 1987. Lee SH, Kim JH, Rhee CH, et al: Loss of heterozygosity on chromosome 10, 13q, Rb), 17p and p53 gene mutations in human brain gliomas. J Korean Med Sci 10:442-448, 1995. Levine AJ: The tumor suppressor genes. Ann. Rev. Biochem 62:623-651, 1993. Louis DN, von Deimling A, Chung RY: Comparative study of p53 gene and protein alterations in human astrocytomas. J Neuropathol Exp Neurol 52:31-38, 1993. Mihara K, Cao X-R, Yen A, et al: Cell cycle-dependent regulation of phosphorylation of the human retinoblastoma gene product. Science 246:1300-1303, 1982. Nakamura M, Konishi N, Tsunoda S, et al: Immunohistochemical evaluation of pRb, p16, INK4 and cyclin D1 as a prognostic factor in astrocytic tumors, Abst, J Neurooncol 30:111, 1996. Rasheed ABK, Mchendon RE, Herndon JE, et al: Alterations of the TP53 gene in human gliomas. Cancer Res 54:1324-1330, 1994. Trudel M, Mulligass L, Cavenee WK, et al: Retinoblastoma and p53 gene product expression in breast carcinoma: Immunohistochemical analysis and clinicopathologic correlation. Human Pathol 23:388-1394, 1992. Tsuzuki T, Tsunoda S, Sakaki T, et al: Alterations of retinoblastoma, p53, p16, CDKN2), and p15 genes in human astrocytomas. Cancer 78:287-293, 1996. Watanabe K, Tachibana O, Yonekawa Y, et al: Role of gemistocytes in astrocytoma progression. Lab Invest 76:277-284, 1997. Whyte P, Buchkovich KJ, Horowitz JM, et al: Association between an oncogene and an anti-oncogene: the adenovirus EIA proteins bind to the retinoblastoma gene product. Nature 334:124-129, 1988. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 21 EP 27 PG 7 ER PT J AU Vosianov, FA Romanenko, MA Zabarko, BL Szende, B Wang, YCh Landas, S Haas, PG AF Vosianov, F Alexander Romanenko, M Alina Zabarko, B Larisa Szende, Bela Wang, Y Ching Landas, Steven Haas, P Gabriel TI Prostatic Intraepithelial Neoplasia and Apoptosis in Benign Prostatic Hyperplasia Before and After the Chernobyl Accident in Ukraine SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prostate; PIN; BPH; apoptosis; ionizing radiation ID prostate; PIN; BPH; apoptosis; ionizing radiation AB The prevalence of prostatic intraepithelial neoplasia (PIN) in men who underwent surgery for benign prostatic hyperplasia (BPH) before and after the Chernobyl nuclear accident was studied. BPH samples were obtained by adenomectomy from 45 patients operated in 1984 before the accident (Group I), and 47 patients from the low contamined Kiev City (Group II) and 76 from high contaminated area (Group III) operated between 1996 and 1998. Their BPH samples were examined histologically and immunohistochemically. The incidences of prostatic intraepithelial neoplasia (PIN) and high grade PIN (HGPIN) were 15.5 and 11.1% in Group I, 29.8 and 14.9% in Group II, and 35.5 and 19.7% in Group III. The difference between the incidences of PIN in Group I and III is signficant (p<0.02). There was increased apoptosis in areas of PIN in Group II and III as compared to Group I (p<0.001). Since apoptosis has been shown to be associated with ionizing radiation and it is now found to be associated with PIN in patients diagnosed after the Chernobyl nuclear accident, this suggests that lon-term low dose internal ionizing radiation potentially may cause prostate cancer. C1 [Vosianov, F Alexander] Academy of Medical Sciences of Ukraine, Departments of Pathology and Urology, Institute of Urology and NephrologyKiev, Ukraine. [Romanenko, M Alina] Academy of Medical Sciences of Ukraine, Departments of Pathology and Urology, Institute of Urology and NephrologyKiev, Ukraine. [Zabarko, B Larisa] Academy of Medical Sciences of Ukraine, Departments of Pathology and Urology, Institute of Urology and NephrologyKiev, Ukraine. [Szende, Bela] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H1085 Budapest, Hungary. [Wang, Y Ching] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H1085 Budapest, Hungary. [Landas, Steven] State University of New York Health Science Center, Departments of Urology and PathologySyracuse, USA. [Haas, P Gabriel] State University of New York Health Science Center, Departments of Urology and PathologySyracuse, USA. RP Szende, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H1085 Budapest, Hungary. EM bszende@korb1.sote.hu CR Rundo J: A survey of the metabolism of Cesium in man. Brit J Radiol 37:214, 1964. Nikiforov Y, Gnepp DR: Pediatric thyroid cancer after the Chernobyl disaster. Pathomorphologic study of 84 cases, 1991- 1992, from the Republic of Belarus. Cancer 74:748-766, 1994. Mettler FA, Williamson MR, Royal HD: Thyroid nodules in the population living around Chernobyl. JAMA 268:616-619, 1992. Williams ED: Chernobyl, eight years on. Nature 371:556, 1994. Williams ED: Fallout from Chernobyl. Thyroid cancer in children increased dramatically in Belarus. Brit Med J 309:1298- 1300, 1994. Goldman M: The Russian radiation legacy: its integrated impact and lessons. Environ Health Perspectives 105:1385, 1997. Vozianov AF, Romanenko AM, Wester K, et al: Long-term low doses irradiation and urothelium in patients with chronic cystitis. J Acad Med Sci Ukraine 2:421-436, 1996. Romanenko AM, Vozianov AF, Wester K, et al: Immunohistochemical changes in bladder urothelium in patients from radiocontaminated areas in Ukraine. Pathol Res Practice 191:763, 1995. Abbas F, Scardino PT: The natural history of Clinical Prostate Carcinoma. Cancer 80:827-833, 1997. Bostwick DG: High Grade Prostatic Intraepithelial Neoplasia: The most likely precursor of prostate cancer. Cancer 75:1823- 1836, 1995. Gaudin PB, Sesterhenn IA, Wojno KJ, et al: Incidence and clinical significance of high-grade prostatic intraepithelial neoplasia in TURP specimens. Urology 49:558-563, 1997. Pacelli A, Bostwick DG: Clinical significance of high-grade prostatic intraepithelial neoplasia in transurethral resection specimens. Urology 50:355-359, 1997. Steel RGD, Torrie, JH: Principles and Procedures of Statistics. New York: McGraw-Hill, 1960. Sakr WA, Grignon DJ, Haas GP, et al: Age and racial distribution of prostatatic intraepithelial neoplasia. Eur Urol 30:138- 144, 1996. Myers RB, Grizzle WE: Changes in biomarker expression in the development of prostatic adenocarcinoma. Biotech Histochem 72:86-95, 1997. Humphrey PA, Swanson PE: Immunoreactive p53 protein in high-grade prostatic intraepithelial neoplasia. Pathol Res Practice 191:881-887, 1995. Maity A, Kao GD, Muschel RJ, et al: Potential molecular targets for manipulating the radiation response. Int J Radiation Oncol Biol Phys 37:639-653, 1997. Stewart BW: Mechanisms of Apoptosis: Integration of Genetic, Biochemical and Cellular Indicators. J Natl Cancer Inst 86:1286-1292, 1994. Ron E, Preston DL, Mabuchi K, et al: Cancer incidence in atomic bomb survivors. Part IV: comparison of cancer incidence and mortality. Radiation Res 137:s98-112, 1994. Raes F, De Cort M, Graziani G: Multi-fractal nature of radioactivity deposition on soil after the Chernobyl accident. Health Phys, 61:271-274, 1991. Shizuma K, Iwatani K, Hasai H, et al: Fallout in the hypocenter area of the Hiroshima atomic bomb. Health Phys 57:1013-1016, 1989. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 28 EP 31 PG 4 ER PT J AU Magyarosy, E Martin, WJ Chu, WE Martin, ES AF Magyarosy, Edina Martin, W John Chu, W Elizabeth Martin, E Sue TI Differential Diagnostic Significance of The Paucity of HLA-I Antigens on Metastatic Breast Carcinoma Cells in Effusions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HLA-A; B; C antigens; breast cancer; metastasis; cytology ID HLA-A; B; C antigens; breast cancer; metastasis; cytology AB Distinction between benign reactive mesothelial cells and metastatic breast adenocarcinoma cells in effusions from patients with a known prior history of breast cancer is not the easiest task in diagnostic pathology. Here, we report the usefulness of testing the expression of class I HLA antigens (HLA A, B, C) in this respect. Cytospins were prepared from effusions of patients without the history of breast cancer (5 cases) and from effusions of patients with infiltrating ductal carcinoma (11 cases). Three effusions from cancerous patients were not malignant cytologically. The expression of HLA-A, B, C, HLA-DR and ß2-microglobulin as well as the macrophage antigen, CD14, was evaluated by immunocytochemistry. In 10 of 11 effusions the cytologically malignant cells expressed very weak or undetectable HLA-A,B,C as compared to the mesothelial cells and macrophages. The paucity of expression of HLA-A, B, C was detectable in those 3 cases where a definitive cytological diagnosis of malignancy could not be established. In contrast, mesothelial cells and macrophages from all samples were uniformly and strongly positive for both HLA-A, B, C and ß2-microglobulin. We conclude that the paucity of HLA-I antigens provides a marker helpful in distinguishing metastatic breast carcinoma cells from reactive mesothelial cells in effusions. C1 [Magyarosy, Edina] National Cancer Institute of Health, Department of PathologyBethesda, USA. [Martin, W John] University of Health Sciences, Department of PathologyBethesda, USA. [Chu, W Elizabeth] National Cancer Institute of Health, Department of PathologyBethesda, USA. [Martin, E Sue] National Cancer Institute of Health, Department of PathologyBethesda, USA. RP Magyarosy, E (reprint author), National Cancer Institute of Health, Department of Pathology, Bethesda, USA. CR Corsdon JM, Pinkus GS: Mesothelioma: profile of keratin proteins and carcinoembryonic antigen. Am J Pathol 108:80-87, 1982. Said JW, Nash G, Banks-Schlegel S, et al: Keratin in human lung tumors: patterns of lokalization of different molecular-weight keratin proteins. Am J Pathol 113:27-32, 1983. To A, Coleman DV, Dearnaley DP, et al: Use of antisera to epithelial membrane antigen for the cytodiagnosis of malignancy in effusions. J Clin Pathol 34:1326-1332, 1981. Walts AE, Said JW: Specific tumor markers in diagnostic cytology: immunoperoxidase studies of carcinoembryonic antigen, lysozyme and other tissue antigens in effusions, washes and aspirates. Acta Cytol 33:356-360, 1980. Concha A, Esteban F, Cabrera T, et al: Tumor aggressiveness and MHC class I and II antigens in laryngeal and breast cancer. Semin Cancer Biol 2:47-54, 1991. Kaklamanis L, Leek R, Koukourakis M, et al: Loss of transporter in antigen processing 1 transport protein and major histocompatibility complex class I molecules in metastatic versus primary breast cancer. Cancer Res 55:5191-5194, 1995. Vitale M, Rezzani R, Rodella L, et al: HLA class I antigen and transporter associated with antigen processing downregulation in high-grade primary breast carcinoma lesions. Cancer Res 58:737-742, 1998. Joseph MG, Banerjee D, Harris P, et al: Multiparameter flow cytometric DNA analysis of effusions: a prospective study of 36 cases compared with routine cytology and immunocytochemitry. Mod Pathol 8:686-693, 1995. Chen LM, Lazcano O, Katzman JA, et al: The role of conventional cytology, immunocytochemistry and flow cytometric DNA ploidy in the evaluation of body cavity fluids: a prospective study of 52 patients. Am J Clin Pathol 109:712-721, 1998. 9. Fleming KA, McMichael A, Morton JA, et al: Distribution of HLA class I antigens in normal human tissue and in mammary cancer. J Clin Pathol 34:779-784, 1981. Travers PJ, Arble J, Trowsdale J, et al: Lack of expression of HLA-A,b,c antigens in choriocarcinoma and other human cell lines. Natl Cancer Inst Monogr 60:175-185, 1981. Lampson LA, Fisher CA, Whelan JP: Striking paucity of HLAA, B,C and b2 microglobulin on human neuroblastoma lines. J Immunol 130:2471-2478, 1983. Amiot L, Onno M, Lamy T, et al: Loss of HLA molecules in B lymphomas is associated with an aggressive clinical course. Br J Haematol 100:655-663, 1998. Glasova M, Konikova E, Stasakova J, et al: The relationship of HLA-DR, CD38 and CD71 markers to activation, proliferation and differentiation of some human leukemia and lymphoma cells. Neoplasma 45:88-95, 1998. Klein T, Levin I, Niska A, et al: Correlation between tumour and serum beta2m expression in patients with breast cancer. Eur J Immunogenet 23:417-423, 1996. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 32 EP 35 PG 4 ER PT J AU Dogusoy, G Karayel, AF Gocener, S Goksel, S AF Dogusoy, Gulen Karayel, Anik Ferah Gocener, Selda Goksel, Suha TI Histopathologic Features and Expression of Bcl-2 and p53 Protein in Primary Gastric Lymphomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lymphoma; MALT; stomach; histopathology; bcl-2; p53 ID lymphoma; MALT; stomach; histopathology; bcl-2; p53 AB The aim of this study is to present a histopathologic and immunohistochemical analysis of primary gastric lymphomas which were reclassified according to the concept of mucosa associated lymphoid tissue (MALT). The resected specimens from 41 patients with primary gastric lymphoma were investigated retrospectively. Immunohistochemical study was done to analyze the immunophenotype and bcl-2 and p53 proteins expression. Twenty three of the cases had tumors mainly located in the antrum. Histologically, 12 were low grade and 20 were high grade B-cell lymphoma of MALT, 9 other B-cell nonHodgkin’s lymphomas. Helicobacter pylori was identified in 72% of the cases. According to Musshoff’s modification, most of the MALT lymphoma cases had stage I or II disease. There was significant difference between low and high grade cases, in respect to depth of invasion in gastric wall. Immunohistochemically, the neoplastic cells in all MALT lymphomas, expressed B-cell phenotype. bcl-2 protein was found to be expressed in 59% and p53 protein expression was detected in 72% of cases. Among the B-cell lymphoma of MALT, bcl-2 positivity decreased and p53 positivity increased significantly as the histological grade advanced. So, an inverse correlation was observed between the expression of bcl-2 and p53. In conclusion, most primary gastric lymphomas are low or high grade B-cell MALT lymphomas and appear to arise in MALT acquired as a reaction to Helicobacter pylori infection. Expression of bcl-2 and p53 in gastric lymphomas may be associated with transformation from low-grade to high-grade disease. C1 [Dogusoy, Gulen] Istanbul Medical Faculty, Department of Pathology, Cerrahpasa Tip Fakultesi, Patoloji ABD, 34303 Istanbul, Aksar, Turkey. [Karayel, Anik Ferah] Istanbul Medical Faculty, Department of Pathology, Cerrahpasa Tip Fakultesi, Patoloji ABD, 34303 Istanbul, Aksar, Turkey. [Gocener, Selda] Istanbul Medical Faculty, Department of Pathology, Cerrahpasa Tip Fakultesi, Patoloji ABD, 34303 Istanbul, Aksar, Turkey. [Goksel, Suha] Istanbul Medical Faculty, Department of Pathology, Cerrahpasa Tip Fakultesi, Patoloji ABD, 34303 Istanbul, Aksar, Turkey. RP Dogusoy, G (reprint author), Istanbul Medical Faculty, Department of Pathology, 34303 Istanbul, Turkey. CR Chan JKC, Isaacson P: Relationship between high-grade lymphoma and low-grade B-cell mucosa-associated lymphoid tissue lymphoma, MALToma, of the stomach. Am J Pathol 136:1153-1164, 1990. Du BM, Peng H, Singh N, et al: The accumulation of p53 abnormalities is associated with progression of mucosa-associated lymphoid tissue lymphoma. Blood 86:4587-4593, 1995. Genta RM, Hamner W, Graham DY: Gastric lymphoid follicles in Helicobacter pylori infection. Hum Pathol 24:577-583, 1993. Harris NL, Jaffe ES, Stein H, et al: A revised European-American classification of lymphoid neoplasms: A proposal from the international lymphoma study group. Blood 84:1361-1392, 1994. Hsi ED, Eisbruch A, Greenson JK, et al: Classification of primary gastric lymphomas according to histologic features. Am J Surg Pathol 22:17-27, 1998. Isaacson P, Wright D: Malignant lymphoma of mucosa-asociated lymphoid tissue. A distinctive type of B-cell lymphoma. Cancer 52:1410-1416, 1983. Isaacson PG, Spencer J, Finn T: Primary B-cell gastric lymphoma. Hum Pathol 17:72-82, 1986. Isaacson P: Gastrointestinal lymphoma. Hum Pathol 25:1020- 1029, 1994. Isaacson P: Recent developments in our understanding of gastric lymphomas. Am J Surg Pathol 20, Suppl.1): S1-S7, 1996. Laszewski MJ, Kamat D, Kemp JD, et al: Immunophenotypic and genotypic characterization of primary non-Hodgkins lymphoma of the gastrointestinal tract. Modern Pathology 3:423- 429, 1990. Lavergne A, Kanavaros P, Galian A: Primary B-cell gastric lymphomas of mucosa-associated lymphoid tissue. Histological and immunohistochemical study of ten cases on surgical specimens. Histol Histopath 7:129-136, 1992. Mielke B, Moller P: Histomorphologic and immunophenotypic spectrum of primary gastrointestinal B-cell lymphomas. Int J Cancer 47:334-343, 1991. Nakamura S, Akazawa K, Yao T et al: Primary gastric lymphoma. A clinicopathologic study of 233 cases with special reference to evaluation with the MIB-1 index. Cancer 76:1313- 1324, 1995. Nakamura S, Akazawa K, Kinukawa N, et al: M: Inverse correlation between the expression of bcl-2 and p53 proteins in primary gastric lymphoma. Hum Pathol 27:225-233,1996. Nakamura S, Yao T, Aoyagi K, et al: Helicobacter pylori and primary gastric lymphoma. A histopathologic and imunohistochemical analysis of 237 patients. Cancer 79:3-11, 1997. Narita M, Yatabe Y, Asai J, et al: Primary gastric lymphomas: Morphologic, immunohistochemical and immunogenetic analyses. Pathology International 46:623-629, 1996. Pan L, Diss TC, Cunningham D, et al: The bcl-2 gene in primary B cell lymphoma of mucosa-associated lymphoid tissue, MALT). Am J Pathol 135:7-11, 1989. Radaszkiewicz T, Dragosics B, Bauer P: Gastrointestinal malignant lymphomas of the mucosa-associated lymphoid tissue: factors relevant to prognosis. Gastroenterology 102:1628-1638, 1992. Spencer J, Diss TC, Isaacson PG: Primary B cell gastric lymphoma. A genotypic analysis. Am J Pathol 135:557-564, 1989. Wotherspoon AC, Hidalgo CO, Falzon MR: Helicobacter pylori- associated gastritis and primary B cell gastric lymphoma. Lancet 338:1175-1176, 1991. Wotherspoon AC, Doglioni C, Diss TC, et al: Regression of primary low-grade B-cell gastric lymphoma of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori. Lancet 342:575-577, 1993. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 36 EP 40 PG 5 ER PT J AU Guran, S Tali, ET AF Guran, Sefik Tali, E Turgut TI p53 and p16INKA4A Mutations During the Progression of Glomus Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE glomus tumor; p53; p16INKA4A; LOH ID glomus tumor; p53; p16INKA4A; LOH AB Glomus tumors are significantly rare tumors of carotid body. The great majority of these tumors are benign in character. Here we present two brothers with hereditary glomus jugulare tumor who had consanguineous parents. Radiotherapy was applied approximately 8 and 10 years ago for treatment in both cases. Eight years later, one of these cases came to our notice due to relapse. The mutation pattern of p53, p57KIP2, p16INK4A and p15NK4B genes which have roles in the cell cycle, was analyzed in tumor samples obtained from the two affected cases in the initial phase and from one of these cases at relapse. The DNA sample obtained from the case in initial diagnosis phase revealed no p53, p57KIP2, p16INK4A or p15INK4B mutation. He is still in remission phase. Despite the lack of p53, p57KIP2, p16INK4A and p15INK4B mutation at initial diagnosis the tumor DNA of the other case in relapse revealed p53 codon 243 (ATG->ATC; met->ile) and p16 codon 97 (GAC->AAC; asp->asn) missense point mutations. No loss of heterozygosity in p53 and p16INK4A was observed by microsatellite analysis of tumoral tissues in these cases. P53 and p16IINK4A mutations observed in relapse phase were in conserved regions of both genes. No previous reports have been published with these mutations in glomus tumor during progression. The mutation observed in this case may due to radiotherapy. In spite of this possibility, the missense point mutations in conserved region of p53 and p16INK4A genes may indicate the role of p53 and p16INK4A in tumor progression of glomus tumors. C1 [Guran, Sefik] Gulhane Medical Faculty, Department of Medical Biology and Genetics, 06018 Ankara, Etlik, Turkey. [Tali, E Turgut] Gazi Medical Faculty, Department of RadiologyAnkara, Turkey. RP Guran, S (reprint author), Gulhane Medical Faculty, Department of Medical Biology and Genetics, 06018 Ankara, Turkey. EM gurans@gata.edu.tr CR Hirschfeld A, Kornblith P: Uncommon tumors of the nervous system. In Textbook of uncommon cancer. Eds: Williams CJ, Krikorian JG, Green MR, Raghavan D. New York John Wiley and Sons 1988, pp 590-612. Baysal BE, von Schorthost FM, Farr JF, et al: A high resolution STS, FST and gene based physical map of the hereditary paraganglioma region on chromosome 11q23. Genomics 44:214- 221, 1997. Oosterwijk JC, Jansen JC, von Schothorst FM, et al: First experience with genetic counseling based on predictive DNA diagnosis in hereditary glomus tumorsparagangliomas. J Med Genet 33:379-383, 1996. Wang CC: Paraganglioma of the head and neck, In Textbook of uncommon cancer Eds: Williams CJ, Krikorian JG, Green MR, Raghavan D. New York, John Wiley and Sons. 1988, pp 725- 729. Hiruta N, Kameda N, Takudome T, et al: Malignant glomus tumor: a case report and review of the literature. Am J. Surg Pathol 21:1096-1103, 1997. Imamura J, Miyashi I, Koeffler PH: P53 in haematologic malignancies. Blood 84:2412-2421, 1994. Marx J: How p53 suppress cell growth. Science 262:1644-1645, 1993. el-Deiry DW, Harper JW, OConnor PM, et al: WAF1/ CIP 1 is induced in p 53-mediated G1 arrest and apopitosis. Cancer Res 54:1169-1174, 1994. Morgan DO: Principles of CDK regulation. Nature 374:131- 134, 1995. Nurse P: Ordering S phase and M phase in the cell cycle. Cell 79:547-550, 1994. Sherr CS: G1 phase progression: Cycling on clue. Cell 79:551- 555, 1994. Hunter T and Pines J: Cyclins and cancer II: Cyclin D and CDK inhibitors come of age. Cell 79:573-582, 1994. Guan KL, Jenkins CW, Li Y, et al: Growth suppression by p18, p16(INK4/MTS1, and p14, INK4B/MTS2)-related CDK6 inhibitor, correlates with wide-type pRb function. Genes Dev 8:2939-2952, 1994. Lee MH, Reynisdottir I, Massague J: Cloning of p57KIP2, a cyclin dependent kinase inhibitor with unique domain structure and tissue distribution. Genes Dev 9:639-649, 1995. Matsouka S, Edwards MC, Bai C, et al: p57KIP2, a structurally distinct member of the p21 CIP1 Cdk inhibitor family, is a candidate tumor suppressor gene. Genes Dev 9:650-662, 1995. Zhang Y, Xiong Y, Yarbrough WG: ARF promotes MDM2 degredation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways. Cell 92:725- 734, 1998. Pomerantz J, Schreiber-Agus N, Silverman A, et al: The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2’ s inhibition of p53. Cell 92:713-725, 1998. Lo Coco F, Gaidano G, Louie DC, et al: P53 mutations are associated with histologic transformation of follicular lymphoma. Blood 82:2289-2295, 1993. Guran ª, Bahce M, Beyan C, et al: During the progression of Chronic Myeloid Leukemia, p53, p15, p16 and p57 mutations. Haemotologia, in press). Guran ª, Pak I: Cumulation of TP53 mutations and p15p16 homozygous deletions in HPV type 16 positive scrotal cancers. Cancer Genet Cytogenet, in press). Tali ET, ªener RN, Ibiº E, et al: Familial bilateral glomus jugulare tumors, Neuroradiology 33:171-172, 1991. Sambrook J, Fritsch EF, Maniatis T: Molecular Cloning. New York. Cold Spring Harbor Lab Press 1998. Wright DK, Manos MM: Simple preparation from paraffinembedded tissues. In PCR Protocols, a guide to methods and applications. Eds. Innis MA. San Diego, Academic Press Inc. pp. 153-158 1990. Liu L, Lassam NJ, Slingerland JM, et al: Germline p16 INK4A mutation and protein dysfunction in a family with inherited melanoma. Oncogene 11:405-412, 1995. Buchman VL, Chumakov PM, Ninkina NN, et al: A variation in the structure of the protein-coding region of human p53 gene. Gene 70:245-252, 1988. Lee PM, De Baun M, Randhava G, et al: Low frequency of p57KIP2 mutation in Beckwith-Wiedemann Syndrome. Am J Hum Genet 61:304-309, 1997. Orita M, Suzuki Y, Sekiya T, et al: Rapid and sensitive detection of point mutations and DNA polymorphism using the polymerase chain reaction. Genomics 5:874-879, 1989. Piqueras JF, Santos J, Castro IP, et al: Frequent allelic losses of 9p21 markers and low insidence of mutations at p16, CDKN2, gene in non-Hodgkin’s lymphomas of B-cell lineage. Cancer Genet Cytogenet 98:63-68, 1987. Weissenbach J, Gyapay G, Dib D, et al: A second-generation linkage map of the human genome. Nature 359, 798-801, 1992. Liu Q, Neuhausen S, Mc Clure M, et al: CDKN2, MST1, tumor suppressor gene mutations in human cancer cell lines. Oncogene 10:1061-1067, 1995. Nobori T, Miura K, Wu DJ, et al: Deletions of the cyclin-dependent kinase -4 inhibitor gene in multiple human cancers. Nature 368, 753-756, 1994. Kwiatkowski DJ, Diaz MO: Dinucleotide repeat polymorphism at the IFNA locus, 9p22). Human Mol Genet 1:658, 1992. Hahn M, Serth J, Fislage R, et al: Polymerase chain reaction detection of a highly polymorphic VNTR segment in intron 1 of the human p53 gene, letter). Clin Chem 39:549-550, 1993. Mc Daniel T, Carbone D, Takahashi T, et al: The Msp I polymorphism in intron 6 of p53, TP53, detected by digestion of PCR products. Nucleic Acids Res 19:4796, 1991. Barel D, Avigad S, Mor C, et al: A novel germ-line mutation in the non-coding region of the p53 gene in a Li-Fraumeni family. Cancer Genet Cytogenet 103:1-6, 1998. Kiyosawa T, Umebetashi Y, Nakayama Y, et al: Hereditary multiple glomus tumors involving the gleans penis. A case report and review of the literature. Dermatol Surg 21:895-899, 1995. Brathwaite CD, Poppiti RS: Malignant glomus tumor. A case report of widespread metastases in a patient with multiple glomus body hamartomas. Am J Surg Pathol 20:233-238, 1996. Sorensen BS, Hovic E: CDKN2A, P16INK4A, somatic and germline mutations. Human Mutation 7:294-303, 1996. Alberts B, Bray D, Lewis J, et al: Cancer eds. Alberts B, Bray D, Lewis J, Raff M, Roberts K, Watson JD in Molecular biology of the cell. New York. Garland Publishing. 1994 pp. 1255- 1291. Gabriel FM, Sampson JH, Dodd IG, et al: Glomus jugulare tumor metastatic to the sacrum after high-dose radiation therapy: a case report. Neurosurgery 37:1001-1005, 1995. Levine AS, Momand S, Finlay AC: The p53 tumor suppressor gene. Nature 351:453-458, 1991. Lilischikis R, Sorcevic R, Kennedy C, et al: Cancer-associated missense and deletion mutations impair p16 INK4 CDK inhibitory activity. Int J Cancer 66:294-304, 1996. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 41 EP 45 PG 5 ER PT J AU Karthikeyan, K Masilamani, V Govindasamy, S AF Karthikeyan, K. Krishnamoorthy Masilamani, Vadivel Govindasamy, Swaminathan TI Spectofluorimetric Detection of DMBA-Induced Mouse Skin Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mouse skin carcinoma; plasma; erythrocytes; erythrocyte membrane; fluorescent intensity; fluorophors ID mouse skin carcinoma; plasma; erythrocytes; erythrocyte membrane; fluorescent intensity; fluorophors AB An attempt has been made to evaluate the normal and cancer blood samples of 7,12-dimethylbenz(a)anthracene (DMBA)-induced mouse skin carcinoma by spectrofluorimetric method. Analysis of acetone extracts of plasma, erythrocyte and erythrocyte membrane showed an alteration around 630 nm when excited at 400 nm by cancer samples, compared to normal samples. The ratio of fluorescent intensity at 530 nm/630 nm was found to be decreased in erythrocyte and plasma and increased in erythrocyte membrane. These changes are not detectable in both hemolysates. It has been suggested that erythrocyte may be the carriers of fluorophors that accumulate in the cancer tissue and may be useful in the diagnosis and treatment of malignancies. C1 [Karthikeyan, K. Krishnamoorthy] University of Madras, Department of Biochemistry and Molecular Biology, 600 025 Madras, India. [Masilamani, Vadivel] Anna University, Department of Physics, Centre for Laser TechnologyMadras, India. [Govindasamy, Swaminathan] University of Madras, Department of Biochemistry and Molecular Biology, 600 025 Madras, India. RP Govindasamy, S (reprint author), University of Madras, Department of Biochemistry and Molecular Biology, 600 025 Madras, India. EM sgsamy@unimad.ernet.in CR Balasubramanian S, Elangovan V, Govindasamy S: Fluorescence spectroscopic identification of 7,12-dimethylbenz(a)anthracene induced hamster buccal pouch carcinogenesis. Carcinogenesis 16:2461-2465, 1995. Fox SB, Gatter KC, Harris AL: Tumor angiogenesis. J Pathol 179:232-237, 1996. Holstein CSV, Nilsson AMK, Andersson-Engel S, et al: Detection of adenocarcinoma in Barett’s esophagus by means of laser induced fluorescence. Gut 39:711-716, 1996. Ingrams DR, Dhingra JK, Roy K, et al: Autofluorescence characteristics of oral mucosa. Head and Neck 19:27-32, 1997. Karthikeyan K, Ravichandran P, Govindasamy S: Chemopreventive effect of Ocimum sanctum on DMBA-induced hamster buccal pouch carcinogenesis. Oral Oncol 35:112-119, 1999. Vengadesan N, Aruna P, Ganesan S: Characterization of native fluorescence from DMBA-treated hamster cheek pouch buccal mucosa for measuring tissue transformation. Br J Cancer 77:391-395, 1998. Wenghong L: Some fluorescence observation on the canceration tissue and blood of cancer patients. SPIE 1054:196-199, 1989. Xu X, Meng JW, Hou S: The characteristic fluorescence of the serum of cancer patients. J Lumin 40-41, 219-220, 1988. Yang, YL, et al: Characteristic autofluorescence for cancer diagnosis and its origin. Lasers Surg Med 7:528-532, 1987. Zonios G: Spectral pathology. Ann NY Acad Sci 838:108-115, 1998. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 46 EP 48 PG 3 ER PT J AU Onoda, MJ Kantak, SS Diglio, AC AF Onoda, M James Kantak, S Seema Diglio, A Clement TI Radiation Induced Endothelial Cell Retraction in vitro: Correlation with Acute Pulmonary Edema SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE radiation; lung; edema ID radiation; lung; edema AB We determined the effects of low dose radiation (<200 cGy) on the cell-cell integrity of confluent monolayers of pulmonary microvascular endothelial cells (PMEC). We observed dose- and time-dependent reversible radiation induced injuries to PMEC monolayers characterized by retraction (loss of cell-cell contact) mediated by cytoskeletal F-actin reorganization. Radiation induced reorganization of F-actin microfilament stress fibers was observed >30 minutes post irradiation and correlated positively with loss of cell-cell integrity. Cells of irradiated monolayers recovered to form contact inhibited monolayers >24 hours post irradiation; concomitantly, the depolymerized microfilaments organized to their pre-irradiated state as microfilament stress fibers arrayed parallel to the boundaries of adjacent contact-inhibited cells. Previous studies by other investigators have measured slight but significant increases in mouse lung wet weight >1 day post thoracic or whole body radiation (>500 cGy). Little or no data is available concerning time intervals <1 day post irradiation, possibly because of the presumption that edema is mediated, at least in part, by endothelial cell death or irreversible loss of barrier permeability functions which may only arise >1 day post irradiation. However, our in vitro data suggest that loss of endothelial barrier function may occur rapidly and at low dose levels (<200 cGy). Therefore, we determined radiation effects on lung wet weight and observed significant increases in wet weight (standardized per dry weight or per mouse weight) in <5 hours post thoracic exposure to 50-200 cGy x-radiation. We suggest that a single fraction of radiation even at low dose levels used in radiotherapy, may induce pulmonary edema by a reversible loss of endothelial cell-cell integrity and permeability barrier function. C1 [Onoda, M James] Wayne State University, Department of Radiation OncologyDetroit, USA. [Kantak, S Seema] Wayne State University, Department of Radiation OncologyDetroit, USA. [Diglio, A Clement] Wayne State University, Department of Radiation OncologyDetroit, USA. RP Onoda, MJ (reprint author), Wayne State University, Department of Radiation Oncology, Detroit, USA. CR Jochelson MS, Tarbell MJ, Weinstei, HJ: Unusual thoracic radiographic findings in children treated for Hodgkin’s disease. J Clin Oncol 4:6-12, 1986. Fulkerson WJ, McLendon RE, Posnitz LR: Adult respiratory distress syndrome after limited thoracic radiotherapy. Cancer 57:1841-1846, 1986. Shankar PG, Kimler BF, Giri UP, et al: Comparison of single fractionated and hyperfractionated irradiation on the development of normal tissue damage in rat lung. Int J Rad Onc Phy 11:527-534, 1984. Ward WF, Sharplin J, Franko AJ, et al: Radiation-induced pulmonary endothelial dysfunction and hydroxyproline accumulation in four strains of mice. Rad Res 120:113-120, 1989. Penny DP, Siemann DW, Rubin P, et al: Morphological correlates of fractionated radiation of the mouse lung: early and late effects. Int J Rad Oncol Biol Phys 29:789-804, 1994. Down JD, Nicholas D, Steel GG: Lung damage after hemithoracic irradiation: Dependence on the mouse strain. Radiother Oncol 6:43-50, 1986. Vergera JA, Raymond U, Thet LA: Changes in lung morphology and cell number in radiation pneumonitis and fibrosis: A quantitative ultrastructural study. Int J Rad Oncol Biol Phy 13:723- 732, 1987. Law MP, Ahler RG: Vascular and epithelial damage in the lung of mouse after x-ray or neutrons. Radiat Res 117:128-144, 1989. Yi ES, Bedoya A, Lee H, et al: Radiation-induced lung injury in vivo expression of transforming growth factor-beta precedes fibrosis. Inflammation 20:339-352, 1996. Gross NJ, Holloway NO, Narine KR: Effects of some nonsteroidal anti-inflammatory agents on experimental radiation pneumonitis. Radiat Res 127:317-324, 1991. Fauroux B, Clement A, Tournier G: Pulmonary toxicity of drugs and thoracic irradiation in children. Rev Mal Respir 13:235-242, 1996. Green GM, Finkelstein JZ, Yefft MF, at al: Diffuse interstitial pneumonitis after pulmonary irradiation for metastatic Wilm’s tumor. Cancer 63:450-453, 1989. Van Houtte P: Radiation and chemotherapy induced lung toxicity. Int J Rad Oncol Biol Phy 13:647-649, 1987. Aubin JE, Alders E, Heersche JNM: A primary role for microfilaments, but not microtubules, in hormone-induced cytoplasmic retraction. Exp Cell Res 143:439-450, 1993. Shasby MD, Shasby SS, Sullivan JM, et al: Role of endothelial cytoskeleton in the control of endothelial permeability. Circ Res 51:657-661, 1982. Shasby MD, Lind SE, Shasby SS, et al: Reversible oxidantinduced increases in albumin transfer across cultured endothelium: Alterations in cell shape and calcium homeostasis. Blood 3:605-614, 1985. Wong WKK, Gotlieb AI: Endothelial cell monolayer integrity. I. Characterization of the dense peripheral band of microfilaments. Arteriosclerosis 6:212-221, 1986. Friedman M, Saunders S, Madden MC, et al: Effects of ionizing radiation on the pulmonary endothelial cell uptake of alpha aminoisobutyric acid and synthesis of prostacyclin. Radiat Res 106:171-181, 1986. Degowin RL, Lewis LJ, Hoak JC, et al: Radiosensitivity of human endothelial cells in culture. J Lab Clin Med 84:42-48, 1974. Hahn GL, Menconi MJ, Cahill M, et al: Influence of gamma radiation on arachidonic acid release and prostacyclin synthesis. Prostaglandins 25:783-791, 1983. Eldor A, Vlodavsky I, Hyam E, et al: Effect of radiation on prostacyclin production by cultured endothelial cells. Prostaglandins 25:263-279, 1983. Farrukh IS, Michael JR, Peters SP, et al: The role of cyclooxygenase and lipoxygenase mediators in oxidant-induced lung injury. Am Rev Respir Dis 137:1343-1349, 1988. Ward PA, Sulavik MC, Johnson KJ: Rat neutrophil activation and effects of lipoxygenase and cyclooxygenase inhibitors. Am J Pathol 116:223-233, 1984. Kantak SS, Diglio CA, Onoda JM: Low dose radiationinduced endothelial cell retraction. Int J Radiat Biol 64:319- 328, 1993. Siemann DW, Hill RP, Penny DP: Early and late pulmonary toxicity in mice evaluated 180 and 420 days following lung radiation. Radiat Res 89:386-407, 1982. Jennings FL, Arden A: Development of radiation pneumonitis. Arch Path 74:351-360, 1962. Gross NJ: Pulmonary effects of radiation therapy. Ann Int Med 86:81-92, 1977. Germon PA, Brady LW: Physiologic changes before and after radiation treatment for carcinoma of the lung. J Am Med Assoc 206:809-814, 1968. Prato FS, Kurdyak R, Saibil EA, et al: Physiologic and radiologic assessment during the development of pulmonary radiation and fibrosis. Radiology 122:398-397, 1977 Mah K, VanDyke J, Keane T, et al: Acute radiation-induced pulmonary damage:A clinical study on the response of fractionated radiation therapy. Int J Rad Onc Biol Phy 13:179-188, 1987. Fenessey FJ: Irradiation damage to the lung. J Thoracic Imag 2:68-79, 1987. Evans ML, Graham MM, Mahler PA, et al: Use of steroids to suppress vascular response to radiation. Int J Rad Onc Biol Phy 13:563-567, 1987. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 49 EP 55 PG 7 ER PT J AU Graf, M Blaeker, H Herwart, FO AF Graf, Matthias Blaeker, Hendrik Herwart, F Otto TI Extraurinal Metastasizing Ependymoma of the Spinal Cord SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE extraneural metastases; intraspinal tumor; ependymoma ID extraneural metastases; intraspinal tumor; ependymoma AB This paper reports a case of the rare entity of an extraneural metastasizing ependymoma of the spinal cord. The tumor which arose in the conus medullaris and in the cauda equina was first diagnosed in 1956 when a thoracolumbar myeloresection was performed. At autopsy, 40 years after the primary diagnosis, a massive local tumor recurrence with extraneural metastases in the lungs, the pleura, the liver, and the thoracal and abdominal lymph nodes were found. Immunohistochemical stains of the extraneural metastases showed a strongly cytoplasmatic expression of glial fibrillary acidic protein (GFAP). Neither the primary tumor nor its metastases showed any of the conventional morphological criteria of malignancy. Reviewing the literature we discuss the possible mechanism of extraneural tumor spread and the incidence of metastases with regard to the tumor type. C1 [Graf, Matthias] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. [Blaeker, Hendrik] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. [Herwart, F Otto] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. RP Graf, M (reprint author), University of Heidelberg, Department of Pathology, D-69120 Heidelberg, Germany. EM matthias_graf@ukl.uni-heidelberg.de CR Bailey P, Cushing H: A classification of the tumors of the glioma group on a histogenetic basis with a correlation study of prognosis. J. B. Lippincott, Philadelphia, 1926. Berger MS, Baumeister B, Geyer JR, et al: The risks of metastases from shunting in children with primary central nervous system tumors. J Neurosurg 74:872-877, 1991. Birchmeier W, Weidner KM, Hulsken J, et al: Molecular mechanisms leading to cell junction, cadherin, deficiency in invasive carcinomas. Semin Cancer Biol 4:231-239, 1993. Choi BH, Holt JT, McDonald JV: Occult malignant astrocytoma of pons with extracranial metastasis to bone prior to craniotomy. Acta Neuropathol Berl 54:269-273, 1981. Dolman CL: Lymph node metastasis as first manifestation of glioblastoma. J Neurosurg 41:607-609, 1974. Farrier SE, Agosti S, Morgan M, et al: Metastatic ependymoma manifested by pancytopenia. South Med J 87:1262-1263, 1994. Friedlander DR, Zagzag D, Shiff B, et al: Migration of brain tumor cells on extracellular matrix proteins in vitro correlates with tumor type and grade and involves alpha V and beta 1 integrins. Cancer Res 56:1939-1947, 1996. Hoffman HJ, Duffner PK: Extraneural metastases of central nervous system tumors. Cancer 56:1778-1782, 1985. Hulbanni S, Goodman PA: Glioblastoma multiforme with extraneural metastases in the absence of previous surgery. Cancer 37:1577-1583, 1976. 10 Janisch W, Schreiber D, Guthert H: Neuropathologie – Tumoren des Nervensystems. Fischer, Stuttgart – New York, 1988. 11. Kung PC, Lee JC, Bakay L: Vascular invasion by glioma cells in man: an electron microscopic study. J Neurosurg 31:339-345, 1969. 12. Liwnicz BH, Rubinstein LJ: The pathways of extraneural spread in metastasizing gliomas: A report of three cases and critical review of the literature. Human Pathology 10:453-467, 1979. 13. Lossinsky AS, Mossakowski MJ, Pluta R, et al: Intercellular adhesion molecule-1, ICAM-1, upregulation in human brain tumors as an expression of increased blood-brain barrier permeability. Brain Pathol 5:339-344, 1995. 14. Mavroudis C, Townsend JJ, Wilson CB: A metastasizing ependymoma of the cauda equina. J Neurosurg 47:771-775, 1977. 15. McComb JG: Recent research into the nature of cerebrospinal fluid formation and absorption. J Neurosurg 59:369-383, 1983. 16. Mork SJ, Loken AC: Ependymoma: a follow-up study of 101 cases. Cancer 40:907-915, 1977. 17. Morris DM, Steinert HR, Wiernik PH: Ineffectiveness of chemotherapy in patients with metastatic ependymoma of the cauda equina. J Surg Oncol 22:33-36, 1983. 18. Newton HB, Henson J, Walker RW: Extraneural metastases in ependymoma. J Neurooncol 14:135-142, 1992. 19. Newton HB, Rosenblum MK, Walker RW: Extraneural metastases of infratentorial glioblastoma multiforme to the peritoneal cavity. Cancer 69:2149-2153, 1992. 20. Oehmichen M, Wietholter H, Gruninger H, et al: Destruction of intracerebrally applied red blood cells in cervical lymph nodes. Experimental investigations. Forensic Sci Int 21:43-57, 1983. 21. Oehmichen M, Gruninger H, Wietholter H, et al: Lymphatic efflux of intracerebrally injected cells. Acta Neuropathol Berl 45:61-65, 1979. 22. Pasquier B, Le Marc Hadour F, Dieny A, et al: Diffuse bone metastases. Cancer 68:2490-2491, 1991. 23. Paterson RJ Jr, Campbell WG Jr, Parsons H: Ependymoma of the cauda equina with multiple visceral metastases. J Neurosurgery 18:145-150, 1961. 24. Rubinstein LJ, Logan WJ: Extraneural metastases in ependymoma of the cauda equina. J Neurol Neurosurg Psychiatry 33:763-770, 1970. 25. Schreiber D, Schneider J, Heller T, et al: Intrakranielles Ependymom mit extra-neuralen Metastasen. Zentralbl Allg Pathol Pathol Anat 135:57-64, 1989. 26. Schroder R, Lorenzen J, Ostertag H, et al: Extraneurale Metastasierung von Hirn- und Ruckenmarktumoren. Bericht uber 2 Falle. Pathologe 16:223-229, 1995. 27. Schweitzer JS, Batzdorf U: Ependymoma of the cauda equina region: diagnosis, treatment, and outcome in 15 patients. Neurosurgery 30:202-207, 1992. 28. Sevick RJ, Johns RD, Curry BJ: Primary spinal primitive neuroectodermal tumor with extraneural metastases. Am J Neuroradiol 8:1151-1152, 1987. 29. Sharman KD: A metastasizing ependymoma of the cauda equina. Indian J Med Sci 10:639-641, 1956. 30. Sgouros S, Malluci CL, Jackowski A: Spinal ependymomas. The value of postoperative radiotherapy for residual disease control. Br J Neurosurg 10:559-566, 1996. 31. Sonneland PR, Scheithauer BW, Onofrio BM: Myxopapillary ependymoma. A clinico-pathologic and immunocytochemical study of 77 cases. Cancer 56:883-893, 1985. 32. Streit M, Schmidt R, Hilgenfeld RU, et al: Adhesion receptors in malignant transformation and dissemination of gastrointestinal tumors. Recent Results Cancer Res 142:19-50, 1996. 33. Vanneste JLA: Subacute bilateral malignant exophtalmus due to orbital medulloblastoma metastases. Arch Neurol 40:441-443, 1983. 34. Weiss L: A metastasizing ependymoma of the cauda equina. Cancer 8:161-171, 1955. 35. Wight DG, Holley KJ, Finbow JA: Metastasizing ependymoma of the cauda equina. J Clin Pathol 26:929-935, 1973. 36. Willis RA: The spread of tumors in the human body, 2th ed, Mosby, St Louis, 1952. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 56 EP 60 PG 5 ER PT J AU Rabczynsky, J Ziolkowski, P AF Rabczynsky, Jerzy Ziolkowski, Piotr TI Primary Endometrioid Carcinoma of Fallopian tube SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Fallopian tube carcinoma; endometrioid carcinoma; histology; immunohistology; p53 protein; c-erbB-2 ID Fallopian tube carcinoma; endometrioid carcinoma; histology; immunohistology; p53 protein; c-erbB-2 AB Twenty cases of primary Fallopian tube endometrioid carcinoma (PFTEC) are presented in the paper. This accounts for 42.5% of all histologic forms of primary Fallopian tube carcinoma (PFTC) found in our Department. The youngest patient was 38, and the oldest 68 years (mean: 56 years). Seven patients were nulliparas. Only two cases were bilateral. According to FIGO staging, 13 cases were evaluated as stage I, 4 as II, and 3 as stage III. Due to the histologic grading, 8 tumors were classified as well, 7 as moderately, and 5 as poorly differentiated. In the time of preparation of the manuscript, 12 women were still alive, 2 of them with recurrent disease. The follow-up of patients without recurrence ranged from 4 to 120 months (median: 63). Eight patients had died (survival time: from 4 to 65 months; median: 26). Metastases were found in 8 patients, especially to ovaries. In 14/20 cases of PFTEC various forms of tubal wall invasion were observed. Blood or lymphatic vessels involvement was found in 9 patients. Six out of them had died and one is alive with the symptoms of disease. Immunohistochemical detection of the mutant form of p53 protein and oncogene product, c-erbB-2, was studied in 17 cases. Nine patients exhibited simultaneous p53 protein accumulation and c-erbB-2 expression. 2/9 of these patients are alive with recurrent tumors and 4/9 died. Endometrioid carcinoma of the Fallopian tube can be characterized by a tendency to superficial invasion of tubal wall and in a half of the cases by invasion of vessels. The majority of these tumors were diagnosed at an early stage tumors. C1 [Rabczynsky, Jerzy] Wroclaw Medical University, Department of Pathology, ul. Marcinkowskiego 1, 50-386 Wroclaw, Poland. [Ziolkowski, Piotr] Wroclaw Medical University, Department of Pathology, ul. Marcinkowskiego 1, 50-386 Wroclaw, Poland. RP Ziolkowski, P (reprint author), Wroclaw Medical University, Department of Pathology, 50-386 Wroclaw, Poland. CR Barakat RR, Rubin SC, Saigo PE, et al: Cisplatin-based combination chemotherapy in carcinoma of the fallopian tube. Gynecol Oncol 42:156-160, 1991. Benedet JL, White GW, Fairey RN, et al: Adenocarcinoma of the Fallopian tube. Experience with 41 patients. Obstet Gynecol 50:654-657, 1977. Dembo AJ, Davy M, Stenwig AF, et al: Prognostic factors in patients with stage I epithelial ovarian cancer. Obstet Gynecol 75:263-273, 1990. Eltabbakh GH, Belinson JL, Kennedy AW, et al: p53 overexpression is not an independent prognostic factor for patients with primary ovarian epithelial cancer. Cancer 80:892-898, 1997. Hellstrom AC, Silfversward C, Nilsson B, et al: Carcinoma of the Fallopian tube. A clinical and histopathologic review. The Radiumhemmet series. Int J Gynecol Cancer 4:395-400, 1994. Hu CY, Taymor ML, Hertig AT: Primary carcinoma of the Fallopian tube. Am J Obstet Gynecol 59:58-67, 1950. Jereczek B, Jassem J, Kobierska A: Primary cancer of the Fallopian tube. Report of 26 patients. Acta Obstet Gynecol Scand 75:281-286, 1996. Kojs Z, Urbanski K, Karolewski K, et al: Pierwotny rak jajowodu. Analiza 32 przypadkow. Gin Pol 67:612-614, 1996. Koonings PP, Campbell K, Mishell DR, et al: Relative frequency of primary ovarian neoplasms: a 10-year review. Obstet Gynecol 74:921-926, 1989. Lacy MQ, Hartmann LC, Keeney GL, et al: c-erbB-2 and p53 expression in Fallopian tube carcinoma. Cancer 75:2891-2896, 1995. Levesque MA, Katsaros D, Yu H, et al: Mutant p53 protein overexpression is associated with poor outcome in patients with well or moderately differentiated ovarian carcinoma. Cancer 75:1327-1338, 1995. Lukes AS, Kohler MF, Pieper CF, et al: Multivariable analysis of DNA ploidy, p53, and HER-2/neu as prognostic factors in endometrial cancer. Cancer 73:2380-2385, 1994. Mittal KR, Barwick KW: Diffusely infiltrating adenocarcinoma of the endometrium. A subtype with poor prognosis. Am J Surg Pathol 12:754- 758, 1988. Navani SS, Alvarado-Cabbero I, Young RH, et al: Endometrioid carcinoma of the Fallopian tube: a clinicopathologic analysis of 26 cases. Gynecol Oncol 63:371-378, 1996. Nordin AJ: Primary carcinoma of the Fallopian tube: a 20-year literature review. Obstet Gynecol Surv 49:349-361, 1994. Peters WA, Andersen WA, Hopkins MP, et al: Prognostic features of carcinoma of the Fallopian tube. Obstet Gynecol 71:757-762, 1988. Poulsen HE, Taylor CW, Sobin LH: Histological typing of female genital tract tumors.WHO Geneva 1975, 74-77. Rabczynski J, Ziokowski P, Kowalski P, et al: Primary carcinoma of the fallopian tube – clinico-morphological review of 41 cases. Med Sci Monit 1998, in press). Rorat E, Wallach RC: Endometrioid carcinoma of the Fallopian tube: pathology and clinical outcome. Int J Gynecol Obstet 32:163-167, 1990. Rosen A., Klein M., Lahousen M., et al: Das primare tubenkarzinom – eine oesterreichische multizenterstudie. Geburtsh u Frauenheilk 53:321- 325, 1993. Salvesen HB, Akslen LA, Albrektsen G, et al: Poorer survival of nulliparous women with endometrial carcinoma. Cancer 82:1328-1333, 1998. Schiller HM, Silverberg SG: Staging and prognosis in primary carcinoma of the Fallopian tube. Cancer 28:389-395, 1971. Scully RE, Bonfiglio TA, Kurman RJ, et al: Histological typing of female genital tract tumors 2nd ed., Springer Verlag. Berlin, Heidelberg, New York, London, Paris, Tokyo, Hong Kong, Barcelona, Budapest, 1994, 15. Serov SF, Scully RE, Sobin LH: International Histological Classification of Tumors, no. 9: Histological Typing of Ovarian tumors. WHO Geneva 1973. Sedlis A: Primary carcinoma of the Fallopian tube. Obstet Gynecol Surv 16:209-226, 1961. Tidy J, Mason WP: Endometrioid carcinoma of the ovary: a retrospective study. Brit J Obst Gynecol 95:1165-1169, 1988. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 61 EP 66 PG 6 ER PT J AU Nemes, B Podder, H Jaray, J Dabasi, G Lazar, L Schaff, Zs Sotonyi, P Perner, F AF Nemes, Balazs Podder, Hemangshu Jaray, Jeno Dabasi, Gabriella Lazar, Laura Schaff, Zsuzsa Sotonyi, Peter Perner, Ferenc TI Primary Hepatic Carcinoid in a Renal Transplant Patient SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE renal transplant; hepatic carcinoid ID renal transplant; hepatic carcinoid AB There seems to be a world-wide increase in the incidence of tumors among immunosuppressed patients. Of 1350 renal allografts transplanted in the past 23 years at the Department of Transplantation and Surgery, 56 cases had malignant tumors. The case of a 58-year-old female patient is reported, with disseminated primary carcinoid in the liver detected 86 days after renal transplantation. According to the literature only 39 patients with primary liver carcinoids have been reported until 1997, but this is the first where the carcinoid developed in an immunosuppressed patient. The rapid progression of the carcinoid could be associated with the immunosuppression. C1 [Nemes, Balazs] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23., H-1082 Budapest, Hungary. [Podder, Hemangshu] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23., H-1082 Budapest, Hungary. [Jaray, Jeno] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23., H-1082 Budapest, Hungary. [Dabasi, Gabriella] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23., H-1082 Budapest, Hungary. [Lazar, Laura] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H1085 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sotonyi, Peter] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23., H-1082 Budapest, Hungary. [Perner, Ferenc] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23., H-1082 Budapest, Hungary. RP Nemes, B (reprint author), Semmelweis University, Department of Transplantation and Surgery, H-1082 Budapest, Hungary. EM nemes@mailtrans.sote.hu CR Alpert LI, Zak FG, Werthammer S, et al: Cholangiocarcinoma: a clinico-pathologic study of five cases with ultrastructural observations. Hum Pathol 5:709-728, 1974. Andreola S, Lombardi L, Audisio RA, et al: A clinicopathologic study of primary hepatic carcinoids. Cancer 65:1211-1218, 1990. Artopoulos JG, Destuni C: Primary mixed hepatocellular carcinoma with carcinoid characteristics. A case report. Hepatogastroenterology 41:442-444, 1994. de Gennes JL, Kiortsis DN, Dairon F et al: Metastatic pulmonary carcinoma revealed by Cushing syndrome initially considered to have pituitary origin. Presse Med 24:1605-1617, 1995. Godwin JD II: Carcinoid tumors: an analysis of 2837 cases. Cancer 36:560, 1975. Goldblum JR, Lloyd RV: Primary renal carcinoid. Case report and review of the literature. Arch Pathol Lab Med 117:855-858, 1993. Hayashi R, Hanyu N, Moriyama S: Efficacy of steroid therapy on liver metastasis of thymic carcinoid. Intern Med 33:45-47, 1994. Himeno H, Yamamoto Y: Primary hormon producing hepatic tumor. Ryoikibetsu Shokogun Shirizu 7:511-514, 1995. Holbrook RF, Koo K, Ryan JH: Resection of malignant primary liver tumors. Am J Surg 171:453-455, 1996. Hsueh CI, Tan XD, Gonzalez Crussi F: Primary hepatic neuroendocrine carcinoma in a child. Morphologic, immuncytochemical and molecular biologic studies. Cancer 71:2660-2665, 1993. Imaoka I, Sugimura K, Tamura K: Case report. MR imaging of a carcinoid tumor of the liver. Clin Radiol 47:287-289, 1993. Inoue Y, Nakamura H, Mizumoto S, et al: Primary hepatic carcinoid with production of gastrin: a case report. Radiat Med 11:102-106, 1993. Krishnamurthy SC, Dutta V, Pai SA, et al: Primary carcinoid tumor of the liver. Report of four resected cases. J Surg Oncol 62:218-221, 1996. Lemaire LCJM, Pols HAP, Tilanus HW: Carcinoid tumor presenting as giant hepatic cyst. Br J Surg 82:133, 1995. Marthur SK, Supe AN, Nagral SS, et al: Primary hepatic carcinoid, a case report). Indian J Gastroenterol 11(abstr.8), 1992. Mauer CA, Baer HU, Dyong TH, et al: Carcinoid of the pancreas. Eur J Cancer 32A:1109-16, 1996. Pi ZM: 20 rare primary hepatic malignant tumors. Chung Hua Chung Liu Tsa Chih 14:210-212, 1992. Shima K, Ota G: Primary hepatic carcinoid. Ryoikibetsu Shokogun Shirizu 7:450-452, 1995. Sioutos N, Virta S, Kessimian N: Primary hepatic carcinoid tumor. An electron microscopic and immunohistochemical study. Am J Clin Pathol 95:172-175, 1991. Takayashu K, Muramatsu Y, Sakamoto M, et al: Findings in primary hepatic carcinoid tumor: US, CT, MRI and angiography. J Comp Ass Tomography 16:99-102, 1992. Toth A, Alfoldy F, Jaray J, et al: Malignant tumors after renal transplantation. Acta Chir Hung 33:211-216, 1992. Volte A, Iglicki F, Sevenet F, et al: Treatment of metastatic carcinoid with interferon alpha. Presse Med 25:63-67, 1996. Yamashita Y, Takahashi M, Tsuji A, et al: Primary carcinoid tumor of the liver: a case report. J Comput Tomogr 10:313-317, 1986. Yasoshima H, Uematsu K, Sakurai K, et al: Primary hepatic carcinoid. Acta Pathol Jpn 43:783-789, 1993. Yu-Ping X, Ji-Yao Y: Primary neuroendocrine carcinoma of the liver. Ultrastruct Pathol 10:313-317, 1986. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 67 EP 69 PG 3 ER PT J AU Rath-Wolfson, L Koren, R Yaniv, E Sadov, R Gal, R AF Rath-Wolfson, Lea Koren, Rumelia Yaniv, Eitan Sadov, Rima Gal, Rivka TI A New Rapid Technique for the Fixation of Thyroid Gland Surgical Specimens SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE lymph node reveling solution; thyroid; fixation ID lymph node reveling solution; thyroid; fixation AB One of the main diagnostic problems in thyroid pathology is to distinguish between follicular adenoma and follicular carcinoma. Thorough sampling of the nodule's capsule is recommended in order to identify capsular invasion. However, during the hardening of the tissue, by the usual fixatives the capsule shrinks and rolls downwards and sometimes the capsule separates from the remaining tissue. The present work evaluates the use of ''Lymph Node Revealing Solution'' (LNRS) for the rapid fixation (2h) of different thyroid lesions as compared to that of formalin. Fifty-one unselected consecutive cases of thyroid nodules, which included various benign and malignant lesions, were examined. Each specimen was cut in two equal parts; one was fixed in LNRS, the other in formalin. Fixation in LNRS for 2 hours gave adequate results in sectioning and staining of the tissue, and excellent immunostains. Its advantage over formalin is the conservation of the natural relationship between the capsule and the rest of the tissue, on the same plane, as well as the short time required for the final diagnosis. C1 [Rath-Wolfson, Lea] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of PathologyTel Aviv, Israel. [Koren, Rumelia] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of PathologyTel Aviv, Israel. [Yaniv, Eitan] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Otolaryngology-Head and Neck SurgeryTel Aviv, Israel. [Sadov, Rima] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Otolaryngology-Head and Neck SurgeryTel Aviv, Israel. [Gal, Rivka] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of PathologyTel Aviv, Israel. RP Koren, R (reprint author), Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Pathology, Tel Aviv, Israel. CR Koren R, Halpern M, Klein B, et al: A new rapid technique for the fixation of lymph nodes. Cell Vision 3:437-438, 1996. Mc Marcus JFA, Mowry RW: Staining methods, histologic and histochemical. Harper & Brothers, New York, 1960, pp 19-21. Lilie: Histologic technic and practical histochemistry. 3rd edition. Mc Grow-Hill Book Company, New York, 1965, pp 48-53. Luna LG, ed): Manual of histologic staining methods of the Armed Forces Institute of Pathology. 3rd edition. McGraw-Hill Book Company, New York, pp 1968, 4-5. Hsu SM, Raine L, Fanger H: Use of avidin-biotin-peroxidase complex, ABC, in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody, PAP, procedures. J Histochem Cytochem 29:577-580, 1981. Bugis SP, Young JEM, Archibald SD, et al: Diagnostic accuracy of fine-needle aspiration biopsy versus frozen section in solitary nodules. Am J Surg 152:411-416, 1986. Rosai J: Ackerman’s surgical pathology. Thyroid gland frozen section. 8th edition. Mosby, St. Louis, 1996, pp 545-546. Prophet EB, ed): Laboratory methods in histotechnology: Armed Forces Institute of Pathology, Washington DC. 1992. Noguchi M, Furuya S, Takeuchi T, et al: Modified formalin and methanol fixation methods for molecular biological and morphological analyses. Pathol Int 47:685-691, 1997. Horbin RW: Problems and artifacts of microwave accelerated procedures in neurohistotechnology and resolution. Methods 15:101-106, 1998. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 70 EP 72 PG 3 ER PT J AU Mikala, G Xie, J Berencsi, Gy Kiss, Cs Marton, I Domjan, Gy Valyi-Nagy, I AF Mikala, Gabor Xie, Jiuru Berencsi, Gyorgy Kiss, Csongor Marton, Ildiko Domjan, Gyula Valyi-Nagy, Istvan TI Human Herpesvirus 8 in Hematologic DiseasesFNT1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE HHV-8; tumorigenesis ID HHV-8; tumorigenesis AB Human herpesvirus type 8 (HHV-8), also known as Kaposi's sarcoma-associated herpesvirus (KSHV) is a new member of the gamma-herpesvirus family. It is an unusual herpesvirus in that it carries a large number of genes that encode oncoproteins or cell signaling proteins. In addition to being the causative agent of both HIV-associated and non-HIV-associated Kaposi's sarcoma this DNA tumor virus has been implicated in the pathogenesis of several diseases. These include multiple myeloma (MM), Waldenstrom's macroglobulinemia (WM), multicentric Castleman's disease (MCD), body cavity-based lymphoma (BCBL), and various other conditions such as sarcoidosis and pemphigus. While the causative role of the viral infection is fairly certain in the development of BCBL and multicentric Castleman's disease, HHV-8 may act through a different mechanism to induce plasma cell malignancies. It has been suggested - though the finding is still controversial - that infection of bone marrow stromal dendritic cells by HHV-8 might be a key factor in the etiology and pathogenesis of monoclonal gammopathies. The aim of this review is to provide a short introduction into the tumorigenic potential of HHV-8 as well as to detail the available data and possible mechanisms on the involvement of this virus in different hematologic diseases. C1 [Mikala, Gabor] Imre Haynal University of Health Sciences, First Department of Internal Medicine, Szabolcs u. 35., H-1135 Budapest, Hungary. [Xie, Jiuru] Bela Johan National Institute of HygieneBudapest, Hungary. [Berencsi, Gyorgy] Bela Johan National Institute of HygieneBudapest, Hungary. [Kiss, Csongor] Medical and Health Science Center, University of Debrecen, Department of PediatricsDebrecen, Hungary. [Marton, Ildiko] University Medical School of Debrecen, Department of StomatologyDebrecen, Hungary. [Domjan, Gyula] Imre Haynal University of Health Sciences, First Department of Internal Medicine, Szabolcs u. 35., H-1135 Budapest, Hungary. [Valyi-Nagy, Istvan] Imre Haynal University of Health Sciences, First Department of Internal Medicine, Szabolcs u. 35., H-1135 Budapest, Hungary. RP Valyi-Nagy, I (reprint author), Imre Haynal University of Health Sciences, First Department of Internal Medicine, H-1135 Budapest, Hungary. CR Agbalika F, Mariette X, Marolleau J-P, et al: Detection of human herpesvirus-8 DNA in bone marrow biopsies from patients with multiple myeloma and Waldenstroms macroglobulinemia. Blood 91:4393-4394, 1998. Alsina M, Gao SJ, Montalvo EA, et al: Increased prevalence of antibodies to Kaposi’s sarcoma-associated herpesvirus, human herpesvirus 8, in patients with multiple myeloma. Blood 90, Suppl 1):87a, 1997. Arvanikatis L, Mesri EA, Nador RG, et al: Establishment and characterization of a primary effusion, body cavity-based, lymphoma cell line, BC-3, harboring Kaposi’s sarcoma-associated herpesvirus, KSHV/HHV-8, in the absence of Epstein-Barr virus. Blood 88:2648-2654, 1996. Asuo H, Said JW, Yang R, et al: Mechanisms of growth control of Kaposi’s sarcoma-associated herpesvirus-associated primary effusion lymphoma cells. Blood 91:2475-2481, 1998. Bais C, Santomasso B, Coso O, et al: G-protein-coupled receptor of Kaposi’s sarcoma-associated herpesvirus is a viral oncogene and angiogenesis activator. Nature 391:86-89, 1998. Brooks LA, Wilson AJ and Crook T: Kaposi’s sarcoma-associated herpesvirus, KSHV, / Human herpesvirus 8, HHV-8, - A new human tumor virus. J Pathol 182:262-265, 1997. Brousset P, Meggetto F, Attal M, et al: Kaposi’s sarcoma-associated herpesvirus infection and multiple myeloma. Science 278:1972-1973, 1997. Brousset P, Theriault C, Roda D, et al: Kaposi’s sarcomaassociated herpesvirus, KSHV, in bone marrow biopsies of patients with Waldenstroms macroglobulinemia. Br J Haematol 102:795-797, 1998. Burger R, Neipel F, Fleckenstein B, et al: Human herpesvirus type 8 interleukin-6 homologue is functionally active on human myeloma cells. Blood 91:1858-1863, 1998. Cathomas G, Stalder A and Kurrer MO: Multiple myeloma and HHV8 infection. Blood 91:4391, 1998. Cheng EH-Y, Nicholas J, Bellows DS, et al: A Bcl-2 homolog encoded by Kaposi sarcoma-associated virus, human herpesvirus 8, inhibits apoptosis but does not heterodimerize with Bax or Bak. Proc Natl Acad Sci USA 94:690-694, 1997. DeGreef C, Bakkus M, Heirman C, et al: The absence of Kaposi sarcoma-associated herpesvirus, KSHV, DNA sequences in leukapheresis products and ex vivo expanded CD34+ cells in multiple myeloma, MM, patients. Blood 90, Suppl 1):86a, 1997. DePond W, Said JW, Tasaka T, et al: Kaposi sarcoma-associated herpesvirus and human herpesvirus 8, KSHV/HHV-8)-associated lymphoma of the bowel. Am J Surg Pathol 21:719-724, 1997. DiAlberti L, Piattelli A, Artese L, et al: Human herpesvirus 8 variants in sarcoid tissues. Lancet 350:1655-1661, 1997. Flore O, Rafii S, Ely S, et al: Transformation of primary human endothelial cells by Kaposi’s sarcoma-associated herpesvirus. Nature 394:588-592, 1998. Gao SJ, Alsina M, Deng JH, et al: Antibodies to Kaposi’s sarcoma- associated herpesvirus, human herpesvirus 8, in patients with multiple myeloma. J Infect Dis 178:846-849, 1998. Gessain A, Sudaka A, Briere J, et al: Kaposi sarcoma-associated herpes-like virus, human herpesvirus type 8, DNA sequences in multicentric Castlemans disease: Is there any relevant association in non-human immunodeficiency virus-infected patients? Blood 87:414-416, 1996. Gura T: Causing cancer by remote control? Science 276:1788- 1789, 1997. Hyjek E, Fata F, Niesvisky R, et al: Low prevalence of antibodies of Kaposi’s sarcoma-associated herpesvirus, KSHV/HHV-8, in patients with multiple myeloma. Blood 90, Suppl 1):87a, 1997. Kikuta H, Itakura O, Taneichi K, et al: Tropism of human herpesvirus 8 for peripheral blood lymphocytes in patients with Castlemans disease. Br J Haematol 99:790-793, 1997. Lee H, Veazey R, Williams K, et al: Deregulation of cell growth by the K1 gene of Kaposi’s sarcoma-associated herpesvirus. Nat Med 4:435-440, 1998. Lee H, Guo J, Li M, et al: Identification of an immunoreceptor tyrosine-based activation motif of K1 transforming protein of Kaposi’s sarcoma-associated herpesvirus. Mol Cell Biol 18:5219-5228, 1998. Lennette ET, Blackburn DJ and Levy JA: Antibodies to human herpesvirus type 8 in the general population and in Kaposi’s sarcoma patients. Lancet 348:858-861, 1996. MacKenzie J, Sheldon J, Morgan G, et al: HHV-8 and multiple myeloma in the UK. Lancet 350:1144, 1997. Marcelin A-G, Dupin N, Bouscary D, et al: HHV-8 and multiple myeloma in France. Lancet 350:1144, 1997. Moore SP, Boshoff C, Weiss RA, et al: Molecular mimicry of human cytokine and cytokine response pathway genes by KSHV. Science 274:1739-1743, 1996. Muralidhar S, Pumfery AM, Hassani M, et al: Identification of kaposin, open reading frame K12, as a human herpesvirus 8, Kaposi’s sarcoma associated herpesvirus, transforming gene. J Virol 72:4980-4988, 1998. Nador RG, Cesarman E, Chadburn A, et al: Primary effusion lymphoma: A distinct clinicopathologic entity associated with the Kaposi’s sarcoma-associated herpesvirus. Blood 88:645- 656, 1996. Parravicini C, Corbellino M, Paulli M, et al: Expression of a virus-derived cytokine, KSHV vIL-6, in HIV-seronegative Castlemans disease. Am J Pathol 151:1517-1521, 1997. Rabkin CS, Schultz TF, Whitby D, et al: Interassay correlation of human herpesvirus 8 serologic tests. HHV-8 Interlaboratory Collaborative Group. J Infect Dis 178:304-309, 1998. Rettig MB, Ma HJ, Vescio RA, et al: Kaposi’s sarcoma-associated herpesvirus infection of bone marrow dendritic cells from multiple myeloma patients. Science 276:1851-1854, 1997. Rettig M, Vescio R, Ma H, et al: Detection of Kaposi’s sarcomaassocited herpesvirus in the dendritic cells of Waldenstroms macroglobulinemia and primary amyloidosis patients. Blood 90, Suppl 1):86a, 1997. Rettig M, Vescio R, Ma H, et al: Sequence variability of HHV- 8 from the dendritic cells of multiple myeloma patients. Blood 90, Suppl 1):86a, 1997. Russo JJ, Bohenzky RA, Chien M-C, et al: Nucleotide sequence of the Kaposi sarcoma-associated herpesvirus, HHV-8). Proc Natl Acad Sci USA 93:14862-14867, 1996. Said JW, Rettig MR, Heppner K, et al: Localization of Kaposi’s sarcoma-associated herpesvirus in bone marrow biopsy samples from patients with multiple myeloma. Blood 90:4278-4282, 1997. Schultz TF: Kaposi’s sarcoma-associated herpesvirus, human herpesvirus-8). J Gen Virol 79:1573-1591, 1998. Smith KJ, et al: Human herpesvirus 8 DNA sequences in pemphigus: The role of the virus in oncogenic and autoimmune manifestations. Arch Dermatol 134:751-754, 1998. Swanton C, Mann DJ, Fleckenstein B, et al: Herpes viral cyclin / Cdk6 complexes evade inhibition by CDK inhibitor proteins. Nature 390:184-187, 1997. Tarte K, Olsen SJ, Lu ZY, et al: Clinical-grade functional dendritic cells from patients with multiple myeloma are not infected with Kaposi’s sarcoma-associated herpesvirus. Blood 91:1852-1857, 1998. Weiss RA, Whitby D, Talbot S, et al: Human herpesvirus type 8 and Kaposi’s sarcoma. J Natl Cancer Inst Monogr 23:51-54, 1998. Yi Q, Ekman M, Anton D, et al: Blood dendritic cells from myeloma patients are not infected with Kaposi’s sarcoma-associated herpesvirus, KSHV/HHV-8). Blood 92:402-404, 1998. Zimring JC, Goodburn S and Offerman MK: Human herpesvirus encodes an interferon regulatory factor, IRF, homolog that represses IRF-1 mediated transcription. J Virol 72:701-707, 1998. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 1999 VL 5 IS 1 BP 73 EP 79 PG 7 ER PT J AU Matolcsy, A AF Matolcsy, Andras TI Primary Effusional Lymphoma: A New Non-Hodgkin’s Lymphoma Entity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE lymphoma; effusional ID lymphoma; effusional AB A distinct non-Hodgkin’s lymphoma (NHL) entity that grow in the body cavities as lymphomatous effusions in the absence of clinically identifiable tumor masses has been defined as primary effusional lymphoma (PEL). This lymphoma characterized by distinetive morphology, immunophenotype, genotype and association with Kaposi’s sarcoma-associated herpesvirus (KHSV)/human herpesvirus-8 (HHV-8) infection. In this minireview, the clinico-pathological and biological characteristics of PELs are summarized. C1 [Matolcsy, Andras] University of Pecs, Department of Pathology, Szigeti ut 12, H-7624 Pecs, Hungary. RP Matolcsy, A (reprint author), University of Pecs, Department of Pathology, H-7624 Pecs, Hungary. EM amatolc@pathology.pote.hu CR Knowles DM, Inghirami G, Ubriaco A, et al: Molecular genetic analysis of three AIDS-associated neoplasms of uncertain lineage demonstrates their B-cell derivation and the possible pathogenetic role of the Epstein-Barr virus. Blood 73:792-799, 1989. Walts AE, Shintaku IP, Said JW: Diagnosis of malignant lymphoma in effusions from patients with AIDS by gene rearrangement. Am J Clin Pathol 94:170-175, 1990. Nador RG, Cesarman E, Chadburn A, et al: Primary effusion lymphoma: a distinct clinicopathologic entity associated with the Kaposi’s sarcoma-associated herpes virus. Blood 88:645- 656, 1996. Cesarman E, Chang Y, Moore PS, et al: Kaposi’s sarcoma-associated herpesvirus-like DNA sequences in AIDS-related bodycavity- based lymphomas. N Engl J Med 332:1186-1191, 1995. Jaffe ES, Harris NL, Diebold J, et al: World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues. A progress report. Am J Clin Pathol 111(suppl. 1):8-12, 1999. Carbone A, Gloghini A, Vaccher E, et al: Kaposi’s sarcomaassociated herpesvirus DNA sequences in AIDS-related and AIDS-unrelated lymphomatous effusions. Br J Haematol 94:533-543, 1996. Nador RG, Cesarman E, Knowles DM, et al: Herpes-like DNA sequences in a body-cavity-based lymphoma in an HIV-negative patient. N Engl J Med 333:943, 1996. Ansari MQ, Dawson DB, Nador R, et al: Primary body cavitybased AIDS-related lymphomas. Am J Clin Pathol 105:221- 229, 1996. Green I, Espiritu E, Ladanyi M, et al: Primary lymphomatous effusions in AIDS: a morphological, immunophenotypic, and molecular study. Modern Pathol 8:39-45, 1995. Matolcsy A, Nador RG, Cesarman E, et al: Immunoglobulin VH gene mutational analysis suggest that primary effusion lymphomas derive from different stages of B cell maturation. Am J Pathol 153:1609-1614, 1998. Gaidan G, Capello D, Cilia AM, et al: Genetic characterization of HHV-8/KSHV-positive primary effusion lymphoma reveals frequent mutations of BCL6:implications for disease pathogenesis and histogenesis. Genes Chromosomes Cancer 24:16-23, 1999. Moor PS, Gao S-J, Dominguez G, et al: Primary characterization of a herpesvirus agent associated with Kaposi’s sarcoma. J Virol 70:549-558, 1996. Ambroziak JA, Herndier BG, Glogau RG, et al: Herpes-like sequences in HIV-infected and uninfected Kaposi’s sarcoma patients. Science 268:582-583, 1995. Cesarman E, Nador RG, Bai F, et al: Kaposi’s sarcoma associated herpesvirus contains G protein-coupled receptor and cyclin D homologs which are expressed in Kaposi’s sarcoma and malignant lymphoma. J Virology 70:8218-8223, 1996. Chang Y, Moore PS, Talbot SJ, et al: Cyclin encoded by KS herpesvirus. Nature 382:410, 1996. Horenstein MG, Nador RG, Chadburn A, et al: Epstein-Barr virus latent gene expression in primary effusion lymphomas containing Kaposi’s sarcoma-associated herpesvirus/human herpesvirus-8. Blood 90:1186-1191, 1997. Wilson JB, Bell JL, Levine AJ: Expression of Epstein-Barr virus nuclear antigen-1 induces B cell neoplasia in transgenic mice. EMBO J 15:3117-3122, 1996. Asou HA, Said JW, Yang R, et al: Mechanisms of growth control of Kaposi’s sarcoma-associated herpes virus-associated primary effusional lymphoma cells. Blood 91:2475-2481, 1998. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 87 EP 89 PG 3 ER PT J AU Tanyi, J Tory, K Bankfalvi, Shroder, W Rath, W Fuzesi, L AF Tanyi, Janos Tory, Kalman Bankfalvi, Agnes Shroder, Willibald Rath, Werner Fuzesi, Laszlo TI Analysis of p53 Mutation and Cyclin D1 Expression in Breast Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; fibroadenoma; p53; mutation; cyclin D1; immunohistochemistry ID breast cancer; fibroadenoma; p53; mutation; cyclin D1; immunohistochemistry AB P53 and cyclin D1 are interacting regulatory genes and both are frequently altered in breast cancer. We analysed p53 mutation by SSCP and sequencing methods as well as p53 protein accumulation immunohistochemically in 34 consecutively operated breast tumors. None of 4 fibroadenomas revealed p53 mutation or p53 protein accumulation. Mutation of p53 was present in 7 carcinomas. Immunohistochemistry revealed accumulation of p53 protein in 6 carcinomas and there was a significant correlation between p53 mutation and protein accumulation. Overexpression of cyclin D1 protein was observed in 11 carcinomas by immunohistochemistry and no correlation was observed between cyclin D1 overexpression and p53 mutation or accumulation. Our data support the concept that the p53-cyclin D1 signal pathway and the cyclin D1 cascade are disregulated in breast cancer. C1 [Tanyi, Janos] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Baross str. 27., H-1088 Budapest, Hungary. [Tory, Kalman] Medical Faculty of the Technical University of Aachen, Department of PathologyAachen, Germany. [Bankfalvi, Agnes] Medical Faculty of the Technical University of Aachen, Department of GynaecologyAachen, Germany. [Shroder, Willibald] University of Munster, Department of PathologyMunster, Germany. [Rath, Werner] University of Munster, Department of PathologyMunster, Germany. [Fuzesi, Laszlo] Medical Faculty of the Technical University of Aachen, Department of PathologyAachen, Germany. RP Tanyi, J (reprint author), Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, H-1088 Budapest, Hungary. EM tanyi©noi1.sote.hu CR Andersen TI, Holm R, Nesland JM, et al: Prognostic significance of TP53 alterations in breast carcinoma. Br J Cancer 68:540-548, 1993. Bates S, Parry D, Bonetta L, et al: Absence of cyclin D/cdk complexes in cells lacking functional retinoblastoma protein. Oncogene 9:1633-1640, 1994. Bankfalvi A, Navabi H, Bier B, et al: Wet autoclave pretreatment for antigen retrieval in diagnostic immunohistochemistry. J Pathol 174:223-228, 1994. Chen X, Bargonetti J, Prives C, et al: P53, through p21, WAF1/CIP1, induces cyclin D1 synthesis. Cancer Res 55:4257-6342, 1995. Cordon-Cardo C: Mutation of cell cycle regulators. Am J Pathol 147:545-560, 1995. El-Deiry WS, Tokino T, Velculescu V E, et al: WAF1, a potential mediator of p53 tumor suppression. Cell 75:817-825, 1993. Fantl V, Smith R, Brokes S, et al: Chromosome 11q13 abnormalities in human breast cancer. Cancer Surv 18:77-94, 1993. Gillett C, Smith P, Gregory W, et al: Cyclin D1 and prognosis in human breast cancer. Int J Cancer 69:92-99, 1996. Ggrana K, Reddy EP: Cell cycle control in mammalian cells: role of cyclins, cyclin dependent kinases, CDKs), growth suppressor genes and cyclin-dependent kinase inhibitors, CKIs). Oncogene 11:211-219, 1995. Greenblatt MS, Bennett WP, Hollstein M, et al: Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res 54:4855-4878, 1994. Hollstein M, Sidransky D, Vogelstein B, et al: p53 mutation in human cancers. Science 253:49-53, 1991. Jacquemier J, Moles JP, Adelaide J, et al: P53 immunohistochemical analysis in breast cancer with four monoclonal antibodies: comparison of staining and PCR-SSCP results. Br J Cancer 89:846-852, 1994. Kastan MB, Zhan Q, El-Deiry WS, et al: A mammalian cell cycle checkpoint pathway utilizing p53 and GADD45 is defective in ataxia-teleangiectasia. Cell 71:587-597, 1992. Kovach JS, Hartmann A, Blaszik H, et al: Mutation detection by highly sensitive method indicates that p53 gene mutations in breast cancer can have important prognostic value. Proc Natl Acad Sci USA 93:1093-1096, 1996. Kranenburg O, van der Eb AJ, Zantema A: Cyclin D1 is an essential mediator of apoptotic neuronal cell death. EMBO J 15:46-54, 1996. Lane DP: P53 guardien of genome. Nature 359:15-16, 1992. Li Y, Jenkins C, Nichols M, et al: Cell cycle expression and p53 regulation of the cyclin-dependent kinase inhibitor p21. Oncogene 9:2261-2268, 1994. Lidereau R, Mathieu D, Escot C, et al: Genetic variability of proto-oncogenes for breast cancer risk and prognosis. Biochemie 70:951-959, 1988. Lucibello FC, Sewing A, Brusselbach S, et al: Deregulation of cyclin D1 and E and suppression of cdk2 and cdk4 in senescenct human fibroblasts. J Cell Sci 105:123-133, 1993. Matsushime H, Quelle DE, Shurtleff SA, et al: D-type cyclindependent kinase activity in mammalian cells. Mol Cell Biol 14:2066-2076, 1994. McIntosh GG, Anderson JJ, Milton I, et al: Determination of the prognostic value of cyclin D1 overexpression in breast cancer. Oncogene 11:885-891, 1995. Michalides R, Hageman P, Tinteren H, et al: A clinicopathological study on overexpression of cyclin D1 and p53 in a series of 248 patients with operable breast cancer. Brit J Cancer 73:728-734, 1996. Schauer IE, Siriwardana S, Langan TA, et al: Cyclin D1 overexpression vs. retinoblastoma inactivation: implications for growth control evasion in non-small cell and small cell lung cancer. Proc Natl Acad Sci USA 91:7827-7831, 1994. Shuuring E, Verhoeven E, van Tinteren H, et al: Amplification of genes within the chromosome 11q13 region is indicative of poor prognosis in patients with operable breast cancer. Cancer Res 52:5229-5234, 1992. Thor AD, Moore DH, Edgerton SM, et al: Accumulation of p53 tumor suppressor gene protein: an independent marker of prognosis in breast cancer. JNCI 84:845-855, 1992. Toguchida J, Yamaguchi T, Rithchie B, et al: Mutation spectrum of the p53 gene in bone and soft tissue sarcomas. Cancer Res 52:6194-6199, 1992. Vogelstein B, Kinzler KW: P53 function and disfunction. Cell 70:523-526, 1992. Waga S, Hannon GJ, Beach D, et al: The p21 inhibitor of cyclin dependent kinases controls DNA replication by interaction with PCNA. Nature 369:574-578, 1994. Wang TC, Cardiff RD, Zukerberg L, et al: Mammary hyperplasia and carcinoma in MMTV-cyclin D1 transgenic mice. Nature 369:669-671, 1994. Wu X, Bayle JH, Olson D, et al: The p53-mdm2 autoregulatory feedback loop. Genes Dev 7:1126-1132, 1993. Xiong Y, Hannon GJ, Zhang H, et al: P21 is a universal inhibitor of cyclin kinases. Nature 366:701-704, 1993. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 90 EP 94 PG 5 ER PT J AU Nair, P Nair, MK Jayaprakash, GP Pillai, MR AF Nair, Prapid Nair, M Krishnan Jayaprakash, G Puthuveetil Pillai, M Radhakrishna TI Decreased Programmed Cell Death in the Uterine Cervix Associated with High Risk Human Papillomavirus Infection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HPV; apoptosis; cervical cancer ID HPV; apoptosis; cervical cancer AB The relationship between apoptosis, apoptosis regulatory proteins, cell proliferation and human papillomavirus infection during various phases of tumor progression in the uterine cervix was studied. Apoptosis was defined by morphological criteria and the TUNEL assay. Expression of p53, bcl-2, bax, cyclin D1, Ki 67 and E6 protein was evaluated by immunocytochemistry. Presence of mutant p53 was detected using a mutant specific ELISA. Type of HPV infection was determined by PCR using type specific primers. Apoptosis showed significant negative correlation with increasing histological abnormality (p=0.0005). Higher tumor cell proliferation was associated with increasing histological abnormality (p=0.001 for Ki 67 and cyclin D1). There was significant correlation between histological grade and immunoreactivity of p53 (p=0.0001 ) and bcl-2 (p=0.0002). However, mutant p53 was expressed by only 12 of the 230 samples. Expression of bax and the bax/bcl-2 ratio showed an inverse correlation to histological grade (p=0.0003 and 0.0001, respectively). There was also an inverse correlation between extent of apoptosis and immunoreactivity of p53 (p=0.0001) and bcl-2 (p=0.0001). A significant positive correlation between expression of the bax protein and apoptosis was evident (p=0.0001). HPV infection significantly correlated to the extent of histological abnormality (p=0.0001). High risk HPV-E6 protein also showed this significant correlation (p=0.0002). There was an inverse correlation between apoptosis and HPV infection (p=0.0002). High risk HPV infection was associated with decreased apoptosis and also increased human cell proliferation. Lowest levels of bax/bcl-2 ratio was also associated with HPV 16 and 18 infection (p=0.0001). Modulation of apoptosis and apoptotic regulatory proteins by high risk HPV infection may be an important factor in the development of cervical cancer. C1 [Nair, Prapid] Regional Cancer Centre, Division of Laboratory Medicine, 695011 Thiruvananthapuram, Kerala State, India. [Nair, M Krishnan] Regional Cancer Centre, Division of Radiation OncologyThiruvananthapuram, India. [Jayaprakash, G Puthuveetil] Regional Cancer Centre, Division of Radiation OncologyThiruvananthapuram, India. [Pillai, M Radhakrishna] Regional Cancer Centre, Division of Laboratory Medicine, 695011 Thiruvananthapuram, Kerala State, India. RP Pillai, MR (reprint author), Regional Cancer Centre, Division of Laboratory Medicine, 695011 Thiruvananthapuram, India. EM mrpillaie@vsnl.com CR Bosch FX, Manos MM, Munoz N, et al: Prevalence of human papillomavirus in cervical cancer: a worldwide perspective. International Biological Study on Cervical Cancer, IBSCC, Study Group. J Natl Cancer Inst 87:796-802, 1995. Cotter TG, Samali A: Oncogenes, apoptosis and cancer. Forum Trends Exp Clin Med 7:4-17, 1997. Furuya H, Yaboshita H, Noguchi M, et al: Apoptosis and cell growth fraction in normai, dysplastic and neoplastic squamous epithelium of uterine cervix. Nippon Rinsha, 54:1916-1921, 1996. Guchelaar HJ, Vermes A, Vermes I, et al: Apoptosis: Molecular mechanisms and implications for cancer chemotherapy. Pharmacy World and Science, 19:119-125, 1997. Isacson C, Kessis TD, Hedrick L, et al: Both cell proliferation and apoptosis increase with lesion grade in cervical neoplasia but do not correlate with human papillomavirus type. Cancer Res 56:669-674, 1996. Jayant K, Rao RS, Nene BM, et al: Improved stage at diagnosis of cervical cancer with increased cancer awareness in a rural Indian population. Int J Cancer 63:161-163, 1995. Kagawa S, Fujiwara T, Hizuta A, et al: P53 expression overcomes p21, WAF1/ CIP1)-mediated G-1 arrest. Oncogene 15:1903-1909, 1997. Kerr JFR, Winterford CM, Harmon BV: Apoptosis: Its significance in cancer and cancer therapy. Cancer 73:2013-2026, 1994. Kesari AL, Chellam VG, Nair PP, et al: Expression of the antiapoptotic protein bcl-2 is not dependent on the tumor suppressor protein p53 in Indian breast carcinoma. Pathobiol 65:108- 112, 1997. Kurvinen K, Tervahauta A, Syrjanen S, et al: The state of the p53 gene in human papillomavirus, HPV)- positive and HPVnegative genital precancer lesions and carcinomas as determined by single-strand conformation polymorphism analysis and sequencing. Anticancer Res 14:177-181, 1994. Lakshmi S, Nair BS, Jayaprakash PG, et al: P53 protein and tumorigenesis in the uterine cervix. Gen Diag Pathol 142:281- 287, 1997. Lane DP: p53, guardian of the genome. Nature 358:15-16, 1992. Lechner MS, Mack DH, Finicle AB, et al: Human papillomavirus E6 proteins bind p53 in vivo and abrogate p53-mediated repression of transcription. EMBO J 11:3045-3052, 1992. Liang XH, Volkmann M, Klein R, et al: Co-localization of the tumor suppressor protein p53 and human papillomavirus E6 protein in human cervical carcinoma cell lines. Oncogene 8:2645-2652, 1993. Matlashewski G: Human papillomavirus update, 15th Annual Intemational Papillomavirus Workshop, Gold Coast Australia, 1996. Mietz JA, Unger T, Huibregtse JM, et al: The transcriptional transactivation function of wild type p53 is inhibited by SV40 large T-antigen and by HPV-16 E6 oncoprotein. EMBO J 11:5013-5020, 1992. Miyashita T, Krajewski S, Krajewska M, et al: Tumor suppressor p53 is a regulator of bcl-2 and bax gene expression in vitro and in vivo. Oncogene 9:1799-1805, 1994. Nelson WG, Kastan MB: DNA strand breaks: The DNA template alterations that trigger p53-dependent DNA damage response pathways. Mol Cell Biol 14:1815-1823, 1994. Pan H, Yin C, Van-Dyke T: Apoptosis and cancer mechanisms. Cancer Surveys 29:305-327, 1997. Picksley SM, Lane DP: p53 and Rb, their cellular roles. Curr Opin Cell Biol 6:853-858, 1994. Pillai MR, Kesari AL, Chellam VG, et al: Spontaneous programmed cell death in infiltrating duct carcinoma: association with p53, bcl-2, hormone receptors and tumor proliferation. Pathology Res Pract, in press). 1998. Pillai R, Halabi S, McKalip A, et al: The Presence of Human Papillomavirus 16/18 E6, p53 and Bcl-2 protein in cervicovagina, smears from patients with invasive cervical cancer. Cancer Epidemiol, Biomark Prevent, 5:329-335, 1996. Ravi D, Nalinakumari KR, Rajaram RS, et al. Expression of programmed cell death regulatory p53 and bcl-2 proteins in oral lesions. Cancer Letters 105:139-146, 1996. Ravi D, Ramdas K, Mathew BS, et al: Angiogenesis during tumor progression in the oral cavity is related to reduced apoptosis and high tumor cell proliferation. Eur J Cancer Part I I, Oral Oncol, in press). 1998. Reihsaus E, Kohler M, Kraiss S, et al: Regulation of the level of the oncoprotein p53 in non-transformed and transformed cells. Oncogene 5:137-145, 1990. Sanchez-Perez AM, Soriano S, Clarke AR, et al: Disruption of the human papillomavirus type 16 E2 gene protects cervica! carcinoma cells from E2F-induced apoptosis. J General Virol 78:3009-3018, 1997. Sheets E, Crum CP, Yeh J: Association between cervical neoplasia and apoptosis as detected by in situ nuclear labelling. Gynecol Oncol, 63:94-100, 1996. Shibata DK, Arnheim N, Martin WJ: Detection of human papilloma virus in paraffin embedded tissue using the polymerase chain reaction. J Exp Med 167:225-230, 1988. Shoji Y, Saegusa M, Takano Y, et al: Correlation of apoptosis with tumor cell differentiation, progression and HPV infection in cervical carcinoma. J Clin Pathol 49:134-138, 1996. Sidransky D, Hollstein M: Clinical implications of the p53 gene. Annu Rev Med 47:285-301, 1996. Srinivas G, Kusumakumary P, Nair MK, et al: BCL-2 protein and apoptosis in paediatric acute lymphoblastic leukaemia. Internat J Mol Med, in press), 1998. Takahashi R: Role of p53 tumor suppressor gene and Fas I Apo- 1 in induction of apoptosis and differentiation of cancer cells. Leukemia 11:331-333, 1997. Ting Y, Manos MM: Detection and typing of genital human papillomavirus, in PCR protocols: A Guide to Methods and Applications, New York, Academic Press, pp 356-367, 1990. Wang TT: Differential effects of chemotherapeutic agents on bcl-2/bax apoptosis pathway in breast cancer cell. Proc Annu Meet Am Assoc Cancer Res 38:A769, 1997. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 95 EP 103 PG 9 ER PT J AU Dursun, A Poyraz, A Celik, B Akyol, G AF Dursun, Ayse Poyraz, Aylar Celik, Betul Akyol, Gulen TI Expression of c-erbB-2 Oncoprotein in Gastric Carcinoma: Correlation with Histopathologic Characteristics and Analysis of Ki-67 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE c-erbB-2; Ki-67; gastric carcinomas ID c-erbB-2; Ki-67; gastric carcinomas AB Amplification and overexpression of the c-erbB-2 gene has been demonstrated in several tumors and thought to be important determinants of biologic behaviors of carcinomas. In this study, correlation between c-erbB-2 expression und histopathologic parameters, including proliferative activity of gastric carcinomas was evaluated. Paraffin-embedded tissue sections from 62 patients who underwent curative resection of gastric carcinoma were analyzed immunohistochemically for the expression of c-erbB-2 and Ki-67. Strong membrane staining for c-erbB-2 was detected in 11 of 62 gastric carcinomas (17,7%) and no positive reaction was evident in noncancerous tissue. The incidence of c-erbB-2 pos- itivity in intestinal type carcinomas (24,3%) washigher than that of diffuse type carcinomas (4,76%). Positive staining for c-erbB-2 was present in one of the 9 (11,1%) early gastric carcinomas and 10 of 53 (18,8%) advanced gastric carcinomas. However, no statistically significant relationships were found between c-erbB-2 expression and histopathologic type, depth on invasion, the tumor size or lymph node metastases. Among the metastatic lymph nodes, 3 were positively stained with c-erbB-2 whereas the primary tumors of two cases had been found to be negative. Additionally, no correlation was found between c-erbB-2 reactivity and proliferative activity of carcinoma cells. The results suggest that expression of c-erbB-2 protein may occur selectively in intestinal type of gastric carcinomas. However, c-erbB-2 expression is not a reliable marker of malignant potential in gastric carcinomas. C1 [Dursun, Ayse] Gazi University Faculty of Medicine, Department of Pathology, Turgut Reis Caddesi 16/8, Mebusevleri Tandogan, 06580 Ankara, Turkey. [Poyraz, Aylar] Gazi University Faculty of Medicine, Department of Pathology, Turgut Reis Caddesi 16/8, Mebusevleri Tandogan, 06580 Ankara, Turkey. [Celik, Betul] Gazi University Faculty of Medicine, Department of Pathology, Turgut Reis Caddesi 16/8, Mebusevleri Tandogan, 06580 Ankara, Turkey. [Akyol, Gulen] Gazi University Faculty of Medicine, Department of Pathology, Turgut Reis Caddesi 16/8, Mebusevleri Tandogan, 06580 Ankara, Turkey. RP Dursun, A (reprint author), Gazi University Faculty of Medicine, Department of Pathology, 06580 Ankara, Turkey. CR Chariyalertsak S, Sugano K, Ohkura H, et al: Comparison of c-erbB-2 oncoprotein expression in tissue and serum of patients with stomach cancer. Tumor Biol 15:294-303, 1994. Chen BF, Marrogi AJ, Freeman SM, et al: Gastric carcinoma recept issues in prognostic factors. J Lou State Med Soc 147:138-145, 1995. Falck VG, Gullick WJ: C-erbB-2 oncogene product staning in gastric adenocarcinoma: An immunohistochemical study. J. Pathol 159:107-111, 1989. Flejou JF, Paraf F, Muzeau F, et al: Expression of c erb B-2 oncogene product in Barrett’s adenocarcinoma: Pathological and prognostic correlations. J Clin Pathol 47:23-26, 1994. Kameda T, Yasui W, Yoshida K, et al: Expression of ERBB2 in human gastric carcinomas: relationship between p185ERBB2 expression and the gene amplification. Cancer Res 50:8002- 8009, 1990. Lauren P: The two histological main types of gastric carcinoma: diffuse and so-called intestinal type carcinoma. Acta Pathol Microbiol Scand 64:31-49, 1965. Lee EY, Cibull ML, Strodel WE, et al: Expression of HER-2/neu oncoprotein and epidermal growth factor receptor and prognosis in gastric carcinoma. Arch Patol Lab Med 118:235-239, 1994. Lemoine NR, Jain S, Silvestre F, et al: Amplification and overexpression of the EGF receptor and c-erbB-2 proto-oncogenes in human stomach cancer. Br J Cancer 64:79-83, 1991. Mizutani T, Onda M, Tokunaga A, et al: Relationship of c-erbB- 2 protein expression and gene amplification to invasion and metastasis in human gastric cancer. Cancer 72:2083-2088, 1993. Mori S, Akiyama T, Miorishita Y, et al: Light and electron microscopical demonstration of c-erbB-2 gene product like immunoreactivity in human malignant tumors. Virchows Arch B Cell Pathol 54:8-15, 1987. Motojima K, Furui J, Kohara N, et al: erb-2 expression in welldifferentiated adenocarcinoma of the stomach predicts shorter survival after curative resection. Surgery 11S: 349-354, 1994. Ohguri T, Sato Y, Koizumi W, et al: An immunohistochemical study of c erb B-2 protein in gastric carcinoma and lymph node metastasis: Is the c-erbB-2 protein really a prognostic indicator? Int J Cancer 53:75-79, 1993. Orita H, Maehara Y, Emi Y, et al: C-erbB-2 expression is predictive for lymphatic spread of clinical gastric carcinoma. Hepatogastroenterology 44:294-298, 1997. Tsugawa K, Fushida S, Yonemura Y: Amplification of the c-erbB-2 gene in gastric carcinoma: Correlation with survival. Oncology 50:418-425, 1993. Uchino S, Tsuda H, Maruyama K, et al: Overexpression of c-erbB-2 protein in gastric cancer. Cancer 72:3179-3184, 1993. Wright C, Agnus B, Nicholson S, et al: Expression of c-erbB-2 oncoprotein; a prognostic indicator in human breast cancer. Cancer Res. 49:2087-2090, 1989. Yamamoto T, Ikawa S, Akiyama T: Similarity of protein encoded by the human c-erbB-2 gene to epidermal growth factor receptor. Nature 319:230-234, 1986. Yonemura Y, Ninomiya I, Ohoyama S, et al: Expression of c-erbB-2 oncoprotein in gastric carcinoma. Cancer 67:2914-2918, 1991. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 104 EP 106 PG 3 ER PT J AU Karabelyos, Cs Dobozy, O Szalai, Cs Klenjanszki, K Varju, K Hadhazi, Kiss, Fulop, KA Madarasz, B Falus, A AF Karabelyos, Csaba Dobozy, Otto Szalai, Csaba Klenjanszki, Katalin Varju, Kornelia Hadhazi, Akos Kiss, Arpad Fulop, Kristof Andras Madarasz, Balint Falus, Andras TI Elevated Hepatic Glucocorticoid Receptor Expression During Liver Regeneration in Rats SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE glucocorticoid receptor; liver; regeneration; monoclonal antibody; mRNA ID glucocorticoid receptor; liver; regeneration; monoclonal antibody; mRNA AB In rats within the first week of partial hepatectomy reconstruction of the normal histological structure of the liver already starts. To approach the possible role of endogenous glucocorticoids in the process of regeneration we measured the changes in the expression of steroid glucocorticoid receptor gene after various regeneration intervals. After partial hepatectomy, between 0.5–168 hours from the surgery, the gene expression (mRNA) of glucocorticoid receptor was determined by reverse transcription followed by PCR and normalized to that of glycerolphoshate dehydrogenase. Two peaks of glucocorticoid receptor mRNA were detected first, between 3 and 6 hours (first peak) and a second between 24 and 36 hours. Immunoreactive glucocorticoid receptor was detected by immunohistochemistry using monoclonal anti-glucocorticoid receptor. Three days after the surgery immunohistochemical studies showed substantially more immunoreactive GcR protein in the regenerated liver than in the controls. These semiquantitative data provide evidence suggesting elevation of glucocorticoid receptor expression during regeneration of liver at mRNA and protein levels. C1 [Karabelyos, Csaba] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4.Budapest, Hungary. [Dobozy, Otto] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4.Budapest, Hungary. [Szalai, Csaba] Heim Pal Children's HospitalBudapest, Hungary. [Klenjanszki, Katalin] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4.Budapest, Hungary. [Varju, Kornelia] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4.Budapest, Hungary. [Hadhazi, Akos] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4.Budapest, Hungary. [Kiss, Arpad] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4.Budapest, Hungary. [Fulop, Kristof Andras] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4.Budapest, Hungary. [Madarasz, Balint] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4.Budapest, Hungary. [Falus, Andras] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4.Budapest, Hungary. RP Falus, A (reprint author), Semmelweis University, Department of Genetics, Cell and Immunobiology, Budapest, Hungary. EM faland@net.sote.hu CR Apostolakos MJ, Schuermann WH, Frampton MW, et al: Measurement of gene expression by multiplex competitive polymerase chain reaction. Anal Biochem 213:277-284, 1993. Berki T, Kumanovics G, Kumanovics A, et al: Production and flow cytometric application of a monoclonal anti-glucocorticoid receptor antibody. J Immun Meth 214:19-27, 1998. Chomczynski P, Sacchi N: Single-step method of RNA isolation by acid guanidium thyocyanate-phenol-chloroform extraction. Anal Biochem 162:156-159, 1987. Cressman DE, Greenbaum LE, DeAngelis RA, et al: Liver failure and defective hepatocyte regeneration in interleukin-6-deficient mice. Science 274:1379-1383, 1996. Dobozy O, Csaba G, Inczefi-Gonda A, et al: Impact of a single insulin treatment, imprinting, applied during liver regeneration on hepatic insulin receptor development, blood glucose level and liver function parameters in adult rats. Acta Phys Hung 79:331-338, 1992. Golding M, Sarraf CE, Lalani EN, et al: Oval cell differentiation into hepatocytes in the acetylaminofluorene-treated regenerating rat liver. Hepatology 4 Pt 1:1243-1253, 1995. Higgins GM, Anderson RM. Experimental pathology of the liver: I. Restoration of the liver of the white rat following partial surgical removal. Arch Pathol, 12:186-202, 1931. Iimuro Y, Nishiura T, Hellerbrand C, et al: NFkappaB prevents apoptosis and liver dysfunction during liver regeneration. J Clin Invest 101:802-811, 1998. Lau EC, Li ZQ, Santos V, et al: Messenger RNA phenotyping for semi-quantitative comparison of glucocorticoid receptor transcript levels in the developing embryonic mouse palate. J Steroid Biochem Mol Biol 46:751-758, 1993. Okamoto K, Isohashi F, Tsukanaka K, et al: Alteration in the inducibility by dexamethasone of the activity of tyrosine aminotransferase and tryptophan oxygenase in regenerating rat liver after partial hepatectomy. Endocrinology 112:336-340, 1983. Snyers L, De Wit L, Content J: Glucocorticoid up-regulation of high affinity interleukin-6 receptors on human epithelial cells. Proc Natl Acad Sci USA 87:2838-2842, 1991. Stocklin E, Wissler M, Gouilleux F, et al: Functional interactions between Stat5 and the glucocorticoid receptor. Nature 383:726- 728, 1996. Rakasz E, Gal A, Biro J, et al: Modulation of glucocorticosteroid binding in human lymphoid, monocytoid and hepatoma cell lines by inflammatory cytokines interleukin, IL)-1, IL-6 and tumor necrosis factor, TNF, Scand. J.Immunology, 37:684-689, 1993. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 107 EP 109 PG 3 ER PT J AU Pacor, S Gagliardi, R Spessotto, P Zabucchi, G Sava, G AF Pacor, Sabrina Gagliardi, Renato Spessotto, Paola Zabucchi, Giuliano Sava, Gianni TI Paracrine Effects of IL-4 Transfection on TS/A Adenocarcinoma Cells Mediate Reduced In Vivo Growth SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article AB The in vitro/in vivo growth capacity and phenotype of TS/A and the IL4-transfected TS/A-IL4 cell lines were studied by cell cycle analysis, expression of ICAM-1/CD54, transferrin receptor/CD71 and E-cadherin and by histology of the primary tumors. TS/A-IL4, unlike the TS/A line, shows in vitro a marked increase in the fibroblastoid cell type and a decreased E-cadherin expression. Administration of conditioned medium containing IL4 obtained from the TS/A-IL4 cell line, stimulates CD54 expression in the TS/A cell line. TS/A-IL4 tumors grow more slowly in vivo and are ultimately rejected. These processes are accompanied by a marked increase in collagen and extracellular matrix proteins and increased recruitment and degranulation of mast cells. The paracrine effect of IL4, released by the transfected tumor cells, might be responsible for the reduced in vivo growth of the TS/A cell line in the presence of TS/A-IL4 cells. C1 [Pacor, Sabrina] University of Trieste, Department of Biomedical Sciences, via L. Giorgieri 7-9, 34127 Trieste, Italy. [Gagliardi, Renato] Institutes of Biological Research, Callerio FoundationTrieste, Italy. [Spessotto, Paola] Institutes of Biological Research, Callerio FoundationTrieste, Italy. [Zabucchi, Giuliano] University of Trieste, Department of Physiology and PathologyTrieste, Italy. [Sava, Gianni] University of Trieste, Department of Biomedical Sciences, via L. Giorgieri 7-9, 34127 Trieste, Italy. RP Sava, G (reprint author), University of Trieste, Department of Biomedical Sciences, 34127 Trieste, Italy. EM gsava@fc.univ.trieste.it CR Nanni P, De Giovanni C, Lollini PL et al: TS/A: a new metastasizing cell line from BALB/c spontaneous mammary adenocarcinoma. Clin Exp Metastasis 1:373-380, 1983. Pericle F, Giovarelli M, Colombo MP et al: An efficient Th2- type memory follows CD8+ lymphocyte-driven and eosinophil- mediated rejection of a spontaneous mouse mammary adenocarcinoma engineered to release IL4. J Immunol 153:5659- 5673, 1994. Allione A, Consalvo M, Nanni P et al: Immunizing and curative potential of replicating and nonreplicating murine mammary adenocarcinoma cells engineered with interleukin, IL)-2, IL-4, IL-6, IL-7, IL-10, Tumor Necrosis Factor a, Granulocyte-Macrophage Colony-stimulating Factor, and g-Interferon Gene or admixed with conventional adjuvants. Cancer Res 54:6022- 6026, 1994. Colombo MP, Forni G: Immunotherapy I: Cyclosine gene transfer strategies. Cancer Met Rev 15:317-328, 1996. Tepper RI, Pattengale PK, Leder P: Murine interleukin-4 displays potent anti-tumor activity in vivo. Cell 57:503-512, 1989. Tepper RI, Coffman RL, Leder P: An eosinophil-dependent mechanism for the antitumor effect of interleukin-4. Science 257:548-551, 1992. Sosman JA, Bartemes K, Offord KP et al: Evidence for eosinophil activation in cancer patients receiving recombinant interleukin- 4: effects of interleukin-4 alone and following interleukin- 2 administration. Clin Cancer Res 1:805-812, 1995. Brown MA, Hural J: Functions of IL-4 and control of its expression. Crit Rev Immunol 17:1-32, 1997. Paul WE, Ohara J: B-cell stimulatory factor/interleukin 4. Annu Rev Immunol 5:429-435, 1987. Sempowski GD, Derdak S, Phipps RP: Interleukin-4 and interferon- discordantly regulate collagen biosynthesis by functionally distinct lung fibroblast subsets. J Cell Physiol 167:290-296, 1996. Stein M, Keshav S, Harris N, Gordon S: Interleukin 4 potently enhances murine macrophage mannose receptor activity: a marker of alternative immunologic macrophage activation. J Exp Med 176:287-292, 1992. Tiggelman AM, Boers W, Linthorst C, et al: Collagen synthesis by human liver, myo)fibroblasts in culture: evidence for a regulatory role of IL-1 beta, IL-4, TGFbeta and IFN gamma. J Hepatol 23:307-317, 1995. Postlethwaite AE, Holnes MA, Katai H, et al: Human fibroblasts synthesize elevated levels of extracellular matrix proteins in response to interleukin 4. J Clin Invest 90:1479-1485, 1992. Obiri NJ, Tandon N, Puri RK: Up-regulation of intercellular adhesion molecule 1, ICAM-1, on human renal cell carcinoma cells by interleukin-4. Int J Cancer 61:635-642, 1995. Dustin MJ, Rothlein R, Bhan AK et al: Induction by IL-1 and interferon-g: tissue distribution, biochemistry and function of a natural adherence molecule, ICAM-1). J Immunol 137:245- 254, 1986. DHHS, Guide for the care and use of laboratory animals DHHS publ., NIH Bethesda, MD, pp. 23-86, 1985. Mc Bride WH, Economou JS, Syljuasen RG et al: The effects of cytokine gene transfer into tumors on host cell infiltration and regression. Anticancer Res; 16:1139-1144, 1996. Kruger-Krasagakes S, Li W, Richter G et al: Eosinophils infiltrating interleukin-5 gene-transfected tumors do not suppress tumor growth. Eur J Immunol 23:992-995, 1993. Noffz G, Quin Z, Kopf M, et al: Neutrophils but not eosinophils are involved in growth suppression of IL-4 secreting tumors. J Immunol 160:345-350, 1998. Chong AS, Boussy IA, Jiang XL et al: CD54/ICAM-1 is a costimulator of NK cell-mediated cytotoxicity. Cell Immunol 157:92-105, 1994. Natali P, Nicotra MR, Cavaliere R, et al: Differential expression of intercellular adhesion molecule 1 in primary and metastatic melanoma lesions. Cancer Res 50:1271-1278, 1990. Tomita Y, Nishyiama T, Watanabe H, et al: Expression of intercellular adhesion molecule 1, ICAM-1, on renal cell cancer: possible significance in host immune responses. Int J Cancer 46:1001-1006, 1990. Schardt C, Heymanns J, Schardt C, et al: Differential expression of the intercellular adhesion molecule 1, ICAM-1, in lung cancer cell lines of various histological types. Eur J Cancer 29A:2250-2255, 1993. Steinbach F, Alexander J, Tanabe K, et al: Expression of cell adhesion molecules in an established and characterized new human renal cell cancer line, CCF-RC7. Urol Res 23:175-183, 1995. Johnson JP, Stade BG, Holzmann B, et al: De novo expression of intercellular-adhesion molecule 1 in melanoma correlates with increased risk of metastasis. Proc Natl Acad Sci USA 86:641-644, 1989. Huang YW, Richardson J, Vitetta ES: Anti-CD54, ICAM-1, has antitumor activity in SCID mice with human myeloma cells. Cancer Res 55:610-616, 1995. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 110 EP 116 PG 7 ER PT J AU Tompa, A Major, J Jakab, GM AF Tompa, Anna Major, Jeno Jakab, G Matyas TI Is Breast Cancer Cluster Influenced by Environmental and Occupational Factors Among Hospital Nurses in Hungary? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ethylene oxide; exchange aberrations; genotoxicology monitoring; ionizing radiation; risk assessment ID ethylene oxide; exchange aberrations; genotoxicology monitoring; ionizing radiation; risk assessment AB An unusual cluster of 8 breast cancer and 8 other malignant tumor cases (ovarian, uterus, lung, colon and brain tumors and malignant melanoma) developed in a period of 12 years among 98 nurses exposed to ethylene oxide (EtOx) for 5–15 years in a unit using gas sterilizer in a hospital of the archiepiscopal city of Eger, Hungary. EtOx concentration in air samples of the working area varied from 5 to 150 mg/m3. The question was, if there was any causal relationship between the elevated incidence of breast cancer and the EtOx exposure, the other possibility was, that this cluster appeared accidentally. EtOx is a human carcinogen, however, no increased breast cancer incidence in EtOx-exposed subjects was reported in the literature. We followed up for two consecutive years the 27 non cancer patients, EtOx-exposed nurses and 11 unexposed hospital controls with the aid of a multiple genotoxicology monitor including chromosomal aberration, sister-chromatide exchange, HPRT point mutation and DNA repair studies. The results were compared with data from 30 local historical controls, 48 historical controls from Budapest, 14 hospital controls and 9 EtOx exposed nurses from Budapest. Significantly high chromosome aberration yields (especially chromosome type exchanges) were alike detected in EtOx-exposed and the two other control groups in Eger. These results could not be interpreted as a consequence of EtOx exposure only, since in the EtOx-exposed group from Budapest, beside an increased total aberration frequency, the obtained exchange type aberration yields were as low as the historical controls. A plausible explanation can be the natural low dose radioactivity (222Rn) of the local tap-water due to a specific geological situation in Eger. The spontaneous breast cancer incidence in Hungary doubled in the last 10 years compared with the previous 20 years (1960–1980), especially in Eger. The appearence of the high breast cancer incidence in the hospital of Eger indicates the combined effect of EtOx and a more common local etiologic factor, such as the naturally radioactive tap-water. However, since the reported studies did not involve the investigation either of the genetic predisposition, or the effects of other possible environmental, occupational, and/or life style confounding factors, further studies (partly in progress) are necessary to clarify the importance of these factors. C1 [Tompa, Anna] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Kemiai Biztonsagi Intezet, Citogenetikai es Molekularis Toxikologiai Osztaly, H-1450 Budapest, Hungary. [Major, Jeno] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Kemiai Biztonsagi Intezet, Citogenetikai es Molekularis Toxikologiai Osztaly, H-1450 Budapest, Hungary. [Jakab, G Matyas] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Kemiai Biztonsagi Intezet, Citogenetikai es Molekularis Toxikologiai Osztaly, H-1450 Budapest, Hungary. RP Tompa, A (reprint author), Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Kemiai Biztonsagi Intezet, Citogenetikai es Molekularis Toxikologiai Osztaly, H-1450 Budapest, Hungary. EM okbi@elender.hu CR WHO: World Health Statistics Annuals 1984-88. UN World Health Organisation, Geneva. 1988. KSH: Demografiai Evkonyv 1970. Kozponti Statisztikai Hivatal, Budapest. 1972., in Hungarian). KSH: Demografiai Evkonyv 1990. Kozponti Statisztikai Hivatal, Budapest. 1992., in Hungarian). Wolff MS: Environmental exposures and breast cancer. In: Accomplishments in cancer research 1996., Eds: Fortner JG and Sharp PA), Lippincott – Raven Publ, Philadelphia-New York, 1997, pp. 136-144. Tokunaga M, Land CE, Tokuoka S, et al.: Incidence of female breast cancer among atomic bomb survivors, 1950-1985. Radiation Res 138:208-223, 1994. Tompa A, Major J, Jakab MG: Environmental genotoxicity and breast cancer risk in Hungary. Cancer Res. submitted for publication., 1999). Hagmar L, Brogger A, Hansteen I-L, et al.: Cancer risk in humans predicted by increased levels of chromosomal aberrations in lymphocytes: Nordic study group on the health risk of chromosome damage. Cancer Res 54:2919-2922, 1994. Bonassi S, Abbondandolo A, Camurri L, et al.: Are chromosome aberrations in circulating lymphocytes predictive of future cancer onset in humans? Preliminary results of an Italian cohort study. Cancer Genet Cytogenet 79:133-135, 1995. Brusick DJ: Value of short-term mutagenicity tests in human population monitoring. In: Chemical mutagenesis, human population monitoring and genetic risk assessment. Progress in Mutation Research, Vol 3., Eds: Bora KC, Douglas GR and Nestmann ER), Elsevier Biomedical Press, Amsterdam-Oxford- New York, 1982, pp. 261-274. Major J, Jakab MG, Tompa A: Genotoxicological investigation of hospital nurses occupationally exposed to ethylene-oxide: I. Chromosome aberrations, sister-chromatid exchanges, cell cycle kinetics, and UV-induced DNA synthesis in peripheral blood lymphocytes. Environ Molec Mutagen 27:84-92, 1996. Agyagasi D: Balneological issues at Eger. Balneologia Gyogyfurdugy Gyogyidegenforgalom 1:3-16. 1993., in Hungarian, abstract in English). IARC: Cancer: Causes, Occurrence and Control. Chapter 8. Radiation. In: IARC Scientific Publications No 100., Eds: Tomatis L, Aitio A, Day NE, Haseltine E, Kaldor J, Miller AB, Parkin DM, Riboli E), WHO, IARC, Lyon, 1990, pp. 157-158. Tompa A, Major J, Jakab MG: Monitoring of benzene-exposed workers for genotoxic effects of benzene: Improved-workingcondition- related decrease in the frequencies of chromosomal aberrations in peripheral blood lymphocytes. Mutation Res 304:159-165, 1994. Moorhead PS, Nowell PC, Mellman, et al.: Chromosome preparations of leukocytes cultured from human peripheral blood. Exp Cell Res 20:613-616, 1960. Perry P, Wolff S: New Giemsa method for the differential staining of sister chromatids. Nature 258:121-125, 1974. Carrano AV, Natarajan AT: Considerations for population monitoring using cytogenetic techniques. ICPEMC Publ. No. 14. Mutation Res 204:379-406, 1988. Muller E, Bertok A: Az egri korhazban elfordult daganatos megbetegedesek - higienes viszonyok, konzekvenciak. Korhaz es Orvostechnika 33:17-22, 1995., in Hungarian). Lerda D, Rizzi R: Cytogenetic studies of persons occupationally exposed to ethylene oxide. Mutation Res 281:31-37, 1992. Major J, Jakab MG, Tompa A: Genotoxicological investigation of hospital nurses occupationally exposed to ethylene-oxide: II. Significance of environmental radon exposure in development of inceased exchange aberration frequencies obtained. Environ Molec Mutagen submitted for publication. 1999. KSH: Demografiai Evkonyv 1996. Kozponti Statisztikai Hivatal, Budapest. 1998., in Hungarian). Gombkoto G: Tumor morbidity and mortality among people living in settlements in the vicinity of town Eger. Egeszsegtudomany 39:154-171, 1995., in Hungarian, abstract in English) Gombkoto G, Szeremi M: A Heves megyei lakossag megbetegedesi – halalozasi viszonyainak elemzese nemzetkozi es magyarorszagi osszehasonlitasban. NEKAP, Eger, 1997., in Hungarian). Kertesz M: Data of the Air Quality Monitoring Network IV. 1996 – IX. 1996. Egeszsegtudomany 41:78-95, 1997., in Hungarian, abstract in English). Norman SA, Berlin JA, Soper KA, et al.: Cancer incidence in a group of workers potentially exposed to ethylene oxide. Int J Epidemiol 24:276-284, 1995. IARC Monographs on the Evaluation of the Carcinogenic Risk of Chemicals to Humans. Vol. 36. Allyl compounds, Aldehydes, Epoxides and Peroxides. Ethylene oxide. IARC, Lyon. 1988. pp. 198-226. Bojtor I, Gidali J, Feher I, et al.: Comparison of hematological parameters between “normal” population and residents consuming tap water of Ra content. Egeszsgtudomany 24:223-235, 1980., in Hungarian, abstract in English). Koteles G, Nikl I, Szerbin P, et al.: Az egri egeszsegugyi problemak sugaregeszsegugyi vonatkozasai. Korhaz- es Orvostechnika 33:92-97, 1995., In Hungarian). WHO: World Health Statistics Annuals 1984-88. UN World Health Organisation, Geneva. 1988. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 117 EP 121 PG 5 ER PT J AU Ozguroglu, M Ersavasti, EG Demir, G Aki, H Demirelli, F Kanberoglu, K Mandel, N Buyukunal, E Serdengecti, S Berkarda, B AF Ozguroglu, Mustafa Ersavasti, Esen Gul Demir, Gokhan Aki, Hilal Demirelli, Fuat Kanberoglu, Kaya Mandel, Nil Buyukunal, Evin Serdengecti, Suheyla Berkarda, Bulent TI Magnetic Resonance Imaging of Bone Marrow Versus Bone Marrow Biopsy in Malignant Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE malignant lymphoma; bone marrow biopsy; magnetic resonance imaging ID malignant lymphoma; bone marrow biopsy; magnetic resonance imaging AB Bone marrow involvement is a frequent finding in malignant lymphoma. Bone marrow biopsy of the posterior iliac crest is routinely performed for staging. Abnormal magnetic resonance imaging (MRI) signals of bone marrow was also reported to be indicative of bone marrow involvement. This study included 60 patients with malignant lymphoma. Unilateral bone marrow biopsy of the posterior iliac crest was performed. MRI of lumbar spine was studied within 24 hours of bone marrow biopsy. 22 healthy controls were used for the detection of MRI objectivity during visual evaluation. In 83% of patients (50/60), biopsy and MRI results agreed completely. In two patients, histologic sections failed to show any evidence of bone marrow involvement despite abnormal MRI signals suggestive of involvement. In three patients, MRI was completely normal despite biopsy proven bone marrow infiltration. False negativity (3/60) and false positivity (2/60) rates were very low. Negative biopsy findings with positive or equivocal MRI results should not exclude bone marrow involvement and needs further evaluation with bilateral or guided biopsy. Thus, we conclude that MRI of bone marrow is a fairly sensitive, noninvasive modality and might be of potential value in detecting bone marrow infiltration in malignant lymphoid neoplasms which can be utilized as a useful adjunct to standard staging procedures. C1 [Ozguroglu, Mustafa] Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, Yogurtcu Basi Sokagi, Akcira apt. 20/3ˆ, 81030 Istanbul, Dalyan-Fenerbahce, Turkey. [Ersavasti, Esen Gul] Cerrahpasa Medical School, Istanbul University, Department of RadiologyIstanbul, Turkey. [Demir, Gokhan] Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, Yogurtcu Basi Sokagi, Akcira apt. 20/3ˆ, 81030 Istanbul, Dalyan-Fenerbahce, Turkey. [Aki, Hilal] Istanbul Medical Faculty, Department of PathologyIstanbul, Turkey. [Demirelli, Fuat] Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, Yogurtcu Basi Sokagi, Akcira apt. 20/3ˆ, 81030 Istanbul, Dalyan-Fenerbahce, Turkey. [Kanberoglu, Kaya] Cerrahpasa Medical School, Istanbul University, Department of RadiologyIstanbul, Turkey. [Mandel, Nil] Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, Yogurtcu Basi Sokagi, Akcira apt. 20/3ˆ, 81030 Istanbul, Dalyan-Fenerbahce, Turkey. [Buyukunal, Evin] Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, Yogurtcu Basi Sokagi, Akcira apt. 20/3ˆ, 81030 Istanbul, Dalyan-Fenerbahce, Turkey. [Serdengecti, Suheyla] Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, Yogurtcu Basi Sokagi, Akcira apt. 20/3ˆ, 81030 Istanbul, Dalyan-Fenerbahce, Turkey. [Berkarda, Bulent] Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, Yogurtcu Basi Sokagi, Akcira apt. 20/3ˆ, 81030 Istanbul, Dalyan-Fenerbahce, Turkey. RP Ozguroglu, M (reprint author), Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, 81030 Istanbul, Turkey. EM ozguroglu@superonline.com CR Shipp MA, Mauch PM, Harris NL: Non-Hodgkin’s Lymphomas. In: DeVita VT Jr, Hellman S, Rosenberg SA, eds. Cancer, Principles and Practice of Oncology. 5th ed. Lippincott-Raven, Philadelphia, 1997:2165-2220. DeVita VT Jr, Mauch PM, Harris NL: Hodgkin’s Disease. In: De Vita VT Jr, Hellman S, Rosenberg SA, eds. Cancer, Principles and Practice of Oncology. 5th ed. Lippincott-Raven, Philadelphia, 1997:2242-2283. Bartl R, Frisch B, Burkhardt R, et al: Lymphoproliferations in the bone marrow: identification and evolution, classification and staging. J Clin Pathol 37:233-254, 1984. Burkhardt R, Frisch B, Bartl R: Bone marrow biopsy in hematologic disorders. J Clin Pathol 8: 257-284, 1982. Westerman MP: Bone marrow needle biopsy: an evaluation and critique. Semin Hematol 18:293-300, 1981. Coller BS, Chabner BA, Gralnick HR: Frequencies and patterns of bone marrow involvement in non-Hodgkin lymphoma: observations on the value of bilateral biopsies. Am J Pathol 3:105-119, 1977. Brunning RD, Bloomfield CD, McKenna RW, et al: Bilateral trephine bone marrow biopsies in lypmhoma and other neoplastic diseases. Ann Intern Med 82:365-366, 1975. Rosenberg SA: Hodgkin’s disease of the bone marrow. Cancer Res 31:1733-1736, 1971. Datz FL, Taylor A: The clinical use of radionuclide bone marrow imaging. Semin Nucl Med 15:239-259, 1985. McAfee JG, Subramanian G, Aburano T, et al: A new formulation of Tc-99m minimicroaggregated albumin for marrow imaging: comparison with other colloids, In-111 and Fe-59. J Nucl Med 23:21-28, 1982. Naughton MJ, Hess JL, Zutter MM, et al: Bone marrow staging in patients with non-Hodgkin’s lymphoma. Is flow cytometry a useful test? Cancer 82:1154-1159, 1998. 12 Smith SR, Roberts N, Percy DF, et al: Detection of bone marrow abnormalities in patients with Hodgkin’s disease by T1 mapping of MR images of lumbar vertebral bone marrow. Br J Cancer 65:246-251, 1992. 13. Linden A, Zankovich R, Theissen P, et al: Schicha H. Malignant lymphoma: Bone marrow imaging versus biopsy. Radiology 173:335-339, 1989. 14. Daffner RH, Lupetin AR, Dash N, et al: MRI in the detection of malignant infiltration of bone marrow. Amer J Roentgenol 6;146:353-358, 1986. 15. Murphy WA, Totty WG: Musculoskeletal magnetic resonance imaging. Magn Reson Ann:1-35, 1986. 16. Vogler JB, Murphy WA: Bone marrow imaging. Radiology 168:679-693, 1988. 17. Steiner RM, Mitchell DG, Rao VM, et al: Magnetic resonance imaging of bone marrow, diagnostic value in diffuse hematologic disorders. Magnet. Reson. Quart. 6:17, 1990. 18. Moore SG, Gooding CA, Brasch RC, et al: Bone marrow in children with acute lymphocytic leukemia: MR relaxation times. Radiology 160:237-240, 1986. 19. Richards MA, Webb JAW, Jewell SE, et al: Low field strength magnetic resonance imaging of bone marrow in patients with malignant lymphoma. Br J Cancer 57:412, 1988. 20. Non-Hodgkin’s lymphoma pathologic classification project. National Cancer Institute sponsored study of classifications of non-Hodgkin’s lymphomas: summary and description of a Working Formulation for clinical usage. Cancer 49:2112, 1982. 21. Kapadia SB, Krause JR: Hodgkin’s disease. In: Krause JR, ed. Bone marrow biopsy, Churchill Livingstone, Edinburgh, 145- 155 1981. 22. Williams WJ, Nelson DA: Examination of the marrow. In: Williams WJ, Beutler E, Erslev AJ, Lichtman MA, eds. Hematology, 4th edition, McGraw Hill, USA, 24-31, 1991. 23. Fineberg S, Marsh E, Alfonso F, et al: Immunophenotypic evaluation of the bone marrow in non-Hodgkin’s lymphoma. Hum Pathol 24:636-642, 1993. 24. Christy M: Active bone marrow distribution as a function of age in humans. Phys Med Biol 26:389-400, 1981. 25. Trubowitz S, Davis S: The bone marrow matrix. In: Trubowitz S, Davis S, eds.The human marrow anatomy, physiology and pathophysiology. Vol 1. Boca raton: CRC, 43-76, 1982. 26. Brateman L. Chemical shift imaging: a review. Amer J Roentgenol 146:971-981, 1986. 27. McKinstry CS, Steiner RE, Young AT, et al: Bone marrow in leukemia and aplastic anemia: MR imaging before, during and after treatment. Radiology 162:701-707, 1987. 28. Rahmouni A, Divine M, Mathieu D, et al: MR appearance of multiple myeloma of the spine before and after treatment. Amer J Roentgenol 160:1053-1057, 1993. 29. Moulopoulos LA, Varma DGK, Dimopoulos MA, et al: Multiple myeloma: spinal mr imaging in patients with untreated newly diagnosed disease. Radiology 185:833-840, 1992. 30. Shields AF, Porter BA, Churchley S, et al: The detection of bone marrow involvement by lymphoma using magnetic resonance imaging. J Clin Oncol 5:225-230, 1987. 31. Rosen BR, Fleming DM, Kushner DC, et al: Hematologic bone marrow disorders: Quantitative chemical shift mr imaging. Radiology 169:799-804, 1988. 32. Sugimura K, Yamasaki K, Kitagaki H, et al: Bone marrow diseases of the spine: Differentiaiton with T1 and T2 relaxation times in MR imaging. Radiology 165:541-544, 1987. 33. Simon JH, Szumowski J: Proton, fat/water, chemical shift imaging in medical magnetic resonance imaging: current status. Invest Radiol 27:865-874, 1992. 34. Mirowitz SA, Apicella P, Reinus WR, et al: MR imaging of bone marrow lesions: Relative conspicuousness on T1-weighted, fat suppressed T2-weighted and STIR images. Amer J Roentgenol 162:215-221, 1994. 35. Staller DW, Porter BA, Steinkirchner TM: Marrow Imaging In: Staller DW. Magnetic Resonance Imaging in Orthopaedics and Sports Medicine. J.B. Lippincott Company, Philadelphia, 1996, 2nd ed. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 123 EP 128 PG 6 ER PT J AU Cserni, G AF Cserni, Gabor TI Estimating the Overlap Between Sentinel Lymph Nodes and Axillary Node Samples in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; axillary clearance; axillary sampling; staging ID breast cancer; axillary clearance; axillary sampling; staging AB Management of the axilla in breast cancer patients is a controversial issue. Axillary sampling and sentinel lymphadenectomy are both conservative surgical approaches which aim to stage the disease. These procedures target selective treatment of node-positive patients and seem to allow the omission of axillary clearance in node-negative ones. In this way, they reduce the rate of complications in an otherwise overtreated subset of patients. Forty consecutive patients with palpable T1 and T2 breast carcinoma underwent sentinel lymphadenectomy following mapping with Patent blue dye, with subsequent axillary clearance and excision of the tumor or mastectomy. Then the largest/firmest 3,4,5 and 6 nodes were selected from all the lymph nodes in order to model an axillary sample. It was suggested that these are the nodes that are the most likely to be included in the specimen during sampling, because of their size and consistency. The probability of the sentinel lymph nodes falling into the sample of the 3-6 largest/firmest nodes was calculated. The sentinel nodes predicted the axillary nodal status in 95%, while the samples of the largest 3, 4, 5 and 6 nodes were predictive in 95, 96, 98 and 98%, respectively. The two methods of evaluation displayed a considerable overlap, as the sentinel node would have been included in the 3–6 largest/firmest nodes in 79–92% of the cases, depending on the number of largest nodes evaluated. The overlap was greater after fine needle aspiration of the primary tumor. Although the two alternative staging procedures of 3, 4, 5 or 6 node sampling and sentinel lymphadenectomy with the vital blue dye technique cannot be simultaneously done without one influencing the other, and the first method was only modeled, the results suggest that there is a considerable overlap between the two; axillary sampling may often remove the sentinel lymph nodes. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., H-6000 Kecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.c3.hu CR Albertini JJ, Lyman GH, Cox C, et al: Lymphatic mapping and sentinel node biopsy in the patient with breast cancer. JAMA 276:1818-1822, 1996. American Joint Committee on Cancer: Breast. In: AJCC Cancer Staging Manual, Stn edition., Eds: Fleming ID, Cooper JS, Henson DE, et al), Lippincott Raven Publishers, Philadelphia, 1997, pp. 171-180. Blichert-Toft M, Smola MG, Cataliotti L, et al: Principles and guidelines for 4 surgeons – management of symptomatic breast cancer. On behalf of the European Society of Surgical Oncology. Eur J Surg Oncol 23:101-109, 1997. Borgstein P, Meijer S: Historical perspective of lymphatic tumour spread and the emergence of the sentinel node concept. Eur J Surg Oncol 24:85-89, 1998. Cady B: Use of primary breast carcinoma characteristics to predict lymph node metastases. Editorial. Cancer 79:1856-1861, 1997. Cady B, Stone MD, Schuler JG, et al: The new era in breast cancer: invasion, size and nodal involvement dramatically decreasing as a result of mammographic screening. Arch Surg 131:301-308, 1996. Cserni G: Changing trends in lymph node recovery from axillary clearance specimens in breast cancer: possible implications for the quantitative axillary status from a 17 year retrospective study. Eur J Oncol 2:403-408, 1997. Cserni G: Histopathologic basis for axillary sampling. Eur J Cancer 34(Suppl. 5):S 109, 1998. Davidson T: Why I favour axillary node clearance in the management of breast cancer. Eur J Surg Oncol 21:5-7, 1995. Davies GC, Millis RR, Hayward JL: Assessment of axillary lymph node status. Ann Surg 192:148-151, 1980. Fisher CJ, Boyle S, Burke M, et al: Intraoperative assessment of nodal status in the selection of patients with breast cancer for axillary clearance. Br J Surg 80:457-458, 1993. Forrest APM, Everington D, McDonald CC, et al: The Edinburgh randomized trial of axillary sampling or clearance after mastectomy. Br J Surg 82:1504-1508, 1995. Forrest APM, Roberts MM, Cant E, et al: Simple mastectomy and pectoral node biopsy. Br J Surg 63:569-575, 1976. Giuliano AE, Dale PS, Turner RR, et al: Improved axillary staging of breast cancer with sentinel lymphadenectomy. Ann Surg 180:700-704, 1995. Giuliano AE, Kirgan DM, Guenther JM, et al: Lymphatic mapping and sentinel lymphadenectomy for breast cancer. Ann Surg 220:391-401, 1994. Greenall MJ: Why I favour axillary node sampling in the management of breast cancer. Eur J Surg Oncol 21:2-5, 1995. Harris JR, Osteen RT: Patients with early breast cancer benefit from effective axillary treatment. Breast Cancer Res Treat 5:17- 21, 1985. Kingston RD, Bridger J, Smith CJ, et al: Axillary sampling and radiotherapy does not compromise the patient with early breast cancer: Trafford database experience. Eur J Surg Oncol 21:364- 367, 1995. Kinne DW: Controversies in primary breast cancer management. Am J Surg 166:502-508, 1993. Kiricuta CI, Tausch J: A mathematical model of axillary lymph node involvement based on 1446 complete axillary dissections in patients with breast carcinoma. Cancer 69:2496-2501, 1992. Kissin MW, Thompson PH, Price AB, et al: The inadequacy of axillary sampling in breast cancer. Lancet i:1210-1212, 1982. Kjaergaard J, Blichert-Toft M, Andersen JA, et al: Probability of false negative nodal staging in conjunction with partial axillary dissection in breast cancer. Br J Surg 72:365-367, 1985. Krag DN, Weaver DL, Alex JC, et al: Surgical resection and radiolocalization of sentinel lymph node in breast cancer using a gamma probe. Surg Oncol 2:335-340, 1993. Margolese RG: Surgery and Adjuvant Chemotherapy. In: Comprehensive Textbook of Oncology, 2nd edition., Eds: Moossa AR, Schimpff SC, Robson MC), Williams and Wilkins, Baltimore, 1991, pp.800-810. Mathiesen O, Carl J, Bonderup O, et al: Axillary sampling and the risk of erroneous staging of breast cancer. Acta Oncol 29:721-725, 1990. Moore MP, Kinne DW: Axillary lymphadenectomy: a diagnostic and therapeutic procedure. J Surg Oncol 66:2-6, 1997. Pigott J, Nichols MD, Maddox WA, et al: Metastases to the upper levels of axillary nodes in carcinoma of the breast and its implications for nodal sampling procedures. Surg Gynecol Obstet 158:255-259, 1984. Ridings P, Bucknall TE: Modern trends in breast cancer therapy: towards less lymphoedema? Eur J Surg Oncol 24:21-22, 1998. Statman R, Giuliano AE: The role of the sentinel lymph node in the management of patients with breast cancer. Adv Surg 30:209-221, 1997. Steele RJC, Forrest APM, Gibson T, et al: The efficacy of lower axillary sampling in obtaining lymph node status in breast cancer; a controlled randomised trial. Br J Surg 72:368-369, 1985. The Breast Surgeons Group of the British Association of Surgical Oncology: Guidelines for surgeons in the management of symptomatic breast disease in the United Kingdom. Eur J Surg Oncol 21(Suppl A):1-13, 1995. Turner RR, Ollila DW, Krasne DL, et al: Histopathologic validation of the sentinel lymph node hypothesis for breast carcinoma. Ann Surg 226:271-276, 1997. Veronesi U, Paganelli G, Galimberti V, et al: Sentinel-node biopsy to avoid axillary dissection in breast cancer with clinically negative lymph-nodes. Lancet 349:1864-1867, 1997. Veronesi U, Zurrida S, Galimberti V: Consequences of sentinel node in clinical decision making in breast cancer and prospects for future studies. Eur J Surg Oncol 24:93-95, 1998. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 129 EP 133 PG 5 ER PT J AU Sharma, S Karak, KA Singh, R Mehta, SV Sarkar, Ch Schmitt, PH AF Sharma, Suash Karak, K Asis Singh, Rajvir Mehta, Singh Veer Sarkar, Chitra Schmitt, P Horst TI A Correlative Study of Gliomas Using In Vivo Bromodeoxyuridine Labeling Index and Computer-aided Malignancy Grading SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gliomas; in vivo BrdU LI; malignancy classification; histologic grading; computer aided grading ID gliomas; in vivo BrdU LI; malignancy classification; histologic grading; computer aided grading AB An in vivo bromodeoxyuridine (BrdU) labeling index (LI) was estimated in 43 cases of astrocytic tumors and mixed gliomas by one hour intra-operative intravenous infusion at a dose of 200 mg/m2 and correlated with (a) histological grading using a computer aided malignancy classifier TESTAST-268; and (b) histological typing using WHO classification. The lowest BrdU LI was seen in pilocytic and gemistocytic astrocytomas followed by astrocytomas, anaplastic astrocytomas and glioblastoma multiforme in that order. Mixed oligoastrocytomas followed the pattern of their astrocytic counterparts. Tumors of similar histological type showed different BrdU LI values especially amongst astrocytomas and glioblastomas. A statistically significant difference in the BrdU LI was also noted between the higher TESTAST grades of astrocytomas (T III and IV) versus the lower TESTAST grades (T II). Unlike earlier reports in literature, in the present study the category of BrdU LI of <1 contained no case of anaplastic astrocytoma or glioblastoma multiforme (TESTAST grades III and IV). Likewise, the category of BrdU LI >5 contained only anaplastic astrocytoma and glioblastoma multiforme (TESTAST grades III and IV). Maximum spread of cases was seen in the BrdU LI category of 1-5, not only in terms of histological types but also TESTAST grades. Thus there appeared to be a positive trend of increasing BrdU LI values both with histological types and increasing TESTAST grades. Further, an interesting observation was that by using a combination of TESTAST grades and BrdU LI, the histologically homogenous glioblastoma group could be further subdivided into 4 categories which showed a trend towards prognostic correlation. Thus, this study though preliminary with number of cases being small in some groups, highlights the possible usefulness of combined histological typing, TESTAST grading and in vivo BrdU LI for prognostication of gliomas especially glioblastoma multiforme. C1 [Sharma, Suash] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Karak, K Asis] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Singh, Rajvir] All India Institute of Medical Sciences, Department of BiostatisticsNew Delhi, India. [Mehta, Singh Veer] All India Institute of Medical Sciences, Department of NeurosurgeryNew Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Schmitt, P Horst] University of Heidelberg, Department of NeuropathologyHeidelberg, Germany. RP Sarkar, Ch (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM sarkarch@medinst.ernet.in CR Burger PC, Scheithauer BW: Tumors of the central nervous system. Armed Forces Institute of Pathology, Washington, 1994. Burger PC, Vogel FS, Green SB, et al: Glioblastoma multiforme and anaplastic astrocytoma : Pathologic criteria and prognostic implications. Cancer 56:1106-1111, 1985. Daumos-Duport C, Scheithauer B, OFallen J, et al: Grading of astrocytomas: A simple and reproducible method. Cancer 62:2152-2165, 1988. Dinda AK, Kharbanda K, Sarkar C, et al: In-vivo proliferative potential of primary human brain tumors : Its correlation with histological classification and morphological features. I. Gliomas. Pathology 25:4-9, 1993. Dinda AK, Kharbanda K, Sarkar C, et al: In-vivo proliferative potential of primary human brain tumors: Its correlation with histological classification and morphological features. II. Nonglial Tumors. Pathology 25:10-14, 1993. Gilles FH, Leviton A, Hedley-Whyte ET, et al: Childhood braintumor update. Hum Pathol 14:834-845, 1983. Gol A: The relative benign astrocytomas of cerebrum: A clinical study of 194 verified cases. J Neurosurg 18:501-506, 1961. Hoshino T: A commentary on biology and growth kinetics of low grade and high grade gliomas. J Neurosurg 61:895-900, 1994. Hoshino T, Nagashima T, Murovic JA, et al: In situ cell kinetic studies on human neuroectodermal tumors with bromodeoxyuridine labeling. J Neurosurg 64:453-59, 1986. Hoshino T, Prados M, Wilson CB, et al: Prognostic implications of low bromodeoxyuridine labeling index of human gliomas. J Neurosurg 71:335-341, 1989. Hoshino T, Rodriguez LA, Cho KC, et al: Prognostic implications of the proliferative potential of low grade astrocytomas. J Neurosurg 69:839-1342, 1988. Hoshino T, Townsend JJ, Muraoka I, et al: An autoradiographic study of human gliomas : growth kinetics of anaplastic astrocytomas and glioblastoma multiforme. Brain 103:967-984, 1980. Hoshino T, Wilson CB: Review of basic concepts of cell kinetics as applied to brain tumors. J Neurosurg 42:123-131, 1975. Hoshino T, Wilson CB: Cell kinetic analysis of human malignant brain tumors, gliomas). Cancer 44:956-962, 1979. Kernohan JW, Mabon RF, Svien HJ, et al: A simplified classification of gliomas. Proc Staff Meet Mayo Clin 24:71-75, 1949. Kernohan JW, Sayre GP: Tumors of Central Nervous System. Fascicle 35, Atlas of Tumor Pathology. Armed Forces Institute of Pathology, Washington, p17-129, 1952. Kleihues P, Burger PC, Scheithauer BW: Histological typing of tumors of the central nervous system. International classification of tumors. 2nd ed. World Health Organisation. Springer Verlag, Berlin, 1993. Louis DN: A molecular genetic model of astrocytoma histopathology. Brain Pathol 7:755-764, 1997. Morimura T, Kitz K, Budka H: In situ cell kinetics in human brain tumors. A comparative immunocytochemical study of Sphase cells by a new in vitro bromodeoxyuridine labeling technique and of proliferating pool cells by monocolonal antibody Ki-67. Acta Neuropathol 77:276-282, 1989. Murovic JA, Nagashima T, Hoshino T, et al: Pediatric central nervous system tumors: A cell kinetic study with bromodeoxyuridine. Neurosurg 19:900-904, 1986. Nagashima T, DeArmond SJ, Murovic J, et al: Immunocytochemical demonstration of S-phase cells by anti-bromodeoxyuridine monoclonal antibody in human brain tissues. Acta Neuropathol 67:155-159, 1985. Nelson JS, Tuskada Y, Schoenfeld D, et al: Necrosis as a prognostic criteria in malignant supratentorial astrocytic gliomas. Cancer 52:550-554, 1983. Ringertz J: Grading of gliomas. Acta Pathol Microbiol Scand 27:51-64, 1950. Russell DS, Rubinstein LJ: Tumors of central neuroepithelial origin. In: Pathology of tumors of the Nervous System. Edward Arnold, London, 198; p 83-247. Scanlon PW, Taylor WF: Radiotherapy of intracranial astrocytomas. Analysis of 417 cases treated from 1960 through 1969. Cancer 5:301-307, 1979. Schmitt HP: Numerical classification of malignancy in astrocytomas: An attempt to improve and to validate the classifier TESTAST. Studien Zur Klassifikation. Indek Verlag, Frankfurt, p361-368, 1989. Schmitt HP, Oberwittler C: Computer aided classification of malignancy in astrocytomas : II. The value of categorically evaluating histologic and non-histologic features for a numerical classifier. Analyt Cell Pathol 4:409-419, 1992. Schmitt HP, Oberwittler C, Schad LR: Computer aided classification of malignancy in astrocytomas: I. The value of nuclear parameters obtained by automated black and white image analysis. Analyt Cell Pathol 4:397-407, 1992. Sharma S, Karak AK, Sarkar C, et al: A grading study of gliomas using histologic morphometry and malignancy classification. J Neuro-Oncol 27:75-85, 1996. Shapiro JR: Biology of gliomas: heterogenity, oncogenes, growth factors. Semin Oncol 133:4-15, 1986. Watanabe K, Tachibona O, Yonekawa Y, et al: Role of gemistocytes in astrocytoma progression. Lab Inv 76:277-284, 1977. Wier B: The relative significance of factors affecting post-operative survival in astrocytoma grade III and IV. J Neurosurg 38:448-452, 1973. Wier B, Grace M: The relative significance of factors affecting operative survival in astrocytomas, grades I and II. Can J Neurol Sci 33:47-50, 1976. Zulch KJ: Histological typing of tumors of central nervous system. International Histologic Classification of Tumors. World Health Organisation, Geneva, p14-50, 1979. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 134 EP 141 PG 8 ER PT J AU Moazzeni, MS Amirzargar, AA Shokri, F AF Moazzeni, M Seyed Amirzargar, Ali-Akbar Shokri, Fazel TI HLA Antigens in Iranian Patients with B-cell Chronic Lymphocytic Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HLA antigens; chronic lymphocytic leukemia ID HLA antigens; chronic lymphocytic leukemia AB The frequency of HLA class-I and class-II antigens was investigated in 32 Iranian patients with B-cell chronic lymphocytic leukemia (B-CLL), using the microlymphocytotoxicity method. A significant increase in the HLA-B13 (P<0.01) and DR53 (P < 0.05) and a significant negative association with the A11 (P < 0.05), B35 (P < 0.05), Cw3 (P < 0.05), and DR1 (P < 0.02) antigens were observed in these patients, compared to the control normal population. These results suggest involvement of some HLA antigens in the multifactorial process of predisposition to B-CLL. C1 [Moazzeni, M Seyed] School of Medical Sciences, Tarbiat Modarres University,, Department of ImmunologyTehran, Iran. [Amirzargar, Ali-Akbar] Ahwaz University of Medical Sciences, Department of ImmunologyAhwaz, Iran. [Shokri, Fazel] School of Medical Sciences, Tarbiat Modarres University,, Department of ImmunologyTehran, LR, Iran. RP Shokri, F (reprint author), School of Medical Sciences, Tarbiat Modarres University,, Department of Immunology, Tehran, Iran. CR Catovsky D, Foa R: The lymphoid leukemias. Buttenworth, London, 1990, pp. 73-111. Cuttner J, Skerrett D, Rosina O, et al: Increased incidence of HLA antigen B35 in patients with chronic lymphocytic leukemia. Leukemia Res 18:565-567, 1994. Dighiero G: Hypogammaglobulinemia and disordered immunity in CLL. In: Chronic lymphocytic leukemia, scientific advances and clinical developments., Ed: Chosen BD). Marcel Dekker, New York, 1993, pp. 167-180. Dorak MT, Chalmers EA, Gaffney D, et al: Human major histocompatibility complex contains several leukemia susceptibility genes. Leuk Lymph 12:211-222, 1994. Dorak MT, Owen G, Galbraith I, et al: Nature of HLA-associated predispoisition to childhood acute lymphoblastic leukemia. Leukemia 9:875-878, 1995. Dorak MT, Machulla HKJ, Hentschel M, et al: Influence of the major histocompatibility complex on age at the onest of chronic lymphocytic leukemia. Int J Cancer 65:134-139, 1996. Dyer PA, Ridway JC, Flanagan NG: HLA-A, B and DR antigens in chronic lymphocytic leukemia. Dis Marker 4:231-237, 1986. Gale RP, Foon KA, Arbor A: Chronic lymphocytic leukemia. Recent advances in biology and treatment. Ann Intern Med 103:101-120, 1985. Gunz FM: The epidemiology and genetics of the chronic leukemias. Clin Hematol 69:3-20, 1977. Jones HP, Whittaker JA: Chronic lymphocytic leukemia: an investigation of HLA antigen frequencies and white cell differential counts in patients, relatives and controls. Leuk Res 15:543- 549, 1991. Kilpatrick DC, Dewar AE, Stockdill G, et al: Histocompatibility antigen frequencies in patient with chronic lymphocytic leukemia: Possible identification of a subgroup with relatively benign disease. Scand J Haematol 33:391-396, 1984. Khamesipur G, Mozdarani H. Shokri F: Cytogenetic analysis of malignant B-cells from Iranian patients with chronic lymphocytic leukemia. Irn J Med Sci 22:50-55, 1997. Linet MS, Bias WB, Dorgan JF, et al: HLA antigens in chronic lymphocytic leukemia. Tissue Antigens 31:71-78, 1988. Neuland CY, Blattner WA, Mann DL, et al: Familial chronic lymphocytic leukemia. J Natl Cancer Inst 71:1143-1150, 1983. Nikbin B, khosravi F, Kamgouyan M, et al: The dirstribution of class I and class II HLA antigens in Iranian population using 3rd. A.O.H.W.C. antisera. In: The proceeding of the 3rd Asia – Oceania histocompatability workshop conference., Ed: Aizawa M), Hokkaido University Press, Sapporo, 1986, pp. 552-555. Oomen LCJM, Van Der Valk MA, Hart AAM, et al: Influence of mouse major histocompatibility complex, H-2, on N-ethyl, N-nitrosourea induced tumor formation in various organs. Cancer Res 48:6634-6641, 1988. Pawelec G, Rehbein A, Schaudt K, et al: Frequencies of HLA - DP allels in four major types of leukaemia. Tissue Antigens 34:138-140, 1989. Raghoebier S, Van-Krieken JH, Kluin-Neleman JC, et al: Oncogene rearrangements in chronic B-cell leukemia. Blood 77:1560-1565, 1991. Rai KR, Sawitsky A, Cronkite EP, et al: Clinical staging of chronic lymphocytic leukemia. Blood 46:219-234, 1975. Schroeder HW, Dighiero G: The pathogenesis of chronic lymphocytic leukemia: analysis of the antibody repertoire. Immunol Today 15:288-294, 1994. Seremetis S, Cuttner J, Winchester R: Definition of a possible genetic basis for susceptibility to acute myelogenous leukemia associated with the presence of polymorphic Ia epitope. J Clin Invest 76:1391-1397, 1985. Shokri F, Mageed RA, Richardson P, et al: Modulation and high frequency expression of autoantibody associated cross – reactive idiotypes linked to VH1 subgroup in CDS-expressing Blymphocytes from patients with chronic lymphocytic leukemia, B-CLL). Stand J Immunol 37:673-679, 1993. Sullivan KA, Amos DB: The HLA system and it’s detection. In: Manual of clinical laboratory immunology., Eds: Rose NR, Friedman H, Fahey JL), 3rd ed., American Society for Microbiolgy, Washington, 1986, pp. 835-846. Svejgaard A: HLA and disease. In: Manual of clinical laboratory immunology., Eds: Rose NR, Friedman H, Fahey JL), 3rd ed. American Society for Microbiology, Washington, 1986, pp.912-920. Terasaki PI, Bernoco D, Parks MS, et al: Microdroplet testing for HLA- A, B, C and DR antigens. Am J Clin Pathol 69:103- 107, 1978. Van Eijk MJT, Stewart-Hayens JA, Beever JE, et al: Development of persistent lymphocytosis in cattel is closely associated with DRB2. Immunogenetics 37:64-68, 1992. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 142 EP 145 PG 4 ER PT J AU Zalatnai, A AF Zalatnai, Attila TI Epidermal Growth Factor Receptor, Somatostatin and Bcl-2 in Human Pancreatic Tumor Xenografts SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE pancreas tumor; xenogaft; somatostatin; bcl-2 ID pancreas tumor; xenogaft; somatostatin; bcl-2 AB Xenografted human pancreatic tumors (5 ductal adenocarcinomas, 1 leiomyosarcoma, altogether 26 samples) were investigated about their immunohistochemical expression of epidermal growth factor receptor (EGFR), somatostatin (SS) and bcl-2 protein. The expression of the EGFR varied from tumor to tumor. One originally negative carcinoma became immunoreactive during passagings, one tumor has lost its early positive expression, and in 3 cancer lines a phenotypically constant pattern was seen. SS immunoreactivity was practically absent in all tumor samples. Concerning bcl-2 expression, different staining patterns were observed among the carcinomas, but the leiomyosarcoma has retained its strong positivity during xenograftings. In the PZX-5 carcinoma line that was originally negative, the one month Sandostatin treatment induced the strong expression of bcl-2 protein suggesting a development of an acquired resistance against programmed cell death in this tumor. C1 [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Zalatnai, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM zalatnai@korb1.sote.hu CR Bell K, Bronner MP, Pasha T, et al: Expression of proliferating cell nuclear antigen in gastrointestinal tract lesions and its relationship to bcl-2 expression. Pathobiology 64:91-98, 1996. Berchem GJ, Bosseler M, Sugars LY, et al: Androgens induce resistance to bcl-2-mediated apoptosis in LNCaP prostate cancer cells. Cancer Res 55:735-738, 1995. Bocsi J, Zalatnai A: Establishment and long term xenografting of human pancreatic carcinomas in immunosuppressed mice: Changes and stability in morphology, DNA ploidy and proliferative activity. J Cancer Res Clin Oncol 125:9-19, 1999. Chester JF, Gaissert HA, Ross JS, et al: Pancreatic cancer in the Syrian golden hamster induced by N-Nitrosobis(2-oxopropyl, amine: cocarcinogenic effect of epidermal growth factor. Cancer Res 46:2954-2957, 1986. Fisher WE, Doran TA, Muscarella II P, et al: Expression of somatostatin receptor subtype 1–5 genes in human pancreatic cancer. J Natl Cancer Inst 90:322-324, 1998. Gansauge F, Gansauge S, Schmidt E, et al: Prognostic significance of molecular alterations in human pancreatic carcinoma – an immunohistochemical study. Langenbecks Arch Surg 383:152-155, 1998. Gower WR jr, Risch RM, Godellas CV, et al: HPAC, a new human glucocorticoid-sensitive pancreatic ductal adenocarcinoma cell line. In Vitro Cell Developm Biol Anim 30A:151- 161, 1994. Hockenbery DM, Zutter M, Hickey W, et al: BCL-2 protein is topographically restricted in tissues characterized by apoptotic cell death. Proc Natl Acad Sci USA 88:6961-6966, 1991. Kuwashima Y, Kobayashi Y, Kurosumi M, et al: Inverse correlation between bcl-2 expression and cell growth fraction in human endometrial adenocarcinoma tissue. Anticancer Res 17:3773-3776, 1997. Lemoine NR, Hughes CM, Barton CM, et al: The epidermal growth factor receptor in human pancreatic cancer. J Pathol 166:7-12, 1992. Makinen K, Hakala T, Lipponen P, et al: Clinical contribution of bcl-2, p53 and Ki-67 proteins in pancreatic ductal adenocarcinoma. Anticancer Res 18:615-618, 1998. Matsuo H, Maruo T, Samoto T: Increased expression of Bcl-2 protein in human uterine leiomyoma and its up-regulation by progesterone. J Clin Endocrinol Metab 82:293-299, 1997. Mejia MC, Navarro S, Pellin A, et al: Study of bcl-2 protein expression and the apoptosis phenomenon in neuroblastoma. Anticancer Res 18:801-806, 1998. Nakanishi H, Ohsawa M, Naka N, et al: Immunohistochemical detection of bcl-2 and p53 proteins and apoptosis in soft tissue sarcoma: their correlations with prognosis. Oncology 54:238- 244, 1997. Ohshio G, Suwa H, Imamura T, et al: An immunohistochemical study of bcl-2 and p53 protein expression in pancreatic carcinomas. Scand J Gastroenterol 33:535-539, 1998. Poston GJ, Townsend CM jr, Rajareman S: Effect of somatostatin and tamoxifen on the growth of human pancreatic cancers in nude mice. Pancreas 5:151-157, 1990. Qin Y, Ertl T, Groot K, et al: Somatostatin analog RC-160 inhibits growth of CFPAC-1 human pancreatic cancer cells in vitro and intracellular proliferation of cyclic adenosine monophosphate. Int J Cancer 60:694-700, 1995. Redding TW, Schally AV: Inhibition of growth of pancreatic carcinomas in animal models by analogs of hypothalamic hormones. Proc Natl Acad Sci USA 81:248-252, 1984. Reed JC: Bcl-2 family proteins and their hormonal control of cell life and death in normalcy and neoplasia. Vitamins and Hormones 53:99-138, 1997. Robertson JF, Watson SA, Hardcastle JD: Effect of gastrointestinal hormones and synthetic analogues on the growth of pancreatic cancer. Int J Cancer 63:69-75, 1995. Rosenberg L, Barkun AN, Denis MH, et al: Low dose octreotide and tamoxifen in the treatment of adenocarcinoma of the pancreas. Cancer 75:23-28, 1995. Saegusa M, Okayasu I: Bcl-2 is closely correlated with favorable prognostic factors and inversely associated with p53 protein accumulation in endometrial carcinomas: immunohistochemical and polymerase chain reaction/loss of heterozygosity findings. J Cancer Res Clin Oncol 123:429-434, 1997. Sinicrope FA, Evans DB, Leach SD, et al: Bcl-2 and p53 expression in resectable pancreatic adenocarcinomas: association with clinical outcome. Clin Cancer Res 2:2015-2022, 1996. Stefanaki K, Rontogiannis D, Vamvouka C, et al: Immunohistochemical detection of bcl-2, p53, mdm2 and p21/waf1 proteins in small-cell lung carcinomas. Anticancer Res. 18:1167-1173, 1998. Szende B, Zalatnai A, and Schally AV: Programmed cell death, apoptosis, in pancreatic cancers of hamsters after treatment with analogs of both luteinizing hormone-releasing hormone and somatostatin. Proc Natl Acad Sci USA 86:1643-1647, 1989. Takeda Y, Escribano MJ: Effects of insulin and somatostatin on the growth and the colony formation of two human pancreatic cancer cell lines. J Cancer Res Clin Oncol 117:416-420, 1991. Teixeira C, Reed JC, Pratt MA: Estrogen promotes chemotherapeutic drug resistance by a mechanism involving Bcl-2 protooncogene expression in human breast cancer cells. Cancer Res 55:3902-3907, 1995. Uegaki K, Nio Y, Inoue Y, et al: Clinicopathological significance of epidermal growth factor and its receptor in human pancreatic cancer. Anticancer Res 17:3841-3847, 1997. Upp JR, Olson D, Poston GJ, et al: Inhibition of growth of two human pancreatic adenocarcinomas in vivo by somatostatin analog SMS 201-995. Am J Surg 155:29-35, 1983. Vidal C, Rauly I, Zeggari M, et al: Up-regulation of somatostatin receptors by epidermal growth factor and gastrin in pancreatic cancer cells. Mol Pharmacol 46:97-104, 1994. Vinter-Jensen L, Juhl CO, Teglbjaerg PS, et al: Systemic treatment with epidermal growth factor in pigs induces ductal proliferations in the pancreas. Gastroenterology 113:1367-1374, 1997. Wada M, Doi R, Hosotani R, et al: Expression of bcl-2 and PCNA in duct cells after pancreatic duct ligation in rats. Pancreas 15:176-182, 1997. Wada M, Hosotani R, Lee JU, et al: An exogenous cdk inhibitor, butyrolactone-1, induces apoptosis with increased Bax/Bcl-2 ratio in p53-mutated pancreatic cancer cells. Anticancer Res 18:2559-2566, 1998. Wang DG, Johnston CF, Buchanan KD: Oncogene expression in gastroenteropancreatic neuroendocrine tumors: implications for pathogenesis. Cancer 80:668-675, 1997. Weckbecker G, Tolcsvai L, Liu R, et al: Preclinical studies on the anticancer activity of somatostatin analogue octreotide, SMS 201-995). Metabolism 41: Suppl. 2., 99-103, 1992. Witty JP, Jensen RA and Johnson AL: Expression and localization of Bcl-2 related proteins in human ovarian cancers. Anticancer Res 18:1223-1230, 1998. Yan JJ, Chen FF, Tsai YC, Jin YT: Immunohistochemical detection of bcl-2 protein in small cell carcinomas. Oncology 53:6-11, 1996. Zalatnai A, Bocsi J, Timar F, et al: Establishment and characterization of a new transplantable pancreatic cancer xenograft, PZX-5, in immunosuppressed mice. Internatl J Pancreatol 23:51-62, 1998. Zalatnai A, Kovacs M, Flautner L, et al: Pancreatic leiomyosarcoma. Case report with immunohistochemical and flow cytometric studies. Virchows Arch 432:469-472, 1998. Zalatnai A, Schally AV: Treatment of the N-Nitrosobis/2-oxopropyl/ amine-induced pancreatic cancer in Syrian golden hamsters with D-Trp-6-LH-RH and somatostatin analog RC-160 microcapsules. Cancer Res 49:1810-1815, 1989. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 146 EP 151 PG 6 ER PT J AU Romics, I AF Romics, Imre TI A Case of Bilateral Testicular Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE testes; lymphoma ID testes; lymphoma AB Authors report on a 75-year-old man with bilateral testicular lymphoma. He complained of painless right testicular enlargement. Orchidectomy was indicated by ultrasound examination and the diagnosis (large cell, non-Hodgkin lymphoma B-cell origin) was established by histology and immunohistochemistry. Two months later, the left testis enlarged, orchidectomy was performed, and a lymphoma with identical histology was found. PET revealed retroperitoneal spread of the tumor. Irradiation (18 Gy) was applied. Three months later, because of gastric metastases of the lymphoma the patient underwent CVP and CAVP (Cyclophosphamide, Adriablastin, Vincristin, Prednisolone) chemotherapy. Despite of the repeated courses, eleven months after the primary diagnosis the patient died due to of multiple metastases. C1 [Romics, Imre] Semmelweis University, Department of Urology, Ulloi ut 78/b, 1082 Budapest, Hungary. RP Romics, I (reprint author), Semmelweis University, Department of Urology, 1082 Budapest, Hungary. CR Adolphs HD, Lindenfelser R, Steffens L, et al: Immunoblastic sarcoma with clinically primary manifestation in the testis, author’s transl). Urol Int 32:8-17, 1977. Bach D, Weissbach L, Adolphs HD: Malignant lymphoma of the testis an urological or nonurological disease? Z Urol Nephrol 71:201-106, 1978. Crellin AM, Hudson BV, Bennett MH, et al: Non-Hodgkins lymphoma of the testis. Radiother Oncol 27:99-106, 1993. Bentley RC, Devlin B, Kaufman RE, et al: Genotypic divergence precedes clinical dissemination in a case of synchronous bilateral B-cell malignant lymphoma of the testes. Hum Pathol 24:675- 678, 1993. Read G: Lymphomas of the testis – results of tretment 1960-77. Clin Radiol 32:687-692, 1981. Frang D, Czvalinga I, Nagy Z, et al: Bilateral testicular tumours. Acta Chir Acad Sci Hung 19:271-280, 1978. Sussman EB, Hajdu SI, Lieberman PH, et al: Malignant lymphoma of the testis: a clinicopathologic study of 37 cases. J Urol 118:1004-1007, 1977. Ito H, Fuse H, Hirano S, et al: Malignant lymphoma of the testis: report of two cases. Hinyokika Kiyo 43:599-603, 1997. Swierz J, Zielinski H, Dabek A: Malignant lymphoma of both testes. Pol Merkuriusz Lek 2:181-182, 1997. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 152 EP 154 PG 3 ER PT J AU Elek, G Slowik, F Eross, L Toth, Sz Szabo, Z Balint, K AF Elek, Gabor Slowik, Felicia Eross, Lorand Toth, Szabolcs Szabo, Zerind Balint, Katalin TI Central Neurocytoma with Malignant Course SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE central neurocytoma; atypical central neurocytoma; glial and neuronal differentiation; ganglioglioma ID central neurocytoma; atypical central neurocytoma; glial and neuronal differentiation; ganglioglioma AB Central neurocytoma is a benign neuronal tumor of young adults in the lateral cerebral ventricles with characteristic X ray and light microscopic findings. In many respects typical central neurocytoma is reported below, with recurrence in the third month requiring reoperation. Death ensued in the fifth postoperative month. Subsequent histology proved progressive vascular proliferation and increasing, unusual glial differentiation of the neuronal tumor. At autopsy tumorous seeding blocked the liquor circulation. A thin tumorous layer covered the surface of all ventricles, the cerebellum and medulla oblongata. The GFAP positive cells out-numbered the synaptophysin positive ones. Increase of GFAP positivity and vascular proliferation of the central neurocytoma may be alarming signs suggesting a malignant course in addition to the other atypical features. C1 [Elek, Gabor] Central Railway Hospital and Polyclinic, Department of Pathology, Pathology of MAV Hospital at Pest, Podmaniczky u 11, H-1062 Budapest, Hungary. [Slowik, Felicia] National Institute of NeurosurgeryBudapest, Hungary. [Eross, Lorand] Hospital of Hungarian Railways, Department of Neurosurgery and Dr Toth Zoltan FoundationBudapest, Hungary. [Toth, Szabolcs] Hospital of Hungarian Railways, Department of Neurosurgery and Dr Toth Zoltan FoundationBudapest, Hungary. [Szabo, Zerind] Hospital of Hungarian Railways, Department of Neurosurgery and Dr Toth Zoltan FoundationBudapest, Hungary. [Balint, Katalin] National Institute of NeurosurgeryBudapest, Hungary. RP Elek, G (reprint author), Central Railway Hospital and Polyclinic, Department of Pathology, H-1062 Budapest, Hungary. CR Akimoto J, Itoh H, Itoh Y, et al: Histogenesis of central neurocytoma: an immunohistochemical and electronmicroscopic study of four cases. No Shinkei Geka 23:1083-1091, 1995. Alleyne CH, Hunter S, Olson JJ, et al: Lipomatosus glioneurocytoma of the posterior fossa with divergent differentiation. Neurosurgery 42:639-643, 1998. Barbosa DM, Balsitis M, Jaspan T, et al: Intraventricular neurocytoma: a clinical and pathological study of three cases. Neurosurgery 26:1045-1054, 1990. Burger PC, Scheithauer BW: Tumors of the central nervous system. Atlas of tumor pathology, third series, AFIP, Fascicle 10:178-184 and 193-194, 1994. Deimling A, Kleihues P, Saremaslani P, et al: Histogenesis and differentiation potential of central neurocytomas. Lab Invest 64:585-591, 1991. Deimling A, Janzer R, Kleihues P, et al: Patterns of differentiation in central neurocytoma. Acta Neuropathol 79:473-479, 1990. Dodds D, Nonis J, Mehta M, et al: Central neurocytoma: a clinical study of response to chemotherapy. J Neurooncol 34:279- 283, 1997. Eng DY, DeMonte F, Ginsberg L, et al: Craniospinal dissemination of central neurocytoma. J Neurosurg 86:547-552, 1997. Figarella-Branger D, Pellissier JF, Daumas-Duport C, et al: Central neurocytomas: critical evaluation of a small cell neuronal tumor. Am J Surg Pathol 16:97-106, 1992. Fujimaki T, Matsuno A, Sasaki T, et al: Proliferative activity of central neurocytoma: doubling time, MIB-1 and bromodeoxyuridine labeling index. J Neurooncol 32:103-109, 1997. Funato H, Inoshita N, Okeda R, et al: Cystic ganglioneurocytoma outside the ventricular region. Acta Neuropathol 94:95- 98, 1997. Giangaspero F, Cenacchi G, Losi L, et al: Extraventricular neoplasms with neurocytoma features. Am J Surg Pathol 21:206- 212, 1997. Hassoun J, Soylemezoglu F, Gambarelli D, et al:Central neurocytoma: a synopsis of clinical and histological features. Brain Pathol 3:297-306, 1993. Horoupian DS, Shuster DL, Kaarsoo-Herrick M, et al: Central neurocytoma: one associated with a fourth ventricular PNET/medulloblastoma and the second mixed with adipose tissue. Hum Pathol 28:1111-1114, 1997. Kim DG, Kim JS, Chi JG, et al: Central neurocytoma: proliferative potential and biological behavior. J Neurosurg 84:742- 747, 1996. Kim DG, Paek SH, Kim IH, et al: Central neurocytoma: the role of radiation therapy and long term outcome. Cancer 79:1995- 2002, 1997. Kim DH, Suh YL: Pseudopapillary neurocytoma of temporal lobe with glial differentiation. Acta Neuropathol 94:187-191, 1997. Kubota T, Hayashi M, Kawano H, et al: Central neurocytoma: immunohistochemical and ultrastructural study. Acta Neuropathol 81:418-427, 1991. Langford LA: Central nervous system noplasms: indications for electron microscopy. Ultrastruct Pathol 20:35-46, 1996. Louis DN, Swearingen B, Linggood RM, et al: Central nervous system neurocytoma and neuroblastoma in adults. J Neurooncol 9:231-238, 1990. Mineura K, Sasajima T, Itoh Y, et al: Blood flow and metabolism of central neurocytoma. Cancer 76:1224-1232, 1995. Mrak RE: Malignant neurocytic tumor. Hum Pathol 25:747- 752, 1994. Nishio S, Fujiwara S, Tashima T, et al: Tumors of the lateral ventricular wall, especially the septum pellucidum. Neurosurgery 27:224-230, 1990. Nishio S, Takeshita I, Fukui M: Primary cerebral ganglioneurocytoma in an adult.Cancer 66:358-362, 1990. Pal L, Santosh V, Gayathri N, et al: Neurocytoma/rhabdomyoma of the cerebellum. Acta Neuropathol 95:318-323, 1998. Schild SE, Scheithauer BW, Haddock MG, et al: Central neurocytomas. Cancer 79:790-795, 1997. Schweitzer JB, Davies KG: Differentiating central neurocytoma. J Neurosurg 86:534-546, 1997. Sgouros S, Carey M, Aluwihare N, et al: Central neurocytoma: correlative clinicopathologic and radiologic analysis. Surg Neurol 49:197-204, 1998. Soylemezoglu F, Scheithauer BW, Esteve J, et al: Atypical central neurocytoma. J Neuropathol Exp Neurol 56:551-556, 1997. Tatter SB, Borges LF, Louis DN: Central neurocytomas of the cervical spinal cord. J Neurosurg 81:288-293, 1994. Tomura N, Hirano H, Watanabe O, et al: Central neurocytoma with clinically malignant behavior. Am J Neuroradiol 18:1175- 1178, 1997. Tsuchida T, Matsamuto M, Shirayama Y, et al: Neuronal and glial characteristics of central neurocytoma. Acta Neuropathol 91:573-577, 1996. Yamamoto T, Komori T, Shibata N, et al: Multifocal neurocytoma/ gangliocytoma with extensive leptomeningeal dissemination in the brain and spinal cord. Am J Surg Pathol 20:363-370, 1996. Yasargil MG, Ammon K, Deimling A, et al: Central neurocytoma: histopathological variants and therapeutic approaches. J Neurosurg 76:32-37, 1992. Valdueza JM, Westphal M, Vortmeyer A, et al: Central neurocytoma: clinical, immunohistologic and biologic findings of a human neuroglial progenitor tumor. Surg Neurol 45:49-56, 1996. Zentner J, Peiffer J, Roggendorf W, et al: Periventricular neurocytoma. Surg Neurol 38:38-42, 1992. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 155 EP 159 PG 5 ER PT J AU Blaker, H Dragoje, S Laissue, JA Herwart, FO AF Blaker, Hendrik Dragoje, Susanne Laissue, Jean-Albert Herwart, F Otto TI Pericardial Involvement by Thymomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE pericardium; thymoma; glomeruloid structures ID pericardium; thymoma; glomeruloid structures AB Thymomas are usually found in the anterior mediastinum, the normal location of the thymus. Involvement of the pericardium by thymic tumors is seen in invasive or metastasized thymoma. Very rarely, thymomas arise primarily in the pericardium. These tumors are believed to derive from thymic tissue which was misplaced in the pericardium during embryologic development. In contrast to patients with orthotopic thymoma who commonly suffer from paraneoplastic diseases, especially myasthenia gravis, patients with intrapericardial thymoma manifestations mainly have symptoms of congestive heart failure which are caused by local complications of tumor growth. In this study, we present two cases of thymoma involving the pericardium. Both tumors were polygonal-oval cell thymomas. In one of the cases diagnosis of an entirely intrapericardial thymoma was established by autopsy. In the other case, explorative thoracotomy revealed massive pericardial and pleural tumor manifestations. The latter tumor showed a peculiar histological pattern with multiple glomeruloid bodies, a finding reported only once for thymomas. C1 [Blaker, Hendrik] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. [Dragoje, Susanne] University of Bern, Department of PathologyBern, Switzerland. [Laissue, Jean-Albert] University of Bern, Department of PathologyBern, Switzerland. [Herwart, F Otto] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. RP Blaker, H (reprint author), University of Heidelberg, Department of Pathology, 69120 Heidelberg, Germany. EM hendrik_blaeker@krzmail.krz.uni-heidelberg.de CR Burke A, Virmani R: Tumors of the heart and the great vessels. Third series, fascicle 16. Washington DC: Armed Forces Institute of Pathology, 1996. Chow W-H, Chow T-C, Chiu S-W: Pericardial metastasis and effusion as the initial manifestation of of malignant thymoma: identification by cross-sectional echocardiography. Int J Cardiol 37:258-260, 1992. Eglen DE: Pericardial based thymoma: diagnosis by fine needle aspiration. Indiana Med 79:526-528, 1986. Gaudin PB, Rosai J: Florid vascular proliferation associated with neural and neuroendocrine neoplasms. Am J Surg Pathol 19:642-652, 1995. Iliceto S, Quagliara D, Calabrese P, et al: Visualization of pericardial thymoma and evaluation of chemotherapy by two dimensional echocardiography. Am Heart J 107:605-606, 1984. Loire R, Hellal H: Pericardites neoplastique. Etude par thoracotomie et biopsie daps 80 cas. Presse Med 22:244-248, 1993. Marino M, Muller Hermelink HK: Thymoma and thymic carcinoma. Virch Arch [A] 407:119-149, 1985. McAllister HA, Fenoglio JJ: Atlas of Tumor Pathology. Tumors of the cardiovascular system. Second series, fascicle 15. Washington DC: Armed Forces Institute of Pathology, 1978. Mirra M, Zanella M, Bussani R, et al: Intrapericardial thymoma. Arch Pathol Lab Med 121:59-63, 1997. Nishimura T, Kondo M, Miyazaki S, et al: Two-dimensional echocardiographic findings in cardiovascular involvement by invasive thymoma. Chest 81:752-754, 1982. Russel DS, Rubinstein LJ: Pathology of tumors of the nervous system. Baltimore, MD: Williams and Wilkins, 1989. Shimosato Y, Mukai K: Atlas of tumor pathology: Tumors of the mediastinum. Third series, fascicle 21. Washington DC: Armed Forces Institute of Pathology, 1997. Venegas RJ, Sun NC: Cardiac tamponade as a presentation of malignant thymoma. Acta Cytol 32:257-262, 1988. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 1999 VL 5 IS 2 BP 160 EP 163 PG 4 ER PT J AU Larsen, KA Gobert, C AF Larsen, K Annette Gobert, Celine TI DNA Topoisomerase I in Oncology: Dr Jekyll or Mr Hyde? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE topoisomerase I; multifunctional proteins; oncogenesis; antitumor agents ID topoisomerase I; multifunctional proteins; oncogenesis; antitumor agents AB Mammalian DNA topoisomerase I is a multifunctional enzyme which is essential for embryonal development. In addition to its classical DNA nicking-closing activities which are needed for relaxation of supercoiled DNA, topoisomerase I can phosphorylate certain splicing factors. The enzyme is also involved in transcriptional regulation through its ability to associate with other proteins in the TFIID-, and possibly TFIIH-, transcription complexes, and is implicated in the recognition of DNA lesions. Finally, topoisomerase I is a recombinase which can mediate illegitimate recombination. A crucial reaction intermediate during relaxation of DNA is the formation of a DNA-topoisomerase I complex (the cleavable complex) where topoisomerase I is covalently linked to a 3’-end of DNA thereby creating a single stranded DNA break. Cleavable complexes are also formed in the vicinity of DNA lesions and in the presence of the antitumor agent, camptothecin. While formation of cleavable complexes may be necessary for the initial stages of the DNA damage response, these complexes are also potentially dangerous to the cell due to their ability to mediate illegitimate recombination, which can lead to genomic instability and oncogenesis. Thus the levels and stability of these complexes have to be strictly regulated. This is obtained by maintaining the enzyme levels relatively constant, by limiting the stability of the cleavable complexes through physical interaction with the oncogene suppressor protein p53 and by degradation of the topoisomerase I by the proteasome system. Emerging evidence suggest that these regulatory functions are perturbed in tumor cells, explaining at the same time why topoisomerase I activities so often are increased in certain human tumors, and why these cells are sensitized to the cytotoxic effects of camptothecins. C1 [Larsen, K Annette] Institut Gustave-Roussy, Laboratory of Biology and Pharmacology of DNA Topoisomerases, 39, Rue Camille Desmoulins, 94805 Villejuif, Cedex, France. [Gobert, Celine] Institut Gustave-Roussy, Laboratory of Biology and Pharmacology of DNA Topoisomerases, 39, Rue Camille Desmoulins, 94805 Villejuif, Cedex, France. RP Larsen, KA (reprint author), Institut Gustave-Roussy, Laboratory of Biology and Pharmacology of DNA Topoisomerases, 94805 Villejuif, France. EM aklarsen@igr.fr CR Rossi F, Labourier E, Forne T, et al: Specific phosphorylation of SR proteins by mammalian DNA topoisomerase I. Nature 381:80-82, 1996. Wang JC: Interaction between DNA and an Escherichia coli protein omega. J Mol Biol 14:523-533, 1971. Champoux JJ, Dulbecco R: An activity from mammalian cells that untwists superhelical DNA; a possible swivel for DNA replication. Proc Natl Acad Sci USA 69:143-146, 1972. Pulleyblank DE, Morgan AR: Partial purification of “omega” protein from calf thymus. Biochemistry 14:5205-5209, 1975. Durnford JM, Champoux JJ: The DNA untwisting enzyme from Saccharomyces cerevisiae. Partial purification and characterization. J Biol Chem 253:1086-1089, 1978. Javaherian K, Tse YC, Vega J: Drosophila topoisomerase I: isolation, purification and characterization. Nucleic Acids Res 10:6945-6955, 1982. Liu LF: HeLa toposiomerase I. Methods Enzymol 100:133-137, 1983. Riou GF, Gabillot M, Douc-Rasy S, et al: A type I DNA topoisomerase from Trypanosoma cruzi. Eur J Biochem 134:479- 484, 1983. Shuman S, Moss B: Identification of a vaccinia virus gene encoding a type I DNA topoisomerase. Proc Natl Acad Sci USA 84:7478-7482, 1987. Wallis JW, Chrebet G, Brodsky G, et al: A hyper-recombination mutation in S. cerevisiae identifies a novel eukaryotic topoisomerase. Cell 58:409-419, 1989. Hanai R, Caron PR, Wang JC: Human TOP3: a single-copy gene encoding DNA topoisomerase III. Proc Natl Acad Sci USA 93:3653-3657, 1996. Stewart L, Ireton GC, Parker LH, et al: Biochemical and biophysical analyses of recombinant forms of human topoisomerase I. J Biol Chem 271:7593-7601, 1996. Redinbo MR, Stewart L, Kuhn P, et al: Crystal structures of human topoisomerase I in covalent and noncovalent complexes with DNA. Science 279:504-513, 1998. Alsner J, Svejstrup JQ, Kjeldsen E, et al: Identification of an Nterminal domain of eukaryotic DNA topoisomerase I dispensable for catalytic activity but essential for in vivo function. J Biol Chem 26:2408-2411, 1992. Labourier E, Rossi F, Gallouzi IE, et al: Interaction between the N-terminal domain of human DNA topoisomerase I and the arginine-serine domain of its substrate determines phosphorylation of SF2/ASF splicing factor. Nucleic Acids Res 26:2955- 2962, 1998. Rossi F, Labourier E, Gallouzi IE, et al: The C-terminal domain but not the tyrosine 723 of human DNA topoisomerase I active site contributes to kinase activity. Nucleic Acids Res 26:2963- 2970, 1998. Juan CC, Hwang J, Liu AA, et al: Human DNA topoisomerase I is encoded by a single-copy gene that maps to chromosome region 20q12-13.2. Proc Natl Acad Sci USA 85:8910-8913, 1988. Kunze N, Yang G, Dolberg M, et al: Structure of the human type I DNA topoisomerase gene. J Biol Chem 266:9610-9616, 1991. Kunze N, Klein M, Richter A, et al: Structural characterization of the human DNA topoisomerase I gene promoter. Eur J Biochem 194:323-330, 1990. Heiland S, Knippers R, Kunze N: The promoter region of the human type-I DNA-topoisomerase gene. Protein-binding sites and sequences involved in transcriptional regulation. Eur J Biochem 21:813-822, 1993. Hwong CL, Chen CY, Shang HF, et al: Increased synthesis and degradation of DNA topoisomerase I during the initial phase of human T lymphocyte proliferation. J Biol Chem 268:18982- 18986, 1993. Kaufmann SH, Charron M, Burke PJ, et al: Changes in topoisomerase I levels and localization during myeloid maturation in vitro and in vivo. Cancer Res 55:1255-1260, 1995. Romig H, Richter A: Expression of the topoisomerase I gene in serum stimulated human fibroblasts. Biochim Biophys Acta 1048:274-280, 1990. Desai SD, Liu LF, Vazquez-Abad D, et al: Ubiquitin-dependent destruction of topoisomerase I is stimulated by the antitumor drug camptothecin. J Biol Chem 272:24159-24164, 1997. Heck MM, Hittelman WN, Earnshaw WC: Differential expression of DNA topoisomerases I and II during the eukaryotic cell cycle. Proc Natl Acad Sci USA 85:1086-1090, 1988. Baker SD, Wadkins RM, Stewart CF, et al: Cell cycle analysis of amount and distribution of nuclear DNA topoisomerase I as determined by fluorescence digital imaging microscopy. Cytometry 19:134-145, 1995. Bronstein IB, Vorobyev S, Timofeev A, et al: Elevations of DNA topoisomerase I catalytic activity and immunoprotein in human malignancies. Oncol Res 8:17-25, 1996. Muller MT, Pfund WP, Mehta VB, et al: Eukaryotic type I topoisomerase is enriched in the nucleolus and catalytically active on ribosomal DNA. EMBO J 4:1237-1243, 1985. Hirano T, Konoha G, Toda T, et al: Essential roles of the RNA polymerase I largest subunit and DNA topoisomerases in the formation of fission yeast nucleolus. J Cell Biol 108:243-253, 1989. Meyer KN, Kjeldsen E, Straub T, et al: Cell cycle-coupled relocation of types I and II topoisomerases and modulation of catalytic enzyme activities. J Cell Biol 136:775-788, 1997. Uemura T, Yanagida M: Isolation of type I and II DNA topoisomerase mutants from fission yeast: single and double mutants show different phenotypes in cell growth and chromatin organization. EMBO J 3:1737-1744, 1984. Thrash C, Bankier AT, Barell BG, et al: Cloning, characterization and sequence of the yeast topoisomerase I gene. Proc Natl Acad Sci USA 82:4374-4378, 1985. Kim RA, Wang JC: Function of DNA topoisomerases as replication swivels in Saccharomyces cerevisiae. J Mol Biol 208:257-267, 1989. DiMauro EG, Camilloni, EG, Verdone L, et al: DNA topoisomerase I controls the kinetics of promoter activation and DNA topology in Saccharomyces cerevisiae. Mol Cell Biol 13:6702- 6710, 1993. Lee MP, Brown SD, Chen A, et al: DNA topoisomerase I is essential in Drosophila melanogaster. Proc Natl Acad Sci USA 90:6656-6660, 1993. Morham SG, Kluckman KD, Voulomanos N, et al: Targeted disruption of the mouse topoisomerase I gene by camptothecin selection. Mol Cell Biol 16:6804-6809, 1996. Yang L, Wold MS, Li JJ, et al: Roles of DNA topoisomerases in simian virus 40 DNA replication in vitro. Proc Natl Acad Sci USA 84:950-954, 1987. Gilmour DS, Pflugfelder G, Wang JC, et al: Topoisomerase I interacts with transcribed regions in Drosophila cells. Cell 44:401-407, 1986. Garg LC, DiAngelo S, Jacob ST: Role of DNA topoisomerase I in the transcription of supercoiled rRNA gene. Proc Natl Acad Sci USA 84:3185-3188, 1987. Merino A, Madden KR, Lane WS, et al: DNA topoisomerase I is involved in both repression and activation of transcription. Nature 365:227-232, 1993. Shykind BM, Kim J, Stewart L, et al: Topoisomerase I enhances TFIID-TFIIA complex assembly during activaton of transcription. Genes Dev 11:397-407, 1997. Kretzschmar M, Meisternst M, Roeder RG: Identification of human DNA topoisomerase I as a cofactor for activator-dependent transcription by RNA polymerase II. Proc Natl Acad Sci USA 90:11508-11512, 1993. Bullock P, Champoux JJ, Botchan M: Association of crossover points with topoisomerase I cleavage sites: a model for nonhomologous recombination. Science 230:954-958, 1985. Cheng C, Kussie P, Pavletich N, et al: Conservation of structure and mechanism between eukaryotic topoisomerase I and sitespecific recombinases. Cell 92:841-850, 1998. Zhu J, Schiestl RH: Topoisomerase I involvement in illegitimate recombination in Saccharomyces cerevisiae. Mol Cell Biol 16:1805-1812, 1996. Bullock P, Forrester W, Botchan M: DNA sequence studies of simian virus 40 chromosomal excision and integration in rat cells. J Mol Biol 174:55-84, 1984. Hino O, Ohtake K, Rogler, CE: Features of two hepatitis B virus, HBV, DNA integrations suggest mechanisms of HBV integration. J Virol 63:2638-2643, 1989. Wang HP, Rogler CE: Topoisomerase I-mediated integration of hepadnavirus DNA in vitro. J Virol 65:2381-2392, 1991. Anderson RD, Berger NA: International Commission for Protection Against Environmental Mutagens and Carcinogens. Mutagenicity and carcinogenicity of topoisomerase-interactive agents. Mutat Res 309:109-142, 1994. Lanza A, Tornaletti S, Rodolfo C, et al: Human DNA topoisomerase I-mediated cleavages stimulated by ultraviolet light-induced DNA damage. J Biol Chem 271:6978-6986, 1996. Pourquier P, Pilon AA, Kohlhagen G, et al: Trapping of mammalian topoisomerase I and recombinations induced by damaged DNA containing nicks or gaps. Importance of DNA end phosphorylation and camptothecin effects. J Biol Chem 272:26441-26447, 1997. Rosenstein BS, Subramanian D, Muller MT: The involvement of topoisomerase I in the induction of DNA-protein crosslinks and DNA single-strand breaks in cells of ultraviolet-irradiated human and frog cell lines. Radiat Res 148:575-579, 1997. Subramanian D, Rosenstein BS, Muller MT: Ultraviolet-induced DNA damage stimulates topoisomerase I-DNA complex formation in vivo: possible relationship with DNA repair. Cancer Res 58:976-984, 1998. Vichi P, Coin F, Renaud JP, et al: Cisplatin and UV-damaged DNA lure the basal transcription factor TFIID/TBP. EMBO J 16:7444-7456, 1997. Castano IB, Brzoska PM, Sadoff BU, et al: Mitotic chromosome condensation in the rDNA requires TRF4 and DNA topoisomerase I in Saccharomyces cerevisiae. Genes Dev 10:2564-2576, 1996. Kimura K, Hirano T: ATP-dependent positive supercoiling of DNA by 13S condensin: a biochemical implication for chromosome condensation. Cell 90:625-634, 1997. Durban E, Goodenough M, Mills J, et al: Topoisomerase I phosphorylation in vitro and in rapidly growing Novikoff hepatoma cells. EMBO J 4:2921-2926, 1985. Kaiserman HB, Ingebritsen TS, Benbow RM: Regulation of Xenopus laevis DNA topoisomerase I activity by phosphorylation in vitro. Biochemistry 27:3216-3222, 1988. Samuels DS, Shimizu Y, Shimizu N: Protein kinase C phosphorylates DNA topoisomerase I. FEBS Lett 259:57-60, 1989. Pommier Y, Kerrigan D, Hartman KD, et al: Phosphorylation of mammalian DNA topoisomerase I and activation by protein kinase C. J Biol Chem 265:9418-9422, 1990. Tournier MF, Sobczak J, de Nechaud B, et al: Comparison of biochemical properties of DNA-topoisomerase I from normal and regenerating liver. Eur J Biochem 210:359-364, 1992. D’Arpa P, Liu LF: Cell cycle-specific and transcription-related phosphorylation of mammalian topoisomerase I. Exp Cell Res 217:125-131, 1995. Krupitza G, Cerutti P: ADP-ribosylation of ADPR-transferase and topoisomerase I in intact mouse epidermal cells JB6. Biochemistry 28:2034-2040, 1989. Boothman DA, Fukunaga N, Wang M: Down-regulation of topoisomerase I in mammalian cells following ionizing radiation. Cancer Res 54:4618-4626, 1994. Ferro AM, Higgins NP, Olivera BM: Poly(ADP-ribosylation, of a DNA topoisomerase. J Biol Chem 258:6000-6003, 1983. Jongstra-Bilen J, Ittel ME, Niedergang C, et al: DNA topoisomerase I from calf thymus is inhibited in vitro by poly(ADPribosylation). Eur J Biochem 136:391-396, 1983. Kasid UN, Halligan B, Liu LF, et al: Poly(ADP-ribose)-mediated post-translational modification of chromatin-associated human topoisomerase I. Inhibitory effects on catalytic activity. J Biol Chem 264:18687-18692, 1989. Adamietz P: Poly(ADP-ribose, synthase is the major endogenous nonhistone acceptor for poly(ADP-ribose, in alkylated rat hepatoma cells. Eur J Biochem 169:365-372, 1987. Ferro AM, Olivera BM: Poly(ADP-ribosylation, of DNA topoisomerase I from calf thymus. J Biol Chem 259:547-554, 1984. Javaherian K, Liu LF: Association of eukaryotic DNA topoisomerase I with nucleosomes and chromosomal proteins. Nucleic Acids Res 11:461-472, 1983. Rose KM, Szopa J, Han FS, et al: Association of DNA topoisomerase I and RNA polymerase I: a possible role for topoisomerase I in ribosomal gene transcription. Chromosoma 96:411- 416, 1988. Bharti AK, Olson MO, Kufe DW, et al: Identification of a nucleolin binding site in human topoisomerase I. J Biol Chem 271:1993-1997, 1996. Kordiyak GJ, Jakes S, Ingebritsen TS, et al: Casein kinase II stimulates Xenopus laevis DNA topoisomerase I by physical association. Biochemistry 33:13484-13491, 1994. Gobert C, Bracco L, Rossi F, et al: Modulation of DNA topoisomerase I activity by p53. Biochemistry 35:5778-5786, 1996. Albor A, Kaku S, Kulesz-Martin M: Wild-type and mutant forms of p53 activate human topoisomerase I: a possible mechanism for gain of function in mutants. Cancer Res 58:2091-2094, 1998. Larsen AK, Gobert C, Gilbert C, et al: DNA topoisomerases as repair enzymes: Mechanism(s, of action and regulation by p53. Acta Biochim Pol 45:535-544, 1998. Gobert C, Skaldanowski A, Larsen AK: The interaction between p53 and DNA topoisomerase I is regulated differently in cells with wild-type and mutant p53. Proc Natl Acad Sci USA, in press, 1999. McLeod HL, Douglas F, Oates M, et al: Topoisomerase I and II activity in human breast, cervix, lung and colon cancer. Int J Cancer 59:607-611, 1994. Vassal G, Pondarre C, Cappelli C, et al: DNA topoisomerase I, a new target for the treatment of neuroblastoma. Eur J Cancer 33:2011-2015, 1997. Husain I, Mohler JL, Seigler H, et al: Elevation of topoisomerase I messenger RNA, protein, and catalytic activity in human tumors: demonstration of tumor-type specificity and implications for cancer chemotherapy. Cancer Res 54:539-546, 1994. Giovanella BC, Steblin JS, Wall ME, et al: DNA topoisomerase I targeted chemotherapy of human colon cancer in xenografs. Science 246:1046-1048, 1989. 82 Lynch BJ, Komaromy-Hiller G, Bronstein IB, et al: Expression of DNA topoisomerase I, DNA topoisomerase II-alpha, and p53 in metastatic malignant melanoma. Hum Pathol 29:1240- 1245, 1998. 83. McLeod HL, Keith WN: Variation in topoisomerase I gene copy number as a mechanism for intrinsic drug sensitivity. Br J Cancer 74:508-512, 1996. 84. Liu LF, Desai SD, Wu JX, et al: The roles of ubiquitin-dependent proteolysis in determining the sensitivity/resistance of tumor cells to topoisomerase inhibitors. Proc Am Ass Cancer Res 40:775, 1999. 85. Larsen AK, Skladanowski A: Cellular resistance to topoisomerase- targeted drugs: from drug uptake to cell death. Biochem Biophys Acta, 1400:257-274, 1998. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 171 EP 178 PG 8 ER PT J AU Eerola, AK Ruokolainen, H Soini, Y Raunio, H Paakko, P AF Eerola, Anna-Kaisa Ruokolainen, Henni Soini, Ylermi Raunio, Hannu Paakko, Paavo TI Accelerated Apoptosis and Low Bcl-2 Expression Associated with Neuroendocrine Differentiation Predict Shortened Survival in Operated Large Cell Carcinoma of the Lung SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mcl-1; bax; bak; neuroendocrine differentiation; prognosis ID Mcl-1; bax; bak; neuroendocrine differentiation; prognosis AB In order to test the hypothesis that increased apoptotic activity is connected with neuroendocrine differentiation and low differentiation degree in large cell carcinoma (LCLC) and is regulated by bcl-2 family proteins, we analysed the extent of apoptosis and tumor necrosis and their relation to the expression of bcl-2, bax, bak and mcl-1 in 35 LCLCs, of which 20 were classified as large cell neuroendocrine lung carcinomas (LCNEC) and 15 as large cell non-neuroendocrine lung carcinomas (LCNNEC). The extent of apoptosis was determined by detecting and counting the relative and absolute numbers of apoptotic cells and bodies using in situ 3'-end labelling of the apoptotic DNA. The extent and intensity of expression of the bcl-2, bax, bak and mcl-1 proteins were studied by immunohistochemistry. Also the relative volume density of necrosis was evaluated and correlated with the other parameters. Finally, all the parameters were evaluated as prognostic markers and correlated with data on the survival of the patients. Relatively high apoptotic indices were seen in both tumor types (average for both 2.53%, range 0.09–27.01%). Significantly higher bcl-2 and bak indices were detected more often in LCNECs than in LCNNECs. Immunohistochemically detected bax, bcl-2 and bak expression was independent of apoptotic index in both tumor types, while there was a statistically significant positive association between mcl-1 expression and apoptotic index in LCNNEC but not in LCNEC. There was a statistically significant association between high apoptotic index and shortened survival in LCLC. However, no association was found between tumor stage and apoptosis. The patients with LCNEC and low bcl-2 protein expression had a significantly shorter survival time than those with high bcl-2 indices. There was also a clear association between shortened survival and necrotic LCNNEC. LCLCs show relatively high apoptotic activity, which is associated with shortened survival. The expression of bcl-2, bak and mcl- 1 is associated with neuroendocrine differentiation in LCLC. Finally, our results support some previous reports suggesting that bcl-2 expression in combination with some other markers involved in apoptosis and/or proliferation may be of prognostic value in cases of lung carcinoma with neuroendocrine differentiation. C1 [Eerola, Anna-Kaisa] University of Oulu, Department of PathologyOulu, Finland. [Ruokolainen, Henni] University of Oulu, Department of PathologyOulu, Finland. [Soini, Ylermi] University of Oulu, Department of Pathology, Kajaanintie 52 D, FIN-90401 Oulu, Finland. [Raunio, Hannu] University of Oulu, Department of Pharmacology and ToxicologyOulu, Finland. [Paakko, Paavo] University of Oulu, Department of Pathology, Kajaanintie 52 D, FIN-90401 Oulu, Finland. RP Paakko, P (reprint author), University of Oulu, Department of Pathology, FIN-90401 Oulu, Finland. EM paavo.paakko@oulu.fi CR Aihara M, Truong ID, Dunn JK, et al: Frequency of apoptotic bodies positively correlates with Gleason grade in prostate cancer. Hum Pathol 25:797-801, 1994. Basolo F, Pollina L, Fontanini G, et al: Apoptosis and proliferation in thyroid carcinoma: correlation with bcl-2 and p53 protein expression. Br J Cancer 75:537-541, 1997. Brambilla E, Negoescu A, Gazzeri S, et al: Apoptosis-related factors p53, bcl2 and bax in neuroendocrine lung tumors. Am J Pathol 149:1941-1952, 1996. Chittenden T, Harrington EA, O´Connor R, et al: Induction of apoptosis by the bcl-2 homologue bak. Nature 374:733-736, 1995. Downey RS, Sewell W, Mansour KA: Large cell carcinoma of the lung: a highly aggressive tumor with dismal prognosis. Ann Thorac Surg 47:806-808, 1989. Eerola AK, Tormanen U, Rainio P, et al: Apoptosis in operated small cell lung carcinoma is inversely related to tumor necrosis and p53 immunoreactivity. J Pathol 181:172-177, 1997. Fleming MV, Guinee DG, Chu WS, et al: Bcl-2 immunohistochemistry in a surgical series of non-small cell lung cancer patients. Hum Pathol 29:60-64, 1998. Fontanini G, Vignati S, Bigini D, et al: Bcl2 protein: a prognostic factor inversely correlated to p53 in non-small cell lung cancer. Br J Cancer 71:1003-1007, 1995. Hellquist HB, Olejnicka B, Jadner M, et al: Fas receptor is expressed in human lung squamous cell carcinomas, whereas bcl-2 and apoptosis are not pronounced. Br J Cancer 76:175-179, 1997. Ikegaki N, Katsumata M, Minna J, et al: Expression of bcl-2 in small cell lung carcinoma cells. Cancer Res 54:6-8, 1994. Jiang SX, Kameya T, Sato Y, et al: Bcl-2 protein expression in lung cancer and close correlation with neuroendocrine differentiation. Am J Pathol 148:837-846, 1996. Joensuu H, Pylkkanen L, Toikkanen S: Bcl-2 protein expression and long-term survival in breast cancer. Am J Pathol 45:1191- 1198, 1994. Kiefer MC, Brauer MJ, Powers VC, et al: Modulation of apoptosis by the widely distributed bcl-2 homologue bak. Nature 374:736-739, 1995. King ED, Matteson J, Jacobs SC, Kyprianou N: Incidence of apoptosis, cell proliferation and bcl-2 expression in transitional cell carcinoma of the bladder: association with tumor progression. J Urol 155:316-320, 1996. Krajewska M, Krajewski S, Epstein JI, et al: Immunohistochemical analysis of bcl-2, bax, bcl-x and mcl-1 expression in prostate cancers. Am J Pathol 148:1567-1576, 1996. Kroemer G: The proto-oncogene bcl-2 and its role in regulating apoptosis., Review, Nature Med 3:614-620, 1997. Laemmli UK: Cleavage of structural proteins during the assembly of the head of bacteriophage Y4. Nature 227:680-685, 1970. Lipponen PK, Aaltomaa S: Apoptosis in bladder cancer as related to standard prognostic factors and prognosis. J Pathol 173:333- 339, 1994. Longthorne VL, Williams GT: Caspase activity is required for commitment to Fas-mediated apoptosis. EMBO J 16:3805-3812, 1997. Mustonen M, Raunio H, Paakko P, et al: The extent of apoptosis is inversely associated with bcl-2 expression in premalignant and malignant breast lesions. Histopathology 31:347-354, 1997. Niehans GA, Brunner T, Frizelle SP, et al: Human lung carcinomas express Fas ligand. Cancer Res 57:1007-1012, 1997. Oltvai ZN, Milliman CL, Korsmeyer SJ: Bcl-2 heterodimerizes in vivo with a conserved homolog, bax, that accelerates programmed cell death. Cell 74:609-619, 1993. Pezzella F, Turley H, Kuzu I, et al: Bcl-2 protein in non-small-cell lung carcinoma. N Engl J Med 329:690-694, 1993. Paakko P, Sormunen R, Risteli L, et al: Malotilate prevents accumulation of type III pN-collagen, type IV collagen, and laminin in carbon tetrachloride-induced pulmonary fibrosis in rats. Am Rev Respir Dis 139:1105-1111, 1989. Rampino N, Yamamoto H, Ionov Y, et al: Somatic frameshift mutations in the bax gene in colon cancers of the microsatellite mutator phenotype. Science 275:967-969, 1997. Reed JC: Bcl-2 and regulation of programmed cell death. J Cell Biol 124:1-6, 1994. Reed JC, Meister L, Tanaka S, et al: Differential expression of bcl- 2 protooncogene in neuroblastoma and other human tumor cell lines of neural origin. Cancer Res 15:6529-38, 1991. Sato T, Hanada M, Bodrug S, et al: Interactions among members of the bcl-2 protein family analyzed by two-hybrid system. Proc Natl Acad Sci USA 91:9238-9242, 1994. Segal NH, Cohen RJ, Haffejee Z, et al: Bcl-2 proto-oncogene expression in the prostatic neuroendocrine cell. Arch Pathol Lab Med 118:616-618, 1994. Silvestrini R, Veneroni S, Daidone MG, et al: The bcl-2 protein: a prognostic indicator strongly related to p53 protein in lymph node-negative breast cancer patients. J Natl Cancer Inst 86:499- 504, 1994. Soini Y, Paakko P, Lehto V-P: Histopathological evaluation of apoptosis in cancer., Review). Am J Pathol 153:1041-1053, 1998. Soini Y, Tormanen U, Paakko P: Apoptosis is inversely related to bcl-2 but not to bax expression in salivary gland tumors. Histopathology 32:28-34, 1998. Soini Y, Virkajarvi N, Lehto VP, et al: Hepatocellular carcinomas with a high proliferation index and a low degree of apoptosis and necrosis are associated with a shortened survival. Br J Cancer 73:1025-1030, 1996. Stammler G, Volm M: Apoptosis in non-small cell lung cancer as related to drug resistance and prognosis. Apoptosis 1:95-99, 1996. Staunton MJ, Gaffney EF: Tumor type is a determinant of susceptibility to apoptosis. Am J Clin Pathol 103:300-307, 1995. Towbin H, Staehelin T, Gordon J: Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: Procedure and some applications. Proc Natl Acad Sci USA 76:4350-4354, 1979. Travis WD, Linnoila I, Tsokos MG, et al: Neuroendocrine tumors of the lung with proposed criteria for large cell neuroendocrine carcinoma. Am J Surg Pathol 15:529-553, 1991. Travis WD, Rush W, Flieder DB, et al: Survival analysis of 200 pulmonary neuroendocrine tumors with clarification of criteria for atypical carcinoid and its separation from typical carcinoid. Am J Surg Pathol 22:934-944, 1998. Tormanen U, Eerola AK, Rainio P, et al: Enhanced apoptosis predicts shortened survival in non-small cell lung carcinoma. Cancer Res 55:5595-5602, 1995. Virkajarvi N, Paakko P, Soini Y: Associaton between p53 overexpression, cell proliferation, tumor necrosis and extent of apoptosis in operated pancreatic carcinoma. APMIS 105:765-772, 1997. Wang DG, Johnston CF, Sloan JM, et al: Expression of bcl-2 in lung neuroendocrine tumors: comparison with p53. J Pathol 184:247-251, 1998. World Health Organization. Histological Typing of Lung Tumors. International Classification of Tumors. No. 1. 2nd edn. Geneva: World Health Organization 1981. Yin X-M, Oltvai ZN, Korsmeyer SJ: BH1 and BH2 domains of bcl- 2 are required for inhibition of apoptosis and heterodimerization with bax. Nature 369:321-323, 1994. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 179 EP 186 PG 8 ER PT J AU Tanyi, J Tory, K Rigo, J Nagy, B Papp, Z AF Tanyi, Janos Tory, Kalman Rigo, Janos Nagy, Balint Papp, Zoltan TI Evaluation of the Tyrosine Kinase Domain of the Met Proto-oncogene in Sporadic Ovarian Carcinomas* SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Met proto-oncogene; epithelial ovarian tumor; tyrosine kinase domain; mutation ID Met proto-oncogene; epithelial ovarian tumor; tyrosine kinase domain; mutation AB Most of the ovarian cancers originate from the ovarian surface epithelium derived from the coelomic mesothelium. The Met proto-oncogene encodes a transmembrane tyrosine kinase receptor (Met) that has the capacity to regulate cell proliferation and differentation and it is activated by hepatocyte growth factor. Trisomy of chromosome 7 and Met protein overexpression have been were observed in ovarian carcinomas, the papillary renal cancers and other solid tumors. Frequent mutations of Met proto-oncogene have been found in hereditary papillary renal cancer (HPRC) and most of the mutations are located in the tyrosine kinase domain. The aim of this study to perform a mutation analysis of exons 17–19 of Met proto-oncogene in epithelial ovarian tumors (EOTs). We have examined 24 tumor samples from patients, operated with EOTs. Mutation was detected in exon 18 in only one sample of 24 EOTs. Our results indicate that mutations located in the Met proto-oncogene is not a common event in EOT. It is not clear whether the mutation plays a role in the tumorigenesis or progression of EOT or not. C1 [Tanyi, Janos] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Baross str. 27., H1088 Budapest, Hungary. [Tory, Kalman] N-Gene R&DNew York, USA. [Rigo, Janos] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Baross str. 27., H1088 Budapest, Hungary. [Nagy, Balint] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Baross str. 27., H1088 Budapest, Hungary. [Papp, Zoltan] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Baross str. 27., H1088 Budapest, Hungary. RP Tanyi, J (reprint author), Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, H1088 Budapest, Hungary. EM tanyi@noi1.sote.hu CR Bergerheim USR, et al: Chromosomal gains and losses in sporadic and hereditary papillary renal carcinomas by comperative genomic hybridization. J Urol 155:543A, 1996. Bladt F, Rietmacher D, Isenmann S, et al: Essential role for the c-Met receptor in the migration of myogenic precursor cells into the limb bud. Nature 376:768-771, 1995. Bottaro DP, Rubin JS, Faletto DL, et al: Identification of the hepatocyte growth factor receptor as the c-Met proto-oncogene product. Science 251:802-804, 1991. Chi-Ho Mok S, Bell DA, Knapp RC, et al: Mutation of K-ras protooncogene in human ovarian tumors of borderline malignancy. Cancer Res 53:1489-1492, 1993. Di Renzo MF, Olivero M, Katsaros D, et al: Overexpression of the Met/HGF receptor in ovarian cancer. Int J Cancer 58:658- 662, 1994. Di Renzo MF, Olivero M, Ferro S, et al.: Overexpression of the c-Met/Hgf receptor gene in human thyroid carcinomas. Oncogene 7:2549-53, 1992. Ebert M, Yokohama M, Friess H, et al: Coexpression of the c- Met proto-oncogene and hepatocyta growth factor in human pancreatic cancer. Cancer Research 54:5775-5778, 1994. Giordano S, Ponzetto C, Di Renzo MF et al: Tyrosine kinase receptor indistinguishable from the c-Met protein. Nature 339:155-156, 1989. Hofstra RM, Landsvater RM, Ceccherini I, et al.: A mutation in the RET proto-oncogene associated with multiple endocrine neoplasia type 2B and sporadic medullary thyroid carcinoma. Nature 367:375-376, 1994. Jeffers M, Rong S, Vande Woude G.: Enhanced tumorigenicity and invasion-Metastasis by hepatocyte growth factor/scatter factor-Met signalling in human cells concomittant with induction of the urokinase proteolysis network. Mol Cell Biol 16:1115-1125, 1996. Kovacs G: Molecular cytogenetics of renal tumors. Adv Cancer Res 62:89-124, 1993. Liang JC, Kurzrock R, Gutterman JU, et al: Trisomy 12 correlates with elevated expression of p21ras in human adenosquamosus carcinoma of the lung. Cancer 23:183-188, 1986. Naldini L, Weidner KM, Vigna E, et al: Scatter factor and hepatocyte growth factor are indistinguishable ligands for the MET receptor. EMBO 10:2867-2878, 1991. Natali PG, Prat M, Nicotra MR, et al: Overexpression of the Met/HGF receptor in renal cell carcinomas. Int J Cancer 69:212-217, 1996. Nusrat A, Parkos CA, Bacarra AE, et al: Hepatocyte growth factor/ scatter factor effects on epithelia. Regulation of intercellular junctions in transformed and nontransformed cell lines, basolateral polarisation of c-Met receptor in transformed and natural intestinal epithelia, and induction of rapid wound repair in a transformed model epithelium. J Clin Invest 93:2056-2065, 1994. Pejovic T, Iosif Cs, Mitelman F, et al: Karyotypic characteristics of borderline malignant tumors of the ovary. Cancer Genet Cytogenet 92:95-98, 1996. Perez RP, Godwin AK, Hamilton TC, et al: Ovarian cancer biology. Semin Oncol 18:186-204, 1991. Persons DL, Hartmann LC, Herath JF, et al: Interphase molecular cytogenetic analysis of epithelial ovarian carcinomas. Am J Pathol 142:733-741, 1993. Prat M, Narsimhan RP, Crepladi T, et al: The receptor encoded by the human c-Met oncogene is expressed in hepatocytes, epithelial cells and solid tumors. Int J Cancer 49:323-328, 1991. Santoro M, Carlomagno F, Romano A, et al: Activation of RET as a dominant transforming gene by germline mutations of MEN2A and MEN2B. Science 267:381-383, 1995. Schmidt L, Fuh-Mei Duh, Fan C, et al: Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nature Genet 16:68-73, 1997. Serov SF, Scully RE, Sobin LH: International histological classification and staging of tumors. No. 9. In: Histologic typing of ovarian tumors. Geneva: World Health Organization, 1973. Sonnenberg E, Meyer D, Weidner KM, rt al: Scatter factor/hepatocyte growth factor and its receptor, the c-Met tyrosine, can mediate a signal exchange between mesenchyme and epithelial cell during mouse development. J Cell Biol 123:223-235, 1993. Tanyi J, Tory K, Rigo J, et al:Mutation in the tyrosine kinase domain of MET proto-oncogene in a sporadic Epithelial ovarian cancer. Submitted to J Reprod Med. Weidner KM, Sachs M, Birchmeier WJ: The Met receptor tyrosine kinase transduces motility, proliferation, and morphogenic signals of scatter factor/hepatocyte growth factor in epithelial cells. Cell Biol 121:145-154, 1993. Woolf AS, Kolatsi-Joannou M, Hardman P, et al: Roles of hepatocyte growth factor/scatter factor and the Met receptor in the early development of the Metanephros. J Cell Biol 128:171- 184, 1995. Zhuang Z, Park WS, Pack S, et al: Trisomy 7-harbouring nonrandom duplication of the mutatnt MET allele in hereditary papillary renal carcinomas. Nat Genet 20:66-69, 1998. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 187 EP 191 PG 5 ER PT J AU Chetty, R Simelane, S AF Chetty, Runjan Simelane, Sobusiso TI p53 and Cyclin A Protein Expression in Squamous Carcinoma of the Oesophagus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE oesophagus; squamous carcinoma; cell cycle; p53; cyclin A ID oesophagus; squamous carcinoma; cell cycle; p53; cyclin A AB The aim of this study was to explore the relationship between p53 and cyclin A immunostaining in squamous carcinomas of the oesophagus. It has been shown that both these proteins are overexpressed in poorly differentiated endometrial carcinomas. Fifty oesophagectomy specimens were analysed for p53 and cyclin A immunoexpression. This was correlated with patient age and gender and tumor stage and grade. Forty-two percent of cases were p53 positive, while 94% of the squamous cancers expressed cyclin A protein. Neither protein showed any statistically significant correlation with clinicopathological parameters. This study has demonstrated that only 42% of oesophageal squamous carcinomas from South Africa express p53 protein, while the vast majority (94%) express cyclin A protein. Neither of these proteins showed any relationship to each other or any clinical feature or the tumor grade or stage. C1 [Chetty, Runjan] University of Natal Medical School, Private Bag 7, 4013 Durban, Congella, South Africa. [Simelane, Sobusiso] University of Natal Medical School, Private Bag 7, 4013 Durban, Congella, South Africa. RP Chetty, R (reprint author), University of Natal Medical School, 4013 Durban, South Africa. EM chettyr@med.und.ac.za CR Stemmermann G, Heffelfinger SC, Noffsinger A, et al: The molecular biology of esophageal and gastric cancer and their precursors: oncogenes, tumor suppressor genes, and growth factors. Hum Pathol 25:968-981, 1994. Motokura T, Arnold A: Cyclins and oncogenesis. Biochima et Biphysica Acta 1155:63-78, 1993. Shiozawa T, Xin L, Nikaido T, et al: Immunohistochemical detection of cyclin A with reference to p53 expression in endometrial endometrioid carcinomas. Int J Gynecol Pathol 16:348-353, 1997. Hollstein MA, Metcalf RA, Welsh JA, et al: Frequent mutation of the p53 gene in human esophageal cancer. Proc Natl Acad Sci USA 87:9958-9961, 1990. Meltzer SJ, Yin J, Huang Y, et al: Reduction to homozygosity involving p53 in esophageal cancers demonstrated by the polymerase chain reaction. Proc Natl Acad Sci USA 88:4976-4980, 1991. Uchino S, Saito T, Inomata M, et al: Prognostic significance of the p53 mutation in esophageal cancer. Jpn J Clin Oncol 26:287-292, 1996. Wang DY, Xiang YY, Tanaka M, et al: High prevalence of p53 protein expression in patients with esophageal cancer in Linxian, China and its relationship to progression and prognosis. Cancer 74:3089-3096, 1994. Sarbia M, Porschen R, Borchard F, et al: p53 protein expression and prognosis in squamous cell carcinoma of the esophagus. Cancer 74:2218-2223, 1994. Shimaya K, Shiozaki H, Inoue M, et al: Significance of p53 expression as a prognostic factor in oesophageal squamous cell carcinoma. Virchows Archiv Pathol Anat Histopathology 422:271-276, 1993. Chetty R, Chetty S: Cyclin D1 and retinoblastoma protein expression in oesophageal squamous carcinoma. J Clin Pathol: Mol Pathol 50:257-260, 1997. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 193 EP 196 PG 4 ER PT J AU Croce, VM Colussi, GA Price, RM Segal-Eiras, A AF Croce, Virginia Maria Colussi, Gladys Andrea Price, R Mike Segal-Eiras, Amada TI Identification and Characterization of Different Subpopulations in a Human Lung Adenocarcinoma Cell Line (A549) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE A549; lung adenocarcinoma; cell subpopulations ID A549; lung adenocarcinoma; cell subpopulations AB The morphology, cell growth, antigenic expression and tumorigenicity of cell subpopulations from the A549 lung adenocarcinoma isolated by Percoll gradient separation have been analysed. Four subpopulations were obtained (subpopulations A, B, C and D). Immunocytochemical analysis of several antigens was performed with monoclonal antibodies (MAbs): MUC1 mucin (C595, HMFG1 and HMFG2), MUC5B (PANH2); gp230 (PANH4); carbohydrate antigens including sialyl Lewis x (KM93), Tn antigen (83D4), Lewis y (C14); 5, 6, 8, 17 and 19 cytokeratins and p53. The cell population D tended to form cell aggregates that piled up on the monolayer similar to overgrowth cultures of the A549 parental cell line, whereas A, B and C cell subpopulations formed well spread monolayers. Both parental A549 and subpopulation D secreted abundant mucus. The topographic distribution and secretion production were correlated with tumorigenic assays since only subpopulation D grew in nude mice exhibiting reduced latency period; these characteristics correlated with the fast growth of the subpopulation D in vitro. Immunocytochemical analysis demonstrated that subpopulation D showed greater expression of MUC1 mucin and carbohydrate antigens such as Tn antigen, sialyl Lewis x and Lewis y and less expression of cytokeratins, p53, MUC5B and gp230; conversely, subpopulations A, B and C showed the opposite antigenic profile. Our results illustrate heterogeneity in the A549 cell line; subpopulations A, B and C retained characteristics of more differentiated adenocarcinoma while subpopulation D displayed features of a less differentiated tumor line. C1 [Croce, Virginia Maria] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Colussi, Gladys Andrea] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Price, R Mike] University of Nottingham, School of Pharmaceutical Sciences, Cancer Research LaboratoriesNottingham, UK. [Segal-Eiras, Amada] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. RP Segal-Eiras, A (reprint author), Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), La Plata, Argentina. EM as-eiras@netverk.com.ar CR Abe K, Hakomori S, Ohshiba S: Differential expression of difucosyl type 2 chain, Le y, defined by monoclonal antibody AH6 in different locations of colonic epithelia, various histological types of colonic polyps and adenocarcinomas. Cancer Res 46:2636-2644, 1986. Alvarez-Fernandez E, Carretero-Albinana I: Expression of blood group antigens in normal bronchopulmonary tissues and common forms of pulmonary carcinomas. Arch Pathol Lab Med 115:42-49, 1991. Berger JT, Horger T, Voynow JA, et al: Respiratory carcinoma cell lines express MUC5/5AC mRNA and secrete MUC5/5AC mucins. 5th International Workshop on Carcinoma Associated Mucins, Cambridge, UK. 1998. Blobel GA, Moll R, Franke WW, et al: Cytokeratins in normal lung and lung carcinomas. I. Adenocarcinomas, squamous cell carcinomas and cultured cell lines. Virchows Arch. B Cell Pathol. Incl. Mol. Pathol. 45(4):407-29, 1984. Broers JL, Rot MK, Oostendorp T, et al: Spontaneous changes in intermediate filament protein expression patterns in lung cancer cell lines. J Cell Sci 91:91-108, 1988. Brown A, Feizi T, Gooi HC: A monoclonal antibody against human colonic adenoma recognises a difucosyalated type-2 blood group chains. Biosci Rep 3:163-169, 1983. Caltagirone S, Ranelleti FO, Rinelli A, et al: Interaction with type II estrogen binding sites and antiproliferative activity of tamoxifen and quercetin in human non-small-cell lung cancer. Am J Respir Cell Mol. Biol. 217:51-9, 1997. Croce MV, Colussi AG, Price M, et al: Immunohistopathological characterization of spontaneous metastases in a human lung mucoepidermoid adenocarcinoma, HLMC, xenograft. Pathol Oncol Res 4:259-266, 1998. Feickert HJ, Anger BR, Cordon-Cardo C, et al: Cell-surface antigens of human lung tumors detected by mouse monoclonal antibodies: definition of blood-group- and non-blood-grouprelated antigenic systems. Int J Cancer 46:1007-1013, 1990. Galbraith SC, Foley J, Kats Y, et al: Clonal variation of p53 expression and proliferative phenotype in A 253 squamous cell carcinoma cells. Oncol Res 8:353-361, 1996. Giard DJ, Aaronson SA, Todaro GJ, et al: In vitro cultivation of human tumors: establishment of cell lines derived from a series of solid tumors. J Natl Cancer Inst 51:1417-1423, 1973. Guzman K, Bader T, Nettesheim P: Regulation of MUC5 and MUC1 gene expression correlation with airway mucous differentiation. Am J Physiol 1846-1853, 1996. Hanai N, Shitara K, Yoshida H: Generation of monoclonal antibodies against human lung squamous cell carcinoma and adenocarcinoma using mice rendered tolerant to normal human lung. Cancer Res 46:4438-4443, 1986. Hakomori S: Aberrant glycosylation in cancer cell membranes as focused on glycolipids: overview and perspectives. Cancer Res 45:2406-2414, 1985. Hirohashi S, Clausen H, Yamada T, et al: Blood group A; cross reacting epitope defined by monoclonal antibodies NCC-lu-35 and -81 expressed in cancer of blood group O or B individuals: its identification as Tn antigen. Proc Natl Acad Sci 82:7039- 7043, 1985. Iggo R, Gatter K, Bartek J, et al: Increased expression of mutant forms of p53 oncogene in primary lung cancer. Lancet 335:675-679, 1990. Izumi Y, Taniuchi Y, Tsuji T, et al: Characterization of human colon carcinoma variant cells selected for sialyl Lex carbohydrate antigen: liver colonization and adhesion to vascular endothelial cells. Exp Cell Res 216:215-21, 1995. Jarrard JA, Linnoila RI, Lee H, et al: MUC1 is a novel marker for the type II pneumocyte lineage during lung carcinogenesis. Cancer Res 58:5582-5589, 1998. Katakoa H, Itoh H, Seguchi K, et al: Establishment and characterization of a human lung adenocarcinoma cell line, LC- 2/ad),producing alpha 1-antitrypsin in vitro. Acta Pathol. Jpn 43(10):566-73, 1993. Kawai T, Suzuki M, Kase K, et al: Expression of carbohydrate antigens in human pulmonary adenocarcinoma. Cancer 72:1581-1587, 1993. Kuan S-F, Byrd JC, Basbaum CB, et al: Characterization of quantitative mucins variants from a human colon cancer cell line. Cancer Res 47:5715-5724, 1987. Ledinko N, Constantino RL: Modulation of p53 gene expression and cytokeratin 18 in retinoid-mediated invasion suppressed lung carcinoma cells. Anticancer Res 10:1335-1339, 1990. Lesuffleur T J, Porchet N, Aubert JP, et al: Differential expression of the human mucin genes MUC1 to MUC5 in relation to growth and differentiation of different mucus-secreting HT-29 cell subpopulations. J Cell Sci 106:771-783, 1993. Lieber M, Smith B, Szakal A, et al: A continuous tumor-cell line from a human lung carcinoma with properties of type II alveolar epithelial cells. Int. J. Cancer 17:62-70, 1976. Masuda N, Fukoaka M, Takada M, et al: Establishment and characterization of 20 human non-small cell lung cancer cell lines in a serum-free defined medium, ACL-4). Chest 100:429- 438, 1991. Nakamori S, Kameyama M, Imaoka S, et al: Involvement of carbohydrate antigen sialyl Lewis x in colorectal cancer metastasis. Dis Colon Rectum 40:420-31, 1997. Nielsen PA, Mandel U, Therkildsen MH, et al: Differential expression of human high-molecular-weight salivary mucin, MG1, and low-molecular–weight salivary mucin, MG2). J Dent Res 75:1820-1826, 1996. Nielsen PA, Mandel U, Therkildsen MH, et al: Loss of a novel mucin-like epithelial glycoprotein in oral and cervical squamous cell carcinoma. Cancer Research 57:634-640, 1997. Pancino GF, Osinaga E, Vorauher W, et al: Production of a monoclonal antibody as immunohistochemical marker on paraffin embedded tissues using a new immunization method. Hybridoma, 9:389, 1990. Price MR, Pugh J, Hudecz F, et al: Immunological and structural features of the protein core of human polymorphic epithelial mucin. Mol Immunol 27:795-801, 1990. Quinlan DC, Davidson AG, Summers CL, et al: Accumulation of p53 protein correlates with a poor prognosis in human lung cancer. Cancer Res 52:4828-4831, 1992. Ramet M, Castren K, Jarvinen K, et al: P53 protein expression is correlated with benzo[a] pyrene-DNA adducts in carcinoma cell lines. Carcinogenesis 16:2117-24, 1995. Resnicoff M, Medrano EE, Podhajcer OL, et al: Subpopulations of MCF7 cells separated by Percoll gradient centrifugation: a model to analize the heterogeneity of human breast cancer. Proc Natl Acad Sci USA 84:7295-7299, 1987. Springer GF: T and Tn, general carcinoma autoantigens. Science 224:1198-1206, 1984. Stranahan PL, Howard RB, Pfenninger O, et al: Mucin gel formed by tumorigenic squamous lung carcinoma cells has Le a-x oligosaccharides and excludes antibodies from underlaying cells. Cancer Res 52:2923-2930, 1992. Svitacheva N, Davies JR, Lesuffleur T, et al: Characterization of mucins produced by the human HT29-MTX cell line. 5th International Workshop on Carcinoma-Associated Mucins, Cambridge, UK, 1998. Takada A, Ohmori K, Yoneda T, et al: Contribution of carbohydrate antigens sialyl Llewis a and sialyl Lewis x to adhesion of human cancer cells to vascular endothelium. Cancer Res. 53:354-361, 1993. Taylor-Papadimitriou J, Peterson J, Arklie J, et al.: Monoclonal antibodies to epithelium-specific components of the human milk fat globule membrane: production and reaction with cells in culture. Int J Cancer, 28:17, 1981. Thompson LH: Mutant isolation. Methods Enzymol 58:308- 322, 1979. Yoshinari K, Kimura H, Sugawara I, et al: Generation of human monoclonal antibodies recognising membranous antigens of the lung adenocarcinoma cell line A549 using an AMeX immunostaing method, Br J Cancer 74:359-367, 1996. Young LR, Wang Y, Voynow J: Neutrophil elastase up-regulates MUC5AC gene expression, 4th International Workshop on Carcinoma- Associated Mucins, Cambridge, UK., 1996. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 197 EP 204 PG 8 ER PT J AU Paraskevakou, H Saetta, A Skandalis, K Tseleni, S Athanassiadis, A Davaris, SP AF Paraskevakou, Helen Saetta, Angelica Skandalis, Kostas Tseleni, Sofia Athanassiadis, Aristides Davaris, S Panayotis TI Morphological-Histochemical Study of Intestinal Carcinoids and K-ras Mutation Analysis in Appendiceal Carcinoids SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE carcinoids; morphology; histogenesis; p53 mutations; ras oncogenes ID carcinoids; morphology; histogenesis; p53 mutations; ras oncogenes AB Intestinal carcinoids are potentially malignant neoplasms. Their histogenesis and pathogenesis are currently uncertain. The morphological and histochemical characteristics of twenty intestinal carcinoids are studied. The primary sites of three mucin-producing tumors were examined by electron microscope. Furthermore 11 appendiceal carcinoids were analysed by the polymerase chain reaction (PCR) for the detection of ras and p53 point mutations. Microscopically all carcinoids were of mixed type. Focal mucin production was evident in three carcinoids that metastasised to regional lymph nodes. HID-Alcian blue staining proved that mucin in both primary and secondary foci did not belong to the sulphated group. The secretory granules and mucin droplets found in a single neoplastic cell suggest that carcinoids of the small intestine and some of the appendix arise from the endoderm. Neither ras nor p53 mutations were detected. It seems that ras oncogenes are probably not involved in the pathogenesis of appendiceal carcinoids. C1 [Paraskevakou, Helen] University of Athens, Department of Pathology, Medical School, 29 Deliyianni Str., GR-145 62 Kifissia, Greece. [Saetta, Angelica] University of Athens, Department of Pathology, Medical School, 29 Deliyianni Str., GR-145 62 Kifissia, Greece. [Skandalis, Kostas] University of Athens, Department of Pathology, Medical School, 29 Deliyianni Str., GR-145 62 Kifissia, Greece. [Tseleni, Sofia] University of Athens, Department of Pathology, Medical School, 29 Deliyianni Str., GR-145 62 Kifissia, Greece. [Athanassiadis, Aristides] University of Athens, Department of Pathology, Medical School, 29 Deliyianni Str., GR-145 62 Kifissia, Greece. [Davaris, S Panayotis] University of Athens, Department of Pathology, Medical School, 29 Deliyianni Str., GR-145 62 Kifissia, Greece. RP Paraskevakou, H (reprint author), University of Athens, Department of Pathology, Medical School, GR-145 62 Kifissia, Greece. EM asaettae@cc.uoa.gr CR Ackerman’s: Surgical Pathology, ed. Rosai J, The CV Mosby Company, 8th edition, 1995. Alpert S, Hanahan D, Teitelman G: Hybrid insulin genes reveal a developmental lineage for pancreatic endocrine cells and imply a relationship with neurons. Cell 53:295-308, 1998. Alumets J, Falkmer S, Hakanson R, et al: Immunohistochemical evidence of peptide hormones in endocrine tumors of the rectum. Cancer 48:2409-2415, 1981. Anderson N, Somerville J, Johnston C, et al: Appendiceal goblet cell carcinoids: a clinicopathological and immunohistochemical study. Histopathology 18:61-65, 1991. Aubock L, Hofler H: Extraepithelial intraneural endocrine cells a starting points for gastrointestinal carcinoids. Virchows Archiv Pathol Anat 401:17-33, 1983. Aubock L, Ratzenhofer M: Extraepithelial enterochromaffin cell-herve-fibre. Complexes in the normal human appendix and in neurogenic appendicopathy. J Pathol 136:217-226, 1982. Baylin S: “APUD” cells. Fact and fiction. TEM March/April 198-203, 1990 Bos J: The ras gene family and human carcinogenesis. Mutation Res 195:255-271, 1988. Cook H: Histological Demonstration Techniques Butterworths and Co Ltd, 1974. Cross SS, Hughes AD, Williams GT Williams ED: Endocrine cell hyperplasia and appendiceal carcinoids. J Pathol 156:325- 329, 1988. Dawson I: The endocrine cells of the gastroinestinal tract and the neoplasms which arise from them. Current Topics in Pathology 63:22, 1976. Erlandson R: Diagnostic transmission. Electron microscopy of tumors. Raven Press 1994. Klappenbach RS, Kurman RJ, Sinclair CF, et al: Composite carcinoma- carcinoid tumors of the gastrointestinal tract. Am J Clin Pathol 84:137-143, 1985. Laurent-Puig P, Olschwang S, Delattre O Validere: Association of Ki-ras mutation with differentiation and tumor-formation pathways in colorectal carcinoma. Int J Cancer 49:220-223, 1991. Listinsky C, Riddell R: Patterns of mucin secretion in neoplastic and non-neoplastic diseases of the colon. Hum Pathol 12:923-929, 1981. Lohmann DR, Fesseler B, Putz B: Infrequent mutations of p53 gene in pulmonary carcinoid tumors. Cancer Res 53:5797- 5801, 1993. Lohmann DR, Funk A, Niedermeyer HP: Identification of p53 gene mutations in gastrointestinal and pancreatic carcinoids by nonradioisotopic SSCA. Virchows Arch B Cell Pathol Incl Mol Pathol 64:293-296, 1993. Lundqvist M, Wilander E: Subepithelial neuroendocrine cells and carcinoid tumors of the human small intestine and appendix. A comparative immunohistochemical study with regard to serotonin, neuron-specific enolase and S-100 protein reactivity. J Pathol 148:141-147, 1986. Moyana T, Satkunam N: A comparative immunohistochemical study of jejunoileal and appendiceal carcinoids. Implications for histogenesis and pathogenesis. Cancer 70:1081-1088, 1992. Nash S, Said J: Gastroenteropancreatic neuroendocrine tumors. A histochemical and immunohistochemical study of epithelial, keratin proteins, carcinoembryonic antigen, and neuroendocrine, neuron-specific enolase, bombesin and chromogranin, markers in foregut, midgut and hindgut tumors. Am J Clin Pathol 86:415-422, 1986. O’Dowd G, Gosney JR: Absence of overexpression of p53 protein by intestinal carcinoid tumors. J Pathol 175:403-404, 1995. Pearse AGE, Pollak JM: Endocrine tumors of neural crest origin- neurolophomas, apudomas and the APUD concept. Med Biol 52:3-18, 1974. Raju G, Lee Y: Gastrointestinal carcinoid tumors: Histological, histochemical and immunohistochemical study. Ann Acad Med Singapore 17:81-88, 1988. Rode J, Dhillon AP, Papadaki L: Serotonin-immunoreactive cells in the lamina propria plexus of the appendix. Human Pathol 145:464-469, 1983. Rodenhuis S, Slebos R: Clinical significance of ras oncogene activation in human lung cancer. Cancer Res 52(Suppl.):2665- 2669, 1992. Rodriquez F, Sarma D, Lunseth J: Goblet cell carcinoid of the appendix. Hum Pathol 13:286-288, 1982. Schmechel D, Marangos PJ, Brightman M: Neuron-specific enolase is a molecular marker for peripheral and central neuroendocrine cells. Nature 276:834-836, 1978. Shamsuddin A, Phelps P, Trump B: Human large intestinal epithelium light microscopy, histochemistry and ultrastructure. Hum Pathol 13: 790-803, 1982. Shaw P: Carcinoid tumors of the appendix are different. J Pathol 162:189-190, 1990. Shaw P: The topografical and age distribution of neuroendocrine cells in normal human appendix. J Pathol 164:235-239, 1991. Sherman SP, Li C-Y, Corney JA: Microproliferation of enterochromaffin cells and the origin of carcinoid tumors of the ileum. Arch Pathol Lab med 103:639-641, 1979. Soga J, Tazawa K: Pathologic analysis of carcinoids. Histologic reevaluation of 62 cases. Cancer 28:990-998, 1971. Spicer S: Diamine methods for differentiating mucosubstances histochemically. J Histochem Cytochem 13:211-234, 1965. Wagner S, Muller R, Boehm J: Neuroendocrine neoplasms of the lung are not associated with point mutations at codon 12 of the Ki-ras gene. Virch Archiv B Cell Pathol 63:325-329, 1993. Wang DG, Johnston CF, Anderson N, et al: Overexpression of the tumour suppressor gene p53 is not implicated in neuroendocrine tumour carcinogenesis. J Pathol 175:397-401, 1995. Younes N, Fulton N, Tanaka R, et al: The presence of K-12 ras mutations in duodenal adenocarcinomas and the absence of ras mutations in other small bowel adenocarcinomas and carcinoid tumors. Cancer 79:1804-1808, 1997. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 205 EP 210 PG 6 ER PT J AU Gunawan, B Granzen, B Keller, U Steinau, G Fuzesi, L Schumpelick, V Mertens, R AF Gunawan, Bastian Granzen, Bernd Keller, Uma Steinau, Gerd Fuzesi, Laszlo Schumpelick, Volker Mertens, Rolf TI Clinical Aspects of Alveolar Rhabdomyosarcoma with Translocation t(1;13)(p36;q14) and Hypotetraploidy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE alveolar rhabdomyosarcoma; soft tissue; cytogenetics; chromosome ID alveolar rhabdomyosarcoma; soft tissue; cytogenetics; chromosome AB Although most cases of alveolar rhabdomyosarcoma (RMS) are characterized by the chromosomal translocation t(2;13)(q35;q14), several cases have been reported with a variant t(1;13)(p36;q14). We present the clinical, morphological and cytogenetic features of an alveolar RMS in a 4-year-old boy. Chromosomal analysis revealed a hypertriploid to hypotetraploid karyotype with a t(1;13)(p36;q14) in all tumor cells. It appears that alveolar RMS with t(1;13) occurs in younger children and displays a higher incidence to upper and lower extremity than tumors with t(2;13). C1 [Gunawan, Bastian] Georg-August-University, Department of PathologyGottingen, Germany. [Granzen, Bernd] Medical Faculty of the Technical University, Department of Pediatrics, RWTH, Pauwelsstr. 30, D-52057 Aachen, Germany. [Keller, Uma] Medical Faculty of the Technical University, Department of Pediatrics, RWTH, Pauwelsstr. 30, D-52057 Aachen, Germany. [Steinau, Gerd] Medical Faculty of the Technical University, Department of SurgeryAachen, Germany. [Fuzesi, Laszlo] Georg-August-University, Department of PathologyGottingen, Germany. [Schumpelick, Volker] Medical Faculty of the Technical University, Department of SurgeryAachen, Germany. [Mertens, Rolf] Medical Faculty of the Technical University, Department of Pediatrics, RWTH, Pauwelsstr. 30, D-52057 Aachen, Germany. RP Mertens, R (reprint author), Medical Faculty of the Technical University, Department of Pediatrics, D-52057 Aachen, Germany. CR Anderson J, Renshaw J, McManus A, et al: Amplification of the t(2;13, and t(1;13, translocations of alveolar rhabdomyosarcoma in small formalin-fixed biopsies using a modified reverse transcriptase polymerase chain reaction. Am J Pathol 150:477- 482, 1997. Biegel JA, Meek RS, Parmiter AH, et al: Chromosomal translocation t(1;13)(p36;q14, in a case of rhabdomyosarcoma. Genes Chromosomes Cancer 3:483-484, 1991. Dal Cin P, Brock P, Aly MS, et al: A variant, 2;13, translocation in rhabdomyosarcoma. Cancer Genet Cytogenet 55:191-195, 1991. Davis RJ, D’Cruz CM, Lovell MA, et al: Fusion of PAX7 to FKHR by the variant t(1;13)(p36;q14, translocation in alveolar rhabdomyosarcoma. Cancer Res 54:2869-2872, 1994. De Zen L, Sommaggio A, d’Amore ESG, et al: Clinical relevance of DNA ploidy and proliferative activity in childhood rhabdomyosarcoma: A retrospective analysis of patients enrolled onto the Italien Cooperative Rhabdomyosarcoma Study RMS88. J Clin Oncol 15:1198-1205, 1997. Douglass EC, Rowe ST, Valentine M, et al: Variant translocations of chromosome 13 in alveolar rhabdomyosarcoma. Genes Chromosom Cancer 3:480-482, 1991. Douglass EC, Valentine M, Etcubanas E, et al: A specific chromosomal abnormality in rhabdomyosarcoma. Cytogenet Cell Genet 45:148-155, 1987. Douglass EC, Shapiro DN, Valentine M, et al: Alveolar Rhabdomyosarcoma with the t(2;13): cytogenetic findings and clinicopathologic correlations. Med Pediatr Oncol 21:83-87, 1993. Enzinger FM, Weiss S: Soft Tissue Tumors. Third edition. St. Louis: Mosby-Year Book, Inc., 1995. ISCN, 1995): An International System for Human Cytogenetic Nomenclature, Mitelman F, ed). S. Karger, Basel, 1995. Kelly KM, Womer RB, Sorensen PHB, et al: Common and variant gene fusions predict distinct clinical phenotypes in rhabdomyosarcoma. J Clin Oncol 15:1831-1836, 1997. Klingebiel T, Pertl U, Hess CF, et al: Treatment of children with relapsed soft tissue sarcoma: report of the german CESS/CWS REZ 91 trial. Med Pediatr Oncol 30:269-275, 1998. Multizentrische Therapiestudie zur Behandlung von Kindern und Jugendlichen mit Weichteilsarkomen, Cooperative Weichteilsarkomstudie CWS-91, Fassung vom 10. Mai 1991. Newton WA Jr, Gehan EA, Webber BL, et al: Classification of rhabdomyosarcomas and related sarcomas. Pathologic aspects and proposal for a new classification – an Intergroup Rhabdomyosarcoma Study Cancer 76:1073-1085, 1995. Shapiro DN, Parham DM, Douglass EC, et al: Relationship of tumor cell ploidy to histologic subtype and treatment outcome in children and adolescents with unresectable rhabdomyosarcoma. J Clin Oncol 9:159-166, 1991. Shapiro DN, Sublett JE, Baitao LI, et al: Fusion of PAX3 to a member of the forkhead family of transcription factors in human alveolar rhabdomyosarcoma. Cancer Res, 53:5108-5112, 1993. Turc-Carel C, Lizard-Nacol S, Justrabo E, et al: Consistent chromosomal translocation in alveolar rhabdomyosarcoma. Cancer Genet Cytogenet 19:361-362, 1986. Wang-Wuu S, Soukup S, Ballard E, et al: Chromosomal analysis of sixteen human rhabdomyosarcomas. Cancer Res 48:983- 987, 1988. Whang-Peng J, Knutsen T, Theil K, et al: Cytogenetic studies in subgroups of rhabdomyosarcoma. Genes Chromosom Cancer 5: 299-310, 1992. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 211 EP 213 PG 3 ER PT J AU Du, B Wang, P Guo, X Du, B AF Du, Bo Wang, Ping Guo, Xiaofeng Du, Baodong TI Expression of Membrane type 1-Matrix Metalloproteinase in Laryngeal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE laryngeal carcinoma; MT1-MMP; invasion; metastasis ID laryngeal carcinoma; MT1-MMP; invasion; metastasis AB Membrane type 1-matrix metalloproteinase (MT1-MMP) is a member of the recently identified unique membrane-type subgroup in the matrix metalloproteinase (MMP) family. MT1-MMP has proteolytic activity against components of the extracellular matrix and activates progelatinase A (MMP-2) at the cell surface. In this study, we analyzed the expression of MT1-MMP mRNA in 45 cases of laryngeal carcinoma by RT-PCR and investigated the relationship between MT1-MMP expression and survival in 18 cases. The result showed that the expression of MT1-MMP mRNA was higher in tumor tissue than in corresponding normal tissue. The tumoral expression in clinical stage III was higher than in stage II. The tumoral expression level of MT1-MMP mRNA in patients with lymph node metastasis was signigicantly higher than those with negative lymph nodes. The patients with high expression level showed significantly poorer 5 year survival than those with low expression level. Collectively, our findings suggest that the high level of MT1-MMP expression is closely related to the invasion and metastasis of laryngeal carcinoma, and indicates poorer prognosis. C1 [Du, Bo] Norman Bethune University of Medical Sciences, Department of ENT & HN, First Clinical Hospital, 130021 Changchun, China. [Wang, Ping] Norman Bethune University of Medical Sciences, Department of ENT & HN, First Clinical Hospital, 130021 Changchun, China. [Guo, Xiaofeng] Norman Bethune University of Medical Sciences, Department of ENT & HN, First Clinical Hospital, 130021 Changchun, China. [Du, Baodong] Norman Bethune University of Medical Sciences, Department of ENT & HN, First Clinical Hospital, 130021 Changchun, China. RP Du, B (reprint author), Norman Bethune University of Medical Sciences, Department of ENT & HN, First Clinical Hospital, 130021 Changchun, China. EM dubo2000@hotmail.com CR Matrisian LM: Metalloproteinases and their inhibitors in matrix remodeling. Trends Genet 6:121-125, 1990. Woessner JFJ: Matrix metalloproteinases and their inhibitors in connective tissue remodeling. FASEB J 5:2145-2154, 1991. Cao J, Sato H, Takino T, et al: The C-terminal region of membrane type matrix metalloproteinase is a functional transmembrane domain required of pro-gelatinase A activation. J Biol Chem 270:801-805, 1995. Emonard HP, Remacle AG, Noel AC, et al: Tumor cell surfaceassociated binding site for the Mr 72.000 type IV collagenase. Cancer Res 52:5845-5848, 1992. Pei D, Weiss S: Transmembrane-deletion mutants of the membrane- type matrix metalloproteinase-1 process proglatianse A and express intrinsic matrix-degrading activity. J Biol Chem 271:9135-9140, 1996. Kinoshita T, Sato H, Takino T, et al: Processing of a precursor of 72kDa type IV collagenase/gelatinase A by a recombinant membrane type 1 matrix metalloproteinase. Cancer Res 56:2535-2538, 1996. Sato H, Takino T, Okada Y, et al: A matrix metalloproteinase expressed on the surface of invasive tumor cells. Nature 370:61-65, 1994. Bando E, Yonemura Y, Endou Y, et al: Immunohistochemical study of MT-MMP tissue status in gastric carcinoma and correlation with survival analyzed by univariate and multivariate analysis. Oncol Rep 5: 1483-1488, 1998. Mori M, Mimori K, Shiraishi T, et al: Analysis of MT1-MMP and MMP2 expression in human gastric cancers. Int J Cancer 74:316-321, 1997. Yamamoto M, Mahanam S, Sawaya R, et al: Differential expression of membrane-type matrix metalloproteinase and its correlation with gelatinase A activation in human malignant brain tumors in vivo and in vitro. Cancer Res 56:384-392, 1996. Gilles C, Polette M, Piette J, et al: High level of MT1-MMP expression is associated with invasiveness of cervical cancer cells. Int J Cancer 65:209-213, 1996. Tokuraku M, Sato H, Shinagawa A, et al: Activation of the precursor of gelatinase A/72 kDa type IV collagenase/MMP-2 in lung carcinoma correlation with the expression of membranetype matrix metalloproteinase 1, MT1-MMP, and with lymph node metastasis. Int J Cancer 64: 355-359, 1995. Yoshizaki T, Sato H, Maruyama Y, et al: Increased expression of membrane type 1-matrix metalloproteinase in head and neck carcinoma. Cancer 79:139-144, 1977. Repassy G, Forster-Horvath Cs, Juhasz A, et al. Expression of invasion markers CD44v6/v3, nm23 and MMP2 in laryngeal and hypopharyngeal carcinoma. Pathol Oncol Res 4:14-22, 1998. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 214 EP 217 PG 4 ER PT J AU Banfalvi, T Gilde, K Boldizsar, M Kremmer, T Otto, Sz AF Banfalvi, Teodora Gilde, Katalin Boldizsar, Mariann Kremmer, Tibor Otto, Szabolcs TI Serum levels of S-100 protein and 5-S-cysteinyldopa as markers of melanoma progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE melanoma; S-100 protein; 5-S-cysteinyldopa ID melanoma; S-100 protein; 5-S-cysteinyldopa AB Serum S-100 protein is widely used as a marker of melanoma and since 5-S-cysteinyldopa (5-S-CD) is a precursor of melanin its serum and urinary levels can reflect melanoma progression. In this study we examined the concentration changes of serum S-100 protein and 5-S-CD in 252 melanoma patients of different clinical stages. Serum samples were taken from 252 melanoma patients at 860 times, from June 1996 to July 1998. The serum S-100 protein was measured by the immunoluminometric assay, levels of 5-S-CD was determined by HPLC. The value of S-100 protein in patients with primary melanoma (0.11m mg/l) and in patients without symptoms (0.15 m mg/l) ranged around the normal level (0.01–0.12 m mg/l). There was a significant difference between the values of patients with or without symptoms. There was a similarly significant difference between the S-100 values of clinical Stage I (0.11 mg/l) and Stage III (2.91 mg/l) as well as between those of clinical Stage II (0.47 mg/l) and Stage III (2.91 mg/l), respectively. Analyzing the values of patients with symptoms we observed significant difference between the S-100 protein values of patients with primary tumor and those with solitary or multiple distant metastases. In case of 5-S-CD significant difference was found between clinical Stage I and III as well as clinical Stage II and III. Furthermore, there was a significant difference between the mean marker values of patients with primary tumor, lymph node, lung metastasis and clinical stage III. C1 [Banfalvi, Teodora] National Institute of Oncology, Department of Dermatology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Gilde, Katalin] National Institute of Oncology, Department of Dermatology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Boldizsar, Mariann] National Institute of Oncology, Department of Biochemistry, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Kremmer, Tibor] National Institute of Oncology, Department of Biochemistry, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Otto, Szabolcs] National Institute of Oncology, Central Clinical Laboratory, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. RP Banfalvi, T (reprint author), National Institute of Oncology, Department of Dermatology, H-1122 Budapest, Hungary. EM banfalvi@oncol.hu CR Agrup G, Agrup P, Andersson T, et al: 5 years experience of 5- S-cysteinyldopa in melanoma diagnosis. Acta Dermatovener, Stockholm, 59:381-388, 1979. Benathan M: Modulation of 5-S-cysteinyldopa formation by tyrosinase activity and intracellular thiols in human melanoma cells. Melanoma Res 6:183-189, 1996. Bonfrer JM, Korse CM, Israels SP: Serum S-100 has prognostic significance in malignant melanoma. Anticancer Res 17:2975-7, 1997. Buer J, Probst M, Franzke A, et al: Elevated serum levels of S- 100 and survival in metastatic malignant melanoma. Br J Cancer 75:1373-6, 1997. Cho K, Hashimoto K, Taniguchi Y, et al: Immunohistochemical study of melanocytic nevus and malignant melanoma with monoclonal antibodies against S-100 subunit. Cancer 66:765- 771, 1990. Fagnart CO, Sindic MJ, Laterre CH: Particle counting immunassay of S-100 protein in serum. Possible relevance in tumours and ischemic disorders of the central nervous system. Clin Chem 34:1387-1391, 1988. Gaynor R, Irie R, Morton D: S-100 protein is present in cultured human malignant melanomas. Nature 286:400-401, 1980. Gaynor R, Irie R, Morton D, et al: S-100 protein: A marker for human malignant melanoma? Lancet 18:869-871, 1981. Guo HB, Stoffel-Wagner B, Bierwirth T, et al: Clinical significance of serum S-100 in metastatic malignant melanoma. Eur J Cancer 31:1898-1902, 1995. Hanson LO, Schoultz E, Djuren E, et al: Prognostic value of serum analyses of S-100 protein beta in malignant melanoma. Anticancer Res 17:3071-3073, 1997. Hasegawa M, Takata M, Hatta N, et al: Simultaneous measurement of serum 5-S-cysteinyldopa, circulating intercellular adhesion molecule-1 and soluble interleukin-2 receptor levels in Japanese patients with malignant melanoma Melanoma Res 7:243-251, 1997. Henze G, Dummer R, Joller-Jemelka H, et al: Serum S-100 – a marker for disease monitoring in metastatic melanoma. Dermatology 194:208-212, 1997. Hirai S, Kageshita T, Kimura T, et al: Serum levels of s-ICAM, and 5-S-cysteinyldopa as markers of melanoma progression. Melanoma Res 7:58-62, 1997. Horikoshi T, Ito Sh, Wakamatsu K, et al: Evaluation of melaninrelated metabolites as markers of melanoma progression. Cancer 73:629-636, 1994. Kagedal B, Pettersson A: Determination of urinary 5-S-cysteinyldopa by high performance liqiud chromatography Chromatogr 272:287-297, 1983. Ketcham AS, Moffat FL, Balch CM: Classification and staging In: CM Balch Cutaneous melanoma Philadelphia, JB Lippincott, 1992, pp. 213-220. Meyerhoffer S, Lindberg Z, Hager A, et al: Urinary excretion of 5-S-cysteinyldopa and 6-hydroxy-5-methoxyindole-2-carboxylic acid in children Acta Derm Venereol 78:31-35, 1998. Miliotes G, Lyman GH, Cruse CW, et al: Evalutaiton of new putative tumor markers for melanoma Ann Surg Oncol 3:558-563, 1996. Nakajima T, Watanabe S, Sato Y, et al: Immunohistochemical demonstration of S-100 protein in malignant melanoma and pigmented nevus and its diagnostic application. Cancer 50:912- 918, 1982. Nyberg L, Kroon L, Ullen A, et al: Sangtec LIA a new sensitive monoclonal assay for measuring protein S 100 in patients with malignant melanoma in Final Program and Abstract Book of the XXIV Meeting of ISOBM. San Diego, 1996. Peterson L, Woodward W, Fletcher W, et al: Plasma 5-S-cysteinyldopa differentietes patients with primary and metastatic melanoma from patients with dysplasticus naevus syndroma and normal subjects. J Am Acad Dermatol 19:509-515, 1988. Schoultz E, Hansson LO, Djureen E, et al: Prognostic value of serum analyses of S-100 beta protein in malignant melanoma. Melanoma Res 6:133-137, 1996. Schoultz ES, Diepgen TL, Von Den Driesch P: Clinical and prognostic relevance of serum S-100 beta protein in malignant melanoma Br J Dermatol 138:426-430, 1998. Sasaki Y, Shimizu H, Naka W, et al: Evaluation of the clinical usefulness of measuring urinary excretion of 5-S-cysteinyldopa in melanoma: ten years experience of 50 patients Acta Derm Venerol 77:379-381, 1998. Wakamatsu K, Ito S: Seasonal variations in serum concentration of 5-S- cysteinyldopa and 6-hydroxy-5-methoxyindole-2-carboxyl acid in healthy Japanese. Pigment Cell Res 8:132-134, 1995. Wimmer I, Meyer CJ, Seifert B, et al.: Prognostic value of serum 5-S-cysteinyldopa for monitoring human metastatic melanoma during immunochemoterapy. Cancer Res 57:5073-5076, 1997. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 218 EP 222 PG 5 ER PT J AU Durmaz, R Deliorman, S Isiksoy, S Uyar, R Erol, K Tel, E AF Durmaz, Ramazan Deliorman, Selda Isiksoy, Serap Uyar, Ruhi Erol, Kevser Tel, Esref TI Antiproliferative Properties of the Lazaroids U-83836E and U-74389G on Glioma Cells In Vitro SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE glioma; C6; MTT; 21-aminosteroids; U-83836E; U-74389G ID glioma; C6; MTT; 21-aminosteroids; U-83836E; U-74389G AB The 21-aminosteroids (lazaroids) are a new family of steroid compounds that inhibit lipid peroxidation reactions. They are novel antioxidant agents, which have been shown to have antiproliferative properties on cancer cells and also are thought to prevent free radical-mediated blood-brain barrier damage. In order to understand the effect of lazaroids on glioma, we tested U-83836E and U-74389G at doses ranging between 0.1–100 m mM on primary cultures of glioblastoma multiforme from three patients, rat C6 glioma cell line, and 5 th subculture established from one of the patients. The effects of both compounds on cell proliferation were determined using 3-(4,5-dimethyl thiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) colorimetric assay. U-83836E in the primary cultures was found to have 50% inhibitory concentrations (IC50 ) of 6.30, 6.75 and 6.50 m mM, respectively. The IC50 value of U-74389G was calculated as 91 m mM in only one of the patients. On C6 glioma cells, while the IC50 of U-83836E was 45 m mM, U-74389G showed no cytotoxic effect. On the 5 th subculture, U-83836E had an IC50 of 37.5 m mM, but the cytotoxic effects of U-74389G was less than in that of the primary culture. In conclusion, these compounds were found to be more cytotoxic in primary culture than the cell lines and there were also differences between their members in the inhibition of cell survival. C1 [Durmaz, Ramazan] Medical Faculty of Osmangazi University, Department of Neurosurgery, 90-26480 Eskisehir, Turkey. [Deliorman, Selda] Medical Faculty of Osmangazi University, Department of PhysiologyEskisehir, Turkey. [Isiksoy, Serap] Eskisehir Osmangazi University, Faculty of Medicine, Department of PathologyEskisehir, Turkey. [Uyar, Ruhi] Medical Faculty of Osmangazi University, Department of PhysiologyEskisehir, Turkey. [Erol, Kevser] Medical Faculty of Osmangazi University, Department of PharmacologyEskisehir, Turkey. [Tel, Esref] Medical Faculty of Osmangazi University, Department of Neurosurgery, 90-26480 Eskisehir, Turkey. RP Durmaz, R (reprint author), Medical Faculty of Osmangazi University, Department of Neurosurgery, 90-26480 Eskisehir, Turkey. EM rdurmaz@ogu.edu.tr CR Alley MC, Scudiero DA, Monks A, et al: Feasibility of drug screening with panels of human tumor cell lines using a microculture tetrazolium assay. Cancer Res 48:589-601, 1988. Arbit E, Rubinstein A, Di Resta G, et al: The effect of the aminosteroids U-78517G on peritumoral brain oedema. Acta Neurochir, Wien, Suppl 51:158-159, 1990. Braughler JM, Pregenzer JF: The 21-Aminosteroid inhibitors of lipid peroxidation: reactions with lipid peroxyl and phenoxy radicals. Free Rad Biol Med 7:125-130, 1989. Braughler JM, Hall ED, Jacobson EJ, et al: The 21-aminostroids: Potent inhibitors of lipid peroxidation for the treatment of central nervous system trauma and ischemia. Drugs of Future 14:143-152, 1989. Buatti JM, Friedman WA, Theele DP, et al. The lazaroid U- 74389G protects normal brain from stereotactic radiosurgeryinduced radiation injury. Int J Radiation Oncology Biol Phys 34:591-597, 1996. Carmichael J, DeGraff WG, Gazdar AF, et al: Evaluation of tetrazolium- based semiautomated colorimetric assay: assesment of chemosensitivity testing. Cancer Res 47:936-942, 1987. Chen TT, Mealey J: Microculture of human brain tumors. Cancer Chemother Rep 54: 9-14, 1970. Donalt TJ Jr, Higgins GM: The effects of high levels of certain steriods on induced lymphocytic leukemia in the rat. Cancer Res 11:937-942, 1951. Durmaz R, Inal M, Angin K, et al: The effects of MK-801 and U-83836E on post-ischemic reperfusion injury in rat brain. Acta Neurobiol Exp 59:99-104, 1999. Franks LM, Wilson PD: Origin and ultrasutructure of cells in vitro. In: International review of cytology., Eds: Bourne GH, Danielli FJ, Jeon KW), Academic Press, 1977, pp. 55-139. Freshney RI: Effects of glucocorticoids on glioma cells in culture. Minireview on cancer research. Exp Cell Biol 52:286-292, 1984. Giese H, Mertsch K, Blasig IE: Effect of MK-801 and U83836E on a porcine brain capillary endothelial cell barrier during hypoxia. Neuroscience Letters 191:169-172, 1995. Glick RP, Gettleman R, Patel K, et al: Insulin and insulin-like growth factor I in brain tumors: Binding and in vitro effects. Neurosurgery 24:791-797, 1989. Hall ED, Andrus PK, Smith SL, et al: Neuroprotective efficacy of microvasculary-localized versus brain-penetrating antioxidants. Acta Neurochir, suppl, 66:107-113, 1996. Hall ED, Braughler JM, Yonkers PA, et al: U-78517F: A potent inhibitor of lipid peroxidation with activity in experimental brain injury and ischemia. J Pharmacol Exp Ther 258:688-694, 1991. Hall ED, Yonkers PA, McCall JM, et al: Effects of the 21- aminosteroid U74006F on experimental head injury in mice. J Neurosurg 68:456-461, 1988. Kim RS, Zaborniak CL, Begleiter A, et al: Antiproliferative properties of aminosteroid antioxidant on cultured cancer cells. Cancer Lett 64:61-66, 1992. King WA, Black KL, Ikezaki K, et al: Novel 21-aminosteroids prevent tumor associated neurological dysfunction. Acta Neurochir, wien, Suppl 51:160-162,1990. Kornblith PL: Role of tissue culture in prediction of malignancy. Clin Neurosurg 25:346-375, 1978. Maciunas RJ, Mericle RA, Sneed CL, et al: Determination of the lethal dose of dexamethasone for early passage in vitro human glioblastoma cell cultures. Neurosurgery 33:485-488, 1993. Megyesi JF, Farrell CL, Del Maestro RF: Investigation of inhibitor of lipid peroxidation U74006F on tumor growth and protein extravasation in the C6 astrocytoma spheroid implantation glioma model. J Neuro-Oncol 8:133-137, 1990. Mossmann T: Rapid colorimetric assay of cellular growth and survival: Application to proliferation and cytotoxicity assays. J Immun Method 65:55-63, 1983. Motulsky HJ, Stannard P, Neubig R, et al: GraphPad Software, Inc, San Diego, 1994-1995. Muller TB, Haraldseth O, Jones RA, et al: Perfusion and diffusion- weighted MR imaging for in vitro evaluation of treatment with U-74389G in rat stroke model. Stroke 26:1453-1458, 1995. Pertuiset B, Dougherty D, Cromeyer C, et al: Stem cell studies of human malignant brain tumors. Part 2: proliferation kinetics of brain-tumor cells in vitro in early-passage cultures. J Neurosurg 63:426-432, 1985. Russell DS, Rubinstein LJ: Pathology of tumours of the nervous system. Edward Arnold, London, 1989, pp 133-140. Smith SL, Scherch HM, Hall ED: Protective effect of tirilazad mesylate and metabolite U-89678 against blood-brain barrier damage after subarachnoid hemorrrage and lipid peroxidative neuronal injury. J Neurosurg 84:229-233, 1996. Thomas DGT, Darling JL, Paul EA, et al: Assay of anticancer drugs tissue culture: relationship of relapse free interval, RFI, and in vitro chemosensitivity in patients with malignant cerebral glioma. Br J Cancer 51:525-532, 1985. Tsuruo T, Iida H, Tsukagoshi S, et al: Overcoming of vincristine resistance in P388 leukemia in vivo and in vitro through enhanced cytotoxicity of vincristine and vinblastine by verapamil. Cancer Res 41:1967-1972, 1981. Unterman TG, Glick RP, Waites GT, et al: Production of insulinlike growth factor-binding proteins by human central nervous system tumors. Cancer Res 51:3030-3036, 1991. Vincent AJPE, Esandi MC, Avezaat CJJ, et al: Preclinical testing of recombinant adenoviral herpes simplex virus-thymidine kinase gene therapy for central nervous system malignancies. Neurosurgery 41:442-452, 1997. Wright RL, Shaumba B, Keller J: The effect of glucocorticosteroids on growth and metabolism of experimental glial tumors. J Neurosurg 30:140-145, 1969. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 223 EP 228 PG 6 ER PT J AU Ustun, C Ozcan, M Gurman, G Cakir, M Erekul, S Akyol, G Arat, M Celebi, H Idlman, R Ilhan, O Demirer, T Beksac, M Koc, H AF Ustun, Celalettin Ozcan, Muhit Gurman, Gunhan Cakir, Mehtap Erekul, Selim Akyol, Gulen Arat, Mutlu Celebi, Harika Idlman, Ramazan Ilhan, Osman Demirer, Taner Beksac, Meral Koc, Haluk TI Differences in Liver Pathology and Clinical Outcome Between Two Patients with Hepatitis B Virus and Graft Versus Host Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE alloBMT; hepatitis B virus; graft versus-host disease ID alloBMT; hepatitis B virus; graft versus-host disease AB Our two patients undergoing allogeneic bone marrow transplantation (AlloBMT) had both Hepatitis B virus (HBV) and graft-versus-host disease (GVHD). In the first patient, liver enzymes elevated three months after AlloBMT, and GVHD was diagnosed. Two weeks after the diagnosis of GVHD, HBsAg appeared in his serum. At that time, liver biopsy was not able to discriminate two disorders, but his sequential liver biopsies disclosed GVHD. Despite the patient was treated with cyclosporin A (CsA), he died for chronic GVHD. In contrast to the first patient, the second patient had HBsAg prior to GVHD. His liver enzymes deterioration was detected in the first month after AlloBMT, and reached the highest level in the third month while withdrawing CsA. In the fifth month he developed scleradermatous skin changes, and skin biopsy revealed chronic GVHD, whereas concurrent liver biopsy revealed chronic active hepatitis. This observation showed that immunosuppressive conditions such as GVHD or its prophylaxis may affect the appearance of liver pathology caused by HBV, which depends on the time of GVHD development, and the duration and depth of GVHD prophylaxis. C1 [Ustun, Celalettin] Ankara University, School of Medicine, Department of Hematology, Ibni Sina Hospital, 06100 Ankara, Sihhiye, Turkey. [Ozcan, Muhit] Ankara University, School of Medicine, Department of Hematology, Ibni Sina Hospital, 06100 Ankara, Sihhiye, Turkey. [Gurman, Gunhan] Ankara University, School of Medicine, Department of Hematology, Ibni Sina Hospital, 06100 Ankara, Sihhiye, Turkey. [Cakir, Mehtap] Ankara University, School of Medicine, Department of PathologyAnkara, Turkey. [Erekul, Selim] Ankara University, School of Medicine, Department of PathologyAnkara, Turkey. [Akyol, Gulen] Gazi University, School of Medicine, Department of PathologyGazi, Turkey. [Arat, Mutlu] Ankara University, School of Medicine, Department of Hematology, Ibni Sina Hospital, 06100 Ankara, Sihhiye, Turkey. [Celebi, Harika] Ankara University, School of Medicine, Department of Hematology, Ibni Sina Hospital, 06100 Ankara, Sihhiye, Turkey. [Idlman, Ramazan] Ankara University, School of Medicine, Department of GastroenterologyAnkara, Turkey. [Ilhan, Osman] Ankara University, School of Medicine, Department of Hematology, Ibni Sina Hospital, 06100 Ankara, Sihhiye, Turkey. [Demirer, Taner] Ankara University, School of Medicine, Department of Hematology, Ibni Sina Hospital, 06100 Ankara, Sihhiye, Turkey. [Beksac, Meral] Ankara University, School of Medicine, Department of Hematology, Ibni Sina Hospital, 06100 Ankara, Sihhiye, Turkey. [Koc, Haluk] Ankara University, School of Medicine, Department of Hematology, Ibni Sina Hospital, 06100 Ankara, Sihhiye, Turkey. RP Koc, H (reprint author), Ankara University, School of Medicine, Department of Hematology, 06100 Ankara, Turkey. EM ustun@pallas.dialup.ankara.edu.tr CR Atkinson K: Recostruction of the haemopoietic and immune systems after marrow transplantation. Bone Marrow Transplant 5:209-226, 1990. Chen PM, Fan S, Liu JH, et al: Reactivation of hepatitis B virus in two chronic GVHD patients after transplant. Int J Hematol 58:183-188, 1993. Chen PM, Fan S, Liu CJ, et al: Changing of hepatitis B virus markers in patients with bone marrow transplantation. Transplantation 49:708-713, 1990. Gerber MA, Thung SN: Biology of disease molecular and cellular pathology of hepatitis B.Lab Invest 53:572-590, 1985. Hoofnagle JH, Dusheiko GM, Schafer DF, et al: Reactivation of chronic hepatitis B virus infection by cancer chemotherapy. Ann Int Med 96:447-449, 1982. Lau GKK, Liang R, Chiu EKW, et al: Hepatic events after bone marrow transplantation in patients with hepatitis B infection: a case controlled study. Bone Marrow Transplant 19:795-799, 1997. Lawrence GL: Immune recovery after bone marrow transplantation. Hematol/Oncol Clin N Am 4:659-675, 1990. Locasciulli A, Alberti A, Bock R, et al. Impact of liver disease and hepatitis infections on allogeneic bone marrow transplantation in Europe: a survey from the European Bone Marrow Transplantation, EBMT, Groupe – Infections Diseases Working Party. Bone Marrow Transplant 14:833-837, 1994. Locasciulli A, Bacigalupo A, Vanlint MT, et al: Hepatitis B virus infection and liver disease after allogeneic bone marrow transplantation. A report of 30 cases. Bone Marrow Transplant 6:25- 29, 1990. Pariente EA, Goudeau, Dubois F, et al: Fulminant hepatitis due to reactivation of chronic hepatitis B virus infection after allogeneic bone marrow transplantation. Dig Dis Sci 33:1185- 1191, 1988. Ustun C, Koc H, Karayalcyn S, et al: Hepatitis B virus infection in allogeneic bone marrow transplantation. Bone Marrow Transplant 20:289-296, 1997. Ustun C, Idilman R, Gurman G, et al. Hematopoietic stem cell transplantation in patients with non-replicative hepatitis B virus carriers is safe. J Hepatol, in press). Webster A, Brenner MK, Prentice HG, et al: Fatal hepatitis B reactivation after autologous bone marrow transplantation. Bone Marrow Transplant 49:708-713, 1989. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 229 EP 232 PG 4 ER PT J AU Strausz, J Soltesz, I AF Strausz, Janos Soltesz, Ibolya TI Bronchial Capillary Hemangioma in Adults SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE hemangioma; bronchus; hemoptysis ID hemangioma; bronchus; hemoptysis AB Two cases with capillary hemangioma of the trachea and the left upper lobe bronchus are presented. The adult patients were referred to the hospital because of hemoptysis and cough. The chest radiographs were normal in both cases. The bronchoscopic examination revealed circumscribed lesions with a capillarized surface protruding into the lumen of the trachea and the left upper lobe bronchus, respectively. The lesions were excised in toto with flexible bronchoscopic forceps. The specimens contained typical capillary hemangiomas without any signs of malignancy. Capillary hemangioma in the bronchial tree is an extremely rare benign lesion in adults. Nevertheless, it should be considered as a possible cause of hemoptysis and cough. C1 [Strausz, Janos] National Koranyi Institute of Pulmonology, Piheno u. l., H-1529 Budapest, Hungary. [Soltesz, Ibolya] National Koranyi Institute of Pulmonology, Piheno u. l., H-1529 Budapest, Hungary. RP Strausz, J (reprint author), National Koranyi Institute of Pulmonology, H-1529 Budapest, Hungary. EM str12196@helka.iif.hu CR Dail DH: Uncommon Tumors. ln: Dail DH, Hammar SP, Eds. Pulmonary Pathology. Springer-Verlag, New York, 847-972, 1988. Franks R, Rothera M: Cardiopulmonary bypass for resection of low tracheal haemangioma. Arch Dis Child 65:630-632, 1990. Harding IR, Williams J, Seal RM: Pedunculated capillary haemangioma of the bronchus. Br J Dis Chest 72:336-342, 1978. Mori S: Sclerosing hemangioma of the lung. Dis Chest 54:71-74, 1968. Paul KP, Borner C, Muller KM, et al: Capillary hemangioma of the right main bronchus treated by sleeve resection in infancy. Am Rev Respir Dis 143:876-879, 1991. Prakash UBS, Freitag L: Hemoptysis and Bronchoscopy-Induced Hemorrhage. In: Prakash UBS, Ed. Bronchoscopy. Raven Press New York. 227-251, 1994. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 233 EP 234 PG 2 ER PT J AU Chuang, ShS Lin, ChN Chu, ChH Chen, FF AF Chuang, Shih-Sung Lin, Ching-Nan Chu, Chien-Hui Chen, Fen-Fen TI Small Cell Carcinoma of the Gallbladder: Report of Two Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE gallbladder; small cell carcinoma; endocrine cell carcinoma; neuroendocrine carcinoma ID gallbladder; small cell carcinoma; endocrine cell carcinoma; neuroendocrine carcinoma AB Two Taiwanese patients with gallbladder small cell carcinoma are reported. One is a 79 year-old male, the other, a 86 year-old female. They both presented with the symptom/signs of acute cholecystitis and underwent cholecystectomy. An intramural mass in the gallbladder neck region was found in the first patient, while the second patient had a transmural indurated tumor in the gallbladder body with extension to the neck region. Characteristic histological and immunohistochemical features of small cell carcinoma were present in both, and electron dense neurosecretory granules were identified in the second. To our knowledge, the second patient is the oldest ever reported. The first patient received chemotherapy directed toward the initial erroneous diagnosis of non-Hodgkin’s lymphoma and developed liver metastasis in two months. The second patient did not receive chemotherapy due to her poor general condition and local recurrence occurred in six weeks. Both passed away three and five months after surgery, respectively. C1 [Chuang, Shih-Sung] Chi-Mei Foundation Hospital, Department of Pathology, 901, Chung Hwa Road, 710 Yung Kang City, Tainan county, Taiwan, Republic of China. [Lin, Ching-Nan] Chi-Mei Foundation Hospital, Department of Pathology, 901, Chung Hwa Road, 710 Yung Kang City, Tainan county, Taiwan, Republic of China. [Chu, Chien-Hui] Sin-Lau Christian Hospital, Department of SurgeryTaichung, Taiwan, Republic of China. [Chen, Fen-Fen] National Cheng-Kung University Hospital, Department of PathologyTainan, Taiwan, Republic of China. RP Chuang, ShS (reprint author), Chi-Mei Foundation Hospital, Department of Pathology, 710 Yung Kang City, Taiwan, Republic of China. EM ssc6061@msl6.hinet.net CR Tahara E: Endocrine tumors of the gastrointestinal tract: classification, function and biological behavior. Digest Dis Pathol 1:121-47, 1988. Yamamoto M, Nakajo S, Miyoshi N, et al: Endocrine cell carcinoma, carcinoid, of the gallbladder. Am J Surg Pathol 13:292- 302, 1989. Ron IG, Wigler N, Ilie B, et al: Small cell carcinoma of the gallbladder: clinical course and response to chemotherapy. Tumori 78:207-210, 1992. Albores-Saavedra J, Molberg K, Henson DE: Unusual malignant epithelial tumors of the gallbladder. Semin Diagno Pathol.. 13:326-338, 1996. Nishigami T, Yamada M, Nakasho K, et al: Carcinoid tumor of the gall bladder. Int Med 35:953-956, 1996. Cavazzana AO, Fassina AS, Tollot M, et al: Small-cell carcinoma of the gallbladder: An immunocytochemical and ultrastructural study. Pathol Res Pract 187:472-476, 1991. Nishihara K, Nagai E, Tsuneyoshi M, et al: Small cell carcinoma combined with adenocarcinoma of the gallbladder. Arch Pathol Lab Med. 118:177-181, 1994. Muraina UI, Tank R, Dhingra C, et al: Small cell carcinoma of gallbladder: report of two cases. Am J Gastroenterol 91:792- 794, 1996. Albores-Saavedra J, Cruz-Ortiz H, Alcantara-Yazques A, et al: Unusual types of gallbladder carcinoma. Arch Pathol Lab Med 105:287-293, 1981. Cavazzana AO, Fassina AS, Tollot M, et al: Small cell carcinoma of gallbladder. An immunohistochemical and ultrastructural study. Path Res Pract 187:472-476, 1991. Cox WF Jr., Pierce GB: The endodermal origin of the endocrine cell of an adenocarcinoma of the colon of the rat. Cancer 50: 1530-1538, 1982. Nash SV, Said JW: Gastropancreatic neuroendocrine tumors. A histochemical and immunohistochemical study of epithelial, keratin proteins, carcinoembryonic antigen, and neuroendocrine, neuron specific enolase, bombesin and chromogranin, markers in foregut, midgut and hindgut tumors. Am J Clin Pathol 86:415- 22, 1986. Mosnier JF, Brousse N, Sevestre C, et al: Primary low-grade Bcell lymphoma of the mucosa-associated lymphoid tissue arising in the gallbladder. Histopathology 1992; 20:273-275, 1992. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 235 EP 238 PG 4 ER PT J AU Mandoky, L AF Mandoky, Laszlo TI Amphicrine Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE amphicrine; adenocarcinoid; neuroendocrine; mucus granules; dense-core granules ID amphicrine; adenocarcinoid; neuroendocrine; mucus granules; dense-core granules AB The term amphicrine refers to cells, and tumors, which show both exocrine and endocrine features. Author’s aim was to analyse the characteristics of these neoplasms. 40 suspicious cases were reviewed. Mucin-stains (PAS, diastase-PAS, Stains-all, Alcian-blue), immunohistochemistry (antibodies against Neuron-Specific Enolase (NSE), and Chromogranin A (CGA), and electronmicroscopic studies were performed to demonstrate exocrine and/or endocrine features of the tumor cells. By means of these methods, 16 cases turned out to be amphicrine tumors. Among them, there were 4 sinonasal, 1 bronchial, 1 mediastinal, 8 gastrointestinal and 2 suprarenal gland neoplasms. In connection to the subject, a brief review is given of amphicrine tumor, regarding its etiological and pathological aspects. These tumors form a distinct clinicopathological entity and should be separated from both neuroendocrine tumors and adenocarcinomas. C1 [Mandoky, Laszlo] Fovarosi Uzsoki utcai Korhaz, Pathologia, Uzsoki utca 29., H-1145 Budapest, Hungary. RP Mandoky, L (reprint author), Fovarosi Uzsoki utcai Korhaz, Pathologia, H-1145 Budapest, Hungary. EM mandlasz@mail.matav.hu CR Al-Talib RK, Mason CH, and Theaker JM: Combined goblet cell carcinoid and mucinous cystadenoma of the appendix. J Clin Pathol 48:869-870, 1995. Andrew A: A study of the development relationships between enterochromaffin cells and the neural crest. J Embryol Exp Morphol 11:307-324, 1963. Azzopardi JG, and Hou LT: Intestinal metaplasia with argentaffin cell in cervical adenocarcinoma. J Pathol Bact 90:686-690, 1965. Borch K, Renvall H, Kullman E, et al: Gastric carcinoid associated with the syndrome of hypergastrinemic atrophic gastritis. A prospective analysis of 11 cases. Am J Surg Pathol 11:435- 444, 1987. Borchard FZ: Das diffuse endokrine System und die Karzinoide des Gastro-intestinaltraktes. Gastroenterologie 20:187-205, 1982. Bordi C, D`Addo T, Pilato FP, et al: Carcinoid, ECL cell, tumor of the oxynic mucosa of the stomach: a hormone dependent neoplasm? In: Progress in surgical pathology., Eds: Fenoglio- Preister C, Wolff M, and Rilke F, Vol 8. Philadelphia: Field & Wood, 1988, pp. 177-195. De Bruine AP, Dinjens WNM, Zijlema JH L, et al: Renewal of enterochromaffin cells in the rat caecum. Anat Rec 233:75-82, 1992b. De Bruine A P, Wiggers Th, Beek C et al.: Endocrine cells in colorectal adenocarcinomas: incidence, hormone profile and prognostic relevance. Int J Cancer 54:765-771, 1993. Capella C, Eusebi V, Mann B, et al: Endocrine differentiation in mucoid carcinoma of the breast. Hystopathol 4:613-630, 1980. Capella C, Usellini L, Papotti M, et al.: Ultrastructural Features of Neuroendocrine Differentiated Carcinomas of the Breast. Ultrastruct Pathol 14:321-334, 1990. Chejfec C, Falkmer S, Askensten U, et al: Neuroendocrine tumors of the gastrointestinal tract. Pathol Res Pract 183:143-154, 1988. Cox WF Jr., and Pierce B: The endodermal origine of the endocrine cells of an adenocarcinoma of the colon of the rat. Cancer 50:1530-1538, 1982. Eusebi V, Mambelli V, Tison V, et al.: Endocrine differentation in basal cell carcinoma. Tumori 65:191-199, 1979. Faverly DRGS, Manni JJ, Smedts, et al.: Adeno-carcinoid or Amphicrine Tumors of the Middle Ear a New Entity? Path Res Pract 188:162-171, 1992. Feyrter F: Uber diffuse endokrine epitheliale Organe. Johann Ambrosius Barth, Leipzig, 1938. Fontaine J, and LeDouarin NM: Analysis of endoderm formation in the avian blastoderm by the use of quail-chick chimaeras. The problem of the neuroectodermal origin of the cells of the APUD series. J Embryol Exp Morphol 41:209-222, 1977. Golouh R, Us-Krasovec M, Auersperg M, et al.: Amphicrine — Composite Calcitonin and Mucin-Producing – Carcinoma of the Thyroid. Ultrastruct Pathol 8:197-206, 1985. Gouzi J-L, Laigneau P, Delalande J-P, et al.: Indications for right hemicolectomy in carcinoid tumors of the appendix. Surg Gynecol Obstet 176:543-547, 1993. Grace C, Yang H, Rotterdam H: Mixed, composite, Gladular- Endocrine Cell Carcinoma of the Stomach. Am J Surg Pathol. 15:592-598, 1991. Hidvegi J, Zsolnai B, Szinay G: Amphicrine Tumor of the Vulva. Path Res Pract 183:505-508, 1988. Hirt RS, Evans LD, Buroker RA, Oleson FB: Gastric enterochromaffin- like cell hyperplasia and neoplasia in the rat: an indirect effect of the histamine H2-antagonist BL-6341. Toxicol Pathol 16:273-287, 1988. Itsuno M, Watanabe H, Iwafuchi M, et al.: Multiple carcinoids and endocrine cell micronests in type A gastritis. Their morphology, histogenesis, and natural history. Cancer. 63:881-890, 1989. Kyung-Whan Min: Usefulness of Electron Microscopy in the Diagnosis of “Small” Round Cell Tumors of the Sinonasal Region. Ultrastruct Pathol 19:347-363, 1995. Laine VJO, Ekfors TO, Gullichsen R, et al: Immunohistochemical characterization of an amphicrine mucinous islet-cell carcinoma of the pancreas. APMIS 100:335-340, 1992. Lewin KJ: Carcinoid tumors and the mixed, composite, glandular- endocrine cell carcinomas. Am J Surg Pathol 11(Suppl 1): 71-86, 1987. Mendelsohn G, De La Monte S, Dunn JL, et al: Gastric carcinoid tumors, endocrine cell hyperplasia, and associated intestinal metaplasia. Histologic, histochemical, and immunohistochemical findings. Cancer 60:1022-1031, 1987. Muller K-M, Fisseler-Eckhoff A: What`s New in Lung Tumor Heterogeneity? Path.Res.Pract. 184:108-115, 1989. Ordonez NG, Balsaver AM, Mackay B: Mucinous islet cell, amphicrine, carcinoma of the pancreas associated with watery diarrhea and hypokalaemia syndrome. Hum Pathol 19:1458- 1461, 1988. Paladugu R, Nathwani B, Goodstein J, et al: Carcinoma of the larynx with mucosubstance production and neuroendocrine differentiation. Cancer 42:343-349, 1982. Papadimitriou JC, Weihing RR, Choi C, et al: Prostatic Marker- Negative Amphicrine Carcinoma of the Prostate. Ultrastruct Pathol 18:357-363, 1994. Pasquinelli G, Santini D, Preda P, et al.: Composite gastric carcinoma and precursor lesions with amphicrine features in chronic atrophic gastritis. Ultrastruct Pathol 17:9-24, 1993. Ratzenhofer M: Uber enterale hyperplasien und Geschwulste der disseminerten endokrinen, parakrinen, Hellen Zellen Feyrters unter Berucksichtigung amphikriner Zellwucherungen. Verh Dtsch Ges Pathol. Tag Erlangen, 61:7-24, 1977. Ratzenhofer M, Aubock L: The amphicrine, endo-exocrine, cells in the human gut, with a short reference to amphicrine neoplasias. Acta Morphologica Acad Sci Hung 28:37-58, 1980. Poynter D, Pick CR, Harcourt RA, et al: Association of long lasting unsur-mountable histamine H2 blockade and gastric carcinoid tumors in the rat. Gut. 26:1284-1295, 1985. Sakamoto F, Ito M, Matumura G, et al.: Ultrastructural study of a mucinous carcinoid of the skin J Cutan Pathol 18:128-133, 1991. Schmid KO, Aubock L, Albegger K: Endocrine-Amphicrine Enteric Carcinoma of the Nasal Mucosa. Virchows Arch A Path Anat Histol 383:329-343, 1979. Sheppard MN, Thurlow NP, Dewar A: Amphicrine Differentiation in Bronchiolo-alveolar Cell Carcinoma. Ultrastruct Pathol 18:437-441, 1994. Sidhu GS: The endodermal origin of digestive and respiratory tract APUD cells. Am J Pathol 96:5-20, 1979. Sjoblom SM, Sipponen P, Karonen SL, et al: Mucosal argyrophil endocrine cells in pernicious anaemia and upper gastrointestinal carcinoid tumors. J Clin Pathol 42:371-377, 1989. Soga J, Tazawa K: Pathologic analysis of carcinoid. Histologic re-evaluation of 62 cases. Cancer 28:990-998, 1971. Solcia E, Capella C, Sessa F, et al: Gastric carcinoids and related endocrine growths. Digestion. 35(Suppl): 3-22, 1986. Solcia E, Bordi C, Creutzfeldt W, et al: Histopathological classification of non-antral gastric endocrine growths in man. Digestion. 41:185-200, 1988. Solcia E, Rindi G, Fiocca R, et al: Distinct patterns of chronic gastritis associated with carcinoid and cancer and their role in tumorigenesis. Yale J Biol Med. 65:793-804, 1992. Wassef M, Kanavaros P, Polivka M, et al: Middle Ear Adenoma – A Tumor Displaying Mucinous and Neuroendocrine Differentiation. Am J Pathol 13:838-847, 1989. Watson PH, Alguacil-Garcia A: Mixed crypt cell carcinoma. A clinicopathological study of the so-called ”Goblet cell carcinoid” Virchows Arch A 412:175-182, 1987. Wisniewski M, Toker C: Composite tumor of the gallbladder exhibiting both carcinomatous and carcinoidal patterns. Am J Gastroenterol 58:633-637, 1972. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 1999 VL 5 IS 3 BP 239 EP 244 PG 6 ER PT J AU Nagy, P Jenei, A Damjanovich, S Jovin, MTh Szolosi, J AF Nagy, Peter Jenei, Attila Damjanovich, Sandor Jovin, M Thomas Szolosi, Janos TI Complexity of Signal Transduction Mediated by ErbB2: Clues to the Potential of Receptor-Targeted Cancer Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE erbB proteins; erbB2; homoassociation; heteroassociation; breast cancer; Herceptin ID erbB proteins; erbB2; homoassociation; heteroassociation; breast cancer; Herceptin AB The erbB2 oncogene belongs to the type I trans-membrane tyrosine kinase family of receptors. Its medical importance stems from its widespread over-expression in breast cancer. This review will focus on the signal transduction through this protein, and explains how the overexpression of erbB2 may result in poor prognosis of breast cancer, and finally it will summerize our current understanding about the therapeutic potential of receptor-targeted therapy in breast cancer. ErbB2 does not have any known ligand which is able to bind to it with high affinity. However the kinase activity of erbB2 can be activated without any ligand, if it is overexpressed, and by heteroassociation with other members of the erbB family (erbB1 or epidermal growth factor receptor, erbB3 and erbB4). This interaction substantially increases the efficiency and diversity of signal transduction through these receptor complexes. In addition, erbB2 forms large scale receptor clusters containing hundreds of proteins. These receptor islands may take part in recruiting cytosolic factors which relay the signal towards the nucleus or the cytoplasm. Overexpression of erbB2 was linked to higher transforming activity, increased metastatic potential, angiogenesis and drug resistence of breast tumor in laboratory experiments. As a corollary of these properties, erbB2 amplification is generally thought to be associated with a poor prognosis in breast cancer patients. These early findings lead to the development of antibodies that down-regulate erbB2. Such a therapeutic approach has already been found effective in experimental tumor models and in clinical trials as well. Further understanding of the importance of erbB2 and growth factor receptors in the transformation of normal cells to malignant ones may once give us a chance to cure erbB2 over-expressing breast cancer. C1 [Nagy, Peter] University Medical School of Debrecen, Department of Biophysics and Cell Biology, H4012 Debrecen, Hungary. [Jenei, Attila] University Medical School of Debrecen, Department of Biophysics and Cell Biology, H4012 Debrecen, Hungary. [Damjanovich, Sandor] University Medical School of Debrecen, Department of Biophysics and Cell Biology, H4012 Debrecen, Hungary. [Jovin, M Thomas] Max Planck Institute for Biophysical Chemistry, Department of Molecular BiologyGottingen, Germany. [Szolosi, Janos] University Medical School of Debrecen, Department of Biophysics and Cell Biology, H4012 Debrecen, Hungary. RP Nagy, P (reprint author), University Medical School of Debrecen, Department of Biophysics and Cell Biology, H4012 Debrecen, Hungary. CR Ahmad I, Longenecker M, Samuel J: Antibody-targeted delivery of doxorubicin entrapped in sterically stabilized liposomes can eradicate lung cancer in mice. Cancer Res 53:1484-1488, 1993. Alimandi M, Romano A, Curia MC, et al: Cooperative signaling of ErbB3 and ErbB2 in neoplastic transformation and human mammary carcinomas. Oncogene 10: 1813-1821, 1995. Allred DC, Clark GM, Molina R, et al: Overexpression of HER-2/neu and its relationship with other prognostic factors change during the progression of in situ to invasive breast cancer. Hum Pathol 23:974-979, 1992. Allred DC, O’Connell P, Fuqua SA: Biomarkers in early breast neoplasia. J Cell Biochem Suppl 17G:125-131, 1993. Andrulis IL, Bull SB, Blackstein ME, et al. neu/erbB-2 amplification identifies a poor-prognosis group of women with node-negative breast cancer. Toronto Breast Cancer Study Group. J Clin Oncol 16:1340-1349, 1998. Bacus SS, Gudkov AV, Zelnick CR, et al: Neu differentiation factor, heregulin, induces expression of intercellular adhesion molecule 1: implications for mammary tumors. Cancer Res 53:5251-5261, 1993. Bacus SS, Huberman E, Chin D, et al.: A ligand for the erbB- 2 oncogene product, gp30, induces differentiation of human breast cancer cells. Cell Growth Differ 3:401-411, 1992. Bacus SS, Stancovski I, Huberman E, et al: Tumor-inhibitory monoclonal antibodies to the HER-2/Neu receptor induce differentiation of human breast cancer cells. Cancer Res 52:2580- 2589, 1992. Bacus SS, Yarden Y, Oren M, et al: Neu differentiation factor, Heregulin, activates a p53-dependent pathway in cancer cells. Oncogene 12:2535-2547, 1996. Bargmann CI, Weinberg RA: Increased tyrosine kinase activity associated with the protein encoded by the activated neu oncogene. Proc Natl Acad Sci USA 85:5394-5398, 1988. Barnes DM: c-erbB-2 amplification in mammary carcinoma. J.Cell Biochem Suppl 17G:132-138, 1993. Baselga J, Norton L, Albanell J et al: Recombinant humanized anti-HER2 antibody, Herceptin, enhances the antitumor activity of paclitaxel and doxorubicin against HER2/neu overexpressing human breast cancer xenografts. Cancer Res 58:2825- 2831, 1998. Baselga J, Seidman AD, Rosen PP, et al: HER2 overexpression and paclitaxel sensitivity in breast cancer: therapeutic implications. Oncology Huntingt 11:43-48, 1997. Baselga J, Tripathy D, Mendelsohn J, et al: Phase II study of weekly intravenous recombinant humanized anti- p185HER2 monoclonal antibody in patients with HER2/neu- overexpressing metastatic breast cancer. J Clin Oncol 14:737-744, 1996. Baulida J, Kraus MH, Alimandi M, et al: All ErbB receptors other than the epidermal growth factor receptor are endocytosis impaired. J Biol Chem 271:5251-5257, 1996. Beerli RR, Hynes NE: Epidermal growth factor-related peptides activate distinct subsets of ErbB receptors and differ in their biological activities. J Biol Chem 271: 6071-6076, 1996. Benz CC, Scott GK, Sarup JC, et al: Estrogen-dependent, tamoxifen- resistant tumorigenic growth of MCF-7 cells transfected with HER2/neu. Breast Cancer Res Treat 24:85-95, 1993. Berns EM, Foekens JA, van Staveren IL, et al: Oncogene amplification and prognosis in breast cancer: relationship with systemic treatment. Gene 159:11-18, 1995. Bianco AR, De Laurentiis M, Carlomagno C: 20 year update of the Naples Gun trial of adjuvant breast cancer therapy: evidence of interaction between c-erb-B2 expression and tamoxifene efficacy. Proc Am Soc Clin Oncol 17:97a1998. Bourguignon LY, Zhu H, Chu A, et al: Interaction between the adhesion receptor, CD44, and the oncogene product, p185HER2, promotes human ovarian tumor cell activation. J Biol Chem 272:27913-27918, 1997. Brower ST, Ahmed S, Tartter PI, et al: Prognostic variables in invasive breast cancer: contribution of comedo versus noncomedo in situ component. Ann Surg Oncol 2:440-444, 1995. Burke CL, Stern DF: Activation of Neu, ErbB-2, mediated by disulfide bond-induced dimerization reveals a receptor tyrosine kinase dimer interface. Mol Cell Biol 18:5371-5379, 1998. Campiglio M, Tagliabue E, Srinivas U, et al: Colocalization of the p185HER2 oncoprotein and integrin alpha 6 beta 4 in Calu- 3 lung carcinoma cells. J Cell Biochem 55:409-418, 1994. Canman CE, Gilmer TM, Coutts SB, et al: Growth factor modulation of p53-mediated growth arrest versus apoptosis. Genes Dev 9:600-611, 1995. Carraway KL, Sliwkowski MX, Akita R et al: The erbB3 gene product is a receptor for heregulin. J Biol Chem 269:14303- 14306, 1994. Chakrabarti A, Matko J, Rahman NA, et al: Self-association of class I major histocompatibility complex molecules in liposome and cell surface membranes. Biochemistry 31:7182-7189, 1992. Chamberlin SG, Davies DE: A unified model of c-erbB receptor homo- and heterodimerisation. Biochim Biophys Acta 1384:223-232, 1998. Chen LI, Webster MK, Meyer AN, et al: Transmembrane domain sequence requirements for activation of the p185c-neu receptor tyrosine kinase. J Cell Biol 137:619-631, 1997. Cohen BD, Kiener PA, Green JM, et al: The relationship between human epidermal growth-like factor receptor expression and cellular transformation in NIH3T3 cells. J Biol Chem 271:30897-30903, 1996. Crovello CS, Lai C, Cantley LC, et al: Differential signaling by the epidermal growth factor-like growth factors neuregulin-1 and neuregulin-2. J Biol Chem 273:26954-26961, 1998. D’Souza B, Berdichevsky F, Kyprianou N, et al: Collageninduced morphogenesis and expression of the alpha 2-integrin subunit is inhibited in c-erbB2-transfected human mammary epithelial cells. Oncogene 8:1797-1806, 1993. D’Souza B, Taylor Papadimitriou J: Overexpression of ERBB2 in human mammary epithelial cells signals inhibition of transcription of the E-cadherin gene. Proc Natl Acad Sci USA 91:7202-7206, 1994. Dalifard I, Daver A, Goussard J, et al: p185 overexpression in 220 samples of breast cancer undergoing primary surgery: comparison with c-erbB-2 gene amplification. Int J Mol Med 1:855-861, 1998. Daly JM, Jannot CB, Beerli RR et al: Neu differentiation factor induces ErbB2 down-regulation and apoptosis of ErbB2-overexpressing breast tumor cells. Cancer Res 57:3804-3811, 1997. Damjanovich S, Vereb G, Schaper A, et al: Structural hierarchy in the clustering of HLA class I molecules in the plasma membrane of human lymphoblastoid cells. Proc Natl Acad Sci USA 92:1122-1126, 1995. De Corte V, De Potter C, Vandenberghe D, et al: A 50 kDa protein present in conditioned medium of COLO-16 cells stimulates cell spreading and motility, and activates tyrosine phosphorylation of Neu/HER-2, in human SK-BR-3 mammary cancer cells. J Cell Sci 107:405-416, 1994. De Potter CR, Quatacker J: The p185erbB2 protein is localized on cell organelles involved in cell motility. Clin Exp Metastasis 11:453-461, 1993. Di Fiore PP, Segatto O, Lonardo F, et al: The carboxy-terminal domains of erbB-2 and epidermal growth factor receptor exert different regulatory effects on intrinsic receptor tyrosine kinase function and transforming activity. Mol Cell Biol 10:2749-2756, 1990. Discafani CM, Carroll ML, Floyd MB, Jr. et al: Irreversible inhibition of epidermal growth factor receptor tyrosine kinase with in vivo activity by N-[4-[(3-bromophenyl)amino]-6-quinazolinyl]- 2-butynamide, CL-387,785). Biochem Pharmacol 57:917-925, 1999. Disis ML, Grabstein KH, Cheaver MA: HER2/neu peptide vaccines elicit T cell immunity to the HER-2/neu protein in patients with breast and ovarian cancer. Proc Am Soc Clin Oncol 17:97a1998. Drebin JA, Link VC, Stern DF, et al: Down-modulation of an oncogene protein product and reversion of the transformed phenotype by monoclonal antibodies. Cell 41:697-706, 1985. Dykins R, Corbett IP, Henry JA et al: Long-term survival in breast cancer related to overexpression of the c-erbB-2 oncoprotein: an immunohistochemical study using monoclonal antibody NCL-CB11. J Pathol 163:105-110, 1991. Edidin M: Lipid microdomains in cell surface membranes. Curr Opin Struct Biol 7:528-532, 1997. Engelman JA, Lee RJ, Karnezis A, et al: Reciprocal regulation of neu tyrosine kinase activity and caveolin-1 protein expression in vitro and in vivo. Implications for human breast cancer. J Biol Chem 273: 20448-20455, 1998. Ethier SP, Langton BC, Dilts CA: Growth factor-independent proliferation of rat mammary carcinoma cells by autocrine secretion of neu-differentiation factor/heregulin and transforming growth factor-alpha. Mol Carcinog 15:134-143, 1996. Falcioni R, Antonini A, Nistico P, et al: Alpha 6 beta 4 and alpha 6 beta 1 integrins associate with ErbB-2 in human carcinoma cell lines. Exp Cell Res 236:76-85, 1997. Fan Z, Baselga J, Masui H, et al: Antitumor effect of anti-epidermal growth factor receptor monoclonal antibodies plus cisdiamminedichloroplatinum on well established A431 cell xenografts. Cancer Res 53:4637-4642, 1993. Fendly BM, Winget M, Hudziak RM, et al: Characterization of murine monoclonal antibodies reactive to either the human epidermal growth factor receptor or HER2/neu gene product. Cancer Res 50: 1550-1558, 1990. French AR, Tadaki DK, Niyogi SK, et al: Intracellular trafficking of epidermal growth factor family ligands is directly influenced by the pH sensitivity of the receptor/ligand interaction. J Biol Chem 270:4334-4340, 1995. Gadella TW, Jr., Jovin TM: Oligomerization of epidermal growth factor receptors on A431 cells studied by time-resolved fluorescence imaging microscopy. A stereochemical model for tyrosine kinase receptor activation. J Cell Biol 129:1543-1558, 1995. Gamett DC, Pearson G, Cerione RA, et al: Secondary dimerization between members of the epidermal growth factor receptor family. J Biol Chem 272:12052-12056, 1997. Gassmann M, Casagranda F, Orioli D, et al: Aberrant neural and cardiac development in mice lacking the ErbB4 neuregulin receptor. Nature 378:390-394, 1995. Giani C, Casalini P, Pupa SM, et al: Increased expression of cerbB- 2 in hormone-dependent breast cancer cells inhibits cell growth and induces differentiation. Oncogene 17:425-432, 1998. Graus Porta D, Beerli RR, Daly JM, et al: ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J 16:1647-1655, 1997. Graus Porta D, Beerli RR, Hynes NE: Single-chain antibodymediated intracellular retention of ErbB-2 impairs Neu differentiation factor and epidermal growth factor signaling. Mol Cell Biol 15:1182-1191, 1995. Gronowski AM, Bertics PJ: Modulation of epidermal growth factor receptor interaction with the detergent-insoluble cytoskeleton and its effects on receptor tyrosine kinase activity. Endocrinology 136: 2198-2205, 1995. Grunt TW, Saceda M, Martin MB, et al: Bidirectional interactions between the estrogen receptor and the cerbB-2 signaling pathways: heregulin inhibits estrogenic effects in breast cancer cells. Int J Cancer 63:560-567, 1995. Gullick WJ: Prevalence of aberrant expression of the epidermal growth factor receptor in human cancers. Br Med Bull 47:87-98, 1991. Gusterson BA, Gelber RD, Goldhirsch A, et al: Prognostic importance of c-erbB-2 expression in breast cancer. International, Ludwig, Breast Cancer Study Group. J Clin Oncol 10:1049-1056, 1992. Haerslev T, Jacobsen GK: c-erbB-2 oncoprotein is not an independent prognostic parameter in primary breast carcinoma. An immunohistochemical study. APMIS 102:612-622, 1994. Harbeck N, Dettmar P, Thomssen C, et al: Prognostic impact of tumor biological factors on survival in node-negative breast cancer. Anticancer Res 18:2187-2197, 1998. Harder T, Simons K: Caveolae, DIGs, and the dynamics of sphingolipid-cholesterol microdomains. Curr Opin Cell Biol 9:534-542, 1997. Hartmann F, Horak EM, Cho C, et al: Effects of the tyrosinekinase inhibitor geldanamycin on ligand-induced Her-2/neu activation, receptor expression and proliferation of Her-2-positive malignant cell lines. Int J Cancer 70:221-229, 1997. Heldin CH: Dimerization of cell surface receptors in signal transduction. Cell 80:213-223, 1995. Hellyer NJ, Cheng K, Koland JG: ErbB3, HER3, interaction with the p85 regulatory subunit of phosphoinositide 3-kinase. Biochem J 333:757-763, 1998. Houston SJ, Plunkett TA, Barnes DM,et al: Overexpression of cerbB2 is an independent marker of resistance to endocrine therapy in advanced breast cancer. Br J Cancer 79:1220-1226, 1999. Hudziak RM, Lewis GD, Winget M, et al: p185HER2 monoclonal antibody has antiproliferative effects in vitro and sensitizes human breast tumor cells to tumor necrosis factor. Mol Cell Biol 9:1165-1172, 1989. Hwang J, Gheber LA, Margolis L, et al: Domains in cell plasma membranes investigated by near-field scanning optical microscopy. Biophys J 74:2184-2190, 1998. Hynes NE, Stern DF: The biology of erbB-2/neu/HER-2 and its role in cancer. Biochim Biophys Acta 1198:165-184, 1994. Kallioniemi OP, Holli K, Visakorpi T, et al: Association of cerbB- 2 protein over-expression with high rate of cell proliferation, increased risk of visceral metastasis and poor long-term survival in breast cancer. Int J Cancer 49:650-655, 1991. Kameda T, Yasui W, Yoshida K, et al: Expression of ERBB2 in human gastric carcinomas: relationship between p185ERBB2 expression and the gene amplification. Cancer Res 50:8002- 8009, 1990. Karunagaran D, Tzahar E, Beerli RR, et al: ErbB-2 is a common auxiliary subunit of NDF and EGF receptors: implications for breast cancer. EMBO J 15:254-264, 1996. Kenworthy AK, Edidin M: Distribution of a glycosylphosphatidylinositol- anchored protein at the apical surface of MDCK cells examined at a resolution of <100 A using imaging fluorescence resonance energy tranfer. J Cell Biol 142:69- 84, 1998. Kim HH, Vijapurkar U, Hellyer NJ, et al: Signal transduction by epidermal growth factor and heregulin via the kinase-deficient ErbB3 protein. Biochem J 334:189-195, 1998. Kirpotin D, Park JW, Hong K, et al: Sterically stabilized anti- HER2 immunoliposomes: design and targeting to human breast cancer cells in vitro. Biochemistry 36:66-75, 1997. Klapper LN, Vaisman N, Hurwitz E, et al: A subclass of tumorinhibitory monoclonal antibodies to ErbB-2/HER2 blocks crosstalk with growth factor receptors. Oncogene 14:2099-2109, 1997. Knowlden JM, Gee JM, Seery L, et al: c-erbB3 and c-erbB4 expression is a feature of the endocrine responsive phenotype in clinical breast cancer. Oncogene 17:1949-1957, 1998. Kraus MH, Issing W, Miki T, et al: Isolation and characterization of ERBB3, a third member of the ERBB/epidermal growth factor receptor family: evidence for overexpression in a subset of human mammary tumors. Proc Natl Acad Sci USA 86:9193-9197, 1989. Kuan CT, Pastan I: Recombinant immunotoxin containing a disulfide-stabilized Fv directed at erbB2 that does not require proteolytic activation. Biochemistry 35:2872-2877, 1996. Lee KF, Simon H, Chen H, et al: Requirement for neuregulin receptor erbB2 in neural and cardiac development. Nature 378:394-398, 1995. Lemmon MA, Bu Z, Ladbury JE, et al: Two EGF molecules contribute additively to stabilization of the EGFR dimer. EMBO J 16:281-294, 1997. Lemmon MA, Schlessinger J: Regulation of signal transduction and signal diversity by receptor oligomerization. Trends Biochem Sci 19:459-463, 1994. Levkowitz G, Klapper LN, Tzahar E, et al: Coupling of the c- Cbl protooncogene product to ErbB-1/EGF-receptor but not to other ErbB proteins. Oncogene 12:1117-1125, 1996. Lewis GD, Figari I, Fendly B, et al: Differential responses of human tumor cell lines to anti-p185HER2 monoclonal antibodies. Cancer Immunol Immunother 37:255-263, 1993. Li W, Park JW, Nuijens A, et al: Heregulin is rapidly translocated to the nucleus and its transport is correlated with c-myc induction in breast cancer cells. Oncogene 12:2473-2477, 1996. Lipponen HJ, Aaltomaa S, Syrjanen S, et al: c-erbB-2 oncogene related to p53 expression, cell proliferation and prognosis in breast cancer. Anticancer Res 13:1147-1152, 1993. Liu X, Pogo BG: Inhibition of erbB-2-positive breast cancer cell growth by erbB-2 antisense oligonucleotides. Antisense Nucleic Acid Drug Dev 6:9-16, 1996. Liu Y, Martindale JL, Gorospe M, et al: Regulation of p21WAF1/CIP1 expression through mitogen-activated protein kinase signaling pathway. Cancer Res 56:31-35, 1996. Lofts FJ, Hurst HC, Sternberg MJ, et al: Specific short transmembrane sequences can inhibit transformation by the mutant neu growth factor receptor in vitro and in vivo. Oncogene 8:2813-2820, 1993. Lupu R, Colomer R, Kannan B, et al: Characterization of a growth factor that binds exclusively to the erbB-2 receptor and induces cellular responses. Proc Natl Acad Sci USA 89:2287- 2291, 1992. Marte BM, Jeschke M, Graus Porta D,et al: Neu differentiation factor/heregulin modulates growth and differentiation of HC11 mammary epithelial cells. Mol Endocrinol 9:14-23, 1995. McNeil C: Herceptin raises its sights beyond advanced breast cancer. J Natl Cancer Inst 90:882-883, 1998. Meyer D, Birchmeier C: Multiple essential functions of neuregulin in development. Nature 378:386-390, 1995. Murali R, Brennan PJ, Kieber Emmons T, et al: Structural analysis of p185c-neu and epidermal growth factor receptor tyrosine kinases: oligomerization of kinase domains. Proc Natl Acad Sci USA 93:6252-6257, 1996. Nagy P, Bene L, Balazs M, et al: EGF-induced redistribution of erbB2 on breast tumor cells: Flow and image cytometric energy transfer measurements. Cytometry 32:120-131, 1998. Nagy P, Jenei A, Kirsch AK, et al: Activation-dependent clustering of the erbB2 receptor tyrosine kinase detected by scanning near-field optical microscopy. J Cell Sci 112:1733-1741, 1999. Niemann C, Brinkmann V, Spitzer E, et al: Reconstitution of mammary gland development in vitro: requirement of c-met and c-erbB2 signaling for branching and alveolar morphogenesis. J Cell Biol 143:533-545, 1998. Norton L: Kinetic concepts in the systemic drug therapy of breast cancer. Semin Oncol 26:11-20, 1999. Nowak F, Jacquemin Sablon A, Pierre J: Expression of the activated p185erbB2 tyrosine kinase in human epithelial cells leads to MAP kinase activation but does not confer oncogenicity. Exp Cell Res 231:251-259, 1997. Olayioye MA, Graus Porta D, Beerli RR, et al: ErbB-1 and ErbB-2 acquire distinct signaling properties dependent upon their dimerization partner. Mol Cell Biol 18:5042-5051, 1998. Ouyang X, Gulliford T, Huang G, et al: Transforming growth factor-alpha short-circuits downregulation of the epidermal growth factor receptor. J Cell Physiol 179:52-57, 1999. Papahadjopoulos D, Allen TM, Gabizon A, et al: Sterically stabilized liposomes: improvements in pharmacokinetics and antitumor therapeutic efficacy. Proc Natl Acad Sci USA 88:11460-11464, 1991. Park JW, Hong K, Carter P : Development of anti-p185HER2 immunoliposomes for cancer therapy. Proc Natl Acad Sci USA 92:1327-1331, 1995. Pegram MD, Finn RS, Arzoo K,et al: The effect of HER-2/neu overexpression on chemotherapeutic drug sensitivity in human breast and ovarian cancer cells. Oncogene 15:537-547, 1997. Pegram MD, Lipton A, Hayes DF, et al: Phase II study of receptor-enhanced chemosensitivity using recombinant humanized anti-p185HER2/neu monoclonal antibody plus cisplatin in patients with HER2/neu-overexpressing metastatic breast cancer refractory to chemotherapy treatment. J Clin Oncol 16:2659-2671, 1998. Peiper M, Goedegebuure PS, Linehan DC, et al: The HER2/neuderived peptide p654-662 is a tumor-associated antigen in human pancreatic cancer recognized by cytotoxic T lymphocytes. Eur J Immunol 27:1115-1123, 1997. Petit AM, Rak J, Hung MC, et al: Neutralizing antibodies against epidermal growth factor and ErbB-2/neu receptor tyrosine kinases down-regulate vascular endothelial growth factor production by tumor cells in vitro and in vivo: angiogenic implications for signal transduction therapy of solid tumors. Am J Pathol 151:1523-1530, 1997. Pietras RJ, Arboleda J, Reese DM, et al: HER-2 tyrosine kinase pathway targets estrogen receptor and promotes hormone- independent growth in human breast cancer cells. Oncogene 10:2435-2446, 1995. Pietras RJ, Fendly BM, Chazin VR, et al: Antibody to HER- 2/neu receptor blocks DNA repair after cisplatin in human breast and ovarian cancer cells. Oncogene 9:1829-1838, 1994. Pietras RJ, Pegram MD, Finn RS, et al: Remission of human breast cancer xenografts on therapy with humanized monoclonal antibody to HER-2 receptor and DNA-reactive drugs. Oncogene 17:2235-2249, 1998. Pinkas Kramarski R, Shelly M, Glathe S, et al: Neu differentiation factor/neuregulin isoforms activate distinct receptor combinations. J Biol Chem 271:19029-19032, 1996. Plowman GD, Culouscou JM, Whitney GS, et al: Ligand-specific activation of HER4/p180erbB4, a fourth member of the epidermal growth factor receptor family. Proc Natl Acad Sci USA 90:1746-1750, 1993. Press MF, Hung G, Godolphin W, et al: Sensitivity of HER- 2/neu antibodies in archival tissue samples: potential source of error in immunohistochemical studies of oncogene expression. Cancer Res 54:2771-2777, 1994. Press MF, Pike MC, Chazin VR, et al: Her-2/neu expression in node-negative breast cancer: direct tissue quantitation by computerized image analysis and association of overexpression with increased risk of recurrent disease. Cancer Res 53:4960- 4970, 1993. Qian X, Dougall WC, Hellman ME, et al: Kinase-deficient neu proteins suppress epidermal growth factor receptor function and abolish cell transformation. Oncogene 9:1507-1514, 1994. Qian X, O’Rourke DM, Fei Z, et al: Domain-specific interactions between the p185(neu, and epidermal growth factor receptor kinases determine differential signaling outcomes. J Biol Chem 274: 574-583, 1999. Quenel N, Wafflart J, Bonichon F, et al: The prognostic value of c-erbB2 in primary breast carcinomas: a study on 942 cases. Breast Cancer Res Treat 35:283-291, 1995. Revillion F, Bonneterre J, Peyrat JP: ERBB2 oncogene in human breast cancer and its clinical significance. Eur J Cancer 34:791-808, 1998. Riethmacher D, Sonnenberg Riethmacher E, et al: Severe neuropathies in mice with targeted mutations in the ErbB3 receptor. Nature 389:725-730, 1997. Ross JS, Fletcher JA: The HER-2/neu oncogene in breast cancer: prognostic factor, predictive factor, and target for therapy. Stem Cells 16:413-428, 1998. Sarup JC, Johnson RM, King K, et al: Characterization of an anti-p185HER2 monoclonal antibody that stimulates receptor function and inhibits tumor cell growth. Growth Regul 1:72- 82, 1991. Schechter AL, Hung MC, Vaidyanathan L, et al: The neu gene: an erbB-homologous gene distinct from and unlinked to the gene encoding the EGF receptor. Science 229:976-978, 1985. Schroeder JA, Lee DC: Dynamic expression and activation of ERBB receptors in the developing mouse mammary gland. Cell Growth Differ 9:451-464, 1998. Shackney SE, A, Smith CA, et al: Intracellular coexpression of epidermal growth factor receptor, Her-2/neu, and p21ras in human breast cancers: evidence for the existence of distinctive patterns of genetic evolution that are common to tumors from different patients. Clin Cancer Res 4:913-928, 1998. Shelly M, Pinkas Kramarski R, Guarino BC, et al: Epiregulin is a potent pan-ErbB ligand that preferentially activates heterodimeric receptor complexes. J Biol Chem 273:10496-10505, 1998. Silletti S, Paku S, Raz A: Tumor cell motility and metastasis. Autocrine motility factor as an example of ecto/exoenzyme cytokines. Pathol Oncol Res 3:230-254, 1997. Simons K, Ikonen E: Functional rafts in cell membranes. Nature 387:569-572, 1997. Sjogren S, Inganas M, Lindgren A, et al: Prognostic and predictive value of c-erbB-2 overexpression in primary breast cancer, alone and in combination with other prognostic markers. J Clin Oncol 16:462-469, 1998. Skarpen E, Johannessen LE, Bjerk K, et al: Endocytosed epidermal growth factor, EGF, receptors contribute to the EGFmediated growth arrest in A431 cells by inducing a sustained increase in p21/CIP1. Exp Cell Res 243:161-172, 1998. Slamon DJ, Clark GM, Wong SG, et al: Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235: 177-182, 1987. Sliwkowski MX, Schaefer G, Akita RW, et al: Coexpression of erbB2 and erbB3 proteins reconstitutes a high affinity receptor for heregulin. J Biol Chem 269:14661-14665, 1994. Soltoff SP, Carraway KL, Prigent SA, et al: ErbB3 is involved in activation of phosphatidylinositol 3-kinase by epidermal growth factor. Mol Cell Biol 14:3550-3558, 1994. Srinivasan R, Poulsom R, Hurst HC, et al: Expression of the c-erbB-4/HER4 protein and mRNA in normal human fetal and adult tissues and in a survey of nine solid tumour types. J Pathol 185:236-245, 1998. Stancovski I, Hurwitz E, Leitner O, et al: Mechanistic aspects of the opposing effects of monoclonal antibodies to the ERBB2 receptor on tumor growth. Proc Natl Acad Sci USA 88:8691- 8695, 1991. Szollosi J, Balazs M, Feuerstein BG,et al: ERBB-2, HER2/neu, gene copy number, p185HER-2 overexpression, and intratumor heterogeneity in human breast cancer. Cancer Res 55:5400- 5407, 1995. Szollosi J, Horejsi V, Bene L, et al: Supramolecular complexes of MHC class I, MHC class II, CD20, and tetraspan molecules, CD53, CD81, and CD82, at the surface of a B cell line JY. J Immunol 157:2939-2946, 1996. Tagliabue E, Centis F, Campiglio M, et al: Selection of monoclonal antibodies which induce internalization and phosphorylation of p185HER2 and growth inhibition of cells with HER2/NEU gene amplification. Int J Cancer 47:933-937, 1991. Tan M, Grijalva R, Yu D: Heregulin beta1-activated phosphatidylinositol 3-kinase enhances aggregation of MCF-7 breast cancer cells independent of extracellular signal-regulated kinase. Cancer Res 59:1620-1625, 1999. Tan M, Yao J, Yu D: Overexpression of the c-erbB-2 gene enhanced intrinsic metastasis potential in human breast cancer cells without increasing their transformation abilities. Cancer Res 57:1199-1205, 1997. Taylor SL, Platt Higgins A, Rudland PS, et al: Cytoplasmic staining of c-erbB-2 is not associated with the presence of detectable c-erbB-2 mRNA in breast cancer specimens. Int J Cancer 76:459-463, 1998. Tetu B, Brisson J: Prognostic significance of HER-2/neu oncoprotein expression in node-positive breast cancer. The influence of the pattern of immunostaining and adjuvant therapy. Cancer 73:2359-2365, 1994. Timar J, Trikha M, Szekeres K, et al: Expression and function of the high affinity alphaIIbbeta3 integrin in murine melanoma cells. Clin Exp Metastasis 16:437-445, 1998. Tiwari RK, Borgen PI, Wong GY, et al: HER-2/neu amplification and overexpression in primary human breast cancer is associated with early metastasis. Anticancer Res 12:419-425, 1992. Tzahar E, Pinkas Kramarski R, et al: Bivalence of EGF-like ligands drives the ErbB signaling network. EMBO J 16:4938- 4950, 1997. Tzahar E, Waterman H, Chen X, et al: A hierarchical network of interreceptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor. Mol Cell Biol 16:5276-5287, 1996. Vaughn JP, Iglehart JD, Demirdji S et al: Antisense DNA downregulation of the ERBB2 oncogene measured by a flow cytometric assay. Proc Natl Acad Sci USA 92:8338-8342, 1995. Verbeek BS, Adriaansen Slot SS, Vroom TM, et al: Overexpression of EGFR and c-erbB2 causes enhanced cell migration in human breast cancer cells and NIH3T3 fibroblasts. FEBS Lett 425:145-150, 1998. Warri AM, Laine AM, Majasuo KE, et al: Estrogen suppression of erbB2 expression is associated with increased growth rate of ZR-75-1 human breast cancer cells in vitro and in nude mice. Int J Cancer 49:616-623, 1991. Waterman H, Sabanai I, Geiger B, et al: Alternative intracellular routing of ErbB receptors may determine signaling potency. J Biol Chem 273:13819-13827, 1998. Wiechen K, Zimmer C, Dietel M: Selection of a high activity c-erbB-2 ribozyme using a fusion gene of c-erbB-2 and the enhanced green fluorescent protein. Cancer Gene Ther 5:45- 51, 1998. Willsher PC, Pinder SE, Gee JM, et al: C-erbB2 expression predicts response to preoperative chemotherapy for locally advanced breast cancer. Anticancer Res 18:3695-3698, 1998. Witters LM, Kumar R, Chinchilli VM, et al: Enhanced antiproliferative activity of the combination of tamoxifen plus HER-2-neu antibody. Breast Cancer Res Treat 42:1-5, 1997. Worthylake R, Opresko LK, Wiley HS: ErbB-2 amplification inhibits down-regulation and induces constitutive activation of both erbB-2 and epidermal growth factor receptors. J Biol Chem 274:8865-8874, 1999. Wright C, Cairns J, Cantwell BJ, et al: Response to mitoxantrone in advanced breast cancer: correlation with expression of c-erbB-2 protein and glutathione S-transferases. Br J Cancer 65:271-274, 1992. Ye D, Mendelsohn J, Fan Z: Augmentation of a humanized anti-HER2 mAb 4D5 induced growth inhibition by a humanmouse chimeric anti-EGF receptor mAb C225. Oncogene 18:731-738, 1999. Yen L, Nie ZR, You XL, et al: Regulation of cellular response to cisplatin-induced DNA damage and DNA repair in cells overexpressing p185(erbB-2, is dependent on the ras signaling pathway. Oncogene 14:1827-1835, 1997. Yu D, Liu B, Tan M, et al: Overexpression of c-erbB-2/neu in breast cancer cells confers increased resistance to Taxol via mdr- 1-independent mechanisms. Oncogene 13:1359-1365, 1996. Zafrani B, Leroyer A, Fourquet A, et al: Mammographicallydetected ductal in situ carcinoma of the breast analyzed with a new classification. A study of 127 cases: correlation with estrogen and progesterone receptors, p53 and c-erbB-2 proteins, and proliferative activity. Semin Diagn Pathol 11:208-214, 1994. Zidovetzki R, Johnson DA, Arndt Jovin DJ, et al: Rotational mobility of high-affinity epidermal growth factor receptors on the surface of living A431 cells. Biochemistry 30:6162-6166, 1991. Zidovetzki R, Yarden Y, Schlessinger J, et al: Microaggregation of hormone-occupied epidermal growth factor receptors on plasma membrane preparations. EMBO J 5:247-250, 1986. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 255 EP 271 PG 17 ER PT J AU Saleh, AH Khatib, G Jackson, H Banerjee, M AF Saleh, A Husain Khatib, Ghada Jackson, Hershel Banerjee, Mousumi TI Correlation of bcl-2 Oncoprotein Immunohistochemical Expression with Proliferation Index and Histopathologic Parameters in Colorectal Neoplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colon carcinoma; adenoma; bcl-2; Ki-67; proliferation index; immunohistochemistry ID colon carcinoma; adenoma; bcl-2; Ki-67; proliferation index; immunohistochemistry AB Thebcl-2oncogene plays an important role in carcinogenesis by inhibiting cell death (apoptosis). It was initially discovered in follicular B cell lymphoma with t(14,18), and subsequently found in other malignant and premalignant lesions. Alteration of the normal controls of cell proliferation is also a significant factor in the multistep process of tumorigenesis. The proliferative activity of a given lesion is commonly valuated by MIB1, a monoclonal antibody to Ki67 proliferation antigen. Immuno-histochemical (IHC) staining expression of bcl-2 and Ki67 was retrospectively investigated in a series of 52 colorectal carcinomas and 56 adenomas according to the avidin-biotin-complex method. The aim of the study was twofold: 1) to investigate any correlation between MIB1 and bcl-2 immunostaining expression in colonic adenomas and carcinomas, 2) to identify any relationship between either marker and several histopathologic parameters including tumor size, pathologic stage, lymph node metastasis, angio-lymphatic invasion, tumor grade and differentiation in colon carcinomas. Bcl-2 was consistently higher in adenomas than in carcinomas. There were 44/56 (78.6%) adenomas, and 27/52 (51.9%) carcinomas positive for bcl-2 (p=0.004).The mean Ki67 labeling index (LI) was 30.05±7.6 and 38.12±11.01 in adenomas and carcinomas, respectively (p=0.0001). Expression of bcl-2 in carcinoma was significantly associated with a lower mean Ki67 LI and with favorable histopathologic parameters. We conclude that bcl-2 oncoprotein expression is probably an early step in the process of colon carcinogenesis, and its expression may be associated with a favorable clinical course. Furthermore, an inverse relationship exists between bcl-2 and Ki67 in colonic neoplasia. Evaluation of bcl-2 and Ki67 IHC expression in colonic carcinoma should be performed prospectively to determine if their expression is of value in predicting the clinical course in these patients. C1 [Saleh, A Husain] Wayne State University, Department of PathologyDetroit, Michigan, USA. [Khatib, Ghada] Wayne State University, Department of PathologyDetroit, Michigan, USA. [Jackson, Hershel] Grace Hospital, Department of Medicine, Section of GastroenterologyDetroit, Michigan, USA. [Banerjee, Mousumi] Wayne State University, Department of PathologyDetroit, Michigan, USA. RP Saleh, AH (reprint author), Wayne State University, Department of Pathology, Detroit, USA. CR Lauwers GY, Kandemir O, Kulbilis P, et al. Cellular Kinitics in Barrett’s Epithelium Carcinogenic Sequence: Roles of Apoptosis, bcl-2 protein, and Cellular Proliferation. Mod Pathol 10:1201-1208, 1997. Suster S, Fisher C, Moran CA: Expression of bcl-2 Oncoprotein in Benign and Malignant Spindle Cell Tumors of Soft Tissue, Skin, Serosal Surfaces and Gastrointestinal Tract; Am J Surg Pathol 22:863-872, 1998. Berardo MD, Elledge RM, Moor C, et al: Bcl-2 and Apoptosis in Lymph Node Positive Breast Carcinoma. Cancer 82:1296- 1302, 1998. Kinokawa S, Akazawa K, Kinukawa N, et al: Inverse Correlation Between the Expression of bcl-2 and p53 proteins in primary Gastric Lymphoma. Hum Pathol 27:225-233, 1996. Barreton GB, Diebold J, Christoforis G, et al: Apoptosis and Immunohistochemical bcl-2 Expression in Colorectal Adenomas and Carcinomas. Cancer 77:255-264, 1996. Nakamura T, Nomura S, Sakai T, et al: Expression of bcl-2 Oncoprotein in Gastrointestinal and Uterine Carcinomas and their Premalignant Lesions. Hum Pathol 28:309-315, 1997. Bosari S, Moneghini L, Graziani D, et al: bcl-2 Oncoprotein in Colorectal Hyperplastic Polyps, Adenomas, and Adenocarcinomas. Hum Pathol 26:534-540, 1995. Palazzo JP, Kafka NJ, Grasso L, et al: The Role of p53, p21 WAF1/C1PI, and bcl-2 in Radioresistent Colorectal Carcinoma. Hum Pathol 28:1189-1195, 1997. Lu QL, Abel P, Foster C, et al: Bcl-2: Role in Epithelial Differentiation and Oncogenesis. Hum Pathol 27:102-110, 1996. Bronner MP, Culin C, Reed JC, et al: The bcl-2 Proto-Oncogene and the Gastrointestinal Epithelial Tumor Progression Model. Am J Pathol 146:20-26, 1995. Carr NJ, Monihan MJ, Nzeako UC, et al: Expression of Proliferative cell nuclear antigen in Hyperplastic Polyps, Adenoma and Inflammatory Cloacogenic Polyps of the Large Intestine. J Clin Pathol 48:46-52, 1995. Porschen R, Lohe B, Hengels KJ, et al: Assessment of Cell Proliferation in Colorectal Carcinomas Using the monoclonal Antibody Ki-67. Correlation With Pathohistologic Criteria and Influence of Irradiation. Cancer 64:2501-2505, 1989. Sahin AA, RoJY, Brown RW, et al: Assessment of Ki67 Derived Tumor Proliferative Activity in Colorectal Adenocarcinomas. Mod Pathol 7:17-22, 1994. Beer TW, Buchanan R, Matthews AW, et al: Prognosis in malignant Mesothelioma Related to MIB 1 Proliferative Index and Histological Subtype. Hum Pathol 29:246-251, 1998. Shepherd NA, Richman PI, England J: Ki67 Derived Proliferative Activity in Colorectal Adenocarcinoma with Prognostic Correlation. J Pathol 155:213-219, 1988. Suzuki H, Matsumoto K, Terabe M: Ki67 antibody labeling index in colorectal carcinoma. J Clin Gastroenterology 15:317- 320, 1992. Berenzi A, Benetti A, Bertalot G, et al: Ki67 immunohistochemical evaluation in colorectal cancer and normal colonic mucosa. Possible clinical applications. Pathologica 84:155-163, 1992. Benetti A, Berenzi A, Grigolato P: Growth fraction of colorectal carcinoma, Ki67): a comparative study. Int J Biol Markers 7:93-96, 1992. Lanza G Jr, Cavazzini L, Borghi L, et al: Immunohistochemical assessment of growth fractions in colorectal adenocarcinomas with monoclonal antibody Ki67. Relation to clinical and pathologic variables. Pathol Res Pract 186:608-618, 1990. Kyzer S, Gordon PH: Determination of proliferative activity in colorectal carcinoma using monoclonal antibody Ki67. Dis Colon Rectum 40:322-325, 1997. Johnston PG, O’Brien MJ, Dervan PA, et al: Immunohistochemical analysis of cell kinetic parameters in colonic adenocarcinomas, adenomas, and normal mucosa. Hum Pathol 20:696-700, 1989. Hoang C, Polivka M, Valleur P, et al: Immunohistochemocal detection of proliferating cells in colorectal carcinomas and adenomas with the monoclonal antibody Ki67. Preliminary data. Virchows Arch A Pathol Anat Histopathol 414:423-428, 1989. Ishida H, Irie K, Itoh T. The Prognostic significance of p53 and bcl-2 expression in lung adenocarcinoma and its correlation with Ki67 growth fraction. Cancer 80:1034-1045, 1997. Linden MD, Ma CK, Kubus J, et al: Ki67 and proliferating cell nuclear antigen tumor proliferative indices in DNA diploid colorectal adenocarcinomas. Correlation with histopathologic characteristics and cell cycle analysis with two-color DNA flow cytometry. Am J Clin Pathol 100:206-212, 1993. Gerdes J: Ki67 and other proliferating markers useful for immunohistological diagnostic and prognostic evaluations in human malignancies. Caner Biol 1:199-206, 1990. Isola JJ, Helin HJ, Helle MJ, et al: Evaluation of cell proliferation in breast carcinoma. Comparison of Ki67 immunohistochemical study, DNA flow cytometric analysis, and mitotic count. Cancer 65:1180-1184, 1990. Hitchcock CL: Ki67 staining as a means to simplify analysis of tumor cell proliferation, Editorial). Am J Clin Pathol 96:444- 446, 1991. Yang H, Hsu P, Chan S, et al: Growth Kinetics of colorectal adenoma- carcinoma sequence: An immunohistochemical study of proliferating cell nuclear antigen expression. Hum Pathol 27:1071-1076, 1996. Watson AJ, Merritt AJ, Jones LS, et al: Evidence of reciprocity of bcl-2 and p53 expression in human colorectal adenomas and carcinomas. Br J Cancer73:889-895, 1996. Duenas-Gonzalez A, Abad-Hernandez M, Cruz-Hernandez JJ, et al: Analysis of bcl-2 in sporadic breast carcinoma. Cancer 80:2100-2108, 1997. Hao XP, Ilyas M, Talbot IC: Expression of bcl-2 and p53 in the colorectal adenoma-carcinoma sequence. Pathobiology 65:140- 145, 1997. Sinicrope FA, Ruan SB, Cleary KR, et al: Bcl-2 and p53 oncoprotein expression during colorectal tumorigenesis. Cancer Res 55:237-241, 1995. Hawkins N, Lees J, Hargrave R, et al: Pathological and genetic correlates of apoptosis in the progression of colorectal neoplasia. Tumour Biol 18:146-156, 1997. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 274 EP 279 PG 6 ER PT J AU Abd El All, H Rye, A Duvillard, P AF Abd El All, Howayda Rye, Annie Duvillard, Pierre TI p53 Immunohistochemical Expression of Egyptian Cervical Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cervical carcinoma; p53; FIGO staging ID cervical carcinoma; p53; FIGO staging AB Data concerning the expression of p53 in cervical carcinoma, one of the leading cause of death in developing countries, are still confusing. This study was designed to identify p53 in Egyptian cervical carcinoma in an attempt to evaluate its prognostic significance. Eleven chronic cervicitis and 38 invasive carcinoma (31 squamous cell carcinoma (sqcc) and 7 adenocarcinoma, ranging from stage IB to IVB), were stained with the monoclonal antibody anti p53, DO7, using the microwave for antigen retrieval. No immunoreactivity was detected in chronic cervicitis, while nuclear p53 reactivity was detected in all carcinoma and in squamous intra-epithelial lesions (SIL) overlying 8 sqcc. P53 immunohistochemical (IHC) expression was more pronounced in early clinical stages (p=0.007) and in adenocarcinoma compared to sqcc (p=0.015). A positive correlation was present between p53 and heat shock protein 70 (hsp70) expressions (p=0.005). No correlation could be found between p53 expression and tumor infiltrating lymphocytes, the presence or absence of either schistosomiasis or HPV infections. It can be concluded, that in the Egyptian population, p53 immunoreactivity appears to be an early event in cervical neoplasm, and seems to play an important role together with other cell regulatory proteins in the process of carcinogenesis, which could be different between sqcc and adenocarcinoma. C1 [Abd El All, Howayda] Suez Canal University, Faculty of Medicine, Department of PathologyIsmailia, Egypt. [Rye, Annie] Institute Gustave Roussy, Department of StatisticsVillejuif, France. [Duvillard, Pierre] Institute Gustave Roussy, Department of PathologyVillejuif, France. RP Abd El All, H (reprint author), Suez Canal University, Faculty of Medicine, Department of Pathology, Ismailia, Egypt. EM howayda@intouch.com CR Abd El All H, Prade M, Khatab T, et al: Immunological study of NK cells in cervical carcinoma. Ph.D. Thesis, Faculty of Medicine, Suez Canal University, Ismailia, Egypt, 1992. Abd El All H, Rey A, Duvillard P: Expression of heat shock protein 70 and c-myc in cervical carcinoma. Anticancer Res 18:1533-1536, 1998. Agoff S,N, Hou J, Linzer DI, et al: Regulation of the human hsp70 promoter by p53. Science 259:5091 84-87, 1993. Beaudry GA, Bertelsen AH, Sherman MI: Therapeutic targeting of the p53 tumor suppressor gene. Curr Opin Biotechnol 7:592-600, 1996. Bosari S, Roncalli M, Viale G, et al: p53 immunoreactivity in inflammatory and neoplastic diseases of the uterine cervix. J Pathol 169:425-430, 1993. Crook T, Wrede D, Vousden HK: p53 point mutation in HPV negative human cervical carcinoma cell lines. Oncogene 6:873-875, 1991. Dittmer D, Pati S, Zambetti G, et al: Gain of function mutations in p53. Nat Genet 4:42-46, 1993. Elledge RM, Clark GM, Fuqua SA, et al: p53 protein accumulation detected by five different antibodies: relationship to prognosis and heat shock protein 70 in breast cancer. Cancer Res 54:3752-3757, 1994. Farias-Eisner RP, Walker DL, Berek JS: Gynecol Cancers. In: Manuel of Clinical Oncology. Ed Casciato DA and Lowitz BB. 3rd edition. Little, Brown Co, Boston- Massachusetts. pp 200-227, 1995. Fourie AM, Hupp TR, Lane DP, et al: HSP70 binding sites in the tumor suppressor protein p53. J Biol Chem 272:19471-19479, 1997. Helland A, Holm R, Kristensen G, et al: Genetic alterations of the TP53 gene, p53 protein expression and HPV infection in primary cervical carcinomas. J Pathol 171:105-114, 1993. Helland A, Karlsen F, Due EU, et al:Mutations in the TP53 gene and protein expression of p53, MDM 2 and p21/WAF-1 in primary cervical carcinomas with no or low human papillomavirus load. Br J Cancer 78:69-72, 1998. Holm R, Skomedal H, Helland A, et al: Immunohistochemical analysis of p53 protein overexpression in normal, premalignant, and malignant tissues of the cervix uteri. J Pathol 169:21-26, 1993. Hunt CR, Hale RJ, Buckley CH et al: p53 expression in carcinoma of the cervix. J Clin Pathol 49:971-974, 1996. Iwaya K, Tsuda H, Fujita S, et al: Natural state of mutant p53 protein and heat shock protein 70 in breast cancer tissues. Lab Invest 72:707-714, 1995. Jeffers MD, Richmond J, Farquharson M, et al: p53 immunoreactivity in cervical intraepithelial neoplasia and non-neoplastic cervical squamous epithelium. J Clin Pathol 47:1073-1076, 1994. Kainz C, Kohlberger P, Gitsch G, et al:Mutant p53 in patients with invasive cervical cancer stages IB to IIB. Gynecol Oncol 57:212- 214, 1995. Kubbutat MH, Vousden KH: Keeping an old friend under control: regulation of p53 stability. Mol Med Today 4:250-256, 1998. Lakshmi S, Nair MB, Jayaprakash PG, et al:p53 protein and tumorigenesis in the uterine cervix. Gen Diagn Pathol 142:281-287, 1997. Lambkin HA, Mothersill CM, Kelehan P: Variations in immunohistochemical detection of p53 protein overexpression in cervical carcinomas with different antibodies and methods of detection. J Pathol 172:13-18, 1994. Lane DP: P53, guardian of the genome. Nature 358:15-16, 1992. Levine AJ, Momand J, Finlay CA: P53 tumor suppressor gene. Nature 351:453-451, 1991. McCluggage G, McBride H, Maxwell P, et al: Immunohistochemical detection of p53 and bcl-2 proteins in neoplastic and non-neoplastic endocervical glandular lesions. Int J Gynecol Pathol 16:22- 27, 1997. Mittal KR, Lin O, Chan W, et al: Cervical squamous dysplasias and carcinomas with immunodetectable p53 frequently contain HPV. Gynecol Oncol 58:289-294, 1995. Mokhtar N: Cancer Pathology Registry 1985-1989. Ed Department of Pathology NCI, 1991. Moubayed P, Ziehe A, Peters J, et al: Carcinoma of the uterine cervix associated with schistosomiasis and induced by human papillomaviruses. Int J Gynaecol Obstet 49:175-179, 1995. Ngan HY, Tsao SW, Liu SS, et al:Abnormal expression and mutation of p53 in cervical cancer-a study at protein, RNA and DNA levels. Genitourin Med 73:54-58, 1997. Oka K, Nakano T, Arai T: p53CM1 expression is not associated with prognosis in uterine cervical carcinoma. Cancer 72:160-164, 1993. Pignatelli M, Stamp GW, Kafiri G, et al: Over-expression of p53 nuclear oncoprotein in colorectal adenomas. Int J Cancer 50:683- 688, 1992. Pillai MR, Halabi S, McKalip A, et al:The presence of human papillomavirus- 16/-18 E6, p53, and Bcl-2 protein in cervicovaginal smears from patients with invasive cervical cancer. Cancer Epidemiol Biomarkers Prev 5:329-335, 1996. Pinhasi-Kimhi O, Michalovitz D, Ben-Zeev A, et al: Specific interaction between the p53 cellular tumour antigen and major heat shock proteins. Nature 320:182-184, 1986. Quinn MA: Adenocarcinoma of the cervix- Are there arguments for a different treatment policy? Current Opinion in Obst Gynecol 9:21- 24, 1997. Raju GC, Teh M and Wee A: An immunohistochemical study of p53 protein in cervical intraepithelial neoplasia and squamous cell carcinoma. Pathology 28:17-19, 1996. Rosin MP, Anwar WA, Ward AJ: Inflammation, chromosomal instability, and cancer: the schistosomiasis model. Cancer Res 54:1929s- 1933s, 1994. Shan L, Nakamura Y, Zhang Z, et al: Overexpression of p53 protein correlates with a high risk of malignant transformation of adenomas in patients with multiple colorectal adenomas. Pathol Int 48:281- 286, 1998. Uchiyama M, Iwasaka T, Matsuo N, et al: Correlation between human papillomavirus positivity and p53 gene overexpression in adenocarcinoma of the uterine cervix. Gynecol Oncol 65:23-29, 1997. Woo RA, McLure KG, Lees-Miller SP, et al:DNA-dependent protein kinase acts upstream of p53 in response to DNA damage. Nature 394:700-704, 1998. Wynford-Thomas D: Oncogenes and anti-oncogenes; the molecular basis of tumour behaviour. J Pathol 165:187-201, 1991. Wynford-Thomas D: P53 in tumour pathology: can we trust immunocytochemistry? J Pathol 166:329-330, 1992. Youssef AF, Fayad MM and Shafeek M: Bilharziasis of the uterine cervix. J Obstet Gynaecol Brit Commonw 77:847-851, 1970. Zur Hausen H:Papilloma viruses in anogenital cancer as a model to understand the role of viruses in human cancers. Cancer Res 49:4677-851, 1989. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 280 EP 284 PG 5 ER PT J AU Dursun, AB Memis, L Dursun, A Bayiz, H Ozkul, M AF Dursun, A Berna Memis, Leyla Dursun, Ayse Bayiz, Hulya Ozkul, Mine TI Clinical Importance of Correlations Between p53 Immunoreactivity and Clinicopathological Parameters in Lung Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p53; lung cancer; clinicopathology ID p53; lung cancer; clinicopathology AB Many studies have revealed the frequency of p53 abnormalities in lung cancer. However, clinico-pathological studies of p53 abnormalities have yielded conflicting results. We examined the p53 immunoreactivity and studied the correlations of p53 status and clinicopathological parameters in 76 primary lung cancers. By using DO-7 antibody, different degrees of p53 immunoreactivity was detected in 8 of 30 small cell lung cancer (SCLC, 26.6%) and 22 of 46 non-small cell lung cancer (NSCLC, 47.8%), 6 of 19 adenocarcinoma, 16 of 27 epidermoid carcinoma cases. In the whole group, no correlation was detected between the p53 status and the histological types of tumor, local tumor invasion, nodal status, and distant metastasis and patient characteristics, such as age, gender or smoking habit. P53 status was also found to have no effect on survival. However, in the NSCLC group, there was a significantly higher p53 immunoreactivity in well- and moderately-differentiated tumors (p<0.05). Patients with p53 immunoreactivity had a poor therapeutic response in the whole group. We concluded that, although p53 immunreactivity may be found in NSCLC, this does not correlate with clinicopathological parameters except therapeutic response. In SCLC p53 immunreactivity can be negligible. C1 [Dursun, A Berna] Ankara Ataturk Training and Research Hospital, Ergin sok. 43/4, Mebu-sevleri/Tandogan, 06580 Ankara, Turkey. [Memis, Leyla] Gazi University Faculty of Medicine, Department of PathologyAnkara, Turkey. [Dursun, Ayse] Gazi University Faculty of Medicine, Department of PathologyAnkara, Turkey. [Bayiz, Hulya] Ankara Ataturk Training and Research Hospital, Ergin sok. 43/4, Mebu-sevleri/Tandogan, 06580 Ankara, Turkey. [Ozkul, Mine] Ankara Ataturk Training and Research Hospital, Ergin sok. 43/4, Mebu-sevleri/Tandogan, 06580 Ankara, Turkey. RP Dursun, AB (reprint author), Ankara Ataturk Training and Research Hospital, 06580 Ankara, Turkey. EM ebdursun@superonline.com CR Brambilla E, Gazzeri S, Moro D, et al: Immunohistochemical study of p53 in human lung carcinomas. Am J Pathol 143:199- 210, 1993. Caamano J, Ruggeri B, Momiki S, et al: Detection of p53 in primary lung tumors and nonsmall cell lung carcinoma cell lines. Am J Pathol 139:839-845, 1991. Carbone DP, Mitsudomi T, Chiba I, et al: p53 immunostaining positivity is associated with reduced survival and is imperfectly correlated with gene mutations in resected non-small cell lung cancer. A preliminary report of LCSG 871. Chest 106(6 Suppl): 377S-381S, 1994. Costa A, Silvestrini R, Mochen C, et al: p53 expression, DNA ploidy and S-phase cell fraction in operable locally advanced non-small-cell lung cancer. Br J Cancer 73:914-919, 1996. Dalquen P, Sauter G, Torhorst J, et al: Nuclear p53 overexpression is an independent prognostic parameter in node-negative non-small cell lung carcinoma. J Pathol 178:53-58, 1996. Ebina M, Steinberg SM, Mulshine JL and Linnoila RI: Relationship of p53 overexpression and up-regulation of proliferating cell nuclear antigen with the clinical course of non-small cell lung cancer. Cancer Res 54:2496-2503, 1994. El-Deiry WS: Role of oncogenes in resistance and killing by cancer therapeutic agents. Curr Opin Oncol 9:79-87, 1997. Fontanini G, Bigini D, Vignati S, et al: p53 expression in non small cell lung cancer: clinical and biological correlations. Anticancer Res 13:737-742, 1993. Horio Y, Takahashi T, Kuroishi T, et al: Prognostic significance of p53 mutations and 3p deletions in primary resected nonsmall cell lung cancer. Cancer Res 53:1-4, 1993. Iggo R, Gatter K, Bartek J, et al: Increased expression of mutant forms of p53 oncogene in primary lung cancer. Lancet 335:675-679, 1990. Johnson BE, Kelley MJ, Johnson EB: Biology of lung cancer. In: Lung Cancer., Ed: Johnson EB), Wiley-Liss Inc, 1995, pp. 15-41. Kern JA, Filderman AE: Oncogenes and growth factors in human lung cancer. Clin Chest Med 14:31-41, 1993. Kern JA, McLennan G. Genetic and molecular changes of human lung cancer. In: Fishman’s Pulmonary Diseases and Disorders., Ed: Fishman AP), McGraw-Hill Book Co, 1998, pp. 1695-1705. Korkolopoulou P, Oates J, Crocker J, et al: p53 expression in oat and non-oat small cell lung carcinomas: correlations with proliferating cell nuclear antigen. J Clin Pathol 46:1093-1096, 1993. Lee JS, Yoon A, Kalapurakal SK, et al: Expression of p53 oncoprotein in non-small-cell lung cancer: a favorable prognostic factor. J Clin Oncol 13:1893-1903, 1995. Liloglou T, Ross H, Prime W, et al: p53 gene aberrations in nonsmall- cell lung carcinomas from a smoking population. Br J Cancer 75:1119-1124, 1997. Marchetti A, Buttitta F, Merlo G, et al: p53 alterations in nonsmall cell lung cancers correlate with metastatic involvement of hilar and mediastinal lymph nodes. Cancer Res 53:2846- 2851, 1993. McLaren R, Kuzu I, Dunnill M, et al: The relationship of p53 immunostaining to survival in carcinoma of the lung. Br J Cancer 66:735-738, 1992. Morkve O, Halvorsen OJ, Skjaerven R, et al: Prognostic significance of p53 protein expression and DNA ploidy in surgically treated non-small cell lung carcinomas. Anticancer Res 13:571- 578, 1993. Nishio M, Koshikawa T, Kuroishi T, et al: Prognostic significance of abnormal p53 accumulation in primary, resected nonsmall- cell lung cancers. J Clin Oncol 14:497-502, 1996. Nuorva K, Soini Y, Kamel D, et al: Concurrent p53 expression in bronchial dysplasias and squamous cell lung carcinomas. Am J Pathol 142:725-732, 1993. Quinlan DC, Davidson AG, Summers CL, et al: Accumulation of p53 protein correlates with a poor prognosis in human lung cancer. Cancer Res 52:4828-4831, 1992. Passlick B, Izbicki JR, Haussinger K, et al: Immunohistochemical detection of P53 protein is not associated with a poor prognosis in non-small-cell lung cancer. J Thorac Cardiovasc Surg 109:1205-1211, 1995. Pavelic ZP, Gluckman JL, Gapany M, et al: Improved immunohistochemical detection of p53 protein in paraffin—embedded tissues reveals elevated levels in most head and neck and lung carcinomas: correlation with clinicopathological parameters. Anticancer Res 12:1389-1394, 1992. Postmus EP: Epidemiology of lung cancer. In: Fishman’s Pulmonary Diseases and Disorders., Ed: Fishman AP). McGraw- Hill Book Co, 1998, pp. 1707-1717. Rusch V, Klimstra D, Venkatraman E, et al: Aberrant p53 expression predicts clinical resistance to cisplatin-based chemotherapy in locally advanced non-small cell lung cancer. Cancer Res 55:5038-5042, 1995. Seaton A, Seaton D, Leitch AG: Cancer of the lung. In: Crofton and Douglas’s Respiratory Diseases., Eds: Seaton A, Seaton D and Leitch AG), Blackwell Scientific Publications, 1989, pp. 912-974. Smith ML, Fornace AJ Jr: Genomic instability and the role of p53 mutations in cancer cells. Curr Opin Oncol 7:69-75, 1995. Vega FJ, Iniesta P, Caldes T, et al: p53 exon 5 mutations as a prognostic indicator of shortened survival in non-small-cell lung cancer. Br J Cancer 76:44-51, 1997. Volm M, Efferth T, Mattern J: Oncoprotein, c-myc, c-erbB1, cerbB2, c-fos, and suppressor gene product, p53, expression in squamous cell carcinomas of the lung. Clinical and biological correlations. Anticancer Res 12:11-20, 1992. Walker C, Robertson LJ, Myskow MW, et al: p53 expression in normal and dysplastic bronchial epithelium and in lung carcinomas. Br J Cancer 70:297-303, 1994. Westra WH, Offerhaus GJ, Goodman SN, et al: Overexpression of the p53 tumor suppressor gene product in primary lung adenocarcinomas is associated with cigarette smoking. Am J Surg Pathol 17:213-220, 1993. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 285 EP 290 PG 6 ER PT J AU Cserni, G Vajda, K Tarjan, M Bori, R Svebis, M Baltas, B AF Cserni, Gabor Vajda, Kornel Tarjan, Miklos Bori, Rita Svebis, Mihaly Baltas, Bela TI Nodal Staging of Colorectal Carcinomas from Quantitative and Qualitative Aspects. Can Lymphatic Mapping Help Staging? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal carcinoma; staging; sentinel lymph node; lymphatic mapping ID colorectal carcinoma; staging; sentinel lymph node; lymphatic mapping AB Retrospective data analysis was performed to determine the minimum number of lymph nodes required for the staging of colorectal carcinomas, and a prospective feasibility study was carried out to identify sentinel nodes in order to clarify whether these may predict the nodal status. From among 240 colorectal carcinoma specimens investigated between 1996 and 1998, 224 tumors were analyzed for their nodal status. Lymphatic mapping with vital patent blue dye injection into the peritumoral sub-serosal layer was performed in 25 patients. Blue nodes were identified by the pathologist in the unfixed specimen immediately after the resection of the bowel and were assessed separately. Of the 123 node-positive carcinomas, 40 had more than 3 nodes involved. The nodal positivity increased substantially when more than 6 nodes were assessed. The cumulative percentage analysis demonstrated that ideally 16 and 13 nodes should be obtained for the identification of any nodal involvement or the involvement of more than 3 nodes, respectively. Lymphatic mapping was successful in 24 patients (96%). Blue nodes were predictive of the nodal status in 19 cases (79%), and were the only sites of metastasis in 2 patients (15% of the node-positive cases). Lymphatic mapping with the vital blue dye technique does not seem to facilitate the staging of colorectal cancers, at least in our patient population with relatively large and deeply infiltrating tumors, and unless the technique is improved or other selective features of lymph nodes are found, all lymph nodes should be assessed. A minimum of 6 nodes, and an optimum of 16 nodes or more, are suggested from these series. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, H-6000 Kecskemet, Hungary. [Vajda, Kornel] Bacs-Kiskun County Hospital, Department of SurgeryKecskemet, Hungary. [Tarjan, Miklos] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, H-6000 Kecskemet, Hungary. [Bori, Rita] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, H-6000 Kecskemet, Hungary. [Svebis, Mihaly] Bacs-Kiskun County Hospital, Department of SurgeryKecskemet, Hungary. [Baltas, Bela] Bacs-Kiskun County Hospital, Department of SurgeryKecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.c3.hu CR American Joint Committee on Cancer: Colon and rectum. In: AJCC cancer staging manual, 5th edition., Eds: Fleming ID, Cooper JS, Henson DE et al), Lippincott-Raven Publishers, Philadelphia-New York, 1997, pp. 83-90. Astler VR, Coller FA: The prognostic significance of direct extension of carcinoma of the colon and rectum. Ann Surg 139:846-851, 1954. Bilchik AJ, Giuliano AE, Essner R, et al: Universal application of intraoperative lymphatic mapping and sentinel lymphadenectomy in solid neoplasms. Cancer J Sci Am 4:351-358, 1998. Borgstein PJ, Pijpers R, Comans EF, et al: Sentinel lymph node biopsy in breast cancer: guidelines and pitfalls of lymphoscintigraphy and gamma probe detection. J Am Coll Surg 186:275-283, 1998. Cabanas RM: An approach for the treatment of penile carcinoma. Cancer 39:456-466, 1977. Caplin S, Cerottini J-P, Bosman FT, et al: For patients with Dukes’B, TNM stage II, colorectal carcinoma, examination of six or fewer lymph nodes is related to poor prognosis. Cancer 83:666-672, 1998. Cox CE, Pendas S, Cox JM, et al: Guidelines for sentinel node biopsy and lymphatic mapping of patients with breast cancer. Ann Surg 227:645-653, 1998. Cserni G: Changing trends in lymph node recovery from axillary clearance specimens in breast cancer: possible implications for the quantitative axillary status from a 17 year retrospective study. Eur J Oncol 2:403-408, 1997. Cserni G: Lymph node harvest reporting in patients with carcinoma of the large bowel. A French population-based study. Letter. Cancer 85:243-244, 1999. Dukes CE: The classification of cancer of the rectum. J Pathol Bacteriol 35:323-332, 1932. Fielding LP, Arsenault PA, Chapuis PH, et al: Clinicopathological staging for colorectal cancer: an International Documentation System, IDS, and an International Comprehensive Anatomical Terminology, ICAT). J Gastroenterol Hepatol 6:325-344, 1991. Gabriel WB, Dukes C, Bussey HJR: Lymphatic spread in cancer of the rectum. Br J Surg 23:395-413, 1935. Giuliano AE, Dale PS, Turner RR, et al: Improved axillary staging of breast cancer with sentinel lymphadenectomy. Ann Surg 180:700-704, 1995. Giuliano AE, Kirgan DM, Guenther JM, et al: Lymphatic mapping and sentinel lymphadenectomy for breast cancer. Ann Surg 220:391-398, 1994. Goldstein NS, Sanford W, Coffey M, et al: Lymph node recovery from colorectal resection specimens removed for adenocarcinoma. Trends over time and a recommendation for a minimum number of lymph nodes to be recovered. Am J Clin Pathol 106:209-216, 1996. Hermanek P, Giedl J, Dworak O: Two programmes for examination of regional lymph nodes in colorectal carcinoma with regard to the new pN classification. Path Res Pract 185:867- 873, 1989. Jass JR: Prognostic factors in rectal cancer. Eur J Cancer 31A:862-863, 1995. Joosten JJA, Stobbe LJA, Wauters CAP, et al: Intraoperative lymphatic mapping and the sentinel node concept in colorectal carcinoma. Br J Surg 86:482-486, 1999. Kelemen PR, Van Herle AJ, Giuliano AE: Sentinel lympadenectomy in thyroid malignant neoplasms. Arch Surg 133:288-292, 1998. Krag DN, Weaver DL, Alex JC, et al: Surgical resection and radiolocalization of sentinel lymph node in breast cancer using a gamma probe. Surg Oncol 2:335-340, 1993. Levenback C, Burke TW, Gershenson DM, et al: Intraoperative lymphatic mapping for vulvar cancer. Obstet Gynecol 84:163- 167, 1994. Mainprize KS, Hewavisinthe J, Savage A, et al: How many lymph nodes to stage colorectal carcinomas? J Clin Pathol 51:165-166, 1998. Maurel J, Launoy G, Grosclaude P, et al: Lymph node harvest reporting in patients with carcinoma of the large bowel. A French population-based study. Cancer 82:1482-1486, 1998. Morton DL, Wen DR, Wong J, et al: Technical details of intraoperative lymphatic mapping for early stage melanoma. Arch Surg 127:392-399, 1992. Oberg A, Stenling R, Tavelin B, et al: Are lymph node micrometastases of any clinical significance in Dukes stages A and B colorectal cancer? Dis Colon Rectum 41:1244-1249, 1998. O’Hea BJ, Hill AD, El-Shirbiny AM, et al: Sentinel lymph node biopsy in breast cancer: initial experience at Memorial Sloan- Kettering Cancer Center. J Am Coll Surg 186:423-427, 1998. Reuhl T, Kaisers H, Markwardt J, et al: Axillaausraumung bei klinisch nodal-negativen Mammakarzinom. Kann die Indikation durch „sentinel node“-Nachweis individualisiert werden? Dtsch Med Wochenschr 123:583-587, 1998. Scott KWM, Grace RH: Detection of lymph node metastases in colorectal carcinoma before and after fat clearance. Br J Surg 76:1165-1167, 1989. Veronesi U, Zurrida S, Galimberti V. Consequences of sentinel node in clinical decision making in breast cancer and prospects for future studies. Eur J Surg Oncol 24:93-95, 1998. Wolmark N, Fisher ER, Wieand HS, Fisher B and contributing NSABP investigators: The relationship of depth of penetration and tumor size to the number of positive nodes in Dukes C colorectal cancer. Cancer 53:2707-2712, 1984. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 291 EP 296 PG 6 ER PT J AU Kristt, D Winston, JG Mellov, MM Veltman, V Koren, R AF Kristt, Don Winston, J Ginger Mellov, M Moshe Veltman, Vladimir Koren, Rumelia TI Patterns of Proliferative Changes in Crypts Bordering Colonic Tumors: Zonal Histology and Cell Cycle Marker Expression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal carcinoma; colonic polyps; aberrant crypt foci; crypt replication cycles ID colorectal carcinoma; colonic polyps; aberrant crypt foci; crypt replication cycles AB Proliferative crypt changes have been noted in mucosa bordering colonic carcinomas, but their biological significance is disputed. We anticipated that zonal patterning of histological changes and cell cycle marker expression would provide clues to the pathogenesis of these border changes. 81 specimens were examined including carcinomas, adenomatours polyps, adenomas with early carcinoma, flat adenomas and aberrant crypt foci. The spatial distribution and frequency of micro-architectural features, and mucosal thickness were determined in a border domain of 150–300 sequential crypts/specimen. Immunocytochemical expression of Ki67 and p53 antigens in crypts also was semi-quantitatively examined. We found that in 100% of carcinomas two histologically abnormal zones (Proximate and Middle) separated tumor from normal mucosa. Differences in the feature frequency between zones were statistically significant (p<0.05). Both zones showed mild increases in crypt cell expression of Ki67, with a statistically significant relationship to zonal patterning (p<0.005). Weak expression of p53 only appeared in rare cells. Crypt elongation with mucosal thickening (1.9x normal, p<0.001) in the Proximate and Middle zones distinguished carcinomas from border changes in all benign lesions, except flat adenomas. Since this change occurs in all cases of carcinoma, there is no correlation with tumor stage or grade. Also in carcinomas, elaborate complexes of attached crypts (connected crypt structures) were characteristic of the Middle zone, so that proximate zone was always architecturally simpler. We conclude, that despite continuous carcinoma growth, the invaded border mucosa maintains a prototypical zonal organization of molecular and histological crypt changes This spatially organized reaction pattern is likely to reflect an interplay between regulated growth and destructive processes in response to advancing carcinoma. Compared to the edges of benign colonic tumors, the edges of carcinomas are distinctive and consistent enough to be diagnostically useful. C1 [Kristt, Don] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Winston, J Ginger] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Mellov, M Moshe] Hasharon Hospital, Rabin Medical Center (Golda Campus), Department of Nuclear MedicinePetach Tikvah, Israel. [Veltman, Vladimir] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Koren, Rumelia] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. RP Koren, R (reprint author), A, Rabin Medical Center (Golda Campus), Department of Pathology, Petach Tikvah, Israel. EM rumelia@isdnmail.co.il CR Eide TJ: Remnants of adenomas in colorectal carcinomas. Cancer 51:1866-1872, 1983. Muto T, Bussey H, Morson B: The evolution of cancer of the colon and rectum. Cancer 36:2251-2270, 1975. Katada N, Hinder RA, Smyrk TC, et al: Apoptosis is inhibited early in the dysplasia-carcinoma sequence of Barrett esophagus. Arch Surg 132:728-733, 1997. Bronner MP, Culin C, Reed JC, et al: The bcl-2 proto-oncogene and the gastrointestinal epithelial tumor progression model. Am J Pathol 146:20-26, 1995. Fenoglio C, Lane N: The anatomical precursor of colorectal carcinoma. Cancer 34:819-827, 1974. Madara J, Harte P, Deasy J: Evidence for an adenoma-carcinoma sequence in dimethylhydrazine-induced neoplasms of rat intestinal epithelium. Am J Pathol 110:230-235, 1983. Shimoda T, Ikegami M, Fujisaki J, et al: Early colorectal carcinoma with special reference to its development de novo. Cancer 64:1138-1146, 1989. Spratt JS Jr., Ackerman LV: Small primary adenocarcinomas of the colon and rectum. JAMA 179: 337-346, 1962. Hamilton SR: The molecular genetics of colorectal neoplasia. Gastroenterology 105:3-7, 1993. Saffos RO, Rhatigan RM: Benign, nonpolypoid, mucosal changes adjacent to carcinomas of the colon. Hum Pathol 8:441-449, 1977. Dansky-Ullmann C, Salgaller M, Adams S, et al: Synergistic effects of IL-6 and IFN-gamma on cacinoembryonic antigen and HLA expression by human colorectal carcinoma cells: role for endogenous IFN-beta. Cytokine 7:118-129, 1995. Brown LF, Berse B, Jackman RW, et al: Expression of vascular permeability factor, vascular endothelial growth factor, and it receptors in adenocarcinomas of the gastrointestinal tract. Cancer Res 53:4727-4735, 1993. Heldin C-H, Westermark B: Autocrine stimulation of growth of normal and transformed cells.In: Habenicht A, ed. Growth factors, differentiation factors and cytokines. Berlin, GDR: Springer-Verlag; 1990:267-278. Bird RP: Observations and quantification of aberrant crypts in the murine colon treated with a colon carcinogen. Cancer Lett 37:147-151, 1987. Roncucci L, Stamp D, Medline A, et al: Identification and quantification of aberrant crypt foci and microadenomas in the human colon. Hum Pathol 22:287-294, 1991. Kristt D, Kingsley B, Cal R: Colonic aberrant crypts may originate from imparied fisstoning: relevance to increased risk of neoplasia. Human Pathol 30:1450-1462, 1999. Pretlow TP, Barrow BJ, Scott, et al: Aberrant crypt foci: Putative preneoplastic foci in human colonic mucosa. Cancer Res 51:1564-1567, 1991. Shpitz B, Bomstein Y, Mekori Y, et al: Aberrant Crypt foci in human colons: distribution and histomorphologic characteristics. Hum Pathol 29:469-475, 1998. Gruber SB, Petersen GM, Kinzler KW, et al: Cancer, crash sites, and the new genetics of neoplasia. Gastroenterol 116:210-212, 1999. Dervan PA, Magee HM, Carney DN: Proliferating cell nuclear antigen counts in formalin-fixed paraffin-embedded tissue correlate with Ki-67 in fresh tissue. Pathology Patterns. Am J Clin Pathol 97:S21-S25, 1992. El-Deiry WS, Tokina T, Waldman T, et al: Topological control of p21WAF1/CIP1 expression in normal and neoplastic tissues. Cancer Res 55:2910-2919, 1995 Polyak K, Hamilton SR, Vogelstein B, et al: Early alteration of cell-cycle-regulated gene expression in colorectal neoplasia. Am J Pathol 149:381-387, 1996 Shi, SR:Antigen retrieval in formalin-fixed, paraffin-embedded tissues: an enhancement method for immunohistochemical staining based on microwave oven heating of tissue sections. J Histochem Cytochem 39:741-743, 1991 Riddel RH, Goldman H, Ransohoff DF, et al: Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Hum Pathol 14:931-966, 1983. Wong MH, Stappenbeck TS, Gordon JI: Living and commuting in intestinal crypts. Gastroenterol 116:208-210, 1999 Cheng H, Bjerknes M, Amar J, et al: Crypt production in normal and diseased human colonic epithelium. Anat Rec 216:44- 48, 1986. Park H-S, Goodlad RA, Wright NA: Crypt fission in the small intestine and colon: a mechanism for the emergence of G6PD locus-mutated crypts after treatment with mutagens. Am J Pathol 147:1416-1427, 1995. Yardley JH, Donovitz M: Colorectal biopsy in inflammatory bowel disease. In: Yardley JH, Morson BC, eds. The gastrointestinal tract. International Academy of Pathology Monograph, No. 18. Baltimore, Md: Williams and Wilkins; 50-94, 1977. Bjerknes M, Cheng H, Hay K, et al: APC mutation and crypt cycle in murine and human intestine. Amer J Pathol 150:833- 839, 1997. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 297 EP 303 PG 7 ER PT J AU Rokkas, Th Liatsos, Ch Karameris, A Petridou, E Lazaris, A Antoniades, D Kalafatis, E AF Rokkas, Theodore Liatsos, Christos Karameris, Andreas Petridou, Evangeija Lazaris, Andreas Antoniades, Dimitris Kalafatis, Evangeios TI Proliferating Cell Nuclear Antigen (PCNA) Immunostaining in Helicobacter PyloriInfection: Impact of Eradication SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Helicobacter pylori infection; H. pylorieradication; PCNA immunostaining; gastric proliferation; gastric carcinogenesis ID Helicobacter pylori infection; H. pylorieradication; PCNA immunostaining; gastric proliferation; gastric carcinogenesis AB Despite the fact that the association of Helicobacter pylori (H. pylori)with an increased risk of gastric cancer has been well documented, the exact mechanisms of this association have not been fully elucidated. The aim of the present prospective study was to contribute to the exploration of these mechanisms by studying the relationship between H. pylori infection and proliferating cell nuclear antigen (PCNA) immunostaining in endoscopic biopsies in gastric antrum. Furthermore, we examined the impact of H. pylorieradication on this relationship. We studied 28 H. pyloripositive patients and the results were compared with 22 endoscopically and histologically normal H. pylorinegative patients (control group) who were comparable to the H. pyloripositive group for age and sex. In addition all H. pyloripositive patients were examined before and after treatment aiming to eradicate H. pylori. In the H. pylori(+) patients the median PCNA index was 35 (range 8-58) and this was significantly higher than the respective number in the control group [5.5 (2–14), p<0.001]. In patients studied before and after successful eradication of H. pylori(n=10) the corresponding numbers were 35 (8-56) and 7 (4–13) (p<0.01) the latter not being significantly different from the control group of H. pylori(-)patients. On the contrary, in patients without successful H. pylori eradication (n=18) the PCNA indices before and after treatment were similar [35.5 (21-58) vs 31.5 (20-56)]. It is concluded that H. pyloriinfection alters the replication cycle of the gastric mucosa inducing hyperproliferation, which return towards normal after successful H. pylori eradication. C1 [Rokkas, Theodore] 401 Army General Hospital, Gastroenterology Unit and Histopathology Laboratory, 192B Alexandras Ave., 115 21 Athens, Greece. [Liatsos, Christos] 401 Army General Hospital, Gastroenterology Unit and Histopathology Laboratory, 192B Alexandras Ave., 115 21 Athens, Greece. [Karameris, Andreas] 401 Army General Hospital, Gastroenterology Unit and Histopathology Laboratory, 192B Alexandras Ave., 115 21 Athens, Greece. [Petridou, Evangeija] 401 Army General Hospital, Gastroenterology Unit and Histopathology Laboratory, 192B Alexandras Ave., 115 21 Athens, Greece. [Lazaris, Andreas] 401 Army General Hospital, Gastroenterology Unit and Histopathology Laboratory, 192B Alexandras Ave., 115 21 Athens, Greece. [Antoniades, Dimitris] 401 Army General Hospital, Gastroenterology Unit and Histopathology Laboratory, 192B Alexandras Ave., 115 21 Athens, Greece. [Kalafatis, Evangeios] 401 Army General Hospital, Gastroenterology Unit and Histopathology Laboratory, 192B Alexandras Ave., 115 21 Athens, Greece. RP Rokkas, Th (reprint author), 401 Army General Hospital, Gastroenterology Unit and Histopathology Laboratory, 115 21 Athens, Greece. EM sakkor@compulink.gr CR Marshall BJ, McGechie DB, Rogers PA, et al: Pyloric campylobacter infection and gastroduodenal disease. Med J Aust 142:439-444, 1985. Rokkas T, Sladen G: Infection with campylobacter pylori. J Royal Coll Phys London 22:97-100, 1988. Graham D: Campylobacter pylori and peptic ulcer disease. Gastroenterology 96:615-625, 1989. Raws EA, Tytgat GNJ: Cure of duodenal ulcer associated with eradication of Helicobacter pylori. Lancet 335:1233-1235, 1990. Tytgat GNJ, Lee A, Graham DY, et al: The role of infectious agents in peptic ulcer disease. Gastroenterol Internat 1993; 6:76-89, 1993. Sipponen P, Kosunen TU, Valle J, et al: Helicobacter pylori infection and chronic gastritis in gastric cancer. J Clin Pathol 45:319-323, 1992 Graham D: Pathogenic mechanisms leading to Helicobacter pylori-induced inflammation. Eur J Gastroenterol Hepatol 4, suppl 2):S9-S16, 1992. Forman D, Newell DG, Fullerton F, et al: Association between infection with H. pylori and risk of gastric cancer. Evidence from a prospective investigation. Brit Med J 302:1302-1305, 1991. Parsonnet J, Friedman GD, Vandersteen DP, et al: H. pylori infection and the risk of gastric carcinoma. N Engl J Med 325:1127-1135, 1991 Nomura A, Stemmerman GN, Chyou PH, et al: H. pylori and gastric carcinoma among Japanese Americans in Hawaii. N Engl J Med 325:1132-1136, 1991 Talley NJ, Zinsmeister AR, Weaver A, et al: Gastric adenocarcinoma and H. pylori infection. J Natl Cancer Inst 83:1734-1739, 1991. THE EUROGAST Study Group. An international association between Helicobacter pylori infection and gastric cancer: an international study. Lancet 341:1359-1362, 1993. Hansson LE, Engstrand L, Nyren O, et al: Helicobacter pylori infection: Independent risk indicator of gastric adenocarcinoma. Gastroenterology 105:1098-1103, 1993. International Agency for Research on Cancer. Schistosomes, liver flukes and Helicobacter pylori. Evaluation of carcinogenic risks to humans. IARC Monogr Eval Carcinog Risks Hum 61, 1994. Correa P, Ruiz B, Shi TY, et al: Helicobacter pylori and nucleolar organizer regions in the gastric antral mucosa. Am J Clin Pathol 101:656-660, 1994. Garcia RL, Coltrera MD, Gown AM: Analysis of proliferative grade using anti-PCNA / Cyclin in fixed, embedded tissues. Comparison with flow cytometric analysis. Am J Pathol 134:733-739, 1989. Hall PA, Levison DA, Woods AL, et al: Proliferating cell nuclear antigen, PCNA, immunolocalization in paraffin sections: an index of cell proliferation with evidence of deregulated expression in some neoplams. J Pathol 162:285-294, 1990. Woods AL, Hall PA, Shepherd NA, et al: The assessment of proliferating cell nuclear, PCNA, immunostaining in primary gastrointestinal lymphomas and its relationship to histological grade, S+G2+M phase fraction, flow cytometric analysis, and prognosis. Histopathology 19:21-27, 1991. Kamel OW, LeBrun DP, Davis RE, et al: Growth fraction estimation of malignant lymphomas in formalin-fixed paraffinembedded tissue using anti-PCNA / Cyclin 19A2. Correlation with Ki-67 labelling. Am J Pathol 138:1471-1477, 1993. Battersby S, Anderson TJ: Correlation of proliferative activity in breast tissue using PCNA/Cyclin. Hum Pathol 21:781, 1990. Coltrera MD, Gown AM: PCNA/Cyclin expression and BrdU uptake define different subpopulations in different cell lines. J Histochem Cytochem 39:23-30, 1991. Marshall BJ, Francis G, Langton S, et al: Rapid urease test in the management of Campylobacter pyloridis associated gastritis. Am J Gastroenterol 3:200-210, 1987. Potters HVPJ, Loffeld RJLF, Stobbeling E, et al: Rapid staining of Campylobacter pyloridis. Histopathology 11:1223, 1987. Logan RP: Urea breath tests in the management of Helicobacter pylori infection. Gut 43, Suppl 1):S47-50, 1998. Dixon MF, Genta RM, Yardley JH, et al: Classification and grading of gastritis. The updated Sydney system. Amer J Surg Pathol 20:1161-1181, 1996. Motalsky H. Intuitive Biostatistics. Oxford University press. New York, Oxford, 1995. Fraser AG, Sim R, Sankey EA, et al: Effect of eradication of Helicobacter pylori on gastric epithelial cell proliferation. Aliment Pharmacol Ther 8:167-173, 1994. Cahill RJ, Xia H, Kilgallen C, et al: Effect of eradication of Helicobacter pylori infection on gastric epithelial cell proliferation. Dig Dis Sci 40:1627-1631, 1995. Lynch DA, Mapstone NP, Clarke AM, et al: Cell proliferation in Helicobacter pylori associated gastritis and the effect of eradication therapy. Gut 36:346-350, 1995. Havard TJ, Sarsfield P, Wotherspoon AC, et al: Increased gastric epithelial cell proliferation in Helicobacter pylori-associated follicular gastritis. J Clin Pathol 49:68-71, 1996. Bechi P, Balzi M, Beccioni A, et al:Helicobacter pylori and cell proliferation of the gastric mucosa: possible implications for gastric carcinogenesis. Am J Gasroenterol 91:271-276, 1996. Chow KW, Bank S, Ahn J, et al: Helicobacter pylori infection does not increase gastric antrum mucosal cell proliferation. Am J Gastroenterol 90:64-66, 1995. Peek RM Jr, Moss SF, Tham KT, et al: Helicobacter pylori cagA+ strains and dissosiasion of gastric epithelial cell proliferation from apoptosis. J Natl Cancer Inst 89:863-868, 1997. Konturek PC, Bobrzynski A, Konturek SJ, et al: Epidermal growth factor and transforming growth factor alpha in duodenal ulcer and non-ulcer dyspepsia patients before and after Helicobacter pylori eradication. Scand J Gastroenterol 33:143-151, 1998. Moss SF, Scholes JV, Holt PR: Abnormalities of epithelial apoptosis in multistep colorectal neoplasia demonstrated by terminal deoxyuridine nick end labeling. Dig Dis Sci 41:2238-2247, 1996. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 304 EP 308 PG 5 ER PT J AU Maity, P Chakraborty, S Bhattacharya, P AF Maity, Putul Chakraborty, Sunit Bhattacharya, Pritha TI Angiogenesis - a Putative New Approach in Glutamine Related Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE angiogenesis; glutaminase; murine tumor ID angiogenesis; glutaminase; murine tumor AB Angiogenesis or the generation of new blood vessels, is an important factor regarding the growth of a tumor. Hence, it becomes a necessary parameter of any kind in therapeutic studies. Glutamine is an essential nutrient of tumor tissue and glutamine related therapy involves clearance of circulatory glutamine by glutaminase. So, whether this enzyme has any effect on angiogenesis of a tumor or not becomes an obvious question. To address this question, this study has been carried out with different murine tumor models.The results indicate that purified glutaminase reduces tumor volume as well as restricts the generation of new blood vessels. Glutaminase is effective in the case of solid as well as ascites tumor models. In the case of induced cancer, the host exhibits delayed onset of neoplasia following enzyme treatment and tumor host interactions determine the intensity of the neovascularisation process. Therefore, it can be concluded that this enzyme might be an effective agent against cancer metastasis. C1 [Maity, Putul] Chittaranjan National Cancer Institute, Department of Metabolic Regulation, 37, S. P. Mukherjee Road, 700026 Calcutta, India. [Chakraborty, Sunit] Chittaranjan National Cancer Institute, Department of Metabolic Regulation, 37, S. P. Mukherjee Road, 700026 Calcutta, India. [Bhattacharya, Pritha] Chittaranjan National Cancer Institute, Department of Metabolic Regulation, 37, S. P. Mukherjee Road, 700026 Calcutta, India. RP Maity, P (reprint author), Chittaranjan National Cancer Institute, Department of Metabolic Regulation, 700026 Calcutta, India. EM cncinstegiasceos@vsul.net.in CR Bossi P, Viale G, Lee AK, et al: Angiogenesis in colorectal tumors: microvessel quantitation in adenomas and carcinomas with clinicopathological correlations. Cancer Res 55:5049- 5053, 1995. Torry RJ, Rongish BJ: Angiogenesis in the uterus : potential regulation and relation to tumor angiogenesis. Amer J Reprod Immunol 27:171-179, 1992. Folkman J, Shing Y: Angiogenesis. J Biol Chem 26:10931- 10934, 1992. Goldie H, Felix MD: Growth characteristics of free tumor cells transferred serially in the peritoneal fluid of the mouse. Cancer Res 11:73-81, 1951. Nagy JA, Morgan ES, Herzberg KT, et al: Pathogenesis of ascites tumor growth : angiogenesis, vascular remodelling and stroma formation in the peritoneal lining. Cancer Res 55:376- 385, 1995. Miguez M, Davel L, Sacerdote de Lustig E: Lymphocyte-Induced Angiogenesis: Correlation with the metastatic incidence of two murine Mammary Adenocarcinomas. Invasion Metast 6:313- 320, 1986. Folkman J: What is the role of angiogenesis in metastasis from Cutaneous Melanoma. Eur J Cancer Clin Oncol 23:361-363, 1987. Netland PA, Zetter BR: Tumor-cell interactions with blood vessels during cancer metastasis. Fundamental Aspects of Cancer Dordrecht: Kluwer Academic Publishers. 84-97, 1989. Greenblat M, Shubik P: Tumor Angiogenesis : Transfilter diffusion studies in the hamster by transparent chamber techniques. J Natl Cancer Inst 41:111-124, 1968. Roberts J, Holcenberg JS, Dolowy WC: Antineoplastic activity of highly purified bacterial glutaminase. Nature 227:1136- 1137, 1970. Maity P, Chakraborty S, Bhattacharya P, et al: Isolation and purification of phosphate dependent glutaminase from Sacoma- 180 tumor and its antineoplastic effects on murine model system. J Exp Clin Cancer Res, communicated). Murphy ED: Studies in carcinogen induced carcinoma of the cervix in mice. Am J Pathol 29:608, 1953. Murphy ED: Carcinogenesis of the uterine cervix in mice effect of diethylstilbestrol after limited application of 3-methylcholanthrene. J Natl Cancer Inst 27:611-653,1961. Quesada AR, Sanchez-Jimenez F, Perez-Rodriguez J, et al: Purification of phosphate dependent glutaminase from isolated mitochondria of Ehrlich ascites tumor cells. Biochem J 256:1031- 1036, 1988. Lund P: In methods of enzymatic analysis. Verlag Chemie: Weinheim 8:357-369, 1985. Lowry OH, Rosenbrough NJ, Farr AL, et al: Protein measurements with folin phenol reagent. J Biol Chem 193:265-275, 1951 Danielsen T, Rofstad EK: VEGF, bFGF and EGF in the angiogenesis of human melanoma xenografts. Int J Cancer 76:836- 841, 1998. Folkman J: What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst 82:4-6, 1990. Burger MM, Folkman J: UICC study group on basic and clinical research: Tumor Angiogenesis. Int J Cancer 56:311-313, 1994. Nagy JA, Brown LF, Senger DR, et al: Pathogenesis of tumor stroma generation : a critical role for leaky blood vessels and fibrin deposition. Biochim Biophys Acta 948:305-326,1998. Dvorak HF, Harvey VS, Estrella P, et al: Fibrin containing gels induce angigenesis. Lab Invest 57:673-686, 1987. Neidle A, Waelsch H: J Biol Chem 234:586, 1959. Matar P, Celoria GC, Font MT, et al: Angiogenesis induced by tumor host interaction in the rat. J Exp Clin Cancer Res 16:249- 253, 1997. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 309 EP 314 PG 6 ER PT J AU Sarkady, E Sapi, Z Toth, V Kiss, S AF Sarkady, Emese Sapi, Zoltan Toth, Vera Kiss, Sandor TI Warthin-like Tumor of the Thyroid - a Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Warthin-like tumor; thyroid gland; papillary carcinoma; Hashimoto thyroiditis; metastasis ID Warthin-like tumor; thyroid gland; papillary carcinoma; Hashimoto thyroiditis; metastasis AB A case of Warthin-like tumor of the thyroid (WaLTT) with cervical lymph node metastasis is presented. The problems of the FNA diagnosis of this type of tumor is discussed as well as the histogenesis, nature and behaviour of this peculiar tumor. C1 [Sarkady, Emese] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93, H-1091 Budapest, Hungary. [Sapi, Zoltan] St John's Hospital, Department of PathologyBudapest, Hungary. [Toth, Vera] St John's Hospital, Department of PathologyBudapest, Hungary. [Kiss, Sandor] St. John Hospital, Department of SurgeryBudapest, Hungary. RP Sarkady, E (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM se@korb2.sote.hu CR Apel LR, Asa LS, Livolsi VA: Papillary Hurthle cell carcinoma with lymphocytic stroma. Am J Surg Pathol 19:810-814, 1995. Jarry A, Cerf-Bensussan N, Brousse N, et al: Same peculiar subset of HML1+ lymphocytes present within normal intestinal epithelium is associated with tumoral epithelium of gastrointestinal carcinomas. Gut 29:1632-1638, 1988. Kradin RL, Bhan AK: Tumor infiltrating lymphocytes. Lab Invest 69:635-638, 1993. Okayashu I, Fujiwara M, Hara Y, et al: Association of chronic lymphocytic thyroiditis and thyroid papillary carcinoma . A study of surgical cases among Japanese, and white and African Americans. Cancer 76:2312-2318, 1995. Ostrowski ML, Merino MJ: Tall cell variant of papillary thyroid carcinoma. A reassessment and immunohistochemical study with comparison to the ususal type of papillary carcinoma of thyroid. Am J Surg Pathol 20:964-974, 1996. Ozaki O, Ito K, Mimura T, et al: Papillary carcinoma of the thyroid. Tall cell variant with extensive lymphocyte infiltration. Am J Surg Pathol 20:695-698, 1996. Schroder S, Schwarz W, Rehpenning W, et al: Dendritic/Langerhans cells and prognosis in patients with papillary thyroid carcinomas: immunohistochemical study of 106 thyroid neoplasms correlated to follow-up data. J Clin Pathol 89:295-300, 1988. Vera-Sempere FJ, Prieto M, Camanas A: Warthin-like tumor of the thyroid: a papillary carcinoma with mitochondrion-rich cells and abundant lymphoid stroma. A case report. Pathol Res Pract 194:341-347, 1998. Wirtschafter A, Schmidt R, Rosen D, Kundu N, et al: Expression of the RET/PTC fusion gene as a marker for papillary carcinoma in Hashimoto’s thyroiditis. Laryngoscope 107:95-100, 1997. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 315 EP 317 PG 3 ER PT J AU Jylling, BAM Jorgensen, L Holund, B AF Jylling, Bak Anne Marie Jorgensen, Lars Holund, Berit TI Mucinous Cystadenocarcinoma in Combination with Hemangiosarcoma in the Ovary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE ovary; cystadenocarcinoma; mucinous; hemangiosarcoma ID ovary; cystadenocarcinoma; mucinous; hemangiosarcoma AB The ovary is the sixth most frequent site of cancer in women in Denmark with an incidence of approximately 600 cases per year. Carcinomas predominate whereas sarcomas are rare. We describe a case of the combination mucinous cystadenocarcinoma and hemangiosarcoma in a 37-year old woman, who had a right-sited oophorectomy because of a cyst. Clinically there was no suspicion of malignancy. The macro- and microscopic findings are described as well as the immunohistochemical stainings performed to confirm the diagnosis. The case shows the importance of careful sampling at the macroscopic examination, especially from areas with a striking appearance. C1 [Jylling, Bak Anne Marie] Odense University Hospital, Department of Pathology, Winsloewparken 15, DK-5000 Odense, Denmark. [Jorgensen, Lars] Odense University Hospital, Department of Pathology, Winsloewparken 15, DK-5000 Odense, Denmark. [Holund, Berit] Odense University Hospital, Department of Pathology, Winsloewparken 15, DK-5000 Odense, Denmark. RP Jylling, BAM (reprint author), Odense University Hospital, Department of Pathology, DK-5000 Odense, Denmark. EM anne.marie.bak.jylling@ouh.dk CR Ongkasuwan C, Taylor J, Tang C: Angiosarcomas of the Uterus and Ovary: Clinicopathologic report. Cancer 49:1469-1475, 1982. Nucci M, Krausz T, Lifschitz-Mercer B: Angiosarcoma of the ovary. Am J Surg Pathol 22:620-630, 1998. Prat J, Scully R: Sarcomas in ovarian mucinous tumors. Cancer 44:1327-1331, 1979. Prat J, Scully R: Ovarian mucinous tumors with sarcoma-like mural nodules. Cancer 44:1332-1344, 1979. Haines & Taylor: Obstetrical and Gynaecological Pathology. Churchill Livingstone, 1995. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 318 EP 319 PG 2 ER PT J AU Lazaris, ChA Paraskevakou, H Davaris, SP AF Lazaris, Ch Andreas Paraskevakou, Helen Davaris, S Panayiotis TI A Solitary Cutaneous Tumor with Distinct Areas of Verruca and Seborrheic Keratosis-like Lesion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE HPV; seborrheic keratosis; skin; in situ hybridization ID HPV; seborrheic keratosis; skin; in situ hybridization AB A single, exophytic, cutaneous tumor on the thigh of a 52-year-old man was examined by light microscopy, in situ hybridization and immunohistochemistry. It demonstrated distinct areas of verruca and of seborrheic keratosis-like morphology simultaneously. Focally, architectural abnormalities were noted in some deeper parts of the tumor, but there was no morphological evidence of malignancy. The patient has remained disease-free for two and a half years after surgery. Biotinylated full genomicDNA probes of HPV confirmed the presence of types 6/11 exclusively in the verrucous portion of the neoplasm. In the verrucous component p53 protein was overexpressed and, additionally, increased Ki-67 immunopositive signals were detected, being localized below the HPV-DNA-expressing spinous cells. C1 [Lazaris, Ch Andreas] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str., Goudi, GR-115 27 Athens, Greece. [Paraskevakou, Helen] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str., Goudi, GR-115 27 Athens, Greece. [Davaris, S Panayiotis] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str., Goudi, GR-115 27 Athens, Greece. RP Lazaris, ChA (reprint author), University of Athens, Medical School, Department of Pathology, GR-115 27 Athens, Greece. EM alazaris@cc.uoa.gr CR Jacyc WK, Dreyer L, de Villiers EM: Seborrheic keratoses of black patients with epidermodysplasia verruciformis contain human papillomavirus DNA. Am J Dermatopathol 15:1-6, 1993. Kirkham N: Tumors and cysts of the epidermis. In: Lever's Histopathology of the Skin., Eds: Elder D, Elenitsas R, Jaworsky C and Johanson B Jr), Lippincott-Raven, Philadelphia, USA, 1997, pp. 716-717. Lassus J, Ranki A: Simultaneously detected aberrant p53 tumor suppressor protein and HPV-DNA localize mostly in separate keratinocytes in anogenital and common warts. Exp Dermatol 5:72-78, 1996. Leonardi Ch, Lhu WY, Kinsey WH, et al: Seborrheic keratoses from the genital region may contain human papillomavirus DNA. Arch Dermatol 127:1203-1206, 1991. Li J, Ackerman AB: "Seborrheic keratoses" that contain human papillomavirus are condylomata acuminata. Am J Dermatopathol 16:398-405, 1994. Lizard G, Usson Y, Chignol MC, et al: Improvements in visualisation and localisation of human papillomavirus DNA in CaSki cells by fluorescence in situ hybridization, laser scanning confocal microscopy and three-dimensional image reconstruction. Anal Cell Pathol 7:53-61, 1994. Lu S, Syrjanen K, Havu VK, et al: Expression of PCNA is associated with the presence of HPV DNA in skin warts. Arch Dermatol Res 289:35-39, 1996. Payne DA, Sanchez R, Tyring SK: Cutaneous verruca with genital human papillomavirus in a 2 year-old girl. Am J Dermatopathol 19:258-260, 1997. Tsambaos D, Monastirli A, Kapranos N, et al: Detection of human papillomavirus DNA in congenital seborrhoeic keratoses. Arch Dermatol Res 287:612-615, 1995. Xia MY, Zhu WY, Lu JY, et al: Ultrastructure and human papillomavirus DNA in papillomatosis of external auditory canal. Int J Dermatol 35:337-339, 1996. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 1999 VL 5 IS 4 BP 320 EP 323 PG 4 ER PT J AU Robert, L AF Robert, Ladislas TI Cellular and Molecular Mechanisms of Aging and Age Related Diseases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE aging; matrix; cell communication ID aging; matrix; cell communication AB This review on aging is focused on those cellular and molecular mechanisms which concern age related pathologies. The central question addressed is the relationship between ''normal” aging and age-related pathologies such as osteoarthritis, cardiovascular diseases, emphysema, malignant tumors and cognitive decline, dementias. The mechanisms recognized as most important in cell and tissue aging are briefly outlined. Emphasis is laid on the importance of post-synthetic modifications of the macromolecules of the extracellular matrix and on cell matrix interactions. Loss of intercellular communication and cell-matrix interactions as a result of receptor decay and receptor uncoupling were recently recognized as key events. Unavoidable poly-pathology at advanced age may be the answer to the above question. C1 [Robert, Ladislas] Universite de Pierre et Marie Curie, Laboratoire de Recherche sur les Therapeutiques Substitutives en Ophtalmologie, 6, Hotel-Dieu, 1 Place du Parvis Notre-Dame, 75181 Paris, Paris cedex 04., France. RP Robert, L (reprint author), Universite de Pierre et Marie Curie, Laboratoire de Recherche sur les Therapeutiques Substitutives en Ophtalmologie, 75181 Paris, France. CR Allal M, Robert L, Labat-Robert R: Fragmentation de la fibronectine dans la mucoviscidose. C R Acad Sci 314:587- 592, 1992. Bailey AJ, Robins SP: Development and maturation of the crosslinks in the collagen fibres of skin, In: Frontiers of Matrix Biology, Eds: Robert L, Robert B). Karger, Basel 1973, pp. 1:130-156. Balo J: Connective tissue changes in atherosclerosis. In: International review of connective tissue research., Ed: Hall DA). Academic Press, New York, 1963, pp. l:241-306. Barlati S, Adamoli A, De Petro G: Presence and role of fibronectin fragments in transformed cells. In: Structural glycoproteins in cell matrix interactions. Frontiers of Matrix Biology,, Eds: Timpl R, Labat-Robert J, Robert L). Karger, Basel, 1986, pp. 11:174-182. Bizbiz L, Alperovitch A, Robert L. et al: Aging of the vascular wall: serum concentration of elastin peptides and elastase inhibitors in relation with cardiovascular risk factor. The EVA study, Atherosclerosis 131:73-78, 1997. Bodnar AG, Ouellette M, Frolkis M, et al: Extension of lifespan by introduction of telomerase into normal human cells. Science 279:349-352, Brenner S. ed., Telomeres and telomerase, 1997, Ciba Foundation Symposium 211, John Wiley & Sons, Chichester. Deguine V, Menasche M, Ferrari M, et al: Free radical depolymerization of hyaluronan by Maillard reaction products. Role in liquefaction of aging vitreous. Int J Biol Macromol 22:17-22, 1998. Emerit I, Chance B:Free radicals and aging. Birkhauser Verlag, Basel, 1992. Faury G, Chabaud A, Ristori MT, et al: Effect of age on the vasodilatory action of elastin peptides Mechanisms of Ageing and Development 95:31-42, 1997. Faury G, Ristori MT, Verdetti J, et al: Effect of elastin peptides on vascular tone. J Vasc Res 32:112-119, 1995. Fodil-Bourahla I, Drubaix I, Robert L: Effect of in vitro aging on the biosynthesis of glycosaminoglycans by human skin fibroblasts. Modulation by the elastin-laminin receptor. Mechanisms of Ageing and Development 106:241-260, 1999. Fulop T, Jacob MP, Varga Zs, et al: Effect of elastin peptides on human monocytes : Ca2+ mobilization, stimulation of respiratory burst and enzyme secretion. Biochem Biophys Res Commun 141:92-98, 1986. Fulop T, Wei SM, Robert L, Jacob MP: Determination of elastin peptides in normal and arteriosclerotic human sera by ELISA. Clin Physiol Biochem 8:273-282, 1990. Fulop T, Jacob MP, Khalil A, et al: Biological effects of elastin peptides. Pathol Biol 46:497-506, 1998. Gariepy J, Simon A, Massonneau M, et al: Wall thickening of carotid and femoral arteries in male subjects with isolated hypercholesterolemia. Atherosclerosis 113:141-151, 1995. Gershon D: Current status of age altered enzymes: alternative mechanisms. Mech Aging Develop 9:189-196, 1979. Gershon H, Gershon D: Detection of inactive enzymes in ageing organisms. Nature 227:1214-1217, 1970. Gown AM, Tsukada T, Ross R: Human atherosclerosis II. Immunocytochemical analysis of the cellular composition of human atherosclerotic lesions. Am J Pathol 125:191-207, 1986. Haranghy L: Gerontological studies on Hungarian centenarians. Akademia Kiado, Budapest, 1965. Hayflick L, Moorhead PS: The serial cultivation of human diploid cell strains. Expl Cell Res 25:585-621, 1961. Homandberg GA, Hui F: High concentrations of fibronectin fragments cause short-term catabolic effects in cartilage tissue while lower concentrations cause continuous anabolic effects. Arch Biochem Biophys 311:213-218, 1994. Homandberg GA, Meyers R, Xie DL: Fibronectin fragments cause chondrolysis of bovine articular cartilage slices in culture. J Biol Chem 267:3597-3604, 1992. Hornebeck W, Derouette JC, Roland J, et al: Correlation entre l’age, l’arteriosclerose et l’activite elastinolytique de la paroi aortique humaine. C R. Acad Sci 292:2003-2006, 1976. Hornebeck W, Tixier JM, Robert L: Inducible adhesion of mesenchymal cells to elastic fibers: elastonectin. Proc Natl Acad Sci 83:5517-5520, 1986. Huzella T: Zwischenzellige Organisation. Jena Verlag von Gustav Fischer, 1941. Ikan R., Ed. The Maillard reaction. Consequences for the chemical and life sciences. John Wiley & Sons, Chichester, 1996. Jacob MP, Hornebeck W, Robert L: Studies on the interaction of cholesterol woth soluble and insoluble elastins. Int J Biol Macromol 5:275-278, 1983. Keil-Dlouha V, Planchenault T: Potential proteolytic activity in human plasma fibronectin. Proc Natl Acad Sci 83:5377-5381, 1986. Labat-Robert J, Kern P, Robert L: Biomarkers of connective tissue aging: biosynthesis of fibronectin, collagen type III and elastase. Ann New York Acad Sci 673:16-22, 1992. Labat-Robert J, Potazman JP, Derouette JC, Robert L: Agedependent increase of human plasma fibronectin. Cell Biol Int 5:969-973, 1981. Lakatta EG: Deficient neuroendocrine regulation of the cardiovascular system with advancing age in healthy humans. Circulation 87:631-636, 1993. Lopez-Armada MJ, Gonzalez E, Gomez-Guerrero C, et al: The 80-kD fibronectin fragment increases the production of fibronectin and tumour necrosis factor-alpha, TNF-?, in cultured mesangial cells. Clin Exp Immunol 107:398-403, 1997. Macieira-Coelho A: Biology of normal proliferating cells in vitro relevance for in vivo aging. Karger, Basel, 1998. Macieira-Coelho A: Cancer and aging. Exper Gerontol 21:483- 495, 1986. Martin GM, Austad SN, Johnson TE: Genetic analysis of ageing: role of oxidative damage and environmental stresses. Nature Genetics 13:25-34, 1996. Orgel LE: The maintenance of the accuracy of protein synthesis and its relevance to aging. Proc Nat Acad Sci. USA, 49:517- 521 et 67: 1476. 1963. Peterszegi G, Robert L: Cell death induced in lymphocytes expressing the elastin-laminin receptor by excess agonists: necrosis and apoptosis. Biomed & Pharmacother 52:369-377, 1998. Peterszegi G, Tixier S, Robert L: Cell death by overload of the elastin-laminin receptor on human activated lymphocytes: protection by lactose and melibiose. Eur J Clin Invest 29:166-172, 1999. Peterszegi G, Texier S, Robert L: Human helper and memory lymphocytes exhibit an inducible elastin-laminin receptor. Int Arch Allergy and Immunol 114:218-223, 1997. Peterszegi G, Texier S, Robert AM, et al: Increased elastase and cathepsin G activity in activated lymphocytes from aged patients. Role of denutrition and dementia. Arch Gerontol Geriat 25:285-298, 1997. Ponten J: Abnormal cell growth. In: Handbook of the biology of aging., Eds: Finch CE, Hayflick L). van Nostrand-Reinhold, New York, 1977. pp. 536-560. Raines EW, Ross R: Mechanisms of plaque formation: cellular changes and the possible role of growth-regulatory molecules. In: Atherosclerosis Rev.(Eds: Weber PC, Leaf A). Raven Press, New York, 1991, 23:143-152. Robert B, Derouette JC, Robert L: Mise en evidence d’une protease a activite elastolytique dans les extraits d’aortes humaines et animales. C R Acad Sci 278:3251-3254, 1974. Robert L, Robert AM: Elastin, elastase and arteriosclerosis. In:“Frontiers of Matrix Biologie“., Eds: Robert AM, Robert L). Karger, Basel, 1980, 8:130-173 Robert L: Aging of the vascular-wall and atherosclerosis. Experimental Gerontology 34:491-501, 1999. Robert L: Aging of vascular wall and atherogenesis. Role of the elastin-laminin receptor. Atherosclerosis 123:169-179, 1996. Robert L, Jacob MP, Fulop T, Jr, et al: Elastonectin and the elastin receptor. Pathol Biol 37:736-741, 1989. Robert L: Le fibroblaste, definition de son phenotype par son „programme“ de biosynthese de la matrice extracellulaire. Pathol Biol 40:851-858, 1992. Robert L: Le vieillissement du cerveau et les demences, 1998, Flammarion, Paris. Robert L: Le vieillissement. 1994, CNRS, Belin, Paris. Robert L: Les horloges biologiques. 1989, Flammarion, Paris. Robert L: Mecanismes cellulaires et moleculaires du vieillissement. 1983, Masson, Paris. Robert L: Mechanisms of aging of the extracellular matrix. Role of the elastin-laminin receptor. Novartis Price Lecture Gerontology 44:307-317, 1998. Robert L, Robert AM, Jacotot B: Elastin-elastase atherosclerosis revisited. Atherosclerosis 140:281-295, 1998. Roth GS: Changes in tissue reesponsiveness to hormones and neurotransitters during aging. Exper Gerontol 30:361-368, 1995. Svanborg A: Health and vitality. 1988, MFR, Gotheborg. Svanborg A: Seventy-year old people in Gotheborg. A population study in an industrialized Swedish city II. General presentation of social and medical conditions. Acta Med Scand 611:3, 1977. Timar J, Diczhazi Cs, Ladanyi A, et al: Interaction of tumour cells with elastin and the metastatic phenotype, In: “The molecular biology and pathology of elastic tissues”,, Ed: Robert L). John Wiley & Sons, Chichester 1995, pp. 321-337 Urry DW:, Sequential polypeptides of elastin: structural properties and molecular pathologies. In: Frontiers of Matrix Biology, Eds:Robert AM, Robert L). Karger, Basel, 1980. 8:78-103. Varga Z, Jacob MP, Csongor J, et al: Altered phosphatidylinositol breakdown after K-elastin stimulation in PMBls of elderly. Mech Age Dev 52:61-70, 1990. Varga Z, Jacob MP, Robert L, Fulop T, Jr: Identification and signal transduction mechanism of elastin peptide receptor in human leukocytes. FEBS Letters 258:5-8, 1989. Varga Z, Kovacs EM, Paragh G, et al: Effect of elastin peptides and N-Formyl-Methionyl-Leucyl phenylalanine on cytosolic free calcium in polymorphonuclear leukocytes of healthy middle- aged and elderly subjects. Clin Biochem 21:127-130, 1988. Varga Zs, Jacob MP, Robert L: Age-dependent changes of kelastin stimulated effector functions of human phagocytic cells: relevance for atherogenesis Exper Gerontol 32: 653-662, 1997. Vaziri H, Schachter F, et al: Loss of telomeric DNA during aging of normal and trisomy 21 human lymphocytes. Am J Genet 52:661-667, 1993. Verzar F: Aging of the collagen fiber. In: International review of connective tissue research., Ed: Hall DA). Academic Press, New York, 1964, 2:243-300. Virchow R., Ackerknecht: Virchow-bibliographie,, Ed: Schwalbe J). Arno Press, New York, 1981. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 3 EP 9 PG 7 ER PT J AU Marchisone, Ch Del Grosso, F Masiello, L Prat, M Santi, L Noonan, MD AF Marchisone, Chiara Del Grosso, Federica Masiello, Luciana Prat, Maria Santi, Leonardo Noonan, M Douglas TI Phenotypic Alterations in Kaposi’s Sarcoma Cells by Antisense Reduction of Perlecan SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Kaposi-sarcoma; perlecan; antisense ID Kaposi-sarcoma; perlecan; antisense AB Metastasis is a sequence of events including proliferation, migration, adhesion, invasion and subsequent metastatic growth of tumour cells in distant organs. We previously showed that highly metastatic variants of murine melanoma cells express higher levels of the basement membrane proteoglycan perlecan than low or non metastatic variants and expression of an antisense perlecan can reduce metastatic potential. In contrast, antisense expression of perlecan in fibrosarcoma cells was reported to enhance tumorigenesis. To better understand the role of perlecan in angiogenesis we have transfected KS-IMM, an immortalized cell line derived from a human Kaposi’s sarcoma, with an antisense perlecan construct and investigated the positive/negative role of perlecan in KS. KS-IMM cells were transfected with either empty vector (neo) or the antisense perlecan construct and clones were isolated. Immuno-blot analysis showed a reduction of perlecan levels in two (AP3 and AP4) isolated clones, in Northern blot analysis endogenous perlecan was undetectable in the AP3 and AP4 clones, while it was present in the neo control clones. AP clones had a reduced migration to HGF in Boyden chambers as compared to neo clones. Proliferation in low serum or serum-free conditions was strongly reduced in the AP clones as compared to the neo control cells. The neotransfected cells showed rapid proliferation in low serum supplemented with HGF and VEGF, while antisense transfected clones showed little response. Finally, AP-trasfected KS-IMM cells had significantly reduced migration to VEGF and HGF with respect to controls. In contrast, when the AP transfected cells were injected in nude mice they paradoxically showed enhanced tumor growth as compared to controls. Our preliminary data indicate that perlecan reduction plays a crucial role on Kaposi’s sarcoma cell migration and proliferation in vitro. However, in vivo KS-IMM depleted of perlecan had a growth advantage. A possible hypothesis is that perlecan is necessary for growth of KS-IMM cells in vitro, however its down-regulation might promote angiogenesis through increased angiogenic growth factor diffusion, resulting in enhanced tumor growth in vivo. C1 [Marchisone, Chiara] Istituto Nazionale per la Ricerca sul Cancro, Modulo di Progressione Neoplastica, c/o Centro di Biotecnologie Avanzate, Largo Rosana Benzi, n. 10, 16132 Genova, Italy. [Del Grosso, Federica] Centro di Biotecnologie AvanzateGenova, Italy. [Masiello, Luciana] Istituto Nazionale per la Ricerca sul Cancro, Modulo di Progressione Neoplastica, c/o Centro di Biotecnologie Avanzate, Largo Rosana Benzi, n. 10, 16132 Genova, Italy. [Prat, Maria] Universita del Piemonte Orientale A. Avogadro, Dep. of Medical Sciences, Laboratorio di IstologiaNovara, Italy. [Santi, Leonardo] Istituto Nazionale per la Ricerca sul CancroGenova, Italy. [Noonan, M Douglas] Istituto Nazionale per la Ricerca sul Cancro, Modulo di Progressione Neoplastica, c/o Centro di Biotecnologie Avanzate, Largo Rosana Benzi, n. 10, 16132 Genova, Italy. RP Noonan, MD (reprint author), Istituto Nazionale per la Ricerca sul Cancro, Modulo di Progressione Neoplastica, 16132 Genova, Italy. EM noonan@ermes.cba.unige.it CR Adatia R, Albini A, Giunciuglio D, et al: Suppression of invasive behaviour of melanoma cells by stable expression of antisense perlecan cDNA. Annals Oncology 8:1257-1261, 1997. Albini A: Tumor and endothelial cell invasion of basement membranes. Pathology Oncology Res. 4:1-12, 1998. Albini A, Paglieri I, Orengo G, et al: The b-core fragment of human Chorionic Gonadotropin, hCG, inhibits growth of Kaposi’s sarcoma-derived cells and a new immortalized Kaposi’s sarcoma cell line. AIDS 11:713-721, 1997. Arikawa-Hirasawa E, Watanabe H, Takami H, et al: Perlecan is essential for cartilage and cephalic development. Nat Genet 23:354-358, 1999. Armes J: A review of Kaposi’s sarcoma. Advan. Cancer Res. 53:73-87, 1989. Aviezer D, Hecht D, Safran M, et al: Perlecan, basal lamina proteoglycan, promotes basic fibroblast growth factor-receptor binding, mitogenesis, and angiogenesis. Cell 79:1005-1013, 1994. Aviezer D, Iozzo RV, Noonan DM, et al: Suppression of autocrine and paracrine functions of basic fibroblast growth factor by stable expression of perlecan antisense cDNA. Mol. Cell Biol. 17:1938-1946, 1997. Aviezer D, Levy E, Safran M, et al: Differential structural requirements of heparin and heparan sulfate proteoglycans that promote binding of basic fibroblast growth factor to its receptor. J Biol Chem 269:114-121, 1994. Chomczynski P, Sacchi, N: Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal. Biochem. 162:156-159, 1987. Cornali E, Zietz C, Benelli R, et al: Vascular endothelial growth factor regulates angiogenesis and vascular permeability in Kaposi’s sarcoma. Am J Path 149:1851-1869, 1996. Costell M, Gustafsson E, Aszodi A, et al: Perlecan maintains the integrity of cartilage and some basement membranes. J Cell Biol 147:1109-1122, 1999. Ensoli B, Gendelman R, Markham P, et al: Synergy between basic fibroblast growth factor and HIV-1 tat protein in induction of Kaposi’s sarcoma. Nature 371:674-680, 1994. Ferrara N: Molecular and biological properties of vascular endothelial growth factor. J Mol Med 77:527-543, 1999. Flore O, Rafii S, Ely S, et al: Transformation of primary human endothelial cells by Kaposi’s sarcoma- associated herpesvirus. Nature 394:588-92, 1998. Gallo RC: Some aspects of the pathogenesis of HIV-1-associated Kaposi’s sarcoma. J Natl Cancer Inst Monogr 1998:55-57, 1998. Gunning P, Leavitt J, Muscat G, et al: A human b-actin expression vector system directs high-level accumulation of antisense transcripts. Proc Natl Acad Sci USA 84:4831-4835, 1987. Inki P, Jalkanen M: The role of syndecan-1 in malignancies. Ann Med 28:63-67, 1996. Joseph SJ, Ford MD, Barth C, et al: A proteoglycan that activates fibroblast growth factors during early neuronal development is a perlecan variant. Development 122:3443-3452, 1996. Kjellen L, Lindahl, U: Proteoglycans: structures and interactions. Annu Rev Biochem 60:443-475, 1991. Maier JAM, Mariotti M, Albini A, et al: Over-expression of Hepatocyte Growth Factor in human Kaposi’s Sarcoma. Int J Cancer 65:168-172, 1996. Mali M, Elenius K, Miettinen HM, et al: Inhibition of basic fibroblast growth factor-induced growth promotion by overexpression of syndecan-1. J Biol Chem 268:24215-24222, 1993. Masood R, Cai J, Zheng T, et al: Vascular endothelial growth factor/vascular permeability factor is an autocrine growth factor for AIDS-Kaposi sarcoma. Proc Natl Acad Sci USA 94:979- 984, 1997. Mathiak M, Yenisey C, Grant DS, et al:A role for perlecan in the suppression of growth and invasion in fibrosarcoma cells. Cancer Res 57:2130-2136, 1997. Naidu YM, Rosen EM, Zitnick R, et al: Role of scatter factor in the pathogenesis of AIDS-related Kaposi Sarcoma. Proc Natl Acad Sci USA 91:5281-5285, 1994. Noonan D, Albini A, Horigan E, et al: The basement membrane heparan sulfate proteoglycan: structure and expression in melanoma cells. Proceedings of the AACR 31:82, 1990. Noonan DM, Fulle A, Valente P, et al: The complete sequence of perlecan, a basement membrane heparan sulfate proteoglycan, reveals extensive similarity with laminin A chain, low density lipoprotein-receptor and the neural cell adhesion molecule. J Biol Chem 266:22939-22947, 1991. Nurcombe V, Ford MD, Wildschut JA, et al: Developmental regulation of neural response to FGF-1 and FGF-2 by heparan sulfate proteoglycan. Science 260:103-106, 1993. Rapraeger AC, Krufka A, Olwin BB: Requirement of heparan sulfate for bFGF-mediated fibroblast growth and myoblast differentiation. Science 252:1705-1708, 1991. Sakata H, Stahl SJ, Taylor WG, et al: Heparin binding and oligomerization of hepatocyte growth factor/scatter factor isoforms. Heparan sulfate glycosaminoglycan requirement for Met binding and signaling. J Biol Chem 2729457-9463, 1997. Schulz TF: Epidemiology of Kaposi’s sarcoma-associated Herpesvirus/ human Herpesvirus 8. Adv Cancer Res 76:121-160, 1999. Sharma B, Handler M, Eichstetter I, et al: Antisense targeting of perlecan blocks tumor growth and angiogenesis In vivo. J Clin Invest 102:1599-608, 1998. Steinfeld R, Van Den Berghe H, David G: Stimulation of fibroblast growth factor receptor-1 occupancy and signaling by cell surface-associated syndecans and glypican. J Cell Biol 133:405-416, 1996. Tessler S, Rockwell P, Hicklin D, et al: Heparin modulates the interaction of VEGF165 with soluble and cell associated flk-1 receptors. J Biol Chem 289:12456-12461, 1994. Yanagishita M, Hascall, VC: Cell surface heparan sulfate proteoglycans. J Biol Chem 267:9451-9454, 1992. Yayon A, Klagsbrun M, Esko JD, et al: Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinty receptor. Cell 64:841-848, 1991. Zhou FY, Owens RT, Hermonen J, et al: Is the sensitivity of cells for FGF-1 and FGF-2 regulated by cell surface heparan sulfate proteoglycans? Eur J Cell Biol 73:166-174, 1997. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 10 EP 17 PG 8 ER PT J AU Molnar, L Berki, T Hussain, A Nemeth, P Losonczy, H AF Molnar, Lenke Berki, Timea Hussain, Alizadeh Nemeth, Peter Losonczy, Hajna TI Detection of TNFa Expression in the Bone Marrow and Determinationof TNFa Production of Peripheral Blood Mononuclear Cells in Myelodysplastic Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Myelodysplastic syndrome; MDS; apoptosis; TNFa ID Myelodysplastic syndrome; MDS; apoptosis; TNFa AB TNFa is a highly active cytokine which plays an important role in the regulation of apoptotic cell death, a mechanism involved in the pathophysiology of myelodysplastic syndrome (MDS). In this study we investigated the expression of TNFa of the bone marrow trephine biopsies by immunohistochemical method and the TNFa production of peripheral blood mononuclear cells by ELISA method in 15 patients affected by MDS. Five of seven patients without excess of blasts showed high or intermediate TNFa expression in the bone marrow biopsies, whereas two patients with excess of blasts were negative and one had low expression. The five CMML patients revealed low or intermediate expression. The production of TNFa by the PBMC was analysed in 10 patients, four patients with RA and two with CMML produced higher level of TNFa which increased after stimulation with phorbol myristic acetate, but none of the RAEB patients revealed increase in TNFa production. In conclusion we suppose that increased TNFa expression and production by PBMC may be an indirect evidence of the role of increased apoptosis in low risk MDS patients. C1 [Molnar, Lenke] University of Pecs, I. Department of Internal Medicine, Ifjusag str. 13, 7624 Pecs, Hungary. [Berki, Timea] University of Pecs, Faculty of Medicine, Department of Immunology and BiotechnologyPecs, Hungary. [Hussain, Alizadeh] University of Pecs, I. Department of Internal Medicine, Ifjusag str. 13, 7624 Pecs, Hungary. [Nemeth, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and BiotechnologyPecs, Hungary. [Losonczy, Hajna] University of Pecs, I. Department of Internal Medicine, Ifjusag str. 13, 7624 Pecs, Hungary. RP Molnar, L (reprint author), University of Pecs, I. Department of Internal Medicine, 7624 Pecs, Hungary. CR Alexandrakis M, Coulocheri S, Xylouri I, et al: Elevated serum TNF-a concentrations are predictive of shortened survival in patients with high-risk myelodysplastic syndromes. Haematologia 29:13-24, 1998. Bebok Zs, Markus B, Nemeth P: Prognostic relevances of transforming growth factor alpha, TGFa, and tumor necrosis factor alpha, TNFa, detected in breast cancer tissues by immunohistochemistry. Breast Cancer Res Treat 29:229-235, 1994. Broxmeyer HE, Williams DE, Lu I, et al: The suppressive influence of human tumor necrosis factor on bone marrow hematopoietic progenitor cells from normal donors and patients with leukemia: synergysm of tumor necrosis factor and interferon- gamma. J Immunol 136, 4487-4495, 1986. Feneaux P: Molecular biology and apoptosis in myelodysplastic syndromes. ISH-EHA, Amsterdam, Education Session, 81- 84, 1998. Ganser A: Myelodysplastic syndromes, MDS): therapeutic drug interactions in the anti-death programme. Trends in Onco- Hemat. 6. 26-29, 1998. Gersuk GM, Beckham C, Loken MR, et al: A role for tumour necrosis factor-a, Fas and Fas-ligand in marrow failure associated with myelodysplastic syndrome. Br J Haemat 103:176- 188, 1998. Han J, Thompson P, Beutler B: Dexamethasone and pentoxifylline inhibit endotoxin-induced cachectin/tumor necrosis factor synthesis at separate points in the signaling pathway. J Exp Med 172:391-394, 1990. Kitagawa M, Saito I, Kuwata T, et al: Overexpression of tumor necrosis factor, TNF)-aand interferon, IFN)-gby bone marrow cells from patients with myelodysplastic syndromes. Leukemia 11:2049-2054, 1997. Koike M, Ishiyama T, Tomoyasu S, et al: Spontaneous cytokine overproduction by peripheral blood mononuclear cells from patients with myelodysplastic syndromes and aplastic anemia. Leuk Res 19:639-644, 1995. List AF, Brasfield F, Heaton R, et al: Stimulation of hematopoiesis by amifostine in patients with myelodysplastic syndrome. Blood 90:3364-3369, 1997. Maciejewski J, Selleri C, Anderson S, et al: Fas antigen expression on CD34+ human marrow cells is induced by interferon g and tumor necrosis factor and potentiates cytokine-mediated hematopoietic suppression in vitro. Blood 85:3183-3190, 1995. Metcalf D: The molecular control of cell division, differentiation commitent and maturation in haemopoietic cells. Nature 339:27-30, 1989. Musto P, Sanpaolo G, D’Arena G, et al. Serum levels of cytokines with hematopoietic activity in myelodysplastic syndromes, MDS). Brit J Haemat 102/1, suppl, 343 1998. Ogata K, Tamura H, Yokose N, et al: Effects of interleukin-12 on natural killer cell cytotoxicity and the production of interferon- g and tumour necrosis factor-a in patients with myelodysplastic syndromes. Br J Haemat 90:15-21, 1995. Ohmori S, Ohmori M, Yamagishi M, et al: MDS-macrophage derived inhibitory activity on myelopoiesis of MDS abnormal clones. Br J Haemat 83:388-391, 1993. Parker JE, Foshlock KL, Mijovic A, et al: ‘Low risk’ myelodysplastic syndrome is associated with excessive apoptosis and an increased ratio of pro- versus anti-apoptotic bcl-2-related proteins. Br J Haematol 103:1075-1082, 1998. Peddie CM, Wolf R, McLellan LI, et al: Oxidative DNA damage in CD34+ myelodysplastic cells is associated with intracellular redox changes and elevated plasma tumour necrosis factorconcentration. Br J Haemat 99:625-631, 1997. Rajapaksa R, Ginzton N, Rott LS, et al: Altered oncoprotein expression and apoptosis in myelodysplastic syndrome marrow cells. Blood 88:4275-4287, 1996. Raza A, Gezer S, Mundle S, et al: Apoptosis in the bone marrow biopsy samples involving stromal and hematopoietic cells in 50 patients with myelodysplastic syndromes. Blood 86:268- 276, 1995. Raza A, Gregory SA, Preisler HD: The myelodysplastic syndromes in 1996: complex stem cell disorders confounded by dual actions of cytokines. Leuk Res 20:881-890, 1996. Raza A, Mundle S, Iftikar A, et al: Simultaneous assessment of cell kinetics and programmed cell death in bone marrow biopsies of myelodysplastic syndrome reveals extensive apoptosis as the probable basis for ineffective hematopoiesis. Am J Hemat 48:143-154, 1995. Sachs I: The control of hematopoiesis and leukemia: from basic biology to the clinic Proc Natl Acad Sci USA 93:4742-4749, 1996. Seipelt G, Ganser A, Duranceyk H, et al: Induction of TNF-a in patients with myelodysplastic syndromes undergoing treatment with interleukin-3. Brit J Haemat 84:749-751, 1993. Shetty V, Mundle S, Alvi S, et al: Measurement of apoptosis, proliferation and three cytokines in 46 patients with myelodysplastic syndromes. Leuk Res 20:891-900, 1996. Stasi R, Brunetti M, Bussa S, et al: Serum levels of tumour necrosis factor-alpha predict response to recombinant human erythropoietin in patients with myelodysplastic syndrome. Clin Lab Haematol 19:197-201, 1997. Tsoplou P, Kouraklis-Symoenidis A, Thanopoulou E, et al: Apoptosis in patients with myelodysplastic syndromes: differential involvement of marrow cells in ‘good’ versus ‘poor’ prognosis patients and correlation with apoptosis-related genes. Leukemia 13:1554-1563, 1999. Verhoef GEG, De Schouwer P, Ceuppens JL, et al: MA: Measurement of serum cytokine levels in patients with myelodysplastic syndromes. Leukemia 6:1268-1272, 1992. Visani G, Zauli G, Tosi P, et al: Impairment of GM-CSF production in myelodysplastic syndromes Br J Haemat 84:227- 231, 1993. Yoshida Y, Mufti GJ: Apoptosis and its significance in MDS: controversies revisited. Leuk. Res 23:777-785, 1999. Zoumbos N, Symeonidis A, Kourakli A, et al:Increased levels of soluble interleukin-2 receptors and tumor necrosis factor in serum of patients with myelodysplastic syndromes. Blood 77:413-414, 1991. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 18 EP 23 PG 6 ER PT J AU Kato, Y Frankenne, F Noel, A Sakai, N Nagashima, Y Koshika, Sh Miyazaki, K Foidart, JM AF Kato, Yasumasa Frankenne, Francis Noel, Agnes Sakai, Naoki Nagashima, Yoji Koshika, Shinri Miyazaki, Kaoru Foidart, Jean-Michel TI High Production of SPARC/osteonectin/BM-40 in Mouse Metastatic B16 Melanoma Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SPARC; osteonectin; BM-40; melanoma; metastasis ID SPARC; osteonectin; BM-40; melanoma; metastasis AB Production of SPARC/osteonectin/BM-40 was determined in mouse B16 melanoma clones BL6 and F10 (high metastatic) and F1 (low metastatic). SPARC was produced greater amount in BL6 and F10 than in F1 cells, showing a good agreement with their metastatic potentials. Moreover, SPARC production was not influenced by culture pH, even in the acidic conditions (=pH 5.9). Although tumor tissues show often low pH due to excessive amount of acidic metabolites such as lactate, most studies have been done in neutral pH. High SPARC production in the acidic medium, therefore, is thought to be an important potential for tumor invasive behaviour. C1 [Kato, Yasumasa] University of Liege, Faculty of Medicine, Laboratory of Tumor and Developmental BiologyLiege, Belgium. [Frankenne, Francis] University of Liege, Faculty of Medicine, Laboratory of Tumor and Developmental BiologyLiege, Belgium. [Noel, Agnes] University of Liege, Faculty of Medicine, Laboratory of Tumor and Developmental BiologyLiege, Belgium. [Sakai, Naoki] Yokohama City University School of Medicine, Department of UrologyYokohama, Japan. [Nagashima, Yoji] Yokohama City University School of Medicine, Department of PathologyYokohama, Japan. [Koshika, Shinri] Kanagawa Dental College, Department of Biochemistry, 82 Inaoka-cho, 238-8580 Yokosuka, Japan. [Miyazaki, Kaoru] Yokohama City University, Kihara Institute for Biological Research, Division of Cell BiologyYokohama, Japan. [Foidart, Jean-Michel] University of Liege, Faculty of Medicine, Laboratory of Tumor and Developmental BiologyLiege, Belgium. RP Kato, Y (reprint author), University of Liege, Faculty of Medicine, Laboratory of Tumor and Developmental Biology, Liege, Belgium. EM yasumasa@kdcnet.ac.jp CR Termine JD, Kleinman HK, Whitson SW, et al: Osteonectin, a bone-specific protein linking mineral to collagen. Cell 26:99- 105, 1981. Mason IJ, Taylor A, Williams JG, et al: Evidence from molecular cloning that SPARC, a major product of mouse embryo parietal endoderm, is related to an endothelial cell 'culture shock' glycoprotein of Mr 43,000. EMBO J 5:1465-1472, 1986. Mann K, Deutzmann R, Paulsson M, Timpl R: Solubilization of protein BM-40 from a basement membrane tumor with chelating agents and evidence for its identity with osteonectin and SPARC. FEBS Lett 218:167-172, 1987. Bellahcene A, Castronovo V: Increased expression of osteonectin and osteopontin, two bone matrix proteins, in human breast cancer. Am J Pathol 146:95-100, 1995. Porte H, Chastre E, Prevot S, et al: Neoplastic progression of human colorectal cancer is associated with overexpression of the stromelysin-3 and BM-40/SPARC genes. Int J Cancer 64:70-75, 1995. Porte H, Triboulet JP, Kotelevets L, et al: Overexpression of stromelysin-3, BM-40/SPARC, and MET genes in human esophageal carcinoma: implications for prognosis. Clin Cancer Res 4:1375-1382, 1998. Ledda F, Bravo AI, Adris S, et al: The expression of the secreted protein acidic and rich in cysteine, SPARC, is associated with the neoplastic progression of human melanoma. J Invest Dermatol 108:210-214, 1997. Massi D, Franchi A, Borgognoni L, et al: Osteonectin expression correlates with clinical outcome in thin cutaneous malignant melanomas. Hum Pathol 30:339-344, 1999. Rempel SA, Golembieski WA, Ge S, et al: SPARC: a signal of astrocytic neoplastic transformation and reactive response in human primary and xenograft gliomas. J Neuropathol Exp Neurol 57:1112-1121, 1998. Rempel SA, Ge S, Gutierrez JA: SPARC: a potential diagnostic marker of invasive meningiomas. Clin Cancer Res 5:237-241, 1999. Le Bail B, Faouzi S, Boussarie L, et al: Osteonectin/SPARC is overexpressed in human hepatocellular carcinoma. J Pathol 189:46-52, 1999. Ledda MF, Adris S, Bravo AI, et al: Suppression of SPARC expression by antisense RNA abrogates the tumorigenicity of human melanoma cells. Nature Med 3:171-176, 1997. Tremble PM, Lane TF, Sage EH, Werb Z: SPARC, a secreted protein associated with morphogenesis and tissue remodeling, induces expression of metalloproteinases in fibroblasts through a novel extracellular matrix-dependent pathway. J Cell Biol 121:1433-1444, 1993. Shankavaram UT, DeWitt DL, Funk SE, et al: Regulation of human monocyte matrix metalloproteinases by SPARC. J Cell Physiol 173:327-334, 1997. Gilles C, Bassuk JA, Pulyaeva H, et al: SPARC/osteonectin induces matrix metalloproteinase 2 activation in human breast cancer cell lines. Cancer Res 58:5529-5536, 1998. Kato Y, Sakai N, Baba M, et al: Stimulation of motility of human renal cell carcinoma by SPARC/osteonectin/BM-40 associated with type IV collagen. Invasion Metastasis 18:105-114, 1999. Jacob K, Webber M, Benayahu D, Kleinman HK: Osteonectin promotes prostate cancer cell migration and invasion: a possible mechanism for metastasis to bone. Cancer Res 59:4453- 4457, 1999. Kato Y, Nakayama Y, Umeda M, Miyazaki K: Induction of 103- kDa gelatinase/type IV collagenase by acidic culture conditions in mouse metastatic melanoma cell lines. J Biol Chem 267:11424-11430, 1992. Kato Y, Ozono S, Shuin T, Miyazaki K: Slow induction of gelatinase B mRNA by acidic culture conditions in mouse metastatic melanoma cells. Cell Biol Int 20:375-377, 1996. Martinez-Zaguilan R, Seftor EA, Seftor RE, et al: Acidic pH enhances the invasive behavior of human melanoma cells. Clin Exp Metastasis 14:176-186, 1996. Sasaki T, Gohring W, Mann K, et al: Limited cleavage of extracellular matrix protein BM-40 by matrix metalloproteinases increases its affinity for collagens. J Biol Chem 272:9237-9243, 1997. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 24 EP 26 PG 3 ER PT J AU Ongrady, J Laird, MH Szilagyi, FJ Horvath, A Bendinelli, M AF Ongrady, Joseph Laird, M Helen Szilagyi, F Joseph Horvath, Attila Bendinelli, Mauro TI Unique Morphological Alterations of the HTLV-I Transformed C8166 Cells by Infection with HIV-1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE C8166; HIV-1; leukemia; surface projections; cellular mimicry; aberrant virions ID C8166; HIV-1; leukemia; surface projections; cellular mimicry; aberrant virions AB C8166 cells express T lymphocyte markers, a monocyte-specific esterase, taxpolypeptide of HTLV-I. In spite of this transactivator, their HIV-1 yield is low. Their culture conditions were modified, and infected cells were immobilized on a poly-L-lysine sheet under semisolid overlays to study their phenotypic alterations and HIV-1 production by microscopy and electron microscopy. Another lymphoid cultures (MT-4, CEM, CEM-ss, AdCEM) similarly treated were infected with either HIV-1/RF or IIIB. Specificity of HIV-1 was compared to the effects of vesicular stomatitis virus (VSV). Unlike other cultures, HIV-1/RF infected C8166 cells in Eagle’s MEM exhibited surface projections resembling hairy leukemia cells, which was followed by balloon degeneration and apoptosis. Immobilized HIV-1 infected cultures formed flat syncytia with several interdigitating dendritic projections. Syncytia shrunk with condensed nuclear material and axon-like filaments characteristic for infected macrophages. VSV induced enlargement and necrotic lysis of all cell types. Early postinfection with HIV-1, electron microscopy revealed irreversible membrane fusion above cell nuclei, and transient fusion between filaments. Transient presence of coated vesicles containing intact HIV-1 particles, Birbeck granule-like structures of Langerhans cells, fibrillar-lamellar structures resembling hairy leukemia or Sezary cells were detected. Late postinfection, high proportion of HIV-1 bud from polarized cytoplasm was empty particle, while that bud and entrapped in cytoplasmic vacuoles contained two or multiple cores in a fused envelope. The effect of early gene products of HIV-1 on HTLV-I and C8166 cells might elicit their latent potentials for monocyte or interdigitating dendritic cells, while in the later phase HTLV-I products might alter HIV-1 virion assembly. C1 [Ongrady, Joseph] National Institute for Dermato-Venereology, Maria utca 41., 1085 Budapest, Hungary. [Laird, M Helen] University of Glasgow, Department of Veterinary PathologyGlasgow, UK. [Szilagyi, F Joseph] University of Glasgow, Institute of VirologyGlasgow, UK. [Horvath, Attila] National Institute for Dermato-Venereology, Maria utca 41., 1085 Budapest, Hungary. [Bendinelli, Mauro] University of Pisa, Department of BiomedicinePisa, Italy. RP Ongrady, J (reprint author), National Institute for Dermato-Venereology, 1085 Budapest, Hungary. EM ongjos@hotmail.com CR Beilke MA, Japa S, Vinson DG: HTLV-I and HTLV-II virus expression increase with HIV-1 coinfection. J. Acquir. Immune Defic. Syndr. Hum. Retrovir. 17:391-397, 1998. Bjornson CRR, Rietze RL, Reynolds BA, et al: Turning brain into blood: A hematopoietic fate adopted by adult neural stem cells in vivo. Science 283:534-537, 1999. Blauvelt A, Asada H, Saville MW, et al: Productive infection of denritic cells by HIV-1 and their ability to capture virus are mediated through separate pathways. J. Clin. Investig. 100:2043-2053, 1997. Charbonnier AS, Verrier B, Jacquet C, et al: In vitro HIV1 infection of CD34+ progenitor-derived dendritic/Langerhans cells at different stages of their differentiation in the presence of GM-CSF/TNF. Res. Virol. 147:89-95, 1996. Clapham PR, Weiss RA, Dalgleish AG et al: Human immunodeficiency virus infection of monocytic and T-lymphocytic cells: receptor modulation and differentiation induced by phorbol ester. Virology 158:44-51, 1987. Cloyd MW, Lynn WS: Perturbation of host cell membrane is a primary mechanism of HIV cytopathology. Virology 181:500- 511, 1991. De Vita VT, Hallman S and Rosenberg SA: Cancer. 5th ed., Lippincott- Raven Publ., Philadelphia, 1997. Dianzani F, Antonelli G, Capobianchi MR, De Marco F: Replication of human immunodeficiency virus: yield of infectious virus under single growth cycle conditions. Arch. Virol. 103:127-131, 1988. Dolei A, Biolchini A, Serra C, et al: Increased replication of Tcell- tropic HIV strains and CXC-chemokine receptor-4 induction in T cells treated with macrophage inflammatory protein, MIP)- 1, MIP-1 and RANTES -chemokines. AIDS 12:183-190, 1998. Edinger AL, Hoffman TL, Sharron M, et al: An orphan seventransmembrane domain receptor expressed widely in the brain functions as a coreceptor for human immunodeficiency virus type 1 and simian immunodeficiency virus. J. Virol. 72:7934- 7940, 1998. Eibl MM, Kupcu Z, Mannhalter JW, et al: Dual tropism of HIV- 1 IIIB for chimpanzee lymphocytes and monocytes. AIDS Res. Human Retrovir. 8:69-75, 1992. Gendelman HE, Orenstein JM, Baca LM, et al: The macrophage in the persistence and pathogenesis of HIV infection. AIDS 3:475-495, 1989. Hamaia S, Casse H, Gazzolo L, et al: The human T-cell leukemia virus type 1 rex regulatory protein exhibits an impaired functionality in human lymphoblastoid Jurkat T cells. J. Virol. 71:8514-8521, 1997. Haraguchi Y, Takeuchi Y, Hoshino H: Inhibition of plating of human T cell leukemia virus type I and syncytium-inducing types of human immunodeficiency virus type 1 by polycations. AIDS Res. Hum Retrovir 13:1517-1523, 1997. Harris JR, Kitchen AD, Harrison JF, et al: Viral release from HIV-1-induced syncytia of CD4+ C8166 cells. J Med Virol 28:81-89, 1989. Henderson ES, Lister TA, Greaves MF: Leukemia, 6th ed., W.B. Saunders Co., Philadelphia, 1996. Hildreth JEK, Subramanium A, Hampton RA: Human T-cell lymphotropic virus type 1, HTLV-1)-induced syncytium formation mediated by vascular cell adhesion molecule-1: Evidence for involvement of cell adhesion molecules in HTLV-1 biology. J Virol 71:1173-1180, 1997. Hoffman R, Benz Ejjr, Shattil SJ, et al: Haematology, 2nd ed., Churchill-Livingstone, New York, 1995. Kiernan R, Marshall J, Bowers R, et al: Kinetics of HIV-1 replication and intracellular accumulation of particles in HTLV-I transformed cells. AIDS Res. Human Retrovir. 6:743-752, 1990. Kinchington D, Barker W, Galpin S, et al: Temperature enhancement of syncytium formation by HIV and Sendai virus. J Med Virol 36:44-48, 1992. Larsen CE, Alford DR, Young LJ, et al: Fusion of simian immunodeficiency virus with liposomes and erythrocyte ghost membranes: effects of lipid composition, pH and calcium. J Gen Virol 71:1947-1955, 1990. Laurent-Crawford AG, Krust B, Muller S, et al.: The cytopathic effect of HIV is associated with apoptosis. Virology 185:829- 839, 1991. Levy JA: HIV and the pathogenesis of AIDS. ASM Press, Washington, 1998. Lodge R, Delamarre L, Lalonde J-P, et al: Two distinct oncornaviruses harbor an intracytoplasmic tyrosine-based basolateral targeting signal in their viral envelope glycoprotein. J Virol 71:5696-5702, 1997. Ludewig B, Gelderblom HR, Becker Y, et al: Transmission of HIV-1 from productively infected mature Langerhans cells to primary CD4+ T lym-phocytes results in altered T-cell responses with enhanced production of IFN-g and IL-10. Virology 215:51-60, 1996. Ludewig B, Holzmeister J, Gentile M, et al: Replication pattern of human immunodeficiency virus type 1 in mature Langerhans cells. J Gen Virol 76:1317-1325, 1995 McLain L, Dimmock NJ: A human CD4+ T-cell line expresses functional CD64, FcgRI), CD32, FcgRII), and CD16, FcgRIII, receptors but these do not enhance their infectivity of HIV-1- IgG complexes. Immunology 90:109-114, 1997. Nagata K, Ide Y, Takagi T, et al.: Complex formation of human T-cell leukemia virus type I p40tax transactivator with cellular polypeptides. J Virol 66: 1040-1049, 1992. Nowotny A: Basic excercises in immunochemistry. Springer- Verlag, Berlin-Heidelberg-New York, 1979. Ongradi J, Ceccherini-Nelli L, Szilagyi JF, et al: The influence of cell culture and storage conditions on HIV-1 infectivity and fusogenic activity. Acta Microbiol Hung 39:207-221, 1993. Ongradi J, Ceccherini-Nelli L, Pistello M, et al: Acid sensitivity of cell-free and cell-associated HIV-1: Clinical implications. AIDS Res. Human Retrovir 6:1433-1436, 1990. Ongradi J, Cunningham C, Szilagyi JF: The role of polypeptides L and NS in the transcription process of vesicular stomatitis virus New Jersey using the temprature sensitive mutant tsE1. J Gen Virol 66:1011-1023, 1985. Ongradi J, Szilagyi JF, Laird HM, et al: Studies on the course of HIV-1 infection in tissue cultures. Hung Med J 134:803-809, 1993. Poo W-J, Conrad L, Jeneway CA: Receptor-directed focusing of lymphokine release by helper T cells. Nature 332:378-380, 1988. Pope M, Betjes MGH, Romani N, et al: Conjugates of dendritic cells and memory T lymphocytes from skin facilitate productive infection with HIV-1. Cell 78:389-398, 1994. Ramazzotti E, Marconi A, Re MC, et al: In vitro infection of human epidermal Langerhans’ cells with HIV-1. Immunology 85:94-98, 1995. Randolph GJ, Beaulieu S, Lebecque S, et al: Differentiation of monocytes into dendritic cells in a model of transendothelial trafficking. Science 282:480-483, 1998. Rubin E, Farber JL: Pathology, J.B. Lippincott Co., Philadelphia, 1988. Salahuddin SZ, Markham PD, Wong-Staal F, et al: Restricted expression of human T-cell leukemia-lymphoma virus, HTLV, in transformed human umbilical cord blood leukocytes. Virology 129: 51-64, 1983. Sato H, Orenstein J, Dimitrov D, et al: Cell-to-cell spread of HIV-1 occurs within minutes and may not involve the participation of virus particles. Virology 186:712-724, 1992. Sinkovics JG: Medical Oncology, Vol. I, Marcel Dekker Inc., New York-Basel, 1986, p 126-131. Sodroski JG, Rosen CA, Haseltine WA: Trans-acting transcriptional activation of the long terminal repeat of human T lymphotropic viruses in infected cells. Science 225:381-385, 1984. Sodroski J, Goh WC, Rosen C, et al: Role of the HTLV-III/LAV envelope in syncytium formation and cytopathicity. Nature 322:470-474, 1986. Tacchetti C, Favre A, Moresco L, Meszaros P, et al: HIV is trapped and masked in the cytoplasm of lymph node follicular dendritic cells. Am J Pathol 150:533-542, 1997. The HTLV European Network: Seroepidemiology of the human T-cell leukaemia/ lymphoma viruses in Europe. J Acquir Immune Defic Syndr Hum Retrovir 13:68-77, 1996. Weissman D, Li Y, Ananworanich J, Zhou L-J, et al.: Three populations of cells with dendritic morphology exist in peripheral blood, only one of which is infectable with human immunodeficiency virus type 1. Proc Natl Acad Sci USA 92:826-830, 1995. Zucker-Franklin D, Greaves MF, Grosci CE, Marmont AM: Le cellule del sangue. Edi-Ermes, Milano, 1988. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 27 EP 37 PG 11 ER PT J AU Kaminska, J Kowalska, M Nowacki, PM Chwalinski, M Rysinska, A Fuksiewicz, M AF Kaminska, Janina Kowalska, M. Maria Nowacki, P Marek Chwalinski, G. Maciej Rysinska, Alicja Fuksiewicz, Malgorzata TI CRP, TNF-alpha, IL-1ra, IL-6, IL-8 and IL-10 in Blood Serum of Colorectal Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer; colorectal; cytokines; CRP ID cancer; colorectal; cytokines; CRP AB Blood serum cytokines: TNFa a, IL-1ra, IL-6, IL-8, IL-10 as well as CRP were investigated in patients with colorectal cancer, prior treatment and 1, 10 and 42 days after surgery. There was an increase of the levels of CRP, IL-6 and IL-10 in most patients 24 hours after surgery. The levels of IL-1ra were elevated in patients in stage C and in several patients in stage B of the disease and there was a decrease of circulating TNFa ain stage B patients. On day 10 and 42 after surgery, the levels of cytokines followed various patterns. C1 [Kaminska, Janina] Maria-Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Department of Tumor Markers, Roentgen 5, 02-781 Warsaw, Poland. [Kowalska, M. Maria] Maria-Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Department of Tumor Markers, Roentgen 5, 02-781 Warsaw, Poland. [Nowacki, P Marek] Maria-Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Department of Colorectal CancerWarsaw, Poland. [Chwalinski, G. Maciej] Maria-Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Department of Colorectal CancerWarsaw, Poland. [Rysinska, Alicja] Maria-Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Department of Tumor Markers, Roentgen 5, 02-781 Warsaw, Poland. [Fuksiewicz, Malgorzata] Maria-Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Department of Tumor Markers, Roentgen 5, 02-781 Warsaw, Poland. RP Kaminska, J (reprint author), Maria-Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Department of Tumor Markers, 02-781 Warsaw, Poland. CR Grandis JR, Snyderman CH, Johnson JT, et al: Postoperative wound infection. Cancer 70:2166-2170, 1992. Grzelak I, Olszewski WL, Zaleska M, et al: Blood cytokine levels rise even after minor surgical trauma. J Clin Immunol 16:159-164, 1996. Haupt W, Hohenberger W, Mueller R, et al: Association between preoperative acute phase response and postoperative complications. Eur J Surg 163:39-44, 1997. Jass JR: Prognostic variables in large bowel cancer. J Clin Pathol 42:1006-1007, 1989. Khan S, Raza A, Petrelli N, et al: In vivo determinations of labelling index of metastatic colorectal carcinoma and normal mucosa using intravenous infusion of bromodeoxyuridine. J Surg Oncol 39:114-118, 1988. McMillan DC, Wotherspoon HA, Fearon KCH, et al: A prospective study of tumor recurrence and the acute-phase response after apparently curative colorectal cancer surgery. Amer J Surg 170:319-322, 1995. Nakazaki H: Preoperative and postoperative cytokines in patients with cancer. Cancer 70:709-713, 1992. Tisi E, Lissoni P, Angeli M, et al: Postoperative increase in soluble interleukin-2 receptor serum levels as predictor for early recurrence in non-small cell lung carcinoma. Cancer 69:2458- 2462, 1992. Young B, Gleeson M, Cripps AW: C-reactive protein: a critical review. Pathology 23:118-124, 1991. Zhou D, Munster AM, Winchurch RA: Inhibitory effects of interleukin 6 on immunity. Possible implications in burn patients. Arch Surg 127:65-69, 1992. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 38 EP 41 PG 4 ER PT J AU Yasasever, V Tas, F Duranyildiz, D Camlica, H Kurul, S Dalay, N AF Yasasever, Vildan Tas, Faruk Duranyildiz, Derya Camlica, Hakan Kurul, Sidika Dalay, Nejat TI Serum Levels of the Soluble Adhesion Molecules in Patients with Malignant Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD 44; adhesion molecules; malignant melanoma ID CD 44; adhesion molecules; malignant melanoma AB The incidence of malignant melanoma has been steadily increasing over the past decades. CD 44 is a transmembrane glycoprotein which is implicated in a number of adhesive and migratory events. Downregulation of CD 44 is implicated in the metastatic process. P-Selectin is a member of the selectin family of cell surface molecules. The levels of P-Selectin in biological fluids may be elevated in subjects with a variety of pathological conditions. In malignant melanoma, elevation of the plasma level of soluble intercellular adhesion molecule-1 (sICAM-1) has been associated with a reduction in disease-free survival. This study was performed to investigate the differences in the serum concentrations of the adhesion molecules in patients with malignant melanoma.The study group consisted of 52 patients with malignant melanoma and 20 healthy subjects. No meaningful difference was observed for P-selectin and sICAM 1 levels. A statistically significant decrease was observed in the cancer patients for serum CD 44 levels. C1 [Yasasever, Vildan] Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. [Tas, Faruk] Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. [Duranyildiz, Derya] Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. [Camlica, Hakan] Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. [Kurul, Sidika] Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. [Dalay, Nejat] Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. RP Dalay, N (reprint author), Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. EM ndalay@yahoo.com CR Altemonte M, Colizzi F, Esposito G, et al: Circulating intercellular adhesion molecule 1 as a marker of disease progression in cutaneous melanoma. N Engl J Med 327-959, 1992. Borland G, Ross JA, Guy K: Forms and function of CD 44. Immunology 9:139–148, 1998. Guo YJ, Ma J, Wang JH, et al: Inhibition of human melanoma growth and metastasis in vitro by anti CD44 monoclonal antibody. Cancer Res 54:1561-1565, 1994. Harning R, Mainolfi E, Bystryn JC, et al: Serum levels of circulating ICAM-1 in human melanoma. Cancer Res 51:5003- 5005, 1991. Hart I, Birch M, Marshall JF:Cell adhesion receptor expression during melanoma progression and metastasis. Cancer Metastasis Rev 10:115-130, 1991. Haynes BF, Hale LP, Patton KL, et al: Measurement of an adhesion molecule as an indicator of inflammatory disease activity. Up-regulation of the receptor for hyaluronate, CD44, in rheumatoid arthritis. Arthritis Rheum 34:1434-1443, 1991. Herrlich P, Zoller M, Palls ST, et al: CD 44 splice variants: metastases meet lymphocytes. Immunol Today 14:395-399, 1993. Kageshita T, Yoshii A, Kimura T, et al: Clinical relevance of ICAM-1 expression in primary lesions and serum of patients with malignant melanoma. Cancer Res 53:4927-4932, 1993. Letner C: Introduction to statistics statistical tables: Geigy scientific tables. CIBA-GEIGY Ltd. Basel, Switzerland, 93:1982. Ley K, Tedder TF: Leukocyte interactions with vascular endothelium. New insights into selectin-mediated attachment and rolling. J Immunol 155:525- 530,1995. Mackay CR , Terpe HJ , Stauder R , et al: Expression and modulation of CD44 variant isoforms in humans. J Cell Biol 124:71-82, 1994. Manten-Horst E, Danen EHJ, Smit L, et al: Expression of CD44 splice variants in human cutaneous melanoma and melanoma cell lines is related to tumor progression and metastatic potential. Int J Cancer 64:182-188, 1995. Nooijen PT, Westphal JR, Eggermont AM, et al: Endothelial PSelectin expression is reduced in advanced primary melanoma and melanoma metastasis. Am J Pathol 152:679-682, 1998. Price EA, Coombe DR, Murray JC: Endothelial CD 44H mediates adhesion of a melanoma cell line to quiescent human endothelial cells in vitro. Int J Cancer 64:513-518, 1996. Ristamaki R, Joensuu H, Jalkanen S: Does soluble CD44 reflect the clinical behavior of human cancer. Curr Top Microbiol Immunol 213:155-166, 1996. Schadendorf D, Heidel J, Gawlik C, et al: Association with clinical outcome of expression of VLA-4 in primary cutaneous malignant melanoma as well as P-Selectin and E-Selectin on intratumoral vessels. J Natl Cancer Inst 87:366-371, 1995. Schadendorf D, Diehl S, Zuberbier T, et al: Quantitative detection of soluble adhesion molecules in sera of melanoma patients correlates with clinical stage. Dermatology 192:89-93, 1996. Sliutz G, Temfer C, Winkler S, et al: Immunohistochemical and serological evaluation of CD44 splice variants in human ovarian cancer. Br J Cancer 72:1494-1497, 1995. Sy MS, Mori H, Liu D: CD44 as a marker in human cancers. Curr Opinion Oncol 9:108-112, 1997. Tan PH, Santos EB, Rossbach HC, et al: Enhancement of natural killer activity by an antibody to CD 44. J Immunol 150:812-820, 1993. Tedder TF: The selectins: Vascular adhesion molecules. FASEB J 9:866,1995. Viac J, Gueniche A, Faure M, Claudy A. Soluble intercellular adhesion molecule 1 and malignant melanoma. Cancer Lett 72:191-194,1993. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 42 EP 45 PG 4 ER PT J AU Karak, KA Singh, R Tandon, NP Sarkar, Ch AF Karak, Kumar Asis Singh, Rajvir Tandon, N Prakash Sarkar, Chitra TI A Comparative Survival Evaluation and Assessment of Interclassification Concordance in Adult Supratentorial Astrocytic Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Brain tumors; astrocytic tumors; classification; grading; survival ID Brain tumors; astrocytic tumors; classification; grading; survival AB Classification and grading of astrocytic tumors has been the subject of several controversies and no universally accepted classification system is yet available. Nevertheless, acceptance of a common system is important for assessing prognosis as well as easy comparative evaluation and interpretation of the results of multi-center therapeutic trials. We report the results of a single center study on comparative survival evaluation along with assessment of inter-classification concordance in 102 cases of supratentorial astrocytic tumors in adults (³ ³16 years of age). Hematoxylin and eosin (H&E) stained slides of these 102 cases were reviewed independently by two pathologists and each case classified or graded according to four different classification systems viz. Kernohan, Daumas-Duport (SAM-A), TESTAST-268 and WHO. The histological grading was then correlated with the survival curves as estimated by the Kaplan-Meier method. The most important observation was that similar survival curves were obtained for any one grade of tumor by all the four classification systems. Fifty three of the 102 cases (51.9%) showed absolute grading concordance using all 4 classifications with maximum concordant cases belonging to grades 2 and 4. Intra-classification grade-wise survival analysis revealed a statistically significant difference between grade 2 and grades 3 or 4, but no difference between grades 3 and 4 in any of the classification systems. It is apparent from the results of this study that if specified criteria related to any of the classification systems is rigorously adhered to, it will produce comparable results. Hence, preferential adoption of any one classification system in practice will be guided by the relative ease of histologic feature value evaluation with maximum possible objectivity and reproducibility. We recommend the Daumas-Duport (SAM-A) system since it appears to be the simplest, most objectivized for practical application and highly reproducible with relative ease. C1 [Karak, Kumar Asis] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Singh, Rajvir] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Tandon, N Prakash] All India Institute of Medical Sciences, Department of NeurosurgeryNew Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. RP Sarkar, Ch (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM sarkarcs@hotmail.com CR Burger PC, Scheithauer BW: Tumors of the central nervous system. In: Atlas of Tumor Pathology, Third series, Fascicle 10. Eds. Rosai J, Sobin LH. Armed Forces Institute of Pathology, Washington DC, 1994. Burger PC, Vogel FS, Green SB, et al:Glioblastoma multiforme and anaplastic astrocytoma: Pathologic criteria and prognostic implications. Cancer 56:1106-1111, 1985. Daumas-Duport C, Scheithauer B, O’Fallon J, et al:Grading of astrocytomas. A simple and reproducible method. Cancer 62:2152-2165, 1988. Garcia DM, Fulling KH, Marks JE: The value of radiation therapy in addition to surgery for astrocytomas of the adult cerebrum. Cancer 55:919-927, 1985. Gol A: The relatively benign astrocytomas of the cerebrum: A clinical study of 194 verified cases. J Neurosurg 18:501-506, 1961. Kernohan JW, Mabon RF, Svien HJ, et al: A simplified classification of gliomas. Proc Staff Meet Mayo Clin 24:71-75, 1949. Kernohan JW, Sayre GP: Tumors of the central nervous system. In: Atlas of Tumor Pathology, Section 10, Vols 35 & 37. Washington DC. Armed Forces Institute of Pathology, 1952. Kim TS, Halliday AL, Hedley-Whyte ET, et al: Correlates of survival and the Daumas-Duport grading system for astrocytomas. J Neurosurg 74:27-37, 1991. Kleihues P, Burger PC, Scheithauer BW: Histological typing of tumors of the central nervous system. New York: Springer-Verlag, 1993. Krauseneck P, Mertens HG: Results of chemotherapy of malignant brain tumors in adults. In: Jellinger K, ed. Therapy of malignant brain tumors, Wien, New York. Springer 349-395, 1987. Nelson DF, Nelson JS, Davis DR, et al: Survival and prognosis of patients with astrocytoma with atypical or anaplastic features. J Neuro-Oncol 3:99-104, 1985. Nelson JS, Tsukada Y, Schoenfeld D, et al: Necrosis as a prognostic criteria in malignant supratentorial astrocytic gliomas. Cancer 52:550-554, 1983. Ringertz J: Grading of gliomas: Acta Pathol Microbiol Scand 27:51-64, 1950. Russell DS, Rubinstein LJ: Pathology of Tumors of the Nervous System. ed.5. Baltimore: Williams and Wilkins. 83-161, 1989. Scanlon PW, Taylor WF: Radiotherapy of intracranial astrocytomas: Analysis of 417 cases treated from 1960 through 1969. Cancer 5:301-307, 1979. Schmitt HP, Oberwittler C: Computer-aided classification of malignancy in astrocytomas: II. The value of categorically evaluated histologic and non-histologic features for a numerical classifier. Analytical Cell Pathol 4:409-419, 1992. Schmitt HP, Oberwittler Ch, Schad LR: Computer-aided classification of malignancy in astrocytomas. I. The value of nuclear parameters obtained by automated black and white image analysis. Analytical Cell Pathol 4:397-407, 1992. Sharma S, Karak AK, Sarkar C, et al: A grading study of gliomas using computer aided malignancy classification and histologic morphometry. J Neuro-Oncol 27:75-85, 1996. Walker MD, Green SB, Byar DP, et al: Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant glioma after surgery. N Engl J Med 303:1323-1329, 1980. Weir B: The relative significance of factors affecting post operative survival in astrocytomas grades 3 and 4. J Neurosurg 38:448-452, 1973. Zulch KJ: Histological Typing of Tumors of the Central Nervous System. Geneva: World Health Organization, 17–57, 1979. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 46 EP 52 PG 7 ER PT J AU Nemeth, Zs Somogyi, A Takacsi-Nagy, Z Barabas, J Nemeth, Gy Szabo, Gy AF Nemeth, Zsolt Somogyi, Andras Takacsi-Nagy, Zoltan Barabas, Jozsef Nemeth, Gyorgy Szabo, Gyorgy TI Possibilities of Preventing Osteoradionecrosis During Complex Therapy of Tumors of the Oral Cavity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tumors of the oral cavity; mandibular resection; radiotherapy; osteoradionecrosis; prevention ID Tumors of the oral cavity; mandibular resection; radiotherapy; osteoradionecrosis; prevention AB In recent years, there has been a dramatic increase in the number of tumors of the head and neck. Their successful treatment is one of the greatest challenges for physicians dealing with oncotherapy. An organic part of the complex therapy is preoperative or postoperative irradiation. Application of this is accompanied by a lower risk of recurrences, and by a higher proportion of cured patients. Unfortunately, irradiation also has a disadvantage: the development of osteoradionecrosis, a special form of osteomyelitis, in some patients (mainly in those cases where irradiation occurs after bone resection or after partial removal of the periosteum). Once the clinical picture of this irradiation complication has developed, its treatment is very difficult. A significant result or complete freedom from complaints can be attained only rarely. Attention must therefore be focussed primarily on prevention, and the oral surgeon, the oncoradiologist and the patient too can all do much to help prevent the occurrence of osteoradionecrosis. Through coupling of an up-to-date, functional surgical attitude with knowledge relating to modern radiology and radiation physics, the way may be opened to forestall this complication that is so difficult to cure. C1 [Nemeth, Zsolt] Semmelweis University, Department of Oral and Maxillofacial Surgery, Maria u. 52., H-1085 Budapest, Hungary. [Somogyi, Andras] Semmelweis University, Department of Radiology and OncotherapyBudapest, Hungary. [Takacsi-Nagy, Zoltan] Semmelweis University, Department of Radiology and OncotherapyBudapest, Hungary. [Barabas, Jozsef] Semmelweis University, Department of Oral and Maxillofacial Surgery, Maria u. 52., H-1085 Budapest, Hungary. [Nemeth, Gyorgy] Semmelweis University, Department of Radiology and OncotherapyBudapest, Hungary. [Szabo, Gyorgy] Semmelweis University, Department of Oral and Maxillofacial Surgery, Maria u. 52., H-1085 Budapest, Hungary. RP Nemeth, Zs (reprint author), Semmelweis University, Department of Oral and Maxillofacial Surgery, H-1085 Budapest, Hungary. CR Aitasalo K, Niinikoski J, Grenman R, et al: A modified protocol for early treatment of osteomyelitis and osteoradionecrosis of the mandible. Head-Neck 20:411-417, 1988. Bachmann G, Rossler R, Klett R, et al: The role of magnetic resonance imaging and scintigraphy in the diagnosis of pathologic changes of the mandible after radiation therapy. Int J Oral Maxilofac Surg 25:189-195, 1996. Behar RA, Martin PJ, Fee WE, et al: Irridium-192 interstitial implant and external beam radiation therapy in the management of squamous cell carcinomas of the tonsil and soft palate. Int J Radiat Oncol Biol Physics 28:221-227, 1994. Bras J, Jonge de HKT, Merkesteyn JPR: Osteoradionecrosis of the mandible: pathogenesis. Amer J Otolaryngology 11:244- 250, 1990. Epstein JB, Mc Bride BC, Stevenson-Moore P, et al: The efficacy of chlorhexidine gel in reduction os Streptocossus mutans and Lactobacillis species in patients treated with radiation therapy. Oral Surgery, Oral Medicine, Oral Pathology 71:172-178, 1991. Happonen RR, Viander M, Pelliniemi L, et al: Actinomyces israelii in osteoradionecrosis of the jaws. Histopathologic and immunocytochemical study of five cases. Oral Surgery, Oral Medicine, Oral Pathology 55:580-588, 1983. Hartmann A, Almeling M, Carl UM: Hyperbaric oxygenation, HBO, in the treatment of radiogenic side effects. Strahlenther Oncol 172:641-648, 1996. Hermans R, Fossion E, Ioannides C, et al: CT findings in osteoradionecrosis of the mandible. Skeletal Radiol 25:31-36, 1988. Horiot JC, Bone MC, Ibrahim E, et al: Systemic dental management in head and neck irradiation. Int J Radiat Oncol Biol Physics 7:1025-1029, 1981. Jesse RH, Fletcher GH, Lindberg RD, et al: Cancer of the head and neck. In Cancer Patient Care at M.D. Anderson Hospital and Tumor Institute, ed. by Clark RL and Howe CD Chicago, Yearbook Medical Publishers, Inc. 92-93, 1976. Jones SJ, Boyde A, Ali NN: The resorption of biological and non-biological substrates by cultured avian and mammation osteoclasts. J Anatomy Embriology 170:247-256, 1984. Keene HJ, Fleming TJ: Prevalence of caries associated micraflora after radiotherapy in patients with cancer of the head and neck. Oral Surgery, Oral Medicine, Oral Pathology 64:421-426, 1987. Marx RE: Osteoradionecrosis: A new concept of its pathophysiology. J Oral Maxillofacial Surgery 41:283-288, 1983. McKenzie MR, Wong FLW, Epstein JB, et al: Hyperbaric oxygen and postradiation osteonecrosis of the mandible. Oral Oncology, European J Cancer 29B:201-207, 1993. Meyer I: Infectious diseases of the jaw. J Oral Surgery 28:17- 26, 1970 Morton ME: Osteoradionecrosis : A study of the incidence in the North West of England. Br J Oral Maxillofacial Surgery 24: 323-331, 1986 Murray CG, Herson J, Daly TE, et al: Radiation necrosis of the mandible: A 10 year study. Part I. Factors Influencing the onset of necrosis. Int J Radiat Oncol Biol Physics 6:543-548, 1980. Murray CG, Herson J, Daly TE, et al: Radiation necrosis of the mandible: A 10 year study. Part II. Dental factors; Onset, duration and management of necrosis. Int J Radiat Oncol Biol Physics 6:549-553, 1980. Regaud C: Sin la necrose des os attrens par un processus cancereux et traites par les radiations. Compt Rend Soc De Biol 87: 427, 1922. Santamaria E, Wei FC, Chen HC: Fibula osteoseptocutaneous flap for reconstruction of osteoradionecrosis of the mandible. Plast Reconstr Surg 101:921-929, 1998. Shaha AR, Cordeiro PG, Hidalgo DA, et al: Resection and immediate microvascular reconstruction in the management of osteoradionecrosis of the mandible. Head-Neck 19:406-11, 1997. Szabo G, Kovacs A, Dori F: Behandlung der Osteoradionekrose im Unterkieferbereich. Septische Mund-Kiefer-Gesichtschirurgie, Band XXIX. Georg Thieme Verlag, Stuttgart, New York 138-140, 1984. Szabo G, Nemeth G, Le van Tach, et al: Az also allcsont helyreallitasanak szempontjai sugarkezeles utan. Magyar Radiologia 58:9-17, 1984. van Merkesteyn JPR, Bakker DJ, Borgmeijer-Hoelen AMMJ: Hyperbaric oxygen treatment of osteoradionecrosis of the mandible. Experience in 29 patients. Oral Surgery, Oral Medicine, Oral Pathology 80:12-16, 1995. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 53 EP 58 PG 6 ER PT J AU Elpek, G Gelen, T Aksoy, HN Karpuzoglu, T Keles, N AF Elpek, Ozlem Gulsum Gelen, Tekinalp Aksoy, Hikmet Nazif Karpuzoglu, Tuncer Keles, Nuran TI Microvessel Count, Proliferating Cell Nuclear Antigen and Ki-67 Indices in Gastric Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microvessel count; angiogenesis; proliferation; gastric carcinoma ID microvessel count; angiogenesis; proliferation; gastric carcinoma AB The aim of the present study was to immunohistochemically investigate the prognostic value of neovascularization (expressed as microvessel count-MVC) and tumor cell proliferation (expressed as PCNA labeling index – PLI and Ki-67 labeling index – KLI) in gastric adenocarcinoma. Correlations with clinicopathologic features were also evaluated. Tumor specimens from 74 patients diagnosed as gastric adenocarcinoma were included in this study. Formalin fixed, paraffin embedded tissue sections stained immunohistochemically with F-VIII, PC10 and MIB-1 monoclonal antibodies. By ocular grid subdivided into 100 areas, number of microvessels and PC10, MIB-1 positive and negative cells were counted at x400 magnification. Chi-square test, Kaplan-Meier method and cox regression analysis were used for statistical analysis. The results showed that, MVC and PLI had a significant correlation with invasion and lymph node metastasis. The prognosis was significantly worse in patients with high MVC (>14 ) and with high PLI (>49%). However any relationship was not observed between KLI (38%) and clinicopathologic parameters, so KLI failed to predict the prognosis. Cox model showed that, MVC and PLI were independent prognostic variables. Ki-67 labeling index in gastric carcinomas has no prognostic relevance. However, the evaluation of microvessel count and proliferating cell nuclear antigen index in gastric carcinomas could be reliable indicators of prognosis. C1 [Elpek, Ozlem Gulsum] Akdeniz University, Faculty of Medicine, Department of Pathology, Typ Fakultesi, Patoloji ABD, Yeni Typ, Dekanlyk, 07070 Antalya, Turkey. [Gelen, Tekinalp] Akdeniz University, Faculty of Medicine, Department of Pathology, Typ Fakultesi, Patoloji ABD, Yeni Typ, Dekanlyk, 07070 Antalya, Turkey. [Aksoy, Hikmet Nazif] Akdeniz University, Faculty of Medicine, Department of Pathology, Typ Fakultesi, Patoloji ABD, Yeni Typ, Dekanlyk, 07070 Antalya, Turkey. [Karpuzoglu, Tuncer] Akdeniz University, Medical School, Department of General SurgeryAntalya, Turkey. [Keles, Nuran] Akdeniz University, Faculty of Medicine, Department of Pathology, Typ Fakultesi, Patoloji ABD, Yeni Typ, Dekanlyk, 07070 Antalya, Turkey. RP Elpek, G (reprint author), Akdeniz University, Faculty of Medicine, Department of Pathology, 07070 Antalya, Turkey. EM elpek@hipokrat.med.akdeniz.edu.tr CR Bicknell R, Harris AL: Novel growth regulatory factors and tumor angiogenesis. Eur J Cancer 27:781-74, 1991. Braun N, Papadopoulos, Muller-Hermelink HK: Cell cycle dependent distribution of the proliferation-associated Ki-67 antigen in human embryonic lung cells. Virchows Archiv Cell Pathol 56:25-33, 1988. Bravo R, MacDonald-Bravo H:Existence of two populations of cyclin/proliferating cell nuclear antigen during cell cycle: association with DNA replication sites. J Cell Biol 105:1549-54, 1987. Brien TP, Depowski PL, Sheehan CE: Prognostic factors in gastric cancer. Mod Pathol 11:870-877, 1998. Broll R, Mahlke C, Best R, et al: Assessment of the proliferation index in gastric carcinomas with the monoclonal antibody MIB 1. J Cancer Res Clin Oncol 124:49-54, 1998. Brown DC, Gatter KC: .Monoclonal antibody: Its use in histopathology. Histopathology 17:489-503, 1990. Celis J, Madsen P: Increased nuclear cyclin/PCNA antigen staining of non-S phase transformed human amnion cells engaged in nucleotide excision DNA repair. FEBS Lett 209:277-283, 1986. de Jong JS, van Diest PJ, Baak JP: Heterogeneity and reproducibility of microvessel counts in breast cancer. Lab Invest 73: 922-926, 1995. Folkman J: What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst 82:4-6, 1990. Gerdes J, Schwab U, Lemke H: Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer 31:13-20, 1983. Hall PA, Levinson DA: Review: assessment of cell proliferation in histological material. J Clin Pathol 43:184-192, 1990. Hall PA, Levison DA, Wood AL: Proliferating cell nuclear antigen, PCNA, immunolocalization in paraffin sections: an index of cell proliferation with evidence of deregulated expression in some neoplasmas. J Pathol 162:285-94, 1990. Hermaneck P, Sobin LH: IUCC classification of malignant tumors. Springer-Verlag Publ Co, Berlin ,1992. Ikeguchi M, Cai J, Yamane N: Clinical significance of spontaneous apoptosis in advanced gastric adenocarcinoma.Cancer 85:2329-2335,1999. Inada T, Imura J, Ichikawa A: Proliferative activity of gastric cancer assessed by immunostaining for proliferating cell nuclear antigen. J Surg Oncol 54:146-152, 1993. Kakeji Y, Korenaga D, Tsujitani S: Predictive value of Ki-67 and argyrophylic nucleolar organiser region staining for lymph node metastasis in gastric cancer.Cancer Res 51:3503-3506, 1991. Kakeji Y, Maehara Y, Adachi Y: Proliferative activity as a prognostic factor in Borrmann type 4 gastric carcinoma. Br J Cancer 69:749-753, 1994. Kawai T, Suzuki M, Kono S: Proliferating cell nuclear antigen and Ki-67 in lung carcinoma: Correlation with DNA flow cytometric analysis. Cancer 74:2468-2475,1994. Kikuyama S, Kubota T, Shimizu K: Ki-67 antigen expression in relation to clinicopathological variables and prognosis in gastric cancer. Oncol Rep 5:867-870, 1998. Lauren P. The two histological main types of gastric carcinoma: an attemp at a histo-clinical classification. Acta Pathol Microbiol Scand 64:31-39, 1965. Leonardi E, Girlando S, Serio C: PCNA and Ki67 expression in breast carcinoma: Correlation with clinical and biological variables. J Clin Pathol 45:416-9,1992. Lu C, Tanigawa N: Spontaneous apoptosis is inversely related to intratumoral microvessel density in gastric carcinoma. Cancer Res 57:221-224, 1997. Maeda K, Chung Y, Onoda N: Proliferating cell nuclear antigen labeling index of preoperative biopsy specimens in gastric carcinoma with special reference to prognosis. Cancer 73:528-33, 1994. Maeda K, Chung Y, Takatsuka S: Tumor angiogenesis as a predictor of recurrence in gastric carcinoma. J Clin Oncol 13:477- 481, 1995. Mangham DC, Rowlands DC, Newbold KM: Expression of proliferating cell nuclear antigen, PCNA, in gastric carcinoma: no evidence for prognostic value. J Clin Pathol 47:473-474, 1994. Moriki T, Takahashi T, Kataoka H: Proliferation marker MIB-1 correlates well with proliferative activity evaluated by BrdU in breast cancer:An immunohistochemical study including correlation with PCNA, p53, c-erbB-2 and estrogen receptor status. Pathol Int 46:953-961, 1996. Okazaki N, Yosbino M, Yoshida T: Evaluation of the prognosis for small hepatocellutar carcinoma based on tumor volume doubline time. Cancer 63:2207-2210, 1989. Oya M, Yao T, Nagai E: Metastasizing intramucosal gastric carcinomas. Well differentiated type and proliferative activity using proliferative cell nuclear antigen and Ki-67. Cancer 75:926-35, 1995. Porchen R, Kriegel A, Langen C: Assessment of proliferative activity in carcinomas of the human alimentary tract by Ki-67 immunostaining. Int J Cancer 47:686-696, 1991. Quinn CM, Wright NA: The clinical assessment of proliferation and growth in human tumours: Evaluation of methods and applications as prognostic variables. J Pathol 160:93-102, 1990. Ramires M, David L, Leitao D: Ki-67 Labelling index in gastric carcinomas. An immunohistochemical study using double staining for the evaluation of the proliferatif activity of diffusetype carcinomas. J Pathol 182: 62- 67, 1997 Rosa JC, Mendes R, Filipe MI: Measurement of cell proliferation in gastric carcinoma: comparative analysis of Ki-67 and proliferative cell nuclear antigen, PCNA). Histochem J 24:93- 101, 1992. Rudas M, Gnant MFX, Mittlboeck M: Thymidine labeling index and Ki-67 growth fraction in breast cancer: Comparison and correlation with prognosis. Breast Cancer Res Treat 32:165-75, 1994. Schutte B, Reynders MJ, Wiggers T: Retrospective analysis of the prognostic significance of DNA content and proliferative activity in large bowel carcinoma. Cancer Res 47:5494-5496, 1987. Tanigawa N, Amaya N, Matsumura M: Extent of tumor vascularization correlates with prognosis and hematogenous metastasis in gastric carcinomas. Cancer Res 56:2671-2676, 1996. Tanigawa N, Amaya H, Matsumura M: Association of tumour vasculature with tumour progression and overall survival of patients with non-early gastric carcinomas. Br J Cancer 75:566- 571, 1997. Tipoe GL, Jin Y, White FH: The relationship between vascularity and cell proliferation in human normal and pathological lesions of the oral cheek epithelium. Eur J Cancer 1:24-31, 1996. Toschi L, Bravo R: Changes in cyclin/PCNA distribution during DNA repair synthesis. J Cell Biol 107:1623-1628, 1988. Vermeulen PB,Verhoeven D, Hubens G: Microvessel density, endothelial cell proliferation and tumor cell proliferation in human colorectal adenocarcinomas. Ann Oncol 6:59-64, 1995. Victorzon M, Roberts PJ, Hanglund C: Ki-67 immunoreactivity, ploidy and S-Phase fraction as prognostic factors in patients with gastric carcinoma. Oncology 53:182-191, 1996. Weidner N: Tumor Angiogenesis: Review of current applications in tumor prognostication. Semin Diag Pathol. 10:302-313, 1993. Xiangming C, Hokita S, Natsugoe S: Angiogenesis as an unfavorable factor related to lymph node metastasis in early gastric cancer. Ann Surg Oncol 5:585-589, 1998. Yonemura Y, Ohyama S, Sugiyama K: Growth fractions in gastric carcinomas determined with monoclonal antibody Ki-67. Cancer 65:1130-1134, 199. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 59 EP 64 PG 6 ER PT J AU Kristt, D Turner, I Koren, R Ramadan, E Gal, R AF Kristt, Don Turner, Isaac Koren, Rumelia Ramadan, Edward Gal, Rivka TI Overexpression of Cyclin D1 mRNA in Colorectal Carcinomas and Relationship to Clinicopathological Features: An In Situ Hybridization Analysis* SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cyclin D1; in situ hybridization; mRNA; colon cancer; colorectal carcinoma; survival; grade; stage ID cyclin D1; in situ hybridization; mRNA; colon cancer; colorectal carcinoma; survival; grade; stage AB Increased expression of a key cell cycle regulator, cyclin D1, may have relevance to carcinogenesis and clinicopathological characteristics of some cancers. This study represents the first application of in situ hybridization, ISH, to detect cyclin D1 mRNA in tissue sections from colorectal carcinomas. This approach was selected because of its unique potential to clarify whether increased expression of cyclin D1 mRNA correlates with clinical and pathological parameters. The ISH ofa non-radioactive oligonucleotide probe (Biogenex) was immunocytochemically detected in paraffin embedded sections from biopsy or resection specimens. Tumors ranged from well to poorly differentiated, and from stages A, B, C, and D. Ten year survival data were available on the majority of patients. Intensity of tumor and background (smooth muscle) signals were independently scored from 0 to 3. Overexpressed cyclin D1 mRNA was seen in 86% of cases compared to background. This frequency is similar to that reported for pancreatic carcinoma. The average signal intensity score in tumor foci was 1.9 with a background score of 0.05 (p<001). All cases showed specific staining judged by the cytoplasmic localization and a tumor signal:background ratio > 1. Expression did not differentiate cancers based on grade, stage or survival (p>1), but did differentiate carcinoma and severe dysplasia from mild dysplasia. We conclude that ISH of cyclin D1 mRNA is an effective and relatively specific means of detecting activity of this gene in colonic neoplasms. The high frequency of overexpression implies that gene activity by itself is not likely to predict a tumor’s biological or clinical behavior. On the other hand, these data suggest that increased cyclin D1 gene activity may be an early event in colorectal carcinogenesis. They also are consistent with findings showing cyclin D1 is inducible by a variety of oncogene products. C1 [Kristt, Don] A, Rabin Medical Center (Golda Campus), Department of Pathology, HaSharon HospitalPetach Tikvah, Israel. [Turner, Isaac] A, Rabin Medical Center (Golda Campus), Department of Pathology, HaSharon HospitalPetach Tikvah, Israel. [Koren, Rumelia] A, Rabin Medical Center (Golda Campus), Department of SurgeryPetach Tikvah, Israel. [Ramadan, Edward] A, Rabin Medical Center (Golda Campus), Department of SurgeryPetach Tikvah, Israel. [Gal, Rivka] A, Rabin Medical Center (Golda Campus), Department of Pathology, HaSharon HospitalPetach Tikvah, Israel. RP Kristt, D (reprint author), A, Rabin Medical Center (Golda Campus), Department of Pathology, Petach Tikvah, Israel. EM pdkristt@netvision.net.il CR Alperts B, Bray D, Lewis J, et al: Molecular biology of the cell. 2nd ed. Garland Publ, New York-London, 481-549, 1989. Arber N, Hibshoosh H, Moss SF, et al: Increased expression of cyclin D1 is an early event in multistage colorectal carcinogenesis. Gastroenterology 110:669-674, 1996. Bartkova J, Lukas J, Strauss M, et al: The PRAD-1/cyclin D1 oncogene product accumulates aberrantly in a subset of colorectal carcinomas. Int J Cancer 58:568-573, 1994. Bartkova J, Lukas J, Strauss M, et al: Cell cycle-related variation and tissue restricted expression of human cyclin D1 protein. J Pathol 172:237-245, 1994. Bates S, Bonetta L, MacAllan D, et al: CDK6, PLSTIRE, and CDK4, PSK-J3, are a distinct subset of the cyclin-dependent kinases that associate with cyclin D1. Oncogene 9:71-79, 1994. Betticher D, Heighway J, Hasleton PS, et al: Prognostic significance of CCND1, cyclin D1, overexpression in primary resected non-small-cell lung cancer. Br J Cancer 73:294-300, 1996. Bodrug S, Warner B, Bath M, et al: Cyclin D1 transgene impedes lymphocyte maturation and collaborates in lymphomagenesis with the myc gene. EMBO J 13:2124-2130, 1994. Chen X, Bargonetti J, Prives C: p53, through p21, WAF1/CIP1), induces cyclin D1 synthesis. Cancer Res. 55:4257-4263, 1995. Dowdy SF, Hinds PW, Louie K, et al: Physical interaction of the retinoblastoma proteins with human D cyclins. Cell 73:499-511, 1993. Filmus J, Robles AL, Shi W, et al: Induction of cyclin D1 overexpression by activated ras. Oncogene 9:3627-3633, 1994. Gansauge S, Gansauge F, Ramadani M, et al: Overexpression of cyclin D1 in human pancreatic carcinoma is associated with poor prognosis. Cancer Res 57:1634-1637, 1997. Gillett C, Fantl V, Smith R, et al: Amplification and overexpression of cyclin D1 in breast cancer detect by immunohistochemical staining. Cancer Res 54:1812-1817, 1994. Hosokawa Y, Arnold A: Mechanism of cyclin D1, CCND1, PRAD1, overexpression in human cancer cells: analysis of allele -specific expression. Genes Chromosomes Cancer 22:66- 71, 1998. Hunter T, Pines J: Cyclins and cancer II: cyclin D and cdk inhibitors come of age. Cell 79:573-582, 1994. Jiang W, Kahn SM, Tomita N, et al: Amplification and expression of the human cyclin D gene in esophageal cancer. Cancer Res 52:2980-2983, 1992. Kallakury BV, Sheehan CE, Ambros RA, et al: The prognostic significance of p34cdc2 and cyclin D1 protein expression in prostate adenocarcinoma. Cancer 80:753-763, 1997. Kristt DA, Winston GJ, Melloul, et al: Patterns of proliferative changes in crypts bordering colonic tumors: zonal histology and cell cycle marker expression. Pathol Oncol Res 5:297-303, 1999. Kuroda H, Komatsu H, Nakamura S, et al: The positive nuclear staining observed with monoclonal antibody against PRAD1/cyclin D1 correlates with mRNA expression in mantle cell lymphoma. Jpn J Cancer Res. 86:890-898, 1995. Lovec H, Sewing A, Lucibello FC, et al: Oncogenic activity of cyclin D1 revealed through cooperation with Ha-ras: Link between cell cycle control and malignant transformation. Oncogene 9:323-326, 1994. Margiotta M, Forde A, Gallery F, et al: Comparison of three commercial kits for in situ detection of viral DNA. J Histochem 19:139-142, 1996. Matsushime H, Roussel MF, Ashmun RA, et al: Colony-stimulating factor 1 regulates novel cyclins during the G1 phase of the cell cycle. Cell 65:701-715, 1991. Naitoh H, Shibata J, Kawaguchi A, et al: Overexpression and localization of cyclin D1 mRNA and antigen in esophageal cancer. Am J Pathol 146:1161-1169, 1995. Pagano M, Theodoras AM, Tam SW, et al: Cyclin D1-mediated inhibition of repair and replication DNA-synthesis in human fibroblasts. Genes Dev 8:1627-1639, 1994. Sutter T, Doi S, Carnevale KA, et al: Expression of cyclins D1 and E in human colon adenocarcinomas. J Med 28:285-309, 1997. Zhang Y-J, Jiang W, Chen CJ, et al: Amplification and overexpression of cyclin D1 in human hepatocellular carcinoma. Biochem Biophys Res Comm 196:1010-1016, 1993. Zhou P, Jiang W, Zhang Y-J, et al: Antisense to cyclin D1 inhibits growth and reverses the transformed phenotype of human esophageal cancer cells. Oncogene 11:571-580, 1995. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 65 EP 70 PG 6 ER PT J AU Nemes, B Zalatnai, A Podder, H Jaray, J Sotonyi, P Schaff, Zs Foldes, K Perner, F AF Nemes, Balazs Zalatnai, Attila Podder, Hemangshu Jaray, Jeno Sotonyi, Peter Schaff, Zsuzsa Foldes, Katalin Perner, Ferenc TI Papillary Microcarcinoma of the Thyroid Gland in Renal Transplant Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE papillary microcarcinoma; thyroid cancer; immunosuppression; renal transplantation ID papillary microcarcinoma; thyroid cancer; immunosuppression; renal transplantation AB Among organ transplant recipients there is a world wide increase in the number of de novo tumors as well as a decrease in the time of the first appearance after the transplantation. Between 1973 and the 31th of August 1999 1709 cadaver renal allograft transplantations were perfomed in our Department. Four thyroid cancers were detected among the renal transplanted patients. Two of them proved to be papillary microcarcinomas. Although the elevated risk of thyroid cancers is well established in the literature papillary microcarcinomas have never been reported before in an immunosuppressed patient. Authors highlight that the thyroid gland should always be carefully checked in organ transplant recipients, since better survival might be achieve even in the immunosuppressed population. Metastatic tumor is relatively benign which is in correlation with the literature, but there has been little experience in organ transplanted patients so far. C1 [Nemes, Balazs] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Podder, Hemangshu] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Jaray, Jeno] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Sotonyi, Peter] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Foldes, Katalin] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Perner, Ferenc] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. RP Nemes, B (reprint author), Semmelweis University, Department of Transplantation and Surgery, H-1082 Budapest, Hungary. EM nemes@mailtrans.sote.hu CR Auteliano F, Spagnoli LG, Santeusinaio G, et al: Occult carcinoma of the thyroid gland: an epidemiological study of autopsy material. Ann Ital Chir 61:141-146, 1990. Brunner FP, Landais P, Selwood NH: Malignancies after renal transplantation: the EDTA-ERA registry experience. European Dialysis and Transplantation Association-European Renal Association. Nephrol Dial Transplant 10:74-80,, suppl 1), 1995. Buccianti G, Maisonneuve P, Ravasi B, et al: Cancer among patients on renal replacement therapy: a population based survey in Lombardi, Italy. Int J Cancer 66:591-593, 1996. Carcangiu ML, Zampi G, Pupi A, et al:, 1985, Papillary Carcinoma of the thyroid. A clinicopathologic Study of 241 Cases Treated at the Universitiy of Florence, Italy. Cancer 55:805-828, 1985. Engel U, Kucharski A, Zornig C: Eingeschrankte Radikalitat bei differezierten Schilddrusencarcinomen. Eine prospektive Studie an 107 Patienten. Chirurg 65:1095-1098, 1994. Esik O, Szentirmay Z, Tusnady G, et al: Malignus pajzsmirigytumorok Magyarorszagon:morbiditas es mortalitas, Malignant thyroid tumors in Hungary:morbidity and mortality, Orv Hetil 137:905-911, 1996. Fahey TJ3rd, Reeve TS, Delbridge L: Increasing incidence and changing presentation of thyroid cancer over a 30 year period. Br J Surg 82:518-20, 1995. Franceschi S, La Vecchia C: Thyroid cancer. Cancer Surv 19- 20:393-422, 1994. Hazard JB. Small papillary carcinoma of the thyroid: A study with special reference to so-called non-encapsulated sclerosing tumor. Lab Invest 8:86-97, 1960. Hedinger C: Histological typing of thyroid tumors, 2nd edition, Springer-Verlag, Berlin. Heidelberg, New York, London, Paris, Tokyo, 1988. Hefer T, Joachims HZ, Eitan A, et al: Are the morphology of papillary thyroid carcinoma and the tumor’s behavior correlated? J Laryngol Otol 110:704-705, 1996. Klinck GH, Winship T: Occult sclerosing carcinoma of the thyroid. Cancer 8:701-766, 1955. Matsuda M, Nagumo S, Kayama H, et al: Occult thyroid cancer discovered by fine needle aspiration cytology of cervical lymph node: a report of three cases. Diagn Cytopathol 7:299-303, 1991. Pelizzo MR, Piotto A, Rubello D, et al: High prevalence of occult papillary thyroid carcinoma in surgical series for benign thyroid diseases. Tumori 76:255-257, 1990. Penn I: Neoplastic complications of transplantation. Semin Respir Infect 8:233-239, 1993. Penn I: The effect of immunosuppression on pre-existing cancers. Transplantation. 55:742-747, 1993. Sampson RJ, Key CR, Buncher CR, et al: Smallest forms of papillary carcinoma of the thyroid. Arch. Pathol. 91:334-339, 1971. Sasaki H, Sirakusa T, Suzuki K, et al: Occult papillary carcinoma of the thyroid presenting as a solitary pulmonary metastasis. Nippon Naibunpi Gakkai Zasshi 67:655-663, 1991. Stio F, Battisti G, Felisci A, et al: Thyroid cancer and its incidence in nodular thyroid diseases: our experience. G Chir 11:487-490, 1990. Torre G, Borgonovo G, Amato A, et al: Differentiated thyroid cancer: surgical treatment of 190 patients. Eur J Surg Oncol 22:276- 281, 1996. Woolner LB, Beahrs OH, Black BM, et al:Classification and prognosis of thyroid carcinoma. A study of 885 cases observed in a thirty year period. Am J Surg 102:354-387, 1981. Yamamoto Y, Maeda T, Izumi K, et al: Occult papillary carcinoma ot the thyroid. Cancer 65:1173-1179, 1990. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2000 VL 6 IS 1 BP 72 EP 75 PG 4 ER PT J AU Timar, J Toth, J AF Timar, Jozsef Toth, Jeannette TI Tumor Sinuses - Vascular Channels SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE tumor sinus; vascular channel; melanoma; breast cancer; pathomechanism ID tumor sinus; vascular channel; melanoma; breast cancer; pathomechanism AB The existence of tumor cell-lined sinuses (vascular channels) in various experimental and human cancer is known for almost a half a century described by a Hungarian pathologist, Bela Kellner. Meanwhile, even the existence as well as the pathomechanism and the possible functional significance of these sinuses are heavily challenged in the recent literature. Ultrastructural studies however provide evidence for the presence of tumor cell-lined sinuses in human melanoma and breast cancer. The generation of such sinuses can be suggested in two ways: by de novoformation, when tumor cells recapitulate an embryonic geno- and phenotype by reexpressing endothelial genes or by a secondary mechanism, where the incorporated microvessels degenerate due to the predominant expression of anti-angiogenic factors. Literature data are available for the potential diagnostic and clinical significance of the tumor sinuses (vascular channels) stimulating further studies on this issue. C1 [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Budapest, Rath Gy. u. 7-9., H-1122 Budapest, Hungary. [Toth, Jeannette] Semmelweis University, Department of OphthalmologyBudapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. CR Folkman J: Tumor angiogenesis: therapeutic implications. N Engl J Med 285:1182-1186, 1971. Hanahan D, Folkman J: Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86:353-364, 1996 Bergers G, Javaherian K, Folkman J, et al: Effects of angiogenesis inhibitors on multistage carcinogenesis in mice. Science 284:808-812,1999. Maniotis AJ, Folberg R, Hess A, et al. Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 155:739-752, 1999. Folberg R, Hendrix MJ, Maniotis AJ: Vasculogenic mimicry and tumor angiogenesis. Am J Pathol 156:361-381, 2000. Hendrix MJC, Seftor EA, Seftor REB, et al. Regulation of uveal melanoma interconverted phenotype by hepatocyte growth factor/ scatter factor, HGF/SF). Am J Pathol 152:855-863, 1998. McDonalds DM, Munn L, Jain RK:Vasculogenic mimicry: how convincing, how novel, and how significant. Am J Pathol 156:383-388, 2000. Barinaga M: A face-off over tumor blood supply. Science 287:783-785, 2000. Kellner B: Die Fettmorphologie der Sarkome. Z Krebsforsch 52:240-246, 1941. Warren BA, Shubik P: The growth of the blood supply to melanoma transplants in the hamster cheek pouch. Lab Invest 15:464-478,1966. Radnot M, Antal M: Vessels of intraocular malignant melanomas. Am J Ophthalmol 88:472-478, 1979. Sato T, Takusagawa K, Asoo N, et al. Ultrastructure of the Lewis lung carcinoma. Eur J Canc Clin Oncol 18:369-376,1982. Van Nesselrooij JH, Hendrixen GJ, Feron VJ, et al: Pathogenesis of blood-filled cavities in estrogen-induced anterior pituitary tumors in male Sprague-Dawley rats. Toxicol Pathol 20:71-80,1992. Nagano N, Sasaki H, Aoyaki M, et al: Invasion of experimental rat brain tumor: early morphological changes following microinjection of C6 glioma cells. Acta Neuropathol 86:117- 125, 1993. Wesseling P, van der Laak JA, de Leeuw H, et al. Quantitative immunohistological analysis of the microvasculature in untreated human glioblastoma multiforme. Computer-assisted image analysis of whole-tumor sections. J Neurosurg 81:902-909, 1994. Holmgren L, O’Reilly MS, Folkman J: Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1:149-153,1995. Pezzella F, Pastorino U, Tagliabue E, et al. Non-small-cell lung carcinoma tumor growth without morphological evidence of neo-angiogenesis. Am J Pathol 151:1417-1423, 1997. Holash J, Maisonpierre PC, Compton D, et al. Vessel cooption, regression and growth in tumors mediated by angiopoietins and VEGF. Science 284:1994-1998, 1999. Folberg R, Pe’er J, Gruman LM, et al: The morphologic characteristics of tumor blood vessels as a marker of tumor progression in primary human uveal melanoma: a matched casecontrol study. Hum Pathol 23:1298-1305,1992. Folberg R, Fleck M, Mehaffey MG, et al: Mapping prognostically significant vascular patterns in ciliary body and choroidal melanomas. Pathol Oncol Res 2:229-236,1996. Bissell MJ: Tumor plasticity allows vasculogenic mimicry, a novel form of angiogenic switch – a rose by any other name? Am J Pathol 155:675-679,1999. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 83 EP 86 PG 4 ER PT J AU Cox, G Walker, AR Muller, S Abrams, RK Steward, PW O’Byrne, JK AF Cox, Giles Walker, A Rosemary Muller, Salli Abrams, R Keith Steward, P William O’Byrne, J Kenneth TI Does Immunointensity Account for the Differences in Prognostic Significance of Bcl-2 Expression in Non-Small Cell Lung Cancer? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bcl-2; apoptosis; prognosis; non-small cell lung cancer ID Bcl-2; apoptosis; prognosis; non-small cell lung cancer AB Bcl-2 is an oncogenic protein that plays a central role in apoptosis. The association of Bcl-2 expression and prognosis in non-small cell lung cancer (NSCLC) is unclear, with some studies showing improved outcome whilst others show no survival advantage. We evaluated 178 surgically resected NSCLC specimens for Bcl-2 and p53 immunoexpression. Bcl-2 staining was present in 34.9% of cases (weakly staining 24.2%, strongly staining 10.7%), nuclear p53 in 43.3% and cytoplasmic p53 in 10.7%. There was no association between p53 and survival. Bcl-2 immunoexpression correlated with improved outcome (p=0.04). A sub-group of strongly Bcl-2 staining cases had a poor survival compared to those that stained weakly (p=0.01). The strongly staining cases had a similar survival to negative cases. Immunointensity may therefore account for the disparity in results regarding the prognostic significance of Bcl-2 demonstrated in previous studies. C1 [Cox, Giles] Leicester Royal Infirmary, Department of Medical Oncology, Welford Road, LE1 5WW Leicester, UK. [Walker, A Rosemary] Glenfield Hospital, Department of PathologyLeicester, UK. [Muller, Salli] Glenfield Hospital, Department of PathologyLeicester, UK. [Abrams, R Keith] University of Leicester, Department of EpidemiologyLeicester, UK. [Steward, P William] Leicester Royal Infirmary, Department of Medical Oncology, Welford Road, LE1 5WW Leicester, UK. [O’Byrne, J Kenneth] Leicester Royal Infirmary, Department of Medical Oncology, Welford Road, LE1 5WW Leicester, UK. RP O’Byrne, JK (reprint author), Leicester Royal Infirmary, Department of Medical Oncology, LE1 5WW Leicester, UK. EM ken.obyrne@lri.org.uk CR Amstad PA, Lik H, Ichimiya M, et al: bcl-2 enhancement of malignant transformation in mouse epidermal JB-6 cells. Mol Carcinogenesis 20:231-239, 1997. Anton RC, Brown RW, Younes M, et al: Absence of prognostic significance of bcl-2 immunopositivity in non-small cell lung cancer: Analysis of 427 cases. Hum Pathol 28:1079-1082, 1997. Apolinario RM, van der Valk P, de Jong JS, et al: Prognostic value of the expression of p53, bcl-2, and baxoncoproteins, and neovascularization in patients with radically resected nonsmall- cell lung cancer. J Clin Oncol 15:2456-2466, 1997. Dobbs SP, Brown LJR, Ireland D, et al: Bcl-2, platelet-derived growth factor, PD-ECGF), vascular endothelial growth factor, VEGF), and angiogenesis in cervical intraepithelial neoplasia, CIN, and squamous cell carcinoma, SCC). Br J Cancer 78:S1,49, 1998. Evan GI, Wyllie AH, Gilbert CS, et al: Induction of apoptosis in fibroblasts by c-myc protein. Cell 69:119-128,1992. Fleming MV, Guinee DG, Chu WS, et al: Bcl-2 immunohistochemistry in a surgical series of non-small cell lung cancer patients. Human Pathol 29:60-64, 1998. Fontanini G, Boldrini L, Vignati S, et al: Bcl-2 and p53 regulate vascular endothelial growth factor, VEGF)-mediated angiogenesis in non-small cell lung carcinoma. Eur J Cancer 34:718- 723, 1998. Kobayashi N, Ishii M, Ueno Y, et al: Co-expression of Bcl-2 protein and vascular endothelial growth factor in hepatocellular carcinomas treated by chemoembolization. Liver 19:25-31, 1999. Koukourakis MI, Giatromanolaki A, O’Byrne KJ, et al: Potential role of bcl-2 as a suppressor of tumor angiogenesis in nonsmall cell lung cancer. Int J Cancer 74:565-570, 1997. Lotem J, Sachs L: Regulation by bcl-2, c-myc, and p53 of susceptibility to induction of apoptosis by heat shock and cancer chemotherapy compounds in differentiation-competent and defective myeloid leukaemia cells. Cell Growth Different 4:41- 47, 1993. Minn AJ, Swain RE, Ma A, et al: Recent progress on the regulation of apoptosis by Bcl-2 family members. Advances in Immunology 70:245-279, 1998. Miyashita T, Reed JC: Bcl-2 gene transfer increases relative resistance of S49.1 and WEH17.2 lymphoid cells to cell death and DNA fragmentation induced by glucocorticoids and multiple chemotherapeutic regimes. Cancer Res 52:5407-5411, 1992. Mountain CF: Revisions in the international system for staging lung cancer. Chest 111:1711-1717, 1997. Naumovski L, Cleary ML: The p53-binding protein 53BP2 also interacts with Bcl-2 and impedes cell cycle progression at G2/M. Mol Cell Biol 16:3884-3892, 1996. Nunez G, London L, Hockenberry D, et al: Deregulated bcl-2 gene expression selectively prolongs survival of growth factordeprived haemopoeitic cell lines. J Immunol 144:3602-3610, 1990. Ohsaki Y, Toyoshima E, Fujiuchi S, et al: bcl-2 and p53 protein expression in non-small cell lung cancers: correlation with survival time. Clin Cancer Res 2:915-920, 1996. O’Neill AJ, Staunton MJ, Gaffney EF: Apoptosis ocurs independently of bcl-2 and p53 over-expression in non-small cell lung cancer. Histopathology 29:45-50, 1996. Pezzella F, Turley I, Kuzu I, et al: Bcl-2 protein in non-small cell lung carcinoma. N Eng J Med 329:690-694, 1993. Ryan JJ, Prochownik E, Gottlieb CA, et al: C-myc and bcl-2 modulate p53 function by altering p53 subcellular trafficking during the cell cycle. Proc Natl Acad Sci USA 91:5878-5882, 1994. Saegusa M, Takano Y, Hashimura M, et al: The possible role of bcl-2 expression in the progression of tumors of the uterine cervix. Cancer 76:2297-2303, 1995. Silvestrini R, Benini E, Veneroni S, et al: p53 and bcl-2 expression correlates with clinical outcome in a series of node-positive breast cancer patients. J Clin Oncol 14:1604-1610, 1996. Walker C, Robertson L, Myskow M, et al: Expression of the Bcl- 2 protein in normal and dysplastic bronchial epithelium and in lung carcinomas. Br J Cancer 72:164-169, 1995. World Health Organisation: The World Health Organisation histological typing of lung cancer. Am J Clin Pathol 77:123- 136, 1982. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 87 EP 92 PG 6 ER PT J AU Croce, VM Price, RM Segal-Eiras, A AF Croce, V Maria Price, R Mike Segal-Eiras, Amada TI Detection and Isolation of MUC1 Mucin from Larynx Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE laryngeal carcinoma; tumor antigens; MUC1mucin ID laryngeal carcinoma; tumor antigens; MUC1mucin AB The progression from uncontrolled cell proliferation to invasion and metastasis of epithelial tumors is partially understood. Alteration of epithelial mucin expression have been described in different malignant localizations but only few attempts have been made to identify mucin expression in malignant laryngeal tumors. In the present report, results are shown of studies on the expression of mucins and carbohydrate related antigens in laryngeal cancer and on the isolation of MUC1 mucin from this tumor tissue. Malignant laryngeal specimens were processed for immunohistochemical analysis and for extranuclear membrane fractions (ENM) which were obtained by ultracentrifugation. Subsequently, ENM samples were centrifuged in density-gradient; the analysis of fractions was performed by means of SDS-PAGE and Western-blotting. The panel of monoclonal antibodies (MAbs) included anti MUC1 mucin, anti Lewis x, anti sialyl Lewis x, anti Lewis y, anti MUC-5B, anti oral mucin (gp230), anti Tn hapten, anti p53 and anti cytokeratins. By immunohistochemistry, it was possible to detect MUC1 mucin, Lewis x and Lewis y showing strong reactions while sialyl-Lewis x and Tn antigen only reacted weakly in a few cells; cytokeratins were detected in all samples. In ENM derived fractions obtained by CsCl centrifugation, MUC1 was demonstrated by Western blotting. Conclusions: (1) laryngeal cancer antigenic expression comprises mostly MUC1 mucin, Lewis x, Lewis y as well as Tn antigen and (2) the methodology here employed is useful to isolate MUC1 from tumor samples. C1 [Croce, V Maria] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Price, R Mike] University of Nottingham, School of Pharmaceutical Sciences, Cancer Research LaboratoriesNottingham, UK. [Segal-Eiras, Amada] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. RP Segal-Eiras, A (reprint author), Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), La Plata, Argentina. EM as-eiras@netverk.com.ar CR Aksoy N, Corfield AP, Paraskeva C, et al: Human intestinal mucin MUC2 in in vitro two adenoma-carcinoma sequences originated from a single benign colonic adenoma cell line: In vitro evidence for the neotransformation. 5th International Workshop on Carcinoma Associated Mucins, Cambridge, UK, 1998. Barnd DL, Lan M, Metzgar R, et al: Specific, MHC-unrestricted recognition of tumor associated mucins by human cytotoxic T cells. Proc Natl Acad Sci USA; 86:7159-7163, 1989. Bradford CR: Predictive factors in head and neck cancer. Hemotol Oncol Clin North Am 13:777-785, 1999. Brown A, Feizi T, Gooi HC: A monoclonal antibody against human colonic adenoma recognizes a difucosylated type-2 blood group chains. Biosci Rep 3:163-169, 1983. Chiesa F, Mauri S, Tradati N, et al: Surfing prognostic factors in head and neck cancer at the Millennium. Oral Oncol 35:590- 596, 1999. Creeth JM, Ramakrishnan Bhaskar K, et al: The separation and characterization of bronchial glycoproteins by density-gradient methods. Biochem J 167:557-569, 1977. Croce MV, Price MR, Segal-Eiras A: Expression of monoclonal antibody-defined antigens in fractions isolated from human breast carcinoma and patients’ serum. Cancer Immunol Immunother; 40:132-137, 1995. Croce MV, Colussi AG, Price M, et al: A Immunohistopathological characterization of spontaneous metastases in a human lung mucoepidermoid adenocarcinoma, HLMC, xenograft. Pathol Oncol Res 4:259-266, 1998. Dolcetti R, Doglioni C, Maestro R, et al: A p53 over expression is an early event in the development of human squamous-cell carcinoma of the larynx: genetic and prognostic implications. Int J Cancer 52:178-182, 1992. Federici MF, Kudryashov, Saigo PE, et al: Selection of carbohydrate antigens in human epithelial ovarian cancers for immunotherapy: serous and mucinous tumors exhibit distinctive patterns of expression. Int J Cancer 81:193-198, 1999. Feickert HJ, Anger BR, Cordon-Cardo, et al: Cell-surface antigens of human lung tumors detected by mouse monoclonal antibodies: definition of blood-group- and non-blood-grouprelated antigenic systems. Int J Cancer: 46:1007-1013, 1990. Gendler SJ, Taylor-Papadimitriou J, Duhig T, et al: A highly immunogenic region of human polymorphic mucin expressed by carcinomas is made up of tandem repeats. J Biol Chem 263:12820-12823, 1988. Gendler SJ, Spicer AP: Epithelial mucin genes: Annu Rev Physiol 57:607-634, 1995. Hakomori S, Nudelman E, Levery SB, et al: Novel fucolipids accumulating in human adenocarcinoma: I. Glycolipids with dior trifucosylated type 2 chain. J Biol Chem 259:4672-4680, 1984. Hanai N, Shitara K, Yoshida H: Generation of monoclonal antibodies against human lung squamous cell carcinoma and adenocarcinoma using mice rendered tolerant to normal human lung. Cancer Res 46:4438-4443, 1986. Hilkens J, Ligtenberg JL, Vos HL, et al: Cell membrane-associated mucins and their adhesion-modulating property. Trends Biochem Sci 17:359-363, 1992. Hurlimann J, Saraga EP: Expression of p53 protein in gastric carcinomas. Am J Surg Pathol 18:1247-1253, 1994. International Union Against Cancer, UICC, Classification of Malignant Tumors. Ed. L.H. Sobin and Ch. Wittekind, 5th edition, New York, 1997. Ioannides CG, Fisk B, Jerome KR, Irimura T: Cytotoxic T cells from ovarian malignant tumors can recognize polymorphic epithelial mucin core peptides. J Immunol 151: 3693-3703, 1993. Itoh T, Yonezawa S, Nemoto M, et al: Expression of mucin antigen and Lewis x-related antigens in carcinomas and dysplasia of the pharynx and larynx. Pathol Int 46: 646-655, 1996. Jerome KR, Barnd DL, Bendt KM: Cytotoxic T-lymphocytes derived from patients with breast adenocarcinoma recognize an epitope present on the protein core of a mucin molecule preferentially expressed by malignant cells. Cancer Res 5:2908-2916, 1991. Johnson NW: Orofacial neoplasms: global epidemiology, risk factors and recommendations for research. Int Dental J 41:365, 1991. Kotera Y, Darrell Fontenot J, Pecher G, et al: Humoral immunity against a tandem repeat epitope of human mucin MUC1 in sera from breast, pancreatic and colon cancer patients. Cancer Res 54:2856-2860, 1994. Laemmli UK: Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature, Lond, 227:680- 681, 1980. McDonald LA, Walker DM, Gibbins JR: Cervical lymph node involvement in head and neck cancer detectable as expression of a spliced transcript of type II keratin K5. Oral Oncol 34:276- 283, 1998. Nees M, Homann N, Discher H, Andl T, et al: Expression of mutated p53 occurs in tumor-distant epithelia of head and neck cancer patient: a possible molecular basis for the development of multiple tumors. Cancer Res 53:4189-4196, 1993. Nielsen PA, Mandel U, Therkildsen MH, et al: Differential expression of human high-molecular-weight salivary mucin, MG1, and low-molecular–weight salivary mucin, MG2). J.Dent Res 75:1820-1826, 1996. Nielsen PA, Mandel U, Therkildsen MH, et al: Loss of a novel mucin-like epithelial glycoprotein in oral and cervical squamous cell carcinoma. Cancer Res 57:634-640, 1997. Nylander K, Stenling R, Gustafsson H, et al:p53 expression and cell proliferation in squamous cell carcinomas of the head and neck. Cancer 75:87-93, 1994. Pancino GF, Osinaga E, Vorauher W, et al: Production of a monoclonal antibody as immunohistochemical marker on paraffin embedded tissues using a new immunization method. Hybridoma 9:389-395, 1990. Parkin DM, Pisani P, Ferlay J: Estimates of the worldwide incidence of eighteen major cancers in 1985. Int J Cancer 54:594-599, 1993. Price MR, Edwards S, Owainati A, et al: Multiple epitopes on a human breast carcinoma associated antigen. Int J Cancer 36:567-574, 1985. Price MR, Pugh J, Hudecz F, et al: Immunological and structural features of the protein core of human polymorphic epithelial mucin. Mol Immunol 27:795-801, 1990. Quinlan DC, Davidson AG, Summers CL, et al: Accumulation of p53 protein correlates with a poor prognosis in human lung cancer. Cancer Res 52:4828-4831, 1992. Ravindranath NH, Nishimoto K, Chu K, et al: Density of cell surface expression of sialyl Lewis antigens and complement restriction factors in oral squamous cell carcinoma. Proc Am Ass for Cancer Res 40:357, 1999. Regimbald LH, Pilarski LM, Longenecker BM: The breast mucin MUC1 as a novel adhesion ligand for endothelial intercellular adhesion molecule 1 in breast cancer. Cancer Res 56:4244-4249, 1996. Rowley H, Poland NJ, Helliwell TR, et al: P53 protein expression in tumors from head and subsites, larynx and hypopharynx and differences in relationship to survival. Clin Otolaryngol 23:57-62, 1998. Rughetti A, Turchi V, Apollonj Ghetti C: Human B-cell immune response to the polymorphic epithelial mucin. Cancer Res 53:2457-2459, 1993. Suo Z, Holm R, Nesland JM: Squamous cell carcinomas. An immunohistochemical study of cytokeratins and involucrin in primary and metastatic tumors. Histopathol 23:45-54, 1993. Svitacheva N, Davies JR, Lesuffleur T: Characterization of mucins produced by human HT29-MTX cell line. 5th International Workshop on Carcinoma Associated Mucins Cambridge, UK, 1998. Takada A, Ohmori K, Yoneda T: Contribution of carbohydrate antigens sialyl Lewis a and sialyl Lewis x to adhesion of human cancer cells to vascular endothelium. Cancer Res 53:354-361, 1993. Taylor KL, Mall AS, Barnard RA: Immunohistochemical detection of gastric mucin in normal and disease states. Oncol Res, 10:465-473, 1998. Taylor-Papadimitriou J, Peterson J, Arklie J: Monoclonal antibodies to epithelium-specific components of the human milk fat globule membrane: production and reaction with cells in culture. Int J Cancer 28:17-21, 1981. Taylor-Papadimitriou J, Finn O: Biology, biochemistry and immunology of carcinoma associated mucins. Immunol Today 18:105-107, 1997. Thornton DJ, Devine PL, Hanski Ch, et al: Identification of two major populations of mucins in respiratory secretions. Am J Respir Crit Care Med 150:823-832, 1994. Towbin H, Staehelin T, Gordon J: Electrophoretic transfer of proteins from polyacrilamide gels to nitrocellulose sheets: procedures and some applications. Proc Natl Acad Sci USA 76:4350-4354, 1979 Whitehouse C, Burchell J, Gschmeissner S: A transfected sialyltransferase that is elevated in breast cancer and localizes to the medial/trans Golgi apparatus inhibits the development of core-2-based O-glycans. J Cell Biol 137:1229-1241, 1997. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 93 EP 99 PG 7 ER PT J AU Athanassiadou, P Gonidi, M Liossi, A Petrakakou, E Nakopoulou, Ly Zerva, Ch Athanassiades, P AF Athanassiadou, Pauline Gonidi, Maria Liossi, Anna Petrakakou, Efthalia Nakopoulou, Lydia Zerva, Cherry Athanassiades, Peter TI Moc-31, Fibronectin and CEA in the Differential Diagnosis of Malignant Effusions: An Immunocytochemical Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MOC-31; CEA; fibronectin; immunocytochemistry; effusions; reactive atypical mesothelial cells ID MOC-31; CEA; fibronectin; immunocytochemistry; effusions; reactive atypical mesothelial cells AB In discriminating benign and malignant origins of cytologically suspicious effusion smears a panel of antibodies against carcinoembryonic antigen (CEA), Fibronectin (F) and MOC-31 was used with immuno-cytochemical techniques. One hundred and thirty seven effusions were studied of which 107 had a malignant and 30 a benign aetiology as determined by clinical and histological examination. Cytologically 24 were diagnosed as benign, 97 as malignant and 14 as suspicious. Staining for F was positive in all effusions of benign and 3 of malignant origin. MOC-31 was positive in 95 (88.8%) of effusions of malignant origin but none of benign origin. Positive CEA was observed in 43% of effusions of malignant origin and in 10 of benign origin. The combination of MOC-31 positivity measured the sensitivity and specificity of the cytological examination in cases where the cytological examination result was suspicious as did F positivity improve the sensitivity for a benign origin of the effusion. Positivity or negativity for CEA is less valuable than the other parameters. C1 [Athanassiadou, Pauline] University of Athens, Cytology Department, Pathology Laboratory, 75 M.Asias Str., GR-115 27 Athens, Greece. [Gonidi, Maria] University of Athens, Cytology Department, Pathology Laboratory, 75 M.Asias Str., GR-115 27 Athens, Greece. [Liossi, Anna] University of Athens, Cytology Department, Pathology Laboratory, 75 M.Asias Str., GR-115 27 Athens, Greece. [Petrakakou, Efthalia] University of Athens, Cytology Department, Pathology Laboratory, 75 M.Asias Str., GR-115 27 Athens, Greece. [Nakopoulou, Lydia] University of Athens, Cytology Department, Pathology Laboratory, 75 M.Asias Str., GR-115 27 Athens, Greece. [Zerva, Cherry] University of Athens, Cytology Department, Pathology Laboratory, 75 M.Asias Str., GR-115 27 Athens, Greece. [Athanassiades, Peter] University of Athens, Alexandra Hospital, Clinical TherapeuticsAthens, Greece. RP Athanassiadou, P (reprint author), University of Athens, Cytology Department, Pathology Laboratory, GR-115 27 Athens, Greece. CR Bedrossian CWM: Malignant effusions: A multimodal approach to cytologic diagnosis. New York, Igaku-Shoin Medical Publishers, 1994. de Leij L, Broers J, Ramaekers F, et al: Monoclonal antibodies in clinical and experimental pathology of lung cancer. In: Ruiter DJ, Fleuren GJ, Warnaar SO, eds. Application of Monoclonal Antibodies in Tumor Pathology. Dordrecht, the Netherlands: Martinus Nijhoff Publishers, 1987:191-207. Hsu SM, Raine L, Fanger H: Use of avidin-biotin peroxidase complex, ABC, in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody, PAP, procedures. J Histochem Cytochem 29:577-80, 1981. Kyrkou K, Iatridis S, Athanassiadou P et al: Detection of benign or malignant origin of ascites with combined indirect immunoperoxidase assays of carcinoembryonic antigen and lysozyme. Acta Cytol 29:57-62, 1985. Kyrkou K, Iatridis S, Athanassiadou P, et al: The cytologic application of carcinoembryonic antigen for the discrimination of malignant from benign serous effusions. Cancer Det Prev 8:247-54, 1985. Lee JS, Nam JH, Lee MC et al: Immunocytochemical panel for distinguishing between carcinoma and reactive mesothelial cells in serous effusions. Acta Cytol 40:631-636, 1996. Mafisso AA, Carder PJ: Monoclonal antibodies in the cytodiagnosis of serous effusions. Cytopathology 1:119-28, 1990. Mezger J, Stotzer O, Schilli G et al: Identification of carcinoma cells in ascitic and pleural fluid: Comparison of four panepithelial antigens with carcinoembryonic antigen. Acta Cytol 36:75- 81, 1992. Naylor B. Pleural, peritoneal and pericardial fluids. In: Bibbo M, ed. Comprehensive Cytopathology. Philadelphia: Saunders Co; 1991, 541-614. Ordonez NG, Mackay B: The roles of immunohistochemistry and electron microscopy in distinguishing epithelial mesothelioma of the pleura from adenocarcinoma. Adv Anat Pathol 3:273-93, 1996. Ordonez NG: Value of the Ber-EP4 antibody in differentiating epithelial pleural mesothelioma from adenocarcinoma. The MD Anderson experience and a critical review of the literature. Am J Clin Pathol 109:85-89, 1998. Ruitenbeek T, Gouw A, Poppema S: Immunocytology of body cavity fluids. MOC-31, a monoclonal antibody discriminating between mesothelial and epithelial cells. Arch Pathol Lab Med 118:265-269, 1994. Sehested M, Ralfkieaer E, Rasmussen J: Immunoperoxidase demonstration of carcinoembryonic antigen in pleural and peritoneal effusions. Acta Cytol 27:124-27, 1983. Souhami RL, Beverley PCL, Bobrow LG, et al: Antigens of lung cancer results of the Second International Workshop on Lung Cancer Antigens. J Natl Cancer Inst 3:609-12, 1991. Wirth PR, Legier J, Wright GL: Immunohistochemical evaluation of seven monoclonal antibodies for differentiation of pleural mesothelioma from lung adenocarcinoma. Cancer 67:655-62, 1991. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 100 EP 103 PG 4 ER PT J AU Salehnia, M Farzad, RT Tachikhani, M Torghaban, ShSh Al-Traihi, T AF Salehnia, Mogdeh Farzad, R Tahereh Tachikhani, Mohamad Torghaban, Sh Shams Al-Traihi, Taki TI Alkaline Phosphatase Histochemistry and Biochemistry in the Diagnosis of Complete Hydatidiform Mole SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE alkaline phosphatase; placental alkaline phosphatase; complete hydatidiform mole; gestational trophoblastic disease ID alkaline phosphatase; placental alkaline phosphatase; complete hydatidiform mole; gestational trophoblastic disease AB The purpose of this study was a complementary method to the diagnosis and prognosis of complete hydatidiform mole (CHM) and differentiate it from the other cases of gestational trophoblatic diseases. This was done by examining the quality and quantity of the total and the placental alkaline phosphatase activity. The ALP in the tissues and sera from 12 patients were compared with 13 control normal non-pregnant and 30 control pregnant females. The enzyme activities were determined by biochemical and histochemical examination.The placental tissues were obtained from uterine curettage, or after delivery which then were frozen in a liquid nitrogen and processed for biochemical study. Cryosections were histochemically stained for ALP and PLAP by the azo coupling method. Isoenzyme specificity was evaluated by heating the tissue at 65°C for 15 min while the including L-phenylalanine (50 mM), D-phenylalanine (50 mM) and L-homoarginine (50 mM) were used for chemical inhibition study. The activity of ALP and PLAP of patients were reduced in comparison with pregnant control group (P<0.05). There was no significant difference between the patients and non-pregnant control (P<0.05) group. The localization of enzyme activities in cryosections of all groups were in the basal, apical, and the cytoplasm of syncytiotrophoblast cells. The ALP in all the groups was thermostable (65°C for 15 min) and was inhibited by L-phenylalanine, but no inhibition was seen with L – homoarginine in patients group only. These findings suggest that the PLAP is a useful marker in the diagnosis and prognosis of hydatidiform mole. C1 [Salehnia, Mogdeh] School of Medical Sciences, Tarbiat Modarres University, Department of HistologyTehran, Iran. [Farzad, R Tahereh] School of Medical Sciences, Tarbiat Modarres University, Department of HistologyTehran, Iran. [Tachikhani, Mohamad] School of Medical Sciences, Tarbiat Modarres University, Department of HistologyTehran, Iran. [Torghaban, Sh Shams] School of Medical Sciences, Tarbiat Modarres University, Department of HistologyTehran, Iran. [Al-Traihi, Taki] School of Medical Sciences, Tarbiat Modarres University, Department of HistologyTehran, Iran. RP Salehnia, M (reprint author), School of Medical Sciences, Tarbiat Modarres University, Department of Histology, Tehran, Iran. CR Beckstead JH: Alkaline phosphatase histochemistry in human germ cell neoplasms. AM J Surg Pathol 7:341-349, 1983. Bracken MB: Incidence and aetiology of hydatidiform mole: an epidemiological review. Br J Obstet Gynaecol 94:1123-1135, 1987. Buckley JD: The epidemiology of molar pregnancy and choriocarinoma. Clin Obstet Gynecol 27:153, 1984. Bur GE, Hertig AT, F.A.C.O.G, et al: Histochemical aspects of hydatidiform mole and choriocarcinoma. Obstet Gynecol 19:156-181, 1962. Byers DA, Fernley HN, Walker PG: Studies on alkaline phosphatase inhibition of human placental phosphoryl phosphatase by L – phenyl alanine. Eur J Biochem 29:197-204, 1972. Cadeau MJ, Malkin A: Relative heat stability for the identification of serum alkaline phosphatase isoenzymes. Clin Chim Acta 45:235-242, 1973. Chase JS, Check JH, Nowroozi KH, Wu CH: First trimester serum levels of the -subunit of human chorionic gonadotropin in a tubal molar pregnancy. Am J Obstet Gynecol 157:910, 1987. Cheah PL, Looi LM. Expression of placental proteins in complete and partial hydatidiform moles. Pathol 26: 115-118, 1994. Check JH., Nowroozi KH., Chase JS, et al. False – positive human chorionic gonadotropin levels caused by a heterophile antibody with the immunoradiometric assay. Am J Obstet Gynecol 158: 99-100, 1988. Danihel L, Porubsky J, Vojtassak J, et al: Trophoblastic disease I. Use of imunohistochemistry in the diagnosis of complete hydatidiform moles. Cesk patol 30:76-79, 1994. Danihel L, Porubsky J, Vojtassak J, et al: Trophoblastic disease II. Immunohistochemical and cytogenetic parameters of partial hydatidiform moles. Cesk Patol 30:80-84, 1994. Fukunaga M, Migazawa Y, Sugisbita M, et al: Immunohistochemistry of molar and non-molar placentas with special reference to their differenial diagnosis. Acta Pathol Jpn 43:683-689, 1993. Grimes DA: Epidemiology of gestational trophoblastic disease. Am J Obstet Gynecol 150:309-318, 1984. Ilancheran A, Singh P, Sen DK, et al: Up date on chemotherapy of gestational trophoblastic disease. Singapore J Obstet Gynecol 19:53-60, 1988. Kaldor G: Clinical enzymology New York: Prager publisher 87- 112, 1983. Koid O, Iwai S, Kanno T, et al: Isoenzymes of alkaline phosphatase in germinoma cells. Am J Clin Pathol 89:611-616, 1988. Lin CW, Fishman WH: L – homoarginine, an organospecific, uncompetitive inhibitor of human liver and bone alkaline phosphohydrolases. J Biol Chem 247:3082-3087, 1972. Losch A, Kainz C: Immunohistochemistry in the diagnosis of the gestational trophoblatic disease. Acta Obstet Gynecol Scand 75:753-756, 1996. Mckay DG, Hertig AT, Adams EC, et al: Histochemical observations on the germ cells of human embryos. Anat Rec 117: 201-219, 1953. Mc Laughlin PJ, Gee H, Johnson PM: Placental – type alkaline phosphatase in pregnancy and malignancy plasma: specific estimation using a monoclonal antibody in a solid phase enzyme immunoassay. Clinica Chimica Acta 130:199-209, 1983. Ming Chu T: Biochemical Markers for cancer. Clinical and biochemical analysis; 11, New York: Marcel Dekker, INC publisher 93-115, 1982. Ozturk M, Berkowitz R, Goldstein O, et al. Differential production of human chorionic gonadotropin and free subunits in gestational trophoblastic disease . Am J Obstet Gynecol 158:193- 198, 1988. Sabellek WM: Alkaline phosphatase, laboratory and clinical lmplications. J Chromato 429:419-444, 1988. Smith DB, O Reilly SM, Newland FS: Current approaches to diagnosis and treatment of gestational trophoblastic disease. Curr Opin Obstet Gynecol 5:84-91, 1993. Sonnenwright AC, Jarett L: Gradwohl’s Clinical laboratory methods and diagnosis. 8th ed London: C.V. Mosby Co. 256- 271, 1980. Wang TH, Wag HS: Gestational trophoblastic diseases: Current trends and perspectives. J Formos Med Assoc 94: 449-457, 1995 Yusoff Dawood M, Facog, Saxena BB, et al: Human chorionic gonadotropin and its subunits in hydatidiform mole and choriocarcinoma. Obstet Gynecol 50:172-180, 1977. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 105 EP 110 PG 6 ER PT J AU Szabo, J Hegedus, Gy Bartok, K Kerenyi, T Vegh, A Romics, I Szende, B AF Szabo, Janos Hegedus, Gyorgy Bartok, Katalin Kerenyi, Tibor Vegh, Attila Romics, Imre Szende, Bela TI Improving Diagnostic Accuracy of Prostate Carcinoma by Systematic Random Map-biopsy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE map-biopsy; prostate; carcinoma; RDE ID map-biopsy; prostate; carcinoma; RDE AB Systematic random rectal ultrasound directed map-biopsy of the prostate was performed in 77 RDE (rectal digital examination) positive and 25 RDE negative cases, if applicable. Hypoechoic areas were found in 30% of RDE positive and in 16% of RDE negative cases. The score for carcinoma in the hypoechoic areas was 6.5% in RDE positive and 0% in RDE negative cases, whereas systematic ''map” biopsy detected 62% carcinomas in RDE positive, and 16% carcinomas in RDE negative patients. The probability of positive diagnosis of prostate carcinoma increased in parallel with the number of biopsy samples/case. The importance of systematic map biopsy is emphasized. C1 [Szabo, Janos] Central Military Hospital, Department of UrologyBudapest, Hungary. [Hegedus, Gyorgy] Central Military Hospital, Department of UrologyBudapest, Hungary. [Bartok, Katalin] Military Hospital, Department of PathologyBudapest, Hungary. [Kerenyi, Tibor] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Vegh, Attila] Central Military Hospital, Department of UrologyBudapest, Hungary. [Romics, Imre] Semmelweis University, Department of UrologyBudapest, Hungary. [Szende, Bela] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Szende, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM bszende@korb1.sote.hu CR Resnick MI: Transrectal ultrasound guided versus digitally directed prostatic biopsy: A comparative study. J Urol 139:754- 757, 1988. Hodge KK, McNeal JE, Stamey TA: Ultrasound guided transrectal core biopsies of the palpably abnormal prostate. J Urol 142:66-70, 1989. Lippman HR, Ghiatas AA, Sarosdy MF: Systematic transrectal ultrasound guided prostate biopsy after negativ digitally directed prostate biopsy. J Urol 147:827-829, 1992. Terris MK, McNeal JE, Stamey TA: Detection of clinically significant prostate cancer by transrectal ultrasound guided systematic biopsies. J Urol 148:829-832, 1992. Irwin MB, Trapasso JG, Stamey TA: Identification of insignificant prostate cancers. Analysis of preoperative parameters. Urology 44:862-868, 1994. Devonec M, Fendler JP, Monsallier M, et al: The significance of the prostatic hypoechoic area: results in 226 ultrasonically guided prostatic biopsies. J Urol 143:316-319, 1990. Hodge KK, McNeal JE, Terris MK, et al: Random systematic versus directed ultrasound guided transrectal core biopsies of the prostate. J Urol 142:71-75, 1989. Hammerer P, Huland H, Cooner WH: Systematic sextant biopsies in 651 patients reffered for prostate evaluation. J Urol 151:99-104, 1994. Vallancien G, Prapotnich D, Veillon B, et al: Systematic prostatic biopsies in 100 men with no suspicion of cancer on digital rectal examination. J Urol 146:1308-1312, 1991. Gasman D, Abbou CC: Biopsies prostatiques: technique, interets et complitatious. Entretiens de Bichat Urologie. Abstract. 1995. Jewett HJ: Significance of the palpable prostatique nodule. JAMA 160:838-839, 1956. Lee F, Trop-Pedersen S, Littrup PJ, et al: Hypoechoic lesions of the prostate: clinical relevance of tumor size, digital rectal examination, and prostate specific antigen. Radiology 170:29- 32, 1989. Holm HH, Gammelgaard J: Ultrasound guided precise needle placement in the prostate and seminal vesicles. J Urol 125:385- 387, 1981. Cooner WH, Mosley BR, Rutherford Jr CL, et al: Prostate cancer detection in a clinical urological practice by ultrasonography, digital rectal examination and prostate specific antigen. J Urol 143:1146-1154, 1990. Topr-Pedersen S, Lee F, Littrup PJ, et al: Transrectal biopsy of the prostage guided with transrectal US: Longitudinal and multiplanar scanning. Radiology 170:23-27, 1989. Coplen DE, Andriole GL, Yuan JJJ, et al: The ablity of systematic transrectal ultrasound guided biopsy to detect prostate cancer in men with the clinical diagnosis of benign prostatic hyperplasia. J Urol 146:75-77, 1991. Vashi AR, Wojno KJ, Henricks W, et al: Determination of the reflex range and appropriate cutpoints for percent free PSA prostate specific antigen in 413 men referred for prostatic evaluation using the AxSYM system. Urology 49:19-27, 1997. William J, Catalona, et al: Evaluation of percentage of free serum prostatic specific antigen to improve specificity of prostate cancer screening. JAMA 274:1214-1220, 1995. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 111 EP 113 PG 3 ER PT J AU Perambakam, S Naresh, K Nerurkar, A Nadkarni, J AF Perambakam, Supriya Naresh, N. Kikkeri Nerurkar, Ashutosh Nadkarni, Jayshree TI Intra-tumoral Cytolytic Cells: Pattern of Distribution in B-cell Non Hodgkin’s Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lymphoma; T-cells; CD4; CD8; TIA-1; NK cells; tumor infiltrating lymphocytes; cytotoxic cells; B-cell ID lymphoma; T-cells; CD4; CD8; TIA-1; NK cells; tumor infiltrating lymphocytes; cytotoxic cells; B-cell AB Non-Hodgkin’s lymphomas (NHLs) constitute a heterogeneous group of lymphoid neoplasms and a majority of them in India are of B-cell phenotype. Varying numbers of T lymphocytes and natural killer (NK) cells are consistently present within the lymph nodes (LNs). The role of these ''reactive'' cells is becoming understood. TIA-1 is a cytotoxic granule associated RNA binding protein, the expression of which is restricted to cytotoxic T lymphocytes (CTLs) and NK cells. Snap frozen lymph node biopsies obtained from 41 B-cell NHLs were localized for intra-tumoral TIA-1 + cytolytic cells by immunohistochemistry. Distribution of T cell subsets and NK cells were also quantified. Cells expressing TIA-1 antigen was observed in all the cases, seen as a strong granular cytoplasmic signal. Results indicate significantly higher number of TIA-1 cytolytic cells outside (periphery of the follicle and interfollicular areas) than within the neoplastic follicle in follicular lymphomas (p<0.001). In small lymphocytic lymphomas, cytolytic cells were mainly seen as uniformly scattered single cells, distributed throughout the tumor environment. In mantle cell and diffuse large B-cell lymphomas these were most often seen as small clusters and less frequently as singly scattered cells. Higher numbers of CD4 + than the CD8 + T cells were observed in most cases. Contrary to the follicles in follicular hyperplasia, CD57 + NK cells were predominantly observed outside the neoplastic follicle in follicular lymphomas (FLs). These results outline specific interactions between the potential anti-tumoral cytolytic and the malignant cells of B-cell NHLs. C1 [Perambakam, Supriya] Tata Memorial Centre, Cancer Research Institute, 400 012 Parel, Mumbai, India. [Naresh, N. Kikkeri] Tata Memorial HospitalParel, Mumbai, India. [Nerurkar, Ashutosh] Tata Memorial HospitalParel, Mumbai, India. [Nadkarni, Jayshree] Tata Memorial Centre, Cancer Research Institute, 400 012 Parel, Mumbai, India. RP Nadkarni, J (reprint author), Tata Memorial Centre, Cancer Research Institute, 400 012 Parel, India. EM cri3@soochak.ncst.ernet.in CR Anderson P, Nagler-Anderson C, O’Brien C, et al: A monoclonal antibody reactive with a 15-kDa cytoplasmic granuleassociated protein defines a subpopulation of CD8+ T lymphocytes. J Immunol 144:574-582, 1990. Diaz JI, Edinger MG, Stoler MH, et al: Activated T cell subsets in lymphoid hyperplasias and B-cell Non-Hodgkin’s Lymphomas. Am J Pathol 139:503-509, 1991. Diaz J, Tubbs R, Stoller M, et al: Cytolytic, TIA-1+, tumor infiltrating lymphocytes in B Cell Non-Hodgkin’s Lymphomas: SWOG central repository members, Leukemia Lymphoma 9:91-94, 1993. Dvoretsky P, Wood GS, Levy R, et al:T lymphocyte subsets in follicular lymphomas compared with those in non-neoplastic lymph nodes and tonsils. Hum Pathol 13:618-625, 1982. Felgar RE, Steward KR, Cousar JB, et al: T-cell rich large Bcell lymphomas contain non activated CD8 + T cells, show increased tumor cell apoptosis, and have lower Bcl-2 expression than diffuse B-cell lymphomas. Am J Pathol 153:1707- 1715, 1998. Garda CF, Lowder J, Meeker TC, et al: Differences in host infiltrates among lymphoma patients treated with anti-idiotype antibodies: Correlation with treatment response. J Immunol 135:4252-4260, 1985. Harcis NL, Bhan A: Distribution of T cell subsets in follicular and diffuse lymphomas of B cell type, Am J Pathol 113:172- 180,1983. Harris NL, Jaffe ES, Stein H, et al: A revised European American Classification of lymphoid neoplasms: A proposal from the International lymphoma study group. Blood 84:1361-1392, 1994. Leger-Ravet M-8, Devergne O, Peuchmaur M, et al: In situ detection of activated cytotoxic ceils in follicular lymphomas, Am J Pathol 144:492-499, 1994. Lindemalm C, Mellstedt H, Biberfeld P, et al: Blood and lymph node T-lymphocyte subsets in Non-Hodgkin’s lymphomas. Scand J Hematol 30:68-78, 1983. Medeiros LJ, Picker LJ, Gelb AB, et al: Numbers of host helper T cells and proliferating cefls predict survival in diffuse small cefl lymphomas. J Clin Oncol 7:1009-1017, 1989. Perambakam S, Amin K, Naresi K, et al: Auto-tumor reactive cytotoxic T cell responses in B-cell Non-Hodgkin’s Lymphoma. Leukemia Lymphoma 27:145-152, 1997. Perambakam S, Naresh K, Soman C,et al: Non-Hodgkin’s lymphoma: Cytotoxic T lymphocyte mediated activity correlated with histopathologic classification and staging. Cancer Biother and Radiopharmaceuticals 12:13-18, 1997. Tian Q, Streuli M, Saito H, et al: A polyadenylate binding protein localized to the granules of cytolytic lymphocytes induces DNA fragmentation in target cells. Cell 67: 629-639, 1991. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 114 EP 117 PG 4 ER PT J AU Oz, B Karayel, AF Gazio, LN Ozlen, F Balci, K AF Oz, Buge Karayel, Anik Ferah Gazio, LU Nurperi Ozlen, Fatma Balci, Kerem TI The Distribution of Extracellular Matrix Proteins and CD44S Expression in Human Astrocytomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE astrocytic tumors; extracellular matrix proteins; CD44; tenascin; immunohistochemistry; anaplastic astrocytoma; glioblastoma ID astrocytic tumors; extracellular matrix proteins; CD44; tenascin; immunohistochemistry; anaplastic astrocytoma; glioblastoma AB Aims of the study were: 1. to establish the prevalence of CD44 protein expression in human astrocytomas; 2. to compare the distribution of the extracellular matrix in these tumors; 3. to investigate the relation between CD 44, the extracellular matrix proteins and the histological grade of the tumor. CD44, Type IV Collagen (Col IV), Laminin (LN), Fibronectin (FN), and Tenascin (TN) expression were detected by immunohistochemistry in formalin fixed paraffin embedded tissue samples of 52 astrocytic tumors: 35 glioblastomas (GB), 7 Anaplastic astrocytomas (AA) and 10 astrocytomas (A). The localization of Col IV was observed in the basement membrane of the vessel walls in most of the astrocytomas (88.4%) with a similar pattern obtained with LN staining. 7 of 10 A (70%), 2 of 7 AA (28%) and 9 of 35 GB (25.7%) showed LN positivity. There was a negative correlation between LN expression and tumor grade (p=0.03). FN was either localized in the basement membrane or showed thick multi-layered immunoreactivity of the vessel walls. FN expression was seen in 6 A (60%), 4 AA (57%) and all of 35 GB (100%). The FN distribution was not uniform and its staining intensity showed decrease in GB. 3A (30%), 3 AA (42%), 27 GB (77.1%) showed TN expression in the vessel walls and in some tumor cells of 19 GBs. TN expression was positively correlated with the degree of vascular endothelial proliferation in GB (p<0.05). The expression of CD44s wasseen as plasma membrane positivity of glioma cells in 5 of 10A (50%), 3 of 7AA (42.3%) and 29 of 35 GB (82.8%). The intensity of immunoreaction was quite strong especially near the vessels. There was a good correlation between TN and CD44s expression in human astrocytic tumors (p=0.005). No relationship was observed between GFAP, ECM proteins and CD44s expression. Both CD44s and TN expression showed increase with malignancy in astrocytomas. These findings indicated that the histological malignancy of the astrocytomas was correlated with expression of TN and CD44s. It was suggested that in astrocytomas there was a biological relationship only between CD44 and TN, but none with the other ECM proteins. TN may play a role in angiogenesis in human astrocytic tumors. C1 [Oz, Buge] Istanbul Medical Faculty, Department of Pathology, Aksaray, 34304 Istanbul, Turkey. [Karayel, Anik Ferah] Istanbul Medical Faculty, Department of Pathology, Aksaray, 34304 Istanbul, Turkey. [Gazio, LU Nurperi] Istanbul University, Cerrahpaºa Medical Faculty, Department of NeurosurgeryIstanbul, Turkey. [Ozlen, Fatma] Istanbul University, Cerrahpaºa Medical Faculty, Department of NeurosurgeryIstanbul, Turkey. [Balci, Kerem] Istanbul Medical Faculty, Department of Pathology, Aksaray, 34304 Istanbul, Turkey. RP Oz, B (reprint author), Istanbul Medical Faculty, Department of Pathology, 34304 Istanbul, Turkey. EM ferhanoz@tkbbv.org.tr CR Ariza A, Lopez D, Mate JL et al: Role of CD44 in Invasiveness of Glioblastoma Multiforme and the Noninvasivness of Meningioma. Hum Pathol 26:1144-1147, 1995. Attanoos RL, Webb R, Gibss AR: CD44H Expression in Reactive Mesothelium, Pleural Mesothelioma and Pulmonary Adenocarcinoma. Histopathol 30:260-263, 1997. Bellon G, Caulet T, Cam T, et al: Immunohistochemical Localization of Macromolecules of the Basement Membrane and Extracellular Matrix of Human Gliomas and Meningiomas. Acta Neuropathol, Berl, 66:245-252, 1985. Brodkey JA, Laywell FD, O’Brien TF, et al: Focal Brain Injury and Upregulation of a Developmentally Regulated Extracellular Matrix Protein. J Neurosurg 82:106-112, 1995. Broll R, Meyer S, Neubner M, et al: Expression of Tenascin in Tumors of the Esophagus, Small Intestine and Colorectum. Gen Diagn Pathol 141:111-119, 1995. Burger PC, Scheithauer BW: Tumors of Neuroglia and Choroid Plexus Epithelium. Ed. Tumor of the Central Nervous System. Atlas of Tumor Pathology. Armed Forces Institute of Pathology. Washington D.C. Bethesta, Maryland,1994 p: 25-68. Chiquet-Ehrismann R, Kalla P, Pearson CA, et al: Tenascin Interferes with Fibronectin Action. Cell 53:383-390, 1988. Chiquet-Ehrismann R, Mackie EJ, Pearson CA, et al: Tenascin: An Extracellular Matrix Protein Involved in Tissue Interactions during Fetal Development and Oncogenesis. Cell 47:131-139, 1986. Darai E, Walker-Combrouze F, Fauconnier A, et al: Analysis of CD44 Expression in Serous and Mucinous Borderline Tumors of the Ovary: Comparision with Cystadenomas and Overt Carcinomas. Histopathol 32:151-159, 1998. Faissner A, Scholze A, Gotz B:Tenascin Glycoproteins in Developing Neural Tissues: Only Decoration? Perspect Dev Neurobiol 2:53-66, 1994. Fasana M, Sabatini MT, Wieczorek R, et al: CD44 and Its v6 Spliced Variant in Lung Tumors. Cancer 80:34-41, 1997. Fox SB, Fawcett J, Jackson DG, et al: Normal Human Tissues, in Addition to some Tumors, Express Multiple Different CD44 Isoforms. Cancer Res 54:4539-4546, 1994. Fox SB, Gatter KC, Jackson DG, et al: CD44 and Cancer Screening, letters to the Editor, Lancet 342:548-549, 1993. Girgrah N, Letarte M, Becker LE, et al: Localization of the Cd44 Glycoprotein to Fibrous Astrocytes in Normal White Matter and to Reactive Astrocytes in Active Lesions in Multiple Sclerosis. J Neuropathol Exp Neurol 50:779-792, 1991. Glese A, Rief MD, Loo MA, et al: Determination of Human Astrocytoma Migration. Cancer Res 54:3897-3904, 1994. Hankard GF, Cezard JP, AigramY, et al:CD44 Variam Expression in Inflammatory Colonic Mucosa is not Disease Specific but Associated with Increased with Crypt Cell Proliferation. Histopathol 32:317-321, 1998. Higuchi M, Ohnishi N, Arita S, et al: Expression of Tenascin in Human Gliomas: Its relation to Histologic Malignancy, Tumor Dedifferentiation and Angiogenesis. Acta Neuropathol 85:481- 487, 1993. Howeedy AA, Virtanen I, Laitinen L, et al: DifFerential Distribution of Tenascin in Normal, Hyperplastic and Neoplastic Breast. Lab Invest 63:798-806, 1990. Iczkowski KA, Shanks JH, Bostwick DG: Loss of CD44 Variant 6 Expression Differentiates Small Cell Carcinoma of Urinary Bladder from Urothelial, transitional cell, Carcinomas. Histopathol 32:322-327, 1998. Ingle J, Jennings TA, Goodman ML, et al: CD44 Expression in Sinonasal Inverted Papillomas and Associated Squamous Cell Carcinoma. Am J Clin Pathol 109:309-314, 1998. Khoshyomn S, Penar PL, Wadsworth MP, et al: Localization of Cd44 at the Invasive Margin of Glioblastomas by Immunelectron Microscopy. Ultrastruct Pathol 21:517-425, 1997. Kleihues P, Burger PC, Scheithauer BW: Histological typing of Tumors of the Central Nervous System.World Health Organisation. Berlin, Springer-Verlag, 1993. Koukoulis GK, Gould VE, Bhattacharyya A, et al: Tenascin in Normal, Reactive, Hyperplastic and Neoplastic Tissues. Hum Pathol 22:636-643, 1997. Kupper MC, Van-Meir E, Gaultier T, et al: Differential Expression of the CD44 Molecule in Human Brain Tumors. Int J Cancer 50:572-577, 1992. Lagorge-Pages C, Paraf F, Dubuis S, et al: Expression of CD44 In Premalignant and Malignant Barret’s Oesephagus. Histopathol 37:7-14, 1998. Lesley J, Hyman R, Kincade PW: Cd44 and its Interaction with Extracellular Matrix. Adv Immunol 54:271-335, 1993. Li H, Hamou MF, Tribolet N, et al: Variant CD44 Adhesion Molecules are Expressed in Human Brain Metastases but not in Glioblastomas. Cancer Res 53:5345-5349, 1993. Mackie EJ, Halfter W, Liverani D: Induction of Tenascin in Healing Wounds. J Cell Biol 107:2757-2767, 1988. Maenpa A, Kovanen PE, Paetau A, et al: Lymphocyte Adhesion Molecule Ligands and Extracellular Matrix Proteins in Gliomas and Normal Brain: Expression of VCAM-1 in Gliomas. Acta Neuropathol, Berl, 94:216-225, 1997. Mahesparan R, Tysnes BB, Edvardsen K, et al: Role of High Molecular Weight Extracellular Matrix Proteins in Glioma cell Migration. Neuropath Appl Neurobiol 23:102-112, 1997. Merzak A, Koochekpour S, Pilkington GJ: Adhesion of Glioma Cells to Fibronectin, Laminin, Vitronectin and Collagen I is Modulated by Gangliosides in vitro. Cell Adhes Commun 3:27- 43, 1995. Natali PG, Zardi L: Tenascin: a Hexameric Adhesive Glycoprotein. Int J Cancer Suppl 4:66-68, 1989. Penno MB, August JT, Baylin SB, et al: Expression of CD44 in Human Lung Tumors. Cancer Res 54:1381-1387, 1994. Russell DS, Rubinstein LJ: Tumors of Central Neuro-epithelial Origin: In pathology of Tumours of the Nervous System.5. Ed. Edward Arnould, London, 1989 pp:83-350. Ventimiglia JB, Wikstrand CJ, Ostrowski LE, et al: Tenascin Expression in Human Glioma Cell Lines and Normal Tissue. J Neuroimmunol 36:41-55, 1992. Yamamoto H, Ohnisi T, Arita N, et al: Migration and Adhesivnes of Malignant Glioma Cells to Fibronectin or Vitronectin and Their Expression of Integrin Subunits. Nippon Rinsho 53:1683- 1687, 1995. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 118 EP 124 PG 7 ER PT J AU Suda, K Nobukawa, B Yamasaki, Sh Suzuki, F Shimizu, H Takase, M AF Suda, Koichi Nobukawa, Bunsei Yamasaki, Shigetaka Suzuki, Fujihiko Shimizu, Hideo Takase, Masaru TI Pre-existing Histological Type and Developmental Mechanism of Mucinous Noncystic Carcinoma of Pancreas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE pancreas cc; intraductal papillary-mucinous cc; ductal adenocc; mucin overproduction; mucinous degeneration; MUC immunoreactivity ID pancreas cc; intraductal papillary-mucinous cc; ductal adenocc; mucin overproduction; mucinous degeneration; MUC immunoreactivity AB Eleven cases with mucinous noncystic carcinoma (MC) of the pancreas were studied by histology and mucin immunohistochemistry, to elucidate the mechanism, or route of development, and pre-existing histological type of MC of the pancreas. These MCs were observed in close approximation to, or surrounding, intraductal papillary-mucinous carcinomas (IPMCs), and were centrally situated among ductal adenocarcinomas (DAs). Hence, the 11 cases originated from 8 IPMCs and 3 DAs. The mechanism and routes to MC were divided into four types as follows: IPMC directly invaded the stroma (4 cases), over-production of mucin in IPMC expanded the branches of the pancreatic duct possibly resulting in rupture (3 cases), DA underwent extreme mucinous degeneration (3 cases), and a recurrent form, as MC, at the surgical stump of IPMC (one case). The outcomes of MC cases with IPMC had variable survival rates, while those from DA had short durations. MUC immunoreactivity in MC was divided into three categories; anti-MUC1-positive only (2 IPMCs, 2 DAs), mixed anti-MUC1 and anti-MUC2-positive (3 IPMCs, one DA) and anti-MUC-positive only (3 IPMCs). Pre-existing MC histological types included both IPMC and DA. These two pre-existing types of MC involved mucin overproduction and mucinous degeneration. MUC immunoreactivity in MC revealed three patterns, which may be related to variable outcomes. C1 [Suda, Koichi] Juntendo University School of Medicine, Department of Pathology, 2-1-1 Hongo, Bunkyo-ku, 113-8421 Tokyo, Japan. [Nobukawa, Bunsei] Juntendo University School of Medicine, Department of Pathology, 2-1-1 Hongo, Bunkyo-ku, 113-8421 Tokyo, Japan. [Yamasaki, Shigetaka] Juntendo University School of Medicine, Department of Pathology, 2-1-1 Hongo, Bunkyo-ku, 113-8421 Tokyo, Japan. [Suzuki, Fujihiko] Juntendo University School of Medicine, Department of Pathology, 2-1-1 Hongo, Bunkyo-ku, 113-8421 Tokyo, Japan. [Shimizu, Hideo] Juntendo University School of Medicine, Department of Pathology, 2-1-1 Hongo, Bunkyo-ku, 113-8421 Tokyo, Japan. [Takase, Masaru] Yamanashi Prefectural Central Hospital, Clinical Laboratory, Pathology DivisionYamanashi, Japan. RP Suda, K (reprint author), Juntendo University School of Medicine, Department of Pathology, 113-8421 Tokyo, Japan. EM ksuda@med.juntendo.ac.jp CR Kloppel G: Pancreatic and non-endocrine tumors. In Pancreatic pathology., Eds . Kloppel G and Heitz PU), Churchill Livingstone, Edinburgh, 1984, pp. 79-113. Solcia E, Capella C, Kloppel G: Tumor of the pancreas. Atlas of tumor pathology, 3rd series, Fascicle 20. Armed Forces Institute of Pathology, Washington DC, 1997, pp. 88. Hasegawa H, Takada T, Uchiyama K: Mucin-producing pancreatic cancer. Int J Pancreatol 3, Suppl 2): 249-250, 1988. Ohhashi K, Murakami Y, Maruyama M, et al: Four cases of mucous secreting pancreatic cancer, in Japanses English abstract). Prog Dig Endosc 20:348-351, 1982. Yamada M, Kozuka S, Yamao K, et al: Mucin-producing tumor of the pancreas. Cancer 68:159-168, 1991. Sessa F, Solcia E, Capella C, et al: Intraductal papillary-mucinous tumors represent a distinct group of pancreatic neoplasms: an investigation of tumor cell differentiation and K-ras, p53 and c-erbB-2 abnormalities in 26 patients. Virchows Archiv 425:357-367, 1994. Swallow DM, Gendler S, Griffiths B, et al: The human tumorassociated epithelial mucin are codes by an expressed hypervariable gene locus PUM. Nature 328:82-84, 1987. Gum JR, Byrd JC, Hicks JW, et al: Molecular cloning of human intestinal mucin cDNAs. Sequence analysis and evidence for genetic polymorphism. J Biol Chem 246:6480-6487, 1989. Gum JR, Hicks JW, Swallow DM, et al: Molecular cloning of cDNAs derived from a novel human intestinal mucin gene. Biochem Biophys Res Commun 171:407-415, 1990. Osako M, Yonezawa S, Siddiki B, et al: Immunohistochemical study of mucin carbohydrates and core proteins in human pancreatic tumors. Cancer 71:2191-2199, 1993. Abe M, Kufe DW: Identification of a family of high molecular weight tumor-associated glycoprotein. J Immunol 139:257- 261, 1987. Xing PX, Prenzoska J, Layton GT, et al: Second generation monoclonal antibodies to intestinal MUC2 peptide reactive with colon cancer. J Natl Cancer Inst 84:699-703, 1992. Fukushima N, Mukai K, Kanai Y, et al: Intraductal papillary tumors and mucinous cystic tumors of the pancreas: Clinicopathologic study of 38 cases. Hum Pathol 28:1010-1017, 1997. Suda K, Hirai S, Matsumoto Y, et al: Variant of intraductal carcinoma, with scant mucin production, is of main pancreatic duct origin: A clinicopathological study of four patients. Am J Gastroenterol 91:798-800, 1996. Muir EG: ’Colloid’ carcinoma of the pancreas. Br J Surg 40:177, 1952. Ho SB, Niehans GA, Lyftogt C, et al: Heterogeneity of mucin gene expression in normal and neoplastic tissues. Cancer Res 53:641-651, 1993. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 125 EP 129 PG 5 ER PT J AU Ehrmann, J Galuszkova, D Ehrmann, J Krc, I Jezdinska, V Vojtesek, B Murray, GP Kolao, Z AF Ehrmann, Jioi Galuszkova, Dana Ehrmann, Jioi Krc, Ivo Jezdinska, Vera Vojtesek, Booivoj Murray, G Paul Kolao, Zdenek TI Apoptosis-related Proteins, BCL-2, BAX, FAS, FAS-L and PCNA in Liver Biopsies of Patients with Chronic Hepatitis B Virus Infection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apoptosis; hepatitis B virus; FAS; FAS-L ID apoptosis; hepatitis B virus; FAS; FAS-L AB While the elimination of hepatitis B virus (HBV) is a common phenomenon at the end of the acute phase of disease, the persistence of HBV is characteristic for chronic hepatitis (CHB). Recent evidence indicates that the elimination of HBV is achieved by FAS/FAS-L induced apoptosis of infected hepatocytes. The aim of this study was to test the hypothesis that HBV persistence in the hepatocytes of CHB patients is due to the delayed onset of apoptosis caused by altered FAS/FAS-L interactions between lymphocytes and hepatocytes. The expression of FAS, FAS-L, BAX, BCL-2, ICE and PCNA in the liver biopsies of 55 patients (14 HBsAg positive, 20 patients with alcoholic hepatopathy, 21 patients with other hepatopathies) was tested by immunohistochemistry. Apoptosis of hepatocytes was evaluated by morphological as well as by TUNEL method. The results were correlated with a grading/staging score and analysed statistically using a one way analysis of variance and the Duncan test. Significantly highernumbers of BAX positive hepatocytes were observed in HBsAg positive patients when compared to control groups. Similarly, both BAX and FAS positive lymphocytes were more frequent in HBsAg positive patients. FAS-L positive lymphocytes and hepatocytes were numerous in all patient groups. Increased numbers of BAX positive hepatocytes in CHB may reflect the increased readiness of these cells to undergo apoptosis. However, the increased numbers of both BAX and FAS positive lymphocytes in CHB suggest that these cells may be particularly sensitive to FAS-L mediated apoptosis potentially resulting in lowered viability of these lymphocytes. This may explain, at least in part, the defective removal of virus-infected cells in chronic hepatitis. However, we cannot rule out the possibility that survival of hepatocytes during CHB may be due to other mechanisms such as defects in apoptosis activation triggered by CD40, defects involving DNase and/or other caspases downstream in the apoptotic cascade within these cells, or to defects in CTL function. C1 [Ehrmann, Jioi] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular Pathology, Hnivotinska 3, CZ-77515 Olomouc, Czech Republic. [Galuszkova, Dana] Faculty of Medicine, PalackyUniversity, Second Department of Internal MedicineOlomouc, Czech Republic. [Ehrmann, Jioi] Faculty of Medicine, PalackyUniversity, Second Department of Internal MedicineOlomouc, Czech Republic. [Krc, Ivo] Faculty of Medicine, PalackyUniversity, Second Department of Internal MedicineOlomouc, Czech Republic. [Jezdinska, Vera] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular Pathology, Hnivotinska 3, CZ-77515 Olomouc, Czech Republic. [Vojtesek, Booivoj] Masaryk Memorial HospitalBrno, Czech Republic. [Murray, G Paul] School of Health Sciences, University of Wolverhampton, Division of Biomedical SciencesWolverhampton, UK. [Kolao, Zdenek] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular Pathology, Hnivotinska 3, CZ-77515 Olomouc, Czech Republic. RP Ehrmann, J (reprint author), Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular Pathology, CZ-77515 Olomouc, Czech Republic. EM erman@tunw.upol.cz CR Adams JM, Cory S: The Bcl –2 protein family: arbiters of cell survival. Science 281:1322-26, 1998. Afford SC, Randhawa S, Eliopoulos AG, et al: CD40 activation induces apoptosis in cultured human hepatocytes via induction of cell surface fas ligand expression and amplifies fas-mediated hepatocyte death during allograft rejection. J Exp Med 189:441-6, 1999. Ando K, Guidotti LG, Wirth S, et al: Class I-restricted cytotoxic T lymphocytes are directly cytopathic for their target cells in vivo. J Immunol. 152:3245-3253, 1994. Bellgrau D, Gold D, Selawry H, et al: A role for CD95 ligand in preventing graft rejection. Nature 377: 630-632, 1995 Berke G: The CTL’s kiss of death Cell 81:9-12, 1995. Beutler B, van Huffel C: Unraveling function in the TNF ligand and receptor families. Science 264:667-668, 1994. Dayan CM: FasL expression on epithelial cells: the Botazzo- Feldmann hypothesis revisited. Immunology Today 18:203, 1997. Dervan PA, Magee HM, Buckley C, et al: Proliferating cell nuclear antigen counts in formalin-fixed paraffin-embedded tissue correlate with Ki-67 in fresh tissue. Am J Clin Pathol 97, Suppl. 1): S21-S28, 1992. Ehrmann J jr, Jezdinska V, Ehrmann J, et al: Immunohistochemical study of the expression of BCL-2, PCNA and p53 proteins in the patients with hepatitis B. The pilot study. Acta Univ Palacki Olomuc, Fac Med 10:83-85, 1996. Enari M, Hug H, Nagata S: Involvement of an ICE-like protease in FAS-mediated apoptosis. Nature 375:78-81, 1995. Ferrari C, Penna A, Bertoletti A, et al: Hepatitis B virus-specific immune response: from antigen recognition to liver damage. Forum 6:203-216, 1996. French LE, Tschopp J: Thyroiditis and hepatitis: FAS on the road to disease. Nature Medicine 3:387-388, 1997. Galle PR, Hofmann WJ, Walczak H, et al: Involvement of the CD 95, APO-1/Fas, receptor and ligand in liver damage. J Exp Med 182:1223-30, 1995. Golstein P: Controlling cell death. Science 275:1081-1082, 1997. Griffith TS, Brunner T, Fletcher SM, et al: Fas ligand-induced apoptosis as a mechamism of immune privilege. Science 270:1189-1192, 1995. Guidotti LG, Borrow P, Hobbs MV, et al: Viral cross talk: intracellular inactivation of the hepatitis B virus during an unrelated viral infection of the liver. Immunology 93:4589-4594, 1996. Guidotti LG, Chisari FV: The transgenic mouse model of hepatitis B virus infection. Forum 6:189-200, 1996. Guidotti LG, Ishikawa T, Hobbs MV, et al: Intracellular inactivation of the hepatitis B virus by cytotoxic T lymphocytes. Immunity 4:25-26, 1996. Guilhot S, Miller T, Cornman G, et al: Apoptosis induced by tumor necrosis factor in rat hepatocyte cell lines expressing hepatitis B virus. Am J Pathol 148:801-814, 1996. Hahne M, Rimoldi D, Schroter M, et al: Melanoma cell expression of FAS, Apo-1/CD95, ligand : implications for tumor immune escape. Science 274:1363-1366, 1996. Hiramatsu N, Hayashi A, Katayama K, et al: Immunohistochemical detection of Fas antigen in liver tissue of patients with chronic hepatitis C. Hepatology 19:1354-1359, 1994. Chisari F: Hepatitis B virus transgenic mice: insights into the virus and disease. Hepatology 22:1316-1325, 1995. Ishak K, Baptista A, Bianchi L, et al: Histological grading and staging of chronic hepatitis. J Hepatol 22:696-699, 1995. Kondo T, Suda T, Fukuyama H, et al: Essential roles of the FAS ligand in the development of hepatitis. Nature Medicine 3:409- 413, 1997. Koziel MJ: Immunology of viral hepatitis. Am J Med 100:98- 109, 1996. Kroemer G: The proto-oncogene Bcl-2 and its role in regulating apoptosis. Nature Medicine 3:614-620, 1997. Lacronique V, Mignon A, Fabre A, et al: Bcl-2 protects from lethal hepatic apoptosis induced by an anti-Fas antibody in mice. Nature Medicine 2:80-86, 1996. Liu CC, Walsh CM, Young JDE: Perforin: structure and function. Immunol Today 16:194-201, 1995. Luo KX, Zhu YF, Zhang LX, et al: In situ investigation of Fas/FasL expression in chronic hepatitis B infection and related liver diseases. J Viral Hepat 4:303-7, 1997. Los M, Van de Craen M, Penning LC, et al: Requirement of an ICE/CED-3 protease for FAS/APO-1-mediated apoptosis. Nature 375:81-83, 1995. Marinos G, Torre F, Chokshi S, et al: Induction of T-helper cell response to hepatitis B core antigen in chronic hepatitis B: a major factor in activation of the host immune response to the hepatitis B virus. Hepatology 22:1040-1049, 1995. Mochizuki K, Hayashi N, Hiramatsu N, et al: Fas antigen expression in liver tissues of patients with chronic hepatitis B. J Hepatology 24:1-7, 1996. Muschen M, Warskulat U, Douillard P, et al: Regulation of CD95, APO-1/Fas, receptor and ligand expression by lipopolysaccharide and dexamethasone in parenchymal and nonparenchymal rat liver cells. Hepatology 27:200-208, 1998. Nagata S, Golstein P: The FAS death factor. Science 267:1449- 1456, 1995. Nakamoto Y, Guidotti LG, Pasquetto V, et al: Differential target cell sensitivity to CTL-activated death pathways in hepatitis B virus transgenic mice. J Immunol 158:5692-7, 1997. Paroli M, Nisini R, Accapezzato D, et al: Viral escape from the immune system`s control. Forum 6:122-127, 1996. Patel T, Gores GJ, Kaufmann SH: The role of proteases during apoptosis. FASEB J 10:587-597, 1996. Peters M: Actions of cytokines on the immune response and viral interactions: an overview. Hepatology 23:909-916, 1996. Reed JC: Double identity for proteins of the Bcl-2 family. Nature 387:773-776, 1997. Rehermann B, Lau D, Hoofnagle JH, et al: Cytotoxic T lymphocyte responsiveness after resolution of chronic hepatitis B virus infection. J Clin Invest 7:1655-1665, 1996. Romero R, Lavine JE: Cytokine inhibition of the hepatitis B virus core promoter. Hepatology 23:17-23, 1996. Ryo K, Kamogawa Y, Ikeda I, et al: FAS and FAS ligand strongly expressed in liver of fulminant hepatitis patients. Hepatology 22: AASLD Abstracts: 230 A, 1995. Shibata S, Kyuwa S, Lee S, Toyota Y, et al: Apoptosis induced in mouse hepatitis virus infected cells by a virus specific CD8+ cytotoxic lymphocyte clone. J Virol 68:7540-7545, 1994. Schaff Z, Lotz G, Schulte-Herman R: Pathomorphological characteristic and pathogenesis of viral hepatitis. Pathology Oncology Research 2:132-143, 1996. Smyth MJ, Trapani JA: Granzymes: exogenous proteinases that induce target cell apoptosis. Immunology Today 16:202-206, 1995. Strand S, Hofmann WJ, Hug H, et al: Lymphocyte apoptosis induced by CD95, Apo-1/Fas, ligand-expressing tumor cells – a mechanism of immune evasion? Nature Medicine 2:1361- 1366, 1996. Streilein JW: Unraveling immune privilege. Science 270:1158- 1159, 1995. Tang DG, Porter AT: Apoptosis: a current molecular analysis. Pathology Oncology Research 2:117-131, 1996. Vaux DL: Ways around rejection. Nature 377:576-577, 1995. Williams N: Tumor cells fight back to beat immune system. Science 274:1362-1363, 1996. Yoffe B, Noonan CA: Hepatitis B virus – new and evolving issues. Dig Dis Sciences 37:1-9, 1992. Yoong KF, Afford SC, Randhawa S, et al: DH: Fas/Fas ligand interaction in human colorectal hepatic metastases: A mechanism of hepatocyte destruction to facilitate local tumor invasion. Am J Pathol 154:693-703, 1999. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 130 EP 135 PG 6 ER PT J AU Sharma, ChM Arora, R Lakhtakia, R Mahapatra, KA Sarkar, Ch AF Sharma, Chand Mehar Arora, Rina Lakhtakia, Ritu Mahapatra, Kumar Ashok Sarkar, Chitra TI Ependymoma with Extensive Lipidization Mimicking Adipose Tissue: A Report of Five Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Ependymoma; adipocyte; metaplasia; lipomatous change; signet ring cell; intracranial tumor ID Ependymoma; adipocyte; metaplasia; lipomatous change; signet ring cell; intracranial tumor AB Lipomatous ependymoma is a recently described entity and only 3 cases of this variant have been reported in the literature. We report 5 cases of this rare variant of ependymoma. Patients’ age ranged from 4 years to 45 years and, interestingly, all of them were males. Two tumors were supratentorial in location, 2 in the fourth ventricle and 1 was intramedullary. Microscopically all of them showed the classical histology of ependymoma along with lipomatous differentiation. The lipomatous component was composed of cells with a large clear vacuole pushing the nucleus to the periphery and giving a signet ring cell appearance. This component demonstrated positivity for GFAP and S-100 protein thereby confirming its glial lineage. Three of the 5 tumors were high grade (WHO-grade III), had a high MIB-1 labelling index (MIB-1 LI) and showed recurrence on follow-up. However, 2 were low grade (WHO grade II) and patients are free of disease till the last follow up. C1 [Sharma, Chand Mehar] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Arora, Rina] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Lakhtakia, Ritu] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Mahapatra, Kumar Ashok] All India Institute of Medical Sciences, Department of NeurosurgeryNew Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. RP Sarkar, Ch (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM sarkarch@medinst.ernet.in CR Budka H: Intracranial lipomatous hamartomas, intracranial “lipomas”). Acta Neuropathol 28:205–222, 1974. Cenacchi G, Cerasoli S, Giangaspero F: Cerebral mixed neuronal glial neoplasm with lipidization. An immunohistochemical and ultrastructural study, Abstract 85). J Neuropathol Exp Neurol 55:626, 1996. Chimelli L, Hahn MD, Budka H: Lipomatous differentiation in a medulloblastoma. Acta Neuropathol 81:471-473, 1991. Davis DG, Wilson D, Schmitz M, et al: Lipidized medulloblastoma in adults. Human Pathol 24:900-995, 1993. Ellison DW, Zygmunt SC, Weller RO: Neurocytoma/Lipoma, neurolipocytoma, of the cerebellum. Neuropathol Appl Neurobiol 19:95-98, 1993. Giangaspero F, Cenacchi G, Roncarfil F, et al. Medullocytoma, lipidized medulloblastoma). A cerebellar neoplasm of adults with favourable prognosis. Am J Surg Pathol 20:656-64, 1996. Hirato H, Nakazato Y, Iizima M: An unusual variant of ependymoma with extensive tumor cell vacuolization. Acta Neuropathol 93:310-316, 1997. Kepes JJ, Rubinstein LJ, Eng LF: Pleomorphic xanthastrocytoma: a distinctive meningocerebral glioma of young subjects with relatively favourable prognosis. Cancer 44:1839-1852, 1979. Kepes JJ, Rubinstein LJ: Malignant gliomas with heavily lipidised, foamy, tumor cells: a report of three cases with immunoperoxidase study. Cancer 47:2451-2459, 1981. Kleihues P, Burger PC, Soheithauer BW: Histological typing of tumors of central nervous system. Ed. 2nd, Springer-Verlag. Berlin Heidelberg New York; 1993. Roda JM, Gutierrez–Molina M: Multiple intraspinal low grade astrocytomas mixed with lipoma, astrolipoma, Case report. J Neurosurgery 82:891-894, 1995. Ruchoux MM, Kepes JJ, Dhellemmes P, et al: Lipomatous differentiation in ependymomas. A report of three cases and comparison with similar changes reported in other central nervous system neoplasms of neuroectodermal origin. Am J Surg Pathol 22:338-346, 1998. Russel DS, Rubinstein LJ: Pathology of tumors of the nervous system. Vol. 2nd Ed. 6th Ed, Arnold, London; 1998. Sharma MC, Gaikwad S, Mahapatra AK, et al: Hypothalamic hamartoma: Report of a case with unusual histological features. Am J Surg Pathol 22:1538-1541, 1998. Shuster D, Herrick M, Horoupian D: Two unusual cases of central neurocytoma: a concomitantly occurring under fourth ventricular PNET, b-mixed with lipoma. Brain Pathol 4:433, 1994. Soylemezoglu F, Soffer D, Onol B, et al: Lipomatous medulloblastoma in adults. A distinct clinicopathological entity. Am J Surg Pathol 20:413-418, 1996. Walter A, Dingermans KP, Weinstein HC, Troost D: Cerebellar astrocytoma with extensive lipidization mimicking adipose tissue. Acta Neuropathol 88: 485-489, 1994. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 136 EP 140 PG 5 ER PT J AU Elek, G Minik, K Pajor, L Parlagi, Gy Varga, I Vetesi, F Zombori, J AF Elek, Gabor Minik, Karoly Pajor, Laszlo Parlagi, Gyula Varga, Istvan Vetesi, Ferenc Zombori, Janos TI New Human Dirofilariosesin Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE parasite; zoonosis; mosquito transmitted; Dirofilaria repens; histology ID parasite; zoonosis; mosquito transmitted; Dirofilaria repens; histology AB About ten cases of filariosis have recently been recorded in the Hungarian medical literature, six of them caused by Dirofilaria repens. Dirofilaria repensis a mosquito-transmitted filaroid worm in the subcutaneous tissue of dogs and cats in the temperate areas of the Old World. It accidentally infects man, too, and can remain unidentified due to physicians , poor knowledge of the parasite. In the last two years six new Dirofilaria repensinfections have been found in various parts of the country: five localised dermally and one in the deep tissues. Two of the cases might have been acquired in Italy during summer travels. Four patients, however, have never been abroad, these cases must be considered autochtonous infections. The thickness of the multilayered cuticle of the worm, diameter of the body and the size, form and number of the longitudinal ridges on its surface are used in the histological diagnosis of the the parasite. C1 [Elek, Gabor] Military Hospital, Department of Pathology, Podmaniczky u 111., H-1062 Budapest, Hungary. [Minik, Karoly] County Hospital of Borsod-Abauj-ZemplenMiskolc, Hungary. [Pajor, Laszlo] University of Pecs, Department of PathologyPecs, Hungary. [Parlagi, Gyula] Municipal Hospital, Del-Pest, Department of PathologyBudapest, Hungary. [Varga, Istvan] Szent Istvan University, Faculty of Veterinary Medicine, Department of Pathology and Forensic Veterinary MedicineBudapest, Hungary. [Vetesi, Ferenc] Municipal Hospital, Hodmezovasarhely, Department of PathologyHodmezovasarhely, Hungary. [Zombori, Janos] Veterinary University, Parasitological DepartmentBudapest, Hungary. RP Elek, G (reprint author), Military Hospital, Department of Pathology, H-1062 Budapest, Hungary. CR Ashford RW, Dowse JA, Rogers WN, et al: Dirofilariasis of the breast. The Lancet, I:1198, 1989. Avdiukhina TI, Supriaga VG, Postnova WN: Dirofilariasis in the countries of the CIS 1915-1996. Medsk Parazitol, Russia, 4:3-7, 1997. Bardach H, Heimbucher J, Raff M: Subkutane Dirofilaria, Nochtiella, repens-Infection beim Menschen – Erste Fallbeschreibung in Osterreich und Ubersicht der Literatur. Wien Klin Wschr 93:123-127, 1981. Boreham PFL, Atwell RB: Dirofilariasis. CRC Press, Boca Raton, Florida, 1988. Gutierrez Y: Diagnostic features of zoonotic filariae in tissue sections. Human Pathology 15:514-525, 1984. Kotlan A:On a new case of human filariidosis in Hungary. Acta Vet Acad Sci Hung 1:69-79, 1951. Nemeth B, Kugler S: Ophthalmo-filariasis. Orvosi Hetilap 109: 195-197, 1968. Pampiglione S, Elek G, Palfi P, et al: Human Dirofilaria repens infection in Hungary: a case in the spermatic cord and a review of the literature. Acta Vet Acad Sci Hung 47:77-83, 1999/a. Pampiglione S, Rivasi F, Canestri Trotti G: Pitfalls and difficulties in histological diagnosis of Human Dirofilariasis due to Dirofilaria, Nochtiella, repens. Diagn Microbiol Infect Dis 34:57-64, 1999/b. Pampiglione S, Bortoletti G, Fossarello M, et al: Dirofilariasi umana in Sardegna: 4 nuovi casi. Pathologica 88:472-477, 1996/a. Pampiglione S, Brollo A, Ciancia EM, et al: Dirofilariasi umana sottocutanea, 8 ulteriori casi in Italia. Pathologica 88:91-96, 1996/c. Pampiglione S, Canestri Trotti G, Rivasi F, et al: Human dirofilariasis in Greece: a review of reported cases and a description of a new, subcutaneous case. Ann Trop Med Parasitol 90:319- 328, 1996/b. Pampiglione S, Rivasi F, Paolino S: Human pulmonary dirofilariasis. Histopathology 29:69-72, 1996/d. Pampiglione S, Azzaro S, Bongiorno A, et al: La dirofilariosi oculare umana in Italia: descrizione di 6 nuovi casi. Revisione della casistica Italiana. Annali di Ottalmologia e Clinica Oculista. 121:257-277, 1995. Pampiglione S, Arlotta MR, Ambrosio ED, et al: La dirofilariasi umana nel sud d’Italia. I. Regione Puglia. Pathologica 86:528-532, 1994. Pampiglione S, Fruttaldo L, Mongio F, et al: Due casi clinici di filariasi zoonotica da probabile Dirofilaria repens. Pathologica 85:521-524, 1993. Parlagi Gy, Sumi A, Elek G, et al: Szemuregi dirofilariosis. Magyar Szemeszet, In press, 2000., in Hungarian only) Portnoy LG: Dirofilariasis. In: Pathology of infectious diseases., Eds: Conor DH, Chandler FW, Schwartz DA et al, Appleton and Lange, 1997, vol 2, pp1391-1396. Settnes OP, Engebjerg E: Human subcutaneous dirofilariasis caused by Dirofilaria repens. Apmis 99:364-370, 1991. Szell Z, Sreter T, Csikos K, et al: Autochton Dirofilaria repens fertozottseg kutyakban. Magy Ao Lapja 121:100-104, 1999., in Hungarian only) Zahler M, Glaser B, Gothe R: Eingeschleppte Parasiten bei Hunden: Dirofilaria repens und Dipetalonema reconditum. Tierarztl Prax 25:388-392 1997. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 141 EP 145 PG 5 ER PT J AU Rieker, JR Quentmeier, A Weiss, C Kretzschmar, U Amann, K Mechtersheimer, G Blaker, H Herwart, FO AF Rieker, J Ralf Quentmeier, Armin Weiss, Carsten Kretzschmar, Ulrich Amann, Kerstin Mechtersheimer, Gunhild Blaker, Hendrik Herwart, F Otto TI Cystic Lymphangioma of the Small-Bowel Mesentery SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE lymphangioma; small-bowel mesentery; infection; etiology; differential diagnosis ID lymphangioma; small-bowel mesentery; infection; etiology; differential diagnosis AB Cystic lymphangioma of the small-bowel mesentery is a rare manifestation of an intraabdominal tumor in elderly patients. We present a case of a small-bowel mesentery lymphangioma, causing fever and chills and present clinical and pathologic features. Furthermore, etiology and differential diagnosis of this tumor are discussed. C1 [Rieker, J Ralf] University of Heidelberg, Department of Pathology, Pathologisches Institut, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. [Quentmeier, Armin] St. Josephskrankenhaus, Department of Surgery and RadiologyHeidelberg, Germany. [Weiss, Carsten] St. Josephskrankenhaus, Department of Surgery and RadiologyHeidelberg, Germany. [Kretzschmar, Ulrich] St. Josephskrankenhaus, Department of Surgery and RadiologyHeidelberg, Germany. [Amann, Kerstin] University of Heidelberg, Department of Pathology, Pathologisches Institut, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. [Mechtersheimer, Gunhild] University of Heidelberg, Department of Pathology, Pathologisches Institut, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. [Blaker, Hendrik] University of Heidelberg, Department of Pathology, Pathologisches Institut, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. [Herwart, F Otto] University of Heidelberg, Department of Pathology, Pathologisches Institut, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. RP Rieker, JR (reprint author), University of Heidelberg, Department of Pathology, D-69120 Heidelberg, Germany. EM ralf_rieker@med.uni-heidelberg.de CR Barquist ES, Apple SK, Jensen DM, et al: Jejunal lymphangioma. An unusual cause of chronic gastrointestinal bleeding. Dig Dis Sci 42:1179-1183, 1997. Bill AH, Sumner DS: A unified concept of lymphangioma and cystic hygroma. Surg Gynaecol Obstet 120:79, 1965. Campbell WJ, Irwin ST, Biggart JD: Benign lymphangioma of the jejunal mesentery: an unusual cause of small bowel obstruction. Gut 32:1568, 1991. Chodak P, Hurwitz A: Lymphangiectasis of stomach simulating polypoid neoplasm. Arch Intern Med 113:225-229, 1964. Daniel S, Lazarevic B, Attia A:Lymphangioma of the mesentery of the jejunum: report of a case and a brief review of the literature. Am J Gastroenterol 78:726-729, 1983. Dowed CN. Hygroma cysticum colli. Its structure and etiology. Ann Surg 58:112-132,1913. Godart S: Embryological significance of lymphangiomas. Arch Dis Child 41:204-206, 1966. Greene EI, Kirshenn MM, Greene JM: Lymphangioma of the transverse colon. Am J Surg 103:225-229, 1964. Hanagiri T, Baba M, Shimabukuro T, et al: Lymphangioma in the small intestine: report of a case and review of the Japanese literature. Surg Today 22:363-367, 1992. Hardin WJ, Hardy JD: Mesenteric cysts. Am Surg 26:42-49, 1960. Kindblom LG, Angervall L: Tumors of lymph vessels. Contemp Issues Surg Pathol 18:163, 1991. Kok KY, Mathew VV, Yapp SK: Lymphangioma of the smallbowel mesentery: unusual cause of intestinal obstruction. J Clin Gastroenterol 24:186-187, 1997. Enzinger FM, Weiss SW: Tumors of the lymph vessels. In: Soft tissue tumors., Eds.: Enzinger FM, Weiss SW, Mosby , 1994, pp. 679-700. Nakagawara G, Kojima Y, Mai M, et al: Lymphangioma of the transverse colon treated by transendoscopic polyectomy. Dis Colon Rectum 24:291-295, 1981. Singh S, Maghrabi M: Small bowel obstruction caused by recurrent cystic lymphangioma. Br J Surg 80:1012, 1993. Takiff H, Calabria R, Yin L, et al: Mesenteric cysts and intraabdominal cystic lymphangiomas Arch Surg 120:1266-1269, 1985. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 146 EP 148 PG 3 ER PT J AU Nash, RJ AF Nash, RG John TI Pathology in the New Medical Curriculum: What has replaced the Subject Courses? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE problem-based learning; pathology; medical education ID problem-based learning; pathology; medical education AB In line with the UK General Medical Council recommendations, the traditional, taught curriculum at Liverpool was replaced from 1996 by a new one using problem-based learning (PBL) as its principal method of information transfer. There is integration of clinical and preclinical studies, coupled with a reduction in the factual knowledge content and the disappearance of identifiable separate subject courses. Learning is now student-centred. This requires a new approach to the acquisition of pathology knowledge. 1. Pathology is included in all relevant PBL case scenarios by pathology representation on module planning and review committees. 2. Special study modules (SSMs) allow students to observe the practice of pathology including surgical and autopsy work, carry out a detailed study and write a dissertation. Career selectives are provided for individual students in the final year. 3. Clinicopathological (CPC) teaching meetings are held, with the discussion of case examples, clinicians and students contributing. 4. Assessments include the input of appropriate pathology content, integrated with other subjects. 5. A pathology teaching website is provided, containing images, notes, self-assessment questions, handouts, timetables and information. Although the 1996 intake have not yet completed their studies, the results of in-course assessments have been encouraging. The response to the pathology SSMs has been very positive, and the level of presentations and dissertations reached is of a high standard. With the disappearance of a separate subject course in pathology, the subject is being learned by other routes, and the students will complete their undergraduate course with a sound basis for proceeding with their further studies. C1 [Nash, RG John] University of Liverpool, Department of Pathology, Duncan Building, Daulby Street, L69 3GA Liverpool, UK. RP Nash, RJ (reprint author), University of Liverpool, Department of Pathology, L69 3GA Liverpool, UK. EM jnash@liv.ac.uk CR Bligh JA: Problem based, small group learning. BMJ, Editorial, 311:342-343, 1995. Branda LA: Implementing problem-based learning. J Dental Education 54:548-549, 1990. Catto G: Specialist registrar training., Editorial). BMJ 320:817- 818, 2000. Ferrier BM: Problem-based learning: does it make a difference? J Dental Education 54:550-551, 1990. General Medical Council. ‘Tomorrow’s Doctors’ London, Kiek and Reid,1993. Also on GMC website: www.gmc.org.uk Neame R, Murphy B, Stitt F, Rake M: The impact of informatics. Universities without walls: evolving paradigms in medical education. BMJ 319:1296, 1999. Neufeld VR, Barrows HS: The ‘McMaster philosophy’: an approach to medical education. J Medical Education 49:1040- 1050, 1974. Pallie W, Carr DH: The McMaster medical education philosophy in theory, practice and histprical perspective. Medical Teacher 9:59-71, 1987. Shaughnessy AF, Slawson DC: Are we providing doctors with the training and tools for lifelong learning? BMJ 319:1280, 1999. Spencer JA, Jordan RK: Learner centred approaches in medical education. BMJ 318:1280-1283, 1999. Towle A: Continuing medical education: changes in health care and continuing medical education for the 21st century. BMJ 316:301-304, 1999. Walton HJ, Matthews MB: Essentials of problem-based learning. Medical Education 23:542-558, 1989. Woodward CA: The effects of innovations in medical education at McMaster: a report on follow-up studies. Meducs 2:64-68, 1989. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2000 VL 6 IS 2 BP 149 EP 154 PG 6 ER PT J AU Cohen, AS Trikha, M Mascelli, AM AF Cohen, A Sidney Trikha, Mohit Mascelli, A Mary TI Potential Future Clinical Applications for the GPIIb/IIIa Antagonist, Abciximab in Thrombosis, Vascular and Oncological Indications SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Abciximab; ReoPro; cardiocascular disease; angiogenesis; tumor metastasis ID Abciximab; ReoPro; cardiocascular disease; angiogenesis; tumor metastasis AB Abciximab (ReoPro) is a mouse-human chimeric monoclonal antibody Fab fragment of the parent murine monoclonal antibody 7E3, and was the first of these agents approved for use as adjunct therapy for the prevention of cardiac ischemic complications in patients undergoing percutaneous coronary intervention (PCI). Abciximab binds with high avidity to both the non-activated and activated form of the GPIIb/IIIa receptor of platelets, the major adhesion receptor involved in aggregation. Additional cardiovascular indications for abciximab are unstable angina, carotid stenting, ischemic stroke and peripheral vascular diseases. Abciximab also interacts with two other integrin receptors; the a av b b3 receptor, which is present in low numbers on platelets but in high density on activated endothelial and smooth muscle cells, and a aMb b2 integrin which is present on activated leukocytes. Cell types that express integrins GPIIb/IIIa and a av b b3 such as platelets, endothelial and tumor cells have been implicated in angiogenesis, tumor growth and metastasis. Since abciximab interacts with high avidity to integrins GPIIb/IIIa and a av b b3 , it is reasonable to assume that it may possess anti-angiogenic properties in angiogenesis-related diseases, as well as anti-metastastatic properties in case of disseminating tumors expressing the target integrin receptors. C1 [Cohen, A Sidney] Cenrocor Inc., 200 Great Valley Parkway, 19355 Malvern, PA, USA. [Trikha, Mohit] Cenrocor Inc., 200 Great Valley Parkway, 19355 Malvern, PA, USA. [Mascelli, A Mary] Cenrocor Inc., 200 Great Valley Parkway, 19355 Malvern, PA, USA. RP Mascelli, AM (reprint author), Cenrocor Inc., 19355 Malvern, USA. EM mascelli@centocor.com CR Davies MJ, Thomas AC, Knapman PA, et al: Intra myocardial platelet aggregation in patients with unstable angina suffering sudden ischemic cardiac death. Circulation 73:418-427, 1986. Coller BS: The role of platelets in arterial thrombosis and the rationale for blockade of platelet IIb/IIIa receptors as antithrombotic therapy. Eur Heart J 16:11-15, 1995. International Stroke Trial Collaborative Group: The International Stroke Trial, IST): a randomized trial of aspirin, subcutaneous heparin, both or neither among 19,435 patients with acute ischemic stroke. Lancet 349:1569-1581, 1997. Chinese Acute Stroke Trial, CAST): Randomized placebocontrolled trial of early aspirin use in 20,000 patients with subacute ischemic stroke. Lancet 349:1641-1649, 1997. De Scheerder I, Wang K, Wilczek K, et al: Experimental study of thrombogenicity and foreign body reaction induced by heparin-coated stents. Circulation 95(6):1549-1553, 1997. Michelson AD, Furman MI: Laboratory markers of platelet activation and their clinical significance. Curr Opin Hematol 6:342-348, 1999. Ferguson JJ, Waly HM, Wilson JM: Fundamental of coagulation and glycoprotein IIb/IIIa receptor inhibition. Am Heart J 134:S35-S42, 1998. Lefkovitz J, Plow EF, Topol EJ: Platelet glycoprotein IIb/IIIa receptors in cardiovascular medicine. New Eng J Med 332:1553-1559, 1995. EPIC Investigators: Use of a monoclonal antibody directed against the platelet glycoprotein IIb/IIIa receptor in high-risk coronary angioplasty. N Eng J Med 330:956-961, 1994. The EPILOG Investigators: Platelet glycoprotein IIb/IIIa receptor blockade and low-dose heparin during percutaneous coronary revascularization. New Eng J Med 336:1689-1696, 1997. CAPTURE Investigators: Randomised placebo controlled trial of abciximab before, during and after coronary intervention in refractory unstable angina: the CAPTURE study. Lancet 349:1429-1435, 1997. The EPISTENT Investigators: Randomised placebo-controlled and balloon angioplasty-controlled trial to assess safety of coronary stenting with use of platelet glycoprotein IIb-IIIa blockade. Lancet 352:87-92, 1998. Coller BS, Scudder LE, Beer J et al: Monoclonal antibodies to platelet glycoprotein IIb/IIIa as antithrombotic agents. Ann NY Acad Sci 614:193–213, 1991. Jordan RE, Wagner CL, Mascelli MA, et al: Preclinical development of c7E3 Fab; a mouse/human chimeric monoclonal antibody fragment that inhibits platelet function by blockade of GP IIb/IIIa receptors with observations on the immunogenicity of c7E3 Fab in humans. In: Horton MA, editor. Adhesion receptors as therapeutic targets. New York: CRC Press; 1996, pp. 281–305. Tcheng JE, Ellis SG, George BS, et al: Pharmacodynamics of chimeric glycoprotein IIb/IIIa integrin antiplatelet antibody Fab 7E3 in high-risk coronary angioplasty. Circulation 90:1757-1764, 1994. Mascelli, MA, Lance ET, Damaraju L, et al: The pharmacodynamic profile of short-term abciximab treatment demonstrates prolonged platelet inhibition with gradual recovery from GPIIb/IIIa receptor blockade. Circulation 97:1680-88, 1998. Lincoff AM, Tcheng JE, Califf RM, et al: Sustained suppression of ischemic complications of coronary intervention by platelet GPIIb/IIIa blockade with abciximab. One year outcome of the EPILOG trial. Circulation 99:1951-1958, 1999. Topol EJ, Ferguson JJ, Weisman HF, et al: Long-term prevention from myocardial ischemic events in a randomized trial of brief integrin 3 blockade with percutaneous coronary intervention J Am Med Assoc 278:479-484, 1997. Tam SH, Sassoli PM, Jordan RE, et al: Abciximab, ReoPro, chimeric 7E3 Fab, demonstrates equivalent affinity and functional blockade of glycoprotein IIb/IIIa and avb3 integrins. Circulation 98:1085–1091, 1998. Coller BS, Cheresh DA, Asch E, et al: Platelet vitronectin receptor expression differentiates Iraqi-Jewish from Arab patients with Glanzmann thrombasthemia in Israel. Blood 77:75-83, 1991. Altieri DC, Edgington TS: A monoclonal antibody reacting with distinct adhesion molecules defines a transition in the functional state of the receptor CD11b/CD18, Mac-1). J Immunol. 1414:2656-2660, 1988. Simon DI, Xu H, Ortlepp S, et al: 7E3 monoclonal antibody directed against the platelet glycoprotein IIb/IIIa cross-reacts with the leukocyte integrin Mac-1 and blocks adhesion to fibrinogen and ICAM-1. Arterioscler Thromb Vasc Biol 17:528- 535, 1977. Wagner CL, Mascelli MA, Neblock DS, et al: Analysis of GPIIb/IIIa receptor number by quantification of 7E3 binding to human platelets. Blood 3:907-914, 1996. Fidler IJ, Kumar R, Bielenberg DR, et al: Molecular determinants of angiogenesis in cancer metastasis. Canc J Sci Am 4:S58-S66,1998. Jones A, Harris AL: New developments in angiogenesis: A major mechanism for tumor growth and target for therapy. Canc J Sci Am 4:209-210,1998. Brooks PC, Montgomery AMP, Rosenfeld M, et al: Integrin avb3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell 79:1157- 1164,1994. Kerr JS, Wexler RS, Mousa SA, et al: Novel small molecule v integrin antagonists: Comparative anti-cancer efficacy with known angiogenesis inhibitors. Anticancer Research 19:959- 968,1999. Wu H, Beuerlein G, Nie Y, et al: Stepwise in vitro affinity maturation of Vitaxin, an avb3-specific humanized mAb. Proc Natl Acad Sci USA 95:6037-6042,1998. Stromblad S, Becker JC, Yebra M, et al: Suppression of p53 activity and p21WAF1/CIPI expression by vascular cell integrin alphaVbeta3 during angiogenesis. J Clin Invest 98:426- 433,1996. Brooks PC, Stromblad S, Sanders LC, et al: Localization of matrix metalloproteinase MMP-2 to the surface of invasive cells by interaction with integrin avb3. Cell 85:683-693,1996. Gasparini G, Brooks PC, Biganzoli E, et al: Vascular integrin avb3: A new prognostic indicator in breast cancer. Clin Cancer Res 4:2625-2634,1998. Carron CP, Meyer DM, Pegg JA, et al: A peptidomimetic antagonist of the integrin avb3 inhibits Leydig cell tumor growth and the development of hypercalcemia of malignancy. Cancer Research 58:1930-1935,1998. Gutheil JC, Campbell TN, Pierce, PR, et al: Phase I study of Vitaxin, an anti-angiogenic humanized monoclonal antibody to vascular integrinavb3. Proc ASCO, 1998 Abstract #832. Varner JA, Nakada MT, Jordan RE, et al: Inhibition of angiogenesis and tumor growth by murine 7E3, the parent antibody of c7E3 Fab, abciximab; ReoPro™ ). Angiogenesis 3:53- 60,1999. Hejna M, Raderer M, Zielinski CC: Inhibition of metastases by anticoagulants. J Natl Cancer Inst 91:22-36,1999. Tang DG, Honn KV: Adhesion molecules and tumor metastasis: An update. Invasion Metastasis 95:109-131,1995. Nierodzik ML, Klepfish A, Karpatkin S: Role of platelets, thrombin, integrin IIb-IIIa, fibronectin and Von Willebrand factor on tumor adhesion in vitro and metastasis in vivo. Thrombosis and Haemostasis 74:282-290,1995. Steinert BW, Tang DG, Grossi IM, et al: Studies on the role of platelet eicosanoid metabolism and integrin aIIbb3 in tumorcell- induced platelet aggregation. Int J Cancer 54:92- 101,1993. Timar J, Chopra H, Rong X, et al: Calcium channel blocker treatment of tumor cells induces alterations in the cytoskeleton, mobility of the integrin aIIbb3 and tumor-cell-induced platelet aggregation. J Cancer Res Clin Oncol 118:425- 434,1992. Tang DG, Grossi IM, Tang KQ et al: Inhibition of TPA and 12(S)-HETE-stimulated tumor cell adhesion by prostacyclin and its stable analogs: Rationale for their antimetastatic effects. Int J Cancer 60:418-425,1995. Amirkhosravi A, Amaya M, Siddiqui F, et al: Blockade of GPIIb/IIIa inhibits the release of vascular endothelial growth factor, VEGF, from tumor cell-activated platelets and experimental metastasis. Platelets 10:285-292,1999. Pinedo HM, Verheul HMW, D’Amato RJ, et al: Involvement of platelets in tumour angiogenesis? Lancet 352:1775- 1777,1998. Heyn Adu P, Lotter MG, Badenhorst PN, et al: Kinetics distribution and site of destruction of 111indium-labeled human platelets. Br J Haematol 44:269-280,1980. Chronos N, Marciniak SJ, Nakada MT: Binding specificity and associated effects of platelet GPIIb/IIIa inhibitors. Eur Heart J 1S: E11-E17, 1999. Mohle R, Green D, Moore MAS, et al: Constitutive production and thrombin-induced release of vascular endothelial growth factor by human megakaryocytes and platelets. Proc Natl Acad Sci USA 94:663-668,1997. Maloney JP, Silliman CC, Ambruso DR, et al: In vitro release of vascular endothelial growth factor during platelet aggregation. Am J Physiol 275:H1054-H1061,1998. Hieken TJ, Farolan M, Ronan SG, et al: Integrin expression in melanoma predicts subsequent metastasis. J Surgical Research 63:169-173,1996. Hieken TJ, Ronan SG, Farolan M, et al: Molecular prognostic markers in intermediate-thickness cutaneous malignant melanoma. Cancer 85:375-382,1999. Hsu M-Y, Shih D-T, Meier FE, et al: Adenoviral gene transfer of 3 integrin subunit induces conversion from radial to vertical growth phase in primary human melanoma. Am J Pathol 153:1435-1442,1998. Montgomery AMP, Reisfeld RA, Cheresh DA: Integrin avb3 rescues melanoma cells from apoptosis in three-dimensional dermal collagen. Proc Natl Acad Sci USA 91:8856- 8860,1994. Felding-Habermann B, Mueller BM, Romerdahl CA, et al: Involvement of integrin v gene expression in human melanoma tumorigenicity. J Clin Invest 89:2018-2022,1992. Petitclerc E, Stromblad S, von Schalscha TL, et al: Integrin avb3 promotes M21 melanoma growth in human skin by regulating tumor cell survival. Cancer Research 59:2724- 2730,1999. Puerschel W Ch, Gawaz M, Worret W-I, et al: Immunoreactivity of glycoprotein IIb is present in metastasized but not in non-metastasized primary malignant melanoma. British J Dermatology135: 883-887,1996. Oleksowicz L, Mrowiec Z, Schwartz E, et al: Characterization of tumor-induced platelet aggregation: The role of immunorelated GPIb and GPIIb/IIIa expression by MCF-7 breast cancer cells. Thrombosis Research 79:261-274,1995. Timar J, Trikha M, Szekeres K, et al: Autocrine motility factor signals integrin-mediated metastatic melanoma cell adhesion and invasion. Cancer Research 56:1902-1908,1996. Chen YQ, Trikha M, Gao X, et al: Ectopic expression of platelet integrin IIb3 in tumor cells from various species and histological origin. Int J Cancer 72:642-648,1997. Trikha M, Timar J, Lundy, SK, et al: The high affinity IIb3 integrin is involved in invasion of human melanoma cells. Cancer Research 57:2522-2528,1997. Trikha M, Timar J, Lundy SK, et al: Human prostate carcinoma cells express functional IIb3 integrin. Cancer Research 56:5071-5078,1996. Noguchi CT, Schechter AN: The intracellular polymerization of sickle hemoglobin and its relevance to sickle cell disease. Blood 58:1057-1068, 1981. Deisseroth A, Nienhuis A, Lawrence J, et al: Chromosomal localization of the human globin gene to chromosome 11 in somatic cell hybrids. Proc Natl Acad Sci USA 75:1456-1460, 1978. Kaul DK, Fabry ME, Nagel RL; The pathophysiology of vascular obstruction in the sickle syndromes. Blood 10:29- 48,1996. Luzzatto L: Genetics of red cells and susceptibility to malaria. Blood 54:961-967,1979. Vichinsky E, Hurst D, Earles A, et al: Newborn screening for sickle cell disease: effect on mortality. Pediatrics 81:749- 755,1988. Hoppe C, Styles L, Vichinsky E: The natural history of sickle cell disease. Curr Opin Pediatr 10:49-52,1998. Solovey A, Lin Y, Browne P, et al: Circulating activated endothelial cells in sickle cell anemia. N Eng J Med 337:1585- 1590,1997. Joneckis CC, Ackley Rl, Orringer EP, et al: Integrin alpha 4 beta 1 and glycoprotein IV, CD36, are expressed on circulating reticuloxytes in sickle cell anemia. Blood 82:3548- 3555,1993 Sugihara K, Sugihara T, Mohandas N, et al: Thrombospondin mediates adherence of CD36+ sickle reticulocytes to endothelial cells. Blood 80:2634-2642,1992. Gee BE, Platt OS: Sickle reticulocytes adhere to VCAM-1. Blood 83:268-274,1995. Setty BN, Stuart MJ: Vascular cell adhesion molecule-1 is involved in mediating hypoxia-induced sickle red cell adherence to endothelium: potential role in sickle cell disease. Blood 88:2311-2320,1996. Brittain HA, Eckman JR, Swerlick RA, et al: Thrombospondin from activated platelets promotes sickle erythrocyte adherence to human microvascular endothelium under physiologic flow: a potential role for platelet activation in sickle cell vascular occlusion. Blood 81:2137-2143,1993. Kaul DK, Chen D, Zhan J: Adhesion of sickle cells to vascular endothelium is critically dependent on changes in density and shape of cells. Blood 83:3006-3017,1994. Wick TM, Moake JL, Udden MM, et al: Unusually large von Willebrand factor multimers increase adhesion of sickle erythrocytes to human endothelial cells under controlled flow. J Clin Invest 80:905-910,1987. Kaul DK, Tsai HM, Liu XD, et al: Monoclonal antibodies to avb3, 7E3 and LM609, inhibit sickle red blood cell-endothelium interactions induced by platelet activating factor. Blood 95:368-374,2000. Oh, SO, Ibe BO, Johnson C, et al: Platelet-activating factor in plasma of patients with sickle cell disease in steady state. J Lab Clin Med 130:191-196,1997. Hashemi S, Palmer DS, Aye MT, et al: Platelet-activating factor secreted by DDAVP-treated monocytes mediates von Willebrand factor release from endothelial cells. J Cell Physiol 154:496-505,1993. Wun T, Paglieroni T, Field CL, et al: Platelet-erythrocyte adhesion in sickle cell disease. J Invest Med 47:121-127,1999. Buerling-Harbury C, Schade SG: Platelet activation during crisis in sickle cell anemia patients. Am J Haematol 31:237-241,1989. Sacco RL, Wolf PA, Kannel WB, et al: Survival and recurrence following stroke. The Framingham study. Stroke 13:290-295, 1982. Goldstein LB, Matchar DB: Clinical assessment of stroke. J Am Med Ass 271:1114-1120, 1994. Warlott: Epidemiology of stroke. Lancet 352(Supp III): 1-4, 1998. Broderick J, Brott T, Kothari R, et al: The Greater Cincinnati/ Northern Kentucky Stroke Study: preliminary first-ever and total incidence rates of stroke among blacks. Stroke 29:415-421, 1998. Gresham GE, Phillips TF, Wolf PA, et al: Epidemiologic profile of long-term stroke disability: the Framingham study. Arch Phys Med Rehabil. 60:487-491, 1979. Sandercock PAG, van den Belt AGM, Lindley RI, et al: Antithrombotic therapy in acute ischemic stroke: an overview of the completed randomized trials. J Neurol Neurosurg Psychiatr 56:17-25, 1993. Taylor TN, Davis PH, Torner JC, et al: Lifetime cost of stroke in the United States. Stroke 27:1459-1466, 1996. Broderick JP, Phillips SJ, Whisnant JP, et al: Incidence rates of stroke in the eighties: the end of the decline in stroke? Stroke 20:577-582, 1989. The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group: Tissue plasminogen activator for acute ischemic stroke. N Engl J Med 333:1581-1587, 1995. The NINDS t-PA Stroke Study Group: Intracerebral hemorrhage after intravenous t-PA therapy for ischemic stroke. Stroke 28:2109-2118, 1997. Hacke W, Kaste M, Fieschi C, et al: Intravenous thrombolysis with recombinant tissue plasminogen activator for acute hemispheric stroke. The European Cooperative Acute Stroke Study. J Am Med Assoc 274:1017-1025, 1995. Hacke W, Kaste M, Fieschi C, et al: Randomised double-blind placebo-controlled trial of thrombolytic therapy with intravenous alteplase in acute ischaemic stroke, ECASS II). Second European-Australasian Acute Stroke Study Investigators. Lancet 352:1245-1251, 1998. The Multicenter Acute Stroke Trial – Europe Study Group: Thrombolytic therapy with streptokinase in acute ischemic stroke. N Engl J Med 335:145-150, 1996. Multicentre Acute Stroke Trial – Italy, MAST-I, Group: Randomised controlled trial of streptokinase, aspirin and combination of both in treatment of acute ischemic stroke. Lancet 346:1509-1514, 1995. CAPRIE Steering Committee: A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events, CAPRIE). Lancet 348:1329-39, 1996. Ohman EM, Lincoff AM, Bode C, et al: Enhanced Early Reperfusion at 60 minutes with Low-does Retaplase Combined with Full-dose Abciximab in Acute Myocardial Infarction: Preliminary Results from the GUSTO-4 Pilot, SPEED, Dose-ranging trial, Circulation 98S1:504-511, 1998. Antman EM, Giugliano RP, Gibson CM, et al: Abciximab facilitates the rate and extent of thrombolysis: results of the Thrombolysis in Myocardial Infarction, TIMI, 14 trial. Circulation 99:2720-2732, 1999. Cohen I, Berk DL, White JG: The effect of peptides and monoclonal antibodies that bind to glycoprotein IIb/IIIa complex on the development of clot tension. Blood 73:1880-1887, 1989. Cox AD, Devine DV: Factor XIIIa binding to activated platelets is mediated through the activation of glycoprotein IIb-IIIa. Blood 83:1006-1016, 1994. Adams HP, Bogouslavsky Barnathan E, Polzer J, et al: Preliminary safety report of a randomized, double-blind dose escalation trial of abciximab, ReoPro, in acute ischemic stroke. Cerebrovasc Dis 9S1:27, 1999. Chastain HD, Wong PM, Mathur A, et al: Does abciximab reduce complications of cerebral vascular stenting in high risk sesions? Circulation 96:1-283, 1997. The Global Use of Strategies to Open Occluded Arteries, GUSTO III, Investigators.report Assessment of the Safety and Efficacy of a New Thrombolytic, ASSENT-2, Investigators: A comparison of retaplase with alteplase for acute myocardial infarction. N Engl J Med 337:1118-1123, 1997. The ASSENT-2 double blind randomized trial: Single-bolus tenecteplase compared with front-loaded alteplase in acute myocardial infarction. Lancet 354:716-722, 1999. ISIS-2, Second International Study of Infarct Survival, Collaborative Group: Randomized trial of intravenous streptokinase, oral aspirin, both, or neither among 17,187 cases of suspected acute myocardial infarction: ISIS-2. Lancet 2:349- 360, 1988. Kleiman N, Ohman EM, Califf RM, et al: Profound inhibition of platelet aggregation with monoclonal antibody 7E3 fab after thrombolytic therapy: results of the Thrombolysis and Angioplasty in Myocardial Infarction, TAMI, 8 pilot study. J Am Coll Cardiol 22:381-389, 1993. Ohman EM, Kleiman NS, Gacioch G, et al: Combined accelerated tissue-plasminogen activator and platelet glycoprotein IIb/IIIa integrin receptor blockade with Integrilin in acute myocardial infarction: results of a randomized, placebo-controlled, dose-ranging trial. Circulation 95:846-854, 1997. Moliterno DJ, Harrington RA, Califf RM, et al: Randomized, placebo-controlled study of lamifiban with thrombolytic therapy for the treatment of acute myocardial infarction: rationale and design for the Platelet Aggregation Receptor Antagonist Dose Investigation and Reperfusion Gain in Myocardial Infarction, PARADIGM, study. J Thromb Thrombol 2:165- 169, 1995. Miller JM, Smalling R, Ohman EM, et al: Effectiveness of early coronary angioplasty and abciximab for failed thrombolysis, reteplase and alteplase, during acite myoccardial infarction, results from the GUSTO-III trial). Am J Card 84:779- 784, 1999. Marso SP, Lincoff AM, Ellis SG, et al: Optimizing the percutaneous interventional oucomes for patients with diabetes mellitis: results of the EPISTENT diabetic substudy. Circulation 100:2477-2484, 1999. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 163 EP 174 PG 12 ER PT J AU Andras, Cs Csiki, Z Gal, I Takacs, I Antal, L Szegedi, Gy AF Andras, Csilla Csiki, Zoltan Gal, Istvan Takacs, Istvan Antal, Lajos Szegedi, Gyula TI Retrospective Evaluation of 5-fluorouracil-interferon-a aTreatment of Advanced Colorectal Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE advanced colorectal cancer; interferon; 5-FU; survival ID advanced colorectal cancer; interferon; 5-FU; survival AB The authors describe the retrospective analysis of treatment by 5-fluorouracil and interferon-a aof 34 patients with advanced colorectal cancer. An average of 4.6 treatment cycles (3–12) was applied. Complete remission was not observed; partial remission was observed in 8 patients; in 13 patients no change occurred and progression was detected in 14 cases. Remission rate was 22.8%, mean response time was 5.2 (3–12) months, mean progress-free survival 5.6 (0–22) months. Mean survival from the start of treatment was 11.9 (1–42) months and from the establishment of the diagnosis 26.1 (3–60) months. Severe life-threatening side-effects did not occur; other side-effects such as fever, nausea, diarrhea, leucopenia, and anemia responded to drugs. Treatment by 5-FU and interferon, in accordance with other authors’ findings, improved survival and well-being of patients but no breakthrough has been achieved. C1 [Andras, Csilla] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. Krt. 22., H-4004 Debrecen, Hungary. [Csiki, Zoltan] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. Krt. 22., H-4004 Debrecen, Hungary. [Gal, Istvan] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. Krt. 22., H-4004 Debrecen, Hungary. [Takacs, Istvan] Medical Faculty, University of Debrecen, 2 nd Department of SurgeryDebrecen, Hungary. [Antal, Lajos] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. Krt. 22., H-4004 Debrecen, Hungary. [Szegedi, Gyula] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. Krt. 22., H-4004 Debrecen, Hungary. RP Andras, Cs (reprint author), University of Debrecen, Medical and Health Center, 3rd Department of Medicine, H-4004 Debrecen, Hungary. CR Bleiberg H: Colorectal Cancer – Is there an alternative to 5FU? Eur J Cancer 33:536-541, 1997. Burke D, Allan-Mersh TG. Colorectal liver metastases. Postgrad Med J 72:464-469, 1996. Gamelin EC, Danquechin-Dorval EM, et al: Relationship between 5FU dose intensity and therapeutic response in patients with advanced colorectal cancer receiving infusional therapy containing 5FU. Cancer 77:441-451, 1996. Dufour P and Husseini F.5-fluorouracil plus alpha interferon as treatment of metastatic colorectal carcinoma. Ann Oncol 7:575- 579, 1996. Ferguson JE, Hulse P, Lorigan P, et al: Coninuous infusion of 5 fluorouracil with alpha 2b interferon for advanced colorectal carcinoma. Brit J Cancer 72:193-197, 1995. Jaeck D, Bachellier P, Guiguet M, et al: Long-term survival following resection of coloretal hepatic metastases. British J Surg 84:977-980, 1997. Kemeny N, Jounes A, Seiter K, et al: Interferon alpha 2a and 5 fluorouracil for advanced colorectal carcinoma. Assesment of activity and toxicity. Cancer 66:2470-2475, 1990. Meadows ML, Linley C: Biochemical and pharmacolgic modulation of fluorouracil by alpha interferon. Advanced colorectal cancer: The role of alpha interferons. Schering Plough International 18:10-14, 1992. Niederle N, Kreuser ED, Meadows LM, et al: Advanced colorectal cancer . The role of alpha interferons. Schering Plough International 18:14-18, 1992. Pazdur R, Ajani JA, Patt YZ: Phase II study of fluorouracil and recombinant interferon alpha 2a in previously untreated advanced colorectal carcinoma. J Clin Oncol 8:2027-2031, 1990. Piga A, Cascinu S, Latini L, et al: A phase II randomised trial of 5-fluorouracil with or without intferon alpha-2a in advanced colorectal cancer. British J Cancer 74:971-974, 1996. Raderer M, Scheithauer W: Treatment of advanced colorectal cancer with 5-fluorouracil and interferin alpha: an overview of clinical trials. Eur J Cancer 31:1002-1008, 1995. Ragnhammar P, Blomgren H, Edler D, et al: Different dose regimens of 5-fluorouracil and interferon alpha in patients with metastatic colorectal carcinoma. Eur J Cancer 31:310-320, 1995. Schmoll HJ: Development of treatment of advanced colorectal cancer: 5-FU infusional and the role of new agents. Eur J Cancer 32:18-22, 1996. Schuller J, Czejka M: Influence of interferon 2b on pharmacocintics of 5-Fluorouracil. Advanced colorectal cancer. The role of alpha interferons. Schering Plough International 18:6- 10, 1993. Stadler W, Coffier B, Pazdur R: Therapeutic advantages in oncology. ASCO Congress Report Series 1997; 2-9, 1997. van Triest B, van Groeningen CJ, Pinedo HM: Current chemoterapeutic possibilities in the treatment of colorectal cancer. Eur J Cancer 31:1193-1197, 1995. Wadler S, Wiernik PM: Clinical update on the roleof fluorouracil and recurrent interferon alpha 2a in the treatment of colorectal cancer. Semin Oncol 8:2027-2031,1990. Woll PJ, Pettengell R: Interferons in oncology. J Clin Oncol 15:1432-1438, 1997. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 175 EP 178 PG 4 ER PT J AU Baintner, K Jakab, G Gyori, Zs Kiss, P AF Baintner, Karoly Jakab, Gabor Gyori, Zsuzsa Kiss, Peter TI Binding of FITC-Labelled Lectins to the Gastrointestinal Epithelium of the Rat SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE rat; gut; stomach; lectin; ConA; LCA; PHA; RPA; PNA; WGA; SNA; UEA ID rat; gut; stomach; lectin; ConA; LCA; PHA; RPA; PNA; WGA; SNA; UEA AB Biotechnology uses lectin genes to transfect into crop plants for protection against insects and nematodes. On the other hand, the information is limited on lectin-binding properties of cells in the gastrointestinal tract. Therefore, binding of a panel of FITC-labelled plant lectins to gastrointestinal cells of the rat was studied. In the stomach, cytoplasmic staining of parietal cells by PHA appeared to be due to glycoproteins attached to the tubulovesicles. PNA also stained the parietal cells, but only in the isthmus and neck regions, reacting with desialylated glycoproteins. WGA bound to the mucous neck cells with higher affinity than to the surface and foveolar mucous cells. The mucous cells were also stained by SNA-I, UEA-I and, less intensively, by LCA. Chief cells did not show detectable reaction with any of the applied lectins. Binding of PHA to gastric cells showed differences when compared with the results of in vivostudies. Small intestinal brush border was stained with UEA-I and SNA-I, the latter lectin also strongly stained the surface of small intestinal crypts. Both lectins reacted with the mucus of goblet cells. In the large intestine UEA-I and SNA-I stained the goblet cells at the base and upper part of the crypts, respectively. Accordingly, we provided evidences for the unique lectin-binding phenotype of the various segments of the gastrointestinal tract. C1 [Baintner, Karoly] Faculty of Animal Science, University of Kaposvar, Department of Physiology, H-7401 Kaposvar, Hungary. [Jakab, Gabor] Uzsoki Hospital, Department of NeurologyBudapest, Hungary. [Gyori, Zsuzsa] Uzsoki Hospital, Department of NeurologyBudapest, Hungary. [Kiss, Peter] University of Agriculture, Department of Agricultural ChemistryGodollo, Hungary. RP Baintner, K (reprint author), Faculty of Animal Science, University of Kaposvar, Department of Physiology, H-7401 Kaposvar, Hungary. EM baintner@atk.kaposvar.pate.hu CR Bardocz S, Grant G, Ewen SWB, et al: Reversible effect of phytohaemagglutinin on the growth and metabolism of rat gastrointestinal tract. Gut 37:353-360,1995. Broekaert WF, Nsimba-Lubaki M, Peeters B, Peumans WJ: A lectin from elder, Sambucus nigraL., bark. Biochem J 221:163-169,1984. Callaghan JM, Toh B-H, Pettitt JM, et al: Poly-N-acetyllactosamine- specific tomato lectin interacts with gastric parietal cells. J Cell Science 95:563-576,1990. Ewen SWB, Pusztai A: Effects of diets containing genetically modified potatoes expressing Galanthus nivalis lectin on rat small intestine. Lancet 354:1353-4,1999. Fischer J: Alternative ultrastructural localization of Dolichos biflorus lectin binding sites in proton secreting parietal cells of mice. Biochemistry 87:1-4,1989. Goldkorn I, Gleeson PA and Toh B-H: Gastric parietal cell antigens of 60-90 kDa, and 100-120 kDa associated with autoimmune gastritis and pernicious anaemia. Role of N-glycans in the structure and antigenicity of the 60-90 kDa component. J Biol Chem 264:18768-74, 1989. Karam SM, Yao X, Forte JG: Functional heterogeneity of parietal cells along the pit-gland axis. Am J Physiol 272: G161-G171, 1997. Pusztai, A: Plant Lectins. Cambridge University Press.1991. Pusztai A, Ewen SWB, Grant G et al: The relationship between survival and binding of plant lectins during small intestinal passage and their effectiveness as growth factors. Digestion 46:, suppl. 2, 308-316,1990. Schulte B and Spicer SS: Light microscopic histochemical detection of terminal galactose and N-acetylgalactosamine residues in rodent complex carbohydrates using a galactose oxidase-Schiff sequence and peanut lectin-horseradish peroxidase conjugate. J Histochem Cytochem 31:19-24,1983. Suzuki S, Tsuyama S and Murata F: Post-embedding staining of rat gastric mucous cells with lectins. Histochemistry 73:563-575,1982. Van Damme EJM, Barre A, Smeets K, et al: The bark of Robinia pseudoacacia contains a complex mixture of lectins. Plant Physiol 107:833-843,1995. Van Damme EJM, Peumans WJ, Pusztai A, et al: Handbook of Plant Lectins. John Wiley & Sons, Chichester, 1998. Wang Q, Lu-Gang Y, Campbell BJ, et al: Identification of intact lectin in peripheral venous blood. Lancet 352:1831-1832,1998. Zolotarev AS, Townsend RR, Stuart-Tilley A, et al: HCO3 –dependent conformational change in gastric parietal cell AE2, a glycoprotein naturally lacking sialic acid. Am J Physiol 271:G311- G321,1996. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 179 EP 182 PG 4 ER PT J AU Gharagozloo, S Sharifian, AR Mageed, AR Shokri, F AF Gharagozloo, Soheila Sharifian, A Ramazan Mageed, A Rizgar Shokri, Fazel TI Analysis of the Expressed Immunoglobulin Variable Region Heavy Chain Gene Products in Paraproteins from Iranian Patients with Multiple Myeloma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE multiple myeloma; VH genes; paraprotein; cross-reactive idiotype ID multiple myeloma; VH genes; paraprotein; cross-reactive idiotype AB The frequency of expression of immunoglobulin (Ig) variable region heavy (VH ) chain gene products was studied in 43 Iranian patients with mutiple myeloma (MM). The expressed VH gene families and associated cross-reactive idiotypes (CRI) were analysed by immunoblotting and ELISA, using peptide-induced polyclonal antibodies specific for VH 1-VH 6 gene families and monoclonal antibodies (MAb) recognising CRI linked to theVH 1, VH 3, VH 4 and VH 6 gene families. The results revealed that the VH 3 family (60.5%) was the most predominant gene family. In contrast, no paraproteins were encoded by genes from the VH 2 gene family and only 2.3% were encoded by the VH 5 family. The panel of paraproteins tested rarely expressed the probed VH -associated CRI. Our results suggest that: 1-The Ig VH genes, may not be randomly expressed in the malignant plasma cells from Iranian patients with MM. 2- Some of the genes seem to be negatively selected or highly mutated, as evidenced by the lack of expression of the probed CRI. C1 [Gharagozloo, Soheila] School of Medical Sciences, Tarbiat Modarres University,, Department of Immunology, 14155 Tehran, Iran. [Sharifian, A Ramazan] School of Medicine, Tehran University of Medical Sciences, Clinic of Hematology and OncologyTehran, Iran. [Mageed, A Rizgar] School of Medicine, Tehran University of Medical Sciences, Clinic of Hematology and OncologyTehran, Iran. [Shokri, Fazel] School of Medical Sciences, Tarbiat Modarres University,, Department of Immunology, 14155 Tehran, Iran. RP Shokri, F (reprint author), School of Medical Sciences, Tarbiat Modarres University,, Department of Immunology, 14155 Tehran, Iran. CR Nishimoto N, Suematsu S, Kishimoto T: Plasma cell dyscrasias. Adv Immunol., 64:218-281, 1997. Liu P, Leong T, Quam L, et al: Activating mutations of N-and K-ras in multiple myeloma show different clinical associations Analysis of the Eastern Cooperative Oncology Group phase III trial. Blood 88:2699-2706, 1996. Dao DD, Sawyer JR, Epstein J, et al: Detection of retinoblastoma gene in multiple myeloma. Leukemia 8:1280-1284, 1994. Harada H, Kawano MM, Huang N, et al: Phenotypic difference of normal plasma cells from mature myeloma cells. Blood 81:2658- 2663, 1993. Mahmoud MS, Huang N, Nobuyoshi M, et al: Altered expression of pax-5 gene in human myeloma. Blood 4311-4315, 1996. Juliusson G, Gahrton G: Chromosome abnormalities in multiple myeloma. In: Multiple Myeloma, G. Gahrton and B. Durie, Eds.), Oxford University Press, Oxford, pp: 55-63, 1996. Bergsagel PL, Nardini E, Brents L, et al:IgH translocations in multiple myeloma: a nearly universal event that rarely involves c-myc. Curr Top Microbiol Immunol 224:283-287, 1997. Nishida K, Tamura A, Nakazawa N, et al: The Ig heavy chain gene is frequently involved in chromosomal translocations in multiple myeloma and plasma cell leukemia as detected by in situ hybridization. Blood 90:526-534, 1997. Bakkus MHC, Heirman C, Van Riet I, et al: Evidence that multiple myeloma Ig heavy VDJ genes contain somatic mutations but show no intraclonal variation. Blood 8:2326-2335, 1992. Sahota S, Hamblin T, Oscier DG, et al: Assessment of the role of clonogenic B lymphocytes in the pathogenesis of multiple myeloma. Leukemia 8:1285-1289, 1994. Vesico RA, Cao Hong CH, Lee JC, et al: The heavy chain variable region in multiple myeloma shows evidence of prior somatic mutation and antigen selection but the lack of intraclonal diversity or clonal progression. J Immunol 155:2487-2497, 1995. Cogne M, Guglielme P: Exon skipping without splice site mutation accounting for abnormal Ig chains in nonsecretory human myeloma. Eur J Immunol 23:1289-1293, 1993. Sahota S, Leo R, Hamblin TJ, et al: Myeloma VL and VH gene sequences reveal a complementary imprint of antigen selection in tumor cells. Blood 89:219-226, 1997. Silverman GJ, Goldfein RD, Chen PP, et al: Idiotypic and subgroup analysis of human rheumatoid factors: implications for structural and genetic basis of autoantibodies in humans. J Clin Invest 82:469-475, 1987. Shokri F, Mageed RA, Richardson P, et al: Modulation and high frequency expression of autoantibody associated cross-reactive idiotypes linked to the VH1 subgroup in CD5 expressing B lymphocytes from patients with chronic lymphocytic leukemia. Scand J Immunol 37:673-679, 1993. Ono M, Winearls CG, Amus N, et al: Monoclonal antibodies to restricted and cross-reactive idiotypes on monoclonal RF and their recognition of idiotype-positive cells. Eur J Immunol 17:343-349, 1987. Stevenson FK, Smith GJ, North J, et al: Identification of normal Bcells counterparts of neoplastic cells which secrete cold agglutinins of anti-I and anti-i specificity. Br J Hematol 72:9-15, 1989. Van Es JH, Raaphorst FM, Vantol MJD, et al:Expression pattern of the most JH proximal human VH gene segment, VH6, in the B cell and antibody repertoire suggests a role of VH6-encoded IgM antibodies in early ontogeny. J Immunol 150:1-8, 1993. Brouet JC, Fermand JP: Multiple myeloma. In: Samters Immunologic Disease, M.M. Frank, K.F. Austen, H.N. Claman, E.R. Unanue, Eds., New York: Little Brown, pp: 623-636, 1995. Shokri F, Mageed RA, Maziak BR, et al: Lymphoproliferation in primary Sjogren’s syndrome: Evidence of selective expansion of a B cell subset characterized by the expansion of cross-reactive idiotypes. Arth Rheum 36:1128-1136, 1993. Shokri F, Mageed RA, Jefferis R: A quantitative ELISA for measurment of RF-associated CRI in serum from patients with rheumatic disease. Br J Rheumatol 32:862-869, 1993. Deane M, Baker BVY, Norton JD: Immunoglobulin VH4 gene usage in B lymphoid leukemias. Br J Hematol 84:242-249, 1993. Kipps TJ, Tomhave E, Pratt LF, et al: Developmentary restricted Ig heavy chain variable region gene expressed at high frequency in chronic lymphocytic leukemia. Proc Natl Acad Sci USA 86:5913- 5917, 1989. Shokri F, Mageed RA, Richardson P, et al: Idiotypic and immunophenotypic characterization of the leukemic B-cells from patients with prolymphocytic leukemia: evidence for a selective expression of immunoglobulin variable region, IgV, gene products. Leuk Res 17:669-676, 1993. Moazzeni SM, Mosayyebi G, Stevenson FK, et al: Biased utilization of immunoglobulin variable region heavy and light chain genes by the malignant CD5- B-cells from patients with Burkitt’s lymphoma. Int J Cancer 58:226-232, 1994. Chapman CJ, Zhou JX, Gregory C, et al: VH and VL gene analysis in sporadic Burkitt’s lymphoma shows hypermutation intraclonal heterogeneity, and a role for antigen selection. Blood 88:3562- 3568, 1996. Hsu FJ, Levy R: Preferential use of the VH4 Ig gene family by the diffuse large cell lymphoma. Blood 86:3072-3082, 1995. Rettig MB, Vesico RA, Cao J, et al: VH gene usage in multiple myeloma: Complete absence of the VH4.21, VH4-34, gene. Blood 87:2846-2852, 1996. Cook GP, Tomlinson IM: The human Ig VH repertoire. Immunol Today 16:237-242, 1995. Vargaz-Madrazo E, Lara-Ochoa F, Ramirez-Benites MC:Evolution of the structural repertoire of the human VH and VK germline genes. Int Immunol 9:1801-1815, 1997. Kraj P, Friedman DF, Stevenson FK, et al: Evidence for the overexpression of the VH4-31, VH4.21, Ig gene segment in the normal adult B cell repertoire. J Immunol 154:6406-6420, 1995. Brezinschek HP, Brezinschek RI, Lipsky PE: Analysis of the heavy chain repertoire of human peripheral B cells using single cell PCR. J Immunol 155:190-202, 1995. Schroeder HW, Wang JY: Preferential utilization of conserved Ig heavy chain variable gene segments during human fetal life. Proc Natl Acad Sci USA 87:6146-6150, 1990. Pasqual V, Victor K, Lelsz D, et al: Nucleotide sequence analysis of the V-regions of two IgM cold agglutinins: evidence that the VH4.21 gene segment is responsible for the major cross-reactive idiotypes. J Immunol 146:4385-4391, 1991. Potter KN, Li YC, Capra JD: The cross-reactive idiotopes recognized by the monoclonal antibodies 9G4 and Lc1 are located in framework region 1 of two non-overlapping subsets of human VH4 family encoded antibodies. Scand J Immunol 40:43-49, 1994. Stevenson FK, Spellerberg MB, Chapman CJ, et al: Differential usage of an autoantibody-associated VH gene, VH4-21, by human B-cell tumors. Leuk Lymph 16:379-384, 1995. Fischer M, Kuppers R: Human IgA- and IgM-secreting intestinal plasma cells carry heavily mutated VH region genes. Eur J Immunol 28:2971-2977, 1998. Pratt LF, Szubin R, Carson DA, et al: Molecular characterization of a supratypic cross-reactive idiotype associated with IgM autoantibodies. J Immunol 147:2041-2046, 1991. Deane M, Mackenzie LE, Stevenson FK, et al: The genetic basis of human VH4 gene family-associated CRI expression in CD5+ and CD5-cord blood B-lymphocyte clones. Scand J Immunol 38:348- 358, 1993. Shokri F, Mageed RA, Tunn E, et al: Qualitative and quantitative expression of VH1-associated cross-reactive idiotypes within IgMRF from patients with early synovitis. Ann Rheum Dis 49:150- 154, 1990. Shokri F, Mageed RA, Maziak BR, et al: Expression of VHIII-associated cross-reactive idiotypes on human B-lymphocytes: Association with staphylococcal protein A, SPA, binding and Staphylococcus aureus Cowan I, SAC, stimulation. J Immunol 146:936- 940, 1991. Potter KN, Li Y, Mageed RA, et al: Anti-idiotypic antibody D12 and superantigen SPA both interact with human VH3-encoded antibodies on the external face of the heavy chain involving FR1, CDR2 and FR3. Molec Immunol 35:1179-1187, 1998. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 185 EP 190 PG 6 ER PT J AU Volavsek, M Masera, A Ovcak, Z AF Volavsek, Metka Masera, Andrej Ovcak, Zdenka TI Incidental Prostatic Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE angiogenesis; prostate; incidental carcinoma; transurethral resection ID angiogenesis; prostate; incidental carcinoma; transurethral resection AB Incidental prostatic carcinoma (ICP) has good prognosis related to low stage at diagnosis. Few rogressive cases demanding aggressive treatment need early identification. Neoangiogenesis proved its predictive role in prostatic carcinoma after radical prostatectomy. To reveal its value in ICP authors investigated specimens after transurethral resection of prostate (TURP). Retrospective study was performed on 68 ICP diagnosed in years 1985–1989. Microvessels highlighted by factor VIII were counted in a x200 microscope field (0,8012 mm 2 ) in most active areas of neovascularisation. Microvessel count was correlated with tumor differentiation degree, Gleason score, disease stage, and patients’ survival in at least 9 years after diagnosis. Higher maximal microvessel counts were associated with lower degree of tumor differentiation (p=0,005), Gleason score (p=0,001), and disease stage (0,003). No association with disease progression and patients’ survival was found. Mean microvessel counts showed less significant values when correlated with tumor differentiation degree (p=0,003) and Gleason score (p=0,01), and no correlation with other variables. Microvessel density in TURP specimens of ICP retains its prognostic value already demonstrated in carcinoma of peripheral prostatic lobes. Maximal microvessel counts were prognostically more reliable than mean values. C1 [Volavsek, Metka] University of Ljubljana, Faculty of Medicine, Institute of Pathology, Korytkova 2, 1000 Ljubljana, Slovenia. [Masera, Andrej] University of Ljubljana, Faculty of Medicine, Institute of Pathology, Korytkova 2, 1000 Ljubljana, Slovenia. [Ovcak, Zdenka] University of Ljubljana, Faculty of Medicine, Institute of Pathology, Korytkova 2, 1000 Ljubljana, Slovenia. RP Volavsek, M (reprint author), University of Ljubljana, Faculty of Medicine, Institute of Pathology, 1000 Ljubljana, Slovenia. CR Adolfsson J, Ronstrom L, Carstensen J, et al: The natural course of low grade, non-metastatic prostatic carcinoma. Br J Urol 65:611-614, 1990. Barth PJ, Weingartner K, Kohler HH, et al: Assessment of the vascularisation in prostatic carcinoma: A morphometric investigation. Hum Pathol 27:1306-1310, 1996. Bostwick DG, Qian J: Current and proposed biologic markers in prostate cancer. J Cell Biochem 19(Suppl): 197-201, 1994. Bostwick DG, Wheeler TM, Blute M, et al: Optimized microvessel density analysis improves prediction of cancer stage from prostate needle biopsies. Urology 48:47-57, 1996. Brawer MK, Deering RE, Brown M, et al: Predictors of pathologic stage in prostatic carcinoma. The role of neovascularity. Cancer 73:678-687, 1994. Brawer MK: Quantitative microvessel density. A staging and prognostic marker for human prostatic carcinoma. Cancer 78:345-349, 1996. Byar DP, Corle DK, and Veterans Administration Cooperative Urological Research Group: VACURG randomized trial of radical prostatectomy for stages I and II prostate cancer. Urology 17(Suppl): 7-11, 1981. Eble JN, Epstein JI: Stage A carcinoma of the prostate In: Contemporary issues in surgical pathology. Pathology of the prostate., Ed: Bostwick DG), Churchill Livingstone, 1990, pp. 61-82. Folkman J, Watson K, Ingber D, et al: Induction of angiogenesis during the transition from hyperplasia to neoplasia. Nature 339:58-61, 1989. Folkman J: Tumor angiogenesis. In: The molecular basis of cancer., Eds: Mendelsohn J, Howley PM, Israel MA, Liotta LA), WB Saunders Company, 1995, pp. 206-32. Fox SB: Tumor angiogenesis and prognosis. Histopathology 30:294-301, 1997. Fregene TA, Khanuja PS, Noto AC, et al: Tumor-associated angiogenesis in prostate cancer. Anticancer Res 13:2377-2382, 1993. Furusato M, Wakui S, Sasaki H, et al: Tumour angiogenesis in latent prostatic carcinoma. Br J Cancer 70:1244-1246, 1994. Gleason DF: Histologic grading of prostatic carcinoma. In: Contemporary issues in surgical pathology. Pathology of the prostate., Ed: Bostwick DG), Churchill Livingstone, 1990, pp. 83-93. Greene DR, Egawa S, Neerhut G, et al: The distribution of residual cancer in radical prostatectomy specimens in stage A prostate cancer. J Urol 145:324-329, 1991. Grignon DJ, Sakr WA: Zonal origin of prostatic adenocarcinoma: Are there biologic differences between transition zone and peripheral zone adenocarcinomas of the prostate gland? J Cell Biochem 19(Suppl): 267-269, 1994. Hall MC, Troncoso P, Pollack A, et al: Significance of tumor angiogenesis in clinically localized prostate carcinoma treated with external beam radiotherapy. Urology 44:869-875, 1994. Isaacs JT: Prostatic cancer: An age-old problem. In: The underlying molecular, cellular, and immunological factors in cancer and aging., Eds: Yang SS, Warner HR), Plenum Press, 1993, pp. 167-84. Johnstone PA, Riffenburgh R, Saunders EL, et al: Grading inaccuracies in diagnostic biopsies revealing prostatic adenocarcinoma: implications for definitive radiation therapy. Int J Radiat Oncol 32:479-482, 1995. Jones EC, Young RH: The differential diagnosis of prostatic carcinoma. Its distinction from premalignant and pseudocarcinomatous lesions of the prostate gland. Am J Clin Pathol 101:48-64, 1994. Lerner SP, Seale-Hawkins C, Carlton Jr CE, et al: The risk of dying of prostate cancer in patients with clinically localised diasease. J Urol 146:1040-1045, 1991. Mc Neal JE, Redwine EA, Freiha FS, et al: Zonal distribution of prostatic adenocarcinoma. Correlation with histologic pattern and direction of spread. Am J Surg Pathol 12:897-906, 1988. Mohler JL, Partin AW, Epstein JI, et al: Prediction of prognosis in untreated stage A2 prostatic carcinoma. Cancer 69:511-519, 1992. Mostofi FK, Davis Jr CJ, Sesterhenn IA: Pathology of carcinoma of the prostate. Cancer 70:235-253, 1992. Pompe-Kirn V: Epidemiološke znaeilnosti raka prostate v Sloveniji. In: Rak prostate. 12. onkološki vikend. Zbornik predavanj., Ed: Marolt F), Laško, 1997, pp 25-33. Schlingemann RO, Rietveld FJR, Kwaspen F, et al: Differential expression of markers for endothelial cells, pericytes, and basal lamina in the microvasculature of tumors and granulation tissue. Am J Pathol 138:1335-1347, 1991. Schroder FH: The natural history of incidental prostatic carcinoma. In: Incidental carcinoma of the prostate., Eds: Altwein JE, Faul P, Schneider W), Springer-Verlag, 1991, pp 56-62. Sedmak B: Diagnostieni postopki pri raku prostate. In: Rak prostate. 12. onkološki vikend. Zbornik predavanj., Ed: Marolt F), Laško, 1997, pp 54-61. Siegal JA, Yu E, Brawer MK: Topography of neovascularity in human prostate carcinoma. Cancer 75:2545-2551, 1995. Smith-Mc Cune KK, Weidner N: Demonstration and characterisation of the angiogenic properties of cervical dysplasia. Cancer Res 54:800-804, 1994. Vartanian RK, Weidner N: Endothelial cell proliferation in prostatic carcinoma and prostatic hyperplasia: Correlation with Gleason’s score, microvessel density, and epithelial cell proliferation. Lab Invest 73:844-850, 1995. Weidner N, Semple JP, Welch WR, et al: Tumor angiogenesis and metastasis-correlation in invasive breast carcinoma. N Engl J Med 324:1-8, 1991. Weidner N, Folkman J, Pozza F, et al: Tumor angiogenesis: A new significant and independent prognostic indicator in earlystage breast carcinoma. J Natl Cancer Inst 84:1875-1887, 1992. Weidner N, Carroll PR, Flax J, et al: Tumor angiogenesis correlates with metastasis in invasive prostate carcinoma, Am J Pathol 143:401-409, 1993. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 191 EP 196 PG 6 ER PT J AU Onguru, Celasun, B AF Onguru, Onder Celasun, Bulent TI Intra-hospital Use of a Telepathology System SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE telepathology; frozen section ID telepathology; frozen section AB Utilization of telepathology systems to cover distant geographical areas has increased recently. However, the potential usefulness of similar systems for closer distances does not seem to be widely appreciated. In this study, we present data on the use of a simple telepathology system connecting the pathology department and the intra-operative consultation room within the operating theaters of the hospital. Ninety-eight frozen section cases from a past period have been re-evaluated using a real-time setup. Forty-eight of the cases have been re-evaluated in the customary fashion; allowing both ends to communicate and cooperate freely. Fifty of the cases, however, were evaluated by the consultant while the operating room end behaved like a ''robot”; moving the stage of the microscope, changing and focusing the objectives. The deferral rate was lower than the original frozen section evaluations. Overall, the sensitivity was 100%, specificity 98%, negative predictive value 96,5% and positive predictive value 100%. No significant difference was found for the diagnostic performances between the cooperative and robotic simulation methods.Our results strengthen the belief that telepathology is a valuable tool in offering pathology services to remote areas. The far side of a hospital building can also be a remote area and a low cost system can be helpful for intraoperative consultations. Educational value of such a system is also commendable. C1 [Onguru, Onder] Gulhane Medical Military Academy, Department of Pathology, Etlik, 06018 Ankara, Turkey. [Celasun, Bulent] Gulhane Medical Military Academy, Department of Pathology, Etlik, 06018 Ankara, Turkey. RP Onguru, (reprint author), Gulhane Medical Military Academy, Department of Pathology, 06018 Ankara, Turkey. CR Callas PW, Leslie KO, Mattia AR, et al: Diagnostic accuracy of a rural live video telepathology system. Am J of Surg Pathol 21:812-819, 1997. Celasun B, Aksu A, Safali M, et al: Frozen section as a diagnostic method: Evaluation of 1316 cases. Bull Ankara Pathol Soc 9:41-48, 1992. Dunn BE, Almagro UA, Choi H, et al: Dynamic-robotic telepathology: Department of Veterans Affairs feasibility study. Hum Pathol 28:8-12, 1997. Eusebi V, Foschini L, Erde S, et al: Transcontinental consults in surgical pathology via the Internet. Hum Pathol 28:13-16, 1997. Kayser K, Kayser G: Basic Aspects of and recent developments in telepathology in Europe, with specific emphasis on quality assurance. Analyt Quant Cytol Histol 21:319-328, 1999. Kuakpaetoon T, Stauch G, Visalsawadi P: Image quality and acceptance of telepathology. Adv Clin Path 2:305-312, 1998. Nordrum I: Real-time diagnoses in telepathology. Adv Clin Pathol 2:127-189, 1998. Oberholzer M, Fischer HR, Christen H, et al: Telepathology with an integrated services digital network-A new tool for image transfer in surgical pathology: A preliminary report. Hum Pathol 24:1078-1085, 1993. Raab SS, Robinson RA, Snider TE, et al: Telepathologic review: Utility, diagnostic accuracy, and interobserver variability on a difficult case consultation service. Mod Pathol 10:630-635, 1997. Weinstein RS: Telepathology comes of age in Norway, editorial). Hum Pathol 22:511-513, 1991. 10. Weinstein RS, Bhattacharryya AK, Graham AR, et al: Telepathology: a ten year progress report. Hum Pathol 28:1-7, 1997. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 197 EP 201 PG 5 ER PT J AU Tokes, AM Paku, S Toth, S Paal, E Kulka, J Toth, J Telekes, A AF Tokes, Anna-Maria Paku, Sandor Toth, Sara Paal, Edina Kulka, Janina Toth, Jozsef Telekes, Andras TI Tenascin Expression in Primary and Recurrent Breast Carcinomas and the Effect of Tenascin on Breast Tumor Cell Cultures SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tenascin; breast cancer; cell cultures; focal adhesion plaques ID tenascin; breast cancer; cell cultures; focal adhesion plaques AB Tenascin is generally classified as an anti-adhesive protein. Many cells do not adhere to tenascin or if they adhere they do not spread. In this study we analysed the stromal expression of tenascin-C in primary, second primary and recurrent breast carcinomas and the ability of tenascin-C to stimulate the focal adhesion plaques in MDA-MB-435 breast carcinoma cell line. To assess the tenascin-C expression formalin-fixed, paraffin-embedded specimens of 20 specially selected breast carcinomas and their recurrences (14) or a second primary breast cancer of the same patient (6) were examined with immunohistochemical methods. We also studied the effect of tenascin-C on focal adhesion plaques added to MDA-MB-435 breast carcinoma cell line. During a median 2,9-year patient follow up 14 local recurrences and 6-second primary breast carcinomas developed in the 20 patients. In 3 cases a second recurrence occurred. The presence of tenascin in tumor cells, in the proliferating and some normal ducts, near to the tumor cell nests, in the stroma and in ductal carcinoma in situ component of the invasive carcinoma may suggest the role of tenascin played in tumor cell migration. Soluble tenascin added to the cell culture had minimal or no effect on focal adhesion plaques. Tenascin only seems not to be of prognostic value in predicting the local recurrence of breast cancer. C1 [Tokes, Anna-Maria] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Paku, Sandor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Toth, Sara] Semmelweis University, Department of Genetics, Cell and ImmunobiologyBudapest, Hungary. [Paal, Edina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Toth, Jozsef] National Institute of OncologyBudapest, Hungary. [Telekes, Andras] National Institute of OncologyBudapest, Hungary. RP Tokes, AM (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. CR Bourdon MA, Ruoslahti E: Tenascin mediates cell attachment through an RGD-Dependent Receptor. J Cell Biol 108:1149- 1155, 1989. Canfield AD, Schor AM: Evidence that tenascin and trombospondin- 1 modulate sprouting of endothelial cells. J Cell Sci 108:797-809, 1995. Chi-Rosso G, Gotwals PJ, Yang J, et al: Fibronectin type III repeats mediate RGD-independent adhesion and signaling through activated beta1 integrins. J Biol Chem 272:31447- 31452, 1997. Chiquet M, Fambrought DM: Chick myotendinous antigen II. A novel extracellular glycoprotein complex consisting of large disulfide-linked subunit. J Cell Biol. 98:1937-1946, 1984. Chiquet Ehrismann R, Mackie EJ, et al: Tenascin: an extracellular matrix protein involved in tissue interactions during fetal development and oncogenesis. Cell 47:131-139, 1986. Chiquet-Ehrismann R, Kalla P et al: Tenascin interferes with fibronectin. Cell 53:383-390, 1988. Chiquet-Ehrismann R, Kalla P, Pearson CA: Participation of tenascin and transforming growht factor- in reciprocal epithelial- mesenchymal interactions of MCF7 cells and fibroblasts. Cancer Res 49:4322-4325, 1989. Chiquet-Ehrismann R: Anti-adhesive molecules of the extracellular matrix. Curr Opin Cell Biol 3:800-804, 1991. Chiquet-Ehrismann R, Hagios C, Schenk S: The complexity in regulating the expression of tenascins. BioEssays 17:873-878, 1995. Choung C-M, Chen H-M: Enhanced expression of neural cell adhesion molecules and tenascin, cytotactin, during wound healing. Am J Pathol 138:427-440, 1991. Crossin KL, Hoffmann S, Grumet M, et al: Site restricted expression of cytotactin during development of the chicken embryo. J Cell Biol 102:1917-1930, 1986. Elston CW, Ellis IO: Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long term follow up. Histopathol 19:403-410, 1991. Hahn AW, Kern F, Jonas U, et al: Functional aspect of vascular tenascin-C expression. J Vasc Res 32:162-174, 1995. Hibino S, Kato K, Kudoh S, et al: Tenascin suppresses CD3- mediated T cell activation. Biochem-Biophys-Res-Commun 250:119-124, 1998. Ishihara A, Yoshida T, Tamaki H, et al: Tenascin expression in cancer cells and stroma of human breast cancer and its prognostic significance. Clin Cancer Res 1:1035-1041, 1995. Jahkola T, Toivonen T, Virtanen I, et al: Tenascin-C expression in invasion border of early breast cancer: a predictor of local and distant reccurence. British J Cancer 78:1507-1513, 1998. Jaskiewicz K, Chasen MR, Robson SC: Differential expression of extracellular matrix proteins and integrins in hepatocellular carcinoma and chronic liver disease. Anticancer Res 13:2229- 2237, 1993. Jessen LR, Petersen WO, Bissell MJ: Cellular changes involved in conversion of normal to malignant breast: importance of the stromal reaction. Physiological Reviews 76:69-125, 1996. Jones PL, Crack J, Rabinovitch M: Regulation of tenascin-C, a vascular smooth muscle cell survival factor that interacts with the v3 integrin to promote epidermal growth factor receptor phosphorylation and growth. J of Cell Biol 139:279-293, 1997. Kawakatsu H, Shiurba R, Obara M, et al: Human carcinoma cells synthesize and secrete tenascin in vitro. Jpn J Cancer Res 83:1073-1080, 1992. Koukoulis GK, Howeedy AA, Korhonen M, et al: Distribution of tenascin, cellular fibronectin and integrins in the normal, hyperplastic and neoplastic breast. J Submicrosc Cytol Pathol 25:285-295, 1993. Mackie EJ, Chiquet-Ehrismann R, Pearson CA, et al: Tenascin is a stromal marker for epithelial malignancy in the mammary gland. Proc Natl Acad Sci 84:4621-4625, 1987. Murphy-Ullrich JE, Lightenasciner VA, Aukhil I, et al: Focal adhesion integrity is downregulated by the alternativily spliced domain of human tenascin. J Cell Biol 115:1127-1136, 1991. Price JE, Fabra A, Zhang RD, et al: Characterization of variants of human breast cancer cell line isolated from metastases in different organs of nude mice. Int J Oncol 5:459-467, 1994. Sage HE, Bornstein P: Extracellular proteins that modulate cellmatrix interactions. SPARC, tenascin and trombospondin. J Biol Chem. 266:14831-14834, 1991. Sakakkura T, Ishihara A, Yatani R: TN-Cin mammary gland development: from embryogenesis to carcinogenesis. In Regulatory Mechanism in Breast Cancer, Lippman ME, ed, pp. 383- 400. Advances in cellular and molecular biology of breast cancer. Kluwer Academic Publishers: Boston., 1991 Shaw LM, Chao C, Wewer UM, et al: Function of the integrin alpha 6 beta 1 in metastatic breast carcinoma cells assessed by expression of a dominant-negative receptor. Cancer Res 56:959-963, 1996. Shoji T, Kamiya T, Tsubura A, et al: Immunohistochemical staining patterns of tenascin in invasive breast carcinomas. Virchows Archiv A Pathol Anat. 421:53-56, 1992. Sriramarao P, Mendler M, Bourdon MA: Endothelial cell attachment and spreading on human tenascin is mediated by alpha 2 beta 1 and alpha v beta 3 integrins. J Cell Sci. 105:1001- 1012, 1993. Taylor HC, Lightenasciner VA, Beyer WF, et al: Biochemical and structurel studies of tenascin/hexabranchion proteins. J Cell Biochem 41:71-90, 1989. Tokes A.M, Hortovanyi E, Csordas Gy, et al: Immunohistochemical localisation of tenascin in invasive ductal carcinoma of the breast. Anticancer Res 19:175-180, 1999. Tremble P, Chiquet-Ehrismann R, Werb Z: The extracellular matrix ligands fibronectin and tenascin collaborate in regulating collagenase gene expression in fibroblasts. Mol Biol Cell 5:439-453, 1994. Vacca A, Ribatti D, Fanelli M, et al: Expression of tenascin is related to histologic malignancy and angiogenesis in b-cell non- Hodgkin’s lymphomas. Leuk Lymphoma 22:473-481, 1996. Wewer UM, Shaw LM, Albrechtsen R, et al: The integrin alpha 6 beta 1 promotes the survival of metastatic human breast carcinoma cells in mice. Am J of Pathol 151:1191-1198, 1997. Wong NC, Mueller BM, Barbas CF, et al: Alphav integrins mediate adhesion and migration of breast carcinoma cell lines. Clin Exp Metastasis 16:50-61, 1998. Yoshida T, Ishihara A, Hirokawa Y, et al: Tenascin in breast cancer development-is epithelial tenascin a marker for poor prognosis. Cancer Lett 90:65-73, 1995. Yoshida T, Matsumoto E, Hanamura N, et al: Co-expression of tenascin and fibronectin in epithelial and stromal cells of beningn lesions and ductal carcinomas in the human breast. J Pathol 182:421-428, 1997. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 202 EP 209 PG 8 ER PT J AU Rubio, L Burgos, J Morera, C Vera-Sempere, F AF Rubio, Luis Burgos, S. Javier Morera, Calderon Vera-Sempere, J. Francisco TI Morphometric Study of Tumor Angiogenesis as a New Prognostic Factor in Nasopharyngeal Carcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE angiogenesis; morphometry; nasopharyngeal carcinona ID angiogenesis; morphometry; nasopharyngeal carcinona AB The aim of this study was to determine the possible prognostic significance of tumor angiogenesis (TA) in nasopharyngeal carcinoma (NPC) patients. Fifty-five NPC patients were evaluated in relation to survival. Endothelial cells were immunohistochemically stained with anti-von Willebrand factor (F-VIII), CD-31 and CD-34 antibodies, and microvessels counted in the most active areas of tumor neovascularization or hotspotsusing both a manual and an automatic method. Overall survival analysis calculated by the Kaplan–Meiertest revealed that both methods were correlated with a statistical significance between intratumoral microvessel density (IMD) and overall survival, using either manual (p=0.0141) or automatic counting (p=0.0117). Other angiogenic parameters studied were perimeter, roundness and accumulative area of the microvessels using a morphometric analyzer. Moreover, our results show that cases with high IMD demonstrated a prognostic significance in relation to the accumulative area (p=0.0072). C1 [Rubio, Luis] University of Valencia, Department of Pathology, Avda. Campanar 21, 46009 Valencia, Spain. [Burgos, S. Javier] CSIC-Autonomous University of Madrid, Center of Molecular Biology, Severo OchoaMadrid, Spain. [Morera, Calderon] University Hospital La Fe, Service of ORLValencia, Spain. [Vera-Sempere, J. Francisco] University of Valencia, Department of Pathology, Avda. Campanar 21, 46009 Valencia, Spain. RP Vera-Sempere, F (reprint author), University of Valencia, Department of Pathology, 46009 Valencia, Spain. CR Nicholls JM: Nasopharyngeal carcinoma: classification and histologic appearances. Adv. Anat. Pathol. 4:71-84, 1996. Vera-Sempere FJ, Burgos JS, Botella MS, et al: Immunohistochemical expression of Epstein-Barr virus, EBV)-encoded latent membrane protein, LMP-1, in paraffin sections of EBVassociated nasopharyngeal carcinoma, NPC, in Spanish patients. Oral Oncol Eur J Cancer 32B:163-166, 1996. Vera-Sempere FJ, Burgos JS, Botella MS: Comparative analysis of Epstein-Barr virus, EBV, detection by nested-PCR and nonisotopic in situ hybridization, NISH, in nasopharyngeal carcinoma. Clin Chem Acta 271:119-132, 1998. Liebowitz D: Nasopharyngeal carcinoma: the Epstein-Barr virus association. Semin. Oncol. 21:376-381, 1994. Shanmugaratnam K, Sobin LH: Histological typing of upper respiratory tract tumors. International histological classification of tumors, WHO, Geneva 19:20-32, 1978. Norrby K: Angiogenesis: new aspects relating to its initiation and control. APMIS 105:417-437, 1997. Folkman J: What is the evidence that tumors are angiogenesisdependent? J Natl Cancer Inst 82:4-6, 1990. Weidner N, Semple JP, Welch WR, et al: Tumor angiogenesis and metastasis-correlation in invasive breast carcinoma. N Engl J Med 324:1-8, 1991. Weidner N, Folkman J, Pozza F, et al: Tumor angiogenesis: a new significant and independent prognostic indicator in earlystage breast carcinoma. J Natl Cancer Inst 84:1875-1887, 1992. Toi M, Kashitani J, Tominaga T:Tumor angiogenesis is an independent prognostic indicator in primary breast carcinoma. Int J Cancer 55:371-374, 1993. Ogawa Y, Chung Y-S, Nakata B, et al: Microvessel quantitation in invasive breast cancer by staining for factor VIII-related antigen. Br J Cancer 71:1297-1301, 1995. Fontanini G, Bigini D, Vignati S, et al: Microvessel count predicts metastatic disease and survival in non-small cell cancer. J Pathol 177:57-63, 1995. Simpson JF, Ahn C, Battifora H, et al: Endothelial area as a prognostic indicator for invasive breast carcinoma. Cancer 77:2077-2085, 1996. Micheau C: What’s new in histological classification and recognition of nasopharyngeal carcinoma, NPC)? Path Res Pract 181:249-253, 1986. De Jong JS, Van Diest PJ, Baak JPA: Heterogeneity and reproducibility of microvessel counts in breast cancer. Lab Invest 73:922-926, 1995. Tomisaki S, Ohno S, Ichiyoshi Y, et al: Microvessel quantification and its possible relation with liver metastasis in colorectal cancer. Cancer 77:1722-1728, 1996. Tanigawa N, Matsumara M, Amaya H, et al: Tumor vascularity correlates with the prognosis of patients with esophageal squamous cell carcinoma. Cancer 79:220-225, 1997. Quian CN, Min HQ, Liang XM, et al: Primary study of neovasculature correlating with metastatic nasopharyngeal carcinoma using computer image analysis. J Cancer Res Clin Oncol 123:645-651, 1997. Roychowdhury DF, Tseng Jr. A, Fu KK, et al: New prognostic factors in nasopharyngeal carcinoma. Tumor angiogenesis and C-erbB2 expression. Cancer 77:1419- 1426, 1996. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 210 EP 216 PG 7 ER PT J AU Bhattacharya, P Maity, P AF Bhattacharya, Pritha Maity, Putul TI Localization of Phosphate Dependent Glutaminase in Ascites Fluid of Ovarian Cancer Patient SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ovarian cancer; ascites fluid; glutaminase ID Ovarian cancer; ascites fluid; glutaminase AB Phosphate dependent glutaminase was purified from ascites fluid of ovarian cancer patients. The purified enzyme showed a final specific activity of 110 unit / mg protein with 72 fold purification and 21% yield. Purified enzyme gives one dark band of Mr ~65.5 KD and two light bands of Mr ~47.5 KD and ~45 KD respectively on 10% SDS-PAGE. One major immunoreactive band was found in trans-immunoblot analysis using antibodies against rat kidney and ascites fluid glutaminase raised in rabbit and mice respectively. Phosphate dependent glutaminase enzyme purified from mitochondria of malignant and non malignant ovarian tissue also showed bands of same molecular weight on 10% SDS-PAGE and gave same immunoreactive bands in trans-immunoblot like the purified glutaminase from ascites fluid. This result was confirmed by using the specific activity stain for glutaminase, which indicates that same enzyme activity is probably due to leakage of the same enzyme from malignant tissue into the ascites fluid. The purified enzyme from human peritoneal fluid showed a high specificity toward glutamine, therefore is a true glutaminase. Moreover, ascites fluid taken from patients of different age group with different stages of ovarian carcinoma revealed the presence of same glutaminase on 10% SDS-PAGE, and exhibited immunoreaction on ELISA, trans-immunoblot and dot immunoblot analysis. C1 [Bhattacharya, Pritha] Chittaranjan National Cancer Institute, Department of Metabolic Regulation, 37, S. P. Mukherjee Road, 16, 700 026 Calcutta, India. [Maity, Putul] Chittaranjan National Cancer Institute, Department of Metabolic Regulation, 37, S. P. Mukherjee Road, 16, 700 026 Calcutta, India. RP Maity, P (reprint author), Chittaranjan National Cancer Institute, Department of Metabolic Regulation, 700 026 Calcutta, India. CR Newsholm EA, Parry-Billings M: Properties of glutamine released by skeletal muscle and its importance to the immune system. J Parenter Enter Nutr 14:63S, 1990. Newsholm EA, Crabtree B, Ardawi MSM: Glutamine metabolism in lymphocytes, its biochemical, physiological and clinical importance. Q J Exp Physiol 70:473-489, 1985. Watford M: Glutamine metabolism in the small intestine: synthesis of three carbon end products in isolated enterocytes. Biochim Biophys Acta 120:73-78, 1994. Windmueller HG: Glutamine utilization by the small intestine. Adv Enzymol 53:201-237, 1982. Windmueller HG, Spaeth AE: Uptake and metabolism of plasma glutamine by the small intestine. J Biol Chem 249:5070- 5079,1974. Abou-Khalil WH, Yunis AA, Abou-Khalil S: prominent glutamine oxidation activity in mitochondria of haematopoetic tumors. Cancer Res 43:1990-1993, 1983. Moreadith RW, Lehringer AL: The pathway of glutamate and glutamine oxidation by tumor cell mitochondria. Role of mitochondrial NAD(P)+ dependent malic enzyme. J Biol Chem 259:6215-6221,1984. Lazarus P, Panasci LC: Characterisation of L-threonine and Lglutamine transport in murine p-388 leukemia cells in vitro. Presence of an N-like amino acid transport system. Biochim Biophys Acta 856:488-495,1986. Coles NW, Johnstone RN: Glutamine metabolism on Ehrlich ascites carcinoma cells. Biochem J 83:284-291,1961. Horowitz ML, Knox WE, Morris HP: Glutaminase activities and growth rates of rat hepatomas. Cancer Res 29:1195-1199,1969. Knox WE, Horowitz ML, Fridell GH: The proportionality of glutaminase content to growth rate of morphology of rat neoplasms. Cancer Res 29:669-680, 1969. Kovacevic Z, Morris HP: The role of glutamine in the oxidative metabolism of malignant cells. Cancer Res 32:326-335,1972. Krebs H: Metabolism of amino acids IV. The synthesis of glutamine from glutamic acid and ammonia and the enzymatic hydrolysis of glutamine on animal tissue. Biochem J 29:1951-1969,1935. Robert J, Holcenberg JS, Dolowy WC: Antineoplastic activity of highly purified bacterial glutaminases. Nature, Lond, 227:1136-1137, 1970 Chakraborty P, Srivastava GC: Ratio of plasma glutaminases in liquid and solid tumor bearing mice. Indian J Med Res 60:800- 807,1972. Lund P: In methods of enzymatic analysis, Bergmeyer HE ed, Verlag chemie wenheim 8:357-359,1985. Lowry OH, Rosenbourgh NJ, Farr AL, et al: Protein measurement with pholine phenol reagent. J Biol Chem 193:265-275, 1951. Quesada AR, Sanchez-Jimenez F, Perez-Rodriguez J, et al: Purification of phosphate dependent glutaminase from isolated mitochondria of Ehrlich ascites tumor cells. Biochem J 256:1031-1036, 1988. Laemmli UK: Nature 227:680-685, 1970. Towbin H, Staehelin T, Gordon J: Electrophoretie transfer of proteins from polacrylamide gels to nitrocellulose sheets: Procedure and some application. Proc Natl Acad Sci, USA, 76:4350-4354, 1979. Curthoys NP, Kuhlenschmidt T, Godfrey SS, et al: Phosphate dependent glutaminase from rat kidney. Cause of increased activity in response to acidosis and identity with glutaminase from other issues. Arch Biochem Biophys 172:162-167, 1976a. Haser WG, Shapiro RA, Curthoys NP: Comparison of the phosphate dependent glutaminase obtained from rat kidney and brain. Biochem J 229:399-408, 1985. Curthoys NP, Kuhlenschmidt T, Godfrey SS: Regulation of renal ammoniagenesis. Purification and characterization of phosphate dependent glutaminase from rat kidney. Arch Biochem Biophys 174:82-89,1976. Chiu JF, Boeker EA: Cow brain glutaminase: partial purification and mechanism of action. Arch biochem Biophys 196:493- 500,1979. Kvamme E, Tveit B, Svenneby G: Glutaminase from pig renal cortex. Its purification and general properties. J Biol Chem 245:1871-1877,1970. Svenneby G, Torgner IA, Kvamme E: Purification of phosphate dependent pig brain glutaminase. J Neurochem 20:1217-1224, 1973. Olsen RB, Torgner IA, Christensen TB, et al: J Mol Biol 74:239- 251,1973. Shapiro RA, Haser WG, Curthoys NP: Immunoblot analysis of glutaminase peptides in intact and solubilized mitochondria isolated from various rat tissues. Biochem J 242:743- 747,1987. Heini HG, Gebhardt R, Mecke D: Purification and characterization of rat liver glutaminase. Eur J Biochem 162:541-546, 1987. Rodbard D, Chrambach A: Stain for glutaminase activity. Anal Biochem 54:272-275, 1979. Choudhury L, Srivastava GC: Intracellular localization of glutamine- aminohydrolase in normal and malignant tissues. Experientia 29:856-857, 1973. Bonnie S, Dunbar DS, Eric DS: Preparation of polyclonal antibodies. In MP Dentscher, Ed, Methods In Enzymology, Academic Press California pp. 663-670, 1990. Escribano MJ, Benedi VJ, Cordier J: Assay of antigens antibodies by fixation to nitrocellulose sheets and immunodetection: Immunologia 2:71-76, 1963. Voller A, Biddwell DE, Barlett A: The ELISA Dynatch Laboratories, Inc Alexandria, Virginia ,1979. Maity P, Chakraborty S, Bhattacharya P, et al: Isolation and purification of phosphate dependent glutaminase from Sarcoma- 180 tumor and its antineoplastic effects on murine model system: J Exp Clin Cancer Res 18:475-480,1999. Pal S, Maity P: Investigation on glutamine amidohydrolase, EC3.5.1.2, and glutamine aminotransferase, EC 2.5.1.15, activity in liver and plasma of EAC bearing mice following glutaminase therapy. Cancer Letters 66:225-231,1992. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 217 EP 223 PG 7 ER PT J AU Szekely, E Schaff, Zs Madaras, L Kupcsulik, P Zsirka, A AF Szekely, Eszter Schaff, Zsuzsa Madaras, Lilla Kupcsulik, Peter Zsirka, Attila TI Trabecular Angiomyolipoma Mimicking Hepatic Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE hepatic tumor; angiomyolipoma; trabecular; epithelioid ID hepatic tumor; angiomyolipoma; trabecular; epithelioid AB Hepatic angiomyolipomas are rare tumors, especially in comparison with those occurring in the kidney. Nevertheless, it is important to be aware of their existence, especially when occurring in the liver, where they might have different subtypes. Not infrequently they are composed of rather irregular cells with epithelioid appearance. In these cases hepatocellular carcinoma or the possibility of other malignant tumors has to be ruled out, with the aid of numerous immunohistochemical reactions. The authors present a case of a female patient, whose liver lesion was first diagnosed on cytological examination as a hepatocellular carcinoma. Based on the preoperative cytological diagnosis, a large liver lobe resection was performed. Histological examination found an angiomyolipoma of the above-mentioned type, and the final diagnosis was ascertained with the aid of vimentin, smooth muscle actin (SMA), and HMB-45. C1 [Szekely, Eszter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Madaras, Lilla] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kupcsulik, Peter] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Zsirka, Attila] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. RP Szekely, E (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM szesz@korb2.sote.hu CR Tsui WMS, Colombari R, Portmann BC: Hepatic angiomyolipoma A clinicopathologic study of 30 cases and delineation of unusual morphologic variants Am J Surg Pathol 23:34-48, 1999. Chan JKC, Tsang WYW, Pau MY, et al: Lymphangiomyomatosis and angiomyolipoma:closely related entities characterized by hamartomatous proliferation of HMB-45 positive smooth muscle. Histopathology 22:445-455, 1993. Eble JN, Amin MB, Young RH: Epithelioid angiomyolipoma of the kidney: a report of five cases with a prominent and diagnostically confusing epithelioid smooth muscle component. Am J Surg Pathol 21:1123-1130, 1997. Guinee DG, Thornberry DS, Azumi N, et al: Unique pulmonary presentation of an angiomyolipoma. Am J Surg Pathol 19:476- 480, 1995. Martignoni G, Pea M, Bonetti, et al: Carcinomalike monotypic epithelioid angiomyolipoma in patients without evidence of tuberous sclerosis: a clinicopathologic and genetic study. Am J Surg Pathol 22:663-672, 1998. Nonomura A, Mizukami Y, Muraoka K: Angiomyolipoma of the liver with pleomorphic histological features. Histopathology 24:279-281, 1994. Kaiserling E, Krober S. XiaoJ-C, et al: Angiomyolipoma of the kidney. Immunoreactivity with HMB-45, light and electronmicroscopic findings. Histopathology 25:41-48, 1994. Wick MR, Humphrey PA, Ritter JH: Pathology of pseudoneoplastic lesions. Lippincott-Raven, 1997, pp 163-165. Sturtz CL, Dabbs DJ: Angiomyolipomas: the nature and expression of the HMB45 antigen. Mod, ???, Pathol 7:842-845, 1994. Tsui WMS, Yuen AKT, Ma KF, et al: Hepatic angiomyolipoma with deceptive trabecular pattern and HMB-45 reactivity. Histopathology 21:569-573, 1992. Tsui WMS, Ng IOL, Colombary R, et al: Hepatic angiomyolipomas, letter). Histopathology 22: 602-603, 1993. Ma TKF, Tse MK, Tsui WMS, et al: Fine needle aspiration diagnosis of angiomyolipoma of the liver using a cell block with immunohistochemical study. A case report. Acta Cytol 38:257- 260, 1994. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 224 EP 226 PG 3 ER PT J AU Sergi, C Bohler, Th Schonrich, G Sieverts, H Roth, US Debatin, KM Herwart, FO AF Sergi, Consolato Bohler, Thomas Schonrich, Gunther Sieverts, Hauke Roth, U Stephanie Debatin, Klaus-Michael Herwart, F Otto TI Occult Thyroid Pathology in a Child with Acquired Immunodeficiency Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE HIV; AaS; childhood; thyroid gland; pathology ID HIV; AaS; childhood; thyroid gland; pathology AB A 11-year-old boy with acquired immunodeficiency syndrome (AaS), Varicella-zoster virus (VZV) infection and long-term antiviral treatment suffered from a disorder of contractility of the left ventricle of the heart. Following severe unmanageable vomiting, the patient died and the postmortem examination showed marked involution of the lymphatic system, multiple foci of fibrosis of both ventricles of the heart, and regressive changes of the thyroid gland. Biochemical values of the thyroid gland function were, however, not altered. Neither human immunodeficiency virus-related p24 antigen, nor VZV DNA sequences were found in the thyroid gland. Regressive changes of the thyroid gland can probably occur before its function fails. By analyzing the possible etiologies, the endocrine toxicity of a long-term antiviral treatment should be taken into account. C1 [Sergi, Consolato] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. [Bohler, Thomas] University of Ulm, Department of PediatricsUlm, Germany. [Schonrich, Gunther] University of Heidelberg, Institute of Hygiene and MicrobiologyHeidelberg, Germany. [Sieverts, Hauke] University of Heidelberg, Department of PediatricsHeidelberg, Germany. [Roth, U Stephanie] University of Heidelberg, Department of NeuropathologyHeidelberg, Germany. [Debatin, Klaus-Michael] University of Ulm, Department of PediatricsUlm, Germany. [Herwart, F Otto] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. RP Sergi, C (reprint author), University of Heidelberg, Department of Pathology, D-69120 Heidelberg, Germany. EM Consolato_Sergi@med.uni-heidelberg.de CR Bakshi SS, Britto P, Capparelli E, et al: Evaluation of pharmacokinetics, safety, tolerance, and activity of combinadon of zalcitabine and zidovudine in stable, zidovudinetreated pediatric patients with human immunodeficiency virus infection. AIDS Clinical Trials Group Protocol 190 Team. J Infect Dis 175:1039- 1050, 1997. Blumberg BS, Fox RC: The Daedalus effect: changes in ethical questions relating to hepatitis B virus. Ann Intern Med 102:390-394, 1985 Butler KM, Venzon D, Henry N, et al: Pancreatitis in human immunodeficiency virus-infected children receiving dideoxyinosine. Pediatrics 91:747-751, 1993. Callea F, Fabbretti G, Brisigotti M, et al Valutazione della patologia da farmaci nel bambino. In: „Farmaci e Bambino. Terapia pediatrica essenziale e principi di farmacologia“. Durand P & Cornaglia-Ferraris P, eds). Casa Editrice Ambrosiana, Milano, 1993, pp. 23-33. Cohen J: AIDS therapy. Failure isn’t what it used to be...but neither is success. Science 279:1133-1134, 1998. Desai G, Islam R: The changing pattern of surgical pathology of the thyroid gland in Zambia. Cent Afr J Med 38:240-242, 1992. Domanski MJ, Sloas MM, Follmann DA, et al: Effect of zidovudine and didanosine treatment on heart function in children infetted with human immunodeficiency virus. J Pediatr 127:137-146, 1995. Edelman M, Birkenhauer MC, Steinberg JJ, et al: Microglial nodule encephalitis: limited CNS infection despite disseminated systemic cryptococcosis. Clin Neuropathol 15:30-33, 1996. Englund JA, Baker CJ, Raskino C, et al: Zidovudine, didanosine, or both as the initial treatment for symptomatic HIVinfected children. AIDS Clinical Trials Group, ACTG, Study 152 Team. N Engl J Med 336:1704-1712, 1997. Hannet I, Erkeller-Yuksel F, Lydyard P, et al: Developmental and maturational changes in human blood lymphocyte subpopulations. Immunol Today 13:215-218, 1992. Hirschfeld S: Use of human.recombinant growth hormone and human recombinant insulin-like growth factor-I in patients with human immunodeficiency virus infection. Horm Res 46:215- 221, 1996. Hirschfeld S, Laue L, Cutler GB Jr, et al: Thyroid abnormalities in children infected with human immunodeficiency virus. J Pediatr 128:70-74, 1996. Hoernle EH, Reid TE: Human immunodeficiency virus infection in children. Am J Health Syst Pharm 52:961-979, 1995. Hsu SM, Raine L, Fanger H: Use of avidin-biotin-peroxidase complex, ABC, in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody, PAP, procedures. J Histochem Cytochem 29:577-580, 1981. Joshi VV: Pathology of childhood AIDS. Pediatr Clin North Am 38:97-120, 1991. Kamali F: Clinical pharmacology of zidovudine and other 2’;3’- dideoxynucleoside analogues. Clin Investig 71:392-405, 1993. Keyhani-Rofagha S, Piquero C: Pneumocystis carinii thyroiditis diagnosis by fme needle aspiration cytology: a case report. Acta Cytol 40:307-310, 1996. Lacaille F, Ortigao MB, Debre M, et al: Hepatic toxicity associated with 2’-3’ dideoxyinosine in children with AIDS. J Pediatr Gastroenterol Nutr 20:287-290, 1995. Levin TL, Berdon WE, Tang HB, et al: Dideoxyinosine-induced pancreatitis in human immunodeficiency virus-infected children. Pediatr Radiol 27:189-191, 1997. Luzuriaga K, Bryson Y, McSherry G, et al: Pharmacokinetics, safety, and activity of nevirapine in human immunodeficiency virus type 1-infected children. J Infect Dis 174:713-721, 1996. Matarazzo P, Palomba E, Lala R, et al: Tovo PA: Growth impairment, IGF I hyposecretion and thyroid dysfunction in children with perinatal HIV-1 infection. Acta Paediatr 83:1029-1034, 1994. Mueller BU, Butler KM, Stocker VL, et al: Clinical and pharmacokinetic evaluation of long-term therapy with didanosine in children with HIV infection. Pediatrics 94:724-731, 1994. Pizzo PA, Butler K, Balis F, et al: Dideoxycytidine alone and in an alternating schedule with zidovudine in children with symptomatic human immunodeficiency virus infection. J Pediatr 117:799-808, 1990. Puchhammer-Stockl E: PCR detection of varicella zoster virus. In: Persing DH, Snith TF, Tenover FC, White TJ, eds). Diagnostic molecular microbiology. Principles and applications. American Society for Microbiology, Washington, 1993, pp. 356-360. Schwartz LJ, St Louis Y, Wu R, et al: P: Endocrine function in children with human immunodeficiency virus infection. Am J Dis Child 145:330-333, 1991. Sergi C, Bohler T, Schonrich G, et al: Pediatric Acquired Immunodeficiency Syndrome, PAIDS, as viewed by the pathologist and the immunologist: up-dated review. Pediatr Grenzgeb, Pediatrics and related topics, 37:271-332, 1998. Whitcup SM, Dastgheib K, Nussenblatt RB, et al: A clinicopathologic report of the retinal lesions associated with didanosine. Arch Ophthalmol 112:1594-1598, 1994. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 227 EP 232 PG 6 ER PT J AU Szekely, E Kulka, J Miklos, I Kaliszky, P AF Szekely, Eszter Kulka, Janina Miklos, Imre Kaliszky, Peter TI Leiomyosarcomas of Great Vessels SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE leiomyosarcoma; thoracic aorta; aortic dissection; inferior vena cava; retroperitoneal tumor ID leiomyosarcoma; thoracic aorta; aortic dissection; inferior vena cava; retroperitoneal tumor AB Sarcomas of the great vessels are rare. Altogether 400 such cases have been described in the aorta, the pulmonary artery, and inferior vena cava. The clinical symptoms are generally related to embolic phenomena, aneurysm formation, and widespread metastases, especially to bones. With improved diagnostic modalities more cases are diagnosed and treated surgically. Resection of the tumor may prolong the patient’s life. In this paper authors present two cases of such rare sarcomas. In our first case a tumor has developed in the thoracic aorta with symptoms of imminent aortic dissection. The tumorous nature of the lesion was revealed only histologically, since neither the operation, nor macroscopic picture gave any clue to its tumorous nature. The second case was a male patient with a huge retroperitoneal tumor arising from the inferior vena cava, which was clinically suspected to be a carcinomaarising in the adrenal gland. C1 [Szekely, Eszter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Miklos, Imre] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Kaliszky, Peter] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. RP Szekely, E (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM szesz@korb2.sote.hu CR Neri E, Miracco C, Luzi P: Intimal type primary sarcoma of the thoracic aorta presenting as a saccular false aneurysm: report of a case with evidence of rhabdomyosarcomatous differentiation J Thorac Cardiovasc Surg. 118:371-372, 1999. Miracco C, Laurini L, Santopietro R: Intimal type primary sarcoma of the aorta. Report of a case with evidence of rhabdomyosarcomatous differentiation Virchows Arch. 435:62-66, 1999. Patel KP, Niazi TBM, Griffiths AP: Massive osteolytic bone metastases from a primary aortic sarcoma: a case report. Hum Pathol 28:1306-1310, 1997. Burke AP, Virmani R: Sarcomas of the Great Vessels. A Clinicopathologic study. Cancer 71:1761-1773, 1993. Burke AP, Virmani R: Tumors of the heart and great vessels, AFIP 1996. Roncalori F, Eusebi V: Rhabdomyoblastic differentiation in a leiomyosarcoma of the retroperitoneum Hum Path 27:310-313, 1996. Suzuki M, Shimoda T, Ushigome S: Leiomyosarcoma of the thoracic aorta associated with dissecting aneurysm Acta Pathol Jpn 39:336-341, 1989. Shimoda H, Oka K, Otani S: Vascular leiomyosarcoma arising from the inferior vena cava diagnosed by intraluminal biopsy. Virchows Arch 433:97-100, 1998. Mingoli A, Cavallaro A, Sapienza P: International registry of inferior vena cava leiomyosarcoma: analysis of a world series of 218 patients. Anticancer Res 16:3201-3205, 1996. Glock Y, Laghzaoui A, Wang J: Fissured leiomyosarcoma of the descending thoracic aorta. A propos of a case and review of the literature. Arch Mal Coeur 90:1317-1320, 1997. Majeski J, Crawford ES, Majeski EI: Primary aortic intimal sarcoma of the endothelial cell type with long term survival. J Vasc Surg 27:555-558, 1998. Rochais JP, Icard P, Coffin O: Intimal sarcoma of the thoracic aorta: a case report. Eur J Vasc Endovasc Surg 18:181-182, 1999. Nanjo H, Murakami M, Ebina T: Aortic intimal sarcoma with acute myocardial infarction. Pathol Int 46:673-681, 1996. Raaf HN, Raaf JH: Sarcomas related to the heart and vasculature. Semin Surg Oncol 10:374-382, 1994. Tejada E, Becker GJ, Waller BF: Malignant myxoid emboli as the presenting feature of primary sarcoma of the aorta, myxoid malignant fibrous histiocytoma): a case report and review of the literature. Clin Cardiol 14:425-430, 1991. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2000 VL 6 IS 3 BP 233 EP 236 PG 4 ER PT J AU Rosendaal, M Krenacs, T AF Rosendaal, Martin Krenacs, Tibor TI Regulatory Pathways in Blood-forming Tissue with Particular Reference to Gap Junctional Communication SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Direct cell-cell communication; gap junctions; connexin 43; growth factors; regulation of haemopoesis; haemopoietic microenvironment ID Direct cell-cell communication; gap junctions; connexin 43; growth factors; regulation of haemopoesis; haemopoietic microenvironment AB Blood formation by pluripotent stem cells and their progeny is thought to be regulated by receptor-ligand interactions between cell-substrate, cell-cell and cell-matrix in the bone marrow. Primitive stem cells form progenitors and, in their turn, these give rise to haemopoietic progeny which are more specifically committed in that they can form progressively fewer types of blood cells. Recently we have estab-lished that direct cell-cell communication via gap junctions may be part of this regulatory system. Con-nexin43 gap junctions metabolically couple the three dimensional meshwork of bone marrow stromal cells to form a functional syncytium in which some blood-forming cells are also coupled. The expression of gap junctions in the bone marrow is markedly upregulated when there is an urgent and substantial demand for blood-formation; for example, following cytotoxic injury after 5-fluorouracil or irradiation; or during neonatal blood-formation and in the epiphysis of growing bones. Chemical blockade of gap junctions blocks blood-formation in long-term cultures but is reversible after the blockade has been relieved. This short review highlights briefly the known regulatory mechanisms of blood-formation with especial attention to gap junctional communication. C1 [Rosendaal, Martin] University College London, Department of Anatomy and Developmental Biology, Gower Street, WC1E 6BT London, UK. [Krenacs, Tibor] University of Szeged, Department of PathologySzeged, Hungary. RP Rosendaal, M (reprint author), University College London, Department of Anatomy and Developmental Biology, WC1E 6BT London, UK. EM m.rosendaal@ucl.ac.uk CR Boocock CA, Jones GE, Stanley ER, et al: Colony-stimulating factor-1 induces rapid behavioural responses in the mouse macrophage cell line, BAC1.2F5. J Cell Sci 93:447-456, 1989. Bradley TR, Metcalf D: The growth of mouse bone marrow cells in vitro. Aust J Exp Biol Med Sci 44:287-299, 1966. Bradley TR, Hodgson GS: Detection of primitive macrophage progenitor cells in mouse bone marrow. Blood 54:1446-1450, 1979. Brookes M: The blood supply of bone. Butterworths, London, 1971. Cancelas JA, Koevoet WL, de Koning AE, et al: Connexin-43 gap junctions are involved in multiconnexin-expressing stromal support of hemopoietic progenitors and stem cells. Blood 96:498-505, 2000. Copeland NG, Gilbert DJ, Cho BC, et al: Mast cell growth factor maps near the steel locus on mouse chromosome 10 and is deleted in a number of steel alleles. Cell 63:175-183, 1990. Coulombel L, Vuillet MH, Leroy C, et al: Lineage- and stagespecific adhesion of human hematopoietic progenitor cells to extracellular matrices from marrow fibroblasts. Blood 71:329- 334, 1988. Dexter TM, Allen TD, Lajtha LG: Conditions controlling the proliferation of haemopoietic stem cells in vitro. J Cell Physiol 91:335-344, 1977. Dorshkind K, Green L, Godwin A, et al: Connexin-43-type gap junctions mediate communication between bone marrow stromal cells. Blood 82:38-45, 1993. Dranoff G, Mulligan RC: Activities of granulocyte-macrophage colony-stimulating factor revealed by gene transfer and gene knockout studies. Stem Cells Dayt 1:173-182, 1994. Drzeniek Z, Stocker G, Siebertz B, et al: Heparan sulfate proteoglycan expression is induced during early erythroid differentiation of multipotent hematopoietic stem cells. Blood 93:2884-2897, 1999. Gauthier L, Fougereau M, Tonnelle C: Construction of temperature and Zn-dependent human stromal cell lines that amplify hematopoietic precursors from cord blood CD34+ cells. Exp Hematol 26:534-540, 1998 Gordon MY, Riley GP, Watt SM, et al: Compartmentalization of a haematopoietic growth factor, GM-CSF, by glycosaminoglycans in the bone marrow microenvironment. Nature 326:403- 405, 1987. Green CR, Severs NJ: Robert Feulgen Prize Lecture. Distribution and role of gap junctions in normal myocardium and human ischaemic heart disease. Histochemistry 99:105-120, 1993. Gupta P, Oegema TJ, Brazil JJ, et al: Structurally specific heparan sulfates support primitive human hematopoiesis by formation of a multimolecular stem cell niche. Blood 92:4641- 4651, 1998. Hodgson GS, Bradley TR, Radley JM: The organization of hemopoietic tissue as inferred from the effects of 5-fluorouracil. Exp Hematol 10:26-35, 1982. Jandl JH: Blood. Little Brown, Boston, 1996. Krenacs T, Rosendaal M: Immunohistological detection of gap junctions in human lymphoid tissue: connexin43 in follicular dendritic and lymphoendothelial cells. J Histochem Cytochem 43:1125-1137, 1995. Krenacs T, van Dartel M, Lindhout E, et al: Direct cell/cell communication in the lymphoid germinal center: connexin43 gap junctions functionally couple follicular dendritic cells to each other and to B lymphocytes. Eur J Immunol 27:1489-1497, 1997. Krenacs T, Rosendaal M: Connexin43 gap junctions in normal, regenerating, and cultured mouse bone marrow and in human leukemias. Am J Pathol 152:993-1004, 1998. Kumar NM, Gilula NB: The gap junction communication channel. Cell 84:381-388, 1996. Levesque JP, Leavesley DI, Niutta S, et al: Cytokines increase human hemopoietic cell adhesiveness by activation of very late antigen, VLA)-4 and VLA-5 integrins. J Exp Med 181:1805- 1815, 1995. Levesque JP, Haylock DN, Simmons PJ: Cytokine regulation of proliferation and cell adhesion are correlated events in human CD34+ hemopoietic progenitors. Blood 88:1168-1176, 1996 Lieschke GJ, Grail D, Hodgson G, et al: Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood 84:1737-1746, 1994. Lieschke GJ: CSF-deficient mice–what have they taught us? Ciba Found Symp 204:60-74, 1997. Metcalf D: The Florey Lecture, The colony-stimulating factors: discovery to clinical use. Philos Trans R Soc Lond B Biol Sci 333:147-173, 1991. Metcalf D: The hemopoietic regulators-an embarrassment of riches. Bioessays 14:799-805, 1992. Miyake K, Weissman IL, Greenberger JS, et al: Evidence for a role of the integrin VLA-4 in lympho-hemopoiesis. J Exp Med 173:599-607, 1991. Montecino RE, Leathers H, Dorshkind K: Expression of connexin 43, Cx43, is critical for normal hematopoiesis. Blood 96:917-24, 2000 Papayannopoulou T, Nakamoto B: Peripheralization of hemopoietic progenitors in primates treated with anti-VLA4 integrin. Proc Natl Acad Sci USA 90:9374-9378, 1993. Papayannopoulou T, Craddock C, Nakamoto B, et al: The VLA4/VCAM-1 adhesion pathway defines contrasting mechanisms of lodgement of transplanted murine hemopoietic progenitors between bone marrow and spleen. Proc Natl Acad Sci USA 92:9647-51, 1995. Papayannopoulou T, Priestley GV, Nakamoto B: Anti- VLA4/VCAM-1-induced mobilization requires cooperative signaling through the kit/mkit ligand pathway. Blood 91:2231- 2239, 1998. Ploemacher RE, van der Sluijs J, Voerman JS, Brons NH: An in vitro limiting-dilution assay of long-term repopulating hematopoietic stem cells in the mouse. Blood 74:2755-2763, 1989. Ploemacher RE, van der Loo J, van Buerden C, et al: Wheat germ agglutinin affinity of murine hemopoietic stem cell subpopulations is an inverse function of their long-term repopulating ability in vitro and in vivo. Leukemia 7:120-130, 1993. Pluznik DH, Sachs L: The cloning of normal „mast“ cells in tissue culture. J Cell Physiol 66:319-324, 1965. Reilly JT: Idiopathic myelofibrosis: pathogenesis, natural history and management. Blood Rev 11:233-242, 1997. Roberts R, Gallagher J, Spooncer E, et al: Heparan sulphate bound growth factors: a mechanism for stromal cell mediated haemopoiesis. Nature 332:376-378, 1988. Rosendaal M, Hodgson GS, Bradley TR: Haemopoietic stem cells are organised for use on the basis of their generation-age. Nature 264:68-69, 1976. Rosendaal M, Hodgson GS, Bradley TR: Organization of haemopoietic stem cells: the generation-age hypothesis. Cell Tissue Kinet 12:17-29, 1979. Rosendaal M, Adam J: Haemopoietic progenitors in different parts of one femur perform different functions during regeneration. Blood Cells 12:629-646, 1987. Rosendaal M, Gregan A, Green CR: Direct cell-cell communication in the blood-forming system. Tissue Cell 23:457-470, 1991. Rosendaal M, Green CR, Rahman A, et al: Up-regulation of the connexin43+ gap junction network in haemopoietic tissue before the growth of stem cells. J Cell Sci 107:29-37, 1994. Rosendaal M: Gap junctions in blood forming tissues. Microsc Res Tech 31:396-407, 1995 Rosendaal M, Mayen A, de Koning A, et al: Does transmembrane communication through gap junctions enable stem cells to overcome stromal inhibition? Leukemia 11:1281-1289, 1997. Ruscetti FW, Boggs DR, Torok BJ, et al: Reduced blood and marrow neutrophils and granulocytic colony-forming cells in S1/S1d mice. Proc Soc Exp Biol Med 152:398-402, 1976. Schofield R: The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells 4:7-25, 1978. Stanley ER, Bartocci A, Patinkin D, et al: Regulation of very primitive, multipotent, hemopoietic cells by hemopoietin-1. Cell 45:667-674, 1986. Stanley E, Lieschke GJ, Grail D, et al: Granulocyte/macrophage colony-stimulating factor-deficient mice show no major perturbation of hematopoiesis but develop a characteristic pulmonary pathology. Proc Natl Acad Sci USA 91:5592-5596, 1994. Strobel ES, Mobest D, von Kleist S, et al: Adhesion and migration are differentially regulated in hematopoietic progenitor cells by cytokines and extracellular matrix. Blood 90:3524- 3532, 1997. van der Loo J, Xiao X, McMillin D, et al: VLA-5 is expressed by mouse and human long-term repopulating hematopoietic cells and mediates adhesion to extracellular matrix protein fibronectin. J Clin Invest 102:1051-1061, 1998. Verfaillie CM, McCarthy JB, McGlave PB: Differentiation of primitive human multipotent hematopoietic progenitors into single lineage clonogenic progenitors is accompanied by alterations in their interaction with fibronectin. J Exp Med 174:693- 703, 1991. Watanabe Y: Fine structure of bone marrow stroma. Nippon Ketsueki Gakkai Zasshi 48:1688-1700, 1985. Weinstein R, Riordan MA, Wenc K, et al: Dual role of fibronectin in hematopoietic differentiation. Blood 73:111-1116, 1989 Weiss L: The hematopoietic microenvironment of the bone marrow: an ultrastructural study of the stroma in rats. Anat Rec 186:161-184, 1976. Williams DA, Rios M, Stephens C, et al: Fibronectin and VLA- 4 in haematopoietic stem cell-microenvironment interactions. Nature 352:438-441, 1991. Wilson JG: Adhesive interactions in hemopoiesis. Acta Haematol 97:6-12, 1997. Yong KL, Linch DC: Differential effects of granulocyte- and granulocyte-macrophage colony-stimulating factors, G- and GM-CSF, on neutrophil adhesion in vitro and in vivo. Eur J Haematol 49:251-259, 1992. Zhou YQ, Levesque JP, Hatzfeld A, et al: Fibrinogen potentiates the effect of interleukin-3 on early human hematopoietic progenitors. Blood 82:800-6, 1993. Zsebo KM, Wypych J, McNiece IK, et al: Identification, purification, and biological characterization of hematopoietic stem cell factor from buffalo rat liver-conditioned medium. Cell 63:195-201, 1990. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 243 EP 249 PG 7 ER PT J AU Sergi, C Kahl, P Herwart, FO AF Sergi, Consolato Kahl, Philip Herwart, F Otto TI Immunohistochemical Localization of Transforming Growth Factor-a and Epithelial Growth Factor Receptor in Human Fetal Developing Skin, Psoriasis and Restrictive Dermopathy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TGF-a; EGFR; developing skin; psoriasis; genodermatosis ID TGF-a; EGFR; developing skin; psoriasis; genodermatosis AB Keratinocytes release a number of cytokines interacting with other intra- and subepidermal cells during the initiation and the perpetuation of skin inflammatory reactions. Cultured human keratinocytes overexpressing the transforming growth factor alpha (TGF-alpha) assumed a spindled morphology and displayed increased locomotion. Moreover, the receptor for TGF-alpha, the epithelial growth factor receptor (EGFR), is important for autocrine growth, promotion of cell survival, and regulation of cell migration. The expression of TGF-alpha and EGFR has not been widely studied in human developing skin and their roles in geno-dermatosis are not known. In this study, we investigated the expression of TGF-alpha and EGFR by immunohistochemistry in human developing skin at different gestational ages (14 th week, 20 th week, and 34 th week), in six patients with psoriasis, and, for the first time, in an infant affected with restrictive dermopathy, a very rare lethal genodermatosis, characterized by abnormal skin growth and differentiation with thin, tightly adherent skin. TGF-alpha and EGFR were expressed in the basal layer at the 14 th week and in all epidermal layers at the 20 th and 34 th week of gestation. In psoriasis, TGF-alpha was overexpressed in all layers of epidermis, while EGFR was expressed in the basal and first suprabasal layers. In restrictive dermopathy, we observed no expression of both TGF-alpha and EGFR at the level of the skin. The other organs showed comparable patterns to those of an age-matched infant. In conclusion, TGF-alpha and EGFR interact strictly to promote skin development during the intrauterine life. An interactive autocrine growth cycle between TGF-alpha and EGFR is present in psoriasis. A skin-localized alteration of the expression of TGF-alpha and EGFR may be at the basis of restrictive dermopathy. The delay of growth and differentiation of the skin in restrictive dermopathy may be related to the absent expression of TGF-alpha, which is probably due to a down regulation of EGFR by an abnormal autocrine mechanism. C1 [Sergi, Consolato] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. [Kahl, Philip] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. [Herwart, F Otto] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, D-69120 Heidelberg, Germany. RP Sergi, C (reprint author), University of Heidelberg, Department of Pathology, D-69120 Heidelberg, Germany. EM Consolato_Sergi@med.uni-heidelberg.de CR Dean JC, Gray ES, Stewart KN et al: Restrictive dermopathy: a disorder of skin differentiation with abnormal integrin expression. Clin Genet 44:287-291, 1993. Higashiyama M, Hashimoto K, Matsumoto K, et al: Differential expression of transforming growth factor-alpha, TGF-alpha, and EGF receptor in transitional area of psoriatic epidermis. J Dermatol Sci 7:45-53, 1994. Holbrook KA, Dale BA, Witt DR, et al: Arrested epidermal morphogenesis in three newborn infants with a fatal genetic disorder, restrictive dermopathy). J Invest Dermatol 88:330-339, 1987. Hudson LG, McCawley LJ: Contributions of the epidermal growth factor receptor to keratinocyte motility. Microsc Res Tech 43:444-455, 1998. Ju WD, Schiller JT, Kazempour MK, et al: TGF alpha enhances locomotion of cultured human keratinocytes. J Invest Dermatol 100:628-632, 1993. Mau U, Kendziorra H, Kaiser P, et al: Restrictive dermopathy: report and review. Am J Med Genet 71:179-185, 1997. McKusick VA: Online Mendelian inheritance in man, OMIM). http//www3.ncbi.nlm.nih.gov/omim/ Mead JE, Fausto N: Transforming growth factor alpha may be a physiological regulator of liver regeneration by means of an autocrine mechanism. Proc Natl Acad Sci U S A 86:1558-1562, 1989. Nanney LB, Stoscheck CM, King LE Jr, et al: Immunolocalization of epidermal growth factor receptors in normal developing human skin. J Invest Dermatol 94:742-748, 1990. Sandgren EP, Luetteke NC, Palmiter RD, et al: Overexpression of TGF alpha in transgenic mice: induction of epithelial hyperplasia, pancreatic metaplasia, and carcinoma of the breast. Cell 61:1121-1135, 1990. Sergi C, Kahl P, Otto HF: Contribution of apoptosis and apoptosis- related proteins to the malformation of the primitive intrahepatic biliary system in Meckel syndrome. Am J Pathol 156:1589-1598, 2000. Sergi C, Schmitt HP: The vesicular forebrain, pseudo-aprosencephaly): a missing link in the teratogenetic spectrum of the defective brain anlage and its discrimination from aprosencephaly. Acta Neuropathol 99:277-284, 2000. Sillevis-Smitt JH, van Asperen CJ, Niessen CM, et al: Restrictive dermopathy. Report of 12 cases. Dutch Task Force on Genodermatology. Arch Dermatol 134:577-579, 1998. Witt DR, Hayden MR, Holbrook KA, et al: Restrictive dermopathy: a newly recognized autosomal recessive skin dysplasia. Am J Med Genet 24:631-648, 1986. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 250 EP 255 PG 6 ER PT J AU Rehman, Sh Crow, J Revell, AP AF Rehman, Shazza Crow, Julie Revell, A Peter TI Bax Protein Expression in DCIS of the Breast in Relation to Invasive Ductal Carcinoma and Other Molecular Markers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; ductal carcinoma in situ; Bax; Ki67; oestrogen receptor; p53; cErbB2; Bcl2 ID breast cancer; ductal carcinoma in situ; Bax; Ki67; oestrogen receptor; p53; cErbB2; Bcl2 AB This study describes the incidence of Bax protein expression in a series of 106 cases of breast cancer including 56 cases of ductal carcinoma in situ (DCIS) and 50 cases of invasive ductal carcinoma (IDC). Relationships of Bax expression to the histological grades of DCIS & IDC, and to the expression of Ki67, ER, p53, cerbB2 & Bcl2 are described. The expression of Bax, Ki67, ER, p53, cerbB2 and Bcl2 proteins is determined immunohistochemically. Cases were regarded positive for Bax, Bcl2 and cerbB2 when they showed either moderate or strong staining for these markers. The nuclear stains (Ki67, ER, and p53) were quantified in terms of percentage positive cells and cases for ER and p53 were considered positive when more than 10% cells were labelled. DCIS were graded histologically as well (n=18), intermediately (n=18), and poorly differentiated (n=20) Invasive ductal carcinoma was graded as grade I (well-differentiated) n=7, grade II (intermediate) n=24 and grade III (poorly differentiated) n=19. 65/106 cases (61%) were Bax positive including 37/56 (66%) of DCIS and 28/50 (56%) of IDC. Bax expression did not correlate to increasing histological grades of either DCIS or IDC. It did not correlate to Ki67, ER, p53 or cerbB2 but positive correlation was seen with Bcl2 (p=0.003). Bcl2 immunostaining displayed a negative correlation with increasing histological grades both of DCIS and IDC (p=0.026), (p=0.041) respectively. There was a trend of negative correlation of Bcl2 with Ki67 (p=0.062). It correlated positively with Bax (p=0.003) and ER (p<0.0001). Results suggest that the regulation of apoptosis is important in ductal carcinoma in situ of the breast as well as invasive ductal carcinomas. Bcl2 is associated with good prognostic markers in both DCIS and IDC, whereas the regulation of Bax is complex and does not necessarily correlate with mutant p53. C1 [Rehman, Shazza] University College London Medical School, Department of Histopathology, Row-land Hill Street, NW3 2PF London, UK. [Crow, Julie] University College London Medical School, Department of Histopathology, Row-land Hill Street, NW3 2PF London, UK. [Revell, A Peter] University College London Medical School, Department of Histopathology, Row-land Hill Street, NW3 2PF London, UK. RP Rehman, Sh (reprint author), University College London Medical School, Department of Histopathology, NW3 2PF London, UK. EM shazzarehman@doctors.org.uk CR Oltvai ZN, Milliman CL, Korsmeyer SJ: Bcl2 heterodinerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Structure-function analysis of Blc2 protein. Cell 74:609-619, 1993. Hanada M, Aime-Sempe C, Sato T, et al: Identification of conserved domains important for homodiverization with Bcl2 and heterodiverization with Bax. J Biol Chem 270:11962-11969, 1995. Bargou RC, Daniel PT, Mapara MY: Expression of the bcl-2 gene family in normal and malignant breast tissue: low baxalpha expression in tumor cells correlates with resistance towards apoptosis, Int J Cancer 60:854-859, 1995. Korsmeyer SJ, Shutter JR, Veis DJ, et al: Bcl-2/Bax: a rheostat that regulates an anti-oxidant pathway and cell death. Semin Cancer Biol 46:327-332, 1993. Miyashita T, Reed JC: Tumour suppressor p53 is a direct transcriptonal activator of the human Bax gene. Cell 80:293-299, 1995. Kapranos N, Karaiosifidi H, Valavanis C, et al: Prognostic significance of apoptosis relate1. d proteins Bcl-2 and Bax in node-negative breast cancer patients, Anticancer Research 17:2499-505, 1997. Tsujimoto Y, Cossman J, Jaffe E, et al: Involvement of the Bcl2 gene in human folicular lymphoma. Science 228:1440-1443, 1985. Hockenberg DM, Zutter M, Hickey W, et al: Bcl2 protein is topgraphically restricted in tissues characterised by apoptotic cell death. Proc Natl Acad Sci 88:6961-6965, 1991. Nathan B, Anbazhagan R, Dyer M, et al: Expression of Bcl2 like immunoreactivity in the normal breast and in breast cancer. The Breast 2:134-137, 1993. Doglioni C, DeiTos AP, Laurino L, et al: The prevalence of Bcl2 immunoreactivity in breast carcinomas and its clinicopatholigical correlates, with particular reference to oestrogen receptor status. Virchous Arch 424:47-51, 1994. Gee JM, Robertson JF, Ellis IO: Immunocytochemical localization of Bcl2 protein in human breast cancers and its relationship to a series of prognostic markers and response to endocrine therapy. Int J Cancer 59:619-628, 1994 Silvestrini R, Veneroni S, Daidone MG: The Bcl2 protein: a prognostic indicator strongly related to p53 protein in lymphnode negative breast cancer patients. J Natl Cancer Inst 86:499- 504, 1994. Lipponen P, Pietilainen T, Kosma VM, et al: Apoptosis suppressing protein Bcl2 is expressed in well differentiated breast carcinomas with favourable prognosis. J Pathol 177:49-55, 1995. Lane DP: p53, guardian of the genome. Nature 358:15-16, 1992. Levine AL, Momand J, Finlay CA: The p53 tumour suppression gene. Nature 351:453-456, 1991. Hollestein M, Sidransky D, Vogelstein B, et al:p53 mutations in human cancer. Science 253: 49-53, 1991 Halder S, Negrini M, Moune M, et al: Down regulation of Bcl2 by p53 in breast cancer cells. Cancer Res. 54:2095-2097, 1994 Kaklamanis L, Savage A, Mortensen N, et al: Early expression of Bcl2 protein in the adenoma-carcinoma sequence of colorectal neoplasia. J Pathol 179:10-14, 1996. Shiina H, Igawa M, Urakami S, et al: Immunohistochemical analysis of Bcl2 expression in transitional cell carcinoma of the bladder. J Clin Palliol. 49:395-399, 1996 Thor AD, Moore DH II, Edgerton SM, et al: Accumulation of p53 tumour suppressor gene protein: An independent marker of prognosis in breast cancers. J. Natl Cancer Inst 84:845-855, 1992. Coussens L, Yang-Feng TL, Liao YC, et al: Tyrosine kinase receptor with exclusive homology to EGF receptor shares chromosomal location with neu oncogene. Science 230:1132-1139, 1985. Wolber RA, Dupuis, BA, Wick MR: Expression of cerbB2 oncoprotein in mammary and extramammary Pagets’ disease. Am J Clin Pathol 96:243-247, 1991. Yamamoto T, Ikawa S, Akiyama T, et al: Similarity of protein encoded by the human cerbB2 gene to epidermal growth factor receptor. Nature 319:230-234, 1986. Allred DC, O’Connell P, Fugua AW: Biomarkers in early breast neoplasia. J. Cell Biochem 17:125-131, 1993. Barnes DM: cerbB2 amplification in mammary carcinoma. J Cell Biochem 17:132-138, 1993. Perren TJ: cErbB2 oncogene as a prognostic marker in breast cancer. Br J. Cancer 63:328-332, 1991. Thompson AM: Lemoine N, Neoptolemos J, Cooke T, eds. Cancer, a molecular approach. Breast Cancer. Oxford: Blackwells 163-183, 1994. Slamon DJ, Clark GM, Wong SG, et al: Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu encogene. Science 235:177-182, 1987. Gerdes J, Schwab U, Lemke H, et al: Production of a mouse monclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer 31:13-20, 1983 Nicholson RI, McClelland RA, Finlay P, et al: Relationship between EGFR, cerbB2 protein expression and ki67 immunostaining in breast cancer and hormone sensitivity. Eur J Cancer 29A:1018-1023, 1993 Bouzubar N, Walker KJ, Griffiths K, et al: Ki67 immunostaining in primary breast cancer: pathological and clinical associations. Br J Cancer 59:943-947, 1989. Pinder SE, Wencyk P, Sibbering DM, et al: Assessment of the new proliferation marker MIBI in breast carcinoma using image analysis; associations with other prognostic factors and survival. Br J Cancer 71:146-149, 1995. Locker AP, Birrell K, Bell JA, et al: Ki67 immunoreactivity in breast carcinoma: relationship to prognostic variables and short term survival. Eur J Surg Oncol 1992; 18:224-229, 1992. Nicholson RI, Bouzubar N, Walker KJ, Et al: Hormone sensitivity in breast cancer: influence of heterogeneity of oestrogen receptor expression and cell proliferation. Eur J Cancer 27:908- 913, 1991 Thorpe SM, Rose C, Rasmussen BB, et al: Prognostic value of steroid hormone receptor multivariate analysis of systemically untreated patients with node negative primary breast cancer. Cancer Res 47:6126-6133, 1987. Kiang DT, Frenning DH, Goldman AL, et al: Estrogen receptors and responses to chemotherapy and hormonal therapy in advanced breast cancer. N Engl J Med 299:1330-1333, 1978. Anderson J, Thorp SM, King WJ, et al: The prognostic value of immunohistochemical oestrogen receptor analysis in paraffin embedded and frozen sections versus that of steroid binding assays. Eur J Cancer 26:442-449, 1990. Elledge RM, Green S, Howes L, et al: Bcl2, p53 and response to lamonifen in oestrogen receptor positive metastatic breast cancer: a southwest oncology group study. J Clin Oncol 15:1916- 1922, 1997. Holland R, Peterse JL, Millis RR, et al: Ductal carcinoma in situ: A proposal for a new classification. Seminars in Diagnostic Pathology. 11:167-180, 1994. Elston CW, Ellis IO: Pathological prognostic factors in breast cancer. The value of histological grade in breast cancer: experience from a large study with long term follow up. Histopathology; 19:403-410, 1991 Vilain MO, Delobelle-Deroide A, Bloget F, et al: Immunohistochemical detection of oestrogen and progesterone receptors in formation fixed, paraffin embedded tissues after microwave treatment. Comparison with biochemical assay in a series of 123 breast carcinomas with determination of the positivity cut off. Annales de Pathologie. 17:82-88, 1997. Krajewski S, Krajewska M, Shabaik A, et al: Immunohistochemical determination of in vivo distribution of Bax, a dominant inhibitor of Bcl2. Am J Pathol; 145:1323-1336, 1994. World Health Organisation,, 1981, Histological typing of breast tumours. International Histological Classification of Tumours, 2nd Edition World Health Organisation, Geneva. Reed JC: Bcl2 and the regulation of programmed cell death. J Cell Biol 124:1-6, 1994. Hockenberg DM: Bcl2 in cancer, development and apoptosis. J Cell Sci 18:51-55, 1994. Kerr JF, Winterford CM, Harmon BV: Apoptosis – its significance in cancer and cancer therapy. Cancer 73:2013-2026, 1994. White E: Life, death and the pursuit of apoptosis. A review. Genes and Development 10:1-15, 1996. Yang E, Korsmeyer SK: Molecular thanatopsis: a discourse on the Bcl2 family and cell death. Blood 88:386-401, 1996. Thompson CB: Apoptosis in the pathogensis and the treatment of disease. Science 267:1456-1462, 1995. Broise LH, Gottschalk AR, Quinfans J, Thompson CB: Bcl2 and Bcl2 related proteins in apoptosis regulation. Curr Top Microbiol Immunol 200:107-121, 1995. Craig RW: The Bcl2 gene family. Semin Cancer Biol 6:35-43, 1995. Krajewski S, Blomqvist C, Franssila K, et al: Reduced expression of proapoptotic gene BAX is associated with poor response rates to combination chemotherapy and shorter survival in women with metastatic breast adenocarcinoma. Cancer Res 55:4471-4478, 1995 Kapucuoglu N, Losi L, Eusebi V: Immunohistochemical localization of Bcl-2 and Bax proteins in in situ and invasive duct breast carcinomas. Virchows Arch 430:17-22, 1997 Yang Q, Sakurai T, Jing X, et al: Expression of Bcl2, but not Bax, correlates with estrogen receptor status and tumour proliferation in invasive breast carcinoma, Pathology International, 49;775-780, 1999. Rochaix P, Krajewski S, Reed JC, et al: In vivo patterns of Bcl2 family protein expression in breast carcinoma in relation to apoptosis. J Pathol 87:410-415, 1999. Van Slooten HJ, Clahsen PC, Van Dierendonck JH: Expression of Bcl2 in node negative breast cancer is associated with various prognostic factors, but does not predict response to one course of peri-operative chemotherapy, Br J Cancer 74:78-85, 1996. Joensuu H, Pylkkanen L, Toikkanen S: Bcl2 protein expression and long term survival in breast cancer, Amer J Pathol 145:1191-1198, 1994. Siziopikou KP, Prioleau JE, Harris JR, et al: Bcl2 expression in the spectrum of pre-invasive breast lesions. Cancer 77:499- 506, 1996 Quinn CM, Ostrowski JL, Harkins L, et al: Loss of Bcl2 expression in DCIS of the breast relates to poor histological differentiation and to expression of p53 and cerbB2 proteins. Histopathology 3:531-536, 1998. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 256 EP 263 PG 8 ER PT J AU Lazaris, ChA Chatzigianni, BE Paraskevakou, H Tseleni-Balafouta, S Davaris, SP AF Lazaris, Ch Andreas Chatzigianni, B Emmy Paraskevakou, Helen Tseleni-Balafouta, Sofia Davaris, S Panayiotis TI Lectin Histochemistry as a Predictor of Dysplasia Grade in Colorectal Adenomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal adenomas; lectin binding; phaseolous vulgaris leucoagglutinin; ulex europaeus; wheat germ agglutinin; peanut agglutinin ID colorectal adenomas; lectin binding; phaseolous vulgaris leucoagglutinin; ulex europaeus; wheat germ agglutinin; peanut agglutinin AB Lectins are sugar-binding proteins that bind to specific cellular carbohydrates, commonly affecting cellular physiology. Phaseolus vulgaris leucoagglutinin (PHA), ulex europaeus isoagglutinin-I (UEA-I), wheat germ agglutinin (WGA) and peanut agglutinin (PNA) are among the most well studied lectins in various tissues. The purpose of this study was to detect the above lectins’ binding sites and so examine alterations in glycoconjugate expression in neoplastic cells of 52 colorectal adenomas with various clinicopathologic characteristics and proliferation rates. Lectin histochemistry was performed in paraffin sections with and without neuraminidase treatment. Proliferative fraction was determined by immunolabelling for Proliferating Cell Nuclear Antigen. PHA was the more frequently positive lectin in the examined specimens; however, it was simultaneously detected in normal colonic mucosa and so was WGA. The frequency of high grade dysplasia was significantly greater in older patients and in samples with UEA-I positivity without neuraminidase pretreatment. UEA-I-reactive adenomas were generally characterized by high cell proliferation rates. A statistical model based on patients’ age and UEA-I binding without neuraminidase treatment can generally predict grade of dysplasia in 83% of adenomas and particularly high grade dysplasia in up to 93% of adenomas; so, such a model may be potentially useful for the early detection of neoplasia, for instance in exfoliative cells from the large intestine. C1 [Lazaris, Ch Andreas] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str., Goudi, GR-115 27 Athens, Greece. [Chatzigianni, B Emmy] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str., Goudi, GR-115 27 Athens, Greece. [Paraskevakou, Helen] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str., Goudi, GR-115 27 Athens, Greece. [Tseleni-Balafouta, Sofia] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str., Goudi, GR-115 27 Athens, Greece. [Davaris, S Panayiotis] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str., Goudi, GR-115 27 Athens, Greece. RP Lazaris, ChA (reprint author), University of Athens, Medical School, Department of Pathology, GR-115 27 Athens, Greece. EM alazaris@cc.uoa.gr CR Kinzler KW, Vogelstein B: Colorectal tumors. In: The Genetic Basis of Human Cancer., Eds: Vogelstein B and Kinzler KW, New York: McGraw- Hill, 1998, pp 565-587. Ryder SDD, Parker N, Ecclestone D, et al: Peanut lectin stimulates proliferation in colonic explants from patients with inflammatory bowel disease and colon polyps. Gastroenterology 106:117-124, 1994. Ohannesian DW, Lotan D, Thomas P, et al: Carcinoembryonic antigen and other glycoconjugates act as ligands for galectin-3 in human colon carcinoma cells. Cancer Research 55:2191- 2199, 1995. Mitchell BS, Schumacher U: The use of lectin Helix pomatia agglutinin, HPA, as a prognostic indicator and as a tool in cancer research. Histol Histopathol 14:217-226, 1999. Li WP, Zuber C, Heitz PU, Roth J: Cytochemical staining for b1,6 branching of asparagine-linked oligosaccharides in variants of metastatic human colon carcinoma cells. Am J Pathol 145:470-480, 1994. Rosai J: Ackerman’ s Surgical Pathology. Mosby, St. Louis, 1996. Kiss R, Camby I, Duckworth C, et al: In vitro influence of Phaseolous vulgaris, Griffonia simplicifolia, Concavalin A, Wheat germ and Peanut agglutinins on HTC-15, LoVo, and SW837 human colorectal cancer cell growth. Gut 40:253- 261,1997. Ryder SD, Smith JA, Rhodess JM: Peanut lectin: a mitogen for normal human colonic epithelium and human HT29 colorectal cancer cells. J Natl Cancer Inst 84:1410-1416, 1992. Jordinson M, El-Harity I, Calnan D, et al: Vicia faba agglutinin, the lectin present in broad beans, stimulates differentiation of undifferentiated colon cancer cells. Gut 44 :709-714, 1999. Said IT, Shamsuddin AM, Sherief MA, et al: Comparison of different techniques for detection of Gal-Gal N Ac, an early marker of colonic neoplasia. Histol Histopathol 14:351-357, 1999. Sivridis E, Agnantis N: The loss of lectin reactivity from human endometrium is a feature of malignant change. Path Res Pract 192:989-997, 1996. Lazaris ACh, Davaris P, Nakopoulou L, et al: Correlation between immunohistochemical expression of proliferating cell nuclear antigen and flow cytometry parameters in colorectal neoplasia. Dis Colon Rectum 37:1083-1089, 1994. Brink U, Bosbach R, Korabiowska M, et al: Histochemical study of expression of lectin-reactive carbohydrate epitopes and glycoligand-binding sites in normal human appendix vermiformis, colonic mucosa, acute appendicitis and colonic adenoma. Histol Histopathol 11:919-930,1996. Lance P, Leu R: Colonic oligosaccharide structures deduced from lectin-binding studies before and after desialylation. Hum Pathol 22:307-312, 1991. Desilets DJ, Davis KE, Nair PP, et al: Lectin binding to human colonocytes is predictive of colonic neoplasia. Am. J. Gastroenterol. 94: 744-750, 1999. Ryder SD, Smith JA, Rhodes EGH, et al: Proliferative responses of HT29 and Caco2 human colorectal cancer cells to a panel of lectins. Gastroenterology 106:85-93, 1994. Kellokumpu IH, Aandersoon LC, Kellokumpu SJ: Detection of colorectal neoplasia with Peanut-Agglutinin, PNA)-reactive carbohydrate structures in rectal mucus. Int J Cancer, Pred. Oncol., 74:648-653, 1997. Sams JS, Lynch HT, Burt RW, et al: Abnormalities in lectin histochemistry in familial polyposis coli and hereditary nonpolyposis colorectal cancer. Cancer 66:502-508,1990. Boland CR, Roberts JA: Quantitation of lectin binding sites in human colon mucins by use of peanut and wheat germ agglutinins. J Histochem Cytochem 36:1305-1307,1988. Drackenberg CB, Papadimitriou JC: Aberrant pattern of lectin binding in low and high grade prostatic intraepithelial neoplasia. Cancer 75:2539-2544, 1995. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 265 EP 271 PG 7 ER PT J AU Demirkan, CN Colakoglu, N Duzcan, E AF Demirkan, Calli Nese Colakoglu, Nagihan Duzcan, Ender TI Value of p53 Protein in Biological Behavior of Basal Cell Carcinoma and in Normal Epithelia Adjacent to Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ageing; basal cell carcinoma; p53 protein ID ageing; basal cell carcinoma; p53 protein AB Mutations in p53 gene are the most frequent gene alterations in human cancer. In this study, we have used the monoclonal antibody (DO7) to evaluate the role of the p53 gene mutation in the progression of basal cell carcinomas towards invasion. We tested the positivity for p53 protein in tumor cells in six cases of basosquamous cell carcinoma (BSCC), in twelve cases of infiltrative basal cell carcinoma (IBCC) and twenty-four cases of non-infiltrative basal cell carcinoma (NIBCC) in order to evaluate its potential prognostic significance. We also tested the expression of p53 protein in normal epithelia adjacent to carcinomas in order to determine its role in tumor progression. p53 protein staining with some peripheral accentuation was identified in 42,9% of all groups. No correlation was found between the immunreactivity of p53 protein and recurrence, pattern of tumor, diameter of the tumors and sex. However, there were statistically significant differences in positivity of p53 protein in normal epithelia adjacent to carcinomas and age of patients (t value: 2,21; p: 0,034). Results of the study suggest that the increasein p53 mutation frequency of morphologically normal epidermis was related to age and was independent of the degree of differentiation of BCC. C1 [Demirkan, Calli Nese] Pamukkale University, Department of Pathology, 269 sok. No: 46-8 Ucyol, 35280 Izmir, Turkey. [Colakoglu, Nagihan] Pamukkale University, Department of Pathology, 269 sok. No: 46-8 Ucyol, 35280 Izmir, Turkey. [Duzcan, Ender] Pamukkale University, Department of Pathology, 269 sok. No: 46-8 Ucyol, 35280 Izmir, Turkey. RP Demirkan, CN (reprint author), Pamukkale University, Department of Pathology, 35280 Izmir, Turkey. EM nesefahir@hotmail.com CR Barret TL, Smith KJ, Hodge JJ, et al: Immunohistochemical nuclear staining for p53, PCNA and Ki-67 in different histologic variants of basal cell carcinoma. J Am Acad Dermatol 37:430-437, 1997. D’Errico M, Calcagnile AS, Corona R, et al: p53 Mutations and chromosome instability in basal cell carcinoma s developed at an early or late age. Cancer Res 57:747-752, 1997. De Rosa G, Staibano S, Barra E, et al: p53 Protein in aggressive and non-aggressive basal cell carcinoma. J Cutan Pathol 20:429-434, 1993. Dixon AY, Lee SH, McGregor DH: Histologic evolution of basal cell carcinoma recurrence. Am J Dermatopathol 13:241,1991. Gunterson BA, Anbazhagan R, Warren W: Expression of p53 in premalignant and malignant squamous epithelium. Oncogene 6:1785-1789, 1986. Helander SD, Peters MS, Pittelkow MR: Expression of p53 protein in benign and malignant epidermal conditions. J Am Acad Dermatol 29:741-748, 1993. Jones MS, Helm KF, Maloney ME: The immunohistochemical characteristics of the basosquamous cell carcinoma. Dermatol Surg 23:181-184, 1997. Kirkham N: Tumors and cysts of the epidermis. In: Lever’s histophology of the skin, Eds: Elder D, Elenistas R, Jaworsky C, Johnson B). 1997, pp 719-731. Philadelphia New York, Lippincott- Raven. Liang SB, Ohtsuki Y, Furihata M, et al: Sun-exposure and ageing- dependent p53 protein accumulation results in growth advantage for tumour cells in carcinogenesis of nonmelanocytic skin cancer. Virchows Arch 434:193-199, 1999 Matsumura Y, Nishigor C, Yagi T, et al: Characterization of p53 gene mutations in basal-cell carcinomas: comparison between sun-exposed and less- exposed skin areas. Int J Cancer 65:778- 780, 1996. Onodera H, Nakamura S, Sugai T: Cell proliferation and p53 protein expressions in cutaneous epithelial neoplasms. Am J Dermatopathol 18:580-588, 1996. Ro YS, Cooper PN, Lee JA, et al: p53 protein expression in benign and malignant skin tumours. Br J Dermatol 128:237-241, 1993. Sexton M, Jones DB, Maloney ME: Histologic pattern analysis of basal cell carcinoma. J Am Acad Dermatol 23:1118-1126, 1990. Shea CR, McNutt NS, Volkenandt M, et al: Overexpression of p53 protein in basal cell carcinomas in human skin. Am J Pothol 141:25, 1992. Urano Y, Asano T, Yoshimoto K, et al: Frequent p53 accumulation in the chronically sun-exposed epidermis and clonal expansion of p53 mutant cells in the epidermis adjacent to basal cell carcinoma. J Invest Dermatol 104:928-932, 1995. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 272 EP 274 PG 3 ER PT J AU Nasierowska-Guttmejer, A Trzeciak, L Nowacki, PM Ostrowski, J AF Nasierowska-Guttmejer, Anna Trzeciak, Lech Nowacki, P Marek Ostrowski, Jerzy TI p53 Protein Accumulation and p53 Gene Mutation in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p53 protein; p53 gene; molecular biology; immunohistochemical studies; colorectal cancer ID p53 protein; p53 gene; molecular biology; immunohistochemical studies; colorectal cancer AB Comparison of immunohistochemical methods for detection of protein p53 accumulation and molecular techniques for analysis p53 gene mutation in colorectal cancer is presented. Thirty eight patients were included: all underwent surgery without preoperative treatment. Sex of patients, tumor localisa-tion, macro and microscopic type of cancer and staging according to Astler-Coller and Jass classifications were evaluated. Protein p53 accumulation was detected by the streptavidin-biotin method using DO-7 (Dako) antibody. The number of cells stained were classified semiquanititatively according to a scoring system: (–)no positive cells, (+) : 10-30% positive cells, (++) : 40-70% positive cells, (+++) : >70% positive cells. For all cancer samples, exons 5 to 9 of p53 gene were amplified from isolated genomic DNA. PCR products were subjected to single standed conformational polymorphism analysis. All product were also directly sequenced on ABI PRISM 377 apparatus using fluorescent dideoxyterminators chemistry. The protein p53 accumulation was detected in 53% (20/38), whereas p53 gene mutation was seen in 55% (21/38). Among them, 15 patients (39%) with overexpression showed mutation in exon 5-8 gene p53. Discrepancies between results were noted in 29%. In conclusion, the necessity of both methods – immunohistochemical and molecular – is indicated for the objective evaluation of functional and structural status of p53 gene and protein. C1 [Nasierowska-Guttmejer, Anna] Medical Center for Postgraduate Education, and Maria Sk³odowska-Curie Memorial Cancer Center and Institute of Oncology, Department of Pathology, Roentgena str. 5, 02-971 Warsaw, Poland. [Trzeciak, Lech] Medical Center for Postgraduate Education and Maria Sk³odowska-Curie Memorial Cancer Center and Institute of Oncology, Department of GastroenterologyWarsaw, Poland. [Nowacki, P Marek] Maria-Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Department of Colorectal CancerWarsaw, Poland. [Ostrowski, Jerzy] Medical Center for Postgraduate Education and Maria Sk³odowska-Curie Memorial Cancer Center and Institute of Oncology, Department of GastroenterologyWarsaw, Poland. RP Nasierowska-Guttmejer, A (reprint author), Medical Center for Postgraduate Education, and Maria Sk³odowska-Curie Memorial Cancer Center and Institute of Oncology, Department of Pathology, 02-971 Warsaw, Poland. CR Agarwall ML, Taylor WR, Chernov MV, et al: The p53 network. J Biol Chem 273:1-4, 1998. Bass IO, Mulder JWR, Offerhaus GJA, et al: An evaluation of six antibodies for immunohistochemistry of mutant p53 gene product in archival colorectal neoplasms. J Pathol 172:5-12, 1994. Bertorelle R, Esposito G, Del Mistro A, et al: Association of p53 gene and protein alterations with metastases in colorectal cancer. Am J Surg Pathol 19:463-471, 1995. Borresen DA, Lothe RA, Meling GI, et al: TP53 and long-term prognosis in colorectal cancer: mutation in the L3 zinc-binding domain predict poor survival. Clin Cancer Res 4:203-210, 1998. Bosari S and Viale G: The clinical significance of p53 aberrations in human tumors. Virchows Arch 427:229-241, 1995. Caldes T, Iniesta P, Vega FJ, et al: Comparative survival analysis of p53 gene mutation and protein accumulation in colorectal cancer. Oncology 55:249-257, 1998. Cripps KJ, Purdie CA, Carder PJ, et al: A study of stabilisation of p53 protein versus point mutation in colorectal carcinoma. Oncogene 9:2739-2743, 1994. Dix B, Robbins P, Corello S, et al: Comparison of p53 gene mutation and protein overexpression in colorectal cancer. Br J Cancer 70:585-590, 1994. El-Deiry WS, Kern SE, Pietenpol JA, et al: Definition of a consensus binding site for p53. Nature Genet 1:45-49, 1992. Goh HS, Yao J and Smith DR: p53 point mutation and survival in colorectal cancer patients. Cancer Res 55:5217-5221, 1995. Greenblatt MS, Bennett WP, Hollstein M, et al: Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res 54:4855-4878, 1994. Hall PA and Lane DP: p53 in tumor pathology: can we trust immunohistochemistry?-revisited! J Pathol 172:1-4, 1994. Hamelin R, Laurent PP and Olschwang S: Association of p53 mutations with short survival in colorectal cancer. Gastroenterology 106:42-48, 1994. Hasegawa H, Ueda M, Furukawa K, et al: p53 gene mutations in early colorectal carcinoma. de novo vs adenoma-carcinoma sequence. Int J Cancer 64:47-51, 1995. Hollstein M, Sidransky D, Vogelstein B, et al: p53 mutations in human cancers. Science 253:49-53,1991. Jass JR, Love SB, Northover JMA: A new classification of rectal cancer. Lancet i: 1303-1306, 1987. Jass J, Sobin WH: Histological typing of intestinal tumours. World Health Organisation. International Histological Classification of Tumours. Springer-Verlag, 1989. Kawasaki Y, Monden T, Morimoto H, et al: Immunohistochemical study of p53 expression in microwave-fixed, parrafinembedded sections of colorectal carcinoma and adenoma. Am J Clin Pathol 97:244-249, 1992. Lewine AJ: p53 , the cellular gatekeeper for growth and division. Cell 88: 23-331, 1997. Locker J: Tumor suppressor genes and the practice of surgical pathology. Hum Pathol 4:359-361, 1995. Nasierowska-Guttmejer A, Roszkowska-Purska K, Gil M, et al: Protein p53 accumulation and proliferative activity in adenomas and early carcinoma of the colorectum. Gastroenterologia Polska 5:341-347, 1998. Oren M, Maltzman W, Levine AJ: Posttranslational regulation of the 54K cellular tumor antigen in normal and transformed cells. Mol Cell Biol 1:101-110, 1981. Prives C, Hall PA: The p53 pathway. J Pathol 187:112-126, 1999. Slebos RJC, Clement M, Polak M, et al: Clinical and pathological associations with p53 tumor-suppressor gene mutations and expression of p21WAF1/Cip1 in colorectal carcinoma. Br J Cancer 74:165-171, 1996. Smith DR, Ji C-Y, Goh H-S: Prognostic significance of p53 overexpression and mutation in colorectal adenocarcinomas. Br J Cancer 74:216-223, 1996. Starzynska T, Bromley M, Marlicz K, et al: Accumulation of p53 in relation to long-term prognosis in colorectal carcinoma. Europ J Gastroent Hepatol, 9:183-186, 1997. Takeda A, Nakajima K, Shimada H, et al: Significance of serum p53 antibody detection on chemosensitivity assay in human colorectal cancer. J Surg. Oncol 71:112-116, 1999. Valentini AM, Pirrelli M, Caruso ML: p53 protein in colorectal cancer: clinicopathological correlation and prognosis significance. J Exp Clin Cancer Res 14:139-144, 1995. Yandell DW, Thor AD: p53 analysis in diagnostic pathology. Biologic implications and possible clinical applications. Diag Molec Pathol 2:1-3, 1993. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 275 EP 279 PG 5 ER PT J AU Kvell, K Balogh, P Nemeth, P AF Kvell, Krisztian Balogh, Peter Nemeth, Peter TI Fine-tuning the EBV+ hu-PBL-SCID Xenogeneic Chimera Model Using InVivo Superinfection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE SCID mice; lymphoproliferative disease; EBV superinfection; B cell proliferation ID SCID mice; lymphoproliferative disease; EBV superinfection; B cell proliferation AB Our purpose was to establish a reproducible xenogeneic chimera model to observe tumors similar to the well-known human posttransplant lymphoproliferative disease (LPD). First we followed the original protocol injecting Epstein-Barr virus positive (EBV+) human peripheral blood lymphocytes (PBL) intraperitoneally into immunodeficient (SCID) mice. Human cells showed T cell phenotype in majority one week after the transfer, whereas one month later a shift towards B cell phenotype was evident according to immunohistochemical and flow cytometric analysis. At this stage the intraperitoneal mass of cells suggested a biologically malignant behaviour infiltrating the liver and the spleen of the host animal. Immunohistochemistry indicated proliferating human lymphatic cells expressing EBV associated proteins and characteristic patterns of invasion within the affected organs. Eventually LPD was lethal to the host animals in 46-67 days. However, the microscopic appearance of experimental LPD was different from the human haemopoietic malignancies: the basic structures of lymphatic organs were preserved and the human T and B cells repopulated the normally T and B dependent areas in mice. The phenotypes of the proliferating cells were human and characteristic for the mature T- and B-lymphocytes. No dominant clone developed during in vitroculturing of the biologically invasive mass of cells removed from the tumor-bearing mice. The results of microscopical, immunological, and flow cytometrical analysis suggested a mature but uncontrolled proliferation of human lymphocytes in SCID mice. The original method for the induction of post-transplant LPD in SCID mice was modified in our further experiments to standardise the experimental technique increasing the efficiency of B cell proliferation and the reducing the number of unspecific factors. Subsequent in vivo EBV superinfection was carried out after the intraperitoneal transfer of a reduced quantity of human PBL from different donors. The same disease developed in our modified chimera model as by the use of original protocol except for some valuable differences. All hosts developed LPD regardless the significantly reduced amount of transplanted PBL and it was lethal in a shorter period of time (41-43 days) compared to the original model. The decreased quantity of transplanted human lymphatic cells was formerly insufficient using the original protocol. Therefore this modified and standardised protocol can lead to a more predictable and reproducible model allowing us to examine fine details of posttransplant lympho-proliferative disease. C1 [Kvell, Krisztian] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Balogh, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Nemeth, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. RP Nemeth, P (reprint author), University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, H-7643 Pecs, Hungary. EM pnemeth@apacs.pote.hu CR Amadori A, Veronesi A, Coppola V, et al: The hu-PBL-SCID mouse in human lymphocyte function and lymphomagenesis studies: achievements and caveats. Seminars in Immunology 8- 4:249-254, 1996. Belagyi J: Medical biometry – an introduction. pp.21, 1999. Boyle TJ, Tamburini M, Berend KR, et al: Human B cell lymphoma in severe combined immunodeficient mice after active infection with Epstein-Barr virus. Surgery 112-2:378-386, 1992. Carlsson R, Martensson C, Kalliomaki S, et al: Human peripheral blood lymphocytes transplanted into SCID mice reconstitute an in vivo culture system exhibiting several parameters found in a normal humoral immune response and are a source of immunocytes for the production of human monoclonal antibodies. The Journal Of immunology 148:1065-1071, 1992. Fuzzati-Armento M-T, Duchosal MA: hu-PBL-SCID mice: an in vivo model of Epstein-Barr virus-dependent lymphoproliferative disease. Histology and Histopathology 13:155-168, 1998. Gleick J: Chaos – the birth of a new science pp.305-335, 1987. Hanto DW, Sakamoto K, Purtilo DT, et al: The Epstien-Barr virus in the pathogenesis of posttransplant lymphoproliferative disorders – clinical, pathologic and virologic correlation. Surgery 90:204-213, 1981. Hesselton RM, Koup RA, Cromwell MA, et al: Human peripheral blood xenografts in the SCID mouse: characterisation of immunologic reconstitution. The Journal of Infectious Diseases 168:630-640, 1993. Joklik WK, Willett HP: Epstein-Barr virus. Microbiology pp.954. Lacerda JF, Ladanyi M, Jagiello C, et al: Administration of rabbit anti-asialo GM1 antiserum facilitates the development of human Epstein-Barr virus-induced lymphoproliferations in xenografted C.B.-17 scid/scid mice. Transplantation 61-3:492- 497, 1996. Malynn BA, Blackwell TK, Fulop GM, et al: The SCID defect affects the final step of the immunoglobulin VDJ recombinase mechanism. Cell 54:453-460, 1988. Martino G, Anastasi J, Feng J, et al: The fate of human peripheral blood lymphocytes after transplantation into SCID mice. European Journal of Immunology 23:1023-1028, 1993. Mosier DE: Humanising the mouse – introduction. Seminars in Immunology 8-4:185, 1996. Murphy WJ, Taub DD, Longo DL: The hu-PBL-SCID mouse as a means to examine human immune function in vivo. Seminars in Immunology 8-4:233-242, 1996. Nakamine H, Okano M, Taguchi Y, et al: Hematopathologic features of Epstein-Barr virus-induced human B-lymphoproliferation in mice with severe combined immune deficiency. Laboratory Investigation 65-4:389-399, 1991. Picchio G, Kobayashi R, Kirven M, et al: Heterogeneity among Epstein-Barr virus-seropositive donors in the generation of immunoblastic B cell lymphomas in SCID mice receiving human peripheral blood leukocyte grafts. Cancer Research 52:2468-2477, 1992. Thorley-Lawson DA, Babcock GJ: A model for persistent infection with Epstein-Barr virus: the stealth virus of human B cells. Life Science 65-14:1433-1453, 1999. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 280 EP 286 PG 7 ER PT J AU Orosz, Zs Rohonyi, B Luksander, A Szanto, J AF Orosz, Zsolt Rohonyi, Bela Luksander, Antal Szanto, Janos TI Pleomorphic Liposarcoma of a Young Woman Following Radiotherapy for Epithelioid Sarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE epithelioid sarcoma; pleomorphic liposarcoma; postirradiation sarcoma ID epithelioid sarcoma; pleomorphic liposarcoma; postirradiation sarcoma AB A case of a metachronous epithelioid sarcoma and pleomorphic liposarcoma in a young woman is described. The first tumor was an epithelioid sarcoma (ES) with focal rhabdoid features localised in the left calf while the second lesion developed seven years later in the same region was diagnosed as pleomorphic liposarcoma resembling myxofibrosarcoma (''myxoid variant of malignant fibrous histiocytoma'') predominantly composed of moderately differentiated spindle cells. Multiple foci of uni- and plurivacuolated lipoblasts were seen. Following the resection of ES the patient received 57 Gy radiation to the region, therefore we regarded the second tumor as a radiation induced liposarcoma. A further interesting feature of this case is that the development of pleomorphic liposarcoma preceded by 6 months the solitary right parabronchial metastasis of ES and after 4 months of metastasectomy a third tumor developed at the site of the first lesion. This tumor showed dedifferentiation toward pleomorphic malignant fibrous histiocytoma. Our case represents a unique case of postirradiation liposarcoma developed on the base of ES. C1 [Orosz, Zsolt] National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Rohonyi, Bela] Erzsebet Hospital, Department of PathologySopron, Hungary. [Luksander, Antal] Erzsebet Hospital, Department of SurgerySopron, Hungary. [Szanto, Janos] University of Debrecen, Department of OncologyDebrecen, Hungary. RP Orosz, Zs (reprint author), National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, H-1122 Budapest, Hungary. EM zso@oncol.hu CR Amendola BE, Amendola MA, McClatchey KD, et al: Radiation- associated sarcoma: a review of 23 patients with postradiation sarcoma over 50 year period. Am J Clin Oncol 12:411- 415, 1989. Arber DA, Kandalaft PL, Mehta P, et al: Vimentin-negative epithelioid sarcoma. The value of an immunohistochemical panel that includes CD34. Am J Surg Pathol 17:302-307, 1993. Cahan WG, Woodward HQ, Mehta P, et al: Vimentin-negative epithelioid sarcoma. The value of an immunohistochemical panel that includes CD34. Am J Surg Pathol 17:302-307, 1993. Chase DR, Enzinger FM: Epithelioid sarcoma: diagnosis, prognostic indicators and treatment. Am J Surg Pathol 9:241-263, 1985. Enzinger FM, Weiss SW, et al: Soft tissue tumors. St. Louis: Mosby, 1995. Evans HL, Baer SC: Epithelioid sarcoma: a clinicopathologic and prognostic study of 26 cases. Semin Diagn Pathol 10:286- 91; 1993. Fletcher CDM: Das histologische Erscheinungsbild bei Lokalrezidiven von Weichgewebssarkomen. Pathologe 15:196- 200, 1994. Guillou L, Wadden C, Coindre J-M, et al: “Proximal-type” epithelioid sarcoma, a distinctive aggressive neoplasm showing rhabdoid features. Am J Surg Pathol 21:130-146, 1997. Halling AC, Wollan PC, Pritchard DJ, et al: Epithelioid sarcoma: a clinicopathologic review of 55 cases. Mayo Clin Proc 71:636-642, 1996 Jacobsen GK, Mellemgard A, Engelholm SA, et al: Increased incidence of sarcoma in patients treated for testicular seminoma. Eur J Cancer 29A:664-669, 1993. Kaldor JM, Day NE, Band P, et al: Second malignancies following testicular cancer, ovarian cancer and Hodgkin’s disease: an international collaborative study among cancer registries. Int J Cancer 39:571-585, 1987. Laskin WB, Silverman TA, Enzinger FM: Postradiation soft tissue sarcomas. An analysis of 53 cases. Cancer 62:2330-2340, 1988. van Leeuwen FE, Stiggelbout AM, van den Belt-Dusebout AW, et al: Second cancer risk following testicular cancer: a follow-up of 1909 patients. J Clin Oncol 11:415-424; 1993. Mark RJ, Poen J, Tran LM, et al: Postirradiation sarcomas. A single-institution study and review of the literature. Cancer 73:2653-2662, 1994 Mirra JM, Kessler S, Bhuta S, et al: The fibroma-like variant of epithelioid sarcoma. Cancer 69:1382-95; 1992. Wiklund TA, Blomqvist CP, Raty J, et al: Postirradiation sarcoma. Analysis of a nation-wide cancer registry material. Cancer 68:524-531; 1991. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 287 EP 291 PG 5 ER PT J AU Arato, G Fulop, V Degrell, P Szigetvari, I AF Arato, Gabriella Fulop, Vilmos Degrell, Peter Szigetvari, Ivan TI Placental Site Trophoblastic Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE placental trophoblastic tumor; choriocarcinoma; immunohistology; intermediate trophoblast ID placental trophoblastic tumor; choriocarcinoma; immunohistology; intermediate trophoblast AB Placental site trophoblastic tumor (PSTT) is the rarest disease of the gestational trophoblast. Our two cases will be interesting not only because of the rarity of the disease, but because both were recognized before operation. Since the tumor cells are lined up tightly side by side, this disease must be distinguished primarily from tumors of epithelial origin. The authors highlight that the diagnosis should rely on intense hPL-positivity as well as the ultrastructural image of the tumor. In histologically equivocal cases, the determination of hPL, hCG, and MIB-1 immunologic markers can be recommended as routinely performed morphological examinations. Serum hCG monitoring is recommended to follow the evolution of the tumor. C1 [Arato, Gabriella] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Szabolcs u. 35., H-1135 Budapest, Hungary. [Fulop, Vilmos] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Degrell, Peter] County Hospital of Borsod-Abauj-Zemplen, Department of PathologyMiskolc, Hungary. [Szigetvari, Ivan] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. RP Arato, G (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1135 Budapest, Hungary. CR Biran H, Dgani R, Wasswemwn JD, et al: Pneumothorax following induction chemotherapy: a case report. Ann Oncol 3:297-300, 1992. Denny LA, Dehaeck K, Neviz J, et al: Placental site trophoblastic tumor: three case report and literature review. Gynecol Oncol 59:300-303, 1995. Fukunaga M, Ushigome S: Malignant trophoblastic tumor. Hum Pathol 24:1098-1106, 1993. Hopkins MP, et al: Malignant placental site trophoblastic tumor associated with placental abruption, fetal distress and elevated CA-125. Gynecol Oncol 47:267-271, 1992. Kashimura M, Kashimura Y, Oikawa K, et al: Placental site trophoblastic tumor: immunohistochemical and nuclear DNA study. Gynecol Oncol 38:262-267, 1990. Kurman RJ: The morphology, biology and pathology of intermediate trophoblast – a look to the present. Human Pathol 22:847-855, 1991. Kurman RJ, Scully RE, Norris HJ: Trophoblastic pseudotumor of the uterus. An exaggerated form of “syncytial endometritis” simulating a malignant tumor. Cancer 38:1214-1226, 1976. Larsen LG, Theilade K, Skibsted L, et al: Malignant placental site trophoblastic tumor. A case report and review of the literature. APMIS Suppl. 23:138-145, 1991. Marchand F: Uber die sogenannten ”decidualen” geschwulste im Anschloss an normale Gebart, Abort, Blasenmole, und Extrauterin Schwangerschaft. Monatsschr Geburtshilfe Gynaekol 1:419, 1985. Orell JM, Sanders DSA: A particularly aggressive placental site trophoblastic tumor. Histopathol 18:559-561, 1991. Remadi S, Lifschitz-Mercer B, Ben-Hur H, et al: Metastasizing placental site trophoblastic tumor: Immunohistochemical and DNA analysis. Arch Gynecol 259:97-103, 1997. Shin IM, Kurman RJ: Expression of melanoma cell adhesin molecule in intermediate trophoblast. Lab Invest 75. Vardar MA, Altintas A: Placental site trophoblastic tumor. Principles of diagnosis, clinical behaviour and treatment. Eur J Gynecol 16:290-295, 1995. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 292 EP 294 PG 3 ER PT J AU Rethy, AL Kalmanchey, R Klujber, V Koos, R Fekete, Gy AF Rethy, A Lajos Kalmanchey, Rozalia Klujber, Valeria Koos, Rozalia Fekete, Gyorgy TI Acid Sphingomyelinase Deficiency in Beckwith-Wiedemann Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Beckwith-Wiedemann syndrome; acidic sphingomyelinase deficiency; embryonal rhabdomyosarcoma; simultaneous occurrence ID Beckwith-Wiedemann syndrome; acidic sphingomyelinase deficiency; embryonal rhabdomyosarcoma; simultaneous occurrence AB We report the association of Beckwith-Wiedemann syndrome (BWS) and a residual acid sphingomyelinase (ASM) activity of about 35% in a 23 months old Hungarian boy. Besides the classical triad of exomphalos, macroglossia and gigantism some other BWS-related features: polyhydramnios (known from the praenatal history), hemihypertrophy, craniofacial dysmorphy, a mild mental retardation, bilaterally undescended testes, cardiac anomalies and a terminally developed, fatal embryonal rhabdomyosarcoma were present in the patient. The decreased activity of the ASM was measured in the patient’s skin fibroblasts. This result, with hepatomegaly, mental retardation, feeding problems, a failure to thrive and muscle-hypotony, partially resembled the ASM-deficient forms of Niemann-Pick disease (NPD). Morphological analysis of the bone-marrow cells gave normal results. There was no chromosomal alteration found by conventional karyotyping of the patient’s lymphocytes.BWS-associated genes as well as the human ASM gene (SMPD1) are all located at 11p15. DNA-studies by region specific markers as well as mutational analysis for the most common NPD-mutations are planned in the future. This is the first report on the simultaneous occurrence of BWS and ASM-deficiency. C1 [Rethy, A Lajos] Bethesda Children's Hospital of the Hungarian Reformed Church, Bethesda u. 3., H-1146 Budapest, Hungary. [Kalmanchey, Rozalia] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Klujber, Valeria] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Koos, Rozalia] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Fekete, Gyorgy] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. RP Rethy, AL (reprint author), Bethesda Children's Hospital of the Hungarian Reformed Church, H-1146 Budapest, Hungary. EM retlaj@yahoo.com CR Eliot M, Maher ER: Beckwith-Wiedemann syndrome. J Med Genet 31:560-564, 1994. Sotelo-Avila C, Gonzalez-Crussi F, Fowler JW: Complete and incomplete forms of Beckwith-Wiedemann syndrome: Their oncogenic potential. J Pediatr 96:47-50, 1980. Wiedemann HR: Tumors and hemihypertrophy associated with Wiedemann-Beckwith syndrome. Eur J Pediatr 414:429, 1983. Reik W, Maher ER: Imprinting in clusters: lessons from Beckwith- Wiedemann syndrome: Trends Genet 13:330-334, 1997. Feinberg AP: Imprinting of a genomic domain of 11p15 and loss of imprinting in cancer: an introduction. Cancer Res 59, Suppl):1743s-1746s, 1999. Schuchman, EH, Desnick RJ: Niemann-Pick disease types A and B: acid sphingomyelinase deficiencies. In: Scriver C.R., Beaudet A.L., Sly W.S., Valle D, eds.): The metabolic and molecular basis of inherited disease, 7th ed. McGraw-Hill, New York, 1995. pp 2601-2624. Gaudray P, Carle GF, Gerhard DS, et al: Report of the Sixth International Workshop on Human Chromosome 11 Mapping 1998. Nice, France, May 2-5, 1998. Cytogenet Cell Genet 86:167-186, 1999. Neri G, Marini R, Cappa M, et al: Simpson-Golabi-Behmel syndrome: an X-linked encephalo-trophoschisis syndrome. Am J Med Genet 30:287-299, 1988. Grundy RG, Pritchard J, Baraitser M, et al: Perlman and Wiedemann-Beckwith syndromes: two distinct conditions associated with Wilms’ tumour. Eur J Pediatr 151:895-898, 1992. Wiedemann HR, Grosse KR, Dibbern H: An atlas of characteristic syndromes. Wolfe Medical Publications, London, 1985. Cohen MM, Jr: A comprehensive and critical assessment of overgrowth and overgrowth syndromes. Adv Hum Genet 29:22-25, 1992. Mannens M, Alders M, Redeker B, et al: Positional cloning of genes involved in the Beckwith-Wiedemann syndrome, hemihypertrophy, and associated childhood tumors. Med Pediatr Oncol 27:490-494, 1996. Bliek J, Alders M, Ryan A, et al: The Beckwith-Wiedemann syndrome: entrance to genes involved in growth regulation. Abstract of the 6th International Workshop on Human Chromosome 11, Nice, France, 1998. URL: http://wwwiag.unice.fr/ workshop/SCW11-6-Abstracts.html Redeker E, Alders M. Hoovers J, et al: Physical mapping of 3 candidate tumour suppressor genes relative to BWS associated chromosomal breakpoints at 11 p 15.3. Cytogenet Cell Genet 68:222-225, 1995. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 295 EP 297 PG 3 ER PT J AU Rethy, AL AF Rethy, A Lajos TI Growth Regulation, Acid Sphingomyelinase Gene and Genomic Imprinting: Lessons from an Experiment of Nature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE acid shingomyelinase; ceramide; Beckwith-Wiedemann Syndrome; growth regulation; apoptosis; conflict theory; imprinting in clusters ID acid shingomyelinase; ceramide; Beckwith-Wiedemann Syndrome; growth regulation; apoptosis; conflict theory; imprinting in clusters AB The author investigated the possible role of acid sphingomyelinase (ASM) gene (SMPD1) in the regulation of growth, in connection with an experiment of nature. The association of a decreased ASM activity and an overgrowth disorder, Beckwith-Wiedemann Syndrome (BWS) with hemihypertrophy has been described at a 23 months old boy in a recent case report (Rethy et al, in this issue). ASM catalyses the production of ceramide, a key molecule of apoptosis, from sphingomyelin. Based on these data it is suggested that the ASM gene (SMPD1) can suppress/counterbalance the anti-apoptotic effects of BWSrelated growth-promoters, like IGF-II, under normal circumstances. Recent literary data support this view. ASM deficient lymphoblasts derived from patients with Niemann-Pick disease (NPD) fail to undergo apoptosis in response to external signals and Fas cross-linking. BWS-related genes are considered to be regulated by genomic imprinting. Therefore the author compared some characteristics of both SMPD1 and imprinted genes. The analyzed features of SMPD1 gene (few and small introns, Alu 1 repeat element, CC-rich regulatory region, alternative splicing) are characteristic to imprinted genes. Hemihypertrophy, mentioned in the referred BWS-case, is distinctive to the involvement of the maternal allele of the second BWS chromosomal region (BWSCR2) at 11p15.3. The SMPDI gene has been localized just distal to the B05 breakpoint of BWSCR2. Furthermore, in BWS-associated tumors, the loss of heterozygosity (LOH) found on 11p15 was always maternal. Thus, in the case referred to with ASM deficiency the maternal allele has certainly been effected. These conclusions are in accordance with the 'cluster-model of imprinting' as well as with the conflict theory of imprinting. Taken together, the above mentioned clinical and experimental data suggest that SMPD1, most likely at 11p15.4, is an imprinted, maternally expressed, BWS- and apoptosis-related growth suppressor gene. Further studies are necessary to prove this hypothesis. C1 [Rethy, A Lajos] Independent Research Group for Genetics -and Immunology, HRC-Bethesda Children's Hospital, Bethesda u. 3., 1146 Budapest, Hungary. RP Rethy, AL (reprint author), Independent Research Group for Genetics -and Immunology, 1146 Budapest, Hungary. EM retlaj@yahoo.com CR Rehthy LA, Kalmanchey R, Khaber V, et al: Acid Sphingomyelinase Deficiency in Beckwith-Wiedemann Syndrome. Pathol Oncol Res 6:295-297, 2000. Haimovitz-Friedman A, Kolesnick RN, Fuks Z: Ceramide signaling in apoptosis. Brit Med Bullet 53:539.553, 1997. Gaudray P, Carle GF, Gerhard DS, et al: Report of the Sixth International Workshop on Human Chromosome 11 Mapping 1998. Cytogenet Cell Genet 86:168-186m, 1999. Mannens M, Alders M, Redeker B, et al: Positional cloning of genes involved in the Beckwith-Wiedemann syndrome, hemihypertrophy, and associated childhood tumors. Med Pediatr Oncol 27:490-94, 1996. S. Reik W, Maher ER: Imprinting in clusters: lessns from Beckwith-Wiedemann syndrome. Trends Genet 13:330-334, 1997. 6. Feinberg AP: Imprinting of a genomic domain of 11p15 and loss of imprinting in cancer: an introduction. Cancer Res 59, Suppl:, 1743s-1746s. 1999. 7. Moore T, Haig D: Genomic imprinting in mammalian development: A parental lug of war. Trends Genet 7:45-49, 1991. 8. http://www.ncbi.nlm.nih.gov/pubmed/ 9. Hurst DL, McVean G, MooreT: Imprinted genes have few and small introns. Nat Genet 12:234-237, 1996. 10. Mannens M, Hoovers JM, Redeker E, et al: Parental imprinting of hum. chromosome region 11p15.3-pter involved in the Beckwith-Wiedemann syndrome and various human neoplasia. Eur J Hum Genet 2:3-23, 1994. 11. Santana P, Pena LA, Haimovitz-Friedman A, et al: Acid sphingomyelinase-deficient human lymphoblasts and mice are defective in radiation-induced apoptosis. Cell 86:189-199, 1996. 12. De Maria R, Rippo MR, Schucman EH, et al : Acidic sphingomyelinase, ASM, is necessary for far-induced GD3 ganglioside accumulation and efficient apoptosis of lymphoid cells. J Exp Med. 187:897-902, 1998. 13. Cock JG. Tepper AD, de Vries E, er al: CD95 Was/APO-II induces ceramide formation and apoptosis in the absence of a functional mid sphingomyelinase. J Biol Chem 273:7560-7565, 1998. 14. Separovic D, Pink JJ, Oleinick NA, et al: Nicmann-Pick human lymphoblasts are resonant to phthatheyanine 4-photodynamic therapy-induced apoposis. Biochem Biophys Res Common 258:506-512, 1999. 15. Malisan F, Testi R: Lipid signaling in CD95-mediated apoptosis. FEBS Lett 452:100-101 1999. 16. McKusick, VA: Mendelian Inheritance in Man. Catalogs of Human Genes and Genetic Disorders. Baltimore: Johns Hopkins University Press. 1998, 12th edition). 17. Schuchman EH, Desnick RJ: Niemann-Pick disease types A and B: acid sphingomyelinase deficiencies. In: Striver C.R.. Braider Sly WS., Valle D, eds.): The metabolic and molecular basis of inherited disease. 7th ed. McGraw-Hill, New York, 1995. pp 2601-2624. 18. Weil D, D'Alessio M, Ramirez F et al: A base substitution in the exon of a oullagen gene causes alternative splicing and generates a structurally abnormal polypeptide in a patient with Ehlers-Danlas syndrome type VII. EMBO J 8:1705-1710, 1989. 19. Neumann B, Kubicka P, Barlow: Characteristics of imprinted genes. Nat Genet 9:12-13, 1995. 20. Ellsworth DL, Hewett-Emmett D, Li WH: Evolution of base composition in the insulin and insulin-like growth factor genes. Mol Biol Evol 11:875-885, 1994. 21. Duret L, Mouchiroud D, Gautier C: Statistical analysis of vertebrate sequences reveals that long genes are scarce in GC richisochores. J Mol Evol 40:308-3M, 1995. 22. Nielsen FC, Ostergaard IL, Nielsen J, et al: Growth-dependent translation of IGF-II mRNA by a rapamycin-sensitive pathway. Nature 377:358-362,1995. 23. Alders M, Bliek J, Redeker B, et al: Cloning of candidate genes involved in the Beckwith-Wiedemann syndrome and childhood tumors. Med Pediatr Oncol 27:495-497, 1996. 24. Redeker E, Alders M, Hoovers J, at al: Physical mapping of 3 candidate tumouniuppressor genes relative to BWS associated chromosomal breakpoints at 1 1p 15.3. Cytogenet Cell Genet 68:222-225, 1995. 25. Lee MP, Feinberg AP: Genomic imprinting of a human apoptosis gene homologue, TSSC3. Cancer Res 58;1052-1056, 1998. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 298 EP 300 PG 3 ER PT J AU Loreto, FM De Martinis, M Corsi, PM Modesti, M Ginaldi, L AF Loreto, Francesca Maria De Martinis, Massimo Corsi, Pia Maria Modesti, Marco Ginaldi, Lia TI Coagulation and Cancer: Implications for Diagnosis and Management SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE thrombosis; malignancy; haemostasis; oncology; therapy ID thrombosis; malignancy; haemostasis; oncology; therapy AB Coagulation disorders are a common problem in neoplastic patients and many factors contribute to increase the risk of thromboembolic events in these patients. An hypercoagulable state is induced by malignant cells interacting directly with hemostatic system and activating the coagulation cascade. More sensitive tests to assess an hypercoagulable state in cancer patients have been developed; even though these tests are always altered in cancer patients, none of them possess a clinical significance in terms of predictive value for the occurence of thromboembolism and disease prognosis in the individual patient. The most frequent thromboembolic complications in cancer patients are deep vein thrombosis of the lower extremities and pulmonary embolism; therefore, disseminated intravascular coagulation, thrombotic thrombocytopenic purpura or haemolytic uremic syndrome are special manifestations of neoplastic disease. Diagnosis of idiopathic deep vein thrombosis, in the absence of other risk factors, could indicate the presence of occult malignant disease; however, the need for an extensive work-up to detect malignancy is still controversial. Neoplastic patients showing a thromboembolic event should be treated with unfractioned heparin or, alternatively, with low molecular weight heparins. In order to prevent recurrence, the administration of heparin should be associated and followed by an oral anticoagulant drug. In recent years new approaches in anti-aggregation therapy have been studied, such as COX-inhibitors, cicaprost and ReoPro; further studies are needed to determine the usefulness of these molecules in treatment of malignancies. C1 [Loreto, Francesca Maria] University of L'Aquila, Department of Internal Medicine and Public Health, Via San Sisto 22/E, 67100 L'Aquila, Italy. [De Martinis, Massimo] University of L'Aquila, Department of Internal Medicine and Public Health, Via San Sisto 22/E, 67100 L'Aquila, Italy. [Corsi, Pia Maria] University of L'Aquila, Department of Internal Medicine and Public Health, Via San Sisto 22/E, 67100 L'Aquila, Italy. [Modesti, Marco] University of L'Aquila, Department of Internal Medicine and Public Health, Via San Sisto 22/E, 67100 L'Aquila, Italy. [Ginaldi, Lia] University of L'Aquila, Department of Internal Medicine and Public Health, Via San Sisto 22/E, 67100 L'Aquila, Italy. RP De Martinis, M (reprint author), University of L'Aquila, Department of Internal Medicine and Public Health, 67100 L'Aquila, Italy. EM demartinis@sscaq1.cc.univaq.it CR Adany R: Intracellular factor XIII: cellular distribution of factor XIII subunit A in humans. Semin Thromb Hemost 22:399- 408, 1996. Bardos H, Juhansz A, Repassy G, et al: Fibrin deposition in squamous cell carcinomas of the larynx and hypopharynx. Thromb Haemost 80:767-772, 1998. Baron JA, Gridley G, Weiderpas E, et al: Venous thromboembolism and cancer. Lancet 351:1077-1080, 1998. Bastida E, Ordinas A, Jamieson GA: Differing platelets aggregating effects by two tumor cells lines: absence of a role for platelet-derived ADP. Am J Hematol 11:367-378, 1981. Bergqvist D, Burmark US: Low-molecular-weight heparin started before surgery as prophylaxis against deep-vein thrombosis: 2500 versus 5000 anti-Xa units in 2070 patients. Br J Surg 82:496-501, 1995. Bertomeu MC, Gallo S, Lauri D Levine MN, et al: Chemotherapy enhances endothelial reactivity to platelets. Clin Expl Metastasis 8:511-518, 1990. Bick RL. Alterations of hemostasis associated with malignancy: etiology, pathophysiology, diagnosis and management. Semin Thromb Hemost 5:1-26, 1978. Bottasso B, Mari D, Coppola R, et al: Hypercoagulability and hyperfibrinolysis in patients with melanoma. Thromb Res 81:345-352, 1996. Brown LF, Detmar M, Claffey KP: Vascular permeability factor/ vascular endothelial growht factor: a multifunctional angiogenesis factor. In: Goldberg ID, Rosen EM, eds. Regulation of Angiogenesis. Basel Birgkhauser Verlag, 233-269; Buccheri G, Ferrigno D, Ginardi C, et al: Haemostatic abnormalities in lung cancer: prognostic implications. Eur J Cancer 33:50-55, 1997. Calvo FA, Hidalgo OF, Gonzales F, et al: Urokinase combination chemotherapy in small cell lung cancer. A phase II study. Cancer 70:2624-2630, 1992. Carey MG, Rodgers GM: Disseminated intravascular coagulation: clinical and laboratory aspects. Am J Hematol 59:65-73, 1998. Chahinian AP, Propert KJ, Ware JH, et al: A randomized trial of anticoagulation with warfarin and alternating chemotherapy in extensive small-cell lung cancer by the Cancer and Leukemia Group B. J Clin Oncol 7:993-1002, 1989. Chan A, Woodruff RK: Complication and failure of anticoagulation therapy in the treatment of venous thromboembolism in patients with disseminated malignancy. Aust NZ J Med 22:119-122, 1992. Chen YQ, Trikha M, Gao X, et al: Ectopic expression of platelet integrin alphaIIb beta3 in tumor cells from various species and histological origin. Int J Cancer 72:642-648, 1997 Clauss M, Gerlach M, Geralach H, et al:Vascular permeabiliy factor: a tumor-derived polypeptide that induce endothelial cell and monocyte procoagulant activity, and promotes monocyte migration. J Exp Med 172:1535-1545, 1990. Colman RW, Rubin RN: Disseminated intravascular coagulation due to malignancy. Semin Oncol 17:140-146, 1990. Cornuz J, Pearson SD, Creager MA, et al: Importance of findings on the initial evaluation for cancer in patients with symptomatic idiopathic deep venous thrombosis. Ann Intern Med 60:257-277, 1996. Cybulsky MI, Chan MKW, Movat HZ. Acute inflammation and microthrombosis induced by endotoxin, interleukin-1 and tumor necrosis factor and their implication in gram negative infection. Lab Invest 58:365-368, 1988. DeLisser HM, Christofidou-Solomidou M, Strieter RM, et al: Involvement of endothelial PECAM-1/CD31 in angiogenesis. Am J Pathol 151:671-677, 1997 Dittman WA, Majerus PW: Structure and function of thrombomodulin: a natural anticoagulant. Blood 75:329-336, 1990. Dolovich L, Ginsberg JS: Low-molecular weight heparins in the treatment of venous thromboembolism. Vessels 3:4-11, 1997. Donati MB, Gambacorti-Passerinin C, et al: Cancer procoagulant in human tumor cells: Evidence from melanoma patients. Cancer Res 46: 6471-6474, 1986. Edwards RL, Rickles FR, Cronlund M: Mononuclear cell tissue factor generation. J Lab Clin Med 98:917-928, 1981. Eldor A: Thrombotic thrombocytopenic purpura: diagnosis, pathogenesis and modern therapy. Baillieres Clin Haematol 11:475-495, 1998. ENOXAN Study Group: Efficacy and safety of enoxaparin versus unfractionated heparin for prevention of deep-vein thrombosis in elective cancer surgery: a double-blind randomised multicentre trial with venography assessment. Br J Surg 84:1099-1103, 1997. Esmon CT: Possible involvment of cytokines indiffuse intravascular coagulation and thrombosis. Baillieres Clin Haematol 7:453-468, 1994. Falanga A, Rickles FR: Pathophysiology of the thrombophilic state in the cancer patient. Semin Thromb Hemost 25:173-182, 1999. Fisher B, Costantino JP, Wickerham L, et al: Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Projects P-1 Study. J Natl Cancer Inst 90:1371-1388, 1998. Fournier DB, Gordon GB: COX-2 and colon cancer: potential targets for chemoprevention. J Cell Biochem Suppl 34:97-102, 2000. Francis JL, Biggerstaff J, Amirkhosravi A: Hemostasis and Malignancy. Semin Thromb Hemost 24:93-109, 1998. Frishman WH, Burns B, Atac B, et al: Novel antiplatelet therapies for treatment of patients with ischemic heart disease: inhibitors of the platelet glycoprotein IIb/IIIa receptor. Am Heart J 130:877-892, 1995. Ghosh BC, Chiffon EE: Malignant tumors with superior vena cava obstruction. NY State J Med 73:283-289, 1973. Girolami A, Prandoni P, Zanon E, et al: Venous thrombosis of upper limbs are more frequently associated with occult cancer as compared with those of lower limbs. Blood Coagul Fibrinolysis 10:455-457, 1999. Glassman AB: Hemostatic abnormalities associated with cancer and its therapy. Ann Clin Lab Sci 27:391-395, 1997. Goldeberg MA, Ginsburg D, Mayer RJ, et al: Is Heparin administration necessary during induction chemotherapy for patients with acute promielocytic leukaemia? Blood 69:187-191, 1987. Goldeberg RJ, Seneff M, Gore JM, et al: Occult malignant neoplasm in patients with deep vein thrombosis. Arch Intern Med 147:251-253, 1987. Gordon LI, Kwaan HC:Thrombotic microangiopaty manifesting as thrombotic thrombocytopenic purpura/ hemolytic uremic syndrome in the cancer patient. Semin Thromb Hemost 25:217-221, 1999. Gouin-Thibault I, Samama MM: Laboratory diagnosis of the thrombophilic state in cancer patients. Semin Thromb Haemost 25:167-172, 1999. Gralnick HR, Abrell BA: Studies of the procoagulant and fibrinolytic activity of promyelocytes in acute promyelocytic leukemia. Br J Hematol 24:89-99, 1973. Griffin MR, Stanson AW, Brown ML, et al: FJ. Deep-vein thrombosis and pulmonary embolism: risk of subsequent malignant neoplasms. Arch Intern Med 147:1907-1901, 1987. Hayes R, Chesebro JH, Fuster V, et al: Anthithrombotic effects of abciximab. Am J Cardiol 85:1167-1172, May 2000. Hasui Y, Marutsuka K, Nishi S, et al: The content of urokinasetype plasminogen activator and tumor recurrence in superficial bladder cancer. J Urol 151:16-19; 1994. Hejna M, Raderer M, Zielinski CC: Inhibition of metastases by anticoagulants. JNCI 91:22-36, 1999. Hienert G, Kirchheimer JC, Christ G, et al: Plasma urokinasetype plasminogen activator correlates to bone scintigraphy in prostatic carcinoma. Eur Urol 15:256-258, 1988. Hiettarachchi JK, Lok J, Prins MH, et al: Undiagnosed malignancy in patients with deep vein thrombosis, Cancer 83:180- 185, 1998. Honn KV, Busse WD, Sloane BF: Prostacyclin and thromboxane. Implications for their role in tumor cell metastasis. Biochem Pharmacol 32:1-11, 1983. Honn KV, Cicone B, Skoff A. Prostacyclin: a potent antimetastatic agent. Science 212:1270-1272, 1981. Honn KV, Tang DG, Crissman JD: Platelets and cancer metastasis: a casual relationship? Cancer Metastasis Rev 11:325- 351, 1992. Hotton KM, Khorsand M, Hank JA, et al: A phase Ib/II trial of granulocyte-macrophage-colony stimulating factor and interleukin- 2 for renal cell carcinoma patients with pulmonary metastases: a case of fatal central nervous system thrombosis. Cancer 88:1892-1901, 2000. Hull R, Delmore T, Carter C, et al: Adjusted subcutaneous heparin versus warfarin sodium in the long term treatment of venous thrombosis. N Engl J Med 306:189-194, 1982. Kakkar AK, Williamson RCN: Prevention of venous thromboembolism in cancer patients. Semin Thromb Hemost 25:239-244, 1999. Karpakin S, Perlstein E:Role of platelets in tumor cells metastases. Ann Intern Med 95:636-641, 1981. Lacotte L, Thierry A, Delwail V, et al: Thrombotic thrombocytopenic purpura during interferon alpha tretment for chronic myelogenous leukemia. Acta Haematol 102:160-162; 2000. Lebeau B, Chastang CL, Brechot JM: Subcutaneous heparin treatment increases complete response rate and overall survival in small cell lung cancer. Lung Cancer 7, Suppl): 129- 134, 1991. Lee AYY, Levine MN: The thrombophilic state induced by therapeutic agents. Semin Thromb Haemost 25:137-145, 1999. Levine M, Gent M, Hirsh J, et al: A comparison of low-molecular weight heparin administered primarily at home with unfractionated heparin administered in the hospital for proximal deep vein thrombosis. N Engl J Med 334:677-681, 1996. Levine M, Hirsh J, Gent M, et al: Double-bind randomized trial of very low-dose warfarin for prevention of thromboembolism in stage IV breast cancer. Lancet 343:886-889, 1994. Levine M, Hirsh J: The diagnosis and treatment of thrombosis in the cancer patient. Semin Oncol 17:160-171, 1990. Levine MN, Lee AYY: Treatment of venous thromboembolism in cancer patients. Semin Thromb Hemost 25:245-249, 1999. Lhindal AK, Sandset PM, Abildgaard U: Indices of hypercoagulation in cancer as compared with those in acute inflammation and acute infarction. Haemostasis 20:253-262, 1990. Lopez Y, Paloma MJ, Rifon J, et al: Measurement of prethrombotic markers in the assessment of acquired hypercoagulable states. Thromb Res 15:71-78, 1999. Ludwig J, Kentos A, Creiner L, et al: Disseminated intravascular coagulation syndrome as manifestation of breast adenocarcinoma metastasis. Rev Med Brux 18:385-388, 1997. Luzzatto G, Schafer AI: The prethrombotic state in cancer. Seminars in Ocology 17:147-159, 1990. Masferrer JL, Leahy KM, Koki AT, et al: Antiangiogenic and antitumor activities of cyclooxygenase-2 inhibitors. Cancer Res 60(5): 1306-11; 2000. Min KW, Gyorkey F, Sato C: Mucin producing adenocarcinomas and nonbacterial thrombotic endocarditis. Cancer 45:2374-2382; 1980. Miyake H, Hara I, Yamanaka K, et al. Elevation of urokinasetype plasminogen activator and its receptor densities as new predictors of disease progression and prognosis in men with prostate cancer. Int J Oncol 14:535-541, 1999. Monreal M, Lafoz E, Casals A, et al: Occult cancer in patients with deep vein thrombosis: a systematic approach. Cancer 67:541-545, 1991. Monreal M, Prandoni P: Venous thromboembolism as first manifestation of cancer. Semin Thromb Hemost 25:131-136; 1999. Muszbek L, Adany R, Mikkola H: Novel aspects of blood coagulation factor XIII. I. Structure, distribution, activation, and function. Crit Rev Clin Lab Sci 33:357-421, 1996. Nicholson GL, Custead SE: Effects of chemotherapeutic drugs on platelet and metastatic tumor cell-endothelial cell interaction as a model for assessing vascular integrity. Cancer Res 45:331-336, 1985. Nie D, Tang K, Szekeres K, et al: Eicosanoid regulation of angiogenesis in human prostate carcinoma and its therapeutic implications. Ann N Y Acad Sci 905:165-176, 2000. Newman PJ: The role of PECAM-1 in vascular cell biology. Ann N Y Acad Sci 714:165-174, 1994. Nordstrom M, Lindblad B, Anderson H, et al: Deep vein thrombosis and occult malignancy: an epidemiological study. BMJ 308:891-894, 1994. Oberhoff C, Winkler UH, Tauchert AM, et al: Adjuvant CMF chemotherapy in patients with breast cancer: result on blood coagulation and fibrinolysis. Zentralbl Gynakol 119:211-217, 1997. O’Connor NT, Cederholm-Williams SA, Fletcher EW, et al: Significance of idiopathic deep venous thrombosis. Postgrad Med J 147:1907-1911, 1987. Ohtake N, Kurita S, Fukabory Y, et al: Clinical study on prostate cancer initially presenting with disseminated intravascular coagulation syndrome. Hinyokika Kiyo 44:387-390, 1998. Owen CA, Bowie EJW: Chronic intravascular coagulation syndromes, a summary. Mayo Clin Proc 49: 673-679, 1974. Ozguroglu M, Arun B, Erzin Y, et al: Serum cardiolipin antibodies in cancer patient with thromboembolic events. Clin Appl Thromb Hemost 5:181-184, 1999. Porta C, Danova M, Riccardi A, et al: Cancer Chemotherapyrelated thrombotic thrombocytopenic purpura: biological evidence of increased nitric oxide production. Mayo Clin Proc 74:570-574, 1999. Prandoni P, Lensing AWA, Buller HR, et al: Comparison of subcutaneous low-molecular weight heparin with intravenous standard heparin in proximal deep vein thrombosis. Lancet 339:441-445, 1992. Prandoni P, Lensing AWA, Buller HR, et al: Deep-vein thrombosis and the incidence of subsequent symptomatic cancer. N Engl J Med 327:1128-1133, 1992. Prandoni P, Lensing AWA, Cogo Cuppini S, et al: The longterm clinical course of acute deep venous thrombosis. Ann Intern Med 125:1-7; 1996. Prandoni P, Piccioli A, Girolami A: Cancer and venous thromboembolism: an overview. Hematologica 84:437-445, 1999. Prandoni P: Antithrombotic strategies in patients with cancer. Thromb Haemost 78:141-144, 1997. Probst-Cousin S, Rickert CH, Gullotta F: Factor XIIIaimmunoreactivity in tumors of the central nervous system. Clin Neuropathol 17:79-84, 1998. Raveh E, Cohen M, Shpitzer T, et al: Carcinoma of the thyroid: a cause of hypercoagulability? Ear Nose Throat J 74:110-112, 1995. Rayson D, Ingle JN, Pruthi RH: Thrombosis of the internal jugular vein: a rare manifestation of the hypercoagulability of malignancy. Cancer Invest 16:319-321, 1998. Rella C, Caviello M, Giotta F, et al: A prothrombotic state in breast cancer patients treated with adjuvant chemotherapy. Breast Cancer Res Treat 40:151-159, 1996. Rickles FR, Edwards RL, Barb C, et al: Abnormalities of blood coagulation in patients with cancer. Cancer 51:301- 307, 1983. Rickles FR, Levine M, Edwards RL: Hemostatic alterations in cancer patients. Cancer Metastasis Rev 11:237-248, 1992. Schafer AI: Bleeding and thrombosis in the myeloproliferative disorders: Blood 69:187-191, 1987. Schneider MR, Schirner M: Antimetastatic prostacyclin analogues. Drugs Future 18:29-48, 1993. Schirner M, Schneider M: Cicaprost does not affect tumor inhibitory potential of cytostatic drugs. Anticancer Res 13:743- 746, 1993. Shirner M, Shneider MR: Cicaprost inhibits metastases of animal tumors. Prostaglandins 42:451-461, 1991. Shirner M, Lichter RB, Schneider MR: The stable prostacyclin analogue cicaprost inhibits metastasis to lungs and lymph nodes in the 1376NF MTLn3 rat mammary carcinoma. Clin Exp Metastasis 12:24-30, 1994. Schulman S, Granqvist S, Holmstrom M, et al:Duration of Anticoagulant Trial Study Group. The duration of oral anticoagulant therapy after a second episode of venous thromboembolism. N Engl J Med 336:393-398, 1997. Semeraro N, Colucci M: Tissue factor in health and disease. Thromb Haemost 78:759-764, 1997. Shands JW: Macrophage procoagulants. Hemostasis 14:373- 377, 1984. Silverstein LR, Nachman RL: Cancer and clotting: Trousseau warning. N Engl J Med 327:1163-1164, 1992. Sorensen HT, Mellemkiaer L, Steffensen FH, et al: The risk of a diagnosis of cancer after primary deep venous thrombosis or pulmonary embolism. N Engl J Med 338:1169-1173, 1998. Skelly MM, Troy A, Duffy MJ, et al: Urokinase-type plasminogen activator in colorectal cancer: relationship with clinicopathological features and patient outcome. Clin Cancer Res 3:1837-1840, 1997. Sun NCJ, McAfee WM, Hum GJ, et al: Hemostatic abnormalities in malignancy, a prospective study of one hundred eight patients. Am J Clin Pathol 71:10-16; 1979. Tang DG, Chen YQ, Newman PJ, et al: Identification of PECAM-1 in solid tumor cells and its potential involvement in tumor cell adhesion to endothelium. J Biol Chem 268:22883- 22894, 1993. Tetu B, Brisson J, Lapointe H, et al: Prognostic significance of stromelysin 3, gelatinase A, and urokinase expression in breast cancer. Hum Pathol 29:979-985, 1998. The Columbus Investigators. Low Molecular weight heparin in the treatment of patients with venous thromboembolism. N Engl J Med 337:657-662, 1997. Thornes RD: Fibrinolytic therapy of leukemia. J R Coll Surg Ireland 6:123-128, 1971. Trikha M, Raso E, Cai Y, et al:Role of alphaII(b)beta3 integrin in prostate cancer metastasis. Prostate 35:185-192, 1998. Trousseau A: Phlegmasia alba dolens. Lecture on clinical medicine, delivered at the Hotel-Dieu, Paris. London: New Sydenham Society, 281-295, 1872. Ustundag Y, Can U, Benli S, et al: N. Internal carotid occlusion in a patient with malignant peritoneal mesothelioma: it is a sign of malignancy-related thrombosis? Am J Med Sci 319:265-267, 2000. Vallejo CT, Rabinovich MG, Perez JE, et al: High-dose cisplatin with dipyridamole in advanced non-small cell lung cancer. A Grupo Oncologico Cooperativo del Sur Study. Am J Clin Oncol Cancer Clin Trial 18:185-188, 1995. Van de Water L, Tracy PB, Aronson D, et al: Tumor cell generation of thrombin via prothrombinase assembly. Cancer Res 45: 5521-5525, 1985. von Tempelhoff GF, Dietrich M, Niemann F, et al: Blood coagulation and thrombosis in patients with ovarian malignancy. Thromb Haemost 77:456-461, 1997. Wada H, Sakuragawa N, Mari Y, et al: Hemostatic molecular markers before the onset of disseminated intravascular coagulation. Am J Hematol 60:273-278, 1999. Winkley JM, Adams HP jr: Potential role of abciximab in ischemic cerebrovascular disease. Am J Cardiol 85:47C-51C, 2000. Wester JPJ, deValk HW, Nieuwenhuis HK, et al: Risk factors for bleed during treatment of acute venous thromboembolism. Thromb Haemost 76:682-688, 1996. Zacharski LR, Henderson WG, Rickles FR, et al: Effect of warfarin anticoagulation on survival in carcinoma of the lung, colon, head and neck, and prostate. Final report of VA Cooperative Study 75. Cancer 53:2046-2052, 1984. Zacharski LR, Wojtukiewicz MZ, Costantini W, et al: Pathways of coagulation/fibrinolysis activation in malignancy. Semin Thromb Hemost 18:104-116, 1992. Zurbron KH, Gram J, Glander K, et al: Influence of cytostatic treatment on the coagulation system and fibrinolysis in patient with non-Hodgkin’s lymphomas and acute leukemias. Eur J Haematol 47:55-59, 1991. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2000 VL 6 IS 4 BP 301 EP 312 PG 12 ER PT J AU Forgacs, E Zochbauer-Muller, S Olah, E Minna, DJ AF Forgacs, Eva Zochbauer-Muller, Sabine Olah, Edit Minna, D John TI Molecular Genetic Abnormalities in the Pathogenesis of Human Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE oncogene; tumor suppressor gene; autocrine; signal transduction; cell cycle; mutation; methylation; telomerase; angiogenesis; preneoplasia ID oncogene; tumor suppressor gene; autocrine; signal transduction; cell cycle; mutation; methylation; telomerase; angiogenesis; preneoplasia AB In the past few years our knowledge of the molecular pathogenesis of lung cancer has significantly increased. There are several molecular mechanisms involved in the multistage carcinogenesis through which respiratory epithelial cells become preneoplastic and then invasive cancer. In this review we summarize some of these changes including, genomic alterations such as loss of heterozygosity and microsatellite alterations, autocrine-paracrine loops, alterations in oncogenes and tumor suppressor genes, tumor angiogenesis, aberrant promoter methylation and inherited predisposition to lung cancer. Translation of these findings to the clinic is also discussed. C1 [Forgacs, Eva] University of Texas Southwestern Medical Center at Dallas, Hamon Center for Therapeutic Oncology Research, 6000 Harry Hines Blvd, 75390-8593 Dallas, TX, USA. [Zochbauer-Muller, Sabine] University of Texas Southwestern Medical Center at Dallas, Hamon Center for Therapeutic Oncology Research, 6000 Harry Hines Blvd, 75390-8593 Dallas, TX, USA. [Olah, Edit] National Institute of Oncology, Department of Biochemistry, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Minna, D John] University of Texas Southwestern Medical Center at Dallas, Hamon Center for Therapeutic Oncology Research, 6000 Harry Hines Blvd, 75390-8593 Dallas, TX, USA. RP Minna, DJ (reprint author), University of Texas Southwestern Medical Center at Dallas, Hamon Center for Therapeutic Oncology Research, 75390-8593 Dallas, USA. EM John.Minna@UTSouthwestern.edu CR Hecht SS: Tobacco smoke carcinogens and lung cancer. J Natl Cancer Inst 91:1194-1210, 1999. Wistuba, II, Lam S, Behrens C: Molecular damage in the bronchial epithelium of current and former smokers. J Natl Cancer Inst 89:1366-1373, 1997. Mao L, Lee JS, Kurie JM: Clonal genetic alterations in the lungs of current and former smokers [see comments]. J Natl Cancer Inst 89:857-862, 1997. Denissenko MF, Pao A, Tang M: Preferential formation of benzo[a]pyrene adducts at lung cancer mutational hotspots in P53. Science 274:430-432, 1996. Wistuba, II, Behrens C, Virmani AK: High resolution chromosome 3p allelotyping of human lung cancer and preneoplastic/ preinvasive bronchial epithelium reveals multiple, discontinuous sites of 3p allele loss and three regions of frequent breakpoints. Cancer Res 60:1949-1960, 2000. Girard L, Zochbauer-Muller S, Virmani AK: Genome-wide allelotyping of lung cancer identifies new regions of allelic loss, differences between small cell lung cancer and non-small cell lung cancer, and loci clustering. Cancer Res 60:4894- 4906, 2000. Sekido Y, Fong KM, Minna JD: Progress in understanding the molecular pathogenesis of human lung cancer. Biochim Biophys Acta 1378:21-59, 1998. Ryberg D, Lindstedt BA, Zienolddiny S: A hereditary genetic marker closely associated with microsatellite instability in lung cancer. Cancer Res 55:3996-3999, 1995. Rosell R, Pifarre A, Monzo M: Reduced survival in patients with stage-I non-small-cell lung cancer associated with DNAreplication errors. Int J Cancer 74:330-334, 1997. Albanell J, Lonardo F, Rusch V: High telomerase activity in primary lung cancers: association with increased cell proliferation rates and advanced pathologic stage. J Natl Cancer Inst 89:1609-1615, 1997. Hiyama K, Hiyama E, Ishioka S: Telomerase activity in smallcell and non-small-cell lung cancers [see comments]. J Natl Cancer Inst 87:895-902, 1995. Fathi Z, Way JW, Corjay MH: Bombesin receptor structure and expression in human lung carcinoma cell lines. J Cell Biochem Suppl 24:237-246, 1996. Richardson GE, Johnson BE: The biology of lung cancer. Semin Oncol 20:105-127, 1993. Cuttitta F, Carney DN, Mulshine J: Bombesin-like peptides can function as autocrine growth factors in human small-cell lung cancer. Nature 316:823-826, 1985. Halmos G, Schally AV: Reduction in receptors for bombesin and epidermal growth factor in xenografts of human small-cell lung cancer after treatment with bombesin antagonist RC-3095. Proc Natl Acad Sci USA 94:956-960, 1997. Shriver SP, Bourdeau HA, Gubish CT: Sex-specific expression of gastrin-releasing peptide receptor: relationship to smoking history and risk of lung cancer. J Natl Cancer Inst 92:24-33, 2000. Rachwal WJ, Bongiorno PF, Orringer MB:Expression and activation of erbB-2 and epidermal growth factor receptor in lung adenocarcinomas. Br J Cancer 72:56-64, 1995. Weiner DB, Nordberg J, Robinson R: Expression of the neu gene-encoded protein, P185neu, in human non-small cell carcinomas of the lung. Cancer Res 50:421-425, 1990. Kern JA, Torney L, Weiner D: Inhibition of human lung cancer cell line growth by an anti-p185HER2 antibody. Am J Respir Cell Mol Biol 9:448-454, 1993. Tateishi M, Ishida T, Mitsudomi T: Immunohistochemical evidence of autocrine growth factors in adenocarcinoma of the human lung. Cancer Res 50:7077-7780, 1990. Damstrup L, Rygaard K, Spang-Thomsen M: Expression of the epidermal growth factor receptor in human small cell lung cancer cell lines. Cancer Res 52:3089-3093, 1992. Lei W, Mayotte JE, Levitt ML: Enhancement of chemosensitivity and programmed cell death by tyrosine kinase inhibitors correlates with EGFR expression in non-small cell lung cancer cells. Anticancer Res 19:221-228, 1999. Ciardiello F, Caputo R, Bianco R: Antitumor effect and potentiation of cytotoxic drugs activity in human cancer cells by ZD-1839, Iressa), an epidermal growth factor receptor- selective tyrosine kinase inhibitor. Clin Cancer Res 6:2053-2063, 2000. Lynch TJ, Jr.: Lung cancer highlights [In Process Citation]. Oncologist 5:274-9, 2000. Harvey P, Warn A, Newman P: Immunoreactivity for hepatocyte growth factor/scatter factor and its receptor, met, in human lung carcinomas and malignant mesotheliomas. J Pathol 180:389-394, 1996. Olivero M, Rizzo M, Madeddu R: Overexpression and activation of hepatocyte growth factor/scatter factor in human nonsmall- cell lung carcinomas. Br J Cancer 74:1862-1868, 1996. Siegfried JM, Weissfeld LA, Singh-Kaw P: Association of immunoreactive hepatocyte growth factor with poor survival in resectable non-small cell lung cancer. Cancer Res 57:433-439, 1997. Quinn KA, Treston AM, Unsworth EJ: Insulin-like growth factor expression in human cancer cell lines. J Biol Chem 271:11477-11483, 1996. Antoniades HN, Galanopoulos T, Neville-Golden J: Malignant epithelial cells in primary human lung carcinomas coexpress in vivo platelet-derived growth factor, PDGF, and PDGF receptor mRNAs and their protein products. Proc Natl Acad Sci USA 89:3942-3946, 1992. Slebos RJ, Kibbelaar RE, Dalesio O: K-ras oncogene activation as a prognostic marker in adenocarcinoma of the lung. N Engl J Med 323:561-565, 1990. Mitsudomi T, Oyama T, Kusano T: Mutations of the p53 gene as a predictor of poor prognosis in patients with non-small-cell lung cancer [see comments]. J Natl Cancer Inst 85:2018-2023, 1993. Rosell R, Li S, Skacel Z: Prognostic impact of mutated K-ras gene in surgically resected non- small cell lung cancer patients. Oncogene 8:2407-2412, 1993. Graziano SL, Gamble GP, Newman NB: Prognostic significance of K-ras codon 12 mutations in patients with resected stage I and II non-small-cell lung cancer. J Clin Oncol 17:668- 675, 1999. Johnson BE, Russell E, Simmons AM: MYC family DNA amplification in 126 tumor cell lines from patients with small cell lung cancer. J Cell Biochem Suppl 24:210-217, 1996. Kaiser U, Schilli M, Haag U: Expression of bcl-2—protein in small cell lung cancer. Lung Cancer 15:31-40, 1996. Pezzella F, Turley H, Kuzu I: bcl-2 protein in non-small-cell lung carcinoma [see comments]. N Engl J Med 329:690-694, 1993. Kok K, Osinga J, Carritt B: Deletion of a DNA sequence at the chromosomal region 3p21 in all major types of lung cancer. Nature 330:578-581, 1987. Naylor SL, Johnson BE, Minna JD: Loss of heterozygosity of chromosome 3p markers in small-cell lung cancer. Nature 329:451-454, 1987. Whang-Peng J, Kao-Shan CS, Lee EC: Specific chromosome defect associated with human small-cell lung cancer; deletion 3p(14-23). Science 215:181-182, 1982. Wistuba, II, Behrens C, Virmani AK: Allelic losses at chromosome 8p21-23 are early and frequent events in the pathogenesis of lung cancer. Cancer Res 59:1973-1979, 1999. Hibi K, Takahashi T, Yamakawa K: Three distinct regions involved in 3p deletion in human lung cancer. Oncogene 7:445- 449, 1992. Todd S, Franklin WA, Varella-Garcia M: Homozygous deletions of human chromosome 3p in lung tumors. Cancer Res 57:1344- 1352, 1997. Wei MH, Latif F, Bader S: Construction of a 600-kilobase cosmid clone contig and generation of a transcriptional map surrounding the lung cancer tumor suppressor gene, TSG, locus on human chromosome 3p21.3: progress toward the isolation of a lung cancer TSG. Cancer Res 56:1487-1492, 1996. Fong KM, Biesterveld EJ, Virmani A: FHIT and FRA3B 3p14.2 allele loss are common in lung cancer and preneoplastic bronchial lesions and are associated with cancer-related FHIT cDNA splicing aberrations. Cancer Res 57:2256-2267, 1997. Lerman IM, Minna DJ: The 630-kb Lung Cancer Homozygous Deletiion Region on Human Chromosome 3p21.3: Identification and Evaluation of the Resident Candidate Tumor Suppressor Genes. Cancer Res 60:6116-6133, 2000. Gao B, Sekido Y, Maximov A: Functional properties of a new voltage- dependent calcium channel alpha(2)delta auxiliary subunit gene, CACNA2D2). J Biol Chem 275:12237-122342, 2000. Sekido Y, Bader S, Latif F: Human semaphorins A(V, and IV reside in the 3p21.3 small cell lung cancer deletion region and demonstrate distinct expression patterns. Proc Natl Acad Sci USA 93:4120-4125, 1996. Shirvan A, Ziv I, Fleminger G: Semaphorins as mediators of neuronal apoptosis. J Neurochem 73:961-971, 1999. Burbee G. D, Forgacs E, Zochbauer-Muller S: Epigenetic Inactivation of RASSFIA in lung and breast cancers and malignant phenotype suppression. J Natl Cancer Inst, In press: 2001. Dammann R, Li C, Yoon JH: Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3 [In Process Citation]. Nat Genet 25:315-9, 2000. Jensen DE, Proctor M, Marquis ST: BAP1: a novel ubiquitin hydrolase which binds to the BRCA1 RING finger and enhances BRCA1-mediated cell growth suppression. Oncogene 16:1097-112, 1998. Papadopoulos N, Nicolaides NC, Wei YF: Mutation of a mutL homolog in hereditary colon cancer [see comments]. Science 263:1625-1629, 1994. Sundaresan V, Roberts I, Bateman A: The DUTT1 Gene, a Novel NCAM Family Member Is Expressed in Developing Murine Neural Tissues and Has an Unusually Broad Pattern of Expression. Mol Cell Neurosci 11:29-35, 1998. Sozzi G, Veronese ML, Negrini M: The FHIT gene 3p14.2 is abnormal in lung cancer. Cell 85:17-26, 1996. Geradts J, Fong KM, Zimmerman PV: Loss of Fhit expression in non-small-cell lung cancer: correlation with molecular genetic abnormalities and clinicopathological features. Br J Cancer 82:1191-1197, 2000. Siprashvili Z, Sozzi G, Barnes LD: Replacement of Fhit in cancer cells suppresses tumorigenicity. Proc Natl Acad Sci USA 94:13771-13776, 1997. Ji L, Fang B, Yen N: Induction of apoptosis and inhibition of tumorigenicity and tumor growth by adenovirus vector-mediated fragile histidine triad, FHIT, gene overexpression. Cancer Res 59:3333-3339, 1999. Sekido Y, Bader S, Latif F:Molecular analysis of the von Hippel- Lindau disease tumor suppressor gene in human lung cancer cell lines. Oncogene 9:1599-1604, 1994. Chevillard S, Radicella JP, Levalois C: Mutations in OGG1, a gene involved in the repair of oxidative DNA damage, are found in human lung and kidney tumours. Oncogene 16:3083- 3086, 1998. Zochbauer-Muller S, Fong M, Virmani K: Aberrant promoter methylation of multiple genes in non-small cell lung cancers. Cancer Res 61:249-255, 2001. Virmani A, Rathi A, Zochbauer-Muller S: Promoter Methylation and Silencing of the Retinoic Acid Receptor-B Gene in Lung Carcinomas. Journal of National Cancer Institute Vol 92.:1303- 1307, 2000. Yunis JJ, Ramsay N: Retinoblastoma and subband deletion of chromosome 13. Am J Dis Child 132:161-163, 1978. Ewen ME: The cell cycle and the retinoblastoma protein family. Cancer Metastasis Rev 13:45-66, 1994. Reissmann PT, Koga H, Takahashi R: Inactivation of the retinoblastoma susceptibility gene in non-small- cell lung cancer. The Lung Cancer Study Group. Oncogene 8:1913-1919, 1993. Cagle PT, el-Naggar AK, Xu HJ: Differential retinoblastoma protein expression in neuroendocrine tumors of the lung. Potential diagnostic implications. Am J Pathol 150:393-400, 1997. Dosaka-Akita H, Hu SX, Fujino M: Altered retinoblastoma protein expression in nonsmall cell lung cancer: its synergistic effects with altered ras and p53 protein status on prognosis. Cancer 79:1329-1337, 1997. Xu HJ, Quinlan DC, Davidson AG: Altered retinoblastoma protein expression and prognosis in early-stage non-small-cell lung carcinoma. J Natl Cancer Inst 86:695-699, 1994. Kratzke RA, Greatens TM, Rubins JB: Rb and p16INK4a expression in resected non-small cell lung tumors. Cancer Res 56:3415-3420, 1996. Shimizu E, Coxon A, Otterson GA: RB protein status and clinical correlation from 171 cell lines representing lung cancer, extrapulmonary small cell carcinoma, and mesothelioma. Oncogene 9:2441-2448, 1994. Sanders BM, Jay M, Draper GJ: Non-ocular cancer in relatives of retinoblastoma patients. Br J Cancer 60:358-365, 1989. Serrano M, Lee H, Chin L: Role of the INK4a locus in tumor suppression and cell mortality. Cell 85:27-37, 1996. Merlo A, Herman JG, Mao L: 5’ CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers [see comments]. Nat Med 1:686-692, 1995. Rusin MR, Okamoto A, Chorazy M: Intragenic mutations of the p16(INK4), p15(INK4B, and p18 genes in primary non-smallcell lung cancers. Int J Cancer 65:734-739, 1996. Marchetti A, Buttitta F, Pellegrini S: Alterations of P16, MTS1, in node-positive non-small cell lung carcinomas. J Pathol 181:178-182, 1997. Shapiro GI, Park JE, Edwards CD: Multiple mechanisms of p16INK4A inactivation in non-small cell lung cancer cell lines. Cancer Res 55:6200-6209, 1995. Okamoto A, Demetrick DJ, Spillare EA: Mutations and altered expression of p16INK4 in human cancer. Proc Natl Acad Sci U S A 91:11045-11049, 1994. Geradts J, Fong KM, Zimmerman PV: Correlation of abnormal RB, p16ink4a, and p53 expression with 3p loss of heterozygosity, other genetic abnormalities, and clinical features in 103 primary non-small cell lung cancers. Clin Cancer Res 5:791-800, 1999. Taga S, Osaki T, Ohgami A: Prognostic value of the immunohistochemical detection of p16INK4 expression in nonsmall cell lung carcinoma. Cancer 80:389-395, 1997. Gazzeri S, Della Valle V, Chaussade L: The human p19ARF protein encoded by the beta transcript of the p16INK4a gene is frequently lost in small cell lung cancer. Cancer Res 58:3926-331, 1998. Sidransky D, Hollstein M: Clinical implications of the p53 gene. Annu Rev Med 47:285-301, 1996. Greenblatt MS, Bennett WP, Hollstein M: Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res 54:4855-4878, 1994. Takahashi T, Nau MM, Chiba I: p53: a frequent target for genetic abnormalities in lung cancer. Science 246:491-494, 1989. Bennett WP, Hussain SP, Vahakangas KH: Molecular epidemiology of human cancer risk: gene-environment interactions and p53 mutation spectrum in human lung cancer. J Pathol 187:8- 18, 1999. Bennett WP, Colby TV, Travis WD: p53 protein accumulates frequently in early bronchial neoplasia. Cancer Res 53:4817-22, 1993. Graziano SL: Non-small cell lung cancer: clinical value of new biological predictors. Lung Cancer 17 Suppl 1:S37-58, 1997. Roth JA, Nguyen D, Lawrence DD: Retrovirus-mediated wildtype p53 gene transfer to tumors of patients with lung cancer. Nat Med 2:985-91, 1996. Roth JA, Swisher SG, Merritt JA: Gene therapy for non-small cell lung cancer: a preliminary report of a phase I trial of adenoviral p53 gene replacement. Semin Oncol 25:33-37, 1998. Forgacs E, Biesterveld EJ, Sekido Y: Mutation analysis of the PTEN/MMAC1 gene in lung cancer. Oncogene 17:1557-1565, 1998. Wu W, Kemp BL, Proctor ML: Expression of DMBT1, a candidate tumor suppressor gene, is frequently lost in lung cancer. Cancer Res 59:1846-1851, 1999. Esteller M, Sanchez-Cespedes M, Rosell R: Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res 59:67-70, 1999. Fontanini G, Vignati S, Boldrini L: Vascular Endothelial Growth Factor Is Associated with Neovascularization and Influences Progression of Non-Small Cell Lung Carcinoma. Clin Cancer Res 3:861-865, 1997. Takanami I, Imamura T, Hashizume T: Immunohistochemical detection of basic fibroblast growth factor as a prognostic indicator in pulmonary adenocarcinoma. Jpn J Clin Oncol 26:293- 297, 1996. Koukourakis MI, Giatromanolaki A, O’Byrne KJ: Plateletderived endothelial cell growth factor expression correlates with tumour angiogenesis and prognosis in non-small-cell lung cancer. Br J Cancer 75:477-481, 1997. Spivack SD, Fasco MJ, Walker VE: The molecular epidemiology of lung cancer. Crit Rev Toxicol 27:319-365, 1997. Spitz MR, Wei Q, Li G: Genetic susceptibility to tobacco carcinogenesis [see comments]. Cancer Invest 17:645-659, 1999. Wu X, Zhao Y, Honn SE: Benzo[a]pyrene diol epoxide-induced 3p21.3 aberrations and genetic predisposition to lung cancer. Cancer Res 58: 605-1608, 1998. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 6 EP 13 PG 8 ER PT J AU John, A Tuszynski, G AF John, Anitha Tuszynski, George TI The Role of Matrix Metalloproteinases in Tumor Angiogenesis and Tumor Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE matrix metalloproteinases; metalloproteinase inhibitors; gelatinases; tumor angiogenesis; metastasis ID matrix metalloproteinases; metalloproteinase inhibitors; gelatinases; tumor angiogenesis; metastasis AB Although a considerable amount of effort has been placed on discovering the etiologies of cancer, the majority of the basic cancer research existing today has focused on understanding the molecular mechanism of tumor formation and metastasis. Metastatic spread of tumors continues to be a major obstacle to successful treatment of malignant tumors. Approximately 30% of those patients diagnosed with a solid tumor have a clinically detectable metastasis and for the remaining 70%, metastases are continually being formed throughout the life of the tumor. Even after the tumor is excised, the threat of death is attributable to the metastasis that may occur through the remaining tumor cells. In addition, treating the metastasis often proves futile since metastasis often vary in size, composition, and anatomical location. New treatments blocking the formation of metastasis will provide greater chances of survival for cancer patients. One family of enzymes that has been shown over the years to play a role in tumor progression is the matrix metalloproteinase (MMP) family. The main function of MMPs, also known as matrixins, is degradation of the extracellular matrix physiologic function involving MMPs include wound healing, bone resorption and mammary involution. MMPs, however, also contribute to pathological conditions including rheumatoid arthritis, coronary artery disease, and cancer. Tumor cells are believed to utilize the matrix degrading capability of these enzymes to spread to distant sites. In addition, MMPs also are thought to promote the growth of these tumor cells once they have metastasized. This review will discuss the role of MMPs and their inhibitors in tumor invasion, angiogenesis and metastasis with special emphasis on the gelatinases, MMP-2 and MMP-9. C1 [John, Anitha] MCP Hahnemann University, Department of PathologyPhiladelphia, USA. [Tuszynski, George] MCP Hahnemann University, Department of PathologyPhiladelphia, USA. RP Tuszynski, G (reprint author), MCP Hahnemann University, Department of Pathology, Philadelphia, USA. EM george.tuszynski@drexel.edu CR Sugarbaker EV: Patterns of metastasis in human malignancies. Cancer Biol Rev 2:235, 1981. Weiss L Gilbert HA, Bone Metastasis: 1981, Boston: G.K. Hall. Liotta LA Stetler-Stevenson WG, Principles of molecular cell biology of cancer: Cancer Metastasis., in Cancer, Principles & Practice of Oncology, VT DeVita, S Hellman, and S.A. Rosenberg, Editors. 1993, Lippincott Co.: Philadelphia. p. 134-149. Delaisse J-M Vaes G: Mechanism of mineral solubilisation and matrix degradation in osteoclastic bone resorption, in Biology and Physiology of the Osteoclast., B.R. Rifkin and C.V. Gay, Editors. 1992, CRC Press: Raton, Florida. p. 290-314. Talhouk RS, Bissell MJWerb Z: Coordinated expression of ECM-degrading proteinases and their inhibitors regulates mammary epithelial function during involution. J Cell Biol 118:1271-1282, 1992. Agren MS, Jorgensen LN, Andersen M, et al: Matrix metalloproteinase 9 level predicts optimal collagen deposition during early wound repair in humans. Brit J Surgery 85:68-71, 1998. Gruber BL, Sorbi D, French DL, et al: Markedly elevated serum MMP-9, gelatinase B, levels in rheumatoid arthritis: a potentially useful laboratory marker. Clinical Immunol Immunopathol 78:161-171, 1996. Parsons SL, Watson SA, Brown PD, et al: Matrix Metalloproteinases. Brit J Surgery 84:160-166, 1997. Tyagi SC: Proteinases and myocardial extracellular matrix turnover. Molecular and Cellular Biochemistry 168:1-12, 1997. Gross J Lapiere CM: Collagenolytic activity in amphibian tissues: a tissue culture assay. Proc Natl Acad Sci USA 48:1014- 1022, 1962. Aimes RT, Quigley JP: Matrix metalloproteinase-2 is an interstitial collagenase. Inhibitor-free enzyme catalyzes the cleavage of collagen fibrils and soluble native type I collagen generating the specific 3/4- and *-length fragments. J Biol Chem 270:5872-5876, 1995. Matrisian L: Metalloproteinases and their inhibitors in matrix remodeling. Trends Genet 6:121-125, 1990. Rooprai HK, McCormick D: Proteases and their inhibitors in human brain tumours: a review. Anticancer Res 17:4151-4162, 1997. Celentano DC, Frishman WH: Matrix metalloproteinases and coronary artery disease: a novel therapeutic target. J Clin Pharmacol 37:991-1000, 1997. Toth M, Gervasi DC, Fridman R: Phorbol ester-induced cell surface association of matrix metalloproteinase-9 in human MCF10A breast epithelial cells. Cancer Res 57:3159-3167, 1997. Miyake H, Yoshimura K, Hara I, et al: Basic fibroblast growth factor regulates matrix metalloproteinases production and in vitro invasiveness in human bladder cancer cell lines. J Urology 157:2351-2355, 1997. Kanno N, Nonomura N, Miki T, et al: Effects of epidermal growth factor on the invasion activity of the bladder cancer cell line. J Urology 159:586-590, 1998. Lamoreaux WJ, Fitzgerald MEC, Reiner A, et al: Vascular endothelial growth factor increases release of gelatinase A and decreases release of tissue inhibitor of metalloproteinases by microvascular endothelial cells in vitro. Microvascular Res 55:29-42, 1998. Song SY, Nomizu M, Yamada Y, et al: Liver metastasis formation by laminin-1 peptide, LQVQLSIR)-adhesion selected B16 - F10 melanoma cells. Int J Cancer 71:436-441, 1997. Tyagi SC, Kumar GS, Glover G: Induction of tissue inhibitor and matrix metalloproteinase by serum in human heart-derived fibroblast and endomyocardial endothelial cells. J Cell Biochem 58:360-371, 1995. Bafetti LM, Young TN, Itoh Y, et al: Intact vitronectin induces matrix metalloproteinase-2 and tissue inhibitor of metalloproteinases- 2 expression and enhanced cellular invasion by melanoma cells. J Biol Chem 273:143-149, 1998. Haas TL, Davis SJ, Madri JA: Three-dimensional type I collagen lattices induce coordinate expression of matrix metalloproteinases MT1-MMP and MMP-2 in microvascular endothelial cells. J Biol Chem 273:3604-3610, 1998. Qian X, Tuszynski GP: Expression of thrombospondin-1 in cancer: a role in tumor progression. Proc Soc Exp Biol Med 212:199-207, 1996. Qian X, Wang TN, Rothman VL, et al: Thrombospondin-1 modulates angiogenesis in vitro by up-regulation of matrix metalloproteinase- 9 in endothelial cells. ExpCell Res 235:403-412, 1997. Sorsa T, Salo T, Koivunen E, et al.: Activation of type IV procollagenases by human tumor-associated trypsin-2. J Biol Chem 272:21067-21074, 1997. Tyagi SC, Kumar S, Katwa L: Differential regulation of extracellular matrix metalloproteinase and tissue inhibitor by heparin and cholesterol in fibroblast cells. J Mol Cell Cardiology 29:391-404, 1997. Mazzieri R, Masiero L, Zanetta L, et al: Control of type IV collagenase activity by components of the urokinase-plasmin system: a regulatory mechanism with cell-bound reactants. EMBO J 16:2319-2332, 1997. Okada Y, Morodomi T, Enghild JJ, et al: Matrix metalloproteinase 2 from human rheumatoid synovial fibroblast: purification and activation of the precursor and enzymatic properties. Eur J Biochem 194:721-730, 1990. Sato H, Seiki M: Membrane-type matrix metalloproteinases, MT-MMPs, in tumor metastasis. J Biochem, Tokyo, 119:209- 215, 1996. Cawston TE, Inhibitors of metalloproteinases, in Proteinase inhibitors, A.J. Barret and G. Salveson, Editors. 1986: Amersterdam. p. 589-610. Gomez DE, Alonso DF, Yoshiji H, et al: Tissue Inhibitors of metalloproteinases: structure, regulation and biological functions. Europ J Cell Biol 74:111-122,1997. Kossakowska AE, Urbanski SJ, Edwards DR: Tissue inhibitor of metalloproteinases, TIMP-1, RNA is expresses at elevated levels in malignant non-Hodgkin’s lymphomas. Blood 77:12475-12481, 1991. Brian J, Wang Y, Smith MR, et al: Suppression of in vivo tumor growth and induction of suspension cell death by tissue inhibitor of metalloproteinases, TIMP-3). Carcinogenesis 9:1805-1811, 1996. Folkman J: Angiogenesis in cancer, vascular, rheumatoid and other disease. Nature Medicine 1:27-31, 1995. Folkman J: Tumor angiogenesis: therapeutic implications. [Review]. New Engl J Med 285:1182-1186, 1971. Fox SB, Gatter KC, Harris AL: Tumour angiogenesis. J Pathology 179:232-237, 1996. Pluda JM: Tumor-associated angiogenesis: mechanisms, clinical implications, and therapeutic strategies. Seminars in Oncology 24:203-218, 1997. Chambers AF, Matrisian LM: Changing views of the role of matrix metalloproteinases in metastasis. J Natl Cancer Inst 89:1260-1270, 1997. Moses MA:The regulation of neovascularization by matrix metalloproteinases and their inhibitors. Stem Cells 15:180-189, 1997. Zucker S, Mirza H, Conner CE, et al: Vascular endothelial growth factor induces tissue factor and matrix metalloproteinase production in endothelial cells – conversion of prothrombin to thrombin results in progelatinase A activation and cell proliferation. Internat J Cancer 75:780-786, 1998. Karelina TV, Goldberg GI, Eisen AZ: Matrix metalloproteinases in blood vessel development in human fetal skin and in cutaneous tumors. J Invest Dermatol 105:411-417, 1995. Taraboletti G, Garofalo A, Belotti D, et al: Inhibition of angiogenesis and murine hemangioma growth by batimastat, a synthetic inhibitor of matrix metalloproteinases. J Natl Cancer Inst 87:293-298,1995. Braunhut SJ, Moses MA: Retinoids modulate endothelial cell production of matrix-degrading proteases and tissue inhibitors of metalloproteinases, TIMP). J Biol Chem 269:13472-13479, 1994. Vacca A, Moretti S, Ribatti D, et al:Progression of mycosis fungoides is associated with changes in angiogenesis and expression of the matrix metalloproteinases 2 and 9. Eur J Cancer 33:1685-1692, 1997. Itoh T, Tanioka M, Yoshida H, et al: Reduced angiogenesis and tumor progression in gelatinase A-deficient mice. Cancer Res 58:1048-1051, 1998. Vu TH, Shipley JM, Bergers G, et al: MMP-9/Gelatinase B is a Key Regulator of Growth Plate Angiogenesis and Apoptosis if Hypertropic Chondrocytes. Cell 93:411-422, 1998. Chu YW, Yang PC, Yang SC, et al: Selection of Invasive and Metastatic Subpopulation from a Human Lung Adenocarcinoma Cell Line. Amer J Resp Cell Mol Biol 17:353-360, 1997. MacDougall JR, Bani MR, Lin Y, et al: The 92-kDa gelatinase B is expressed by advanced stage melanoma cells: Suppression by somatic cell hybridization with early stage melanoma cells. Cancer Res 55:4174-4181, 1995. Ueda Y, Imai K, Tsuchiya H, et al: Matrix metalloproteinase 9, gelatinase B, is expressed in multinucleated giant cells of human giant cell tumor of bone and is associated with vascular invasion. Amer J Pathol 148:611-622, 1996. Kossakowska AE, Hinek A, Edwards DR, et al: Proteolytic activity of human non-Hodgkin’s lymphomas. Amer J Pathol 152:565-576, 1998. Llorens A, Vinyals A, Alia P, et al: Metastatic Ability of MXT Mouse Mammary Subpopulations Coorelates with Clonal Expression and/or Membrane-Association of Gelatinase A. Molecular Carcinogenesis 19: 54-56, 1997. Koshiba T, Hosotani R, Wada M, et al.: Involvement of matrix metalloproteinase-2 activity in invasion and metastasis of pancreatic carcinoma. Cancer 82:642-50, 1998. Deryugina EI, Luo GX, Reisfeld RA, et al: Tumor cell invasion through matrigel is regulated by activated matrix metalloproteinase- 2. Anticancer Res 17:3201-3210, 1997. Koop S, Khokha R, Schmidt EE, et al.: Overexpression of metalloproteinase inhibitor in B16F10 cells does not affect extravasation but reduces tumor growth. Cancer Res 54:4791-4797, 1994. Matrisian LM: Matrix metalloproteinase gene expression. Ann NY Acad Sci 732:42-50, 1993. Zeng ZS, Guillem JG: Distinct pattern of matrix metalloproteinase 9 and tissue inhibitor of metalloproteinase 1 mRNA expression in human colorectal cancer and liver metastases. Bri J Cancer 72:575-582, 1995. Tomita T: Matrix metalloproteinases and tissue inhibitors of metalloproteinases in thyroid C-cells and medullary thyroid carcinomas. Histopathology 31:150-6, 1997. Afzal S, Lalani EN, Poulsom R, et al: MT1-MMP and MMP-2 mRNA expression in human ovarian tumors: possible implications for the role of desmoplastic fibroblasts. Human Pathol 29:155-165, 1998. Harada T, Arii S, Mise M, et al: Membrane-type matrix metalloproteinase- 1, MT1-MMP, gene is overexpressed in highly invasive hepatocellular carcinomas. J Hepatology 28:231-239, 1998. Menashi S, Dehem M, Souliac I, et al: Density-dependent regulation of cell-surface association of matrix metalloproteinase-2, MMP-2, in breast-carcinoma cells. Internat J Cancer 75:259- 265, 1998. Endo K, Maehara Y, Baba H, et al: Elevated levels of serum and plasma metalloproteinases in patients with gastric cancer. Anticancer Res 17:2253-2258, 1997. Gohji K, Fujimoto N, Hara I, et al: Serum matrix metalloproteinase- 2 and its density in men with prostate cancer as a new predictor of disease extension. Int J Cancer 79:96-101, 1998. Kanayama H, Yokota K, Kurokawa Y, et al: Prognostic Values of matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase- 2 expression in bladder cancer. Cancer 82:1359-1366, 1998. Alvarez OA, Carmichael DF, DeClerck YA: Inhibition of collagenolytic activity and metastasis of tumor cells by a recombinant human tissue inhibitor of metalloproteinases. J Natl Cancer Inst 82:589-595, 1990. DeClerck YA, Perez N, Shimada H, et al: Inhibition of invasion and metastasis in cell transfected with an inhibitor of metalloproteinases. Cancer Res 52:701-708, 1992. Tsuchiya Y, Sato H, Endo Y, et al: Tissue inhibitor of metalloproteinase 1 is a negative regulator of the metastatic ability of a human gastric cancer cell line, KKLS, in the chick embryo. Cancer Res 53:1397-1402, 1993. Matsuzawa K, Fukuyama K, Hubbard SL, et al: Transfection of an invasive human astrocytoma cell line with a TIMP-1 cDNA: modulation of astrocytoma invasive potential. J Neuropathol Exp Neurol 55:88-96, 1996. Hoyhtya M, Hujanen E, Turpeenniemi-Hujanen T, et al: Modulation of type-IV collagenase activity and invasive behavior of metastatic human melanoma, A2058, cells in vitro by monoclonal antibodies to type-IV collagenase. Int J Cancer 46:282- 286, 1990. French DL, Ramos-Desimone N, Rozinski K, et al: Matrix metalloproteinase- 9 in tumor cell invasion. Ann NY Acad Sci 732:324-334, 1994. Chirivi RGS, Garofalo A, Crimmin MJ, et al: Inhibition of the metastatic spread and growth of B16-BL6 murine melanoma by a synthetic matrix metalloproteinase inhibitor. Int J Cancer 58:460-464, 1994. Aparicio T, Kermorgant S, Dessirier V, et al: Matrix Metalloproteinase inhibition prevents peritoneal carcinomatosis development and prolongs survival in rats. Carcinogenesis 20:1445-1451, 1999. Steward WP: Marimastat, BB2516): current status of development. Cancer Chemotherapy and Pharmacology 43 Suppl.: 556-560, 1999. Rosemurgy A, Harris J, Langleben A, et al: Marimastat, a novel metalloproteinase inhibitor in patients with advanced carcinoma of the pancreas. 1996. Philadelphia. Rosemurgy A, Harris J, Langleben A, et al: Marimastat in patients with advanced pancreatic cancer: a dose finding study. Amer J Clin Oncol 22:247-252, 1999. Kroep JR, Pinedo HM, VanGroeningen CJ, et al: Experimental drugs and drug combinations in pancreatic cancer. Ann Oncol 10 Suppl:234-238, 1999. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 14 EP 23 PG 10 ER PT J AU Dursun, A Poyraz, A Suer, Sezer, C Akyol, G AF Dursun, Ayse Poyraz, Aylar Suer, Ozlem Sezer, Cem Akyol, Gulen TI Expression of Bcl-2 and c-ErbB-2 in Colorectal Neoplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bcl-2; c-ErbB-2; colorectal neoplasia ID Bcl-2; c-ErbB-2; colorectal neoplasia AB Several studies have been demonstrated the value of c-ErbB-2 and Bcl-2 in predicting the biological behaviour of tumors. The aim of this study was to investigate Bcl-2 and c-ErbB-2 expression in colorectal carcinomas and the correlation between their presence and other clinicopathologic parameters. Eighty-six colorectal carcinomas and 17 adenomas were stained with Bcl-2 and c-ErbB-2 immunohistochemically. Staining patterns were assessed semi-quantitatively and correlated with tumor size, Duke’s classification, tumor differentiation, mucinous characteristic and anatomic locations. We detected Bcl-2 expression in 10 of 17 adenomas (58.8 %) and 31 of 86 carcinomas (36.04 %). Positive staining in normal mucosa was observed only in the compartment of cryptic cells. However neither the difference in the rates of Bcl-2 positivity in adenoma and carcinoma groups, nor the correlation with other mentioned clinicopathological parameters, were found statistically significant. Bcl-2 expression was found to be significantly high in mucinous carcinomas. Expression of c-ErbB-2 was observed in 12 of 86 (13.95 %) carcinomas. It was not detected in adenomas and normal mucosa. Although the incidence of c-ErbB-2 in nonmucinous carcinoma was higher than that of mucinous carcinoma, this was not significant. In addition we were unable to show any significant relation between c-ErbB-2 expression and other clinicopathologic features. Our result suggest that c-ErbB-2 protein expression in colorectal carcinomas, is not very frequent event. There is no correlation between c-ErbB-2 expression and malignant potential of colorectal carcinomas. Higher expressions of Bcl-2 in adenomas than carcinomas suggest us a possible role of Bcl-2 in early carcinogenesis of colon. However since we were unable to find any significant correlation between Bcl-2 expression and other parameters the impact of this gene on biological behavior is still unclear for us. C1 [Dursun, Ayse] Gazi University Faculty of Medicine, Department of Pathology, Turgut Reis Caddesi 16/8 Mebu-sevleri-Tandosan, 06580 Ankara, Turkey. [Poyraz, Aylar] Gazi University Faculty of Medicine, Department of Pathology, Turgut Reis Caddesi 16/8 Mebu-sevleri-Tandosan, 06580 Ankara, Turkey. [Suer, Ozlem] Gazi University Faculty of Medicine, Department of Pathology, Turgut Reis Caddesi 16/8 Mebu-sevleri-Tandosan, 06580 Ankara, Turkey. [Sezer, Cem] Gazi University Faculty of Medicine, Department of Pathology, Turgut Reis Caddesi 16/8 Mebu-sevleri-Tandosan, 06580 Ankara, Turkey. [Akyol, Gulen] Gazi University Faculty of Medicine, Department of Pathology, Turgut Reis Caddesi 16/8 Mebu-sevleri-Tandosan, 06580 Ankara, Turkey. RP Dursun, A (reprint author), Gazi University Faculty of Medicine, Department of Pathology, 06580 Ankara, Turkey. EM aylarpoyraz@superonline.com CR Baretton GB, Diebold J, Christoforis, et al: Apoptosis and immunohistochemical Bcl-2 expression in colorectal adenomas and carcinomas. Cancer 77:255-264, 1996. Bedi A, Pasricha PJ, Akhtar AJ, et al: Inhibition of apoptosis during development of colorectal cancer. Cancer Res. 55:1811- 1816, 1995. Bosari S, Viale G, Bossi P, et al: Cytoplasmic accumulation of p53 protein: an independent prognostic indicator in colorectal adenocarcinomas. J Natl Cancer Inst 86:681-687,1994. Bronner MP, Culin C, Reed JC, et al: The Bcl-2 proto-oncogene and the gastrointestinal epithelial tumor progression model. Am J Pathol 146:20-26, 1995. Fearon ER, Vogelstein B: A genetic model of colorectal carcinogenesis. Cell 61:759-767, 1990. Gorczyca W, Makiewski M, Kram A, et al: Immunohistochemical analysis of Bcl-2 and p53 expression in breast carcinomas: their correlation with Ki-67 growth fraction. Virhows Arch 420:229-233,1995. Hao XP, Ilyas M, Talbot IC, et al: Expression of Bcl-2 and p53 in the colorectal adenoma-carcinoma sequence. Pathobiology 65:140-145,1997. Kapitanovic S, Spaventi R, Poljak L, et al: High c-Erb B-2 protein level in colorectal adenocarcinomas correlates with clinical parameters. Cancer Detection and Prevention 18:97-101, 1994. Kapitanovic S, Radosevic S, Kapitanovic M, et al: The Expression of p185HER-2/neu Correlates With the Stage of Disease and Survival in Colorectal Cancer. Gastroenterology 112:1103-1111, 1997. Lauwers GY, Scott GV, Hendricks J: Immunohistochemical evidence of aberrant Bcl-2 protein expression in gastric epithelial dysplasia. Cancer 73:2900-2904,1994. Manne U, Myers BR, Moron C, et al: Prognostic significance of Bcl-2 expression and p53 nuclear accumulation in colorectal adenocarcinoma. Int J Cancer, Pred. Oncol., 74:346-358, 1997. Menard S, Tagliabue E, Campiglio M, et al: Role of HER2 Gene Overexpression in Breast Carcinoma. J Cell Physol 182:150-162, 2000. Muller-Hocker J: Immunreactivity of p53, Ki67 and Bcl-2 in oncocytic adenomas and carcinomas of the throid gland. Hum Pathol 30:926-933,1999. Reed JC: Bcl-2 and the regulation of programmed cell death. J Cell Biol. 124:1-6,1993. Tollenaar R, Krieken JHJM, Slooten HV, et al: Immunohistochemical detection of p53 and Bcl-2 in colorectal carcinoma: no evidence for prognostic significance. Br J Cancer 77:1842- 1847, 1998. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 24 EP 27 PG 4 ER PT J AU Kleist, B Bankau, A Lorenz, G Poetsch, M AF Kleist, Britta Bankau, Alexander Lorenz, Gerd Poetsch, Micaela TI Multiple Chromosomal Underrepresentations Detected by Interphase Cytogenetics - Possible Prognostic Markers in Head and Neck Tumors? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE head and neck carcinoma; FISH; numerical chromosomal aberrations; TNM status; grading ID head and neck carcinoma; FISH; numerical chromosomal aberrations; TNM status; grading AB Relevant prognostic factors for head and neck squamous cell carcinoma are tumor extension (pT), occurrence of lymph node metastases (pN) and grade of differentiation (G). We tried to correlate these histological characteristics with numerical aberrations of whole chromosomes as demonstrated by fluorescence in situ hybridization techniques (FISH). Therefore, we investigated isolated interphase cells from paraffin sections of squamous cell carcinomas of the head and neck region from 46 patients with centromeric DNA probes for chromosomes 1, 3, 4, 6, 7, 9, 10, 11, 12, 15, 17, 18, X and Y. The majority of tumor samples showed aneuploidy for most chromosomes analyzed. The main chromosomal abnormality was loss of chromosomal material, predominantly of chromosomes 3 (28%), 6 (20%), 9 (26%), 10 (24%) and 18 (33%). Multiple deletions could be demonstrated more frequently in poorly differentiated carcinomas (88% G3-tumors with more than one deletion in contrast to 66% G2-tumors). The occurrence of multiple deletions may also correlate with progression in lymph node metastasis (66% in pN0-tumors vs. 85% in pN2-tumors), whereas the differences between the stages of primary tumor extension were not so obvious. Despite of a some-what disproportionate distribution of tumors in the different pT- and pN-stages and the rather low number of cases, our results suggest a relationship between the quantity of chromosomal underrepresentation, grade of differentiation and higher lymph node stage. Therefore, they underline the importance of chromosomal deletions as a possible additional prognostic marker in head and neck squamous cell carcinoma. C1 [Kleist, Britta] University of Greifswald, Institute of Pathology, F.-Loeffler-Strasse 23e, D-17489 Greifswald, Germany. [Bankau, Alexander] University of Greifswald, Department of OtorhinolaryngologyGreifswald, Germany. [Lorenz, Gerd] University of Greifswald, Institute of Pathology, F.-Loeffler-Strasse 23e, D-17489 Greifswald, Germany. [Poetsch, Micaela] University of Greifswald, Institute of Forensic MedicineGreifswald, Germany. RP Kleist, B (reprint author), University of Greifswald, Institute of Pathology, D-17489 Greifswald, Germany. CR Ambrosch P, Kron M, Fischer G, et al: Micrometastasis in carcinoma of the upper aerodigestive tract: detection, risk of metastasizing, and prognostic value of depth of invasion. Head Neck 17:473-479, 1995. Anneroth G, Batsakis J, Kyba N: Review of the literature and recommended system of malignancy grading in oral squamous cell carcinoma. Scand J Dent Res 95:229-249, 1987. Bailey BJ: Beyond the “new“ TNM classification. Arch Otolaryngol Head Neck Surg 117:369-370, 1991. Berenson JR, Yang J, Mickel RA: Frequent amplification of the bcl-1 locus in head and neck squamous cell carcinomas. Oncogene 4:1111-1116, 1989. Cowan JM, Beckett MA, Ahmed-Swan S, et al: Cytogenetic evidence of the multistep origin of head and neck squamous cell carcinomas. J Natl Cancer Inst 84:793-797, 1992. Fearon ER, Vogelstein B: A genetic model for colorectal tumorigenesis. Cell 61:759-767, 1990. Greenman J, Homer JJ, Stafford ND: Markers in cancer of the larynx and pharynx. Clin Otolaryngol 25:9-18, 2000. Harada Y, Toyomasa K, Ito I, et al:Genetic studies of 457 breast cancers. Clinicopathologic parameters compared with genetic alterations. Cancer 74:2281-2286, 1994. Hunter T: Cooperation between oncogenes. Cell 64:249-270, 1991. Jakobson J, Hansen O, Jorgensen KE, et al: Lymph node metastasis from laryngeal and pharyngeal carcinomas-calculation of burden of metastasis and its impact on prognosis. Acta Oncol 37:489-493, 1998. Jin Y, Mertens F, Mandahl N, et al: Chromosome abnormalities in eighty-three head and neck squamous cell carcinomas: influence of culture conditions on karyotypic pattern. Cancer Res 53:2140-2146, 1993. Li X, Lee NK, Ye Y-W, et al: Allelic loss at chromosomes 3p, 8p, 13q, and 17p associated with poor prognosis in head and neck cancer. J Natl Cancer Inst 86:1524-1529, 1994. Lydiatt WM, Davidson BJ, Shah J, et al:The relationship of loss of heterozygosity to tobacco exposure and early recurrence in head and neck carcinoma. Am J Surgery 168:437-440, 1994. Marshall CJ: Tumor suppressor genes. Cell 64:313-326, 1991. Matthias C, Jahnke V, Hand P, et al: Immunhistologic and molecular genetic studies of the effect of glutathione-S-transferases on the development of squamous epithelial carcinomas in the area of the head and neck. Laryngorhinootologie 78:182-188, 1999. Morales-Angulo C, Val-Bernal F, Buelta L, et al: Prognostic factors in supraglottic laryngeal carcinoma. Otolaryngol Head Neck Surg 119:548-553, 1998. Nicolai P, Redaelli de Zinis LO, Tomenzoli D, et al: Prognostic determinants in supraglottic carcinoma: univariate and Cox regression analysis. Head Neck 19:323-334, 1997. Nishida N, Fukuda Y, Kokuryu H, et al: Accumulation of allelic loss on arms of chromosomes 13q, 16q and 17p in the advanced stages of human hepatocellular carcinoma. Int J Cancer 51:862-868, 1992. Parkin DM, Laara E, Muir CS: Estimates of the world wide frequency of sixteen major cancers in 1980. Int J Cancer 41:184- 97, 1988. Poetsch M, Woenckhaus C, Dittberner T, et al: Increased frequency of numerical chromosomal abnormalities and 1p36 deletions in isolated cells of paraffin sections of malignant melanoma detected by interphase cytogenetics. Cancer Genet Cytogenet 104:146-52, 1998. Poetsch M, Kleist B, Lorenz G, et al: Different numerical chromosomal aberrations detected by FISH in oropharyngeal, hypopharyngeal and laryngeal squamous cell carcinoma. Histopathology 34:234-240, 1999. Raitiola H, Pukander J, Laippala P: Glottic and supraglottic laryngeal carcinoma: differences in epidemiology, clinical characteristics and prognosis. Acta Otolaryngol 119:847-851, 1999. Richter J, Wagner U, Schrami P, et al: Chromosomal imbalances are associated with a high risk of progression in early invasive, pT1, urinary bladder cancer. Cancer Res 59:5687- 5691, 1999. Saretzki G, Hoffmann U, Rohlke P, et al: Identification of allelic loses in benign, borderline, and invasive epithelial ovarian tumors and correlation with clinical outcome. Cancer 80:1241- 1248, 1997. Solano J, Esteban F, Delgado M, et al: Histopathological malignancy and prognosis of laryngeal cancer. Acta Otorrinolaringol Esp 48:375-382, 1997. Takes RP, Baatenburg de Jong RJ, Schuuring E, et al: Differences in expression of oncogenes and tumor suppressor genes in different sites of head and neck squamous cell. Anticancer Res 18:4793-4800, 1998. Van Dyke DL, Worsham MJ, Benninger MS, et al: Recurrent cytogenetic abnormalities in squamous cell carcinomas of the head and neck region. Genes Chrom Cancer 9:192-206, 1994. Vogelstein B, Fearon ER, Hamilton SR, et al: Genetic alterations during colorectal-tumor development. N Engl J Med 319:525-532, 1988. Vogelstein B, Fearon ER, Kern SE, et al: Allelotype of colorectal carcinomas. Science 244:207-211, 1989. Wiernik G, Alcock CJ, Fowler JF, et al: The predictive value of tumor classification compared with results of the British Institute of Radiology fractionation trial in the treatment of laryngopharyngeal carcinoma. Laryngoscope 100:863-872, 1990. Wiernik G, Millard PR, Haybittle JL: The predictive value of histological classification into degrees of differentiation of squamous cell carcinoma of the larynx and hypopharynx compared with the survival of patients. Histopathology 19:411-417, 1991. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 28 EP 32 PG 5 ER PT J AU Bayramoglu, H Duzcan, E AF Bayramoglu, Hatice Duzcan, Ender TI Atypical Epithelial Changes and Mutant p53 Gene Expression in Ovarian Endometriosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ovarian endometriosis; atypia; p53 ID ovarian endometriosis; atypia; p53 AB It has been reported that cases of ovarian endometriosis those with epithelial cytological atypia have potential for malignant transformation. This study was planned to determine the incidence of atypical endometriosis and its cytological criteria, to evaluate the malignant potential of atypical endometriosis via immunohistochemical methods (p53). In this study we evaluated 140 samples obtained from 120 cases of ovarian endometriosis and 10 ovarian endometrioid carcinomas that have been previously diagnosed histopathologically. We re-evaluated endometriosis cases with respect to their epithelial and stromal features, existence of acute or chronic inflammatory cells in endometriotic epithelium or stroma and other accompanying histological findings. We observed atypia in 7 (5.8%) cases; reactive atypia in 37 (30.8%) cases, no atypia in 76 (63.4%) cases. We evaluated immunohistochemical p53 expression in 7 atypical cases, 37 reactive atypical cases, and in 10 of those without atypia and in 10 endometrioid carcinoma cases. We noted no staining in cases with atypia, reactive atypia and without atypia while 3 cases of endometrioid carcinoma had positive staining for p53. We concluded that prominent nucleolus and angulation of nuclear contour could be added to criteria of atypia that were mentioned before in the literature. In our study, even though p53 expression could not be shown with immunohistochemical methods in atypical endometriotic cases; it can not be determined that atypical endometriosis lesions are not premalignant. Still, endometriosis cases should be evaluated carefully by the pathologist for foci of cytological atypia and it should be kept in mind that malignant transformation might occur in these atypical endometriosis cases. C1 [Bayramoglu, Hatice] Pamukkale University, Faculty of Medicine, Department of Pathology, Atakent Mh. Esnaf Sitesi, Sardunya Sk. No:17Denizli, Turkey. [Duzcan, Ender] Pamukkale University, Faculty of Medicine, Department of Pathology, Atakent Mh. Esnaf Sitesi, Sardunya Sk. No:17Denizli, Turkey. RP Bayramoglu, H (reprint author), Pamukkale University, Faculty of Medicine, Department of Pathology, Denizli, Turkey. EM hbayramoglu@hotmail.com CR Clement PB: Diseases of the Peritoneum, In Kurman RJ ed. Blaustein’s Pathology of the Female Genital Tract. New York: Springer-Verlag, 1994, pp660-680. Gerbie AB, Merrill JA: Pathology of endometriosis. Clin Obstet Gynecol 31:779-786, 1988. Clement PB, Young RH, Scully RE: Peritoneum. In Sternberg SS ed. Diagnostic Surgical Pathology. New York, Raven Press Ltd., 1994, pp 2311-2315. Moll UM, Chumas JC, Chalas E, Mann WJ: Ovarian carcinoma arising in atypical endometriosis. Obstet Gynecol 75:537- 539, 1990. LaGrenade A, Silverberg SG: Ovarian tumors associated with atypical endometriosis. Human Pathol 19:1080-1084, 1988. Heaps JM, Nieberg RK, Berek JS: Malignant neoplasms arising in endometriosis. Obstet Gynecol 75:1023-1028, 1990. De La Cuesta RS, Eichhorn JH, Rice LW, et al: Histologic transformation of benign endometriosis to early epithelial ovarian cancer. Gynecol Oncol 60:238-244, 1996. Granai CO, Walters MD, Safai H, et al: M: Malignant transformation of vaginai endometriosis. Obstet Gynecol 64:592-595, 1984. Brooks JJ, Wheeler JE: Malignancy arising in extragonadal endometriosis. Cancer 10:3065-3073, 1977. Horiuch A, Osada R, Nakayama K, et al: Ovarian yolk sac tumor with endometrioid carcinoma arising from endometriosis in a postmenopausal woman, with special reference to expression of alpha-fetoprotein, sex steroid receptora, and p53. Gynecol Oncol 70:295-299, 1998. Han AC, Hovenden S, Rosenblum NG, Salazar H: Adenocarcinoma arising in extragonadal endometriosis: an immunohistochemical study. Cancer 83:1163-1169, 1998. Czernobilsky B, Morns WJ: A histologic study of ovarian endometriosis with emphasis on hyperplastic and atypical changes. Obstet Gynecol 53:318-323, 1979. Chalas E, Chumas J, Barbien R, Mann WJ: Nucleolar organizer regions in endometriosis, atypical endometriosis, and clear cell and endometrioid carcinoma. Gynecol Onco140:260-263, 1991. Ballouk F, Ross JS, Wolf BC: Ovarian Endometriotic cysts, An analysis of cytologic atypia and DNA polidy patterns. Am J Clin Pathol 102:415-419, 1994 Nigro JM, Baker SJ, Preisinger AC, et al: Mutations in the p53 gene occur in diverse human tumour types. Nature 342:705- 708, 1989. Kawai A, Noguchi M, Beppu Y, et al: Nuclear immunoreaction of p53 protein in soft tissue sarcomas. Cancer 73:2499-2505, 1994. Deb S, Jackson CT, Subler MA, Martin DW: Modulation of cellular and viral promoters by mutant human p53 proteins found in tumor cella. J Virol 66:6164-6170, 1992. Harris AL: p53 expression in human breast cancer. Advances Cancer Res 59:69-88, 1992. Rotter V, Prokocimer M: p53 and human malignancies. Advances Cancer Res 57: 257-272, 1991. Walker C, Robertson LJ, Myskow MW, et al: p53 expression in normal and dysplastic bronchial epithelium and in lung carcinomas. Br J Cancer 70:297-303, 1994. Fontanini G, Vignati S, Bigini D, et al: Human non- small cell lung cancer: p53 protein accumulation is an early event and parsists durrog metastatic progression. J Pathol 174:23-31, 1994. Bennett Wp, Hollstein MC, Metcalf RA, et al: p53 mutation and protein accumulation durrog multistage human esophageal carcinogenesis. Cancer Res 52:6092-6097, 1992. Fukunaga M, Nomura K, Ishikawa E, et al: Ovarian atypical endometriosis: its close association with malignant epithelial tumours. Histopathology 30:249-255, 1997. Seidmann JD: Prognostic importence of hyperplasia and atypia in endometriosis. Int J Gynecol Pathol 15:1-9, 1996. Erhan Y, Postaci H: Over endometriozlarmda histolojik ozellikler, Ízmir Devlet Hastanesi Mecmuasi. 21:254-261, 1983. Karseladze AI: Ovarian endometriosis. Arch Pathol 52:24-29, 1990. Marks JR, Davidoff AM, Kerns BJ, et al. Overexpression and mutation of p53 in epithelial ovarian cancer. Cancer Res 51:2979-2984, 1991. Harlozinska A, Bar JK, Sedlaczek P, Gerber J: Expression of p53 protein and Ki-67 reactivity in ovarian neoplasms. Am J Clin Pathol 105:3340-3345, 1996. Koshiyama M, Konishi I, Mandai M, et al: Immunohistochemical analysis of p53 protein and 72 kDa heat shock protein, HSP72, expression in ovarian carcinomas. Correlation with clinicopathology and sex steroid receptor status. Virchows Arch 425:603-609, 1995. Niwa K, Itoh M, Murase T, et al: Alteration of p53 gene in ovarian carcinoma: clinicopathological correlation and prognostic significance. Br J Cancer 70:1191-7, 1994. Kappes S, Milde-Langosch K, Kressin P, et al: p53 mutations in ovarian tumors, detected by temperatere-gradient gel electrophoresis, direct sequencing and immunohistochemistry. Int J Cancer 64:52-59, 1995. Kargi HA, Sagol O, Ozuysal S: The immunohistochemical expression of proliferating cell nuclear antige c-erbB2 and p53 in benign, borderline and malignant epithelial ovarian neoplasms. Turkish J Cancer 27:13-18, 1997. Eccles DM, Brett L, Lessek A, et al: Overexpression of the p53 protein and allele loss at 17p13 in ovarian carcinoma. Br J Cancer 65:40-44, 1992. Schneider J, Jimenez E, Rodriguez F, et al: C-myc, c-erb-B2, nm23 and p53 expression in human endometriosis. Oncol Rep 5:49-52, 1998. Kupryjanczyk J, Bell DA, Yandell DW, et al: p53 expression in ovarian borderline tumor and stage I carcinomas. Am J Clin Pathol 102:671-676, 1994. Cattoretti G, Rilke F, Andreola S, et al: p53 expression in breast cancer. Int J Cancer 41:178-183, 1988. Haerslev T, Jacobsen GK: An immunohistochemical study of p53 with correlations to histopathological parameters, c-erb-2, proliferating cell nuclear antigen, and prognosis. Hum Pathol 26:295-301, 1995. Akasofu M, Oda Y: Immunohistochemical detection of in cervical epithelial lesions with or without infection of human papillomavirus types 16-18. Virchows-Arch 425:593-602, 1995. Martmez J, Georgoff I, Martmez J, et al: Cellular localization and cell cycle regulation by a temperature-sensitive p53 protein. Genes Development 5:151-159, 1991. Iggo R, Gatter K, Bartek J, et al: Increased expression of mutant forms of p53 oncogene in primary lung cancer. Lancet 335;675-679, 1990. Wynford-Thomas D: p53 in tumour pathology: Can we trust immunocytochemistry? J Pathol 166:329-330, 1992. Zhao M, Zhang NX, Laissue SA, Zimmermann A: Immunohistochemical analysis of p53 protein over expression in liver cell dysplasia and in hepatocellular carcinoma. Virchows Archiu 424:613-621, 1994. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 33 EP 38 PG 6 ER PT J AU Ghaderi, AA Talei, AAR Gharesi-Fard, B Farjadian, HF Amirzargar, AA Vasei, M AF Ghaderi, Ali Abbas Talei, Abd-Al-Rasoul Gharesi-Fard, Behronz Farjadian, H F Amirzargar, Ali-Akbar Vasei, Mohammad TI HLA-DRB 1 Alleles and the Susceptibility of Iranian Patients with Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; HLA-DRB1; PCR-SSP ID breast cancer; HLA-DRB1; PCR-SSP AB Breast cancer is considered a major malignancy among women worldwide. The contribution of genetic elements to the onset of familial breast cancer has already been established. The current study investigate the alfele frequency of HLA-DRB 1 in 36 primary operable female breast cancer patients from southern Iran by polymerase chain reaction using sequence specific primers (PCR-SSP). Results were compared with those of 36 female control subjects. Statistical analysis was performed and P values were determined for each character. Our results indicated that the frequency of HLA-DRB 1*12 allele is significantly higher in the patient group (p<0.03) compared to the control group. In addition, HLA-DRB1*11 appeared to be as the most frequent allele in the control group (29.2%) and had approximately the same distribution among the patient group (22.5%). C1 [Ghaderi, Ali Abbas] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Talei, Abd-Al-Rasoul] Medical School, Shiraz University of Medical Sciences, Department of SurgeryShiraz, Iran. [Gharesi-Fard, Behronz] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Farjadian, H F] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Amirzargar, Ali-Akbar] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Vasei, Mohammad] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. RP Ghaderi, AA (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Immunology, Shiraz, Iran. EM immunol@sums.ac.ir CR Garfinkel L, Catherine C, Boring MPH, et al: An overview of breast cancer incidence and mortality. Cancer 74:222-227, 1994. Szabo CI, King MC: Population genetics of BRCA1 and BRCA2. Am J Hum Genet 60:1013-1020, 1997. Miki Y, Swense J, Shatuck-Eidens D, et al: A strong candidate for the breast and ovarian cancer susceptibility. Science 266:66-71, 1994. Reiter DJ, Brocker EB, Ferrone S: Expression and susceptibility of modulation by interferones of HLA-class I and II antigens on melanoma cells. Immunohistochemical analysis and clinical relevance. J Immunogenetics 13:229-234, 1986. Glew SS, Duggan-Keen M, Cabrera T, et al: HLA-class II antigen expression in human papilomavirus-associated cervical cancer. Cancer Res 52:4009-4016, 1992. Paterson AC, Sciot R, Kew MC, et al: HLA expression in human hepatocellular carcinoma. Br J Cancer 57:369-373, 1988. van den Ingh HF, Ruiter DJ, Griffioen G, et al: HLA antigens in colorectal tumours-low expression of HLA class I antigens in mucinous colorectal carcinomas. Br J Cancer 55:125-130, 1987. Tokumoto H: Analysis of HLA-DRB1-related alleles in Japanese patients with lung cancer-relationship to genetic susceptib~ lity and resistance to lung cancer. J Cancer Res Clin Oncol 124:511-516, 1988. Graham DE:The isolation of high molecular weight DNA from whole organisms or large tissue masses. Anal Biochem 85:609- 613, 1978. Miller SA, Dykes DD, Polesky IF: A simple sahing out procedure for extracting DNA from human nucleated cells. Nucl Acid Res 16:1215, 1988. Olerup O, Zetterquist H: HLA-DR typing by PCR amplification with PCR-SSP in two hours. Tissue Antigens 39:225-235, 1992. Casoli C, Zanelli P, Adorni A, et al: Serological and molecular study on the HLA phenotype of female breast cancer patients. Europ J Cancer 30A:1207-1208, 1994. Pardon DM, Topalian SL: The role of CD4+ T cells responses in antitumor immunity. Curr Opin Immunol 10:588-589, 1998. Armstrong TD, Klements VK, Ostrand-Rosenberg S:MHC class II-transfected tumor cells directly present antigen to tumor specific CD4+ T lymphocytes. J Immunol 160:660-666, 1998. Armstrong TD, Klements VK, Ostrand-Rosenberg S: ClassIItransformed tumor cells directly present endogenous antigen to CD4+ T cells in vitro and are APCs for tumor-encoded antigen in vivo. J Immunother 21:218-224, 1998. Feinmesser M, Sulkes A, Morgenstern S, et al: HLA-DR and beta 2 microglobulin expression in medullary and atypical medullary carcinoma of the breast: histopathologically similar but biologically distinct entities. J Clin Pathol 53:286-291, 2000. Dadmarz R, Sgagias MK, Rosenberg SA, et al: CD4+ T lymphocytes infiltrating human breast cancer recognise autologous tumor in an MHC-class-II restricted fashion. Cancer Immunol Immunother 40:1-9, 1995. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 39 EP 41 PG 3 ER PT J AU Magyarlaki, T Buzogany, I Nagy, J AF Magyarlaki, Tamas Buzogany, Istvan Nagy, Judit TI Specific von Hippel-Lindau Protein Expression of Clear Cell Renal Cell Carcinoma with ''Immunogenic” Features SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE clear cell renal cell carcinoma; von Hippel-Lindau protein; tumor infiltrating lymphocytes; immune complexes ID clear cell renal cell carcinoma; von Hippel-Lindau protein; tumor infiltrating lymphocytes; immune complexes AB Human clear cell renal cell carcinoma (CCRCC) is characterized by specific von Hippel-Lindau (VHL) gene alterations and ''immunogenic” features. In the present study, the immunohistochemical expression of the von Hippel-Lindau gene protein (pVHL) was compared with the presence of major histocompatibility complex (MHC I-II), tumor infiltrating lymphocytes (TIL) and tumoral immune complexes (TIC) in CCRCC. Native tumor tissues of 132 RCC patients (95 with the common clear cell subtype), diagnosed according to the Heidelberg classification, were obtained for immunohistochemistry. Tumor stainings with pVHL, MHC I-II and tumor infiltrating lymphocytes (T and B lymphocytes, monocytes) were detected by immunoperoxidase methods using monoclonal antibodies. Tumoral immune complexes (IgG, IgA, IgM and C1q, C3 complement proteins) were visualized by fluorescent polyclonal antibodies. Immune stainings were semiquantitatively evaluated. Specificity and sensitivity of these markers in relation to the common histological subtype of RCC (CCRCC) were calculated. CCRCC was characterized by specific pVHL expression. At the same time, CCRCC was associated with constitutional MHC I-II expression and highly specific degree of TIL and TIC. It is concluded that specific pVHL expression of CCRCC is frequently associated with ''immunogenic” features. Immunohistochemical analysis aims the initial tumor staging of RCC patients to achieve better patient selection for immunotherapy. However, the association of pVHL expression with the ''immunogenic” CCRCC is statistically relevant, the mechanism and its clinical relevance in immunotherapy still remains to be tested. C1 [Magyarlaki, Tamas] University of Pecs, Institute of Biochemistry and Medical Chemistry, Ifjusag utja 13, 7624 Pecs, Hungary. [Buzogany, Istvan] University of Pecs, Department of UrologyPecs, Hungary. [Nagy, Judit] University of Pecs, 2nd Department of Medicine and Nephrological CenterPecs, Hungary. RP Magyarlaki, T (reprint author), University of Pecs, Institute of Biochemistry and Medical Chemistry, 7624 Pecs, Hungary. EM mlaki@clinics.pote.hu CR Belldegrun A, Tao CL, Kaboo R, et al: Natural immune reactivity- associated therapeutic response in patients with metastatic renal cell carcinoma receiving tumor-infiltrating lymphocytes and interleukin-2-based therapy. J Immunother Emphasis Tumor Immunol 19:149-161, 1996. van Bezooijen RL, Goey H, Stoter G, et al: Prognostic markers for survival in patients with metastatic renal cell carcinoma treated with interleukin-2. Cancer Immunol Immunother 43:293-298, 1996. Brauch H, Weirich G, Brieger J, et al: VHL alterations in human clear cell renal cell carcinoma: association with advanced tumor stage and a novel hot spot mutation. Cancer Res 60:1942-1948, 2000. Buszello H, Ackermann R:Immunohistochemical studies on the expression of HLA Class I antigen in renal cell carcinoma: comparison of primary and metastatic tumor tissue. Eur Urol 25:158-163, 1994. Corless CL, Kibel AS, Iliopoulos O, et al: Immunostaining of the von Hippel-Lindau gene product in normal and neoplastic human tissues. Hum Pathol 28:459-463, 1996. Van den Hove LE, van Gool S.W, van Poppel H, et al: Identification of an enriched CD4+ CD8alpha++ CD8beta+ T-cell subset among tumor-infiltrating lymphocytes in human renal cell carcinoma. Int J Cancer 71:178-182, 1997. Van den Hove LE, van Gool SW, van Poppel H, et al: Phenotype, cytokine production and cytolytic capacity of fresh, uncultured, tumor-infiltrating T lymphocytes in human renal cell carcinoma. Clin Exp Immunol 109:501-509, 1997. Jantzer P, Schendel DJ: Human renal cell carcinoma antigenspecific CTLs: antigen-driven selection and long-term persistence in vivo. Cancer Res 58:3078-3086, 1998. Kawata N, Akimoto Y, Hirano, et al: Immunological effect of recombinant interferon-gamma on tumor infiltrating lymphocytes of renal cell carcinoma – relationshship with clinical stage, Japanese). Hinyokika-Kiyo 42:1-4, 1996. Kovacs Gy, Akhtar M, Beckwith BJ, et al: The Heidelberg classification of renal cell tumors. J Pathol 183:131-133, 1997. Kowalczyk D, Skorupski W, Kwias Z, et al: Flow cytometric analysis of tumor-infiltrating lymphocytes in patients with renal cell carcinoma. Br J Urol 80:543-547, 1997. Licht MR, Novick AC, Tubbs RR, et al: Renal oncocytoma: clinical and biological correlates. J Urol 150:1380-1383, 1993. Magyarlaki T, Mosolits S, Baranyay F, et al: Immunohistochemistry of complement responses on human renal cell carcinoma biopsies. Tumori 82:473-480, 1996. Magyarlaki T, Kiss B, Buzogany I, et al: Renal cell carcinoma and paraneoplastic IgA nephropathy. Nephron 82:127-130, 1999. Toliou T, Stravoravdi P, Polyzonis M, et al: Natural killer cell activation after interferon administration in patients with metastatic renal cell carcinoma: an ultrastructural and immunohistochemical study. Eur Urol 29:252-256, 1996. Vogelzang NJ, Stadler WM: Kidney cancer. Lancet 352:1691- 1696, 1998. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 42 EP 45 PG 4 ER PT J AU Gupta, N Sarkar, Ch Singh, R Karak, KA AF Gupta, Nimisha Sarkar, Chitra Singh, Rajvir Karak, Kumar Asis TI Evaluation of Diagnostic Efficiency of Computerized Image Analysis Based Quantitative Nuclear Parameters in Papillary and Follicular Thyroid Tumors Using Paraffin-Embedded Tissue Sections SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE thyroid tumor; image analysis; quantitative nuclear morphometry; papillary carcinoma; follicular neoplasm ID thyroid tumor; image analysis; quantitative nuclear morphometry; papillary carcinoma; follicular neoplasm AB Computerized image analysis (IA) system has emerged in recent years as a very powerful tool for objective and reproducible quantification of histological features. It has shown considerable potential for diagnostic application in diverse histological situations. The objectives of the present study were to evaluate the discriminatory diagnostic efficiency of computerized image analysis based quantitative subvisual nuclear parameters in papillary and follicular neoplasms of thyroid. A total of 60 cases were studied. Forty-four cases belonged to training set and 16 cases belonged to a test set. A minimum of 100 nuclei was analyzed in each case using uniform 5 m mm thick hematoxylin stained sections. The IA workstation comprised of an Olympus microscope, a 10 bit digital video camera, an image grabber card and a pentium 120 MHz computer. Optimas 5.2 software was utilized for data collection on 8 morphometric and 8 densitometric parameters. Multivariate stepwise discriminant statistical analysis of data was done with the help of BMDP statistical software release 7.0. Results from a training set revealed correct classification rates of 98.0%, 84.5% and 61.2% for the histological groups of hyperplastic papillae versus papillae of papillary carcinoma (group I), follicular variant of papillary carcinoma versus the broad category of follicular neoplasms consisting of both follicular adenoma and follicular carcinoma (group II) and follicular adenoma versus follicular carcinoma (group III), respectively. Results of test set revealed correct classification rates of 100%, 80% and 50% for groups I, II and III respectively. It was concluded that computerized nuclear IA parameters have potential usefulness for discriminating benign versus malignant papillary lesions of thyroid, follicular variant of papillary carcinoma versus follicular adenoma and/or follicular carcinoma but are of no value in discriminating between follicular adenoma and follicular carcinoma. C1 [Gupta, Nimisha] All India Institute of Medical Sciences, Department of Pathology and Biostatistics, 110029 New Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology and Biostatistics, 110029 New Delhi, India. [Singh, Rajvir] All India Institute of Medical Sciences, Department of Pathology and Biostatistics, 110029 New Delhi, India. [Karak, Kumar Asis] All India Institute of Medical Sciences, Department of Pathology and Biostatistics, 110029 New Delhi, India. RP Karak, KA (reprint author), All India Institute of Medical Sciences, Department of Pathology and Biostatistics, 110029 New Delhi, India. EM akkarak@hotmail.com CR Ain KB: Papillary thyroid carcinoma. Etiology, assessment and therapy. Endocrinol Metab Clin North Amer 24:711-760, 1995. Artacho-Perula E, Roldan-Villalobos R, Blanco-Garcia F, et al: Objective differential classification of thyroid lesions by nuclear quantitative assessment. Histol. Hisopathol 12:425- 431, 1997. Auer G, Askensten U, Ahrens O: Cytophometry. Hum Pathol 20:518-527, 1989. Baak JP, Blanco AA, Kurver PH, et al: Quantitation of borderline and malignant mucinous ovarian tumors. Histopathology 5:353-360, 1981. Baak JP, Kurver PH, Diegenbach PC, et al: Discrimination of hyperplasia and carcinoma of endometrium by quantitative microscopy. A feasibility study. Histopathology 5: 61-68, 1981. Baak JP, Kurver PH, Overdiep SH, et al: Quantitative, microscopical, computer-aided diagnosis of endometrial hyperplasia or carcinoma in individual patients. Histopathology 5:689-695, 1981. Cabaret V, Vilain MO, Delobelle-Deroide A, et al: Immunohistochemical demonstration of ceruloplasmin and lactoferrin in a series of 59 thyroid tumors. Ann Pathol 12:347-352, 1992. Carcangiu ML, Zampi G, Pupi A, et al: Papillary carcinoma of the thyroid: A clinicopathologic study of 241 cases treated at the University of Florence, Italy. Cancer 55:805-828, 1985. Cavallari V, Maiorana A, La Rosa GL, et al: Morphometric studies on fine-needle aspirates from follicular proliferative lesions of the thyroid. Pathologica 81:441-446, 1989. Chem KT, Rosai J: Follicular variant of thyroid papillary carcinoma. A clinicopathologic study of six cases. Am J Surg Pathol 1:123-130, 1977. Colgan TJ, Norris HJ, Foster W, et al: Predicting the outcome of endometrial hyperplasia by quantitative analysis of nuclear features using linear discriminant function. Int J Gynecol Pathol 1:347-352, 1983. Collin F, Salmon I, Rahier I, et al. Quantitative nuclear cell image analysis of thyroid tumors from archival material. Hum Pathol 22:191-196, 1991. Dawson AE, Austin RE Jr, Weinberg DS: Nuclear grading of breast carcinoma by image analysis: Classification by multivariate and neural network analysis. Am J Clin Pathol 95:529- 37, 1991. De Santis M, Sciarretta F, Sudano L, et al: Morphometric evaluation of histologic sections of thyroid gland in benign and malignant follicular lesions. Diagn Cytopathol 3:60-67, 1987. Diamond DA, Berry SJ, Umbricht C, et al: Computerized image analysis of nuclear shape as a prognostic factor for prostatic cancer. Prostate 3:321-332, 1982. Epstein JI, Berry SJ, Eggleston JC: Nuclear roundness factor : A predictor of progression in untreated stage A2 prostate cancer. Cancer 54:1666-1671, 1984. Erler BS, Truong HM, Kim SS, et al:A study of hepatocellular carcinoma using morphometric and densitometric image analysis. Am J Clin Pathol 100:151-157, 1993. Fassina AS, Montesco MC, Ninfo V, et al: Histological evaluation of thyroid carcinomas : reproducibility of the ``WHO’’ classification. Tumori 79:314-320, 1993. Ferrer-Roca O, Ballester Guardia E, Martin J: Nuclear chromatin texture to differentiate follicular and papillary carcinoma of the thyroid. Pathol Res Pract 185:561-566, 1989. Ferrer-Roca O, Ballester-Guardia E, Martin-Rodriguez JA: Morphometric, densitometric and flow cytometric criteria for the automated classification of thyroid lesions. Analyt Quant Cytol Histol 12:48-55, 1990. Fugazzola L, Pierotti MA, Vigano E, et al: Molecular and biochemical analysis of RET/PTC4, a novel oncogenic rearrangement between Ret and Ele 1 genes in post-Chernobyl papillary thyroid cancer. Oncogene 13:1093-1097, 1996. Galera-Davidson H, Bibbo M, Bartels PH, et al: Differential diagnosis between follicular adenoma and follicular carcinoma of the thyroid by marker features. Anal Quant Cytol Histol 8:195-200, 1986. Ghali VS, Jimenez EJS, Garch RL: Distribution of Leu-7 antigen, HNK-1, in thyroid tumors: Its usefulness as a diagnostic marker for follicular and papillary carcinomas. Hum Pathol 23:21- 25, 1992. Gil J: Image analysis in anatomic pathology : what are the issues? Hum Pathol 20:203-204, 1989. Grebe SK, Hay ID: Follicular thyroid cancer. Endocrinol Metab Clin North Amer 24:761-801, 1995. Gurley AM, Hidvegi DF, Bacus JW, et al: Comparison of the Papanicolaou and Feulgen staining methods for DNA quantification by image analysis. Cytometry 11:468-474, 1990. Hedinger CE, Williams ED, Sobin LH: Histologic Typing of Thyroid Tumors. Springer-Verlag, Berlin, 1988. Holschbach A, Kriete A, Schaffer R: Differential diagnosis of papillary carcinomas of the thyroid using image analysis and three dimensional reconstruction from serial sections. Verh Dtsch Ges Pathol 74:270-274, 1990. Jenkins RB, Hay ID, Herath JF, et al: Frequent occurrence of cytogenetic abnormalities in sporadic non-medullary thyroid carcinoma. Cancer 66:1213-1220, 1990. Koss LG, Bartels PH, Bibbo M, et al: Computer discrimination between benign and malignant urothelial cells. Acta Cytol 19:378-391, 1975. Kotani T, Asada Y, Aratake Y, et al. Diagnostic usefulness of dipeptidyl aminopeptidase IV monoclonal antibody in paraffin embedded thyroid follicular tumors. J Pathol 168:41-45, 1992. Kriete A, Romen W, Schaffer R, et al: Computer analysis of chromatin arrangement and nuclear texture in follicular thyroid tumors. Histochemistry 78:227-230, 1983. La Rosa GL, Cavallari V, Giuffrida D, et al: The morphometric analysis of cell nuclei from fine needle aspirates of thyroid follicular lesions does not improve the diagnostic accuracy of traditional cytologic examination. J Endocrinol Invest 13:701- 707, 1990. Livolsi VA: Surgical Pathology of the thyroid. W B Saunders, Philadelphia, 1990. Livolsi VA, Asa SL: The demise of follicular carcinoma of the thyroid gland. Thyroid 4:233-236, 1994. Luna LG:Manual of Histologic Staining Methods of the Armed Forces Institute of Pathology. McGraw Hill Publications, New York, 1968. Marchevsky AM, Hauptman E, Gil J, et al: Computerized interactive morphometry as an aid in the diagnosis of pleural effusions. Acta Cytol. 31:131-136, 1987. McLaren KM, Cossar DW: The immunohistochemical localization of S-100 in the diagnosis of papillary carcinoma of the thyroid. Hum Pathol 27:633-636, 1996. Miettinen M, Franssila K, Lehto VP, et al: Expression of intermediate filament proteins in thyroid gland and thyroid tumors. Lab Invest 50:262-270, 1984. Montironi R, Alberti R, Sisti S, et al. Discrimination between follicular adenoma and follicular carcinoma of the thyroid: preoperative validity of cytometry on aspiration smears. Appl Pathol 7:367-374, 1989. Nafe R, Fritsch RS, Soudah B, et al: Histomorphometry in paraffin sections of thyroid tumors. Pathol Res Pract 188:1042- 1048, 1992. Nagashima T, Suzuki M, Oshida M, et al: Morphometry in cytologic evaluation of thyroid follicular lesions. Cancer 84:115- 118, 1998. Pasini B, Ceccherini I, Romeo G: Ret mutations in human disease. Trends Genet 12:138-144, 1996. Raphael SJ, McKeown-Eyssen G, Asa SL: High molecularweight cytokeratin and cytokeratin-19 in diagnosis of thyroid tumors. Mod Pathol 7:295-300, 1994. Rosai J, Carcangiu ML: Pitfalls in the diagnosis of thyroid neoplasms. Pathol Res Pract 182:169-179, 1987. Rosai J, Carcangiu ML, Delellis RA: Papillary carcinoma. In: Tumors of the thyroid gland., Eds: Rosai J and Sobin LH), Armed Forces Institute of Pathology, 1992, pp. 65-121. Salmon I, Kiss R, Franc B, et al: Comparison of morphonuclear features in normal, benign and neoplastic thyroid tissue by digital cell image analysis. Analyt Quant. Cytol Histol. 14:47-54, 1992. Saxen E, Franssilla K, Bjarnason O, et al: Observer variation in histologic classification of thyroid cancer. Acta Path Microbiol Scand, A, 86:483-486, 1978. Schuurmann G, Mattfeldt T, Feichter G, et al: Differential diagnosis of follicular neoplasm of the thyroid. Langenbecks Arch Chir 375:95-101, 1990. Schurmann G, Mallfeldt T, Feichter G, et al: Stereology, flow cytometry and immunohistochemistry of follicular neoplasms of the thyroid gland. Hum Pathol 22:179-184, 1991. Sozzi G, Miozzo M, Cariani TC, et al: A t(2:3)(q12-13; p24-25, in follicular thyroid adenomas. Cancer Genet Cytogenet 64:38- 41, 1992. Stanta G, Carcangiu ML, Rosai J: The biochemical and immunohistochemical profile of thyroid neoplasia. Pathol Annual 23, Part I): 129-157, 1988. Tsybrovskyy O, Vassilenko I, Mannweiler S, et al: Multivariate karyometric approach in differential diagnosis of follicular thyroid neoplasms. A study of 31 cases. Virchows Arch 433:135- 143, 1998. Tuccari G, Barresi G: Tissue polypeptide antigen in thyroid tumors of follicular cell origin: an immunohistochemical reevaluation for diagnostic purposes. Histopathology 16:377- 381, 1990. Walts AE, Marchevsky AM: Computerized interactive morphometry: An expert system for diagnosis of lymphoid-rich effusions. Am J Clin Pathol 92:765-772, 1989. Wied GL, Bibbo M, Bartels PH: Computer analysis of microscopic images: Application in cytopathology. Pathol Ann 16:367-409, 1981. Xu XC, el-Naggar AK, Lotan R : Differential expression of galectin-1 and galectin-3 in thyroid tumors. Am J Pathol 147: 815-822, 1995 Yamshita H, Noguchi S, Murakami N, et al. Immunohistological differentiation of benign thyroid follicular cell tumors from malignant ones: usefulness of anti-peroxidase and JT-95 antibodies. Acta Pathol Jpn 43:670-673, 1993. Zou M, Shi Y, Farid NR: Low rate of Ret proto-oncogene activation, PTC/ret TPC, in papillary carcinomas from Saudi Arabia. Cancer 73:176-180, 1994. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 46 EP 55 PG 10 ER PT J AU Kravchick, S Gal, R Cytron, Sh Peled, R Weissman, Y Mukamel, E Koren, R AF Kravchick, Sergey Gal, Rivka Cytron, Shmuel Peled, Ronit Weissman, Yona Mukamel, Eliahu Koren, Rumelia TI Increased Incidence of Diabetes Mellitus in the Patients with Transitional Cell Carcinoma of Urinary Bladder SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Urinary bladder; transitional cell carcinoma; diabetes mellitus ID Urinary bladder; transitional cell carcinoma; diabetes mellitus AB The progression of bladder cancer to invasive disease is highly dependent on its ability to penetrate basement membrane of urothelium. Studies on diabetic nephropathy have shown a reduction in pro-teoglycan content of the glomerular basement membrane. Based on the well-known fact that proteoglycans are one of the main components of basement membrane and extracellular matrix we assessed the relationship between diabetes mellitus, bladder cancer incidence and its behavior. These studies include 252 patients with microscopically confirmed transitional cell carcinoma of bladder, and 549 patients with other urological disorders who served as controls. The prevalence of diabetes mellitus in each group was assessed. The group of patients suffering from transitional cell carcinoma was divided according to etiological risk factors such as cigarette smoking, diabetes and patients that were non-smokers and did not suffer from diabetes mellitus. We assessed the features of bladder cancer behavior in each group. Logistic regression model estimation for statistical analysis was used, with transitional cell carcinoma as a dependent binary variable and age, sexes smoking and diabetes as independent variables. Statistical significance was considered at two levels: p £ £0.001 and p £ £0.05. Odds ratio (OR) adjusted to age, sex, cigarette smoking, diabetes mellitus and 95% Confidence Interval (CI) were calculated for TCC. In the TCC group 22.2% of the patients suffered from diabetes mellitus. In the control group 10.38% suffered from diabetes mellitus. Logistic regression analysis, OR and 95% CI showed a statistically significant relationship between diabetes and TCC. These data are comparable only with smoking (OR – 2.3; 95% CI – 1.6 – 3.5 and OR– 1.58; 95% CI – 1.08 – 2.4 correspondingly). Based on these data we suggest that diabetes mellitus may be considered an etiological risk factor for bladder cancer development. C1 [Kravchick, Sergey] Barzialy Medical Center, Urology DepartmentAshkelon, Israel. [Gal, Rivka] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Cytron, Shmuel] Barzialy Medical Center, Urology DepartmentAshkelon, Israel. [Peled, Ronit] Barzilay Medical Center, Epidemiology UnitAshkelon, Israel. [Weissman, Yona] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Mukamel, Eliahu] Hasharon Hospital, Department of UrologyPetach Tikvah, Israel. [Koren, Rumelia] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. RP Gal, R (reprint author), A, Rabin Medical Center (Golda Campus), Department of Pathology, Petach Tikvah, Israel. EM rumelia@isdn.mail.co.il CR Cotran RS, Kumar V, Robbins SL, et al: Pathologic basis of disease, 5th ed, WB Saunders Co, Philadelphia, Pennsylvania, 1994. Hayes RB, Friedell GH, Zahm SH, et al: Are the known bladder cancer risk factors associated with more advanced bladder cancer? Cancer Causes Control 4:157-162, 1993. Schulte PA, Ringen K, Hemstreet GP, et al: Risk factors for bladder cancer in a cohort exposed to aromatic amines. Cancer 58:21-56, 1986. Silverman DT, Hartge P, Morrison AS, et al: Epidemiology of bladder cancer. Hematol Oncol Clin North Am 6:1-30, 1992. Wilkens LR, Kadir MM, Kolonel LN, et al: Risk factors for lower urinary tract cancer: the role of total fluid consumption, nitrites and nitrosamines, and selected foods. Cancer Epidemiol Biomarkers Prev 5:161-166, 1996. Asten P, Barrett J, Symmons D: Risk of developing certain malignancies is related to duration of immunosuppressive drug exposure in patients with rheumatic diseases. J Rheumatol 26:1705-1714, 1999. Liu BC, Liotta LA:Biochemistry of bladder cancer invasion and metastasis. Urol Clin North Am 19:621-627, 1992. Jackson RL, Buch SJ, Cardin AD: Glycosaminogycans: molecular properties, protein interaction, and role in physiological processes. Physiol Rev 71:481-539, 1991. Ruoslahti E, Yamaguchi Y: Proteoglycans as modulators of growth factor activities. Cell 64:867-869, 1991. Vlodavsky I, Eldor A, Haimovitz-Friedman A, et al: Expression of heparanase by platelets and circulating cells of immune system: possible involvement in diapedesis and extravasation. Invasion Metastasis 12:112-127, 1992. Nakajima M, Irimura T, Nicolson GL: Heparanases and tumor metastasis. J Cell Biochem 36:157-167, 1988. Timar J, Diczhazi C, Bartha I, et al: Modulation of heparan-sulphat/ chondroitin-sulphate ratio by glycosaminoglycan biosynthesis inhibitors affects liver metastatic potential of tumor cells. Int J Cancer 62:755-761, 1995 Tamsma JT, van den Born J, Bruijn JA, et al: Expression of glomerular extra cellular matrix components in human diabetic nephropathy: decrease of heparan sulphate in the glomerular basement membrane. Diabetologia 37:313-320, 1994. Uehling DT, Kelly E, Hopkins WJ, et al: Urinary glycosaminoglycan levels following induced cystitis in monkeys. J Urology 139:1103-1105, 1988. Parsons CL, Boychuk S, Hurst R, et al: Bladder surface glycosaminoglycans: an epithelial permeability barrier. J Urol 143:139-142, 1990. Bodenstab W, Kaufman J, Parsons CL: Inactivation of antiadherence effect of bladder surface glycosaminoglycans by a complete urinary carcinogen, N-Methyl – N – Nitrosourea). J Urol 129:200-201, 1983. Messing EM, Hanson P, Ulrich P, et al: Epidermal growth factor interaction with normal and malignant urothelium: in vivo and in situ studies. J Urol 138:1329-1335, 1987. Messing EM: Clinical implication of the expression of epidermal growth factor in human transitional cell carcinoma. Cancer Res 50:2530–2537, 1990. Kristensen JK, Lose G, Lund F, Nexo E: Epidermal growth factor in urine from patients with urinary bladder tumors. Eur Urol 14:313–314, 1988. Lokeshwar VB, Lokeshwar VB, Young MJ, et al: Identification of bladder tumor-derived hyaluronidase: its similarity to HYAL1. Cancer Res 59:4464-4470, 1999. Attia N, Caprio S, Jones TW, et al: Changes in free insulin-like growth factor-1 and leptin concentrations during acute metabolic decompensation in insulin withdrawn patients with type 1 diabetes. J Clin Endocrinol Metab 84:2324-2328,1999. Butler AA, Blakesley VA, Poulaki V, et al: Stimulation of tumor growth by recombinant human insulin-like growth factor-I, IGF-I, is dependent on the dose and the level of IGF-I receptor expression. Cancer Res 58:3021-3027, 1998. Kamel HK, Rodriguez-Saldana J, Flaherty JH, et al: Diabetes mellitus among ethnic seniors: contrasts with diabetes in whites. Clin Geriatr Med 15:265-278, 1999. Sinclair AJ: Diabetes in the elderly: A perspective from the United Kingdom. Clin Geriatr Med 15:225-37, 1999. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 56 EP 59 PG 4 ER PT J AU Kabukcuoglu, F Sungun, A Senturk, AB Evren, I Ilhan, R AF Kabukcuoglu, Fevziye Sungun, Aysim Senturk, Akan Billur Evren, Ismail Ilhan, Ridvan TI Mixed Germ Cell Tumor of the Ovary with Sarcomatous Component SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Ovary; germ cell; sarcoma ID Ovary; germ cell; sarcoma AB Germ cell tumors constitute a very complicated group of tumors of the ovary and their histogenesis is not yet clarified. Besides their histological heterogeneity, sarcomatous areas have also been described. A right ovarian mass was found in a 23-year-old female, who was being treated in the hospital for miscarriage. Disseminated omental metastases were detected during abdominal laparotomy. Pathological examination of the dissected material revealed the tumor to be a mixed germ cell tumor (immature teratoma and dysgerminoma) with sarcomatous component. Areas resembling granulosa cell tumor were also encountered. This ovarian tumor with many different histopathological features is presented with a review of the literature. The importance of thorough sampling in determining the type and extent of the malignant components is also emphasized due to their direct relation with the prognosis. C1 [Kabukcuoglu, Fevziye] Sisli Etfal Training and Research Hospital, Department of Pathology, Manastirli Ismail Hakki sk No:10/1 Dogancilar/UskudarIstanbul, Turkey. [Sungun, Aysim] Sisli Etfal Training and Research Hospital, Department of Pathology, Manastirli Ismail Hakki sk No:10/1 Dogancilar/UskudarIstanbul, Turkey. [Senturk, Akan Billur] Sisli Etfal Training and Research Hospital, Department of Pathology, Manastirli Ismail Hakki sk No:10/1 Dogancilar/UskudarIstanbul, Turkey. [Evren, Ismail] Sisli Etfal Training and Research Hospital, Department of Pathology, Manastirli Ismail Hakki sk No:10/1 Dogancilar/UskudarIstanbul, Turkey. [Ilhan, Ridvan] Sisli Etfal Training and Research Hospital, Department of Pathology, Manastirli Ismail Hakki sk No:10/1 Dogancilar/UskudarIstanbul, Turkey. RP Kabukcuoglu, F (reprint author), Sisli Etfal Training and Research Hospital, Department of Pathology, Istanbul, Turkey. EM fkabukcuoglu@hotmail.com CR Ahmed T, Bosl GJ, Hadju S: Teratoma with malignant transformation in germ cell tumors in men. Cancer 56:860-863, 1985. Akhtar M, Bakri Y, Rank F: Dysgerminoma of the ovary with rhabdomyosarcoma. Cancer 64:2309-2312, 1989. Bolen JW: Mixed germ cell-sex cord stromal tumor. Am J Clin Pathol 75:565-573, 1981. Kurman RJ, Norris HJ: Malignant mixed germ cell tumors of the ovary. A clinical and pathologic analysis of 30 cases. Obstet Gynecol 48:579-589, 1976. Mostofi FK, Sabin LH: Histological typing of testis tumors. Geneva, WHO 1977. Nogales F: Germ cell tumors of the ovary, In: Obstetrical and Gynaecological Pathology., Eds: Fox H and Wells M): 4th ed, Churchill Livingstone, New York, 1995, pp 847-896. Radford DM, Johnson FE, Janney CG: Sarcomatous change in a teratoma after treatment of testicular carcinoma. Cancer 68:395-399, 1991. Rishi M, Howard LN, Bratthauer GL, Tavassoli FA: Use of monoclonal antibody against human inhibin as a marker for sex cord-stromal tumors of the ovary. Am J Surg Pathol 2185:583- 589, 1997. Talerman A: Germ cell tumors of the ovary. In Blaustein’s Pathology of the Female Genital Tract.(Ed: Kurman RJ), 4th ed, Springer-Verlag, New York 1994, pp 849-914. Terrier-Lacombe MJ, Martinez-Madrigal F, Porta W, et al: Embryonal rhabdomyosarcoma arising in a mature teratoma of the testis: a case report. J Urol 143:1232-1234, 1990. Ulbright TM: Germ cell Neoplasms of the testis. Am J Surg Pathol 17:1075-1091, 1993. Ulbright TM, Clark SA, Einhorn LH: Angiosarcoma associated with germ cell tumors. Hum Pathol 16:268-272, 1985. Ulbright TM, Loehrer PJ, Roth LM, et.al: The development of non-germ cell malignancies within germ cell tumors. A clinicopathologic study of 11 cases. Cancer 54:1824-1833, 1984. Yamashita K, Yamoto M, Shikone T, et al: Production of inhibin A and inhibin B in human ovarian sex cord stromal tumors. Am J Obstet Gynecol 177: 1450-1457, 1997. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 60 EP 62 PG 3 ER PT J AU Cappello, F Bellafiore, M Bucchieri, F Balsano, G Palma, A Zummo, G AF Cappello, Francesco Bellafiore, Marianna Bucchieri, Fabio Balsano, Giuseppe Palma, Antonio Zummo, Giovanni TI Poorly Differentiated Synovial Sarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE synovial sarcoma; poorly differentiated synovial sarcoma; soft tissue tumors; calretinin; apoptosis ID synovial sarcoma; poorly differentiated synovial sarcoma; soft tissue tumors; calretinin; apoptosis AB Poorly differentiated synovial sarcoma is a rare soft tissue tumor. We studied a case arising in the pleural cavity of a young subject, characterised by the presence of spindle cell, small cell, and large epithelioid cell areas. We performed stains for mucosubstances and analysed the expression of cytokeratins 5/6, 7, 8, 18, 19, CEA, CD34, Ber-Ep4 and calretinin to characterize the phenotype of this neoplasm. We furthermore assessed immunohistochemically the presence of p53, Bcl-2, Bax and caspase 3, four apoptotic markers, to evaluate a relationship between apoptotic activity and the behaviour of this tumor. Our findings showed a strong presence of calretinin, p53 and Bcl-2 in all three areas. The possibility that poorly differentiated synovial sarcoma could be calretinin-positive was a new data, to our knowledge, and it could be of some importance in diagnostic pathology. Moreover, the negligible positivity for Bax and caspase 3 suggested that the minor role of programmed cell death could be one of the causes of the aggressive behaviour of this tumor. These data also suggest that the reduction of apoptotic phenomena in poorly differentiated synovial sarcoma could be considered one of the major mechanisms of tumoral growth. C1 [Cappello, Francesco] University of Palermo, Institute of Pathological Anatomy, Via Alla Falconara 120, 90136 Palermo, Italy. [Bellafiore, Marianna] University of Palermo, Institute of Human AnatomyPalermo, Italy. [Bucchieri, Fabio] University of Palermo, Institute of Human AnatomyPalermo, Italy. [Balsano, Giuseppe] University of Palermo, Institute of General and Endoscopic SurgeryPalermo, Italy. [Palma, Antonio] University of Palermo, Institute of Human AnatomyPalermo, Italy. [Zummo, Giovanni] University of Palermo, Institute of Human AnatomyPalermo, Italy. RP Cappello, F (reprint author), University of Palermo, Institute of Pathological Anatomy, 90136 Palermo, Italy. EM FRANCAPP@HOTMAIL.COM CR Abenoza P, Manivel JC, Swanson PE, et al: Synovial sarcoma: ultrastructural study and immunohistochemical analysis by a combined peroxidase-antiperoxidase/avidin-biotin complex procedure. Hum Pathol 17:1107-1115, 1986. Bissonnette RP, Echeverri F: Apoptotic cells induced by c-myc are inhibited by Bcl-2. Nature 359:552-554, 1992. Bubendorf L, Sauter G, Moch H, et al: Prognostic significance of Bcl-2 in clinically localized prostate cancer. Am J Surg Pathol 148:1557-1565, 1996. Chilosi M, Facchetti F, Dei Tos AP, et al: Bcl-2 expression in pleural and extrapleural solitary fibrous tumors. J Pathol 181:362-367, 1997. Dei Tos AP, Wadden C, Calonje E: Immunohistochemical demonstration of glycoprotein p30/30 MIC2, CD99, in synovial sarcoma. A potential cause of diagnostic confusion. Applied immunohistochemistry 3:168-173, 1995. Doglioni C, Dei Tos AP, Laurino L, et al: Calretinin: a novel immunocytochemical marker for mesothelioma. Am J Surg Pathol 20:1037-1046, 1996. Enzinger FM, Weiss SW: Soft tissue tumors. St. Louis, Mosby, 1995. Fisher C: Ultrastructural and immunohistochemical features of epithelial differentiation in monophasic and biphasic tumors. Hum Pathol 17:996-1008, 1986. Folpe AL, Schmidt RA, Chapman D, et al: Poorly differentiated synovial sarcoma. Immunohistochemical distinction from primitive neuroectodermal tumors and high grade malignant peripheral nerve sheath tumors. Am J Surg Pathol 22:673-682, 1998. Gaertner E, Zeren EH, Fleming MV, et al: Biphasic synovial sarcoma arising in the pleural cavity. A clinicopathologic study of five cases. Am J Surg Pathol 138:505-513, 1991. Hall PA, Levison DA: Biphasic tumor: clues to possible histogenesis in developmental processes. J Pathol 159:1-2, 1989. Joensuu H, Pylkkanen L, Toikkanen S: Bcl-2 protein expression and long term survival in breast cancer. Am J Pathol 145:1191- 1198, 1994. Kulig E, Jin L, Qian X, et al: Apoptosis in non-tumorous and neoplastic human pituitaries. Expression of the Bcl-2 family of proteis. Am J Pathol 154:767-774, 1999. Lu QL, Abel P, Foster CS, et al: Bcl-2: role in epithelial differentiation and oncogenesis. Hum Pathol 27:102-110, 1996. McPake CR, Tillman DM, Poquette CA, et al: Bax is an important determinant of chemosensitivity in pediatric tumor cell lines independent of Bcl-2 expression and p53 status. Oncol Res 10:235-244, 1998. Meis-Kindblom JM, Stenman G, Kindblom LG: Differential diagnosis of small round cell tumors. Semin Diagn Pathol 13:213-241, 1996. Nakanishi H, Ohsawa M, Naka N, et al: Immunohistochemical detection of Bcl-2 and p53 proteins and apoptosis in soft tissue sarcoma: their correlation with prognosis. Oncology 54:238- 244, 1997. Nicholson AG, Goldstraw P, Fisher C: Synovial sarcoma of the pleura and its differentiation from other primary pleural tumors: a clinicopathological and immunohistochemical review of three cases. Histopathology 33:508-513, 1998. Oltavi ZN, Milliman CL, Korsmeyer SJ: Bcl-2 hemerodimer in vivo with a conserved homologue, Bax, that accelerates programmed cell death. Cell 74:609-619, 1993. Ordonez NG, Mahfouz SM, MacKay B: Synovial sarcoma: an immunohistochemical and ultrastructural study. Hum Pathol 21:733-749, 1990. Ordonez NG: The immunohistochemical diagnosis of epithelial mesothelioma. Hum Pathol 30:313-323, 1999. Salisbury JR, Isaacson PG: Synovial sarcoma: an immunohistochemical study. J Pathol 147:49-57, 1985. Segers K, Ramael M, Singh Sk, et al: Immunoreactivity for Bcl- 2 protein in malignant mesothelioma and non-neoplastic mesothelium. Virchows Arch 424:631-634, 1994. Singer S, Baldini EH, Demetri GD, et al: Synovial sarcoma: prognostic significance of tumor size, margin sesection and mitotic activity for survival. J Clin Oncol 14:1201-1208, 1996. Suster S, Fisher C, Moran CA: Expression of Bcl-2 oncoprotein in benign and malign spindle cell tumors of soft tissue, skin, serosal surfaces and gastrointestinal tract. Am J Surg Pathol 22:863-872, 1998. Tormanen U, Eerole AK, Rainio P, et al: Enhanced apoptosis predicts shortened survival in non-small cell lung carcinoma. Cancer Res 55:595-602, 1995. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 63 EP 66 PG 4 ER PT J AU Girschick, JH Klein, R Scheurlen, GW Kuhl, J AF Girschick, J Hermann Klein, Rudiger Scheurlen, G Wolfram Kuhl, Joachim TI Cytogenetic and Histopathologic Studies of Congenital Supratentorial Primitive Neuroectodermal Tumors: A Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE PNET; primitive neuroectodermal tumor; brain neoplasm; congenital brain tumor; cytogenetic studies ID PNET; primitive neuroectodermal tumor; brain neoplasm; congenital brain tumor; cytogenetic studies AB Primitive neuroectodermal tumors (PNET) represent about 25% of primary central nervous system tumors in childhood, but congenital PNETs are rare. Cytogenetic studies and studies on molecular pathology have identified several genetic alterations in medulloblastoma, but molecular investigations on supratentorial PNETs are infrequent. We present a male newborn with a large congenital PNET of the right cerebral hemisphere and the molecular analysis of the tumor. Tumor tissue was investigated by routine histology and immunohistochemistry. Fluorescence in-situ hybridization was carried out on native tumor tissue to investigate deletions on chromosome 17p and to analyze c-Myc or N-Myc amplifications. Histologic examination revealed a primitive neuroectodermal tumor with massive extension covering almost the entire right hemisphere. Genetic analysis of the native tumor tissue of our patient excluded a deletion of chromosome 17p. An amplification of the c-Myc or N-Myc oncogene was absent using fluorescence in-situ hybridization. Despite unremarkable genetic analysis in our case prognosis was poor, suggesting that there are additional, yet unknown constitutional genetic aberrations in the pathogenesis of congenital supratentorial PNET. C1 [Girschick, J Hermann] University of Wurzburg, Children’s Hospital, Josef-Schneider-Str. 2, D-97080 Wurzburg, Germany. [Klein, Rudiger] University of Wurzburg, Institute of Pathology, Division of NeuropathologyWurzburg, Germany. [Scheurlen, G Wolfram] University of Mannheim, Children’s HospitalMannheim, Germany. [Kuhl, Joachim] University of Wurzburg, Children’s Hospital, Josef-Schneider-Str. 2, D-97080 Wurzburg, Germany. RP Girschick, JH (reprint author), University of Wurzburg, Children’s Hospital, D-97080 Wurzburg, Germany. EM Hermann.Girschick@mail.uni-wuerzburg.de CR Cohen BH, Zeltzer PM, Boyett JM, et al: Prognostic factors and treatment results for supratentorial primitive neuroectodermal tumors in children using radiation and chemotherapy: A childrens cancer group randomized trial. J Clin Oncol 13:1687- 1696, 1995. Cohen BH, Packer RJ: Chemotherapy for medulloblastomas and primitive neuroectodermal tumors. J Neurooncol 29:55-68, 1996. Di Rocco C, Iannelli A, Papacci F, et al: Prognosis of medulloblastoma in infants. Child’s Nerv Syst 13:388-396, 1997. Dirks PB, Harris L, Hoffmann HJ, et al: Supratentorial primitive neuroectodermal tumors in children. J Neurooncol 29:75- 84, 1996. Evans AE, Jenkin RD, Sposto R, et al: The treatment of medulloblastoma. Results of a prospective randomized trial of radiation therapy with and without CCNU, vincristine, and prednisone. J Neurosurg 72:572-582, 1990. Emadian SM, McDonald JD, Gerken SC, et al: Correlation of 17p loss with clinical outcome in medulloblastomas. Clin Cancer Res 2:1559-1564, 1996. Giangaspero F, Bigner SH, Giordana MT, et al: Medulloblastoma. In: Pathology and Genetics of Tumors of the Nervous System., Eds: Kleihues P, Cavenee WK), International Agency for Research on Cancer, Lyon, 1997, pp. 96-103. Haddad SF, Menezes AH, Bell WE: Brain tumors occuring before 1 year of age: a retrospective review of 22 cases in an 11-year period, 1977-1987). Neurosurgery 29:8-13, 1991. Janisch W, Haas JF, Schreiber D, et al: Primary central nervous system tumors in stillborn and infants. Epidemiological considerations. J Neuroonocol 2:113-116, 1984. Janss AJ, Yachnis AT, Silber JH, et al: Glial differentiation predicts poor clinical outcome in primitive neuroectodermal brain tumors. Ann Neurol 39:481-489, 1996. Joos S, Falk MH, Lichter P, et al: Variable breakpoints in Burkitt lymphoma cells with chromosomal t(8;14, translocation separate c-Myc and the IgH locus up to several hundred kb. Hum Mol Genet 1:625-632, 1992. Krischer JP, Ragab AH, Kun L: Nitrogen mustard, vincristine, procarbazine, and prednisone as adjuvant chemotherapy in the treatment of medulloblastoma. A pediatric oncology group study. J Neurosurg 74:905-909, 1991. Lichter P, Tang CJ, Call K, et al: High-resolution mapping of human chromosome 11 by in situ hybridization with cosmid clones. Science 247:64-69, 1990. Moriuchi S, Shimizu K, Miyao Y, et al: An immunohistochemical analysis of medulloblastoma and PNET with emphasis on NMyc protein expression. Anticancer Res 16:2687-2692, 1996. Neumann E, Kalousek DK, Norman MG, et al: Cytogenetic analysis of 109 pediatric central nervous system tumors. Cancer Genet Cytogenet 71:40-49, 1993. Ohgaki H, Eibl RH, Wiestler OD, et al: p53 mutations in nonastrocytic human brain tumors. Cancer Res 51:6202-6205, 1991. Packer RJ, Sutton LN, Goldwein JW, et al: Improved survival with the use of adjuvant chemotherapy in the treatment of medulloblastoma. J Neurosurg 74:433-440, 1991. Raffel C, Gilles FE, Weinberg KI: Reduction to Homozygosity and Gene amplification in central nervous system primitive neuroectodermal tumors of childhood. Cancer Res 50:587-591, 1990. Rorke LB, Trojanowski JQ, Lee VM: Primitive neuroectodermal tumors of the central nervous system. Brain Pathology 7:765- 784, 1997. Rostomily RC, Bermingham-McDonogh O, Berger MS, et al: Expression of neurogenic basic helix-loop-helix genes in primitive neuroectodermal tumors. Cancer Res 57:3526-3531, 1997. Scheurlen WG, Krauss J, Kuhl J: No preferential loss of one parental allele of chromosome 17p13.3 in childhood medulloblastoma. Int J Cancer 63:372-374, 1995. Scheurlen W, Seranski P, Mincheva A, et al: High resolution deletion mapping of chromosome arm 17p in childhood primitive neuroectodermal tumors, PNET, reveals a common chromosomal disruption within the Smith-Magenis region, an unstable region in band 17p11.2. Genes Chromosomes Cancer 18:50-58, 1997. Scheurlen WG, Schwabe GC, Joos S, et al: Molecular analysis of childhood primitive neuroectodermal tumors defines markers associated with poor outcome. J Clin Oncol 16:2478-2485, 1998. Scheurlen WG, Sorensen N, Roggendorf W, et al: Molecular analysis of medulloblastomas occuring simultaneously in monocygotic twins. Eur J Pediatr 155:880-884, 1996. Schutz BR, Scheurlen W, Krauss J, et al: Mapping of chromosomal gains and losses in primitive neuroectodermal tumors by comparative genomic hybridization. Genes Chromosomes Cancer 16:196-203, 1996. Tait DM, Thornton-Jones H, Bloom HJ, et al: P: Adjuvant chemotherapy for medulloblastoma: the first multi-center control trial of the International Society of Pediatric Oncology, SIOP I). Eur J Cancer 26:464-469, 1990. Tomita T, McLone DG, Yasue M: Cerebral primitive neuroectodermal tumors in childhood. J Neurooncol 6:233-243, 1988. Wasson JC, Saylors III RL, Zeltzer P, et al: Oncogene Amplification in pediatric brain tumors. Cancer Res 50:2987-2990, 1990. Wilgenbus K, Coy J, Mincheva A, et al: Ordering of 66 STSs along the entire short arm of human chromosome 17 and chromosome assignment of a transscribed sequence, FMR1L2, homologous to FMR1. Cytogenet Cell Genet 73:240-243, 1992. Wilgenbus K, Mincheva A, Korn B: IRS-long range PCR: a simple method for amplification of human genomic DNA from complex sources. Meth Mol Cell Biol 5:214-221, 1995. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 67 EP 71 PG 5 ER PT J AU Repassy, LD Csata, S Sterlik, G Ivanyi, A AF Repassy, L Denes Csata, Sandor Sterlik, Gabor Ivanyi, Andras TI Giant Adrenal Myelolipoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE adrenal gland; tumor; myelolipoma ID adrenal gland; tumor; myelolipoma AB Authors present a case of giant adrenal myelolipoma, where the tumor was hormonally inactive but caused abdominal and flank pain. The huge tumor, a 20x18x10 cm mass, was surgically removed. The ipsilateral kidney was preserved. C1 [Repassy, L Denes] Saint Stephen Hospital, Department of Urology, Nagyvarad ter 1., H-1096 Budapest, Hungary. [Csata, Sandor] Saint Stephen Hospital, Department of Urology, Nagyvarad ter 1., H-1096 Budapest, Hungary. [Sterlik, Gabor] Saint Stephen Hospital, Department of Urology, Nagyvarad ter 1., H-1096 Budapest, Hungary. [Ivanyi, Andras] Municipal Hospital, Del-Pest, Department of PathologyBudapest, Hungary. RP Repassy, LD (reprint author), Saint Stephen Hospital, Department of Urology, H-1096 Budapest, Hungary. CR Albala DM, Chung CJ, Sueoka BL, et al: Hemorrhagic myelolipoma of adrenal gland after blunt trauma. Urology 38:559, 1991. Meaglia JP, Schmidt JD: Natural history of an adrenal myelilipoma. J Urol 147:1089, 1992. Goldman HB, Howard RC, Patterson L: Spontenaous retroperitoneal hemorrhage from a giant adrenal myelolipoma. J Urol 155:639, 1995. Lam KY: Lipomatous tumors of the adrenal gland. J Urol Pathol 3:95, 1995. Gould JD, Mitty HA, Pertsemlidis D, et al: Adrenal myelilipoma: diagnosis by fine needly aspiration. Am J Radiol 148:921, 1987. Bennett BD, McKenna RJ, Hough AI: Adrenal myelilipoma associated with Cushing’s disease. Am J Clin Pathol 73:443, 1980. Olsson CA, Krane RJ, Klugo RC, et al: Adrenal myelolipoma. Surgery 73:665-670, 1973. Desai SB, Dourmashkin L, Kabakow BR, et al: Myelolipoma of the adrenal gland: case report. literature review and analysis of diagnostic features. Mt Sinai J Med. 46:155-159, 1979. Vick CW, Zeman RK, Mannes E, et al.: Adrenal myelolipoma: CT and ultrasound findings. Urol Radiol 6:7-13, 1984. Newmann P, Silen W: Myelolipoma of the adrenal gland. Arch Surg 97:637, 1968. Gierke E: Ueber Knochenmarksgewebe in der Nebenniere. Beitr Path Anat. 7:311, 1905. Oberling, C: Les formations myelo-lipomateuses. Bull Assoc Franc Cancer 18:234, 1929. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 72 EP 73 PG 2 ER PT J AU Sharma, ChM Vaish, S Arora, R Gaikwad, S Sarkar, Ch AF Sharma, Chand Mehar Vaish, Sandeep Arora, Reena Gaikwad, Sailesh Sarkar, Chitra TI Composite Pituitary Adenoma and Intrasellar Tuberculoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE adenoma; endocrine; pituitary gland; sella; tuberculoma ID adenoma; endocrine; pituitary gland; sella; tuberculoma AB Tuberculous involvement of the pituitary gland is rare. We report a unique case of a composite lesion consisting of pituitary adenoma and intrasellar tuberculoma. A 24-year-old lady presented with features of acromegaly and amenorrhea. Serum growth hormone levels were found to be raised. Radiological investigations were consistent with a pituitary adenoma. Decompression of the lesion was done through trans-sphenoidal approach. Histological examination revealed a growth hormone secreting pituitary adenoma in association with a granulomatous lesion suggesting of pituitary tuberculoma. No other evidence of tuberculosis was found in the brain or spinal cord. This type of dual pathology has been reported only once in the earlier literature. C1 [Sharma, Chand Mehar] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Vaish, Sandeep] All India Institute of Medical Sciences, Department of NeurosurgeryNew Delhi, India. [Arora, Reena] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Gaikwad, Sailesh] All India Institute of Medical Sciences, Department of NeuroradiologyNew Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. RP Sarkar, Ch (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM sarkarch@medinst.ernet.in CR Ashkan K, Papadopoulos MC, Casey AT, et al: Sellar tuberculoma: report of twocase. Acta Neurochir, Wien, 139: 523-525, 1997. Bhagwati SN: Intracranial tuberculoma. Neurology India 34:161-163, 1996. Brooks MH, Dunlao JS, Bronsky D, et al: Hypophysial tuberculoma with hypopituitarism. Am J Med 54:777-781, 1973. Coleman CC, Meredith JM: Diffuse tuberculosis of the pituitary gland simulating tumor with postoperative recovery. Arch Neurol Psychiatry 44:1076-1085, 1940. Dastur HM: Tuberculoma, Vinken PJ, Bruyn GW [eds]. Handbook of clinical neurology, New York, American Elsevier Publishing Co., Vol. 18, pp 413-426, 1975. DeAngelis LM: Intracranial tuberculoma: case report and review of the literature. Neurology 31: 133-1136, 1981. Delsedine M, Aguggia M, Cantello R: Isolated hypophyseal tuberculoma : case report. Clin Neuropathol 7:311-313, 1988. Eckland DJA, O’ Neil JH, Lightman SL: A pituitary tuberculoma. J Neurol Neurosurg Psychia 50:360-61, 1987. Esposito V, Fraioli B, Ferrante L, Palma L: Intrasellar tuberculoma: case report. Neurosurg 21:721-723, 1987. Gazioglu N, AK II, Oz B, et al: Silent pituitary tuberculoma associated with pituitary adenoma. Act Neurochir, Wien, 141:785-786, 1999. Ghosh S, Chandy MJ: Intrasellar tuberculoma case report. Clin Neurol Neurosurg 94:251-252, 1992. Maurice-Williams RS: Tuberculomas of the brain in Britain. J Postgrad Med 48:679-681, 1972. Pereira J, Vaz R, Carvalho D, et al: Thickening of pituitary stalk: A finding suggestive of intrasellar tuberculoma? Case report. Neurosurg 36:1013-1016, 1995. Petrossians P, Delvenne P, Flandroy P, et al: An unusual pituitary pathology. J Clin Endo Metabolism 83:3454-3459, 1998. Ranjan A, Chandy MJ: Intrasellar tuberculoma. Brit J Neurosurg 8:179-185, 1994. Rohmer V, Chanson P, Dupas B, et al: Intrasellar non-adenomatous expansive process. Ann endocrinol, Paris, 58:11-19, 1997. Sharma MC, Arora R, Mahapatra AK, et al: Intrasellar tuberculoma – an enigmatic pituitary infection: a series of 18 cases. Clin Neurol and Neurosurg 102:72-77;2000. Taparia SC, Tyagi G, Singh AK, et al: Sellar tuberculoma. J Neurol Neurosurg Psychiatr 55:629, 1992. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2001 VL 7 IS 1 BP 74 EP 76 PG 3 ER PT J AU Timar, J Dome, B Fazekas, K Janovics, Paku, S AF Timar, Jozsef Dome, Balazs Fazekas, Karoly Janovics, Agnes Paku, Sandor TI Angiogenesis-Dependent Diseases and Angiogenesis Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE angiogenesis; diseases; pro-angiogenesis; anti-angiogenesis ID angiogenesis; diseases; pro-angiogenesis; anti-angiogenesis AB The discovery of the molecular mechanisms of physiological vasculogenesis and pathological angiogenesis helped to recognize two classes of diseases: one where the therapeutic angiogenesis can repair the tissue damages (arteriosclerosis, myocardial infarction, limb ischemia) and the other one where inhibition of pathological angiogenesis can cure the disease or delay its progression (retinopathies, benign and malignant angiogenic tumors, progression of malignant tumors). Although there are an exponentially growing number of new synthetic molecules characterized mainly by antiangiogenic properties, the discovery of a large battery of natural pro- and anti-angiogenic factors suggests that this may provide a more physiological approach to treat both class of angiogenesis-dependent diseases in the near future. C1 [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gy. U. 7-9., H1122 Budapest, Hungary. [Dome, Balazs] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Fazekas, Karoly] National Institute of Oncology, Department of Tumor Progression, Rath Gy. U. 7-9., H1122 Budapest, Hungary. [Janovics, Agnes] National Institute of Oncology, Department of Tumor Progression, Rath Gy. U. 7-9., H1122 Budapest, Hungary. [Paku, Sandor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H1122 Budapest, Hungary. CR Albini A, Benelli R, Presta M, et al: HIV-tat protein is a heparin binding angiogenic growth factor. Oncogene 12:289-297, 1996. Bae DG, Gho YS, Yoon WH, et al: Arginic-rich anti-vascular endothelial growth factor peptides inhibit tumor growth and metastasis by blocking angiogenesis. J Biol Chem 275:13588- 13596, 2000. Bowder T, Folkman J, Pirie-Shepherd S: The hemostatic system as a regulator of angiogenesis. J Biol Chem 275:1521-1524, 2000. Brown JM: Exploiting the hypoxic cancer cell: mechanisms and therapeutic strategies. Molecular Medicine Today 6:157-162, 2000. Bussolino F, Albini A, Camussi G, et al: Role of soluble mediators in angiogenesis. Eur J Cancer 32A:2401-2412, 1996. Carmeliet P: Mechanisms of angiogenesis and arteriogenesis. Nature Medicine 6:389-395, 2000. Chang YS, di Tomaso E, McDonald DM, et al: Mosaic blood vessels in tumors: frequency of cancer cells in contact with flowing blood. Proc Natl Acad Sci USA 97:14608-14613, 2000. DeNardo SJ, Burke PA, Leigh BR, et al: Neovascular targeting with cycle RGD peptide, cRGDf-ACHA, to enhance delivery of radioimmunotherapy. Cancer Biotherapy Radiopharmaceuticals 15:71-79, 2000. Elicieri BP, Cheresh DA:The role ofav integrins during angiogenesis: insights into potential mechanisms of action and clinical development. J Clin Invest 103:1227-1230, 1999. Fidler IJ, Ellis LM: The implications of angiogenesis for the biology and therapy of cancer metastasis. Cell 79:185-188, 1994. Folkman J: Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med 1:27-31, 1995. Gately S: The contributions of cyclooxygenase-2 to tumor angiogenesis. Cancer Metast Reviews 19:19-27, 2000. Hanahan D, Folkman J: Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86:353-364, 1996. Hiltunen MO, Turunen MP, Laitinen M, et al: Insights into the molecular pathogenesis of atherosclerosis and therapeutic strategies using gene transfer. Vasc Med 5:41-48, 2000. Holash J, Maisonpierre PC, Compton D, et al: Vessel cooption, regression and growth in tumors mediated by angiopoietins and VEGF. Science 284:1994-1998, 1999. Holmgren L, O’Reilly MS, Folkman J: Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1:149-153, 1995. Isner JM, Asahara T: Angiogenesis and vasculogenesis as therapeutic strategies for postnatal neovascularization. J Clin Invest 103:1231-1235, 1999. Jacobson BS: Hereditary hemorrhagic telangiectasia a model for blood vessel growth and enlargement. Am J Path 156:737- 742, 2000. Jimenez B, Volpert OV, Crawford SE et al: Signals leading to apoptosis-dependent inhibition of neovascularization by thrombospondin- 1. Nature Medicine 6:41-48, 2000. Kerbel RS: Tumor angiogenesis: past, present and the near future. Carcinogenesis 21:505-515, 2000. Lee P, Wang CC, Adamis AP: Ocular neovascularization: an epidemiologic review. Surv Ophthalmol 43:245-269, 1998. Li WW, Jaffe M, Li VW, et al: Lessons to be learned from clinical trials of angiogenesis modulators in ischemic diseases. In G.M. Rubanyi,, eds.), Marcel Dekker, Inc. New York, Basel, 2000, pp. 519-536. Li WW: Tumor angiogenesis: molecular pathology, therapeutic targeting and imaging. Acad Radiol 7:800-811, 2000. Luzzi KJ, Varghese HJ, MacDonald IC, et al: Arginie-rich antivascular endothelial growth factor peptides inhibit tumor growth and metastasis by blocking angiogenesis. J Biol Chem 4:373-379, 1998/1999. Malonne H, Langer I, Kiss R, et al: Mechanisms of tumor angiogenesis and therapeutic implications: angiogenesis inhibitors. Clin exper Metastasis 17:1-14, 1999. Maniotis AJ, Folberg R, Hess A, et al: Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 155:739-752, 1999. Nicolaou KC, Trujilo JI, Jandeleit B, et al: Design, synthesis and biological evaluation of nonpeptide integrin antagonists. Bioorg Med Chem 6:1185-1208, 1998. Nie D, Lamberti M, Zacharek A, et al: Thromboxane A(2, regulation of endothelial cell migration, angiogenesis, and tumor metastasis. Biochem Biophys Res Commun 267:245-251, 2000a. Nie D, Tang K, Diglio C, et al: Eicosanoid regulation of angiogenesis: role of endothelial arachidonate 12-lipoxygenase. Blood 95:2304-2311, 2000b. O’Reilly MS, Holmgren L, Shing Y, et al: Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell 79:315-328, 1994. O’Reilly MS, Boehm T, Shing Y, et al: Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell 88:277-285, 1997. Paku S: Current concepts of tumor-induced angiogenesis. Pathol Oncol Res 4:62-75, 1998. Pribluda VS, Gubish ER, LaVallee, TM. Jr, et al: Methoxyestradiol: An endogenous antiangiogenic and antiproliferative drug candidate. Cancer Metast Reviews 19:173-179, 2000. Sasisekharan R, Moses MA, Nugent MA, et al: Heparinase inhibits neovascularization. Proc Nat Acad Sci USA 91:1524-1528, 1994. Schiller J: Early trials of antiangiogenic agents. Anticancer Agents in Oncology 15:3-10, 1999. Schlaeppi J-M, Wood JM: Targeting vascular endothelial growth factor, VEGF, for anti-tumor therapy, by anti-VEGF neutralizing monoclonal antibodies or by VEGF receptor tyrosine-kinase inhibitors. Cancer Metastasis Rev 18:473-381, 1999. Semenza GL: HIF-1: Using two hands to flip the angiogenic switch. Cancer Metast Reviews 19:59-65, 2000. Stephens TD, Fillmore BJ: Hypothesis: Thalidomine embryopathy- proposed mechanism of action. Teratology 61:189-195, 2000. Stetler-Stevenson WG: Matrix metalloproteinases in angiogenesis: a moving target for therapeutic intervention. J Clin Invest 103:1237-1241, 1999. Thompson WD, Li WW, Maragoudakis M: The clinical manipulation of angiogenesis: pathology, side-effects, surprises, and opportunities with novel human therapies. J Pathol 190:330-337, 2000. Timar J, Toth J: Tumor sinuses – vascular channels. Pathol Oncol Res 6:83-86, 2000. Venkov CD, Rankin AB, Vaughan DE: Identification of authentic estrogen receptor in cultured endothelial cells. A potential mechanism for steroid hormone regulation of endothelial function. Circulation 94:727-733, 1996. Xu C, Lee S, Singh TM, et al: Molecular mechanisms of aortic wall remodeling in response to hypertension. J vasc Surg 33:570-578, 2001. Yeh ChH, Peng H-Ch, Huang T-F: Accutin, a new desintegrin, inhibits angiogenesis in vitro and in vivo by acting as integrinavb3 antagonist and inducing apoptosis. Blood 92:3268-3276, 2000. Zhang M, Volpert O, Shi YH, et al: Maspin is an angiogenesis inhibitor. Nature Medicine 6:196-199, 2000. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 85 EP 94 PG 10 ER PT J AU Petak, I Houghton, AJ AF Petak, Istvan Houghton, A Janet TI Shared Pathways: Death Receptors and Cytotoxic Drugs in Cancer Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE apoptosis; tumor; death receptor; Fas; drug; p53 ID apoptosis; tumor; death receptor; Fas; drug; p53 AB Death ligands (TNF, FasL, TRAIL) and their respective death receptor signaling pathways can be used to induce tumor cells to undergo apoptosis. Chemotherapeutic drugs can induce apoptosis and the upregulation of death ligands or their receptors. Downstream events following cytotoxic stress-induced DNA damage and the signaling pathways that lead to the induction of apoptosis may be either dependent or independent of death receptor signaling. The involvement of the Fas signaling pathway in chemotherapy-induced apoptosis has been the most extensively studied, with the current emergence of information on the TRAIL signaling pathway. Fas-mediated and chemotherapy-induced apoptosis can converge at the level of the receptor, FasL, DISC formation, activation of the initiator caspase-8, at the level of the mitochondria, or at the level of downstream effector caspase activation. Convergence is influenced by the specific form of DNA damage, the cellular environment, and the specific pathway(s) by which death receptor-mediated or drug-mediated apoptosis are induced. This review discusses the different levels of interaction between signaling pathways in the different forms of cell death. C1 [Petak, Istvan] St. Jude Children's Research Hospital, Department of Hematology-Oncology, Division of Experimental Hematology, 332 North Lauderdale, 38105 Memphis, TN, USA. [Houghton, A Janet] St. Jude Children's Research Hospital, Department of Hematology-Oncology, Division of Experimental Hematology, 332 North Lauderdale, 38105 Memphis, TN, USA. RP Houghton, AJ (reprint author), St. Jude Children's Research Hospital, Department of Hematology-Oncology, Division of Experimental Hematology, 38105 Memphis, USA. EM janet.houghton@stjude.org CR Ashkenazi A, Dixit VM: Death receptors: Signaling and modulation. Science 281:305-108, 1998. Ware CF, VanArsdale S, VanArsdale TL: Apoptosis mediated by the TNF-related cytokine and receptor families. J Cell Biochem 60:47-55, 1996. McGeehan GM, Becherer JD, Bast RCJr, et al: Regulation of tumor necrosis factor-alpha processing by a metalloproteinase inhibitor. Nature 370:558-561, 1994. Tanaka M, Itai T, Adachi M, et al: Downregulation of Fas ligand by shedding. Nature Med 4:31-36, 1998. Kayagaki N, Kawasaki A, Ebata T, et al: Metalloproteinasemediated release of human Fas ligand. J Exp Med 182:1777- 1783, 1995. Ashkenazi A, Dixit VM: Apoptosis control by death and decoy receptors. Curr Opin Cell Biol 11:255-260, 1999. Nagata S, Golstein P: The Fas death factor. Science 276:1449, 1995. Arai H, Gordon D, Nabel EG, et al: Gene transfer of Fas ligand induces tumor regression in vivo. Proc Natl Acad Sci USA 94:13862-13867, 1997. French LE, Hahne M, Viard I, et al: Fas and Fas ligand in embryos and adult mice: ligand expression in several immuneprivileged tissues and coexpression in adult tissues characterized by apoptotic cell turnover. J Cell Biol 133:335-343, 1996. Moller P, Koretz K, Leithauser F, et al: Expression of Apo-1, CD95), a member of the NGF/TNF receptor superfamily, in normal and neoplastic colon epithelium. Int J Cancer 57:371- 377, 1994. Pitti RM, Marsters SA, Lawrence DA, et al: Genomic amplification of a decoy receptor for Fas ligand in lung and colon cancer. Nature. 396:699-703. 1998. Itoh N, Yonehara S, Ishii A, et al: The polypeptide encoded by the cDNA for human cell surface antigen Fas can mediate apoptosis. Cell 66:233-243, 1991. Boldin MP, Varfolomeev EE, Pancer Z, et al: A novel protein that interacts with the death domain of Fas/APO1 contains a sequence motif related to the death domain. J Biol Chem 270:7795-7798, 1995. Chinnaiyan AM, O’Rourke K, Tewari M, et al: FADD, a novel death domain-containing protein, interacts with the death domain of Fas and initiates apoptosis. Cell 81:505-512, 1995. Boldin MP, Goncharov TM, Goltsev YV, et al: Involvement of MACH, a novel MORT1/FADD-interacting protease, in Fas/APO1 and TNF receptor-induced cell death. Cell 85:803- 815, 1996. Medema JP, Scaffidi C, Kischkel FC, et al: FLICE is activated by association with the CD95 death-inducing signaling complex, DISC). EMBO J 16:2794-2804, 1997. Muzio M, Chinnaiyan AM, Kischkel FC, et al: FLICE a novel FADD-homologous ICE/CED-3-like protease is recruited to the CD95, Fas/Apo-1, death-inducing signaling complex. Cell 85:817-827, 1996. Irmler M, Thome M, Hahne M, et al: Inhibition of death receptor signals by cellular FLIP. Nature 388:190-195. 1997. Scaffidi C, Schmitz I, Krammer PH, et al: The role of c-FLIP in modulation of CD95-induced apoptosis. J Biol Chem 274:1541-1548. 1999. Scaffidi C, Fulda S, Srinivasan A, et al: Two CD95, APO- 1/Fas, signaling pathways. EMBO J 17:1675-1687, 1998. Sun X-M, MacFarlane M, Zhuang J, et al: Distinct caspase cascades are initiated in receptor-mediated and chemicalinduced apoptosis. J Biol Chem 274:5053-5060, 1999. Kuwana T, Smith JJ, Muzio M, et al: Apoptosis induction by caspase-8 is amplified through the mitochondrial release of cytochrome c. J Biol Chem. 27:16589-16594, 1998. Li P, Nijhawan D, Alnemri ES, Wang X: Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 91:479-489, 1997. Jeremias I, Herr I, Boehler T, et al: TRAIL/Apo-2-ligandinduced apoptosis in human T cells. Eur J Immunol 28:143- 152, 1998. Marsters SA, Pitti RM, Donahue CJ, et al: Activation of apoptosis by Apo-2 ligand is independent of FADD but blocked by CrmA. Curr Biol 6:750-752, 1996. Griffith TS, Lynch DH: TRAIL: a molecule with multiple receptors and control mechanisms. Curr Opin Immunol 10:559-563, 1998. Zhang XD, Franco AV, Nguyen T, et al: Differential Localization and Regulation of Death and Decoy Receptors for TNFRelated Apoptosis-Inducing Ligand, TRAIL, in Human Melanoma Cells. J Immunol 164:3961-3970, 2000. Walczak H, Miller RE, Ariail K, et al: Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nature Med 5:157-163, 1999. Walczak H, Degli-Eposti MA, Johnson RS, et al: TRAIL-R2: a novel apoptosis-mediating receptor for TRAIL. EMBO J 16:5386-5397, 1997. Wiley SR, Schooley K, Smolak PJ, et al: Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity 3:673-682, 1995. Pan G, O’Rourke K, Chinnaiyan AM, et al: The receptor for the cytotoxic ligand TRAIL. Science 276:111-113, 1997. Pan G, Ni J, Wei Y-F, et al. An antagonist decoy receptor and a death domain-containing receptor for TRAIL. Science 277:815-818, 1997. Sheridan JP, Marsters SA, Pitti PM, et al: Control of TRAILinduced apoptosis by a family of signaling and decoy receptors. Science 277:818-821, 1997. Degli-Eposti MA, Smolak PJ, Walczak H, et al: Cloning and characterization of TRAIL-R3, a novel member of the emerging TRAIL receptor family. J Exp Med 186:1165- 1170, 1997. Degli-Eposti MA, Dougall WC, Smolak PJ, et al: The novel receptor TRAIL-R4 induces NF-kB and protects against TRAIL-mediated apoptosis, yet retains an incomplete death domain. Immunity 7:813-820, 1997. Marsters SA, Sheridan JP, Pitti RM, et al:A novel receptor for Apo2L/TRAIL contains a truncated death domain. Curr Biol 7:1003-1006, 1997. Frank S, Kohler U, Schackert G, et al: Expression of TRAIL and its receptors in human brain tumors. Biochem Biophys Res Commun 257:454-459, 1999. Griffith TS, Chin WA, Jackson GC, et al. Intracellular regulation of TRAIL-induced apoptosis in human melanoma cells. J Immunol 161:2833-2840, 1998. Thomas WD, Hersey P: TNF-related apoptosis-inducing ligand, TRAIL, induces apoptosis in Fas ligand-resistant melanoma cells and mediates CD4 T cell killing of target cells. J Immunol 161:2195-2200, 1998. Ashkenazi A, Pai RC, Fong S, et al: Safety and antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest 104:155- 162, 1999. Rieger J, Naumann U, Glaser T, et al: APO2 ligand: a novel weapon against malignant glioma? FEBS Lett 427:124-128, 1998. Schneider P, Thome M, Burns K, et al: TRAIL receptors 1, DR4, and 2, DR5, signal FADD-dependent apoptosis and activate NF-kappaB. Immunity 7:831-6, 1997. 42. Schneider P, Thome T, Burns K, et al: TRAIL receptors 1, DR4, and 2, DR5, signal FADD-dependent apoptosis and activate NF-kappaB. Immunity 7:831-836,1997. Kischkel FC, Lawrence DA, Chuntharapai A, et al: Apo2L/TRAIL-dependent recruitment of endogenous FADD and caspase-8 to death receptors 4 and 5. Immunity 12:611- 620, 2000. Sprick MR, Weigand MA, Rieser E, et al: FADD/MORT1 and caspase-8 are recruited to TRAIL receptors 1 and 2 and are essential for apoptosis mediated by TRAIL receptor 2. Immunity 12:599-609, 2000. Bodmer JL, Holler N, Reynard S, et al: TRAIL receptor-2 signals apoptosis through FADD and caspase-8. Nat Cell Biol 2:241-243, 2000. Hickman JA: Apoptosis induced by anticancer drugs. Cancer Metast Rev 11:121-139, 1992. Holzman D: Apoptosis provides new targets for chemotherapy. J Natl Cancer Inst 88:1098-1100, 1996. Sellers WR, Fisher DE: Apoptosis and cancer drug targeting. J Clin Invest 104:1655-1661, 1999. Miyashita T, Reed JC: Bcl-2 oncoprotein blocks chemotherapy- induced apoptosis in a human leukemia cell line. Blood 81:151-157, 1993. Bunz F, Hwang PM, Torrance C, et al: Disruption of p53 in human cancer cells alters the responses to therapeutic agents. J Clin Invest 104:263-269, 1999. Zhu H, Fearnhead HO, Cohen GM: An ICE-like protease is a common mediator of apoptosis induced by diverse stimuli in human monocytic THP.1 cells. FEBS LETT 375:303-308, 1995. Petak I, Mihalik R, Bauer PI, et al: BCNU is a caspase-mediated inhibitor of drug-induced apoptosis. Cancer Res 58:614- 618, 1998. Petak I, Tillman, DM, Harwood FG, et al: Fas-dependent and -independent mechanisms of cell death following DNA damage in human colon carcinoma cells. Cancer Res 60:2643- 2650. 2000. Tounekti O, Pron G, Belehradek J, et al: Bleomycin, an apoptosis- mimetic drug that induces two types of cell death depending on the number of molecules internalized. Cancer Res 53:5462-5469, 1993. Skladanowski A, Konopa, J: Adriamycin and daunomycin induce programmed cell death, apoptosis, in tumour cells. Biochem Pharmacol 46:375-382, 1993. Pellicciari C, Bottone MG, Schaack V, et al: Etoposide at different concentrations may open different apoptotic pathways in thymocytes. Eur J Histochem 40:289-298, 1996. Lock RB, Stribinskiene L: Dual modes of death induced by etoposide in human epithelial tumor cells allow Bcl-2 to inhibit apoptosis without affecting clonogenic survival. Cancer Res 56:4006-4012, 1996. Kasibhatla S, Brunner T, Genestier L, et al: DNA damaging agents induce expression of Fas ligand and subsequent apoptosis in T lymphocytes via the activation of NF-kappa B and AP-1. Mol Cell 1, 543-551. 1998. Faris M, Kokot N, Latinis K, et al: The c-Jun N-terminal kinase cascade plays a role in stress-induced apoptosis in Jurkat cells by up-regulating Fas ligand expression. J Immunol 160:134-144, 1998. Friesen C, Herr I, Krammer PH, et al: Involvement of the CD95, APO-1/Fas, receptor/ligand system in drug-induced apoptosis in leukemia cells. Nature Med 2:574-577, 1996. Fulda S, Sievens H, Frieson C, et al: The CD95, APO1-Fas, system mediates drug-induced apoptosis in neuroblastoma cells. Cancer Res 57:3823-3829, 1997. Villunger A, Egle A, Kos M, et al: Drug-induced apoptosis is associated with enhanced Fas, APO-1/CD9J, ligand expression but occurs independently of Fas, APO-1/CD95, signaling in human T-acute lymphatic leukemia cells. Cancer Res 57:3331-3334, 1997 Eischen CM, Kottke TJ, Martins LM, et al: Comparison of Apoptosis in wild-type and Fas-resistant cells: chemotherapyinduced apoptosis is not demendent on Fas/Fas ligand interactions. Blood 90:935-943, 1997. Wen J, Ramadevi N, Nguyen D, et al: Antileukemic drugs increase death receptor 5 levels and enhance apo-2L-induced apoptosis of human acute leukemia cells. Blood 96:3900-3906, 2000. Nagane M, Pan G, Weddle JJ, et al: Increased death receptor 5 expression by chemotherapeutic agents in human gliomas causes synergistic cytotoxicity with tumor necrosis factorrelated apoptosis-inducing ligand in vitro and in vivo. Cancer Res 60:847-853, 2000. Keane MM, Ettenberg SA, Nau MM, et al: Chemotherapy augmants TRAIL-induced apoptosis in breast cell lines. Cancer Res 59:734-741, 1999. Alderson MR, Tough TW, Davissmith T, et al: Fas ligand mediates activation-induced cell death in human T lymphocytes. J Exp Med 181:71-77, 1995, Brunner T, Mogil RJ, LaFace D, et al: Cell-autonomous Fas, CD95)/Fas-ligand interaction mediates activation-induced apoptosis in T-cell hybridomas. Nature 373:441-444, 1995. Fulda S, Strauss G, Meyer E,et al:Functional CD95 ligand and CD95 death-inducing signaling complex in activation-induced cell death and doxorubicin-induced apoptosis in leukemic T cells. Blood 95:301-308, 2000. Fulda S, Meyer E, Friesen C, et al: Cell type specific involvement of death receptor and mitochondrial pathways in druginduced apoptosis. Oncogene 20:1063-1075, 2001. Houghton JA, Harwood FG, Tillman DM: Thymineless death in colon carcinoma cells is mediated via Fas signaling. Proc Natl Acad Sci USA 94:8144-8149, 1997. Tillman DM, Petak I, Houghton JA: A Fas-dependent component in 5-fluorouracil/leucovorin-induced cytotoxicity in colon carcinoma cells. Clin Cancer Res 5:425-430. 1999. Eichhorst ST, Muller M, Li-Weber M, et al: A novel AP-1 element in the CD95 ligand promoter is required for induction of apoptosis in hepatocellular carcinoma cells upon treatment with anticancer drugs. Mol Cell Biol 20:7826-7837, 2000. Eichhorst ST, Muerkoster S, Weigand MA, et al: The chemotherapeutic drug 5-fluorouracil induces apoptosis in mouse thymocytes in vivo via activation of the CD95(APO-1/Fas, system. Cancer Res 61:243-248. 2001. Harwood FG, Kasibhatla S, Petak I, et al: Regulation of FasL by NF-kappaB and AP-1 in Fas-dependent thymineless death of human colon carcinoma cells. J Biol Chem 275:10023- 10029. 2000. Muller M, Wilder S, Bannasch D, et al: P53 activates the CD95, APO-1/Fas, gene in response to DNA damage by anticancer agents. J Exp Med., 188:2033-2045, 1998. Petak I, Tillman DM, Houghton JA: P53-dependence of Fas induction amd acute apoptosis in response to 5-fluorouracilleucovorin in human colon carcinoma cell lines. Clin Cancer Res 6:4432-4441, 2000. Siltonen T, Mantymaa P, Saily M, et al: Etoposide-induced apoptosis is not associated with the fas pathway in acute myeloblastic leukemia cells. Leukemia Res 24:281-288, 2000. Tolomeo M, Dusonchet L, Meli M, et al: The CD95/CD95 ligand system is not the major effector in anticancer drug-mediated apoptosis. Cell Death Diff 5:735-742, 1998. Shao R-G, Cao C-X, Nieves-Neira W, et al: Activation of the Fas pathway independently of Fas ligand during apoptosis induced by camptothecin in p53 mutant colon carcinoma cells. Oncogene 20:1852-1859, 2001. Wieder T, Essmann F, Prokop A, et al: Activation of caspase-8 in drug-induced apoptosis of B-lymphoid cells is independent of CD95/Fas receptor-ligand interaction and occurs downstream of caspase-3. Blood 97:1378-1387, 2001. Wesselborg S, Engels IH, Rossmann E, et al: Anticancer drugs induce caspase-8/FLICE activation and apoptosis in the absence of CD95 receptor/ligand interaction. Blood 93:3053- 3063, 1999. Tang D, Lahti JM, Kidd VJ: Caspase-8 activation and Bid cleavage contribute to MCF7 cellular execution in a caspase- 3-dependent manner during staurosporine-mediated apoptosis. J Biol Chem 275:9303-9307, 2000. Ferreira CG, Span SW, Peters GJ: Chemotherapy triggers apoptosis in a caspase-8-dependent and mitochondria-controlled manner in the non-small cell lung cancer cell line NCIH460. Cancer Res 60:7133-7141, 2000. Kinoshita H, Yoshikawa H, Shiiki K, et al: Cisplatin, CDDP, sensitizes human osteosarcoma cells to Fas/CD95-mediated apoptosis by downregulating FLIP-L expression. In J Cancer 88:986-991, 2000. Tang D, Lahti JM, Grenet J, et al: Cycloheximide-induced Tcell death is mediated by a Fas-associated death domaindependent mechanism. J Biol Chem 274:7245-7252, 1999. Dorrie J, Schuh W, Keil A, et al: Regulation of CD95 expression and CD95-mediated cell death by interferon-gamma in acute lymphoblastic leukemia with chromosomal translocation t(4;11). Leukemia 13:1539-1547, 1999. Griffith TS, Chin WA, Jackson GC, et al: Intracellular regulation of TRAIL-induced apoptosis in human melanoma cells. J Immunol 161:2833-2840, 1998. Bretz JD, Rymaszewski M, Arscott PL, et al: TRAIL death pathway expression and induction in thyroid follicular cells. J Biol Chem 274:23627-23632, 1999. Kataoka T, Schroter M, Hahne M, et al: FLIP prevents apoptosis induced by death receptors but not by perforin/granzyme B, chemotherapeutic drugs, and gamma radiation. J Immunol 161:3936-3942, 1998. Los M, Herr I, Friesen C, et al: Cross-resistance of CD95- and drug-induced apoptosis as a consequence of deficient activation of caspases, ICE/Ced-3 proteases). Blood 90:3118-3129, 1997. Boesen-de Cock JG, Tepper AD, de Vries E, et al: Common regulation of apoptosis signaling induced by CD95 and the DNA-damaging stimuli etoposide and -radiation downstream from caspase-8 activation. J Biol Chem 274:14255-14261, 1999. Sun X-M, MacFarlane M, Zhuang J, et al: Distinct caspase cascades are initiated in receptor-mediated and chemicalinduced apoptosis. J Biol Chem 274:50530-50560, 1999. Wu X-X, Mizutami Y, Kakehi Y, et al: Enhancement of Fasmediated apoptosis in renal cell carcinoma cells by adriamycin. Cancer Res 60:2912-2918, 2000. Tamura T, Aoyama N, Saya H, et al: Induction of Fas-mediated apoptosis in p53-transfected human colon carcinoma cells. Oncogene, 11:1939-1946, 1995. Owen-Sachaub LB, Zhang W, Cusack JC, et al: Wild-type human p53 and a temperature-sensitive mutant induce Fas/APO-1 expression. Mol Cell Biol 15:3032-3040, 1995. Rakkar ANS, Katayos Y, Kim M, et al: A novel adenoviral vector expressing human Fas/CD95/APO-1 enhances p53-mediated apoptosis. Cell Death Diff 6:326-333, 1999. Fukazawa T, Fujiwara T, Morimoto Y, et al: Differential involvement of the CD95, Fas/APO-1, receptor/ligand system on apoptosis induced by the wild-type p53 gene transfer in human cancer cells. Oncogene, 18:2189-2199, 1999. Shimizu M, Yoshimoto T, Nagata S, et al: A trial to kill tumor cells through Fas, CD95)-mediated apoptosis in vivo. Biochem Biophys Res Commun 228:375-379, 1996. Wu GS, Burns TF, McDonald ER 3rd, et al: KILLER/DR5 is a DNA damage-inducible p53-regulated death receptor gene. Nat Genet 17:141-143, 1997. Nagane M, Pan G, Weddle JJ, et al: Increased death receptor 5 expression by chemotherapeutic agents in human gliomas causes synergistic cytotoxicity with tumor necrosis factorrelated apoptosis-inducing ligand in vitro and in vivo. Cancer Res 60:847-853, 2000. Herr I, Wilhelm D, Bohler T, et al: JNK/SAPK activity is not sufficient for anticancer therapy-induced apoptosis involving CD95-L, TRAIL and TNF-alpha. Int J Cancer 80:417-424, 1999. Friesen C, Fulda S, Debatin KM: Induction of CD95 ligand and apoptosis by doxorubicin is modulated by the redox state in chemosensitive- and drug-resistant tumor cells. Cell Death Differ 6:471-480, 1999. Herr I, Wilhelm D, Bohler T, et al: Activation of CD95, APO- 1/Fas, signaling by ceramide mediates cancer therapy-induced apoptosis. EMBO J 16:6200-6208, 1997. Boland MP, Foster SJ, O’Neill LAJ: Daunorubicin activates NF-kB and induces kB-dependent gene expression in HL-60 promyelocytic and Jurkat T lymphoma cells. J Biol Chem 272:12952-12960, 1997 Miyamoto S, Verma IM: Rel/NF-kappaB/IkappaB story. Adv Cancer Res 66:255-292, 1995. Baldwin AS: The NF-kappaB and I kappaB proteins: new discoveries and insights. Ann Rev Immunol 14:649-683, 1996. Chen Z, Hagler J, Palombella VJ, et al: Signal-induced sitespecific phosphorylation targets I kappa B alpha to the ubiquitin- proteasome pathway. Genes and Dev 9:1586-1597, 1995. Karin M: The regulation of AP-1 activity by mitogen-activated protein kinases. J Biol Chem 270:16483-16486, 1995. Kyriakis J, Banerjee P, Nikolakaki E, et al. The stress-activated protein kinase subfamily of c-Jun kinases. Nature 369:156- 160, 1994. Minden A, Lin A, McMahon M, et al: Differential activation of ERK and JNK mitogen-activated protein kinases by Raf-1 and MEKK. Science 266:1719-1723, 1994. Derijard B, Hibi M, Wu I-H, et al: JNK1: a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain. Cell 76:1025-1037, 1994. Chen Y-R, Wang X, Templeton D, et al:The role of c-Jun N-terminal kinase, JNK, in apoptosis induced by ultraviolet C and radiation. J Biol Chem 271:31929-31936, 1996. Kharbanda S, Ren R, Pandey P, et al: Activation of the c-Abl tyrosine kinase in the stress response to DNA-damaging agents. Nature 376:785-788, 1995. Van Dam H, Wilhelm D, Herr I, et al: ATF-2 is preferentially activated by stress-activated protein kinases to mediate c-jun induction in response to genotoxic agents. EMBO J 14:1798- 1811, 1995. Yu R, Shtil AA, Tan T-H, et al: Adriamycin activates c-jun Nterminal kinase in human leukemia cells: a relevance to apoptosis. Cancer Lett 107:73-81, 1996. Seimiya H, Mashima T, Toho M, et al: c-Jun NH2-terminal kinase-mediated activation of interleukin-1b converting enzyme/CED-3-like protease during anticancer drug-induced apoptosis. J Biol Chem 272:4631-4636, 1997. Sanchez-Perez I, Murguia JR, Perona R: Cisplatin induces a persistent activation of JNK that is related to cell death. Oncogene 16:533-540, 1998. Rudel T, Zenke FT, Chuang T-H, et al: Cutting edge: p21-activated kinase, PAK, is required for Fas-induced JNK activation in Jurkat cells. J Immunol 160:7-11, 1998. Liu Z-G, Baskaran C, Lee-Choe ET, et al: Three distinct signaling responses by murine fibroblasts to genotoxic stress. Nature 384:273-276, 1996. Enari M, Sakahiera H, Yokoyama H, et al: A caspase-activated Dnase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature 391:43, 1998. Lee FS, Hagler J, Chen CJ, et al: Activation of the IkBk kinase complex by MEKK1, a kinase of the JNK pathway. Cell 88:213-222, 1997. Bessho R, Matsubara K, Kubota M, et al: Pyrrolidine dithiocarbamate, a potent inhibitor of nuclear factor kB, NF-kB, activation, prevents apoptosis in human promyelocytic leukemia HL-60 cells and thymocytes. Biochem Pharmacol 48:1883-1889, 1994. Piret B, Piette J: Topoisomerase poisons activate the transcription factor NF-kB in ACH-2 and CEM cells. ucl Acids Res 24:4242-4248, 1996. Brach MA, Kharbanda SM, Herrmann F, et al: Activation of the transcription factor kappaB in human KG-1 myeloid leukemia cells treated with 1-beta-D-arabinofuranosylcytosine. Mol Pharmacol 41:60-63, 1992. Testi R. Sphingomyelin breakdown and cell fate. Trends Biochem Sci 21:468-471, 1996. Bose R, Verheij M, Haimovitz-Friedman A, et al: Ceramide synthase mediates daunorubicin-induced apoptosis: an alternative mechanism for generating death signals. Cell 82:405- 414. 1995. Hannun YA: Functions of ceramide in coordinating cellular responses to stress. Science 274:1855-1859, 1996. Hannun YA, Obeid LM: Mechanisms of ceramide-mediated apoptosis. Adv Exp Med Biol 407:145-149, 1997. Mathias S, Pena LA, Kolesnick RN: Signal transduction of stress via ceramide. Biochem J 335:465-480, 1998. Verheij M, Bose R, Lin XH, et al: Requirement for ceramideinitiated SAPK/JNK signalling in stress-induced apoptosis. Nature 380:75-79, 1996. Wiegmann K, Schutze S, Machleidt T, et al: Functional dichotomy of neutral and acidic sphingomyelinases in tumor necrosis factor signaling. Cell 78:1005-1015, 1994. Spiegel S, Foster D, Kolesnick R: Signal transduction through lipid second messengers. Curr Opin Cell Biol 8:159-167, 1996. Hirschberg K, Rodger J, Futerman AH: The long-chain sphingoid base of sphingolipids is acylated at the cytosolic surface of the endoplasmic reticulum in rat liver. Biochem J 290:751- 757, 1993. Shimeno H, Soeda S, Yasukouchi M, et al: Fatty acyl-Co A: sphingosine acyltransferase in bovine brain mitochondria: its solubilization and reconstitution onto the membrane lipid liposomes. Biol Pharm Bull 18:1335-1339, 1995. Adam-Klages S, Schwandner R, Adam D, et al: Distinct adapter proteins mediate acid versus neutral sphingomyelinase activation through the p55 receptor for tumor necrosis factor. J Leukoc Biol 63:678-682, 1998. Schwandner R, Wiegmann K, Bernardo K, et al: TNF receptor death domain-associated proteins TRADD and FADD signal activation of acid sphingomyelinase. J Biol Chem 273:5916- 5922, 1998. Wiegmann K, Schwandne R, Krut O, et al: Requirement of FADD for tumor necrosis factor-induced activation of acid sphingomyelinase. J Biol Chem 274:5267-5270, 1999. Cuvillier O, Edsall L, Spiegel S:Involvement of sphingosine in mitochondria-dependent Fas-induced apoptosis of type II Jurkat T cells. J Biol Chem 275:15691-15700, 2000. Cecconi F, Alvarez-Bolado G, Meyer BI, et al: Apaf1, CED-4 homolog, regulates programmed cell death in mammalian development. Cell 94:727-737, 1998. Susin SA, Zamzami N, Castedo M, et al: The central executioner of apoptosis: multiple connections between protease activation and mitochondria in Fas/APO-1/CD95- and ceramide-induced apoptosis. J Exp Med 186:25-37, 1997. De Maria R, Lenti L, Malisan F, et al: Requirement for GD3 ganglioside in CD95- and ceramide-induced apoptosis. Science 277:1652-1655, 1997. Zhou H, Summers SA, Birnbaum MJ, et al: Inhibition of Akt kinase by cell-permeable ceramide and its implications for ceramide-induced apoptosis. J Biol Chem 273:16568-16575, 1998. Boland MP, Foster SJ, O’Neill LA: Daunorubicin activates NFkappaB and induces kappaB-dependent gene expression in HL-60 promyelocytic and Jurkat T lymphoma cells. J Biol Chem 272:12952-12960, 1997. Dbaibo GS, Pushkareva MY, Rachid RA, et al: p53-dependent ceramide response to genotoxic stress. J Clin Invest 102:329- 339, 1998. Sawai H, Okazaki T, Yamamoto H, et al: Requirement of AP- 1 for ceramide-induced apoptosis in human leukemia HL-60 cells. J Biol Chem 270:27326-27331, 1995. Scaffidi C, Schmitz I, Zha J, et al: Differential modulation of apoptosis sensitivity in CD95 type I and type II cells. J Biol Chem 274:22532-22538, 1999. Cuvillier O, Pirianov G, Kleuser B, et al: Suppression of ceramide-mediated programmed cell death by sphingosine-1- phosphate. Nature 381:800-803, 1996. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 95 EP 106 PG 12 ER PT J AU Banhegyi, G Mandl, J AF Banhegyi, Gabor Mandl, Jozsef TI The Hepatic Glycogenoreticular System SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE glycogen; glycogenolysis; endoplasmic reticulum; glucuronidation; ascorbate; glucose-6-phosphatase ID glycogen; glycogenolysis; endoplasmic reticulum; glucuronidation; ascorbate; glucose-6-phosphatase AB One of the major liver functions is the ability of hepatocytes to store glucose in the form of glycogen for various purposes. Beside glucose production and secretion, the synthesis of glucuronides and ascorbate has been reported to be dependent on the extent of the glycogen stores and on the rate of glycogenolysis in the liver. It is common that the final steps of these pathways are catalysed by intraluminally orientated enzymes of the endoplasmic reticulum, which are supported by transporters for the permeation of substrates and products. On the basis of the close morphological and functional proximity of glycogen, glycogen-dependent pathways and the (smooth) endoplasmic reticulum we propose to use the term ''glycogenoreticular system'' for the description of this export-orientated hepatocyte-specific metabolic unit. C1 [Banhegyi, Gabor] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, H1444 Budapest, Hungary. [Mandl, Jozsef] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, H1444 Budapest, Hungary. RP Mandl, J (reprint author), Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, H1444 Budapest, Hungary. EM mandl@puskin.sote.hu CR Subcellular Biochemistry, Volume 21. Endoplasmic reticulum., Eds. Borgese M and Harris JR), Plenum Press, New York and London, 1993. Nordlie RC, Jorgenson RA: Glucose-6-phosphatase. In: The Enzymes of Biological Membranes, Ed: Martonosi A), Plenum Press, New York, 1976, Vol. 2., pp. 465-491. Chen YT, Burchell A: Glycogen storage diseases. In: The metabolic basis of inherited disease., Eds. Scriver CR, Beaudet AL, Sly WS, Valle D, McGraw-Hill, New York, 1995, pp. 935-965. Thurman RG, Kauffman FC: Factors regulating drug metabolism in intact hepatocytes. Pharmacol Rev 31:229-251, 1980. Banhegyi G, Garzo T, Antoni F, et al: Glycogenolysis - and not gluconeogenesis - is the source of UDP-glucuronic acid for glucuronidation. Biochim Biophys Acta 967:429-435, 1988. Braun L, Garzo T, Mandl J, et al: Ascorbic acid synthesis is stimulated by enhanced glycogenolysis in murine liver. FEBS Lett. 352:4-6, 1994. Banhegyi G, Puskas R, Garzo T, et al: High amounts of glucose and insulin inhibit p-nitrophenol conjugation in mouse hepatocytes. Biochem Pharmacol 42:1299-1302, 1991. Mandl J, Banhegyi G, Kalapos MP, et al: Increased oxidation and decreased conjugation of drugs in the liver caused by starvation., review, Chem-Biol Interact 96:87-101, 1995. Ziegler DM: Role of reversible oxidation-reduction of enzyme thiols-disulfides in metabolic regulation. Ann Rev Biochem 54:305-329, 1985. Braun L, Puskas F, Csala M, et al: Regulation of glucuronidation by glutathione redox state through the alteration of UDP-glucose supple originating from glycogen metabolism. Arch Biochem Biophys 348:169-173, 1997. Braun L, Csala M, Poussu A, et al: Glutathione depletion induces glycogenolysis dependent ascorbic acid synthesis in isolated murine hepatocytes. FEBS Lett 388:173-176, 1996. Banhegyi G, Braun L, Csala M, et al: Ascorbate metabolism and its regulation in animals. Free Radic Biol Med 23:793-803, 1997. Banhegyi G, Garzo T, Fulceri R, et al: Latency is the major determinant of UDP-glucuronosyl-transferase activity in isolated hepatocytes. FEBS Lett 328:149-152, 1993. Kalant N, Parniak M, Lemieux M: Compartmentation of glucose 6-phosphate in hepatocytes. Biochem J 248:927-931, 1987. Christ B, Jungermann K: Sub-compartmentation of the ‘cytosolic’ glucose 6-phosphate pool in cultured rat hepatocytes. FEBS Lett 221:375-380, 1987. Burchell B, Coughtrie MW: UDP-glucuronosyltransferases. Pharmacol Ther 43:261-289, 1989. Kiuchi K, Nishikimi M, Yagi K: Purification and characterization of L-gulonolactone oxidase from chicken kidney microsomes. Biochemistry 21:5076-5082, 1982. Fawcett DW: Observations on the cytology and electron microscopy of hepatic cells. J Natl Cancer Inst 15:1475-1503, 1955. Cardell RR Jr: Smooth endoplasmic reticulum in rat hepatocytes during glycogen deposition and depletion. Int Rev Cytol 48:221-279, 1977. Babcock MB, Cardell RR Jr: Fine structure of hepatocytes from fasted and fed rats. Am J Anat 143:399-438, 1975. Leskes A, Siekevitz P, Palade GE: Differentiation of endoplasmic reticulum in hepatocytes. I. Glucose-6-phosphatase distribution in situ. J Cell Biol 49:264-287, 1971. Clarke DJ, Burchell B: Conjugation-Deconjugation Reactions in Drug Metabolism and Toxicity., 1994, In: Handbook of Experimental Pharmacology, Vol. 112, Ed. Kauffman FC, Springer Verlag, Budapest, 1994, pp. 3-43. Girard J, Pegorier JP: An overview of early post-partum nutrition and metabolism. Biochem Soc Trans 26:69-74, 1998. Dallner G, Siekevitz P, Palade GE: Biogenesis of endoplasmic reticulum membranes. I. Structural and chemical differentiation in developing rat hepatocyte. J Cell Biol 30:73-96, 1966. Babcock MB, Cardell RR Jr: Fine structure of hepatocytes from fasted and fed rats Am. J Anat 143:399-438, 1975. Berry C, Hallinan T: Summary of a novel, three-component regulatory model for uridine diphosphate glucuronyltransferase. Biochem Soc Trans 4:650-652, 1976. Hemrika W, Wever R: A new model for the membrane topology of glucose-6-phosphatase: the enzyme involved in von Gierke disease. FEBS Lett 409:317-319, 1997. Pan CJ, Lei KJ, Annabi B, et al: Transmembrane topology of glucose-6-phosphatase. J Biol Chem 273:6144-6148, 1998. Banhegyi G, Marcolongo P, Fulceri R, et al: Demonstration of a metabolically active glucose-6-phosphate pool in the lumen of liver microsomal vesicles. J Biol Chem 272:13584-13590, 1997. Gerin I, Veiga da Cunha M, Achouri Y, et al: Sequence of a putative glucose 6-phosphate translocase, mutated in glycogen storage disease type Ib. FEBS Lett 419:235-238, 1998. Bossuyt X, Blanckaert N: Mechanism of stimulation of microsomal UDP-glucuronosyltransferase by UDP-N-acetylglucosamine. Biochem J 305:321-328, 1995. Banhegyi G, Braun L, Marcolongo P, et al: Evidence for an UDP-glucuronic acid - phenol glucuronide antiport in rat liver microsomal vesicles. Biochem J 315:171-176, 1996. Puskas F, Braun L, Csala M, et al: Gulonolactone oxidase activity-dependent intravesicular glutathione oxidation in rat liver microsomes. FEBS Lett 430:293-296, 1998. Guillam MT, Burcelin R, Thorens B: Normal hepatic glucose production in the absence of GLUT2 reveals an alternative pathway for glucose release from hepatocytes. Proc Natl Acad Sci USA 95:12317-12321, 1998. Upston JM, Karjalainen A, Bygrave FL: Efflux of hepatic ascorbate: a potential contributor to the maintenance of plasma vitamin C. Biochem J 342:49-56, 1999. Banhegyi G, Marcolongo P, Puskas F, et al: Dehydroascorbate and ascorbate transport in rat liver microsomal vesicles. J Biol Chem 273:2758-2762, 1998. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 107 EP 110 PG 4 ER PT J AU Croce, VM Price, RM Segal-Eiras, A AF Croce, Virginia Maria Price, R Mike Segal-Eiras, Amada TI Association of a alpha1 Acidic Glycoprotein and Squamous Cell Carcinoma of the Head and Neck SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Head and neck cancer; alpha1 acidic-glycoprotein; disease stage ID Head and neck cancer; alpha1 acidic-glycoprotein; disease stage AB Serum from patients with different malignancies contain an abnormal concentration of a a1-acidic-glycoprotein (AAG) and also, increased levels of AAG are associated with the presence of tumor mass. In the present report, serum levels of AAG were measured by radial immunodiffusion in squamous cell carcinoma of the head and neck (SCCHN) patients taking into account disease status parameters such as tumor localization, stage and extension of disease. Immunohistochemical methods, SDS-PAGE and Western-blotting were employed to study the expression of AAG and a carbohydrate related antigen (sialyl Lewis x) in tumor tissues and derived fractions. AAG showed abnormal levels in 7/15 oral cavity tumor patients’ sera, 2/5 oropharynx and 5/10 larynx tumors; increased AAG serum levels belonged to patients with disseminated disease. On the other hand, the presence of AAG and sialyl Lewis x were demonstrated in carcinoma cells and in derived fractions from tumor tissues belonging to patients with elevated AAG serum levels. In the present study, we have found elevated levels of AAG in serum samples from SCCHN patients; these neoplastic cells are capable to express AAG. C1 [Croce, Virginia Maria] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120, 1900 La Plata, Argentina. [Price, R Mike] University of Nottingham, School of Pharmaceutical Sciences, Cancer Research LaboratoriesNottingham, UK. [Segal-Eiras, Amada] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120, 1900 La Plata, Argentina. RP Segal-Eiras, A (reprint author), Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), 1900 La Plata, Argentina. EM as-eiras@netverk.com.ar CR Bennett M, Schmid K: Immunosuppression by human plasma a1 acid glycoprotein: importance of the carbohydrate moiety. Proc Natl Acad Sci USA 77:6109-6114, 1980. Caldani C, Thyss A, Schneider M, et al: Orosomucoid: prealbumin ratio- a marker of the host-tumor relationship in head and neck cancer. Eur J Cancer Clin Oncol 24:653-657, 1988. Croce MV, Segal-Eiras A: Identification of acute–phase proteins, APP, in circulating immune complexes, CIC, in esophageal cancer patients’ sera. Cancer Invest 4:421-426, 1997. Croce MV, Colussi AG, Price M, et al: An immunohistopathological characterization of spontaneous metastases in a human lung mucoepidermoid adenocarcinoma, HLMC, xenograft. Pathol Oncol Res 4:259-266, 1998. Chandrasekaran EV, Davila M, Nixon D, et al: Structures of oligosaccharide chains of two forms of a1 acidic glycoprotein purified from liver metastases of lung, colon and breast tumors. Cancer Res 44:1557-1567, 1984. Cheresh DA, Haynes D, Distasio JA: Interaction of an acute phase reactant a1 acid glycoprotein [orosomucoid] with the lymphoid cell surface a model for non-specific immune suppression. Immuno 51:541-547, 1984 Dage JL, Ackermann BL, Halsall HB: Site localization of sialyl Lewisx antigen ona1 acid glycoprotein by high performance liquid chromatography-electrospray mass spectometry. Glycobiol 8:755-760, 1998. De Graaf TW, Van der Stelt ME, Anbergen MG: Inflammation induced expression of sialyl-Lewis x-containing glycan structures on alpha 1 acidic glycoprotein, orosomucoid, in human sera. J Exp Med 177:657-666, 1993. Elg SA, Mayer AR, Carson LF, et al: a1-acidic glycoprotein is an immunosuppressive factor found in ascites from ovarian carcinoma. Cancer 80:1448-1456, 1997. Feickert HJ, Anger BR, Cordon-Cardo C, et al: Cell-surface antigens of human lung tumours detected by mouse monoclonal antibodies: definition of blood-group- and non-blood-grouprelated antigenic systems. Int J Cancer 46:1007-1013, 1990. Feizi T: Demonstration by monoclonal antibodies that carbohydrate structures of glycoproteins and glycolipids are oncodevelopmental antigens. Nature 314:53-57, 1985. Fujii M, Takahashi N, Hayashi H, et al: Comparative study of alpha1-acidic glycoprotein molecular variants in ascitic fluid of cancer and non-cancer patients. Anticancer Res 8:303-306, 1988. Hakomori S, Nudelman E, Levery SB: Novel fucolipids accumulating in human adenocarcinoma: I. Glycolipids with di- or trifucosylated type 2 chain. J Biol Chem 259:4672-4680, 1984. Hanai N, Shitara K, Yoshida H: Generation of monoclonal antibodies against human lung squamous cell carcinoma and adenocarcinoma using mice rendered tolerant to normal human lung. Cancer Res 46:4438-4443, 1986. International Union Against Cancer, UICC, Classification of Malignant Tumours. Ed. LH Sobin and Ch Wittekind, 5th edition, New York, 1997. Irimura T, McIsaac AM, Matsushita Y, et al: Colorectal cancer metastasis determined by carbohydrate–mediated cell adhesion: role of sialyl-Lex antigens. Semin CancerBiol 4:319-324, 1993. Itoh T, Yonezawa S, Nomoto M, et al: Expression of mucin antigens and Lewis x related antigens in carcinomas and dysplasia of the pharynx and larynx. Pathol Int 46:646-655, 1996. Kannagi R: Carbohydrate-mediated cell adhesion involved in hematogenous metastasis of cancer. Glycoconjugate J 14:577- 584, 1997. Krecicki T, Leluk M: Acute phase reactant proteins-an aid to monitoring surgical treatment of laryngeal carcinoma. J Laryngol Otol 106:613-619, 1992. Laemmli UK: Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature, Lond, 227:680- 681, 1980. Ljungberg B, Grankvist K, Rasmuson T: Serum interleukin-6 in relation to acute-phase reactants and survival in patients with renal cell carcinoma. Eur J Cancer 33:1794-1798, 1998. Makino K, Ogata T, Miyake H, et al: Expression of tumor associated glycoantigen, sialyl Lewis a, in human head and neck squamous cell carcinoma and its application to tumor immunotherapy. Jpn J Cancer Res 85:887-891, 1994. Matsushita Y, Cleary KR, Ota DM, et al: Sialyl-dimeric Lewis- X antigen expressed on mucin-like glycoproteins in colorectal cancer metastases. Lab Invest 63:780-790, 1990. Nakamori S, Kameyama M, Imaoka S, et al: Involvement of carbohydrate antigen sialyl Lewis, x, in colorectal cancer metastasis. Dis Col Rectum 40:420-431, 1997. Phillips ML, Nuddelman E, Gaeta FCA, et al: ELAM-1 mediates cell adhesion by recognition of a carbohydrate ligand, Sialyl- Lex. Science 250:1130-1132, 1990. Price MR, Edwards S, Owainati A, et al: Multiple epitopes on a human breast carcinoma associated antigen. Int J Cancer 36:567-574, 1985. Schultz D, Arnold P: Properties of four acute phase proteins: Creactive protein, serum amyloid A protein, a1-acidic glycoprotein and fibrinogen. Sem Arthr Rheum 20:129-147, 1990. Stamatidis A, Toumanidou M, Vyssoulis G: Value of serum acute-phase proteins and CEA in preoperative staging of colorectal cancer. Cancer 65:2055-2057, 1990. Stanciu L, Dumitrascu D, Radu D: Non-specific tumoral markers in hepato-cellular carcinoma. Med Intern: 28:139-144, 1990. Suarez-Nieto C, Cuesta-Garcia A, Fernandez-Bustillo E, et al: Serum glycoproteins and prognosis in cancer of the head and neck. Clin Otolaryngol 11:41-45, 1986. Svitacheva N, Davies JR, Lesuffleur T: Characterization of mucins produced by human HT29-MTX cell line. 5th International Workshop on Carcinoma Associated Mucins Cambridge, UK, 1998. Towbin H, Staehelin T, Gordon J: Electrophoretic transfer of proteins from polyacrilamide gels to nitrocellulose sheets: procedures and some applications. Proc Natl Acad Sci USA 76:4350-4354, 1979. Woods KV, El Naggar A, Clayman GL, et al: Variable expression of cytokines in human head and neck squamous cell carcinoma cell lines and consistent expression in surgical specimens. Cancer Res 58:3132-3141, 1998. Zar JH: Biostatistical Analysis, 3rd ed, Prentice Hall, New Jersey, USA, 1996. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 111 EP 117 PG 7 ER PT J AU Magyarlaki, T Buzogany, I Kaiser, L Sukosd, F Dobronte, R Simon, B Fazekas, A Nagy, J AF Magyarlaki, Tamas Buzogany, Istvan Kaiser, Laszlo Sukosd, Farkas Dobronte, Robert Simon, Barbara Fazekas, Attila Nagy, Judit TI Prognostic Histological and Immune Markers of Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE renal cell carcinoma; histology; immunohistochemistry; immunotherapy; clinicopathology ID renal cell carcinoma; histology; immunohistochemistry; immunotherapy; clinicopathology AB Recent development on the fields of molecular genetics and immunology of human renal cell carcinoma (RCC) have resulted in more successful treatment of advanced and metastatic RCCs. Re-evaluation of the prognostic/predictive data aim the initial tumor staging of RCC patients to achieve better patient selection for immune and gene therapy. 125 RCC patients diagnosed according to the Heidelberg histological classification, graded, Robson staged, immune treated (Interferon-a a+ Vinblastin or Broncho-Waxom/Decaris) were followed-up clinically for 36 months. Tumor immunity markers by immunohistochemistry of tumor infiltrating lymphocytes (TIL) were detected by immunoperoxidase methods using monoclonal antibodies. Tumoral immune complexes (TIC) were visualized by fluorescent polyclonal antibodies. Histologically oncocytomas defined a better (p<0.02) and sarcomatous RCCs a worse (p<0.01) follow-up prognosis. Basically, the metastatic status (related with the stage and grade) determined the clinical outcome (p<0.00002) of the RCC patients. Tumoral immune complexes (TIC) were weak positive, while tumor infiltrating lymphocytes (TIL) weak negative predictors of the succes of Broncho-Waxom/Decaris immune therapy. Molecular genetic based histological classification, grade, stage and metastatic status parameters together with some tumor immunity parameters (TIL, TIC) can predict the success of immunotherapy of RCC patients. C1 [Magyarlaki, Tamas] University of Pecs, Institute of Biochemistry and Medical Chemistry, Ifjusag utja 13.Pecs, Hungary. [Buzogany, Istvan] University of Pecs, Department of UrologyPecs, Hungary. [Kaiser, Laszlo] University of Pecs, Department of PathologyPecs, Hungary. [Sukosd, Farkas] University of Pecs, Department of PathologyPecs, Hungary. [Dobronte, Robert] University of Pecs, Institute of Biochemistry and Medical Chemistry, Ifjusag utja 13.Pecs, Hungary. [Simon, Barbara] University of Pecs, Institute of Biochemistry and Medical Chemistry, Ifjusag utja 13.Pecs, Hungary. [Fazekas, Attila] University of Pecs, 2nd Department of Medicine and Nephrological CenterPecs, Hungary. [Nagy, Judit] University of Pecs, 2nd Department of Medicine and Nephrological CenterPecs, Hungary. RP Magyarlaki, T (reprint author), University of Pecs, Institute of Biochemistry and Medical Chemistry, Pecs, Hungary. EM kellerm@apacs.pote.hu CR Angevin E, Kreme, F, Gaudin C, et al: Analysis of T-cell immune response in renal cell carcinoma: polarisation to type 1-like differentiation pattern, clonal T-cell expansion and tumor-specific cytotoxicity. Int J Cancer 72:431-440, 1997. Belldegrun A, Tao CL, Kaboo R, et al: Natural immune reactivity- associated therapeutic response in patients with metastatic renal cell carcinoma receiving tumor-infiltrating lymphocytes and interluekin-2-based therapy. J Immunother Tumor Immunol 19:149-161, 1996. Bezooijen van RL, Goey H, Stoter G, et al: Prognostic markers for survival in patients with metastatic renal cell carcinoma treated with interleukin-2. Cancer Immunol Immunother 43:293-298, 1996. Brouwenstijn N, Gaugler B, Kruse KM, et al: Renal-cell carcinoma- specific lysis by cytotoxic T-lymphocyte clones isolated from peripheral blood lymphocytes and tumor infiltrating lymphocytes. Int J Cancer 68:177-182, 1996. Buszello H, Ackermann R:Immunohistochemical studies on the expression of HLA Class I antigen in renal cell carcinoma: comparison of primary and metastatic tumor tissue. Eur Urol 25:158-163, 1994. Buzogany I, Czvalinga I, Gotz F: Ambulanter vegzett interferon mono- es kombinacios terapia eredmenye elorehaladott vesedaganat eseten. Orv Hetil 138:67-70, 1997. Chang AE, Aruga A, Cameron MJ, et al: Adoptive immunotherapy with vaccine-primed lymph node cells secondarily activated with anti-CD3 and interleukin-2. J Clin Oncol 15:796- 807, 1997. Corless CL, Kibel AS, Iliopoulos O, et al: Immunostaining of the von Hippel-Lindau gene product in normal and neoplastic human tissues. Hum Pathol 28:459-463, 1996. Crotty TB, Farrow GM, Liber HJ: Chromophobe cell renal carcinoma: clinicopathological features of 50 cases. J Urol 154:964-967, 1996. Curnow RT: Clinical experience with CD64-directed immunotherapy. An overview. Cancer Immunol Immunther 45:210- 215, 1997. Hove van den LE, van Gool SW, van Poppel, et al: Phenotype, cytokine production and cytolytic capacity of fresh, uncultured, tumor-infiltrating T lymphocytes in human renal cell carcinoma. Clin Exp Immunol 109:501-509, 1997. Hove van den LE, van Gool SW, van Poppel, et al: Identification of an enriched CD4+ CD8alpha++ CD8beta+ T-cell subset among tumor-infiltrating lymphocytes in human renal cell carcinoma. Int J Cancer 71:178-182, 1997. Jantzer P, Schendel DJ: Human renal cell carcinoma antigenspecific CTLs: antigen-driven selection and long-term persistence in vivo. Cancer Res 58:3078-3086, 1998. Kawata N, Akimoto Y, Hirano D, et al: Immunological effect of recombinant interferon-gamma on tumor infiltrating lymphocytes of renal cell carcinoma – relationshship with clinical stage, Japanese). Hinyokika-Kiyo 42:1-4, 1996. Knebelman B, Ananth S, Cohen HT, et al: Transforming growth factoc alpha is a target fro the von Hippel-Lindau tumor suppressor. Cancer Res 58:226-231, 1998. Kovacs Gy, Akhtar M, Beckwith BJ, et al: The Heidelberg classification of renal cell tumors. J Pathol 183:131-133, 1997. Kovacs Gy, Erlandsson R, Boldog F, et al: Consistent chromosome 3p deletion and loss of heterozygositiy in renal cell carcinoma. Proc Natl Acad Sci USA 85:1571-1575, 1988. Kowalczyk D, Skorupski W, Kwias Z, et al: Flow cytometric analysis of tumor-infiltrating lymphocytes in patients with renal cell carcinoma. Br J Urol 80:543-547, 1997. Krumm A, Groudine M: Tumor suppression and transciption elongation: the dier consequences of changing partners. Science 269:1400-1401, 1995. Licht MR, Novick AC, Tubbs RR, et al: Renal oncocytoma: clinical and biological correlates. J Urol 150:1380-1383, 1993. Lubensky IA, Schmidt L, Zhuang Z, et al: Hereditary and sporadic papillary renal carcinomas with c-met mutations share a distinct morphological phenotype. Am J Pathol 155:517-526, 1999. Magyarlaki T, Kiss B, Buzogany I, et al: Renal cell carcinoma and paraneoplastic IgA nephropathy. Nephron 82:127-130, 1999. Magyarlaki T, Mosolits S, Baranyay F, et al: Immunohistochemistry of complement responses on human renal cell carcinoma biopsies. Tumori 82:473-480, 1996. Moch H, Schraml P, Bubendorf L, et al: Intratumoral heterogeneity of von Hippel-Lindau gene deletions in renal carcinoma detected by fluorescence in situ hybridization. Cancer Res 58:2304-2309, 1998. Neumann HPH, Bender BU, Berger DP, et al:Prevalence, morphology and biology of renal cell carcinoma in von Hippel-Lindau disease compared to sporadic reanl cell carcinoma. J Urol 160:1248-1254, 1998. Olive C, Nikol D, Falk MC: Characterisation of gamma delta T cells in renal cell carcinoma patinets by polymerase chain reaction analysis of T cell receptor transcripts. Cancer Immunol Immunother 44:27-34, 1997. Siemeister G, Weindel K, Mohrs K, et al: Reversion of deregulated expression of vascular endothelial growth factor in human renal carcinoma cells by von Hippel-Lindau suppressor protein. Cancer Res 56:2299-2301, 1996. Surfus JE, Hank JA, Oosterwijk E, et al: Anti-renal-cell carcinoma chimeric antibody G250 facilitates antibody-dependent cellular cytotoxicity with in vitro and in vivo interleukin-2-activated effectors. J .Immunother Tumor Immunol 19:184-191, 1996. Toliou T, Stravoravd P, Polyzonis M, et al: Natural killer cell activation after interferon administration in patients with metastatic renal cell carcinoma: an ultrastructural and immunohistochemical study. Eur Urol 29:252-256, 1996. Vogelzang NJ, Stadler WM: Kidney cancer. Lancet 352:1691- 1696, 1998. Weijtens ME, Willemsen RA, van Krimpen BA, et al: Chimeric scFv/gamma receptor-mediated T-cell lysis of tumor cells is coregulated by adhesion and accessory molecules. Int J Cancer 77:181-187, 1998. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 118 EP 124 PG 7 ER PT J AU Khan, E Mapara, Z Khan, Sh Naveeda, A Siddiqui, T Pervez, Sh AF Khan, Enver Mapara, Zohair Khan, Shaista Naveeda, Arshad Siddiqui, Tariq Pervez, Shahid TI DNA Ploidy Analyses in 218 Consecutive Pakistani Breast Cancer Patients: Does it Add Anything? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DNA ploidy; breast carcinoma; Pakistan ID DNA ploidy; breast carcinoma; Pakistan AB An analysis was made to evaluate the significance of DNA ploidy in the biology and prognosis of breast carcinoma. This was done by estimating the correlation of DNA ploidy with other established prognostic markers of breast cancer, namely tumor size, tumor grade, lymph node metastasis and S-phase fraction. From 1995 up to year 2000 ploidy analysis was performed on 218 consecutive cases of infiltrating breast carcinoma by flow cytometry using formalin fixed paraffin embedded material. From the laboratory record, data regarding other pathological variables was retrieved. No correlation could be found between DNA ploidy and tumor grade, nor could there be found a correlation with tumor size. For lymph node metastasis there was a significant difference between the proportion of aneuploids and diploids having metastasis in more than 4 lymph nodes. However, no significant difference was found in axillary lymph node positive and negative groups when number of positive lymph nodes was not taken into account. The mean value of S-phase fraction for the aneuploids and the diploids was also insignificantly different. In conclusion DNA ploidy alone did not add much to predict tumor behaviour in terms of known pathologic variables. C1 [Khan, Enver] The Aga Khan University, Department of Pathology and MicrobiologyKarachi, Pakistan. [Mapara, Zohair] The Aga Khan University, Department of Pathology and MicrobiologyKarachi, Pakistan. [Khan, Shaista] The Aga Khan University, Department of SurgeryKarachi, Pakistan. [Naveeda, Arshad] The Aga Khan University, Department of Pathology and MicrobiologyKarachi, Pakistan. [Siddiqui, Tariq] The Aga Khan University Medical Centre, Department of MedicineKarachi, Pakistan. [Pervez, Shahid] The Aga Khan University, Department of Pathology and MicrobiologyKarachi, Pakistan. RP Pervez, Sh (reprint author), The Aga Khan University, Department of Pathology and Microbiology, Karachi, Pakistan. EM shahid.pervez@aku.edu CR Naila Z, Azhar M, Parveen A, et al: Pattern of malignant tumors in Karachi- is it different? JCPSP 10:338-341, 2000. Bhurgri Y, Bhurgi A, Hassan SH, et al: Cancer Incidence in Karachi, Pakistan: first results form Karachi Cancer Registry. Int J Cancer 85:325-329, 2000. Brower ST, Tartter PI, Ahmed S, et al: Proliferative indices and oncoprotein expression in benign and malignant breast biopsies. Ann Surg Oncol 2:416-423, 1995. Utada Y, Yoshimoto M, Kasumi F, et al: Relationship between DNA ploidy and survival in breast cancer. Gan To Kagaku Ryoho 25 Suppl: 431-435, 1998. Camplejohn RS, Ash CM, Gillett CE, et al: The prognostic significance of DNA flow cytometry in breast cancer: results from 881 patients treated in a single centre. Br J Cancer 71:140-145, 1995. Wong SW, Rangan AM, Bilous AM, et al: The value of S-phase and DNA ploidy analysis as prognostic markers for node-negative breast cancer in the Australian setting. Pathology 31:90-94, 1999. Shiao YH, Chen VW, Lehmann HP, et al: Patterns of DNA ploidy and S-phase fraction associated with breast cancer survival in blacks and whites. Clin Cancer Res. 3:587-592, 1997. Chen CM, Chang HT, Mok KT, et al: Analysis of prognostic factors in Chinese women with breast cancer in southern Taiwan. Chung Hua I Hsueh Tsa Chih Taipei 62:717-723, 1999. Krieger N, van den Eeden SK, Zava D, et al: Race/ethnicity, social class and prevalence of breast cancer prognostic biomarkers: A study of white, black, and Asian women in the San Francisco bay area. Ethn Dis 7:137-149, 1997. Stanton PD, Cooke TG, Oakes SJ, et al: Lack of prognostic significance of DNA ploidy and S phase fraction in breast cancer. Br J Cancer 66:925-929, 1992. Stal O, Brisfors A, Carstensen J, et al: Relationships of DNA ploidy, S-phase fraction and hormone receptor status to tumor stage in breast cancers detected by population screening. The South-East Sweden Breast Cancer Group. Int J Cancer 51:28- 33, 1992. Feichter GE, Mueller A, Kaufmann M, et al: Correlation of DNA flow cytometric results and other prognostic factors in primary breast cancers. Int J Cancer 41:823-828, 1988. Kallioniemi OP, Blanco G, Alavaikko M, et al: Improving the prognostic value DNA flow cytometry in Breast Cancer by combining DNA index and S-Phase fraction. Cancer 62:2183- 2190, 1988. 13. Dressler LG, Seamer LC, Owens MA, et al: A flow cytometry and prognostic factors in 1331 frozen breast cancer specimens. Cancer 61:420-427, 1988. Redkar AA, Balsara BR, Bhisey AN, et al: DNA analysis of breast cancer by flow cytometry and correlation with other prognostic parameters. Indian J Med Res 96:223-229, 1992. Dittadi R, Calderazzo F, Cabrelle A, et al:C-erbB-2/neu protein expression, DNA ploidy and S phase in breast cancer. Cell Prolif 29:403-412, 1996. Wenger CR, Beardslee S, Owens MA, et al: DNA ploidy, Sphase, and steroid receptors in more than 127,000 breast cancer patients. Breast Cancer Res Treat 28:9-20, 1993. Beerman H, Kluin PM, Hermans J, et al: Prognostic Significance of DNA ploidy in a series of 690 primary breast cancer patients. Int J Cancer 45:34-945, 1990. Fallenius AG, Franzen SA, Auer GU: Predictive Value of Nuclear DNA content in Breast Cancer in relation to Clinical and Morphological factors. Cancer 62:521-530, 1988. Ewers SB, Langstrom E, Baldetorp B, et al: Flow cytometric DNA analysis in Primary Breast Carcinoma and Clinicopatholgical Correlation. Cytometry 5:408-419, 1984. Bergers E, van Diest PJ, Baak JP: Cell cycle analysis of 932 flow cytometric DNA histograms of fresh frozen breast carcinoma material. Correlations between flow cytometric, clinical, and pathologic variables. Cancer. 1996; 77(11): 2258-66. Brotherick I, Shenton BK, Cowan WK, et al: The relationship between flow-cytometric and immunohistochemically detected c-erbB-2 expression, grade and DNA ploidy in breast cancer. Cancer Immunol Immunother 41:137-145, 1995. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 125 EP 128 PG 4 ER PT J AU Tenderenda, M Rutkowski, P Jesionek-Kupnicka, D Kubiak, R AF Tenderenda, Micha Rutkowski, Piotr Jesionek-Kupnicka, Dorota Kubiak, Robert TI Expression of CD34 in Gastric Cancer and its Correlation with Histology, Stage, Proliferation Activity, p53 Expression and Apoptotic Index SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD 34 immunostaining; gastric cancer ID CD 34 immunostaining; gastric cancer AB The formation of new blood vessels is essential for tumor growth and progression. Until today there are only few studies of the immunohistochemical assessment of angiogenesis in gastric cancer by the evaluation of the expression of CD34 antigen. The aim of this study was to analyze the relationship between microvessel density (MVD) expressed as the mean count of CD34 immunostained vessels and clinicopathologic features of gastric tumors (the histological type according to the Lauren classification, tumor grade – G; presence of lymph node metastases – N; depth of tumor invasion; stage of disease (UICC-AJCC 1988–1992), p53 expression, tumor cell proliferative activity described as the Ki67 labelling index and apoptotic index of tumor cells – TUNEL method). We assessed formalin-fixed, paraffin-embedded tissue samples obtained during potentially radical gastrectomy from 58 patients with primary gastric adenocarcinoma. The representative tissue blocks from each tumor were used for the immunohistochemical assay and examined by two pathologists independently. MVD was counted in five tumor areas of the most intensive neovascularization (x 200 field by light microscopy) and the mean counts were recorded. The mean MVD (CD34 expression value±SD) in this study was 43,15±19,8 per x 200 field. The study demonstrated the statistically significant correlation between MVD and two main histological parameters: tumor grading (p < 0.001) and tumor histological type according to Lauren’s classification (p<0.05). In well – and moderately – differentiated tumors (G1/2) MVD was significantly lower in comparison to the group of poorly differentiated cancer – G3 (mean value: 31,62 vs. 49,89). MVD was higher in diffuse type of gastric cancer comparing to intestinal type (50.05±19,03 vs. 39.17±20,09). However, the authors failed to find a significant correlation between MVD and other investigated histopathological features in malignant gastric tumors. The close relationship between CD34 immunostaining, gastric cancer tumor vascularity and main histological parameters was shown in this study. It can be stated that analysis of expression of angiogenesis in gastric cancer may be helpful for better estimation of hematogenous recurrence and the selection of the group of patients for adjuvant antiangiogenic treatment. C1 [Tenderenda, Micha] Medical University of Lodz, Department of Oncological SurgeryLodz, Poland. [Rutkowski, Piotr] M. Sklodowska-Curie Memorial Cancer Center-Institute, Department of Soft Tissue/Bone Tumors, Roentgena Str. 5, 02-781Warsaw, Poland. [Jesionek-Kupnicka, Dorota] Medical University of Lodz, Department of PathologyLodz, Poland. [Kubiak, Robert] Medical University of Lodz, Department of PathologyLodz, Poland. RP Rutkowski, P (reprint author), M. Sklodowska-Curie Memorial Cancer Center-Institute, Department of Soft Tissue/Bone Tumors, Warsaw, Poland. EM rutkowskip@coi.waw.pl CR Roder JD, Bottcher K, Siewert JR, et al: Prognostic factors in gastric carcinoma. Results of the German Gastric Carcinoma Study 1992. Cancer 72:2089-97, 1993. Folkman J: What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst 82:4-6, 1990. Hansen S, Grabau DA, Sorensen FB, et al: Vascular grading of angiogenesis; prognostic significance in breast cancer. Br J Cancer 82:339-347, 2000. Matsuyama K, Chiba Y, Sasaki M, et al: Tumor angiogenesis as a prognostic marker in operable non-small cell lung cancer. Ann Thorac Surg 65:1405-1409, 1998. Chung YS, Maeda K, Sowa M: Prognostic value of angiogenesis in gastro-intestinal tumors. Eur J Cancer 32A:2501-2505, 1996. Schlingemann RO, Rietveld FJR, de Waal RMW, et al: Leukocyte antigen CD34 is expressed by a subset of cultured endothelial cells and on endothelial abluminal microprocesses in the tumor stroma. Lab Invest 62:690-696, 1990. Lauren P: The two main types of gastric carcinoma: diffuse and so-called intestinal type carcinoma Acta Pathol Microbiol Scand 64:31-49, 1965. Rutkowski P, Jesionek-Kupnicka D, Tenderenda M: A study on the relationship between Ki67 labelling index and selected clinicopathological features in gastric cancer. Onkol Pol 2:143-146, 1999. Weidner N, Semple JP, Weich WR, et al: Tumor angiogenesis and metastasis – correlation in invasive breast carcinoma. N Engl J Med 324:1-8, 1991. Vermeulen PB, Gasparini G, Fox SB, et al: Quantification of angiogenesis in solid human tumors: an international consensus on the methodology and criteria of evaluation. Eur J Cancer 32A:2474-2484, 1996. Banner BF, Whitehouse R, Baker SP, et al: Tumor angiogenesis in stage II colorectal carcinoma. Association with survival. Am J Clin Pathol 109:733-737, 1998. Tanigawa N, Amaya H, Matsamura M, et al: Extent of tumor vascularization correlates with prognosis and hematogenous metastasis in gastric carcinomas. Cancer Res 56:2671-2676, 1996. Kitadai Y, Haruma K, Sumii K, et al: Expression of interleukin- 8 correlates with vascularity in human gastric carcinomas. Am J Pathol 152:93-96, 1998. Inoue K, Ozeki Y, Suganuma T, et al: Vascular Endothelial Growth Factor Expression in Primary Esophageal Squamous Cell Carcinoma. Cancer 79:206-213, 1997. Tanigawa N, Amaya H, Matsumura M, et al: Correlation Between Expression of Vascular Endothelial growth Factor and Tumor Vascularity and Patients Outcome in Human Gastric Carcinoma. J Clin Oncol 15:826-832, 1997. Ikeguchi M, Oka S, Saito H, et al: The expression of vascular growth factor and proliferative activity of cancer cells in gastric cancer. Langebecks Arch Surg 384:264-270, 1999. Heimburg S, Oehler MK, Papadopoulos T, et al: Prognostic relevance of the endothelial marker CD 34 in ovarian cancer. Anticancer Res 19:2527-2529, 1999. Engel CJ, Bennett ST, Chambers AF, et al: Tumor angiogenesis predicts recurrence in invasive colorectal cancer when controlled for Dukes stage. Am J Surg Pathol 20:1260-1265, 1996. Setala LP, Kosma V-M, Marin S, et al: Prognostic factors in gastric cancer; the value of vascular invasion, mitotic rate and lymphoplasmatic infiltration. Br J Cancer 74:766-772, 1996. Gabbert HE, Meier S, Gerharz CD, et al: Tumor-cell dissociation at the invasive front: a new prognostic parameter in gastric cancer patients Int J Cancer 50:202-207, 1992 Rak J, Filmus J, Kerbel RS: Reciprocal paracrine interactions between tumor cells endothelial cells: the „angogenesis progression“ hypothesis. Eur J Cancer 32A:2438-2450, 1996 Dvorak HF, Siossat TM, Brown LF, et al: Distribution of vascular permeability factors in tumors: concentration in tumor blood vessels. J Exp Med 174:1275-1278, 1991 Connolly DT, Heuvelman DM, Nelson R, et al: Tumor vascular permeability factor stimulates endothelial cell growth and angiogenesis. J Clin Invest 84:1470-1478, 1989. Haraguchi M, Miyadera K, Uemura K, et al: Angiogenic activity of enzymes. Nature, Lond., 368:198, 1994. Bussolino F, Albini A, Camussi G, et al: Role of soluble mediators in angiogenesis. Eur J Cancer 32A, 2401-2412, 1996. Maeda K, Kang S-M, Ogawa M, et al: Combined analysis of vascular endothelial growth factors and platelet-derived endothelial cell growth factor expression in gastric carcinoma. Int J Cancer, Pred Oncol, 74:545-550, 1997. Pritchard AJ, Chatterjee T, Wilkinson M, et al: Evidence for weak angogenic response to human colorectal cancer. Br J Cancer 71:1081-1086, 1995. Ohtani H, Sasano N: Stromal cell changes in human colorectal adenomas and carcinomas. Virchows Arch A Pathol Anat Histopathol 401:209-222, 1983. Nakayama H, Enzan H, Miyazaki E, et al: Differential expression of CD34 in normal colorectal tissue, peritumoral inflammatory tissue, and tumor stroma. J Clin Pathol 53:626-629, 2000. Ikeguchi M, Cai J, Yamane N, et al: Clinical significance oif spontaneous apoptosis in advanced gastric adenocarcinoma Cancer 85:2329-2323, 1999. Lu C, Tanigawa N: Spontaneous Apoptosis is Inversely Related to Intratumoral Density in Gastric Carcinoma Cancer Res 57:221-224, 1997. Rosa JC, Mendes R, Filipe MI, et al: Measurement of cell proliferation in gastric carcinoma; comparative analysis of Ki67 and proliferative cell nuclear antigen, PCNA, Histochem J 24:93-101, 1992. Kirsch D, Kastan M: Tumor-suppressor p53; implications for tumor development and prognosis. J Clin Oncol 1998; 16:3158- 3168, 1998. Tanigawa N, Amaya H, Matsumura M, Shimomatsuya T: Association of tumor vasculature with tumor progression overall survival of patients with non-early gastric carcinomas Br J Cancer 75:556-571, 1997. Tanigawa N, Amaya H, Matsumura M, et al: Tumor angiogenesis and mode of metastasis in patients with colorectal cancer Cancer Res 57:1043-1046, 1997. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 129 EP 134 PG 6 ER PT J AU Gharagozloo, S Khoshnoodi, J Shokri, F AF Gharagozloo, Soheila Khoshnoodi, Jalal Shokri, Fazel TI Hepatitis C virus Infection in Patients with Essential Mixed Cryoglobulinemia, Multiple Myeloma and Chronic Lymphocytic Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatitis C virus; Lymphoproliferative disorders; Anti-HCV antibody ID Hepatitis C virus; Lymphoproliferative disorders; Anti-HCV antibody AB Increased prevalence of HCV infection in some lymphoproliferative diseases has been recently reported. In the present study, the frequency of anti-HCV antibody (Ab) together with hepatitis B surface (HBs) antigen (Ag) and anti-HBs Ab were determined in 42, 45 and 23 patients with essential mixed cryoglobulinemia (EMC), multiple myeloma (MM) and B-cell chronic lymphocytic leukemia (B-CLL), respectively. Thirty hospitalized patients with chronic rheumatoid arthritis (RA) were also included as a control. Specific antibodies to HCV antigens were detected by enzyme linked immunosorbent assay (ELISA) and positive results were confirmed by a recombinant immunoblot assay (RIBA). Our results demonstrated anti-HCV positivity in 69%, 11% and 4.3% of the EMC, MM and B-CLL samples tested, respectively. None of the RA patients were found to be anti-HCV positive. No significant differences were observed between the patients’ groups regarding the frequency of HBs Ag and anti-HBs Ab. Considering the low incidence of HCV infection in the control group and the normal population, these results confirm and extend previous reports on the possible role of HCV infection in the etiology of EMC and further suggest involvement of this virus in a subset of MM. C1 [Gharagozloo, Soheila] School of Medical Sciences, Tarbiat Modarres University,, Department of Immunology, 14155 Tehran, Iran. [Khoshnoodi, Jalal] School of Medical Sciences, Tarbiat Modarres University,, Department of Immunology, 14155 Tehran, Iran. [Shokri, Fazel] School of Medical Sciences, Tarbiat Modarres University,, Department of Immunology, 14155 Tehran, Iran. RP Shokri, F (reprint author), School of Medical Sciences, Tarbiat Modarres University,, Department of Immunology, 14155 Tehran, Iran. CR Yoshida M, Miyoshi I, Hinuma Y: Isolation and characterization of retrovirus from cell lines of human adult T-cell leukemia and its implication in the disease.Proc Natl Acad Sci USA70:2031- 2035, 1982. Chisari FV, Ferrari C: Hepatitis B virus immunopathogenesis. Ann Rev Immunol 13:29-60, 1995. Magrath I. The pathogenesis of Burkitt’s lymphoma. Adv Cancer Res 55:133-270, 1990. Choo QL, Kuo G, Weiner AJ, et al: Isolation of a cDNA derived from a blood-born non-A non-B viral hepatitis genome. Science 244:359-362, 1989. Kuo G, Choo QL, Alter HJ, et al. An assay for circulating antibodies to a major etiologic virus of human non-A, non-B hepatitis. Science 244:362-364, 1989. Choo QL, Weiner AJ, Overby LR, et al: Hepatitis C virus: The major causative agent of viral non-A non-B hepatitis. Br Med Bull 46:423-441, 1990. Tsukuma H, Hiyama T, Tanak S, et al: Risk factors for hepatocellular carcinoma among patients with chronic liver disease.N Engl J Med 328:1797-1801, 1993. Saito I, Miyamura T, Ohbayashi A, et al.Hepatitis C virus Infection is associated with the development of hepatocellular carcinoma. Proc Natl Acad Sci USA 87:6547-6549, 1990. Pascual M, Perrin L, Giotra E, et al: Hepatitis C virus in patients with cryoglobulinemia type II. J Inf Dis 162:5695, 1990. Ferri C, Greco F, Longombardo G, et al. Antibodies to hepatitis C virus in patients with mixed cryoglobulinemia. Arth Rheum 34:1606-1610, 1991. Ferri C, Zignego AL: Relation between infection and autoimmunity in mixed cryoglobulinemia.Curr opin Rheumatol 12:53- 60, 2000. Dammaco F, Sansonno D: Antibodies to hepatitis C virus in essential mixed cryoglobulinemia. Clin Exp Immunol 87:352- 356, 1992. Montagnino G: Reappraisal of the clinical expression of mixed cryoglobulinemia. Springer Semin Immunopathol 10:1-19, 1988. Dammaco F, Gatti P, Sansonno D: Hepatitis C virus infection, mixed cryoglobulinemia, and non-Hodgkin’s lymphoma: an emerging picture. Leuk Lymph 31:463-476, 1998. Arico M, Maggiore G, Silini E, et al. Hepatitis C virus infection in children treated for acute lymphoblastic leuemia. Blood 84:2919-2922, 1994. Luppi L, Longo G, Ferrari MG, et al: Additional neoplasms and HCV infection in low-grade lymphoma of MALT type. Br J Haematol 94:373-375, 1996. Gorevic PD, Kassab HJ, Levo Y et al. Mixed cryoglobulinemia: Clinical aspects and long- term follow-up of 40 patients. Am J Med 69:287-308, 1980. Brouet JC, Fermand JP: Multiple myeloma. In: Frank MM, Austen KF, Claman HN, Unanue ER, eds. Samters Immunologic Diseases. New York: Little Brown, 623-636, 1995. Rai KR, Sawitsky A, Gronkite EP, et al: Clinical staging of chronic lymphocytic leukemia. Blood 46:219-234, 1975. Shokri F, Mageed RA, Richardson P, et al: Modulation and high frequency expression of autoantibody associated cross-reactive idiotypes linked to the VH1 subgroup in CD5-expressing B lymphocytes from patients with chronic lymphocytic leukemia, B-CLL). Scand J Immunol 37:673-679, 1993. Shokri F, Mageed RA, Kitas GD, et al: Quantitation of crossreactive idiotype-positive rheumatoid factor produced in autoimmune diseases. An indicator of clonality and B cell proliferative mechanisms. Clin Exp Immunol 85:20-27, 1991. Shokri F, Mageed RA, Maziak BR, et al. Lymphoproliferation in primary Sjogren’s syndrome: Evidence of selective expansion of a B-cell subset characterized by the expression of cross-reactive idiotypes. Arth Rheum 36:1128-1136, 1993. Shokri F, Mageed RA, Jefferis R: A quantitative ELISA for measurement of rheumatoid factor associated cross-reactive idiotypes in serum from patients with rheumatic disease. Br J Rheum 32:862-869, 1993. Gharagozlou S, Khoshnoodi J, Sharifian RA, et al: Distribution of immunoglobulin light and heavy chain isotypes and subclasses in paraproteins isolated from Iranian patients with multiple myeloma. Ir J Med Sci 21:105-111, 1996. Sansonno D, De vita S, Cornacchiulo V, et al: Detection and distribution of hepatitis C virus-related proteins in lymph nodes of patients with type II mixed cryoglobulinemia and neoplastic or non-neoplastic lymphoproliferation. Blood 88:4638-4645, 1996. Artini M, Natoli G, Avantaggiati MI: Detection of replicative intermediates of viral RNA in peripheral blood mononuclear cells from chronic hepatitis C virus carriers. Arch Virol8:23-29, 1993. Karavattathayyil SJ, Kalkeri G, Liu HJ, et al: Detection of hepatitis C virus RNA sequences in B-cell non-Hodgkin lymphoma. Am J Clin Pathol 113:391-398, 2000. Paydas S, Kilic B, Sahin B, et al: Prevalence of hepatitis C virus infection in patients with lymphoproliferative disorders in southern Turkey. Br J Cancer 80:1303-1305, 1999. Nagayama R, Tsuda F, Okamoto H, et al: Genotype dependence of hepatitis C virus antibodies detectable by the first-generation enzyme-linked immunosorbent assay with C 100-3 protein. J Clin Invest 92:1529-1535, 1993. Lechmann M, Ihlenfeldt HG, Braunschweiger I, et al: T- and Bcell responses to different HCV antigens in patients with chronic hepatitis C infection and in healthy anti-HCV-positive blood donors without viremia. Hepatology 24:790-795, 1996. Weiner AJ, Geysen HM, Christopherson C, et al: Evidence for immune selection of hepatitis C virus putative envelope glycoprotein variants: Potential role in chronic HCV infections. Proc Natl Acad Sci USA 89:3468-3472, 1992. Rezvan H, Ahmadi J, Farhadi M: A preliminary study on the prevalence of anti-HCV amongst healthy blood donors in Iran. Vox Sang 67, Suppl.2): 100, Abstr), 1994. Luppi M, Longo G, Ferrari MG, et al: Clinico-pathological characterization of hepatitis C virus-related B-cell non- Hodgkin’s lymphomas without symptomatic cryoglobulinemia. Ann Oncol 9:495-498, 1998. Silvestri F, Barillari G, Fanin R, et al. Risk of hepatitis C virus infection, Waldenstrom’s macroglobulinemia, and monoclonal gammopathies. Blood 88:1125-1126, 1996. Mussini C, Ghini M, Mascia MT, et al: Monoclonal gammopathies and hepatitis C virus infection. Blood 85:1144-1145, 1995. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 135 EP 139 PG 5 ER PT J AU Gati, I Leel-Ossy, L AF Gati, Istvan Leel-Ossy, Lorant TI Heat Shock Protein 60 in Corpora Amylacea SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HSP 60; corpora amylacea; neurological disease ID HSP 60; corpora amylacea; neurological disease AB Heat shock protein 60 representation in the corpora amylacea of the brain was investigated in five different neurological diseases. In the cases with cerebral infarct, amyotrophic lateral sclerosis, multiple sclerosis, acute disseminated encephalomyelitis and primary tumors of the nervous system the corpora amylacea showed similar appearance with strong HSP-60 positivity in all investigated disorders at the predilection sites. In the inflammatory diseases, besides corpora amylacea, several cellular elements exhibited HSP-60 immunostaining too. In these cases, the widespread HSP-60 immunoreactivity associated with relative moderate corpora amylacea production as compared to other diseases. From this contradiction we concluded the corpora amylacea participate in the cellular stress reaction but stress protein synthesis certainly is not the primary event in corpora amylacea formation. In the development of the corpora amylacea the incipient process is most probably degenerative in nature, which later on is accompanied by stress protein synthesis and slow growing of these round structures designated for a protective role in the brain. However, the role of the stress protein synthesis in the corpora amylacea formation and growth was not unequivocally answered in this study. It is necessary to perform further comparative investigations of the stress protein representation and corpora amylacea formation in different diseases which may help in discovering useful pathogenetic data and the biological role of this degenerative structure. C1 [Gati, Istvan] University of Pecs, Department of Neurology, Ret u. 2., H-7623 Pecs, Hungary. [Leel-Ossy, Lorant] St. Borbala University Hospital, Department of Pathology, Neuropathological LaboratoryTatabanya, Hungary. RP Gati, I (reprint author), University of Pecs, Department of Neurology, H-7623 Pecs, Hungary. CR Ananthan J, Goldberg AL, Voellmy R: Abnormal proteins serve as eukaroyotic stress signals and trigger the activation of heat shock genes. Science 232:522-524, 1986. Bergstedt K, Hu B-R, Wieloch T: Initiation of protein synthesis and heat-shock protein-72 expression in the rat brain following severe insulin-induced hypoglycemia. Acta Neuropathol. 86:145-153, 1993. Chiechanover A: The ubiquitin-mediated proteolytic pathway. Brain Pathology 3:67-75, 1993. Chopp M: The roles of heat shock proteins and immediate early genes in the central nervous system normal function and pathology. Curr Opinion in Neurol Neurosurg. 6:6-10, 1993. Cisse S, Pery G, Lacoste-Royal G, et al: Immunochemical identification of ubiquitin and heat-shock proteins in corpora amyleacea from normal aged and Alzheimer’s disease brains. Acta Neuropathol. 85:233-240, 1993. DeNagel DC, Pierce SK: Heat Shock proteins in immune responses. Critical Rev Immunol. 13:71-81, 1993. Garofalo O, Kennedy PGE: Ubiquitin and heat shock protein expression in amyotrophic lateral sclerosis. Neuropathol Appl Neurobiol. 17:39-45, 1991. Jaattela M, Wissinf D: Emerging role of heat shock protein in biology and medicine. Annal Medicine 24:249-258, 1992. Leel-Ossy L, Gati I: Corpus amylaceum, polyglucosan body, in the peripheral olfactory system., Abstr., Brain Pathology 7:1113, 1997. Leel-Ossy L: Pathological significance and characteristics of corpus amylaceum. Neuropathology 11:105-114, 1991. Leel-Ossy L: The occurrence of corpus amylaceum, polyglucosan body, in diabetes mellitus. Neuropathology 15:108-111, 1995. Lowe J, Mayer RJ, Landon M: Ubiquitin in Neurodegenerative diseases. Brain Pathology 3:55-65, 1993. Lukacs KV, Lowrie DB, Stokes RW, et al: Tumor cells transfected with a bacterial heat-shock gene lose tumorigenicity and induce protection against tumors. J. Exp Med. 343-348, 1993. Martin JE, Mather KM, Swash MS, et al: Heat shock protein expression in corpora amylacea in the central nervous system: clues to their origin. Neuropathol Appl Neurobiol. 17:113-119, l991. Matsumoto S, Goto S, Kusaka H, et al: Ubiquitin-positive inclusion in the anterior horn cells in subgroups of motor neuron diseases: a comparative study of adult-onset amyotrophic lateral sclerosis, juvenile amyotrophic lateral sclerosis and Werdnig- Hoffmann disease. J Neurol Sci. 115:208-213, 1993. Ostermann J, Horowich AL, Neupert W, et al: Protein folding in mitochondria requres complex formation with hsp 60 and ATP hydrolysis. Nature 341:125-130, 1989. Peetermans WE: Heat-shock proteins in the medicine. Acta Clin Belg. 50:131-135, 1995. Pelham HRG: Speculation on the functions of the major heat shock and glucose-regulated proteins. Cell 46:959-961, 1986. Prabhakar S, Kurian E, Gupta RS: Heat shock protein immunoreactivity in CSF: Correlation with oligoclonal banding and demyelinating disease. Neurology 44:1644-1648 Santoro MG, Garaci E, Amici C, et al: Induction of heat shock protein synthesis by prostaglandins with antineoplastic and antiviral activity. Adv Prostagl Thromboxane Leukotr Res. 21:867-874, 1990. Tytell M, Barbee MF, Brown IR: Stress, Heat Shock, protein accumulation in the central nervous system. Its relationship to cell stress and damage. Adv in Neurol. 59:293-303, 1993. Yee WM, Frim DM, Isacson O: Relationships between stress protein induction and NMDA-mediated neuronal death in the entorhinal cortex. Exp Brain Res. 94:193-202, 1993. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 140 EP 144 PG 5 ER PT J AU Leel-Ossy, L AF Leel-Ossy, Lorant TI New Data on the Ultrastructure of the Corpus Amylaceum (Polyglucosan Body) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Corpus amylaceum; ultrastructure; scanning EM; polyglucosan body ID Corpus amylaceum; ultrastructure; scanning EM; polyglucosan body AB During the semiquantitative evaluation of the occurrence of the corpus amylaceum (CA) in a large quantity of autopsy and biopsy material (1,407 cases), electromicroscopical (EM) and scanning EM examinations were carried out on 8 autopsied cases where CA was demonstrated. The EM examinations appeared to underline the astrocytic origin of the CA formation, which is initiated in the astrocytic fiber system by glycogen and other carbohydrate polymers. The biophysics of the development of the CA is indicative of the same mechanisms as for (mainly intracellular) inclusion bodies. The large amount of CA that develops at the predilection sites is a consequence of metabolic damage, a large quantity of cerebrospinal fluid and recurring disturbances in the barrier functions. The abundant CA may cause secondary blood-brain barrier disturbances. This working hypothesis demands further investigations and the continuation of research by modern immunocytochemical and ultrastructural methods recommended. C1 [Leel-Ossy, Lorant] University Hospital of County Borsod-A.-Z., I. Department of Neurology and NeurosurgeryMiskolc, Hungary. RP Leel-Ossy, L (reprint author), University Hospital of County Borsod-A.-Z., I. Department of Neurology and Neurosurgery, Miskolc, Hungary. CR Anzil A, Herlinger H, Blinzinger K: Intraneuritic corpora amylacea. Virchows Arch. Path. Anat. 364:297-301, 1974. Austin JH, Sakai M: Corpora amylacea. In: Pathology of the Nervous System. Vol. 3., Ed.: Minckler J., McGraw-Hill Book Co., New York, 1972, pp. 2961-68. Cavanagh JB: Copora-amylacea and the family of polyglucosan diseases. Brain Research Review 29:255-295, 1999. Cervos-Navarro J: Polyglukosaneinschlusse im Nervengewebe. In: Pathologie des Nervensystems. V., Ed.: Cervos-Navarro J., Springer Verl, Berlin, Heidelberg, etc. 1991, S. 99-115. Cohrs P: Lehrbuch der spezielle pathologische Anatomie der Haustiere. VEB G. Fischer Verl, Jena 1954, S. 441 u. 690. Daems WTh, Perslin P: Demonstration of PAS-positive material in electron microscopy with lead staining. Histochemie 3:70-88, 1952. Gray F, Gherardi R, et al: Adult polyglucosan body disease, APBD). J Neuropathol Exp Neurol 47:459-474, 1988. Gueft B, Ghidoni JG: The site of formation and ultrastructure of amyloid. Amer J Pathol 43:837-854, 1963. Hara M: Microscopic globular bodies in the human brain. Ann Neurol 45:169-178, 1986. Hirano A: Some fine structural alterations of the central nervous system in aging. In: Proc. 7th Internat. Congr. Neuropathol., Eds.: Kornyey St, Tariska St, Gosztonyi G, Excerpta Med. Amsterdam, Akademiai Kiado, Budapest, 1975, pp. 83-90. Hirano A: Praktischer Leitfaden der Neuropathologie. Springer Verl, Berlin, Heidelberg, etc. 1983, S. 244. Hirano A Personal communication, Toronto 1992. Ikeda K, Kosaka K, Hori A: Intraneuronal polyglucosan bodies and peculiar inclusions in the midbrain-tegmentum of the aged. J Psychiatry Neurol. 41:719-723, 1987. Ketz HA: Die Altersveranderungen im Zentralnervensystem der Haustiere. Z Altersforschung 13:103-236, 1959. Kikuchi E: Uber die Altersveranderungen im Gehirn des Pferdes. Arch wis prakt Tierheilk. 58:541-573, 1957. Leel-Ossy L: The origin and the pathoogical significance of the corpus amylaceum. Acta Neuropathol., Berl, Suppl. II. 396- 399, 1981. Leel-Ossy L: Further observation on the structure and nature of the corpus amylaceum. Zbl allg Pathol pathol Anat 27:131-395, 1986. Leel-Ossy L: Pathological significance and characteristics of corpus amylaceum. Neuropathol., Japan, 11:105-114, 1991. Leel-Ossy L: A corpus amylaceum elofordulasanak statisztikai ertekelese es patologiai jelentosege. Ideggyogy Szemle, Hung, 50:90-102, 1997. Palmucc L, Anzil AP, Luh S: Intraastrocytic glycogen granules and corpora amylacea stain positively for polyglucosan: a cytochemical contribution on the fine structural polymorphism of particulate polysaccharides. Acta Neuropathol, Berl, 57:99- 102, 1982. Ramsey HI: Ultrastructure of corpora amylacea. J Neuropathol Exp Neurol. 24:25-39, 1965. Robitaille Y, Carpenter S, Karpati G, et al: A distinct form of adult polyglucosan body disease with massive involvement of central and peripheral neuronal processes and astrocytes. Brain 103:315-336, 1980. Roukema PA, Oderkerk CH: Isolation and preliminary characterization of corpora amylacea from human barin. Psychiat Neurol Neurochir. 73:87-96, 1970. Sakai M, Austin J, Witmer F, et al: Studies of corpora amylacea. I. Isolation and preliminary characterization by chemical and histochemical techniques. Arch Neurol. 21:526-544, 1969. Sharbati A, Carner M, Coletti V: Extrusion of corpora amylacea from the marginal glia at the vestibular root entry. J Neuropathol Exp Neurol. 55:196-201, 1996. Schochet SS, Jr: Neuronal inclusions. In: Structure and function of nervous tissue. Vol. 4., Ed: Bourne GH, Academic Press, New York, 1972, pp.129-177. Schwalbe HP, Quadbeck G: Die Corpora amylacea im menschlichen Gehirn. Virchows Arch Path Anat. 366:305-311, 1975. Seitelberger F: Myoclonus body disease. In: Pathology of the nervous system. Vol. 1., Ed: Minckler J, McGraw-Hill, New York, 1968, pp. 1121-1134. Takahashi K, Agari M, Nakamura H: Intra-axonal corpora amylacea in ventral and lateral horns of the spinal cord. Acta Neuropathol., Berl, 30:151-158, 1975. Tomlinson BE: The ageing brain. In: Recent advances in neuropathology., Eds: Thomas IW, et al., Churchill Livingstone, Edinburgh, London, New York, 1979, pp. 229-259. Tunon T, Bengoechea O, Narbona J: Glycogenosis with amylopectinoid deposits in a 13-year-old girl. Clin Neuropathol., Berl, 7:100-104, 1988. Yagashita S, Itoh Y, et al: Pleomorphic intra-neuronal polyglucosan bodies mainly restricted to the pallidum. Acta Neuropathol., Berl, 62:159-163, 1983. Yoshimura T: Beitrage zu den klinisch - und histopathologischen Untersuchungen uber die Falle der Lafora-ahnlichen Einschlusskorperchen. Folia Psychiat Neurol Jpn 1977. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 145 EP 150 PG 6 ER PT J AU Szekely, E Madaras, L Kulka, J Jaray, B Nagy, L AF Szekely, Eszter Madaras, Lilla Kulka, Janina Jaray, Balazs Nagy, Lajos TI Leiomyosarcoma of the Female Breast SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE breast tumors; leiomyosarcoma ID breast tumors; leiomyosarcoma AB Leiomyosarcomas of the breast are rare tumors. Less than 15 such cases have been reported in the literature so far. In this paper authors describe a case of leiomyosarcoma of a female breast presenting as a firm lobulated mass, mimicking a phylloid tumor radiographically. By fine needle aspiration biopsy, on the smears discohesive malignant looking cells were conclusive to a poorly differentiated invasive ductal carcinoma of the breast. The mastectomy specimen contained a lobulated mass, microscopically showing a partly epithelioid spindle cell tumor, immunoreactive for vimentin, desmin, smooth muscle actin antibodies, and negative for epithelial markers, hormone and growth factor receptors. Axillary lymph nodes were free of tumor. A primary leiomyosarcoma of the breast was diagnosed. C1 [Szekely, Eszter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Madaras, Lilla] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Jaray, Balazs] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Nagy, Lajos] Semmelweis University, 3rd Department of SurgeryBudapest, Hungary. RP Szekely, E (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM szesz@korb2.sote.hu CR Blanchard DK, Budde JM, Hatch GF: 3rd Tumors of the small intestine. World J Surg 24:421-429, 2000. Burke AP, Virmani R: Sarcomas of the Great Vessels. A Clinicopathologic study. Cancer, 71:1761-1773, 1993. Burke AP, Virmani R: Tumors of the heart and great vessels, AFIP, 1996. Falconieri G, Della Libera D, Zanconati F, Bittesini L: Leiomyosarcoma of the Female Breast. Am J Surg Pathol 108:19- 25, 1997. Glock Y, Laghzaoui A, Wang J: Fissured leiomyosarcoma of the descending thoracic aorta. A propos of a case and review of the literature. Arch Mal Coeur 90:1317-1320, 1997. Palacios G: Leiomyosarcoma of the Female Breast. Am J Surg Pathol 109:650-651, 1998. Majeski J, Crawford ES, Majeski EI: Primary aortic intimal sarcoma of the endothelial cell type with long term survival. J Vasc Surg 27:555-558, 1998. Mentzel T, Calonje E, Fletcher CDM: Leiomyosarcoma with prominent osteoclast-like giant cells Am J Surg Pathol 18:258- 265, 1994. Miracco C, Laurini L, Santopietro R: Intimal type primary sarcoma of the aorta. Report of a case with evidence of rhabdomyosarcomatous differentiation Virchows Arch 435:62-66, 1999. Patel KR, Niazi TBM, Anthony P: Griffiths.Massive osteolytic bone metastases from a primary aortic sarcoma: a case report. Hum Pathol 28:1306-1310, 1997. Pautier P, Genestie C, Rey A: Analysis of clinicopathologic prognostic factors for 157 uterine sarcomas and evaluation of a grading score validated for soft tissue sarcoma. Cancer 88:1425-1431, 2000. Rochais JP, Icard P, Coffin O: Intimal sarcoma of the thoracic aorta: a case report. Eur J Vasc Endovasc Surg 18:181-182, 1999. Roncaroli F, Eusebi V: Rhabdomyoblastic differentiation in a leiomyosarcoma of the retroperitoneum Hum Pathol 27:310- 313, 1996. Shimoda H, Oka K, Otani S: Vascular leiomyosarcoma arising from the inferior vena cava diagnosed by intraluminal biopsy. Virchows Arch 433:97-100, 1998. Soh LT, Chew SH, Ang L: Uterine leiomyosarcoma-a Singapore experience. Aust NZJ Obstet Gynecol 39:246-248 1999. Ugras S, Dilek ON, Karaayvaz M, et al: Primary Leiomyosarcoma of the Breast. Surg Today 27:1082-1085, 1997. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2001 VL 7 IS 2 BP 151 EP 153 PG 3 ER PT J AU Chatziantoniou, DV AF Chatziantoniou, D Vassilios TI Telomerase: Biological Function and Potential Role in Cancer Management SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE telomerase; telomeres; cancer; function; regulation; therapy ID telomerase; telomeres; cancer; function; regulation; therapy AB Telomeres are the specialized ends of eukaryotic chromosomes, thought to have many functions, most importantly serving as a clock signaling entry into cellular senescence. These structures are maintained by the reverse transcriptase telomerase, a peculiar enzyme in both structure, since it contains its own template RNA and function, since it is inactivated in most normal tissues but activated in the vast majority of malignant tumors. These features have made telomerase a subject of intense investigation, both to understand its cellular role and regulation and to exploit its activation in cancer to develop drugs or diagnostic methods based on telomerase. This work gathers all the information currently available in the biological and clinical fields of telomerase research. C1 [Chatziantoniou, D Vassilios] Semmelweis UniversityBudapest, Hungary. RP Chatziantoniou, DV (reprint author), Semmelweis University, Budapest, Hungary. CR Blackburn EH: Structure and function of telomeres. Nature 350:569-572, 1991. Kipling D., ed): The telomere. Oxford University Press, Oxford, 1995. Griffith JK, Comeau L, Rosenfield S, et al: Mammalian telomeres end in a large duplex loop. Cell 97:503-514, 1999. Jun-Ping Liu: Studies of the molecular mechanisms in the regulation of telomerase activity. FASEB J 13:2091-2104, 1999. Blackburn EH: Telomerases. Annu Rev Biochemistry 61:113- 129, 1992. Bhattacharyya A, Blackburn EH: A functional telomerase RNA swap in vivo reveals the importance of nontemplate RNA domains. Proc Natl Acad Sci USA 94:2823-2827, 1997. Beattie TL, Zhou W, Robinson MO, et al: Reconstitution of human telomerase activity in vitro. Current Biology 8:177-180, 1998. Kilian A, Bowtell DD, Abud HE, et al: Isolation of a candidate human telomerase catalytic subunit gene, which reveals complex splicing patterns in different cell types. Human Mol Genetic 6:2011-2019, 1997. Biessmann H, Walter MF, Mason JM: Drosophila telomere elongation. Ciba Fndn Symp 211:53-70, 1997. Zakian VA: Structure, function and replication of Saccharomyces cerevisiae telomeres. Annu Rev Genet 30:141-172, 1996. Wang J, Xie LY, Allan S, et al: Myc activates telomerase. Genes Dev 12:1769-1774, 1998. Kyo S, Takakura M, Kanaya T, et al: Estrogen activates telomerase. Cancer Res 59:5917-5921, 1999. Li H, Cao Y, Berndt MC, et al: Molecular interactions between telomerase and the tumor suppressor protein p53 in vitro. Oncogene 18:6785-6794, 1999. Van Steensel B, De Lange T: Control of telomere length by the human telomeric protein TRF1. Nature 385:740-743, 1997. Smith S, Giriat I, Schmitt A, et al: Tankyrase, a poly(ADPribose, polymerase at human telomeres. Science 282:1484- 1487, 1998. Wynford-Thomas D: p53: Guardian of cellular senescence. J Pathol 180:118-121, 1996. Li H, Zhao LL., Funder JW, Liu JP: Protein phosphatase 2A inhibits nuclear telomerase activity in human breast cancer cells. J Biol Chem 272:16729-16732, 1997. Li H, Zhao LL, Yang Z, et al: Telomerase is controlled by protein kinase Calpha in human breast cancer cells. J Biol Chem 273:33436-33442, 1998. Bickenbach JR, Vornwald D, Bachor C, et al: Telomerase is not an epidermal stem cell marker and is downregulated by calcium. J Invest Dermatol 111:1045-1052, 1998. Martens UM, Zijlmans JM, Poon SS, et al: Short telomeres on human chromosome 17p. Nature Genet 18:76-80, 1998. Vaziri H, Schachter F, Uchida I, et al: Loss of telomeric DNA during aging of normal and trisomy 21 human lymphocytes. Am J Human Genet 52:661-667, 1993. Hahn WC, Counter CM, Lundberg AS, et al: Creation of human tumour cells with defined genetic elements. Nature 400:464- 468, 1999. McKenzie KE, Umbricht CB, Sukumar S: Applications of telomerase research in the fight against cancer. Mol Med Today 5:114- 122, 1999. Davis AJ, Siu LL: Telomerase: therapeutic potential in cancer. Cancer Invest 18:269-277, 2000. Shay JW, Wright WE:Telomerase activity in human cancer.Curr Opin Oncol 8:66-71, 1996. Lichtsteiner SP, Lebkowski JS, Vasserot AP: Telomerase: A target for anticancer therapy. Ann NY Acad Sci 886:1-11, 1999. Minev B, Hipp J, Firat H, et al: Cytotoxic T cell immunity against telomerase reverse transcriptase in humans. Proc Natl Acad Sci 97:4796-4801, 2000. Hahn WC, Stewart WC, Brooks MW, et al.: Inhibition of telomerase limits the growth of human cancer cells. Nature Med 5:1164-1170, 1999 Duncan VE, Ajmani PS, Hughes JA: Cellular delivery is a major obstacle for oligodeoxynucleotide inhibition of telomerase activity. Anticancer Res 18:4105-4108, 1998. Sawant SG, Gregoire V, Dhar S, et a.: Telomerase activity as a measure for monitoring radiocurability of tumor cells. FASEB J 13:1047-1054, 1999. Greider CW: Telomere length regulation. Annu Rev Biochem 65:337-365, 1996. Harley CB, Futcher AB, Greider CW: Telomeres shorten during aging of human fibroblasts. Nature 345:458-460, 1990. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2001 VL 7 IS 3 BP 161 EP 170 PG 10 ER PT J AU Esteva, JF Hortobagyi, NG Sahin, AA Smith, LT Chin, MD Liang, ShY Pusztai, L Buzdar, UA Bacus, SS AF Esteva, J Francisco Hortobagyi, N Gabriel Sahin, A Aysegul Smith, L Terry Chin, Mon Dot Liang, Shang-Ying Pusztai, Lajos Buzdar, U Aman Bacus, S Sarah TI Expression of erbB/HER Receptors, Heregulin and P38 in Primary Breast Cancer using Quantitative Immunohistochemistry SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tumor markers; biological; proto-oncogene proteins; immunohistochemistry; ligands; breast neoplasm ID Tumor markers; biological; proto-oncogene proteins; immunohistochemistry; ligands; breast neoplasm AB The purpose of this study was to investigate the frequency of expression of the erbB/HER family of growth factor receptors, their ligand heregulin, and the two different signaling pathways p38 and mitogen-activated protein kinase (MAPK), as well as the status of HER-2 phosphorylation in tumor specimens from patients with primary breast cancer. The level of expression of these proteins was measured by quantitative immunohistochemistry combined with microscope-based image analysis in paraffin-embedded breast cancer tissue from 35 patients. The frequency of expression was: EGFR (51%), HER-2 (54%), P-HER-2 (48%), HER-3 (48%), HER-4 (57%), heregulin (48%), p38 (17%), MAPK (48%). There was evidence of associations among the coexpression of heregulin, EGFR, HER-2, and HER-3. Also, there was evidence of a positive association between P-MAPK and HER-4. HER-3 was expressed at high levels in patients younger than 50 years of age. There was a trend for expression of higher levels of HER-4 in tumors larger than 2 cm. The expression of EGFR, HER-2, heregulin, p38 and MAPK was independent of age, tumor size, number of lymph nodes involved or hormone receptor status. The HER family of growth factor receptors appear to be regulated independently in invasive breast cancer. Assessing the expression of multiple tumor markers by quantitative immuno-histochemistry is feasible. Further research is needed to determine the prognostic and predictive roles of the various associations between HER receptors, their ligands and signal transduction molecules in patients with early-stage breast cancer. C1 [Esteva, J Francisco] The University of Texas, M. D. Anderson Cancer Center, Department of Breast Medical Oncology, 1515 Holcombe Boulevard, 77030-4095 Houston, Texas, USA. [Hortobagyi, N Gabriel] The University of Texas, M. D. Anderson Cancer Center, Department of Breast Medical Oncology, 1515 Holcombe Boulevard, 77030-4095 Houston, Texas, USA. [Sahin, A Aysegul] The University of Texas M.D. Anderson Cancer Center, Department of PathologyHouston, Texas, USA. [Smith, L Terry] The University of Texas M.D. Anderson Cancer Center, Department of BiostatisticsHouston, Texas, USA. [Chin, Mon Dot] Quantitative Diagnostics LaboratoryElmhurst, Illinois, USA. [Liang, Shang-Ying] The University of Texas M.D. Anderson Cancer Center, Department of BiostatisticsHouston, Texas, USA. [Pusztai, Lajos] The University of Texas, M. D. Anderson Cancer Center, Department of Breast Medical Oncology, 1515 Holcombe Boulevard, 77030-4095 Houston, Texas, USA. [Buzdar, U Aman] The University of Texas, M. D. Anderson Cancer Center, Department of Breast Medical Oncology, 1515 Holcombe Boulevard, 77030-4095 Houston, Texas, USA. [Bacus, S Sarah] The University of Texas, M. D. Anderson Cancer Center, Department of Breast Medical Oncology, 1515 Holcombe Boulevard, 77030-4095 Houston, Texas, USA. RP Esteva, JF (reprint author), The University of Texas, M. D. Anderson Cancer Center, Department of Breast Medical Oncology, 77030-4095 Houston, USA. CR Bacus SS, Gudkov AV, Esteva FJ, et al: Expression of erb-B receptors and their ligands in breast cancer: implications to biological behavior and therapeutic response. Breast Disease 11:63-75, 2000. Schechter AL, Hung MC, Vaidyanathan L, et al: The neu gene: an erbB-homologous gene distinct from and unlinked to the gene encoding the EGF receptor. Science 229:976-978, 1985. Dickson RB, Lippman ME: Growth factors in breast cancer. Endocr Rev 16:559-589, 1995. Slamon DJ, Clark GM, Wong SG, et al: Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235:177-182, 1987. Gasparini G, Gullick WJ, Bevilacqua P, et al: Human breast cancer: prognostic significance of the c-erb B-2 oncoprotein compared with epidermal growth factor receptor, DNA ploidy, and conventional pathologic features. J Clin Oncol 10:686-695, 1992. Harris AL, Nicholson S, Sainsbury R, et al: Epidermal growth factor receptor and other oncogenes as prognostic markers. J Natl Cancer Inst 181-187, 1992. Muss HB, Thor AD, Berry DA, et al: c-erbB-2 expression and response to adjuvant therapy in women with node-positive early breast cancer. N Engl J Med 330:1260-1266, 1994. Thor AD, Berry DA, Budman DR, et al: erbB-2, p53, and efficacy of adjuvant therapy in lymph node-positive breast cancer. J Natl Cancer Inst 90:1346-1360, 1998. Paik S, Bryant J, Park C, et al: erbB-2 and response to doxorubicin in patients with axillary lymph node-positive, hormone receptor-negative breast cancer. J Natl Cancer Inst 90:1361- 1370, 1998. Gusterson BA, Gelber RD, Goldhirsch A, et al: Prognostic importance of c-erbB-2 expression in breast cancer. International, Ludwig, Breast Cancer Study Group. J Clin Oncol 10:1049- 1056, 1992. Baselga J, Seidman A, Rosen PP, et al: HER2 overexpression and paclitaxel sensitivity in breast cancer: therapeutic implications. Oncology 11:43-48, 1997. Carlomagno C, Perrone F, Gallo C, et al: c-erb B2 overexpression decreases the benefit of adjuvant tamoxifen in early-stage breast cancer without axillary lymph node metastases. J Clin Oncol 14:2702-2708, 1996. Elledge RM, Green S, Ciocca D, et al: HER-2 expression and response to tamoxifen in estrogen receptor-positive breast cancer: a Southwest Oncology Group Study. Clin Cancer Res 4:7- 12, 1998. Lemoine NR, Barnes DM, Hollywood DP, et al: Expression of the ERBB3 gene product in breast cancer. Br J Cancer 66:1116- 1121, 1992. Kumar NB, Cantor A, Allen K, et al: Android obesity at diagnosis and breast carcinoma survival – Evaluation of the effects of anthropometric variables at diagnosis, including body composition and body eat distribution and weight gain during life span, and survival from breast carcinoma. Cancer 88:2751- 2757, 2000. Gullick WJ: The c-erbB3/HER3 receptor in human cancer. Cancer Surveys 27:339-349, 1996. Kew TY, Bell JA, Pinder SE, et al: c-erbB-4 protein expression in human breast cancer. British Journal of Cancer 82:1163- 1170, 2000. Xia Z, Dickens M, Raingeaud J, et al: Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science 270:1326- 1331, 1995. Bacus SS, Plowman G, Yarden Y: Expression of erbB-2 receptor family and their ligands: implication to breast cancer biological behavior, Meeting abstract). Breast Cancer Res Treat 32:93-93, 1994. Tang MTC, Weiss NS, Daling JR, et al: Case-control differences in the reliability of reporting a history of induced abortion. Amer J Epidemiol 151:1139-1143, 2000. Drumea KC, Levine E, Bernstein J, et al: ATM heterozygosity and breast cancer: screening of 37 breast cancer patients for ATM mutations using a non-isotopic RNase cleavage-based. Breast Cancer Res Treat 61:79-85, 2000. Wang TTY, Jeng JJ: Coordinated regulation of two TRAILR2/ KILLER/DR5 mRNA isoforms by DNA damaging agents, and 17 beta-estradiol in human breast cancer cells. Breast Cancer Research and Treatment 61:87-96, 2000. Plowman GD, Culouscou JM, Whitney GS, et al: Ligand-specific activation of HER4/p180erbB4, a fourth member of the epidermal growth factor receptor family. Proc Natl Acad Sci USA 90:1746-1750, 1993. Knowlden JM, Gee JM, Seery LT, et al: c-erbB3 and c-erbB4 expression is a feature of the endocrine responsive phenotype in clinical breast cancer. Oncogene 17:1949-1957, 1998. Lupu R, Lippman ME: William L. McGuire Memorial Symposium. The role of erbB2 signal transduction pathways in human breast cancer. Breast Cancer Res Treat 27:83-93, 1993. Peles E, Ben-Levy R, Tzahar E, et al: Cell-type specific interaction of Neu differentiation factor, NDF/heregulin, with Neu/HER-2 suggests complex ligand-receptor relationships. EMBO J 12:961-971, 1993. Bacus SS, Gudkov AV, Zelnick CR, et al: Neu differentiation factor, heregulin, induces expression of intercellular adhesion molecule 1: implications for mammary tumors. Cancer Res 53:5251-5261,1993. Bacus SS, Gudkov AV, Esteva FJ, et al: Expression of erb-B Receptors and Their Ligands in Breast Cancer: Implications to Biological Behavior and Therapeutic Response, in Liu E, ed): The Breast, 2000. Daly JM, Olayioye MA, Wong AM, et al: NDF/heregulininduced cell cycle changes and apoptosis in breast tumour cells: role of PI3 kinase and p38 MAP kinase pathways. Oncogene 18:3440-3451, 1999. Liu W, Li J, Roth RA: Heregulin regulation of Akt/protein kinase B in breast cancer cells. Biochem Biophys Res Com 261:897- 903, 1999. Altiok S, Batt D, Altiok N, et al: Heregulin induces phosphorylation of BRCA1 through phosphatidylinositol 3-Kinase/AKT in breast cancer cells. J Biol Chem 274:32274-32278, 1999. Aguilar Z, Akita RW, Finn RS, et al: Biologic effects of heregulin/ neu differentiation factor on normal and malignant human breast and ovarian epithelial cells. Oncogene 18:6050- 6062, 1999. Bacus SS, Stancovski I, Huberman E, et al: Tumor-inhibitory monoclonal antibodies to the HER-2/Neu receptor induce differentiation of human breast cancer cells. Cancer Res 52:2580- 2589, 1992. Romashkova JA, Makarov SS: NF-kappaB is a target of AKT in anti-apoptotic PDGF signalling. Nature 401: 86-90, 1999. Majewski M, Nieborowska-Skorska M, Salomoni P, et al: Activation of mitochondrial Raf-1 is involved in the antiapoptotic effects of Akt. Cancer Research 59:2815-2819, 1999. Xu FJ, Stack S, Boyer C, et al: Heregulin and agonistic antip185( c-erbB2, antibodies inhibit proliferation but increase invasiveness of breast cancer cells that overexpress p185(cerbB2): increased invasiveness may contribute to poor prognosis. Clin Cancer Res 3:1629-1634, 1997. Harris LN, Yang L, Tang C, et al: Induction of sensitivity to doxorubicin and etoposide by transfection of MCF-7 breast cancer cells with heregulin beta-2. Clin Cancer Res 4:1005- 1012, 1998. Bacus SS, Gudkov AV, Lowe M, et al. Taxol-induced cytotoxicity: apoptotic signaling via MAP kinase pathways. Oncogene. 20:147-155, 2001. Komurasaki T, Toyoda H, Uchida D, et al: Epiregulin binds to epidermal growth factor receptor and ErbB-4 and induces tyrosine phosphorylation of epidermal growth factor receptor, ErbB-2, ErbB-3 and ErbB-4. Oncogene 15:2841-2848, 1997. Tzahar E, Pinkas-Kramarski R, Moyer JD, et al: Bivalence of EGF-like ligands drives the ErbB signaling network. EMBO J 16:4938-4950, 1997. Pinkas-Kramarski R, Lenferink AE, Bacus SS, et al: The oncogenic ErbB-2/ErbB-3 heterodimer is a surrogate receptor of the epidermal growth factor and betacellulin. Oncogene 16:1249- 1258, 1998. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2001 VL 7 IS 3 BP 171 EP 177 PG 7 ER PT J AU Pal, J Somogyi, Cs Szmolenszky, Szekeres, Gy Sipos, J Hegedus, G Martzinovits, I Molnar, J Nemeth, P AF Pal, Jozsef Somogyi, Csilla Szmolenszky, Agnes Szekeres, Gyorgy Sipos, Jozsef Hegedus, Geza Martzinovits, Ilona Molnar, Janos Nemeth, Peter TI Immunohistochemical Assessment and Prognostic Value of Hepatitis B Virus X Protein in Chronic Hepatitis and Primary Hepatocellular Carcinomas using anti-HBxAgMonoclonal Antibody SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE primary hepatocellular carcinoma; hepatitis-B X protein; chronic B virus hepatitis; monoclonal anti-HBx antibody; immunohistochemistry ID primary hepatocellular carcinoma; hepatitis-B X protein; chronic B virus hepatitis; monoclonal anti-HBx antibody; immunohistochemistry AB Hepatitis B virus (HBV) is the most meaningful risk factor in chronic hepatitis, cirrhosis and primary hepatocellular carcinoma (PHC). The hepatitis B virus X protein (HBxAg) is a multifunctional protein with many important functions in hepatocellular carcinogenesis. A monoclonal anti-HBxAg antibody was developed in our laboratory and characterized by different methods. Using this antibody HBxAg was detected in formaldehyde fixed paraffin embedded tissue sections of 72 liver biopsies from patients with acute hepatitis, chronic hepatitis, cirrhosis and primary hepatocellular carcinoma. The co-expression of hepatitis B surface antigen (HBsAg), hepatitis B core antigen (HBcAg) and HBxAg was compared. The histological and cytological localization of the detected HBxAg showed a characteristic distribution in different stages of HBV infection. Strong and diffuse nuclear reaction was detected in PHC cases in contrast to the focal, cytoplasmic and nuclear labeling in the acute and chronic B hepatitis cases. Our antibody seems to be a suitable prognostic marker for routine pathohistological diagnosis and for comparative pathological and epidemiological research on the development of PHC. C1 [Pal, Jozsef] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Somogyi, Csilla] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Szmolenszky, Agnes] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Szekeres, Gyorgy] Hisztopatologia KFTPecs, Hungary. [Sipos, Jozsef] County Hospital of Zala, Department of PathologyZalaegerszeg, Hungary. [Hegedus, Geza] County Hospital of Baranya, Department of PathologyPecs, Hungary. [Martzinovits, Ilona] University of Szeged, Albert Szent-Gyorgyi Clinical Centre, Institute of Clinical MicrobiologySzeged, Hungary. [Molnar, Janos] University of Szeged, Albert Szent-Gyorgyi Clinical Centre, Institute of Clinical MicrobiologySzeged, Hungary. [Nemeth, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. RP Nemeth, P (reprint author), University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, H-7643 Pecs, Hungary. EM peter.nemeth@aok.pte.hu CR Andrisani OM, Barnabas S: The transcriptional function of the hepatitits B virus X protein and its role in hepatocarcinogenesis., Review, Internat J Oncol 15:373-379, 1999. Balogh P, Szekeres Gy, Nemeth P: Hapten-mediated identification of cell membrane antigens using an anti-FITC monoclonal antibody. J Immunol Method 169:35-40, 1994. Brechot C: Molecular Mechanisms of Hepatitis B and C Viruses Related to Liver Carcinogenesis. Hepato-Gastroenterology 45:1189-1196, 1998. Diamantis ID, McGandy CE, Chen TJ, et al: Hepatitis B X-gene expression in hepatocellular carcinoma. J Hepatol 15:400-403, 1992. Doria M, Klein N, Lucito R, et al: The hepatitis B virus HBx protein is a dual specificity cytoplasmic activator of Ras and nuclear activator of Transcription factor. EMBO J 14:4747- 4757, 1995. Engwall E, Perlmann P: Enzyme-linked immunossorbent assay, ELISA. 3. Quantitation of specific antibodies by enzyme- labelled anti-immunglobulin in antigen-coated tubes. J Immunol 109:129-135, 1972. Feitelson MA, Clayton MM: X antigen polypeptides in the sera of hepatitis B virus-infeceted patients. Virology 177:367-371, 1990. Feitelson MA, Clayton MM, Blumberg BS: X antigen/antibody markers in hepadnavirus infections. Gastroenterology 98:1071- 1078, 1990. Feitelson MA: Hepatitis B virus infection and primary hepatocellular carcinoma. Clin Microbiol Rev 5:275-301, 1992. Feitelson MA, Zhu M, Duan L-X and London WT: Hepatitis B x antigen and p53 are associated in vitro and in liver tissues from patients with primary hepatocellular carcinoma. Oncogene 8:1109-1117, 1993. Feitelson MA: Hepatitis B Virus in hepatocarcinogenesis J Cellular Physiol 181:188-202, 1999. Feitelson MA, Duan L: Hepatitis B Virus x Antigen in the Pathogenesis of Chronic infections and the Development of Hepatocellular Carcinoma.(Review). Amer J Pathol 150:4, 1997. Gupta A, Mal TK, Jayasuryan N, et al: Assignment of disulphide bonds in the X protein, HBx, of hepatitis B virus. Biochem Biophys Res Comm 212:919-924, 1995. Haruna Y, Hayashi N, Katayama K, et al: Expression of X protein and hepatitis B virus replication in chronic hepatitis. Hepatology 13:417-421, 1991. Haviv I, Shamay M, Doits G, et al: Hepatitis B virus pX targets TFIIB in transcription coactivation. Mol Cell Biol 18:1562- 1569, 1998. Henkler F, Hoare J, Waseem N, et al: A: Intracellular localization of the hepatitis B virus HBx protein. J General Virol 82:871-882, 2001. Hess J, Stemler M, Will H, et al: Frequent detection of antibodies to hepatitis B x-protein in acute, chronic and resolved infections. Medical Microbiol Immunol 177:195-205, 1988. Katayama K, Hayashi N, Sasaki Y, et al: Detection of Hepatitis B Virus X Gene protein and Antibody in Type B Chronic Liver DiseaseGastroenterology 97:990-998, 1989. Kay A, Dinechin SD,Vitvitski-Trepo L, et al: Recognition of the N-Terminal, C-Terminal, and Interior Portions of HBx by Sera From Patients With Hepatitis B. J Med Virol 33:228-235, 1991. Kim CM, Koike K, Saito I, et al: HBx gene of hepatitis B virus induced liver cancer in transgenic mice. Nature 351:317-320, 1991. Kohler G, Milstein C: Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256:495-497, 1975. Kumar V, Jayasuryan N, Reddi H, et al: A Monoclonal Antibody Against the X Protein of Hepatitis B Virus: Fine Mapping of its Epitope and Application in a Quantitative ELISA of the X Protein in Sera of Hepatitis B Patients. Hybridoma 17:2, 1998. Laemmli UK: Cleave of structural proteins during the assembly of the bacteriophage T4. Nature 227:680-685, 1970. Levrero M, Jean-Jean O, Balsano C, et al: Hepatitis B virus polypeptide x gene expression in human cells and anti-HBx antibodies detected in chronic HBV infection. Virology 174:299-304, 1990. Malik AH, William M: Chronic Hepatitis B Virus Infection: Treatment Strategies for the Next Millenium Lee. Ann Intern Med 132:723-731, 2000. Marczinovits I, Somogyi Cs, Patthy A, et al: An alternative purification protocol for producing hepatitis B virus X antigen on a preparative scale in Escherichia coli. J Biotechol 56:81-88, 1997. Murakami S: Hepatitis B Virus X Protein:Sturcture, Function and Biology. Intervirology 42:81-99, 1999. Nomura T, Lin Y, Dorjsuren D, et al: Human Hepatitis B virus X protein is detectable in nuclei of transfected cells, and active for transactivation. Biochim Biophys Acta 1453:330-340, 1999. Pfaff E, Salfeld J, Gmelin K, et al: Synthesis of the X-protein of hepatitis B virus in vitro and detection of anti-X antibodies in human sera. Virology 158:456-460, 1987. Poussin K, Dienes H, Sirma H, et al: Expression of Mutated hepatitis B Virus x Genes in Human Hepatocellular Carcinomas. Int J Cancer 80:497-505 1999. Seo J, Kim K, Murakami S, et al: Lack of Colocalization of HBxAg and Insulin like Growth Factor II in the Livers of Patients with Chronic Hepatitis B, Cirrhosis and Hepatocellular Carcinoma. JKMS 12:523-531, 1997. Siddiqui A, Jameel S, Mapoles J: Expression of the hepatitis B virus X gene in mammalian cells. Proc Natl Acad Sci USA 84:2513-2517, 1987. Sirma H, Weil R, Rosmorduc O, et al: Cytosol is the prime compartment of hepatitis B virus X protein where it colocalizes with proteasome. Oncogene 16:2051-2063, 1998. Stemler M, Weimer T, Tu Z-X, et al: Mapping of B-cell epitopes of the human hepatitis B virus X protein. J Virol 64:2802-2809, 1990. Su Q, Schroder CH, Hoffmann WJ, et al: Expression of Hepatitis B Virus X Protein in HBV-infected human Livers and hepatocellular Carcinomas. Hepatology 27:1109-1120, 1998. Takeuchi M, Fujimoto J, Niwamoto, et al: Frequent Detection of Hepatitis B Virus X-gene DNA in Hepatocellular Carcinoma and Adjacent Liver Tissue in Hepatitis B Surface Antigen-Negative Patients. Digestive Dis Sci 42:2264-2269, 1997. Towbin H, Staehelin T, Gordon J: Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure an some applications. Proc Natl Acad Sci USA 76:4350- 4354, 1979. Unger T, Shaul Y: The X protein of the hepatitis B virus acts as a transcription factor when targeted to its responsive element. EMBO J 9:1889-1895, 1990. Vitvitski-Trepo L, Kay A, et al: Early and Freequent Detection of HBxAg and/or Anti-HBx in Hepatitis B Virus Infection. Hepatology 12:1278-1283, 1990. Wang W, London WT, Lega L, et al: HBxAg in the liver from carrier patients with chronic hepatitis and cirrhosis. Hepatology 14:29-37, 1990. Wang W, London WT, Feitelson, MA: Hepatitis B x Antigen in Hepatitis B Virus Carrier Patients with Liver Cancer Cancer Research 51:4971-4977, 1991. Wang XW, Forrester K, Yeh H, et al: Hepatitis B virus X protein inhibits p53 sequence-specific DNA binding, transcriptional activity, and association with transcription factor ERCC3. Proc Natl Acad Sci USA 91:2230-2234, 1994. Zentgraf H, Herrman G, Klein R, et al:Mouse monoclonal antibody directed against hepatitis B virus X protein synthesized in Escherichia coli: detection of reactive antigen in liver cell carcinoma and chronic hepatitis. Oncology 47:143-148, 1990. Zhu M, London WT, Duan L-X, et al: The value of hepatitis B x antigen as a prognostic marker in the development of hepatocellular carcinoma. Int J Cancer 55:571-576, 1993. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2001 VL 7 IS 3 BP 178 EP 184 PG 7 ER PT J AU Altinoz, AM Ozar, E Taskin, M Bozcali, E Bilir, A Altug, T Aydiner, A Sav, A AF Altinoz, A Meric Ozar, Engin Taskin, Murat Bozcali, Evin Bilir, Ayhan Altug, Tuncay Aydiner, Adnan Sav, Aydin TI Vascularization Pattern of C6 Glioma is Modified with Medroxyprogesterone Acetate and Ibuprofen in Wistar Rat Brain SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE medroxyprogesterone; ibaprofen; glioma; angiogenesis ID medroxyprogesterone; ibaprofen; glioma; angiogenesis AB Beneficial effects of medroxyprogesterone acetate (MPA) in cancer therapy is partly mediated via its antiangiogenic activity. The same is true for the antitumoral action of non-steroidal antiinflammatory drugs. We have studied two liposoluble drugs, MPA and the analgesic ibuprofen, on glioma vascularization in vivo. In this study we have shown that, until the sacrifice at 27. day after tumor inoculation in the right hemisphere, MPA had a slight though insignificant activity to reduce the fatality of C6 glioma, growing in right cerebral hemisphere of male Wistar rats. But ibuprofen both alone or with MPA had no effect on survival with gavage application of a 30 mg/kg/day dosing regime. On histological analysis, intra- and peritumoral vessels were counted. Progesterone seemed to lower intratumoral, but to increase peritumoral vessels, especially glomeruloids, around the tumor mass. Coadministration of ibuprofen acted to suppress the peritumoral vessel increase, and to enhance lymphomonocytic infiltration around tumor vessels. C1 [Altinoz, A Meric] Faculty of Medicine, Istanbul University, Department of Histology and EmbryologyBakirkoy, Turkey. [Ozar, Engin] Bakirkoy Mental Diseases Hospital, 1 st Neurosurgery ClinicBakirkoy, Turkey. [Taskin, Murat] Bakirkoy Mental Diseases Hospital, 1 st Neurosurgery ClinicBakirkoy, Turkey. [Bozcali, Evin] Faculty of Medicine, Istanbul University, Department of Histology and EmbryologyBakirkoy, Turkey. [Bilir, Ayhan] Faculty of Medicine, Istanbul University, Department of Histology and EmbryologyBakirkoy, Turkey. [Altug, Tuncay] Istanbul Medical Faculty, Istanbul University, Oncology InstituteIstanbul, Turkey. [Aydiner, Adnan] Istanbul Medical Faculty, Istanbul University, Oncology InstituteIstanbul, Turkey. [Sav, Aydin] Marmara University, Institute of Neuroscience, Talia Bali Aykan Neuropathology Laboratory, General Sukru Kanatli Sok. No: 36/8Bakirkoy, Turkey. RP Sav, A (reprint author), Marmara University, Institute of Neuroscience, Talia Bali Aykan Neuropathology Laboratory, Bakirkoy, Turkey. EM maltinoz@hotmail.com CR Blei F, Wilson EL, Mignatti P, et al: Mechanism of action of angiostatic steroids: Suppression of plasminogen activator activity via stimulation of plasminogen activator inhibitor synthesis. J Cell Physiology 155:568-578, 1993. Bourdon MA, Wikstrand CJ, Furthmayr H, et al: Human glioma-mesenchymal extracellular matrix antigen defined by monoclonal antibody. Cancer Res 43:2796-2805, 1983. Chow G, Woronick A, Kinkade P, et al: Pharmacological modulation of plasminogen activator secretion by P388D1 cell line. Agents Actions 21:387-389, 1987. Farrell CL, Megyesi JM, Maestro FRD: Effect of ibuprofen on tumor growth in the C6 spheroid implantation glioma model. J Neurosurg 68:925-930, 1998. Fujimoto J, Hosoda S, Fujita H, et al: Effect of medroxyprogesterone acetate on secondary spreading of endometrial cancer. Invasion Metastasis 9:209-215, 1989. Haddad SF, Moore SA, Schelper RL, et al: Smooth muscle can comprise the sarcomatous component of gliosarcomas. J Neuropathol Exp Neurol 51:493-498, 1992. Haddad SF, Moore SA, Schelper RL, et al: Vascular smooth muscle hyperplasia underlies the formation of glomeruloid vascular structures of glioblastoma multiforme. J Neuropathol Exp Neurol 51:493-498, 1992. Hyder SM, Murthy L, Stancel GM, et al: Progestin regulation of vascular endothelial growth factor in human breast cancer cells. Cancer Res 58:392-395, 1998. Iwamoto T, Tamaki K, Nakayama M, et al: Effect of endothelin 1 on fibrinolysis and plasminogen activator inhibitor 1 synthesis in rat mesangial cells. Nephron 73:273-297, 1996. Jikihara H., Terada N., Yamamoto R., et al: Inhibitory effect of medroxyprogesterone acetate on angiogenesis induced by human endometrial cancer. Am J Obstet Gynecol 167:207-211, 1992. Khalid H, Shibata S, Kishikawa M, et al: Immunohistochemical analysis progesterone receptor and Ki-67 labelling index in astrocytic tumors. Cancer 80:2133–2140, 1997. Kono S, Rao JS, Bruner JM, et al: Immunohistochemical localization of plasminogen activator type 1 in human brain tumors. J Neuropathol Exp Neurol 53:256-262, 1994. Nagashima T, Hoshino T, Cho KG : Proliferative potential of vascular components in human glioblastoma multiforme. Acta Neuropathol 73:301-305, 1987. Nguyen G, Delarue F, Berrou J, et al: Specific receptor binding of renin on human mesangial cells in culture increases plasminogen activator inhibitor-1 antigen. Kidney Int 50:1897- 1903, 1996. Okada N, Fushimi M, Nagata Y, et al: Evaluation of angiogenic inhibitors with an in vivo quantitative angiogenesis method using agarose microencapsulation and mouse hemoglobin enzyme-linked immunosorbent assay. Jap J Cancer Res 87:952- 957, 1996. Palayoor ST, Bump EA, Calderwood SK, et al: Combined antitumor effect of radiation and ibuprofen in human prostate carcinoma cells. Clin Canc Res 4:763-771, 1998. Rao BR: Pregnancy associated highly vascularized tumors negatively correlate with the levels of antiangiogenic 17 alphahydroxy- progesterone. Anticancer Res 17:1019-1021, 1999. Rutten EH, Doesburg WH, Slooff JL : Histologic factors in the grading and prognosis of astrocytoma grade I-IV, J Neurooncol 13:223-230, 1992. Selby DM, Woodard CA, Henry ML, et al: Are endothelial cell patterns indicative of grade. In vivo 11:371-375, 1997. Slowik F, Jellinger K, Gaszo L, et al: Gliosarcomas: histological, immunohistochemical, ultrastructural, and tissue culture studies, Acta Neuropathol(Berl), 67:201-210, 1985. Wadgymar A, Halloran PF: Crossreactions between an I-A allospecificity and the cytoskeleton of glomerular epithelium and of vascular smooth muscle. Transplantation 43:93-98, 1987. Yoshida Y, Kumanishi T, Abe S: Glomeruloid blood vessels in ethylnitrosourea-induced rat gliomas. Histological and immunohistochemical studies. Acta Neuropathol 79:240-247, 1989. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2001 VL 7 IS 3 BP 185 EP 189 PG 5 ER PT J AU Aziz, AS Pervez, Sh Khan, Sh Kayani, N Azam, IS Rahbar, HM AF Aziz, Abdus Syed Pervez, Shahid Khan, Shaista Kayani, Naila Azam, Igbal Syed Rahbar, Hussain Mohammed TI Significance of Immunohistochemical c-ErbB-2 Product Localisation Pattern for Prognosis in Human Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE c-ErbB-2; breast cancer; prognosis; drug resistance; nodal metastases ID c-ErbB-2; breast cancer; prognosis; drug resistance; nodal metastases AB Breast cancer is an increasingly important cause of illness and death among women. In recent years several novel prognostic determinants of breast cancer have been identified, including c-ErbB-2. In this study, expression of c-ErbB-2 in breast carcinoma was correlated with axillary lymph node metastases and disease outcome. The expression of c-ErbB-2 oncoprotein was analysed in 315 tumor specimens of infiltrating ductal carcinoma of breast. They were categorized according to the modified Bloom and Richardson criteria into three histological grades. These patients also had axillary lymph nodes sampling. The expression of c-ErbB-2 oncoprotein was analysed immunohistochemically. Over expression of c-ErbB-2 were observed in 39.36% tumors. Axillary lymph node metastasis had significant correlation with intensified positivity of c-ErbB-2. C-ErbB-2 positive patients did show resistance to chemotherapy when compared for recurrence and distant metastases following surgery (p< 0.05). At a median follow-up of 48 months in c-ErbB-2 positive patients, the overall survival was 3.0 years and disease free survival was 2.5 years. c-ErbB-2 negative tumor patients showed a far better survival. In this group the overall survival was 4.44 years and the disease free survival was 3.78 years. These findings reinforce the view that c-ErbB-2 immunohistochemical detection is of help in detecting a subgroup of breast carcinoma patients who are at high risk. This may also be of particular relevance in decisions regarding adjuvant chemotherapy to these patients. C1 [Aziz, Abdus Syed] The Aga Khan University, Department of Pathology and Microbiology, Stadium RoadKarachi, Pakistan. [Pervez, Shahid] The Aga Khan University, Department of Pathology and Microbiology, Stadium RoadKarachi, Pakistan. [Khan, Shaista] The Aga Khan University, Department of SurgeryKarachi, Pakistan. [Kayani, Naila] The Aga Khan University, Department of Pathology and Microbiology, Stadium RoadKarachi, Pakistan. [Azam, Igbal Syed] The Aga Khan University, Department of CHSKarachi, Pakistan. [Rahbar, Hussain Mohammed] The Aga Khan University, Department of CHSKarachi, Pakistan. RP Pervez, Sh (reprint author), The Aga Khan University, Department of Pathology and Microbiology, Karachi, Pakistan. EM shahid.pervez@aku.edu CR Guido S, Georg F, Joachim T: Fluorescence in Situ Hybridization for detecting C-ErbB-2 amplification in breast tumor fine needle aspiration biopsies. Acta Cytologica 40:164-173, 1996. Muss HB, Thor AD, Berry DA: Cerb-B-2 expression and response to adjuvant therapy in women with node-positive early breast cancer. N Engl J Med 330:1260-1266, 1994. William C: More than one way to look for HER-2. Cap Today 13:31-54, 1999. Perren TJ: C-ErbB-2 oncogene as a prognostic marker in breast cancer. Br J Cancer 63:328-332, 1991. van de Vijver MJ, Peterse JL, Mooi WJ: Neu-protein over expression in breast cancer. Association with comedo-type ductal carcinoma in situ and limited prognostic value in stage II breast cancer. N Engl J Med 319:1239-1245, 1988. Aryandono-T, Harijadi, Ghozali-A: Correlation of clinical, pathological status, hormone receptor and C-ErbB-2 oncoprotein in breast cancer patients. Gan-To-Kagaku-Ryoho Suppl 2:600-606, 2000. Looi-LM, Cheah-PL: C-ErbB-2 oncoprotein amplification in infiltrating ductal carcinoma of breast relates to high histological grade and loss of oestrogen receptor protein. Malays J Pathol. 20:19-23, 1998. Wright C, Angus B, Nicholson S:Expression of c-ErbB-2- oncoprotein: a prognostic indicator in human breast cancer. Cancer Res 49:2087-2090, 1989. Walker RA, Gullick WJ, Varley JM: An evaluation of immunoreactivity for C-ErbB-2 protein as a marker of poor short-term prognosis in breast cancer. Br J Cancer 60:426-429, 1989. Barnes DM, Lammie GA, Millis RR: An immunohistochemical evaluation of C-ErbB-2 gene expression in human breast carcinoma. Br J Cancer 58:448-452, 1988. Gusterson BA, Gelber RD, Goldhirsch A: Prognostic significance of C-ErbB-2 expression in breast cancer. J Clin Oncol 10:1046-1049, 1992. Tandon AK, Clark GM, Chamness GC: HER-2/neu oncogene protein and prognosis in breast cancer. J Clin Onco 1989; 17:1120-1128, 1989. Tsuda H, Hirohashi S, Shimasto Y: Correlation between longterm survival in breast cancer patients and amplification of two putative oncogene-coamplification units, hst-1 / int-2 and CErbB- 2 / ear-1. Cancer Res 49:3104-3110, 1989. Borg A, Tandon AK, Sigurdsson H: HER-2/ neu amplification predicts poor survival in node-negative breast cancer. Cancer Res 50:4332-4337, 1990. Slamon DJ, Clark GM, Wong SG: Human breast cancer: Correlation of relapse and survival with amplification of HER-2/neu oncogene. Science 235:177-183, 1987. Wright C, Angus B, Nicholson S:Expression of c-ErbB-2- oncoprotein: a prognostic indicator in human breast cancer. Cancer Res 49:2087-2090, 1989. Gullick WL, Wright C, Barnes D: C-ErbB-2 protein overexpression in breast carcinoma is a risk factor in patients with involved and uninvolved lymph nodes. Br J Cancer 63:434- 438, 1991. Cobleigh MA, Vogel CL, Tripathy D:Efficacy and safety of herceptin as a single agent in 222 women with HER-2 over expression who relapsed following chemotherapy for metastatic breast cancer. ASCO, Los Angeles California 1998; Abstract 376. Slamon D, Leyland-Jones B, Shak S: Addition of herceptin to first line chemotherapy for HER-2 over expression metastatic breast cancer markedly increases anticancer activity; A randomised multinational controlled phase III trial. ASCO, Los Angeles California 1998; Abstract 377. Jakic-Razumovic-J, Petrovecki-M, Uzarevic-B: Mutual predictive value of c-ErbB-2 over expression and various prognostic factors in ductal invasive breast carcinoma. Tumori 86:30-36, 2000. King CR, Kraus MH, Aaronson SA: Amplification of a v-erbB related gene in human mammary carcinoma. Science 229:974- 976, 1985. Berger MS, Locher GW, Saurer S: Correlation of C-ErbB-2 gene amplification and protein expression in human breast carcinoma with nodal status and nuclear grading. Cancer Res 48:1238- 1243, 1985. Slamon DJ, Godolphin W, Lones LA: Studies of HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 244:707-712, 1989. Ali IU, Campbell G, Lidereau R: Amplification of C-ErbB-2 and aggressive human breast tumors. Science 235:1795-1798, 1988. Bernard T, Jacques B: Prognostic significance of HER-2 /neu oncoprotein expression in node-positive breast cancer. Cancer 73:2359-2365, 1994. Sharma BK, Ray A, Kaur S: Immunohistochemical co-expression of c-ErbB-2/Neu oncoprotein, altered tumor suppressor, p53, protein, EGF-R and EMA in histological subtypes of infiltrating duct carcinoma of the breast. Indian J Exp Biol 37: 223-227, 1999. Shimizu C, Fukutomi T, Tsuda H: c-ErbB-2 protein over expression and p53 immunoreaction in primary and recurrent breast cancer tissues. J Surg Oncol 73:17-20, 2000. Baselga J, Tripathy D, Mendelsohn J: Phase II study of weekly intravenous trastuzumab, Herceptin, in patients with HER2/neu-overexpressing metastatic breast cancer. Semin Oncol 26, Suppl 12): 78-83, 1992. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2001 VL 7 IS 3 BP 190 EP 196 PG 7 ER PT J AU Popovic, M Hrcenjak, MT Babic, T Kos, J Grdica, M AF Popovic, Maja Hrcenjak, Mihaela Terezija Babic, Tomislav Kos, Josip Grdica, Mira TI Effect of Earthworm (G-90) Extract on Formation and Lysis of Clots Originated from Venous Blood of Dogs with Cardiopathies and with Malignant Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE homeostasis; cardiopathy; malignant tumors; earthworm extract (G-90) ID homeostasis; cardiopathy; malignant tumors; earthworm extract (G-90) AB The stability of homeostasis is important to keep a balance between coagulation and fibrinolysis. A disorder of homeostasis leads to different physiological changes and causes different diseases such as cardiopathies and malignant tumors. Cardiopathies is characterized by a hypercoagulation. In the malignant tumors, besides the hypercoagulation due to plasminogen activators (PA) formed inside the tumor, a disorder of homeostasis leads also to acceleration of the fibrinolysis. The variety of internal and external factors in both cases determine the deviation of time for the clots formation, as well as the lyses of blood and fibrin clots. In this study the venous blood as well as the blood and the fibrin clots, derived from healthy dogs, the dogs with cardiopathies and with malignant tumors, were examined for the time of coagulation and fibrinolysis by adding different substances. In these experiments we used a glycolipoprotein extract from earthworm tissue homogenate (G-90) and the proteolytic enzymes P I and P II, isolated from G-90. The efficacy of the tested substances was comparable with the clinically administered anticoagulants. The most significant differences in clotting time among the three tested groups of dogs were obtained by application of the original G-90. The results suggest a possibility that G-90, along with the fibrinolytic enzymes and other biologically active factors, also contains a factor that decelerates the formation of clot in a specific medium, such as the blood from the dogs with malignant tumors. C1 [Popovic, Maja] Faculty of Veterinary Medicine, Department of Biology, Heinzelova 55, 10000 Zagreb, Croatia. [Hrcenjak, Mihaela Terezija] Faculty of Veterinary Medicine, Department of Biology, Heinzelova 55, 10000 Zagreb, Croatia. [Babic, Tomislav] Faculty of Veterinary Medicine, Ruder Bockovic Institute,, Department of SurgeryZagreb, Croatia. [Kos, Josip] Faculty of Veterinary Medicine, Ruder Bockovic Institute,, Department of SurgeryZagreb, Croatia. [Grdica, Mira] Rudjer Boskovic Institute, Department of Molecular MedicineZagreb, Croatia. RP Popovic, M (reprint author), Faculty of Veterinary Medicine, Department of Biology, 10000 Zagreb, Croatia. EM mpopovic@vef.hr CR Brommer EJP, Emeis JJ, Verheijen JH, et al: Progress in Clinical Fibrinolysis. In: 7 Recent Advances in Blood Coagulation., Eds. Poller L and Ludlam CA), Churchill Livingstone, 1997, pp 161-182. Gandolfo GM, Conti L, Vercillo M: Fibrinolysis Components as Prognostic Markers in Breast Cancer and Colorectal Carcinoma. Anticancer Res 16:2155-2160, 1992. Glassman A: Hemostatic abnormalities associated with cancer and its therapy. Ann Clini Labo Sci 27:391-395, 1997. Dickneite G, Nicolay U, Friesen HJ, et al: Development of an anti-bleeding agent for recombinant hirudin induced skin bleeding in the pig. Thrombo Haemost 80:192-198, 1998. Du Z, Li X, Qian J, et al: Effect of earthworm fibrinolytic enzyme given orally on the activity of plasmin and on platelet aggregation in beagles dogs. Acta Acade Medi Shang 25:229- 231, 1998. Hrženjak T, Hrženjak M, Kašuba V, et al: A new source of biologically active compounds-earthworm tissue, Eisenia foetida, Lumbricus rubelus). Comp Biochem Physiol 102A:441-447, 1992. Hrženjak M, KobrehelÐ, Levanat S: Mitogenicity of the earthworm’s, Eisenia foetida, insulin-like proteins. Comp Biochem Physiol 104B:723-729,1993. Hrženjak T, PopoviaeM, BožiaeT, et al: Fibrinolytic and anticoagulative activities from the earthworm Eisenia foetida. Comp Biochem Physiol 119B:825-832, 1998. Hrženjak T, PopoviaeM, Tiška-Rudman LJ: Fibrinolytic activity of earthworms extract, G-90, on lysis of fibrin clots originated from the venous blood of patients with malignant tumors. Pat Oncol Res 3:206-211, 1998. Oberhoff C, Szymeczek J, Hoffmann O, et al: Adjuvant antiestrogen treatment with tamoxifen in postmenopausal women with breast cancer: A longitudinal study of blood coagulation and fibrinolysis. Breast Canc Res Treat 50:73-81, 1998. Okita Y, Takamoto S, Ando M, et al: Coagulation and fibrinolysis system in aortic surgery under deep hypothermic ciculatory arrest with aprotinin. The importance of adequate heparinization. Circulation 96:376-381, 1997. PopoviaeM, Grdiša M, VukoviaeS, et al: Adhesins of immunoglobulin- like superfamily from earthworm Eisenia foetida. Gen Pharmacol 30: 795-800, 1997. Sagripanti A, Capri A, Zacharski LR: The Pathophysiology of the haemostatic system in cancer patients: Insights gained from studies using plasmatic markers of haemostatic system activation. In: Cancer and Blood Coagulation: basic and clinical aspect., Eds. Sagripanti A, Carpi A and Zacharski LR), Ets Editrice, 1990, pp 69-92. Van Zutphen LFM, Baumans V, Beynen AC: Principles of Laboratory animal science., Eds. Van Zutphen LFM, Baumans V and Beynen A C), Elsevier Science Publishers, 1993. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2001 VL 7 IS 3 BP 197 EP 202 PG 6 ER PT J AU Arora, S Mathew, R Mathur, M Chattopadhayay, KT Ralhan, R AF Arora, Sonia Mathew, Robin Mathur, Meera Chattopadhayay, Kant Tushar Ralhan, Ranju TI Alterations in MDM2 Expression in Esophageal Squamous Cell Carcinoma: Relationship with p53 Status SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MDM2; p53; mutation; oncogene; esophageal squamous cell carcinoma ID MDM2; p53; mutation; oncogene; esophageal squamous cell carcinoma AB In view of the significance of MDM2 as a regulator as well as critical target of wild type p53, this study was undertaken to determine the alteration in MDM2 expression in esophageal squamons cell carcinoma (ESCC) and its relationship to clinicopathological parameters as well as p53gene and protein status. Immunohistochemical analysis of MDM2 and p53 proteins on paraffin embedded sections from 64 surgically resected ESCCs and matched histologically normal tissues showed overexpression of MDM2 protein in 23/64 (36%) ESCCs, while the histopathologically normal esophageal tissues did not show detectable level of MDM2 immunoreactivity. Interestingly, MDM2 – /p53 + phenotype was observed in 37/64 (58%) cases. None of the cases with p53 mis-sense mutations (12/30, 40%) showed detectable level of MDM2 protein. Missense p53 mutations were significantly associated with discordant p53 + /MDM2 – immunophenotype (p= 0.004). The most intriguing feature of the study was accumulation of MDM2 in the absence of detectable p53 in 11% of and overexpression of MDM2 and p53 in 25% of ESCCs, suggesting a p53-independent role for MDM2 in a subset of tumors. These results underscore the involvement of MDM2 in p53-dependent and -independent pathways in the pathogenesis of esophageal cancer in the Indian population. C1 [Arora, Sonia] All India Institute of Medical Sciences, Department of Biochemistry, 110029 Ansari Nagar, New Delhi, India. [Mathew, Robin] All India Institute of Medical Sciences, Department of Biochemistry, 110029 Ansari Nagar, New Delhi, India. [Mathur, Meera] All India Institute of Medical Sciences, Department of PathologyAnsari Nagar, New Delhi, India. [Chattopadhayay, Kant Tushar] All India Institute of Medical Sciences, Department of SurgeryAnsari Nagar, New Delhi, India. [Ralhan, Ranju] All India Institute of Medical Sciences, Department of Biochemistry, 110029 Ansari Nagar, New Delhi, India. RP Ralhan, R (reprint author), All India Institute of Medical Sciences, Department of Biochemistry, 110029 Ansari Nagar, New Delhi, India. EM ralhanr@hotmail.com CR Barak Y, Juven T, Hafner R, et al: MDM2 expression is induced by wild type p53 activity. EMBO J 12:461-468, 1993. Cahilly Snyder L, Yang-Feng T, Francke U, et al: Molecular analysis and Chromosomal mapping of amplified genes isolated from a transformed mouse 3T3 cell line. Somat Cell Molec Genet 13:235-244, 1987. Cordon-Cardo C, Latres E, Drobnjak M, et al: Molecular abnormalities of mdm-2 and p53 genes in adult soft tissue sarcomas. Canc Res 54:794-799, 1994. El-Deiry WS, Harper JW, O’Connor PM, et al: p21 is induced in p53-mediated G1 arrest and apoptosis. Canc Res 54:1169, 1994. Fakhar-Zadeh SS, Trusko SP, George DC: Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line. EMBO J 10:1565-1569, 1991. Foulkes WD, Stamp G WH, Afzal S, et al: MDM2 overexpression is rare in ovarian carcinoma irrespective of TP53 mutation status. Br J Cancer 72:883-888, 1995. Ganguli G, Abecassis J, Wasylyk B: MDM2 induces hyperplasia and premalignant lesions when expressed in the basal layer of the epidermis. EMBO J 19:5135-5147, 2000. Gaur D, Arora S, Mathur M, et al: High prevalence of p53 gene alterations and protein accumulation in human esophageal cancer: correlation with dietary risk factors in India. Clin Canc Res 3:2129-2136, 1997. Haines DS: The mdm2 proto-oncogene. Leukemia Lymphoma 26:227-238, 1997. Haital A, Wiener GH, Baethge U, et al: Mdm2 expression as a prognostic indicator in clear cell renal cell carcinoma: comparison with p53 overexpression and clinicopathological parameters. Clin Cancer Res 6:1840-1844, 2000. Itoshima T, Fujiwara T, Waku T, et al: Induction of apoptosis in human esophageal cancer cells by sequential transfer of the wild-type p53 E2F-1 Genes: Involvement of p53 accumulation via ARF-mediated MDM2 down-regulation. Clin Canc Res 6:2851-2859, 2000. Jones SN, Hancock AR, Vogel H, et al: Overexpression of Mdm2 in mice reveals a p53-independent role for Mdm2 in tumorigenesis. Proc Nat Acad Sci USA 95:15608-15612, 1998. Kern SE, Pietenpol JA, Thiagalingan S, et al: Oncogenic forms of p53 as a sequence-specific DNA-binding protein. Science 256:827, 1992. Kern SE, Kinzler KW, Bruskin A, et al: Identification of p53 as a sequence-specific DNA-binding protein. Science 252:1708, 1992. Landis SH, Murray T, Bolden S, et al: Cancer Statistics-1999, CA- J Clini. Ame Canc Soci 49:57-58, 1999. Leach FS, Tokino T, Meltzer P, et al: p53 Mutation and MDM2 amplification in human soft tissue sarcomas. Cancer Res 53:2231-2234, 1993. Marchett A, Buttitta F, Girlando S, et al: Mdm gene alterations and mdm2 protein expression in breast carcinomas. J Pathol 17:31-38, 1995 Michalides R:Prognosis for G1 cell–cycle regulators:Useful for predicting course of disease and for assessment of therapy in Cancer. J Pathol 188:341-343, 1999. Momand J, Wu HH, Dasgupta G: MDM2-master regulator of the p53 tumor suppressor protein. Gene 242:15-29, 2000. Oliner JD, Kinzler KW, Meltzer PS, et al: Amplification of a gene encoding a p53-associated protein in human sarcomas. Nature 358:80-83, 1992. Ralhan R, Agarwal S, Mathur M, et al: Induction of MDM2-P2 transcripts correlates with stabilized wild type p53 in Betel-tobacco Related Human Oral Cancer. Am J Pathol 157:587-596, 2000. Ralhan R, Arora S, Chattopadhyay TK, et al: Circulating p53 antibodies, p53 gene mutational profile and product accumulation in esophageal squamous-cell carcinoma in India. Int J Cancer, 85:791-795, 2000. Sandhya Agarwal, Mathur M, Shukla NK, et al: MDM2/p53 coexpression in oral premalignant and malignant lesions: potential prognostic implications. E J Canc Oral Oncol 35:209–216, 1999. Sherr CJ, Weber JD: The ARF/p53 pathway. Curr Opin Genet Dev 10, 94-99, 2000. Villuendas R, Pezzella F, Gatter K, et al:p21waf1/cip1 and MDM2 expression in non-Hodgkin’s lymphoma and their relationship to p53 status: A p53+, MDM2-, p21-immunophenotype associated with missense p53 mutations. J Pathol 181:51-61, 1997. Wu X, Bayle JH, Olson D, Leumi AJ: The p53-mdm2 autoregulatory feedback loop. Genes Dev 7:1126-1132, 1993. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2001 VL 7 IS 3 BP 203 EP 208 PG 6 ER PT J AU Karaburun Paker, S Kilicarslan, B Ciftcioglu, MA Oztekin, S Sargin, CF Erdogru, T Baykara, M AF Karaburun Paker, Semra Kilicarslan, Bahar Ciftcioglu, M Akif Oztekin, Sevim Sargin, C Figen Erdogru, Tibet Baykara, Mehmet TI Relationship Between Apoptosis Regulator Proteins (bcl-2 and p53) and Gleason Score in Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p53; bcl-2; prostate cancer; apoptosis ID p53; bcl-2; prostate cancer; apoptosis AB Cellular proliferation programmed cell death (apoptosis) are associated with tumor growth in general, and prostate cancer growth in particular. The aim of this study was to examine the expression of the apoptosis regulating genes bcl-2 and p53 and Gleason score in core needle biopsy specimens of prostate cancer using immunohistochemistry. We studied bcl-2 and p53 expression in 12 cases of low grade (Gleason score 2-5), 12 cases of intermediate grade (Gleason score 6-7) and 8 cases of high grade (Gleason score 8-10) prostate cancer. Overexpression of bcl-2 was noted in 3 of 32 patients (9.32%). One of them was high grade; others were intermediate grades. Expression of p53 was observed in 3 of low grades; others were high grade. The statistical analysis of present data suggest that there is no significant relation between p53 and bcl-2 expression and Gleason score in prostate cancer. C1 [Karaburun Paker, Semra] Akdeniz University, Faculty of Medicine, Department of PathologyAntalya, Turkey. [Kilicarslan, Bahar] Akdeniz University, Faculty of Medicine, Department of PathologyAntalya, Turkey. [Ciftcioglu, M Akif] Akdeniz University, Faculty of Medicine, Department of PathologyAntalya, Turkey. [Oztekin, Sevim] Akdeniz University, Faculty of Medicine, Department of PathologyAntalya, Turkey. [Sargin, C Figen] Akdeniz University, Faculty of Medicine, Department of PathologyAntalya, Turkey. [Erdogru, Tibet] Akdeniz University, Faculty of Medicine, Department of UrologyAntalya, Turkey. [Baykara, Mehmet] Akdeniz University, Faculty of Medicine, Department of UrologyAntalya, Turkey. RP Kilicarslan, B (reprint author), Akdeniz University, Faculty of Medicine, Department of Pathology, Antalya, Turkey. EM bkilicarslan@hotmail.com CR Bauer JJ, Sesterhenn IA, Mostofi FK, et al: Elevated levels of apoptosis regulator proteins p53 and bcl-2 are independent prognostic biomarkers in surgically treated clinically localized prostate cancer. J Urol 156:1511-1516, 1996. Boring CC, Squires T: Cancer statistics 1994. CA Cancer J Clin 44:7, 1994. Bubendorf L, Sauter G, Moch H, et al: Prognostic significance of bcl-2 in clinically localized prostate cancer. Am J Pathol 148:1557, 1996. Cheng L, Leibovich BC, Bergstralh EJ, et al. P53 Alteration in regional lymph node metastases from prostate carcinoma. Cancer 85:2455-2459, 1999. Cheng L, Sebo JT, Pisansky T, et al: P53 Protein Overexpression Is Associated with Increased Cell Proliferation in Patients with Locally Recurrent Prostate Carcinoma after Radiation Therapy. Cancer 85:1293-1299, 1999. Fox SB, Persad RA, Royds J, et al: p53 and c-myc expression in stage A1 prostatic adenocarcinoma: Useful prognostic determinants? J Urol 150:490-494, 1993. Grignon DJ, Hammond EH: Collage of American pathologists conferance XXVI on clinical relevance of prognostic markers in solid tumors: Report of prostate cancer working group. Arch Pathol Lab Med 119:1122-1126, 1995. Grossfeld GD, Olumi AF, Connolly JA, et al: Locally recurrent prostate tumors following either radiation therapy or radical prostatectomy have changes in Ki-67 labelling index, p53 and bcl-2 immunoreactivity. J Urology 159:1437-1443, 1998. Hockenbery D, Nunez G, Milliam C, et al: Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature 348:334, 1990. Issacs JT: Molecular markers for prostate cancer metastasis: Devoloping diagnostic methods for predicting the aggressiveness of prostate cancer. Am J Pathol 150:1511-1521, 1997. Kallakury BV, Figge J, Ross JS, et al: Association of p53 immunoreactivity with high Gleason tumor grade in prostatic adenocarcinoma. Human Pathol 25:92,1994. Levine AJ: P53, the cellular gatekeeper for growth and division. Cell 88:323, 1997. Masuda M, Takano Y, Iki M, et al: Prognostic Significance of Ki-67, p53 and bcl-2 expression in prostate cancer patients with lymph node metastases: A retrospective immunohistochemical analysis. Pathol Int:48:41-46,1998. Matsushima H, Kitamura T, Goto T, et al: Combined anaysis with bcl-2 and p53 immunostaining predicts poorer prognosis in prostatic carcinoma. J Urology 158:2278-2283, 1997. Prendergest NJ, Atkins MR, Schatte EC, et al: p53 immunohistochemical and genetic alterations are associated at high incidence with post-irradiated locally persistent prostate carcinoma. J Urol 155:1685-92,1996. Piris MA, Pezzella F, Martinez-Montero JC, et al: p53 and bcl- 2 expression in high grade B-cell lymphomas. Br J Cancer, 69:337, 1994. Shurbaji MS, Kalbfleisch JH, Thurmond S: Immunohistochemical detection of p53 protein as a prognostic indicator in prostate cancer. Human Path 26:106-109, 1995. Soini Y, Paakko P: Extent of Apoptosis in Relation to p53 and bcl-2 Expression in Germ Cell Tumors. Hum Pathol 27:1221- 1226, 1996. Von Eschenbach AC: The biologic dilemma of early carcinoma of prostate. Cancer 78:326-329, 1996. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2001 VL 7 IS 3 BP 209 EP 212 PG 4 ER PT J AU Resmiye, K Pesterel, EH Erdogan, G Gulkesen, HK Karavel, S AF Resmiye, Kaya Pesterel, Elif Hadice Erdogan, Gulgun Gulkesen, Hakan Kemal Karavel, Seyda TI Proliferating Activity in Differential Diagnosis of Benign Phyllodes Tumor and Cellular Fibroadenomas: Is It Helpful? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Phyllodes tumor; fibroadenoma; proliferating activity ID Phyllodes tumor; fibroadenoma; proliferating activity AB Benign phyllodes tumors and fibroadenomas are two types of fibroepithelial tumors of breast that are usually difficult to differentiate. The purpose of this study is to evalute the proliferative activity of these tumors and to find out if it helps in differential diagnosis. Thirty-one benign phyllodes tumors and twelve cellular fibroadenomas were retrieved from the archives of Pathology Department of Akdeniz University, School of Medicine. Proliferating activity of epithelial and stromal cells were evaluated by using labeling index (LI) of proliferating cell nuclear antigen (PCNA) and Ki-67 antigen by immunohistochemistry. The results were compared with other clinicopathologic findings. There was not any significant difference between the proliferating activity of phyllodes tumor and cellular fibroadenomas. Mean LI of PCNA was 28.01 (±22.85) in stromal cells and 56.57 (±30.98) in epithelial cells of phyllodes tumor where it was 28.92 (±24.02) and 62.53 (±32.56) in fibroadenomas. Ki-67 indices were 0.05 (±0.19) in stromal cells, 2.65 (±12.53) in epithelial cells of phyllodes tumors and 0.0 (±0) in stromal cells, 0.43 (±0.63) in epithelial cells of fibroadenomas. There was no correlation between the diameter of tumors and proliferating activity in both groups. Proliferating activity, determined by immunohistochemistry with PCNA and Ki-67 antibodies did not reveal significant difference between phyllodes tumor and fibroadenoma. C1 [Resmiye, Kaya] Akdeniz University, Faculty of Medicine, Department of Pathology, Akdeniz Universitesi Tip Fakultesi, Patoloji Anabilim Dali, ArapsuyuAntalya, Turkey. [Pesterel, Elif Hadice] Akdeniz University, Faculty of Medicine, Department of Pathology, Akdeniz Universitesi Tip Fakultesi, Patoloji Anabilim Dali, ArapsuyuAntalya, Turkey. [Erdogan, Gulgun] Akdeniz University, Faculty of Medicine, Department of Pathology, Akdeniz Universitesi Tip Fakultesi, Patoloji Anabilim Dali, ArapsuyuAntalya, Turkey. [Gulkesen, Hakan Kemal] Akdeniz University, Faculty of Medicine, Department of Pathology, Akdeniz Universitesi Tip Fakultesi, Patoloji Anabilim Dali, ArapsuyuAntalya, Turkey. [Karavel, Seyda] Akdeniz University, Faculty of Medicine, Department of Pathology, Akdeniz Universitesi Tip Fakultesi, Patoloji Anabilim Dali, ArapsuyuAntalya, Turkey. RP Pesterel, EH (reprint author), Akdeniz University, Faculty of Medicine, Department of Pathology, Antalya, Turkey. EM epestereli@superonline.com CR Tavassoli FA: Biphasic tumors. Pathology of The Breast 2nd ed. Appleton Lange, Stanford, Connecticut, 1999, pp. 571-631. Hanby AM, Millis RR, Oberman HA: The Breast. Diagnostic Surgical Pathology, Ed. Sternberg SS. Lippincott Williams Wilkins 3rd Ed. Philadelphia, USA, 1999 pp. 319-385. Hasebe T, Imoto S, Sasaki S, et al: Proliferative activity and tumor angiogenesis is closely correlated to stromal cellularity of fibroadenoma: Proposal fibroadenoma, cellular variant. Pathol Int 49:435-443, 1999. Keelan PA, Mayers JL, Wold LE, et al: Phyllodes tumor: Clinicopathological review of 60 patients and flow cytometric analysis in 30 patients. Hum Pathol 23:1048-1054, 1992. Kocova L, Skalova A, Fakan F, et al: Phyllodes tumor of the breast: Immunohistochemical study of 37 tumors using MIB1 antibody. Pathol Res Pract 94:97-104, 1998. Layfield LJ, Hart J, Neuwirth H, et al:. Relation between DNA ploidy and the clinical behavior of phyllodes tumors. Cancer 64:1486-1489, 1989. Nakano T, Oka K: Differential values of Ki-67 and mitotic index of proliferating cell population. Cancer 72:2401-2408, 1993. Noguchi S, Yokouchi H, Aihara T, et al: Progression of fibroadenoma to phyllodes tumor demonstrated by clonal analysis. Cancer 76:1779-1785, 1995. Perlich G, Tan CK, Kostura M, et al:Functional identity of proliferating cell nuclear antigen and a DNA polymerase delta auxiliary protein. Nature 326:517-520, 1987. Quinn CM, Wright NA: The clinical assessment of proliferation and growth in human tumors: Evaluation of methods and applications as prognostic variables. J Pathol 160:93-102, 1990. Salvadori B, Cusumano F, Bo RD, et al: Surgical treatment of phyllodes tumor of the breast. Cancer 63:2532-2536, 1989. Sawyer EJ, Hanby AM, Ellis P, et al: Molecular analysis of phyllodes tumors reveals distinct changes in the epithelial and stromal components. Am J Pathol 156:1093-1098, 2000. Umekita Y, Yoshida Y: Immunohistohcemical study of MIB1 expression in phyllodes tumor and fibroadenoma. Pathol Int 49:807-810, 1999. Umekita Y, Yoshida Y: Immunohistochemical study of hormoneregulated protein expression in phyllodes tumor: comparison with fibroadenoma. Virchows Arch 433:311-314, 1998. Witte F, Honig A, Mirecka J, et al. Cystosarcoma pyhllodes of the breast: Prognostic significance of proliferation and apopitosis associated genes. Anticancer Res 19:3355-3360, 1999. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2001 VL 7 IS 3 BP 213 EP 216 PG 4 ER PT J AU Timar, J Csuka, O Orosz, Zs Jeney, A Kopper, L AF Timar, Jozsef Csuka, Orsolya Orosz, Zsolt Jeney, Andras Kopper, Laszlo TI Molecular Pathology of Tumor Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE metastatic cascade; extracellular matrix interaction; metastatic phenotype; lung cancer; breast cancer; colon cancer; prostate cancer; malignant melanoma ID metastatic cascade; extracellular matrix interaction; metastatic phenotype; lung cancer; breast cancer; colon cancer; prostate cancer; malignant melanoma AB Millenium reviews of oncology agreed that the last century produced major developments mainly in the management of the primary tumor, but despite all of these results, cancer still remains among the leading causes of death due to the failure of clinical management of disseminated disease. This failure is primarily due to the lack of detailed information on the molecular mechanisms of tumor metastasis. Therefore, one of the hottest fields in experimental oncology is metastasis research, which provides more and more information about the molecular mechanisms. However, this information is fragmented and is not yet exploited in clinical practice. A new field of diagnostic pathology recently emerged, which translates basic research data to diagnostic practice to provide clinically relevant information on the biological potential (in this case metastatic potential) of the malignant tumors. Since tumor cell-extracellular matrix interactions are key features of tumor dissemination, expression of genes responsible for them can define the metastatic potential of malignant tumors. This review summarizes our recent knowledge on the metastatic geno- and phenotype of major human solid tumors: lung, colon, breast, prostate cancers and malignant melanoma. C1 [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gy. U. 7-9., H-1122 Budapest, Hungary. [Csuka, Orsolya] National Institute of Oncology, Department of Tumor Progression, Rath Gy. U. 7-9., H-1122 Budapest, Hungary. [Orosz, Zsolt] National Institute of Oncology, Department of Tumor Progression, Rath Gy. U. 7-9., H-1122 Budapest, Hungary. [Jeney, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. CR Adachi M, Taki T, Haung C, et al: Reduced integrin alpha3 expression as a factor of poor prognosis of patients with adenocarcinoma of the lung. J Clin Oncol 16:1060-1067, 1998. Balch ChM, Reintgen DS, Kirkwood JM, et al: Cutaneous melanoma. In:, DeVita, VT, Hellman S, Rosenberg SA, eds)): Principles & Practice of Oncology, Fifth Edition, Lippincott- Raven Publ. Philadelphia, Vol.1. 1947-94, 1997. Bankfalvi A, Terpe HJ, Breukelmann D et al: Gains and losses of CD44 expression during breast carcinogenesis and tumour progression. Histopathology 33:107-116, 1998. Barnes DM, Gillett CE: Cyclin D1 in breast cancer. Breast Cancer Res Treat 52:1-15, 1998. Bellahcene A, Albert V, Pollina L, et al: Ectopic expression of bone sialoprotein in human tyroid cancer. Thyroid 8:637-641, 1998. Bishop NJA: Molecular pathology of melanoma. Cancer Metast Rev 16:141-154, 1997. Caputi M, Greoger AM, Esposito V, et al: Prognostic role of cyclin D1 in lung cancer. Relationship to proliferating cell nuclear antigen. Am J Respir Cell Mol Biol. 20:746- 750,1999. Chana JS, Wilson GD, Cree IA et al: c-myc, p53, and Bcl-2 expression and clinical outcome in uveal melanoma. Br J Ophthalmol 83:110-114,1999. Clezardin P: Recent insights into the role of integrins in cancer metastasis. CMLS Cell. Mol Life Sci 53:5412-548,1998. Cochran A, Bailly Ch, Paul E, et al: Characteristics that relate to prognosis. In:, Cochran AJ, Bailly Ch, Paul E, Remotti F, Bhuta S, Eds.): Melanocytic tumors. A guide to diagnosis. Lippincott-Raven Publishers, Philadelphia, New York, pp. 261-286, 1997. Cohen AM, Minsky BR, Schilsky RI: Cancer of the colon. In:, DeVita VT, Hellman S, Rosenberg, SA, eds)): Principles Practice of Oncology, Fifth Edition, Lippincott-Raven Publ. Philadelphia, Vol.1. 1144-1162, 1997. Coppola D, Hyacinthe H, Fu L, et al: CD44V6 expression in human colorectal carcinoma. Hum Pathol 29:627-635, 1998. Cox LA, Chen G, Le EY: Tumor suppressor genes and their role in breast cancer. Breast Cancer Res Treat 32:19-38,1994. D’Amico TA, Masse M, Herndon JE et al: A biologic risk model for stage I lung cancer: immunohistochemical analysis of 408 patients with the use of ten molecular markers. J Thorac Cardiovasc Surg 117:736-743, 1999. De Wit PEJ, Van Muijen GNP, De Wall: Pathology of malignant melanoma, including new markers and techniques in diagnosis and prognosis. Curr Op Oncol 8:143-151, 1996. Dome B, Somlai B, Ladanyi A, et al. Expression of CD44v3 splice variant is associated with the visceral metastatic phenotype of human melanoma. Virchows Arch, in press) Dong JT, Rinker-Schaeffer CW, Ichikawa T, et al: Prostate cancer– biology of metastasis and its clinical implications. World J Urol 14:182-189, 1996. Duffy MJ: Proteases as prognostic markers in cancer. Clin Cancer Res 2:613-618, 1996. Ewing J: Neoplatic diseases. WB Saunders, Philadelphia, 1928, 1928. Fazekas K, Csuka O, Koves I, et al.: Experimental and clinicopathological studies on the function of the HGF receptor in human colon cancer metastasis. Clin Exp Metast 2001, in press) Fidler IJ: Molecular Biology of Cancer: Invasion and Metastasis. Cancer In:, DeVita VT, Hellman S, Rosenberg SA, eds)): Principles & Practice of Oncology, Fifth Edition, Lippincott- Raven Publ. Philadelphia, 1:135-152, 1997. Fidler IJ, Radinsky R: Search for genes that suppress cancer metastasis. J Natl Cancer Inst 88:1700-1703, 1996. Fleisch H: Osteolytic tumor bone disease In: Felisch H, ed): Bisphosphonates in bone disease. The Parthenon Publishing Group, New York, London, pp.86-106, 1997. Foekens JA, Dall P, Klijn JG, et al: Prognostic value of CD44 variant expression in primary breast cancer. Int J Cancer 84:209-215, 1999. Folkman J: How is blood vessel growth regulated in normal and neoplastic tissue? Cancer Res 46:467-477, 1986. Fredersdorf S, Burns J, Milne AM, et al: High level expression of p27(kip1, and cyclin D1 in some human breast cancer cells: inverse correlation between the expression of p27, kip1, and degree of malignancy in human breast and colorectal cancers. Proc Natl Acad Sci USA 93:6380-6385, 1997. Freije JM, MacDonald NJ, Steeg PS: Nm23 and tumour metastasis: basic and translational advances. Biochem Soc Symp 63:261-271, 1998. Gasparini G, Brooks PC, Biganzoli E, et al: Vascular integrin alpha(v)beta3: a new prognostic indicator in breast cancer. Clin Cancer Res 4:2625-2634, 1998. Ghoussoub RA, Dillon DA, D’Aquila T, et al: Expression of cmet is a strong independent prognostic factor in breast carcinoma. Cancer 82:1513-1520, 1998. Gillespie MT, Thoma RJ, Pu ZY, et al: Calcitonin receptors, bone sialoprotein and osteopontin are expresswed in primary breast cancer. Int J Cancer 73:812-815, 1997. Ginsberg RJ, Vokes, EE, Raben A: Non-Small Cell Lung Cancer. In:, DeVita, VT., Hellman, S., Rosenberg, SA, eds)): Principles & Practice of Oncology, Fifth Edition, Lippincott- Raven Publ. Philadelphia, Vol.1. pp. 852-911, 1997. Glinsky GV: Cell adhesion and metastasis: is the site specificity of cancer metastasis determined by leukocyte-endothelial cell recognition and adhesion. Clin Rev Oncol/Hematol 14:229-278, 1993. Glinsky GV: Apoptosis in metastatic cancer cells. Clin Rev Oncol/Hematol 24:175-186, 1997. Gohji K, Fujimoto N, Hara I, et al: Serum matrix metalloproteinase- 2 and its density in men with prostate cancer as a new predictor of disease extension. Int J Cancer 79:96-101, 1998. Goldmann T, Ribbert D, Suter L, et al: Tumor characteristics involved in the metastatic behaviour as an improvement in primary cutaneous melanoma prognostics. J Exp Clin Cancer Res 17:483-489, 1998. Gui GP, Puddefoot JP, Vinson GP, et al: Alterd cell-matrix contact: a prerequisite for breast cancer metastasis? Br J Cancer 75:623-633, 1997. Guilford P: E-cadherin downregulation in cancer: fuel on the fire: Mol Medicine Today 5:172-177, 1999. Harris Y, Marrow M, Norton L: Malignant tumor of the breast. In:, DeVita VT, Hellman S, Rosenberg SA, eds)): Principles Practice of Oncology, Fifth Edition, Lippincott-Raven Publ. Philadelphia, Vol.1. 1557-1616,1997. Hart IR: Perspective: tumour spread – the problems of latency. J Pathology 187:91-94, 1999. Hashimoto S, Koji T, Kohara N, et al: Frequency of apoptosis relates inversely to invasiveness and metastatic activity in human colorectal cancer. Virchows Arch 43:241-248, 1997. Hofmann M, Rudy W, Zoller M, et al: CD44 splice variants confer metastatic behaviour in rats: homologous sequences are expressed in human tumor cell lines. Cancer Res 51:5292- 5297, 1991. Honn KV, Aref A, Chen Q, et al: Prostate cancer old problems and new approaches. Pathology Oncology Research 2:191- 211, 1996a. Honn KV, Aref A, Chen YQ et al: Prostate Cancer. Old problems and new approaches. Part III. Prevention and Treatment. Pathol Oncol Res 2:276-292, 1996b. Houston SJ, Plunkett TA, Barnes, DM, et al: Overexpression of c-erbB2 is an independent marker of resistance to endocrine therapy in advanced breast cancer. Br J Cancer 79:1220-1226, 1999. Ishigami SI, Arii S, Furutani M, et al: Predictive value of vascular endothelial growth factor, VEGF, in metastasis and prognosis of human colorectal cancer. Br J Cancer 78:1379- 1384, 1998. Jalkanen S, Reichert RA, Gallatin WM, et al: Homing receptors and the control of lymphocyte migration. Immunol Rev 91:39-60, 1986. Jansen RL, Hupperets PS, Arends JW, et al: MIB-1 labelling index is an independent prognostic marker in primary breast cancer. Br J Cancer 78:460-465, 1998. Juliano RL, Varner JA: Adhesion molecules in cancer: the role of integrins. Curr Op Cell Biol 5:812-818, 1993. Kim YC, Park KO, Kern JA, et al: The interactive effect of Ras, HER2, P53, and Bcl-2 expression in predicting the survival of non-small cell lung cancer patients. Lung Cancer 22:181-190, 1998. Koeneman KS, Yeung F, Chung LW: Osteomimetic properties of prostate cancer cells: a hypothesis supporting the predilection of prostate cancer metastasis and growth in the bone environment. Prostate 39:246-261, 1999. Kumar S, Ghellal A, Li C, et al: Breast carcinoma: vascular density determined using CD105 antibody correlates with tumor prognosis. Cancer Res 59:856-861, 1999. Lee J-H, Miele ME, Hicks DJ, et al: KiSS-1, a novel human malignant melanoma metastasis-suppressor gene. J Natl Cancer Inst 88:1731-1737, 1996. Liapis H, Flath A, Kitazawa S: Integrin alphav beta3 expression by bone resident breast cancer metastases. Diagn Mol Pathol 5:127-135, 1996. Liotta LA: Tumor invasion and metastasis: role extracellular matrix. Cancer Res 46:1-14, 1986. Maguire TM, Shering SG, Duggan CM, et al: High levels of cathepsin B predict poor aoutcome in patients with breast cancer. Int J Biol Markers 13:139-144, 1998. Menard S, Tagliabue E, Colnaghi MI: The 67 kDa laminin receptor as a prognostic factor in human cancer. Breast Cancer Res Treat 52:137-145, 1998. Meyer T, Hart IR: Mechanisms of tumour metastasis. Eur J Cancer 34:214-221, 1998. Miyake H, Hara I, Yamanaka K, et al: Elevation of serum levels of urokinase-type plasminogen activator and its receptor is associated with disease progression and prognosis in patients with prostate cancer. Prostate 39:123-129, 1999. Miyake M, Adachi M, Huang C, et al: A novel molecular staging protocol for non-small cell lung cancer. Oncogene 18:2397-2404, 1999. Nakamori S, Kameyama M, Imaoka S, et al: Involvement of carbohydrate antigen sialyl Lewis(x, in colorectal cancer metastasis. Dis Colon Rectum 40:420-431, 1997. Nakamori S, Watanabe H, Kameyama M, et al: Expression of autocrine motility factor receptor in colorectal cancer as a predictor for disease recurrence. Cancer 74:1855-1862, 1994. Nicolson GL: Paracrine and autocrine growth mechanisms in tumor metastasis to specific sites with particular emphasis on brain and lung metastasis. Cancer Metast Rev 12:325-343, 1993. Nihei Z, Ichikawa W, Kojima K et al: The positive relationship between the expression of CD44 variant 6 and prognosis in colorectal cancer. Surg Today 26:760-761, 1996. Noordzij MA, van Steenbrugge GJ, Verkaik NS, et al: The prognostic value of CD44 isoforms in prostate cancer patients treated by radical prostatectomy. Clin Cancer Res 3:805-815, 1997. Oesterling J, Fuks ZVI, Lee CT, Scher HI: In:, DeVita VT, Hellman S, Rosenberg SA, eds)): Principles & Practice of Oncology, Fifth Edition, Lippincott-Raven Publ. Philadelphia, Vol. 1. 1222-1385, 1997. Ohta Y, Endo Y, Tanaka M, et al: Significance of vascular endothelial growth factor messenger RNA expression in primary lung cancer. Clin Cancer Res 2:1411-1416, 1996. Paget S. The distribution of secondary growths in cancer of the breast. Lancet 1:571-573, 1889. Pappot H, Francis D, Brunner N, et al: p53 protein in nonsmall cell lung cancer as quantitated by enzyme-linked immunosorbent assay: relation to prognosis. Clin Cancer Res 2:155-160, 1996. Parsons SL, Watson SA, Collins HM et al: Gelatinase, MMP-2 and -9, expression in gastrointestinal malignancy. Br J Cancer 78:1495-502, 1998. Pegram DM, Pauletti G, Slamon DJ: HER-2/neu as a predictive marker of response to breast cancer therapy. Breast Cancer Res Treat 52:65-77, 1998. Ponta H, Sleeman J, Herrlich P: Tumor metastasis formation: cell-surface proteins confer metastasis-promoting or -suppressisng properties. Biochem Biophys Acta 1198:1-10, 1994. Radinsky R: Paracrine growth regulation of human colon carcinoma organ-specifric metastasis. Cancer Metast Rev 12:345-361, 1993. Raso E, Tovari J, Toth K, et al. Ectopic aIIbb3 integrin signaling involves 12-lipoxygenase and PKC-mediated serine phosphorylation events in melanoma cells. Thromb Haemostas 85:1037-1042, 2001. Ravioli A, Bagli L, Zucchini A, et al: Prognosis and prediction of response in breast cancer: the current role of the main biological markers. Cell Prolif 31:113-126, 1998. Ropponen KM, Eskelinen MJ, Lipponen PK, et al: Expression of CD44 and variant proteins in human colorectal cancer and its relevance for prognosis. Scand J Gastroenterol 33:301-309, 1998. Rudolph P, Alm P, Heidebrecht HJ, et al: Immunologic proliferation marker Ki-S2 as prognostic indicator for lymph nodenegative breast cancer. J Natl Cancer Inst 91:271-278, 1999. Saito M, Yamaguchi A, Goi T, et al: Expression of DCC protein in colorectal tumors and its relationship to tumor progression and metastasis. Oncology 56:134-141, 1999. Saitoh K, Takasashi H, Sawada N: Detection of the c-met proto-oncogene product in normal skin and tumours of melanocytic origin. J Pathology 174:191-199, 1994. Satoh Y, Higashi T, Nouso K, et al: Cathepsin B in the growth of colorectal cancer: increased activity of cathepsin B in human colorectal cancer. Acta Med Okayama 50:305-311, 1996. Seiki M, Sato H, Liotta LA, et al: Comparison of autocrine mechanisms promoting motility in two metastatic cell lines: human melanoma and ras-transfected NH3T3 cells. Int J Cancer 49:717-720, 1991. Shiina H, Igawa M, Shigeno K, et al: Relationship of serum testosterone level with proliferating cell nuclear antigen and nm23 protein in human prostatic carcinoma tissue. Oncology 54:482-489, 1997. Shivers SC, Wang X, Li W, et al: Molecular staging of malignant melanoma: correlation with clinical outcome. JAMA 280:1410-1415,1998. Siegfried JM, Weissfeld LA, Luketich JD, et al: The clinical significance of hepatocyte growth factor for non-small cell lung cancer. Ann Thorac Surg 66:1915-1918, 1998. Silletti S, Paku S, Raz A: Tumor cell motility and metastasis. Autocrine motility factor as an example of ecto/exoenzyme cytokines. Pathology Oncology Research 3:230-254, 1997. Sloane BF, Moin K, Krepela E, et al: Cathepsin B and its endogenous inhibitors: the role in tumor malignancy. Cancer Metastasis Rev 9:333-352, 1990. Smythe WR, Wasf, D, Bavaria JE et al: Loss of alpha-v integrin expression and recurrence in node-negative lung carcinoma. Ann Thorac Surg 64:949-953, 1997. Steeg PS, Bevilacqua G, Kopper L, et al: Evidence for a novel gene associated with low tumor metastatic potential. J Natl Cancer Inst 80:200-204, 1988. Stephens RW, Nielsen HJ, Christensen IJ, et al: Plasma urokinase receptor levels in patients with colorectal cancer: relationship to prognosis. J Natl Cancer Inst 91:869-874, 1999. Sun XF, Ekberg H, Zhanh H, et al: Overexpression of ras is an independent prognostic factor in colorectal adenocarcinoma. APMIS 106:657-664, 1998. Takanami I, Tanaka F, Hashizume T, et al: Tumor angiogenesis in pulmonary adenocarcinomas: relationship with basic fibroblast growth factor, its receptor, and survival. Neoplasma 44:295-298, 1997. Tan P, Cody B, Wanner M, et al: The cell cycle inhibitor p27 is an independent prognostic marker in small, T1a,b, invasive breast carcinomas. Cancer Res 57:1259-1263, 1997. Tang DG, Honn KV: Adhesion molecules and tumor metastasis: an update. Invasion Metast 14:109-122, 1995. Tanigawa S, Hirokazu A, Matsumura M, et al: Tumor angiogenesis and mode of metastasis in Patients with colorectal cancer. Cancer Res 57:1043-1046, 1997. Telvensaari-Mattila A, Paakk, P, Hoyhtya M, et al: Matrix metalloproteinase- 2 immunoreactive protein: a marker of aggressiveness in breast carcinoma. Camcer 83:1153-1162, 1998. Timar J, Silletti S, Bazaz R, et al: Regulation of melanoma-cell motility by the lipoxygenase metabolite 12-(S)-HETE. Int J Cancer 55:1003-1010, 1993. Timar J, Toth Sz, Tovari J, et al. Autocrine motility factor, neuroleukin, phosphohexose isomerase, induces cell movement through 12-lipoxygenase-dependent tyrosine phosphorylation and serine dephosphorylation events. Clin Exp Metast 17:809- 816, 1999. Timar J, Raso E, Gilde K, et al. Expression and function of the AMR-AMFR system in human melanoma in experimental and clinical systems. Clin Exp Metast, in press). Tovari J, Paku S, Raso E, et al: Role of sinusoidal heparan sulfate proteoglycan in liver metastasis formation Int J Cancer 71:825-831, 1997. Trikha M, Timar J, Lundy SK, et al: Human prostate carcinoma cells express functional aIIbb3 integrin. Cancer Res 56:5071-5078, 1996. Trikha M, Cai Y, Grignon D, et al.: Identification of a novel truncated aIIb integrin. Canc Res 58:4771-4775, 1998. Vaisanen A, Tuominen H, Kallioinen M, Turpeenniemi-Hujanen T: Matrix metallo-proteinase-2, 72 kD type IV collagenase, expression occurs in the early stage of human melanocytic tumour progression and may have prognostic value. J Pathol 180:283-289, 1996. Volm M, Mattern J, Koomagi R: Association between nm23- H1 expression, proliferation and apoptosis in non-small cell lung carcinomas. Clin Exp Metastasis 16:595-602, 1998. Watanabe H, Takehana K, Date M, et al: Tumor cell autocrine motility factor is the neuroleukin / phosphohexose isomerase polypeptide. Cancer Res 46:2960-2963, 1996. Yamaguchi A, Ding K, Maehara M, et al: Expression of nm23- H1gene and Sialyl Lewis X antigen in breast cancer. Oncology 55:357-362, 1998. Yu H, Levesque MA, Clark GM, et al: Prognostic value of prostate-specific antigen for women with breast cancer: a large United States cohort study. Clin Cancer Res 4:1489-1497, 1997. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2001 VL 7 IS 3 BP 217 EP 230 PG 14 ER PT J AU Polgar, Cs Fodor, J Major, T Orosz, Zs Nemeth, Gy AF Polgar, Csaba Fodor, Janos Major, Tibor Orosz, Zsolt Nemeth, Gyorgy TI The Role of Boost Irradiation in the Conservative Treatment of Stage I-II Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE breast cancer; radiotherapy; boost; brachytherapy; electron; local control ID breast cancer; radiotherapy; boost; brachytherapy; electron; local control AB In this article, we review the current status, indication, technical aspects, controversies, and future prospects of boost irradiation after breast conserving surgery (BCS). BCS and radiotherapy (RT) of the conserved breast became widely accepted in the last decades for the treatment of early invasive breast cancer. The standard technique of RT after breast conservation is to treat the whole breast up to a total dose of 45 to 50 Gy. However, there is no consensus among radiation oncologists about the necessity of boost dose to the tumor bed. Generally accepted criteria for identification of high risk subgroups, in which boost is recommended, have not been established yet. Further controversy exists regarding the optimal boost technique (electron vs. brachytherapy), and their impact on local tumor control and cosmesis. Based on the results of numerous retrospective and recently published prospective trials, the European brachytherapy society (GEC-ESTRO), as well as the American Brachytherapy Society has issued their guidelines in these topics. These guidelines will help clinicians in their medical decisions. Some aspects of boost irradiation still remain somewhat controversial. The final results of prospective boost trials with longer follow-up, involving analyses based on pathologically defined subgroups, will clarify these controversies. Preliminary results with recently developed boost techniques (intraoperative RT, CT-image based 3D conformal brachytherapy, and 3D virtual brachytherapy) are promising. However, more experience and longer follow-up are required to define whether these methods might improve local tumor control for breast cancer patients treated with conservative surgery and RT. C1 [Polgar, Csaba] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Fodor, Janos] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Major, Tibor] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Orosz, Zsolt] National Institute of Oncology, Department of Diagnostic Pathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Nemeth, Gyorgy] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. RP Polgar, Cs (reprint author), National Institute of Oncology, Center of Radiotherapy, H-1122 Budapest, Hungary. EM polgar@oncol.hu CR Arriagada R, Le MG, Rochard F, et al: Conservative treatment versus mastectomy in early breast cancer: patterns of failure with 15 years of follow-up data. J Clin Oncol 14:1558-1564, 1996. Blichert-Toft M, Rose C, Andersen JA, et al: Danish randomized trial comparing breast conservation therapy with mastectomy: Six years of life-table analysis. J Natl Cancer Inst Monogr 11:19-25, 1992. Fisher B, Anderson S, Redmond CK, et al: Reanalysis and results after 12 years of follow-up in a randomized clinical trial comparing total mastectomy with lumpectomy with or without irradiation in the treatment of breast cancer. N Engl J Med 333:1456-1461, 1995. Lichter AS, Lippman ME, Danforth DN, et al: Mastectomy versus breast-conserving therapy in the treatment of stage I and II carcinoma of the breast: a randomized trial at the National Cancer Institute. J Clin Oncol 10:976-983, 1992. Van Dongen JA, Bartelink H, Fentiman IS, et al. Randomized clinical trial to assess the value of breast-conserving therapy in stage I and II breast cancer, EORTC 10801 Trial. J Natl Cancer Inst Monogr 11:15-18, 1992. Veronesi U, Salvadori B, Luini A, et al: Breast conservation is a safe method in patients with small cancer of the breast: Longterm results of three randomised trials of 1973 patients. Eur J Cancer 31A: 1574-1579, 1995. NIH Consensus Conference: Treatment of early-stage breast cancer. JAMA 265:391-395, 1991. Hammer J, Mazeron JJ, Van Limbergen E: Breast boost – why, how, when…? Strahlenther Onkol 175:478-483, 1999. Clark RM, Whelan T, Levine M, et al: Randomized clinical trial of breast irradiation following lumpectomy and axillary dissection for node-negative breast cancer: an update. J Natl Cancer Inst 88:1659-1664, 1996. Fisher B, Wickerham DL, Deutsch M, et al: Breast tumor recurrence following lumpectomy with and without breast irradiation: an overview of recent NSABP findings. Semin Surg Oncol 8:153-160, 1992. Liljegren G, Holmberg L, Adami HO, et al: Sector resection with or without postoperative radiotherapy for stage I breast cancer: five-year results of a randomized trial. J Natl Cancer Inst 86:717-722, 1994. Pezner RD, Lipsett JA, Desai K, et al: To boost or not to boost: decreasing radiation therapy in conservative breast cancer treatment when “inked” tumor resection margins are pathologically free of cancer. Int J Radiat Oncol Biol Phys 14:873-877, 1988. Collette L, Fourquet A, Horiot JC, et al: Impact of boost dose of 16 Gy on local control in patients with early breast cancer: the EORTC “Boost versus no boost” trial., Abstract, Radiother Oncol 56, Suppl. 1): 46, 2000. Polgar C, Orosz Z, Fodor J, et al: The effect of high-dose-rate brachytherapy and electron boost on local control and side effects after breast conserving surgery: First results of the randomized Budapest breast boost trial. Radiother Oncol 60(Suppl. 1): 10, 2001. Romestaing P, Lehingue Y, Carrie C, et al: Role of a 10-Gy boost in the conservative treatment of early breast cancer: results of a randomized clinical trial in Lyon, France. J Clin Oncol 15:963-968, 1997. Nagykalnai T, Nemeskeri Cs, Mayer A, et al: Effectivity of boost radiotherapy on the local recurrence rate following breast conserving surgery plus whole breast irradiation. Proceedings of the 2nd European Congress on Senology, 1994, pp. 591-596. Teissier E, Hery M, Ramaioli A, et al: Boost in conservative treatment: 6 year results of randomized trial. Breast Cancer Res Treat 50:345, 1998. Berberich W, Schnabel K, Berg D, et al: Boost irradiation of breast carcinoma: teletherapy vs. brachytherapy. Eur J Obstet Gynecol Reprod Biol 94:276-282, 2001. Clarke DH, Vicini F, Jacobs H, et al: High dose rate brachytherapy for breast cancer. In: High dose rate brachytherapy: a textbook., Ed: Nag S), Futura Publ Co, 1994, pp. 321-329. De la Rochefordiere A, Abner A, Silver B, et al: Are cosmetic results following conservative surgery and radiation therapy for early breast cancer dependent on technique? Int J Radiat Oncol Biol Phys 23:925-931, 1992. Frazier RC, Kestin LL, Kini V, et al: Impact of boost technique on outcome in early-stage breast cancer patients treated with breast-conserving therapy. Am J Clin Oncol 24:26-32, 2001. Jacobs H: HDR afterloading experience in breast conservation therapy. Selectron Brachy-therapy J 6:14-17, 1992. Mansfield CM, Komarnicky LT, Schwartz GF, et al: Ten-year results in 1070 patients with stages I and II breast cancer treated by conservative surgery and radiation therapy. Cancer 75:2328-2336, 1995. Nag S, Kuske RR, Vicini FA, et al: Brachytherapy in the treatment of breast cancer. Oncology, Huntingt, 15:195-202, 2001. Perez CA, Taylor ME, Halverson K, et al: Brachytherapy or electron beam boost in conservation therapy of carcinoma of the breast: a nonrandomized comparison. Int J Radiat Oncol Biol Phys 34:995-1007, 1996. Sarin R, Dinsaw KA, Shrivastava SK, et al: Therapeutic factors influencing outcome and late complications in the conservative management of early breast cancer. Int J Radiat Oncol Biol Phys 27:285-292, 1993. Touboul E, Belkacemi Y, Lefranc JP, et al: Early breast cancer: influence of type of boost, electrons vs iridium-192 implant, on local control and cosmesis after conservative surgery and radiation therapy. Radiother Oncol 34:105-113, 1995. Vicini FA, Horwitz EM, Lacerna MD, et al: Long-term outcome with interstitial brachy-therapy in the management of patients with early-stage breast cancer treated with breast-conserving therapy. Int J Radiat Oncol Biol Phys 37:845-852, 1997. Wazer DE, Kramer B, Schmid C, et al: Factors determining outcome in patients treated with interstitial implantation as a radiation boost for breast conservation therapy. Int J Radiat Oncol Biol Phys 39:381-393, 1997. Wazer DE, Schmidt-Ullrich RK, Ruthazer R, et al: Factors determining outcome for breast-conserving irradiation with margin-directed dose escalation to the tumor bed. Int J Radiat Oncol Biol Phys 40:851-858, 1998. Proceedings of the Consensus Meeting on Breast Cancer: To boost or not to boost and how to do it. GEC-ESTRO, 2001, pp. 1-114 Holland R, Veling SHJ, Mravunac M, et al: Histologic multifocality of Tis, T1-2 breast carcinomas: Implication for clinical trials of breast-conserving surgery. Cancer 56:979-990, 1985. Holland R, Conolly J, Gelman R: Nature and extent of residual cancer in the breast related to the intraductal component in the primary tumor. Int J Radiat Oncol Biol Phys 15:182-183, 1988. Liljegren G, Holmberg L, Bergh J, et al: 10-year results after sector resection with or without postoperative radiotherapy for stage I breast cancer: a randomized trial. J Clin Oncol 17:2326- 2333, 1999. Fisher ER, Anderson S, Redmond C, et al: Ipsilateral breast tumor recurrence and survival following lumpectomy and irradiation: pathological findings from NSABP Protocol B-06. Semin Surg Oncol 8:161-166, 1992. Schmidt-Ullrich RK, Wazer DE, DiPetrillo T, et al: Breast conservation therapy for early stage breast carcinoma with outstanding 10-year locoregional control rates: a case for aggressive therapy to the tumor bearing quadrant. Int J Radiat Oncol Biol Phys 27:545-552, 1993. Clarke DH, Le MG, Sarrazin D, et al: Analysis of local regional relapses in patients with early breast cancers treated by excision and radiotherapy. Experience of the Institute Gustave Roussy. Int J Radiat Oncol Biol Phys 11:137-145, 1985. Recht A, Silver B, Schnitt SJ, et al: Breast relapse following primary radiation therapy for early breast cancer I. Classification, frequency and salvage. Int J Radiat Oncol Biol Phys 11:1271- 1276, 1985. Van Limbergen E, Van den Bogaert W, Van der Schueren E, et al: Tumor excision and radiotherapy as primary treatment of breast cancer. Analysis of patient and treatment parameters and local control. Radiother Oncol 8:1-9, 1987. Van Limbergen E: The evidence for dose response relationship for local control rates after breast conserving surgery and radiotherapy. What is than the optimal dose when look at cosmetic outcome? Proceedings of the Consensus Meeting on Breast Cancer: To boost or not to boost and how to do it. GECESTRO, 2001, pp. 47-57. Salvadori B, Veronesi U: Conservative methods for breast cancer of small size: the experience of the National Cancer Institute, Milan, 1973-1998). Breast 8:311-314, 1999. Forrest P, Stewart H, Everington D, et al: Randomized controlled trial of conservation therapy for breast cancer: 6-year analysis of the Scottish trial. Lancet 348:708-713, 1996. Bartelink H: Higher radiation dose reduces recurrence rate in patients with early breast cancer: The EORTC ”Boost versus No Boost” Trial., Abstract, Proceedings of the Consensus Meeting on Breast Cancer: To boost or not to boost and how to do it. GEC-ESTRO, 2001, pp. 21. Fodor J, Major T, Polgar C, et al: The impact of radiotherapy on the incidence and time of occurrence of local recurrence in early-stage breast cancer after breast conserving therapy. Neoplasma 47:181-186, 2000. Borger J, Kemperman H, Hart A, et al: Risk factors in breastconservation therapy. J Clin Oncol 12:653-660, 1994. Elkhuizen PHM, Van de Vijver MJ, Hermans J, et al: Local recurrence after breast-conserving therapy for invasive breast cancer: high incidence in young patients and association with poor survival. Int J Radiat Oncol Biol Phys 40:859-867, 1998. Kollias J, Elston CW, Ellis IO: Early-onset breast cancer – histopathological and prognostic considerations. Br J Cancer 75:1318-1323, 1997. De la Rochefordiere A, Asselain B, Campana F, et al: Age as a prognostic factor in premenopausal breast carcinoma. Lancet 341:1039-1043, 1993. Smitt MC, Nowels KW, Zdeblick MJ, et al: The importance of the lumpectomy surgical margin status in long term results of breast conservation. Cancer 76:259-267, 1995. Spivack B, Khanna MM, Tafra L, et al: Margin status and local recurrence after breast-conserving surgery. Arch Surg 129:952- 957, 1994. Anscher MS, Jones P, Prosnitz LR, et al: Local failure and margin status in early-stage breast carcinoma treated with conservation surgery and radiation therapy. Ann Surg 218:22-28, 1993. DiBiase SJ, Komarnicky LT, Schwartz GF, et al: The number of positive margins influences the outcome of women treated with breast preservation for early stage breast carcinoma. Cancer, 82:2212-2220, 1998. Schnitt SJ, Abner A, Gelman R, et al: The relationship between microscopic margins of resection and the risk of local recurrence in patients with breast cancer treated with breast-conserving surgery and radiation therapy. Cancer 74:1746-1751, 1994. Fowble BL, Solin LJ, Schultz DJ, et al: Ten year results of conservative surgery and irradiation for stage I and II breast cancer. Int J Radiat Oncol Biol Phys 21:269-277, 1991. Eberlein TJ, Conolly JL, Schnitt SJ, et al: Predictors of local recurrence following conservative breast surgery and radiation therapy. Arch Surg 125:771-777, 1990. Krishnan L, Jewell WR, Krishnan EC, et al: Breast cancer with extensive intraductal component: treatment with immediate interstitial boost irradiation. Radiology, 183:273-276, 1992. Taghian AG, Powell SN: The role of radiation therapy for primary breast cancer. Surg Clin N Am 79:1091-1115, 1999. Perez CA, Taylor ME: Breast: Stage Tis, T1, and T2 tumors. In: Perez CA, Brady LW, eds. Principles and practice of radiation oncology, Lippincott-Raven Publishers, Philadelphia pp 1269- 1415, 1998. Fodor J, Major T, Polgar Cs, et al: Tumor bed relapse after breast conserving surgery: evaluation of radiation therapy. Hungarian Oncol 42:225-228, 1998., in Hungarian) Schnitt SJ, Conolly JL, Harris JR, et al: Pathologic predictors of early local recurrence in stage I and II breast cancer treated by primary radiation therapy. Cancer 53:1049-1057, 1984. Clemente CG, Boracchi P, Del Vecchio M, et al: Peritumoral lymphatic invasion in patients with node-negative mammary duct carcinoma. Cancer 69:1396-1403, 1992. Sastre-Garau X, Jouve M, Asselain B, et al: Infiltrating lobular carcinoma of the breast: clinicopathologic analysis of 975 cases with reference to data on conservative therapy and metastatic patterns. Cancer 77:113-120, 1996. Polgar C: Radiotherapy of the tumor bed after breast conserving surgery for stage I-II breast cancer: Analysis of efficacy of conventional and novel radiotherapy methods. PhD Theses, Budapest, 2001., in Hungarian) Gould EW, Robinson PG: The pathologist’s examination of the ”lumpectomy”: the pathologists’view of surgical margins. Semin Surg Oncol 8:129-135, 1992. EORTC Breast Cancer Cooperative Group: Manual for clinical research and treatment in breast cancer. 4th edition, 2000. Carter D: Margins of ”lumpectomy” for breast cancer. Human Pathol 17 :330-332, 1986. Elston CW, Ellis IO: Pathological prognostic factors in breast cancer I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19:403-410, 1991. Hammer J, Seewald DH, Track C, et al: Breast cancer: Primary treatment with external-beam radiation therapy and high-dose-rate iridium implantation. Radiology 193:573-577, 1994. Hammer J, Track C, Seewald DH, et al: 192-iridium HDR boost in breast cancer treatment - experience from 644 patients. Radiother Oncol 55, Suppl. 1): 32, 2000. Hennequin C, Durdux C, Espie M, et al: High-dose-rate brachytherapy for early breast cancer: an ambulatory technique. Int J Radiat Oncol Biol Phys 45:85-90, 1999. Manning MA, Arthur DW, Schmidt-Ullrich RK, et al: Interstitial high-dose-rate brachytherapy boost: the feasibility and cosmetic outcome of a fractionated outpatient delivery scheme. Int J Radiat Oncol Biol Phys 48:1301-1306, 2000. Van Limbergen E: What are the optimal boost methods in relation to boost target depth in the breast? Proceedings of the Consensus Meeting on Breast Cancer: To boost or not to boost and how to do it. GEC-ESTRO, 2001, pp. 105-114. DeBiose DA, Horwitz EM, Martinez AA, et al: The use of ultrasonography in the localization of the lumpectomy cavity for interstitial brachytherapy of the breast. Int J Radiat Oncol Biol Phys 38:755-759, 1997. Rabinovitch R, Finlayson C, Pan Z, et al: Radiographic evaluation of surgical clips is better than ultrasound for defining the lumpectomy cavity in breast boost treatment planning: a prospective clinical study. Int J Radiat Oncol Biol Phys 47:313- 317, 2000. Harrington KJ, Harrison M, Bayle P, et al: Surgical clips in planning the electron boost in breast cancer: a qualitative and quantitative evaluation. Int J Radiat Oncol Biol Phys 34:579- 584, 1996. Regine WF, Ayyangar KM, Komarnicky LT, et al: Computer-CT planning of the electron boost in definitive breast irradiation. Int J Radiat Oncol Biol Phys 20:121-125, 1991. Sedlmayer F, Rahim HBK, Kogelnik D, es mtsai: Quality assurance in breast cancer brachytherapy: geographic miss in the interstitial boost treatment of the tumor bed. Int J Radiat Oncol Biol Phys 34:1133-1139, 1996. Polgar C, Major T, Somogyi A, et al: CT-image based conformal brachytherapy of breast cancer: the significance of semi-3D and 3-D treatment planning. Strahlenther Onkol 176:118-124, 2000. Vicini FA, Jaffray DA, Horwitz EM, et al: Implementation of 3D-virtual brachytherapy in the management of breast cancer: a description of a new method of interstitial brachytherapy. Int J Radiat Oncol Biol Phys 40:629-635, 1998. Krishnan L, Jewell WR, Krishnan EC, et al: Breast cancer with extensive intraductal component: Treatment with immediate interstitial boost irradiation. Radiology 183:273-276, 1992. Polgar C, Sulyok Z, Major T, et al: Reexcision and perioperative brachytherapy in the treatment of local relapse after breast conservation: a possible alternative to salvage mastectomy. Hungarian Surgery 53:120-123, 2000., in Hungarian) Gatzemeier W, Orecchia R, Gatti G, et al: Intra-operative radiation therapy, IORT, in the treatment of breast cancer – a new therapeutic alternative in the conservative treatment of breast cancer? Its potential role and future perspectives. Experiences from the European Institute of Oncology, EIO), Milan. Strahlenther Onkol 177:330-337, 2001. Hammer J, Van Limbergen, Mazeron JJ: Statements of the GEC-ESTRO Consenus Meeting on Breast Cancer: ”To boost or not to boost and how to do it”. Radiother Oncol, in preparation). NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2001 VL 7 IS 4 BP 241 EP 250 PG 10 ER PT J AU Qian, X Rothman, LV Nicosia, FR Tuszynski, PG AF Qian, Xiaohua Rothman, L Vicki Nicosia, F Roberto Tuszynski, P George TI Expression of Thrombospondin-1 in Human Pancreatic Adenocarcinomas: Role in Matrix Metalloproteinase-9 Production SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE extracellular matrix; thrombospondin-1; matrix metalloproteinase; pancreatic cancer; tumor invasion; tissue inhibitor of metalloproteinase ID extracellular matrix; thrombospondin-1; matrix metalloproteinase; pancreatic cancer; tumor invasion; tissue inhibitor of metalloproteinase AB Human pancreatic adenocarcinoma, an aggressive malignant disease, shows a strong desmoplastic reaction characterized by a remarkable proliferation of interstitial connective tissues. Thrombospondin-1 (TSP-1), a 450 kDa platelet and matrix glycoprotein, has been implicated in tumor invasion, angiogenesis and metastasis. TSP-1 and MMP-9 expression in pancreatic adenocarcinoma and control pancreas tissues was measured by immunohistochemistry. TSP-1 expression in pancreatic carcinoma cell lines, fibroblasts, and endothelial cells was measured by a competitive TSP-1 enzyme linked immunosorbent assay (ELISA). The effect of TSP-1 on MMP-9 production in pancreatic carcinoma cell lines was measured by zymography and Western blot analysis. Eighty five per cent (23/27) of cases of pancreatic adenocarcinoma showed increased TSP-1 staining in the desmoplastic stroma adjacent to tumor cells. No specific positive staining for TSP-1 was observed in the normal pancreatic tissues and the inflammatory areas. TSP-1 localized in tumor stroma surrounding the tumor cells expressing MMP-9. Using TSP-1 competitive ELISA, the secretion of TSP-1 by different pancreatic cancer cell lines into culture medium varied from 11.45 ± 14.08 to 275.82 ± 45.56 ng/10 6 cells/24 hours. The amounts of TSP-1 detected in both culture media and cell extracts from fibroblasts or endothelial cells were at least 2-3 fold higher than those from pancreatic cancer cells. TSP-1 augmented the production of matrix metalloproteinase-9, a matrix degrading enzyme, in pancreatic cancer cells in vitro. Stromally-derived TSP-1 up-regulates the production of MMP-9 by pancreatic adenocarcinoma. These data are consistent with the conclusion that TSP-1-rich stroma is involved in regulating matrix remodeling in tumor invasion. C1 [Qian, Xiaohua] MCP Hahnemann University, Department of PathologyPhiladelphia, Pennsylvania, USA. [Rothman, L Vicki] MCP Hahnemann University, Department of Surgery, Broad and Vine Streets, 19102-1192 Philadelphia, Pennsylvania, USA. [Nicosia, F Roberto] MCP Hahnemann University, Department of PathologyPhiladelphia, Pennsylvania, USA. [Tuszynski, P George] MCP Hahnemann University, Department of Surgery, Broad and Vine Streets, 19102-1192 Philadelphia, Pennsylvania, USA. RP Tuszynski, PG (reprint author), MCP Hahnemann University, Department of Surgery, 19102-1192 Philadelphia, USA. EM george.tuszynski@drexel.edu CR Parker SL, Tong T, Bolden S, Wingo PA: Cancer Statistics, 1996. CA-A Cancer J Clin 65:5-27, 1996. Almoguera C, Shibata D, Forrester K: Most human carcinomas of the human exocrine pancreas contain mutant of c-K-ras genes. Cell 53:549-554, 1988. Brentnall TA, Chen R, Lee JG, et al: Microsatellite instability and K-ras mutations associated with pancreatic adenocarcinoma and pancreatitis. Cancer Res 55:4264-4267, 1995. Hohne MW, Halatsch M, Kahl GF, et al: Frequent loss of expression of the potential tumor suppressor gene DCC in ductal pancreatic adenocarcinoma. Cancer Res 52:2616-2619, 1992. Ruggeri BA, Huang LY, Berger D, et al: Molecular pathology of primary and metastatic ductal pancreatic lesions. Analyses of mutations and expression of the p53, mdm-2, and p21/WAF-1 genes in sporadic and familial lesions. Cancer 79:700-716, 1997. Hahn SA, Schutte M, Hoque ATMS, et al: DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 271:350-353, 1996. Hall PA, Lemoine NR: Models of Pancreatic Cancer. Can Surveys 16:135-155, 1993. Stetler-Stevenson WG, Aznavoorian S, Liotta LA: Tumor cell interactions with the extracellular matrix during invasion and metastasis. Annu Rev Cell Biol 9:541-573, 1993. Tuszynski GP, Nicosia RF: Localization of thrombospondin and its cysteine-serine-valine- threonine-cysteine-glycine-specific receptor in human breast carcinoma. Lab Invest 70:228-233, 1994. Bertin N, Clezardin P, Kubiak R, et al: Thrombospondin-1 and -2 messenger RNA expression in normal, benign, and neoplastic human breast tissues: correlation with prognostic factors, tumor angiogenesis, and fibroblastic desmoplasia. Cancer Res 57:596-399, 1997. Qian XH, Tuszynski GP: Expression of thrombospondin-1 in cancer: A role in tumor progression. Proc Soc Exp Biol Med 212:199-207, 1996. Roberts DD: Regulation of tumor growth and metastasis by thrombospondin-1; FASEB J 10:1183-1191, 1996. Wang TN, Qian XH, Granick MS, et al: Thrombospondin-1, TSP-1, promotes the invasive properties of human breast cancer. J Surg Res 63:39-43, 1996. Qian X, Wang TN, Rothman VL, et al: Thrombospondin-1 modulates angiogenesis in vitro by up-regulation of matrix metalloproteinase- 9 in endothelial cells. Exp Cell Res 235:403-412, 1997. Albo D, Rothman VL, Roberts DD, et al: Tumor cell thrombospondin- 1 regulates tumor cell adhesion and invasion through the urokinase plasminogen activator receptor. Br J Cancer 83:298-306, 2000. Ray JM, Stetler-Stevenson WG: The role of matrix metelloproteases and their inhibitors in tumor invasion, metastasis and angiogenesis. Europ Resp J 7:2062-2072, 1994. Murphy G, Crabbe T: Gelatinases A and B. Methods Enzymol 248:470-484, 1995. Okada Y, Gonoji Y, Naka K, et al: Matrix metalloproteinase 9, 92-kDa Gelatinase/type IV collagenase, from HT 1080 human fibrosarcoma cells; J Biol Chem 267:21712-21719, 1992. Hua J, Muschel RJ: Inhibition of matrix metalloproteinase 9 expression by a ribozyme blocks metastasis in a rat sarcoma model system. Cancer Res 56:5279-5284, 1996. Lengyel E, Gum R, Juarez J, et al: Induction of Mr 92,000 type IV collagenase expression in a squamous cell carcinoma cell line by fibroblasts. Cancer Res 55:963-967, 1995. Segain JP, Harb J, Gregoire M, et al: Induction of fibroblast gelatinase B expression by direct contact with cell lines derived from primary tumor but not from metastases. Cancer Res 56:5506-5512, 1996. Tuszynski GP, Switalska HI, Knudsen K: Methods of studying platelet-secreted proteins and the platelet cytoskeleton. Modern Methods Pharmacol 4:267-286, 1987. Tuszynski GP, Srivastava S, Switalska HI, et al: The interaction of human platelet thrombospondin with fibrinogen. J Biol Chem 260:12240-12245, 1985. Murphy-Ullrich JE, Schultz-Cherry S, Hook M; Transforming growth factor-b complexes with thrombospondin. Mol Biol Cell 3:181-188, 1992. Kinoshita T, Sato H, Takino T, et al: Processing of a precursor of 72-kilodalton type IV collagenase/gelatinase A by a recombinant membrane-type 1 matrix metalloproteinase. Cancer Res 56:2535-2538, 1996. Kleiner DE, Stetler-Stevenson WG: Quantitative zymography: Detection of Picogram quantities of gelatinases. Anal Biochem 218:325-329, 1994. Himelstein BP, Canete-Soler R, Bernhard EJ, et al: Metalloproteinases in tumor progression: the contribution of MMP-9. Invas Metast 14:246-258, 1994. Vischer P, Beeck H, Voss B: Synthesis, intracellular processing and secretion of thrombospondin in human endothelial cells. Eur J Biochem 153:435-443, 1985. Clezardin P: Expression of thrombospondin by cells in culture. In: Lahav J, ed. Thrombospondin. Boca Raton: CRC Press, 41- 61, 1993. Goldberg GI, Strongin A, Collier IE, et al: Interaction of 92- kDa type IV collagenase with the tissue inhibitor of metalloproteinases prevents dimerization, complex formation with interstitial collagenase, and activation of the proenzyme with stromelysin; J Biol Chem 267:4583-4591, 1992. Lyons-Giordano B, Brinker JM, Kefalides NA: Heparin increases mRNA levels of thrombospondin but not fibronectin in human vascular smooth muscle cells; Biochem Biophys Res Comm 162:1100-1104, 1989. Clezardin P, Frappart L, Clerget M, et al: Expression of thrombospondin, TSP1, and its receptors, CD36 and CD51, in normal, hyperplastic, and neoplastic human breast; Cancer Res 53:1421-1430, 1993. Friess H, Yamanaka Y, Buchler M, et al: Enhanced expession of transforming growth factor b isoforms in pancreatic cancer correlates with decreased survival. Gastroenterology 105:1846- 1856, 1993. Tikhonenko AT, Black DJ, Linial ML: Viral Myc oncoproteins in infected fibroblasts down-modulate thrombospondin-1, a possible tumor suppressor gene; J Biol Chem 271:30741-30747, 1996. John AS, Tuszynski GP: The Role of Matrix Metalloproteinases in Tumor Angiogenesis and Tumor Metastasis. Pathol Oncol Res 7:14-23, 2001. Murphy G, Willenbrock F: Tissue inhibitors of matrix metalloendopeptidases. Methods Enzymol 248:496-510, 1995. Gress TM, Muller-Pillasch F, Lerch MM, et al: Expression and in-situ localization of genes coding for extracellular matrix proteins and extracellular matrix degrading proteases in pancreatic cancer. Int J Cancer 62:407-413, 1995. Hirata M, Itoh M, Tsuchida A, et al: Cholecystokinin receptor antagonist, loxiglumide, inhibits invasiveness of human pancreatic cancer cell lines. FEBS Lett 383:241-244, 1996. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2001 VL 7 IS 4 BP 251 EP 259 PG 9 ER PT J AU Ladanyi, A Gallai, M Paku, S Nagy, OJ Dudas, J Timar, J Kovalszky, I AF Ladanyi, Andrea Gallai, Monika Paku, Sandor Nagy, O Julianna Dudas, Jozsef Timar, Jozsef Kovalszky, Ilona TI Expression of a Decorin-Like Moleculein Human Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE decorin; human melanoma; mRNA; protein; TGF-beta ID decorin; human melanoma; mRNA; protein; TGF-beta AB Decorin, a member of the family of small leucin-rich proteoglycans, has originally been described as a secreted proteoglycan component of the connective tissues, and has been implicated in the negative regulation of cell proliferation directly or via interactions with TGF-b. It was reported to be generally absent from tumor cells. Here we show that human melanoma cell lines express a decorin-like molecule. We detected decorin mRNA by RT-PCR in 7 out 7 human melanoma lines characterized by various metastatic potential. Using polyclonal antiserum against the core protein of decorin, the typical 80-120 kD glycanated form as well as a high molecular weight aberrant version (200-210 kD) of decorin were demonstrated by Western blot technique in the culture supernatants as well as in lysates of human melanoma cells. Finally, decorin epitope was also demonstrated immunohistochemically in human melanoma xenografts, as well as in tumor cells of surgically resected melanomas but not in melanocytes of nevi.The expression of this aberrant decorin did not inhibit the in vitroor in vivogrowth of human melanoma cells, and it was independent of their metastatic potential. Human melanoma cell lines expressing aberrant decorin retained sensitivity to the antiproliferative and gelatinase-stimulatory effects of exogenous TGF-b b. C1 [Ladanyi, Andrea] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Gallai, Monika] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Paku, Sandor] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular PathologyBudapest, Hungary. [Nagy, O Julianna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Dudas, Jozsef] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. CR Krusius T, Ruoslahti E: Primary structure of an extracellular matrix proteoglycan core protein deduced from cloned cDNA. Proc Natl Acad Sci USA 83:7683-7687, 1986. Iozzo RV: The family of the small leucine-rich proteoglycans: key regulators of matrix assembly and cellular growth. Crit Rev Biochem Mol Biol 32:141-174, 1997. Yamaguchi Y, Mann DM, Ruoslahti E: Negative regulation of transforming growth factor-b by the proteoglycan decorin. Nature 346:281-284, 1990. Border WA, Noble NA, Yamamoto T, et al: Natural inhibitor of transforming growth factor-bprotects against scarring in experimental kidney disease. Nature 360361-364, 1992. Takeuchi Y, Kodama Y, Matsumoto T: Bone matrix decorin binds transforming growth factor-b and enhances its bioactivity. J Biol Chem 269:32634-32638, 1994. Hausser H, Groning A, Hasilik A. et al.: Selective inactivity of TGF-b/decorin complexes. FEBS Lett 353:243-245, 1994. Coppock DL, Kopman C, Scandalis S, et al: Preferential gene expression in quiescent human lung fibroblasts. Cell Growth Differ 4:483-493, 1993. Mauviel A, Santra M, Chen YQ. et al: Transcriptional regulation of decorin gene expression. Induction by quiescence and repression by tumor necrosis factor-a. J Biol Chem 270:11692- 11700, 1995. Santra M, Mann DM, Mercer EW, et al: Ectopic expression of decorin protein core causes a generalized growth suppression in neoplastic cells of various histogenetic origin and requires endogenous p21, an inhibitor of cyclin-dependent kinases. J Clin Invest 100:149-157, 1997. Yamaguchi Y, Ruoslahti E:Expression of human proteoglycan in Chinese hamster ovary cells inhibits cell proliferation. Nature 336:244-246, 1988. De Luca A, Santra M, Baldi A, et al: Decorin-induced growth suppression is associated with up-regulation of p21, an inhibitor of cyclin-dependent kinases. J Biol Chem 271:18961- 18965, 1996. Adany R, Heimer R, Caterson B, et al.: Altered expression of chondroitin sulfate proteoglycan in the stroma of human colon carcinoma. Hypomethylation of PG-40 gene correlates with increased PG-40 content and mRNA levels. J Biol Chem 265:11389-11396, 1990. Pluschke G, Vanek M, Evans A, et al: Molecular cloning of a human melanoma-associated chondroitin sulfate proteoglycan. Proc Natl Acad Sci USA 93:9710-9715, 1996. Iida J, Meijne AML, Knutson JR, et al: Cell surface chondroitin sulfate proteoglycans in tumor cell adhesion, motility and invasion. Semin Cancer Biol 7:155-162, 1996. Timar J, Ladanyi A, Lapis K, et al: Differential expression of proteoglycans on the surface of human melanoma cells characterized by altered experimental metastatic potential. Am J Pathol 141:467-474, 1992. Caux F, Timar J, Lapis K, Moczar M: Proteochondroitin sulphate in human melanoma cell cultures. Biochem Soc Transact 18:293-294, 1990. Timar J, Jeney A, Kovalszky I, Kopper L: Role of proteoglycans in tumor progression. Pathol Oncol Res 1:85-93, 1995. Ladanyi A, Timar J, Paku S, et al: Selection and characterization of human melanoma lines with different liver-colonizing capacity. Int J Cancer 46:456-461, 1990. Ladanyi A, Timar J, Bocsi J. et al: Sex-dependent liver metastasis of human melanoma lines in SCID mice. Melanoma Res 5:83-86, 1995. Ladanyi A, O. Nagy J, Jeney A, et al: Cytokine sensitivity of metastatic human melanoma cell lines – simultaneous inhibition of proliferation and enhancement of gelatinase activity. Pathol Oncol Res 4:108-114, 1998. Vogel KG, Ordog A, Pogany G, et al: Proteoglycans in the compressed region of human tibialis posterior tendon and in ligaments. J Orthop Res 11:68-77, 1984. Mueller BM, Romerdahl CA, Trent JM, et al: Suppression of spontaneous melanoma metastasis in scid mice with an antibody to the epidermal growth factor receptor. Cancer Res 51:2193-2198, 1991. Bani MR, Rak J, Adachi D, et al: Multiple features of advanced melanoma recapitulated in tumorigenic variants of early stage, radial growth phase, human melanoma cell lines: evidence for a dominant phenotype. Cancer Res 56:3075-3086, 1996. Timar J, Kovalszky I, Paku S, et al.: Two human melanoma xenografts with different metastatic capacity and glycosaminoglycan pattern. J Cancer Res Clin Oncol 115:554-557, 1989. Voss B, Glossl J, Cully Z, Kresse H: Immunocytochemical investigation on the distribution of small chondroitin sulfatedermatan sulfate proteoglycan in the human. J Histochem Cytochem 34:1013-1019, 1986. Imai K, Hiramatsu A, Fukushima D, et al: Degradation of decorin by matrix metalloproteinases: identification of the cleavage sites, kinetic analyses and transforming growth factor- 1 release. Biochem J 322:809-814, 1997. Danielson KG, Fazzio A, Cohen I, et al: The human decorin gene: intron-exon organization, discovery of two alternatively spliced exons in the 5’ untranslated region, and mapping of the gene to chromosome 12q23. Genomics 15:146-160, 1993. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2001 VL 7 IS 4 BP 260 EP 266 PG 7 ER PT J AU Ralte, MA Sharma, ChM Karak, KA Mehta, SV Sarkar, Ch AF Ralte, Mercy Angela Sharma, Chand Mehar Karak, Kumar Asis Mehta, Singh Veer Sarkar, Chitra TI Clinicopathological Features, MIB-1 Labeling Index and Apoptotic Index in Recurrent Astrocytic Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE astrocytic tumors; recurrent; MIB-1 labeling index; apoptotic index; interval to recurrence; progression ID astrocytic tumors; recurrent; MIB-1 labeling index; apoptotic index; interval to recurrence; progression AB This is a study of 64 cases of recurrent astrocytic tumors of all four WHO grades wherein a comparative evaluation of initial vs. recurrent tumor was done with respect to histological grading, MIB-1 labeling index (LI) and apoptotic index (AI). The aim was to identify factor/s that could influence interval to recurrence and/or malignant progression. Recurrence was noted in all grades and upon recurrence, 93.3% of grade II (low grade diffuse) astrocytomas and 63.6% of grade III anaplastic astrocytomas underwent malignant progression. However, none of the Grade I tumors showed evidence of malignant progression. Though interval to recurrence varied considerably, there was a correlation with histological grade of the initial tumor in that grade I and II tumors had a significantly longer mean interval to recurrence (43 months and 54.8 months respectively) as compared to grade III and IV (glioblastoma multiforme) tumors (17.6 and 12.8 months respectively). The interval to recurrence was also longer for grade II and III tumors which showed progression on recurrence (55.3 months for Grade II->Grade III; 54 months for Grade II->Grade IV and 20.6 months for Grade III->IV) as compared to tumors which recurred to the same grade (12.5 months for Grade III->Grade III and 12.8 months for Grade IV->Grade IV). A statistically significant inverse correlation of MIB-1 LI with interval to recurrence was noted. Higher the MIB-1 LI, shorter was the interval to recurrence. Further a cut off MIB-1 LI value of 2.8% could be proposed in predicting recurrence free survival. Interestingly, MIB-1 LI of grade II tumors, which had progressed to grade IV was significantly higher than MIB-1 LI of grade II tumors which had progressed to grade III. Thus, this study establishes the potential role of MIB-1 LI of the initial tumor in determining interval to recurrence. However, apoptotic index has no role in predicting either interval to recurrence or malignant progression. C1 [Ralte, Mercy Angela] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Sharma, Chand Mehar] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Karak, Kumar Asis] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Mehta, Singh Veer] All India Institute of Medical Sciences, Department of NeurosurgeryNew Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. RP Sarkar, Ch (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM sarkarcs@hotmail.com CR Afra D, Muller W: Recurrent low grade gliomas. Dedifferentiation and prospects of re-operation. In: Glioma. [Edt. Karim and Laws]. Springer Verlag, Berlin, Heidelberg. pp 189-203, 1991. Afra D, Muller W, Benoist G, et al: Supratentorial recurrences of gliomas : Results of re-operation on astrocytomas and oligodendrogliomas. Acta Neurochir, Wien, 43:217-227, 1978. Carrol RS, Zhang J, Chauncey BW, et al: Apoptosis in astrocytic neoplasms. Acta Neurochir, Wien, 139:845-850, 1997. Ellison DW, Steart PV, Bateman AC, et al: Prognostic indicators in a range of astrocytic tumors: An immunohistochemical study with Ki-67 and p53 antibodies. J Neurol Neuro Surg Psychiatry 59:413-419, 1995. Ellison DW, Steart PV, Gatter KC, et al: Apoptosis in cerebral astrocytic tumor and its relationship to expression of the Bcl-2 and p53 proteins. Neuropathol 21:352-361, 1995. Enestrom S, Vavruch L, Franlund B, et al:Ki-67 antigen expression as a prognostic factor in primary and recurrent astrocytomas. Neurochir 44:25-30, 1998. Heesters MA, Koudstaal J, Go KG, et al: Analysis of proliferation and apoptosis in brain gliomas : prognostic and clinical value. J Neurooncol 44:255-266, 1999. Hilton DA, Love S, Barber R, et al: Accumulation of p53 and Ki67 expression do not predict survival in patients with fibrillary astrocytomas or the response of these tumors to radiotherapy. Neurosurgery 42:724-729, 1998. Hoshi M, Yoshida K, Shimazaki K: Correlation between MIB-1 staining indices and recurrence in low grade astrocytomas. Brain Tumor Pathol.14:47-51, 1997. Hoshino T, Ahn D, Prados MD, et al: Prognostic significance of the proliferative potential of intracranial gliomas measured by Bromodeoxyuridine labeling. Int J Cancer 53:550-555, 1993. Hoshino T, Rodriguez LA, Cho KG, et al: Prognostic implications of the proliferative potential of low grade astrocytomas. J Neuro Surg 69:839-842, 1988. Ito S, Chandler KL, Prados MD, et al: Proliferative potential and prognostic evaluation of low grade astrocytomas. J Neuro Oncol 19:1-9, 1994. Iuzzolino PO, Ghimenton C, Nicolato A, et al: p53 in low grade astrocytomas : a study with long-term follow-up. Br J Cancer 69:586-591, 1994. Jaros E, Perry RH, Adam L, et al:Prognostic implication of p53 protein, EGFR and Ki67 labeling in brain tumors. Br J Cancer 66:373-380, 1992. Kaluza J, Adamek D, Pyrich M: Ki-67 as a marker of proliferation activity in tumor progression of recurrent gliomas of supratentorial localization. Immunocytochemical quantitative studies. Pol J Pathol 48:31-36, 1997. Cavenee WK, Bigner DD, Newcomb EW, et al: Diffuse astrocytomas. In : Pathology and genetics tumors of the nervous system., Eds. Kleihues P and Cavenee WK). International Agency for Research on Cancer, Lyon, 1997, pp 2-9. Krans JA, Bolln C, Wolf HK, et al: TP53 alterations and clinical outcome in low grade astrocytoma. Genes Chromosomes 10:143-149, 1994. Laws ER Jr, Taylor WG, Clinjton MB, et al: Neurosurgical management of low grade astrocytoma of the cerebral hemispheres. J Neurosug 61:665-73, 1984. Litofsky NS, Mix TC, Baker SP, et al: Ki-67 proliferation index in recurrent glial neoplasms : no prognostic significance. Surg Neurol 50:579-585, 1998. McKeever PE, Strawderman MS, Yamini B, et al: MIB-1 proliferation index predicts survival among patients with grade II astrocytoma. J Neuropathol Exp Neurol 57:931-936, 1998. Montine TJ, Vandersteenhoven JJ, Aguzzi A, et al: Prognostic significance of Ki 67 proliferation index in supratentorial fibrillary astrocytic neoplasms. Neurosurgery 34:674-678, 1994. Muller W, Afra D, Schroeder R: Supratentorial recurrences of gliomas. Morphological studies in relation to time intervals with astrocytomas. Acta Neurochir 37:75-91, 1977. Mustonen M, Rainio H, Paakko P, et al: The extent of apoptosis is inversely associated with bcl-2 expression in pre-malignant and malignant breast lesions. Histopathology 31:347-354, 1997. Nakamura M, Konishi N, Tsunoda S, et al: Retinoblastoma protein expression and MIB-1 correlate with survival of patients with malignant astrocytomas. Cancer 80:242-249, 1997. Rasheed BKA, McLendon RK, Herndon JD, et al: Alterations of the TP53 gene in human gliomas. Cancer Res 54:1324-1330, 1994. Recht LD, Law R, Smith TW: Suspected low grade glioima : Is deferring treatment safe? Ann Neurol 31:431-436, 1992. Rieger J, Ohgaki H, Kleihues P, et al: Human astrocytic brain tumors express Apo2L/Trail. Acta Neuropathol, Berl, 97:1-4, 1999. Reifenberger J, Ring GU, Gies U, et al: Analysis of p53 mutation and epidermal growth factor receptor amplification in recurrent gliomas with malignant progression. J Neuropathol Exp Neurol 55:822-831, 1996. Schiffer D, Chio A, Giordano MT, et al: Prognostic value of histologic features in adult cerebral astrocytomas. Cancer 61:1386-1393, 1988. Soffietti R, Chio A, Giordano MT, et al: Prognostic factors in well differentiated cerebral astrocytomas in the adult. Neurosurgery 24:686-692, 1989. Tandon PN, Mahapatra AK, Sarkar C, et al: Recurrence of supratentorial gliomas. Neurol India 45:141-149, 1997. Vertosik FT, Selker RG, Arena VC: Survival of patients with well differentiated astrocytomas diagnosed in the era of computed tomography. Neurosurgery 28:496-501, 1991. von Deimling A, Eibl RH, Ohgaki H, et al: p53 mutations are associated with 17p allelic loss in grade II and grade III astrocytomas. Cancer Res 52:2987-2990, 1992. von Deimling A, Louis DN, Menon AG, et al: Allelicloss on the long arm of chromosome 17 in pilocytic astrocytoma. Acta Neuropathol 76:81, 1993. Watanabe K, Peraud A, Gratas C, et al: p53 and PTEN mutations in gennistocytic astrocytomas. Acta Neuropathol, Berl, 95:559-560, 1998. Watanabe K, Tachibana O, Yonekawa, et al: Role of gemistocytes in astrocytoma progression. Lab Invest 76:277-284, 1997. Winger MJ, Macdonald DR, Cairncross JG: Supratentorial anaplastic gliomas in adults. The prognostic important of extent of resection and prior low grade glioma. J Neurosurg 71:787- 793, 1989. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2001 VL 7 IS 4 BP 267 EP 278 PG 12 ER PT J AU Kaya, H Ragazzini, T Aribal, E Guney, I KOTILOGLU, E AF Kaya, Handan Ragazzini, Teresa Aribal, Erkin Guney, Ilter KOTILOGLU, Esin TI Her-2/neuGene Amplification Compared with HER-2/neu Protein Overexpression on Ultrasound Guided Core-Needle Biopsy Specimens of Breast Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast carcinoma; FISH; immunohistochemistry; Her-2/neu ID breast carcinoma; FISH; immunohistochemistry; Her-2/neu AB Genomic amplification and oncoprotein overexpression of Her-2/neuwas studied on ultrasound core needle biopsy specimens of the infiltrative ductal carcinomas of the breast. We performed '' two colour '' fluorescence in situ hybridization (FISH) for Her-2/neuand chromosome 17 and compared the FISH results with the immunohistochemical overexpression of Her-2/neuprotein by 2 antibodies (DAKO HercepTest and the BioGenex monoclonal antibody AM 134-5M). Furthermore, following radical mastectomy with axillary dissection, Her-2/neustatus of the patients were compared with the well known histopathological prognostic factors such as histologic grade, tumor stage, lympho/ vascular invasion, surgical margin status and Paget’s disease. Amplification was demonstrated 27% of the cases. Her-2/neu protein overexpression was detected in 47% and 80% of the cases with CB11 and HercepTest respectively. We revealed statistically significant association between the tumor, oncoprotein expression and oncogene amplification (p<0.05). The results of our study showed that combination of IHC and FISH methods enhances the evaluation of tumor genetics at both gene and protein level for the analysis of Her-2/neuin breast carcinoma. C1 [Kaya, Handan] Marmara University, School of Medicine, Department of Pathology, Altunizade, 81190 Istanbul, Turkey. [Ragazzini, Teresa] University of Bologna, Department of OncologyBologna, Italy. [Aribal, Erkin] Marmara University Hospital, Department of RadiologyIstanbul, Turkey. [Guney, Ilter] Marmara University, School of Medicine, Department of Medical BiologyIstanbul, Turkey. [KOTILOGLU, Esin] Marmara University, School of Medicine, Department of Pathology, Altunizade, 81190 Istanbul, Turkey. RP Kaya, H (reprint author), Marmara University, School of Medicine, Department of Pathology, 81190 Istanbul, Turkey. EM hkaya@superonline.com CR Pettine S, Place R, Babu S, et al: Stereotactic breast biopsy accurate, minimally invasive, and cost effective. Am J Surg 171:474-476, 1996. Wallace JE, Sayler C, McDowell NG, et al: The role of stereotactic biopsy assessment of nonpalpable breast lesions. Am J Surg 172:471-473, 1996. Timothy WJ, Kalliopi PS, Prioleau JE, et al: Do prognostic marker studies on core needle biopsy specimens of breast carcinoma accurately reflect the marker status of the tumor. Mod Pathol 11:259-264, 1998. Bargmann CI, Hung MC, Weinberg RA: The neu oncogene encodes an epidermal growth factor receptor-related protein. Nature 319:226-230, 1986. Slamon DJ, Clark GM, Wong SG, et al: Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235:177-182, 1987. Evans AJ, Pider SE, Ellis IO, et al: Correlations between the mammographic features of ductal carcinoma in situ, DCIS, and C-erbB-2 oncogene expression. Nottingham Breast Team: Clin Radiol 49:559-562, 1994. Pauletti G, Godolphin W, Press MF, et al: Detection and quantitation of HER-2/neu gene amplification in human breast cancer archival material using fluorescence in situ hybridization. Oncogene 13:63-72, 1996. Bankfalvi A, Simon R, Brandt B, et al: Comparative methodological analysis of erbB-2/HER-2 gene dosage, chromosomal copy number and protein overexpression in breast carcinoma tissues for diagnostic use. Histopathol 37:411-419, 2000. Jimenez RE, Wallis T, Tabasczka P, et al: Determination of Her- 2/neu status in breast carcinoma: comparative analysis of immunohistochemistry and fluorescent in situ hybridization. Mod Pathol 13:37-45, 2000. Press MF, Bernstein L, Thomas PA, et al: HER-2/neu gene amplification characterized by fluorescence in situ hybridization: poor prognosis in node-negative breast carcinomas. J Clin Oncol 15:2894-2904, 1997. Persons DL, Borelli KA, Hsu PH: Quantitation of HER-2/neu and c-myc gene amplification in breast carcinoma using fluorescence in situ hybridization. Mod Pathol 10:720-727, 1997. Hoang MP, Sahin AA, Ordonez NG, et al: Her-2/neu gene amplification compared with Her-2/neu protein overexpression and interobserver reproducibility in invasive breast carcinoma. Am J Clin Pathol 113:852-859, 2000. Ratcliffe N, Wells W, Wheeler K, et al:The combination of in situ hybridization and immunohistochemical analysis: an evaluation of Her2/neu expression in paraffin-embedded breast carcinomas and adjacent normal appearing breast epithelium. Mod Pathol 10:1247-1252, 1997. Scorilas A, Yotis J, Strvolemos K, et al: c-erbB-2 overexpression may be used as an independent prognostic factor for breast cancer patients. Anticancer Res 15:1543-1648, 1995. An H-X, Niederacher D, Dominik SI, et al: Int-2 and c-erbB-2 gene amplification detected in 70 frozen human breast carcinomas by quantitative polymerase chain reaction. Anticancer Res 17:3133-3136, 1997. Slamon DJ, Godolphin W, Jones LA, et al: Studies of the HER- 2/neu proto-oncogene in human breast and ovarian cancer. Science 244:707-712, 1989. Hopman AHN, Ramaekers FCS, Raap AK, et al: In situ hybridization as a tool to study numerical chromosome aberrations in solid bladder tumors. Histochem 89:307-316, 1988. De Potter CR, Schelfout AM: The neu-protein and breast cancer. Virchows Arch 426:107-115, 1995. Ross JS, Fletcher JA: The HER-2/neu oncogene in breast cancer: prognostic factor, predictive factor, and target for therapy. Stem Cells 16:413-428, 1998. Press MF, Hung G, Godolphin W, et al: Sensitivity of HER- 2/neu antibodies in archival tissue samples:potential source of error in immunohistochemical studies of oncogene expression. Cancer Res 54:2771-2777, 1994. Tubbs RR, Pettay JD, Roche PC, et al: Discrepancies in clinical laboratory testing of eligibility for transtuzumab therapy: apparent immunohistochemical false positives do not get the message. J Clin Oncol 15:2714-2721, 2001. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2001 VL 7 IS 4 BP 279 EP 283 PG 5 ER PT J AU Croce, VM Rabassa, EM Price, RM Segal-Eiras, A AF Croce, V Maria Rabassa, E Martin Price, R Mike Segal-Eiras, Amada TI MUC1 Mucin and Carbohydrate Associated Antigens as Tumor Markers in Head and Neck Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Head and Neck Squamous Cell Carcinoma (HNSCC); MUCI mucin; carbohydrate associated antigens ID Head and Neck Squamous Cell Carcinoma (HNSCC); MUCI mucin; carbohydrate associated antigens AB An immunological analysis to study MUC1 mucin core protein and carbohydrate associated antigens as tissue tumor markers in head and neck carcinoma was performed. Twenty nine patients with the following tumor localizations were included: tongue (n=10), larynx (n=8), oral cavity (n=4), maxillary sinus (n=3), tonsillar ring (n=3) and pharynx (n=1); seven samples of epithelium obtained from normal organs at the same localizations were studied as controls. Immunohistochemical analysis was performed following standard procedures and reaction was graded according to staining intensity and distribution. From each tissue section, membrane, cytoplasmic and nuclear moieties were obtained by differential centrifugation with subsequent fractionation by density gradient centrifugation (6M guanidium chloride-CsCl); subcellular moieties and CsCl derived fractions were analyzed by immunoblotting. Monoclonal antibodies (MAbs) reacting with the core protein of MUCI (C595) and associated carbohydrate antigens were: Tn, 83D4 MAb; Lewis y antigen (Le y), C14 MAb; Lewis x antigen (Le x), KM380 MAb and sialyl Lewis x (sLe x), KM93 MAb. Statistical analysis was undertaken by Spearman rank correlation. In tumor samples, the immunohistochemical identification of MUCl core protein and associated antigens was extended; differences were found in the pattern and intensity of expression; results were corroborated by immunoblotting although in a few samples there was not coincidence between both methods. Localization, tumor mass or node involvement did not show significant differences for any of the antigens studied. Conclusions: 1) head and neck carcinoma expressed MUCI and associated carbohydrate antigens in high levels; 2) no relationship between antigenic expression and tumor status was found. C1 [Croce, V Maria] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120, 1900 La Plata, Argentina. [Rabassa, E Martin] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120, 1900 La Plata, Argentina. [Price, R Mike] University of Nottingham, School of Pharmaceutical SciencesNottingham, UK. [Segal-Eiras, Amada] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120, 1900 La Plata, Argentina. RP Segal-Eiras, A (reprint author), Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), 1900 La Plata, Argentina. EM as-eiras@netverk.com.ar CR Brakenhoff RH, Stroomer JGW, den Brink C, et al: Sensitive detection of squamous cells in bone marrow and blood of head and neck cancer patients by E48 reverse transcriptase polymerase chain reaction. Clin Cancer Res 5:725-732, 1999. Brennan JA, Mao L, Hruban RH, et al: Molecular assessment of histopathological staging in squamous cell-carcinoma of the head and neck. N Engl J Med 332:429-435, 1995. Brown A, Feizi T, Gooi HC: A monoclonal antibody against human colonic adenoma recognises a difucosylated type-2 blood group chains. Biosci Rep 3:163-169, 1983. Burchell J, Durbin H, Taylor-Papadimitriou J: Complexity of expression of antigenic determinants recognised by monoclonal antibodies HMFG-1 and HMFG-2 in normal and malignant human mammary epithelial cells. J Immunol 131:508-513, 1983. Croce MV, Price M R, Segal-Eiras A: Expression of monoclonal antibody-defined antigens in fractions isolated from human breast carcinoma and patients’ serum. Cancer Immunol Immunother 40:132-137, 1995. Croce MV, Colussi AG, Price MR, et al: Immunohistopathological characterization of spontaneous metastases in a human lung mucoepidermoid adecarcinoma, HLMC, xenograft. Pathol Oncol Res 4:259-266, 1998. Croce MV, Price MR, Segal-Eiras A: Detection and isolation of MUC 1 mucin from larynx squamous cell carcinoma. Pathol Oncol Res 2:93-99, 2000. Federici MF, Kudryashov V, Saigo P, et al: Selection of carbohydrte antigens in human epithelial ovarian cancers as targets for immunotherapy: serous and mucinous tumors exhibit distinctive patterns of expression. Int J Cancer 81:193-198, 1999. Feickert HJ, Anger BR, Cordon-Cardo C, et al: Cell-surface antigens of human lung tumors detected by mouse monoclonal antibodies: definition of blood-group- and non-blood-grouprelated antigenic systems. Int J Cancer 46: 1007-1013, 1990. Fukuda M: Possible roles of tumor-associated carbohydrate antigens. Cancer Res 56:2237-2244, 1996. Gendler SJ, Spicer AP: Epithelial mucin genes. Ann Rev Physiol 57:607-634, 1995. Griffiths A, Burchell J, Gendler S, et al: Immunological analysis of mucin molecules expressed by normal and malignant epithelial cells. Int J Cancer 40:319-327, 1987. Hanai N, Shitara K, Yoshida H: Generation of monoclonal antibodies against human lung squamous cell carcinoma and adenocarcinoma using mice rendered tolerant to normal human lung. Cancer Res., 46:4438-4443, 1986. Hakomori S: Aberrant glycosylation in cancer cell membranes as focused on glycolipids: overview and perspectives. Cancer Res. 45:2406-2414, 1985. Hilkens J, Ligtenberg JL, Vos HL, Litvinov S: Cell membraneassociated mucins and their adhesion-modulating property. Trends Biochem Sci 1992; 17:359-63, 1992. Ikeda Y, Kuwano H, Ikebe M, et al: Immunohistochemical detection of CEA, CA19-9 and DF3 in esophageal carcinoma limited to the submucosal layer. J Sur Oncol 56:7-12, 1994. International Union Against cancer, UICC, Classification of Malignant tumors. ED.L.H. Sobin and Ch. Wittekind, 5th edition, New York, 1997. Ioannides CG, Fisk B, Jerome KR, Irimura T: Cytotoxic T cells from ovarian malignant tumors can recognize polymorphic epithelial mucin core peptides. J Immunol 151:3693-3703, 1993. Itoh T, Yonezawa S, Nemoto M et al: expression of mucin antigen and Lewis x-related antigens in carcinoma s and dysplasia of the pharynx and larynx. Pathol Int 46: 646-655, 1996. Jeannon JP, Stafford FW, Soames JV, Wilson JA: Altered MUC1 and MUC2 glycoprotein expression in laryngeal cancer. Otolaryngol Head and Neck Surg 124:199-202, 2001. Jerome KR, Barnd DL, Bendt KM: Cytotoxic T-lymphocytes derived from patients with breast adenocarcinoma recognize an epitope present on the protein core of a mucin molecule preferentially expressed by malignant cells. Cancer Res 5:2908-2916, 1991. Laemmli UK: Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature, Lond, 227:680- 681, 1980. Nakagoe T, Sawai T, Tsuji T, et al: Preoperative serum levels of sialyl Lewis, a), sialyl Lewis(x, and sialyl Tn antigens as prognostic markers after curative resection for colorectal cancer. Cancer Detect Prev 25:299-308, 2001. Nakamori S, Kameyama M, Imaoka S, et al:Involvement of carbohydrate antigen sialyl Lewis x in colorectal cancer metastasis. Dis Colon Rectum 40:420-431, 1997. Nitta T, Sugihara K, Tsuyama S et al: Immunohistochemical study of MUC1 mucin in premalignant oral lesions and oral squamous cell carcinoma. Cancer 88:245-254, 2000. Pancino GF, Osinaga E, Vorauher W, et al: Production of a monoclonal antibody as immunohistochemical marker on paraffin embedded tissues using a new immunization method. Hybridoma 9:389, 1990. Price MR, Edwards S, Owainati A, et al: Multiple epitopes on a human breast carcinoma associated antigen. Int J Cancer 36:567, 1985. Price MR, Pugh J, Hudecz F, O’Sullivan, C et al: Immunological and structural features of the protein core of human polymorphic epithelial mucin. Mol Immunol 27:795-801, 1990. Regimbald LH, Pilarski LM, Longenecker BM: The breast mucin MUC1 as a novel adhesion ligand for endothelial intercellular adhesion molecule 1 in breast cancer. Cancer Res 56:4244-4249, 1996. Rubin JS, Bloor BK, Hart IR, Morgan PR: Muo-1 gene expression in head and neck squamous cell carcinoma. J Laryngol Otol 114:772-776, 2000. Svitacheva N, Davies JR, Lessuileur T: Characterization of mucins produced by human HT29-MTX cell line. 5`h International workshop on Carcinoma Associated Mucins, Cambridge, LJK 1998. Swallow DM, Griffiths B, Bramwell ME, et al: Detection of the urinary „pum“polymorphism by the tumour binding monoclonal antibodies Cal, Ca2, HMFG-I and HMFG-2. Dis Markers 4:247-254, 1986. Takada A, Ohmori K, Yoneda T, et al: Contribution of carbohydrate antigens sialyl Lewis a and sialyl Lewis x to adhesion of human cancer cells to vascular endothelium. Cancer Res. 53:354-361, 1993. Taylor-Papadimitriou J, Finn O: Biology, biochemistry and immunology of carcinoma associated mucins. Immunol Today 18:105-107, I 997. Towbin H, Staehelin T, Gordon J: Electrophoretic transfer of proteins from polyacrilamide gels to nitrocellulose sheets: procedures and some applications. Proc Natl Acad Sci USA 76:4350-4354, 1979. Vernham, GA, Crowther JA: Head and neck carcinoma: stage at presentation. Clin Otolaryngol 19:120-124, 1994. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2001 VL 7 IS 4 BP 284 EP 291 PG 8 ER PT J AU Berki, T David, M Bone, B Losonczy, H Vass, J Nemeth, P AF Berki, Timea David, Marianna Bone, Beata Losonczy, Hajna Vass, Janos Nemeth, Peter TI New diagnostic tool for differentiation of idiopathic SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE hypereosinophilic syndrome; IL-5; INFg; Th1-Th2 balance; flow cytometry ID hypereosinophilic syndrome; IL-5; INFg; Th1-Th2 balance; flow cytometry AB The hypereosinophilic syndrome (HES) is a very rare disease, characterized by persistent eosinophilia with tissue involvement and organ dysfunction which often precedes a subsequent T cell lymphoma. Interleukin-5 secreted by a T lymphocyte subpopulation has been described in previous reports as the most important factor responsible for the prolonged lifespan of the eosinophils. The goal of the present study was to describe a fast, simple diagnostic method for the differentiation of HES and secondary eosinophilic states. Beside the surface marker analysis of peripheral blood mononuclear cells (PBMC) we measured surface bound IgE molecules on lymphocytes and eosinophil cells, intracellular cytokines (IL-5, INFg g) in CD4+ lymphocytes and eosinophil major basic protein (MBP) in eosinophils using flow cytometric detection method. The appearance of an IL-5 producing cell population with a decreased number of INFg gpositive lymphocytes was characteristic for the blood samples of HES patients. Predominance of Th2 cells with the appearance of a CD8+/CD3–/CD56+ cell population was restricted for the HES cases and could not be detected in secondary eosinophilic individuals. Our flow cytometric cytokine detection method (with parallel cell surface marker analysis) does not require cell separation or long term cell culture steps previously described for the detection of IL-5 producing cells. Therefore it seems to be a more appropriate approach for the differential diagnosis of primary and secondary eosinophilic states. C1 [Berki, Timea] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12., H-7643 Pecs, Hungary. [David, Marianna] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Bone, Beata] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12., H-7643 Pecs, Hungary. [Losonczy, Hajna] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Vass, Janos] University of Pecs, Department of PathologyPecs, Hungary. [Nemeth, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12., H-7643 Pecs, Hungary. RP Berki, T (reprint author), University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, H-7643 Pecs, Hungary. EM timea.berki@aok.pte.hu CR Bain BJ: Eosinophilic leukemias and the idiopathic hypereosinophilic syndrome Br J Haematol 95:2-9, 1996. Bain BJ:Hypereosinophilia. Curr Opin Hematol 7:21-5, 2000. Berki T, Kumanovics G, Kumanovics A, et al: Production and flow cytometric application of a monoclonal anti-glucocorticoid receptor antibody. J Immunol Methods 214:19-27, 1998. Brugnoni D, Airo P, Rossi G, et al:A case of hypereosinophilic syndrome is associated with the expansion of a CD3- CD4+ T-cell population able to secrete large amounts of interleukin-5. Blood 87:1416-1422, 1996. Chusid MJ, Dale DC, West BC, et al: The hypereosinophilic syndrome: analysis of fourteen cases with review of the literature. Medicine, Baltimore, 54:1-27, 1975. Clutterbuck EJ, Hirst EM, Sanderson CJ: Human interleukin-5, IL- 5, regulates the production of eosinophils in human bone marrow cultures: comparison and interaction with IL-1, IL-3, IL-6 and GMCSF. Blood 73:1504-1512, 1989. Cogan E, Schandene L, Crusiaux A, et al:Clonal proliferation of type 2 helper T cells in a man with the hypereosinophilic syndrome. N Engl J Med 330:535-538, 1994. Desreumaux P:Synthesis of interleukin-5 by activated eosinophils in patients with eosinophilic heart diseases. Blood 82:1553-1560, 1993. Kim CJ, Park SH, Chi JG: Idiopathic hypereosinophilic syndrome terminating as disseminated T-cell lymphoma. Cancer 67:1064-1069, 1991. Kitano K, Ishikawa N, Shimodaira S, et al: Eosinophilia associated with clonal T-cell proliferation. Leukemia Lymphoma 27:335-342, 1997. Krug N, Thurau AM, Lackie P, et al: A flow cytometric method for the detection of intracellular basic proteins in unseparated peripheral blood and bone marrow eosinophils. J Immunol Methods 190:245- 254, 1996. Menssen HD, Renkl HJ, Rieder H, et al: Distinction of eosinophilic leukemia from idiopathic hypereosinophilic syndrome by analysis of Wilms’ tumor gene expression. Br J Haematol 101:325-334. 1998. Metz J, McGrath KM, Savoia HF, et al: T cell lymphoid aggregates in bone marrow in idiopathic hypereosinophilic syndrome. J Clin Path 46:955-958, 1993. Oliver JW, Deol I, Morgan DL, et al: Chronic eosinophilic leukemia and hypereosinophilic syndromes. Proposal for classification, literature review and report of a case with a unique chromosomal abnormality. Cancer Gen Cytogen 107:111-117, 1998. Owen WF, Rothenberg ME, Petersen J: Interleukin-5 and phenotipically altered eosinophils in the blood of patients with the hypereosinophilic syndrome. J Exp Med 170:343-348, 1989. Pinto A, Aldinucci D, Gloghini A, et al:The role of eosinophils in the pathobiology of Hodgkin’s disease. Ann Oncol 8:89-96, 1997. Raghavachar A, Fleicher S, Frickhofen N, et al: T lymphocyte control of human eosinophilic granulopoiesis. Clonal analysis in an idiopathic hypereosinophilic syndrome. J Immunol 139:3753-3758, 1987. Rauch AE, Amyot KM, Ducc HG, et al:Hypereosinophilic syndrome and myocardial infarction in a 15-year-old. Pediatric Pathol Lab Med 17:469-486, 1997. Roufosse F, Schandene L, Sibille C, et al: T-cell receptor-independent activation of clonal Th2 cells associated with chronic hypereosinophilia. Blood 94:994-1002, 1999. Roufosse F, Schandene L, Sibille C, et al:Clonal Th2 lymphocytes in patients with the idiopathic hypereosinophilic syndrome. Br J Haematol 109:540-8, 2000. Satoh T, Sun L, Li MS, et al: Interleukin-5 mRNA levels in blood and bone marrow mononuclear cells from patients with the idiopathic hypereosinophilic syndrome. Immunology 83:308-312, 1994. Schrezenmeier H, Thome SD, Tewald F, et al:Interleukin-5 is the predominant eosinophilopoietin produced by cloned T lymphocytes in hypereosinophilic syndrome. Exp Hematol 21:358-.365. 1993 Simon HU, Plotz SG, Dummer R, et al: Abnormal clones of T cells producing interleukin-5 in idiopathic eosinophilia. N Engl J Med 341:1112-1120, 1999. Sornasse T, Larenas PV, Davis KA, et al: Differentiation and stability of Th1 and Th2 cells derived from naive human neonatal CD4+ T cells, analyzed at the single cell level. J Exp Med 84:473-483, 1996. Staynov DZ, Lee TH: Expression of interleukin-5 and granulocytemacrophage colony-stimulating factor in human peripheral blood mononuclear cells after activation with phorbol myristate acetate. Immunology 75:196-201, 1992. Tai PC, Sun L, Spry CJ: Effects of IL-5, granulocyte/macrophage colony stimulating factor, GM-CSF, and IL-3 on the survival of human blood eosinophils in vitro. Clin Exp Immunol 85:312-316, 1991. Trainor KJ, Brisco MJ, Wan JH, et al: Gene rearrangement in B- and T-lymphoproliferative disease detected by the polymerase chain reaction. Blood 78:192-196, 1991. Walsh GM: Human eosinophils: Their accumulation, activation and fate. Br J Haematol 97:701-709, 1997. Weller PF, Bubley GJ: The idiopathic hypereosinophilic syndrome. Blood 83:2759-2779, 1994. Weller PF: The immunobiology of eosinophils. N Engl J Med 324:1110-1118, 1991. Yamaguchi Y, Suda T, Ohta S, et al: Analysis of the survival of mature human eosinophils: Interleukin-5 prevents apoptosis in mature human eosinophils. Blood 78:2542-2547, 1991. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2001 VL 7 IS 4 BP 292 EP 297 PG 6 ER PT J AU Ribeiro-Silva, A Luzzatto, F Chang, D Zucoloto, S AF Ribeiro-Silva, Alfredo Luzzatto, Felipe Chang, Daniel Zucoloto, Sergio TI Limitations of Fine-Needle Aspiration Cytology to Diagnose Metaplastic Carcinoma of the Breast SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE metaplastic carcinoma; breast; fine-needle aspiration ID metaplastic carcinoma; breast; fine-needle aspiration AB Metaplastic carcinoma is a very rare breast neoplasm that is often confused with benign and others malignant entities on both clinical and conventional histopathologic basis. Three cases of metaplastic carcinoma of breast are presented. The difficulties found on fine needle aspiration cytology and the limitations of this procedure are discussed as well the main features of this tumor. C1 [Ribeiro-Silva, Alfredo] Sao Paulo University School of Medicine, Department of Pathology, Av. Bandeirantes, s/n – Campus Universitario Monte Alegre., 14048-900 Sao Paulo, Brazil. [Luzzatto, Felipe] Sao Paulo University School of Medicine, Department of Pathology, Av. Bandeirantes, s/n – Campus Universitario Monte Alegre., 14048-900 Sao Paulo, Brazil. [Chang, Daniel] Sao Paulo University School of Medicine, Department of Pathology, Av. Bandeirantes, s/n – Campus Universitario Monte Alegre., 14048-900 Sao Paulo, Brazil. [Zucoloto, Sergio] Sao Paulo University School of Medicine, Department of Pathology, Av. Bandeirantes, s/n – Campus Universitario Monte Alegre., 14048-900 Sao Paulo, Brazil. RP Ribeiro-Silva, A (reprint author), Sao Paulo University School of Medicine, Department of Pathology, 14048-900 Sao Paulo, Brazil. EM ardsilva@rpa.fmrp.usp.br CR Castella E, Gomez-Plaza MC, Urban A, et al: Fine-needle aspiration biopsy of metaplastic of the breast: Report of a case with abundant myxoid ground substance. Diagn Cytopathol 14:325- 327, 1996. Yen H, Florentine B, Kelly LK, et al: Fine-needle aspiration of a metaplastic breast carcinoma with extensive melanocytic differentiation: a case report. Diagn Cytopathol 23:46-50, 2000. Brenner RJ, Turner RR, Schiller V, et al. Metaplastic carcinoma of the breast: report of three cases. Cancer 82:1082-1087, 1998. Park JM, Han BK, Moon WK, et al: Metaplastic carcinoma of the breast: mammographic and sonographic findings. J Clin Ultrasound 28:176-186, 2000. Chao TC, Wang CS, Chen SC, et al: Metaplastic carcinomas of the breast. J Surg Oncol 71:220-225, 1999. Rayson D, Adjei AA, Suman VJ, et al: Metaplastic breast cancer: prognosis and response to systemic therapy. Ann Oncol 10:413-419, 1999. Chhieng DC, Cangiarella JF, Waisman J, et al: Fine-needle aspiration cytology of spindle cell lesions of the breast. Cancer 87:359-371, 1999. Jebsen PW, Hagmar BM, Nesland JM: Metaplastic breast carcinoma. A diagnostic problem in fine needle aspiration biopsy. Acta Cytol 35:396-402, 1991. Straathof D, Yakimets WW, Mourad WA: Fine-needle aspiration cytology of sarcomatoid carcinoma of the breast: a cytologically overlooked neoplasm. Diagn Cytopathol 16:242-246, 1997. Gupta RK: Cytodiagnosis patterns of metaplastic breast carcinoma in aspiration samples: a study of 14 cases. Diagn Cytopathol 20:10-12, 1999. Chhieng C, Cranor M, Lesser ME, et al: Metaplastic carcinoma of the breast with osteocartilaginous heterologous elements. Am J Surg Pathol 22:188-194, 1998. Gupta RK: Fine needle aspiration cytodiagnosis of primary and metastatic squamous cell carcinoma of the breast. Acta Cytol 41:692-696,1997. Kokufu I, Yamamoto M, Fukuda K, et al: Squamous cell carcinoma of the breast: three case reports. Breast cancer 6:63-68, 1999. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2001 VL 7 IS 4 BP 298 EP 300 PG 3 ER PT J AU Kravchick, S Cytron, Sh Lobik, L Altshuler, A Kravchenko, Y Ben-Dor, D AF Kravchick, Sergey Cytron, Shmuel Lobik, Leonid Altshuler, Alexander Kravchenko, Yakov Ben-Dor, David TI Clot Retention and Spontaneous Rupture with Secondary Pneumatosis of Bladder Wall Following Routine Cystoscopy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE bladder neoplasm; cystoscopy; bladder injury; pneumatosis ID bladder neoplasm; cystoscopy; bladder injury; pneumatosis AB In this article we report an unusual case of spontaneous rupture of bladder wall following office-cystoscopy. It took place in a patient who suffered from low-stage highgrade carcinoma of bladder with the different aggressive behavior. Finally, he underwent radical cystectomy, which showed micropapillary carcinoma and pneumatosis within the bladder wall. The cause of the latter finding is rather puzzling and has been never reported previously. C1 [Kravchick, Sergey] Barzialy Medical Center, Urology Department, Ha Histadrut 1, 78306 Ashkelon, Israel. [Cytron, Shmuel] Barzialy Medical Center, Urology Department, Ha Histadrut 1, 78306 Ashkelon, Israel. [Lobik, Leonid] Barzialy Medical Center, Urology Department, Ha Histadrut 1, 78306 Ashkelon, Israel. [Altshuler, Alexander] Barzilay Medical Center, Department of RadiologyAshkelon, Israel. [Kravchenko, Yakov] Barzialy Medical Center, Pathology DepartmentAshkelon, Israel. [Ben-Dor, David] Barzialy Medical Center, Pathology DepartmentAshkelon, Israel. RP Kravchick, S (reprint author), Barzialy Medical Center, Urology Department, 78306 Ashkelon, Israel. CR Smith DP, Goldman SM, Fishman EK: Rupture of the urinary bladder following cystoscopic clot evacuation : report of two cases diagnosed by CT. Abdom Imaging 19:177, 1994. Hasui Y, Osada Y, Kitada S, Nishi S: Significance of invasion to the mucosae on the progression of superficial bladder cancer. Urology 43:782, 1994. Maranchie JK, Bouyounes BT, Zhang PL, et al: Clinical and pathological characteristics of micropapillary transitional cell carcinoma: a highly aggressive variant. J Urol 163:748, 2000. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2001 VL 7 IS 4 BP 301 EP 302 PG 2 ER PT J AU Sauer, R AF Sauer, Rolf TI Adjuvant and Neoadjuvant Radiotherapy and Concurrent Radiochemotherapy for Rectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE rectal cancer; radiotherapy; radiochemotherapy ID rectal cancer; radiotherapy; radiochemotherapy AB Radical surgery with negative margins remains the most important prognostic factor in the treatment of rectal cancer. Combined modality treatment is the recommended standard adjuvant therapy for patients with locally advanced rectal cancer in the USA and in Germany. During the last decade substantial progress has been made in treatment modalities: surgical management currently includes a broad spectrum of operative procedures ranging from radical operations to innovative sphincter-preserving techniques. Specialized groups have reported excellent local control rates with total mesorectal excision (TME) alone. New and improved radiation techniques (conformal radiotherapy, intraoperative radiotherapy) and innovative schedules (protracted intravenous infusion, chronomodulated infusion) and combinations (oxaliplatin, irinotecan) of chemotherapy may have the potential to further increase the therapeutic benefit of adjuvant treatment. Moreover, the basic issue of timing of radio-(chemo-)therapy - preoperative versus postoperative - within a multimodality regimen is currently being addressed in prospective trials. Evidently, the current monolithic approaches, established by studies conducted more than a decade ago, to apply either the same schedule of postoperative radiochemotherapy to all patients with stage II/III rectal cancer or to give preoperative intensive short-course radiation according to the Swedish concept for all patients with resectable rectal cancer irrespective of tumor stage and treatment goal (e.g. sphincter preservation), need to be questioned. This review will discuss different irradiation settings in more recent and ongoing studies of perioperative radiotherapy for rectal cancer and will focus on the issue which patient should receive radiotherapy at all, and if so, how and when? C1 [Sauer, Rolf] University of Erlangen, Department of Radiation Oncology, Universitatsstr. 27, 91054 Erlangen, Germany. RP Sauer, R (reprint author), University of Erlangen, Department of Radiation Oncology, 91054 Erlangen, Germany. CR NIH Consensus Conference: Adjuvant therapy for patients with colon and rectal cancer. JAMA 264:1444-1450, 1990. Junginger T, Hossfeld DK, Sauer R, Hermanek P: Adjuvante Therapie bei Kolon- und Rektumkarzinom. Dt Arztebl 96:A- 698-700, 1999. MacFarlane JK, Ryall RDH, Heald RJ: Mesorectal excision for rectal cancer. Lancet 341:457-460, 1993. Enker WE:Total mesorectal excision – the new golden standard of surgery for rectal cancer. Ann Med 29:127-133, 1997. Kockerling F, Reymond MA, Altendorf-Hofmann A, et al: Influence of surgery on metachronous distant metastases and survival in rectal cancer. J Clin Oncol 16:324-329, 1998. Willett CG, Badizadegan K, Ancukiewicz M, et al: Prognostic factors in stage T3N0 rectal cancer. Do all patients require postoperative pelvic irradiation and chemotherapy? Dis Colon Rectum 42:167-173, 1999. Hermanek P, Wiebelt H, Staimmer D, et al: Prognostic factors of rectum carcinoma. Experience of the German Multicenter Study SGCRC. Tumori 81:60-64, 1995. Hohenberger W: The effect of specialization or organization in rectal cancer surgery. In Soreide O, Norstein J, eds): “Rectal cancer surgery”. Berlin: Springer; 1996. p. 353-363. Douglass HO, Moertel CG, Mayer RJ, et al: Survival after postoperative combination treatment of rectal cancer. New Engl J Med 315:1294-1299, 1986. Krook JE, Moertel CG, Gunderson LL, et al: Effective surgical adjuvant therapy for high-risk rectal carcinoma. New Engl J Med 324:709-715, 1991. Tveit KM, Guldvog I, Hagen S,et al: Randomized controlled trial of postoperative radiotherapy and short-term time-scheduled 5-fluorouracil against surgery alone in the treatment of Dukes B and C rectal cancer. Br J Surg 84:1130-1135, 1997. Gastrointestinal Tumor Study Group, GITSG): Radiation therapy and fluorouracil with or without semustine for the treatment of patients with surgical adjuvant adenocarcinoma of the rectum. J Clin Oncol 10:549-557, 1992. O`Connell MJ, Martenson JA, Wieand HS, et al: Improving adjuvant therapy for rectal cancer by combining protractedinfusion fluorouracil with radiation therpy after curative surgery. New Engl J Med 331:502-507, 1994. Kapiteijn E, Marijnen CAM, Nagtegaal ID, et al: Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. N Engl J Med 345:638-646, 2001. Wolmark N, Wieand HS, Hyams DM, et al: Randomized trial of postoperative adjuvant chemotherapy with or without radiotherapy for carcinoma of the rectum: National Surgical Adjuvant Breast and Bowel Project Protocol R-02. J Natl Cancer Inst 92:388-396, 2000. Fisher B, Wolmark N, Rockette H, et al: Postoperative adjuvant chemotherapy or radiation therapy for rectal cancer: Results from NSABP Protocol R-01. J Natl Cancer Inst 80:21-29, 1988. Kollmorgen CF, Meagher AP, Wolf BG, et al: The long-term effect of adjuvant postoperative chemoradiotherapy for rectal carcinoma on bowel function. Ann Surg 220:676-682, 1994. Ooi BS, Tjandra JJ, Green MD: Morbidities of adjuvant chemotherapy and radiotherapy for resectable rectal cancer. An overview. Dis Colon Rectum 42:403-418, 1999. Withers HR, Peters LJ, Taylor MG: Dose-response relationship for radiation therapy of subclinical disease. Int J Radiat Oncol Biol Phys 31:353-359, 1995. Tepper JE, O‘Connell MJ, Petron GR, et al: Adjuvant postoperative fluorouracil-modulated chemotherapy combined with pelvic radiation therapy for rectal cancer: initial results of Intergroup 0114. J Clin Oncol 15:2030-2039, 1997. Link KH, Staib L, Bernhart H, et al: Acceptance and toxicity of postoperative adjuvant therpy in colon and rectal cancer. Onkologie 20:235-238, 1997. Douillard JY, Cunningham D, Roth AD, et al: A randomized phase III trial comparing irinotecan + 5-FU/folinic acid to the same schedule of 5-FU/folinic acid in patients with metastatic colorectal cancer as front line therapy [Abstract]. Proc Am Soc Clin Oncol 18:233a, 1999. Giacchetti S, Perpoint B, Zidani R, et al: Phase III multicenter randomized trial of oxaliplatin added to chronomodulated fluorouracil- leucovorin as first-line treatment of metastatic colorectal cancer, J Clin Oncol 18:136-147, 2000. Rodel C, Fietkau R, Grabenbauer GG, et al: Akuttoxizitat der simultanen Radiochemotherapie des Rektumkarzinoms [Acute toxicity of simultaneous radiochemotherapy for rectal cancer]. Strahlenther Onkol 173:414-420, 1997. Minsky BD, Cohen AM, Kemeny N, et al: Combined modality therapy of rectal cancer: decreased acute toxicity with the preoperative approach. J Clin Oncol 10:1218-1224, 1992. Pahlman L: Neoadjuvant and adjuvant radio- and radiochemotherapy of rectal carcinomas [Review]. Int J Colorectal Dis 15:1-8, 2000. Swedish Rectal Cancer Trial: Improved survival with preoperative radiotherapy in resectable rectal cancer. New Engl J Med 336:980-987, 1997. Minsky BD: Adjuvant therapy for rectal cancer – a good first step [letter]. New Engl J Med 14:1016-1017, 1997. Rodel C, Hohenberger W, Sauer R:Adjuvante und neoadjuvante Therapie des Rektumkarzinoms [Review] [Adjuvant and neoadjuvant therapy for rectal cancer]. Strahlenther Onkol 174:497-504, 1998. Francois Y, Nemoz CJ, Baulieux J, et al: Influence of the interval between preoperative radiation therapy and surgery on downstaging and the rate of sphincter-sparing surgery for rectal cancer: the Lyon R90-01 randomized trial. J Clin Oncol 17:2396-2402, 1999. Frykholm-Jansson G, Sintorn K, Montelius A, et al: Acute lumbosacral plexopathy after preoperative radiotherapy in rectal carcinoma. Radiother Oncol 38:121-130, 1996. Sauer R, Fietkau R, Wittekind C, et al: Adjuvant versus neoadjuvant radiochemotherapy for locally advanced rectal cancer. A progress report of a phase-III randomized trial, protocol CAO/ARO/AIO-94). Strahlenther Onkol 177:173-18, 2001. Dahlberg M, Glimelius B, Graf W, et al: Preoperative irradiation affects functional results after surgery for rectal cancer. Dis Colon Rectum 41:543-551, 1998. Chan A, Wong A, Langevin J, et al: Preoperative concurrent 5- fluorouracil infusion, mitomycin C and pelvic radiation therapy in tethered and fixed rectal carcinoma. Int J Radiat Oncol Biol Phys 25:791-799, 1993. Minsky BD, Cohen AM, Kemeny N, et al: The efficacy of preoperative 5-fluorouracil, high-dose leucovorin, and sequential radiation therapy for unresectable rectal cancer. Cancer 71:3486-3492, 1993. Marsh RD, Chu NM, Vauthey JN, et al. Preoperative treatment of patients with locally advanced unresectable rectal adenocarcinoma utilizing continuous chronobiologically shaped 5-fluorouracil infusion and radiation therapy. Cancer 78:217-25, 1996. Videtic GM, Fisher BJ, Perera FE, et al. Preoperative radiation with concurrent 5-fluorouracil continuous infusion for locally advanced unresectable rectal cancer. Int J Radiat Oncol Biol Phys 42:319-324, 1998. Rodel C, Grabenbauer GG, Schick CH, et al: Preoperative radiation with concurrent 5-fluorouracil for locally advanced T4- primary rectal cancer. Strahlenther Onkol 176:161-167, 2000. Minsky BD, Cohen AM, Kemeny N et al: Enhancement of radiation induced downstaging of rectal cancer by fluorouracil and high-dose leucovorin chemotherapy. J Clin Oncol 10: 79-84, 1992. Rau B, Wust P, Gellermann J, et al: Preoperative radio-chemothermo- therapy in locally advanced rectal cancer. Strahlenther Onkol 174:556-565, 1998. Ahmad NR, Nagle DA: Preoperative radiation therapy followed by local excision. Semin Radiat Oncol 8:36-38, 1998. Maghfoor I, Wilkes J, Kuvshinoff B, et al: Neoadjuvant chemoradiotherapy with sphincter-sparing surgery for low lying rectal cancer [Abstract]. Proc Am Soc Clin Oncol 16:274, 1997. Wagman R, Minsky BD, Cohen AM, et al: Sphincter preservation with preoperative radiation therapy and coloanal anastomosis: Long tern follow-up. Int J Radiat Oncol Biol Phys 42:51-57, 1998. Grann A, Minsky BD, Cohen AM: Preliminary results of preoperative 5-fluorouracil, 5-FU), low dose leucovorin, and concurrent radiation therapy for resectable T3 rectal cancer. Dis Colon Rectum 40:515-522, 1997. Rouanet P, Fabre JM, Dubois JB: Conservative surgery for low rectal carcinoma after high-dose radiation. Ann Surg 221:67- 73, 1995. Hyams DM, Mamounas EP, Petrelli N, et al: A clinical trial to evaluate the worth of preoperative multimodality therapy in patients with operable carcinoma of the rectum: a progress report of NSABP R-03. Dis Colon Rectum 40:131-139, 1997. Minsky BD: Adjuvant therapy of rectal cancer [Review]. Semin Oncol 26:540-544, 1999. Pahlman L, Glimelius B: Pre- or postoperative radiotherapy in rectal and rectosigmoid carcinoma. Ann Surg 211:187-195, 1990. Bosset JF, Pavy JJ, Bolla M, et al: Four arms phase III clinical trial for T3-T4 resectable rectal cancer comparing preoperative pelvic irradiation to preoperative irradiation combined with fluorouracil and leucovorin, with or without postoperative adjuvant chemotherapy. EORTC Radiotherapy Cooperative Group. Protocol no. 22921. EORTC Datacenter. Brussels, October 1992. Neve de W, Martijn H, Lybeert MM, et al: Incompletely resected rectum, rectosigmoid, or sigmoid carcinoma: results of postoperative radiotherapy and prognostic factors. Int J Radiat Oncol Biol Phys 21:1297-1302, 1991. Hu KS, Harrison LB: Results and complications of surgery combined with intraoperative radiation therapy for the treatment of locally advanced or recurrent cancers in the pelvis [Review]. Semin Surg Oncol 18:269-278, 2000. Harrison LB, Minsky BD, Enker WE, et al: High dose rate intraoperative radiation therapy, HDR-IORT, as part of the management strategy for locally advanced primary and recurrent rectal cancer. Int J Radiat Oncol Biol Phys 42:325-330, 1998. Gunderson LL: Indications for and results of combined modality treatment of colorectal cancer [Review]. Acta Oncologica 38:7-21, 1999. Gunderson LL, Nelson H, Martenson JA, et al: Locally advanced primary colorectal cancer: intraoperative electron and external beam irradiation +/- 5-FU. Int J Radiat Oncol Biol Phys 37:601-614, 1997. Gunderson LL, Nelson H, Martenson JA, et al: Intraoperative electron and external beam irradiation with or without 5-fluorouracil and maximum surgical resection for previously unirradiated, locally recurrent colorectal cancer. Dis Colon Rectum 39:1379-1395, 1996. Farouk R, Nelson H, Gunderson LL: Aggressive multimodality treatment for locally advanced irresectable rectal cancer [Review]. Br J Surg 84:741-749, 1997. Atektiar KM, Zelefsky MJ, Paty PB, et al: High-dose-rate intraoperative brachytherapy for recurrent colorectal cancer. Int J Radiat Oncol Biol Phys 48:219-226, 2000. Mannaerts GH, Martijn H, Crommelin MA, et al: Intraoperative electron beam radiation therapy for locally recurrent rectal carcinoma. Int J Radiat Oncol Biol Phys 45:297-308, 1999. Mannaerts GH, Martijn H, Crommelin MA, et al: Feasibility and first results of multimodality treatment, combining EBRT, extensive surgery, and IORT in locally advanced primary rectal cancer. Int J Radiat Oncol Biol Phys 47:425-433, 2000. Eble MJ, Lehnert T, Treiber M, et al: Moderate dose intraoperative and external beam radiotherapy for locally recurrent rectal carcinoma. Radiother Oncol 49:167-174,1998. Hohenberger W, Gunther R, Fietkau R: Is Radiochemotherapy necessary in the treatment of rectal cancer? Eur J Cancer 34:441-446, 1998. Tveit KM, Nordlinger B, Penna B, Schmoll HJ: Current conversies in Cancer. Is there a stamdard adjuvant treatment for rectal cancer. Eur J Cancer 34:1827-1853, 1998. Agence Nationale pour le Developpement de l’Evaluation Medicale: Le choix des therapeutiques du cancer du rectum. Conference de Consensus, Paris, 1994. Willett CG: Technical advances in the treatment of patients with rectal cancer. Int J Radiat Ooncol Biol Phys 45:1107-1108, 1999 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 7 EP 17 PG 11 ER PT J AU Wheatley, ND Campbell, E AF Wheatley, N Denys Campbell, Elaine TI Arginine Catabolism, Liver Extracts and Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE arginine; arginase; arginine deiminase; arginine decarboxylase; deprivation; cell cycle checkpoints; cell death; cancer; leukemia ID arginine; arginase; arginine deiminase; arginine decarboxylase; deprivation; cell cycle checkpoints; cell death; cancer; leukemia AB Although it is self evident that cells will not grow in amino acid deficient medium, an observation less well appreciated is that malignant cells are particularly vulnerable to such deprivation, which can lead to their rapid demise. Indeed, the more flagrantly malignant the phenotype (anaplastic the tumor), the more susceptible the cells seem to be to deprivation. While some attempts to employ this strategy in cancer treatment have been made, the difference between normal and malignant cells should be more fully exploited as a means of selectivelyeliminating tumor cell populations. To be successful, information on differences between the normal and the deranged cell cycle engine and checkpoints, especially how these are affected by deprivation, is of crucial importance. Since it is only recently that the controls at restriction points have been elucidated, it is little surprise that earlier attempts to control tumor cell growth by limiting the availability of an essential amino acid have met with limited success. Studies havebeen sporadic and isolated, often with little more than anecdotal descriptions as far as clinical work was concerned. This review concentrates on what has been accomplished primarily in vitro and since about 1950 with regard to argininecatabolism, while recognising that other essential amino acids have also been the focus of attention by some investigators. Treatments have included medium and plasma manipulation, dietary control, enzymatic degradation, and the use of liver extracts. On some occasions, substitution of amino acid analogues has been explored. It is argued that current knowledge, combined with past experience, calls for a much closer examination of the full potential of amino acid (and specifically arginine) deprivation as a means of controlling tumor growth, with greater attention to protocols that might be used to treat human cancers. C1 [Wheatley, N Denys] University of Aberdeen, Department of Cell Pathology, MacRobert Building, 581 King Street, AB24 5UA Aberdeen, UK. [Campbell, Elaine] University of Aberdeen, Department of Cell Pathology, MacRobert Building, 581 King Street, AB24 5UA Aberdeen, UK. RP Wheatley, ND (reprint author), University of Aberdeen, Department of Cell Pathology, AB24 5UA Aberdeen, UK. EM wheatley@abdn.ac.uk CR Wheatley DN Scott LA, Lamb JL, Smith S: Single amino, arginine, restriction: growth and death of cultured HeLa and normal human diploid fibroblasts. Cell Physiol Biochem 10:37-55, 2000. Currie GA, Basham C: Differential arginine dependence and the selective cytotoxic effects of activated macrophages for malignant cell in vitro. Brit J Cancer 38:653-659, 1978. Brittenden J, Heys S, Eremin O: L-arginine and malignant disease: a potential therapeutic role? Eur J Surg Oncol 20:189- 192, 1994. Heys S, Ogston K, Miller I, et al: Potentiation of the response to chemotherapy in patients by dietary supplementation with Larginine; results of a randomised controlled trial. Int J Oncol 12:221-225, 1998. Daly JM, Reynolds J, Thom A, et al: Immune and metabolic effects of arginine in the surgical patient. Arch Surg 208:512- 523, 1988. Anon, Editorial): Aminoacid manipulation and cancer. The Lancet 303-304, 1973. Milner JA: Metabolic aberrations associated with arginine deficiency. J Nutr 115:516-523, 1985. Bach SJ, Lasnitzki I: Some aspects of the role of arginine and arginase in mouse carcinoma 63. Enzymologia 12:198-205, 1947. Vrat V: Inhibitory effects of arginase on mammary carcinoma transplants in strain “A” mice. Permanente Found Med Bul 9:56-59, 1951. Wiswell OB: Effects of intraperitoneally injected arginase on growth of mammary carcinoma implants in mice. Proc Soc Exp Biol Med 76:588-589,1951. Irons WG, Boyd EF: Arginase as anticarcinogenic agent in mice and human beings. Ariz Med 9:39-43, 1952. Bach SJ, Maw GA: Creatine synthesis by tumor-bearing rats. Biochim Biophys Acta 11:69-78, 1953. Bach SJ, Simon-Reuss I:: Arginase, an antimitotic agent in tissue culture. Biochim Biophys Acta 11:396-402,1953. Bach SJ, Killip JD: Purification and crystallisation of arginase. Biochim Biophys Acta 29:273-280, 1958. Bach SJ, Hawkins RA, Swaine D, A short method for the purification of arginase from ox liver. Biochem J 89:263-265, 1963. Bach SJ, Swaine D: The effect of arginase on the retardation of tumor growth. Brit J Cancer 19:379-386,1965. Gilbert DA, Bergel F: The chemistry of xanthine oxidase. Biochem J 90:350-353, 1964. Lieberman I, Ove P: Inhibition of growth of cultured mammalian cells by liver extracts. Biochem Biophys Acta 38:153- 157, 1960. Freed JJ, Schatz SA: Chromosome aberrations in cultured cells deprived of single essential amino acids. Exp Cell Res 55:393- 409, 1969. Holley RW: Evidence that a rat liver “inhibitor” of the synthesis of DNA in cultured mammalian cells is arginase. Biochim Biophys Acta 145:525-527, 1967. Umeda M, Diringer H, Heidelberger: Inhibition of the growth of cultured cells by arginase and soluble proteins from mouse skin. Isr J Med Sci 4:1216-1222, 1968. Freed JJ, Sorof S: The nature of the inhibition of replication of cultured cells by a liver macromolecule. Wistar Inst Symp Monogr 7:15-24, 1967. Sasada M, Terayama H: The nature of inhibitors of DNA synthesis in rat-liver hepatoma cells. Biochim Biophys Acta 190:73-87, 1969. Miyamoto M, Terayama H: Nature of rat liver cell sap factors inhibiting the DNA synthesis in tumor cells. Biochim Biophys Acta 228:324-330, 1971. Otsuka H: Difference of the inhibitor of DNA synthesis in liver extract from liver arginase. Cancer Res 29:265-266, 1969. McMahon JB, Iype PT: Specific inhibition of proliferation of non-malignant rat hepatic cells by a factor from rat liver. Cancer Res 40:1249-1254, 1980. Osunkoya BC, Adler WH, Smith RT: Effect of arginine deficiency on synthesis of DNA and immunoglobulin receptor of Burkitt lymphoma cells. Nature 227:398-400, 1970. Storr JM, Burton AF: The effects of arginine deficiency on lymphoma cells. Br J Cancer 30:50-59, 1974. Koji T, Terayama H: Arginase as one of the inhibitory principles in the density-dependent as well as plasma membrane-mediated inhibition of liver cell growth in vitro. Experimental Cell Research 155:359-370, 1984. Barra R, Parsons J, Koch MR, Lea MA: Soluble factors from liver and hepatomas which inhibit [3H]-thymidine incorporation into DNA of Novikoff hepatoma cells. Cancer Res 39:1655-1660, 1979. Miyamoto M, Terayama H: Nature of rat liver cell sap factors inhibiting the DNA synthesis in tumor cells. Biochim Biophys Acta 228:324-330, 1971. Terayama H, Koji T, Kontani M, Okumoto T: Arginase is an inhibitory principle in liver plasma membranes arresting the growth of various mammalian cells in vitro. Biochim Biophys Acta 720:188-192, 1982. Savoca KV, Davis FF, Van Es T, et al: Cancer therapy with chemically modified enzyme: II. The therapeutic effectiveness of arginase modified by the covalent attachment of polyethylene glycol, on a Taper liver tumor and the L5178Y murine leukaemia. Cancer Biochem Biophys 7:261-268, 1984. Scott LA, Lamb JL, Smith S, Wheatley DN: Single amino, arginine, deprivation: rapid and selective cells death of cultured transformed and malignant cells. Brit J Cancer 83:800-810, 2000. Snodgrass PJ, Lin RC: Differing effects of arginine deficiency on the urea cycle enzymes of rat liver, cultured hepatocytes and hepatoma cells. J Nutr 117:1827-1837, 1987. Hollta KG, Pohjanpelto P: Polyamine dependence of Chinese hamster ovary cells in serum-free culture is due to deficient arginase activity. Biochim Biophys Acta 721:321-327, 1982. Anehus S, Pohjanpelto P, Baldetrop B, et al. Polyamine starvation prolongs the S and G2 phases pf polyamine-dependent, arginase-deficient, CHO cells. Mol Cell Biol 4:915-922, 1984. Finlay IG, Seifert JK, Stuart GJ, Morris DL: Resection with cryotherapy of colorectal hepatic metastasis has the same survival as hepatic resection alone. Eur J Surg Oncol 26:199-202, 1998. Pohjanpelto P, Hollta E: Arginase activity of different cells in tissue culture. Biochem Biophys Acta 757:191-195, 1983. Leu S-Y, Wang S-R: Clinical significance of arginase in colorectal cancer. Cancer 70:733-736, 1992. Straus B, Cepelak I, Festa G: Arginase: a new marker for mammary carcinoma. Clin Chem Acta 210:5-12, 1992. Dvorak HF: Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 315:1650-1659, 1986. Barbul A, Wasserkrug HL, Seifter E, et al: Immunostimulatory effects of arginine in normal and injured rats. J Surg Res 29:228-235, 1980. Barile MF, Levinthan BG: Possible mechanism of Mycoplasm inhibition of lymphocyte transformation induced by phytohaemagglutinin. Nature 219:751-753, 1968. Kraemer PM, Defendi V, Hayflick L, Manson LA: Mycoplasma, PPLO, strains with lytic activity for murine lymphoma cells in vitro. Proc Soc Exp Biol Med 112:381-387, 1963. Gill P, Pan J: Inhibition of cell division in L5178Y cells by arginine-degrading mycoplasma: the role of arginine deiminase. Can J Microbiol 16:415-419, 1970. Miyazaki K, Takaku H, Umeda M, et al: Potent growth inhibition of human tumor cells in culture by arginine deiminase purified from a culture medium of a mycoplasma-infected cell line. Cancer Res 50:4522-4527, 1990. Sugimura K, Ohno T, Kusuyama T, Azuma I: High sensitivity of human melanoma cell lines to the growth inhibitory activity of arginine deiminase in vitro. Melanoma Res 2:190-196,1992. Takaku H, Takase M, Abe SI, et al: In vivo anti-tumor activity of arginine deiminase purified from Mycoplasma argini. Int J Cancer 51:244-249,1992. Claesson MH, Tscherning T, Nissen MH, Lind K: Inhibitory effect of mycoplasma-released arginase. Activity in mixedlymphocyte and tumor cell cultures. Scand J Immunol 32:623- 630, 1990. Komada Y, Zhang XL, Zhou YW, et al: Apoptotic cell death of human T lymphoblastoid cells induced by arginine deiminase. Int J Hematol 65:129-141, 1997. Gong H, Zolzer F, von Recklinghausen G, et al: Arginine deiminase inhibits cell proliferation by arresting cell cycle and inducing apoptosis. Biochem Biophys Res Comm 261:10-14, 1999. Gong H, Zolzer F, von Recklinghausen G, et al: Arginine deiminase inhibits proliferation of human leukaemia cells more potently than asparginase by inducing cell cycle arrest and apoptosis. Leukemia 14:826-829, 2000. Muller HJ, Boos J: Use of L-asparaginase in childhood ALL. Crit Rev Oncol Hematol 28:97-113, 1998. Schimke RT: Enzymes of arginine metabolism in mammalian cell culture. 1. Repression of argininosuccinate synthease and argininosuccinase. J Biol Chem 239:136-145, 1964. Blethen SL, Boeker EA, Snell EE: Arginine decarboxylase from Escherichia coli. J Biol Chem 243:1671-1677, 1968. Wu WH, Morris DR: Biosynthetic arginine decarboxylase from Escherichia coli. J Biol Chem 248:1687-1695, 1973. Morrissey J, McCracken R, Ishidoya S, et al: Partial cloning and characterization of an arginine decarboxylase in the kidney. Kidney Internat 47:1458-1461, 1995. Buch JK, Boyle SM: Biosynthetic arginine decarboxylase in Escherichia coli is synthesized as a precursor and located in the cell envelope. J Bacteriol 163:522-527, 1985. Galea E, Regunathan S, Eliopoulos V, et al: Inhibition of mammalian nitric oxide synthases by agmatine, an endogenous polyamine formed by decarboxylation of arginine. Biochemical J 316:247-249, 1996. Wheatley DN: Dietary restriction, amino acid deprivation, and cancer. The Cancer J 11:183-189, 1998. Joshi M: The importance of L-arginine metabolism in melanoma: an hypothesis for the role of nitric oxide and polyamines in tumor angiogenesis. Free Radic Biol Med 22:573-578, 1997. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 18 EP 25 PG 8 ER PT J AU Park, SH Kim, H Song, BJ AF Park, Sung-Hye Kim, Hanseong Song, Byung-Joo TI Down Regulation of Bcl2 Expression in Invasive Ductal Carcinomas Is Both Estrogen- and Progesterone-Receptor Dependent and Associated with Poor Prognostic Factors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bcl2; estrogen receptor; invasive ductal carcinoma; ductal carcinoma in situ; progesterone receptor ID Bcl2; estrogen receptor; invasive ductal carcinoma; ductal carcinoma in situ; progesterone receptor AB In normal breast, Bcl2 is expressed in the non-pregnant and non-involuting mammary epithelium. The exact mechanism and the effect of the down regulation of the Bcl2 expression on breast cancer cells are not clearly defined. We compared down regulation as well as the persistent expression of Bcl2 with ER, PR, p53, and c-erb-B2 overexpression and clinicopathologic variables, and tumor stage in 11 cases of ductal carcinomas in situ (DCIS) and 44 cases of invasive ductal carcinomas (IDC) of Korean women by immunohistochemical studies. Bcl2 down regulation was found in 39% of IDC and in 18% of DCIS cases. In IDC, while persistent Bcl2 expression was displayed in 95% and 78.9% of ER and PR immunoreactive ones and 71.9 % of c-erb-B2 immnonegative ones. Seventeen cases of Bcl2 down regulated IDC had a significant correlation with ER negativity (94.1%), PR negativity, (76.5%), and high nuclear (61.1% is grade III) and histological grade (76% is grade III). However, in DCIS, no significant correlation between the Bcl2 expression and various parameters were obtained, probably due to small sample size. In conclusion, the Bcl2 expression was both ER and PR dependent and down regulation of Bcl2 in IDC was significantly correlated with poor prognostic factors. C1 [Park, Sung-Hye] Inje University, College of Medicine, Ilsan Paik Hospital, Department of Anatomical Pathology, 2240 Daewha-dong, Ilsan-gu, 411-706 Koyang-city, Kyunggi-do, South Korea. [Kim, Hanseong] Inje University, College of Medicine, Ilsan Paik Hospital, Department of Anatomical Pathology, 2240 Daewha-dong, Ilsan-gu, 411-706 Koyang-city, Kyunggi-do, South Korea. [Song, Byung-Joo] Inje University, College of Medicine, Ilsan Paik Hospital, Department of General SurgeryKyunggi-do, South Korea. RP Park, SH (reprint author), Inje University, College of Medicine, Ilsan Paik Hospital, Department of Anatomical Pathology, 411-706 Koyang-city, South Korea. EM sunghye@ilsanpaik.ac.kr CR Strasser A, Huang DC, Vaux DL: The role of the bcl-2/ced-9 gene family in cancer and general implications of defects in cell death control for tumourigenesis and resistance to chemotherapy. Biochim Biophys Acta 1333:F151-178, 1997. Hockenbery DM, Zutter M, Hickey W, et al: BCL2 protein is topographically restricted in tissues characterized by apoptotic cell death. Proc Natl Acad Sci 88:6961-6965, 1991. Chao DT, Korsmeyer SJ:BCL-2 family: regulators of cell death. Annu Rev Immunology 16:395-419, 1998. Bonnotte B, Favre N, Moutet M, et al: Bcl-2-mediated inhibition of apoptosis prevents immunogenicity and restores tumorigenicity of spontaneously regressive tumors. J Immunol 161:1433-1438, 1998. Gross A, McDonnell JM, Korsmeyer SJ: BCL-2 family members and the mitochondria in apoptosis. Genes Dev 13:1899- 1911, 1999. Knowlton K, Mancini M, Creason S, et al: Bcl-2 slows in vitro breast cancer growth despite its antiapoptotic effect. J Surg Res 76:22-26, 1998. Kumar R, Vadlamudi RK, Adam L: Apoptosis in mammary gland and cancer. Endocr Relat Cancer 7:257-69, 2000. Hickman JA: Apoptosis and chemotherapy resistance. Eur J Cancer 32A:921-926, 1996. Jansen B, Wacheck V, Heere-Ress E, et al: Chemosensitisation of malignant melanoma by BCL2 antisense therapy. Lancet 356:1728-1733, 2000. Rehman S, Crow J, Revell PA: Bax protein expression in DCIS of the breast in relation to invasive ductal carcinoma and other molecular markers. Pathol Oncol Res 6:256-263, 2000. Castiglione F, Sarotto I, Fontana V, et al.: Bcl2, p53 and clinical outcome in a series of 138 operable breast cancer patients. Anticancer Res 19:4555-4563, 1999. Mauri FA, Maisonneuve P, Caffo O, et al: Prognostic value of estrogen receptor status can be improved by combined evaluation of p53, Bcl2 and PgR expression: an immunohistochemical study on breast carcinoma with long-term follow-up. Int J Oncol 15:1137-1147, 1999. Olopade OI, Adeyanju MO, Safa AR, et al: Overexpression of BCL-x protein in primary breast cancer is associated with high tumor grade and nodal metastases. Cancer J Sci Am 3:230-237, 1997. Gasparini G, Barbareschi M, Doglioni C, et al: Expression of bcl- 2 protein predicts efficacy of adjuvant treatments in operable node-positive breast cancer. Clin Cancer Res 1:189-198, 1995. Silvestrini R, Veneroni S, Daidone MG, et al.: The Bcl-2 protein: a prognostic indicator strongly related to p53 protein in lymph node-negative breast cancer patients. J Natl Cancer Inst 86: 499-504, 1994. Bhargava V, Kell DL, van de Rijn M, et al: Bcl-2 immunoreactivity in breast carcinoma correlates with hormone receptor positivity. Am J Pathol 145:535-540, 1994. Bronner MP, Culin C, Reed JC, et al:The bcl-2 proto-oncogene and the gastrointestinal epithelial tumor progression model. Am J Pathol 146:20-26, 1995. Teixeira C, Reed JC, Pratt MA: Estrogen promotes chemotherapeutic drug resistance by a mechanism involving Bcl-2 protooncogene expression in human breast cancer cells. Cancer Res 55:3902-3907, 1995. Luna-More S, Casquero S, Perez-Mellado A, et al: Importance of estrogen receptors for the behavior of invasive micropapillary carcinoma of the breast. Review of 68 cases with followup of 54. Pathol Res Pract 196:35-39, 2000. Leal C, Henrique R, Monteiro P, et al: Apocrine ductal carcinoma in situ of the breast: histologic classification and expression of biologic markers. Hum Pathol 32:487-493, 2001. Sumantran VN, Ealovega MW, Nunez G, et al: Overexpression of Bcl-XS sensitizes MCF-7 cells to chemotherapy-induced apoptosis. Cancer Res 55:2507-2510, 1995. Leung LK, Wang TT: Differential effects of chemotherapeutic agents on the Bcl-2/Bax apoptosis pathway in human breast cancer cell line MCF-7. Breast Cancer Res Treat 55:73-83, 1999. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 26 EP 30 PG 5 ER PT J AU Hong, A Lee, S AF Hong, Angela Lee, C. Soon TI Kaposi's Sarcoma: Clinico-pathological Analysis of Human Immunodeficiency Virus (HIV) and Non-HIV Associated Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Kaposi’s sarcoma; human immunodeficiency virus; HIV; neoplasia; human herpesvirus 8; HHV8 ID Kaposi’s sarcoma; human immunodeficiency virus; HIV; neoplasia; human herpesvirus 8; HHV8 AB Kaposi's sarcoma (KS) is a angioformative lesion that classically occurs in elderly Eastern European and Mediterranean males but is also common in immunosuppressed individuals particularly human immunodeficiency virus (HIV)-infected patients. This study investigates the clinical and histopathological features of 47 patients with Kaposi’s sarcoma from a teaching hospital in Sydney, Australia, in which 44 cases had adequate clinical follow-up information over a 10-year period. Most of the lesions were of late stage (37/47 cases; 79%), consisting of 11 cases of plaque stage KS and 26 cases of nodular stage KS with only 10 cases of early or patch stage KS. The majority of the HIV-positive cases (23/33; 70%) and all of the HIV-negative (14/14; 100%) cases had late stage lesions (p=0.020; X 2 -test). The histopathological features that were more common in the KS lesions of HIV-negative patients were lesional cell mitosis (p=0.0002), single cell necrosis (p=0.001), apoptosis (p=0.0001) and single cell anaplasia (p=0.0001). The KS lesions in HIV-positive patients tended to have dissecting blood vessels (14/33 cases; 42%) unlike those seen in HIV-negative patients (0/14 cases; 0%) (p=0.004). Most HIV-positive cases (30/33; 90%) were males (p=0.0068); and all these patients (33/33 cases; 100%) were <60 years old, in contrast to HIV-negative patients (1/11 cases; 9%) (p=0.0001). HIV status does not affect the occurrence of multiplicity of KS lesions. However, extracutaneous or visceral KS lesions were more likely to occur in HIV-positive patients (p=0.027). The number of cases of histologically proven KS cases has decreased markedly over the recent 5 year period of 1995-1999 (n=14), which was less than half of the number of the preceding 5 year period, 1990-1994 (n=33). In summary, there are distinct differences in the clinical and histopathological features of Kaposi’s sarcoma lesions in HIV-positive and HIV-negative patients. Despite the recent discovery of the HHV8 virus as the initiating and promoting factor of most of the KS lesions, these differences indicated that there might be different mechanisms that occur in HIV-positive and HIV-negative patients in the development of this lesion. C1 [Hong, Angela] Royal Prince Alfred Hospital, Department of Radiation Oncology, Mis-senden Road, Camperdown, 2050 New South Wales, Australia. [Lee, C. Soon] Royal Prince Alfred Hospital,, Department of Anatomical Pathology, Mis-senden Road, Camperdown, 2050 New South Wales, Australia. RP Lee, S (reprint author), Royal Prince Alfred Hospital,, Department of Anatomical Pathology, 2050 New South Wales, Australia. EM slee@csls.rpa.cs.nsw.gov.au CR Friedman-Birnbaum R, Bergman R, Bitterman-Deutsch O, et al: Classic and iatrogenic Kaposi’s sarcoma. Histopathological patterns as related to clinical course. Am J Dermatopathol 15:523-527, 1993. Santucci M, Pimpinelli N, Moretti S, Giannotti B: Classic and immunodeficiency-associated Kaposi’s sarcoma. Clinical, histologic, and immunologic correlations. Arch Pathol Lab Med 112:1214-1220, 1988. Fauci AS, Macher AM, Longo DL, et al: NIH conference. Acquired immunodeficiency syndrome: epidemiologic, clinical, immunologic, and therapeutic considerations. Ann Intern Med 100:92-106, 1984. Antman K, Chang Y: Kaposi’s sarcoma. N Engl J Med 342:1027-1038, 2000. Chor PJ, Santa Cruz DJ: Kaposi’s sarcoma. A clinicopathologic review and differential diagnosis. J Cutan Pathol 19:6-20, 1992. Blumenfeld W, Egbert BM, Sagebiel RW: Differential diagnosis of Kaposi’s sarcoma. Arch Pathol Lab Med 109:123-127, 1985. Templeton AC: Kaposi’s sarcoma. Pathol Annu 16:315-336, 1981. DiGiovanna JJ, Safai B: Kaposi’s sarcoma. Retrospective study of 90 cases with particular emphasis on the familial occurrence, ethnic background and prevalence of other diseases. Am J Med 71:779-783, 1981. Gange RW, Jones EW: Kaposi’s sarcoma and immunosuppressive therapy: an appraisal. Clin Exp Dermatol 3:135-146, 1978. Gupta AK, Cardella CJ, Haberman HF: Cutaneous malignant neoplasms in patients with renal transplants. Arch Dermatol 122:1288-1293, 1986. Hoshaw RA, Schwartz RA: Kaposi’s sarcoma after immunosuppressive therapy with prednisone. Arch Dermatol 116:1280- 1282, 1980. Klepp O, Dahl O, Stenwig JT: Association of Kaposi’s sarcoma and prior immunosuppressive therapy: a 5-year material of Kaposi’s sarcoma in Norway. Cancer 42:2626-2630, 1978. Lutzner MA: Skin cancer in immunosuppressed organ transplant recipients. J Am Acad Dermatol 11:891-893, 1984. Penn I: Kaposi’s sarcoma in organ transplant recipients: report of 20 cases. Transplantation 27:8-11, 1979. Safai B, Johnson KG, Myskowski PL, et al. The natural history of Kaposi’s sarcoma in the acquired immunodeficiency syndrome. Ann Intern Med 103:744-750, 1985. Safai B, Sarngadharan MG, Koziner B, et al. Spectrum of Kaposi’s sarcoma in the epidemic of AIDS. Cancer Res 45:4646s-4648s, 1985. Gottlieb GJ, Ackerman AB: Kaposi’s sarcoma: an extensively disseminated form in young homosexual men. Hum Pathol 13:882-892, 1982. Francis ND, Parkin JM, Weber J, Boylston AW: Kaposi’s sarcoma in acquired immune deficiency syndrome, AIDS). J Clin Pathol 39:469-474, 1986. Cox FH, Helwig EB: Kaposi’s sarcoma. Cancer 12:289-298, 1959. Reynolds WA WR, Soule EH: Kaposi’s sarcoma: a clinicopathologic study with particular reference to its relationship to the reticuloendothelial system. Medicine 44:419-443, 1965. Hong A, Lee CS: The emerging role of the human herpesvirus 8, HHV8, in human neoplasia. Pathology 33:460-467, 2001. Moore PS. The emergence of Kaposi’s sarcoma-associated herpesvirus, human herpesvirus 8). N Engl J Med 343:1411-3, 2000. Kedes DH, Ganem D, Ameli N, et al: The prevalence of serum antibody to human herpesvirus 8, Kaposi sarcoma-associated herpesvirus, among HIV-seropositive and high-risk HIVseronegative women. JAMA: 478-481, 1997. Kedes DH, Operskalski E, Busch M, et al: The seroepidemiology of human herpesvirus 8, Kaposi’s sarcoma-associated herpesvirus): distribution of infection in KS risk groups and evidence for sexual transmission. Nat Med 2: 18-924, 1996. Martin JN, Ganem DE, Osmond DH, et al: Sexual transmission and the natural history of human herpesvirus 8 infection. N Engl J Med 1998; 338:948-954, 1998. Prakash O, Tang ZY, He Y, et al: Human Kaposi’s sarcoma cellmediated tumorgenesis in human immunodeficiency type 1 Tatexpressing transgenic mice. J Natl Cancer Inst 92:721-728, 2000. Weiss R, Boshoff C: Addressing controversies over Kaposi’s sarcoma. J Natl Cancer Inst 92:677-678, 2000. Modlin RL, Crissey JT, Rea TH: Kaposi’s sarcoma. Int J Dermatol 22:443-448, 1983. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 31 EP 35 PG 5 ER PT J AU Akisik, E Bavbek, S Dalay, N AF Akisik, Elif Bavbek, Sevil Dalay, Nejat TI CD44 Variant Exons in Leukemia and Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD44; RT-PCR; leukemia; lymphoma ID CD44; RT-PCR; leukemia; lymphoma AB CD44 is a cell surface glycoprotein expressed on different cell types that functions in lymphocyte activation and homing, extracellular matrix adhesion and cellular migration. CD44 is encoded by a single gene composed of at least 20 exons. The standard CD44 protein (CD44S or CD44H) is the hematopoietic form of CD44 in lymphoid cells. Variant isoforms (designated from v1 to v10) are formed by addition of new exons to the extracellular domain. High levels of CD44v6 expression has been observed in some tumors and are associated with metastatic spread. The aim of the present study was to investigate and evaluate expression of the CD44v6 and v6-containing variants as a possible marker in chronic myeloid leukemia and lymphoma by reverse transcription-polymerase chain reaction. CD44 exon v6 was detected in all patients and all individuals in the control group. CD44v6-v10 mRNA was observed in 25 patients but in none of the subjects in the control group. CD44v6/v9-10, CD44v6-v7, CD44v6/v10 transcripts were detected in 11, 6, and 2 patients, respectively. CD44v6-7/v9-10 transcripts were not observed in either the patients or the healthy individuals. We conclude that CD44v6-v10 expression may be associated with hematologic malignancies. C1 [Akisik, Elif] Istanbul Medical Faculty, Istanbul University, Oncology Institute, Capa, 34390 Istanbul, Turkey. [Bavbek, Sevil] Istanbul Medical Faculty, Istanbul University, Oncology InstituteIstanbul, Turkey. [Dalay, Nejat] Istanbul Medical Faculty, Istanbul University, Oncology Institute, Capa, 34390 Istanbul, Turkey. RP Dalay, N (reprint author), Istanbul Medical Faculty, Istanbul University, Oncology Institute, 34390 Istanbul, Turkey. EM ndalay@istanbul.edu.tr CR Arch R, Wirth K, Hofmann M, et al: Participation in normal immune response of a metastasis-inducing splice variant of CD44. Science 257:682-685, 1992. Bartolazzi A, Jackson D, Bennett K, et al: Regulation of growth and dissemination of human lymphoma by CD44 splice variants. J Cell Sci 108:1723-1733, 1995. Bolodeoku J: PCR analysis of CD44 variants in tumors. Clin Appl PCR 16:189-202, 1998. Borland G, Ross JA, Guy K: Forms and functions of CD44. Immunology 9:139-148, 1998. Chomczynski P, Sacchi N: Single-step method of RNA isolation by guanidium thiocyanate-phenol-chloroform extraction. Anal Biochem 162:156-159, 1987. Dall P, Heider KH, Sinn HP, et al: Comparison of immunohistochemistry and RT-PCR for detection of CD44v expression, a new prognostic factor in human breast cancer. Int J Cancer 60:471-477, 1995. Dammrich J, Vollmers HP, Heider KH, et al: Importance of different CD44v6 expression in human gastric intestinal and diffuse type cancers for metastatic lymphogenic spreading. J Mol Med 73:395-401, 1995. Finke Lh, Zorb C, Haensch W, et al: Expression of CD44 isoforms: a new histopathologic parameter in colorectal carcinoma ? Zbl Chir 121:450-454, 1996. Fox SB, Fawcett J, Jackson D, et al: Normal human tissues, in addition to some tumors, express multiple different CD44 isoforms. Cancer Res 54:4539-4546, 1994. Fukuse T, Hirata T, Naiki H, et al: Expression of proliferating cell nuclear antigen and CD44 variant isoforms in the primary and metastatic sites of non-small cell lung carcinoma with intrapulmonary metastasis. Cancer 86:1174-1181, 1999. Gotley DC, Fawcett J, Walsh MD, et al: Alternatively spliced variants of the cell adhesion molecule CD44 and tumor progression in colorectal cancer. Br J Cancer 74:342-351, 1996. Guriec N, Gairard B, Marcellin L, et al: CD44 isoforms with exon v6 and metastasis of primary N0M0 breast carcinomas. Breast Cancer Res Treat 44:261-268, 1997. Gunthert U, Hofmann M, Rudy W, et al: A new variant of the glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell 65:13-24, 1991. Harn HJ, Ho LI, Chang JY, et al: Differential expression of the human metastasis adhesion molecule CD44 v in normal and carcinomatous stomach mucosa of the Chinese subjects. Cancer 75:1065-1071, 1995. Haynes BF, Telen MJ, Hale LP, et al: CD44; A molecule involved in leucocyte adherence and T Cell activation. Immunol Today 10:423-428, 1989. Heider KH, Hofmann M, Hors E, et al: A human homologue of the rat metastasis – associated variant of CD44 is expressed in colorectal carcinomas and adenomateous polyps. J Cell Biol 120:227-233, 1993. Herrlich P, Pals S, Ponta H: CD44 in colon cancer. Eur J Cancer 31A, 1110-1112, 1995. Herrlich P, Zoller M, Pals S, et a : CD44 splice variants : metastases meet lymphocytes. Immunol Today 14:395-399, 1993. Hirata F, Fukuse T, Naiki H, et al: Expression of CD44 variant exon 6 in stage I non-small cell lung carcinoma as a prognostic factor. Cancer Res 58:1108-1110, 1998. Hofmann M, Rudy W, Zoller M, et al: CD44 splice variants confer metastatic behaviour in rats: homologuous sequences are expressed in human tumor cell lines. Cancer Res 51:4292-5297, 1991. Imazeki F, yokosuka O, Yamaguchi T, et al: Expression of variant CD44 messenger RNA in colorectal adenocarcinomas and adeonomatous polyps in humans. Gastroenterology 110:362- 368, 1996. Khaldoyanidi S, Achtnich M, Hehlmann R, et al: Expression of CD44 variant isoforms in peripheral blood leukocytes in malignant lymphoma and leukemia: inverse correlation between expression and tumor progression. Leuk Res 20:839-851, 1996. Koopman G, Heider KH, Horst E, et al: Activated human lymphocytes and aggressive Non-Hodgkin’s lymphomas express a homologue of the rat metastatic-associated variant of CD44. J Exp Med 177:897-904, 1993. Lesley J, Hyman R: CD44 structure and function. Front Biosci 3:616-630, 1998. Mackay CR, Terpe HJ, Stauder R, et al: Expression and modulation of CD44 variant isoforms in humans. J Cell Biol 124:71- 82, 1994. Matsumura Y, Tarin D: Significance of CD44 gene products for cancer diagnosis and disease evaluation. Lancet 340:1053- 1058, 1992. Miwa T, Watanabe A, Yamada Y, et al: Progression in gastric carcinoma relative to the ratio of CD44 epithelial variant transcript to CD44 hematopoietic variant transcript. Cancer 77:25-29, 1996. Mulder JWR, Kruyt PM, Sewnath M, et al: Colorectal cancer prognosis and expression of exon-v6-containing CD44 proteins. Lancet 344:1470-1472, 1994. Okamoto I, Morisaki T, Sasaki J, et al: Molecular detection of cancer cells by competitive reverse-transcription polymerase chain reaction analysis of specific CD44 variant mRNAs. J Natl Cancer Inst 90:307-315, 1998. Orzechowski HD, Beckenbach C, Herbs H, et al: Expression of CD44v6 is associated with cellular dyspleasia in colorectal epithelial cells. Eur J Cancer 31A, 2073-2079, 1995. Ponta H, Herrlich P: The CD44 protein family: Roles in embryogenesis and tumor progression. Front Biosci 3:650-656, 1998. Ristamaki R, Joensuu H, Hagberg H, et al: Clinical significance of circulating CD44 in non-Hodgkin’s lymphoma. Int J Cancer 79:221-225, 1998. Ristamaki R, Joensuu H, Jalkanen S: Serum CD44 in non- Hodgkin’s lymphoma. Leuk Lymphoma 33:433-440, 1999. Ristamaki R, Joensuu H, Salmi M et al: Serum CD44 in malignant lymphoma: an association with treatment response. Blood 84:238-243, 1994. Ristamaki R, Joensuu H, Soderstrom Ko, et al: CD44v6 expression in non-Hodgkin’s lymphoma: an association with low histological grade and poor prognosis. J Pathol 176:259-267, 1995. Rodriguez C, Monges G, Rouanet P, et al: CD44 expression patterns in breast and colon tumors: a PCR-based study of splice variants. Int J Cancer 64:347-354, 1995. Rudy W, Hofmann M, Schwartz-Albiez R, et al: The two major CD44 proteins expressed on a metastatic rat tumor cell line are derived from different splice variants: each one individually suffices to confer metastatic behaviour. Cancer Res 53:1262- 1268, 1993. Salmi M, Gron-Virta K, Sointu P, et al: Regulated expression of CD44v6 containing isoforms of CD44 in man : downregulation during malignant transformation of tumors of squamocellular origin. J Cell Biol 122:431-442, 1993. Schumacher U, Horny HP, Horst HA, et al: A CD44 variant exon 6 epitope as a prognostic indicator in breast cancer. Eur J Surg Oncol 22:259-261, 1996. Seiter S, Arch R, Reber S, et al: Prevention of tumor metastasis formation by anti-variant CD44. J Exp Med 177:443-455, 1993. Shermann L, Sleeman J, Dall P, et al: The CD44 proteins in embryonic development and in cancer. Curr Top Microbiol Immunol 213:249-269, 1996. Sleeman J, Moll J, Shermann L, et al : The role of CD44 splice variants in human metastatic cancer. Ciba Found Symp 189:142-156, 1995. Stauder R, Eisterer W, Thaler J, et al: CD44 variant isoforms in Non-Hodgkin’s lymphomas: A new independent prognostic factor. Blood 85:2885-2899, 1995. Stauder R, Van Driel M, Schwarzler C, et al: Different CD44 splicing patterns define prognostic subgroups in multiple myeloma. Blood 88:3101-3108, 1996. Sy MS, Mori H, Liu D: CD44 as a marker in human cancers. Curr Opin Oncol 9:108-112, 1997. Tran TA, Kallakury BV, Sheehan CE, et al: Expression of CD44 standard form and variant isoforms in non-small cell lung carcinomas. Hum Pathol 28:804-814, 1997. Underhill C: CD44: The hyaluronan receptor. J Cell Sci 103:293-298, 1992. Wimmel A, Kogan E, Ramaswamy A, et al: Variant expression of CD44 in preneoplastic lesions of the lung. Cancer 92:1231- 1236, 2001. Woodman AC, Sugiyama M, Yoshida K, et al: Analysis of anomalous CD44 gene expression in human breast, bladder and colon cancer and correlation of observed mRNA and protein isoforms. Am J Pathol 149:1519-1530, 1996. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 36 EP 40 PG 5 ER PT J AU Nagy, ZsZ Toth, J Nagymihaly, A Suveges, I AF Nagy, Zsolt Zoltan Toth, Jeannette Nagymihaly, Attila Suveges, Ildiko TI The Role of Ultraviolet-B in Corneal Healing Following Excimer Laser in situ Keratomileusis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Laser in situ Keratomileusis (LASIK); Ultraviolet-B; corneal transparency; subepithelial haze; activated ker-atocytes; extracellular matrix ID Laser in situ Keratomileusis (LASIK); Ultraviolet-B; corneal transparency; subepithelial haze; activated ker-atocytes; extracellular matrix AB Corneal photoablation with the 193 nm argon fluorid excimer laser during photorefractive keratectomy (PRK) in high diopter range is frequently associated with subepithelial haze and consequent refractive regression due to avascular corneal wound healing. The wound healing response can be augmented by Ultraviolet-B (UV-B) exposure originating from sun or solarium. Clinically Laser in situ Keratomileusis (LASIK) even in high diopter range is associated with less subepithelial haze and regression than PRK. In an animal model, the morphologic changes of the rabbit cornea were evaluated following LASIK and secondary UV-B exposure. Light microsopic changes were found to be insignificant. Transmission electron microscopy (TEM) normal epithelium, epithelial adhesion structures and normal anterior stroma showed in the LASIK treated UV-B irradiated rabbit eyes. Around the peripheral LASIK cut, migrating keratocytes with pseudopodia were observed. Under the flap (160 µm depth) the overall stromal collagen structure was normal, some activated keratocytes and mild extracellular matrix formation within and around keratocytes were noted. Within activated keratocytes TEM showed prominent rough endoplasmic reticulum, Golgi apparatus, mitochondria and extracellular vacuoles, which showed resolution with time. These changes were much milder than in PRK treated-UV-B irradiated eyes. Secondary UV-B caused no long-term disturbance in corneal transparency in LASIK and UV-B treated rabbit eyes. C1 [Nagy, Zsolt Zoltan] Semmelweis University, Department of Ophthalmology, Tomo u. 25-29, 1083 Budapest, Hungary. [Toth, Jeannette] Semmelweis University, Department of Ophthalmology, Tomo u. 25-29, 1083 Budapest, Hungary. [Nagymihaly, Attila] Semmelweis University, Department of Ophthalmology, Tomo u. 25-29, 1083 Budapest, Hungary. [Suveges, Ildiko] Semmelweis University, Department of Ophthalmology, Tomo u. 25-29, 1083 Budapest, Hungary. RP Nagy, ZsZ (reprint author), Semmelweis University, Department of Ophthalmology, 1083 Budapest, Hungary. CR Seiler T, Kriegerowski M, Kahle G, et al: Excimer laser keratomileusis for myopia correction. Results and complications. Klin Monatsbl Augenheilk 199:153-159, 1999. Seiler T, Holschbach A, Derse M, et al: Complications of myopic photorefractive keratectomy with the excimer laser. Ophthalmology 101:153-160, 1994. Tsubota K, Toda I, Itoh S: Reduction of subepithelial haze after photorefractive keratectomy by cooling the cornea. Am J Ophthalmol 115:820-821, 1993. Nagy ZZ, Nemeth J, Suveges I, et al: Examination of subepithelial haze after photorefractive keratectomy with the ultrasound biomicroscope. Klin Monatsbl Augenheilk 209:283-285, 1996. Durrie DS, Lesher MP, Cavanaugh TB: Classification of variable clinical response after photorefractive keratectomy for myopia. J Cataract Refract Surg 11:341-347, 1995. Grimm B, Waring Go III, Ibrahim O: Regional variation in corneal topography and wound healing following photorefractive keratectomy. J Refract Surg 11:348-357, 1995. Siganos DS, Katsanevaki VJ, Pallikaris IG: Correlation of subepithelial haze and refractive regression 1 month after photorefractive keratectomy. J Refract Surg 15:338-342, 1999. Wachtlin J, Langenbeck K, Schrunder S, et al:Immunohistology of corneal wound healing after photorefractive keratectomy and laser in situ keratomileusis. J Refract Surg 15:451-458, 1999. Pallikaris IG, Siganos DS: Excimer laser in situ keratomileusis and photorefractive keratectomy for correction of high myopia. J Refract Corneal Surg 10:498-510, 1994. Nagy Z Z, Hiscott P, Seitz B, et al: Ultraviolet-B enhances stromal response to 193-nm excimer laser treatment. Ophthalmology 104:375-380, 1997. Knorz MC, Liermann A, Seiberth V, et al: Laser in situ keratomileusis to correct myopia of –6.00 to –29.00 diopters. J Refract Surg 12:575-584, 1996. Marinho A, Pinto MC, Pinto R, et al: LASIK for high myopia: one year experience. Ophthalmic Surg Lasers 1996;27:S517-520. Kim HM, Jung HR: Laser assisted in situ keratomileusis for high myopia. Ophthalmic Surg Lasers 1996;27:S508-511. Hersh PS, Brint SF, Maloney RK, et al: Photorefractive keratectomy versus laser in situ keratomileusis for moderate to high myopia. A randomized prospective study. Ophthalmology 105:1512-1522, 1998. Knorz MC, Wiesinger B, Liermann A, et al: Laser in situ keratomileusis for moderate and high myopia and myopic astigmatism. Ophthalmology 105:932-940, 1998. McGhee CN, Craig JP, Sachdev N, et al: Functional, psychological and satisfaction outcomes of laser in situ keratomileusis for high myopia. J Cataract Refarct Surg 26:497-509, 2000. Hanna KD, Pouliquen YM, Waring GO III, et al: Corneal wound healing in monkeys after repeated excimer laser photorefractive keratectomy. Arch Ophthalmol 110:1286-1291, 1992. Chayet AS, Assil KK, Montes M, et al: Regression and its mechanism after laser in situ keratomileusis in moderate and high myopia. Ophthalmology 105:1194-1199, 1998. Maldonado MJ, Ruiz-Oblitas L, Munuera JM, et al: Optical coherence tomography evaluation of the corenal cap and stromal bed features after laser in situ keratomileusis for high myopia and astigmatism. Ophthalmology 107:81-87, 2000. Wang Z, Chen J, Yang B: Comparision of laser in situ keratomileusis and photorefractive keratectomy to correct myopia from -1.25 to -6.0 diopters. J Refract Surg 13:528-534, 1997. Corbett MC, O’Brart DP, Warburton FG, Marshall J: Biologic and environmental risk factors for regression after photorefractive keratectomy. Ophthalmology 103:1381-1391, 1996. Fagerholm P: Wound healing after photorefractive keratectomy. J Cataract Refract Surg 26:432-447, 2000. Fagerholm P, Hamberg-Nystrom H, Tengroth B: Wound healing and myopic regression following photorefractive keratectomy. Acta Ophthalmol Copenh 72:229-234, 1994. Weber BA, Fagerholm P, Johansson B: Colocalization of hyaluronan and water in rabbit corneas after photorefractive keratectomy by specific staining for hyaluronan and by quantitative microradiography. Cornea 16:560-563, 1997. Helena MC, Baerveldt F, Kim WJ, Wilson SE: Keratocyte apoptosis after corneal surgery. Invest Ophthalmol Vis Sci 39:276- 283, 1998. Wilson SE: Keratocyte apoptosis in refractive surgery. Everett Kinsey Lecture. CLAO 24:181-185, 1998. Podskochy A, Fagerholm P: Cellular response and reactive hyaluronan production in UV-exposed rabbit corneas. Cornea 17:640-645, 1998. Podskochy A, Gan L, Fagerholm P: Apoptosis in UV-exposed rabbit corneas. Cornea 2000. 19:99-103. Fitzsimmons TD, Molander N, Stenevi U, et al: Endogenous hyaluronan in corneal disease. Invest Ophthalmol Vis Sci 35:2774-2782, 1994. Vinciguerra P, Azzolini M, Radice P, et al: A method for examining surface and interface irregularities after photorefractive keratectomy and laser in situ keratomileusis: a predictor of optical and functional outcomes. J Refract Surg 14 S204-206, 1998. Amm M, Wetzel W, Winter M, et al: Histopathological comparision of photorefractive keratectomy and laser in situ keratomileusis. J Refract Surg 12:758-766, 1996. Helena MC, Meisler D, Wilson SE: Epithelial growth within the lamellar interface after in situ keratomileusis, LASIK). Cornea 16:300-305, 1997. Wilson SE, Mohan RR, Mohan RR, et al: The corneal wound healing response: cytokine-mediated interaction of epithelium, stroma, and inflammatory cells. Progress Ret Eye Res 20:625- 637, 2001. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 41 EP 46 PG 6 ER PT J AU Strauss, L Fuenzalida, M Illanes, J Dabancens, A Diaz, E Lemus, D Guerrero, A AF Strauss, Laura Fuenzalida, Marcela Illanes, Julio Dabancens, Alfredo Diaz, Eugenia Lemus, David Guerrero, Anibal TI Effect of Sulfated Beta-cyclodextrin, a Water Soluble Cycloamylose, on the Promotion and/or Inhibition of Angiogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE angiogenesis; cancer; sulfated b-cyclodextrin ID angiogenesis; cancer; sulfated b-cyclodextrin AB Previous studies have reported that sulfated b b-cyclodextrin, a naturally occurring cycloamylose built up from six to eight glucopyranose units, when administered alone promotes angiogenesis, but administered with an angiostatic steroid inhibits angiogenesis in the cick embryo bioassay. In our experiments sulfated b b-cyclodextrin has been shown to posses many properties unrelated to its classical functions in the promotion and inhibition of angiogenesis that were not previously described. We studied the angiogenic and angiostatic properties of b b-cyclodextrin in a subcutaneosus plastic sponge model in mice. We realized two set of experiments. In each set mice were randomized into five groups (n= 5 mice). The first group was treated with sulfated b b-cyclodextrin (200 ng), the second group was treated with sulfated b b-cyclodextrin (2000 ng), the third group received unsubstituted b b-cyclodextrin (2000 ng), the fourth group was treated with sulfated b b-cyclodextrin (20000 ng) and the last group was used as a control group. In all groups compounds were administered intraperitonally 4 days after subcutaneous implantation of a sterile polyvinyl sponge on day 0, controls were not treated. Cyclodextrin administered alone at low drug concentration (200 ng) promoted angiogenesis and increased the development of venules in the sponge matrix. However, cyclodextrin administered at high drug concentration (2000 and 20 000 ng) reduced the vessel index in the sponge and areas of microhemorraghes were observed. From our results we propose that b b-cyclodextrin contains both a promoter and an inhibitor of angiogenesis and that the activation of both is drug concentration dependent. C1 [Strauss, Laura] Faculty of Medicine, University of Chile, Santiago de Chile, Department of Experimental Medicine and Department of Experimental Morphology, 60, rue de Longchamps, F-92200 Neuilly sur Seine, France. [Fuenzalida, Marcela] Faculty of Medicine, University of Chile, Santiago de Chile, Department of Experimental Medicine and Department of Experimental Morphology, 60, rue de Longchamps, F-92200 Neuilly sur Seine, France. [Illanes, Julio] Faculty of Medicine, University of Chile, Santiago de Chile, Department of Experimental Medicine and Department of Experimental Morphology, 60, rue de Longchamps, F-92200 Neuilly sur Seine, France. [Dabancens, Alfredo] Faculty of Medicine, University of Chile, Santiago de Chile, Department of Experimental Medicine and Department of Experimental Morphology, 60, rue de Longchamps, F-92200 Neuilly sur Seine, France. [Diaz, Eugenia] Faculty of Medicine, University of Chile, Santiago de Chile, Department of Experimental Medicine and Department of Experimental Morphology, 60, rue de Longchamps, F-92200 Neuilly sur Seine, France. [Lemus, David] Faculty of Medicine, University of Chile, Santiago de Chile, Department of Experimental Medicine and Department of Experimental Morphology, 60, rue de Longchamps, F-92200 Neuilly sur Seine, France. [Guerrero, Anibal] Faculty of Medicine, University of Chile, Santiago de Chile, Department of Experimental Medicine and Department of Experimental Morphology, 60, rue de Longchamps, F-92200 Neuilly sur Seine, France. RP Strauss, L (reprint author), Faculty of Medicine, University of Chile, Santiago de Chile, Department of Experimental Medicine and Department of Experimental Morphology, F-92200 Neuilly sur Seine, France. EM strauss_ewers@hotmail.com CR Bender ML, Kaniyama M: Cyclodextrin Chemistry. Springer Verlag, Berlin, 1978. Bergeron RJ: In Inclusion Compounds, Arwood JL, Davies JED, MacNicol DD, eds); Academic Press, New York, Vol. 3, pp 391-443, 1984. Bishop DK, Jutila MA, Sedmak DD, et al: Lymphocyte entry, NTO Inflammatory Tissues in vivo. Qualitative Differences of High Endothelial venule-like vessels in Sponge Matrix Allografts vs Isografts. J Immunol 142:4219-4224, 1989. Boyle EA, Mangan FR: The effect of a novel, non-steroidal anti-inflammatory compound, nabumetol, BRL14777, , on cellular infiltration into 24-hour polyvinyl sponge implants in the rat , compared with some steroidal and non-steroidal antiinflammatory drugs. J Pharm Pharmacol.34:570-575, 1982 Carmeliet P, Rakesh JK: Angiogenesis in cancer and other diseases. Nature 407:249-256, 2000. Dierendonck JH: A new method to detect apoptosis in poraffin sections: in situ end-labelling of fragmented DNA. J Histochem Cytochem 41, 7, 1993. Folkman J, Langer R, Linhardt RJ, et al: Angiogenesis inhibition and tumor regression caused by heparin or a heparin fragment in the presence of cortisone. Science 221:719-725, 1983. Folkman J, et al: Control of Angiogenesis with Synthetic Heparin Substitutes. Science 243:1490-1493, 1989. Frank SG, Kavaliunas DR: Investigation of the beta-cyclodextrin hydrocortisone inclusion compound. J Pharm Sci 72:1215, 1983. Freeman PC, Mangan FR, Wathius DK:Some effects of hydrocortisone on the early development of the rat cotton pellet granuloma. Biochem Pharmacol 29:372-373, 1979. Gavrieli Y, et al: Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. J Cell Biol 119: 493-501, 1992. Griffioen AW, Molena G: Angiogenesis: Potentials for Pharmacologic Intervention in the Treatment of Cancer, Cardiovascular Diseases, and Chronic Inflammation. Pharmacological Rev 52: 238-268, 2000. Guerrero A, et al: Actividad antineoplasica ejercida por quinacrina oral sobre tumors malignos transplantables en raton. Revista Chilena de Cancerologia 2:119-122, 1992. Koch AE, Halloran MM, Haskell C:Angiogenesis mediated by soluble forms of E-selectin and vascular cell adhesion molecule- 1. Nature 376:517-519, 1995. Lubec G: Interactions of N-CAM with heparin–like molecule. Nature 323: 743-474, 1986. Nangia–Makker P, Hanjo Y, et al: Galectin-3 Induces Endothelial Cell Morphogenesis and angiogenesis. Am J Pathol 156:899-909, 2000. Nguyen M, Stubel NA, Bischoff J: A role for sialyl Lewis–X/A glycoconjugates in capillary morphogenesis. Nature 365:267- 269, 1996. O’Reilly MS, et al: Angiostatin: A circulating endothelial cell inhibitor that suppresses angiogenesis and tumor growth. In: Cold Spring Harbor Symposia Quant Biol 59:471-482, 1994. Wijsman JA, Janker RR, Keijzer R, et al: A new method to detect apoptosis in paraffin sections : In situ end- labelling of fragmented DNA. J Histochem and Cytochem 41:7-12, 1993. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 47 EP 53 PG 7 ER PT J AU Patyanik, M Mayer, Polgar, I AF Patyanik, Mihaly Mayer, Arpad Polgar, Istvan TI Results of Ovary Tumor Treatment With Abdominally Administered 198Au Evaluated on the Basis of Long Term Follow Up SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ovary tumor; intraperitoneal therapy; radionuclide therapy ID ovary tumor; intraperitoneal therapy; radionuclide therapy AB In the period between 1959 and 1980 165 patients – previously operated with ovarian tumor – were treated by intraperitoneally administered 198Au in the Oncoradiological Centre of the Uzsoki Hospital. The stage distribution of the 158 patients with common epithelial histology was as it follows: Stage I/A 31; Stage I/B 9; Stage I/C 59; Stage II/A 19; Stage II/B 11; Stage II/C 7, Stage III/A 22. The five year survival result is the next: Stage I/A 90%; Stage I/B 78%; Stage I/C 58%; Stage II/A 26%; Stage II/B 27%; Stage II/C 14%; Stage III/A 18%. From the other 7 patients six had sex cord tumor and one lipid cell tumor. The number of the side effects is in good agreement with the data in literature. The use of 198Au for intraperitoneal treatment of ovary tumors is not contemporary today because of gamma radiation of radiogold, but intraperitoneal radiation treatment should not be forgotten. C1 [Patyanik, Mihaly] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Uzsoki utca 29, 1145 Budapest, Hungary. [Mayer, Arpad] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Uzsoki utca 29, 1145 Budapest, Hungary. [Polgar, Istvan] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Uzsoki utca 29, 1145 Budapest, Hungary. RP Patyanik, M (reprint author), Fovarosi Onkormanyzat Uzsoki utcai Korhaza, 1145 Budapest, Hungary. CR Chamberlain RH: Outline of the applicants of radioactive colloidal gold 198 to intraperioneal, intrapleural and intrathecal use. In. A preliminary manual of the properties and use of colloidal gold 198:23-31, 1951. Currie JR, Bayne F, Harris C et al: Radioactive chromic phosphate suspension: Studies on distribution, dose, absorption and effective therapeutic radiation in phantoms, dogs and patients. Gynecol Oncol 12:193-218, 1981. Decker DG, Webb MJ, Holbrook MA: Radiogold treatment of epithelial cancer of the ovary. Am J Obstet Gynecol 115:751- 756, 1973. Dembo AJ: Abdominopelvic radiotherapy in ovarium cancer. A 10 year experience. Cancer 55:2285-2290, 1985. Eltringham JR: Radiation therapy for ovarium carcinoma. Clin Obstet Gynecol 22:967-992, 1979. Epenetos AA, Munro AJ, Steward S: Antibody-guided irradiation of advanced ovarium cancer with intraperitoneally administered radiolabelled monoclonal antibodies. J Clin Oncol 12:1890-1899, 1987. Fletcher GM: Textbook of Radiology. Lea and Febiger, Philadelphia, 1973. Fuks Z: Patterns of spread of ovarium carcinoma relation to therapeutic strategies. Adv Biosci 26:39-51, 1980. Kalebi IA, et al: Radioimmunotherapy of solid cancers. Acta Oncolo 35:343-355, 1996. Klaassen D, Shelley W, Starreveld A, et al: External beam pelvic radiotherapy plus intraperitoneal radioactive chromic phosphate in early stage ovarium cancer. A toxic combination. Int J Radiol Oncol Biol Phys 11:1801-1804, 1985. Moor DW, Langley H: Routine use of radiogold following operation for ovarium cancer. Am J Obstet Gynecol 98:624-628, 1967. Muller JH: Curative aim and results of routine intraperitoneal radiocolloid administration in the treatment of ovarian cancer. Am J Roentgenology, Radiat Ther Nucl Med 89:533-540, 1963. Muller JL: Weitere Entwicklungen der Therapie von peritoneal Carcinosen bei ovarial Carcinom mit kunstlicher Radioaktivitat, 198Au). Gynecologia 129: 89-294, 1950. Munnell EW: Is conservative therapy ever justified in Stage I, Ia, cancer of the ovary? Am J Obstet Gynecol 103:641-650, 1969. Ozols RF: Carboplatin and Paclitaxel in ovarium Cancer. Seminars in Oncology 22: Suppl 6, 78-84, 1995. Pagenelli G, Belloni C, Magnani P, et al: Two-step tumor targeting in ovarian cancer patients using biotinylated monoclonal antibodies and radioactive streptavidine. Eur J Nucl Med 19:322, 1992. Perez CA, Walz BJ, Jacobson PL: Radiation therapy in management of carcinoma of ovary, Symposium of Ovarium Cancer NCI Monograph 42:119-125, 1975. Pezner RD, Stevens KR Jr., Tong D, Allen CV: Limited epithelial carcinoma of the ovary treated with curative intent by the intraperitoneal instillation of radio colloids. Cancer 42:2563- 2571, 1978. Piver MS, Barlow JJ, Lelle SB: Incidency of subclinical metastasis in Stage I and II ovarium carcinoma. Obstet Gynecol 52:100-104, 1978. Rosenshein NB: Radioisotopes in the treatment of ovarium cancer. Clin Obstet Gynaecol 10:279-295, 1983. Steven M. Larson: Choosing right radionuclid and antibody for intraperitoneal radioimmuntherapy. J Nat Cancer Inst 83:1662- 1664, 1991. Varia M: Intraperitoneal chromic phosphate therapy after second- look laparatomy for ovarian cancer. Cancer 61:919-927. 1992. Zalutsky R. et al: Radioimmuntherapy with a-particle emitting radioimunoconjugates. Acta Oncolo 35:373-379, 1996. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 54 EP 57 PG 4 ER PT J AU Dehaghani, SA Amirzargar, AA Farjadian, Sh Ghaderi, A AF Dehaghani, Samsami Alamtaj Amirzargar, Ali-Akbar Farjadian, Shirin Ghaderi, Abbas TI HLA-DQBl Alleles and Susceptibility to Cervical Squamous Cell Carcinoma in Southern Iranian Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cervix carcinoma; HLA-DQ; PCR-SSP ID cervix carcinoma; HLA-DQ; PCR-SSP AB The association of HLA class II with various autoimmune diseases has been extensively investigated. Despite the importance and functions of HLA genes in the evolution of cancer, the allele specific association of HLA molecules in cancer patients has not been well investigated. In this study the HLA-class II alleles frequency was investigated in Iranian patients with cervical squamous cell carcinoma. HLA typing was carried out by PCR amplification using sequence specific primers (PCR-SSP). DRB1, DQA1 and DQB1 typing was performed for 23 patients. The allele frequencies were calculated and compared with 36 healthy Iranian female controls. A positive association was observed between the existence of HLA-DQB1* 0601 and squamous cell carcinoma of the cervix (p<0.04, RR=1.94). Moreover, analysis of HLA-DRB1, DQA1 and DQB1 haplotypes indicated that none of the putative haplotypes were significantly associated with either patient or control group. Positive association of cervical carcinoma with a single allele of HLA-DQ provides evidence on the importance of HLA class II molecules and the immune response in squamous cell carcinoma of cervix. C1 [Dehaghani, Samsami Alamtaj] Shiraz University of Medical Sciences, Department of Obstetrics and GynecologyShiraz, Iran. [Amirzargar, Ali-Akbar] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Farjadian, Shirin] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Ghaderi, Abbas] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. RP Ghaderi, A (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Immunology, Shiraz, Iran. EM ghaderia@sums.ac.ir CR Torres LM, Cabrera T, Concha A, et al: HLA class I expression and HPV-16 sequences in premalignant and malignant lesions of the cervix. Tissue Antigens 41:65-71, 1993. Reid R: Immunology and Immunotherapy. In: practical Gynecology Oncology. 1st Edition. Williams and Wilkins Co, 1989. Wank R, Thomssen C: High risk of squamous cell carcinoma of the cervix for women with HLA-DQw3. Nature 352:723-725, 1991. Glew SS, Stern PL, Davidson JA, Dyers PA: HLA antigens and cervical carcinoma. Nature 356:22, 1992. Gregoire L, Lawrence WD, Kukuruga D, et al: Association between HLA-DQB 1 alleles and risk for cervical cancer in African-American women. Int J Cancer 57:504-507, 1994. Nawa A, Nishiyama Y, Kobayashi T, et al:Association of human leukocyte antigen-DQB1*03 with cervical cancer in Japanese women aged 35 years and younger. Cancer 75:518-521, 1995. Vandenvelde C, De Foor M, Van Beers D: Precision about the association between cervical carcinoma and HLA-DQBl*03 alleles. Lancet 342:553, 1993. Wank R, Meulen JT, Luande J, et al: Cervical intraepithelial neoplasia, cervical carcinoma and risk for patients with HLADQB1* 0602, *0301, *0303 alleles. Lancet 341:1215, 1993. Odunsi K, Terry G, Ho L, et al: Susceptibility to human papillomavirus- associated cervical intra-epithelial neoplasia is determined by specific HLA-DR-DQ alleles. Int J Cancer 67:595-602, 1996. Graham DE: The isolation of high molecular weight DNA from whole organisms or tissue masses. Anal Biochem 85:609- 613, 1978. Miller SA, Dykes DD, Polesky HE: A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16:1215-1221, 1989. Olerup O, Zetterquist H: HLA-DR typing by PCR amplification with sequence-specific primers, PCR-SSP, in 2 hours: an alternative to serological DR-typing in clinical practice including donor-recipient matching in cadaveric Transplantation. Tissue Antigens 39:225-235, 1992. Zetterquest H, Olerup O: Identification of the HLA-DRB1*04, DRB1*07 and DRB1*09 alleles by PCR amplification with sequence-specific primers, PCR-SSP, in 2 hours. Hum Immunol 34:64-74, 1992. Beck HP, Felger I, Barker M, et al: Evidence of HLA-class II association with antibody response against the malaria vaccine SPF66 in a naturally exposed population. Am J Trop Med Hyg 53:284-288, 1995. Naruse TK, Matsuzawa Y, Ota M, et al: HLA-DQB1*0601 is primarily associated with the susceptibility to cardiac sarcoidosis. Tissue Antigens 56:52-57, 2000. Illeni MT, Pasquali M, La Monica G, et al: HLA antigens in ovarian adenocarcinoma patients. Eur J Gynecol Oncol 6:121- 125, 1985. Montoya L, Saiz I, Rey G, et al: Cervical carcinoma: human papillomavirus infection and HLA-associated risk factors in the Spanish population. Eur J Immunogenet 25:329-37, 1998. Hamsikova E, Sramkova I, Loudova M, et al: The frequency of HLA-DRB 1 and -DQB 1 alleles in Cervical Cancer cases in the Czech Republic. Folia Biol 45:31-34, 1999. Neuman RJ, Huettner PC, Li L, et al: Association between DQB 1 and cervical cancer in patients with human papillomavirus and family controls. Obstet Gynecol 95:134-140, 2000. Amirzargar A, Mytilineos J, Farjadian Sh, et al: HLA Class-II allele frequencies and haplotype association in Iranian normal population. Hum Immunol 62:1234-1238, 2001. Furgyik S, Grubb R, Kullander S, et al: Familial occurrence of cervical cancer, stage 0-IV. Acta Obstet Gynecol Scand 65:223-227, 1986. zur Hausen H, Schneider A: The role of papillomaviruses in human anogenital cancer. In: The papovaviridae: the papillomaviruses, eds. Salzman NP, and Howley PM, Vol: 2, pp. 245- 263, Plenum Press, New York, 1987. Farahmandbeigi M: The incidence rate of registered cancer in Fars province. Disease Control Unit, Shiraz University Press, Iran, 2000. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 58 EP 61 PG 4 ER PT J AU Bhurgri, Y Hasan, HSh Pervez, Sh Kayani, N Hussainy, ShA Muzaffar, S Khurshid, M AF Bhurgri, Yasmin Hasan, H Sheema Pervez, Shahid Kayani, Naila Hussainy, Shah Akbar Muzaffar, Suhail Khurshid, Mohammad TI Large-Scale Pathology-Based Cancer Data - a Reflection of Population-Based Cancer Data SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer; pathology ID cancer; pathology AB Pathology-based cancer data is a high quality reflection of the patterns of cancer in the population it represents, provided the demographic details of the patients are carefully recorded. Relative frequency data is neither a replacement for population-based data nor a suggested alternative; it simply enhances the quality of population data and in very large data sets reflects the cancer patterns observed in the representative populations. Aware of the standard shortfalls of pathology-based data, the department of pathology, ‘The Aga Khan University Hospital’ (AKUH) standardized its data, representing 53.4% of the cancer data of Karachi Division (Pakistan) and also reflecting the cancer pattern of other provinces of Pakistan. This data was compared with 4 different population and institutional-based data sets. The findings substantiate the observation that ''despite the problems of interpretation of data from pathology laboratories, they are an invaluable source of information on cancer patterns in much of the world where incidence data are unavailable''. If developing countries, unable to organize National Population- Based Registry should as an alternate develop National Pathology-based Registers a well targeted and monitored, a Cancer Control Program would be possible. A good quality, large-scale pathology data with demographic details of the patient recorded can also be extended to give coverage to the population. C1 [Bhurgri, Yasmin] The Aga Khan University, Department of Pathology and Microbiology, Stadium Rd., 74800 Karachi, Pakistan. [Hasan, H Sheema] The Aga Khan University, Department of Pathology and Microbiology, Stadium Rd., 74800 Karachi, Pakistan. [Pervez, Shahid] The Aga Khan University, Department of Pathology and Microbiology, Stadium Rd., 74800 Karachi, Pakistan. [Kayani, Naila] The Aga Khan University, Department of Pathology and Microbiology, Stadium Rd., 74800 Karachi, Pakistan. [Hussainy, Shah Akbar] The Aga Khan University, Department of Pathology and Microbiology, Stadium Rd., 74800 Karachi, Pakistan. [Muzaffar, Suhail] The Aga Khan University, Department of Pathology and Microbiology, Stadium Rd., 74800 Karachi, Pakistan. [Khurshid, Mohammad] The Aga Khan University, Department of Pathology and Microbiology, Stadium Rd., 74800 Karachi, Pakistan. RP Bhurgri, Y (reprint author), The Aga Khan University, Department of Pathology and Microbiology, 74800 Karachi, Pakistan. CR Parkin DM: Cancer Occurrence in Developing Countries. IARC Technical Report No. 75. IARC Lyon, 1986. Bhurgri Y., Bhurgri A., Hassan SH, et al:Cancer Incidence in Karachi, Pakistan: First Results from Karachi Cancer Registry. Int. J. Cancer 85:325-329, 2000. Bhurgri Y: Cancer Registration in Pakistan. Asia Pacific J. Cancer Prev, 2 IARC Supplement, 51-54, 2000. Bhurgri Y: Epidemiology of Cancers in Karachi 1995-1999, KCR Technical Report No. 5; Karachi Cancer Registry, 2001. Jensen OM, Parkin DM, MacLennan R, et al, eds.): Cancer Registration: Principles and Methods;IARC Scientific Publications No. 95 Lyon IARC, 1991. Parkin DM, Chen VW, Ferley J,(eds.): Comparability and Quality Control in Cancer Registration, IARC Technical Report No.19, Lyon IARC, 1994. WHO, International Classification of Diseases for Oncology, Ed. 2, Geneva, WHO 1990. Rothwell DJ, ed.): Systemic Nomenclature of Medicine, SNOMED, microglossary for surgical pathology, College of American Pathologists, Library of Congress Number: 80, 68528 WHO, 1992, International Classification of Diseases, 10th Revision, Geneva, WHO 1992. Census Bulletin-1, Population and Housing Census of Pakistan 1998, Population Census Organisation Statistics division, Federal Bureau of Statistics, Government of Pakistan, 1998. Jafarey NA, Mahmood Z, Zaidi SHM: Habits and dietary patterns of cases of carcinoma of the oral cavity and oropharynx. J Pak Med Assoc 27:340-343, 1977. Jafarey NA: Betel quid, betel nuts and other chewing habits. Postgradu Doctor Africa 15:42-440, 1994. Alam SE: Prevalence and pattern of smoking in Pakistan. J Pak Med Assoc 48:64-66, 1998. Merchant AT, Luby SP, Perveen G: Smoking in Pakistan: more than cancer and heart disease. J Pak Med Assoc 43:77-79, 1998. Sankaranarayanan R, Duffy SW, Padmakumari G, et al: Tobacco chewing, alcohol and nasal stuff in cancer of the gingiva in Kerala, India. Br J Cancer 60:638-643, 1989. Sankaranarayanan R, Duffy SW, Padmakumari G, et al:. Risk factors for cancer of the buccal and labial mucosa in Kerala, Ind J Epidemiol Comm Health 13:286-292, 1990. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 62 EP 67 PG 6 ER PT J AU Gombas, P Skepper, NJ Hegyi, L AF Gombas, Peter Skepper, N Jeremy Hegyi, Laszlo TI The Image Pyramid System - an Unbiased, Inexpensive and Broadly Accessible Method of Telepathology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE education; image pyramid; remote consultation; telemedicine; telepathology ID education; image pyramid; remote consultation; telemedicine; telepathology AB Although computerised information technology, including the Internet is broadly used and globally accessible it is still not a significant form of professional communications in diagnostic histopathology. The high cost of interactive dynamic telepathology systems makes their use limited outside the richest economies. In contrast static telepathology systems are relatively cheap but in practice their information content can be heavily biased by the choice of images sent by the consulting pathologist. The degree of this bias may be regarded simply as the amount of information transferred to a remote location expressed as a percentage of the total information present in the histological sample. We refer to this as the percentage of explicit versus implicit information. Another major source of bias may be found in the information transmitted in written or verbal discussion with a remote consultant. We have developed a system of static telepathology, the image pyramid, which attempts to minimise bias by transferring all of the information in a section to the consultant. It is inexpensive and should prove to be widely accessible. C1 [Gombas, Peter] Military Hospital, Department of Pathology, Varosligeti Fasor 9-11, H-1071 Budapest, Hungary. [Skepper, N Jeremy] University of Cambridge, Department of Anatomy, Multi-Imaging Centre, CB2 3DY Cambridge, UK. [Hegyi, Laszlo] University of Cambridge, Department of Medicine, Addenbrooke’s Hospital, Level 6, ACCI, Hills Road, CB2 2QQ Cambridge, UK. RP Gombas, P (reprint author), Military Hospital, Department of Pathology, H-1071 Budapest, Hungary. EM pgombas@bm.gov.hu CR Weinstein RS, Bloom KJ, Rozek LS: Telepathology and the networking of pathology diagnostic services. Arch Pathol Lab Med 111:646-652, 1987. Kayser K, Szymas J, Weinstein R: Telepathology. Telecommunication, Electronic Education and Publication in Pathology., 1st ed). Springer Verlag: Berlin, 97-150, 1999. Wells CA, Softer C: Telepathology: a diagnostic tool for the millennium? J Pathol 191:1-7, 2000. Raggett J, Bains W: Artificial Intelligence from A to Z. Chapman and Hall, London. 1992. Martin E, Dusserre P, Fages A, et al: Telepathology: a new tool of pathology? Presentation of a French national network. Zentralbl Pathol 138:419-423, 1992. Kayser K, Oberholzer M, Weisse G, et al: Long distance image transfer: First results of its use in histopathological diagnosis. APMIS 99:808-814, 1991. Kayser K, Drileck M, Rahn W: Aids of telepathology in intraoperative histomorphological tumor diagnosis and classification. In Vivo 7:395-398, 1993. Eide TJ, Nordrum I: Current status of telepathology. APMIS 102:881-890, 1994. Weinstein RS, Bhattacharyya AK, Graham AR, et al: Telepathology: a ten-year progress report. Hum Pathol 28:1-7, 1997. Gombas P, Szende B, Stotz G: Future aspects and benefits of telematic networks used in pathology for countries of central Europe, CCE). Elec J Pathol Histol; 2:963-968, 1996. Schubert E, Gross W, Siderits RH, et al: A pathologist-designed imaging system for anatomic pathology signout, teaching, and research. Semin Diagn Pathol 11:263-73, 1994. Bashshur RL: Telemedicine effects: cost, quality, and access. J Med Syst 19: 81-91, 1995. Halliday BE, Bhattacharyya AK, Graham AR, et al: Diagnostic accuracy of an international static-imaging telepathology consultation service. Hum Pathol 28:17-21. 1997. Viellefond A, Staroz F, Fabre M, et al: Martin-Pop V, Martin E, Got C, Franc B. Reliability of the anatomopathological diagnosis by static image transfer. Arch Anat Cytol Pathol 43:246-50, 1995. Callas PW, Leslie KO, Mattia AR, et al: Diagnostic accuracy of a rural live video telepathology system. Am J Surg Pathol 21:812-819, 1997 Dunn BE, Almagro UA, Choi H, et al: Dynamic-robotic telepathology. Department of Veterans Affairs feasibility study. Hum Pathol 28:8-12, 1997. Poremba C, Pickhardt N: Economic evaluation of telepathology. Pathologe 19: 18-24, 1998. Sowter C, Wells CA: Telepathology: assessment of the implications and applications of telepathology for practical diagnostic pathology. [editorial ] J Clin Pathol 51: 14-715, 1998. Petersen I, Wolf G, Roth K, Schluns K: Telepathology by the Internet. J Pathol 191:8-11, 2000. Weinstein RS, Bloom KJ, Krupinski EA et al: Human performance studies of the video microscope component of a dynamic telepathology system. Zentralbl Pathol 138:399-403, 1992. Mairinger T, Netzer TT, Schoner W et al:. Pathologist’ attitudes to implementing telepathology. J Telemed Telecare 4:41-46, 1998. Weinberg DS, Allaert FA, Dussere P et al: Telepathology diagnosis by means of digital still images: an international validation study. Hum Pathol 27: 111-118, 1996. Weinstein RA: Futurist meets the 21 century: love at first sight [editorial ] Hum Pathol 31: 1-2, 2000. Katt EC, Dennis SE: Web-based pathology education. Arch Pathol Lab Med 122:745-749, 1998. Kayser K, Kayser G. Recent development of telepathology in Europe with specific emphasis on quality assurance. Anal Quant Cytol Histol 21:319-21, 1999. Gombas P, Skepper JN, Hegyi L: The Image Pyramid – digital slide in the surgical pathology. http://ultra.obuda.kando.hu/~medinfo/tp/ 2001. Gombas P: Informational aspects of telepathology on routine surgical pathology Anal Cell Pathol 21:141-147, 2000. Harris T, Leaven T, Heidger P, Kreiter C: Comparison of a virtual microscope laboratory to a regular microscope laboratory for teaching histology Anat Rec 265:10-4, 2001. Leong FJ, J. McGee JO: Automated complete slide digitization: a medium for simultaneous viewing by multiple pathologists. J Pathol 195:508-514, 2001. Saltz HJ: Digital Pathology – The big picture Hum Pathol; 2000; 31:779-780. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 68 EP 73 PG 6 ER PT J AU Cserni, G AF Cserni, Gabor TI SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE ductal carcinoma in situ (DCIS); solid papillary carcinoma; breast cancer; revertant DCIS ID ductal carcinoma in situ (DCIS); solid papillary carcinoma; breast cancer; revertant DCIS AB A case of invasive carcinoma of mixed papillary and not otherwise specified ductal type with areas of solid papillary ductal carcinoma in situ(DCIS) is reported. The solid papillary areas were predominantly of low nuclear grade, but a small area of intermediate-grade solid papillary neoplasm was also seen within the tumor, together with an area suggestive of microinvasion. The massive regional nodal tumor load consisted of invasive papillary carcinoma and revertant low-grade solid papillary carcinoma with no myoepithelial cells around the circum-scribed solid papillary areas. This is the first report of a solid papillary pattern simulating intraductal carcinoma in lymph nodes, and the first time that a solid papillary carcinoma is reported in association with invasive papillary carcinoma. The case suggests that mammary carcinomas with a solid papillary pattern may sometimes be of higher grade than usual, and do not always represent a DCIS, but may be invasive. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., H-6000 Kecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.hu CR Barsky SH, Bose S: Should LCIS be regarded as a heterogeneous disease? Breast J 5:407-412, 1999. Barsky SH, Doberneck SA, Sternlicht MD, et al: “Revertant” DCIS in human axillary breast carcinoma metastases. J Pathol 183:188-194, 1997. Buerger H, Otterbach F, Simon R, et al: Different genetic pathways in the evolution of invasive breast cancer are associated with distinct morphological subtypes. J Pathol 189:521-526, 1999. Capella C, Eusebi V, Mann B, et al: Endocrine differentiation in mucoid carcinoma of the breast. Histopathology 4:613-630, 1980. Cross AS, Azzopardi JG, Krausz T, et al: A morphological and immunohistochemical study of a distinctive variant of ductal carcinoma in situ of the breast. Histopathol 9:1-37, 1985. Diest PJ van: Ductal carcinoma in situ in breast carcinogenesis. J Pathol 187:383-384, 1999. Douglas-Jones AG, Gupta SK, Attanoos RL, et al: A critical appraisal of six modern classifications of ductal carcinoma in situ of the breast, DCIS): correlation with grade of associated invasive carcinoma. Histopathol 29: 97-409, 1996. Ellis IO, Eslton CW, Poller DN: Ductal carcinoma in situ. In: Elston CW, Ellis IO, Eds, The Breast, Systemic Pathology, Volume 13, 3rd ed., pp 249-281. Churchill Livingstone, Edinburgh, 1998. Elston CW, Ellis IO: Assessment of histological grade. In: Elston CW, Ellis IO, Eds, The Breast, Systemic Pathology, Volume 13, 3rd ed., pp 365-384. Churchill Livingstone, Edinburgh, 1998. Lampejo OT, Barnes DM, Smith P, et al: Evaluation of infiltrating carcinomas with a DCIS component: correlation of the histologic type of the in situ component with the grade of the infiltrating component. Semin Diagn Pathol 11: 215-222, 1994 Lee AHS, Telfer TP, Millis RR: Metastatic breast carcinoma with appearance resembling micropapillary ductal carcinoma in situ. J Clin Pathol 48:380-382, 1995. Maluf HM, Koerner FC: Solid papillary carcinoma of the breast. A form of intraductal carcinoma with endocrine differentiation frequently associated with mucinous carcinoma. Am J Surg Pathol 19:1237-1244, 1995. Maluf HM, Zukerberg LR, Dickersin GR, et al: Spindle-cell argyrophilic mucin-producing carcinoma of the breast. Histological, ultrastructural, and immunohistochemical studies of two cases. Am J Surg Pathol 15:677-686, 1991. National Coordinating Group for Breast Screening Pathology. Pathology reporting in breast cancer screening, 2nd ed., p65. NHSBSP Publications, Sheffield, 1997. Pinder SE, Elston CW, Ellis IO: Invasive carcinoma - usual histological types. In: Elston CW, Ellis IO, Eds, The Breast, Systemic Pathology, Volume 13, 3rd ed., pp 283-337. Churchill Livingstone, Edinburgh, 1998. Rosen PP, Oberman HA: Tumors of the mammary gland. Fascicle 7, 3rd series, p217. AFIP, Washington DC, 1993. Roylance R, Gorman P, Harris W, et al: Comparative genomic hybridisation of breast tumors stratified by histological grade reveals new insights into the biological progression of breast cancer. Cancer Res 59:1433-1436, 1999. Saphir O: Mucinous carcinoma of the breast. Surg Gynecol Obstet 72:908-914, 1941. Sternlicht MD, Kedeshian P, Shao ZM, et al: The human myoepithelial cell is a natural tumor suppressor. Clin Cancer Res 3:1949- 1958, 1997. Tsang WYW, Chan JKC: Endocrine ductal carcinoma in situ, EDCIS, of the breast. Am J Surg Pathol 20:921-943, 1996. Yaziji H, Gown AM, Sneige N: Detection of stromal invasion in breast cancer: the myoepithalial markers. Adv Anat Pathol 7:100- 109, 2000. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2002 VL 8 IS 1 BP 74 EP 77 PG 4 ER PT J AU Dank, M AF Dank, Magdolna TI The Role of Aromasin in the Hormonal Therapy of Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE exemestane; aromasin; hormonal therapy; breast cancer ID exemestane; aromasin; hormonal therapy; breast cancer AB In the last 40 years tamoxifen and progestogens constituted the basis of hormonal therapy. Introduction of the third generation, selective, anti-aromatase agents added effective drugs of good tolerability to the anti-cancer armamentarium. Exemestane, an oral steroidal-type aromatase inhibitor - which irreversibly blocks aromatase - is very effective in the treatment of metastatic breast cancer. As a second line therapy, exemestane is more effective and causes less side effects than megestrol-acetate. Its administration as first line therapy gave promising results. The role of exemestane in adjuvant treatment has not yet been soundly established but trials are ongoing. It may be effective as neoadjuvant treatment in selected groups of patients. Future studies will clarify exemestane’s role in chemoprevention and in the treatment of post-menopausal women administered together with cytostatic agents. C1 [Dank, Magdolna] Semmelweis University, Department of Radiology and Oncotherapy, Ulloi ut 78a, H1082 Budapest, Hungary. RP Dank, M (reprint author), Semmelweis University, Department of Radiology and Oncotherapy, H1082 Budapest, Hungary. EM danke@radi.sote.hu CR Buzdar A, Howell A: Advances in aromatase inhibition: Clinical efficacy and tolerability in the treatment of breast cancer. Clin Cancer Res 7:2620-2635, 2001. Beatson GT: On the treatment of inoperable cases of carcinoma of the mamma, suggestions for a new method of treatment with illustrative cases. Lancet 2:104-107, 1896. Huggins C, Dao T L-Y: Adrenalectomy and oophorectomy in treatment of advanced carcinoma of the breast. JAMA 151:1388-1394, 1953. Luft R, Olivecrona H, Sjogren B: Hypophysectomy in man. Nord Med 14:351-354, 1952. NissenMeyer R, Vogt JH: Cortisone treatment of metastatic breast cancer. Acta Unio Contra Cancrum 15:1140-1144, 1959. Nosaquo ND: Androgens and estrogens in the treatment of disseminated mammary carcinoma. JAMA 172:135-147, 1960. Cole MP, Jones CTA, Todd IDH: A new anti-oestrogenic agent in late breast cancer. An early clinical appraisal of ICI46474. Br J Cancer 25:270-275, 1971 Cash R, Brough AJ, Cohen MNP, et al: Aminogluthetimide is an inhibitor or adrenal steroidogenesis: mechanim of action and therapeutic trial. J Clin Endocrinol Metab 27:1239-1248, 1967. Panutti F, Martoni A, Lenaz GR, et al: A possible new approach to the treatment of metastatic breast cancer: massive doses of medroxyprogesterone acetate. Cancer Treat Rep 62:499-504, 1978. Coombes RC, Goss P, Dowsett M, et al: 4-hydroxyandrostendione in treatment of postmenopausal patients with advanced breast cancer. Lancet 2:1237-1239, 1984. Honig SF: Treatment of metastatic disease. In Harris JR, et al, eds. Diseases of the Breast. Philadelphia PA, Lippincott-Raven Publishers, Boston, 1996, pp 669-734. Robertson JFR, Lee D: Proc Am Soc Clin Oncol 16: 151a, 1997. Shenton KC, Dowsett M, Lu Q, et al: Comparison of biochemical activity with aromatase immunhistochemistry in human breast carcinoma. Breast Cancer Res Treat 49:S101-S107,1998. Brueggemeier RW: Aromatase inhibitors - mechanisms of steroidal inhibitors. Breast Cancer Res Treat 30:31-42,1994. Brodie AM, Garrett WM, Hendrickson JR, et al: Inactivation of aromatase in vitro by 4-hydroxy-androstene-3,17-dione and 4- acetaxy-4-androstene-3,17-dione and sustained effects in vitro. Steroids 38:693-702, 1981. Harada N, Honda S-I, Hatano O: Aromatase inhibitors and enzyme stability. Endocrine Related Cancer 6:211-218, 1999. Sasano H, Sato S, Ito K, et al: Effects of aromatase inhibitors on the pathobiology of human breast endometrial and ovarian carcinoma. Endocrine Related Cancer 6:197-204,1999. Miller WR: Biology of aromatase inhibitors: pharmacology/endocrinology within the breast. Endocrine Related Cancer 6:187-195,1999. Lonning: Breast 74:1286-1291,1996. Geisler J, King N, Anker G, et al: In vivo inhibition of aromatization by exemestane, a novel irreversible aromatase inhibitor in postmenopausal breast cancer patients. Clin Cancer Res 4:2089-2093, 1994. diSalle E, Ornati G, Giudici D, et al: Exemestane, FCE 24304, a new steroidal aromatase inhibitor. J Steroid Biochem Mol Biol 49:289-294, 1994. Giudici D, Ornati G, Briatico G, et al: 6-Methylenandrosta- 1,4-diene-3,17-dion, FCE 24304): a new irreversible aromatase inhibitor. J Steroid Biochem 30:391-394, 1988. diSalle E, Briatico G, Giudici D, Ornati G: Novel aromatase and 5-reductase inhibitors. J Steroid Biochem Mol Biol 49:289-294, 1994. Johannessen DC, Engan T, diSalle E: Endocrine and clinical effects of exemestane, PNU 155971), a novel steroidal aromatase inhibitor, in postmenopausal breast cancer patients: a PhaseI study Clin Cancer Res 3:1101-1108, 1997. Lundgren S, Helle SI, Lonning PE: Profound suppression of plasma estrogen by megestrol acetate in postmenopausal breast cancer patients. Clin Cancer Res 2:1515-1521, 1996. Buzdar A: Exemestane in advanced breast cancer. AntiCancer Drug 11:609-616, 2000. Thurlimann B, Paradaens R, Serin D: Third-line treatment with exemestane in postmenopausal patients with advanced breast cancer progressing on aminogluthetimide. A Phase II multicentre multinationale study. Eur J Cancer 33:1767-1773, 1997. Lonning PE, Bajetta E, Murray R: Activity of exemestane, Aromasin®, in metasttic breast cancer after failure of nonsteroidal aromatase inhibitors: a phase II trial J Clin Oncol 18: 2234-2244, 2000. Jones S, Chang A, Lusch C: A phase II confirmatory study of antitumor efficacy and safety of exemestane, EXE, as third line hormonal treatment of postmenopausal patients, PTS, with metastatic breast cancer, MBC, refractory to tamoxifen, T, and Megace®San Antonio Breast Cancer Conference, 1998B Jones S, Vogel C, Arkhipov A: Phase II trial of exemestane as third line hormonal therapy of postmenopausal women with metastatic breast cancer J Clin Oncol 17:3418-3425,1999. Kvinnsland S, Anker G, Dirix L-Y: High activity and tolerability demonstrated for exemestane in postmenopausal women with metastatic breast cancer failing tamoxifen Eur J Cancer 36:976- 982, 2000. Jones S, Belt R, Cooper B: A phase II study of antitumor efficacy and safety of exemestane, EXE, as a second line hormonal treatment of postmenopausal patients with metastatic breast cancer, MBC, refractory to tamoxifen, Tam, San Antonio Breast Conference, 1998A Kaufmann M, Bajetta E, Dirix L-Y: Exemestane is superior to megestrol acetate following tamoxifen failure in postmenopausal women with advanced breast cancer. Results of Phase III randomised double-blind trial. J Clin Oncol 18:1399- 1344, 2000. Tedeschi M, Kvinnsland S, Jones SE: Hormonal therapy in breast cancer and predominant visceral disease: effectiveness of the new oral aromatase inactivator, Aromasin®, exemestane, in advanced breast cancer patients having progressed on antioestrogens. 10th European Cancer Conference, Vienna, Austria 1999. Paradiens R, Dirix L-Y, Beex L: Exemestan, Aromasin, is active and well tolerated as first line hormonal therapy, HT, in metastatic breast cancer, MBC, patients, Pt): result of a randomised phase II trial. Proc Am Soc Clin Oncol 19:83a,2000. Early Breast Cancer Trialists’ Collaborative Group: Tamoxifen for early breast cancer: an overview of the randomised trials. Lancet 351:1451-1467,1998. Dixon M, Anderson T, Miller WR: Phase IIb study of neoadjuvant exemestan in locally advanced breast cancer. ASCO Abstr 1908, 2001. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 87 EP 92 PG 6 ER PT J AU Esik, O Tusnady, G Tron, L Boer, A Szentirmay, Z Szabolcs, I Racz, K Lengyel, E Szekely, J Kasler, M AF Esik, Olga Tusnady, Gabor Tron, Lajos Boer, Andras Szentirmay, Zoltan Szabolcs, Istvan Racz, Karoly Lengyel, Erzsebet Szekely, Judit Kasler, Miklos TI Markov Model-based Estimation of Individual Survival Probability for Medullary Thyroid Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE medullary thyroid carcinoma; prognosis; multivariate analysis; Markov model; individual survival probability; MEDUPRED ID medullary thyroid carcinoma; prognosis; multivariate analysis; Markov model; individual survival probability; MEDUPRED AB The relatively benign, but occasionally rapidly fatal clinical course of medullary thyroid cancer (MTC) has raised the need for individual survival probability estimation. A retrospective study on 91 MTC clinical case histories with a mean follow-up of 6 years indicated prevalences of local, regional and distant residual tumor on primary care completion of 23%, 54% and 54%, respectively. Local, regional and distant relapses during follow-up occurred in 8%, 23% and 26% of the patients, with a cause-specific death in 26% of the cases. Prognostic factors statistically significantly influencing the cause-specific survival were selected by uni- and multivariate analysis. A Markov method-based model was developed for the estimation of individual time-dependent local, regional and distant relapse-free and cause-specific survival probability functions, with parameters numerically determined via a maximum likelihood procedure. These parameters include relative risk factors related to prognosticators, a residual or recurrent local/regional/distant tumor, and combinations of these entities. In multivariate studies, the patient’s age and gender, the genetic basis of the dis-ease, lymph node involvement, the existence of a general symptom (diarrhoea) at presentation, and the dosage of external irradiation proved to be prognosticators. The cause-specific survival function of the study population indicated mean 5, 10 and 15-year survival probabilities of 69%, 62% and 58%. CONCLUSION: Survival probabilities can be predicted for extrastudy cases provided that the same laws and principles govern the clinical course of these cases and those comprising the study. For individual survival probability estimation, a Pascal program (MEDUPRED) was written and is available on the home page of the National Institute of Oncology, Budapest (www.oncol.hu). C1 [Esik, Olga] Semmelweis University, Department of OncologyBudapest, Hungary. [Tusnady, Gabor] Hungarian Academy of Sciences, Renyi Alfred Mathematical InstituteBudapest, Hungary. [Tron, Lajos] University of Debrecen, PET CenterDebrecen, Hungary. [Boer, Andras] National Institute of Oncology, Department of Head and Neck SurgeryBudapest, Hungary. [Szentirmay, Zoltan] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Szabolcs, Istvan] National Health Centre, Department of EndocrinologyBudapest, Hungary. [Racz, Karoly] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Lengyel, Erzsebet] National Institute of Oncology, Center of Radiotherapy, H-1122 Budapest, Hungary. [Szekely, Judit] National Institute of Oncology, Center of Radiotherapy, H-1122 Budapest, Hungary. [Kasler, Miklos] National Institute of Oncology, Department of Head and Neck SurgeryBudapest, Hungary. RP Esik, O (reprint author), Semmelweis University, Department of Oncology, Budapest, Hungary. CR Anderson PK, Borgan O, Gill RD, et al: Statistical models based on counting processes. Springer Verlag, Berlin-Heidelberg- New York-London-Paris-Tokyo-Hong Kong-Barcelona- Budapest, 1992. Backdahl M, Carstensen J, Auer G, et al: Statistical evaluation of the prognostic value of nuclear DNA content in papillary, follicular, and medullary thyroid tumors. World J Surg 10:974- 980, 1986. Beressi N, Campos JM, Beressi JP, et al: Sporadic medullary microcarcinoma of the thyroid: a retrospective analysis of eighty cases. Thyroid 8:1039-1044, 1998. Bergholm U, Adami H-O, Auer G, et al: Histopathologic characteristics and nuclear DNA content as prognostic factors in medullary thyroid carcinoma. A nationwide study in Sweden. Cancer 64:135-142, 1989. Bergholm U, Bergstrom R, Ekbom A: Long term follow-up of patients with medullary carcinoma of the thyroid. Cancer 79:132-138, 1997. BMDP Statistical Software Inc. Los Angeles, 1990. Bottger T, Klupp J, Sorger K, et al: Therapie und Prognose des medullaren Schilddrusenkarzinoms. Med Klin 86:8-14, 1991. Brierley J, Tsang R, Simpson WJ, et al: Medullary thyroid cancer: analyses of survival and prognostic factors and the role of radiation therapy in local control. Thyroid 6:305-310, 1996. Cox DR: Regression models and life tables. J Roy Stat Soc B 34 Series B:187-220, 1972. Dabrowska DM, Sun G-w, Horowitz MM: Cox regression in a Markov renewal model: an application to the analysis of bone marrow transplant data. J Am Stat Ass 89: 867-877, 1994. Dralle H, Damm I, Scheumann GFW, et al: Compartment-oriented microdissection of regional lymph nodes in medullary thyroid carcinoma. Surg Today 24:112-121, 1994. Ekman ET, Bergholm U, Backdahl M, et al: Nuclear DNA content and survival in medullary thyroid carcinoma. Cancer 65:511-517, 1990. El-Naggar AK, Ordonez NG, McLemore D, et al: Clinicopathologic and flow cytometric DNA study of medullary thyroid carcinoma. Surgery 108: 981-985, 1990. Eng C, Clayton D, Schuffenecker I, et al: The relationship between specific RET proto-oncogene mutations and disease phenotype in multiple endocrine neoplasia type 2. International RET Mutation Consortium analysis. JAMA 276:1575-1579, 1996. Esik O, Tusnady G, Daubner K, et al: Survival chance in papillary thyroid cancer in Hungary: individual survival probability estimation using the Markov method. Radiother Oncol 44:203-212, 1997. Esik O, Szavcsur P, Szakall S Jr, et al. Angiography effectively supports the diagnosis of hepatic metastases in medullary thyroid cancer. Cancer 91:2084-2095, 2001. Gharib H, McConahey WM, Tiegs RD, et al: Medullary thyroid carcinoma: clinicipathologic features and long-term follow-up of 65 patients treated during 1946 through 1970. Mayo Clinic Proc 67:934-940, 1992. Gilliland FD, Hunt WC, Morris DM, et al: Prognostic factors for thyroid carcinoma. A population-based study of 15,698 cases from the Surveillance, Epidemiology and End Results, SEER, Program 1973-1991. Cancer 79:564-573, 1997. Gimm O, Ukkat J, Dralle H: Determinative factors of biochemical cure after primary and reoperative surgery for sporadic medullary thyroid carcinoma. World J Surg 22:562-568, 1998. Grebe SKG, Hay ID: Thyroid cancer nodal metastases. Biologic significance and therapeutic considerations. Surg Oncol Clin North Am 5:43-63, 1996. Grigsby PW, Luk KH: Thyroid. In: Principles and practice of radiation oncology., Eds: Perez CA and Brady LW, 3rd ed, Lippincott Co, Philadelphia, PA, 1998, pp 1157-1180. Hannequin P, Liehn JC, Delisle MJ: Multifactorial analysis of survival in thyroid cancer. Pitfalls of applying the results of published studies to another population. Cancer 58:1749-1755, 1986. Hay ID, Ryan JJ, Grant CS, et al: Prognostic significance of nondiploid DNA determined by flow cytometry in sporadic and familial medullary thyroid carcinoma. Surgery 108:972- 980, 1990. Kallinowski F, Buhr HJ, Meybier H, et al: Medullary carcinoma of the thyroid – therapeutic strategy derived from fifteen years of experience. Surgery 114:491-496, 1993. Kaplan EL, Meier P: Nonparametric estimation from incomplete observations. J Am Stat Ass 53:457-481, 1958. Klein I, Esik O, Homolya V, et al: Molecular genetic diagnostic program of MEN2a and FMTC syndromes in Hungary. J Endocrinol 170:661-666, 2001. Lippman SM, Mendelsohn G, Trump DL, et al: The prognostic and biological significance of cellular heterogeneity in medullary thyroid carcinoma: a study of calcitonin, L-DOPA decarboxylase, and histaminase. J Clin Endocrinol Metab 54:233-240, 1982. Marsh DJ, Learoyd DL, Robinson BG: Medullary thyroid carcinoma: recent advances and management update. Thyroid 5:407-424, 1995. Mendelsohn G, Wells SA Jr, Baylin SB: Relationship of tissue carcinoembryonic antigen and calcitonin to tumor virulence in medullary thyroid carcinoma. An immunohistochemical study in early, localized, and virulent disseminated stages of the disease. Cancer 54:657-662, 1984. Pyke CM, Hay ID, Goellner JR, et al: Prognostic significance of calcitonin immunoreactivity, amyloid staining, and flow cytometric DNA measurements in medullary thyroid carcinoma. Surgery 110:964-971, 1991. Rejto L, Tusnady G: On the Cox regression. In: Asymptomatic methods in probability and statistics., Ed: Szyszkowicz B, Elsevier Science Pbl, BV North-Holland, 1998, pp 621-637. Rougier P, Parmentier C, Laplanche A, et al: Medullary thyroid carcinoma: prognostic factors and treatment. Int J Radiat Oncol Biol Phys 9:161-169, 1983. Russel CF, van Heerden JA, Sizemore GW, et al: The surgical management of medullary thyroid carcinoma. Ann Surg 197:42-48, 1983. Saad MF, Ordonez NG, Rashid RK, et al: Medullary carcinoma of the thyroid. A study of the clinical features and prognostic factors in 161 patients. Medicine 63:319-342, 1984. Samaan NA, Schultz PN, Hickey RC: Medullary thyroid carcinoma: prognosis of familial versus sporadic disease and the role of radiotherapy. J Clin Endocrinol Metab 67:801-805, 1988. Samaan NA, Schultz PN, Hickey RC: Medullary thyroid carcinoma: prognosis of familial versus sporadic disease and the role of radiotherapy. Hormone Metab Res 21(Suppl): 21-25, 1989. Schroder S, Bocker W, Baisch H, et al: Prognostic factors in medullary thyroid carcinomas. Survival in relation to age, sex, stage, histology, immunocytochemistry, and DNA content. Cancer 61:806-816, 1988. Scopsi L, Sampietro G, Boracchi P, et al: Multivariate analysis of prognostic factors in sporadic medullary carcinoma of the thyroid. A retrospective study of 109 consecutive patients. Cancer 78:2173-2183, 1996. Simpson WJ, Palmer JA, Rosen IB, et al: Management of medullary carcinoma of the thyroid. Am J Surg 144:420-422, 1982. Spiessl B, Beahrs OH, Hermanek P, et al, eds). TNM atlas. Illustrated guide to the TNM/pTNM classification of malignant tumors. 3rd ed., Springer Verlag, Berlin-Heidelberg-New York- London-Paris-Tokyo-Hong Kong-Barcelona, 1990, pp 56-61. Szakall S Jr, Esik O, Bajzik G, et al: 18F-FDG PET detection of lymph mode metastasis in medullary thyroid carcinoma. J Nucl Med 43:66-71, 2002. Tusnady G, Esik O: MEDUPRED, software for individual survival probability estimation of medullary thyroid cancer patients, www@oncol.hu: Budapest, 2000. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 93 EP 104 PG 12 ER PT J AU Pasquale, AM Weppler, D Smith, J Icardi, M Amador, A Gonzalez, M Tomoaki, K Tzakis, A Ruiz, P AF Pasquale, A Melissa Weppler, Debbie Smith, Jon Icardi, Michael Amador, Alexandra Gonzalez, Monica Tomoaki, Kato Tzakis, Andreas Ruiz, Phillip TI Burkitt's Lymphoma Variant of Post-transplant Lymphoproliferative Disease (PTLD) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PTLD; Burkitt’s lymphoma ID PTLD; Burkitt’s lymphoma AB The occurrence of posttransplant lymphoproliferative disorder (PTLD) in solid organ allograft recipients can be quite varied in clinical presentation, histopathological characteristics and frequency. A variety of lymphomas can develop as a PTLD although some types appear infrequently and remain poorly understood in this clinical setting. In this report, we describe two cases of Burkitt’s lymphoma presenting as a PTLD following liver transplantation. The recipients were 12 and 44 years of age and displayed gastrointestinal involvement by the tumors several years following transplant. The tumors displayed the typical histological features of Burkitt’s lymphoma and were markedly positive for EBV. The tumors displayed similar immunophenotypic characteristics by flow cytometry and had rearrangements of the immunoglobulin J-H heavy chain. The tumors required aggressive chemotherapy and a cessation of immunosuppressive therapy. This report demonstrates that Burkitt’s type lymphomas can develop in the posttransplant setting and that these tumors contain morphologic, cytofluorographic and molecular features identical to Burkitt’s lymphomas that occur in non-transplant patients. Our experience is that these PTLD- Burkitt’s lymphomas behave aggressively and require intensive chemotherapeutic intervention. C1 [Pasquale, A Melissa] University of Miami, Department of Pathology, Jackson Memorial Hospital - Holtz Center, Room 2101, 33136 Miami, Florida, USA. [Weppler, Debbie] University of Miami, Department of SurgeryMiami, Florida, USA. [Smith, Jon] University of Miami, Department of Pathology, Jackson Memorial Hospital - Holtz Center, Room 2101, 33136 Miami, Florida, USA. [Icardi, Michael] University of Miami, Department of Pathology, Jackson Memorial Hospital - Holtz Center, Room 2101, 33136 Miami, Florida, USA. [Amador, Alexandra] University of Miami, Department of Pathology, Jackson Memorial Hospital - Holtz Center, Room 2101, 33136 Miami, Florida, USA. [Gonzalez, Monica] University of Miami, Department of SurgeryMiami, Florida, USA. [Tomoaki, Kato] University of Miami, Department of SurgeryMiami, Florida, USA. [Tzakis, Andreas] University of Miami, Department of SurgeryMiami, Florida, USA. [Ruiz, Phillip] University of Miami, Department of Pathology, Jackson Memorial Hospital - Holtz Center, Room 2101, 33136 Miami, Florida, USA. RP Ruiz, P (reprint author), University of Miami, Department of Pathology, 33136 Miami, USA. EM pruiz@med.miami.edu CR Jain A, Reyes J, Kashyap R, et al: Liver transplantation under tacrolimus in infants, children, adults and seniors: long-term results, survival and adverse events in 1000 consecutive patients. Transplant Proc 30:1403-1404, 1998. Ho M, Jaffe R, Miller G, et al: The frequency of Epstein-Barr virus infection and associated lymphoproliferative syndrome after transplantation and its manifestations in children. Transplantation 45:719-727, 1988. Cacciarelli TV, Green M, Jaffe R, et al: Management of posttransplant lymphoproliferative disease in pediatric liver transplant recipients receiving primary tacrolimus, FK506, therapy. Transplantation 66:1047-1052, 1998. Malatack JF, Gartner JC Jr, Urbach AH, Zitelli BJ: Orthotopic liver transplantation, Epstein-Barr virus, cyclosporine, and lymphoproliferative disease: a growing concern. J Pediatr 118:667-675, 1991. Harris NL, Ferry JA, Swerdlow SH: Posttransplant lymphoproliferative disorders: summary of Society for Hematopathology Workshop. Semin Diag Pathol 1997;14(1):8-14. Penn I. Cancers complicating organ transplantation. NEJM 323:1767-1769, 1990. Leblond V, Sutton L, Dorent R, et al: Lymphoproliferative disorders after organ transplantation: a report of 24 cases observed in a single center. J Clin Oncol 13:961-968, 1995. Frank D, Cesarman E, Lie YF, et al: Posttransplantation lymphoproliferative disorders frequently contain Type A and not Type B Epstein-Barr virus. Blood 85:1396-1403, 1995. Walker RC, Paya, CV, Marshall WF, et al: Pretransplantation seronegative Epstein-Barr virus status is the primary risk factor for posttransplantation lymphoproliferative disorder in adult heart, lung, and othe rsolid organ transplantations. J Heart Lung Transplant 14:214-221, 1995. Harris NL, Jaffe ES, Diebold J, et al: World Health Organization classification of neoplastic diseases of the haematopoietic and lymphoid tissues: report of the Clinical Advisory Committee Meeting, Airlie House, Virginia, November 1997. Histopathology 36:69-86, 2000. Berard, et al. Histopathological definition of Burkitt’s Tumour. Bull WHO 40:601-607, 1969. Cossman J, Uppenkamp M, Sundeen J, et al: Molecular genetics and the diagnosis of lymphoma. Arch Pathol Lab Med 112:117-127, 1988. Wu TC, Mann RB, Epstein JI, et al: Abundant expression of EBER1 am11 nuclear RNA in nasopharyngeal carcinoma, a morphologically distinctive target for detection of Epstein-Barr virus in formalin-fixed paraffin-embedded carcinoma specimens. Am J Pathol 46:1310-1313, 1991. Rea D, Fourcade C, Leblond V, et al: Epstein-Barr virus latent and replicative gene expression in posttransplant lymphoproliferative disorders and AIDS-related non-Hodgkin’s lymphomas. Ann Oncol 5, Suppl 1): S113-S116, 1994. Cen H, Williams PA, McWilliams HP, et al: Evidence of restricted Epstein-Barr virus latent gene expression and anti- EBNA antibody response in solid organ transplant recipients with posttransplant lymphoproliferative disorders. Blood 81:1393-1403, 1993. Smets F, Vajro P, Cornu G, et al: Indications and results of chemotherapy in children with posttransplant lymphoproliferative disease after liver transplantation. Transplantation 69:982- 984, 2000. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 105 EP 108 PG 4 ER PT J AU Teni, T Pawar, S Sanghvi, V Saranath, D AF Teni, Tanuja Pawar, Sagar Sanghvi, Vikram Saranath, Dhananjaya TI Expression of Bcl-2 and Bax In Chewing Tobacco-Induced Oral Cancers and Oral Lesions from India SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bcl-2; bax; p53; oral cancer; oral lesions; immunohistochemistry ID bcl-2; bax; p53; oral cancer; oral lesions; immunohistochemistry AB Deregulation of oncogenes and tumor suppressor genes involved in apoptosis has been associated with tumor development and progression. To investigate the involvement of apoptosis regulating proteins in oral cancer in Indian patients, primarily associated with chewing tobacco habits, immunohistochemical expression of bcl-2 and bax was examined in 63 oral squamous cell carcinomas, and 31 putative premalignant lesions. Our studies revealed overexpression of tumor specific cytoplasmic bcl-2 in 56% and bax in 43% oral cancers. The oral cancers in the Indian patients are preceded by premalignant oral lesions; hence oral lesions were examined for bcl-2 and bax expression. We observed aberrant expression of bcl-2 in 16% oral lesions comprising leukoplakias and SMF and bax in 55% oral lesions. We have already reported, p53 expression in these oral cancers and lesions. It was noteworthy that 30% oral cancers demonstrated a p53+bcl2+ pattern, and 14% samples exhibited p53+bcl2+bax+ pattern. However, none of the oral lesions showed concurrent deregulation of p53 and bcl-2 or all the three genes. Interestingly 45% oral lesions were p53-bax+ as compared to 18% oral cancers; while 39% oral lesions were bcl2-bax+ as compared to 14% oral cancers, indicating overexpression of bax in oral lesions, in the absence of p53 and bcl-2 proteins. Significant correlation was observed between positive nodal status and bcl2+ (p=0.047) and p53+bcl-2+ (p=0.01) in oral cancers. Kaplan Meier survival analysis showed significantly (p=0.059) higher survival in patients with p53- oral tumors than with p53+ tumors. Our studies thus indicate frequent overexpression of apoptosis regulators bcl-2, bax and p53 proteins in oral cancers, and a subset of oral lesions, representing early events in oral car-cinogenesis. The aberrant bcl-2 expression and loss of p53 function observed, may play an important role in the tumorigenesis of oral cancers by allowing escape from apoptosis and enabling additional genetic alterations to accrue. C1 [Teni, Tanuja] Tata Memorial Centre, Cancer Research Institute, Laboratory of Cancer Genes, Jaslok Hospital and Research Centre, 15, Dr.G.Deshmukh Marg, 400026 Parel, Mumbai, India. [Pawar, Sagar] Tata Memorial Centre, Cancer Research Institute, Laboratory of Cancer Genes, Jaslok Hospital and Research Centre, 15, Dr.G.Deshmukh Marg, 400026 Parel, Mumbai, India. [Sanghvi, Vikram] Tata Memorial Hospital, Tata Memorial Centre, Department of SurgeryParel, Mumbai, India. [Saranath, Dhananjaya] Tata Memorial Centre, Cancer Research Institute, Laboratory of Cancer Genes, Jaslok Hospital and Research Centre, 15, Dr.G.Deshmukh Marg, 400026 Parel, Mumbai, India. RP Saranath, D (reprint author), Tata Memorial Centre, Cancer Research Institute, Laboratory of Cancer Genes, 400026 Parel, India. EM dsaranath@jaslokhospital.net CR Moore SR, Johnson NW, Pierce AM, Wilson DF: The epidemiology of mouth cancer: a review of global incidence. Oral Diseases 6:65-74, 2000. National Cancer Registry Programme: Consolidated Report of the Population Based Cancer Registries : 1990–1996, Incidence and Distribution of Cancer under the ICMR, New Delhi, August 2001. Gupta PC, Bhonsle RB, Murti PR, et al: An epidemiological assessment of cancer risk of oral precancerous lesions in India with special reference to nodular leukoplakia. Cancer 63:2247- 2251, 1989. Tradati N, Grigolat R, Calabrase L, et al : Oral leukoplakia to treat or not? Oral Oncol 33:317- 321, 1997. Saranath D, Panchal RG, Nair R, et al: Oncogene amplification in squamous cell carcinoma of the oral cavity. Jpn J Cancer 80:430-437, 1989. Saranath D, Panchal RG, Nair R, et al: Amplification and overexpression of epidermal growth factor receptor in human oropharyngeal cancer. Eur J Cancer B Oral Oncol. 28:139-143, 1992. Saranath D, Chang SE, Bhoite LT, et al: High frequency mutation in codon 12 and 61 of H-ras oncogene in chewing tobacco related human oral carcinomas in India. Br J Cancer 63:573- 578, 1991. Saranath D, Bhoite LT, Mehta AR, et al: Loss of allelic heterozygosity at the Harvey locus in human oral carcinomas. J Cancer Res Clin Oncol. 117:483-488, 1991. Saranath D, Bhoite LT and Deo MG: Molecular lesions in human oral cancer: the Indian scene. Eur J Cancer Oral Oncol. 29:107-112,1993. Tandle A and Saranath D: Genetic alterations on chromosome 3, p53 tumor suppressor gene and telomerase activity in oral cancers from India. Asian Pacific Journal of Cancer Prevention, Vol. I APCC Supplement 219-225, 2000. Mahale A and Saranath D: Microsatellite alterations on chromosome 9 in chewing tobacco -induced oral cell carcinomas from India. Oral Oncol 36:199-206, 2000. 12 Saranath D, Tandle AT and Deo MG: Loss of p53 gene as a biomarker of high risk oral leukoplakia. Indian J Biochem Biophy 34:266-273, 1997. 13. Saranath D, Tandle AT, Teni TR, et al: p53 inactivation in chewing tobacco-induced oral cancers and leukoplakias from India. Oral Oncol. 35:242-250, 1999. 14. Field JK, Spandidos DA and Stell PM: Overexpression of the p53 gene in head and neck cancer, linked with heavy smoking and drinking. Lancet 339:502-503, 1992. 15. Miyashita T, Krajewski S, Krajweska M, et al: Tumor suppressor p53 is a regulator of bcl2 and bax expression in vitro and in vivo. Oncogene 9:1799 –1805, 1994. 16. Wang Y, Szekely L, Okan J, Klien G et al: Wild type p53 triggered apoptosis is inhibited by bcl2 in v-myc induced T-cell lymphoma lines. Oncogene 8:3427-3431, 1993. 17. Pollina L, Pacini F, Fontanini G et al: Bcl2, p53 and proliferating cell nuclear antigen expression is related to the degree of differentiation in thyroid carcinomas. Br J Cancer 73:139-143, 1996. 18. Qi-Lung, Abel P, Foster CB, et al: Bcl-2: Role in epithelial differentiation and oncogenesis. Human Pathol. 27:102-109, 1996. 19. Jordan RCK, Catzavelos GC, Barrett AW, et al: Differential expression of bcl2 and bax in squamous cell carcinomas of the oral cavity. Oral Oncol. 32B:394-400, 1996. 20. Stoll C, Baretton G, Abrens C, et al: Prognostic significance of apoptosis and associated factors in oral squamous cell carcinoma. Virchows Arch 436:102-108, 2000. 21. Birchall MA, Schock E, Harmon BV et al : Apoptosis, Mitosis, PCNA and bcl2 in normal, leukoplakic and malignant epithelia of the human oral cavity: prospective, in-vivo study. Oral Oncol 3:419-425, 1997. 22. Pena J, Thompson CB, Recant W, et al: Bcl-xL and Bcl2 expression in squamous cell carcinoma of head and neck. Cancer 85:164-170, 1999. 23. Yao L, Iwai M and Furuta I: Correlation of bcl2 and p53 expression with the clinico pathological features in tongue squamous cell carcinomas. Oral Oncol. 35:56-62, 1999. 24. Ravi D, Ramadas K, Mathew BS, et al : De novo programmed cell death in oral cancer. Histopathol. 14:241-249, 1999. 25. Kannan K, Lakshmi Latha PN and Shanmugan G : Expression of bcl2 oncoprotein in Indian oral squamous cell carcinomas. Oral Oncol 34:373-377, 1998. 26. McAlinden RL, Maxwell P, Napiet S, et al: Bcl-2 expression in sequential biopsies of potentially malignant oral mucosal lesions assessed by immunocytochemistry. Oral Diseases 6:318-326, 2000. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 109 EP 114 PG 6 ER PT J AU Nagy, G Szekeres, Gy Kvell, K Berki, T Nemeth, P AF Nagy, Gergely Szekeres, Gyorgy Kvell, Krisztian Berki, Timea Nemeth, Peter TI Development and Characterisation of a Monoclonal Antibody Family Against Aquaporin 1 (AQP1) and Aquaporin 4 (AQP4) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE aquaporin 1; aquaporin 4; anti-aquaporin antibodies; monoclonal antibody; immunohistochemistry ID aquaporin 1; aquaporin 4; anti-aquaporin antibodies; monoclonal antibody; immunohistochemistry AB Recent studies have discovered the existence of water-channel molecules, the so called aquaporins (AQP) presumably involved in active, ATP dependent water transport between the intracellular and extracellular compartments. Both genetic and protein sequences and structures of the AQPs are known and crystallographic analyses of some members of the AQP family have been performed. Specific antibodies are required to examine their histological locations and analyse their roles in physiological and pathological pathways of water transportation and osmotic regulation. Until recently some polyclonal antibodies have been developed against certain members of the AQP family. However, to date highly specific monoclonal antibodies against aquaporins do not exist. We have developed a monoclonal antibody family against the aquaporin 1 (AQP1) and aquaporin 4 (AQP4) molecules. Well-conserved epitop sequences of AQP1 and AQP4 proteins were selected by computer analysis and their synthetic peptide fragments were used as the antigens of immunisation and the following screening. Antibodies were characterised by immunoserological methods (ELISA, dot-blot and immunoblot), flow cytometry and immunohistochemistry of formaldehyde-fixed and paraffin-embedded tissue samples. RT-PCR tests controlled the specificity of the immune reactions. Our monoclonal antibodies recognised AQP1 and AQP4 in all the techniques mentioned above and apparently they are useful both in various quantitative and qualitative measurements of the expressions of AQP1 and AQP4 in several species (human, rat, mouse, invertebrates, even plants). According to preliminary immunohistochemical studies our monoclonal anti-AQP1 and anti-AQP4 antibodies are appropriate tools of patho-morphological examinations on routine formol-paraffin tissue samples. C1 [Nagy, Gergely] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Szekeres, Gyorgy] Hisztopatologia KFTPecs, Hungary. [Kvell, Krisztian] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Berki, Timea] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Nemeth, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. RP Nemeth, P (reprint author), University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, H-7643 Pecs, Hungary. EM peter.nemeth@aok.pte.hu CR Agre, P, Brown D, Nielsen S: Aquaporin water channels: unanswered questions and unresolved controversies. Curr Op Cell Biol 7:472-483, 1995. Agre, P, Preston GM, Smith BL, et al: Aquaporin CHIP: the arce type molecular water channel. Am J Physiol 265:463-476, 1993. Bodis B, Nagy G, Nemeth P, et al: Ative water selective channels in the stomach: investigation of aquaporins after ethanol and capsaicin treatment in rats. J Phys Paris 95:271-278, in press). Borgnia M, Nielsen S, Engel A et al: Cellular and molecular biology of the aquaporin water channels. Ann Rev Biochem 68:425-458, 1999. Dyson, HJ, Wright PE: Antigenic peptides. FASEB J 9:37-42, 1995. Endo M, Jain RK, Witwer B et al: Water channel, aquaporin 1, expression and distribution in mammary carcinomas and glioblastomas. Microvasc Res 58:89-98, 1999. Engel A: Structural analyses of the aquaporin super-family. Nephrol Dial Transplant. 15 Suppl. 6:23-5, 2000. Fujita, Horio Y, Nielsen S, et al: High-resolution immunogold cytochemistry indicates that AQP4 is concentrated along the basal membrane of parietal cell in rat stomach. FEBS Lett 459:305-9, 1999. Greisz V, Burghardt B, Ferguson CJ, et al: Expression of aquaporin 1, AQP1, water channels in human labial salivary glands. Arch Oral Biol 44:S53-57, 1999. Greisz V, Kwon TH, Hurley PT, et al: Identification and localization of aquaporin water channels in human salivary glands. Am J Phys Gatrointest Liver Phys 281:G247-254, 2001. Gristen, S, Woodside M, Sardet C, et al:. Activation of Na+/H+ antiport during cell volume regulation. Evidece for phosphorylation- independent mechanism. J Biol Chem 267:23823- 23828, 1992. Hasegawa, H, Skach W, Baker O, et al: A multi-functional aqueous channel formed by CFTR. Science 258:1477-1479, 1992. Hopp, TP: Protein surface analysis. Methods for identifying antigenic determinants and other interaction sites. J Immunol Methods 88:1-18,1986. King LS, Yasui M, Agre P: Aquaporins in health and disease. Mol Med Today 6:60-5, 2000. Kirazov LP, Venkov LG, Kirazov EP: Comparison of the Lowry and the Bradford protein assays as applied for protein estimation of membrane-containing fractions. Anal Biochem 208:44-48, 1993. Knepper, MA: The aquaporin family of molecular water channels. Proc Natl Acad Sci US 91:6255-6258, 1994. Koyama Y, Yamamoto T, Tani T, et al: Expression and localisation of aquaporins in rat gastrointestinal tract, Am J Physiol 276:621-627, 1999. Lane, D: Antibodies: A laboratory Manual. Cold Spring Harbor Press, Cold Spring Harbor N.Y. 1988. Laver, WG, Air GM, Webster RG et al: Epitopes on protein antigens: misconceptions and realities. Cell 61:553-556, 1990. Loo, DD, Hirayama BA,. Meinild AK et al: Passive water and ion transport by cotransporter. J Physiol 518:195-202, 1999. Lopez JM, Imperial S, Valderrama R, et al: An improved Bradford protein assay for collagen proteins. Clin Chim Acta 220:91-100, 1993. Marshall T, Williams KM: Bradford protein assay and the transition from an insoluble to a soluble dye complex: effects of sodium dodecyl sulphate and other additives. J Biochem Biophys Methods 26:237-240, 1993. Najbauer, J., Tigyi, G., Nemeth, P.: Antigen-specific cell adherence assay: a new method for separation of antigen-specific hybridoma cells. Hybridoma 5:361-370, 1986. Vajda Z, Promeneur D, Doczi T et al: Increased Aquaporin-4 immunoreactivity in rat brain in response to systematic hyponatremia. Biochem Biophys Res Comm 270:495-503, 2000. Verkman, AS: Water channels in cell membranes Ann Rev Physiol 54:97-108, 1992. Vizuete ML, Venero JL, Vargas C et al: Astrocytes and aquaporin- 4, AQP4, play a significant role in brain ion homeostasis. Neurobiol Dis 6:245-58, 1999. Work, TS, Work E, editors. Laboratory techniques in Biochemistry and Molecular Biology, Vol. 11. Elsevier Science, Oxford. 37-41, 94-96, 1982. Yang, B, and Verkman AS: Urea transporter UT3 functions as an efficient water channel: direct evidence for a common water/urea pathway. J Biol Chem 273:9369-9372, 1998. Zhang, R, Alper S, Thorens B, et al:Evidence from oocyte expression that the erythrocyte water channel is distinct from band and the glucose transporter. J Clin Invest 88:1553-1558, 1991. Zor T, Selinger Z: Linearization of the Bradford protein assay increases its sensitivity: theoretical and experimental studies. Anal Biochem 236: 302-308, 1996. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 115 EP 124 PG 10 ER PT J AU Madhavan, M Srinivas, P Abraham, E Ahmed, I Vijayalekshmi, RN Balaram, P AF Madhavan, Maya Srinivas, Priya Abraham, Elizabeth Ahmed, Iqbal Vijayalekshmi, Rugmini Narayana Balaram, Prabha TI Down Regulation of Endothelial Adhesion Molecules in Node Positive Breast Cancer: Possible Failure of Host Defence Mechanism SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE VCAM; ICAM; E-selectin; invasion; breast cancer ID VCAM; ICAM; E-selectin; invasion; breast cancer AB Endothelial cell adhesion molecules (CAMs) are important in tumorigenesis and host defense mechanism. Their status in breast cancer with regard to nodal invasion is not yet known. Hence we looked at the expression of three important CAMs: VCAM, ICAM and E-selectin. A downregulation of all these CAMs was noted in node positive breast cancer in comparison to node negative cases. This suggests shedding of these molecules in cases with nodal metastasis which might help the tumor cells to escape the host defense mechanism. On multi-variate analysis, VCAM alone emerged as an independent predictor of nodal metastasis. C1 [Madhavan, Maya] Regional Cancer Centre, Division of Cancer Research, 695011 Trivandrum, Kerala, India. [Srinivas, Priya] Regional Cancer Centre, Division of Cancer Research, 695011 Trivandrum, Kerala, India. [Abraham, Elizabeth] Regional Cancer Centre, Division of HistopathologyTrivandrum, Kerala, India. [Ahmed, Iqbal] Regional Cancer Centre, Division of Surgical OncologyTrivandrum, Kerala, India. [Vijayalekshmi, Rugmini Narayana] University of Kerala, Department of BiochemistryTrivandrum, Kerala, India. [Balaram, Prabha] Regional Cancer Centre, Division of Cancer Research, 695011 Trivandrum, Kerala, India. CR Nicolson GL: Organ colonization and cell-surface properties of malignant cells. Biochim Biophys Acta 695:113-176, 1982. Pauli BU, Augustin-Voss HG, El-Sabban ME, et al: Organ preference of metastasis. The role of endothelial cell adhesion molecules. Cancer Metastasis Review 9:175-189, 1990. Nicolson GL: Metastatic tumor cell interaction with endothelium, basement membrane and tissue. Curr Opin Cell Biol 1:1009-1019, 1989. Liotta LA: Tumor invasion and metastasis of the extracellular matrix. Cancer Res 46:1-7,1986. Harning R, Mainolfi E, Brystryn JC, et al: Serum levels of circulating intercellular adhesion molecule 1 in human malignant melanoma Cancer Res 51:5003-5005, 1991. Srivastava MD, Srivastava A, Srivastava BI: Soluble interleukin 2 receptor, soluble CD8 and soluble intercellualr adhesion molecule 1 levels in hematologic malignancies Leuk Lymphoma 12:241-251, 1994. Koyama S, Ebihara T, Fukao K: Expression of intercellular adhesion molecule1, ICAM-1)during the development of invasion and/or metastasis of gastric carcinoma. J Cancer Res Clin Oncol 118:609-614, 1992. Pober JS, Cotran RS: Immunologic interactions of T lymphocytes with vascular endothelium. Adv Immunol 50:261-302, 1991. Stoolman LM: Adhesion molecules controlling lymphoctye migration. Cell 56:907-910, 1989. Osborn L: Leukocyte adhesion to endothelium in inflammation. Cell 62:3-6, 1990. Osborn L , Hession C, Tizard R, et al: Direct expression cloning of Vascular cell adhesion molecule-1, a cytokine induced endothelial protein that binds to lymphocytes. Cell 59:1203-1211,1989. Vogetseder W, Feichtinger H, Schulz TF, et al: Expression of 7F7-antigen, a human adhesion molecule identical to Intercellular adhesion molecule-1, ICAM-1, in human carcinomas and their stromal fibroblasts. Int J Cancer 43:768-773,1989. Makgoba MW, Sanders ME, Ginther Luce GE, et al: Functional evidence that intercellular adhesion molecule(ICAM-1, is a ligand for LFA-1 dependent adhesion in T-cell mediated cytotoxicity. Eur J Immunol 18:637-640, 1988. Bevilacqua MP, Pober JS, Mendrick DL, et al: Identification of an inducible endothelial-leukocyte adhesion molecule. Proc Nat Acad Sci, Wash, 84:9238-9242, 1987. Lasky LA: Selectins: interpreters of cell specific carbohydrate information during inflammation. Science 258:964-969,1992. Tsujisaki M, Imai K, Hirata H, et al: Detection of circulating intercellular adhesion molecule-1 antigen in malignant diseases. Clin Exp Immunol 85:3-8, 1991. Viac J, Gueniche A, Faure M, et al: Soluble intercellular adhesion molecule 1(sICAM-1, and malignant melanoma. Cancer Letters 72:191-194, 1993. Gruss HJ, Dolken G, Brach MA, et al: Serum levels of circulating ICAM-1 are increased in Hodgkins disease. Leukemia 7:1245-1249, 1993. Wittig BM, Treichel U, Blaheta R, et al: Soluble E-selectin enhances ICAM-1 expression in human tumor cell lines. Exp Cell Res 237:364-70,1997. Lawson C, Ainsworth M, Yacoub M, et al: Ligation of ICAM- 1 on endothelial cells leads to expression of VCAM-1 via a nuclear factor-KB independent mechanism. J Immunol 162:2990-6.1999. Rothlein R, Mainolf EA, Czajkowski M, Marlin SD: A form of circulating ICAM-1 in human serum. J Immunol 147:3788- 3793, 1991. Seth R, Raymond FD, Mukgoba MW: Cirulating ICAM-1 isoforms: diagnostic prospects for inflammatory and immune disorders. Lancet 338:83-84, 1991. Kageshita Y, Yoshii A, Kimura T, et al: Analysis of expression and soluble form of intercellular adhesion molecule-1 in malignant melanoma. J Dermatol 19:836-840, 1992. Anastossiou G, Schilling H, Stang A, et al: Expression of the cell adhesion molecules ICAM-1, VCAM-1 and NCAM in uveal melanoma: A clinicopathological study. Oncology 58:83-88, 2000. Pizzolo G, Vinante F, Nadali G, et al: ICAM-1 tissue overexpression associated with increased serum levels of its soluble form in Hodgkins disease. Br J Haematol 84:161-162, 1993. Vogetseder W, Feichtinger H, Schulz TF, et al: Expression of 7F7 antigen, a human adhesion molecule identical to intercellular adhesion molecule-1, ICAM-1, in human carcinomas and their stromal fibroblasts. Int J Cancer 43:768-773, 1989. Gearing AJH, Newman W: Circulating adhesion molecules in disease. Immunol Today 14:506-512, 1993. Banks RE, Gearing AJH, Hemingway IK, et al: Circulating intercellular adhesion molecule-1, ICAM-1), E-selectin and vascular cell adhesion molecule-1(VCAM-1, in human malignancies. Br J Cancer 68:122-124, 1993. Liu CM, Sheen TS, Ko JY, et al: Circulating intercellular adhesionmolecule1( ICAM-1), E-selectin and vascular cell adhesion molecule1(VCAM-1, in head and neck cancer. Br J Cancer 79:360-362,1999. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 125 EP 128 PG 4 ER PT J AU Dekel, Y Koren, R Kugel, V Livne, MP Gal, R AF Dekel, Yoram Koren, Rumelia Kugel, Valentina Livne, M Pinhas Gal, Rivka TI Significance of Angiogenesis and Microvascular Invasion in Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE angiogenesis; factor VIII; renal cell carcinoma; prognosis ID angiogenesis; factor VIII; renal cell carcinoma; prognosis AB The aim of this study is to evaluate the relationship between tumor angiogenesis and microvascular invasion, and the subsequent development of metastatic disease in patients undergoing surgery for renal cell carcinoma (RCC). The study group consisted of 102 patients who underwent surgery for RCC between the years 1990 and 1997 in our institute with a mean follow up period of 81.3 months. Paraffin blocks were stained for Factor VIII - related antigen and CD34 which decorate endothelial cells in order to assess angiogenesis and microvascular invasion and their relevance for developing metastatic disease. When Factor VIII- related antigen staining was used we found that the microvessel count correlated with the development of metastatic disease with a mean count of 49.7 for patients with no evidence of disease and a mean count of 95.5 for patients who developed metastatic disease (p<0.05). We also found that microvascular invasion correlated with the development of metastatic disease. It was demonstrated in 55.5% of patients who developed metastatic disease versus 23.8% of patients with no evidence of disease with Factor VIII staining (p<0.05), and in 33.3% and 7.1%, respectively (p<0.05) with CD34 staining. This study suggest that demonstration of intense angiogenesis and micro-vascular invasion may be a predictor of a more aggressive tumor mandating closer follow up and consideration of adjuvant therapy. C1 [Dekel, Yoram] Hasharon Hospital, Department of Urology, Rabin Medical Center, Campus Golda, Petah Tikva, and Sackler Faculty of MedicinePetach Tikvah, Israel. [Koren, Rumelia] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Kugel, Valentina] Hasharon Hospital, Department of Urology, Rabin Medical Center, Campus Golda, Petah Tikva, and Sackler Faculty of MedicinePetach Tikvah, Israel. [Livne, M Pinhas] Hasharon Hospital, Department of Urology, Rabin Medical Center, Campus Golda, Petah Tikva, and Sackler Faculty of MedicinePetach Tikvah, Israel. [Gal, Rivka] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. RP Koren, R (reprint author), A, Rabin Medical Center (Golda Campus), Department of Pathology, Petach Tikvah, Israel. EM rumelia@isdnmail.co.il CR Bex A, Luboldt H, Sudermann T, et al: Influence of linomide on local tumor growth and metastasis of the human hormoneresistant prostate cancer cell line PC3 in an orthotopic model. Eur Urol 37:628-633, 2000. Bochner BH, Cote RJ, Weidner N, et al: Angiogenesis in bladder cancer: relationship between microvessel density and tumor prognosis. J Natl Cancer Inst 87:1603-1612, 1995. Campbell SC: Advances in angiogenesis research: relevance to urological oncology. J Urol 158:1663-1674, 1997. Crew JP: Vascular endothelial growth factor: An important angiogenic mediator in bladder cancer. Eur Urol 35:2-8, 1999. DeKernion JB, Derry D: The diagnosis and treatment of renal cell carcinoma. Cancer 45:1947-1953, 1980. DeKernion JB, Mukamel E: Selection of initial therapy for renal cell carcinoma. Cancer 60:539-546, 1987. Fuhrman SA, Lasky LC, Limas C: Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol 6:655-660, 1992. Guinan PD, Vogelzang NJ, Freman AM: Renal cell carcinoma: tumor size, stage and survival. J Urol 153:901-905, 1995. Hatcher PA, Anderson EE, Paulson DF, et al: Surgical management and prognosis of renal cell carcinoma invading the vena cava. J Urol 145:20-25, 1991. Hollingsworth HC, Kohn EC, Steinberg SM, et al: Tumor angiogenesis in advanced stage ovarian carcinoma. Am J Pathol 147:33-41, 1995. Imazano Y, Takebayashi Y, Nishiyama K, et al: Correlation between thymidine phosphorylase expression and prognosis in human renal cell carcinoma. J Clin Oncol15:2570-2578, 1997. Lang H, Lindner V, Saussine C, et al: Microscopic venous invasion: A prognostic factor in renal cell carcinoma. Eur Urol 38:600-605, 2000. Maldazys JD, deKernion JB: Prognostic factors in metastatic renal carcinoma. J Urol 136:376-380, 1986. Mancilla-Jimenez R: Papillary renal cell carcinoma: a clinical, radiologic and pathologic study of 34 cases. Cancer 38:246- 270, 1976. Marstic C, Salamon J, Weber R, et al: Microscopic venous infiltration as predictor of relapse in renal cell carcinoma. J Urol 148:271-276, 1992. Medeiros LJ, Gelb AB, Weiss LM: Renal cell carcinoma: prognostic significance of morphologic parameters in 121 cases. Cancer 61:1639-1644, 1988. Muss HB: The role of biological response modifiers in metastatic renal cell carcinoma. Semin Oncol 15:30-34, 1988. Myers GH, Fehrenbaker LG, Kellais PP: Prognostic significance of renal vein invasion by hypernephroma. J Urol 100:420-423, 1968. Page DL: Prognosis and breast cancer. Recognition of lethal and favorable prognostic types. Am J Surg Pathol 15:334-349, 1991. Patel NP, Lavengood RW: Renal cell carcinoma. Natural history and results of treatment. J Urol 119:722-727, 1978. Richie AWS: Current treatment approaches to renal cell carcinoma - the role of surgery. In Progress in the treatment of renal cell carcinoma. Proceeding of the National Symposium, Queens College, Cambridge, Massachusetts 1-8, 1989. Robson C: Radical nephrectomy for renal cell carcinoma. J Urol 89:37-41, 1963. Samma S, Yoshida K, Ozono S, et al: Tumor thrombus and microvascular invasion as prognostic factor in renal cell carcinoma. Jap J Clin Oncol 21:340-345, 1991. Sanchez De La Muela P, Zudaire JJ, Robles IE, et al: Renal cell carcinoma: vena cava invasion and prognostic factors. Eur Urol 19:284-290, 1991. Sevnic M, Kirkali Z, Yorukoglu K, et al:Prognostic significance of microvascular invasion in localized renal cell carcinoma. Eur Urol 38:728-733, 2000. Selli C, Hinshaw WM, Woodard BH, Paulson DF: Stratification of risk factors in renal cell carcinoma. Cancer 52:899-903, 1983. Sirgi KE, Wick MR, Swanson PE: B72.3 and CD34 immunoreactivity in malignant epithelioid soft tissue tumors. Adjuncts in the recognition of endothelial neoplasms. Am J Surg Pathol. 17:179-185, 1993. Skinner DG, Colvin RB, Vermillion CD, et al: Diagnosis and management of renal cell carcinoma a clinical and pathological study of 309 cases. Cancer 28:1165-1170, 1971. Skinner DG. Pfister RF, Colvin R: Extension of renal cell carcinoma into the vena cava. The rationale for aggressive surgical management. J Urol 107: 11-714, 1972. Slaton JW, Inoue K, Perrotte P, et al: Expression levels of genes that regulate metastasis and angiogenesis correlate with advanced pathological stage of renal cell carcinoma. Am J Pathol. 158:735-743, 2001. Srivastava A, Hughes LE, Woodcock JP, et al: Vascularity in cutaneous melanoma detected by Doppler sonography and histology: correlation with tumor behaviour. Br J Cancer 59:89- 91, 1989. Takebayashi Y, Natugoe S, Baba M, et al: Angiogenesis in esophageal squamous cell carcinoma. Oncol Rep 5:401-404, 1998. Tanigawa N, Amaya H, Matsumura M, et al : Tumor angiogenesis and expression of thymidine phosphorylase/platelet derived endothelial cell growth factor in human gastric carcinoma. Cancer 108:281-290, 1996. Van Poppel H, Vandendriessche H, Boel K, et al: Microscopic vascular invasion is the most relevant prognosticator after radical nephrectomy for clinically non-metastatic renal cell carcinoma. J Urol 158:45-49, 1997. Wenig BM, Abbondanzo SL, Heffess CS: Epithelioid angiosarcoma of the adrenal glands. A clinicopathologic study of nine cases with a discussion of the implications of finding “epithelial- specific” markers. Am J Surg Pathol. 18:62-73, 1994. Wingo PA, Tong T, Bolden S: Cancer Statistics, 1995. Cancer J Clin 45:8-30, 1995. World Health Organization: International Histologic Classification of Tumors. 2nd edition. Spring Verlag, Berlin, 1990. Yoshino S, Kato M, Okada K: Prognostic significance of microvessel count in low stage renal cell carcinoma. Int J Urol 2:156-160, 1995. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 129 EP 132 PG 4 ER PT J AU Arista-Nasr, J Gutierrez-Villalobos, L Nuncio, J Maldonaldo, H Bornstein-Quevedo, L AF Arista-Nasr, Julian Gutierrez-Villalobos, Lisa Nuncio, Juan Maldonaldo, Hector Bornstein-Quevedo, Leticia TI Fibrolamellar Hepatocellular Carcinoma in Mexican Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE fibrolamellar; hepatocellular carcinoma; liver; neoplasm ID fibrolamellar; hepatocellular carcinoma; liver; neoplasm AB The aim of this report is to describe the frequency, clinical, and morphologic characteristics of fibrolamellar hepatocellular carcinoma in Mexican patients. Fibrolamellar hepatocellular carcinoma (FLHCC) is a rare variant of hepatocellular carcinoma. Although this tumor appears to be predominant among the Caucasian population of the U.S, FLHCC has been described in many other countries. The frequency and characteristics of FLHCC in Latin American population is almost unknown. The clinico-pathologic characteristics of seven (5.8%) Mexican patients with FLHCC, obtained among 121 hepato-cellular carcinomas are described. The frequency of these tumors was compared with the frequency reported in other geographic areas in the international literature between 1980 and 1999. There were four women and three men. Two patients had taken oral contraceptives for six months and a year prior to diagnosis; another patient had positive serology for the hepatitis B virus. Common symptoms included a palpable mass, abdominal pain and weight loss; two patients presented jaundice. In two patients the tumor had been removed eight and three years previously, and they were readmitted when FLHCC recurred. In three patients the diagnosis was suspected in radiological studies (computed tomography and/or magnetic resonance). Laboratory tests were non-specific. In four patients, resection of the tumor was performed, and in the remaining three the neoplasm was diagnosed by percutaneous hepatic biopsy. Two patients had died of disease at the time of the study, and another was alive with recurrent disease. Conclusions: fibrolamellar hepatocarcinoma is an uncommon, but not an exceptional neoplasm in our population and represents 5.8% of all hepatocarcinomas reviewed. C1 [Arista-Nasr, Julian] Instituto National de Ciencias Medicas y Nutricion, Salvador Zubiran, Department of Pathology, Vasco de Quiroga No.15 Coi. Seccion XVI, 14000 Mexico City, Mexico. [Gutierrez-Villalobos, Lisa] Instituto National de Ciencias Medicas y Nutricion, Salvador Zubiran, Department of Pathology, Vasco de Quiroga No.15 Coi. Seccion XVI, 14000 Mexico City, Mexico. [Nuncio, Juan] Instituto National de Ciencias Medicas y Nutricion, Salvador Zubiran, Department of Pathology, Vasco de Quiroga No.15 Coi. Seccion XVI, 14000 Mexico City, Mexico. [Maldonaldo, Hector] Instituto National de Ciencias Medicas y Nutricion, Salvador Zubiran, Department of Pathology, Vasco de Quiroga No.15 Coi. Seccion XVI, 14000 Mexico City, Mexico. [Bornstein-Quevedo, Leticia] Instituto National de Ciencias Medicas y Nutricion, Salvador Zubiran, Department of Pathology, Vasco de Quiroga No.15 Coi. Seccion XVI, 14000 Mexico City, Mexico. RP Arista-Nasr, J (reprint author), Instituto National de Ciencias Medicas y Nutricion, Salvador Zubiran, Department of Pathology, 14000 Mexico City, Mexico. EM pipa5@hotmail.com CR Saab S, Yao F: Fibrolamellar hepatocellular carcinoma. Case reports and a review of the literature. Dig Dis Sci 41:1981- 1985, 1996. Ruffin MT: Fibrolamellar hepatoma. Am J Gastroenterol 85: 577-581, 1990. Teilez-Zenteno JF, Hernandez-Ronquillo L, Tapia-Rangel B, et al. Hepatocarcinoma variedad fibrolamelar en una joven de 17 anos de edad. Gac Med Mex 135: 83-87, 1999. Chedid A, Ryan LM, Dayal Y, et al: Morphology and other prognostic factors of hepatocellular carcinoma. Arch Pathol Lab Med 123:524-8, 1999. Wood WJ, Raelings M, Evans H, et al: Hepatocellular carcinoma: importance of histologic classification as a prognostic factor. Am J Surg 155: 663-666, 1988. Yoshida K, Amemiya A, Kobayashi S, et al. Fibrolamellar carcinoma of the liver in the Orient. J Surg Oncol 39: 187-189, 1988. Kearney D, Donlon JB, Mendelson RM: A case of fibrolamellar hepatocelullar carcinoma. Australas Radiol 35: 88-91, 1991. Barsu M: Fibrolamellar hepatocarcinoma: case report and review of the literature. Rom J Morphol Embryol 37:161-166, 1991. Tanaka J, Baba N, Aru S, et al: Typical fibrolamellar hepatocellular carcinoma in Japanese patients: report of two cases. Surg Today 24: 459-463, 1994. Kaczynski J, Gustavsson B, Hansson G, et al. Fibrolamellar hepatic carcinoma in an area with a iow incidence of primary liver cancer: a retrospective study. Eur J Surg 162: 367-371, 1996. Lemmer ER, Krige JE, Hall PM, et al: Surgical resection for hepatocelullar carcinoma in Cape Town- a clinical and histopathoiogical study. S Afr Med J 88: 1575-1580, 1998. Yamaguchi R, Tajika T, Kanda H, et al: Fibrolamellar carcinoma of the liver. Hepatogastroenterology 46: 1706-1709, 1999. Ichikawa T, Federle MP, Graziou L, et al. Fibrolamellar hepatocellular carcinoma: imaging and pathologic findings in 31 recent cases. Radiology 213: 352-361, 1999. Kufkarni MS, Mathur SK, Nagral SS, et al: Fibrolamellar hepatocellular carcinoma. Indian J Gastroenterol 13: 148-149, 1994. Arslan I, Oner K, Kilicturgay S. et al: Fibrolamellar hepatocellular carcinoma. “It is not a ordinary case for Mediterranean countries”. HPB Surg 9: 51-53, 1995. Ashraf SJ. Arya SC, el-Sayed M, et al. A profile of primary hepatocellular carcinoma patients in the Gizan Area of Saudi Arabia. Cancer 58:2163-2168, 1986. Lapis K, Schaff Z, Kopper L, et al: Fibrolamellar liver carcinoma. Zentralbl Allg Pathol 136: 135-149, 1990. Van Leeuwen DJ, Bos RJ, Vidacovic-Vucic MM: Hepatocellular carcinoma in the Amsterdam area. A retrospective analysis in 61 patients. Scand J Gastroenterol Suppl 188:108-17, 1991. Yuan CY, Yuan CC, Shiou GF, et al: Fibrolamellar variant of hepatocellular carcinoma-report of a Chinese patient. Hepatogastroenterology 42:182-184, 1995. Haratake J, Horie A, Lee SD, et al: Fibrolamellar carcinoma of the fiver in a middle-aged Korean man. J UOEH 12:349-54, 1990. Andreola S, Audisio RA, Lombardi L: A light microscopic and ultrastructural study of two cases of fibrolamellar hepatocellular carcinoma. Tumori 72: 609-6016. 1986. Vlalondra AB, Picornell MF, Pons JT, et al: European cases of fibrolamellar hepatocellular carcinoma., Letter). Ann Intern Med 110:324, 1989. Caballero T, Aneiros J, Lopez-Caballero J, et al: Fibrolamellar hepatocellular carcinoma: an immunohistochemical and ultrastructuraf study. Histopathology 9:445-456, 1985. Klompmaker IJ, Haagsma EB, Verwer R, et al: Fibrolamellar hepatoma in Europe., Letter). Am J Gastroenterol 86:790-791, 1991. Guix Pericas M, Sancho Poch F, Pladellorens Roca M, et al: Hepatocarcinoma fibrolamelar: presentacion de dos casos. Gastroenterol Hepatol 6:475-479, 1983. Vecchio FM, Fabiano A, Ghirlanda G, et al. Fibrolamellar carcinoma of the liver: the malignant counterpart of focal nodular hyperplasia with oncocytic change. Am J Clin Pathol 81:521- 526, 1984. Soreide O, Czerniak A, Bradpiece H, et al. Characteristics of fibrolamellar hepatocellular carcinoma. A study of nine cases and a review of the literature: Am J Surg 151:518-523, 1986. Bore VB, Chasle J, Raffy P, et al: Le carcinoma fibrofamillaire du fori, a propos dune observation. Arch Anat Cytol Pathol 30:109-112, 1982. Ratia-Gimenez T, Herrera CJ, Pereira PF, et al: Liver transplant in fibrolamellar hepatocarcinoma. A case report and review of the literature. Rev Esp Enferm Apar Dig 75:273-276, 1989. Kahle M, Heilman KL, Filler RD:Das fibrolamellare leberzellkarzinom Ein failbericht. Z Gastroenterol 27:341-2, 1989. Zheng ZZ: Fibrolamellar carcinoma of the liver: Clinicopathologic analysis of six cases. Ching Hua Ko Tsa Chih 26:617- 619, 1988. Sifva H, Leon G, Naquira N: Fibrolamellar hepatocellular carcinoma. Report of four cases. Rev Med Chile 116:153-156, 1988. Berman MA, Burnham JA, Sheahan DG: Fibrolamellar carcinoma of the liver. An immunohistochemical study of nineteen cases and a review of the literature. Human Pathol 19:784-94, 1988. Dunk AA, Spuiadis H, Sherlock S, et al: Hepatocellular carcinoma: cfinicaf, aetio4ogical and pathological features in British patients. Int J Cancer 41:17-23, 1988. Kahno H, Nagasue N, Taniura H, et al. Fibrolamellar carcinoma of the liver-a case report from Japan and a review of the literature. HPB Surg 1:77-80, 1988. Eckstein RP, Bambach CP; Stiel D, et al: Fibrolamellar carcinoma as a cause of bile duct obstruction. Pathology 20:326-31, 1988. Azais-Nobunski B; Dahan D, Tubiana JM: Fibrolamellar carcinoma of the liver. Apropos of 2 cases. Review of the literature. Ann Radiol, Paris, 30:395-399, 1897. Grases PJ, Ruiz MC, Aparcero M, et al. Fibrolamellar carcinoma of the liver apropos of a case with successful resection. GEN 40:75-82, 1986. 40. Perez Blanco FJ, Urbano Jimenez FJ, Caballero Morates T, et al: A new case of fibrofamellar hepatocarcinoma. Rev Esp Enfer Apar Dig 67:551-555, 1985. 41. Denis J, Grippon P, Legendre C, et al: Fibrolamellar carcinoma of the liver: hepatocellular carcinoma with favorable prognosis. Gastroenterol Clin Biol 8:920-924, 1984. 42. Schechter D, Shefer R, Yosipov J, et al. Fibrolamellar carcinoma of the liver. Nareua 113:160-1, 1987. 43. Eyskens E, Van der Stighelen Y, Bourgeois N: Icterogenic hepatocellular carcinoma and polygonal cell carcinoma with fibrous stroma, fibrolamellar hepatocarcinoma). Acta Chir Belg 87:19-25, 1987. 44. Berman MM, Libbey NP, Foster JH: Hepatocelullar carcinoma polygonal cell type with fibrous stroma – an atypical variant with a favorable prognosis. Cancer 46:1448-1455, 1980 45. Borges AM. Swaroop VS, Desai PB: Response to Dr. Ruffin., Letter). Am J Gastroenterol 86:789-790, 1991. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 133 EP 137 PG 5 ER PT J AU Demokan, S Muslumanoglu, M Yazici, H Igci, A Dalay, N AF Demokan, Semra Muslumanoglu, Mahmut Yazici, Hulya Igci, Abdullah Dalay, Nejat TI Investigation of Microsatellite Instability in Turkish Breast Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microsatellite instability; breast cancer ID microsatellite instability; breast cancer AB Multiple somatic and inherited genetic changes that lead to loss of growth control may contribute to the development of breast cancer. Microsatellites are tandem repeats of simple sequences that occur abundantly and at random throughout most eucaryotic genomes. Microsatellite instability (MI), characterized by the presence of random contractions or expansions in the length of simple sequence repeats or microsatellites, is observed in a variety of tumors. The aim of this study was to compare tumor DNA fingerprints with constitutional DNA fingerprints to investigate changes specific to breast cancer and evaluate its correlation with clinical characteristics. Tumor and normal tissue samples of 38 patients with breast cancer were investigated by comparing PCR-amplified microsatellite sequences D2S443 and D21S1436. Microsatellite instability at D21S1436 and D2S443 was found in 5 (13%) and 7 (18%) patients, respectively. Two patients displayed instability at both marker loci. No association was found between MI and age, family history, lymph node involvement and other clinical parameters. C1 [Demokan, Semra] Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. [Muslumanoglu, Mahmut] Istanbul Faculty of Medicine, Istanbul University, Department of SurgeryIstanbul, Turkey. [Yazici, Hulya] Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. [Igci, Abdullah] Istanbul Faculty of Medicine, Istanbul University, Department of SurgeryIstanbul, Turkey. [Dalay, Nejat] Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. RP Dalay, N (reprint author), Oncology Institute, Department of Basic Oncology, 34390 Capa-Istanbul, Turkey. EM ndalay@yahoo.com CR Dickson RB, Lippman ME. Molecular basis of breast cancer. In: The Molecular Basis of Cancer, WB Saunders, 358-384, 1995. Hovig E, Smith-Sorensen B, Uitterlinden AG, et al: Detection of DNA variation in cancer. Pharmacogenetics 2:317-328, 1992. Paulson TG, Wright FA, Parker BA, et al: Microsatellite instability correlates with reduced survival and poor disease prognosis in breast cancer. Cancer Res 56:4021-4026, 1996. Ali S, Muller CR, Epplen JT. DNA fingerprinting by oligonucleotide probes specific for simple repeats. Hum Genet 74:239- 243, 1986. Decker RA, Moore J, Ponder B, et al: Linkage mapping of human chromosome 10 microsatellite polymorphisms. Genomics 12:604-606, 1992. Weber JL, Wong C. Mutation of human short tandem repeats. Hum Mol Genet 2:1123-1128, 1993. Branch P, Aquilina G, Bignami M, et al: Defective mismatch binding and a mutator phenotype in cells tolerant to DNA damage. Nature 362:652-654, 1993. Fey MF, Wells RA, Wainscoat JS, et al: Assessment of clonality in gastrointestinal cancer by DNA fingerprinting. J Clin Invest 82:1532-1537, 1988. Jeffreys AJ, Neumann R, Wilson V. Repeat unit sequence variation in minisatellites: A novel source of DNA polymorphism for studying variation and mutation by single molecule analysis. Cell 60:473-485, 1990. Samowitz WS, Curtin K, Ma KN, et al: Microsatellite instability in sporadic colon cancre is associated with an improved prognosis at the population level. Cancer Epidem Biomarkers Prev 10:917-923, 2001. van Rhijn BWG, Lurkin I, Kirkels WJ, et al: Microsatellite analysis – DNA test in urine competes with cystoscopy in follow- up of superficial bladder carcinoma. Cancer 92:768-775, 2001. Han HJ, Yanagisawa A, Kato Y, et al: Genetic instability in pancreatic cancer and poorly differentiated type of gastric cancer. Cancer Res 53:5087-5089, 1993. Gonzalez R, Silva JM, Sanchez A, et al: Microsatellite alterations and TP53 mutations in plasma DNA of small cell lung cancer patients: Follow-up study and prognostic significance. Annals Oncol 11:1097-1104, 2000. Contegiacomo A, Palmirotta R, De Marchis L, et al: Microsatellite instability and pathological aspects of breast cancer. Int J Cancer 64:264-268, 1995. Vaurs-Barriere C, Penault-Llorca F, Laplace-Marieze V, et al: Low frequency of microsatellite instability in BRCA1 mutated breast tumors. J Med Genet 37:1-3, 2000. Chen P, Hurst T, Khoo SK. Detection of somatic DNA alterations in ovarian cancer by DNA fingerprint analysis. Cancer 67:1551-1555, 1991. Fearon ER, Vogelstein B. Tumor suppressor and DNA repair gene defects in human cancer. In : Holland JF, Bast RC, Morton DL, Frei III E, Kufe DW, Weichselbaum RR, eds). Cancer Medicine. 4. ed. Williams and Wilkins, 97-119, 1997. Alexander J, Watanabe T, Wu TT, et al: Histopathological identification of colon cancer with microsatellite instability. Am J Pathol 158:527-535, 2001. Seripa D, Parrella P, Gallucci M, et al: Sensitive detection of transitional cell carcinoma of the bladder by microsatellite analysis of cells exfoliated in urine. Int J Cancer 95:364-369, 2001. Rha SH, Dong SM, Jen J, et al: Molecular detection of cervical intraepithelial neoplasia and cervical carcinoma by microsatellite analysis of Papanicolaou smears. Int J Cancer 93:424-429, 2001. Gonzalez-Zulueta M, Ruppert JM, et al: Microsatellite instability in bladder cancer. Cancer Res 53:5620-5623, 1993. Sourvinos G, Kiaris H, Tsikkinis A, et al: Microsatellite instability and loss of heterozygosity in primary breast tumors. Tumor Biol 18:157-166, 1997. Thibodeau SN, Bren G, Schaid D. Microsatellite instability in cancer of the proximal colon. Science 260:816-819, 1993. Rush EB, Calvano JE, Van Zee KJ, et al: Microsatellite instability in breast cancer. Ann Surg Oncol 4:310-315, 1997. Shaw JA, Walsh T, Chappell SA, et al: Microsatellite instability in early sporadic breast cancer. Br J Cancer 73:1393-1397, 1996. Kasami M, Vnencak-Jones CL, Manning S, et al: Loss of heterozygosity and microsatellite instability in breast hyperplasia. Am J Pathol 150:1925-1931, 1997. Krajinovic M, Richer C, Gorska-Flipot I, et al: Genomic loci susceptible to replication errors in cancer cells. Br J Cancer 78:981-985, 1998. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 138 EP 141 PG 4 ER PT J AU Cserni, G Kovacs, RB Tarjan, M Sapi, Z Domjan, Zs Szabo, Z AF Cserni, Gabor Kovacs, Rita Beata Tarjan, Miklos Sapi, Zoltan Domjan, Zsolt Szabo, Zoltan TI Sarcomatoid Renal Cell Carcinoma with Foci of Chromophobe Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE chromophobe renal cell carcinoma; sarcomatoid renal carcinoma; DNA-ploidy; p53 ID chromophobe renal cell carcinoma; sarcomatoid renal carcinoma; DNA-ploidy; p53 AB Both chromophobe carcinoma and sarcomatoid carcinoma of the kidney are rare. The former is characterized by a relatively good prognosis, while the latter is a highly aggressive tumor. Coexistence of the two components in one renal tumor, which has been reported only rarely, is therefore paradoxical. Both sarcomatoid and chromophobe renal carcinoma were diagnosed in a 52-year-old woman following nephrectomy and resection of metastases in the right lobe of the liver. She died of the disease two months after the first operation; only the sarcomatoid component of her tumor was seen in the liver metastasis and the recurrent carcinoma. Differences in phenotype, immunophenotype and DNA-ploidy patterns of the two components are reported. The intensive p53 staining observed only in the sarcomatoid area supports the role of the TP53 gene in the transformation of chromophobe renal carcinoma to sarcomatoid carcinoma. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., H-6000 Kecskemet, Hungary. [Kovacs, Rita Beata] St John's Hospital, Department of PathologyBudapest, Hungary. [Tarjan, Miklos] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., H-6000 Kecskemet, Hungary. [Sapi, Zoltan] St John's Hospital, Department of PathologyBudapest, Hungary. [Domjan, Zsolt] Bacs-Kiskun County Teaching Hospital, Department of UrologyKecskemet, Hungary. [Szabo, Zoltan] Bacs-Kiskun County Teaching Hospital, Department of UrologyKecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.c3.hu CR Akhtar M, Kardar M, Linjawi T, et al: Chromophobe cell carcinoma of the kidney: a clinicopathologic study of 21 cases. Am J Surg Pathol 19:1245-1256, 1995. Akhtar M, Kfoury H, Kardar A, et al: Sarcomatoid chromophobe cell carcinoma of the kidney. J Urol Pathol 4:155-166, 1996. Akhtar M, Tulbah A, Kardar AH, Ali MA: Sarcomatoid renal carcinoma: the chromophobe connection. Am J Surg Pathol 21:1188-1195, 1997. Bertoni F, Ferri C, Benati A, et al: Sarcomatoid carcinoma of the kidney. J Urol 137:25-28, 1987. Crotty TB, Farrow GM, Liber MM: Chromophobe cell renal carcinoma, clinicopathological features of 50 cases. J Urol 154:964-967, 1995. Gomez-Roman JJ, Mayorga-Fernandez M, Mayorga-Fernendez F, Val-Bernal JF: Sarcomatoid chromophobe cell renal carcinoma: immunohistochemical and lectin study in one case. Gen Diagn Pathol 143:63-69, 1997. Hirokawa M, Shimizu M, Sakurai T, et al: Sarcomatoid renal carcinoma with chromophobe cell foci. Report of a case. APMIS 106:993-996, 1998. Kovacs G: Molecular differential pathology of renal cell tumors Histopathology 22:1-8, 1993. Kuroda N, Hayashi Y, Itoh H: A case of chromophobe renal cell carcinoma with sarcomatoid foci and a small daughter lesion. Pathol Int 48:812-817, 1998. Mai KT, Veinot JP, Collins JP: Sarcomatous transformation of chromophobe cell renal carcinoma. Histopathology 34:557- 559, 1999. Murphy WM, Beckwith JB, Farrow GM: Atlas of tumor pathology. Tumors of the kidney, bladder, and related structures. Third series. Armed Forces Institute of Pathology, Washington, 1994. Renshaw AA, Henske EP, Loughlin KR, et al: Aggressive variants of chromophobe renal cell carcinoma. Cancer 78:1756- 1761, 1996. Ro JY, Ayala AG, Sella A, et al: Sarcomatoid renal cell carcinoma: clinicopathologic study of 42 cases. Cancer 59:516-526, 1987. Tardio JC: Chromophobe cell renal carcinomas with sarcomatoid areas: Histopathology 35:184-185, 2000. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 142 EP 144 PG 3 ER PT J AU Kabukcuoglu, F Baksu, A Yilmaz, B Aktumen, A Evren, I AF Kabukcuoglu, Fevziye Baksu, Alparslan Yilmaz, Banu Aktumen, Alpay Evren, Ismail TI Malignant Struma Ovarii SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Ovary; malignant struma ovarii; germ cell tumor ID Ovary; malignant struma ovarii; germ cell tumor AB Malignant struma ovarii is a rare form of ovarian germ cell tumors. Because of its rarity, diagnosis and management of the tumor has not been clearly defined. A 52-year-old female with follicular variant of papillary carcinoma arising in struma ovarii is presented. There was no evidence of clinical malignancy or metastases. However on the basis of histopathologic findings, the case was diagnosed as malignant struma ovarii. It was accepted as stage Ia tumor and no other treatment was considered after radical excision. Due to its fairly good prognosis, surgical excision has been the preferred treatment for those who do not have disseminated disease. Nevertheless, long-term follow-up is necessary to determine the course of the disease. C1 [Kabukcuoglu, Fevziye] Etfal Training and Research Hospital, Pathology Department, M. Ismail Hakki sk No: 10/1 Dogancilar, 81160 Uskudar, Istanbul, Turkey. [Baksu, Alparslan] Etfal Training and Research Hospital, Pathology Department, M. Ismail Hakki sk No: 10/1 Dogancilar, 81160 Uskudar, Istanbul, Turkey. [Yilmaz, Banu] Etfal Training and Research Hospital, Pathology Department, M. Ismail Hakki sk No: 10/1 Dogancilar, 81160 Uskudar, Istanbul, Turkey. [Aktumen, Alpay] Etfal Training and Research Hospital, Pathology Department, M. Ismail Hakki sk No: 10/1 Dogancilar, 81160 Uskudar, Istanbul, Turkey. [Evren, Ismail] Etfal Training and Research Hospital, Pathology Department, M. Ismail Hakki sk No: 10/1 Dogancilar, 81160 Uskudar, Istanbul, Turkey. RP Kabukcuoglu, F (reprint author), Etfal Training and Research Hospital, Pathology Department, 81160 Uskudar, Istanbul, Turkey. EM etfaly@superonline.com CR Ayhan A, Yany k F, Tuncer R, et al:Struma ovarii. Int J Gynecol Obstet 42:143-146, 1993. Berghella V, Ngadiman S, Rosenberg H, et al: Malignant struma ovarii. A case report and review of the literature. Gynecol Obstet Invest 43:68-72; 1997. Dardik RB, Dardik M, Westra W, Montz FJ: Malignant struma ovarii: Two case reports and a review of the literature. Gynecol Oncol 73:447-451, 1999. Devaney K, Snyder R, Norris HJ, Tavassoli FA: Proliferative and histologically malignant struma ovarii: a clinicopathologic study of 54 cases. Int J Gynecol Pathol 12:333-343, 1993. Gonzales A, Kaufman RH, Braungardt CD, et al: Adenocarcinoma of thyroid arising in struma ovarii, malignant struma ovarii). Report of two cases and review of the literature. Obstet Gynecol 21:567-576, 1963. Karseladze AI, Kulinitch SI: Peritoneal strumosis. Path Res Pract 190:1082-1085, 1994. Nogales FF: Germ cell tumours of the ovary, In: Obstetrical and Gynecological Pathology., Eds: Fox H and Wells M): 4th ed, Churchill Livingstone, New York, 1995, pp 847-896. O’Connell MEA, Fisher C, Harmer CL: Malignant struma ovarii: presentation and management. Br J Radiol 63:360-363, 1990. Pardo-Mindan FJ, Vazquez JJ: Malignant struma ovarii Light and electron microscopic study. Cancer 51:337-343, 1983. Rose PG, Arafah B, Abdul-Karim FW: Malignant struma ovarii: recurrence and response to treatment monitored by thyroglobulin levels. Gynecol Oncol 70:425-427, 1998. Szyfelbein WM, Young RH, Scully RE:Struma ovarii simulating ovarian tumors of other types. A report of 30 cases. Am J Surg Pathol 19(1): 21-29, 1995. Talerman A: Germ cell tumors of the ovary: In Blaustein’s Pathology of the Female Genital Tract., Ed: Kurman RJ), 4th ed, Springer-Verlag, New York 1994, pp 849-914. Thomas RD, Batty VB: Metastatic malignant struma ovarii. Two case reports. Clin Nucl Med 17:577-578. Vadmal MS, Smilari TF, Lovecchio JL, et al: Diagnosis and treatment of disseminated struma ovarii with malignant transformation. Gynecol Oncol 64:541-546, 1997. Willemse PHB, Oosterhuis JW, Aalders JG, et al: Malignant struma ovarii treated by ovariectomy, thyroidectomy, and 131I administration. Cancer 60:178-182, 1987. Woodruff JD, Rauh T, Markley RL: Ovarian struma. Obstet Gynecol 27:194-201, 1966. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 145 EP 147 PG 3 ER PT J AU Agarwal, N Shatma, CM Deol, P Mehta, SV Sarkar, Ch AF Agarwal, M. Nitesh Shatma, C M Deol, S. Parminder Mehta, Singh Veer Sarkar, Chitra TI Epithelioid Sarcoma of the Sciatic Nerve Perineural Sheath: a Mimic of Nerve Sheath Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE epithelioid sarcoma; sciatic nerve; soft tissue tumor; sarcoma ID epithelioid sarcoma; sciatic nerve; soft tissue tumor; sarcoma AB We report herein a rare case of epithelioid sarcoma, in a 39-year old lady involving the sciatic nerve. Clinically and radiologically it stimulated a nerve sheath tumor. Involvement of a nerve by an epithelioid sarcoma is extremely uncommon. To the best of our knowledge, this is the first case of an epithelioid sarcoma involving the sciatic nerve and needs documentation. C1 [Agarwal, M. Nitesh] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Shatma, C M] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Deol, S. Parminder] All India Institute of Medical Sciences, Department of NeuroradiologyNew Delhi, India. [Mehta, Singh Veer] All India Institute of Medical Sciences, Department of NeurosurgeryNew Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. RP Sarkar, Ch (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM sarkarcs@hotmail.com CR Enzinger FM: Epithelioid sarcoma. A sarcoma simulating a granuloma or a carcinoma. Cancer 26:1029-1041, 1970. Laskowski J: Sarcoma aponeuroticum. Nowotwary 11: 61-67, 1961. Enzinger FM, Weiss SW: Soft tissue tumors, 3rd Ed. Mosby, St. Louis, Baltimore, New York pp 1074-1083; 1995. Prat J, Woodruff JM, Matcove RC: Epithelioid sarcoma: An analysis of 22 cases indicating the prognostic significance of vascular invasion and regional lymph node metastasis. Cancer 41:1472-1487, 1978. Chase DR, Enzinger FM: Epithelioid sarcoma: Diagnosis, prognostic indicators and treatment. Am J Surg Pathol 9:241- 263, 1985. Bos GD, Pritchard DJ, Reiman HM, et al: Epithelioid sarcoma. An analysis of fifty-one cases. J Bone Joint Surg 70:862-874, 1988. Halling AC, Wollen PC, Pritchard DJ: Epithelioid sarcoma. A clinicopathological review of 55 cases. Mayo Clin Proc 71:636-642, 1996. Bryan RS, Soule EH, Dobyns JH, et al: Primary epithelioid sarcoma of the hand and forearm. A review of thirteen cases. J Bone Joint Surg 56:458-465, 1974 Brennan MF. Soft tissue sarcoma involving the sciatic nerve. Brit J Surg 87:993, 2000. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 148 EP 150 PG 3 ER PT J AU Kaya, H Aribal, E Yegen, C AF Kaya, Handan Aribal, Erkin Yegen, Cumhur TI Apocrine Differentiation in Invasive Pleomorphic Lobular Carcinoma with In Situ Ductal and Lobular Apocrine Carcinoma: Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE lobular carcinoma; pleomorphic lobular carcinoma; apocrine differentiation ID lobular carcinoma; pleomorphic lobular carcinoma; apocrine differentiation AB Invasive pleomorphic lobular carcinoma (PLC) is a distinctive aggressive subtype of invasive lobular carcinomas (ILC). We report one case of PLC with in situ PLC and ductal carcinoma in situ with apocrine features. C1 [Kaya, Handan] Marmara University, School of Medicine, Department of Pathology, Altunizade, 81190 Istanbul, Turkey. [Aribal, Erkin] Marmara University Hospital, Department of RadiologyIstanbul, Turkey. [Yegen, Cumhur] Marmara University, School of Medicine, Department of General SurgeryIstanbul, Turkey. RP Kaya, H (reprint author), Marmara University, School of Medicine, Department of Pathology, 81190 Istanbul, Turkey. EM hkaya@superonline.com CR Damiani S, Dina R, Eusebi V: Eosinophilic and granular cell tumors of the breast. Semin Diag Pathol 16:117-125, 1999. Eusebi V, Magalhaes F, Azzopardi JG: Pleomorphic lobular carcinoma of the breast: An aggressive tumor showing apocrine differentiation. Hum Pathol 23:655-662, 1992. Losi L, Lorenzini P, Bussolati G: Apocrine differentiation in invasive carcinomas of the breast. Comparison of monoclonal and polyclonal gross cystic disese fluid protein 15 with prolactin inducible protein in RNA gene expression. Appl Immunohistochem 3:91-98, 1995. Middleton LP, Palacios DM, Bryant BR, et al: Pleomorphic lobular carcinoma: morphology, immunohistochemistry, and molecular analysis. Am J Surg Pathol 24:650-1656, 2000. Radhi JM: Immunohistochemical analysis of pleomorphic lobular carcinoma: higher expression of p53 and chromogranin and lower expression of ER and PgR. Histopath 36:156-160, 2000. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD AUG PY 2002 VL 8 IS 2 BP 151 EP 152 PG 2 ER PT J AU Pisch, J Moskovitz, T Esik, O Homel, P Keller, S AF Pisch, Julianna Moskovitz, Tibor Esik, Olga Homel, Peter Keller, Steven TI Concurrent Paclitaxel-Cisplatin and Twice-a-DayIrradiation in Stage IIIA and IIIB NSCLC Shows Improvement in Local Control and Survival with Acceptable Hematologic Toxicity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article AB Non-small-cell lung cancer (NSCLC) has one of the highest death rates among the various forms of cancer. In attempts to improve on this unsatisfactory outcome, different radiation schedules and chemo-therapy agents have been examined in phase II or III studies. These have led to modest improvements in local control and survival, but combined therapies are associated with substantial hematologic toxicity. In this phase II study, 80 consecutive stage IIIA or IIIB NSCLC patients were treated with concomitant chemotherapy and twice-a-day irradiation in a total dose of 60 Gy in 1.5 Gy fractions. Patients scheduled for surgery received 45 Gy only. Paclitaxel (30 mg/m 2 ) on days 1-4 and cisplatin (100 mg/m 2 ) on day 5 were administered in the first and fourth weeks of treatment. Granulocyte colony stimulating factor (30 ng/m 2 ) was given on days 10-15. The local control, the 1- and 2-year survival rates and the occurrence of acute hematologic toxicity in the non-surgically treated patients were examined. Fifty-two patients were treated without and 28 with surgery. Among the non-surgically treated cases, 43 were evaluable for response and 47 for acute toxicity during a median follow-up of 22 months. The rate of local control was 65% (28/43), and the 1- and 2-year survival rates proved to be 68% and 48%, respectively, with a median survival of 28 months. Severe acute grade 3-4 toxicities included grade 4 leukopenia in 6 cases (13%), grade 3 leukopenia in 4 cases (9%), grade 3 esophagitis in 3 cases (6%) and grade 3 anemia in 3 cases (6%). Our results and the relevant data from the literature support the application of twice-a-day irradiation with concomitant chemotherapy in stage IIIA and IIIB NSCLC. Local control and survival were improved relative to once-a-day irradiation with sequential or concomitant chemotherapy. C1 [Pisch, Julianna] Beth Israel Medical Center, Department of Radiation Oncology, First Avenue at 16 Street, NY 10003 New York, USA. [Moskovitz, Tibor] Beth Israel Medical Center, Department of Medical OncologyNew York, USA. [Esik, Olga] Beth Israel Medical Center, Department of BiostatisticsNew York, USA. [Homel, Peter] Semmelweis University, Department of OncologyBudapest, Hungary. [Keller, Steven] Montefiore Medical Center, Albert Einstein College of Medicine, Department of Thoracic SurgeryNew York, USA. RP Pisch, J (reprint author), Beth Israel Medical Center, Department of Radiation Oncology, NY 10003 New York, USA. EM JPisch@bethisraelny.org CR Arriagada R, Le Chevalier T, Quoix E, et al: ASTRO Plenary: Effect of chemotherapy on locally advanced non-small cell lung carcinoma: a randomized study of 353 patients. GETCB, Groupe d’Etude et Traitment des Cancers Bronchiques), FNCLC, Federation Nationale des Centres de Lutte contre le Cancer, and the CEBI trialists. Int J Radiat Oncol Biol Phys 20:1183-1190, 1991 Byhardt RW, Pajak TF, Emami B, et al: A phase I/II study to evaluate accelerated fractionation via concomitant boost for squamous, adeno, and large cell carcinoma of the lung: report of Radiation Therapy Oncology Group 84-07. Int J Radiat Oncol Biol Phys 26:459-468, 1993 Byhardt RW, Scott CB, Ettinger DS, et al: Concurrent hyperfractionated irradiation and chemotherapy for unresectable nonsmall cell lung cancer. Results of Radiation Therapy Oncology Group 90-15. Cancer 75:2337-2344, 1995 Cancer Therapy Evaluation Program. Common Toxicity Criteria, Version 2.0., 1997, http://ctep.info.nih.gov Choy H, Devore RF III, Hande KR, et al: A phase II study of paclitaxel, carboplatin, and hyperfractionated radiation therapy for locally advanced inoperable non-small-cell lung cancer, a Vanderbilt Cancer Center Affiliate Network Study). Int J Radiat Oncol Biol Phys 47:931-937, 2000 Clamon G, Herndon J, Cooper R, et al: Radiosensitization with carboplatin for patients with unresectable stage III nonsmall- cell lung cancer: a phase III trial of the Cancer and Leukemia Group B and the Eastern Cooperative Oncology Group. J Clin Oncol 17: 4-11, 1999 Cox JD, Azarnia N, Byhardt RW, et al: A randomized phase I/II trial of hyperfractionated radiation therapy with total doses of 60.0 Gy to 79.2 Gy: possible survival benefit with greater than or equal to 69.6 Gy in favorable patients with Radiation Therapy Oncology Group stage III non-small-cell lung carcinoma: report of Radiation Therapy Oncology Group 83-11. J Clin Oncol 8:1543-55, 1990 Cox JD, Azarnia N, Byhardt RW, et al: Altered fractionation for non-small-cell carcinoma of the lung. Chest 96:68S-69S, 1999 Dillman RO, Seagren SL, Propert KJ, et al: A randomized trial of induction chemotherapy plus high-dose radiation versus radiation alone in stage III non-small-cell lung cancer. N Engl J Med 323: 940-945, 1990 Greenlee R, Hill-Harmon MB, Murray T, Thun M: Cancer statistics, 2001. CA Cancer J Clin 51:15-36, 2001 Hazuka MB, Crowley JJ, Bunn PA Jr, et al: Daily low—dose cisplatin plus concurrent high-dose thoracic irradiation in locally advanced unresectable non-small-cell lung cancer: results of a phase II Southwest Oncology Group Study. J Clin Oncol 12:1814-1820, 1994 International Commission on Radiation Units and Measurements. ICRU Report 50. Prescribing, recording, and reporting photon beam therapy. Bethesda, MD, 1993 International Commission on Radiation Units and Measurements, eds: Wambersie A, Landberg T, ICRU Report 62. Prescribing, recording, and reporting photon beam therapy, Supplement to ICRU Report 50, Bethesda, MD, 1999 Jeremic B, Shibamoto Y, Acimovic L, et al:Hyperfractionated radiation therapy and concurrent low-dose, daily carboplatin/etoposide with or without weekend carboplatin/etoposide chemotherapy in stage III non-small-cell lung cancer: a randomized trial. Int J Radiat Oncol Biol Phys 50: 19-25, 2001 Kaplan EL, Meier P: Nonparametric estimation from incomplete observations. J Am Stat Ass 54: 457-481, 1958 Kelly K, Hazuka M, Pan Z, et al: A phase I study of daily carboplatin and simultaneous accelerated, hyperfractionated chest irradiation in patients with regionally inoperable nonsmall cell lung cancer. Int J Radiat Oncol Biol Phys 40:559- 567, 1998 Komaki R, Scott C, Ettinger D, et al: Randomized study of chemotherapy/radiation therapy combinations for favorable patients with locally advanced inoperable nonsmall cell lung cancer: Radiation Therapy Oncology Group, RTOG, 92-04. Int J Radiat Oncol Biol Phys 38:149-155, 1997 Langer CJ, Curran WJ, Keller SM, et al: Report of phase II trial of concurrent chemoradiotherapy with radical thoracic irradiation, 60 Gy), infusional fluorouracil, bolus cisplatin and etoposid for clinical stage IIIB and bulky IIIA non-small cell lung cancer. Int J Radiat Oncol Biol Phys 26: 469-478, 1993 Martel MK, Ten Haken RK, Hazuka MB, et al: Estimation of tumor control probability model parameters from 3D dose distributions of non-small cell lung cancer patients. Lung Cancer 24: 31-37, 1999 Mehta MP, Tannehill SP, Adak S, et al: Phase II trial of hyperfractionated accelerated radiation therapy for nonresectable non-small-cell lung cancer: results of Eastern Cooperative Oncology Group 4593. J Clin Oncol 11: 3518-3523, 1998 Perez CA, Pajak TF, Rubin P, et al: Long-term observations of the patterns of failure in patients with unresectable non-oat cell carcinoma of the lung treated with definitive radiotherapy. Report by the Radiation Therapy Oncology Group. Cancer 59: 1874-1881, 1987 Pisch J, Berson AM, Malamud S, et al: Chemoradiation in advanced nonsmall cell lung cancer. Int J Radiat Oncol Biol Phys 33: 183-188, 1995 Saunders M, Dische S, Barrett A, et al: Continuous hyperfractionated accelerated radiotherapy, CHART, versus conventional radiotherapy in non-small-cell lung cancer: a randomised multicenter trial. CHART Steering Committee. Lancet 350:161-165, 1997 Sause WT, Scott C, Taylor S, et al: Phase II trial of combination chemotherapy and irradiation in non-small-cell lung cancer, Radiation Therapy Oncology Group 88-04. Am J Clin Oncol 15:163-167, 1992 Sause W, Kolesar P, Taylor S IV, et al: Final results of phase III trial in regionally advanced unresectable non-small cell lung cancer: Radiation Therapy Oncology Group, Eastern Cooperative Oncology Group, and Southwest Oncology Group. Chest 117:358-364, 2000 Schaake-Koning C, van den Bogaert W, Dalesio O, et al: Effects of concomitant cisplatin and radiotherapy on inoperable nonsmall- cell lung cancer. N Engl J Med 326: 524-530, 1992 Trott KR, Kummermehr J. The time factor and repopulation in tumors and normal tissues. Semin Radiat Oncol 3: 115-125, 1993 Withers HR, Taylor JM, Maciejewski B: The hazard of accelerated tumor clonogen repopulation during radiotherapy. Acta Oncol 27: 131-146, 1988 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2002 VL 8 IS 3 BP 163 EP 169 PG 7 ER PT J AU Khoursheed, M Mathew, Th Makar, RR Sonia, L H, A Asfar, S Al-Sayer, MH Dashti, H Al-Bader, A AF Khoursheed, Mousa Mathew, C. Thazhumpal Makar, Ray Ragai Sonia, L H, Abul Asfar, Sami Al-Sayer, Musaed Hilal Dashti, M. Hussain Al-Bader, Abdullatif TI Expression of CD44s in Human Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article AB CD44s is a cell adhesion molecule, which belongs to the family of hyaluronan binding proteins. Anti-body to CD44s is used to establish the association of its expression with the clinicopathological characteristics of colorectal cancer using immunohistochemical methods. The aim of this study is to investigate the expression of the standard form of CD44 (CD44s) in colorectal cancer tissues as compared to adjacent normal colonic tissues. Furthermore, the level of expression of CD44s in colorectal cancer tissues was correlated with the degree of histological differentiation, Duke’s classification, sex, size and site of the tumor. Immunohistochemical analysis for CD44s was carried out in 49 paraffin-fixed sections of neoplastic colorectal tissues and non-neoplastic ones adjacent to the lesion, by the standard peroxidase-antiperoxidase method. Expression of these antigens were compared in normal and malignant epithelium and stromal cells. The results show that the level of CD44s in the epithelial and stromal cells was significantly higher in the colorectal cancer tissues than the normal ones. However, there was no association between the percentages of expressions of CD44s and the degree of histological differentiation, Duke’s classification, sex or size of the tumor. There was however, a significantly higher expression of CD44s in the epithelium of rectal cancer than that of colonic cancer. This study indicates that the expression of CD44s is significantly higher in colorectal cancer tissues. However, further studies are required to understand its role in tumor progression and metastasis of this disease. C1 [Khoursheed, Mousa] Kuwait University, Faculty of Medicine, Department of Surgery, 13110 Safat, Kuwait. [Mathew, C. Thazhumpal] Kuwait University, Faculty of Medicine, Faculty of Allied Health SciencesSafat, Kuwait. [Makar, Ray Ragai] Kuwait University, Faculty of Medicine, Department of PathologySafat, Kuwait. [Sonia, L] Kuwait University, Faculty of Medicine, Department of Surgery, 13110 Safat, Kuwait. [H, Abul] Kuwait University, Faculty of Medicine, Faculty of Allied Health SciencesSafat, Kuwait. [Asfar, Sami] Kuwait University, Faculty of Medicine, Department of Surgery, 13110 Safat, Kuwait. [Al-Sayer, Musaed Hilal] Kuwait University, Faculty of Medicine, Department of Surgery, 13110 Safat, Kuwait. [Dashti, M. Hussain] Kuwait University, Faculty of Medicine, Department of Surgery, 13110 Safat, Kuwait. [Al-Bader, Abdullatif] Kuwait University, Faculty of Medicine, Department of PathologySafat, Kuwait. RP Khoursheed, M (reprint author), Kuwait University, Faculty of Medicine, Department of Surgery, 13110 Safat, Kuwait. EM khoursheed@hsc.kuniv.edu.kw CR Hart I, Goode N, Wilson R: Molecular aspects of metastatic cascade. Biochem Biophys Acta 989: 65-84, 1989 Screaton GR, Bell MV, Jackson DG, et al: Genomic structure of DNA encoding the lymphocyte homing receptor CD44 reveals at least 12 alternatively spliced axons. Proc Natl Acad Sci USA 89:12160-12164, 1992 Tolg C, Hofmann M, Herrlich P, Ponta H: Splicing choice from ten variant exons establishes CD44 variability. Nucleic Acids Res 21: 1225-1229, 1993 Sleeman J, Moll J , Sherman L, et al. The role of CD44 splice variants in human metastatic cancer: Cell adhesion and human disease. Chichester, Ciba Foundation Symposium 189), Wiley, pp 142-56, 1995 Bartolazzi A, Jackson D, Bennett K, et al:Regulation of growth and dissemination of a human lymphoma by CD44 splice variants. J Cell Sci 108:1723-1733, 1995 Gunthert U, Hofmann M, Rudy W, et al: A new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell 65: 13-24, 1991 Seiter S, Arch R, Reber S, et al: Prevention of tumor metastasis formation by anti-variant CD44. J Exp Med 177: 443-455, 1993 Haynes B, Telen M, Hale L, Denning S: CD44-a molecule involved in leukocyte adherence and T-cell activation. Immunol Today 10: 423-428, 1989 Ropponen KM, Eskelinen MJ, Lipponen PK, et al: Expression of CD44 and variant proteins in human colorectal cancer and its relevance for prognosis. Scan J Gastroenterol 33: 301-309, 1998 Trowbridge I, Lesly J, Schulte R, et al: Biochemical characterization and cellular distribution of a polymorphic, murine cellsurface glycoprotein expressed in lymphoid tissues. Immunogenetics 15: 299-312, 1982 Herrlich P, Pals S, Ponta H: CD44 in colon cancer. Eur J Cancer 31:1110-1112, 1995 Boland CR, Kim YS: Transitional mucosa of the colon and tumor growth factors. Med Hypotheses 22: 237-243, 1987 Fox S, Fawcett J, Jackson DG: Normal human tissues, in addition to some tumors, express multiple different CD44 isoforms. Cancer Res 54:4539-4546, 1994 Hamilton PW, Watt PC, Allen DC: A morphometric assessment of transitional mucosa in the colon. Histopathology 13: 519- 530, 1988 151.Isaacson P, Attwood PR: Failure to demonstrate specificity of the morphological and histochemical changes in mucosa adjacent to colonic carcinoma, transitional mucosa). J Clin Pathol 32: 214-218, 1979 16. Sugino T, Gorham H, Yoshida K, et al. Progressive loss of CD44 gene expression in invasive bladder cancer. Am J Path 149:873-882, 1996 17. Wielenga VJM, Heider KH, Offerhaus GJA, et al. Expression of CD44 variant protein in human colorectal cancer is related to tumor progression. Cancer Res 53: 4754-756, 1993 18. Herrlich P, Zoller M, Pals ST, Ponta H. CD44 slice variants: Metastases meet lymphocytes. Immunol Today 14: 395-399, 1993 19. Tanabe KK, Ellis LM, Saya H:Expression of CD44RI adhesion molecule in colon carcinomas and metastases. Lancet 341:725- 726, 1993 20. East J, Hart IR: CD44 and its role in tumor progression and metastasis. Eur J Cancer 29A: 1921-1922, 1993 21. Mulder JW, Kruyt PM, Sewnath M, et al: Colorectal cancer prognosis and expression of exon-v6-containing CD44 proteins. Lancet 344: 1470-1472, 1994 22. Koopman G, Heider KH, Horst E, et al. Activated human lymphocytes and aggressive non-Hodgkin’s lymphomas express a homologue of the rat metastasis-associated variant of CD44. J Exp Med 177: 897-904, 1993 23. Orzechowski HD, Beckenbach C, Herbst H, et al: Expression of CD44v6 is associated with cellular dysplasia in colorectal epithelial cells. Eur J Cancer 31A: 2073-2079, 1995 24. Mulder JW, Wielenga VJ, Polak MM, et al: Expression of mutant p53 protein and CD44 variant proteins in colorectal tumorigenesis. Gut 36:76-80 1995 25. Gotley DC, Fawcett J, Walsh MD, et al: Alternatively spliced variants of the cell adhesion molecule CD44 and tumor progression in colorectal cancer. Br J Cancer 74: 342-351, 1996 26. Abbasi AM, Forbes A, Chester KA, et al. Putatively metastatic specific CD44V6 is also expressed in normal colon and in human colorectal tumors. J Pathol 175(suppl): 137A, 1995 27. Imazeki F, Yokosuka O, Yamaguchi T, et al. Expression of variant CD44-massanger RNA in colorectal adenocarcinomas and adenomatous polyps in humans. Gastroenterology 110: 362- 368, 1996 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2002 VL 8 IS 3 BP 170 EP 174 PG 5 ER PT J AU Elpek, G Gelen, T Karpuzoglu, G Karpuzoglu, T Aksoy, HN Keles, N AF Elpek, Ozlem Gulsum Gelen, Tekinalp Karpuzoglu, Gulten Karpuzoglu, Tuncer Aksoy, Hikmet Nazif Keles, Nuran TI Clinicopathologic Evaluation of CDw75 Antigen Expression in Colorectal Adenocarcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article AB CDw75, a B lymphocyte surface antigen, is a sialylated carbohydrate epitope, which is generated by the enzyme b bgalactosyl a a2,6 sialyltransferase (Sia-T1). In colon carcinomas, although higher levels of Sia-T1 has been described and found to be correlated with metastatic potential of tumor cells, the expression of CDw75 antigen still remains unknown. To address this issue, we investigated immunohistochemically CDw75 antigen expression in 195 colorectal adenocarcinomas and their nodal metastases. The correlation between CDw75 antigen expression with selected clinicopathologic variables was analyzed by using Chi-square and Fisher’s exact tests. Positive staining was observed in 101 cases. Non-neoplastic mucosa was negative consistently. The frequency of positivity was decreased according to the degree of differentiation (p<0.001). Antigen expression was found to be associated with deeper penetration (p<0.006), positive lymph nodes (p<0.001), distant metastases (p<0.006) and advanced stage (p<0.001). Same relationships were detected in well and moderately differentiated tumors when CDw75 immunoreactivity was evaluated in each histologic grade separately. Our findings indicate that CDw75 antigen expression may be a good indicator of the biological aggressiveness of colorectal adenocarcinomas especially in tumors with well and moderately differentiated morphology. C1 [Elpek, Ozlem Gulsum] Akdeniz University, Faculty of Medicine, Department of Pathology, Akdeniz Universitesi, Typ Fakultesi, Patoloji ABD, Yeni Tip, Dekanlik,, 07070 Antalya, Turkey. [Gelen, Tekinalp] Akdeniz University, Faculty of Medicine, Department of Pathology, Akdeniz Universitesi, Typ Fakultesi, Patoloji ABD, Yeni Tip, Dekanlik,, 07070 Antalya, Turkey. [Karpuzoglu, Gulten] Akdeniz University, Faculty of Medicine, Department of Pathology, Akdeniz Universitesi, Typ Fakultesi, Patoloji ABD, Yeni Tip, Dekanlik,, 07070 Antalya, Turkey. [Karpuzoglu, Tuncer] Akdeniz University, Medical School, Department of General SurgeryAntalya, Turkey. [Aksoy, Hikmet Nazif] Akdeniz University, Faculty of Medicine, Department of Pathology, Akdeniz Universitesi, Typ Fakultesi, Patoloji ABD, Yeni Tip, Dekanlik,, 07070 Antalya, Turkey. [Keles, Nuran] Akdeniz University, Faculty of Medicine, Department of Pathology, Akdeniz Universitesi, Typ Fakultesi, Patoloji ABD, Yeni Tip, Dekanlik,, 07070 Antalya, Turkey. RP Elpek, G (reprint author), Akdeniz University, Faculty of Medicine, Department of Pathology, 07070 Antalya, Turkey. EM elpek@med.akdeniz.edu.tr CR Bast BJEG, Zhou LJ, Freeman GJ: The HB-6, CDw75, and CD76 differentiation antigens are unique cell-surface carbohydrate determinants generated by the beta-galactoside alpha 2,6- sialyltransferase. J Cell Biol 116: 423-435, 1992 Bears OS, Henson DE, Hutter RVP: Manual for staging cancer. JB Lippincott Publ Co, Philadelphia, 1992 Cooper HS: Peanut lectin-binding sites in large bowel carcinoma. Lab Invest 47: 383-390, 1982 Dall’Olio F, Malagolini N, Di Stefano G: Increased CMPNeuAc: Gal1, 4GlcNac-R 2,6 sialyltranferase activity in human colorectal cancer tissues. Int J Cancer 44: 434-39, 1989 Dall’Olio F, Malagolini N and Di Stefano G: Post natal development of rat colon epithelial cells is associated with changes in the expression of a1,4-N-Acetylgalactosaminyltransferase involved in the synthesis of Sda antigen and of a2,6 sialyltransferase towards N-acetyllactosamine. Biochem J 270:519- 524, 1990 Dall’Olio F, Malagolini N, Di Stefano: a2,6 sialylation of Nacetyllactosaminic sequences in human colorectal cancer cell lines. Relationships with non-adherent growth. Int J Cancer 47: 291-297, 1991 Dall’Olio F, Trere D: Expression of a2,6 sialylated sugar chains in normal and neoplastic tissues. Detection by digoxigenin- conjugated Sambucus nigra agglutinin. Eur J Histochem 35: 257-265, 1993 Dall’olio F, Malagolini N, Guerrini S: Differentiation-dependent expression of human b-galactoside a2,6-sialyltransferase mRNA in colon carcinoma CaCo-2 cells. Glycoconjugate J 13: 115-121, 1996 David L, Nesland JM, Funderud S: CDw75 antigen expression in human gastric carcinoma and adjacent mucosa. Cancer 72: 1522-1527, 1993 Dennis JW, Laferte S: Tumor cell surface carbohydrates and the metastatic phenotype. Cancer Met Rev 5: 185-204, 1987 Dennis JW: Changes in glycosylation associated with malignant transformation and tumour progression. In: Cell surface carbohydrates and cell development., Ed: Fukuda M), Boca- Raton CRC press Co, 1992, pp: 161-194 Elpek GO, Gelen T, Karpuzoðlu G: Clinicopathologic evaluation of CDw75 antigen expression in patients with gastric carcinoma. J Pathol 193: 169-174, 2001 Erikstein BK, Funderud S, Beiske K: Cell cycle-dependent regulation of CDw75, beta-galactoside alpha-2,6-sialyltransferase, on human lymphocytes. Eur J Immunol 22: 1149-1155,1992 Evans IM, Hilf R, Murphy M: Correlation of serum, tumor, and liver serum glycoprotein: N-acetylneuraminic acid transferase activity with growth of the R3230AC mammary tumor in rats and relationship of the serum activity to tumor burden. Cancer Res 40: 3103-3111,1980 Feizi T: Demonstration by monoclonal antibodies that carbohydrate structures of glycoproteins and glycolipids are oncodevelopmental antigens. Nature 314: 53-57,1985 Fogel M, Altevogt P, Schirrmacher V: Metastatic potential severely altered by changes in tumour cell adhesiveness and cell-surface sialylation. J Exp Med 157: 371-376, 1983 Gangopadhyay A, Perera SP, Thomas P: Differential expression of alpha2,6 sialyltransferase in colon tumors recognized by a monoclonal antibody. Hybridoma 17: 117-123,1998 Ganzinger U, Deutsch E: Serum sialyltransferase levels as a parameter in the diagnosis and follow-up of gastrointestinal tumors. Cancer Res 40: 1300-1304,1980 Hakamori S: Tumor-associated carbohydtrate antigens. Ann Rev Immunol 2: 103-126, 1984 Hakomori S: Aberrant glycosylation in tumors and tumourassociated antigens. Adv Cancer Res 52: 257-331, 1989 Harvey BE, Toth CA, Wagner HE:Sialyltransferase activity and hepatic tumour growth in nude mouse model of colorectal cancer. Cancer Res 52: 1775-1779, 1992 Irimura T, Nakamori S, Matsushita Y: Colorectal cancer metastasis determined by carbohydrate-mediated cell adhesion: role of sialyl-LeX antigens. Semin Cancer Biol 4: 319- 324, 1993 Itzkowitz SH, Yuan M, Ferrell LD: Cancer associated alteration of blood group antigen expression in human colorectal polyps. Cancer Res 46: 5976-5984, 1986 Itzkowitz SH, Yuan M, Montgomery CK: Expression of Tn, sialosyl-Tn and T antigens in human colon cancers. Cancer Res 49: 197-204, 1989 Itzkowitz S, Bloom EJ, Kokal WA: Sialosyl-Tn. A novel mucin antigen associated with prognosis in colorectal cancer patients. Cancer 66: 1960-1966,1990 Jass JR, Sobin LH: International histological classification of tumors: Histological typing of intestinal tumors. Springer-Verlag Publ Co, New York, 1989 Kellokumpu I, Karhi K, Andersson LC: Lectin binding sites in normal, hyperplastic, adenomatous and carcinomatous human colorectal mucosa. APMIS 94: 271-280, 1986 Kemmer W, Kruck D, Schlag P: Different sialyltransferase activities in human colorectal carcinoma cells from surgical specimens detected by specific glycoprotein and glycoproteins acceptors. Clin Exp Metastasis 12: 245-254, 1994 Kijima-Suda I, Miyamoto Y, Toyoshima S: Inhibition of experimental pulmonary metastasis of mouse colon carcinoma 26 subline by a sialic acid nucleoside conjugate having sialyltransferase inhibiting activity. Cancer Res 46: 858-862, 1986 Kjeldsen T, Clausen H, Hirohashi S: Preparation and characterization of monoclonal antibodies directed to the tumour-associated O-linked sialosyl-2-6?-N-acetylgalactosaminyl, Sialosyl- Tn, epitope. Cancer Res 48: 2214-2220, 1988 Li M, Andersen V, Lance P: Expression and regulation of glycosyltransferases for N-glycosyl oligosaccharides in fresh human surgical and murine tissues and cultured cell lines. Clin Sci 89: 397-404, 1995 Mechtersheimer G, Kruger KH, Born IA: Antigenic profile of mammary fibroadenoma and cystosarcoma phyllodes. A study using antibodies to estrogen and progesterone receptors and to a panel of cell surface molecules. Pathol Res Pract 186: 427- 438, 1990 Ming L, Vemulapalli R, Ullah A: Downregulation of a human colonic sialyltransferase by a secondary bile acid and a phorbol ester. Am J Physiol 274: G599-G606, 1998 Miyagi T, Koseki M, Tsuiki S: Comparative studies of the levels of sialyltransferases responsible for the formation of sugar chains in glycoproteins and gangliosides in rat liver and hepatoma. Jpn J Cancer Res 79: 742-749, 1988 Munro S, Bast BJEG, Colley KJ: The B lymphocyte surface antigen CD75 is not an alpha-2,6-sialyltransferase but is a carbohydrate antigen, the production of which requires the enzyme. Cell 68: 1003, 1992 Murayama T, Zuber C, Seelentag WKF: Colon carcinoma glycoproteins carrying a2,6-linked sialic acid reactive with Sambucus Nigra agglutinin are not constituvely expressed in normal human colon mucosa and are distinct from sialyl-Tn antigen. Int J Cancer 70: 575-581, 1997 Nakamori S, Kameyama M, Imaoka S: Increased expression of sialyl Lewis x antigen correlates with poor survival in patients with colorectal carcinoma: clinicopathological and immunohistochemical study. Cancer Res 53: 3632-3637, 1993 Paulson JC, Colley KJ: Glycosyltransferases. Structure, localization and control of cell type-specific glycosylation. J Biol Chem 264: 17615-18, 1989 Paulson JC, Weinstein J, Schauer A: Tissue specific expression of sialyltransferases. J Biol Chem 264: 10931-10934, 1992 Pierce M, Arango J: Rous sarcoma virus-transformed baby hamster kidney cells express higher levels of asparaginelinked, tri-and tetraantennary glycopeptides containing Glc- Nac-b1,6, Man-a1-6)Man and poly-N-acetyllactosamine sequences than baby hamster kidney cells. J Biol Chem 261:10772-10777, 1986 Reed W, Erikstein BK, Funderud S: CDw75 antigen expression in breast lesions. Pathol Res Pract 189: 394-398, 1993 420.Sata T, Roth J, Zuber C: Expression of 2,6-linked sialic acid residues in neoplastic but not in normal human colonic mucosa. A lectin-gold cytochemical study with Sambucus nigra and Maackia amurensis lectins. Am J Pathol 139: 1435-1448, 1991 43. Sgroi D, Varki A, Braesch-Andersen S: CD22 a B-cell specific immunoglobulin superfamily member, is a sialic acid binding lectin. J Biol Chem 268: 7011-7018, 1993 44. Smets LA, Van Beek WP: Carbohydrates of the tumor cell surface. Biochim Biophys Acta 738: 237-249, 1984 45. Stamenkovic I, Sgroi D, Aruffo A: The B-lymphocyte adhesion molecule CD22 interacts with leukocyte common antigen CD45RO on T cells and a2-6 sialyltransferase, CD75, on B cells. Cell 66: 1133-1144, 1991 46. Thampoe IJ, Furukawa K, Vellve I: Sialyl-transferse levels and ganglioside expression in melanoma and other cultured cancer cells. Cancer Res 49: 6258-6264, 1989 47. Weinstein J, Lee EU, McEntee K: Primary structure of b-galactoside ?-2,6 sialyltransferase. J Biol Chem 262: 17735- 17743, 1987 48. Yamada N, Chung Y-S, Takatsuka S: Increased sialyl Lewis A expression and fucosyltransferase activity with acquisition of a high metastatic capacity in a colon cancer cell line. Br J Cancer 76: 582-587, 1997 49. Yamashita K, Ohkura T and Tachibana Y: A comparative study of the oligosaccharides released from baby hamster kidney cells and their polyoma transformants by hydrazinolysis. J Biol Chem 259: 10834-10840, 1984 50. Yogeeswaran G and Salk PL: Metastatic potential is positively correlated with cell surface sialylation of altered murine tumor cell lines. Science 212: 1514-1516, 1981 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2002 VL 8 IS 3 BP 175 EP 182 PG 8 ER PT J AU Banfalvi, T Boldizsar, M Gergye, M Gilde, K Kremmer, T Otto, Sz AF Banfalvi, Teodora Boldizsar, Mariann Gergye, Maria Gilde, Katalin Kremmer, Tibor Otto, Szabolcs TI Comparison of prognostic significance of serum 5-S-Cysteinyldopa, LDH and S-100B protein in Stage III-IV malignant melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article AB 5-S-cysteinyldopa is a precursor of pheomelanin. S-100B protein is a low molecular weight, acidic, calcium binding, cytoplasmatic protein. LDH was defined as the most important serum parameter in disseminated melanoma. The aim of the present study was to compare the prognostic values of serum 5-S-Cysteinyldopa, S-100B and LDH concentrations in Stage III-IV melanoma patients. Serum samples were taken from 179 Stage III-IV melanoma patients at diagnosis. Serum 5-S-CD concentrations were determined by HPLC, S-100B protein by immunoluminometric assay while LDH by UV kinetic method. The mean/median concentrations of LDH, S-100B protein and 5-S-CD in Stage III patients ranged around the normal level. In Stage IV, the markers ranked as S100B = 5-S-CD > LDH for sensitivity, S-100B > LDH > 5-S-CD for specificity and LDH = S100B = 5-S-CD for positive predictive value, respectively. Furthermore, mean marker concentrations of patients with progressive disease differed significantly from nonprogresssive cases (when staging categories have been disregarded). Survival analysis indicated, that the initially elevated LDH and S-100B level in Stage IV disease predicts comparably short survival. Results of our study suggest that these serum marker values correlate well with Stages and disease progression. In Stage IV melanoma, the markers had appropriate sensitivity, high specificity as well as important positive predictive value. Among the studied serum markers S-100B protein and LDH proved to be similarly reliable in respect to the clinical outcome. C1 [Banfalvi, Teodora] National Institute of Oncology, Department of Dermatology, Rath Gyorgy u.7-9.Budapest, Hungary. [Boldizsar, Mariann] National Institute of Oncology, Department of BiochemistryBudapest, Hungary. [Gergye, Maria] National Institute of Oncology, Central Clinical LaboratoryBudapest, Hungary. [Gilde, Katalin] National Institute of Oncology, Department of Dermatology, Rath Gyorgy u.7-9.Budapest, Hungary. [Kremmer, Tibor] National Institute of Oncology, Department of BiochemistryBudapest, Hungary. [Otto, Szabolcs] National Institute of Oncology, Central Clinical LaboratoryBudapest, Hungary. RP Banfalvi, T (reprint author), National Institute of Oncology, Department of Dermatology, Budapest, Hungary. EM banfalvit@freemail.hu CR Balch CM, Buzaid AC, Atkins MB, et al:A new American Joint Committee on cancer staging system cutaneous melanoma. Cancer 88: 1484-1491, 2000 Banfalvi T, Gilde K, Boldizsar M, et al: Serum levels of S-100 protein and 5-S-cysteinyldopa as markers of melanoma progression. Pathol Oncol Res 5: 218-223, 1999 Banfalvi T, Gilde K, Boldizsar M, et al: Serum concentration of 5-S-cysteinyldopa in patients with melanoma. Eur J Clin Invest 30: 900-904, 2000 Benathan M, Labidi F: Modulation of 5-S-cysteinyldopa formation by tyrosinase activity and intracellular thiols in human melanoma cells. Melanoma Res 6: 183-189, 1996 Berking C, Schlupen E, Schrader A, et al: Tumor markers in peripheral blood of patients with malignant melanoma: Multimarker RT-PCR versus a luminometric assay for S-100. Arch Dermatol Res 291: 479-484, 1999 Buer J, Probst M, Franzke A et al: Elevated serum levels of S- 100 and survival in metastatic malignant melanoma. Br J Cancer 75: 1373-1376, 1997 Buzaid AC, Ross MI, Balch CM, et al: Critical analysis of the current American Joint Committee on Cancer staging system for cutaneous melanoma and proposal of new staging system. J Clin Oncol 15: 1039-1051, 1997 Clark WH Jr, From L, Bernardino EA, Mihm MC: The histogenesis and biologic behaviour of primary human malignant melanomas of the skin. Cancer Res 29: 705-726, 1991 Deichmann M, Benner A, Bock Mjackel A, et al: S-100 beta, melanoma inhibiting activity and lactate dehydrogenase discriminate progressive from nonprogressive AJCC Stage IV melanoma. J Clin Oncol 17: 1891-1896, 1999 Deichmann M, Benner A, Kuner N et al: Are responses to therapy of metastatized melanoma reflected by decreasing serum values of S-100 beta or MIA? Melanoma Res. 11: 291-296, 2001 Djukanovic D, Hofmann U, Sucker A, et al: Comparison of S- 100 protein and MIA protein as serum markers in malignant melanoma. Anticancer Res 20: 2203-2207, 2000 Franzke A, Probst-kepper M, Buer J, et al: Elevated pretreatment serum levels of soluble vascular cell adhesion molecule 1 and lactate dehydrogenase as predictors of survival in cutaneous metastatic malignant melanoma. Br J Cancer 78: 40-45, 1998 Guo HB, Stoffel-Wagner B, Bierwirth T, et al: Clinical significance of serum S-100 in metastatic malignant melanoma. Eur J Cancer 31A: 1898-1902, 1995 140.Hansson C, Edholm L, Agrup G, et al: The quantitative determination of 5-S-cysteinyl-dopa and dopa in normal serum and in serum from patients with malignant melanoma by means of high performance liquid chromatography. Clin Chim Acta 88: 419-421, 1978 15. Hasegawa M, Takata M, Hatta N, Wakamatsu K, et al: Simultaneous measurement of serum 5-S-cysteinyldopa, circulating intercellular adhesion molecule-1 and soluble interleukin-2 receptor levels in Japanese patients with malignant melanoma. Melanoma Res 7: 243-251, 1997 16. Hauschild A: The use of serological tumor markers for malignant melanoma. Onkologie 20: 462-465, 1997 17. Hauschild A, Michaelsen J, Brenner W, et al: Prognostic significance of serum S-100B detection compared to routin blood parameters in advanced metastatic melanoma patients. Melanoma Res 9: 155-161, 1999 18. Horikoshi T, Ito Sh, Wakamatsu K, et al: Evaluation of melaninrelated metabolites as markers of melanoma progression. Cancer 73: 629-636, 1994. 19. Karnell R, von Schoultz E, Hansson LO, et al.: S 100 B protein, 5-S-cysteinyldopa and 6-hydroxy-5-metoxyindole-2-carboxylic acid as biochemical markers for survival prognosis in patients with malignant melanoma. Melanoma Res 7: 393-399, 1997 20. Karnell R, Kagedal B., Lindholm C, et al: The value of cysteinyldopa in the follow up of disseminated malignant melanoma. Melanoma Res 10: 363-369, 2000 21. Krahn G, Kaskel P, Sander S, et al: S-100 beta is a more reliable tumor marker in peripherial blood of patients with newly occurred melanoma metastases comparing to MIA, albumin and lactat dehydrogenase. Anticancer Res 21: 311-1316, 2001 22. Martenson ED, Hansson LO, Nilsson B, et al.: Serum S-100b protein as prognostic marker in malignant cutaneuos melanoma. J Clin Oncol 19: 824-831, 2001 23. Meyerhoffer S, Lindberg Z, Hager A, et al: Urinary excretion of 5-S-cysteinyldopa and 6-hydroxy-5-methoxyindole-2-carboxylic acid in children. Acta Derm Venereol 78: 31-35, 1998 24. Sasaki Y, Shimizu H, Naka W, et al: Evaluation of the clinical usefulness of measuring urinary excretion of 5-S-cysteinyldopa in melanoma: ten years experience of 50 patients. Acta Derm Venerol 77: 379-381, 1998 25. Seregeni E, Massaron S, Martinetti A, et al: S-100 protein serum levels in cutaneous malignant melanoma. Oncol Rep 5: 601-604, 1998 26. Schmitz C, Brenner W, Henze E, et al: Comparative study on the clinical use of protein S-100 B and MIA in melanoma patients. Anticancer Res 20: 5059-5063, 2000 27. Schoultz ES, Diepgen TL, von den Dresch P: Clinical prognostic relevance of serum S-100b protein in malignant melanoma. Br J Dermatol 38: 426-430, 1998 28. Wakamatsu K, Ito S: Seasonal variations in serum concentration of 5-S-cysteinyldopa and 6-hydroxy-5-methoxyindole-2- carboxyl acid in healthy Japanese. Pigment Cell Res 8: 132- 134, 1995 29. Wibe E, Hannisdal E, Paus E, Aamdal S: Neuron-specific enolase as prognostic factor in metastatic melanoma. Eur J Cancer 28: 1692-1695, 1992 30. Wimmer I, Meyer CJ, Seifert B, et al.: Prognostic value of serum 5-S-cysteinyldopa for monitoring human metastatic melanoma during immunochemoterapy. Cancer Res 57: 5073- 5076, 1997 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2002 VL 8 IS 3 BP 183 EP 187 PG 5 ER PT J AU Gumurdulu, D Zeren, H Cagle, TP Kayaselcuk, F Alparslan, N Kocabas, A Tuncer, I AF Gumurdulu, Derya Zeren, E. Handan Cagle, T Philip Kayaselcuk, Fazilet Alparslan, Nazan Kocabas, Ali Tuncer, Ilhan TI Specificity of MOC-31 and HBME-1 Immunohistochemistry in the Differential Diagnosis of Adenocarcinoma and Malignant Mesothelioma: a Study on Environmental Malignant Mesothelioma Cases from Turkish Villages SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article AB Histological diagnosis of malignant mesothelioma (MM) and differentiation from adenocarcinoma is often difficult. A number of clinical, radiologic, histologic and histochemical criteria have been used as diagnostic aids, but most cases cannot be readily classified on the basis of these characteristics. In recent years, a panel of immunohistochemical anti-bodies have been increasingly applied for the differential diagnosis of these two tumors. MOC-31 has been recently used as specific for adenocarcinomas while reacting with a minimal number of benign and malignant mesothelial proliferations, and HBME-1 has also been presented as a mesothelial cell marker. In this study, we aimed to show the importance of these two antibodies among the environmental MM cases from Southeastern Turkey. Fifty five cases of MM and twenty adenocarcinomas were included in this study. Histochemical (PAS, PAS-D, mucicarmine) and immunohistochemical (Keratin, EMA,CEA, MOC-31, HBME-1) stains have been performed on each case. Keratin was positive in all cases. EMA stained 50 of 55 MM and all the adenocarcinoma cases. According to our results, dPAS, mucicarmen, CEA and MOC-31 positivity was statistically significant in the diagnosis of adenocarcinoma whereas HBME-1 was demonstrable in most MM cases (52/55) and 11 adenocarcinoma cases. This study confirmed that in the diagnostic distinction between MM and adenocarcinoma, immuno-histochemistry is an important diagnostic tool, however, a panel of antibodies must be used rather than any single antibody. HBME-1 should be included in this panel; MOC-31 can be used where CEA is not available or to doublecheck the reactivity of this antibody. C1 [Gumurdulu, Derya] Cukurova University, School of Medicine, Pathology Department, 01330 Adana, Turkey. [Zeren, E. Handan] Cukurova University, School of Medicine, Pathology Department, 01330 Adana, Turkey. [Cagle, T Philip] Univeristy of Houston, Department of PathologyHouston, USA. [Kayaselcuk, Fazilet] Cukurova University, School of Medicine, Pathology Department, 01330 Adana, Turkey. [Alparslan, Nazan] Cukurova University, Faculty of Medicine, Department of BiostatisticsAdana, Turkey. [Kocabas, Ali] Cukurova University, Faculty of Medicine, Department of Pulmonary MedicineAdana, Turkey. [Tuncer, Ilhan] Cukurova University, School of Medicine, Pathology Department, 01330 Adana, Turkey. CR Sheibani K, Battifora H, Burke JS: Antigenic phenotype of malignant mesothelioma and pulmonary adenocarcinomas. Am J Pathol 123: 212-219, 1986 Sosolik RC, Mcgaughy VR, De Young BR: Anti-MOC-31: A potential addition to the pulmonary adenocarcinoma versus mesothelioma immunohistochemistry panel. Mod Pathol 10: 716-719, 1997 Donna A, Betta PG, Chiodera P, et al: Newly marketed tissue markers for malignant mesothelioma: Immunoreactivity of rabbit AMAD-2 antiserum, compared with monoclonal antibody HBME-1 and a review of the literature on so-called antimesothelioma antibodies. Hum Pathol 28: 929-937, 1997 Zeren EH, Gumurdulu D, Roggli VL, et al: Environmental Malignant Mesothelioma in Southern Anatolia: A study of fifty cases. Environ Health Perspect 108: 1047-1050, 2000 Warnock ML, Stoloff A, Thor A: Differentiation of adenocarcinoma of the lung from mesothelioma: periodic acid schiff, monoklonal antibodies B 72.3 and leu M1. Am J Pathol 133: 30-38, 1988 Brown RW, Clark GM, Tandon AK, et al: Multiple-marker immunohistochemical phenotypes distinguishing malignant pleural mesothelioma from pulmonary adenocarcinoma. Hum Pathol 24: 347-354, 1993 Skov BG, Lauritzen AF, Hirsch F, et al: The histopathological diagnosis of malignant mesothelioma v.pulmonary adenocarcinoma: reproducibility of the histopathological diagnosis. Histopathology 24: 553-557, 1994 Skov B, Lauritzen AF, Hirsch F, et al: Differantiation of adenocarcinoma of the lung and malignant mesothelioma: Predictive value and reproducibility of immunoreactive antibodies. Histopathology 25: 431-437, 1994 Wirth PR, Legier J, Wright GL: Immunohistochemical evaluation of seven monoclonal antibodies for differentiation of pleural mesothelioma from lung adenocarcinoma. Cancer 67: 655-662, 1991 Zeng L, Feith J, Monnet I, et al: Immunocytochemical characterization of cell lines from human malignant mesothelioma: characterization of human mesothelioma cell lines by immunocytochemistry with a panel of monoclonal antibodies. Hum Pathol 25:3: 227-234, 1994 Cagle PT, Truong LD, Roggli VL, et al: Immunohistochemical differentiation of sarcomatoid mesotheliomas from other spindle cell neoplasms. Am J Clin Pathol 92: 566-571, 1989 Sheibani K, Esteban JM, Bailey A, et al: Immunopathologic and molecular studies as an aid to the diagnosis of malignant mesothelioma. Hum Pathol 23: 107-116, 1992 Bateman AC, Al-Talib RK, Newman T: Immunohistochemical phenotype of malignant mesothelioma: predictive value of CA 125 and HBME-1 expression. Histopathology 30: 49-56, 1997 Dejmek A, Hjerpe A: Immunohistochemical reactivity in mesothelioma and adenocarcinoma: A stepwise logistic regression analysis. APMIS 102: 255-264, 1994 Battifora H, Kopinski MI: Distinction of mesothelioma from adenocarcinoma. Cancer 55: 1679-1685, 1985 Kortsik CS, Werner P, Freudenberg N, et al: Immunocytochemical characterization of malignant mesothelioma and carcinoma metastatic to the pleura: IOB3- a new tumor marker. Lung 173: 79-87, 1995 Moch H, Oberholzer M, Dalquen P, et al: Diagnostic tools for differentiating between pleural mesothelioma and lung adenocarcinoma in paraffin embedded tissue. Part I: immunohistochemical findings. Virchows Arch A Pathol Anat 423: 19-27, 1993 Mezger J, Lamerz R, Permanetter W: Diagnostic significance of carcinoembryonic antigen in the differantial diagnosis of malignant mesothelioma. J Thorac Cardiovasc Surg 100: 860- 866, 1990 Morgan RL, De Young BR, McGaughy VR, et al: MOC-31 aids in the differentiation between adenocarcinoma and reactive mesothelial cells. Cancer 87: 390-394, 1999 Edwards C, Oates J: OV 632 and MOC-31 in the diagnosis of mesothelioma and adenocarcinoma: an assessment of their use in formalin fixed and paraffin wax embedded material. J Clin Pathol 48: 626-630, 1995 Ordonez NG: Value of the MOC-31 monoclonal antibody in differentiating epithelial pleural mesothelioma from lung adenocarcinoma. Hum Pathol 29: 166-169, 1998 Attanoos RL, Goddard H, Gibbs AR: Mesothelioma-binding antibodies: thrombomodulin, OV 632 and HBME-1 and their use in the diagnosis of malignant mesothelioma. Histopathology 29: 209-215. 1996 Kennedy AD, King G, Kerr KM: HBME-1 and antithrombomodulin in the differential diagnosis of malignant mesothelioma of pleura. J Clin Pathol 50: 859-862, 1997 Fetsch PA, Abati A, Hijazi YM: Utility of the antibodies CA 19- 9, HBME-1 and thrombomodulin in the diagnosis of malignant mesothelioma and adenocarcinoma in cytology. Cancer 84: 101-108, 1998 Riera JR, Astengo-Osuna C, Longmate JA,et al: The immunohistochemical diagnostic panel for epithelial mesothelioma: a reevaluation after heat induced epitope retrieval. Am J Surg Pathol 21: 1409-1419, 1997 Cury PM, Butcher DN, Fisher C, et al: Value of the mesothelium- associated antibodies thrombomodulin, cytokeratin 5/6, calretinin and CD44H in distinguishing epithelioid pleural mesothelioma from adenocarcinoma metastatic to the pleura. Mod Pathol 13: 107-112, 2000 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2002 VL 8 IS 3 BP 188 EP 193 PG 6 ER PT J AU Noorali, S Nausheen, Y Nasir, IM Moatter, T Pervez, Sh AF Noorali, Samina Nausheen, Yaqoob Nasir, Israr Muhammad Moatter, Tariq Pervez, Shahid TI Prevalence of Mycosis Fungoides and Its Association with EBV and HTLV-1 in Pakistanian Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article AB Mycosis fungoides (MF) is an indolent T cell lymphoma that is distinguished from other lymphomas by its initial appearance on the skin. The histologic diagnosis of MF may be difficult because there is significant overlap in the histologic features of neoplastic T-cell infiltrates and inflammatory dermatoses. This T-cell neoplasm commonly occurs in a mixed, reactive background and can show only a subtle degree of cytologic atypia, rendering histologic diagnosis difficult. In this study MF constituted 0.86% of all non-Hodgkin’s lymphoma (NHL) both T and B, as compared to the Western studies which have reported 0.5% prevalence for MF of all NHL. Polymerase chain reaction (PCR) technique was used to assess T-cell clonality in paraffin-embedded skin biopsies clinically and pathologically suspicious for early MF. Out of the 14 cases diagnosed as MF, amplifiable DNA was isolated from 6 cases, which were further studied for T-cell receptor (TcR) – b b, g g, and d dchain gene rearrangements. Clonal product was seen in 4 out of 6 cases for b b, g g, and d dTcR chain genes. Association for Epstein Barr virus (EBV) was observed in 3 out of 6 cases (50%) of MF. Although these 3 cases were positive for EBV by PCR, but were negative by in-situ hybridization (ISH). No heterogeneity was noted in these 3 cases of MF for BamHI E, K, N, and Z regions of EBV. All six cases were negative for HTLV-1 (tax region) by PCR. It was concluded that the prevalence of MF in Pakistani population is comparable to the Western data, and that EBV association to MF cases was higher than in Western studies. C1 [Noorali, Samina] The Aga Khan University, Department of Pathology and Microbiology, 74800 Karachi, Pakistan. [Nausheen, Yaqoob] The Aga Khan University, Department of Pathology and Microbiology, 74800 Karachi, Pakistan. [Nasir, Israr Muhammad] The Aga Khan University, Department of Pathology and Microbiology, 74800 Karachi, Pakistan. [Moatter, Tariq] The Aga Khan University, Department of Pathology and Microbiology, 74800 Karachi, Pakistan. [Pervez, Shahid] The Aga Khan University, Department of Pathology and Microbiology, 74800 Karachi, Pakistan. RP Pervez, Sh (reprint author), The Aga Khan University, Department of Pathology and Microbiology, 74800 Karachi, Pakistan. EM shahid.pervez@aku.edu CR The Non-Hodgkin’s Lymphoma Pathologic Classification: National Cancer Institute sponsored study of classification of non-Hodgkin’s lymphomas. Summary and description of a working formulation for clinical usage. Cancer 49: 2112-2135, 1982 Kim YH, Hoppe RT: Mycosis Fungoides and Sezary Syndrome. Semin Oncol 26: 276-289,1999 Maurice B. Mycosis Fungoides: Diagnosis and Pathogenesis. Am J Clin Pathol 99: 452-458, 1993 Nasu K, Said J, Vonderheid E, Olerud J, Sako D, Kadin M: Immunopathology of Cutaneous T-cell lymphomas. Am J Pathol 119: 436-447, 1985 Sandberg AA. The chromosomes in human cancer, ed 2 New York: Elsevier, 1990, pp 405-409 Kanavaros P, Ioannidou D, Tzardi M, et al: Mycosis Fungoides: expression of c-myc, p62, p53, bcl-2 and PCNA proteins and absence of association with Epstein-Barr virus. Pathol Res Pract 190: 767-774, 1994 Peris K, Niedermeyer H, Cerroni L, et al: Detection of Epstein Barr virus genome in primary cutaneous T and B cell lymphomas and pseudolymphomas. Arch Dermatol Res 286: 364-368, 1994 Dreno B, Celerier P, Fleischmann M, et al: Presence of Epstein-Barr virus in cutaneous lesions of Mycosis Fungoides and Sezary syndrome. Acta Derm Venereol 74: 355-357, 1994 Ranki A, Niemi KM, Nieminen P, Krohn K: Antibodies against retroviral core proteins in relation to disease outcome in-patients with mycosis fungoides. Arch Dermatol Res 282:532-538, 1990 Wood GS, Salvekar A, Schaffer J, et al: Evidence against a role for HTLV-1 in the pathogenesis of American cutaneous T-cell lymphoma. J invest Dermatol 107: 301-307, 1996 Persing DH, Smith TF, Tenover FC, White TJ: PCR detection and typing of Epstein-Barr virus. Diagnostic Molecular Microbiology: Principles and Applications 344-349, 1993 Kwok S, Ehrlich G, Poiesz B, et al: Enzymatic amplification of HTLV-1 viral sequences from peripheral blood mononuclear cells and infected tissues. Blood 72: 1117-1123, 1988 Chang KL, Chen YY, Shibata D, Weiss LM: Description of an insitu hybridization methodology for detection of Epstein-Barr virus RNA in paraffin-embedded tissues, with a survey of normal and neoplastic tissue. Diag Mol Pathol 1: 246-255, 1992 Dimitris T, Alison W. Boulter, Jair et al: Diversity of naturally occurring Epstein-Barr virus revealed by nucleotide sequence polymorphism in hypervariable domains in the Bam HI K and N subgenomic regions. J Gen Virol 79: 2809-17, 1998 Wrightham MN, Stewart JP, Janjua NJ, et al: Antigenic and sequence variation in the C-terminal unique domain of the Epstein-Barr virus nuclear antigen EBNA-1. Virology 208: 521-530,1995 Hu LF, Zabarovsky ER, Chen F, et al: Isolation and sequencing of the Epstein-Barr virus BNLF-1 gene, LMP1, from a Chinese nasopharyngeal carcinoma. J Gen Virol 72: 2399-2409, 1991 Packham G, Brimmell M, Cook D, et al: Strain variation in Epstein-Barr virus immediate early genes. Virology 192: 541- 550,1993 Sample J, Young L, Martin B, et al: Epstein-Barr virus types 1 and 2 differ in their EBNA-3A, EBNA-3B, and EBNA-3C genes. J Virol 64: 4084-4092, 1990 Slack DN, McCarthy KP, Wiedemann LM, Sloane JP: Evaluation of sensitivity, specificity and reproducuibility of an optimized method for detecting clonal rearrangements of immunoglobulin and T cell receptor genes in formalin fixed, paraffin embedded sections. Diag Mol Path 2:223-232,1993 McCarthy KP, Sloane JP, Karbarowski JHS, et al: The rapid detection of Clonal T-cell proliferations in patients with lymphoid disorders. Am J Pathol 138: 821-828, 1991 McCarthy KP, Sloane JP, Karbarowski JHS, et al: A simplified method of detection of clonal rearrangements of the T cell receptor gamma chain gene. Diag Mol Path 1: 173-179, 1992 Goudie RB, Karim SN, Mills K, et al: A sensitive method of screening for dominant T cell clones by amplification of T cell gamma gene rearrangements with the polymerase chain reaction. J Pathol 162: 191-196, 1990 Campana D, Yokota S, Coustan S, et al: The detection of residual acute lymphoblastic leukaemia cells with immunologic methods and polymerase chain reaction. Leukemia 4: 609-614, 1990 Langerak AW, Szezepanski T, Burg MV, et al: Heteroduplex PCR analysis of rearranged T cell receptor genes for clonality assessment in suspect T-cell proliferations. Leukemia11: 2192- 2199, 1997 Harris NL, Jaffe ES, Stein H, et al. A Revised European-American Classification of Lymphoid Neoplasms: A proposal from the International Lymphoma Study Group. Blood 84: 1361- 1392, 1994 Harris NL, Jaffe ES, Diebold J, et al: World Health Organization Classification of Neoplastic Diseases of Hematopoietic and Lymphoid Tissues: Report of the Clinical Advisory Committee Meeting- Airlie House, Virginia, November 1997. J Clin Oncol 17: 3835-3849, 1999 Pandolfino TL, Siegel RS, Kuzel TM, et al: Primary Cutaneous B cell Lymphoma: Review and Current Concepts. J Clin Oncol 18 : 2152-2168, 2000 Diamandidou E, Cohen PR, Kurzrock R: Mycosis Fungoides and Sezary Syndrome. Blood 88: 2385-2409, 1999 Ishiji T, Takagi Y, Niimura M: Cutaneous lymphomas in Tokyo: analysis of 62 cases in a dermatology clinic. Int J Dermatol 40: 37-40, 2001 Kikuchi A, Shimizu H, Nishikawa T: Mycosis fungoides with marked hyperpigmentation. Dermatology 192: 360-363, 199. Katz B, Raab-Traub A, Miller G: Latent and replicating forms of Epstein-Barr virus DNA in lymphomas and lymphoroliferative diseases. J Infect Dis, 160: 589-598, 1989 Weiss LM, Strickler JG, Warnke RA, et al: Epstein-Barr viral DNA in tissues of Hodgkin’s disease. Am J Pathol 129: 86-91, 1987 Angel CA, Slater DN, Royds JA, et al: Absence of Epstein-Barr viral encoded RNS, ERER, in primary cutaneous T-cell lymphoma. J Pathol 178: 173-175, 1996 Mouly F, Baccard M, Rybojad M, et al: Aggressive cutaneous T-cell lymphoma associated with the presence of Epstein-Barr virus. 2 cases. Ann Dermatol Venereol 123: 574-576, 1996 Anagnostopoulos I, Hummel M, Kaudewitz P, et al: Low incidence of Epstein-Barr virus presence in primary cutaneous T-cell lymphoproliferations. Br J Dermatol 134: 276-281, 1996 Ashton-Key M, Diss TC, Du MQ, et al: The value of the polymerase chain reaction in the diagnosis of cutaneous T-cell infiltrate. Am J Surg Pathol 21: 743-747, 1997 Jorge RT, Micheal B, Lynn S, et al: T-cell lymphoma of the skin. Arch Dermatol 136: 1024-1032, 2000 Thor Straten P, E. Ralfkiaer, J. Hendriks, et al: T-cell receptor variable region genes in Cutaneous T-cell Lymphomas. Br J Dermatol 138: 3-12, 1998 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2002 VL 8 IS 3 BP 194 EP 199 PG 6 ER PT J AU Kacar, F Meteoglu, I Sen, S Levi, E AF Kacar, Furuzan Meteoglu, Ibrahim Sen, Serdar Levi, Edi TI Primary Neuroendocrine Carcinoma of the Mediastinum SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports AB A mediastinal mass was found in a 37 year old male who presented with fever, weight loss and fatigue. The chest CT revealed a 9x6x4 cm well circumscribed mass located paratracheally in the upper mid-mediastinum. The mass was removed by right thoracotomy. Macroscopically the tumor weighed 195 g and measured 9x6x4 cm. Microscopically the tumor consisted of small blue cells in solid and trabeculer patern. Immunohistochemical studies performed for differential diagnosis of small blue cell tumors. The tumor was diagnosed as primary neuroendocrine carcinoma of the mediastinum. This case is presented for its rare recurrence in that particular location. C1 [Kacar, Furuzan] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Meteoglu, Ibrahim] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Sen, Serdar] Adnan Menderes University, Faculty of Medicine, Department of Thoracic SurgeryAydin, Turkey. [Levi, Edi] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. RP Kacar, F (reprint author), Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. EM furuzankacar@hotmail.com CR Modlin IM, Sandor A: An analysis of 8305 cases of carcinoid tumors. Cancer 79: 813-829, 1997 Wick MR, Rosai J: Neuroendocrine neoplasms of the mediastinum. Semin Diagn Pathol 8: 35-51, 1991 Yazima A, Toki T, Morinaga S, et al: A retroperitoneal endocrine carcinoma. Cancer 54: 2040-2042, 1984 Horenstein MG, Erlandson RA, Gonzalez-Cueto DM, Rosai J: Presacral carcinoid tumors: report of three cases and review of the literature. Am J Surg Pathol 22: 251-255, 1998 Enzinger FM, Weiss SW: Peripheral neuroepithelioma. In: Soft tissue tumors. 3rd ed. New York, NY Mosby; 945-951, 1995 Horie Y, Keto M: Neuroendocrine carcinoma of the posterior mediastinum. A possible primary lesion. Arch Pathol Lab Med 123: 933-936, 1999 Hill DA, Pfeifer JD, Marley EF et al: WT1 staining reliably differentiates desmoplastic small round cell tumor from Ewing Sarcoma/PNET. Am J Clin Pathol 114: 345-353, 2000 Wick MR, Scheithauer BW. Thymic carcinoid: a histologic, immunohistochemical and ultrastructural study of 12 cases. Cancer 53: 475-484, 1984 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2002 VL 8 IS 3 BP 200 EP 201 PG 2 ER PT J AU Repassy, LD Ivanyi, A Csata, S Tamas, Gy AF Repassy, Laszlo Denes Ivanyi, Andras Csata, Sandor Tamas, Gyorgy TI Combined Occurrence of Prostate Carcinoma and Malacoplakia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports AB This is a case of a 56-year-old male patient suffering from a combination of prostate carcinoma and malacoplakia. A 56-year-old male patient was admitted in our Departement because ischuria due to the enlargement of the prostate. Perineal needle biopsy was done resulting carcinoma of prostatae. Radical prostatectomy was performed. Histology proved carcinoma and a great part of the enlarged prostate consisted of malacoplakia. Questions related to the morphology, tissue structure as well as diagnosis are briefly surveyed. This is the first report on the rarely occurring combination of prostate carcinoma and malacoplakia treated by radical retropubic prostatectomy. C1 [Repassy, Laszlo Denes] Saint Stephen Hospital, Department of Urology, Nagyvarad ter 1, H-1096 Budapest, Hungary. [Ivanyi, Andras] Municipal Hospital, Del-Pest, Department of PathologyBudapest, Hungary. [Csata, Sandor] Saint Stephen Hospital, Department of Urology, Nagyvarad ter 1, H-1096 Budapest, Hungary. [Tamas, Gyorgy] Saint Stephen Hospital, Department of Urology, Nagyvarad ter 1, H-1096 Budapest, Hungary. RP Repassy, LD (reprint author), Saint Stephen Hospital, Department of Urology, H-1096 Budapest, Hungary. CR Long JP Jr, Althausen AF: Malacoplakia: a 25-year experience with a review of the literature. J Urol 14:1328-1331, 1989 Michaelis L, Gutmann C: Uber einschliesse in Blasentumoren. Ztschr Klin Med 47:20-215, 1902 Von Hansemann D: Uber Malakoplakie der Harnblase. Wirch Arch Path Anat 173:302-308, 1903 Sarma HN, Ramesh K, al Fituri O, et al: Malakoplakia of the prostate gland. Report of two cases and review of the literature. Scand J Urol Nephrol 30:155-157, 1996 Suzuki Z., Kubota Y, Sasagawa I, et al: Malacoplakia of the prostate. Int Urol Nephrol 27:551-555, 1995 Koga S, Arakaki Y, Matsouka M, Ohyama C: Malakoplakia of prostate. Urology 27:160-161, 1986 Thrasher JB, Donatucci CF: Malakoplakia and carcinoma of the prostate. Br J Urol 73:111-112, 1994 Liu S, Christmas TJ, Kirby RS: Malakoplakia and carcinoma of the prostate. Br J Urol 72:120-121, 1993 Sujka SK, Malin BT, Asirwatham JE: Prostatic malakoplakia associated with prostatidc adenocarcinoma and multiple prostatic abscesses. Urology 34:159-161, 1989 Chantelois AE, Parker SH, Sims JE, Horne DW: Malacoplakia of the prostate sonographically mimicking carcinoma. Radiology 177:193-195, 1990 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2002 VL 8 IS 3 BP 202 EP 203 PG 2 ER PT J AU Timar, J Csuka, O Orosz, Zs Jeney, A Kopper, L AF Timar, Jozsef Csuka, Orsolya Orosz, Zsolt Jeney, Andras Kopper, Laszlo TI Molecular Pathology of Tumor Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture AB Molecular Pathology of Tumor Metastasis With the development of non-invasive methods, diagnosis of metastasis from various solid malignancies has become a routine task for diagnostic pathology. However, the differential diagnosis between primary and metastatic cancers and the precise identification of various metastatic cancer types requires the coordinated use of various morphological (light- and electron microscopic-), immunological and molecular techniques. The detection of the lymphatic spread of the primary tumor may now based on the sentinel lymph node technology while the identification of the hematogenous progression may be based on the analysis of the peripheral blood and the bone marrow. More and more frequently these techniques employ highly sensitive immunological and molecular techniques. Accordingly, clinical staging is now confronted with the results of molecular staging, where the only techniques which are able to detect cancer cells are immunocytochemistry or nucleic acid-based methodology. Although several clinical studies have provided evidences for the impact of the immunocytochemistry-based identification of micrometastases on the survival of patients with various type of cancers, none of these methods have become part of standard diagnostic protocols. Although more sensitive molecular techniques are being introduced to identify micrometastasis, their clinical significance is yet unknown. Multicentric clinical trials are now warranted to establish the clinical impact of molecular staging in various cancer types. Without the integration of these methods into the prognostic/predictive pathological protocols it is difficult to envision significant improvement in the results of cancer therapy. C1 [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Csuka, Orsolya] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Orosz, Zsolt] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Jeney, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. CR Bernini A, Spencer M, Frizelle S et al: Evidence for colorectal cancer micrometastases using reverse transcriptase-polymerase chain reaction analysis of MUC2 in lymph nodes. Cancer Detect Prev 24: 72-79, 2000 Hardingham JE, Kotasek D, Farmer B et al: Immunobead- PCR: A technique for the detection of circulating tumor cells using immunomagnetic beads and the polymerase chain reaction. Cancer Res 53: 3455-3458,1993 Hildebrandt M, Mapara MY, Korner IJ et al: Reverse transcriptase- polymerase chain reaction, RT-PCR)-controlled immunomagnetic purging of breast cancer cells using the magnetic cell separation, MACS, system: A sensitive method for monitoring purging efficiency. Exp Hematol 25: 57-65,1997 Tsuchiya A, Sugano K, Kimijima I, Abe R: Immunohistochemical evaluation of lymph node micrometastases from breast cancer. Acta Oncol 35: 425-428,1996 Hashimoto T, Kobayashi Y, Ishikawa Y et al. Prognostic value of genetically diagnosed lymph node micrometastasis in nonsmall cell lung carcinoma cases. Cancer Res 60:6472-8, 2000 Min CJ, Tafra L, Verbanac KM: Identification of superior markers for polymerase chain reaction detection of breast cancer metastases in sentinel lymph nodes. Cancer Res 58: 4581- 4584,1998 Mitas M, Mikhitarian K, Walters C et al: Quantitative realtime RT-PCR detection of breast cancer micrometastasis using a multigene marker panel. Int J Cancer 93: 162-171, 2001 Blaheta HJ, Schittek B, Breuninger H et al: Lymph node micrometastases of cutaneous melanoma: increased sensitivity of molecular diagnosis in comparison to immunohistochemistry. Int J Cancer 79: 318-323,1998 Biegliek SC, Ghossein RA, Bhattacharya S, Coi DG: Detection of tyrosinase mRNA by reverse transcriptase polymerase chain reaction, RT-PCR, in melanoma sentinel nodes. Ann Surg Oncol 6: 232-240,1999 Zeidman I, Buss JM. Experimental studies of the spread of cancer in the lymphatic system. Cancer Res 1954, 14: 403-405 Cserni G, Vajda K, Tarjan M et al. Nodal staging of colorectal carcinomas from quantitative and qualitative aspects. Can lymphatic mapping help staging? Pathol Oncol Res 5:291-6, 1999 Gould EA, Winship T, Philbin PH, Kerr HH: Observations on a ‘sentinel node’ in cancer of the parotid. Cancer 13: 77-78, 1960 Cabanas RM. An approach for the treatment of penile carcinoma. Cancer 39: 456-466, 1977 Morton DL, Wen DR, Wong et al: Technical details of intraoperative lymphatic mapping for early stage melanoma. Arch Surg 127: 392-399, 1992 Giuliano AE, Kirgan DM, Guenther JM, Morton DL. Lymphatic mapping and sentinel lymphadenectomy for breast cancer. Ann Surg 1994, 220: 391-398 Krag DN, Weaver DL, Alex JC, Fairbank JT. Surgical resection and radiolocalization of the sentinel lymph node in breast cancer using a gamma probe. Surg Oncol 1993, 2: 335-339 Peley G, Farkas E, Teglas M et al: Feasibility and accuracy of the combined radioisotope and blue-dye guided sentinel lymph node biopsy in breast cancer. Magyar Sebeszet 53:241- 246,2000 Tanis P J, Nieweg OE, Olmos RAV et al: History of sentinel node and validation of the technique. Breast Cancer Res 3: 109-112,2001 Dowlatshahi K, Fan M, Snider HC, Habib FA. Lymph node micrometastases from breast carcinoma: reviewing the dilemma. Cancer 80:1188-1198,1997 Cote RJ, Peterson HF, Chaiwun B et al: Role of immunohistochemical detection of lymph-node metastases in management of breast cancer. International Breast Cancer Study Group. Lancet 354:896-900, 1999 Rampaul RS, Miremadi A, Pinder SE et al: Pathological validation and significance of micrometastasis in sentinel nodes in primary breast cancer. Breast Cancer Res 3:113-6, 2001 Silverberg SG. Sentinel node Processing. Recommendations for pathologists. Am J Surg Pathol 26:383-385, 2002 Yared MA, Middleton LP, Smith TL et al:Recommendation for sentinel lymph node processing in breast cancer. Am J Surg Pathol 26:377-382, 2002 Cserni G. Metastases in axillary sentinel lymph nodes in breast cancer as detected by intensive histopathological work up.J Clin Pathol 52: 922-924, 1999 Veronesi U, Paganelli G, Viale G et al: Sentinel lymph node biopsy and axillary dissection in brest cancer: results in a large series. J Natl Cancer Inst 91:368-73,1999 Ratanawichitrasin A, Biscotti CV, Levy L Crowe JP. Touch imprint cytologic analysis of sentinel lymph nodes for detecting axillary metastases in patients with breast cancer. Br J Surg 86:1346-8,1999 Huvos AG, Hutter RV, Berg JW. Significance of axillary macrometastases and micrometastases in mammary cancer. Ann Surg 173:44-46,1971 Fleming ID, Cooper JS, Henson DE et al: AJCC Cancer Staging Manual, Fifth Edition, Lippincott Williams Wilkins, 1998 Association of Directors of Anatomic and Surgical Pathology. ADASP recommendations for processing and reporting lymph node specimens submitted for evaluation of metastatic disease. Am J Surg Pathol 25:961-3, 2001 Weaver DL. Sentinel lymph node biopsy in breast cancer: creating controversy and defining new standards. Adv Anat Pathol 8:65-73,2001 Papa, R: Bone marrow metastases. A review. Cancer 74: 2403-2413, 1994 Pantel K, Schlimok G, Angstwurm M et al: Methodological analysis of immunocytochemical screening for disseminated epithelial tumor cells in bone marrow. J Hematother 3: 165- 173,1994 Mathieu MC, Friedman S, Bosq J et al: Immunohistochemical staining of bone marrow biopsies for detection of occult metastasis in breast cancer. Breast Cancer Res Treat 15: 21-26,1990 Neumaier M, Gerhard M, Wagener C: Diagnosis of micrometastases by the amplification of tissue-specific genes. Gene 159: 43-47,1995 Kaplan JC, Kahn A, Chelly J: Illegitimate transcription: its use in the study of inherited disease. Hum Mutat 1: 357- 360,1992 Ruud P, Fodstad O, Hovig E: Identification of a novel cytokeratin 19 pseudogene that may interfere with reverse transcriptase- polymerase chain reaction assays used to detect micrometastatic tumor cells. Int J Cancer 80: 119-125,1999 Bostick PJ, Chatterjee S, Chi DD et al: Limitations of specific reverse-transcriptase polymerase chain reaction markers in the detection of metastases in the lymph nodes and blood of breast cancer patients. J Clin Oncol 16: 2632-2640,1998 Ghossein RA, Scher HI, Gerald, WL et al: Detection of circulating tumor cells in patients with localized and metastatic prostatic carcinoma: clinical implications. J Clin Oncol 13: 1195-1200,1995 Soeth E, Vogel I, Roder C et al: Comparative analysis of bone marrow and venous blood isolates from gastrointestinal cancer patients for the detection of disseminated tumor cells using reverse transcriptase PCR. Cancer Res 57: 3106-3610,1997 Mansi J, Gogas H, Bliss J et al: Outcome of primary-breastcancer patients with micrometastases: a long-term follow-up study. Lancet 354: 197-202, 1999 O’Sullivan GC, Collins JK, Kelly J et al: Micrometastases: marker of metastatic potential or evidence of residual disease? Gut 40: 512-515,1997 Pantel K and Otte M: Occult micrometastasis: enrichment, identification and characterization of single disseminated tumour cells. Semin Cancer Biol 11:327-337, 2001 Pantel K, Cote RJand Fodstad : Detection and clinical importance of micrometastatic disease. J Natl Cancer Inst 91: 1113- 1124, 1999 Flatmark K, Bjornland, Johannessen HO. et al: Immunomagnetic detection of Micrometastatic cells in bone marrow of colorectal cancer patients. Clin Cancer Res 8: 444-449, 2002 Fodstad O, Faye R, Hoifordt HK. et al: Immunobead-based detection and characterization of circulating tumor cells in melanoma patients. Recent Results Cancer Res 158: 40-50, 2001 Diel IJ: Bone marrow staging for breast cancer: is it better than axillary node disssection? Semin Oncol 28: 236-244, 2001 Cote RJ, Rosen PP, Leser ML. et al: Prediction of early relapse in patients with operable breast cancer by detection of occult bone marrow micrometastases. J Clin Oncol 9: 1749- 1756, 19991 Lindemann F, Schlimok G, Dirschedl P. et al: Prognostic significance of micrometastatic tumour cells in bone marrow of colorectal cancer patients. Lancet 340: 685-689, 1992 Braun S, Cevatli BS, Assemi C. et al: Comparative analysis of micrometastasis to the bone marrow and lymph nodes of node-negative breast cancer patients receiving no adjuvant therapy. J Clin Oncol 19: 1468-1475, 2001 Gerber B, Krause A, Muller H. et al: Simultaneous immunohistochemical detection of tumor cells in lymph nodes and bone marrow aspirate in breast cancer and its correlation with other prognostic factors. J Clin Oncol 19: 960-971, 2001 Mansi JL, Gogas H, Bliss JM. et al: Outcome of primarybreast- cancer patients with micrometastases: a long-term follow- up study. Lancer 354:197-202, 1999 Harbeck N, Untch M, Pache L. et al: Tumour cell detection in the bone marrow of breast cancer patients at primary therapy: results of a 3-year median follow-up. Br J Cancer 69: 566-571, 1994 Liefers GJ, Cleton-Jansen AM, van de Velde CJ. et al: Micrometastases and survival in stage II colorectal cancer. N Engl J Med 339:223-228, 1998 Jauch KW, Heiss MM, Gruetzner U. et al: Prognostic significance of bone marrow micrometastases in patients with gastric cancer. J Clin Oncol 14: 1810-1817, 1996 Schlimok G, Funke I, Pantel K. et al: Micrometastatic tumour cells in bone marrow of patients with gastric cancer: methodological aspects of detection and prognostic significance. Eur J Cancer 27: 1461-1465, 1991 Schwarz G. Cytomorphology and cell yield in a new cytocentrifugal technique allowing the collection of the cell-free supernatant. Lab med 15:45-50,1991. Pantel K, Izbicki J, Passlick B. et al: Frequency and prognostic significance of isolated tumour cells in bone marrow of patients with non-small-cell lung cancer without overt metastases. Lancer 347: 649-653, 1996 Osaki T, Oyama T, Gu CD. et al: Prognostic impact of micrometastatic tumor cells in the lymph nodes and bone marrow of patients with completely resected stage I non-smallcell lung cancer. J Clin Oncol 20: 2930-2936, 2002 Diel IJ, Cote RJ: Bone marrow and lymph node assessment for minimal residual disease in patients with breast cancer. Cancer Treat Rev 26:53-65, 2000 Diell IJ, Kaufmann M, Costa SD. et al: Micrometastatic breast cancer cells in bone marrow at primary surgery: prognostic value in comparison with nodal status. J Natl Cancer Inst 88: 1652-1658, 1996 Cote RJ, Rosen PP, Lesser M, et al: Prediction of early relapse in patients with operable breast cancer by detection of occult bone marrow micrometastases. J Clin Oncol 9: 1749- 1756,1991 Mori M, Mimori K, Ueo H et al: Molecular detection of circulating solid carcinoma cells in the peripheral blood: The concept of early sysemic disease. Int J Cancer 68: 739- 743,1996 Mori M, Mimori K, Ueo H et al: Clinical significance of molecular detection of carcinoma cells in lymph nodes and peripheral blood by reverse transcription-polymerase chain reaction in patients with gastrointestinal or breast carcinoma. J Clin Oncol 16: 128-132, 1998 Racila E, Euhus D, Weiss AJ et al: Detection and characterization of carcinoma cells in blood. Proc Natl Acad Sci USA 95: 4589-4594,1998 Berois N, Varangot M, Aizen B et al: Molecular detection of cancer cells in bone marrow and peripheral blood of patients with operable breast cancer. Comparison of CK19, MUC1 and CEA using RT-PCR. Eur J Cancer 36: 717-723, 2000 Watson MA, Fleming TP: Mammaglobin, a mammary-specific member of the uteroglobin gene family, is overexpressed in human breast cancer. Cancer Res 56: 860-865,1996 Taback B, Chan AD, Kuo CT et al: Detection of occult metastatic breast cancer cells in blood by a multimolecular marker assay: correlation with clinical stage of disease. Cancer Res 61: 8845-8850, 2001 Aihara T, Fujiwara Y, Ooka M et al: Mammaglobin B as a novel marker for detection of breast cancer micrometastases in axillary lymph nodes by reverse transcription-polymerase chain reaction. Breast Cancer Res Treat 58: 137-140, 1999 Zach O, Kasparu H, Krieger O, et al: Detection of circulating mammary carcinoma cells in the peripheral blood of breast cancer patients via a nested reverse transcriptase polymerase chain reaction assay for mammaglobin mRNA. J Clin Oncol 17: 2015-2019, 1999 Leitzel K, Lieu B, Curley E: Detection of cancer cells in peripheral blood of breast cancer patients using reverse transcription polymerase chain reaction for epidermal growth factor receptor. Clin Cancer Res 4: 3037-3043, 1998 Bouizar Z, Spyratos F, Deytieux, S et al: Polymerase chain reaction analysis of parathyroid hormone-related protein gene expression in breast cancer patients and occurrence of bone metastases. Cancer Res 53: 5076-5078, 1993 Wulf GG, Jurgens B, Liersch T et al: Reverse transcriptasepoly- merase chain reaction analysis of parathyroid hormonerelated protein for the detection of tumor cell dissemination in the peripheral blood and bone marrow of patients with breast cancer. J Cancer Res Clin Oncol 123: 514-521, 1997 Noguchi, S, Aihara, T, Motomura, K et al: Detection of breast cancer micrometastases in axillary lymph nodes by means of reverse transcriptasepolymerase chain reaction: Comparison between MUCI mRNA and keratin 19 mRNA amplification. Am J Pathol 148: 649-656, 1996 Lonn U, Lonn S, Nilsson B, Stenkvist B: Prognostic value of erbB2 and myc amplification in breast cancer imprints. Cancer 75: 2681-2687, 1995 Slamon DJ, Clark GM, Wong SG et al: Human breast cancer: correlation of relapse and survival with amplification of the HER2/neu oncogene. Science 235: 177-182, 1987 Saiki RK, Gelfand DH, Stoffel S et al: Primer-directed enzymatic amplification of DNA with a thermostable DNA polymerase. Science 239: 487-491, 1988 Slade MJ, Smith BM, Sinnett HD, Cross NCP, Coombes RC: Quantitative polymerase chain reaction for the detection of micrometastases in patients with breast cancer. J Clin Oncol 17: 870-879, 1999 Burchill SA, Bradbury MF, Pittman K et al: Detection of epithelial cancer cells in peripheral blood by reverse transcriptase polymerase chain reaction. Brit J Cancer 71: 278- 281, 1995 Smith B, Selby P, Southgate J et al: Detection of melanoma cells in peripheral blood by means of reverse transcriptase and polymerase chain reaction. Lancet 338: 1227-1229, 1991 Kunter U, Buer J, Probst M: Peripheral blood tyrosinase messenger RNA detection and survival in malignant melanoma. J Natl Cancer Inst 88: 590-594, 1996 Farthman B, Eberle J, Krasagakis K et al: RT PCR for tyrosinase mRNA positive cells in peripheral blood: evaluation strategy and correlation with known prognostic markers in 123 melanoma patients. J Invest Dermatol 110: 263-267, 1998 Mellado B, Guttierrez L, Castel T et al: Prognostic significance of the detection of circulating malignant cells by reverse transcriptase-polymerase chain reaction in long-term clinically disease-free melanoma patients. Clin Cancer Res 5: 1843- 1848, 1999 Calaluce R, Miedema BW, Yesus YW et al: Micrometastasis in colorectal carcinoma: a review. J Surg Oncol 67: 194-202, 1998 Denis MG, Lipart C, Leborgne J et al: Detection of disseminated tumor cells in peripheral blood of colorectal cancer patients. Int J Cancer 74: 540-544, 1997 McCann J: Molecular markers may improve colon cancer staging, screening. J Natl Cancer Inst 92: 1039-1040, 2000 Weitz J, Kienle P, Magener A et al: Detection of disseminated colorectal cancer cells in lymph nodes, blood and bone marrow. Clin Cancer Res 5: 1830-1836, 1999 Liefers GJ, Cleton-Jansen AM, van de Velde JH, et al: Micrometastases and survival in stage II colorectal cancer. N Engl J Med 339: 223-228, 1998 Bilchik AJ, Nora D, Tollenaar RA et al: Ultrastaging of early colon cancer using lymphatic mapping and molecular analysis. Eur J Cancer 38: 977-985, 2002 Bilchik AJ, Saha S, Wiese D, et al: Molecular staging of early colon cancer on the basis of sentinel node analysis: a multicenter phase II trial. J Clin Oncol 19: 1128-1136, 2001 Liefers GJ, Tollenar RA, Cleton-Jansen AM: Molecular staging of colorectal cancer: a step forward. Gastroenterology 116: 769-770, 1999 Futamura M, Takagi Y, Koumura, H et al: Spread of colorectal cancer micrometastases in regional lymph nodes by reverse transcriptase-polymerase chain reactions for carcinoembryonic antigen and cytokeratin. J Surg Oncol 68: 34-40, 1998 Gunn J, McCall JL, Yun K, Wright PA: Detection of micrometastases in colorectal cancer patients by K19 and K20 reverse-transcription polymerase chain reaction. Lab Invest 75: 611-616, 1996 Jung R, Petersen K, Kruger W et: Detection of micrometastasis by cytokeratin 20 RT-PCR is limited due to stable background transcription in granulocytes. Br J Cancer 81: 870- 873, 1999 Rosenberg R, Hoos A, Mueller J, Nekarda H: Impact of cytokeratin- 20 and carcinoembryonic antigen mRNA detection by RT-PCR in regional lymph nodes of patients with colorectal cancer. Br J Cancaer 83: 1323-1329, 2000 Wyld DK, Selby P, Perren TJ et al: Detection of colorectal cancer cells in peripheral blood by reverse-transcriptase polymerase chain reaction for cytokeratin 20. Int.J Cancer79: 288- 293, 1998 Merrie AE, Yun K, van Rij AM, McCall JL: Detection and significance of minimal residual disease in colorectal cancer. Histol Histopathol 14: 561-569, 1999 Soeth E, Roder C, Juhl H et al: The detection of disseminated tumor cells in bone marrow from colorectal-cancer patients by a cytokeratin-20-specific nested reverse-transcriptase-polymerasechain reaction is related to the stage of disease. Int J Cancer 69: 278-282, 1996 Tsavellas G, Patel H, Allen-Mersh TG: Detection and clinical significance of occult tumour cells in colorectal cancer. Br J Surg 88: 1307-1320, 2001 Gerhard M, Juhl H, Kalthoff H et al:Specific detection of carcinoembryonic antigen expressing tumor cells in bone marrow aspirated by polymerase chain reaction. J Clin Oncol 12: 725- 729, 1994 Wong LS, Cantrill JE, Odogwu S et al: Detection of circulating tumour cells and nodal metastasis by reverse transcriptase-polymerase chain reaction technique. Br J Surg 84: 834-839, 1997 Castells A, Boix L, Bessa X et al: Detection of colonic cells in peripheral blood of colorectal cancer patients by means of reverse transcriptase and polymerase chain reaction. Br J Cancer 78: 1368-1372, 1998 Hammar PS: Metastatic adenocarcinoma of unknown primary origin. Hum Path 29: 1319-1402, 1998 Gorstein F. Diagnostic electron microscopy of neoplasms. Hum Pathol 29:1335-1366,1999. Yu H, Levesque MA, Clark GM, et al.: Prognostic value of prostate-specific antigen for women with breast cancer: a large United States cohort study. Clin Cancer Res 4: 1489-1497, 1998 Leong ASY, Wick MR, Swanson PE: Immunohistology and ultrastructural features in site-specific epithelial neoplasms - an algorithmic approach. In:, Leong A.S-Y, Wick M.R, Swanson P.E., Eds)): Immunohistology and electron microsacopy of anaplastic and pleomorphic tumours. Cambridge Univ Press, Cambridge, 1997, pp.209-241. Hendrix MJ, Seftor EA, Seftor RE, et al.: Biologic determinants of uveal melanoma metastatic phenotype: role of intermediate filaments as predictive markers. Lab Invest 78: 153- 163, 1998 Gillespie MT, Thoma RJ, Pu ZY, et al.: Calcitonin receptors, bone sialoprotein and oesteopontin are expressed in primary breast cancer. Int J Cancer 73: 812-815, 1997 Koeneman KS, Yeung F, Chung LW: Osteomimetic properties of prostate cancer cells: a hypophesis supporting the predilection of prostate cancer metastasis and growth in the bone environment. Prostate 39: 246-261, 1999 Bertucci F, Houlgatte R, Nguyen C, et al: Gene expression profiling of cancer by use of DNA arrays: how far from the clinic? Lancet Oncol 2: 674-682, 2001 Ladanyi M, Chan WC, Triche TJ, Gerald, WL: Expression profiling of human tumors: the end of surgical pathology? J Mol Diagnostics 3: 92-97, 2001 Takemasa I, Higuchi H, Yamamoto H, et al.: Consruction of preferential cDNA microarray specialized for human colorectal carcinoma: molecular sketch of colorectal cancer. Biochem Biophys Res Commun 289: 1244-1249, 2001 Zang H, Yu C-Y, Singer B, Xiong M: Recursive partitioning for tumor classification with gene expression microarray data. PNAS 98: 6730-6735, 2001 Garber ME, Troyanskaya OG, Schluens K, et al.: Diversity of gene expression in adenocarcinoma of the lung. PNAS 98: 13784-13789, 2001 Bull JH, Ellison G, Patel A, et al.: Identification of potential diagnostic markers of prostate cancer and prostatic intraepithelial neoplasia using cDNA microarray. Br J Cancer 84: 1512-1519, 2001 Dhanasekaran SM, Barrette RT, Ghosh D, et al.: Delineation of prognostic biomarkers in prostate cancer. Nature 412: 822- 826, 2001 Ross DT, Scherf U, Eisen MB, et al.: Systematic variation in gene expression patterns in human cancer cell lines. Nature Gen 24: 227-235, 2000 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2002 VL 8 IS 3 BP 204 EP 219 PG 16 ER PT J AU Zvara, Hackler, L Nagy, BZs Micsik, T Puskas, GL AF Zvara, Agnes Hackler, Laszlo Nagy, B Zsolt Micsik, Tamas Puskas, G Laszlo TI New Molecular Methods for Classification, Diagnosis and Therapy Prediction of Hematological Malignancies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE molecular methods; DNA-chip; oncohematology ID molecular methods; DNA-chip; oncohematology AB Normal functions of the cell are based on the precise regulation of various genes. If this strict regulation and the hierarchy of genes becomes upset due to flaws in this system, the result will be cellular dysfunction which eventually may lead to carcinogenic transformation. Two basic challenges of the classification of cancers are the discovery of new molecular markers characteristic to defined disease groups and the classification of already diagnosed or new cases into existing groups. This precise classification may open the door to tailored treatment or project the expected outcome of the disease. Today there is unlimited access available to the databases containing sequences and localization of the genes within the confines of Human Genome project. It provides significant help for the discovery of chromosome abnormalities and systematic analysis of gene expression patterns. This is important not only to understand normal functions of the cells, but it also contributes to the identification of new genes that are characteristic to given disease groups as markers and that are potential drug targets. Until the second half of the twentieth century the study of the function and regulation of genes was based on step-by-step investigation of individual genes. Regarding the fact, that the genomes of an increasing number of organisms have become known in whole or in part, numerous new techniques have been developed that facilitated the systematic analysis of gene functions. The aim of this study is to summarize the new, molecular based possibilities for classification, diagnosis and prognosis of hematological malignancies, as well as to summarize the main results of these areas. C1 [Zvara, Agnes] University of Szeged, Department of Medical Genetics, Temesvari krt. 62, H-6726 Szeged, Hungary. [Hackler, Laszlo] University of Szeged, Department of Medical Genetics, Temesvari krt. 62, H-6726 Szeged, Hungary. [Nagy, B Zsolt] University of Szeged, Department of Medical Genetics, Temesvari krt. 62, H-6726 Szeged, Hungary. [Micsik, Tamas] University of Szeged, Department of Medical Genetics, Temesvari krt. 62, H-6726 Szeged, Hungary. [Puskas, G Laszlo] University of Szeged, Department of Medical Genetics, Temesvari krt. 62, H-6726 Szeged, Hungary. RP Puskas, GL (reprint author), University of Szeged, Department of Medical Genetics, H-6726 Szeged, Hungary. CR Adorjan P, Distler J, Lipscher E, et al:Tumour class prediction and discovery by microarray-based DNA methylation analysis. Nucleic Acids Res 30:e21, 2002. Albala JS and Humphrey-Smith I: Array-based proteomics: high-throughput expression and purification of IMAGE consortium cDNA clones. Curr Opin Mol Ther 680–684, 1999. Alizadeh AA, Ross DT, Perou CM, et al: Towards a novel classification of human malignancies based on gene expression patterns. J Pathol 195: 41-52, 2001. Alwine JC, Kemp DJ, Stark GR: Method for detection of specific RNAs in agarose gels by transfer to diazobenzyloxymethyl- paper and hybridization with DNA probes. Proc Natl Acad Sci USA 74: 5350-5354, 1977. Appelbaum FR: Molecular Diagnosis and Clinical Decisions in Adult Acute Leukemia, Seminars in Hematology 36: 401-410, 1999. Arico M, Valsecchi MG, Camitta B et al: Outcome of treatment in children with Philadelphia chromosome-positive acute lymphoblastic leukemia. N Engl J Med 342: 998-1006, 2000. Aubele M, Auer G, Braselmann H et al: Chromosomal imbalances are associated with metastasis-free survival in breast cancer patients. Anal Cell Pathol 24: 77-87, 2002. Bittner M, Meltzer P, Chen Y et al: Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature 406: 536-540, 2000. Blohm DH, Guiseppi-Elie A: New developments in microarray technology. Curr Opin Biotechnol 12: 41-47, 2001. Bruckert P, Kappler R, Scherthan H et al: Double minutes and c-MYC amplification in acute myelogenous leukemia: Are they prognostic factors? Cancer Genet Cytogenet 120: 73-79, 2000. Cahill DJ: Protein arrays: a high-throughput solution for proteomics research? Proteomics: A Trends Guide, 47–51, 2000. Chan MF, Liang G and Jones PA: Relationship between transcription and DNA methylation. Curr Top Microbiol Immunol 249: 75–86, 2000. Chen KT, Lin JD, Chao TC et al: Identifying differentially expressed genes associated with metastasis of follicular thyroid cancer by cDNA expression array. Thyroid 11: 41-46, 2001. Clark J, Edwards S, John M et al: Identification of amplified and expressed genes in breast cancer by comparative hybridization onto microarrays of randomly selected cDNA clones. Genes Chromosomes Cancer 34: 104-114, 2002. Costello JF, Fruhwald MC, Smiraglia DJ et al: Aberrant CpGisland methylation has non-random and tumour-type-specific patterns. Nature Genet 24: 132–138, 2000. DeRisi J, Penland L, Brown P et al: Use of a cDNA microarray to analyse gene expression patterns in human cancer. Nature Genet 14: 457-460, 1996. Dunican DS, McWilliam P, Tighe O et al: Gene expression differences between the microsatellite instability, MIN, and chromosomal instability, CIN, phenotypes in colorectal cancer revealed by high-density cDNA array hybridization. Oncogene 21: 3253-3257, 2002. Eads CA, Danenberg KD, Kawakami K et al: MethyLight: a high-throughput assay to measure DNA methylation. Nucleic Acids Res 28-c32, 2000. Emili AQ and Cagney G: Large-scale functional analysis using peptide or protein arrays. Nat. Biotechnol 18: 393–397, 2000. Ernst T, Hergenhahn M, Kenzelmann M et al: Decrease and gain of gene expression are equally discriminatory markers for prostate carcinoma: a gene expression analysis on total and microdissected prostate tissue. Am J Pathol 160: 2169-2180, 2002. Esteller M, Corn PG, Baylin SB et al: A gene hypermethylation profile of human cancer. Cancer Res 61: 3225–3229, 2001. Ferrando AA, Look AT: Clinical implications of recurring chromosomal and associated molecular abnormalities in acute lymphoblastic leukemia. Semin Hematol 37: 381-95, 2000. Ferrando AA, Neuberg DS, Staunton J, et al: Gene expression signatures define novel oncogenic pathways in T cell acute lymphoblastic leukemia. Cancer Cell, 1: 75-87, 2002. Furak JI, Trojan T, Szoke L et al: Development of Brain Metastasis 5 Years Before the Appearance of the Primary Lung Cancer: Messenger Metachronous Metastasis. Annals Thoracic Surg, in press). Gitan RS, Shi H, Chen CM et al: Methylation-specific oligonucleotide microarray: a new potential for high-throughput methylation analysis. Genome Res 12: 158-164, 2001. Gilliland DG: Molecular Genetics of Human Leukemia. Leukemia 12: S7-S12, 1998. Golub TR: The Genetics of AML: An Update in Proceedings of the American Society of Hematology, pp. 102-111, 1999. Golub TR, Slonim DK, Tamayo P et al.: Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. Science 286: 531-537, 1999. Haab BB Dunham MJ, Brown PO: Protein microarrays for highly parallel detection and quantitation of specific proteins and antibodies in complex solutions. Genome Biol 2: Research 0004.1–0004.13, 2001. Hanash SM, Madoz-Gurpide J, Misek DE: Identification of novel targets for cancer therapy using expression proteomics. Leukemia 16: 478-485, 2002. Hedenfalk I, Duggan D, Chen Y et al: Gene-expression profiles in hereditary breast cancer. N Engl J Med 344: 539-548, 2001. Hedrick SM, Cohen DI, Nielsen EA et al: Isolation of cDNA clones encoding T cell-specific membrane-associated proteins. Nature 308: 149-153, 1984. Hodgson G, Hager JH, Volik S et al: Genome scanning with array CGH delineates regional alterations in mouse islet carcinomas. Nat Genet 29: 459-464, 2001. Houldsworth J, Chaganti RS: Comparative genomic hybridization: an overview. Am J Pathol 145: 1253-1260, 1994. Huang TH-M, Perry MR and Laux DE: Methylation profiling of CpG islands in human breast cancer cells. Hum Mol Genet 8: 459–470, 1999. Huang Y, Prasad M, Lemon WJ et al: Gene expression in papillary thyroid carcinoma reveals highly consistent profiles. Proc Natl Acad Sci USA 98: 15044-15049, 2001. Jiang Y, Harlocker SL, Molesh DA et al: Discovery of differentially expressed genes in human breast cancer using subtracted cDNA libraries and cDNA microarrays. Oncogene 21: 2270-2282, 2002. Jones PA: DNA methylation errors and cancer. Cancer Res 65: 2463–2467, 1996. Kanerva J, Niini T, Vettenranta K et al: Loss at 12p detected by comparative genomic hybridization, CGH): association with TEL-AML1 fusion and favorable prognostic features in childhood acute lymphoblastic leukemia, ALL). A multi-institutional study. Med Pediatr Oncol 37: 419-425, 2001. Kitajka K, Puskas LG, Zvara A et al: The role of n-3 polyunsaturated fatty acids in brain: Modulation of rat brain gene expression by dietary n-3 fatty acids. Proc Natl Acad Sci USA 99, 2619-2624, 2002. Kopper L, Timar J: Gene expression profiles in the diagnosis and prognosis of cancer. Magy Onkol 46:3-9, 2002. Kozian DH, Kirschbaum BJ: Comparative gene-expression analysis. Trends Biotechnol 17: 73-78, 1999. Kroll T, Odyvanova L, Clement JH et al: Molecular characterization of breast cancer cell lines by expression profiling. J Cancer Res Clin Oncol 128: 125-134, 2002. Liang P, Pardee A et al: Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction. Science 257: 967-971, 1992. Lin YM, Furukawa Y, Tsunoda T et al: Molecular diagnosis of colorectal tumors by expression profiles of 50 genes expressed differentially in adenomas and carcinomas. Oncogene 21: 4120-4128, 2002. Lisitsyn N, Wigler M et al: Cloning the differences between two complex genomes. Science 259: 946-951, 1993. Luo JH, Yu YP, Cieply K et al: Gene expression analysis of prostate cancers. Mol Carcinog 33: 25-35, 2002. Mitelman F, Mertens F, Johansson B: A breakpoint map of recurrent chromosomal rearrangements in human neoplasia. Nat Genet Spec No: 417-474, 1997. Novak U, Oppliger Leibundgut E, Hager J et a: A high-resolution allelotype of B-cell chronic lymphocytic leukemia, BCLL). Blood 100: 1787-1794, 2002. Pinkel D, Segraves R, Sudar D et al: High resolution analysis of DNA copy number variation using comparative genomic hybridization to microarrays. Nat Genet 20: 207-11, 1998. Prashar Y, Weissman S et al: Analysis of differential gene expression by display of 3’ end restriction fragments of cDNAs. Proc Nat Acad Sci USA 93: 659-663, 1996. Pui CH, Campana D, Evans WE: Childhood acute lymphoblastic leukaemia current status and future perspectives. Lancet Oncol 10: 597-607, 2001. Puskas LG, Zvara A, Hackler JrL et al: RNA amplification results in reproducible microarray data with slight ratio biases. Biotechniques 32: 1330-1342, 2002. Puskas LG, Zvara A, Hackler JrL et al: Production of bulk amounts of universal RNA for DNA-microarrays. Biotechniques 33: 898-900, 902, 904, 2002. Ramaswamy S, Tamayo P, Rifkin R et al: Multiclass cancer diagnosis using tumor gene expression signatures. Proc Natl Acad Sci USA 98: 15149-15154, 2001. Rubnitz JE and Pui C-H: Leukemias, in Principles of Molecular Medicine, pp. 233-239., J.L. Jameson ed., Humana Press, NJ, 1998. Schlossman SF, Chess L, Humphreys RE et al: Distribution of Ia-like molecules on the surface of normal and leukemic human cells. Proc Natl Acad Sci USA 73: 1288-1292, 1976. Schwarze SR, DePrimo SE, Grabert LM et al: Novel pathways associated with bypassing cellular senescence in human prostate epithelial cells. J Biol Chem 277: 14877-14883, 2002. Shipp MA, Ross KN, Tamayo P et al: Diffuse large B-cell lymphoma outcome prediction by gene-expression profiling and supervised machine learning. Nat Med. 8: 68-74, 2002. Shou J, Soriano R, Hayward SW et al: Expression profiling of a human cell line model of prostatic cancer reveals a direct involvement of interferon signaling in prostate tumor progression. Proc Natl Acad Sci USA 99: 2830-2835, 2002. Sidransky D: Emerging molecular markers of cancer. Nature Rev Cancer 2: 210-219, 2002. Stremmel C, Wein A, Hohenberger W et al: DNA microarrays: a new diagnostic tool and its implications in colorectal cancer. Int J Colorectal Dis 17: 131-136, 2002. Suzuki H, Gabrielson E, Chen W et al: A genomic screen for genes upregulated by demethylation and histone deacetylase inhibition in human colorectal cancer. Nat Genet 31: 141-149, 2002. Swaroop A, Xu JZ, Agarwal N et al.: A simple and efficient cDNA library subtraction procedure: isolation of human retinaspecific cDNA clones. Nucleic Acids Res 19: 1954, 1991. Sweetser DA, Chen CS, Blomberg AA et al: Loss of heterozygosity in childhood de novo acute myelogenous leukemia. Blood 98: 1188-94, 2001. Takano T, Hasegawa Y, Matsuzuka F et al: Gene expression profiles in thyroid carcinomas Br J Cancer 83: 1495-502, 2000. Toyota M, Ho C, Ahuja N et al: Identification of differentially methylated sequences in colorectal cancer by methylated CpG island amplification. Cancer Res 59: 2307–2312, 1999. Tsukasaki K, Krebs J, Nagai K et al: Comparative genomic hybridization analysis in adult T-cell leukemia/lymphoma: correlation with clinical course. Blood 97: 3875-3881, 2001. Yeoh EJ, Ross ME, Shurtleff SAA et al: Classification, subtype discovery, and prediction of outcome in pediatric acute lymphoblastic leukemia by gene expression profiling. Cancer Cell 1: 133-143, 2002. Velculescu VE, Zhang L, Vogelstein B et al: Serial analysis of gene expression. Science 270: 464-467, 1995. Vogeli-Lange R, Burckert N, Boiler T et al: Rapid selectionand classification of positive clones generated by mRNA differential display. Nucleic Acids Res 24: 1385-1386, 1996. Weinberger SR, Morris TS, Pawlak M: Recent trends in protein biochip technology. Pharmacogenomics 1: 395-416, 2000. Wieser R: Rearrangements of chromosome band 3q21 in myeloid leukemia. Leuk Lymphoma 43: 59-65, 2002. Wilhelm M, Veltman JA, Olshen AB et al: Array-based comparative genomic hybridization for the differential diagnosis of renal cell cancer. Cancer Res 62: 957-960, 2002. de Wit NJ, Burtscher HJ, Weidle UH et al: Differentially expressed genes identified in human melanoma cell lines with different metastatic behaviour using high density oligonucleotide arrays. Melanoma Res 12: 57-69, 2002. Wreesmann VB, Ghossein RA, Patel SG et al: Genome-wide appraisal of thyroid cancer progression. Am J Pathol 161: 1549-1556, 2002. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 231 EP 240 PG 10 ER PT J AU Girolami, F Passerini, I Gargano, D Frusconi, S Villari, D Nicita, G Torricelli, F AF Girolami, Francesca Passerini, Ilaria Gargano, Dorotea Frusconi, Sabrina Villari, Donata Nicita, Giulio Torricelli, Francesca TI Microsatellite Analysis of Chromosome 3p Region in Sporadic Renal Cell Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microsatellite instability; renal cell carcinoma; LOH; RER ID microsatellite instability; renal cell carcinoma; LOH; RER AB The etiology and progression of renal carcinomas (RCC) is still poorly understood. RCC have been classified into several pathological entities. The most frequent type, clear cell carcinoma, accounts for about 80% of sporadic RCC and shows several chromosome abnormalities documented both by conventional cytogenetics, loss of eterozygosity (LOH) and replication error (RER) studies. In 10 clear cell type sporadic RCC we evaluated LOH and RER using a set of 10 microsatellite markers covering the chromosome 3p region, which has been suggested for interstitial deletions. Electrophoresis was performed by automated sequencer ABI Prism 377 and data were analyzed with Genescan and Genotyper 2.5 softwares. We revealed allelic loss in 48,7% of informative microsatellites and a single case of RER. We found the highest LOH frequency in 3p25-26 region where maps Von Hippel-Lindau (VHL) oncosuppressor gene. In addition, DNA hypermethylation, an alternative mechanism of VHL gene silencing, was evaluated by methylation-specific PCR. However hypermethylation status was not detected in any of our tumor samples. C1 [Girolami, Francesca] Azienda Ospedaliera Careggi and II Urological Clinic, Cytogenetics and Genetics Unit, V.le Morgagni n.85, 50134 Firenze, Italy. [Passerini, Ilaria] Azienda Ospedaliera Careggi and II Urological Clinic, Cytogenetics and Genetics Unit, V.le Morgagni n.85, 50134 Firenze, Italy. [Gargano, Dorotea] Azienda Ospedaliera Careggi and II Urological Clinic, Cytogenetics and Genetics Unit, V.le Morgagni n.85, 50134 Firenze, Italy. [Frusconi, Sabrina] Azienda Ospedaliera Careggi and II Urological Clinic, Cytogenetics and Genetics Unit, V.le Morgagni n.85, 50134 Firenze, Italy. [Villari, Donata] Azienda Ospedaliera Careggi and II Urological Clinic, Cytogenetics and Genetics Unit, V.le Morgagni n.85, 50134 Firenze, Italy. [Nicita, Giulio] Azienda Ospedaliera Careggi and II Urological Clinic, Cytogenetics and Genetics Unit, V.le Morgagni n.85, 50134 Firenze, Italy. [Torricelli, Francesca] Azienda Ospedaliera Careggi and II Urological Clinic, Cytogenetics and Genetics Unit, V.le Morgagni n.85, 50134 Firenze, Italy. RP Girolami, F (reprint author), Azienda Ospedaliera Careggi and II Urological Clinic, Cytogenetics and Genetics Unit, 50134 Firenze, Italy. EM citogenbibl@ao-careggi.toscana.it CR Erlandsson R: Molecular genetics of renal cell carcinoma. Cancer Genet Cytogenet 104: 1-18, 1998 Diakoumis E, Sourvinos G, Kiaris H, et al: Genetic instability in renal cell carcinoma. Eur Urol 33: 227-232, 1998 Chudek J, Wilhelm M, Bugert P, et al: Detailed microsatellite analysis of chromosome 3p region in non-papillary renal cell carcinomas. Int J Cancer 73: 225-229, 1997 Orikasa k, Orikasa S, and Horii A: Identification of a 700-kb region of common allelic loss in chromosome bands 3p14.3- p21.1 in human renal cell carcinoma. Cancer Genet Cytogenet 104: 104-110, 1998 Wilhelm M, Bugert P, Kenck C, et al: Terminal deletion of chromosome3p sequences in nonpapillary renal cell carcinomas: a breakpoint cluster between loci D3S1285 and D3S1603. Cancer Research 55: 5383-5385, 1995 Bugert P, Kench C, Wilhelm M, et al: Refining a proximal breakpoint cluster at chromosome 3p11.2 in non-papillary renal cell carcinomas. Int J Cancer 68: 723-726, 1996 Bernues M, Casadeval C, Miro’ R, et al: Analysis of 3p allelic losses in renal carcinomas: comparison with cytogenetic results. Cancer Genet Cytogenet 107: 121-124, 1998 Willers CP, Siebert R, Bardenheuer W, et al: Genetic instability of 3p12-p21 specific microsatellite sequences in renal cell carcinoma. Br J Urol 77: 524-529, 1996 Van den Berg A and Buys CHCM: Involvement of multiple loci on chromosome 3 in renal cell cancer development. Genes Chromosom Cancer 19: 59-76, 1997 Kok K, Draaijers TG, Mosselaar A, et al: Inclusion of new microsatellite repeats in allelic loss analysis escludes retention of heterozygosity in the renal cell carcinoma critical region in 3p21. Cancer Genet Cytogenet 116: 40-43, 2000 Canzian F, Salovaara R, Hemminki A, et al: Semiautomated assessment of loss of heterozygosity and replication error in Tumors. Cancer Research 56: 3331-3337, 1996 Clifford SC, Prowse AH, Affara NA, et al: Inactivation of the Von Hippel Lindau, VHL, tumor suppressor gene and allelic losses at chromosome arm 3p in primary renal cell carcinoma: evidence for a VHL indipendent pathway in clear cell renal tumorigenesis. Genes Chromosomes Cancer 22: 200-209, 1998 Prowse A H, Webster A R, Richards F M, et al: Somatic inactivation of the VHL gene in Von Hippel-Lindau disease tumors. Am J Hum Genet 60:765-771, 1997 Martinez A, Fullwood P, Kondo K, et al: Role of chromosome 3p12-p21 tumor suppressor genes in clear cell renal cell carcinoma: analysis of VHL dependent and VHL independent pathways of tumorigenesis. Pathol 53: 137-144, 2000 Bugert P, and Kovacs G: Molecular differential diagnosis of renal cell carcinomas by microsatellite analysis. Am J Pathol 149: 2081-2088, 1996 Zatyka M, Morrissey C, Kuzmin I, et al: Genetic and functional analysis of the von Hippel-Lindau, VHL, tumour suppressor gene promoter. J Med Genet 39: 463-472, 2002 Velickovic M, Delahunt B, Storkel S, et al: VHL and FHIT locus loss of heterozygosity in common in all renal cancer morphotypes but differs in pattern and prognostic significance. Cancer Res 61: 4815-4819, 2001 Meyer AJ, Hernandez A, Florl AR, et al: Novel mutations of the Von Hippel Lindau tumor- suppressor gene and rare DNA hypermethylation in renal cell carcinoma cell lines of the clearcell type. Int J Cancer 87:650-653, 2000 Chino K, Esumi M, Ishida H, et al: Characteristic loss of heterozygosity in chromosome 3p and low frequency of replication errors in sporadic renal cell carcinoma. J Urol 162: 614- 618, 1999 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 241 EP 244 PG 4 ER PT J AU Smardova, J Pavlova, S Koukalova, H AF Smardova, Jana Pavlova, Sarka Koukalova, Hana TI Determination of Optimal Conditions for Analysis of p53 Status in Leukemic Cells Using Functional Analysis of Separated Alleles in Yeast SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tumor suppressor p53; FASAY; DNA polymerase fidelity; mRNA stability ID tumor suppressor p53; FASAY; DNA polymerase fidelity; mRNA stability AB Tumor suppressor p53 is transcription factor that participates in control of many cellular functions. Somatic mutations of the p53 gene are frequently detected in human cancers. Several methods can be used for identification of p53 mutations, including FASAY - functional analysis of separated alleles in yeast. FASAY distinguishes yeast colonies expressing functional p53 protein from colonies producing a dysfunctional p53 protein simply on the basis of color. The validity of the method depends on a low background level. There are several sources of background as PCR-induced point mutations, low quality of RNA and alternative splicing of intron 9 affecting the p53 carboxy-terminus. In the present work we show that FASAY can be successfully used for analysis of mRNA isolated from blood samples that were collected and stored for 24 hours at 0°C without undesired increase of background. We also measured fidelity of several commonly used DNA polymerases and determined the most suitable kinds of Pfu DNA polymerases for FASAY. Reaction conditions described in this report allow routine analysis of p53 status in leukemic cells using FASAY. C1 [Smardova, Jana] Masaryk Memorial Cancer Institute, Department of Experimental Oncology, Zluty kopec 7, 656 53 Brno, Czech Republic. [Pavlova, Sarka] Masaryk Memorial Cancer Institute, Department of Experimental Oncology, Zluty kopec 7, 656 53 Brno, Czech Republic. [Koukalova, Hana] Masaryk Memorial Cancer Institute, Department of Experimental Oncology, Zluty kopec 7, 656 53 Brno, Czech Republic. RP Smardova, J (reprint author), Masaryk Memorial Cancer Institute, Department of Experimental Oncology, 656 53 Brno, Czech Republic. EM smardova@mou.cz CR Baker SJ, Markowitz S, Fearon ER, et al: Suppression of human colorectal carcinoma cell growth by wild-type p53. Science 249: 912-915, 1990 Bi S, Hughes T, Bungey J, et al: p53 in chronic myeloid leukemia cell lines. Leukemia 6: 839-842, 1992 Camplejohn RS, Rutheford J: p53 functional assay: detecting p53 mutations in both the germline and in sporadic tumours. Cell Prolif 34: 1-14, 2001 Di Como CJ, Prives C: Human tumor-derived p53 proteins exhibit binding site selectivity and temperature sensitivity for transactivation in a yeast-based assay. Oncogene 16: 2527- 2539, 1998 Duddy PM, Hanby AM, Barnes DM, Camplejohn RS: Improving the detection of p53 mutations in breast cancer by use of the FASAY, a functional Assay J Mol Diagnostics 2: 139-144, 2000 Flaman JM, Frebourg T, Moreau V, et al: A rapid PCR fidelity assay. Nucl Acid Res 22: 3259-3260, 1994 Flaman JM., Frebourg T, Moreau V, et al: A simple p53 functional assay for screening cell lines, blood, and tumors. Proc Natl Acad Sci USA 92: 3963-3967, 1995 Flaman JM, Waridel F, Estreicher A, et al: The human tumour suppressor gene p53 is alternatively spliced in normal cells. Oncogene 12: 813-818, 1996 Flaman JM, Robert V, Lenglet S, et al: Identification of of human p53 mutations with differential effects on the bax and p21 promoters using functional assay in yeast. Oncogene 16: 1369-1372, 1998 Inga A, Iannone R, Monti P, et al: Determining mutational fingerprints at the human p53 locus with a yeast functional assay: A new tool for molecular epidemiology. Oncogene 14: 1307- 1313, 1997 Ishioka C, Frebourg T, Yan YX, et al:Screening patients for heterozygous p53 mutations using a functional assay in yeast. Nat.Genet. 5: 124-129, 1993 Kashiwazaki H, Tonoki H, Tada M, et al: High frequency of p53 mutations in human oral epithelial dysplasia and primary squamous cell carcinoma detected by yeast functional assay. Oncogene 15: 2667-2674, 1997 Meinhold-Heerlein I, Ninci E, Ikenberg H, et al: Evaluation of methods to detect p53 mutations in ovarian cancer. Oncology 60: 176-188, 2001 O´Connor PM, Jackman J, Bae I, et al: Characterization of the p53 tumor suppressor pathway in cell lines of the National cancer institute anticancer drug screen and correlations with the growth inhibitory potency of 123 anticancer agents. Cancer Res 57: 4285-4300, 1997 Rizzo MG, Zepparoni A, Cristofanelli B, et al: Wt-p53 action in human leukaemia cell lines corresponding to different stages of differentiation. Brit J Cancer 77: 1429-1438, 1998 Smardova J, Vagunda V, Jandakova E, et al:p53 status in breast carcinomas revealed by FASAY correlates well with p53 protein accumulation determined by immunohistochemistry. Neoplasma 46: 384-389, 1999 Smardova J, Nemajerova A, Trbusek M, et al: Rare somatic p53 mutation identified in breast cancer:A case report. Tumor Biol 22: 59-66, 2001 Soussi T, Legros Y, Lubin R, et al: Multifactorial analysis of p53 alteration in human cancer: A review. Int. J. Cancer Inst. 88: 173-182, 1996 Stotz A, Linder P: ADE2 gene from Saccharomyces cerevisiae: Sequence and new vwctors. Gene 95: 91-98, 1990 Waridel F, Estreicher A, Bron L, et al: Field cancerisation and polyclonal p53 mutation in the upper aerodigestive tract. Oncogene 14: 163-169, 1997 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 245 EP 251 PG 7 ER PT J AU Ghaderi, A Vasei, M Maleck-Hosseini, AS Gharesi-Fard, B Khodami, M Doroudchi, M Modjtahedi, H AF Ghaderi, Abbas Vasei, Mohammad Maleck-Hosseini, A S Gharesi-Fard, Behronz Khodami, Maliheh Doroudchi, Mehrnoosh Modjtahedi, Helmout TI The Expression of c-erbB-1 and c-erbB-2 in Iranian Patients with Gastric Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE c-erbB-1; c-erbB-2; gastric carcinoma; southern Iranian ID c-erbB-1; c-erbB-2; gastric carcinoma; southern Iranian AB To assess the significance of epidermal growth factor receptor family members, the overexpression of c-erbB-1 and c-erbB-2 was retrospectively investigated in 146 southern Iranian gastric cancer patients. Indirect immunostaining was used to evaluate the expression of these two receptors in formalin-fixed paraffin-embedded tissue samples. c-ErbB-1 expression was observed in 47 (32.2%) and c-erbB-2 expression was observed in 24 (16.4%) of tumors. Significant positive correlations were observed between c-erbB-1 expression and tumor size, local invasion, lymph node involvement and tumor stage. There was also a negative correlation between c-erbB-2 expression and tumor stage. These results may suggest the contribution of c-erbB-1 molecule in progression of gastric carcinomas in southern Iranian patients. Moreover, the relatively high percentage of c-erbB-2 positive tumors may provide a useful target for the immunotherapy of these cancers. C1 [Ghaderi, Abbas] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Vasei, Mohammad] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Maleck-Hosseini, A S] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Gharesi-Fard, Behronz] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Khodami, Maliheh] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Doroudchi, Mehrnoosh] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Modjtahedi, Helmout] Medical School, Shiraz University of Medical Sciences, Department of SurgeryShiraz, Iran. RP Ghaderi, A (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Pathology, Shiraz, Iran. CR Farahmandbeigi M, Kadivar MR: The incidence rate of registered cancers in Fars province. Disease Control Unit, Shiraz University Press, Iran, 2000. Koyama S, Maruyama T, Adachi S: Expression of epidermal growth factor receptor and CD44 splicing variants sharing exons 6 and 9 on gastric and esophageal carcinomas: a twocolor flow-cytometric analysis. J Cancer Res Clin Oncol 125: 47-54, 1999. Prakash I, Mathur RP, Kar P, et al: Comparative evaluation of cell proliferative indices and epidermal growth factor receptor expression in gastric carcinoma. Indi J Pathol Microbiol 40:481-490, 1997. Jang WI, Yang WI, Lee CI, et al: Immunohistochemical detection of p53 protein, c-erbB-2 protein, epidermal growth factor receptor protein and proliferating cell nuclear antigen in gastric carcinoma. J Korean Med Sci 8: 293-304, 1993. Oshima CT, Lanzoni VP, Iriya K, Forones NM:C-erbB-2 oncoprotein in gastric carcinoma: correlation with clinical stage and prognosis. Int J Biol Markers 16: 250-254, 2001. Wu MS, Shun CT, Sheu JC, et al: Overexpression of mutant p53 and c-erbB-2 proteins and mutations of the p15 and p16 genes in human gastric carcinoma: with respect to histological subtypes and stages. J Gastroenterol Hepatol 13: 305-310, 1998. Lee HR, Kim JH, Uhm HD, et al: Overexpression of c-ErbB-2 protein in gastric cancer by immunohistochemical stain. Oncology 53: 192-197, 1996. Aoyagi K, Kohfuji K, Yano S, et al: Evaluation of the epidermal growth factor receptor, EGFR, and c-erbB-2 in superspreading- type and penetrating-type gastric carcinoma. Kurume Med J 48:197-200, 2001. Motojima K, Furui J, Kohara N, et al: erbB-2 expression in welldifferentiated adenocarcinoma of the stomach predicts shorter survival after curative resection. Surgery 115:349-354, 1994. Yoshida K, Yasui W, Ito H, Tahara E: Growth factors in progression of human esophageal and gastric carcinomas. Exp Pathol 40: 291-300, 1990. Lee EY, Cibull ML, Strodel WE, Haley JV: Expression of HER- 2/neu oncoprotein and epidermal growth factor receptor and prognosis in gastric carcinoma. Arch Pathol Lab Med 118: 235-239, 1994. Orita H, Maehara Y, Emi Y, et al: c-erbB-2 expression is predictive for lymphatic spread of clinical gastric carcinoma. Hepatogastroenterology 44: 294-298, 1997. Tokunaga A, Onda M, Okuda T, et al: Prevention of growth of a human gastric cancer xenograft in nude mice with anti-EGF receptor ntibody. In: Takahashi T, editor. Recent advances in management of digestive cancers. Tokyo: Springer-Verlag, 403-405, 1993. Teramoto T, Onda M, Tokunaga A, Asano G: Inhibitory effect of anti-epidermal growth factor receptor antibody on a human gastric cancer. Cancer 77s: 1639-1645, 1996. Ross JS, McKenna BJ: The HER-2/neu oncogene in tumors of the gastrointestinal tract. Cancer Invest 19:554-568, 2001. Modjtahedi H, Hickish T, Nicolson M, et al: Phase I trial and tumour localisation of the anti-EGFR monoclonal antibody ICR62 in head and neck or lung cancer. Br J Cancer 73: 228- 235, 1996. Bianco C, Bianco R, Tortora G, et al: Antitumor activity of combined treatment of human cancer cells with ionizing radiation and anti-epidermal growth factor receptor monoclonal antibody C225 plus type I protein kinase A antisense oligonucleotide. Clin Cancer Res 6: 4343-4350, 2000. Ciardiello F, Bianco R, Damiano V, et al: Antiangiogenic and antitumor activity of anti-epidermal growth factor receptor C225 monoclonal antibody in combination with vascular endothelial growth factor antisense oligonucleotide in human GEO colon cancer cells. Clin Cancer Res 6: 3739-47, 2000. Ciardiello F, Bianco R, Damiano V, et al: Antitumor activity of sequential treatment with topotecan and anti-epidermal growth factor receptor monoclonal antibody C225. Clin Cancer Res 5: 909-916, 1999. Falcey J, Pfister D, Cohen R, et al: A study of anti-epidermal growth factor receptor, EGFr, monoclonal antibody C225 and cisplastin in patients with head and neck or lung carcinomas. Proc Am Soc Clin Oncol 16: 1364, 1997. Cobleigh MA, Vogel CL, Tripathy D, et al: Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol 17: 2639-2648, 1999. Slamon DJ, Leyland-Jones B, Shak S, et al: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 344: 783-792, 2001. Vogel CL, Cobleigh MA, Tripathy D, et al: Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J Clin Oncol 20: 719-726, 2002. Cook-Bruns N: Retrospective analysis of the safety of Herceptin immunotherapy in metastatic breast cancer. Oncology 61s: 58-66, 2001. Azzoli CG, Krug LM, Miller VA, et al:Trastuzumab in the treatment of non-small cell lung cancer. Semin Oncol 29s: 59-65, 2002. Zinner RG, Kim J, Herbst RS: Non-small cell lung cancer clinical trials with trastuzumab: their foundation and preliminary results. Lung Cancer 37: 17-27, 2002. Leyland-Jones B: Maximizing the response to Herceptin therapy through optimal use and patient selection. Anticancer Drugs 4s: 11-17, 2001. Modjtahedi H, Eccles SA, Box G, et al: Antitumor activity of combinations of antibodies directed against different epitopes on the extracellular domain of the human EGF receptor. Cell Biophys 22: 129-146, 1993. Styles JM, Harrison S, Gusterson BA, Dean CJ: Rat monoclonal antibodies to the external domain of the product of the C-erbB-2 proto-oncogene. Int J Cancer 45: 320-324, 1990. Walker RA:The erbB/HER type1 tyrosine kinase receptor family. J Pathol 185: 234-235, 1998. Peles E, Yarden Y: Neu and its ligands: from an oncogene to neural factors. Bioessays 15: 815-824, 1993. Takehana T, Kunitomo K, Kono K, et al: Status of c-erbB-2 in gastric adenocarcinoma: a comparative study of immunohistochemistry, fluorescence in situ hybridization and enzymelinked immuno-sorbent assay. Int J Cancer 98: 833-837, 2002. Gharesi-Fard B, Vasei M, Talei A, et al: The expression and prognostic significance of c-erbB-2 molecules in patients with breast cancer in Iran. Irn J Med Sci 25: 31-35, 2000. Khademi B, Shirazi FM, Vasei M, et al: The expression of p53, c-erbB-1 and c-erbB-2 molecules and their correlation with prognostic markers in patients with head and neck tumors. Cancer Lett 184: 223-230, 2002. Shimada E, Kato M, Saito Y: Immunohistochemical study of the c-erbB-2 protein in human gastric carcinoma. Nippon Geka Gakkai Zasshi 94: 33-40, 1993. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 252 EP 256 PG 5 ER PT J AU Elo, J Balatoni, Zs Kotai, Zs Bartfai, R AF Elo, Janos Balatoni, Zsuzsa Kotai, Zsuzsa Bartfai, Reka TI Considerations in the Treatment of the Node-negative (N0) Neck in Glottic Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glottic cancer; N0 neck; predictive factors; therapy ID Glottic cancer; N0 neck; predictive factors; therapy AB Treatment of lymph node negative (N0) glottic carcinoma has raised numerous controversy for decades. Prevention is one of the oldest axioms in medicine. On the other hand, overtreatment can cause unnecessary harm to patients. This retrospective study was performed in 206 patients having glottic cancers with clinically node-negative (N0) necks. The aim of this assessment is to deal with the diagnosis, predictive factors and surgical therapy of occult metastases of squamous cell cancers originating from the glottic region. The examinations were performed in three phases. Preoperative clinical, histological - and in selective cases - imaging were carried out to separate high-risk patients. Intraoperative cases of open surgery after U-shaped skin preparation up to the hyoid bone with direct inspection of jugular lymph node chain (JLNCh) where the neck was staged. The enlarged suspicious nodes were submitted for immediate frozen section. The types of neck dissection were based on the size, shape, number and histological diagnosis of regional nodes. The postoperative additional management was decided according to the results of definitive pathological findings from serial sections of the dissected specimen. Endolaryngeal LASER surgery was carried out in 87 patients based on clinical, histological and imaging criteria. In the course of two years follow-up 2 occult metastases became clinically apparent. At 119 cases open surgery were performed. In 51 patients we could not see enlarged lymph nodes (N< 2 mm) with direct examination, and thus the JLNCh remained intact. In 68 patients elective neck dissections (END) were carried out. In cases of extracapsular spread (ECS) and/or multiple nodal involvements additional radiotherapy was given. C1 [Elo, Janos] Uzsoki University Teaching Hospital, Department of ENT and Head-Neck Surgery, Uzsoki u. 29, H-1145 Budapest, Hungary. [Balatoni, Zsuzsa] Uzsoki University Teaching Hospital, Department of ENT and Head-Neck Surgery, Uzsoki u. 29, H-1145 Budapest, Hungary. [Kotai, Zsuzsa] Uzsoki University Teaching Hospital, Department of ENT and Head-Neck Surgery, Uzsoki u. 29, H-1145 Budapest, Hungary. [Bartfai, Reka] Uzsoki University Teaching Hospital, Department of ENT and Head-Neck Surgery, Uzsoki u. 29, H-1145 Budapest, Hungary. RP Elo, J (reprint author), Uzsoki University Teaching Hospital, Department of ENT and Head-Neck Surgery, H-1145 Budapest, Hungary. CR Ganzer U, Sandrowski P, Vosteen KH, Meyer-Breiting F: Die kombinierte radio-chirurgische Behandlung des fortgeschrittenen Kehlkopfkarzinoms, T3-T4, Laryngol Rhinol 60:63-70, 1981 Snow GB: The N0 neck in head and neck cancer patients. Eur Arch Otorhinolaryngol 250:423-424, 1993 Stringer SP: Current concepts in surgical management of neck metastases from head and neck cancer. Oncology 9:547-553, 1995 Giacomarra V, Tirelli G, Papanikolla L, Bussani R: Predictive factors of nodal metastases in oral cavity and oropharynx carcinomas. Laryngoscope 109:795-799, 1999 Som PM, Curtin HD, Mancuso AA: An imaging-based classification for the cervical nodes designed as an adjunct to recent clinically based nodal classification. Arch Otolaryngol Head Neck Surg 125:388-396, 1999 Lenz M, Kersting-Sommerhof B, Gross MD: Diagnosis and treatment in N0-neck in carcinoma of the upper aerodigestive tract: current status of diagnostic procedures. Eur Arch Otorhinolaryngol 250:432-438, 1993 Steiner W, Hommerich CP: Diagnoses and treatment of the N0 neck of carcinomas of the upper aerodigestive tract. Eur Arch Otorhinolaryngol 250:450-456, 1993. van den Breckel MWM, Stel HV, Castelijns JA, et al: Cervical lymph node metastasis. Assessment of Radiologic Criteria. Radiology 177:379-384, 1990. Friedman M, Mafee MF, Pacella BL: Rationale for elective neck dissection in 1990. Laryngoscope 100:54-59, 1990. Issing PR, Kettling T, Kempt HG, et al. Sonographische Dignitatsbeurteilung von Halslymphknoten unter besonderer Berucksichtigung der Farbdopplersonographie. Laryngo- Rhino-Otol 78:566-572, 1999. Glanz HK: Carcinoma of the larynx. Adv Otorhinolaryngol 32:1-23, 1984 Ambrosch P, Brinck U: Detection of nodal micrometastases in head and neck cancers by serial sectioning and immunostaining. Oncology 10:1221-1226, 1996 Busaba NY, Fabian RL: Extent of Lymphadenectomy achived by various modification of neck dissection: A pathologic analysis. Laryngoscope 109: 212-215, 1999. Czigner J, Savay L: Endoscopic CO2- laser surgery for vocal cord cancer. Diagnostic and therapeutic Endoscopy 1:69-74, 1994. Elo J: Relationship between “dynamic histology” and metastases of laryngeal tumors. Acta Otolaryngol 112:383-386, 1992. Robbins KT:Classification of neck dissection: current concepts and future consideration. Oto-laryngol Clin North Am 31:639- 656, 1998 Werner JA, Dunne AA, Brandt D, et al: Untersuchungen zum Stellenwert der Sentinel Lymphonodektomie bei Karzinom des Pharynx und Larynx Laryngo-Rhino-Otol 78: 663-670, 1999. Hagen R: Die lymphatische Drainage des Larynx – Einfluß auf die Therapieentscheidung beim Larynx-Karcinom. HNO 44:55-60,1996. Mann W: Das Lymphsystem des Kehlkopfes. Arch Otorhinolaryngol 225:165-180, 1979. Werner JA, Schuncke M, Lippert BM, et al: Das laryngeale Lymphghefaasystem des Menschen. Eine morphologische und lymphographische Untersuchung unter klinischen Gesichtspuncten. HNO 43:525-531, 1995. O’Brien ChJ, Traynor SJ, McNeil ÍE, et al: The use of clinical criteria alone in the management of clinically negative neck among patients with squamous cell carcinoma of the oral cavity and oropharynx. Ach Otolaryngol Head Neck Surg 126:360-365, 2000. Elo J: Die Bedeutung des Malignitatsgrades und Proliferationsaktivitat des Larynxcarcinomas fur regionare Lymphknotenmetastasen. Laryngol Rhinol Otol 62:244-245,1983. Schantz SP: Biologic markers, cellular differentation and metastatic head and neck cancer. Eur Arch Otorhinolaryngol 250:424-428, 1993. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 257 EP 261 PG 5 ER PT J AU Canoz, Belenli, O Patiroglu, ET AF Canoz, Ozlem Belenli, Olcay Patiroglu, E Tahir TI General Features of Gastric Carcinomas and Comparison of HSP70 and NK cell Immunoreactivity with Prognostic Factors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gastric carcinoma; HSP70; NK-cell; prognostic factors ID gastric carcinoma; HSP70; NK-cell; prognostic factors AB During the period of 1996-1998 ninety-four gastrectomy specimens with gastric carcinoma referred to Erciyes University, Medical Faculty, Department of Pathology, were examined histopathologically, histochemically and immunohistochemically. General characteristics of gastric carcinomas and prognostic factors were studied. According the Lauren classification, of the 94 cases of gastric carcinomas, 56 were intestinal type, 21 were diffuse type and 17 were mixed type carcinoma. The association rates of Helicobacter pylori, chronic atrophic gastritis and intestinal metaplasia with gastric carcinomas were high. There was strong immunorectivity with HSP70 in 62,5% of the intestinal type carcinomas. This ratios were lower in diffuse and mixed type carcinomas (p<0.05). The more tumor size and invasion depth increased, the more HSP70 immunoreactivity was obtained (p<0.05). HSP70 immunorectivity was considerably higher in the patients having lymph node metastasis and vascular invasion (p<0.05). It was found that the NK cell number was low in the tumor but higher around the tumor in early gastric carcinomas, compared with advanced carcinomas (p>0.05). In the tumors larger than 10 cm with vascular invasion, NK cell number was lower around the tumor (p>0.05). Defining prognostic factors of gastric carcinomas is of importance to clinicians. It is thought that HSP70 immunoreactivity, besides invasion depth, lymph node metastasis, vascular invasion, tumor size and inflammatory reaction against the tumor, is important in prognosis and associated with advanced stage. C1 [Canoz, Ozlem] Medical Faculty of Erciyes University, Pathology Department, MKP Bulvari, Zumrut mah, Hukukcular Sit. B-Blok No:15Kocasinan / Kayseri, Turkey. [Belenli, Olcay] Medical Faculty of Duzce Abant Izzet Baysal University, Pathology DepartmentDuzce, Turkey. [Patiroglu, E Tahir] Medical Faculty of Erciyes University, Pathology Department, MKP Bulvari, Zumrut mah, Hukukcular Sit. B-Blok No:15Kocasinan / Kayseri, Turkey. RP Canoz, (reprint author), Medical Faculty of Erciyes University, Pathology Department, Kocasinan / Kayseri, Turkey. EM ocanoz@erciyes.edu.tr CR Davessar K, Pezzullo JC, Kessimian N, et al: Gastric adenocarcinoma: prognostic significance of several pathologic parameters and histologic classifications. Hum Pathol 21:325-332, 1990 Grabiec J, Owen DA: Carcinoma of the stomach in young persons. Cancer 56:388-390, 1985 Lauren PA, Nevalainen TJ: Epidemiology of intestinal and diffuse types of gastric carcinoma. Cancer 71:2926-2933, 1993 Lundergardh G, Lindgren A, Rohul A, et al: Intestinal and diffuse types of gastric cancer: secular trends in Sweden since 1951. Br J Cancer 64:1182-1186, 1991 Antonioli DA, Goldman H. Changes in the location and type of gastric adenocarcinoma Cancer 50:775-781, 1982 Zanghieri G, Di Gregorio C, Sacchetti C, et al. Familial occurence of gastric cancer in the 2-year experience of a population- based registry. Cancer 66:2047-2051, 1990 Lu XP, Omar RA, Chang WWL. Immunocytochemical expression of the 70 kD heat-shock protein in ischaemic bowel disease. J Pathol 179:409-413, 1996 Crawford JM. The Gastrointestinal Tract. In: Cotran R, Kumar V, Robbins S, eds), Pathologic Basis of Disease, 5th ed). WB Saunders Company, Philadelphia 1994, pp 21, 779-783. Robert J, Menoret A, Basu S, et al: Phylogenetic conservation of the molecular and immunological properties of the chaperones gp96 and hsp70. Eur J Immunol 31:186-195, 2001 Monson JRT, Ramsden CW, Giles GR, Brennan TG. Lymphokin activated killer, LAK, cells in patients with gastrointestinal cancer. Gut 28:1420-1425, 1987 Coca S, Perez-Piqueras J, Martinez D, et al. The prognostic significance of intratumoral natural killer cells in patients with colorectal carcinoma. Cancer 79:2320-2328, 1997 Lauren P: The two histological main types of gastric carcinoma: diffuse and so called intestinal-type carcinoma. Acta Pathol Microbiol lmmunol Scand [B] 64:31-49, 1965 Holmes EC: Immunology of tumor infiltrating lymphocytes. Ann Surg 201:158-163, 1985 Nakamura K, Ueyama T, Yao T, et al: Pathology and prognosis of gastric carcinoma. Cancer 70:1030-1037, 1992 Setala LP, Kosma VM, Marin S, et al: Prognostic factors in gastric cancer: the value of vascular invasion, mitotic rate and lymphoplasmocytic infiltration. Br J Cancer 74:766-772, 1996 Rotterdam H: Stomach. In: Silverberg SG, ed), Biopsy Diagnosis of the Digestive Tract, 2nd ed). Raven Press, New York pp 188-214, 1993 Sipponen P, Kekki M, Siurala M: Atrophic chronic gastritis and intestinal metaplasia in gastric carcinoma. Cancer 52:1062-1068, 1983 Iida F, Kusama J: Gastric carcinoma and intestinal metaplasia. Cancer 50:2854-2858, 1982 Huang CB, Xu J, Huang JF, Meng XY: Sulphomucin colonic type intestinal metaplasia and carcinoma in the stomach. Cancer 57:1370-1375, 1986 Hattori T: Devolepment of adenocarcinomas in the stomachs. Cancer 57:1528-1534, 1986 Oohara T, Tohma H, Aono G, et al: Intestinal metaplasia of the regenerative epithelia in 549 gastric ulcers. Hum Pathol 14:1066-1071, 1983 Filipe MI, Potet F, Bogomoletz WV, et al. Incomplete sulphomucin secreting intestinal metaplasia for gastric cancer. Gut 6:1319-1326, 1985 Tanaka F, Yamaguchi K, Urano Y: Statistical analysis of the surgical materials of stomach. Cancer 59:1978-1982, 1987 Murayama H, Kikuchi M, Enjoji M, et al: Changes in gastric mucosa that antedate gastric carcinoma. Cancer 66:2017-2026, 1990 Karnes WE, Samloff IM, Sivrala M, Kekki M: Positive antibody and negative tissue staining for Helicobacter pylori in subjects with atrophic body gastritis. Gastroenterology 101:167-174, 1991 Go MF: What are the host factors that place an individual at risk for Helicobacter pylori-associated disease? Gastroenterology 113: 15-20, 1997 Shibata T, Imoto I, Ohuchi Y, et al: Helicobacter pylori infection in patients with gastric carcinoma in biopsy and surgical resection specimens. Cancer 77:1044-1049, 1996 Lee PM, Jang JJ, Kim JS, et al: Association of Helicobacter pylori infection with gastric adenocarcinoma. Jpn J Cancer Res 89:597-603, 1998 Wee A, Kang JY, Teh M: Helicobacter pylori and gastric cancer: correlation with gastritis, intestinal metaplasia, and tumour histology. Gut 33:1029-1032, 1992 Asaka M, Takeda H, Sugiyama T, Kato M: What role does Helicobacter pylori play in gastric cancer? Gastroenterology 113:56-60, 1997 Multhoff G, Botzler C, Jennen L, et al. Heat-shock protein 72 on tumor cells. J Immunol 158:4341-4350, 1997 Kawanishi K, Shiozaki H, Doki Y, et al: Prognostic significance of heat shock proteins 27 and 70 in patients with squamous cell carcinoma of the esophagus. Cancer 85: 1649-1657, 1999 Park CS, Joo IS, Song SY, et al: An immunohistochemical analysis of heat shock protein 70, p 53 and estrogen status in carcinoma of the uterine cervix. Gynecol Oncol 74: 53-60, 1999 Ciocca DR, Clark GM, Tandon AK, et al: Heat shock protein hsp70 in patient with axillary lymph node-negative breast cancer: prognostic implications. J Nat Cancer Ins 85:570-574, 1996 Chin Y, Janseens J, Vandepitte J, et al. Phenotypic analysis of tumor-infiltrating lymphocytes from human breast cancer. Anticancer Res 12:1463-1466, 1992 Brittenden J, Heys SD, Ross J, Eremin O: Natural killer cells and cancer. Cancer 77:1226-1243, 1996 Toliohu Th, Stravoravdi P, Polyzonis M, Vakalikos J: Natural killer cell activation after interferon administration in patients with metastatic renal cell carcinoma. Eur Urol 29:252-256, 1996 Karimine N, Arinaga S, Inove H, et al: Lymphokine-activated killer cell function of peripheral blood mononuclear cells, spleen cells and regional lymph node cells in gastric cancer patients. Clin Exp Immunol 96:484-490, 1994 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 262 EP 269 PG 8 ER PT J AU Romics, I Riesz, P Szelepcsenyi, J Nyirady, P AF Romics, Imre Riesz, Peter Szelepcsenyi, Janos Nyirady, Peter TI Bilateral Renal Cell Carcinoma in a Horseshoe Kidney SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE renal cell carcinoma; horseshoe kidney; radical nephrectomy ID renal cell carcinoma; horseshoe kidney; radical nephrectomy AB We report a case of bilateral renal cell carcinoma in a horseshoe kidney. To the best of our knowledge this is the second reported case in the international literature. We performed different radiological examinations preoperatively to identify of blood supply, because correct preoperative location of vessels is mandatory. C1 [Romics, Imre] Semmelweis University, Department of Urology, Ulloi ut 78/b, H-1082 Budapest, Hungary. [Riesz, Peter] Semmelweis University, Department of Urology, Ulloi ut 78/b, H-1082 Budapest, Hungary. [Szelepcsenyi, Janos] General PractitionerNagykata, Hungary. [Nyirady, Peter] Semmelweis University, Department of Urology, Ulloi ut 78/b, H-1082 Budapest, Hungary. RP Romics, I (reprint author), Semmelweis University, Department of Urology, H-1082 Budapest, Hungary. EM romimre@urol.sote.hu CR Bauer S, Perlmutter A, Retik A: Anomalies of the upper urinary tract. In: Campbell’s Urology, 6th ed., Eds: Walsh PC, Retik AB, Stamey TA, Vaughan ED Jr), W.B. Saunders Philadelphia, pp 1357-1442, 1992 Brum FA, Becker M, Uglione A, Da Ros CT: Polycystic horseshoe kidney. J Urol 158: 2229-2232, 1997 Cuckow PM, Nyirady P: Embryology of the Urogenital Tract. In: Pediatric Urology., Eds: Gearhart JP, Rink RC, Mouriquand PDE), W.B. Saunders, Philadelphia, pp. 3-13, 2001 Fisher RM, Frank JD: Abnormal Migration and Fusion of the Kidneys. In: Pediatric Urology., Eds: Gearhart JP, Rink RC, Mouriquand PDE), W.B. Saunders, Philadelphia, pp. 275-278, 2001 Schubert RA, Soldner J, Steiner T, et al: WA: Bilateral renal cell carcinoma in a horseshoe kidney: preoperative assessment with MRI and digital substraction angiography. Eur Radiol 8:1694- 1697, 1998 Smith RB, deKernion JB, Ehrlich RM, et al: Bilateral renal cell carcinoma and renal cell carcinoma in the solitary kidney. J Urol 132: 450-455, 1984 Thomas DFM, Cook JA: Perinatal Urology. In Textbook of Genitourinary Surgery,2nd ed., Eds: Whitfield HN, Hendry WF, Kirby RS, Duckett JW), Blackwell Science, Oxford, pp. 133- 143, 1998 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 270 EP 271 PG 2 ER PT J AU Arista-Nasr, J Nuncio, J Martinez, B AF Arista-Nasr, Julian Nuncio, Juan Martinez, Braulio TI Atypical Histiocytic Infiltration Simulating Diffuse-type Carcinoma in a Gastric Ulcer due to Non-Steroidal Anti-Inflammatory Drugs SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE atypical; histiocytic; gastric; ulcer; NSAID ID atypical; histiocytic; gastric; ulcer; NSAID AB A 83-year old man treated with naproxen during two years was admitted because of hypovolemia and peritoneal irritation. A panendoscopic study was performed and an ulcer localized at the large curvature of the stomach was disclosed. In the gastrectomy specimen the ulcer showed necrosis, edema, fibrosis, chronic inflammatory infiltrate with lymphocytes and plasma cells. Additionally, atypical cells with irregular and hyperchromatic nuclei or vacuolated cytoplasm were seen in the lamina propia and infiltrating the muscular layers; isolated signet-ring-like cells were also seen. Histochemical study with periodic acid-Schiff, mucicarmin, and colloidal stains revealed mucosubstances in these cells. A poorly differentiated carcinoma was initially diagnosed. However, the immunohistochemical study were positive for histiocytic markers (CD-68, S-100 protein) and negative for epithelial markers (cytokeratin; and epithelial membrane antigen). The positivity of mucus stains in the histiocytes could be explained in this case by phagocytosis of mucous substances released from broken hyperplastic glands in the vicinity of the ulcer. To our knowledge, atypical histiocytic infiltration in gastric ulcers has not been previously described; thus, it should be included in the group of gastric carcinoma mimicks. C1 [Arista-Nasr, Julian] Instituto Nacional de la Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No.15, Tlalpan, 14000 Mexico City, Mexico. [Nuncio, Juan] Instituto Nacional de la Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No.15, Tlalpan, 14000 Mexico City, Mexico. [Martinez, Braulio] Instituto Nacional de la Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No.15, Tlalpan, 14000 Mexico City, Mexico. RP Arista-Nasr, J (reprint author), Instituto Nacional de la Nutricion Salvador Zubiran, Department of Pathology, 14000 Mexico City, Mexico. EM pipa5@hotmail.com CR Humphries TJ. Gastric xantomata: The need for recognition. J Clin Gastroenterol 4:275-276, 1982. Drude RB, Balart LA, Herrington JP, et al: Gastric xantoma: histologic similarity to signet ring cell carcinoma. J Clin Gastroenterol 4:217-221, 1982. Arista-Nasr J, Jimenez A, Keirns C, et al: The role of endoscopic biopsy in the diagnosis of gastric lymphoma. Hum Pathol 22:339-348, 1991. Zamboni G, Franzin G, Scarpa A, et al: Carcinoma-like signetring cells in gastric mucosa-associated lymphoid tissue, MALT, lymphoma. Am J Surg Pathol 20:588-598, 1996. Arista-Nasr J, Romero-Lagarza P, Pichardo R. Artifactual signet-ring-like cells in endoscopic biopsy of gastric lymphoma. Arch Pathol Lab Med 121:623-625, 1997. Taha AS, Nakshabendi I, Lee FD et al. Chemical gastritis and Helicobacter pylori related gastritis in patients receiving nonsteroidal anti-inflammatory drugs: comparison and correlation with peptic ulceration. J Clin Pathol 45:135-139, 1992. Lanza FL, Royer GL, Nelson RS: Endoscopic evaluation of the effect of aspirin, buffered aspirin and enteric-coated aspirin on gastric and duodenal mucosa. N Engl J Med 303:136-138, 1980. Dixon MF, O´Connor HJ, Axon ATR et al: Reflux gastritis: distinct histopathological entity?. J Clin Pathol 39:524-530, 1986. Quinn M, Bjarnason I, Price AB: Gastritis in patients on nonsteroidal anti-inflammatory drugs. Histopathology 23:341-348, 1993. El-Zimaity HMT, Genta RM, Graham DY: Histological features do not define NSAID-induced gastritis. Hum Pathol 27:1348- 1354, 1996 Ball PAJ, James AH: The histological background to gastric ulcer. Lancet 1:1365-1367, 1961 Ogawa Y, Inada K, Tsutsumi Y: CD68 reactive histiocytosis complicating early gastric cancer. Pathol Int. 45:698-701, 1995. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 272 EP 274 PG 3 ER PT J AU Kayaselcuk, F Tuncer, I Toyganozu, Y Bal, N Gursel, AF Kayaselcuk, Fazilet Tuncer, Ilhan Toyganozu, Yavuz Bal, Nebil Gursel, Ozgur TI Carcinosarcoma of the Stomach SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE carcinosarcoma; immunohistochemistry; stomach ID carcinosarcoma; immunohistochemistry; stomach AB In the gastrointestinal tract, carcinosarcomas are most frequently seen in the esophagus. Carcinosarcoma in the stomach is a rare tumor. We report a carcinosarcoma of the antrum of stomach. The tumor was polypoid and exophytic in appearance and located in the antrum. Immunohistochemical studies showed positivity for cytokeratin, epithelial membrane antigen and cytoplasmic carcinoembryonic antigen in the epithelial component. Positive staining with vimentin, desmin and focal smooth muscle actin and negative staining with chromogranin were observed in spindle cells. Nuclear positive staining was observed with p53 and Ki-67 in both glandular and spindle atypical cells. C1 [Kayaselcuk, Fazilet] Baskent University Faculty of Medicine, Department of Pathology, Adana Hospital, Seyhan Hospital, Dadaloglu mah. 39 Sk. No 6, 01250 Yuregir, Adana, Turkey. [Tuncer, Ilhan] Baskent University Faculty of Medicine, Department of Pathology, Adana Hospital, Seyhan Hospital, Dadaloglu mah. 39 Sk. No 6, 01250 Yuregir, Adana, Turkey. [Toyganozu, Yavuz] Department of SurgeryAdana, Turkey. [Bal, Nebil] Baskent University Faculty of Medicine, Department of Pathology, Adana Hospital, Seyhan Hospital, Dadaloglu mah. 39 Sk. No 6, 01250 Yuregir, Adana, Turkey. [Gursel, Ozgur] Department of GastroenterologyAdana, Turkey. RP Kayaselcuk, F (reprint author), Baskent University Faculty of Medicine, Department of Pathology, Adana Hospital, Seyhan Hospital, 01250 Yuregir, Adana, Turkey. EM faziletks@yahoo.com CR Robey-Cafferty SS, Gringon DJ, Ro JY, et al: Sarcomatoid carcinoma of the stomach. A report of three cases with immunohistochemical and ultrastructural observations. Cancer 65: 1601- 1606, 1990. Bansal M, Kaneko M, Gordon RE: Carcinosarcoma and separate carcinoid tumor of the stomach. A case report with light and electron microscopic studies. Cancer 50: 1876-1881, 1982. Fenoglio-Preiser CM, Noffsinger AE, Stemmermann GN, et al: Gastointestinal Pathology. Second ed, 1999; Philadelphia, pp: 262. Xu LT, Sun CF, Wu LH, et al: Clinical and pathological characteristics of carcinosarcoma of the esophagus: report of four cases. Ann Thorac Surg 37: 197-203, 1984. Tsuneyama K, Sasaki M, Sabit A, et al: A case report of gastric carcinosarcoma with rhabdomyosarcomatous and neuroendocrinal differentiation. Pathol Res Pract 195: 93-97, 1999. Nakayama Y, Murayama H, Iwasaki H, et al: Gastric carcinosarcoma, sarcomatoid carcinoma, with rhabdomyoblastic and osteoblastic differentiation. Pathol Int 47: 557-563, 1997. Matsukuma S, Wada R, Hase K, Sakai Y, Ogata S, Kuwabara N: Gastric stump carcinosarcoma with rhabdomyosarcomatous differentiation. Pathol Int 47: 73-77, 1997. Melato M, Bucconi S, Grillo BP, et al: Carcinosarcoma and separate neuroendocrine malignant tumor of a malignancy promoter, the gastric stump. Anticancer Res 13: 2485-2488, 1993. Tanimura H, Furuta M: Carcinosarcoma of the stomach. Am J Surg 113: 702-709, 1967. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 275 EP 277 PG 3 ER PT J AU Bogner, B Hegedus, G AF Bogner, Barna Hegedus, Geza TI Ciliated Hepatic Foregut Cyst SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE liver; benign hepatic cysts; ciliated hepatic foregut cyst ID liver; benign hepatic cysts; ciliated hepatic foregut cyst AB Ciliated hepatic foregut cyst is a rare, benign, most often solitary and unilocular, rarely multilocular cyst made up of a ciliated pseudostratified columnar epithelium, a subepitheial connective tissue layer, a smooth muscle layer and an outer fibrous capsule. The lesion is usually found incidentally by ultrasonography, during surgical exploration or autopsy. Recent publications characterizes of its fine needle aspiration biopsy features. The lesion is mostly asymptomatic, however one case caused portal vein compression and another which showed malignant trasformation through squamous metaplasia which warns to examine these lesions cautiously. As the lesion is extremely rare it is difficult to estimate its prevalence and its nature, so every single case presentation could be important. C1 [Bogner, Barna] County Hospital of Baranya, Department of Pathology, Rakoczi ut 2, 7623 Pecs, Hungary. [Hegedus, Geza] County Hospital of Baranya, Department of Pathology, Rakoczi ut 2, 7623 Pecs, Hungary. RP Bogner, B (reprint author), County Hospital of Baranya, Department of Pathology, 7623 Pecs, Hungary. EM bognerb@pathodg.hu CR Friedrich N: Cyste mit flimmerepithel in der Leber. Archiv Pathol Anat 11: 466-469, 1857. Hornstein A, Batts KP, Linz LJ et al:Fine needle aspiration diagnosis of ciliated hepatic foregut cysts: a report of three cases. Acta Cytol 40: 576-580, 1996. Kimura A, Makuuchi M, Takayashu K, et al: Ciliated hepatic foregut cyst with soild tumor appearance on CT. J Comput Assist Tomogr 14: 1016-1018, 1990. Kadoya M, Matsui O, Nakamuna Y et al: Ciliated hepatic foregut cyst: radiologic features. Radiology 175: 475-477, 1990. Vick DJ, Goodman ZD, Deavers MT et al.: Ciliated hepatic foergut cyst. Am J Surg Pathol 23: 671-677, 1999. Vick DJ, Goodman ZD, Ishak KG: Squamous cell carcinoma arising in a ciliated hepatic foregut cyst. Arch Pathol Lab Med 11: 1115-1117, 1999. Wheeler DA, Edmondson HA.: Ciliated hepatic foregut cyst. Am J Surg Pathol 8: 467-70, 1984. Zaman SS, Langer JE, Gupta PK: Report of a case with findings on fine needle aspiration. Acta Cytol 39: 781-78 1995. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 278 EP 279 PG 2 ER PT J AU Culhaci, N Levi, E Sen, S Kacar, F Meteoglu, I AF Culhaci, Nil Levi, Edi Sen, Serdar Kacar, Furuzan Meteoglu, Ibrahim TI Pulmonary Lymphomatoid Granulomatosis Evolving to Large Cell Lymphoma in the Skin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE lymphomatoid granulomatosis; Epstein-Barr virus; large cell lymphoma; angiocentric lymphoma ID lymphomatoid granulomatosis; Epstein-Barr virus; large cell lymphoma; angiocentric lymphoma AB Lymphomatoid granulomatosis is an angiodestructive, angioinvasive lymphoproliferative disorder. It involves most frequently lungs, central nervous system and skin. Recent studies indicate that lymphomatoid granulomatosis is an Epstein-Barr virus associated B cell disorder with a background of reactive T lymphocytes. In a 49 year old woman presenting with fever, malaise and pulmonary masses the diagnosis of lymphomatoid granulomatosis was established histologically by open lung biopsy. Following the initial diagnosis the patient was found to have gastric and skin involvement. The skin lesion was diagnosed as diffuse large B-cell lymphoma. C1 [Culhaci, Nil] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Levi, Edi] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Sen, Serdar] Adnan Menderes University, Faculty of Medicine, Department of Thoracic SurgeryAydin, Turkey. [Kacar, Furuzan] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Meteoglu, Ibrahim] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. RP Culhaci, N (reprint author), Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. EM nculhaci@hotmail.com CR Patchefsky AS: Nonneoplastic pulmonary disease. In: Sternberg SS, ed). Diagnostic surgical pathology, third ed. Philadelphia: Lippincott Williams&Wilkins pp 1035-1037, 1999 Jaffe ES, Wilson WH: Lymphomatoid granulomatosis: pathogenesis, pathology and clinical implications. Cancer Surv 30: 233-247, 1997 Wilson WH, Kingma D, Raffeld M, et al: Association of lymphomatoid granulomatosis with Epstein-Barr viral infection of B lymphocytes and response to interferon-2b. Blood 87: 4531- 4537, 1996 Kingma D, Fishback N, Wallberg K, et al: Pulmonary lymphomatoid granulomatosis. Evidence for a proliferation of Epstein-Barr virus infected B-lymphocytes with a prominent Tcell component and vasculitis. Am J Surg Pathol 18: 753-764, 1994 Karnak I, Ciftci AO, Talim B, et al: Pulmonary lymphomatoid granulomatosis in a 4 year old. J Pediatr Surg 34: 1033-1035, 1999 Lipford EH, Margolick JB Jr, Longo DL, et al: Angiocentric immunoproliferative lesions: a clinicopathologic spectrum of post-thymic T-cell proliferations. Blood 72: 1674-1681, 1988 Guinee DG, Perkins SL, Travis WD, et al: Proliferation and cellular phenotype in lymphomatoid granulomatosis. Implications of a higher proliferation index in B cells. Am J Surg Pathol 22: 1093-1100, 1998 Taniere Ph, Thivolet-Bejui F, Vitrey D, et al: Lymphomatoid granulomatosis- a report on four cases: evidence for B phenotype of the tumoral cells. Eur Respir J 12: 102-106, 1998 Beaty MW, Toro J, Sorbora L, et al: Cutaneous lymphomatoid granulomatosis: correlation of clinical and biologic features. Am J Surg Pathol 25: 1111-1121, 2001 Saxena A, Dyker KM, Angel S, et al: Posttransplant diffuse large B-cell lymphoma of "Lymphomatoid Granulomatosis" type. Virch Arch 441:622-628, 2002 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 280 EP 282 PG 3 ER PT J AU Dahiya, S Kumar, R Sarkar, Ch Ralte, MA Sharma, ChM AF Dahiya, Sonika Kumar, Rajiv Sarkar, Chitra Ralte, Mercy Angela Sharma, Chand Mehar TI Clear Cell Odontogenic Carcinoma: a Diagnostic Dilemma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE clear cell odontogenic tumors; clear cell ameloblastoma; maxilla; young age ID clear cell odontogenic tumors; clear cell ameloblastoma; maxilla; young age AB A 26-year-old man presented with a swelling in the right side of face and CT scan revealed a destructive tumor in the right maxilla. Tumor recurred within 5 years of its excision and histopathological examination revealed a clear cell odontogenic carcinoma. The rarity of this tumor, occurrence in maxilla and young age of the patient are some of the rare features which need documentation. The importance of its diagnosis and various differential diagnoses are discussed. C1 [Dahiya, Sonika] All India Institute of Medical Sciences, Departments of Pathology and E.N.T., 110029 New Delhi, India. [Kumar, Rajiv] All India Institute of Medical Sciences, Departments of Pathology and E.N.T., 110029 New Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Departments of Pathology and E.N.T., 110029 New Delhi, India. [Ralte, Mercy Angela] All India Institute of Medical Sciences, Departments of Pathology and E.N.T., 110029 New Delhi, India. [Sharma, Chand Mehar] All India Institute of Medical Sciences, Departments of Pathology and E.N.T., 110029 New Delhi, India. RP Sharma, ChM (reprint author), All India Institute of Medical Sciences, Departments of Pathology and E.N.T., 110029 New Delhi, India. EM meharsharma@hotmail.com CR Kramer IRH, Pindborg JJ, Shear M: WHO histological typing of odontogenic tumors. 2nd Ed. Berlin, Germany; Springer Verlag; 1992 Hansen LS, Eversole LR, Green TL, Powell HS: Clear cell odontogenic tumor- A new histological variant with aggressive potential. Head Neck Surg 8: 115-123, 1985 Waldron CA, Small IA, Silverman H: Clear cell ameloblastoma: An odontogenic carcinoma. J Oral Maxillofac Surg 43: 707- 717, 1985 Mari A, Escutia E, Carrera M, Pericot J: Clear cell ameloblastoma or odontogenic carcinoma. J Cranio Maxillofacial Surg 23: 387-390, 1995 Muramatsu T, Hashimoto S, Inoue T, et al: Clear cell odontogenic carcinoma in the mandible: histochemical and immuno histochemical observations with a review of the literature. J Oral Pathol Med 25: 516-521, 1996 Li TJ, Yu SF, Gao Y, Wang E-B: Clear cell odontogenic carcinoma. A clinicopathological and immunohistochemical study of 5 cases. Arch Pathol Lab Med 125: 1566-1571, 2001 Bang G, Koppang HS, Hansen LS et al: Clear cell odontogenic carcinoma : Report of three cases with pulmonary and lymphnode metastases. J Oral Pathol Med18: 113-118, 1989 Eversole LR, Duffey DC, Powell NB. Clear cell odontogenic carcinoma: A clinicopathological analysis. Arch Otololaryngol Head Neck Surg121: 685-689, 1995. Fan J, Kubota E, Imamura H, et al:Clear cell odontogenic carcinoma: A case report with massive invasion of neighbouring organs and lymph node metastasis. Oral Surg Oral Med Oral Pathol 74: 768-775, 1992. Milles M, Doyle JL, Mesa M, Raz S:Clear cell odontogenic carcinoma with lymph node metastasis. Oral Surg Oral Med Oral Pathol 76: 82-89, 1993. Piatteli A, Sesenna E, Trisi P: Clear cell odontogenic carcinoma: Report of a case with lymph node and pulmonary metastases. Eur J Canc Oral Oncol 30B: 278-280, 1994. Eversole LR: Malignant epithelial odontogenic tumors. Sem Diag Pathol 16: 317-324, 1999 Muller H, Salootweg P: Clear cell differentiation is an ameloblastoma. J Maxillofac Surg 14: 158-160, 1986 Ng KH, Siar CH: Peripheral ameloblastoma with clear cell differentiation. Oral Surg Oral Med Oral Pathol 70: 210-213, 1999 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2002 VL 8 IS 4 BP 283 EP 285 PG 3 ER PT J AU Lengyel, E Gilde, K Remenar, Esik, O AF Lengyel, Erzsebet Gilde, Katalin Remenar, Eva Esik, Olga TI Malignant Mucosal Melanoma of the Head and Neck - a Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE head and neck; mucosal melanoma; malignant melanoma; surgery radiotherapy; chemotherapy ID head and neck; mucosal melanoma; malignant melanoma; surgery radiotherapy; chemotherapy AB Mucosal melanomas comprise about 1% of all malignant melanomas and exhibit far more aggressive behaviour than that of skin melanomas: they are more inclined to metastatize into regional and distant sites or recur locally, regionally or in distant locations, resulting in a high rate of cause-specific death. Mucosal melanomas in the head and neck region account for half of all mucosal melanomas, occurring mainly in the upper respiratory tract, oral cavity and pharynx. They appear with equal gender distribution and with a peak incidence in the age range 60-80 years. In consequence of their hidden location, they are usually diagnosed in a locoregionally advanced clinical stage, with a rate of 5-48% of regional and 4-14% of distant dissemination. The typical therapeutic approach is surgery, postoperative irradiation and systemic therapy. Local control with either surgery or radiotherapy is frequently (60- 70%) achieved, but the rates of local, regional and distant recurrences are high (50-90%, 20-60% and 30-70%, respectively). The reported 5-year actual survival rates are poor (17-48%), which is attributed mainly to a haematogenous dissemination. These characteristics demonstrate that identification of the precursor lesions and more effective local and systemic approaches are needed to improve the therapeutic results. C1 [Lengyel, Erzsebet] National Institute of Oncology, Center of Radiotherapy, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Gilde, Katalin] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Remenar, Eva] National Institute of Oncology, Department of Head and Neck SurgeryBudapest, Hungary. [Esik, Olga] National Institute of Oncology, Center of Radiotherapy, Rath Gy. u. 7-9, 1122 Budapest, Hungary. RP Lengyel, E (reprint author), National Institute of Oncology, Center of Radiotherapy, 1122 Budapest, Hungary. EM homokora2001@yahoo.com CR Agarwala SS, Atkins MB, Kirkwood JM: Current approaches to advanced and high-risk melanoma. Proc Am Soc Clin Oncol 2000. Ang KK, Byers RM, Peters LJ, et al: Regional radiotherapy as adjuvant treatment for head and neck malignant melanoma. Arch Otolaryngol Head Neck Surg 116:169-172, 1990. Banfalvi T, Boldizsar M, Gergye M, et al: Comparison of prognostic significance of serum 5-S-Cysteinyldopa, LDH and S- 100B protein in Stage III-IV malignant melanoma. Pathol Oncol Res 8:183-187, 2002. Barranco SC, Romsdahl MM, Humphrey RM: The radiation response of human malignant melanoma cells grown in vitro. Cancer Res 31:830-831, 1971. Barton RT: Mucosal melanomas of the head and neck. Laryngoscope 85:93-99, 1975. Batsakis JD, Regezi JA, Solomon AR, et al: The pathology of head and neck tumors-part 13: mucosal melanomas. Head Neck Surg 4:404-412, 1982. Berking C, Schlupen E, Schrader A, et al: Tumor markers in peripheral blood of patients with malignant melanoma: Multimarker RT-PCR versus a luminometric assay for S-100. Arch Dermatol Res 291:479-484, 1999. Blatchford SJ, Koopman CF, Coulthard SW: Mucosal melanoma of the head and neck. Laryngoscope 96:929- 934,1986. Brandwein MS, Rothstein A, Lawson W, et al: A clinicopathologic study of 25 cases and literature meta-analysis. Arch Otolaryngol Head Neck Surg 123:290-296, 1997. Britten CD, Rowinsky EK, Baker SD, et al: A phase I and pharmacokinetic study of temozolomide and cisplatin in patients with advanced solid malignancies. Clin Cancer Res 5:1629, 1999. Chang AE, Karnell LH, Menck HR: The National Cancer Data Base Report on cutaneous and noncutaneous melanoma. Cancer 83:1664-1678, 1998. Chapman PB, Einhorn LH, Meyers ML, et al: Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol 17:2745, 1999. Chaundhry AP, Hampel A, Gorlin RJ: Primary melanoma of the oral cavity. Cancer 11:923-928, 1958. Cocconi G, Bella M, Calabresi F, et al: Treatment of metastatic malignant melanoma with dacarbazine plus tamoxifen. N Engl J Med 327:516, 1992. Eneroth CM, Lungberg C: Mucosal malignant melanomas of the head and neck. Acta Otolaryngol 80:452-458, 1975. Falkson CI, Falkson G, Falkson HC:Improved results with the addition of interferon alfa-2b to dacarbazine in the treatment of patients with metastatic malignant melanoma. J Clin Oncol 9:1403, 1991. Falkson CI, Ibrahim J, Kirkwood JM, et al: Phase III trial of dacarbazine versus dacarbazine with interferon -2b versus dacarbazine with interferon -2b and tamoxifen in patients with metastatic malignant melanoma: an Eastern Cooperative Oncology Group study, E3690). J Clin Oncol 16:1743, 1998. Freedman HM, DeSanto LW, Devine KD, et al: Malignant melanoma of the nasal cavity and paranasal sinuses. Arch Otolaryngol 97: 322-325, 1973. Gilligan D, Slevin NJ: Radical radiotherapy for 28 cases of mucosal melanoma in the nasal cavity and sinuses. Br J Radiol 64:1147-1150, 1991. Greco FA, Hainsworth JD: Cancer of unknown primery site. In: Cancer: principles and practice of oncology, Eds.: deVita VT Jr, Hellman S, Rosenberg SA, 6th ed, Lippincott Williams and Wilkins, Philadelphia–Baltimore-New York-London-Buenos Aires-Hong Kong–Sydney-Tokyo, 2001, pp. 2537-2569. Goldman JA, Lawson W, Zak FG, et al: The presence of melanocytes in human larynx. Laryngoscope 82:824-835, 1972 Gotshalk HC, Tessmer CF, Smith JW: Malignant melanoma of palate. Arch Pathol 30:762, 1940. Guzzo M, Grandi C, Licitra L, et al: Mucosal malignant melanoma of head and neck: forty-eight cases treated at Instituto Nazionale Tumori of Milan. Eur J Surg Oncol 19:316-319,1993. Hall EJ. Radiobiology for the radiologists. 5th ed. Philadelphia: Lippincott-Williams and Wilkins, 2000. Harrison DFN, Lund VJ: Tumors of the upper jaw. Edinburgh, London: Churchill Livingstone, 1993, pp. 332. Keilholz U, Eggermont AM: The role of interleukin-2 in the management of stage IV melanoma: the EORTC melanoma cooperative group program. Canc J Scient Am 6:S99, 2000. Kingdom TT, Kaplan MJ: Mucosal melanoma of the nasal cavity and paranasal sinuses. Head Neck 17:184-189, 1995. Lange JR, Raubitschek AA, Pockaj BA, et al: A pilot study of the combination of interleukin-2-based immunotherapy and radiation therapy. J Immunother 12:265, 1992. Lattanzi SC, Tosteson T, Chertoff J, et al: Dacarbazine,cisplatin and carmustine, with or without tamoxifen, for metastatic melanoma: 5-year follow up. Melanoma Res 5:365, 1995. Lee SP, Shimizu KT, Tran LM, et al: Mucosal melanoma of the head and neck: the impact of local control on survival. Laryngoscope 104:121-126, 1994. Legha SS, Ring S, Eton O, et al: Development of a biochemotherapy regimen with concurrent administration of cisplatin, vinblastine, dacarbazine, interferon alfa and interleukin- 2 for patients with metastatic melanoma. J Clin Oncol 16:1752, 1998. Liversedge RL: Oral malignant melanoma. Br J Oral Surg 13:40-55, 1975. Lotze MT, Dallal RM, Kirkwood JM et al: Cutaneous melanoma. In: Cancer: principles and practice of oncology, Eds.: deVita VTJr, Hellman S, Rosenberg, SA, 6th ed, Lippincott Williams and Wilkins, Philadelphia–Baltimore-New York-London-Buenos Aires-Hong Kong–Sydney-Tokyo, 2001, 2012-2069. Lund VJ: Malignant melanoma of the nasal cavity and paranasal sinuses. J Laryngol Otol 96:347-355,1982. Lund VJ: Malignant melanoma of the nasal cavity and paranasal sinuses. ENT J 72:285-290, 1993. Lund VJ, Howard DJ, Harding L, et al: Management options and survival in malignant melanoma of the sinonasal mucosa. Laryngoscope 109:208-211, 1999. Middleton MR, Grob JJ, Aaronson N, et al: Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma. J Clin Oncol 18:158, 2000. Pandey M, Mathew A, Iype EM, et al: Primary malignant mucosal melanoma of the head and neck region: pooled analysis of 60 published cases from India and review of literature. Eur J Cancer Prevention 11:3-10, 2002. Patel SG, Prasad ML, Escrig M et al: Primary mucosal malignant melanoma of the head and neck. Head Neck 24:247-257, 2002. Perez CA, Chao KSC. Mucosal Melanoma. In Principle and Practice of Radiation Oncology., Eds.: Perez CA, Brady LW., 3rd ed, Philadelphia-New York: Lippincott-Raven, 1997, pp.1122-1123. Rinaldo A, Shaha AR, Patel SG, et al: Primary mucosal melanoma of the nasal cavity and paranasal sinuses. Acta Otolaryngol 121:979-982, 2001. Rosenberg Sa, Yanelli Jr, Yang JC, et al: Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin-2. J Natl Cancer Inst 86:1159, 1994. Rosenberg SA, White DE: Vitiligo in patients with melanoma: normal tissue antigens can be targets for cancer immuntherapy. Journal of Immuntherapy with Emphasis on Tumor Immunology 19:81, 1996. Rosenberg Sa, Yang JC, Schwartzentruber DJ, et al: Prospective randomized trial of the treatment of patients with metastatic melanoma using chemotherapy with cisplatin, dacarbazine, and tamoxifen alone or in combination with interleukin-2 and interferon alfa-2b. J Clin Oncol 17:968, 1999. Shah JP, Huvos AG, Strog EW: Mucosal melanomas of the head and neck. Am J Surg 134:531-535, 1977. Shibuya H, Takeda M, Matsumoto S, et al: The efficacy of radiation therapy for malignant melanoma in the mucosa of the upper jaw: an analytic study. Int J Radiat Oncol Biol Phys 25:35-39, 1992. Stern SJ, Guillamondegui OM: Mucosal melanoma of the head and neck. Head Neck 13:22-27, 1991. Tagliaferri P, Barile C, Caraglia M, et al: Daily low-dose subcutaneous recombinant interleukin-2 by alternate weekly administration: antitumor activity and immunomodulatory effects. Am J Clin Oncol 21:48, 1998. Takagi M, Ishikawa G, Mori W: Primary malignant melanoma of the oral cavity in Japan. Cancer 34:358, 1974. Timar J, Csuka O, Orosz Z, et al: Molecular pathology of tumor metastasis. I. Predictive pathology. Pathol Oncol Res 7:217-230, 2001. Trapp TK, Fu Y-S, Calcaterra TC: Melanoma of the nasal and paranasal sinus mucosa. Arch Otolaryngol Head Neck Surg 113:1086-1089, 1987. Trott KR, von Lieven H, Kummermehr J, Skopal D, et al: The radiosensitivity of malignant melanomas. Part I: Experimental studies. Int J Radiat Oncol Biol Phys 7:9-13, 1981. Trott KR, von Lieven H, Kummermehr J, et al. Radiosensitivity of malignant melanomas. Part II: Clinical studies. Int J Radiat Oncol Biol Phys 7:15-20, 1981. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2003 VL 9 IS 1 BP 7 EP 12 PG 6 ER PT J AU De Silva, MCh Reid, R AF De Silva, MV Chandu Reid, Robin TI Gastrointestinal Stromal Tumors (GIST): C-kit Mutations, CD117 Expression, Differential Diagnosis and Targeted Cancer Therapy with Imatinib SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastrointestinal stromal tumors; c-kit; CD117; imatinib ID Gastrointestinal stromal tumors; c-kit; CD117; imatinib AB Gastrointestinal stromal tumors (GISTs) have been recognised as a biologically distinctive tumor type, different from smooth muscle and neural tumors of the gastrointestinal tract. They constitute the majority of gastrointestinal mesenchymal tumors. They are defined and diagnosed by the expression of a protooncogene protein called CD117 detected by immunohistochemistry. It is now believed that GISTs originate from gastrointestinal pacemaker cells known as interstitial cells of Cajal, that control gut motility or from a precursor of these cells. The identification of mutations mostly in exon 11 and to a lesser extent in exons 9 and 13 of the c-kit protooncogene coding for c-kit (CD117) in many GISTs, has resulted in a better understanding of their oncogenic mechanisms.The finding of remarkable antitumor effects of the molecular inhibitor, imatinib (Glivec™ ) in metastatic and inoperable GISTs, has necessitated accurate diagnosis of GISTs and their distinction from other gastrointestinal mesenchymal tumors. To achieve this, pathologists need to be familiar with the spectrum of histological appearances shown by GISTs and have a high index of suspicion for these tumors. This review summarises recent advances in knowledge regarding the histogenesis, pathology, molecular biology, genetics and differential diagnosis of GISTs and the basis for the novel targeted cancer therapy with imatinib. C1 [De Silva, MV Chandu] Western Infirmary, University Department of Pathology, G11 6NT Glasgow, UK. [Reid, Robin] Faculty of Medicine, University of Colombo, University Department of PathologyColombo, Sri Lanka. RP Reid, R (reprint author), Faculty of Medicine, University of Colombo, University Department of Pathology, Colombo, Sri Lanka. CR Andersson J, Sjogren H, Meis-Kindblom JM, et al: The complexity of KIT gene mutations and chromosome rearrangements and their clinical correlation in gastrointestinal stromal, pacemaker cell, tumors. Am J Pathol 160: 15-22, 2002. Appleman HD: Smooth muscle tumors of the gastrointestinal tract: what we now know that Stout didn’t know. Am J Surg Pathol 10, Suppl 1): 83-99, 1986. Beghini A, Tibiletti MG, Roversi G, et al: Germline mutation in the juxtamembrane domain of the kit gene in a family with gastrointestinal stromal tumors and urticaria pigmentosa. Cancer 92: 657-662, 2001. Cooper PN, Quirke P, Hardy GJ, Dixon MF: A flow cytometric, clinical and histologic study of stromal neoplasms of the gastrointestinal tract. Am J Surg Pathol 16: 163-170, 1992. Corless CL, McGreeney L, Haley A, et al: KIT mutations are common in incidental gastrointestinal stromal tumors one centimeter or less in size. Am J Pathol 160:1567-1572, 2002. Cunningham RE, Federspiel BH, McCarthy WF, et al: Predicting prognosis of gastrointestinal smooth muscle tumors. Role of clinical and histologic evaluation, flow cytometry, and image cytometry. Am J Surg Pathol 17: 588-594, 1993. Dematteo RP, Heinrich MC, El-Rifai WM, Demetri G: Clinical management of gastrointestinal stromal tumors: before and after STI-571. Human Pathol 33: 466-477, 2002. El-Rifai W, Sarlomo-Rikala M, Miettinen M, et al: DNA copy number losses in chromosome 14: an early change in gastrointestinal stromal tumors. Cancer Res 56: 3230-3233, 1996. Emory TS, Sobin LH, Lukes L, et al: Prognosis of gastrointestinal smooth muscle, stromal, tumors: dependence on anatomic site. Am J Surg Pathol 23: 82-87, 1999. Fletcher CDM, Berman JJ, Corless C, et al: Diagnosis of gastrointestinal stromal tumors: a consensus approach. Hum Pathol 33: 459-465, 2002. Franquemont DW: Differentiation and risk assessment of gastrointestinal stromal tumors. Am J Clin Pathol 103: 41-47, 1995. Hasegawa T, Matsuno Y, Shimoda T, Hirohashi S: Gastrointestinal stromal tumor: Consistent CD117 immunostaining for diagnosis and prognostic classification based on tumor size and MIB-1 grade. Hum Pathol 33: 669-676, 2002. Heinrich MC, Blanke CD, Druker BJ, Corless CL: Inhibition of KIT tyrosine kinase activity: a novel molecular approach to the treatment of KIT-positive malignancies. J Clin Oncol 20: 1692-1703, 2002. Heinrich MC, Rubin BP, Longley BJ, Fletcher JA:Biology and genetic aspects of gastrointestinal stromal tumors: KIT activation and cytogenetic alterations. Human Pathol 33: 484-495, 2002. Hirota S, Isozaki K, Moriyama Y, et al: Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 279: 577-580, 1998. Hornick JL and Fletcher CDM:Immunohistochemical staining for KIT, CD117, in soft tissue sarcomas is very limited in distribution. Am J Clin Pathol 117: 188-193, 2002. Isozaki K, Terris B, Belghiti J, et al: Germline-activating mutation in the kinase domain of KIT gene in familial gastrointestinal stromal tumors. Am J Pathol 157: 1581-1585, 2000. Joensuu H, Roberts PJ Sarlomo-Rikala M, et al:Effect of tyrosine kinase inhibitor ST1-571 in a patient with a metastatic gastrointestinal stromal tumor. N Engl J Med 344:1052-1056, 2001. Kindblom LG, Remotti HE, Aldenborg F, Meis-Kindblom JM: Gastrointestinal pacemaker cell tumor, GIPACT): Gastrointestinal stromal tumors show phenotypic characteristics of the interstitial cells of Cajal. Am J Pathol 152: 1259-1269, 1998. Lasota J, Jasinski M, Sarlomo-Rikala M, Meittinnen M: Mutations in exon 11 of c-kit occur preferentially in malignant versus benign gastrointestinal stromal tumors and do not occur in leiomyomas and leiomyosarcomas. Am J Pathol 154: 53-60, 1999. Lauwers GY, Erlandson RA, Casper ES, et al: Gastrointestinal autonomic nerve tumors: a clinicopathologic, immunohistochemical and ultrastructural study of 12 cases. Am J Surg Pathol 17: 887-893, 1993. Lecoin L, Gabella G, Le Douarin N: Origin of the c-kit positive interstitial cells in the avian bowel. Development 122:725- 733, 1996. Lee JR, Joshi V, Griffin JW Jr, et al: Gastrointestinal autonomic nerve tumor: immunohistochemical and molecular identity with gastrointestinal stromal tumor. Am J Surg Pathol 25:979- 987, 2001. Lux ML, Rubin BP, Biase TL, et al: KIT extracellular and kinase domain mutations in gastrointestinal stromal tumors. Am J Pathol 156: 791-795, 2000. Maeda H, Yamagata A, Nishikawa S, et al: Requirement of ckit for development of intestinal pacemaker system. Development 156: 791-792, 1992. Mazur MT, Clark HB: Gastric stromal tumors. Reappraisal of histogenesis. Am J Surg Pathol 7: 507-519, 1983. Miettinen M, El-Rifai W, Sobin LH, Lasota J: Evaluation of malignancy and prognosis of gastrointestinal stromal tumors: a review. Hum Pathol 33:478-483, 2002. Miettinen M, Furlong M, Sarlomo-Rikala M, et al: Gastrointestinal stromal tumors, intramural leiomyomas and leiomyosarcomas in the rectum and anus. A clinicopathologic, immunohistochemical and molecular genetic study of 144 cases. Am J Surg Pathol 25: 1121-1133, 2001. Miettinen M, Lasota J: Gastrointestinal stromal tumors-definition, clinical, histological, immunohistochemical, and molecular genetic features and differential diagnosis. Virchows Arch 438: 1-12, 2001. Miettinen M, Sarlomo-Rikala M, Lasota J: Gastrointestinal stromal tumors. Annales Chirurgiae et Gynaecologiae 87: 278- 281, 1998. Miettinen M, Sarlomo-Rikala M, Lasota J: Gastrointestinal stromal tumors:recent advances in understanding of their biology. Hum Pathol 30: 1213-1220, 1999. Miettinen M, Sarlomo-Rikala M, Sobin LH, Lasota J: Esophageal stromal tumors: a clinicopathologic, immunohistochemical and molecular genetic study of 17 cases and comparison with esophageal leiomyomas and leiomyosarcomas. Am J Surg Pathol 24: 211-222, 2000. Miettinen M, Virolainen M, Sarlomo-Rikala M: Gastrointestinal stromal tumors-value of CD34 antigen in their identification and separation from true leiomyomas and schwannomas. Am J Surg Pathol 19: 207-216, 1995. Mikhael AI, Bacchi CE, Zarbo RJ, et al: CD34 expression in stromal tumors of the gastrointestinal tract. Appl Immunohistochem 2: 89-93, 1994. Ming KW: Small intestinal stromal tumors with skeinoid fibers. Clinicopathological, immunohistochemical and ultrastructural investigations. Am J Surg Pathol 16:145-155, 1992. Montgomery E, Torbenson MS, Kaushal M, et al: -catenin immunohistochemistry separates mesenteric fibromatosis from gastrointestinal stromal tumor and sclerosing mesenteritis. Am J Surg Pathol 26: 1296-1301, 2002. Moskaluk CA, Tian Q, Marshall CR, et al: Mutations of c-kit JM domain are found in a minority of human gastrointestinal stromal tumors. Oncogene 18: 1897-1902, 1999. Nakahara M, Isozaki K, Hirota S, et al: A novel gain-of-function mutation of c-kit gene in gastrointestinal stromal tumors. Gastroenterology 115: 1090-1095, 1998. Nishida T, Hirota S, Tangiguchi M, et al: Familial gastrointestinal stromal tumors with germline mutations of the KIT gene. Nat Genet 19: 323-324, 1998. Reith JD, Goldblum JR, Lyles RH, Weiss SW: Extragastrointestinal, soft tissue, stromal tumors: an analysis of 48 cases with emphasis on histological predictors of outcome. Mod Pathol 13: 577-585, 2000. Rosai J. Ackerman’s surgical pathology, 8th ed). Mosby, St Louis, 1995, pp.645-647. Rubin BP, Singer S, Tsao C, et al: KIT activation is a ubiquitous feature of gastrointestinal stromal tumors. Cancer Res 81: 8118-8121, 2001. Debiec-Rychter M, Lasota J, Sarlomo-Rikala M, et al: Chromosomal aberrations in malignant gastrointestinal tumors correlation with c-KIT gene mutation. Cancer Genet Cytogenet 128: 24-30, 2001. Sakurai S, Fukusawa T, Chong JM, et al: Embyonic form of smooth muscle myosin heavy chain, SEmb/MCH-B, in gastrointestinal stromal tumor and interstitial cells of Cajal. Am J Pathol 154: 23-28, 1999. Sarlomo-Rikala M, El-Rifai W, Andersson LC, et al: Different patterns of DNA copy changes in gastrointestinal stromal tumors, leiomyomas and schwannomas. Hum Pathol 29: 476- 481, 1998. Sarlomo-Rikala M, Kovatich AJ, Barusevicius A, Miettinen M: CD117: a sensitive marker for gastrointestinal stromal tumors that is more specific than CD34. Mod Pathol 11: 728-734, 1998. Savage DG, Antman KH: Imatinib mesylate-a new oral targeted therapy. N Engl J Med 346: 683-693, 2002. Sircar K, Hewlett BR, Huizinga JD, et al: Interstitial cells of Cajal as precursors for gastrointestinal stromal tumors. Am J Surg Pathol 23: 377-389, 1999. Suster S: Gastrointestinal stromal tumors. Semin Diagn Pathol 13:297-313, 1996. Tsuura Y, Hiraki H, Watanabe K, et al: Preferential localization of c-kit product in tissue mast cells, basal cells of the skin, epithelial cells of the breast, small cell lung carcinoma and seminoma/dysgerminoma in human: immunohistochemical study of formalin-fixed paraffin-embedded tissues. Virchows Arch 424: 135-141, 1994. Tworek JA, Goldblum JR, Weiss SW, et al: Stromal tumors of the abdominal colon: A clinicopathologic study of 20 cases. Am J Surg Pathol 23: 937-945, 1999. Tworek JA, Goldblum JR, Weiss SW, et al: Stromal tumors of the anorectum: A clinicopathologic study of 22 cases. Am J Surg Pathol 23: 946-954, 1999. van de Rijn M, Hendrickson MR, Rouse RV: The CD34 expression by gastrointestinal stromal tumors. Hum Pathol 25: 766- 771, 1994. Van Oosterom AT, Judson I, Verweij J, et al: Safety and efficacy of imatinib, ST1-571, in metastatic gastrointestinal stromal tumors: A phase 1 study. Lancet 358: 1421-1423,2001. Vliagoftis H, Worobec AS, Metcalfe DD: The proto-oncogene c-kit and c-kit ligand in human disease. J Alleg Clin Immunol 100: 435-440, 1997. Walker P, Dvorak AM: Gastrointestinal autonomic nerve, GAN, tumor: ultrastructural evidence for a newly recognised entity. Arch Pathol Lab Med 110:309-316, 1986. Wardelmann E, Neidt I, Bierhoff E, et al: C-kit mutations in gastrointestinal stromal tumors occur preferentially in the spindle rather than in the epithelioid cell variant. Mod Pathol 15: 125-136, 2002. Yantiss RK, Spiro IJ Compton CC, Rosenberg AE: Gastrointestinal stromal tumor versus intra-abdominal fibromatosis of the bowel wall. Am J Surg Pathol 24: 947-957, 2000. Young HM, Ciampoli D, Southwell BR, Newgreen DF: Origin of interstitial cells of Cajal in the mouse intestine. Dev Biol 180:97-107, 1996. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2003 VL 9 IS 1 BP 13 EP 19 PG 7 ER PT J AU Ribeiro-Silva, A Zamzelli Ramalho, NL Garcia, BS Zucoloto, S AF Ribeiro-Silva, Alfredo Zamzelli Ramalho, N Leandra Garcia, Britto Sergio Zucoloto, Sergio TI Is p63 reliable in detecting microinvasion in ductal carcinoma in situ of the breast? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p63; smooth-muscle actin; ductal carcinoma is situ; microinvasion; breast ID p63; smooth-muscle actin; ductal carcinoma is situ; microinvasion; breast AB P63, a p53 homologue, is considered to be a marker of myoepithelial cells in breast tissue. This study was carried out to determine the sensitivity of p63 in detecting myoepithelial cells in DCIS and to compare the results obtained with smooth-muscle actin (1A4) in an attempt to verify the reliability of p63 as a possible marker of microinvasion in breast carcinoma. Fifteen DCIS of the breast were submitted to immunohistochemical analysis with anti-p63 and 1A4 antibodies and to a double immunolabeling study using p63 with 1A4. The double immunolabeling study showed that the same cells positive for p63 were also positive for 1A4. The three cases of DCIS with micro-invasion were negative for p63 and 1A4 in the foci of invasiveness. P63 staining was continuous in five of twelve cases of DCIS without microinvasion, being focal and discontinuous in 6 cases and completely negative in one case. Smooth-muscle actin staining was continuous in nine of twelve cases, including the five cases positive for p63. Smooth-muscle actin was focal and discontinuous in only two cases, which were also discontinuous for p63. The DCIS negative for p63 was also negative for 1A4. In conclusion, our results confirm the data of literature that p63 is a specific marker of myoepithelial cells in breast tissue. However, p63 is not as sensitive as 1A4 in staining myoepithelial cells and lack of p63 expression cannot be used as a reliable marker of invasiveness in ductal carcinoma in situ of the breast. C1 [Ribeiro-Silva, Alfredo] Sao Paulo University School of Medicine, Department of Pathology, Ribeirao Preto - SP, 14048-900 Sao Paulo, Brazil. [Zamzelli Ramalho, N Leandra] Sao Paulo University School of Medicine, Department of Pathology, Ribeirao Preto - SP, 14048-900 Sao Paulo, Brazil. [Garcia, Britto Sergio] Sao Paulo University School of Medicine, Department of Pathology, Ribeirao Preto - SP, 14048-900 Sao Paulo, Brazil. [Zucoloto, Sergio] Sao Paulo University School of Medicine, Department of Pathology, Ribeirao Preto - SP, 14048-900 Sao Paulo, Brazil. RP Ribeiro-Silva, A (reprint author), Sao Paulo University School of Medicine, Department of Pathology, 14048-900 Sao Paulo, Brazil. EM ardsilva@rpa.fmrp.usp.br CR Barbareschi M, Pecciarini L, Cangi MG, et al.: P63, a p53 homologue, is a selective nuclear marker of myoepithelial cells of the human breast. Am J Surg Pathol 25: 1054-1060, 2001. Damiani S, Ludvikova M, Tomasic G, et al: Myoepithelial cells and basal lamina in poorly differentiated is situ carcinoma of the breast. An immunocytochemical study. Virchows Arch 434: 227-234, 1999. De Mascarel I, MacGrogan G, Mathoulin-Pelissier S, et al: Breast ductal carcinoma in situ with microinvasion: a definition supported by a long-term study of 1248 serially sectioned ductal carcinomas. Cancer 94: 2134-2142, 2002. Di Como CJ, Urist MJ, Babayan I, et al: P63 expression profiles in human normal and tumor tissues. Clin Cancer Res 8: 494-501, 2002. Douglas-Jones AG, Gupta SK, Attanoos RL, et al: A critical appraisal of six modern classifications of ductal carcinoma in situ of the breast, DCIS): Correlation with grade of associated invasive carcinoma. Histopathology 29: 397-409, 1996. Foschini MP, Scarpellini F, Gown AM, et al.: Differential expression of myoepithelial markers in salivary, sweat and mammary glands. Int J Surg Pathol 8: 293-7, 2000. Hoda SA, Prasad ML, Moore A, et al: Microinvasive carcinoma of the breast: can it be diagnosed reliably and its clinically significant? Histopathology 35: 468-472, 1999. Irwin MS, Kaelin WG Jr: P53 family update: p73 and p63 develop their own identities. Cell Growth Differ 12: 337-349, 2001. Lazard D, Sastre X, Frid MG, et al: Expression of smooth musclespecific proteins in myoepithelium and stromal myofibroblasts of normal and malignant human breast tissue. Proc Natl Acad Sci USA 90: 999-1003, 1993 Little NA, Jochemsen AG:P63. Int J Biochem Cell Biol 34: 6-9, 2002. Miller NA, Chapman JA, Fish EB, et al: In situ carcinoma of the breast: clinical and histopathologic factors and association with recurrent carcinoma. Breast J 7: 292-302, 2001. Mills AA, Zheng B, Wang XJ, et al: P63 is a p53 homologue required for limb and epidermal morphogenesis. Nature 398: 708-713,1 999. Moritani S, Kushima R, Sugihara H, et al: Availability of CD10 immunohistochemistry as a marker of breast myoepithelial cells on paraffin sections. Mod Pathol 15: 397-405, 2002. Polyak K: On the birth of breast cancer. Biochim Biophys Acta 1552: 1-13, 2001. Reis-Filho JS, Albergaria A, Milanezi F, et al: Naked nuclei revisited: p63 immunoexpression. Diagn Cytopathol 27:135-138, 2002. Reis-Filho JS, Milanezi F, Amendoeira I, et al: P63 staining of myoepithelial cells in breast fine needle aspirates: a study of its role in differentiating in situ from invasive ductal carcinomas of the breast. J Clin Pathol 55: 936-939, 2002. Reis-Filho JS, Schmitt FC: Taking advantage of basic research: p63 is a reliable myoepithelial and stem cell marker. Adv Anat Pathol 9: 280-289, 2002. Signoretti S; Waltregny D, Dilks J, et al: P63 is a prostate basal cell marker and is required for prostate development. Am J Pathol 157: 1769-1775, 2000. 190.Silver SA, Tavassoli F: Mammary ductal carcinoma in situ with microinvasion. Cancer 82:2382-2390, 1998. 20. Silverstein MJ, Poller DN, Waisman J, et al.: Prognostic classification of breast ductal carcinoma in situ. The Lancet 345:1154-1157, 1995. 210.Skinner KA, Silverstein MJ: The management of ductal carcinoma in situ of the breast. Endocr Relat Cancer 8: 33-45, 2001. 22. Smith GH, Chepko G: Mammary epithelial stem cells. Microsc Res Tech 52: 190-197, 2001. 23. Yang A, Schweitzer R, Sun D, et al: P63 is essential for regenerative proliferation in limb, craniofacial and epithelial development. Nature 398: 714-718, 1999. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2003 VL 9 IS 1 BP 20 EP 23 PG 4 ER PT J AU Bencsath, M Blaskovits, A Borvendeg, J AF Bencsath, Marta Blaskovits, Aladar Borvendeg, Janos TI Biomolecular Cytokine Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chronic inflammation; therapy; cytokines; infliximab; etanercept ID chronic inflammation; therapy; cytokines; infliximab; etanercept AB As for the chronicity of inflammatory-immune diseases, the medication of them needs to be longterm and thus, quite safe with respect to side effects due to drug actions. Therapy of these diseases includes steroid and non steroid anti-inflammatories given in monotherapy or in combination with cytotoxic antimetabolites. Longterm administration of these active substances cumulate in side effects, not to speak of the probability of developing unresponsiveness to the drug in use. In principle, the earlier the intervention, the better the outcome of medication in therapy. In harmony with this principle, biopharmacology focuses on specific targets in early (acute) phase of inflammatory-immune diseases. One of these targets is the proinflammatory cascade of cytokines (IL1beta, IL6, IL8, IL12, TNFalpha). Among them, the overproduction of tumor necrosis factor (TNFalpha) is suggested to orchestrate and escalate the disease phenotype. Hence, targeting of TNFa may restrict or stop the propagation of pathological reactions. TNFalpha in its excess can be captured at transcription, translation, secretion levels as well as in the extracellular soluble form. This latter approach is supported by clinical records emphasizing the use of recombinant antibodies and soluble receptors in trapping extra amounts of TNFalpha. This review serves as an illustration for the efficacy and safety of infliximab (antibody) and etanercept (soluble receptor) in the example of rheumatoid arthritis (RA). C1 [Bencsath, Marta] National Institute of Pharmacy, Zrinyi u. 3, 1051 Budapest, Hungary. [Blaskovits, Aladar] National Institute of Pharmacy, Zrinyi u. 3, 1051 Budapest, Hungary. [Borvendeg, Janos] National Institute of Pharmacy, Zrinyi u. 3, 1051 Budapest, Hungary. RP Bencsath, M (reprint author), National Institute of Pharmacy, 1051 Budapest, Hungary. EM bencsath.marta@ogyi.hu CR Barclay AN, Brown MH, Law SKA, et al: The Leukocyte Antigen Facts Book. Academic Press, 2nd edition, 1997. Baud V, Liu Z-G, Bennett B, et al: Signaling by proinflammatory cytokines: oligomerization of TRAF2 and TRAF6 is sufficient for JNK and IKK activation and target gene induction via an amino-terminal effector domain. Genes Develop 13: 1297- 1308, 1999. Brown SL, Greene MH, Gershon SK, et al: Tumor necrosis factor antagonist therapy and lymphoma development : twenty-six cases reported to the Food and Drug Administration. Arthritis Rheum 46: 3151-3158, 2002. Beyaert R, Fiers W: Tumor necrosis factor and lymphotoxin. In: Cytokines, Eds: Mire-Sluis A, Thorpe R, Academic Press, 1998, pp. 335–360. Budd RC: Death receptors couple to both cell proliferation and apoptosis. J Clin Invest 109: 437–442, 2002. Choy EHS, Panayi GS: Cytokine pathways and joint inflammation in rheumatoid arthritis. N Engl J Med 344: 907–916, 2001. Economides AN,Carpenter LR,Rudge JS, et al: Cytokine traps: multi-component, high-affinity blockers of cytokine action. Nat Med 9: 47-52, 2003. Goodman Gilman A, Rall TW, Nies AS, Taylor P: The pharmacological basis of therapeutics. Pergamon Press – 8th edition, 1990. Hayashida K, Nanki T, Girschick H, et al: Synovial stromal cells from rheumatoid arthritis patients attract monocytes by producing MCP and IL8. Arthritis Res 3: 118–126, 2001. Jensen PE, Weber DA, Thayer WP, et al: Peptide exchange in MHC molecules. Immunol Rev 172: 229–238, 1999. Keane J, Gershon S, Wise RP, et al: Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent. N Engl J Med 345: 1098-1104, 2001. Kvien TK, Uhlig T, Kristiansen IS: Criteria for TNF-targeted therapy in rheumatoid arthritis. Drugs 61: 1711-1720, 2001. Lee DM, Weinblatt ME: Rheumatoid arthritis – Review. Lancet 358: 903–911, 2001. Li Q, Verma IM: NFB regulation in the immune system. Nat Rev Immunol 2: 725-734, 2002. Liu ZG, Hsu H, Goeddel DV, Karin M: Dissection of TNF receptor 1 effector functions: JNK activation is not linked to apoptosis while NF-kappaB activation prevents cell death. Cell 87: 565–576, 1996. Morita Y, Yang J, Gupta R, et al: Dendritic cells genetically engineered to express IL-4 inhibit murine collagen-induced arthritis. J Clin Invest 107: 1275–1284, 2001. Nakajima A, Seroogy CM, Sandora MR, et al: Antigen-specific T cell-mediated gene therapy in collagen-induced arthritis. J Clin Invest 107: 1293–1301, 2001. Nanki T, Lipsky PE: Cytokine, activation marker, and chemokine receptor expression by individual CD4+ memory T cells in rheumatoid arthritis synovium. Arthritis Res 2: 415–423, 2000. PubMed/Swissprot Accession No P20333. Reed JC: Apoptosis-based therapies. Nat Rev Drug Disc 1: 111-121, 2002. Yung RL: Etanercept Immunex. Curr Opin Investig Drugs 2: 216–221, 2001. Vingsbo-Lundberg C, Nordquist N, Olofsson P, et al: Genetic control of arthritis onset, severity and chronicity in a model for rheumatoid arthritis in rats. Nat Genet 4: 401-404, 1998. Wagner UG, Koetz K, Weyand CM, Goronzy JJ: Perturbation of the T cell repertoire in rheumatoid arthritis. Proc Natl Acad.Sci USA 95: 14447–14452, 1998. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2003 VL 9 IS 1 BP 24 EP 29 PG 6 ER PT J AU NG, Y Husain, I Waterfall, N AF NG, Yeung Husain, Imtiaz Waterfall, Naeha TI Diabetes Mellitus and Bladder Cancer - An Epidemiological Relationship? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article AB An epidemiological association between diabetes mellitus and transitional cell carcinoma of the bladder has been proposed. This study looked retrospectively at 125 patients with transitional cell carcinoma of the bladder as a study group and 80 other hospital patients with conditions not specifically associated with diabetes mellitus as a control group. Diabetic patients had an increased, significant odds ratio for bladder cancer compared with non diabetics even after adjustment for smoking and age [OR: 2.69 p=0.049 (95% CI 1.006-7.194)] A history of smoking OR 2.16 p=0.013 (95% C.I. 1.175-3.964) is a significant independent association with transitional cell carcinoma of the bladder as is age: p=0.001 OR 1.07. We propose potential pathogenic pathways for transitional cell carcinoma of the bladder in diabetic patients based on altered integrin and cadherin distribution in urothelial cells in diabetic patients. A larger study is planned to confirm an association between diabetes mellitus and transitional cell carcinoma of the bladder. C1 [NG, Yeung] University of Dundee, Department of Surgery and Molecular OncologyDundee, UK. [Husain, Imtiaz] Bedford General HospitalBedford, UK. [Waterfall, Naeha] Bedford General HospitalBedford, UK. RP NG, Y (reprint author), University of Dundee, Department of Surgery and Molecular Oncology, Dundee, UK. EM y.ng@dundee.ac.uk CR Kravchik S, Gal R, Cytron S et al: Increased Incidence of Diabetes Mellitus in Patients with TCC of Urinary Bladder. Path Oncol Res 7: 56-59, 2001. Rische H, Burch J; Miller A et al: Dietary Factors and the Incidence of Cancer of the Urinary Bladder. Am J Epidemiol 127: 1179-119, 1988. Glashan R, Cartwright R: Occupational Bladder Cancer and Cigarette Smoking in West Yorkshire. Br J Urology 53: 602- 604, 1981. Clavel J, Cordier S, Boccon-Gibod L, Hemon D: Tobacco and Bladder Cancer in Males: Increased Risk for Inhalers and Smokers of Black Tobacco. Int J Cancer 44: 605-610, 1989. Singh AK, Mo W, Dunea G et al: Effect of Glycated Proteins on the Matrix of Glomerular Epithelial Cells. J Am Soc Nephrol 9: 802-10, 1998. Chen HC, Chen CA, Guy JY et al: Altering Expession of Alpha1Beta3 Integrin on Podocytes of Human and Rats with Diabetes. Life Sci 67: 2345-2353, 2000. Lipponen PK, Eskelinen MJ: Reduced Expression of E-cadherin is Related to Invasive Disease and and Frequent Recurrence in Bladder Cancer. J Cancer Res Clin Oncol 53: 3241- 3245, 1995. Bringuier PP, Umbas R, Schaafsma HE et al: Decresed Cadherin Immunoreactivity correlates with poor survival in Patients with Bladder Tumours. Cancer Res 56: 4154-4158, 1996. Byrne RR, Shariat SF, Brown R et al. E-cadherin immunostaining of bladder transitional cell carcinoma, carcinoma in situ and lymph node metastases with long term followup. J Urol 165: 1473-1479, 2001. Liebert M, Washington R et al: Loss of Colocalisation of Alpha 6 Beta 4 Integrin and Collagen VII in Bladder Cancer. Am J Pathol 144: 287-299, 1994. Jin DK, Fish AJ, Wayner EA et al: Distribution of Integrin Subunits in Human Diabetic Kidneys. J Am Soc Nephrol 7: 2636- 2645, 1996. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2003 VL 9 IS 1 BP 30 EP 31 PG 2 ER PT J AU Elek, G Gyori, S Toth, B Pap, AF Elek, Gabor Gyori, Sandor Toth, Bernadett Pap, Akos TI Histological Evaluation of Preoperative Biopsies from Ampulla Vateri SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE papilla Vateri; preoperative diagnosis; sampling error; papillectomy; biopsy of Vater papilla; pathohistology; forceps biopsy ID papilla Vateri; preoperative diagnosis; sampling error; papillectomy; biopsy of Vater papilla; pathohistology; forceps biopsy AB Frequency of the lesions of the papilla Vateri is increasing in Hungary because of epidemiological reasons. Over two years nearly 300 ampullary endoscopic biopsies were taken in our hospital. In 36 percent of the patients the papillary specimens demonstrated acute or chronic inflammation, in 44 percent adenoma, including 5 percent with severe dysplasia, in 5 percent adenomatous hyperplasia and in 7 percent adenomyosis or other benign tumors (2%) were found. Around 7 percent of the ampullary samples proved to be malignant, but only in 2.6 percent were the malignancy of intraampullary origin. Nearly 25 percent of biopsies were repeated once and 10 percent twice or more. Concordance of endoscopic and pathologic diagnoses was 69 percent on average but it increased to 83 percent after including repeated biopsies. In the adenoma-carcinoma group the concordance was 90 percent. The sensitivity of the pathological diagnosis with forceps biopsy was only 77 percent, but it became at least 86 percent following papillectomy. In order to improve diagnostic reliability more extensive use of papillectomy is proposed with close cooperation between the endoscopist and pathologist. C1 [Elek, Gabor] Central Railway Hospital and Polyclinic, Department of Pathology, Podmaniczky street 111, 1062 Budapest, Hungary. [Gyori, Sandor] Central Railway Hospital and Polyclinic, Department of Pathology, Podmaniczky street 111, 1062 Budapest, Hungary. [Toth, Bernadett] Central Railway Hospital and Polyclinic, Department of Pathology, Podmaniczky street 111, 1062 Budapest, Hungary. [Pap, Akos] Central Railway Hospital and Polyclinic, Department of Gastroenterology, Podmaniczky street 111, 1062 Budapest, Hungary. RP Pap, (reprint author), Central Railway Hospital and Polyclinic, Department of Gastroenterology, 1062 Budapest, Hungary. EM papakos@kkk.sote.hu CR Albores-Saavedra J, Henson DE, Klimstra DS: Tumors of the Gallbladder, extrahepatic bile ducts, and ampulla of Vater. Atlas of tumor pathology, Third series, Fascicle 27, Washington, 2000, pp 10-19, 44, 149-156, 207-209 and 245-358. Albores-Saavedra J, Murakata L, Krueger JE, et al: Noninvasive and minimally invasive papillary carcinomas of the extrahepatic bile ducts. Cancer 89: 508-515, 2000. Alarcon JF, Burke CA, Church JM, et al: Familial adenomatous polyposis. Dis Colon Rectum 42: 1533-1536, 1999. Allgaier HP, Schwacha H, Kleinschmidt M, et al: Ampullary hamartoma. Digestion 60: 497-500, 1999. Avisse C, Flament JB, Delattre JF: Ampulla of Vater. Surgical clinics of North America 80: 201-212, 2000. Bajtai A, Juhasz L, Lonovics J, et al: The papilla of Vater. Melania Publishing Ltd, Budapest, 1995, pp 16-81. Balgha V, Topa L, Simon K, et al: A Vater papilla tumoros megbetegedesei. Orv Hetil 138: 1387-1391, 1997. Beger HG, Treitschke F, Gansauge F, et al: Tumor of the ampulla of Vater. Arch Surg 134: 526-532, 1999. Benchamiche AM, Jouve JL, Manfredi S, et al: Cancer of the ampulla of Vater: results of a 20-year population based study. Eur J Gastroenterol Hepatol 12: 75-79, 2000. Berczi L, Bocsi J, Lapis K, et al: Relationship between the survival and the clinicopathological parameters of the patients with tumors in the pancreatic head region. Acta chirurgica Hung 38: 235-241,1999. Clary BM, Tyler DS, Dematos P, et al: Local anpullary resection with careful intraoperative frozen section evaluation for presumed benign ampullary neoplasms. Surgery 127: 628-633, 2000. Gouma DJ, Obertrop H: Centralisation of surgery for periampullary malignancy. Br J Surg 86: 1361-1362, 1999. Holle G: Neue Befunde zur Morphologie und Physiologie der Vaterschen Papille. Zschr arztl Fortbild 57: 402-409, 1963. Kim HJ, Kim MH, Lee KS, et al: Mucin hypersecreting bile duct tumor characterized by a striking homology with an intraductal papillary mucinous tumor of the pancreas. Endoscopy 32: 389-393, 2000. Kimchi NA, Mindrul V, Brodie E, et al: The contribution of endoscopy and biopsy to the diagnosis of periampullary tumors. Endoscopy 30: 538-543, 1998. Kimura W, Otsubo K: Incidence, sites of origin, and immunohistochemical and histochemical characteristics of atypical epithelium and minute carcinoma of the papilla of Vater. Cancer 61: 1394-1402, 1988. Kubo H, Chijiiwa Y, Akahoshi K, et al: Pre-operative staging of ampullary tumours by endoscopic ultrasound. Br J Radiol 72: 443-447, 1999. Maacke H, Kessler A, Schmiegel W, et al: Overexpression of p53 protein during pancreatitis. Br J Cancer 75: 1501-1504, 1997. Matsubayashi H, Watanabe H, Yamaguchi T, et al: Differences in mucus and K-ras mutation in relation to phenotypes of tumors of the papilla of Vater. Cancer 86: 596-607, 1999. Matsumoto T, Iida M, Nakamura S, et al: Natural history of ampullary adenoma in famialial adenomatous polyposis. Am J Gastroenterol 95: 1557-1562, 2000. Mouzas I, Skordilis P, Frangiadakis N, et al: Carcinoma of the ampulla of Vater in Crete. Anticancer Res 19: 4501-4505, 1999. Pap A, Burai M: Endoscopic papillectomy for adenoma of papilla Vateri. Z Gastroenterol 37: 438, 1999. Park SH, Kim YI, Park YH, et al: Clinicopathological correlation of p53 overexpression in adenoma and carcinoma of the ampulla of Vater. World J Surg 24: 54-59, 2000. Park SW, Song SY, Chung JB, et al: Endoscopic snare resection for tumors of the ampulla of Vater. Yonsei Med J 41: 213-218, 2000. Perzin KH, Bridge MF: Adenomas of the small intestine. Cancer 48: 799-819, 1981. Sato T, Konishi K, Kimura H, et al: Adenoma and tiny carcinoma in adenoma of the papilla of Vater - p53 and PCNA. Hepato- Gastroenterol 46: 1959-1962, 1999. Sellner F: Investigations on the significance of the adenoma-carcinoma sequence in the small bowel. Cancer 66: 702-715, 1990. Sellner F, Riegler FM, Machacek E: Implications of histological grade of tumour for the prognosis of radically resected periampullary adenocarcinoma. Eur J Surg. 165: 865-870, 1999. Simon K, Balgha V, Gal I, et al: Value of p53 immunohistochemistry in Vater papilla tumors. Z Gastroenterol 35: 438, 1997. Talbot IC, Neoptolemos JP, Shaw DE: The histopathology and staging of carcinoma of the ampulla of Vater. Histopathology 12: 155-165, 1988. Tascilar M, Sturm PDJ, Caspers E, et al: Diagnostic p53 immunostaining of endobiliary brush cytology. Cancer 87: 306-311, 1999. Yamaguchi K, Enjoji: Carcinoma of the ampulla of Vater. Cancer 59: 506- 515, 1987. Varsanyi M, Gyokeres T, Burai M, et al: Diagnostic and therapeutic pitfalls of periampullary tumours. Z Gastroenterol 39: 428, 2001. Will U, Bosseckert H, Schroder H, et al: Probleme in der Diagnostik und Therapie des juxtapapillaren, tubulovillosen Adenoms. Z. Gastroenterol 37: 1013-1017, 1999. Witzigmann H, Mobius Ch, Uhlmann D, et al: Behandlungskonzept von Adenomen der Papilla Vateri. Chirurg 71: 197-201, 2000. Zhao B, Kimura W, Futakawa N, et al: p53 and p21/ Waf1 protein expression and K-ras codon 12 mutation in carcinoma of the papilla of Vater. Am J Gastroenterol 94: 2128-2134, 1999. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2003 VL 9 IS 1 BP 32 EP 41 PG 10 ER PT J AU Hortovanyi, E Illyes, Gy Kadar, A AF Hortovanyi, Eszter Illyes, Gyorgy Kadar, Anna TI Early Atherosclerosis and Chlamydia Pneumoniae Infection in the Coronary Arteries SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chlamydia pneumoniae; early; atherosclerosis; hypertrophy ID chlamydia pneumoniae; early; atherosclerosis; hypertrophy AB In non-atheromatous segments of coronary arteries a sequence of preatherosclerotic changes was identified which consisted of medial thickening followed by intimal thickening. More recently, Chlamydia pneumoniae seropositivity was associated with enhanced intima-media thickness of arteries. In the present study the intimal and medial thickness of coronary artery of young adults were measured, and were correlated with the presence of Chlamydia pneumoniae antigens. Proximal and distal segments of the left anterior descending coronaries (LAD) obtained at autopsy from young adults (15-34 years) were studied. The thickness and cellular density of the intima and of the media without clear-cut atherosclerotic changes were measured by image analysis. The hypertrophy index was calculated as the ratio of cell density and the thickness of the respective layer. Atherosclerotic lesions occurring elsewhere in the same coronary were noted and graded by severity. The presence of Chlamydia pneumoniae verified by immunohistochemistry was correlated with the severity of lesions and with the hypertrophy index. In the proximal segments, atherosclerosis of LAD was associated with the widening of both the intima and the media of lesion free-sites. In the distal coronary segments the proportion of the intimal thickening had a significant association with atherosclerosis. Compared to non-infected arteries, Chlamydia pneumoniae infection was associated with higher hypertrophy index in the intima as well as in the media. The rate of Chlamydia pneumoniae positivity increased with the severity of lesions. As a conclusion: in the LAD coronary, the intimal thickening is the main preatherosclerotic change. Chlamydia pneumoniae may favour arterial wall hypertrophy and plays a role in lesion progression. C1 [Hortovanyi, Eszter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. [Illyes, Gyorgy] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. [Kadar, Anna] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. RP Kadar, A (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM kadann@korb2.sote.hu CR Berger M, Schroder B, Daeschlein G, Schneider Wet al.: Chlamydia pneumoniae DNA in non-coronary atherosclerotic plaques and circulating leukocytes. J Lab Clin Med 136: 194- 200, 2000. Davidson M, Kuo CC, Middaugh J et al.: Confirmed previous infection with Chlamydia pneumoniae, TWAR, and its presence in early coronary atherosclerosis. Circulation 98: 628-33, 1998. Gurfinkel E, Bozovich G, Darcoca A et al.: Randomized trial of roxithromycin in non-Q-wave coronary syndromes: ROXIS pilot study. Lancet 350: 404-7, 1997. Ericson K, Saldeen TG, Lindquist O et al.: Relationship of Chlamydia pneumoniae infection to severity of human coronary atherosclerosis. Circulation 101: 2568-71, 2000. Espinola-Klein C, Rupprecht HJ, Blankenberg S et al.: Are morphological or functional changes in the carotid artery wall associated with Chlamydia pneumoniae, Helicobacter pylori, cytomegalovirus, or herpes simplex virus infection? Stroke 31:2127-33, 2000. Hortovanyi E, Illyes Gy, Glasz T, Kadar A: Chlamydia pneumoniae in different coronary artery segments in the young. Pathol Res Pract 198: 19-23, 2002. Kadar A., Mozes G, Illyes G et al.: Histomorphometry and histochemistry of atherosclerotic lesions in coronary arteries and the aorta in young population. World Health Organization, WHO, and the World Heart Federation, WHF, Pathobiological Determinants of Atherosclerosis in Youth Study, WHO/WHF PBDAY Study, 1986-1996. Nutr Metab Cardivasc Dis 9:220-227, 1999. Kuo CC, Grayston JT, Campbell LA et al.: Chlamydia pneumoniae, TWAR, in coronary arteries of young adults, 15-34 years old, Proc Natl Acad Sci USA 92: 6911-14, 1995. Mayr M, Kiechl S, Willeit Jet al.: Associations of antibodies to Chlamydia pneumoniae, Helicobacter pylori, and Cytomegalovirus with immune reactions to Heat-shock protein 60 and carotid or femoral atherosclerosis. Circulation 102:833-39, 2000 Molestina RE, Dean D, Miller RD et al.: Characterization of a strain of Chlamydia pneumoniae isolated from a coronary atheroma by analysis of the omp1 gene and biological activity in human endothelial cells. Infect Immun 66:1370-6, 1998, Ross R: Atherosclerosis – an inflammatory disease. N Engl J Med 340: 115-25, 1999. Stary HC: Composition and classification of human atherosclerotic lesions. Virchows-Archiv-A-Pathol-Anat 421:277, 1992. Stary HC: The Histological classification of atherosclerotic lesions in human coronary arteries. Atherosclerosis and Coronary Artery Disease Chapter 26 pp 463-474. Lippincott-Raven Publishers, Philadelphia, 1996. Tracy RE: Medial thickness of coronary arteries as a correlate of atherosclerosis. Atherosclerosis 139:11-19. 1998. Tracy RE: Medial thickenings of coronary artery and the aging risk factor for atherosclerosis. Atherosclerosis 155: 337–346, 2001. Vehmaan-Kreula P, Puolakkainen M, Sarvas M et al.: Chlamydia pneumoniae proteins induce secretion of the 92-kDa gelatinase by human monocyte-derived macrophages. Arterioscler Thromb Vasc Biol 21:1-8, 2001. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2003 VL 9 IS 1 BP 42 EP 46 PG 5 ER PT J AU Pecorella, I Lucas, BS Ciardi, A Memeo, L Miller, FR AF Pecorella, Irene Lucas, B Sebastian Ciardi, Antonio Memeo, Lorenzo Miller, F Robert TI Calcium Oxalate Precipitates in a Renomedullary Interstitial Cell Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE calcium oxalate; kidney; renomedullary interstitial cell tumor ID calcium oxalate; kidney; renomedullary interstitial cell tumor AB We report a case of calcium oxalate deposition in a renomedullary interstitial cell tumor (RICT) in a patient dying of full-blown AIDS. The precipitates showed birefringence using a partially polaris ed light and were stained black in Yasue’s silver nitrate-rubeanic acid method. The combination of calcium oxalosis and RICT has not been reported before and might possibly be due to systemic biochemical alterations of the glycosaminoglycans as a result of profound metabolic disturbances in AIDS patients. C1 [Pecorella, Irene] Universita degli Studi La Sapienza, Dipartimento di Medicina Sperimentale e Patologia, Viale Regina Elena, 324, 00161 Rome, Italy. [Lucas, B Sebastian] Guy's Kings and St Thomas's School of Medicine, St Thomas's Hospital, Department of HistopathologyLondon, UK. [Ciardi, Antonio] Universita degli Studi La Sapienza, Dipartimento di Medicina Sperimentale e Patologia, Viale Regina Elena, 324, 00161 Rome, Italy. [Memeo, Lorenzo] Universita degli Studi La Sapienza, Dipartimento di Medicina Sperimentale e Patologia, Viale Regina Elena, 324, 00161 Rome, Italy. [Miller, F Robert] Windeyer Institute of Medical Sciences, Royal Free and University College Medical School and Camden and Islington Community Health Services Trust, Department of Sexually Transmitted DiseasesLondon, UK. RP Pecorella, I (reprint author), Universita degli Studi La Sapienza, Dipartimento di Medicina Sperimentale e Patologia, 00161 Rome, Italy. EM irenepecorella@virgilio.it CR Chaplin AJ. Histopathological occurrence and characterisation of calcium oxalate: a review. J Clin Pathol 30: 800-811, 1977. Lerman RJ, Pitcock JA, Stephenson P, Muirhead EE. Renomedullary interstitial cells tumor, formerly fibroma of renal medulla). Hum Pathol 3: 559-568, 1972. Memeo L, Pecorella I, Ciardi A,et al: Calcium oxalate microdeposition in failing kidney grafts. Transpl Proc 33: 1262-1265, 2001. Pecorella I, McCartney ACE, Lucas S, et al: Histological study of oxalosis in the eye and adnexa of AIDS patients. Histopathology 27: 431-438, 1995. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2003 VL 9 IS 1 BP 47 EP 48 PG 2 ER PT J AU Timar, J Ladanyi, A Petak, I Jeney, A Kopper, L AF Timar, Jozsef Ladanyi, Andrea Petak, Istvan Jeney, Andras Kopper, Laszlo TI Molecular Pathology of Tumor Metastasis III. SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE metastasis therapy; molecular target; cancer cell; host cell ID metastasis therapy; molecular target; cancer cell; host cell AB Therapy of tumor progression and the metastatic disease is the biggest challenge of clinical oncology. Discovery of the diverse molecular pathways behind this complex disease outlined an approach to better treatment strategies. The development of combined cytotoxic treatment protocols has produced promising results but no breakthrough in the clinical management of metastatic disease. The multiple - specific and non-specific pathways and cellular targets of tumor progression are outlined in this review. Such an approach, individually designed for various cancer types, may have a better chance to treat or even cure cancer patients with progressive disease. C1 [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. [Ladanyi, Andrea] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. [Petak, Istvan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Jeney, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, 1122 Budapest, Hungary. CR Timar J, Csuka O, Orosz Zs et al: Molecular pathology of tumor metastasis. I. Predictive Pathology. Pathol Oncol Res 7: 217-230, 2001. Timar J, Csuka O, Orosz Zs et al: Molecular pathology of tumor metastasis. II. Molecular staging and differential diagnosis. Pathol Oncol Res 8: 204-219, 2002. Thierry JP: Epithelial mesenchymal transitions in tumor progression. Nature Rev Cancer 2: 442-454, 2002. Chambers AF, Groom AC, MacDonald IC: Dissemination and growth of cancer cells in metastatic sites. Nature Rev Cancer 2: 563-572, 2002. Ramaswamy S, Ross KN, Golub TR: A molecular signature of metastasis in primary tumors. Nature Genetics 33: 1-6, 2003. Fidler IJ and Kripke ML: Metastasis results from pre-existing variant cells within a malignant tumor. Science 197:893-895, 1977. Liotta A and Kohn EC: The microenvironment of the tumorhost interface. Nature 411:375-379, 2001. Braun et al: Cytokeratin positive cells in the bone marrow and survival of patiens with stage I, II, or III breast cancer. N England J Med 329:257-263, 2000. Hood JD and Cheresh DA: Role of integrins in cell invasion and migration. Nature Rev Cancer 2: 91-100, 2002. Yamamoto Y, Tsutsumi Y, Mayumi T: Molecular design of bioconjugated cell adhesion peptide with a water-soluble polymeric modifier for enhancement of antimetastatic effect. Curr Drug Targets 3:123-130, 2002. Buerkle MA, Pahernik SA, Sutter A et al: Inhibition of the alpha-nu integrins with a cyclic RGD peptide impairs angiogenesis, growth and metastasis of solid tumours in vivo. Br J Cancer 86: 788-795, 2002. Yi M and Ruoslahti E: A fibronectin fragment inhibits tumor growth, angiogenesis, and metastasis. PNAS 98: 620-624, 2001. Trikha M, De Clerck YA, Markland FS: Contortrostatin, a snake venom disintegrin, inhibits beta 1 integrin-mediated human metastatic melanoma cell adhesion and blocks experimental metastasis. Cancer Res 54: 4993-4998, 1994. Zhou Q, Sherwin RP, Parrish C et al: Contortrostatin, a dimeric disintegrin from Agkistrodon contortrix inhibits breast cancer progression. Breast Cancer Res Treat 61: 249- 260, 2000. Correa MC Jr, Maria DA, Moura-da-Silva AM et al: Inhibition of melanoma cells tumorigenicity by the snake venom toxin jararhagin. Toxicon 40: 739-748, 2002. Ritter MR, Zhou Q, Markland FS Jr: Contortrostatin, a homodimeric disintegrin, actively disrupts focal adhesion and cytoskeletal structure and inhibits cell motility through a novel mechanism. Cell Commun Adhes 8: 71-86, 2001. Gutheil JC, Campbell TN, Pierce PR et al: Targeted antiangiogenic therapy for cancer using Vitaxin: a humanized monoclonal antibody to the integrin alphavbeta3. Clin Cancer Res 30: 3056-3061, 2000. Cohen SA, Trikha M, Mascelli MA: Potential future clinical applications for the GPIIb/IIIa antagonist, abciximab in thrombosis, vascular and oncological indications. Pathol Oncol Res 6: 163-174, 2000. Trikha M, Zhou Z, Timar J, et al: Multiple roles for platelet GPIIb/IIIa and avb3 integrins in tumor growth, angiogenesis, and metastasis. Cancer Res 62: 2824-2833, 2002. Chen YO, Trikha M, Gao X, et al.: Ectopic expression of platelet integrin aIIibb3 in tumor cells from various species and histological origin. Int J Cancer 72: 642-648, 1997. Damiano JS: Integrins ad novel drug targets for overcoming innate drug resistance. Current Cancer Drug Targets 2: 37-43, 2002. Favoni RE, de Cupis A: The role of polypeptide growth factors in human carcinomas: New targets for a novel pharmacological approach. Pharmacol Rev 52: 179-206, 2000. Zugmaier G, Lippman ME, Wellstein A:Inhibition by pentosan polysulfate, PRS, of heparin-binding growth factors relased from tumor cells and blockage by PRS of tumor growth in animals. J Natl Cancer Res 84: 1716-1724, 1992. Timar J, Dome B, Fazekas K et al: Angiogenesis-dependent diseases and angiogenesis therapy. Pathol Oncol Res 7: 85-94, 2001. Baselga J: Why the epidermal growth factor receptor? The rationale for cancer therapy. Oncologist 7: 2-8, 2002. Shak S, and the Herceptin Multinational Investigator Study Group: Overview of the trastuzumab, Herceptin, anti-HER2 monoclonal antibody clinical program in HER2-overexpressing metastatic breast cancer. Semin Oncol 26: 71-77, 1999. Sliwkowski MX, Lofgren JA, Lewis GD et al: Nonclinical studies addressing the mechanism of action of trastuzumab, Herceptin). Semin Oncol 26(Suppl 12): 60-70, 1999. Herbst RS, Hong WK: IMC-C225, an anti-epidermal growth factor receptor monoclonal antibody for treatment of head and neck cancer. Semin Oncol 29(Suppl 14): 18-30, 2002. Huang SM, Li J, Harari PM: Molecular inhibition of angiogenesis and metastatic potential in human squamous cell carcinomas after epidermal growth factor receptor blockade. Mol Cancer Ther 1: 507-514, 2002. Fazekas K, Csuka O, Koves I et al: Experimental and clinicopathologic studies on the function of the HGF receptor in human colon cancer metastasis. Clin Exp Metast 18: 639-649, 2001. Maehara N, Nagai E, Mizumoto K et al: Gene transduction of NK4, HGF antagonist, inhibits in vitro invasion and in vivo growth of human pancreatic cancer. Clin Exp Metast 19: 417- 426, 2002. Saimura M, Nagai E, Mizumoto K et al: Tumor suppression through angiogenesis inhibition by SUIT-2 pancreatic cancer cells genetically engineered to secrete NK4. Clin Cancer Res 8: 3243-3249, 2002. Fazekas K, Raso E, Zarandi M et al.: Basic HGF-like peptides inhibit generation of liver metastases in murine and human tumor models. Anticancer Res 22:2575-2580, 2002. Fazekas K, Janovics A, Dome B et al: Effect of HGF-like basic hexapeptides on angiogenesis. Microvasc Res 62: 440- 444, 2001. Webb CP, Hose CD, Koochekpour S et al: The geldanamycins are poteint inhibitors of the hepatocyte growth factor/scatter factor-met-urokinase plasminogen activator-plasmin proteolytic network. Cancer Res 60: 342-349, 2000. Timar J, Lapis K, Dudas J et al: Proteoglycans and tumor progression: Janus-faced molecules with contradictory functions in cancer. Semin Cancer Biol 12: 173-186, 2002. Sasisekharan R, Shriver Z, Venkataraman G, Narayanasami U: Roles of heparan-sulphate glycosaminoglycans in cancer. Nature Rev Cancer 2: 521-528, 2002. Timar J, Tovari J, Pogany G et al: The antimetabolite tiazofurin, TR, inhibits glycoconjugate biosynthesis and invasiveness of tumour cells. Eur J Cancer 32A: 152-159, 1996. Jeney A, Timar J, Pogany G, et al: Glycosaminoglycans as novel target in antitumor therapy. Tokai J Exp Clin Med 15:167-177, 1990. Jeney A, Kopper L, Hidvegi E et al: Pharmacobiochemical properties of 5-alkyl-2’ deoxyuridine on tumor growth and glycoconjugate synthesis. Int J Exp Clin Chemother 4: 32-39, 1991. Timar J, Diczhazi Cs, Bartha I et al: Modulation of heparansulphate/ chondroitin-sulphate ratio by glycosaminoglycan biosynthesis inhibitors affects liver metastatic potential of tumor cells. Int J Cancer 62: 755-761, 1995. Pumphrey CY, Theus AM, Li S et al: Neoglycans, carbodiimide- modified glycosamino-glycans: A new class of anticancer agents that inhibit cancer cell proliferation and induce apoptosis. Cancer Res 62: 3722-3728, 2002. Guilford P:E-cadherin downregulation in cancer. Fuel on fire. Mol Med Today 5: 172-177, 1999. Mbalaviele G, Dunstan CR, Sasaki A et al:E-cadherin expression in human breast cancer cells suppresses the development of osteolytic metastasis in an experimental metastasis model. Cancer Res 56: 4063-4070. 1996. Vleminckx KL, Deman JJ, Bruyneel EA, Vandenbossche GM: Enlarged cell associated proteoglycans abolish E-cadherin functionality in invasive tumor cells, Cancer Res 54: 873-877, 1994. Steeg PS: Metastasis suppressors alter the signal transduction of cancer cells. Nature Rev 3: 55-63, 2003. Ranson M: ZD 1839, Iressa): for more than just non-small cell lung cancer. Oncologist 7: 16-24, 2002. Boyer SJ: Small molecule inhibitors of KDR, VEGFR-2, kinase: an overview of structure activity relationships. Curr Top Med Chem 2: 973-1000, 2002. O’Dwyer ME, Druker BJ: The role of the tyrosine kinase inhibitor STI571 in the treatment of cancer. Curr Cancer Drug Targets 1: 49-57, 2001. Favoni RE, de Culpis A: The role of polypeptide growth factors in human carcinoma: New targets for a novel pharmacological approach. Pharmacol Rev 2000, 53: 179-205. Downward J: Targeting ras signaling pathways in cancer therapy. Nature Rev Cancer 3: 11-22, 2002. Aplin AE, Howe A, Alahari SK, Juliano RL: Signal transduction and signal modulation by cell adhesion receptors: The role of integrins, cadherins, immunoglobulin-cell adhesion molecules and selectins. Pharmacol Rev 50:197-263, 1998. Nam JS, Ino Y, Sakamoto M, Hirohashi S: Src family kinase inhibitor PP2 restores the E-cadherin/catenin cell adhesion system in human cancer cells and reduces cancer metastasis. Clin Cancer Res 8: 2430-2430, 2002. Tokumura A, Majima E, Kariya Y et al: Identification of human plasma lysophospholipase D, lysophosphatidic acidproducing enzyme, as autotaxin, a multifunctional phosphodiesterase. J Biol Chem 277: 39436-39442, 2002. Umezu-Goto M, Kishi Y, Taira A et al: Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production. J Cell Biol 158: 227-233, 2002. Shimizu K, Tani M, Watanabe H et al: The autocrine motility factor receptor gene encodes a novel type of seven transmembrane protein. FEBS 456: 295- 300, 1999. Kohn EC, Liotta LA: Invasion and metastasis: New approaches to an old problem. Oncology 7: 47-52, 1993. Otto T, Lummen G, Bex A et al: Tumor cell motility as a novel target in cancer – experimental and clinical results. Onkologie 25: 172-177, 2002. Murata K, Kameyama M, Fukui F et al: Phosphodiesterase type III inhibitor, cilostazol, inhibits colon cancer cell motility. Clin Exp Metast 17: 525-530, 1999. Jansen B, Schlagbauer-Wadl H, Kahr H et al: Novel Ras antagonist blocks human melanoma growth. Proc Natl Acad Sci USA 96: 14019-14024, 1999. Nam JS, Ino Y, Sakamoto M, Hirohashi S: Ras farnesylation inhibitor FTI-277 restores the E-cadherin/catenin cell adhesion system in human cancer cells and reduces cancer metastasis. JPN J Cancer Res 93: 1020-1028, 2002. Zhang B, Prendergast GC, Fenton RG: Farnesyltransferase inhibitors reverse Ras-mediated inhibition of Fas gene expression. Cancer Res 62: 450-458, 2002. Davies H, Bignell GR, Cox C et al: Mutation of the BRAF gene in human cancer. Nature 417: 949-954, 2002. Honn KV, Tang DG, Gao X et al: 12-lipoxygenases and 12(S)- HETE: role in cancer metastasis. Cancer Metast Rev 13: 365- 396, 1994. Raso E, Tovari J, Toth K et al: Ectopic aIIbb3 integrin signaling involves 12-lipoxygenase- and PKC-mediated serine phosphorylation events in melanoma cells. Thromb Haemost 85: 1037-1042, 2001. Pidgeon GP, Kandouz M, Meram A, Honn KV: Mechanisms controlling cell cycle arrest and induction of apoptosis after 12-lipoxygenase inhibition in prostate cancer cells. Cancer Res 62: 2721-2727, 2002. Timar J, Raso E, Fazakas Zs et al: Multiple use of a signal transduction pathway in tumor cell invasion. Anticancer Res 16: 3299-3306, 1996. Timar J, Toth Sz, Tovari J et al: Autocrine motility factor induces cell movement through 12-lipoxygenase-dependent tyrosine phosphorylation and serine dephosphorylation events. Clin Exp Metast 17: 809-816, 1999. Tang DG, Li L, Zhu Z et al. BMD188, a novel hydroxamic acid compound, demonstrates potent anti-prostate cancer effects in vitro and in vivo by inducing apoptosis: requirements for mitochondria, reactive oxygen species and proteases. Pathol Oncol Res 4: 179-190, 1998. Timar J, Raso E, Dome B et al: Expression, subcellular localization and putative function of platelet-type 12-lipoxygenase in human prostate cancer cell lines of different metastatic potential. Int J Cancer 87: 37-43, 2000. Timar J, Dome B, Fazekas K et al: Angiogenesis-dependent diseases and angiogenesis therapy. Pathol Oncol Res 7: 85-95, 2001. Chen WS, Wei SJ, Liu JM et al: Tumor invasiveness and liver metastasis of colon cancer cell correlated with cyclooxygenase- 2, COX-2, expression and inhibited by a COX-2-selective inhibitor, etodolac. Int J Cancer 91: 894-899, 2001. Nagatsuka I, Yamada N, Shimizu S et al: Inhibitory effect of a selective cyclooxygenase-2 inhibitor on liver metastasis of colon cancer. Int J Cancer 100: 515-519, 2002. Bedikian AY, Plager C, Stewart JR et al: Phase II evaluation of bryostatin-1 in metastatic melanoma. Melanoma Res 11: 183- 188, 2001. Zonder JA, Shields AF, Zalupski M et al: A phase II trial of bryostatin 1 in the treatment of metastatic colorectal cancer. Clin Cancer Res 7: 38-42, 2001. Gopalakrishna R, Gundimeda U: Protein kinase C as a molecular target for cancer prevention by selenocompounds. Nutr Cancer 40: 55-63, 2001. Carafoli E: Calcium signaling: A tale for all seasons. PNAS 99: 1115-1122, 2002. Cullen PJ, Lockeyer PJ: Integration of calcium and Ras signaling. Nat Rev Mol Cell Biol 3: 339-348, 2002. Alessandro R, Masiero L, Liotta LA, Kohn EC:The role of calcium in the regulation of invasion and angiogenesis. In Vivo 10: 153-160, 1996. Onoda JM, Nelson KK, Taylor JD, Honn KV: In vivo characterization of combination antitumor chemotherapy with calcium channel blockers and cis-diaminedichloroplatinum, II). Cancer Res 49:2844-2850, 1989. Timar J, Chopra H, Rong X et al: Calcium channel blocker treatment of tumor cells induces alterations in the cytoskeleton, mobility of the integrin aIIbb3 and tumor cell-induced platelet aggregation. J Cancer Res Clin Oncol 118: 425-434, 1992. Kohn EC, Figg WD, Sarosy GA et al: Phase I trial of micronized formulation carboxyamidotriazole in patients with refractory solid tumors: pharmacokinetics, clinical outcome, and comparison of formulations. J Clin Oncol 15: 1985-1993, 1997. Wu Y, Palad AJ, Wasilenko WJ et al: Inhibition of head and neck squamous cell carcinoma growth and invasion by the calcium influx inhibitor carboxyamido-triazole. Clin Cancer Res 3: 1915-1921, 1997. Kohn EC, Reed E, Sarosy GA et al: A phase I trial of carbvoxyamido- triazole and paclitaxel for relapsed solid tumors: potential efficacy of the combination and demonstration of pharmacokinetic interaction. Clin Cancer Res 7: 1600-1609, 2001. Egeblad M, Werb Z: New functions for the matrix metalloproteinases in cancer progression. Nature Rev Cancer 2: 161-172, 2002. Coussens LM, Fingleton B, Matrisian LM: Matrix metalloproteinase inhibitors and cancer: Trials and tribulations. Science 295: 2387-2392, 2002. Jeney A, Kovalszky I, Kopper L et al: Anticollagenase oligodeoxynucleotid, CRIC-2, modifies microinvasiveness and drug sensitivity of malignant cells. Ann Oncol 7: 72, 1996. Otvos L, Sagi J, Sagi Gy et al: Enzymatic hydrolysis and biological activity of oligonucleotides containing 5-substitutesd pyrimidine bases. Nucleosides and Nucleotides 18: 1655- 1666, 1999. Elkin M, Reich R, Nagler A et al: Inhibition of matrix metalloproteinase- 2 expression and bladder carcinoma metastasis by halofuginone. Clin Cancer Res 5: 1982-1988, 1999. Falardeau P, Champagne P, Poyet P et al: Neovastat, a naturally occurring multifunctional antiangiogenic drug, in phase III clinical trials. Semin Oncol 28: 620-625, 2001. Garbisa S, Biggin S, Cavallarin N et al: Tumor invasion: molecular shears blunted by green tea. Nat Med 5: 1216, 1999. Timar F, Botyanszky J, Suli-Vargha H et al: The antiproliferative action of melphalan hexapeptide with collagenase cleavable site. Cancer Chemother Pharmacol 41: 292-298, 1998. Kos J, Werle B, Lah T, Brunner N: Cysteine proteinases and their inhibitors in extracellular fluids: markers for diagnosis and prognosis in cancer. Int J Biol Markers 15: 84-89, 2000. Rochefort H, Liaudet-Coopman E: Cathepsin D in cancer metastasis: a protease and a ligand. APMIS 107: 86-95, 1999. Montaser M, Lalmanach G and Mach L: CA-074, but not its methyl ester CA-074Me, is a selective inhibitor of cathepsin B within living cells. Biol Chem 383: 1305-1308, 2002. Bromme D, Kaleta J: Thiol-dependent cathepsins: pathophysiological implication and recent advances in inhibitor design. Curr Pharm Des 8: 1639-1658, 2002. Del Rosso M, Fibbi G, Pucci M et al: Multiple pathways of cell invasion are regulated by multiple families of serine proteases. Clin Exp Metast 19: 193-207, 2002. Scotton CJ, Wilson JL, Scott K et al: Multiple actions of chemokine CXCL12 on epithelial tumor cells in human ovarian cancer. Cancer Res 62: 5930-5938, 2002. Martin TJ: Manipulating the environment of cancer cells in bone: a novel therapeutic approach. J Clin Invest 110: 1399- 1401, 2002. Mundy GR. Metastasis to bone: causes, consequences and therapeutic opportunities. Nature Rev Cancer 2: 584-593, 2002. Guise TA, Yin JJ, Taylor SD et al: Evidence for a causal role of parathyroid hormone-related protein in the pathogenesis of human breast cancer-mediated osteolysis. J Clin Invest 98: 1544-1549, 1996. Yin JJ, Selander K, Chirgwin JM et al: TGF-bsignaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development. J Clin Invest 103: 197-206, 1999. Lindemann RK, Ballschmieter P, Nordheim A, Dittmer J: Transforming growth factor regulates parathyroid hormonerelated protein expression in MDA-MB-231 breast cancer cells through a novel Smad/Ets synergism. J Biol Chem 276: 46661-46670, 2001. Henderson MA, Danks J, Moseley J et al: Parathyroid hormone- related protein production by breast cancers, improved survival, and reduced bone metastases. J Natl Cancer Inst 93: 234-237, 2001. Neville-Webbe HL, Holen I, Coleman RE: The anti-tumour activity of bishophonates. Cancer Treat Rev 28: 305-319, 2002. Paterson AH: Adjuvant bisphosphonate therapy: the future. Semin Oncol 28: 81-85, 2001. El Abdaimi KE, Dion N, Papavasiliou V et al: The vitamin D analogue EB 1089 prevents skeletal metastasis and prolongs survival time in nude mice transplanted with human breast cancer cells. Cancr Res 60: 4412-4418, 2000. Gallwitz WE, Guise TA, Mundy GR: Guanosine nucleotides inhibit different syndromes of PTHrP excess caused by human cancer in vivo. J Clin Invest 110: 1559-1572, 2002. Griepp P et al: A single subcutaneous dose of an osteoprotegerin, OPG, construct, AMGN-0007, causes a profound and sustained decrease of bone resorption comparable to standard intravenous bisphosphonate in patient with multiple myeloma. Blood 92(Suppl. 1): A3227, 1998. Oyajobi BO, Anderson DM, Traianedes K et al: Therapeutic efficacy of a soluble receptor activator of nuclear factor kappaB- IgG Fc fusion protein in suppressing bone resorption and hypercalcemia in a model of humoral hypercalcemia of malignancy. Cancer Res 61: 2572-2578, 2001. Kakkar AK and Williamson RCN: Antithrombotic therapy in cancer. BMJ 318: 1571-1572, 1999. Letai A and Kuter DJ: Cancer, coagulation, and anticoagulation. Oncologist 4: 443-449, 1999. Loreto MF, De Martinis M, Corsi MP et al: Coagulation and cancer: Implications for diagnosis and management. Pathol Oncol Res 6: 301-312, 2000. Nash GF, Walsh DC, Kakkar AK: The role of the coagulation system in tumour angiogenesis. Lancet Oncol 2: 608-613, 2001. Hejna M, Raderer M, Zielinsky ChC: Inhibition of metastases by anticoagulants J Natl Cancer Inst 91: 22-36, 1999. 115. Cosgrove RH, Zacharski LR, Racine E, Andersen JC: Improved cancer mortality with low-molecular weight heparin treatment: a review of the evidence. Semin Thromb Haemost 28: 79-87, 2002. 117. von Tempelhoff GF, Harenberg J, Niemann F et al: Effect of low molecular weight heparin, Certoparin, versus unfractionated heparin on cancer survival following breast and pelvic cancer surgery: A prospective randomized double-blind trial. Int J Oncol 16: 815-824, 2000. 118. Borsing L, Wong R, Feramisco J et al: Heparin and cancer revisited: Mechanistic connections involving platelets, P-selectin, carcinoma mucins, and tumor metastasis. PNAS 98: 3352-3357, 2001. 119. Smorenburg SM, von Noorden CJF: The complex effects of heparins on cancer progression and metastasis in experimental studies. Pharmacol Rev 53: 93-105, 2001. 120. Varki A, Varki NM: P-selectin, carcinoma metastasis and heparin: novel mechanistic connections with therapeutic implications. Braz J Med Biol Res 34: 711-717, 2001. 121. De Larco JE, Wuertz BRK, Manivel JC et al: Dissemination and growth of cancer cells in metastatic sites. Nature Rev Cancer 2: 563-572, 2002. 122. Trikha M, Nakada MT: Platelets and cancer: Implications for antiangiogenic therapy. Semin Thromb Haemost 28: 39-44, 2002. 123. Cohen SA, Trikha M, Mascelli MA: Potential future clinical applications for the GPIIb/IIIa antagonist, abciximab in thrombosis, vascular and oncological indications. Pathol Oncol Res 6: 163-174, 2000. 124. Steinert BW, Tang DG, Grossi IM et al: Studies on the role of platelet eicosanoid metabolism and integrin aIIbb3 in tumorcell- induced platelet aggregation. Int J Cancer 54: 92-101, 1993. 125. Schneider MR, Tang DG, Schirner M, Honn KV: Prostacyclin and its analogues: antimetastatic effects and mechanism of action. Cancer Metast Rev 13: 349-364, 1994. 126. Jeney A, Lapis K, Kopper L et al: Tumor progression and prostaglandins. In: Hemostasis and Cancer, Ed: Muszbek L, CRC Press, 1987, pp. 207-214. 127. Moller P, Koretz K, Leithauser F et al: Expression of APO-1, CD95), a member of the NGF/TNF receptor superfamily, in normal and neoplastic colon epithelium. Int J Cancer 57: 371- 377, 1994. 128. Muller M, Wilder S, Bannasch D et al: p53 activates the CD95, APO-1/Fas, gene in response to DNA damage by anticancer drugs. J Exp Med 188: 2033-2045, 1998. 129. Lowin B, Hahne M, Mattmann C, and Tschopp J: Cytolytic Tcell cytotoxicity is mediated through perforin and Fas lytic pathways. Nature 370: 650-652, 1994. 130. Harwood FG, Kasibhatla S, Petak I et al: Regulation of FasL by NF-kappaB and AP-1 in Fas-dependent thymineless death of human colon carcinoma cells. J Biol Chem 275: 10023- 10029, 2000. 131. Petak I, Tillman DM, and Houghton, JA: p53 dependence of Fas induction and acute apoptosis in response to 5-fluorouracil- leucovorin in human colon carcinoma cell lines. Clin Cancer Res 6: 4432-4441, 2000. 132. Tillman DM, Petak I, and Houghton JA: A Fas-dependent component in 5-fluorouracil/leucovorin-induced cytotoxicity in colon carcinoma cells. Clin Cancer Res 5: 425-430, 1999. 133. Schwartzberg LS, Petak I, Stewart C et al: Modulation of the Fas signaling pathway by IFN-gamma in therapy of colon cancer: phase I trial and correlative studies of IFN-gamma, 5-fluorouracil, and leucovorin. Clin Cancer Res 8: 2488- 2498, 2002. 134. Kettelhut IC, Fiers W, and Goldberg AL: The toxic effects of tumor necrosis factor in vivo and their prevention by cyclooxygenase inhibitors. Proc Natl Acad Sci USA 84: 4273- 4277, 1987. 135. Ogasawara J, Watanabe-Fukunaga R, Adachi M et al: Lethal effect of the anti-Fas antibody in mice. Nature, 364: 806-809, 1993. 136. Ashkenazi A, Pai RC, Fong S et al: Safety and antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest 104: 155-162, 1999. 137. Marsters SA, Pitti RA, Sheridan JP, and Ashkenazi A: Control of apoptosis signaling by Apo2 ligand. Recent Prog Horm Res 54:225-234, 1999. 138. Pollack IF, Erff M, and Ashkenazi, A: Direct stimulation of apoptotic signaling by soluble apo2l/tumor necrosis factorrelated apoptosis-inducing ligand leads to selective killing of glioma cells. Clin Cancer Res 7: 1362-1369, 2001. 139. Keane MM, Ettenberg SA, Nau MM et al: Chemotherapy augments TRAIL-induced apoptosis in breast cell lines. Cancer Res 59: 734-741, 1999. 140. Jo M, Kim TH, Seol DW et al: Apoptosis induced in normal human hepatocytes by tumor necrosis factor-related apoptosis- inducing ligand. Nat Med 6: 564-567, 2000 141. Lawrence D, Shahrokh Z, Marsters S et al: Differential hepatocyte toxicity of recombinant Apo2L/TRAIL versions. Nat Med 7: 383-385, 2001. 142. Tai YT, Strobel T, Kufe D, and Cannistra SA: In vivo cytotoxicity of ovarian cancer cells through tumor-selective expression of the BAX gene. Cancer Res 59: 2121-2126, 1999. 143. Teitz T, Wei T, Valentine MB et al: Caspase 8 is deleted or silenced preferentially in childhood neuroblastomas with amplification of MYCN. Nat Med 6: 529-535, 2000. 144. Petak I, Douglas L, Tillman DM et al: Pediatric rhabdomyosarcoma cell lines are resistant to Fas-induced apoptosis and highly sensitive to TRAIL-induced apoptosis. Clin Cancer Res 6: 4119-4127, 2000. 145. Mazumder S. and Almasan A: Is caspase-8 a neuroendocrine lung tumor suppressor? Cancer Biol Ther 1: 70-71, 2002. 146. Shivapurkar N, Toyooka S, Eby MT et al: Differential inactivation of caspase-8 in lung cancers. Cancer Biol Ther 1: 65- 69, 2002. 147. Fulda S, Kufer MU, Meyer E et al: Sensitization for death receptor- or drug-induced apoptosis by re-expression of caspase- 8 through demethylation or gene transfer. Oncogene 20: 5865-5877, 2001. 148. Yanagisawa J, Takahashi M, Kanki H et al: The molecular interaction of Fas and FAP-1. A tripeptide blocker of human Fas interaction with FAP-1 promotes Fas-induced apoptosis. J Biol Chem 272: 8539-8545, 1997. 149. Li Y, Kanki H, Hachiya T et al: Negative regulation of Fasmediated apoptosis by FAP-1 in human cancer cells. Int J Cancer 87: 473-479, 2000. 150. Tschopp J, Irmler M, and Thome M: Inhibition of fas death signals by FLIPs. Curr Opin Immunol 10: 552-558, 1998. 151. Que FG, Phan VA, Phan VH et al: Cholangiocarcinomas express Fas ligand and disable the Fas receptor. Hepatology 30: 1398-1404, 1999. 152. Banerjee D: Genasense, Genta Inc). Curr Opin Invest Drugs 2: 574-580, 2001. 153. Olie RA, and Zangemeister-Wittke U: Targeting tumor cell resistance to apoptosis induction with antisense oligonucleotides: progress and therapeutic potential. Drug Resist Updat 4: 9-15, 2001. 154. Yamamoto T and Tanigawa N: The role of survivin as a new target of diagnosis and treatment in human cancer. Med Electron Microsc 34: 207-212, 2001. 155. Khwaja A: Akt is more than just a Bad kinase. Nature 401: 33- 34, 1999. 156. Capdeville R, Buchdunger E, Zimmermann J et al: Glivec, STI571, imatinib), a rationally developed, targeted anticancer drug. Perspectives on the development of a molecularly targeted agent. Nat Rev Drug Discov 1: 493-502, 2002. 157. Fox SB, Gasparini G and Harris AL: Angiogenesis: pathological, prognostic, and growth-factor pathways and their link to tiral design and anticancer drugs. Lancer Oncol 2: 278-289, 2001 158. Stopeck A, Sheldon M, Vahedian M et al: Results of a phase I dose-escalating study of the antiangiogenic agent, SU5416, in patients with advanced malignancies. Clin Cancer Res 8: 2798-1805, 2002. 159. Holmgren L, Reilly MSO, Folkman J: Dormancy of micrometastases: Balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1: 149-153, 1995. 160. Liao F, Doddy JF, Overholser J et al: Selective targeting of angiogenic tumor vasculature by vascular endothelial-cadherin antibody inhibits tumor growth without affecting vascular permeability. Cancer Res 62: 2567-2575, 2002. 161. Thorpe PE, Ran S: Tumor infarction by targeting tissue factor to tumor vasculature. Cancer J 6, Suppl 3): S237-244, 2000. 162. Weenendaal LM, Jin H, Ran S et al: In vitro and in vivo studies of a VEGF121/rGelonin chimeric fusion toxin targeting the neovasculature of solid tumors. PNAS 99:7866-7871, 2002. 163. Burke PA, DeNardo SJ, Miers LA et al: Cilengitide targeting of alpha(v)beta(3, integrin receptor synergizes with radioimmunotherapy to increase efficacy and apoptosis in breast cancer xenografts. Cancer Res 62: 4263-4272, 2002. 164. Schraa AJ, Kok RJ, Moorlag HE et al: Targeting of RGDmodified proteins to tumor vasculature: pharmacokinetic and cellular distribution study. Int J Cancer 102: 469-475, 2002. 165. Semenza GL: HIF-1 and mechanisms of hypoxia sensing. Curr Opin Cell Biol 13: 167-171, 2001. 166. Harris AL: Hypoxia – A key regulatory factor in tumour growth. Nature Rev Cancer 2: 38-47, 2001. 167. Fyles AW, Milosevic M, Wong R et al: Oxygenation predicts radiation response and survival in patients with cervix cancer. Radiother Oncol 48: 149-156, 1998. 168. Kung AL, Wang S, Klco JM et al: Suppression of tumor growth through disruption of hypoxia-inducible transcription. Nat Med 6: 1335-1340, 2000. 169. Dunst J: Hemoglobin level and anemia in radiation oncology: prognostic impact and therapeutic implications. Semin Oncol 27: 4-8, 2000. 170. Girinski T, Pejovic-Lenfant MH, Bourhis J et al: Prognostic value of hemoglobin concentrations and blood transfusions in advanced carcinoma of the cervix treated by radiation therapy: results of a retrospective study of 386 patients. Int J Radiat Oncol Biol Phys 16: 37-42, 1989. 171. Grogan M, Thomas GM, Melamed I et al: The importance of hemoglobin levels during radiotherapy for carcinoma of the cervix. Cancer 86: 1528-1536, 1999. 172. Obermair A, Handisurya A, Kaider A et al: The relationship of pretreatment serum hemoglobin level to the survival of epithelial ovarian carcinoma patients. Cancer 83: 726-731, 1998. 173. Clark O, Adams JR, Bennett CL, Djulbegovic B: Erythropoetin, uncertainty principle and cancer related anaemia. BMC Cancer 2: 23-31, 2002. 174. hews O, Koenig R, Kelleher DK et al: Enhanced radiosensitivity in experimental tumours following erythropoietin treatment of chemotherapy-induced anaemia. Br J Cancer 78: 752-756, 1998. 175. Tisdale MJ: Cachexia in cancer patients. Nature Rev Cancer 2: 862-871, 2002. 176. Cabrero A, Alegret M, Sanchez RM et al: Down-regulation of uncoupling protein-3 and H-2 by thiazolidinediones in C2C12 myotubes. FEBS Lett 484: 37-42, 2000. 177. Hasselgren PO, Fischer JE: Muscle cachexia: current concepts of intracellular mechanisms and molecular regulation. Ann Surg 232: 9-17, 2001. 178. Guttridge DC, Mayo MV, Madrid LV et al: NF-kB-induced loss of MyoD messenger RNA: possible role in muscle decay and cachexia. Science 289: 2363-2366, 2000. 179. Zimmers TA, Davies MV, Koniaris LG et al: Induction of cachexia in mice by systemically administered myostatin. Science 296: 1486-1488, 2002. 180. Lorite MJ, Smith HJ, Arnold JA et al: Activation of ATP-ubiquitin- dependent proteolysis in skeletal muscle in vivo and murine myoblasts in vitro by a proteolysis-inducing factor, PIF). Br J Cancer 85: 297-302, 2001. 181. Wigmore SJ, Barber MD, Ross JA et al: Effect of oral eicosapentaenoic acid on weight loss in patients with pancreatic cancer. Nutr Cancer 36: 177-184, 2000. 182. Smith HJ, Lorite MJ, Tisdale MJ: Effect of a cancer cachectic factor on protein synthesis/degradation in murine C2C12 myoblasts: modulation by eicosapentaenoic acid. Cancer Res 59: 5507-5513, 1999. 183. Tan C and Waldmann TA: Proteasome inhibitor PS-341, a potential therapeutic agent for adult T-cell leukaemia. Cancer Res 62: 1083-1086, 2002. 184. Russell ST, Hirai K and Tisdale MJ: Role of b-adrenergic receptors in the action of a tumour lipid mobilizing factor. Br J Cancer 86: 424-428, 2002. 185. Marincola FM, Jaffee EM, Hicklin DJ, et al: Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Adv Immunol 74:181-273, 2000. 186. Brandau S, Bohle A: Therapy of bladder cancer with BCG: the mechanism behind a successful immunotherapy. Mod Asp Immunobiol 2:37-41, 2001. 187. Nathan PD, Eisen TG: The biological treatment of renal-cell carcinoma and melanoma. Lancet Oncol 3:89-96, 2002. 188. Rosenberg SA, Yang JC, White DE, et al: Durability of complete responses in patients with metastatic cancer treated with high-dose interleukin-2. Ann Surg 228:307-319, 1998. 189. Panelli MC, Wang E, Phan G, et al: Gene-expression profiling of the response of peripheral blood mononuclear cells and melanoma metastases to systemic IL-2 administration. Genome Biol 3:research0035.1-0035.17. 190. Berd D: Autologous, hapten-modified vaccine as a treatment for human cancers. Vaccine 19:2565-2570, 2001. 191. Soiffer R, Lynch T, Mihm M, et al: Vaccination with irradiated autologous melanoma cells engineered to secrete human granulocyte- macrophage colony-stimulating factor generates potent antitumor immunity in patients with metastatic melanoma. Proc Natl Acad Sci USA 95:13141-13146, 1998. 192. Nelson WG, Simons JW, Mikhak B, et al: Cancer cells engineered to secrete granulocyte-macrophage colony-stimulating factor using ex vivo gene transfer as vaccines for the treatment of genitourinary malignancies. Cancer Chemother Pharmacol 46(Suppl):S67-S72, 2000. 193. Chan AD, Morton DL: Active immunotherapy with allogeneic tumor cell vaccines: present status. Semin Oncol 25:611- 622, 1998. 194. Rosenberg SA, Yang JC, Schwartzentruber DJ, et al: Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat Med 4:321-327, 1998. 195. Marchand M, van Baren N, Weynants P, et al: Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1. Int J Cancer 80:219-230, 1999. 196. Jager E, Ringhoffer M, Altmannsberger M, et al: Immunoselection in vivo: independent loss of MHC class I and melanocyte differentiation antigen expression in metastatic melanoma. Int J Cancer 71:142-147, 1997. 197. Riker A, Cormier J, Panelli M, et al: Immune selection after antigen-specific immunotherapy of melanoma. Surgery 126:112-120, 1999. 198. Rosenberg SA, Zhai Y, Yang JC, et al: Immunizing patients with metastatic melanoma using recombinant adenoviruses encoding MART-1 or gp100 melanoma antigens. J Natl Cancer Inst 90:1894-1900, 1998. 199. Marshall JL, Hawkins MJ, Tsang KY, et al: Phase I study in cancer patients of a replication-defective recombinant vaccine that expresses human carcinoembryonic antigen. J Clin Oncol 17:332-337, 1999. 200. Nestle FO, Alijagic S, Gilliet M, et al: Vaccination of melanoma patients with peptide- or tumor pulsed dendritic cells. Nat Med 4:328-332, 1998. 201. Thurner B, Haendle I, Roder C, et al: Vaccination with Mage- 3A1 peptide-pulsed mature monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J Exp Med 190:1669-1678, 1999. 202. Holtl L, Zelle-Rieser C, Gander H, et al: Immunotherapy of metastatic renal cell carcinoma with tumor lysate-pulsed autologous dendritic cells. Clin Cancer Res 8:3369-3376, 2002. 203. Jefford M, Maraskovsky E, Cebon J, et al: The use of dendritic cells in cancer therapy. Lancet Oncol 2:343-353, 2001. 204. Murphy GP, Tjoa BA, Simmons SJ, et al: Infusion of dendritic cells pulsed with HLA-A2-specific prostate-specific membrane antigen peptides: a phase II prostate cancer vaccine trial involving patients with hormone-refractory metastatic disease. Prostate 38:73-78, 1999. 205. Kugler A, Stuhler G, Walden P, et al: Regression of human metastatic renal cell carcinoma after vaccination with tumor cell-dendritic cell hybrids. Nat Med 6:332-336, 2000. 206. Rosenberg SA, Yannelli JR, Yang JC, et al: Treatment of patients with metastatic melanoma with autologous tumorinfiltrating lymphocytes and interleukin 2. J Natl Cancer Inst 86:1159-1166, 1994. 207. Figlin RA, Pierce WC, Kaboo R, et al: Treatment of metastatic renal cell carcinoma with nephrectomy, interleukin-2 and cytokine-primed or CD8+ selected tumor-infiltrating lymphocytes from primary tumor. J Urol 158:740-745, 1997. 208. Figlin RA, Thompson JA, Bukowski RM, et al: Multicenter, randomized, phase III trial of CD8+ tumor-infiltrating lymphocytes in combination with recombinant interleukin-2 in metastatic renal cell carcinoma. J Clin Oncol 17:2521-2529, 1999. 209. Dudley ME, Wunderlich JR, Robbins PF, et al: Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 298:850-854, 2002. 210. Yee C, Thompson JA, Byrd D, et al: Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: In vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci USA 99:16168-16173, 2002. 211. Lee K-H, Wang E, Nielsen M-B, et al: Increased vaccine-specific T cell frequency after peptide-based vaccination correlates with increased susceptibility to in vitro stimulation but does not lead to tumor regression. J Immunol 163:6292-6300, 1999. 212. Clay TM, Hobeika AC, Mosca PJ, et al: Assays for monitoring cellular immune responses to active immunotherapy of cancer. Clin Cancer Res 7:1127-1135, 2001. 213. Keilholz U, Weber J, Finke JH, et al: Immunologic monitoring of cancer vaccine therapy: results of a workshop sponsored by the Society for Biological Therapy. J Immunother 25:97-138, 2002. 214. Lapis K, Timar J, Papay J et al: Experimental metastasis inhibition by the pretreatment of the host. Arch Geschwulstforsch 60: 97-102, 1990. 215. Gately S and Kerbel R: Antiangiogenic scheduling of lower dose cancer chemotherapy. Cancer J 7: 427-436, 2001. 216. Fidler IJ, Ellis LM: Chemotherapeutic drugs: more really is not better. Nat Med 6: 500-502, 2000. 217. Browden T, Butterfield CE, Kraling BM et al: Antiangiogenic scheduling of chemotherapy improves efficacy against experimental drug-resistant cancer. Cancer Res 60: 1878-1886, 2000. 218. Gasparini G: Metronomic scheduling: the future of chemotherapy? Lancet Oncol 2: 733-740, 2001. 219. Sun LM, Yeh SA, Wang CJ et al: Postoperative radiation therapy for medulloblastoma high recurrence rate in the subfrontal region. J Neurooncol 58: 77-85, 2002. 220. Crane CH, Macdonald KO, Vauthey JN et al: Limitations of conventional doses of chemoradiation for unresectable biliary cancer. Int J Radiat Oncol Biol Phys 53: 969-974, 2002. 221. Bhattathiri VN: Cumulative interfraction interval analysis of influence of time and interruptions on radiotherapy result in oral cancers. Int J Radiat Oncol Biol Phys 52: 1251-1256, 2002. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2003 VL 9 IS 1 BP 49 EP 72 PG 24 ER PT J AU Nishimura, Y Itoh, K Yoshioka, K Tokuda, K Himeno, M AF Nishimura, Yukio Itoh, Kazuyuki Yoshioka, Kiyoko Tokuda, Kazuo Himeno, Masaru TI Overexpression of ROCK in Human Breast Cancer Cells: Evidence that ROCK Activity Mediates Intracellular Membrane Traffic of Lysosomes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ROCK; RhoA; Cathepsin D; LIMPII; lysosomes; endosomes; cytoskeleton ID ROCK; RhoA; Cathepsin D; LIMPII; lysosomes; endosomes; cytoskeleton AB Small GTPase Rho and its downstream effectors, ROCK family of Rho-associated serine-threonine kinases, are thought to participate in cell morphology, motility, and tumor progression through regulating the rearrangement of actin cytoskeleton. Here we present evidence that transfection of human breast cancer cells with cDNA encoding a dominant active mutant of ROCK causes dispersal of lysosomal vesicles throughout the cytoplasm without perturbing the machinery of the endocytic pathway. The intracellular distribution of lysosomes and endocytosed transferrin, an early endosomal marker, were further assessed by confocal immunofluorescence microscopy. In the active ROCK transfected cells the lysosomal proteins, cathepsin D, LIMPII, and LAMP1, were found throughout the cytoplasm in dispersed small vesicles, which were accessible to the endocytosed Texas Red-labeled transferrin. 3D-image analysis of lysosomal distribution in the active ROCKtransfectants revealed abundant punctate signals in the peripheral region of the basal plasma membrane. Cells expressing vector alone did not exhibit these alterations. Wortmannin, a phosphatidylinositol 3-kinase inhibitor, induced LIMPII-positive/ transferrin negative large vacuoles in the perinuclear region, and disappearence of the dispersed small vesicular structures. To our knowledge, this is the first evidence that increasing ROCK expression contributes to selective cellular dispersion of lysosomes in invasive breast cancer cells. C1 [Nishimura, Yukio] Kyushu University, Faculty of Pharmaceutical Sciences, 3-1-1 Maidashi, Higashi-ku, 812-8582 Fukuoka, Japan. [Itoh, Kazuyuki] Osaka Medical Center for Cancer and Cardiovascular Diseases, Research InstituteOsaka, Japan. [Yoshioka, Kiyoko] Osaka Medical Center for Cancer and Cardiovascular Diseases, Research InstituteOsaka, Japan. [Tokuda, Kazuo] Kyushu University, Faculty of Pharmaceutical Sciences, 3-1-1 Maidashi, Higashi-ku, 812-8582 Fukuoka, Japan. [Himeno, Masaru] Kyushu University, Faculty of Pharmaceutical Sciences, 3-1-1 Maidashi, Higashi-ku, 812-8582 Fukuoka, Japan. RP Nishimura, Y (reprint author), Kyushu University, Faculty of Pharmaceutical Sciences, 812-8582 Fukuoka, Japan. EM nishimur@bioc.phar.kyushu-u.ac.jp CR Paterson HF, Self AJ, Garrett MD et al: Microinjection of recombinant p21rho induces rapid changes in cell morphology. J Cell Biol 111: 1001-1007, 1990. Perona R, Esteve P, Jimene, B et al: Tumorigenic activity of rho genes from Aphysia california. Oncogene 8: 1285-1292, 1993. Kishi K, Sasaki T, Kuroda S et al: Regulation of cytoplasmic division of Xenopus Embryo by rhop21 and its inhibitory GDP/GTP exchange protein, rhoGDI). J Cell Biol 120: 1187-1195,1993. Takaishi K, Kikuchi A, Kuroda S et al: Involvement of rhop21 and its inhibitory GDP/GTP exchange protein, rhoGDI, in cell motility. Mol Cell Biol 13: 72-79, 1993 Hall, A. Small GTP-binding proteins and the regulation of the actin cytoskeleton. Annu Rev Cell Biol 10: 31-54, 1994 Prendergast GC, Khosravi-Far R, Solski PA, et al: Critical role of Rho in cell transformation by oncogenic Ras. Oncogene 10: 2289- 2296, 1995 Fritz G, Just I, Kaina B: Rho GTPases are over-expressed in human tumors. Int J Cancer 81: 682-687, 1999 Yoshioka K, Nakamori S, Itoh K: Overexpression of small GTPbinding protein RhoA promotes invasion of tumor cells. Cancer Res 59: 2004-2010, 1999 Yoshioka K, Matsumura F, Akedo H, Itoh K: Small GTP-binding protein Rho stimulates the actomyosin system, leading to invasion of tumor cells. J Biol Chem 273: 5146-5154, 1998. Leung T, Manser E, Tan L, Lim L: A novel serine/threonine kinase binding the Ras-related RhoA GTPase which translocates the kinase to the peripheral membranes. J Biol Chem 270: 29051- 29054, 1995. Itoh K, Yoshioka K, Akedo H et al: An essential part for Rho-associated kinase in the transcellular invasion of tumor cells. Nature Medicine 5: 221-225, 1999 Sloane BF, Moin K, Sameni M et al: Membrane association of cathepsin B can be induced by transfection of human breast epithelial cells with c-Ha-Ras oncogene. J Cell Sci 107: 373-384, 1994 Sameni M, Elliott E, Ziegler G et al: Cathepsin B and D are localized at the surface of human breast cancer cells. Pathol Oncol Res 1: 43-53, 1995 Taniguchi S, Nishimura Y, Takahashi, T et al: Augmented excretion of procathepsin L of a fos-transferred highly metastatic cell line. Biochem Biophys Res Commun 168: 520-526, 1990 Kageshita T, Yoshii A, Kimura T et al: Biochemical and immunohistochemical analysis of cathepsins B, H, L and D in human melanocytic tumours. Arch Dermatol Res 287: 266-272, 1995 Albini A, Graf J, Kitten GT et al: 17ß-Estradiol regulates and v- Ha-ras transfection constitutively enhances MCF7 breast cancer cell interactions with basement membrane. Proc Natl Acad Sci US 83: 8182-8186, 1986 Ichikawa T, Kyprianou N, Isaacs JT: Genetic instability and the acquisition of metastatic ability by rat mammary cancer cells following v-H-ras oncogene transfection. Cancer Res 50: 6349- 6357, 1990. Nishimura Y, Sameni M, Sloane BF: Malignant transformation alters intracellular trafficking of lysosomal cathepsin D in human breast epithelial cells. Pathol Oncol Res 4: 283-296, 1998 Nishimura Y, Itoh K, Yoshioka,K et al: Small guanosine triphosphatase Rho/Rho-associated kinase as a novel regulator of intracellular redistribution of lysosomes in invasive tumor cells. Cell Tissue Res 342: 341-351, 2000. Uehata M, Ishizaki T, Satoh H et al: A key role for p160ROCKmediated Ca++sensitization of smooth muscle in hypertension. Nature 389: 990-994, 1997. Matsui T, Amano M, Yamamoto T et al: Rho-associated kinase, a novel serine threonine kinase, as a putative target for the small GTP binding protein Rho. EMBO J 15: 2208-2216, 1996 Nakagawa O, Fujisawa K, Ishizaki et al: ROCK-I and ROCK-II; two isoforms of Rho-associated coiled-coil forming protein serine/ threonine kinase in mice. FEBS Lett 392: 189-193, 1996 Amano M, Chihara K, Kimur, Y et al: Formation of actin stress fibers and focal adhesions enhanced by Rho-kinase. Science 275: 1308-1311,1997. Ishizaki T, Naito M, Fujisaw, K et al: p160ROCK, a Rho-associated coiled-coil forming protein kinase, works downstream of Rho and induces focal adhesion. FEBS Lett 404: 118-124, 1997 Nishimura Y, Itoh K, Yoshioka K, et al: A role for small GTPase RhoA in regulating intracellular membrane traffic of lysosomes in invasive rat hepatoma cells. Histochem J 34: 189-213, 2002. Lippincott-Schwart J, Fambrough DM: Lysosomal membrane dynamics: structure and interorganellar movement of a major lysosomal membrane glycoprotein. J Cell Biol 102: 1593-1605, 1986. Griffiths G, Hoflack B, Simon, K et al: The mannose 6-phosphate receptor and the biogenesis of lysosomes. Cell 52: 329-341, 1998 Kornfeld S, Mellman I: The biogenesis of lysosomes. Ann Rev Cell Biol 5: 483-525, 1989. Okazaki I, Himeno M, Ezaki J et al: Purification and characterization of an 85 kDa sialoglycoprotein in rat liver. J Biochem 111: 763-769, 1992. Sandoval IV, Arredondo JJ, Alcalde J et al: The residues Leu(Ile)475-Ile(Leu)476, contained in the extended carboxyl cytoplasmic tail, are critical for targeting of the resident lysosomal membrane protein LIMPII to lysosomes. J Biol Chem 269: 6622- 6631, 1994 Tabuchi N, Akasaki K, Tsuji H: Two acidic amino acid residues, Asp(470, and Glu(471), contained in the carboxyl cytoplasmic tail of a major lysosomal membrane protein, LGP85/LIMPII, are important for its accumulation in secondary lysosomes. Biochem Biophys Res Commun 270: 557-563, 2000. Soule HD, Vazguez J, Long et al.:, A human cell line from a pleural effusion derived from a breast carcinoma. J Natl Cancer Inst 51: 1409-1416, 1973. Nishimura Y, Higaki M, Kato K: Identification of a precursor form of cathepsin D in microsomal lumen: characterization of enzymatic activation and proteolytic processing in vitro. Biochem Biophys Res Commun 148: 335-343, 1987 Nishimura Y, Kawabata T, Kato K: Identification of latent procathepsins B and L in microsomal lumen: characterization of enzymatic activation and proteolytic processing in vitro. Arch Biochem Biophys 261: 64-71, 1988 Matteoni R, Kreis TE: Translocation and clustering of endosomes and lysosomes depends on microtubules. J Cell Biol 105: 1253-1265, 1987 Mu FT, Callaghan JM, Steele-Mortimer O et al: EEA1, an early endosome-associated protein. EEA1 is a conserved alpha-helical peripheral membrane protein flanked by cysteine “fingers” and contains a calmodulin-binding IQ motif. J Biol Chem 270: 13503-13511, 1995 Stack JH, Emr SD: Genetic and biochemical studies of protein sorting to the yeast vacuole. Curr Biol 5: 641-646,1993. Brown WJ Emr SD, Plutner H, Balch WE: Role for phosphatidylinositol 3-kinase in the sorting and transport of newly synthesized lysosomal enzymes in mammalian cells. J Cell Biol 130: 797-805, 1995 Davidson H: Wortmannin causes mis-targeting of procathepsin D. Evidence for the involvement of a phosphatidylinositol 3- kinase in vesicular transport to lysosomes. J Cell Biol 130: 797- 805, 1995 Reaves B, Bright NA, Mullock B, Luzio JP: The effect of wortmannin on the localization of lysosomal type I integral membrane glycoproteins suggests a role for phosphoinositide 3- kinase activity in regulating membrane traffic late in the endocytic pathway. J Cell Sci 109: 749-762, 1996 Pfeffer SR: GTP-binding proteins in intracellular transport. Trends Cell Biol 2: 41-46, 1992 Zerial M, Stenmark H: Rab GTPases in vesicular transport. Curr Opin Cell Biol 5: 613-620, 1993 Chavrier P, Goud B: The role of ARF and Rab GTPases in membrane transport. Curr Opin Cell Biol, 11: 466-475, 1999 Pfeffer SR: Transport-vesicle targeting: tethers before SNAREs. Nature Cell Biol 1: E17-E22, 1999. Bucci C, Thomsen P, Nicoziani P et al.: Rab7: A key to lysosome biogenesis. Mol Biol Cell 11: 467-480, 2000 Press B, Feng Y, Hoflack B, Wandinger-Nes A: Mutant Rab7 causes the accumulation of cathepsin D and cation-independent mannose 6-phosphate receptor in an early endocytic compartment. J Cell Biol 140: 1075-1089, 1998. Hirose M, Ishizaki T, Watanabe N et al: Molecular dissection of the Rho-associated kinase, p160ROCK)-regulated neurite remodeling in neuroblastoma N1E-115 cells. J Cell Biol 141: 1625-1636, 1998 Hollenbeck PJ, Swanson JA: Radial extension of macrophage tubular lysosomes supported by kinesin. Nature 346: 864-866, 1990 Nakata T, Hirokawa N: Point mutation of adenosine triphosphate- binding motif generated rigor kinesin that selectively blocks anterograde lysosome membrane transport. J Cell Biol 131: 1039-1053, 1995 Hotta K, Tanak K, Mino A, et al: Interaction of the Rho family small G proteins with kinectin, an anchoring protein of kinesin motor. Biochem Biophys Res Commun 225: 69-74, 1996 Vignal E, Blangy A, Martin M, et al: Kinectin is a key effector of RhoG microtubule-dependent cellular activity. Mol Cell Biol 21: 8022-8034, 2001 Timar J, Tang D, Bazaz R, et al.: PKC mediates 12-(S)-HETEinduced cytoskeletal rearrangement in B16a melanoma cells. Cell Motil Cytoskel 26: 49-65, 1993 Timar J, Bazaz R, Kimler V, et al: Immunomorphological characterization and effects of 12-(S)-HETE on a dynamic intracellular pool of the aIIbß3-integrin in melanoma cells. J Cell Sci 108: 2175-2186, 1995 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2003 VL 9 IS 2 BP 83 EP 95 PG 13 ER PT J AU Raso, E Paku, S Kopper, L Timar, J AF Raso, Erzsebet Paku, Sandor Kopper, Laszlo Timar, Jozsef TI Trace Elements Improve Survival of DTIC-Treated Mice with Overt Liver Metastases of Lewis Lung Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE 3LL-HH tumor; mice; trace element mixture; liver metastasis; survival; 5-FU; DTIC ID 3LL-HH tumor; mice; trace element mixture; liver metastasis; survival; 5-FU; DTIC AB Trace elements have been previously shown to have specific antimetastatic effects in a mouse 3LLHH liver metastasis model. Here we have analyzed the effect on the survival of animals with liver metastases. Trace elements administered per os at 500-5000 mg/kg/day did not affect the survival of animals with liver metastases. However, when trace element treatment was combined with dacarbazine (DTIC) administration, the survival of animals was significantly improved (55%). This effect was specific for DTIC since trace elements did not influence the effect of 5-fluorouracil on survival in this liver metastasis model. These data and those found in the literature all suggest that trace elements can specifically modulate the antitumoral/antimetastatic effects of chemotherapeutic agents. C1 [Raso, Erzsebet] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy 7-9, H-1122 Budapest, Hungary. [Paku, Sandor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy 7-9, H-1122 Budapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, H-1122 Budapest, Hungary. CR Gal K, Bertok L: The effect of X-radiation on reticuloendothelial system and its treatment with radiotoxified endotoxin and trace elements in rats. Acta Microbiol Immunol Hung 41: 457-463, 1994 Falus A, Beres J Jr: A trace element preparation containing zinc increases the production of interleukon-6 in human monocytes and glial cells. Trace Element Res 51: 293-301, 1996 Grinevich JA, Beres J. Jr, Bendyug GD: A trace element preparation increases antitumor activity in mice. Pathol Oncol Res 3: 34-37, 1997 Timar J, Raso E, Paku S , Kopper L: Oral administration of a trace element preparation and zinc inhibit liver metastasis of 3LL-HH murine tumor cells. Int J Molec Med 2: 105-108, 1998 Pal K, Kopper L, Lapis K: Increased metastatic capacity of Lewis lung tumor cells by in vivo selection procedure. Invasion Metast 3: 174-182, 1983 Lapis K, Papay J, Paku S et al: Effect of Lentinan on the metastasis of Lewis lung carcinoma. Int J ImmunotherV: 195-201, 1989 Pal K, Kopper L, Lapis K: Chemotherapeutic sensitivity of liver metastases from intrasplenically-growing Lewis lung tumor. Clin Exp Metast 1341-1347, 1983 Satoh M, Nagamura A, Imura N: Modulation of adriamycin toxicity by tissue-specific induction of metallothionein synthesis in mice. Life Sci 67:627-634, 2000 Ali MM, Frei E, Straub J, et al: Induction of metallothionein by zinc protects from daunorubicin toxicity in rats. Toxicology 179:85-93,2002 Katano K, Kondo A, Safaei R et al:Acquisition of resistance to cisplatin is accompanied by changes in the extracellular pharmacology of copper. Cancer Res 62:6559-6565, 2002 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2003 VL 9 IS 2 BP 96 EP 99 PG 4 ER PT J AU Cengiz-Boduroglu, E Irkkan, Bilir, G AF Cengiz-Boduroglu, Esin Irkkan, Cigdem Bilir, Gulay TI Is Nothingham Prognostic Index Correlated with Apoptosis and P53 Expression in Infiltrating Ductal Carcinoma of the Breast? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p53; apoptosis; disease free survival; prognostic index ID p53; apoptosis; disease free survival; prognostic index AB The role of p53 as a prognostic factor is not clear. P53 named as ''guardian of the genome” plays an important role in many intracellular regulatory systems, one of which is apoptosis, having an impact on tumor kinetics. A retrospective study was undertaken to assess the relationship of the Nothingham Prognostic Index (NPI) to p53 expression and apoptotic cell counts. To conduct the study, 160 successive cases of infiltrating ductal carcinoma of the breast were included. P53 was assessed on AP-AAP stained sections. Apoptotic cell counting (ACC) was done on the HE stained routine sections in 10 HPFs. Clinical data were derived from the hospital files. Apoptotic cell counts were higher in the p53 positive group but the difference was not significant (p=0.079). P53 positivity was found to be related to the disease-free survival (DFS) (p=0.008). NPI was significantly higher in apoptotic cell containing group (p=0.006). There was a positive linear correlation between ACC and NPI scores (p=0.004). This correlation was not present between apoptosis and disease free survival. P53 expression was found to be related with DFS but not with the NPI which is a score composed of the best prognostic indicators known today. In contrast to this, ACC was found to be closely and linearly associated to the known prognostic factors. This may suggest that the apoptotic cell counts done on routine sections may be used as a part of prognosis assessment in infiltrating ductal carcinoma. C1 [Cengiz-Boduroglu, Esin] Dr. Abdurrahman Yurtarslan Ankara Oncology Education and Research Hospital, Department of Pathology, Ortadogu Camlik Sitesi No: 88 Karakusunlar, 06530 Ankara, Turkey. [Irkkan, Cigdem] Dr. Abdurrahman Yurtarslan Ankara Oncology Education and Research Hospital, Department of Pathology, Ortadogu Camlik Sitesi No: 88 Karakusunlar, 06530 Ankara, Turkey. [Bilir, Gulay] Dr. Abdurrahman Yurtarslan Ankara Oncology Education and Research Hospital, Department of Pathology, Ortadogu Camlik Sitesi No: 88 Karakusunlar, 06530 Ankara, Turkey. RP Cengiz-Boduroglu, E (reprint author), Dr. Abdurrahman Yurtarslan Ankara Oncology Education and Research Hospital, Department of Pathology, 06530 Ankara, Turkey. EM ebodur@ada.net.tr CR Askmalm-Stenmark M, Stal O, Sullivan S, Ferraud L, et al.: Cellular accumulation of p53 protein: an independent prognostic factor in stage II breast cancer. Eur J Cancer 30A:175-180, 1994 Baak JP, Dop H, Kurver PHJ: The value of morphometry to classic prognosticators in breast cancer. Cancer 56:374-382, 1985 Barnes DM, Dublin AE, Fisher CJ, et al.: Immunohistochemical detection of p53 protein in mammary carcinoma: An important new independent indicator of prognosis? Hum Pathol 24:469-476, 1993 Cummings MC, Winterford CM, Walker NI: Apoptosis. Am J Surg Pathol 21:88-101, 1997 Elston CW, Ellis IO: Pathological prognostic factors in breast cancer. Histopathology 19:403-410, 1991 Gonzales-Campora R, Galera Ruiz MR, Vasquez RF, et al.: Apoptosis in breast carcinoma. Pathol Res Pract 196: 167-174, 2000 Haybittle JL, Blamey RW, Elston CW:A prognostic index in primary breast cancer. Br J Cancer 45:361-366, 1982 Kovach JS, Hartmann A, Blaszyk H: Mutation detection by highly sensitive methods indicates that p53 gene mutations in breast cancer can have important prognostic value. Proc Natl Acad Sci USA 93:1093-1096, 1996 Lane DP: p53, guardian of the genome. Nature 358:15-16, 1992 Lipponen P, Aaltomaa S, Kosma VM, Syrjanen K: Apoptosis in breast cancer as related to histopathologic characteristics and prognosis. Eur J Cancer 30A:2068-2073, 1994 Moll UM, Ostermeyer AG, Ahomadegbe JC: P53 mediated tumor cell response to chemotherapeutic DNA damage: a preliminary study in matched pairs of breast cancer biopsies. Hum Pathol 26:1293-1301, 1995 O’Malley FP, Saad Z, Kerkvliet N, Doig G, et al.: The predictive power of semiquantitative immunohistochemical assessment of p53 and c-erbB2 in lymph node negative breast cancer. Hum Pathol 27:955-963, 1996 Ozbun MA, Butel JS: Tumor suppressor p53 mutations and breast cancer: critical analysis. Adv Cancer Res 66:71-7, 1995 Sher CJ: Cancer cell cycles. Science 274:1672-1677, 1996 Thorlacius S, Thorgilsson B, Bjornsson J, et al.: TP53 mutations and abnormal p53 protein staining in breast carcinomas related to prognosis. Eur J Cancer 31A:1856-1861, 1995 Zhang GJ, Kimijima I, Abe R, Watanabe T, et al.: Apoptotic index correlates to bcl-2 and p53 protein expression, histological grade and prognosis in invasive breast cancers. Anticancer Res 18: 1989-1998, 1998 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2003 VL 9 IS 2 BP 100 EP 103 PG 4 ER PT J AU Szomor, Zenou, P Roda, D Al Saati, T Csanaky, Gy Pajor, L Kelenyi, G Delsol, G Losonczy, H AF Szomor, Arpad Zenou, Prune Roda, Daniel Al Saati, Talal Csanaky, Gyorgy Pajor, Laszlo Kelenyi, Gabor Delsol, Georges Losonczy, Hajna TI Genotypic Analysis in Primary Systemic Anaplastic Large cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE anaplastic large cell lymphoma; TCR g- and bgene rearrangement; IgH gene rearrangement; t(2; 5)(p23; q35) and t(1; 2)(q25; p23) translocation ID anaplastic large cell lymphoma; TCR g- and bgene rearrangement; IgH gene rearrangement; t(2; 5)(p23; q35) and t(1; 2)(q25; p23) translocation AB This report presents an experience of polymerase chain reaction (PCR) analysis of T-cell receptor g- and bgene (TCR g, TCR b), and immunoglobulin heavy chain (IgH) gene rearrangements in 9 cases of primary systemic anaplastic large cell lymphoma. We showed 2 clonal IgH, 2 TCR g, 1 TCR b rearrangements. The genotype was B/T-cell in 1, T-cell in 1, B-cell in 1 and null cell-type in 6 cases. We used reverse transcriptase PCR (RT-PCR) to detect t(2;5)(p23;q35) and t(1;2)(q25;p23) translocations. T(2;5) translocation was demonstrated in 2 cases, there was no t(1;2) translocation. In most cases the molecular genetic results were found to be in agreement with immunophenotypic data. C1 [Szomor, Arpad] University of Pecs, I. Department of Internal Medicine, Ifjusag u.13., 7624 Pecs, Hungary. [Zenou, Prune] Central Hospital University Purpan, Laboratory of Anatomic PathologyToulouse, France. [Roda, Daniel] Central Hospital University Purpan, Laboratory of Anatomic PathologyToulouse, France. [Al Saati, Talal] Central Hospital University Purpan, Laboratory of Anatomic PathologyToulouse, France. [Csanaky, Gyorgy] Markusovszky Hospital, Department of PathologySzombathely, Hungary. [Pajor, Laszlo] University of Pecs, Department of PathologyPecs, Hungary. [Kelenyi, Gabor] University of Pecs, Department of PathologyPecs, Hungary. [Delsol, Georges] Central Hospital University Purpan, Laboratory of Anatomic PathologyToulouse, France. [Losonczy, Hajna] University of Pecs, I. Department of Internal Medicine, Ifjusag u.13., 7624 Pecs, Hungary. RP Szomor, (reprint author), University of Pecs, I. Department of Internal Medicine, 7624 Pecs, Hungary. EM aszomor@clinics.pote.hu CR Falini B: Anaplastic large cell lymphoma: pathological, molecular and clinical features. Br J Haematol 114: 741-760, 2001. Harris NL, Jaffe ES, Diebold J, et al: World Health Organization classification of neoplastic diseases of the hematopoetic and lymphoid tissues: report of the Clinical Advisory Committee Meeting – Airlie House, Virginia, November 1997. J Clin Oncol 17: 3835-3849, 1999. Lamant L, Dastugue N, Pulford K, et al: A new fusion gene TPM3-ALK in anaplastic large cell lymphoma created by a, 1;2)(q25;p23, translocation. Blood 93: 3088-3095, 1999. Lamant L, Meggetto F, Al Saati T, et al: High incidence of the t(2;5)(p23;q35, translocation in anaplastic large cell lymphoma and its lack of detection in Hodgkin’s disease. Comparison of cytogenetic analysis, reverse trancriptase-polymerase chain reaction, and P-80 immunostaining. Blood 87: 284-291, 1996. Morris SW, Kirstein MN, Valentine MB, et al: Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non- Hodgkin’s lymphoma. Science 263: 1281-1284, 1994. Nagy M, Feher K, Laszlo T, et al: Analysis of T-cell receptorg- gene rearrangement in lymphoproliferative disorders using polymerase chain reaction. Orv Hetil 140: 2441-2444, 1999. Pulford K, Lamant L, Morris SW, et al: Detection of anaplastic lymphoma kinase, ALK, and nucleolar protein nucleophosmin, NPM)-ALK proteins in normal and neoplastic cells with the monoclonal antibody ALK1. Blood 89: 1394-1404, 1997. Stein H, Foss HD, Durkop H, et al: CD30+ anaplastic large cell lymphoma: a review of its histopathologic, genetic and clinical features. Blood 96: 3681-3695, 2000. Stein H, Mason DY, Gerdes J, et al: The expression of the Hodgkin’s disease associated antigen Ki-1 in reactive and neoplastic lymphoid tissue: evidence that Reed-Sternberg cells and histiocytic malignancies are derived from activated lymphoid cells. Blood 66: 848-858, 1985. Theriault C, Galoin S, Valmary S, et al: PCR analysis of immunoglobulin heavy chain, IgH, and TcR-gamma chain rearrangements in the diagnosis of lymphoproliferative disorders of a study of 525 cases. Mod Pathol 13: 1269-1279, 2000. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2003 VL 9 IS 2 BP 104 EP 106 PG 3 ER PT J AU Kurup, KR Nair, AR Kurup, AP AF Kurup, Kumar Ravi Nair, A Rekha Kurup, Achutha Parameswara TI Isoprenoid Pathway Related Cascade in Multiple Myeloma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE digoxin; dolichol; ubiquinone; multiple myeloma ID digoxin; dolichol; ubiquinone; multiple myeloma AB This study assessed the changes in the isoprenoid pathway and its metabolites digoxin, dolichol and ubiquinone in multiple myeloma. The following parameters were assessed: isoprenoid pathway metabolites, tyrosine and tryptophan catabolites, glycoconjugate metabolism, RBC membrane composition and free radical metabolism. There was elevation in plasma HMG CoA reductase activity, serum digoxin and dolichol and a reduction in RBC membrane Na+ - K+ ATPase activity, and serum ubiquinone levels. Serum tryptophan, serotonin, nicotine, strychnine and quinolinic acid were elevated while tyrosine, dopamine, noradrenaline and morphine were decreased. The total serum glycosaminoglycans and glycosaminoglycan fractions, the activity of GAG degrading enzymes and glycohydrolases, carbohydrate residues of glycoproteins and serum glycolipids were elevated. The RBC membrane glycosaminoglycans, hexose and fucose residues of glycoproteins, cholesterol and phospholipids were reduced. The activity of all free radical scavenging enzymes, concentration of glutathione, iron binding capacity and ceruloplasmin decreased significantly while the concentration of lipid peroxidation products and NO increased. Hyperdigoxinemia related altered intracellular Ca++ mediated oncogene activation, dolichol induced altered glycoconjugate metabolism and ubiquinone deficiency related mitochondrial dysfunction can contribute to the pathogenesis of multiple myeloma. The biochemical findings reported could be the cause or the consequence of multiple myeloma. C1 [Kurup, Kumar Ravi] Medical College Hospital, Department of NeurologyKerala, India. [Nair, A Rekha] Regional Cancer Center, II. Department of PathologyKerala, India. [Kurup, Achutha Parameswara] Metabolic Disorders Research Center, Gouri Sadan, T.C.4/1525, North of Cliff House, Kattu Road, Kowdiar P.O., TrivandrumKerala, India. RP Kurup, AP (reprint author), Metabolic Disorders Research Center, Kerala, India. EM kvgnair@satyam.net.in CR Haupert GT: Sodium pump regulation by endogenous inhibition. Topics in Membrane Transport 34: 345-348, 1989 Cowen PJ, Wood AJ: Biological markers of depression. Psychol Med 21:831-836, 1991 Kramer HJ, Meyer Lehnert H et al: Endogenous natriuretics and ouabain like factors and their roles. Am J Hypertens 1: 81- 89, 1991 Tamura H, Shimoyama S, Sunaga Y: Digoxin like immunoreactive substance in urine of patients with mucocutaneous lymphnode syndrome. Angiology 10: 856-865, 1992 Rhee HM: Parallelism between transport inhibition and L1210 cell growth by ouabain. Adv Exp Med Biol 22 : 727-734, 1988 Buchwald H:Cholesterol inhibition, cancer and chemotherapy. Lancet 339: 1154-1156, 1992 Barinaga M. From Bench Top to Bedside. Science 278: 1036- 1038, 1997 Jacob RA. Nutrition, health and antioxidants. INFORM 5:1271-1275, 1994 Linstinsky JL, Siegal GP, Listinsky CM. Alpha-L-Fucose a potentially critical molecule in pathologic process including neoplasia. Am J Clin Pathol 110: 425-440, 1998 Teoh G, Anderson KC. Interaction of tumor and host cells with adhesion and extracellular matrix molecules in the development of multiple myeloma. Hematol Oncol Clin N Amer 11: 27-39,1997 Hisaka A, Kasamatu, Takenaga N: Absorption of a novel prodrug DOPA. Drug Metab Disp 18: 621-625, 1990 Saito K, Crowley JS, Markey S, Heyes MP: A mechanism for increased quinolinic acid formation following acute systemic immune stimulation. J Biol Chem 268: 15496-503, 1993 Felton D, Cohen N, Ader R: Psychoneuroimmunology. Academic Press, New York 1991 Arun P, Ravikumar A, Leelamma S, Kurup PA: Endogenous alkaloids in the brain of rats loaded with tyrosine/tryptophan and in the serum of patients of neurodegenerative and psychiatric disorders. Ind J Med Res 107: 231-238, 1998b Rao AV, Ramakrishnan S: Estimation of HMG CoA reductase activity. Clin Chem 21:1523-1528, 1975 Wallach DFH, Kamath VB. Methods Enzymol 8.Academic Press, New York, 1966. Arun P, Ravikumar A, Leelamma S, Kurup PA: Identification and estimation of endogenous EDLF in biological fluids and tissues by TLC and HPLC. Ind J Biochem Biophys 35: 308- 312, 1998a Palmer DN, Maureen AA, Robert DJ. Separation of some neutral lipids by normal phase high performance liquid chromatography on a cyanopropyl column ubiquinone, dolichol and cholesterol levels in sheep liver. Anal Biochem 140:315- 319, 1984 Bloxam DL, Warren WH. Error in the determination of tryptophan by the method of Denkala and Dewey. A revised procedure. Anal Biochem 60: 621-625, 1974 Wong PWK, O’Flynn ME, Inouye R. Flourimetric method for Tyrosine. Clin Chem 10: 1098-1100, 1964 Curzon G, Green AR.Rapid method for the determination of 5- hydroxy tryptophan and 5-hydroxy indoleacetic acid in certain regions of rat brain. Br J Pharmacol 39: 653-655, 1979 Well-Malherbe R.Methods of Biochemical Analysis. Inter Science, New York, 1971 Manoj AJ, Kurup PA. Changes in the glycosaminoglycans and glycoproteins in the rat brain during protein calorie malnutrition. Clin Biochem Nutr 25: 149-157,1998 Lowenstein JM. Methods in Enzymology. Academic Press, New York, 1969 Kakkar P, Das B, Viswanathan PN: A modified spectrophotometric assay of SOD. Ind J Biochem Biophys 21: 130, 1984 Maehly AC, Chance B: The assay of catalase and peroxidase. Methods Biochemical Analysis 2: 357, 1954 Paglia DE, Valentine WN. Studies on quantitative and qualitative characterisation of erythrocyte glutathione peroxidase. J Lab Clin Med 70: 158, 1967 Horn H D, Burns FH: Methods of Enzymatic Analysis. Academic Press, New York 1978 Brien P J O. Estimation of conjugated dienes and hydroperoxide. Can J Biochem 47: 485, 1969 Beutler E, Duran O, Kelley BM: Modified procedure for the estimation of reduced glutathione. J Lab Clin Med 61: 882,1963 Colin G, Rammel R, Cunlifee B, Keiboom AJ.Determination of alpha tocopherol in biological specimens by high performance liquid chromatography. J Liq Chromatogr 6: 1123,1983 Gabor G, Allon N: Spectrofluorometric method for NO determination. Anal Biochem 220: 16, 1994 Wootton IDP: Estimation of iron binding capacity. Microanal Med Biochem 4: 124, 1964 Henry RJ, Chiamori N, Jacobs SL, Segalov M. Estimation of ceruloplasmin. Proc Soc Exp Biol Med 104: 620, 1960 Falholt K, Lund B, Fatholt W. Estimation of free fatty acids. Clin Chem Acta 46: 105, 1973 Haga H. Effects of dietary magnesium supplementation on diurnal variation of BP and plasma Na+-K+ ATPase activity in essential hypertension. Jap Heart J 33: 785-798, 1992 Feinman RM, Sawyer J, Hardin J, Tricot G: Cytogenetics and molecular genetics in multiple myeloma. Hemat Oncol Clin N Am 11: 1-21, 1997 Ravikumar A., Deepadevi KV, Arun P et al: Tryptophan and tyrosine catabolic pattern in neuropsychiatric disorders. Neurol Ind 48: 231-238, 2001 Irwin M, Lacher U, Caldwell C. Depression and decreased natural killer cell cytotoxicity. A longitudinal study of depressed patients and control subjects. Psychol Med 22:1045-1050, 1992 Lauder JM. Neurotransmitters as growth regulatory signals: role of receptors and second messengers. Trends Neurosci 16: 233-238, 1993 Greenamyre JT, Poter RHP. Anatomy and Physiology of glutamate in CNS. Neurol 44 ,S8: S7-S13, 1994 Monia BP, Ecker J, Crooks ST. Ubiquitination Enzymes. Biotechnol 8:209-215, 1990 Ploegh HL. Viral strategies for immune evasion. Science 280:248-253, 1998 Rettig MB, Ma HJ, Vesico RA et al.: Kaposi’s sarcoma associated herpesvirus infection of bone marrow dendritic cells from multiple myeloma patients. Science 276:1851-1857, 1997 Green DR, Reed JC: Mitochondria and apoptosis. Science 281:1309-1316, 1998 Mukhopadhyay CK, Attieh ZK, Fox PL: Role of ceruloplasmin in cellular iron uptake. Science 279 : 714-717, 1998 Finkel TH: T-cell development and transmembrane signalling. Changing biological responses through a unchanging receptor. Immunol Today 12: 79-86, 1991 Barut BA, Zon LI, Cochran MK. Role of interleukin – 6 in the growth of myeloma derived cell lines. Leukaem Res 16: 951,1992 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2003 VL 9 IS 2 BP 107 EP 114 PG 8 ER PT J AU Esik, O Csere, T Stefanits, K Lengyel, Zs Safrany, G Vonoczky, K Lengyel, E Nemeskeri, Cs Repa, I Tron, L AF Esik, Olga Csere, Tibor Stefanits, Klara Lengyel, Zsolt Safrany, Geza Vonoczky, Katalin Lengyel, Erzsebet Nemeskeri, Csaba Repa, Imre Tron, Lajos TI A Review on Radiogenic Lhermitte's Sign SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE radiation myelopathy; permanent Lhermitte's sign; magnetic resonance imaging; positron emission tomography; [18F]fluorodeoxyglucose; [15O]butanol; [11C]methionine ID radiation myelopathy; permanent Lhermitte's sign; magnetic resonance imaging; positron emission tomography; [18F]fluorodeoxyglucose; [15O]butanol; [11C]methionine AB Radiation myelopathy is a rare, but extremely serious side-effect of radiotherapy. Recovery from radiation-induced motor sequelae is rare, whereas, the regeneration of sensory losses is relatively frequent. Among the sensory radiogenic injuries of the spinal cord, Lhermitte's sign (LS) is most frequent. This review describes the clinical picture and diagnostic imaging signs of radiogenic LS. There have been only a few studies on large patient groups with radiogenic LS, demonstrating a rate of occurrence of 3.6-13%, relating mainly to mantle irradiation or the radiotherapy of head and neck tumors. These cases typically manifest themselves 3 months following radiotherapy and gradually disappear within 6 months. Only 3 LS cases have been described in the English literature with extraordinarily severe symptoms lasting for more than 1 year. MRI, a sensitive tool in the detection of demyelination, failed to reveal any pathological sign accompanying radiogenic LS. However, positron emission tomography demonstrated increased [18F]fluorodeoxyglucose accumulation and [15O]butanol perfusion, but a negligible [11C]methionine uptake in the irradiated spinal cord segments in patients with long-standing LS. These imaging data are suggestive of a close direct relationship between the regional perfusion and metabolism of the spinal cord, very much like the situation in the brain. We postulate that an altered, energy-demanding conduction along the demyelinated axons of patients with chronic radiogenic LS may explain the increased metabolism and perfusion. C1 [Esik, Olga] Semmelweis University, Department of Oncology, Rath Gyorgy u 7-9, H-1122 Budapest, Hungary. [Csere, Tibor] University of Pecs, Department of OncologyPecs, Hungary. [Stefanits, Klara] University of Pecs, Department of OncologyPecs, Hungary. [Lengyel, Zsolt] University of Debrecen, PET CenterDebrecen, Hungary. [Safrany, Geza] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Frederic Joliot-Curie Sugarbiologiai es Sugaregeszsegugyi Kutato IntezetBudapest, Hungary. [Vonoczky, Katalin] National Institute of Oncology, Outpatient Department of NeurologyBudapest, Hungary. [Lengyel, Erzsebet] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Nemeskeri, Csaba] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai KozpontBudapest, Hungary. [Repa, Imre] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. [Tron, Lajos] University of Debrecen, PET CenterDebrecen, Hungary. RP Esik, O (reprint author), Semmelweis University, Department of Oncology, H-1122 Budapest, Hungary. EM esik@oncol.hu CR Arasil E, Tascioglu AO: Spontaneous migration of an intracranial bullet to the cervical spinal canal causing Lhermitte’s sign. Case report. J Neurosurg 56:158-159, 1982 Ballantyne AJ: Late sequelae of radiation therapy in cancer of the head and neck with particular reference to the nasopharynx. Am J Surg 130:433-436, 1975 Bentzen SM: Potential clinical impact of normal-tissue intrinsic radiosensitivity testing. Radiother Oncol 43:121-131, 1997 Blackburn CRB, McLeod JG: CNS lesions in cystinuria. Arch Neurol 34:638-639, 1977 Blanco G, Peters HA: Myeloneuropathy and macrocytosis associated with nitrous oxide abuse. Arch Neurol 40:416-418, 1983 Boden G: Radiation myelitis of the cervical spinal cord. Brit J Radiol 21:464-469, 1948 Brock WA, Tucker SL, Geara FB, et al: Fibroblast radiosensitivity versus acute and late normal skin responses in patients treated for breast cancer. Int J Radiat Oncol Biol Phys 32:1371- 1379, 1995 Busch D: Genetic susceptibility to radiation and chemotherapy injury: diagnosis and management. Int J Radiat Oncol Biol Phys 30:997-1002, 1994 Butler WM, Taylor HG, Diehl LF: Lhermitte’s sign in cobalamin, vitamin B12, deficiency. JAMA 245:1059, 1981 Carmel RJ, Kaplan HS: Mantle irradiation in Hodgkin’s disease. An analysis of technique, tumor eradication, and complications. Cancer 37:2813-2825, 1976 Chan RC, Steinbok P: Delayed onset Lhermitte’s following head and/or neck injuries. Report of four cases. J Neurosurg 60:609-612, 1984 Dewar J, Lunt H, Abernethy DA, et al: Cisplatin neuropathy with Lhermitte’s sign. J Neurol Neurosug Psy 49:96-99, 1986 Di Chiro G, Oldfield E, Bairamian D, et al: Metabolic imaging of the brain stem and spinal cord: studies with positron emission tomography using 18F-2-deoxyglucose in normal and pathological cases. J Comput Assist Tomogr 7:937-945, 1983 Dynes JB, Smedal MI: Radiation myelitis. Am J Roentgenol 83:78-87, 1960 Eeles R, Tait DM, Peckham MJ: Lhermitte’s sign as a complication of cisplatin-containing chemotherapy for testicular cancer. Cancer Treat Rep 70:905-907, 1986 Esik O, Emri M, Csornai M, et al: Radiation myelopathy with partial functional recovery: PET evidence of long-term increased metabolic activity of the spinal cord. J Neurol Sci 163:39-43, 1999 Esik O, Csere T, Stefanits K, et al: Increased metabolic activity in the spinal cord of patients with long-standing Lhermitte’s sign. Strahlenther Onkol, in press Fein DA, Marcus RB Jr, Parsons JT, et al: Lhermitte’s sign: incidence and treatment variables influencing risk after irradiation of the cervical spinal cord. Int J Radiat Oncol Biol Phys 27:1029-1033, 1993 Felts PA, Baker TA, Smith KJ: Conduction in segmentally demyelinated mammalian central axons. J Neurosci 17:7267- 7277, 1997 Fowler JF: The linear-quadratic formula and progress in fractionated radiotherapy. Brit J Radiol 62:679-694, 1989 Glanzmann C, Aberle HG, Horst W: The risk of chronic progressive radiation myelopathy. Strahlenther 152:363-372, 1976 Gutrecht JA, Zamani AA, Slagado ED: Anatomic-radiologic basis of the Lhermitte’s sign in sclerosis multiplex. Arch Neurol 50:849-851, 1993 Hall EJ: Radiobiology for the radiologists. 5th ed. Philadelphia: Lippincott-Williams and Wilkins, 2000 Hendry JH: Genomic instability: potential contributions to tumour and normal tissue response, and second tumours, after radiotherapy. Radiother Oncol 59:117-126, 2001 Higano S, Shishido F, Nagashima M, et al: PET evaluation of spinal cord tumor using 11C-methionine. J Comput Assist Tomogr 14:297-299, 1990 Inbar M, Merimsky O, Wigler N, et al: Cisplatin-related Lhermitte’s sign. Anticancer Drugs 3:357-377, 1992 Jellinger K, Sturm KW: Delayed radiation myelopathy in man. Report of twelve necropsy cases. J Neurol Sci 14:389-408, 1971 Jones A: Transient radiation myelopathy, with reference to Lhermitte’s sign of electrical paraesthesia). Brit J Radiol 37:727-744, 1964 Kachandani R, Howe JG: Lhermitte’s sign in multiple sclerosis: a clinical survey and review of the literature. J Neurol Neurosurg Psy 45:308-312, 1982 Kamoto Y, Sadato N, Yonekura Y, et al: Visualization of the cervical spinal cord with FDG and high-resolution PET. J Comput Assist Tomogr 22:487-491, 1998 Kiltie AE, Ryan AJ, Swindell R, et al: A correlation between residual radiation-induced DNA double-strand breaks in cultured fibroblasts and late radiotherapy reactions in breast cancer patients. Radiother Oncol 51:55-65, 1999 Kristensson K, Molin B, Sourander P: Delayed radiation lesions of the human spinal cord. Report of five cases. Acta Neuropathol 9:34-44, 1967 Kunkel M, Grotz KA, Forster GJ, et al: Therapy monitoring with 2-(18F)-FDG positron emission tomography after neoadjuvant radiation treatment of mouth carcinoma. Strahlenther Onkol 177:145-152, 2001 Lang O, Bihl H, Hultenschmidt B, et al: Clinical relevance of positron emission tomography, PET, in treatment control and relapse of Hodgkin’s disease. Strahlenther Onkol 177:138-144, 2001 Layzer RB: Myeloneuropathy after prolonged exposure to nitrous oxide. Lancet 2:1227-1230, 1978 Leaver RC, Loeser JD: Lhermitte’s phenomenon after high velocity missile injuries of the brain. J Neurosurg 34:159-163, 1981 Lengyel Z, Reko G, Majtenyi K, et al: Autopsy verifies demyelination but lack of vascular damage in partially reversible radiation myelopathy. Spinal Cord, in press Lewanski CR, Sinclair JA, Stewart JS: Lhermitte’s sign following head and neck radiotherapy. Clin Oncol, R Coll Radiol, 12:98-103, 2000 Lhermitte J: Sur l’hypertrophie musculaire consecutive aux lesions grave des nerfs peripherique: une observation nouvelle. Press Med 26:64, 1918 Lhermitte J, Bollak M, Nicolas M: Les douleurs a type de decharge electrique consecutives a la flexion cephalique dans la sclerose en plaques. Un cas de form sensitive de la sclerose multiple. Rev Neurol 39:56-62, 1924 List AF, Kummet TD: Spinal cord toxicity complicating treatment with cisplatin and etoposid. Am J Clin Oncol 13:256- 258, 1990 Lossos A, Siegal T: Electric shock-like sensations in 42 cancer patients: clinical characteristics and distinct etiologies. J Neurooncol 29:175-181, 1996 Marie P, Chatelin C: Note sur certains symptomes vraisemblament d’origine radiculaire chez les blessees du crane. Rev Neurol 24:143-144, 1917 Meltzer CC, Townsend DW, Kottapally S, et al: FDG imaging of spinal cord primitive neuroectodermal tumor. J Nucl Med 39:2121-2127, 1998 Mendenhall WM, Million RR, Bova FJ: Carcinoma of the cervical esophagus treated with radiation therapy using fourfield box technique. Int J Radiat Oncol Biol Phys 8:1435- 1439, 1982 National Cancer Institute. Cancer Therapy Evaluation Program: Common toxicity criteria, Version 2.0. http://ctep.info.nih.gov, 1997) Newton HB, Rea GL: Lhermitte’s sign as a presenting symptom of primary spinal cord tumor. J Neurooncol 29:183-188, 1996 Page NGR, Spiteri MA: Lhermitte’s sign in Behcet’s disease. Brit Med J 284:704-705, 1982 Palmer JJ: Radiation myelopathy. Brain 95:109-122, 1972 Parry GJ, Bredesen DE: Sensory neuropathy with low-dose pyridoxine. Neurology 35:1466-1468, 1985 Rades D, Kuhnel G, Wildfang I, et al: The value of positron emission tomography, PET, in the treatment of patients with cancer of unknown primary, CUP). Strahlenther Onkol 177:525-529, 2001 Rahn AN, Baum RP, Adamietz IA, et al: Value of [18F]fluorodeoxyglucose positron emission tomography in radiotherapy planning of head-neck tumors. Strahlenther Onkol 174:358- 364, 1998 Read CF: Multiple sclerosis with Lhermitte’s sign. Arch Neurol Psychiat 27:227-228, 1932 Reagan TJ, Thomas JE, Colby MY Jr: Chronic progressive radiation myelopathy. Its clinical aspects and differential diagnosis. JAMA 203:128-132, 1968 Roland PE: Brain activation. New York: Wiley-Liss Inc, 469- 504, 1993 Sasajima T, Mineura K, Itoh Y, et al: Spinal cord ependymoma: a positron emission tomographic study with, 11C-methyl)-Lmethionine. Neuroradiology 38:53-55, 1996 Schultheiss TE, Stephens LC, Maor MH: Analysis of the histopathology of radiation myelopathy. Int J Radiat Oncol Biol Phys 14:27-32, 1988 Schultheiss TE, Kun LE, Ang KK, et al: Radiation response of the central nervous system. Int J Radiat Oncol Biol Phys 31:1093-1112, 1995 Ševeikova L, Bolješikova E, Chorvath M, et al: Long-term, 15 years, results of extended field radiotherapy in Hodgkin’s disease. Neoplasma 47:253-256, 2000 Signorini GC, Beltramello A, Pinna G, et al: The significance of preoperative neurological disorders in predicting outcome of cervical spondylotic myelopathy after surgery. J Neurosurg Sci 28:89-92, 1984 Solheim OP: Radiation injury of the spinal cord. Acta Radiol Ther Phys Biol 10:474-480, 1971 Thornton AF, Zimberg SH, Greenberg HS, et al: Protracted Lhermitte’s sign following head and neck irradiation. Arch Otolaryngol Head Neck Surg 117:1300-1303, 1991 Traynelis VC, Hitchon PW, Yuh WT, et al: Magnetic resonance imaging and posttraumatic Lhermitte’s sign. J Spinal Disord 3:376-379, 1990 Valk PE, Dillon WP: Diagnostic imaging of central nervous system radiation injury. In: Gutin PH, Leibel SA, Sheline GE, eds, Radiation injury to the nervous system. New York: Raven Press Ltd, 211-237, 1991 van den Bent MJ, Hilkens PHE, Sillevis Smitt PAE, et al: Lhermitte’s sign following chemotherapy with docetaxel. Neurology 50:563-564, 1998 Ventafridda V, Caraceni A, Martini C, et al: On the significance of Lhermitte’s sign in oncology. J Neurooncol 10:133-137, 1991 Vollmer TL, Brass LM, Waxman SG: Lhermitte’s sign in a patient with herpes zoster. J Neurol Sci 106:153-157, 1991 Walther PJ, Rossitch E Jr, Bullard DE: The development of Lhermitte’s sign during cisplatin chemotherapy: possible druginduced toxicity causing spinal cord demyelination. Cancer 60:2170-2172, 1987 Waxman SG: Demyelinating diseases – new pathological insights, new therapeutic targets. N Eng J Med 338:323-325, 1998 Weber WA, Avril N, Schwaiger M: Relevance of positron emission tomography, PET, in oncology. Strahlenther Onkol 175:356-373, 1999 Wen PY, Blanchard KL, Block CC, et al: Development of Lhermitte’s sign after bone marrow transplantation. Cancer 69:2262-2266, 1992 Wilmshurst JM, Barrington SF, Pritchard D, et al: Positron emission tomography in imaging spinal cord tumors. J Child Neurol 15:465-472, 2000 Woesler B, Kuwert T, Probst-Cousin S, et al: Spinal metastases of a high grade astrocytoma visualized with FDG-PET. Clin Nucl Med 22:863-864, 1997 Wong CS, van Dyk J, Milosevic M, et al: Radiation myelopathy following single courses of radiotherapy and retreatment. Int J Radiat Oncol Biol Phys 30:575-581, 1994 Word JA, Kalokhe UP, Aran BS, et al: Transient radiation myelopathy, Lhermitte’s sign, in patients with Hodgkin’s disease treated by mantle irradiation. Int J Radiat Oncol Biol Phys 6:1731-1733, 1980 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2003 VL 9 IS 2 BP 115 EP 120 PG 6 ER PT J AU Farjadian, Sh Asadi, E Doroudchi, M Samsami Dehaghani, A Tabei, S Kumar, VP Ghaderi, A AF Farjadian, Shirin Asadi, E Doroudchi, Mehrnoosh Samsami Dehaghani, Alamtaj Tabei, Z. Seyed Kumar, Vijayananda Perikala Ghaderi, Abbas TI High Risk HPV Types in Southern Iranian Patients with Cervical Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cervical cancer; HPV ID cervical cancer; HPV AB This study was undertaken to assess the rate of HPV infection in cervical carcinoma among southern Iranian patients. 101 archival cervical carcinoma tissue samples of a 10 year period were studied for the presence of HPV DNA in southern Iran by a polymerase chain reaction method. In addition, the presence of high risk HPV-16 and HPV-18 genotypes was investigated. In total, 88 (87.1%) of the samples were HPV DNA positive, of which 83 were squamous cell carcinomas and 5 were adenocarcinomas. HPV-16 genotype was detected in 26.7% of HPV positive cervical carcinomas; however, none of the samples were positive for the existence of HPV-18 genotype. Collectively, these results suggest that HPV-16 and HPV-18 are not the frequent high risk HPV types in our patients and circulating HPV types in southern Iranian population are different from many other populations. C1 [Farjadian, Shirin] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Asadi, E] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Doroudchi, Mehrnoosh] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Samsami Dehaghani, Alamtaj] Shiraz University of Medical Sciences, Department of Obstetrics and GynecologyShiraz, Iran. [Tabei, Z. Seyed] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Kumar, Vijayananda Perikala] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Ghaderi, Abbas] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. RP Ghaderi, A (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Immunology, Shiraz, Iran. EM ghaderia@sums.ac.ir CR Parkin DM: Global cancer statistics in the year 2000. Lancet Oncol 2:533-543, 2001 Farahmandbeigi M, Kadivar MR: The incidence rate of registered cancer in Fars province. Disease Control Unit, Shiraz University Press, Iran, 2000 Bosch FX, Munoz N. The viral etiology of cervical cancer. Virus Res 89:183-190, 2002 Walboomers JM, Jacobs MV, Manos MM, et al. Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189:12-19, 1999 Southern SA, Herrington CS. Disruption of cell cycle control by human papillomaviruses with special reference to cervical carcinoma. Int J Gynecol Cancer 10:263-274, 2000 Park TW, Fujiwara H, Wright TC. Molecular biology of cervical cancer and its precursors. Cancer 76:1902-1913, 1995 IARC working Group, Human papilloma viruses, IARC Monograph on the evaluation of carcinogenic risks to humans. Lyon, France: International Agency for Research on Cancer, 65, 1995 Badaracco G, Venuti A, Sedati A, Marcante ML. HPV16 and HPV18 in genital tumors: Significantly different levels of viral integration and correlation to tumor invasiveness. J Med Virol 67:574-582, 2002 Munoz N, Franceschi S, Bosetti C, et al. International Agency for Research on Cancer. Multicentric Cervical Cancer Study Group Role of parity and human papillomavirus in cervical cancer: the IARC multicentric case-control study.Lancet 359:1093-1101, 2002 Castle PE, Wacholder S, Lorincz AT, et al.A prospective study of high-grade cervical neoplasia risk among human papillomavirusinfected women. J Natl Cancer Inst 94:1406-1414, 2002 Rong S, Chen W, Wu L, et al. Analysis of risk factors for cervical cancer in Xiangyuan County, Shanxi Province Zhonghua Yu Fang Yi Xue Za Zhi 36:41-43, 2002 Mandelblatt JS, Lawrence WF, Womack SM, et al. Benefits and costs of using HPV testing to screen for cervical cancer. JAMA 287:2372-2381, 2002 Castellsague X, Menendez C, Loscertales MP, et al. Human papillomavirus genotypes in rural Mozambique. Lancet 358:1429-1430, 2001 Snijders PJ, van den Brule AJ, Schrijnemakers HF, et al. The use of general primers in the polymerase chain reaction permits the detection of a broad spectrum of human papillomavirus genotypes. J Gen Virol 71:173-181, 1990 Samoylova EV, Shaikhaiev GO, Petrov SV, et al. HPV infection in cervical cancer cases in Russia. Int J Cancer 61:337-341, 1995 Ferenczy A, Franco E. Cervical-cancer screening beyond the year 2000. Lancet Oncol 2:27-32, 2002 Sebbelov AM, Davidson M, Kruger Kjaer S, et al. Comparison of human papillomavirus genotypes in archival cervical cancer specimens from Alaska natives, Greenland natives and Danish Caucasians. Microbes Infect 2:121-126, 2000 Goncalves MA, Massad E, Burattini MN, Villa LL. Relationship between human papillomavirus, HPV, genotyping and genital neoplasia in HIV-positive patients of Santos City, Sao Paulo, Brazil. Int J STD AIDS 10:803-807, 1999 Lo KW, Wong YF, Chan MK, et al. Prevalence of human papillomavirus in cervical cancer: a multicenter study in China. Int J Cancer 100:327-331, 2002 Baay MF, Tjalma WA, Weyler J, et al. Human papillomavirus infection in the female population of Antwerp, Belgium: prevalence in healthy women, women with premalignant lesions and cervical cancer. Eur J Gynaecol Oncol 22:204-208, 2001 Chen S, O’Sullivan H, Tabrizi SN, et al. Prevalence and genotyping of HPV in cervical cancer among Australian women. Int J Gynaecol Obstet 67:163-168, 1999 Chabaud M, Le Cann P, Mayelo V, et al. Detection by PCR of human papillomavirus genotypes in cervical lesions of Senegalese women. J Med Virol 49:259-263, 1996 Beskow AH, Gyllensten UB. Host genetic control of HPV 16 titer in carcinoma in situ of the cervix uteri. Int J Cancer 101:526-531, 2002 Zwaveling S, Ferreira Mota SC, Nouta J, et al. Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. J Immunol 169:350-358, 2002 Dehaghani AS, Amirzargar A, Farjadian S, Ghaderi A. HLADQBl alleles and susceptibility to cervical squamous cell carcinoma in Southern Iranian patients. Pathol Oncol Res 8:58- 61, 2002 Amirzargar A, Mytilineos J, Farjadian Sh, et al. Human Leukocyte Antigen class II allele frequencies and haplotype association in Iranian population. Hum Immunol 62:1234- 1238, 2001 Hording U, Teglbjaerg CS, Visfeldt J, Bock JE. Human papillomavirus types 16 and 18 in adenocarcinoma of the uterine cervix. Gynecol Oncol 46:313-316, 1992 Bjersing L, Rogo K, Evander M, et al. HPV 18 and cervical adenocarcinomas. Anticancer Res 11:123-127, 1991 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2003 VL 9 IS 2 BP 121 EP 125 PG 5 ER PT J AU Otto, Sz AF Otto, Szabolcs TI Cancer Epidemiology in Hungary and the Bela Johan National Program for the Decade of Health SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer epidemiology; Hungary; morbidity; mortality ID cancer epidemiology; Hungary; morbidity; mortality AB According to a recent survey, in 1995 Hungary lead cancer mortality statistics in men in Europe, while was on the second place in the case of women. The figures for cancer morbidity were highly similar. According to cancer types, the Hungarian mortality rates are the worst in the case of lung, oral cavity laryngeal and pancreatic cancers among men and oral cavity colorectal and thyroid cancers in women. Between 1999 and 2001 in Hungary the cancer mortality list is topped by lung and colorectal cancers among men and by breast and colorectal cancers among women. The National Cancer Registry started to provide reliable morbidity data which indicate that in 2001 in Hungary the men's most frequent cancer types are lung, colorectal and lip and mouth cancers while among women breast, colorectal and lung cancers. These shocking cancer mortality and morbidity figures outlined the primary targets of the recently lunched national public health program for this decade. C1 [Otto, Szabolcs] National Institute of Oncology, Rath Gy. u. 7-9., H-1122 Budapest, Hungary. RP Otto, Sz (reprint author), National Institute of Oncology, H-1122 Budapest, Hungary. EM sz.otto@oncol.hu CR Andersen RE. Healthy people 2010. Phys Sportsmed 28: 7-8, 2000 Bray F, Sankila R, Ferlay J, Parkin DM. Estimates of cancer incidence and mortality in Europe in 1995. Eur J Cancer 38:99- 166, 2002 Demografiai evkonyvek KSH, Volumes of the Demographical Yearbook, KSH – Central Statistics Office, Budapest 1975- 2000 in Hungary,, in Hungarian) Public Health Program “For a Healthy Population” 2001-2010. Target Program for the Prevention of Public Health Problem. Egeszsegugyi Kozlony, Health Bulletin in Hungary, 51:2237- 2322, 2001, in Hungarian) Greenlee RT, Hill-Harmon MB, Murray T, Thun M. Cancer Statistics, 2001. CA Cancer J Clin 51:15-36, 2001 Otto S, Schumann B: Planning and development of tumormarker examinations. Lege Artis Med 9:114-117, 1999, in Hungarian) Otto S, Kasler M: Cancer mortality and Incidence in Hungary in relation to international data. Hungarian Oncol 46:111-117, 2002, in Hungarian) Jozan P: The basic trend of mortality in the 20th century and mortality conditions at the turn of the millennium in Hungary. Hungarian Science 4:419-439, 2002, in Hungarian) Dobrossy L: Cancer mortality in central-eastern Europe: facts behind the figures. Lancet Oncol 3:374-381, 2002 Kopp MS, Skrabski A and Szedmak S: Psychosocial risk factors, inequality and self-rated morbidity in a changing society. Soc Sci Med 51: 1351-1361, 2000 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2003 VL 9 IS 2 BP 126 EP 130 PG 5 ER PT J AU Gopcsa, L Barta, A Banyai, A Dolgos, J Halm, G Paloczi, K AF Gopcsa, Laszlo Barta, Aniko Banyai, Aniko Dolgos, Janos Halm, Gabriella Paloczi, Katalin TI Salvage Chemotherapy with Donor Lymphocyte Infusion and STI 571 In a Patient Relapsing with B-Lymphoblastic Phase Chronic Myeloid Leukemia after Allogeneic Bone Marrow Transplantation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE CML; blastic phase; allogeneic HSCT; DLI; STI 571 ID CML; blastic phase; allogeneic HSCT; DLI; STI 571 AB Relapse is the main cause of treatment failure following hematopoietic stem cell transplantation for blastic phase chronic myeloid leukemia. Treatment options including donor lymphocyte infusion, second transplantation, interferon- and re-induction chemotherapy are often unsuccessful. We report a patient with blastic phase chronic myeloid leukemia relapsing after allogeneic stem cell transplantation. The post-transplant leukemia was characterized with B-lymphoid markers and multiple genetic abnormalities including double Ph-chromosomes. The disease was treated with three courses of salvage chemotherapy combined with donor lymphocyte infusion and bcr-abl tyrosine kinase inhibitor. The leukemia proved to be non-responsive both to immune therapy and STI 571. The presented case demonstrates the need for combination approaches in post-transplant relapsed leukemia and discusses the possible contributing mechanisms of STI-571 resistance. C1 [Gopcsa, Laszlo] National Medical Center, Department of Immunology, H-1519 Budapest, Hungary. [Barta, Aniko] National Medical Center, Department of Immunology, H-1519 Budapest, Hungary. [Banyai, Aniko] National Medical Center, Department of Immunology, H-1519 Budapest, Hungary. [Dolgos, Janos] National Medical Center, Department of Immunology, H-1519 Budapest, Hungary. [Halm, Gabriella] National Medical Center, Department of Immunology, H-1519 Budapest, Hungary. [Paloczi, Katalin] National Medical Center, Department of Immunology, H-1519 Budapest, Hungary. RP Gopcsa, L (reprint author), National Medical Center, H-1519 Budapest, Hungary. EM k.paloczi@ohvi.hu CR Baron F, Frere P, Fillet G et al: Treatment of leukaemia relapse after allogeneic hematopoietic stem cell transplantation by donor lymphocyte infusion and STI 571, Haematologica 86: 993-994, 2001 Carlens S, Remberger M, Aschan I et al: The role of disease stage in the response to donor lymphocyte infusions as treatment for leukemic relapse. Biol Blood Marrow Transplant 7: 31-38, 2001 Druker BJ, Sawyers CL, Kantarjian H et al: Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. New Engl J Med 344:1038- 1042, 2001 Elmaagacli AH, Beelen DW and Schaefer UW: A retrospective single centre study of the outcome of five different therapy approaches in 48 patients with relapse of chronic myelogenous leukemia after allogeneic bone marrow transplantation. Bone Marrow Transplant 20: 1045-1055, 1997 Gopcsa L, Barta A, Banyai A et al: Acute myeloid leukaemia of donor cell origin developing five years after allogeneic bone marrow transplantation for chronic myeloid leukaemia. Bone Marrow Transplant 29:449-452, 2002 Kantarjian H, Melo JV, Tura S et al: Chronic myelogenous leukemia: disease biology and current and future therapeutic strategies. Hematology, Am Soc Hematol Educ Program, 90- 109, 2000 Mughal TI and Goldman JM: Chronic myeloid leukaemia: STI 571 magnifies the therapeutic dilemma. Eur J Cancer 37:561- 568, 2001 Thomas ED and Clift RA: Allogeneic transplantation for chronic myeloid leukemia. In: Hematopoietic cell transplantation., Eds: Thomas ED, Blume KG and Forman SJ, Blackwell Scientific Publications, Malden, 1999, pp. 807-816 Visani G, Rosti G, Bandini G et al: Second chronic phase before transplantation is crucial for improving survival of blastic phase chronic myeloid leukaemia. Br J Haematol 109: 722-728, 2000 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2003 VL 9 IS 2 BP 131 EP 133 PG 3 ER PT J AU Halbauer, JD Meszaros, I Doczi, T Kajtar, P Pajor, L Kovacs, K Gomori, AF Halbauer, J D Meszaros, Istvan Doczi, Tamas Kajtar, Pal Pajor, Laszlo Kovacs, Krisztina Gomori, Eva TI Rare Sellar Region Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE intracranial midline tumor; pituitary adenoma; granular cell tumor; germinoma ID intracranial midline tumor; pituitary adenoma; granular cell tumor; germinoma AB We present three cases of rare intracranial midline tumor in the sellar region, often mimiking pituitary adenomas clinically. We describe their symptoms, radiological and pathomorphological features. The first case is a pituitary adenoma producing growth hormone with ganglion cell differentiation. In addition, a rare intracranial granular cell tumor of sellar region and germinoma of pituitary fossa are also presented. All tumors were resected and histologically analyzed. Their biological behaviour was favorable with a 10-year follow-up demonstrating no recurrent tumor mass. C1 [Halbauer, J D] University of Pecs, Department of Pathology, 12. Szigeti u., H- 7643 Pecs, Hungary. [Meszaros, Istvan] Pecsi Tudomanyegyetem, Idegsebeszeti KlinikaPecs, Hungary. [Doczi, Tamas] Pecsi Tudomanyegyetem, Idegsebeszeti KlinikaPecs, Hungary. [Kajtar, Pal] University of Pecs, Department of PediatricsPecs, Hungary. [Pajor, Laszlo] University of Pecs, Department of Pathology, 12. Szigeti u., H- 7643 Pecs, Hungary. [Kovacs, Krisztina] University of Pecs, Department of Pathology, 12. Szigeti u., H- 7643 Pecs, Hungary. [Gomori, Eva] University of Pecs, Department of Pathology, 12. Szigeti u., H- 7643 Pecs, Hungary. RP Gomori, (reprint author), University of Pecs, Department of Pathology, H- 7643 Pecs, Hungary. EM geva@pathology.pote.hu CR Albrecht S, Armstrong DL, Mahoney DH et al: Cytogenetic demonstration of gene amplification in a primary intracranial germ cell tumor. Genes Chromosome Cancer 6: 61-63, 1993. Asa SL, Kovacs K, Horvath E: Human fetal adenohypophysis. Electronmicroscopic and ultrastructural immunocytochemical analysis. Neuroendocrinology 48: 423-431, 1988. Barrande G, Kujas M, Gancel A et al: Granular cell tumors. Rare tumors of the neurohypophysis. Press Med 24: 1676-1680, 1995. Becker DH, Wilson CB: Symptomatic parasellar granular cell tumors. Neurosurgery 8: 173-179, 1981. Dickson DV, Suzuki KI, Kanner R et al: Cerebral granular cell tumor: immunohistochemical and electron microscopic study. J Neuropathol Exp Neurol 45: 304-314, 1986. Geddes JF, Jansen GH, Robinson SFD et al: Gangliocytoma of the pituitary: a heterogeneous group of lesions with differing histogenesis. Am J Surg Path 24: 607-613, 2000. Giangaspero F, Cennachi G: Oncocytic and granular cell neoplasms of the central nervous system and pituitary gland. Semin Diagn Pathol 16: 91-97, 1999. Gomori E, Halbauer DJ, Doczi T et al: Cytogenetic profile of primary pituitary germinoma. J Neuro-Oncol 50: 251-255, 2000. Horvath E, Kovacs K, Scheithauer BW et al: Pituitary adenoma with neuronal choristoma, PANCH): composite lesion or lineage infidelity? Ultrastruct Pathol 18: 565-574, 1994. Iwase T, Nishizawa S, Baba S: Intrasellar neuronal choristoma associated with growth hormone-producing pituitary adenoma containing amyloid deposits. Hum Pathol 26: 925-928, 1995. Kohler L: Case of the month. April 1996-pituitary tumor. Brain Pathol 6: 531-532, 1996. Matsutani M, Sano K, Takakura K et al: Primary intracranial germ cell tumors: a clinical analysis of 153 histologically verified cases. J Neurosurg 86: 446-455, 1997. Nakashima T, Nishimura Y, Sakai N et al: Germinoma in cerebral hemisphere associated with Down syndrome. Childs Nerv Syst 13: 563-566, 1997. Puchner MJ, Ludecke DK, Saeger W et al: Gangliocytomas of the sellar region – a rewiev. Exp Clin Endocrinol Diabetes 103: 129-149, 1995. Rickert CH, Kuchelmeister K, Gillotta F: Morphological and immunohistochemical characterisation of granular cell in nonhypophyseal tumors of the central nervous system. Histopathology 30: 464-471, 1997. Sainati L, Bolcato S, Montaldi A et al: Cytogenetics of pediatric central nervous system tumors. Cancer Genet Cytogenet 91: 13-27, 1996. Tomita T, Gates E: Pituitary adenoma and granular cell tumor. Incidence, cell type and location of tumor in 100 pituitary glands at autopsy. Am J Clin Pathol 111: 817-825, 1999. Towfighi J, Salam MM, McLendon RE et al:Ganglion cell-containing tumors of the pituitary gland. Arch Pathol Lab Med 120: 369-377, 1996. Wong TT, Ho DM, Chang TK et al: Familial neurofibromatosis 1 with germinoma involving the basal ganglion and thalamus. Childs Nerv Syst 11: 456-458, 1995. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2003 VL 9 IS 2 BP 134 EP 137 PG 4 ER PT J AU Marchetti, D Denkins, Y Reiland, J Greiter-Wilke, A Galjour, J Murry, B Blust, J Roy, M AF Marchetti, Dario Denkins, Yvonne Reiland, Jane Greiter-Wilke, Andrea Galjour, Jennifer Murry, Brian Blust, Jason Roy, Madhuchhanda TI Brain-Metastatic Melanoma: a Neurotrophic Perspective SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE brain metastasis; malignant melanoma; neurotrophins; neurotrophin receptors; p75NTR; heparanase; astrocytes ID brain metastasis; malignant melanoma; neurotrophins; neurotrophin receptors; p75NTR; heparanase; astrocytes AB The brain is a unique microenvironment enclosed by the skull and maintaining a highly regulated vascular transport barrier. To metastasize to the brain, malignant tumor cells must attach to microvessel endothelial cells, invade the blood-brain barrier (BBB), and respond to brain survival and growth factors. Neurotrophins (NT) are important in brain invasion because they stimulate this process. In brain-metastatic melanoma cells, NT can promote invasion by enhancing the production of extracellular matrixdegradative enzymes such as heparanase, an enzyme capable of locally destroying both the extracellular matrix and the basement membrane of the BBB. We have examined human and murine melanoma cell lines exhibiting varying abilities to form brain metastases, and have found that they express low-affinity neurotrophin receptor p75NTR in relation to their brain-metastatic potentials. They do not, however, express trkA, the gene encoding the tyrosine kinase receptor TrkA, the high-affinity receptor for nerve growth factor (NGF), the prototypic NT. Presence of functional TrkC, the putative receptor for the invasion-promoting neurotrophin NT-3, was also expressed in these cells. Brain-metastatic melanoma cells can also produce autocrine factors and inhibitors that influence their growth, invasion, and survival in the brain. Synthesis of these factors may influence NT production by brain cells adjacent to the neoplastic invasion front, such as oligodendrocytes and astrocytes. In brain biopsies, we observed increased amounts of NGF and NT-3 in tumor-adjacent tissues at the invasion front of human melanoma tumors. Additionally, we found that astrocytes contribute to the brain-metastatic specificity of melanoma cells by producing NT-regulated heparanase. Trophic, autocrine, and paracrine growth factors may therefore determine whether metastatic cells can successfully invade, colonize, and grow in the central nervous system (CNS). C1 [Marchetti, Dario] Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, LA 70803 Baton Rouge, USA. [Denkins, Yvonne] Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, LA 70803 Baton Rouge, USA. [Reiland, Jane] Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, LA 70803 Baton Rouge, USA. [Greiter-Wilke, Andrea] Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, LA 70803 Baton Rouge, USA. [Galjour, Jennifer] Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, LA 70803 Baton Rouge, USA. [Murry, Brian] Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, LA 70803 Baton Rouge, USA. [Blust, Jason] Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, LA 70803 Baton Rouge, USA. [Roy, Madhuchhanda] Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, LA 70803 Baton Rouge, USA. RP Marchetti, D (reprint author), Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, LA 70803 Baton Rouge, USA. EM dmarchetti@vetmed.lsu.edu CR Prados M, Wilson C: Neoplasms of the central nervous system. In: Holland, J.F., Frei, III.E., Bast, Jr.R.C., Kufe, D.W., Morton, D.L., Weischselbaum, R.R., Eds.), Cancer Medicine. Philadelphia: Lea & Febiger, pp. 1080-1119, 1993. Sawaya R, Ligon, BL, Bindal, AK, et al.: Surgical treatment of metastatic brain tumors. J. Neurooncol 27: 269-277, 1996. Soffietti R, Ruda, R, and Mutani, R. Management of brain metastases. J Neurol. 249: 1357-1369, 2002. Steck, P, and Nicolson G: Metastasis to the central nervous system. In: Levine, A., Schmidek, H., Eds.), Molecular Genetics of Nervous System Tumors. New York: Wiley and Sons, pp. 371-379, 1993 Fidler IJ: The pathogenesis of cancer metastasis: the “seed and soil” hypothesis revisited. Nature Rew Cancer 3: 1-6, 2003. Nicolson GL, Menter DG, Herrmann, JL, et al.: Brain metastasis: role of trophic, autocrine, and paracrine factors in tumor invasion and colonization of the central nervous system. Curr Top Microbiol Immunol 213: 89-115, 1996. Yano S, Shinohara H, Herbst RS, et al.: Expression of vascular endothelial growth factor is necessary but not sufficient for production and growth of brain metastasis. Cancer Res 60: 4959-4967, 2000. Albino AP, Davis BM, and Nanus DM: Induction of growth factor RNA expression in human malignant melanoma: markers of transformation. Cancer Res 51: 4815-4820, 1991. Herlyn M, Thurin J, Balaban G, et al.: Characteristics of cultured human melanocytes isolated from different stages of tumor progression. Cancer Res 45: 5670-5676, 1985. Bradshaw RA, Blundell TL, Lapatto R, et al.: Nerve growth factor revisited. Trends Biochem Sci 18: 48-52, 1993. Lee R, Kermani P, Teng KK, and Hempstead BL: Regulation of cell survival by secreted proneurotrophins. Science 294: 1945-1948, 2001. Raff MC: Social controls on cell survival and cell death. Nature 356: 397-400, 1992. Raff MC, Barres BA, Burne, JF, et al.: Programmed cell death and the control of cell survival: lessons from the nervous system. Science 262: 695-700, 1993. Snider WD: Functions of the neurotrophins during nervous system development: what the knockouts are teaching us. Cell 77, 627-638, 1994. Jones KR, Farinas I, Backus C, and Reichardt LF: Targeted disruption of the BDNF gene perturbs brain and sensory neuron development but not motor neuron development. Cell 76: 989-999, 1994. DiCicco-Bloom E, Friedman, WJ, and Black IB: NT-3 stimulates sympathetic neuroblast proliferation by promoting precursor survival. Neuron 11: 1101-1111, 1993. Chao MV, and Bothwell M: Neurotrophins: to cleave or not to cleave. Neuron 33: 9-12, 2002. Johnson D, Lanahan, A, Buck, CR, et al.: Expression and structure of the human NGF receptor. Cell 47: 545-554, 1986. Maher PA: Nerve growth factor induces protein-tyrosine phosphorylation. Proc Natl Acad Sci U.S.A. 85: 6788-6791, 1988. Miyasaka T, Chao, MV, Sherline P, and Saltiel AR: Nerve growth factor stimulates a protein kinase in PC-12 cells that phosphorylates microtubule-associated protein-2. J Biol Chem 265:4730-4735, 1990. Ohmichi M, Decker SJ, and Saltiel AR: Nerve growth factor stimulates the tyrosine phosphorylation of a 38-kDa protein that specifically associates with the src homology domain of phospholipase C-gamma 1. J Biol Chem 267:21601-21606, 1992. Barbacid M: Nerve growth factor: a tale of two receptors. Oncogene 8: 2033-2042, 1993. Chao MV: Neurotrophins and their receptors: a convergence point for many signalling pathways. Nature Rev 4: 299-309, 2003. Meakin SO, and Shooter EM: The nerve growth factor family of receptors. Trends Neurosci 15: 323-331, 1992. Saltiel AR, and Decker SJ: Cellular mechanisms of signal transduction for neurotrophins. Bioassays 16: 405-411, 1994. Birren SJ, Lo L, and Anderson DJ: Sympathetic neuroblasts undergo a developmental switch in trophic dependence. Development 119: 597-610, 1993. Kalcheim C, Carmeli C, and Rosenthal A: Neurotrophin-3 is a mitogen for cultured neural crest cells. Proc Natl Acad Sci U.S.A. 89: 1661-1665, 1992. Schnell L, Schneider R, Kolbeck R, et al.: Neurotrophin-3 enhances sprouting of corticospinal tract during development and after adult spinal cord lesion. Nature 367: 170-173, 1994. Ernfors P, Lee KF, Kucera J, and Jaenisch R: Lack of neurotrophin- 3 leads to deficiencies in the peripheral nervous system and loss of limb proprioceptive afferents. Cell 77: 503- 512, 1994. Klein R, Silos-Santiago I, Smeyne RJ, et al.: Disruption of the neurotrophin-3 receptor gene trkC eliminates la muscle afferents and results in abnormal movements. Nature 368:249-251, 1994. Ernfors P, Lee KF, and Jaenisch R: Mice lacking brainderived neurotrophic factor develop with sensory deficits. Nature 368: 147-150, 1994. Klein R, Smeyne RJ, Wurst W, et al.: Targeted disruption of the trkB neurotrophin receptor gene results in nervous system lesions and neonatal death. Cell 75: 113-122, 1993. Buchman VL, and Davies AM: Different neurotrophins are expressed and act in a developmental sequence to promote the survival of embryonic sensory neurons. Development 118: 989-1001, 1993. Crowley C, Spencer SD, Nishimura MC, et al.: Mice lacking nerve growth factor display perinatal loss of sensory and sympathetic neurons yet develop basal forebrain cholinergic neurons. Cell 76:1001-1011, 1994. Smeyne RJ, Klein R, Schnapp A, et al.: Severe sensory and sympathetic neuropathies in mice carrying a disrupted Trk/NGF receptor gene. Nature 368: 246-249, 1994. Peacocke M, Yaar M, Mansur CP, et al.: Induction of nerve growth factor receptors on cultured human melanocytes. Proc Natl Acad Sci U.S.A. 85: 5282-5286, 1988. Yaar M, Eller MS, DiBenedetto P, et al.: The trk family of receptors mediates nerve growth factor and neurotrophin-3 effects in melanocytes. J Clin Invest 94: 1550-1562, 1994. Yaar M, and Gilchrest BA:Human melanocyte growth and differentiation: a decade of new data. J Invest Dermatol 97: 611- 617, 1991. Herrmann JL, Menter DG, Hamada J, et al.: Mediation of NGF-stimulated extracellular matrix invasion by the human melanoma low-affinity p75 neurotrophin receptor: melanoma p75 functions independently of trkA. Mol Biol Cell 4: 1205- 1216, 1993. Marchetti D, Murry B, Galjour J, and Wilke-Greiter A: Human melanoma TrkC: Its association with a purine-analogsensitive kinase activity. J Cell Biochem 88: 865-872, 2003. Bibel M, Hoppe E, and Barde, YA: Biochemical and functional interactions between the neurotrophin receptors Trk and p75NTR. EMBO J 18: 616-622, 1999. Marchetti D, McQuillan DJ, Spohn WC, et al.: Neurotrophin stimulation of human melanoma cell invasion: selected enhancement of heparanase activity and heparanase degradation of specific heparan sulfate subpopulations. Cancer Res 56: 2856-2863, 1996. Marchetti D, and Nicolson GL: Human melanoma cell invasion: selected neurotrophin enhancement of invasion and heparanase activity. J Invest Dermatol Symp Proc 2: 99-105, 1997. Marchetti D, and Nicolson GL: Neurotrophin stimulation of human melanoma cell invasion: selected enhancement of heparanase activity and heparanase degradation of specific heparan sulfate subpopulations. Adv Enzyme Reg 37: 111- 134, 1997. Nicolson GL, Menter DG, Herrmann J, et al.: Tumor metastasis to brain: role of endothelial cells, neurotrophins, and paracrine growth factors. Crit Rev Oncol 5: 451-471, 1994. Hempstead BL, Martin-Zanca D, Kaplan DR, Parada LF, and Chao MV: High-affinity NGF binding requires coexpression of the trk proto-oncogene and the low-affinity NGF receptor. Nature 350: 678-683, 1991. Lee KF, Li E, Huber LJ, Landis SC, et al.: Targeted mutation of the gene encoding the low affinity NGF receptor p75 leads to deficits in the peripheral sensory nervous system. Cell 69: 737-749, 1992. Lee KF, Bachman K, Landis S, and Jaenisch R: Dependence on p75NTR for innervation of some sympathetic targets. Science 263: 1447-1449, 1994. Verdi JM, Birren SJ, Ibanez CF, et al.: p75LNGFR regulates Trk signal transduction and NGF-induced neuronal differentiation in MAH cells. Neuron 12: 733-745, 1994. Ohmichi M, Decker SJ, Pang L, and Saltiel AR:Phospholipase C-gamma 1 directly associates with the p70 trk oncogene product through its src homology domains. J Biol Chem 266: 14858-14861, 1991. Ohmichi M, Decker SJ, and Saltiel AR: Activation of phosphatidylinositol- 3 kinase by nerve growth factor involves indirect coupling of the trk proto-oncogene with src homology 2 domains. Neuron 9: 769-777, 1992. Avruch J, Zhang XF, and Kyriakis JM: Raf meets Ras: completing the framework of a signal transduction pathway. Trends Biochem Sci 19:279-283, 1994. Batistatou A, Volonte C, and Greene LA: Nerve growth factor employs multiple pathways to induce primary response genes in PC12 cells. Mol Biol Cell 3: 363-371, 1992. Borrello MG, Pelicci G, Arighi E, et al.: The oncogenic versions of the Ret and Trk tyrosine kinases bind Shc and Grb2 adaptor proteins. Oncogene 9: 1661-1668, 1994. Lange-Carter CA, and Johnson GL: Ras-dependent growth factor regulation of MEK kinase in PC12 cells. Science 265: 1458-1461, 1994. Obermeier A, Lammers R, Wiesmuller KH, et al: Identification of Trk binding sites for SHC and phosphatidylinositol 3- kinase and formation of a multimeric signaling complex. J Biol Chem 268: 22963-22966, 1993. Obermeier A, Halfter H, Wiesmuller KH, et al.: Tyrosine 785 is a major determinant of Trk-substrate interaction. EMBO J 12: 933-941, 1993. Obermeier A, Bradshaw RA, Seedorf K, et al.: Neuronal differentiation signals are controlled by nerve growth factor receptor/Trk binding sites for SHC and PLC gamma. EMBO J 13: 1585-1590, 1994. Ohmichi M, Matuoka K, Takenawa T, and Saltiel AR: Growth factors differentially stimulate the phosphorylation of Shc proteins and their association with Grb2 in PC-12 pheochromocytoma cells. J Biol Chem 269:1143-1148, 1994. Rozakis-Adcock M, McGlade J, Mbamalu G, et al.: Association of the Shc and Grb2/Sem5 SH2-containing proteins is implicated in activation of the Ras pathway by tyrosine kinases. Nature 360: 689-692, 1992. Satoh T, Nakafuku M, and Kaziro Y: Function of Ras as a molecular switch in signal transduction. J Biol Chem 267: 24149- 24152, 1992. Stephens RM, Loeb DM, Copeland TD, et al.: Trk receptors use redundant signal transduction pathways involving SHC and PLC-gamma 1 to mediate NGF responses. Neuron 12: 691-705, 1994. Taylor LK, Swanson KD, Kerigan J, et al.: Isolation and characterization of a nerve growth factor-regulated Fos kinase from PC12 cells. J Biol Chem 269: 308-318, 1994. Kaplan DR, and Miller, FD: Neurotrophin signal transduction in the nervous system. Curr Opin Neurobiol 10: 381-391, 2000. Berg MM, Sternberg DW, Hempstead BL, and Chao MV: The low-affinity p75 nerve growth factor, NGF, receptor mediates NGF-induced tyrosine phosphorylation. Proc. Natl. Acad. Sci. U. S. A. 88, 7106-7110, 1991. Hempstead BL, Schleifer LS, and Chao MV: Expression of functional nerve growth factor receptors after gene transfer. Science 243, 373-375, 1989. von Bartheld CS, Kinoshita Y, Prevette D, et al.: Positive and negative effects of neurotrophins on the isthmo-optic nucleus in chick embryos. Neuron 12:639-654, 1994. Rabizadeh S, Oh J, Zhong, LT, Yang J, et al.: Induction of apoptosis by the low-affinity NGF receptor. Science 261: 345-348, 1993. Kannan Y, Usami K, Okada M, et al.: Nerve growth factor suppresses apoptosis of murine neutrophils. Biochem Biophys Res Commun 186: 1050-1056, 1992. Feinstein DL, and Larhammar D: Identification of a conserved protein motif in a group of growth factor receptors. FEBS Lett 272: 7-11, 1990. Knipper M, Beck A, Rylett J, and Breer H: Neurotrophin induced cAMP and IP3 responses in PC12 cells: different pathways. FEBS Lett 32: 147-152, 1993. Hantzopoulos PA, Suri C, Glass DJ, et al.: The low-affinity NGF receptor, p75, can collaborate with each of the Trks to potentiate functional responses to the neurotrophins. Neuron 13: 187-201, 1994. Hempstead BL, Patil N, Thiel B, and Chao, MV: Deletion of cytoplasmic sequences of the nerve growth factor receptor leads to loss of high-affinity ligand binding. J Biol Chem 265: 9595-9598, 1990. Beutler B, and van Huffel C: Unraveling function in the TNF ligand and receptor families. Science 264: 667-668, 1994. Smith CA, Farrah T, and Goodwin RG: The TNF receptor superfamily of cellular and viral proteins: activation, costimulation, and death. Cell 76: 959-962, 1994. Barrett GL, and Bartlett PF: The p75 nerve growth factor receptor mediates survival or death depending on the stage of sensory neuron development. Proc Natl Acad Sci U.S.A. 91: 6501-6505, 1994. Volonte’ C, Ross AH, and Greene LA: Association of a purineanalogue- sensitive protein kinase activity with p75 nerve growth factor receptors. Mol. Biol Cell 4: 71-78, 1993. Volonte’ C, and Greene LA: Nerve growth factor-activated protein kinase N. Characterization and rapid near homogeneity purification by nucleotide affinity-exchange chromatography. J Biol Chem 267: 21663-21670, 1992. Dobrowsky RT, Werner MH, Castellino AM, Chao MV, and Hannun YA: Activation of the sphingomyelin cycle through the low-affinity neurotrophin receptor. Science 265: 1596- 1599, 1994. Wolff RA, Dobrowsky RT, Bielawska A, Obeid LM, and Hannun YA: Role of ceramide-activated protein phosphatase in ceramide-mediated signal transduction. J Biol Chem 269: 19605-19609, 1994. Ross AH, Grob P, Bothwell M, et al.: Characterization of nerve growth factor receptor in neural crest tumors using monoclonal antibodies. Proc Natl Acad Sci U.S.A. 81: 6681-6685, 1984. Morse HG, Gonzalez R, Moore GE, and Robinson WA: Preferential chromosome 11q and/or 17q aberrations in short-term cultures of metastatic melanoma in resections from human brain. Cancer Genet Cytogenet 64: 118-126, 1992. Ishikawa M, Dennis JW, Man S, and Kerbel RS: Isolation and characterization of spontaneous wheat germ agglutinin-resistant human melanoma mutants displaying remarkably different metastatic profiles in nude mice. Cancer Res 48: 665-670, 1988. Marchetti D, Menter D, Jin L, et al.: Nerve growth factor effects on human and mouse melanoma cell invasion and heparanase production. Int J Cancer 55: 692-699, 1993. Gladson CL, Wilcox JN, Sanders L: Cerebral microenvironment influence et al.: expression of the vitronectin gene in astrocytic tumors. J Cell Sci 108: 947-956, 1995. Liotta LA, Steeg PS, and Stetler-Stevenson WG:Cancer metastasis and angiogenesis: an imbalance of positive and negative regulation. Cell 64, 327-336, 1991. Liotta LA, Thorgeirsson UP, and Garbisa S: Role of collagenases in tumor cell invasion. Cancer Metastasis Rev 1: 277- 288, 1982. Powell WC, and Matrisian LM: Complex roles of matrix metalloproteinases in tumor progression. Curr Top Microbiol Immunol 213: 1-21, 1996. Sloane BF, and Honn KV, Cysteine proteinases and metastasis. Cancer Metastasis Rev 3: 249-263, 1984. Timar J, Lapis K, Dudas J, et al.: Proteoglycans and tumor progression: Janus-faced molecules with contradictory functions in cancer. Semin Cancer Biol 12: 173-186, 2002. Bernfield M, Gotte M, Park PW, et al.: Functions of cell surface heparan sulfate proteoglycans. Ann Rev Biochem 68: 729-777, 1999. Iozzo RV: Heparan sulfate proteoglycans: intricate molecules with intriguing functions. J Clin Invest 108. 165-167, 2001. McKeehan WL, and Kan M: Heparan sulfate fibroblast growth factor receptor complex: structure-function relationships. Mol Reprod Dev 39: 69-81, 1994. Yanagishita M, and Hascall VC: Cell surface heparan sulfate proteoglycans. J Biol Chem 267: 9451-9454, 1992. Vlodavsky I, and Friedmann Y: Molecular properties and involvement of heparanase in cancer metastasis and angiogenesis. J Clin Invest 108: 341-347, 2001. Marchetti D, Liu S, Spohn WC, and Carson DD: Heparanase and a synthetic peptide of heparan sulfate-interacting protein recognize common sites on cell surface and extracellular matrix heparan sulfate. J Biol Chem 272: 15891-15897, 1997. Nakajima M, Irimura T, and Nicolson GL: Heparanases and tumor metastasis. J Cell Biochem 36: 157-167, 1988. Nakajima M, Irimura T, and Nicolson GL: A solid-phase substrate of heparanase: its application to assay of human melanoma for heparan sulfate degradative activity. Anal Biochem 157: 162-171, 1986. Nakajima M, Irimura T, and Nicolson GL: Tumor metastasisassociated heparanase, heparan sulfate endoglycosidase, activity in human melanoma cells. Cancer Lett 31: 277-283, 1986. Vlodavsky I, Friedmann Y, Elkin M, et al.: Mammalian heparanase: gene cloning, expression and function in tumor progression and metastasis. Nature Med 5: 793-802, 1999. Hulett MD, Freeman C, Hamdorf BJ, et al.: Cloning of mammalian heparanase, an important enzyme in tumor invasion and metastasis. Nature Med 5: 803-809, 1999. Kussie PH, Hulmes JD, Ludwig DL, et al.: Cloning and functional expression of a human heparanase gene. Biochem Biophys Res Comm 261: 183-187, 1999. Toyoshima M, and Nakajima M: Human heparanase. Purification, characterization, cloning, and expression. J Biol Chem 274: 24153-24160, 1999. Folkman J: Angiogenesis-dependent diseases. Semin Oncol 28: 536-542, 2001. Aviezer D, Iozzo RV, Noonan DM, and Yayon A: Suppression of autocrine and paracrine functions of basic fibroblast growth factor by stable expression of perlecan antisense cDNA. Mol Cell Biol 17: 1938-1946, 1997. Rodeck U, Becker D, and Herlyn M: Basic fibroblast growth factor in human melanoma. Cancer Cells 3: 308-311, 1991. Gospodarowicz D, and Cheng J: Heparin protects basic and acidic FGF from inactivation. J Cell Physiol 128: 475-484, 1986. Marchetti D, Reiland J, Erwin B, and Roy M: Inhibition of heparanase activity and heparanase-induced angiogenesis by suramin analogues. Int J Cancer 104: 167-174, 2003. Menter DG, Herrmann JL, Marchetti D, and Nicolson GL: Involvement of neurotrophins and growth factors in brain metastasis formation. Invasion Metastasis 14: 372-384, 1994. Marchetti D, McCutcheon I, Ross JM, and Nicolson GL: Inverse expression of neurotrophin receptor and at the invasion front of brain-metastatic human melanoma tissues. Int J Oncol 7: 87-94, 1995. Yoshida K, and Gage FH: Cooperative regulation of nerve growth factor synthesis and secretion in fibroblasts and astrocytes by fibroblast growth factor and other cytokines. Brain Res 569: 14-25, 1992. McCarthy KD, and de Vellis J: Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. J Cell Biol 85: 890-902, 1980. Marchetti D, Li J, and Shen R: Astrocytes contribute to the brain-metastatic specificity of melanoma cells by producing heparanase. Cancer Res 60: 4767-4770, 2000. Marchetti D, and Nicolson GL: Human heparanase: a molecular determinant of brain metastasis. Adv Enzyme Reg 41: 343-359, 2001. Norenberg MD: Astrocyte responses to CNS injury. J Neuropathol Exp Neurol 53: 213-220, 1994. Kettenmann H, Orkand RK, and Schachner M: Coupling among identified cells in mammalian nervous system cultures. J Neurosci 3: 506-516, 1983. Wilkin GP, Marriott DR, and Cholewinski AJ: Astrocyte heterogeneity. Trends Neurosci 13: 43-46, 1990. Hirano A, Kawanami T, and Llena JF: Electron microscopy of the blood-brain barrier in disease. Microsc Res Tech 27: 543- 556, 1994. Kimelberg HK, and Ransom BR: Physiological aspects of astrocyte swelling. In: Fedoroff, S., Verandakis, A., Eds.), Astrocytes. Orlando: Academic Press, pp. 129-166, 1986 Lantos PL, Luthert PJ, and Deane BR: Vascular permeability and cerebral edema in experimental brain tumors. In: Inaba, Y., Klatzo, I., Spatz, I., Eds.), Brain Edema, New York: Springer-Verlag, pp. 40-47, 1984 Klatzo I, Chui E, Fujiwara K, and Spatz M: Resolution of vasogenic brain edema. Adv Neurol 28: 359-373, 1980. Walch ET, Albino AP, and Marchetti D: Correlation of overexpression of the low-affinity p75 neurotrophin receptor with augmented invasion and heparanase production in human malignant melanoma cells. Int J Cancer 82: 112-120, 1999. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 147 EP 158 PG 12 ER PT J AU Makar, RR Saji, T Junaid, T AF Makar, Ray Ragai Saji, Tessy Junaid, A. Tola TI Epstein-Barr Virus Expression in Hodgkin's Lymphoma in Kuwait SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hodgkin's lymphoma; Epstein Barr virus; in situ hybridization; Epstein Barr virus encoded RNA; immunohistochemistry; Epstein Barr virus latent membrane protein 1 ID Hodgkin's lymphoma; Epstein Barr virus; in situ hybridization; Epstein Barr virus encoded RNA; immunohistochemistry; Epstein Barr virus latent membrane protein 1 AB The epidemiology of Hodgkin's lymphoma (HL) shows wide geographic variation in histological subtypes and in its association with the Epstein-Barr virus (EBV). The proportion of EBV positive HL is low in industrialized countries, high in non-industrialized countries and intermediate in early-industrialized countries. Reports from the Persian Gulf and Middle East are very limited. The aim of this study was to determine the epidemiology of HL in Kuwait, an early-industrialized country in the Persian Gulf, and to delineate the extent of its association with EBV. We reviewed 134 cases of HL for histological classification and demographic data. 107 cases were examined for the presence of EBV using immunohistochemistry (IHC) for the latent membrane protein I (LMPI) and in-situ hybridization (ISH) for EBVencoded RNA (EBER). 70.4% of the patients were males and 29.6% were females. The male: female ratio was 2.4:1. The mean age was 30.6 years (range, 4-71 years). Mixed cellularity HL (MCHL) was the most common subtype (45.5%), followed by nodular sclerosis (37.3%), nodular lymphocyte predominant (6.7%), lymphocyte rich (3%) and lymphocyte depletion (3%). 4.5% of cases were unclassifiable. EBV expression was seen in 56%, was significantly higher in MCHL, in children, and in males. Our findings suggest that the frequency of EBV expression in HL in Kuwait is similar to other early-industrialized countries. Further research from other countries in the Persian Gulf and the Middle East should shed more light on the epidemiology of HL and its relation to EBV in this region. C1 [Makar, Ray Ragai] Faculty of Medicine, Kuwait University, Department of Pathology, 1330 SW Third Avenue #1210, OR 97201 Portland, OR, USA. [Saji, Tessy] Faculty of Medicine, Kuwait University, Department of Pathology, 1330 SW Third Avenue #1210, OR 97201 Portland, OR, USA. [Junaid, A. Tola] Faculty of Medicine, Kuwait University, Department of Pathology, 1330 SW Third Avenue #1210, OR 97201 Portland, OR, USA. RP Makar, RR (reprint author), Faculty of Medicine, Kuwait University, Department of Pathology, OR 97201 Portland, USA. EM rosymakar@aol.com CR A1-Idrissi HY, Ibrahim EM: Hodgkin’s disease in adults in Saudi Arabia. Clinical features, prognostic factors and an analysis of therapy. Outcome of combination chemotherapy only for both localized and advanced disease. Int J Cancer 47: 822-826, 1991. Azzam SA: High Incidence of Hodgkin’s disease in children in Lebanon. Cancer Research 26: 1202-1203, 1966. Bamanikar S, Thunold S, Devi KR, Bamanikar A: The pattern of malignant lymphoma in Oman. J Trop Med Hyg 98: 351- 354, 1995. Benharroch D, Brousset P, Goldstein J, et al: Association of the Epstein-Barr virus with Hodgkin’s disease in Southern Israel. Int J Cancer 71: 138-141, 1997. Chang KL, Albujar PF, Chen YY, Johnson RM, Weiss LM: High prevalence of Epstein-Barr virus in the Reed-Sternberg cells of Hodgkin’s disease occurring in Peru. Blood 81: 496-501, 1993. Enblad G, Sandvej K, Sundstrom C, et al: Epstein-Barr virus distribution in Hodgkin’s disease in an unselected Swedish population. Acta Oncologica 38: 425-429, 1999. Flavell KJ, Murray PG. Hodgkin’s disease and the Epstein- Barr virus: J Clin Pathol: Mol Pathol 53: 262-269, 2000. Gad-El-Mawla N, El-Deeb BB, Abu-Gabal A, et al: Pediatric Hodgkin’s Disease in Egypt. Cancer 52: 1129-1131, 1983. Glaser SL, Lin RJ, Stewart SL, et al: Epstein-Barr virus-associated Hodgkin’s disease: epidemiologic characteristics in international data. Int J of Cancer 70: 375-382, 1997 Harris NL: The many faces of Hodgkin’s disease around the world: what have we learned from its pathology. Annals of Oncology 9(suppl5): S45-S56, 1998. Kandil A, Bazarbashi S, Mourad W: The correlation of Epstein- Barr virus expression and lymphocyte subsets with the clinical presentation of nodular sclerosing Hodgkin disease. Cancer 91: 1957-1963, 2001. Kawa K: Epstein-Barr virus – associated diseases in humans. Int J Hematol 71: 108-117, 2000. Khan G, Norton AJ, Slavin G: Epstein-Barr virus in Hodgkin Disease. Relation to age and subtype. Cancer 71: 3124-3129, 1993. Kusuda M, Toriyama K, Kamidigo NO, et al: A Comparison of epidemiologic, histologic and virologic studies on Hodgkin’s disease in Western Kenya and Nagasaki, Japan. Am J Trop Med Hyg 59: 801-807, 1998. Leoncini L, Spina D, Nyong A, et al: Neoplastic cells of Hodgkin’s disease show differences in EBV expression between Kenya and Italy. Int J Cancer 65: 781-784, 1996. Liu SM, Chow KC, Chiu CF, et al: Expression of Epstein-Barr virus in patients with Hodgkin’s disease in Taiwan. Cancer 83: 367-71, 1998. MacMahon B: Epidemiology of Hodgkin’s disease. Cancer Research 26: 1189-1200, 1966. Marafioti T, Hummel M, Foss HD, et al: Hodgkin and Reed- Sternberg cells represent an expansion of a single clone originating from a germinal center B-cell with functional immunoglobulin gene rearrangements but defective immunoglobulin transcription. Blood 95: 1443-1450, 2000. Morente MM, Piris MA, Abraira V, et al: Adverse clincial outcome in Hodgkin’s Disease is associated with loss of retinoblastoma protein expression, high Ki67 proliferation index, and absence of Epstein-Barr virus latent membrane protein 1 expression. Blood 90:2429-2436, 1997. Motawy MS, Omar YT: Hodgkin’s disease in children in Kuwait. Cancer 57:2255-2259, 1986. Mourad W, Alsohaibani MO, Saddik M, et al: Epsztein-Barr Virus Expression in Hodgkin’s disease: correlation with histologic subtypes and T and B lymphocyte distribution. Ann Saudi Med 18: 296-300, 1998. Mueller NE, Grufferman S: The Epidemiology of Hodgkin’s disease. In: Mauch PM, Armitage JD, Diehl V et al. Hodgkin’s Diseases, pp. 61-77. Philadelphia, USA: Lippincott Williams & Wilkins, 1999. Mughal TI, Robinson WA, Padmos MA: Adult Hodgkin’s disease in Saudi Arabia. Eur J Surg Oncol 11:41-45, 1985. Murray PG, Billingham LJ, Hassan HT, et al: Effect of Epstein-Barr virus Infection on Response to Chemotherapy and Survival in Hodgkin’s disease. Blood 94: 442-447, 1999. Niedobitek G, Meru N, Delecluse HJ: Epstein-Barr virus infection and human malignancies. Int J Exp Pathol 82: 149-170, 2001. Okano M: Epstein-Barr virus infection and its role in the expanding spectrum of human diseases. Acta Paediatr 87: 11- 18, 1998. Oudejans JJ, Jiwa NM, Meijer CJ. Editorial: Epstein-Barr virus in Hodgkin’s disease: more than just an innocent bystander. J Pathol 181: 353-356, 1997. Parkin DM, Whelan SL, Ferlay J, et al: Cancer Incidence in Five Continents Vol VII. International Agency for Research on Cancer, IARC, Press, Lyon, 1997. Peh SC, Looi LM, Pallesen G: Epstein-Barr virus, EBV, and Hodgkin’s disease in a multi-ethnic population in Malaysia. Histopathology 30: 227-233, 1997 Pileri SA, Ascani S, Leoncini L, et al.Hodgkin’s lymphoma: the pathologist’s viewpoint: J Clin Pathol 55: 162-176, 2002. Stein H, Delsol G, Pileri S, et al:Hodgkin Lymphoma. In: Jaffe ES, Harris NL, Stain H, Vardiman JW. Pathology and genetics of tumors of haematopoietic and lymphoid tissues. International Agency for Research on Cancer, IARC, Press, Lyon, 2001. pp. 23 7-253 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 159 EP 165 PG 7 ER PT J AU Boer, K Lang, I Juhos, Pinter, T Szanto, J AF Boer, Katalin Lang, Istvan Juhos, Eva Pinter, Tamas Szanto, Janos TI Adjuvant Therapy of Breast Cancer with Docetaxel-Containing Combination (TAC) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE early breast cancer; adjuvant chemotherapy; comparative study; docetaxel ID early breast cancer; adjuvant chemotherapy; comparative study; docetaxel AB The adjuvant chemotherapy of breast cancer changed in the past two decades. Docetaxel containing regimens are highly active in metastatic breast cancer. A logical approach was their incorporation into trials of early breast cancer adjuvant therapy. The authors present the Hungarian interim analysis and experience with the BCIRG 001 randomized, multicentric, phase III clinical trial comparing TAC (docetaxel, doxorubicin, cyclophosphamide) and FAC (5-fluorouracil, doxorubicin, cyclophosphamide) in the adjuvant treatment of node positive breast cancer patients. The results are presented compared to the international data. Three Hungarian centers - Szt. Margit Hospital, Budapest, National Institute of Oncology, Budapest, Petz Aladar Hospital, Gyor - participated in the international trial. Between June 1997 and June 1999, 61 patients with node positive breast cancer were enrolled in the study after the surgery. Thirty-four patients were randomized to TAC (75/50/500 mg/m2 6xq3wk) and 27 patients were randomized to FAC (500/50/500 mg/m2 6x q3wk) chemotherapy, with prospective stratification by node (1-3, 4+). Patients with hormone receptor positive tumors received tamoxifen for 5 years after the chemotherapy. Radiotherapy was performed after the 6th cycle of chemotherapy. 33 months of follow up was performed. In both arms the hematological toxicity was more frequent. The TAC group showed a higher incidence of neutropenia (76%) compared to the FAC (22%), as well as a higher incidence of febrile neutropenia (26 % versus none), without grade 3-4 infection and there was no cases of septic death. More grade 3-4 nausea and vomiting was observed in the FAC group. At three years follow up, results indicated improvement in disease-free survival (88% vs. 76%) in favour of TAC, and similar tendency was observed in the case of overall survival (97% vs. 88%). Based on the international data analysis TAC was superior to FAC chemotherapy, the results show statistically significant differences between the two arms. This benefit with TAC was seen regardless of hormone receptor status. Additional follow up data will evaluate the role of TAC in the adjuvant setting of early breast cancer treatment. C1 [Boer, Katalin] Szent Margit Hospital, V. Department of Internal Medicine - Oncology, Becsi ut 132, H-1032 Budapest, Hungary. [Lang, Istvan] National Institute of OncologyBudapest, Hungary. [Juhos, Eva] National Institute of OncologyBudapest, Hungary. [Pinter, Tamas] Petz Aladar County HospitalGyor, Hungary. [Szanto, Janos] Szent Margit Hospital, V. Department of Internal Medicine - Oncology, Becsi ut 132, H-1032 Budapest, Hungary. RP Boer, K (reprint author), Szent Margit Hospital, V. Department of Internal Medicine - Oncology, H-1032 Budapest, Hungary. EM kboer@freemail.hu CR Amadori D, Nanni O, Marangolo M et al: Disease-free survival advantage of adjuvant cyclophosphamide, methotrexate and fluorouracil in patients with node-negative rapidly proliferating breast cancer. A randomized multicenter study. J Clin Oncol 18: 3125-3134, 2000 Baki M, Rado J, Gyergyay F es mtsai: Tapasztalataink elorehaladott stadiumu emlodaganatos betegek Adriblastin, A, es Docetaxel, D, osszehasonlito vizsgalata soran. Magy Onkol Tars XXII. Nemzeti Kongr Magyar Onkol 41: 300-301, 1997, in Hungarian) Bonadonna G, Valagussa P: The contribution of medicine to the primary treatment of breast cancer. Cancer Res 48: 2314- 2324, 1998 Bonneterre, Roche H, Monnier A et al: Taxotere(TXT, versus 5-Fluorouracil + Navelbine, FUN, as second-line chemotherapy, CT, in patients, pts, with metastatic breast cancer, MBC). Proc Am Soc Clin Oncol 16:162a, 1997 Chan S, Friedrich K, Noel D et al: Prospective randomized trial of docetaxel versus doxorubicin in patients with metastatic breast cancer. J Clin Oncol 17: 2341-2354, 1999 Early Breast Cancer Trialists Collaborative Group: Polychemotherapy for early breast cancer: an overview of the randomized trials. Lancet. 352: 930-942, 1998 Early Breast Cancer Trialists Collaborative Group: 2000 analysis overview results. Fifth Meeting of the Early Breast Cancer Trialists Collaborative Group, Oxford, United Kingdom, September 21-23, 2000 Early Breast Cancer Trialists Collaborative Group: Tamoxifen for early breast cancer : an overview of the randomized trials. Lancet 351:1451-1507, 1998 Eckhardt S: A docetaxel hatasa a daganatokra. Orv Hetilap 139: 867-872, 1998, in Hungarian) Fumoleau P, Chevalier B, Kerbrat P et al: A multicentre phase II study of the efficacy and safety of docetaxel as first –line treatment of advanced breast cancer. A report of the Clinical Screening Group of the EORTC. Ann Oncol 7: 165- 171, 1996 Goldhirsch A, Glick JH, Gelber RD, Senn HJ: Highlights. International consensus panel on treatment of primary breast cancer. J Natl Cancer Inst 90: 1601-1608, 1998 Nabholtz JM, Mackey JR, Smylie M et al: Final results of phase II study of docetaxel, doxorubicin and cyclophosphamide, TAC, as 1st line chemotherapy in metastatic breast cancer patients. Breast Cancer Res Treat 50:227, 1998 Nabholtz JM, Pienkowski T, Mackey J et al: Phase III trial comparing TAC, docetaxel, doxorubicin, cyclophosphamide, with FAC, 5-fluorouracil, doxorubicine, cyclophosphamide, in the adjuvant treatment of node positive breast cancer, BC, patients: interim analysis of the BCIRG 001 study. Proc Am Soc Clin Oncol, abstract No 141, 21: 36a, 2002 Piccart MJ, Lohrisch C, Duchateau L et al: Taxanes in the adjuvant setting: why not yet ? J Natl Cancer Inst Monogr 30: 88-95, 2001 Sjostrom J, Mouridsen H, Pluzanska A et al: Taxotere, T, versus methotrexate-5 fluorouracil, MF, in patients with advanced anthracycline- resistant breast cancer. Proc Am Soc Clin Oncol 17:111a, 1998 Szanto J, Pinter T, Szanto J: Doxorubicin + docetaxel-, illetoleg doxorubicin + cyclophosphamid-kezelessel elerheto eredmenyek tavoli attetet ado emlorakban. Orvosi Hetilap 142; 723-726, 2001, in Hungarian) NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 166 EP 169 PG 4 ER PT J AU Wrobel, T Mazur, G Surowiak, P Wolowiec, D Jelen, M Kuliczkowsky, K AF Wrobel, Tomasz Mazur, Grzegorz Surowiak, Pawel Wolowiec, Dariusz Jelen, Michal Kuliczkowsky, Kazimierz TI Increased Expression of Vascular Endothelial Growth Factor (VEGF) in Bone Marrow of Patients with Myeloproliferative Disorders (MPD) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE angiogenesis; myeloproliferative disorders; VEGF ID angiogenesis; myeloproliferative disorders; VEGF AB Angiogenesis is a multistep process of the development of capillaries from established blood vessels. Angiogenesis probably plays a significant role in the development and progression of hematopoietic malignancies. Higher microvascular density and increased serum levels of proangiogenic factors such as vascular endothelial growth factor (VEGF) or basic fibroblasts growth factor (bFGF) have been reported in acute and chronic leukemias, myeloproliferative and myelodysplastic disorders, multiple myeloma and lymphomas. The microvessel density of bone marrow stroma in myeloproliferative disorders is increased and VEGF is considered as the most potent endothelial cell activator. The purpose of this study was to examine the expression of VEGF in bone marrow of patients with MPD. 60 paraffinembedded bone marrow core biopsy specimens from newly diagnosed patients with MPD were evaluated. In addition 10 bone marrow core biopsy specimens from adult patients without evidence of malignancy were used as controls. Bone marrow sections were stained immunohistochemically for VEGF (PharMingen, USA). Obtained data show that MPD are associated with an increased expression of VEGF in the bone marrow. This observation support previous studies suggesting that angiogenesis may play a role in the pathophysiology of myeloproliferative disorders. Clinical significance of this phenomenon needs further investigation however thus provides rationale for use of angiogenesis inhibitors in MPD therapy. C1 [Wrobel, Tomasz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, ul. Pasteura 4, 50-367 Wroclaw, Poland. [Mazur, Grzegorz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, ul. Pasteura 4, 50-367 Wroclaw, Poland. [Surowiak, Pawel] Wroclaw Medical University, Department of Human Morphology and EmbryologyWroclaw, Poland. [Wolowiec, Dariusz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, ul. Pasteura 4, 50-367 Wroclaw, Poland. [Jelen, Michal] Wroclaw Medical University, Department of PathologyWroclaw, Poland. [Kuliczkowsky, Kazimierz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, ul. Pasteura 4, 50-367 Wroclaw, Poland. RP Wrobel, T (reprint author), Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, 50-367 Wroclaw, Poland. EM wrobelt@hemat.am.wroc.pl CR Belgore FM, Lip G, Bareford D, et al: Plasma levels of vascular endothelial growth factor, VEGF, in haematological cancers. Br J Haematol 110: 496-497, 2000. Elliot MA, Mesa RA, Li CY, et al: Thalidomide treatment in myelofibrosis with myeloid metaplasia. Br J Haematol 117: 288-296, 2002. Faderl S, Talpaz M, Estrov Z, et al: The biology of chronic myeloid leukemia. N Engl J Med 341: 164-172, 1999. Folkman J: Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med 1: 27-31, 1995. Hsu SM, Raine L, Fanger H: Use of avidin-biotin-peroxidase complex, ABC, in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody, PAP, procedures. J Histochem Cytochem 29: 577-582, 1981. Janowska-Wieczorek A, Majka M, Marquez-Curtis L, et al: Bcr-abl-positive cells secrete angiogenic factors including matrix metalloproteinases and stimulate angiogenesis in vivo in Martigel implants. Leukemia 16: 1160-1166, 2002. Li CY: The role of morphology, cytochemistry and immunochemistry in the diagnosis of chronic myeloproliferative diseases. Int J Haematol 76,, Suppl II): 6-8, 2002. Lundberg LG, Lerner R, Sundelin P, et al: Bone marrow in polycythemia vera, chronic myeloid leukemia and myelofibrosis has an increased vascularity. Am J Pathol 157: 15-19, 2000. McMahon G: VEGF receptor signaling in tumor angiogenesis. Oncologist 5: 3-10, 2000. Molica S, Santoro R, Iuliano F, et al: Serum levels of vascular endothelial growth factor in chronic leukemias. A comparative study with emphasis on myeloproliferative disorders. Hematologica 86: 771, 2001. Murphy S, Peterson P, Iland H, et al: Experience of the Polycytaemia Vera Study Group with essential thrombocytaemia: A final report on diagnostic criteria, survival and leukemic transition by treatment. Semin Hematol 34: 29-35, 1997. Musolino C, Calabro L, Bellomo G, et al: Soluble angiogenic factors: implications for chronic myeloproliferative disorders. Am J Hemat 69: 159-163, 2002. Pearson TC, Messinezy M: The diagnostic criteria of polycythemia rubra vera. Leuk Lymphoma 22,, suppl1): 87-94, 1996. Perez-Atayde AR, Sallan SE, Tedrow U, et al: Spectrum of tumor angiogenesis in bone marrow of children with acute lymphoblastic leukemia. Am J Pathol 150: 815-821, 1997. Di Raimondo F, Palumbo G, Molica S, et al: Angiogenesis in chronic myeloproliferative diseases. Acta Haematol 106: 177- 183, 2001. Remmele W, Stenger HE: Vorschlag zur einheitlichen Definition eines Immunreaktiven Score, IRS, fuer den immunohistochemischen Oestrogenrezeptor – Nachweis, ER – ICA, im Mammakarzinomgewebe. Pathologe 8: 138-140, 1987. Shi SR, Key ME, Kalra KL: Antigen retrieval in formalin-fixed, paraffin-embedded tissues: an enhancement method for immunohistochemical staining based on microvave oven heating of tissue sections. J Histochem Cytochem 39: 741-746, 1991. Tefferi A: The Philadelphia chromosome negative chronic myeloproliferative disorders: A practical overview. Mayo Clin Proc 73: 1177-1184, 1998. Thomas DA, Giles FJ, Cortes J, et al: Antiangiogenic therapy in leukemia. Acta Haematol 106: 190-207, 2001. Wrobel T, Mazur G, Surowiak P, et al: Increased expression of vascular endothelial growth factor, VEGF, in bone marrow of multiple myeloma, MM, patients. Eur J Internal Med 14: 98- 100, 2003. Verstovsek S, Kantarijan H, Manshourti T, et al:Prognostic significance of cellular vascular endothelial growth factor expression in chronic phase chronic myeloid leukemia. Blood 99: 2265-2267, 2002. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 170 EP 173 PG 4 ER PT J AU Kereskai, L Vass, AJ Kneif, M Pajor, L AF Kereskai, Laszlo Vass, A Janos Kneif, Maria Pajor, Laszlo TI Correlation Between BCR-ABL Expression and Tumor Burden is Restricted to the Transition from Minor to Major Cytogenetic Response in Interferon Treated CML Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chronic myeloid leukaemia; molecular monitoring; quantitative polymerase chain reaction; fluorescence in situ hybridisation ID chronic myeloid leukaemia; molecular monitoring; quantitative polymerase chain reaction; fluorescence in situ hybridisation AB The interferon treatment of chronic myeloid leukaemia has been monitored by investigating the tumour burden as revealed by fluorescence in situ hybridization and the expression of BCR-ABL chimera determined by quantitative reverse transcription polymerase chain reaction. These parameters were obtained from the peripheral blood of 51 untreated and 104 follow-up patient samples. Poor corrrelation (r=0.31) was found between BCR-ABL expression and tumor load in all samples as well as in untreated patients, and this correlation was even less in all follow-up cases (r=0.28). Regarding chimera expression five order of magnitude difference existed in the untreated patients and this value dropped to two in those with complete cytogenetic response. Only the major and the complete cytogenetic response groups differed significantly (p<0.001) in the BCR-ABL expression from that of patients at diagnosis. Among the different cytogenetic response groups the only significant difference (p<0.01) in the BCRABL expression was obtained between the major and the minor responders. In the individual patients not only correlated changes of residual tumour mass and chimera expression, but mainly independent changes of these two parameters were observed. This indicates that the BCR-ABL expression and the tumor burden are largely independent variables. C1 [Kereskai, Laszlo] University of Pecs, Department of Pathology, 12 Szigeti Str, H-7624 Pecs, Hungary. [Vass, A Janos] University of Pecs, Department of Pathology, 12 Szigeti Str, H-7624 Pecs, Hungary. [Kneif, Maria] University of Pecs, Department of Pathology, 12 Szigeti Str, H-7624 Pecs, Hungary. [Pajor, Laszlo] University of Pecs, Department of Pathology, 12 Szigeti Str, H-7624 Pecs, Hungary. RP Pajor, L (reprint author), University of Pecs, Department of Pathology, H-7624 Pecs, Hungary. EM pajor@pathology.pote.hu CR Bedi A, Zehnbauer BA, Collector MI, et al: BCR-ABL gene rearrangement and expression of primitive hematopoietic progenitors in chronic myeloid leukemia. Blood 81: 2898-2902, 1993 Bentz M, Cabot G, Moos M,et al: Detection of chimeric BCRABL genes on bone marrow samples and blood smears in chronic myeloid and acute lymphoblastic leukaemia by in situ hybridization. Blood 83: 1922-1928, 1994 Brizard F, Chomel JC, Veinstein A, et al: Does BCR-ABL genomic rearrangement persist in CML patients in complete remission after interferon therapy? Leukemia 12: 1076-1080, 1998 Cuneo A, Bigoni R, Emmanuel B, et al: Fluorescence in situ hybridization for the detection and monitoring of the Ph-positive clone in chronic myelogenous leukemia: comparison with metaphase banding analysis. Leukemia 12: 1718-1723, 1998 Dewald GW, Schad CR, Christensen ER, et al: The application of fluorescent in situ hybridization to detect Mbcr/abl fusion in variant Ph chromosomes in CML and ALL. Cancer Genet Cytogen 71: 7-14, 1993 Faderl S, Talpaz M, Kantarjian HM, et al: Should polymerase chain reaction analysis to detect minimal residual disease in patients with chronic myelogenous leukemia be used in clinical decision making? Blood 93: 2755-2759, 1999 Furukawa Y, Iwase S: Antileukemic effect of interferon-a is mediated through down-modulation of E2F activity. Leukemia 11: 446-448, 1997 Gaiger A, Henn T, Horth E, et al: Increase of BCR-ABL chimeric mRNA expression in tumor cells of patients with chronic myeloid leukemia precedes disease progression. Blood 86: 2371-2378, 1995 Garcia-Isidoro M, Tabernero MD, Garcia JL,et al: Detection of the Mbcr/abl translocation in chronic myeloid leukemia by fluorescence in situ hybridization: Comparison with conventional cytogenetics and implication for minimal residual disease detection. Hum Pathol 28: 154-159, 1997 Hermans A, Gow J, Selleri L, et al: Bcr-abl oncogene activation in Philadelphia chromosome-positiv acute lymphoblastic leukemia. Leukemia 2: 628-633, 1988 Hochhaus A, Lin F, Reiter A, et al: Quantification of residual disease in chronic myelogeneous leukemia patients on Interferon- a therapy by competitive polymerase chain reaction. Blood 87: 1549-1555, 1996 Hochhaus A, Reiter A, Skladny H, et al: Molecular heterogeneity in complete cytogenetic responders after interferon—therapy of chronic myelogenous leukemia: levels of minimal residual disease predict risk of relapse. Blood 88, suppl. 1): 664a., abstr. No 2644), 1996 Hochhaus A, Lin F, Reiter A, et al: Variable numbers of BCRABL transcripts persist in CML patients who achieve complete cytogenetic remission with interferon-. Brit J Haematol 91: 126-131, 1995 Hochhaus A, Weisser A, La Rosee P, et al: Detection and quantification of residual disease in chronic myelogeneous leukemia. Leukemia 14: 998-1005, 2000 Issaad C, Ahmed M, Novault S, et al: Biological effects induced by variable levels of BCR-ABL protein in the pluripotent hematopoietic cell line UT-7. Leukemia 14: 662-670, 2000 Jakab Z, Balogh E, Kiss C, et al: Biclonal chromosomal aberrations in a child with myelodysplastic syndrome. Cancer Genet Cytogen 108: 13-18, 1999 Kantarjian HM, Deisseroth A, Kurzrock R, et al: Chronic myelogenous leukemia: A concise update. Blood 82: 691-703, 1993 Kantarjian HM, OBrien S, Anderlini P, et al: Treatment of chronic myelogenous leukemia: current status and investigational options. Blood 87: 3069-3081, 1996 Kantarjian HM, Smith TL, OBrian S, et al: Prolonged survival in chronic myelogenous leukemia after cytogenetic response to Interferon- therapy. Ann Intern Med 122: 254-261, 1995 Keating A, Xing-Hua Wang XH, Laraya P: Variable transcription of BCR-ABL by Ph+ cells arising from hematopoietic progenitors in chronic myeloid leukemia. Blood 83: 1744-1749 Lion T, Gaiger A, Henn T, et al: Use of quantitative polymerase chain reaction to monitor residual disease in chronic myelogenous leukemia during treatment with interferon. Leukemia 9: 1353-1360, 1995 Moore S, Haylock DN, Levesque JP, et al: Stem cell factor as a single agent induces selective proliferation of the Philadelphia chromosome positive fraction of chronic myeloid leukemia CD34(+, cells. Blood 92: 2461-2470, 1998 Muhlmann J, Thaler J, Hilbe W, et al: Fluorescence in situ hybridization, FISH, on peripheral blood smears for monitoring Philadelphia chromosome-positive chronic myeloid leukemia, CML, during Interferon treatment : A new strategy for remission assessment. Gene Chromosome Canc 21: 90- 100, 1998 Pajor L, Szuhai K, Mehes G, et al: Combined metaphase, interphase cytogenetic, and flow cytometric analysis of DNA content of pediatric acute lymphoblastic leukaemia. Cytometry 34: 87-94, 1998 Pajor L, Vass JA, Kereskai L, et al: Silent Philadelphia chromosome: A distinct developmental stage in a Philadelphia chromosome-positive chronic myeloproliferation? Cancer Genet Cytogen 118: 14-19, 2000 Reiter A, Marley SB, Hochhaus A, et al: BCR-ABL-positive progenitors in chronic myeloid leukaemia patients in complete cytogenetic remission after treatment with interferon-alpha. Brit J Haematol 102: 1271-1278, 1998 Shtivelman E, Gale RP, Dreazen O, et al: bcr-abl RNA in patients with chronic myelogenous leukaemia. Blood 69: 971- 973, 1987 Talpaz M, Kantarjian HM, Kurznock R, et al: Interferon-alfa produces sustained cytogenetic responses in chronic myelogenous leukaemia Ph-chromosome positive patients. Ann Intern Med 114: 532-538, 1991 Tchirkov A, Giollant M, Tavernier F, et al: Interphase cytogenetics and competitive RT-PCR for residual disease monitoring in patients with chronic myeloid leukaemia during interferontherapy. Brit J Haematol 101: 552-557, 1998 The Italian Cooperative Study Group on Chronic Myeloid Leukemia: Interferon Alfa-2a as compared with conventional chemotherapy for the treatment of chronic myeloid leukemia. New Engl J Med 330: 820-825, 1994 Verfaillie CM, Bhatia R, Miller W, et al: BCR/ABL-negative primitive progenitors suitable for transplantation can be selected from the marrow of most early-chronic phase but not accelerated- phase chronic myelogenous leukemia patients. Blood 87: 4770-4779, 1996 Yanagi M, Shinjo K, Takeshita A, et al:Simple and reliably sensitive diagnosis and monitoring of Philadelphia chromosomepositive cells in chronic myeloid leukemia by interphase fluorescence in situ hybridization of peripheral blood cells. Leukemia 13: 542-552, 1999 Zhang JG, Lin F, Chase A, et al: Comparison of genomic DNA and cDNA for detection of residual disease after treatment of chronic myeloid leukemia with allogeneic bone marrow transplantation. Blood 87: 2588-2593, 1996 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 174 EP 179 PG 6 ER PT J AU Gorog, D Nagy, P Peter, A Perner, F AF Gorog, Denes Nagy, Peter Peter, Antal Perner, Ferenc TI Influence of Obesity on Lymph Node Recovery from Rectal Resection Specimens SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE rectal resection; lymph node; obesity ID rectal resection; lymph node; obesity AB Careful lymph node dissection from colorectal resection specimens is important procedure for cancer staging. Present study intended to assess the impact of surgical technique and patient’s obesity on this process. Number of lymph nodes harvested by manual dissection from resection specimens of 141 patients with rectal cancer and the rate of nodal metastases were analyzed and compared in different groups of patients selected by length of resection specimen and body mass index. The median and mean number of lymph nodes found per patient were 6 and 6.7. The shorter resection specimens (<16 cm after formalin fixation) yielded significantly lower number of nodes than those with length > 16 cm (5.7 versus 7.9). Most significant reduction in mean number of lymph nodes was observed in obese patients with short specimens (4.8). This subset of patients presented the lowest rate of nodal metastases (38%). The surgical technique seems to be an important factor for lymph node recovery from rectal resections specimens. The patient’s obesity had an unfavourable impact on this procedure. Standardized surgery and histopathological examination are needed even in non-specialized centers to harvest adequate number of lymph nodes. C1 [Gorog, Denes] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Nagy, Peter] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Peter, Antal] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Perner, Ferenc] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. RP Gorog, D (reprint author), Semmelweis University, Department of Transplantation and Surgery, H-1082 Budapest, Hungary. EM gorogd@freemail.hu CR Blenkinsopp WK, Stewart-Brown S, Blesovsky L, et al: Histopathology reporting in large bowel cancer. J Clin Pathol 34: 509-513, 1981 Calle EE, Thun MJ, Petrelli JM, et al: Body mass index and mortality in a prospective cohort of US adults. N Eng J Med 341: 1097-1105, 1999 Caplin S, Cerottini J-P, Bosman FT, et al: For patients with Dukes’B, TNM stage II, colorectal carcinoma, examination of six or fewer lymph nodes is related to poor prognosis. Cancer 83: 666-672, 1998 Casillas S, Pelley RJ, Milsom JW: Adjuvant therapy for colorectal cancer with lymph node metastases. Cancer 73: 2076- 2082, 1994 Cawthorn SJ, Gibbs NM, Marks CG: Clearance technique for the detection of lymph nodes in colorectal cancer. Br J Surg 73: 58-60, 1986 Cianchi F, Palomba A, Boddi V, et al: Lymph node recovery from colorectal tumor specimens: recommendation for a minimum number of lymph nodes to be examined. World J Surg 26: 384-389, 2002 Goldstein NS, Sanford W, Coffey M, et al: Lymph node recovery from colorectal resection specimens removed for adenocarcinoma: Trends over time and a recommendation for a minimum number of lymph nodes to be recovered. Am J Clin Pathol 106: 209-216, 1996 Hermanek P: pTNM and residual tumor classifications: Problems of assessement and prognostic significance. World J Surg 19: 184-190, 1995 Hermanek P, Henson DE, Hutter RVP, et al: UICC TNM Supplement 1993: A commentary on uniform use. Springer-Verlag, New York, 1993. Hernanz F, Revuelta S, Redondo C, et al: Colorectal adenocarcinoma: Quality of the assessment of lymph node metastases. Dis Colon Rectum 37: 373-377, 1994 Herrera L, Villareal JR, Cert RT: Incidence of metastases from rectal adenocarcinoma in small lymph nodes detected by clearing technique. Dis Colon Rectum 35: 783-788, 1992 Hodgson DC, Zhang W, Zaslavsky AM, et al: Relation of hospital volume to colostomy rates and survival for patients with rectal cancer. J Natl Cancer Inst 95: 708-716, 2003 Jass JR: Prognostic factors in rectal cancer. Eur J Cancer 31A: 862-863, 1995 Maurel J, Launoy G, Grosclaude P, et al: Lymph node harvest reporting in patients with carcinoma of the large bowel. Cancer 82: 1482-1486, 1998 Newland RC, Dent OF, Lyttle MNB, et al: Pathologic determinants of survival associated with colorectal cancer with lymph node metastases. Cancer 73:2076-2082, 1994 Scott KWM, Grace RH: Detection of lymph node metastases in colorectal carcinoma before and after fat clearance. Br J Surg 76: 1165-1167, 1989 Tepper JE, O’Connel MJ, Niedzwiecki D, et al:Impact of number of nodes retrieved on outcome in patients with rectal cancer. J Clin Oncol 19: 157-163, 2001 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 180 EP 183 PG 4 ER PT J AU Liszkay, G Peley, G Sinkovics, I Peter, I Orosz, Zs Fejos, Zs Horvath, B Koves, I Gilde, K Kasler, M AF Liszkay, Gabriella Peley, Gabor Sinkovics, Istvan Peter, Ilona Orosz, Zsolt Fejos, Zsuzsa Horvath, Bela Koves, Istvan Gilde, Katalin Kasler, Miklos TI Clinical Significance of Sentinel Lymph Node Involvement in Malignant Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE sentinel lymph node; melanoma; prognosis; tumor thickness ID sentinel lymph node; melanoma; prognosis; tumor thickness AB In the period 1997-2002, sentinel lymph node (SLN) surgery was performed on 179 primary skin melanoma patients, one to two months after the removal of the primary. Staining with patent blue was combined with an isotope technique. Histological evaluation of the sentinel lymph nodes was performed in serial sections. Immunohistochemical detection of S100, HMB-45, or Melan-A was used in the case of suspected micrometastases. Demonstration of positive sentinel lymph node was followed, preferably within 2-3 weeks, by regional block dissection. In these cases interferon-a2 in low doses or BCG immune therapy were applied as adjuvant therapy. Bimonthly follow-up of the patients included physical examination and the use of imaging techniques as specified in the melanoma protocol. Sentinel lymph node surgery was successful in 177/179 cases (98%). Positive sentinel lymph node was identified in 26/177 patients (14.7%). In node positive patients the thickness of the primary tumour was significantly greater than that of node negative ones (p<0.00001). Patients with micrometastases had significantly poorer symptom-free and overall survival by the Mantel-Cox test than those of the other group (p=0.0001 and p=0.0007 respectively). Comparison of the tumor thickness and positive SLN by discriminance analysis, yielded 81.7% and 79.9%, respectively for correct classification rates. Based on our study and data from the literature, we suggest SLN-positivity as equally strong poor prognosis factor for skin melanoma as the tumor thickness. C1 [Liszkay, Gabriella] National Institute of Oncology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Peley, Gabor] National Institute of Oncology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Sinkovics, Istvan] National Institute of Oncology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Peter, Ilona] National Institute of Oncology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Orosz, Zsolt] National Institute of Oncology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Fejos, Zsuzsa] National Institute of Oncology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Horvath, Bela] National Institute of Oncology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Koves, Istvan] National Institute of Oncology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Gilde, Katalin] National Institute of Oncology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Kasler, Miklos] National Institute of Oncology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. RP Liszkay, G (reprint author), National Institute of Oncology, H-1122 Budapest, Hungary. EM liszkay@oncol.hu CR Alazraki MT, Eshima LA: Lymphoscintigraphy, the sentinel node concept, and the intraoperative gamma probe in melanoma, breast cancer, and other potential cancers. Semin Nucl Med 27: 55-67, 1997 Alex JC, Krag DN: Gamma-probe-guided localization of lymph nodes. Surg Oncol 2: 137-147, 1993 Association od Directors of Anatomic and Surgical Pathology. ADASP recommendations for processing and reporting lymph node specimens submitted for evaluation of metastatic disease. Am J Surg Pathol 25: 961-963, 2001 Buzaid AC, Balch CM: Proposed change of TNM classification. Mel Res 11 Suppl: S4-S5, 2001 Cabanas RM: An approach for the treatment of penile cancer. Cancer 39: 456-466, 1977 Coldiron MB:Sentinel node biopsy: who needs it? Int J of Dermatol 39: 807-811, 2000 Kirkwood JM, Hunt Strawdwrman M, Ernstoff MS, et al. Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: The Eastern Cooperative Oncology Group Trial EST 1684. J Clin Oncol 14:7-17, 1996 Krag DN, Meijer SJ, Weaver DL, et al: Minimal access surgery for staging of malignant melanoma. Arch Surg 130: 654-658, 1995 Lens MB, Dawes M, Newton-Bishop JA: Tumour thickness as a predictor of occult lymph node metastases in patients with stage I and II melanoma undergoing sentinel lymph node biopsy. Br J Surg 89:1223-1227, 2002 Liszkay G, Peley G, Farkas E, et al. Sentinel node biopsy for melanoma in 103 patients. Early follow-up study. EJSO 28: 357, 2002 Moore MP, Kinne DW. Axillary lymphadenectomy: A diagnostic and therapeutic procedure. J Surg Oncol 66: 2-6, 1997 Morton DL, Wen DR, Wong JH, et al. Technical details of intraoperative lymphatic mapping for early stage melanoma. Arch Surg 127: 392-399, 1992 Reintgen D, Balch CM, Kirkwood J, et al. Recent advances in the care of the patient with malignant melanoma. Ann Surg 225: 1-14, 1997 Ronan GS, Eng MA, Briele AH, et al. Thin malignant melanomas with regression and metastases. Arch Dermatol 123: 1326- 1330, 1987 Ross MI. Surgical management of stage I and II melanoma patients: approach to the regional lymph node basin. Semin Surg Oncol 12: 394-401, 1996 Ross MI, Reintgen D, Balch Ch: Selective lymphadenectomy: Emerging role of lymphatic mapping and sentinel node biopsy in the management of early stage melanoma. Semin Surg Oncol 9: 219-223, 1993 Starz H, Gerstel C, Bachter D, et al: Two simple morphometric parameters permit a clinically relevant staging of the sentinel node metastases in malignant melanoma and merkel cell carcinoma. Eur J Nucl Med 26, Suppl): S68, 1988 Taylor A, Murray D, Herda S, et al. Dynamic lymphoscintigraphy to identify the sentinel and satellite nodes. Clin Nucl Med 21: 755-758, 1996 Timar J, Csuka O, Orosz Z et al:Molecular pathology of tumor metastasis. II. Molecular staging and differential diagnosis. Pathol Oncol Res 8: 204-219, 2002 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 184 EP 187 PG 4 ER PT J AU Mannweiler, S Dinges, PH Beham-Schmid, Ch Hauser, H Starlinger, M Regauer, S AF Mannweiler, Sebastian Dinges, Peter Hans Beham-Schmid, Christine Hauser, Hubert Starlinger, Michael Regauer, Sigrid TI Colliding / Concomitant Tumors of the Intestine: Report of 3 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE adenocarcinoma; non-Hodgkin lymphoma; colon; ileum ID adenocarcinoma; non-Hodgkin lymphoma; colon; ileum AB Collision/concomitant tumors of the intestine involving lymphomas are very rare. For these cases molecular genetic analyses are valuable diagnostic adjuncts. We report one collision tumor of the rectum (adenocarcinoma and peripheral T-cell lymphoma, unspecified), and two cases of concomitant tumors (carcinoma in the cecum and lymphoma in the ileum; carcinoma in the sigmoid and lymphoma in the ileum). The collision tumor (poorly differentiated adenocarcinoma and a peripheral T-cell lymphoma, unspezified) showed a variable proportion of the anaplastic tumor cells expressing lymphatic markers as well as cytokeratin. Only polymerase chain reaction (PCR) analysis revealing T-cell monoclonality of the anaplastic part of the colliding tumor allowed the correct diagnosis. In the second case, a moderately differentiated adenocarcinoma in the cecum with a concomitant B-cell Non-Hodgkin lymphoma in the ileum, PCR analysis was non contributary. In the third case (adenocarcinoma in the sigmoid colon and a follicular center lymphoma in the ileum) PCR analysis revealed gene rearrangement of the immunoglobulin heavy chain gene. We would like to emphasize that collision and concomitant tumors of the gut are rare and that molecular genetic analysis may be mandatory for correct diagnosis. It is our impression, that these tumors may be diagnosed more often in the intestinal tract if molecular genetic analysis and immunohistochemistry are used routinely, at least for all anaplastic tumors. C1 [Mannweiler, Sebastian] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. [Dinges, Peter Hans] Hospital Klagenfurt, Department of PathologyKlagenfurt, Austria. [Beham-Schmid, Christine] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. [Hauser, Hubert] University of Graz, Surgical DepartmentGraz, Austria. [Starlinger, Michael] Landeskrankenhaus Klagenfurt, Surgical DepartmentKlagenfurt, Austria. [Regauer, Sigrid] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. RP Mannweiler, S (reprint author), Medical University of Graz, Department of Pathology, A-8036 Graz, Austria. EM sebastian.mannweiler@uni-graz.at CR Aoki T, Sasano M, Ikeuchi D, et al: Coexistence of primary colonic malignant lymphoma and rectal adenocarcinoma. Jpn J Gastroenterol Surg 34: 54-58, 2001. Beham-Schmid C, Beham A, Jakse R, et al: Extranodal follicular dendritic cell tumour of the nasopharynx. Virchows Arch 432: 293-298, 1998. Camunas Mohinelo F, Requena P M, Zaragoza J M, et al: A. Tumor de colision gastrico con metaplasia osea. Rev Esp Enf Digest 89: 317-319, 1997. Carr N, Remotti H, Sobin L: Dual carcinoid/epithelial neoplasia of the appendix. Histopathology 27: 557-562, 1995. Chazouilleres O, Andreani T, Boucher J, et al: Adenocarcinome rectal associe a un lymphome, “collision tumor”). Gastroenterol Clin Biol 14: 185-186, 1990. Corsi A, Bosman C: Adenocarcinoma and atypical carcinoid: morphologic study of a gastric collision-type tumor in the carcinoma- carcinoid spectrum. Ital J Gastroenterol 27: 303-308, 1995. Frierson H, Bellafiore F, Gaffey M, et al: Cytokeratin in anaplastic large cell lymphoma. Mod Pathol 7: 317-321, 1994. Furuya M, Saito Y, Funakosi K, et al: A caseof lymphomatous polyposis of the gastrointestinal tract. Endosc Forum Dig Dis 13: 200-204, 1997. Ghose A, Dasgupta J, Konar A, et al: Collision tumor of gastrointestinal tract. Indian J Gastroenterol 13: 29-30, 1994. Goteri G, Ranaldi R, Rezai B, et al: Synchronous mucosa-associated lymphoid tissue lymphoma and adenocarcinoma of the stomach. Am J Surg Pathol 21: 505-509, 1997. Lasota J, Hyjek E, Koo C, et al: Cytokeratin-positive large-cell lymphomas of B-cell lineage. A study of five phenotypically unusual cases verified by polymerase chain reaction. Am J Surg Pathol 20: 346-354, 1996. Mir-Madjlessi S, Vafai M, Khademi J, et al: Coexisting primary malignant lymphoma and adenocarcinoma of the large intestine in an IgA-deficient boy. Dis Colon Rectum 27: 822-824, 1984. Morishita Y, Tanaka T, Kato K, et al: Gastric collision tumor, carcinoid and adenocarcinoma, with gastritis cystica profunda. Arch Pathol Lab Med 115: 1006-1010, 1991. Nakamura S, Aoyagi K, Iwanaga S, et al: Synchronous and metachronous primary gastric lymphoma and adenocarcinoma: A clinicopathologic study of 12 patients. Cancer 79: 1077- 1085, 1997. Nishino N, Konno H, Baba S, et al: Synchronous lymphoma and adenocarcinoma occuring as a collision tumor in the stomach: Report of a case. Surg Today 26: 508-512, 1996. Popper H, Winter E, Hofler G: DNA of mycobacterium tuberculosis in formalin-fixed, paraffin-embedded tissue in tuberculosis and sarcoidosis detected by polymerase chain reaction. Am J Clin Pathol 101: 738- 741, 1994. Schmid C, Sargent C, Isaacson PL: H cells of nodular lymphocyte predominant Hodgkin´s disease show immunoglobulin light-chain restriction. Am J Pathol 139: 1281-1289, 1991. Wagle S, Mohandas K, Vazifdar K, et al: Synchronous adenocarcinoma and lymphoma of the colon. Indian J Gastroenterol 16: 28-29, 1997. Williams I, Williams N, Stock D, et al: Collision tumour of the ampulla of Vater: Carcinoid and adenocarcinoma. HPB Surg 10: 241-244, 1997. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 188 EP 192 PG 5 ER PT J AU Vargas-Gonzalez, R Sotelo-Avila, C Coria, SA AF Vargas-Gonzalez, Roberto Sotelo-Avila, Cirilo Coria, Solis Araceli TI Renal Medullary Carcinoma in a Six-Year-Old Boy with Sickle Cell Trait SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE sickle cell disease; renal medullary carcinoma; collecting duct carcinoma ID sickle cell disease; renal medullary carcinoma; collecting duct carcinoma AB Renal medullary carcinoma (RMC), an aggressive malignant epithelial neoplasm, first emerged as a distinct clinicopathologic entity in 1995. It affects individuals 40 years of age or younger and is strongly associated with sickle cell disease or trait. The majority of patients with RMC have widely disseminated disease at the time of diagnosis and most fail to respond to both chemotherapy and radiotherapy. Mortality approaches 100%, and death usually occurs within a few months to a year of diagnosis. We report a 6-year-old African-American boy with a history of gross hematuria who died four weeks after diagnosis of disseminated metastatic disease. Autopsy showed a 4.4-cm renal mass with metastases to the contra lateral kidney, liver, lungs and multiregional lymph nodes. RMC should be included in the differential diagnosis of any patient 40 years old or younger with a history of hemoglobinopathy and gross hematuria and/or abdominal or flank pain. A brief discussion of the differential diagnosis, histogenesis and treatment is presented in this study. C1 [Vargas-Gonzalez, Roberto] Hospital Para el Nino Poblano, Department of PathologyPuebla, Mexico. [Sotelo-Avila, Cirilo] Cardinal Glennon Children’s Medical Center, Department of Pathology, 63104-1095 St. Louis, Missouri, USA. [Coria, Solis Araceli] Hospital Infantil de Mexico Federico Gomez, Department of PediatricsMexico City, Mexico. RP Sotelo-Avila, C (reprint author), Cardinal Glennon Children’s Medical Center, Department of Pathology, 63104-1095 St. Louis, USA. EM Cirilo_Sotelo_MD@ssmhc.com CR Berman LB: Sickle cell nephropathy. JAMA 228:1279, 1974. Davis CJJ, Mostofi FK, Sesterhenn IA: Renal medullary carcinoma: the seventh sickle cell nephropathy. Am J Surg Pathol 19: 1-11, 1995. Avery RA, Harris JE, Davis CJJ, et al: Renal medullary carcinoma: clinical and therapeutic aspects of a newly described tumor. Cancer 78: 128-132, 1996. Dimashkieh H, Choe J, Mutema G: Renal medullary carcinoma a report of two cases and review of the literature. Arch Pathol Lab Med 127:e 135-138, 2003. Swartz MA, Karth J, Schneider DT et al.Renal medullary carcinoma: Clinical, pathologic, immunohistochemical and genetic analysis with pathogenic implications. Urology 60: 1083-89, 2002. Davidson AJ, Choyke PL, Hartman DS, Davis CJ Jr. Renal medullary carcinoma associated with sickle cell trait: radiologic findings. Radiology 195: 83-85, 1995. Srigley JR, Eble JN: Collecting duct carcinoma of kidney. Semin Diagn Pathol 15: 54-67, 1998. Mostofi FK, Vorder Bruegge CF, Diggs LW: Lesions in kidneys removed for unilateral hematuria in sickle cell disease. Arch Pathol 63: 336-51, 1957. Pham PT, Pham PC, Wilkinson AH, et al: Renal abnormalities in patients in sickle cell disease. Kidney Int 57: 1-8, 2000. Ataga KI, Orringer EP: Renal abnormalities in sickle cell disease. Am J Hematol 63: 205-211, 2000. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 193 EP 195 PG 3 ER PT J AU Bal, N Erdogan, A Kayaselcuk, F Tarim, E Tuncer, I AF Bal, Nebil Erdogan, Aslan Kayaselcuk, Fazilet Tarim, Ebru Tuncer, Ilhan TI Vulvar Syringoma Aggravated by Pregnancy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE pregnancy; syringoma; vulva ID pregnancy; syringoma; vulva AB Syringoma is a benign tumors of eccrine sweat gland. They appear as multiple, tiny, firm, skin-colored papules. Vulvar involvement of syringoma is rare. Only 24 cases with vulvar syringoma have been previously reported in the literature. The majority of patients with vulvar syringomas are asymptomatic. A case of syringoma of the vulva exacerbated during pregnancy is presented. The case appears remarkable for the experienced aggravated pruritic symptoms of the patient during her pregnancy. C1 [Bal, Nebil] Baskent University, Department of Pathology, Adana Hastanesi, Yuredir, 01250 Adana, Turkey. [Erdogan, Aslan] Baskent University Faculty of Medicine, Adana Teaching and Medical Research Center, Department of Gynecology and ObstetricAdana, Turkey. [Kayaselcuk, Fazilet] Baskent University, Department of Pathology, Adana Hastanesi, Yuredir, 01250 Adana, Turkey. [Tarim, Ebru] Baskent University Faculty of Medicine, Adana Teaching and Medical Research Center, Department of Gynecology and ObstetricAdana, Turkey. [Tuncer, Ilhan] Baskent University, Department of Pathology, Adana Hastanesi, Yuredir, 01250 Adana, Turkey. RP Bal, N (reprint author), Baskent University, Department of Pathology, 01250 Adana, Turkey. EM nebilbal@yahoo.com CR Gerdsen R, Wenzel J, Uerlich M et al. Periodic genital pruritus caused by syringoma of the vulva. Acta Obstet Gynecol Scand 81:369-370, 2002 Tay YK, Tham SN, Teo R. Localized vulvar syringoma – an unusual cause of pruritus vulvae. Dermatology 192:62-63, 1996 Turan C, Ugur M, Kutluay L et al. Vulvar syringoma exacerbated during pregnancy. Eur J Obstet Gynecol Reprod Biol 64:141- 142, 1996 Belardi MG, Maglione MA, Vighi S, et al. Syringoma of the vulva. A case report. J Reprod Med 39:957-959, 1994 Nogita T, Kawashima M. Subclinical syringoma coexisting with nevocellular nevus on the vulva. J Dermatol 20:188-189, 1993 Trager JD, Silvers J, Reed J et al. Neck and vulvar papules in an 8-year-old girl. Arch Dermatol 135:203-206, 1999 Kurman RJ. Blaustein’s Pathology of the Female Genital Tract, Fifth ed. New York: Springer-Verlag Press, 2002. Elder D. Lever’s Histopathology of the Skin, Eight ed. Philadelphia: Lippincott-Raven Publishers, 1997. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 196 EP 197 PG 2 ER PT J AU Megyesi, M Berta, M Khoor, A AF Megyesi, Maria Berta, Maria Khoor, Andras TI Endobronchial Large Cell Neuroendocrine Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE neuroendocrine carcinoma; lung ID neuroendocrine carcinoma; lung AB Lung tumors with neuroendocrine morphology include typical carcinoid, atypical carcinoid, large cell neuroendocrine carcinoma, and small cell carcinoma. The World Health Organization emphasizes the importance of mitotic count in differentiating these tumors. We studied the case of a 58-year-old male nonsmoker with recurrent pneumonia and an endobronchial mass, which was removed by right middle lobectomy. The patient was alive with no recurrent disease at 36-month follow-up. Histologically, the tumor showed welldeveloped neuroendocrine morphology but contained up to 20 mitoses per 10 high-power fields and was therefore diagnosed as a large cell neuroendocrine carcinoma. However, several features, including the carcinoid-like morphology and endobronchial location of the tumor, absence of smoking history, and promising clinical course, were more characteristic of an atypical carcinoid than of a large cell neuroendocrine carcinoma. It may be necessary to redefine histologic criteria to allow a higher mitotic rate for classification as an atypical carcinoid. C1 [Megyesi, Maria] Central Railway Hospital and Polyclinic, Department of PathologyBudapest, Hungary. [Berta, Maria] Central Railway Hospital and Polyclinic, Department of PathologyBudapest, Hungary. [Khoor, Andras] Mayo Clinic, Department of Laboratory Medicine and Pathology, 4500 San Pablo Road, 32224 Jacksonville, Florida, USA. CR Travis WD, Sobin LH: Histological Typing of Lung and Pleural Tumours. 3rd ed. Berlin, Germany: Springer-Verlag; 1999 Colby TV, Koss MN, Travis WD: Tumors of the Lower Respiratory Tract. Washington, DC: Armed Forces Institute of Pathology; 1995 Travis WD, Linnoila RI, Tsokos MG, et al: Neuroendocrine tumors of the lung with proposed criteria for large-cell neuroendocrine carcinoma: an ultrastructural, immunohistochemical, and flow cytometric study of 35 cases. Am J Surg Pathol 15:529- 553, 1991 Jung KJ, Lee KS, Han J, et al: Large cell neuroendocrine carcinoma of the lung: clinical, CT, and pathologic findings in 11 patients. J Thorac Imaging 16:156-162, 2001 Travis WD, Rush W, Flieder DB, et al: Survival analysis of 200 pulmonary neuroendocrine tumors with clarification of criteria for atypical carcinoid and its separation from typical carcinoid. Am J Surg Pathol 22:934-944, 1998 Dresler CM, Ritter JH, Patterson GA, Ross E, Bailey MS, Wick MR: Clincal-pathologic analysis of 40 patients with large cell neuroendocrine carcinoma of the lung. Ann Thorac Surg 63:180- 185, 1997 Mazieres J, Daste G, Molinier L, et al. Large cell neuroendocrine carcinoma of the lung: pathological study and clinical outcome of 18 resected cases. Lung Cancer 37:287-292, 2002 Takei H, Asamura H, Maeshima A, et al. Large cell neuroendocrine carcinoma of the lung: a clinicopathologic study of eightyseven cases. J Thorac Cardiovasc Surg 124:285-292, 2002 Arrigoni MG, Woolner LB, Bernatz PE. Atypical carcinoid tumors of the lung. J Thorac Cardiovasc Surg 64:413-421, 1972 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 198 EP 200 PG 3 ER PT J AU Bilgic, B Ates, EL Demiryont, M Ozger, H Dizdar, Y AF Bilgic, Bilge Ates, Esberk Lora Demiryont, Misten Ozger, Harzem Dizdar, Yavuz TI Malignant Peripheral Nerve Sheath Tumors Associated With Neurofibromatosis Type 1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Neurofibromatosis; malignant peripheral nerve sheath tumours; sarcoma; nonossifying fibroma ID Neurofibromatosis; malignant peripheral nerve sheath tumours; sarcoma; nonossifying fibroma AB We report 4 cases of malignant peripheral nerve sheath tumors (MPNST) with neurofibromatosis type 1 (NF1). Mean age was 29.5. Two of them had a family history. Three of them were male. All of them had enlarging mass and pain in the background of neurofibromas. Locations were popliteal, thigh and forearm. The masses were greater than 5 cm in diameter in each case. In two cases the mass was showing continuity with a nerve. One patient had a nonossifying fibroma as well as a MPNST. Wide excision and radiotherapy were applied to three of the patients. One of them did not take any therapy after surgical resection. Two of the patients died of lung metastases after a mean period of 12.5 months. In a majority of NF1 patients MPNST emerges from a preexisting neurofibroma. The patients with NF1 are at greatest risk for developing sarcomas, so they should be followed closely. C1 [Bilgic, Bilge] Istanbul Medical Faculty, Department of Pathology, Capa, 34390 Istanbul, Turkey. [Ates, Esberk Lora] Istanbul Medical Faculty, Department of Pathology, Capa, 34390 Istanbul, Turkey. [Demiryont, Misten] Istanbul Medical Faculty, Department of Pathology, Capa, 34390 Istanbul, Turkey. [Ozger, Harzem] Istanbul University, Istanbul Medicine Faculty, Department of Orthopaedic SurgeryIstanbul, Turkey. [Dizdar, Yavuz] Istanbul University, Istanbul Medicine Faculty, Institute of Oncology, Department of Radiation OncologyIstanbul, Turkey. RP Ates, EL (reprint author), Istanbul Medical Faculty, Department of Pathology, 34390 Istanbul, Turkey. EM loraates@hotmail.com CR Blatt J, Jaffe R, Deutsch M, et al: Neurofibromatosis and childhood tumors. Cancer 57: 1225–1229, 1986 D’Agostina AN, Soule EH, Miller RH: Sarcomas of the peripheral nerves and soft tissues associated with multiple neurofibromatosis, Von Recklinghausen’s Disease). Cancer 16: 1015–1027, 1963 Ducatman BS, Scheithauer BW: Malignant peripheral nerve sheath tumors with divergent differentiation. Cancer 54: 1049–1057, 1984 Ducatman BS, Scheithauer BW, Piepgras DG, et al: Malignant peripheral nerve sheath tumors a clinicopathologic study of 120 cases. Cancer 57: 2006–2011, 1986 Ferner RE, Lucas JD, O’Doherty MJ, et al: Evaluation of 18 Fluorodeoxyglucose positron emission tomography, 18 FDG PET, in the detection of malignant peripheral nerve sheath tumours arising from within plexiform neurofibromas in neurofibromatosis 1. J Neurol Neurosurg Psychiatry 68: 353-357, 2000 Halliday AL, Sobel RA, Martuza RL: Benign spinal nerve sheath tumors: their occurrence sporadically and in neurofibromatosis types 1 and 2. J Neurosurg 74: 248–253, 1991 Hajdu SI: Peripheral nerve sheath tumors, histogenesis, classification and prognosis. Cancer 72: 3549–3552, 1993 Hruban RH, Shiu MH, Senie RT, et al: Malignant peripheral nerve sheath tumors of the buttock and lower extremity, A study of 43 Cases. Cancer 66: 1253–1265, 1990 King AA, DeBaun MR, Riccardi VM, et al: Malignant peripheral nerve sheath tumors in neurofibromatosis 1. Am J Med Genet 93: 388-392, 2000 Nussbaum RL, McInnes RR, Willard HF: Genetics In Medicine. Genetics and Cancer, W. B. Saunders Company: Philadelphia, 2001. Rasmussen SA, Overman J, Thomson SAM, et al: Chromosome 17 loss-of-heterozygosity studies in benign and malignant tumors in neurofibromatosis type 1. Genes, Chromosomes, Cancer 28: 425–431, 2000 Schmidt H, Wurl P, Taubert H, et al: Genomic imbalances of 7p and 17q in malignant peripheral nerve sheath tumors are clinically relevant. Genes, Chromosomes, Cancer 25: 205–211, 1999 Topsakal CD, Akdemir I, Tiftikci M, et al: Malignant schwannoma of the sciatic nerve originating in a spinal plexiform neurofibroma associated with neurofibromatosis type 1. Neurol Med Chir, Tokyo, 41: 551–555, 2001 Wanebo JE, Malik JM, VandenBerg SR, et al: Malignant peripheral nerve sheath tumors, A clinicopathologic study of 28 cases. Cancer 71: 1247–1253, 1993 Weiss SW, Goldblum JR: Benign tumors of the peripheral nerves. Malignant tumors of the peripheral nerves. Enzinger and Weiss’s Soft Tissue Tumors. Mosby, St Louis, 2001. Wick MR, Swanson PE, Scheithauer BW, et al: Malignant peripheral nerve sheath tumor. An immunohistochemical study of 62 cases. Am J Clin Pathol 87: 425–433, 1987 Woodruff JM, Christensen WN: Glandular peripheral nerve sheath tumors. Cancer 72: 3618–3628, 1993 Woodruff JM: Pathology of tumors of the peripheral nerve sheath in type 1 neurofibromatosis. Am J Med Genet, Semin Med Genet, 89: 23–30, 1999 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2003 VL 9 IS 3 BP 201 EP 205 PG 5 ER PT J AU Szabo, E Lotz, G Paska, Cs Kiss, A Schaff, Zs AF Szabo, Erzsebet Lotz, Gabor Paska, Csilla Kiss, Andras Schaff, Zsuzsa TI Viral Hepatitis: New Data on Hepatitis C Infection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE viral hepatitis; hepatitis C; chronic hepatitis ID viral hepatitis; hepatitis C; chronic hepatitis AB Viral hepatitis (VH) is almost as old as human beings, at least as old as known human history. However, the natural history and the epidemiology of the disease has undergone changes during the centuries and even recently we have been facing several new aspects. The estimated global prevalence is around 3-5%, which means that approximately 400 million patients are infected with hepatitis B virus and that there are 170 million infections with hepatitis C virus. The mortality figures are projected to show a 2- to 3-fold increase over the next two decades as hepatitis C virus-infected patients develop cirrhosis, which makes this the leading indication for liver transplantation. These data point to the importance of VH being a significant public health problem worldwide. The list of hepatotropic viruses is well known, including hepatitis A (HAV), B (HBV), C (HCV), D (HDV), E (HEV), G (HGV) and F (HFV). HGV and HFV are excluded from the present review, mainly because they are questionable in relation to the causation of liver disease. Our knowledge of HAV, HBV, HDV and HEV has accumulated over the last decade, so the present discussion is focused on HCV, which is currently generating considerable concern and controversy, and is the leading cause of chronic liver disease worldwide. The main questions to be discussed, are: the characterization of the agents’ viral genotypes/subtypes, the viral-cell interaction, the pathogenesis of VH, the extrahepatic manifestations of viral infection and hepatocarcinogenesis. C1 [Szabo, Erzsebet] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93., H-1091 Budapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93., H-1091 Budapest, Hungary. [Paska, Csilla] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93., H-1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93., H-1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93., H-1091 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM schaff@korb2.sote.hu CR Agnello V, Abel G, Elfahal M, et al: Hepatitis C virus and other flaviviridae viruses enter cells via low density lipoprotein receptor. Proc Natl Acad Sci USA 96: 12766-12771, 1999 Adinolfi LE, Utili R, Andreana A, et al: Relationship between genotypes of hepatitis C virus and histopathological manifestations in chronic hepatitis C patients. Eur J Gastroenterol Hepatol 12: 299-304, 2000 Alison MR, Poulsom R, Jeffery R, et al: Hepatocytes from nonhepatic adult stem cells. Nature 406: 257, 2000 Alter MJ, Kruszon-Moran D, Nainon OV, et al: The prevalence of hepatitis C virus infection in United States, 1988 through 1994. N Engl J Med 321: 556-562, 1999 Ander RA, Yerian LM, Tretiakova M, et al: cDNA microarray analysis of macroregenerative and dysplastic nodules in endstage hepatitis C virus-induced cirrhosis. Am J Pathol 162: 991-1000, 2003 Armstrong GL, Alter MJ, McQuillan GM, et al: The past incidence of hepatitis C virus infection: implications for the future burden of chronic liver disease in the United States. Hepatology 31: 777-782, 2000 Barba G, Harper F, Harada T, et al: Hepatitis C virus core protein shows a cytoplasmic localization and associates to cellular lipid storage droplets. Proc Natl Acad Sci USA 94:1200-1205, 1997 Bartenschlager R, Lohmann V: Replication of hepatitis C virus. J Gen Virol 81: 1631-1648, 2000 Bedossa P, Poynard T: The METAVIR cooperative study group. An algorithm for the grading of activity in chronic hepatitis C. Hepatology 24: 289-293, 1996 Bisgaard HC, Holmskov U, Santoni-Rugiu E, et al: Heterogeneity of ductular reactions in adult rat and human liver revealed by novel expression of deleted in malignant brain tumor 1. Am J Pathol 161: 1187-1198, 2002 Buendia MA: Genetics of hepatocellular carcinoma. Semin Cancer Biol 10: 185-200, 2000 Chang M, Williams O, Mittler J, Qiuntanilla A, Carithers RL, Perkins J, Corey L, Gretch DR: Dynamics of hepatitis C virus replication in human liver. Am J Pathol 163: 433-444, 2003 Choi J, Xu Z, Ou J-H: Triple decoding of hepatitis C virus RNA by programmed translational frameshifting. Mol Cell Biol 23: 1489-1497, 2003 Crowson AN, Nuovo G, Ferri C, et al: The dermatopathologic manifestations of hepatitis C infection: A clinical, histological, and molecular assessment of 35 cases. Hum Pathol 34: 573- 579, 2003 De Martino L, Sampaolo S, Tucci C, et al: Viral RNA in nerve tissues of patients with hepatitis C infection and peripheral neuropathy. Muscle and Nerve 27: 102-104, 2003 De Vos R, Verslype C, Depla E, et al: Ultrastructural visualization of hepatitis C virus components in human and primate liver biopsies. J Hepatol 37: 370-379, 2002 Di Bisceglie AM: Hepatitis C – Virology and future antiviral targets. Am J Med 107: 45S-48S, 1999 Dudas J, Kovalszky I, Gallai M, et al: Expression of decorin, transforming growth factor beta 1, tissue inhibitor metalloproteinase 1, 2 and type IV collagenases in chronic hepatitis. Am J Clin Path 115: 725-735, 2001 Edamoto Y, Hara A, Biernat W, et al: Alterations of RB1, p53 and Wnt pathways in hepatocellular carcinomas associated with hepatitis C, hepatitis B and alcoholic liver cirrhosis. Int J Cancer 106: 334-341, 2003 Forbes S, Vig P, Poulsom R, et al: Hepatic stem cells. J Pathol 197: 510-518, 2002 Gao G, Buskel Z, Seef L et al: Drift in the hypervariable region of the hepatitis C virus during 27 years in two patients. J Med Virol 68: 60-67, 2002 Gardner JP, Durso RJ, Arrigale RR, et al: L-sign, CD 209L, is a liver-specific capture receptor for hepatitis C virus. Proc Natl Acad Sci USA 100: 4498-4503, 2003 Gasztonyi B, Par A, Kiss K, et al: Activation of the nuclear factor kappa B – key role in oncogenesis? Chronic hepatitis C virus infection and lymphomagenesis. Orv Hetil 144: 863-868, 2003 Gervain J, Simon G, Papp I, et al: Analysing the type and subtype of hepatitis virus C of chronic viral hepatitis patients in Hungary. Orv Hetil 142: 1315-1319, 2001 Gervain J, Czibula A, Simon J, et al: Structure analysis of the PKR-binding region of HCV 1b samples from patients with chronic hepatitis C and the correlation with IFN-sensitivity. Orv Hetil 144: 1179-1184, 2003 He Y, Katze MG: To inferfere and to anti-interfere: The interplay between hepatitis C virus and interferon. Viral Immunol 15: 95-119, 2002 Houghton M: Hepatitis C viruses. In: Fields Virology, ed 3., Eds: Fields BN, Knipe DM, Howley PM), Lippincott-Raven, Philadelphia, 1996, pp. 1035-1068 International Hepatology Informatics Group: Diseases of the Liver and Biliary Tract. Standardization of Nomenclature, Diagnostic Criteria, and Prognosis. Raven Press, New York, 1994. Ishak K, Baptista A, Bianchi L, et al: Histological grading and staging of chronic hepatitis. J Hepatol 22: 696-699, 1995 Iwarson S, Schaff Zs, Seto BJ, et al: Retrovirus-like particles in hepatocytes of patients with transfusion-aquired non-A, non-B hepatitis. J Med Virol 16: 37-45, 1985 Jarmay K, Gallai M, Karacsony G, et al: Decorin and actin expression and distribution in chronic hepatitis C following interferon-alfa-2b treatment. J Hepatol 32: 993-1002, 2000 Kapadia SB, Brideau-Andersen A, Chisari FV: Interference of heptitis C virus RNA replication by short interfering RNAs. Proc Natl Acad Sci USA 100: 2014-2018, 2003 Knodell RG, Ishak KG, Black WC, et al: Formulation and application of a numerical scoring system for assessing histological activity in asymptomatic chronic active hepatitis. Hepatology 1: 431-435, 1981 Korbling M, Katz RL, Khanna A, et al:Hepatocytes and epithelial cells of donor origin in recipients of peripheral-blood stem cells. N Engl J Med 346: 738-746, 2002 Lotz G, Kiss A, Kaposi Novak P, et al: Hepatitis viruses and hepatocarcinogenesis. J Physiol-Paris 95: 417-422, 2001 Lotz G, Simon Zs, Szalay F, et al: Localization of hepatitis C virus RNA on human red blood cells by RT-in situ PCR technique. Scand J Gastroenterol 37: 578-584, 2002 Lowes KN, Brennan BA, Yeoh GC, et al: Oval cell numbers in human chronic liver diseases are directly related to disease severity. Am J Pathol 154: 537-541, 1999. Mihm S, Fayyazi A, Hartmann H, et al: Analysis of histopathological manifestations of chronic hepatitis C virus infection with respect to virus genotype. Hepatology 25: 735-739, 1997. Muller HM, Pfaff E, Goeser T, et al: Peripheral blood leucocytes serve as a possible extrahepatic site for hepatitis C virus replication. J Gen Virol 74: 669-676, 1993. Nagy P,Schaff Zs, Lapis K: Immunohistochemical detection of transforming growth factor-ß1 in fibrotic liver diseases. Hepatology 14: 269-273, 1991 Paku S, Schnur J, Nagy P, et al: Origin and structural evolution of the early proliferating oval cells in rat liver. Am J Pathol 158: 1313-1323, 2001 Par A: Hepatitis C virus, HCV, infection – after 12 years. Advances in the management of chronic hepatitis C. Orv Hetil 143: 2667-2674, 2002 Par A, Telegdy L, Dalmi L, et al: Therapy for chronic hepatitis C. J Physiol 95: 399-405, 2001 Pileri P, Uematsu Y, Campagnoli S, et al: Binding of hepatitis C virus to CD81. Science 282: 938-941, 1998 Pina Dore M, Realdi G, Mura D, et al: Genomic instability in chronic viral hepatitis and hepatocellular carcinoma. Hum Pathol 32: 698-703, 2001 Podanyi B: Extrahepatic manifestations and skin diseases associated with hepatitis C virus infection. Orvoskepzes 1: 1-48, 2003 Poynard T, Bedossa P, Opolon P: Natural history of liver fibrosis progression in patients with chronic hepatitis C. Lancet 349: 825-832, 1997 Ramalho F, Costa A, Pires A, et al: Correlation of genotypes and route of transmission with histologic activity and disease stage in chronic hepatitis C. Digest Dis Sci 45: 182-187, 2000 Rozario R, Ramakrishna B: Histopathological study of chronic hepatitis B and C: a comparison of two scoring systems. J Hepatol 38: 223-229, 2003 Rubbia-Brandt L, Leandro G, Spahr L, et al: Liver steatosis in chronic hepatitis C: a morphological sign suggesting infection with HCV genotype 3. Histopathology 39: 119-124, 2001 Scarselli E, Ansuini H, Cerino R, et al: The human scavenger receptor class B type I is a novel candidate receptor for the hepatitits C virus. EMBO J 21: 5017-5025, 2002 Schaff Zs, Hsia CC, Sarosi I, et al: Overexpression of transforming growth factor-alpha in hepatocellular carcinoma and focal nodular hyperplasia from European patients. Hum Pathol 25: 644-651, 1994 Schaff Zs, Lotz G, Eder G, et al: Pathomorphology and apoptosis in viral hepatitis. In: Therapies for Viral Hepatitis., Eds: Schinazi RF, Sommadossi JP, Thomas H), Int. Med. Press Ltd, London, 1998, pp. 77-86 Schaff Zs, Tabor E, Jackson DR, et al: Ultrastructural alterations in serial liver biopsy specimens from chimpanzees experimentally infected with a human non-A, non-B hepatitis agent. Virchows Arch B 45: 301-312, 1984 Scheuer PJ: Assessment of liver biopsies in chronic hepatitis: how is it best done? J Hepatol 38: 240-242, 2003 Scheuer PJ, Davies SE, Dhillon AP: Histopathological aspects of viral hepatitis. J Viral Hep 3: 277-283, 1996 Serfaty L, Andreani T, Giral P, et al: Hepatitis C virus induced hypobetalipoproteinemia: a possible mechanism for steatosis in chronic hepatitis C. J Hepatol 34: 428-434, 2001 Shimotohno K: Hepatitis C virus and its pathogenesis. Semin Cancer Biol 10: 233-240, 2000 Smith MW, Yue ZN, Geiss GK, et al: Identification of novel tumor markers in hepatitis C virus-associated hepatocellular carcinoma. Cancer Res 63: 859-864, 2003 Tabor E: Hepatocellular carcinoma: global epidemiology. Digest Liver Dis 33: 115-117, 2001 Wanless IR, Nakashima E, Sherman M: Regression of human cirrhosis. Arch Pathol Lab Med 124: 1599-1607, 2000 Wasley A, Alter MJ: Epidemiology of hepatitis C: geographic differences and temporal trends. Semin Liver Dis 20: 1-16, 2000 WHO Global surveillance and control of hepatitis C. Report of a WHO consultation organized in collaboration with the Viral Hepatitis Prevention Board, Antwerp, Belgium. J Viral Hepatol 6: 35-47, 1999 Wilson JA, Jayasena S, Khvorova A et al: RNA inferference blocks gene expression and RNA synthesis from hepatitis C replicons propagated in human liver cells. Proc Natl Acad Sci 100: 2783-2788, 2003 Yen T, Keeffe E, Ahmed A: The epidemiology of hepatitis C virus infection. J Clin Gastroenterol 36: 47-53, 2003. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2003 VL 9 IS 4 BP 215 EP 221 PG 7 ER PT J AU Keresztes, K Takacs, M Horanyi, M Miltenyi, Zs Illes, AF Keresztes, Katalin Takacs, Maria Horanyi, Margit Miltenyi, Zsofia Illes, Arpad TI HCV and HGV Infection in Hodgkin's Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hodgkin’s disease; hepatitis C virus; hepatitis G virus ID Hodgkin’s disease; hepatitis C virus; hepatitis G virus AB Numerous observations imply that the pathogenesis of malignant lymphomas is multifactorial and that viruses probably play an important etiologic role. Besides Epstein-Barr virus, there might be other viruses among the causes of Hodgkin's disease. A total of 111 randomly selected patients with Hodgkin's disease were included in this study, and hepatitis C and G viruses were tested with polymerase chain reaction. The results were compared to hepatitis C and G virus infection ratios assessed by polymerase chain reaction in the Hungarian blood bank. Hepatitis C virus was diagnosed in 10 (9%) patients, and hepatitis G virus in 9 (8,1%), which is a 12-fold and a 1,5-fold infection rate as compared to that of the Hungarian blood bank, respectively. There was no significant difference between hepatitis positive and negative patients concerning mean age at the time of diagnosis, sex, disease stage, histology type, treatment applied, risk factors in the history of the infection and liver enzymes. Hepatitis C virus positivity in patients with Hodgkin's disease differs significantly from that in blood donors. Based on these results and data in the literature, no definite statement can be made on the etiological role of viruses, but further studies are needed. C1 [Keresztes, Katalin] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22., H-4004 Debrecen, Hungary. [Takacs, Maria] Bela Johan National Institute of Hygiene, Department of VirologyBudapest, Hungary. [Horanyi, Margit] OGYK, Virus Nucleic Acid LaboratoryBudapest, Hungary. [Miltenyi, Zsofia] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22., H-4004 Debrecen, Hungary. [Illes, Arpad] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22., H-4004 Debrecen, Hungary. RP Keresztes, K (reprint author), University of Debrecen, Medical and Health Center, 3rd Department of Medicine, H-4004 Debrecen, Hungary. EM katalin@iiibel.dote.hu CR Barna TK, Ozsvar Z, Szendrenyi V, et al: Hepatitis C virus antibody in the serum of blood donors. Orv Hetil 10: 507-511, 1996 Cividini A, Rebucci C, Silini E, et al: Is the natural history of hepatitis C virus carriers with normal aminotransferase really benign? Gastroenterology 121: 1526-1527, 2001 Fuchs K, Motz M, Schreier E, et al: Characterization of nucleotide sequences from European hepatitis C virus isolates. Gene 103: 163-169, 1991 Gasztonyi B, Par A, Szomor A, et al: Hepatitis C virus infection associated with B-cell non-Hodgkin’s lymphoma in Hungarian patients. Br J Haemat 110: 498-499, 2000 Halasz R, Weiland O, and Sallberg M: GB virus C/hepatitis G virus. Scand J Infect Dis 33: 572-580, 2001 Jarrett RF, and MacKenzie J: Epstein-Barr virus and other candidate viruses in the pathogenesis of Hodgkin’s disease. Sem Haematol 36: 26-29, 1999 Keenan RD, Harrison P, Joffre L, et al: Hepatitis G virus, HGV, and lymphoproliferative disorders. Br J Haematol 99: 710, 1997 Khudyakov YE, Cong ME, Bonafonte MT, et al: Sequence variation within a nonstructural region of the hepatitis G virus genome. J of Virology 71: 6875-6880, 1997 Markovic S, Drozina G, Vovk M, et al: Reactivation of hepatitis B but not hepatitis C in patients with malignant lymphoma and immunosuppressive therapy. A prospective study in 305 patients. Hepatogastroenterology 46: 2925-2930, 1999 Par A, Telegdy L, Gogl A, et al: Interferon therapy of chronic viral hepatitis in Hungary: 5-year experience. A multicenter study. Orv Hetil 140: 1227-1233, 1999 Persico M, De Renzo A, Persico E, et al: Hepatitis G virus in patients with Hodgkin’s lymphoma. Br J Haematol 103: 1206, 1998 Pozzato G, Mazzaro C, Santini G, et al: Hepatitis C virus and non-Hodgkin’s lymphomas. Leuk Lymph 22: 53-60, 1996 Szabo A, Heemann U, Muller V, et al: Hepatitis G virus infection in adults and children after kidney transplantation. Orv Hetil 140: 1619-1623, 1999 Zuckerman E, Zuckerman T, Douer D, et al: Liver dysfunction in patients with hepatitis C virus undergoing chemotherapy for hematologic malignancies. Cancer 83: 1224-1230, 1998 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2003 VL 9 IS 4 BP 222 EP 225 PG 4 ER PT J AU Al-Moundhri, M Nirmala, V Al-Mawaly, K Ganguly, Sh Burney, I Rizvi, A Grant, Ch AF Al-Moundhri, Mansour Nirmala, Vadakkepat Al-Mawaly, K Ganguly, Shyam Burney, A. Ikram Rizvi, J. Azhar Grant, Christopher TI Significance of p53, Bcl-2, and HER-2/neu Protein Expression in Omani Arab Females with Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; p53; bcl-2; HER-2; arab-females ID breast cancer; p53; bcl-2; HER-2; arab-females AB Racial disparity in the presentation of breast cancer and the outcome of its treatment is well established. However, the causes remain unexplained. The scarcity of reports about the prognostic significance of p53, bcl-2, and HER-2/neu in Arab females with breast cancer has been the impetus to this study. We evaluated the prognostic significance of altered expression of p53, bcl-2, HER-2/neu in Omani Arab females with non-metastatic breast cancer with correlation to other established prognostic factors. We have retrospectively analyzed the immunohistochemical expression of p53, HER-2/neu and bcl-2 in paraffin embedded blocks of 72 females diagnosed with invasive breast cancer between 1992 and 2002. The expression of the above proteins was correlated with other prognostic factors and univariate and multivariate analysis was carried out for all prognostic factors. Overexpression of p53 significantly correlated with younger age (<40), pre-menopausal status, poor differentiation with inverse correlation with bcl-2 expression. Expression of bcl-2 immunopostivity significantly correlated to low histological grade and positive estrogen and progesterone receptor status. On univariate and multivariate p53 overexpression and lack of bcl-2 immunostaining resulted in worse survival outcome, but not Her-2/neu overexpression. Expression patterns of p53 and bcl-2 are independent predictors of survival in Omani Arab population which may help to stratify these patients into different risk groups. C1 [Al-Moundhri, Mansour] Sultan Qaboos University, Department of Medicine, Medical Oncology Unit, 123 Al-Khod, Oman. [Nirmala, Vadakkepat] Sultan Qaboos University, Department of PathologyMuscat, Oman. [Al-Mawaly, K] Sultan Qaboos University, Department of PathologyMuscat, Oman. [Ganguly, Shyam] Sultan Qaboos University, Department of Epidemiology and Medical StatisticsMuscat, Oman. [Burney, A. Ikram] Sultan Qaboos University, Department of Medicine, Medical Oncology Unit, 123 Al-Khod, Oman. [Rizvi, J. Azhar] Sultan Qaboos University, Department of Medicine, Medical Oncology Unit, 123 Al-Khod, Oman. [Grant, Christopher] Sultan Qaboos University, Department of Surgery College of MedicineMuscat, Oman. RP Al-Moundhri, M (reprint author), Sultan Qaboos University, Department of Medicine, Medical Oncology Unit, 123 Al-Khod, Oman. CR Rose DP, Royak-Schaler R: Tumor biology and prognosis in black breast cancer patients: a review. Cancer Detect Prev 25:16- 31, 2001 Ikpatt OF, Kuopio T, Collan Y: Proliferation in African breast cancer: biology and prognostication in Nigerian breast cancer material. Mod Pathol 15:78, 2002 Simon MS SR: Racial differences in breast cancer survival: the interaction of socio-economic status and tumor biology. Am J Obstet Gynecol 176:233-236, 1997 Cheng SH, Tsou MH, Liu MC, et al: Unique features of breast cancer in Taiwan. Breast Cancer Res Treat 63:213-23, 2000 Chow LW TA, Cheung KL, Au GK, et al: Current status of breast cancer in Hong Kong. Chin Med J, Engl, 110:474-8, 1997 Ezzat A RM, Zwaan F, Brigden M, et al: The lack of age as a significant prognostic factor in non-metastatic breast cancer. Eur J Surg Oncol 24:23-7, 1998 Fakhro AE FB, Al-Asheeri N, Al-Ekri SA: Breast cancer: patient characteristics and survival analysis at Salmaniya Medical Complex, Bahrain. Eastern Mediterranean Health Journal 5:430-439, 1999 Maalej M FH, Ben Salem S, Daoud J, et al: Breast cancer in Tunisia: clinical and epidemiological study. Bull Cancer 86:302- 6., 1999 Malik IA: Clinico-pathological features of breast cancer in Pakistan. J Pak Med Assoc 52:100-4, 2002 Slamon DJ, deKernion JB, Verma IM, et al: Expression of cellular oncogenes in human malignancies. Science 224:256-62, 1984 Bankfalvi A, Tory K, Kemper M, et al: Clinical relevance of immunohistochemical expression of p53-targeted gene products mdm-2, p21 and bcl-2 in breast carcinoma. Pathol Res Pract 196:489-501, 2000 Barbareschi M, Caffo O, Veronese S, et al: Bcl-2 and p53 expression in node-negative breast carcinoma: a study with long-term follow-up. Hum Pathol 27:1149-55, 1996 Ferrero-Pous M, Hacene K, Bouchet C, et al: Relationship between c-erbB-2 and other tumor characteristics in breast cancer prognosis. Clin Cancer Res 6:4745-54, 2000 Jansen RL, Joosten-Achjanie SR, Volovics A, et al: Relevance of the expression of bcl-2 in combination with p53 as a prognostic factor in breast cancer. Anticancer Res 18:4455-62, 1998 Le MG, Mathieu MC, Douc-Rasy S, et al: c-myc, p53 and bcl-2, apoptosis-related genes in infiltrating breast carcinomas: evidence of a link between bcl-2 protein over-expression and a lower risk of metastasis and death in operable patients. Int J Cancer 84:562-7, 1999 Nunes RA, Harris LN: The HER2 extracellular domain as a prognostic and predictive factor in breast cancer. Clin Breast Cancer 3:125-35, 2002 Ross JS, Fletcher JA: The HER-2/neu oncogene in breast cancer: prognostic factor, predictive factor, and target for therapy. Stem Cells 16:413-28, 1998 Sahin AA: Biologic and clinical significance of HER-2/neu, cerbB-2, in breast cancer. Adv Anat Pathol 7:158-66, 2000 Silvestrini R, Veneroni S, Daidone MG, et al: The Bcl-2 protein: a prognostic indicator strongly related to p53 protein in lymph node-negative breast cancer patients. J Natl Cancer Inst 86:499- 504, 1994 Sjogren S, Inganas M, Lindgren A, et al: Prognostic and predictive value of c-erbB-2 overexpression in primary breast cancer, alone and in combination with other prognostic markers. J Clin Oncol 16:462-9, 1998 Zhang GJ, Tsuda H, Adachi I, et al: Prognostic indicators for breast cancer patients with one to three regional lymph node metastases, with special reference to alterations in expression levels of bcl-2, p53 and c-erbB-2 proteins. Jpn J Clin Oncol 27:371-7, 1997 el-Ahmady O, el-Salahy E, Mahmoud M, et al: Multivariate analysis of bcl-2, apoptosis, p53 and HER-2/neu in breast cancer: a short-term follow-up. Anticancer Res 22:2493-9, 2002 Temmim L, Baker H, Sinowatz F: Immunohistochemical detection of p53 protein expression in breast cancer in young Kuwaiti women. Anticancer Res 21:743-8, 2001 Doglioni C, Dei Tos AP, Laurino L, et al: The prevalence of BCL-2 immunoreactivity in breast carcinomas and its clinicopathological correlates, with particular reference to oestrogen receptor status. Virchows Arch 424:47-51, 1994 al-Lawati JA, Santhosh-Kumar CR, Mohammed AJ, et al: Cancer incidence in Oman, 1993-1997. East Mediterr Health J 5:1035-41, 1999 Rosanelli GP, Steindorfer P, Wirnsberger GH, et al: Mutant p53 expression and DNA analysis in human breast cancer comparison with conventional clinicopathological parameters. Anticancer Res 15:581-6, 1995 Fresno M, Molina R, Perez del Rio MJ, et al: p53 expression is of independent predictive value in lymph node-negative breast carcinoma. Eur J Cancer 33:1268-74, 1997 Bosari S, Lee AK, Viale G, et al: Abnormal p53 immunoreactivity and prognosis in node-negative breast carcinomas with longterm follow-up. Virchows Arch A Pathol Anat Histopathol 421:291-5, 1992 Quinn CM, Ostrowski JL, Harkins L, et al: Loss of bcl-2 expression in ductal carcinoma in situ of the breast relates to poor histological differentiation and to expression of p53 and c-erbB-2 proteins. Histopathology 33:531-6, 1998 Hellemans P, van Dam PA, Weyler J, et al: Prognostic value of bcl-2 expression in invasive breast cancer. Br J Cancer 72:354- 60, 1995 Ioachim EE, Malamou-Mitsi V, Kamina SA, et al: Immunohistochemical expression of Bcl-2 protein in breast lesions: correlation with Bax, p53, Rb, C-erbB-2, EGFR and proliferation indices. Anticancer Res 20:4221-5, 2000 Haldar S, Negrini M, Monne M, et al: Down-regulation of bcl-2 by p53 in breast cancer cells. Cancer Res 54:2095-7, 1994 Vairo G, Innes KM, Adams JM: Bcl-2 has a cell cycle inhibitory function separable from its enhancement of cell survival. Oncogene 13:1511-9, 1996 Miyashita T, Kitada S, Krajewski S, Horne WA, Delia D, Reed JC: Overexpression of the Bcl-2 protein increases the half-life of p21 Bax J Biol Chem 270:26049–26052, 1995 Thor AD, Berry DA, Budman DR, et al: erbB-2, p53, and efficacy of adjuvant therapy in lymph node-positive breast cancer. J Natl Cancer Inst 90:1346-60, 1998 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2003 VL 9 IS 4 BP 226 EP 231 PG 6 ER PT J AU Arista-Nasr, J Martinez-Benitez, B Valdes, S Hernandez, M Bornstein-Quevedo, L AF Arista-Nasr, Julian Martinez-Benitez, Braulio Valdes, Samuel Hernandez, Mercedes Bornstein-Quevedo, Leticia TI Pseudohyperplastic Prostatic Adenocarcinoma in Transurethral Resections of the Prostate SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE pseudohyperplastic prostatic adenocarcinoma; diagnosis ID pseudohyperplastic prostatic adenocarcinoma; diagnosis AB Pseudohyperplastic prostatic adenocarcinoma is a recently described variety of adenocarcinoma that has been studied in core-needle biopsies and prostatectomy specimens. It is characterized by malignant glands that simulate benign hyperplastic glands with complex, medium to large-sized glands with papillary infoldings, luminal undulations, branching or cystic dilatations, and columnar cells with macronucleoli and nuclear enlargement. Our aim was to define frequency, tumor volume, and histologic features of pseudohyperplastic prostatic adenocarcinoma in transurethral resections of prostate. We studied 250 specimens from transurethral resections; 150 specimens were originally diagnosed as benign glandular hyperplasia, and 100 as conventional prostate adenocarcinomas. Of the 150 biopsies originally diagnosed as benign glandular hyperplasia, two (1.3%) had areas of pseudohyperplastic carcinoma. In both cases the neoplasm was limited to two chips and measured 3 and 4 mm in diameter, respectively. Both patients were asymptomatic 2 and 4 years after diagnosis. Of the 100 biopsies with adenocarcinoma, areas of pseudohyperplastic carcinoma were found in three cases. In the first two these areas were found in two fragments, and in the other case they were found in three chips, and measured 3, 4, and 6 mm, respectively. The clinical course in these cases was unfavorable, and two patients had metastasis. Main histologic findings included crowded glands (5/5), papillary projections (5/5), nuclear enlargement (5/5) macronucleoli (4/5) cystic glandular dilatation (4/5) straight luminal borders (4/5), pink amorphous secretions (4/5) nuclear hyperchromasia (3/5) and transition to small acinar pattern of adenocarcinoma (3/5). In conclusion, pseudohyperplastic prostate carcinoma is rare in transurethral resection specimens and is found in scarce chips. Frequency of false negative results in biopsies originally diagnosed as benign glandular hyperplasia was 1.3%. In biopsies diagnosed as carcinoma, this frequency was 3%. These patients had an adverse clinical course, apparently due to association with areas of conventional adenocarcinoma. C1 [Arista-Nasr, Julian] Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No. 15, 14000 Tlalpan, Mexico. [Martinez-Benitez, Braulio] Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No. 15, 14000 Tlalpan, Mexico. [Valdes, Samuel] Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No. 15, 14000 Tlalpan, Mexico. [Hernandez, Mercedes] Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No. 15, 14000 Tlalpan, Mexico. [Bornstein-Quevedo, Leticia] Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No. 15, 14000 Tlalpan, Mexico. RP Arista-Nasr, J (reprint author), Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Pathology, 14000 Tlalpan, Mexico. EM pipa5@hotmail.com CR Muir EG. Carcinoma of the prostate. Lancet 1:667-72,1934 Willis RA. Pathology of tumors. ST. Louis: CV Mosby; 1948, p587. Ackerman LV.Surgical Pathology. St. Louis: CV Mosby;1964:651. Humphrey PA, Kaleem Z, Swanson PE, Vollmer RT. Pseudohyperplatic prostatic adenocarcinoma. Am J Surg Pathol 22: 1239-46, 1998 Levi A, Epstein JI. Pseudohyperplastic prostatic adenocarcinoma on needle biopsy and simple prostatectomy. Am J Surg Pathol 24:1039-46,2000 Arista-Nasr, Cortes E, Picardo R. Low grade adenocarcinoma simulating benign glandular lesions in needle prostatic biopsy. Rev Invest Clin 49:37-40,1997 Wojno KJ, Epstein JI.The utility of basal cell specific anticytokeratin antibody, 34 beta E12, in the diagnosis of prostate cancer: a review of 228 cases. Am J Surg Pathol 19:251-60,1995 Epstein JI. Variants of prostatic adenocarcinoma. In: Prostate Biopsy Interpretation. New York, NY: Lippincott-Raven, 1989, pp169-73. Epstein JI. Evaluation of transurethral resection specimens. In: Prostate Biopsy Interpretation. Philadelphia: Lippincott-Raven, 1995, pp143-4. Bostwick DG Amin MB, Dundore P et al. Architectural patterns of high grade prostatic intraepithelial neoplasia. Hum Pathol 24:298- 310,1993 Bostwick DG, Srigely J, Grignon D et al. Atypical adenomatous hyperplasia of the prostate: morphological criteria for its distinction from well differentiated carcinoma. Hum Pathol 24:819- 32,1993 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2003 VL 9 IS 4 BP 232 EP 235 PG 4 ER PT J AU Kamory, E Kolacsek, O Otto, Sz Csuka, O AF Kamory, Eniko Kolacsek, Orsolya Otto, Szabolcs Csuka, Orsolya TI hMLH1 and hMSH2 Somatic Inactivation Mechanisms in Sporadic Colorectal Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE sporadic colorectal cancer; mismatch repair; hMLH1; hMSH2; microsatellite instability; allelic imbalance; promoter hypermethylation and inactivation ID sporadic colorectal cancer; mismatch repair; hMLH1; hMSH2; microsatellite instability; allelic imbalance; promoter hypermethylation and inactivation AB Much is known about the role of germline inactivation in mismatch repair (MMR) genes in hereditary non-polyposis colorectal cancer (HNPCC), but the impact of somatic MMR gene changes on sporadic colorectal cancer remains to be elucidated. In hereditary cases the hMLH1 and hMSH2 genes were shown to have a great importance, and in order to examine the somatic inactivation mechanisms of the two MMR genes hMLH1 and hMSH2 we screened 37 Hungarian sporadic colorectal cancer patients for allelic imbalance (AI), microsatellite instability (MSI), hMLH1 promoter hypermethylation and somatic mutations. Thirteen of the examined tumours (35%) were characterized by low-level MSI and none of the cases belonged to the high MSI group. Nine (24%) and seven (19%) cases had AI at the hMLH1 and hMSH2 genes, respectively. Seven tumours (19%) showed dense promoter hypermethylation of hMLH1, but only two patients had somatic mutations, one for each MMR gene. According to our study on this limited set of cases the most prominent mismatch repair inactivation mechanism in sporadic colorectal cancer patients is the hMLH1 promoter hypermethylation which may have a role in the carcinogenesis of sporadic colorectal cancer. C1 [Kamory, Eniko] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Kolacsek, Orsolya] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Otto, Szabolcs] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Csuka, Orsolya] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. RP Csuka, O (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, H-1122 Budapest, Hungary. EM csuka@oncol.hu CR Konishi M, Kikuchi-Yanoshita R, Tanaka K, et al: Molecular nature of colon tumors in hereditary nonpolyposis colon cancer, familial polyposis, and sporadic colon cancer. Gastroenterology 111: 307-317, 1996. Bevilacqua RAU, Simpson AJG: Methylation of the hMLH1 promoter but no hMLH1 mutations in sporadic gastric carcinomas with high-level microsatellite instability. Int J Cancer 87: 200-203, 2000. Kastan MB: Genetic Instability and Tumorigenesis. Springer- Verlag Berlin Heidelberg, 1997. Fishel R, Lescoe MK, Rao MRS, et al: The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell 75: 1027-1038, 1993. Bronner CE, Baker SM, Morrison PT, et al: Mutation in the DNA mismatch repair gene homologue hMLH1 is associated with hereditary non-polyposis colon cancer. Nature 368: 258-261, 1994. Hemminki A, Peltomaki P, Mecklin JP, et al: Loss of the wild type MLH1 gene is a feature of hereditary nonpolyposis colorectal cancer. Nat Genet 8: 405-410, 1994. Kolodner RD, Hall NR, Lipford J, et al: Structure of the human MLH1 locus and analysis of a large hereditary nonpolyposis colorectal carcinoma kindred for mlh1 mutations. Cancer Res 55: 242-248, 1995. Parsons R, Li GM, Longley M, et al: Mismatch repair deficiency in phenotipically normal human cells. Science 268: 738-740, 1995. Wong IHN: Methylation profiling of human cancers in blood: Molecular monitoring and prognostication. Int J Oncol 19: 1319-1324, 2001. Kondo E, Furukawa T, Yoshinaga K, et al: Not hMSH2 but hMLH1 is frequently silenced by hypermethylation in endometrial cancer but rarely silenced in pancreatic cancer with microsatellite instability. Int J Oncol 17: 535-541, 2000. Esteller M, Levine R, Baylin SB, et al: hMLH1 promoter hypermethylation is associated with microsatellite instability phenotype in sporadic endometrial carcinoma. Oncogene 16: 2413- 2417, 1998. Cunningham JM, Christensen ER, Tester DJ, et al: Hypermethylation of the hMLH1 promoter in colon cancer with microsatellite instability. Cancer Res 58: 3455-3460, 1998. Maniatis T, Fritsch EF, Sambrook J: Molecular Cloning, A Laboratory Manual. Cold Spring Harbor Laboratory, New York, USA, 1982. Tomlinson IPM, Lambros MBK, Roylance RR: Loss of heterozygosity analysis: practically and conceptually flawed? Genes Chromosomes Cancer 34: 349-353, 2002. Boland CR, Thibodeau SN, Hamilton SR, et al: A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58: 5248-5257, 1998. Kane MF, Loda M, Gaida GM, et al: Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic colon tumors and mismatch repair-defective human tumor cell lines. Cancer Res 57: 808-811, 1997. Yanagisawa Y, Akiyama Y, Iida S, et al: Methylation of the hMLH1 promoter in familial gastric cancer with microsatellite instability. Int J Cancer 85: 50-53, 2000. Beck NE, Tomlinson IPM, Homfray T, et al: Use of SSCP analysis to identify germline mutations in HNPCC families fulfilling the Amsterdam criteria. Hum Genet 99: 219-224, 1997. Parc YR, Halling KC, Wang L, et al: HMSH6 alterations in patients with microsatellite instability-low colorectal cancer. Cancer Res 60: 2225-2231, 2000. Kuismanen SA, Holmberg MT, Salovaara R, et al: Genetic and epigenetic modification of MLH1 accounts for a major share of microsatellite-unstable colorectal cancers. Am J Pathol 56: 1773-1779, 2000. Furukawa T, Konishi F, Masubuchi S, et al: Densely methylated MLH1 promoter correlates with decreased mRNA expression in sporadic colorectal cancers. Genes Chromosomes Cancer 35: 1-10, 2002. Porfiri E, Scartozzi M, Piga A, et al: Missense mismatch repair gene alterations, microsatellite instability, and hereditary nonpolyposis colorectal cancer. J Clin Oncol 20: 3178- 3179, 2002. Brueckl WM, Limmert T, Brabletz T, et al: Mismatch repair deficiency in sporadic synchronous colorectal cancer. Anticancer Res 20: 4727-4732, 2000. Toyota M, Ohe-Toyota M, Ahuja N, et al: Distinct genetic profiles in colorectal tumors with or without the CpG island methylator phenotype. PNAS 97: 710-715, 2000. Han HJ, Maruyama M, Baba S, et al: Genomic structure of human mismatch repair gene, hMLH1, and its mutation analysis in patients with hereditary non-polyposis colorectal cancer, HNPCC). Hum Mol Genet 14: 237-242, 1995. Maliaka YK, Chudina AP, Belev NF, et al: CpG dinucleotides in the hMSH2 and hMLH1 genes are hotspots for HNPCC mutations. Hum Genet 97: 251-255, 1996. http://www.nfdht.nl/database/mdbchoice.htm) Liu T, Wahlberg S, Rubio C, et al: DGGE screening of mutations in mismatch repair genes, hMSH2 and hMLH1, in 34 Swedish families with colorectal cancer. Clin Genet 53: 131- 135, 1998. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2003 VL 9 IS 4 BP 236 EP 241 PG 6 ER PT J AU Bilgic, B Mete, Ozturk, AS Demiryont, M Keles, N Basaran, M AF Bilgic, Bilge Mete, Ozgur Ozturk, A Settar Demiryont, Misten Keles, Nesil Basaran, Mert TI Synovial Sarcoma: a Rare Tumor of Larynx SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE larynx; synovial sarcoma ID larynx; synovial sarcoma AB Synovial sarcoma is a soft tissue sarcoma of unknown histogenesis and occurs predominantly in the lower extremities of young adults. The head and neck is a relative rare location. There are about 10 cases with laryngeal localization in the literature. We present a 24 year-old male with an endolaryngeal mass. Incisional biopsy and the hemilaryngectomy material revealed a biphasic synovial sarcoma. One year later a local recurrence occurred. Tumor excision and neck dissection were performed. Radiotherapy was added. Six months later lung metastases was discovered on thoracic CT. The patient received chemotherapy for 6 courses. The metastases responded well to chemotherapy and the patient is now alive without tumor on radiological and clinical examination after 3.5 years of follow-up. C1 [Bilgic, Bilge] Istanbul Medical Faculty, Department of Pathology, 34390 Istanbul, Turkey. [Mete, Ozgur] Istanbul Medical Faculty, Department of Pathology, 34390 Istanbul, Turkey. [Ozturk, A Settar] Istanbul Medical Faculty, Department of Pathology, 34390 Istanbul, Turkey. [Demiryont, Misten] Istanbul Medical Faculty, Department of Pathology, 34390 Istanbul, Turkey. [Keles, Nesil] Istanbul Medical Faculty, ENT SurgeryIstanbul, Turkey. [Basaran, Mert] Istanbul Medical Faculty, Istanbul University, Oncology InstituteIstanbul, Turkey. RP Bilgic, B (reprint author), Istanbul Medical Faculty, Department of Pathology, 34390 Istanbul, Turkey. EM eminbilgic@superonline.com CR Aubert S: Parapharyngeal synovial sarcoma. Ann Pathol 21: 71-75, 2001 Danninger R, Hurner U, Stammberger H: Das Synovialsarkom, ein seltener Tumor des Larynx. Falldarstellung und differentialdiagnostische Uberlegungen. Laryngorhinootologie 73: 442-444, 1994 Dei Tos A, Dal Cin P, Sciot R, et al: Ann Otol Rhinol Laryngol 107: 1080-1085, 1998 Ferlito A, Caruso G: Endolaryngeal synovial sarcoma: An update on diagnosis and treatment. ORL, J Otorhinolaryngol Relat Spec 53: 116-119, 1991 Garenstein A, Neel HB 3rd, Weiland LH, et al: Sarcomas of the larynx. Arch Otolaryngol 106: 8-12, 1980 Gatti WM, Strom CG, Orfei E: Synovial sarcoma of the laryngopharynx. Arch Otolaryngol 101: 633-636, 1975 Geahchan NE, Lambert J, Micheau C, et al: Synovialome malin du larynx. Ann Otolaryngol Chir Cervicofac 100: 61-65, 1983 Ianniello F, Ferri E, Armato E, et al: Carcinosarcoma of the larynx: immunohistochemical study, clinical considerations, therapeutic strategies. Acta Otorhinolaryngol Ital 21: 192-197, 2001 Kitsmaniuk ZD, Volkov I, Demochko VB, et al: Synovial sarcoma of the larynx. Vestn Otorinolaryngol; 2: 61-62, 1985 Kleinasser O: Tumors of the larynx and hypopharynx. Stuttgart Thieme 1988: pp 326-328 Miller LH, Santaella-Latimer L, Miller T: Synovial sarcoma of the larynx. Trans Am Acad Ophtalmol Otolaryngol 80: 448- 451, 1975 Morland B, Cox G, Randall C, et al: Synovial sarcoma of the larynx in a child: case report and histological appearances. Med Pediatr Oncol 23: 64-68, 1994 Pruszczynski M, Manni JJ, Smedt F: Endolaryngeal synovial sarcoma: Case report with immunohistochemical studies. Head Neck 11: 76-80, 1989 Quinn HJ: Synovial sarcoma of the larynx treated by partial laryngectomy. Laryngoscope 94: 1158-1161, 1984 Schneider-Stock R, Onnasch D, Haeckel C, et al: Prognostic significance of p53 gene mutations and p53 protein expression in synovial sarcoma. Virchows Arch 435: 407-412, 1999 Skytting B: Synovial sarcoma: A Scandinavian Sarcoma Group Project. Acta Orthopaedica Scandinavia. Supplementum 291: 1-28, 2000 Skytting BT, Bauer HC, Perfekt R, et al: Ki-67 is stongly prognostic in synovial sarcoma; analysis based on 86 patients from the Scandinavian Sarcoma Group Register. Br J Cancer 80: 1809-1814, 1999 Thompson LD: Diagnostically challenging lesions in head and neck pathology. Eur Arch Otorhinolaryngol 254: 357-366, 1997 Weiss SW, Goldblum JR: Malignant soft tissue tumors of uncertain type. In: Enzinger and Weiss’s Soft Tissue Tumors, 4th ed. Mosby St Louis 2001 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2003 VL 9 IS 4 BP 242 EP 245 PG 4 ER PT J AU Szendroi, A Rusz, A Szekely, E Riesz, P Kelemen, Zs AF Szendroi, Attila Rusz, Andras Szekely, Eszter Riesz, Peter Kelemen, Zsolt TI Renal Tumor Causing Haematuria and Sepsis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE haematuria; pregnancy; kidney tumor; Bellini duct cancer ID haematuria; pregnancy; kidney tumor; Bellini duct cancer AB A 28 year old female patient developed hematuria in the 32th week of her pregnancy. She was given antibiotic treatment, since a urinary tract infection was suspected. After delivery symptoms of acute pyelonephritis, then sepsis developed, and conservative therapy had no effect. Ultrasound examination showed unusual renal destruction, so nephrectomy was performed. Surgical intervention revealed the presence of an advanced tumor of the kidney, while histological examination confirmed a Bellini duct carcinoma of the kidney. C1 [Szendroi, Attila] Semmelweis University, Department of Urology, Ulloi ut 78/b, H-1078 Budapest, Hungary. [Rusz, Andras] Semmelweis University, Department of Urology, Ulloi ut 78/b, H-1078 Budapest, Hungary. [Szekely, Eszter] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Riesz, Peter] Semmelweis University, Department of Urology, Ulloi ut 78/b, H-1078 Budapest, Hungary. [Kelemen, Zsolt] Semmelweis University, Department of Urology, Ulloi ut 78/b, H-1078 Budapest, Hungary. RP Szendroi, A (reprint author), Semmelweis University, Department of Urology, H-1078 Budapest, Hungary. EM atsszendroi@freemail.hu CR Antonelli A, Portesi E, Cozzoli A, et al: The collecting duct carcinoma of the kidney: a cytogenetical study. Eur Urol 43:680- 685, 2003 Belldegrun A, deKernion JB: Renal tumors In: Campbell’s Urology, 7th ed. Edited by PC Walsh, AB Retik, TA Stamey and ED Vaughan. Philadelphia: WB Saunders Co., vol.3, Chapt 76, pp. 2283-2327, 1998 Cheville JC, Lohse CM, Zincke H, et al: Comparisons of outcome and prognostic features among histologic subtypes of renal cell carcinoma Am J Surg Pathol 27:612-24, 2003 El Fettouh HIA, Cherullo E, El-Jack M, et al: Sporadic renal cell carcinoma in young adults: presentation, treatment and outcome. Urology 60: 806-810, 2002 Kovacs G, Akhtar M, Beckwith BJ, et al: The Heidelberg classification of renal cell tumors. J Path 183:131, 1997 Mejean A, Roupret M, Larousserie F,et al: Is there a place for radical nephrectomy in the presence of metastatic collecting duct, Bellini, carcinoma? J Urol 169:1287-90, 2003 Mickisch GHJ: Rational selection of a control arm for randomised trials in metastatic renal cell carcinoma Eur Urol 43:670-679, 2003 Poel van der H.G, Roukema JA, Horenblas S, et al: Metastectomy in renal cell carcinoma: a multicenter retrospective analysis. Eur Urol 35:197-203, 1999 Romics I, Goepel M: A here es vesedaganatok diagnosztikaja es terapiaja. Akademiai Kiado, 1998 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2003 VL 9 IS 4 BP 246 EP 248 PG 3 ER PT J AU Vargas-Gonzalez, R Solis-Coria, A AF Vargas-Gonzalez, Roberto Solis-Coria, Araceli TI Giant Cell Fibroblastoma in a 3-Year-Old Boy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE tumor; giant cell; giant cell fibroblastoma; dermatofibrosarcoma protuberans ID tumor; giant cell; giant cell fibroblastoma; dermatofibrosarcoma protuberans AB Giant cell fibroblastoma (GCF) is a rare soft tissue tumor most often discovered during the first two decades of life. We present a case of a 3-year-old boy with a history of a recurrent lesion in the knee, the tumor growth progressively and enlarged to 2.1 cm in the previous two years before diagnosis. It involved the subcutaneous tissue, had infiltrative borders and extended into the superficial dermis. The tumor was surgically excised with free margins. There was no evidence of local recurrence, and a metastatic workup was negative after 10 years of follow up. We review herein the clinicopathologic features, histogenesis, differential diagnosis and relationship to dermatofibrosarcoma protuberans (DFSP). C1 [Vargas-Gonzalez, Roberto] Hospital Para el Nino Poblano, Department of Pathology, Km 1.5 Carretera Federal Puebla-Atlixco, 72190 Puebla, Mexico. [Solis-Coria, Araceli] Hospital Para el Nino Poblano, Department of Pathology, Km 1.5 Carretera Federal Puebla-Atlixco, 72190 Puebla, Mexico. RP Vargas-Gonzalez, R (reprint author), Hospital Para el Nino Poblano, Department of Pathology, 72190 Puebla, Mexico. EM soncoy@msn.com CR Shmookler BM, Enzinger FM: Giant cell fibroblastoma: a peculiar childhood tumor. Lab Invest 46:76- xx, 1982. Shmookler BM, Enzinger FM, Weiss SW: Giant cell fibroblastoma: a juvenile form of dermatofibrosarcoma protuberans. Cancer 64:2154-61,1989. Kholova I, Ryska A, Dedic K: Composite tumor consisting of dermatofibrosarcoma protuberans and giant cell fibroblastoma associated with intratumoral endometriosis. Report of a case. Pathol Res Pract 197:263-7,2001. Maeda T, Hirose T, Furuya K, et al: Giant cell fibroblastoma associated with dermatofibrosarcoma protuberans: a case report. Mod Pathol 11:491-5,1998. Fletcher CD: Giant cell fibroblastoma of soft tissue: a clinicopathological and immunohistochemical study. Histopathology 13:499-508,1988. Weiss SW, Nickoloff BJ: CD 34 is expressed by a distinctive cell population in peripheral nerve, nerve sheath tumors, and related lesions. Am J Surg Pathol 17:1039.1993. Abdul-Karim W, Evans HL, Silva EG: Giant cell fibroblastoma: a report of three cases. Am J Clin Pathol 83:165-70,1985. Nguyen CM, Burch JM, Fitzpatrick JE, et al: Giant cell fibroblastoma in a child misdiagnosed as a dermatofibroma. Ped Dermatol 19:28-32,2002. Goldblum JR: Giant cell fibroblastoma a report of three cases with histologic and immunohistochemical evidence of a relationship to dermatofibrosarcoma protuberans. Arch Pathol Lab Med 120:1052-55,1996. Beham A, Fletcher CDM: Dermatofibrosarcoma protuberans with areas resembling giant cell fibroblastoma: report of two cases. Histopathology 17:165-7,1990. Cin PD, Sciot R, De Wever I, et al: Cytogenetic and immunohistochemical evidence that giant cell fibroblastoma is related to dermatofibrosarcoma protuberans. Genes Chromosomes Cancer 15:73,1996. Terrier-Lacombe MJ, Guillou L, Maire G, et al: Dermatofibrosarcoma protuberans, giant cell fibroblastoma and hybrid lesions in children: clinicopathologic comparative analysis of 28 cases with molecular data-a study from the French federation of cancer centers sarcoma group. Am J Surg Pathol 27:27- 39,2003. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2003 VL 9 IS 4 BP 249 EP 251 PG 3 ER PT J AU Zalatnai, A AF Zalatnai, Attila TI Pancreatic Cancer - a Continuing Challenge in Oncology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE pancreatic cancer; risk factors; PanIN; hormones; chemoprevention; review ID pancreatic cancer; risk factors; PanIN; hormones; chemoprevention; review AB Pancreatic cancer is still one of the major health problems because of its rising incidence and the modest therapeutic results. The paper surveys the statistical data, the risk factors, the preneoplastic ductal lesions, the hormonal sensitivity, the possible transdifferentiation in the endocrine and exocrine parts and the possibilities for chemoprevention. Hungary is peculiar among the European countries because during the last 50 years the incidence of pancreatic cancer has displayed a 15-fold increase. Apart from smoking, additional risk factors seem to be important, and recently a puzzling association between Helicobacter pylori seropositivity and pancreatic cancer was found. First-degree relatives of patients with pancreatic cancer are also at increased risk of this tumor. The term pancreatic intraepithelial neoplasia (PanIN) seems yet to be established, but the dynamics of these lesions needs to be further elucidated. Several lines of firmly established data indicate the hormonal sensitivity of this tumor, but still an unexplained discrepancy exists between the experimental and the clinical results. In addition to the somatostatin analogs, anti-gastrin vaccine is being tested. The mixed exocrine-endocrine tumors might suggest a real possibility of transdifferentiation between different compartments of the pancreas. Finally, the paper outlines the available data about the possibility of chemoprevention, including the role of cyclooxygenase inhibitors. C1 [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Zalatnai, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM zalatnai@korb1.sote.hu CR Ahmad NA, Lewis JD, Ginsberg GG, et al: Long term survival after pancreatic resection for pancreatic adenocarcinoma. Am J Gastroenterol 96: 2609-2615, 2001 Alanen KA, Joensuu: Long-term survival after pancreatic adenocarcinoma – often a misdiagnosis? Br J Cancer 68: 1004- 1005, 1993 Anderson KE, Hammons GJ, Kadlubar FF, et al: Metabolic activation of aromatic amines by human pancreas. Carcinogenesis 18: 1085-1092, 1997 Anderson KE, Johnson TW, Lazovich D, et al: Association between nonsteroidal anti-inflammatory drug use and the incidence of pancreatic cancer. J Natl Cancer Inst 94: 1168-1171, 2002 Anderson KE, Sinha R, Kulldorff M, et al: Meat intake and cooking techniques: associations with pancreatic cancer. Mutat Res 30: 225-231, 2002 Anttila A, Pukkala E, Sallmen M, et al: Cancer incidence among Finnish workers exposed to halogenated hydrocarbons. J Occup Environ Med 37: 797-806, 1995 Arundel SE, Kinnier-Wilson LM: Parental occupations and cancer: a review of the literature. J Epidemiol Community Health 40: 30-36, 1986 Biankin AV, Kench JG, Morey AL, et al: Overexpression of p21, WAF1/CIP1, is an early event in the development of pancreatic intraepithelial neoplasia. Cancer Res 61: 8830-8837, 2001 Bouwens L: Transdifferentiation versus stem cell hypothesis for the regeneration of islet beta-cells in the pancreas. Microsc Res Tech 43: 332-336, 1998 Boyle P, Maisonneuve P, Bueno de Mesquita B, et al: Cigarette smoking and pancreas cancer: a case control study of the search programme of the IARC. Int J Cancer 67: 63-71, 1996 Brat DJ, Lillemoe KD, Yeo CJ, et al: Progression of pancreatic intraductal neoplasias to infiltrating adenocarcinoma of the pancreas. Am J Surg Pathol 22: 163-169, 1998 Brett BT, Smith JC, Bouvier CV, et al: Phase II study of antigastrin- 17 antibodies, raised to G17DT, in advanced pancreatic cancer. J Clin Oncol 20: 4225-4235, 2002 Burghardt B, Barabas K, Marcsek Z, et al: Inhibitory effect of a long-acting somatostatin analogue on EGF-stimulated cell proliferation in Capan-2 cells. J Physiol Paris 94: 57-62, 2000 Buscail L, Saint-Laurent N, Chastre E, et al: Loss of ss2 somatostatin receptor gene expression in human pancreatic and colorectal cancer. Cancer Res 56: 1823-1827, 1996 Cantor KP, Silberman W: Mortality among aerial pesticide applicators and flight instructors: follow-up from 1965-1988. Am J Ind Med 36: 239-247, 1999 Chen J, Baithun SI, Ramsay MA: Histogenesis of pancreatic carcinomas: a study based on 248 cases. J Pathol 146: 65-76, 1985 Cheung NW, Boyages SC: Somatostatin-14 and its analog octreotide exert a cytostatic effect on GH3 rat pituitary tumor cell proliferation via a transient G0/G1 cell cycle block. Endocrinology, 136: 4174-4181, 1995 Clary T, Ritz B: Pancreatic cancer mortality and organochlorine pesticide exposure in California, 1989-1996. Am J Ind Med 43: 306-313, 2003 Cottliar AS, Fundia AF, Moran C, et al: Evidence of chromosome instability in chronic pancreatitis. J Exp Clin Cancer 19: 513-517, 2000 Cubilla AL, Fitzgerald PJ: Morphological lesions associated with human primary invasive nonendocrine pancreas cancer. Cancer Res 36: 2690-2698, 1976 Curphey TJ, Kuhlmann ET, Roebuck BD, et al: Inhibition of pancreatic and liver carcinogenesis in rats by retinoid- and selenium-supplemented diets. Pancreas 3: 36-40, 1988 Day JD, Digiuseppe JA, Yeo C, et al: Immunohistochemical evaluation of HER-2/neu expression in pancreatic adenocarcinoma and pancreatic intraepithelial neoplasms. Hum Pathol 27: 119-124, 1996 De Gonzalez AB, Sweetland S, Spencer E: A meta-analysis of obesity and the risk of pancreatic cancer. Br J Cancer 89: 519- 523, 2003 Delesque N, Buscail L, Esteve JP, et al: Sst2 somatostatin receptor expression reverses tumorigenicity of human pancreatic cancer cells. Cancer Res 57: 956-962, 1997 Dell L, Teta MJ: Mortality among workers at a plastics manufacturing and research and development facility: 1946-1988. Am J Ind Med 28: 373-384, 1995 Ding XZ, Adrian TE: Resveratrol inhibits proliferation and induces apoptosis in human pancreatic cancer cells. Pancreas 25: 71-76, 2002 Ding XZ, Hennig R, Adrian TE: Lipoxygenase and cyclooxygenase metabolism: new insights in treatment and chemoprevention of pancreatic cancer. Mol Cancer 2: 1-10, 2003 Ding XZ, Tong WG, Adrian TE: Blockade of cycloxygenase-2 inhibits proliferation and induces apoptosis in human pancreatic cancer cells. Anticancer Res 20: 2525-2631, 2000 Eibl G, Reber HA, Wente MN, et al: The selective cyclooxygenase- 2 inhibitor nimesulide induces apoptosis in pancreatic cancer cells independent of COX-2. Pancreas 26: 33-41, 2003 vanEijck CHJ, Lamberts SWJ, Lemaire LCJ, at al: The use of somatostatin receptor scintigraphy in the differential diagnosis of pancreatic duct cancers and islet cell tumors. Ann Surg 224: 119-124, 1996 Ekbom A, McLaughilin JK, Karlsson BM,et al: Pancreatitis and pancreatic cancer. A population-based study. J Natl Cancer Inst 86: 625-627, 1994 Engeland A: Trends in the incidence of smoking-associated cancers in Norway, 1954-93. Int J Cancer 68: 39-46, 1996 Ewing J: Neoplastic diseases. A treatise on tumors. 3rd ed., W.B. Saunders, Philadelphia, London, 1928 Faivre J, Forman D, Esteve J, et al: Survival of patients with primary liver cancer, pancreatic cancer and biliary tract cancer in Europe. Eur J Cancer 34: 2184-2190, 1998 Fazeny B, Baur M, Prohaska M, et al: Octreotide combined with goserelin in the therapy of advanced pancreatic cancer – results of a pilot study and review of the literature. J Cancer Res Clin Oncol 123: 45-52, 1997 Fenoglio CM, King DW: Somatostatin: an update. Human Pathol 14: 475-479, 1983 Fernandez E, La Vecchia C, Porta M, et al: Trends in pancreatic cancer mortality in Europe, 1955-1989. Int J Cancer 57: 786-792, 1994 Fisher WE, Muscarella P, Boros LG, et al: Gastrointestinal hormones as potential adjuvant treatment of exocrine pancreatic adenocarcinoma. Int J Pancreatol 24: 169-180, 1998 Fisher WE, Doran TA, Muscarella P, et al: Expression of somatostatin receptor subtype 1-5 genes in human pancreatic cancer. J Natl Cancer Inst 90: 322-324, 1998 Fueger BJ, Hamilton G, Raderer M, et al: Effects of chemotherapeutic agents on expression of somatostatin receptors in pancreatic tumor cells. J Nucl Med 42: 1856-1862, 2001 Garabrant DH, Held J, Langholz B, et al: DDT and related compounds and risk of pancreatic cancer. J Natl Cancer Inst 84: 764-771, 1992 Ghadirian P, Boyle P, Simard A, et al: Reported family aggregation of pancreatic cancer within a population-based casecontrol study in the Francophone community in Montreal, Canada. Int J Pancreatol 10: 183-196, 1991 Ginsberg GG: New developments in pancreatic cancer. Semin Gastrointest Dis 11: 162-167, 2000 Grapin-Botton A, Majithia AR, Melton DA: Key events of pancreas formation are triggered in gut endoderm by ectopic expression of pancreatic regulatory genes. Gen Dev 15: 444- 454, 2001 Greenway B, Iqbal MJ, Johnson PJ, et al: Oestrogen receptor proteins in malignant and fetal pancreas. Br Med J 283: 751- 753, 1981 Gudjonsson B: Survival statistics gone awry: pancreatic cancer, a case in point. J Clin Gastroenterol 35: 180-184, 2002 Guillermet J, Saint-Lauren N, Rochaix P, et al: Somatostatin receptor subtype 2 sensitizes human pancreatic cancer cells to death ligand-induced apoptosis. Proc Natl Acad Sci USA 100, 155-160, 2003 Gullo L: Risk of pancreatic and periampullary cancer following cholecystectomy. Ann Oncol 10:, Suppl 4), 127-128, 1999 Haddock G, Harrison DJ, Carter DC: The effect of the somatostatin analogue SMS 201-995 on experimental pancreatic carcinogenesis in the Syrian golden hamster. Carcinogenesis 12: 1103-1107, 1991 Hahn SA, Greenhalf B, Ellis I, et al: BRCA2 germline mutations in familial pancreatic carcinoma. J Natl Cancer Inst 95: 214-221, 2003 Hall PA, Lemoine NR: Rapid acinar to ductal transdifferentiation in cultured human exocrine pancreas. J Pathol 166: 97- 103, 1992 Harnack LJ, Anderson KE, Zheng W, et al: Smoking, alcohol, coffee, and tea intake and incidence of cancer of the exocrine pancreas: the Iowa Women’s Health Study. Cancer Epidemiol Biomarkers Prev 6: 1081-1086, 1997 Hecht SS, Hoffmann D: N-nitroso compounds and tobaccoinduced cancers in man. IARC Sci Publ 105:54-61, 1991 Hemminki K, Li X: Familial and second primary pancreatic cancers: a nationwide epidemiologic study from Sweden. Int J Cancer 103: 525-530, 2003 Howatson AG, Carter DC: Pancreatic carcinogenesis – enhancement by cholecystokinin in the hamster-nitrosamine model. Br J Cancer 51: 107-114, 1985 Howatson AG, Carter DC: Pancreatic carcinogenesis: effect of secretin in the hamster-nitrosamine model. J Natl Cancer Inst 78: 101-105, 1987 Hruban RH, Adsay NV, Albores-Saavedra J, Compton C, et al: Pancreatic intraepithelial neoplasia. A new nomeclature and classification system for pancreatic duct lesions. Am J Surg Pathol 25: 579-586, 2001 Hruban RH, Canto MI, Yeo CJ: Prevention of pancreatic cancer and strategies for management of familial pancreatic cancer. Dig Dis 19: 76-84, 2001 Huynh H, Pollak M: Enhancement of tamoxifen-induced suppression of insulin-like growth factor 1 gene expression and serum level by a somatostatin analogue. Biochem Biophys Res Comm 203: 253-259, 1994 Imaizumi Y: Longitudinal Gompertzian analysis of mortality from pancreatic cancer in Japan, 1955-1993. Mech Ageing Dev 90: 163-181, 1996 Janes RH, Niederhuber JE, Chmiel JS, et al: National patterns of care for pancreatic cancer. Results of a survey by the Commission on Cancer. Ann Surg 223: 261-272, 1996 Jemal A, Murray T, Samuels A,, et al: Cancer statistics, 2003. CA Cancer J Clin 53: 5-26, 2003 Ji BT, Chow WH, Hsing AW, et al: Green tea consumption and the risk of pancreatic and colorectal cancers. Int J Cancer 27: 255-258, 1997 Kamisawa T, Tu Y, Egawa N, et al: Ductal and acinar differentiation in pancreatic endocrine tumors. Dig Dis Sci 47: 2254- 2261, 2002 Keri G, Erchegyi A, Horvath A, et al: A tumor-selective somatostatin analog, TT-232, with strong in vitro and in vivo antitumor activity. Proc Natl Acad Sci USA 93: 12513-12518, 1996 Kernan GJ, Ji BT, Dosemeci M, et al: Occupational risk factors for pancreatic cancer: a case-control study based on death certificates from 24 U.S. states. Am J Ind Med 36: 260-270, 1999 Kikutsuji T, Harada M, Tashiro S, et al: Expression of somatostatin receptor subtypes and growth inhibition in human exocrine pancreatic cancer. J Hepatobiliary Pancreat Surg 7: 496-503, 2000 Klein AP, Beaty TH, Bailey-Wilson JE, et al: Evidence for a major gene influencing risk of pancreatic cancer. Genet Epidemiol 23: 133-149, 2002 Klein AP, Hruban RH, Brune KA, et al: Familial pancreatic cancer. Cancer J 7: 266-273, 2001 Klein WM, Hruban RH, Klein-Szanto AJ, et al: Direct correlation between proliferative activity and dysplasia in pancreatic intraepithelial neoplasia, PanIN): additional evidence for a recently proposed model of progression. Modern Pathol 15: 441-447, 2002 Klimstra DS, Longnecker DS: K-ras mutations in pancreatic ductal proliferative lesions. Am J Pathol 145: 1547-1548, 1994 Kokawa A, Kondo H, Gotoda T, et al: Increased expression of cyclooxygenase-2 in human pancreatic neoplasms and potential for chemoprevention by cyclooxygenase inhibitors. Cancer 91: 333-338, 2001 Lee JU, Hosotani R, Wada M, et al: Antiproliferative activity induced by the somatostatin analogue, TT-232, in human pancreatic cancer cells. Eur J Cancer 38: 1526-1534, 2002 Levi F, Decarli A, La Vecchia C: Trends in cancer mortality in Switzerland, 1951-1984. Rev Epidemiol Sante Publique 36: 15-25, 1988 Lillemoe KD, Yeo CJ, Cameron JL: Pancreatic cancer: stateof- the-art care. CA Cancer J Clin 50: 241-268, 2000 Lin Y, Tamakoshi A, Kawamura T, Inaba Y, et al: Risk of pancreatic cancer in relation to alcohol drinking, coffee consumption and medical history: findings from the Japan collaborative cohort study for evaluation of cancer risk. Int J Cancer 99: 742-746, 2002 Lippman SM, Lee JJ, Sabichi AL: Cancer chemoprevention: progress and promise. J Natl Cancer Inst 90: 1514-1528, 1998 Lopez F, Esteve JP, Buscail L, et al: Molecular mechanisms of antiproliferative effect of somatostatin: involvement of a tyrosine phosphatase. Metabolism, 45:, Suppl. 1., 14-16, 1996 Luttges J, Diederichs A, Menke MA, et al: Ductal lesions in patients with chronic pancreatitis show K-ras mutations in a frequency similar to that in the normal pancreas and lack nuclear immunoreactivity for p53. Cancer 88: 2495-2504, 2000 Lyn-Cook BD, Rogers T, Yan Y, et al: Chemopreventive effects of tea extracts and various components on human pancreatic and prostate tumor cells in vitro. Nutr Cancer 35: 80-86, 1999 Maitra A, Ashfaq R, Gunn CR, et al: Cyclooxygenase 2 expression in pancreatic adenocarcinoma and pancreatic intraepithelial neoplasia: an immunohistochemical analysis with automated cellular imaging. Am J Clin Pathol 118: 194-201, 2002 Majima T, Tsutsumi M, Nishino H, et al: Inhibitory effects of beta-carotene, palm carotene, and green tea polyphenols on pancreatic carcinogenesis initiated by N-nitrosobis(2-oxopropyl, amine in Syrian golden hamsters. Pancreas 16: 13-18, 1998 Manes G, Balzano A, Vaira D: Helicobacter pylori and pancreatic disease. J Pancreas, Online, 4: 111-116, 2003 Meijers M, Woutersen RA, van Garderen-Hoetmer A, et al: Effects of sandostatin and castration on pancreatic carcinogenesis in rats and hamsters. Int J Cancer 50: 246-251, 1992 Menezes RJ, Huber KR, Mahoney MC, et al: Regular use of aspirin and pancreatic cancer risk. BMC Public Health 2: 1- 18, 2002 Michaud DS, Giovannucci E, Willett WC, et al: Coffee and alcohol consumption and the risk of pancreatic cancer in two prospective United States cohorts. Cancer Epidemiol Biomarkers Prev 10: 429-437, 2001 Molina MA, Sitja-Arnau M, Lemoine MG, et al: Increased cyclooxygenase-2 expression in human pancreatic carcinomas and cell lines: growth inhibition by nonsteroidal anti-inflammatory drugs. Cancer Res 59: 4356-4362, 1999 Morimoto H, Nio Y, Tsubono M, et al: Inhibitory effects of cholecystokinin antagonist, loxiglumide, CR-1505), on the growth of freshly separated and xenografted human pancreatic cancer. J Surg Oncol 53: 47-53, 1993 Mouria M, Gukovskaya AS, Jung Y, et al: Food-derived polyphenols inhibit pancreatic cancer growth through mitochondrial cytochrome C release and apoptosis. Int J Cancer 98: 761-769, 2002 Mulligan NJ, Yang S, Andry C, et al: The role of p21ras in pancreatic neoplasia and chronic pancreatitis. Hum Pathol 30: 602-610, 1999 Nilsson HO, Stenram U, Ihse I, et al: Re: Helicobacter pylori seropositivity as a risk factor for pancreatic cancer. J Natl Cancer Inst 94: 632-633, 2002 Okada Y, Mori H, Tsutsumi A: Duct-acinar-islet cell tumor of the pancreas. Pathol Int 45: 669-676, 1995 Orth M, Gansauge F, Gansauge S, et al: K-ras mutations at codon 12 are rare events in chronic pancreatitis. Digestion 59: 120-124, 1998 Parkash O: Carcinoma of pancreas. A study based on autopsy data from 1928 to 1972 in Vienna, Austria. Digestion 13: 137-144, 1975 Patel PC, Barrie R, Hill N, et al: Postreceptor signal transduction mechanisms involved in octreotide-induced inhibition of angiogenesis. Surgery 116: 1148-1152, 1994 Pettersson F, Dalgleish AG, Bissonnette RP, et al: Retinoids cause apoptosis in pancreatic cancer cells via activation of RAR-gamma and altered expression of Bcl2/Nax. Br J Cancer 87: 555-561, 2002 Pilichowska M, Kimura N, Schindler M, et al: Somatostatin type 2A receptor immunoreactivity in human pancreatic adenocarcinomas. Endocrine Pathol 12: 147-155, 2001 Pleðko I, Dimitrova E, Somogyi J, et al: Geographic patterns and trends of liver, biliary tract and pancreas cancer mortality in Slovakia. Neoplasma 30: 97-104, 1983 Pollak MN, Schally AV: Mechanisms of antineoplastic action of somatostatin analogs. Proc Soc Exp Biol Med 217: 143- 152, 1998 Popiela T, Kedra B, Karcz D, et al: Long-term results of pancreatic cancer surgery. Przegl Lek 57:, Suppl. 5, 22-26, 2000 Poston GJ, Townsend CM jr, Rajaraman S, et al: Effect of somatostatin and tamoxifen on the growth of human pancreatic cancer in nude mice. Pancreas 5: 151-157, 1990 Pour PM, Schmied BM, Ulrich AB, et al: Abnormal differentiation of islet cells in pancreatic cancer. Pancreatology 1: 110-116, 2001 Prokopczyk B, Hoffmann D, Bologna M, et al: Identification of tobacco-derived compounds in human pancreatic juice. Chem Res Toxicol 15: 677-85, 2002 Qin Y, Ertl T, Groot K, et al: Somatostatin analog RC-160 inhibits growth of CFPAC-1 human pancreatic cancer cells in vitro and intracellular production of cyclic adenosine monophosphate. Int J Cancer 60: 694-700, 1995 Queneau PE, Adessi GL, Thibault P, et al: Early detection of pancreatic cancer in patients with chronic pancreatitis: diagnostic utility of a K-ras point mutation in the pancreatic juice. Am J Gastroenterol 96: 700-704, 2001 Raderer M, Wrba F, Kornek G, et al: Association between Helicobacter pylori infection and pancreatic cancer. Oncology 55: 16-19, 1998 Radulovic S, Comaru-Schally AM, Milovanovic S, et al: Somatostatin analogue RC-160 and LH-RH antagonist SB-75 inhibit growth of MIA PaCa-2 human pancreatic cancer xenografts in nude mice. Pancreas 8: 88-97, 1993 Rautalahti MT, Virtamo JR, Taylor PR, et al: The effects of supplementation with alpha-tocopherol and beta-carotene on the incidence and mortality of carcinoma of the pancreas in a randomized, controlled trial. Cancer 86: 37-42, 1999 Recchia F, Sica G, Casucci D, et al: Advanced carcinoma of the pancreas: phase II study of combined chemotherapy, beta-interferon, and retinoids. Am J Clin Oncol 21: 275-278, 1998 Reubi JC: Relevance of somatostatin receptors and other peptide receptors in pathology. Endocrine Pathol 8: 11-20, 1997 Reubi JC, Horisberger U, Essed EC, et al: Absence of somatostatin receptors in human exocrine pancreatic adenocarcinomas. Gastroenterology 95: 760-763, 1988 Reubi JC, Kappeler A, Waser B, et al: Immunohistochemical localization of somatostatin receptors sst2A in human tumors. Am J Pathol 153: 233-245, 1998 Riecken EO, Rosewicz S: Retinoids in pancreatic cancer. Ann Oncol 10: Suppl. 4, 197-200, 1999 Robertson JF, Watson SA, Hardcastle JD: Effect of gastrointestinal hormones and synthetic analogues on the growth of pancreatic cancer. Int J Cancer 63: 69-75, 1995 Robinson EK, Grau AM, Evans DB, et al: Cell cycle regulation of human pancreatic cancer by tamoxifen. Ann Surg Oncol 5: 342-349, 1998 Rohan T, McMichael AZ: Alimentary tract cancer mortality in Australia, 1908-1978. An epidemiological appraisal. Med J Aust 1: 232-235, 1981 Rooman L, Heremans Y, Heumberg H, et al: Modulation of rat pancreatic acinoductal transdifferentiation and expression of PDX-1 in vitro. Diabetologia 43: 907-914, 2000 Rosenberg, L, Barkun AN, Denis MH, et al: Low dose octreotide and tamoxifen in the treatment of adenocarcinoma of the pancreas. Cancer 75: 23-28, 1995 Rulyak SJ, Lowenfels AB, Maisonneuve P, et al: Risk factors for the development of pancreatic cancer in familial pancreatic cancer kindreds. Gastroenterology 124: 1292-1299, 2003 Schlosser W, Schoenberg MH, Rhein E, et al: Pankreaskarzinom bei chronischer Pankreatitis mit entzundlichem Pankreaskopftumor. Z Gastroenterol 34: 3-8, 1996 Schmied BM, Ulrich A, Matsuzaki H, et al: Transdifferentiation of human islet cells in a long-term culture. Pancreas 23: 157-171, 2001 Selenskas S, Teta MJ, Vitale JN: Pancreatic cancer among workers processing synthetic resins. Am J Ind Med 28: 385- 98, 1995 Singh P, Townsend CM jr, Poston GJ, et al: Specific binding of cholecystokinin. estradiol and somatostatin to human pancreatic cancer xenografts. J Steroid Biochem Mol Biol 39: 759-767, 1991 Smith JP, Verderame MF, Zagon IS: Antisense oligonucleotides to gastrin inhibit growth of human pancreatic cancer. Cancer Lett 135: 107-112, 1999 Stolzenberg-Solomon RZ, Blaser MJ, Limburg PJ, et al: Helicobacter pylori seropositivity as a risk factor for pancreatic cancer. J Natl Cancer Inst 93: 937-941, 2001 Su GH, Hruban RH, Bansal RK, Bova GS, et al: Germline and somatic mutations of the STK11/LKB1 Peutz-Jeghers gene in pancreatic and biliary cancers. Am J Pathol 154: 1835-1840, 1999 Szende B, Srkalovic G, Schally AV, et al: Inhibitory effects of analogs of luteinizing hormone-releasing hormone and somatostatin on pancreatic cancers in hamsters. Events that accompany regression. Cancer 65: 2279-2290, 1990 Szende B, Zalatnai A, Schally AV: Programmed cell death, apoptosis, in pancreatic cancers of hamsters after treatment with analogs of both luteinizing hormone-releasing hormone and somatostatin. Proc Natl Acad Sci USA 86: 1643-1647, 1989 Szepeshazi K, Lapis K, Schally, AV: Effect of combination treatment with analogs of luteinizing hormone-releasing hormone, LH-RH, or somatostatin and 5-fluorouracil on pancreatic cancer in hamsters, Int J Cancer 49: 260-266, 1991 Tabata T, Fujimori T, Maeda S, et al: The role of Ras mutation in pancreatic cancer, precancerous lesions, and chronic pancreatitis. Int J Pancreatol, 14: 237-244, 1993 Takeda Y, Escribano MJ: Effects of insulin and somatostatin on the growth and the colony formation of two human pancreatic cancer cell lines. J Cancer Res Clin Oncol 117: 416-20, 1991 Tang C, Biemond I, Verspaget HW, Offerhaus GJ, et al: Expression of somatostatin receptors in human pancreatic tumor. Pancreas 17: 80-84, 1998 Tezel E, Nagasaka T, Tezel G, et al: Characterization of scattered neuroendocrine cells in ductal carcinoma of the pancreas. Pancreas 25: 136-141, 2002 Tomao S, Romiti A, Massidda B, et al: A phase II study of gemcitabine and tamoxifen in advanced pancreatic cancer. Anticancer Res 22: 2361-2364, 2002 Upp JR, Olson D, Poston GJ, et al: Inhibition of growth of two human pancreatic adenocarcinomas in vivo by somatostatin analog SMS 201-995. Am J Surg 155: 29-35, 1988 Van Laethem JL, Bourgeois V, Parma J, et al: Relative contribution of Ki-ras gene analysis and brush cytology during ERCP for the diagnosis of biliary and pancreatic diseases. Gastrointest Endosc 47: 479-485, 1998 Vernejoul F, Faure P, Benali N, et al: Antitumor effect of in vivo somatostatin receptor subtype 2 gene transfer in primary and metastatic pancreatic cancer models. Cancer Res 62: 6124-6131, 2002 Wakasugi H, Funakoshi A, Iguchi H, et al: Pancreatic carcinoma associated with chronic pancreatitis. Intern Med 38: 951- 956, 1999 Wang M, Abbruzzese JL, Friess H, et al: DNA adducts in human pancreatic tissues and their potential role in carcinogenesis. Cancer Res 58: 38-41, 1998 Wang R, Li J, Rosenberg L: Factors mediating transdifferentiation of islets of Langerhans to duct epithelial-like structures. J Endocrinol 171: 309-318, 2001 Weiderpass E, Vainio H, Kauppinen T, et al: Occupational exposures and gastrointestinal cancers among Finnish women. J Occup Environ Med 45: 305-15, 2003 Welch HG, Schwartz LM, Woloshin S: Are increasing 5-year survival rates evidence of success against cancer? JAMA 283: 2975-2978, 2000 Wenger FA, Jacobi CA, Siderow A, et al: Hormontherapie des postoperative rezidivierten Pankreaskarzinoms mit Octreotid und Tamoxifen. Chirurg 70: 694-699, 1999 Wenger FA, Kilian M, Mautsch I, et al: Influence of octreotide and tamoxifen in tumor growth and liver metastasis in Nnitrosobis( 2-oxopropyl)amine induced pancreatic cancer in Syrian hamsters. Horm Res 54: 74-77, 2000 Woutersen RA, Appel MJ, Van Garderen-Hoetmer A: Modulation of pancreatic carcinogenesis by antioxidants. Food Chem Toxicol 37: 981-984, 1999 Yao GY, Zhou JL, Lai MD, et al: Neuroendocrine markers in adenocarcinomas: an investigation of 356 cases. World J Gastroenterol 9: 858-861, 2003 Yip-Schneider MT, Sweeney CJ, Jung SH, et al: Cell cycle effects of nonsteroidal anti-inflammatory drugs and enhanced growth inhibition in combination with gemcitabine in pancreatic carcinoma cells. J Pharmacol Exp Ther 298: 976-985, 2001 Zalatnai A, Pogany V: Apoptosis-induction and phosphorylation state in human pancreatic carcinoma xenografts following octreotide treatment. Anticancer Res 21: 477-480, 2001 Zalatnai A, Schally AV: Responsiveness of the hamster pancreatic cancer to the treatment with microcapsules of D-Trp-6- LH-RH and somatostatin analog RC-160: histological evidence of improvement. Int J Pancreatol 4: 149-160, 1989 Zalatnai A, Schally AV: Treatment of the N-Nitrosobis(2-oxopropyl, amine-induced pancreatic cancer in Syrian golden hamsters with D-Trp-6-LH-RH and somatostatin analog RC- 160 microcapsules. Cancer Res 49: 1810-1815, 1989 Zalatnai A, Szegedi Z, Bocsi J: Flow cytometric evidence of apoptosis in human pancreatic cancer xenografts treated with Sandostatin, octreotide). Anticancer Res 20: 1663-1666, 2000 Zalatnai A, Timar F: In vitro antiangiogenic effect of Sandostatin, octreotide, on the proliferation of the placental vessels. Anticancer Res 22: 4225-4228, 2002 Zugmaier G, Jager R, Grage B, Gottardis MM, Havemann K et al: Growth-inhibitory effects of vitamin D analogues and retinoids on human pancreatic cancer cells. Br J Cancer 73: 1341-1346, 1996 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2003 VL 9 IS 4 BP 252 EP 263 PG 12 ER PT J AU Szabo, E Paska, Cs Kaposi Novak, P Schaff, Zs Kiss, A AF Szabo, Erzsebet Paska, Csilla Kaposi Novak, Pal Schaff, Zsuzsa Kiss, Andras TI Similarities and Differences in Hepatitis B and C Virus Induced Hepatocarcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE hepatitis B virus; hepatitis C virus; hepatocellular carcinoma; hepatocarcinogenesis ID hepatitis B virus; hepatitis C virus; hepatocellular carcinoma; hepatocarcinogenesis AB Hepatocellular carcinoma (HCC), the major manifestation of primary liver cancer, is one of the most frequent and malignant diseases worldwide. Among other environmental factors, hepatitis viruses, as the hepatitis B (HBV) and hepatitis C (HCV) viruses, are to be listed in the etiology of HCC. Both of these viruses cause a wide spectrum of clinical manifestations, ranging from healthy carrier state to acute and chronic hepatitis, cirrhosis and HCC. HBV and HCV are different viruses in structure: HBV contains a DNA genome which replicates through an RNA intermediate and requires an active viral reverse transcriptase (RT) polymerase enzyme, while HCV is an RNA virus which has no RT activity and replicates on the cellular membrane by RNA replication. In this review we discuss how these two biologically diverse viruses use common pathways to induce hepatocarcinogenesis despite their significant structural and viral cycle differences. A summary is also given of several observable common and different features. Direct integration of HBV viral sequences into the host genome increases the genomic instability, which does not occur in HCV infection. However, viral proteins may directly play a significant role in the induction of carcinogenesis by both viruses. C1 [Szabo, Erzsebet] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93., H-1091 Budapest, Hungary. [Paska, Csilla] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93., H-1091 Budapest, Hungary. [Kaposi Novak, Pal] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93., H-1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93., H-1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93., H-1091 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM schaff@korb2.sote.hu CR Agnello V, Abel G, Elfahal M et al: Hepatitis C virus and other flaviviridae viruses enter cells via low density lipoprotein receptor. Proc Natl Acad Sci USA 96: 12766-12771, 1999 Aizawa Y, Shibamoto Y, Takagi I et al: Analysis of factors affecting the appearance of hepatocellular carcinoma in patients with chronic hepatitis C. Cancer 89: 53-59, 2000 Andrisani OM, Barnabas S: The transcriptional function of the hepatitis B virus X protein and its role in hepatocarcinogenesis. Int J Oncol 15: 373-379, 1999 Anthony PP: Hepatocellular carcinoma: an overview. Histopathology 39: 109-118, 2001 Bosch FX: Global epidemiology of hepatocellular carcinoma. In: Liver Cancer., Eds: Okuda K, Tabor E), Churchill Livingstone, New York, 1997, pp. 13-28 Brechot C, Gozuacik D, Murakami Y et al: Molecular bases for the development of hepatitis B virus, HBV)-related hepatocellular carcinoma, HCC). Semin Cancer Biol 10: 211-231, 2000 Buendia MA: Genetics of hepatocellular carcinoma. Semin Cancer Biol 10: 185-200, 2000 Chen P-J, Chen D-S: Hepatitis B virus infection and hepatocellular carcinoma: Molecular genetics and clinical perspectives. Semin Liver Dis 19: 253-262, 1999 Cohen J: The scientific challenge of hepatitis C. Science 285: 26-30, 1999 Conjeevaram HS, Lok AS: Management of chronic hepatitis B. J Hepatol 38: S90-S103, 2003 Edamoto Y, Hara A, Biernat W et al: Alterations of RB1, p53 and Wnt pathways in hepatocellular carcinomas associated with hepatitis C, hepatitis B and alcoholic liver cirrhosis. Int J Cancer 106: 334-341, 2003 El-Serag HB, Mason AC, Key C: Trends in survival of patients with hepatocellular carcinoma between 1977 and 1996 in the United States. Hepatology 3: 62-65, 2001 Faivre J, Forman D, Esteve J et al: Survival of patients with primary liver cancer, pancreatic cancer and biliary tract cancer in Europe. EUROCARE Working Group. Eur J Cancer 34: 2184-2190, 1998 Feitelson MA: Hepatitis B virus in hepatocarcinogenesis. J Cell Physiol 181: 188- 202, 1999 Feo F, Pascale RM, Simile MM et al: Genetic alterations in liver carcinogenesis: implications for new preventive and therapeutic strategies. Crit Rev Oncogen 11: 19-62, 2000 Ferber MJ, Montoya DP, Yu C et al: Integrations of the hepatitis B virus, HBV, and human papillomavirus, HPV, into the human telomerase reverse transcriptase, h TERT, gene in liver and cervical cancers. Oncogene 22: 3813-3820, 2003 Ferlay J, Bray F, Pisani P et al: GLOBOCAN 2000: Cancer incidence, mortality and prevalence worldwide, Version 1.0., IARC CancerBase No. 5, IARC Press, Lyon, 2001 Gelatti U, Donato F, Tagger A et al: Etiology of hepatocellular carcinoma influences clinical and pathologic features but not patient survival. Am J Gastroenterol 98: 907-914, 2003 Giannelli G, Fransvea E, Marinosci F et al: Transforming growth factor-1 triggers hepatocellular carcinoma invasiveness via 31 integrin. Am J Pathol 161: 183-193, 2002 Idilman R, DeMaria N, Colantoni A et al: Pathogenesis of hepatitis B and C-induced hepatocellular carcinoma. J Viral Hepat 5: 285-299, 1998 Iizuka N, Oka M, Yamada-Okabe H et al: Differential gene expression in distinct virologic types of hepatocellular carcinoma: association with liver cirrhosis. Oncogene 22: 3007-3014, 2003 Laurent-Puig P, Legoix P, Bluteau O et al: Genetic alterations associated with hepatocellular carcinomas define distinct pathways of hepatocarcinogenesis. Gastroenterology 120: 1763- 1773, 2001 Moradpour D, Wands JR: Molecular pathogenesis of hepatocellular carcinoma. In: Hepatology. A Textbook of Liver Disease, ed 4., Eds: Zakim D, Boyer TD), Saunders, Philadelphia, 2003, pp. 1333-1354 Nair S, Perrillo RP:Hepatitis B and D. In: Hepatology. A Textbook of Liver Disease, ed 4., Eds: Zakim D, Boyer TD), Saunders, Philadelphia, 2003, pp. 959-1016 Nakamoto Y, Kanekos S: Mechanisms of viral hepatitis induced liver injury. Curr Mol Med 3: 537-544, 2003 Ochiai T, Urata Y, Yamano T et al: Clonal expansion in evolution of chronic hepatitis to hepatocellular carcinoma as seen at an X-chromosome locus. Hepatology 31: 615-621, 2000 Okuda K: Hepatocellular carcinoma. J Hepatol 32, Suppl 1): 225-237, 2000 Ozturk M: Genetic aspects of hepatocellular carcinogenesis. Semin Liver Dis 19: 235-242, 1999 Paradis V, Bieche I, Dargere D et al: Molecular profiling of hepatocellular carcinomas, HCC, using a large-scale real-time RT-PCR approach. Am J Pathol 163: 733-741, 2003 Parkin DM, Bray FI, Devesa SS: Cancer burden in the year 2000. The global picture. Eur J Cancer 37: S4-S66, 2001 Pileri P, Uematsu Y, Campagnoli S et al: Binding of hepatitis C virus to CD81. Science 282: 938-941, 1998 Pina Dore M, Realdi G, Mura D et al: Genomic instability in chronic viral hepatitis and hepatocellular carcinoma. Hum Pathol 32: 698-703, 2001 Pineau P and Buendia MA: Studies of genetic defects in hepatocellular carcinoma: recent outcomes and new challenges. J Hepatol 33: 152-156, 2000 Poussin K, Dienes H, Sirma H et al: Expression of mutated hepatitis B virus X genes in human hepatocellular carcinomas. Int J Cancer 80: 497-505, 1999 Qui W, David D, Zhou B et al: Down-regulation of growth arrest DNA damage-inducible gene 45 expression is associated with human hepatocellular carcinoma. Am J Pathol 162: 1961- 1974, 2003 Rosen HR, Gretch DR: Hepatitis C virus: current understanding and prospects for future therapies. Mol Med Today 5: 393- 399, 1999 Saunier B, Triyatni M, Ulianich L et al: Role of the asialoglycoprotein receptor in binding and entry of hepatitis C virus structural proteins in cultured human hepatocytes.J Virol 77: 546-559, 2003 Scarselli E, Ansuini H, Cerino R et al: The human scavenger receptor class B type I is a novel candidate receptor for the hepatitis C virus. EMBO J 21: 5017-5025, 2002 Schulze zur Wiesch J, Schmitz H, Borowski E, Borowski P: The proteins of the hepatitis C virus: Their features and interactions with intracellular protein phosphorylation. Arch Virol 148: 1247-1267, 2003 Szabo E, Lotz G, Paska C et al: Viral hepatitis: New data on hepatitis C. Pathol Oncol Res 9: 215-221, 2003 Tabor E: Interferon for preventing and treating hepatocellular carcinoma associated with the hepatitis B and C viruses. Dig Liver Dis 35: 297-305, 2003 Thorgeirsson SS, Grisham JW: Molecular pathogenesis of human hepatocellular carcinoma. Nat Genet 31: 339-346,2002 Torbenson M, Marinopoulos S, Dang DT et al: Smad4 overexpression in hepatocellular carcinoma is strongly associated with transforming growth factor beta II receptor immunolabeling. Hum Pathol 33: 871-876, 2002 Tornillo L, Carafa V, Richter J et al: Marked genetic similarities between hepatitis B virus-positive and hepatitis C viruspositive hepatocellular carcinomas. J Pathol 192: 307-312, 2000 van Regenmortel MHV, Fauquet CM, Bishop DHL et al: Virus Taxonomy. The VIIth Report of the International Committee on Taxonomy of Viruses. Academic Press, San Diego, 2000 Wagayama, H, Shiraki K, Sugimoto K et al: High expression of p21WAF1/CIP1 is correlated with human hepatocellular carcinoma in patients with hepatitis C virus-associated chronic liver diseases. Hum Pathol 33: 429-434, 2002 Waris G, Siddiqui A: Regulatory mechanisms of viral hepatitis B and C. J Biosci 28: 311-321, 2003 Xu X-R, Huang J, Xu Z-G et al: Insight into hepatocellular carcinogenesis at transcriptome level by comparing gene expression profiles of hepatocellular carcinoma with those of corresponding noncancerous liver. Proc Natl Acad Sci USA 98: 15089-15094, 2001 Ye Q-H, Qin L-X, Forgues M et al: Predicting hepatitis B viruspositive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learning. Nat Med 9: 416-423, 2003 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 5 EP 11 PG 7 ER PT J AU Mehes, K Kosztolanyi, Gy AF Mehes, Karoly Kosztolanyi, Gyorgy TI Clinical Manifestations of Genetic Instability Overlap One Another SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE genetic instability; cancer-prone diseases; mild errors of morphogenesis; premature centromere division; predisposition to cancer ID genetic instability; cancer-prone diseases; mild errors of morphogenesis; premature centromere division; predisposition to cancer AB Cancer syndromes are characteristic associations of specific malignancies with various congenital anomalies. In addition to such diseases, an increased prevalence in general of chromosomal instability, malformations, immunodeficiencies, altered growth and development, and reproductive loss has been observed in both childhood leukemias and solid tumors. The overlap among these congenital disorders suggests their common prenatal, possibly genetic origin and thus the existence of a nonspecific genetic instability leading to various clinical manifestations of disturbancies in cell division. Seeking for related features in family members of a patient with malignancy may be of clinical value in detecting predisposition to cancer. C1 [Mehes, Karoly] University of Pecs, Department of Medical Genetics and Child Development, Jozsef Attila u. 7, H-7623 Pecs, Hungary. [Kosztolanyi, Gyorgy] University of Pecs, Department of Medical Genetics and Child Development, Jozsef Attila u. 7, H-7623 Pecs, Hungary. RP Mehes, K (reprint author), University of Pecs, Department of Medical Genetics and Child Development, H-7623 Pecs, Hungary. EM karoly.mehes@aok.pte.hu CR Al-Nemri AR, Kilani RA, Salih MAM, et al: Embryonal rhabdomyosarcoma and chromosomal breakage in a newborn infant with possible Dubowitz syndrome. Am J Med Genet 92:107-110, 2000 Alter BP: Molecular medicine and bone marrow failure syndromes. J Pediatr 136:275-276, 2000 Altmann AE, Halliday JL, Giles GG: Associations between congenital malformations and childhood cancer. A registerbased case-control study. Br J Cancer 78:1244-1249, 1998 Anuradha N, Satyanarayana M, Manjunatha KR: Premature centromere division and abortions. Int J Hum Genet 1:191-193, 2001 Atkin NB: Microstallite instability. Cytogenet Cell Genet 92:177-181, 2002 Bajnoczky K, Gardo S: „Premature anaphase” in a couple with recurrent miscarriages. Hum Genet 92:388-390, 1993 Ben-Yosef T, Benvenisty N: Hereditary cancer and developmental abnormalities. Biol Neonate 77:1-11, 2000 Buhler EM, Fessler R, Beutler C, et al: Incidental finding of double minutes, DM), single minutes, SM), homogenously staining regions, HSR), premature chromosome condensation, PCC), and premature centromere division, PCD)? Ann Genet, Paris, 30:75-79, 1987 Carey JC: Invited editorial comment: Study of minor anomalies in childhood malignancy. Eur J Pediatr 144:250-251, 1985 Cillo C, Faiella A, Cantile M, et al: Homeobox genes and cancer. Exp Cell Res 248:1-9, 1999 Cloos J, Nieuwenhuis EJ, Boomsma DI et al.: Inherited susceptibility to bleomycin-induced chromatid breaks in cultured peripheral blood lymphocytes. J Natl Cancer Inst 91:1125- 1130, 1999 Cnattingius S., Zack MM, Ekbom A, et al.: Prenatal and neonatal risk factors for childhood lymphatic leukemia. J Natl Cancer Inst 87:908-914, 1995 Cope JU, Tsokos M, Helman LJ, et al.: Inguinal hernia in patients with Ewing sarcoma: A clue to etiology. Med Pediatr Oncol 34:195-199, 2000 Davies SM: More on congenital abnormalities and childhood cancer [comment]. Med Pediatr Oncol 32:49-50, 1999 Duesberg P, Li R:Multistep carcinogenesis: a chain reaction of aneuploidizations. Cell Cycle 2:202-210, 2003 Duker NJ: Chromosomal breakage syndromes and cancer. Am J Med Genet, Semin Med Genet, 115:125-129, 2002 Evans G, Burnell L, Campbell R et al.: Congenital anomalies and genetic syndromes in 173 cases of medulloblastoma. Med Pediatr Oncol 21:433-434, 1993 Fearon ER: Human cancer syndromes: clues to the origin and nature of cancer. Science 278:1043-1050, 1997 Fekete G, Rethy L, Batta I: Minor anomalies in children with leukaemia and malignant tumors [in Hungarian]. Orv Hetil 128:725-729, 1987 Fitzgerald PH: Premature centromere division and aneuploidy. In: Aneuploidy, Part A, Incidence and Etiology., Eds: Vig BK, Sandberg AA). Alan R Liss. New York, 1987, pp. 249-264. Ford HL: Homeobox genes: a link between development, cell cycle, and cancer? Cell Biol Int 22:397-400, 1998 Frias JL, Carey JC: Mild errors of morphogenesis. Adv Ped 43:27-75, 1996 Friedman JM: Genetics and epidemiology, congenital anomalies and cancer [invited editorial] Am J Hum Genet 60:469- 473, 1997 Ghali MH, Yoo K-Y, Flannery JT, et al: Association between childhood rhabdomyosarcoma and maternal history of stillbirths. Int J Cancer 50:365-368, 1992 Gisselsson D: Chromosome instability in cancer: how, when, and why? Adv Cancer Res 87:1-29, 2003 Hartley AL, Birch JM, Blair V, et al: Malformations in children with soft tissue sarcoma and in their parents and siblings. Ped Perinat Epidemiol 8:423-432, 1994 Hartley AL, Birch JM, Blair V, et al.: Foetal loss and infant deaths in families of children with soft-tissue sarcoma. Int J Cancer 56:646-649, 1994 Hisama FM, Weissman SM, Martin GM, Eds): Chromosomal instability and aging. Marcel Dekker, Inc., New York, Basel, 2003 Jakab Z, Szegedi I, Balogh E, et al.: Duchenne muscular dystrophy/ rhabdomyosarcoma, ichthyosis vulgaris/acute monoblastic leukemia: Association of rare genetic disorders and childhood malignant diseases. Med Pediatr Oncol 39:66-68, 2002 Jones SM, Phillips PC, Molloy PT, et al.: Congenital anomalies and genetic disorders in families of children with central nervous system tumours. J Med Genet 32:627-632, 1995 Kajii T, Ikeuchi T, Yang Z-Q, et al.: Cancer-prone syndrome of mosaic variegated aneuploidy and total premature chromatid separation: Report of five infants. Am J Med Genet 104:57-64, 2001 Kajtar P, Weisenbach J, Mehes K: Association of Wilms tumour with spina bifida occulta. Eur J Pediatr 149:594-595, 1990 Kajtar P, Mehes K: Bilateral Coats retinopathy associated with aplastic anaemia and mild dyskeratotic signs. Am J Med Genet 49:374-377, 1994 Kaye SA, Robison LL, Smithson WA, et al.: Maternal reproductive history and birth characteristics in childhood acute lymphoblastic leukemia. Cancer 68:1351-1355, 1991 Kirchner GI, Wagner S, Flemming P, et al.: COACH syndrome associated with multifocal liver tumors. Am J Gastroenterol 97:2664-2669, 2002 Kobayashi N, Furukawa T, Takatsu T: Congenital anomalies in children with malignancy. Pediat Univ Tokyo 16:31-37, 1968 Kosztolanyi G: Does a „ring syndrome” exist? An analysis of 207 case reports on patients with a ring autosome. Hum Genet 75:174-179, 1987 Lengauer C, Kinzler KW, Vogelstein B: Genetic instabilities in human cancers. Nature 398:643-649, 1998 Madan K, Lindhout D, Palan A: Premature centromere division, PCD): a dominantly inherited cytogenetic anomaly. Hum Genet 77:193-196, 1987 Major J, Jakab MG, Tompa A: The frequency of induced premature centromere division in human populations occupationally exposed to genotoxic chemicals. Mutat Res, Genet Toxicol Environm Mutagenesis, 445:241-249, 1999 Martin GM, Oshima J: Lessons from human progeroid syndromes. Nature 408:263-266, 2000 Makitie O, Pukkala E, Teppo L, et al: Increased incidence of cancer in patients with cartilage-hair hypoplasia. J Pediatr 134:315-318, 1999 Mehes K: Non-random centromere division: A mechanism of non-disjunction causing aneuploidy? Hum Hered 28:255-260, 1978 Mehes K: Mild errors of morphogenesis in malignancy: Macroscopic manifestation of genetic instability? [editorial]. Med Pediatr Oncol 34:111-112, 2000 Mehes K, Signer E, Pluss HJ, et al.: Increased prevalence of minor anomalies in childhood malignancy. Eur J Pediatr 144:243-249, 1985 Mehes K, Szijjarto L, Kajtar P: Sex difference in the height of children with acute lymphoblastic leukemia. Acta Med Auxol 26:69-75, 1994 Mehes K, Gyurkovits K, Kosztolanyi G, et al: Fruhzeitige Zentromertrennung bei Kindern mit Immundefizienz [abstract]. Mschr Kinderheilk 141:S55, 1993 Mehes K, Buhler EM. Premature centromere division: a possible manifestation of chromosomal instability. Am J Med Genet 56:76-79, 1995 Mehes K, Kajtar P, Sandor G, et al.: Excess of mild errors of morphogenesis in childhood lymphoblastic leukemia. Am J Med Genet 75:22-27, 1998 Mehes K, Kajtar P, Kosztolanyi G: Association of nonsyndromic Wilms tumor with premature centromere division, PCD). Am J Med Genet 112:215-216, 2002 Mehes K, Weisenbach J, Kajtar P: Association of Wilms tumor with spinal dysraphism. Pediatr Hematol Oncol 20:261-264, 2003 Merlob P: Mild errors of morphogenesis: one of the most controversial subjects in dysmorphology. In Kalter H, Ed, Issues and Reviews in Teratology, Plenum, New York, 1994;7:57-102 Mertens AC, Wen W, Davies SM, et al.: Congenital abnormalities in children with acute leukemia: a report from the Children’s Cancer Group. J Pediatr 133:617-623, 1998 Miller RW: Down’s syndrome, mongolism), other congenital malformations and cancer among the sibs of leukemic children. New Engl J Med 268:393-401, 1963 Ming JE, Stiehm ER, Graham JM Jr.: Syndromes associated with immunodeficiency. Adv Pediatr 46:271-351, 1999 Murakami S, Terakado M, Misumi M, et al.: Situs inversus totalis with malignant lymphoma of the stomach: report of a case. Surg Today 33:533-536, 2003 Narod SA, Hawkins MM, Robertson CM, et al: Congenital anomalies and childhood cancer in Great Britain. Am J Hum Genet 60:474-485, 1997 Nishi M, Miyake H, Takeda T, et al: Congenital malformations and childhood cancer. Med Pediatr Oncol 34:250-254, 2000 Nunes FD, de Almeida FC, Tucci R, et al: Homeobox genes: link between development and cancer. Pesqui Odontol Brasil 17:94-98, 2003 Ondrus D, Kuba D, Chrenova S, et al:Familial testicular cancer and developmental anomalies. Neoplasma 44:59-61, 1997 Opitz JM:Invited editorial comment: study of minor anomalies in childhood malignancy. Eur J Pediatr 144:252-254, 1985 Opitz JM: Heterogeneity and minor anomalies. Am J Med Genet 91:254-255, 2000 Pakakasama S, Tomlinson GE: Genetic predisposition and screening in pediatric cancer. Pediatr Clin N Am 49:1393- 1413, 2002 Pavithran K, Kapoor G: Acute myeloid leukemia in a child with Goldenhar syndrome, Letter to the editor). Med Pediatr Oncol 38:451-452, 2002 Perrillat F, Clavel J, Jaussent I et al.: Breast-feeding, fetal loss and childhood acute leukaemia. Eur J Pediatr 161:235-237, 2002 Plaja A, Vendrell T, Smeets D, et al.: Variegated aneuploidy related to premature centromere division, PCD, in expressed in vivo and is a cancer-prone disease. Am J Med Genet 98:216- 223, 2001 Ramesh KH, Bhargava MK: Cytogenetic damage in peripheral blood lymphocytes of cancer patients prior to radiotherapy. Cancer Genet Cytogenet 60:86-88, 1992 Roganovic J, Radojcic-Badovinac A, Ahel V: Increased prevalence of minor anomalies in children with hematologic malignancies. Med Pediatr Oncol 38:128-130, 2002 Sandberg AA, Chen Z: Cytogenetics and molecular genetics of human cancer. Am J Med Genet, Semin Med Genet, 115:111- 112, 2002 Sandri A, Manazza AD, Bertin D, et al.: Schinzel-Giedion syndrome with sacrococcygeal teratoma. J Pediatr Hematol Oncol 25:558-561, 2003 Sen S: Aneuploidy and cancer. Curr Opin Oncol 12:82-88, 2000 Sieber OM, Heinimann K, Tomlinson IP: Opinion: Genomic instability – the engine of tumorigenesis? Nat Rev Cancer 3:701-708, 2003 Steinwachs EF, Amos C, Johnston D, et al.: Nonsyndromic cleft lip and palate is not associated with cancer or other birth defects. Am J Med Genet 90:17-24, 2000 Strauss A, Girschick G, Fuchshuber S, et al.: Prune-Belly-Syndrom und kongenitale Nierenturmoren. Z Geburtsh Neonatol 205:195199, 2001 Tollerud DJ, Blattner WA, Fraser MC, et al.: Familial testicular cancer and urogenital developmental anomalies. Cancer 55:1849-1854, 1985 Van Steensel-Moll HA, Valkenburg HA, Vanderbroucke JP, et al: Are maternal fertility problems related to childhood leukaemia? Int J Epidem 14:555-560, 1985 Vig BK, Paweletz N and Schroeter D: The centromere-kinetochore complex in cancer. Cancer J 6:243-252, 1993 Wertelecki W, Mantel SK, Grush DC: Increased birth weight in leukemia. Pediatr Res 7:132-138, 1973 Yeazel MW, Buckley JD, Woodes WG, et al.: History of maternal fetal loss and increased risk of childhood acute leukaemia at an early age: a report from the Children’s Cancer Group. Cancer 75:1718-172, 1995 Zhu JL, Basso O, Hasle H et al.: Do parents of children with congenital malformations have a higher cancer risk? A nationwide study in Denmark. Br J Cancer 87:524-528, 2002 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 12 EP 16 PG 5 ER PT J AU Ebinger, M Senf, L Wachowski, O Scheurlen, W AF Ebinger, Martin Senf, Leonore Wachowski, Olga Scheurlen, Wolfram TI Promoter Methylation Pattern of Caspase-8, P16INK4A, MGMT, TIMP-3, and E-Cadherin in Medulloblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE aberrant promoter methylation; medulloblastoma; caspase-8; p16INK4A; MGMT; TIMP-3; CDH1 ID aberrant promoter methylation; medulloblastoma; caspase-8; p16INK4A; MGMT; TIMP-3; CDH1 AB Methylation of promoter regions of CpG-rich sites is an important mechanism for silencing of tumor suppressor genes (TSG). To evaluate the role of tumor suppressor genes caspase-8 (CASP8), TIMP-3, E-cadherin (CDH1), p16INK4A, and MGMT in medulloblastoma tumorigenesis, 51 medulloblastomas (46 primary tumor specimens, 5 cell lines) were screened for methylation of promoter linked CpG-islands. For CASP8, we examined the 5' UTR region that has been shown to be associated with expression of CASP8. As detected by methylation specific PCR, methylation rate was low for TIMP-3 (3% of tumor samples; 1/5 cell lines), for MGMT (0% of tumor samples; 1/5 cell lines), for p16INK4A (2% of tumor samples; 2/5 cell lines) and for CDH1 (8% of tumor samples; 1/4 cell lines). CASP8, however, was methylated in 90% of tumor samples and 4/5 cell lines examined. Screening other tumor entities for CASP8 methylation, we found a similarly high level in 6 neuroblastoma cell lines in contrast to 5 osteosarcoma-, 4 Ewing's sarcoma- and 6 non-embryonic tumor cell lines without any increased promoter methylation. From our results we conclude that methylation of the CASP8 5' UTR region may play a role in inactivation of CASP8 in neural crest tumors. C1 [Ebinger, Martin] Institute of Pathology, Department of Molecular PathologyTubingen, Germany. [Senf, Leonore] Cnopf'sche Kinderklinik, St. Johannis-Muhlgasse 19, 90419 Nurnberg, Germany. [Wachowski, Olga] University Hospital Gießen, Zentrum fur Kinderheilkunde und JugendmedizinGiessen, Germany. [Scheurlen, Wolfram] Cnopf'sche Kinderklinik, St. Johannis-Muhlgasse 19, 90419 Nurnberg, Germany. RP Scheurlen, W (reprint author), Cnopf'sche Kinderklinik, 90419 Nurnberg, Germany. EM wolfram.scheurlen@nbg.diakonieneuendettelsau.de CR Bachman KE, Herman JG, Corn PG et al: Methylation-associated silencing of the tissue inhibitor of metalloproteinase-3 gene suggests a suppressor role in kidney, brain, and other human cancers. Cancer Res 59: 798-802, 1999. Banelli B, Casciano I, Croce M et al: Expression and methylation of CASP8 in neuroblastoma: identification of a promoter region. Nat Med 8: 1333-1335; author25 reply 1335, 2002. Barker FG, Chen P, Furman F et al: P16 deletion and mutation analysis in human brain tumors. J Neurooncol 31: 17-23, 1997. Berx G, Staes K, van Hengel J et al: Cloning and characterization of the human invasion suppressor gene E-cadherin, CDH1). Genomics 26: 281-289, 1995. Bobola MS, Berger MS, Ellenbogen RG et al: O6-methylguanine- DNA methyltransferase in pediatric primary brain tumors: relation to patient and tumor characteristics. Clin Cancer Res 7: 613-619, 2001. Bodmer JL, Holler N, Reynard S et al: TRAIL receptor-2 signals apoptosis through FADD and caspase-8. Nat Cell Biol 2: 241-243, 2000. Costello JF, Fruhwald MC, Smiraglia DJ et al: Aberrant CpGisland methylation has non-random and tumor-type-specific patterns. Nat Genet 25: 132-138, 2000 Esteller M, Sanchez-Cespedes M, Rosell R et al: Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res 59: 67-70, 1999. Esteller M, Garcia-Foncillas J, Andion E et al: Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med 343: 1350-1354, 2000. Friesen C, Herr I, Krammer PH, Debatin KM: Involvement of the CD95, APO-1/FAS, receptor/ligand system in druginduced apoptosis in leukemia cells. Nat Med 2: 574-577, 1996. Fruhwald MC, O’Dorisio MS, Dai Z et al: Aberrant promoter methylation of previously unidentified target genes is a common abnormality in medulloblastomas – implications for tumor biology and potential clinical utility. Oncogene 36: 5033-5042, 2001. Fulda S, Sieverts H, Friesen C et al: The CD95, APO-1/Fas, system mediates drug-induced apoptosis in neuroblastoma cells. Cancer Res 57: 3823-3829, 1997. Fulda S, Debatin KM: IFN sensitizes for apoptosis by upregulating caspase-8 expression through the STAT1 pathway. Oncogene 15: 2295-2308, 2002. Grenet J, Teitz T, Wei T et al: Structure and chromosome localization of the human CASP8 gene. Gene 226: 225–232, 1999. Grotzer MA, Eggert A, Zuzak TJ et al: Resistance to TRAILinduced apoptosis in primitive neuroectodermal brain tumor cells correlates with a loss of caspase-8 expression. Oncogene 19: 4604-4610, 2000. Harada K, Toyooka S, Shivapurkar N et al: Deregulation of caspase 8 and 10 expression in pediatric tumors and cell lines. Cancer Res 62: 5897-5901, 2002. Herman JG, Graff JR, Myohanen S et al: Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 93: 9821-9826, 1996. Jones PA, Baylin SB: The fundamental role of epigenetic events in cancer. Nat Rev Genet 3: 415-428, 2002. Katzenellenbogen RA, Baylin SB, Herman JG: Hypermethylation of the DAP-kinase CpG island is a common alteration in B-cell malignancies. Blood 93: 4347-4353, 1999. Mannello F, Gazzanelli G: Tissue inhibitors of metalloproteinases and programmed cell death: conundrums, controversies and potential implications. Apoptosis 6: 479-482, 2001. Miller SA, Dykes DD, Polesky HF: A simple salting out procedure for extracting DNA from human nucleated cells. Nucl Acid Res 16: 1215, 1988. Packer RJ: Childhood medulloblastoma: progress and future challenges. Brain Dev 21: 75-81, 1999. Ramchandani S, MacLeod AR, Pinard M et al: Inhibition of tumorigenesis by a cytosine-DNA, methyltransferase, antisense oligodeoxynucleotide. Proc Natl Acad Sci USA 94: 684-689, 1997. Schutz BR, Scheurlen W, Krauss J et al: Mapping of chromosomal gains and losses in primitive neuroectodermal tumors by comparative genomic hybridization. Genes Chromosomes Cancer 16: 196-203, 1996. Teitz T, Wei T, Valentine MB et al: Caspase-8 is deleted or silenced preferentially in childhood neuroblastomas with amplification of MYCN. Nat Med 6: 529-535, 2000. Zochbauer-Muller S, Fong KM, Virmani AK et al: Aberrant promoter methylation of multiple genes in non-small cell lung cancers. Cancer Res 61: 249-255, 2001. Zuzak TJ, Steinhoff DF, Sutton LN et al: Loss of caspase-8 mRNA expression is common in childhood primitive neuroectodermal brain tumour/medulloblastoma. Eur J Cancer 38: 83- 91, 2002. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 17 EP 21 PG 5 ER PT J AU Raso, E Meszaros, L Albini, A Timar, J AF Raso, Erzsebet Meszaros, Livia Albini, Adriana Timar, Jozsef TI A WT1 Expressing Metastatic Human Kaposi Sarcoma Xenograft Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Kaposi sarcoma; xenograft model; WT1 expression; metastasis ID Kaposi sarcoma; xenograft model; WT1 expression; metastasis AB We have established a non-metastatic and a metastatic human Kaposi sarcoma (KS) xenograft model in SCID mice by injecting KS-Imm cells subcutaneously and intrasplenically, respectively. KS-Imm cells expressed endothelial markers, CD34 and vWF in vivo. Futhermore, we have shown that these cells express all the splice variants of the WT1 gene and WT1wt protein in vitro and in vivo detected by nested PCR and immunohistochemistry. WT1 expression in the peripheral blood was only detectable in case of metastatic KS model suggesting it as a molecular marker of progression. C1 [Raso, Erzsebet] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Meszaros, Livia] National Institute of Oncology, Department of Diagnostic PathologyBudapest, Hungary. [Albini, Adriana] Istituto Nazionale per la Ricerca sul CancroGenova, Italy. [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. CR Bais C, van Geelen A, Eroles P et al.: Kaposi’s sarcoma associated herpesvirus G protein-coupled receptor importalized human endothelial cells by activation of the VEGF receptor-2/ KDR. Cancer Cell 3:131-143, 2003 Montaner S, Sodhi A, Molinolo A et al.: Endothelial infection with KSHV genes in vivo reveals that vGPCR initiates Kaposi’s sarcomagenesis and can promote the tumorigenic potential of viral latent genes. Cancer Cell 3: 23-36, 2003 Moses AV, Jarvis MA, Raggo C et al: Kaposi’s sarcoma-associatd herpesvirus-induced upregulation of the c-kit proto-oncogene, as identified by gene expression profiling, is essential for the tansformation of endothelial cells. J Virol 76:8383-8399, 2002 Folpe AL, Veikkola T, Valtola R, Weiss SW: Vascular endothelial growth factor receptor-3, VEGFR-3): a marker of vascular tumors with presumed lymphatic differentiation, including Kaposi’s sarcoma, kaposiform and dabska-type hemangioendotheliomas, and a subset of angiosarcomas. Mod Pathol 13:180-185,2000 Jussila L, Valtola R, Partanen TA et al: Lymphatic endothelium and Kaposi’s sarcoma spindle cells detected by antibodies against the vascular endothelial growth factor receptor-3. Cancer Res 58: 1599- 04, 1998 6. Kahn HJ, Bailey D and Marks A: Monoclonal antibody D2-40, a new maker of lymphatic endothelium, reacts with Kaposi’s sarcoma and a subset of angiosarcomas. Mod Pathol 15: 434-440, 2002 Simonart T, Degraef C, Heenen M et al: Expression of the fibroblast/ macrophage marker 1B10 by spindle cells in Kaposi’s sarcoma lesions and by Kaposi’s sarcoma-derived tumor cells. J Cutan Pathol 29: 72-78, 2002 Maurer U, Brieger J, Wiedmann E et al: The Wilm’s tumor gene is expressed in a subset of CD34+ progenitors and downregulated in the course of differentiation in vitro. Exp Hematol 25:945-950, 1997. Gehling UM, Ergun S, Schumacher U, Wagener Ch et al: In vitro differentiation of endothelial cells from AC133-positive progenitor cells. Blood 95:3106-3112, 2000 Moore MAS: Putting the neo into neoangiogenesis. J Clin Invest 109:313-213,2002 Rafii S, Lyden D, Benezra R et al: Vascular and hamatopoietic stem cells: novel targets for anti-angiogenesis therapy? Nat Rev Cancer 2:826-835, 2002 Albini A, Paglieri I, Orengo G et al: The beta core fragment of human choriogonadotrophin inhibits growth of Kaposi Sarcoma derived cells and a new immortalized Kaposi sarcoma cell line. AIDS 11:713-721, 1997 Lunardi-Iskandar Y, Gill P, Lam VH et al: Isolation and characterization of an immortal neoplastic cell, KS Y-1, from AIDS-Associated Kaposi’s sarcoma. J Natl Cancer Inst 87: 974-949, 1995 Possati L, Campioni D, Sola F et al. Antiangiogenic, antitumoural and antimetasttaic effects of the distamycin A derivatives with anti- HIV-1 Tat activity in a Kaposi’s sarcoma-like murine model. Clin Exp Metast 17:575-582,1999. Kovacs L, Szende B, Elek G et al: Working experience with a new vacuum-accelerated microwave histoprocessor. J Pathol 180:108- 110, 1996 Hirose M: The role of Wilm’s tumor genes. J Med Invest 46:130- 140,1999. Ellisen LW, Carlesso N, Cheng T et al: The Wilms tumor suppressor WT1 directs stage-specific quiescence and differentiation of human hematopoietic progenitor cells. EMBO J 20:1897-1909, 2001 Magyarosy E, Varga N, Timar J, Raso E. Follow-up of minimal residual disease in acute childhood lymphoblastic leukemia by WT1 gene expression in the peripheral blood: The Hungarian experience. Pediatric Haematol Oncol 20:65-74,2003 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 22 EP 25 PG 4 ER PT J AU Wu, Q Suo, Z Risberg, B Karlsson, GM Villman, K Nesland, MJ AF Wu, Qinghua Suo, Zhenhe Risberg, Bjorn Karlsson, G Mats Villman, Kenneth Nesland, M Jahn TI Expression of Ephb2 And Ephb4 in Breast Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EphB2; EphB4; breast carcinoma; RT-PCR; immunohistochemistry ID EphB2; EphB4; breast carcinoma; RT-PCR; immunohistochemistry AB Eph receptor tyrosine kinases and their cell-surfacebound ligands, the ephrins, play key roles in diverse biological processes. Eph receptors comprise the largest family of receptor tyrosine kinases consisting of eight EphA receptors (with five corresponding ephrinA ligands) and six EphB receptors (with three corresponding transmembrane ephrinB ligands). Originally identified as neuronal pathfinding molecules, EphB receptors and ephrinB ligands are later proved to be crucial regulators of vasculogenesis and embryogenesis. More studies indicate that Eph receptors are involved in angiogenesis and tumorigenesis. This study aimed to investigate the expression of EphB2 and EphB4 in breast carcinomas. Semiquantitative RT-PCR and immunohistochemistry were used to examine the expression patterns of EphB2 and EphB4. Clinicopathological and survival correlations were statistically analyzed in a series of 94 breast carcinomas, 9 normal specimens and 4 breast carcinoma cell lines. 1(1%), 16(17%), 29(31%), 48(51%) of the 94 tumors were negative, weak, moderate and strong EphB2 protein expression, respectively. 6(6%), 27(29%), 28(30%), 33(35%) of the tumors were negative, weak, moderate and strong EphB4 expression, respectively. Both EphB2 and EphB4 RTPCR products could be detected in all specimens. Increased EphB2 protein expression was negatively associated with overall survival, and there was a trend that increased EphB2 protein expression was correlated with shorter disease free survival, while EphB4 protein expression was associated with histological grade and stage. EphB4 membrane staining was increased with S phase fraction and associated with DNA aneuploidy. These findings indicate that both EphB2 and EphB4 are involved in the development of breast cancer and that both molecules could be potential predictive markers. C1 [Wu, Qinghua] The Norwegian Radium Hospital, University of Oslo, Department of Pathology, N-0310 Oslo, Norway. [Suo, Zhenhe] The Norwegian Radium Hospital, University of Oslo, Department of Pathology, N-0310 Oslo, Norway. [Risberg, Bjorn] The Norwegian Radium Hospital, University of Oslo, Department of Pathology, N-0310 Oslo, Norway. [Karlsson, G Mats] Orebro Medical Center Hospital, Department of PathologyOrebro, Sweden. [Villman, Kenneth] Orebro Medical Center Hospital, Department of OncologyOrebro, Sweden. [Nesland, M Jahn] The Norwegian Radium Hospital, University of Oslo, Department of Pathology, N-0310 Oslo, Norway. RP Suo, Z (reprint author), The Norwegian Radium Hospital, University of Oslo, Department of Pathology, N-0310 Oslo, Norway. EM zhenhes@labmed.uio.no CR van der Geer P, Hunter T, Lindberg RA: Receptor protein-tyrosine kinases and their signal transduction pathways. Ann Rev Cell Biol 10: 251-337, 1994. Gale N., Holland SJ., Valenzuela DM, et al: Eph receptors and ligands comprise two major specificity subclasses and are reciprocally compartmentalized during embryogenesis. Neuron 17: 9-19, 1996. Davy A, Gale NW, Murray EW, et al: Compartmentalized signaling by GPI-anchored ephrin-A5 requires the Fyn tyrosine kinase to regulate cellular adhesion. Genes Dev 13: 3125-3135, 1999. Davy A., Robbins SM: Ephrin-A5 modulates cell adhesion and morphology in an integrin-dependent manner. Embo J 19: 5396-5405, 2000. Kullander K, Klein R: Mechanisms and functions of Eph and ephrin signalling. Nat Rev Mol Cell Biol 3: 475-486, 2002. Flanagan JG, Vanderhaeghen P: The ephrins and Eph receptors in neural development. Ann Rev Neurosci 21: 309-345, 1998. Tessier-Lavigne M: Eph receptor tyrosine kinases, axon repulsion, and the development of topographic maps.Cell 82: 345-348, 1995. Wang HU, Anderson DJ: Eph family transmembrane ligands can mediate repulsive guidance of trunk neural crest migration and motor axon outgrowth. Neuron 18: 383-396, 1997. Winslow JW, Moran P, Valverde J, et al: Cloning of AL-1, a ligand for an Eph-related tyrosine kinase receptor involved in axon bundle formation. Neuron 14: 973-981, 1995. Chinoy MR, Graybill MM, Miller SA, et al: Angiopoietin-1 and VEGF in vascular development and angiogenesis in hypoplastic lungs. Am J Physiol Lung Cell Mol Physiol 283: L60-66, 2002. Li H, Fredriksson L, Li X, et al: PDGF-D is a potent transforming and angiogenic growth factor. Oncogene 22: 1501-1510, 2003. Goede V., Fleckenstein G, Dietrich M, et al: Prognostic value of angiogenesis in mammary tumors. Anticancer Res 18: 2199- 2210, 1998. Kuehn R, Lelkes PI, Bloechle C, et al: Angiogenesis, angiogenic growth factors, and cell adhesion molecules are upregulated in chronic pancreatic diseases: angiogenesis in chronic pancreatitis and in pancreatic cancer. Pancreas 18: 96-103, 1999. Weidner N, Folkman J, Pozza F, et al: Tumor angiogenesis: a new significant and independent prognostic indicator in earlystage breast carcinoma. J Natl Cancer Inst 84: 1875-1882, 1992. Zelinski DP, Zantek ND, Stewart JC, et al: EphA2 overexpression causes tumorigenesis of mammary epithelial cells. Cancer Res 61: 2301-2311, 2001. Miyazaki T, Kato H, Fukuchi M, et al: EphA2 overexpression correlates with poor prognosis in esophageal squamous cell carcinoma. Int J Cancer 103: 657-668, 2003. Walker-Daniels J, Coffman K, Azimi M, et al: Overexpression of the EphA2 tyrosine kinase in prostate cancer. Prostate 41: 275-281, 1999. Ikegaki N, Tang XX, Liu XG, et al: Molecular characterization and chromosomal localization of DRT, EPHT3): a developmentally regulated human protein-tyrosine kinase gene of the EPH family. Hum Mol Genet 4: 2033-2041, 1995. Nikolova Z, Djonov V, Zuercher G, et al: Cell-type specific and estrogen dependent expression of the receptor tyrosine kinase EphB4 and its ligand ephrin-B2 during mammary gland morphogenesis. J Cell Sci 111: 2741-2750, 1998. Cromer A, Carles A, Millon R, et al: Identification of genes associated with tumorigenesis and metastatic potential of hypopharyngeal cancer by microarray analysis. Oncogene, in press), 2004. Kiyokawa E, Takai S, Tanaka M, et al: Overexpression of ERK, an EPH family receptor protein tyrosine kinase, in various human tumors. Cancer Res 54: 3645-3652, 1994. Tang XX, Brodeur GM, Campling BG, et al: Coexpression of transcripts encoding EPHB receptor protein tyrosine kinases and their ephrin-B ligands in human small cell lung carcinoma. Clin Cancer Res 5: 455-463, 1999. Tang XX, Evans AE, Zhao H, et al: High-level expression of EPHB6, EFNB2, and EFNB3 is associated with low tumor stage and high TrkA expression in human neuroblastomas. Clin Cancer Res 5: 1491-1501, 1999. Tang XX, Evan, AE, Zhao H, et al: Association among EPHB2, TrkA, and MYCN expression in low-stage neuroblastomas. Med Pediatr Oncol 36: 80-86, 2001. Liu W, Ahmad SA, Jung YD, et al: Coexpression of ephrin-Bs and their receptors in colon carcinoma. Cancer 94: 934-943, 2002. Kataoka H, Tanaka M, Kanamori M, et al: Expression profile of EFNB1, EFNB2, two ligands of EPHB2 in human gastric cancer. J Cancer Res Clin Oncol 128: 343-352, 2002. Bennett BD, Wang Z, Kuang WJ, et al: Cloning and characterization of HTK, a novel transmembrane tyrosine kinase of the EPH subfamily. J Biol Chem 269: 14211-14220, 1994. Bercla G, Karamitopoulou E, Mazzucchelli L, et al: Activation of the receptor protein tyrosine kinase EphB4 in endometrial hyperplasia and endometrial carcinoma. Ann Oncol 14: 220- 227, 2003. Stephenson SA, Slomka S, Douglas EL, et al: Receptor protein tyrosine kinase EphB4 is up-regulated in colon cancer. BMC Mol Biol 2: 15-21, 2001. Berclaz G, Flutsch B, Altermatt HJ, et al: Loss of EphB4 receptor tyrosine kinase protein expression during carcinogenesis of the human breast. Oncol Rep 9: 985-993, 2002. Elston CW, Elli, IO: Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19: 403-411, 1991. Suo Z, Berner HS, Risberg B, et al: Estrogen receptor-alpha and C-ERBB-4 expression in breast carcinomas. Virchows Arch 439: 62-73, 2001. Himanen JP, Rajashankar KR, Lackmann M, et al: Crystal structure of an Eph receptor-ephrin complex. Nature 414: 933- 939, 2001. Kalo MS, Pasquale EB: Multiple in vivo tyrosine phosphorylation sites in EphB receptors. Biochemistry 38: 14396-14401, 1999. Dodelet VC, Pazzagli C, Zisch AH, et al: A novel signaling intermediate, SHEP1, directly couples Eph receptors to R-Ras and Rap1A. J Biol Chem 274: 31941-31946, 1999. Holland SJ, Gale NW, Gish GD, et al: Juxtamembrane tyrosine residues couple the Eph family receptor EphB2/Nuk to specific SH2 domain proteins in neuronal cells. Embo J 16: 3877-3884, 1997. Ellis C, Kasmi F, Ganju P, et al: A juxtamembrane autophosphorylation site in the Eph family receptor tyrosine kinase, Sek, mediates high affinity interaction with p59fyn. Oncogene 12: 1727-1733, 1996. Steinle JJ, Meininger CJ, Forough R, et al: Eph B4 receptor signaling mediates endothelial cell migration and proliferation via the phosphatidylinositol 3-kinase pathway. J Biol Chem 277: 43830-43836, 2002. Vindis C, Cerretti DP, Daniel TO, et al: EphB1 recruits c-Src and p52Shc to activate MAPK/ERK and promote chemotaxis. J Cell Biol 162: 661-669, 2003. Stein E, Cerretti DP, Daniel TO: Ligand activation of ELK receptor tyrosine kinase promotes its association with Grb10 and Grb2 in vascular endothelial cells. J Biol Chem 271: 23588-23596, 1996. Stein E, Huynh-Do U, Lane AA, et al: Nck recruitment to Eph receptor, EphB1/ELK, couples ligand activation to c-Jun kinase. J Biol Chem 273: 1303-1312, 1998. Hock B, Bohme B, Karn T, et al: PDZ-domain-mediated interaction of the Eph-related receptor tyrosine kinase EphB3 and the ras-binding protein AF6 depends on the kinase activity of the receptor. Proc Natl Acad Sci U S A 95: 9779-9785, 1998. Stein E, Lane AA, Cerretti DP, et al: Eph receptors discriminate specific ligand oligomers to determine alternative signaling complexes, attachment, and assembly responses. Genes Dev 12: 667-672, 1998. Wybenga-Groot LE, Baskin B, Ong SH, et al: Structural basis for autoinhibition of the Ephb2 receptor tyrosine kinase by the unphosphorylated juxtamembrane region. Cell 106: 745-753, 2001. Matsuzaki M, Nagase S, Abe T, et al: Detailed deletion mapping on chromosome 1p32-p36 in human colorectal cancer: identification of three distinct regions of common allelic loss. Int J Oncol 13: 1229-1238, 1998. Praml C, Finke LH, Herfarth C, et al: Deletion mapping defines different regions in 1p34.2-pter that may harbor genetic information related to human colorectal cancer. Oncogene 11: 1357-1367, 1995. Oba SM, Wang YJ, Song JP, et al: Genomic structure and loss of heterozygosity of EPHB2 in colorectal cancer. Cancer Lett 164: 97-109, 2001. Adams RH: Vascular patterning by Eph receptor tyrosine kinases and ephrins. Semin Cell Dev Biol 13: 55-63, 2002. Wang HU, Chen ZF, Anderson DJ: Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4. Cell 93: 741- 753, 1998. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 26 EP 33 PG 8 ER PT J AU Mazur, G Wozniak, Z Wrobel, T Maj, J Kuliczkowski, K AF Mazur, Grzegorz Wozniak, Zdzislaw Wrobel, Tomasz Maj, Joanna Kuliczkowski, Kazimierz TI Increased Angiogenesis in Cutaneous T-cell Lymphomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE angiogenesis; microvessel density; cutaneous T-cell lymphoma; mycosis fungoides ID angiogenesis; microvessel density; cutaneous T-cell lymphoma; mycosis fungoides AB Primary cutaneous T-cell lymphomas (CTCL) represent a heterogeneous group of neoplasms derived from skin-homing T cells. CTCL behave similarly to indolent B-cell lymphomas. There is increasing evidence that angiogenesis may be important in lymphoproliferative disorders. The aim of the study was to evaluate microvessel density (MVD) as a parameter of tumor angiogenesis measured by the expression of CD34 in the skin samples in CTCL patients. Formaldehyde-fixed, paraffin-embedded skin tumor biopsy specimens from 25 patients (16 men, 9 women) with CTCL (mycosis fungoides), and 8 skin samples from healthy volunteers were analysed. The preparations were stained with haematoxylin and eosin, and evaluated histopathologically. Staining for endothelial cells with monoclonal antibody against CD34 revealed a mean number of 134 dots per mm2 for CTCL and 106 dots/mm2 for controls; the difference was statistically significant (p=0.0388). Our study shows a higher number of microvessels in primary CTCL compared with normal skin. Microvascular endothelial cells have become an important target in cancer therapy. Increased MVD in the skin of CTCL patients indicate that angiogenesis may play a role in the growth of CTCL, and raises the possibility of using angiogenesis inhibitors in CTCL therapy. C1 [Mazur, Grzegorz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, Pasteura 4, 50-367 Wroclaw, Poland. [Wozniak, Zdzislaw] Wroclaw Medical University, Department of PathologyWroclaw, Poland. [Wrobel, Tomasz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, Pasteura 4, 50-367 Wroclaw, Poland. [Maj, Joanna] Wroclaw Medical University, Department of Dermatology, Venereology and AllergologyWroclaw, Poland. [Kuliczkowski, Kazimierz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, Pasteura 4, 50-367 Wroclaw, Poland. RP Mazur, G (reprint author), Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, 50-367 Wroclaw, Poland. EM grzegmaz@hemat.am.wroc.pl CR Folkman J: Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med 1:27-31, 1995 Folkman J, Klagsbrun M: Angiogenic factors. Science 235: 442-447, 1987 Kim YH, Hoppe RT: Mycosis fungoides and the Sezary Syndrome. Semin Oncol 26: 276-289, 1999 Mangi MH, Newland AC: Angiogenesis and angiogenic mediators in haematological malignancies. Br J Haematol 111: 43- 51, 2000 Ribatti D, Vacca A, Nico B, et al: Angiogenesis spectrum in the stroma of B cell NHL. An immunohistochemical and ultrastructure study. Eur J Haematol 56: 45-53, 1996 Ridell B, Norrby K: Intratumoral microvascular density in malignant lymphomas of B-cell origin. APMIS 109: 66-72, 2001 Salven P, Teerenhovi L, Joensuu H: A high pretreatment serum basic fibroblast growth factor concentration is an independent predictor of poor prognosis in non-Hodgkin lymphoma. Blood 94: 3334-3339, 1999 Schaerer L, Hess-Schmid M, Mueller B, et al: Angiogenesis in cutaneous lymphoproliferative disorders: microvessel density discriminates between cutaneous B-cell lymphomas and B-cell pseudolymphomas. Am J Dermatopathol 22: 140-143, 2000 Siegel RS, Pandolfino T, Guitart J, et al: Primary cutaneous Tcell lymphoma: review and current concepts. J Clin Oncol 18: 2908-2925, 2000 Vacca A, Moretti S, Ribatti D et al: Progression of mycosis fungoides is associated with changes in angiogenesis and expression of the matrix metalloproteinases 2 and 9. Eur J Cancer 33: 1685-1692, 1997 Vacca A, Ribatti D, Roncali L, Dammacco F: Angiogenesis in B cell lymphoproliferative diseases. Biological and clinical studies. Leuk Lymphoma 20: 27-38, 1995 Weidner N, Semple JP, Welch WR, Folkman J: Tumor angiogenesis and metastasis: correlation in invasive breast carcinoma. N Engl J Med 324:1-8, 1991 Willemze R, Kerl H, Sterry W, et al: EORTC classification for primary cutaneous lymphomas: a proposal from the cutaneous lymphoma study group of the European Organization for Research and Treatment. Blood 90: 354-371,1997 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 34 EP 36 PG 3 ER PT J AU Amirghofran, Z Monabati, A Gholijani, N AF Amirghofran, Zahra Monabati, Ahmad Gholijani, Naser TI Androgen Receptor Expression in Relation to Apoptosis and the Expression of Cell Cycle Related Proteins in Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prostate carcinoma; androgen receptor; apoptosis; p53; bcl-2; bax; Ki-67 ID prostate carcinoma; androgen receptor; apoptosis; p53; bcl-2; bax; Ki-67 AB The expression of several genes involved in the regulation of cell cycle and apoptosis may be regulated via the androgen receptor (AR) in the prostate. AR may have a role in the prognosis of prostatic carcinoma. The aim was to examine AR expression status and its relationship with markers of proliferation, apoptosis and cell cycle control in prostate cancer. Expression of AR, bcl-2, bax, Ki-67 and p53 was examined in paraffin-embedded tissues from 50 cases of prostate carcinoma by immunohistochemistry and evaluated using an index of staining. Detection of apoptotic cells was performed by TUNEL method. Correlation between AR expression and apoptosis, proliferation index, bcl-2, bax and p53 and also clinicopathological parameters including stage, pathological grade and Gleason score were determined. AR expression was observed in all cases with mean expression of 81%+/-15 and mean staining index of 141+/-65. No correlation was found between AR expression and apoptosis detected in patients. The mean AR staining index was 170+/-72 in bcl-2 positive tumors versus 120+/-53 in bcl-2 negative tumors showing a significant association between AR and bcl-2 expression (p=0.015). AR expression also showed a significant association with bcl-2/bax ratio (r=0.321, p=0.023) and Ki-67 proliferation staining index (r=0.396, p=0.004). Although a significant correlation between Ki-67 and p53 with differentiation status of the tumors was observed (p<0.004) no correlation was found with AR. AR expression showed no prognostic value regarding its correlation with stage and differentiation status of the prostate carcinoma. However, its significant correlation with Ki-67 and bcl-2 that are markers of cell survival suggest its contribution to tumor cell progression. C1 [Amirghofran, Zahra] Medical School, Shiraz University of Medical Sciences, Department of Immunology, 71345-1798 Shiraz, Iran. [Monabati, Ahmad] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Gholijani, Naser] Medical School, Shiraz University of Medical Sciences, Department of Immunology, 71345-1798 Shiraz, Iran. RP Amirghofran, Z (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Immunology, 71345-1798 Shiraz, Iran. EM amirghz@sums.ac.ir CR Buttyan R, Shabsigh A, Perlman H, Colombel M. Regulation of Apoptosis in the Prostate Gland by Androgenic Steroids. Trends Endocrinol Metab 10:47-54, 1999 Chang C, Saltzman A, Yeh S et al.: Androgen receptor: an overview. Crit Rev Eukaryot Gene Exp 5: 97-125, 1995 Eder IE, Culig Z, Putz T et al.: Molecular biology of the androgen receptor: from molecular understanding to the clinic. Eur Urol 40:241-51, 2001 Klocker H, Culig Z, Eder IE et al: Mechanism of androgen receptor activation and possible implications for chemoprevention trials. Eur Urol 35: 413-419, 1999. Kyprianou N, Isaacs JT: Activation of programmed cell death in the rat ventral prostate after castration. Endocrinology 122: 552-562, 1988 Gelmann EP. Molecular biology of the androgen receptor. J Clin Oncol 20: 3001-3015, 2002 Feng Z, Joos HJ, Vallan C et al: Apoptosis during castrationinduced regression of the prostate is Fos dependent. Oncogene 19: 17: 2593-600, 1998 Huggins C, Hodges CV. Studies on prostatic cancer: I. The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. J Urol 168: 9-12, 2002 Culig Z, Klocker H, Bartsch G et al: Androgen receptors in prostate cancer. J Urol 170: 1363-1369 2003. Culig Z, Hobisch A, Erdel M et al: Studies on androgen receptor mutations and amplification in human prostatecancer. Methods Mol Med 81: 267-275, 2003 Sadi MV, Barrack ER: Image analysis of androgen receptor immunostaining in metastatic prostate cancer. Heterogeneity as a predictor of response to hormonal therapy. Cancer 15: 71: 2574-2580, 1993 Miyamoto KK, McSherry SA, Dent GA et al.: Immunohistochemistry of the androgen receptor in human benign and malignant prostate tissue. J Urol 149:1015-1019, 1993 de Winter JA, Trapman J, Brinkmann AO et al: Androgen receptor heterogeneity in human prostatic carcinomas visualized by immunohistochemistry. J Pathol 160: 329-332, 1990 Shaffer DR, Scher HI. Prostate cancer: a dynamic illness with shifting targets. Lancet Oncol 4: 407-414, 2003 Magi-Galluzzi C, Xu X, Hlatky L et al: Heterogeneity of androgen receptor content in advanced prostate cancer. Mod Pathol 10: 839-845, 1997 Sweat SD, Pacelli A, Bergstralh EJ et al: Androgen receptor expression in prostatic intraepithelial neoplasia and cancer. J Urol 161: 1229-1232, 1999 Yoshikawa H, Ikeuchi T, Kai Y. Immunohistochemical study of androgen receptor in adenocarcinoma of the human prostatic cancer. Nippon Hinyokika Gakkai Zasshi 87: 956-963, 1996 van der Kwast TH, Schalken J, Ruizeveld de Winter JA et al: Androgen receptors in endocrine-therapy-resistant human prostate cancer. Int J Cancer 48: 189-193, 1991 Takeda H, Akakura K, Masai M et al.: Androgen receptor content of prostate carcinoma cells estimated by immunohistochemistry is related to prognosis of patients with stage D2 prostate carcinoma. Cancer 77: 934-940, 1996 Finnegan NM, Curtin JF, Prevost G et al: Induction of apoptosis in prostate carcinoma cells by BH3 peptides which inhibit Bak/Bcl-2 interactions. Br J Cancer 85: 115-121, 2001 DiPaola RS, Aisner J. Overcoming bcl-2- and p53-mediated resistance in prostate cancer. Semin Oncol 26(Suppl, 2: 112- 116, 1999 Kolar Z, Murray PG, Madarova J et al: Nuclear receptors in early hormone refractory prostate cancer and their relationship to apoptosis-related proteins. Neoplasma 49:172-177, 2002 Fizazi K, Martinez LA, Sikes CR et al: The association of p21, WAF-1/CIP1, with progression to androgen-independent prostate cancer. Clin Cancer Res 8: 775-781, 2002 Tanji N. Yokoyama M. Sagamoto T et al: Apoptosis in prostatic adenocarcinoma; A study of relationship to Ki-67 and bcl-2 protein expression. Anticancer Res 18:1111-1116, 1998 Kaltz-Wittmer C, Klenk U, Glaessgen A et al: FISH analysis of gene aberrations, MYC, CCND1, ERBB2, RB, and AR, in advanced prostatic carcinomas before and after androgen deprivation therapy. Lab Invest 80: 1455-1464, 2000 Baretton GB, Klenk U, Diebold J et al: Proliferation- and apoptosis-associated factors in advanced prostatic carcinomas before and after androgen deprivation therapy: prognostic significance of p21/WAF1/CIP1 expression. Br J Cancer 80: 546- 555, 1999 Cadwell C, Zambetti GP: The effects of wild-type p53 tumor suppressor activity and mutant p53 gain-of-function on cell growth. Gene 17: 15-30, 2001 Brooks JD, Bova GS, Ewing CM et al: An uncertain role for p53 gene alterations in human prostate cancers. Cancer Res 56: 3814-3822. 1996 Navone NM, Troncoso P, Pisters LL, et al.: p53 protein accumulation and gene mutation in the progression of human prostate carcinoma. J Natl Cancer Inst 85: 1657-1669, 1993 Aprikian AG, Sarkis AS, Fair WR et al: Immunohistochemical determination of p53 protein nuclear accumulation in prostatic adenocarcinoma. J Urol 151: 1276-1280, 1994 Bargonetti J, Manfredi JJ. Multiple roles of the tumor suppressor p53. Curr Opin Oncol 14: 86-91, 2002 Howell SB. Resistance to apoptosis in prostate cancer cells. Mol Urol 4: 225-229, 2000 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 37 EP 41 PG 5 ER PT J AU Esik, O Emri, M Szakall, Sz Herzog, H Safrany, G Lengyel, E Boer, A Liszkay, G Tron, L Lengyel, Zs Repa, I AF Esik, Olga Emri, Miklos Szakall, Szabolcs Herzog, Hans Safrany, Geza Lengyel, Erzsebet Boer, Andras Liszkay, Gabriella Tron, Lajos Lengyel, Zsolt Repa, Imre TI PET Identifies Transitional Metabolic Change in the Spinal Cord Following a Subthreshold Dose of Irradiation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE radiotherapy; spinal cord; positron emission tomography; [18F]fluorodeoxyglucose; [11C]methionine; [15O]butanol; radiobiology ID radiotherapy; spinal cord; positron emission tomography; [18F]fluorodeoxyglucose; [11C]methionine; [15O]butanol; radiobiology AB Positron emission tomographic (PET) investigations were performed to obtain in vivo information on symptomless radiation-induced pathological changes in the human spinal cord. PET investigations were carried out prior to radiotherapy and during the regular follow-up in an early hypopharyngeal cancer patient (the spinal cord was irradiated with a biologically effective dose of 80 Gy2), with [18F]fluorodeoxyglucose (FDG), [11C]methionine and [15O]butanol as tracers; radiosensitivity and electroneuronographic (ENG) studies were also performed. A very low background FDG accumulation (mean standardized uptake values, i.e. SUV: 0.84) was observed in the spinal cord before the initiation of radiotherapy. An increased FDG uptake was measured 2 months after the completion of radiotherapy (mean SUV: 1.69), followed by a fall-off, as measured 7 months later (mean SUV: 1.21). By 44 months after completion of irradiation, the FDG accumulation in the irradiated segments of the spinal cord had decreased to a level very close to the initial value (mean SUV: 1.11). The simultaneous [15O]butanol uptake results demonstrated a set of perfusion changes similar to those observed in connection with the FDG accumulation. The patient exhibited an extremely low [11C]methionine uptake within the irradiated and the nonirradiated spinal cord during the clinical course. She has not had any neurological symptoms, and the results of central ENG measurements before radiotherapy and 2 months following its completion proved normal. Radiobiological investigations did not reveal unequivocal signs of an increased radiosensitivity. A transitory increased spinal cord FDG uptake following radiotherapy may be related to the posttherapeutic mild inflammatory and regenerative processes. The normal [11C]methionine accumulation observed is strong evidence against intensive cell proliferation. The high degree of normalization of the temporarily increased FDG uptake of the irradiated spinal cord segments by 44 months is in good agreement with the results of monkey studies, which demonstrated a nearly complete recovery from radiation-induced spinal cord injury. C1 [Esik, Olga] University of Pecs, Department of Oncology, Edesanyak utja 17., H-7624 Pecs, Hungary. [Emri, Miklos] University of Debrecen, PET CenterDebrecen, Hungary. [Szakall, Szabolcs] University of Debrecen, PET CenterDebrecen, Hungary. [Herzog, Hans] Research Center, Institute of Medicine, PET LaboratoryJulich, Germany. [Safrany, Geza] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Frederic Joliot-Curie Sugarbiologiai es Sugaregeszsegugyi Kutato IntezetBudapest, Hungary. [Lengyel, Erzsebet] Semmelweis University, Department of Radiology and OncotherapyBudapest, Hungary. [Boer, Andras] National Institute of Oncology, Department of Head and Neck SurgeryBudapest, Hungary. [Liszkay, Gabriella] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Tron, Lajos] University of Debrecen, PET CenterDebrecen, Hungary. [Lengyel, Zsolt] University of Debrecen, PET CenterDebrecen, Hungary. [Repa, Imre] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. RP Esik, O (reprint author), University of Pecs, Department of Oncology, H-7624 Pecs, Hungary. EM olga.esik@aok.pte.hu CR Jones A: Transient radiation myelopathy, with reference to Lhermitte’s sign of electrical paraesthesia). Br J Radiol 37:727-744, 1964 Schultheiss TE, Stephens LC, Jiang GL, et al: Radiation myelopathy in primate treated with conventional fractionation. Int J Radiat Oncol Biol Phys 19:935-940, 1990 Schultheiss TE, Stephens LC, Ang KK, et al: Volume effects in rhesus monkey spinal cord. Int J Radiat Oncol Biol Phys 30:67-72, 1994 Ang KK, Price RE, Stephens LC, et al: The tolerance of primate spinal cord to re-irradiation. Int J Radiat Oncol Biol Phys 15:459- 464, 1993 Ang KK, Jiang G-L, Feng Y, et al: Extent and kinetics of recovery of occult spinal cord injury. Int J Radiat Oncol Biol Phys 50:1013- 1020, 2001 Lossos A, Siegal T: Electric shock-like sensations in 42 cancer patients: clinical characteristics and distinct etiologies. J Neurooncol 29:175-181, 1996 Traynelis VC, Hitchon PW, Yuh WT, et al: Magnetic resonance imaging and posttraumatic Lhermitte’s sign. J Spinal Disord 3:376-379, 1990 van den Bent MJ, Hilkens PHE, Sillevis Smitt PAE, et al: Lhermitte’s sign following chemotherapy with docetaxel. Neurology 50:563- 564, 1998 Fowler JF: The linear-quadratic formula and progress in fractionated radiotherapy. Br J Radiol 62:679-694, 1989 Lipinski B, Herzog H, Rota Kops E, et al: Expectation maximization reconstruction of positron emission tomography images using anatomical magnet resonance information. IEEE Trans Med Imaging 16:129-136, 1997 Freshney RI: Culture of animal cells: a manual of basic techniques. AR Liss Inc, New York, pp 113-115, 1987 Klaude M, Eriksson S, Nygren J, et al: The comet assay: mechanisms and technical considerations. Mutat Res 363:89-96, 1996 Singh NP, McCoy MT, Tice RR, et al: A simple technique for quantitation of low levels of DNA damage in individual cells. Exp Cell Res 175:184-191, 1988 Elkind MM, Whitmore GF: The radiobiology of cultured mammalian cells. Gordon and Breach, New York, 1967 Barendsen GW, Beusker TLJ, Vergroesen AJ, et al: Effects of different ionizing radiations on human cells in tissue culture. II. Biological experiments. Radiat Res 13:841-849, 1960 Di Chiro G, Oldfield E, Bairamian D, et al: Metabolic imaging of the brain stem and spinal cord: studies with positron emission tomography using 18F-2-deoxyglucose in normal and pathological cases. J Comput Assist Tomogr 7:937-945, 1983 Kamoto Y, Sadato N, Yonekura Y, et al: Visualization of the cervical spinal cord with FDG and high-resolution PET. J Comput Assist Tomogr 22:487-491, 1998 Meltzer CC, Townsend DW, Kottapally S, et al: FDG imaging of spinal cord primitive neuroectodermal tumor. J Nucl Med 39:2121- 2127, 1998 Wilmshurst JM, Barrington SF, Pritchard D, et al:Positron emission tomography in imaging spinal cord tumors. J Child Neurol 15:465- 472, 2000 Woesler B, Kuwert T, Probst-Cousin S, et al: Spinal metastases of a high grade astrocytoma visualized with FDG-PET. Clin Nucl Med 22:863-864, 1997 Higano S, Shishido F, Nagashima M, et al: PET evaluation of spinal cord tumor using 11C-methionine. J Comput Assist Tomogr 14:297- 299, 1990 Sasajima T, Mineura K, Itoh Y, et al: Spinal cord ependymoma: a positron emission tomographic study with, 11C-methyl)-L-methionine. Neuroradiology 38:53-55, 1996 Esik O, Emri M, Csornai M, et al: Radiation myelopathy with partial functional recovery: PET evidence of long-term increased metabolic activity of the spinal cord. J Neurol Sci 163:39-43, 1999 Esik O, Lengyel Z, Safrany G, et al:A PET study on the characterization of partially reversible radiogenic lower motor neurone disease. Spinal Cord 40:468-473, 2002 Roland PE: Brain Activation. Wiley-Liss Inc, New York, pp 469- 504, 1993 Janus TJ, Kim EE, Tilbury R, et al: Use of 18Ffluorodeoxyglucose positron emission tomography in patients with primary malignant brain tumors. Ann Neurol 33:540-548, 1993 Godwin-Austen RB, Howell DA, Worthington B: Observations on radiation myelopathy. Brain 98:557-568, 1975 Jellinger K, Sturm KW: Delayed radiation myelopathy in man. Report of twelve necropsy cases. J Neurol Sci 14:389-408, 1971 Kristensson K, Molin B, Sourander P: Delayed radiation lesions of the human spinal cord. Report of five cases. Acta Neuropathol 9:34- 44, 1967 Palmer JJ: Radiation myelopathy. Brain 95:109-122, 1972 Reagan TJ, Thomas JE, Colby MY Jr: Chronic progressive radiation myelopathy. Its clinical aspects and differential diagnosis. JAMA 203:128-132, 1968 Schultheiss TE, Stephens LC, Maor MH: Analysis of the histopathology of radiation myelopathy. Int J Radiat Oncol Biol Phys 14:27-32, 1988 Schultheiss TE, Kun LE, Ang KK, et al: Radiation response of the central nervous system. Int J Radiat Oncol Biol Phys 31:1093-1112, 1995 Keirstead HS, Blakemore WF: The role of oligodendrocytes and oligodendrocyte progenitors in CNS remyelination. Adv Exp Med Biol 468:183-197, 1999 Jeffery ND, Blakemore WF: Locomotor deficits induced by experimental spinal cord demyelination are abolished by spontaneous remyelination. Brain 120:27-37, 1997 Yamamoto S, Nagao M, Sugimori M, et al: Transcription factor expression and Notch-dependent regulation of neural progenitors in the adult rat spinal cord. J Neurosci 21:9814-9823, 2001 Yamamoto S, Yamamoto N, Kitamura T, et al: Proliferation of parenchymal neural progenitors in response to injury in the adult rat spinal cord. Exp Neurol 172:115-127, 2001 Sasaki M, Honmou O, Akiyama Y, et al: Transplantation of an acutely isolated bone marrow fraction repairs demyelinated adult rat spinal cord axons. Glia 35:26-34, 2001 Ishii K, Toda M, Nakai Y, et al:Increase of oligodendrocyte progenitor cells after spinal cord injury. J Neurosci Res 65:500-507, 2001 Sanyal B, Pant GC, Subrahmaniyam K, et al: Radiation myelopathy. J Neurol Neurosurg Psy 42:413-418, 1979 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 42 EP 46 PG 5 ER PT J AU , E Celikel, C Gulluoglu, B AF , Eren Celikel, Cigdem Gulluoglu, Bahadir TI Neuroendocrine Differentiation in Gastric Adenocarcinomas; Correlation with Tumor Stage and Expression of VEGF and p53 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric adenocarcinoma; neuroendocrine; p53; VEGF ID Gastric adenocarcinoma; neuroendocrine; p53; VEGF AB Studies on neuroendocrine differentiation (NED) in conventional gastric adenocarcinomas and its significance on tumor behavior are limited. Our aim was to search for the expression of neuroendocrine differentiation in conventional gastric adenocarcinomas and correlate it with tumor type, stage and expression of VEGF and p53. Forty-two gastrectomy specimens with gastric adenocarcinoma were stained with chromogranin A to detect neuroendocrine differentiation and 45% of the cases were found to be NED (+). No significant correlation was found between NED and tumor type. However, NED was more frequent in advanced stage cases independently of tumor type. VEGF expression was also considerably more frequent in NED (+) tumors compared to NED (-) ones (84% vs. 56%). Moreover, we found a significant correlation between NED and the presence of lymph node metastases. P53 expression in NED (+) tumors was 68%. There was no significant correlation between VEGF and p53 in NED (+) cases. In conclusion, neuroendocrine differentiation is a frequent finding in conventional gastric adenocarcinomas, and although it does not seem to play a specific role in tumor progression, it seems that neuroendocrine cells are one of the factors contributing to angiogenesis by expressing VEGF, especially in advanced stage cases, affecting the incidence of lymph node metastases. Further studies with larger series should be performed to confirm this observation. C1 [, Eren] Marmara University, School of Medicine, Department of Pathology, Bagdat Cad. 277-1 Dostlar apt. Daire 2, Caddebostan, 34728 Istanbul, Turkey. [Celikel, Cigdem] Marmara University, School of Medicine, Department of Pathology, Bagdat Cad. 277-1 Dostlar apt. Daire 2, Caddebostan, 34728 Istanbul, Turkey. [Gulluoglu, Bahadir] Marmara University, School of Medicine, Department of General SurgeryIstanbul, Turkey. RP , E (reprint author), Marmara University, School of Medicine, Department of Pathology, 34728 Istanbul, Turkey. EM fyeren@superonline.com CR Allen FJ, Van Velden DJ, Heyns CF: Are neuroendocrine cells of practical value as an independent prognostic parameter in prostate cancer? Br J Urol 75: 751-754, 1995 Blumenfeld W, Chandhoke D, Jageman P: Neuroendocrine differentiation in gastric adenocarcinomas. An immunohistochemical study. Arch Pathol Lab Med 120: 478-481, 1996 Bostwick DG, Qion J, Pacelli A, et al: Neuroendocrine expression in node positive prostate cancer: Correlation with systemic progression and survival. J Urol 168: 1204-1211, 2002 Hamilton K, Chiappori A, Olson S, et al: Prevalence and prognostic significance of neuroendocrine cells in esophageal adenocarcinoma. Mod Pathol 13: 475-481, 2000 Helge L, Waldum, Brenna E, Sandvik A: Relationship of ECL cells and gastric neoplasia. Yale J Biol Med 71: 325-335, 1998 Hiroshima K, Iyoda A, Shibuya K, et al: Prognostic significance of neuroendocrine differentiation in adenocarcinoma of the lung. Ann Thorac Surg 73: 1732-1735, 2002 Indinnimeo M, Cichini C, Memeo L, et al: Correlation between chromogranin A expression and pathological variables in human colon carcinoma. Anticancer Res 22: 395-398, 2002 Kyokane K, Ito M, Sato Y, et al: Expression of bcl-2 and p53 correlates with the morphology of gastric neoplasia. J Pathol 184: 382-389, 1998 Maehara Y, Kabashima A, Koga T, et al: Vascular invasion and potential for tumor angiogenesis and metastasis in gastric carcinoma. Surgery 128: 408-416, 2000 Maeda K, Kong SM, Onoda N, Ogawa M: Expression of p53 and vascular endothelial growth factor associated with tumor angiogenesis and prognosis in gastric adenocarcinoma. Oncology 55: 594-599, 1998 Miremadi A, Pinder SE, Lee AH, et al: Neuroendocrine differentiation and prognosis in breast carcinoma. Histopathology 40: 211-214, 2002 Ooi A, Hayashi H, Katsuda S, et al: Gastric adenocarcinoma cells with endocrine differentiation show no evidence of proliferation. Hum Pathol 23: 736-741, 1992 Ooi A, Mai M, Ogino T, et al: Endocrine differentiation of gastric carcinoma. The prevalence as evaluated by immunoreactive chromogranin A and its biologic significance. Cancer 62: 1096-1104, 1998 Park JG, Choe GY, Helmon LJ, et al: Chromogranin-A expression in gastric and colon cancer tissues. Int J Cancer 51: 189- 194, 1992 Qvigstad G, Arne K, Sandvik A, et al: Detection of chromogranin A in human gastric adenocarcinomas using a sensitive immunohistochemical technique. Histochemical J 32: 551-556, 2000 Saito H, Tujitani S, Ikeguchi M, et al: Neoangiogenesis and relationship to nuclear p53 accumulation and vascular endothelial growth factor expression in advanced gastric carcinoma. Oncology 57: 164-172, 1999 Waldum HL, Brenna E, Sandvik A: Relationship of ECL cells and gastric neoplasia. Yale J Biol Med 71: 325-335, 1998 Waldum HL, Aase S, Kuetnoi I: Neuroendocrine differentiation in human gastric carcinoma. Cancer 83: 435-444, 1998 Wu MS, Shen OJ, Sheu JC: Overexpression of mutant p53 and C-erbB-2 proteins and mutations of the p15 and p16 genes in human gastric carcinoma. J Gastroenterol Hepatol 13: 305- 310, 199 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 47 EP 51 PG 5 ER PT J AU Pal, J Marczinovits, I Hudecz, F Toth, KG Mezo, G Molnar, J Nemeth, P AF Pal, Jozsef Marczinovits, Ilona Hudecz, Ferenc Toth, K Gabor Mezo, Gabor Molnar, Janos Nemeth, Peter TI Modeling of Main Characteristics of Bullous Pemphigoid Antigen-2 (BPAG2) Peptide Structure in Serological Recognition by Autoantibodies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE carrier effect; BPAG2 epitope; autoantibodies; immunological recognition; cross-reaction ID carrier effect; BPAG2 epitope; autoantibodies; immunological recognition; cross-reaction AB The serum level of autoantibodies against autoantigens of the bullous pemphigoid peptides 1 and 2 (BPAG1 and BPAG2) is a relevant diagnostic marker. Twelve representative sera of BP were tested against the RSILPYGDSMDRIEKDRLQMAP amino acid sequence that is an epitope fragment of the NC16A domain of BPAG2 (AC Q02802; 507-528) to find the most suitable antigenic form for specific detection of autoantibodies of BP patients’ sera by quantitative ELISA system. The antigenic epitope sequence was presented as an antigen in a carrier free form of dimeric peptide (BP22), dimeric peptide fused to glutathione S-transferase (GST-BP22) or dimeric peptide chemically conjugated to polyLys(Ser-DL-Alam) (SAK-BP22). The intensity of ELISA reaction was highest against the recombinant fusion antigen GSTBP22; the chemically conjugated SAK-BP22 performed less well than the free dimeric form of the peptide. In the case of the GST-BP22 antigen, the (GST-BP22)-(GST)492nm optical density values were determined. There was no significant difference between the mean ODs of the GST-BP22 and the SAK-BP22 (0.888 vs. 0.892, p= 0.9726). Conjugating the epitope peptide with the synthetic carrier SAK was advantageous, as it abrogated cross-reactivity with GST carrier protein. Consequently, the SAKBP22 conjugate appears to be the most reliable assay component, avoiding cross-reactivity with GST and simplifying the detection and evaluation of BP autoantibodies in routine ELISA diagnostic system. C1 [Pal, Jozsef] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u 12., H-7643 Pecs, Hungary. [Marczinovits, Ilona] University of Szeged, Department of PhysiologySzeged, Hungary. [Hudecz, Ferenc] Eotvos Lorand University, Hungarian Academy of Sciences, MTA-ELTE Research Group of Peptide ChemistryBudapest, Hungary. [Toth, K Gabor] University of Szeged, Department of Medical ChemistrySzeged, Hungary. [Mezo, Gabor] Eotvos Lorand University, Hungarian Academy of Sciences, MTA-ELTE Research Group of Peptide ChemistryBudapest, Hungary. [Molnar, Janos] University of Szeged, Department of PhysiologySzeged, Hungary. [Nemeth, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u 12., H-7643 Pecs, Hungary. RP Nemeth, P (reprint author), University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, H-7643 Pecs, Hungary. EM peter.nemeth@aok.pte.hu CR Dopp R, Schmidt E, Chimanovitch I et al: IgG4 and IgE are the major immunoglobulins targeting the NC16A domain of BP180 in bullous pemphigoid: Serum levels of these immunoglobulins reflect disease activity. J Am Acad Dermatol 42:577-583, 2000 Hancock DC, O’Relly NJ, Evan GI: Synthetic peptides in biochemical research. Mol Biotechnol 4:73-86, 1995 Hudecz F, Pimm MV, Rajnavolgyi E et al: Carrier design: New generation of polycationic branched polypeptides containing OH groups with prolonged blood survival and diminished in vitro cytotoxicity. Bioconjug Chem 10:781-790, 1999 Hudecz F: Manipulation of epitope function by modification of peptide structure. A minireview. Biologicals 29:197-207, 2001 Husz S, Kiss M, Molnar K et al: Development of a system for detection of circulating antibodies against hemidesmosomal proteins in patients with bullous pemphigoid. Arch Dermatol Res 292:217-224, 2000 Laczko I, Vass E, Toth G K et al: Conformational consequences of coupling bullous pemphigoid antigenic peptides to glutathione- S-transferase and their diagnostic significance. J Pept Sci 6:378-386, 200. Marczinovits I, Somogyi C, Patthy A et al: An alternative purification protocol for producing hepatitis B virus X antigen on a preparative scale in Escherichia coli. J Biotechnol 56:81- 88, 1997 McCauliffe DP, Yin H, Wang LX, Lucas L: Autoimmune sera react with multiple epitopes on recombinant 52 and 60 kDa Ro(SSA, proteins. J Rheumatol 21:1073-1080, 1994 Molnar J, Marczinovits I, Kiss M et al: Recombinant antigens by fusion of antigenic epitopes to a GST partner. In: Stability and Stabilization of Biocatalysts, Eds: Ballesteros A, Plou FJ, Iborra JL and Halling PJ). Prog Biotechnol 15:691-696, 1998 Nakatani C, Muramatsu T and Shirai T: Immunoreactivity of bullous pemphigoid, BP, autoantibodies against the NC16A and Cterminal domains of the 180 kDa BP antigen, BP180): immunoblot analysis and enzyme-linked immunosorbent assay using BP180 recombinant proteins. Br J Dermatol 139:365-370, 1998 Zillikens D, Mascaro JM, Rose PA et al: A highly sensitive enzyme-linked immunosorbent assay for the detection of circulating anti-BP180 autoantibodies in patients with bullous pemphigoid. J Invest Dermatol 109:679-683, 1997 Zillikens D, Rose P A, Balding S D et al: Tight clustering of extracellular BP180 epitopes recognized by bullous pemphigoid autoantibodies. J Invest Dermatol 109:573-579, 1997 Wesierska-Gadek J, Lindner H, Hitchman E et al: Anti-SLA seropositive autoimmune hepatitis sera recognize distinct subunits of glutathione S-transferase: high prevalence of the Ya autoantigen. Cell Mol Biol 48:301-307, 2002 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 52 EP 56 PG 5 ER PT J AU Brittig, F Ajtay, E Jakso, P Kelenyi, G AF Brittig, Ferenc Ajtay, Elvira Jakso, Pal Kelenyi, Gabor TI Follicular Dendritic Reticulum Cell Tumor Mimicking Inflammatory Pseudotumor of the Spleen SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Follicular dendritic reticulum cells ID Follicular dendritic reticulum cells AB In the course of a routine clinical check up of the 54 year old male a splenic well circumscribed tumor like mass of 12 cm in diameter was discovered. Splenectomy with removal of splenic hilar lymph nodes and liver wedge biopsy were performed. Four years later the patient is symptom free. In the removed spleen the tumor like lesion showed a pattern consistent with the diagnosis of inflammatory pseudotumor. However, besides lymphocytes, plasma cells, macrophages, eosinophils and myofibroblasts a high number of slightly polymorphic, frequently binucleated cells positive for CD21 and CD23 were seen. These cells which were scattered or formed smaller or larger groups and fascicles were considered to represent follicular dendritic reticulum cells (FDRCs) and the lesion a FDRC tumor. Flow cytometric DNA ploidy analysis showed a hyperdiploid cell population inside the tumor like lesion. Besides FDRC tumors of high and of intermediate malignancy the present case may represent a low grade type of moderate proliferation activity. The FDRCs of the lesion and a few smaller spindle cells were EBER positive indicative of the presence of EBV. No EBER positive cells were seen in the uninvolved spleen. C1 [Brittig, Ferenc] County Hospital, Department of PathologyVeszprem, Hungary. [Ajtay, Elvira] County Hospital, Department of Internal MedicineVeszprem, Hungary. [Jakso, Pal] University of Pecs, Department of Pathology, Szigeti ut 12., H-7624 Pecs, Hungary. [Kelenyi, Gabor] University of Pecs, Department of Pathology, Szigeti ut 12., H-7624 Pecs, Hungary. RP Kelenyi, G (reprint author), University of Pecs, Department of Pathology, H-7624 Pecs, Hungary. CR Al-Nafussi A, Wong NACS: Intraabdominal spindle cell lesions: a review and practical aids to diagnosis. Histopath 38: 387-402, 2001 Andriko J-A, Kaldijian EP, Tsokos M et al: Reticulum cell neoplasms of lymph nodes. Am J Surg Pathol 22: 1048-1058, 1998 Arber DA, Weiss LM, Chang KL: Detection of Epstein-Barr virus in inflammatory pseudotumor. Semin Diagn Pathol 15: 155-160, 1998 Bajema IM and Brujin JA: What stuff is this! A historical perspective on fibrinoid necrosis. J Pathol 191: 235-238, 2000 Biselli R, Boldrini R, Ferlini C, et al: Myofibroblastic tumors: neoplasias with divergent behaviour, ultrastructural and flow cytometric analysis. Pathol Res Pract 195: 619-632, 1999 Biselli R, Ferlini C, Fattorossi A, et al.: Inflammatory myofibroblastic tumor, inflammatory pseudotumor): DNA flow cytometric analysis of nine pediatric cases. Cancer 77: 778-784, 1996 Bofill M, Akbar AN and Amiot PL: Follicular dendritic cells share a membrane-bound protein with fibroblats. J Pathol 191: 217-226, 2000 Chan JKC, Tsang WYW, Ng CS: Follicular dendritic cell tumor and vascular neoplasm complicating hyaline-vascular Castleman’s disease. Am J Surg Pathol 18: 517-525, 1994 Chan JKC, Fletcher CDM, Nayler SJ et al: Follicular dendritic cell sarcoma. Clinicopathologic analysis of 17 cases suggesting malignant potential higher than currently recognized. Cancer 79: 294-313, 1997 Cheuk W, Woo PCY, Yuen KY, et al: Intestinal inflammatory pseudotumor with regional lymph node involvement: identification of a new bacterium as the aetiological agent. J Pathol 192: 289-292, 2000 Cheuk W, Chan JKC, Shek TWH, Chang JH, et al: Inflammatory pseudotumor-like follicular dendritic cell tumor. Am J Surg Pathol 25: 721-731, 2000 Coffin CM, Humprey PA, Dehner LP: Extrapulmonary inflammatory myofibroblastic tumor: a clinical and pathological review. Semin Diagn Pathol 15: 85-101, 1998 Coffin CM, Watterson J, Priest JR, et al: Extrapulmonary inflammatory myofibroblastic tumor, inflammatory pseudotumor): a clinicopathologic and immunohistochemical study of 84 cases. Am J Surg Pathol 19: 859-872, 1995 Dehner LP: The enigmatic inflammatory pseudotumors: the current state of our understanding, or misunderstanding. J Pathol 192: 277- 279. 2000 Dominis M, Dzebro S, Kusic B, et al: Inflammatory pseudotumor of the spleen. Acta Cytol 42:1053-1056, 1998 Gomez-Roman JJ, Sanchez-Velasco P, Ocejo-Vinyals G, et al: Human herpesvirus-8 genes are expressed in pulmonary inflammatory myofibroblastic tumor, inflammatory pseudotumor). Am J Surg Pathol 25: 624-629, 2001 Jones D, Jorgensen JL, Shahsafaci A, Dorfman DM: Characteristic proliferations of reticular and dendritic cells in angioimmunoblastic lymphoma. Am J Surg Pathol 22: 956-964, 1998 KalmanE, Kelenyi G, Pajor L: Flow cytometric analysis of the DNA conent of formol fixed malignant lymphoma tissues. Blut 61: 333, 1990 Lawrence B, Perez-Alayde A, Hibbard MK et al.:TPM3- ALK and TPM4-ALK oncogenes in inflammatory myofibroblastic tumors. Am J Pathol 157: 377-384,2000 Leung CY, Ho FCS, Srivastava G, Loke SL et al: Usefulness of follicular dendritic cell pattern in classification of peripheral T cell lymphomas. Histopathol 23: 433-437, 1993 Menke DM, Griesser H, Araujo I, et al: Inflammatory pseudotumors of lymph node origin show macrophage-derived spindle cells and lymphocyte derived cytokine transcripts without evidence of T-cell receptor gene rearrangements. Am J Clin Pathol 105: 430-439, 1996 Moran CA, Suster S, Abondanzo SL: Inflammatory pseudotumor of lymph nodes: a study of 25 cases with emphasis on morphological heterogeneity. Hum. Pathol 28: 332-338, 1997 Noguchi H, Kondo H, Shiraiwas M, et al.: Inflammatory pseudotumor of the spleen: a case report. Jpn J Clin Oncol 30: 190-203, 2000 Overton WR, Catalano E and McCoy P: Method to make paraffinembedded breast and lymph node tissue mimic fresh tissue in DNA analysis. Cytometry 26: 166-171, 1996 Pileri SA, Grogan TM, Harris NL, et al: Tumors of histiocytes and accessory dendritic cells: an immunohistochemical approach to classifcation from the International Lymphoma Study Group based on 61 cases. Histopathology 41: 1-29, 2000 Radhi J, Hadjis N, Anderson L, et al: Retroperitoneal actinomycosis masquerading as inflammatory pseudotumor. J Pediatr Surg 32: 618- 620, 1997 Raymond I, Al Salati T, Tkaczuk J, et al: CNA.42, a new monoclonal antibody directed against a fixative-resistant antigen of follicular dendritic reticulum cells. Am J Pathol 151: 1577-1585, 1997 Sciot R, Dal Cin P, Fletcher CDM, et al: Inflammatory myofibroblastic tumor of the bone. Report of two cases with evidence of clonal chromosomal changes. Am J Surg Pathol 21: 1166-1172, 1999 Selves J, Meggetto F, Brousset P, et al: Inflammatory pseudotumor of the liver. Evidence for follicular dendritic reticulum cell proliferation associated with clonal Epstein-Barr virus. Am J Surg Pathol 20: 747- 753, 1996 Shek TWH, Ho FCS, Ng IOL, et al: Follicular dendritic reticulum cell tumor of the liver. Evidence of an Epstein-Barr virus-related clonal prliferation of follicular dendritic cells. Am J Surg Pathol 20: 313- 324, 1996 Snyder CS, Dell’Agulia M, Haghighi P, et al: Clonal changes in inflammatory pseudotumor of the lung. A case report. Cancer 76: 1545-1549, 1995 Su L, Sheldon S, Weiss SW: Inflammatory myofibroblastic tumor. Cytogenetic evidence supporting clonal origin. Mod Pathol 8: 12A,1995 Tornocky T, Kelenyi G, Pajor L: EBER oligonucleotide RNA in situ hybridisation in EBV associated neoplasms. Path Oncol Res 3: 201- 205,1998 Trainor KJ, Brisco, MJ, Wan JH, et al: Gene rearrangement in B- and T-lymphoproliferative disease detected by the polymerase chain reaction. Blood 78: 192-196, 1991 Vaideeswar P, Madiwale CV, Desai AP, et al: Inflammatory pseudotumor of the lymph node in an HIV-positive individual. Histopathology 36: 374-375, 2000 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2004 VL 10 IS 1 BP 57 EP 60 PG 4 ER PT J AU Kopper, L Hajdu, M AF Kopper, Laszlo Hajdu, Melinda TI Tumor Stem Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE stem cell; tumor ID stem cell; tumor AB Stem cells possess two basic characteristics: they are able to renew themselves and to develop into different cell types. The link between normal stem cells and tumor cells could be examined in three aspects: what are the differences and similarities in the control of self-renewal capacity between stem cells and tumor cells; whether tumor cells arise from stem cells; do tumorous stem cells exist? Since tumor cells also exhibit self-renewal capacity, it seems plausible that their regulation is similar to that of the stem cells. The infinite self-renewal ability (immortalization) is assured by several, so far only partly known, mechanisms. One of these is telomerase activity, another important regulatory step for survival is the inhibition of apoptosis. Other signal transduction pathways in stem cell regulation may also play certain roles in carcinogenesis: e.g. Notch, Sonic hedgehog (SHH), and Wnt signals. Existence of tumor stem cells was suggested since it is simpler to retain the self-renewal capacity than to reactivate the immortality program in an already differentiated cell. Moreover, stem cells live much longer than the differentiated ones, and so they are exposed for a long period of time to impairments, collecting gene errors leading to the breakdown of the regulation. However, it is still an open question whether all cells in the tumor possess the capacity that produces this tissue or not, that is: are there tumor stem cells or there are not. If tumor stem cells exist, they would be the main target for therapy: only these must be killed since the other tumor cells possess limited proliferative capacity, therefore limited life span. The only problem is that during tumor progression stem-like cells can develop continuously and the identification but mainly the prevention of their formation is still a great challenge. C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Hajdu, Melinda] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular PathologyBudapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM kopper@korb1.sote.hu CR Thomson JA, Ilskovitz-Eldor M, Shapiro SS, et al: Embryonic stem cell lines derived from human blastocysts. Science 282: 1145-1147, 1998 Lagasse E, Connors H, Al-Dhalimy M, et al: Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat Med 6: 1229-1234, 2000 Ferrari G, Cusella-De Angelis G, Coletta M, et al: Muscle regeneration by bone marrow-derived myogenic progenitor. Science 279: 1528-1530, 1998 Alison MR, Poulsom R, Jeffery R, et al: Hepatocytes from nonhepatic adult stem cells. Nature 406: 257, 2000 Eglitis MA, Mezey E: Hematopoietic cells differentiate into both microglia and macroglia in the brains of adult mice. Proc Natl Acad Sci USA 94: 4080-4085, 1997 Orlic D, Kajsturo J, Chimenti S, et al: Bone marrow cells regenerate infarcted myocardium. Nature 410: 701-704, 2001 Poulsom R, Forbes SJ, Hodivala-Dilke K, et al: Bone marrow contributes to renal parenchymal turnover and regeneration. J Pathol 195: 229-235, 2001 Preston SL, Alison MR, Forbes SJ, et al: The new stem cell biology: something for everyone. J Clin Pathol: Mol Pathol 56: 86-96, 2003 Trosko JE, Chang CC: Stem cell theory of carcinogenesis. Toxicol Lett 49: 283, 1989 Domen J, Gandy KL, Weissmann IL: Systemic overexpression of BCL2 in the hematopoetic system protects transgenic mice from the consequences of lethal irradiation. Blood 91: 2272- 2282, 1998 Domen J, Weissman IL: Hematopoietic stem cellss need two signals to prevent apoptosis; BCL-2 can provide one of these, Kit/c-Kit signaling the other. J Exp Med 192: 1707-1718, 2000 Reya T, Morrison RJ, Clarke MF, Weissman IL:Stem cells, cancer, and cancer stem cells. Nature 414: 105-111, 2001 Taipale J, Beachy PA: The Hedgehog and Wnt signaling pathways in cancer. Nature 411: 349-354, 2001 Varnum-Finney B, et al: Pluripotent, cytokine-dependent, hematopoetic stem cells are immortalized by constitutive Notch signaling. Nat Med 6: 1278-1281, 2000 Karanu FN, et al: The Notch ligand Jagged-1 represents a novel growth factor of human hematopoetic stem cells. J Exp Med 192: 1365-1372, 2000 Spradling A, Drummond-Barbosa D, Kai T. Stem cells find their niche. Nature 414: 98-104, 2001 Dick JE. Self-renewal writ in blood. Nature, 423: 231, 2003 Lessard J, Sauvageau G: Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature 423: 255- 260, 2003 Gat U, DasGupta R, Deegenstein I, et al: De novo hair follicle morphogenesis and hair tumor sin mice expressing a truncated beta-catenin in skin. Cell 95: 605-614, 1998 Kojika S, Griffin JD: Notch receptors and hematopoiesis. Exp Hematol 29: 1041, 2001 Artavanis-Tsakonas S, Rand MD, Lake RJ: Notch Signaling: Cell Fate Control and Signal Integration in Development. Science 284: 770-776, 1999 Hitoshi S, Alexson T, Tropepe V, Donoviel D et al. Notch pathway molecules are essential for the maintenance, but no the generation, of mammalian neural stem cells. Genes Dev 16: 846, 2002 Ingham PW:. Trancducing Hedgehog: the story so far. EMBO J 17: 3505, 1998 Bhardwaj G, Murdoch B, Wu D, Baker DP, et al: Sonic hedgehog induces the proliferation of primitive human hematopoietic cells via BMP regulation. Nat Immunol 2: 172, 2001 Nowell PC: The clonal evolution of tumor cell population. Science 194: 23-28, 1976 Miyamoto T, Weissman IL, Akashi K: AML1/ETO expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc Natl Acad Sci USA 97: 7521-7526, 2000 Traver D, Akashi K, Weissman IL, Lagasse E: Mice defective in two apoptosis pathways in the myeloid lineage develop acute myeloblastic leukemia. Immunity 9: 47-57, 1998 Al-Hajj M, Wicha MS, Benito-Hernandez A, et al: Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 100: 3983-3988, 2003 Dick JE. Breast cancer stem cells revealed. Proc Natl Acad Sci USA 100: 3547-3549, 2003 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2004 VL 10 IS 2 BP 69 EP 73 PG 5 ER PT J AU Jin, R Huang, J Tan, PH Bay, BH AF Jin, Rongxian Huang, Jingxiang Tan, Puay-Hoon Bay, Boon-Huat TI Clinicopathological Significance of Metallothioneins in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE MT isoforms; biochemistry; biomarker; prognosis; chemoresistance; carcinogenesis ID MT isoforms; biochemistry; biomarker; prognosis; chemoresistance; carcinogenesis AB Metallothioneins (MTs) are a family of metal binding proteins that play an important role in maintaining transition metal ion homoeostasis, redox balance in the cell and fundamental cellular processes such as proliferation and apoptosis. In humans, there are 4 groups of MT proteins which are encoded by 10 functional MT isoforms. In breast tissues, MT is primarily expressed in myoepithelial and malignant epithelial cells. Immunohistochemical studies have revealed that 26% to 100% of invasive ductal breast cancers express the MT protein. The MT-1F and MT-2A isoforms have been reported to be associated with higher histological grade in breast cancer, whereas higher MT-1E mRNA expression was found in estrogen receptor-negative tumors compared to their estrogen receptor-positive counterparts. A number of studies have shown that MT expression in breast cancer is associated with poorer prognosis. In addition, metallothionein expression may have a potential role in protecting the breast cancer cell from chemotherapeutic threats to survival. C1 [Jin, Rongxian] National University of Singapore, Department of Anatomy, 4 Medical Drive, Block MD10, S 117 597 Singapore, Singapore. [Huang, Jingxiang] National University of Singapore, Department of Anatomy, 4 Medical Drive, Block MD10, S 117 597 Singapore, Singapore. [Tan, Puay-Hoon] National University Hospital, Department of PathologySingapore, Singapore. [Bay, Boon-Huat] National University of Singapore, Department of Anatomy, 4 Medical Drive, Block MD10, S 117 597 Singapore, Singapore. RP Bay, BH (reprint author), National University of Singapore, Department of Anatomy, S 117 597 Singapore, Singapore. EM antbaybh@nus.edu.sg CR Kojima Y, Binz P-A, Kagi JHR: Nomenclature of metallothionein: proposal for a revision. In: Klaassen, C.D. eds. Metallothionein IV, Birkhauser Verlag, Basel, Switzerland. pp 3-6, 1999. Pande J, Vasak M, Kagi JH: Interaction of lysine residues with the metal thiolate clusters in metallothionein. Biochemistry 24: 6717-6722, 1985. Robbins AH, McRee DE, Williamson M, et al.: Refined crystal structure of Cd, Zn metallothionein at 2.0A resolution. J Mol Biol 221: 1269-1293, 1991. Binz P-A, Kagi JHR: Metallothionein: molecular evolution and classification. In: Klaassen, C.D. eds. Metallothionein IV, Birkhauser Verlag, Basel, Switzerland. pp 7-13, 1999. West AK, Stallings R, Hildebrand CE, et al.: Human metallothionein genes: structure of the functional locus at 16q13. Genomics 8: 513-518, 1990. Stennard FA, Holloway AF, Hamilton J, et al.: Characterisation of six additional human metallothionein genes. Biochim Biophys Acta 1218: 357-365, 1994. Mididoddi S, McGuirt JP, Sens MA, et al.: Isoform-specific expression of metallothionein mRNA in the developing and adult human kidney. Toxicol Lett 85:17-27, 1996. Palmiter RD, Findley SD, Whitmore TE, et al.: MT-III, a brainspecific member of the metallothionein gene family. Proc Natl Acad Sci USA 89: 6333-6337, 1992. Quaife CJ, Findley SD, Erickson JC, et al.: Induction of a new metallothionein isoform, MT-IV, occurs during differentiation of stratified squamous epithelia. Biochemistry 33: 7250-7259, 1994. Kagi JH, Hunziker P. Mammalian metallothionein. Biol Trace Elem Res 21: 111-118, 1989. Braun W, Wagner G, Worgotter E, et al.: Polypeptide fold in the two metal clusters of metallothionein-2 by nuclear magnetic resonance in solution. J Mol Biol 187: 125-129, 1986 Schultze P, Worgotter E, Braun W, et al.: Conformation of [Cd7]-metallothionein-2 from rat liver in aqueous solution determined by nuclear magnetic resonance spectroscopy. J Mol Biol 203: 251-268, 1988 Braun W, Vasak M, Robbins AH et al.: Comparison of the NMR solution structure and the X-ray crystal structure of rat metallothionein- 2. Proc Natl Acad Sci USA 89: 10124-10128, 1992. Tapiero H, Tew KD: Trace elements in human physiology and pathology: zinc and metallothioneins. Biomed Pharmacother 57: 399-411, 2003. Roesijadi G. Metal transfer as a mechanism for metallothionein- mediated metal detoxification. Cell Mol Biol, Noisy-legrand, 46: 393-405, 2000. Kagi JH, Kojima Y. Chemistry and biochemistry of metallothionein. Experientia 52: 25-61, 1987. Kondoh M, Imada N, Kamada K, et al.: Property of metallothionein as a Zn pool differs depending on the induced condition of metallothionein. Toxicol Lett 142: 11-18, 2003. Maret W: Metallothionein/disulfide interactions, oxidative stress, and mobilization of cellular zinc. Neurochem Int 27: 111-117, 1995. Maret W, Vallee BL: Thiolate ligands in metallothionein confer redox activity on zinc clusters. Proc Natl Acad Sci USA 95: 3478-3482, 1998. Miura T, Muraoka S, Ogiso T: Antioxidant activity of metallothionein compared with reduced glutathione. Life Sci 60: 301-309, 1997. Hussain S, Slikken JW, Ali SF. Role of metallothionein and the antioxidants in scavenging superoxide radicals and their possible role in neuroprotection. Neurol Chem Int 29: 145–152, 1996. Abel J, Ruiter N: Inhibition of hydroxy radical generated DNA degradation by metallothionein. Toxicol Lett 47: 191-196, 1989. Cai L, Klein JB, Kang YJ: Metallothionein inhibits peroxynitrite- induced DNA and lipoprotein damage. J Biol Chem 275: 38957-38960, 2000. Mendez-Armenta M, Villeda-Hernandez J, Barroso-Moguel R, et al.: Brain regional lipid peroxidation and metallothionein levels of developing rats exposed to cadmium and dexamethasone. Toxicol Lett 144: 151-157, 2003. Garrett SH, Sens MA, Shukla D,et al.: Metallothionein isoform 1 and 2 gene expression in the human prostate: downregulation of MT-1X in advanced prostate cancer. Prostate 43: 125-135, 2000. Tai SK, Tan OJ, Chow VT, et al.: Differential expression of metallothionein 1 and 2 isoforms in breast cancer lines with different invasive potential: identification of a novel nonsilent metallothionein-1H mutant variant. Am J Pathol 163: 2009- 2019, 2003. Haq F, Mahoney M, Koropatnick J: Signaling events for metallothionein induction. Mutat Res 533: 211-226, 2003. Garrett SH, Somji S, Todd JH, et al.: Differential expression of human metallothionein isoform I mRNA in human proximal tubule cells exposed to metals. Environ Health Perspect 106: 825-832, 1998. Garrett SH, Belcastro M, Sens MA, et al.: Acute exposure to arsenite induces metallothionein isoform-specific gene expression in human proximal tubule cells. J Toxicol Environ Health A 64: 343-355, 2001. Jahroudi N, Foster R, Price-Haughey J, et al.: Cell-type specific and differential regulation of the human metallothionein genes. Correlation with DNA methylation and chromatin structure. J Biol Chem 265: 6506-6511, 1990. Barnes NL, Ackland ML, Cornish EJ: Metallothionein isoform expression by breast cancer cells. Int J Biochem Cell Biol 32: 895-903, 2000. Fresno M, Wu W, Rodriguez JM, et al.: Localization of metallothionein in breast carcinomas. An immunohistochemical study. Virchows Arch A Pathol Anat Histopathol 423: 215-219, 1993. Jin R, Bay BH, Chow VT, et al.: Significance of metallothionein expression in breast myoepithelial cells. Cell Tissue Res 303: 221-226, 2001. Bier B, Douglas-Jones A, Totsch M, et al.: Immunohistochemical demonstration of metallothionein in normal human breast tissue and benign and malignant breast lesions. Breast Cancer Res Treat 30: 213-221, 1994. Schmid KW, Ellis IO, Gee JM, et al.: Presence and possible significance of immunocytochemically demonstrable metallothionein over-expression in primary invasive ductal carcinoma of the breast. Virchows Arch A Pathol Anat Histopathol 422: 153-159, 1993. Zhang R, Zhang H, Wei H, et al.:Expression of metallothionein in invasive ductal breast cancer in relation to prognosis. J Environ Pathol Toxicol Oncol 19: 95-97, 2000. Vazquez-Ramirez FJ, Gonzalez-Campora JJ, Hevia-Alvarez E, et al.: P-glycoprotein, metallothionein and NM23 protein expressions in breast carcinoma. Pathol Res Pract 196: 553- 559, 2000. Jin R, Chow VT, Tan PH, et al.: Metallothionein 2A expression is associated with cell proliferation in breast cancer. Carcinogenesis 23: 81-86, 2002. Ioachim E, Tsanou E, Briasoulis E, et al.: Clinicopathological study of the expression of hsp27, pS2, cathepsin D and metallothionein in primary invasive breast cancer. Breast 12: 111- 119, 2003. Jin R, Bay BH, Chow VTK, et al.: Metallothionein 1F mRNA expression correlates with histological grade in breast carcinoma. Breast Caner Res Treat 66: 265-272, 2001. Sens MA, Somji S, Garrett SH, et al.: Metallothionein isoform 3 overexpression is associated with breast cancers having a poor prognosis. Am J Pathol 159: 21-26, 2001. Nartey N, Cherian MG, Banerjee D: Immunohistochemical localization of metallothionein in human thyroid tumors. Am J Pathol 129: 177-182, 1987 Cherian MG, Huang PC, Klaassen CD, et al.: National Cancer Institute workshop on the possible roles of metallothionein in carcinogenesis. Cancer Res 53: 922-925, 1993 Cherian MG, Howell SB, Imura N, et al.: Role of metallothionein in carcinogenesis. Toxicol Appl Pharmacol 126: 1-5, 1994 Fan LZ, Cherian MG. Potential role of p53 on metallothionein induction in human epithelial breast cancer cells. Br J Cancer 87: 1019-1026, 2002. Kagi JH: Overview of metallothionein. Methods Enzymol. 205: 613-26, 1991. Schwarz MA, Lazo JS, Yalowich JC, et al.: Metallothionein protects against the cytotoxic and DNA-damaging effects of nitric oxide. Proc Natl Acad Sci USA 92: 4452-4456, 1995. Andrews GK, Adamson ED, Gedamu L: The ontogeny of expression of murine metallothionein: comparison with the alpha-fetoprotein gene. Dev Biol 1984; 103: 294-303. Nagel WW, Vallee BL: Cell cycle regulation of metallothionein in human colonic cancer cells. Proc Natl Acad Sci USA 92:579-583, 1995. Abdel-Mageed A, Agrawal KC: Antisense down-regulation of metallothionein induces growth arrest and apoptosis in human breast carcinoma cells. Cancer Gene Ther 4: 199-207, 1997. Gurel V, Sens DA, Somji S, et al.: Stable transfection and overexpression of metallothionein isoform 3 inhibits the growth of MCF-7 and Hs578T cells but not that of T-47D or MDA-MB- 231 cells. Breast Cancer Res Treat 80: 181-191, 2003. Cai L, Wang GJ, Xu ZL, et al.: Metallothionein and apoptosis in primary human hepatocellular carcinoma from northern China. Anticancer Res 18: 4667-4672, 1998. Jayasurya A, Bay BH, Yap WM, et al.: Correlation of metallothionein expression with apoptosis in nasopharyngeal carcinoma. Br J Cancer 82: 1198-1203, 2000. Huang J, Tan PH, Thiyagarajan J, et al.: Prognostic significance of glutathione S-transferase-pi in invasive breast cancer. Mod Pathol 16: 558-565, 2003. Abdel-Mageed A, Agrawal KC: Activation of nuclear factor kappaB: potential role in metallothionein-mediated mitogenic response. Cancer Res 58: 2335-2338, 1998. Sakurai A, Hara S, Okano N, et al.: Regulatory role of metallothionein in NF-kappaB activation. FEBS Lett 455:55-58, 1999. Butcher HL, Kennette WA, Collins O, et al.: Metallothionein mediates the level and activity of nuclear factor {kappa}B, NFkB, in murine fibroblasts. J Pharmacol Exp Ther 2004 Mar 23 [Epub ahead of print] Kim CH, Kim JH, Lee J, et al.: Zinc-induced NF-kappaB inhibition can be modulated by changes in the intracellular metallothionein level. Toxicol Appl Pharmacol 190: 189-196, 2003. Cui Y, Wang J, Zhang X, et al.: ECRG2, a novel candidate of tumor suppressor gene in the esophageal carcinoma, interacts directly with metallothionein 2A and links to apoptosis. Biochem Biophys Res Commun 302:904-915, 2003. Hainaut P, Mann K: Zinc binding and redox control of p53 structure and function. Antioxid Redox Signal 3: 611-623, 2001. Meplan C, Richard MJ, Hainaut P: Metalloregulation of the tumor suppressor protein p53: zinc mediates the renaturation of p53 after exposure to metal chelators in vitro and in intact cells. Oncogene 19: 5227-5236, 2000. Jacob C, Maret W, Vallee BL: Control of zinc transfer between thionein, metallothionein, and zinc proteins. Proc Natl Acad Sci USA 95: 3489-3494, 1998. Haerslev T, Jacobsen GK, Zedeler K: The prognostic significance of immunohistochemically detectable metallothionein in primary breast carcinomas. APMIS 103: 279-285, 1995. Oyama T, Take H, Hikino T, et al.: Immunohistochemical expression of metallothionein in invasive breast cancer in relation to proliferative activity, histology and prognosis. Oncology 53: 112-117, 1996. Ioachim E, Kamina S, Demou A, et al.: Immunohistochemical localization of metallothionein in human breast cancer in comparison with cathepsin D, stromelysin-1, CD44, extracellular matrix components, P53, Rb, C-erbB-2, EGFR, steroid receptor content and proliferation. Anticancer Res 19: 2133- 2139, 1999. Goulding H, Jasani B, Pereira H, et al.: Metallothionein expression in human breast cancer. Br J Cancer 72: 968-972, 1995. Douglas-Jones AG, Navabi H, Morgan JM, et al.: Immunoreactive p53 and metallothionein expression in duct carcinoma in situ of the breast. No correlation. Virchows Arch 430: 373- 379, 1997. Jin R, Bay BH, Chow VT, et al.: Metallothionein 1E mRNA is highly expressed in oestrogen receptor-negative human invasive ductal breast cancer. Br J Cancer 83: 319-323, 2000. Cree IA, Knight L, Di Nicolantonio F, et al.: Chemosensitization of solid tumors by modulation of resistance mechanisms. Curr Opin Investig Drugs 3: 634-640, 2002. Kishi K, Doki Y, Miyata H, et al.: Prediction of the response to chemoradiation and prognosis in oesophageal squamous cancer. Br J Surg 89:597-603, 2002. Siu LL, Banerjee D, Khurana RJ, et al.: The prognostic role of p53, metallothionein, P-glycoprotein, and MIB-1 in muscleinvasive urothelial transitional cell carcinoma. Clin Cancer Res 4: 559-565, 1998. Joseph MG, Banerjee D, Kocha W, et al.: Metallothionein expression in patients with small cell carcinoma of the lung: correlation with other molecular markers and clinical outcome. Cancer 92: 836-842, 2001. Andrews PA, Murphy MP, Howell SB. Metallothionein-mediated cisplatin resistance in human ovarian carcinoma cells. Cancer Chemother Pharmacol 19: 149-154, 1987. Germain I, Tetu B, Brisson J, et al.: Markers of chemoresistance in ovarian carcinomas: an immunohistochemical study of 86 cases. Int J Gynecol Pathol 15: 54-62, 1996. Wrigley E, Verspaget HW, Jayson GC, et al.: Metallothionein expression in epithelial ovarian cancer: effect of chemotherapy and prognostic significance. J Cancer Res Clin Oncol 126: 717-721, 2000. Shimoda R, Achanzar WE, Qu W, et al.:Metallothionein is a potential negative regulator of apoptosis. Toxicol Sci 73: 294-300, 2003. Cherian MG, Jayasurya A, Bay BH: Metallothioneins in human tumors and potential roles in carcinogenesis. Mutat Res 533: 201-209, 2003. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2004 VL 10 IS 2 BP 74 EP 79 PG 6 ER PT J AU Helal, EATh Khalf Alla, EA Laban, AM Fahmy, MR AF Helal, El-A Thanaa Khalf Alla, E Ali Laban, A Mohamed Fahmy, M Remon TI Immunophenotyping of Tumor-Infiltrating Mononuclear Cells in Ovarian Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ovarian cancer; benign ovarian tumors; CD8+ cells; CD68+ cells ID Ovarian cancer; benign ovarian tumors; CD8+ cells; CD68+ cells AB Infiltrating mononuclear cells play an important role in many types of cancer. The aim of this work was to determine the immunologic characteristics of mononuclear cellular infiltrate in ovarian cancer as compared to benign ovarian tumors. Paraffin-embedded tissues obtained from 52 ovarian carcinomas and 21 benign ovarian neoplasms were examined immunohistochemically to demonstrate suppressor/cytotoxic T cells and macrophages by using CD8 and CD68 monoclonal antibodies, respectively. The mean percentage of CD8+ cells was much higher in the malignant than in the benign group (P=0.00009). Similarly, the mean level of CD68+ cells was significantly higher in carcinomas than in benign cases (P=0.006). There was a significant negative correlation between the percentage of CD8+ cells and CD68+ cells in the malignant group (P=0.000002). Conversely, no correlation could be obtained between the values of these two cell types in the benign lesions. In the malignant group, although the percentages of CD8+ cells and CD68+ cells were not related to tumor differentiation, they were significantly related to tumor type. CD8+ cells were significantly higher in the serous (P=0.02), and CD68+ cells were higher in the mucinous carcinomas (P=0.0005). CD8+ T cells and macrophages constitute a major component of the infiltrating mononuclear cells in ovarian carcinoma. Their frequency seems to be related to the tumor type rather than the degree of tumor differentiation. C1 [Helal, El-A Thanaa] Ain Shams Faculty of Medicine, Department of PathologyCairo, Egypt. [Khalf Alla, E Ali] Ain Shams Faculty of Medicine, Department of Obstetrics and GynecologyCairo, Egypt. [Laban, A Mohamed] Ain Shams Faculty of Medicine, Department of Obstetrics and GynecologyCairo, Egypt. [Fahmy, M Remon] Ain Shams Faculty of Medicine, Department of Obstetrics and GynecologyCairo, Egypt. RP Helal, EATh (reprint author), Ain Shams Faculty of Medicine, Department of Pathology, Cairo, Egypt. EM thanaahelal@hotmail.com CR Dietl J, Horny HP, Ruck P, et al: Dysgerminoma of the ovary. An immunohistochemical study of tumor-infiltrating lymphoreticular cells and tumor cells. Cancer 71:2562-2568, 1997. Balch CM, Riley LB, Bae YJ, et al: Patterns of human tumorinfiltrating lymphocytes in 120 human cancers. Arch Surg 125:200-205, 1990. Freedman RS and Platsoucas CD: Immunotherapy for peritoneal ovarian carcinoma metastasis using ex vivo expanded tumor infiltrating lymphocytes. Cancer Treat Res 82:115- 146, 1996. Negus R, Stamp GW, Hadley J, et al: Quantitative assessment of the leukocyte infiltrate in ovarian cancer and its relationship to the expression of C-C chemokines. Am J Pathol 150:1723-1732, 1997. Bonta IL and Ben-Efraim S: Involvement of inflammatory mediators in macrophage antitumor activity. J Leukoc Biol 54:613-626, 1993. Scully RE, Fox HR, Russel AR, et al: International histopathological classification of ovarian tumors. Curr Opin Obstet Gynecol 4: 608-615, 1992. Hsu SM, Raine L, Fanger H: Use of avidin-biotin-peroxidase complex, ABC, in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody, PAP, procedures. J Histochem Cytochem 29: 577-580, 1981. Naukkarinen A, Syrjanen KJ: Quantitative immunohistochemical analysis of mononuclear infiltrates in breast carcinomas – correlation with tumor differentiation. J Pathol 160: 217-222, 1990. Chouaib S, Asselin-Paturel C, Mami-Chouaib F, et al: The host-tumor immune conflict: from immunosuppression to resistance and destruction. Immunol Today 18: 493-497, 1997. Chen SY, Koffler D, Cohen CJ: Cell-mediated immunity in patients with ovarian carcinoma. Am J Obstet Gynecol 115: 467-470, 1973. Kabawat SE, Bast RC, Welch WR, et al: Expression of major histocompatibility antigens and nature of inflammatory cellular infiltrate in ovarian neoplasms. Int J Cancer 32: 547-554, 1983. Svennevig JL, Svaar H: Content and distribution of macrophages and lymphocytes in solid malignant human tumors. Int J Cancer 24: 754-758, 1979. Meogi AJ, Marrogi AJ, Ramesh R, et al: Tumor-host interaction: analysis of cytokines, growth factors, and tumor-infiltrating lymphocytes in ovarian carcinomas. Hum Pathol 28: 321-331, 1997. Mantovani A, Bottazzi B, Colotta F, et al: The origin and function of tumor-associated macrophages. Immunol Today 13: 265-270, 1992. Zusman I, Gurevich P, Gurevich E, Ben-Hur H: The immune system, apoptosis and apoptosis-related proteins in human ovarian tumors, a review). Int J Oncol 18: 965-972, 2001. Kullander S, Rausing A: Phenotypic characteristics of mononuclear cells in human ovarian tumors – with special reference to cystic and ascitic fluid. Int J Gynecol Cancer 4: 298-305, 1994. Lockhart DC, Chan AK, Mak S, et al: Loss of T-cell receptor- CD3 and T-cell function in tumor-infiltrating lymphocytes but not in tumor-associated lymphocytes in ovarian carcinoma. Surgery 129: 749-756, 2001. Brasanac D, Markovic-Lipkovski J, Hadzi-Djokic J, et al: Immunohistochemical analysis of HLA class II antigens and tumor infiltrating mononuclear cells in renal cell carcinoma: correlation with clinical and histopathological data. Neoplasma 46: 173-178, 1999. Csiszar A, Szentes T, Haraszti B, et al: Characterisation of cytokine mRNA expression in tumour-infiltrating mononuclear cells and tumour cells freshly isolated from human colorectal carcinomas. Eur Cytokine Netw 12: 87-96, 2001. Puccetti L, Manetti R, Parronchi P, et al: Role of low nuclear grading of renal carcinoma cells in the functional profile of tumor-infiltrating T cells. Int J Cancer 98: 674-681, 2002. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2004 VL 10 IS 2 BP 80 EP 84 PG 5 ER PT J AU Moldvay, J Jackel, M Bogos, K Soltesz, I Agocs, L Kovacs, G Schaff, Zs AF Moldvay, Judit Jackel, Marta Bogos, Krisztina Soltesz, Ibolya Agocs, Laszlo Kovacs, Gabor Schaff, Zsuzsa TI The Role of TTF-1 in Differentiating Primary and Metastatic Lung Adenocarcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TTF-1; immunohistochemistry; lung adenocarcinoma; metastasis ID TTF-1; immunohistochemistry; lung adenocarcinoma; metastasis AB Thyroid transcription factor-1 (TTF-1) is a sensitive marker for pulmonary and thyroid adenocarcinomas. The aim of this work was to determine its usefulness in distinction between primary and metastatic lung adenocarcinomas. We have examined the expression of TTF-1 in 100 solitary pulmonary nodules. They included 50 stage I peripheral primary bronchial adenocarcinomas (30 men, 20 women, mean age: 60 years) and 50 metastatic pulmonary adenocarcinomas (21 men, 29 women, mean age: 57 years) of different origins, such as breast (13), colon (13), rectum (13), kidney (7), stomach (2), and thyroid gland (2). TTF-1 immunohistochemistry was performed on formalin-fixed, paraffin-embedded tissues. In primary bronchial adenocarcinomas we found immunopositivity in 46/50 cases, among them 30 cases showed strong nuclear immunostaining. In four primary adenocarcinoma cases the observed immunopositivity was localized to the cytoplasm. Out of the metastatic adenocarcinomas all but the 2 thyroid cancers were negative. Both thyroid tumors showed strong immunopositivity. Our results confirm that TTF-1 immunohistochemistry is a very sensitive and highly specific method in the differential diagnosis of primary and metastatic lung adenocarcinomas and should be used in the everyday clinical practice. C1 [Moldvay, Judit] County Hospital of Pulmonology, Munkacsy M. u. 70., 2045 Torokbalint, Hungary. [Jackel, Marta] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Bogos, Krisztina] National Koranyi Institute of PulmonologyBudapest, Hungary. [Soltesz, Ibolya] National Koranyi Institute of PulmonologyBudapest, Hungary. [Agocs, Laszlo] National Koranyi Institute of PulmonologyBudapest, Hungary. [Kovacs, Gabor] National Koranyi Institute of PulmonologyBudapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Moldvay, J (reprint author), County Hospital of Pulmonology, 2045 Torokbalint, Hungary. EM drmoldvay@hotmail.com CR Abutaily AS, Addis BJ, Roche WR: Immunohistochemistry in the distinction between malignant mesothelioma and pulmonary adenocarcinoma: a critical evaluation of new antibodies. J Clin Pathol 55: 662-668, 2002. Afify AM, al-Khafaji BM: Diagnostic utility of thyroid transcription factor-1 expression in adenocarcinomas presenting in serous fluids. Acta Cytol 46: 675-678, 2002. Bejarano PA, Mousavi F: Incidence and significance of cytoplasmic thyroid transcription factor-1 immunoreactivity. Arch Pathol Lab Med 127: 193-195, 2003. Cai YC, Banner B, Glickman J, et al: Cytokeratin 7 and 20 and thyroid transcription factor 1 can help distinguish pulmonary from gastrointestinal carcinoid and pancreatic endocrine tumors. Hum Pathol 32: 1087-1093, 2001. Castro CY, Moran CA, Flieder DG, et al: Primary signet ring cell adenocarcinomas of the lung: a clinicopathological study of 15 cases. Histopathology 39: 397-401, 2001. Chhieng DC, Cangiarella JF, Zakowski MF, et al: Use of thyroid transcription factor 1, PE-10, and cytokeratins 7 and 20 in discriminating between primary lung carcinomas and metastatic lesions in fine-needle aspiration biopsy specimens. Cancer 93: 330-336, 2001. Fujita J, Ohtsuki Y, Bandoh Set al: Expression of thyroid transcription factor-1 in 16 human lung cancer cell lines. Lung Cancer 39: 31-36, 2003. Goldstein NS, Thomas M: Mucinous and nonmucinous bronchioloalveolar adenocarcinomas have distinct staining patterns with thyroid transcription factor and cytokeratin 20 antibodies. Am J Clin Pathol 116: 319-325, 2001. Gomez-Fernandez C, Jorda M, Delgado PI, et al: Thyroid transcription factor 1: a marker for lung adenoarinoma in body cavity fluids. Cancer 96: 289-293, 2002. Haque AK, Syed S, Lele SM, et al: Immunohistochemical study of thyroid transcription factor-1 and HER2/neu in non-small cell lung cancer: strong thyroid transcription factor-1 expression predicts better survival. Appl Immunohistochem Mol Morphol 10: 103-109, 2002. Hecht JL, Pinkus JL, Weinstein LJ, et al: The value of thyroid transcription factor-1 in cytologic preparations as a marker for metastatic adenocarcinoma of lung origin. Am J Clin Pathol 116: 483-488, 2001. Jang KY, Kang MJ, Lee DG, et al: Utility of thyroid transcription factor-1 and cytokeratin 7 and 20 immunostaining in the identification of origin in malignant effusions. Anal Quant Cytol Histol 23: 400-404, 2001. Lau SK, Desrochers MJ, Luthringer DJ: Expression of thyroid transcription factor-1, cytokeratin 7, and cytokeratin 20 in bronchioloalveolar carcinomas: an immunohistochemical evaluation of 67 cases. Mod Pathol 15: 538-542, 2002. Lau SK, Luthringer DJ, Eisen RN: Thyroid transcription factor- 1: a review. Appl Immunohistochem Mol Morphol 10: 97- 102, 2002. Merchant SH, Amin MB, Tamboli P, et al: Primary signet-ring cell carcinoma of lung: immunohistochemical study and comparison with non-pulmonary signet-ring cell carcinomas. Am J Surg Pathol 25: 1515-1519, 2001. Nakamura N, Miyagi E, Murata S, et al: Expression of thyroid transcription factor-1 in normal and neoplastic lung tissues. Mod Pathol 15: 1058-1067, 2002. Ng WK, Chow JC, Ng PK: Thyroid transcription factor-1 is highly sensitive and specific in differentiating metastatic pulmonary from extrapulmonary adenocarcinoma in effusion fluid cytology specimens. Cancer 96: 43-48, 2002. Ordonez NG: Value of thyroid transcription factor-1 immunostaining in distinguishing small cell lung carcinomas from other small cell carcinomas. Am J Surg Pathol 24: 1207-1223, 2000. Srodon M, Westra WH: Immunohistochemical staining for thyroid transcription factor-1: a helpful aid in discerning primary site of tumor origin in patients with brain metastases. Hum Pathol 33: 642-645, 2002. Sturm N, Lantuejoul S, Laverriere MH, et al: E. Thyroid transcription factor 1 and cytokeratins 1, 5, 10, 14, 34betaE12, expression in basaloid and large-cell neuroendocrine carcinomas of the lung. Hum Pathol 32: 918-925, 2001. Wu M, Wang B, Gil J, et al: p63 and TTF-1 immunostaining. A useful marker panel for distinguishing small cell carcinoma of lung from poorly differentiated squamous cell carcinoma of lung. Am J Clin Pathol 119: 696-702, 2003. Yatabe Y, Mitsudomi T, Takahashi T: TTF-1 expression in pulmonary adenocarcinomas. Am J Surg Pathol 26: 767-773, 2002. Zamecnik J, Kodet R: Value of thyroid transcription factor-1 and surfactant apoprotein A in the differential diagnosis of pulmonary carcinomas: a study of 109 cases. Virchows Arch 440: 353-361, 2002. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2004 VL 10 IS 2 BP 85 EP 88 PG 4 ER PT J AU Kacar Ozkara, S Corakci, A AF Kacar Ozkara, Sevgiye Corakci, Aydin TI Significantly Decreased P27 Expression In Endometrial Carcinoma Compared to Complex Hyperplasia with Atypia (correlation with p53 expression) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrium; carcinoma; hyperplasia; p27; p53 ID Endometrium; carcinoma; hyperplasia; p27; p53 AB P27 expression was examined on paraffin-embedded specimens in proliferative, secretory, hyperplastic and neoplastic human endometrium by immunohistochemistry. The results of p27 immunoreactivity in endometrial carcinomas were compared with clinicopathological indicators as well as with p53 expression. Thirty-eight cases of endometrial carcinoma, 30 normal functional (15 proliferative, 15 secretory), 24 hyperplastic endometrium (12 without atypia, 12 with atypia) specimens were studied by using monoclonal p27 and p53 antibodies. The streptavidin-biotin-peroxidase detection system was used and the intensity and the distribution of immunoreactivity was evaluated semiquantitatively. p27 expression was present both in the proliferative and secretory phases; the expression being stronger in the secretory period. In complex hyperplasia with atypia, p27 expression was even higher and it was significantly reduced in the endometrial carcinoma group (p<0.05). No significant correlation was found between p27 expression and any of the clinicopathologic prognostic parameters (p>0.05). Nuclear p53 expression was detected in 13 (34.2%) patients with endometrial carcinoma and was higher in non-endometrioid carcinomas and in tumors with increasing FIGO grade (p<0.05). High expression of p53 was not found to be a significant prognostic indicator of survival (p>0.05). No p53 expression was detected in the endometria with proliferation, secretion or hyperplasia either simple without atypia or complex with atypia. Surprisingly, tumors with absent/low p27 expression showed absent/low p53 expression. Our data suggest that p27 is necessary to control the proliferation of endometrium and its loss of expression seems to play a role in some aspects of endometrial carcinogenesis. C1 [Kacar Ozkara, Sevgiye] Kocaeli University School of Medicine, Department of Pathology, Mustafa Pasa Mah, Bagdat Cad. 0712. Sok. No. 19/6, 41400 Kocaeli, Gebze, Turkey. [Corakci, Aydin] Kocaeli University School of Medicine, Department of Gynecology and ObstetricsKocaeli, Turkey. RP Kacar Ozkara, S (reprint author), Kocaeli University School of Medicine, Department of Pathology, 41400 Kocaeli, Turkey. EM emrecan1999@yahoo.com CR Al Kushi A, Lim P, Aquino-Parsons C, et al: Markers of proliferative activity are predictors of patient outcome for low-grade endometrioid adenocarcinoma but not papillary serous carcinoma of endometrium. Mod Pathol 15: 365-371, 2002 Bamberger AM, Riethdorf L, Milde-Langosch K, et al: Strongly reduced expression of the cell cycle inhibitor p27 in endometrial neoplasia. Virchows Arch 434: 423-428, 1999 Berchuck A, Kohler MF, Marks JR, et al: The p53 tumor suppressor gene frequently is altered in gynecologic cancers. Am J Obstet Gynecol 170: 246-252, 1994 Cinel L, Polat A, Aydin O, et al: Bcl-2, iNOS, p53 and PCNA expression in normal, disordered proliferative, hyperplastic and malignant endometrium. Pathology International 52: 384-389, 2002 Dubowy RL, Feinberg RF, Keefe DL, et al: Improved endometrial assessment using cyclin E and p27. Fertil Steril 80: 146- 156, 2003 Elhafey AS, Papadimitriou JC, El-hakim MS, et al: Computerized image analysis of p53 and proliferating cell nuclear antigen expression in benign, hyperplastic and malignant endometrium. Arch Pathol Lab Med 125: 872-879, 2001 Enomoto T, Fujita M, Inoue M, et al: Alterations of the p53 tumor suppressor gene and its association with activation of the c-K-ras-2 protooncogene in premalignant and malignant lesions of the human uterine endometrium. Cancer Research 53: 1883-1888, 1993 Erdem O, Erdem M, Dursun A, et al: Angiogenesis, p53, and bcl-2 expression as prognostic indicators in endometrial cancer: comparison with traditional clinicopathologic variables. Int J Gynecol Pathol 22: 254-260, 2003 Kaku T, Kamura T, Hirakawa T, et al: Endometrial carcinoma associated with hyperplasia - Immunohistochemical study of angiogenesis and p53 expression. Gynecol Oncol 72: 51-5, 1999 Kato N, Watanabe J, Jobo T, et al: Immunohistochemical expression of cyclin E in endometrial adenocarcinoma, endometrioid type, and its clinicopathological significance. J Cancer Res Clin Oncol 129: 222-226, 2003 [Epub 2003 Apr 08] Kimura F, Watanabe J, Hata H, et al: PTEN immunohistochemical expression is suppressed in G1 endometrioid adenocarcinoma of the uterine corpus. J Cancer Res Clin Oncol [Epub ahead of print] , 2003 Dec 20 Li SF, Shiozawa T, Nakayama K, et al: Stepwise abnormality of sex steroid hormone receptors, tumor suppressor gene products, p53 and Rb), and cyclin E in uterine endometrioid carcinoma. Cancer 77: 321-9, 1996 Masciullo V, Susini T, Zamparelli A, et al: Frequent loss of expression of the cyclin-dependent kinase inhibitor p27(Kip1, in estrogen-related endometrial adenocarcinomas. Clin Cancer Res 9: 5332-5338, 2003 Miturski R, Semczuk A, Tomaszewski J, et al: Bcl-2 protein expression in endometrial carcinoma: the lack of correlation with p53. Cancer Letters 133: 63-69, 1998 Oshita T, Shigemasa K, Nagai N, et al: p27, cyclin E, and CDK2 expression in normal and cancerous endometrium. Int J Oncol 21: 737-743, 2002 Saegusa M, Machida D, Okayasu I: Age-dependent differences in tumor cell polarity in endometrial carcinomas. J Cancer Res Clin Oncol 128: 205-213, 2002 Shiozawa T, Horiuchi A, Kato K, et al: Up-regulation of p27Kip1 by progestins is involved in the growth suppression of the normal and malignant human endometrial glandular cells. Endocrinology 142: 4182-4188, 2000 Shiozawa T, Nikaido T, Nakayama K, et al: Involvement of cyclin-dependent kinase inhibitor p27Kip1 in growth inhibition of endometrium in the secretory phase and of hyperplastic endometrium treated with progesterone. Mol Hum Reprod 4: 899-905, 1998 Siufi AA, Cotrim G, Da Silva ID, et al: Effects of tamoxifen therapy on the expression of p27 protein in the endometrium of women with primary breast cancer. Int J Oncol 23: 1545-1551, 2003 Suzuki C, Matsumoto T, Sonoue H, et al: Prognostic significance of the infiltrative pattern invasion in endometrioid adenocarcinoma of the endometrium. Pathol Int 53: 495-500, 2003 Watanabe J, Sato H, Kanai T, et al: Paradoxical expression of cell cycle inhibitor p27 in endometrioid adenocarcinoma of the uterine corpus – correlation with proliferation and clinicopathological parameters. Br J Cancer 87: 81-85, 2002 Zhuang YH, Sarca D, Weisz A, et al: Cell type-specific induction of cyclin D and cyclin-dependent kinase inhibitor p27(kip1, expression by estrogen in rat endometrium. J Steroid Biochem Mol Biol 78: 193-199, 2001 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2004 VL 10 IS 2 BP 89 EP 97 PG 9 ER PT J AU Szende, B Farid, P Vegso, Gy Perner, F Kopper, L AF Szende, Bela Farid, Parvaneh Vegso, Gyula Perner, Ferenc Kopper, Laszlo TI Apoptosis and P53, Bcl-2 and Bax Gene Expression in Parathyroid Glands of Patients with Hyperparathyroidism SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Parathyroid; adenoma; hyperplasia; carcinoma; mitosis; apoptosis; Bcl-2; p53; Bax ID Parathyroid; adenoma; hyperplasia; carcinoma; mitosis; apoptosis; Bcl-2; p53; Bax AB Altogether 107 patients were operated on at the Department of Transplantation and Surgery of Semmelweis University in the past four years, for clinical symptoms of hyperparathyroidism. Clinical and laboratory data of the patients supported the diagnosis of primary or secondary hyperparathyroidism. Chronically impaired renal function was found in 52 cases. The removed parathyroid glands showed hyperplasia in 54, adenoma in 50 and carcinoma in 3 cases. The majority of parathyroid lesions in primary hyperparathyroidism were adenomas (41 cases) and in secondary hyperparathyroidism were hyperplasias (43 cases). The ratio of oxyphil to chief cells as well as occasional mitotic and apoptotic figures were determined. The oxyphil component was present in both hyperplastic and tumorous lesions. Apoptosis and mitosis were rarely seen in hyperplasias and adenomas (under 2%), whereas in carcinomas 3% of the tumor cells were apoptotic and 4% showed mitosis. Cytoplasmic p53 positivity could be observed in 3 of the adenomas and in 2 of the hyperplasias. The carcinomas, four adenomas and 3 hyperplasias showed nuclear p53 positivity. Bcl-2 and Bax were detected in the cytoplasm of the tumor cells in the majority of adenomas and in the cells of hyperplasias. Oxyphil cells were more frequently positive than chief cells or clear cells. Colocalization of Bcl-2 and Bax was found randomly in all types of lesions. The very low incidence of carcinoma, the low mitotic and apoptotic ratio in adenomas and hyperplasias suggest a potent antiproliferative defense mechanism in the parathyroid cell population. This may also be reflected in the cytoplasmic colocalization of various gene products which regulate cell death and cell proliferation. No significant differences in the p53, Bcl-2 and Bax spectrum were found between the primary and secondary (i.e. renal failure) parathyroid alterations. C1 [Szende, Bela] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, Ulloi u. 26, H-1085 Budapest, Hungary. [Farid, Parvaneh] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, Ulloi u. 26, H-1085 Budapest, Hungary. [Vegso, Gyula] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Perner, Ferenc] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Kopper, Laszlo] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, Ulloi u. 26, H-1085 Budapest, Hungary. RP Szende, B (reprint author), Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, H-1085 Budapest, Hungary. EM bszende@korb1.sote.hu CR Roth SJ: The parathyroid gland. In Silverberg, S G.Principals and practice of surgical pathology. Second edition. Churchill Livingstone, New York, pp; 1923-1955, 1990 Reiss E, Counterbury I M. Spectrum of hyperparathyroidism. Am J Med 56: 784-810, 1974 Koea JB, Shaw JH: Parathyroid cancer: Biology and management. Surg Oncol 8:155-165, 1999 Bocsi J, Perner F, Szucs J, et al: DNA content of parathyroid tumors, Anticancer Res 18: 2901-2904, 1998 Ricci F, Mingazzini PL, Sebastiani V, et al: P53 as a marker of differentiation between hyperplastic and adenomatous lesions of parathyroids. Int Diag Pathol 6:229-235, 2002 Stojadinovic A, Hoos A, Nissan A, et al: Cordon-Cardo C, Shaha AR, Brennan MF, Singh B, Gossein RA. Parathyroid neoplasms: Clinical, histopathological, and tissue microarraybased molecular analysis.Hum Pathol 34: 54-64, 2003 Abbona GC, Papotti M, Gasparri G, et al: Proliferative activity in parathyroid tumors as detected by Ki-67 immunostaining. Hum Pathol 26: 135-138, 1995 Vargas MP, Vargas HI, Kleiner DE, et al: The role of prognostic markers, MIB-1, RB, and Bcl-2, in the diagnosis of parathyroid tumors. Mod Pathol 10: 12-17, 1997 Erikson LA, Jin L, Wollan P, et al: Parathyroid hyperplasia, adenomas and carcinomas: Differential expression of P27 protein. Am J Surg Pathol 23: 288-295 1999 Schantz A, Castleman B: Parathyroid carcinoma: A study of 70 cases. Cancer 31: 600-605, 1973 Snover DC,Foukar K: Mitotic activity in benign parathyroid disease. Am J Clin Pathol 75: 355-347, 1981 Zang Y, Xiong Y:P53 amino terminal nuclear signal inhibited by DNA damage-induced phosphorylation. Science 292: 1910- 1915, 2000 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2004 VL 10 IS 2 BP 98 EP 103 PG 6 ER PT J AU Ghosh, S Maity, P AF Ghosh, Sonali Maity, Putul TI Isolation and Purification of Vascular Endothelial Growth Factor (VEGF) from Ascitic Fluid of Ovarian Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE VEGF; ascitic fluid; ovarian cancer ID VEGF; ascitic fluid; ovarian cancer AB Vascular Endothelial Growth Factor (VEGF) or Vascular Permeability Factor (VPF) is an angiogenic cytokine expressed by many human and animal tumors. Because of the importance of VEGF in animal tumors, we purified VEGF/VPF from ascitic fluid of ovarian cancer patients with heparin sepharose column. The purified protein gave protein bands of 37 and 26 kD, respectively in 12% SDS PAGE. The specificity of the purified protein was determined with dot blot, trans-immunoblot and ELISA using polyclonal goat anti-VEGF antibody (Santa Cruz Biotechnology). The vasodilatatory effect of the purified protein was confirmed by a vascular permeability assay on mouse. A polyclonal mouse antibody was raised against the purified protein, which recognized the same protein by ELISA, transimmunoblot and dot-blot analysis. It has been also found that the raised polyclonal antibody in mouse- and the commercial VEGF polyclonal antibody (Santa Cruz Biotechnology) both inhibited in vitrocell proliferation of human MCF-7 cell line. This study shows for the first time an effort to purify VEGF from human source. C1 [Ghosh, Sonali] Chittaranjan National Cancer Institute, Department of Metabolic Regulation, 37, S. P. Mukherjee Road, 700026 Kolkata, India. [Maity, Putul] Chittaranjan National Cancer Institute, Department of Metabolic Regulation, 37, S. P. Mukherjee Road, 700026 Kolkata, India. RP Maity, P (reprint author), Chittaranjan National Cancer Institute, Department of Metabolic Regulation, 700026 Kolkata, India. EM putulmaity@yahoo.co.in CR Beliveau R, Gingras D, Kruger E A, et al: The antiangiogenic agent Neovastat, AE-941, inhibits vascular endothelial growth factor mediated biological effects. Clin Cancer Res 8: 1242- 1250, 2002. Claffey KP, Brown LF, del Aguila LF et al: Expression of vascular permeability factor/ vascular endothelial growth factor by melanoma cells increases tumor growth, angiogenesis, and experimental metastasis. Cancer Res 56: 172-181, 1996. Escribano MJ, Benedi VJ, Cordier J: Assay of antigen antibodies by fixation to nitrocellulose sheets and immunodetection. Immunologia 2: 71-76, 1963. Ferrara N: Vascular endothelial growth factor. Eur J Cancer 32A:2413-2422, 1996 Hanahan D and Folkman J: Pattern and emerging mechanism of angiogenic switch during tumerogenesis. Cell 86: 353-364, 1996. Luo JC, Yamaguchi S, Shinkai A et al.: Significant expression of vascular endothelial growth factor in mouse ascitic tumors. Cancer Res 58: 2652-2660, 1998. Kim KJ, Li B, Winer J, et al.: Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumor growth in vivo. Nature 362:841-844,1993. Kraft A, Weindel K, Ochs A, et al.: Vascular endothelial growth factor in the sera and effusions of patients with malignant and nonmalignant disease. Cancer 95: 178-187, 1999. Kondo S, Asano M, Matsuo K, Ohmori I, Suzuki H: Vascular endothelial growth factor/ vascular permeability factor is detectable in sera of tumor bearing mice and cancer patients. Biochem Biophys Acta 1221:211-214, 1994. Laemmli UK:Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227: 680-685, 1993. Leung DW, Cachinaes G, Kuang WJ, Goeddel DV, Ferara N: Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 246: 1306-1309,1989 Nagy JA, Masse EM, Herzberg KT, et al: Pathogenesis of ascitic tumor growth: Vascular permeability factor, vascular hyperpermeability and ascitic fluid accumulation. Cancer Res 55: 360-368, 1995. Obermair A, Tempfer C, Hefler L, et al.: Concentration of vascular endothelial growth factor, VEGF, in the serum of patients with suspected ovarian cancer. Br J Cancer 77:1870- 1874, 1988. Senger D R, Van de WaterL, Brown LF, et al.: Vascular permeability factor, VPF, VEGF, in tumor biology. Cancer Metast Rev 12: 303-324,1993 Takahashi Y, Kitachi Y, Bucana CD et al: Expression of vascular endothelial growth factor and its receptor KDR correlates with vascularity, metastasis, and proliferation of human colon cancer. Cancer Res 55: 3964-3968, 1995. Toi M, Hoshina S, Takayanagi T, et al:: Association of vascular endothelial growth factor expression with tumor angiogenesis and with early relapse in primary breast cancer. Jpn J Cancer Res 85: 1045-1049, 1994. Towbin H, Staehelin T, Gordon J: Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets. Procedure and some application. Proc Natl Acad Sci, USA, 76: 4350-4354, 1979. Voller A, Biddwell DE, Barlett A: The ELISA Dynatch Laboratories Inc., Alexandria, Virginia, 1979. Wood M J, Bold G, Buchdunger E, Cozens R, Ferrari S, Frei J et al. PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases impairs vascular endothelial growth factor induced responses and tumor growth after oral administration. Cancer Res 60: 2178-2189, 2000. Yamamoto Y, Toi M, Kondo S et al. Concentration of vascular endothelial growth factor in sera of normal controls and cancer patients. Clin Cancer Res 2: 821-826, 1996. Yabushita H, Shimazu M, Noguchi M, et al.: Vascular endothelial growth factor activating matrix metalloproteinase in ascitic fluid during peritoneal dissemination of ovarian cancer. Oncol Rep10: 89-95, 2003. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2004 VL 10 IS 2 BP 104 EP 108 PG 5 ER PT J AU Csiszar, A Szentes, T Haraszti, B Balazs, A Petranyi, GGy Pocsik, AF Csiszar, Anna Szentes, Tamas Haraszti, Bea Balazs, Annamaria Petranyi, G Gyozo Pocsik, Eva TI The Pattern of Cytokine Gene Expression in Human Colorectal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cytokine; gene expression; colorectal carcinoma; RT-PCR ID cytokine; gene expression; colorectal carcinoma; RT-PCR AB Systemic and local cytokine environment may modulate the immunogenicity of colorectal cancer cells, and affect anti-tumor immune functions of tumorinfiltrating lymphocytes. We therefore investigated cytokine mRNA expression patterns in tumors and peripheral blood mononuclear cells (PBMC) from patients with colorectal adenocarcinoma. IL-2, IFN-g, tumor necrosis factor-a (TNF-a), IL-4, IL-6, IL-8, IL-10 and IL-1b mRNAs in single cell suspension of freshly isolated colorectal cancer tissue were studied by RT-PCR. Frequencies of cytokine gene expression were compared to those in normal colonic mucosa from tumor patients. The frequencies of IL-2, IFN-g, IL-4 and IL-10 gene expression were also determined in peripheral blood mononuclear cells from patients with colorectal adenocarcinoma and compared to those of healthy individuals. Tumor samples were more frequently positive for IFN-g, IL-2, TNF-a and IL-10 gene expression than normal mucosa (p=0.0001, p=0.0118, p=0.001 and p<0.0001, respectively). Frequencies of IL-2 and TNF-a gene expressions were significantly higher in tumors with a diameter <5 cm, than in those with a diameter >5 cm. The genes for IL-6, IL-1b and IL-8 were commonly expressed in both tumor tissue and normal colonic mucosa. IFN-g transcripts were detected in more PBMC samples from patients with colorectal cancer than those from normal controls (p=0.0449). Thus, colorectal cancer tissue is characterized by a specific pattern of cytokine gene expression. It is likely that multiple interactions between pro- and anti-inflammatory cytokines regulate tumor growth and the functional activity of tumor-infiltrating lymphocytes. C1 [Csiszar, Anna] New York Medical College, Department of PhysiologyValhalla, USA. [Szentes, Tamas] Szent Imre Hospital, Department of SurgeryBudapest, Hungary. [Haraszti, Bea] National Medical Center, Department of Hematology and Stem Cell Transplantation, 64 Dioszegi ut, H-1113 Budapest, Hungary. [Balazs, Annamaria] New York Medical College, Department of PhysiologyValhalla, USA. [Petranyi, G Gyozo] National Medical Center, Department of Hematology and Stem Cell Transplantation, 64 Dioszegi ut, H-1113 Budapest, Hungary. [Pocsik, Eva] National Medical Center, Department of Hematology and Stem Cell Transplantation, 64 Dioszegi ut, H-1113 Budapest, Hungary. RP Pocsik, (reprint author), National Medical Center, Department of Hematology and Stem Cell Transplantation, H-1113 Budapest, Hungary. EM pocsik@ohvi.hu CR Hilgenfeld RU and Kreuser ED: Immunological and biochemical modulation in the treatment of advanced colorectal cancer: Update and future directions. Curr Topics Microbiol Immunol 213: 217-240, 1996. Berghella AM, Pellegrini P, Del Beato T et al.: The significance of an increase in soluble interleukin-2 receptor level in colorectal cancer and its biological regulating role in the physiological switching of the immune response cytokine network from TH1 to TH2 and back. Cancer Immunol Immunother 45: 241-249, 1998 Shibata M, Nezu T, Takekawa M et al: Serum levels of interleukin- 10 and interleukin-12 in patients with colorectal cancer. Ann N Y Acad Sci 795: 410-412,1996. Tsushima H, Kawata S, Tamura S et al: High levels of transforming growth factor 1 in patients with colorectal cancer: association with disease progression. Gastroenterology 110: 375-382, 1996. Zaloudik J, Lauerova L, Janakova L et al: Significance of pretreatment immunological parameters in colorectal cancer patients with unresectable metastases to the liver. Hepatogastroenterology 46: 220-227, 1999. Heriot AG, Marriott JB, Cookson S et al: Reduction in cytokine production in colorectal cancer patients: association with stage and reversal by resection. Br J Cancer 82: 1009- 1012, 2000. Lahm H, Schindel M, Frikart L et al: Selective suppression of cytokine secretion in whole blood cell cultures of patients with colorectal cancer. Br J Cancer 78: 1018-1023, 1998. O’Hara RJ, Greenman J, Drew PJ et al: Impaired interleukin- 12 production is associated with a defective anti-tumor response in colorectal cancer. Dis Colon Rectum 41: 460-463, 1998. Brew R, Southern SA, Flanagan BF et al: Detection of interleukin- 8 mRNA and protein in human colorectal carcinoma cells. Eur J Cancer 32A: 2142-2147, 1996. Gastl GA, Abrams JS, Nanus DM et al: Interleukin-10 production by human carcinoma cell lines and its relationship to interleukin- 6 expression. Int J Cancer 55: 96-101, 1993. Langerak AD, Garewal HS: Transforming growth factor-b1: a useful tumor marker in patients with colorectal carcinoma? Cancer 85: 517-519, 1999. Mauerer MJ, Walter W, Martin D et al: Interleukin-7, IL-7, in colorectal cancer: IL-7 is produced by tissues from colorectal cancer and promotes preferential expansion of tumour infiltrating lymphocytes. Scand J Immunol 45: 182-192, 1997. Coca S, Perez-Piqueras J, Martinez D et al: The prognostic significance of intratumoral natural killer cells in patients with colorectal carcinoma. Cancer 79: 2320-2328, 1997. Di Giorgio A, Botti C, Tocchi A et al: The influence of tumor lymphocytic infiltration on long term survival of surgically treated colorectal cancer patients. Int Surg 77: 256-260, 1992. Jass JR: Lymphocytic infiltration and survival in rectal cancer. J Clin Pathol 39: 585-589, 1986. Naito Y, Saito K, Shiiba K et al: CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res 58: 3491-3494, 1998. Goedegebuure PS, Eberlein TJ: The role of CD4+ tumor-infiltrating lymphocytes in human solid tumors. Immunol Res 14: 119-131, 1995. Mosmann TR, Li L, Sad S: Function of CD8 T-cell subsets secreting different cytokine patterns. Semin Immunol 9: 87-92, 1997. Xie K: Interleukin-8 and human cancer biology. Cytokine Growth Factor Rev 12: 375-391, 2001. Rosen EM, Zitnik R J, Elias JA et al: The interaction of HGFSF with other cytokines in tumor invasion and angiogenesis. EXS 65: 301-310, 1993. Opdenakker G, Van den Steen PE, Dubois B et al: Gelatinase B functions as regulator and effector in leukocyte biology. J Leukoc Biol 69: 851-859, 2001. Kossakowska AE, Urbanski SJ, Janowska-Wieczorek A: Matrix metalloproteinases and their tissue inhibitors - expression, role and regulation in human malignant non-Hodgkin’s lymphomas. Leuk Lymphoma 39: 485-493, 2000. Wilson J, Balkwill F: The role of cytokines in the epithelial cancer microenvironment. Semin Cancer Biol 12: 113-120, 2002. Anasagasti MJ, Olaso E, Calvo F et al: Interleukin 1-dependent and -independent mouse melanoma metastases. J Natl Cancer Inst 89: 645-651, 1997. Pocsik E, Mihalik R, Gyodi E et al: Activation of lymphocytes after platelet allotransfusion possessing only class I MHC product. Clin Exp Immunol 82:102-107, 1990. Paloczi K, Pocsik E, Kotlan B et al: The pattern of activation antigen expression on T-lymphocyte subpopulation in infectious mononucleosis. Haematologia 24: 83-90, 1991. Luo J S, Kammerer R, von Kleist S: Comparison of the effects of immunosuppressive factors from newly established colon carcinoma cell cultures on human lymphocyte proliferation and cytokine secretion. Tumour Biol 21: 11-20, 2000. Csiszar A, Szentes T, Haraszti B et al: Characterization of cytokine mRNA expression in tumour-infiltrating mononuclear cells and tumour cells isolated freshly from human colorectal carcinomas. Eur Cyt Netw 12: 87-96, 2001. Csiszar A, Nagy G, Gergely P, Pozsonyi T, Pocsik E: Increased interferon-, IFN-g), IL-10 and decreased IL-4 mRNA expression in peripheral blood mononuclear cells, PBMC, from patients with systemic lupus erythematosus, SLE). Clin Exp Immunol 122: 464-470, 2000. Chomczynski P: A reagent for the single-step simultaneous isolation of RNA, DNA and proteins from cell and tissue samples. Biotechniques 15: 532-534, 536-537, 1993. Zou W, Durand-Gasselin I, Dulioust A et al: Quantification of cytokine gene expression by competitive PCR using a colorimetric assay. Eur Cyt Netw 6: 257-264, 1995. Shire D, Legoux P, Minty A J: Standardisation of messenger RNA quantification using an RT-PCR method involving coamplification with a multi-specific internal control. In: Modern Applications of DNA Amplification Techniques: Problems and New Tools., Eds. Lassner D, Pustowoit B, and Rolfs A), Plenum Press, New York, 1997, pp. 25-35. Barth RJ Jr, Camp BJ, Martuscello TA et al: The cytokine microenvironment of human colon carcinoma. Lymphocyte expression of tumor necrosis factor-a and interleukin-4 predicts improved survival. Cancer 78: 1168-1178, 1996. Piancatelli D, Romano P, Sebastiani P et al: Local expression of cytokines in human colorectal carcinoma: evidence of specific interleukin-6 gene expression. J Immunother 22: 25-32, 1999. Numata A, Minagawa T, Asano M et al: Functional evaluation of tumor-infiltrating mononuclear cells. Detection of endogenous interferon- and tumor necrosis factor-a in human colorectal adenocarcinomas. Cancer 68: 1937-1943, 1991. Barth RJ Jr, Mule JJ, Spiess PJ, Rosenberg SA: Interferon-g and tumor necrosis factor have a role in tumor regressions mediated by murine CD8+ tumor-infiltrating lymphocytes. J Exp Med 173: 647-658, 1991. Takagi K, Tomita K, Fukushima et al: Endogenous TNF inducibility and prognosis of colorectal cancer. Anticancer Res 18: 4141-4146, 1998. Etoh T, Shibuta K, Barnard GF et al: Angiogenin expression in human colorectal cancer: the role of focal macrophage infiltration. Clin Cancer Res 6: 3545-3551, 2000. Minami S, Furui J, Kanematsu T: Role of carcinoembryonic antigen in the progression of colon cancer cells that express carbohydrate antigen. Cancer Res 61: 2732-2735, 2001. Suzuki S, Mita S, Kamohara H et al: IL-6 and IFN-gregulation of IL-10 production by human colon carcinoma cells. Int J Oncol 18: 581-586, 2001. Tran-Thang C, Kruithof E, Lahm H et al: Modulation of the plasminogen activation system by inflammatory cytokines in human colon carcinoma cells. Br J Cancer 74: 846-852, 1996. Isaacs KL, Sartor RB, Haskill S: Cytokine messenger RNA profiles in inflammatory bowel disease mucosa detected by polymerase chain reaction amplification. Gastroenterology 103: 1587-1595, 1992. Eckmann L, Jung HC, Schurer-Maly C et al: Differential cytokine expression by human intestinal epithelial cell lines: regulated expression of interleukin 8. Gastroenterology 105: 1689-1697, 1993. Fox SH, Whalen GF, Sanders MM et al: Angiogenesis in normal tissue adjacent to colon cancer. J Surg Oncol 69: 230-234, 1998. Ono M, Torisu H, Fukushi J et al: Biological implications of macrophage infiltration in human tumor angiogenesis. Cancer Chemother Pharmacol 43 Suppl: S69-71, 1999. Opdenakker G, Van Damme J: Chemotactic factors, passive invasion and metastasis of cancer cells. Immunol Today 13: 463-464, 1992. Salazar-Onfray F: Interleukin-10: a cytokine used by tumors to escape immunosurveillance. Med Oncol 16: 86-94, 1999. Huang S, Ullrich SE, Bar-Eli M: Regulation of tumor growth and metastasis by interleukin-10: the melanoma experience.J Interferon Cytokine Res 19: 697-703, 1999. Elgert KD, Alleva DG, Mullins DW: Tumor-induced immune dysfunction: the macrophage connection. J Leukoc Biol 64:275-290, 1998. Kiessling R, Wasserman K, Horiguchi S et al: Tumor-induced immune dysfunction. Cancer Immunol Immunother 48: 353-632, 1999. Melichar B, Touskova M, Vesely P: Effect of irinotecan on the phenotype of peripheral blood leukocyte populations in patients with metastatic colorectal cancer. Hepatogastroenterology 49: 967-970, 2002. De Vita F, Orditura M, Galizia G et al: Serum interleukin-10 levels in patients with advanced gastrointestinal malignancies. Cancer 86: 1936-1943, 1999. Giltay EJ, Fonk JC, von Blomberg BM, et al: In vivo effects of sex steroids on lymphocyte responsiveness and immunoglobulin levels in humans. J Clin Endocrinol Metab 85: 1648- 1657, 2000. Danis VA, Franic GM, Rathjen DA, et al: Effects of granulocyte- macrophage colony-stimulating factor, GM-CSF), IL-2, interferon-g, IFN-g), tumour necrosis factor-a, TNF-a, and IL-6 on the production of immunoreactive IL-1 and TNF-aby human monocytes. Clin Exp Immunol 85: 143-150, 1991. McGee DW, Bamberg T, Vitkus SJ, McGhee JR: A synergistic relationship between TNF-a, IL-1b , and TGF-b 1 on IL-6 secretion by the IEC-6 intestinal epithelial cell line. Immunology 86: 6-11, 1995. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2004 VL 10 IS 2 BP 109 EP 116 PG 8 ER PT J AU Vermes, G Acs, N Szabo, I Langmar, Z Jaray, B Banhidy, F AF Vermes, Gabor Acs, Nandor Szabo, Istvan Langmar, Zoltan Jaray, Balazs Banhidy, Ferenc TI Simultaneous Bilateral Occurrence of a Mixed Mesodermal Tumor and Cystadenocarcinoma in the Ovary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE malignant mixed mesodermal tumor; ovary; simultaneous occurrence ID malignant mixed mesodermal tumor; ovary; simultaneous occurrence AB The mixed mesodermal tumor is a very uncommon malignancy. The aggressiveness of this lesion is illustrated by extremely poor prospects for afflicted patients: postoperative survival is usually shorter than 24 months. According to the literature, malignant mixed tumor of the ovary is rather rare and its occurrence with other malignancy is exceptional. We report here a case of a 62-years old woman with serous cystadenocarcinoma in the right ovary and a heterologous malignant mixed mesodermal tumor in the left one. Both tumors expressed cytokeratins, while only the mesodermal tumor expressed S-100 and focal NSE. C1 [Vermes, Gabor] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi u. 78/a, H-1082 Budapest, Hungary. [Acs, Nandor] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi u. 78/a, H-1082 Budapest, Hungary. [Szabo, Istvan] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi u. 78/a, H-1082 Budapest, Hungary. [Langmar, Zoltan] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi u. 78/a, H-1082 Budapest, Hungary. [Jaray, Balazs] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Banhidy, Ferenc] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi u. 78/a, H-1082 Budapest, Hungary. RP Vermes, G (reprint author), Semmelweis University, 2nd Department of Obstetrics and Gynecology, H-1082 Budapest, Hungary. CR Fowler JM, Nathan L, Nieberg RK, Berek JS. Mixed mesodermal sarcoma of the ovary in a young patient. Eur J Obstet Gynecol Reprod Biol 65:249-253, 1966 Le T, Krepart GV, Lotocki RJ, Heywood MS. Malignant mixed mesodermal ovarian tumor treatment and prognosis: A 20 – year experience. Gynecol Oncology 65:237-240, 1997 Ariad S, Rabinovitz A,Yanai-Inbar I, Piura B. Mixed uterine mesodermal sarcoma in the population of southern Israel in the years 1996-1971–clinical and pathological charasteristics. Harefuah 134:93-6, 159, 1998 Nakata Y, Morita M, Mori Y. A malignant mixed mesodermal tumor of the ovary. Gan No Rinsho 36:2210-2217, 1990 Li H, Shi S, Zhang W. Malignant mixed mesodermal tumors of the ovary: a clinical analysis of 12 cases. Zhonghua Zhong Liu Za Zhi 20:460-462,1998 Piura B, Rabinovich A, Yanai-Inbar I, et al. Primary sarcoma of the ovary: report of five cases and review of the literature. Eur J Gynecol Oncol 19:257-261,1998 Kaji Y, Sugimura K, Yamamoto N, et al: A case of malignant mixed mesodermal tumor, MMMT, of the ovary: MR features before and after chemotherapy. Radiat Med 17:81-3,1999 Krigman HR, Coogan AC, Marks JR: Simultaneous endometrial malignant mixed mesodermal tumor and ovarian serous adenocarcinoma. Arch Pathol Lab Med 119:99-103, 1995 Ergeneli MH, Demirhan B, Duran EH, Coskun M: Malignant mixed mesodermal tumor arising in a benign cystic teratoma. Eur J Obstet Gynecol Reprod Biol 83:191-4,1999 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2004 VL 10 IS 2 BP 117 EP 120 PG 4 ER PT J AU Kabukcuoglu, F Kabukcuoglu, Y Yilmaz, B Erdem, Y Evren, I AF Kabukcuoglu, Fevziye Kabukcuoglu, Yavuz Yilmaz, Banu Erdem, Yesim Evren, Ismail TI Mazabraud's Syndrome: Intramuscular Myxoma Associated with Fibrous Dysplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Mazabraud's syndrome; intramuscular myxoma; fibrous dysplasia ID Mazabraud's syndrome; intramuscular myxoma; fibrous dysplasia AB The association of fibrous dysplasia and intramuscular myxoma is a rare disease known as Mazabraud's syndrome. Both lesions tend to occur in the same anatomical region. The relationship between fibrous dysplasia and myxoma remains unclear, where an underlying localized error in tissue metabolism has been proposed to explain this occasional coexistence. Another example of this syndrome in a 52 year-old woman is reported. The patient presented with a soft tissue mass at the anteromedial mid part of the left thigh. After excision of the mass, three separate bone lesions were detected in her control MRI. The soft tissue mass was misdiagnosed as liposarcoma in another center, and the bone lesions were interpreted as metastasis. The hypocellularity and the indistinct vascular pattern of the lesion were consistent with myxoma. The Jam-Shidi needle biopsies of the osseous lesions were diagnosed as fibrous dysplasia. The recognition of this entity is important for appropriate management of the patient. Patients with soft tissue myxomas should be thoroughly examined for fibrous dysplasia. The greater risk of sarcomatous transformation in fibrous dysplasia with Mazabraud's syndrome should also be kept in mind. C1 [Kabukcuoglu, Fevziye] Sisli Etfal Training and Research Hospital, Department of Pathology, No:10/1 Dogancilar, Uskudar, 81160 Istanbul, Turkey. [Kabukcuoglu, Yavuz] Sisli Etfal Training and Research Hospital, Department of Orthopedics and TraumatologyIstanbul, Turkey. [Yilmaz, Banu] Sisli Etfal Training and Research Hospital, Department of Pathology, No:10/1 Dogancilar, Uskudar, 81160 Istanbul, Turkey. [Erdem, Yesim] Sisli Etfal Training and Research Hospital, Department of Pathology, No:10/1 Dogancilar, Uskudar, 81160 Istanbul, Turkey. [Evren, Ismail] Sisli Etfal Training and Research Hospital, Department of Pathology, No:10/1 Dogancilar, Uskudar, 81160 Istanbul, Turkey. RP Kabukcuoglu, F (reprint author), Sisli Etfal Training and Research Hospital, Department of Pathology, 81160 Istanbul, Turkey. EM ykabukcuoglu@yahoo.com CR Blasier DR, Ryan JR, Schaldenbrand MF: Multiple myxomata of soft tissue associated with polyostotic fibrous dysplasia: A case report. Clin Orthop 206: 211-214, 1986 Cabral CE, Guedes P, Fonseca T, et al: Polyostotic fibrous dysplasia associated with intramuscular myxomas: Mazabraud’s syndrome. Skeletal Radiol. 27: 278-282, 1998 Cohen MM, Siegal GP: Congenital and inherited syndromes. In: Pathology and Genetics of Tumours of Soft Tissue and Bone, Eds: Fletcher CDM, Unni KK, Mertens F, WHO Classification of Tumours, IARC Press, Lyon, 2002, pp 357-359 Court-Payen M, Jensen LI, Bjerregaard B, et al: Intramuscular myxoma and fibrous dysplasia of bone – Mazabraud’s syndrome. Acta Radiol. 38: 368-371, 1997 Enzinger FM, Weiss SW: Benign Soft Tissue Tumors and Psedotumors of Miscellaneous Type. In: Enzinger and Weiss’s Soft Tissue Tumors., Eds: Weiss SW, Goldblum JR), Ed 4. St Louis, Mosby, 2001, pp.1419-1481 Gianoutsos MP, Thompson JF, Marsden FW: Mazabraud’s Syndrome: Intramuscular myxoma associated with fibrous dysplasia of bone. Aust N Z J Surg 60: 825-828, 1990 Iwasko N, Steinbach LS, Disler D, et al: Imaging findings in Mazabraud’s syndrome: seven new cases. Skeletal Radiol. 31:81-87, 2002 Jhala DN, Eltoum I, Carrol AJ, et al: Osteosarcoma in a patient with McCune-Albright syndrome and Mazabraud’s syndrome: a case report emphasizing the cytological and cytogenic findings. Hum Pathol. 34:1354-1357, 2003 Lopez-Ben R, Pitt MJ, Jaffe KA and Siegal GP: Osteosarcoma in a patient with McCune-Albright syndrome and Mazabraud’s syndrome. Skeletal Radiol. 28: 522-526, 1999 Mazabraud A, Semat P, Roze R: A propos de l’association de fibromyxomes des tissus mous a la dysplasie fibreuse des os. Presse Med 75: 2223-2228, 1967 Miettinen M, Hockerstedt K, Reitamo J and Totterman S: Intramuscular myxoma – a clinicopathological study of twentythree cases. Am J Clin Pathol 84: 265-272, 1985 Prayson MA, Leeson MC: Soft-tissue myxomas and fibrous dysplasia of bone. Clin Orthop 291: 222-228, 1993 Silver WP, Harrelson JM, Scully SP: Intramuscular myxoma: a clinicopathologic study of 17 patients. Clin Orthop 403: 191- 197, 2002 Sundaram M, McDonald DJ, Merenda G: Intramuscular myxoma: a rare but important association with fibrous dysplasia of bone. AJR Am J Roentgenol 153:107-108, 1989 Wirth UA, Leavitt D, Enzinger FM: Multiple intramuscular myxomas. Another extraskeletal manifestation of fibrous dysplasia. Cancer 27:1167, 1971. Witkin GB, Guiliford WB, Siegal GP: Osteogenic sarcoma and soft tissue myxoma in a patient with fibrous dysplasia and hemoglobins. J Baltimore and S Clin Orthop 209: 245-252, 1986 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2004 VL 10 IS 2 BP 121 EP 123 PG 3 ER PT J AU Bruheim, S Bruland, SO Breistol, K Maelandsmo, MG Fodstad, O AF Bruheim, Skjalg Bruland, S Oyvind Breistol, Knut Maelandsmo, M Gunhild Fodstad, Oystein TI Human Osteosarcoma Xenografts and Their Sensitivity to Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE osteosarcoma; human tumor xenograft; chemotherapy; drug resistance ID osteosarcoma; human tumor xenograft; chemotherapy; drug resistance AB Despite the increased survival rates of osteosarcoma patients attributed to adjuvant chemotherapy, at least one third of the patients still die due to their disease. Further improvements in the management of osteosarcoma may rely on a more individualised treatment strategy, as well as on the introduction of new drugs. To aid in the preclinical evaluation of new candidate substances against osteosarcoma, we have established 11 human osteosarcoma xenograft lines and characterised them with regard to response to five different reference drugs. Doxorubicin, cisplatin methotrexate, ifosfamide and lomustine were effective in 3/11, 3/11, 1/10, 5/11 and 4/11 of the xenografts, respectively. Five xenografts were resistant to all compounds tested. We also assessed the mRNA expression levels of the xenografts for the O6-Methylguanine DNA Methyltransferase (MGMT), DNA topoisomerase II- (Topo II)-a, Gluthathione-S-transferase (GST)-p, Multidrug-resistance related protein (MRP) 1 and Multidrug-resistance (MDR) 1 genes. There was an inverse correlation between the transcript levels of GST-p and doxorubicin growth inhibition (r=-0.66; p<0.05), and between the transcript levels of MGMT and the effect of lomustine (r=-0.72; p<0.01), whereas the expression of MRP1 and cisplatin growth inhibition was positively correlated (r=0.82; p<0.005). This panel of xenografts should constitute a good tool for pharmacological and molecular studies in osteosarcoma. C1 [Bruheim, Skjalg] The Norwegian Radium Hospital, Department of Tumor Biology, Montebello, 0310 Oslo, Norway. [Bruland, S Oyvind] The Norwegian Radium Hospital, Department of Medical Oncology and RadiotherapyOslo, Norway. [Breistol, Knut] The Norwegian Radium Hospital, Department of Tumor Biology, Montebello, 0310 Oslo, Norway. [Maelandsmo, M Gunhild] The Norwegian Radium Hospital, Department of Tumor Biology, Montebello, 0310 Oslo, Norway. [Fodstad, Oystein] The Norwegian Radium Hospital, Department of Medical Oncology and RadiotherapyOslo, Norway. RP Bruheim, S (reprint author), The Norwegian Radium Hospital, Department of Tumor Biology, 0310 Oslo, Norway. EM skjalg.bruheim@labmed.uio.no CR Malawer MM, Link MP, Donaldson SS. Sarcomas of the Bone. In DeVita VT, Hellmann S, Rosenberg SA, eds. Cancer: Principles and Practice of Oncology 2001; 6th Edition, Volume 2):18911936. Souhami R, Cannon S. Osteosarcoma. In Peckham M, Pinedo HM, Veronesi U, eds. Oxford Textbook of Oncology. Oxford, Oxford University Press 1995: 1969-1976. Bruland OS, Pihl A. On the current management of osteosarcoma. A critical evaluation and a proposal for a modified treatment strategy. Eur J Cancer 33:1725-1731, 1997 Sambrook J, Fritsh EF, Maniatis T. Molecular Cloning: a Laboratory Manual. Cold Spring Harbor, NY: Cold Spring Harbour Laboratory Press 1989; 2nd Edition. Feinberg AP, Vogelstein B. A technique for radiolabeling DNA restriction endonuclease fragments to high specific activity. Anal Biochem 132:6-13, 1983 Church GM, Gilbert W. Genomic sequencing. Proc Natl Acad Sci USA 81:1991-1995, 1984 Tsai-Pflugfelder M, Liu LF, Liu AA, et al: Cloning and sequencing of cDNA encoding human DNA topoisomerase II and localization of the gene to chromosome region 17q21-22. Proc Natl Acad Sci USA 85:7177-7181, 1988 Moscow JA, Townsend AJ, Goldsmith ME, et al: Isolation of the human anionic glutathione S-transferase cDNA and the relation of its gene expression to estrogen-receptor content in primary breast cancer. Proc Natl Acad Sci USA 85:6518-6522, 1988 Tano K, Shiota S, Collier J, et al: Isolation and structural characterization of a cDNA clone encoding the human DNA repair protein for 06-alkylguanine. Proc Natl Acad Sci USA 87:686- 690, 1990 Zaman GJ, Flens MJ, van Leusden MR, et al: The human multidrug resistance-associated protein MRP is a plasma membrane drug-efflux pump. Proc Natl Acad Sci USA 91:8822- 8826, 1994 Fuqua SA, Fitzgerald SD, McGuire WL. A simple polymerase chain reaction method for detection and cloning of low-abundance transcripts. Biotechniques 9:206-211, 1990 Budach W, Budach V. Association of tumor growth on nude mice and poor clinical outcome in soft tissue sarcoma patients. J Cancer Res Clin Oncol 127:523-530, 2001 Hoffmann J, Schmidt-Peter P, Hansch W, et al: Anticancer drug sensitivity and expression of multidrug resistance markers in early passage human sarcomas. Clin Cancer Res 5:2198-204, 1999 Boven E, Pinedo HM, van Hattum AH, et al: Characterization of human soft-tissue sarcoma xenografts for use in secondary drug screening. Br J Cancer 78:1586-1593, 1998 Budach W, Budach V, Stuschke M, el al: Efficacy of ifosfamide, dacarbazine, doxorubicin and cisplatin in human sarcoma xenografts. Br J Cancer 70:29-34, 1994 Meyer WH, Houghton JA, Houghton PJ, et al: Development and characterization of pediatric osteosarcoma xenografts. Cancer Res 50:2781-2785, 1990 Harris MB, Cantor AB, Goorin AM, et al: Treatment of osteosarcoma with ifosfamide: comparison of response in pediatric patients with recurrent disease versus patients previously untreated: a Pediatric Oncology Group study. Med Pediatr Oncol 24:87-92, 1995 Scotlandi K, Serra M, Nicoletti G, et al: Multidrug resistance and malignancy in human osteosarcoma. Cancer Res 56:2434- 2439, 1996 Scotlandi K, Manara MC, Serra M, et al: The expression of P-glycoprotein is causally related to a less aggressive phenotype in human osteosarcoma cells. Oncogene 18:739-746, 1999 Serra M, Scotlandi K, Reverter-Branchat G, et al: Value of Pglycoprotein and clinicopathologic factors as the basis for new treatment strategies in high-grade osteosarcoma of the extremities. J Clin Oncol 21:536-542, 2003 Serra M, Maurici D, Scotlandi K, et al: Relationship between P-glycoprotein expression and p53 status in high-grade osteosarcoma. Int J Oncol 14:301-307, 1999 Chan HS, Grogan TM, Haddad G, et al: P-glycoprotein expression: critical determinant in the response to osteosarcoma chemotherapy. J Natl Cancer Inst 89:1706-1715, 1997 Baldini N, Scotlandi K, Barbanti-Brodano G, et al: Expression of P-glycoprotein in high-grade osteosarcomas in relation to clinical outcome. N Engl J Med 333:1380-1385, 1995 Posl M, Amling M, Grahl K, et al: P-glycoprotein expression in high grade central osteosarcoma and normal bone cells. An immunohistochemical study. Gen Diagn Pathol 142:317-325, 1997 Wunder JS, Bull SB, Aneliunas V, et al: MDR1 gene expression and outcome in osteosarcoma: a prospective, multicenter study. J Clin Oncol 18:2685-2694, 2000 Gorlick R, Huvos AG, Heller G, et al: Expression of HER2/erbB-2 correlates with survival in osteosarcoma. J Clin Oncol 17:2781-2788, 1999 Suto R, Abe Y, Nakamura M, et al: Multidrug resistance mediated by overexpression of P-glycoprotein in human osteosarcoma in vivo. Int J Oncol 12:287-291, 1998 Radig K, Hackel C, Herting J, et al: Expression of P-glycoprotein in high grade osteosarcomas with special emphasis on chondroblastic subtype. Gen Diagn Pathol 142:139-145, 1997 Borst P, Evers R, Kool M, Wijnholds J. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst 92:1295-302, 2000 Ifergan I, Meller I, Issakov J, Assaraf YG. Reduced folate carrier protein expression in osteosarcoma: implications for the prediction of tumor chemosensitivity. Cancer 98:1958-1966, 2003 Batist G, Tulpule A, Sinha BK, et al: Overexpression of a novel anionic glutathione transferase in multidrug-resistant human breast cancer cells. J Biol Chem 261:15544-15549, 1986 Goto S, Ihara Y, Urata Y, et al. Doxorubicin-induced DNA intercalation and cavenging by nuclear glutathione S-transferase pi. Faseb J 15:2702-2714, 2001 Saburi Y, Nakagawa M, Ono M, et al. Increased expression of glutathione S-transferase gene in cis-diamminedichloroplatinum( II)-resistant variants of a Chinese hamster ovary cell line. Cancer Res 1989;49(24 Pt 1):7020-5. Wang YY, Teicher BA, Shea TC, et al: Cross-resistance and glutathione- S-transferase-pi levels among four human melanoma cell lines selected for alkylating agent resistance. Cancer Res 49:6185-6192, 1989 Puchalski RB, Fahl WE. Expression of recombinant glutathione S-transferase pi, Ya, or Ybl confers resistance to alkylating agents. Proc Natl Acad Sci USA 87:2443-2447, 1990 Ban N, Takahashi Y, Takayama T, et al: Transfection of glutathione S-transferase, GST)-pi antisense complementary DNA increases the sensitivity of a colon cancer cell line to adriamycin, cisplatin, melphalan, and etoposide. Cancer Res 56:3577-3582, 1996 Gilbert L, Elwood LJ, Merino M, et al: A pilot study of pi-class glutathione S-transferase expression in breast cancer: correlation with estrogen receptor expression and prognosis in nodenegative breast cancer. J Clin Oncol 11:49-58, 1993 Shiga H, Heath El, Rasmussen AA, et al: Prognostic value of p53, glutathione S-transferase pi, and thymidylate synthase for neoadjuvant cisplatin-based chemotherapy in head and neck cancer. Clin Cancer Res 5:4097-4104, 1995 Green JA, Robertson LJ, Clark AH. Glutathione S-transferase expression in benign and malignant ovarian tumors. Br J Cancer 68:235-239, 1993 Bai F, Nakanishi Y, Kawasaki M, et al: Immunohistochemical expression of glutathione S-transferase-Pi can predict chemotherapy response in patients with nonsmall cell lung carcinoma. Cancer 78:416-421, 1996 Uozaki H, Horiuchi H, Ishida T, et al: Overexpression of resistance- related proteins, metallothioneins, glutathione-S-transferase pi, heat shock protein 27, and lung resistance-related protein, in osteosarcoma. Relationship with poor prognosis. Cancer 79:2336-2344, 1997 Kellner U, Sehested M, Jensen PB, et al: Culprit and victim – DNA topoisomerase II. Lancet Oncol 3:235-243, 2002 Gerson SL. Clinical relevance of MGMT in the treatment of cancer. J Clin Oncol 20:2388-2399, 2002 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2004 VL 10 IS 3 BP 133 EP 141 PG 9 ER PT J AU Mehes, G Speich, N Bollmann, M Bollmann, R AF Mehes, Gabor Speich, Norbert Bollmann, Magdolna Bollmann, Reinhard TI Chromosomal Aberrations Accumulate in Polyploid cells of High-grade Squamous Intraepithelial Lesions (HSIL) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HPV; aneuploidy; chromosome; cytology; cervix ID HPV; aneuploidy; chromosome; cytology; cervix AB Persistant infection with human papillomavirus (HPV) of the uterine cervix is related with cytological atypia (SIL), the oncogenic potential of which is unclear in a given time point of monitoring. HPVinduced genetic instability result in polyploidization as well as in low frequency random chromosome aberrations in squamous cells. In the present work we analyzed whether highly polyploid/aneuploid cells reflect genomic changes at the chromosomal level. 13 samples with the cytological diagnosis of HSIL were analyzed for HPV type and nuclear DNA content measured by laser scanning cytometry (LSC). Hyperdiploid cells with >5c and with >9c DNA content were further analyzed for numerical aberrations of the chromosomes 3 and 17 by fluorescence in situ hybridization (FISH) following repositioning. Cells with >5c DNA content were found more frequently than cells with >9c DNA content (5-98 and 1-44 cells, respectively). The FISH analysis demonstrated frequent polysomies, however, the rate of aneusomy (other than 2, 4, 8 or 16 chromosome copies) was significantly higher in cells with >9c DNA content than in cells with >5c DNA content or the normal diploid cells. The imbalance of chromosome 3 and 17 copy number was also increased in cells with >9c DNA content. Moreover, in three out of the 13 analyzed HSIL samples, recurrent abnormal chromosome 3/17 ratio was demonstrated in a significant part of the cells, indicating a common origin of these cells. Highly polyploid/aneuploid cells in HSIL accumulate cytogenetic aberrations detectable by FISH analysis. These cells may reflect early changes with tumorigenic potential in a very concentrated fashion. C1 [Mehes, Gabor] Institute of PathologyBonn-Duisdorf, Germany. [Speich, Norbert] University of Bonn, Institute of PathologyBonn, Germany. [Bollmann, Magdolna] Institute of PathologyBonn-Duisdorf, Germany. [Bollmann, Reinhard] Institute of PathologyBonn-Duisdorf, Germany. RP Mehes, G (reprint author), Institute of Pathology, Bonn-Duisdorf, Germany. EM mehesg@pathology.pote.hu CR zur Hausen H: Human papillomaviruses in the pathogenesis of anogenital cancer. Virology 184: 9-13, 1991 Ferenczy A, Franco E: Persistent human papillomavirus infection and cervical neoplasia. Lancet Oncol 3:11-167, 2002 Bosch FX, Lorincz A, Munoz N, et al: The causal relation between human papillomavirus and cervical cancer. J Clin Pathol 55:244- 265, 2002 4 Zielinsky GD, Snijders PJF, Rozendaal L, et al. HVP presence precedes abnormal cytology in women developing cervical cancer and signals false negative smears. Br J Cancer 85: 398-404, 2001 5. The 1988 Bethesda System for reporting cervical/vaginal cytologic diagnoses. Acta Cytol 33: 567-72, 1989 6. Solomon D, Davay D, Kurman R, et al: The Bethesda 2001 Workshop: The 2001 Bethesda System: terminology of reporting results of cervical cytology. JAMA 287: 2114-2119, 2002 7. Bocking A, Chatelain R: Diagnostic and prognostic value of DNA cytometry in gynaecologic cytology. Anal Quant Cytol Histol 11: 177-186, 1989 8. Hanselaar AG, Vooijs GP, Mayall BH, et al: DNA changes in progressive cervical intraepithelial neoplasia. Anal Cell Pathol 4: 315- 324, 1992 9. Winkler B, Crum CP, Fujii T, et al: Koilicytic lesions of the cervix: the relationship of mitotic abnormalities to the presence of papillomavirus antigens and nuclear DNA-content. Cancer 53: 1081-1087, 1984 10. Bibbo M, Dytch HE, Alenghat E, et al: DNA ploidy profiles as indicators in CIN lesions. Am J Clin Pathol 92: 261-265, 1989 11. Bollmann R, Bollmann M, Henson DE, Bodo M: DNA-cytometry confirms the utility of the Bethesda System for the classification of Papanicolaou smears. Cancer 93: 222-228, 2001 12. Bollmann R, Mehes G, Torka R, et al: HPV typing and DNA ploidy determination in squamous intraepithelial lesions, SIL, in liquid fixed cytological samples. Cancer 99: 57-62, 2003 13. Bollmann R, Mehes G, Torka R, et al: Determination of features indicating progression in ASCUS: HPV typing and DNA ploidy analysis from liquid based cytological samples. Cancer 99: 113- 117, 2003 14. Tarnok A, Gerstner AO: Clinical applications of laser scanning cytometry. Cytometry 50: 133-143, 2002 15. Hanselaar AG, Bocking A, Gundlach H, et al: International Consensus Conference on the Fight Against Cervical Cancer IAC Task Force 8 Summary, Chicago, Illinois, USA: Summary statement on quantitative cytochemistry, DNA and molecular biology): Task Force 8. Acta Cytol 45: 499-501, 2001 16. Feoli-Fonseca JC, Oligny LL, Filion M, et al: Two-tier polymerase chain reaction direct sequencing method for detecting an typing human papillomaviruses in pathological specimens. Diagn Mol Pathol 7: 317-323, 1998 17. Jacobs MV, Snijders PJ, van den Brule JC, et al: A general Primer GP5+/GP6+-Mediated PCR-Enzyme Immunoassay Method for Rapid Detection of 14 High-Risk and 6 Low-Risk Human Papillomavirus Genotypes in Cervical Scrapings. J Clin Microbiol 35: 791-795, 1997 18. Bauer HM, Greer CE, Manos MM: Determination of genital HPV infection using consensus PCR. In: Herrington CS and McGee JO, ed.): Diagnostic molecular pathology: a practical approach. Oxford University Press, Oxford, 1992 19. Duensing S, Duensing A, Flores ER, et al: Centrosome abnormalities and genomic instability by episomal expression of human papillomavirus type 16 in raft cultures of human keratinocytes. J Virol 75: 7712-7716, 2001 20. Skyldberg B, Fujioka K, Hellstrom AC, et al: Human papillomavirus infection, centrosome aberration and genetic stability in cervical lesions. Mod Pathol 14: 279-284, 2001 21. Southern SA, Herrington CS: Differential cell cycle regulation by low – and high-risk human papillomaviruses in low-grade squamous intraepithelial lesions of the cervix. Cancer Res 58: 2941- 2945, 1998 22. Giannoudis A, Evans MF, Southern SA, Herrington CS: Basal keratinocyte tetrasomy in low-grade squamous intra-epithelial lesions of the cervix is restricted to high and intermediate risk HPV infection but is not type-specific. Br J Cancer 82: 424-428, 2000 23. Hopman AH, Voorter CE, Ramaekers FC: Detection of genomic changes in cancer by in situ hybridization. Mol Biol Rep 19: 31-44, 1994 24. Bollmann R, Torka R, Schmitz J, et al: Determination of ploidy and steroid receptor status in breast cancer by laser scanning cytometry. Cytometry 50: 210-215, 2002 25. Atkin NB, Baker MC, Fox MF: Chromosome changes in 43 carcinomas of the cervix uteri. Cancer Genet Cytogenet 44: 229-241, 1990 26. Umayahara K, Numa F, Suehiro Y, et al: Comparative genomic hybridization detects genetic alterations during early stages of cervical cancer progression. Genes Chrom Cancer 33: 98-102, 2002 27. Kirchhoff M, Rose H, Petersen BL, et al: Comparative genomic hybridization reveals a recurrent pattern of chromosomal aberrations in severe dysplasia/carcinoma in situ of the cervic and in advanced stage cervical carcinoma. Genes Chromosomes Cancer 24: 144-150, 1999 28. Hidalgo A, Schewe C, Petersen S, et al: Human papillomavirus status and chromosomal imbalances in primary cervical carcinomas and tumour cell lines. Eur J Cancer 36: 542-548, 2000 29. Brink AATP, Wiegant JCAG, Szuhai K, et al: Simultaneous mapping of human papillomavirus integration sites and molecular karyotyping in short term cultures of cervical carcinomas by using 49- color combined binary ratio labeling fluorescence in situ hybridization. Cancer Genet Cytogenet 134: 145-150, 2002 30. Bulten J, Poddighe PJ, Robben JC,et al: Interphase cytogenetic analysis of cervical intraepithelial neoplasia. Am J Pathol 152: 495- 503, 1998 31. Aubele M, Zitzelsberger H, Schenck U, et al: Distinct cytogenetic alterations in squamous intraepithelial lesions of the cervix revealed by laser–assisted microdissection and comparative genomic hybridization. Cancer 84: 375-379, 1998 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2004 VL 10 IS 3 BP 142 EP 148 PG 7 ER PT J AU Lorincz, T Timar, J Szendroi, M AF Lorincz, Tamas Timar, Jozsef Szendroi, Miklos TI Alterations of Microvascular Density in Bone Metastases of Adenocarcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bone metastasis; microvessel density; breast cancer; lung cancer; renal cell cancer ID bone metastasis; microvessel density; breast cancer; lung cancer; renal cell cancer AB Bone may provide an extremely fertile microenvironment for angiogenesis. Experimental investigations indicate angiogenesis as a major regulator of bone metastasis development. Vascularization and angiogenic potential is known for most of the primary tumor types, but no studies investigated angiogenesis in bone metastases of human cancers. We have evaluated microvessel density of bone metastases of various cancer types (all adenocarcinomas) and compared to their primary tumors in paraffin samples of 39 patients. Microvessel density was determined by using the hot spot method and the blood vessel marker, CD34. The most vascularized adenocarcinoma was found to be renal cell cancer followed by lung adenocarcinoma, while breast cancer was heterogenous in this respect. Two patterns of modulation of the angiogenic phenotype in the bone metastases emerged in this study, which seemed to be cancer type specific: decreased angiogenic potential characterizing 45% of renal cell cancers and breast cancers of high vascularity in their primary, and increased angiogenic potential characterizing 40% of lung adenocarcinomas and breast cancers of low vascularity in their primary lesion. Our data demonstrate that i., the vascularization of bone metastases is frequently altered compared to the primary tumors, ii., patterns are different in the case of various cancer types. The tumor-type specific alterations of the angiogenic phenotype of cancers, metastatic to the bone, can have a clinical significance when angiosuppressive therapies are considered. C1 [Lorincz, Tamas] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Szendroi, Miklos] Semmelweis University, Department of OrthopedicsBudapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. CR Hasan J, Byers R, Jayson GC. Intra-tumoral microvessel density in human solid tumours. Br J Cancer 86:1566-1577, 2002 Yoneda T. Cellular and molecular mechanisms of breast and prostate cancer metastasis to bone. Eur J Cancer 34:240-245, 1998 Saaristo A, Karpanen T, Alitalo K. Mechanisms of angiogenesis and their use in the inhibition of tumor growth and metastasis. Oncogene 19:6122-6129, 2000 Winding B, Misander H, Sveigaard C et al. Human breast cancer induced angiogenesis, recruitment, and activation of osteoclasts in osteolytic metastasis. J Cancer Res Clin Oncol 126:631-640, 2000 Van der Pluijm G, Sijmons B, Vloedgraven H, et al: Monitoring metastatic behavior of human tumor cells in mice with species-specific polymerase chain reaction: elevated expression of angiogenesis and bone resorption stimulators by breast cancer in bone metastases. J Bone Miner Res 16:1077-1091, 2001 Woodhouse EC, Chuaqui RF, Liotta LA. General mechanisms of metastasis. Cancer 80:1529-1537, 1997 Ellis LM, Liu W, Ahmad SA et al. Overview of angiogenesis: Biologic implications for antiangiogenic therapy. Semin Oncol 28:94-104, 2001 Kovacs L, Szende B, Elek G et al. Working experience with a new vacuum-accelerated microwave histoprocessor. J Pathol 180:106-110, 1996 Weidner N, Semple JP, Welch WR, Folkman J. Tumor angiogenesis and metastasis-correlation in invasive breast carcinoma. N Engl J Med 324:1-8, 1991 Mundy GR. Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer 2:584-593, 2002 Yang JC, Haworth L, Sherry RM et al. A randomised trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med 349:427-434, 2003 Hurwitz H, Fehrenbacher L, Novotny W et al. Bevacizumab plus irinotecan, fluorouracil and leucovorin for metastatic colorectal cancer. N Engl J Med 350:2335-23342, 2004. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2004 VL 10 IS 3 BP 149 EP 153 PG 5 ER PT J AU Amirghofran, Z Monabati, A Khezri, A Malek-Hosseini, Z AF Amirghofran, Zahra Monabati, Ahmad Khezri, Abdolaziz Malek-Hosseini, Zahra TI Apoptosis in Transitional Cell Carcinoma of Bladder and its Relation to Proliferation and Expression of P53 and Bcl-2 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Transitional cell carcinoma of bladder; apoptosis; bcl-2; p53 ID Transitional cell carcinoma of bladder; apoptosis; bcl-2; p53 AB Transitional cell carcinoma of bladder (TCC) is a relatively common cancer among men. Tumor progression is associated with expression or modulation of several gene products that control apoptosis and proliferation. Apoptosis is a negative growth regulatory mechanism in tumors. The aim of this study is to examine apoptosis and related regulatory molecular markers in a group of patients with TCC. Paraffinembedded tissues from 49 patients with TCC were examined for the expression of bcl-2, p53 and Ki-67 by immunohistochemistry. Apoptosis was detected by TUNEL method. Correlation between apoptotic index (AI), proliferation index (PI) and bcl-2 and p53 expression with each other and with pathological grade was determined. Apoptosis was observed in 28.1% of TCC cases. The mean AI of all cases was 13.7±24. No correlation was found between apoptosis and differentiation status of carcinoma. Bcl-2 expression was weakly detected in only one sample. P53 expression was detected in 26 of cases with mean staining index of 102±96. A significant correlation between p53 and Ki-67 staining indices was observed (r=0.521, p=0.001). Both p53 and Ki-67 expression showed a good association with the pathological grade (p=0.0001 and p=0.004, respectively). None of the markers showed significant correlation with AI and no correlation was found between the ratio of AI to PI and other parameters either. In conclusion, the frequency of apoptosis in TCC of bladder appears not to be associated with tumor grade, and with bcl-2, p53 and Ki-67 expression. C1 [Amirghofran, Zahra] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Monabati, Ahmad] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Khezri, Abdolaziz] Medical School, Shiraz University of Medical Sciences, Department of UrologyShiraz, Iran. [Malek-Hosseini, Zahra] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. RP Amirghofran, Z (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Immunology, Shiraz, Iran. CR Hickman JA. Apoptosis and tumorigenesis. Curr Opin Genet Dev 12:67-72, 2002 Rossi D, Gaidano G. Messengers of cell death: apoptotic signaling in health and disease. Haematologica 88:212-218, 2003 Burlacu A. Regulation of apoptosis by Bcl-2 family proteins. J Cell Mol Med 7:249-257, 2003 Batinac T, Gruber F, Lipozencic J et al. Protein p53—structure, function, and possible therapeutic implications. Acta Dermatovenerol Croat 11:225-230, 2003 Laiho M, Latonen L. Cell cycle control, DNA damage checkpoints and cancer. Ann Med 35:391-397, 2003 Sherr CJ. Principles of tumor suppression. Cell 23; 116:235- 246, 2004 Rodriguez-Alonso A, Pita-Fernandez S, Gonzalez-Carrero J, Nogueira-March JL: Multivariate analysis of survival, recurrence, progression and development of metastasis in T1 and T2a transitional cell bladder carcinoma. Cancer 15; 94:1677- 1684, 2002 Lin Z, Kim H, Park H, et al. The expression of bcl-2 and bcl- 6 protein in normal and malignant transitional epithelium. Urol Res 31:272-275, 2003 Yu DS, Chang SY. The expression of oncoproteins in transitional cell carcinoma: its correlation with pathological behavior, cell cycle and drug resistance. Urol Int 69:46-50, 2002 Llopis J, Alcaraz A, Ribal MJ, et al. p53 expression predicts progression and poor survival in T1 bladder tumors. Eur Urol 37:644-653, 2000 World Health Organization. Histological typing of urinary bladder tumors. 10, 1973 Budd RC. Activation-induced cell death. Curr Opin Immunol 13:356-362, 2001 King ED, Matteson J, Jacobs SC, Kyprianou N. Incidence of apoptosis, cell proliferation and bcl-2 expression in transitional cell carcinoma of the bladder: association with tumor progression. J Urol 155:316-320, 1996 Korkolopoulou P, Konstantinidou AE, Christodoulou P, et al. Apoptosis in bladder carcinomas detected with monoclonal antibody to single-stranded DNA: relation to cell cycle regulators and survival. Urology 1; 56:516-520, 2000 Roman S, Petrusca D, Moldovan I, et al. Evaluation of apoptosis of tumor and of apparently normal cells in human renal carcinoma. Immunol Lett 15; 67:15-22, 1999 Kong C, Zhang X, Takenaka I. Apoptotic cell death and Smad4 expression in transitional cell carcinoma of the renal pelvis and ureter. Int J Urol 8:386-390, 2001 Zhang X, Kong C, Takenaka I. Evaluation of cell proliferation, apoptosis, and angiogenesis in transitional cell carcinoma of the renal pelvis and ureter. Urology 57:981-985, 2001 Townson JL, Naumov GN, Chambers AF. The role of apoptosis in tumor progression and metastasis. Curr Mol Med 3:631- 642, 2003 Vermeulen K, Berneman ZN, Van Bockstaele DR. Cell cycle and apoptosis. Cell Prolif 36165-36175, 2003 Bargonetti J, Manfredi JJ. Multiple roles of the tumor suppressor p53. Curr Opin Oncol 14:86-91, 2002 Lu QL, Abel P, Foster CS, Lalani EN. bcl-2: role in epithelial differentiation and oncogenesis. Hum Pathol 27:102-110, 1996 Uchida T, Minei S, Gao JP, et al. Clinical significance of p53, MDM2 and bcl-2 expression in transitional cell carcinoma of the bladder. Oncol Rep 9:253-9, 2002 Ong F, Moonen LM, Gallee MP, et al. Prognostic factors in transitional cell cancer of the bladder: an emerging role for Bcl-2 and p53. Radiother Oncol 61:169-175, 2001 Asci R, Yildiz L, Sarikaya S, et al. p53 and bcl-2 overexpression as associated risk factors in patients 40 years old or less with transitional cell carcinoma of the bladder. Urol Int 67:34- 40, 2001 Kirsh EJ, Baunoch DA, Stadler WM. Expression of bcl-2 and bcl-X in bladder cancer. J Urol 159:1348-1353, 1998 Pfister C, Flaman JM, Dunet F, et al. p53 mutations in bladder tumors inactivate the transactivation of the p21 and Bax genes, and have a predictive value for the clinical outcome after bacillus Calmette-Guerin therapy. J Urol 162:69-73, 1999 Harada H, Grant S. Apoptosis regulators. Rev Clin Exp Hematol 7:117-138, 2003 Smith ND, Rubenstein JN, Eggener SE, Kozlowski JM. The p53 tumor suppressor gene and nuclear protein: basic science review and relevance in the management of bladder cancer. J Urol 169: 1219-1228, 2003 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2004 VL 10 IS 3 BP 154 EP 159 PG 6 ER PT J AU Hegyi, L Hockings, DP Benson, GM Busza, LA Overend, P Grimsditch, CD Burton, JK Lloyd, H Whelan, AG Skepper, NJ Vidgeon-Hart, PM Carpenter, TA Reid, GD Suckling, EK Weissberg, LP AF Hegyi, Laszlo Hockings, D Paul Benson, G Martin Busza, L Albert Overend, Philip Grimsditch, C David Burton, J Katherine Lloyd, Heather Whelan, A Greg Skepper, N Jeremy Vidgeon-Hart, P Martin Carpenter, T Adrian Reid, G David Suckling, E Keith Weissberg, L Peter TI Short Term Arterial Remodelling in the Aortae of Cholesterol Fed New Zealand White Rabbits Shown in vivo by High-Resolution Magnetic Resonance Imaging - Implications for Human Pathology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE animal models of human disease; imaging; MRI; atherosclerosis; remodelling ID animal models of human disease; imaging; MRI; atherosclerosis; remodelling AB High-resolution, non-invasive imaging methods are required to monitor progression and regression of atherosclerotic plaques. We investigated the use of MRI to measure changes in plaque volume and vessel remodelling during progression and regression of atherosclerosis in New Zealand White rabbits. Atherosclerotic lesions were induced in the abdominal aorta by balloon injury and cholesterol feeding. MR images (2D) of the abdominal aorta were acquired with cardiac and respiratory gating using a fast spin echo sequence with and without fat-suppression. In an initial study on rabbits treated for 30 weeks we imaged the aortae with a spatial resolution of 250x250 micrometers with a slice thickness of 2 mm and achieved a close correlation between MRI-derived measurements and those made on perfusion pressure-fixed histological sections (r1= 0.83, slope p1 < 0.01). We subsequently imaged 18 rabbits before and periodically during 12 weeks of cholesterol feeding (progression) followed by 12 weeks on normal diet (regression). Aortic wall (atherosclerotic lesion) volume increased significantly during progression and decreased during regression. In contrast, lumen volume increased during progression and did not change during regression. In conclusion, this study confirms that non-invasive, high-resolution MRI can be used to monitor progression and regression of atherosclerosis, each within 3 months and shows, for the first time in a short-term model, that positive remodelling occurs early during progression and persists through regression of atherosclerotic lesions. C1 [Hegyi, Laszlo] University of Cambridge, Department of Medicine, Division of Cardiovascular Medicine, Hammersmith Hospital, Du Cane Road, W12 0HS Cambridge, UK. [Hockings, D Paul] GlaxoSmithKline, Imaging GroupThe Frythe, Welwyn, UK. [Benson, G Martin] GlaxoSmithKline, Atherosclerosis DepartmentStevenage, Hertfordshire, UK. [Busza, L Albert] GlaxoSmithKline, Imaging GroupThe Frythe, Welwyn, UK. [Overend, Philip] GlaxoSmithKline, Atherosclerosis DepartmentStevenage, Hertfordshire, UK. [Grimsditch, C David] GlaxoSmithKline, Atherosclerosis DepartmentStevenage, Hertfordshire, UK. [Burton, J Katherine] GlaxoSmithKline, Laboratory Animal SciencesThe Frythe, Welwyn, UK. [Lloyd, Heather] GlaxoSmithKline, Laboratory Animal SciencesThe Frythe, Welwyn, UK. [Whelan, A Greg] GlaxoSmithKline, Laboratory Animal SciencesThe Frythe, Welwyn, UK. [Skepper, N Jeremy] University of Cambridge, Department of Anatomy, Multi-Imaging CentreCambridge, UK. [Vidgeon-Hart, P Martin] GlaxoSmithKline, Atherosclerosis DepartmentStevenage, Hertfordshire, UK. [Carpenter, T Adrian] University of Cambridge, Wolfson Brain Imaging CentreCambridge, UK. [Reid, G David] GlaxoSmithKline, Imaging GroupThe Frythe, Welwyn, UK. [Suckling, E Keith] GlaxoSmithKline, Atherosclerosis DepartmentStevenage, Hertfordshire, UK. [Weissberg, L Peter] University of Cambridge, Department of Medicine, Division of Cardiovascular Medicine, Hammersmith Hospital, Du Cane Road, W12 0HS Cambridge, UK. RP Hegyi, L (reprint author), University of Cambridge, Department of Medicine, Division of Cardiovascular Medicine, W12 0HS Cambridge, UK. EM lhegyi@doctors.org.uk CR Tadrous PJ: Methods for imaging the structure and function of living tissues and cells: 3. Confocal microscopy and microradiology. J Pathol 191:345-354, 2000. Kobayashi N, Hosoya T, Adachi M, et al: Virtual MR microscopy for unruptured aneurysm. Comput Methods Programs Biomed 66:99-103, 2001 Cova M, Toffanin R: MR microscopy of hyaline cartilage: current status. Eur Radiol 12:814-823, 2002 Ezawa H, Yoneyama R, Kandatsu S, et al: Introduction of autopsy imaging redefines the concept of autopsy: 37 cases of clinical experience. Pathol Int 53:865-873, 2003 Thali MJ, Yen K, Vock P, et al: Image-guided virtual autopsy findings of gunshot victims performed with multi-slice computed tomography and magnetic resonance imaging and subsequent correlation between radiology and autopsy findings. Forensic Sci Int 138:8-16, 2003 Glagov S, Weisenberg E, Zarins CK, et al: Compensatory enlargement of human atherosclerotic coronary arteries. N Engl J Med 316:1371-1375, 1987 Fayad ZA, Fallon JT, Shinnar M, et al: Noninvasive in vivo high-resolution magnetic resonance imaging of atherosclerotic lesions in genetically engineered mice. Circulation 98:1541- 1547, 1998 McConnell MV, Aikawa M, Maier SE, et al: MRI of rabbit atherosclerosis in response to dietary cholesterol lowering. Arterioscler Thromb Vasc Biol 19:1956-1959, 1999 Fayad ZA, Nahar T, Fallon JT, et al: In vivo magnetic resonance evaluation of atherosclerotic plaques in the human thoracic aorta: a comparison with transesophageal echocardiography. Circulation 101:2503-2509, 2000 Worthley SG, Helft G, Fuster V, et al: Serial in vivo MRI documents arterial remodeling in experimental atherosclerosis. Circulation 101:586-589, 2000 Kolodgie FD, Katocs AS Jr, Largis EE, et al: Hypercholesterolemia in the rabbit induced by feeding graded amounts of low-level cholesterol. Methodological considerations regarding individual variability in response to dietary cholesterol and development of lesion type. Arterioscler Thromb Vasc Biol 16:1454-1464, 1996 Groot PH, van Vlijmen BJ, Benson GM, et al: Quantitative assessment of aortic atherosclerosis in APOE*3 Leiden transgenic mice and its relationship to serum cholesterol exposure. Arterioscler Thromb Vasc Biol 16:926-933, 1996 Stary HC, Chandler AB, Dinsmore RE, et al: A definition of advanced types of atherosclerotic lesions and a histological classification of atherosclerosis. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation 92:1355-1374, 1995 Helft G, Worthley SG, Fuster V, et al: Atherosclerotic aortic component quantification by noninvasive magnetic resonance imaging: an in vivo study in rabbits. J Am Coll Cardiol 37:1149-1154, 2001 Trouard TP, Altbach MI, Hunter GC, et al: MRI and NMR spectroscopy of the lipids of atherosclerotic plaque in rabbits and humans. Magn Reson Med 38:19-26, 1997 Hockings PD, Roberts T, Galloway GJ, et al: Repeated threedimensional magnetic resonance imaging of atherosclerosis development in innominate arteries of low-density lipoprotein receptor-knockout mice. Circulation 106:1716-1721, 2002 Helft G, Worthley SG, Fuster V, et al: Progression and regression of atherosclerotic lesions: monitoring with serial noninvasive magnetic resonance imaging. Circulation 105:993-998, 2002 Choudhury RP, Aguinaldo JG, Rong JX, et al: Atherosclerotic lesions in genetically modified mice quantified in vivo by noninvasive high-resolution magnetic resonance microscopy. Atherosclerosis 162:315-321, 2002 Saito D, Oka T, Kajiyama A, et al: Factors predicting compensatory vascular remodelling of the carotid artery affected by atherosclerosis. Heart 87:136-139, 2002 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2004 VL 10 IS 3 BP 159 EP 165 PG 7 ER PT J AU Pires, RF Perez, EdCD de Almeida, PO Kowalski, PL AF Pires, Ramoa Fabio Perez, Elias da Cruz Danyel de Almeida, Paes Oslei Kowalski, Paulo Luiz TI Estrogen Receptor Expression in Salivary Gland Mucoepidermoid Carcinoma and Adenoid Cystic Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mucoepidermoid carcinoma; adenoid cystic carcinoma; salivary gland tumors; immunohistochemistry; estrogen; estrogen receptor ID mucoepidermoid carcinoma; adenoid cystic carcinoma; salivary gland tumors; immunohistochemistry; estrogen; estrogen receptor AB Estrogen receptor (ER) expression in salivary gland carcinomas is controversial, and most published studies considered no more than 10 cases. We analyzed ER expression by immunohistochemistry in 136 mucoepidermoid carcinomas and 72 adenoid cystic carcinomas. All cases were negative. These results do not support a role for estrogens in salivary gland mucoepidermoid carcinoma and adenoid cystic carcinoma. C1 [Pires, Ramoa Fabio] School of Dentistry, State University of Rio de Janeiro, Oral PathologyRio de Janeiro, Brazil. [Perez, Elias da Cruz Danyel] State University of Campinas, School of Dentistry, Department of Oral DiagnosisPiracicaba, Brazil. [de Almeida, Paes Oslei] State University of Campinas, School of Dentistry, Department of Oral DiagnosisPiracicaba, Brazil. [Kowalski, Paulo Luiz] A. C. Camargo Cancer Hospital, Department of Head and Neck Surgery and Otorhinolaryngology, Rua Prof Antonio Prudente, 211, Liberdade, 01509-900 Sao Paulo, Brazil. RP Kowalski, PL (reprint author), A. C. Camargo Cancer Hospital, Department of Head and Neck Surgery and Otorhinolaryngology, 01509-900 Sao Paulo, Brazil. EM lp_kowalski@uol.com.br CR Arpino G, Clark GM, Mohsin S, et al: Adenoid cystic carcinoma of the breast: molecular markers, treatment, and clinical outcome. Cancer 94: 2119-2227, 2002 Barnes L, Rao U, Contis L, et al: Salivary duct carcinoma. Part II. Immunohistochemical evaluation of 13 cases for estrogen and progesterone receptors, cathepsin D, and c-erbB-2 protein. Oral Surg Oral Med Oral Pathol 78: 74-80, 1994 Dimery IW, Jones LA, Verjan RP, et al: Estrogen receptors in normal salivary gland and salivary gland carcinoma. Arch Otolaryngol Head Neck Surg 113: 1082-1085, 1987 Dori S, Trougouboff P, David R, Buchner A: Immunohistochemical evaluation of estrogen and progesterone receptors in adenoid cystic carcinoma of salivary gland origin. Oral Oncol 36: 450-453, 2000 Ellis GL, Auclair P: Tumors of the Salivary Glands. Atlas of Tumor Pathology, 3rd Series, Fascicle 17, eds). Washington, DC: Armed Forces Institute of Pathology, 1996 Gaffney EV, Pinkston JA, Eidson JJ: Estrogen receptors in parotid tumors. Endocr Res 21: 635-643, 1995 Glas AS, Hollema H, Nap RE, Plukker JT: Expression of estrogen receptor, progesterone receptor, and insulin-like growth factor receptor-1 and of MIB-1 in patients with recurrent pleomorphic adenoma of the parotid gland. Cancer 94: 2211-2216, 2002 Jeannon JP, Soames JV, Bell H, Wilson JA: Immunohistochemical detection of oestrogen and progesterone receptors in salivary tumours. Clin Otolaryngol 24: 52-54, 1999 Lamey PJ, Leake RE, Cowan SK, et al: Steroid hormone receptors in human salivary gland tumours. J Clin Pathol 40: 532- 534, 1987 Miller AS, Hartman GG, Chen SY, et al: Estrogen receptor assay in polymorphous low-grade adenocarcinoma and adenoid cystic carcinoma of salivary gland origin. An immunohistochemical study. Oral Surg Oral Med Oral Pathol 77: 36- 40, 1994 Nasser SM, Faquin WC, Dayal Y: Expression of androgen, estrogen, and progesterone receptors in salivary gland tumors. Frequent expression of androgen receptor in a subset of malignant salivary gland tumors. Am J Clin Pathol 119: 801-806, 2003 Ozono S, Sato K, Ito Y, et al: Immunohistochemical evidence that tumors elicit the synthesis of estrogen receptors in the submandibular gland of female rats. Experientia 51:220-222, 1995 Shick PC, Riordan GP, Foss RD: Estrogen and progesterone receptors in salivary gland adenoid cystic carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 80: 440-444, 1995 Wilson JA, Rogers MJC, Hawkins RA, et al: Epidermal growth factor receptors and oestrogen receptors in the head and neck. Clin Otolaryngol 18: 66-68, 1993 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2004 VL 10 IS 3 BP 166 EP 168 PG 3 ER PT J AU Kovacs, J Varga, A Bessenyei, M Gomba, Sz AF Kovacs, Judit Varga, Attila Bessenyei, Maria Gomba, Szabolcs TI Renal Cell Cancer Associated with Sarcoid-like Reaction SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE renal cancer; sarcoid-like granuloma; immunohistochemistry ID renal cancer; sarcoid-like granuloma; immunohistochemistry AB An unusual granulomatous reaction within a conventional clear cell renal cancer in a 62 year-old woman is reported. Using immunohistochemical evaluation, cells of the granuloma were CD68 (Kp1), carboxypeptidase M and CD3 positive. No signs of sarcoidosis were found in other organs. According to the few publications that mention cancer associated sarcoid-like reaction, such lesions do not influence the prognosis. Our patient is still well for a 15 months follow-up. C1 [Kovacs, Judit] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98., H-4012 Debrecen, Hungary. [Varga, Attila] University of Debrecen, Medical and Health Science Center, Department of UrologyDebrecen, Hungary. [Bessenyei, Maria] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98., H-4012 Debrecen, Hungary. [Gomba, Szabolcs] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98., H-4012 Debrecen, Hungary. RP Kovacs, J (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, H-4012 Debrecen, Hungary. CR Marinides GN, Hajdu I, Gand RO: A unique association of renal cell carcinoma with sarcoid reaction in the kidney. Nephron 67:477-480, 1994 Ryska A, Seifert G: Adenolymphoma, Whartin’s tumor, with multiple sarcoid-like granulomas. Pathol Res Pract 195:835- 839, 1999 Ludvikova M, Ryska A and Drovakova E: Focal sarcoid-like change of the thyroid gland. A possible consequence of aspiration cytology? Pathol Res Pract 198:479-482, 2002 Moder KG, Litin SC and Gaffey TA: Renal cell carcinoma associated with sarcoid-like tissue reaction. Mayo Clin Proc 65:1498-1501, 1990 Bottone AC, Labarbera M, Asasourian A et al: Renal sarcoidosis coexisting with hypernephroma. Urology 41:157-159, 1993 Campbell F and Douglas-Jones AG: Sarcoid-like granulomas in primary renal cell carcinoma. Sarcoidosis 10:128-131, 1992 Lucci S, Rivolta R and Fazi M: Sarcoidosis and clear cell carcinoma of the kidney: the sixth case? G Chir 23:75-78, 2002 Shigematsu H, Kurita A, Omura Y et al: Gastric cancer with sarcoid reactions in the regional lymphnodes, the stomach and the splenic parenchyma. Report of a case. Surgery Today- Tokyo 29:549-552, 1999 Kamiyoshihara M, Hirai T and Kawashima O: Sarcoid reactions in primary pulmonary carcinoma. Report of seven cases. Oncology Reports 5:177-180, 1998 Bassler K and Birke F: Histopathology of tumor associated sarcoid- like stromal reaction in breast cancer. An analysis of 5 cases with immunohistochemical investigation. Virchows Arch A Pathol Anat Histopathol 412:231-239, 1988 Rayson D, Burch PA and Richardson RL: Sarcoidosis and testicular carcinoma. Cancer 83:337-343, 1998 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2004 VL 10 IS 3 BP 169 EP 171 PG 3 ER PT J AU Brittig, F Weinkauf, P Karchner, H AF Brittig, Ferenc Weinkauf, Philip Karchner, Hannelore TI Ideal Cooling Process for Paraffin-Embedded Tissues SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE sectioning; paraffin block; cooling technique ID sectioning; paraffin block; cooling technique AB Back in the seventies everybody was convinced that it was no longer necessary to cool paraffinembedded tissues, because of the new advances in the production of paraffin. The reason for this assumption was the addition of plastic polymers and dimethyl sulfoxide. The quality of tissue sectioning improved because of these additives. The daily routine in the histology laboratories shows that it is impossible to produce good quality cutting without cooling. Cooling the paraffin by means of cooling plates or ice dishes creates drastic improvement; the cutting quality improves and it is much easier. Both improvements speed up the process and save time for the technicians and the physician. Long-term study indicates that not only the temperature but also the methodology of cooling and the cooling rate are playing a decisive role in the cutting quality. C1 [Brittig, Ferenc] County Hospital, Department of Pathology, H-8200 Veszprem, Hungary. [Weinkauf, Philip] Design engineerVeszprem, Hungary. [Karchner, Hannelore] Design engineerVeszprem, Hungary. RP Brittig, F (reprint author), County Hospital, Department of Pathology, H-8200 Veszprem, Hungary. CR Lamb RA: Waxes for Histology, Histopathology), selected topics, Ed.: H. C. Cook. Lailliere Tindall, London, 1973 Arnold M: Histochemie, Leydenfrostisches Phanomen, Springer Verlag, 1968 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2004 VL 10 IS 3 BP 172 EP 173 PG 2 ER PT J AU Sinkovics, GJ AF Sinkovics, G Joseph TI Chondrosarcoma Cell Differentiation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE chondrocytes; chondrosarcoma cells; immune T cell; NK cell; cytotoxic lymphocytes; tumor cell differentiation; telomerase ID chondrocytes; chondrosarcoma cells; immune T cell; NK cell; cytotoxic lymphocytes; tumor cell differentiation; telomerase AB A mixed population of lymphocytes from a healthy donor co-existed with an established culture of allogeneic chondrosarcoma cells, during which time the tumor cells changed from malignantly transformed to benign fibroblast-like morphology; from multilayered to a monolayered growth pattern; lost their potency to grow in colonies in soft agar; and showed signs of senescence. A discussion of possible molecular mechanisms for this event is offered. If there are as yet undiscovered lymphokines that can induce reversal of the malignant geno/phenotype, the cognate gene(s) should be cloned for genetic engineering and for the mass production of the corresponding molecular mediators for clinical trials. C1 [Sinkovics, G Joseph] The University of South Florida College of Medicine, Departments of Medicine and Medical Microbiology-Immunology, Dr Martin Luther King Jr Blvd, 3001 W Tampa, FL, USA. RP Sinkovics, GJ (reprint author), The University of South Florida College of Medicine, Departments of Medicine and Medical Microbiology-Immunology, 3001 W Tampa, USA. CR Adams CS, Shapiro IM: The fate of the terminally differentiated chondrocyte: evidence for microenvironmental regulation of chondrocyte apoptosis. Crit Rev Oral Biol Med 13:465-473, 2002. Aigner T, Loos S, Muller S et al.: Cell differentiation and matrix gene expression in mesenchymal chondrosarcomas. Am J Pathol 156:1327-1335, 2000. Aigner, T., Dertinger, S., Belke, J., Kirchner, T: Chondrocytic cell differentiation in clear cell chondrosarcoma. Hum Pathol 27:1301-1305,1996 Aigner T, Unni KK: Is dedifferentiated chondrosarcoma a ‘dedifferentiated’ chondrosarcoma? J Pathol 189:454-462, 1999. Aikawa T, Segre GV, Lee K: Fibroblast growth factor inhibits chondrocyte growth through induction of p21 and subsequent inactivation of cycline E-cdk2. J Biol Chem 275:29347- 29352, 2001. Akiyama B, Chaboissier MC, Martin JF et al: The transcription factor Sox9 has essential roles in successive steps of the chondrocyte differentiation pathway and is required for expression of Sox5 and Sox6. Genes Dev 16:2813-2828, 2002. Algros MP, Collonge-Rame MA, Bedgejian T et al: Neuroectodermal differentiation of extraskeletal myxoid chondrosarcoma: a classical feature? Ann Pathol 23:244-248, 2003. Amling M, Posl M, Hentz M et al: PTHrP and Bcl-2: essential regulatory molecules in chondrocyte differentiation and chondrogenic tumors. Verh Dtsch Ges Pathol 82:160-169, 1998. Archer C W, Francis-West P: The chondrocyte. Int J Biochem Cell Biol 35:401-404, 2003. Aszodi A, Hunziker EB, Brakebusch C, Fassler R: b1 Integrins regulate chondrocyte rotation, GH1 progression and cytokinesis. Genes Dev 17:2465-2479, 2003. Beier F, Lee RJ, Taylor AC, et al: Identification of the cyclin D1 gene as a target of activating transcription factor 2 in chondrocytes. Proc Natl Acad Sci USA 96:1433-1438, 1999. Benz K, Breit S, Lukoschek M, et al: Molecular analysis of expansion, differentiation, and growth factor treatment of human chondrocytes identifies differentiation markers and growth-related genes. Biochem Biophys Res Commun 293:284-292, 2002. Blasenbreu S, Baretton GB, Bender C et al: TP53 gene aberrations in chondromatous neoplasms: correlation with immunohistochemical p53 accumulation and MDM2 expression. Verh Dtsch Ges Pathol 82:284-289, 1998. Bovee JV, van den Broek LJ, Cleton-Jansen AM, Hogendoorn PC: Up-regulation of PTHrP and Bcl-2 expression characterizes the progression of osteochondroma toward peripheral chondrosarcoma and is a late event in central chondrosarcoma. Lab Invest 80:1925-1934, 2000. Bovee JVMG, Cleton-Jansen A-M, Wuyts W et al: EXT-mutation analysis and loss of heterozygosity in sporadic and hereditary osteochondromas and secondary chondrosarcomas. Am J Hum Genet 65:689-698, 1999. Campi G, Crosti M, Consogno G et al: CD4+ T cells from healthy subjects and colon cancer patients recognize a carcinoembryonic antigen-specific immunodominant epitope. Cancer Res 63:8481- 8486, 2003. Casorzo L, Chiecchio L, Pisacane A et al: Cytogenetic findings in a case of dedifferentiated chondrosarcoma. Cancer Genet Cytogenet 144:61-64, 2003. Chang SH, Oh CD, Yang MS et al: Protein kinase C regulates chondrogenesis of mesenchymes via mitogen-activated protein kinase signaling. J Biol Chem 273:19213-19219, 1998. Church V, Nohno T, Linker C, et al: Wnt regulation of chondrocyte differentiation. J Cell Sci 115:4809-4818, 2002. Cormier S, Delezoide A-L, Benoist-Lasselin C et al: Parathyroid hormone receptor type 1/indian hedgehog expression is preserved in the growth plate of human fetuses affected with fibroblast growth factor type 3 activating mutations. Am J Pathol 161:1325-1335, 2002. Cunha GR, Hayward SW, Wang YZ, Ricke WA: Role of the stromal microenvironment in carcinogenesis of the prostate. Int J Cancer 107:1-10, 2003. Dailey L, Laplantine E, Priore R, Basilico C: A network of transcriptional and signaling events is activated by FGF to induce chondrocyte growth arrest and differentiation. J Cell Biol 161:1053-1066, 2003. Deckelbaum RA, Chan G, Miao D et al: Ihh enhances differentiation of CFK-2 chondrocytic cells and antagonizes PTHrP-mediated activation of PKA. J Cell Sci 115:3015-3025, 2002. Domanski HA, Carlen B, Martens F, Akerman M: Extraskeletal myxoid chondrosarcoma with neuroendocrine differentiation: a case report with fine-needle aspiration biopsy, histopathology, electron microscopy, and cytogenetics. Ultrastruct Pathol 27: 363-368, 2003. Eguchi T, Kubota S, Kondo S et al: Regulatory mechanism of human connective tissue growth factor, CTGF/Hcs24, gene expression in a human chondrocytic cell, HCS-2/8. J Biochem 130:79-87, 2001. Ellsworth JL, Berry J, Bukowski T et al: Fibroblast growth factor- 18 is a trophic factor for mature chondrocytes and their progenitors. Osteoarthr Cartil 10:308-320, 2002. Enomoto-Iwamoto M, Kitagaki J, Koyama E et al: The Wnt antagonist Frzb-1 regulates chondrocyte maturation and long bone development during limb skeletogenesis. Dev Biol 251:142-156, 2002. Ferrari D, Kosher RA: D1x5 is a positive regulator of chondrocyte differentiation during enchondral ossification. Dev Biol 252:257-270, 2002. Ghert MA, Qi WN, Erickson HP et al:Tenascin-C expression and distribution in cultured human chondrocytes and chondrosarcoma cells. J Orthop Res 20:834-841, 2002. Grimaud E, Heymann D, Redini, F: Recent advances in TGF-b effects on chondrocyte metabolism. Potential therapeutic roles of TGF-b in cartilage disorders. Cytokine Growth Factor Rev 13:241-257, 2002. Grube M, Rezvani H, Sconocchia G et al: Autoreactive, cytotoxic T lymphocytes specific for peptides derived from normal Bcell differentiation antigens in healthy individuals and patients with B-cell malignancies. Clin Cancer Res 10: 1047-1056, 2004. Guicheux J, Palmer G, Relic B et al: Primary human articular chondrocytes, dedifferentiated chondrocytes, and synoviocytes exhibit differential responsiveness to interleukin-4: correlation with the expression pattern of the common receptor gamma chain. J Cell Physiol 192: 93-101, 2002. Guo W, Gorlick R, Ladanyi M et al: Expression of bone morphogenetic proteins and receptors in sarcomas. Clin Orthop 365:175- 183, 1999. Hatakeyama Y, Nguyen J, Wang X, et al: Smad signaling in mesenchymal and chondroprogenitor cells. J Bone Joint Surg Am 3:13-18, 2003. Hecht JT, Hogue D, Wang Y et al: Hereditary multiple exostoses, EXT): mutational studies of familial EXT1 cases and EXT-associated malignancies. Am J Hum Genet 60:80-86, 1997. Hiraoka K, Zenmyo M, Komiya S et al: Relationship of p21, waf1/cip1, and differentiation in chondosarcoma cells. Virchows Arch 440:285-290, 2002. Hoang MP, Suarez PA, Donner LR et al: Mesenchymal chondrosarcoma: a small cell neoplasm with polyphenotypic differentiation. Int J Surg Pathol 8:291-301, 2000. Hoodless PA, Haerry T, Abdollah S et al: MADR1, a MAD-related protein that functions in BMP2 signaling pathways. Cell 85:489-500, 1996. Huh YH, Kim SH, Kim SJ, Chun JS: Differentiation status-dependent regulation of cyclooxygenase-2 expression and prostaglandin E2 production by epidermal growth factor via mitogen-activated protein kinase in articular chondrocytes. J Biol Chem 278:9691-9697, 2003. Ionescu AM, Schwartz EM, Zuscik MJ et al: ATF-2 cooperates with Smad3 to mediate TGFbeffects on chondrocyte maturation. Exp Cell Res 288:198-207, 2003. Ivkovic S, Yoon BS, Popoff SN et al: Connective tissue growth factor coordinates chondrogenesis and angiogenesis during skeletal development. Development 130: 2779-2791, 2003. Kalil RK, Inwards CY, Unni KK et al: Dedifferentiated clear cell chondrosarcoma. Am J Surg Pathol 2:1079-1086, 2000. Kato Y, Iwamoto M: Fibroblast growth factor is an inhibior of chondrocyte terminal differentiation. J Biol Chem 265:5903- 5909, 1990. Kawashima H, Ogose A, Hayami T et al: Effect of dexamethasone on growth inhibition and chondrogenesis of human chondrosarcoma. J Orthop Sci 8:341-345, 2003. Kay HD, Cabiness JR, Ervin F et al: Do lymphocytes presensitized to tumor antigens occur in normal individuals? Proc Ann Meeting Am Soc Microbiol p 111, #M227, 1973. Kay HD, Cabiness JR, Kitowski T, Sinkovics JG: Cytotoxicity to, and immune stimulation of, cultured tumor cells by normal human lymphocytes and serum factors. Proceeding 65th AACR 15:64, #254, 1974. Kay HD, Sinkovics JG, Cabiness JR et al: The concept of immune surveillance and its experimental testing. Clin Res 22/1: 44A, 1974. Kay HD, Sinkovics JG: Cytotoxic lymphocytes from normal donors. Lancet 2: 296-297, 1974. Kay HD, Thota H, Romero JJ, Sinkovics JG: Lymphocytes from normal donors are frequently cytotoxic to tumor cells in vitro. Clin Res 23/3:340A, 1975. Kenny PC, Bissell MJ:Tumor reversion: Correction of malignant behavior by microenvironmental cues. Int J Cancer 107:688-695, 2003. Kim DW, Lassar AB: Smad-dependent recruitment of a histone de-acetylase/Sin3A complex modulates the bone morphogenetic protein-dependent transcriptional repressor activity of Nkx3.2. Mol Cell Biol 23:8704-8717, 2003. Kim SJ, Chun JS: Protein kinase Ca and x regulate oxideinduced NFeB activation that mediates cyclooxygenase-2 expression and apoptosis but not dedifferentiation in articular chondrocytes. Biochem Biophys Res Commun 303:206-211, 2003. Kim SJ, Hwang SG, Kim IC, Chun JS: Actin cytoskeletal architecture regulates nitric oxide-induced apoptosis, dedifferentiation and cyclooxygenase-2 expression in articular chondrocytes via mitogen-avtivated protein kinase and protein kinase C pathways. J Biol Chem 278:42448-42456, 2003. Kim SJ, Kim HG, Oh CD et al: p38 kinase-dependent and –independent inhibition of protein kinase Cx and a regulates nitric oxide-induced apoptosis and dedifferentiation of articular chondrocytes. J Biol Chem 277:30375-30381, 2002. Kinisada T, Miyakazi M, Mihara K et al: A new human chondrosarcoma cell line, OUMS-27, that maintains chondrocytic differentiation. Int J Cancer 77:854-859, 1998. Kobayashi T, Chung, UI, Schipani, E et al: PTHrP and indian hedgehog control differentiation of growth plate chondrocytes at multiple steps. Development 129:2977-2986, 2002. Koshizuka Y, Yamada T, Hoshi K et al: Cystatin 10, a novel chondrocyte- specific protein, may promote the last steps of the chondrocyte differentiation pathway. J Biol Chem 278:48259-48266, 2003. Kunisada T, Moseley JM, Slavin Jl et al: Co-expression of parathyroid hormone-related protein, PTHrP, and PTH/PTHrP receptor in cartilaginous tumours: a marker for malignancy? Pathology 34:133-137, 2002. Ladanyi A, Somlai B, Gilde K et al: T-cell activation marker expression on tumor-infiltrating lymphocytes as prognostic factor in cutaneous malignant melanoma. Clin Cancer Res 15:521- 530, 2004. Li N, Shen LH, Zhu QF: Overexpression of c-erb-2 proto-oncogene product in chondrosarcoma. Zhoghua Bing Li Xue Za Zhi 23:37-39, 1994. Li X, Ionescu AM, Schwartz EM et al: Smad6 is induced by BMP-2 and modulates chondrocyte differentiation. J Orthop Res 21:908-913, 2003. Liu Z, Xu J, Colvin JS, Ortiz DM: Coordination of chondrogenesis and osteogenesis by fibroblast growth factor 18. Genes Dev 16:859-869, 2002. Loeser RF, Pacione CA, Chubinskaya S: The combination of insulin-like growth factor 1 and osteogenic protein 1 promotes increased survival of, and matrix synthesis by, normal osteoarthritic human articular chondrocytes. Arth Rheum 48:2188-2196, 2003. Longobardi L, Torello M, Buckay C et al: A novel insulin-like growth factor, IGF)-independent role for IGF binding protein-3 in mesenchymal chondroprogenitor cell apoptosis. Endocrinol 144:1695-1702, 2003. Maclean HE, Kim JI, Glimcher MKJ et al: Absence of transcription factor c-maf causes abnormal terminal differentiation of hypertrophic chondrocytes during enchondral bone development. Dev Biol 262:51-63, 2003. Martin JA, Buckwalter JA: Human chondrocyte senescence and osteoarthritis. Biorheology 39:145-152, 2002. Merz D, Liu R, Johnson K, Terkeltaub R: IL-8/CXCL8 and growth-related oncogene a/CXCL1 induce chondrocyte hypertrophic differentiation. J Immunol 171:4406-4415, 2003. Minina E, Kreschel C, Naski MC, et al: Interaction of FGF, Ihh/PTHrP and BMP signaling integrates chondrocyte proliferation and hypertrophic differentiation. Dev Cell 3:439-449, 2002. Miyamoto M, Ito H, Mukai S, Kobayashi T et al: Simultaneous stimulation of EP2 and EP4 is essential to the effect of prostaglandin E2 in chondrocyte differentiation. Osteoarthr Cartil 11:644-652, 2003. Nagorsen D, Scheibenbogen C, Marincola FM, Letsch, A, Keilholz U: Natural T cell immunity against cancer. Clin Cancer Res 9:4296-4303, 2003. Nakanishi T, Nishida T, Shimo T et al: Effects of CTGF/Hcs24, a product of a hypertrophic chondrocyte-specific gene, on the proliferation and differentiation of chondrocytes in culture. Endocrinology 141:264-273, 2000. Nielsen GP, Keel SB, Dickersin GR et al: Chondromyxoid fibroma: a tumor showing myofibroblastic, myochondroblastic, and chondrocytic differentiation. Mod Pathol 12:514-517, 1999. Nishida T, Kubota S, Nakanishi T et al: CTGF/Hcs24, a hypertrophic chondrocyte-specific gene product, stimulates proliferation and differentiation, but not hypertrophy of cultured articular chondrocytes. J Cell Physiol 192:55-63, 2002. O’Donovan M, Russel JM, O’Leary JJ et al: Abl expression, tumor grade, and apoptosis in chondrosarcoma. Mol Pathol 52:341-344, 1999. Oh CD, Chun JS:Signaling mechanisms leading to the regulation of differentiation and apoptosis of articular chondrocytes by insulin-like growth factor-1. J Biol Chem 278:36563-36571, 2003. Ohbayashi N, Shibayama M, Kurotaki Y et al: FGF18 is required for normal cell proliferation and differentiation during osteogenesis and chondrogenesis. Genes Dev 16:870-879, 2002. Park H-R, Kim Y-W, Jung WW et al: Evaluation of HER-2/neu status by real-time quantitative PCR in malignant cartilaginous tumors. Int J Oncol 24:575-580, 2004. Raucci A, Laplantine E, Mansukhani A, Basilico C:Activation of the ERK1/2 and p38 mitogen-activated protein kinase pathways mediates fibroblast growth factor-induced growth arrest of chondrocytes. J Biol Chem 279:1747-1756, 2004 Raulet DH: Chapter 12. Natural killer cells. In: Fundamental Immunology. W. E. Paul, editor. 5th edition. Lippincott Williams & Wilkins, Philadelphia, PA. Pp 365-391, 2003. Reguly T, Wrana JL: In or out? The dynamics of Smad nucleocytoplasmic shuttling. Trends Cell Biol 13:216-220, 2003. Reiter I, Tzukerman M, Maor G: Spontaneously differentiating primary chondrocytic tissue culture; a model for enchondral ossification. Bone 31:333-339, 2002. Rozenblatt-Rosen O, Mosonego-Ornan E, Sadot E et al: Induction of chondrocyte growth arrest by FGF: transcriptional and cytoskeletal alterations. J Cell Sci 115:553-562, 2002. Schaefer JF, Millham M, de Crombrugghe B, Buckbinder L: FGF signaling antagonizes cytokine-mediated repression of Sox9 in SW1353 chondrosarcoma cells. Osteoarthr Cartil 11:233-241, 2003. Schuerwegh AJ, Dombrecht EJ, Stevens WJ et al: Influence of pro-inflammatory IL-1a, IL-6, TNFa, IFNg and anti-inflammatory, IL-4, cytokines on chondrocyte function. Osteoarth Cartil 11: 681-687, 2003. Schulze-Tanzil G, de Souza P, Villegas Castrejon H et al: Redifferentiation of dedifferentiated human chondrocytes in high density cultures. Cell Tissue Res 308:371-379, 2002. Segat D, Comai R, Di Marco E et al: Integrins a6Ab1 and a6Bb1 promote different stages of chondrogenic cell differentiation. J Biol Chem 277:31612-31622, 2002. Shakunaga T, Ozaki T, Ohara N et al: Expression of connective tissue growth factor in cartilaginous tumors. Cancer 89:1466- 1473, 2000. Sims WW, Ordonez NG, Johnston D et al: p53 expression in dedifferentiated chondrosarcoma. Cancer 76:223-227, 1995. Sinkovics JG: Acquisition of resistance by human tumor cells to lymphocyte-mediated cytotoxicity. Proc AACR 17: 99, #393, 1976. Sinkovics JG: Cytotoxic lymphocytes. Ann Clin Lab Sci 16:488- 496, 1986. Sinkovics JG: Interferon. In: Medical Oncology, an Advanced Course. 2nd edition, vol. I & II. Sinkovics JG, author. Marcel Dekker, New York, N. Y. Pp 1455, 1986. Sinkovics JG: A reappraisal of cytotoxic lymphocytes in human tumor immunology. In: Cancer Biology and Therapeutics, ed. Cory JG, Szentivanyi A, Plenum Press, New York, N.Y., pp 225- 253, 1987. Sinkovics JG: Oncogenes and growth factors. CRC Crit Rev Immunol 8:217-298, 1988. Sinkovics JG: Malignant lymphoma arising from natural killer cells: report of the first case in 1970 and newer developments in the FasL®FasR system. Acta Microbiol Immunol Hung 44:295- 307, 1997 Sinkovics JG: New biological therapeutics: competitors or collaborators of the viral therapy for human cancers. In: Viral Therapy of Human Cancers. Horvath JC, Sinkovics JG, editors. Marcel Dekker, New York, N.Y., in print, 2004. Sinkovics JG, Cabiness JR, Shullenberger CC: Monitoring in vitro of immune reactions to solid tumors. Front Rad Ther Oncol 7:141-154, 1972. Sinkovics JG, Dreyer DA, Shirato E et al: Cytotoxic lymphocytes. I. Destruction of neoplastic cells by lymphocytes in cultures of human origin. Texas Rep Biol Med 29:227-242, 1971. Sinkovics JG, Gyorkey F, Kusyk C, Siciliano MJ: Growth of human tumor cells in established cultures. Methods Cancer Res 14:243-323, 1978. Sinkovics J, Gyorkey F, Shullenberger CC: Project M23/gm7. The etiologic agents, immunology and spread of human leukemia and sarcoma. In: Research Report. The University of Texas System Cancer Center M. D. Anderson Hospital and Tumor Institute, Houston, TX. Printing Division, The University of Texas, Austin, TX. Pp 321-322, 1978. Sinkovics JG, Horvath JC: Virological and immunological connotations of apoptotic and anti-apoptotic forces in neoplasia. Int J Oncol 19:473-488, 2001. Sinkovics JG, Shirato E, Cabiness JR, Martin RG: Rhabdomyosarcoma after puberty: clinical, tissue culture and immunological studies. J Med Exp Clin 1:313-326, 1970. Sinkovics JG, Shirato E, Gyorkey F et al: Relationship between lymphoid neoplasms and immunologic functions. In: Leukemia- Lymphoma. 14th Annual Clinical Conference, The University of Texas M. D. Anderson Hospital and Tumor Institute, Houston, TX, 1969. Year Book, Medical Publishers, Chicago, IL. Pp 53- 92, 1970. Sinkovics JG, Shirato E, Martin RG, et al: Chondrosarcoma. Immune reactions of a patient to autologous tumor. Cancer 27:782-793, 1971. Sinkovics JG, Shirato E, Martin RG, White EC: Chondrosarcoma. I. A brief review of eighty-three patients. J Med Exp Clin 1:15-25, 1970. Sinkovics JG, Tebbi K, Cabiness JR: Cytotoxicity of lymphocytes to established cultures of human tumors: evidences for specificity. Natl Cancer Inst Monogr 37:9-18, 1973. Sinkovics JG, Thota H, Romero JJ, Waldinger R: Bone sarcomas: etiology and immunology. Can J Surg 20:494-503, 1977. Soderstrom M, Bohling T, Ekfors T et al: Molecular profiling of human chondrosarcomas for matrix production and cancer markers. Int J Cancer 100:144-151, 2002. St-Jacques B, Hammerschmidt M, McMahon RT: Indian hedgehog signaling regulates proliferation and differentiation of chondrocytes and is essential for bone formation. J Cell Sci 110:2691- 2701, 1997. Stanton LA, Underhill TM, Beier F: MAP kinases in chondrocyte differentiation. Dev Biol 263:165-175, 2003. Stheneur C, Dumontier MF, Guedes C et al: Basic fibroblast growth factor as a selective inducer of matrix G1a protein gene expression in proliferative chondrocytes. Biochem J 369: 63-79, 2003. Sitcheran R, Cogswell PC, Baldwin Jr AS: NFeB mediates inhibition of mesenchymal cell differentiation through a posttranscriptional gene silencing mechanism. Genes Dev 17:2368-2373, 2003. Takigawa M, Okawa T, Pan H et al: Insulin-like growth factors I and II are autocrine factors stimulating proteoglycan synthesis, a marker of differeniated chondrocytes, acting through their respective receptors on a clonal human chondrosarcoma-derived chondrocyte cell line, HCS-2/8. Endocrinology 138:4390-4400, 1997. Tarkkanen M, Wiklund T, Virolainen M et al: Dedifferentiated chondrosarcoma with t(9;22)(q34;q11-12). Genes Chromosomes Cancer 9:136-140, 1994. Tebbi CK: Purification of lymphocytes: a new technique. Lancet 1:1392, 1973. Tuli R, Tuli S, Nandi S et al: Transforming growth factor b-mediated chondrogenesis of human mesenchymal progenitor cells involves N-cadherin and mitogen-activated protein kinase and Wnt signaling cross-talk. J Biol Chem 278:41227-41236, 2003. Uyama Y, Yagami K, Hatori et al: Recombinant human bone morphogenetic protein-2 promotes Indian hedgehog-mediated osteochondrogenic differentiation of a human chondrocytic cell line in vivo and in vitro. Differentiation 72:32-40, 2004. Valcourt U, Gouttenoire J, Moustakas A, et al.: Functions of transforming growth factor type I receptors and Smad proteins in the hypertrophic maturation and osteoblastic differentiation of chondrocytes. J Biol Chem 277:33545-33558, 2002. Vonderheide RH, Domchek SM, Schultze JL et al: Vaccination of cancer patients against telomerase induces functional antitumor CD8+ T lymphocytes. Clin Cancer Res 10:828-839, 2004. Wehrli BM, Huang W, de Crombrugghe D, et al. Sox9, a master regulator of chondrogenesis, distinguishes mesenchymal chondrosarcoma from small blue round cell tumors. Hum Pathol 34: 263-269, 2003. Wuyts W, Van Hul W, De Boulle K et al: Mutations in the EXT1 and EXT2 genes in hereditary multiple exostoses. Am J Hum Genet 62:346-354, 1998. Yang Y, Topol L, Lee H, Wu J: Wnt5a and Wnt5b exhibit distinct activities in coordinating chondrocyte proliferation and differentiation. Development 130:1003-1015, 2003. Yates KE, Forbes RL, Glowacki J: New chondrocyte genes discovered by representational difference analysis of chondroinduced human fibroblasts. Cells Tissues Organs 176:41-53, 2004. Yosimichi G, Nakanishi T, Nishida T et al: CTGF/Hcs24 induces chondrocyte differentiation through a p38 mitogen-activated protein kinase, p38MAPK), and proliferation through a p44/42MAPK/extracellular signal-regulated kinase. Eur J Biochem 268:6058-6065, 2001. Zhang L, Duan, CJ, Binkley C et al: A transforming growth factor b-induced Smad3/Smad4 complex directly activates protein kinase A. Mol Cell Biol 24:2169-2180, 2004. Zwaveling S, Vierboom MP, Ferreira M et al: Antitumor efficacy of wild-type p53-specific CD4+ T-helper cells. Cancer Res 62:6187-6193, 2002. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2004 VL 10 IS 3 BP 174 EP 187 PG 14 ER PT J AU Gundy, S Babosa, M Baki, M Bodrogi, I AF Gundy, Sarolta Babosa, Maria Baki, Marta Bodrogi, Istvan TI Increased Predisposition to Cancer in Brothers and Offspring of Testicular Tumor Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE testicular cancer; familial cancer risk; childhood tumors ID testicular cancer; familial cancer risk; childhood tumors AB Cancer susceptibility was examined in first-degree relatives of 293 testicular tumor patients (TTPs) and 586 age-matched healthy males. Significantly increased risk was found in the families of TTPs (OR: 1.4; CI: 1.08-1.79), however, except for testicular cancer of 7 brothers (OR: 11.7; CI: 1.42-256.5), and 6 various childhood tumors (bilateral Wilms' tumor, neuroblastoma, medulloblastoma, ALL, histiocytosis-X, testicular tumor) of 200 offspring (OR: 12.9; CI: 1.54-286.2), no association with other malignancies was observed. No differences were seen between the fertility of patients and controls when occupational or socio-economic status of the families was taken into account. However, the majority of the controls (85%) fathered the first child between 20-30 years of age, while only 61% of TTPs had the first child in the same age group. TTPs fathered more girls than boys (P=0.009), and the lower male - higher female ratio of index children was also identical, irrespective of the conception taking place before or after the father’s treatment. Occupations did not, but smoking might have influenced cancer susceptibility of the patients. Aggregation of fraternal testicular tumors, and both dramatically increased cancer risk and altered sex ratio of the offspring indicate a remarkable role of hereditary factors in tumorigenesis and later consequences of a certain portion of testicular malignancies, which must be refined by molecular studies. C1 [Gundy, Sarolta] National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, Rath Gy. u. 7-9., H-1122 Budapest, Hungary. [Babosa, Maria] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Baki, Marta] National Institute of Oncology, Department of Chemotherapy and ClinicopharmacologyBudapest, Hungary. [Bodrogi, Istvan] National Institute of Oncology, Department of Chemotherapy and ClinicopharmacologyBudapest, Hungary. RP Gundy, S (reprint author), National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, H-1122 Budapest, Hungary. EM gundy@oncol.hu CR Hoff Wanderas E, Tretli S, Fossa SD: Trends in incidence of testicular cancer in Norway 1955-1992. Eur J Cancer 31A: 2044-2048, 1995 Bergstrom R, Adami HO, Mohner M, et al: Increase in testicular cancer incidence in six European countries: a birth cohort phenomenon. J Natl Cancer Inst 88: 727-733, 1996 Heimdal K, Olsson H, Tretli S, et al: Familial testicular cancer in Norway and southern Sweden. Br J Cancer 73: 964-969, 1996 Sonnenveld DJA, Sleijfer DTh, Schrafford Koops H, et al: Familial testicular cancer in a single-centre population. Eur J Cancer 35:1368-1373, 1999 Dong C, Lonnstedt I, Hemminki K: Familial testicular cancer and second primary cancers in testicular cancer patients by histological type. Eur J Cancer 37: 1878-1885, 2001 Spermon JR, Witjes JA, Nap M, Kiemeney LALM: Cancer incidence in relatives of patients with testicular cancer in the Eastern part of Netherlands. Urology 57: 747-752, 2001 Rapley EA, Crockford GP, Easton DF, et al: Localisation of susceptibility genes for familial testicular cancer. APMIS 111: 128-135, 2001 Buetow SA: Epidemiology of testicular cancer. Epidem Rev 17: 433-449, 1995 Bokemeyer C, Schmoll H-J, Schoffsky P, et al: Bilateral testicular tumors: Prevalence and clinical implications. Eur J Cancer 29A: 874-876, 1993 Geczi L, Gomez F, Baki M, Bodrogi I: The incidence, prognosis, clinical and histological characteristics, treatment, and outcome of patients with bilateral germ cell testicular cancer in Hungary. J Cancer Res Clin Oncol 129:309-315, 2003 Nicholson PW, Harland SJ: Inheritance and testicular cancer. Br J Cancer 71:421-426, 1995 Khoury MJ, Beaty TH, Liang KY: Can familial aggregation of disease be explained by familial aggregation of environmental risk factors? Am J Epidemiol 127: 674-683, 1988 Swerdlow AJ, De Stavola BL, Swanwick MA, et al: Risk factors for testicular cancer: a case-control study in twins. Br J Cancer 80: 1098-1102, 1999. Huyghe E, Matsuda T, Thonneau, P: Increasing incidence of testicular cancer worldwide: a review. J Urol 170: 5-11, 2003 Swerdlow AJ, Douglas AJ, Huttly SRA, Smith PG: Cancer of the testis, socioeconomic status, and occupation. Br J Ind Med 48: 670-674, 1991 “GraphPad Software, InStat guide to choosing and interpreting statistical tests, 1998, GraphPad Software, Inc., San Diego California USA, www.graphpad.com Gundy S, Babosa M, Baki M. Offspring of testicular cancer patients: A model for male-mediated mutagenesis and carcinogenesis, In: Reproductive Toxicology., Ed: Richardson M), Weinheim, New York, Basel, Cambridge, Tokyo, VCH, 1993, pp. 143-151 Babosa M, Baki M, Bodrogi I, Gundy S: A study of children fathered by men treated for testicular cancer, conceived before, during and after chemotherapy. Med Ped Oncol 22: 33-38, 1994 Senturia YD, Peckham CS, Peckham MJ: Children fathered by men treated for testicular cancer. Lancet 2: 766-769, 1985 Fried P, Steinfeld R, Casileth B, Steinfeld A: Incidence of developmental handicaps among the offspring of men treated for testicular seminoma. Int J Androl 10: 385-387, 1987 Heimdal K, Olsson H, Tretli S, et al: Risk of cancer in relatives of testicular cancer patients. Br J Cancer 73: 970-973, 1996 Hemminki K, Vaittinen P: Effect of paternal and maternal cancer on cancer in the offspring: a population-based study. Cancer Epidem Biom Prev 6: 993-997, 1997 Olsen JH, Boice JD, Seersholm N, et al: Cancer in the parents of children with cancer. New Eng J Med 333:1594-1599, 1995 Moller H: Clues to the aetiology of testicular germ cell tumors from descriptive epidemiology. Eur Urol 233: 8-13, 1993 Chaganti RSK, Houldsworth J: Genetics and biology of adult human male germ cell tumors. Cancer Res 60: 1475-1482, 2000 Kreidberg JA, Sariola H, Loring JM, et al: WT-1 is required for early kidney development. Cell 74: 679-691, 1993 Chompret A: The Li-Fraumeni syndrome. Biochimie 84: 75- 82, 2000 Knudson AGJ, Strong LC: Mutation and cancer: neuroblastoma and pheochromocytoma. Am J Hum Genet 24: 514-522, 1972 Kim E, Nichols KE, Malkin D, et al: Germ-line p53 mutations predispose to a wide spectrum of early-onset cancers. Cancer Epidem Biom Prev 10: 83-87, 2001 Heimdal K, Lothe RA, Lystad S, et al: No germ line TP53 mutations detected in familial and bilateral testicular cancer. Genes Chromosomes Cancer 6: 92-97, 1993 Jones RH, Vasey P: New directions in testicular cancer; molecular determinants of oncogenesis and treatment success. Eur J Cancer 39: 147-156, 2003 Peltomaki P, Lothe R, Borresen AL, et al: Altered dosage of the sex chromosomes in human testicular cancer: a molecular genetic study. Int J Cancer 47: 518-522, 1991 Moller H: Trends in sex-ratio, testicular cancer and male reproductive hazards: Are they connected? APMIS 106: 232- 239, 1998 Moller H, Skakkebaek NE: Risk of testicular cancer in subfertile men. Br Med J 318: 559-562, 1999 Fossa SD, Kravdal O: Fertility in Norwegian testicular cancer patients. Br J Cancer 82: 737-741, 2000 Demographic Yearbook. Hungarian Central Statistical Office, 2003, Budapest Hemminki K, Vaittinen P, Dong C, Easton D: Sibling risks in cancer: clues to recessive or X-linked genes? Br J Cancer 84: 388-391, 2001 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 197 EP 203 PG 7 ER PT J AU Kida, A Ohashi, K Kobayashii, T Sakai, M Yamashita, T Akiyama, H Kishida, Sh Sakamaki, H AF Kida, Aiko Ohashi, Kazuteru Kobayashii, Takeshi Sakai, Miwa Yamashita, Takuya Akiyama, Hideki Kishida, Shuji Sakamaki, Hisashi TI Incapacitating Lower Limb Pain Syndrome in Cord Blood Stem Cell Transplant Recipients with Calcineurin Inhibitor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE calcineurin inhibitor; lower limb pain; pruritus; cord blood stem cell transplantation ID calcineurin inhibitor; lower limb pain; pruritus; cord blood stem cell transplantation AB Calcineurin-inhibitor induced pain syndrome (CIPS) is a newly described entity with a characteristic feature of sudden onset of severe lower limb pain, and high levels of cyclosporine or tacrolimus may be involved in the pathogenesis. This syndrome is rarely seen in recipients of hematopoietic stem cell transplantation (HSCT) compared with other organ transplant recipients, however, heightened awareness of this complication after HSCT may be needed for hematologists, as misdiagnosis can result in catastrophic consequences. We report herein two cases of lower limb pain syndrome, with some clinical features resembling CIPS, occurring during the early phase of cord blood stem cell transplantation for hematological malignancy. C1 [Kida, Aiko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Ohashi, Kazuteru] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kobayashii, Takeshi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Sakai, Miwa] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Yamashita, Takuya] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Akiyama, Hideki] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kishida, Shuji] Tokyo Metropolitan Komagome Hospital, Neurology DivisionTokyo, Japan. [Sakamaki, Hisashi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. RP Ohashi, K (reprint author), Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 113-8677 Tokyo, Japan. EM k.ohashi@cick.jp CR Kunzendorf U, Brockmoller J, Jochimsen F, et al: Cyclosporin metabolites and central-nervous-system toxicity. Lancet 1: 1223-1223, 1988 Walker RW, Brochstein JA: Neurologic complications of immunosuppressive agents. Neurol Clin 6: 261-278, 1988 Reece DE, Frei-Lahr DA, Shepherd JD, et al: Neurologic complications in allogeneic bone marrow transplant patients receiving cyclosporin. Bone Marrow Transplant 8: 393-401, 1991 Bechstein WO: Neurotoxicity of calcineurin inhibitors: impact and clinical management. Transpl Int 13: 313-326, 2000 Grotz WH, Breitenfeldt MK, Braune SW, et al: Calcineurininhibitor induced pain syndrome, CIPS): a severe disabling complication after organ transplantation. Transpl Int 14: 16-23, 2001 Lucas VP, Ponge TD, Plougastel-Lucas ML, et al: Musculoskeletal pain in renal-transplant recipients. N Engl J Med 325: 1449-1450, 1991 Sanches EN, Criado AB, Guajardo AS, et al: Leg bone pain syndrome due to cyclosporine in a renal transplant patient. Clin Exp Rheum 12: 653-656, 1994 Gauthier VJ, Barbosa LM: Bone pain in transplant recipients responsive to calcium channel blockers. Ann Intern Med 121: 863-865, 1994 Stevens JM, Hilson AJW, Sweny P: Post-renal transplant distal limb bone pain. Transplantation 60: 305-307, 1995 Goffin E, Vande Berg B, Pirson Y, et al: Epiphyseal impaction as a cause of severe osteoarticular pain of lower limbs after renal transplantation. Kidney Int 44: 98-106, 1993 Villaverde V, Cantalejo M, Balsa A, et al: Leg bone pain syndrome in a kidney transplant patient treated with tacrolimus, FK506). Ann Rheum Dis 58: 653-654, 1999 Malat GE, Dupuis RE, Kassman B, et al: Tacrolimus-induced pain syndrome in a pediatric orthotopic liver transplant patient. Pediatr Transplant 6: 435-438, 2002 Blombery PA: A review of reflex sympathetic dystrophy. Aust Fam Physician 24: 1651-1655, 1995 Munoz-Gomez J, Collado A, Gratacos J, et al: Reflex sympathetic dystrophy syndrome of the lower limbs in renal transplant patients treated with cyclosporin A. Arthritis Rheum 34: 625-630, 1991 Shinoda K, Sugiyama E, Taki H, et al: Resting T cells negatively regulate osteoclast generation from peripheral blood monocytes. Bone 33: 711-720, 2003 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 204 EP 206 PG 3 ER PT J AU Surowiak, P Paluchowski, P Wysocka, T Wojnar, A Zabel, M AF Surowiak, Pawel Paluchowski, Piotr Wysocka, Teresa Wojnar, Andrzej Zabel, Maciej TI Steroid Receptor Status, Proliferation and Metallothionein Expression in Primary Invasive Ductal Breast Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; hormone receptor status; metallothionein; Ki-67 ID Breast cancer; hormone receptor status; metallothionein; Ki-67 AB The most important immunocytochemical prognostic and predictive factors in cases of breast cancer include estrogen receptor alpha (ER) and progesterone receptor (PgR). The present study aimed at examining the relationship between the manifestation intensity of proliferation markers (Ki-67 and nucleolar organizer regions - AgNORs) on one hand, and expression of ER and PgR on the other in a uniform group of invasive ductal breast cancers of G2 grade. Moreover, the study aimed at examining the relationship between the above mentioned markers and expression of metallothionein (MT). The studies were performed on samples of invasive ductal breast cancers of G2 grade, originating from 60 females. In paraffin sections originating from the studied cases immunocytochemical reactions were performed using monoclonal antibodies to ER, PgR, Ki-67 and MT, and silver staining was conducted to localize AgNORs. The obtained results were subjected to statistical analysis using Statistica software. Results indicate that manifestation of AgNORs does not correlate with any of the studied antigens (ER, PgR, Ki-67, MT) (p>0.05). Moreover, no relationship could be demonstrated between the intensity of MT expression and proliferation markers or steroid receptor status (p>0.05). A negative correlation was shown between the expression of ER and Ki-67 (p=0.0009). The most intense proliferative activity was demonstrated in cases of breast cancer showing PgR expression but no ER expression (p=0.015), while the lowest proliferative activity was detected in breast cancers with expression of both ER and PgR (p<0.05). C1 [Surowiak, Pawel] Wroclaw Medical University, Department of Human Morphology and Embryology, ul. Chalubinskiego 6a,, 50-356 Wroclaw, Poland. [Paluchowski, Piotr] Regio Hosptital PinnebergPinneberg, Germany. [Wysocka, Teresa] Wroclaw Medical University, Department of Human Morphology and Embryology, ul. Chalubinskiego 6a,, 50-356 Wroclaw, Poland. [Wojnar, Andrzej] Regio Hosptital PinnebergPinneberg, Germany. [Zabel, Maciej] Wroclaw Medical University, Department of Human Morphology and Embryology, ul. Chalubinskiego 6a,, 50-356 Wroclaw, Poland. RP Surowiak, P (reprint author), Wroclaw Medical University, Department of Human Morphology and Embryology, 50-356 Wroclaw, Poland. CR Biesterfeld S, Farokhzad F, Klueppel D, et al: Improvement of breast cancer prognostication using cell kinetic-based silver-stainable nucleolar organizer region quantification of the MIB-1 positive tumor cell compartment. Virchows Arch 438: 478-484, 2001 Clahsen PC, Van de Velde CJH, Duval C, et al: The utility of mitotic index, oestrogen receptor and Ki67 measurements in the creation of novel prognostic indices for node-negative breast cancer. Eur J Surg Oncol 25: 356-363, 1999 Detreb S, King N, Salter J, et al: Immunohistochemical and biochemical analysis of the oestrogen regulated protein pS2, and its relation with oestrogen receptor and progesterone receptor in breast cancer. J Clin Pathol 47: 240-244, 1994 Fitzgibbons PL, Page DL, Weaver D, et al: Prognostic factors in breast cancer. College of American Pathologist consensus statement 1999. Arch Pathol Lab Med 124: 966-978, 2000 Friedline JA, Garret SH, Somji S, et al: Differential expression of the MT-1E gene in estrogen-receptor-positive and -negative human breast cancer cell lines. Am J Pathol 152: 23-27, 1998 Harris H, Henderson RA, Bhat RA, Komm BS: Regulation of metallothionein II messenger ribonucleic acid measures exogenous estrogen receptor beta activity in SAOS-2 and LNCaPLN3 cells. Endocrinology 142: 645-652, 2001 Hishikawa Y, Koji T, Dhar DK, et al: Metallothionein expression correlates with metastatic and proliferative potential in squamous cell carcinoma of the oesophagus. Br J Cancer 81: 712-720, 1999 Ioachim E, Assimakopoulos D, Peschos D, et al: Immunohistochemical expression of metallothionein in benign, premalignant and malignant epithelium of the larynx: correlation with p53 and proliferative cell nuclear antigen. Pathol Res Pract 195: 809-814, 1999 Jensen EV, Cheng G, Palimieri C, et al: Estrogen receptors and proliferation markers in primary and recurrent breast cancer. Proc Natl Acad Sci USA 98: 15197-15202, 2001 Jin R, Bay BH, Chow VT, Tan PH: Metallothionein 1F mRNA expression correlates with histological grade in breast carcinoma. Breast Cancer Res Treat 66: 265-272, 2001 Jin R, Chow VT, Tan PH, et al: Metallothionein 2A expression is associated with cell proliferation in breast cancer. Carcinogenesis 23: 81-86, 2002 Kagi JHR: Overview of metallothionein. Methods Enzymol 205: 613-626, 1991 Keshigan AA, Cnaan A: Estrogen receptor-negative, progesterone receptor-positive breast carcinoma. Poor clinical outcome. Arch Pathol Lab Med 120: 970-973, 1996 Ploton D, Menager M, Jeannesson P, et al: Improvement in the staining and in the visualisation of the argyrophilic proteins of the nucleolar organizer region at the optical level. Histochem J 18: 5-14, 1986 Remmele W, Stegner HE: Recommendation for uniform definition of an immunoreactive score, IRS, for immunohistochemical estrogen receptor detection, ER-ICA, in breast cancer tissue. Pathologe 8: 138-140, 1987 Schmidt CJ, Hamer DH: Cell specificity and an effect of ras on human metallothionein gene expression. Proc Natl Acad Sci USA 83: 3346-3350, 1986 Schwint A, Folco A, Morales A, et al: AgNOR mark epithelial foci in malignant transformation in hamster cheek pouch carcinogenesis. J Oral Pathol Med 25: 20-24, 1996 Surowiak P, Dzizgiel P, Zabel M, et al: Analysis of estrogen receptor, ER, and estrogen-dependent pS2 protein expression in cells of mammary ductal carcinoma. Fol Histochem Cytobiol 39: 145-146, 2001 Zas³awski R, Surowiak P, Dziegiel P, et al: Analysis of the expression of estrogen and progesterone receptors, and of PCNA and Ki67 proliferation antigens, in uterine myomata cells in relation to the phase of the menstrual cycle. Med Sci Monit 7: 784-789, 2001 Zhang R, Zhang H, Wei H, Luo X: Expression of metallothionein in invasive ductal breast cancer in relation to prognosis. J Environ Pathol Toxicol Oncol 19: 95-97, 2000 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 207 EP 211 PG 5 ER PT J AU Wu, D Qiao, Y Kristensen, BG Li, Sh Troen, G Holm, R Nesland, MJ Suo, Z AF Wu, Dan Qiao, Yuhuan Kristensen, B Gunnar Li, Shanshan Troen, Gunhild Holm, Ruth Nesland, M Jahn Suo, Zhenhe TI Prognostic Significance of Dysadherin Expression in Cervical Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE dysadherin; squamous cell cervical carcinoma; immunohistochemistry; RT-PCR; laser capture microdissection ID dysadherin; squamous cell cervical carcinoma; immunohistochemistry; RT-PCR; laser capture microdissection AB The protein and mRNA expression of dysadherin was studied in a series of squamous cell cervical carcinomas, and their clinicopathological associations and prognostic value were explored. Immunohistochemistry was used to assess protein expression of dysadherin in 206 patients with squamous cell cervical carcinoma, FIGO stage Ia-IVb. Frozen tissues from 20 cases in which the tumors showed variable dysadherin protein expression were used for laser capture microdissection (LCM) and processed for RTPCR detection of dysadherin mRNA. Immunohistochemically, all the dysadherin-positive staining was membranous. Positive cell membranous dysadherinpositive staining was often observed at the edge of tumor nests, although strong immunoreactivity throughout whole tumor nests was also seen in some tumors. Basal cells of the normal cervical epithelia were positive for dysadherin while its expression in the squamous cell cervical carcinomas was variable. Among the 206 tumors, 23 (11.2%) were negative, 53 (25.7%) were scored 1+, 54 (26.2%) were scored 2+ and 76 (36.9%) were scored 3+. In the 20 tumors analyzed, mRNA expression of dysadherin basically corresponded to its protein expression. No significant correlation between expression of dysadherin and age, FIGO stage or lymph node status was observed. Higher level of dysadherin expression, however, was significantly associated with shorter overall survival (p=0.04). We conclude that there is dysadherin protein expression in basal and parabasal cells of normal cervical epithelia, and higher level of dysadherin protein expression in squamous cell cervical carcinoma is predictive of a shorter overall survival, indicating that dysadherin may be a valuable prognostic marker in cervical carcinoma. C1 [Wu, Dan] The Norwegian Radium Hospital, University of Oslo, Department of Pathology, Montebello, 0310 Oslo, Norway. [Qiao, Yuhuan] The First Teaching Hospital, Zhengzhou University, Department of Obstetrics and GynecologyHenan, China. [Kristensen, B Gunnar] The Norwegian Radium Hospital, University of Oslo, Department of Gynecologic OncologyOslo, Norway. [Li, Shanshan] The First Teaching Hospital, Zhengzhou University, Department of PathologyHenan, China. [Troen, Gunhild] The Norwegian Radium Hospital, University of Oslo, Department of Pathology, Montebello, 0310 Oslo, Norway. [Holm, Ruth] The Norwegian Radium Hospital, University of Oslo, Department of Pathology, Montebello, 0310 Oslo, Norway. [Nesland, M Jahn] The Norwegian Radium Hospital, University of Oslo, Department of Pathology, Montebello, 0310 Oslo, Norway. [Suo, Zhenhe] The Norwegian Radium Hospital, University of Oslo, Department of Pathology, Montebello, 0310 Oslo, Norway. RP Suo, Z (reprint author), The Norwegian Radium Hospital, University of Oslo, Department of Pathology, 0310 Oslo, Norway. EM zhenhe.suo@labmed.uio.no CR Jemal A, Murray T, Samuels A et al: Cancer statistics, 2003. CA Cancer J Clin 53:5-26, 2003 Flores-Luna L, Salazar-Martinez E, Escudero-De los Rios P et al: Prognostic factors related to cervical cancer survival in Mexican women. Int J Gynaecol Obstet 75:33-42, 2001 Kawagoe T, Kashimura M, Matsuura Y et al: Clinical significance of tumor size in stage IB and II carcinoma of the uterine cervix. Int J Gynecol Cancer 9:421-426, 1999 Hirohashi S, Kanai Y: Cell adhesion system and human cancer morphogenesis. Cancer Sci 94:575-581, 2003 Birchmeier W. E-cadherin as a tumor, invasion, suppressor gene. Bioessays 17:97-99, 1995 Christofori G, Semb H: The role of the cell-adhesion molecule E-cadherin as a tumor-suppressor gene. Trends Biochem Sci 24:73-76, 1999 Yoshiura K, Kanai Y, Ochiai A et al: Silencing of the E-cadherin invasion-suppressor gene by CpG methylation in human carcinomas. Proc Natl Acad Sci USA 92:7416-7419, 1995 Graff JR, Gabrielson E, Fujii H et al: Methylation patterns of the E-cadherin 5’ CpG island are unstable and reflect the dynamic, heterogeneous loss of E-cadherin expression during metastatic progression. J Biol Chem 275:2727-2732, 2000 Becker KF, Atkinson MJ, Reich U et al: E-cadherin gene mutations provide clues to diffuse type gastric carcinomas. Cancer Res 54:3845-3852, 1994 Matsuyoshi N, Hamaguchi M, Taniguchi S et al: Cadherinmediated cell-cell adhesion is perturbed by v-src tyrosine phosphorylation in metastatic fibroblasts. J Cell Biol 118:703-714, 1992 Ino Y, Gotoh M, Sakamoto M et al: Dysadherin, a cancer-associated cell membrane glycoprotein, down-regulates E-cadherin and promotes metastasis. Proc Natl Acad Sci USA 99:365-370, 2002 Aoki S, Shimamura T, Shibata T et al: Prognostic significance of dysadherin expression in advanced colorectal carcinoma. Br J Cancer 88:726-732, 2003 Shimamura T, Sakamoto M, Ino Y et al: Dysadherin overexpression in pancreatic ductal adenocarcinoma reflects tumor aggressiveness: relationship to e-cadherin expression. J Clin Oncol 21:659-667, 2003 Sato H, Ino Y, Miura A et al: Dysadherin: expression and clinical significance in thyroid carcinoma. J Clin Endocrinol Metab 88:4407-4412, 2003 Pecorelli S, Odicino F: Cervical cancer staging. Cancer J 9:390-394, 2003 Tsuiji H, Takasaki S, Sakamoto M et al: Aberrant O-glycosylation inhibits stable expression of dysadherin, a carcinomaassociated antigen, and facilitates cell-cell adhesion. Glycobiology 13:521-527, 2003 Adams CL, Nelson WJ, Smith SJ: Quantitative analysis of cadherin- catenin-actin reorganization during development of cellcell adhesion. J Cell Biol 135:1899-1911, 1996 Carico E, Atlante M, Bucci B et al: E-cadherin and alphacatenin expression during tumor progression of cervical carcinoma. Gynecol Oncol 80:156-161, 2001 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 212 EP 218 PG 7 ER PT J AU Sharma, ChM Ralte, MA Gaekwad, Sh Santosh, V Shankar, KS Sarkar, Ch AF Sharma, Chand Mehar Ralte, Mercy Angela Gaekwad, Shailesh Santosh, Vani Shankar, K S Sarkar, Chitra TI Subependymal Giant Cell Astrocytoma - a Clinicopathological Study of 23 Cases with Special Emphasis on Histogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tuberous sclerosis; SEGA; epilepsy; lateral ventricle tumor; immunohistochemistry ID tuberous sclerosis; SEGA; epilepsy; lateral ventricle tumor; immunohistochemistry AB Subependymal giant cell astrocytomas (SEGAs) are relatively rare tumors but occur commonly in the setting of the familial syndrome of tuberous sclerosis complex (TSC). In view of its varied morphology, i.e. resemblance to astrocytic and ganglion cells, its histogenesis remains controversial. We studied 23 cases of SEGA, 19 from our own institute and 4 from NIMHANS, Bangalore. These 19 cases of SEGAs were collected over a period of 23 years (1979 to 2001), and accounted for 0.16% of intracranial tumors and 0.51% of all gliomas reported at our center. The majority of patients presented with visual disturbances (19/23, 82.6%) in the form of decreased vision (60.8%) and blindness (21.7%), generalized tonic clonic seizures (43.4%) and focal motor seizures (4.37%). Age ranged from 4 to 37 years (mean 13.2 years) with male predominance (M:F 2.2:1), and the duration of symptoms varied from 1 month to 96 months (mean 17.2 months). Lateral ventricular involvement was the most common site (91.3%), followed by the third ventricle (8.6%). Nine patients (39.1%) had stigmata of tuberous sclerosis (6 at the time of diagnosis and 3 in the follow-up period). Two patients died due to surgical complications, while the rest were alive and well in the follow-up period ranging from 3 to 264 months (mean 37.1 months). Two patients experienced recurrences, one two years and another 22 years after surgery. Microscopic examination showed varied histology consisting of sweeping bundles of spindle cells, gemistocyte and ganglion-like cells with interspersed inflammatory cell component. The inflammatory cell component on special staining turned out to be an admixture of mast cells and T lymphocytes. Six cases showed areas of necrosis and/or mitosis, but were not indicative of aggressive nature of this tumor. Immunoreactivity for GFAP, NF, S-100, NSE and synaptophysin indicates that this is a hybrid tumor with glial and neuronal differentiation. None of the tumors was immunopositive for HMB-45. The significance of the presence of T lymphocytes and mast cells is not clear. It could be related to tumor immunology and may indicate a favorable prognosis. C1 [Sharma, Chand Mehar] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Ralte, Mercy Angela] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Gaekwad, Shailesh] All India Institute of Medical Sciences, Department of NeuroradiologyNew Delhi, India. [Santosh, Vani] National Institute of Mental Health and Neurosciences, Department of NeuropathologyBangalore, India. [Shankar, K S] National Institute of Mental Health and Neurosciences, Department of NeuropathologyBangalore, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. CR Shepherd CW, Scheithauer BW, Gomez MR, et al: Subependymal giant cell astrocytoma: a clinical, pathological and flow cytometric study. Neurosurg 28: 868-864, 1991 Chow CW, Klug GL, Lewis EA: Subependymal giant cell astrocytoma in children: An unusual discrepancy between histological and clinical features. J Neurosurg 68: 880-883, 1988 Bonnin JM, Rubinstein LJ, Papasozomenos SC and Marangos PJ: Subependymal giant cell astrocytomas. Significance and possible cytogenetic implications of an immunohistochemical study. Acta Neuropathol 62: 185-193, 1984 Fuziwara S, Takaki T, Hikita T, Nishio S: Subependymal giant cell astrocytoma associated with tuberous sclerosis: do subependymal nodules grow? Child’s nervous system 5: 43- 44, 1989 Morimoto K, Mogami H: Sequential CT study of subependymal giant cell astrocytoma associated with tuberous sclerosis: case report. J Neurosurg 65: 874-877, 1986 Lopes MBS, Altermatt HJ, Scheithauer BW, et al: Immunohistochemical characterisation of subependymal giant cell astrocytomas. Acta Neuropathol 91: 368-375, 1996 Bender BL, Yunis EJ: Central nervous system pathology of tuberous sclerosis in children. Ultrastruct Pathol 1: 287-299, 1980 Halmagyi GM, Bignold LP, Allospi JP: Recurrent subependymal giant cell astrocytoma in the absence of tuberous sclerosis. J Neurosurg 50: 106-109, 1979 Sima AAF, Robertson DM: Subependymal giant cell astrocytoma. Case report with ultrastructural study. J Neurosurg 50: 240-245, 1979 Trombley IK, Mirra SS: Ultrastructure of tuberous sclerosis: cortical tuber and subependymal tumor. Ann Neurol 9: 174- 181, 1981 Bancel B, Belin MF, Meiniel A, et al: Contribution a l’etude de l’histogenese des gliomes sous ependymaires de la sclerose tubereuse de Bourneville. Ann Pathol 10: 109-116, 1990 Iwasaki Y, Yoshikawa H, Sasaki M, et al: Clinical and immunohistochemical studies of subependymal giant cell astrocytoma associated with tuberous sclerosis. Brain Dev 12: 478-481, 1990 Stefanssan K, Wollmann RL: Distribution of neuronal specific protein 14-3.2 in central nervous system lesions of tuberous sclerosis. Acta Neuropathol, Berl, 53: 113-117, 1981 Nakamura S, Tsubokawa T: Ultrastructure of subependymal giant cell astrocytoma associated with tuberous sclerosis. J Clin Electron Microscope 20: 5-6, 1987 Bonetti F, Chiodera PL, Pea M, et al: Transbronchial biopsy in lymphangiomyomatosis of the lung: HMB- 45 for diagnosis. Am J Surg Pathol 17:1092-1102, 1993 Gyure KA, Hart WR, Kennedy AW: Lymphangiomyomatosis of the uterus associated with tuberous sclerosis and malignant neoplasia of the female genital tract: a report of two cases. Int J Gynecol Pathol 14: 344-351, 1995 Pea M, Bonetti F, Zamboni G, et al: Melanocyte marker HMB- 45 is regularly expressed in angiomyolipoma of the kidney. Pathology 23: 185-188, 1991 Weeks DA, Chase DR, Malott RL, et al: HMB-45 staining in angiomyolipoma, cardiac rhabdomyoma, other mesenchymal processes and tuberous sclerosis associated brain lesions. Int J Surg Pathol 1:191-198, 1994 Al-Saleem T, Wessner LL, Scheithauer BW, et al: Malignant tumors of the kidney, brain, and soft tissues in children and young adults with the tuberous sclerosis complex. Cancer 83: 2208-2216, 1988 Kingsley DPE, Kendall BE, Fitz CR: Tuberous sclerosis: A clinico-radiological evaluation of 110 cases with particular reference to atypical presentation. Neuroradiology 28: 38-46, 1986 Boesel CP, Paulson GW, Kosnik EJ, Earle KM: Brain hamartomas and tumors associated with tuberous sclerosis. Neurosurgery 4: 410-417, 1979 Padmalatha C, Harsuff RC, Ganick D, Hafez GR: Glioblastoma multiforme with tuberous sclerosis. Arch Lab Med 105: 645-650, 1980 Brown JM: Tuberous sclerosis with malignant astrocytoma. Med J Austr 1:811-814, 1975. Medhkour A, Traul D, Husain M: Neonatal subependymal giant cell astrocytoma. Ped Neurosurg 36: 271-274, 2002 Gyure KA, Prayson RA: Subependymal giant cell astrocytoma: A clinicopathologic study with HMB-45 and MIB-1 immunohistochemical analysis. Mod Pathol 10: 313-317, 1997 Bacchi CE, Bonetti F, Pea M, et al: HMB-45: a review. Appl Immunohistochem 4: 73-85, 1996 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 219 EP 224 PG 6 ER PT J AU Sharifzadeh, S Owji, MS Pezeshki, MA Malek-Hoseini, Z Kumar, VP Ghayumi, MAS Ghaderi, A AF Sharifzadeh, Sedigheh Owji, Mohammad Seyed Pezeshki, Mohammad Abdul Malek-Hoseini, Zahra Kumar, Vijayananda Perikala Ghayumi, Mohammad Ali Sied Ghaderi, Abbas TI Establishment and Characterization of a Human Large Cell Lung Cancer Cell Line with Neuroendocrine Differentiation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cell line; lung cancer; large cell carcinoma; neuroendocrine differentiation ID cell line; lung cancer; large cell carcinoma; neuroendocrine differentiation AB Characterization of a human lung cancer cell line is reported. This cell line was established from a patient referred to Nemazi Hospital of Shiraz University of Medical Sciences with a diagnosis of poorly differentiated carcinoma. Sterile sample from peritoneal effusion was taken and immediately cultured in RPMI-1640 medium containing 20% FBS, at 37C with 5% CO2. This cell line has been in continuous culture for more than one year and has been named as Mehr-80. Several features of the cell line were investigated, including growth characteristics, electron microscopic features, cloning efficiency in soft agar, expression of various antigenic markers, chromosomal and DNA analysis. On the basis of morphological and immunohistochemical analysis of Mehr-80, it is possible to conclude that this cell line is characterized by features similar to those reported for large cell carcinoma with neuroendocrine differentiation (LCCND). This cell line will be a valuable in vitro tool for further studies on lung cancers. C1 [Sharifzadeh, Sedigheh] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Owji, Mohammad Seyed] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Pezeshki, Mohammad Abdul] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Malek-Hoseini, Zahra] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Kumar, Vijayananda Perikala] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Ghayumi, Mohammad Ali Sied] Shiraz University of Medical Sciences, Department of Internal MedicineShiraz, Iran. [Ghaderi, Abbas] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. RP Ghaderi, A (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Immunology, Shiraz, Iran. EM ghaderia@sums.ac.ir CR Pisani P, Parkin DM, Bary F, Ferlay J: Estimates of the worldwide mortality from 25 cancers in 1990. Int J Cancer 83:18- 29, 1999 Ferlay J, Bray F, Pisani P, Parkin DM: GLOBOCAN 2000: Cancer Incidence, Mortality and Prevalence Worldwide, Version 1.0 IARC Cancer Base No.5. IARC Press, Lyon 2001 Travis WD, Colby TV, Corrin B: Histological typing of lung and pleural tumors. 3rd ed., Berlin, Springer 1999 Iyoda A, Hiroshima K, Baba M, et al: Pulmonary large cell carcinomas with neuroendocrine features are high-grade neuroendocrine tumors. Ann Thorac Surg 73: 1049-1054, 2002 Difilippantonio S, Chen Y, Pietas A, et al: Gene expression profiles in human non-small and small-cell lung cancers. Eur J Cancer 39: 1936-1947, 2003 Yamagata N, Shyr Y, Yanagisawa K, et al: A training-testing approach to the molecular classification of resected non-small cell lung cancer. Clin Cancer Res 9: 4695-4704, 2003 Lim EH, Aggarwal A, Agasthian T, et al: Feasibility of using low-volume tissue samples for gene expression profiling of advanced non-small cell lung cancers. Clin Cancer Res 9: 5980-5987, 2003 Bonner AE, Lemon WJ, Devereux TR, et al: Molecular profiling of mouse lung tumors: association with tumor progression, lung development, and human lung adenocarcinoma. Oncogene 3:1166-1176, 2004 Yesner R: Heterogeneity of so-called neuroendocrine lung tumors. Exp Mol Pathol 70: 179-182, 2001 Macikova I, Perzelova A, Mraz P, et al: Establishment, morphological, growth and cytoskeletal properties of 135-BCA carcinoma cell line derived from lung brain metastasis. Neoplasma 48: 479-482, 2001 Harada R, Uemura Y, Kobayashi M, et al: Establishment and characterization of a new lung cancer cell line, MI-4, producing high levels of granulocyte colony stimulating factor. Jpn J Cancer Res 93: 667-676, 2002 Kozaki K, Miyaishi O, Tsukamoto T, et al: Establishment and characterization of a human lung cancer cell line NCI-H460- LNM35 with consistent lymphogenous metastasis via both subcutaneous and orthotopic propagation. Cancer Res 60:2535-2540, 2000 Sekio Y, Sato M, Usami N, et al: Establishment of a large cell lung cancer cell line, Y-ML-1B, producing granulocyte colony-stimulating factor. Cancer Genet Cytogenet 137: 33- 42, 2002 Shijubo N, Inoue Y, Hirasawa M, et al: Granulocyte colonystimulating factor-producing large cell undifferentiated carcinoma of the lung. Intern Med 3: 277-280, 1992 Suzuki A, Takahashi T, Okuno Y, et al: Analysis of abnormal expression of g-csf gene in a novel tumor cell line, KHC 287, elaborating G-CSF, IL-1 and IL-6 with co-amplification of cmyc and c-ki-ras. Int J Cancer 48: 428-433, 1991 Shibuya T: Establishment of a G-CSF and GM-CSF producing cell line from human large cell line from human large cell carcinoma of the lung. Eur J Haematol 43: 182-183, 1989 Visscher DW, Zabro RJ, Trojanowski JQ, et al: Neuroendocrine differentiation in poorly differentiated lung carcinoma: a light microscopic and immunohistologic study. Mod Pathol 3: 508- 512, 1990 Jiang SX, Kameya T, Shoji M, et al: Large cell neuroendocrine carcinoma of the lung: A histologic and immunohistochemical study of 22 cases. Am J Surg Pathol 22: 526-537, 1998 Upton MP, Hirohashi S, Tome Y, et al: Expression of vimentin in surgically resected adenocarcinomas and large cell carcinomas of lung. Am J Surg Pathol 10: 560-567, 1986 Roberts HL, Komaki R, Allen P, et al: Prognostic significance of DNA content in stage I adenocarcinoma of the lung. Int J Radiat Oncol Biol Phys 41: 573-578, 1998 Yousem SA, Taylor SR: Typical and atypical carcinoid tumors of lung: a clinicopathologic and DNA analysis of 20 tumors. Mod Pathol 3: 502-507, 1990 Volm M, Mattern J, Sonka J, et al: DNA distribution in nonsmall- cell lung carcinomas and its relationship to clinical behavior. Cytometry 6: 348-356, 1985 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 225 EP 230 PG 6 ER PT J AU Dekel, Y Rath-Wolfson, L Rudniki, C Koren, R AF Dekel, Yoram Rath-Wolfson, Lea Rudniki, Carlos Koren, Rumelia TI Talc Inhalation is a Life-Threatening Condition SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE talc inhalation; fibrosis; adult respiratory distress syndrome ID talc inhalation; fibrosis; adult respiratory distress syndrome AB A case of rapidly progressive disease and pulmonary hypertension due to chronic cosmetic talc inhalation is presented. Although an uncommon cause of pulmonary hypertension, talc, especially through intravenous administration, should be included in the etiology of parenchymal pulmonary hypertension. In our case talc inhalation was inadvertent, causing fulminant disease leading to the patient’s death. To our knowledge, this is the first case of inadvertent talc inhalation causing death in adult patient. C1 [Dekel, Yoram] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Rath-Wolfson, Lea] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Rudniki, Carlos] Tel Aviv University, Sackler Faculty of Medicine, Ramat Aviv, 69978 Tel Aviv, Israel. [Koren, Rumelia] Tel Aviv University, Sackler Faculty of Medicine, Ramat Aviv, 69978 Tel Aviv, Israel. RP Koren, R (reprint author), Tel Aviv University, Sackler Faculty of Medicine, 69978 Tel Aviv, Israel. EM rumelia@isdnmail.co.il CR Abraham JL, Brambilla C: Particle size for differentiation between inhalation and injection pulmonary talcosis. Environ Res 21: 94-96, 1980 Arnett EN, Battle WE, Russo JV, et al: Intravenous injection of talccontaining drugs intended for oral use. A cause of pulmonary granulomatosis and pulmonary hypertension. Am J Med 60: 711-718, 1976 Avolio G, Galietti F, Oliaro A, et al: Talcosis as an occupational disease. Case histories. Minerva Med 80: 269-273, 1989 Bignon J, Brochard P: Pulmonary fibrosis and inorganic particles. Rev Fr Mal Respir 11: 371-382, 1983 Canessa PA, Torraca A, Lavecchia MA, et al: Primary acute pulmonary cavitation in asymptomatic sarcoidosis. Sarcoidosis 6: 158- 160, 1989 Cotton WH, Davidson PJ: Aspiration of baby powder. N Engl J Med 313: 1662, 1985 Crouch E, Churg A: Progressive massive fibrosis of the lung secondary to intravenous injection of talc. A pathologic and mineralogic analysis. Am J Clin Pathol 80: 520-526, 1983 Egan AJ, Tazelaar HD, Myers JL, et al: Munchausen syndrome presenting as pulmonary talcosis. Arch Pathol Lab Med 123: 736-738, 1999 Gibbs AE, Pooley FD, Griffiths DM, et al: Talc pneumoconiosis: a pathologic and mineralogic study. Hum Pathol 23:1344-1354, 1992 Gutermuth M, Schirg E, Steinbacher D, et al: Accidental aspiration of baby powder. Radiographic findings, preventive and therapeutic aspects. Intensivmed Prax 2: 83-92, 1980 Hollinger MA: Pulmonary toxicity of inhaled and intravenous talc. Toxicol Lett 52: 121-127, 1990 Honda Y, Beall C, Delzell E, et al: Mortality among workers at a talc mining and milling facility. Ann Occup Hyg 46: 575-585, 2002 Hopkins GB: Pulmonary angiothrombotic granulomatosis in drug offenders. JAMA 221: 909-911, 1972 Lamb D, Roberts G: Starch and talc emboli in drug addicts’ lungs. J Clin Pathol 25: 876-881, 1972 Lundquest DE, Young WK, Edland JF: Maternal death associated with intravenous methylphenidate, Ritalin, and pentazocine, Talwin, abuse. J Forensic Sci 32: 798-801, 1987 Magnan A, Ottomani A, Garbe L, et al: Respiratory failure in a HIV seropositive heroin addict female. Ann Fr Anesth Reanim 10: 74- 76, 1991 Motomatsu K, Adachi H, Uno T: Two infant deaths after inhaling baby powder. Chest 75: 448-450, 1979 Oubeid M, Bickel JT, Ingram EA, et al: Pulmonary talc granulomatosis in a cocaine sniffer. Chest 98: 237-239, 1990 Pairaudeau PW, Wilson RG, Hall MA, et al: Inhalation of baby powder: an unappreciated hazard. BMJ 302: 1200-1201, 1991 Pfenninger J, D’Apuzzo V: Powder aspiration in children. Report of two cases. Arch Dis Child 52: 157-159, 1977 Pickrell JA, Snipes MB, Benson JM, et al: Talc deposition and effects after 20 days of repeated inhalation exposure of rats and mice to talc. Environ Res 49: 233-245, 1989 Reyes de la Rocha S, Brown MA: Normal pulmonary function after baby powder inhalation causing adult respiratory distress syndrome. Pediatr Emerg Care 5: 43-48, 1989 Scancarello G, Romeo R, Sartorelli E: Respiratory disease as a result of talc inhalation. J Occup Environ Med 38: 610-614, 1996 Steele AA: Suicidal death by aspiration of talcum powder. Am J Forensic Med Pathol 11: 316-318, 1990 Wehner AP: Biological effects of cosmetic talc. Food Chem Toxicol 32: 1173-1184, 1994 Zazenski R, Ashton WH, Briggs D, et al: Talc: occurrence, characterization, and consumer applications. Regul Toxicol Pharmacol 21: 218-229, 1995 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 231 EP 233 PG 3 ER PT J AU Culhaci, N Meteoglu, I Kacar, F Ozbas, S AF Culhaci, Nil Meteoglu, Ibrahim Kacar, Furuzan Ozbas, Serdar TI Abscess of the Spleen SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE abscess; spleen; immunosuppression ID abscess; spleen; immunosuppression AB Abscess of the spleen is a very rare lesion. In this study, 4 cases of splenic abscess are presented and discussed along with the literature. The cases were between 16 and 55 years-old and two of them had hematologic malignancy. All of them had been operated on because of acute abdomen, and in two cases splenic rupture was present. Only in one of the cases was salmonellosis detected by microbiological methods. By histological examination, expansion and congestion in splenic sinusoids, and foci of abscess including wide areas of necrosis and inflammatory infiltration by neutrophils were seen in all cases. The most frequent cause of splenic abscess is septic embolism arising from bacterial endocarditis. There are also a few splenic abscess cases seen with malignancies. While splenic abscess is seen rarely, it has a high rate of mortality when it is diagnosed late. C1 [Culhaci, Nil] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Meteoglu, Ibrahim] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Kacar, Furuzan] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Ozbas, Serdar] Adnan Menderes University Medical School, Department of SurgeryAydin, Turkey. RP Culhaci, N (reprint author), Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. EM nculhaci@hotmail.com CR Spleen. In: Ackerman’s Surgical Pathology., Ed: Rosai J), Mosby-Year Book, 8th ed., St. Louis, MO, 1996, pp. 1777-1778 Changchien CS, Tsai TL, Hu TH, et al: Sonographic patterns of splenic abscess: an analysis of 34 proven cases. Abdom Imaging 27: 739-745, 2002. Smyrniotis V, Kehagias D, Voros D, et al: Splenic abscess. An old disease with new interest. Dig Surg 17: 354-357, 2000 Ramakrishnan MR, Sarathy TKP, Balu M: Percutaneous drainage of splenic abscess: case report and review of literature. Pediatrics 79: 1029-1030, 1987 Aessopos A, Politou M, Farmakis D, et al: Staphylococcus aureus abscess of the spleen in a beta-thalassemia patient. Scand J Infect Dis 34: 466-480, 2002 Colmenero JD, Queipo-Ortuno MI, Reguera JM, et al: Chronic hepatosplenic abscess in brucellosis. Clinico-therapeutic features and molecular diagnostic approach. Diagn Microbiol Infect Dis 42:159-167, 2002 Yayly G, Yºler M, Oyar O: Medically treated splenic abscess due to Brucella melitensis. Scand J Infect Dis 34: 133-135, 2001 Shedda S, Campbell I, Skinner I. Clostridium difficile splenic abscess. Aust NZ J Surg 70: 147-148, 2000 Wald BR, Ortega JA, Ross L, et al: Candidal splenic abscess complicating acute leukemia of childhood treated by splenectomy. Pediatrics 67:296-99, 1981 Bessho H, Ichihara I, Takii M: A case of splenic abscess due to Chlamydia pneumoniae. Diagn Microbiol Infect Dis 39: 261- 264, 2001 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 234 EP 236 PG 3 ER PT J AU Vargas Gonzalez, R San Martin-Brieke, W Gil-Orduna, C Lara-Hernandez, F AF Vargas Gonzalez, Roberto San Martin-Brieke, Walter Gil-Orduna, Claudia Lara-Hernandez, Fabiola TI Desmoplastic Fibroma-Like Tumor of Maxillofacial Region Associated with Tuberous Sclerosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE desmoplastic fibroma; desmoid tumor; bone tumor; tuberous sclerosis ID desmoplastic fibroma; desmoid tumor; bone tumor; tuberous sclerosis AB Desmoplastic fibroma is a rare primary tumor of bone that histologically and biologically mimics the extra-abdominal desmoid tumor of soft tissue. It usually presents in patients during the first three decades of life and often involves the mandible or long bones of the skeleton. Its clinical behavior is characterized by a locally aggressive, infiltrating, and destructing course, often with invasion of surrounding tissues but without metastasis. We present herein the clinicopathological features of a desmoplastic fibroma-like tumor involving the left maxillofacial region in a 14-year-old Hispanic boy with tuberous sclerosis. C1 [Vargas Gonzalez, Roberto] Hospital Para el Nino Poblano, Department of Pathology, Km 1.5 Carretera Federal Puebla-Atlixco, 72190 Puebla, Mexico. [San Martin-Brieke, Walter] Hospital Para el Nino Poblano, Department of Maxillofacial SurgeryPuebla, Mexico. [Gil-Orduna, Claudia] Hospital Para el Nino Poblano, and Facultad de Estomatologia B.U.A.P., Department of EstomatologyPuebla, Mexico. [Lara-Hernandez, Fabiola] Hospital Para el Nino Poblano, Department of Pathology, Km 1.5 Carretera Federal Puebla-Atlixco, 72190 Puebla, Mexico. RP Vargas Gonzalez, R (reprint author), Hospital Para el Nino Poblano, Department of Pathology, 72190 Puebla, Mexico. EM soncoy@msn.com CR Miyamoto Y, Satomura K, Rikimaru K, Hayashi Y: Desmoplastic fibroma of the mandible associated with tuberous sclerosis. J Oral Pathol Med 24: 93-96, 1995 Damm DD, Tomich CE, White DK, et al: Intraosseous fibrous lesions of the jaws: a manifestation of tuberous sclerosis. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 87: 334-340, 1999 Jaffe HL: Tumors and Tumorous Conditions of the Bones and Joints. Lea & Febiger, Philadelphia, PA, 1958, pp 298–303 Inwards CY, Unni KK, Beabout JW, Sim FH: Desmoplastic fibroma of bone. Cancer 68: 1978–1983, 1991 Salzer Kuntschik M: Desmoplastishes Fibrom. In: Witt AN, Rettig H, Schlegel KF, Hackenbroch M, Hupfauer W, eds. Orthopadie in Praxis und Klinik, Band III: Allgemeine Orthopadie, Teil 2: Tumoren und tumorahnliche Erkrankungen, 2nd ed. Stuttgart, Germany: Georg Thieme Verlag 1: 258–265, 1984 Dahlin DC, Unni KK: Bone Tumors. 4th ed. Thomas, Springfield, IL, 1986, pp 375–378 Bohm P, Krober S, Greschniok A. et al: Desmoplastic fibroma of the bone: a report of two patients, review of the literature, and therapeutic implications. Cancer 78: 1011–1023, 1996 Crim JR, Gold RH, Mirra JM, et al: Desmoplastic fibroma of bone: radiographic analysis. Radiology 172: 827-832, 1989 Young JWR, Aisner SC, Levine AM, et al: Computed tomography of desmoid tumors of bone: desmoplastic fibroma. Skeletal Radiol 17: 333–337, 1998 Yeung RS: Multiple roles of the tuberous sclerosis complex genes. Genes Chromosomes Cancer 38: 368-375, 2003 Lygidakis NA, Lindenbaum RH: Oral fibromatosis in tuberous sclerosis. Oral Surg Oral Med Oral Pathol 68: 725-728, 1989 Scully C: Orofacial manifestations in tuberous sclerosis. Oral Surg Oral Med Oral Pathol 44:706-716, 1977 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 237 EP 239 PG 3 ER PT J AU Bolat, F Kayaselcuk, F Erkan, NA Cagici, AC Bal, N Tuncer, I AF Bolat, Filiz Kayaselcuk, Fazilet Erkan, Nabi Alper Cagici, Alper Can Bal, Nebil Tuncer, Ilhan TI Epidermoid Carcinoma Arising in Warthin’s Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Epidermoid carcinoma; Warthin’s tumor ID Epidermoid carcinoma; Warthin’s tumor AB Warthin’s tumor is a well-defined salivary gland neoplasm consisting of benign epithelial and lymphoid components. However, malignant transformation is extremely rare and the differential diagnosis of metastasis from an epidermoid carcinoma in Warthin’s tumor is important. We present a case with epidermoid carcinoma arising in Warthin’s tumor of parotid gland in a 48-year-old woman, and differential diagnosis is discussed. C1 [Bolat, Filiz] Baskent University, Department of Pathology, Adana Hastanesi, Yure?ir, 01250 Adana, Turkey. [Kayaselcuk, Fazilet] Baskent University, Department of Pathology, Adana Hastanesi, Yure?ir, 01250 Adana, Turkey. [Erkan, Nabi Alper] Baskent University Faculty of Medicine, Adana Teaching and Medical Research Center, Department of Ear-Throat-NoseAdana, Turkey. [Cagici, Alper Can] Baskent University Faculty of Medicine, Adana Teaching and Medical Research Center, Department of Ear-Throat-NoseAdana, Turkey. [Bal, Nebil] Baskent University, Department of Pathology, Adana Hastanesi, Yure?ir, 01250 Adana, Turkey. [Tuncer, Ilhan] Baskent University, Department of Pathology, Adana Hastanesi, Yure?ir, 01250 Adana, Turkey. RP Bolat, F (reprint author), Baskent University, Department of Pathology, 01250 Adana, Turkey. EM drfilizbolat@yahoo.com CR Huvos AG, Paulino AFG: Salivary Glands. In: Diagnostic Surgical Pathology., Eds: Stenberg SS), 3rd edition, Lippincott Williams&Wilkins, Philedelphia, 1999, pp 861-862 Gunduz M, Yamanaka N, Hotomi M, et al: Squamous cell carcinoma arising in a Warthin’s tumor. Auris Nasus Larynx 26:355- 360, 1999 Skalova A, Michal M, Nathansky Z: Epidermoid carcinoma arising in Warthin’s tumour: a case study. J Oral Pathol Med 23:330- 333, 1994 Morrison GAJ, Shaw HJ: Squamous carcinoma arising within a Warthin’s tumor of the parotid gland. J Laryngol Otol 102:1189- 1191, 1988 Damjanov I, Sneff EM, Delerme AN: Squamous cell carcinoma arising in Warthin’s tumor of the parotid gland. A light, electron microscopic, and immunohistochemical study. Oral Surg Oral Med Oral Pathol 55:286-290, 1983 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 240 EP 242 PG 3 ER PT J AU Swiatoniowski, G Mazur, G Halon, A Rozumek, G Dabrowska, M Zawisza, R Prudlak, E AF Swiatoniowski, Grzegorz Mazur, Grzegorz Halon, Agnieszka Rozumek, Grzegorz Dabrowska, Maria Zawisza, Radoslaw Prudlak, Edmun TI Malignant Melanoma with Gall Bladder Metastasis as a Second Neoplasm in the Course of Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE prostate cancer; malignant melanoma; hormone therapy; second neoplasm ID prostate cancer; malignant melanoma; hormone therapy; second neoplasm AB Malignant melanoma is a neoplasm with an often unpredictable course and metastases potentially affecting all organs of the human body. Metastases into the gall bladder are rare. The role of hormonal background in the development and progression of malignant melanoma has not been established. The authors present a case of a 63-year-old man who had initially undergone long-term hormone therapy for the treatment of prostate cancer, and later presented with melanoma metastatic to the gall bladder. C1 [Swiatoniowski, Grzegorz] 4th Academic Military Hospital, Department of Internal MedicineWroclaw, Poland. [Mazur, Grzegorz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, Pasteura 4, 50-367 Wroclaw, Poland. [Halon, Agnieszka] Wroclaw Medical University, Department of PathologyWroclaw, Poland. [Rozumek, Grzegorz] 4th Academic Military Hospital, Department of Internal MedicineWroclaw, Poland. [Dabrowska, Maria] 4th Academic Military Hospital, Department of Internal MedicineWroclaw, Poland. [Zawisza, Radoslaw] 4th Academic Military Hospital, Department of SurgeryWroclaw, Poland. [Prudlak, Edmun] 4th Academic Military Hospital, Department of Internal MedicineWroclaw, Poland. RP Mazur, G (reprint author), Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, 50-367 Wroclaw, Poland. EM grzegmaz@hemat.am.wroc.pl CR Feskanich D, Hunter DJ, Willett WC, et al: Oral contraceptive use and risk of melanoma in premenopausal women. Br J Cancer 81: 918-923, 1999 Grin CM, Driscoll MS, Grant-Kels JM: The relationship of pregnancy, hormones, and melanoma. Semin Cutan Med Surg 17: 167-171, 1998 Grostern RJ, Slusker-Shternfeld I, Bacus SS, et al: Absence of type I estrogen receptors in choroidal melanoma: analysis of Collaborative Ocular Melanoma Study, COMS, eyes. Am J Ophthalmol 131: 788-791, 2001 Heath DI, Womack C: Primary malignant melanoma of the gall bladder. J Clin Pathol 41: 1073-1077, 1988 Higgins CM, Strutton GM: Malignant melanoma of the gall bladder – does primary melanoma exist? Pathology 27: 312- 314, 1995 Jones JA, Albright KD, Christen RD, et al: Synergy between tamoxifen and cisplatin in human melanoma cells is dependent on the presence of antiestrogen-binding sites. Cancer Res 57: 2657-2660, 1997 Kanter-Lewensohn L, Girnita L, Girnita A, et al: Tamoxifeninduced cell death in malignant melanoma Wells: possible involvement of the insulin-like growth factor 1, IGF-1, pathway. Mol Cell Endocrinol 165: 131-137, 2000 Kohler U, Jacobi T, Sebastian G, Nagel M: Laparoscopic cholecystectomy in isolated gallbladder metastasis of malignant melanoma. Chirurg 71: 1517-1520, 2000 Lama G, Angelucci C, Bruzzese N, et al: Sensitivity of human melanoma cells to oestrogens, tamoxifen and quercetin: is there any relationship with type I and II estrogen binding site expression? Melanoma Res 8: 313-322, 1999 Langley RG, Bailey EM, Sober AJ: Acute cholecystitis from metastatic melanoma to the gall-bladder in a patient with a low-risk melanoma. Br J Dermatol 136: 279-282, 1997 Miller JG, Gee J, Price A, et al: Investigation of estrogen receptors, sex steroids and soluble adhesion molecules in the progression of malignant melanoma. Melanoma Res 7: 197-208, 1997 Ostick DG, Haqqani MT: Obstructive cholecystitis due to metastatic melanoma. Postgrad Med J 52: 710-712, 1976 Richardson B, Price A, Wagner M, et al: Investigation of female survival benefit in metastatic melanoma. Br J Cancer 80: 2025-2033, 1999 Rusthoven JJ: The evidence for tamoxifen and chemotherapy as treatment for metastatic melanoma. Eur J Cancer 34, Suppl 3): S31-S36, 1998 Seelig MH, Schonleben K: Laparoscopic cholecystectomy for a metastasis of a malignant melanoma in the gallbladder. Z Gastroenterol 35: 673-675, 1997 Toma S, Ugolini D, Palumbo R: Tamoxifen in the treatment of metastatic malignant melanoma: still a controversy? Int J Oncol 15: 321-337, 1999 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2004 VL 10 IS 4 BP 243 EP 245 PG 3 ER PT J AU Kopper, L Timar, J AF Kopper, Laszlo Timar, Jozsef TI Genomics of Lung Cancer may Change Diagnosis, Prognosis and Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE lung cancer; genomics ID lung cancer; genomics AB Despite significant improvements in tumor management in general, the prognosis of lung cancer patients remains dismal. It is a hope that our increasing knowledge in molecular aspects of tumor development, growth and progression will open new targets for therapeutic interventions. In this review we discuss some of the more recent results of this field. This includes the susceptibility factors, an association between genetic changes in EGFR pathway and tyrosine kinase inhibitors, the role of gene hypermethylation and genetic profiling, as well as different molecular aspects of tumor progression. Available data all support that lung cancer is a group of diseases with not only distinct histological but with similarly different genetic characters. Accordingly, the diagnosis, prognosis and therapy must accommodate this heterogeneity. C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor ProgressionBudapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM kopper@korb1.sote.hu CR Schwartz AG: Genetic predisposition to lung cancer. Chest 125: 86S-89S, 2004 Olak J, Colson Y: Gender differences in lung cancer: Have we really come a long way, baby? J Thorac Cardiovasc Surg 128: 346-351, 2004 Minna JD, Roth JA, Gazdar AF: Focus on lung cancer. Cancer Cell 1: 49-52, 2002 Balsara BR, Testa JR: Chromosomal imbalances in human lung cancer. Oncogene 21: 6877-6883, 2002 Yokota J, Kohno T: Molecular footprints of human lung cancer progression. Cancer Sci 95: 197-204, 2004 Kazanjian A, Wallis D, Au N, et al: Growth factor independence- 1 is expressed in primary human neuroendocrine lung carcinomas and mediates the differentiation of murine pulmonary neuroendocrine cells. Cancer Res 64: 3874-6882, 2004 Marchetti A, Martella C, Felicioni L, et al:EGFR mutations in non-small-cell lung cancer: analysis of a large series of cases and development of a rapid and sensitive method for diagnostic screening with potential implications on pharmacologic treatment. J Clin Oncol 23: 857-865, 2005 Lynch TJ, Bell DW, Sordella R, et al: Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350: 2129-2139, 2004 Paez JG, Janne PA, Lee JC, et al:EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304: 1497-1500, 2004 Amann J, Kalyankrishna S, Massion PP, et al: Aberrant epidermal growth factor receptor signaling and enhanced sensitivity to EGFR inhibitors in lung cancer. Cancer Res 65: 226-235, 2005 Cancer Genome Project and Collaborative Group. Nature 431: 525-526, 2004 West KA, Linnoila IR, Brognard J, et al: Tobacco carcinogeninduced cellular transformation increases Akt activation in vitro and in vivo. Chest 125: 101S-102S, 2004 De Benedetti A, Graff JR: eIF expression and its role in malignancies and metastases. Oncogene 23: 3189-3199, 2004 Sugita M, Geraci M, Gao B, et al: Combined use of oligonucleotide and tissue microarray identifies cancer/testis antigens as biomarkers in lung carcinoma. Cancer Res 62: 3971-3979, 2002 Belinsky SA: Gene-promoter hypermethylation as a biomarker in lung cancer. Nature Rev Cancer 4: 707-717, 2004 Mazieres J, He B, You L, et al: Wnt inhibitory factor-1 is silenced by promoter hypermethylation in lung cancer. Cancer Res 64: 4717-4720, 2004 Garber ME, Troyanskaya OG, Schluens K, et al: Diversity of gene expression in adenocarcinoma of the lung. Proc Natl Acad Sci USA 98: 13784-13789, 2001 Bhattacharjee A, Richards WG, Staunton J, et al:Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. Proc Natl Acad Sci USA 98: 13790-13795, 2001 Wigle DA, Jurisica I, Radulovich N, et al: Molecular profiling of non-small cell lung cancer and correlation with disease-free survival. Cancer Res 62: 3005-3008, 2002 Kaminski N, Krupsky M: Gene expression patterns, prognostic and diagnostic markers, and lung cancer biology. Chest 125: 111S-115S, 2004 Beer DG, Kardia SL, Huang CC, et al: Gene expression profiles predict survival of patients with lung adenocarcinoma. Nat Med 8: 816-824, 2002 Gordon GJ, Jensen RV, Hsiao LL, et al: Translation of microarray data into clinically relevant cancer diagnostic tests using gene expression ratios in lung cancer and mesothelioma. Cancer Res 62: 4963-4967, 2002 Martin B, Paesmans M, Mascaux C et al: Ki-67 expression and patients survival in lung cancer: systematic review of the literature with meta-analysis. Br J Cancer 91:2018-2025, 2004 Lu C, Soria J-C, Tang X, et al: Prognostic factors in resected stage I non-small-cell lung cancer: a multivariate analysis of six molecular markers. J Clin Oncol 22:4575-4583, 2004 Mehdi SA, Tatum AH, Newman NB et al: Prognostic markers in resected stage I and II non-small-cell lung cancer: an analysis of 260 patients with 5 year follow-up. Clin Lung Cancer 1:59- 67, 1999 Masuya D, Huang C, Liu D, et al: The tumour-stromal interaction between intratumoral c-Met and stromal hepatocyte growth factor associated with tumour growth and prognosis in nonsmall- cell lung cancer patients. Br J Cancer 90:1555-1562, 2004 Fukuoka J, Fujii T, Shih JH et al:Chromatin remodeling factors and BRM/BRG1 expression as prognostic indicators in nonsmall cell lung cancer. Clin Cancer Res 10:4314-4324, 2004 Goncharuk VN, del-Rosario A, Kren L et al: Co-downregulation of PTEN, KAI-1, and nm23-H1 tumor/metastasis suppressor proteins in non-small cell lung cancer. Ann Diagn Pathol 8:6-16, 2004 Diederichs S, Bulk E, Steffen B et al: S100 family members and trypsinogens are predictors of distant metastasis and survival in early-stage non-small cell lung cancer. Cancer Res 64:5564- 5569, 2004 Simi L, Andreani M, Davini F et al: Simultaneous measurement of MMP9 and TIMP1 mRNA in human non small cell lung cancers by multiplex real time RT-PCR. Lung Cancer 45:171- 179, 2004 Takahashi H, Ebihara S, Okazaki T et al: Clinical significance of heparanase activity in primary resected non-small cell lung cancer. Lung Cancer 45:207-214, 2004 Shah L, Walter KL, Borczuk AC et al: Expression of Syndecan- 1 and expression of epidermal growth factor receptor are associated with survival in patients with nonsmall cell lung carcinoma. Cancer 101:1632-1638, 2004 O-charoenrat P, Rusch V, Talbot SG et al: Casein kinase II alpha subunit and C1-inhibitor are independent predictors of outcome in patients with squamous cell carcinoma of the lung. Clin Cancer Res 10:5709-5803, 2004 Au NHC, Cheang M, Huntsman DG et al: Evaluation of immunohistochemical markers in non-small cell lung cancer by unsupervised hierarchical clustering analysis: a tissue microarray study of 284 cases and 18 markers. J Pathol 204:101-109, 2004 Shabman MS, Srinivasan R, Wali A et al: Expression of p53 protein and the apoptotic regulatory molecules Bcl-2, Bcl-XL, and Bax in locally advanced squamous cell carcinoma of he lung. Lung Cancer 45:181-188, 2004 Toledo G, Sola JJ, Lorenzo MD et al: Loss of FHIT protein expression is related to high proliferation, low apoptosis and worse prognosis in non-small-cell lung carcinoma. Mod Pathol 17:440-448, 2004 Aljada IS, Rammath N, Donohue K et al: Upregulation of the tissue inhibitor of metalloproteinase-1 protein is associated with progression of human non-small-cell lung cancer. J Clin Oncol 22:3218-3229, 2004 Tsutsumida H, Goto M, Kitajima S et al: Combined status of MUC1 mucin and surfactant apoprotein A expression can predict the outcome of patients with small-size lung adenocarcinoma. Histopathology 44: 147-155, 2004 Ramaswamy S, Ross KN, Lander ES, Golub H:A molecular signature of metastasis in primary solid tumors. Nat Genet 33:49- 54, 2003 Araki K, Hashimoto K, Ardyanto TD, et al: Co-expression of Cox-2 and EGFR in stage I human bronchial adenocarcinomas. Lung Cancer 45:161-169, 2004 Yamaguchi NH, Lichtens AJ, Dmarchi LM et al:COX-2, MMP- 9, and Noguchi classification provide additional prognostic information about adenocarcinoma of the lung. A study of 117 patients from Brazil. Am J Clin Pathol 121:78-86, 2004 Pelosi G, Barisella M, Pasini F et al: CD117 immunoreactivity in stage I adenocarcinoma and squamous cell carcinoma of the lung: relevance to prognosis in a subset of adenocarcinoma patients. Mod Pathol 17:711-721, 2004 Jones MH, Virtanen C, Honjoh D et al: Two prognostically significant subtypes of high-grade lung neuroendocrine tumours independent of small-cell and large-cell neuroendocrine carcinomas identified by gene expression profiles. Lancet 363:775- 781, 2004 Rohr U-P, Rehfeld N, Pflugfelder L et al: Expression of the tyrosine kinase C-kit is an independent prognostic factor in patients with small cell lung cancer. Int J Cancer 111:159-263, 2004 He P, Varticovski L, Bowman ED, et al: Identification of carboxypeptidase E and g-glutamyl hydrolase as biomarkers for pulmonary neuroendocrine tumors by cDNA microarray. Hum Pathol 35:1196-1209, 2004 Koukourakis MI, Giatromanolaki A, Sivridis E et al: Lactate dehydrogenase-5, LDH-5, overexpression in non-small cell lung cancer tissues is linked to tumour hypoxia, angiogenetic factor production and poor prognosis. Br J Cancer 89:877-885, 2003 Nakashima T, Huan C, Liu D et al: Expression of vascular endothelial growth factor-A and vascular endothelial growth factor-C as prognostic factors for non-small cell lung cancer. Med Sci Monit 10:BR157-165, 2004 Iwasaki A, Kuwahara M, Yoshinaga Y, Shirakusa T: Basic fibroblast growth factor, bFGF, and vascular endothelial growth factor, VEGF, levels, as prognostic indicators in NSCLC. Eur J Cardio-thoracic Surg 25:443-448, 2004 Ogawa E, Takenaka K, Yanagihara K et al: Clinical significance of VEGF-C status in tumor cells and stromal macrophages in non-small cell lung cancer patients. Br J Cancer 91:498-503, 2004 Mineto TC, Ambrogi V, Baldi A et al: Prognostic impact of VEGF, CD31, CD34, and CD105 expression and tumour vessel invasion after radical surgery for IB-IIA non-small cell lung carcinoma. J Clin Pathol 57:591-597, 2004 Tanaka F, Yanagihara K, Otake Y et al: Prognostic factors in resected pathologic, p-, stage IIIA-N2, non-small-cell lung carcinoma. Ann Surg Oncol 11:612-618, 2004 Stefanou D, Batistatou A, Arkoumani E et al: Expression of vascular endothelial growth factor, VEGF, and association with microvessel density in small-cell and non-small cell lung carcinoma. Histol Histopathol 19:37-42, 2004 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2005 VL 11 IS 1 BP 5 EP 10 PG 6 ER PT J AU Orosz, Zs Tornoczky, T Sapi, Z AF Orosz, Zsolt Tornoczky, Tamas Sapi, Zoltan TI Gastrointestinal Stromal Tumors: A Clinicopathologic and Immunohistochemical Study of 136 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE c-kit; CD117; gastrointestinal stromal tumor (GIST); inflammatory fibroid polyp; leiomyoma; leiomyosarcoma; schwannoma ID c-kit; CD117; gastrointestinal stromal tumor (GIST); inflammatory fibroid polyp; leiomyoma; leiomyosarcoma; schwannoma AB The clinicopathologic features of 136 gastrointestinal stromal tumors were analyzed. The tumors occurred in 60 women and 76 men, ranging in age from 19 to 88 years (median 59 years, mean 59.2 years). Sixty-one cases arose from stomach, 38 from small intestine and 11 from colon or rectum. Abdominal cavity was indicated as tumor site in 10 cases, but the extragastrointestinal origin using strict criteria was not proved. Four locally recurrent cases and 12 metastatic samples were also included. The primary and recurrent tumors ranged in size from 0.5 to 30 cm (mean 8.3 cm). The large number of high-grade cases (85 of 112 classifiable) is alarming and emphasize the importance of oncology care. Histologically, ninety-two cases were classified as spindle cell while 11 as epithelioid GIST. Mixed cellularity was seen in 33 cases. Skeinoid fibers were present in 14 and coagulation necrosis in 40 primary cases. Ulceration observed by microscopic examination was common (36 of 110 cases, 32.7%), explaining the clinically frequently observed gastrointestinal bleeding. Unusual histological features such as stromal hyalinization and nuclear palisading were present in 30 and 27 cases, respectively. Immunohistochemical CD117 (c-kit) positivity was documented in 133 cases. Three cases with CD117 negative results were included, because their morphology was most consistent with GIST and immunohistochemical reactions excluded the possibility of other neoplasms. CD34 positivity was seen in 70%, alpha-smooth muscle actin positivity in 39.6% of examined cases. Only one case showed desmin reactivity and seven had S100 positive tumor cells. For h-caldesmon 39 cases proved to be positive (60.9% of the tested cases). C1 [Orosz, Zsolt] National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Tornoczky, Tamas] University of Pecs, Department of PathologyPecs, Hungary. [Sapi, Zoltan] St John's Hospital, Department of PathologyBudapest, Hungary. RP Orosz, Zs (reprint author), National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, H-1122 Budapest, Hungary. EM zso@oncol.hu CR Ceballos KM, Nielsen GP, Selig MK, O’Connell JX: Is anti-hcaldesmon useful for distinguishing smooth muscle and myofibroblastic tumors? An immunohistochemical study. Am J Clin Pathol 114: 746-753, 2000 Corless CL, Fletcher JA, Heinrich MC: Biology of gastrointestinal stromal tumors. J Clin Oncol 22: 3813-3825, 2004 Daum O, Hes O, Vanecek T, et al: Vanek’s tumor, inflammatory fibroid polyp). Report of 18 cases and comparison with three cases of original Vanek’s series. Mod Pathol 16:366-375, 2003 DeMatteo RP, Lewis JJ, Leung D, et al: Two hundred gastrointestinal stromal tumors: recurrence patterns and prognostic factors for survival. Ann Surg 231:51-58, 2000 de Silva CM, Reid R: Gastrointestinal stromal tumors, GIST): C-kit mutations, CD117 expression, differential diagnosis and targeted cancer therapy with Imatinib. Pathol Oncol Res 9:13- 19, 2003 Eckhardt S, Papai Zs, Bodoky Gy, et al: Az imatinib kezeles hatasa gastrointestinalis stroma eredetu daganatokban [Effect of imatinib treatment of gastrointestinal stromal tumors] Orv Hetil 144:2207-2212, 2003 Emile JF, Theou N, Tabone S, et al: Clinicopathologic, phenotypic, and genotypic characteristics of gastrointestinal mesenchymal tumors. Clin Gastroenterol Hepatol 2:597-605, 2004 Fletcher CDM, Berman JJ, Corless C, et al: Diagnosis of gastrointestinal stromal tumors: A consensus approach. Hum Pathol 33: 459-465, 2002 Fujimoto Y, Nakanishi Y, Yoshimura K, Shimoda T: Clinicopathologic study of primary malignant gastrointestinal stromal tumor of the stomach, with special reference to prognostic factors: analysis of results in 140 surgically resected patients. Gastric Cancer 6: 39-48, 2003 Greenson JK: Gastrointestinal stromal tumors and other mesenchymal lesions of the gut. Mod Pathol 16: 366-375, 2003 Hasegawa T, Matsuno Y, Shimoda T, Hirohashi S: Gastrointestinal stromal tumor: consistent CD117 immunostaining for diagnosis, and prognostic classification based on tumor size and MIB-1 grade. Hum Pathol 33: 669-676, 2002 Hirota S, Isozaki K, Moriyama Y, et al: Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 279: 577–580, 1998 Hjermstad BM, Sobin LH, Helwig EB: Stromal tumors of the gastrointestinal tract: myogenic or neurogenic? Am J Surg Pathol 11:383-386, 1987 Joensuu H, Roberts PJ, Sarlomo-Rikala M, et al: Effect of the tyrosine kinase inhibitor STI571 in a patient with a metastatic gastrointestinal stromal tumor. N Engl J Med. 344:1052-1056, 2001 Kindblom LG, Remotti HE, Aldenborg F, Meis-Kindblom JM: Gastrointestinal pacemaker cell tumor, GIPACT): gastrointestinal stromal tumors show phenotypic characteristics of the interstitial cells of Cajal. Am J Pathol 152: 1259-1269, 1998 Kwon MS, Lee SS, Ahn GH: Schwannomas of the gastrointestinal tract: clinicopathological features of 12 cases including a case of esophageal tumor compared with those of gastrointestinal stromal tumors and leiomyomas of the gastrointestinal tract. Pathol Res Pract 198: 605-613, 2002 Lasota J, Wasag B, Dansonka-Mieszkowska A, et al: Evaluation of NF2 and NF1 tumor suppressor genes in distinctive gastrointestinal nerve sheath tumors traditionally diagnosed as benign schwannomas: s study of 20 cases. Lab Invest. 83: 1361-71, 2003 Lee JR, Joshi V, Griffin JW Jr, et al: Gastrointestinal autonomic nerve tumor: immunohistochemical and molecular identity with gastrointestinal stromal tumor. Am J Surg Pathol 25: 979- 987, 2001 Mazur MT, Clark HB. Gastric stromal tumors: reappraisal of histogenesis. Am J Surg Pathol 7:507-519, 1983 Medeiros F, Corless CL, Duensing A, et al: KIT-negative gastrointestinal stromal tumors: proof of concept and therapeutic implications. Am J Surg Pathol 28:889-894, 2004 Miettinen M, Furlong M, Sarlomo-Rikala M, et al: Gastrointestinal stromal tumors, intramural leiomyomas, and leiomyosarcomas in the rectum and anus: a clinicopathologic, immunohistochemical, and molecular genetic study of 144 cases. Am J Surg Pathol 25:1121-1133, 2001 Miettinen M, Kopczynski J, Makhlouf HR, et al: Gastrointestinal stromal tumors, intramural leiomyomas, and leiomyosarcomas in the duodenum: a clinicopathologic, immunohistochemical, and molecular genetic study of 167 cases. Am J Surg Pathol 27:625-641, 2003 Miettinen M, Lasota J: Gastrointestinal stromal tumors – definition, clinical, histological, immunohistochemical, and molecular genetic features and differential diagnosis. Virchows Arch 438:1-12, 2001 Miettinen M, Shekitka KM, Sobin LH: Schwannomas in the colon and rectum: a clinicopathologic and immunohistochemical study of 20 cases. Am J Surg Pathol 25: 846-855, 2001 Miettinen M, Sobin LH, Sarlomo-Rikala M: Immunohistochemical spectrum of GISTs at different sites a their differential diagnosis with a reference to CD117, KIT). Mod Pathol 13: 1134-1142, 2000 Miettinen M, Virolainen M, Sarlomo-Rikala M: Gastrointestinal stromal tumors: Value of CD34 antigen in their identification and separation from true leiomyomas and schwannomas. Am J Surg Pathol 19:207-216, 1995 Mikhael AI, Bacchi CE, Zarbo RJ, et al: CD34 expression in stromal tumors of the gastrointestinal tract. Appl Immunohistochem 2:89-93, 1994 Reith, JD, Goldblum JR, Lyles RH, Weiss SW: Extragastrointestinal, soft tissue, stromal tumors: Analysis of 48 cases with emphasis on histologic predictors of outcome. Mod Pathol 13:577–585, 2000 Rudolph P, Chiaravalli AM, Pauser U, et al: Gastrointestinal mesenchymal tumors - immunophenotypic classification and survival analysis. Virchows Arch 441: 238-248, 2002 Sapi Z, Kovacs RB, Bodo M: Gastrointestinal stromal tumors. Observations on the basis of 29 cases Orv Hetil 142: 2479- 2485, 2001 Stout AP: Bizarre smooth muscle tumors of the stomach. Cancer 15:400-409, 1962 Tornoczky T, Kover E, Pajor L: Frequent occurrence of low grade cases among metastatic gastrointestinal stromal tumours J Clin Pathol 56: 363–367, 2003 West RB, Corless CL, Chen X, et al: The novel marker, DOG1, is expressed ubiquitously in gastrointestinal stromal tumors irrespective of KIT or PDGFRA mutation status. Am J Pathol 165: 107-113, 2004 Wong NA, Young R, Malcomson RD, et al: Prognostic indicators for gastrointestinal stromal tumours: a clinicopathological and immunohistochemical study of 108 resected cases of the stomach. Histopathology. 43:118-126, 2003 Yamaguchi U, Hasegawa T, Masuda T, et al: Differential diagnosis of gastrointestinal stromal tumor and other spindle cell tumors in the gastrointestinal tract based on immunohistochemical analysis. Virchows Arch 445:142-150, 2004 Yantiss RK, Spiro IJ, Compton CC, Rosenberg AE: Gastrointestinal stromal tumor versus intra-abdominal fibromatosis of the bowel wall: a clinically important differential diagnosis. Am J Surg Pathol 24:947-957, 2000 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2005 VL 11 IS 1 BP 11 EP 21 PG 11 ER PT J AU Demokan, S Demir, D Suoglu, Y Kiyak, E Akar, U Dalay, N AF Demokan, Semra Demir, Deniz Suoglu, Yusufhan Kiyak, Erkan Akar, Ugur Dalay, Nejat TI Polymorphisms of the XRCC1 DNA Repair Gene in Head and Neck Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE polymorphism; XRCC1; head and neck cancer ID polymorphism; XRCC1; head and neck cancer AB Inherited polymorphisms in the genes controlling the cell cycle or functioning in the DNA repair mechanisms may impair their function and contribute to genetic susceptibility. Abnormalities in the DNA repair have been reported in head and neck cancer. The XRCC1 gene functions in singlestrand break and base excision repair processes. In this study, two polymorphisms of the XRCC1 gene, Arg194Trp and Arg399Gln were investigated in 95 patients with head and neck carcinoma. The polymorphic regions were amplified by PCR followed by digestion with methylation-specific restriction enzymes, and analyzed electrophoretically. Genotype and allele frequencies were calculated, and association with cancer risk or clinical parameters was investigated. No association was observed between the genotypes and head and neck cancer for either polymorphism. Distribution of the alleles did not significantly differ between the patients and the control group. A significant association was only found for the Trp194 allele among the smoking individuals. Our data indicate that the Arg194Trp and Arg399Gln polymorphisms do not confer a significant risk for head and neck carcinogenesis. C1 [Demokan, Semra] Istanbul Medical Faculty, Istanbul University, Oncology Institute, Capa, 34390 Istanbul, Turkey. [Demir, Deniz] Istanbul Medical Faculty, Istanbul University, Department of OtolaryngologyIstanbul, Turkey. [Suoglu, Yusufhan] Istanbul Medical Faculty, Istanbul University, Department of OtolaryngologyIstanbul, Turkey. [Kiyak, Erkan] Istanbul Medical Faculty, Istanbul University, Department of OtolaryngologyIstanbul, Turkey. [Akar, Ugur] Istanbul Medical Faculty, Istanbul University, Department of Medical BiologyIstanbul, Turkey. [Dalay, Nejat] Istanbul Medical Faculty, Istanbul University, Oncology Institute, Capa, 34390 Istanbul, Turkey. RP Dalay, N (reprint author), Istanbul Medical Faculty, Istanbul University, Oncology Institute, 34390 Istanbul, Turkey. EM ndalay@yahoo.com CR Abdel-Rahman SZ, Soliman AS, Bondy ML, et al: Inheritance of the 194Trp and the 399Gln variant alleles of the DNA repair gene XRCC1 are associated with increased risk of early-onset colorectal carcinoma in Egypt. Cancer Lett 159: 79-86, 2000 Bohr VA: DNA repair fine structure and its relations to genomic instability. Carcinogenesis 16: 2885-2892, 1995 Butkiewicz D, Rusin M, Enewold L, et al:Genetic polymorphisms in DNA repair genes and risk of lung cancer. Carcinogenesis 22: 593-597, 2001 Caldecott KW, Aoufouchi S, Johnson P, Shall S: XRCC1 polypeptide interacts with DNA polymerase beta and possibly poly, ADPribose, polymerase, and DNA ligase III is a novel molecular ‘nicksensor’ in vitro. Nucleic Acids Res 24: 4387-4394, 1996 Cappelli E, Taylor R, Cevasco M, et al: Involvement of XRCC1 and DNA ligase III gene products in DNA base excision repair. J Biol Chem 272: 23970-23975, 1997 Crowe DL: Molecular pathology of head and neck cancer. Histol Histopathol 17: 909-914, 2002 David-Beabes GL, London SJ:Genetic polymorphism of XRCC1 and lung cancer risk among African-Americans and Caucasians. Lung Cancer 34: 333-339, 2001 Divine KK, Gilliland FD, Crowell RE, et al:The XRCC1 399 glutamine allele is a risk factor for adenocarcinoma of the lung. Mutat Res 461: 273-278, 2001 Duell EJ, Millikan RC, Pittman GS, et al: Polymorphisms in the DNA repair gene XRCC1 and breast cancer. Cancer Epidemiol Biomarkers Prev 10: 217-222, 2001 Goode EL, Ulrich CM, Potter JD:Polymorphisms in DNA repair genes and associations with cancer risk. Cancer Epidemiol Biomarkers Prev 11: 1513-1530, 2002 Hu JJ, Mohrenweiser HW, Bell DA, et al: Symposium Overview: Genetic polymorphisms in DNA repair and cancer risk. Toxicol Appl Pharmacol 185: 64-73, 2002 Kubota Y, Nash RA, Klungland A, et al: Reconstitution of DNA base excision-repair with purified human proteins: interaction between DNA polymerase beta and the XRCC1 protein. EMBO J 15: 6662-6670, 1996 Lee SG, Kim B, Choi J, et al:Genetic polymorphisms of XRCC1 and risk of gastric cancer. Cancer Lett 187: 53-60, 2002 Miller MC, Mohrenweiser HW, Bell DA: Genetic variability in susceptibility and response to toxicants. Toxicol Lett 120: 269- 280, 2001 Misra RR, Ratnasinghe D, Tangrea JA, et al: Polymorphisms in the DNA repair genes XPD, XRCC1, XRCC3, and APE/ref-1, and the risk of lung cancer among male smokers in Finland. Cancer Lett 191:171-178, 2003 Nelson HH, Kelsey KT, Mott LA, Karagas MR: The XRCC1 Arg399Gln polymorphism, sunburn, and non-melanoma skin cancer: evidence of gene-environment interaction. Cancer Res 62:152-155, 2002 Olshan AF, Watson MA, Weissler MC, Bell DA:XRCC1 polymorphisms and head and neck cancer. Cancer Lett 178: 181-186, 2002 Park JY, Muscat JE, Ren Q, et al: CYP1A1 and GSTM1 polymorphisms and oral cancer risk. Cancer Epidemiol Biomarkers Prev 6: 791-797, 1997 Ratnasinghe D, Yao SX, Tangrea JA, et al:Polymorphisms of the DNA repair gene XRCC1 and lung cancer risk. Cancer Epidemiol Biomarkers Prev 10:119-123, 2001 Scully C, Field JK, Tanzawa H: Genetic aberrations in oral or head and neck squamous cell carcinoma, SCCHN): 1. Carcinogen metabolism, DNA repair and cell cycle control. Oral Oncol 36: 256-263, 2000 Scully C, Field JK, Tanzawa H: Genetic aberrations in oral or head and neck squamous cell carcinoma: 2. Chromosomal aberrations. Oral Oncol 36: 311-327, 2000 Shen MR, Jones IM, Mohrenweiser H: Nonconservative amino acid substitution variants exist at polymorphic frequency in DNA repair genes in healthy humans. Cancer Res 58: 604–608, 1998 Shen H, Xu Y, Qian Y, et al: Polymorphisms of the DNA repair gene XRCC1 and risk of gastric cancer in a Chinese population. Int J Cancer 88:601-606, 2000 Shu XO, Cai Q, Gao YT, et al: A population based case control study of the Arg399Gln polymorphism in DNA repair gene XRCC1 and risk of breast cancer. Cancer Epidemiol Biomarkers Prev 12: 1462-1467, 2003 Stern MC, Umbach DM, van Gils CH, et al: DNA repair gene XRCC1 polymorphisms, smoking, and bladder cancer risk. Cancer Epidemiol Biomarkers Prev 10:125-131, 2001 Sturgis EM, Castillo EJ, Li L, et al: Polymorphisms of DNA repair gene XRCC1 in squamous cell carcinoma of the head and neck. Carcinogenesis 20: 2125-2129, 1999 Sturgis EM, Wei Q:Genetic susceptibility-molecular epidemiology of head and neck cancer. Curr Opin Oncol 14: 310-317, 2002 Taningher M, Malacarne D, Izzotti A, et al: Drug metabolism polymorphisms as modulators of cancer susceptibility. Mutat Res 436: 227–261, 1999 Xing D, QI J, Miao X, et al:Polymorphisms of DNA repair genes XRCC1 and XPD and their associations with risk of esophageal squamous cell carcinoma in a Chinese population. Int J Cancer 100: 600-605, 2002 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2005 VL 11 IS 1 BP 22 EP 25 PG 4 ER PT J AU Sobel, G Szabo, I Paska, Cs Kiss, A Kovalszky, I Kadar, A Paulin, F Schaff, Zs AF Sobel, Gabor Szabo, Istvan Paska, Csilla Kiss, Andras Kovalszky, Ilona Kadar, Anna Paulin, Ferenc Schaff, Zsuzsa TI Changes of Cell Adhesion and Extracellular Matrix (ECM) Components in Cervical Intraepithelial Neoplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE occludin; claudin; cervical intraepithelial neoplasia; syndecan-1; carcinogenesis ID occludin; claudin; cervical intraepithelial neoplasia; syndecan-1; carcinogenesis AB Cell-cell and cell-extracellular matrix interaction is crucial in tumor progression. Tight junction (TJ) proteins as occludin and claudins (CLDNs) play important role in this process together with several extracellular matrix components, as syndecan. Our previous work suggested significant changes in the expression of claudins even in the early stages of cervical carcinogenesis. The aim of our present work was to study the expression of occludin and syndecan-1, as compared to CLDNs, in early phases of cervical carcinogenesis. Paraffin sections of 50 samples were studied by immunohistochemistry, including cervical intraepithelial neoplasias (CINI-II-III), in situ carcinomas (CIS) and normal cervical samples. Occludin and CLDN-2 were found colocalized in the basal layer, while syndecan-1 and CLDN-1, -4 and -7 were coexpressed in the parabasal and intermedier layers in normal epithelia. Intensity of occludin staining decreased in CIN/CIS lesions, although it was more extended towards the upper epithelial layers with inverse relation with grades, as seen in the case of CLDN-2 expression. CLDN-1, -2, -4 and -7 were detected in the entire epithelium in CIN, showing decrease in CIS. The progression of CIN was associated with reduced syndecan-1 expression, in contrast to CLDN-1, -4 and -7 which increased toward CIS. The obtained data suggest that significant changes occur in the composition of cell adhesion complexes even in early stages of cervical carcinogenesis. The pattern of expression is characteristic for the alteration, the changes in the different components, however, are not parallel with each other. C1 [Sobel, Gabor] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Szabo, Istvan] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Paska, Csilla] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kadar, Anna] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Paulin, Ferenc] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. CR Acharya P, Beckel J, Ruiz WG, et al: Distribution of the tight junction proteins ZO-1, occludin, and claudin-4, -8, and -12 in bladder epithelium. Am J Physiol Renal Physiol 287: F305-F318, 2004 Anttonen A, Kajanti M, Heikkila P, et al: Syndecan-1 expression has a prognostic significance in head and neck carcinoma. Br J Cancer 79: 558-564, 1999 Anttonen A, Heikkila P, Kajanti M, et al: High syndecan-1 expression is associated with favourable outcome in squamous cell lung carcinoma treated with radical surgery. Lung Cancer 32: 297-305, 2001 Bazer-Garner IB, Dilaz B, Sanderson RD and Smoller BR: Syndecan- 1 expression is decreased with increasing aggressiveness of basal cell carcinoma. Am J Dermatopathol 22: 119-122, 2000 Billings SD, Walsh SV, Fisher C, et al: Aberrant expression of tight junction-related proteins ZO-1, claudin-1 and occludin in synovial sarcoma: an immunohistochemical study with ultrastructural correlation. Mod Pathol 17: 141-149, 2004 Chiba H, Gotoh T, Kojima T, et al: Hepatocyte nuclear factor, HNF)-4a triggers formation of functional tight junctions and establishment of polarized epithelial morphology in F9 embryonal carcinoma cells. Exp Cell Res 286: 288-297, 2003 Fujiya M, Watari J, Ashida T, et al: Reduced expression of syndecan1 affects metastatic potential and clinical outcome in patients with colorectal cancer. Jpn J Cancer Res 92: 1078- 1081, 2001 Furuse M, Fujita K, Hiiragi T, et al: Claudin-1 and -2: Novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol 141: 1539-1550, 1998 Giroglou T, Florin L, Schafer F, et al: Human papillomavirus infection requires cell surface heparan sulfate. J Virol 75: 1565- 1570, 2001 Gumbiner BM: Cell adhesion: The molecular basis of tissue architecture and morphogenesis. Cell 84: 345-357, 1996 Hirohashi S, Kanai Y: Cell adhesion system and human cancer morphogenesis. Cancer Sci 94: 575-581, 2003 Hodivala KJ, Watt FM: Evidence that cadherins play a role in the downregulation of integrin expression that occurs during keratinocyte terminal differentiation. J Cell Biol 124: 589-600, 1994 Hughes DE, Rebello G, Al-Nafussi A: Integrin expression in squamous neoplasia of the cervix. J Pathol 173: 97-104, 1994 Inki P, Stenback F, Grenman S, Jalkanen M: Immunohistochemical localization of syndecan in normal and pathological human uterine cervix. J Pathol 172: 349-355, 1994 Johnson LG: Applications of imaging techniques to studies of epithelial tight junctions. Adv Drug Deliver Rev 57: 111-121, 2005 Kominsky SL, Argani P, Korz D, et al: Loss of the tight junction protein claudin-7 correlates with histological grade in both ductal carcinoma in situ and invasive ductal carcinoma of the breast. Oncogene 22: 2021-2033, 2003 Langbein L, Grund C, Kuhn C, et al: Tight junctions and compositionally related junctional structures in mammalian stratified epithelia and cell cultures derived therefrom. Europ J Cell Biol 81: 419-435, 2002 Litvinov SV, van Driel W, van Rhijn CM, et al: Expression of Ep- CAM in cervical squamous epithelia correlates with an increased proliferation and the disappearance of markers for terminal differentiation. Am J Pathol 148: 865-875, 1996 Mikami S, Ohashi K, Usui Y, et al: Loss of syndecan-1 and increased expression of heparanase in invasive esophageal carcinomas. Jpn J Cancer Res 92: 1062-1073, 2001 Miwa N, Furuse M, Tsukita S, et al: Involvement of claudin-1 in the beta-catenin/Tcf signaling pathway and its frequent upregulation in human colorectal cancers. Oncol Res 12: 469- 476, 2000 Morita K, Tsukita S, Miyachi Y: Tight junction-associated proteins, occludin, ZO-1, claudin-1, claudin-4, in squamous cell carcinoma and Bowen’s disease. Br J Dermatol 151: 328-334, 2004 Nakanishi K, Yoshioka N, Oka K, Hakura A: Reduction of syndecan- 1 mRNA in cervical-carcinoma cells is involved with the 3’ untranslated region. Int J Cancer 80: 527-532, 1999 Nichols LS, Ashfaq R and Iacobuzio-Donahue CA: Claudin 4 protein expression in primary and metastatic pancreatic cancer: support for use as a therapeutic target. Am J Clin Pathol 121: 226-230, 2004 Numa F, Hirabayashi K, Kawasaki K, et al: Syndecan-1 expression in cancer of the uterine cervix: association with lymph node metastasis. Int J Oncol 20: 39-43, 2002 Paska C, Bogi K, Szilak L, et al: Effect of formalin, acetone and RNAlater fixatives on tissue preservation and different size amplicons by real-time PCR from paraffin-embedded tissue. Diagn Mol Path 13: 234-240, 2004 Peppi M, Ghabriel MN: Tissue specific expression of the tight junction proteins claudins and occludin in the rat salivary glands. J Anat 205: 257-266, 2004 Rangel LB, Agarwal R, D’Souza T, et al: Tight junction proteins claudin-3 and claudin-4 are frequently overexpressed in ovarian cancer but not in ovarian cystadenomas. Clin Cancer Res 9: 2567-2575, 2003 Rintala J, Inki P, Klemi P, et al: Association of syndecan-1 with tumor grade and histology in primary invasive cervical carcinoma. Gynecol Oncol 75: 372-378, 1999 Schneeberger EE, Lynch RD: The tight junction: a multifunctional complex. Am J Physiol Cell Physiol 286: C1213-C1228, 2004 Shafti-Keramat S, Handisurya A, Kriehuber E, et al: Different heparan sulfate proteoglycans serve as cellular receptors for human papillomaviruses. J Virol 77: 13125-13135, 2003 Sobel G, Szabo I, Kiss A, et al: Increased expression of claudins in cervical squamous intraepithelial neoplasia and invasive carcinoma. Hum Pathol 36: 162-169, 2005 Timar J, Lapis K, Dudas J, et al: Proteoglycans and tumor progression: Janus-faced molecules with contradictory functions in cancer. Semin Cancer Biol 12: 173-186, 2002 Tobioka H, Isomura H, Kokai Y, et al: Occludin expression decreases with the progression of human endometrial carcinoma. Hum Pathol 35: 159-164, 2004 Tobioka H, Tokunaga Y, Isomura H, et al: Expression of occludin, a tight-junction-associated protein, in human lung carcinomas. Virchows Arch 445: 472-476, 2004 Tokes A-M, Kulka J, Paku S, et al: Claudin-1, -3 and -4 proteins and mRNA expression in benign and malignant breast lesions: a research study. Breast Cancer Res 7: R296-R305, 2005 Tsukita S, Furuse M, Itoh M: Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol 2: 285-293, 2001 Turksen K, Troy T-C: Barriers built on claudins. J Cell Sci 117: 2435-2447, 2004 Wiksten JP, Lundin J, Nordling S, et al: Epithelial and stromal syndecan-1 expression as predictor of outcome in patients with gastric cancer. Int J Cancer 95: 1-6, 2001 Woodworth CD, Waggoner S, Barnes W, et al: Human cervical and foreskin epithelial cells immortalized by human papillomavirus DNAs exhibit dysplastic differentiation in vivo. Cancer Res 50: 3709-3715, 1990 Yang Y, Yaccoby S, Liu W, et al: Soluble syndecan-1 promotes growth of myeloma tumors in vivo. Blood 100: 610-617, 2002 Zheng JY, Yu D, Foroohar M, et al: Regulation of the expression of the prostate-specific antigen by claudin-7. J Membr Biol 194: 187-197, 2003 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2005 VL 11 IS 1 BP 26 EP 31 PG 6 ER PT J AU Bircan, S Ensari, A Ozturk, S Erdogan, N Dundar, I Ortac, F AF Bircan, Sema Ensari, Arzu Ozturk, Sibel Erdogan, Nural Dundar, Ilkkan Ortac, Firat TI Immunohistochemical Analysis of c-Myc, c-Jun and Estrogen Receptor in Normal, Hyperplastic and Neoplastic Endometrium SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE human endometrium; estrogen receptor; c-myc; c-jun ID human endometrium; estrogen receptor; c-myc; c-jun AB To evaluate the role of c-jun and c-myc proto-oncogenes in normal, hyperplastic and neoplastic endometrium in relation to estrogen receptor (ER) status and to investigate whether these genes can be related to other histopathological features of endometrial carcinoma, 32 endometrial carcinomas, 38 endometrial hyperplasias and 22 cyclic endometria (10 proliferative and 12 secretory) were evaluated histologically. Endometrial hyperplasia cases were classified as simple and complex hyperplasia without atypia, and atypical hyperplasia. Endometrial carcinoma cases were subtyped according to the International Society of Gynecological Pathologists. Modified FIGO system was used for both grading and staging. Immunohistochemical examination was performed using antibodies to ER-alpha, c-myc and c-jun with streptavidin-biotin-peroxidase technique. The mean percentage of ER-alpha positive cells changed cyclically during the menstrual cycle, and it was the highest (96%) and the lowest (31.6%) in proliferative and carcinomatous endometrium, respectively. There was a statistically significant difference between proliferative and secretory phases and proliferative and carcinomatous endometrium in relation to ER-alpha staining (p<0.05). There was also a statistically significant difference with respect to ERalpha reactivity between secretory phase and each hyperplastic group, as well as between the carcinoma group and each hyperplastic group (p<0.05). Although not significant, the mean percentage of c-myc expressing cells in the carcinoma group was higher (15.3%) than that of proliferative phase and hyperplastic groups. The mean percentage of c-jun positive cells in proliferative endometrium was slightly higher than in secretory endometrium, and it was the highest in atypical hyperplastic endometrium (28.3%), but there was no statistically significant difference between the groups. In carcinoma cases, a positive correlation was observed between c-jun positivity and tumor grade (p=0.027, r=0.3908), but such a correlation with c-myc was not found. A positive correlation was detected between ER-alpha and c-myc expression (p=0.038, r=0.3686). A progressive loss of ER seems to be correlated with increasing malignant transformation. C-myc expression might play a role in the development of endometrial carcinoma via ER. The association between c-jun and ER appears to be lost in endometrial carcinoma. The relationship between c-myc, c-jun and ER appears to be altered in endometrial carcinoma compared to that of menstrual endometrium. C1 [Bircan, Sema] Ankara University, School of Medicine, Department of Pathology, Suleyman Demirel Universitesi, Tip Fakultesi, Patoloji Anabilim Dali, IspartaAnkara, Turkey. [Ensari, Arzu] Ankara University, School of Medicine, Department of Pathology, Suleyman Demirel Universitesi, Tip Fakultesi, Patoloji Anabilim Dali, IspartaAnkara, Turkey. [Ozturk, Sibel] Ankara University, School of Medicine, Department of Pathology, Suleyman Demirel Universitesi, Tip Fakultesi, Patoloji Anabilim Dali, IspartaAnkara, Turkey. [Erdogan, Nural] Ankara University, School of Medicine, Department of Pathology, Suleyman Demirel Universitesi, Tip Fakultesi, Patoloji Anabilim Dali, IspartaAnkara, Turkey. [Dundar, Ilkkan] School of Medicine, University of Ankara, Department of Gynecological OncologyAnkara, Turkey. [Ortac, Firat] School of Medicine, University of Ankara, Department of Gynecological OncologyAnkara, Turkey. RP Bircan, S (reprint author), Ankara University, School of Medicine, Department of Pathology, Ankara, Turkey. EM bircans2000@yahoo.com CR Nephew KP, Choi CM, Polek TC, et al: Expression of fos and jun proto-oncogenes in benign versus malignant human uterine tissue. Gynecol Oncol 76: 388-396, 2000 Shiozawa T, Miyamoto T, Kashima H, et al: Estrogen-induced proliferation of normal endometrial glandular cells is initiated by transcriptional activation of cyclin D1 via binding of c-jun to an AP-1 sequence. Oncogene 23: 8603-8610, 2004 Udou T, Hachisuga T, Tsujioka H, Kawarabayashi T: The role of c-jun protein in proliferation and apoptosis of the endometrium throughout the menstrual cycle. Gynecol Obstet Invest 57: 121-126, 2004 Yamashita S, Takayanagi A, Shimizu N: Temporal and cell-type specific expression of c-fos and c-jun protooncogenes in the mouse uterus after estrogen stimulation. Endocrinology 137: 5468-5475, 1996 Subramaniam M, Harris SA, Rasmussen K, Spelsberg TC: Rapid down-regulation of c-jun protooncogene transcription by progesterone in the avian oviduct. Endocrinology 133: 2049- 2054, 1993 Webb DK, Moulton BC, Khan SA: Estrogen induced expression of the c-jun proto-oncogene in the immature and mature rat uterus. Biochem Biophys Res Commun 168: 721-726, 1990 Morishita S, Niwa K, Ichigo S, et al: Overexpressions of cfos/ jun mRNA and their oncoproteins, Fos/Jun, in the mouse uterus treated with three natural estrogens. Cancer Lett 97: 225- 231, 1995 Fujimoto J, Hori M, Ichigo S, et al: Clinical implication of fos and jun expressions and protein kinase activity in endometrial cancers. Eur J Gynaecol Oncol 16: 138-146, 1995 Kumar V, Abbas AK, Fausto N: Robbins and Cotran Pathologic Basis of Disease. Elsevier Saunders, Philadelphia, 2005 Salmi A, Heikkila P, Lintula S, Rutanen EM: Cellular localization of c-jun messenger ribonucleic acid and protein and their relation to the proliferation marker Ki-67 in the human endometrium. J Clin Endocrinol Metab 83: 1788-1796, 1998 Wisdom R, Johnson RS, Moore C: C-Jun regulates cell cycle progression and apoptosis by distinct mechanisms. EMBO J 18: 188-197, 1999 Vogt PK, Bos TJ: The oncogene jun and nuclear signaling. TIBS 14: 172-175, 1989 Travers MT, Knowler JT: Oestrogen-induced expression of oncogenes in the immature rat uterus. FEBS Letters 211: 27- 30, 1987 Huet-Hudson YM, Andrews GK, Dey SK: Cell type-specific localization of c-myc protein in the mouse uterus: modulation by steroid hormones and analysis of the periimplantation period. Endocrinology 125: 1683-1690, 1989 Cole MD: The myc oncogene: its role in transformation and differentiation. Ann Rev Genet 20:361-384, 1986 Fujimoto J, Hori M, Ichigo S, et al: Tissue differences in the expression of mRNAs of Ha-ras, c-myc, fos and jun in human uterine endometrium, myometrium and leiomyoma under the influence of estrogen/progesterone. Tumor Biol 15: 311-317, 1994 Salmi A, Carpen O, Rutanen E: The association between c-fos and c-jun expression and estrogen and progesterone receptors is lost in human endometrial cancer. Tumor Biol 20: 202-211, 1999 Murphy LJ, Murphy LC, Friesen HG: Estrogen induction of Nmyc and c-myc proto-oncogene expression in the rat uterus. Endocrinology 120: 1882-1888, 1987 Silverberg SG: Hyperplasia and carcinoma of the endometrium. Semin Diagn Pathol 5: 135-153, 1988 Silverberg SG, Kurman RJ: Tumors of the uterine corpus and gestational trophoblastic disease. AFIP, Washington DC, 1992 Mikuta JJ: International Federation of Gynecology and Obstetrics Staging of Endometrial Cancer 1988. Cancer 71: 1460- 1463, 1993 Salmi A, Rutanen F: C-fos and c-jun expression in human endometrium and myometrium. Mol Cell Endocrinol 117: 233-240, 1996 Thornton JG, Wells M: Oestrogen receptor in glands and stroma of normal and neoplastic human endometrium: a combined biochemical, immunohistochemical, and morphometric study. J Clin Pathol 40: 1437-1442, 1987 Huang SJ, Cheng L, Lewin KJ, Fu YS: Immunohistochemical estrogen receptor assessment in hyperplastic, neoplastic, and physiologic endometria. Pathol Res Pract 187: 487-495, 1991 Papadimitriou CS, Athanasiadou S, Stylianidou A, et al: Immnohistochemical detection of estrogen receptors on paraffin sections of normal, hyperplastic and carcinomatous endometrium. Oncology 49:196-202, 1992 Nunobiki O, Taniguchi E, Ishii A, et al: Significance of hormone receptor status and tumor vessels in normal, hyperplastic and neoplastic endometrium. Pathol Int 53: 846-852, 2003 Neis KJ, Brandner P, Ulrich-Winkelspecht K: The oestrogen receptor, ER, in normal and abnormal uterine tissue. Eur J Cancer 36: S27-36, 2000 Uchikawa J, Shiozawa T, Shih H, et al: Expression of steroid receptor coactivators and corepressors in human endometrial hyperplasia and carcinoma with relevance to steroid receptors and Ki-67 expression. Cancer 98: 2207-2213, 2003 Snijders MP, De Goeij AF, Koudstaal J, et al: Oestrogen and progesterone receptor immunocytochemistry in human hyperplastic and neoplastic endometrium. J Pathol 166: 171-177, 1992 Punnonen R, Mattila J, Kuoppala T, Koivula T: DNA ploidy, cell proliferation and steroid hormone receptors in endometrial hyperplasia and early adenocarcinoma. J Cancer Res Clin Oncol 119: 426-429, 1993 Sivridis E, Giatromanolaki A: Prognostic aspects on endometrial hyperplasia and neoplasia. Virchows Arch 439: 118-126, 2001 Carcangiu ML, Chambers JT, Voynick IM, et al: Immunohistochemical evaluation of estrogen and progesterone receptor content in 183 patients with endometrial carcinoma. Part I: Clinical and histologic correlations. Am J Clin Pathol 94: 247-254, 1990. Chambers JT, Carcangiu ML, Voynick IM, Schwartz PE: Immunohistochemical evaluation of estrogen and progesterone receptor content in 183 patients with endometrial carcinoma. Part II: Correlation between biochemical and immunohistochemical methods and survival. Am J Clin Pathol 94: 255-260, 1990 Bigsby RM, Li A: Differentially regulated immediate early genes in the rat uterus. Endocrinology 134: 1820-1826, 1994 Nephew KP, Tang M, Khan SA: Estrogen differentially affects c-jun expression in uterine tissue compartments. Endocrinology 134: 1827-1834, 1994 Yokoyama Y, Sagara M, Sato S, Saito Y: Value of glutathione Stransferase π and the oncogene products c-jun, c-fos, H-ras, and c-myc as a prognostic indicator in endometrial carcinomas. Gynecol Oncol 68: 280-287, 1998 Odom LD, Barrett JM, Pantazis CG, et al: Immunocytochemical study of ras and myc proto-oncogene polypeptide expression in the human menstrual cycle. Am J Obstet Gynecol 161: 1663-1668, 1989 Jack AS, Kerr IB, Evan G, Lee FD: The distribution of the cmyc oncogene product in malignant lymphomas and various normal tissues as demonstrated by immunocytochemistry. Br J Cancer 53: 713-719, 1986 Schenken RS, Johnson JV, Riehl RM: C-myc protooncogene polypeptide expression in endometriosis. Am J Obstet Gynecol 164: 1031-1037, 1991 Bai MK, Costopoulos JS, Christoforidou BP, Papadimitriou CS: Immunohistochemical detection of the c-myc oncogene product in normal, hyperplastic and carcinomatous endometrium. Oncology 51: 314-319, 1994 Loke SL, Neckers LM, Schwab G, Jaffe ES: C-myc protein in normal tissue: Effects of fixation on its apparent subcellular disribution. Am J Pathol 131: 29-37, 1988 Sato S, Ito K, Ozawa N, Yajima A, Sasano H: Expression of cmyc, epidermal growth factor receptor and c-erbB-2 in human endometrial carcinoma and cervical adenocarcinoma. Tohoku J Exp Med 165: 137-145, 1991 Watson JV, Curling OM, Munn CF, Hudson CN: Oncogene expression in ovarian cancer: A pilot study of c-myc oncoprotein in serous papillary cancer. Gynecol Oncol 28: 137-150, 1987 Monk BJ, Chapman JA, Johnson GA, et al: Correlation of cmyc and HER-2/neu amplification and expression with histopathologic variables in uterine corpus cancer. Am J Obstet Gynecol 171: 1193-1198, 1994 Borst MP, Baker VV, Dixon D, et al: Oncogene alterations in endometrial carcinoma. Gynecol Oncol 38: 364-366, 1990 Ambros RA: C-myc gene expression in stage I endometrioid adenocarcinoma of the uterus. Materia Medica Polona 24: 76- 78, 1992 Geisler JP, Geisler HE, Manahan KJ, et al: Nuclear and cytoplasmic c-myc staining in endometrial carcinoma and their relationship to survival. Int J Gynecol Cancer 14: 133-137, 2004 Sasano H, Comerford J, Wilkinson DS, et al: Serous papillary adenocarcinoma of the endometrium. Analysis of proto-oncogene amplification, flow cytometry, estrogen and progesterone receptors, and immunohistochemistry. Cancer 65: 1545-1551, 1990 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2005 VL 11 IS 1 BP 32 EP 39 PG 8 ER PT J AU Brnic, Z Gasparov, S Lozo, VP Anic, P Patrlj, L Ramljak, V AF Brnic, Zoran Gasparov, Slavko Lozo, Vladislav Petar Anic, Petar Patrlj, Leonardo Ramljak, Vesna TI Is Quadrant Biopsy Sufficient in Men Likely to Have Advanced Prostate Cancer? Comparison with Extended Biopsy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prostatic neoplasms; needle biopsy ID prostatic neoplasms; needle biopsy AB We hypothesized that quadrant prostate biopsy (QPB) provides sufficient first-line pathological evaluation of patients with presumed advanced prostate cancer (PC). The aim of this study was to investigate whether the reduction of core number in first-line PB from 6-12 to 4 in patients with presumed advanced PC leads to loss of clinically relevant information. We retrospectively studied 113 men that underwent PB, classified in two groups: ''H” (high) and ''L” (low likelihood of having advanced PC), according to PSA, digital rectal and transrectal ultrasound findings. Pathological results of 6-12-core PB and QPB were retrospectively compared for the presence of malignancy, percentage of positive cores, Gleason score (GS), and the presence of high-grade prostatic intraepithelial neoplasia (HGPIN). PC detection rate was not impaired in group H but dropped significantly in group L, and the percentage of positive cores was not significantly changed in group H (p=0.39), but decreased in group L (p=0.04), due to sampling scheme reduction. No HGPIN was missed with QPB in group H, while 2 HGPINs were missed in group L. No significant change in GS in either group was observed (p=0.12, p=0.13) due to reduction to QPB. We conclude that in patients with presumed advanced PC, reduction of the number of cores in PB may be an acceptable diagnostic strategy, but further studies are needed to analyze the impact of PB scheme reduction on other relevant pathological information obtained from PB. C1 [Brnic, Zoran] University Hospital Merkur, Department of Diagnostic and Interventional Radiology, Zajeeva 19, 10000 Zagreb, Croatia. [Gasparov, Slavko] “Merkur” University Hospital, Department of Clinical Pathology and CytologyZagreb, Croatia. [Lozo, Vladislav Petar] Poliklinika Lozo, Department of UltrasoundZadar, Croatia. [Anic, Petar] University Hospital Merkur, Department of Diagnostic and Interventional Radiology, Zajeeva 19, 10000 Zagreb, Croatia. [Patrlj, Leonardo] University Hospital Merkur, Department of Diagnostic and Interventional Radiology, Zajeeva 19, 10000 Zagreb, Croatia. [Ramljak, Vesna] University Hospital for TumorsZagreb, Croatia. RP Brnic, Z (reprint author), University Hospital Merkur, Department of Diagnostic and Interventional Radiology, 10000 Zagreb, Croatia. EM zoran.brnic@zg.t-com.hr CR Aus G, Ahlgren G, Hugosson J, et al: Diagnosis of prostate cancer: optimal number of prostate biopsies related to serum prostate-specific antigen and findings on digital rectal examination. Scand J Urol Nephrol 31: 541-544, 1997 Aus G, Bergdahl S, Hugosson J, et al: Outcome of laterally directed sextant biopsies of the prostate in screened males aged 50-66 years. Implications for sampling order. Eur Urol 39: 655- 661, 2001 Babaian RJ, Toi A, Kamoi K, et al: A comparative analysis of sextant and an extended 11-core multisite directed biopsy strategy: J Urol 163: 152-157, 2000 Borboroglu PG, Amling CL: Correlation of positive sextant biopsy locations to site of positive surgical margins in radical prostatectomy specimens. Eur Urol 39: 648-653, 2001 Chon CH, Lai FC, McNeal JE, Presti JC Jr: Use of extended systematic sampling in patients with a prior negative prostate needle biopsy. J Urol 167: 2457-2460, 2002 Damiano R, Autorino R, Perdona S, et al: Are extended biopsies really necessary to improve prostate cancer detection? Prostate Cancer Prostatic Dis 6: 250-255, 2003 Djavan B, Kadeski K, Klopukh B, et al: Gleason scores from prostate biopsies obtained with 18-gauge biopsy needles poorly predict Gleason scores of radical prostatectomy specimens. Eur Radiol 33: 261-270, 1998 Djavan B, Waldert M, Zlotta AR, et al: Safety and morbidity of first and repeat transrectal ultrasound-guided prostate needle biopsies: results of the prospective European Prostate Cancer Detection Study. J Urol 166: 856-860, 2001 Eastham JA, May R, Robertson JL, et al: Development of a nomogram that predicts the probability of positive prostate biopsy in men with an abnormal digital rectal examination and a prostate-specific antigen between 0 and 4 ng/mL. Urology 54: 709-713, 1999 Gerstenbluth RE, Seftel AD, Hampel N, et al: The accuracy of the increased prostate specific antigen level, greater than or equal to 20 ng./ml., in predicting prostate cancer: is biopsy always required? J Urol 168: 1990-1993, 2002 Gore JL, Shariat SF, Miles BJ, et al: Optimal combinations of systematic sextant and laterally directed biopsies for the detection of prostate cancer. J Urol 165:1554-1559, 2001 Grossklaus DJ, Coffey CS, Shappell SB, et al: Percent of cancer in the biopsy set predicts pathological finding after prostatectomy. J Urol 167: 2032-2035, 2002 Grossklaus DJ, Coffey CS, Shappell SB, et al: Prediction of tumor volume and pathological stage in radical prostatectomy specimens is not improved by taking more prostate needlebiopsy cores. BJU Int 88: 722-726, 2001 Naughton CK, Ornstein DK, Smith DS, Catalona WJ: Pain and morbidity of transrectal ultrasound guided prostate biopsy: a prospective randomized trial of 6 versus 12 cores. J Urol 163: 168-171, 2000 Nava L, Montorsi F, Consonni P, et al: Results of a prospective randomised study comparing 6, 12, 18 transrectal ultrasound guided sextant biopsies in patients with elevated PSA, normal DRE and normal prostatic ultrasound. J Urol 157(Suppl): 59A, 1997 Peyromaure M, Ravery V, Messas A, et al: Pain and morbidity of an extensive prostate 10-biopsy protocol: a prospective study of 289 patients. J Urol 167: 218-221, 2002 Potter SR, Horniger W, Tinzl M, et al: Age, prostate-specific antigen, and digital rectal examination as determinants of the probability of having prostate cancer. Urology 57: 1100-1104, 2001 Ravery V, Goldblatt L, Royer B, et al: Extensive biopsy protocol improves the detection rates of prostate cancer. J Urol 164: 393-396, 2000 Ruijter E, van Leenders G, Miller G, et al: Errors in histologic grading by prostatic needle biopsy specimens: frequency and predisposing factors. J Pathol 192: 229-233, 2000 San Francisco IF, DeWolf WC, Rosen S, et al:Extended prostate needle biopsy improves concordance of Gleason grading between prostate needle biopsy and radical prostatectomy. J Urol 169: 136-140, 2003 Soloway MC, Obek C: Periprostatic local anesthesia before ultrasound guided prostate biopsy. J Urol 163: 172-173, 2000 Szabo J, Hegedus G, Bartok K, et al: Improving diagnostic accuracy of prostate carcinoma by systematic random mapbiopsy. Pathol Oncol Res 6: 111-113, 2000 Tigrani VS, Bhargava V, Shinohara K, Presti JC Jr: Number of positive systematic sextant biopsies predicts surgical margin status at radical prostatectomy. Urology 54: 689-693, 1999 Tombal B, Tajeddine N, Cosins JP, et al: Does site-specific labelling and individual processing of sextant biopsies improve the accuracy of prostate biopsy in predicting pathological stage in patients with T1c prostate cancer? BJU Int 89: 543-548, 2002 Yamamoto T, Ito K, Ohi M, et al:Diagnostic significance of digital rectal examination and transrectal ultrasonography in men with prostate-specific antigen levels of 4 ng/mL or less. Urology 58: 994-998, 2001 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2005 VL 11 IS 1 BP 40 EP 44 PG 5 ER PT J AU Hadar, T Shvero, J Yaniv, E Ram, E Shvili, I Koren, R AF Hadar, Tuvia Shvero, Jacob Yaniv, Eitan Ram, Eduard Shvili, Itzhak Koren, Rumelia TI Expression of p53, Ki-67 and Bcl-2 in Parathyroid Adenoma and Residual Normal Tissue SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE parathyroid adenoma; normal tissue; p53; Ki-67; bcl-2 ID parathyroid adenoma; normal tissue; p53; Ki-67; bcl-2 AB The aim of this study was to investigate the expression of Ki-67, bcl-2 and p53 in parathyroid adenomas and their residual rim of normal parathyroid tissue. Specimens from 26 parathyroid adenomas were studied by immunohistochemical analysis for Ki-67, bcl-2 and p53 expression. Positive findings were noted for p53 in 4 (15%) adenomas and none of the residual rims of normal parathyroid tissue (p = 0.055); for Ki-67 in 15 (56%) adenomas and none of the residual rims of normal parathyroid tissue (p = 0.00002); and for bcl-2 in 19 (73%) adenomas and 8 (31%) residual rims of normal parathyroid tissue (p < 0.01). The high rate of Ki-67 expression may indicate susceptibility of parathyroid adenomas to clonal proliferation. The weak immunoreactive expression of p53, combined with a relatively strong expression of bcl-2, may contribute to the characteristic slow progression of these tumors. C1 [Hadar, Tuvia] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Otolaryngology-Head and Neck SurgeryTel Aviv, Israel. [Shvero, Jacob] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Otolaryngology-Head and Neck SurgeryTel Aviv, Israel. [Yaniv, Eitan] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Otolaryngology-Head and Neck SurgeryTel Aviv, Israel. [Ram, Eduard] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Surgery ATel Aviv, Israel. [Shvili, Itzhak] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Otolaryngology-Head and Neck SurgeryTel Aviv, Israel. [Koren, Rumelia] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Pathology, Ramat Aviv, 69978 Tel Aviv, Israel. RP Koren, R (reprint author), Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Pathology, 69978 Tel Aviv, Israel. EM rumelia@isdumail.co.il CR Abbona GC, Papotti M, Gasparri G, et al: Proliferative activity in parathyroid tumors as detected by Ki-67 immunostaining. Hum Pathol 26:135-138, 1995 Arnold A, Staunton CE, Kim HG, et al: Monoclonality and abnormal parathyroid hormone genes in parathyroid adenomas. N Engl J Med 318: 658-662, 1988 Arnold A, Kim HG: Clonal loss of one chromosome 11 in a parathyroid adenoma. J Clin Endocrinol Metab 69:496-499, 1989 Carson DA, Ribeiro JM: Apoptosis and disease. Lancet 341:1251-1254, 1993 Cryns VL, Rubio MP, Thor AD, et al: p53 abnormalities in human parathyroid carcinoma. J Clin Endocrinol Metab 78:1320-1324, 1994 Dervan PA, Magee HM, Buckley C, et al: Proliferating cell nuclear antigen counts in formalin-fixed paraffin-embedded tissue correlate with Ki-67 in fresh tissue. Am J Clin Pathol 97:S21-28, 1992 Ghandur-Mnaymneh L, Kimura N: The parathyroid adenoma. A histopathologic definition with a study of 172 cases of primary hyperparathyroidism. Am J Pathol 115:70-83, 1984 Greenblatt MS, Bennett WP, Hollstein M, et al: Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res 54:855-4878, 1994 Gulkesen KH, Kilicarslan B, Altunbas HA, et al: EGFR and p53 expression and proliferative activity in parathyroid adenomas; an immunohistochemical study. APMIS 109:870-874, 2001 Hakim JP, Levine MA: Absence of p53 point mutations in parathyroid adenoma and carcinoma. J Clin Endocrinol Metab 78:103-106, 1994 Hockenbery DM, Zutter M, Hickey W, et al: BCL2 protein is topographically restricted in tissues characterized by apoptotic cell death. Proc Natl Acad Sci USA 88:6961-6965, 1991 Hollstein MC, Peri L, Mandard AM, et al: Genetic analysis of human esophageal tumors from two high incidence geographic areas: frequent p53 base substitutions and absence of ras mutations. Cancer Res 51:4102-4106, 1991 Karak AK, Sarkar C, Chumber S, Tandon N: MIB-1 proliferative index in parathyroid adenoma & hyperplasia. Ind J Med Res 105: 235-238, 1997 Kayath MJ, Martin LC, Vieira JG, et al: A comparative study of p53 immunoexpression in parathyroid hyperplasias secondary to uremia, primary hyperplasias, adenomas and carcinomas. Eur J Endocrinol 139:78-83, 1998 Kishikawa S, Shan L, Ogihara K, et al: Overexpression and genetic abnormality of p53 in parathyroid adenomas. Pathol Int 49:853-857, 1999 LeBrun DP, Warnke RA, Cleary ML: Expression of bcl-2 in fetal tissues suggests a role in morphogenesis. Am J Pathol 142:743-753, 1993 Lloyd RV, Carney A, Ferreio JA, et al: Immunohistochemical Analysis of the cell cycle-associated antigens ki-67 and retinoblastoma protein in parathyroid carcinomas and adenomas. Endocrine Pathol 6: 279-287, 1995 Loda M, Lipman J, Cukor B, et al: Nodular foci in parathyroid adenomas and hyperplasias: an immunohistochemical analysis of proliferative activity. Hum Pathol 25:1050-1056, 1994 Lu QL, Abel P, Foster CS, et al: bcl-2: role in epithelial differentiation and oncogenesis. Hum Pathol 27:102-110, 1996 Martin LN, Kayath MJ, Vieira JG, et al: Parathyroid glands in uraemic patients with refractory hyperparathyroidism: histopathology and p53 protein expression analysis. Histopathology 33:46-51, 1998 Naccarato AG, Marcocci C, Miccoli P, et al: Bcl-2, p53 and MIB-1 expression in normal and neoplastic parathyroid tissues. J Endocrinol Invest 21:136-141, 1998 Nigro JM, Baker SJ, Preisinger AC, et al: Mutations in the p53 gene occur in diverse human tumor types. Nature 342:705-708, 1989 Novack DV, Korsmeyer SJ: Bcl-2 protein expression during murine development. Am J Pathol 145:61-73, 1994 Ricci F, Mingazzini PL, Sebastiani V, et al: p53 as a marker of differentiation between hyperplastic and adenomatous parathyroids. Ann Diagn Pathol 6:229-235, 2002 Stojadinovic A, Hoos A, Nissan A, et al: Parathyroid neoplasms: clinical, histopathological, and tissue microarray-based molecular analysis. Hum Pathol 34:54-64, 2003 Szende B, Farid P, Vegso G, et al: Apoptosis and P53, Bcl-2 and Bax gene expression in parathyroid glands of patients with hyperparathyroidism. Pathol Oncol Res 10: 98-103, 2004 Szende B: The occurrence and significance of apoptosis in tumors. Magy Onkol 48: 215-219, 2004 Tsujimoto Y, Cossman J, Jaffe E, et al: Involvement of the bcl- 2 gene in human follicular lymphoma. Science 228:1440-1443, 1985 Vargas MP, Vargas HI, Kleiner DE, et al: The role of prognostic markers, MIB-1, RB, and bcl-2, in the diagnosis of parathyroid tumors. Mod Pathol 10:12-17, 1997 Wang W, Johansson H, Kvasnicka T, et al: Detection of apoptotic cells and expression of Ki-67 antigen, Bcl-2, p53 oncoproteins in human parathyroid adenoma. APMIS 104:789-796, 1996 Williams GT: Programmed cell death: apoptosis and oncogenesis. Cell 65:1097-1098, 1991 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2005 VL 11 IS 1 BP 45 EP 49 PG 5 ER PT J AU Koch, JH Hau, P AF Koch, J Horst Hau, Peter TI ROC Analysis as an Additional Method to Characterize Time to Event Data SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE ROC; Kaplan-Meier; brain tumor ID ROC; Kaplan-Meier; brain tumor AB The receiver operating characteristic (ROC) is predominantly used to assess the discriminant power of diagnostic tests. The present paper proposes this method as an additional alternative to compare survival data of in oncology or related fields. Survival data of brain tumors were analysed with conventional Kaplan-Meier method and ROC. The Area und the Curve (ROC-curve) gives additional illustrative information to distinguish between two therapeutic approaches and low or high grade brain tumors. C1 [Koch, J Horst] Neurologische Universitatsklinik Regensburg, Universitatsstrasse 84, 93053 Regensburg, Germany. [Hau, Peter] Neurologische Universitatsklinik Regensburg, Universitatsstrasse 84, 93053 Regensburg, Germany. RP Koch, JH (reprint author), Neurologische Universitatsklinik Regensburg, 93053 Regensburg, Germany. EM horst.koch@medbo.de CR Altman D. Practical Statistics for Medical Research. Chapman and Hall. NY 1991 Jensen K, Muller H-H, Schafer H. Regional confidence bands for ROC curves. Statistics in Medicine 19: 493-509, 2000 Kalbfleisch JD, Prentice RL. The Statistical Analysis of Failure Time Data. J. Wiley, NY 1980 Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc 53: 457-481, 1958 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2005 VL 11 IS 1 BP 50 EP 52 PG 3 ER PT J AU Bal, N Kayaselcuk, F Polat, A Bolat, F Yilmaz, Z Tuncer, I AF Bal, Nebil Kayaselcuk, Fazilet Polat, Ay?e Bolat, Filiz Yilmaz, Zerrin Tuncer, Ilhan TI Familial Cystic Nephroma in Two Siblings with Pleuropulmonary Blastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Cystic nephroma; familial tumors; pleuropulmonary blastoma ID Cystic nephroma; familial tumors; pleuropulmonary blastoma AB Cystic nephroma (CN) and pleuropulmonary blastoma (PPB) are rare tumors. In the cases presented here, a 13-month-old boy underwent right radical nephrectomy for CN. From the family history we learned that four years ago the patient’s older sister underwent left radical nephrectomy for CN at a different center when she was 4 years old. A lung tumor was detected in the sister one year after nephrectomy. Biopsy from the lung tumor revealed PPB, and the sister died within one year after biopsy. To the knowledge of the authors, these cases represent the second reported familial occurrence of CN and the fourth of CN and PPB. C1 [Bal, Nebil] Baskent University, Department of Pathology, Adana Hastanesi, Yure ir, 01250 Adana, Turkey. [Kayaselcuk, Fazilet] Baskent University, Department of Pathology, Adana Hastanesi, Yure ir, 01250 Adana, Turkey. [Polat, Ay?e] Mersin University School of Medicine, Department of PathologyMersin, Turkey. [Bolat, Filiz] Baskent University, Department of Pathology, Adana Hastanesi, Yure ir, 01250 Adana, Turkey. [Yilmaz, Zerrin] Baskent University Faculty of Medicine, Department of Medical Biology and GeneticsAnkara, Turkey. [Tuncer, Ilhan] Baskent University, Department of Pathology, Adana Hastanesi, Yure ir, 01250 Adana, Turkey. RP Bal, N (reprint author), Baskent University, Department of Pathology, 01250 Adana, Turkey. EM nebilbal@baskent-adn.edu.tr CR Delahunt B, Thomson KJ, Ferguson AF, et al: Familial cystic nephroma and pleuropulmonary blastoma. Cancer 71: 1338- 1342, 1993 Ishida Y, Kato K, Kigasawa H, et al: Synchronous occurrence of pleuropulmonary blastoma and cystic nephroma: possible genetic link in cystic lesions of the lung and the kidney. Med Pediatr Oncol 35: 85-87, 2000 Kiziltepe TT, Patrick E, Alvarado C, et al: Pleuropulmonary blastoma and ovarian teratoma. Pediatr Radiol 29: 901-903, 1999 Priest JR, Watterson J, Strong L, et al: Pleuropulmonary blastoma: a marker for familial disease. J Pediatr 128: 220-224, 1996 Manivel JC, Priest JR, Watterson J, et al: Pleuropulmonary blastoma. The so-called pulmonary blastoma of childhood. Cancer 62: 1516-1526, 1988 Lallier M, Bouchard S, Di Lorenzo M, et al: Pleuropulmonary blastoma: a rare pathology with an even rarer presentation. J Pediatr Surg 34: 1057-1059, 1999 Knudson AG. On a new genetic syndrome. Med Pediatr Oncol 35: 428, 2000 Sebire NJ, Rampling D, Malone M, et al: Gains of chromosome 8 in pleuropulmonary blastomas of childhood. Pediatr Dev Pathol 5: 221-222, 2002 Sacher P, Willi UV, Niggli F, et al: Cystic nephroma: a rare benign renal tumor. Ped Surg Int 13: 197-199, 1998 Kural AR, Obek C, Ozbay G, et al: Multilocular cystic nephroma: an unusual localization. Urology 52: 897-899, 1998 Fujimoto K, Samma S, Fukui Y, et al: Spontaneously ruptured multilocular cystic nephroma. Int J Urol 9: 183-186, 2002 Sharma S, Nagar R, Singh K, et al: Cystic nephroma: an unusual renal lesion. J Urol 163: 1860, 2000 Polat P, Suma S, Celenk C, et al: Quiz case of the month. Multilocular cystic nephroma, MLCN). Eur Radiol 7: 757-758, 1997 Verma RS, Babu A: Human Chromosomes Principles and Techniques 2nd Edition. Mc Graw Hill, New York, 1995, pp. 6-71 Verma RS, Babu A: Human Chromosomes Principles and Techniques 2nd Edition. Mc Graw Hill, New York, 1995, pp. 72-133 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2005 VL 11 IS 1 BP 53 EP 56 PG 4 ER PT J AU Rieker, JR Aulmann, S Schnabel, AP Sack, FU Herwart, FO Mechtersheimer, G Schirmacher, P Blaker, H AF Rieker, J Ralf Aulmann, Sebastian Schnabel, A Philipp Sack, Falk-Udo Herwart, F Otto Mechtersheimer, Gunhild Schirmacher, Peter Blaker, Hendrik TI Cystic Thymoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE thymic cysts; cystic thymoma; micronodular thymoma ID thymic cysts; cystic thymoma; micronodular thymoma AB Thymic cysts are rare lesions located along the anatomical course of the third pharyngeal pouch. While most of the cases represent congenital cysts, they may also be related to neoplasms. We report a case of a micronodular thymoma with lymphoid stroma, which was completely built of small cysts, discuss the pathologic features of this tumor type and review the etiology and other aspects of thymic cysts. C1 [Rieker, J Ralf] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Aulmann, Sebastian] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Schnabel, A Philipp] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Sack, Falk-Udo] University Hospital, Department of Cardiac SurgeryHeidelberg, Germany. [Herwart, F Otto] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Mechtersheimer, Gunhild] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Schirmacher, Peter] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Blaker, Hendrik] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. RP Rieker, JR (reprint author), University of Heidelberg, Department of Pathology, 69120 Heidelberg, Germany. EM ralf.rieker@med.uni-heidelberg.de CR Bradford ML, Mahon HW, Grow JB: Mediastinal cysts and tumors. Surg Gynecol Obstet 85: 467-491, 1947 Dubois P: Du diagnostic de la syphilis consideree comme une des causes possibles de la mort du foetus. Gaz Med de Paris 21:392-395, 1850 Guba AM jr, Adam AE, Jaques DA, et al: Cervical presentation of thymic cysts. Am J Surg 136:430-436, 1978 Hyde TL, Sellers ED, Owen M: Thymic cyst of the neck: report of a case. Tex State J Med 39:539-540, 1944 Indeglia RA, Shea MA, Grage TB: Congenital cysts of the thymus gland. Arch Surg 94:149-152, 1967 Inoue M, Starostik P, Zettl A, et al: Correlating genetic aberrations with World Health Organization-defined histology and stage across the spectrum of thymomas. Cancer Res 63:3708- 3715, 2003. Krech WG, Storey CF, Umiker WC: Thymic cysts: a review of the literature and report of two cases. J Thorac Surg 27: 477- 493, 1954 Leong AS, Brown JH: Malignant transformation in a thymic cyst. Am J Surg Pathol 8:471-475, 1984 Marx A, Stroebel P. Marino M, et al: Micronodular thymoma with lymphoid stroma. In: Pathology and Genetics of Tumours of the Lung, Pleura, Thymus and Heart., Eds: WD Travis, E Brambilla HK Muller-Hermelink, CC Harris, World Health Organization of Tumors. IARC Press, Lyon 2004, pp. 167-168 Moran CA, Suster S: Thymoma with prominent cystic and hemorrhagic changes and areas of necrosis and infarction: a clinicopathologic study of 25 cases. Am J Surg Pathol 25:1086- 1090, 2001 Murray JA, Parker AC: Mediastinal Hodgkin’s disease and thymic cysts. Acta Haematol, Basel, 71:282-284, 1984 Penzel R, Hoegel J, Schmitz W, et al: Clusters of chromosomal imbalances in thymic epithelial tumours are associated with the WHO classification and the staging system according to Masaoka. Int J Cancer 105:494-498, 2003 Pollosson A, Piery M: Un cas d’epithelioma primitif du Thymus. Provinc Med 16:1-4, 1901 Rippert H: Die Entwicklungsstorung der Thymusdruse bei kongenitaler Lues. Frankfurt Z Pathol 11:209-218, 1912 Rosai J, Levine GD: Tumors of the thymus. In: Atlas of Tumor Pathology, 2nd series, fasc 13. Armed Forces Institute of Pathology, Washington DC, 1976, pp 48-49, 207-211, Smart J: A case of large thymic cyst successfully removed from the anterior mediastinum. Br J Tuberc 41: 84-88, 1947 Suster S, Moran CA: Primary thymic epithelial neoplasms showing combined features of thymoma and thymic carcinoma. A clinicopathologic study of 22 cases. Am J Surg Pathol 20:1469-1480, 1996 Suster S, Moran CA: Micronodular thymoma with lymphoid Bcell hyperplasia: clinicopathologic and immunohistochemical study of eighteen cases of a distinctive morphologic variant of thymic epithelial neoplasm. Am J Surg Pathol 23:955-962, 1999 Suster S, Rosai J: Multilocular thymic cyst: an acquired reactive process. Study of 18 cases. Am J Surg Pathol 15: 388-398, 1991 Suster S, Rosai J: Cystic thymomas. A clinicopathologic study of ten cases. Cancer 69:92-97, 1992 Tateyama H, Saito Y, Fujii Y, et al: The spectrum of micronodular thymic epithelial tumours with lymphoid B-cell hyperplasia. Histopathology 38: 519-527, 2001 Thomas De Montpreville V, Zemoura L, Dulmet E: Thymoma with epithelial micronodules and lymphoid hyperplasia: six cases of a rare and equivocal subtype. Ann Pathol 22:177-182, 2002 Wychulis AR, Payne WS, Clagett OT, Woolner LB: Surgical treatment of mediastinal tumors - a 40 year experience. J Thorac Cardiovasc Surg 62: 379-392, 1971 Zettl A, Strobel P, Wagner K, et al: Recurrent genetic aberrations in thymoma and carcinoma. Am J Pathol 157:257-266, 2000 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2005 VL 11 IS 1 BP 57 EP 60 PG 4 ER PT J AU Kopper, L Zalatnai, A Timar, J AF Kopper, Laszlo Zalatnai, Attila Timar, Jozsef TI Genomics of Pancreatic Cancer: Does It Make Any Improvement in Diagnosis, Prognosis and Therapy? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE pancreatic cancer; genomics ID pancreatic cancer; genomics AB Pancreatic cancer (PanC) is an extremely lifethreatening neoplasm due to its late discovery, rapid progression and resistance to chemo- and radiotherapy. In the past years a significant research attention turned to this cancer. Extensive genomic analysis of PanC revealed numerous alterations, however, none of them emerged yet as a key regulator of tumor progression. Our increasing knowledge on the molecular targets in various cancer types started to change their management. Examples of success of the molecular therapies (in CML, GIST, NSCLC) may initiate more activity in pancreatic cancer as well. C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor ProgressionBudapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM kopper@korb1.sote.hu CR Schleger C, Arens N, Zentgraf H, et al: Identification of frequent chromosomal aberrations in ductal adenocarcinoma of the pancreas by comparative genomic hybridization, CGH). J Pathol 191: 27-32, 2000 Mahlamaki EH, Hoglund M, Gorunova L, et al: Comparative genomic hybridization reveals frequent gains of 20q, 8q, 11q, 12p, and 17q, and losses of 18q, 9p, and 15q in pancreatic cancer. Genes Chromosomes Cancer 204: 383-391, 1997 Holtzmann K, Kohlhammer H, Schwaenen C, et al: Genomic DNA-chip hybridization reveals a higher incidence of genomic amplifications in pancreatic cancer than conventional comparative genomic hybridization and leads to the identification of novel candidate genes. Cancer Res 64: 4428-4444, 2004 Okami J, Simeone DM, Logsdon CD: Silencing of the hypoxiainducible cell death protein BNIP3 in pancreatic cancer. Cancer Res 64: 5338-5346, 2004 Prasad NB, Biankin AV, Fukushima N, et al: Gene expression profiles in pancreatic intraepithelial neoplasia reflect the effects of Hedgehog signaling on pancreatic ductal epithelial cells. Cancer Res 65: 1619-1626, 2005 Bardeesy N, DePinho RA: Pancreatic cancer biology and genetics. Nat Rev Cancer 2: 897-909, 2002 Juhasz M, Nitsche B, Malfertheiner P, Ebert MPA: Implications of the growth factor alterations in the treatment of pancreatic cancer. Mol Cancer 2: 5, 2003 Iacobuzio-Donahue CA, Asfaq R, Maitra A, et al: Highly expressed genes in pancreatic ductal adenocarcinomas: a comprehensive characterization and comparison of the transcription profiles obtained from three major technologies. Cancer Res 63: 8614-8622, 2003 Shen J, Person MD, Zhu J, et al: Protein expression profiles in pancreatic adenocarcinoma compared with normal pancreatic tissue affected by pancreatitis as detected by two-dimensional gel electrophoresis and mass spectrometry. Cancer Res 64: 9018-9026, 2004. Suwa H, Ohshio G, Arao S, et al: Immunohistochemical localization of P-glycoprotein and expression of the multidrug resistance- 1 gene in human pancreatic cancer: relevance to indicator of better prognosis. Jpn J Cancer Res 87: 641-649, 1996 Watanabe I, Hasabe T, Sasaki S et al: Advanced pancreatic ductal cancer: fibrotic focus and b-catenin expression correlate with outcome. Pancreas 26: 326-333, 2003 Ougolkov AV, Fernandez-Zapico ME, Savoy DN, et al: Glycogen synthase kinase-3b participates in nuclear factor kB-mediated gene transcription and cell survival in pancreatic cancer cells. Cancer Res 65: 2076-2081, 2005 Muerkoster S, Arlt A, Sipos B, et al: Increased expression of the E3-ubiquitin ligase receptor subunit bTRCP1 relates to constitutive nuclear factor-kB activation and chemoresistance in pancreatic carcinoma cells. Cancer Res 65: 1316-1324, 2005 Bai J, Sui J, Demirjian A, et al: Predominant Bcl-XL knockdown disables antiapoptotic mechanisms: tumor necrosis factor- related apoptosis-inducing ligand-based triple chemotherapy overcomes chemoresistance in pancreatic cancer cells in vitro. Cancer Res 65: 2344-2352, 2005 Jiang XT, Tao HQ, Zou SC: Detection of serum tumor markers in the diagnosis and treatment of patients with pancreatic cancer. Hepatobiliary Pancreatic Dis Inst 3: 363-468, 2004 Louhimo J, Alfthan H, Stenman UH, Haglund C: Serum HCG beta and CA 72-4 are stronger prognostic factors than CEA, CA 19-9 and CA 242 in pancreatic cancer. Oncology 66: 126- 131, 2004 Suzuki K, Aiura K, Kitagou M, et al: Platelets counts closely correlate with the disease-free survival interval of pancreatic cancer patients. Hepatogastroenterology 51: 847-853, 2004 Timar J, Tovari J, Raso E, et al: Thrombocyte-mimicry of cancer cells: rationale for the prevention and treatment of hematogenous dissemination. Oncology, 2005, in press) Logsdon CD, Simeone DM, Binkley C, et al: Molecular profiling of pancreatic adenocarcinoma and chronic pancreatitis identified multiple genes differentially regulated in pancreatic cancer. Cancer Res 63: 2649-2657, 2003 Li YJ, Meng YX, Ji XR. Relationship between expression of Ecadherin and alpha-catenin and biological behaviors of human pancreatic cancer. Hepatobiliary Pancreat Dis Int 2: 471-477, 2003 Keleg S, Buchler P, Ludwig R, et al: Invasion and metastasis in pancreatic cancer. Mol Cancer 2:14, 2003 Stanton KJ, Sidner RA, Miller GA, et al: Analysis of Ki-67 antigen expression, DNA proliferative fraction, and survival in resected cancer of the pancreas. Am J Surg 186: 486-492, 2003 Fukumoto A, Ikeda N, Sho M, et al: Prognostic significance of localized p27Kip1 and potential role of Jab1/CSN5 in pancreatic cancer. Oncol Rep 11: 277-284, 2004 Juuti A, Nordling S, Louhimo J, et al: Loss of p27 expression is associated with poor prognosis in stage I-II pancreatic cancer. Oncology 65: 371-377, 2003 Jinfeng M, Kimura W, Hirai I, et al: Expression off MUC5AC and MUC6 in invasive ductal carcinoma of the pancreas and relationship with prognosis. Int J Gastrointest Cancer 34: 9-18, 2003 Ueda S, Ogata S, Tsuda H, et al: The correlation between cytoplasmic overexpression of epidermal growth factor receptor and tumor aggressiveness: poor prognosis in patients with pancreatic ductal adenocarcinoma. Pancreas 29: e1-8, 2004 Schlieman MG, Fahy BN, Ramsamooj R, et al: Incidence, mechanism and prognostic value of activated AKT in pancreas cancer. Br J Cancer 89: 2110-2115, 2003 Grutzmann R, Luttges J, Sipos B, et al: ADAM9 expression in pancreatic cancer is associated with tumour type and is a prognostic factor in ductal adenocarcinoma. Br J Cancer 90: 1053- 1058, 2004 Fukunaga A, Miyamoto M, Cho Y, et al: CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas 28: e26-31, 2004 Jaster R: Molecular regulation of pancreatic stellate cell function. Mol Cancer 3: 26, 2004 Sausville EA: The challenge of pathway and environmentmediated drug resistance. Cancer Metastasis Rev 20: 117-122, 2001 Elad-Sfadia G, Haklai R, Ballan E, et al: Galectin-1 augments Ras activation and diverts Ras signals to Raf-1 at the expense of phosphoinositide 3-kinase. J Biol Chem 277: 37169-37175, 2002 Kitada T, Seki S, Sakaguchi H, et al: Clinicopathological significance of hypoxia-inducible factor-1a expression in human pancreatic carcinoma. Histopathology 43: 550-555, 2003 Duffy JP, Eibl G, Reber HA, Hines OJ: Influence of hypoxia and neoangiogenesis on the growth of pancreatic cancer. Mol Cancer 2: 12, 2003 Silletti S, Paku S, Raz A: Tumor cell motility and metastasis. Autocrine motility factor as an example of ecto/exoenzyme cytokines. Pathol Oncol Res 3: 230-254, 1997 Semenza GL: Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3: 721-732, 2003 Kuwahara K, Sasaki T, Kuwada Y, et al: Expression of angiogenic factors in pancreatic ductal carcinoma: a correlative study with clinicopathologic parameters and patient survival. Pancreas 26: 344-349, 2003 Ikeda N, Nakajima Y, Tokuhara T, et al: Clinical significance of aminopeptide N/CD13 expression in human pancreatic carcinoma. Clin Cancer Res 9: 1503-1508, 2003 Uehara H, Miyamoto M, Kato K, et al: Expression of pigment epithelium-derived factor decreases liver metastasis and correlates with favorable prognosis for patients with ductal pancreatic adenocarcinoma. Cancer Res 64: 3533-3537, 2004 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 69 EP 73 PG 5 ER PT J AU Croce, VM Rabassa, EM Pereyra, A Segal-Eiras, A AF Croce, V Maria Rabassa, E Martin Pereyra, Adrian Segal-Eiras, Amada TI Influence of Sialic Acid Removal on MUC1 Antigenic Reactivity in Head and Neck Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE head and neck carcinoma; MUC1; carbohydrate antigens; sialic acid ID head and neck carcinoma; MUC1; carbohydrate antigens; sialic acid AB To investigate the influence of sialic acid removal on MUC1 peptidic and carbohydrate epitope reactivity in head and neck squamous cell carcinoma (HNSCC), tumor samples belonging to 24 HNSCC patients were studied by standard immunohistochemistry (IHC) with and without desialylation with 0.1 U/ml neuraminidase. From each tumor sample, subcellular fractions were obtained and analyzed by SDS-PAGE and Western blotting (WB). Three monoclonal antibodies (MAbs) were used: C595 MAb directed to MUC1 protein core, an anti-Tn hapten MAb, and an anti-sTn hapten MAb; a comparative analysis between desialylated and sialylated samples was performed. By IHC without neuraminidase treatment, 19 of 24 samples reacted with anti-MUC1 peptidic epitope, while Tn hapten was not detected and sTn was found in 1 of 24 cases. Desialylation increased either the number of reacting cells or the intensity of the reaction with C595 and anti-Tn MAbs, and some negative samples became positive. On the other hand, sTn expression decreased with desialylation. By WB, several bands from >200 to 25 kDa were found; desialylation increased high-molecular-weight bands, diminishing the detection of low-molecular-weight ones. The use of desialylation is a suitable treatment that contributes to the exposure of MUC1-associated epitopes, which may be related to the spreading of HNSCC. C1 [Croce, V Maria] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Rabassa, E Martin] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Pereyra, Adrian] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Segal-Eiras, Amada] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. RP Segal-Eiras, A (reprint author), Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), La Plata, Argentina. EM as-eiras@netverk.com.ar CR Bramwell ME, Wiseman G, Shotton DM: Electron-microscopic studies of the CA antigen, epitectin. J Cell Sci 86:249-261, 1986. Brockhausen I, Yang J-M, Burchell J et al: Mechanisms underlying aberrant glycosylation of MUC1 mucin in breast cancer cells. Eur J Biochem 223:607-617, 1995. Bryne M, Gravdahi C, Koppang HS et al: Is the carbohydrate sialosyl-Tn a marker for altered, non-malignant activity in squamous epithelium in the head and neck region? J Pathol 175:237-242, 1995 Burchell J and Taylor-Papadimitriou J: Effect of modification of carbohydrate side chains on the reactivity of antibodies with core-protein epitopes of the MUC1 gene product. Epithelial Cell Biol 2:115-162, 1993. Ciborowski P, Konitzi WM, Magarian Blander J, et al: Screening of anti-MUC1 antibodies for reactivity with native, ascites, and recombinant, Baculovirus, MUC1 and for blocking MUC1 specific cytotoxic T-lymphocytes. Tumor Biol 19, Suppl 1):147-151, 1998. Croce MV, Colussi A, Price MR, et al: Expression of tumor associated antigens in normal, benign and malignant human mammary epithelial tissue: a comparative study. Anticancer Res 17:4287-4292, 1997. Croce MV, Rabassa ME, Price MR, et al: MUC1 mucin and carbohydrate associated antigens as tumor markers in head and neck squamous cell carcinoma. Pathol Oncol Res 7:284-291, 2001. Croce MV, Isla-Larrain M, Rua C, et al: Patterns of MUC1 cytoplasmic tail monoclonal antibody in breast cancer. J Histochem Cytochem 5:1-8, 2003. Dabelsteen E, Clausen H, Mandel U: Aberrant glycosylation in oral malignant and premalignant lesions. J Oral Path Med 20:361-368, 1991. Dalziel M, Whitehouse C, Mcfarlane I, et al: The relative of the C2Gn T1 and ST3Gal-I glycosyltransferases determine O-glycan structure and expression of a tumor-associated epitope on MUC1. J Biol Chem 276:11007-11015, 2001. Farmer RW, Richtsmeier WJ, Scher RL: Identification of sialyl Lewis-x in squamous cell carcinoma of the head and neck. Head Neck 20:726-731, 1998. Feickert HJ, Anger BR, Cordon-Cardo C, et al: Cell-surface antigens of human tumors detected by mouse monoclonal antibodies: definition of blood-group- and non-blood-group-related antigenic systems. Int J Cancer 46:1007-1013, 1990. Gendler SJ, Taylor-Papadimitriou J, Duhig T: A highly immunogenic region of human polymorphic mucin expressed by carcinomas is made up of tandem repeats. J Biol Chem 263:12820-12823, 1988. Gendler S: MUC1, the renaissance molecule. J Mamm Gland Biol Neoplasia 6:339-353, 2001. Gleich LL, Ryzenman J, Gluckman JL et al: Recurrent advanced, T3 or T4, head and neck squamous cell carcinoma: is salvage possible? Arch Otolaryngol Head Neck Surg 130:35-38, 2004. Hakomori S-I: Aberrant glycosylation in tumors and tumor-associated carbohydrate antigens. Adv Cancer Res 52:257-331, 1989. Ho JJL, Cheng S, Kim YS: Access to peptide regions of a surface mucin, MUC1, is reduced by sialic acids. Biochem Biophys Res Commun 210:866-873, 1995. Irimura T, Denda K, Iida S, et al: Diverse glycosylation of MUC1 and MUC2: potential significance in tumor immunity J Biochem 126:975-985, 1999. Itoh T, Yonezawa S, Nomoto M, et al: Expression of mucin antigens and Lewis X-related antigens in carcinomas and dysplasia of the pharynx and larynx. Pathol Int 46:646-655, 1996. Itzkowitz SH, Bloom EJ, Kokal WA, et al: Sialosyl Tn: a novel mucin antigen associated with prognosis in colorectal cancer patients. Cancer 66:1960-1966, 1990. Jeannon J-P, Stafford FW, Soames JV, et al: Altered MUC1 and MUC2 glycoprotein expression in laryngeal carcinoma. Otolaryngol Head Neck 124: 199-202, 2000. Kam JL, Regimbald LH, Hilgers JH, et al: MUC1 synthetic peptide inhibition of intercellular adhesion molecule-1 and MUC1 binding requires six tandem repeats. Cancer Res 58: 5577-5581, 1998. Kjeldsen T, Clausen H, Hirohashi S, et al: Preparation and characterization of monoclonal antibodies directed to the tumorassociated O-linked sialosyl-2-6 alpha-N-acetylgalactosaminyl, sialosyl-Tn, epitope. Cancer Res 48:2214-20, 1988. Kunz H: Synthetic glycopeptides for the development of tumorselective vaccines. J Pept Sci 9: 563-573, 2003. Kurahara S, Shinohara M, Ikebe T, et al: Immunohistochemical study of sialyl Le(a, and sialyl Le(x, antigen in oral squamous cell carcinoma: the association of sialyl Le(a, expression with the metastatic potential. Head Neck 21:330-337, 1999. Laemmli UK: Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227:680-685, 1970. Lloyd KL, Burchell J, Kudryashov V, et al: Comparison of Olinked carbohydrate chains in MUC-1 mucin from normal breast epithelial cell lines and breast carcinoma cell lines. Demonstration of simpler and fewer glycan chains in tumor cells. J Biol Chem 271:33325-33334, 1996. Luna-More S, Rius F, Weil B, et al: EMA: a differentiation antigen related to node metastatic capacity of breast carcinomas. Pathol Res Pract 197:419-425, 2001. Mandel U, Petersen OW, Sorensen H, et al: Simple mucin-type carbohydrates in oral stratified squamous and salivary gland epithelia. J Invest Dermatol 97:713-721, 1991. Muller S and Hanisch F-G: Recombinant MUC1 probe authentically reflects cell-specific O-glycosylation profiles of endogenous breast cancer mucin. J Biol Chem 277:26103-26112, 2002. Musselli C, Livingston PO, Ragupathi G: Keyhole limpet hemocyanin conjugate vaccines against cancer: the Memorial Sloan Kettering experience. J Cancer Res Clin Oncol 127, Suppl 2):R20-26, 2001. Ogawa H, Ghazizadeh M, Araki T: Tn and sialyl-Tn antigens as potential prognostic markers in human ovarian carcinoma. Gynecol Obstet Invest 41:278-283, 1996. Price MR, Pugh JA, Hudecz F, et al: C595 - a monoclonal antibody against the protein core of human urinary epithelial mucin commonly expressed in breast carcinomas. Br J Cancer 61: 681-686, 1990. Regimbald LH, Pilarski LM, Longenecker BM: The breast mucin MUC1 as a novel adhesion ligand for endothelial intercellular adhesion molecule 1 in breast cancer. Cancer Res 56:4244-4249, 1996. Renkonen J, Paavonen T, Renkonen R: Endothelial and epithelial expression of sialyl Lewis x and sialyl Lewis a in lesions of breast carcinoma. Int J Cancer 74:296-300, 1997. Rubin JS, Bloor BK, Hart IR, et al: muc-1 gene expression in head and neck squamous cell carcinoma. J Laryngol Otol 114:772-776, 2000. Shi SR, Cote RJ, Taylor CR: Antigen retrieval techniques: current perspectives. Histochem Cytochem 49:931-937, 2001. Spencer DI, Price MR, Tendler SJ, et al: Effect of glycosylation of a synthetic MUC1 mucin-core-related peptide on recognition by anti-mucin antibodies. Cancer Lett 100:11-15, 1996. Stenersen TC, Dabelsteen E: Changes in the glycosylation pattern of histo-blood group antigens in benign, premalignant and malignant laryngeal epithelial lesions. APMIS 27(Suppl):139- 148, 1992. Taylor-Papadimitriou J, Burchell J, Miles DW, et al: MUC1 and cancer. Biochim Biophys Acta 1455:301-313, 1999. Towbin H, Staehelin T, Gordon J: Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc Natl Acad Sci USA 76:4350-4354, 1979. Wesseling J, van der Van SW, Vos HL, et al: Episialin, MUC1, overexpression inhibits integrin-mediated cell adhesion to extracellular matrix components. J Cell Biol 129:255-265, 1995. Zhang S, Walberg LA, Ogata S, et al: Immune sera and monoclonal antibodies define two configurations for the sialyl Tn tumor antigen. Cancer Res 55:3364-3368, 1995. Zhang K, Baeckstrom D, Brevinge H, Hanasson GC: Secreted MUC1 mucins lacking their cytoplasmic part and carrying sialyl- Lewis a and x epitopes from a tumor cell line and sera of colon carcinoma patients can inhibit HL-60 leukocyte adhesion to E-selectin-expressing endothelial cells. J Cell Biochem 60:538-549, 1996. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 74 EP 81 PG 8 ER PT J AU Komlosi, K Kellermayer, R Maasz, A Havasi, V Hollody, K Vincze, O Merkli, H Pal, E Melegh, B AF Komlosi, Katalin Kellermayer, Richard Maasz, Anita Havasi, Viktoria Hollody, Katalin Vincze, Olga Merkli, Hajnalka Pal, Endre Melegh, Bela TI Maternally Inherited Deafness and Unusual Phenotypic Manifestations Associated with A3243G Mitochondrial DNA Mutation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mtDNA A3243G; tRNALeu(UUR); mitochondrial disease; MELAS; deafness ID mtDNA A3243G; tRNALeu(UUR); mitochondrial disease; MELAS; deafness AB The mitochondrial DNA A3243G transition is a fairly common mutation which often associates with a MELAS (mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes) phenotype, however, a broad variety in the associated clinical picture has also been described. The patient reported here developed a generalized seizure at age 12, which was followed by bilateral hearing loss and occasional fatigue. The maternal inheritance pattern of hearing loss pointed to a possible mitochondrial origin, which was confirmed by molecular analysis of the mitochondrial DNA, revealing a heteroplasmic A3243G transition. Interestingly, muscle biopsy showed ragged-red fibers in the proband, which is unusual in the deafness-associated forms of this mitochondrial disorder. In addition to hearing impairment in four generations of the family, fatal cerebral embolization in the mother and fatal heart attack in the maternal grandmother (both at age 33) also occurred. On the contrary, diabetes, which usually accompanies the hearing loss variant, was specifically absent in all generations. The unusual manifestations associated with this mutation somewhat differentiate this family from the already known variants. C1 [Komlosi, Katalin] University of Pecs, Department of Medical Genetics and Child Development, H-7624 Pecs, Hungary. [Kellermayer, Richard] University of Pecs, Department of Medical Genetics and Child Development, H-7624 Pecs, Hungary. [Maasz, Anita] University of Pecs, Department of Medical Genetics and Child Development, H-7624 Pecs, Hungary. [Havasi, Viktoria] University of Pecs, Department of Medical Genetics and Child Development, H-7624 Pecs, Hungary. [Hollody, Katalin] University of Pecs, Department of NeurologyPecs, Hungary. [Vincze, Olga] County Hospital of BaranyaPecs, Hungary. [Merkli, Hajnalka] University of Pecs, Department of NeurologyPecs, Hungary. [Pal, Endre] University of Pecs, Department of NeurologyPecs, Hungary. [Melegh, Bela] University of Pecs, Department of Medical Genetics and Child Development, H-7624 Pecs, Hungary. RP Melegh, B (reprint author), University of Pecs, Department of Medical Genetics and Child Development, H-7624 Pecs, Hungary. EM Bela.Melegh@aok.pte.hu CR Bene J, Nadasi E, Kosztolanyi G, et al: Congenital cataract as the first symptom of a neuromuscular disease caused by a novel single large-scale mtDNA deletion. Eur J Hum Genet 11: 375- 379, 2003 Chinnery PF, Turnbull DM: Epidemiology and treatment of mitochondrial disorders. Am J Med Genet 106:94-101, 2001 Damian MS, Seibel P, Reichmann H, et al: Clinical spectrum of the MELAS mutation in a large pedigree. Acta Neurol Scand 92: 409-415, 1995 DiMauro S, Hirano M, Schon EA: Mitochondrial encephalomyopathies: therapeutic approaches. Neurol Sci 21: S901-S908, 2000 DiMauro S, Schon EA: Mitochondrial DNA mutations in human disease. Am J Med Genet 106: 18-26, 2001 DiMauro S, Hirano M, Kaufmann P, et al: Clinical features and genetics of myoclonic epilepsy with ragged red fibers. Adv Neurol 89: 217-229, 2002 DiMauro S, Schon EA: Mitochondrial respiratory-chain diseases. N Engl J Med 348: 2656-2668, 2003 Fischel-Ghodsian N: Mitochondrial deafness mutations reviewed. Hum Mut 13: 261-270, 1999 Gerbitz KD, van den Ouweland JM, Maassen JA, et al: Mitochondrial diabetes mellitus: a review. Biochim Biophys Acta 1271: 253-260, 1995 Goto Y, Nonaka I, Horai S: A mutation in the tRNA(Leu)(UUR, gene associated with the MELAS subgroup of mitochondrial encephalomyopathies. Nature 348: 651-653, 1990 Hirano M, Ricci E, Koenigsberger MR, et al: Melas: an original case and clinical criteria for diagnosis. Neuromuscul Disord 2:125-135, 1992 Huang CC, Kuo HC, Chu CC, et al: Clinical phenotype, prognosis and mitochondrial DNA mutation load in mitochondrial encephalomyopathies. J Biomed Sci 9: 527-533, 2002 Ihara Y, Namba R, Kuroda S, et al: Mitochondrial encephalomyopathy, MELAS): Pathological study and successful therapy with coenzyme Q10 and idebenone. J Neurol Sci 90: 263- 271, 1989 Jacobs HT: Mitochondrial deafness. Ann Med 29: 438-491, 1997 Kobayashi Y, Momoi MY, Tominaga K, et al: A point mutation in the mitochondrial tRNA(Leu)(UUR, gene in MELAS, mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes). Biochem Biophys Res Commun 173: 816-822, 1990 Komlosi K, Bene J, Havasi V, et al: The A3243G mitochondrial DNA mutation in a Hungarian family, In Hungarian). Orv Heti 35: 1805-1809, 2004 Lam CW, Lau CH, Williams JC, et al.: Mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes, MELAS, triggered by valproate therapy. Eur J Pediatr 156: 562-564, 1997 Macmillan C, Lach B, Shoubridge EA: Variable distribution of mutant mitochondrial DNAs, tRNA(Leu[3243]), in tissues of symptomatic relatives with MELAS: the role of mitotic segregation. Neurology 43: 1586-1590, 1993 Matthews PM, Hopkin J, Brown RM, et al: Comparison of the relative levels of the 3243, A—>G, mtDNA mutation in heteroplasmic adult and fetal tissues. J Med Genet 31: 41-44, 1994 McFarland R, Taylor RW, Turnbull DM: The neurology of mitochondrial DNA disease. Lancet Neurol 1: 343-351, 2002 Montagna P, Gallassi R, Medori R, et al: MELAS syndrome: characteristic migrainous and epileptic features and maternal transmission. Neurology 38: 751-754, 1998 Munnich A, Rustin P: Clinical spectrum and diagnosis of mitochondrial disorders. Am J Med Genet 106: 4-17, 2001 Oldfors A, Tulinius M: Mitochondrial encephalomyopathies. J Neuropathol Exp Neurol 62: 217-227, 2003 Ozawa M, Nonaka I, Goto Y: Single muscle fiber analysis in patients with 3243 mutation in mitochondrial DNA: comparison with the phenotype and the proportion of mutant genome. J Neurol Sci 159: 170-175, 1998 Pavlakis SG, Phillips PC, DiMauro S, et al: Mitochondrial myopathy, encephalopathy, lactic acidosis, and strokelike episodes: a distinctive clinical syndrome. Ann Neurol 16: 481- 488, 1984 Prezant TR, Agapian JV, Bohlman MC, et al: Mitochondrial ribosomal RNA mutation associated with both antibioticinduced and non-syndromic deafness. Nat Genet 4: 289-294, 1993 Pronicki M, Sykut-Cegielska J, Mierzewska H, et al: Diversity of clinical symptoms in A3243G mitochondrial DNA mutation, MELAS syndrome mutation). Med Sci Monit 8: CR767- CR773, 2002 Reardon W, Ross RJM, Sweeney MG, et al: Diabetes mellitus associated with a pathogenic point mutation in mitochondrial DNA. Lancet 340: 1376-1379, 1992 Silvestri G, Bertini E, Servidei S, et al: Maternally inherited cardiomyopathy: a new phenotype associated with the A to G AT nt.3243 of mitochondrial DNA, MELAS mutation). Muscle Nerve 20: 221-225, 1997 Takahashi K, Merchant SN, Miyazawa T, et al: Temporal bone histopathology and quantitative analysis of mitochondrial DNA in MELAS. Laryngoscope 113: 1362-1368, 2003 Thorburn DR, Dahl HH: Mitochondrial disorders: genetics, counseling, prenatal diagnosis and reproductive options. Am J Med Genet 106: 102-114, 2001 van den Ouweland JMW, Lemkes HHPJ, Ruitenbeek W, et al: Mutation in mitochondrial tRNALeu(UUR, gene in a large pedigree with maternally transmitted Type II diabetes mellitus and deafness. Nat Genet 1: 368-371, 1992 Willems PJ: Genetic causes of hearing loss. N Engl J Med 342: 1101-1109, 2000 Yamasoba T, Tsukuda K, Oka Y, et al: Cochlear histopathology associated with mitochondrial transfer RNA(Leu(UUR), gene mutation. Neurology 52: 1705-1707, 1999 Zeviani M, Spinazzola A, Carelli V: Nuclear genes in mitochondrial disorders. Curr Opin Genet Dev 13: 262-270, 2003 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 82 EP 86 PG 5 ER PT J AU Bircan, S Candir, O Kapucuoglu, N AF Bircan, Sema Candir, Ozden Kapucuoglu, Nilgun TI The Effect of Tumor Invasion Patterns on Pathologic Stage of Bladder Urothelial Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE urinary bladder; urothelial carcinoma; invasion pattern ID urinary bladder; urothelial carcinoma; invasion pattern AB The aim of this study was to investigate tumor invasion pattern, its heterogeneity and association with histopathological features and stage in invasive urothelial carcinoma of the bladder. We studied 62 cases of invasive urothelial carcinoma of the bladder. World Health Organization (WHO) 1973, WHO/ISUP 1998 and WHO 1999 systems were used for tumor grading. Pathologic staging of each case was done according to 1997 TNM system. During evaluation of the slides three main tumor invasion patterns were detected: ''nodular”, ''trabecular” and ''infiltrative”. In addition, homogeneity or heterogeneity of invasion patterns was also recorded for each case. Of sixty-two invasive cases, 17 (27%) had nodular, 36 (58%) trabecular, and 9 (15%) infiltrative invasion pattern. There was a statistically significant difference between invasion patterns in relation to pathologic stage (pT) (p=0.001), but not to grade. Of the 17 cases with nodular invasion pattern and 36 tumors with trabecular invasion pattern, 13 (77%) and 26 (72%) were pT1, respectively, whereas 8 of 9 infiltrative cases (89%) were advanced stage (pT2-3). According to heterogeneity, forty-two cases (68%) had homogeneous, while the remaining 20 (32%) had heterogeneous invasion pattern. Of the 42 homogeneous cases 34 (81%) were pT1, whereas 14 of 20 heterogeneous cases (70%) were advanced stage (p=0.000). The different invasion patterns seem to have a large impact on pathologic stage, especially the infiltrative pattern. In addition, invasion heterogeneity appears to be of value in determination of biologic aggressiveness in urothelial carcinoma. C1 [Bircan, Sema] Suleyman Demirel University, Faculty of Medicine, Department of PathologyIsparta, Turkey. [Candir, Ozden] Suleyman Demirel University, Faculty of Medicine, Department of PathologyIsparta, Turkey. [Kapucuoglu, Nilgun] Suleyman Demirel University, Faculty of Medicine, Department of PathologyIsparta, Turkey. RP Bircan, S (reprint author), Suleyman Demirel University, Faculty of Medicine, Department of Pathology, Isparta, Turkey. EM bircans2000@yahoo.com CR Lapham RL, Grignon D, Ro JY: Pathologic prognostic parameters in bladder urothelial biopsy transurethral resection, and cystectomy specimens. Semin Diagn Pathol 14: 109-122, 1997 Lopez-Beltran A, Cheng L: Stage pT1 bladder carcinoma: diagnostic criteria, pitfalls and prognostic significance. Pathology 35: 484-491, 2003 Cheng L, Weaver AL, Bostwick DG: Predicting extravesical extension of bladder carcinoma: A novel method based on micrometer measurement of the depth of invasion in transurethral resection specimens. Urology 55: 668-672, 2000 Bostwick DG, Mikuz G: Urothelial papillary, exophytic, neoplasms. Virchows Arch 441: 109-116, 2002 Amin MB, Gomez JA, Young RH: Urothelial transitional cell carcinoma with endophytic growth patterns: A discussion of patterns of invasion and problems associated with assessment of invasion in 18 cases. Am J Surg Pathol 21: 1057-1068, 1997 Epstein JI, Amin MB, Reuter VR, et al: The Bladder Consensus Conference Committee. The World Health Organization/International Society of Urological Pathology Consensus Classification of Urothelial, Transitional Cell, Neoplasms of the Urinary Bladder. Am J Surg Pathol 22: 1435-1448, 1998 Mazzucchelli L, Bacchi M, Studer UE, et al: Invasion depth is the most important prognostic factor for transitional cell carcinoma in a prospective trial of radical cystectomy and adjuvant chemotherapy. Int J Cancer 57: 15-20, 1994 Farrow GM, Utz DC: Observations on microinvasive transitional cell carcinoma of the urinary bladder. Clin Oncol 1: 609- 615, 1982 Bostwick DG, Ramnani D, Cheng L: Diagnosis and grading of bladder cancer and associated lesions. Urol Clin North Am 26: 493-507, 1999 Mostofi FK: Pathology and spread of carcinoma of the urinary bladder. In: Cancer of the Genitourinary Tract., Eds: Johnson DE, Samuels ML), Raven, New York, 1979, pp 303-308 Jimenez RE, Gheiler E, Oskanian P, et al: Grading the invasive component of urothelial carcinoma of the bladder and its relationship with progression-free survival. Am J Surg Pathol 24: 980-987, 2000 Cheng L, Neumann RM, Nehra A, et al: Cancer heterogeneity and its biologic implications in the grading of urothelial carcinoma. Cancer 88: 1663-1670, 2000 Billis A, Carvalho RB, Mattos AC, et al: Tumor grade heterogeneity in urothelial bladder carcinoma: Proposal of a system using combined numbers. Scand J Urol Nephrol 35: 275-279, 2001 Mostofi FK, Sobin LH, Torloni H: Histological typing of urinary bladder tumours. International Histological Classification of Tumours, No. 10. Geneva, Switzerland, World Health Organization, 1973 Busch C, Algaba F: The WHO/ISUP 1998 and WHO 1999 systems for malignancy grading of bladder cancer. Scientific foundation and translation to one another and previous systems. Virchows Arch 441: 105-108, 2002 Sobin LH, Wittekind C: International Union Against Cancer, UICC). TNM Classification of Malignant Tumours. 5th ed. New York: Wiley-Liss, 1997, pp187-190 Cheng L, Neumann RM, Weaver AL, et al: Predicting cancer progression in patients with stage T1 bladder carcinoma. J Clin Oncol 17: 3182-3187, 1999 Cheng L, Weaver AL, Neumann RM, et al: Substaging of T1 bladder carcinoma based on the depth of invasion as measured by micrometer: A new proposal. Cancer 86: 1035-1043, 1999 Smits G, Schaafsma E, Kiemeney L, et al: Microstaging of pT1 transitional cell carcinoma of the bladder: Identification of subgroups with distinct risks of progression. Urology 52: 1009- 1014, 1998 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 87 EP 91 PG 5 ER PT J AU Baki, M AF Baki, Marta TI What is the Role of Letrozole in Adjuvant Breast Carcinoma Setting? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports AB A meeting for Letrozole clinical investigators was organized in Orlando, joined to ASCO Conference, 2005, in order to review the newest data in adjuvant breast cancer trials. C1 [Baki, Marta] Szent Margit Hospital, V. Department of Internal Medicine - OncologyBudapest, Hungary. RP Baki, M (reprint author), Szent Margit Hospital, V. Department of Internal Medicine - Oncology, Budapest, Hungary. CR Goss PE, Ingle JN, Martino S, et al. A randomized trial of letrozole in postmenopausal women after five years of tamoxifen therapy for early-stage breast cancer. New Engl J Med 349: 1793-1802, 2003 Wasan KM, Goss PE, Pritchard PH, et al. The influence of letrozole on serum lipid concentrations in postmenopausal women with primary breast cancer who have completed 5 years adjuvant tamoxifen, NCIC CTG MA.17L). Ann Oncol 16: 707-715, 2005 Thurlimannn B et al for the BIG I-98 Collaborative coordinated by the International Breast Cancer Study Group. Letrozole as adjuvant endocrine therapy for postmenopausal women with receptor positive breast cancer. First results of IBCSG 18-98/BIG I-98. Presented at 9th International Conference on Primary Therapy of Early Breast Cancer: January 26-29, 2005, St. Gallen, Abstr. S4 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 91 EP 91 PG 1 ER PT J AU Simsa, P Teillaud, JL Stott, ID Toth, J Kotlan, B AF Simsa, Peter Teillaud, Jean-Luc Stott, I David Toth, Jozsef Kotlan, Beatrix TI Tumor-Infiltrating B Cell Immunoglobulin Variable Region Gene Usage in Invasive Ductal Breast Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunoglobulin variable region; breast ductal carcinoma; tumor-infiltrating lymphocytes ID immunoglobulin variable region; breast ductal carcinoma; tumor-infiltrating lymphocytes AB A major focus of tumor immunology is to reveal the potential role and capacity of immunocompetent cells found in different solid tumor tissues. The most abundant infiltrating cells (TIL), the T lymphocytes have been investigated in details concerning T-cell receptor usage and specificity. However, B cells have hardly been investigated in this respect, although high cellular B-cell infiltration has been correlated with improved patients’ survival in some breast carcinomas. This led to our objectives to study variable region gene usage of the tumor-infiltrating B cells in different breast carcinoma types. By defining the immunoglobulin repertoire of the tumor-infiltrating B lymphocytes in the most common invasive ductal carcinoma (IDC) of the breast we compared it to the rare medullary breast carcinoma (MBC). After phenotyping infiltrating ductal carcinomas, B cells were obtained from tumor tissue by microdissection technique. Numerous rearranged TIL-B immunoglobulin heavy chain V genes (VH) were amplified, cloned, sequenced, and comparatively analyzed. Some characteristics were found for both breast carcinoma types. The immunoglobulins produced by TIL-B in ductal carcinoma are highly matured and oligoclonal. We conclude that Ig variable region gene usage reveals similar and distinguishable characteristics of TIL-B immunoglobulin repertoires, which are representative of the nature of the immune responses in invasive ductal and medullary breast carcinomas. C1 [Simsa, Peter] National Medical Center, Department of Hematology and Stem Cell Transplantation, Szabolcs u 33-35., H-1135 Budapest, Hungary. [Teillaud, Jean-Luc] Centre de Recherches Biomedicales des Cordeliers, INSERM U255Paris, France. [Stott, I David] University of Glasgow, Western Infirmary, Division of Immunology, Infection and InflammationGlasgow, Scotland, UK. [Toth, Jozsef] National Institute of OncologyBudapest, Hungary. [Kotlan, Beatrix] National Medical Center, Department of Hematology and Stem Cell Transplantation, Szabolcs u 33-35., H-1135 Budapest, Hungary. RP Kotlan, B (reprint author), National Medical Center, Department of Hematology and Stem Cell Transplantation, H-1135 Budapest, Hungary. EM KotlanB@netscape.net CR Hadden JW: The immunology and immunotherapy of breast cancer: an update. Int J Immunopharmacol 21: 79-101, 1999 Ben-Hur H, Cohen O, Schneider D, et al: The role of lymphocytes and macrophages in human breast tumorigenesis: an immunohistochemical and morphometric study. Anticancer Res 22: 1231–1238, 2002 Di Girolamo V, Lauens RP, Coronato S, et al: Quantitative and functional study of breast cancer axillary lymph nodes and those draining other malignant tumors. J Exp Clin Cancer Res 19:155-159, 2000 Allan CP, Turtle CJ, Mainwaring PN, et al: The immune response to breast cancer, and the case for DC immunotherapy. Cytotherapy 6:154-163, 2004 Balch CM, Riley LB, Bae YJ, et al: Patterns of human tumorinfiltrating lymphocytes in 120 human cancers. Arch Surg 125:200-205, 1990 Schwartzentruber DJ, Solomon D, Rosenberg SA, et al: Characterization of lymphocytes infiltrating human breast cancer: specific immune reactivity detected by measuring cytokine secretion. J Immunother 12:1-12, 1992 Lin E, Pollard JW: Role of infiltrated leucocytes in tumour growth and spread. Br J Cancer 90:2053-8, 2004 Ladanyi A: Function and prognostic significance of immune cells infiltrating human tumors., In Hungarian, Magy Onkol 48:49-56, 2004 Filaci G, Fravegy M, Fenoglio D, et al: Non-antigen specific CD8+ T suppressor lymphocytes. Clin Exp Med 4:86-92, 2004 Whitford P, Mallon EA, George WD, et al: Flow cytometric analysis of tumor infiltrating lymphocytes in breast cancer. Br J Cancer 62:971–975, 1990 Lee RS, Schlumberger M, Caillou B, et al: Phenotypic and functional characterisation of tumor-infiltrating lymphocytes. Eur J Cancer 32: 1233-1239, 1996 Chin Y, Janssens J, Bleus J, et al: Characterization of tumor infiltrating lymphocytes in human breast cancer. Eur J Cancer Prev 2, Suppl.33): 27-31, 1993 Grekou AN, Toliou T, Stravoravdi P, et al: Correlation of apoptosis with the distribution and composition of lymphocytic infiltrate in human breast carcinomas. Anticancer Res 16:3991- 3995, 1996 Ridolfi RL, Rosen PP, Port A, et al: Medullary carcinoma of the breast. A clinicopathology study with 10 year follow-up. Cancer 40:1365-1385, 1977 Coronella JA, Spier C, Welch M, et al: Antigen-driven oligoclonal expansion of tumor-infiltrating B cells in infiltrating ductal carcinoma of the breast. J Immunol 169:1829-1836, 2002 Nzula S, Going JJ, Stott DI: Antigen driven clonal proliferation, somatic hypermutation, and selection of B lymphocytes infiltrating human ductal breast carcinomas. Cancer Res 63: 3275–3280, 2003 Moore OS, Foote FW: The relatively favourable prognosis of medullary carcinoma of the breast. Cancer 2: 635-640, 1949 Marks JD, Tristem M, Karpas A, et al: Oligonucleotide primers for polymerase chain reaction amplification of human immunoglobulin variable genes and design of family-specific oligonucleotide probes. Eur J Immunol 21:985-991, 1991 Sims GP, Shiono H, Willcox N, et al: Somatic hypermutation and selection of B cells in thymic germinal centers responding to acetylcholine receptor in myasthenia gravis. J Immunol 167:1935-1944, 2001 Tomlinson IM, Williams SC, Ignatovich O, et al: Human VBASE directory of immunoglobulin genes. Cambridge, UK: MRC Centre for Protein Engineering, 1997 Kotlan B, Gruel N, Zafrani B, et al: Immunoglobulin variable regions usage by B-lymphocytes infiltrating a human breast medullary carcinoma. Immunol Lett 65:143-151, 1999 Cuisinier AM, Guigou V, Boubli, et al: Preferential expression of VH5 and VH6 immunoglobulin genes in early human B-cell ontogeny. Scand J Immunol 30: 493-497, 1989 Kotlan B, Simsa P, Foldi J, et al: Immunoglobulin repertoire of B lymphocytes infiltrating breast medullary carcinoma. Human Antibodies 12: 113-121, 2003 Ito T, Saga S, Nagayoshi S, et al: Class distribution of immunoglobulin-containing plasma cells in the stroma of medullary carcinoma of breast. Breast Cancer Res Treat 7:97- 103, 1986 Lazzaro B, Anderson AE, Kajdacsy-Balla A, et al: Antigenic characterization of medullary carcinoma of the breast: HLADR expression in lymph node positive cases. Appl Immunohistochem Mol Morphol 9: 234-241, 2001 Gaffey MJ, Frierson HF, Mills SE, et al: Medullary carcinoma of the breast: identification of lymphocyte subpopulations and their significance. Mod Pathol 6:721-728, 1993 Berek C, Berger A, Apel M: Maturation of the immune response in germinal centers. Cell 67:1121-1129, 1991 O’Brien PM, Tsirimonaki E, Coomber DW, et al: Immunoglobulin genes expressed by B-lymphocytes infiltrating cervical carcinomas show evidence of antigen-driven selection. Cancer Immunol Immunother 50:523–532, 2001 Friedman DF, Kraj P, Silberstein LE: VH4.21 Expression in the normal human B-cell repertoire. In: Immunoglobulin Gene Expression in Development and Disease., Eds: Casali P, Silberstein LE, Ann New York Acad Sc 764:264-274, 1995 Stollar BD: The expressed heavy chain V gene repertoire of circulating B cells in normal adults. In: Immunoglobulin Gene Expression in Development and Disease., Eds: Casali P, Silberstein LE, Ann New York Acad Sc 764:264- 274, 1995 Kotlan B, Simsa P, Teillaud JL et al: Novel ganglioside antigen identified by B cells in human medullary breast carcinomas. The proof of principle concerning the infiltrating B lymphocytes. J Immun, 2005, in press) NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 92 EP 97 PG 6 ER PT J AU Kamali-Sarvestani, E Gharesi-Fard, B Sarvari, J Talei, AAR AF Kamali-Sarvestani, Eskandar Gharesi-Fard, Behrouz Sarvari, Jamal Talei, Abd-Al-Rasoul TI Association of TNF-α and TNF-β Gene Polymorphism with Steroid Receptor Expression in Breast Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; steroid receptors; TNF; polymorphism ID breast cancer; steroid receptors; TNF; polymorphism AB The presence of estrogen and progesterone receptors is correlated with good prognosis in breast cancer. The effect of TNF-α on down-regulation of estrogen receptor and blocking the proliferative response of breast cancer cells to estradiol have been demonstrated. However, the effect of TNFA and TNFB gene polymorphisms on the expression of steroid receptors in breast cancer cells is not well documented. Therefore, 160 breast cancer patients were recruited to investigate the association of TNFA and TNFB gene polymorphism with the level of steroid receptor expression. This association was not found to be significant for TNFA polymorphism and estrogen receptor expression (p=0.07). However, when combined genotypes of TNFA and TNFB polymorphism was considered, homozygous patients for lower TNF-α producer genotypes (TNFA1/A1 and TNFB1/B1) showed significantly higher progesterone receptor expression (p=0.041). Our findings indicate that TNFA and TNFB polymorphisms may be associated with the levels of steroid receptor expression in breast cancer patients. Further studies on a larger group of breast cancer patients are recommended. C1 [Kamali-Sarvestani, Eskandar] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Gharesi-Fard, Behrouz] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Sarvari, Jamal] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Talei, Abd-Al-Rasoul] Medical School, Shiraz University of Medical Sciences, Department of SurgeryShiraz, Iran. RP Kamali-Sarvestani, E (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Immunology, Shiraz, Iran. EM immunol2@sums.ac.ir CR Akerblom, IW, Mellon, PL: Nuclear Hormone Receptors, Ed: Parker MG), Academic Press Ltd, London 1991, pp 175-196 Ackerman GE, Smith ME, Mendelson CR, et al: Aromatization of androstenedione by human adipose tissue stromal cells in monolayer culture. J Clin Endocrinol Metab 53: 412-417, 1981 Zhao Y, Nichols JE, Bulnn SE, et al: Aromatase P450 gene expression in human adipose tissue. J Biol Chem 270: 16449–16457, 1995 Newman SP, Purohit A, Ghilchik MW, et al: Regulation of steroid sulphatase expression and activity in breast cancer. J Steroid Biochem Mol Biol 75: 259–264, 2000 Duncan LJ, Coldham NG, Reed MJ: The interaction of cytokines in regulating oestradiol 17β-hydroxy steroid dehydrogenase activity in MCF-7 cells. J Steroid Biochem Mol Biol 49: 63–68, 1994 Danforth DN Jr, Sgagias MK: Tumor necrosis factor-alpha modulates oestradiol responsiveness of MCF-7 breast cancer cells in vitro. J Endocrinol 138: 517-528, 1993 Smith CA, Farrah T, Goodwin RG: The TNF receptor superfamily of cellular and viral proteins: activation, costimulation, and death. Cell 25: 959-962, 1994 Wilson AG, Symons JA, McDowell TL, et al: Effects of a polymorphism in the human tumor necrosis factor alpha promoter on transcriptional activity. Proc Natl Acad Sci USA 94: 3195-3199, 1997 Messer G, Spengler U, Jung MC, et al: Polymorphic structure of the tumor necrosis factor, TNF, locus: an NcoI polymorphism in the first intron of the human TNF-beta gene correlates with a variant amino acid in position 26 and a reduced level of TNF-beta production. J Exp Med 173: 209-219, 1991 Stuber F, Petersen M, Bokelmann F, Schade U. A: genomic polymorphism within the tumor necrosis factor locus influences plasma tumor necrosis factor-α concentrations and outcome of patients with severe sepsis. Crit Care Med 24: 381-384, 1996 Verjans GM, Brinkman BM, Van Doornik CE, et al: Polymorphism of tumor necrosis factor-alpha, TNF-alpha, at position -308 in relation to ankylosing spondylitis. Clin Exp Immunol 97: 45-47, 1994 Dexter DL, Kowalski HM, Blazar BA, et al: Heterogeneity of tumor cells from a single mouse mammary tumor. Cancer Res 38: 3174- 3181, 1978 Roodi N, Bailey LR, Kao WY, et al: Estrogen receptor gene analysis in estrogen receptor-positive and receptor-negative primary breast cancer. J Natl Cancer Inst 15: 446-451, 1995 Kalkhoven E, Wissink S, van der Saag PT, et al: Negative interaction between the RelA, p65, subunit of NF-kB and the progesterone receptor. J Biol Chem 271: 6217-6224, 1996 Wilson AG, Symons JA, McDowell TL, et al: Effects of a tumor necrosis factor, TNF-α, promoter base transition on transcriptional activity. Br J Rheumatol 33:89, 1994 Bouma G, Crusius JBA, Pool MO, et al: Secretion of tumour necrosis factor alpha and lymphotoxin alpha in relation to polymorphisms in the TNF gene and HLA-DR alleles: Relevance for immunology of bowel disease. Scand J Immunol 43: 456-463, 1996 Gadducci A, Ferdeghini M, Castellani C, et al: Serum levels of tumor necrosis factor, TNF), soluble receptors for TNF, 55- and 75- kDa sTNFr), and soluble CD14, sCD14, in epithelial ovarian cancer. Gynecol Oncol 58: 184-188, 1995 Okamoto T, Watanabe N, Yamauchi N, et al: Endogenous tumor necrosis factor exerts its protective function intracellularly against the cytotoxicity of exogenous tumor necrosis factor. Cancer Res 52: 5278-5281, 1992 Clark GM, McGuire WL: Steroid receptors and other prognostic factors in primary breast cancer. Semin Oncol 15: 20-25, 1998 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 99 EP 102 PG 4 ER PT J AU Cavalcanti, dBCF Alves, AFV Pereira, J Kanamura, TC Wakamatsu, A Saldanha, BL AF Cavalcanti, de Barros Correia Fernanda Alves, Avancini Ferreira Venancio Pereira, Julio Kanamura, T Cristina Wakamatsu, Alda Saldanha, Balthazar Luis TI Proliferative Lesions of Prostate: a Multivariate Approach to Differential Diagnosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prostatic hyperplasia; prostatic intraepithelial neoplasia; atypical small acinar proliferation; prostate cancer; histological features ID prostatic hyperplasia; prostatic intraepithelial neoplasia; atypical small acinar proliferation; prostate cancer; histological features AB Prostatic needle biopsies from 142 patients were studied: 61 cases were ''benign”, 19 atypical small acinar proliferation, 31 high-grade prostatic intraepithelial neoplasia, and 31 adenocarcinoma. Using univariate analysis of 46 previously described morphological features, 16 variables were selected, which were followed by multivariate discriminant analysis. Of these parameters, seven (glandular fusion, crystalloids, nucleolomegaly, papillary architecture, visibility of basal cell layer, areas of normal luminal cell nucleus/cytoplasm ratio and areas of high luminal cell nucleus/cytoplasm ratio) remained significant in discriminating the groups. Multivariate analysis selected a small panel of histological features as those most helpful in the differential diagnosis of proliferative lesions in prostate biopsies. C1 [Cavalcanti, de Barros Correia Fernanda] Sao Paulo University School of Medicine, Department of Pathology, Av. Dr. Arnaldo 455, 01246-802 Sao Paulo, Brazil. [Alves, Avancini Ferreira Venancio] Sao Paulo University School of Medicine, Department of Pathology, Av. Dr. Arnaldo 455, 01246-802 Sao Paulo, Brazil. [Pereira, Julio] Sao Paulo University School of Medicine, Department of Pathology, Av. Dr. Arnaldo 455, 01246-802 Sao Paulo, Brazil. [Kanamura, T Cristina] Sao Paulo Public Health Service, Adolfo Lutz Institute, Division of PathologySao Paulo, Brazil. [Wakamatsu, Alda] Sao Paulo Public Health Service, Adolfo Lutz Institute, Division of PathologySao Paulo, Brazil. [Saldanha, Balthazar Luis] Sao Paulo University School of Medicine, Department of Pathology, Av. Dr. Arnaldo 455, 01246-802 Sao Paulo, Brazil. RP Alves, AFV (reprint author), Sao Paulo University School of Medicine, Department of Pathology, 01246-802 Sao Paulo, Brazil. EM venancio@uol.com.br CR IBGE: DPE/DEPIS; MS/InCA/CONPREV/Divisao de Epidemiologia e Avaliacao S/SIM- Sistema de Informacao sobre mortalidade [http:www.ibge.gov.br/], 2000 Arista-Nasr J, Cortes E, Pichardo R: Low grade adenocarcinoma simulating benign glandular lesions in needle prostatic biopsy. Rev Invest Clin 49:37-40, 1997 Gilbert ES: On discrimination using qualitative variables. J Am Stat Assoc 63: 1399-1412, 1968 Moore DH: Evaluation of five discrimination procedures for binary variables. J Am Stat Assoc 68: 399, 1973 Nasir A, Copeland J, Gillespine JW, et al: Preneoplastic lesions of the prostate – clinical, pathological and molecular biological aspects. In Vivo 6:557-566, 2002 Brawer MK: Prostatic intraepithelial neoplasia: A premalignant lesion. Hum Pathol 23: 242-248, 1992 Cheville JC, Bostwick DG: Postatrophic Hyperplasia of the Prostate. Am J Surg Pathol 19:1068-1076, 1995 Cleary KR, Choi HY, Ayala AG: Basal cell hyperplasia of the prostate. Am J Clin Pathol 80: 850-854, 1983 Gleason DF, 1992). Histologic grading of prostate cancer. A perspective. Hum Pathol 23: 273-279, 1992 Gleason DF, Mellinger GT, Veterans Administration Cooperative Urological Research Group: Prediction of prognosis for prostatic adenocarcinoma by combined histological grading and clinical staging. J Urol 111: 58-64, 1974 Grignon DJ, Ro JY, Ordonez NG, et al: Basal cell hyperplasia, adenoid basal cell tumor, and adenoid cystic carcinoma of the prostate gland: An immunohistochemical study. Hum Pathol 19: 1425-1433, 1988 Iczkowski KA, Bassler TJ, Schwob VS, et al: Diagnosis of “suspicious for malignancy” in prostate biopsies: predictive value for cancer. Urology 51:749-757, 1998 Iczkowski KA, MacLennan GT, Bostwick DG: Atypical small acinar proliferation suspicious for malignancy in prostate needle biopsies: clinical significance in 33 cases. Am J Surg Pathol 21:1489-1495, 1997 Moore RA: Benign hypertrophy of the prostate - a morphological study. J Urol 50: 580-710, 1943 Purnell DM, Heatfield BM, Anthony RL, Trump BF: Immunohistochemistry of the cytoskeleton of human prostatic epithelium. Am J Pathol 126:384-395, 1987 Totten RS, Heineman MW, Hudson PB, et al: Microscopic differential diagnosis of latent carcinoma of prostate. Arch Pathol 55: 131-141, 1953 Varma M, Lee MW, Tamboli P, et al: Morphologic criteria for the diagnosis of prostatic adenocarcinoma in needle biopsy specimens. A study of 250 consecutive cases in a routine surgical pathology practice. Arch Pathol Lab Med 126:554-561, 2002 Cheng L, Cheville JC, Bostwick DG: Diagnosis of prostate cancer in needle biopsies after radiation therapy. Am J Surg Pathol 23: 1173- 1183, 1999 Bostwick DG, Sgriley J, Grignon D et al: Atypical adenomatous hyperplasia of the prostate: Morphologic criteria for its distinction from well differentiated carcinoma. Hum Pathol 24: 819-832, 1993 Helpap B: Differential diagnosis of glandular proliferations in the prostate. A conventional and immunohistochemical approach. Virchows Arch 433:397-405, 1998 Oliai BR, Kahane H, Epstein JI: Can basal cells be seen in adenocarcinoma of prostate?: an immunohistochemical study using high molecular weight cytokeratin, clone 34bE12, antibody. Am J Surg Pathol 26:1151-1160, 2002 Googe PB, Mcginley KM, Fitzgibbon JF: Anticytokeratin antibody 34 b E12 staining in prostate carcinoma. Am J Clin Pathol 107: 219- 223, 1997 Amin MB, Schultz DS, Zarbo RJ: Analysis of cribriform morphology in prostatic neoplasia using antibody to high molecular weight cytokeratins. Arch Pathol Lab Med 118: 260-264, 1994 Millar EKA, Sharma NK, Lessels AM: Ductal, endometrioid, adenocarcinoma of the prostate: a clinicopathological study of 16 cases. Histopathology 29: 11-19, 1996 Bostwick DG: High grade prostatic intraepithelial neoplasia and early invasion in prostatic carcinoma. Cancer 59:788-794, 1987 Mc Neal JE, Bostwick DG: Intraductal dysplasia: a premalignant lesion of the prostate. Hum Pathol 17:64-71, 1986 Montironi R, Pomante R, Colanzi P, et al: Diagnostic distance of high grade prostatic intraepithelial neoplasia from normal prostate and adenocarcinoma. J Clin Pathol 50:775-782, 1997 Rubin MA, de La Taille A, Bagiella E, et al: Cribriform carcinoma of the prostate and cribriform prostatic intraepithelial neoplasia: incidence and clinical implications. Am J Surg Pathol 22:840-848, 1998 Brawer MK, Peehl DM, Stamey TA, Bostwick DG: Keratin immunoreactivity in the benign and neoplastic human prostate. Cancer Res 45: 3663-3667, 1985 Wojno K.J, Epstein J: The utility of basal cell specific anti-cytokeratin antibody, 34 B E 12, in the diagnosis of prostate cancer - a review of 228 cases. Am J Surg Pathol 19: 251-260, 1995 Kahane H, Sharp JW, Shuman GB, et al: Utilization of high molecular weight cytokeratin on prostate needle biopsies in an independent laboratory. Urology 45: 981-986, 1995 O’Malley FP, Grignon DJ, Shum DT: Usefulness of immunoperoxidase staining with high molecular weight cytokeratin in the differential diagnosis of small acinar lesions of the prostate gland. Virchows Arch Pathol Anat 417:191-196, 1990 Ximing JY, Lecksell K, Gaudin P, Epstein J: Rare expression of high molecular weight cytokeratin in adenocarcinoma of the prostate gland. A study of 100 cases of metastatic and locally advanced prostate cancer. Am J Surg Pathol 23:147-152, 1999 Amin MB, Tamboli P, Varma M, Srigley JR: Postatrophic hyperplasia of the prostate gland: a detailed analysis of its morphology in needle biopsy specimens. Am J Surg Pathol 23:925-931, 1999 Troxel DB, Sabella JD: Problem areas in pathology practice uncovered by a review of malpractice claims. Am J Surg Pathol 18:821- 831, 1994 Ruska KM, Sauvageot J, Epstein JI: Histology and cellular kinetics of prostatic atrophy. Am J Surg Pathol 22:1073-1077, 1998 Leroy X, Aubert S, Villens A, et al: Minimal focus of adenocarcinoma on prostate biopsy: clinicopathological correlations. J Clin Pathol 56: 230-232, 2003 Anton RC, Kattan MW, Chakraborty S, Wheeler TM: Postatrophic hyperplasia of the prostate: lack of association with prostate cancer. Am J Surg Pathol 23:932-936, 1993 Bostwick DG, Amin MB, Dundore P, et al: Architectural patterns of high grade prostatic intraepithelial neoplasia. Hum Pathol 24:298- 310, 1993 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 103 EP 107 PG 5 ER PT J AU Matusan, K Dordevic, G Mozetic, V Lucin, K AF Matusan, Koviljka Dordevic, Gordana Mozetic, Vladimir Lucin, Ksenija TI Expression of Osteopontin and CD44 Molecule in Papillary Renal Cell Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE papillary renal cell carcinoma; cell adhesion; CD44s antigen; CD44v6 antigen; osteopontin; immunohistochemistry ID papillary renal cell carcinoma; cell adhesion; CD44s antigen; CD44v6 antigen; osteopontin; immunohistochemistry AB The aim of the study was to analyze the expression of CD44 adhesion molecule and its ligand osteopontin in papillary renal cell tumors, and to assess the possible prognostic significance of CD44 and osteopontin expression in papillary renal cell carcinomas. The expression of the standard and v6 exon containing isoforms of CD44 molecule, as well as of its ligand osteopontin, was immunohistochemically evaluated in 43 papillary renal cell tumors, which included 5 adenomas and 38 carcinomas. In order to assess their prognostic significance, the results obtained in papillary renal cell carcinomas were compared to usual clinicopathological parameters such as tumor size, histological grade, pathological stage, and Ki-67 proliferation index. Normal renal tissue was negative for CD44s and v6 isoforms, while the expression of osteopontin was found in distal tubular epithelial cells in the form of cytoplasmic granular positivity. CD44s and v6 isoforms were upregulated in 22 (58%) and 12 (32%) out of 38 carcinomas, respectively. Among all clinicopathological parameters examined, we only found significant association of CD44s-positive carcinomas with lower pathological stage (p=0.026). Papillary renal cell adenomas were generally negative for CD44s, except for focal positivity found in one sample. The osteopontin protein was detected in all adenomas and all papillary renal cell carcinomas, except one. Our results show constitutive expression of osteopontin in papillary renal tumors, including papillary renal cell adenomas. The upregulation of CD44s and v6 isoforms, although found in a considerable number of papillary renal cell carcinomas, does not appear to have any prognostic value in this type of renal cancer. C1 [Matusan, Koviljka] Rijeka University School of Medicine, Department of Pathology, Brace Branchetta 20, 51 000 Rijeka, Croatia. [Dordevic, Gordana] Rijeka University School of Medicine, Department of Pathology, Brace Branchetta 20, 51 000 Rijeka, Croatia. [Mozetic, Vladimir] Clinical Hospital Center, Department of UrologyRijeka, Croatia. [Lucin, Ksenija] Rijeka University School of Medicine, Department of Pathology, Brace Branchetta 20, 51 000 Rijeka, Croatia. RP Lucin, K (reprint author), Rijeka University School of Medicine, Department of Pathology, 51 000 Rijeka, Croatia. EM ksenijal@medri.hr CR Agrawal D, Chen T, Irby R, et al: Osteopontin identified as lead marker of colon cancer progression, using pooled sample expression profiling. J Natl Cancer Inst 94:513-521, 2002 Amin MB, Corless CL, Renshaw AA, et al: Papillary, chromophil, renal cell carcinoma: histomorphologic characteristics and evaluation of conventional pathologic prognostic parameters in 62 cases. Am J Surg Pathol 21:621-635, 1997 Ariztia EV, Subbarao V, Solt DB, et al: Osteopontin contributes to hepatocyte growth factor-induced tumor growth and metastasis formation. Exp Cell Res 288:257-267, 2003 Brown LF, Papadopoulos-Sergiou A, Berse B, et al: Osteopontin expression and distribution in human carcinomas. Am J Pathol 145:610-623, 1994 Coppola D, Szabo M, Boulware D, et al: Correlation of osteopontin protein expression and pathological stage across a wide variety of tumor histogenesis. Clin Cancer Res 10:184-190, 2004 Craig AM, Denhardt DT: The murine gene encoding secreted phosphoprotein 1, osteopontin): promoter structure, activity, and induction in vivo by estrogen and progesterone. Gene 100:163-171, 1991 Delahunt B, Eble JN: Papillary renal cell carcinoma: A clinicopathologic and immunohistochemical study of 105 tumors. Mod Pathol 10:537-544, 1997 Eble JN, Sauter G, Epstein JI, Sesterhenn IA: WHO Classification of Tumours. Pathology and Genetics of Tumours of the Urinary System and Male Genital Organs. 6st vol. IARC Press, Lyon, France), 2004, pp: 9-87 Fuhrman S, Lasky L, Lumas C: Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol 6:655-663, 1982 Gilcrease MZ, Guzman-Paz M, Niehans G, et al: Correlation of CD44s expression in renal clear cell carcinomas with subsequent tumor progression or recurrence. Cancer 86:2320-2326, 1999 Glukhova L, Goguel AF, Chudoba I, et al: Overexpression of 7q31 and 17q in renal cell carcinoma. Genes Chromosomes Cancer 5:348- 356, 1998 Guinan P, Sobin LH, Algaba F, et al: TNM staging of renal cell carcinoma: Workgroup No. 3. Union International Contre le Cancer, UICC, and the American Joint Committee on Cancer, AJCC). Cancer 80:992- 993, 1997 Gunthert U, Stauder R, Mayer B, et al: CD44 variant isoforms involved in human tumor progression? Cancer Surv 24:19-42, 1995 Guo X, Zhang YP, Mitchell DA, et al: Identification of a ras-activated enhancer in the mouse osteopontin promoter and its interaction with a putative ETS-related transcription factor whose activity correlates with the metastatic potential of the cell. Mol Cell Biol 15:476- 487, 1995 Heider KH, Ratschek M, Zatloukal K, et al: Expression of CD44 isoforms in human renal cell carcinomas. Virchows Arch 428:267-273, 1996 Kovacs G, Fuzesi L, Emanual A, et al: Cytogenetics of papillary renal cell tumors. Genes Chromosomes Cancer 3:249-255, 1991 Lubensky IA, Schmidt L, Zhuang Z, et al: Hereditary and sporadic papillary renal carcinomas with c-met mutations share a distinct morphological phenotype. Am J Pathol 155:517-526, 1999 Lucin K, Matusan K, Dordevic G, et al: The expression of osteopontin in renal cell carcinomas. In: Proceedings of the Second Intercontinental Congress of Pathology., Eds: Soares F, Vassallo J, Torres LFB), Medimond, 2004, p: 151-154 Lucin K, Matusan K, Dordevic G, et al: Prognostic significance of CD44 molecule in renal cell carcinoma. Croat Med J 45:703-708, 2004 Oldberg A, Franzen A, Heinegard D: Cloning and sequence analysis of rat bone sialoprotein, osteopontin, cDNA reveals and Arg-Gly-Asp cell-binding sequence. Proc Natl Acad Sci USA 83:8819-8823, 1986 Pan HW, Ou YH, Peng SY, et al: Overexpression of osteopontin is associated with intrahepatic metastasis, early recurrence, and poorer prognosis of surgically resected hepatocellular carcinoma. Cancer 98:119- 127, 2003 Paradis V, Ferlicot S, Ghannam E, et al: CD44 is an independent prognostic factor in conventional renal cell carcinomas. J Urology 161:1984-1987, 1999 Rittling SR, Chambers AF: Role of osteopontin in tumour progression. Br J Cancer 90:1877-1881, 2004 Rudland PS, Platt-Higgins A, El-Tanani M, et al: Prognostic significance of the metastasis-associated protein osteopontin in human breast cancer. Cancer Res 62:3417-3427, 2002 Terpe HJ, Storkel S, Zimmer U, et al: Expression of CD44 isoforms in renal cell tumors. Positive correlation to tumor differentiation. Am J Pathol 148:453-463, 1996 Zbigniew R, Serge J: CD44 and the adhesion of neoplastic cells. J Clin Pathol: Mol Pathol 50:57-71, 1997 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 108 EP 113 PG 6 ER PT J AU Cambruzzi, E Zettler, GC Alexandre, OPC AF Cambruzzi, Eduardo Zettler, Galleano Claudio Alexandre, Osmar Pereira Claudio TI Expression of Ki-67 and Squamous Intraepithelial Lesions are Related with HPV in Endocervical Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE endocervical adenocarcinoma; bcl-2; Ki-67; p53; HPV ID endocervical adenocarcinoma; bcl-2; Ki-67; p53; HPV AB To estimate the association between human papillomavirus (HPV) status and the expression of p53, Ki-67 and bcl-2 in cases of endocervical adenocarcinoma, and the relation with squamous intraepithelial lesions (SIL) and age, 229 cases were selected, treated between 1995 and 2003 in the Hospital Nossa Senhora da Conceicao. All samples were evaluated by polymerase chain reaction to determine HPV status. Immunohistochemical technique was used to investigate the expression of p53, Ki-67 and bcl-2. The joint occurrence of endocervical adenocarcinoma and SIL were estimated too. In the 229 evaluated cases, 182 cases (79.48%) were associated with the presence of the HPV. The most common types were HPV18 (93 cases – 51.09%) and HPV16 (62 cases - 34.06%). Expression of Ki-67 (p=0.009) and the presence of SIL (p=0.018) were associated to HPV infection. Expression of p53 (p=0.647) and bcl-2 (p=0.671) were not related to HPV status. The mean age of the patients was 53.2 years, without clear correlation between age group and HPV (p=0.095). The presence of HPV, especially type 18 in endocervical adenocarcinoma suggests that this agent can be an important cofactor in the development and progression of glandular neoplasms of the uterine cervix. The joint occurrence of endocervical adenocarcinoma and SIL may support this hypothesis. HPV may promote an increased proliferation index in endocervical adenocarcinoma, shown by the expression of Ki-67. C1 [Cambruzzi, Eduardo] Fundacao Faculdade Federal de Ciencias Medicas de Porto Alegre, Av. Loureiro da Silva, 1500/1308, B. Centro, 90050-240 Porto Alegre, Rio Grande do Sul, Brazil. [Zettler, Galleano Claudio] Fundacao Faculdade Federal de Ciencias Medicas de Porto Alegre, Av. Loureiro da Silva, 1500/1308, B. Centro, 90050-240 Porto Alegre, Rio Grande do Sul, Brazil. [Alexandre, Osmar Pereira Claudio] Fundacao Faculdade Federal de Ciencias Medicas de Porto Alegre, Av. Loureiro da Silva, 1500/1308, B. Centro, 90050-240 Porto Alegre, Rio Grande do Sul, Brazil. RP Cambruzzi, E (reprint author), Fundacao Faculdade Federal de Ciencias Medicas de Porto Alegre, 90050-240 Porto Alegre, Brazil. EM dudacambruzzi@yahoo.com.br CR Alfsen GC, Thoresen SO, Kristensen GB: Histopathologic subtyping of cervical adenocarcinomas reveals increasing incidence rates of endometrioid tumors in all age groups. A population based study with review of all nonsquamous tumors in Norway from 1966 to 1970, 1976 to1980, and 1986 to 1990. Cancer 89:1291-1299, 2000 Loh TO, Wang PH, Yen MS, et al: Identifying local tumor variables for operable node-negative, margin-free patients with bulky cervical carcinoma of FIGO stage IB, IIA, and IIB without adjuvant therapies. Eur J Gynecol Oncol 22:420-422, 2001 Schoolland M, Alpress S, Sterret, GF: Adenocarcinoma of the cervix. Cancer 96:5-13, 2002 Colgan TJ, Auger M, McLaughlin JR: Histopathologic classification of cervical carcinomas and recognition of mucin-secreting squamous carcinoma. Int J Gynecol Pathol 12:64-69, 1993 Vesterinen E, Forss M, Nieminen U: Increase of cervical adenocarcinoma: a report of 520 cases of cervical carcinoma including 112 tumors with glandular elements. Gynecol Oncol 33:49-53, 1989 Vizcaino AP, Moreno V, Bosch FX: International trends in the incidence of cervical cancer: I. Adenocarcinoma and adenosquamous cell carcinomas. Int J Cancer 75:536-545, 1998 Young RH, Clement PB: Endocervical adenocarcinoma and its variants: their morphology and differential diagnosis. Histopathology 41:185-207, 2002 Andersson S, Rylander E, Larsson, B, et al: The role of human papillomavirus in cervical adenocarcinoma carcinogenesis. Eur J Cancer 37:246-250, 2001 Clarke B, Chetty R: Cell cycle aberrations in the pathogenesis of squamous cell carcinoma of the uterine cervix. Gynecol Oncol 82:238-246, 2001 Crum CP: Contemporary theories of cervical carcinogenesis: the virus, the host and the stem cell. Mod Pathol 13:243-251, 2000 Cuschieri KS, Cubie HA, Whitley MW, et al: Multiple high risk HPV infections are common in cervical neoplasia and young women in a cervical screening population. J Clin Pathol 57:68- 72, 2004 Gorstein F: Precursor lesions of squamous cell carcinoma of the cervix: are there reliable predictors of biologic behavior? Hum Pathol 31:1339-1340, 2000 Ali-Fehmi R, Qureshi F, Lawrence WD, Jacques SM: Apoptosis, proliferation and expression of p53 and bcl-2 in endocervical glandular intraepithelial lesions and invasive endocervical adenocarcinoma. Int J Gynecol Pathol 23:1-6, 2004 Bykov VJN, Wiman KG: Novel cancer therapy by reactivation of the p53 apoptosis pathway. Ann Med 35:458-465, 2003 Cheung TH, Chung TKH, Lo KWK, et al: Apoptosis related proteins in cervical intraepithelial neoplasia and squamous cell carcinoma of the cervix. Gynecol Oncol 86:14-18, 2002 Cho NH, Lim SY, Kim YT, et al: G2 check point in uterine cervical cancer with HPV 16 E6 according to p53 polymorphism and its screening value. Gynecol Oncol 90:15-22, 2003 Cordon-Cardo C: Mutations of cell cycle regulators: Biological and clinical implications for human neoplasia. Am J Pathol 147:545-555, 1995 Dimitrakakis C, Kymionis G, Diakomanolis E: The possible role of p53 and bcl-2 expression in cervical carcinomas and their premalignant lesions. Gynecol Oncol 77:129-136, 2000 Kurvinen K, Syrjanen S: P53 and bcl-2 proteins as prognostic markers in human papillomavirus associated cervical lesions. J Clin Oncol 14:2120-2130, 1996 Nakamura T, Nomura S, Sakai T, Nariya S: Expression of bcl-2 oncoprotein in gastrointestinal and uterine carcinomas and their premalignant lesions. Hum Pathol 28:309-316, 1997 Suzuko M, Olga IB, Satoru S, et al: Mitotic activity and apoptosis in endocervical glandular lesions. Int J Gynecol Pathol 21:12-133, 2002 Cameron RI, Maxwell P, Jenkins D, McCluggage WG: Immunohistochemical staining with MIB1, bcl-2 and p16 assists in the distinction of cervical glandular intraepithelial neoplasia from tubo-endometrial metaplasia, endometriosis and microglandular hyperplasia. Histopathology 41:313-321, 2002 Davidson B, Goldberg I, Lerner-Geva L, et al: Expression of topoisomerase II and Ki-67 in cervical carcinoma – clinicopathological study using immunohistochemistry. APMIS 108:209-215, 2000 Keating JT, Cviko A, Riethdorf S, et al: Ki-67, cyclin E and p16INK4 are complementary surrogate biomarkers for human papilloma virus-related cervical neoplasia. Am J Surg Pathol 25:884-891, 2001 McCluggage WG, Maxwell P, McBride HA, et al: Monoclonal antibodies Ki-67 and MIB1 in the distinction of tuboendometrial metaplasia from endocervical adenocarcinoma and adenocarcinoma in situ in formalin fixed material. Int J Gynecol Pathol 14:209-216, 1995 Heatley MK: Proliferation in the normal cervix and in preinvasive cervical lesions. J Clin Pathol 49:957, 1996 Colgan TJ, Lickrish GM: The topography and invasive potential of cervical adenocarcinoma in situ with and without associated squamous dysplasia. Gynecol Oncol 36:246-249, 1990 Lee MF, Chang MXC, Wu CH: Detection of human papillomavirus types in cervical adenocarcinoma by the polymerase chain reaction. Int J Gynaecol Obstet 63:265-270, 1998 Cook-Glenn CL, Keyhani-Rofagha S: Adenocarcinoma of the uterine associated with pregnancy: a retrospective 10 year investigative study. Diagn Cytopathol 18:393-397, 1998 Madeleine MM, Daling JR, Schwartz SM, et al: Human papillomavirus and long term oral contraceptive use increase the risk of adenocarcinoma in situ of the cervix. Cancer Epidemiol Biomarkers Prev 10:171-177, 2001 Nair P, Nair MK, Jayaprakash PG, et al: Decreased programmed cell death in the uterine cervix associated with high risk human papillomavirus infection. Pathol Oncol Res 5:95-103, 1999 Riethford L, Riethford S, Lee KR: Human papillomaviruses, expression of p16, and early endocervical glandular neoplasia. Hum Pathol 33:899-904, 2002 Stanczuk G, Kay P, Sibanda E, et al: Typing of human papillomavirus in Zimbabwean patients with invasive cancer of the uterine cancer. Acta Obstet Gynecol Scand 82:762-769, 2003 Nakagawa H, Sugano K, Fujii T, et al: Frequent detection of human papilloma viruses in cervical dysplasia by PCR singlestrand DNA-conformational polymorphism analysis. Anticancer Res 22:1655-1660, 2002 Nagai Y, Maehama T, Asato T, Kanazawa K: Detection of human papillomavirus DNA in primary and metastatic lesions of carcinoma of the cervix in women from Okinawa, Japan. Am J Clin Oncol 24:160-166, 2001 Lacey JV, Brinton LA, Abbas FM, et al: Oral contraceptives as risk factors for cervical adenocarcinomas and squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev 8:1079-1085, 1999 Andersson S, Larson B, Hjerpe A, et al: Adenocarcinoma of the uterine cervix: the presence of human papillomavirus and the method of detection. Acta Obstet Gynecol Scand 82:960-965, 2003 Al-Sader MH, Gorman G, Kay E, et al: Prognostic significance of mitosis, apoptosis and proliferation in cervical intraepithelial neoplasia, grade 3. J Pathol 178:50, 1996 Crum CP: Binary, Bethesda, system for cervical precursor lesions: a histologic perspective. Diagn Cytopathol 13:379- 385, 1995 McCluggage WG, McBride H, Maxwell P, et al: Immunohistochemical detection of p53 and bcl-2 proteins in neoplastic and non-neoplastic endocervical glandular lesions. Int J Gynecol Pathol 16:22-27, 1997 Grace VMB, Shalini JV, Iekha TTS, et al: Co-overexpression of p53 and bcl-2 proteins in HPV-induced squamous cell carcinoma of the uterine cervix. Gynecol Oncol 91:51-58, 2003 Koyamatsu Y, Yokoyama M, Nakao Y, et al: A comparative analysis of human papillomavirus type 16 and 18 and expression of p53 gene and Ki-67 in cervical, vaginal and vulvar carcinomas. Gynecol Oncol 90:547-551, 2003 Tjalma W, De Cuyper E, Weyler J, et al: Expression of bcl-2 in invasive and in situ carcinoma of the uterine cervix. Am J Obstet Gynecol 178:113-117, 1998 Cina SJ, Richardson MS, Austin MR: Immunohistochemical staining for Ki-67, carcinoembryonic antigen and p53 in the differential diagnosis of glandular lesions of the cervix. Mod Pathol 10:176-180, 1997 Hamilton A, Piccart M: The contribution of molecular markers to the prediction of response in the treatment of breast cancer: a review of the literature on HER-2, p53 and bcl-2. Ann Oncol 11:647-663, 2000 Park CS, Joo IS, Song SY, et al: An immunohistochemical analysis of heat shock protein 70, p53 and estrogen receptor status in carcinoma of the uterine cervix. Gynecol Oncol 74:53- 60, 1999 Toki T, Zhai YL, Park JS, Fujii S: Infrequent occurrence of highrisk human papillomavirus and of p53 mutation in minimal deviation adenocarcinoma of the cervix. Internat J Gynecol Pathol 18:215-219, 1999 Saegusa M, Takano Y, Hashimura M, et al: The possible role of bcl-2 expression in the progression of tumors of the uterine cervix. Cancer 76:2297-2303, 1995 Franquemont DW, Ward BE, Andersen WA: Prediction of high risk cervical papillomavirus infection by biopsy morphology. Am J Clin Pathol 92:577-582, 1989 Harris TG, Kulasingam SL, Kiviat NB, et al: Cigarette smoking, oncogenic human papillomavirus, Ki-67 antigen, and cervical intraepithelial neoplasia. Am J Epidemiol 159:834-842, 2004 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 114 EP 120 PG 7 ER PT J AU Culhaci, N Meteoglu, I Dundar, M Kocak, I AF Culhaci, Nil Meteoglu, Ibrahim Dundar, Mehmet Kocak, Izzet TI Histopathological Evaluation of Renal Vascular Changes in Rats Exposed to Passive Smoking SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE passive smoking; renal vasculature; rat ID passive smoking; renal vasculature; rat AB Cigarette smoking is an important risk factor for renal damage due to its effects on small interlobular arteries. We investigated the effects of long-term passive smoking on renal vascular structures in healthy rats exposed to smoke soon after birth. Forty-two Sprague-Dawley rats (21 males and 21 females) exposed to passive smoking comprised the experimental group and 33 non-exposed rats (17 males and 16 females) comprised the control group. The number of renal vessels, as well as the level of glomerular capillary sclerosis, hyalinosis of arterioles, and myointimal hyperplasia of arteries was assessed in renal biopsy specimens. The mean number of renal vessels in male and female rats exposed to passive smoke (21.71 and 13.81, respectively) did not significantly differ from the mean number of renal vessels in male and female control rats (22.47 and 13.06, respectively) (p>0.05). Levels of glomerulosclerosis, hyalinosis, and myointimal hyperplasia also did not differ between the experimental and control groups (p>0.05). Histopathologic evidence of renal vascular damage was not found in young rats exposed to passive smoke for 4 months. A longer or higher degree of exposure to cigarette smoke components may be required before such changes manifest, and aging and primary renal disease may play a role. C1 [Culhaci, Nil] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Meteoglu, Ibrahim] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Dundar, Mehmet] Adnan Menderes University, Faculty of Medicine, Department of UrologyAydin, Turkey. [Kocak, Izzet] Adnan Menderes University, Faculty of Medicine, Department of UrologyAydin, Turkey. RP Culhaci, N (reprint author), Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. EM nculhaci@hotmail.com CR Bartecchi CE, Mac Kenzie TD, Schrier RW: The human costs of tobacco use, first of two parts). N Engl J Med 330: 907-912, 1994 Orth SR, Ritz E, Schrier RW: The renal risks of smoking. Kidney Int 51: 1669-1677, 1997 Orth SR: Smoking – a risk factor for progression of renal disease. Kidney Blood Pres R 23: 202-204, 2000 Halimi J, Giraudeau B, Vol S, et al: Effects of current smoking and smoking discontinuation on renal function and proteinuria in the general population. Kidney Int 58: 1285-1292, 2000 Markowitz GS, Lin J, Valeri AM, et al. Idiopathic nodular glomerulosclerosis is a distinct clinicopathologic entity linked to hypertension and smoking. Hum Pathol 33: 826-835, 2002 Kasiske BL, Klinger D: Cigarette smoking in renal transplant recipients. J Am Soc Nephrol 11: 753-759, 2000 Lhotta K, Rumpelt HJ, Konig P, et al: Cigarette smoking and vascular pathology in renal biopsies. Kidney Int 61: 648-654, 2002 Gambaro G, Verlato F, Budakovic A, et al: Renal impairment in chronic cigarette smokers. J Am Soc Nephrol 9: 562-567, 1998 Remuzzi G: Cigarette smoking and renal function impairment. Am J Kidney Dis 33: 807-811, 1999 Black HR, Zeevi GR, Silten RM, Smith GJW: Effect of heavy cigarette smoking on renal and myocardial arterioles. Nephron 34: 173-179, 1983 Petrik PV, Gelabert HA, Moore WS, et al: Cigarette smoking accelerates carotid artery intimal hyperplasia in a dose-dependent manner. Stroke 26: 1409-1414, 1995 Raitakari OT, Adams MR, McCredie RJ, et al: Arterial endothelial dysfunction related to passive smoking is potentially reversible in healthy young adults. Ann Intern Med 130: 578-581, 1999 Oberai B, Adams CWM, High OB: Myocardial and renal arteriolar thickening in cigarette smokers. Atherosclerosis 52: 185- 190, 1984 Stengel B, Couchoud C, Cenee S, Hemon D: Age, blood pressure and smoking effects on chronic renal failure in primary glomerular nephropathies. Kidney Int 57: 2519-2526, 2000 Bleyer AJ, Shemanski LR, Burke GL, et al: Tobacco, hypertension, and vascular disease: risk factors for renal functional decline in an older population. Kidney Int 57: 2072-2079, 2000 Pfarrer C, Macara L, Leiser R, Kingdom J: Adaptive angiogenesis in placentas of heavy smokers. Lancet 354: 303, 1999 Odoni G, Ogata H, Viedt C, et al: Cigarette smoke condensate aggravates renal injury in the renal ablation model. Kidney Int 61: 2090-2098, 2002 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2005 VL 11 IS 2 BP 121 EP 124 PG 4 ER PT J AU Sapi, Z Papai, Zs Hruska, A Antal, I Bodo, M Orosz, Zs AF Sapi, Zoltan Papai, Zsuzsa Hruska, Anett Antal, Imre Bodo, Miklos Orosz, Zsolt TI Her-2 Oncogene Amplification, Chromosome 17 and DNA Ploidy Status in Synovial Sarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE synovial sarcoma; Her-2 oncogene; FISH; DNA ploidy; immunohistochemistry; prognostic factor ID synovial sarcoma; Her-2 oncogene; FISH; DNA ploidy; immunohistochemistry; prognostic factor AB The treatment options for synovial sarcoma (SS) are very limited, though this type of sarcoma seems to be more heterogeneous than it has been traditionally considered. The present study investigates the Her-2 oncogene status of 20 cases of SS, to determine whether Her-2 amplification can be considered as a prognostic factor. Her-2 oncogene amplification was determined on smears (frozen material was used from our tumor bank in each case), using fluorescence in situ hybridization technique (dual color FISH with centromeric probe for chromosome 17 and specific probe for Her-2 oncogene). Moreover, protein expression was assessed by immunohistochemistry, and DNA ploidy status was measured using image analysis. We had 5 biphasic and 15 monophasic SSs, patients’ age ranged from 13 to 68 years (mean, 39.8 years). Tumor size was larger than 5 cm in each case. Follow-up time ranged from 6 to 78 months (mean, 38.5 months). For statistical analysis the chi-square test was used. Her-2 oncogene amplification was found in three cases (15.0%) of 20 SSs. These cases proved to be 2+ positive by immunohistochemistry, but massive amplification, characteristic of a subset of breast carcinomas, was not observed. Her-2 oncogene amplification was significantly associated with a lower risk of developing metastasis (P<0.05) (none of the 3 amplified cases had metastases), while no association was found with recurrence. Six cases proved to be aneuploid and 14 were diploid, but no association was found between Her-2 amplification status and ploidy, and between ploidy status and metastasis or recurrence. Our results emphasize and confirm that Her-2 oncogene amplification is a rare event in SS, but the small subset of SS with Her-2 amplification has a better overall prognosis. Furthermore, this may open a theoretically new treatment possibility with Trastuzumab for Her-2-amplified cases of SS. C1 [Sapi, Zoltan] St John's Hospital, Department of Pathology, Diosarok ut 1., H-1125 Budapest, Hungary. [Papai, Zsuzsa] National Health Institute, Department of OncologyBudapest, Hungary. [Hruska, Anett] Semmelweis University, Institute of Morphology and PhysiologyBudapest, Hungary. [Antal, Imre] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Bodo, Miklos] St John's Hospital, Department of Pathology, Diosarok ut 1., H-1125 Budapest, Hungary. [Orosz, Zsolt] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. RP Sapi, Z (reprint author), St John's Hospital, Department of Pathology, H-1125 Budapest, Hungary. CR Bergh P, Meis-Kindblom JM, Gherlinzoni F, et al: Synovial sarcoma: identification of low and high risk groups. Cancer 85:2596-2607, 1999 Lewis JJ, Antonescu CR, Leung DHY, et al: Synovial sarcoma: a multivariate analysis of prognostic factors in 112 patients with primary localized tumors of the extremity. J Clin Oncol 18:2087-2094, 2000 Spillane AJ, Ahern R, Judson IR, et al: Synovial sarcoma: a clinicopathologic, staging and prognostic assessment. J Clin Oncol 18:3794-3803, 2000 Spurrell EL, Fisher C, Thomas JM, et al: Prognostic factors in advanced synovial sarcoma: an analysis of 104 patients treated at the Royal Marsden Hospital. Ann Oncol 16:437-444, 2005 Panagopoulos I, Mertens F, Isakesson M, et al: Clinical impact of molecular and cytogenetic findings in synovial sarcoma. Genes Chromosomes Cancer 31:362-372, 2001 Haroske G, Baak JPA, Danielsen H, et al: Fourth updated ESACP consensus report on diagnostic DNA image cytometry. Anal Cell Pathol 23:89-95, 2001 Kindermann D, Hilgers CH: Glare-correction in DNA image cytometry. Anal Cell Pathol 6:165-180, 1994 Barbashina V, Benevenia J, Aviv H, et al: Oncoproteins and proliferation markers in synovial sarcomas: a clinicopathologic study of 19 cases. J Cancer Res Clin Oncol 128:610-616, 2002 Allander SV, Illei PB, Chen Y, et al: Expression profiling of synovial sarcoma by cDNA microarrays: association of ERBB2, IGFBP2, and ELF3 with epithelial differentiation. Am J Pathol 161:1587-1595, 2002 Thomas DG, Giordano TJ, Sanders D, et al: Expression of receptor tyrosine kinases epidermal growth factor receptor and HER-2/neu in synovial sarcoma. Cancer 103:830-838, 2005 Nuciforo PG, Pellegrini C, Fasani R, et al: Molecular and immunohistochemical analysis of HER-2/neu oncogene in synovial sarcoma. Hum Pathol 34:639-645, 2003 Coombs LM, Pigott DA, Sweeney E, et al: Amplification and over-expression of c-erbB-2 in transitional cell carcinoma of the urinary bladder. Br J Cancer 63:601-608, 1991 Kay EW, Mulcaby H, Walsh CB, et al: Cytoplasmic c-erbB-2 protein expression correlates with survival in Dukes’ B colorectal carcinoma. Histopathology 25:455-461, 1996 Keshgegian AA, Cnaan A: erbB-2 oncoprotein expression in breast carcinoma. poor prognosis associated with high degree of cytoplasmic positivity using CB-11 antibody. Am J Clin Pathol 108:456-463, 1997 Roychowdhury DF, Tseng A Jr, Fu KK, et al: New prognostic factors in nasopharyngeal carcinoma. Tumor angiogenesis and c-erbB-2 expression. Cancer 77:1419-1426, 1994 Kilpatrick SE, Geisinger KR, King TS, et al: Clinicopathologic analysis of HER-2/neu immunoexpression among various histologic subtypes and grades of osteosarcoma. Mod Pathol 14:1277-1283, 2001 Sugg SL, Ezzat S, Zheng L, et al: Cytoplasmic staining of erbB- 2 but not mRNA levels correlates with differentiation in human thyroid neoplasia. Clin Endocrinol 49:629-637, 1998 El-Naggar AK, Ayala AG, Abdul-Karim FW, et al: Synovial sarcoma. A DNA flow cytometric study. Cancer 65:2295-2300, 1990 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 133 EP 138 PG 6 ER PT J AU Nicolson, LG AF Nicolson, L Garth TI Lipid Replacement/Antioxidant Therapy as an Adjunct Supplement to Reduce the Adverse Effects of Cancer Therapy and Restore Mitochondrial Function SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lipids; antioxidants; cancer therapy; dietary supplement; fatigue; mitochondria ID lipids; antioxidants; cancer therapy; dietary supplement; fatigue; mitochondria AB The most common complaints of cancer patients undergoing chemo- or radiotherapy are fatigue, nausea, vomiting, malaise, diarrhea and headaches. These adverse effects are thought to be due to damage of normal tissues during the course of therapy. In addition, recent evidence indicates that fatigue is related to reduced mitochondrial function through loss of efficiency in the electron transport chain caused by membrane oxidation, and this occurs during aging, in fatiguing illnesses and in cancer patients during cytotoxic therapy. Lipid Replacement Therapy administered as a nutritional supplement with antioxidants can prevent oxidative membrane damage to normal tissues, restore mitochondrial and other cellular membrane functions and reduce the adverse effects of cancer therapy. Recent clinical trials using patients with chronic fatigue have shown the benefit of Lipid Replacement Therapy plus antioxidants in restoring mitochondrial electron transport function and reducing moderate to severe chronic fatigue by protecting mitochondrial and other cellular membranes from oxidative and other damage. In cancer patients a placebo-controlled, cross-over clinical trial using Lipid Replacement Therapy plus antioxidants demonstrated that the adverse effects of chemotherapy can be reduced in 57-70% of patients. Dietary use of unoxidized membrane lipids plus antioxidants is recommended for patients undergoing cancer therapy to improve quality of life but should not be taken at the same time of day as the therapy. C1 [Nicolson, L Garth] The Institute for Molecular Medicine, Department of Molecular Pathology, 16371 Gothard St. H, CA 92647 Huntington Beach, California, USA. RP Nicolson, LG (reprint author), The Institute for Molecular Medicine, CA 92647 Huntington Beach, USA. EM gnicolson@immed.org CR Buckingham R, Fitt J, Sitzia J: Patients’ experience of chemotherapy: side-effects of carboplatin in the treatment of carcinoma of the ovary. Eur J Cancer Care 6: 59-71, 1997. Vogelzang N, Breitbart W, Cella D, et al: Patient caregiver and oncologist perceptions of cancer-related fatigue: results of a tripart assessment survey. Semin Hematol 34, Suppl 2): 4-12, 1997. Romanelli A, Bozzone A, Magrone G, et al: Cancer-related fatigue: evaluation and treatment. Rays 29: 453-455, 2004. Bender CM, Ergyn FS, Rosenzweig MQ, et al: Symptom clusters in breast cancer across 3 phases of the disease. Cancer Nurs 28: 219-225, 2005. Ahlberg K, Ekman T, Gaston-Johansson F: Fatigue, psychological distress, coping resources, and functional status during radiotherapy for uterine cancer. Oncol Nurs Forum 32: 633- 640, 2005. Shafqat A, Einhorn LH, Hanna N, et al: Screening studies for fatigue and laboratory correlates in cancer patients undergoing treatment, dagger). Ann Oncol 16:1545-1550, 2005. Liu L, Marler MR, Parker BA, et al: The relationship between fatigue and light exposure during chemotherapy. Support Care Cancer in press, 2005. Von Roenn JH, Paice JA: Control of common, non-pain cancer symptoms. Semin Oncol 32: 200-210, 2005. Kroenke K, Wood DR, Mangelsdorff AD, et al: Chronic fatigue in primary care. Prevalence, patient characteristics, and outcome. JAMA 260: 929-934, 1988. Morrison JD: Fatigue as a presenting complaint in family practice. J Family Pract 10: 795-801, 1980. McDonald E, David AS, Pelosi AJ, Mann AH: Chronic fatigue in primary care attendees. Psychol Med 23:987-998, 1993. Piper BF, Linsey AM, Dodd MJ: Fatigue mechanism in cancer. Oncol Nursing Forum 14: 17-23, 1987. Piper BF, Dribble SL, Dodd MJ, et al: The revised Piper Fatigue Scale: psychometric evaluation in women with breast cancer. Oncol Nursing Forum 25: 667-684, 1998. Nicolson GL: Lipid replacement as an adjunct to therapy for chronic fatigue, anti-aging and restoration of mitochondrial function. J Am Nutraceut Assoc 6: 22-28, 2003. Huang H, Manton KG: The role of oxidative damage in mitochondria during aging: a review. Front Biosci 9: 1100-1117, 2004. Richter C, Par JW, Ames B: Normal oxidative damage to mitochondrial and nuclear DNA is extensive. Proc Natl Acad Sci USA 85: 6465-6467, 1998. Wei YH, Lee HC: Oxidative stress, mitochondrial DNA mutation and impairment of antioxidant enzymes in aging. Exp Biol Med 227: 671-682, 2002. Harman D: Aging: A theory based on free radical and radiation chemistry. J Gerontol 2: 298-300, 1956. Halliwell B: Role of free radicals in the neurodegenerative diseases: therapeutic implications for antioxidant treatment. Drugs Aging 18: 685-716, 2001. Tan, NSS, Vinckenbosch NS, Liu N, et al: Selective cooperation between fatty acid binding proteins and peroxisome proliferator- activated receptors in regulating transcription. Mol Cell Biol 22: 5114-5127, 2002. Chen D, Cao G, Hastings T, et al: Age-dependent decline of DNA repair activity for oxidative lesions in rat brain mitochondria. J Neurochem 81: 1273-1284, 2002. Xu D, Finkel T: A role for mitochondria as potential regulators of cellular life span. Biochem Biophys Res Commun 294: 245- 248, 2002. Logan AC, Wong C: Chronic fatigue syndrome: oxidative stress and dietary modifications. Altern Med Rev 6: 450-459, 2001. Manuel y Keenoy B, Moorkens G, Vertommen J, et al: Antioxidant status and lipoprotein peroxidation in chronic fatigue syndrome. Life Sci 68: 2037-2049, 2001. Richards RS, Roberts TK, McGregor NR, et al: Blood parameters indicative of oxidative stress are associated with symptom expression in chronic fatigue syndrome. Redox Rep 5: 35-41, 2000. Felle S, Mecocci P, Fano G, et al: Specific oxidative alterations in vastus lateralis muscle of patients with the diagnosis of chronic fatigue syndrome. Free Radical Biol Med 29: 1252- 1259, 2000. Pall ML: Elevated, sustained peroxynitrite levels as the cause of chronic fatigue syndrome. Med Hypotheses 54: 115-125, 2000. Castro L, Rodriguez M, Radi R: Aconitase is readily inactivated by peroxynitrite, but not by its precursor, nitric oxide. J Biol Chem 269: 29409-29415, 1994. Radi R, Rodriguez M, Castro L, Telleri R: Inhibition of mitochondrial electronic transport by peroxynitrite. Arch Biochem Biophys 308: 89-95, 1994. Manuel y Keenoy B, Moorkens G, Vertommen J, et al: Magnesium status and parameters of the oxidant-antioxidant balance in patients with chronic fatigue: effects of supplementation with magnesium. J Am Coll Nutr 19: 374-382, 2000. Kanno T, Sato EE, Muranaka S, et al: Oxidative stress underlies the mechanism for Ca(2+)-induced permeability transition of mitochondria. Free Radical Res 38: 27-35, 2004. Nicolson GL, Poste G, Ji T: Dynamic aspects of cell membrane organization. Cell Surface Rev 3: 1-73, 1977. Subczynski WK, Wisniewska A: Physical properties of lipid bilayer membranes: relevance to membrane biological functions. Acta Biochim Pol 47: 613-625, 2000. Ellithorpe RR, Settineri R, Nicolson GL: Reduction of fatigue by use of a dietary supplement containing glycophospholipids. J Am Nutraceut Assoc 6: 23-28, 2003. Agadjanyan M, Vasilevko V, Ghochikyan A, et al: Nutritional supplement, NTFactor, restores mitochondrial function and reduces moderately severe fatigue in aged subjects. J Chronic Fatigue Syndr 11: 23-26, 2003. Hajri T, Abumrad NA: Fatty acid transport across membranes: relevance to nutrition and metabolic pathology. Annu Rev Nutr 22: 383-415, 2002. Hamilton JA: Fatty acid transport: difficult or easy? J Lipid Res 39: 467-481, 1998. Fellmann P, Herve P, Pomorski T, et al: Transmembrane movement of diether phospholipids in human erythrocytes and human fibroblasts. Biochemistry 39: 4994-5003, 2000. Conner SD, Schmid SL: Regulated portals of entry into the cell. Nature 422: 37-44, 2003. Mansbach CM, Dowell R: Effect of increasing lipid loads on the ability of the endoplasmic reticulum to transport lipid to the Golgi. J Lipid Res 41: 605-612, 2000. De AK, Darad R: Age-associated changes in antioxidants and antioxidative enzymes in rats. Mech Ageing Dev 59:123-128, 1991. Ames BM: Micronutrients prevent cancer and delay aging. Toxicol Lett 102: 1035-1038, 1998. Sharman EH, Bondy SC: Effects of age and dietary antioxidants on cerebral electron transport chain activity. Neurobiol Aging 22: 629-634, 2001. Sugiyama S, Yamada K, Ozawa T: Preservation of mitochondrial respiratory function by coenzyme Q10 in aged rat skeletal muscle. Biochem Mol Biol Int 37: 1111-1120, 1995. Lin MT, Simon DK, Ahn CH, et al: High aggregate burden of somatic mtDNA point mutations in aging and Alzheimer’s disease brain. Hum Mol Genet 11: 133-145, 2002. Seidman M, Khan MJ, Tang WX, Quirk WS: Influence of lecithin on mitochondrial DNA and age-related hearing loss. Otolaryngol Head Neck Surg 127: 138-144, 2002. Miquel J: Can antioxidant diet supplementation protect against age-related mitochondrial damage? Ann NY Acad Sci 959: 317-347, 2002. Nicolson GL, Ellithorpe R: Lipid replacement and antioxidant nutritional therapy for restoring mitochondrial function and reducing fatigue in chronic fatigue syndrome and other fatiguing illnesses. J Chronic Fatigue Syndr, in press, 2005. Colodny L, Lynch K, Farber C, et al: Results of a study to evaluate the use of Propax to reduce adverse effects of chemotherapy. J Am Nutraceut Assoc 2: 17-25, 2000. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 139 EP 144 PG 6 ER PT J AU Elek, G Gyokeres, T Schafer, E Burai, M Pinter, F Pap, AF Elek, Gabor Gyokeres, Tibor Schafer, Eszter Burai, Maria Pinter, Ferenc Pap, Akos TI Early Diagnosis of Pancreatobiliary Duct Malignancies by Brush Cytology and Biopsy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE intraductal biopsy; intraductal cytology; pancreatobiliary strictures; sensitivity and specificity; sampling-; processing- and interpretive errors ID intraductal biopsy; intraductal cytology; pancreatobiliary strictures; sensitivity and specificity; sampling-; processing- and interpretive errors AB Two hundred and five preoperative intraductal samplings (brushing and biopsy) were evaluated from 113 patients with biliary or Wirsung duct strictures. One hundred and three strictures could be specified by histology of the operative specimens, autopsy, or by the patients’ clinical course. Preoperative diagnostic efficacy depended on the tumor location (it was the best for ampullary and parapapillary tumors), but the average quantitative indices for sensitivity, absolute sensitivity, specificity, positive and negative predictive values, diagnostic accuracy of cytology were 53%, 20%, 100%, 100%, 25%, 59%, respectively. The same values for biopsy were 43%, 34%, 100%, 100%, 36% and 56%. These figures improved after simultaneous cytology and biopsy. Close cooperation with the endoscopist was necessary in cases of negative, inconclusive and dysplastic (27%) samples. Repetition of sampling improved the results by 8%. Among the 26 preoperative false negative cases, sampling-, technical- and interpretative errors occurred in 84%, 4% and 12%, respectively. Revision of samples revealed 4 malignant cases among the false negative cytologic brushings. Reclassification of specimens considering the latest criteria - primary and secondary malignant features, pancreatic intraepithelial neoplasia (PanINs), etc. - resulted in improvement of the diagnostic efficiency. C1 [Elek, Gabor] Central Railway Hospital and Polyclinic, Department of PathologyBudapest, Hungary. [Gyokeres, Tibor] Central Railway Hospital and Polyclinic, Department of Gastroenterology, Podmaniczky u. 111., H-1062 Budapest, Hungary. [Schafer, Eszter] Central Railway Hospital and Polyclinic, Department of Gastroenterology, Podmaniczky u. 111., H-1062 Budapest, Hungary. [Burai, Maria] Central Railway Hospital and Polyclinic, Department of Gastroenterology, Podmaniczky u. 111., H-1062 Budapest, Hungary. [Pinter, Ferenc] Central Railway Hospital and Polyclinic, Department of Gastroenterology, Podmaniczky u. 111., H-1062 Budapest, Hungary. [Pap, Akos] Central Railway Hospital and Polyclinic, Department of Gastroenterology, Podmaniczky u. 111., H-1062 Budapest, Hungary. RP Pap, (reprint author), Central Railway Hospital and Polyclinic, Department of Gastroenterology, H-1062 Budapest, Hungary. EM papakos@kkk.sote.hu CR Adsay V, Logani S, Sarkar F, et al: Foamy gland pattern of pancreatic ductal adenocarcinoma. Am J Surg Pathol 24: 493-504, 2000. Albores-Saavedra J, Henson DE, Klimstra DS: Tumors of the gallbladder, extrahepatic bile ducts and ampulla of Vater. In: Atlas of Tumor Pathology. Third series, Fascicle 27, Armed Forces Inst. Washington, 2000, pp 44-53, 147-150, 177, 191- 194, 250-256. Al-Kaisi N, Siegler EE: Fine needle aspiration cytology of the pancreas. Acta Cytol 33: 145-152, 1989. Andea A, Sarkar F, Adsay VN: Clinicopathological correlates of pancreatic intraepithel neoplasia. Mod Pathol 16: 996-1006, 2003. Bajtai A, Nemesanszky E: The future of the pathology – from the side of clinical pathology. Orv Hetil 143: 867-873, 2002. Bardales RH, Stanley MW, Simpson DD, et al: Diagnostic value of brush cytology in the diagnosis of duodenal, biliary and ampullary neoplasms. Am J Clin Pathol 109: 540-548, 1998. Chang KJ, Nguyen P, Erickson RA, et al: Clinical utility of endoscopic ultrasound guided fine-needle aspiration in the diagnosis and staging of pancreatic carcinoma. Gastrointest Endosc 45: 387-393, 1997. Cohen MB, Wittchow RJ, Johlin FC, et al: Brush cytology of the extrahepatic biliary tract: comparison of cytologic features of adenocarcinoma and benign biliary strictures. Mod Pathol 8: 498-502, 1995. de Peralta-Venturina MN, Wong DK, Purslow MJ, et al: Biliary tract cytology in specimens obtained by direct cholangiographic procedures. Diagn Cytopathol 14: 334-348, 1996. Domagk D, Poremba C, Dietl KH, et al: Endoscopic transpapillary biopsies and intraductal ultrasonography in the diagnostics of bile duct strictures. Gut 51: 240-244, 2002. Earnhardt RC, McQuone SJ, Minasi JS, et al: Intraoperative fine needle aspiration of pancreatic and extrahepatic biliary masses. Surg Gynecol Obstet 177: 147-152, 1993. Elek G, Gyori S, Toth B, et al: Histological evaluation of preoperative biopsies from ampulla Vateri. Pathol Oncol Res 9: 32-41, 2003. Glasbrenner B, Ardan M, Boeck W, et al: Prospective evaluation of brush cytology of biliary strictures during endoscopic retrograde cholangiopancreatography. Endoscopy 31: 712-717, 1999. Gyokeres T, Schafer E, Gelley A, et al: The sensitivity of the intraductal cytology and biopsy in pancreatobiliary malignancies. Endoscopy 33, Suppl I): 1833-A, 2001. Henke AC, Jensen CS, Cohen MB: Cytologic diagnosis of adenocarcinoma in biliary and pancreatic duct brushings. Adv Anat Pathol 9: 301-308, 2002. Hruban RH, Adsay NV, Albores-Saavedra J, et al: Pancreatic intraepithelial neoplasia: a new nomenclature and classification system for pancreatic duct lesions. Am J Surg Pathol 25: 579- 586, 2001. Jailwala J, Fogel EL, Sherman S, et al: Triple tissue sampling at ERCP in malignant biliary obstruction. Gastrointest Endosc 51: 383-390, 2000. Kocjan G, Smith AN: Bile duct brushings cytology: potential pitfalls in diagnosis. Diagn Cytopathol 16: 358-363, 1997. Kovacs F, Gyokeres T, Elek G, et al: Sphincter of Oddi dysfunction – prolonged medical treatment or early endoscopic sphincter ablation. Orv Hetil 143: 2829-2834, 2002. Kubota Y, Takaoka M, Tani K, et al: Endoscopic transpapillary biopsy for diagnosis of patients with pancreaticobiliary ductal strictures. Am J Gastroenterol 88: 1700-1704, 1993. Kurzawinski T, Deery A, Davidson BR: Diagnostic value of cytology for biliary stricture. Br J Surg 80: 414-421, 1993. Layfield LJ, Wax TD, Lee JG, et al: Accuracy and morphologic aspects of pancreatic and biliary duct brushings. Acta Cytol 39: 11-18, 1995. Lee JG, Leung JW, Baillie J, et al: Benign, dysplastic, or malignant – making sense of endoscopic brush cytology. Am J Gastroenterol 90: 722-726, 1995. Logrono R, Kurtycz DF, Molina CP, et al: Analysis of falsenegative diagnoses on endoscopic brush cytology of biliary and pancreatic duct strictures. Arch Pathol Lab Med 124: 387- 392, 2000. Mansfield JC, Griffin SM, Wadehra V, et al: A prospective evaluation of cytology from biliary strictures. Gut 40: 671- 677, 1997. McGuire DE, Venu RP, Brown RD, et al: Brush cytology for pancreatic carcinoma: an analysis of factors influencing results. Gastrointest Endosc 44: 300-304, 1996. Merchant NB, Conlon KC, Saigo P, et al: Positive peritoneal cytology predicts unresectability of pancreatic adenocarcinoma. J Am Coll Surg 188: 421-426, 1999. Okonkwo AM, De Frias DVS, Gunn R, et al: Reclassification of „atypical” diagnoses in endoscopic retrograde cholangiopancreatography. Acta Cytol 47: 435-442, 2003. Osnes M, Serck-Hanssen A, Kristensen O, et al: Endoscopic retrograde brush cytology in patients with primary and secondary malignancies of the pancreas. Gut 20: 279-284, 1979. Ponchon T, Gagnon P, Berger F, et al: Value of endobiliary brush cytology and biopsies for the diagnosis of malignant bile duct stenosis: results of a prospective study. Gastrointest Endosc 42: 565-572, 1995. Ponsioen CY, Vrouenraets SME, van Milligen de Wit AWM, et al: Value of brush cytology for dominant strictures in primary sclerosing cholangitis. Endoscopy 31: 305-309, 1999. Pugliese V, Conio M, Nicolo G, et al: Endoscopic retrograde forceps biopsy and brush cytology of biliary strictures: a prospective study. Gastrointest Endosc 42: 520-526, 1995. Renshaw AA, Madge R, Jiroutek M, et al: Bile duct brushing cytology: statistical analysis of proposed diagnostic criteria. Am J Clin Pathol 110: 635-640, 1998. Solcia E, Capell C, Kloppel G: Tumors of the pancreas. In: Atlas of Tumor Pathology. Third series, Fascicle 27, Armed Forces Inst. Washington, 2000, pp 53-64, 253. Stewart CJR, Mills PR, Carter R, et al: Brush cytology in the assessment of pancreatico-biliary strictures: a review of 406 cases. J Clin Pathol 54: 449-455, 2001. Stewart CJR, Burke GM: Value of p53 immunostaining in pancreatico- biliary brush cytology specimens. Diagn Cytopathol 23: 308-313, 2000. Stewart CJR, Stephen MR, Ferrier RK: Hepatocellular carcinoma diagnosis in bile duct brush cytology. Diagn Cytopathol 19: 149-150, 1998. Stewart CJR, Carter R, Imrie CW, et al: Brush cytology of intraduct papillary mucinous neoplasm of the pancreas. Cytopathology 8: 343-348, 1997. Sturm PDJ, Rauws EAJ, Hruban RH, et al: Clinical value of Kras codon 12 analysis and endobiliary brush cytology for the diagnosis of malignant extrahepatic bile duct stenosis. Clin Cancer Res 5: 629-635, 1999. Sugiyama M, Atomi Y, Wada N, et al: Endoscopic transpapillary bile duct biopsy without sphincterotomy for diagnosing biliary strictures: a prospective comparative study with bile and brush cytology. Am J Gastroenterol 91: 465-467, 1996. Tamada K, Kurihara K, Tomiyama T, et al: How many biopsies should be performed during percutaneous transhepatic cholangioscopy to diagnose biliary tract cancer? Gastrointest Endosc 50: 653-658, 1999. Tascilar M, Sturm PDJ, Caspers E, et al: Diagnostic p53 immunostaining of endobiliary brush cytology. Cancer 87: 306-311, 1999. van Es JM, Polak MM, van den Berg FM, et al: Molecular markers for diagnostic cytology of neoplasms in the head region of the pancreas: mutation of K-ras and overexpression of the p53 protein product. J Clin Pathol 48: 218-222, 1995. Vandervoort J, Soetikno RM, Montes H, et al: Accuracy and complication rate of brush cytology from bile duct versus pancreatic duct. Gastrointest Endosc 49: 322-327, 1999. Vellet D, Leiman G, Mair S, et al: Fine needle aspiration cytology of mucinous cystadenocarcinoma of the pancreas. Acta Cytol 32: 43-48, 1988. Winternitz T, Jarai B, Szekely E, et al: The role of the preoperative cytology at patients with pancreatic head mass. Z Gastroenterol 41: 146-A, 1993. Ylagan LR, Liu LH, Maluf HM: Endoscopic bile duct brushing of malignant pancreatic biliary strictures: retrospective study with comparison of conventional smear and ThinPrep® techniques. Diagn Cytopathol 28: 196-204, 2003. Zalatnai A: Pancreatic cancer – a continuing challenge in oncology. Pathol Oncol Res 9: 252-263, 2003. Zabo A, Barocsai G, Bodo M, et al: About the bile cytology in connection with three cases. Magyar Sebeszet 51: 207-210, 1998., In Hungarian only) Zen Y, Harada K, Sasaki M, et al: IgG4-related sclerosing cholangitis with and without hepatic inflammatory pseudotumor and sclerosing pancreatitis associated sclerosing cholangitis. Am J Surg Pathol 28: 1193-1203, 2004. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 145 EP 155 PG 11 ER PT J AU Behjati, F Atri, M Najmabadi, H Nouri, K Zamani, M Mehdipour, P AF Behjati, Farkhondeh Atri, Morteza Najmabadi, Hossein Nouri, Keramat Zamani, Mahdi Mehdipour, Parvin TI Prognostic Value of Chromosome 1 and 8 Copy Number in Invasive Ductal Breast Carcinoma among Iranian Women: An Interphase FISH Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; Iranian women; interphase FISH; clinicopathological parameters; chromosome 1 and 8 aneusomy; prognosis ID breast cancer; Iranian women; interphase FISH; clinicopathological parameters; chromosome 1 and 8 aneusomy; prognosis AB Breast cancer is amongst the leading causes of death in women worldwide and the most common cancer amongst Iranian women. Unfortunately, the current clinical and histological criteria can only help 60 percent of women with breast cancer in diagnosis and long-term treatment. Therefore, genetic markers both at single gene and chromosomal level can play an important role in improving the diagnosis and prognosis of breast cancer patients. The aim of this retrospective study was to investigate the role of chromosome 1 and 8 copy number assessed by interphase fluorescence in situ hybridization (FISH), as prognostic parameters in 50 Iranian women, aged 35 to 64 years, with sporadic invasive ductal breast carcinoma. Chromosome 1 and 8 copy numbers were evaluated in relation to established clinicopathological parameters, the immunohistochemical markers ER, PR, P53 and cathepsin D, DNA index by flow cytometry, age and survival status of the patients. FISH using centromeric probes for chromosomes 1 and 8 was applied to interphase cell suspensions prepared from archived, Carnoyfixed tumor cells and selected paraffin-embedded tumor sections. Aneusomy for chromosomes 1 and 8 was present in all 50 patients to different levels. The total abnormality rate for chromosome 1 was 33.92 percent (4.24 percent monosomy and 29.68 percent polysomy), whereas for chromosome 8 this rate was 28.30 percent (6.48 percent monosomy and 21.82 percent polysomy). Statistically significant association (p<0.05) was demonstrated between monosomy 1 and patients’ age below 50 years, and between monosomy 1 and poor survival, respectively. Disomy 8 was significantly associated with P53 expression. A borderline significant correlation was demonstrated between polysomy 8 and diploid DNA content, as well as between disomy 1 and disease-free status of the patients. Chromosome 1 and 8 copy numbers may be considered as useful prognostic markers in invasive ductal carcinoma of the breast. C1 [Behjati, Farkhondeh] Tehran University of Medical Sciences, Faculty of Medicine, Department of Medical GeneticsTehran, Iran. [Atri, Morteza] Cancer Institute, Faculty of Medicine, Tehran University of Medical Sciences, Department of SurgeryTehran, Iran. [Najmabadi, Hossein] University of Social Welfare and Rehabilitation Sciences, Genetics Research CenterTehran, Iran. [Nouri, Keramat] School of Public Health, Tehran University of Medical Sciences, Department of Epidemiology and BiostatisticsTehran, Iran. [Zamani, Mahdi] Tehran University of Medical Sciences, Faculty of Medicine, Department of Medical GeneticsTehran, Iran. [Mehdipour, Parvin] Tehran University of Medical Sciences, Faculty of Medicine, Department of Medical GeneticsTehran, Iran. RP Mehdipour, P (reprint author), Tehran University of Medical Sciences, Faculty of Medicine, Department of Medical Genetics, Tehran, Iran. EM mehdipor@sina.tums.ac.ir CR Adeyinka A, Mertens F, Idvall I, et al: Multiple polysomies in breast carcinoma: preferential gain of chromosomes 1, 5, 6, 7, 12, 16, 17, 18, and 19. Cancer Genet Cytogenet 111: 144-148, 1999. Atri M, Jafarimojarrad E, Javidroozi M, Mehdipour P: Lack of association between early onset of breast cancer and numbers of affected relatives in an Iranian population. Fam Cancer 2: 117-118, 2003. Bertucci F, Nasser V, Granjeaud S, Houlgatte R: Gene expression profiles of poor prognosis primary breast cancer correlate with survival. Hum Mol Genet 11: 863-872, 2002. Botti C, Pescatore B, Mottolese M, et al: Incidence of chromosomes 1 and 17 aneusomy in breast cancer and adjacent tissue with interphase cytogenetic study. J Am Coll Surg 190: 530- 539, 2000. Dies PJV, Baak JPA, Cok PM, et al: Reproducibility of mitosis counting in 2,469 breast cancer specimens. Hum Pathol 23: 603-607, 1992. Dominguez D, Silva J, Silve JM, et al: Clinicopathological characteristics of breast carcinoma with allelic loss in the p73 region. Breast Cancer Res Treat 63: 17-22, 2000. Elston CW: Grading of invasive carcinoma of the breast. In: Diagnostic Histopathology of the Breast., Eds: Page DL, Anderson TJ), Churchill-Livingstone, Edinburgh, 1987, pp 300-311. Fehm T, Morrison L, Saboorian H, et al: Patterns of aneusomy for three chromosomes in individual cells from breast cancer tumors. Breast Cancer Res Treat 75: 227-239, 2002. Ferti AD, Stamouli MJ, Panani AD, et al: Molecular cytogenetic analysis of breast cancer: a combined multicolour fluorescence in situ hybridization and G-banding study of uncultured tumor cells. Cancer Genet Cytogenet 149: 28-37, 2004. Fitzgibbons PL, Page DL, Weaver D: Prognostic factors in breast cancer, College of American Pathologists Consensus Statement 1999. Arch Pathol Lab Med 124: 966-978, 2000. Harirchi I, Karbakhsh M, Kashefi A, Momtahen AJ: Breast cancer in Iran: results of a multi-center study. Asian Pac J Cancer Prev 5: 24-27, 2004. Harrison MM, Magee HM, O’loughlin JF: Comparison of chromosome 1 aneusomy detected by interphase cytogenetics and DNA ploidy in carcinoma of the breast. Histopathology 3: 221-226, 1997. Haskell CM: Cancer Treatment, 5th edition. Saunders Company, Philadelphia, 2001, pp 7-25. Hopman AHN, Remaekers FCS, Raap AK, et al: In situ hybridization as a tool to study numeric chromosomal aberrations in solid bladder tumors. Histochemistry 89: 307-316, 1988. Hsu SM, Raine L, Fanger H: Use of avidin-biotin peroxidase complex, ABC, in immunoperoxidase technique: a comparison between ABC and unlabelled antibody, PAP, procedures. J Histochem Cytochem 29: 577-580, 1981. Hudelist G, Kostler W, Czervenka K, et al: Predicting the clinical course of breast cancer patients undergoing trastuzumabbased therapy: an outlook. Methods Find Exp Clin Pharmacol, 26: 201-210, 2004. Leger I, Thomas M, Ronot X, Brugal G: Detection of chromosome 1 aberration by fluorescent in situ hybridization, FISH, in the human breast cancer cell line MCF-7. Anal Cell Pathol 5: 299-309, 1993. Jain AN, Chin K, Borresen- Dale AL, et al: Quantitative analysis of chromosomal CGH in human breast tumors associates copy number abnormalities with P53 status and patient survival. Proc Natl Acad Sci 98: 7952-7957, 2001. Kahan E, Ibrahim AS, EI Najjar K, et al: Cancer patterns in the Middle East – Special report from the Middle East Cancer Society. Acta Oncol 36: 631-636, 1997. Lalloo F: Genetics for Oncologists. Remedica, London, 2002. Levine AJ: The tumor suppressor genes. Ann Rev Biochem 62: 623-651, 1993. Marinho AF, Botelho M, Schmitt FC: Evaluation of numerical abnormalities of chromosome 1 and 17 in proliferative epithelial breast lesions using fluorescence in situ hybridization. Pathol Res Pract 196: 227-233, 2000. Mark HF, Taylor W, Afify A, et al: Stage I and II infiltrating ductal carcinoma of breast analysed for chromosome 8 copy number using fluorescent in situ hybridization. Pathobiology 65: 184-189, 1997. Massoner A, Augustin F, Duba HC, et al: FISH cytogenetics and prognosis in breast and non-small cell lung cancers. Cytometry 62B: 52-56, 2004. Mehdipour P, Atri M, Jafarimojarrad E, et al: Laddering through pedigrees: family history of malignancies in primary breast cancer patients. Asian Pac J Cancer Prev 4: 185-192, 2003. Montazeri A, Ebrahimi M, Mehrdad N, et al. Delayed prevention in breast cancer: A study in Iranian women. BMC Womens Health, 3: 4, 2003. Nakopoulou L, Giannopoulou I, Trafalis D, et al: Evaluation of numeric alterations of chromosomes 1 and 17 by in situ hybridization in invasive breast carcinoma with clinicopathologic parameters. Appl Immunohistochem Mol Morphol 10: 20-28, 2002. Nakopoulou L, Tsirmpa I, Giannopoulou I, et al: Aneuploidy of chromosome 20 in invasive breast cancer correlates with poor outcome. Cancer Genet Cytogenet 134: 127-132, 2002. Persons DL, Hartmann LC, Herath JF, et al. Fluorescence in situ hybridization analysis of trisomy 12 in ovarian tumors. Am J Clin Pathol 102: 775-779, 1994. Persons DL, Robinson RA, Hsu P, et al. Chromosome specific aneusomy in carcinoma of the breast. Clin Cancer Res 2: 883- 888, 1996. Roka S, Fiegl M, Zojer N, et al. Aneuploidy of chromosome 8 as detected by interphase fluorescence in situ hybridization is a recurrent finding in primary and metastatic breast cancer. Breast Cancer Res Treat 48: 125-133, 1998. Santora LM, Mahoney MC, Lawvere S, et al. Breast cancer screening beliefs by practice location. BMC Public Health 3: 9, 2003. Sigurdsson S, Bodvarsdottir SK, Eyfjord J: P53 abnormality and chromosomal instability in the same breast tumor cells. Cancer Genet Cytogenet 12: 150-155, 2000. Summary of Report on Cancer Incidence in Iran. Cancer and Genetics Administration, Non-Communicable Diseases Sector of Iranian Center for Prevention and Control of Diseases. Deputy of Health, Ministry of Health, Treatment and Education, Islamic Republic of Iran, 2000. Takami S, Kawasome C, Kinoshita M, et al: Chromosomal instability detected by fluorescence in situ hybridization in Japanese cancer patients. Clin Chim Acta 308: 127-131, 2001. Truong K, Vielh P, Guilly MN, et al. Quantitative FISH analysis on interphase nuclei may improve diagnosis of DNA diploid breast cancers. Diagn Cytopathol 26: 213-216, 2002. Union International Contre Cancer. TNM classification of International Union Against Cancer., Eds: Harmanek P, Sobin LH). TNM Atlas, Berlin, Springer-Verlag, 1992. World Health Organization. Histologic typing of breast tumors. In: International Histologic Classification of Tumors., Eds: Hartman WH, Urello L, Sobin LH, Stalberg H), Geneva, WHO, 1981, pp 15-25. Yamamoto D, Senzaki H, Nakagawa H, et al. Detection of chromosomal aneusomy by fluorescence in situ hybridization for patients with nipple discharge. Cancer 97: 690-694, 2003. Fan Y-S: Molecular cytogenetics in medicine. In: Methods in Molecular Biology. Vol 204, Molecular Cytogenetics: Protocols and Applications, Humana Press, Totowa, NJ, 2002. Yoshihito U, Mitsuru E, Masataka Y: Allelic loss at 1p34-36 predicts poor prognosis in node-negative breast cancer. Clin Cancer Res 6: 3193-3198, 2000. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 157 EP 163 PG 7 ER PT J AU Zalatnai, A AF Zalatnai, Attila TI P-glycoprotein Expression is Induced in Human Pancreatic Cancer Xenografts During Treatment with a Cell Cycle Regulator, Mimosine SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE P-glycoprotein; pancreatic cancer; xenografts; human tumors; mimosine ID P-glycoprotein; pancreatic cancer; xenografts; human tumors; mimosine AB Application of several cell cycle checkpoint regulators seem to be promising in various experimental models including pancreatic cancer, and they are being evaluated in Phase I-II clinical trials. Among these compounds, mimosine, a plant-derived amino acid has shown an antineoplastic effect on human lung or pancreatic cancer xenografts in addition to cell cycle arrest in the late G1 phase. In the present study, immunosuppressed CBA mice bearing subcutaneously growing human ductal pancreatic adenocarcinomas were treated with 30 mg/kg L-mimosine for 34 days. The treatment resulted in retardation of tumor growth, accompanied by a significantly diminished proliferative activity (22.6% ± 1.7% Ki-67 positivity vs. 29.9% ± 1.1% in controls, mean ± SEM, P < 0.007) and an increased apoptotic rate (14.5 ± 1.1 apoptotic cells/mm2 vs. 3.8 ± 0.4/mm2 in the controls, P < 0.0001). The immunohistochemical expression of the multidrug resistance gene (MDR1)-encoded Pglycoprotein (p170) was studied. The parental and the untreated tumors did not express p170 protein, but in the mimosine-treated samples 30 to 60% of the carcinoma cells displayed a linear, membranebound positivity. The results indicate that P-glycoprotein is inducible by a cell cycle regulator, creating an acquired resistant phenotype. C1 [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Zalatnai, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM zalatnai@korb1.sote.hu CR Abe S, Kubota T, Otani Y, et al: UCN-01, 7-hydroxystaurosporine, inhibits in vivo growth of human cancer cells through selective perturbation of G1 phase checkpoint machinery. Jpn J Cancer Res 92: 537-545, 2001. Abolhoda A, Wilson AE, Ross H, et al: Rapid activation of MDR1 gene expression in human metastatic sarcoma after in vivo exposure to doxorubicin. Clin Cancer Res 5: 3352-3356, 1999. Bocsi J, Zalatnai A: Establishment and long term xenografting of human pancreatic carcinomas in immunosuppressed mice: Changes and stability in morphology, DNA ploidy and proliferative activity. J Cancer Res Clin Oncol 125: 9-19, 1999. Buolamwini JK: Cell cycle molecular targets in novel anticancer drug discovery. Curr Pharm Des 6: 379-392, 2000. Chang HC, Lee TH, Chuang LY, et al: Inhibitory effect of mimosine on proliferation of human lung cancer cells is mediated by multiple mechanisms. Cancer Lett 145: 1-8, 1999. Chang HC, Weng CF, Yen MH, et al: Modulation of cell cycle regulatory protein expression and suppression of tumor growth by mimosine in nude mice. Int J Oncol 17: 659-665, 2000. Chaudhary PM, Roninson IB: Induction of multidrug resistance in human cells by transient exposure to different chemotherapeutic drugs. J Natl Cancer Inst 85: 632-639, 1993. Chung HC, Rha SY, Kim JH, et al: P-glycoprotein: the intermediate end point of drug response to induction chemotherapy in locally advanced breast cancer. Breast Cancer Res Treat 42: 65-72, 1997. Fardel O, Lecureur V, Daval S, et al: Up-regulation of P-glycoprotein expression in rat liver cells by acute doxorubicin treatment. Eur J Biochem 246: 186-192, 1997. Feldman ST, Schonthal A: Negative regulation of histone H1 kinase expression by mimosine, a plant amino acid. Cancer Res 54: 494-498, 1994. Goldstein LJ, Galski H, Fojo A, et al: Expression of a multidrug resistance gene in human cancers. J Natl Cancer Inst 81: 116-124, 1989. Granzotto M, Drigo I, Candussio L, et al: Rifampicin and verapamil induce the expression of P-glycoprotein in vivo in Ehrlich ascites tumor cells. Cancer Lett 205: 107-115, 2004. Jimenez RE, Zalupski MM, Frank JJ, et al: Multidrug resistance phenotype in high grade soft tissue sarcoma: correlation of P-glycoprotein immunohistochemistry with pathologic response to chemotherapy. Cancer 86: 976-981, 1999. Krude T: Mimosine arrests proliferating human cells before onset of DNA replication in a dose-dependent manner. Exp Cell Res 247: 148-159, 1999. Lage H, Dietel M: Multiple mechanisms confer different drugresistant phenotypes in pancreatic carcinoma cells. J Cancer Res Clin Oncol 128: 349-357, 2002. Leonard GD, Fojo T, Bates SE: The role of ABC transporters in clinical practice. Oncologist 8: 411-424, 2003. Licht T, Fiebig HH, Bross KJ, et al: Induction of multiple-drug resistance during anti-neoplastic chemotherapy in vitro. Int J Cancer 49: 630-637, 1991. Lu Z, Kleeff J, Shrikhande S, et al: Expression of the multidrugresistance 1, MDR1, gene and prognosis in human pancreatic cancer. Pancreas 21: 240-247, 2000. Mealey KL, Barhoumi R, Burghardt RC, et al: Doxycycline induces expression of P glycoprotein in MCF-7 breast carcinoma cells. Antimicrob Agents Chemother 46: 755-761, 2002. Monden N, Abe S, Hishikawa Y, et al: The role of P-glycoprotein in human gastric cancer xenografts in response to chemotherapy. Int J Surg Invest 1: 3-10, 1999. Nawrocki ST, Sweeney-Gotsch B, Takamori R, McConkey DJ: The proteasome inhibitor bortezomib enhances the activity of docetaxel in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther 3: 59-70, 2004. Ng IO, Lam KY, Ng M, et al: Expression of P-glycoprotein, a multidrug-resistance gene product, is induced by radiotherapy in patients with oral squamous cell carcinoma. Cancer 83: 851- 857, 1998. Nielsen D, Eriksen J, Maare C, et al. P-glycoprotein expression in Ehrlich ascites tumour cells after in vitro and in vivo selection with daunorubicin. Br J Cancer 78: 1175-1180, 1998. Owa T, Yoshino H, Yoshimatsu K, Nagasu T: Cell cycle regulation in the G1 phase: a promising target for the development of new chemotherapeutic anticancer agents. Curr Med Chem 8: 1487-1503, 2001. Ruiz van Haperen VW, Veerman G, Eriksson S, et al. Induction of resistance to 2’,2’-difluorodeoxycytidine in the human ovarian cancer cell line A2780. Semin Oncol 22(Suppl 11): 35-41, 1995. Sandor V, Bakke S, Robey RW, et al: Phase I trial of the histone deacetylase inhibitor, depsipeptide, FR901228, NSC 630176), in patients with refractory neoplasms. Clin Cancer Res 8: 718- 728, 2002. Sandor V, Robbins AR, Robey R, et al. FR901228 causes mitotic arrest but does not alter microtubule polymerization. Anticancer Drugs 11: 445-545, 2000. Sato N, Ohta T, Kitagawa H, et al: FR901228, a novel histone deacetylase inhibitor, induces cell cycle arrest and subsequent apoptosis in refractory human pancreatic cancer cells. Int J Oncol 24: 679-685, 2004. Schondorf T, Neumann R, Benz C, et al: Cisplatin, doxorubicin and paclitaxel induce mdr1 gene transcription in ovarian cancer cell lines. Recent Results Cancer Res 161: 111-116, 2003. Sun D, Urrabaz R, Buzello C, Nguyen M: Induction of DNA ligase I by 1-beta-D-arabinosylcytosine and aphidicolin in Mia- PaCa human pancreatic cancer cells. Exp Cell Res 280: 90-96, 2002. Suwa H, Ohshio G, Arao S, et al: Immunohistochemical localization of P-glycoprotein and expression of the multidrug resistance- 1 gene in human pancreatic cancer: relevance to indicator of better prognosis. Jpn J Cancer Res 87: 641-649, 1996. Thomas H, Coley HM: Overcoming multidrug resistance in cancer: an update on clinical strategy on inhibiting P-glycoprotein. Cancer Control 10: 159-165, 2003. Tomida A, Naito M, Tsuruo T: Acute induction of adriamycin-resistance in human colon carcinoma HT-29 cells exposed to a sublethal dose of adriamycin. Jpn J Cancer Res 86: 224-232, 1995. Tsvetkov LM, Russev GC, Anachkova BB: Effect of mimosine on DNA synthesis in mammalian cells. Cancer Res 57: 2252- 2255, 1997. Wada M, Hosotani R, Lee JU, et al: An exogenous cdk inhibitor, butyrolactone-I, induces apoptosis with increased Bax/Bcl2 ratio in p53-mutated pancreatic cancer cells. Anticancer Res 18: 2559-2566, 1998. Wang G, Miskimins R, Miskimins WK: Mimosine arrests cells in G1 by enhancing the levels of p27(Kip1). Exp Cell Res 254: 64-71, 2000. Wartenberg M, Fischer K, Hescheler J, Sauer H: Modulation of intrinsic P-glycoprotein expression in multicellular prostate tumor spheroids by cell cycle inhibitors. Biochim Biophys Acta 1589: 49-62, 2002. Watson PA, Hanauske-Abel HH, Flint A, Lalande M: Mimosine reversibly arrests cell cycle progression at the G1-S phase border. Cytometry 12: 242-246, 1991. Zalatnai A: Epidermal growth factor receptor, somatostatin and bcl-2 in human pancreatic tumor xenografts. An immunohistochemical study. Pathol Oncol Res 5: 146-151, 1999. Zalatnai A: Potential role of cell cycle synchronizing agents in combination treatment modalities of malignant tumors. In vivo 19: 85-91, 2005. Zalatnai A, Bocsi J: Mimosine, a plant-derived amino acid induces apoptosis in human pancreatic cancer xenografts. Anticancer Res 23: 4007-4010, 2003. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 164 EP 169 PG 6 ER PT J AU Dundar, M Kocak, I Culhaci, N Erol, H AF Dundar, Mehmet Kocak, Izzet Culhaci, Nil Erol, Haluk TI Determination of Apoptosis Through Bax Expression in Cryptorchid Testis: an Experimental Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bax; spermatogonia; spermatocyte; undescended testis ID Bax; spermatogonia; spermatocyte; undescended testis AB The aim was to determine the expression of Bax in germ cells of rats with unilateral experimental cryptorchidism, and to evaluate the role of apoptosis in germ cell loss. Twenty-one prepubertal rats were randomly subdivided into three groups after the execution of the left cryptorchidism model. Group 1 (n=8), group 2 (n=6) and group 3 (n=7) rats were killed at the end of the first, second and third month, respectively. Bax expression was assessed in Sertoli cells, spermatogonia and spermatocytes by immunohistochemistry. Percentages of Bax expression in spermatocytes and spermatogonia were decreased in the left testicles in the 2nd and 3rd months compared to results obtained in the 1st month (p<0.05). Percentage of Bax expression in the left testicles of group 1, at the level of both spermatogonia and spermatocytes was higher than that in the right one (p<0.05). However, in groups 2 and 3, the higher Bax expression on the left side was only seen in the spermatocytes (p<0.05). In all groups, the mean weight of the left testicle was lower than that of the scrotal counterpart where the difference was significant only in groups 1 and 3 (p<0.05). The weight of the left and right testicles was increasing with time. In this model of cryptorchidism, the affected testis had a decreased weight compared to the normal one. Based on the increased Bax expression, we think that apoptosis may play a role in the germ cell loss. C1 [Dundar, Mehmet] Adnan Menderes University, Faculty of Medicine, Department of Urology, 09100 Aydin, Turkey. [Kocak, Izzet] Adnan Menderes University, Faculty of Medicine, Department of Urology, 09100 Aydin, Turkey. [Culhaci, Nil] Adnan Menderes University, Medical Faculty, Pathology DepartmentAydin, Turkey. [Erol, Haluk] Adnan Menderes University, Faculty of Medicine, Department of Urology, 09100 Aydin, Turkey. RP Dundar, M (reprint author), Adnan Menderes University, Faculty of Medicine, Department of Urology, 09100 Aydin, Turkey. EM medundar2002@yahoo.com CR Heiskanen P, Billig H, Toppari J, et al: Apoptotic cell death in the normal and cryptorchid human testis: The effect of human chorionic gonadotropin on testicular cell survival. Pediatr Res 40: 351-356, 1996 Shikone T, Billig H, Hsueh JW: Experimentally induced cryptorchidism increases apoptosis in rat testis. Biol Reprod 51: 865-872, 1994 Hikim AP, Lue Y, Yamamoto CM, et al: Key apoptotic pathways for heat-induced programmed germ cell death in the testis. Endocrinology 144: 3167-3175, 2003 Hikim AP, Lue Y, Diaz-Romero M, et al: Deciphering the pathways of germ cell apoptosis in the testis. J Steroid Biochem Mol Biol 85: 175- 182, 2003 Damavandi E, Hishikawa Y, Izumi S, et al: Involvement of Bax redistribution in the induction of germ cell apoptosis in neonatal mouse testes. Acta Histochem Cytochem 35: 449-459, 2002 Miura M, Sashagawa I, Suzuki Y, et al: Apoptosis and expression of apoptosis-related genes in the mouse testis following heat exposure. Fertil Steril 77: 787-793, 2002 Zini A, Abitbol J, Schulsinger D, et al: Restoration of spermatogenesis after scrotal replacement of experimentally cryptorchid rat testis. Assessment of germ cell apoptosis and eNOS expression. Urology 53: 223-227, 1999 Knudson CM, Tung KS, Tourtellote WG, et al: Bax deficient mice with lymphoid hyperplasia and male germ cell death. Science 270: 96-99, 1995 Dundar M, Kocak I, Culhaci N: A new experimental model for cryptorchidism: inguinoscrotal approach. Urol Res 29: 178-181, 2001 Hellstrom WJG, Monga M: Cryptorchidism and infertility. Male Infertility and Sexual Dysfunction., Ed: Hellstrom WJG), Springer, 1997, pp 307-322 Billig H, Furuta I, Rivier C, et al: Apoptosis in testis germ cells: developmental changes in gonadotropin dependence and localization to selective tubule stages. Endocrinology 136: 5-12, 1995 Majno G, Joris I: Apoptosis, oncosis and necrosis. An overview of cell death. Am J Pathol 146: 3-15, 1995 Tomomasa H, Adachi Y, Oshio S, et al: Germ cell apoptosis in undescended testis. The origin of its impaired spermatogenesis in the TS inbred rat. J Urol 168: 343-347, 2002 Ogi S, Tanji N, Yokoyama M, et al: Involvement of Fas in the apoptosis of mouse germ cells induced by experimental cryptorchidism. Urol Res 26: 17-21, 1998 Wang ZQ, Todani T, Watanabe A, et al: Germ cell degeneration in experimental unilateral cryptorchidism: role of apoptosis. Pediatr Surg Int 14: 9-13, 1998 Yan W, Samson M, Jegou B, Toppari J: Bcl-w forms complexes with Bax and Bak, and elevated ratios of Bax/Bcl-w and Bak/Bcl-w correspond to spermatogonial and spermatocyte apoptosis in the testis. Mol Endocrinol 14: 682-699, 2000 Xu J, Xu Z, Jiang Y, et al: Cryptorchidism induces mouse testicular germ cell apoptosis and changes in bcl-2 and Bax protein expression. J Environ Pathol Toxicol Oncol 19: 25-33, 2000 Ito K, Tanemura K, Gotoh H, et al: Apoptosis-like cell death in experimentally- induced cryptorchidism in adult mice. J Vet Med Sci 59: 353-359, 1997 Wang QZ, Watanabe Y, Toki A, et al: Altered distribution of Sertoli cell vimentin and increased apoptosis in cryptorchid rats. J Pediatr Surg 37: 648-652, 2002 Ohta Y, Nishikawa A, Fukazawa Y, et al: Apoptosis in adult mouse testis induced by experimental cryptorchidism. Acta Anat 157: 195- 204, 1996. Barqawi A, Trummer H, Meacham R: Effect of prolonged cryptorchidism on germ cell apoptosis and testicular sperm count. Asian J Androl 6: 47-51, 2004 Kocak I, Dundar M, Hekimgil M, et al: Assessment of germ cell apoptosis on cryptorchid rats. Asian J Androl 4: 183-186, 2002 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 170 EP 173 PG 4 ER PT J AU Vari, S Cserneky, M Kadar, A Szende, B AF Vari, Sandor Cserneky, Maria Kadar, Anna Szende, Bela TI Development of Present and Future of Telepathology in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE telepathology; consultations; diagnosis; database ID telepathology; consultations; diagnosis; database AB Pathologists in Hungary evaluate several hundred thousand histological specimens yearly, and a second opinion is requested in 5-10 percent of cases. Application of multimedia systems (i.e. telepathology) is convenient and efficient to establish a correct diagnosis in such cases. The first telepathology connection in Hungary has been established between the 1st Department of Pathology and Experimental Cancer Research, Semmelweis University and the Department of Pathology, Central Hospital of the Ministry of the Interior. Further development occurred in the course of various projects, supported by the EU (Interpath, Retransplant, BePro): new stations were established in three university institutes and six county hospitals. Electronic fixation of the images and their transmission by telephone line (ISDN) is easily available and an important feature of the multimedia system applied in telepathology. The system used by us is suited to evaluate frozen or paraffin-embedded histologic sections, as well as immunohistochemical and cytologic specimens, if necessary supplemented with transmission of macroscopic pictures. Our experience with bilateral consultations has proven the importance of telepathology. The telepathology system established in Hungary is now ready to join the telepathology network of the EU. C1 [Vari, Sandor] Varimed LtdBudapest, Hungary. [Cserneky, Maria] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Kadar, Anna] International Academy of Pathology, Hungarian DivisionBudapest, Hungary. [Szende, Bela] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Szende, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM bszende@korb1.sote.hu CR Fischer SI, Nandedkar MA, Williams BH, Abbondanzo L: Telepathology in a clinical consultative practice. Hum Pathol 32: 1327-1333, 2001 Allen EA, Ollayos CW, Tellado MV, et al: Characteristics of a telecytology consultation service. Hum Pathol 32: 1323-1326, 2001 Weinstein RS, Descour MR, Lian C, et al: Telepathology overview: From concept to implementation. Hum Pathol 23: 1283-1299, 2001 Kayser K, Fritz P, Drlicek M: Aspects of telepathology in routinary diagnostic work with specific emphasis on ISDN. Arch Anat Cytol Pathol 43: 216-218, 1995 Schwarzmann P, Schmid J, Schnorr C, et al: Telemicroscopy stations for telepathology based on broadband and ISDN connections. Arch Anat Cytol Pathol 43: 209-215, 1995 Gombas P, Szende B, Stotz G: Telecommunicative support of decision in diagnostic pathology. Experiences with the first Hungarian telepathology system, in Hungarian). Orvosi Hetilap 137: 2299-2303, 1996 Otto S, Kasler M: Cancer mortality and incidence in Hungary, related to European data, in Hungarian). Magyar Onkologia 46: 111-117, 2002 Molnar B, Berczi L, Diczhazy C, et al: Digital slide and virtual microscopy based routine and telepathology evaluation of routine gastrointestinal biopsy specimens. J Clin Pathol 56: 1-6, 2003 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 174 EP 177 PG 4 ER PT J AU Venizelos, I Tamiolakis, D Lambropoulou, M Nikolaidou, S Bolioti, S Papadopoulos, H Papadopoulos, N AF Venizelos, Ioannis Tamiolakis, Demetrio Lambropoulou, Maria Nikolaidou, Sylva Bolioti, Sophia Papadopoulos, Hlias Papadopoulos, Nikolas TI An Unusual Case of Posttransplant Peritoneal Primary Effusion Lymphoma with T-cell Phenotype in a HIV-negative Female, not Associated with HHV-8 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE primary effusion lymphoma; non-Hodgkin’s lymphoma; immunohistochemistry ID primary effusion lymphoma; non-Hodgkin’s lymphoma; immunohistochemistry AB Primary effusion lymphoma (PEL) is a recently individualized form of non-Hodgkin’s lymphoma (WHO classification) that mainly develops in HIV infected males, more frequently in homosexuals and advanced stages of the disease (total CD4+ lymphocyte count below 100-200/µL). Occasionally, it appears in other immunodepressive states (such as solid organs transplant period) and even, although very rarely, in immunocompetent patients. From a pathogenetic point of view, PEL has been related to Kaposi’s sarcoma associated herpes virus (also named human herpesvirus 8, HHV-8), an etiological factor of Kaposi’s sarcoma. The relative infrequency of this disease, the absence of wide casuistics allowing a better characterization, and its unfavorable outcome support the need of a deeper knowledge. We present here the clinical-biological findings of a patient, HIVseronegative, who was diagnosed with peritoneal PEL of T-cell origin, and not HHV-8-associated, five years after renal transplantation. C1 [Venizelos, Ioannis] Ippokratio General Hospital of Salonica, Department of PathologySalonica, Greece. [Tamiolakis, Demetrio] General Hospital of Chania, Department of CytologyChania, Greece. [Lambropoulou, Maria] Democritus University of Thrace, Department of Histology-Embryology, Dragana, 68100 Alexandroupolis, Greece. [Nikolaidou, Sylva] General Hospital of Chania, Department of CytologyChania, Greece. [Bolioti, Sophia] General Hospital of Chania, Department of CytologyChania, Greece. [Papadopoulos, Hlias] Democritus University of Thrace, Department of Histology-Embryology, Dragana, 68100 Alexandroupolis, Greece. [Papadopoulos, Nikolas] Democritus University of Thrace, Department of Histology-Embryology, Dragana, 68100 Alexandroupolis, Greece. RP Papadopoulos, N (reprint author), Democritus University of Thrace, Department of Histology-Embryology, 68100 Alexandroupolis, Greece. EM npapad@med.duth.gr CR Carbone A, Cilia AM, Gloghini A, et al: Primary effusion lymphoma cell lines harbouring human herpesvirus type 8. Leuk Lymphoma 36: 447-456, 2000. Fassone L, Bhatia K, Gotierrez M, et al: Molecular profile of Epstein-Barr virus infection in HHV-8-positive primary effusion lymphoma. Leukemia 14: 271-277, 2000. Ascoli V, Scalzo CC, Danese C, et al: Human herpes virus-8 associated primary effusion lymphoma of the pleural cavity in HIV-negative elderly men. Eur Respir J 14: 1231-1234, 1999. Chang Y, Cesarman E, Pessin MS, et al: Identification of herpesvirus- like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science 266:1865-1869, 1994 Frank D, Cesarman E, Liu YF, et al: Post-transplantation lymphoproliferative disorders frequently contain type A and not type B Epstein-Barr virus. Blood 85: 1396-1403, 1995. Gaidano G, Ballerini P, Gong JZ, et al: p53 mutations in human lymphoid malignancies: association with Burkitt lymphoma and chronic lymphocytic leukemia. Proc Natl Acad Sci USA 88: 5423-5417, 1991. Ohshima K, Ishiguro M, Yamasaki S, et al: Chromosomal and comparative genomic analyses of HHV-8-negative PEL in 5 HIVnegative Japanese patients. Leuk Lymphoma 43: 595-601, 2002 Shimazaki M, Fujita M, Tsukamoto K, et al: An unusual case of PEL in a HIV-negative patient not pathogenetically associated with HHV-8. Eur J Haematol 71: 62-67, 2003. Said JW, Shintaku IP, Asou H, et al: Herpesvirus 8 inclusions in primary effusion lymphoma: report of a unique case with T-cell phenotype. Arch Pathol Lab Med 123: 257-260, 1999 Kinney MC, Kadin ME. The pathologic and clinical spectrum of anaplastic large cell lymphoma and correlation with ALK gene dysregulation. Am J Clin Pathol. 1999;111, suppl 1): S56- S67. Chan AC, Chan JK, Yan KW, Kwong YL: Anaplastic large cell lymphoma presenting as a pleural effusion and mimicking primary effusion lymphoma. A report of two cases. Acta Cytol 47: 809-816, 2003. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 178 EP 181 PG 4 ER PT J AU Aydin, MV Cekinmez, M Kizilkilic, O Kayaselcuk, F Sen, O Altinors, N AF Aydin, M Volkan Cekinmez, Melih Kizilkilic, Osman Kayaselcuk, Fazilet Sen, Orhan Altinors, Nur TI Unusual Case of Skull Metastasis Secondary to Pancreatic Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE adenocarcinoma; metastasis; pancreas; skull ID adenocarcinoma; metastasis; pancreas; skull AB Skull metastasis must be kept in mind when considering the differential diagnosis of a skull tumor. Skull metastases cause local swelling that is usually painless, and rarely they lead to neurologic dysfunction. Despite the fact that hematogenous skull metastases can be caused by nearly all types of tumors (lung, prostate, thyroid carcinoma, malignant melanoma), breast cancer is associated with the highest rate of metastatic skull lesions. We report an extremely rare case of skull metastasis from a pancreatic adenocarcinoma, in a 65-year-old woman, presented with painless frontoparietal scalp swelling which developed within three months. To the best of our knowledge, this is the second case involving the skull secondary to a pancreatic adenocarcinoma, and the first case when skull metastasis was the first evidence of a pancreatic adenocarcinoma. C1 [Aydin, M Volkan] Baskent University, Departments of Neurosurgery, Yuregir, 1250 Adana, Turkey. [Cekinmez, Melih] Baskent University, Departments of Neurosurgery, Yuregir, 1250 Adana, Turkey. [Kizilkilic, Osman] Baskent University, Department of RadiologyAdana, Turkey. [Kayaselcuk, Fazilet] Baskent University, Department of PathologyAdana, Turkey. [Sen, Orhan] Baskent University, Departments of Neurosurgery, Yuregir, 1250 Adana, Turkey. [Altinors, Nur] Baskent University, Departments of Neurosurgery, Yuregir, 1250 Adana, Turkey. RP Aydin, MV (reprint author), Baskent University, Departments of Neurosurgery, 1250 Adana, Turkey. EM volkan8@hotmail.com CR Bontoux D, Plazanet F, Azais I: Distribution of bone metastases of cancers. A scintigraphic study of 376 cases. Bull Acad Natl Med 182: 997- 1008, 1998 Constans JP, Donzelli R: Surgical features of cranial metastases. Surg Neurol 15: 35-38, 1981 Galicich JH, Arbit E, Wronski M: Metastatic brain tumors. In: Neurosurgery., Eds: Wilkins RH, Rengarachary SS), Vol 1, 2nd ed, New York, McGraw-Hill, 1996, pp 807-821 Gloria-Cruz TI, Schachern PA, Paparella MM, et al: Metastases to temporal bones from primary nonsystemic malignant neoplasms. Arch Otolaryngol Head Neck Surg 6: 209-214, 2000 Hornig K: Extensive osteolytic calotte destructions in primary breast cancer. Radiologe 28: 132-133, 1988 Igarashi M, Card GG, Johnson PE, Alford BR: Bilateral sudden hearing loss and metastatic pancreatic adenocarcinoma. Arch Otolaryngol 105: 196-199, 1979 Michael JB, Gokaslan ZL, DeMonte F, et al: Surgical resection of calvarial metastases overlying dural sinuses. Neurosurgery 48: 745-755, 2001 Stark AM, Eichmann T, Mehdorn HM: Skull metastases: clinical features, differential diagnosis, and review of the literature. Surg Neurol 60: 219-226, 2003 Wong GKC, Boet R, Poon WS, Ng HK: Lytic skull metastasis secondary to thyroid carcinoma in an adolescent. HKMJ 8: 149-151, 2002 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 182 EP 183 PG 2 ER PT J AU Terracciano, L Bouygue, RG Startari, R Guerriero, F Bouygue, MC AF Terracciano, Luigi Bouygue, R Gabriel Startari, Rosario Guerriero, Francesca Bouygue, MR Colette TI A Case of Fulminant ''Talc Pneumoconiosis”: Where is the Smoking Gun? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review C1 [Terracciano, Luigi] University of Milan Medical School, Melloni Paediatria, 52 Via Melloni, 20129 Milan, Italy. [Bouygue, R Gabriel] University of Milan Medical School, Melloni Paediatria, 52 Via Melloni, 20129 Milan, Italy. [Startari, Rosario] University of Milan Medical School, Melloni Paediatria, 52 Via Melloni, 20129 Milan, Italy. [Guerriero, Francesca] University of Milan Medical School, Melloni Paediatria, 52 Via Melloni, 20129 Milan, Italy. [Bouygue, MR Colette] University of Milan Medical School, Melloni Paediatria, 52 Via Melloni, 20129 Milan, Italy. RP Terracciano, L (reprint author), University of Milan Medical School, Melloni Paediatria, 20129 Milan, Italy. CR Dekel Y, Rath-Wolfson L, Rudniki Carlos, Koren R. Talc inhalation is a life-threatening condition. Path Oncol Res 10:231-233, 2004 Deer WA, Howie RA, Zussman J: Rock-Forming Minerals, vol III: Sheet Silicates. London, 1963, Longmans Awai K, Yamane K, Nishioka Y, et al: Analysis of pyrophillitosis by highresolution computed tomography. Nippon Igaku Hosahsen Gakkai Zasshi 51: 656-662, 1991 Kurahashi M, Mizutani J, Hioki A: Determination of trace elements in sediment reference materials by monochromatic X-ray excitation X-ray fluorescence spectrometry. Anal Sci 21: 827-832, 2005 Karik AB: Immune response to exposure to occupational and environmental agents. ICMR Bulletin 31: 73-79, 2001 Miller A, Teirstein AS, Bader ME, et al: Talc pneumoconiosis. Significance of sublight microscopic mineral particles. Am J Med 50: 395-402, 1971 Fine LJ, Peters JM, Burgess WA, Di Berardinis LJ: Studies of respiratory morbidity in rubber workers. Part IV. Respiratory morbidity in talc workers. Arch Environ Health 31: 195-200, 1976 Accessed from: http://minerals.usgs.gov/minerals/pubs/commodity/ talc NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 184 EP 184 PG 1 ER PT J AU Koren, R AF Koren, Rumelia TI Reply SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture C1 [Koren, Rumelia] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Pathology, Ramat AvivTel Aviv, Israel. RP Koren, R (reprint author), Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Pathology, Tel Aviv, Israel. CR Dimopoulos G, Piagnerelli M, Berre J, et al: Post mortem examination in the intensive care unit: still useful? Intensive Care Med 30: 2080-2085, 2004 Scancarello G, Romeo R, Sartorelli E: Respiratory disease as a result of talc inhalation. J Occup Environ Med 38: 610-614, 1996 Pairaudeau PW, Wilson RG, Hall MA, Milne M: Inhalation of baby powder: an unappreciated hazard. Br Med J 302: 1200- 1201, 1991 Steele AA: Suicidal death by aspiration of talcum powder. Am J Forensic Med Pathol 11: 316-318, 1990 Hollinger M: Pulmonary toxicity of inhaled and intravenous talc. Toxicol Lett 52: 121-127; discussion 117-119, 1990 Cotton WH, Davidson PJ: Aspiration of baby powder. N Engl J Med 313: 1662, 1985 Motomatsu K, Adachi H, Uno T: Two infant deaths after inhaling baby powder. Chest 75:448-450, 1979 Pfenninger J, D’Apuzzo V: Powder aspiration in children. Report of two cases. Arch Dis Child 52:157-159, 1977 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 185 EP 185 PG 1 ER PT J AU Nagy, Zs AF Nagy, Zsuzsanna TI Zoledronic acid (ZOMETA): a Significant Improvement in the Treatment of Bone Metastases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline C1 [Nagy, Zsuzsanna] St. Imre Hospital, Department Clinical Oncology, Tetenyi u. 12-16, H-1115 Budapest, Hungary. RP Nagy, Zs (reprint author), St. Imre Hospital, Department Clinical Oncology, H-1115 Budapest, Hungary. CR Coleman RE: Metastatic bone disease: clinical features, pathophysiology and treatment strategies. Cancer Treat Rev 27: 165- 176, 2001 Russell RG, Rogers MJ: Bisphosphonates: From the laboratory to the clinic and back again. Bone 25: 97-106, 1999 Hurst M, Noble S: Clodronate: A review of its use in breast cancer. Drugs Aging 15: 143-167, 1999 Theriault RL, Lipton A, Hortobagyi GN, et al: Pamidronate reduces skeletal morbidity in women with advanced breast cancer and lytic bone lesions: A randomized, placebo-controlled trial – Protocol 18 Aredia Breast Cancer Study Group. J Clin Oncol 17: 846-854, 1999 Gordon D, Rosen L, Coleman RE, et al: Long-term efficacy and safety of zoledronic acid compared with pamidronate in treatment of skeletal complications in patients with advanced multiple myeloma of breast cancer, Proc Am Soc Clin Oncol 22: 47, 2003, abstr 188) Perry CM, Figgitt DP: Zoledronic acid: A review of its use in patients with advanced cancer. Drugs 64: 1197-1211, 2004 Kohno N, Aogi K, Minami H, et al: Zoledronic acid significantly reduces skeletal complications compared with placebo in Japanese women with bone metastases from breast cancer: a randomized, placebo-controlled trial. J Clin Oncol 23: 3314- 3321, 2005 Seneratne SG, Colston KW: Direct effects of bisphosphonates on breast cancer cells. Breast Cancer Res 4: 18-23, 2002 Powles T, McCloskey E, Kurkilahti M: Oral clodronate for adjuvant treatment of operable breast cancer: results of a randomized, double-blind, placebo-controlled multicenter trial. Proc Am Soc Clin Oncol 23: 9, 2004, abstr 528) Jaschke A, Bastert G, Solomayer EF, et al: Adjuvant clodronate treatment improves the overall survival of primary breast cancer patients with micrometastases to bone marrow – a longtime follow-up. Proc Am Soc Clin Oncol 23: 9, 2004, abstr 529) Coleman R, Gralow J, Bell R, Lipton A: Zoledronic acid is being investigated for the prevention of bone metastases in patients with early stage cancer. 7th workshop on Bisphosphonates, 24-26 March 2004, Davos, Switzerland, abstr 60) Brufsky A, Harker GW, Beck J, et al: Zoledronic acid for prevention of cancer treatment induced bone loss in postmenopausal women with early breast cancer receiving adjuvant letrozole: preliminary results of the Z-FAST trial. 27th Annual San Antonio Breast Cancer Symposium 2004, San Antonio, abstr 1114) Brufsky A, Harker W, Beck J, et al: Zoledronic acid, ZA, effectively inhibits cancer treatment-induced bone loss, CTIBL, in postmenopausal women, PMW, with early breast cancer, BCa, receiving adjuvant Letrozole, Let): 12 mos BMD results of the FAST trial. ASCO Annual Meeting, Orlando, 2005, abstr 533) Botteman MF, Aapro MS, Hay JW, et al: Cost effectiveness of intravenous, IV, zoledronic acid vs other IV bisphosphonates for the prevention of bone complications in breast cancer patients with bone metastases: A Markov model from the UK perspective. ASCO Annual Meeting, Orlando, 2005, abstr 721) Body J, Lichinitser M, Tjulandin S, et al: Effect of oral ibandronate versus intravenous, i.v., zoledronic acid on markers of bone resorption with breast cancer and bone metastases: Results from a comparative phase III trial. ASCO Annual Meeting, Orlando, 2005, abstr 534) Lipton A, Hei Y, Coleman R, et al: Suppression of bone turnover markers by zoledronic acid and correlation with clinical outcome. ASCO Annual Meeting, Orlando, 2005, abstr 532) Jung J, Hwang G, Lee Y, et al: Pamidronate as adjuvant treatment for prevention of bone metastasis in breast cancer. ASCO Annual Meeting, Orlando, 2005, abstr 888) Hortobagyi GN: Progress in the management of bone metastases: One continent at a time? J Clin Oncol 23, 1-3, 2005 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2005 VL 11 IS 3 BP 186 EP 186 PG 1 ER PT J AU Kopper, L Timar, J AF Kopper, Laszlo Timar, Jozsef TI Genomics of Prostate Cancer: Is There Anything to ''Translate''? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE prostate cancer; progression; genomics; proteomics ID prostate cancer; progression; genomics; proteomics AB This review provides an up-dated collection of data concerning the genetic and epigenetic changes during development, growth and progression of prostate cancer. Hereditary and susceptibility factors have a long list, similarly to the expression of single genes connected to various cell functions. It was a hope that covering a large set of genes, array technologies would clarify very rapidly the role of genetics in malignant diseases, offering targets for molecular diagnostics and therapy. The power of high-throughput techniques for the detection and global analysis of gene expression is unquestionable, interesting, astonishing as well as puzzling data have already been obtained. However, the standardization of the procedures is still missing and the reproducibility is rather low in many instances. Moreover, the different array methods can select different gene expression profiles, which makes the decision rather difficult. Another important question is, coming again from the array technologies, how far the genotype (the gene profiles or fingerprints) can reflect the actual phenotype in a highly complex and readily changing disease as cancer. Proteomics will provide a closer look to this seemingly unanswerable problem. We are at the beginning of the exploration of the behavior of cancer cells in order to apply a more effective therapy based on a more reliable set of diagnostic and prognostic informations. C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 78., H-1085 Budapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor ProgressionBudapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM kopper@korb1.sote.hu CR Arakawa A, Soh S, Chakaborty S, et al: Prognostic significance of angiogenesis in clinically localized prostate cancer, staining for Factor VIII-related antigen and CD34 antigen). Prostate Cancer Prostatic Dis 1:32-38, 1997 Ashida S, Nakagawa H, Katagiri T et al: Molecular features of the transition from prostate intraepithelial neoplasia, PIN, to prostate cancer: genome-wide gene-expression profiles of prostate cancers and PINs. Cancer Res 64: 5963-5972, 2004 Banez LL, Srivastava S, Moul JW: Proteomics in prostate cancer. Curr Opin Urol 15: 151-156, 2005 Bernard D, Pourtier-Manzanedo A, Gil J and Beach DH: Myc confers androgen-independent prostate cancer cell growth. J Clin Invest 112:1724-1731, 2003 Calvo A, Gonzales-Moreno O, Yoon C-Y et al: Prostate cancer and the genomic revolution: Advances using microarray analyses. Mutat Res 576: 66-79, 2005 Chen G, Shukeir N, Potti A, et al: Up-regulation of Wnt-1 and beta-catenin production in patients with advanced metastatic prostate carcinoma: potential pathogenetic and prognostic implications. Cancer 101:1345-1356, 2004 Chiorino G, Acquadro F, Mello Grand M, et al: Interpretation of expression-profiling results obtained from different platforms and tissue sources: examples using prostate cancer data. Eur J Cancer 40: 2592-2603, 2004 DeMarzo AM, Nelson WG, Isaacs WB et al: Pathological and molecular aspects of prostate cancer. Lancet 361: 955-964, 2003 Deutsch E, Maggiorella L, Eschwege P et al: Environmental, genetic and molecular features of prostate cancer. Lancet Oncol 5: 303-313, 2004 Dhanasekaran SM, Barrette TR, Ghosh D, et al: Delineation of prognostic biomarkers in prostate cancer. Nature 412:822-826, 2001 Di Lorenzo G, Tortora G, D’Armiento F, et al: Expression of epidermal growth factor receptor correlates with disease relapse and progression to androgen-independence in human prostate cancer. Clin Cancer Res 8:3438-3444, 2002 Dong Y, Zhang H, Hawthorn L, et al: Delineation of the molecular basis for selenium-induced growth arrest in human prostate cancer cells by oligonucleotide array. Cancer Res 63: 52-59, 2003 Edwards J, Krishna NS, Witton CJ and Bartlett JMS: Gene amplifications associated with the development of hormoneresistant prostate cancer. Clin Cancer Res 9:2571-2581, 2003 Edwards SM, Eeles RA: Unravelling the genetics of prostate cancer. Am J Med Genet C, Semin Med Genet 129: 65-73, 2004 Furuya Y, Nagakawa O and Fuse H: Prognostic significance of serum soluble Fas level and its change during regression and progression of advanced prostate cancer. Endocrine J 50:629- 633, 2003 Glinsky GV, Glinskii AB, Stephenson AJ, et al: Gene expression profiling predicts clinical outcome of prostate cancer. J Clin Invest 113: 913-923, 2004 Gravdal K, Halvorsen OJ, Haukaas SA and Akslen LA: Expression of bFGF/FGFR-1 and vascular proliferation related to clinicopathologic features and tumor progression in localized prostate cancer. Virchows Arch, 2005, in press) Gregory CW, Fei X, Ponguta L, et al: Epidermal growth factor increases coactivation of the androgen receptor in recurrent prostate cancer. J Biol Chem 179:7119-7130, 2004 Gsur A, Feik E, Madersbacher S: Genetic polymorphisms and prostate cancer risk. World J Urol 21: 414-423, 2004 Henshall SM, Afar DEH, Hiller J, et al: Survival analysis of genome-wide gene expression profiles of prostate cancers identifies new prognostic targets of disease relapse. Cancer Res 63:4196-4203, 2003 Hughes C, Murphy A, Martin C, et al: Molecular pathology of prostate cancer. J Clin Pathol 58: 673-684, 2005 Jennbacken K, Vallbo C, Wang W and Damber JE: Expression of vascular endothelial growth factor C, VEGF-C, and VEGF receptor-3 in human prostate cancer is associated with regional lymph node metastasis. Prostate 65:110-116, 2005 Jung K, Lein M, Stephan C, et al: Comparison of 10 serum bone turnover markers in prostate carcinoma patients with bone metastatic spread: diagnostic and prognostic implications. Int J Cancer 111:783-791, 2004 Karayi MK, Markham AF: Molecular biology of prostate cancer. Prostate Cancer Prostatic Dis 7: 6-20, 2004 Kaushal V, Mukunyadzi P, Dennis RA, et al: Stage-specific characterization of the vascular endothelia growth factor axis in prostate cancer: expression of lymphangiogenic markers is associated with advanced-stage disease. Clin Cancer Res 11:584-593, 2005 Keshava C, McCanlies EC, Weston A: CYP3A4 polymorphisms – potential risk factors for breast and prostate cancer: a HuGE review. Am J Epidemiol 160: 825-841, 2004 Kil PJ, Goldschmidt HM, Wieggers BJ et al: Tissue polypeptide- specific antigen, TPS, determinations before and during intermittent maximal androgen blockade in patients with metastatic prostatic carcinoma. Eur Urol 43:31-38, 2003 Klezovitch O, Chevillett J, Mirosevitch J, et al: Hepsin promotes prostate cancer progression and metastasis. Cancer Cell 6:186-195, 2004 Koivisto PA, Hyytinen ER, Matikainen M, et al: Germline mutation analysis of the androgen receptor gene in Finnish patients with prostate cancer. J Urol 171:431-433, 2004 Kwak C, Jeong SJ, Park MS, et al: Prognostic significance of the nadir prostate specific antigen after hormone therapy for prostate cancer. J Urol 168:995-1000, 2002 Lara PN Jr, Meyers FJ, Gray CR, et al: Her-2/neu is overexpressed infrequently in patients with prostate carcinoma. Cancer 94:2584-2589, 2002 Lehrer S, Diamond EJ, Mamkine B, et al: Serum interleukin-8 is elevated in men with prostate cancer bone metastases. Technol Cancer Res Treat 3:411, 2004 Li LC, Okino ST, Dahiya R: DNA methylation in prostate cancer. Biophys Biochem Acla 1704: 87-102, 2004 Li Y, Sarkar FH: Gene expression profiles of genistein-treated PRCA3 prostate cancer cells. J Nutr 132: 3623-3631, 2002 Liede A, Karlan BY, Narod SA: Cancer risks for male carriers of germline mutations in BRCA1 or BRCA2. J Clin Oncol 22: 735-742, 2004 Lorenzo GD, Bianco R, Torora G and Ciardillo F: Involvement of growth factor receptors of the epidermal growth factor receptor family in prostate cancer development and progression to androgen independence. Clin Prostate Cancer 2:50-57, 2003 Mehta PB, Jenkins BL, McCarthy L, et al: MEK5 overexpression is associated with metastatic prostate cancer, and stimulates proliferation, MMP-9 expression and invasion. Oncogene 22:1381-1389, 2003 Miyata Y, Sakai H, Hayashi T and Kanetake H: Serum insulinlike growth factor binding protein-3/prostate-specific antigen ratio is a useful predictive marker in patients with advanced prostate cancer. Prostate 54:125-132, 2003 Mohler JL, Gregory CW, Ford III OH, et al: The androgen axis in recurrent prostate cancer. Clin Cancer Res 10:440-448, 2004 Nakamura T, Scorilas A, Stephan C, et al: Quantitative analysis of macrophage inhibitory cytokine-1, MIC-1, gene expression in human prostatic tissues. Br J Cancer 88:1101-1104, 2003 Nakayama M, Gonzalgo, Yegnasubramanian S, et al: GSTP1 CpG island hypermethylation as a molecular biomarker for prostate cancer. J Cell Biochem 91: 540-552, 2004 Nie D, Che M, Zacharek A, et al: Differential expression of thromboxane synthase in prostate carcinoma: role in tumor cell motility. Am J Pathol 164:429-439, 2004 Offesen BV, Borre M, Brandt F, et al: Comparison of methods of microvascular staining and quantification in prostate carcinoma: relevance to prognosis. APMIS 110:177-185, 2002 Porkka KP, Visakorpi T: Molecular mechanisms of prostate cancer. Eur Urol 45: 683-691, 2004 Roodman GD: Mechanisms of bone metastasis. N Engl J Med 350:1655-1664, 2004 Ross JS, Kallakury BV, Sceehan CE, et al: Expression of nuclear factor-kappa B and I kappa B alpha proteins in prostatic adenocarcinomas: correlation of nuclear factor-kappa B immunoreactivity with disease recurrence. Clin Cancer Res 10:2466-2472, 2004 Rubin MA, Buyyounouski M, Bagiella E, et al: Microvessel density in prostate cancer: lack of correlation with tumor grade, pathologic stage, and clinical outcome. Urology 53:542-547, 1999 Rubin MA, Mucci NR, Figurski J, et al: E-cadherin expression in prostate cancer: a broad survey using high-density tissue microarray technology. Hum Pathol 32:690-697, 2001 Shappell SB, Manning S, Boeglin W, et al: Alterations in lipoxygenase and cyclooxygenase-2 catalytic activity and mRNA expression in prostate carcinoma. Neoplasia 3:287-303, 2001 Shariat SF, Roudier MP, Wilcox GE, et al: Comparison of immunohistochemistry with reverse transcription-PCR for the detection of micrometastatic prostate cancer in lymph nodes. Cancer Res 63:4662-4671, 2003 Stefanou D, Batistatou A, Kamina S, et al: Expression of vascular endothelial growth factor, VEGF, and association with microvessel density in benign prostatic hyperplasia and prostate cancer. In Vivo 18:155-160, 2004 Stewart DA, Cooper CR and Sikes RA: Changes in extracellular matrix, ECM, and ECM-associated proteins in the metastatic progression of prostate cancer. Reprod Biol Endocrinol 2:1-14, 2004 Straume O, Chappuis PO, Salvesen HB, et al: Prognostic importance of glomeruloid microvascular proliferation indicates an aggressive angiogenic phenotype in human cancers. Cancer Res 62:6808-6811, 2002 Subbarayan V, Xu XC, Kim J, et al: Inverse relationship between 15-lipoxygenase-2 and PPAR-gamma gene expression in normal epithelia compared with tumor epithelia. Neoplasia 7:280-293, 2005 Thompson J, Hyytinen ER, Haapala K, et al: Androgen receptor mutations in high-grade prostate cancer before hormonal therapy. Lab Invest 83:1709-1713, 2003 Timar J, Raso E, Dome B, et al: Expression, subcellular localization and putative function of platelet-type 12-lipoxygenase in human prostate cancer cell lines of different metastatic potential. Int J Cancer 87:37-43, 2000 Trikha M, Timar J, Lundy SK, et al: Human prostate carcinoma cells express functional αIIbβ3 integrin. Cancer Res 56:5071- 5078, 1996 Uzgare AR, Isaacs JT: Enhanced redundancy in Akt and mitogen- activated protein-kinase-induced survival of malignant versus normal prostate epithelial cells. Cancer Res 64: 6190- 6199, 2004 Uzgare AR, Isaacs JT: Prostate cancer: potential targets of antiproliferative and apoptotic signaling pathways. Int J Biochem Cell Biol 37: 707-714, 2005 Wang W, Bergh A and Damber JE: Cyclooxygenase-2 expression correlates with local chronic inflammation and tumor neovascularization in human prostate cancer. Clin Cancer Res 11:3250-3256, 2005 Welch DR and Hunger KW: A new member of the growing family of metastasis suppressors identified in prostate cancer. J Natl Cancer Inst 95:839-891, 2003 Xin W, Rhodes DR, Ingold C, et al: Dysregulation of annexin family is associated with prostate cancer progression. Am J Pathol 162: 255-261, 2003 Yu EY, Yu E, Meyer GE and Brawer MK: The relation of p53 protein nuclear accumulation and angiogenesis in human prostatic carcinoma. Prostate Cancer Prostatic Dis 1:39-44, 1997 Yu YP, Landsittel D, Jing L, et al: Gene expression alteration in prostate cancer predicting tumor aggression and preceding development of malignancy. J Clin Oncol 22:2790-1799, 2004 Zellweger T, Ninck C, Mirlacher M, et al: Tissue microarray analysis reveals prognostic significance of syndecan-1 expression in prostate cancer. Prostate 55:20-29, 2003 Zeng Y, Opeskin K, Baldwin ME, et al: Expression of vacular endothelial growth factor receptor-3 by lymphatic endothelial cells is associated with lymph node metastasis in prostate cancer. Clin Cancer Res 10:5137-5144, 2004 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2005 VL 11 IS 4 BP 197 EP 203 PG 7 ER PT J AU Mehes, G AF Mehes, Gabor TI Chromosome Abnormalities with Prognostic Impact in B-cell Chronic Lymphocytic Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE CLL; chromsomal deletions; IgVH; prognosis ID CLL; chromsomal deletions; IgVH; prognosis AB The detailed analysis of the biologic features led to a rapid increase in clinically relevant information in CLL. The recognition of the prognostic role of IgVH hypermutation status and related phenotypic changes (CD38, ZAP-70 expression) as well as of chromosome abnormalities defined by cytogenetic analysis enabled a refined classification of the disease. Improvements in karyotyping and the introduction of fluorescence in situ hybridization (FISH) in routine hematological diagnostics raised the detection rate of chromosomal aberrations to approx. 60-80% in CLL. Among them, deletions of 17p and 11q have been associated with unfavorable prognosis. The deletion of the p53 locus (17p13) was described as the strongest independent predictor for aggressive behavior, resistance to chemotherapy and early death. On the contrary, an isolated deletion at 13q14 or a normal karyotype was related with a long survival. Classical and molecular cytogenetic analysis became an important tool for individual risk estimation. Unlike any other approaches, cytogenetic monitoring reflects the genetic heterogeneity and clonal growth dynamics during the course of the disease. C1 [Mehes, Gabor] University of Pecs, Department of Pathology, Szigeti ut 12., H-7602 Pecs, Hungary. RP Mehes, G (reprint author), University of Pecs, Department of Pathology, H-7602 Pecs, Hungary. EM gabor.mehes@freemail.hu CR Bacher U, Kern W, Schoch C, et al: Discrimination of chronic lymphocytic leukemia, CLL, and CLL/PL by cytomorphology can clearly be correlated to specific genetic markers as investigated by interphase fluorescence in situ hybridization, FISH). Ann Hematol 83: 349-355, 2004. Bea S, Lopez-Guillermo A, Ribas M, et al: Genetic imbalances in chronic lymphocytic leukemia and transformed large-cell lymphoma, Richter’s syndrome). Am J Pathol 161: 957-968, 2002. Byrd JC, Stilgenbauer S, Flinn IW: Chronic lymphocytic leukemia. Hematology 163-183, 2004. Calin GA, Liu CG, Sevignani C, et al. MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proc Natl Acad Sci USA 101: 11755-11760, 2004. Capello D, Guarini A, Berra E, et al: Evidence of biased immunoglobulin variable gene usage in highly-stable B-cell chronic leukemia. Leukemia 18: 1941-1947, 2004. Cuneo A, Rigolin GM, Bigoni R, et al: Chronic lymphocytic leukemia with 6q- shows distinct hematological features and intermediate prognosis. Leukemia 18: 476-483, 2004. De Jong D, van’t Veer LJ: Tools for molecular risk-stratification for clinical purposes: CLL as a prototype. Eur J Hum Genet 12: 423, 2004. De la Fuente MT, Casanova B, Cantero E, et al: Involvement of p53 in alpha4beta1 integrin-mediated resistance of B-CLL cells to fludarabine. Biochem Biophys Res Commun 311: 708- 712, 2003. Dewald GW, Brockman SR, Paternoster SF, et al: Chromosome anomalies detected by fluorescence in situ hybridisation: correlation with significant biological features in B-cell chronic lymphocytic leukemia. Br J Haematol 121: 287-295, 2003. Dohner H, Stilgenbauer S, Dohner K, et al: Chromosome aberrations in B-cell chronic lymphocytic leukemia: reassessment based on molecular cytogenetic analysis. J Mol Med 77: 266- 281, 1999. Glassman AB, Hayes KJ: The value of fluorescence in situ hybridization in the diagnosis and prognosis of chronic lymphocytic leukemia. Cancer Genet Cytogenet 158: 88-91, 2005. Hammarsund M, Corcoran MM, Wilson W, et al: Characterization of a novel B-CLL candidate gene – DLEU – in the 13q14 tumor suppressor locus. FEBS Lett 556: 75-80, 2004. Haslinger C, Schweifer N, Stilgenbauer S, et al: Microarray gene expression profiling of B-cell chronic lymphoid leukemia subgroups defined by genomic aberrations and VH mutation status. J Clin Oncol 22: 3937-3949, 2004. Hernandez JM, Mecucci C, Criel A, et al: Cytogenetic analysis of B-cell chronic lymphocytic leukemias classified according to morphologic and immunophenotypic criteria. Leukemia 9: 2140-2146, 1995. Karhu R, Tobin G, Thunberg U, et al: More extensive genetic alterations in unmutated than in hypermutated cases of chronic lymphocytic leukemia. Genes Chromosomes Cancer 37: 417- 420, 2003. Krober A, Seiler T, Benner A, Bullinger L, et al: VH mutation status, CD38 expression level, genomic aberrations and survival in chronic lymphocytic leukemia. Blood 100: 1410-1416, 2002. Matolcsy A: High-grade transformation of low-grade non- Hodgkin’s lymphomas: mechanisms of tumor progression. Leuk Lymphoma 34: 251-259, 1999. Matrai Z: CD38 as a prognostic marker in CLL. Hematology 10: 39-46, 2005. Matthews C, Catherwood M, Morris TC, Alexander HD: Routine analysis of IgVH mutational status in CLL patients using BIOMED-2 standardized primers and protocols. Leuk Lymphoma 45: 1899-1904, 2004. Messmer BT, Albesiano E, Efremov DG, et al: Multiple distinct sets of stereotyped antigen receptors indicate a role for antigen in promoting chronic lymphocytic leukemia. J Exp Med 200: 519-525, 2004. Orchard JA, Ibbotson RE, Davis Z, et al: ZAP-70 expression and prognosis in chronic lymphocytic leukemia. Lancet 363: 105-111, 2004. Ottaggio L, Viaggi S, Zunino A, et al: Chromosome aberrations evaluated by comparative genomic hybridization in B-cell chronic lymphocytic leukemia: correlation with CD38 expression. Haematologica 88: 769-777, 2003. Que TH, Marco JG, Ellis J, et al: Trisomy 12 in chronic lymphocytic leukemia detected by fluorescence in situ hybridization: analysis by stage, immunophenotype and morphology. Blood 82: 571-575, 1993. Rosenwald A, Alizadeh AA, Widhopf G, et al: Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med 194: 1625- 1638, 2001. Rosenwald A, Chuang EY, Davis RE, et al: Fludarabine treatment of patients with chronic lymphocytic leukemia induces p53 dependent gene expression response. Blood 104: 1428- 1434, 2004. Schwaenen C, Nessling M, Wessendorf S, et al: Automated array-based genomic profiling in chronic lymphocytic leukemia: development of a clinical tool and discovery of recurrent genomic alterations. Proc Natl Acad Sci USA 101: 1039- 1044, 2004. Stankovic T, Stewart GS, Byrd P, et al: ATM mutations in sporadic lymphoid tumors. Leuk Lymphoma 42: 1563-1571, 2002. Stankovic T, Hubank M, Cronin D, et al: Microarray analysis reveals that TP-53 and ATM-mutant B-CLLs share a defect in activating proapoptotic responses after DNA damage but are distinguished by major differences in activating prosurvival responses. Blood 103: 291-300, 2004. Timar B, Fulop Z, Csernus B, et al: Relationship between the mutational status of VH genes and the pathogenesis of diffuse large B-cell lymphoma in Richter’s syndrome. Leukemia 18: 326-30, 2004. Tsimberiodu AM, Keating MJ: Richter syndrome. Biology, incidence and therapeutic strategies. Cancer 103: 216-228, 2004. Thornton PD, Gruszka-Westwood AM, Hamoudi RA, et al: Characterization of TP53 abnormalities in chronic lymphocytic leukemia. Hematol J 5: 47-54, 2004. Valganon M, Giraldo P, Agirre X, et al: P53 aberrations do not predict individual response to fludarabine in patients with Bcell chronic lymphocytic leukaemia in advanced stages Rai III/IV. Br J Haematol 129: 53-9, 2005. Wiestner A, Rosenwald A, Barry TS, et al: ZAP-70 expression identifies a chronic lymphocytic leukemia subtype with unmutated immunoglobulin genes, inferior clinical outcome and distinct gene expression profile. Blood 101: 4944-4951, 2003. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2005 VL 11 IS 4 BP 205 EP 210 PG 6 ER PT J AU Naghibalhossaini, F Pakdel, A Ghaderi, AA Saberi Firoozi, M AF Naghibalhossaini, Fakhraddin Pakdel, Abbas Ghaderi, Ali Abbas Saberi Firoozi, Mehdi TI Effective Production of Carcinoembryonic Antigen by Conversion of the Membrane-bound Into a Recombinant Secretory Protein by Site-specific Mutagenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CEA; GPI; site-specific mutagenesis; immunotherapy ID CEA; GPI; site-specific mutagenesis; immunotherapy AB Carcinoembryonic antigen (CEA), the most widely used human tumor marker, is a heavily glycosylated protein over-expressed by a wide range of tumors. It has been indicated that CEA might be a useful target for human anti-tumor immunotherapy. CEA assay for research as well as clinical trials demands a continuous source of CEA protein preparations. In a multi-purpose research program to provide a reliable source for large production of CEA, we converted the membrane-bound carcinoembryonic antigen into a secretory protein by site-specific mutagenesis. We made the secretory CEA protein by introducing a new stop codon at 99 bp upstream of the original stop codon in CEA cDNA by PCR-based mutagenesis. The glycosylation of recombinant CEA proteins, especially those destined for administration to human trials is crucially important. To produce CEA with the same glycosylation pattern and immunogenicity as the native CEA expressed by human tumors in vivo, the truncated CEA cDNA which does not encode the last C-terminal 33-amino acid hydrophobic domain was transfected into HT29, a human colon carcinoma cell line by the calcium phosphate method. Stable transfectants were selected and pooled. CEA secretion from the cells was verified by analysis of the transfectant culture supernatant for CEA protein. As determined by ELISA, 16 µg/L of recombinant CEA was secreted per 106 transfectants within 48 hrs, an increase over 40 times relative to the untransfected cells. The size of the recombinant CEA secreted by HT29 transfectants in our experiment is identical to that of reference CEA secreted from tumors and is fully antigenic. It seems that the C-terminal truncation does not affect CEA glycosylation in HT29 cells. It is predicted that human cancer immunotherapy using recombinant CEA expressed in this system would be more effective than the commercial protein which is usually prepared from bacterial or other heterologous expression systems. C1 [Naghibalhossaini, Fakhraddin] Shiraz University of Medical Sciences, Department of Biochemistry, P.O. Box 1167, 71345 Shiraz, Iran. [Pakdel, Abbas] Shiraz University of Medical Sciences, Department of Biochemistry, P.O. Box 1167, 71345 Shiraz, Iran. [Ghaderi, Ali Abbas] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Saberi Firoozi, Mehdi] Shiraz University of Medical Sciences, Gastroenterohepatology Research CenterShiraz, Iran. RP Naghibalhossaini, F (reprint author), Shiraz University of Medical Sciences, Department of Biochemistry, 71345 Shiraz, Iran. EM fakhraddin.naghibalhossaini@elf.mcgill.ca CR Benchimol S, Fuks A, Jothy S, et al: Carcinoembryonic antigen, a human tumor marker, functions as an intercellular adhesion molecule. Cell 57: 327-334, 1989. Berinstein NL: Carcinoembryonic antigen as a target for therapeutic anticancer vaccines: a review. J Clin Oncol 20: 2197- 2207, 2002. Bohme U, Cross GA: Mutational analysis of the variant surface glycoprotein GPI-anchor signal sequence in Trypanosoma brucei. J Cell Sci 115: 805-816, 2002. Chowdhury S, Chester KA, Bridgewater J, et al: Efficient retroviral vector targeting of carcinoembryonic antigen positive tumors. Mol Ther 9: 85-92, 2004. Ford CH, Macdonald F, Griffin JA, et al: Immunoadsorbent purification of carcinoembryonic antigen using a monoclonal antibody: a direct comparison with a conventional method. Tumour Biol 8: 241-250, 1987. Garcia M, Seigner C, Bastid C, et al: Carcinoembryonic antigen has a different molecular weight in normal colon and in cancer cells due to N-glycosylation differences. Cancer Res 51: 5679-5686, 1991. Gouin E, Ouary M, Pogu S, et al: Release of carcinoembryonic antigen from human tumor cells by phosphatidylinositol-specific phospholipase C: highly effective extraction and upregulation from LS-174T colonic adenocarcinoma cells. Arch Biochem Biophys 306: 125-132, 1993. Hefta LJ, Schrewe H, Thompson JA, et al: Expression of complementary DNA and genomic clones for carcinoembryonic antigen and nonspecific cross-reacting antigen in Chinese hamster ovary and mouse fibroblast cells and characterization of the membrane-expressed products. Cancer Res 50: 2397-2403, 1990. Hefta SA, Hefta LJ, Lee TD, et al: Carcinoembryonic antigen is anchored to membranes by covalent attachment to a glycosylphosphatidylinositol moiety: identification of the ethanolamine linkage site. Proc Natl Acad Sci USA 85: 4648-4652, 1988. Ikeda S, Kuroki M, Haruno M, et al: Epitope mapping of the carcinoembryonic antigen with various related recombinant proteins expressed in Chinese hamster ovary cells and 25 distinct monoclonal antibodies. Mol Immunol 29:229-240, 1992. Ilantzis C, DeMarte L, Screaton RA, et al: Deregulated expression of the human tumor marker CEA and CEA family member CEACAM6 disrupts tissue architecture and blocks colonocyte differentiation. Neoplasia 4: 151-163, 2002. Jessup JM, Ishii S, Mitzoi T, et al: Carcinoembryonic antigen facilitates experimental metastasis through a mechanism that does not involve adhesion to liver cells. Clin Exp Metastasis 17:481-488, 1999. Jessup JM, Petrick AT, Toth CA, et al: Carcinoembryonic antigen: enhancement of liver colonisation through retention of human colorectal carcinoma cells. Br J Cancer 67: 464-470, 1993. Keep PA, Leake BA, Rogers GT: Extraction of CEA from tumour tissue, foetal colon and patients’ sera, and the effect of perchloric acid. Br J Cancer 37: 171-189, 1978. Liu KJ, Wang CC, Chen LT, et al: Generation of carcinoembryonic antigen, CEA)-specific T-cell responses in HLA-A*0201 and HLA-A*2402 late-stage colorectal cancer patients after vaccination with dendritic cells loaded with CEA peptides. Clin Cancer Res 10: 2645-2651, 2004. Kuroki M, Murakami M, Wakisaka M, et al: Immunoreactivity of recombinant carcinoembryonic antigen proteins expressed in Escherichia coli. Immunol Invest 21: 241-257, 1992. Kuroki M, Kuwahara M, Tsuruta Y, et al: Effective purification of nonspecific cross-reacting antigens with phosphatidylinositol- specific phospholipase C. Prep Biochem 23: 333-349, 1993. Laemmli UK: Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227: 680-685, 1970. Leconte A, Garambois V, Ychou M, et al: Involvement of circulating CEA in liver metastases from colorectal cancers reexamined in a new experimental model. Br J Cancer 80: 1373- 1379, 1999. Matsuoka Y, Matsuo Y, Okamoto N, et al: Highly effective extraction of carcinoembryonic antigen with phosphatidylinositol- specific phospholipase C. Tumour Biol 12: 91-98, 1991. Mayer A, Chester KA, Flynn AA, Begent RH: Taking engineered anti-CEA antibodies to the clinic. J Immunol Methods 231:261-273, 1999. Nap M, Mollgard K, Burtin P, et al: Immunohistochemistry of carcino-embryonic antigen in the embryo, fetus and adult. Tumour Biol 9: 145-153, 1988. Rudd PM, Wormald MR, Stanfield RL, et al: Roles for glycosylation of cell surface receptors involved in cellular immune recognition. J Mol Biol 293: 351-366, 1999. Samanci A, Yi Q, Fagerberg J, et al: Pharmacological administration of granulocyte/macrophage-colony-stimulating factor is of significant importance for the induction of a strong humoral and cellular response in patients immunized with recombinant carcinoembryonic antigen. Cancer Immunol Immunother 47: 131-142, 1998. Sarobe P, Huarte E, Lasarte JJ, et al: Carcinoembryonic antigen as a target to induce anti-tumor immune responses, Review. Curr Cancer Drug Targets 4: 443-454, 2004. Shively JE, Beatty JD: CEA-related antigens: molecular biology and clinical significance. Crit Rev Oncol Hematol 2: 355- 399, 1985. Terskikh A, Mach JP, Pelegrin A: Marked increase in the secretion of a fully antigenic recombinant carcinoembryonic antigen obtained by deletion of its hydrophobic tail. Mol Immunol 30: 921-927, 1993. Udenfriend S, Micanovic R, Kodukula K: Structural requirements of a nascent protein for processing to a PI-G anchored form: studies in intact cells and cell-free systems. Cell Biol Int Rep 15: 739-759, 1991. Udenfriend S, Kodukula K: How glycosylphosphatidylinositolanchored membrane proteins are made. Annu Rev Biochem 64: 563-591, 1995. Ullenhag GJ, Frodin JE, Jeddi-Tehrani M, et al: Durable carcinoembryonic antigen, CEA)-specific humoral and cellular immune responses in colorectal carcinoma patients vaccinated with recombinant CEA and granulocyte/macrophage colonystimulating factor. Clin Cancer Res 10: 3273-3281, 2004. Witherspoon LR, Shuler SE, Alyea K, et al: Carcinoembryonic antigen: assay following heat compared with perchloric acid extraction in patients with colon cancer, non-neoplastic gastrointestinal diseases, or chronic renal failure. J Nucl Med 24:916-921, 1983. Wong JY, Chu DZ, Williams LE, et al: Pilot trial evaluating an 123I-labeled 80-kilodalton engineered anticarcinoembryonic antigen antibody fragment, cT84.66 minibody, in patients with colorectal cancer. Clin Cancer Res 10: 5014-5021, 2004. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2005 VL 11 IS 4 BP 211 EP 217 PG 7 ER PT J AU Raso, E Tovari, J Ladanyi, A Varga, N Timar, J AF Raso, Erzsebet Tovari, Jozsef Ladanyi, Andrea Varga, Norbert Timar, Jozsef TI Ligand-Mimetic Anti-αIIbβ3 Antibody PAC-1 Inhibits Tyrosine Signaling, Proliferation and Lung Colonization of Melanoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ?IIbß3; integrin; ligand-mimetic; melanoma ID ?IIbß3; integrin; ligand-mimetic; melanoma AB β3 integrin expression is the hallmark of melanoma and may serve as a potential therapeutic target. While αvβ3 integrin expression seems to be constitutive in melanoma, ectopic expression of platelet-αIIbβ3 is dependent on progression. B16a murine melanoma is a suitable model for studies on αIIbβ3 treatment strategies since αvβ3 is not expressed in this cell line. Here we have used a ligand-mimetic anti-αIIbβ3 monoclonal antibody, PAC-1, to test the biological consequences of αIIbβ3 modulation in melanoma cells. We have previously reported that in B16a cells FAK is constitutively active and tyrosine-phosphorylated. Upon PAC-1 binding to the surface αIIbβ3, which is in the active conformation, FAK became dephosphorylated through a process of PKC-dependent phosphatase activation. Furthermore, PAC-1 binding to B16a cells induced a significant decrease in phosphotyrosine-positive melanoma cells within 30 min. Treatment of B16a cells in vitro with PAC-1 significantly inhibited proliferation by decreasing the mitotic index but not affecting apoptotic rate. Incubation of B16a cells with PAC-1 decreased their lung colonization potential, suggesting a profound alteration in their biological behavior under the effect of this antibody. These preclinical data suggest that the ectopic expression of αIIbβ3 in melanoma cells can be exploited as a novel target of antibody therapy of melanoma. C1 [Raso, Erzsebet] National Institute of Oncology, Department of Tumor Progression, Rath Gy. u. 7-9., H-1122 Budapest, Hungary. [Tovari, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gy. u. 7-9., H-1122 Budapest, Hungary. [Ladanyi, Andrea] National Institute of Oncology, Department of Tumor Progression, Rath Gy. u. 7-9., H-1122 Budapest, Hungary. [Varga, Norbert] National Institute of Oncology, Department of Tumor Progression, Rath Gy. u. 7-9., H-1122 Budapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gy. u. 7-9., H-1122 Budapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. CR Hood JD and Cheresh DA: Role of integrins in cell invasion and migration. Nat Rev Cancer 2: 91-100, 2002 Hin J and Varner J: Integrins: roles in cancer development and as treatment targets. Br J Cancer 90: 561-565, 2005 Danen EH: Integrins: regulators of tissue function and cancer progression. Curr Pharm Des 11: 881-891, 2005 Guo W and Giancotti FG: Integrin signaling during tumour progression. Nat Rev Mol Cell Biol 5: 816-826, 2004 Goel HL and Languino LR: Integrin signaling in cancer. Cancer Treat Res 119: 15-31, 2004 Kuphal S, Bauer R and Rosserhoff AK: Integrin signaling in malignant melanoma. Cancer Metast Rev 24:195-222, 2005 Albelda SM, Mette SA, Elder DE, et al: Integrin distribution in malignant melanoma: Association of the a3 subunit with tumor progression. Cancer Res 50: 6757-6774, 1990 Puerschel WC, Gawaz M, Worret W-I and Ring J: Immunoreactivity of glycoprotein IIb is present in metastasized but not in nonmetastasized primary malignant melanoma. Br J Dermatol 135: 883-887, 1996 Trikha M, Timar J, Lundy SK, et al: The high affinity αIIbβ3 integrin is involved in invasion of human melanoma cells. Cancer Res 57: 2522-2528, 1997 Trikha M, Timar J, Zacharek A, et al: Role for β3 integrins in human melanoma growth and survival. Int J Cancer 101: 156-167, 2002 Timar J, Trikha M, Szekeres K, et al: Expression and function of the high affinity αIIbβ3 integrin in murine melanoma cells. Clin Exp Metast 16: 437-455, 1998 Raso E, Tovari J, Toth K, et al: Ectopic αIIbβ3 integrin signaling involves 12-lipoxygenase and PKC mediated serine phosphorylation events in melanoma cells. Thromb Haemost 85: 1037-1042, 2001 Dome B, Raso E, Dobos J, et al. Parallel expression of αIIbβ3 and αvβ3 integrins in human melanoma cells upregulates bFGF expression and promotes their angiogenic phenotype. Int J Cancer 116: 27- 35, 2005 Mikecz K: Vitaxin applied molecular evolution. Curr Opin Investig Drugs 1: 199-203, 2000 Gutheil JC, Campbell TN, Pierce PR, et al: Targeted antiangiogenic therapy for cancer using vitaxin: a humanized monoclonal antibody to the integrin αvβ3. Clin Cancer Res 6: 3056-3061, 2000 Tucker GC: Alpha v integrin inhibitors and cancer therapy. Curr Opin Investig Drugs 4: 722-731, 2003 Tam SH, Sassoli PH, Jordan RE and Nakada MT: Abciximab, Reo- Pro, Chimeric 7E3 Fab, demonstrates equivalent affinity and functional blockade of glycoprotein IIb/IIa and αvβ3 integrins. Circulation 97: 1085-1091, 1998 Cohen SA, Trikha M and Mascelli MA: Potential future clinical applications for the GPIIb/IIIa antagonist, abciximab in thrombosis, vascular and oncological indications. Pathol Oncol Res 6: 163-174, 2000 Trikha M, Zhou Z, Timar J, et al: Multiple roles for platelet GPIIb/IIIa and αvβ3 integrins in tumor growth, angiogenesis, and metastasis. Cancer Res 62:2824-2833, 2002 Harms JF, Welch DR, Samant RS, et al: A small molecule antagonist of the αvβ3 integrin suppresses MDA-MB-435 skeletal metastasis. Clin Exp Metast 21: 119-128, 2004 Carron CP, Meyer DM, Englemann WW, et al: Peptidomimetic antagonists of avb3 inhibit bone resorption by inhibiting osteoclast bone resorptive activity, not osteoclast adhesion to bone. J Endocrinol 165: 587-598, 2000 Engleman VW, Nickols GA, Ross FP, et al: A peptidomimetic antagonist of the αvβ3 integrin inhibits bone resorption in vitro and prevents osteoporosis in vivo. J Clin Invest 99: 2284-2292, 1997 Reinmuth N, Liu WB, Ahmad SA, et al: αvβ3 integrin antagonist S247 decreases colon cancer metastasis and angiogenesis and improves survival in mice. Cancer Res 63:2079-2087, 2003 Gomes N, Vassy J, Lebos C, et al: Breast adenocarcinoma cell adhesion to the vascular subendothelium in whole blood and under flow conditions: Effects of αvβ3 and αIIbβ3 antagonists. Clin Exp Metast 21: 553-561, 2004 Amirkhosravi A, Mousa SA, Amaya M, et al: Inhibition of tumor cellinduced platelet aggregation and lung metastasis by the oral GpIIb/IIIa antagonist XV454. Thromb Haemost 90: 549-554, 2003 Shattil SJ, Hoxie JA, Cunningham M and Brass LF: Changes in the platelet membrane glycoprotein IIb.IIIa complex during platelet activation. J Biol Chem 260: 11107-11114, 1985 Hubert P, Grenot P, Autran B and Debre P: Analysis by flow cytometry of tyrosine-phosphorylated proteins in activated T-cell subsets on whole blood samples. Cytometry 29: 83-91, 1997 Shattil SJ, Ginsberg MH and Brugge JS: Adhesive signaling in platelets. Curr Opin Cell Biol 6: 695-704, 1994 Gao J, Zoller KE, Ginsberg MH, et al: Regulation of the pp72syk protein tyrosine kinase by platelet integrin alpha IIb beta 3. EMBO J 16: 6414-6425, 1997 Law DA, Nannizzi-Aleimo L and Phillips DR: Outside-in integrin signal transduction. Alpha IIb beta 3-(GP IIb IIIa, tyrosine phosphorylation induced by platelet aggregation. J Biol Chem 271: 10811- 10815, 1996 Hughes PE, Diaz-Gonzalez F, Leong L, et al. Breaking the integrin hinge. J Biol Chem 271:6571-6574, 1996 Honn KV, Timar J: Integrin expression in tumor progression role of signaling mechanisms. In: Tumor matrix biology., Ed. R. Adany), CRC Press, Boca Raton, 1995, pp 145-171 Carman CV and Springer TA: Integrin avidity regulation: are changes in affinity and conformation underemphasized? Curr Opin Cell Biol 15: 547-556, 2003 Schwarz M, Katagiri Y, Kotani M, et al: Reversibility versus persistence of GPIIb/IIIa blocker-induced conformational change of GPIIb/IIIa, αIIbβ3, CD41/CD61). J Pharmacol Exp Ther 308: 1002- 1011, 2004 Timar J, Tovari J, Raso E, et al. Platelet-mimicry of cancer cells: epiphenomenon with clinical significance. Oncology 69: 185-201, 2005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2005 VL 11 IS 4 BP 218 EP 223 PG 6 ER PT J AU Yilmaz, Z Sahin, IF Atalay, B Ozen, Caner, H Bavbek, M Demirhan, B Altinors, N AF Yilmaz, Zerrin Sahin, Iffet Feride Atalay, Basar Ozen, Ozlem Caner, Hakan Bavbek, Murad Demirhan, Beyhan Altinors, Nur TI Chromosome 1p36 and 22qter Deletions in Paraffin Block Sections of Intracranial Meningiomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE meningioma; 22q; 1p36; fluorescence in situ hybridization; grading ID meningioma; 22q; 1p36; fluorescence in situ hybridization; grading AB Meningiomas are the most frequent benign tumors of the intracranial cavity. The classification and underlying pathogenetic mechanisms have been reported to be investigated by both pathological and genetic methods. In this study, we aimed to detect 1p36 and 22qter deletions by fluorescence in situ hybridization (FISH) in archival materials of 50 intracranial meningioma patients. The clinical material consisted of paraffin-embedded tissue sections from 50 patients who were surgically treated and had histopathologic diagnosis of an intracranial meningioma. We observed 1p36 deletion in 23/50 (46%) and 22qter deletion in 33/50 (66%) patients. In addition, we observed 22qter deletion in 26/36 (72.2%) patients with meningothelial meningioma. This finding implies that 22qter deletion might play an important role in the pathogenesis of meningothelial meningioma. On the other hand, no alterations were documented in the frequency of these chromosomal alterations according to the grade of meningiomas, suggesting that malignant progression of these tumors depends on other, more relevant, genetic changes. C1 [Yilmaz, Zerrin] Baskent University Faculty of Medicine, Department of Medical Biology and Genetics, Kubilay Sokak No: 36 06570-MaltepeAnkara, Turkey. [Sahin, Iffet Feride] Baskent University Faculty of Medicine, Department of Medical Biology and Genetics, Kubilay Sokak No: 36 06570-MaltepeAnkara, Turkey. [Atalay, Basar] Baskent University Faculty of Medicine, Department of NeurosurgeryAnkara, Turkey. [Ozen, Ozlem] Baskent University Faculty of Medicine, Department of PathologyAnkara, Turkey. [Caner, Hakan] Baskent University Faculty of Medicine, Department of NeurosurgeryAnkara, Turkey. [Bavbek, Murad] Baskent University Faculty of Medicine, Department of NeurosurgeryAnkara, Turkey. [Demirhan, Beyhan] Baskent University Faculty of Medicine, Department of PathologyAnkara, Turkey. [Altinors, Nur] Baskent University Faculty of Medicine, Department of NeurosurgeryAnkara, Turkey. RP Sahin, IF (reprint author), Baskent University Faculty of Medicine, Department of Medical Biology and Genetics, Ankara, Turkey. EM feridesahin@hotmail.com CR Bello MJ, de Campos JM, Vaquero J et al: High-resolution analysis of chromosome arm 1p alterations in meningioma. Cancer Genet Cytogenet 120: 30-36, 2000 Bostrom J, Muhlbauer A, Reifenberger G: Deletion mapping of the short arm of chromosome 1 identifies a common region of deletion distal to D1S496 in human meningiomas. Acta Neuropathol 94: 479-485, 1997 Cerda-Nicolas M, Lopez-Ginez C, Perez-Bacete M, et al: Histologically benign metastatic meningioma: morphological and cytogenetic study. J Neurosurg 98: 194-198, 2003 Dumanski JP, Carlbom E, Collins VP, Nordenskjold M: Deletion mapping of a locus on human chromosome 22 involved in the oncogenesis of meningioma. Proc Natl Acad Sci USA 84: 9275-9279, 1987 Durmaz R, Arslantaº A, Artan S, et al: The deletion of 22q13 region in both intracranial and spinal meningiomas in a patient, case report). Clin Neurol Neurosurg 100: 219-223, 1998 Freiler A, Zang KD: Monosomy 7p in meningiomas: a rare constituent of tumor progression. Cancer Genet Cytogenet 144:65-68, 2003 Ishino S, Hashimoto N, Fushiki S, et al: Loss of material from chromosome arm 1p during malignant progression of meningioma revealed by fluorescent in situ hybridization. Cancer 83: 360-366, 1998 Khan J, Parsa NZ, Harada T, et al: Detection of gains and losses in 18 meningiomas by comparative genomic hybridization. Cancer Genet Cytogenet 103:95-100, 1998 Kleihues P, Louis DN, Scheithauer BW et al: The WHO classification of tumors of the nervous system. J Neuropathol Exp Neurol 61: 215-225, 2002 Lamszus K, Kluwe L, Matschke J, et al: Allelic losses at 1p, 9q, 10q, 14q, and 22q in the progression of aggressive meningiomas and undifferentiated meningeal sarcomas. Cancer Genet Cytogenet 110:103-110, 1999 Lomas J, Bello J, Arjona D, et al: Analysis of p73 gene in meningiomas with deletion at 1p. Cancer Genet Cytogenet 129: 88-91, 2001 Lopez-Gines C, Cerda-Nicolas M, Gil-Benso R, et al: Association of loss of 1p and alterations of chromosome 14 in meningioma progression. Cancer Genet Cytogenet 148: 123-128, 2004 Lopez-Gines C, Cerda-Nicolas M, Gil-Benso R, et al: Loss of 1p in recurrent meningiomas: a comparative study in successive recurrences by cytogenetics and fluorescence in situ hybridization. Cancer Genet Cytogenet 125:119-124, 2001 Louis DN, Scheithauer BW, Budka H, et al: Meningiomas. In: WHO Classification of Tumors. Pathology & Genetics Tumours of the Nervous System., Eds: Kleihues P, Cavenee WK), IARC Press , Lyon, 2000 Martin AJ, Summersgill BM, Fisher C, et al: Chromosomal imbalances in meningeal solitary fibrous tumors. Cancer Genet Cytogenet 135:160-164, 2002 Murakami M, Hashimoto N, Takahashi Y, et al: A consistent region of deletion on 1p36 in meningiomas: identification and relation to malignant progression. Cancer Genet Cytogenet 140:99-106, 2003 Perry A, Stafford SL, Schithauer BW, et al: The prognostic significance of MIB-1, p53, and DNA flow cytometry in completely resected primary meningiomas. Cancer 82: 2262- 2269, 1998 Perry A, Jenkins RB, Dahl RJ, et al: Cytogenetic analysis of aggressive meningiomas. Possible diagnostic and prognostic implications. Cancer 77: 2567-2573, 1996 Sawyer JR, Husain M, Lukacs JL, et al: Telomeric function as a mechanism for the loss of 1p in meningioma. Cancer Genet Cytogenet 145:38-48, 2003 Sayagues JM, Tabernero MD, Diaz P, et al: Incidence of numerical chromosome aberration in meningioma tumors as revealed by fluorescence in situ hybridization using 10 chromosome- specific probes. Cytometry, Clinical Cytometry, 50:153-159, 2002 Simeone A: Detection of m-RNA in tissue sections with radiolabelled riboprobes. In: In Situ Hybridization; A Practical Approach. Second ed., Ed: Wilkison DG), Oxford University Press, New York, 1999, pp. 70-86. Weber RG, Bostrom J, Wolter M, et al: Analysis of genomic alterations in benign, atypical, and anaplastic meningiomas: Toward a genetic model of meningioma progression. Proc Natl Acad Sci USA 94: 14719-14724, 1997 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2005 VL 11 IS 4 BP 224 EP 228 PG 5 ER PT J AU Behrem, S Zarkovic, K Eskinja, N Jonjic, N AF Behrem, Senija Zarkovic, Kamelija Eskinja, Neven Jonjic, Nives TI Distribution Pattern of Tenascin-C in Glioblastoma: Correlation with Angiogenesis and Tumor Cell Proliferation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tenascin; glioblastoma multiforme; angiogenesis; proliferation ID tenascin; glioblastoma multiforme; angiogenesis; proliferation AB Tenascin-C (TN-C) is an extracellular matrix protein which participates in different processes like normal fetal development, wound healing, inflammation, keloids and rheumatoid arthritis. Furthermore, the immunostaining for TN-C is seen in the stroma of various malignant tumors as in glioblastoma multiforme (GBM), however, the significance of these findings is still not clear. In this study 62 GBM samples were analyzed immunohistochemically for distribution patterns of TN-C and correlated with angiogenesis and tumor cell proliferation. Tenascin-C in GBM localizes in two compartments, perivascular and intercellular space. Intercellular tenascin-C (TN-C ic) showed focal distribution in 66%, and diffuse one in 34% of cases. Perivascular tenascin-C (TN-C pv) showed strong correlation with microvascular density (MVD) and vascular endothelial growth factor (VEGF) expression. Moreover, it seems that TN-C pv enhanced the effect of VEGF. Intercellular TN-C did not correlate with MVD and VEGF expression, but showed strong correlation with proliferation index. Furthermore, tumors with diffuse TN-C ic expression had higher proliferation indices than tumors with focal TN-C expression. Our results indicate that TN-C plays a role in angiogenesis and tumor cell proliferation, but beside the intensity of expression, the distribution patterns are also important in these processes. This study also suggests that perivascular and intercellular TN-C compartments have probably different sources and different roles in GBM. C1 [Behrem, Senija] Rijeka University School of Medicine, Department of Pathology, Braaee Branchetta 20, 51 000 Rijeka, Croatia. [Zarkovic, Kamelija] Clinical Medical Center, Department of NeuropathologyZagreb, Croatia. [Eskinja, Neven] Clinical Medical Center Rijeka, Department of NeurosurgeryRijeka, Croatia. [Jonjic, Nives] Rijeka University School of Medicine, Department of Pathology, Braaee Branchetta 20, 51 000 Rijeka, Croatia. RP Jonjic, N (reprint author), Rijeka University School of Medicine, Department of Pathology, 51 000 Rijeka, Croatia. EM nives@medri.hr CR Brooks PC, Dark RA, Cheresh DA: Requirement of vascular integrin alfa v beta 3 for angiogenesis. Science 264: 569-571, 1994 Castellani P, Siri A, Zardi L, et al: Distribution of tenascin in human gliomas is not related to cell proliferation. J Neurol Neurosurg Psychiatry 62: 290-291, 1997 Castellon R, Caballero S, Hamdi KH, et al: Effects of tenascin- C on normal and diabetic retinal endothelial cells in culture. Investigative Ophthalmol Vis Sci 43: 2758-2766, 2002 Chintala SK, Rao JS: Invasion in human glioma: role of extracellular matrix. Front Biosci 11: 324-339, 1996 Chiquet-Ehrismann R, Mackie EJ, Pearson CA, Sakakura T: An extracellular matrix protein involved in tissue interactions during fetal development and oncogenesis. Cell 147: 131-139, 1986 Crossin KL: Cytotactin binding: inhibition of stimulated proliferation and intracellular alkalinization in fibroblasts. Proc Natl Acad Sci USA 88: 11403-11407, 1991 Ehrismann RC, Chiquet M: Tenascins: regulation and putative functions during pathological stress. J Pathol 200: 488-499, 2003 Fischer D, Brown-Ludi M, Schulthess T, Chiquet-Ehrismann R: Concerted action of tenascin-C domains in cell adhesion, antiadhesion and promotion of neurite outgrowth. J Cell Sci 110: 1513-1522, 1997 Giese A, Berens ME, Loo MA, et al: Contrasting migratory response of astrocytoma cells to tenascin mediated by different integrins. J Cell Sci 109:2161-2168, 1996 Gladson CL: The extracellular matrix of gliomas: modulation of cell function. J Neuropathol Exp Neurol 58: 1029-1040, 1999 Herold-Mende, Mueller MM, Bonsanto MM, et al: Clinical impact and functional aspects of tenascin-c expression during glioma progression. Int J Cancer 98: 362-369, 2002 Higuchi M, Ohnishi T, Arita N, et al: Expression of tenascin in human gliomas: its relation to histological malignancy, tumor dedifferentiation and angiogenesis. Acta Neuropathol 85: 481- 487, 1993 Huang W, Chiquet-Ehrismann R, Moyano V, et al: Interference of tenascin-C with syndecan-4 binding to fibronectin blocks cell adhesion and stimulates tumor cell proliferation. Cancer Res 61: 8586-8594, 2001 Ishihara A, Yoshida T, Tamaki H, Sakakukra T: Tenascin expression in cancer cells and stroma of human breast cancer and its prognostic significance. Clin Cancer Res 1: 1035-1041, 1995 Kim CH, Bak KH, Kim JM, et al: Expression of tenascin-C in astrocytic tumors: its relevance to proliferation and angiogenesis. Surg Neurol 54: 235-240, 2000 Kumar P, Wang J, Bernabeu C: CD105 and angiogenesis. J Pathol 178:363-366, 1996 Kumar S, Ghellal A, Li C, et al: Vascular density determined using CD105 antibody correlates with tumor prognosis. Cancer Res 59: 856-861, 1999 Lotz MM, Burdsal CA, Erickson HP, Mc Clay DR: Cell adhesion to fibronectin and tenascin: quantitative measurements of initial binding and subsequent strengthening response. J Cell Biol 109: 1795-1805, 1989 Mai J, Sameni M, Mikkelsen T, Sloane BF: Degradation of extracellular matrix protein tenascin-C by cathepsin B: an interaction involved in the progression of gliomas. Biol Chem 383: 1407-1413, 2002 McLendon RE, Wikstrand J, Matthews Mr, et al: Glioma associated antigen expression in oligodendroglial neoplasms: tenascin and epidermal growth factor receptor. J Histochem Cytochem 48: 1103-1110, 2000 Orend G, Huang W, Olayioye MA, et al: Tenascin-C blocks cellcycle progression of anchorage-dependent fibroblasts on fibronectin through inhibition of syndecan-4. Oncogene 22: 3917-3926, 2003 Pilch H, Schaffer U, Schlenger K, et al: Expression of tenascin in human cervical cancer: association of tenascin expression with clinicopathological parameters. Gynecol Oncol 73: 415- 421, 1999 Roijani AM, Dorovini-Zis K: Glomeruloid vascular structures in glioblastoma multiforme: an immunohistochemical and ultrastructural study. J Neurosurg 85:1078-1084, 1996 Scheetz FJ, LLoyd PJ: The tenascin family of ECM glycoproteins: structure, function, and regulation during embryonic development and tissue remodeling. Developmental Dynamics 218: 235-259, 2000 Schenk S, Chuquet-Ehrismann R, Battegay EJ: The fibrinogen globe of tenascin-C promotes basic fibroblast growth factorinduced endothelial cell elongation. Mol Biol Cell 10: 2933- 2943, 1999 Tanaka F, Yanagihara OK, Kawano Y, et al: Evaluation of angiogenesis in non-small cell lung cancer: comparison between anti-CD34 antibody and anti-CD105 antibody. Clin Cancer Res 7: 3410-3415, 2001 Tanaka K, Hiraiwa N, Hashimoto H, et al: Tenascin -C regulates angiogenesis in tumor through the regulation of vascular endothelial growth factor expression. Int J Cancer 108: 31-40, 2004 Uhm JH, Gladson LC, Rao JS: The role of integrins in the malignant phenotype of gliomas. Front Biosci 4: 188-199, 1999 Ventimiglia JB, Wikstrand CJ, Ostrowski LE, et al: Tenascin expression in human glioma cell lines and normal tissues. J Neuroimmunol. 36:41-45, 1992 Vitolo D, Paradiso P, Uccini S, et al: Expression of adhesion molecules and extracellular matrix in glioblastomas: relation to angiogenesis and spread. Histopathology 28: 521- 528, 1996 Wehrle-Haller B, Chiquet M: Dual function of tenascin: simultaneous promotion of neural growth and inhibition of glial migration. J Cell Sci 106: 597-610, 1993 Yoshyida T, Yoshimura E, Numata H, et al: Involvement of tenascin-C on proliferation and migration of laryngeal carcinoma cells. Virchows Arch 435: 496-500, 1993 Zagzag D, Friedlander DR, Miller DC, et al: Tenascin expression in astrocytomas correlates with angiogenesis. Cancer Res 55: 907-914, 1995 Zagzag D, Capo V: Angiogenesis in the central nervous system: a role for endothelial growth factor/vascular permeability factor and tenascin-C, common molecular effectors in cerebral neoplastic and non-neoplastic “angiogenic diseases”. Histol Histopathol 17: 301-321, 2002 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2005 VL 11 IS 4 BP 229 EP 235 PG 7 ER PT J AU Ghayumi, ASM Mehrabi, S Doroudchi, M Ghaderi, A AF Ghayumi, Ali Sied Mohammad Mehrabi, Samrad Doroudchi, Mehrnossh Ghaderi, Abbas TI Diagnostic Value of Tumor Markers for Differentiating Malignant and Benign Pleural Effusions of Iranian Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE pleural effusion; tumor markers ID pleural effusion; tumor markers AB In order to evaluate the diagnostic yield of tumor markers in differentiating malignant and benign pleural effusions, we carried out a prospective study in a group of Iranian people. Pleural and serum levels of carcinoembryonic antigen (CEA), carbohydrate antigen 15-3 (CA 15-3), neuron-specific enolase (NSE) and cancer antigen 125 (CA 125) were assayed prospectively in patients with pleural effusion (40 malignant and 37 benign). The highest sensitivity was obtained with a combination of CA 15-3 in serum, and CA 15-3 and CEA in pleural fluid (80%), also with combination of CA 15-3 in serum, and CA 15-3, NSE and CEA in pleural fluid (80%). The highest specificity was obtained with combination of CA 15-3 in serum, and CA 15-3 and NSE in pleural fluid (100%), and also with combination of CA 15-3 in serum, and CA15-3, NSE and CEA in pleural fluid (100%). C1 [Ghayumi, Ali Sied Mohammad] Shiraz University of Medical Sciences, Department of Internal MedicineShiraz, Iran. [Mehrabi, Samrad] Shiraz University of Medical Sciences, Department of Internal MedicineShiraz, Iran. [Doroudchi, Mehrnossh] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Ghaderi, Abbas] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. RP Ghayumi, ASM (reprint author), Shiraz University of Medical Sciences, Department of Internal Medicine, Shiraz, Iran. EM ghayyoumim@sums.ac.ir CR Chernow B, Sahn SA: Carcinomatous involvement of the pleura: an analysis of 96 patients. Am J Med 63: 695-702, 1977 Marel M, Stastny B, Melinova L, et al: Diagnosis of pleural effusions. Experience with clinical studies, 1986 to 1990. Chest 107: 1598-1603, 1995 Maskell NA, Butland RJ: Pleural Diseases Group, Standards of Care Committee, British Thoracic Society. BTS guidelines for the investigation of a unilateral pleural effusion in adults. Thorax 58, Suppl 2): 8-17, 2003 Loddenkemper R: Medical thoracoscopy. In: Textbook of Pleural Diseases., Eds: Light RW, Gray Lee, YC), 2003, pp 498-512 Prakash UB, Reiman HM: Comparison of needle biopsy with cytologic analysis for the evaluation of pleural effusion: analysis of 414 cases. Mayo Clin Proc 60:158-164, 1985 Johanston WW: Cancer, 1985; Light RW, Approach to the patient. In: Pleural Diseases., Ed: Light RW), 3rd ed, William & Wilkins, Baltimore, 1995, pp 75-82 Porcel JM, Vives M, Esquerda A, et al: Use of a panel of tumor markers, carcinoembryonic antigen, cancer antigen 125, carbohydrate antigen 15-3, and cytokeratin 19 fragments, in pleural fluid for the differential diagnosis of benign and malignant effusions. Chest 126: 1757-1763, 2004 Richard WL: Tumor markers in undiagnosed pleural effusions. Chest 126: 1721-1722, 2004 Ferrer J, Villarino MA, Encabo G, et al: Diagnostic utility of CYFRA 21-1, carcinoembryonic antigen, CA 125, neuron specific enolase, and squamous cell antigen level determinations in the serum and pleural fluid of patients with pleural effusions. Cancer 86: 1488-1495, 1999 Garcia-Pachon E, Padilla-Navas I, Dosda MD, et al: Elevated level of carcinoembryonic antigen in nonmalignant pleural effusions. Chest 111: 643-647, 1997 Alatas F, Alatas O, Metintas M, et al: Diagnostic value of CEA, CA 15-3, CA 19-9, CYFRA 21-1, NSE and TSA assay in pleural effusions. Lung Cancer 31: 9-16, 2001 Ryu JS, Lee HJ, Cho JH, et al: The implication of elevated carcinoembryonic antigen level in pleural fluid of patients with non-malignant pleural effusion. Respirology 8: 487-491, 2003 Villena V, Lopez-Encuentra A, Echave-Sustaeta, et al: Diagnostic value of CA 72-4, carcinoembryonic antigen, CA 15-3, and CA 19-9 assay in pleural fluid. A study of 207 patients. Cancer 78: 736-740, 1996 Shimokata K, Totani Y, Nakanishi K, et al: Diagnostic value of cancer antigen 15-3, CA15-3, detected by monoclonal antibodies, 115D8 and DF3, in exudative pleural effusions. Eur Respir J 4: 341-344, 1988 Romero S, Fernandez C, Arriero JM, et al: CEA, CA 15-3 and CYFRA 21-1 in serum and pleural fluid of patients with pleural effusions. Eur Respir J 9: 17-23, 1996 Hernandez L, Espasa A, Fernandez C, et al: CEA and CA 549 in serum and pleural fluid of patients with pleural effusion. Lung Cancer 36: 83-89, 2002 Pinto MM, Bernstein LH, Rudolph RA, et al: Diagnostic efficiency of carcinoembryonic antigen and CA125 in the cytological evaluation of effusions. Arch Pathol Lab Med 116: 626- 631, 1992 McKenna JM, Chandrasekhar AJ, Henkin RE: Diagnostic value of carcinoembryonic antigen in exudative pleural effusions. Chest 78: 587-590, 1980 Romero Candeira S, Hernandez Blasco L, Senent Espanol C, et al: Clinical usefulness of tumor markers in the diagnosis of pleural effusions. Carcinoembryonic antigen, alpha-fetoprotein and orosomucoid. Med Clin 86: 439-443, 1986 Garcia-Pachon E, Padilla Navas I, Querol M, Custardoy J. The behavior of carcinoembryonic antigen in pleural empyema. Med Clin 101: 237-238, 1993 Whiteside TL, Dekker A: Diagnostic significance of carcinoembryonic antigen levels in serous effusions. Correlation with cytology. Acta Cytol 23: 443-448, 1979 Niwa Y, Kishimoto H, Shimokata K: Carcinomatous and tuberculous pleural effusions. Comparison of tumor markers. Chest 87: 351-355, 1985 Faravelli B, Nosenzo M, Razzetti A, et al: The role of concurrent determinations of pleural fluid and tissue carcinoembryonic antigen in the distinction of malignant mesothelioma from metastatic pleural malignancies. Eur J Cancer Clin Oncol 21: 1083-1087, 1985 Mezger J, Calavrezos A, Drings P, et al: Value of serum and effusion fluid CEA levels for distinguishing between diffuse malignant mesothelioma and carcinomatous pleural metastases. Lung 172: 183-184, 1994 Kuralay F, Tokgoz Z, Comlekci A: Diagnostic usefulness of tumor marker levels in pleural effusions of malignant and benign origin. Clin Chim Acta 300: 43-55, 2000 Fenton KN, Richardson JD: Diagnosis and management of malignant pleural effusions. Am J Surg 170: 69-74, 1995 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2005 VL 11 IS 4 BP 236 EP 241 PG 6 ER PT J AU Ates, EL Kapran, Y Erbil, Y Barbaros, U Dizdaroglu, F AF Ates, Esberk Lora Kapran, Yersu Erbil, Yesim Barbaros, Umut Dizdaroglu, Ferhunde TI Cystic Lymphangioma of the Right Adrenal Gland SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Adrenal gland; adrenal cyst; lymphangioma ID Adrenal gland; adrenal cyst; lymphangioma AB Lymphangiomas are benign malformations of the vessels. They are commonly located in the neck, axillary region and mediastinum. Lymphangioma of the adrenal gland is very rare. These lesions were first discovered as incidental autopsy findings. As the imaging techniques have improved, they now appear as incidental findings at abdominal ultrasonography and computed tomography scan examinations. They are usually asymptomatic. We present a 26-year-old woman admitted to the hospital, complaining of weakness, putting on weight, and lumbago. Her laboratory findings were within normal limits. Radiological examination revealed a 7 cm cystic lesion located in the right adrenal gland. Right adrenalectomy was performed. Histopathological examination and immunohistochemical analysis of the cystic lesion was consistent with a lymphangioma. C1 [Ates, Esberk Lora] Istanbul Medical Faculty, Department of Pathology, Temel Bilimler Binasi, Patoloji Anabilim Dali, Capa, 34390 Istanbul, Turkey. [Kapran, Yersu] Istanbul Medical Faculty, Department of Pathology, Temel Bilimler Binasi, Patoloji Anabilim Dali, Capa, 34390 Istanbul, Turkey. [Erbil, Yesim] Istanbul Faculty of Medicine, Istanbul University, Department of SurgeryIstanbul, Turkey. [Barbaros, Umut] Istanbul Faculty of Medicine, Istanbul University, Department of SurgeryIstanbul, Turkey. [Dizdaroglu, Ferhunde] Istanbul Medical Faculty, Department of Pathology, Temel Bilimler Binasi, Patoloji Anabilim Dali, Capa, 34390 Istanbul, Turkey. RP Ates, EL (reprint author), Istanbul Medical Faculty, Department of Pathology, 34390 Istanbul, Turkey. EM loraates@hotmail.com CR Fauquenot-Nollen AM, Plaisier ML, Tjon A, Tham RT: Combined thoracic and abdominal lymphangioma in an adult. JBRBTR 85: 130-131, 2002 Gaffey MJ, Mills SE, Fechner RE, et al: Vascular adrenal cysts. A clinicopathologic and immunohistochemical study of endothelial and hemorrhagic, pseudocystic, variants. Am J Surg Pathol 13: 740-747, 1989 Katz RL, Mackay B, Zornoza J: Fine needle aspiration cytology of the adrenal gland. Acta Cytol 28: 269-282, 1984 Khoda J, Hertzanu Y, Sebbag G, et al: Adrenal cysts: diagnosis and therapeutic approach. Int Surg 78: 239-242, 1993 Lack EE: Other neoplasm and tumor-like lesions of the adrenal glands. In: Atlas of Tumor Pathology, Tumors of the Adrenal Gland and Extraadrenal Paraganglia., Ed Rosai J), Armed Forces Institute of Pathology, Washington DC, 1997 Longo JM, Jafri SZ, Bis KB: Adrenal lymphangioma: a case report. Clin Imag 24: 104-106, 2000 Plaut A: Hemangiomas and related lesions of the adrenal gland. Virchows Arch Path Anat 335: 345-355, 1962 Plaut A: Locally invasive lymphangioma of adrenal gland. Cancer 15: 1165-1169, 1962 Poncelet V: Retroperitoneal cystic lymphangioma. J Belge Radiol 81: 245, 1998 Rozenblit A, Morehouse HT, Amis ES: Cystic adrenal lesions. Radiology 201: 541-548, 1996 Satou T, Uesugi T, Nakai Y, et al: Case of adrenal lymphangioma with atypical lymphocytes in aspirate cytology. Diagn Cytopathol 29: 87-90, 1996 Tagge DU, Baron PL: Giant adrenal cyst: Management and review of the literature. Am Surg 63: 744-746, 1997 Torres C, Ro JY, Batt MA, et al: Vascular adrenal cysts: a clinicopathologic and immunohistochemical study of six cases and review of the literature. Mod Pathol 10: 530-536, 1997 Tung GA, Pfister RC, Papanicolaou N, et al: Adrenal cysts: imaging and percutaneous aspiration. Radiology 173: 107-110, 1989 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2005 VL 11 IS 4 BP 242 EP 244 PG 3 ER PT J AU Eckhardt, S AF Eckhardt, Sandor TI Viral Therapy of Human Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review AB Viral Therapy of Human Cancer JG Sinkovics, JC Horvath (eds), Marcel Dekker New York, 2005, pp 1-829 C1 [Eckhardt, Sandor] National Institute of OncologyBudapest, Hungary. RP Eckhardt, S (reprint author), National Institute of Oncology, Budapest, Hungary. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2005 VL 11 IS 4 BP 245 EP 245 PG 1 ER PT J AU Kopper, L Timar, J AF Kopper, Laszlo Timar, Jozsef TI Genomics of Renal Cell Cancer - Does It Provide Breakthrough? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE renal cell cancer; genomics; prognostic markers; targeted therapy ID renal cell cancer; genomics; prognostic markers; targeted therapy AB It is a strong hope that the more we characterize the pathways in an individual tumor, the better we will be able to evaluate the response to a specific therapy. Different array technologies could be powerful tools to achieve this goal, i.e. selecting patients on the basis of the genomic and/or proteomic profiles who would really benefit from the target-designed therapy. Genomic analysis of RCC accumulated ample of data which now can be exploited in clinical management of a previously almost uncontrollable disease. Beside the previously identified genetic abnormalities (VHL, MET, EGFR), CAIX seems to be a novel molecular marker of RCC. Array studies also outlined a small set of tumor markers, vimentin, galectin-3, CD74 and parvalbumin, which can define the individual histologic subtypes of RCC. We are at the beginning to take advantage of the genomic results. Some new approaches will interfere with the progression of RCC (anti-VEGF, anti-VEGFR or anti-EGFR therapies). Further novel molecular targets are available, such as HIF, HSP90 or the IFN-regulated genes, which can be used to the fine-tuning of RCC therapy. C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor ProgressionBudapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM kopper@korb1.sote.hu CR Beniers AJ, Peelen WP, Debruyne FM, Schalken JA: HLAclass- I and -class-II expression on renal tumor xenografts and the relation to sensitivity for alpha-IFN, gamma-IFN and TNF. Int J Cancer 48: 709-716, 1991 Boer JM, Huber WK, Sultmann H, et al: Identification and classification on differentially expressed genes in renal cell carcinoma by expression profiling on a global human 31,500-element cDNA array. Genome Res 11:1861-1870, 2001 Bromwich E, McMillan DC, Lamb GW, et al: The systemic inflammatory response, performance status and survival in patients undergoing alpha-interferon treatment for advanced renal cancer. Br J Cancer 91:1236-1238, 2004 Bromwich EJ, McArdle PA, Canna K, et al: The relationship between T-lymphocyte infiltration, stage, tumour grade and survival in patients undergoing curative surgery for renal cell cancer. Br J Cancer 89:1906-1908, 2003 Bui MH, Seligson D, Han KR, et al: Carbonic anhydrase IX is an independent predictor of survival in advanced renal clear cell carcinoma: implications for prognosis and therapy. Clin Cancer Res 9: 802-811, 2003 Bui MH, Visapaa H, Seligson D, et al: Prognostic value of carbonic anhydrase IX and KI67 as predictors of survival for renal clear cell carcinoma. J Urol 171:2461-2466, 2004 Campbell L, Gumbleton M, Griffiths DF: Caveolin-1 overexpression predicts poor disease-free survival of patients with clinically confined renal cell carcinoma. Br J Cancer 89:1909-1913, 2003 Castillo M, Petit A, Mellado B, et al: C-kit expression in sarcomatoid renal cell carcinoma: potential therapy with imatinib. J Urol 171:2176-2180, 2004 Chautard D, Dalifard I, Chassevent A, et al: Prognostic value of uPA, PAI-1, and DNA content in adult renal cell carcinoma. Urol 63:1055-1060, 2004 Dancey JE: Epidermal growth factor receptor and epidermal growth factor receptor therapies in renal cell carcinoma: do we need a better mouse trap? J Clin Oncol 22: 2975-2977, 2004 Dawson NA, Guo C, Zak R, et al: A phase II trial of gefitinib, Iressa, ZD1839, in stage IV and recurrent renal cell carcinoma. Clin Cancer Res 10: 7812-7819, 2004 Donskov F, Bennedsgarrd KM, Hokland M, et al: Leukocyte orchestration in blood and tumour tissue following interlukin-2 based immunotherapy in metastatic renal cell carcinoma. Cancer Immunol Immunother 53: 729-739, 2004 Foon KA, Yand XD, Wiener LM, et al: Preclinical and clinical evaluations of ABX-EGF, a fully human anti-epidermal growth factor receptor antibody. Int J Radiat Oncol Biol Phys 58: 984-990, 2004 Gollob JA: Sorafenib: scientific rationales for single-agent and combination therapy in clear-cell renal cell carcinoma. Clin Genitourin Cancer 4: 167-174, 2005 Jacobsen J, Grankvist K, Rasmuson T, et al: Expression of vascular endothelial growth factor protein in human renal cell carcinoma. BJU Int 93:297-302, 2004 Jermann M, Stahel RA, Salzberg M, et al: A phase II, openlabel study of gefitinib, IRESSA, in patients with locally advanced, metastatic, or relapsed renal-cell carcinoma. Cancer Chemother Pharmacol 57: 533-539, 2006 Joo HJ, Oh DK, Kim YS, et al: Increased expression of caveolin- 1 and microvessel density correlates with metastasis and poor prognosis in clear cell renal cell carcinoma. BJU Int 93:291-296, 2004 Kawata N, Yagasaki H, Hirakata H, et al: The impact of angiogenesis on the prognosis of advanced renal cell carcinoma. Hinyokika Kiyo 50:157-163, 2004 Kim HL, Seligson D, Liu X, et al: Using protein expression to predict survival in clear cell renal carcinoma. Clin Canc Res 10: 5464-5471, 2004 Kovacs G: Molecular differential pathology of renal tumors. Histopathology 22: 1-8, 1993 Kovacs G, Akhtar M, Beckwith BJ, et al: The Heidelberg classification of renal cell tumors. J Pathol 183: 131-133, 1997 Lam JS, Belldegrun AS, Figlin RA: Tissue array-based predictions of pathobiology, prognosis, and response to treatment for renal cell carcinoma therapy. Clin Cancer Res 10: 6304s-6309s, 2004 Langner C, Ratschek M, Rehak P, et al: Steroid hormone receptor expression in renal cell carcinoma: an immunohistochemical analysis of 182 tumors. J Urol 171:611-614, 2004. Langner C, Ratschek M, Rehak P, et al: Are heterogenous results of EGFR immunoreactivity in renal carcinoma related to non-standardised criteria for staining evaluation? J Clin Pathol 57:773-775, 2004 Lee S-W, Lee K-I, Kim JY: Revealing urologic diseases by proteomic technique. J Chromatogr B Analyt Technol Biomed Life Sci 815: 203-213, 2005 Linehan WM, Vasselli J, Srinivasan R, et al: Genetic basis of cancer of the kidney: disease-specific approaches to therapy. Clin Cancer Res 10, 6282s-6289s, 2004 Liou LS, Shi T, Duan Z-H, et al: Microarray gene expression profiling and analysis in renal cell carcinoma. BMC Urol 4:1- 11, 2004 Lorincz T, Timar J, Szendroi M: Alterations of microvascular density in bone metastases of adenocarcinoma. Pathol Oncol Res 10: 149-153, 2004 Mekhail TM, Kawanishi-Tabata R, Tubbs R, et al: Renal cell carcinoma, RCC, and telomerase activity: relationship to stage. Urol Oncol 21:424-430, 2003 Moch H, Sauter G, Buchholz N, et al: Epidermal growth factor receptor expression is associated with rapid tumor cell proliferation in renal cell carcinoma. Hum Pathol 28: 1255-1259, 1997 Moch H: Genomic alterations in renal tumours: what have we learned in the era of comparative genomic hybridisation? Pathology 36: 51-57, 2004 Motzer RJ, Michaelson D, Redman BG, et al: Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma. J Clin Oncol 24: 16-24, 2006 Negrier S, Perol D, Menetier-Caux C, et al: Interleukin-6, interleukin-10, and vascular endothelial growth factor in metastatic renal cell carcinoma: prognostic value of interleukin-6 – from the Groupe Francais d’Immunotherapie. J Clin Oncol 22:2371-2378, 2004 Padrik P: Prognostic factors of immunotherapy in metastatic renal cell carcinoma. Med Oncol 20:325-334, 2003 Pantuck AJ, Zeng G, Belldegrun AS, Figlin RA: Pathobiology, prognosis, and targeted therapy for renal cell carcinoma: exploiting the hypoxia-induced pathway. Clin Cancer Res 9:4641-4652, 2003 Paul R, Necknig U, Busch R, et al: Cadherin-6: a new prognostic marker for renal cell carcinoma. J Urol 171:97-101, 2004 Pavlovich CP, Schmidt LS: Searching for the hereditary causes of renal cell carcinoma. Nat Rev Cancer 4: 381-393, 2004 Ramp U, Krieg T, Caliskan E, et al: XIAP expression is an independent prognostic marker in clear-cell renal carcinomas. Hum Pathol 35:1022-1088, 2004 Richard S, Lidereau R, Giraud S: The growing family of hereditary renal cell carcinoma. Nephrol Dial Transplant 19: 2954- 2958, 2004 Rini BI, Small EJ: Biology and clinical development of vascular endothelial growth factor-targeted therapy in renal cell carcinoma. J Clin Oncol 23: 1028-1043, 2005 Rioux-Leclercq N, Delcros JG, Bansard JY, et al: Immunohistochemical analysis of tumor polyamines discriminates highrisk patients undergoing nephrectomy for renal cell carcinoma. Hum Pathol 35:1279-1284, 2004 Roemer A, Schwetmann L, Jung M, et al: Increased mRNA expression of ADAMs in renal cell carcinoma and their association with clinical outcome. Oncol Rep 11:529-536, 2004 Royston P, Sauerbrei W, Ritchie A: Is treatment with interferon-α effective in all patients with metastatic renal carcinoma? A new approach to the investigation of interactions. Br J Cancer 90:794-799, 2004 Sakaeda T, Okumura N, Gotoh A, et al: EGFR mRNA is upregulated, but somatic mutations of the gene are hardly found in renal cell carcinoma in Japanese patients. Pharm Res 22: 1757- 1761, 2005 Shimazui T, Yoshikawa K, Uemura H, et al: The level of cadherin- 6 mRNA in peripheral blood is associated with the site of metastasis and with the subsequent occurrence of metastases in renal cell carcinoma. Cancer 101:963-968, 2004 Staehler M, Rohrmann K, Haseke N, et al: Target agents for the treatment of advanced renal cell carcinoma. Curr Drug Targets 6: 835-846, 2005 Staller P, Sulitkova J, Lisztwan J, et al: Chemokine receptor CXCR4 downregulated by von Hippel-Lindau tumor suppressor pVHL. Nature 425: 307-311, 2003 Sufan RI, Jewett MAS, Ohh M: The role of von Hippel-Lindau tumor suppressor protein and hypoxia in renal clear cell carcinoma. Am J Physiol Renal Physiol 287: F1-F6, 2004 Takahashi M, Rhodes DR, Furge KA, et al: Gene expression profiling of clear cell renal cell carcinoma: gene identification and prognostic classification. Proc Natl Acad Sci USA 98: 9754-9759, 2001 Tan M-H, Rogers CG, Cooper JT, et al: Gene expression profiling of renal cell carcinoma. Clin Cancer Res 10: 6315s-6321s, 2004 Twine NC, Stover JA, Marshall B, et al: Disease-associated expression profiles in peripheral blood mononuclear cells from patients with advanced renal cell carcinoma. Cancer Res 63: 6069-6075, 2003 Uhlman DL, Nguyen P, Manivel JC, et al: Epidermal growth factor receptor and transforming growth factor alpha expression in papillary and nonpapillary renal cell carcinoma: correlation with metastatic behavior and prognosis. Clin Cancer Res 1: 913-920, 1995 Vasselli JR, Shih JH, Iyengar SR, et al: Predicting survival in patients with metastatic kidney cancer by gene-expression profiling in the primary tumor. Proc Natl Acad Sci USA 100: 6958-6963, 2003 Vogelzang NJ: Treatment options in metastatic renal carcinoma: an embarrassment of riches. J Clin Oncol 24: 1-3, 2006 Wittnebel S, Jalil A, Thiery J, et al: The sensitivity of renal cell carcinoma cells to interferon alpha correlates with p53-induction and involves Bax. Eur Cytokine Netw 16: 123-127, 2005 Yildiz E, Gokce G, Kilicarslan H, et al: Prognostic value of the expression of Ki-67, CD44 and vascular endothelial growth factor, and microvessel invasion, in renal cell carcinoma. BJU Int 93:1087-1093, 2004 Zigeuner R, Ratschek M, Rehak P, et al: Value of p53 as a prognostic marker in histologic subtypes of renal cell carcinoma: a systematic analysis of primary and metastatic tumor tissue. Urology 63:651-655, 2004 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2006 VL 12 IS 1 BP 5 EP 11 PG 7 ER PT J AU Koksal, TI Ates, M Danisman, A Sezer, C Ciftcioglu, A Karpuzoglu, G Sevuk, M AF Koksal, Turker Ismail Ates, Mutlu Danisman, Ahmet Sezer, Cem Ciftcioglu, Akif Karpuzoglu, Gulten Sevuk, Metin TI Reduced E-cadherin and α-catenin Expressions Have No Prognostic Role in Bladder Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE E-cadherin; α-catenin; transitional cell carcinoma; urinary bladder ID E-cadherin; α-catenin; transitional cell carcinoma; urinary bladder AB In various human cancers, dysfunction of the E-cadherin-catenin complex is associated with a decrease in cellular and tissue differentiation, and with higher invasive and metastatic potentials. The objective of this study was to investigate E-cadherin and α-catenin expression in superficial noninvasive papillary TCC and invasive TCC, and correlate these results with pathological and clinical parameters. We have used immunohistochemistry to localize Ecadherin and α-catenin in 56 formalin-fixed, paraffin-embedded tissue blocks from 41 patients with superficial bladder cancer and 15 with invasive bladder cancer. The 46 male and 10 female patients had a mean age of 67 years, with range of 40 to 82 years. The mean follow-up time was 33.4 (range 5-120) months. Tumor grade 1:2:3 ratios were 5:32:19. In superficial bladder tumor, abnormal expression of E-cadherin and α-catenin was demonstrated in 37 and 71% of the tumors, respectively. In advanced bladder tumor, abnormal expression of E-cadherin and α-catenin was demonstrated in 80 and 100% of the tumors, respectively. Differences in expression of E-cadherin and α-catenin could be discerned between superficial and advanced bladder tumors (p=0.004, p=0.024, respectively). However, the association between E-cadherin and α-catenin expression and tumor grade was not statistically significant (p>0.05). In addition, the expression of E-cadherin and α-catenin did not correlate with tumor number and size (p>0.05). We have demonstrated that abnormal expression of E-cadherin and/or α-catenin occurs in more than 85% of bladder carcinomas and correlates significantly only with advanced stage. Nevertheless, these observations need to be confirmed in larger prospective clinical studies. C1 [Koksal, Turker Ismail] Akdeniz University, Faculty of Medicine, Department of Urology, Dumlupinar Bulvari, Campus, 07070 Antalya, Turkey. [Ates, Mutlu] Akdeniz University, Faculty of Medicine, Department of Urology, Dumlupinar Bulvari, Campus, 07070 Antalya, Turkey. [Danisman, Ahmet] Akdeniz University, Faculty of Medicine, Department of Urology, Dumlupinar Bulvari, Campus, 07070 Antalya, Turkey. [Sezer, Cem] Akdeniz University, Faculty of Medicine, Department of PathologyAntalya, Turkey. [Ciftcioglu, Akif] Akdeniz University, Faculty of Medicine, Department of PathologyAntalya, Turkey. [Karpuzoglu, Gulten] Akdeniz University, Faculty of Medicine, Department of PathologyAntalya, Turkey. [Sevuk, Metin] Akdeniz University, Faculty of Medicine, Department of Urology, Dumlupinar Bulvari, Campus, 07070 Antalya, Turkey. RP Koksal, TI (reprint author), Akdeniz University, Faculty of Medicine, Department of Urology, 07070 Antalya, Turkey. EM tkoksal@akdeniz.edu.tr CR Abel PD, Henderson D, Bennett MK, et al: Differing interpretations by pathologists of the pT category and grade of transitional cell cancer of the bladder. Br J Urol 623: 39-42, 1988. Bringuier PP, Giroldi LA, Umbas R, et al: Mechanisms associated with abnormal E-cadherin immunoreactivity in human bladder tumors. Int J Cancer 83: 591-595, 1999. Bringuier PP, Umbas R, Schaafsma HE, et al: Decreased Ecadherin immunoreactivity correlates with poor survival in patients with bladder tumors. Cancer Res 53: 3241-3245, 1993. Fujisawa M, Miyazaki J, Takechi Y, et al: The significance of Ecadherin in transitional-cell carcinoma of the human urinary bladder. World J Urol 14: 12-15, 1996. Giroldi LA, Schalken JA: Decreased expression of the intercellular adhesion molecule E-cadherin in prostate cancer: biological significance and clinical implications. Cancer Metastasis Rev 12: 29-37, 1993. Hirano S, Kimoto N, Shimoyama Y, et al: Identification of a neural alpha-catenin as a key regulator of cadherin function and multicellular organization. Cell 70: 293-301, 1992. International Union Against Cancer, Hermanek P, Sobin LH:, eds, TNM Classification of Malignant Tumors, 5th ed, Springer-Verlag, Germany, 1997. Koksal IT, Ozcan F, Kilicaslan I, Tefekli A: Expression of E-cadherin in prostate cancer in formalin-fixed, paraffinembedded tissues: correlation with pathological features. Pathology 34: 233-238, 2002. Krishnadath KK, Tilanus HW, van Blankenstein M, et al: Reduced expression of the cadherin-catenin complex in oesophageal adenocarcinoma correlates with poor prognosis. J Pathol 182: 331-338, 1997. Landis SH, Murray T, Bolden S, Wingo PA: Cancer statistics, 1999. CA Cancer J Clin 49: 8-31, 1999. Lipponen PK: Review of cytometric methods in the assessment of prognosis in transitional cell bladder cancer. Eur Urol 21: 177-183, 1992. Lipponen PK, Eskelinen MJ: Reduced expression of E-cadherin is related to invasive disease and frequent recurrence in bladder cancer. J Cancer Res Clin Oncol 121:303-308, 1995. Lopez-Beltran A, Croghan GA, Croghan I, et al: Prognostic factors in survival of bladder cancer. Cancer 70: 799-807, 1992 Mansouri A, Spurr N, Goodfellow PN, Kemler R: Characterization and chromosomal localization of the gene encoding the human cell adhesion molecule uvomorulin. Differentiation 38: 67-71, 1988. Mialhe A, Louis J, Montlevier S, et al: Expression of E-cadherin and alpha-, beta- and gamma-catenins in human bladder carcinomas: are they good prognostic factors? Invasion Metastasis 17: 124-137, 1997. Mialhe A, Louis J, Pasquier D, et al: Expression of three cell adhesion molecules in bladder carcinomas: correlation with pathological features. Anal Cell Pathol 13: 125-136, 1997. Millan-Rodriguez F, Chechile-Toniolo G, Salvador-Bayarri J, et al: Multivariate analysis of the prognostic factors of primary superficial bladder cancer. J Urol 163: 73-78, 2000. Mostofi FK, Sobin LH, Torloni H: Histological typing of urinary bladder tumours, In: International Histological Classification of Tumors, 10. World Health Organisation, Geneva, 1973. Narayana AS, Loening SA, Slymen DJ, Culp DA: Bladder cancer: factors affecting survival. J Urol 130: 56-60, 1983. Oka H, Shiozaki H, Kobayashi K, et al: Expression of E-cadherin cell adhesion molecules in human breast cancer tissues and its relationship to metastasis. Cancer Res 53: 1696-1701, 1993. Otto T, Birchmeier W, Schmidt U, et al: Inverse relation of Ecadherin and autocrine motility factor receptor expression as a prognostic factor in patients with bladder carcinomas. Cancer Res 54: 3120-3123, 1994. Ozawa M, Baribault H, Kemler R: The cytoplasmic domain of the cell adhesion molecule uvomorulin associates with three independent proteins structurally related in different species. EMBO J 8: 1711-1717, 1989. Ozawa M, Ringwald M, Kemler R: Uvomorulin-catenin complex formation is regulated by a specific domain in the cytoplasmic region of the cell adhesion molecule. Proc Natl Acad Sci USA 8742: 46-50, 1990. Ross JS, del Rosario AD, Figge HL, et al: E-cadherin expression in papillary transitional cell carcinoma of the urinary bladder. Hum Pathol 26: 940-944, 1995. Shimoyama Y, Nagafuchi A, Fujita S, et al: Cadherin dysfunction in a human cancer cell line: possible involvement of loss of alpha-catenin expression in reduced cell-cell adhesiveness. Cancer Res 52: 5770-5774, 1992. Shimazui T, Schalken JA, Giroldi LA, et al: Prognostic value of cadherin-associated molecules, alpha-, beta-, and gammacatenins and p120cas, in bladder tumors. Cancer Res 56: 4154- 4158, 1996. Soloway MS: Intravesical therapy for bladder cancer. Urol Clin North Am 15: 661-669, 1988. Syrigos KN, Harrington K, Waxman J, et al: Altered gammacatenin expression correlates with poor survival in patients with bladder cancer. J Urol 160: 1889-1893, 1998. Syrigos KN, Krausz T, Waxman J, et al: E-cadherin expression in bladder cancer using formalin-fixed, paraffin-embedded tissues: correlation with histopathological grade, tumour stage and survival. Int J Cancer 64: 367-370, 1995. Takaishi M: Cadherin cell adhesion receptor as a morphogenetic regulator. Science 251:1451-1455, 1991. Thrasher JB, Crawford ED: Current management of invasive and metastatic transitional cell carcinoma of the bladder. J Urol 149: 957-972, 1993. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2006 VL 12 IS 1 BP 13 EP 19 PG 7 ER PT J AU Elagoz, S Egilmez, R Koyuncu, A Muslehiddinoglu, A Arici, S AF Elagoz, Sahande Egilmez, Reyhan Koyuncu, Ayhan Muslehiddinoglu, Ahmet Arici, Sema TI The Intratumoral Microvessel Density and Expression of bFGF and nm23-H1 in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; microvascular density; basic fibroblast growth factor; nm23-H1 ID Colorectal cancer; microvascular density; basic fibroblast growth factor; nm23-H1 AB It has previously been reported that intratumoral microvessel density (IMD), and the expression of bFGF and nm23-H1 are useful prognostic markers in colorectal cancer (CRC). In this study, a total of 100 CRCs were evaluated histopathologically, and IMD, bFGF and nm23-H1 expression were assessed by immunohistochemistry. IMD of patients increased with grade and stage, and this increase was statistically significant (p<0.05). A significantly higher incidence of high bFGF expression scores was also associated with increasing grade and stage (p<0.05). However, there was no significant difference between the grades in nm23-H1 expression (p=0.234). nm23-H1 expression occurred with lower incidence in stages C1, C2 and D than in stages B1 and B2 (p<0.05). Thus, a negative correlation was found between nm23-H1 expression and stage or lymph node metastasis (LNM) (p<0.05). IMD and bFGF expression were positively correlated with grade, stage, LNM, and lymphovascular invasion. Although positive correlation was found between IMD and bFGF, nm23-H1 expression negatively correlated with both of them. As a result, in clinical practice, increased IMD and bFGF expression and decreased nm23-H1 expression may provide valuable information in characterizing the malignant phenotype. C1 [Elagoz, Sahande] Cumhuriyet University School of Medicine, Department of Pathology, 58140 Sivas, Turkey. [Egilmez, Reyhan] Cumhuriyet University School of Medicine, Department of Pathology, 58140 Sivas, Turkey. [Koyuncu, Ayhan] Cumhuriyet University School of Medicine, Department of General SurgerySema, Turkey. [Muslehiddinoglu, Ahmet] Cumhuriyet University School of Medicine, Department of Pathology, 58140 Sivas, Turkey. [Arici, Sema] Cumhuriyet University School of Medicine, Department of Pathology, 58140 Sivas, Turkey. RP Elagoz, S (reprint author), Cumhuriyet University School of Medicine, Department of Pathology, 58140 Sivas, Turkey. EM selagoz@cumhuriyet.edu.tr CR Harpaz N, Saxena R: Large intestine. In: Modern Surgical Pathology., Eds: Weidner N, Cote RJ, Suster S, Weis LM). 1st Edition, Saunders, Philadelphia, 2003, pp 749-852 Vermeulen PB, Van den Eynden GG, Huget P, et al: Prospective study of intratumoral microvessel density, p53 expression and survival in colorectal cancer. Br J Cancer 79:316-322, 1999 Weidner N, Semple JP, Welch WR, et al: Tumor angiogenesis and metastasis – correlation in invasive breast carcinoma. N Eng J Med 324: 1-8, 1991 Graham CH, Rivers J, Kerbel RS, et al: Extent of vascularization as a prognostic indicator in thin, <0.76 mm, malignant melanomas. Am J Pathol 145: 510-514, 1994 Smith-McCune KK, Weidner N: Demonstration and characterization of the angiogenic properties of cervical dysplasia. Cancer Res 54:800-804, 1994 Goi T, Fujioka M, Satoh Y, et al: Angiogenesis and tumor proliferation/ metastasis of human colorectal cancer cell line SW620 transfected with endocrine glands derived vascular endothelial growth factor, as a new angiogenic factor. Cancer Res 64: 1906-1910, 2004 Saad RS, Liu YL, Nathan G, et al: Endoglin, CD105, and vascular endothelial growth factor as prognostic markers in colorectal cancer. Mod Pathol 17: 197-203, 2004 Fenjvesi A: Prognostic significance of tumor–induced angiogenesis in colorectal carcinoma. Med Pregl 56: 263-268, 2003 Aotake T, Lu C, Chiba Y, et al: Changes of angiogenesis and tumor cell apoptosis during colorectal carcinogenesis. Clin Cancer Res 5: 135-142, 1999 Shan YS, Lee JC, Chow NH, et al: Immunohistochemical microvessel count is not a reliable prognostic predictor in colorectal carcinoma. Hepatogastroenterol 50: 1316-1320, 2003 Banner BF, Whitehause R, Baker SP, et al: Tumor angiogenesis in stage II colorectal carcinoma. Association with survival. Am J Clin Pathol 109: 733-737, 1998 Bossi P, Viale G, Lee AKC, et al: Angiogenesis in colorectal tumors: microvessel quantitation in adenomas and carcinomas with clinicopathological correlations. Cancer Res 55: 5049- 5053, 1995 Saito S, Tsuno N, Nagawa H, et al: Expression of platelet-derived endothelial cell growth factor correlates with good prognosis in patients with colorectal carcinoma. Cancer 88: 42-49, 2000 Leung DW, Cachianes G, Kuang WJ, et al: Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 246: 1306-1309, 1989 New B, Yeoman L: Identification of basic fibroblast growth factor sensitivity and receptor and ligand expression in human colon tumor cell lines. Cell Physiol 150: 320-326, 1992 Steeg PS, Bevilacqua G, Kopper L et al: Evidence for a novel gene associated with low tumor metastatic potential. J Natl Cancer Inst 80: 200-204, 1998 Tannapfel A, Kockerling F, Katalinic A, et al: Expression of nm23-H1 predicts lymph node involvement in colorectal carcinoma. Dis Colon Rectum 38: 651-654, 1995 Ayhan A, Yasui W, Yokozaki H, et al: Reduced expression of nm23-H1 protein is associated with advanced tumor stage and distant metastases in human colorectal carcinomas. Virchows Arch B Cell Pathol 63: 213-218, 1993 Heide I, Theide C, Poppe K, et al: Expression and mutational analysis of nm23-H1 in liver metastases of colorectal cancer. Br J Cancer 70: 1267-1271, 1994 Lee JC, Lin YJ, Chow NH, et al: Reappraisal of the role of nm23-H1 in colorectal cancers. J Surg Oncol 76: 58-62, 2001 Garinis GA, Manolis EN, Spanakis NE, et al: High frequency of concomitant nm23-H1 and E-cadherin transcriptional inactivation in primary non-inheriting colorectal carcinomas. J Mol Med 81: 256-263, 2003 George ML, Dzik-Jurasz ASK, Padhani AR, et al: Non-invasive methods of assessing angiogenesis and their value in predicting response to treatment in colorectal cancer. Br J Surg 88: 1628- 1636, 2001 Lackner C, Jukic Z, Tsybrovskyy O, et al.: Prognostic relevance of tumor-associated macrophages and von Willebrand factorpositive microvessels in colorectal cancer. Virchows Arch 445:160-167, 2004 Hawighorts H, Knapstein PG, Knopp MV, et al: Uterine cervical carcinoma: Comparison of standard and pharmacokinetic analysis of time intensity curves for assessment of tumor angiogenesis and patient survival. Cancer Res 58: 3598-3602, 1998 Tomisaki S, Ohno S, Ichiyoshi Y, et al: Microvessel quantification and its possible relation with liver metastasis in colorectal cancer. Cancer 77: 1722-1728, 1996 Tanigawa N, Amaya H, Matsumara M, et al: Tumor angiogenesis and mode of metastasis in patients with colorectal cancer. Cancer Res 57:1043-1046, 1997 Lindmark G, Gerdin B, Sundberg C, et al: Prognostic significance of the microvascular count in colorectal cancer. J Clin Oncol 14: 461-466, 1996 Takahashi Y, Kitadai Y, Bucana CD, et al: Expression of vascular endothelial growth factor and its receptor, KDR, correlates with vascularity, metastasis, and proliferation of human colon cancer. Cancer Res 55: 3964-3968, 1995 Frank RE, Saclarides TJ, Leurgans S, et al: Tumor angiogenesis as a predictor of recurrence and survival in patients with node-negative colon cancer. Ann Surg 222: 695-699, 1995 White JL, Hewett PL, Kosuge D, et al: Vascular endothelial growth factor-D expression is an independent prognostic marker for survival in colorectal carcinoma. Cancer Res 62:1669-1675, 2002 Li Z-P, Meng Q-F, Sun C-H, et al: Tumor angiogenesis and dynamic CT in colorectal carcinoma: Radiologic-pathologic correlation. World J Gastroenterol 11: 1287-1291, 2005 Basilico C, Moscatelli D: The FGF family of growth factors and oncogenes. Adv Cancer Res 59: 115-125, 1992 Landriscina M, Cassano A, Ratto C et al: Quantitative analysis of basic fibroblast growth factor and vascular endothelial growth factor in human colorectal cancer. Br J Cancer 78: 765- 770, 1998 Zheng S, Han MY, Xiao ZX, et al: Clinical significance of vascular endothelial growth factor expression and neovascularization in colorectal carcinoma. World J Gastroenterol 9: 1227- 1230, 2003 Takahashi Y, Tucker SL, Kitadai Y, et al: Vessel counts and expression of vascular endothelial growth factor as prognostic factors in node-negative colon cancer. Arch Surg 132: 541-546, 1997 Stahl JA, Leone A, Rosengard AM, et al: Identification of a second human nm23-H1 gene, nm23-H1-H2. Cancer Res 51: 445- 449, 1991 Xerri L, Grob JJ, Battyani Z, et al.: nm23-H1 expression in metastasis of malignant melanoma is a predictive prognostic parameter correlated with survival. Br J Cancer 70: 1224-1228, 1994 Yamaguchi A, Urano T, Goi T, et al: Expression of human nm23- H1 and nm23-H1-H2 proteins in hepatocellular carcinoma. Cancer 73: 2280-2284, 1994 Kodera Y, Isobe KI, Yamauchi M et al: Expression of nm23-H1 RNA levels in human gastric cancer tissues. Cancer 73: 259- 265, 1994 Zou M, Shi Y, al-Sedairy S, et al: High levels of nm23-H1 gene expression in advanced stage of thyroid carcinomas. Br J Cancer 68: 385-388, 1993 Engel M, Theisinger B, Seib T, et al: High levels of nm23-H1 and nm23-H1-H2 messenger RNA in human squamous-cell lung carcinoma are associated with poor differentiation and advanced tumor stages. Int J Cancer 55: 375-379, 1993 Zeng ZS, Hsu S, Zhang ZF, et al: High level of nm23-H1 gene expression is associated with local colorectal cancer progression not with metastasis. Br J Cancer 70: 1025-1030, 1994 Yamaguchi A, Urana T, Fushida S, et al: Inverse association of nm23-H1 expression by colorectal cancer with liver metastasis. Cancer 68: 1020-1024, 1993 Lindmark G: nm23-H1 immunohistochemistry is not useful as predictor of metastatic potential of colorectal cancer. Br J Cancer 74: 1413-1418, 1996 Sarris M, Lee CS: Nm23-H1 protein expression in colorectal carcinoma metastasis in regional lymph nodes and the liver. Eur J Surg Oncol 27: 170-174, 2001 Soliani P, Ziegler S, Romani A et al: Prognostic significance of nm23-H1 gene product expression in patients with colorectal carcinoma treated with radical intent. Oncol Rep 11: 1193- 1200, 2004 Dursun A, Akyurek N, Gunel N, et al: Prognostic implication of nm23-H1 expression in colorectal carcinomas. Pathology 34: 427-432, 2002 Dusonchet L, Corsale S, Migliavacca M et al: Nm23-H1 expression does not predict clinical survival in colorectal cancer patients. Oncol Rep 10: 1257-1263, 2003 Royds JA, Cross SS, Silcocs PB, et al: Nm23-H1 “anti-metastatic” gene product expression in colorectal carcinoma. J Pathol 172: 261-266, 1994 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2006 VL 12 IS 1 BP 21 EP 27 PG 7 ER PT J AU Pezeshki, A Sari-Aslani, F Ghaderi, A Doroudchi, M AF Pezeshki, Abdulmohammad Sari-Aslani, Fatemeh Ghaderi, Abbas Doroudchi, Mehrnoosh TI p53 Codon 72 Polymorphism in Basal Cell Carcinoma of the Skin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE basal cell carcinoma; p53; polymorphism; sun exposure ID basal cell carcinoma; p53; polymorphism; sun exposure AB Basal cell carcinoma (BCC) is the most prevalent cancer in Iran. A common polymorphism at codon 72 of exon 4 of p53 tumor suppressor gene has been reported to be associated with increased inheritable susceptibility to several cancers. In the present study the frequency of p53 codon 72 polymorphism in 91 patients with BCC of skin, compared to 465 healthy normal individuals, was investigated. In total, there was no significant difference in the p53 genotypes between patients and controls. However, there was an apparent increase in the Arg/Arg genotype among those BCC patients who had a history of occupational sun exposure, compared to non-exposed patients (46.3% vs. 23.1%, P=0.11). A trend of increase in the frequency of Arg allele among sun-exposed patients was also observed (69.4% vs. 53.8%, P=0.07). Comparison of the genotype frequencies between sunexposed patients and normal controls confirmed the accumulation of Arg/Arg genotype in these patients (46.3% vs. 34.8%, P = 0.07). In addition, the frequency of Arg allele was significantly higher in sunexposed patients compared to controls (69.4% vs. 58.2%, P=0.03). Our results suggest that Arg allele at codon 72 of p53 gene might affect the risk of ultraviolet-induced basal cell carcinoma. C1 [Pezeshki, Abdulmohammad] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Sari-Aslani, Fatemeh] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Ghaderi, Abbas] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Doroudchi, Mehrnoosh] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. RP Doroudchi, M (reprint author), Shiraz University of Medical Sciences, Institute for Cancer Research, Shiraz, Iran. EM mdoroud@sums.ac.ir CR de Gruijl FR, van Kranen HJ, Mullenders LH: UV-induced DNA damage, repair, mutations and oncogenic pathways in skin cancer. J Photochem Photobiol 63: 19-27, 2001 Kim MY, Park HJ, Baek SC, et al: Mutations of the p53 and PTCH gene in basal cell carcinomas: UV mutation signature and strand bias. J Dermatol Sci 29: 1-9, 2002 McGregor JM, Harwood CA, Brooks L, et al: Relationship between p53 codon 72 polymorphism and susceptibility to sunburn and skin cancer. J Invest Dermatol 119: 84-90, 2002 Murata M, Tagawa M, Kimura M, et al: Analysis of a germ line polymorphism of the p53 gene in lung cancer patients; discrete results with smoking history. Carcinogenesis 17: 261-264, 1996 Dumont P, Leu JI, Della Pietra AC, et al: The codon 72 polymorphic variants of p53 have markedly different apoptotic potential. Nat Genet 33: 357-365, 2003 Buchman VL, Chumakov PM, Ninkina NN, et al: A variation in the structure of the protein-coding region of the human p53 gene. Gene 70: 245-252, 1988 Marin MC, Jost CA, Brooks LA, et al: A common polymorphism acts as an intragenic modifier of mutant p53 behaviour. Nat Genet 25: 47-54, 2000 Tada M, Furuuchi K, Kaneda M, et al: Inactivate the remaining p53 allele or the alternate p73? Preferential selection of the Arg72 polymorphism in cancers with recessive p53 mutants but not transdominant mutants. Carcinogenesis 22: 515-517, 2001 Marshall SE, Bordea C, Wojnarowska F, et al: p53 codon 72 polymorphism and susceptibility to skin cancer after renal transplantation. Transplantation 69: 994-996, 2000 Makni H, Franco EL, Kaiano J, et al: p53 polymorphism in codon 72 and risk of human papillomavirus-induced cervical cancer: effect of inter-laboratory variation. Int J Cancer 87: 528-533, 2000 Nagpal JK, Sahni S, Das BR: p53 codon 72 polymorphism and susceptibility to development of human papilloma virus-associated cervical cancer in Indian women. Eur J Clin Invest 32: 943-948, 2002 Qie M, Zhang Y, Wu J: Study on the relationship between cervical cancer and p53 codon 72 polymorphism. Hua Xi Yi Ke Da Xue Xue Bao 33: 274-275, 2002 Li T, Lu ZM, Guo M, et al: p53 codon 72 polymorphism, C/G, and the risk of human papillomavirus-associated carcinomas in China. Cancer 95: 2571-2576, 2002 Pegoraro RJ, Moodley M, Rom L, et al: p53 codon 72 polymorphism and BRCA 1 and 2 mutations in ovarian epithelial malignancies in black South Africans. Int J Gynecol Cancer 13: 444-449, 2003 Suzuki K, Matsui H, Ohtake N, et al: A p53 codon 72 polymorphism associated with prostate cancer development and progression in Japanese. J Biomed Sci 10: 430-435, 2003 Cenci M, French D, Pisani T, et al: p53 polymorphism at codon 72 is not a risk factor for cervical carcinogenesis in central Italy. Anticancer Res 23: 1385-1387, 2003 Abba MC, Villaverde LM, Gomez MA, et al: The p53 codon 72 genotypes in HPV infection and cervical disease. Eur J Obstet Gynecol Reprod Biol 109: 63-66, 2003 Anzola M, Cuevas N, Lopez-Martinez M, et al: Frequent loss of p53 codon 72 Pro variant in hepatitis C virus-positive carriers with hepatocellular carcinoma. Cancer Lett 193: 199-205, 2003 Buyru N, Budak M, Yazici H, Dalay N: p53 gene mutations are rare in human papillomavirus-associated colon cancer. Oncol Rep 10: 2089-2092, 2003 Humbey O, Cairey-Remonnay S, Guerrini JS, et al: Detection of the human papillomavirus and analysis of the TP53 polymorphism of exon 4 at codon 72 in penile squamous cell carcinomas. Eur J Cancer 39: 684-690, 2003 Soulitzis N, Sourvinos G, Dokianakis DN, Spandidos DA: p53 codon 72 polymorphism and its association with bladder cancer. Cancer Lett 179: 175-183, 2002 International Agency for Research on Cancer: Solar and Ultraviolet Radiation, vol. 55, International Agency for Research on Cancer, Lyon, 1992 Weihrauch M, Bader M, Lehnert G, et al: Carcinogen-specific mutation pattern in the p53 tumor suppressor gene in UV radiation- induced basal cell carcinoma. Int Arch Occup Environ Health 75: 272-276, 2002 Ananthaswamy HN, Loughlin SM, Cox P, et al: Sunlight and skin cancer: inhibition of p53 mutations in UV-irradiated mouse skin by sunscreens. Nat Med 3: 510-514, 1997 Feynman RP, Leighton SM, In: The Feynman lectures on Physics, Vol. III. Addison-Weseley, Reading, MA, 1967, pp.15-7-15-12 Ziegler AD, Leffel DJ, Kunala S, et al: Mutation hotspots due to sunlight in the p53 gene of non-melanoma skin cancers. Proc Natl Acad Sci USA 90: 4216-4220, 1993 Farahmandbeigi M, Kadivar MR: The incidence rate of registered cancers in Fars province. Disease Control Unit, Shiraz University Press, Iran, 2000 Baron JA, Greenberg ER: Prevention of non-melanoma skin cancer. Arch Dermatol 136: 245-246, 2000 Argorastos T, Lambropoulos AF, Constantinidis TC, et al: p53 codon 72 polymorphism and risk of intra-epithelial and invasive cervical neoplasia in Greek women. Eur J Cancer Prev 9: 113-118, 2000 Armstrong BK, Kricker A: The epidemiology of UV induced skin cancer. J Photochem Photobiol B 63: 8-18, 2001 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2006 VL 12 IS 1 BP 29 EP 33 PG 5 ER PT J AU Pal, J Nyarady, Z Marczinovits, I Par, A Ali, SY Berencsi, Gy Kvell, K Nemeth, P AF Pal, Jozsef Nyarady, Zoltan Marczinovits, Ilona Par, Alajos Ali, Saleh Younes Berencsi, Gyorgy Kvell, Krisztian Nemeth, Peter TI Comprehensive Regression Analysis of Hepatitis B Virus X Antigen Level and Anti-HBx Antibody Titer in the Sera of Patients with HBV Infection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatitis B virus; HBxAg; immunoserology; epitope mapping; antibody response ID Hepatitis B virus; HBxAg; immunoserology; epitope mapping; antibody response AB Although the pathogenetic significance of hepatitis B virus x protein (HBxAg) in chronic hepatitis, liver cirrhosis, and primary hepatocellular carcinoma has already been studied, the comparative analyses of both the actual serum HBxAg levels and antibody production against various HBx epitopes have been examined to lesser extent. We have simultaneously investigated the relationship between antibody production (IgG and IgM) against the HBxAg fragments and HBxAg level in the sera of patients with acute (14) or chronic hepatitis (80) and symptomless carriers (12). A recently developed sandwich-type ELISA was used for the quantitative measurements of HBxAg. Overlapping recombinant and synthetic antigens were used to map the fine epitope specificities of circulating anti-HBx antibodies. In acute hepatitis, we have found high and homogenous correlation in the IgM type immune responses against all the examined HBxAg regions. Moreover, strong correlation has been observed between IgG type immune responses to a characteristic C-terminal region (C1: 79-117) and the longest fragment (X: 10-143). Moderate correlation has been found between HBxAg concentration and the IgG type anti-HBx antibody levels against C-terminus of HBxAg in patients with chronic hepatitis. In the case of symptomless carriers, there were also demonstrable associations in the immune responses against the C-terminal sequences; however, significant correlations were found for antibody production against the N-terminal region as well. The examinations show that the C-terminal sequence, responsible for transactivation, promotes an efficient IgG antibody response in all three groups of patients, whereas the negative regulator N-terminal part of the HBxAg molecule for the most part does not trigger antibody production. This suggests that the immune responses against various - biologically active - epitopes of the HBxAg may have a different role in the pathogenesis of hepatitis and may be used as prognostic markers in human HBV infections. C1 [Pal, Jozsef] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Nyarady, Zoltan] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Marczinovits, Ilona] University of Szeged, Department of AnatomySzeged, Hungary. [Par, Alajos] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Ali, Saleh Younes] University of Garyounis, Faculty of MedicineBenghazi, Libyan Arab Jamabiriya. [Berencsi, Gyorgy] ANTSZ Regional Institute of State Public Health Service, Regional Laboratory of VirologyPecs, Hungary. [Kvell, Krisztian] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. [Nemeth, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti u. 12., H-7643 Pecs, Hungary. RP Nemeth, P (reprint author), University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, H-7643 Pecs, Hungary. EM peter.nemeth@aok.pte.hu CR Altschul SF, Madden TL, Schaffer AA, et al: “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs”. Nucleic Acids Res 25: 3389-3402,1997 Bichko V, Pushko P, Dreilina D, et al: Subtype ayw variant of hepatitis B virus. DNA primary structure analysis. FEBS Lett 185: 208- 212, 1985 Caselmann WH: Trans-activation of cellular genes by hepatitis B virus proteins: A possible mechanism of hepatocarcinogenesis. Adv Virus Res 47: 253-302, 1996 Feitelson MA, Clayton MM, Blumberg BS: X antigen/antibody markers in hepadnavirus infections. Presence and significance of the hepadnavirus X gene product(s, in serum. Gastroenterology 98: 1071-1078, 1990 Hann HW, Lee J, Bussard A, et al: Preneoplastic markers of hepatitis B virus-associated hepatocellular carcinoma. Cancer Res 64: 7329-7335, 2004 Hwang GY, Lin CY, Huang LM, et al: Detection of the hepatitis B virus X protein, HBx, antigen and anti-HBx antibodies in cases of human hepatocellular carcinoma. J Clin Microbiol 41: 5598-5603, 2003 Jung MC, Stemler M, Weimer T, et al: Immune response of peripheral blood mononuclear cells to HBx-antigen of hepatitis B virus. Hepatology 13: 637-643, 1991 Kumar V, Jayasuryan N, Kumar R: A truncated mutant, residues 58- 140, of the hepatitis B virus X protein retains transactivation function. Proc Natl Acad Sci USA 93:5647-5652, 1996 Malik AH, Lee WM: Chronic hepatitis B virus infection: treatment strategies for the next Millennium. Lee Ann Intern Med 132: 723- 731, 2000 Marczinovits I, Somogyi C, Patthy A, et al: An alternative purification protocol for producing hepatitis B virus X antigen on a preparative scale in Escherichia coli. J Biotechnol 56: 81-88, 1997 Misra KP, Mukherji A, Kumar V: The conserved amino-terminal region, amino acids 1-20, of the hepatitis B virus X protein shows a transrepression function. Virus Res 105: 157-165, 2004 Murakami S, Cheong JH, Kaneko S: Human hepatitis virus X gene encodes a regulatory domain that represses transactivation of X protein. J Biol Chem 269: 15118-15123, 1994 Murakami S: Hepatitis B virus X protein. Structure, function and biology. Intervirology 42: 81-99, 2001 Murakami S: Hepatitis B virus X protein: a multifunctional viral regulator. J Gastroenterol 36: 651-660, 1999 Ordog K, Szendroi A, Szarka K, et al: Perinatal and intrafamily transmission of hepatitis B virus in three generations of a low-prevalence population. J Med Virol 70: 194-204, 2003 Pal J, Czompoly T, Nyarady Z, et al: Determination of the fine epitope specificity of an anti-hepatitis B virus X protein monoclonal antibody using microanalytical and molecular biological methods. Mol Immunol 40: 241-246, 2003 Pal J, Palinkas L, Nyarady Z, et al:Sandwich type ELISA and a fluorescent cytometric microbead assay for quantitative determination of Hepatitis B virus X antigen level in human sera. J Immunol Methods, 2005, accepted for publication) Pal J, Somogyi C, Szmolenszky AA, et al: Immunohistochemical assessment and prognostic value of hepatitis B virus X protein in chronic hepatitis and primary hepatocellular carcinomas using anti- HBxAg monoclonal antibody. Pathol Oncol Res 7: 178-184, 2001 Song CZ, Wang QW, Song CC, Bai ZL: Viral replication modulated by synthetic peptide derived from hepatitis B virus X protein. World J Gastroenterol 10: 389-392, 2004 Stemler M, Weimer T, Tu ZX, et al: Mapping of B-cell epitopes of the human hepatitis B virus X protein. J Virol 64: 2802-2809, 1990 Tiollais P, Pourcel C, Dejan A: The hepatitis B virus. Nature 317: 489-495, 1985 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2006 VL 12 IS 1 BP 34 EP 40 PG 7 ER PT J AU Saqui-Salces, M Martinez-Benitez, B Gamboa-Dominguez, A AF Saqui-Salces, Milena Martinez-Benitez, Braulio Gamboa-Dominguez, Armando TI EBV+ Lymphoepithelial Carcinoma of the Parotid Gland in Mexican Mestizo Patients with Chronic Autoimmune Diseases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE lymphoepithelial carcinoma; Epstein-Barr virus; autoimmune disease; salivary glands ID lymphoepithelial carcinoma; Epstein-Barr virus; autoimmune disease; salivary glands AB Lymphoepithelial carcinomas of the salivary gland are rare tumors constantly associated with Epstein-Barr virus (EBV) and mainly identified in Asiatic and Greenlander population. Four cases have been described in Caucasians, only two with EBV infection. We describe two cases of parotid gland lymphoepithelial carcinomas in Mexican mestizo women in which chronic latent EBV infection was documented by immunohistochemistry and in situ hybridization. One patient had primary Sjogren’s syndrome and the other systemic lupus erythematosus of six and three years of evolution, respectively. Epithelial neoplastic cells showed latency pattern II (LMP1+, EBNA-2-, EBER+) with a dense inflammatory infiltrate composed mainly by CD8+ T lymphocytes. Follow-up excluded nasopharyngeal involvement in both patients. This report expands the ethnic groups in which salivary lymphoepithelial carcinomas associated with chronic latent EBV infection have been described, and illustrates for the first time its association with autoimmune diseases in two women living in a region non-endemic for this unusual neoplasm. C1 [Saqui-Salces, Milena] Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No 15, 14000 Tlalpan, Mexico. [Martinez-Benitez, Braulio] Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No 15, 14000 Tlalpan, Mexico. [Gamboa-Dominguez, Armando] Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Pathology, Vasco de Quiroga No 15, 14000 Tlalpan, Mexico. RP Gamboa-Dominguez, A (reprint author), Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Pathology, 14000 Tlalpan, Mexico. EM agamboad@quetzal.innsz.mx CR Abu-Shakra M, Ehrenfeld M, Shoenfeld Y: Systemic lupus erythematosus and cancer: associated or not? Lupus 11:137-144, 2002 Albeck H, Nielsen NH, Hansen HE, et al: Epidemiology of nasopharyngeal and salivary carcinoma in Greenland. Artic Med Res 51:189-195, 1992 Bialas M, Sinczak A, Choinska-Stefanska A, Zygulska A: EBVpositive lymphoepithelial carcinoma of salivary gland in a woman of a non-endemic area—a case report. Pol J Pathol 53:235-238, 2002 Bjornadal L, Lofstrom B, Yin L, et al: Increased cancer incidence in a Swedish cohort of patients with systemic lupus erythematosus. Scand J Rheumatol 31:66-71, 2002 Chan JKC, Yip TTC, Tsang WYW: Specific association of Epstein-Barr virus with lymphoepithelial carcinoma among tumors and tumor like lesions of the salivary gland. Arch Pathol Lab Med 118: 994-997, 1994 Hamilton-Dutoit SJ, Therkildsen MH, Nielsen NH: Undifferentiated carcinoma of the salivary gland in Greenlandic Eskimos: demonstration of Epstein-Barr virus DNA by in situ nucleic hybridization. Hum Pathol 22: 811-815, 1991 Jen KY,Cheng J, Li J, et al: Mutational events in LMP1 gene of Epstein-Barr virus in salivary gland lymphoepithelial carcinomas. Int J Cancer 105: 654-660, 2003 Kang I, Quan T, Nolasco H: Defective control of latent Epstein- Barr virus infection in systemic lupus erythematosus. J Immunol 172: 1287-1294, 2004 Kotsianti A, Costopoulos J, Morgello S, Papadimitriou C: Undifferentiated carcinoma of the parotid gland in a white patient: detection of Epstein-Barr virus by in situ hybridization. Hum Pathol 27: 87-90, 1996 Kuo T, Hsue C: Lymphoepithelioma-like salivary gland carcinoma in Taiwan: a clinicopathological study of nine cases demonstrating a strong association with Epstein-Barr virus. Histopathology 31: 75-82, 1997 Leung SY, Chung LP, Yuen ST: Lymphoepithelial carcinoma of the salivary gland: in situ detection of EBV. J Clin Pathol 48:1022-1027, 1995 Lin CL, Lo WF, Lee TH: Immunization with EBV peptidepulsed dendritic cells induces functional CD8+T-Cell immunity and may lead to tumor regression in EBV-positive NPC. Cancer Res 62:6952-6958, 2002 Niedobitek G, Meru N, Delecluse HJ: Epstein-Barr virus infection and human malignancies. Int J Exp Pathol 82:149-170, 2001 Quintanilla-Martinez L, Gamboa-Dominguez A, Gamez-Ledesma I, et al: Prevalencia alta del virus de Epstein-Barr en una poblacion mexicana con enfermedad de Hodgkin. Rev Invest Clin 46: 355-362, 1994 Saku T, Cheng J, Jen KY, et al: Epstein-Barr virus infected lymphoepithelial carcinomas of the salivary gland in the Russia- Asia area: a clinicopathologic study of 160 cases. Arkh Patol 65: 35-39, 2003 Tsai C-C, Chen C-L, Hsu H-C: Expression of Epstein Barr virus in carcinomas of salivary glands: a strong association with lymphoepithelioma-like carcinoma. Hum Pathol 27:258- 262, 1996 Villa AR, Kraus A, Jimenez-Corona A: Malignant neoplasms in autoimmune rheumatic diseases: examination of the risk of developing a malignancy among five different rheumatic diseases in one institution. J Clin Rheumatol 6:176-183, 2000 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2006 VL 12 IS 1 BP 41 EP 45 PG 5 ER PT J AU Sobel, G Halasz, J Bogdanyi, K Szabo, I Borka, K Molnar, P Schaff, Zs Paulin, F Banhidy, F AF Sobel, Gabor Halasz, Judit Bogdanyi, Katalin Szabo, Istvan Borka, Katalin Molnar, Peter Schaff, Zsuzsa Paulin, Ferenc Banhidy, Ferenc TI Prenatal Diagnosis of a Giant Congenital Primary Cerebral Hemangiopericytoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE congenital brain tumors; infantile hemangiopericytoma ID congenital brain tumors; infantile hemangiopericytoma AB Congenital primary intracranial hemangiopericytomas are exceptionally rare tumors. We present a case of a fetus, with the prenatal sonogram at 33 weeks of gestation revealing a large cerebral tumor. Because of the enlarged head, a cesarean section was performed. The tumor was confirmed by postnatal ultrasound, magnetic resonance imaging (MRI) and biopsy. Elevated intracranial pressure and hemorrhage led to death on the 11th day. Autopsy revealed a 10x9 cm large inhomogeneous tumor located centrally, mainly in the posterior fossa. Histology showed a hypercellular and hypervascular tumor with extended necrosis and high mitotic rate. The tumor cells were positive for vimentin and CD34 antigens and negative for several neurological markers, desmin and CD31. The diagnosis of a congenital primary cerebral hemangiopericytoma was confirmed. C1 [Sobel, Gabor] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Halasz, Judit] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Bogdanyi, Katalin] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Szabo, Istvan] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Borka, Katalin] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Molnar, Peter] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Paulin, Ferenc] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Banhidy, Ferenc] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM schaff@korb2.sote.hu CR Janisch W, Haas JF, Schreiber D, et al: Primary central nervous system tumors in stillborns and infants. Epidemiological considerations. J Neurooncol 2: 113-116, 1984 Jellinger K, Sunder-Plassmann M: Connatal intracranial tumours. Neuropaediatrie 4: 46-63, 1973 Lee D-Y, Kim Y-M, Yoo S-J, et al: Congenital glioblastoma diagnosed by fetal sonography. Child’s Nerv Syst 15: 197-201, 1999 Nakayama K, Nakamura Y: Localization of congenital glioblastomas in the Japanese: a case report and review of the literature. Child’s Nerv Syst 18: 149-152, 2002 Winters JL, Wilson D, Davis DG: Congenital glioblastoma multiforme: a report of three cases and a review of the literature. J Neurol Sci 188: 13-19, 2001 Leins AM, Kainer R, Weis S, et al: Sonography and neuropathology of a congenital brain tumor: report of a rare incident. Ultrasound Obstet Gynecol 17: 245-247, 2001 Aouad N, Vital C, Rivel J, et al: Giant supratentorial meningeal haemangiopericytoma in a newborn. Acta Neurochirurg 112: 154-156, 1991 Herzog CE, Leeds NE, Bruner JM, et al: Intracranial hemangiopericytomas in children. Ped Neurosurg 22: 274-279, 1995 Hodaie M, Becker L, Teshima I, et al: Total resection of an intracerebral hemangioendothelioma in an infant. Case report and review of the literature. Pediatr Neurosurg 34: 104-112, 2001 Kirk IR, Dominguez R, Castillo M: Congenital primary cerebral angiosarcoma: CT, US, and MR findings. Pediatr Radiol 22: 134-135, 1992 Mena H, Ribas JL, Enzinger FM, et al: Primary angiosarcoma of the central nervous system. J Neurosurg 75: 73-76, 1991 Suzuki Y, Yoshida YK, Shirane R, et al: Congenital primary cerebral angiosarcoma. J Neurosurg 92: 466-468, 2000 Alen JF, Lobato RD, Gomez PA, et al: Intracranial hemangiopericytoma: study of 12 cases. Acta Neurochir, Wien, 143: 575-586, 2001 Calonje E, Fletcher CDM: Tumors of blood vessels and lymphatics. In: Diagnostic Histopathology of Tumors., Eds: Fletcher CDM), Vol. 1, Second ed., Churchill Livingstone, London, 2000, pp. 45-86 Ordonez NG, Mackay B, el-Naggar AK, et al: Congenital hemangiopericytoma. An ultrastructural, immunocytochemical, and flow cytometric study. Arch Pathol Lab Med 117: 934-937, 1993 Johnson MD, Mitchell AR, Troup EC, et al: Congenital cystic hemangioblastomas of the cerebral hemisphere in a neonate without alteration in the VHL gene. Pediatr Neurosurg 40: 124-127, 2004 Weiss SW, Goldblum JR: Perivascular tumors. In: Enzinger and Weiss’s Soft Tissue Tumors., Eds: Weiss SW and Goldblum JR), Fourth ed., Mosby, St. Louis, 2001, pp. 985-1037 Beckner ME, Sasatomi E, Swalsky PA, et al: Loss of heterozygosity reveals non-VHL allelic loss in hemangioblastomas at 22q13. Hum Pathol 35: 1105-1111, 2004 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2006 VL 12 IS 1 BP 46 EP 49 PG 4 ER PT J AU Culhaci, N Ozkara, E Yuksel, H Ozsunar, Y Unal, E AF Culhaci, Nil Ozkara, Esra Yuksel, Hasan Ozsunar, Yelda Unal, Emel TI Spontaneously Ruptured Uterine Angioleiomyoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE angioleiomyoma; uterus; genital tract ID angioleiomyoma; uterus; genital tract AB Angioleiomyoma is an uncommon type of leiomyoma of the uterus that originates from smooth muscle cells and contains thick-walled vessels. A 45-year-old woman with the complaint of lower abdominal pain was admitted to the hospital. In the operation a ruptured, bleeding uterine tumor was seen. She underwent total hysterectomy and bilateral salpingo-oophorectomy. On pathologic examination of the specimen, the tumor was diagnosed as angioleiomyoma. Here, an unusual clinical presentation of uterine angioleiomyoma was reported. C1 [Culhaci, Nil] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Ozkara, Esra] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. [Yuksel, Hasan] Adnan Menderes University, Faculty of Medicine, Department of Obstetrics and GynecologyAydin, Turkey. [Ozsunar, Yelda] Adnan Menderes University, Faculty of Medicine, Department of RadiologyAydin, Turkey. [Unal, Emel] Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. RP Culhaci, N (reprint author), Adnan Menderes University, Medical Faculty, Pathology Department, 09100 Aydin, Turkey. EM nculhaci@adu.edu.tr CR Weiss SW, Goldblum JR: Benign tumors of smooth muscle. In: Enzinger and Weiss’s soft tissue tumors. 4th ed., Mosby, St. Louis, 2001, pp 699-700 Zaloudek C, Hendrickson MR: Mesenchymal tumors of the uterus. In: Blaustein’s Pathology of the Female Genital Tract., Ed.: Kurman R), 5th ed, Springer-Verlag, New York, 2002, pp 574 Hennig Y, Caselitz J, Stern C, et al: Karyotype evolution in a case of uterine angioleiomyoma. Cancer Genet Cytogenet 108: 79-80, 1999 Wang CP, Chang YL, Sheen TS: Vascular leiomyoma of the head and neck. Laryngoscope 114: 661-665, 2004 Ide F, Mishima K, Saito I: Angiomyoma in the submandibular gland: a rare location for a ubiquitous tumour. J Laryngol Otol 117: 1001-1002, 2003 Hsieh CH, Lui CC, Huang SC, et al: Multiple uterine angioleiomyomas in a woman presenting with severe menorrhagia. Gynecol Oncol 90: 348-352, 2003 Stewart EA, Nowak RA: Leiomyoma-related bleeding: a classic hypothesis updated for the molecular era. Hum Reprod Update 2: 295-306, 1996 Agorastos T, Dinas K, Patsiaoura K: Cystic degenerated angioleiomyoma mimicking ovarian pathology. Acta Obstet Gynecol Scand 80: 863-865, 2001 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2006 VL 12 IS 1 BP 50 EP 51 PG 2 ER PT J AU Szilasi, M Dolinay, T Nemes, Z Strausz, J AF Szilasi, Maria Dolinay, Tamas Nemes, Zoltan Strausz, Janos TI Pathology of Chronic Obstructive Pulmonary Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE chronic bronchitis; obstructive bronchiolitis; emphysema; inflammation ID chronic bronchitis; obstructive bronchiolitis; emphysema; inflammation AB Chronic obstructive pulmonary disease is one of the leading causes of death and morbidity worldwide. Despite intensive investigation, its pathology and pathophysiology are not well understood. The hallmarks of the disease are irreversible airflow limitation and chronic inflammation. Small airway obstruction due to progressive inflammation and fibrosis, and the loss of elastic recoil mediated by elastolysis and apoptosis equally contribute to pathologic changes. However, it is debated to what extent the obstruction of large airways leads to altered lung function. Three morphologic entities are described in the literature under one disease; chronic bronchitis, obstructive bronchiolitis and emphysema may appear in the same patient at the same time. The authors review pathologic changes observed in chronic obstructive pulmonary disease, including acute exacerbations and secondary pulmonary hypertension as severe but common complications of the disease. Furthermore, we detail recent scientific evidences for major cellular and molecular inflammatory pathway activation. These mechanisms result in accelerated apoptosis, remodeling and increased proinflammatory cytokine release. Targeting intracellular pathological changes may lead to the discovery of a new generation of drugs that could reduce chronic obstruction before airway irreversibility is established. C1 [Szilasi, Maria] University of Debrecen Medical and Health Science Center, Department of Pulmonary Medicine, Moricz Zs. krt. 22., H-4004 Debrecen, Hungary. [Dolinay, Tamas] University of Debrecen Medical and Health Science Center, Department of Pulmonary Medicine, Moricz Zs. krt. 22., H-4004 Debrecen, Hungary. [Nemes, Zoltan] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Strausz, Janos] National Koranyi Institute of PulmonologyBudapest, Hungary. RP Szilasi, M (reprint author), University of Debrecen Medical and Health Science Center, Department of Pulmonary Medicine, H-4004 Debrecen, Hungary. EM mszilasi@jaguar.unideb.hu CR Aoshiba K, Yokohori N, Nagai A: Alveolar wall apoptosis causes lung destruction and emphysematous changes. Am J Respir Cell Mol Biol 28: 555-562, 2003 Boszormenyi Nagy G: Chronic obstructive pulmonary disease. In: Epidemiology and Functional Data in Pulmonology Institutions in year 2004 [book in Hungarian], Eds: Jonas J, Fodor K, Turgyei M and Nyari L), Orszagos Koranyi TBC es Pulmonologiai Intezet, 2005, pp. 50-56 Chodosh S: Treatment of acute exacerbations of chronic bronchitis: state of the art. Am J Med 91: 87S, 1991 Daheshia M: Therapeutic inhibition of matrix metalloproteinases for the treatment of chronic obstructive pulmonary disease, COPD). Curr Med Res Opin 21: 587-593, 2005 de Dodoy I, Donahoe M, Calhoun WJ, et al: Elevated TNFalpha production by peripheral blood monocytes of weightloosing COPD patients. Am J Respir Crit Care Med 153: 633- 637, 1996 Deshmukh HS, Case LM, Wesselkamper SC: Metalloproteinases mediate mucin 5AC expression by epidermal growth factor receptor activation. Am J Respir Crit Care Med 171: 305-314, 2005 Di Stefano A, Capelli A, Lusuardi M, et al: Severity of airflow limitation is associated with severity of airway inflammation in smokers. Am J Respir Crit Care Med 158: 1277-1285, 1998 Drost EM, Skwarski J, Sauleda J, et al: Oxidative stress and airway inflammation in severe extracerbations of COPD. Thorax 60: 293-300, 2005 Enelow RI, Mohammed AZ, Stoler MH, et al: Structural and functional consequences of alveolar cell recognition by CD8+ T lymphocytes in experimental lung disease. J Clin Invest 102: 1653-1661, 1998 Fletcher C, Peto R, Tinker C, et al: The natural history of chronic bronchitis and emphysema: an eight-year study of early chronic obstructive lung disease in working men in London. Oxford University Press, Oxford, England: 93, 1976 Giaid A, Yanagisawa M, Langleben D, et al: Expression of endothelin-1 in lungs of patients with pulmonary hypertension. N Engl J Med 328: 1732-1739, 1993 Gump DW, Philips CA, Forsyth BR: Role of infection in chronic bronchitis. Am Rev Respir Dis 113: 465, 1976 Hansel TT, Barnes PJ, Celli BR: Introduction: definitions, burden and causation. In: An Atlas of Chronic Obstructive Pulmonary Disease, Eds: Hansel TT, Barnes PJ and Celli BR), The Parthenon Publishing Group, 2004, pp. 1-19 Haraguchi M, Shimura S, Shirato K: Morphometric analysis of bronchial cartilage in chronic obstructive pulmonary disease and bronchial asthma. Am J Respir Crit Care Med 159: 1005- 1013, 1999 Hogg JC, Chu F, Utokaparch S, et al: The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med 350: 2645-2653, 2004 Hogg JC, Macklem PT, Thurlbeck WM: Site and nature of airway obstruction in chronic obstructive lung disease. N Engl J Med 278: 1355-1360, 1968 Hubbard RC, Fells G, Gadek J, et al: Neutrophil accumulation in the lung in alpha-1 antitrypsin deficiency. Spontaneous release of leukotriene B4 by alveolar macrophages. J Clin Invest 88: 891-897, 1991 Jeffery PK: Remodeling in asthma and chronic obstructive lung disease. Am J Respir Crit Care Med 164: S28-S38, 2001 Kasahara Y, Tuder RM, Cool CD, et al: Endothelial cell death and decreased expression of vascular endothelial growth factor and vascular endothelial growth factor receptor 2 in emphysema. Am J Respir Crit Care Med 163: 737-744, 2001 Kasahara Y, Tunder RM, Taraseviciene-Stewart L, et al: Inhibition of VEGF receptors causes lung cell apoptosis and emphysema. J Clin Invest 106: 1311-1319, 2000 Keatings VM, Collins PD, Scott DM, et al: Differences in interleukin- 8 and tumor necrosis factor-alpha in induced sputum from patients with chronic obstructive pulmonary disease or asthma. Am J Respir Crit Care Med 153: 530-534, 1996 Kim WD, Eidelman DH, Izquierdo JL, et al: Centrilobular and panlobular emphysema in smokers. Two distinct morphological and functional entities. Am Rev Respir Dis 144: 1385-1390, 1991 Kranenburg AR, de Boer WI, Alagappan VK, et al: Enhanced bronchial expression of vascular endothelial growth factor and receptors, Flk-1 and Flt-1, in patients with chronic obstructive pulmonary disease. Thorax 60: 106-113, 2005 Ladner KJ, Caligiuri MA, Guttridge DC: TNF regulated biphasic activation of NF-kB is required for cytokine-induced loss of skeletal muscle gene products. J Biol Chem 24278: 2294-2303, 2003 Lams BE, Sousa AR, Rees PJ, Lee TH: Immunopathology of the small-airway submucosa in smokers with and without chronic obstructive pulmonary disease. Am J Respir Crit Care Med 158: 1518-1523, 1998 Lams BE, Sousa AR, Rees PJ, Lee TH: Subepithelial immunopathology of the large airways in smokers with and without chronic obstructive pulmonary disease. Eur Respir J 15: 512- 516, 2000 Langen RC, Schols AM, Kelders MC, et al: Inflammatory cytokines inhibit myogenic differentiation through activation of nuclear factor-kappaB. Faseb J 15: 1169-1180, 2001 Lees AW, McNaught W: Bacteriology of lower respiratory tract secretions, sputum and upper respiratory tract secretions in "normals" and chronic bronchitis. Lancet 2: 1112, 1959 Li YP, Schwartz RJ, Waddell ID, et al: Skeletal muscle myocytes undergo protein loss and reactive oxygen-mediated NFkappa B activation in response to tumor necrosis factor alpha. Faseb J 12: 871-880, 1998 Liu AN, Mohammed AZ, Rice WR, et al: Perforin-independent CD8(+, T-cell-mediated cytotoxicity of alveolar epithelial cells is preferentially mediated by tumor necrosis factor-alpha. Am J Respir Cell Mol Biol 20: 849-858, 1999 Matsuba K, Thurlbeck WM: The number and dimension of small airways in emphysematous lungs. Am J Pathol 67: 265- 275, 1972 McCrory DC, Brown C, Gelfand SE, et al: Management of acute exacerbation of COPD: a summary and appraisal of published evidence. Chest 119: 1190-1209, 2001 Medical Research Council: Definition and classification of chronic bronchitis for clinical and epidemiological purposes. Lancet i: 775-779, 1965 Miotto D, Hollenberg MD, Bunnett NW, et al: Expression of protease activated receptor-2, PAR-2, in central airways of smokers and non-smokers. Thorax 57: 146-151, 2002 NHLBI/WHO workshop report: Global initiative for chronic obstructive lung disease. National Heart, Lung and Blood Institution Publication Nr. 2701, 2001 Niewoehner DE, Klienerman J, Rice D: Pathologic changes in the peripheral airways of young cigarette smokers. N Engl J Med 291: 755-758, 1974 Ning W, Li CJ, Kaminski N, et al: Comprehensive gene expression profiles reveal pathways related to the pathogenesis of chronic obstructive pulmonary disease. Proc Natl Acad Sci U S A 101: 14895-14900, 2004 O'Shaughnessy TC, Ansari TW, Barnes NC, et al: Reticular basement membrane thickness in moderately severe asthma and smokers' chronic bronchitis with or without airflow obstruction [abstract]. Am J Respir Crit Care Med 153: A879, 1996 Oudijk EJ, Lammers JW, Koenderman L: Systemic inflammation in chronic obstructive pulmonary disease. Eur Respir J Suppl 46: 5s-13s, 2003 Payne D, Roger AV, Jaffe A, et al: Reticular basement membrane thickness in children with severe asthma [abstract]. Arch Dis Child 82: A42, 2000 Peinado VI, Barbera JA, Abate P, et al: Inflammatory reaction in pulmonary muscular arteries of patients with mild chronic obstructive pulmonary disease. Am J Respir Crit Care Med 159: 1605-1611, 1999 Qiu Y, Zhu J, Bandi V, et al: Biopsy neutrophilia, neutrophil chemokine and receptor gene expression in severe exacerbations of chronic obstructive pulmonary disease. Am J Respir Crit Care Med 168: 968-975, 2003 Reid L: Measurement of bronchial mucous gland layer: a diagnostic yardstick in chronic bronchitis. Thorax 6: 132-141, 1960 Reid L: Pathology of chronic bronchitis. Lancet 266: 274-278, 1954 Richter A, O'Donnell RA, Powell RM, et al: Autocrine ligands for the epidermal growth factor receptor mediate interleukin-8 release from bronchial epithelial cells in response to cigarette smoke. Am J Respir Cell Mol Biol 27: 85-90, 2002 Rohde G, Wiethege A, Borg I: Respiratory viruses in exacerbations of chronic obstructive pulmonary disease requiring hospitalization: a case-control study. Thorax 58: 37, 2003 Rooks SW, Bayley DL, Hill SL, et al: Bronchial inflammation in acute bacterial exacerbation of chronic bronchitis: the role of leukotriene B4. Eur Respir J 15: 274-280, 2000 Saetta M: Airway inflammation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 160: S17-S20, 1999 Saetta M, Baraldo S, Corbino L, et al: CD8+ve cells in the lungs of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 160: 711-717, 1999 Saetta M, Di Stefano A, Maestrelli P, et al: Activated T-lymphocytes and macrophages in bronchial mucosa of subjects with chronic bronchitis. Am Rev Respir Dis 147: 301-306, 1993 Saetta M, Di Stefano A, Maestrelli P, et al: Airway eosinophilia in chronic bronchitis during exacerbations. Am J Respir Crit Care Med 150: 1646-1652, 1994 Saetta M, Di Stefano A, Turato G, et al: CD8+ T-lymphocytes in peripheral airways of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 157: 822-826, 1998 Saetta M, Turato G, Baraldo S, et al: Goblet cell hyperplasia and epithelial inflammation in peripheral airways of smokers with both symptoms of chronic bronchitis and chronic airflow limitation. Am J Respir Crit Care Med 161: 1016-1021, 2000 Saetta M, Turato G, Maestrelli P, et al: Cellular and structural bases of chronic obstructive pulmonary disease. Am J Respir Crit Care Med 163: 1304-1309, 2001 Saint S, Bent S, Vittinghoff E, et al: Antibiotics in chronic obstructive pulmonary disease exacerbation: A meta-analysis. JAMA 273: 957-960, 1995 Santos S, Peinado VI, Ramirez J, et al: Enhanced expression of vascular endothelial growth factor in pulmonary arteries of smokers and patients with moderate chronic obstructive pulmonary disease. Am J Respir Crit Care Med 167: 1250-1256, 2003 Segura-Valdez L, Pardo A, Gaxiola M: Upregulation of gelatinases A and B, collagenases 1 and 2, and increased parenchymal cell death in COPD. Chest 117: 684-694, 2000 Seti S, Evans N, Grant BJ, et al: New strains of bacteria and exacerbations of chronic obstructive pulmonary disease. N Engl J Med 347: 465-473, 2002 Snider GL: Chronic obstructive pulmonary disease: a continuing challenge. Am Rev Respir Dis 133: 942-944, 1986 Takeyama K, Dabbagh K, Lee HM, et al: Epidermal growth factor system regulates mucin production in airways. Proc Natl Acad Sci U S A 96: 3081-3086, 1999 ten Hacken NH, Postma DS, Timens W: Are asthma and chronic obstructive pulmonary disease different diseases?--Con. Monaldi Arch Chest Dis 54: 551-558, 1999 Tuder RM, Zhen L, Cho CY, et al: Oxidative stress and apoptosis interact and cause emphysema due to vascular endothelial growth factor receptor blockade. Am J Respir Cell Mol Biol 29: 88-97, 2003 Vestbo J, Lange P: Can GOLD Stage 0 provide information of prognostic value in chronic obstructive pulmonary disease? Am J Respir Crit Care Med 66: 329-332, 2002 Vestbo J, Prescott E, Lange P: Association of chronic mucus hypersecretion with FEV1 decline and chronic obstructive pulmonary disease morbidity. Am J Respir Crit Care Med 153: 1530-1535, 1996 Vlahovic G, Russell ML, Mercer RR, et al: Cellular and connective tissue changes in alveolar septal wall in emphysema. Am J Respir Crit Care Med 160: 2086-2092, 1999 Vliagoftis H, Schwingshackl A, Milne CD, et al: Proteinaseactivated receptor-2-mediated matrix metalloproteinase-9 release from airway epithelial cells. J Allergy Clin Immunol 106: 537-545, 2000 Zheng T, Kang MJ, Crothers K, et al: Role of cathepsin Sdependent epithelial cell apoptosis in IFN-gamma-induced alveolar remodeling and pulmonary emphysema. J Immunol 174: 8106-8115, 2005 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2006 VL 12 IS 1 BP 52 EP 60 PG 9 ER PT J AU Kopper, L Timar, J AF Kopper, Laszlo Timar, Jozsef TI Professor Karoly LAPIS, a Hungarian pathologist, is 80 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor ProgressionBudapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2006 VL 12 IS 1 BP 61 EP 61 PG 1 ER PT J AU Sulkowska, M Golaszewska, J Wincewicz, A Koda, M Baltaziak, M Sulkowski, S AF Sulkowska, Mariola Golaszewska, Jolanta Wincewicz, Andrzej Koda, Mariusz Baltaziak, Marek Sulkowski, Stanislaw TI Leptin - From Regulation of fat Metabolism to Stimulation of Breast Cancer Growth SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE leptin; leptin receptor; signal transducer and activator of transcription 3; estrogen receptor alpha; antiestrogen resistance ID leptin; leptin receptor; signal transducer and activator of transcription 3; estrogen receptor alpha; antiestrogen resistance AB Leptin restricts intake of calories as a satiety hormone. It probably stimulates neoplastic proliferation in breast cancer, too. Growth of malignant cells could be regulated by various leptin-induced second messengers like STAT3 (signal transducers and activators of transcription 3), AP-1 (transcription activator protein 1), MAPK (mitogen-activated protein kinase) and ERKs (extracellular signal-regulated kinases). They seem to be involved in aromatase expression, generation of estrogens and activation of estrogen receptor ? (ER?) in malignant breast epithelium. Leptin may maintain resistance to antiestrogen therapy. Namely, it increased activation of estrogen receptors, therefore, it was suspected to reduce or even overcome the inhibitory effect of tamoxifen on breast cell proliferation. Although several valuable reviews have been focused on the role of leptin in breast cancer, the status of knowledge in this field changes quickly and our insight should be continuously revised. In this summary, we provide refreshed interpretation of intensively reported scientific queries of the topic. C1 [Sulkowska, Mariola] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Golaszewska, Jolanta] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Wincewicz, Andrzej] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Koda, Mariusz] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Baltaziak, Marek] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Sulkowski, Stanislaw] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. RP Sulkowski, S (reprint author), Medical University of Bialystok, Department of General Pathomorphology, 15-269 Bialystok, Poland. EM sulek@zeus.amb.edu.pl CR Zhang Y, Proenca R, Maffei M, et al: Positional cloning of the mouse obese gene and its human homologue. Nature 372: 425- 432, 1994. Tartaglia LA: The leptin receptor. J Biol Chem 272: 6093- 6096, 1997. Gaja A and Chury Z: The importance of leptin in oncology— hypothesis or facts? Vnitr Lek 47: 245-249, 2001. Tessitore L, Vizio B, Pesola D, et al: Adipocyte expression and circulating levels of leptin increase in both gynaecological and breast cancer patients. Int J Oncol 24: 1529-1535, 2004. O’Brien SN, Welter BH and Price TM: Presence of leptin in breast cell lines and breast tumors. Biochem Biophys Res Commun 259: 695-698, 1999. Das UN: Is obesity an inflammatory condition? Nutrition 17: 953-966, 2001. Cleary MP, Grande JP, Juneja SC and Maihle NJ: Dietinduced obesity and mammary tumor development in MMTVneu female mice. Nutr Cancer 50: 174-180, 2004. Szymczak J, Milewicz A, Thijssen JH, et al: Concentration of sex steroids in adipose tissue after menopause. Steroids 63: 319-321, 1998. Surmacz E and Bartucci M: Role of estrogen receptor alpha in modulating IGF-I receptor signaling and function in breast cancer. J Exp Clin Cancer Res 23: 385-394, 2004. Koda M, Sulkowski S, Kanczuga-Koda L, et al: Expression of ERalpha, ERbeta and Ki-67 in primary tumors and lymph node metastases in breast cancer. Oncol Rep 11: 753-759, 2004. Stattin P, Soderberg S, Biessy C, et al: Plasma leptin and breast cancer risk: a prospective study in northern Sweden. Breast Cancer Res Treat 86: 191-196, 2004. Somasundar P, Yu AK, Vona-Davis and McFadden DW: Differential effects of leptin on cancer in vitro. J Surg Res 113: 50- 55, 2003. Caldefie-Chezet F, Damez M, de Latour M, et al: Leptin: A proliferative factor for breast cancer? Study on human ductal carcinoma. Biochem Biophys Res Commun 334: 737-741, 2005. Markowska A, Malendowicz K and Drews K: The role of leptin in breast cancer. Eur J Gynaecol Oncol 25: 192-194, 2004. Cleary MP, Phillips FC, Getzin SC, et al: Genetically obese MMTV-TGF-alpha/Lep(ob)Lep(ob, female mice do not develop mammary tumors. Breast Cancer Res Treat 77: 205-215, 2003. Ishikawa M, Kitayama J and Nagawa H: Enhanced expression of leptin and leptin receptor, OB-R, in human breast cancer. Clin Cancer Res 10: 4325-4331, 2004. Goodwin PJ, Ennis M, Fantus IG, et al: Is leptin a mediator of adverse prognostic effects of obesity in breast cancer? J Clin Oncol 23: 6037-6042, 2005. Miyoshi Y, Funahashi T, Tanaka S, et al: High expression of leptin receptor mRNA in breast cancer tissue predicts poor prognosis for patients with high, but not low, serum leptin levels. Int J Cancer 118: 1414-1419, 2006. Mantzoros CS, Bolhke K, Moschos S and Cramer DW: Leptin in relation to carcinoma in situ of the breast: a study of premenopausal cases and controls. Int J Cancer 80: 523-526, 1999. Sauter ER, Garofalo C, Hewett J, et al: Leptin expression in breast nipple aspirate fluid, NAF, and serum is influenced by body mass index, BMI, but not by the presence of breast cancer. Horm Metab Res 36: 336-340, 2004. Kumar S, Kishimoto H, Chua HL, et al: Interleukin-1 alpha promotes tumor growth and cachexia in MCF-7 xenograft model of breast cancer. Am J Pathol 163: 2531-2541, 2003. Catalano S, Marsico S, Giordano C, et al: Leptin enhances, via AP-1, expression of aromatase in the MCF-7 cell line. J Biol Chem 278: 28668-28676, 2003. Catalano S, Mauro L, Marsico S, et al: Leptin induces, via ERK1/ERK2 signal, functional activation of estrogen receptor alpha in MCF-7 cells. J Biol Chem 279: 19908-19915, 2004. Masso-Welch PA, Zangani D, Ip C, et al: Isomers of conjugated linoleic acid differ in their effects on angiogenesis and survival of mouse mammary adipose vasculature. J Nutr 134: 299- 307, 2004. Yin N, Wang D, Zhang H, et al: Molecular mechanisms involved in the growth stimulation of breast cancer cells by leptin. Cancer Res 64: 5870-5875, 2004. Hu X, Juneja SC, Maihle NJ and Cleary MP: Leptin—a growth factor in normal and malignant breast cells and for normal mammary gland development. J Natl Cancer Inst 94: 1704- 1711, 2002. Laud K, Gourdou I, Pessemesse L, et al: Identification of leptin receptors in human breast cancer: functional activity in the T47-D breast cancer cell line. Mol Cell Endocrinol 188: 219- 226, 2002. Dieudonne MN, Machinal-Quelin F, Serazin-Leroy V, et al: Leptin mediates a proliferative response in human MCF7 breast cancer cells. Biochem Biophys Res Commun 293: 622-628, 2002. Magoffin DA, Weitsman SR, Aagarwal SK and Jakimiuk AJ: Leptin regulation of aromatase activity in adipose stromal cells from regularly cycling women. Ginekol Pol 70: 1-7, 1999. Garofalo C, Sisci D and Surmacz E: Leptin interferes with the effects of the antiestrogen ICI 182,780 in MCF-7 breast cancer cells. Clin Cancer Res 10: 6466-6475, 2004. Nunez NP, Jelovac D, Macedo L, et al: Effects of the antiestrogen tamoxifen and the aromatase inhibitor letrozole on serum hormones and bone characteristics in a preclinical tumor model for breast cancer. Clin Cancer Res 10: 5375-5380, 2004. Cleary MP, Juneja SC, Phillips FC, et al: Leptin receptor-deficient MMTV-TGF-alpha/Lepr(db)Lepr(db, female mice do not develop oncogene-induced mammary tumors. Exp Biol Med 229: 182-193, 2004. Coskun U, Gunel N, Toruner FB, et al. Serum leptin, prolactin and vascular endothelial growth factor, VEGF, levels in patients with breast cancer. Neoplasma 50: 41-46, 2003. Ozet A, Arpaci F, Yilmaz MI, et al. Effects of tamoxifen on the serum leptin level in patients with breast cancer. Jpn J Clin Oncol 31: 424-427, 2001. Marttunen MB, Andersson S, Hietanen P, et al. Antiestrogenic tamoxifen and toremifene increase serum leptin levels in postmenopausal breast cancer patients. Maturitas 35: 175-179, 2000. Gunel N, Coskun U, Toruner FB, et al: Serum leptin levels are associated with tamoxifen-induced hepatic steatosis. Curr Med Res Opin 19: 47-50, 2003. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 69 EP 72 PG 4 ER PT J AU Szekely, E Torok, V Szekely, T Riesz, P Romics, I AF Szekely, Eszter Torok, Virag Szekely, Tamas Riesz, Peter Romics, Imre TI E-Cadherin Expression in Transitional Cell Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bladder cancer; E-cadherin; transitional cell carcinoma ID bladder cancer; E-cadherin; transitional cell carcinoma AB The authors analyzed the expression of E-cadherin, one of the most important cell adhesion molecules, on histological slides of tumors of bladder cancer patients. The aim of the study was to see whether there is any association between E-cadherin expression and tumor grade, stage, age and gender of the patients, number of recurrences, or overall survival. The samples were examined in 51 primary bladder transitional cell carcinomas (TCC) of 50 patients, resected by transurethral resection (TUR) between January 1, 1996 and January 1, 1997. Immunoreactions were performed with monoclonal anti-human E-cadherin antibody. Forty of the fifty patients could be clinically followed. The analysis of the results on these forty patients was performed by contingency analysis and significance was assessed by ?2 test. No significant association between E-cadherin expression and tumor grade, stage, age or gender of the patients, the number of recurrences, or overall survival could be seen. C1 [Szekely, Eszter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Torok, Virag] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Szekely, Tamas] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Riesz, Peter] Semmelweis University, Department of UrologyBudapest, Hungary. [Romics, Imre] Semmelweis University, Department of UrologyBudapest, Hungary. RP Szekely, E (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM szeszter@gmail.com CR Acs G, LiVolsi VA: Loss of membrane expression of E-cadherin in leukemic erythroblasts. Arch Pathol Lab Med 125:198–201, 2001 Bringuier PP, Giroldi LA, Umbas R et al: Mechanisms associated with abnormal E-cadherin immunoreactivity in human bladder tumors. Int J Cancer 83: 591-595, 1999 Bringuier PP, Umbas R, Schaafsma HE et al: Decreased E-cadherin immunoreactivity correlates with poor survival inpatients with bladder tumors. Cancer Res 53: 3241-3245, 1993 Conacci-Sorrell M, ZhurinskyJ, Ben-Ze’ev A.: The cadherincatenin adhesion system in signaling and cancer. J Clin Invest 109:987-991, 2002 Fearon ER.: Connecting estrogen receptor function, transcriptional repression, and E-cadherin expression in breast cancer. Cancer Cell 3: 307-310, 2003 Giroldi LA, Bringuier PP, Shimazu T et al: Changes in cadherin- catenin complexes in the progression of human bladder carcinoma. Int J Cancer 82: 70-76, 1999 Han AC, Soler AP, Tang CK et al: Nuclear localization of Ecadherin expression in Merkel cell carcinoma. Arch Pathol Lab Med 124: 1147–1151, 2004 Handschuh G, Candidus S, Luber et al: Tumour-associated Ecadherin mutations alter cellular morphology, decrease cellular adhesion and increase cellular motility Oncogene 18:4301- 4312, 1999 Horikawa Y, Sugano K, Shigyo M et al: Hypermethylation of an E-cadherin, CDH1, promoter region in high grade transitional cell carcinoma of the bladder comprising carcinoma in situ. J Urol 169:1541-1545, 2003 Jou TS, Stewart DP, Stappert J et al: Genetic and biochemical dissection of protein linkages in the cadherin-catenin complex. Proc Natl Acad Sci USA 92: 5067-5071, 1995 Koksai IT, Ates M, Danisman A et al: Reduced E-cadherin and a-catenin expressions have no prognostic role in bladder carcinoma. Pathol Oncol Res 12: 13-19, 2006 Koksai IT, Ozcan F, Kilicaslan I et al: Expression of E-cadherin in prostate cancer in formalin-fixed, paraffin-embedded tissues: correlation with pathological features. Pathology 34 : 233-238, 2002 Krishnadath KK, Tilanus HW, Blankenstein M et al: Reduced expression of the cadherin-catenin complex in oesophageal adenocarcinoma correlates with poor prognosis. J Pathol 182: 331-338, 1997 Landis SH, Murray T, Bolden S: Cancer statistics. CA Cancer J Clin 49: 8-31, 1999 Laskin WB, Miettinen M: Epithelial-type and neural-type cadherin expression in malignant noncarcinomatous neoplasms with epithelioid features that involve the soft tissues. Arch Pathol Lab Med 126:425–431, 2002 Lipponen PK, Eskelinen MJ: Reduced expression of E-cadherin is related to invasive disease and frequent recurrence in bladder cancer. J Cancer Res Clin Oncol 121: 303-308, 1995 Mialhe A, Louis J, Pasquier D et al: Expression of E-cadherin and alpha-, beta-, and gamma-catenins in human bladder carcinomas: are they good prognostic factors? Invasion Metastasis 17: 124-137, 1997 Mialhe A, Louis J, Pasquier D et al: Expression of three cell adhesion molecules in bladder carcinomas: correlation with pathological features. Anal Cell Pathol 14:1225-1227, 1997 Ozawa M, Ringwald M, Kemler R: Uvomorulin-catenin complex formation is regulated by a specific domain in the cytoplasmic region in the cell adhesion molecule. Proc Natl Acad Sci USA 87:4246-4250 , 1990 Ropke M, Boltze C, Neumann HW et al: Genetic and epigenetic alteration in tumor progression in a dedifferentiated chondrosarcoma. Pathol Res Pract 199:437-444, 2003 Ross JS, del Rosario AD, Figge HL et al: E-cadherin expression in papillary transitional cell carcinoma of the urinary bladder. Hum Pathol 26: 940-944, 1995 Soloway MS: Intravesical therapy for bladder cancer. Urol Clin North Am 15: 661-669, 1988 Takeichi M: Cadherins in cancer: implications for invasion and metastasis. Curr Opin Cell Biol 5:806-811, 1993 Thrasher JB, Crawford ED: Current management of invasive and metastatic transitional cell carcinoma of the bladder. J Urol 149: 957-992, 1993 Yoo J, Park S, Kang CS et al: Expression of E-cadherin and p53 proteins in human soft tissue sarcomas. Arch Pathol Lab Med 1261: 33-38, 2002 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 73 EP 77 PG 5 ER PT J AU Baccar Harrath, A Yacoubi Loueslati, B Troudi, W Hmida, S Sedkaoui, S Dridi, A Jridi, A Ben Ayed, F Ben Rhomdhane, K Ben Ammar Elgaaied, A AF Baccar Harrath, Amal Yacoubi Loueslati, Besma Troudi, Wafa Hmida, Slama Sedkaoui, Syrine Dridi, Amel Jridi, Afef Ben Ayed, Farhat Ben Rhomdhane, Khaled Ben Ammar Elgaaied, Amel TI HLA Class II Polymorphism: Protective or Risk Factors to Breast Cancer in Tunisia? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HLA; DRB1; DQB1; breast cancer ID HLA; DRB1; DQB1; breast cancer AB HLA system plays a key role in the tumor cells' escape from immune surveillance. Herein is the first report on the correlation of the susceptibility to breast cancer with HLA class II markers in Tunisia. Molecular typing of HLA-DRB1 and -DQB1 loci was undertaken for 70 Tunisian female patients. Comparison of allele and haplotype distribution between patients and 70 female control subjects reveals a negative association between HLADRB1* 07-DQB1*02 and the incidence of breast cancer in the Tunisian population. C1 [Baccar Harrath, Amal] University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 1060 Tunis, Tunisia. [Yacoubi Loueslati, Besma] University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 1060 Tunis, Tunisia. [Troudi, Wafa] University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 1060 Tunis, Tunisia. [Hmida, Slama] Tunisian Center of Blood TransfusionTunis, Tunisia. [Sedkaoui, Syrine] University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 1060 Tunis, Tunisia. [Dridi, Amel] Tunisian Center of Blood TransfusionTunis, Tunisia. [Jridi, Afef] Tunisian Center of Blood TransfusionTunis, Tunisia. [Ben Ayed, Farhat] Salah Azeiz Oncology InstituteTunis, Tunisia. [Ben Rhomdhane, Khaled] Salah Azeiz Oncology InstituteTunis, Tunisia. [Ben Ammar Elgaaied, Amel] University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 1060 Tunis, Tunisia. RP Baccar Harrath, A (reprint author), University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 1060 Tunis, Tunisia. EM amal.baccar@laposte.net CR Abrahamova J, Majsky A: HLA system and some neoplastic diseases. Acta Univ Carol Med Monogr 123: 1-80, 1988 Apple RJ, Erlich HA, Klitz W, et al: HLA DR-DQ associations with cervical carcinoma show papillomavirus-type specificity. Nat Genet 6:157-162, 1994 Bidwell JL, Soong TW, Raymond PA, et al: HLA genotyping of colorectal carcinoma in the Chinese population. Hum Immunol 34:19- 23, 1992 Biswal BM, Kumar R, Julka PK, et al: Human leucocytic antigens, HLA, in breast cancer. Int J Med Sci 52:177-183, 1998 Bodmer WF: The HLA system: structure and function. J Clin Pathol 40:948-958, 1987 Bouillence C, Deneufbourg JM: Positive correlation between breast cancer incidence and HLA antigens. Oncology 36:156-159, 1979 Buyse I, Dacorte R, Baens M, et al: Rapid DNA typing of class II antigens using the polymerase chain reaction and reverse dot blot hybridization. Tissue Antigens 41:1-14, 1993 Casoli C, Zanelli P, Adorni A, et al: Serological and molecular study on the HLA phenotype of female breast cancer patients. Eur J Cancer 30: 1207-1208, 1994 Campbell RD, Trowsdale J: Map of the human MHC. Immunol Today 14 349, 1993 Chaudhuri S, Cariappa A, Tang M, et al: Genetic susceptibility to breast cancer, HLA DQB*03032 and HLA DRB1*11 may represent protective alleles. Proc Natl Acad Sci USA 97: 11451-11454, 2000 De Jong MM, Nolte IM, De Vries EGE, et al: The HLA class III subregion is responsible for an increased breast cancer risk. Hum Mol Genet 12:2311-2319, 2003 Feinmesser M, Sulkes A, Morgenstern S, et al: HLA-DR and beta 2 microglobulin expression in medullary and atypical medullary carcinoma of the breast: histopathologically similar but biologically distinct entities. J Clin Pathol 53: 286-291, 2000 Ferguson TA, Green DR, Griffith TS: Cell death and immune privilege. Int Rev Immunol 21:153-172, 2002 Ghaderi A, Talei A, Gharesi-Fard B, et al: HLA-DRB1 alleles and the susceptibility of Iranian patients with breast cancer. Pathol Oncol Res 7: 39-41, 2001 Hammond MG, Appadoo B, Brain P: HLA and cancer in South African Indians. Tissue Antigens 14:296-302, 1979 Hmida S, Gauthier A, Dridi A, et al: HLA class II polymorphism in Tunisians. Tissue Antigens 45: 63-68, 1995 Igney FH, Krammer PH: Death and anti-death: tumor resistance to apoptosis. Nat Rev Cancer 2:277-288, 2002 Maniatis T, Fritsch EF, Sambrook J: Molecular Cloning. A Laboratory Manual. Cold Spring Laboratory Press, Cold Spring Harbor NY, 1982 Mestiri S, Bouaouina N, Ahmed SB, et al: Genetic variation in the tumor necrosis factor-alpha promoter region and in the stress protein hsp 70-2: susceptibility and prognostic implications in breast carcinoma. Cancer 91: 672-678, 2001 Park KS, Mok JW, Ko HE, et al: Polymorphisms of tumor necrosis factors A and B in breast cancer. Eur J Immunogenet 29: 7-10, 2002 Pellegris G, Illeni MT, Vaglini M, et al: HLA antigens in malignant melanoma patients. Tumori 66: 51-58, 1980 Ragoussis J, Monaco A, Mockridge I, et al: Cloning of the HLA class II region in yeast artificial chromosomes. Proc Natl Acad Sci USA 88: 3753-3757, 1991 Sastre-Garau X, Loste MN, Vincent-Salomon A, et al: Decreased frequency of HLA-DRB1 13 alleles in Frenchwomen with HPV-positive carcinoma of the cervix. Int J Cancer 69: 159-164, 1996 Subira ML, Crisci CD, Zornoza G, et al: Breast cancer and histocompatibility antigens. Allegrol Immunopathol, Madr, 7: 411-416, 1979 Wu MS, Hsieh RP, Huang SP, et al: Association of HLA-DQB1*0301 and HLA-DQB1*0602 with different subtypes of gastric cancer in Taiwan. Jpn J Cancer Res 93:404-410, 2002 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 79 EP 81 PG 3 ER PT J AU Sughayer, AM Al-Khawaja, MM Massarweh, S Al-Masri, M AF Sughayer, A Maher Al-Khawaja, M Maha Massarweh, Suleiman Al-Masri, Mahmoud TI Prevalence of Hormone Receptors and HER2/neu in Breast Cancer Cases in Jordan SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; hormone receptors; HER2/neu ID breast cancer; hormone receptors; HER2/neu AB The management and prognosis of breast cancer nowadays require the evaluation of Estrogen (ER), Progesterone Receptors (PR) and HER2/neu. Ethnic variation in the expression of these receptors is well documented. The aim of this study is to determine the prevalence of ER, PR and HER2/neu among Jordanian women with breast cancer of ductal and lobular types. A retrospective analysis was performed on 267 cases of breast cancer referred for treatment at King Hussein Cancer Center, Jordan between the period of June 2003 and June 2004. Standard immune stains were used for evaluation of hormone receptors and HER2/neu. In addition, evaluation of HER2/neu was done by FISH in selected cases. Of these 267 cases, 240 (89.9%) were ductal carcinomas of various histological grades, 122 (50.8%) of which were ER-positive, 138 (57.5%) PRpositive and 42 (17.5%) HER2/neu-positive. Twentytwo (8.2%) of all cases were lobular carcinomas, 15 (68%) of which were ER-positive, 20 (90.9%) PRpositive and 3 (13.6%) HER2/neu-positive. Five (1.9%) of the total cases were of mixed lobular and ductal types, 4 (80%) of which were ER-positive, 3 (60%) PR-positive and none were positive for HER2/neu. The prevalence of hormone receptor positivity in breast cancer of Jordanian women is lower than that of the western populations and close to other populations such as the Chinese and the minor ethnic groups of Northern America (African Americans). C1 [Sughayer, A Maher] King Hussein Cancer Center, Department of Pathology, Queen Rania AlAbdullah Str., 11941 Amman, Jordan. [Al-Khawaja, M Maha] King Hussein Cancer Center, Department of Pathology, Queen Rania AlAbdullah Str., 11941 Amman, Jordan. [Massarweh, Suleiman] Department of Medical OncologyAmman, Jordan. [Al-Masri, Mahmoud] Department of Surgical OncologyAmman, Jordan. RP Sughayer, AM (reprint author), King Hussein Cancer Center, Department of Pathology, 11941 Amman, Jordan. CR Lazennec G, Bresson D, Lucas A, et al: ER beta inhibits proliferation and invasion of breast cancer cells. Endocrinology 142: 4120-4130, 2001 Anderson E, Clarke RB, Howell A: Estrogen responsiveness and control of normal human breast proliferation. Review. J Mammary Gland Biol Neoplasia 3: 23-35, 1998 Osborne CK, Yochmowitz MG, Knight WA 3rd, McGuire WL: The value of estrogen and progesterone receptors in the treatment of breast cancer. Cancer 46: 2884-2888, 1980 Wittliff JL: Steroid-hormone receptors in breast cancer. Cancer 53, 3 Suppl): 630-643, 1984 Slamon DJ, Clark GM, Wong SG, et al: Human breast cancer: correlation of relapse and survival with amplification of the HER- 2/neu oncogene. Science 235: 177-182, 1987 Shak S: Overview of the trastuzumab, Herceptin, anti-HER2 monoclonal antibody clinical program in HER2-overexpressing metastatic breast cancer. Herceptin Multinational Investigator Study Group. Review. Semin Oncol 4 Suppl 12: 71-77, 1999 Cobleigh MA, Vogel CL, Tripathy D, et al: Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol 9: 2639-2648, 1999 Tavassoli FA, Devilee P, Eds.): World Health Organization Classification of Tumours. Pathology and Genetics of Tumours of the Breast and Female Genital Organs. IARC Press: Lyon, 2003 Pegoraro RJ, Karnan V, Nirmul D, Joubert SM: Estrogen and progesterone receptors in breast cancer among women of different racial groups. Cancer Res 46: 2117-2120, 1986 Ruder AM, Lubin F, Wax Y, et al: Estrogen and progesterone receptors in breast cancer patients. Epidemiologic characteristics and survival differences. Cancer 64: 196-202, 1989 Gapstur SM, Dupuis J, Gann P, et al: Hormone receptor status of breast tumors in black, Hispanic, and non-Hispanic white women. An analysis of 13,239 cases. Cancer 77: 1465-71, 1996 Joslyn SA: Hormone receptors in breast cancer: racial differences in distribution and survival. Breast Cancer Res Treat 73: 45-59, 2002 Li CI, Malone KE, Daling JR: Differences in breast cancer hormone receptor status and histology by race and ethnicity among women 50 years of age and older. Cancer Epidemiol Biomarkers Prev 7: 601-607, 2002 Chu KC, Anderson WF, Fritz A, et al: Frequency distributions of breast cancer characteristics classified by estrogen receptor and progesterone receptor status for eight racial/ethnic groups. Cancer 92: 37-45, 2001 Stierer M, Rosen H, Weber R, et al: Immunohistochemical and biochemical measurement of estrogen and progesterone receptors in primary breast cancer. Correlation of histopathology and prognostic factors. Ann Surg 218: 13-21, 1993 Chow LW, Ho P: Hormonal receptor determination of 1,052 Chinese breast cancers. J Surg Oncol 75: 172-175, 2000 Lertsanguansinchai P, Chottetanaprasith T, Chatamra K, et al: Estrogen and progesterone receptors status in Thai female breast cancer patients: an analysis of 399 cases at King Chulalongkorn Memorial Hospital. J Med Assoc Thai 85, Suppl 1: S193-202, 2002 Ikpatt OF, Ndoma-Egba R: Oestrogen and progesterone receptors in Nigerian breast cancer: relationship to tumour histopathology and survival of patients. Cent Afr J Med 49: 122-126, 2003 al-Alwan NA, al-Kubaisy W, al-Rawaq K: Assessment of response to tamoxifen among Iraqi patients with advanced breast cancer. East Mediterr Health J 6: 475-82, 2000 Abadjian G, Antoun R: Breast carcinoma: evaluation of hormone receptors and pS2, erb-B2, P-glycoprotein and Ki-67 markers. J Med Liban 44: 10-15. 1996 in French Samir S Amr, Abdul Rahman M. Sa'di, Fazal Ilahi, et al: Spectrum of Breast Diseases In Saudi Arab Females: A 26 Year Pathological Survey At Dhahran Health Center. Ann Saudi Med 15: 125-132, 1995 McCarthy NJ, Yang X, Linnoila IR, et al: Microvessel density, expression of estrogen receptor alpha, MIB-1, p53, and c-erbB-2 in inflammatory breast cancer. Clin Cancer Res 8: 3857-3862, 2002 Arpino G, Bardou VJ, Clark GM, Elledge RM: Infiltrating lobular carcinoma of the breast: tumor characteristics and clinical outcome. Breast Cancer Res 6: 149-156, 2004 Ross JS, Fletcher JA: HER-2/neu, c-erb-B2, gene and protein in breast cancer. Am J Clin Pathol 112(Suppl 1): S53-67, 1999 Ameyaw MM, Tayeb M, Thornton N, et al: Ethnic variation in the HER-2 codon 655 genetic polymorphism previously associated with breast cancer. J Hum Genet 47: 172-175, 2002 Sellami M, Gamoudi M, Krichen K, et al: Incidence of amplification of the C-erb B2/Her-2/neu gene in human breast cancer, in French). Arch Inst Pasteur Tunis 68: 33-41, 1991 el-A Helal T, Khalifa A, Kamel AS: Immunohistochemical expression of p53 and c-erbB2 proteins in breast cancer in Egypt. Anticancer Res 20: 2145-2150, 2000 Middleton LP, Palacios DM, Bryant BR, Krebs P, Otis CN, Merino MJ: Pleomorphic lobular carcinoma: morphology, immunohistochemistry, and molecular analysis. Am J Surg Pathol 24: 1650-1656, 2000 Feuer EJ, Wun LM: DEVCAN: Probability of Developing or Dying of Cancer. Version 4.0. Bethesda MD: National Cancer Institute. 1999. Al-Kayed S, Al-Hijawi B, Al-Haliq T: Incidence of cancer in Jordan 2001. National Cancer Registry, Ministry of Health/Jordan, 2001 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 83 EP 86 PG 4 ER PT J AU Hiraishi, Y Wada, T Nakatani, K Negoro, K Fujita, Sh AF Hiraishi, Yukihiro Wada, Takeshi Nakatani, Ken Negoro, Kenji Fujita, Shigeyuki TI Immunohistochemical Expression of EGFR and p-EGFR in Oral Squamous Cell Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR; p-EGFR; oral squamous cell carcinomas ID EGFR; p-EGFR; oral squamous cell carcinomas AB Epidermal growth factor receptor (EGFR) is a tyrosine kinase receptor of the ErbB family, which is expressed or highly expressed in a variety of solid tumors, including oral cancers. High EGFR expression has been correlated with tumor size, metastasis and survival. In recent years, EGFR has been considered a promising target for monoclonal antibody therapy. A total of 52 patients with oral squamous cell carcinoma (OSCC) were selected for EGFR and phosphorylated EGFR (p-EGFR) detection. Immunohistochemical staining was performed to evaluate EGFR and p-EGFR expression. Positive EGFR and p-EGFR staining was present in 92.3% (48/52) and 98.0% (51/52) of all cases, respectively. High EGFR and p-EGFR expression was present in 63.4% (33/52) and 69.2% (36/52) of all cases, respectively. EGFR and p-EGFR expression did not correlate with the clinical factors tumor stage, regional lymph node metastasis, or distant metastasis. However, a statistically significant correlation was identified between high EGFR expression and the pathologic factor tumor invasion. As a conclusion, the majority of OSCCs highly express EGFR and p-EGFR, indicating the importance of studying the efficacy of anticancer therapy targeting these signal factors. C1 [Hiraishi, Yukihiro] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 kimiidera, 641-8509 Wakayama, Japan. [Wada, Takeshi] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 kimiidera, 641-8509 Wakayama, Japan. [Nakatani, Ken] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 kimiidera, 641-8509 Wakayama, Japan. [Negoro, Kenji] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 kimiidera, 641-8509 Wakayama, Japan. [Fujita, Shigeyuki] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 kimiidera, 641-8509 Wakayama, Japan. RP Hiraishi, Y (reprint author), Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 641-8509 Wakayama, Japan. EM hiraishi@wakayama-med.ac.jp CR Salomon DS, Brandt R, Ciardiello F, Normanno N: Epidermal growth factor- related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol 19: 183-232, 1995 Mendelsohn J, Baselga L: Status of epidermal growth factor receptor antagonists in the biology and treatment of cancer. J Clin Oncol 14: 2787- 2799, 2003 Irish JC, Bernstein A: Oncogenes in head and neck cancer. Laryngoscope 103: 42-52, 1993 Yamamoto E, Kohama G, Iwai M, Hiratsuka H: Mode of invasion, Bleomycin sensitivity and clinical course in squamous cell carcinoma of the oral cavity. Cancer 51: 2175-2180, 1983 Wada T: Nature of mononuclear cell infiltrates in oral squamous cell carcinoma and their clinical significance. Wakayama Med Rep 30: 103-117, 1989 Putti TC, To KF, Hsu HC, et al: Expression of epidermal growth factor receptor in head and neck cancers correlates with clinical progression: a multicentre immunohistochemical study in the Asia-Pacific region. Histopathology 41: 144-151, 2002 van Oijen MG, Rijksen G, ten Broek FW, Slootweg PJ: Increased expression of epidermal growth factor receptor in normal epithelium adjacent to head and neck carcinomas independent of tobacco and alcohol abuse. Oral Dis 4: 4-8, 1998 Wen QH, Nishimura T, Nagayama I, Furukawa M: Expression of EGF, EGFR and PCNA in laryngeal lesions. J Laryngol Otol 109: 630-636, 1995 Lee CS, Redshaw A, Boag G: Epidermal growth factor immunoreactivity in human laryngeal squamous cell carcinoma. Pathology 29: 251-254, 1997 Kearsley JH, Leonard JH, Walsh MD, Wright GR: A comparison of epidermal growth factor receptor, EGFR, and c-erbB-2 oncogene expression in head and neck squamous cell carcinomas. Pathology 23: 189-194, 1991 Zheng X, Hu L, Chen F, Christensson B: Expression of Ki67 antigen, epidermal growth factor receptor and Epstein-Barr virus-encoded latent membrane protein, LMP1, in nasopharyngeal carcinoma. Eur J Cancer B Oral Oncol 30: 290-295, 1994 Miyaguchi M, Olofsson J, Hellquist HB: Expression of epidermal growth factor receptor in glottic carcinoma and its relation to recurrence after radiotherapy. Clin Otolaryngol 16: 466-469, 1991 Downward J, Parker P, Waterfield MD: Autophosphorylation sites on the epidermal growth factor receptor. Nature 311: 483-485, 1984 Kanematsu T, Yano S, Uehara H, et al: Phosphorylation, but not overexpression, of epidermal growth factor receptor is associated with poor prognosis of non-small cell lung cancer patients. Oncol Res 13: 289-298, 2003 Olayioye MA, Neve RM, Lane HA, Hynes NE: The ErbB signaling network: Receptor heterodimerization in development and cancer. EMBO J 19: 3159-3167, 2000 Kolch W: Meaningful relationships: The regulation of the Ras/Raf/MEK/ ERK pathway by protein interactions. Biochem J 351: 289-305, 2000 Zwick E, Bange J, Ullrich A: Receptor tyrosine kinases as targets for anticancer drugs. Trends Mol Med 8: 17-23, 2002 Busse D, Doughty RS, Ramsey TT, et al: Reversible G1 arrest induced by inhibition of the epidermal growth factor receptor tyrosine kinase requires up-regulation of p27KIP1 independent of MAPK activity. J Biol Chem 275: 6987-6995, 2000 Khazaie K, Schirrmacher V, Lichtner RB: EGF receptor in neoplasia and metastasis. Cancer Metastasis Rev 12: 255-274, 1993 Storkel S, Reichert T, Reiffen KA, Wagner W: EGFR and PCNA expression in oral squamous cell carcinomas–a valuable tool in estimating the patient’s prognosis. Eur J Cancer B Oral Oncol 29: 273-277, 1993 Xia W, Lau YK, Zhang HZ, et al: Combination of EGFR, HER-2/neu, and HER-3 is a stronger predictor for the outcome of oral squamous cell carcinoma than any individual family members. Clin Cancer Res 5: 4164- 4174, 1999 Christensen ME: The EGF receptor system in head and neck carcinomas and normal tissues. Immunohistochemical and quantitative studies. Dan Med Bull 45:121-134, 1998 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 87 EP 91 PG 5 ER PT J AU Kahan, Zs Varga, K Dudas, R Nyari, T Thurzo, L AF Kahan, Zsuzsanna Varga, Katalin Dudas, Rita Nyari, Tibor Thurzo, Laszlo TI Collaborative/Active Participation per se Does Not Decrease Anxiety in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE anxiety; early breast cancer; information needs; metastatic breast cancer; treatment decision-making ID anxiety; early breast cancer; information needs; metastatic breast cancer; treatment decision-making AB The information needs of breast cancer patients on their disease, its treatment, the prognosis, and their attitude to decision-making concerning treatment were assessed. One hundred and fifty early and 45 metastatic breast cancer patients were recruited into the study. The amount of information and role in the treatment decision-making process preferred by the patient were independently estimated by the patient and the oncologist, using questionnaires. Information was provided in accordance with the wishes of the patient as perceived by the physician. Test of anxiety was performed before, and one week after the consultation. Most of the patients claimed to anticipate the provision of extensive information and an active role in the decision-making, but real interest during the consultation was found less frequently. The post-consultation anxiety test revealed a significant decrease in situational anxiety; this was not related to the patient’s information needs or her attitude to the decision-making concerning treatment. Our study demonstrates that a significant decrease in anxiety may be achieved via a consultation tailored to the needs of the patient. Loading the patient with information and involvement in the decision regarding therapy as much as the patient seems comfortable with lowers distress. C1 [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, Koranyi fasor 12., H-6720 Szeged, Hungary. [Varga, Katalin] University of Szeged, Department of Oncotherapy, Koranyi fasor 12., H-6720 Szeged, Hungary. [Dudas, Rita] University of Szeged, Department of Oncotherapy, Koranyi fasor 12., H-6720 Szeged, Hungary. [Nyari, Tibor] University of Szeged, Department of Medical InformaticsSzeged, Hungary. [Thurzo, Laszlo] University of Szeged, Department of Oncotherapy, Koranyi fasor 12., H-6720 Szeged, Hungary. RP Kahan, Zs (reprint author), University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. EM kahan@onko.szote.u-szeged.hu CR Holland JC: IPOS Sutherland Memorial Lecture: an international perspective on the development of psychosocial oncology: overcoming cultural and attitudinal barriers to improve psychosocial care. Psychooncology 13: 445-459, 2004 Ruckdeschel JC, Blanchard CG, Albrecht T: Psychosocial oncology research. Where we have been, where we are going, and why we will not get there. Cancer 74(suppl 4): S1458- S1463, 1994 The Steering Committee on Clinical Practice Guidelines for the Care and Treatment of Breast Cancer. A Canadian consensus document. CMAJ 158(suppl 3): S3-S64, 1998 Coates A: Management of early breast cancer: An Australian consensus report. Oncology 52: 82-85, 1995 Fallowfield LJ, Hall A, Maguire GP, et al: Psychological outcomes of different treatment policies in women with early breast cancer outside a clinical trial. BMJ 301: 575-580, 1990 Brown RF, Butow PN, Dunn SM, et al: Promoting patient participation and shortening cancer consultations: a randomised trial. Br J Cancer 85: 1273-1279, 2001 Siminoff LA, Ravdin P, Colabianchi N, et al: Doctor-patient communication patterns in breast cancer adjuvant therapy discussions. Health Expect 3: 26-36, 2000 Wallberg B, Michelson H, Nystedt M, et al.: Information needs and preferences for participation in treatment decisions among Swedish breast cancer patients. Acta Oncol 39: 467-476, 2000 Hagerty RG, Butow PN, Ellis PA, et al: Cancer patient preferences for communication of prognosis in the metastatic setting. J Clin Oncol 22: 1721-1730, 2004 Bruera E, Sweeney C, Calder K, et al: Patient preferences versus physician perceptions of treatment decisions in cancer care. J Clin Oncol 19: 2883-2885, 2001 Blanchard CG, Labrecque MS, Ruckdeschel JC, et al: Physician behaviors, patient perceptions, and patient characteristics as predictors of satisfaction of hospitalized adult cancer patients. Cancer 65: 186-192, 1990 Jansen SJ, Otten W, van de Velde CJ, et al: The impact of the perception of treatment choice on satisfaction with treatment, experienced chemotherapy burden and current quality of life. Br J Cancer 91: 56-61, 2004 Mesters I, van den Borne B, De Boer M, et al: Measuring information needs among cancer patients. Patient Educ Cons 43: 253-262, 2001 Spielberger CD: Manual for the state trait anxiety inventory, Form Y). Consulting Psychologists Press, Palo Alto, CA, 1983 Degner LF, Sloan JA: Decision making during serious illness: what role do patients really want to play? J Clin Epidemiol 45: 941-950, 1992 Ravdin PM, Siminoff LA, Davis GJ, et al: Computer program to assist in making decisions about adjuvant therapy for women with early breast cancer. J Clin Oncol 19: 980-991, 2001 Whelan T, Sawka C, Levine M, et al: Helping patients make informed choices: a randomized trial of a decision aid for adjuvant chemotherapy in lymph node-negative breast cancer. J Natl Cancer Inst 95: 581-587, 2003 Whelan T, Levine M, Willan A, et al: Effect of a decision aid on knowledge and treatment decision making for breast cancer surgery: a randomized trial. JAMA 292: 435-441, 2004 Beaver K, Bogg J, Luker KA: Decision-making role preferences and information needs: a comparison of colorectal and breast cancer. Health Expect 2: 266-276, 1999 Beaver K, Luker KA, Owens RG, et al: Treatment decision making in women newly diagnosed with breast cancer. Cancer Nurs 19:8-19, 1996 Butow PN, Dunn SM, Tattersall MHN, et al: Patient participation in the cancer consultation: evaluation of a question prompt sheet. Ann Oncol 5: 199-204, 1994 Butow PN, Kazemi JN, Beeney LJ, et al.: When diagnosis is cancer. Patient communication experiences and preferences. Cancer 77: 2630-2637, 1996 Moumjid N, Carrere MO, Charavel M, et al: Clinical issues in shared decision-making applied to breast cancer. Health Expect 6: 222-227, 2003 Lam W, Fielding R, Chan M, et al: Participation and satisfaction with surgical treatment decision-making in breast cancer among Chinese women. Breast Cancer Res Treat 80: 171-180, 2003 Cassileth BR, Zupkis RV, Sutton-Smith K, et al: Information and participation preferences among cancer patients. Ann Int Med 92:832-836, 1980 Bilodeau BA, Degner LF: Information needs, sources of information, and decisional roles in women with breast cancer. Oncol Nurs Forum 23:691-696, 1996 Johnson JD, Roberts CS, Cox CE, et al: Breast cancer patients’ personality style, age, and treatment decision making. J Surg Oncol 63:183-186, 1996 Maly RC, Umezawa Y, Leake B, et al: Determinants of participation in treatment decision-making by older breast cancer patients. Breast Cancer Res Treat 85: 201-209, 2004 Sepucha KR, Belkora JK, Tripathy D, et al: Building bridges between physicians and patients: results of a pilot study examining new tools for collaborative decision making in breast cancer. J Clin Oncol 18: 1230-1238, 2000 Liang W, Burnett CB, Rowland JH, et al: Communication between physicians and older women with localized breast cancer: implication for treatment and patient satisfaction. J Clin Oncol 20: 1008-1016, 2002 Bruera E, Willey JS, Palmer JL, et al: Treatment decisions for breast carcinoma: patient preferences and physician perceptions. Cancer 94: 2076-2080, 2002 Leighl N, Gattellari M, Butow P, et al: Discussing adjuvant cancer therapy. J Clin Oncol 19: 1768-1778, 2001 Butow PN, Dowsett S, Hagerty R, et al: Communicating prognosis to patients with metastatic disease: what do they really want to know? Support Care Cancer 10: 161-168, 2002 Janz NK, Wren PA, Copeland LA, et al: Patient-physician concordance: preferences, perceptions, and factors influencing the breast cancer surgical decision. J Clin Oncol 22: 3091-3098, 2004 Protiere C, Viens P, Genre D, et al: Patient participation in medical decision-making: a French study in adjuvant radiochemotherapy for early breast cancer. Ann Oncol 11: 39-45, 2000 Molenaar S, Sprangers MA, Rutgers EJ, et al: Decision support for patients with early-stage breast cancer: effects of an interactive breast cancer CDROM on treatment decision, satisfaction, and quality of life. J Clin Oncol 19: 1676-1687, 2001 EUSOMA Position Paper. The requirements of a special breast unit. Eur J Cancer 36: 2288-2293, 2000 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 93 EP 101 PG 9 ER PT J AU Borka, K Patai, K Rendek, A Sobel, G Paulin, F AF Borka, Katalin Patai, Kalman Rendek, Aniko Sobel, Gabor Paulin, Ferenc TI Pleomorphic Rhabdomyosarcoma of the Uterus in a Postmenopausal Patient SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE pleomorphic rhabdomyosarcoma; uterus; adult; immunohistochemistry; postmenopausal bleeding ID pleomorphic rhabdomyosarcoma; uterus; adult; immunohistochemistry; postmenopausal bleeding AB Pure rhabdomyosarcomas occurring in the adult uterus are very rare, with poor prognosis. We present a case of a 67-year-old woman with postmenopausal vaginal bleeding caused by pleomorphic rhabdomyosarcoma of the uterus, treated with hysterectomy, bilateral salpingo-oophorectomy, pelvic/paraaortic lymphadenectomy and partial sigmoidectomy. Postoperative chemotherapy (Doxorubicin) was given according to protocol. Follow-up examinations one year after surgery revealed no abnormalities or tumor recurrence. The rarity of this histological entity makes the presented case worthy of publication. C1 [Borka, Katalin] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Patai, Kalman] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Rendek, Aniko] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Sobel, Gabor] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Paulin, Ferenc] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. RP Borka, K (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM bk@korb2.sote.hu CR Holcomb K, Francis M, Ruiz J, et al: Pleomorphic rhabdomyosarcoma of the uterus in a postmenopausal woman with elevated serum CA-125. Gynecol Oncol 74: 499-501, 1999 Chiarle R, Godio L, Fusi D, et al: Pure alveolar rhabdomyosarcoma of the corpus uteri: description of a case with increased serum level of CA-125. Gynecol Oncol 66: 320-323, 1997 Sternberg SS, ed.): Diagnostic Surgical Pathology. Raven Press, New York, 1989 Rosai J, ed.): Rosai and Ackerman’s Surgical Pathology. Mosby, Edinburgh, 2004 Kulka EW, Douglas GW: Rhabdomyosarcoma of the corpus uteri: report of a case, associated with adenocarcinoma of the cervix, with review of the literature. Cancer 5: 727-736, 1952 McCluggage WG, Lioe TF, Mcclelland HR, et al: Rhabdomyosarcoma of the uterus: report of two cases, including one of the spindle cell variant. Gynecol Oncol 12: 128-132, 2002 Podczaski E, Sees J, Kaminski P, et al: Rhabdomyosarcoma of the uterus in a postmenopausal patient. Gynecol Oncol 37: 439- 442, 1990 Ng TY, Loo KT, Leung TW, et al: Alveolar rhabdomyosarcoma of the cervix. Gynecol Oncol 91: 623-626, 2003 Hart WR, Craig JR: Rhabdomyosarcomas of the uterus. Am J Clin Pathol 70: 217-223, 1987 Cavazzana AO, Schmidt D, Ninfo V, et al: Spindle cell rhabdomyosarcoma: a prognostically favorable variant of rhabdomyosarcoma. Am J Surg Pathol 16: 229-235, 1992 Montag TW, D’ablaing G, Schlaerth JB, et al: Embryonal rhabdomyosarcoma of the uterine corpus and cervix. Gynecol Oncol 25: 171-194, 1986 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 102 EP 104 PG 3 ER PT J AU Dundar, E Acikalin, FM Can, C AF Dundar, Emine Acikalin, Fuat Mustafa Can, Cavit TI The Nested Variant of Urothelial Carcinoma: an Aggressive Tumor Closely Simulating Benign Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE urothelial carcinoma; nested variant ID urothelial carcinoma; nested variant AB The ''nested'' variant is a rare form of urothelial carcinoma and its biologic behavior is highly aggressive. Herein two new cases of nested variant of urothelial carcinoma with immunohistochemical examination are presented. In one of the cases, the tumor extended through the bladder wall into the perivesicular soft tissue, prostatic urethra and left vesicula seminalis, and metastasized to obturator lymph nodes. In the other case, invasion of muscular layer was observed and three recurrences were developed during a follow-up period of 23 months. Both tumors of our study demonstrated high p53 and Ki-67 indices, supporting the aggressive nature of such tumors. C1 [Dundar, Emine] Eskisehir Osmangazi University, Faculty of Medicine, Department of Pathology, Fakultesi Patoloji Anabilim DaliEskisehir, Turkey. [Acikalin, Fuat Mustafa] Eskisehir Osmangazi University, Faculty of Medicine, Department of Pathology, Fakultesi Patoloji Anabilim DaliEskisehir, Turkey. [Can, Cavit] Eskisehir Osmangazi University, Faculty of Medicine, Department of UrologyEskisehir, Turkey. RP Dundar, E (reprint author), Eskisehir Osmangazi University, Faculty of Medicine, Department of Pathology, Eskisehir, Turkey. EM edundar_99@yahoo.com CR Talbert ML, Young RH: Carcinomas of the urinary bladder with deceptively benign-appearing foci. A report of three cases. Am J Surg Pathol 13: 374-381, 1989 Lin O, Cardillo M, Dalbagni G, et al: Nested variant of urothelial carcinoma: A clinicopathologic and immunohistochemical study of 12 cases. Mod Pathol 16: 1289-1298, 2003 Murphy WM, Deana DG: The nested variant of transitional cell carcinoma: A neoplasm resembling proliferation of Brunn’s nests. Mod Pathol 5: 240-243, 1992 Xiao GQ, Savage SJ, Gribetz ME, et al: The nested variant of urothelial carcinoma. Clinicopathology of 2 cases. Arch Pathol Lab Med 127: 333-336, 2003 Holmang S, Johansson SL: The nested variant of transitional cell carcinoma. A rare neoplasm with poor prognosis. Scand J Urol Nephrol 35:102-105, 2001 Volmar KE, Chan TY, De Marzo AM, Epstein JI: Florid von Brunn nests mimicking urothelial carcinoma. A morphologic and immunohistochemical comparison to the nested variant of urothelial carcinoma. Am J Surg Pathol 27: 1243-1252, 2003 Drew PA, Furman J, Civantos F, Murphy WM: The nested variant of transitional carcinoma: an aggressive neoplasm with innocuous histology. Mod Pathol 9: 989-994, 1996 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 105 EP 107 PG 3 ER PT J AU Glisic, A Atanackovic, J AF Glisic, Andreja Atanackovic, Jasmina TI Krukenberg tumor in pregnancy. The lethal outcome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Krukenberg tumor; pregnancy; lethal outcome ID Krukenberg tumor; pregnancy; lethal outcome AB Krukenberg tumor refers to gastrointestinal cancer metastatic to the ovaries and its prognosis is uniformly poor. This case report concerns a 38-year-old pregnant woman suffering from abdominal pain and iterative vomiting episodes. She presented with a large abdominopelvic tumor. Because of suspected ovarian torsion, we performed urgent surgery. At laparotomy, bilateral ovarian tumors, ascites and gastric cancer located at the cardia and the lesser curvature invading the serosa were identified. We performed right ovariectomy, resection of the left ovary, and gastric biopsy. Histological examination of the specimen yielded diagnosis of Krukenberg tumor. Ten days later the patient underwent an elective Cesarean section in the 25th gestational week because of fetal asphyxia and very poor maternal life prognosis. We performed Cesarean delivery and extracted a vital female newborn of 31 cm, 600 g, Ap score 3, with virilization. Few days later the baby died at the intensive care unit. Two weeks later the mother died because of pulmonary failure. C1 [Glisic, Andreja] Clinical Center of Serbia, Department of Obstetrics and Gynecology, Visegradska 26, 11000 Belgrade, Serbia. [Atanackovic, Jasmina] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia. RP Glisic, A (reprint author), Clinical Center of Serbia, Department of Obstetrics and Gynecology, 11000 Belgrade, Serbia. EM gan@eunet.yu CR McGill F, Ritter DB, Rickard C, et al: Management of Krukenberg tumors: an 11-year experience and review of the literature. Prim. Care Update Ob Gyns 5:157-158, 1998 McGill F, Ritter DB, Rickard C, et al: Krukenberg tumors: can management be improved? Gynecol Obstet Invest 48: 61-65, 1999 Agarwal N, Parul, Kriplani A, et al: Management and outcome of pregnancies complicated with adnexal masses. Arch Gynecol Obstet 267: 148-152, 2003 Chou MM, Ho ES, Lin NF, Lee YH: Color Doppler sonographic appearance of a Krukenberg tumor in pregnancy. Ultrasound Obstet Gynecol 11: 459-460, 1998 Mackey JR, Hugh J, Smylie M: Krukenberg tumor complicated by pregnancy. Gynecol Oncol 61: 153-155, 1996 De Palma P, Wronski M, Bifermino V, Bovani I: Krukenberg tumor in pregnancy with virilization. A case report. Eur J Gynaecol Oncol 16: 59-64, 1995 Tamussino K, Scholl W, Reich O, Winter R: Gastric carcinoma presenting as a Krukenberg tumor in the 24th week of gestation. Eur J Obstet Gynecol Reprod Biol 62: 251-252, 1995 Cheng CY, Chen TY, Lin CK, et al: Krukenberg tumor in pregnancy with delivery of a normal baby: a case report. Zhonghua Yi Xue Za Zhi, Taipei, 54: 424-427, 1994 Scharl A, Huber P, Lorenyen J, Gohring UJ: Gastric cancer during early pregnancy. Two case reports. Arch Gynecol Obstet 258: 151-154, 1996 Sandmaier D, Lobrinus JA, Vial Y, et al: Bilateral Krukenberg tumor of the ovary during pregnancy. Eur J Gynecol Oncol 21: 58-60, 2000 Cosme A, Ojeda E, Bujanda L, et al: Krukenberg tumor secondary to gastric carcinoma in a woman in her eighth month of pregnancy. Gastroenterol Hepatol 24: 63-65, 2001 Okutomi T, Hoshino Y, Amano K, et al: Intrathecal fentanyl/ meperidine combined with low-dose epidural bupivacaine for Cesarean section in a patient with advanced Krukenberg tumors. Acta Anaesthesiol Scand 46: 1272-1275, 2002 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 108 EP 110 PG 3 ER PT J AU Vargas-Gonzalez, R Sanchez-Sosa, S AF Vargas-Gonzalez, Roberto Sanchez-Sosa, Sergio TI Fibrocartilaginous Dysplasia (Fibrous Dysplasia with Extensive Cartilaginous Differentiation) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Fibrocartilaginous dysplasia; fibrous dysplasia; fibrochondrodysplasia; fibrous dysplasia with cartilaginous differentiation ID Fibrocartilaginous dysplasia; fibrous dysplasia; fibrochondrodysplasia; fibrous dysplasia with cartilaginous differentiation AB Fibrocartilaginous dysplasia is a variant of fibrous dysplasia in which extensive cartilaginous differentiation is identified. The amount of cartilage varies from case to case, however, no percentage has been proposed to consider this diagnosis. We present a 6-year-old girl with a two-year history of hip pain. Initial imaging studies of the right femur revealed a lucent lesion of the proximal shaft that extended into the femoral neck with ill-defined borders but wellmaintained cortex. Computed tomography scan demonstrated increased density of the medullary cavity but the cortex appeared intact. Curettage of the lesion was performed and fragments with cartilaginous appearance were obtained, weighing 45 g in total. Microscopically, the tumor revealed a cartilaginous (60%) and a fibro-osseous (40%) component; the former had increased cellularity and some chondrocytes displayed moderate atypia and binucleation, while the latter showed features of fibrous dysplasia. Areas of endochondral ossification and calcification were also identified. After five years of surgery this child is well and without evidence of recurrence. We discuss the differential diagnosis of this variant of fibrous dysplasia in the pediatric group. C1 [Vargas-Gonzalez, Roberto] Hospital Para el Nino Poblano, Department of Pathology, Km 1.5 Carretera Federal Puebla - Atlixco, 72190 Puebla, Mexico. [Sanchez-Sosa, Sergio] Hospital Para el Nino Poblano, Department of Pathology, Km 1.5 Carretera Federal Puebla - Atlixco, 72190 Puebla, Mexico. RP Vargas-Gonzalez, R (reprint author), Hospital Para el Nino Poblano, Department of Pathology, 72190 Puebla, Mexico. EM soncoy@msn.com CR Alman BA, Greel DA, Wolfe HJ: Activating mutations of Gs protein in monostotic fibrous lesions of bone. J Orthop Res 14: 311-315, 1996 Bianco P, Riminucci M, Majolagbe A, et al: Mutations of the GNAS1 gene, stromal cell dysfunction, and osteomalacic changes in non-McCune-Albright fibrous dysplasia of bone. J Bone Miner Res 15:120-128, 2000 Lichtenstein L, Jaffe HL: Fibrous dysplasia of bone. Arch Pathol 33: 777-816, 1942 Dorfman HD, Czeniak B: Bone Tumors. Mosby, St. Louis, 1998, pp. 441-481 Forest M, Tomeno B, Vanel D: Orthopedic surgical pathology: diagnosis of tumors and pseudotumoral lesions of bone and joints. Churchill Livingstone, Edinbourgh, 1998, pp. 595-612 Kransdorf MJ, Moser RP, Gilkey FW: Fibrous dysplasia. Radiographics 10: 519-537, 1990 Harris WH, Dudley HR Jr, Barry RJ: The natural history of fibrous dysplasia. J Bone Joint Surg Am 44:207-233, 1962 Sanerkin NG, Watt I: Enchondromata with annular calcification in association with fibrous dysplasia. B J Radiol 54: 1027- 1033, 1981 Pelzmann KS, Nagel DZ, Salyer WR: Case report 114. Skeletal Radiol 5:116-118, 1980 Drolshagen LF, Reynolds WA, Marcus NW: Fibrocartilaginous dysplasia of bone. Radiology 156: 32, 1985 Hermann G, Klein M, Abdelwahab IF, Kenan S: Fibrocartilaginous dysplasia. Skeletal Radiol 25: 509-511, 1996 Ishida T, Dorfman HD: Massive chondroid differentiation in fibrous dysplasia of bone, fibrocartilaginous dysplasia). Am J Surg Pathol 17: 924-930, 1993 De Smet AA, Travers H, Neff JR: Chondrosarcoma occurring in a patient with polyostotic fibrous dysplasia. Skeletal Radiol 7: 197-201, 1981 Teldorf ED: A case of osteitis fibrosa, with formation of hyaline cartilage). Br J Surg 18:409-414, 1930 Kyriakos M, Mc Donald DJ, Sundaram M: Fibrous dysplasia with extensive cartilaginous differentiation, “fibrocartilaginous dysplasia”): a review, with an illustrative case followed for 18 years. Skeletal Radiol 33: 51-62, 2004 Bulychova IV, Unni KK, Bertoni F, Beabout JW: Fibrocartilaginous mesenchymoma of bone. Am J Surg Pathol 17: 830-836, 1993 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 111 EP 114 PG 4 ER PT J AU Merkli, H Pal, E Gati, I Czopf, J AF Merkli, Hajnalka Pal, Endre Gati, Istvan Czopf, Jozsef TI Distal Myopathy with Rimmed Vacuoles and Cerebellar Atrophy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE ataxia; cerebellar atrophy; distal myopathy; rimmed vacuole ID ataxia; cerebellar atrophy; distal myopathy; rimmed vacuole AB Distal myopathies constitute a clinically and pathologically heterogeneous group of genetically determined neuromuscular disorders, where the distal muscles of the upper or lower limbs are affected. The disease of a 41-year-old male patient started with gait disturbances, when he was 25. The progression was slow, but after 16 years he became seriously disabled. Neurological examination showed moderate to severe weakness in distal muscles of all extremities, marked cerebellar sign and steppage gait. Muscle biopsy resulted in myopathic changes with rimmed vacuoles. Brain MRI scan showed cerebellar atrophy. This case demonstrates a rare association of distal myopathy and cerebellar atrophy. C1 [Merkli, Hajnalka] University of Pecs, Department of Neurology, Ret u.2.,, H-7623 Pecs, Hungary. [Pal, Endre] University of Pecs, Department of Neurology, Ret u.2.,, H-7623 Pecs, Hungary. [Gati, Istvan] University of Pecs, Department of Neurology, Ret u.2.,, H-7623 Pecs, Hungary. [Czopf, Jozsef] University of Pecs, Department of Neurology, Ret u.2.,, H-7623 Pecs, Hungary. RP Pal, E (reprint author), University of Pecs, Department of Neurology, H-7623 Pecs, Hungary. EM endre.pal@aok.pte.hu CR Ikeuchi T, Asaka T, Saito M, et al: Gene locus for autosomal recessive distal myopathy with rimmed vacuoles maps to chromosome 9. Ann Neurol 41: 432-437, 1997. Kumamoto T, Fukuhara N, Nagashima M, et al: Distal myopathy. Histochemical and ultrastructural studies. Arch Neurol 39: 367-371, 1982. Kumamoto T, Ito T, Horinouchi H, et al: Increased lysosomerelated proteins in the skeletal muscles of distal myopathy with rimmed vacuoles. Muscle Nerve 23: 1686-1693, 2000. Mastaglia FL, Laing NG: Distal myopathies: clinical and molecular diagnosis and classification. J Neurol Neurosurg Psychiatry 67: 703-709, 1999. Mizusawa H, Kurisaki H, Takatsu M, et al: Rimmed vacuolar distal myopathy: a clinical, electrophysiological, histopathological and tomographic study of seven cases. J Neurol 234: 129-136, 1987. Mizusawa H, Kurisaki H, Takatsu M, et al: Rimmed vacuolar distal myopathy. An ultrastructural study. J Neurol 234: 137- 145, 1987. Nishino I, Noguchi S, Murayama K, et al: Distal myopathy with rimmed vacuoles is allelic to hereditary inclusion body myopathy. Neurology 59: 1689-1693, 2002. Nonaka I, Murakami N, Suzuki Y, et al: Distal myopathy with rimmed vacuoles. Neuromusc Disord 8: 333-337, 1998. Sunohara N, Nonaka I, Kamei N, Satoyoshi E: Distal myopathy with rimmed vacuole formation. Brain 112: 65-83, 1989. Werneck LC, Marrone CD, Scola RH: Distal myopathies: clinical, laboratory, electromyographic, histologic-histochemical analysis of 8 cases. Arch Neuropyschiatr 51: 475-486, 1993. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 115 EP 117 PG 3 ER PT J AU Lumniczky, K Safrany, G AF Lumniczky, Katalin Safrany, Geza TI Cancer Gene Therapy: Combination with Radiation Therapy and the Role of Bystander Cell Killing in the Anti-tumor Effect SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Lecture DE gene therapy; bystander effect; gap junction ID gene therapy; bystander effect; gap junction AB Current anti-cancer modalities such as surgery, chemo- and radiation therapies have only limited success in cancer treatment. Gene therapy is a promising new tool to improve outcomes. In this review, first we summarize the various strategies to kill tumor cells, and then focus on the bystander effect of gene therapy. A variety of strategies, such as gene-directed enzyme pro-drug therapy, activation of an anti-tumor immune attack, application of replication-competent and oncolytic viral vectors, tumor-specific as well as radiation- and hypoxiainduced gene expression, might be applied to target tumor cells. We put special emphasis on the combination of these approaches with local tumor irradiation. Using the available vector systems, only a small portion of cancer cells contains the therapeutic genes under clinical situations. However, cells directly targeted by gene therapy will transfer death signals to neighboring cancer cells. This bystander cell killing improves the efficiency of cancer gene therapy. Death signals are delivered by cell-to-cell communication through gap junction intercellular contacts, release of toxic metabolites into the neighborhood or to larger distances, phagocytosis of apoptotic bodies, and the activation of the immune system. Bystander cell killing can be enhanced by the introduction of gap junction proteins into cells, by further activating the immune system with immune-stimulatory molecules, or by introducing genes that help the transfer of cytotoxic genes and/or metabolites into bystander cells. In conclusion, although bystander cell killing can improve therapeutic effects, there should be additional developments in cancer gene therapy for a more efficient clinical application. C1 [Lumniczky, Katalin] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Frederic Joliot-Curie Sugarbiologiai es Sugaregeszsegugyi Kutato Intezet, Anna u. 5., H-1221 Budapest, Hungary. [Safrany, Geza] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Frederic Joliot-Curie Sugarbiologiai es Sugaregeszsegugyi Kutato Intezet, Anna u. 5., H-1221 Budapest, Hungary. RP Safrany, G (reprint author), Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Frederic Joliot-Curie Sugarbiologiai es Sugaregeszsegugyi Kutato Intezet, H-1221 Budapest, Hungary. EM safrany@hp.osski.hu CR Vile RG, Russell SJ, Lemoine NR: Cancer gene therapy: hard lessons and new courses. Gene Ther 7: 2-8, 2000 Inaba M, Sawaa H, Sadata A, Hamada H: Circumvention of 5- fluorouracil resistance in human stomach cancer cells by uracil phosphoribosyl transferase gene transduction. Jpn J Cancer Res 90: 349-354, 1999 Aghi M, Hochberg F, Breakfield XO: Prodrug activation enzymes in cancer gene therapy. J Gene Med 2: 148-164, 2000 Aghi M, Kramm CM, Chou T, et al: Synergistic anticancer effects of ganciclovir/thymidine kinase and 5-fluorocytosine/ cytosine deaminase gene therapies. J Natl Cancer Inst 90: 370- 380, 1998 Takamiya Y, Short MP, Ezzeddine ZD, et al: Gene therapy of malignant brain tumors: a rat glioma line bearing the herpes simplex virus type 1-thymidine kinase gene and wild type retrovirus kills other tumor cells. J Neurosci Res 33: 493-503, 1992 Kanai F, Kawakami T, Hamada H, et al: Adenovirus-mediated transduction of Escherichia coli uracil phosphoribosyltransferase gene sensitizes cancer cells to low concentrations of 5- fluorouracil. Cancer Res 58: 1946-1951, 1998 Maron A, Gustin T, Le Roux A, et al: Gene therapy of rat C6 glioma using adenovirus-mediated transfer of the herpes simplex virus thymidine kinase gene: long-term follow-up by magnetic resonance imaging. Gene Ther 3: 315-322, 1996 Desaknai S, Lumniczky K, Esik O, et al: Local tumor irradiation enhances the anti-tumor effect of a double-suicide gene therapy system in a murine glioma model. J Gene Med 5: 377-385, 2003 Shawler DL, Fakhrai H, Van Beveren C, et al: Gene therapy approaches to enhance antitumor immunity. Adv Pharmacol 40: 309-337, 1997 Dranoff G, Jaffee E, Lazenby A, et al: Vaccination with irradiated tumor cells engineered to secrete murine granulocytemacrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc Natl Acad Sci USA 90: 3539-3543, 1993 Allione A, Consalvo M, Nanni P, et al: Immunizing and curative potential of replicating and non-replicating murine mammary adenocarcinoma cells engineered with interleukin IL-2, IL-4, IL-6, IL- 7, IL-10, tumor necrosis factor alpha, granulocyte-macrophage colony-stimulating factor, and gamma-interferon gene or admixed with conventional adjuvants. Cancer Res 54: 6022-6026, 1994 Li J, Andres ML, Fodor I, et al: Evaluation of pGL1-TNF-alpha therapy in combination with radiation. Oncol Res 10: 379-387, 1998 Staba MJ, Mauceri HJ, Kufe DW, et al: Adenoviral TNF-alpha gene therapy and radiation damage tumor vasculature in a human malignant glioma xenograft. Gene Ther 5: 293-300, 1998 Lumniczky K, Desaknai S, Mangel L, et al: Local tumor irradiation augments the anti-tumor effect of cytokine producing autologous cancer cell vaccines in a murine glioma model. Cancer Gene Ther 9: 44-52, 2002 Hann B, Balmain A: Replication of an E1B 55-kilodalton proteindeficient adenovirus ONYX-015 is restored by gain-of-function rather than loss-function p53 mutants. J Virol 77: 11588-11595, 2003 Nemunaitis J, Khuri F, Ganly I, et al: Phase II trial of intratumoral administration of ONYX-015, a replication-selective adenovirus, in patients with refractory head and neck cancer. J Clin Oncol 19: 289-298, 2001 Nemunaitis J, Cunningham C, Buchanan A, et al: Intravenous infusion of a replication-selective adenovirus ONYX-015 in cancer patients: safety, feasibility and biological activity. Gene Ther 8: 746-759, 2001 Rogulski KR, Freytag SO, Zhang K, et al: In vivo antitumor activity of ONYX-015 is influenced by p53 status and is augmented by radiotherapy. Cancer Res 60: 1193-1196, 2000 Geoerger B, Grill J, Opolon P, et al: Potentiation of radiation therapy by the oncolytic adenovirus dl 1520 ONYX-015 in human malignant glioma xenografts. Br J Cancer 89: 577-584, 2003 Lin E, Nemunaitis J: Oncolytic viral therapies. Cancer Gene Ther 11: 643-664, 2004 Stanziale SF, Petrowsky H, et al: Ionizing radiation potentiates the antitumor efficacy of oncolytic herpes simplex virus G207 by upregulating ribonucleotide reductase. Surgery 132: 353-359, 2002 Timiryasova TM, Gridley DS, Chen B, et al: Radiation enhances the anti-tumor effects of vaccinia-p53 gene therapy in glioma. Technol Cancer Res Treat 2: 223-235, 2003 Robson T, Hirst DG: Transcriptional targeting in cancer gene therapy. J Biomed Biotechnol 110-137, 2003 Hallahan DE, Mauceri HJ, Seung LP, et al: Spatial and temporal control of gene therapy using ionizing radiation. Nat Med 1: 786- 791, 1995 Weichselbaum RR, Hallahan DE, Beckett MA, et al: Gene therapy targeted by radiation preferentially radiosensitizes tumor cells. Cancer Res 54: 4266-4269, 1994 Horsman MR, Bohm L, Margison GP, et al: Tumor radiosensitizers – current status of development of various approaches: report of an International Atomic Energy Agency meeting. Int J Radiat Oncol Biol Phys 64: 551-561, 2006 Chastel C, Jiricny J, Jaussi R: Activation of stress-responsive promoters by ionizing radiation for deployment in targeted gene therapy. DNA Repair 3: 201-215, 2004 Manome Y, Kunieda T, Wen PY, et al: Transgene expression in malignant glioma using a replication-defective adenoviral vector containing the Egr-1 promoter: activation by ionizing radiation or uptake of radioactive iodo-deoxyuridine. Hum Gene Ther 9: 1409-1417, 1998 Weichselbaum RR, Kufe DW, Hellman S, et al: Radiation-induced tumor necrosis factor-a expression: clinical application of transcriptional and physical targeting of gene therapy. Lancet Oncol 3: 665-671, 2002 Joki T, Nakamura M, Ohno T: Activation of the radiosensitive EGR-1 promoter induces expression of the herpes simplex virus thymidine kinase gene and sensitivity of human glioma cells to ganciclovir. Hum Gene Ther 6: 1507-1513, 1995 Worthington J, Robson T, O’Keeffe M, Hirst DG: Tumor cell radiosensitization using constitutive CMV and radiation inducible WAF1 promoters to drive the iNOS gene: a novel suicide gene therapy. Gene Ther 9: 263-269, 2002 Worthington J, McCarthy HO, Barrett E, et al: Use of the radiation- inducible WAF1 promoter to drive iNOS gene therapy as a novel anti-cancer treatment. J Gene Med 6: 673-680, 2004 Marples B, Greco O, Joiner MC, Scott SD: Radiogenetic therapy: strategies to overcome tumor resistance. Curr Pharm Des 9: 2105- 2112, 2003 Shibata T, Giaccia AJ, Brown JM: Development of a hypoxiaresponsive vector for tumor-specific gene therapy. Gene Ther 7: 493-498, 2000 J Gene Med Clinical Trial Site. Rainov NG: A phase III clinical evaluation of herpes simplex virus type 1 thymidine kinase and ganciclovir gene therapy as an adjuvant to surgical resection and radiation in adults with previously untreated glioblastoma multiforme. Hum Gene Ther 20: 2389-2401, 2000 Immonen A, Vapalahti M, Tyynela K, et al: AdvHSV-tk gene therapy with intravenous ganciclovir improves survival in human malignant glioma: a randomised, controlled study. Mol Ther 10: 967-972, 2004 Palmer DH, Young LS, Mautner V: Cancer gene-therapy: clinical trials. Trends Biotechnol 24: 76-82, 2006 Laheru D, Jaffe EM: Immunotherapy for pancreatic cancer – science driving clinical progress. Nat Rev Cancer 5: 459-467, 2005 Pulkkanen KJ, Yla-Herttuala S: Gene therapy for malignant glioma: current clinical status. Mol Ther 12: 585-598, 2005 Lawler SE, Peruzzi PP, Chiocca EA: Genetic strategies for brain tumor therapy. Cancer Gene Ther 13: 225-233, 2006 Makower D, Rozenblit A, Kaufman H, et al: Phase II clinical trial of intralesional administration of the oncolytic adenovirus ONYX-015 in patients with hepatobiliary tumors with correlative p53 studies. Clin Cancer Res 9: 693-702, 2003 Chiocca EA, Abbed KM, Tatter S, et al: A phase I open-label, dose-escalation, multi-institutional trial of injection with an E1Battenuated adenovirus, ONYX-015, into the peritumoral region of recurrent malignant gliomas, in the adjuvant setting. Mol Ther 10: 958-966, 2004 Khuri FR, Nemunaitis J, Gantly I, et al: A controlled trial of intratumoral ONYX-015, a selectively replicating adenovirus in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat Med 16: 879-885, 2000 Dilber MS, Gahrton G: Suicide gene therapy: possible applications in haematopoietic disorders. J Intern Med 249: 359-367, 2001 van Dillen IJ, Mulder NH, Vaalburg W, et al: Influence of the bystander effect on HSV-TK/GCV gene therapy. A review. Curr Gene Ther 2: 307-322, 2002 Namba H, Iwadate Y, Kawamura K, et al: Efficacy of the bystander effect in the herpes simplex virus thymidine kinasemediated gene therapy is influenced by the expression of connexin43 in the target cells. Cancer Gene Ther 8: 414-420, 2001 Kim YG, Bi W, Feliciano ES, et al: Ganciclovir-mediated cell killing and bystander effect is enhanced in cells with two copies of the herpes simplex virus thymidine kinase gene. Cancer Gene Ther 7: 240-246, 2000 Princen F, Robe P, Lechanteur C, et al: A cell type-specific and gap junction-independent mechanism for the herpes simplex virus-1 thymidine kinase gene/ganciclovir-mediated bystander effect. Clin Cancer Res 5: 3639-3644, 1999 Drake RR, Pitlyk K, McMasters RA, et al: Connexin-independent ganciclovir-mediated killing conferred on bystander effect-resistant cell lines by a herpes simplex virus-thymidine kinaseexpressing colon cell line. Mol Ther 2: 515-523, 2000 Freeman SM, Abboud CN, Whartenby KA, et al: The “bystander effect”: tumor regression when a fraction of the tumor mass is genetically modified. Cancer Res 53: 5274-5283, 1993 Hamel W, Magnelli L, Chiarugi VP, Israel MA: Herpes simplex virus thymidine kinase/ganciclovir-mediated apoptotic death of bystander cells. Cancer Res 56: 2697-2702, 1996 Barba D, Hardin J, Sadelain M, Gage FH: Development of antitumor immunity following thymidine kinase-mediated killing of experimental brain tumors. Proc Natl Acad Sci USA 91: 4348- 4352, 1994 Bi W, Kim YG, Feliciano ES, et al: An HSVtk-mediated local and distant antitumor bystander effect in tumors of head and neck origin in athymic mice. Cancer Gene Ther 4: 246-252, 1997 Park JY, Elshami AA, Amin K, et al: Retinoids augment the bystander effect in vitro and in vivo in herpes simplex virus thymidine kinase/ganciclovir-mediated gene therapy. Gene Ther 4: 909-917, 1997 Robe PA, Princen F, Martin D, et al: Pharmacological modulation of the bystander effect in the herpes simplex virus thymidine kinase/ganciclovir gene therapy system: effects of dibutyryl adenosine 3’,5’-cyclic monophosphate, alpha-glycyrrhetinic acid, and cytosine arabinoside. Biochem Pharmacol 60: 241-249, 2000 Kunishige I, Samejima Y, Moriyama A, et al: cAMP stimulates the bystander effect in suicide gene therapy of human choriocarcinoma. Anticancer Res 18: 3411-3419, 1998 Mesnil M, Piccoli C, Tiraby G, et al: Bystander killing of cancer cells by herpes simplex virus thymidine kinase gene is mediated by connexins. Proc Natl Acad Sci USA 93: 1831-1835, 1996 Duflot-Dancer A, Piccoli C, Rolland A, et al: Long-term connexin- mediated bystander effect in highly tumorigenic human cells in vivo in herpes simplex virus thymidine kinase/ganciclovir gene therapy. Gene Ther 5: 1372-1378, 1998 Sanson M, Marcaud V, Robin E, et al: Connexin 43-mediated bystander effect in two rat glioma cell models. Cancer Gene Ther 9: 149-155, 2002 Walling HW, Swarthout JT, Culver KW: Bystander-mediated regression of osteosarcoma via retroviral transfer of the herpes simplex virus thymidine kinase and human interleukin-2 genes. Cancer Gene Ther 7: 187-196, 2000 Liu CS, Kong B, Xia HH, et al: VP22 enhanced intercellular trafficking of HSV thymidine kinase reduced the level of ganciclovir needed to cause suicide cell death. J Gene Med 3: 145-152, 2001 Hyer ML, Sudarshan S, Schwartz DA, et al: Quantification and characterization of the bystander effect in prostate cancer cells following adenovirus-mediated FasL expression. Cancer Gene Ther 10: 330-339, 2003 Seol JY, Park KH, Hwang CI, et al: Adenovirus-TRAIL can overcome TRAIL resistance and induce a bystander effect. Cancer Gene Ther 10: 540-548, 2003 Park SY, Seol JW, Lee YJ, et al: IFN-gamma enhances TRAILinduced apoptosis through IRF-1. Eur J Biochem 271: 4222- 4228, 2004 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2006 VL 12 IS 2 BP 118 EP 124 PG 7 ER PT J AU Nagy, K Szekely-Szuts, K Izeradjene, K Douglas, L Tillman, M Barti-Juhasz, H Dominici, M Spano, C Luca Cervo, G Conte, P Houghton, AJ Mihalik, R Kopper, L Petak, I AF Nagy, Katalin Szekely-Szuts, Kinga Izeradjene, Kamel Douglas, Leslie Tillman, Mike Barti-Juhasz, Helga Dominici, Massimo Spano, Carlotta Luca Cervo, Gian Conte, Pierfranco Houghton, A Janet Mihalik, Rudolf Kopper, Laszlo Petak, Istvan TI Proteasome Inhibitors Sensitize Colon Carcinoma Cells to TRAIL-Induced Apoptosis via Enhanced Release of Smac/DIABLO from the Mitochondria SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TRAIL; epoxomicin; MG132; bortezomib/PS-341; Smac/DIABLO; colon carcinoma ID TRAIL; epoxomicin; MG132; bortezomib/PS-341; Smac/DIABLO; colon carcinoma AB The synergistic interaction between proteasome inhibitors and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a promising approach to induce cell death in tumor cells. However, the molecular and biochemical mechanisms of this synergism have been proven to be cell type specific. We therefore focused our investigation on TRAIL-resistant colon carcinoma cells in this study. DNA fragmentation, mitochondrial membrane depolarization and increased caspase-3-like enzyme activity was exclusively induced only by combined treatment with proteasome inhibitors (epoxomicin, MG132, bortezomib/PS-341) and TRAIL. The expression level of anti-apoptotic proteins (XIAP, survivin, Bcl-2, Bcl-XL), regulated by NF-κB transcription factor, was not effected by any of these treatments. TRAIL alone induced only partial activation of caspase-3 (p20), while the combination of TRAIL and proteasome inhibition led to the full proteolytic activation of caspase-3 (p17). Only the combination treatment induced marked membrane depolarization and the release of cytochrome c, HtrA2/Omi and Smac/DIABLO. Apoptosis-inducing factor (AIF) was not released in any of these conditions. These results are consistent with a model where the full activation of caspase-3 by caspase-8 is dependent on the release of Smac/DIABLO in response to the combined treatment. This molecular mechanism, independent of the inhibition NF-κB activity, may provide rationale for the combination treatment of colon carcinomas with proteasome inhibitors and recombinant TRAIL or agonistic antibody of TRAIL receptors. C1 [Nagy, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Szekely-Szuts, Kinga] St. Jude Children’s Research Hospital, Department of Hematology-Oncology, Division of Molecular TherapeuticsMemphis, TN, USA. [Izeradjene, Kamel] St. Jude Children’s Research Hospital, Department of Hematology-Oncology, Division of Molecular TherapeuticsMemphis, TN, USA. [Douglas, Leslie] St. Jude Children’s Research Hospital, Department of Hematology-Oncology, Division of Molecular TherapeuticsMemphis, TN, USA. [Tillman, Mike] St. Jude Children’s Research Hospital, Department of Hematology-Oncology, Division of Molecular TherapeuticsMemphis, TN, USA. [Barti-Juhasz, Helga] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Dominici, Massimo] University of Modena & Reggio Emilia, Department of Oncology, Hematology & Respiratory DiseasesModena, Italy. [Spano, Carlotta] University of Modena & Reggio Emilia, Department of Oncology, Hematology & Respiratory DiseasesModena, Italy. [Luca Cervo, Gian] University of Modena & Reggio Emilia, Department of Oncology, Hematology & Respiratory DiseasesModena, Italy. [Conte, Pierfranco] University of Modena & Reggio Emilia, Department of Oncology, Hematology & Respiratory DiseasesModena, Italy. [Houghton, A Janet] St. Jude Children’s Research Hospital, Department of Hematology-Oncology, Division of Molecular TherapeuticsMemphis, TN, USA. [Mihalik, Rudolf] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular PathologyBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Petak, Istvan] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Petak, I (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM petak@kkk.org.hu CR Chaudan D, Hideshima T, Mitsiades C, et al: Proteasome inhibitor therapy in multiple myeloma. Mol Cancer Ther 4:686- 692, 2005 Kloetzel PM: The proteasome and MHC class I antigen processing. Biochim Biophys Acta 1695:225-233, 2004 Roos-Mattjus P, Sistonen L: The ubiquitin-proteasome pathway. Ann Med 36:285-295, 2004 Sun L, Chen ZJ: The novel functions of ubiquitination in signaling. Curr Opin Cell Biol 16:119-126, 2004 Mani A, Gelmann EP: The ubiquitin-proteasome pathway and its role in cancer. J Clin Oncol 23:4776-4789, 2005 Petak I, Vernes R, Szucs KS, et al: A caspase-8-independent component in TRAIL/Apo-2L-induced cell death in human rhabdomyosarcoma cells. Cell Death Differ 10:729-739, 2003 Petak I, Houghton JA: Shared pathways: death receptors and cytotoxic drugs in cancer therapy. Pathol Oncol Res 7:95-106, 2001 Petak I, Douglas L, Tillman DM, et al: Pediatric rhabdomyosarcoma cell lines are resistant to Fas-induced apoptosis and highly sensitive to TRAIL-induced apoptosis. Clin Cancer Res 6:4119-4127, 2000 Zhang L, Fang B: Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Ther 12:228-237, 2005 Guseva NV, Taghiyev AF, Sturm MT, et al: Tumor necrosis factor- related apoptosis-inducing ligand-mediated activation of mitochondria-associated nuclear factor-kappaB in prostatic carcinoma cell lines. Mol Cancer Res 2:574-584, 2004 Aggarwal BB, Bhardwaj U, Takada Y: Regulation of TRAILinduced apoptosis by ectopic expression of antiapoptotic factors. Vitam Horm 67:453-483, 2004 Yang LQ, Fang DC, Wang RQ, Yang SM: Effect of NF-kappaB, survivin, Bcl-2 and caspase3 on apoptosis of gastric cancer cells induced by tumor necrosis factor related apoptosis inducing ligand. World J Gastroenterol 10:22-25, 2004 Bortul R, Tazzari PL, Cappellini A, et al: Constitutively active Akt1 protects HL60 leukemia cells from TRAIL-induced apoptosis through a mechanism involving NF-kappaB activation and cFLIP(L, up-regulation. Leukemia 17:379-89, 2003 Ehrhardt H, Fulda S, Schmid I, et al: TRAIL induced survival and proliferation in cancer cells resistant towards TRAILinduced apoptosis mediated by NF-kappaB. Oncogene 22:3842-3852, 2003 Franco AV, Zhang XD, Van Berkel E, et al: The role of NF-kappa B in TNF-related apoptosis-inducing ligand, TRAIL)-induced apoptosis of melanoma cells. J Immunol 166:5337-5345, 2001 Jeremias I, Kupatt C, Baumann B, et al: Inhibition of nuclear factor kappaB activation attenuates apoptosis resistance in lymphoid cells. Blood 91:4624-4631, 1998 Johnson TR, Stone K, Nikrad M, et al: The proteasome inhibitor PS-341 overcomes TRAIL resistance in Bax and caspase 9-negative or Bcl-xL overexpressing cells. Oncogene 22:4953-4963, 2003 Zhu H, Guo W, Zhang L, et al: Proteasome inhibitors-mediated TRAIL resensitization and Bik accumulation. Cancer Biol Ther 4:781-786, 2005 Zhu H, Zhang L, Dong F, et al: Bik/NBK accumulation correlates with apoptosis-induction by bortezomib, PS-341, Velcade, and other proteasome inhibitors. Oncogene 24:4993- 4999, 2005 Nikrad M, Johnson T, Puthalalath H, et al: The proteasome inhibitor bortezomib sensitizes cells to killing by death receptor ligand TRAIL via BH3-only proteins Bik and Bim. Mol Cancer Ther 4:443-449, 2005 Schneider P: Production of recombinant TRAIL and TRAIL receptor: Fc chimeric proteins. Methods Enzymol 322:325-3245, 2000 Leverkus M, Sprick MR, Wachter T, et al: Proteasome inhibition results in TRAIL sensitization of primary keratinocytes by removing the resistance-mediating block of effector caspase maturation. Mol Cell Biol 23:777-790, 2003 Chauhan D, Li G, Podar K, et al: Targeting mitochondria to overcome conventional and bortezomib/proteasome inhibitor PS-341 resistance in multiple myeloma, MM, cells. Blood 104:2458-2466, 2004 LaVallee TM, Zhan XH, Johnson MS, et al: 2-methoxyestradiol up-regulates death receptor 5 and induces apoptosis through activation of the extrinsic pathway. Cancer Res 63:468-475, 2003 Creagh EM, Murphy BM, Duriez PJ, et al: Smac/Diablo antagonizes ubiquitin ligase activity of inhibitor of apoptosis proteins. J Biol Chem 279:26906-26914, 2004 Du C, Fang M, Li Y, et al: Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 102:33-42, 2000 Ni H, Ergin M, Huang Q, et al: Analysis of expression of nuclear factor kappa B, NF-kappa B, in multiple myeloma: downregulation of NF-kappa B induces apoptosis. Br J Haematol 115:279-286, 2001 Berenson JR, Ma HM, Vescio R: The role of nuclear factor-kappaB in the biology and treatment of multiple myeloma. Semin Oncol 28:626-633, 2001 Tillman DM, Izeradjene K, Szucs KS, et al: Rottlerin sensitizes colon carcinoma cells to tumor necrosis factor-related apoptosis- inducing ligand-induced apoptosis via uncoupling of the mitochondria independent of protein kinase C. Cancer Res 63:5118-5125, 2003 Ganten TM, Koschny R, Haas TL, et al: Proteasome inhibition sensitizes hepatocellular carcinoma cells, but not human hepatocytes, to TRAIL. Hepatology 42:588-597, 2005 Lashinger LM, Zhu K, Williams SA, et al: Bortezomib abolishes tumor necrosis factor-related apoptosis-inducing ligand resistance via a p21-dependent mechanism in human bladder and prostate cancer cells. Cancer Res 65:4902-4908, 2005 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2006 VL 12 IS 3 BP 133 EP 142 PG 10 ER PT J AU Johannessen, LA Torp, HS AF Johannessen, Linn Anne Torp, Helge Sverre TI The Clinical Value of Ki-67/MIB-1 Labeling Index in Human Astrocytomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Brain-tumors; gliomas-immunohistochemistry; Ki-67; prognosis ID Brain-tumors; gliomas-immunohistochemistry; Ki-67; prognosis AB The current WHO classification of human astrocytomas has limitations in predicting prognosis and diagnosis, and there is a need for additional factors. Several studies have investigated the clinical value of proliferative activity in these tumors, especially the Ki-67/MIB-1 labeling index (LI). The aim of this study was to review the literature on this topic to get a survey of the current experience. All studies show increasing values of Ki-67/MIB-1 LI with increasing grade of malignancy. Most of them demonstrate that MIB-1 LI differentiates well between diffuse astrocytomas WHO grade II (AII) and anaplastic astrocytomas (AA) and between AII and glioblastomas (GM), but not between AA and GM. There is, however, considerable overlap of indices between the different malignancy groups. Further, in most studies positive correlations between MIB-1 LI and survival are found, though the proposed cut-off values vary substantially between the reports. The studies reviewed report MIB-1 LI as an important prognostic factor in human astrocytomas. Due to the great spread of values between the various tumor grades, however, MIB-1 LI cannot be used as a diagnostic factor alone but should be used in combination with established criteria of histological malignancy. It may be especially useful in cases where histology reveals a low-grade astrocytoma whereas other parameters indicate a more malignant neoplasm. Thus, it is our opinion that MIB-1 LI should be a part of the routine investigation in patients with astrocytic tumors. Until larger multicenter studies based on standardized immunohistopathological procedures have been completed, each laboratory has to establish its own practice. C1 [Johannessen, Linn Anne] St. Olavs Hospital, Trondheim University Hospital, Department of Pathology and Medical Genetics, Department of Laboratory Medicine, Children’s and Women’s Health, N-7006 Trondheim, Norway. [Torp, Helge Sverre] St. Olavs Hospital, Trondheim University Hospital, Department of Pathology and Medical Genetics, Department of Laboratory Medicine, Children’s and Women’s Health, N-7006 Trondheim, Norway. RP Torp, HS (reprint author), St. Olavs Hospital, Trondheim University Hospital, Department of Pathology and Medical Genetics, Department of Laboratory Medicine, Children’s and Women’s Health, N-7006 Trondheim, Norway. EM sverre.torp@ntnu.no CR Di Nishizaki T, Harada K, et al: Proliferative potentials of glioma cells and vascular components determined with monoclonal antibody MIB-1. J Exp Clin Canc Res 16:389-394, 1997 Enestrom S, Vavruch L, Franlund B, et al: Ki-67 antigen expression as a prognostic factor in primary and recurrent astrocytomas. Neurochir 44:25-30, 1998 Gerdes J, Schwab U, Lemke H, et al: Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer 31:13-20, 1983 Gerdes J, Li L, Schlueter, et al: Immunobiochemical and molecular biologic characterization of the cell proliferation-associated nuclear antigen that is defined by monoclonal antibody Ki- 67. Am J Pathol 138: 867-873, 1991 Hilton DA, Love S, Barber R, et al: Accumulation of p53 and Ki-67 expression do not predict survival in patients with fibrillary astrocytomas or the response of these tumors to radiotherapy. Neurosurgery 42:724-729, 1998 Hsu DW, Louis DN, Efird JT, et al: Use of MIB-1, Ki-67, immunoreactivity in differentiating grade II and grade III gliomas. J Neuropathol Exp Neurol 56:857-865, 1997 Karamitopoulou E, Perentes E, Diamantis I, et al: Ki-67 immunoreactivity in human central nervous system tumors: a study with MIB 1 monoclonal antibody on archival material. Acta Neuropathol 87:47-54, 1994 Key G, Becker MHG, Baron B, et al: New Ki-67-equivalent murine monoclonal antibodies, MIB 1-3, generated against bacterially expressed parts of the Ki-67 cDNA containing three 62 base pair repetitive elements encoding for the Ki-67 epitope. Lab Invest 68: 629-636, 1993 Khalid H, Shibata S, Kishikawa M, et al: Immunohistochemical analysis of progesterone receptor and Ki-67 labeling index in astrocytic tumors. Cancer 80: 2133-2140, 1997 Kleihuses P, Cavanee WK: World Health Organization Classification of Tumors. Pathology and Genetics of Tumors of the Nervous System. IARC Press, Lyon, 2000 McKeever PE, Ross DA, Strawderman MS, et al: A comparison of the predictive power for survival in gliomas provided by MIB-1, bromodeoxyuridine and proliferating cell nuclear antigen with histopathologic and clinical parameters. J Neuropathol Exp Neurol 56:798-805, 1997 McKeever PE, Strawderman MS, Yamini B, et al: MIB-1 proliferation index predicts survival among patients with grade II astrocytoma. J Neuropathol Exp Neurol 57:931-936, 1998 Neder L, Colli BO, Machado HR, et al: MIB-1 labeling index in astrocytic tumors – a clinicopathologic study. Clin Neuropathol 6: 262-270, 2004 Ralte AM, Sharma MC, Karak AK, et al: Clinicopathological features, MIB-1 labeling index and apoptotic index in recurrent astrocytic tumors. Pathol Oncol Res 7:267-278, 2001 Reavey-Cantwell JF, Haroun RI, Zahurak M, et al: The prognostic value of tumor markers in patients with glioblastoma multiforme: analysis of 32 patients and review of the literature. J Neuro-Oncol 55:195-204, 2001 Sallinen PK, Haapasalo HK, Visakorpi T, et al: Prognostication of astrocytoma patient survival by Ki-67, MIB-1), PCNA, and S-phase fraction using archival paraffin-embedded samples. J Pathol 174: 275-282, 1994 Schiffer D, Cavalla P, Chio A, et al: Proliferative activity and prognosis of low-grade astrocytomas. J Neuro-Oncol 34:31-35, 1997 Torp SH: Diagnostic and prognostic role of Ki67 immunostaining in human astrocytomas using four different antibodies. Clin Neouropathol 21:252-257, 2002 Torp SH, Alsaker M: Ki-67 immunoreactivity, basic fibroblast growth factor, bFGF, expression, and microvessel density as supplementary prognostic tools in low-grade astrocytomas. Pathol Res Pract 198:261-265, 2002 Wakimoto H, Aoyagi M, Nakayama T, et al: Prognostic significance of Ki-67 labeling indices obtained using MIB-1 monoclonal antibody in patients with supratentorial astrocytomas. Cancer 77:373-380, 1996 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2006 VL 12 IS 3 BP 143 EP 147 PG 5 ER PT J AU Lorincz, T Toth, J Badalian, G Timar, J Szendroi, M AF Lorincz, Tamas Toth, Jozsef Badalian, Gayane Timar, Jozsef Szendroi, Miklos TI HER-2/neu Genotype of Breast Cancer May Change in Bone Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; bone metastatic; HER-2 ID breast cancer; bone metastatic; HER-2 AB The genotype of breast cancer (BRC) is considered to be relatively stable during tumor progression, accordingly, determination of the estrogen receptor and HER-2/neu status is currently based on the primary tumor. However, recent data suggest that the gene expression profile of the metastatic lesion can be different compared to that of the primary BRC. Accordingly, it is possible that the HER-2/neu status is different in the metastatic lesion and the primary BRC. Since the bone is the most frequent metastatic site during the progression of BRC, we have analyzed the HER-2/neu status of 48 bone metastatic BRC cases by immunohistochemistry and fluorescent in situ hybridization, and it was possible to compare it to the primary site in 23 cases. The frequency of HER-2/neu amplification of BRC in the primary tumors was found to be 17.4% compared to 10.5% in bone metastases. Half of BRC cases with HER-2/neu amplification lost this genotype in bone metastases (4/23 versus 2/23, respectively) and even in the 2 cases where HER-2/neu amplification was retained in the metastases, the copy number was found to be decreased compared to the primary tumor. Based on our data and previous reports in the literature, we suggest to perform HER-2/neu testing both on primary tumor and samples obtained from BRC metastases, at least in case of primary tumors with HER-2/neu amplification, before introduction of HER-2/neu-targeting therapy. C1 [Lorincz, Tamas] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Toth, Jozsef] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Badalian, Gayane] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Szendroi, Miklos] Semmelweis University, Department of OrthopedicsBudapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. CR Mendelsohn J, Baselga J: The EGF receptor family as targets for cancer therapy. Oncogene 19: 6550-6565, 2000 Slamon DJ, Clark GM, Wong SG, et al: Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235:177-182, 1987 Yu D, Hung MC: Overexpression of ErbB2 in cancer and ErbB2-targeting strategies. Oncogene 19: 6115-6121, 2000 Tsuda H, Hirohashi S, Shimotaso Y, et al: Correlation between long term survival in breast cancer patients and amplification of two putative oncogene co-amplification units: hst-1/int-2 and c-erbB-2/ear-1. Cancer Res 49:3104-3108, 1989 de Bono JS, Rowinsky EK: The ErbB receptor family: a therapeutic target for cancer. Trends Mol Med 8, 4 suppl): S19-S26, 2002 Ross JS, Fletcher JA: HER-2/neu, c-erb-B2, gene and protein in breast cancer. Am J Clin Pathol 112: 53-67, 1999 Cobleigh MA, Vogel CL, Triphaty D et al: Multinational study of the efficacy and safety of humanized anti-HER-2/NEU monoclonal antibody in woman who have HER-2/NEU-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol 17: 2639-2648, 1999 Smith IE: Efficacy and safety of Herceptin®in women with metastatic breast cancer: Result from pivotal clinical studies. Anticancer Drugs 12, suppl 4): S3-S10, 2001 Di Leo A, Dowsett M, Horten B, et al: Current status of HER-2/neu testing. Oncology 63, suppl 1): 25-32, 2002 Gancberg M, Di Leo A, Rouas G et al: Comparison of topoisomerase IIα, topoIIα, and HER-2 status between primary, P, breast cancer, BC, and corresponding metastatic, M, sites. Ann Oncol 13: 1036-1043, 2002 Fidler IJ: Review: biologic heterogeneity of cancer metastasis. Breast Cancer Res Treat 9:17-26,1987 Masood S, Bui MM: Assessment of Her-2/neu overexpression in primary breast cancers and their metastatic lesions: an immunohistochemical study. Ann Clin Lab Sci 30: 259-265, 2000 Shimizu C, Fukutomi T, Tsuda H et al: c-erbB-2 protein overexpression and p53 immunoreaction in primary and recurrent breast cancer tissues. J Surg Oncol 73: 17-20, 2000 Carlsson J, Nordgren H, Sjostrom J et al: HER-2/NEU expression in breast cancer primary tumours and corresponding metastases. Original data and literature review. Br J Cancer 90: 2344-2348, 2004 Vincent-Salomon A, Jouve M, Genin P et al: HER-2/NEU status in patients with breast carcinoma is not modified selectively by preoperative chemotherapy and is stable during the metastatic process. Cancer 94: 2169-2173, 2002 Tanner M, Jarvinen P, Isola J: Amplification of Her-2/neu and topoisomerase IIα in primary and metastatic breast cancer. Cancer Res 61:5345-5348, 2001 Kunitomo K, Inoue S, Ichihara F et al: A case of metastatic breast cancer with outgrowth of HER-2/NEU-negative cells after eradication of HER-2/NEU-positive cells by humanized anti-HER-2/NEU monoclonal antibody, trastuzumab, combined with docetaxel. Hum Pathol 35: 379-381, 2004 Zidan J, Dashkovsky I, Stayerman C et al: Comparison of HER-2 overexpression in primary breast cancer and metastatic sites and its effect on biological targeting therapy of metastatic disease. Br J Cancer 93: 552-556, 2005 Latinovic L, Heinze G, Birner P et al: Prognostic relevance of three histological grading methods in breast cancer. Int J Oncol 19: 1271- 1277, 2001 Press MF, Slamon DJ, Flom KJ et al: Evaluation of HER-2/neu gene amplification and overexpression: comparison of frequently used assay methods in a molecularly characterized cohort of breast cancer specimens. J Clin Oncol 20: 3095-3105, 2002 Bruun Rasmussen B, Kamby C: Immunohistochemical detection of oestrogen receptors in paraffin sections from primary and metastatic breast cancer. Pathol Res Pract 185:856-859, 1989 Woelfle U, Cloos J et al: Molecular signature associated with bone marrow micrometastasis in human breast cancer. Cancer Res 63:5679-5684, 2003 Yoneda T: Cellular and molecular mechanism of breast and prostate cancer metastasis to bone. Eur J Cancer 34: 240-245, 1998 Gong Y, Booser DJ, Sneige N: Comparison of HER-2 status determined by fluorescence in situ hybridization in primary and metastatic breast carcinoma. Cancer 103: 1763-1769, 2005 Regitnig P, Schippinger W, Lindbauer M et al. Change of HER-2/neu status in a subset of distant metastases from breast carcinomas. J Pathol 203: 918-926, 2004 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2006 VL 12 IS 3 BP 149 EP 152 PG 4 ER PT J AU Nogueira, CM Guedes Neto, dPE Rosa, WM Zettler, E Zettler, GC AF Nogueira, Cordoni Mauricio Guedes Neto, de Paula Ernesto Rosa, Wengrover Marcos Zettler, Eduardo Zettler, Galleano Claudio TI Immunohistochemical Expression of p16 and p53 in Vulvar Intraepithelial Neoplasia and Squamous Cell Carcinoma of the Vulva SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE vulvar cancer; vulvar intraepithelial neoplasia; p16; p53 ID vulvar cancer; vulvar intraepithelial neoplasia; p16; p53 AB This study was undertaken to examine the expression of p16 and p53 in vulvar intraepithelial neoplasia (VIN) and squamous cell carcinoma (SCC) of the vulva. We also analyzed the relationship between p16 and p53 immunoexpression in women younger vs. older than 55 years of age. Seventyseven histologic samples of vulvar tissue, treated surgically between June 2000 and November 2004 at the Complexo Hospitalar Santa Casa (Porto Alegre, Brazil), were investigated. We analyzed 28 cases of VIN, 37 cases of SCC and 12 normal vulvar tissues. The percentage of immunohistochemical positivity for p16 had the following distribution across the groups: VIN: 21.4% (6/28), cancer: 24.3% (9/37) and control: absent (p=0.202). p53 expression showed the following percentages: VIN: 60.7% (17/28), cancer: 18.9% (7/37) and control: 8.3% (1/12) (p=0.01). p16 expression in the cancer group (mean age: 63.4 years) was positive in 6 and 3 cases of women younger or older than 55 years, respectively (54.5% vs. 11.5%, p=0.01). p53 expression was not detected in young females with cancer, while it was expressed in 7/26 (26.9%) cases of the group of females older than 55 years of age (p=0.08). Our results suggest an increase in the immunohistochemical expression of p16 protein in young women with squamous cell carcinoma of the vulva, and a possible association with a low expression of p53. C1 [Nogueira, Cordoni Mauricio] Complexo Hospitalar Santa Casa de Porto Alegre, Santa Rita Cancer Center, Gynecologic Oncology Group, Rua Prof. Annes Dias, 285, 90020-090 Porto Alegre, Brazil. [Guedes Neto, de Paula Ernesto] Complexo Hospitalar Santa Casa de Porto Alegre, Santa Rita Cancer Center, Gynecologic Oncology Group, Rua Prof. Annes Dias, 285, 90020-090 Porto Alegre, Brazil. [Rosa, Wengrover Marcos] Complexo Hospitalar Santa Casa de Porto Alegre, Santa Rita Cancer Center, Gynecologic Oncology Group, Rua Prof. Annes Dias, 285, 90020-090 Porto Alegre, Brazil. [Zettler, Eduardo] Santa Casa Hospital Complex, Santa Rita Hospital, Department of PathologyPorto Alegre, Brazil. [Zettler, Galleano Claudio] Santa Casa Hospital Complex, Santa Rita Hospital, Department of PathologyPorto Alegre, Brazil. RP Nogueira, CM (reprint author), Complexo Hospitalar Santa Casa de Porto Alegre, Santa Rita Cancer Center, Gynecologic Oncology Group, 90020-090 Porto Alegre, Brazil. EM nogueira@santacasa.tche.br CR Landis SH, Murray T, Bolden S, Wingo PA: Cancer statistics, 1998. CA Cancer J Clin 48:6-29, 1998. Creasman WT, Phillips JL, Menck HR: The National Cancer Data Base report on early stage invasive vulvar carcinoma. The American College of Surgeons Commission on Cancer and the American Cancer Society. Cancer 80:505-513, 1997. National Cancer Institute, NCI): Cancer Facts and Figures 2005. Washington, DC). Available: http://www.nci.nih.gov/ cancertopics/pdq/vulvar. Ministerio da Saude, Brasil): Instituto Nacional do Cancer, INCA, – Cancer no Brasil: Dados dos registros de Base populacional. Available: http://www.inca.gov.br/regpop/2003/index. McNally OM, Mulvany NJ, Pagano R, et al: VIN 3: a clinicopathologic review. Int J Gynecol Cancer 12: 490-495, 2002. Ghurani GB, Penalver MA: An update on vulvar cancer. Am J Obstet Gynecol 185: 294-299, 2001. Al-Ghandi A, Freedman D, Miller D, et al: Vulvar squamous cell carcinoma in young women: a clinicopathologic study of 21 cases. Gynecol Oncol 84:94-101, 2002. Canavan TP, Cohen D: Vulvar cancer. Am Fam Physician 66:1269-1274, 2002. Trimble CL, Hildesheim A, Brinton LA, et al: Heterogeneous etiology of squamous carcinoma of the vulva. Obstet Gynecol 87: 59-64, 1996. International Agency for Research on Cancer, WHO). International Association of Cancer Registries, IARC): Cancer Incidence in Five Continents. Vol. VI, Scientific Publications 120, Lyon, 1992. Nash JD, Curry S: Vulvar cancer. Surg Oncol North Am 7: 335-346, 1998. Grendys EC, Fiorica JV: Innovations in the management of vulvar carcinoma. Curr Opin Obstet Gynecol 12: 15-20, 2000. Pinto AP, Signorello LB, Crum CP, et al: Squamous cell carcinoma of the vulva in Brazil: prognostic importance of host and viral variables. Gynecol Oncol 74: 61-67, 1999. Kaufman RH, Adam E, Vonka V: Human papillomavirus infection and cervical carcinoma. Clin Obstet Gynecol 43: 363-379, 2000. Beutner KR, Tyring S: Human papillomavirus and human disease. Am J Med 5: 9-15, 1997. Bloss JD, Liao SY, Wilczynski SP: Clinical and histologic features of vulva carcinomas analyzed for human papillomavirus status: Evidence that squamous cell carcinoma of the vulva has more than one etiology. Hum Pathol 22: 711-718, 1991. Nobori T, Miura K, Wu DJ, Lois A: Deletions of the cyclindependent kinase-4 inhibitor gene in multiple human cancers. Nature 368: 753-756, 1994. Gasco M, Sullivan A, Repellin C, et al: Coincident inactivation of 14-3-3 sigma and p16INK4 is an early event in vulvar squamous neoplasia. Oncogene 21: 1876-1881, 2002. Schorge JO, Lea JS, Elias KJ, et al: p16 as a molecular biomarker of cervical adenocarcinoma. Am J Obstet Gynecol 190: 668-673, 2004. Fregonesi PA, Teresa DB, Duarte RA, et al: p16(INK4A, immunohistochemical overexpression in premalignant and malignant oral lesions infected with human papillomavirus. J Histochem Cytochem 51: 1291-1297, 2003. Chan MK, Cheung TH, Chung TK, et al: Expression of p16INK4 and retinoblastoma protein rb in vulvar lesions of Chinese women. Gynecol Oncol 68:156-161, 1997. Hollstein M, Sidransky D, Vogelstein B, Harris C: p53 mutations in human cancers. Science 117: 291-294, 1991. Ellis PE, Fong LF, Rolfe KJ, et al: The role of p53 and Ki67 in Paget’s disease of the vulva and the breast. Gynecol Oncol 86: 150-156, 2002. Val IC, Almeida GF, Valiante PM, et al: Vulvar intraepithelial neoplasia p53 expression in recurrent/progressive cases. J Reprod Med 49: 868-874, 2004. Branton PA: College of American Pathologists, CAP). Protocol Revision date: January 2005., Based on WHO, AJCC/UICC, TNM 6th and FIGO Annual Report). Washington, DC);, Protocol applies to pre-malignant and invasive carcinomas of the vulva), 2005. Santos M, Montagut C, Mellado B, et al: Immunohistochemical staining for p16 and p53 in premalignant and malignant epithelial lesions of the vulva. Int J Gynecol Pathol 23: 206- 214, 2004. Seters M, Beurden M, Craen AJ: Is the assumed natural history of vulvar intraepithelial neoplasia III based on enough evidence? A systematic review of 3322 published patients. Gynecol Oncol 97: 645-651, 2005. Kaufman RH: Intraepithelial neoplasia of the vulva. Gynecol Oncol 56:8-21, 1995. Riethdorf S, Neffen E, Cviko A, et al: P16(INK4A, as biomarker for HPV 16-related vulvar neoplasias. Hum Pathol 35: 1477- 1483, 2004. Esrig D, Spruck CH, Nichols PW: p53 nuclear protein accumulation correlates with mutations in the p53 gene, tumor grade and stage in bladder cancer. Am J Pathol 143: 1389-1392, 1993. Pilotti S, Donghi R, Giarola M, et al: Papilloma virus, p53 alteration and primary carcinoma of the vulva. Eur J Cancer 29: 924-925, 1993. Chen TM, Chen CA, Hsieh CY, et al: The state of p53 in primary human cervical carcinomas and its effects in human papillomavirus- immortalized human cervical cells. Oncogene 8: 1511-1518, 1993. Koyamatsu Y, Yokoyama M, Nakao Y, et al: A comparative analysis of human papillomavirus types 16 and 18 and expression of p53 gene and Ki-67 in cervical, vaginal, and vulvar carcinomas. Gynecol Oncol 90: 547-551, 2003. Gualco M, Bonin S, Foglia G: Morphologic and biologic studies on ten cases of verrucous carcinoma of the vulva supporting the theory of a discrete clinicopathologic entity. Int J Gynecol Cancer 13: 317-324, 2003. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2006 VL 12 IS 3 BP 153 EP 157 PG 5 ER PT J AU Szegedi, I Kiss, Cs Karaszi, Vamosi, Gy Szollosi, J Kovacs, P Benko, I AF Szegedi, Istvan Kiss, Csongor Karaszi, Eva Vamosi, Gyorgy Szollosi, Janos Kovacs, Peter Benko, Ilona TI Differential Regulation of Umbilical Cord Blood and Leukemic B Cells by Interferon-Alpha (IFN-α): Observations in Cultured Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apoptosis; B cells; colony formation; interferon-alpha; type II Fcy receptor ID apoptosis; B cells; colony formation; interferon-alpha; type II Fcy receptor AB The exact mechanism of the beneficial therapeutic action of interferon-a (IFN-α) in B-cell-lineage malignancies has not been adequately explained. Here we report on the differential effect of IFN-α2b on non-malignant B cells of umbilical cord blood and leukemic B-cell lines JY, BL-41 and BCBL-1. Leukemic cell proliferation was characterized by colony assay, whereas apoptosis was investigated by flow cytometry of propidium iodide-stained cells. The degree of differentiation was evaluated by measuring the expression level of Fcγ receptor-II (FcγRII) labeled with anti-CD32-FITC monoclonal antibody using flow cytometry. IFN-α protected umbilical cord blood CD19-positive B lymphocytes from apoptotic cell death in vitro. IFN-α significantly decreased colony formation of all three cell lines, and in contrast to normal cells, induced apoptosis in JY and BL-41 and excessive necrosis in HHV-8 infected BCBL-1 cells. FcγRII was upregulated both in normal and in leukemic B cells as indicated by an increase both in the proportion of CD32-positive cells and the mean fluorescence intensity. From our results it seems that antiproliferative, apoptotic and differentiative effects of IFN-α are interrelated but distinct cellular events, which are differentially regulated in normal, leukemic and virus-infected cells of the B-cell lineage. C1 [Szegedi, Istvan] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, H-4012 Debrecen, Hungary. [Kiss, Csongor] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, H-4012 Debrecen, Hungary. [Karaszi, Eva] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Vamosi, Gyorgy] University Medical School of Debrecen, Department of Biophysics and Cell BiologyDebrecen, Hungary. [Szollosi, Janos] University Medical School of Debrecen, Department of Biophysics and Cell BiologyDebrecen, Hungary. [Kovacs, Peter] University of Debrecen, Department of Pharmacology and PharmacotherapyDebrecen, Hungary. [Benko, Ilona] University of Debrecen, Department of Pharmacology and PharmacotherapyDebrecen, Hungary. RP Kiss, Cs (reprint author), Medical and Health Science Center, University of Debrecen, Department of Pediatrics, H-4012 Debrecen, Hungary. EM kisscs@dote.hu CR Balmelli C, Vincent IE, Rau H et al: Fc gamma RII-dependent sensitisation of natural interferon-producing cells for viral infection and interferon-alpha responses. Eur J Immunol 35: 2406-2415, 2005 Benko I, Kovacs P, Szegedi I, et al: Effect of myelotropic and pleiotropic cytokines on colony formation by blast cells of children with acute lymphoblastic leukemia. N-S Arch Pharmacol 363: 499-508, 2001 D’Agostino G, Arico E, Santodonato L et al: Type I consensus IFN, IFN-con1, gene transfer into KSHV/HHV-8-infected BCBL-1 cells causes inhibition of viral lytic cycle activation via induction of apoptosis and abrogates tumorigenicity in SCID mice. J Interferon Cytokine Res 19: 1305-1316, 1999 Heslop HE, Hoffbrand AV: Interferons in haematological malignancy. Recent Adv Haemat 7: 131-148, 1992 Jewell AP: Interferon-alpha, bcl-2 expression and apoptosis in B-cell chronic lymphocytic leukemia. Leuk Lymphoma 21: 43- 47, 1996 Kiss C: Interferons in pediatric oncology: end of the beginning or beginning of the end? Med Pediatr Oncol 30: 364-366, 1998 Kiss C, Surrey S, Schreiber AD et al: Human c-kit ligand, stem cell factor, induces platelet Fc receptor expression in megakaryocytic cells. Exp Hematol 24: 1232-1237, 1996 Martinez-Lostao L, Briones J, Forne I et al: Role of the STAT1 pathway in apoptosis induced by fludarabine and JAK kinase inhibitors in B-cell chronic lymphocytic leukemia. Leuk Lymphoma 46: 435-442, 2005 McSweeney EN, Giles FJ, Worman CP et al: Recombinant alpha 2a interferon therapy in the treatment of patients with early stage B chronic lymphatic leukaemia. Br J Haemat 85: 77-83, 1993 Mikala G, Xie J, Berencsi G, Kiss C et al: Human herpesvirus 8 in hematologic diseases. Pathol Oncol Res 5: 73-79, 1999 Miyauchi J, Kelleher CA, Yang YC et al: The effects of three recombinant growth factors, IL-3, GM-CSF, and G-CSF on the blast cells of acute myeloblastic leukemia maintained in shortterm suspension culture. Blood 70: 657-663, 1987 Pearse RN, Feinman R, Shuai K et al: Interferon g-induced transcription of the high-affinity Fc receptor for IgG requires assembly of a complex that induces the 91-kDa subunit of transcription factor ISGF3. Proc Natl Acad Sci USA 90: 4314- 4318, 1993 Picchio GR, Sabbe RE, Gulizia RJ et al: The KSHV/HHV8- infected BCBL-1 lymphoma line causes tumors in SCID mice but fails to transmit virus to a human peripheral blood mononuclear cell graft. Virology 238: 22-29, 1997 Pouge SL, Preston BT, Stalder J et al: The receptor for type I IFNs is highly expressed on peripheral blood B cells and monocytes and mediates a distinct profile of differentiation and activation of these cells. J Interferon Cytokine Res 24: 131-139, 2004 Pozharskaya VP, Weakland LL, Offermann MK: Inhibition of infectious human herpesvirus 8 production by gamma interferon and alpha interferon in BCBL-1 cells. J Gen Virol 85: 2779- 2787, 2004 Renne R, Zhong W, Herndier B et al: Lytic growth of Kaposi’s sarcoma-associated herpesvirus, Human herpesvirus 8, in culture. Nat Med 2: 342-346, 1996 Santavenere E, Di Pietro R, Centurione MA et al: Synergistic regulatory effects of TNF alpha, IL-1 alpha and IFN alpha on the growth and differentiation of Daudi lymphoma cells. Cell Biol Int 20: 335-338, 1996 Sarmay G, Koncz G, Gergely J: Human type II Fcγ receptors inhibit B cell activation by interacting with the p21ras-dependent pathway. J Biol Chem 271: 30499-30504, 1996 Szollosi J, Horejsi V, Bene L et al: Supramolecular complexes of MHC class I, MHC class II, CD20, and tetraspan molecules, CD53, CD81, and CD82, at the surface of a B cell line JY. J Immunol 157: 2939-2946, 1996 Zimring JC, Goodburn S, Offerman MK: Human herpesvirus encodes an interferon regulatory factor, IRF, homolog that represses IRF-1 mediated transcription. J Virol 72: 701-707, 1998 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2006 VL 12 IS 3 BP 159 EP 163 PG 5 ER PT J AU Malik, A Deb, P Sharma, ChM Sarkar, Ch AF Malik, Ajay Deb, Prabal Sharma, Chand Mehar Sarkar, Chitra TI Neuropathological Spectrum of Pilocytic Astrocytoma - an Indian Series of 120 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pilocytic astrocytoma; vascular spectrum; angiomatous proliferation; glomeruloid vessels; perivascular; hyalinization; hemosiderin ID Pilocytic astrocytoma; vascular spectrum; angiomatous proliferation; glomeruloid vessels; perivascular; hyalinization; hemosiderin AB Pilocytic astrocytomas (PAs) are generally well circumscribed, slowly growing, cystic tumors, occurring in the pediatric age group. Our aims were to retrospectively analyze the neuropathological spectrum of PA, and correlate it with various clinicopathological features. A total of 120 PAs, diagnosed and managed at this center during a 5-year period, were included. The study population had a mean age of 18.9 years, with male predominance (68.3%), and demonstrated predilection for posterior fossa (61.7%). On histopathology, biphasic pattern (89.2%) along with Rosenthal fibers (66.7%) and eosinophilic granular bodies (60%) were present in the majority of cases. Vascular features were characterized by perivascular hyalinization (51.7%), angiomatous proliferation (21.7%) and glomeruloid changes (21.7%). Hemosiderin-laden macrophages were noted in 37.1% of cases. Further, 60.8% showed lymphoplasmacytic infiltration, while atypia and necrosis were present in 25.8% and 1.7% of cases, respectively. Statistical evaluation revealed significant correlation of angiomatous proliferation with age (≤12 and >12-year age groups) (p=0.011); and of hemosiderin deposition with angiomatous proliferation (p=0.006), perivascular hyalinization (p=0.035), and age (≤12 and >12-year age groups) (p=0.028). This study emphasizes that though PAs generally display classical histomorphology, diagnosis may be challenging in patients with unusual clinicopathological features, e.g. in older patients, uncommon location, absence of biphasic pattern, or presence of nuclear atypia, mitotic figures and necrosis, and also in cases of small biopsies. In the absence of diagnostic histology enumerated above, vascular features like angiomatous proliferation, glomeruloid changes and perivascular hyalinization, along with hemosiderinladen macrophages and perivascular lymphocytic infiltration should be considered as surrogate histological markers of PA. C1 [Malik, Ajay] All India Institute of Medical Sciences, Department of Pathology, Ansari Nagar, 110029, New Delhi, India. [Deb, Prabal] All India Institute of Medical Sciences, Department of Pathology, Ansari Nagar, 110029, New Delhi, India. [Sharma, Chand Mehar] All India Institute of Medical Sciences, Department of Pathology, Ansari Nagar, 110029, New Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, Ansari Nagar, 110029, New Delhi, India. RP Sarkar, Ch (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029, New Delhi, India. EM drchitrasarkar@yahoo.com CR Badhe PB, Chauhan PP, Mehta NK: Brainstem gliomas – a clinicopathological study of 45 cases with p53 immunohistochemistry. Indian Cancer 41: 170-174, 2004 Bell D, Chitnavis BP, Al-Sarraj S, et al: Pilocytic astrocytoma of the adult—clinical features, radiological features and management. Br J Neurosurg 18: 613-616, 2004 Berroir S, Lafitte F, Heran F, et al: Pilocytic astrocytoma: unusual feature. J Neuroradiol 28:249-252, 2001 Beutler AS, Hsiang JK, Moorhouse DF, et al: Pilocytic astrocytoma presenting as an extra-axial tumor in the cerebellopontine angle: case report. Neurosurgery 37:125-128, 1995 Boch AL, Cacciola F, Mokhtari K, et al: Benign recurrence of a cerebellar pilocytic astrocytoma 45 years after gross total resection. Acta Neurochirurgica, Wien, 142:341-346, 2000 Burger PC, Scheithauer BW, Paulus W, et al: Pilocytic astrocytoma. In: Pathology and Genetics of Tumors of the Nervous System., Eds: Kleihues P and Cavenee WK), IARC Press, Lyon, France, 2000, pp. 45-47 Burger PC, Scheithauer BW, Vogel FS: The brain tumors. In: Surgical Pathology of the Nervous System and its Coverings., Eds: Burger PC, Scheithauer BW, Vogel FS), Churchill Livingstone, New York, 2002, pp.160-378 Burkhard C, Di Patre PL, Schuler D, et al: A population-based study of the incidence and survival rates in patients with pilocytic astrocytoma. J Neurosurg 98: 1170-1174, 2003 Coakley KJ, Huston J, Scheithauer BW, et al: Pilocytic astrocytomas: well-demarcated magnetic resonance appearance despite frequent infiltration histology. Mayo Clinics Proceedings 70: 747-751, 1995 Darwish B, Koleda C, Lau H, et al: Juvenile pilocytic astrocytoma ‘pilomyxoid variant’ with spinal metastases. J Clin Neurosci 11: 640-642, 2004 Dierssen G, Figolls J, Trigueros F, Vazquez A: A pontine astrocytoma with radiological evidence of very extensive and dense calcification. Case report. Neurochirurgia, Stuttg). 27:190-192, 1984 Forsyth PA, Shaw EG, Scheithauer BW, et al: Supratentorial pilocytic astrocytomas. A clinicopathologic, prognostic, and flow cytometric study of 51 patients. Cancer 72:1335-1342, 1993 Giannini C, Scheithauer BW, Burger PC, et al: Cellular proliferation in pilocytic and diffuse astrocytomas. J Neuropathol Exp Neurology 58:46-53, 1999 Gilles FH, Sobel EL, Tavare CJ, et al: Age-related changes in diagnoses, histological features, and survival in children with brain tumors: 1930-1979. The Childhood Brain Tumor Consortium. Neurosurgery 37: 1056-1068, 1995 Hadjipanayis CG, Kondziolka JC, Flickinger JC, Lunsford LD: The role of stereotactic radiosurgery for low-grade astrocytomas. Neurosurgery Focus 14: e15, 2003 Ideguchi M, Nishizaki T, Harada K, et al: Pilocytic astrocytoma of the velum interpositum. Neurol Med Chir, Tokyo, 38:283- 286, 1998 Jallo G, Benardete EA: Low-Grade Astrocytoma., Topic id 190). http://www.emedicine.com/neuro/topic.190.htm. Kan P, Gottfried O, Blumenthal DT, et al: Oligodendroglioma and juvenile pilocytic astrocytoma presenting as synchronous primary brain tumors. Case report with histological and molecular differentiation of the tumors and review of the literature. J Neurosurg 100:700-705, 2004 Katsetos CD, Krishna L: Lobar pilocytic astrocytomas of the cerebral hemispheres: I. Diagnosis and nosology. Clin Neuropathol 13:295-305, 1994 Kleihues P, Cavenee WK. World Health Organization Classification of Tumors. Pathology and Genetics of Tumors of the Nervous System. IARC Press, Lyon, France, 2000. Komotar RJ, Burger PC, Carson BS et al: Pilocytic and pilomyxoid hypothalamic/chiasmatic astrocytomas. Neurosurgery 54:72-79, 2004 Komotar RJ, Carson BS, Rao C, et al: Pilomyxoid astrocytoma of the spinal cord: report of three cases. Neurosurgery 56:191, 2005 Komotar RJ, Mocco J, Jones JE, et al: Pilomyxoid astrocytoma: diagnosis, prognosis, and management. Neurosurgery Focus 18:e7, 2005 Lee YY, Van Tassel P, Bruner JM, et al: Juvenile pilocytic astrocytomas: CT and MR characteristics. Am J Roentgenol. 152:1263-1270, 1989 Lesniak MS, Klem JM, Weingart J, Carson BS Sr: Surgical outcome following resection of contrast-enhanced pediatric brainstem gliomas. Pediatr Neurosurg 39:314-322, 2003 Lones MA, Verity MA: Fatal hemorrhage in a cerebral pilocytic astrocytoma-adult type. Acta Neuropathol, Berlin, 81:688-690, 1991 Machen SK, Prayson RA: Cyclin D1 and MIB-1 immunohistochemistry in pilocytic astrocytomas: a study of 48 cases. Hum Pathol 29:1511-1516, 1998 Maruyama K, Morita A, Shibahara J, et al: Multifocal pilocytic astrocytomas with ependymal differentiation in the bilateral medial temporal lobes: case report. Neurol Med Chir, Tokyo, 45:411-414, 2005 Ohgaki H, Kleihues P: Epidemiology and etiology of gliomas. Acta Neuropathol, Berl). 109: 93-108, 2005 Philipson MR, Timothy J, Chakrobarthy A, Towns G: Pilocytic astrocytoma of a spinal nerve root. Case report. Neurosurg 97:110-112, 2002 Rickert CH, Paulus W: Epidemiology of central nervous system tumors in childhood and adolescence based on the new WHO classification. Childs Nerv Syst. 17:503-511, 2001 Rosemberg S, Fujiwara D: Epidemiology of pediatric tumors of the nervous system according to the WHO 2000 classification: a report of 1,195 cases from a single institution. Childs Nervous System 21:940-944, 2005 Tomlinson FH, Scheithauer BW, Hayostek CJ, et al: The significance of atypia and histologic malignancy in pilocytic astrocytoma of the cerebellum: a clinicopathologic and flow cytometric study. J Clin Neurol 9: 301-310, 1994 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2006 VL 12 IS 3 BP 164 EP 171 PG 8 ER PT J AU Helal, EATh Fadel, TM El-Sayed, KN AF Helal, El A Thanaa Fadel, T Mona El-Sayed, K Naglaa TI Human Papilloma Virus and p53 Expression in Bladder Cancer in Egypt: Relationship to Schistosomiasis and Clinicopathologic Factors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Egyptian patients; bladder carcinoma; schistosomiasis; p53; HPV ID Egyptian patients; bladder carcinoma; schistosomiasis; p53; HPV AB The aim of the current study was to compare the role of p53 and human papillomavirus (HPV) in schistosomiasis-related and schistosomiasis-unrelated carcinoma of the urinary bladder. To achieve this aim, we investigated 114 bladder carcinomas for p53 oncoprotein expression by immunohistochemistry and for human papillomavirus by in situ hybridization technique. The results revealed that 64 tumors (56.1%) were schistosomiasis-associated. Sixty seven (58.8%) were transitional cell carcinomas and 32 (28%) were squamous cell carcinomas. The remaining 15 tumors (13.2%) included adenocarcinomas and sarcomatoid carcinomas. In both schistosomiasis-associated and non-associated carcinomas, p53 oncoprotein expression was significantly higher in poorly differentiated tumors. However, it was significantly higher in locally more invasive tumors in the schistosomal carcinomas only. HPV types 16/18 could be detected in 1 of the 114 bladder carcinomas (0.95%), which was schistosomiasis-related squamous cell carcinoma in situ. These results suggest that p53 immunohistochemistry can be a prognostic factor in both schistosomal and nonschistosomal bladder cancer. More importantly, HPV does not seem to play a role in the pathogenesis of either type of bladder cancer in our country. C1 [Helal, El A Thanaa] Ain Shams Faculty of Medicine, Department of PathologyCairo, Egypt. [Fadel, T Mona] Ain Shams Faculty of Medicine, Department of PathologyCairo, Egypt. [El-Sayed, K Naglaa] Faculty of Science, Cairo University, Department of ZoologyCairo, Egypt. RP Helal, EATh (reprint author), Ain Shams Faculty of Medicine, Department of Pathology, Cairo, Egypt. EM thanaahelal@hotmail.com CR Khaled HM: Bladder cancer and bilharziasis today. Cancer J 6: 65-71, 1993. El-Bolkainy MN, Ghoneim MA, Mansour MA: Carcinoma of the bilharzial bladder in Egypt. Clinical and pathological features. Br J Urol 44: 561-570, 1972. El-Sebai I: Cancer of the bilharzial bladder. Urol Res 6: 233- 236, 1978. El-Merzabani MM, El-Aaser AA, Zakhary NI: A study on the etiological factors of bilharzial bladder cancer in Egypt. 1. Nitrosoamines and their precursors in urine. Eur J Cancer 15: 287-291, 1979. El-Bolkainy MN, Mokhtar NM, Ghoneim MA, Hussein MH: The impact of schistosomiasis on the pathology of bladder carcinoma. Cancer 48: 2643-2648, 1981. Mostafa MH, Sheweita SA, O’Connor PJ: Relationship between schistosomiasis and bladder cancer. Clin Microbiol Rev 12: 97-111, 1999. Wright C, Mellon K, Johnston P, et al. Expression of mutant p53, c-erb B2 and the epidermal growth factor receptor in transitional cell carcinoma of the human urinary bladder. Br J Cancer 63: 967-970, 1991. Soini Y, Turpeenniemi-Hujamen T, Kamel D, et al: p53 immunohistochemistry in transitional cell carcinoma and dysplasia of the urinary bladder correlates with disease progression. Br J Cancer 68: 1029–1035, 1993. Harney J, Murphy DM, Jones M, Mothersill C: Expression of p53 in urothelial cell cultures from tumor-bearing and tumorfree patients. Br J Cancer 71: 25-29, 1995. Glick SH, Howell LP, White RW: Relationship of p53 and bcl2 to prognosis in muscle-invasive transitional cell carcinoma of the bladder. J Urol 155: 1754-1757, 1996. Hermann GG, Horn T, Steven K: The influence of the level of lamina propria invasion and the prevalence of p53 nuclear accumulation on survival in stage T1 transitional cell bladder cancer. J Urol 159: 91-94, 1998. Niehans GA, Kratzke RA, Froberg-MK, et al: G1 check point protein and p53 abnormalities occur in most invasive transitional cell carcinomas of the urinary bladder. Br J Cancer 80: 1175-1184, 1999. Steiner G, Bierhoff E, Schmidt D, et al: p53 immunoreactivity in biopsy specimens of T1 G3 transitional cell carcinoma of the bladder, a helpful parameter in guiding the decision for or against cystectomy? Eur J Cancer 36: 610-614, 2000. Habuchi T, Takahashi R, Yamada H, et al: Influence of cigarette smoking and schistosomiasis on p53 gene mutation in urothelial cancer. Cancer Res 53: 3795-3799, 1993. Kamel D, Soini Y, Nuorva K, et al: p53 and c-erb2 expression in schistosomal urinary bladder carcinomas and schistosomal cystitis with premalignant lesions. Virchows Arch 424: 349-355, 1994. Warren W, Biggs PJ, el Baz M, et al: Mutations in the p53 gene in schistosomal bladder cancer: a study of 92 tumors from Egyptian patients and a comparison between mutational spectra from schistosomal and non-schistosomal urothelial tumors. Carcinogenesis 16: 1181-1189, 1995. Ramchurren N, Cooper K, Summerhay IC: Molecular events underlying schistosomiasis-related bladder cancer. Int J Cancer 62: 237-244, 1995. Chaudhary KS, Abel PD, Khandan-Nia N, et al: Expression of bcl-2 and p53 oncoproteins in schistosomiasis-associated transitional and squamous cell carcinoma of the urinary bladder. Br J Urol 79: 78-84, 1997. Bryant P, Skelly J, Wilson D: Demonstration of papillomavirus structural antigen in human urinary bladder neoplasia. Br J Urol 60: 405-409, 1987. Kitamura T, Yogo Y, Ueki T, et al: Presence of human papillomavirus type 16 genome in bladder carcinoma in situ of a patient with mild immunodeficiency. Cancer Res 48: 7207-7211, 1988. Bryant P, Davies P, Wilson D. Detection of human papillomavirus DNA in cancer of the urinary bladder by in situ hybridization. Br J Urol 68: 49-52, 1991. Anwar K, Naiki H, Nakakuki K, Inuzuka M: High frequency of human papillomavirus infection in carcinoma of the urinary bladder. Cancer 70: 1967-1973, 1992. Chetsange C, Malnstrom PU, Gyllensten U, et al: Low incidence of human papillomavirus type 16 DNA in bladder tumor detected by the polymerase chain reaction. Cancer 69: 1208-1211, 1992. Lopez-Beltron A, Munoz E: Transitional cell carcinoma of the bladder: low incidence of human papillomavirus DNA detected by the polymerase chain reaction and in situ hybridization. Histopathology 26: 565-569, 1995. Tenti P, Zappatore R, Roagnoli S, et al: p53 overexpression and human papillomavirus infection in transitional cell carcinoma of the urinary bladder: correlation with histological parameters. J Pathol 178: 65-70, 1996. Cooper K, Haffajee Z, Taylor L: Human papillomavirus and schistosomiasis associated bladder cancer. Mol Pathol 50: 145- 148, 1997. Guidici C, Ferrario D, Forlni N, et al: Study of human papillomavirus via chemiluminescence technique and polymerase chain reaction in transitional cell carcinoma of the bladder. Pathologica 90: 776-782, 1998. Aynaud O, Tranboloc P, Orth G: Lack of evidence for a role of human papillomavirus in transitional cell carcinoma of the bladder. J Urol 159: 86-89, 1998. Simoneau M, La Rue H, Fradet Y. Low frequency of human papillomavirus in initial papillary bladder tumors. Urol Res 27: 180-184, 1999. De-Goetani C, Ferrori G, Righi E, et al: Detection of human papillomavirus DNA in urinary bladder carcinoma by in situ hybridization. J Clin Pathol 52: 103-106, 1999. Khaled HM, Raafat A, Mokhtar N, et al: Human papillomavirus infection and overexpression of p53 protein in bilharzial bladder cancer. Tumori 87: 256-261, 2001. Mostofi FK, Sobin LH, Torloni H: Histologic typing of urinary bladder tumors. In: International Histological Classification of Tumors, Vol10). WHO, Geneva, 1973. Mostofi FK, Davis CJ, Sestrhenn IA: Histologic typing of urinary bladder tumors. In: World Health Organization International Histological Classification of Tumors 2nd ed. Heidelberg, Germany: Springer Verlag, Berlin, 1999. Fleming ID, Cooper JS, et al, eds): AJCC Manual for Staging of Cancer. 5th ed., Lippincott Raven, Philadelphia, PA, 1997. Hsu SM, Raine L: Use of avidin-biotin-peroxidase complex, ABC, in immunoperoxidase technique. A comparison between ABC and unlabelled, PAP, procedures. J Hischochem Cytochem 29: 577-580, 1981. Gabert HE, Muller W, Schneiders A, et al: The relationship of p53 expression to the prognosis of 418 patients with gastric carcinoma. Cancer 76: 720-726, 1995. Krager B: Preparation and use of biotinylated oligonucleotide probes. Focus 11: 57-58, 1989. Ross RK, Jones PA, Yu MC: Bladder cancer: Epidemiology and pathogenesis. Semin Oncol 23:536-545, 1996. Koroltchouk V, Stanley K, Stjinsward J, Mott K: Bladder cancer: approaches to prevention and control. Bull World Health Organ 65: 513-520, 1987. Plastiras D, Moutzouris G, Barbatis C, et al: Can p53 nuclear over-expression, bcl2 accumulation and PCNA status be of prognostic significance in high risk superficial and invasive bladder tumors? Eur J Surg Oncol 25: 61-65, 1999. Herr HW, Bajorin DF, Scher HI, et al: Can p53 help select patients with invasive bladder cancer for bladder preservation. J Urol 161: 20-22, 1999. Llopis J, Alcaraz A, Ribal MJ, et al: p53 expression predicts progression and poor survival in T1 bladder tumors. Eur Urol 37: 644-653, 2000. La Rue H, Simoneau M, Fradet Y. Human papillomavirus in transitional cell carcinoma of the urinary bladder. Clin Cancer Res 1: 435-440, 1995. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2006 VL 12 IS 3 BP 173 EP 178 PG 6 ER PT J AU Elek, G Lapis, K AF Elek, Gabor Lapis, Karoly TI A Path or a New Road in Laboratory Diagnostics? Biological Mass Spectrometry: Facts and Perspectives SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE serum proteome; tissue proteome; mass spectrometry; matrix; protein biomarker; SELDI; ProteinChip system; proteomics; genomics ID serum proteome; tissue proteome; mass spectrometry; matrix; protein biomarker; SELDI; ProteinChip system; proteomics; genomics AB Proteins in tissues and biofluids and their many attributes define the proteome. Proteome can be directly correlated to known diseases and histological regions allowing the diagnosis and monitoring of disease progression as well predicting the patient’s response to specific treatments. Proteomics performs large-scale, high-throughput characterization of the human proteome, among others by biological mass spectrometry. Proteinchip technology coupled with bioinformatics is able to screen any protein source for putative disease biomarkers from a small sample volume (microliter range) by surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF MS). This article discusses on a basic level both the technology and reliability of these methods. C1 [Elek, Gabor] Semmelweis University, Kutvolgyi Clinical CentreBudapest, Hungary. [Lapis, Karoly] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. RP Lapis, K (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM klapis@korb1.sote.hu CR Anderson NG, Matheson A, Anderson NL: Back to the future: the human protein index, HPI, and the agenda for post-proteomic biology. Proteomics 1:3-12, 2001. Apweiler R, Bairoch A, Wu CH, et al: UniProt: the universal protein knowledgebase. Nucleic Acid Res 32:D115-D119, 2004. Bonner RF, Emmert-Buck M, Cole K, et al: Laser capture microdissection: molecular analysis of tissue. Science 278:1481-1483, 1997. Caprioli RM, Farmer TB, Gile J: Molecular imaging of biological samples: localization of peptides and proteins using MALDI-TOF MS. Anal Chem 69:4751-4760, 1997. Chaurand P, Schwartz SA, Reyzer ML, et al: Imaging mass spectrometry: principles and potentials. Toxicol Pathol 33:92- 101, 2005. Chaurand P, Sanders ME, Jensen RA, et al: Proteomics in diagnostic pathology. Profiling and imaging proteins directly in tissue sections. Am J Pathol 165:1057-1068, 2004. Chaurand P, Schwartz SA, Bilheimer D, et al: Integrating histology and imaging mass spectroscopy. Anal Chem 76:1145- 1155, 2004. Chaurand P, Schwartz SA, Caprioli RM: Profiling and imaging proteins in tissue sections by mass spectrometry. Anal Chem 76:86A-94A, 2004. Chaurand P, Stoeckli M, Caprioli LM: Direct profiling of proteins in biological tissue sections by MALDI mass spectrometry. Anal Chem 71:5263-5270, 1999. Conrads TP, Fusaro VA, Ross S, et al: High-resolution serum proteomic features for ovarian cancer detection. Endocrine- Related Cancer 11:163-178, 2004. Diamandis EP: Proteomic patterns in biological fluids: do they represent the future of cancer diagnosis? Clin Chem 49:1272- 1275, 2003. Finkelnburg W: Einfuhrung in die Atomphysik. 9 und 10-te Auflage, Springer, Berlin-Gottingen-Heidelberg, 1964, pp 34- 43, 48. Garden RW, Sweedler JV: Heterogeneity within MALDI samples as revealed by mass spectrometric imaging. Anal Chem 72:30-36, 2000. Issaq J, Conrads TP, Prieto DA, et al: SELDI TOF MS for diagnostic proteomics. Anal Chem 75:148A-155A, 2003. Karas M, Hillenkamp F: Laser desorption ionization of proteins with molecular masses exceeding 10,000 daltons. Anal Chem 60:2299-2301, 1988. Lage H: Proteomics in cancer cell research: an analysis of therapy resistance. Pathol Res Pract 200:105-117, 2004. Lander ES, Linton LM, Birren B, et al: Initial sequencing and analysis of the human genome. Nature 409:860-921, 2001. Melle C, Kaufmann R, Hommann M, et al: Proteomic profiling in microdissected hepatocellular carcinoma using protein chip® technology. Int J Oncol 24:885-891, 2004. Palmer-Toy DE, Sarracino DA, Sgroi D, et al: Direct acquisition of matrix-assisted laser desorption/ionization time-offlight mass spectra from laser captured microdissected tissues. Clin Chem 46:1513-1516, 2000. Pandey A, Mann M: Proteomics to study genes and genomes. Nature 405:837-846, 2000. Petricoin EF, Ardekani AM, Hitt BA, et al: Use of proteomic patterns in serum to identify ovarian cancer. Lancet 359:572- 577, 2002. Petricoin EF, Liotta LA: SELDI-TOF-based serum proteomic pattern diagnostics for early detection of cancer. Curr Opin Biotechnol 15:24-30, 2004. Petricoin E 3rd, Liotta LA: The vision for a new diagnostic paradigm. Clin Chem 49:1276-1278, 2003. 24 Pieper R, Gatlin CL, Makusky AJ, et al: The human serum proteome: display of nearly 3700 chromatographically separated protein spots on two dimensional electrophoresis gels and identification of 325 distinct proteins. Proteomics 3:1345-1364, 2003. 25. Rogers MA, Clarke P, Noble J, et al: Proteomic profiling of urinary proteins in renal cancer by surface enhanced laser desorption ionization and neural network analysis: identification of key issues affecting potential clinical utility. Cancer Res 63:6971-6983, 2003. 26. Rosty C, Christa L, Kuzdzal S, et al: Identification of hepatocarcinoma- intestine-pancreas/pancreatitis associated protein I as a biomarker for pancreatic ductal adenocarcinoma by protein biochip technology. Cancer Res 62:1868-1875, 2002. 27. Rocken C, Ebert MPA, Roessner A: Proteomics in pathology, research and practice. Pathol Res Pract 200:69-82, 2004. 28. Reymond MA, Steinert R, Kahne T, et al: Expression and functional proteomics in colorectal cancer. Pathol Res Pract 200:119-127, 2004. 29. Seibert V, Wiesner A, Buschman T, et al: Surface-enhanced laser desorption ionization time-of-flight mass spectrometry, SELDI TOF-MS, and ProteinChip® technology in proteomic research. Pathol Res Pract 200:83-94, 2004. 30. Sheng Y, Jacobs JM, Comp DG, et al: Ultra-high-efficiency strong cation exchange LC/RPLC/MS/MS for high dynamic range characterization of the human plasma proteome. Anal Chem 76:1134-1144, 2004. 31. Yanagisawa K, Shyr Y, Xu BJ, et al: Proteomic patterns of tumour subsets in non-small-cell lung cancer. Lancet 362:433- 439, 2003. 32. Venter JC, Adams MD, Meyers EW, et al: The sequence of the human genome. Science 291:1304-1351, 2001. 33. Villenueva I, Philip I, Entenberg D, et al: Serum peptide profiling by magnetic particle assisted, automated sample processing and MALDI TOF mass spectrometry. Anal Chem 76:1560- 1570, 2004. 34. Wetmore BA, Merrick BA: Toxicoproteomics: proteomics applied to toxicology and pathology. Toxicol Pathol 32:619- 642, 2004. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2006 VL 12 IS 3 BP 178 EP 183 PG 6 ER PT J AU Coutts, AM Borthwick, JN Hungerford, LJ Cree, AI AF Coutts, A Michael Borthwick, J Nicola Hungerford, L John Cree, A Ian TI Post-menopausal Bleeding: a Rare Presentation of Metastatic Uveal Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE melanoma; eye; uveal; endometrium; post-menopausal bleeding; metastasis ID melanoma; eye; uveal; endometrium; post-menopausal bleeding; metastasis AB Uveal melanoma differs from cutaneous melanoma in many ways, including its pattern of metastasis, and exhibits latency with clinical evidence of metastasis sometimes appearing many years after primary diagnosis. Most patients develop metastasis within the liver, but some may present with metastasis to other sites. We report a case of uveal melanoma that presented with post-menopausal bleeding due to metastasis. Further investigation revealed widespread metastatic disease and the patient was not fit for chemotherapy. She died two months after presentation: autopsy revealed metastases in many sites, including the uterus, right ovarian fibroma, kidney, mesentery, liver, lung, thyroid, bone marrow and skin. The immediate cause of death was cardiac tamponade due to a malignant effusion secondary to cardiac metastasis. This case illustrates the widespread metastatic potential of uveal melanoma and highlights the potential for unusual presentation of metastatic disease from this eye tumor. C1 [Coutts, A Michael] Preston Hall Hospital, Maidstone and Tunbridge Wells NHS Trust, Cellular PathologyKent, UK. [Borthwick, J Nicola] University College London, Institute of Ophthalmology, Department of PathologyLondon, UK. [Hungerford, L John] Moorfields Eye Hospital, City Road, Ocular Oncology ServiceLondon, UK. [Cree, A Ian] University College London, Institute of Ophthalmology, Department of PathologyLondon, UK. RP Cree, AI (reprint author), University College London, Institute of Ophthalmology, Department of Pathology, London, UK. EM ian.cree@porthosp.nhs.uk CR Foss AJ, Cree IA, Dolin PJ, et al: Modelling uveal melanoma. Br J Ophthalmol 83:588-594, 1999 Jensen OA: Malignant melanomas of the uvea in Denmark 1943-1952. A clinical, histopathological, and prognostic study. Acta Ophthalmol, Copenh, 43(Suppl 75):1-220, 1963 Jensen OA: Malignant melanomas of the human uvea. Recent follow-up of cases in Denmark, 1943-1952. Acta Ophthalmol, Copenh, 48:1113-1128, 1970 Sato T, Babazono A, Shields JA, et al: Time to systemic metastases in patients with posterior uveal melanoma. Cancer Invest 15:98-105, 1997 Shields JA AJ, Donoso LA, et al: Hepatic metastasis and orbital recurrence of uveal melanoma after 42 years. Am J Ophthalmol 100:666-668, 1985 Jensen OA: Malignant melanomas of the human uvea: 25-year follow-up of cases in Denmark, 1943--1952. Acta Ophthalmol, Copenh, 60:161-182, 1982 Zakka KA, Foos RY, Omphroy CA, et al: Malignant melanoma. Analysis of an autopsy population. Ophthalmology 87:549- 556, 1980 Kujala E, Makitie T, Kivela T: Very long-term prognosis of patients with malignant uveal melanoma. Invest Ophthalmol Vis Sci 44:4651-4659, 2003 Lee WR, Jay JL: A complicated case of ciliary body melanoma and glaucoma. Int Ophthalmol 7:175-181, 1985 Makitie T, Kivela T: Cardiac metastasis from uveal melanoma. Chest 120:2115, 2001 Rednam KR, Jampol LM, Levine RA, et al: Uveal melanoma in association with multiple malignancies. A case report and review. Retina 1:100-106, 1981 Casey JH, Shapiro RF: Metastatic melanoma presenting as primary uterine neoplasm: a case report. Cancer 33:729-731, 1974 Takeda M, Diamond SM, DeMarco M, et al: Cytologic diagnosis of malignant melanoma metastatic to the endometrium. Acta Cytol 22:503-506, 1978 Berker B, Sertcelik A, Kaygusuz G, et al: Abnormal uterine bleeding as a presenting sign of metastasis to the endometrium in a patient with a history of cutaneous malignant melanoma. Gynecol Oncol 93:252-256, 2004 Kumar NB, Hart WR: Metastases to the uterine corpus from extragenital cancers. A clinicopathologic study of 63 cases. Cancer 50:2163-2169, 1982 Karwinski B, Svendsen E: Trends in cardiac metastasis. APMIS 97:1018-1024, 1989 Ruiz RS, El-Harazi S, Albert DM, et al: Cardiac metastasis of choroidal melanoma. Arch Ophthalmol 117:1558-1559, 1999 Chong GT, Kim RJ, Ambati SR, et al: Diagnosis of a solitary cardiac metastasis from ocular melanoma. J Thorac Cardiovasc Surg 130:1727-1728, 2005 Thombs J, Borthwick NJ, Hungerford JL, et al: Recruiting donors for autopsy based cancer research. J Med Ethics 31:360- 361, 2005 Furness PN, Nicholson ML: Obtaining explicit consent for the use of archival tissue samples: practical issues. J Med Ethics 30:561-564, 2004 McLean IW, Zimmerman LE, Evans RM: Reappraisal of Callender’s spindle a type of malignant melanoma of choroid and ciliary body. Am J Ophthalmol 86:557-564, 1978 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2006 VL 12 IS 3 BP 184 EP 187 PG 4 ER PT J AU Singh, PA Saxena, S AF Singh, P Avninder Saxena, Sunita TI Infiltrating Ductal Carcinoma of the Breast, Metastatic to Axillary Lymph Nodes Harboring Primary Tuberculous Lymphadenitis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE breast cancer; infiltrating ductal carcinoma; tuberculous lymphadenitis; metastasis ID breast cancer; infiltrating ductal carcinoma; tuberculous lymphadenitis; metastasis AB A 46-year-old female presented with lump in the left breast. Fine-needle aspiration cytology (FNAC) from breast and axillary lymph node revealed infiltrating ductal carcinoma with metastasis in axillary node. The patient underwent radical mastectomy with axillary lymph node dissection. Histopathological examination showed concomitant presence of metastatic tumor deposits and tubercular lymphadenitis in 8/18 nodes. The case is presented for its rarity and illustrates that FNAC can fail to detect mixed lesions unless multiple punctures from many sites are performed. C1 [Singh, P Avninder] Safdarjung Hospital Campus, Institute of Pathology, 213-B Sukhdev Vihar, 110025 New Delhi, India. [Saxena, Sunita] Safdarjung Hospital Campus, Institute of Pathology, 213-B Sukhdev Vihar, 110025 New Delhi, India. RP Singh, PA (reprint author), Safdarjung Hospital Campus, Institute of Pathology, 110025 New Delhi, India. EM dravninder@yahoo.co.in CR Kaplan MH, Armstrong D, Rosen P: Tuberculosis complicating neoplastic disease: a review of 201 cases. Cancer 33: 850-858, 1974 Warthin AS: The coexistence of tuberculosis and carcinoma of the mammary gland. Am J Med Sci 118: 25, 1899 Ibrahim EM, Uwaydah A, Mulhim FA, et al: Tuberculosis in patients with malignant disease. Ind J Cancer 26: 53-57, 1989 Miller RE, Solomen PF, West JP: The co-existence of carcinoma and tuberculosis of the breast and axillary lymph nodes. Am J Surg 121: 338- 340, 1971 Das DK, Mohil RS, Kashyap V, et al: Colloid carcinoma of the breast with concomitant metastasis and a tubercular lesion in the axillary lymph nodes. A case report. Acta Cytol 36: 399-403, 1992 Pandey M, Abraham EK, Chandramohan K, Rajan B: Tuberculosis and metastatic carcinoma coexistence in axillary lymph node: a case report. World J Surg Oncol 1: 1-3, 2003 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2006 VL 12 IS 3 BP 188 EP 189 PG 2 ER PT J AU Kulka, J Tokes, AM Kaposi-Novak, P Udvarhelyi, N Keller, A Schaff, Zs AF Kulka, Janina Tokes, Anna-Maria Kaposi-Novak, Pal Udvarhelyi, Nora Keller, Aniko Schaff, Zsuzsa TI Detection of HER-2/neu Gene Amplification in Breast Carcinomas Using Quantitative Real-time PCR-A Comparison with Immunohistochemical and FISH Results SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HER-2/neu protein; HER-2/neu DNA amplification; breast carcinoma; immunohistochemistry; FISH; quantitative RT-PCR; LightCycler RT-PCR system ID HER-2/neu protein; HER-2/neu DNA amplification; breast carcinoma; immunohistochemistry; FISH; quantitative RT-PCR; LightCycler RT-PCR system AB The aim of our study was to evaluate the value of quantitative real-time-PCR (qPCR) in the determination of HER-2/neu amplification status of human breast carcinomas by comparing qPCR, FISH and immunohistochemistry results from the same samples. A total of 210 breast carcinomas were examined. Ready-to-use CB11 antibody was applied to detect HER-2/neu oncoprotein expression. In 76 out of 210 cases FISH was performed, and 162 cases were investigated with qPCR. Seventy-five tumors were 2+ or 3+ positive with immunohistochemistry, while 135 samples were either completely negative or 1+. In 45 cases results from all three methods were available. Out of these, in twenty negative and sixteen positive cases both FISH and qPCR led to similar results. The mean qPCR amplification ratio in the concordant positive cases was 5.424 while in the qPCR+/FISH- group the mean ratio was 2.765. Out of 121 samples with scores of 0 or 1+ immunohistochemical result, analyzed also with qPCR, 26 showed HER-2/neu gene amplification. In these cases the mean amplification ratio was 2.53. Comparison of FISH and qPCR together with immunohistochemistry shows that qPCR is more sensitive to detect HER-2/neu gene amplification in tumors scored as 2+ with immunohistochemistry, but the diagnostic cut-off ratio should be defined above 2.7 to avoid high number of false positive cases. Amongst the immunohistochemistry score 2+ cases, 10 of 18 showed gene amplification by qPCR while 10 of 26 by FISH. In conclusion, a well calibrated HER-2/neu qPCR assay may serve as useful alternative to FISH in breast cancer patients. C1 [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Tokes, Anna-Maria] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Kaposi-Novak, Pal] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Udvarhelyi, Nora] National Institute of OncologyBudapest, Hungary. [Keller, Aniko] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. RP Kulka, J (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM kj@korb2.hu CR Allred DC, Clark GM, Molina R, et al: HER-2/neu in nodenegative breast cancer: prognostic significance of overexpression influenced by the presence of in situ carcinoma. J Clin Oncol 10: 599-605, 1992 Bankfalvi A, Boecker W, Reiner A: Comparison of automated and manual determination of HER2 status in breast cancer for diagnostic use: a comparative methodological study using Ventana BenchMark automated staining system and manual tests. Int J Oncol 25: 929-935, 2004 Bankfalvi A, Giuffre G, Ofner D, et al: Relationship between HER2 status and proliferation rate in breast cancer assessed by immunohistochemistry, fluorescence in situ hybridisation and standardised AgNOR analysis. Int J Oncol 23: 1285-1292, 2003 Bankfalvi A: HER-2 diagnostics. Magy Onkol 46: 11-15, 2002 [In Hungarian] Bargmann CI, Hung MC, Weinberg RA: The neu oncogene encodes an epidermal growth factor receptor-related protein. Nature 319: 226-230, 1986 Bartlett JM, Going JJ, Mallon EA, et al: Evaluating HER2 amplification and overexpression in breast cancer. J Pathol 195: 422-428, 2001 Beyser K, Reiser A, Gross C, et al: J. Real-time quantification of HER2/neu gene amplification by LightCycler Polymerase Chain Reaction, PCR, – a new research tool. Biochemica 2: 15-18, 2001 Beyser K, Reiser A, Gross C, et al: PCR is a powerful tool to assess HER2/neu positivity in breast cancer. Eur J Cancer 158: 419-429, 2001 Bilous M, Ades C, Armes J, et al: Predicting the HER2 status of breast cancer from basic histopathology data: an analysis of 1500 breast cancers as part of the HER2000 international study. Breast 12: 92-98, 2003 DiGiovanna MP: Clinical significance of HER-2/neu overexpression: Part I. Principles and Practice of Oncology 13: 1-10, 1999 Elkin EB, Weinstein MC, Winer EP, et al: HER-2 testing and trastuzumab therapy for metastatic breast cancer: a cost-effectiveness analysis. J Clin Oncol 22: 854-863, 2004 Elston CW, Ellis IO: Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long term follow up. Histopathology 19: 403-410, 1991 Esteva FJ, Sahin AA, Smith TL, et al: Prognostic significance of phosphorylated P38 mitogen-activated protein kinase and HER-2 expression in lymph node-positive breast carcinoma. Cancer 100: 499-506, 2004 Gjerdrum LM, Sorensen BS, Kjeldsen E, et al: Real-time quantitative PCR of microdissected paraffin-embedded breast carcinoma. J Mol Diagn 6: 42-51, 2004 Gusterson BA, Machin LG, Gullick WJ, et al: Immunohistochemical distribution of c-erbB-2 in infiltrating and in situ breast cancer. Int J Cancer 42: 842-845, 1988 Hammock L, Lewis M, Phillips C, et al: Strong HER-2/neu protein overexpression by immunohistochemistry often does not predict oncogene amplification by fluorescence in situ hybridization. Hum Pathol 34: 1043-1047, 2003 Harbeck N, Ross JS, Yurdseven S, et al: HER-2/neu gene amplification by fluorescence in situ hybridization allows risk-group assessment in node negative breast cancer. Int J Oncol 14: 663- 671, 1999 Jacobs TW, Gown AM, Yaziji H, et al: HER-2/neu protein expression in breast cancer evaluated by immunohistochemistry; a study of interlaboratory agreement. Am J Clin Pathol 113: 251-258, 2000 Jimenez RE, Wallis T, Tabasczka P, Visscher DW: Determination of Her-2/Neu status in breast carcinoma: comparative analysis of immunohistochemistry and fluorescent in situ hybridization. Mod Pathol 13: 37-45, 2000 Kakar S, Puangsuvan N, Stevens JM, et al: HER-2/neu assessment in breast cancer by immunohistochemistry and fluorescence in situ hybridization: comparison of results and correlation with survival. Mol Diagn 5: 199-207, 2000 Kauraniemi P, Hautaniemi S, Autio R, et al: Effects of Herceptin treatment on global gene expression patterns in HER2- amplified and nonamplified breast cancer cell lines. Oncogene 23: 1010-1013, 2004 Lebeau A, Deimling D, Kaltz C, et al: Her-2/neu analysis in archival tissue samples of human breast cancer: Comparison of immunohistochemistry and in situ hybridization. J Clin Oncol 19: 354-363, 2001 Lopez-Guerrero JA, Navarro S, Noguera R, et al: Histological tumor grade correlates with HER2/c-erB-2 status in invasive breast cancer: a comparative analysis between immunohistochemical, CB11 clone and Herceptest), FISH and differential PCR procedures. Arkh Patol 65: 50-55, 2003 Lu Y, Zi X, Zhao Y, Pollak M: Overexpression of ErbB2 receptor inhibits IGF-I-induced Shc-MAPK signaling pathway in breast cancer cells. Biochem Biophys Res Commun 313: 709- 715, 2004 Luftner D, Henschke P, Kafka A, et al: Discordant results obtained for different methods of HER-2/neu testing in breast cancer – a question of standardization, automation and timing. Int J Biol Markers 19: 1-13, 2004 Ma Y, Lespagnard L, Durbecq V, et al: Polysomy 17 in HER- 2/neu status elaboration in breast cancer: effect on daily practice. Clin Cancer Res 11: 4393-4399, 2005 Nistor A, Watson PH, Pettigrew N, et al: Real-time PCR complements immunohistochemistry in the determination of HER- 2/neu status in breast cancer. BMC Clin Pathol 6:2, DOI 10.1186/1472-6890-6-2, 2006 O’Malley FP, Parkes R, Latta E, et al: Comparison of HER2/neu status assessed by quantitative polymerase chain reaction and immunohistochemistry. Am J Clin Pathol 115: 504-511, 2001 Pavelic ZP, Pavelic L, Lower EE, et al: C-myc, c-erbB-2, and Ki-67 expression in normal breast tissue and in invasive and noninvasive breast carcinoma. Cancer Res 52: 2597-2606, 1992 Perez EA, Roche PC, Jenkins RB, et al: HER2 testing in patients with breast cancer: poor correlation between weak positivity by immunohistochemistry and gene amplification by fluorescence in situ hybridization. Mayo Clin Proc 77: 148-154, 2002 Press MF, Slamon DJ, Flom KJ, et al: Evaluation of HER- 2/neu gene amplification and overexpression: comparison of frequently used assay methods in a molecularly characterized cohort of breast cancer specimens. J Clin Oncol 20: 3095-3105, 2002 Riben MW, Malfetano JH, Nazeer T, et al: Identification of HER-2/neu oncogene amplification by fluorescence in situ hybridization in stage I endometrial carcinoma. Mod Pathol 10: 823-831, 1997 Ridolfi RL, Jamehdor MR, Arber JM: HER-2/neu testing in breast carcinoma: A combined immunohistochemical and fluorescence in situ hybridization approach. Mod Pathol 13: 866- 873, 2000 Ross JS, Fletcher JA: The HER2/neu, c-erbB-2, gene and protein in breast cancer. Am J Clin Pathol 112, Suppl 1): 553-567, 1999 Seshadri R, Figaira FA, Horsfall DJ, et al: Clinical significance of HER-2/neu oncogene amplification in primary breast cancer. The South Australian Breast Cancer Study Group. J Clin Oncol 11: 1936-1942, 1993 Slamon DJ, Clark GM, Wong SG, et al: Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235:177-182, 1987 Slamon DJ, Godolphin W, Jones LA, et al: Studies of the HER- 2/neu proto-oncogene in human breast and ovarian cancer. Science 244: 707-712, 1989 Somerville JE, Clarke LA, Biggart JD: C-erbB-2 overexpression and histological type of in situ and invasive breast carcinoma. J Clin Pathol 45: 16-20, 1992 Suo Z, Daehli KU, Lindboe CF, et al: Real-time PCR quantification of c-erbB-2 gene is an alternative for FISH in the clinical management of breast carcinoma patients. Int J Surg Pathol 12: 311-318, 2004 Taylor SL, Platt-Higgins A, Rudland PS, et al: Cytoplasmic staining of c-erbB-2 is not associated with the presence of detectable c-erbB-2 mRNA in breast cancer specimens. Int J Cancer 76: 459-463, 1998 Tse C, Brault D, Gligorov J, et al: [abstract]. HER-2 gene copy number quantified by real-time quantitative PCR, qPCR, and serum HER-2 concentration by enzyme-linked immunosorbent assay, ELISA): comparison with fluorescent in situ hybridization, FISH, and immunohistochemistry, IHC). 4th European Breast Cancer Conference, Hamburg, 395, 2004 Van de Vijver M, Peterse JL, Mooi WJ, et al: Neu protein overexpression in breast cancer. Association with comedo-type ductal carcinoma in situ and limited prognostic value in stage II breast cancer. N Engl J Med 319: 1239-1245, 1988 Woods Ignatoski KM, Grewal NK, Markwart S, et al: p38MAPK induces cell surface alpha4 integrin downregulation to facilitate erbB-2-mediated invasion. Neoplasia 5:128-134, 2003 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 197 EP 204 PG 8 ER PT J AU Kruslin, B Davor, T Cviko, A Cupic, H Odak, L Belicza, M AF Kruslin, Bozo Davor, Tomas Cviko, Aida Cupic, Hrvoje Odak, Ljubica Belicza, Mladen TI Periacinar Clefting and p63 Immunostaining in Prostatic Intraepithelial Neoplasia and Prostatic Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p63; periacinar clefting; prostatic adenocarcinoma; prostatic intraepithelial neoplasia (PIN) ID p63; periacinar clefting; prostatic adenocarcinoma; prostatic intraepithelial neoplasia (PIN) AB The aim of the present study was to correlate the presence and extent of retraction clefting and the expression of p63 in neoplastic glands and glands with prostatic intraepithelial neoplasia (PIN) in needle core biopsies. We analyzed needle core biopsies from 28 patients with PIN and 41 patients with adenocarcinoma. Neoplastic glands and those with PIN were analyzed on high power field (400x) and classified in three groups according to the extent of clefting. Immunohistochemical staining was performed following Microwave Streptavidin ImmunoPeroxidase (MSIP) protocol on DAKO TechMate Horizon automated immunostainer. Periacinar retraction clefting was significantly more prominent in prostatic carcinoma compared to PIN (p<0.0001) and nonneoplastic glands (p<0.0001). There was no difference between normal glands and PIN regarding clefting (p=0.8064). p63 was positive around the whole circumference in 12 out of 28 cases with PIN, and discontinuously positive in remaining 16 PIN cases suggesting initial disruption of the basal cell layer. p63 immunostaining was also positive in all nonneoplastic glands, and negative in all carcinomas. We conclude that retraction clefting was associated with cancer and lack of basal cells, but not with PIN. The relationship between clefting and p63 immunostaining in prostatic cancer should be further analyzed. C1 [Kruslin, Bozo] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska 29., 10000 Zagreb, Croatia. [Davor, Tomas] Brigham and Women’s Hospital, Department of PathologyBoston, USA. [Cviko, Aida] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska 29., 10000 Zagreb, Croatia. [Cupic, Hrvoje] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska 29., 10000 Zagreb, Croatia. [Odak, Ljubica] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska 29., 10000 Zagreb, Croatia. [Belicza, Mladen] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska 29., 10000 Zagreb, Croatia. RP Kruslin, B (reprint author), University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, 10000 Zagreb, Croatia. EM bkruslin@kbsm.hr CR Bostwick DG, Dundore PA: Biopsy pathology of the prostate. Chapmann&Hall Medical, London, 1997. Bostwick DG, Qian J: High-grade prostatic intraepithelial neoplasia. Mod Pathol 17: 360-379, 2004. Davis LD, Zhang W, Merseburger A, et al: p63 expression profile in normal and malignant prostate epithelial cells. Anticancer Res 22: 3819-3825, 2002. De Marzo AM, Meecker AK, Zha S, et al: Human cancer precursors and pathobiology. Urology 62(Suppl 1): 55-62, 2003. Epstein JI: Diagnosis and reporting of limited adenocarcinoma of the prostate on needle biopsy. Mod Pathol 17: 307-315, 2004. Epstein JI: Diagnosis and reporting of limited adenocarcinoma of the prostate on needle biopsy. Pathol International 54(Suppl 1): S609-S612, 2004. Epstein JI: The diagnosis and reporting of adenocarcinoma of the prostate in core needle biopsy specimens. Cancer 78: 350- 356, 1996. Halpert B, Schmalhorst WR: Carcinoma of the prostate in patients 70 to 79 years old. Cancer 19: 695-698, 1966. Halpert B, Sheehan EE, Schmalhorst WR, Scott R Jr: Carcinoma of the prostate: a survey of 5000 autopsies. Cancer 16: 736- 742, 1963. Kru lin B, Novosel I, Reljiae A, et al: Periacinar cleft-like spaces in prostatic needle core biopsies. Acta Clin Croat 41: 175, 2002. Kru lin B, Tomas D, Rogatsch H, et al: Periacinar retraction clefting in the prostate needle core biopsies: an important diagnostic criterion or a simple artifact? Virchows Arch 443: 524-527, 2003. Kru lin B, Tomas D, Rogatsch H, et al: Correlation of periacinar retraction clefting in needle core biopsies and corresponding prostatectomy specimens of patients with prostatic adenocarcinoma. Int J Surg Pathol 13: 67-72, 2005. Leroy X, Aubert S, Villers A, et al: Minimal focus of adenocarcinoma on prostate biopsy: clinicopathologic correlations. J Clin Pathol 56: 23-32, 2003. McNeal JE, Bostwick DG: Spread of adenocarcinoma within prostatic ducts and acini. Morphologic and clinical correlations. Am J Surg Pathol 20: 802-814, 1996. Oliai BR, Kahane H, Epstein JI: Can basal cells be seen in adenocarcinoma of the prostate?: an immunohistochemical study using high molecular weight cytokeratin, clone 34 beta E12, antibody. Am J Surg Pathol 26: 1151-1160, 2002. Parsa R, Yang A, McKeon F, Green H: Association of p63 with proliferative potential in normal and neoplastic human keratinocytes. J Invest Dermatol 113: 1099-1105, 1999. Rogatsch H, Mairinger T, Horninger W, et al: Optimized preembedding method improves the histologic yield of prostatic core needle biopsies. Prostate 42: 124-129, 2000. Rogatsch H, Moser P, Volgger H, et al: Diagnostic effect of an improved preembedding method of prostate needle biopsy specimens. Hum Pathol 31: 1102-1107, 2000. Sakr WA: Prostatic intraepithelial neoplasia. Pathol International 54(Suppl 1):S612-S615, 2004. Signoretti S, Waltregny W, Dilks J, et al: p63 is a prostate basal cell marker and is required for prostate development. Am J Pathol 157: 1769-1775, 2000. Tacha DE, Miller RT: Use of p63/P504S monoclonal antibody cocktail in immunohistochemical staining of prostate tissue. Appl Immunohistochem Mol Morphol 12: 75-78, 2004. Tomas D, Kru lin B: The potential value of, myo)fibroblastic stromal reaction in the diagnosis of prostatic adenocarcinoma. Prostate 61: 324-331, 2004. Varma M, Jasani B: Diagnostic utility of immunohistochemistry in morphologically difficult prostate cancer: review of current literature. Histopathology 47: 1-16, 2005. Varma M, Lee MW, Tamboli P, et al: Morphologic criteria for the diagnosis of prostatic adenocarcinoma in needle biopsy specimens. Arch Pathol Lab Med 126: 554-561, 2002. Weinstein MA, Signoretti S, Loda M: Diagnostic utility of immunohistochemical staining for p63, a sensitive marker of prostatic basal cells. Mod Pathol 15: 1302-1308, 2002. Zhou M, Shah R, Shen R, Rubin MA: Basal cell cocktail, 34betaE12 + p63, improves the detection of prostate basal cells. Am J Surg Pathol 27: 365-371, 2003. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 205 EP 209 PG 5 ER PT J AU Sahin, IF Yilmaz, Z Yagmurdur, CM Atac, BF Ozdemir, HB Karakayali, H Demirhan, B Haberal, M AF Sahin, Iffet Feride Yilmaz, Zerrin Yagmurdur, Can Mahmut Atac, Belgin Fatma Ozdemir, Handan Binnaz Karakayali, Hamdi Demirhan, Beyhan Haberal, Mehmet TI Clinical Findings and HER-2/neu Gene Amplification Status of Breast Carcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HER-2/neu; breast carcinoma; FISH ID HER-2/neu; breast carcinoma; FISH AB The study group was derived from the archival materials of 48 invasive intraductal breast cancer patients who had undergone partial mastectomy/ axillary dissection. All patients included in the study had clinically T1-2N0M0 invasive ductal carcinoma. To detect HER-2/neu status, fluorescent in situ hybridization was performed using a HER-2/neu locus-specific probe. Signals were counted and patients were classified in three groups according to signal ratios: signal ratio <2, group 1 (n=31); signal ratio 2-4, group 2 (n=11); signal ratio >4, group 3 (n=6). Ratios of axillary metastatic lymph nodes to dissected total lymph nodes were 17%, 23% and 83% in groups 1, 2 and 3 respectively (P=0.003). The number of metastatic axillary lymph nodes, and the ratio of microscopic metastatic lymph nodes were highest in group 3 (P=0.001 and P=0.008, respectively). No significant difference was observed between groups for distant metastasis in a 5-year follow-up period. Signal ratios decreased with estrogen receptor expression (P=0.03). Histopathologically, an irregular growth pattern of the tumor was observed in 100% of the patients in group 3, and in 54% and 60% in groups 1 and 2, respectively (P=0.04). Lymphovascular invasion of the tumor was significantly higher in group 3 compared to the other two groups (P=0.01). The extensive intraductal component ratio was the highest in group 3 (P=0.04). The appearance of desmoplastic reaction and lymphocyte infiltration did not show significant difference between the groups. Our results show that HER-2/neu signal ratio increases with lymphovascular invasion, an extensive intraductal component, irregular growth pattern and axillary metastasis in clinically T1-2N0M0 invasive ductal carcinoma of the breast. C1 [Sahin, Iffet Feride] Baskent University Faculty of Medicine, Department of Medical Biology and Genetics, 06570 Ankara, Turkey. [Yilmaz, Zerrin] Baskent University Faculty of Medicine, Department of Medical Biology and Genetics, 06570 Ankara, Turkey. [Yagmurdur, Can Mahmut] Baskent University Faculty of Medicine, Department of SurgeryAnkara, Turkey. [Atac, Belgin Fatma] Baskent University Faculty of Medicine, Department of Medical Biology and Genetics, 06570 Ankara, Turkey. [Ozdemir, Handan Binnaz] Baskent University Faculty of Medicine, Department of PathologyAnkara, Turkey. [Karakayali, Hamdi] Baskent University Faculty of Medicine, Department of SurgeryAnkara, Turkey. [Demirhan, Beyhan] Baskent University Faculty of Medicine, Department of PathologyAnkara, Turkey. [Haberal, Mehmet] Baskent University Faculty of Medicine, Department of SurgeryAnkara, Turkey. RP Sahin, IF (reprint author), Baskent University Faculty of Medicine, Department of Medical Biology and Genetics, 06570 Ankara, Turkey. EM feridesahin@hotmail.com CR Dandachi N, Dietze O, Hauser-Kronberger C: Chromogenic in situ hybridization: a novel approach to a practical and sensitive method for the detection of HER2 oncogene in archival human breast carcinoma. Lab Invest 82: 1007-1014, 2002. Mariani SM: Conference report - breast cancer markers: What Next? Medscape General Medicine 5: 2003. Dowsett M, Bartlett J, Ellis IO et al: Correlation between immunohistochemistry, HercepTest, for HER-2 in 426 breast carcinomas from 37 centres. J Pathol 199: 418-423, 2003. Sauer T, Wiedswang G, Boudjema G et al: Assessment of HER- 2/neu overexpression and/or gene amplification in breast carcinomas: should in situ hybridization be the method of choice? APMIS 111: 444-450, 2003. Varshney D, Zhou YY, Geller SA et al: Determination of HER- 2 status and chromosome 17 polisomy in breast carcinomas comparing HercepTest and PathVysion FISH assay. Am J Clin Pathol 121: 70-77, 2004. Simeone A: Detection of mRNA in tissue section with radiolabelled riboprobes. In: In Situ Hybridization 2nd ed, Ed: DG Wilkinson), The Practical Approach Series, Oxford University Press, 1998, pp 69-86. Yilmaz Z, Sahin FI, Atalay B, et al: Chromosome 1p36 and 22qter deletions in paraffin block sections of intracranial meningiomas. Pathol Oncol Res 11: 224-228, 2005. Tibiletti MG: Specificity of interphase fluorescence in situ hybridization for detection of chromosome aberrations in tumor pathology. Cancer Genet Cytogenet 155: 143-148, 2004. Sumita G, Zoran G, Amin M et al: FISH for HER-2/neu in breast cancer: Standardization makes the difference! Ind J Cancer 41: 152-158, 2004. Bozzetti C, Nizzoli R, Guazzi A et al: HER-2/neu amplification detected by fluorescence in situ hybridization in fine needle aspirates from primary breast cancer. Ann Oncol 13: 1398- 1403, 2002. Bartlett J, Mallon E, Cooke T: The clinical evaluation of HER- 2 status: which test to use? J Pathol 199: 411-417, 2003. Pauletti G, Dandekar S, RongHM et al: Assessment of methods for tissue-based detection of the HER-2/neu alteration in human breast cancer: A direct comparison of fluorescence in situ hybridization and immunohistochemistry. J Clin Oncol 18: 3651-3664, 2000. Ross JS, Fletcher JA, Linette GP et al: The HER-2/neu gene and protein in breast cancer 2003: Biomarker and target of therapy. The Oncologist 8: 307-325, 2003. Climent MA, Segui MA, Peiro G et al: Prognostic value of HER-2/neu and p53 expression in node-positive breast cancer. HER-2/neu effect on adjuvant tamoxifen treatment. The Breast 10: 67-77, 2001. Dowsett M, Harper-Wynne C, Boeddinghaus I et al: HER-2 amplification impedes the antiproliferative effects of hormone therapy in estrogen receptor-positive primary breast cancer. Cancer Res 61: 8452-8458, 2001. Elledge RM, Green S, Ciocca D et al: HER-2 expression and response to tamoxifen in estrogen receptor-positive breast cancer: a Southwest Oncology Group Study. Clin Cancer Res 4: 7- 12, 1998. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 211 EP 215 PG 5 ER PT J AU Cserni, G Orosz, Zs Kulka, J Sapi, Z Kalman, E Bori, R AF Cserni, Gabor Orosz, Zsolt Kulka, Janina Sapi, Zoltan Kalman, Endre Bori, Rita TI Divergences in Diagnosing Nodular Breast Lesions of Noncarcinomatous Nature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE fibroepithelial breast tumors; nodular lesions; phyllodes tumor; phylloid tumor; fibroadenoma ID fibroepithelial breast tumors; nodular lesions; phyllodes tumor; phylloid tumor; fibroadenoma AB Nodular breast lesions of noncarcinomatous origin are often of fibroepithelial origin. They may cause classification problems when they are hypocellular or hypercellular; the latter setting may also raise the differential diagnosis of phyllodes tumors. Thirty equivocal nodular breast lesions were collected and one hematoxylin and eosin slide from each was assessed by six pathologists with special interest in breast pathology. The overall reproducibility of classifying these lesions into categories of fibroadenoma, phyllodes tumor or anything else was moderate (kappa value: 0.48). The lack of a uniform nomenclature was not felt disturbing for hypocellular lesions, but the discordant diagnosis of tumors resembling or representing phyllodes tumors was acknowledged to require intervention, such as more obvious implication of guidelines and quality assurance programs aiming at assessing diagnoses and prognostic parameters. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., H-6000 Kecskemet, Hungary. [Orosz, Zsolt] National Institute of Oncology, Department of Diagnostic PathologyBudapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Sapi, Zoltan] St John's Hospital, Department of PathologyBudapest, Hungary. [Kalman, Endre] University of Pecs, Department of PathologyPecs, Hungary. [Bori, Rita] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., H-6000 Kecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.hu CR Azzopardi J: Problems in breast pathology. Saunders, London- Philadelphia-Toronto, 1979 Bellocq JP, Magro G: Fibroepithelial tumors. In: WHO Classification of Tumors. Pathology and Genetics of Tumors of the Breast and Female Genital Organs., Eds: Tavassoli FA, Devilee P), IARC Press, Lyon, 2003, pp 99-103 Berean K, Tron VA, Churg A, et al: Mammary fibroadenoma with multinucleated stromal giant cells. Am J Surg Pathol 10: 823-827, 1986 Chen WH, Cheng SP, Tzen CY, et al: Surgical treatment of phyllodes tumors of the breast: retrospective review of 172 cases. J Surg Oncol 91: 185-194, 2005 Cserni G: Adaptation of mitotic scoring of breast carcinomas. Hum Pathol 27:617-618, 1996 Daya D, Trus T, D’Souza TJ, et al: Hamartoma of the breast, an underrecognized breast lesion. A clinicopathologic and radiographic study of 25 cases. Am J Clin Pathol 103: 685-689, 1995 Ellis IO, Pinder SE, Bobrow L, et al: Pathology reporting of breast disease. NHS Publication No 58. NHS Cancer Screening Programmes and the Royal College of Pathologists, Sheffield, 2005 Elston CW, Ellis IO: Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: Experience from a larger study with long term follow up. Histopathology 19:403-410, 1991 Fisher CJ, Hanby AM, Robinson L, et al: Mammary hamartoma – a review of 35 cases. Histopathology 20: 99-106, 1992 Fleiss JL: The measurement of interrater agreement. In: Statistical Methods for Rates and Proportions., Ed: Fleiss JL), 2nd ed., John Wiley and Sons, New York, 1981, pp 212-236 Kuijper A, Buerger H, Simon R, et al: Analysis of the progression of fibroepithelial tumors of the breast by PCR-based clonality assay. J Pathol 197: 575-581, 2002 Landis JR, Koch GG: The measurement of observer agreement for categorical data. Biometrics 33: 159-174, 1977 Moffat CJ, Pinder SE, Dixon AR, et al: Phyllodes tumors of the breast: a clinicopathological review of thirty-two cases. Histopathology 27: 205-218, 1995. National Mammographic Screening Working Party: Guidelines for the diagnosis and treatment of early breast cancer and mammographic breast screening. Orszagos Tisztifoorvosi Hivatal, Budapest, 2005 [In Hungarian] Perry N, Broeders M, et al, eds): European guidelines for breast screening and diagnosis. European Communities, Luxemburg, 2006 Putti TC, Pinder SE, Elston CW, et al: Breast pathology practice: most common problems in a consultation service. Histopathology 47: 445- 457, 2005 Rajan PB, Cranor ML, Rosen PP: Cystosarcoma phyllodes in adolescent girls and young women: a study of 45 patients. Am J Surg Pathol 22: 64-69, 1998 Rosen PP: Rosen’s Breast Pathology, 2nd edition. Lippincott, Williams and Wilkins, Philadelphia, 2001 Rosen PP, Oberman HA: Tumors of the Mammary Gland. Atlas of Tumor Pathology, 3rd series. Armed Forces Institute of Pathology, Washington DC, 1993 Tavassoli FA: Pathology of the Breast, 2nd edition. McGraw-Hill, New York, 1999 Trojani M: Atlas en couleur d’histopathologie mammaire. Maloine, Paris, 1988 Tse GM, Law BK, Ma TK, et al: Hamartoma of the breast: a clinicopathological review. J Clin Pathol 55: 951-954, 2002 Vuitch MF, Rosen PP, Erlandson RA: Pseudoangiomatous hyperplasia of mammary stroma. Hum Pathol 17: 185-191, 1986 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 216 EP 221 PG 6 ER PT J AU Kiani, J Khan, A Khawar, H Shuaib, F Pervez, Sh AF Kiani, Jawad Khan, Afrasyab Khawar, Hina Shuaib, Fawad Pervez, Shahid TI Estrogen Receptor Alpha-Negative and Progesterone Receptor-Positive Breast Cancer: Lab Error or Real Entity? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE estrogen receptor (ER) alpha; progesterone receptor (PR); breast carcinoma ID estrogen receptor (ER) alpha; progesterone receptor (PR); breast carcinoma AB Aretrospective study comparing the estrogen receptor (ER) alpha subtype and progesterone receptor (PR) profile of breast carcinomas amongst 1625 cases over 2.5 years was carried out. Strictly speaking it is generally believed that breast carcinomas can biochemically express PR only if they are ER-positive. However, a few ERalpha-PR+ cases do exist paradoxically. This class of tumors was the focus of our study in which we looked at the possible reasons for such an immunophenotype and compared it with a group of ERalpha+PR+ breast carcinomas. An internationally recognized immunohistochemical method employing monoclonal antibodies against estrogen and progesterone receptors was used. Correlations with established risk factors i.e. menopausal status, grade, tumor size and lymph node status were analyzed for our study group (ERalpha-PR+) and compared with a control (ERalpha+PR+). Out of the total 1625 cases, 29.91% (486) were ERalpha+PR+, 5.11% (83) were ERalpha+PR-, 56.86% (924) were ERalpha-PR- and 8.12% (132) were ERalpha-PR+. Patients’ age was significantly lower in the ERalpha-PR+ group (P=0.002). Statistical analysis of the grading between the two study groups revealed no significant difference (P=0.091), although the ERalpha-PR+ group contained significantly more poorly differentiated tumors than the ERalpha+PR+ one (P=0.032). Tumor size was also significantly larger in the ERalpha-PR+ than in the ERalpha+PR+ group (P=0.046). The frequency of lymph node metastases was independent of receptor profile. In conclusion, our study group does exhibit characteristics which are suggestive of a distinct breast cancer phenotype (ERalpha-PR+) with a different etiology and prognosis. C1 [Kiani, Jawad] The Aga Khan University, Department of Pathology and Microbiology, Stadium Road, 74800 Karachi, Pakistan. [Khan, Afrasyab] The Aga Khan University, Department of Pathology and Microbiology, Stadium Road, 74800 Karachi, Pakistan. [Khawar, Hina] The Aga Khan University, Department of Pathology and Microbiology, Stadium Road, 74800 Karachi, Pakistan. [Shuaib, Fawad] The Aga Khan University, Department of Pathology and Microbiology, Stadium Road, 74800 Karachi, Pakistan. [Pervez, Shahid] The Aga Khan University, Department of Pathology and Microbiology, Stadium Road, 74800 Karachi, Pakistan. RP Pervez, Sh (reprint author), The Aga Khan University, Department of Pathology and Microbiology, 74800 Karachi, Pakistan. EM shahid.pervez@aku.edu CR Ballare C, Bravo, AI, Sorin I, et al: The expression of progesterone receptors coincides with an arrest of DNA synthesis in human breast cancer. Cancer 67: 1352-1358, 1991 Berger U, McLelland RA, Wilson P, et al: Immunohistochemical determination of estrogen receptor, progesterone receptor and 1, 25-dihydroxvitamin D3 receptor in breast cancer and relationship to prognosis. Cancer Res 51: 239-244, 1991 Berkenstam A, Glaumann H, Martin M, et al: Hormonal regulation of estrogen receptor messenger ribonucleic acid in T47Dco and MCF-7 breast cancer cells. Mol Endocrinol 3: 22- 28, 1989 Bloom HJG, Richardson WW: Histological grading and prognosis in breast cancer. Br J Cancer 11:359-377, 1957 deSombre, ER: Steroid receptors in breast cancer. In: The Breast., Eds: McDivitt RW, Oberman HA, Ozello L and Kaufman N), Williams and Wilkins, Baltimore, 1984, pp149-174 Edery M, Goussard J, Dehennin L, et al: Endogenous oestradiol- 17β concentration in breast tumors determined by mass fragmentography and by radioimmunoassay: relationship to receptor content. Eur J Cancer 17: 115-120, 1981 Fishman J, Nisselbaum JS, Menendez-Botet CJ, Schwartz MK: Estrogen and estradiol content in human breast tumors: relationship to estradiol receptors. J Steroid Biochem 8: 893-896, 1977 Greene GL, Press MF: Steroid receptor structure, including monoclonal antibodies and new methods of determination): structure and dynamics of the estrogen receptor. J Steroid Biochem 24: 1-7, 1986 Greene GM, Fitch FW, Jensen EV: Monoclonal antibodies to trophilin: probes for the study of estrogen receptors. Proc Natl Acad Sci USA 77: 157-161, 1980 Horwitz KB, McGuire WL: Estrogen control of progesterone receptor in human breast cancer. Correlation with nuclear processing of estrogen receptor. J Biol Chem 253: 2223-2228, 1978 Horwitz KB: Cellular heterogeneity and mutant oestrogen receptors in hormone resistant breast cancer. Cancer Surv 14: 41-54, 1992 Howat JMT, Harris M, Swindell R, Barnes DM: The effect of oestrogen and progesterone receptors on recurrence and survival in patients with carcinoma of the breast. Br J Cancer 51: 263-270, 1985 Kiang DT, Kollander R: Breast cancers negative for estrogen receptor but positive for progesterone receptor, a true entity? J Clin Oncol 5: 662-666, 1987 Leclercq G, Heuson JC, Deboel MC, et al: Estrogen and progesterone receptors in human breast cancer. In: Progesterone Receptors in Normal and Neoplastic Tissues., Eds: McGuire WL, Raynaud J-P, Baulieu E-E), Raven, New York, 1977, pp 141-153 Leygue E, Dotzlaw H, Watson PH, Murphy LC: Altered estrogen receptor alpha and beta messenger RNA expression during human breast tumorigenesis. Cancer Res 58: 3197-3201, 1998 McGuire WL: Hormone receptors. Their role in predicting prognosis and response to endocrine therapy. Semin Oncol 5: 428-433, 1979 McGuire WL, Horwitz KB, Pearson DH, Segaloff A: Current status of estrogen and progesterone receptors in breast cancer. Cancer 39: 2934-2947, 1977 McGuire WL: Steroid receptors in human breast cancer. Cancer Res 38: 4289-4291, 1978 Nuclear Receptors Nomenclature Committee: A unified nomenclature system for the nuclear receptor superfamily. Cell 97:161-163, 1999 Osborne CK, Yochmowitz MG, Knight WA, McGuire WL: The value of estrogen and progesterone receptors in the treatment of breast cancer. Cancer 46: 2884-2888, 1980 Osborne CK: Receptors. In: Breast Diseases., Eds: Harris JR, Hellman S, Henderson IC, Kinne DW), 2nd ed, JB Lippincott Co, Philadelphia, PA, 1991, pp 301-325 Panko WB, MacLeod RM: Uncharged nuclear receptors for estrogen in breast cancers. Cancer Res 38: 1948-1951, 1978 Perrot-Applanat M, Logeat F, Croyer-Picard MT, Milgrom E: Immunocytochemical study of mammalian progesterone receptor using monoclonal antibodies. Endocrinology 116: 147- 1483, 1985 Pertschuk LP. Kim DS, Kamran Nayer MA, et al: Immunocytochemical estrogen and progestin receptor assays in breast cancer with monoclonal antibodies. Cancer 66: 1663-1670, 1990 Pichon MF, Milgrom E: Oestrogen receptor negative-progesterone receptor positive phenotype in 1,211 breast tumors. Br J Cancer 65: 895-897, 1992 Reiner A, Neumeister B, Spona J, et al: Immunocytochemical localization of estrogen and progestin and prognosis in human primary breast cancer. Cancer Res 50: 7057-7061, 1990 Saez S, Martin PM, Chouvet CD: Estradiol and progesterone receptors in human breast adenocarcinoma in relation to plasma estrogen and progesterone levels. Cancer Res 38: 3468- 3473, 1978 Sarrif AM, Durant JR: Evidence that estrogen-receptor-negative progesterone-receptor-positive breast and ovarian carcinomas contain estrogen receptor. Cancer 48: 1215-1220, 1982 Savouret J-F, Fridlanski F, Atger M, et al: Origin of the high constitutive level of progesterone receptor in T47-D breast cancer cells. Mol Cell Endocrinol 75: 157-162, 1991 Shaaban AM, O’Neill PA, Davies MP, et al: Declining estrogen receptor beta expression defines malignant progression of human breast neoplasia. Am J Surg Pathol 27: 1502-1512, 2003 Shapiro CM, Schiffeling D, Bitran JD, et al: Prognostic value of estrogen receptor value in pathologic stage 1 and 2 adenocarcinoma of the breast. J Surg Oncol 19: 119-121, 1982 Shigehira S, Makiko H, Masakazu T: Clinical significance of estrogen receptor β in breast cancer. Cancer Chemother Pharmacol 56: s21-s26, 2005 Speirs V, Skliris GP, Burdall SE, Carder PJ: Distinct expression patterns of ER alpha and ER beta in normal human mammary gland. J Clin Pathol 55: 371-374, 2002 Thorpe SM: Immunological quantitation of nuclear receptors in human breast cancers: relation to cytosolic estrogen and progesterone receptors. Cancer Res 47: 1830-1835, 1987 Thorsen T: Occupied and unoccupied oestradiol receptor in human breast tumor cytosol. J Steroid Biochem. 24: 1-7, 1986 Toft DO, O’Malley BW: Target tissue receptors for progesterone: The influence of estrogen treatment. Endocrinology 90: 1041-1045, 1972 Vihko R, Janne O, Kontula K, Syrjala P: Female sex steroid receptor status in primary and metastatic breast carcinoma and its relationship to serum steroid and peptide hormone levels. Int J Cancer 26: 13-21, 1980 Watson CS, Medina D, Clark JH: Characterization and estrogen stimulation of cytoplasmic progesterone receptor in the ovarian- dependent MXT-3590 mammary tumour line. Cancer Res 39: 4098-4104, 1979 Zava DT, Chamness GC, Horwitz KB, McGuire WL: Biologically active estrogen receptor in the absence of estrogen. Science 196: 663-664, 1977 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 223 EP 227 PG 5 ER PT J AU Kamory, E Tanyi, M Kolacsek, O Olasz, J Toth, L Damjanovich, L Csuka, O AF Kamory, Eniko Tanyi, Miklos Kolacsek, Orsolya Olasz, Judit Toth, Laszlo Damjanovich, Laszlo Csuka, Orsolya TI Two Germline Alterations in Mismatch Repair Genes Found in a HNPCC Patient with Poor Family History SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bethesda criteria; hereditary nonpolyposis colorectal cancer; hMLH1; hMSH2; microsatellite instability; immunohistochemistry ID Bethesda criteria; hereditary nonpolyposis colorectal cancer; hMLH1; hMSH2; microsatellite instability; immunohistochemistry AB The Bethesda guidelines may offer more useful criteria in patients’ selection for germline mismatch repair gene mutation analysis than guidelines merely based on family background. An early onset double primary colorectal cancer patient with poor family history with MSI-H status was investigated for MLH1 promoter methylation, expression of the MLH1 and MSH2 gene by immunohistochemistry and mutations in the MLH1 and MSH2 genes. The index patient carried two germline alterations, the p.Val716Met in MLH1 and the c.2210+1G>C in MSH2 genes, and both tumors failed to express MLH1 and MSH2 proteins. After subsequent analysis of the whole family of the index patient, the p.Val716Met variant can be defined as a rare polymorphism with the possible contribution of pathogenicity to tumor formation and c.2210+1G>C as a true pathogenic mutation causing an out-of-frame deletion of exon 13. C1 [Kamory, Eniko] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Tanyi, Miklos] University of Debrecen, Medical and Health Science Centre, 1st Department of SurgeryDebrecen, Hungary. [Kolacsek, Orsolya] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Olasz, Judit] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Toth, Laszlo] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Damjanovich, Laszlo] University of Debrecen, Medical and Health Science Centre, 1st Department of SurgeryDebrecen, Hungary. [Csuka, Orsolya] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. RP Kamory, E (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, H-1122 Budapest, Hungary. EM eniko.kamory@gmail.com CR Cunningham JM, Christensen ER, Tester DJ, et al: Hypermethylation of the hMLH1 promoter in colon cancer with microsatellite instability. Cancer Res 58: 3455-3460, 1998. Wang Q, Lasset C, Desseigne F, et al: Prevalence of germline mutations of hMLH1, hMLS2, hPMS1, hPMS2, and hMSH6 genes in French kindreds with nonpolyposis colorectal cancer. Hum Genet 105: 79-85, 1999. Syngal S, Fox EA, Eng C, et al: Sensitivity and specificity of clinical criteria for hereditary non-polyposis colorectal cancer associated mutations in MSH2 and MLH1. J Med Genet 37: 641-645, 2000. Rodriguez-Bigas MA, Boland CR, Hamilton SR, et al: A National Institute workshop on hereditary nonpolyposis colorectal cancer syndrome: meeting highlights and Bethesda guidelines. J Natl Cancer Inst 89: 1758-1762, 1997. Raedle J, Trojan J, Brieger A, et al: Bethesda guidelines: Relation to microsatellite instability and MLH1 promoter methylation in patients with colorectal cancer. Ann Intern Med 135: 566-576, 2001. Montera M, Resta N, Simone C, et al: Mutational germline analysis of hMSh2 and hMLH1 genes in early onset colorectal cancer patients. J Med Genet 37: E7, 2000. Boland CR, Thibodeau SN, Hamilton SR, et al: A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58: 5248-5257, 1998. Kamory E, Kolacsek O, Otto S, et al: hMLH1 and hMSH2 somatic inactivation mechanisms in sporadic colorectal cancer patients. Pathol Oncol Res 9: 236-241, 2003. Eads CA, Lord RV, Wickramasinghe K, et al: Epigenetic patterns in the progression of esophageal adenocarcinoma. Cancer Res 61: 3410-3418, 2001. Beck NE, Tomlinson IPM, Homfray T, et al: Use of SSCP analysis to identify germline mutations in HNPCC families fulfilling the Amsterdam criteria. Hum Genet 99: 219-224, 1997. Yanagisawa Y, Akiyama Y, Iida S, et al: Methylation of the hMLH1 promoter in familial gastric cancer with microsatellite instability. Int J Cancer 85: 50-53, 2000. Wahlberg SS, Schmeits J, Thomas G, et al: Evaluation of microsatellite instability and immunohistochemistry for the prediction of germ-line MSH2 and MLH1 mutations in hereditary nonpolyposis colon cancer families. Cancer Res 62: 3485- 3492, 2002. Hendriks Y, Franken P, Dierssen JW, et al: Conventional and tissue microarray immunohistochemical expression analysis of mismatch repair in hereditary colorectal tumors. Am J Pathol 162: 469-477, 2003. Raut CP, Pawlik TM, Rodriguez-Bigas MA: Clinicopathologic features in colorectal cancer patients with microsatellite instability. Mut Res. 568:275-282, 2004. Hutter P, Couturier A, Membrez V, et al: Excess of hMLH1 germline mutations in Swiss families with hereditary non-polyposis colorectal cancer. Int J Cancer 78: 80-84, 1998. Cederquist K, Emanuelsson M, Goransson I, et al: Mutation analysis of the MLH1, MSH2 and MSH6 genes in patients with double primary cancers of the colorectum and the endometrium: a population-based study in northern Sweden. Int J Cancer 109: 370-376, 2004. http://prodes.toulouse.inra.fr Kurzawski G, Suchy J, Kladny J, et al: Germline MSH2 and MLH1 mutational spectrum in HNPCC families from Poland and the Baltic States. J Med Genet 39: E65, 2002. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 228 EP 233 PG 6 ER PT J AU Sule, N Teszas, A Kalman, E Szigeti, R Miseta, A Kellermayer, R AF Sule, Norbert Teszas, Alexandra Kalman, Endre Szigeti, Reka Miseta, Attila Kellermayer, Richard TI Lithium Suppresses Epidermal SERCA2 and PMR1 Levels in the Rat SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hailey-Hailey disease; Darier disease; lithium; rat; SERCA2; PMR1 ID Hailey-Hailey disease; Darier disease; lithium; rat; SERCA2; PMR1 AB Autosomal dominant mutations in the genes encoding the calcium ATPases SERCA2 and PMRI/SPCA1 cause the genodermatoses Darier disease (DD) and Hailey-Hailey disease (HHD), respectively. Recent observations indicated that the level of the pathogenic proteins greatly decreases in the affected areas of the epidermis in these disorders. Here we addressed how lithium, a recognized exacerbating factor in Darier disease, affects the epidermal expression of SERCA2 and PMR1/SPCA1 in the rat as a model. Standard histologic and immunohistochemical methods were utilized in 3 lithium-treated and 3 control animals. A significant suppression of epidermal SERCA2 and PMR1 levels were observed as a result of lithium therapy in addition to marked qualitative and quantitative changes in the stratum corneum and the granular layer of the epidermis in the treated animals. Our findings suggest that exacerbating factors in calcium ATPase disorders of the skin suppress epidermal SERCA2 and PMR1 levels, further decreasing the already haploinsufficient protein expression to a potentially critical level in Darier disease and Hailey-Hailey disease, respectively. Lithium therapy should specifically be avoided not only in Darier disease, but Hailey-Hailey disease as well. C1 [Sule, Norbert] University of Pecs, Department of PathologyPecs, Hungary. [Teszas, Alexandra] University of Pecs, Department of Medical Genetics and Child Development, Jozsef A. u. 7., H-7623 Pecs, Hungary. [Kalman, Endre] University of Pecs, Department of PathologyPecs, Hungary. [Szigeti, Reka] University of Pecs, Department of Dermatology, Venereology and OncodermatologyPecs, Hungary. [Miseta, Attila] University of Pecs, Department of Laboratory MedicinePecs, Hungary. [Kellermayer, Richard] University of Pecs, Department of Medical Genetics and Child Development, Jozsef A. u. 7., H-7623 Pecs, Hungary. CR Sakuntabhai A, Ruiz-Perez V, Carter S, et al: Mutations in ATP2A2, encoding a Ca2+ pump, cause Darier disease. Nat Genet 21: 271-277, 1999 Hu Z, Bonifas JM, Beech J, et al: Mutations in ATP2C1, encoding a calcium pump, cause Hailey-Hailey disease. Nat Genet 24: 61-65, 2000 Tavadia S, Authi KS, Hodgins MB, Munro CS: Expression of the sarco/endoplasmic reticulum calcium ATPase type 2 and 3 isoforms in normal skin and Darier's disease. Br J Dermatol 151: 440-445, 2004 Porgpermdee S, Yu X, Takagi A, et al: Expression of SPCA1, Hailey-Hailey disease gene product, in acantholytic dermatoses. J Dermatol Sci 40: 137-140, 2005 Mayuzumi N, Ikeda S, Kawada H, et al: Effects of ultraviolet B irradiation, proinflammatory cytokines and raised extracellular calcium concentration on the expression of ATP2A2 and ATP2C1. Br J Dermatol 152: 697-701, 2005 Rubin MB: Lithium-induced Darier's disease. J Am Acad Dermatol 32: 674-675, 1995 Kellermayer R: Hailey-Hailey disease as an orthodisease of PMR1 deficiency in Saccharomyces cerevisiae. FEBS Lett 579: 2021-2025, 2005 Prasad V, Boivin GP, Miller ML, et al: Haploinsufficiency of Atp2a2, encoding the sarco(endo)plasmic reticulum Ca2+- ATPase isoform 2 Ca2+ pump, predisposes mice to squamous cell tumors via a novel mode of cancer susceptibility. Cancer Res 65: 8655-8661, 2005 Burge SM, Schomberg KH: Adhesion molecules and related proteins in Darier's disease and Hailey-Hailey disease. Br J Dermatol 127: 335-343, 1992 Elias PM, Nau P, Hanley K, et al: Formation of the epidermal calcium gradient coincides with key milestones of barrier ontogenesis in the rodent. J Invest Dermatol 110: 399-404, 1998 Green E, Elvidge G, Jacobsen N, et al: Localization of bipolar susceptibility locus by molecular genetic analysis of the chromosome 12q23-q24 region in two pedigrees with bipolar disorder and Darier's disease. Am J Psychiatry 162: 35-42, 2005 Kellermayer R, Szigeti R, Keeling KM, et al: Aminoglycosides as potential pharmacogenetic agents in the treatment of Hailey- Hailey disease. J Invest Dermatol 126: 229-231, 2006 Yeung CK, Chan HH: Cutaneous adverse effects of lithium: epidemiology and management. Am J Clin Dermatol 5: 3-8, 2004 Csutora P, Karsai A, Nagy T, et al: Lithium induces phosphoglucomutase activity in various tissues of rats and in bipolar patients. Int J Neuropsychopharmacol 9:613-619, 2006 Csutora P, Strassz A, Boldizsar F, et al: Inhibition of phosphoglucomutase activity by lithium alters cellular calcium homeostasis and signaling in Saccharomyces cerevisiae. Am J Physiol Cell Physiol 289: C58-C67, 2005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 234 EP 236 PG 3 ER PT J AU Alberth, M Majoros, L Kovalecz, G Borbas, E Szegedi, I J Marton, I Kiss, Cs AF Alberth, Marta Majoros, Laszlo Kovalecz, Gabriella Borbas, Emese Szegedi, Istvan J Marton, Ildiko Kiss, Csongor TI Significance of Oral Candida Infections in Children with Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE child; cancer; oral infection; Candida albicans; non-albicans Candida strains ID child; cancer; oral infection; Candida albicans; non-albicans Candida strains AB Candidiasis is common in children with cancer, particularly during periods of severe immunosuppression and neutropenia. Our aim was to study the microbiological changes in the oral cavity of children with newly diagnosed cancer. The study group consisted of 30 consecutive children and adolescents, 16 with acute lymphoblastic leukemia and 14 with solid tumors. Oral cultures to detect fungi and bacteria were conducted for all patients before treatment, during and after neutropenic episodes. In 23 patients developing fever simultaneous throat, urine and blood sampling was carried out. No pathogens were found in the cultures taken before the outset (30 cultures) or after recovery from (30 cultures) the neutropenic episodes. In the 45 oral cultures taken during the neutropenic episodes 38 (84.4%) proved positive. Fungi were the most frequently isolated oral pathogens: 33/38 yeast and 6/38 bacterial infections were identified. There was no association between the underlying malignancy and the occurrence of the positive cultures. Of the 30 patients, all 23 (76.7%) who have developed moderate-to-severe neutropenia, developed oral fungal colonization or clinically obvious fungal infection at least on one occasion during the study. In addition to oral samples, fungi were identified in 9/23 pharyngeal swabs, 6/23 urine and 1/23 blood cultures. The initial fungal pathogen was exclusively (33/33) Candida albicans. In extended severe neutropenic states, C. albicans was replaced by non-albicans species (C. kefyr, C. lusitaniae, C. sake, C. tropicalis) in 5 patients between 4 to 6 days of the neutropenic episodes. Four of the nonalbicans Candida strains were resistant to azole-type antifungal agents. Neutropenic episodes of children with cancer are associated with an increased risk of developing oral and even systemic infections with C. albicans that can be replaced by azole-resistant nonalbicans strains in prolonged neutropenia contributing to morbidity of these patients. C1 [Alberth, Marta] University of Debrecen, Faculty of DentistryDebrecen, Hungary. [Majoros, Laszlo] University of Debrecen, Faculty of Medicine, Department of Medical MicrobiologyDebrecen, Hungary. [Kovalecz, Gabriella] University of Debrecen, Faculty of DentistryDebrecen, Hungary. [Borbas, Emese] University of Debrecen, Faculty of DentistryDebrecen, Hungary. [Szegedi, Istvan] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98. Nagyerdei krt., H-4032 Debrecen, Hungary. [J Marton, Ildiko] University of Debrecen, Faculty of DentistryDebrecen, Hungary. [Kiss, Csongor] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98. Nagyerdei krt., H-4032 Debrecen, Hungary. RP Kiss, Cs (reprint author), Medical and Health Science Center, University of Debrecen, Department of Pediatrics, H-4032 Debrecen, Hungary. EM kisscs@dote.hu CR Ball K, Sweeney MP, Baxter WP, Bagg J: Fluconazole sensitivity of Candida species isolated from the mouth of terminally ill cancer patients. Am J Hosp Palliat Care 15: 315-319, 1998 Dahiya MC, Redding SW, Dahiya RS, et al: Oropharyngeal candidiasis caused by non-albicans yeast in patients receiving external beam radiotherapy for head-and-neck cancer. Int J Radiat Oncol Biol Phys 57: 79-83, 2003 Davies AN, Brailsford S, Broadley K, Beighton D: Oral yeast carriage in patients with advanced cancer. Oral Microbiol Immunol 17: 79-84, 2002 Edwards JE Jr: Editorial response: Should all patients with Candidemia be treated with antifungal agents? Clin Infect Dis 15: 422-423, 1992 Epstein JB, Hancock PJ, Nantel S: Oral candidiasis in hematopoietic cell transplantation patients: an outcome-based analysis. Oral Surg Oral Med Oral Pathol Radiol Endod 96: 154-163, 2003 Epstein JB, Ransier A, Lunn R, et al: Prophylaxis of candidiasis in patients with leukemia and bone marrow transplants. Oral Surg Oral Med Oral Pathol Radiol Endod 81: 291-296, 1996 Genc A, Atalay T, Gedikoglu G, et al: Leukemic children: clinical and histopathological gingival lesions. J Clin Pediatr Dent 22: 253-256, 1998 Gozdasoglu S, Ertem M, Buyukkececi Z, et al: Fungal colonization and infection in children with acute leukemia and lymphoma during induction therapy. Med Pediatr Oncol 32: 76-77, 2000 Hoppe JE, Klausner M, Klingebiel T, Niethammer D: Retrospective analysis of yeast colonization and infection in pediatric bone marrow transplant recipients. Mycoses 40: 47-54, 1997 Hughes WT: Systemic candidiasis: a study of 109 fatal cases: Pediatr Infect Disease J 1: 11-18, 1982 Michaud M, Baehner RL, Bixler D, Kafrawy AH: Oral manifestations of acute leukemia in children. J Am Dent Assoc 95: 1145-1150, 1977 Ridola V, Chachaty E, Raimondo G, et al: Candida infections in children with conventional chemotherapy for solid tumors, transplant recipients excluded): The Institute Gustave Roussy Pediatrics Department experience. Pediatr Blood Cancer 42: 332-337, 2004 Simon AR, Roberts MW: Management of oral complications associated with cancer therapy in pediatric patients. J Dent Child 58: 384-398, 1991 Sonis ST, Fey EG: Oral complications of cancer therapy. Oncology, Williston Park, 16: 680-686, 2002 Stinnet EA, Childers NK, Wright JT, et al: The detection of oral Candida in pediatric leukemia patients. Pediatr Dent 14: 236- 239, 1992 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 237 EP 241 PG 5 ER PT J AU Derecskei, K Moldvay, J Bogos, K Timar, J AF Derecskei, Katalin Moldvay, Judit Bogos, Krisztina Timar, Jozsef TI Protocol Modifications Influence the Result of EGF Receptor Immunodetection by EGFR pharmDxTM in Paraffin-Embedded Cancer Tissues SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE EGF receptor immunodetection; EGFR pharmDx; antigen retrieval; incubation time ID EGF receptor immunodetection; EGFR pharmDx; antigen retrieval; incubation time AB EGF receptor (EGFR) became a useful target for several recently introduced therapies of various cancer types including colorectal, lung, head and neck cancers and glioblastoma. The successful clinical application of these novel molecularly targeted therapies requires the expression of their target, EGFR, determined by nucleic acid based or immunohistochemical techniques. However, until now, immunohistochemistry has not become a reliable diagnostic approach for this purpose. The golden standard for the determination of EGFR protein expression in paraffin-embedded cancer tissues is the EGFR pharmDxTM kit. Here we show that the recommended protocol may not be optimal for EGFR immunodetection. Microwave antigen retrieval and extended primary antibody incubation time converted four out of eight EGFR-negative tumors into EGFR-positive in a study of 50 lung adenocarcinoma cases. Accordingly, we recommend retesting cases negative for EGFR with EGFR pharmDxTM using protocol modifications optimizing antigen retrieval and the incubation periods. C1 [Derecskei, Katalin] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Moldvay, Judit] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Bogos, Krisztina] National Koranyi Institute of PulmonologyBudapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. CR Baselga J and Arteaga CL: Critical update and emerging trends in epidermal growth factor receptor targeting in cancer. J Clin Oncol 23:2445-2459, 2005 Meropol NJ: Epidermal growth factor receptor inhibitors in colorectal cancer: it’s time to get back on target. J Clin Oncol 23:1791-1793, 2005 http://www.fda.gov/cdrh/mda/docs http://www.ventanamed.com/catalog/antibody Nguyen PL, Swanson PE, Jaszcz W, et al: Expression of epidermal growth factor receptor in invasive transitional cell carcinoma of the urinary bladder. A multivariate survival analysis. Am J Clin Pathol 101:166-176, 1994 Pii K, Andersen FG, Jensen S, Spaulding B: Characterization of a new monoclonal antibody, clone 2-18C9, for the measurement of epidermal growth factor receptor expression in solid tumor. Proc. 95th AACR, Abstr #5029, 2004 Harari PM and Huang S-M: Searching for reliable epidermal growth factor receptor response predictors. Clin Cancer Res 10:428-432, 2004 Dancey JE: Predictive factors for epidermal growth factor receptor inhibitors. The bull’s-eye hits the arrow. Cancer Cell 5:411-415, 2004 Bunn PA Jr, Dziadziuszko R, Varella-Garcia M, et al: Biological markers for non-small cell lung cancer patient selection for epidermal growth factor receptor tyrosine kinase inhibitor therapy. Clin Cancer Res 12:3652-3656, 2006 Chung KY, Shia J, Kemeny NE, et al: Cetuximab shows activity in colorectal cancer patients with tumors that do not express the epidermal growth factor receptor by immunohistochemistry. J Clin Oncol 23:1803-1810, 2005 Baselga J, Trigo JM, Bourhis J, et al: Phase II multicenter study of the antiepidermal growth factor receptor monoclonal antibody cetuximab in combination with platinum-based chemotherapy in patients with platinum-refractory metastatic and/or recurrent squamous cell carcinoma of the head and neck. J Clin Oncol 23:5568-5577, 2005 Robert F, Blumenschein G, Herbst RS, et al: Phase I/IIa study of cetuximab with gemcitabine plus carboplatin in patients with chemotherapy-naive advanced non-small-cell lung cancer. J Clin Oncol 23:9089-9096, 2005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 243 EP 246 PG 4 ER PT J AU Pappa, L Machera, M Tsanou, E Damala, C Peschos, D Bafa, M Malamou-Mitsi, V AF Pappa, Lina Machera, Melpomeni Tsanou, Eleni Damala, Constantina Peschos, Dimitrios Bafa, Maria Malamou-Mitsi, Vassiliki TI Subcutaneous Metastasis of Peritoneal Mesothelioma Diagnosed by Fine-Needle Aspiration SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE mesothelioma; metastatic nodules; FNA; cytology ID mesothelioma; metastatic nodules; FNA; cytology AB Mesothelioma is a rare malignant neoplasm of the serosal membranes, which can give distant metastases in various organs in advanced stages of its course. Subcutaneous tissue is an unusual metastatic site. In the literature, only one case of metastatic mesothelioma to the skin of the face has been reported. We present a case of a 60-year-old female with a prior history of peritoneal malignant mesothelioma, who 6 months after the initial diagnosis presented with a subcutaneous nodule in the lateral chest wall. Cytological examination of the material obtained by FNA from the nodule revealed metastatic mesothelioma. Although subcutaneous metastasis of malignant mesothelioma is a rare entity, one must always keep this possibility in mind and proceed to further investigation of such lesions. In these cases, FNA is a simple diagnostic procedure for the identification of metastatic disease in patients with a prior history of malignancy. C1 [Pappa, Lina] Ioannina University Hospital, Department of Pathology, 45110 Ioannina, Greece. [Machera, Melpomeni] Ioannina University Hospital, Department of Pathology, 45110 Ioannina, Greece. [Tsanou, Eleni] Ioannina University Hospital, Department of Pathology, 45110 Ioannina, Greece. [Damala, Constantina] Ioannina University Hospital, Department of Pathology, 45110 Ioannina, Greece. [Peschos, Dimitrios] University of Ioannina, Department of Forensic Medicine and ToxicologyIoannina, Greece. [Bafa, Maria] Ioannina University Hospital, Department of Pathology, 45110 Ioannina, Greece. [Malamou-Mitsi, Vassiliki] Ioannina University Hospital, Department of Pathology, 45110 Ioannina, Greece. RP Malamou-Mitsi, V (reprint author), Ioannina University Hospital, Department of Pathology, 45110 Ioannina, Greece. EM me00627@cc.uoi.gr CR Ansari N, Derias N: Diagnosis of malignant mesothelioma by fine needle aspiration of a cervical lymph node. A case report. Acta Cytol 44: 70-74, 2000 Antman K: Natural history and epidemiology of malignant mesothelioma. Chest 103, Suppl): 373-376, 1993 Battifora H, Kopinski M: Distinction of mesothelioma from adenocarcinoma: an immunohistochemical approach. Cancer 55: 1679-1685, 1985 Brenner J, Sordillo P, Magill G, Golbey R: Malignant mesothelioma of the pleura: Review of 123 patients. Cancer 49: 2431- 2435, 1982 Chabot J, Beard D, Langlois A. Mesotheliomas of peritoneum epicardium and pericardium induced by strain of MC29 avian leucosis virus. Cancer Res 30: 1287-1308, 1970 Constantopoulos S, Goudevenos J, Saratzis N, et al: Pleural calcification and restrictive lung function in Northwestern Greece. Environ Res 38: 319-331, 1985 Constantopoulos S, Malamou-Mitsi V, Goudevenos J, et al: High incidence of malignant pleural mesothelioma in neighbouring villages of Northwestern Greece. Respiration 51: 266- 271, 1987 Craig F, Fishback N, Scwartz J, Powers C: Occult metastatic mesothelioma – Diagnosis by fine needle aspiration. A case report. Am J Clin Pathol 97: 493-497, 1992 Dutt P, Baxter J, O’Malley F, et al: Distant cutaneous metastasis of pleural malignant mesothelioma. J Cutan Pathol 19: 490- 495, 1992 Edstrom L, Dawson P, Kohler J, DeMester T: Malignant mesothelioma. A metastasis to the face. J Surg Oncol 14: 251- 254, 1980 Garcia-Reija M, Matilla J, DePaz A, et al: Unusual metastasis to the mandibular alveolus of a malignant pleural mesothelioma. Otolaryngol Head Neck Surg 126: 435-437, 2002 Hillerdal G: Malignant mesothelioma 1982: Review of 4710 published cases. Br J Dis Chest 77: 321-343, 1983 Kaye J, Wang A: Malignant mesothelioma with brain metastases. Am J Med 80: 95-97, 1987 Kim B, Varkey B, Choi H: Diagnosis of malignant pleural mesothelioma by axillary lymph node biopsy. Chest 91: 278- 281, 1987 Kubota K, Furuse K, Kawahara M, et al: A case of malignant pleural mesothelioma with metastasis to the orbit. Jpn J Clin Oncol 26: 469-471, 1996 Losi L, Cocchi R, Calbucci F, Eusebi V: Metastasis of pleural malignant mesothelioma to the brain and upper maxilla: description of 2 cases. Pathologica 92: 273-277, 2000 MacDonald AD, Harper A, ElAttar O: Epidemiology of primary malignant mesothelial tumors in Canada. Cancer 26: 914- 919, 1970 McEwen J, Finlayson A, Mair A: Mesothelioma in Scotland. Br Med J 4: 575-578, 1970 Moertel CG: Peritoneal mesothelioma. Gastroenterology 63: 346-350, 1972 Obers V, Leiman G, Girdwood R, Spiro F: Primary malignant pleural tumors, mesotheliomas, presenting as localized masses. Fine needle aspiration cytologic findings, clinical and radiologic features and review of the literature. Acta Cytol 32: 568-575, 1988 Prieto V, Kenet B, Varghese M: Malignant mesothelioma metastatic to the skin presenting as inflammatory carcinoma. Am J Dermatopathol 19: 261-265, 1997 Roberts G: Distant visceral metastases in pleural mesothelioma. Br J Dis Chest 70: 246-250, 1976 Sebbag G, Sugarbaker P: Peritoneal mesothelioma proposal for a staging system. Eur J Surg Oncol 27: 223-224, 2001 Sgrignoli A, Abati A, Pass H, et al: Metastatic mesothelioma presenting as a salivary gland neoplasm. Acta Cytol 42: 818- 820, 1998 Sheibani K, Esteban J, Bailey A, et al: Immunopathologic and molecular studies as an aid to the diagnosis of malignant mesothelioma. Hum Pathol 23: 107-116, 1992 Sheibani K, Shinn S, Kezirian J, Weiss L: BerEP4 antibody as a discriminant in the differential diagnosis of malignant mesothelioma versus adenocarcinoma. Am J Surg Pathol 15: 79-84, 1991 Sterret G, Whitaker D, Shilkin K, Walters MN: Fine needle aspiration cytology of malignant mesothelioma. Acta Cytol 31: 185-193, 1987 Sussman J, Rosai J: Lymph node metastasis as the initial manifestation of malignant mesothelioma. Report of six cases. Am J Surg Pathol 14: 819-828, 1990 Wagner J, Sleggs C, Marshad P: Diffuse pleural mesothelioma and asbestos exposure in the North Western Cape Province. Br J Ind Med 17: 260-271, 1960 Walters K, Martinez A: Malignant fibrous mesothelioma metastatic to brain and liver. Acta Neuropathol 33: 173-177, 1975 Wanebo H, Martini N, Melamed M, et al: Pleural mesothelioma. Cancer 38: 2481-2488, 1976 Yu G, Baloch Z, Gupta P: Cytomorphology of metastatic mesothelioma in fine needle aspiration specimens. Diagn Cytopathol 20: 328-332, 1999 Zanconati F, DelConte A, Bonifacio-Gori D, Falconieri G: Metastatic pleural mesothelioma presenting with solitary involvement of the tongue: a report of a new case and review of the literature. Int J Surg Pathol 11: 51-55, 2003 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 247 EP 250 PG 4 ER PT J AU Kondo, T Tanaka, Y AF Kondo, Takeshi Tanaka, Yoshio TI Malignant Pilomatricoma in the Parietal Area SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE malignant pilomatricoma; pilomatrix carcinoma; basophilic cells; distant metastasis ID malignant pilomatricoma; pilomatrix carcinoma; basophilic cells; distant metastasis AB A 27-year-old Japanese woman presented with a 2.5-cm nodular subcutaneous lesion in the parietal area. The nodule was well demarcated and situated in the dermis and subcutis. Histologically, the tumor was diagnosed as malignant pilomatricoma. The tumor was excised, the postoperative course was uneventful, no evidence of local recurrence or distant metastasis was observed, and the patient continues to be under close follow-up. Malignant pilomatricoma, a locally aggressive counterpart of benign pilomatricoma, is also referred to as pilomatrix carcinoma. Most cases are excised as benign tumors; however, when the excision is incomplete local recurrence is likely, and distant metastases have also been reported. Histologically, the diagnosis can be challenging because no clear histologic criteria are available. Because of the rarity of malignant pilomatricoma, no welldefined standards in the surgical management of this neoplasm have been established. Moreover, since distant metastases have been described, close followup of the lesion is requisite. C1 [Kondo, Takeshi] Kobe University Graduate School of Medicine, Department of Biomedical Informatics, Division of Molecular Pathology, 7-5-1 Kusunoki-cho, Chuo-ku, 650-0017 Kobe, Japan. [Tanaka, Yoshio] Shinko Hospital, Department of Plastic SurgeryKobe, Japan. RP Kondo, T (reprint author), Kobe University Graduate School of Medicine, Department of Biomedical Informatics, Division of Molecular Pathology, 650-0017 Kobe, Japan. EM kondo@med.kobe-u.ac.jp CR Dutta R, Boadle R, Ng T: Pilomatrix carcinoma: case report and review of literature. Pathology 33: 248-251, 2001 Elder D, Elenitsas R, Ragsdale BD: Tumors of the epidermal appendages. In: LEVER’s Histopathology of the Skin. 8th ed., Ed: Elder D), Lippincott-Raven, Philadelphia, 1997, p759 Fujiwara T, Yamamoto H, Hashiro M: Malignant pilomatricoma. Scand J Plast Reconstr Surg Hand Surg 36: 119-121, 2002 Gould E, Kurzon R, Kowalczyk AP, Saldana M:Pilomatrix carcinoma with pulmonary metastasis. Report of a case. Cancer 54: 370-372, 1984 Hardisson D, Linares MD, Cuevas-Santos J, Contreras F: Pilomatrix carcinoma: a clinicopathologic study of six cases and review of the literature. Am J Dermatopathol 23: 394-401, 2001 Lopansri S, Mihm MC Jr: Pilomatrix carcinoma or calcifying epitheliocarcinoma of Malherbe: a case report and review of literature. Cancer 45: 2368-2373, 1980 Malignant pilomatricoma. In: Atlas of Tumor Pathology Non- Melanocytic Tumors of the Skin., Eds: Murphy GF and Elder DE), AFIP, Washington, DC, 1990, pp.147-148 Manivel C, Wick MR, Mukai K: Pilomatrix carcinoma: an immunohistochemical comparison with benign pilomatrixoma and other benign cutaneous lesions of pilar origin. J Cutan Pathol 13: 22-29, 1986 Martelli G, Giardini R: Pilomatrix carcinoma: a case report and review of the literature. Eur J Surg Oncol 20: 703-704, 1994 Mir R, Cortes E, Papantoniou PA, et al: Metastatic trichomatricial carcinoma. Arch Pathol Lab Med 110: 660-663, 1986 Sable D, Snow SN: Pilomatrix carcinoma of the back treated by mohs micrographic surgery. Dermatol Surg 30: 1174-1176, 2004 Sau P, Lupton GP, Graham JH: Pilomatrix carcinoma. Cancer 71: 2491-2498, 1993 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 251 EP 253 PG 3 ER PT J AU Merkli, H Pal, E Gati, I AF Merkli, Hajnalka Pal, Endre Gati, Istvan TI Asymmetric Calf Hypertrophy of Neurogenic Origin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE asymmetric atrophy; muscle biopsy; neuropathy; spinal muscular atrophy ID asymmetric atrophy; muscle biopsy; neuropathy; spinal muscular atrophy AB A 47-year-old male presented with painful swelling of the right calf. His medical history was negative, except for a herniation of disc LIV-V 5 years before. Physical examination revealed unilateral calf hypertrophy with moderate weakness of plantarflexion, mild paresis of dorsiflexion. Electromyography showed a peripheral neurogenic lesion in the right anterior tibial muscle, but normal findings were obtained from the unaffected quadriceps muscle. Histological examination of the right gastrocnemic muscle showed neurogenic changes with typical targetoid fibers, but no pathological changes were present in the quadriceps muscle. Chronic asymmetric spinal muscular atrophy is an infrequent neuromuscular disease and because of asymmetric appearance, it might be difficult to distinguish from other, acquired neurogenic muscle diseases such as radiculopathy caused by intervertebral disc herniation. Our case confirms that muscular hypertrophy can follow partial denervation in humans. C1 [Merkli, Hajnalka] University of Pecs, Department of Neurology, Ret u. 2., H-7623 Pecs, Hungary. [Pal, Endre] University of Pecs, Department of Neurology, Ret u. 2., H-7623 Pecs, Hungary. [Gati, Istvan] University of Pecs, Department of Neurology, Ret u. 2., H-7623 Pecs, Hungary. RP Pal, E (reprint author), University of Pecs, Department of Neurology, H-7623 Pecs, Hungary. EM endre.pal@aok.pte.hu CR Adams RD, Denny-Brown D, Pearson CM: Diseases of Muscle. A Study in Pathology. Second Edition. Harper and Row, Hagerstown, Maryland, 1962, pp. 349-353, 666-669. Barron SA, Heffner RR: Abnormal spontaneous electrical activity and gross enlargement of muscle. Arch Neurol 40: 515-518, 1983. Bernat JL, Ochoa JL: Muscle hypertrophy after partial denervation: a human case. J Neurol Neurosurg Psychiat 41: 719-725, 1978. Bundey S, Lovelace RE: A clinical and genetic study of chronic proximal spinal muscular atrophy. Brain 98: 455-472, 1975. Celesia GG, Andermann F, Wiglesworth FW, Robb JP: Monomelic myopathy: Congenital hypertrophic myotonic myopathy limited to one extremity. Arch Neurol 17: 69-77, 1967. Costa J, Graca P, Evangelista T, de Carvalho M: Pain and calf hypertrophy associated with spontaneous repetitive discharges treated with botulinum toxin. Clin Neurophysiol 116: 2847- 2852, 2005. De Visser M, Verbeten B, Lyppens KC: Pseudohypertrophy of the calf following S1 radiculopathy. Neuroradiol 28: 279-280, 1986. Dubowitz V, Brooke MH, Neville HE: Muscle biopsy: A Modern Approach. Saunders W.B. Co, London, 1973, pp 20-33. Gobbele R, Schoen SW, Schroder JM, et al: S1 radiculopathy as a possible predisposing factor in focal myositis with unilateral hypertrophy of the calf muscles. J Neurol Sci 170: 64-68, 1999. Harding AE, Bradbury PG, Murray NM: Chronic asymmetrical spinal muscular atrophy. J Neurol Sci 59: 69-83, 1983. Korczyn AD, Kuritzky A, Sandbank U: Muscle hypertrophy with neuropathy. J Neurol Sci 36: 399-408, 1978. Mielke U, Ricker K, Emser W, Boxler K: Unilateral calf enlargement following S1 radiculopathy. Muscle Nerve 5: 434-438, 1982. Montagna P, Martinelli P, Rasi F, et al: Muscular hypertrophy after chronic radiculopathy. Arch Neurol 41: 397-398, 1984. Pearn J, Hudgson P: Anterior-horn cell degeneration and gross calf hypertrophy with adolescent onset. A new spinal muscular atrophy syndrome. Lancet 1: 1059-1061, 1978. Pearn JH: Classification of the spinal muscular atrophies. Lancet 1: 919, 1980. Ricker K, Rohkamm R, Moxley RT: Hypertrophy of the calf with S-1 radiculopathy. Arch Neurol 45: 660-664, 1988. Valenstein E, Watson RT, Parker JL: Myokymia, muscle hypertrophy and percussion myotonia in chronic recurrent polyneuropathy. Neurology 28: 1130-1134, 1978. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2006 VL 12 IS 4 BP 254 EP 256 PG 3 ER PT J AU Cserni, G Bianchi, S Vezzosi, V Arisio, R Bori, R Peterse, LJ Sapino, A Castellano, I Drijkoningen, M Kulka, J Eusebi, V Foschini, PM Bellocq, JP Marin, C Thorstenson, S Amendoeira, I Reiner-Concin, A Decker, Th Lacerda, M Figueiredo, P Fejes, G AF Cserni, Gabor Bianchi, Simonetta Vezzosi, Vania Arisio, Riccardo Bori, Rita Peterse, L Johannes Sapino, Anna Castellano, Isabella Drijkoningen, Maria Kulka, Janina Eusebi, Vincenzo Foschini, P Maria Bellocq, Jean-Pierre Marin, Cristi Thorstenson, Sten Amendoeira, Isabel Reiner-Concin, Angelika Decker, Thomas Lacerda, Manuela Figueiredo, Paulo Fejes, Gabor TI Sentinel Lymph Node Biopsy in Staging Small (up to 15 mm) Breast Carcinomas. Results from a European Multi-institutional Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sentinel lymph node; non-sentinel lymph node; breast cancer; pT1 ID Sentinel lymph node; non-sentinel lymph node; breast cancer; pT1 AB Sentinel lymph node (SLN) biopsy has become the preferred method for the nodal staging of early breast cancer, but controversy exists regarding its universal use and consequences in small tumors. 2929 cases of breast carcinomas not larger than 15 mm and staged with SLN biopsy with or without axillary dissection were collected from the authors’ institutions. The pathology of the SLNs included multilevel hematoxylin and eosin (HE) staining. Cytokeratin immunohistochemistry (IHC) was commonly used for cases negative with HE staining. Variables influencing SLN involvement and non-SLN involvement were studied with logistic regression. Factors that influenced SLN involvement included tumor size, multifocality, grade and age. Small tumors up to 4 mm (including in situ and microinvasive carcinomas) seem to have SLN involvement in less than 10%. Non-SLN metastases were associated with tumor grade, the ratio of involved SLNs and SLN involvement type. Isolated tumor cells were not likely to be associated with further nodal load, whereas micrometastases had some subsets with low risk of non-SLN involvement and subsets with higher proportion of further nodal spread. In situ and microinvasive carcinomas have a very low risk of SLN involvement, therefore, these tumors might not need SLN biopsy for staging, and this may be the approach used for very small invasive carcinomas. If an SLN is involved, isolated tumor cells are rarely if ever associated with non-SLN metastases, and subsets of micrometastatic SLN involvement may be approached similarly. With macrometastases the risk of non-SLN involvement increases, and further axillary treatment should be generally indicated. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, H-6000 Kecskemet, Hungary. [Bianchi, Simonetta] AOU Careggi, Department of Human Pathology and OncologyFlorence, Italy. [Vezzosi, Vania] AOU Careggi, Department of Human Pathology and OncologyFlorence, Italy. [Arisio, Riccardo] Sant’Anna Hospital, Department of PathologyTurin, Italy. [Bori, Rita] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, H-6000 Kecskemet, Hungary. [Peterse, L Johannes] The Netherlands Cancer Institute, Department of PathologyAmsterdam, The Netherlands. [Sapino, Anna] University of Turin, Department of Biological Science and Human OncologyTurin, Italy. [Castellano, Isabella] University of Turin, Department of Biological Science and Human OncologyTurin, Italy. [Drijkoningen, Maria] University Hospitals Leuven, Pathologische OntleedkundeLeuven, Belgium. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Eusebi, Vincenzo] Bellaria Hospital, Pathologic Anatomy UnitBologna, Italy. [Foschini, P Maria] Bellaria Hospital, Pathologic Anatomy UnitBologna, Italy. [Bellocq, Jean-Pierre] Hopital de Hautepierre, Service d’Anatomie PathologiqueStrasbourg, France. [Marin, Cristi] Hopital de Hautepierre, Service d’Anatomie PathologiqueStrasbourg, France. [Thorstenson, Sten] Kalmar Hospital, Department of Pathology and CytologyKalmar, Sweden. [Amendoeira, Isabel] University of Porto, Department of Pathology and Immunology of ICBASPorto, Portugal. [Reiner-Concin, Angelika] Donauspital, Pathologisch-Bakteriologisches InstitutVienna, Austria. [Decker, Thomas] University of Munster, Department of PathologyMunster, Germany. [Lacerda, Manuela] Centro Regional De Oncologia De Coimbra, Laboratorio De HistopatologicaCoimbra, Portugal. [Figueiredo, Paulo] Centro Regional De Oncologia De Coimbra, Laboratorio De HistopatologicaCoimbra, Portugal. [Fejes, Gabor] Bacs-Kiskun County Teaching Hospital, Department of InformaticsKecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.hu CR Moore MP, Kinne DW: Axillary lymphadenectomy: a diagnostic and therapeutic procedure. J Surg Oncol 66: 2-6, 1997 Fisher B: Laboratory and clinical research in breast cancer - a personal adventure: the David A. Karnofsky memorial lecture. Cancer Res 40: 3863-3874, 1980 Forrest AP, Stewart HJ, Roberts MM, et al: Simple mastectomy and axillary node sampling, pectoral node biopsy, in the management of primary breast cancer. Ann Surg 196: 371-378, 1982 Ahlgren J, Holmberg L, Bergh J, et al: Five-node biopsy of the axilla: an alternative to axillary dissection of levels I-II in operable breast cancer. Eur J Surg Oncol 28:97-102, 2002 Sato K, Tamaki K, Takeuchi H, et al: Management of the axilla in breast cancer: a comparative study between sentinel lymph node biopsy and four-node sampling procedure. Jpn J Clin Oncol 31: 318-321, 2001 Krag DN, Weaver DL, Alex JC, et al: Surgical resection and radiolocalization of sentinel lymph node in breast cancer using a gamma probe. Surg Oncol 2: 335-340, 1993 Giuliano AE, Kirgan DM, Guenther JM, et al: Lymphatic mapping and sentinel lymphadenectomy for breast cancer. Ann Surg 220: 391-398, 1994 Albertini JJ, Lyman GH, Cox C, et al: Lymphatic mapping and sentinel node biopsy in the patient with breast cancer. JAMA 276: 1818-1822, 1996 Al-Hilaly M, Willsher PC, Robertson JFR, et al: Audit of a conservative management policy of the axilla in elderly patients with operable breast cancer. Eur J Surg Oncol 23: 339-340, 1997 Ragaz J, Jackson SM, Le N, et al: Adjuvant radiotherapy and chemotherapy in node-positive premenopausal women with breast cancer. N Engl J Med 337: 956-962, 1997 Overgaard M, Hansen PS, Overgaard J, et al: Postoperative radiotherapy in high-risk premenopausal women with breast cancer who receive adjuvant chemotherapy. N Engl J Med 337: 949- 955, 1997 Overgaard M, Jensen MB, Overgaard J, et al: Postoperative radiotherapy in high-risk postmenopausal breast-cancer patients given adjuvant tamoxifen: Danish Breast Cancer Cooperative Group DBCG 82c randomised trial. Lancet 353: 1641-1648, 1999 Vinh-Hung V, Verschraegen C: Breast-conserving surgery with or without radiotherapy: pooled-analysis for risks of ipsilateral breast tumor recurrence and mortality. J Natl Cancer Inst 96: 115-121, 2004 Orr RK: The impact of prophylactic axillary node dissection on breast cancer survival - a Bayesian meta-analysis. Ann Surg Oncol 6: 109-116, 1999 Early Breast Cancer Trialists’ Collaborative Group: Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: an overview of the randomised trials. Lancet 366: 2087-2106, 2005 Silverstein MJ, Lagios MD, Recht A, et al: Image-detected breast cancer: state of the art diagnosis and treatment. J Am Coll Surg 201: 586-597, 2005 Schwartz GF, Giuliano AE, Veronesi U, et al: Proceedings of the consensus conference on the role of sentinel lymph node biopsy in carcinoma of the breast, April 19-22, 2001, Philadelphia, Pennsylvania. Cancer 94: 2542-2551, 2002 Pendas S, Dauway E, Giuliano R, et al: Sentinel node biopsy in ductal carcinoma in situ patients. Ann Surg Oncol 7: 15-20, 2000 Klauber-DeMore N, Tan LK, Liberman L, et al: Sentinel lymph node biopsy: is it indicated in patients with high-risk ductal carcinoma- in-situ and ductal carcinoma-in-situ with microinvasion? Ann Surg Oncol 7: 636-642, 2000 Cox CE, Nguyen K, Gray RJ, et al: Importance of lymphatic mapping in ductal carcinoma in situ, DCIS): why map DCIS? Am Surg 67: 513-521, 2001 Intra M, Zurrida S, Maffini F, et al: Sentinel lymph node metastasis in microinvasive breast cancer. Ann Surg Oncol 10: 1160- 1165, 2003 Degnim AC, Griffith KA, Sabel MS, et al: Clinicopathologic features of metastasis in nonsentinel lymph nodes of breast carcinoma patients. Cancer 98: 2307-2315, 2003 Cserni G, Gregori D, Merletti F, et al: Non-sentinel node metastases associated with micrometastatic sentinel nodes in breast cancer: metaanalysis of 25 studies. Br J Surg 91: 1245-1252, 2004 Schrenk P, Konstantiniuk P, Wolfl S, et al: Prediction of non-sentinel lymph node status in breast cancer with a micrometastatic sentinel node. Br J Surg 92: 707-713, 2005 Houvenaeghel G, Nos C, Mignotte H, et al: Micrometastases in sentinel lymph node in a multicentric study: predictive factors of nonsentinel lymph node involvement—Groupe des Chirurgiens de la Federation des Centres de Lutte Contre le Cancer. J Clin Oncol 24: 1814-1822, 2006 Van Zee KJ, Manasseh DM, Bevilacqua JL, et al: A nomogram for predicting the likelihood of additional nodal metastases in breast cancer patients with a positive sentinel node biopsy. Ann Surg Oncol 10: 1140-1151, 2003 Lambert LA, Ayers GD, Hwang RF, et al: Validation of a breast cancer nomogram for predicting nonsentinel lymph node metastases after a positive sentinel node biopsy. Ann Surg Oncol 13: 310-320, 2006 Smidt ML, Kuster DM, van der Wilt GJ, et al: Can the Memorial Sloan-Kettering Cancer Center nomogram predict the likelihood of nonsentinel lymph node metastases in breast cancer patients in the Netherlands? Ann Surg Oncol 12: 1066-1072, 2005 Kocsis L, Svebis M, Boross G, et al: Use and limitations of a nomogram predicting the likelihood of non-sentinel node involvement after a positive sentinel node biopsy in breast cancer patients. Am Surg 70: 1019-1024, 2004 Degnim AC, Reynolds C, Pantvaidya G, et al: Nonsentinel node metastasis in breast cancer patients: assessment of an existing and a new predictive nomogram. Am J Surg 190: 543-550, 2005 Cserni G: Comparison of different validation studies on the use of the Memorial-Sloan Kettering Cancer Center nomogram predicting nonsentinel node involvement in sentinel node positive breast cancer patients. Am J Surg, in press) Cserni G, Amendoeira I, Apostolikas N, et al: Pathological workup of sentinel lymph nodes in breast cancer. Review of current data to be considered for the formulation of guidelines. Eur J Cancer 39: 1654-1667, 2003 Cserni G, Amendoeira I, Apostolikas N, et al: Discrepancies in current practice of pathological evaluation of sentinel lymph nodes in breast cancer. Results of a questionnaire-based survey by the European Working Group for Breast Screening Pathology. J Clin Pathol 57: 695-701, 2004 Cserni G, Bianchi S, Boecker W, et al: Improving the reproducibility of diagnosing micrometastases and isolated tumor cells. Cancer 103: 358-367, 2005 Cserni G, Bianchi S, Vezzosi V, et al: The value of cytokeratin immunohistochemistry in the evaluation of axillary sentinel lymph nodes in patients with lobular breast carcinoma. J Clin Pathol 59: 518-522, 2006 Perry N, Broeders M, de Wolf C, et al, Eds): European Guidelines for Breast Screening and Diagnosis. European Communities, Luxemburg, 2006 Sobin LH and Wittekind C, Eds): UICC TNM Classification of Malignant Tumors, 6th edition. John Wiley and Sons, Inc., New- York, 2002 Ellis IO, Pinder SE, Bobrow L, et al: Pathology reporting of breast disease. http://www.cancerscreening.nhs.uk/breastscreen/publications/ nhsbsp58-low-resolution.pdf., Last assessed: 11 November 2006) Tavassoli FA, Devilee P, Eds): World Health Organization: Tumors of the Breast and Female Genital Organs, 4th edition, IARC Press, Lyon, 2003 Greene FL, Page DL, Fleming ID, et al, Eds): AJCC Cancer Staging Handbook – TNM Classification of Malignant Tumors, 6th edition. Springer Verlag, New York, 2002 Hermanek P, Hutter RVP, Sobin LH, et al: Classification of isolated tumor cells and micrometastasis. Cancer 86: 2668-2673, 1999 Cserni G, Sapino A, Decker T: Discriminating between micrometastases and isolated tumor cells in a regional and institutional setting. Breast 15: 347-354, 2006 Miller WG: OpenStat. http://www.statpages.org/miller/openstat/ OpenStat.htm, Last assessed: 15 August 2006) Cserni G, Burzykowski T, Vinh-Hung V, et al: Axillary sentinel node and tumor-related factors associated with non-sentinel node involvement in breast cancer. Jpn J Clin Oncol 34: 519- 524, 2004 Woodward WA, Vinh-Hung V, Ueno NT, et al: Prognostic value of nodal ratios in node-positive breast cancer. J Clin Oncol 24: 2910- 2916, 2006 Cady B: New era in breast cancer. Impact of screening on disease presentation. Surg Oncol Clin N Am 6: 195-202, 1997 McMasters KM, Chao C, Wong SL, et al: Sentinel lymph node biopsy in patients with ductal carcinoma in situ: a proposal. Cancer 95: 15-20, 2002 Holloway C: Surgery for ductal carcinoma in situ. Curr Probl Cancer 24: 141-162, 2000 Lagios MD, Silverstein MJ: Sentinel node biopsy for patients with DCIS: a dangerous and unwarranted direction. Ann Surg Oncol 8: 275-277, 2001 Intra M, Veronesi P, Mazzarol G, et al: Axillary sentinel lymph node biopsy in patients with pure ductal carcinoma in situ of the breast. Arch Surg 138: 309-313, 2003 Trisal V, Qian D, Wagman LD: Axillary recurrence in DCIS: is axillary lymphadenectomy warranted? Am Surg 70: 876-880, 2004 Leidenius M, Salmenkivi K, von Smitten K, et al: Tumor-positive sentinel node findings in patients with ductal carcinoma in situ. J Surg Oncol 94: 380-384, 2006 Cserni G, Boross G, Maraz R, et al: Sentinel lymph node biopsy for in situ carcinoma of the breast. Experience at the Bacs-Kiskun County Hospital and review of the literature [In Hungarian]. Magy Seb 59: 164-172, 2006 Torok K, Peley G, Matrai Z, et al: The role of sentinel lymph node biopsy for staging patients with ductal carcinoma in situ of the breast. [In Hungarian] Magy Seb 59: 173-178, 2006 Damiani S, Ludvikova M, Tomasic G, et al: Myoepithelial cells and basal lamina in poorly differentiated in situ duct carcinoma of the breast. An immunocytochemical study. Virchows Arch 434: 227-234, 1999 Moran CJ, Kell MR, Kerin MJ: The role of sentinel lymph node biopsy in ductal carcinoma in situ. EJSO 31: 1105-1111, 2005 Silverstein MJ, Rosser RJ, Gierson ED, et al: Axillary lymph node dissection for intraductal breast carcinoma – is it indicated? Cancer 59: 1819-1824, 1987 Arisio R, Sapino A, Cassoni P, et al: What modifies the relation between tumor size and lymph node metastases in T1 breast carcinomas? J Clin Pathol 53: 846-850, 2000 Della Rovere GQ, Bonomi R, Ashley S, et al: Axillary staging in women with small invasive breast tumors. EJSO 32: 733-737, 2006 Diaz LK, Wiley EL, Venta LA: Are malignant cells displaced by large-gauge needle core biopsy of the breast? Am J Roentgenol 173: 1303-1313, 1999 Diaz NM, Mayes JR, Vrcel V: Breast epithelial cells in dermal angiolymphatic spaces: a manifestation of benign mechanical transport. Hum Pathol 36: 310-313, 2005 Nagi C, Bleiweiss I, Jaffer S: Epithelial displacement in breast lesions: a papillary phenomenon. Arch Pathol Lab Med 129: 1465-1469, 2005 Bleiweiss IJ, Nagi CS, Jaffer S: Axillary sentinel lymph nodes can be falsely positive due to iatrogenic displacement and transport of benign epithelial cells in patients with breast carcinoma. J Clin Oncol 24: 2013-2018, 2006 Bleiweiss IJ: Sentinel lymph nodes in breast cancer after 10 years: rethinking basic principles. Lancet Oncol 7: 686-692, 2006 Viale G, Zurrida S, Maiorano E, et al: Predicting the status of axillary sentinel lymph nodes in 4351 patients with invasive breast carcinoma treated in a single institution. Cancer 103: 492-500, 2005 Vinh-Hung V, Verschraegen C, Promish DI, et al: Ratios of involved nodes in early breast cancer. Breast Cancer Res 6: R680- 688, 2004 Calhoun KE, Hansen NM, Turner RR, et al: Nonsentinel node metastases in breast cancer patients with isolated tumor cells in the sentinel node: implications for completion axillary node dissection. Am J Surg 190: 588-591, 2005 van Rijk MC, Peterse JL, Nieweg OE, et al: Additional axillary metastases and stage migration in breast cancer patients with micrometastases or submicrometastases in sentinel lymph nodes. Cancer 107: 467-471, 2006 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 5 EP 14 PG 10 ER PT J AU Wincewicz, A Sulkowska, M Koda, M Sulkowski, S AF Wincewicz, Andrzej Sulkowska, Mariola Koda, Mariusz Sulkowski, Stanislaw TI Clinicopathological Significance and Linkage of the Distribution of HIF-1alpha and GLUT-1 in Human Primary Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HIF-1α; GLUT-1; colorectal cancer; cell survival ID HIF-1α; GLUT-1; colorectal cancer; cell survival AB HIF-1alpha induces GLUT-1 expression, and their presence has been evaluated in colorectal cancer. However, the expressions of GLUT-1 and HIF-1alpha have not been investigated together with reference to clinicopathological characteristics in human colorectal cancer. The aim of our study was to compare the expression of HIF-1alpha and GLUT-1 with various clinicopathological features of colorectal cancer. The presence of HIF-1alpha and GLUT-1 was visualized immunohistochemically in 123 primary tumors. Membranous localization of GLUT-1 was found in multifocally necrotizing cancer samples, while pure cytoplasmic perinuclear, mostly supranuclear GLUT-1 accumulation was characteristic of cancer fields with lack of necrosis. HIF-1alpha was located in the cytoplasm and occasionally in the nuclei of cancer cells. Immunoreactivity to GLUT-1 was significantly higher in node-positive cancers compared with nodenegative ones (p=0.04), confirming our earlier results obtained on a larger number of patients. Non-mucinous adenocarcinomas expressed GLUT-1 and HIF-1alpha with significantly greater frequency than mucinous adenocarcinomas (p=0.002, p=0.0002, respectively). GLUT-1 and HIF-1alpha expression did not differ in relation to tumor stage, location, or patients’ age or gender. In contrast to that of GLUT-1, expression of HIF-1alpha correlated with grade (p=0.00003) without difference with regard to pN status. HIF-1alpha expression correlated with GLUT-1 expression in the whole patient population, as well as in all clinicopathological groups except for the pT1+pT2 group. Although the coexpression of cytoplasmic HIF-1alpha and GLUT-1 does not directly prove the dependence between HIF-1 as a nuclear transcriptional factor and GLUT-1 as its downstream protein, it is evidence of their simultaneous upregulation. The extranuclear accumulation of HIF-1alpha and GLUT-1 requires further studies to explain its significance in colorectal cancer. C1 [Wincewicz, Andrzej] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Sulkowska, Mariola] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Koda, Mariusz] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Sulkowski, Stanislaw] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. RP Sulkowski, S (reprint author), Medical University of Bialystok, Department of General Pathomorphology, 15-269 Bialystok, Poland. EM sulek@zeus.amb.edu.pl CR Wang GL, Jiang BH, Rue EA, Semenza GL: Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci USA 92: 5510- 5514, 1995 Wood SM, Wiesener MS, Yeates KM, Okada N, Pugh CW, Maxwell PH, Ratcliffe PJ: Selection and analysis of a mutant cell line defective in the hypoxia-inducible factor-1 alpha-subunit, HIF-1alpha). Characterization of hif-1alpha-dependent and -independent hypoxia-inducible gene expression. J Biol Chem 273: 8360-8368, 1998 Allen JW, Johnson RS, Bhatia SN: Hypoxic inhibition of 3- methylcholanthrene-induced CYP1A1 expression is independent of HIF-1alpha. Toxicol Lett 155: 151-159, 2005 Chen J, Zhao S, Nakada K, Kuge Y, Tamaki N, Okada F, Wang J, Shindo M, Higashino F, Takeda K, Asaka M, Katoh H, Sugiyama T, Hosokawa M, Kobayashi M: Dominant-negative hypoxia-inducible factor-1 alpha reduces tumorigenicity of pancreatic cancer cells through the suppression of glucose metabolism. Am J Pathol 162: 1283-1291, 2003 Williams KJ, Telfer BA, Airley RE, Peters HP, Sheridan MR, van der Kogel AJ, Harris AL, Stratford IJ: A protective role for HIF- 1 in response to redox manipulation and glucose deprivation: implications for tumorigenesis. Oncogene 21: 282-290, 2002 Vleugel MM, Greijer AE, Shvarts A, van der Groep P, van Berkel M, Aarbodem Y, van Tinteren H, Harris AL, van Diest PJ, van der Wall E: Differential prognostic impact of hypoxia induced and diffuse HIF-1alpha expression in invasive breast cancer. J Clin Pathol 58: 172-177, 2005 Kunkel M, Reichert TE, Benz P, Lehr HA, Jeong JH, Wieand S, Bartenstein P, Wagner W, Whiteside TL: Overexpression of Glut-1 and increased glucose metabolism in tumors are associated with a poor prognosis in patients with oral squamous cell carcinoma. Cancer 97: 1015-1024, 2003 Yoshimura H, Dhar DK, Kohno H, Kubota H, Fujii T, Ueda S, Kinugasa S, Tachibana M, Nagasue N: Prognostic impact of hypoxia-inducible factors 1alpha and 2alpha in colorectal cancer patients: correlation with tumor angiogenesis and cyclooxygenase- 2 expression. Clin Cancer Res. 10: 8554-8560, 2004 Cooper R, Sarioglu S, Sokmen S, Fuzun M, Kupelioglu A, Valentine H, Gorken IB, Airley R, West C: Glucose transporter- 1, GLUT-1): a potential marker of prognosis in rectal carcinoma? Br J Cancer 89: 870-876, 2003 Furudoi A, Tanaka S, Haruma K, Yoshihara M, Sumii K, Kajiyama G, Shimamoto F: Clinical significance of human erythrocyte glucose transporter 1 expression at the deepest invasive site of advanced colorectal carcinoma. Oncology 60: 162-169, 2001 Haber RS, Rathan A, Weiser KR, Pritsker A, Itzkowitz SH, Bodian C, Slater G, Weiss A, Burstein DE: GLUT1 glucose transporter expression in colorectal carcinoma: a marker for poor prognosis. Cancer 83: 34-40, 1998 Palit V, Phillips RM, Puri R, Shah T, Bibby MC: Expression of HIF-1alpha and Glut-1 in human bladder cancer. Oncol Rep 14: 909-913, 2005 Koukourakis MI, Giatromanolaki A, Harris AL, Sivridis E: Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: a metabolic survival role for tumor-associated stroma. Cancer Res 66: 632-637, 2006 Wincewicz A, Sulkowska M, Koda M, Kanczuga-Koda L, Witkowska E, Sulkowski S: Significant co-expression of GLUT- 1, Bcl-xl and Bax in colorectal cancer. Ann N Y Acad Sci 2007, accepted) Fogt F, Wellmann A, Urbanski SJ, Noffsinger A, Poremba C, Zimmerman RL, Alsaigh N: Glut-1 expression in dysplastic and regenerative lesions of the colon. Int J Mol Med 7: 615-619, 2001 Ito H, Duxbury M, Zinner MJ, Ashley SW, Whang EE: Glucose transporter-1 gene expression is associated with pancreatic cancer invasiveness and MMP-2 activity. Surgery 136: 548-556, 2004 Sakashita M, Aoyama N, Minami R, Maekawa S, Kuroda K, Shirasaka D, Ichihara T, Kuroda Y, Maeda S, Kasuga M: Glut1 expression in T1 and T2 stage colorectal carcinomas: its relationship to clinicopathological features. Eur J Cancer 37: 204- 209, 2001 Younes M, Juarez D, Lechago LV, Lerner SP: Glut 1 expression in transitional cell carcinoma of the urinary bladder is associated with poor patient survival. Anticancer Res 21: 575-578, 2001 Grover-McKay M, Walsh SA, Seftor EA, Thomas PA, Hendrix MJ: Role for glucose transporter 1 protein in human breast cancer. Pathol Oncol Res 4: 115-120, 1998 Airley R, Loncaster J, Davidson S, Bromley M, Roberts S, Patterson A, Hunter R, Stratford I, West C: Glucose transporter glut-1 expression correlates with tumor hypoxia and predicts metastasis-free survival in advanced carcinoma of the cervix. Clin Cancer Res 7: 928-934, 2001 Bos R, van der Groep P, Greijer AE, Shvarts A, Meijer S, Pinedo HM, Semenza GL, van Diest PJ, van der Wall E: Levels of hypoxia-inducible factor-1alpha independently predict prognosis in patients with lymph node negative breast carcinoma. Cancer 97: 1573-1581, 2003 Couvelard A, O’Toole D, Turley H, Leek R, Sauvanet A, Degott C, Ruszniewski P, Belghiti J, Harris AL, Gatter K, Pezzella F: Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumours: negative correlation of microvascular density and VEGF expression with tumour progression. Br J Cancer 92: 94-101, 2005 Garofalo C, Koda M, Cascio S, Sulkowska M, Kanczuga-Koda L, Golaszewska J, Russo A, Sulkowski S, Surmacz E: Increased expression of leptin and the leptin receptor as a marker of breast cancer progression: possible role of obesity-related stimuli. Clin Cancer Res 12: 1447-1453, 2006 Sivridis E, Giatromanolaki A, Gatter KC, Harris AL, Koukourakis MI; Tumor and Angiogenesis Research Group: Association of hypoxia-inducible factors 1alpha and 2alpha with activated angiogenic pathways and prognosis in patients with endometrial carcinoma. Cancer 95: 1055-1063, 2002 Yoo YG, Yeo MG, Kim DK, Park H, Lee MO: Novel function of orphan nuclear receptor Nur77 in stabilizing hypoxia-inducible factor-1alpha. J Biol Chem 279: 53365-53373, 2004 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 15 EP 20 PG 6 ER PT J AU Kiss, J Timar, J Somlai, B Gilde, K Fejos, Zs Gaudi, I Ladanyi, A AF Kiss, Judit Timar, Jozsef Somlai, Beata Gilde, Katalin Fejos, Zsuzsanna Gaudi, Istvan Ladanyi, Andrea TI Association of Microvessel Density with Infiltrating Cells in Human Cutaneous Malignant Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE angiogenesis; immunohistochemistry; lymphocytes; macrophages; microvessel density ID angiogenesis; immunohistochemistry; lymphocytes; macrophages; microvessel density AB Vascularization and host response to malignant tumors may have common molecular regulators, therefore, we analyzed the relationship between microvessel density (MVD) and tumor infiltrating cells in cutaneous malignant melanoma. Density of lymphocyte subpopulations, macrophages, dendritic cells and CD34+ microvessels was determined by immunohistochemistry in primary tumor samples from fifty-two patients with melanoma thicker than 1 mm. Intratumoral MVD did not show significant association with infiltration for any of these cell types. In the case of peritumoral reactive cell densities analyzed in the whole patient population, a positive correlation of MVD was found with CD3+ T cell density. This association was stronger in melanomas >4.0 mm and in visceral metastatic tumors. In these subgroups similar phenomenon was observed for CD8+ cells. We found significant correlation of MVD with CD68+ macrophage density only in the highest thickness category, and weak associations with B-cell and dendritic cell infiltration in visceral metastatic cases. MVD did not vary significantly in tumors categorized according to thickness, localization, ulceration or histological type. However, both intratumoral MVD and macrophage infiltration were significantly higher in male patients compared to females. The correlation of immune cell density with tumor vascularization and gender differences in vascularity and macrophage infiltration of melanoma deserve further attention. C1 [Kiss, Judit] National Institute of Oncology, Department of Tumor Progression, 7-9 Rath Gy. u., H-1122 Budapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, 7-9 Rath Gy. u., H-1122 Budapest, Hungary. [Somlai, Beata] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Gilde, Katalin] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Fejos, Zsuzsanna] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Gaudi, Istvan] National Institute of Oncology, National Cancer RegistryBudapest, Hungary. [Ladanyi, Andrea] National Institute of Oncology, Department of Tumor Progression, 7-9 Rath Gy. u., H-1122 Budapest, Hungary. RP Ladanyi, A (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. EM ladanyi@oncol.hu CR Arici A, Senturk LM, Seli E, Bahtiyar MO, Kim G: Regulation of monocyte chemotactic protein-1 expression in human endometrial stromal cells by estrogen and progesterone. Biol Reprod 61: 85-90, 1999 Arvilommi AM, Salmi M, Kalimo K, Jalkanen S: Lymphocyte binding to vascular endothelium in inflamed skin revisited: a central role for vascular adhesion protein-1, VAP-1). Eur J Immunol 26: 825-833, 1996 Ausprunk DH, Folkman J: Migration and proliferation of endothelial cells in performed and newly formed blood vessels during tumor angiogenesis. Microvasc Res 14: 53-65, 1977 Balch CM, Buzaid AC, Soong S-J, Atkins MB, Cascinelli N, Coit DG, Fleming ID, Gershenwald JE, Houghton A Jr, Kirkwood JM, McMasters KM, Mihm MF, Morton DL, Reintgen DS, Ross MI, Sober A, Thompson JA, Thompson JF: Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 19: 3635-3648, 2001 Barnhill RL, Fine JA, Roush GC, Berwick M: Predicting fiveyear outcome for patients with cutaneous melanoma in a population- based study. Cancer 78: 427-432, 1996 Barnhill RL, Levy MA: Regressing thin cutaneous malignant melanomas, < or = 1.0 mm, are associated with angiogenesis. Am J Pathol 143: 99-104, 1993 Brocker EB, Zwadlo G, Holzmann B, Macher E, Sorg C: Inflammatory cell infiltrates in human malignant melanoma at different stages of tumor progression. Int. J. Cancer 41: 562-567, 1988 Busam KJ, Berwick M, Blessing K, Fandrey K, Kang S, Karaoli T, Fine J, Cochran AJ, White WL, Rivers J: Tumor vascularity is not a prognostic factor for malignant melanoma of the skin. Am J Pathol 147: 1049-1056, 1995 Clark WH Jr, Elder DE, Guerry D 4th, Braitman LE, Trock BJ, Schultz D, Synnestvedt M, Halpern AC: Model predicting survival in stage I melanoma based on tumor progression. J Natl Cancer Inst 81: 1893-1904, 1989 Clemente CG, Mihm MC Jr, Bufalino R, Zurrida S, Collini P, Cascinelli N: Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer 77: 1303-1310, 1996 Colombel M, Filleur S, Fournier P, Merle C, Guglielmi J, Courtin A, Degeorges A, Serre CM, Bouvier R, Clezardin P, Cabon F: Androgens repress the expression of the angiogenesis inhibitor thrombospondin-1 in normal and neoplastic prostate. Cancer Res 65: 300-308, 2005 Coukos G, Benencia F, Buckanovich RJ, Conejo-Garcia JR: The role of dendritic cell precursors in tumour vasculogenesis. Br J Cancer 92: 1182-1187, 2005 Curiel TJ, Cheng P, Mottram P, Alvarez X, Moons L, Evdemon- Hogan M, Wei S, Zou L, Kryczek I, Hoyle G, Lackner A, Carmeliet P, Zou W: Dendritic cell subsets differentially regulate angiogenesis in human ovarian cancer. Cancer Res 64: 5535-5538, 2004 Davidson B, Goldberg I, Gotlieb WH, Lerner-Geva L, Ben- Baruch G, Agulansky L, Novikov I, Kopolovic J: Macrophage infiltration and angiogenesis in cervical squamous cell carcinoma – clinicopathologic correlation. Acta Obstet Gynecol Scand 78: 240-244, 1999 Depasquale I, Thompson WD: Microvessel density for melanoma prognosis. Histopathology 47: 186-194, 2005 Dobos J, Timar J, Bocsi J, Burian Z, Nagy K, Barna G, Petak I, Ladanyi A: In vitro and in vivo antitumor effect of 2-methoxyestradiol on human melanoma. Int J Cancer 112: 771-776, 2004 Dome J, Hendrix MJC, Paku S, Tovari J, Timar J: Alternative vascularization mechanisms in cancer: pathology and therapeutic implications. Am J Pathol, 170: 1-15, 2007 Dome B, Paku S, Somlai B, Timar J: Vascularization of cutaneous melanoma involves vessel co-option and has clinical significance. J Pathol 97: 355-362, 2002 Forster-Horvath C, Dome B, Paku S, Ladanyi A, Somlai B, Jalkanen S, Timar J: Loss of vascular adhesion protein-1 expression in intratumoral microvessels of human skin melanoma. Melanoma Res 14: 135-140, 2004 Fotsis T, Zhang Y, Pepper MS, Adlercreutz H, Montesano R, Nawroth PP, Schweigerer L: The endogenous oestrogen metabolite 2-methoxyoestradiol inhibits angiogenesis and suppresses tumour growth. Nature 368: 237-239, 1994 Freeman MR, Schneck FX, Gagnon ML, Corless C, Soker S, Niknejad K, Peoples GE, Klagsbrun M: Peripheral blood T lymphocytes and lymphocytes infiltrating human cancers express vascular endothelial growth factor: a potential role for T cells in angiogenesis. Cancer Res 55: 4140-4145, 1995 Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, Carbone DP: Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med 2: 1096-1103, 1996 Gross J, Azizkhan RG, Biswas C, Bruns RR, Hsieh DS, Folkman J: Inhibition of tumor growth, vascularization, and collagenolysis in the rabbit cornea by medroxyprogesterone. Proc Natl Acad Sci U S A 78: 1176-1180, 1981 Hanada T, Nakagawa M, Emoto A, Nomura T, Nasu N, Nomuray: Prognostic value of tumor-associated macrophage count in human bladder cancer. Int J Urol 7: 263-269, 2000 Ilmonen S, Kariniemi AL, Vlaykova T, Muhonen T, Pyrhonen S, Asko-Seljavaara S: Prognostic value of tumour vascularity in primary melanoma. Melanoma Res 9: 273-278, 1999 Inoshima N, Nakanishi Y, Minami T, Izumi M, Takayama K, Yoshino I, Hara N: The influence of dendritic cell infiltration and vascular endothelial growth factor expression on the prognosis of non-small cell lung cancer. Clin Cancer Res 8: 3480- 3486, 2002 Ishigami S, Aikou T, Natsugoe S, Hokita S, Iwashige H, Tokushige M, Sonoda S: Prognostic value of HLA-DR expression and dendritic cell infiltration in gastric cancer. Oncology 55: 65-69, 1998 Ishigami S, Natsugoe S, Matsumoto M, Okumura H, Sakita H, Nakashima S, Takao S, Aikou T: Clinical implications of intratumoral dendritic cell infiltration in esophageal squamous cell carcinoma. Oncol Rep 10: 1237-1240, 2003 Kashani-Sabet M, Sagebiel RW, Ferreira CM, Nosrati M, Miller JR 3rd: Tumor vascularity in the prognostic assessment of primary cutaneous melanoma. J Clin Oncol 20: 1826-1831, 2002 Kim-Schulze S, McGowan KA, Hubchak SC, Cid MC, Martin MB, Kleinman HK, Greene GL, Schnaper HW: Expression of an estrogen receptor by human coronary artery and umbilical vein endothelial cells. Circulation 94: 1402-1407, 1996 Koch AE, Polverini PJ, Kunkel SL, Harlow LA, DiPietro LA, Elner VM: Interleukin-8 as a macrophage-derived mediator of angiogenesis. Science 258: 1798-1801, 1992 Kono K, Salazar-Onfray F, Petersson M, Hansson J, Masucci G, Wasserman K, Nakazawa T, Anderson P, Kiessling R: Hydrogen peroxide secreted by tumor-derived macrophages down-modulates signal-transducing zeta molecules and inhibits tumor-specific T cell- and natural killer cell-mediated cytotoxicity. Eur J Immunol 26:1308-1313, 1996 Lackner C, Jukic Z, Tsybrovskyy O, Jatzko G, Wette V, Hoefler G, Klimpfinger M, Denk H, Zatloukal K: Prognostic relevance of tumour-associated macrophages and von Willebrand factorpositive microvessels in colorectal cancer. Virchows Arch 445: 160-167, 2004 Ladanyi A, Kiss J, Somlai B, Gilde K, Fejos Z, Mohos A, Gaudi I, Timar J: Density of DC-LAMP+ mature dendritic cells in combination with activated T lymphocytes infiltrating primary cutaneous melanoma is a strong independent prognostic factor. Cancer Immunol Immunother Epub DOI 10.1007/s00262-007- 0286-3, 2007 Ladanyi A, Somlai B, Gilde K, Fejos Z, Gaudi I, Timar J: T-cell activation marker expression on tumor-infiltrating lymphocytes as prognostic factor in cutaneous malignant melanoma. Clin Cancer Res 10:521-530, 2004 Leek RD, Harris AL, Lewis CE: Cytokine networks in solid human tumors: regulation of angiogenesis. J Leukoc Biol 56: 423-435, 1994 Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J, Harris AL: Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res 56: 4625-4629, 1996 Losordo DW, Isner JM: Estrogen and angiogenesis: A review. Arterioscler Thromb Vasc Biol 21: 6-12, 2001 Mabjeesh NJ, Willard MT, Frederickson CE, Zhong H, Simons JW: Androgens stimulate hypoxia-inducible factor 1 activation via autocrine loop of tyrosine kinase receptor/phosphatidylinositol 3’-kinase/protein kinase B in prostate cancer cells. Clin Cancer Res 9: 2416-2425, 2003 Madtes DK, Raines EW, Sakariassen KS, Assoian RK, Sporn MB, Bell GI: Induction of transforming growth factor-alpha in human alveolar macrophages. Cell 53: 285-293, 1988 Makitie T, Summanen P, Tarkkanen A, Kivela T: Tumor-infiltrating macrophages, CD68-positive cells, and prognosis in malignant uveal melanoma. Invest Ophthalmol Vis Sci 42: 1414-1421, 2001 Mantovani A, Bottazzi B, Colotta F, Sozzani S, Ruco L: The origin and function of tumor-associated macrophages. Immunol Today 13: 265-270, 1992 Marcoval J, Moreno A, Graells J, Vidal A, Escriba JM, Garcia- Ramirez M, Fabra A: Angiogenesis and malignant melanoma. Angiogenesis is related to the development of vertical, tumorigenic, growth phase. J Cutan Pathol 24: 212-218, 1997 Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S: Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Adv Immunol 74: 181- 273, 2000 McBride WH: Phenotype and functions of intratumoral macrophages. Biochim Biophys Acta 865: 27-41, 1986 McDonald DM, Foss AJE: Endothelial cells of tumor vessels: abnormal but not absent. Cancer Metastasis Rev 19: 109-120, 2002 Miescher S, Whiteside TL, Carrel S, von Fliedner V: Functional properties of tumor-infiltrating and blood lymphocytes in patients with solid tumors: effects of tumor cells and their supernatants on proliferative responses of lymphocytes. J Immunol 136: 1899-1907, 1986 Ono M, Torisu H, Fukushi J-I, Nishie A, Kuwano M: Biological implications of macrophage infiltration in human tumor angiogenesis. Cancer Chemother Pharmacol 43: 69-71, 1999 Peng SH, Deng H, Yang JF, Xie PP, Li C, Li, Feng DY: Significance and relationship between infiltrating inflammatory cell and tumor angiogenesis in hepatocellular carcinoma tissues. World J Gastroenterol 11: 6521-6524, 2005 Piras F, Colombari R, Minerba L, Murtas D, Floris C, Maxia C, Corbu A, Perra MT, Sirigu P: The predictive value of CD8, CD4, CD68, and human leukocyte antigen-D-related cells in the prognosis of cutaneous malignant melanoma with vertical growth phase. Cancer 104: 1246-1254, 2005 Prehn RT: The paradoxical association of regression with a poor prognosis in melanoma contrasted with a good prognosis in keratoacanthoma. Cancer Res 56: 937-940, 1996 Pribluda VS, Gubish ER Jr, Lavallee TM, Treston A, Swartz GM, Green SJ: 2-Methoxyestradiol: an endogenous antiangiogenic and antiproliferative drug candidate. Cancer Metastasis Rev 19: 173-179, 2000 Qin Z, Schwartzkopff J, Pradera F, Kammertoens T, Seliger B, Pircher H, Blankenstein T: A critical requirement of interferon gamma-mediated angiostasis for tumor rejection by CD8+ T cells. Cancer Res 63: 4095-4100, 2003 Ronan SG, Eng AM, Briele HA, Shioura NN, Das Gupta TK: Thin malignant melanomas with regression and metastases. Arch Dermatol 123: 1326-1330, 1987 Saito H, Tsujitani S, Ikeguchi M, Maeta M, Kaibara N: Relationship between the expression of vascular endothelial growth factor and the density of dendritic cells in gastric adenocarcinoma tissue. Br J Cancer 78: 1573-1577, 1998 Seli E, Pehlivan T, Selam B, Garcia-Velasco JA, Arici A: Estradiol down-regulates MCP-1 expression in human coronary artery endothelial cells. Fertil Steril 77: 542-547, 2002 Seo KH, Lee HS, Jung B, Ko HM, Choi JH, Park SJ, Choi IH, Lee HK, Im SY: Estrogen enhances angiogenesis through a pathway involving platelet-activating factor-mediated nuclear factor-kappaB activation. Cancer Res 64: 6482-6488, 2004 Spyridopoulos I, Sullivan AB, Kearney M, Isner JM, Losordo DW: Estrogen-receptor-mediated inhibition of human endothelial cell apoptosis. Estradiol as a survival factor. Circulation 95: 1505-1514, 1997 Straume O, Salvesen HB, Akslen LA: Angiogenesis is prognostically important in vertical growth phase melanomas. Int J Oncol 15: 595-599, 1999 Sunderkotter C, Steinbrink K, Goebeler M, Bhardwaj R, Sorg C: Macrophages and angiogenesis. J Leukoc Biol 55: 410-422, 1994 Torisu H, Ono M, Kiryu H, Furue M, Ohmoto Y, Nakayama J, Nishioka Y, Sone S, Kuwano M: Macrophage infiltration correlates with tumor stage and angiogenesis in human malignant melanoma: possible involvement of TNFalpha and IL-1alpha. Int J Cancer 85: 182-188, 2000 Varney ML, Johansson SL, Singh RK: Tumour-associated macrophage infiltration, neovascularization and aggressiveness in malignant melanoma: role of monocyte chemotactic protein- 1 and vascular endothelial growth factor-A. Melanoma Res 15: 417-425, 2005 Whiteside TL: Signaling defects in T lymphocytes of patients with malignancy. Cancer Immunol Immunother 48: 346-352, 1999 Zou W: Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer 5:263- 274, 2005 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 21 EP 31 PG 11 ER PT J AU Park, HR Park, YK AF Park, Hye-Rim Park, Yong-Koo TI Differential Expression of Runx2 and Indian Hedgehog in Cartilaginous Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Runx2; Indian hedgehog; cartilaginous tumors ID Runx2; Indian hedgehog; cartilaginous tumors AB Runx2-Cbfa1, a Runt transcription factor, plays important roles during skeletal development. In its absence, chondrocyte hypertrophy is severely impaired and there is no vascularization of cartilage templates during skeletal development. In addition, Indian hedgehog (Ihh) signaling molecules control the space and timing of chondrocyte differentiation. Our goal was to gain a better understanding of the molecular process underlying the development of chondrosarcoma and to investigate whether there is a biological difference among variable types of chondrosarcomas. To accomplish this we collected a series of 10 enchondromas and 57 chondrosarcomas (conventional, n = 17; mesenchymal, n = 20; clear cell, n = 20), and investigated the expression of Runx2 and Ihh in these cartilaginous tumors by immunohistochemistry. Cellular and matrix-rich areas were evaluated separately. Runx2 was expressed in 100% of conventional, mesenchymal, and clear cell chondrosarcomas, and in 30% of enchondromas. Higher levels of expression of Runx2 were found in cellular areas than in matrixrich areas. Expression levels increased with increasing histological grade in conventional chondrosarcoma, suggesting involvement in tumor progression. Ihh was expressed in 100% of conventional and clear cell chondrosarcomas, especially in matrix-rich areas. Mesenchymal chondrosarcomas revealed only focal expression of Ihh in matrix-rich areas. Small cell areas were negative. Ihh was absent or focally expressed in enchondromas. These findings demonstrate that Runx2 expression is active in variable chondrosarcomas compared to enchondromas, suggesting its importance in growth and differentiation of neoplastic cartilage. Ihh expression is considered a marker of the hypertrophic stage of differentiation in these tumor cells. C1 [Park, Hye-Rim] Hallym University, Department of PathologyAnyang, South Korea. [Park, Yong-Koo] Kyung Hee University, College of Medicine, Department of Pathology, #1 Hoeki-dong, Dongdaemun-ku, 130-702 Seoul, South Korea. RP Park, YK (reprint author), Kyung Hee University, College of Medicine, Department of Pathology, 130-702 Seoul, South Korea. EM ykpark@khmc.or.kr CR Bovee JVMG, Cleton-Jansen AM, Taminiau AHM, Hogendoorn PCW: Emerging pathways in the development of chondrosarcoma of bone and implications for targeted treatment. Lancet Oncol 6: 599-607, 2005 Kundu M, Javed A, Jeon JP, Horner A, Shum L, Eckhaus M, Muenk M, Lian JB, Yang Y, Nuckolls GH, Stein GS, Liu PP: Cbfβ interacts with Runx2 and has a critical role in bone development. Nat Genet 32: 639-644, 2002 Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi K, Shimizu Y, Bronson RT, Gao Y, Inada M, Sato M, Okamoto R, Kitamura Y, Yoshiki S, Kishimoto T: Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell 89: 755-764, 1997 Wang WF, Wang YG, Reginato AM, Glotzer DJ, Fukai N, Plotkina S, Karsenty G, Olsen BR: Groucho homologue Grg5 interacts with the transcription factor Runx2-Cbfa1 and modulates its activity during postnatal growth in mice. Dev Biol 270: 364- 381, 2004 Takeda S, Bonnamy JP, Owen MJ, Ducy P, Karsenty G: Continuous expression of Cbfa1 in nonhypertrophic chondrocytes uncovers its ability to induce hypertrophic chondrocyte differentiation and partially rescues Cbfa1-deficient mice. Genes Dev 15: 467-481, 2001 Yoshida CA, Yamamoto H, Fujita T, Furuichi T, Ito K, Inoue KI, Yamana K, Zanma A, Takada K, Ito Y, Komori T: Runx2 and Runx3 are essential for chondrocyte maturation, and Runx2 regulates limb growth through induction of Indian hedgehog. Genes Dev 18: 952-963, 2004 St-Jacques B, Hammerschmidt M, McMahon AP: Indian hedgehog signaling regulates proliferation and differentiation of chondrocytes and is essential for bone formation. Genes Dev 13: 2072-2086, 1999 Kim IS, Otto F, Zabel B, Mundlos S: Regulation of chondrocyte differentiation by Cbfa1. Mech Dev 80: 159-170, 1999 Komori T: Regulation of skeletal development by the Runx family of transcription factors. J Cell Biochem 95: 445-453, 2005 Alexandre C, Jacinto A, Ingham PW: Transcriptional activation of hedgehog target genes in Drosophila is mediated directly by the cubitus interruptus protein, a member of the GLI family of zinc finger DNA-binding proteins. Genes Dev 10: 2003-2013, 1996 Evans HL, Ayala AG, Romsdahl MM: Prognostic factors in chondrosarcoma of bone. A clinicopathologic analysis with emphasis on histologic grading. Cancer 40: 818-831, 1977 Papachristou DJ, Papachristou GJ, Papaefthimiou OA, Agnantis NJ, Basdra EK, Papavassiliou AG: The MAPK-AP- 1/-Runx2 signalling axes are implicated in chondrosarcoma pathobiology either independently or via up-regulation of VEGF. Histopathology 47: 565-574, 2005 Romeo S, Bovee JVMG, Jadnanansing NAA, Taminiau AHM, Hogendoorn PCW: Expression of cartilage growth plate signaling molecules in chondroblastoma. J Pathol 202: 113-120, 2004 Kronenberg HM: Developmental regulation of the growth plate. Nature 423: 332-336, 2003 Kindblom JM, Nilsson O, Hurme T, Ohlsson C, Savendahl L: Expression and localization of Indian hedgehog, Ihh, and parathyroid hormone related protein, PTHrP, in the human growth plate during pubertal development. J Endocrinol 174: R1-6, 2002 Karp SJ, Schipani E, St Jacques B, Hunzelman J, Kronenberg H, McMahon AP: Indian hedgehog coordinates endochondral bone growth and morphogenesis via parathyroid hormone related-protein-dependent and -independent pathways. Development 127: 543-548, 2000 Rozeman LB, Hameetman L, Cleton-Jansen AM, Taminiau AHM, Hogendoorn PCW, Bovee JVMG: Absence of IHH and retention of PTHrP signalling in enchondromas and central chondrosarcomas. J Pathol 205: 476-482, 2005 Tiet TD, Hopyan S, Nadesan P, Gokgoz N, Poon R, Lin AC, Yan T, Andrulis IL, Alman BA, Wunder JS: Constitutive hedgehog signaling in chondrosarcoma up-regulates tumor cell proliferation. Am J Pathol 168: 321-330, 2006 Aigner T: Towards a new understanding and classification of chondrogenic neoplasias of the skeleton – biochemistry and cell biology of chondrosarcoma and its variants. Virchows Arch 441: 219-230, 2002 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 32 EP 37 PG 6 ER PT J AU Arslantas, A Artan, S Oner, Muslumanoglu, HM Ozdemir, M Durmaz, R Arslantas, D Vural, M Cosan, E Atasoy, AM AF Arslantas, Ali Artan, Sevilhan Oner, Ulku Muslumanoglu, Hamza Mahmut Ozdemir, Muhsin Durmaz, Ramazan Arslantas, Didem Vural, Murat Cosan, Erhan Atasoy, Ant Metin TI Genomic Alterations in Low-Grade, Anaplastic Astrocytomas and Glioblastomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE anaplastic astrocytoma; comparative genomic hybridization; genomic imbalances; glioblastoma multiforme; low-grade astrocytoma ID anaplastic astrocytoma; comparative genomic hybridization; genomic imbalances; glioblastoma multiforme; low-grade astrocytoma AB To extend our understanding of potential stepwise genetic alterations that may underlie tumor progression from low-grade astrocytomas to glioblastomas, histopathologic and comparative genomic hybridization analyses were performed on tumor specimens from 68 primary lesions, including 40 glioblastomas, 10 anaplastic and 18 low-grade astrocytomas. The number of aberrations per case increased towards the higher grade tumors (grade II: 1.66±1.49; grade III: 2.80±1.68; grade IV: 3.02±1.07; F=6.955, p=0.002). A gain of 7/7q was common and the most frequently seen aberration in low-grade astrocytomas, whereas loss of 10q was the most frequently seen anomaly in anaplastic astrocytomas and glioblastomas. Chromosome 7p amplification was only detected in glioblastomas. Chromosome 10/10q deletion and combination of 1p, 19q and 17p deletions were specific to high-grade astrocytic tumors. Sequences of chromosome 7 and 10 seem to have pivotal roles in the biology of human gliomas. The genomic copy deletions of chromosomes 1p and 19q might provide an alternative mechanism in the genesis of astrocytomas. C1 [Arslantas, Ali] Medical Faculty of Osmangazi University, Department of Neurosurgery, Hasan Polatkan Bulvari No: 102/C, Ertav apt. D:9Eskisehir, Turkey. [Artan, Sevilhan] Medical Faculty, Osmangazi University, Department of Medical GeneticsEskisehir, Turkey. [Oner, Ulku] Eskisehir Osmangazi University, Faculty of Medicine, Department of PathologyEskisehir, Turkey. [Muslumanoglu, Hamza Mahmut] Medical Faculty, Osmangazi University, Department of Medical GeneticsEskisehir, Turkey. [Ozdemir, Muhsin] Medical Faculty, Osmangazi University, Department of Medical GeneticsEskisehir, Turkey. [Durmaz, Ramazan] Medical Faculty of Osmangazi University, Department of Neurosurgery, Hasan Polatkan Bulvari No: 102/C, Ertav apt. D:9Eskisehir, Turkey. [Arslantas, Didem] Medical Faculty, Osmangazi University, Department of Public HealthEskisehir, Turkey. [Vural, Murat] Medical Faculty of Osmangazi University, Department of Neurosurgery, Hasan Polatkan Bulvari No: 102/C, Ertav apt. D:9Eskisehir, Turkey. [Cosan, Erhan] Medical Faculty of Osmangazi University, Department of Neurosurgery, Hasan Polatkan Bulvari No: 102/C, Ertav apt. D:9Eskisehir, Turkey. [Atasoy, Ant Metin] Medical Faculty of Osmangazi University, Department of Neurosurgery, Hasan Polatkan Bulvari No: 102/C, Ertav apt. D:9Eskisehir, Turkey. RP Arslantas, A (reprint author), Medical Faculty of Osmangazi University, Department of Neurosurgery, Eskisehir, Turkey. EM aali@ogu.edu.tr;aarslanta@ogu.edu.tr CR Arslantas A, Artan S, Oner U, Durmaz R, Cosan E, Atasoy MA, Basaran N, Tel E: The importance of genomic copy number changes in the prognosis of glioblastoma multiforme. Neurosurg Rev 27: 58-64, 2004 Arslantas A, Artan S, Oner U, Durmaz R, Atasoy MA, Basaran N, Tel E: Detection of chromosomal imbalances in spinal meningiomas by comparative genomic hybridization. Neurol Medico- Chirur 43: 12-19, 2003 Arslantas A, Artan S, Oner U, Durmaz R, Atasoy MA, Basaran N, Tel E: Comparative genomic hybridization analysis of genomic alterations in benign, atypical and anaplastic meningiomas. Acta Neurol Bel 102: 53-62, 2002 Backlund LM, Nilsson BR, Liu L, Ichimura K, Collins VP: Mutations in Rb1 pathway-related genes are associated with poor prognosis in anaplastic astrocytomas. Br J Cancer 93: 124-130, 2005 Bigner SH, Matthews MR, Rasheed BK, Wiltshire RN, Friedman HS, Friedman AH, Stenzel TT, Dawes DM, McLendon RE, Bigner DD: Molecular genetic aspects of oligodendrogliomas including analysis by comparative genomic hybridization. Am J Pathol 155: 375-386, 1999 Cairncross JG, Ueki K, Zlatescu MC, Lisle DK, Finkelstein DM, Hammond RR, Silver JS, Stark PC, Macdonald DR, Ino Y, Ramsay DA, Louis DN: Specific genetic predictors of chemotherapeutic response and survival in patients with anaplastic oligodendrogliomas. J Natl Cancer Inst 90: 1473-1479, 1998 Durmaz R, Erken S, Arslantas A, Atasoy MA, Bal C, Tel E: Management of glioblastoma multiforme: with special reference to recurrence. Clin Neurol Neurosurg 99: 117-123, 1997 Ezaki T, Yanagisawa A, Ohta K, Aiso S, Watanabe M, Hibi T, Kato Y, Nakajima T, Ariyama T, Inazawa J, Nakamura Y, Horii A: Deletion mapping on chromosome 1p in well-differentiated gastric cancer. Br J Cancer 73: 424-428, 1996 Fan X, Aalto Y, Sanko SG, Knuutila S, Klatzmann D, Castresana JS: Genetic profile, PTEN mutation and therapeutic role of PTEN in glioblastomas. Int J Oncol 21: 1141-1150, 2002 Hirose Y, Aldape KD, Chang S, Lamborn K, Berger MS, Feuerstein BG: Grade II astrocytomas are subgrouped by chromosome aberrations. Cancer Genet Cytogenet 142:1-7, 2003 Inda MM, Fan X, Munoz J, Perot C, Fauvet D, Danglot G, Palacio A, Madero P, Zazpe I, Portillo E, Tunon T, Martinez-Penuela JM, Alfaro J, Eiras J, Bernheim A, Castresana JS: Chromosomal abnormalities in human glioblastomas: gain in chromosome 7p correlating with loss in chromosome 10q. Mol Carcinog 36: 6-14, 2003 Jen J, Harper JW, Bigner SH, Bigner DD, Papadopoulos N, Markowitz S, Willson JK, Kinzler KW, Vogelstein B: Deletion of p16 and p15 genes in brain tumors. Cancer Res 54: 6353-6358, 1994 Karlbom AE, James CD, Boethius J: Loss of heterozygosity in malignant gliomas involves at least three distinct regions on chromosome 10. Hum Genet 92: 169-174, 1993 Kitange G, Misra A, Law M, Passe S, Kollmeyer TM, Maurer M, Ballman K, Feuerstein BG, Jenkins RB: Chromosomal imbalances detected by array comparative genomic hybridization in human oligodendrogliomas and mixed oligoastrocytomas. Genes Chromosomes Cancer 42: 68-77, 2005 Kunwar S, Mohapatra G, Bollen A, Lamborn KR, Prados M, Feuerstein BG: Genetic subgroups of anaplastic astrocytomas correlate with patient age and survival. Cancer Res 61: 7683- 7688, 2001 Maruno M, Yoshimine T, Muhammad AK, Ninomiya H, Kato A, Hayakawa T: Chromosomal aberrations detected by comparative genomic hybridization, CGH, in human astrocytic tumors. Cancer Lett 135: 61-66, 1999 Mollenhauer J, Wiemann S, Scheurlen W, Korn B, Hayashi Y, Wilgenbus KK, von Deimling A, Poustka A: DMBT1, a new member of the SRCR superfamily, on chromosome 10q25.3-26.1 is deleted in malignant brain tumours. Nat Genet 17: 32-39, 1997 Nagai H, Pineau P, Tiollais P, Buendia MA, Dejean A: Comprehensive allelotyping of human hepatocellular carcinoma. Oncogene 14: 2927-2933, 1997 Nishizaki T, Kubota H, Harada K, Harada K, Ito H, Suzuki M, Sasaki K: Clinical evidence of distinct subgroups of astrocytic tumors defined by comparative genomic hybridization. Hum Pathol 31: 608-614, 2000 Nishizaki T, Ozaki S, Harada K, Ito H, Arai H, Beppu T, Sasaki K: Investigation of genetic alterations associated with the grade of astrocytic tumor by comparative genomic hybridization. Genes Chromosomes Cancer 21: 340-346, 1998 Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL, Burkhard C, Schuler D, Probst-Hensch NM, Maiorka PC, Baeza N, Pisani P, Yonekawa Y, Yasargil MG, Lutolf UM, Kleihues P: Genetic pathways to glioblastoma: a population study. Cancer Res 64: 6892-6899, 2004 Ohgaki H, Schauble B, zur Hausen A, von Ammon K, Kleihues P: Genetic alterations associated with the evolution and progression of astrocytic brain tumours. Virchows Arch 427:113-118, 1995 Sano T, Lin H, Chen X, Langford LA, Koul D, Bondy ML, Hess KR, Myers JN, Hong YK, Yung WK, Steck PA: Differential expression of MMAC/PTEN in glioblastoma multiforme: relationship to localization and prognosis. Cancer Res 59: 1820-1824, 1999 Schmidt MC, Antweiler S, Urban N, Mueller W, Kuklik A, Meyer- Puttlitz B, Wiestler OD, Louis DN, Fimmers R, von Deimling A: Impact of genotype and morphology on the prognosis of glioblastoma. Neuropathol Exp Neurol 61: 321-328, 2002 Schrock E, Thiel G, Lozanova T, du Manoir S, Meffert MC, Jauch A, Speicher MR, Nurnberg P, Vogel S, Janisch W: Comparative genomic hybridization of human malignant gliomas reveals multiple amplification sites and nonrandom chromosomal gains and losses. Am J Pathol 144: 1203-1218, 1994 Ushio Y, Tada K, Shiraishi S, Kamiryo T, Shinojima N, Kochi M, Saya H: Correlation of molecular genetic analysis of p53, MDM2, p126, PTEN and EGFR and survival of patients with anaplastic astrocytoma and glioblastoma. Front Biosci 8: 281- 288, 2003 von Deimling A, Bender B, Jahnke R, Waha A, Kraus J, Albrecht S, Wellenreuther R, Fassbender F, Nagel J, Menon AG: Loci associated with malignant progression in astrocytomas: a candidate on chromosome 19q. Cancer Res 54: 1397-1401, 1994 Wullich B, Sattler HP, Fischer U, Meese E: Two independent amplification events on chromosome 7 in glioma amplification of the epidermal growth factor receptor gene and amplification of the oncogene MET. Anticancer Res 15: 577-580, 1994 Zhou XP, Li YJ, Hoang-Xuan K, Laurent-Puig P, Mokhtari K, Longy M, Sanson M, Delattre JY, Thomas G, Hamelin R: Mutational analysis of the PTEN gene in gliomas: molecular and pathological correlations. Int J Cancer 84: 150-154, 1999 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 39 EP 46 PG 8 ER PT J AU Amirzargar, AA Khosravi, F Dianat, SS Alimoghadam, K Ghavamzadeh, F Ansaripour, B Moradi, B Nikbin, B AF Amirzargar, Ali-Akbar Khosravi, Farideh Dianat, S Saied Alimoghadam, Kamran Ghavamzadeh, Fereidoun Ansaripour, Bita Moradi, Batool Nikbin, Behrooz TI Association of HLA Class II Allele and Haplotype Frequencies with Chronic Myelogenous Leukemia and Age-at-Onset of the Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chronic myelogenous leukemia; genetic susceptibility; HLA-DRB; HLA-DQA1; HLA-DQB1 ID chronic myelogenous leukemia; genetic susceptibility; HLA-DRB; HLA-DQA1; HLA-DQB1 AB Chronic myelogenous leukemia (CML) is characterized by the presence of Philadelphia chromosome resulting from bcr/abl translocation. To clarify the association between HLA class II allele and haplotype frequencies in CML, 50 patients referred to Hematology Oncology and Bone Marrow Transplantation (BMT) center, Shariaty Hospital, Tehran, Iran, were randomly selected and compared with a group of 80 unrelated healthy blood donor subjects. HLA class II alleles were determined by PCR-SSP method. The results showed that the frequencies of DQB1*03011 (P=0.01) and DQA1*0505 (P=0.05) were higher, while that of DQB1*03032 (P=0.04) was lower in patients than in the controls. Regarding age-at-onset, the frequency of HLA-DRB1*07 (P=0.03) and -DQA1*0201 (P=0.03) alleles were higher in patients younger than 35 years. The most frequent haplotypes in our CML patients were HLA-DRB1*11/-DQB1*03011/-DQA1*0505 (P=0.01) and HLA-DRB1*04/-DQB1*0302/-DQA1*03011 (P=0.02). In conclusion, it is suggested that positive and negative association in certain HLA alleles and haplotypes exist in Iranian patients with CML. C1 [Amirzargar, Ali-Akbar] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. [Khosravi, Farideh] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. [Dianat, S Saied] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. [Alimoghadam, Kamran] Tehran University of Medical Sciences, Research Center, Shariati Hospital, Hematology, Oncology and Bone Marrow TransplantationTehran, Iran. [Ghavamzadeh, Fereidoun] Tehran University of Medical Sciences, Research Center, Shariati Hospital, Hematology, Oncology and Bone Marrow TransplantationTehran, Iran. [Ansaripour, Bita] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. [Moradi, Batool] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. [Nikbin, Behrooz] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. RP Amirzargar, AA (reprint author), Tehran University of Medical Sciences, Faculty of Medicine, Department of Immunology, Tehran, Iran. EM Amirzargar_ali@yahoo.com CR Amirzargar A, Mytilineos J, Farjadian S, et al: Human leukocyte antigen class II allele frequencies and haplotype association in Iranian normal population. Hum Immunol 62:1234-1238, 2001 Bortin MM, D’Amaro J, Bach FH, et al: HLA associations with leukemia. Blood 70: 227-232, 1987 ten Bosch GJA, Toornvliet AC, Friede T, et al: Recognition of peptides corresponding to the joining region of p210BCR-ABL protein by human T cells. Leukemia 9: 1344-1348, 1995 Caruso C, Lo Campo P, Botindari C, Modica MA: HLA antigens in Sicilian patients affected by chronic myelogenous leukaemia. J Immunogenet 14: 295-299, 1987 Dorak MT, Chalmers EA, Gaffney D, et al: Human major histocompatibility complex contains several leukemia susceptibility genes. Leuk Lymphoma 12: 211-222, 1994 Haldane JB: The estimation and significance of the logarithm of a ratio of frequency. Ann Hum Genet 20:309-311, 1956 Kurzrock R, Kantarjian HM, Druker BJ, et al: Philadelphia chromosome-positive leukemias: from basic mechanisms to molecular therapeutics. Ann Intern Med 138: 819-830, 2003 Miller SA, Dykes DD, Polesky HF: A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16:1215, 1988 Mundhada S, Luthra R, Cano P: Association of HLA Class I and Class II genes with bcr-abl transcripts in leukemia patients with t(9;22,, q34;q11). BMC Cancer 4: 25, 2004 Oguz FS, Kalayoglu S, Diler AS, et al: HLA system affects the age-at-onset in chronic myeloid leukemia. Am J Hematol 73: 256-262, 2003 Olerup O, Zetterquist H: HLA-DR typing by PCR amplification with sequence-specific primers, PCR-SSP, in 2 hours: an alternative to serological DR typing in clinical practice including donor-recipient matching in cadaveric transplantation. Tissue Antigens 39: 225-235, 1992 Pawelec G, Max H, Halder T, et al: BCR/ABL leukemia oncogene fusion peptides selectively bind to certain HLA-DR alleles and can be recognized by T cells found at low frequency in the repertoire of normal donors. Blood 88: 211-218, 1996 Posthuma EFM, Falkenburg JHF, Apperley JF, et al: HLA-DR4 is associated with a diminished risk of the development of chronic myeloid leukemia, CML). Leukemia 14: 859-862, 2000 Tian H, Zhou SY, Liu ZH, et al: Association of HLA-DPB1 alleles with chronic myelogenous leukemia in southern Chinese Hans. Zhongguo Shi Yan Xue Ye Xue Za Zhi 11: 266-268, 2003 Yasukawa M, Ohminami H, Kojima K, et al: Analysis of HLADRB1 alleles in Japanese patients with chronic myelogenous leukemia. Am J Hematol 63: 99-101, 2000 Yotnda P, Firat H, Garcia-Pons F, et al: Cytotoxic T cell response against the chimeric p210 BCR-ABL protein in patients with chronic myelogenous leukemia. J Clin Invest 101: 2290-2296, 1998 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 47 EP 51 PG 5 ER PT J AU Magyari, L Bene, J Komlosi, K Talian, G Farago, B Csongei, V Jaromi, L Safrany, E Sipeky, Cs Lakner, L Varga, M Gasztonyi, B Melegh, B AF Magyari, Lili Bene, Judit Komlosi, Katalin Talian, Gabor Farago, Bernadett Csongei, Veronika Jaromi, Luca Safrany, Eniko Sipeky, Csilla Lakner, Lilla Varga, Marta Gasztonyi, Beata Melegh, Bela TI Prevalence of SLC22A4 1672T and SLC22A5 -207C Combination Defined TC Haplotype in Hungarian Ulcerative Colitis Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ulcerative colitis; OCTN1; OCTN2; SLC22A4; SLC22A5; TC haplotype ID ulcerative colitis; OCTN1; OCTN2; SLC22A4; SLC22A5; TC haplotype AB Ulcerative colitis (UC) is a chronic inflammatory disease of the gastrointestinal tract. The aim of this study was to verify the prevalence rate of the haplotype called TC, determined by combination of two functional alleles of OCTN cation transporter genes (SLC22A4 1672T and SLC22A5 -207C combination variants) in ulcerative colitis patients and unrelated healthy controls. The ''TC haplotype” has recently been suggested to confer risk for UC. A total of 121 unrelated Hungarian subjects with UC and 110 matched controls were genotyped for the two single nucleotide polymorphisms. The genotypes were determined by using PCR/RFLP assay and direct sequencing. The SLC22A4 1672T allele frequency was 46.7% in the patients with UC and 46.4% in the controls, whereas the SLC22A5 -207C allele occurred in 48.8% of the patients and 51.4% of the controls. The prevalence of the TC haplotype was 19% in the patient group and 22.7% in controls. Since there was no accumulation of the TC haplotype in the patient group, our observation suggests that carrying the TC haplotype is not associated with a higher risk for UC in the Hungarian population. C1 [Magyari, Lili] University of Pecs, Department of Medical Genetics and Child Development, Szigeti ut 12, H-7624 Pecs, Hungary. [Bene, Judit] University of Pecs, Department of Medical Genetics and Child Development, Szigeti ut 12, H-7624 Pecs, Hungary. [Komlosi, Katalin] University of Pecs, Department of Medical Genetics and Child Development, Szigeti ut 12, H-7624 Pecs, Hungary. [Talian, Gabor] University of Pecs, Department of Medical Genetics and Child Development, Szigeti ut 12, H-7624 Pecs, Hungary. [Farago, Bernadett] University of Pecs, Department of Medical Genetics and Child Development, Szigeti ut 12, H-7624 Pecs, Hungary. [Csongei, Veronika] University of Pecs, Department of Medical Genetics and Child Development, Szigeti ut 12, H-7624 Pecs, Hungary. [Jaromi, Luca] University of Pecs, Department of Medical Genetics and Child Development, Szigeti ut 12, H-7624 Pecs, Hungary. [Safrany, Eniko] University of Pecs, Department of Medical Genetics and Child Development, Szigeti ut 12, H-7624 Pecs, Hungary. [Sipeky, Csilla] University of Pecs, Department of Medical Genetics and Child Development, Szigeti ut 12, H-7624 Pecs, Hungary. [Lakner, Lilla] Markusovszky Hospital, Department of Medicine and GastroenterologySzombathely, Hungary. [Varga, Marta] Rethy Pal Hospital, 3rd Department of Medicine and GastroenterologyBekescsaba, Hungary. [Gasztonyi, Beata] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Melegh, Bela] University of Pecs, Department of Medical Genetics and Child Development, Szigeti ut 12, H-7624 Pecs, Hungary. CR Babusukumar U, Wang T, McGuire E, et al: Contribution of OCTN variants within the IBD5 locus to pediatric onset Crohn's disease. Am J Gastroenterol 101:1354-1361, 2006 Bene J, Komlosi K, Havasi V, et al: Changes of plasma fasting carnitine ester profile in patients with ulcerative colitis. World J Gastroenterol 12:110-113, 2006 Bene J, Magyari L, Talian G, et al: Prevalence of SLC22A4, SLC22A5 and CARD15 gene mutations in Hungarian pediatric patients with Crohn's disease. World J Gastroenterol 12:5550- 5553, 2006 Bonen DK, Cho JH: The genetics of inflammatory bowel disease. Gastroenterology 124:521-536, 2003 Danese S, Fiocchi C: Etiopathogenesis of inflammatory bowel diseases. World J Gastroenterol 12:4807-4812, 2006 Eaden JA, Abrams KR, Mayberry JF: The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 48:526-535, 2001 Gazouli M, Mantzaris G, Archimandritis AJ, et al: Single nucleotide polymorphisms of OCTN1, OCTN2, and DLG5 genes in Greek patients with Crohn's disease. World J Gastroenterol 11:7525-7530, 2005 Giallourakis C, Stoll M, Miller K, et al: IBD5 is a general risk factor for inflammatory bowel disease: replication of association with Crohn disease and identification of a novel association with ulcerative colitis. Am J Hum Genet 73:205-211, 2003 Jantchou P, Monnet E, Carbonnel F: [Environmental risk factors in Crohn's disease and ulcerative colitis, excluding tobacco and appendicectomy)], in French). Gastroenterol Clin Biol 30:859-867, 2006 Leung E, Hong J, Fraser AG, et al: Polymorphisms in the organic cation transporter genes SLC22A4 and SLC22A5 and Crohn's disease in a New Zealand Caucasian cohort. Immunol Cell Biol 84:233-236, 2006 Machida H, Tsukamoto K, Wen CY, et al: Association of polymorphic alleles of CTLA4 with inflammatory bowel disease in the Japanese. World J Gastroenterol 11:4188-4193, 2005 Mahid SS, Minor KS, Soto RE, et al: Smoking and inflammatory bowel disease: a meta-analysis. Mayo Clin Proc 81:1462- 1471, 2006 Mahida YR, Rolfe VE: Host-bacterial interactions in inflammatory bowel disease. Clin Sci, Lond, 107:331-341, 2004 Martinez A, Del Carmen MM, Mendoza JL, et al: Association of the organic cation transporter OCTN genes with Crohn's disease in the Spanish population. Eur J Hum Genet 14:222- 226, 2006 Medici V, Mascheretti S, Croucher PJ, et al: Extreme heterogeneity in CARD15 and DLG5 Crohn disease-associated polymorphisms between German and Norwegian populations. Eur J Hum Genet 14:459-468, 2006 Newman B, Gu X, Wintle R, et al: A risk haplotype in the Solute Carrier Family 22A4/22A5 gene cluster influences phenotypic expression of Crohn's disease. Gastroenterology 128:260-269, 2005 Noble CL, Nimmo ER, Drummond H, et al: The contribution of OCTN1/2 variants within the IBD5 locus to disease susceptibility and severity in Crohn's disease. Gastroenterology 129:1854- 1864, 2005 Palmieri O, Latiano A, Valvano R, et al: Variants of OCTN1-2 cation transporter genes are associated with both Crohn's disease and ulcerative colitis. Aliment Pharmacol Ther 23:497-506, 2006 Peltekova VD, Wintle RF, Rubin LA, et al: Functional variants of OCTN cation transporter genes are associated with Crohn disease. Nat Genet 36:471-475, 2004 Pena AS: Contribution of genetics to a new vision in the understanding of inflammatory bowel disease. World J Gastroenterol 12:4784-4787, 2006 Russell RK, Drummond HE, Nimmo ER, et al: Analysis of the influence of OCTN1/2 variants within the IBD5 locus on disease susceptibility and growth indices in early onset inflammatory bowel disease. Gut 55:1114-1123, 2006 Satsangi J, Jewell D, Parkes M, et al: Genetics of inflammatory bowel disease. A personal view on progress and prospects. Dig Dis 16:370-374, 1998 Silverberg MS: OCTNs: will the real IBD5 gene please stand up? World J Gastroenterol 12:3678-3681, 2006 Siminovitch KA: Advances in the molecular dissection of inflammatory bowel disease. Semin Immunol 18:244-253, 2006 Taylor KD, Yang H, Rotter JI: Inflammatory bowel disease. II. Gene mapping. Mol Genet Metab 74:22-44, 2001 Torkvist L, Noble CL, Lordal M, et al: The contribution of OCTN1/2 variants within the IBD5 locus to Crohn's disease in the Swedish population. Gastroenterology 128:A449, 2005 Torok HP, Glas J, Tonenchi L, et al: Polymorphisms in the DLG5 and OCTN cation transporter genes in Crohn's disease. Gut 54:1421-1427, 2005 Tosa M, Negoro K, Kinouchi Y, et al: Lack of association between IBD5 and Crohn's disease in Japanese patients demonstrates population-specific differences in inflammatory bowel disease. Scand J Gastroenterol 41:48-53, 2006 Urcelay E, Mendoza JL, Martinez A, et al: IBD5 polymorphisms in inflammatory bowel disease: association with response to infliximab. World J Gastroenterol 11:1187-1192, 2005 Vermeire S, Pierik M, Hlavaty T, et al: Association of organic cation transporter risk haplotype with perianal penetrating Crohn's disease but not with susceptibility to IBD. Gastroenterology 129:1845-1853, 2005 Waller S, Tremelling M, Bredin F, et al: Evidence for association of OCTN genes and IBD5 with ulcerative colitis. Gut 55:809-814, 2006 Yang H, Taylor KD, Rotter JI: Inflammatory bowel disease. I. Genetic epidemiology. Mol Genet Metab 74:1-21, 2001 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 53 EP 56 PG 4 ER PT J AU Renyi, I Bardi, E Udvardi, E Kovacs, G Bartyik, K Kajtar, P Masat, P Nagy, K Galantai, I Kiss, Cs AF Renyi, Imre Bardi, Edit Udvardi, Erzsebet Kovacs, Gabor Bartyik, Katalin Kajtar, Pal Masat, Peter Nagy, Kalman Galantai, Ilona Kiss, Csongor TI Prevention and Treatment of Hyperuricemia with Rasburicase in Children with Leukemia and Non-Hodgkin’s Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE rasburicase; uric acid; tumor lysis syndrome; leukemia; lymphoma ID rasburicase; uric acid; tumor lysis syndrome; leukemia; lymphoma AB To prevent acute renal failure in children at risk for developing tumor lysis syndrome due to acute lymphoblastic leukemia or non-Hodgkin’s lymphoma treated according to international BFM protocols, we investigated recombinant urate oxidase (rasburicase) in the first Central European openlabeled, prospective, multicenter phase IV trial. Rasburicase was administered intravenously, at 0.2 mg/kg for 5 consecutive days to 36 patients. Blood levels of uric acid, creatinine, phosphorus, calcium, lactate dehydrogenase and complete blood count were measured daily during rasburicase treatment and on days 6, 7 and 12. Initial uric acid level decreased significantly by 4 hours (from 343 micromol/L to 58 micromol/L, p<0.001), except for one steroid-resistant patient who required hemodialysis on day 14 after having introduced combined cytostatic treatment. Comparing the data of a subgroup of 12 patients receiving rasburicase with that of a historic cohort of 14 patients treated with allopurinol indicated the superiority of rasburicase over allopurinol in prophylaxis and treatment of hyperuricemia in children with leukemia and lymphoma. C1 [Renyi, Imre] Semmelweis University, 1st Department of PediatricsBudapest, Hungary. [Bardi, Edit] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98. Nagyerdei Circle, H-4012 Debrecen, Hungary. [Udvardi, Erzsebet] Sanofi-Synthelabo Co.Budapest, Hungary. [Kovacs, Gabor] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Bartyik, Katalin] University of Szeged, Department of PediatricsSzeged, Hungary. [Kajtar, Pal] University of Pecs, Department of PediatricsPecs, Hungary. [Masat, Peter] Markusovszky Vas Country Hospital, Department of PediatricsSzombathely, Hungary. [Nagy, Kalman] BAZ Megyei Korhaz, GyermekosztalyMiskolc, Hungary. [Galantai, Ilona] Madarasz Street Pediatric HospitalBudapest, Hungary. [Kiss, Csongor] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98. Nagyerdei Circle, H-4012 Debrecen, Hungary. RP Bardi, E (reprint author), Medical and Health Science Center, University of Debrecen, Department of Pediatrics, H-4012 Debrecen, Hungary. EM editbardi@hotmail.com CR Goldman SC, Holcenberg JS, Finkleinstein JZ, et al: A randomized comparison between rasburicase and allopurinol in children with lymphoma or leukemia at high risk for tumor lysis. Blood 97: 2998-3003, 2001 Pui CH, Relling MV, Lascombes F, et al: Urate oxidase in prevention and treatment of hyperuricemia associated with lymphoid malignancies. Leukemia 11: 1813-1816, 1997 Pui CH, Mahmoud HH, Wiley JM, et al: Recombinant urate oxidase for the prophylaxis or treatment of hyperuricemia in patients with leukemia or lymphoma. J Clin Oncol 19: 697- 704, 2001 Bosly A, Sonet A, Pinkerton CR, et al: Rasburicase, recombinant urate oxidase, for the management hyperuricemia in patients with cancer: report of an international compassionate use study. Cancer 98: 1048-1054, 2003 Pession A, Barbieri E: Treatment and prevention of tumor lysis syndrome in children. Experience of Associazione Italiana Ematologia Oncologia Pediatrica. Contrib Nephrol 147:80-92, 2005 Schrappe M, Reiter A, Zimmermann M, et al: Long-term results of four consecutive trials in childhood ALL performed by the ALL-BFM study group from 1981 to 1995, Berlin-Frankfurt- Munster. Leukemia 14: 2205-2222, 2000 Reiter A, Schrappe M, Parwaresch R, et al: Non-Hodgkin’s lymphomas of childhood and adolescence: results of a treatment stratified for biologic subtypes and stage – a report of the Berlin-Frankfurt-Munster Group. Clin Oncol 113: 359-372, 1995 Masera G, Jankovic M, Zurlo MG, et al: Urate-oxidase prophylaxis of uric acid-induced renal damage in childhood leukemia. J Pediatr 7: 202-204, 1985 Cohen LF, Balow JE, Magrath IT, et al: Acute tumor lysis syndrome: A review of 37 patients with Burkitt lymphoma. Am J Med 68: 486-491, 1980 Liu CY, Sims-McCallum RP, Schiffer CA: A single dose of rasburicase is sufficient for the treatment of hyperuricemia in patients receiving chemotherapy. Leuk Res 29: 463-465, 2005 Annemans L, Moeremans K, Lamotte M, et al: Pan-European multicentre economic evaluation of recombinant urate oxidase, Rasburicase, in prevention and treatment of hyperuricemia and tumor lysis syndrome in haematological cancer patients. Support Care Cancer 11: 249-257, 2003 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 57 EP 62 PG 6 ER PT J AU Vegso, Gy Toth, M Hidvegi, M Toronyi, Langer, MR Dinya, E Toth, A Perner, F Jaray, J AF Vegso, Gyula Toth, Maria Hidvegi, Marta Toronyi, Eva Langer, M Robert Dinya, Elek Toth, Andras Perner, Ferenc Jaray, Jeno TI Malignancies after Renal Transplantation during 33 Years at a Single Center SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunosuppression; kidney transplantation; posttransplant malignancies; tumor risk; waiting list ID immunosuppression; kidney transplantation; posttransplant malignancies; tumor risk; waiting list AB This study provides an analysis of incidence and characteristics of malignant tumors of 2535 patients who underwent renal transplantation between 1973 and 2007 at the Transplantation Center in Budapest. One hundred ninety-three malignant diseases were found in 188 patients (7.6%). The incidence of thyroid-, renal- hepatic-, skin- and gastric cancers as well as of Kaposi sarcoma and lymphomas increased in our transplant patient cohort compared to the figures of the general population based on the data of our Cancer Registry. On the other hand, colorectal-, oralprostate and lung cancers were underrepresented in our patient cohort. The mean time of diagnosis of malignancies following kidney transplantation was 58.5±44.8 months. One fifth of the tumors were detected within the first year. Patients with malignancies were distributed into four groups based on the immunosuppressive regimen: group I (8.5%), azathioprine + prednisone; group II (59.0%), cyclosporine + prednisone; group III (26.6%), cyclosporine + mycophenolate mofetil + prednisone; group IV (5.9%), tacrolimus + mycophenolate mofetil + prednisone. The mean age of patients was 47.3, 53.5, 55.5 and 58.1 years in group I, II, III and IV, respectively. Oncologic and immunosuppressive therapy was decided individually. Immunosuppression was switched to rapamycin-containing regimens in 63 cases. We lost 92 patients (48.9%) with a mean survival time of 25.8±39.4 months. Cumulative 1- and 5-year survivals were 69.5% and 52%, respectively. The increasing number of cancers seen early after transplantation and the increased risk of developing a cancer due to the older age of recipients draw attention to the importance of regular oncologic screening in patients on the waiting list and after transplantation. C1 [Vegso, Gyula] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Toth, Maria] EGIS Pharmaceuticals PLC, Medical DepartmentBudapest, Hungary. [Hidvegi, Marta] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Toronyi, Eva] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Langer, M Robert] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Dinya, Elek] EGIS Pharmaceuticals PLC, Medical DepartmentBudapest, Hungary. [Toth, Andras] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Perner, Ferenc] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Jaray, Jeno] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. RP Vegso, Gy (reprint author), Semmelweis University, Department of Transplantation and Surgery, H-1082 Budapest, Hungary. EM vegso@trans.sote.hu CR Buell JF, Gross TG, Woodle ES: Malignancy after transplantation. Transplantation 80: S254-264, 2005 Kasiske BL, Snyder JJ, Gilbertson DT, Wang C: Cancer after kidney transplantation in the United States. Am J Transplant 4: 905-913, 2004 Adami J, Gabel H, Lindelof B, Ekstrom K, Rydh B, Glimelius B, Ekbom A, Adami HO, Granath F: Cancer risk following organ transplantation: a nationwide cohort study in Sweden. Br J Cancer 89: 1221-1227, 2003 Montagnino G, Lorca E, Tarantino A, Bencini P, Aroldi A, Cesana B, Braga M, Lonati F, Ponticelli C: Cancer incidence in 854 kidney transplant recipients from a single institution: comparison with normal population and with patients under dialytic treatment. Clin Transpl 10: 461-469, 1996 Lutz J, Heemann U: Tumors after kidney transplantation. Curr Opin Urol 13: 105-109, 2003 Penn I: Cancers in renal transplant recipients. Adv Ren Replace Ther 7: 147-156, 2000 Penn I: Occurrence of cancers in immunosuppressed organ transplant recipients. Clin Transpl 12: 147-158, 1998 Kauffman HM, Cherikh WS, McBride MA, Cheng Y, Hanto DW: Post-transplant de novo malignancies in renal transplant recipients: the past and present. Transpl Int 19: 607-620, 2006 Desoize B: Immunosuppressive agents are also carcinogens. Crit Rev Oncol Hematol 56: 1-4, 2005 Caillard S, Dharnidharka V, Agodoa L, Bohen E, Abbott K: Posttransplant lymphoproliferative disorders after renal transplantation in the United States in era of modern immunosuppression. Transplantation 80: 1233-1243, 2005 Opelz G, Dohler B: Lymphomas after solid organ transplantation: A collaborative transplant study report. Am J Transplant 4: 222-230, 2004 Taylor AL, Marcus R, Bradley JA: Post-transplant lymphoproliferative disorders, PTLD, after solid organ transplantation. Crit Rev Oncol Hematol 56: 155-167, 2005 Mathew T, Kreis H, Friend P: Two-year incidence of malignancy in sirolimus-treated renal transplant recipients: results from five multicenter studies. Clin Transpl 18: 446-449, 2004 Otto S, Kasler M: Trends in cancer mortality and morbidity in Hungarian and international statistics. Characteristics and potential outcome of public health screening programmes., In Hungarian, Hungarian Oncology 49:99-107, 2005 Penn I: Transmission of cancer from organ donors. Ann Transplant 2: 7-12, 1997 Kinlen LJ, Eastwood JB, Kerr DN, Moorhead JF, Oliver DO, Robinson BH, de Wardener HE, Wing AJ: Cancer in patients receiving dialysis. Br Med J 280: 1401-1403, 1980 Fischereder M, Jauch KW: Prevalence of cancer history prior to renal transplantation. Transpl Int 18: 779-784, 2005 Kahan BD, Knight R, Schoenberg L, Pobielski J, Kerman RH, Mahalati K, Yakupoglu Y, Aki FT, Katz S, Van Buren CT: Ten years of sirolimus therapy for human renal transplantation: the University of Texas at Houston experience. Transplant Proc 35: 25S-34S, 2003 Yakupoglu YK, Buell JF, Woodle S, Kahan BD: Individualization of immunosuppressive therapy. III. Sirolimus associated with a reduced incidence of malignancy. Transplant Proc 38: 358-361, 2006 Taylor AL, Watson CJ, Bradley JA: Immunosuppressive agents in solid organ transplantation: Mechanisms of action and therapeutic efficacy. Crit Rev Oncol Hematol 56: 23-46, 2005 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 63 EP 69 PG 7 ER PT J AU Atmaca, FA Akbulut, Z Demirci, A Belenli, O Alici, S Balbay, MD AF Atmaca, Fuat Ali Akbulut, Ziya Demirci, Alparslan Belenli, Olcay Alici, Suleyman Balbay, M Derya TI Multiple Subcutaneous Nodular Metastases from Transitional Cell Carcinoma of the Bladder SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE bladder cancer; skin metastasis; transitional cell carcinoma ID bladder cancer; skin metastasis; transitional cell carcinoma AB Skin metastasis from transitional cell carcinoma (TCC) of the bladder is rare. In this report an uncommon metastasis of TCC of the bladder is presented. C1 [Atmaca, Fuat Ali] Ankara Ataturk Training and Research Hospital, 1st Urology Clinic, 475. sokak 20/20 Cukurambar, 06520 Ankara, Turkey. [Akbulut, Ziya] Ankara Ataturk Training and Research Hospital, 1st Urology Clinic, 475. sokak 20/20 Cukurambar, 06520 Ankara, Turkey. [Demirci, Alparslan] Ankara Ataturk Training and Research Hospital, 1st Urology Clinic, 475. sokak 20/20 Cukurambar, 06520 Ankara, Turkey. [Belenli, Olcay] Ankara Ataturk Training and Research Hospital, 1st Pathology ClinicAnkara, Turkey. [Alici, Suleyman] Ankara Ataturk Training and Research Hospital, Medical Oncology ClinicAnkara, Turkey. [Balbay, M Derya] Ankara Ataturk Training and Research Hospital, 1st Urology Clinic, 475. sokak 20/20 Cukurambar, 06520 Ankara, Turkey. RP Atmaca, FA (reprint author), Ankara Ataturk Training and Research Hospital, 1st Urology Clinic, 06520 Ankara, Turkey. EM alifuatatmaca@yahoo.com CR Akman Y, Cam K, Kavak A, Alper M: Extensive cutaneous metastasis of transitional cell carcinoma of the bladder. Int J Urol 10:103-104, 2003 Brownstein MH, Helwig EB: Spread of tumors to the skin. Arch Dermatol 107:80-86, 1973 Freeman JR, Esrig DE, Stein JP, Simoneau AR, Skinner EC, Chen SC, Groshen S, Lieskovsky G, Boyd SD, Skinner DGl: Radical cystectomy for high risk patients with superficial bladder cancer in the era of orthotopic urinary reconstruction. Cancer 76: 833-839, 1995 Fujita K, Sakamoto Y, Fujime M, Kitagawa R: Two cases of inflammatory skin metastasis from transitional cell carcinoma of the urinary bladder. Urol Int 53:114-116, 1994 Kumar PV, Salimi B, Musallaye A, Tadayyon A: Subcutaneous metastasis from transitional cell carcinoma of the bladder diagnosed by fine needle aspiration biopsy. Acta Cytol 44: 657- 660, 2000 Mueller TJ, Wu H, Greenberg RE, Hudes G, Topham N, Lessin SR, Uzzo RG: Cutaneous metastases from genitourinary malignancies. Urology 63: 1021-1026, 2004 Rebelakos A, Manthopoulos A, Hadjissotiriou GG: Unusual metastasis of bladder cancer. Br J Urol 64: 198, 1989 Rosen T: Cutaneous metastases. Med Clin North Am 64: 885- 900, 1980 Specter JS, Zimbler H, Deleo M, Ross JS: Skin metastases from transitional cell bladder cancer. Urology 29: 215-217, 1987 Wyldes MP, Osborn DE: Solitary cutaneous metastasis from transitional cell carcinoma of the bladder. Br J Urol 61:164, 1988 Serel TA, Ozdemir G, Yaman LS, Ozdemir H: Skin metastasis of bladder carcinoma. A case report. Mater Med Pol 26:113-114, 1994 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 70 EP 72 PG 3 ER PT J AU Kojima, M Matsumoto, M Miyazawa, Y Shimizu, K Itoh, H Masawa, N AF Kojima, Masaru Matsumoto, Morio Miyazawa, Yuri Shimizu, Kazuhiko Itoh, Hideaki Masawa, Nobuhide TI Follicular Lymphoma with Prominent Sclerosis (''Sclerosing Variant of Follicular Lymphoma'') Exhibiting a Mesenteric Bulky Mass Resembling Inflammatory Pseudotumor. Report of Three Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE follicular lymphoma; sclerosis; mesenterium; inflammatory pseudotumor; immunohistochemistry ID follicular lymphoma; sclerosis; mesenterium; inflammatory pseudotumor; immunohistochemistry AB We present three cases of follicular lymphoma (FL) exhibiting prominent sclerosis (sclerosing variant of follicular lymphoma), resembling inflammatory pseudotumor (IPT) of the lymph node, arising from mesenteric lymph node. Clinically all three cases represented bulky masses of the mesenteric lymph node. Histologically, the lesions were characterized by neoplastic lymphoid follicles separated by stromal collagenization and sclerotic process, with cellular infiltrate extending into the adjacent adipose tissue. The lesions contained variable cellular spindle cell proliferation and inflammatory infiltrate including numerous reactive T cells and histiocytes. Small capillary proliferation with vascular change was also noted. Immunohistochemical study demonstrated the myofibroblastic nature of the spindle cells. Moreover, neoplastic follicles were composed of intermediate to medium-sized lymphocytes, somewhat resembling reactive lymphoid aggregates. The overall histomorphological findings of the three lesions were similar to those of IPT of the lymph node. However, CD10, Bcl-2 and Bcl-6 immunostaining demonstrated the neoplastic nature of the lymphoid follicles and the lesions were diagnosed as FL grade 1. The present three cases indicate that the sclerosing variant of grade 1 FL should be added to the differential diagnosis from IPT of the lymph node. C1 [Kojima, Masaru] Gunma Cancer Center Hospital, Department of Pathology and Clinical Laboratories, 617-1, Takabayashinishi-cho, 373-8550 Ohta, Japan. [Matsumoto, Morio] National Nishigunma Hospital, Department of HematologyShibukawa, Japan. [Miyazawa, Yuri] National Nishigunma Hospital, Department of HematologyShibukawa, Japan. [Shimizu, Kazuhiko] Ashikaga Red Cross Hospital, Department of Pathology and Clinical LaboratoriesAshikaga, Japan. [Itoh, Hideaki] Maebashi Red Cross Hospital, Department of Pathology and Clinical LaboratoriesMaebashi, Japan. [Masawa, Nobuhide] Dokkyo Medical University School of Medicine, Department of Diagnostic and Anatomic PathologyMibu, Japan. RP Kojima, M (reprint author), Gunma Cancer Center Hospital, Department of Pathology and Clinical Laboratories, 373-8550 Ohta, Japan. EM mkojima@gunma-cc.jp CR Spencer H: The pulmonary plasma cell/histiocytoma complex. Histopathology 8:903-916,1984 Coffin CM, Watterson J, Priest JR, Dehner LP: Extrapulmonary inflammatory myofibroblastic tumor, inflammatory pseudotumor). A clinicopathologic and immunohistochemical study of 84 cases. Am J Surg Pathol 19:859-872, 1995 Perrone T, De Wolf-Peeters C, Frizzera G: Inflammatory pseudotumor of lymph nodes: A distinct pattern of nodal reaction. Am J Surg Pathol 12:351-361, 1988 Facchetti F, De Wolf Peeters C, De Wever I, Frizzera G: Inflammatory pseudotumor of lymph nodes. Immunohistochemical evidence for its fibrohistiocytic nature. Am J Pathol 137: 281- 289, 1990 Davis RE, Warnke RA, Dorfman RF: Inflammatory pseudotumor of lymph nodes. Additional observations and evidence for an inflammatory etiology. Am J Surg Pathol 15:744-756, 1991 Moran CA, Suster S, Abbondanzo SL: Inflammatory pseudotumor of lymph nodes: a study of 25 cases with emphasis on morphological heterogeneity. Hum Pathol 28:332-338, 1997 Kojima M, Nakamura S, Shimizu K, Hosomura Y, Ohno Y, Itoh H, Yamane N, Yoshida K, Masawa N: Inflammatory pseudotumor of the lymph nodes. Clinicopathologic and immunohistological study of 11 Japanese cases. Int J Surg Pathol 9:207- 214, 2001 Warnke RF, Weiss LM, Chan JKC, Clearre ML, Dorfman RF: Tumor of the lymph nodes and spleen, Atlas of Tumor Pathology, 3rd series, Fascicle 14). Armed Forces Institute of Pathology, Bethesda MD, 1995, pp 63-118 Feller AC, Diebold J: Histopathology of nodal and extranodal non-Hodgkin’s lymphomas. Berlin, Springer, 2003, pp 53-66 Bennet MH: Sclerosis in non-Hodgkin’s lymphomata. Br J Cancer Suppl 31:44-52, 1975 Waldron JA, Newcomer LN, Katz ME: Sclerosing variants of follicular center cell lymphomas presenting in the retroperitoneum. Cancer 52: 712-720, 1983 Keller AR, Hochholzer L, Castleman B: Hyaline-vascular and plasma-cell types of giant lymph node hyperplasia of the mediastinum and other locations. Cancer 29:670-683, 1972 Nozawa Y, Hirao M, Kamimura K, Hara Y, Abe M: Unusual case of follicular lymphoma with hyaline vascular follicles. Pathol Intern 52:794-795, 2002 McCurley TL, Gay RE, Gay S, Glick AD, Haralson MA, Collins RD: The extracellular matrix in sclerosing follicular center cell lymphomas: an immunohistochemical and ultrastructural study. Hum Pathol 17: 930-938, 1986 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2007 VL 13 IS 1 BP 74 EP 77 PG 4 ER PT J AU Tschaharganeh, D Ehemann, V Nussbaum, T Schirmacher, P Breuhahn, K AF Tschaharganeh, Darjus Ehemann, Volker Nussbaum, Tanja Schirmacher, Peter Breuhahn, Kai TI Non-specific Effects of siRNAs on Tumor Cells with Implications on Therapeutic Applicability Using RNA Interference SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE nonsense siRNA; RNA interference; RNAi; non-specific effects; functional aspects; cancer ID nonsense siRNA; RNA interference; RNAi; non-specific effects; functional aspects; cancer AB Elimination of protein expression using RNA interference (RNAi) significantly improves the understanding of gene function and represents a promising technique for the treatment of diseases such as cancer and neurological disorders. Accumulating evidence suggests the so-called interferon-independent non-specific gene silencing of short interfering RNA (siRNA); however, its biological and functional cellular consequences are largely unidentified. We therefore analyzed the effects of different nonsense siRNAs on characteristic bio-parameters such as cell viability, proliferation, cell cycle distribution, apoptosis, and migration of tumor cells. All analyzed cellular aspects have been observed to be significantly affected by the presence of siRNA in an interferon-independent manner: viability, mitosis, and motility were significantly diminished and programmed cell death was significantly elevated. Moreover, all cell cycle stages (G0/G1-, G2/M-, and S-phase) were moderately shifted. Together, these results support the hypothesis that siRNA, due to sequence-specific cellular consequences, modulate bio-functionality independent of the target sequence. This phenomenon affects the design of siRNA experiments for future in vitro but also for in vivo tests as well as for potential therapeutic and preventive strategies. Moreover, monitoring interferon response after transfection of siRNAs is necessary but not sufficient to exclude potential off-target effects in non-diseased cells. C1 [Tschaharganeh, Darjus] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. [Ehemann, Volker] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. [Nussbaum, Tanja] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. [Schirmacher, Peter] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. [Breuhahn, Kai] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. RP Breuhahn, K (reprint author), University of Heidelberg, Department of Pathology, 69120 Heidelberg, Germany. EM kai.breuhahn@med.uni-heidelberg.de CR Sen GL, Blau HM: A brief history of RNAi: the silence of the genes. Faseb J 20:1293-1299, 2006 Watanabe T, Sudoh M, Miyagishi M, et al: Intracellular-diced dsRNA has enhanced efficacy for silencing HCV RNA and overcomes variation in the viral genotype. Gene Ther 13:883- 892, 2006 Xia X, Zhou H, Huang Y, et al: Allele-specific RNAi selectively silences mutant SOD1 and achieves significant therapeutic benefit in vivo. Neurobiol Dis 23:578-586, 2006 Ko K, Furukawa K, Takahashi T, et al: Fundamental study of small interfering RNAs for ganglioside GD3 synthase gene as a therapeutic target of lung cancers. Oncogene 25:6924-6935, 2006 Romano PR, McCallus DE, Pachuk CJ: RNA interferencemediated prevention and therapy for hepatocellular carcinoma. Oncogene 25:3857-3865, 2006 Elbashir SM, Martinez J, Patkaniowska A, et al: Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate RNA interference is mediated by 21- and 22-nucleotide RNAs. Embo J 20:6877-6888, 2001 Kumar A, Haque J, Lacoste J, et al: Double-stranded RNAdependent protein kinase activates transcription factor NFkappa B by phosphorylating I kappa B. Proc Natl Acad Sci U S A 91:6288-6292, 1994 Reynolds A, Anderson EM, Vermeulen A, et al: Induction of the interferon response by siRNA is cell type- and duplex lengthdependent. RNA 12:988-993, 2006 Persengiev SP, Zhu X, Green MR: Nonspecific, concentrationdependent stimulation and repression of mammalian gene expression by small interfering RNAs, siRNAs). RNA 10:12- 18, 2004 Semizarov D, Frost L, Sarthy A, et al: Specificity of short interfering RNA determined through gene expression signatures. Proc Natl Acad Sci U S A 100:6347-6352, 2003 Jackson AL, Bartz SR, Schelter J, et al: Expression profiling reveals off-target gene regulation by RNAi. Nat Biotechnol 21:635-637, 2003 Kern MA, Schubert D, Sahi D, et al: Proapoptotic and antiproliferative potential of selective cyclooxygenase-2 inhibitors in human liver tumor cells. Hepatology 36:885-894, 2002 Ehemann V, Hashemi B, Lange A, et al: Flow cytometric DNA analysis and chromosomal aberrations in malignant glioblastomas. Cancer Lett 138:101-106, 1999 Ehemann V, Sykora J, Vera-Delgado J, et al: Flow cytometric detection of spontaneous apoptosis in human breast cancer using the TUNEL-technique. Cancer Lett 194:125-131, 2003 Nicoletti I, Migliorati G, Pagliacci MC, et al: A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry. J Immunol Methods 139:271-279, 1991 Scacheri PC, Rozenblatt-Rosen O, Caplen NJ, et al: Short interfering RNAs can induce unexpected and divergent changes in the levels of untargeted proteins in mammalian cells. Proc Natl Acad Sci U S A 101:1892-1897, 2004 Doench JG, Petersen CP, Sharp PA: siRNAs can function as miRNAs. Genes Dev 17:438-442, 2003 Fedorov Y, Anderson EM, Birmingham A, et al: Off-target effects by siRNA can induce toxic phenotype. RNA 12:1188-1196, 2006 Ullu E, Tschudi C, Chakraborty T: RNA interference in protozoan parasites. Cell Microbiol 6:509-519, 2004 Jackson AL, Burchard J, Schelter J, et al: Widespread siRNA "off-target" transcript silencing mediated by seed region sequence complementarity. RNA 12:1179-1187, 2006 Jackson AL, Burchard J, Leake D, et al: Position-specific chemical modification of siRNAs reduces "off-target" transcript silencing. RNA 12:1197-1205, 2006 Schwarz DS, Hutvagner G, Haley B, et al: Evidence that siRNAs function as guides, not primers, in the Drosophila and human RNAi pathways. Mol Cell 10:537-548, 2002 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 84 EP 90 PG 7 ER PT J AU Kruger, S Ola, V Feller, CA Fischer, D Friedrich, M AF Kruger, Stefan Ola, Vladislava Feller, C Alfred Fischer, Dorothea Friedrich, Michael TI Expression of Cancer-Testis Antigen CT7 (MAGE-C1) in Breast Cancer: An Immunohistochemical Study with Emphasis on Prognostic Utility SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; cancer-testis antigen; CT7; MAGE-C1; prognosis; immunohistochemistry ID breast cancer; cancer-testis antigen; CT7; MAGE-C1; prognosis; immunohistochemistry AB High expression of the cancer-testis antigen CT7, also referred to as MAGE-C1, has been recently described in a variety of malignant tumors, including breast carcinoma. To our knowledge, no data concerning the prognostic utility of CT7 expression in breast cancer are available. In this retrospective study, we evaluated the relationship between CT7 immunoreactivity and clinicopathological parameters as well as relapse-free survival (RFS) and metastasis-free survival (MFS) of 124 women with invasive breast cancer. A positive CT7 status, defined as immunoreactivity in more than 50% of tumor cells, was found in 18% of cases and correlated significantly with high tumor grade (p=0.004), but with no other clinicopathological parameter. In a univariate analysis, CT7 status showed an association with RFS by trend (p=0.107; relative risk [RR]: 1.85) and a significant association with MFS (p=0.043; RR: 2.02). In a multivariate analysis, tumor grade, stage, nodal status, angioinvasion, HER2 expression as well as estrogen and progesterone receptor expression were identified as significant independent prognostic factors of RFS and/or MFS. In this respect, CT7 expression showed a weak, statistically not significant trend towards an independent prognostic relevance concerning prediction of MFS (p=0.147; RR: 1.95). Our data suggest that estimation of CT7 immunoreactivity is of limited prognostic usefulness in breast cancer. It may provide additional information concerning assessment of MFS in selected cases. C1 [Kruger, Stefan] University of Schleswig-Holstein, Institute of Pathology, Ratzeburger Allee 160, D-23538 Lubeck, Germany. [Ola, Vladislava] University of Schleswig-Holstein, Institute of Pathology, Ratzeburger Allee 160, D-23538 Lubeck, Germany. [Feller, C Alfred] University of Schleswig-Holstein, Institute of Pathology, Ratzeburger Allee 160, D-23538 Lubeck, Germany. [Fischer, Dorothea] University of Schleswig-Holstein, Department of Gynecology and ObstetricsLubeck, Germany. [Friedrich, Michael] University of Schleswig-Holstein, Department of Gynecology and ObstetricsLubeck, Germany. RP Kruger, S (reprint author), University of Schleswig-Holstein, Institute of Pathology, D-23538 Lubeck, Germany. EM krueger@patho.uni-luebeck.de CR Van den Eynde BJ, van der Bruggen P: T cell defined tumor antigens. Curr Opin Immunol 9: 684-693, 1997. Takahashi K, Shichijo S, Noguchi M, et al: Identification of MAGE-1 and MAGE-4 proteins in spermatogonia and primary spermatocytes of testis. Cancer Res 55: 3478-3482, 1995. Scanlan MJ, Gure AO, Jungbluth AA, et al: Cancer/testis antigens: an expanding family of targets for cancer immunotherapy. Immunol Rev 188: 22-32, 2002. Suri A: Cancer testis antigens – their importance in immunotherapy and in the early detection of cancer. Expert Opin Ther 6: 379-389, 2006. Marchand M, van Baren N, Weiynants P, et al: Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1. Int J Cancer 18: 219-230, 1999. Banchereau J, Palucka AK, Dhodapkar M, et al: Immune and clinical responses in patients with metastatic melanoma to CD34(+, progenitor-derived dentritic cell vaccine. Cancer Res 61: 6451-6458, 2001. Coulie PG, Karanikas V, Colau D, et al: A monoclonal cytolytic T-lymphocyte response observed in a melanoma patient vaccinated with a tumor-specific antigenic peptide encoded by gene MAGE-3. Proc Natl Acad Sci USA 98: 10290-10295, 2001. Bettinotti MP, Panelli MC, Ruppe E, et al: Clinical and immunological evaluation of patients with metastatic melanoma undergoing immunization with the HLA-Cw*0702-associated epitope MAGE-A12:170-178. Int J Cancer 105: 210-216, 2003. Marchand M, Punt CJ, Aamdal S, et al: Immunisation of metastatic cancer patients with MAGE-3 protein combined with adjuvant SBAS-2: a clinical report. Eur J Cancer 39: 70- 77, 2003. Kruit WH, van Ojik HH, Brichard VG, et al: Phase 1/2 study of subcutaneous and intradermal immunization with recombinant MAGE-3 protein in patients with detectable metastatic melanoma. Int J Cancer 117: 596-604, 2005. van Baren N, Bonnet MC, Dreno B, et al: Tumoral and immunologic response after vaccination of melanoma patients with an ALVAC virus encoding MAGE antigens recognized by T cells. J Clin Oncol 23: 9008-9021, 2005. Sadanaga N, Nagashima H, Mashino K, et al: Dendritic cell vaccination with MAGE peptide is a novel therapeutic approach for gastrointestinal carcinomas. Clin Cancer Res 7: 2277-2284, 2001. Russo V, Traversari C, Verrecchia A, et al: Expression of the MAGE gene family in primary and metastatic human breast cancer: implications for tumor antigen-specific immunotherapy. Int J Cancer 64: 216-221, 1995. Jungbluth AA, Chen YT, Busam KJ, et al: CT7, MAGE-C1, antigen expression in normal and neoplastic tissues. Int J Cancer 99: 839-845, 2002. Elston CW, Ellis IO: Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19: 403-410, 1991. Chitale DA, Jungbluth AA, Marshall DS, et al: Expression of cancer-testis antigens in endometrial carcinomas using a tissue microarray. Mod Pathol 18: 119-126, 2005. Early Breast Cancer Trialists’ Collaborative Group: Polychemotherapy for early breast cancer: an overview of the randomised trials. Lancet 352: 930-942, 1998. Early Breast Cancer Trialists’ Collaborative Group: Tamoxifen for early breast cancer: an overview of the randomised trials. Lancet 351: 1451-1467, 1998. Danish Breast Cancer Cooperative Group: Study of failure pattern among high-risk breast cancer patients with or without postmastectomy radiotherapy in addition to adjuvant systemic therapy: long-term results from the Danish Breast Cancer Cooperative Group BBCG 82b and c randomized studies. J Clin Oncol: 24: 2268-2275, 2006. Yamanaka K, Miyake H, Hara I, et al: Expression of MAGE genes in renal cell carcinoma. Int J Mol Med 2: 57-60, 1998. Patard JJ, Brasseur F, Gil-Diez S, et al: Expression of MAGE genes in transitional cell carcinomas of the urinary bladder. Int J Cancer 64: 60-64, 1995. Toh Y, Yamana H, Shichijo S, et al: Expression of MAGE-1 gene by esophageal carcinomas. Jpn J Cancer Res 86: 714-717, 1995. Nagao T, Higashitsuji H, Nonoguchi K, et al: MAGE-A4 interacts with the liver oncoprotein gankyrin and suppresses its tumorigenic activity. J Biol Chem 278: 10668-10674, 2003. Peikert T, Specks U, Farver C, et al: Melanoma antigen A4 is expressed in non-small cell lung cancers and promotes apoptosis. Cancer Res 66: 4693-4700, 2006. Monte M, Simonatto M, Peche LY, et al: MAGE-A tumor antigens target p53 transactivation function through histone deacetylase recruitment and confer resistance to chemotherapeutic agents. Proc Natl Acad Sci USA 103: 11160-11165, 2006. Riker A, Cormier J, Panelli M, et al: Immune selection after antigen-specific immunotherapy of melanoma. Surgery 126: 112-120, 1999. El-Shami K, Tirosh B, Bar-Haim E, et al: MHC class I-restricted epitope spreading in the context of tumor rejection following vaccination with a single immunodominant CTL epitope. Eur J Immunol 29: 3295-3301, 1999. Markiewicz MA, Fallarino F, Ashikari A, et al: Epitope spreading upon P815 tumor rejection triggered by vaccination with the single class I MHC-restricted peptide P1A. Int Immunol 13: 625-632, 2001. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 91 EP 96 PG 6 ER PT J AU Badalian, G Barbai, T Raso, E Derecskei, K Szendroi, M Timar, J AF Badalian, Gayane Barbai, Tamas Raso, Erzsebet Derecskei, Katalin Szendroi, Miklos Timar, Jozsef TI Phenotype of Bone Metastases of Non-Small Cell Lung Cancer: Epidermal Growth Factor Receptor Expression and K-RAS Mutational Status SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR immunohistochemistry; K-RAS mutation; NSCLC; bone metastasis ID EGFR immunohistochemistry; K-RAS mutation; NSCLC; bone metastasis AB Bone metastasis is a frequent complication of lung cancer progression, however, studies on bone metastatic tissues are scanty. Here we have collected a small cohort of 11 non-small cell lung cancer cases where primary tumors and corresponding bone metastases were available for pathological analysis. We have tested two molecular markers: EGFR protein expression and K-RAS mutation at codon 12 using immunohistochemistry and RFLPPCR, respectively. We have shown that using improved protocols, EGFR protein (both the extracellular as well as the cytoplasmic domain) is readily detectable in decalcified bone tissue. We found that the EGFR expression status is highly similar in bone metastases compared to the primary tumors, although the expression levels may change. Individual comparison of corresponding primary and metastatic NSCLC tissues indicated that downregulation of EGFR was a rare event (2/11) compared to upregulation (4/11) in bone metastases. On the other hand, our data indicate that the K-RAS mutational status of the primary tumor does not predict the status of the bone metastatic tissue of NSCLC, since we have observed both emergence of mutant clones in metastases from wild-type (wt) primary tumors and loss of mutant clones in metastases from mutant primaries in addition to the maintained mutant status. Our data support that at least two progression models occur in NSCLC, the samegene as well as the clonal selection one. It is noteworthy that in NSCLC cases with wt- or mutant KRAS, downregulation of EGFR expression was a rare event although upregulation in bone metastases was observed more frequently in wt K-RAS cases. C1 [Badalian, Gayane] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Barbai, Tamas] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Raso, Erzsebet] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Derecskei, Katalin] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. [Szendroi, Miklos] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Timar, Jozsef] National Institute of Oncology, Department of Tumor Progression, Rath Gyorgy u. 7-9., H-1122 Budapest, Hungary. RP Timar, J (reprint author), National Institute of Oncology, Department of Tumor Progression, H-1122 Budapest, Hungary. CR Mundy GR: Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer 2:584-593, 2002 Roodman GD: Mechanisms of bone metastasis. N Engl J Med 350:1655-1664, 2004 Garber ME, Troyanskaya OG, Schluens K, Petersen S, Thaesler Z, Pacyna-Gengelbach M, van de Rijn M, Rosen GD, Perou CM, Whyte RI, Altman RB, Brown PO, Botstein D, Petersen I: Diversity of the gene expression in adenocarcinoma of the lung. Proc Natl Acad Sci USA 98:13784-13789, 2001 Beer DG, Kardia SL, Huang CC, Giordano TJ, Levin AM, Misek DE, Lin L, Chen G, Gharib TG, Thomas DG, Lizyness ML, Kuick R, Hayasaka S, Taylor JM, Iannettoni MD, Orringer MB, Hanash S: Gene-expression profiles predict survival of patients with lung adenocarcinoma. Nat Med 8:816-824, 2002 Kopper L, Timar J: Genomics of lung cancer may change diagnosis, prognosis and therapy. Pathol Oncol Res 11:5-10, 2005 Marchetti A, Martella C, Felicioni L, Barassi F, Salvatore S, Chella A, Camplese PP, Iarussi T, Mucilli F, Mezzetti A, Cuccurullo F, Sacco R, Buttitta F: EGFR mutations in non-smallcell lung cancer: analysis of a large series of cases and development of a rapid and sensitive method for diagnostic screening with potential implications on pharmacologic treatment. J Clin Oncol 23:857-865, 2005 Aviel-Ronen S, Blackhall FH, Shepherd FA, Shepherd FA, Tsao MS: K-ras mutations in non-small cell lung carcinoma: a review. Clin Lung Cancer 8:30-38, 2006 Soung YH, Lee JW, Kim SY, Seo SH, Park WS, Nam SW, Song SY, Han JH, Park CK, Lee JY, Yoo NJ, Lee SH: Mutational analysis of EGFR and K-RAS genes in lung adenocarcinomas. Virchows Arch 446:483-488, 2005 Riely GJ, Politi KA, Miller VA, Pao W: Update on epidermal growth factor receptor mutations in non-small cell lung cancer. Clin Cancer Res 12:7232-7241, 2006 Derecskei K, Moldvay J, Bogos K, Timar J: Protocol modifications influence the result of EGF receptor immunodetection by EGFR pharmDxTM in paraffin-embedded cancer tissues. Pathol Oncol Res 12: 243-246, 2006 Molina JR, Adjei AA, Jett JR: Advances in chemotherapy of non-small cell lung cancer. Chest 130:1211-1219, 2006 Bepler G, Li X, Sharma A et al: Clinical value of tumoral RRM1 and ERCC1 expressions for treatment response and therapeutic decisions in NSCLC. In: ASCO 2006 Education Book, Ed.: M.C. Perry, ASCO, Alexandria, 2006, pp 431-435 Patel JD: Epidermal growth factor receptor pathway targeted therapy in patients with aerodigestive malignancies. Curr Opin Oncol 18:609-614, 2006 Fearon ER, Vogelstein B: A genetic model for colorectal tumorigenesis. Cell 61:759-767, 1990 Gray JW: Evidence emerges for early metastasis and parallel evolution of primary and metastatic tumors. Cancer Cell 4:4-6, 2003 Bernards R, Weinberg RA: A progression puzzle. Nature 418:823, 2002 Shiseki M, Kohno T., Nishikawa R, Sameshima Y, Mizoguchi H, Yokota J: Frequent allelic losses on chromosomes 2q, 18q, and 22q in advanced non small cell lung carcinoma. Cancer Res 54:5643-5648, 1994 Petersen S, Aninat-Mayer M, Schluns K, Gellert K, Dietel M, Petersen I: Chromosomal alteration in the clonal evolution of the metastatic stage of squamous cell carcinoma of the lung. Br J Cancer 82:65-73, 2000 Takahashi K, Kohno T, Matsumoto S, Nakanishi Y, Arai Y, Yamamoto S, Fujiwara T, Tanaka N, Yokota J: Clonal and parallel evolution of primary lung cancers and their metastases revealed by molecular dissection of cancer cells. Clin Cancer Res 13:111-120, 2007 Matsumoto S, Takahasi K, Iwakawa R, Matsuno Y, Nakanishi Y, Kohno T, Shimizu E, Yokota J: Frequent EGFR mutation in brain metastases of lung adenocarcinoma. Int J Cancer 15:1491-1494, 2006 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 99 EP 104 PG 6 ER PT J AU Sanada, Y Yoshida, K Ohara, M Tsutani, Y AF Sanada, Yuichi Yoshida, Kazuhiro Ohara, Masahiro Tsutani, Yasuhiro TI Expression of Orotate Phosphoribosyltransferase (OPRT) in Hepatobiliary and Pancreatic Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE OPRT; pancreatic carcinoma; gallbladder carcinoma; hepatocellular carcinoma; pancreatic intraepithelial neoplasia (PanIN) ID OPRT; pancreatic carcinoma; gallbladder carcinoma; hepatocellular carcinoma; pancreatic intraepithelial neoplasia (PanIN) AB The purpose of this study was to clarify the role of orotate phosphoribosyltransferase (OPRT) in the progression of hepatobiliary and pancreatic carcinomas. Representative sections from 8 surgically resected pancreatic carcinomas, 5 gallbladder carcinomas and 19 hepatocellular carcinomas (HCCs) were examined microscopically. Sites of pancreatic intraepithelial neoplasia (PanIN) were counted, and histologic subtypes of invasive ductal carcinoma of the pancreas (IDC) were determined. Gallbladder carcinomas and HCCs were examined histologically, and the subtypes and spread patterns were assessed. Expression of OPRT was examined immunohistochemically. A total of 75 PanINs were identified. Expression of OPRT increased as lesions progressed from early to high-grade PanINs (PanIN-1A and -1B versus PanIN-2 and -3, p=0.0004). Three (37.5%) of the 8 pancreatic IDCs were positive for OPRT. In the remaining 5 cases, OPRT was expressed only in the neoplastic ducts adjacent to PanIN-3s. In gallbladder carcinomas, mucosal neoplastic epithelium showed dense cytoplasmic expression in 4 of the 5 cases, but expression was absent in the deeply invasive lesions. Among HCCs, 15 of the 19 cases were negative for OPRT in the central area of the tumor, but 8 of the 19 cases expressed OPRT in vascularly invasive lesions. Our data suggest that OPRT is involved in early events of pancreatic and gallbladder carcinogenesis and invasion of HCC. C1 [Sanada, Yuichi] Hiroshima University, Department of Surgical Oncology, Research Institution for Radiation Biology and Medicine, 1-2-3 Kasumi, 734-8551 Hiroshima, Minami-ku, Japan. [Yoshida, Kazuhiro] Hiroshima University, Department of Surgical Oncology, Research Institution for Radiation Biology and Medicine, 1-2-3 Kasumi, 734-8551 Hiroshima, Minami-ku, Japan. [Ohara, Masahiro] Hiroshima University, Department of Surgical Oncology, Research Institution for Radiation Biology and Medicine, 1-2-3 Kasumi, 734-8551 Hiroshima, Minami-ku, Japan. [Tsutani, Yasuhiro] Hiroshima University, Department of Surgical Oncology, Research Institution for Radiation Biology and Medicine, 1-2-3 Kasumi, 734-8551 Hiroshima, Minami-ku, Japan. RP Sanada, Y (reprint author), Hiroshima University, Department of Surgical Oncology, Research Institution for Radiation Biology and Medicine, 734-8551 Hiroshima, Japan. EM ysanadasurg@hotmail.com CR Sakamoto K, Sugimoto Y, Miyadera K, et al: Preparation of anti-orotate phosphoribosyltransferase antibody and its application to immunohistochemical detection in human tumor cells. Int J Mol Med 16: 245-249, 2005 Oguri T, Achiwa H, Bessho Y, et al: The role of thymidylate synthase and dihydropyrimidine dehydrogenase in resistance to 5-fluorouracil in human lung cancer cells. Lung Cancer 49: 345-351, 2005 Ishida H, Shirakawa K, Ohsawa T, et al Expression of mRNA levels of thymidylate synthase, dihydropyrimidine dehydrogenase, and orotate phosphoribosyltransferase of colorectal cancer–relationships among mRNA expression with response to 5-FU based treatment. Gan To Kagaku Ryoho 32: 1929-1934, 2005, in Japanese) Kamoshida S, Shiogama K, Shimomura R, et al: Immunohistochemical demonstration of fluoropyrimidine-metabolizing enzymes in various types of cancer. Oncol Rep 14: 1223-1230, 2005 Ochiai T, Sugitani M, Nishimura K, et al: Impact of orotate phosphoribosyl transferase activity as a predictor of lymph node metastasis in gastric cancer. Oncol Rep 14: 987-992, 2005 Nakano J, Huang C, Liu D, et al: Evaluations of biomarkers associated with 5-FU sensitivity for non-small-cell lung cancer patients postoperatively treated with UFT. Br J Cancer 95: 607- 615, 2006 Ochiai T, Nishimura K, Noguchi H, et al: Prognostic impact of orotate phosphoribosyl transferase activity in resectable colorectal cancers treated by 5-fluorouracil-based adjuvant chemotherapy. J Surg Oncol 94: 45-50, 2006 Takaori K, Kobayashi Y, Matsusue S, et al: Pancreatic intraepithelial neoplasia. Pancreas 28: 257-261, 2004 Lemoine NR, Jain S, Hughes CM, et al: K-ras oncogene activation in preinvasive pancreatic cancer. Gastroenterology 102: 230-236, 1992 Wilentz RE, Lacobuzio-Donahue CA, Argani P, et al: Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia: evidence that Dpc4 inactivation occurs late in neoplastic progression. Cancer Res 60: 2002-2006, 2000 Kim GE, Bae HI, Park HU, et al: Aberrant expression of MUC5AC and MUC6 gastric mucin and Sialyl Tn antigen in intraepithelial neoplasms of the pancreas. Gastroenterology 123: 1052-1060, 2002 Sanada Y, Yoshida K, Ohara M, et al: Histopathologic evaluation of stepwise progression of pancreatic carcinoma with immunohistochemical analysis of gastric epithelial transcription factor SOX2: comparison of expression patterns between invasive components and cancerous or nonneoplastic intraductal components. Pancreas 32: 164-170, 2006 Albores-Saavedra J, Shukla D, Carrick K, Henson DE: In situ and invasive adenocarcinomas of gallbladder extending into or arising from Rokitansky-Aschoff sinuses. A clinicopathologic study of 49 cases. Am J Surg Pathol 28: 621-628, 2004 Kojiro M: Histopathology of liver cancers. Best Pract Res Clin Gastroenterol 19: 39-62, 2005 Mizutani Y, Wada H, Fukushima M, et al: Prognostic significance of orotate phosphoribosyl transferase activity in bladder carcinoma. Cancer 100: 723-731, 2004 Takamori H, Kanemitsu K, Tsuji T, et al: 5-fluorouracil intraarterial infusion combined with systemic gemcitabine for unresectable pancreatic cancer. Pancreas 30: 223-226, 2005 Alberts SR, al-Khatib H, Mahoney MR, et al: Gemcitabine, 5- fluorouracil, and leucovorin in advanced biliary tract and gallbladder carcinoma in North Central Cancer Treatment Group phase II trial. Cancer 103: 111-118, 2005 Obi S, Yoshida H, Toure R, et al: Combination therapy of intraarterial 5-fluorouracil and systemic interferon-alpha for advanced hepatocellular carcinoma with portal venous invasion. Cancer 106: 1990-1997, 2006 Sakurai Y, Sakamoto K, Sugimoto Y, et al: Orotate phosphoribosyltransferase levels measured by a newly established enzymelinked immunosorbent assay in gastric carcinoma. Cancer Sci 97: 492-498, 2006 Dhawan P, Singh AB, Deane NG, et al: Claudin-1 regulates cellular transformation and metastatic behavior in colon cancer. J Clin Invest 115: 1765-1776, 2005 Doucas H, Garcea G, Neal CP, et al: Chemoprevention of pancreatic cancer: a review of the molecular pathways involved, and evidence for the potential for chemoprevention. Pancreatology 6: 429-439, 2006 Hidaka S, Yasutake T, Fukushima M, et al: Chromosomal imbalances associated with acquired resistance to fluoropyrimidines in human colorectal cancer cells. Eur J Cancer 39: 975-980, 2003 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 105 EP 113 PG 9 ER PT J AU Jakso, P Kereskai, L Molnar, L Pajor, L AF Jakso, Pal Kereskai, Laszlo Molnar, Lenke Pajor, Laszlo TI Lineage-Specific Clonality Analysis of Chronic Myeloproliferative Disorders and Myelodysplastic Syndrome by Human Androgen Receptor Assay SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chronic myeloproliferative disease; myelodysplasia; HUMARA ID chronic myeloproliferative disease; myelodysplasia; HUMARA AB In myelodysplastic syndrome (MDS) as well as chronic myeloproliferative disorders (CMPD) others than chronic myeloid leukemia the frequency of pathognomonic genetic aberrations is very low and, therefore, X chromosome inactivation (XI) assays may help in assessing the clonality. To establish specific clonality criteria on XI, human androgen receptor assay (HUMARA) was performed on sorted myeloid and lymphoid peripheral blood cells of 21 healthy females. Clonality criteria 1 and 2 conferring at least 90% specificity were set based on the ranges and differences of XI number (XIN) describing the ratio of representation of the two alleles in as well as in between reactive myeloid and lymphoid compartments. Spiking experiments indicated that the test identifies clonality reliably when no more than 40-50% reactive cells are admixed. In the CMPD and MDS cases peripheral myeloid cells were monoclonal by one of the two criteria in 71-100%, whereas lymphoid cells in 28-75%. The results of HUMARA, available in 73% of the female patients, supported the clinicopathological data in 84% as well as proved pluripotent stem cell origin in 31-75% and 21% of CMPDs and MDS, respectively. C1 [Jakso, Pal] University of Pecs, Department of Pathology, 12. Szigeti u., H-7643 Pecs, Hungary. [Kereskai, Laszlo] University of Pecs, Department of Pathology, 12. Szigeti u., H-7643 Pecs, Hungary. [Molnar, Lenke] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Pajor, Laszlo] University of Pecs, Department of Pathology, 12. Szigeti u., H-7643 Pecs, Hungary. RP Pajor, L (reprint author), University of Pecs, Department of Pathology, H-7643 Pecs, Hungary. EM titkar@pathology.pote.hu CR Allen RC, Zoghbi HY, Moseley AB: Methylation of HpaII and HhaI sites near the polymorphic CAG repeat in the human androgen-receptor gene correlates with X chromosome inactivation. Am J Hum Genet 51:1229-1239, 1992 Anderson JE, Gilliland DG, List AF: Myelodysplastic syndrome. Educational Materials by the American Society of Hematology, 1998 Baxter EJ, Scott LM, Campbell PJ: Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 365:1054-1061, 2005 Busque L, Gilliland DG: X-inactivation analysis in the 1990s: promise and potential problems. Leukemia 12:128-135, 1998 Culligan DJ, Cachia P, Wittaker J: Clonal lymphocytes are detectable in only some cases of MDS. Br J Haematol 81:346- 352, 1992 Heaney ML, Golde DW: Myelodysplasia. N Engl J Med 340:1649-1660, 1999 Hellstrom-Lindberg E, Willmann C, Barrett AJ: Achievements in understanding and treatment of myelodysplastic syndromes. Educational Materials by the American Society of Hematology, 2000. Jaffe ES, Harris NL, Stein H, Vardinian JW eds. World Health Organization Classification of Tumours, Pathology & Genetics, Tumours of Haematopoietic and Lymphoid Tissues. IARC Press, Lyon, 2001 James C, Ugo V, Le Couedic JP: A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 434:1144-1148, 2005 Knuutila S, Teerenhovi L, Larramendy ML: Cell lineage involvement of recurrent chromosomal abnormalities in hematological neoplasms. Genes, Chromosomes Cancer 10:95-102, 1994 Kralovics R, Passamonti F, Buser AS: A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 352:1779-1790, 2005 Levine RL, Wadleigh M, Cools J: Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocytemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 7:387-397, 2005 Nilsson L, Astrand-Grundstrom I: Isolation and characterization of hematopoietic progenitor/stem cells in 5q-deleted myelodysplastic syndromes: evidence for involvement at the hematopoietic stem cell level. Blood 156:2012-2021, 2000 Pajor L, Matolcsy A, Vass JA, Mehes G, Marton E, Szabo F: Phenotypic and genotypic analyses of blastic cell population suggest that pure B-lymphoblastic leukemia may arise from myelodysplastic syndrome. Leuk Res 22:13-17, 1998 Pearson TC, Messinezy M, Westwood N: A Polycythemia Vera Update: diagnosis, pathobiology, and treatment. Educational Materials by the American Society of Hematology, 2000. Raskind WH, Steinmann L, Najfeld V: Clonal development of myeloproliferative disorders: clues to hematopoietic differentiation and multistep pathogenesis of cancer. Leukemia 12: 108- 116, 1998 Tonon L, Bergamaschi G, Dellavecchia C, Rosti V, Lucotti C, Malabarba: Unbalanced X-chromosome inactivation in haematopoietic cells from normal women. Br J Haematol 102:996-1003, 1998 Zhao R, Xing S, Li Z: Identification of an acquired JAK2 mutation in Polycythemia vera. J Biol Chem 280:22788-22792, 2005 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 114 EP 122 PG 9 ER PT J AU Sharma, S Sharma, ChM Johnson, HM Lou, M Thakar, A Sarkar, Ch AF Sharma, Suash Sharma, Chand Mehar Johnson, H Maribeth Lou, Mimi Thakar, Alok Sarkar, Chitra TI Esthesioneuroblastoma - a Clinicopathologic Study and Role of DNA Topoisomerase Alpha SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE esthesioneuroblastoma; topoisomerase alpha ID esthesioneuroblastoma; topoisomerase alpha AB Esthesioneuroblastoma (ENB) differs from adrenal neuroblastomas in its histopathologic and biologic characteristics. Hyams grading and Kadish staging have shown correlation with survival. Scant data are available on proliferation indices and prognosis. We retrospectively reviewed the clinicopathologic characteristics of ENB. Both Kadish and UCLA staging systems were used. Hyams grading was simplified into low and high grade. DNA topoisomerase II alpha labeling index (T2alpha LI) was obtained in 8 cases using immunohistochemistry. Of the 19 cases studied, 14 were males and 5 females. Age range was 2 to 62 years (average 27 years). The mass primarily involved the nose in 12 (63%) and paranasal sinuses in 7 cases (37%). Patients presented with nose block in 19 (100%), epistaxis in 10 (53%), proptosis in 9 (47%) and loss of vision in 6 cases (32%). Bony involvement was seen in 7 cases (37%), and intracranial spread in one case (5%). Thirteen (68%) were low-grade tumors and 6 were (32%) high-grade. There was no statistically significant difference between the low- and high-grade ENB in age (years) (p=0.2882), duration of symptoms (months) (p=0.5636), and either in the Kadish (p=0.5456) or the UCLA staging system (p=0.7771). The difference in DNA topoisomerase alpha labeling index between the low- and highgrade ENB (medians: 10.4 and 22.3, respectively) was not statistically significant (p=0.0714), but it was suggestive of a positive association. The results of this study should be interpreted with caution, because of the limited sample size. Three cases recurred locally, one each stage A, B and C, but all low-grade. This preliminary study suggests the need to combine a simplified histologic grading with accurate staging in a reasonable attempt to assess local progression in esthesioneuroblastoma. Larger studies may clarify the role of T2alpha LI in improving histologic grading. C1 [Sharma, Suash] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Sharma, Chand Mehar] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Johnson, H Maribeth] Medical College of Georgia, Biostatistics Consulting CenterAugusta, GA, USA. [Lou, Mimi] Medical College of Georgia, Biostatistics Consulting CenterAugusta, GA, USA. [Thakar, Alok] All India Institute of Medical Sciences, Department of OtolaryngologyNew Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. RP Sarkar, Ch (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM drchitasarkar@yahoo.com CR Argani P, Perez-Ordonez B, Xiao H, Cariana SM, Huvos AG, Ladanyi M: Olfactory neuroblastoma is not related to the Ewing’s family of tumors: absence of EWS/FLI1 gene fusion and MIC2 expression. Am J Surg Pathol 22: 391-398, 1998 Bockmuhl U, You X, Pacyna-Gengelbach M, Arps H, Draf W, Petersen I: CGH pattern of esthesioneuroblastoma and their metastases. Brain Pathol 14: 158-163, 2004 Broich G, Pagliari A, Ottaviani F: Esthesioneuroblastoma: a general review of the cases published since the discovery of the tumor in 1924. Anticancer Res 17: 2683-2706, 1996 Constantinidis J, Steinhart H, Koch M, Buchfelder M, Schaenzer A, Weidenbacher M, Iro H: Olfactory neuroblastoma: The University of Erlangen-Nuremberg experience 1975-2000. Otolaryngol Head Neck Surg 130:567-574, 2004 Dulguerov P, Allal AS, Calacaterra TC: Esthesioneuroblastoma: a meta-analysis and review. Lancet Oncol 2: 683-690, 2001 Dulguerov P, Calcaterra T: Esthesioneuroblastoma: The UCLA experience 1970-1990. Laryngoscope 102: 843-849, 1992 Dias FL, Sa GM, Lima RA, Kligerman J, Leoncio MP, Freitas EQ, Soares JR, Arcuri RA: Patterns of failure and outcome in esthesioneuroblastoma. Arch Otolaryngol Head Neck Surg 129: 1186-1192, 2003 Eden BV, Debo RF, Larner JM, Kelly MD, Levine PA, Stewart FM, Cantrell RW, Constable WC: Esthesioneuroblastoma. Long-term outcome and patterns of failure – the University of Virginia experience. Cancer 73: 2556-2562, 1994 Eriksen JG, Bastholt L, Krogdahl AS, Hansen O, Joergensen KE: Esthesioneuroblastoma – what is the optimal treatment? Acta Oncol 39: 231-235, 2000 Haas I, Ganzer U: Does sophisticated diagnostic workup on neuroectodermal tumors have an impact on the treatment of esthesioneuroblastoma? Onkologie 26: 261-267, 2003 Ho DM, Hsu CY, Ting LT, Chiang H: MIB-1 and DNA topoisomerase II alpha could be helpful for predicting long-term survival of patients with glioblastoma. Am J Clin Pathol 119: 715- 722, 2003 Hwang SK, Paek SH, Kim DG, Jeon YK, Chi JG, Jung HW: Olfactory neuroblastomas: survival rate and prognostic factors. J Neurooncol 59: 217-226, 2002 Hyams VJ: Tumors of upper respiratory tract and ear. In: Atlas of Tumor Pathology, Eds: Hyams VJ, Batsakis JG, Michaels L, eds.). 2nd series, Fascicle 25. Washington, DC, Armed Forces Institute of Pathology, 1988, pp 240-248 Ingeholm P, Theilgaard SA, Buchwald C, Hansen HS, Francis D: Esthesioneuroblastoma: a Danish clinicopathological study of 40 consecutive cases. APMIS 110: 639-645, 2002 Jethanamest D, Morris LG, Sikora AG, Kutler DI: Esthesioneuroblastoma: a population-based analysis of survival and prognostic factors. Arch Otolaryngol Head Neck Surg 133: 276-280, 2007 Kadish S, Goodman M, Wang CC: Olfactory neuroblastoma: a clinical analysis of 17 cases. Cancer 37: 1571-1576, 1976 Koka VN, Julieron M, Bourhis J, Janot F, Le Ridant AM, Marandas P, Luboinski B, Schwaab G: Aesthesioneuroblastoma. J Laryngol Otol 112: 628-633, 1998 Levine PA, Gallagher R, Cantrell RW: Esthesioneuroblastoma: reflections of a 21-year experience. Laryngoscope 109: 1539- 1543, 1999 Liu WS, Tang PZ, Xu GZ: Clinical analysis of 34 cases of esthesioneuroblastoma. Zhonghua Er Bi Yan Hou Ke Za Zhi 39: 328-332, 2004 Loy AH, Reibel JF, Read PW, Thomas CY, Newman SA, Jane JA, Levine PA: Esthesioneuroblastoma: continued follow-up of a single institution’s experience. Arch Otolaryngol Head Neck Surg 132: 134-138, 2006 McElroy EA, Buckner JC, Lewis JE: Chemotherapy for advanced esthesioneuroblastoma: The Mayo Clinic experience. Neurosurgery 42: 1023-1028, 1998 Meneses MS, Thurel C, Mikol J, Ramina R, Maniglia JJ, Arruda WO, Cophignon J: Esthesioneuroblastoma with intracranial extension. Neurosurgery 27: 813-819, 1990 Mezzelani A, Tornielli S, Minoletti F, Pierotti MA, Sozzi G, Pilotti S: Esthesioneuroblastoma is not a member of the primitive peripheral neuroectodermal tumor – Ewing’s group. Br J Cancer 81: 586-591, 1999 Mishima Y, Nagasaki E, Terui Y, Irie T, Takahashi S, Ito Y, Oguchi M, Kawabata K, Kamata S, Hatake K: Combination chemotherapy, cyclophosphamide, doxorubicin, and vincristine with continuous- infusion cisplatin and etoposide, and radiotherapy with stem cell support can be beneficial for adolescents and adults with esthesioneuroblastoma. Cancer 101: 1437-1444, 2004 Miyagami M, Katayama Y, Kinukawa N, Sawada T: An ultrastructural and immunohistochemical study of olfactory neuroepithelioma with rhabdomyoblasts. Med Electron Microsc 35: 160-166, 2002 Miyamoto RC, Gleich LL, Biddinger PW, Gluckana JL: Esthesioneuroblastoma and sinonasal undifferentiated carcinoma: impact of histologic grading and clinical staging on survival and prognosis. Laryngoscope 110: 1262-1265, 2000 Morita A, Ebersold MJ, Olsen KD, Foote RL, Lewis JE, Quast LM: Esthesioneuroblastoma: prognosis and management. Neurosurgery 32: 706-715, 1993 Olsen KE, Knudsen H, Rasmussen BB, Balslev E, Knoop A, Ejlertsen B, Nielsen KV, Schonau A, Overgaard J: Amplification of HER2 and TOP2A and deletion of TOP2A genes in breast cancer investigated by new FISH probes. Acta Oncol 43: 35-42, 2004 Park SH, Suh YL: Expression of cyclin A and topoisomerase II alpha of oligodendrogliomas is correlated with tumor grade, MIB-1 labelling index and survival. Histopathology 42: 395- 402, 2003 Rosenthal DI, Barker JL Jr, El-Naggar AK, Glisson BS, Kies MS, Diaz EM Jr, Clayman GL, Demonte F, Selek U, Morrison WH, Ang KK, Chao KS, Garden AS: Sinonasal malignancies with neuroendocrine differentiation: patterns of failure according to histologic phenotype. Cancer 101: 2567-2573, 2004 Sakata K, Aoki Y, Karasawa K, Nakagawa K, Hasezawa K, Muta N, Terahara A, Onogi Y, Sasaki Y, Akanuma A, et al: Esthesioneuroblastoma: a report of seven cases. Acta Oncol 32: 399-402, 1993 Tatagiba M, Samii M, Dankoweit-Timpe E, Aguiar PH, Osterwald L, Babu R, Ostertag H: Esthesioneuroblastomas with intracranial extension. Proliferative potential and management. Arq Neuropsiquiatr 53: 577-586, 1995 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 123 EP 129 PG 7 ER PT J AU Croce, VM Isla-Larrain, M Rabassa, EM Demichelis, S Colussi, GA Crespo, M Lacunza, E Segal-Eiras, A AF Croce, V Maria Isla-Larrain, Marina Rabassa, E Martin Demichelis, Sandra Colussi, G Andrea Crespo, Marina Lacunza, Ezequiel Segal-Eiras, Amada TI Lewis x is Highly Expressed in Normal Tissues: a Comparative Immunohistochemical Study and Literature Revision SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE carbohydrate epitopes; mucins; normal epithelia; immunohistochemistry ID carbohydrate epitopes; mucins; normal epithelia; immunohistochemistry AB An immunohistochemical analysis was employed to determine the expression of carbohydrate antigens associated to mucins in normal epithelia. Tissue samples were obtained as biopsies from normal breast (18), colon (35) and oral cavity mucosa (8). The following carbohydrate epitopes were studied: sialyl-Lewis x, Lewis x, Lewis y, Tn hapten, sialyl-Tn and Thomsen-Friedenreich antigen. Mucins were also studied employing antibodies against MUC1, MUC2, MUC4, MUC5AC, MUC6 and also normal colonic glycolipid. Statistical analysis was performed and Kendall correlations were obtained. Lewis x showed an apical pattern mainly at plasma membrane, although cytoplasmic staining was also found in most samples. TF, Tn and sTn haptens were detected in few specimens, while sLewis x was found in oral mucosa and breast tissue. Also, normal breast expressed MUC1 at a high percentage, whereas MUC4 was observed in a small number of samples. Colon specimens mainly expressed MUC2 and MUC1, while most oral mucosa samples expressed MUC4 and MUC1. A positive correlation between MUC1VNTR and TF epitope (r=0.396) was found in breast samples, while in colon specimens MUC2 and colonic glycolipid versus Lewis x were statistically significantly correlated (r=0.28 and r=0.29, respectively). As a conclusion, a defined carbohydrate epitope expression is not exclusive of normal tissue or a determined localization, and it is possible to assume that different glycoproteins and glycolipids may be carriers of carbohydrate antigens depending on the tissue localization considered. C1 [Croce, V Maria] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Isla-Larrain, Marina] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Rabassa, E Martin] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Demichelis, Sandra] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Colussi, G Andrea] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Crespo, Marina] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Lacunza, Ezequiel] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. [Segal-Eiras, Amada] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), Calle 60 y 120La Plata, Argentina. RP Segal-Eiras, A (reprint author), Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), La Plata, Argentina. EM as-eiras@netverk.com.ar CR Ajioka Y, Xing P, Hinoda Y, Jass J: Correlative histochemical study providing evidence for the dual nature of human colorectal cancer mucin. Histochem J 29:143-152, 1997 Baldus S, Engelmann K, Hanisch F-G: MUC1 and MUCs: a family of human mucins with impact in cancer biology. Cr Rev Clin Lab Sci 41:189-231, 2004 Baruch A, Hartmann M, Yoeli M, et al: The breast cancer associated MUC1 gene generates both a receptor and its cognate binding protein. Cancer Res 59:1552-1561, 1999 Beum PV, Cheng P-W: Biosynthesis and function of β 1,6 branched mucin-type glycans. In: The Molecular Immunology of Complex Carbohydrates – 2., Ed: Wu AM), Kluwer Academic/ Plenum Publishers, 2001, pp. 279-312 Braga VMM, Pemberton LF, Duhig T, et al: Spatial and temporal expression of an epithelial mucin, Muc-1, during mouse development. Development 115:427-437, 1992 Bresalier RS, Byrd JC, Brodt P, et al: Liver metastasis and adhesion to the sinusoidal endothelium by human colon cancer cells is related to mucin carbohydrate chain length. Int J Cancer 76:556-562, 1998 Brockhausen I, Romero PA, Herscovics A: Glycosyltransferase changes upon differentiation of CaCo-2 human colonic adenocarcinoma cells. Cancer Res 51:3136-3142, 1991 Brown A, Feizi T, Gooi HC, et al: A monoclonal antibody against human colonic adenoma recognizes difucosylated Type-2-blood-group chains. Biosci Rep 3:163-170, 1983 Bryne M, Reibel J, Mandel U, Dabelsteen E: Expression of mucin type carbohydrates may supplement histologic diagnosis in oral premalignant lesions. J Oral Pathol Med 20:120-125, 1991 Burchell J, Gendler S, Taylor-Papadimitrious J, et al: Development and characterization of breast cancer reactive monoclonal antibodies directed against to the core protein of the human milk mucin. Cancer Res 47:5476-5482, 1987 Cao Y, Karsten U, Liebrich W: Expression of Thomsen-Friedenreich- related antigens in primary and metastatic colorectal carcinomas. A reevaluation. Cancer 76:1700-1708, 1995 Cao Y, Stosiek P, Springer G, et al: Thomsen-Friedenreichrelated carbohydrate antigens in normal adult human tissues: a systematic and comparative study. Histochem Cell Biol 106:197-207, 1996 Cao Y, Blohm D, Ghadimi BM: Mucins, MUC1 and MUC3, of gastrointestinal and breast epithelia reveal different and heterogeneous tumor-associated aberrations in glycosylation. J Histochem Cytochem 45:1547-1557, 1997 Carraway K, Perez A, Idris N, et al: Muc4/sialomucin complex, the intramembrane ErbB2 ligand, in cancer and epithelia: to protect and to survive. Prog Nucleic Acid Res Mol Biol 171:149-185, 2000 Chang SK, Dohrman AF, Basbaum CB: Localization of mucin, MUC2 and MUC3, messenger RNA and peptide expression in human normal intestine and colon cancer. Gastroenterol 107:28-36, 1994 Croce MV, Price MR, Segal-Eiras A: Antigen heterogeneous expression in colorectal cancer. J Exp Clin Cancer Res 15:353- 356, 1996 Croce MV, Colussi AG, Price MR, et al: Expression of tumour associated antigens in normal, benign and malignant human mammary epithelial tissue: a comparative immunohistochemical study. Anticancer Res 17:4287-4292, 1997 Croce MV, Colussi AG, Price MR, et al: Identification and characterization of different subpopulations in a human lung adenocarcinoma cell line, A549). Pathol Oncol Res 5:197-204, 1999 Croce MV, Rabassa ME, Price MR, Segal-Eiras A: MUC1 mucin and carbohydrate associated antigens as tumor markers in head and neck squamous cell carcinoma. Pathol Oncol Res 7:284-291, 2001 Croce MV, Isla-Larrain MT, Remes-Lenicov F, et al: MUC1 cytoplasmic tail detection using CT33 polyclonal and CT2 monoclonal antibodies in breast and colorectal tissue. Histol Histopathol 21:849-855, 2006 Dabelsteen E, Roed-Petersen B, Pindborg JJ: Loss of epithelial blood group antigens A and B in oral premalignant lesions. Acta Pathol Microbiol Immunol Scand 83:292-300, 1975 Dabelsteen E, Clausen H, Holmstrup P, et al: Premalignant and malignant oral lesions are associated with changes in the glycosylation pattern of carbohydrates related to ABH blood group antigens. APMIS 96:813-819, 1988 Dabelsteen E: Cell surface carbohydrates as prognostic markers in human carcinomas. J Pathol 179:358-369, 1996 Dabelsteen E, Gron B, Mandel U, et al: Altered expression of epithelial cell surface glyconjugates and intermediate filaments at the margins of mucosal wounds. J Invest Dermatol 111:592- 597, 1998 Feickert HJ, Anger BR, Cordon-Cardo C: Cell-surface antigens of human lung tumors detected by mouse monoclonal antibodies: definition of blood-group- and non-blood-grouprelated antigenic systems. Int J Cancer 46:1007-1013, 1990 Feizi T, Gooi H, Childs R, et al: Tumour-associated and differentiation antigens on the carbohydrate moieties of mucin-type glycoproteins. Biochem Soc Trans 12: 591-596, 1984 Fernandez B, Lund J, Meyers F: Epithelial membrane antigen expression in benign and malignant squamous epithelium of the head and neck. Otolaryngol Head Neck Surg 97:288-293, 1987 Fukuda MN, Dell A, Oates JE, et al: Structures of glycosphingolipids isolated from human granulocytes. The presence of a series of linear poly-N-acetyllactosaminylceramide and its significance in glycolipids of whole blood cells. J Biol Chem 260:1067-1082, 1985 Fukushima K, Hirota M, Terasaki P, et al: Characterization of sialosylated Lewis x as a new tumor-associated antigen. Cancer Res 44:5279-5285, 1984 Gao S, Dabelsteen E, Reibel J, et al: Genotypic characterization of histo-blood group ABO in oral squamous cell carcinomas, abstract). J Dent Res, 2003 Gendler S, Burchell JM, Duhig T, et al: Cloning of partial cDNA encoding differentiation and tumor-associated mucin glycoproteins expressed by human mammary epithelium. Proc Natl Acad Sci USA 84:6060-6064, 1987 Gendler SJ, Lancaster CA, Taylor-Papadimitriou J, et al: Molecular cloning and expression of human tumor-associated polymorphic epithelial mucin. J Biol Chem 265:15286- 15293, 1990 Gendler SJ: MUC1, the renaissance molecule. J Mammary Gland Biol Neoplasia 6:339-353, 2001 Hakomori S: Carbohydrate to carbohydrate interaction, through glycosynapse, as a basis of cell recognition and membrane organization. Glycoconj J 21:125-137, 2004 Hanai N, Shitara K, Yoshida H: Generation of monoclonal antibodies against human lung squamous cell carcinoma and adenocarcinoma using mice rendered tolerant to normal human lung. Cancer Res 46:4438-4443, 1996 Hanisch FG, Uhlenbruck G, Peter-Katalinic J, et al: Structures of neutral O-linked polylactosaminoglycans on human skim milk mucins. A novel type of linearly extended poly-N-acetyllactosamine backbones with Galβ(1–4)GlcNAcβ(1–6, repeating units. J Biol Chem 264:872-877, 1989 Hanisch FG, Muller S: MUC1: The polymorphic appearance of a human mucin. Glycobiol 10:439-449, 2000 Hanski C, Hanski M-L, Zimmer T, et al: Characterization of the major sialyl-Lex-positive mucins present in colon, colon carcinoma and sera of patients with colorectal cancer. Cancer Res 55:928-933, 1995 Ho SB, Kim YS: Carbohydrate antigens on cancer-associated mucin-like molecules. Semin Cancer Biol 2:389-400, 1991 Ho SB, Niehans GA, Lyftogt C, et al: Heterogeneity of mucin gene expression in normal and neoplastic tissues. Cancer Res 53:641-651, 1993 Irimura T, Denda K, Lida S, et al: Diverse glycosylation of MUC1 and MUC2: potential significance in tumor immunity. J Biochem 126:975-985, 1999 Itoh T, Yonezawa S, Nomoto M, et al: Expression of mucin antigens and Lewis X-related antigens in carcinomas and dysplasia of the pharynx and larynx. Pathol Int 46:646-655, 1996 Jeannon JP, Aston V, Stafford FW, et al: Expression of MUC1 and MUC2 glycoproteins in laryngeal cancer. Clin Otolaryngol Allied Sci 26:109-112, 2001 Kufe D, Inghirami G, Abe M, et al: Differential reactivity of a novel monoclonal antibody, DF3, with human malignant versus benign breast tumors. Hybridoma 3:223-232, 1984 Levitin F, Stern O, Weiss M, et al: The MUC1 SEA module is a self-cleaving domain. J Biol Chem 280:33374-33386, 2005 Ligtenberg MJ, Buijs F, Vos HL, et al: Suppression of cellular aggregation by high levels of episialin. Cancer Res 52:2318- 2324, 1992 Liu B, Lague JR, Nunes DP, et al: Expression of membraneassociated mucins MUC1 and MUC4 in major human salivary glands. 50:811-820, 2002 Lloyd O, Burchell J, Kudryashov V, et al: Comparison of O-linked carbohydrate chains in MUC-1 mucin from normal breast epithelial cell lines and breast carcinoma cell lines. J Biol Chem 271:33325-33334, 1996 Lopez-Ferrer A, de Bolos C, Barranco C, et al: Role of fucosyltransferases in the association between apomucin and Lewis antigen expression in normal and malignant gastric epithelium. Gut 47:349-356, 2000 Luna-More S, Rius F, Weil B, et al: EMA: a differentiation antigen related to node metastatic capacity of breast carcinomas. Pathol Res Pract 197:419-425, 2001 Mandel U, Petersen OW, Sorensen H, et al: Simple mucin-type carbohydrates in oral stratified squamous and salivary gland epithelia. J Invest Dermatol 97:713-721, 1991 Meerzaman D, Shapiro PS, Kim KC: Involvement of the MAP kinase ERK2in MUC1 mucin signaling. Am J Physiol Lung Cell Mol Physiol 281:L86-L91, 2001 Moniaux N, Escande F, Porchet N, et al: Structural organization and classification of the human mucin genes. Front Biosci 6:D1192-206, 2001 Nakagoe T, Fukushima K, Hirota M, et al: An immunohistochemical employer monoclonal antibodies against Le(a), sialyl Le(a), Le(x), and sialyl Le(x, antigens in primary colorectal, carcinomas and lymph node and hepatic lesions. J Gastroenterol 29:129-138, 1994 Nitta T, Sugihara K, Tsuyama S, et al: Immunohistochemical study of MUC1 mucin in premalignant oral lesions and oral squamous cell carcinoma: association with disease progression, mode of invasion, and lymph node metastasis. Cancer 88:245-254, 2000 Orntoff TF, Harving N, Langkilde NC: O-linked mucin-type glycoproteins in normal and malignant colon mucosa: lack of T-antigen expression and accumulation of Tn and sialosyl-Tn antigens in carcinomas. Int J Cancer 45:666-672, 1990 Parry S, Silverman H, McDermott K, et al: Identification of MUC1 proteolytic cleavage sites in vivo. Biochem Biophys Res Commun 283:715-720, 2001 Perey L, Hayes DF, Kufe DF: Effects of differentiating agents on cell surface expression of the breast carcinoma associated DF3-P epitope. Cancer Res 52:6365-6370, 1992 Philipsen EK, Jorgensen M, Dabelsteen E: Expression of blood group-related carbohydrate antigens in normal human pancreatic tissue. APMIS 99:931-940, 1991 Podolsky DK: Oligosaccharide structures of isolated human colonic mucin species. J Biol Chem 260:15510-15515, 1985 Price MR, Pugh JA, Hudecz F, et al: C595 – a monoclonal antibody against the protein core of human urinary epithelial mucin commonly expressed in breast carcinomas. Br J Cancer 61:681- 686, 1990 Price MR: Epitopes of carcinoembryonic antigen, CEA, defined monoclonal antibodies. Br J Cancer 57:165-169, 1988 Price MR, Sekowski M, Yang GY, et al: Reactivity of an anti-, human gastric carcinoma, monoclonal antibody with co-related peptides of gastrointestinal mucin. Cancer Immunol Immunother 33:80-84, 1991 Ramsauer VP, Caraway CA, Salas PJ: MUC4/sialomucin complex, the intramembrane ErbB2 ligand, translocates ErbB2 to the apical surface in polarized epithelial cells. J Biol Chem 278:30142-30147, 2003 Reibel J: Prognosis of oral pre-malignant lesions: significance of clinical, histopathological and molecular biological characteristics. Crit Rev Oral Biol Med 14:47-62, 2003 Reis CA, David L, Nielsen P: Immunohistochemical study of MUC5AC expression in human gastric carcinomas using a novel monoclonal antibody. Int J Cancer 74:112-121, 1997 Reis CA, Sorensen T, Mandel U, et al: Development and characterization of an antibody directed to an alpha-N-acetyl-Dgalactosamine glycosylated MUC2 peptide. Glycoconj J 15:51- 62, 1998 Reis CA, David L, Carvalho F: Immunohistochemical study of the expression of MUC6 mucin and coexpression of other secreted mucins, MUC5AC and MUC2, in human gastric carcinomas. J Histochem Cytochem 48:377-388, 2000 Renkonen J, Paavonen T, Renkonen R: Endothelial and epithelial expression of sialyl Lewis(x, and sialyl Lewis(a, in lesions of breast carcinoma. Int J Cancer 74:296-300, 1997 Schroeder JA, Thompson MC, Gardner MM, Gendler SJ: Transgenic MUC1 interacts with epidermal growth factor receptor and correlates with mitogen-activated protein kinase activation in the mouse mammary gland. J Biol Chem 276:13057-13064, 2001 Sengupta A, Valdramidou D, Huntley S, et al: Distribution of MUC1 in the normal human oral cavity is localized to the ducts of minor salivary glands. Arch Oral Biol 46:529-538, 2001 Shimizu M, Yamauchi K: Isolation and characterization of mucin-like glycoprotein in human milk fat globule membrane. J Biochem 91:515-524, 1982 Spielman J, Hull SR, Sheng ZQ, et al: Biosynthesis of a tumor cell surface sialomucin. Maturation and effects of monensin. J Biol Chem 263:9621-9629, 1988 Springer GF: T and Tn, general carcinoma autoantigens. Science 224:1198-1206, 1984 Takada A, Ohmori K, Yoneda T, et al: Contribution of carbohydrate antigens sialyl Lewis A and sialyl Lewis X to adhesion of human cancer cells to vascular endothelium. Cancer Res 53:354-361, 1993 Tatemoto Y, Saka M, Tanimura T, et al: Immunohistochemical observations on binding of monoclonal antibody to epithelial membrane antigen in epithelial tumors of the oral cavity and skin. Oral Surg Oral Med Oral Pathol 64:721-726, 1987 Taylor-Papadimitriou J, Burchell J, Miles DW, et al: MUC1 and cancer. Biochem Biophys Acta 1455:301-313, 1999 Therkildsen MH, Mandel U, Thorn J et al: Simple mucin-type carbohydrate antigens in major salivary glands. 42:1251-1259, 1994 Urdal DL, Brentrall TA, Bernstein ID, Hakomori SI: A granulocyte reactive monoclonal antibody, 1G10, identifies the Galβ-4, Fuc a 1-3, GlcNAc, X determinant, expressed in HL- 60 cells on both glycolipid and glycoprotein molecules. Blood 62:1022-1026, 1983 Yamashita K, Tachibana Y, Hitoi A, Kobata A: Sialic acid-containing sugar chains of hen ovalbumin and ovomucoid. Carbohydr Res 130:271-288, 1984 Yogeeswaran G, Salk PL: Metastatic potential is positively correlated with cell surface sialylation of cultured mouse tumor cell lines. Science 212:1514-1516, 1981 Weed DT, Gomez-Fernandez C, Bonfante E, et al: MUC4, sialomucin complex, expression in salivary gland tumors and squamous cell carcinoma of the upper aerodigestive tract. 124:127- 141, 2001 Zotter S, Hageman PC, Lossnitzer A et al: Tissue and tumor distribution of human polymorphic epithelial mucin. Cancer Reviews 11/12:55-101, 1988 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 130 EP 138 PG 9 ER PT J AU Baintner, K Bodnar, Zs Kiss, P Kiss, LA Lukats, AF Baintner, Karoly Bodnar, Zsofia Kiss, Peter Kiss, L Anna Lukats, Akos TI Effect of Intraperitoneally Administered Plant Lectins on Leukocyte Diapedesis and Visceral Organ Weight in Rats and Mice SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lectin; ConA; WGA; PHA; neutrophil; diapedesis; mesothelial; CINC ID lectin; ConA; WGA; PHA; neutrophil; diapedesis; mesothelial; CINC AB The effects of intraperitoneally administered plant lectins were examined in rats and mice. Intraperitoneally injected ConA transiently decreased the leukocyte count in the peritoneal cavity, due to the agglutination and attachment of cells to the peritoneal lining. Subsequently the total cell count was increased for hours, exceeding initial values. Peritoneal fluid aspartate transaminase (AST) concentration showed little change during the accumulation of ascitic fluid. The most marked histological alterations were found when wheat germ lectin was injected ip. (WGA, 10 mg/kg, 6 h). Neutrophil granulocytes migrated across the wall of both arterioles and venules, but the response was highly variable among adjacent vessels. The wall of the arterioles may have impeded the migration of neutrophil granulocytes, resulting in their accumulation in the muscular layer. Granulocyte accumulation was also observed in patches under the mesothelium and in other sites of the interstitium. Marked dilatation and thrombosis of a few venules were also observed. Kidney bean lectin (PHA) induced similar but less pronounced changes. The neutrophil diapedesis suggests the release of mediator(s) from mesothelial cells and/or peritoneal white cells. The cytokine-induced neutrophil chemoattractant CINC-1, injected as control, resulted in the diapedesis of predominantly mononuclear cells in the omentum within 40 minutes. In rats ip. injected ConA increased the wet weight of spleen and liver within 6 and 10 h, respectively, but kidney weight did not change. Intravascular clumping of red blood cells, thrombosis and organ weight changes also suggest the absorption of ConA into the circulation. The experiments show that plant lectins, used as models of bacterial lectins, can reproduce some aspects of peritonitis. C1 [Baintner, Karoly] Faculty of Animal Science, University of Kaposvar, Department of Physiology, H-7401 Kaposvar, Hungary. [Bodnar, Zsofia] Faculty of Animal Science, University of Kaposvar, Department of Physiology, H-7401 Kaposvar, Hungary. [Kiss, Peter] University of Agriculture, Department of Agricultural ChemistryGodollo, Hungary. [Kiss, L Anna] Faculty of Medicine, Semmelweis University, Institute of Human Morphology and Developmental BiologyBudapest, Hungary. [Lukats, Akos] Faculty of Medicine, Semmelweis University, Institute of Human Morphology and Developmental BiologyBudapest, Hungary. RP Baintner, K (reprint author), Faculty of Animal Science, University of Kaposvar, Department of Physiology, H-7401 Kaposvar, Hungary. EM baintner@mail.atk.u-kaposvar.hu CR Takeda K, Kaisho T, Akira S: Toll-like receptors. Annu Rev Immunol 21:335-376, 2003 Baintner K, Kiss P, Bardocz S, Pusztai A: Effect of orally administered plant lectins on intestinal liquor accumulation and amylase activity in rats. Acta Physiol Hung 91:73-81, 2004 Baintner K, Kiss P, Pikli A, Peumans W, Bardocz S, Pusztai A: Origin and mediation of secretion induced by oral phytohaemagglutinin, PHA, in rats. Acta Physiol Hung 91: 221-233, 2004 Roth J: Ultrahistochemical demonstration of saccharide components of complex carbohydrates at the alveolar cell surface and at the mesothelial cell surface of the pleura visceralis of mice by means of concanavalin A. Exp Pathol, Jena, 8: 157- 167, 1973 Kortsik CS, Freudenberg N, Riede U: Lectin binding sites and immunocytochemical characterization of normal pleural mesothelium. Gen Diagn Pathol 141: 141-146, 1995 Mohr W, Beneke G, Murr L: Transformation of peritoneal and pleural cells by phytohaemagglutinin. Beitr Pathol 142: 90- 113, 1970 Nagaoka I, Ishibashi Y, Yamashita T: Studies on phytohaemagglutinin- induced peritonitis of mice: Dynamics of inflammatory responses and influence of anticomplementary agents and anti-inflammatory drugs. Int J Tissue React 9: 179-188, 1987 Nagaoka I, Kaneko H, Yamashita T: Inhibition of the accumulation of macrophages and the generation of macrophage chemotactic activity by dexamethasone in concanavalin Ainduced peritonitis of mice. Agents Actions 25: 156-163, 1988 Alencar NMN, Assreuy AMS, Alencar VBM, Melo SC, Ramos MV, Cavada BS, Cunha FQ, Ribeiro RA: The galactose-binding lectin from Vatairea macrocarpa seeds induces in vivo neutrophil migration by indirect mechanism. Int J Biochem Cell Biol 35: 1674-1681, 2003 Vretblad P: Purification of lectins by biospecific affinity chromatography. Biochim Biophys Acta 434: 169-176, 1976 Pusztai A, Watt WB, Stewart JC: A comprehensive scheme for the isolation of trypsin inhibitors and the agglutinin from soybean seeds. J Agr Food Chem 39: 862-866, 1991 Bardocz S, Grant G, Pusztai A, Franklin MF, de Carvalho A: The effect of phytohaemagglutinin at different dietary concentrations on the growth, body composition and plasma insulin of the rat. Br J Nutr 76: 613-626, 1996 Smirnova IV, Khaspekova SC, Ignatov VV, Mazurov AV: Interaction of wheat germ agglutinin and concanavalin A with platelets. Stimulation of platelet functional reactions and binding with membrane glycoproteins. Biokhimija 63: 710-718, 1998 Nasreen N, Mohammed KA, Hardwick J, van Horn RD, Sanders KL, Doerschuk CM, Hott JW, Antony VB: Polar production of interleukin-8 by mesothelial cells promotes the transmesothelial migration of neutrophils: role of intercellular adhesion molecule-1. J Infect Dis 183: 1638-1645, 2001 Meager A: Cytokine regulation of cellular adhesion molecule expression in inflammation. Cytokine Growth Factor Rev 10: 27-39, 1999 Ebnet K, Kaldjian EP, Anderson AO, Shaw S: Orchestrated information transfer underlying leukocyte endothelial interactions. Annu Rev Immunol 14: 155-177, 1996 McEver RP: Selectins: lectins that initiate cell adhesion under flow. Curr Opin Cell Biol 14: 581-586, 2002 Knolle PA, Gerken G, Loser E, Dienes HP, Gantner F, Tiegs G, Meyer zum Buschenfelde KH, Lohse AW: Role of sinusoidal endothelial cells of the liver in concanavalin A-induced hepatic injury in mice. Hepatology 24: 824-829, 1996 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 139 EP 143 PG 5 ER PT J AU Kavantzas, N Paraskevakou, H Tseleni-Balafouta, S Aroni, K Athanassiades, P Agrogiannis, G Patsouris, E AF Kavantzas, Nikolaos Paraskevakou, Helen Tseleni-Balafouta, Sofia Aroni, Kyriaki Athanassiades, Pauline Agrogiannis, George Patsouris, Efstratios TI Association between Microvessel Density and Histologic Grade in Renal Cell Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE renal carcinoma; microvascular density ID renal carcinoma; microvascular density AB Angiogenesis seems to contribute to tumor growth and the development of metastases. There may be an association between the vascular density of individual tumors and their prognosis. In the present survey we studied 53 cases of renal cell carcinoma investigating possible relationship between histologic grade and microvessel density (MVD) measured by an image analysis system. According to our results MVD was significantly associated with the histologic grade, higher grades being accompanied with a higher MVD. Further studies are needed to investigate a possible connection of MVD with the prognostic role of grade in RCCs. C1 [Kavantzas, Nikolaos] University of Athens, Medical School, Department of Pathology, 60 Agias Lavras str., GR-157 73 Athens, Greece. [Paraskevakou, Helen] University of Athens, Medical School, Department of Pathology, 60 Agias Lavras str., GR-157 73 Athens, Greece. [Tseleni-Balafouta, Sofia] University of Athens, Medical School, Department of Pathology, 60 Agias Lavras str., GR-157 73 Athens, Greece. [Aroni, Kyriaki] University of Athens, Medical School, Department of Pathology, 60 Agias Lavras str., GR-157 73 Athens, Greece. [Athanassiades, Pauline] University of Athens, Medical School, Department of Pathology, 60 Agias Lavras str., GR-157 73 Athens, Greece. [Agrogiannis, George] University of Athens, Medical School, Department of Pathology, 60 Agias Lavras str., GR-157 73 Athens, Greece. [Patsouris, Efstratios] University of Athens, Medical School, Department of Pathology, 60 Agias Lavras str., GR-157 73 Athens, Greece. RP Agrogiannis, G (reprint author), University of Athens, Medical School, Department of Pathology, GR-157 73 Athens, Greece. EM agrogeorge@panafonet.gr CR Denekamp J, Hobson B: Endothelial-cell proliferation in experimental tumours. Br J Cancer 46: 711-720, 1982 Diaz-Cano SJ, de Miguel M, Blanes A, Galera H, Wolfe HJ: Contribution of the microvessel network to the clonal and kinetic profiles of adrenal cortical proliferative lesions. Hum Pathol 32: 1232-1239, 2001 Gelb A, Sudilovsky D, Daniel Wu C, Weiss L.M, Medeiros JL: Appraisal of intratumoral microvessel density, MIB-1 score, DNA content and p53 protein expression as prognostic indicators in patients with locally confined renal cell carcinoma. Cancer 80: 1768-1775, 1997 Ginnara JR, Ward LM, Porter FD, Wagner JR, Devor DE, Grinberg A, Emmert-Buck MR, Westphal H, Klausner RD, Linehan WM: Defective placental vasculogenesis causes embryonic lethality in VHL-deficient mice. Proc Natl Acad Sci USA 94: 9102-9107, 1997 Haigh JJ, Gerber HP, Ferrara N, Wagner EF: Conditional inactivation of VEGF-A in areas of collagen2α1 expression results in embryonic lethality in the heterozygous state. Development 127: 1445-1453, 2000 Heicappell R: Cadherins in renal cell carcinoma. Anticancer Res 19(2C): 1501-1504, 1999 Herbst C, Kosmehl H, Stiller KJ, Berndt A, Eiselt M, Schubert J, Katenkamp B: Evaluation of microvessel density by computerised image analysis in human renal cell carcinoma. Correlation to pT category, nuclear grade, proliferative activity and occurrence of metastasis. J Cancer Res Clin Oncol 124:141- 147, 1998 Imao T, Egawa M, Takashima H, Koshida K, Namiki M: Inverse correlation of microvessel density with metastasis and prognosis in renal cell carcinoma. Int J Urol. 1:948-953, 2004 Joo HJ, Oh DK, Kim YS, Lee KB, Kim SJ: Increased expression of caveolin-1 and microvessel density correlates with metastasis and poor prognosis in clear cell renal cell carcinoma. BJU Int 93:291-296, 2004 McCue PA, Gorstein F: Genetic markers in renal cell carcinomas. Hum Pathol 32: 1027-1028, 2001 MacLennan GT, Bostwick DC: Microvessel density in renal cell carcinoma: lack of prognostic significance. Urology 46:27-30, 1995 Nativ O, Sabo E, Reiss A, Wald M, Madjar S, Moskovitz B: Clinical significance of tumor angiogenesis in patients with localized renal cell carcinoma. Urology 51: 693-696, 1998 Pantuck A, Zeng G, Belldegrun A, Figlin R: Pathobiology, prognosis and targeted therapy for renal cell carcinomas. Clin Cancer Res 9: 4641-4652, 2003 Paraskevakou E, Kavantzas N, Pavlopoulos PM, Delibasis A, Yova D, Davaris P: Computerized nuclear morphometry of renal cell carcinomas. Gen Diagn Pathol 142: 101-104, 1996 Rioux-Leclercq N, Epstein JI, Bansard JY, Turlin B, Botard JJ, Manunta A, Chan T, Ramee MP, Lobel B, Moulinoux JP: Clinical significance of cell proliferation, microvessel density and CD44 adhesion molecule expression in renal cell carcinoma. Hum Pathol 32: 1209-1215, 2001 Tuna B, Yorukoglu K, Unlu M, Mungan MU, Kirkali Z: Association of mast cells with microvessel density in renal cell carcinomas. Eur Urol 50:530-534, 2006 Van Brussel JP, Mickisch GH: Prognostic factors in renal cell and bladder cancer. BJU Int 83: 902-908, 1999 Yagasaki H, Kawata N, Takimoto Y, Nemoto N: Histopathological analysis of angiogenic factors in renal cell carcinoma. Int J Urol 10:220-227, 2003 Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, Topalian SL, Steinberg SM, Chen HX, Rosenberg SA: A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med 349:427-434, 2003 Yoshino S, Kato M, Okada K: Clinical significance of angiogenesis, proliferation and apoptosis in renal cell carcinoma. Anticancer Res 20: 591-594, 2000 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 145 EP 148 PG 4 ER PT J AU Bodor, Cs Schmidt, O Csernus, B Rajnai, H Szende, B AF Bodor, Csaba Schmidt, Otto Csernus, Balazs Rajnai, Hajnalka Szende, Bela TI DNA and RNA Isolated from Tissues Processed by Microwave-Accelerated Apparatus MFX-800-3 are Suitable for Subsequent PCR and Q-RT-PCR Amplification SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE paraffin embedding; MFX-800-3 histoprocessor; real-time PCR; molecular pathology ID paraffin embedding; MFX-800-3 histoprocessor; real-time PCR; molecular pathology AB Over the past decade, methods of molecular biology have appeared in diagnostic pathology and are routinely applied on formalin-fixed, paraffin-embedded histological samples, processed via conventional embedding methods. Due to its reagent- and cost-effectiveness, embedding techniques that utilize microwave acceleration in one or more steps of histoprocessing are increasingly used by numerous laboratories. The demand arises that tissues processed this way should also be suitable for the requirements of molecular pathology. In this study, both conventionally embedded and MFX-800-3 machine-processed tissue samples from the same source were used for isolation of DNA and RNA and for performing PCR and real-time PCR. PCR amplification of the beta-globin gene, as well as the real-time PCR amplification of the ABL mRNA was successful in all cases. Our conclusion is that samples processed by the vacuum assisted automatic microwave histoprocessor MFX-800-3 are perfectly applicable for DNA and RNA isolation and provide appropriate templates for further PCR and realtime PCR studies. C1 [Bodor, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Schmidt, Otto] MeditestBudapest, Hungary. [Csernus, Balazs] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Rajnai, Hajnalka] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Szende, Bela] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. RP Szende, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM bodor@korb1.sote.hu CR Beillard E, Pallisgaard N, van der Velden VH, et al: Evaluation of candidate control genes for diagnosis and residual disease detection in leukemic patients using ‘real-time’ quantitative reverse-transcriptase polymerase chain reaction, RQPCR, - a Europe against cancer program. Leukemia 17:2474- 2486, 2003 Benchekroun M, DeGraw J, Gao J, et al: Impact of fixative on recovery of mRNA from paraffin-embedded tissue. Diagn Mol Pathol 13:116-125, 2004 Boon ME, Wals-Paap CH, Visinoni FA, et al: The two-step vacuum- microwave method for histoprocessing. Eur J Morphol 33:349-358, 1995 Coindre JM, Hostein I, Terrier P, et al: Diagnosis of clear cell sarcoma by real-time reverse transcriptase-polymerase chain reaction analysis of paraffin embedded tissues: clinicopathologic and molecular analysis of 44 patients from the French sarcoma group. Cancer 107:1055-1064, 2006 Crocker J: Demystified... Molecular pathology in oncology. Mol Pathol 55:337-347, 2002. Ekuni D, Firth JD, Putnins EE, et al: RNA integrity and in situ RT-PCR in dento-alveolar tissues after microwave accelerated demineralisation. Arch Oral Biol 51:164-169, 2006 Gjerdrum LM, Sorensen BS, Kjeldsen E, et al: Real-time quantitative PCR of microdissected paraffin-embedded breast carcinoma: an alternative method for HER-2/neu analysis. J Mol Diagn 6:42-51, 2004 Hsu HC, Peng SY, Shun CT, et al: High quality of DNA retrieved for Southern blot hybridization from microwavefixed, paraffin-embedded liver tissues. J Virol Methods 31:251-261, 1991 Kok LP, Visser PE, Boon ME, et al: Histoprocessing with the microwave oven: an update. Histochem J 20:323-328, 1988 Kovacs L, Szende B, Elek G, et al: Working experience with a new vacuum-accelerated microwave histoprocessor. J Pathol 180:106-110, 1996 Lou YK, Qin H, Molodysky E, et al: Simple microwave and thermal cycler boiling methods for preparation of cervicovaginal lavage cell samples prior to PCR for human papillomavirus detection. J Virol Methods 44:77-81, 1993 Man YG, Burgar A: An antigen unmasking protocol that satisfies both immunohistochemistry and subsequent PCR amplification. Pathol Res Pract 199:815-825, 2003 Mies C: Molecular biological analysis of paraffin-embedded tissues. Hum Pathol 25:555-560, 1994 Miller SA, Dykes DD, Polesky HF, et al: A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16:1215, 1988 Ryan JL, Fan H, Glaser SL, et al: Epstein-Barr virus quantitation by real-time PCR targeting multiple gene segments: a novel approach to screen for the virus in paraffin-embedded tissue and plasma. J Mol Diagn 6:378-385, 2004 Vincek V, Nassiri M, Nadji M, et al: A tissue fixative that protects macromolecules, DNA, RNA, and protein, and histomorphology in clinical samples. Lab Invest 83:1427-1435, 2003 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 149 EP 152 PG 4 ER PT J AU Sitic, S Korac, P Peharec, P Zovko, G Perisa, MM Gasparov, S AF Sitic, Sanda Korac, Petra Peharec, Petra Zovko, Gojko Perisa, Milkovic Marija Gasparov, Slavko TI Bcl-2 and MALT1 Genes are not Involved in the Oncogenesis of Uterine Tumors Resembling Ovarian Sex Cord Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE uterus; ovarian sex cord tumors; Bcl-2; MALT1; translocation ID uterus; ovarian sex cord tumors; Bcl-2; MALT1; translocation AB Uterine tumors resembling ovarian sex cord tumors (UTROSCT) are rare entities. They were described by Clement and Scully in 1976 who classified them into groups I and II. Group I comprises typical endometrial stromal neoplasms with focal areas resembling ovarian sex cord elements and group II are predominantly or completely composed of ovarian sex cord-like elements. We report a case of UTROSCT type II with cytogenetic analysis. The tumor occurred in a 76-year-old woman who presented with vaginal bleeding. The tumor was lobulated, firm, yellow and histologically composed of sex cord-like elements. Tumor cells expressed vimentin, CD10, CD99 and alpha-actin. Cytogenetic analysis in a previously reported case detected translocation t(4;18)(q21.1;q21.3) in the majority of cells. Bcl-2 and MALT1 genes are located at or near the translocation breakpoints, and the aim of this study was to determine whether these genes were involved in chromosomal translocation or tumorigenesis. We did not find IgH translocation or the most common MALT translocations. Bcl-2 was also not involved in this oncogenesis. C1 [Sitic, Sanda] “Merkur” University Hospital, Department of Clinical Pathology and Cytology, Zajceva 19Zagreb, Croatia. [Korac, Petra] “Merkur” University Hospital, Department of Clinical Pathology and Cytology, Zajceva 19Zagreb, Croatia. [Peharec, Petra] University Hospital Center Zagreb, Department of PathologyZagreb, Croatia. [Zovko, Gojko] Clinical Hospital Merkur, University Clinic for Gynecology and Obstetrics, Department of Gynecological OncologyZagreb, Croatia. [Perisa, Milkovic Marija] “Merkur” University Hospital, Department of Clinical Pathology and Cytology, Zajceva 19Zagreb, Croatia. [Gasparov, Slavko] “Merkur” University Hospital, Department of Clinical Pathology and Cytology, Zajceva 19Zagreb, Croatia. RP Sitic, S (reprint author), “Merkur” University Hospital, Department of Clinical Pathology and Cytology, Zagreb, Croatia. EM sitic.st@inet.hr CR Baker RJ, Hildebrandt RH, Rouse RV, Hendrickson MR, Longacre TA: Inhibin and CD99, MIC2, expression in uterine stromal neoplasms with sex-cord-like elements. Hum Pathol 30: 671-679, 1999 Busam KJ, Iversen K, Coplan KA, Old LJ, Stockert E, Chen YT, McGregor D, Jungbluth A: Immunoreactivity for A103, an antibody to melanocytic antigen melan-A, Mart-1), in adrenocortical and other steroid tumors. Am J Surg Pathol 22: 57-63, 1998 Clement PB, Scully RE: Uterine tumors resembling ovarian sex-cord tumors. Am J Clin Pathol 66: 512-525, 1976 Fukunaga M, Miayazawa Y, Ushigome S: Endometrial lowgrade stromal sarcoma with ovarian sex cord-like differentiation: report of two cases with an immunohistochemical and flow cytometric study. Pathol Int 47: 412-415, 1977 Harris NL, Jaffe ES, Diebold J, Flandrin G, Muller-Hermelink HK, Vardiman J: The World Health Organisation classification of neoplastic diseases of the haematopoetic and lymphoid tissue: report of the Clinical Advisory Committee Meeting, Airlie House, Virginia, November 1997. Histopathology 36: 69-97, 2000 Hauptmann S, Nadjari B, Kraus J, Turnwald W, Dietel M: Uterine tumor resembling ovarian sex-cord tumor—a case report and review of the literature. Virchows Arch 439: 97-101, 2001 Hedley DW, Friedlander ML, Taylor IW, Rugg CA, Musgrove EA: Method for analysis of cellular DNA content of paraffinembedded pathological material using flow cytometry. J Histochem Cytochem 31: 1333-1335, 1983 Hiddemann W, Schumann J, Andreeff M, Barlogie B, Herman CJ, Leif RC, Mayall BH, Murphy RF, Sandberg AA: Convention on nomenclature for DNA cytometry. Cytometry 5: 445- 446, 1984 Hillard JB, Malpica A, Ramirez PT: Conservative management of a uterine tumor resembling an ovarian sex cord-stromal tumor. Gynecol Oncol 92: 347-352, 2004 Horn LC, Stegner HE: Uteriner Stromatumor mit ovarieller Keimstrangdifferenzierung. Pathologe 16: 421-425,1995 Isaacson PG: Update on MALT lymphomas. Best Pract Res Clin Haematol 18: 57-68, 2005 Kabbani W, Deavers MT, Malpica A, Burke TW, Liu J, Ordonez NG, Jhingran A, Silva EG: Uterine tumor resembling ovarian sex-cord tumor: report of a case mimicking cervical adenocarcinoma. Int J Gynecol Pathol 22: 297-302, 2003 Kantelip B, Cloup N, Dechelotte P: Uterine tumor resembling ovarian sex cord tumors: Report of a case with ultrastructural study. Hum Pathol 17: 91-94, 1986 Krishnamurthy S, Jungbluth AA, Busam KJ, Rosai J: Uterine tumors resembling ovarian sex-cord tumors have an immunophenotype consistent with true sex-cord differentiation. Am J Surg Pathol 22: 1078-1082, 1998 Mazur MT, Kraus FT: Histogenesis of morphologic variations in tumors of the uterine wall. Am J Surg Pathol 4: 59-74, 1980 Suzuki C, Matsumoto T, Fukunaga M, Itoga T, Furugen Y, Kurosaki Y, Suda K, Kinoshita K: Uterine tumors resembling ovarian sex-cord tumors producing parathyroid hormonerelated protein of the uterine cervix. Pathol Int 52: 164-168, 2002 Wang J, Blakey GL, Zhang L, Bane B, Torbenson M, Li S: Uterine tumor resembling ovarian sex cord tumor: report of a case with t(X;6)(p22.3;q23.1, and t(4;18)(q21.1;q21.3). Diagn Mol Pathol 12: 174-180, 2003 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 153 EP 156 PG 4 ER PT J AU Csire, M Mikala, G Jako, J Masszi, T Janosi, J Dolgos, J Fule, T Tordai, A Berencsi, Gy Valyi-Nagy, I AF Csire, Marta Mikala, Gabor Jako, Janos Masszi, Tamas Janosi, Judit Dolgos, Janos Fule, Tibor Tordai, Attila Berencsi, Gyorgy Valyi-Nagy, Istvan TI Persistent Long-Term Human Herpesvirus 6 (HHV-6) Infection in a Patient with Langerhans Cell Histiocytosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Langerhans cell histiocytosis; eosinophilic granuloma; HHV-6; human herpesviruses; herpesvirus reactivation ID Langerhans cell histiocytosis; eosinophilic granuloma; HHV-6; human herpesviruses; herpesvirus reactivation AB Langerhans cell histiocytosis (eosinophilic granuloma) was first diagnosed in the adolescence of a male patient presented. Several years later persisting human herpesvirus 6 (HHV-6) infection was recognized. The HHV-6 infection could be verified retrospectively in his historical histological samples; the continuous presence of HHV-6 could be established through 17 years of disease course. The patient was operated several times during this period for painful relapses, and developed diabetes insipidus. At variable time points during the clinical course, Varicella zoster (VZV), Epstein-Barr virus (EBV) and human herpesvirus 8 (HHV-8) infections were temporarily detected from blood samples and biopsy specimens. HHV-6 was the only virus continuously identified throughout the entire follow-up period. Antiviral therapy effectively cleared EBV and HHV-8, but HHV-6 remained detectable throughout the disease course. Since DNA sequences of HHV-6 could be detected in the pathologic histiocytes of eosinophilic granuloma, and from other samples taken later on, it is suggested that long-term HHV-6 infection may be associated with development or progression of Langerhans cell histiocytosis. C1 [Csire, Marta] National Center for Epidemiology, Division of Virology, 2-6. Gyali ut, H-1097 Budapest, Hungary. [Mikala, Gabor] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Jako, Janos] National Medical Center, 1st Department of Internal MedicineBudapest, Hungary. [Masszi, Tamas] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Janosi, Judit] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Dolgos, Janos] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Fule, Tibor] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular PathologyBudapest, Hungary. [Tordai, Attila] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Berencsi, Gyorgy] National Center for Epidemiology, Division of Virology, 2-6. Gyali ut, H-1097 Budapest, Hungary. [Valyi-Nagy, Istvan] National Medical Center, 1st Department of Internal MedicineBudapest, Hungary. RP Csire, M (reprint author), National Center for Epidemiology, Division of Virology, H-1097 Budapest, Hungary. EM mcsire@freemail.hu CR Ahlqvist J, Fotheringham J, Akhyani N, Yao K, Fogdell-Hahn A, Jacobson S: Differential tropism of human herpesvirus 6, HHV-6, variants and induction of latency by HHV-6A in oligodendrocytes. J Neurovirol 11:384-394, 2005. Aubin JT, Agut H, Collandre A, Yamanishi K, Chandran B, Montagnier L, Huraux J M: Antigenic and genetic differentiation of the two putative types of herpes virus 6. J Virol Methods 41:223-234, 1993. Chang Y, Cesarman E, Pessin MS, Lee F, Culpepper J, Knowles D M, Moore PS: Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science 266:1865-1869, 1994. De Bolle L, Naesens L, De Clercq E: Update on human herpesvirus 6 biology, clinical features, and therapy. Clin Microbiol Rev 18:217-245, 2005. Donati D, Martinelli E, Cassiani-Ingoni R, Ahlqvist J, Hou J, Major EO, Jacobson S: Variant-specific tropism of human herpesvirus 6 in human astrocytes. J Virol 79:9439-9448, 2005. Glotzbecker MP, Carpentieri DF, Dormans JP: Langerhans cell histiocytosis: a primary viral infection of bone? Human herpes virus 6 latent protein detected in lymphocytes from tissue of children. J Pediatr Orthop 24:123-129, 2004. Glotzbecker MP, Dormans JP, Pawel BR, Wills BP, Josi Y, Elkan M, Hodinka RL: Langerhans cell histiocytosis and human herpesvirus 6, HHV-6), an analysis by real-time polymerase chain reaction. J Ortop Res 24:313-320, 2006. Ishikawa K, Hasegawa K, Naritomi T, Kanai N, Ogawa M, Kato Y, Kobayashi M, Torii N, Hayashi N: Prevalence of herpesviridae and hepatitis virus sequences in the livers of patients with fulminant hepatitis of unknown etiology in Japan. J Gastroenterol 37:523-530, 2002. Jenson HB, McClain KL, Leach CT, Deng JH, Gao SJ: Evaluation of human herpesvirus type 8 infection in childhood langerhans cell histiocytosis. Am J Hematol 64:237-241, 2000. Kakimoto M, Hasegawa A, Fujita S, Yasukawa M: Phenotypic and functional alterations of dendritic cells induced by human herpesvirus 6 infection. J Virol 76:10338-10345, 2002. Leahy MA, Krejci SM, Frednash M, Stockert SS, Wilson H, Huff JC, Wetson W, Brice SL: Human herpesvirus 6 is present in lesions of Langerhans cell histiocytosis J Invest Dermatol 101:642-645, 1993. Mitchell SM, Fox JD, Tedder RS, Gazzard BG, Lightman S: Vitreous fluid sampling and viral genome detection for the diagnosis of viral retinitis in patients with AIDS. J Med Virol 43:336-340, 1994. Pozo F, Tenorio A: Detection and typing of lymphotropic herpesviruses by multiplex polymerase chain reaction. J Virol Methods 79:9-19, 1999. Renne R, Zhong W, Herndier B, Kedes D, McGrath M, Ganem D: Lytic growth of Kaposi’s sarcoma-associated herpesvirus, human herpesvirus 8, in B cell lymphoma cells in culture. Nat Med 2:342-346, 1996. Tetsushi Y: Human herpesvirus 6 infection in haematopoetic stem cell transplant patients. Br J Haematol 124:421-432, 2004. Ward KN: The natural history and laboratory diagnosis of human herpesviruses-6 and -7 infections in the immunocompetent. J Clin Virol 32:183-193, 2005. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 157 EP 160 PG 4 ER PT J AU Andreadis, D Nomikos, A Barbatis, C AF Andreadis, Dimitrios Nomikos, Alexandros Barbatis, Calypso TI Metastatic Renal Clear Cell Carcinoma in the Parotid Gland: A Study of Immunohistochemical Profile and Cell Adhesion Molecules (CAMs) Expression in Two Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE metastatic renal cell carcinoma; parotid gland; cell adhesion molecules; immunohistochemistry ID metastatic renal cell carcinoma; parotid gland; cell adhesion molecules; immunohistochemistry AB Metastasis of renal cell carcinoma (RCC) may involve any organ, including the parotid salivary gland. While the definition of salivary gland neoplasms with clear cell transformation can be concluded by the synchronous presence of areas showing typical morphology, sometimes the definition of a metastatic RCC in the parotid is difficult and the application of immunohistochemistry may support the clinical and radiographic observations in the final diagnosis. The aim of this paper was to describe the heterogeneous immunohistochemical features and, furthermore, to characterize the pattern of expression of cell adhesion molecules (CAMs) E-cadherin, beta4-integrin, desmoglein-2, ICAM-1 and CD44s (HCAM) in two cases of metastatic parotid RCC. C1 [Andreadis, Dimitrios] Hellenic Red Cross Hospital of Athens, Department of Histopathology, 4 Sarantaporou St, 155-61 Athens, Greece. [Nomikos, Alexandros] Hellenic Red Cross Hospital of Athens, Department of Histopathology, 4 Sarantaporou St, 155-61 Athens, Greece. [Barbatis, Calypso] Hellenic Red Cross Hospital of Athens, Department of Histopathology, 4 Sarantaporou St, 155-61 Athens, Greece. RP Andreadis, D (reprint author), Hellenic Red Cross Hospital of Athens, Department of Histopathology, 155-61 Athens, Greece. EM uromorulin@hotmail.com CR Maiorano E, Altini M, Favia G: Clear cell tumors of the salivary glands, jaws, and oral mucosa. Semin Diagn Pathol 14:203-212, 1997 Hessan H, Strauss M, Sharkey FE: Urogenital tract carcinoma metastatic to the head and neck. Laryngoscope 96:1352-1356, 1986 Adil G, Murat D, Ayhan O, Ozgur TM, Ibrahim Y, Fuat PA, Rifki F: Renal cell carcinoma metastatic to the parotid gland. 83:861-862, 1999 Solitary metastasis of renal cell carcinoma to the parotid gland 10 years after radical nephrectomy. Int J Urol 11:894-896, 2004 Parotid gland metastasis from renal cell carcinoma. Laryngoscope 112:453-456, 2002 Clear cell tumors of salivary glands. Ann Otol Rhinol Laryngol 89:196-197, 1980 Ellis GL, Auclair PL: Tumors of the salivary glands. Washington DC: Armed Forces Institute of Pathology, 1996, pp 281-289 Seijas BP, Franco FL, Sastre RM, Garcia AA, Cempanos JLL: Metastatic renal cell carcinoma presenting as a parotid tumor. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 99:554-557, 2005 Nappi O, Mills SE, Swanson PE, Wick MR: Clear cell tumors of unknown nature and origin: a systematic approach to diagnosis. Semin Diagn Pathol 14:203-212, 1997 Albelda SM: Biology of disease: Role of integrins and other cell adhesion molecules in tumor progression and metastasis. Lab Invest 68:4-17, 1993 Pignatelli M, Vessey CJ: Adhesion molecules: Novel molecular tools in tumor pathology. Hum Pathol 25:849-856, 1994 Schafer S, Koch PJ, Franke WW: Identification of the ubiquitous human desmoglein, Dsg2, and the expression catalogue of the desmoglein subfamily of desmosomal cadherins. Exp Cell Res 211:391-399, 1994 Mercurio AM, Rabinovitz I: Towards a mechanistic understanding of tumor invasion—lessons from the alpha6beta4 integrin. Semin Cancer Biol 11:129-141, 2001 Sneath RJS, Mangham DC: The normal structure and function of CD44 and its role in neoplasia. J Clin Pathol: Mol Pathol 51:191- 200, 1998 Thiery JP: The Ig superfamily of adhesion molecules. In Kreis T, Vale R: Guidebook extracellular matrix, anchor and adhesion proteins. 2nd edition, Oxford University Press, 1999, pp 125-128 Ozolek JA, Bastacky SI, Myers EN, Hunt JL: Immunophenotypic comparison of salivary gland oncocytoma and metastatic renal cell carcinoma. Laryngoscope 115:1097-1100, 2005 Rosai J: Rosai and Ackerman’s Surgical Pathology. Ninth Edition. Vol.1. Mosby, London, 2004 Dabbs D: Diagnostic Immunohistochemistry. Churchill Livingstone, New York, 2004 Wang B, Brandwein M, Gordon R, Robinson R, Urken M, Zarbo RJ: Primary salivary clear cell tumors—a diagnostic approach: a clinicopathologic and immunohistochemical study of 20 patients with clear cell carcinoma, clear cell myoepithelial carcinoma, and epithelial-myoepithelial carcinoma. Arch Pathol Lab Med 126:676- 685, 2002 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 161 EP 165 PG 5 ER PT J AU Perez, EdCD Magrin, J de Almeida, PO Kowalski, PL AF Perez, Elias da Cruz Danyel Magrin, Jose de Almeida, Paes Oslei Kowalski, Paulo Luiz TI Multiple Cutaneous Metastases from a Parotid Adenoid Cystic Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE adenoid cystic carcinoma; cutaneous metastasis; cancer; cytokeratin; histopathology; immunohistochemistry ID adenoid cystic carcinoma; cutaneous metastasis; cancer; cytokeratin; histopathology; immunohistochemistry AB Cutaneous metastasis from salivary gland adenoid cystic carcinoma is extremely rare. We report a case of a 39-year-old man that presented multiple cutaneous metastases from a parotid salivary gland adenoid cystic carcinoma. The clinical, histopathological and immunohistochemical features are described and discussed. This case shows the importance of a detailed and periodical skin examination in patients treated for salivary gland adenoid cystic carcinoma. C1 [Perez, Elias da Cruz Danyel] A. C. Camargo Cancer Hospital, Department of Stomatology, Rua Professor Antonio Prudente, 211. Liberdade, 01509-900 Sao Paulo, Brazil. [Magrin, Jose] A. C. Camargo Cancer Hospital, Department of Head and Neck Surgery and OtorhinolaryngologySao Paulo, Brazil. [de Almeida, Paes Oslei] University of Campinas, School of Dentistry of Piracicaba, Department of Oral PathologySao Paulo, Brazil. [Kowalski, Paulo Luiz] A. C. Camargo Cancer Hospital, Department of Head and Neck Surgery and OtorhinolaryngologySao Paulo, Brazil. RP Perez, EdCD (reprint author), A. C. Camargo Cancer Hospital, Department of Stomatology, 01509-900 Sao Paulo, Brazil. EM perezdec2003@yahoo.com.br CR Alcedo JC, Fabrega JM, Arosemena JR, Urrutia A: Imatinib mesylate as treatment for adenoid cystic carcinoma of the salivary glands: report of two successfully treated cases. Head Neck 26:829-831, 2004 Araujo VC, Sousa SOM, Carvalho, Araujo NS: Application of immunohistochemistry to the diagnosis of salivary gland tumors. Appl Immunohistochem Mol Morphol 8:195-202, 2000 Chang CH, Liao YL, Hong HS: Cutaneous metastasis from adenoid cystic carcinoma of the parotid gland. Dermatol Surg 29:775-779, 2003 Doganay L, Bilgi S, Aygit C, Altaner S: Primary cutaneous adenoid cystic carcinoma with lung and lymph node metastases. J Eur Acad Dermatol Venereol 18:369-371, 2004 Ellis GL, Auclair P: Tumors of the salivary glands: Atlas of Tumor Pathology. Washington, DC: Armed Forces Institute of Pathology, 3rd Series, Fascicle 17, 1996 Holst VA, Marshall CE, Moskaluk CA and Frierson HF Jr: KIT protein expression and analysis of c-kit gene mutation in adenoid cystic carcinoma. Mod Pathol 12:956-960, 1999 Hotte SJ, Winquist EW, Lamont E, MacKenzie M, Vokes E, Chen EX, Brown S, Pond GR, Murgo A, Siu LL: Imatinib mesylate in patients with adenoid cystic cancers of the salivary glands expressing c-kit: A Princess Margaret Hospital phase II consortium study. J Clin Oncol 23:585-90, 2005 Khan AJ, DiGiovanna MP, Ross DA, Sasaki CT, Carter D, Son YH, Haffty BG: Adenoid cystic carcinoma: a retrospective clinical review. Int J Cancer 96:149-158, 2001 Kokemueller H, Eckardt A, Brachvogel P, Hausamen JE: Adenoid cystic carcinoma of the head and neck – a 20 years experience. Int J Oral Maxillofac Surg 33:25-31, 2004 Lin CH, Yen RF, Jeng YM, Tzen CY, Hsu C, Hong RL: Unexpected rapid progression of metastatic adenoid cystic carcinoma during treatment with imatinib mesylate. Head Neck 27:1022-1027, 2005 Nakamura M, Miyachi Y: Cutaneous metastasis from an adenoid cystic carcinoma of the lacrimal gland. Br J Dermatol 141:350-351, 1999 Nascimento AG, Amaral ALP, Prado LAF, Kligerman J, Silveira TR: Adenoid cystic carcinoma of the salivary glands. A study of 61 cases with clinicopathologic correlation. Cancer 57:312-319, 1986 Schwartz RA: Cutaneous metastatic disease. J Am Acad Dermatol 33:161-182, 1995 Spiers AS, Esseltine DL, Ruckdeschel JC, Davies JN, Horton J: Metastatic adenoid cystic carcinoma of salivary glands. Case reports and review of the literature. Cancer Control 3:336-342, 1996 Spiro RH: Distant metastasis in adenoid cystic carcinoma of salivary origin. Am J Surg 174:495-498, 1997 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 167 EP 169 PG 3 ER PT J AU Sari, A Uyaroglu, AM Ermete, M Oder, M Girgin, C Dincer, C AF Sari, Aysegul Uyaroglu, Ali Mehmet Ermete, Murat Oder, Mehmet Girgin, Cengiz Dincer, Cetin TI Microcystic Urothelial Carcinoma of the Urinary Bladder Metastatic to the Penis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Urothelial carcinoma; microcystic variant; primary bladder cancer; penile metastasis ID Urothelial carcinoma; microcystic variant; primary bladder cancer; penile metastasis AB Metastatic spread of primary bladder cancer to the penis is an extremely rare event. Microcystic urothelial carcinoma is a very rare variant of urothelial carcinoma. Due to its rareness and insufficient clinical follow-up data, the prognosis of microcystic urothelial carcinoma is still not clear. Here in we report a case of a penile metastasis from microcystic urothelial carcinoma of urinary bladder, in a 56-year-old man who died 6 months after radical cystoprostatectomy and total penectomy. To the best of our knowledge this is the first case report of microcystic variant of urothelial carcinoma which has metastasized to the penis. C1 [Sari, Aysegul] Izmir Ataturk Training and Research Hospital, Department of Pathology, Ataturk caddesi, Firat apt. No: 184, Kat: 3, Daire: 6, 35220 Izmir, Alsancak, Turkey. [Uyaroglu, Ali Mehmet] Izmir Ataturk Training and Research Hospital, Department of Pathology, Ataturk caddesi, Firat apt. No: 184, Kat: 3, Daire: 6, 35220 Izmir, Alsancak, Turkey. [Ermete, Murat] Izmir Ataturk Training and Research Hospital, Department of Pathology, Ataturk caddesi, Firat apt. No: 184, Kat: 3, Daire: 6, 35220 Izmir, Alsancak, Turkey. [Oder, Mehmet] Izmir Ataturk Training and Research Hospital, Department of 1st UrologyIzmir, Turkey. [Girgin, Cengiz] Izmir Ataturk Training and Research Hospital, Department of 1st UrologyIzmir, Turkey. [Dincer, Cetin] Izmir Ataturk Training and Research Hospital, Department of 1st UrologyIzmir, Turkey. RP Sari, A (reprint author), Izmir Ataturk Training and Research Hospital, Department of Pathology, 35220 Izmir, Turkey. EM aysegulakder@gmail.com CR Khan MA, Tao W, Mathews P, Potluri BS: Penile metastasis arising from transitional cell carcinoma of the urinary bladder. Urol Int 66:162-163, 2001 Haddad FS: Tumors of the bladder. Annu Rep Orient Hosp 12:8-19, 1959 Young RH, Eble JN: Unusual forms of carcinoma of the urinary bladder. Hum Pathol 22:948-965, 1991 Young RH, Zukerberg LR: Microcystic transitional cell carcinomas of the urinary bladder. A report of four cases. Am J Clin Pathol 96:635-639, 1991 Paz A, Rath-Wolfson L, Lask D, Koren R, Manes A, Mukamel E, Gal R: The clinical and histological features of transitional cell carcinoma of the bladder with microcysts: analysis of 12 cases. Br J Urol 79:722-725, 1997 Leroy X, Leteurtre E, De La Taille A, Augusto D, Biserte J, Gosselin B: Microcystic transitional cell carcinoma: a report of 2 cases arising in the renal pelvis. Arch Pathol Lab Med 126:859-861, 2002 Radopoulos D, Kalyvas K, Kotakidou R, Panagiotopoulou K, Katsikas V, Papathanasiou M: Case report: microcystic transitional cell carcinoma of the urinary bladder. Int Urol Nephrol 37:291-293, 2005 Pomara G, Pastina I, Simone M, Casale P, Marchetti G, Francesca F: Penile metastasis from primary transitional cell carcinoma of the renal pelvis: first manifestation of systemic spread. BMC Cancer 4:90, 2004 Demuren OA, Koriech O: Isolated penile metastasis from bladder carcinoma. Eur Radiol 9:1596-1598, 1999 Osther PJ, Lontoft E: Metastasis to the penis. Case reports and review of the literature. Int Urol Nephrol 23:161-167, 1991 Haddad FS: Penile metastases secondary to bladder cancer. Review of the literature. Urol Int 39:125-142, 1984 Berger AP, Rogatsch H, Hoeltl L, Steiner H, Bartsch G, Hobisch A: Late penile metastasis from primary bladder carcinoma. Urology 62:145, 2003 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2007 VL 13 IS 2 BP 170 EP 173 PG 4 ER PT J AU Beser, RA Tuzlali, S Guzey, D Dolek Guler, S Hacihanefioglu, S Dalay, N AF Beser, Rehber Asli Tuzlali, Sitki Guzey, Deniz Dolek Guler, Semra Hacihanefioglu, Seniha Dalay, Nejat TI HER-2, TOP2A and Chromosome 17 Alterations in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TOP2A; HER-2; breast cancer ID TOP2A; HER-2; breast cancer AB HER-2 amplification is a biomarker for identifying patients who respond to trastuzumab and has been evaluated as a factor predicting the response to anthracyclines. The relationship between HER-2 and response to anthracycline therapy may also be the result of the close localization of TOP2A on 17q. It has been a matter of debate whether these two genes, HER-2 and TOP2A, behave separately on different amplicons or act together thus making it possible to predict the TOP2A status from the HER-2 status. In this study TOP2A, HER-2 and chromosome 17 aneusomy were investigated by fluorescent in situ hybridization (FISH) in 50 consecutive breast cancer patients. HER-2 amplification was detected in 11 patients (22%) and TOP2A changes were seen in 6 patients (12%); two amplifications and two deletions were observed in HER-2-amplified cases and two deletions in HER-2-nonamplified cases. Three of the TOP2A-deleted cases had polysomy 17. HER-2 copy number was higher than the TOP2A copy number in one patient with co-amplification. Polysomy was observed in 9 cases (18%) and monosomy in 6 cases (12%). Aneusomy was the sole anomaly in 11 patients (22%). We conclude that the TOP2A status cannot be predicted from the HER-2 status and evaluation of the TOP2A status only in patients with HER-2 overexpression may lead to missing cases with TOP2A deletion with possible resistance to therapy. Other factors modulating topo II activity may also affect the response to therapy. Studies evaluating different parameters that can modulate topo II activity and the response to the drugs targeting the enzyme are necessary. C1 [Beser, Rehber Asli] Istanbul Medical Faculty, Istanbul University, Oncology Institute, 34093 Istanbul, Capa, Turkey. [Tuzlali, Sitki] Istanbul Medical Faculty, Department of PathologyIstanbul, Turkey. [Guzey, Deniz] Vakif Gureba Hospital, Department of SurgeryIstanbul, Turkey. [Dolek Guler, Semra] Istanbul Medical Faculty, Istanbul University, Oncology InstituteIstanbul, Turkey. [Hacihanefioglu, Seniha] Istanbul Medical Faculty, Istanbul University, Department of Medical BiologyIstanbul, Turkey. [Dalay, Nejat] Istanbul Medical Faculty, Istanbul University, Oncology Institute, 34093 Istanbul, Capa, Turkey. RP Dalay, N (reprint author), Istanbul Medical Faculty, Istanbul University, Oncology Institute, 34093 Istanbul, Turkey. EM ndalay@yahoo.com CR Bhargava R, Lal P, Chen B: HER-2/neu and topoisomerase IIα gene amplification and protein expression in invasive breast carcinomas: chromogenic in situ hybridization and immunohistochemical analyses. Am J Clin Pathol 123:889-895, 2005 Biersack H, Jensen S, Gromova I, Nielsen IS, Westergaard O: Active heterodimers are formed from human topoisomerase II alpha and II beta isoforms. Proc Natl Acad Sci USA 93: 8288- 8293, 1996 Bose S, Mohammed M, Shintaku P, Rao PN: Her-2/neu gene amplification in low to moderately expressing breast cancers: possible role of chromosome 17/Her-2/neu polysomy. Breast J 7:337-344, 2001 Burden DA, Osheroff N: Mechanism of action of eukaryotic topoisomerase II and drugs targeted to the enzyme. Biochim Biophys Acta 1400:139-154, 1999 Cardoso F, Durbecq V, Larsimont D, Paesmans M, Leroy JY, Rouas G, Sotiriou C, Renard N, Richard V, Picaart MJ, Di Leo A: Correlation between complete response to anthracyclinebased chemotherapy and topoisomerase II alpha gene amplification and protein overexpression in locally advanced/metastatic breast cancer. Int J Oncol 24: 201-209, 2004 Coon JS, Marcus E, Gupta-Burt S, Seelig S, Jacobson K, Chen S, Renta V, Fronda G, Preisler HD: Amplification and overexpression of topoisomerase II predict response to anthracyclinebased therapy in locally advanced breast cancer. Clin Cancer Res 8: 1061-1067, 2002 Di Leo A, Gancberg D, Larsimont D, Tanner M, Jarvinen T, Rouas G, Dolci S, Leroy JY, Paesmans M, Isola J, Piccart MJ: HER-2 amplification and topoisomerase II? gene aberrations as predictive markers in node-positive breast cancer patients randomly treated either with an anthracycline-based therapy or with cyclophosphamide, methotrexate, and 5-fluorouracil. Clin Cancer Res 8: 1107-1116, 2002 Downs-Kelly E, Yoder BJ, Stoler M, Tubbs RR, Skacel M, Grogan T, Roche P, Hicks DG: The influence of polysomy 17 on HER2 gene and protein expression in adenocarcinoma of the breast: A fluorescent in situ hybridization, immunohistochemical, and isotopic mRNA in situ hybridization study. Am J Surg Pathol 29:1221-1227, 2005 Durbecq V, Desmed C, Paesmans M, Cardoso F, Di Leo A, Mano M, Rouas G, Leroy JY, Sotiriou C, Piccart M, Larsimont D: Correlation between topoisomerase-IIalpha gene amplification and protein expression in HER-2 amplified breast cancer. Int J Oncol 25:1473-1479, 2004 Faneyte IF, Chrama JO, Peterse JL, Remijse JL, Rodenhuis S, Van der Vijver MJ: Breast cancer response to neoadjuvant chemotherapy: predictive markers and relation with outcome. Br J Cancer 88: 406-412, 2003 Fehm T, Morrison L, Saboorian H, Hynan L, Tucker T, Uhr J: Patterns of aneusomy for three chromosomes in individual cells from breast cancer tumors. Breast Cancer Res Treat 75: 227- 239, 2002 Greenlee RT, Hill-Harmon MB, Murray T, Thun M: Cancer Statistics 2001. CA Cancer J Clin 51:15-36, 2001 Harris LN, Yang L, Liotcheva V, Pauli S, Iglehart JD, Colvin OM, Hsieh TS: Induction of topoisomerase II activity after ERbB2 activation is associated with a differential response to breast cancer chemotherapy. Clin Cancer Res 7: 1497-1504, 2001 Hicks DG, Yoder BJ, Pettay J, Swain E, Tarr S, Hartke M, Skacel M, Crowe JP, Budd GT, Tubbs RR: The incidence of topoisomerase II-alpha genomic alterations in adenocarcinoma of the breast and their relationship to human epidermal growth factor receptor-2 gene amplification: a fluorescence in situ hybridization study. Hum Pathol 36:348-356, 2005 Hoffken K: The European Experience. J Clin Oncol 19: 112- 117, 2001 Jacobson KK, Morrison LE, Henderson BT, Blondin BA, Wilber KA, Legator MS, O’Hare A, Van Stedum SC, Proffitt JH, Seelig SA, Coon JS Gene copy mapping of the ERBB2/TOP2A region in breast cancer. Genes Chromosomes Cancer 40:19-31, 2004 Jarvinen TA, Holli K, Kuukasjarvi T, Isola JJ: Predictive value of topoisomerase II alpha and other prognostic factors for epirubicin chemotherapy in advanced breast cancer. Br J Cancer 77: 2267-2273, 1998 Jarvinen TAH, Liu ET: HER-2/neu and Topoisomerase IIa – simultaneous drug targets in cancer. Comb Chem High Throughput Screen 6: 79-99, 2003 Jarvinen TAH, Liu ET: Topoisomerase IIα gene, TOP2A, amplification and deletion in cancer – more common than anticipated. Cytopathology 4:309-313, 2003 Jarvinen TAH, Tanner M, Barlund M, Borg A, Isola J: Characterization of topoisomerase IIα gene amplification and deletion in breast cancer: Genes Chromosomes Cancer 26:142-150, 1999 Jarvinen TAH, Tanner M, Rantanen V, Barlund M, Borg A, Grenman S, Isola J: Amplification and deletion of topoisomerase IIα associate with ErbB-2 amplification and affect sensitivity to topoisomerase II inhibitor doxorubicin in breast cancer. Am J Pathol 156: 839-847, 2000 Kellner U, Sehested M, Jensen PB, Gieseler F, Rudolph P: Culprit and victim – DNA topoisomerase II. Lancet Oncol 3:235-243, 2002 Knoop AS, Knudsen H, Balslev E, Rasmussen BB, Overgaard J, Nielsen KV, Schonau A, Gunnarsdottir K, Olsen KE, Mouridsen H, Ejlertsen B; Danish Breast Cancer Cooperative Group: Retrospective analysis of topoisomerase IIα amplifications and deletions as predictive markers in primary breast cancer patients randomly assigned to cyclophosphamide, methotrexate, and fluorouracil or cyclophosphamide, epirubicin, and fluorouracil: Danish Breast Cancer Cooperative Group. J Clin Oncol 23:7483-7490, 2005 MacGrogan G, Rudolph P, de Mascarel I, Maurica L, Durand M, Avril A, Dilhudy JM, Robert J, Mathoulin-Pelissier S, Picot V, Floquet A, Serankowski G, Coindre JM: DNA topoisomerase II alpha expression and the response to primary chemotherapy in breast cancer. Br J Cancer 89: 666-667, 2003 Martin-Richard M, Munoz M, Albanell J, Colomo L, Bellet M, Rey MJ, Tabernero J, Alonso C, Cardesa A, Gascon P, Fernandez PL: Serial topoisomerase II expression in primary breast cancer and response to neoadjuvant anthracycline-based chemotherapy. Oncology 66: 388-394, 2004 Mueller RE, Parkes RK, Andrulis I, O’Malley FP: Amplification of the TOP2A gene does not predict high levels of topoisomerase II alpha protein in human breast tumor samples. Genes Chromosomes Cancer 39:288-297, 2004 Nakopoulou L, Giannopoulou I, Trafalis D, Gakiopoulou H, Keramopoulos A, Davaris P: Evaluation of numeric alterations of chromosomes 1 and 17 by in situ hybridisation in invasive breast carcinoma with clinicopathologic parameters. Appl Immunohistochem Mol Morphol 10:20-28, 2002 Paik S, Bryant J, Park C, Fisher B, Tan-Chiu E, Hyams D, Fisher ER, Lippman ME, Wickerham DL, Wolmark N: erbB2 and response to doxorubicin in patients with axillary lymph node-positive, hormone receptor-negative breast cancer. J Natl Cancer Inst 90: 1320-1327,1998 Petit T, Wilt M, Velten M, Millon R, Rodier JF, Borel C, Mors R, Haegele P, Eber M, Ghnassia JP: Comparative value of tumour grade, hormonal receptors, Ki-67, HER-2 and topoisomerase II alpha status as predictive markers in breast cancer patients treated with neoadjuvant anthracycline-based chemotherapy. Eur J Cancer 40: 205-211, 2004 Ross JS, Fletcher JA: HER-2/neu, c-erb-B2, gene and protein in breast cancer. Am J Clin Pathol 112(Suppl 1):53-67, 1999 Ross JS, Fletcher JA, Bloom KJ, Linette GP, Stec J, Symmans WS, Pusztai L, Hortobagyi GN. Targeted therapy in breast cancer: the HER-2/neu gene and protein. Mol Cell Proteomics 3: 379-398, 2004 Salido M, Tusquets I, Corominas JM, Suarez M, Espinet B, Corzo C, Bellet M, Fabregat X, Serrano S, Sole F: Polysomy of chromosome 17 in breast cancer tumors showing an overexpression of ERBB2: a study of 175 cases using fluorescence in situ hybridization and immunohistochemistry. Breast Cancer Res 7:267-273, 2005 Tanner M, Jarvinen P, Isola J: Amplification of HER-2/neu and topoisomerase IIα in primary and metastatic breast cancer. Cancer Res 61: 5345-5348, 2001 Thor AD, Berry DA, Budman DR, Muss HB, Kute T, Henderson IC, Barcos M, Cirrincione C, Edgerton S, Allred C, Norton L, Liu ET: ERbB-2, p53, and efficacy of adjuvant therapy in lymph nodepositive breast cancer. J Natl Cancer Inst 90: 1346-1360, 1998 Varshney D, Zhou YY, Geller SA, Alsabeh R: Determination of HER-2 status and chromosome 17 polysomy in breast carcinomas comparing HercepTest and PathVysion FISH assay. Am J Clin Pathol 121:70-77, 2004 Wang S, Hossein Saboorian M, Frenkel EP, Haley BB, Siddiqui MT, Gokaslan S, Hynan L, Ashfaq R: Aneusomy 17 in breast cancer: its role in HER-2/neu protein expression and implication for clinical assessment of HER-2/neu status. Mod Pathol 15:137-45, 2002 Watters AD, Going JJ, Cooke TG, Bartlett JMS: Chromosome 17 aneusomy is associated with poor prognostic factors in invasive breast carcinoma. Breast Cancer Res Treat 77: 109-114, 2003 Zhang F, Yang Y, Smith T, Kau SW, McConathy JM, Esteva FJ, Kuerer HM, Symmans WF, Buzdar AU, Hortobagyi GN, Pusztai L: Correlation between HER-2 expression and response to neoadjuvant chemotherapy with 5-fluorouracil, doxorubicin, and cyclophosphamide in patients with breast carcinoma. Cancer 97:1758-1765, 2003 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 180 EP 185 PG 6 ER PT J AU Riesz, P Lotz, G Paska, Cs Szendroi, A Majoros, A Nemeth, Zs Torzsok, P Szarvas, T Kovalszky, I Schaff, Zs Romics, I Kiss, A AF Riesz, Peter Lotz, Gabor Paska, Csilla Szendroi, Attila Majoros, Attila Nemeth, Zsuzsanna Torzsok, Peter Szarvas, Tibor Kovalszky, Ilona Schaff, Zsuzsa Romics, Imre Kiss, Andras TI Detection of Bladder Cancer from the Urine using Fluorescence in situ Hybridization Technique SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bladder cancer; cystoscopy; fluorescence in situ hybridization; molecular pathology ID bladder cancer; cystoscopy; fluorescence in situ hybridization; molecular pathology AB The authors report on their first experiences with the UroVysion fluorescence in situ hybridization (FISH) kit developed for the detection of bladder cancer. This new non-invasive diagnostic application of the FISH technique in the field of urology was elaborated to replace cystoscopy. The special urine examination method detects genetic alterations of the urothelial cells found in the urine, using fluorescent directlabeled DNA probes binding to the peri-centromeric regions of chromosomes 3, 7 and 17 as well as on the 9p21 locus. We aimed to evaluate the utility of UroVysion test in the light of the histological diagnosis. Urine samples from 43 bladder cancer patients and 12 patients with no or benign alterations were studied using a new application of FISH technique: the UroVysion reagent kit. The obtained FISH results were compared with the histological findings of the transurethral surgical resection specimens. The study rated the specificity and sensitivity of the technique 100% and 87%, respectively. Therefore, the technique could well fit into the diagnostic process of bladder carcinomas. Statistical analyses showed significant correlation between tumor progression and the severity of the genetic alterations detected by this FISH technique. Furthermore, positive correlation was found between tumor grade and the proportion of tumor cells showing genetic abnormality. The noninvasiveness, the robustness of evaluation and the high specificity/sensitivity are all in favor of this technique. The disadvantages are the higher costs of the technical background and the required future clinical studies to determine whether this technique can replace cystoscopy. C1 [Riesz, Peter] Semmelweis University, Department of UrologyBudapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Paska, Csilla] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Szendroi, Attila] Semmelweis University, Department of UrologyBudapest, Hungary. [Majoros, Attila] Semmelweis University, Department of UrologyBudapest, Hungary. [Nemeth, Zsuzsanna] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Torzsok, Peter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Szarvas, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. [Romics, Imre] Semmelweis University, Department of UrologyBudapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., H-1091 Budapest, Hungary. RP Lotz, G (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM beka@korb2.sote.hu CR Droller M.J: Bladder cancer: state-of-the-art care. Ca: Cancer J Clin 48: 269-284, 1998 Oosterlinck W, Lobel B, Jakse G, Malmstrom PU, Stockle M, Sternberg C: Guidelines on bladder cancer. Eur Urol 41: 105- 112, 2002 Heney NM, Ahmed S, Flanagan MJ, Frable W, Corder MP, Hafermann MD, Hawkins IR: Superficial bladder cancer progression et recurrence. J Urol 130: 1083-1086, 1983 Halling KC, King W, Sokolova IA, Meyer RG, Burkhardt HM, Halling AC, Cheville JC, Sebo TJ, Ramakumar S, Stewart CS, Pankratz S, O’Kane DJ, Seelig SA, Lieber MM, Jenkins RB: A comparison of cytology and fluorescence in situ hybridization for the detection of bladder cancer. J Urol 164: 1768-1775, 2000 Halling KC, King W, Sokolova IA, Karnes RJ, Meyer RG, Powell EL, Sebo TJ, Cheville JC, Clayton AC, Krajnik KL, Ebert TA, Nelson RE, Burkhardt HM, Ramakumar S, Stewart CS, Pankratz VS, Lieber MM, Blute ML, Zincke H, Seelig SA, Jenkins RB, O'Kane DJ: A comparison of BTA stat, hemoglobin dipstick, telomerase and Vysis UroVysion assays for the detection of urothelial carcinoma in urine. J Urol 167: 2001-2006, 2002 Bollmann M, Heller H, Bankfalvi A, Griefingholt H, Bollmann R: Quantitative molecular urinary cytology by fluorescence in situ hybridization: a tool for tailoring surveillance of patients with superficial bladder cancer? BJU Int 95: 1219-1225, 2005 Parkin DM, Pisani P, Ferlay J: Estimates of the worldwide incidence of 25 major cancers in 1990. Int J Cancer 80: 827- 841, 1999 Glas AS, Roos D, Deutekom M, Zwinderman AH, Bossuyt PM, Kurth KH: Tumor markers in the diagnosis of primary bladder cancer. A systematic review. J Urol 169: 1975-1982, 2003 Grossman HB: New methods for detection of bladder cancer. Semin Urol Oncol 16: 17-22, 1998 Slaton JW, Dinney CP, Veltri RW, Miller CM, Liebert M, O’Dowd GJ, Grossman HB: Deoxyribonucleic acid ploidy enhances the cytological prediction of recurrent transitional cell carcinoma of the bladder. J Urol 158: 806-811, 1997 van Poppel H, Billen J, Goethuys H, Elgamal AA, Gerits M, Mortelmans L, Blanckaert N, Baert L: Serum tissue polypeptide antigen, TPA, as a tumor marker for bladder cancer. Anticancer Res 16: 2205-2208, 1996 Zhang FF, Arber DA, Wilson TG, Kawachi MH, Slovak ML: Toward the validation of aneusomy detection by fluorescence in situ hybridization in bladder cancer: comparative analysis with cytology, cytogenetics, and clinical features predicts recurrence and defines clinical testing limitations. Clin Cancer Res 3: 2317-2328, 1997 Mao L, Schoenberg MP, Scicchitano M, Erozan YS, Merlo A, Schwab D, Sidransky D: Molecular detection of primary bladder cancer by microsatellite analysis. Science 271: 659-662, 1996 Steiner G, Schoenberg MP, Linn JF, Mao L, Sidransky D: Detection of bladder cancer recurrence by microsatellite analysis of urine. Nature Med 3: 621-624, 1997 Skacel M, Fahmy M, Brainard JA, Pettay JD, Biscotti CV, Liou LS, Procop GW, Jones JS, Ulchaker J, Zippe CD, Tubbs RR: Multitarget fluorescence in situ hybridization assay detects transitional cell carcinoma in the majority of patients with bladder cancer and atypical or negative urine cytology. J Urol 169: 2101-2105, 2003 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 187 EP 194 PG 8 ER PT J AU Suba, Zs AF Suba, Zsuzsanna TI Gender-Related Hormonal Risk Factors for Oral Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE oral cancer; fasting glucose; insulin resistance; estrogen deficiency; gender difference ID oral cancer; fasting glucose; insulin resistance; estrogen deficiency; gender difference AB Oral cancer (OC) is a neoplasm with fairly high male to female ratio in most populations. The conspicuously lower incidence of this tumor among women than man is suggestive of certain endocrine involvement in its development. The aim of the present case-control study was to clarify the origin of this gender-specific risk of OC incidence. 2660 inpatients (530 females and 2130 males) with squamous cell OC at the Department of Oral and Maxillofacial Surgery were included in a case-control study. Smoking, alcohol consumption, elevated fasting serum glucose level and menopausal histories of female cases were registered. Smoking and excessive alcohol intake proved to be strong risk factors for OC both in the male and female group. However, moderate alcohol consumption was a weaker risk factor for male patients, and it presented no risk for female cases. Elevated fasting glucose level was not a demonstrable OC risk factor among males, however, it proved to be strong risk factor for OC among female patients, especially in gingival cancer cases. The almost exclusively postmenopausal state of female OC patients and the long mean interval (17 years) between their menopause and OC diagnosis suggested an important role of estrogen deficiency in OC epidemiology. The significantly younger mean age at menopause and the significantly higher rate of hysterectomy among female OC cases in comparison with their controls also support the estrogen deficiency hypothesis. This novel hypothesis of estrogen deficiency and elevated fasting glucose as risk factors for OC in postmenopausal women may provide new insights into the etiology of oral malignancies. C1 [Suba, Zsuzsanna] Semmelweis University, Department of Oral and Maxillofacial Surgery, Maria u. 52., H-1085 Budapest, Hungary. RP Suba, Zs (reprint author), Semmelweis University, Department of Oral and Maxillofacial Surgery, H-1085 Budapest, Hungary. EM suba@szajseb.sote.hu CR Neville BW, Day TA: Tumors and precancerous lesions of the oral cavity. CA Cancer J Clin 52: 195-215, 2002 La Vecchia C, Lucchini F, Negri E et al: Trends in oral cancer mortality in Europe. Oral Oncol 40: 433-439, 2004 Krolls SO, Hoffman S: Squamous cell carcinoma of the oral soft tissues: a statistical analysis of 14,253 cases by age, sex and race of patients. JADA 92: 571-574, 1976 Rich AM, Radden BG: Squamous cell carcinoma of the oral mucosa: a review of 244 cases in Australia. J Oral Pathol 13: 459-471, 1984 Levi F, Lucchini F, Negri E et al: Trends in cancer mortality in the European Union and accession countries, 1980-2000. Ann Oncol 15: 1425-1431, 2004 Johnson NW: Etiology and risk factors for oral cancers and their correlations with smoking and alcohol consumption. Hung Oncol, in Hungarian, 45: 115-122, 2001 Suba Z, Barabas J: Prevention of oral cancer, in Hungarian). Medicina ZRT, Budapest, 2007, pp. 15-20 Ujpal M, Matos O, Bibok G, et al: Diabetes and oral tumors in Hungary: Epidemiological correlations. Diabetes Care 27: 770- 774, 2004. Suba Z, Barabas J, Szabo G, et al: Increased prevalence of diabetes and obesity in patients with salivary gland tumors. Diabetes Care 28: 228, 2005 Mealey BL, Moritz AJ: Hormonal influences: effects of diabetes mellitus and endogenous female sex steroid hormones on the periodontium. Periodontol 2000 32: 59-81, 200311. Eliasson B, Attval S, Taskinen MR, et al: The insulin resistance syndrome in smokers is related to smoking habits. Arterioscler Thromb 14: 1946-1950, 1994 12. Sierksma A, Patel H, Ouchi N, et al: Effect of moderate alcohol consumption on adiponectin, tumor necrosis factor alpha and insulin sensitivity. Diabetes Care 24: 184-189, 2004 13. Beulens JW, Stolk RP, van der Schouw YT et al: Alcohol consumption and risk of type-2 diabetes among older women. Diabetes Care 28: 2933-2938, 2005 14. Nestler JE: Obesity, insulin, sex steroids and ovulation. Int J Obes Relat Metab Disord 24, Suppl 2): S71-S73, 2000 15. Huber JC, Schneeberger C, Tempfer CB: Genetic modelling of the estrogen metabolism as risk factor of hormone-dependent disorders. Maturitas 42: 1-12, 2002 16. Liehr JG: Is estradiol a genotoxic mutagenic carcinogen? Endocrine Rev 21: 40-54, 2000 17. Fernandez E, Gallus S, Bosetti C et al: Hormone replacement therapy and cancer risk: a systematic analysis from a network of case-control studies. Int J Cancer 105: 408-412, 2003 18. Diamanti-Kandarakis E: Hormone replacement therapy and risk of malignancy. Curr Opin Obstet Gynecol 16: 73-78, 2004 19. Ettinger B, Friedman GD, Bush T, Qusenberry CP, Jr: Reduced mortality associated with long-term postmenopausal estrogen therapy. Obstet Gynecol 87: 6-12, 1996 20. Gallus S, Bosetti C, Franceschi S et al: Oesophageal cancer in women: tobacco, alcohol, nutritional and hormonal factors. Br J Cancer 85: 341-345, 2001 21. La Vecchia C, D’Avanzo B, Franceschi S et al: Menstrual and reproductive factors and gastric-cancer risk in women. Int J Cancer 59: 761-764, 1994 22. Parazzini F, La Vecchia C, Negri E et al: Case-control study of estrogen replacement therapy and risk of cervical cancer. Br Med J 315: 85-88, 1997 23. Grodstein F, Clarkson T, Manson J: Understanding the divergent data on postmenopausal hormone therapy. N Eng J Med 348: 645-650, 2003 24. Olsson H, Bladstrom AM, Ingvar C: Are smoking-associated cancers prevented or postponed in women using hormone replacement therapy? Obstet Gynecol 102: 565-570, 2003 25. Reckelhoff JF: Sex steroids, cardiovascular disease, and hypertension. Hypertension 45: 170-180, 2005 26. Austad SN: Why women live longer than men: Sex differences in longevity. Gender Med 3: 79-92, 2006 27. Ebbert JO, Janney CA, Sellers TA et al: The association of alcohol consumption with coronary heart disease mortality and cancer incidence varies by smoking history. J Gener Intern Med 20: 14-20, 2005 28. McGrath M, Michaud DS, De Vivo I: Hormonal and reproductive factors and the risk of bladder cancer in women. Am J Epidemiol 163: 236-244, 2006 29. Gago-Domingez M, Castelao JE, Yuan JM et al: Increased risk of renal cell carcinoma subsequent to hysterectomy. Cancer Epidemiol Biomarkers Prev 8: 999-1003, 1999 30. Faustini-Fustini M, Rochira V, Carani C: Oestrogen deficiency in men: where are we today? Eur J Endocrinol 140: 111-129, 1999 31. McKeown-Eyssen G: Epidemiology of colorectal cancer revisited: are serum triglycerides and plasma glucose associated with risk? Cancer Epidemiol Biomarkers Prev 3: 687-695, 1994 32. Everhart J, Wright D: Diabetes mellitus as a risk factor for pancreatic cancer: a meta-analysis. JAMA 273: 1605-1609, 1995 33. Balkau B, Kahn H, Courbon D et al: Hyperinsulinemia predicts fatal liver cancer but is inversely associated with fatal cancer at some other sites: The Paris Prospective Study. Diabetes Care 24: 843-849, 2001 34. Yeung NG, Husain I, Waterfall N: Diabetes mellitus and bladder cancer – An epidemiological relationship? Pathol Oncol Res 9: 30-31, 2003 35. Barnard RJ, Aronson WJ, Tymchuk CN et al: Prostate cancer: another aspect of the insulin-resistance syndrome. Obes Rev 3: 303-308, 2002 36. Muti P, Quattrin T, Grant BJB et al: Fasting glucose is a risk factor for breast cancer. Cancer Epidemiol Biomarkers Prev 11: 1361-1368, 2002 37. Key TJ, Applyby PN, Reeves GK: Body mass index, serum sex hormones and breast cancer risk in postmenopausal women. J Natl Cancer Inst 95: 1218-1226, 2003 38. Soliman PT, Oh JC, Schmeler KM et al: Risk factors for young premenopausal women with endometrial cancer. Obstet Gynecol 105: 575-580, 2005 39. Solomon CG, Hu FB, Dunaif A et al: Long or highly irregular menstrual cycles as a marker for risk of type-2 diabetes mellitus. JAMA 286: 2421-2426, 2001 40. Bloomgarden ZT: Second World Congress on the Insulin Resistance Syndrome: Mediators, pediatric insulin resistance, the polycystic ovary syndrome, and malignancy. Diabetes Care 28: 1821-1830, 2005 41. Godsland IF: Oestrogens and insulin secretion. Diabetologia 48: 2213-2220, 2005 42. Jones ME, Thorburn AW, Britt KL et al: Aromatase-deficient, ArKO, mice have a phenotype of increased adiposity. Proc Natl Acad Sci USA 97: 12735-12740, 2000 43. Gadducci A, Gargini A, Palla E et al: Polycystic ovary syndrome and gynecological cancers: is there a link? Gynecol Endocrinol 20: 200-208, 2005 44. Freiberg MS, Cabral HJ, Heeren TC: Alcohol consumption and the prevalence of the metabolic syndrome in the US.: A cross-sectional analysis of data from the Third National Health and Nutrition Examination Survey. Diabetes Care 27: 2954- 2959, 2004 45. Onland-Moret NC, Peeters PHM, van der Schouw YT et al: Alcohol and endogenous sex steroid levels in postmenopausal women: A Cross-Sectional Study. J Clin Endocrinol Metab 90: 1414-1419, 2005 46. Tavani A, Gallus S, La Vecchia C et al: Diet and risk of oral and pharyngeal cancer. An Italian case-control study. Eur J Cancer Prev 10: 191-195, 2001 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 195 EP 202 PG 8 ER PT J AU Perez-Gutierrez, S Gonzalez-Campora, R Amerigo-Navarro, J Beato-Moreno, A Sanchez-Leon, M Pareja Megia, MJ Virizuela-Echaburu, AJ Lopez-Beltran, A AF Perez-Gutierrez, Sofia Gonzalez-Campora, Ricardo Amerigo-Navarro, Joaquin Beato-Moreno, Antonio Sanchez-Leon, Maria Pareja Megia, Maria Jesus Virizuela-Echaburu, Antonio Juan Lopez-Beltran, Antonio TI Expression of P-glycoprotein and Metallothionein in Gastrointestinal Stromal Tumor and Leiomyosarcomas. Clinical Implications SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gastrointestinal stromal tumors; GIST; leiomyosarcomas; P-glycoprotein; metallothionein ID gastrointestinal stromal tumors; GIST; leiomyosarcomas; P-glycoprotein; metallothionein AB We investigated the expression of P-glycoprotein (P-GP) and metallothionein (MT) in a series of 92 GIST and 14 gastrointestinal leiomyosarcomas (GILMS) with the purpose to expand our knowledge on the biological bases of GIST chemo-resistance and to ascertain their significance in patients’ prognosis. P-GP expression was more frequent in GIST than in GI-LMS (83.7% vs. 21.4%, p<0.001), with no difference between low- and high-risk GIST (p=1.000) or low- and high-grade GI-LMS (p=0.538). P-GP expression was unrelated to anatomic location (gastric vs. intestinal) in GIST (39/45 vs. 35/43, p=0.770) and in GI-LMS (0/2 vs. 2/6, p=1.000). MT expression was non-significantly higher in GI-LMS than in GIST (35.7% vs. 14.1%, p=0.060), with no difference between low- and high-risk GIST (p=1.000) or low- and high-grade GI-LMS (p=1.000). MT expression was unrelated to the anatomic location (gastric vs. intestinal) in GIST (7/45 vs. 6/43) and GI-LMS (0/2 vs. 1/6) (p=1.000 and p=0.1000, respectively). Overall tumor-specific survival (p< 0.001) and disease-free survival (p<0.001) were different in GIST as compared with GI-LMS, and the number of events was higher in GI-LMS. When the survival analysis took into consideration P-GP or MT expression, the overall survival in GIST was influenced by the expression of MT (p=0.021) but not by that of P-GP (p=0.638). However, in GI-LMS, P-GP expression influenced disease-free survival (p=0.050); in addition, it is important to recognize the limited value of these results because of the low number of cases involved in the study. Differential expression of P-GP and MT might explain the known variability in response to systemic chemotherapy in these tumors. Detection of P-GP and MT seems to add certain prognostic value in GIST (MT) or GI-LMS (P-GP). C1 [Perez-Gutierrez, Sofia] Juan Ramon Jimenez Hospital, Pathology ServiceHuelva, Spain. [Gonzalez-Campora, Ricardo] Virgen Macarena University Hospital and University of Seville Medical School, Department of Pathology, Avda, Dr. Fedriani s/n, 41009 Seville, Spain. [Amerigo-Navarro, Joaquin] Hospital Torrecardenas, Pathology ServiceAlmeria, Spain. [Beato-Moreno, Antonio] University of Seville, Department of Statistic and Operations ResearchSeville, Spain. [Sanchez-Leon, Maria] Virgen Macarena University Hospital and University of Seville Medical School, Department of Pathology, Avda, Dr. Fedriani s/n, 41009 Seville, Spain. [Pareja Megia, Maria Jesus] Virgen Macarena University Hospital and University of Seville Medical School, Department of Pathology, Avda, Dr. Fedriani s/n, 41009 Seville, Spain. [Virizuela-Echaburu, Antonio Juan] Virgen Macarena University Hospital, Oncology ServiceSeville, Spain. [Lopez-Beltran, Antonio] Reina Sofia University Hospital and Cordoba University Medical School, Department of PathologyCordoba, Spain. RP Gonzalez-Campora, R (reprint author), Virgen Macarena University Hospital and University of Seville Medical School, Department of Pathology, 41009 Seville, Spain. EM rcampora@us.es CR Berman J, O´Leary TJ: Gastrointestinal stromal tumor workshop. Hum Pathol 32:578-582, 2001. Dziegiel P, Salwa-Zurawska W, Zurawski J, Wojnar A, Zabel M: Prognostic significance of augmented metallothionein, MT, expression correlated with Ki-67 antigen expression in selected soft tissue sarcomas. Histol Histopathol 2: 83-89, 2005. Edmonson JH, Marks RS, Buckner JC, Mahoney MR: Contrast of response to dacarbazine, mitomycin, doxorubicin and cisplatin, DMAP, plus GM-CSF between patients with advanced malignant gastrointestinal stromal tumors and patients with other advanced leiomyosarcomas. Cancer Invest 20:605-612, 2002. Endresen L, Bakka A, Rugstad HE: Increased resistance to chlorambucil in cultured cells with a high concentration of cytoplasmic metallothionein. 43: 2918-2926, 1983. Fletcher CMD, Berman JJ, Corless C, Gorstein F, Lasota J, Longley BJ, Miettinen M, O'Leary TJ, Remotti H, Rubin BP, Shmookler B, Sobin LH, Weiss SW: Diagnosis of gastrointestinal stromal tumors: a consensus approach. Hum Pathol 33: 459-465, 2002. Gaumann A, Tews DS, Mentzel T, Petrow PK, Mayer E, Otto M, Kirkpatrick CJ, Kriegsmann J: Expression of drug resistance related proteins in sarcomas of the pulmonary artery and poorly differentiated leiomyosarcomas of other origin. Virchows Arch 442: 529-537, 2003. Hegedus T, Orfi L, Seprodi A, Varadi A, Sarkadi B, Keri G: Interaction of tyrosine kinase inhibitors with the human multidrug transporter proteins, MDR1 and MRP1. Biochim Biophys Acta 1587: 318-325, 2002. Heinrich MC, Corless CL, Demetri GD, Blanke CD, von Mehren M, Joensuu H, McGreevey LS, Chen CJ, Van den Abbeele AD, Druker BJ, Kiese B, Eisenberg B, Roberts PJ, Singer S, Fletcher CD, Silberman S, Dimitrijevic S, Fletcher JA: Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumors. J Clin Oncol 21:4342-4349, 2003. Hirota S, Isozaki K: Pathology of gastrointestinal stromal tumors. Pathol Int 56:1-9, 2006. Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Ishiguro S, Kawano K, Hanada M, Kurata A, Takeda M, Muhammad Tunio G, Matsuzawa Y, Kanakura Y, Shinomura Y, Kitamura Y: Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 279: 577-580, 1998. Joensuu H, Roberts PJ, Sarlomo-Rikala M Andersson LC, Tervahartiala P, Tuveson D, Silberman S, Capdeville R, Dimitrijevic S, Druker B, Demetri GD: Effect of the tyrosine kinase inhibitor STI571 in a patient with metastatic gastrointestinal stromal tumors. N Engl J Med 344:1052-1056, 2001. Kagi JHR: Overview of metallothionein. Methods Enzymol 205: 613-626, 1991. Kindblom LG, Remotti HE, Aldenborg F, Meis-Kindblom JM: Gastrointestinal pacemaker cell tumor, GIPACT): gastrointestinal stromal tumors show phenotypic characteristics of the interstitial cells of Cajal. Am J Pathol 152:1259-1269, 1998. Lehnert T: Gastrointestinal sarcoma, GIST, – a review of surgical management. Ann Chir Gynecol 87: 297-305, 1998. Merino V, Relevance of multidrug resistance proteins on the clinical efficacy of cancer therapy. 1: 203-212, 2004. Nilsson B, Bumming P, Meis-Kindblom JM, Oden A, Dortok A, Gustavsson B, Sablinska K, Kindblom LG: Gastrointestinal stromal tumors: the incidence, prevalence, clinical course, and prognostication in the preimatinib mesylate era – a population- based study in western Sweden. Cancer 103: 821-829, 2005. Ning ZQ, Li J, Arceci RJ: Activating mutation of c-kit at codon 816 confers drug resistance in human leukemia cells. Leuk Lymphoma. 41: 513-522, 2001. Plaat BEC, Hollema H, Willemina MM: Soft tissue leiomyosarcomas and malignant gastrointestinal stromal tumors: differences in clinical outcome and expression of multidrug resistance proteins. J Clin Oncol 18: 3211-3220, 2000. Theou N, Gil S, Devocelle A Julie C,Lavergne-Slove A, Beauchet A, Callard P, Farinotti R, Le Cesne A, Lemoine A, Faivre-Bonhomme L, Emile JF: Multidrug resistance proteins in gastrointestinal stromal tumors: site-dependent expression and initial response to imatinib. Clin Cancer Res 11: 7593-7598, 2005. Weiss SW, Goldblum JR: Enzinger and Weiss´s soft tissue tumors, 4th ed., Mosby, St. Louis, 2001. Zwelling A, Slovak ML, Doroshow JH, Hinds M, Chan D, Parker E, Mayes J, Sie KL, Meltzer PS, Trent JM: HT1080/DR4: A P-Glycoprotein-negative human fibrosarcoma cell line exhibiting resistance to topoisomerase II-reactive drugs despite the presence of a drug sensitive topoisomerase II. J Natl Cancer Inst 82: 1553-1561, 1990. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 203 EP 208 PG 6 ER PT J AU Bereczki, L Kis, Gy Bagdi, E Krenacs, L AF Bereczki, Laszlo Kis, Gyongyi Bagdi, Eniko Krenacs, Laszlo TI Optimization of PCR Amplification for B- and T-cell Clonality Analysis on Formalin-fixed and Paraffin-embedded Samples SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PCR optimization; B- and T-cell clonality; archived tissues ID PCR optimization; B- and T-cell clonality; archived tissues AB In many cases, particularly in retrospective studies, only formalin-fixed and paraffin-embedded (FFPE) tissue samples are available for molecular studies. DNA recovered from FFPE tissues generally consists of fragmented small target sequences with chemical alterations. Clonality analysis is not easy on FFPE samples, in fact, it requires even more experience than that of performed on fresh samples or is more complicated than most genomic PCR amplifications for somatic genes. In our study, we have performed a multi-parameter PCR evaluation investigating immunoglobulin heavy chain gene (IgH) and T-cell receptor gamma gene (TCRgamma) rearrangements on non-purified crude lysates of FFPE samples, in order to establish the significance of different variables on performance of PCR amplification. The results showed that a slight decrease in the concentration of primers in combination with a slight increase in MgCl2 and Taq polymerase concentrations, as well as the use diluted crude template and a standard amount of dNTPs can be the modifications of choice while adjusting IgH and TCRgamma clonality tests on poor quality DNA FFPE samples. Using our improved protocol, 74% (17/23) of the tested B-cell lymphomas and 68% (31/46) of the tested T-cell lymphomas demonstrated monoclonal PCR product, proving the applicability of our optimized method. Our experience may be of help during the optimization process in technically difficult cases as well as to determine which parameters and how should be changed to minimize false-negative and false-positive results. C1 [Bereczki, Laszlo] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular Diagnostics, Derkovits fasor 2., H-6726 Szeged, Hungary. [Kis, Gyongyi] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular Diagnostics, Derkovits fasor 2., H-6726 Szeged, Hungary. [Bagdi, Eniko] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular Diagnostics, Derkovits fasor 2., H-6726 Szeged, Hungary. [Krenacs, Laszlo] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular Diagnostics, Derkovits fasor 2., H-6726 Szeged, Hungary. RP Krenacs, L (reprint author), Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular Diagnostics, H-6726 Szeged, Hungary. EM krenacsl@bay.u-szeged.hu CR Arnold A, Cossman J, Bakhsi A, et al: Immunoglobulin-gene rearrangements as unique clonal markers in human lymphoid neoplasms. N Engl J Med 309: 1593-1599, 1983 Korsmeyer SJ: B-lymphoid neoplasms: Immunoglobulin genes as molecular determinants of clonality, lineage, differentiation, and translocation. Adv Intern Med 33: 1-15, 1988 Van Dongen JJM, Langerak AW, Bruggemann M, et al: Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombination in suspect lymphoproliferations: Report of the BIOMED-2 Concerted Action BMH4-CT98-3936. Leukemia 17:2257-2317, 2003 Trainor KJ, Brisco MJ, Story CJ, Morley AA: Monoclonality in B lymphoproliferative disorders detected at the DNA level. Blood 75:2220-2222, 1990 Lehman CM, Sarago C, Nasim S, et al: Comparison of PCR with Southern blot hybridization for the routine detection of immunoglobulin heavy chain gene rearrangements. Am J Clin Pathol 103:171-176, 1995 Hoeve MA, Krol ADG, Philippo K, et al: Limitations of clonality analysis of B cell proliferations using CDR3 polymerase chain reaction. J Clin Pathol:Mol Pathol 53:194-200, 2000 Inghirami G, Szabolcs MJ, Yee HT, et al: Detection of immunoglobulin gene rearrangement of B cell non-Hodgkin’s lymphomas and leukemias in fresh, unfixed and formalin-fixed, paraffinembedded tissue by polymerase chain reaction. Lab Invest 68: 746- 757, 1993 McCarthy KP, Sloane JP, Kabarowski JHS, et al: A simplified method of detection of clonal rearrangements of the T-cell receptorg chain gene. Diagn Mol Pathol 1:173-179, 1992 Diss TC, Pan L, Peng H, et al: Sources of DNA for detecting B cell monoclonality using PCR. J Clin Pathol 47:493-496, 1994 Coates PJ, d’Ardenne AJ, Khan G, et al: Simplified procedures for applying the polymerase chain reaction to routinely fixed paraffin wax sections. J Clin Pathol 44:115-118, 1991 Kuppers R, Zhao M, Rajewsky K, et al: Detection of clonal B cell populations in paraffin-embedded tissues by polymerase chain reaction. Am J Pathol 143:230-239, 1993 Lorenzen J, Jux G, Zhao-Hohn M, et al: Detection of T-cell clonality in paraffin-embedded tissues. Diagn Mol Pathol 3: 93-99, 1994 Solomon MJ, Varshavsky A: Formaldehyde-mediated DNA-protein crosslinking: a probe for in vivo chromatin structures. PNAS 82:6470-6474, 1985 Ben-Ezra J, Johnson DA, Rossi J, et al. Effect of fixation on the amplification of nucleic acids from paraffin-embedded material by the polymerase chain reaction. J Histochem Cytochem 39:351-354, 1991 Bielawski K, Zaczek A, Lisowska U, et al: The suitability of DNA extracted from formalin-fixed, paraffin-embedded tissues for double differential polymerase chain reaction analysis. Int J Mol Med 8:573-578, 2001 Srinivasan M, Sedmak D, Jewell S: Effect of fixatives and tissue processing on the content and integrity of nucleic acids. Am J Pathol 161:1961-1971, 2002 Metz B, Kersten GF, Hoogerhout P, et al: Identification of formaldehyde-induced modifications in proteins. Reactions with model peptides. J Biol Chem 279:6235-6243, 2004 Sepp R, Szabo I, Uda H, Sakamoto H: Rapid techniques for DNA extraction from routinely processed archival tissue for use in PCR. J Clin Pathol 47: 318-323, 1994 Sato Y, Sugie R, Tsuchiya B, et al: Comparison of the DNA extraction methods for polymerase chain reaction amplification from formalin- fixed and paraffin-embedded tissues Diagn Mol Pathol 10:265-271, 2001 Shi S-R, Datar R, Liu C, et al: DNA extraction from archival formalin- fixed, paraffin-embedded tissues: heat-induced retrieval in alkaline solution. Histochem Cell Biol 122: 211-218, 2004 Gonzalez MD, Gonzalez R, Lopez-Perez R, et al: Heteroduplex analysis of VDJ amplified segments from rearranged igh genes for clonality assessments in B-cell non-Hodgkin’s lymphoma. A comparison between different strategies. Hematologica 84:779-784, 1999 Gurbity TP, Bagdi E, Groen NA, et al: Increased sensitivity of Bcell clonality analysis in formalin-fixed and paraffin-embedded Bcell lymphoma samples using an enzyme blend with both 5’->3’ DNA polymerase and 3’->5’ exonuclease activity. Virchows Arch. , 2003 Elenitoba-Johnson KSJ, Bohling SD, Mitchell RS, et al: PCR analysis of the immunoglobulin heavy chain gene in polyclonal processes can yield pseudoclonal bands as an artifact of low B cell number. J Mol Diag 2:92-96, 2000 Saiki RK, Gelfand DH, Stoffel S, et al: Primer-directed enzymatic amplification of DNA with a thermostable DNA polymerase. Science 239 487-499, 1988 Arber DA, Brazile RM, Bagg A, Bijwaard KE: Evaluation of T cell receptor testing in lymphoid neoplasms - Results of a multicenter study of 29 extracted DNA and paraffin-embedded samples. J Mol Diag 3:133-140, 2001 Bagg A, Braziel RM, Arber DA, Bijwaard KE, Chu AY: Immunoglobulin heavy chain gene analysis in lymphomas – A multi-center study demonstrating the heterogeneity of performance of polymerase chain reactions assays. J Mol Diag 4:81-89, 2002 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 209 EP 214 PG 6 ER PT J AU Celikel, C , E Gulluoglu, B Bekiroglu, N Turhal, S AF Celikel, Cigdem , Eren Gulluoglu, Bahadir Bekiroglu, Nural Turhal, Serdar TI Relation of Neuroendocrine Cells to Transforming Growth Factor-Alpha and Epidermal Growth Factor Receptor Expression in Gastric Adenocarcinomas: Prognostic Implications SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE neuroendocrine cells; TGF-alpha; EGFR; gastric carcinoma ID neuroendocrine cells; TGF-alpha; EGFR; gastric carcinoma AB The presence of neuroendocrine (NE) cells in gastric adenocarcinoma (GCa) is well documented, however, their significance is controversial. There is no evidence in the literature concerning the possible effect of these cells on the expression of TGF-alpha and EGFR, which are believed to confer growth advantage to tumor cells. 101 partial or total gastrectomy specimens from patients operated for conventional gastric adenocarcinoma were included in the study. In each case immunohistochemistry was performed on sequential tissue sections for chromogranin A (ChrA), TGF-alpha and EGFR. Samples were graded based on the number of ChrA-positive cells (0-3). TGF-alpha and EGFR expressions were evaluated according to both the intensity (0-2) and quantification of the positively stained areas (0-3). Follow-up data was available in 54 patients. Twenty-seven patients died of disease, while 27 patients were alive with a follow-up of at least 15 months. ChrA expression was detected in 54.4% of the tumor specimens. TGF-alpha was stained positively in 42.6% and EGFR in 49.5% of the cases. NE cells in GCa was related to TGF-alpha (p<0.0001) and EGFR expression (p<0.05), and TGF-alpha/EGFR coexpression (p<0.001). Among histopathologic variables, the presence of NE cells was significantly related to grade, stage and lymph node status. Although the presence of NE cells had no effect on survival, the expression of EGFR (p<0.0001) and TGF-alpha (p=0.002) were related to survival. The results of our study suggest that the presence of NE cells may have an effect on the expression of TGF-alpha and EGFR in GCa, and the autocrine mechanism between TGF-alpha and EGFR plays an important role in the prognosis of gastric carcinoma. C1 [Celikel, Cigdem] Marmara University, School of Medicine, Department of Pathology, Tophanelioglu Cad., 81190 Istanbul, Altunizade, Turkey. [, Eren] Marmara University, School of Medicine, Department of General SurgeryIstanbul, Turkey. [Gulluoglu, Bahadir] Marmara University School of Medicine, Department of BiostatisticsIstanbul, Turkey. [Bekiroglu, Nural] Marmara University School of Medicine, Department of Medical OncologyIstanbul, Turkey. [Turhal, Serdar] Marmara University, School of Medicine, Department of General SurgeryIstanbul, Turkey. RP Celikel, C (reprint author), Marmara University, School of Medicine, Department of Pathology, 81190 Istanbul, Turkey. EM turege@superonline.com.tr CR Adachi Y, Yasuda K, Inomata M, Sato K, Shiraishi N, Kitano S: Pathology and prognosis of gastric carcinoma: well versus poorly differentiated type. Cancer 89:1418-1424, 2000. Back W, Rohr G, Bleyl U: Expression of TGF-alpha in neuroendocrine tumours of the distal colon and rectum. APMIS 111:931-939, 2003. Bonar SF, Sweeney EC: The prevalence, prognostic significance and hormonal content of endocrine cells in gastric cancer. Histopathology 10:53-63, 1986. Carpenter G: Properties of the receptor for epidermal growth factor. Cell 37:357-358, 1984. Chen WS, Lazar CS, Lund KA, Welsh JB, Chang CP, Walton GM, Der CJ, Wiley HS, Gill GN, Rosenfeld MG: Functional independence of the epidermal growth factor receptor from a domain required for ligand-induced internalization and calcium regulation. Cell 59:33-43, 1989. Dawson IMP: Diffuse endocrine and neuroendocrine cell tumors. In: Recent Advances in Histopathology., Eds: Anthony PP, Macween RNM), Churchill Livingstone, Edinburgh, 1984, pp 111-128. Derynck R: Transforming growth factor-alpha: Structure and biological activities. J Cell Biochem 32:293-304, 1986. di Sant’Agnese PA: Neuroendocrine differentiation in prostatic carcinoma: an update. Prostate Suppl 8:74-79, 1998. Eren F, Celikel CA, Gulluoglu B: Neuroendocrine differentiation in gastric adenocarcinomas; correlation with tumor stage and expression of VEGF and p53. Pathol Oncol Res 10:47-51, 2004. Gamboa-Dominguez A, Dominguez-Fonseca C, Quintanilla- Martinez L, Reyes-Gutierrez E, Green D, Angeles-Angeles A, Busch R, Hermannstadter C, Nahriq J, Becker KF: Epidermal growth factor receptor expression correlates with poor survival in gastric adenocarcinoma from Mexican patients: a multivariate analysis using a standardized immunohistochemical detection system. Mod Pathol 17:579-587, 2004. Garcia I, Vizoso F, Martin A, Sanz L, Abdel-Lah O, Raigoso P, Garcia-Muniz JL: Clinical significance of the epidermal growth factor receptor and HER2 receptor in resectable gastric cancer. Ann Surg Oncol 10:234-241, 2003. Grabowski P, Schindler I, Anagnostopoulos I, Foss HD, Riecken EO, Mansmann U, Stein H, Berger G, Buhr HJ, Scherulb H: Neuroendocrine differentiation is a relevant prognostic factor in stage III-IV colorectal cancer. Eur J Gastroenterol Hepatol 13:405-411, 2001. Hirayama D, Fujimori T, Satonaka K, Nakamura T, Kitazawa S, Horio M, Maeda S, Nagasako K: Immunohistochemical study of epidermal growth factor and transforming growth factor-b in penetrating type of early gastric cancer. Hum Pathol 23:681- 685, 1992. Hopfner M, Sutter AP, Gerst B, Zeitz M, Scherubl H: A novel approach in the treatment of neuroendocrine gastrointestinal tumours. Targeting the epidermal growth factor receptor by gefitinib, ZD1839). Br J Cancer 89:1766-1775, 2003. Issing WJ, Liebich C, Wustrow TPU, Ullrich A: Coexpression of epidermal growth factor receptor and TGF-α and survival in upper aerodigestive tract cancer. Anticancer Res 16:283-288, 1996. Ito R, Nakayama H, Yoshida K, Matsumura S, Oda N, Yasui W: Expression of Cbl linking with the epidermal growth factor receptor system is associated with tumor progression and poor prognosis of human gastric carcinoma. Virchows Arch 444:324-331, 2004. James R, Bradshaw RA: Polypeptide growth factors. Annu Rev Biochem 53:259-292, 1984. Jiang SX, Mikami T, Umezawa A, Saegusa M, Kameya T, Okayasu I: Gastric large cell neuroendocrine carcinomas: a distinct clinicopathologic entity. Am J Surg Pathol 30:945-953, 2006. Karameris A, Kanavaros P, Aninos D, Gorgoulis V, Mikou G, Rokas T, Niotis M, Kalogeropoulos N: Expression of epidermal growth factor, EGF, and epidermal growth factor receptor, EGFR, in gastric and colorectal carcinomas: an immunohistochemical study of 63 cases. Path Res Pract 189:133-137, 1993. Kim KM, Kim MJ, Cho BK, Choi SW, Rhyu MG: Genetic evidence for the multi-step progression of mixed glandular-neuroendocrine gastric carcinomas. Virchows Arch 440:85-93, 2002. Krishnamurthy S, Dayal Y: Immunohistochemical expression of transforming growth factor alpha and epidermal growth factor receptor in gastric carcinoids. Am J Surg Pathol 21:327-333, 1997. Kuba T, Watanabe H: Neoplastic argentaffin cells in gastric and intestinal carcinomas. Cancer 27:447-454, 1971. Lechago J: Gastrointestinal neuroendocrine cell proliferations. Hum Pathol 25:1114-1122, 1994. Modlin IM, Tang LH: The gastric enterochromaffin-like cell: an enigmatic cellular link. Gastroenterology 111:783-810, 1996. Murayama H, Imai T, Kikuchi M: Solid carcinomas of the stomach. Cancer 51:1673-1681, 1983. Nilsson O, Wangberg B, Kolby L, Schultz GS, Ahlman H: Expression of transforming growth factor alpha and its receptor in human neuroendocrine tumours. Int J Cancer 60:645- 651, 1995. Ooi A, Mai M, Ogino T, Ueda H, Kitamura T, Takahashi Y, Kawahara E, Nakanishi I: Endocrine differentiation of gastric adenocarcinoma: the prevalence as evaluated by immunoreactive chromogranin A and its biologic significance. Cancer 62:1096-1104, 1988. Papouchado B, Erickson LA, Rohlinger AL, Hobday TJ, Erlichman C, Ames MM, Lloyd RV: Epidermal growth factor receptor and activated epidermal growth factor receptor expression in gastrointestinal carcinoids and pancreatic endocrine carcinomas. Mod Pathol 18:1329-1335, 2005. Park DI, Yun JW, Park JH, Oh SJ, Kim HJ, Cho YK, Sohn CI, Jeon WK, Kim BI, Yoo CH, Son BH, Cho EY, Chae SW, Kim EJ, Sohn JH, Ryu SH, Sepulveda AR: HER-2/neu amplification is an independent prognostic factor in gastric cancer. Dig Dis Sci 51:1371-1379, 2006. Park JG, Choe GY, Helman LJ, Gazdar AF, Yang HK, Kim JP, Park SH, Kim YI: Chromogranin-A expression in gastric and colon cancer tissues. Int J Cancer 51:189-194, 1992. Pueztal L, Lewis CE, Lorenzen J, McGee OD: Growth factors: regulation of normal and neoplastic growth. J Pathol 169:191- 201, 1993. Qvigstad G, Sandvik AK, Brenna E, Aase S, Waldum HL: Detection of chromogranin A in human gastric adenocarcinomas using a sensitive immunohistochemical technique. Histochem J 32:551-556, 2000. Qvigstad G, Qvigstad T, Westre B, Sandvik AK, Brenna E, Waldum HL: Neuroendocrine differentiation in gastric adenocarcinomas associated with severe hypergastrinemia and/or pernicious anemia. APMIS 110:132-139, 2002. Shah T, Hochhauser D, Frow R, Quaglia A, Dhillon AP, Caplin ME: Epidermal growth factor receptor expression and activation in neuroendocrine tumours. J Neuroendocrinol 18:355- 360, 2006. Sobin LH, Wittekind CH, eds): TNM Classification of Malignant Tumors. International Union Against Cancer. 5th edition, John Wiley & Sons, New York, 1997. Tahara E: Growth factors and oncogenes in human gastrointestinal carcinomas. J Cancer Res Clin Oncol 116:121-131, 1990. Tang LH, Modlin LM, Lawton GP, Kidd M, Chinery R: The role of transforming growth factor a in the enterochromaffinlike cell tumor autonomy in an African rodent Mastomys. Gastroenterology 111:1212-1223, 1996. Tielemans Y, Chen D, Sundler F, Hakanson R, Willems G: Reversibility of the cell kinetic changes induced by omeprazole in the rat oxyntic mucosa. An autoradiographic study using tritiated thymidine. Scand J Gastroenterol 27:155-160, 1992. Waldum HL, Aase S, Kvetnol I, Brenna E, Sandvik AK, Syversen U, Johnsen G, Vatten L, Polak JM: Neuroendocrine differentiation in human gastric carcinoma. Cancer 83:435-444, 1998. Waldum HL, Brenna E, Sandvik AK: Relationship of ECL cells and gastric neoplasia. Yale J Biol Med 71:325-335, 1998. Wilson BS, Lloyd RV: Detection of chromogranin in neuroendocrine cells with a monoclonal antibody. Am J Pathol 115:458-468, 1984. Yao GY, Zhou JL, Lai MD, Chen XQ, Chen PH: Neuroendocrine markers in adenocarcinomas: an investigation of 356 cases. World J Gastroenterol 9:858-861, 2003. Yonemura Y, Takamura H, Ninomiya I, Fushida S, Tsugawa K, Kaji M, Nakai Y, Ohoyama S, Yamaguchi A, Miyazaki I: Interrelationship between transforming growth factor-a and epidermal growth factor receptor in advanced gastric cancer. Oncology 49:157-161, 1992. Yoshida K, Kyo E, Tsujino T, Sano T, Niimoto M, Tahara E: Expression of epidermal growth factor, transforming growth factor- a and their receptor genes in human gastric carcinomas; implication for autocrine growth. Jpn J Cancer Res 81:43-51, 1990. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 215 EP 226 PG 12 ER PT J AU Bovari, J Czompoly, T Olasz, K Hans-Henning, A Balogh, P AF Bovari, Judit Czompoly, Tamas Olasz, Katinka Hans-Henning, Arnold Balogh, Peter TI Complex Organizational Defects of Fibroblast Architecture in the Mouse Spleen with Nkx2.3 Homeodomain Deficiency SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE spleen; fibroblast heterogeneity; Nkx2.3 ID spleen; fibroblast heterogeneity; Nkx2.3 AB The capacity of secondary lymphoid organs to provide suitable tissue environment for mounting immune responses is dependent on their compartmentalized stromal constituents, including distinct fibroblasts. In addition to various members of the tumor necrosis factor/lymphotoxin beta family as important morphogenic regulators of peripheral lymphoid tissue development, the formation of stromal elements of spleen is also influenced by the Nkx2.3 homeodomain transcription factor in a tissue-specific fashion. Here we extend our previous work on the role of Nkx2.3-mediated regulation in the development of spleen architecture by analyzing the structure of reticular fibroblastic meshwork of spleen in inbred Nkx2.3-deficient mice. Using immunohistochemistry and dual-label immunofluorescence we found both distributional abnormalities, manifested as poor reticular compartmentalization of T-zone and circumferential reticulum, and developmental blockade, resulting in the absence of a complementary fibroblast subpopulation of white pulp. The disregulated distribution of fibroblasts was accompanied with an increased binding of immunohistochemically detectable complement factor C4 by T-cell zone-associated reticular fibroblasts, distinct from follicular dendritic cells with inherently high-level expression of bound C4. These data indicate that the impact of Nkx2.3 gene deficiency on fibroblast ontogeny within the spleen extends beyond its distributional effects, and that the formation of various white pulp fibroblast subsets is differentially affected by the presence of Nkx2.3 activity, possibly also influencing their role in various immune functions linked with complement activation and deposition. C1 [Bovari, Judit] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7643 Pecs, Hungary. [Czompoly, Tamas] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7643 Pecs, Hungary. [Olasz, Katinka] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7643 Pecs, Hungary. [Hans-Henning, Arnold] Technical University of Braunschweig, Department of Cell & Molecular Biology, Institute of Biochemistry & BiotechnologyBraunschweig, Germany. [Balogh, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7643 Pecs, Hungary. RP Balogh, P (reprint author), University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, H-7643 Pecs, Hungary. EM peter.balogh@aok.pte.hu CR Fu YX, Chaplin DD: Development and maturation of secondary lymphoid tissues. Annu Rev Immunol 17: 399-433, 1999. Mebius RE: Organogenesis of lymphoid tissues. Nat Rev Immunol 3: 292-303, 2003. Weih F, Caamano J: Regulation of secondary lymphoid organ development by the nuclear factor-kappaB signal transduction pathway. Immunol Rev 195: 91-105, 2003. Gommerman JL, Browning JL: Lymphotoxin/light, lymphoid microenvironments and autoimmune disease. Nat Rev Immunol 3: 642-655, 2003. Brendolan A, Ferretti E, Salsi V, Moses K, Quaggin S, Blasi F, Cleary ML, Selleri L: A Pbx1-dependent genetic and transcriptional network regulates spleen ontogeny. Development 132: 3113-3126, 2005. Browning JL, French LE: Visualization of lymphotoxin-beta and lymphotoxin-beta receptor expression in mouse embryos. J Immunol 168: 5079-5087, 2002. Tumanov AV, Grivennikov SI, Shakhov AN, Rybtsov SA, Koroleva EP, Takeda J, Nedospasov SA, Kuprash DV: Dissecting the role of lymphotoxin in lymphoid organs by conditional targeting. Immunol Rev 195: 106-116, 2003. Pabst O, Forster R, Lipp M, Engel H, Arnold HH: NKX2.3 is required for MAdCAM-1 expression and homing of lymphocytes in spleen and mucosa-associated lymphoid tissue. EMBO J 19: 2015-23, 2000. Wang CC, Biben C, Robb L, Nassir F, Barnett L, Davidson NO, Koentgen F, Tarlinton D, Harvey RP: Homeodomain factor Nkx2-3 controls regional expression of leukocyte homing coreceptor MAdCAM-1 in specialized endothelial cells of the viscera. Dev Biol 224: 152-167, 2000. Tarlinton D, Light A, Metcalf D, Harvey RP, Robb L: Architectural defects in the spleens of Nkx2-3-deficient mice are intrinsic and associated with defects in both B cell maturation and T cell-dependent immune responses. J Immunol 170: 4002-4010, 2003. Balogh P, Balazs M, Czompoly T, Weih DS, Arnold HH, Weih F: Distinct roles of lymphotoxin-beta signaling and the homeodomain transcription factor Nkx2.3 in the ontogeny of endothelial compartments in spleen. Cell Tissue Res 328: 473-486, 2007. Balogh P, Horvath G, Szakal AK: Immunoarchitecture of distinct reticular fibroblastic domains in the white pulp of mouse spleen. J Histochem Cytochem 52: 1287-1298, 2004. Taylor PR, Pickering MC, Kosco-Vilbois MH, Walport MJ, Botto M, Gordon S, Martinez-Pomares L: The follicular dendritic cell restricted epitope, FDC-M2, is complement C4; localization of immune complexes in mouse tissues. Eur J Immunol 32: 1888-1896, 2002. Van Vliet E, Melis M, Foidart JM, Van Ewijk W: Reticular fibroblasts in peripheral lymphoid organs identified by a monoclonal antibody. J Histochem Cytochem 34: 883-890, 1986. Katakai T, Hara T, Sugai M, Gonda H, Shimizu A: Lymph node fibroblastic reticular cells construct the stromal reticulum via contact with lymphocytes. J Exp Med 200: 783-795, 2004. Balogh P, Aydar Y, Tew JG, Szakal AK: Ontogeny of the follicular dendritic cell phenotype and function in the postnatal murine spleen. Cell Immunol 214: 45-53, 2001. Yoshida K, Tamahashi N, Matsuura N, Takahashi T, Tachibana T: Antigenic heterogeneity of the reticular meshwork in the white pulp of mouse spleen. Cell Tissue Res 266: 223-229. 1991. Skibinski G, Skibinska A, Stewart GD, James K: Enhancement of terminal B lymphocyte differentiation in vitro by fibroblast- like stromal cells from human spleen. Eur J Immunol 28: 3940-3948, 1998. Nolte MA, Belien JA, Schadee-Eestermans I, Jansen W, Unger WW, van Rooijen N, Kraal G, Mebius RE: A conduit system distributes chemokines and small blood-borne molecules through the splenic white pulp. J Exp Med 198: 505-512, 2003. Lotscher M, Recher M, Hunziker L, Klein MA: Immunologically induced, complement-dependent up-regulation of the prion protein in the mouse spleen: follicular dendritic cells versus capsule and trabeculae. J Immunol 170: 6040-6047, 2003. Wang SL, Kutche M, DiSciullo G, Schachner M, Bogen SA: Selective malformation of the splenic white pulp border in L1-deficient mice. J Immunol 165: 2465-2473, 2000. Kraal G, Schornagel K, Streeter PR, Holzmann B, Butcher EC: Expression of the mucosal vascular addressin, MAd- CAM-1, on sinus-lining cells in the spleen. Am J Pathol 147: 763-771, 1995. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 227 EP 235 PG 9 ER PT J AU Keresztes, K Szollosi, Z Simon, Zs Tarkanyi, I Nemes, Z Illes, AF Keresztes, Katalin Szollosi, Zoltan Simon, Zsofia Tarkanyi, Ilona Nemes, Zoltan Illes, Arpad TI Retrospective Analysis of the Prognostic Role of Tissue Eosinophil and Mast Cells in Hodgkin's Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hodgkin's lymphoma; mast cell; eosinophil cell ID Hodgkin's lymphoma; mast cell; eosinophil cell AB The composition of reactive cell populations, which constitute the majority of tumor load in Hodgkin’s lymphoma (HL), can influence the prognosis of the disease. Besides widely accepted and applied prognostic scores, the authors evaluate biological factors that may have a prognostic impact. Previous data indicate that the rate of eosinophils and mast cells in the reactive cell population, determined already at diagnosis, can be used for this purpose. Histological samples from 104 patients with HL with an average follow-up period of 110 (24-214) months were retrospectively analyzed. Mast cell positivity was associated with better overall survival, although this difference was only of borderline statistical significance (p=0.092). No significant difference was found in parameters like overall survival (OS, p=0.906) or event-free survival (EFS, p=0.307) of eosinophil-positive vs. -negative cases or in EFS (p=0.742) of mast cell-positive vs. -negative individuals (criterion for a positive specimen was more than 5% of appropriate cells in the reactive cell population). Looking at the effect of eosinophilia and mastocytosis together, there was no significant difference between the subgroups categorized according to the combined presence of the two cell types. It seems that tissue eosinophil and mast cell predominance have no prognostic value that could be used in clinical practice, although a tendency for correlation of mast cell positivity with overall survival could be seen. For a definitive statement, multicenter studies should be performed involving a higher number of patients suffering from HL. C1 [Keresztes, Katalin] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, H-4004 Debrecen, Hungary. [Szollosi, Zoltan] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Simon, Zsofia] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, H-4004 Debrecen, Hungary. [Tarkanyi, Ilona] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, H-4004 Debrecen, Hungary. [Nemes, Zoltan] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, H-4004 Debrecen, Hungary. RP Keresztes, K (reprint author), University of Debrecen, Medical and Health Center, 3rd Department of Medicine, H-4004 Debrecen, Hungary. EM keresztesk@gmail.com CR Axdorph U, Porwit-MacDonald A, Grimfors G, et al: Tissue eosinophilia in relation to immunopathological and clinical characteristics in Hodgkin’s disease. Leuk Lymphoma 42: 1055-1065, 2001 Costello R, O’Callaghan T, Sebahoun G: Eosinophils and antitumour response. Rev Med Interne 6: 479-484, 2005 d’Amore ES, Lee CK, Aeppli DM, et al: Lack of prognostic value of histopathologic parameters in Hodgkin’s disease, nodular sclerosis type. A study of 123 patients with limited stage disease who had undergone laparotomy and were treated with radiation therapy. Arch Pathol Lab Med 116: 856-861, 1992 Enblad G, Sundstrom C, Glimelius B: Infiltration of eosinophils in Hodgkin’s disease involved lymph nodes predicts prognosis. Hematol Oncol 11: 187-193, 1993 Fischer M, Juremalm M, Olsson N, et al: Expression of CCL5/RANTES by Hodgkin and Reed-Sternberg cells and its possible role in the recruitment of mast cells into lymphomatous tissue. Int J Cancer 107: 197-201, 2003 Glimelius I, Edstrom A, Fischer M, et al: Angiogenesis and mast cells in Hodgkin lymphoma. Leukemia 12: 2360-2362, 2005 Hanamoto H, Nakayama T, Miyazato H, et al: Expression of CCL28 by Reed-Sternberg cells defines a major subtype of classical Hodgkin’s disease with frequent infiltration of eosinophils and/or plasma cells. Am J Pathol 164: 997-1006, 2004 Hasenclever D, Diehl V: A prognostic score for advanced Hodgkin’s disease. N Engl J Med 339: 1506-1514, 1998 Hedvat CV, Jaffe ES, Qin J, et al: Macrophage-derived chemokine expression in classical Hodgkin’s lymphoma: application of tissue microarrays. Mod Pathol 12: 1270-1276, 2001 Henry-Amar M, Friedman S, Hayat M, et al: Erythrocyte sedimentation rate predicts early relapse and survival in early-stage Hodgkin’s disease. The EORTC Lymphoma Cooperative Group. Ann Intern Med 114: 361-365, 1991 Jundt F, Anagnostopoulos I, Bommert K, et al: Hodgkin/Reed- Sternberg cells induce fibroblasts to secrete eotaxin, a potent chemoattractant for T cells and eosinophils. Blood 94: 2065- 2071, 1999 Kadin M, Butmarc J, Elovic A, et al: Eosinophils are the major source of transforming growth factor-beta 1 in nodular sclerosing Hodgkin’s disease. Am J Pathol 142: 11-16, 1993 Molin D: Bystander cells and prognosis in Hodgkin lymphoma. Review based on a doctoral thesis. Ups J Med Sci 109: 179- 228, 2004 Molin D, Edstrom A, Glimelius I, et al: Mast cell infiltration correlates with poor prognosis in Hodgkin’s lymphoma. Br J Haematol 119: 122-124, 2002 Molin D, Fischer M, Xiang Z, et al: Mast cells express functional CD30 ligand and are the predominant CD30L-positive cells in Hodgkin’s disease. Br J Haematol 114: 616-623, 2001 Mosialos G, Birkenbach M, Yalamanchili R, et al: The Epstein- Barr virus transforming protein LMP1 engages signaling proteins for the tumor necrosis factor receptor family. Cell 80: 389-399, 1995 Pinto A, Aldinucci D, Gloghini A, et al: Human eosinophils express functional CD30 ligand and stimulate proliferation of a Hodgkin’s disease cell line. Blood 88: 3299-3305, 1996 Pinto A, Aldinucci D, Gloghini A, et al: The role of eosinophils in the pathobiology of Hodgkin’s disease. Ann Oncol 8(Suppl 2): 89-96, 1997 Ribatti D, Nico B, Vacca A, et al: Do mast cells help to induce angiogenesis in B-cell non-Hodgkin’s lymphomas? Br J Cancer 77: 1900-1906, 1998 Samoszuk M, Kanakubo E, Chan JK: Degranulating mast cells in fibrotic regions of human tumors and evidence that mast cell heparin interferes with the growth of tumor cells through a mechanism involving fibroblasts. BMC Cancer 5: 121, 2005 Samoszuk M, Ramzi E: IgE, Reed-Sternberg cells, and eosinophilia in Hodgkin’s disease. Leuk Lymphoma 9, 315-319, 1993 Skinnider BF, Mak TW: The role of cytokines in classical Hodgkin lymphoma. Blood 99: 4283-4297, 2002 Sorbo J, Jakobsson A, Norrby K: Mast-cell histamine is angiogenic through receptors for histamine 1 and histamine 2. Int J Exp Pathol 75: 43-50, 1994 Teruya-Feldstein J, Jaffe ES, Burd PR, et al: Differential chemokine expression in tissues involved by Hodgkin’s disease: direct correlation of eotaxin expression and tissue eosinophilia. Blood 93: 2463-2470, 1999 Toth J, Dworak O, Sugar J: Eosinophil predominance in Hodgkin’s disease. Z Krebsforsch Klin Onkol Cancer Res Clin Oncol 89: 107-111, 1977 von Wasielewski R, Seth S, Franklin J, et al: Tissue eosinophilia correlates strongly with poor prognosis in nodular sclerosing Hodgkin’s disease, allowing for known prognostic factors. Blood 95: 1207-1213, 2000 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 237 EP 242 PG 6 ER PT J AU Maasz, A Kisfali, P Horvatovich, K Mohas, M Marko, L Csongei, V Farago, B Jaromi, L Magyari, L Safrany, E Sipeky, Cs Wittmann, I Melegh, B AF Maasz, Anita Kisfali, Peter Horvatovich, Katalin Mohas, Marton Marko, Lajos Csongei, Veronika Farago, Bernadett Jaromi, Luca Magyari, Lili Safrany, Eniko Sipeky, Csilla Wittmann, Istvan Melegh, Bela TI Apolipoprotein A5 T-1131C Variant Confers Risk for Metabolic Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE metabolic syndrome; glucose intolerance; ApoA5; T-1131C ID metabolic syndrome; glucose intolerance; ApoA5; T-1131C AB The -1131C is a naturally occurring variant of the apolipoprotein A5 (ApoA5) gene, which has been shown to associate with increased triglyceride levels. This variant has also been shown to confer risk for development of ischemic heart disease and stroke. The gene is in linkage disequilibrium with factors known to correlate with impaired glucose homeostasis. These observations prompted us to study the prevalence of the ApoA5 -1131C allele in patients with metabolic syndrome. A total of 201 metabolic syndrome patients and 210 controls were studied. In both groups the triglyceride levels of patients with -1131C allele were significantly increased compared to the subjects with -1131T allele (3.22+/-0.43 mmol/l vs. 2.24+/-0.12 mmol/l, p<0.01 in the metabolic syndrome patients; 2.10+/-0.19 mmol/l vs. 1.22+/-0.05 mmol/l, p<0.01 in the controls). In metabolic syndrome patients the prevalence of the ApoA5 -1131C variant was increased compared to the healthy controls (11% vs. 6.20%). Multiplex regression analysis model adjusted for age, gender, serum total cholesterol levels, acute myocardial infarction and stroke events revealed that the examined ApoA5 variant confers risk for the development of metabolic syndrome: the odds ratio at 95% confidence interval was 3.622 (1.200-10.936), p=0.02. Our findings strongly suggest that this variant is a risk factor for the development of hypertriglyceridemia and metabolic syndrome. C1 [Maasz, Anita] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Kisfali, Peter] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Horvatovich, Katalin] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Mohas, Marton] University of Pecs, 2nd Department of Medicine and Nephrological CenterPecs, Hungary. [Marko, Lajos] University of Pecs, 2nd Department of Medicine and Nephrological CenterPecs, Hungary. [Csongei, Veronika] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Farago, Bernadett] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Jaromi, Luca] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Magyari, Lili] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Safrany, Eniko] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Sipeky, Csilla] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Wittmann, Istvan] University of Pecs, 2nd Department of Medicine and Nephrological CenterPecs, Hungary. [Melegh, Bela] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. RP Melegh, B (reprint author), University of Pecs, Department of Medical Genetics and Child Development, H-7624 Pecs, Hungary. EM bela.melegh@aok.pte.hu CR Executive Summary of the Third Report of the National Cholesterol Education Program, NCEP, Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol In Adults, Adult Treatment Panel III): JAMA 285:2486-2497, 2001 Alberti KG, Zimmet PZ: Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med 15:539-553, 1998 Aouizerat BE, Kulkarni M, Heilbron D, et al: Genetic analysis of a polymorphism in the human apoA-V gene: effect on plasma lipids. J Lipid Res 44:1167-1173, 2003 Cameron AJ, Shaw JE, Zimmet PZ: The metabolic syndrome: prevalence in worldwide populations. Endocrinol Metab Clin North Am 33:351-375, 2004 Cavanaugh J: NOD2: ethnic and geographic differences. World J Gastroenterol 12:3673-3677, 2006 Connelly PW, Petrasovits A, Stachenko S, et al: Prevalence of high plasma triglyceride combined with low HDL-C levels and its association with smoking, hypertension, obesity, diabetes, sedentariness and LDL-C levels in the Canadian population. Canadian Heart Health Surveys Research Group. Can J Cardiol 15:428-433, 1999 Eckel RH, Grundy SM, Zimmet PZ: The metabolic syndrome. Lancet 365:1415-1428, 2005 Endo K, Yanagi H, Araki J, et al: Association found between the promoter region polymorphism in the apolipoprotein A-V gene and the serum triglyceride level in Japanese schoolchildren. Hum Genet 111:570-572, 2002 Evans D, Buchwald A, Beil FU: The single nucleotide polymorphism -1131T>C in the apolipoprotein A5, APOA5, gene is associated with elevated triglycerides in patients with hyperlipidemia. J Mol Med 81:645-654, 2003 Genoux A, Dehondt H, Helleboid-Chapman A, et al: Transcriptional regulation of apolipoprotein A5 gene expression by the nuclear receptor RORalpha. Arterioscler Thromb Vasc Biol 25:1186-1192, 2005 Groenendijk M, Cantor RM, De Bruin TW, et al: The apoAICIII- AIV gene cluster. Atherosclerosis 157:1-11, 2001 Havasi V, Szolnoki Z, Talian G, et al: Apolipoprotein A5 gene promoter region T-1131C polymorphism associates with elevated circulating triglyceride levels and confers susceptibility for development of ischemic stroke. J Mol Neurosci 29:177- 183, 2006 Jakel H, Nowak M, Moitrot E, et al: The liver X receptor ligand T0901317 down-regulates APOA5 gene expression through activation of SREBP-1c. J Biol Chem 279:45462- 45469, 2004 Lai CQ, Tai ES, Tan CE, et al: The APOA5 locus is a strong determinant of plasma triglyceride concentrations across ethnic groups in Singapore. J Lipid Res 44:2365-2373, 2003 Martin S, Nicaud V, Humphries SE, et al: Contribution of APOA5 gene variants to plasma triglyceride determination and to the response to both fat and glucose tolerance challenges. Biochim Biophys Acta 1637:217-225, 2003 Merkel M, Heeren J: Give me A5 for lipoprotein hydrolysis! J Clin Invest 115:2694-2696, 2005 Merkel M, Loeffler B, Kluger M, et al: Apolipoprotein AV accelerates plasma hydrolysis of triglyceride-rich lipoproteins by interaction with proteoglycan-bound lipoprotein lipase. J Biol Chem 280:21553-21560, 2005 Nadasi E: Comparison of mtDNA haplogroups in Hungarians with four other European populations: a small incidence of descents with Asian origin. Acta Biologica Hungarica 58:245- 256, 2007 Olivier M, Wang X, Cole R, et al: Haplotype analysis of the apolipoprotein gene cluster on human chromosome 11. Genomics 83:912-923, 2004 Pennacchio LA, Olivier M, Hubacek JA, et al: An apolipoprotein influencing triglycerides in humans and mice revealed by comparative sequencing. Science 294:169-173, 2001 Pennacchio LA, Olivier M, Hubacek JA, et al: Two independent apolipoprotein A5 haplotypes influence human plasma triglyceride levels. Hum Mol Genet 11:3031-3038, 2002 Pennacchio LA, Rubin EM: Apolipoprotein A5, a newly identified gene that affects plasma triglyceride levels in humans and mice. Arterioscler Thromb Vasc Biol 23:529-534, 2003 Permutt MA, Wasson J, Cox N: Genetic epidemiology of diabetes. J Clin Invest 115:1431-1439, 2005 Prieur X, Coste H, Rodriguez JC: The human apolipoprotein AV gene is regulated by peroxisome proliferator-activated receptoralpha and contains a novel farnesoid X-activated receptor response element. J Biol Chem 278:25468-25480, 2003 Reaven G: The metabolic syndrome or the insulin resistance syndrome? Different names, different concepts, and different goals. Endocrinol Metab Clin North Am 33:283-303, 2004 Reaven P: Metabolic syndrome. J Insur Med 36:132-142, 2004 Salas J, Jansen S, Lopez-Miranda J, et al: The SstI polymorphism of the apolipoprotein C-III gene determines the insulin response to an oral-glucose-tolerance test after consumption of a diet rich in saturated fats. Am J Clin Nutr 68:396-401, 1998 Semino O, Passarino G, Quintana-Murci L, et al: MtDNA and Y chromosome polymorphisms in Hungary: inferences from the palaeolithic, neolithic and Uralic influences on the modern Hungarian gene pool. Eur J Hum Genet 8:339-346, 2000 Szalai C, Keszei M, Duba J, et al: Polymorphism in the promoter region of the apolipoprotein A5 gene is associated with an increased susceptibility for coronary artery disease. Atherosclerosis 173:109-114, 2004 Talmud PJ, Hawe E, Martin S, et al: Relative contribution of variation within the APOC3/A4/A5 gene cluster in determining plasma triglycerides. Hum Mol Genet 11:3039-3046, 2002 Waterworth DM, Hubacek JA, Pitha J, et al: Plasma levels of remnant particles are determined in part by variation in the APOC3 gene insulin response element and the APOCI-APOE cluster. J Lipid Res 41:1103-1109, 2000 Wright WT, Young IS, Nicholls DP, et al: SNPs at the APOA5 gene account for the strong association with hypertriglyceridaemia at the APOA5/A4/C3/A1 locus on chromosome 11q23 in the Northern Irish population. Atherosclerosis 185:353-360, 2006 Yamada Y, Kato K, Hibino T, et al: Prediction of genetic risk for metabolic syndrome. Atherosclerosis 191:298-304, 2006 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 243 EP 247 PG 5 ER PT J AU Bardi, E Bobok, I V. Olah, A Kappelmayer, J Kiss, Cs AF Bardi, Edit Bobok, Ildiko V. Olah, Anna Kappelmayer, Janos Kiss, Csongor TI Anthracycline Antibiotics Induce Acute Renal Tubular Toxicity in Children with Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE nephrotoxicity; anthracycline therapy; dexrazoxane; NAGi; microalbuminuria ID nephrotoxicity; anthracycline therapy; dexrazoxane; NAGi; microalbuminuria AB Experimental evidence suggests that anthracyclines, widely used in cancer chemotherapy, may impair kidney function. We assessed kidney function by serum creatinine, urinary N-acetyl-beta-D-glucosaminidase activity indices (NAGi) and microalbuminuria (MA) in 160 serum and urine samples obtained from 66 children with cancer. The effect of dexrazoxane was analyzed in 6 children on dexrazoxane supportive therapy in conjunction with daunorubicin (DNR) treatment, as compared with 6 children not receiving this agent. NAGi was significantly (p<0.05) elevated after treatment by DNR, doxorubicin, epirubicin (EPI) and idarubicin (IDA). MA proved to be a less sensitive indicator of kidney damage than NAGi. DNR resulted in a progressive deterioration of proximal tubular function as determined by linear regression analysis. The mean NAGi in the dexrazoxanetreated group was significantly (p<0.005) lower than in children not receiving dexrazoxane prior to DNR treatment. In conclusion, our study demonstrated that DNR, EPI and IDA induced an acute renal tubular damage similar to known tubulotoxic agents as cisplatin, carboplatin, cyclophosphamide and ifosfamide. The damage was clinically mild and only a minor proportion of patients can be expected to develop long-lasting tubulopathy with negative impact on the quality of life. C1 [Bardi, Edit] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98. Nagyerdei korut, H-4012 Debrecen, Hungary. [Bobok, Ildiko] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98. Nagyerdei korut, H-4012 Debrecen, Hungary. [V. Olah, Anna] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Kappelmayer, Janos] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Kiss, Csongor] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98. Nagyerdei korut, H-4012 Debrecen, Hungary. RP Bardi, E (reprint author), Medical and Health Science Center, University of Debrecen, Department of Pediatrics, H-4012 Debrecen, Hungary. EM editbardi@hotmail.com CR Bardi E, Bobok I, Olah VA, et al: Cystatin C is a suitable marker of glomerular function in children with cancer. Pediatr Nephrol 10:1145-1147, 2004 Deprez-DeCampeneere D, Jaenke R, Trouet A: Comparative cardiac and renal toxicity of daunorubicin in the rat and rabbit. Cancer Treat Rep 66: 395-397, 1982 Kobayashi H: Cisplatin and ovarian carcinoma – early detection of cisplatin-induced nephrotoxicity. Nippon Sanka Fujinka Gakkai Zasshi 37:888-896, 1985 Zoubek A, Holzinger B, Mann G, et al: High-dose cyclophosphamide, adriamycin, and vincristine, HD-CAV, in children with recurrent solid tumor. Pediatr Hematol Oncol 11:613- 623,1994 Kopecna L: Late effects of anticancer therapy on kidney function in children with acute lymphoblastic leukemia. Bratisl Lek Listy 102:357-360, 2001 Kakihara T, Imai C, Uchiyama M, et al: Impaired tubular excretory function as a late side effect of chemotherapy in children. J Pediatr Hematol Oncol 25:209-214, 2003 Bardi E, Olah VA, Bartyik K, et al: Late effects on renal glomerular and tubular function in childhood cancer survivors. Pediatr Blood Cancer 43:668-673, 2004 Schrappe M, Reiter A, Zimmermann M, et al: Long-term results of four consecutive trials in childhood ALL performed by the ALL-BFM study group from 1981 to 1995, Berlin-Frankfurt- Munster. Leukemia 14: 2205-2222, 2000 Creutzig U, Ritter J, Zimmermann M, et al: Improved treatment results in high-risk pediatric acute myeloid leukemia patients after intensification with high-dose cytarabine and mitoxanthrone: results of Study Acute Myeloid Leukemia-Berlin- Frankfurt-Munster 93. J Clin Oncol 19: 2705-2713, 2001 Reiter A, Schrappe M, Parwaresch R, et al: Non-Hodgkin’s lymphomas of childhood and adolescence: results of a treatment stratified for biologic subtypes and stage – a report of the Berlin-Frankfurt-Munster Group. J Clin Oncol 13:359-372, 1995 Schellong G, Potter R, Bramswig J, et al: High cure rates and reduced long-term toxicity in pediatric Hodgkin’s disease: the German-Austrian multicenter trial DAL-HD-90. The German- Austrian Pediatric Hodgkin’s Disease Study Group. J Clin Oncol 17:3736-3744, 1999 Marx M, Langer T, Graf N, et al: Multicentre analysis of anthracycline-induced cardiotoxicity in children following treatment according to the nephroblastoma studies SIOP No. 9/ GPOH and SIOP 93-01/ GPOH. Med Pediatr Oncol 39:18-24, 2002 Ozaki T, Flege S, Kevric M, et al: Osteosarcoma of the pelvis: experience of the Cooperative Osteosarcoma Study Group. J Clin Oncol 21:334-341, 2003 Koscielniak E, Jurgens H, Winkler K, et al: Treatment of soft tissue sarcoma in childhood and adolescence. A report of the German Cooperative Soft Tissue Sarcoma Study. Cancer 70:2557-2567, 1992 Ninane J, Perilongo G, Stalens JP, et al: Effectiveness and toxicity of cisplatin and doxorubicin, PLADO, in childhood hepatoblastoma and hepatocellular carcinoma: a SIOP pilot study. Med Pediatr Oncol 19:199-203, 1991 Jaffe M: On the precipitation that picric acid produces in normal urine, and on a new reaction of creatinine [in German]. Zietschrift Physiol Chem 10: 391-400, 1986 Olah VA, Csathy L, Pocsi I, et al: Reference ranges for urinary NAG indices in healthy children determined with three colorimetric methods. Ann Clin Biochem 31:87-88, 1994 Hensley ML, Schuchter LM, Lindley C, et al: American Society of Clinical Oncology Clinical practice guidelines for the use of chemotherapy and radiotherapy protectants. J Clin Oncol 17:3333-3355, 1999 Herman EH, Zhang J, Chadwick DP, Ferrans VJ: Comparison of the protective effects of amifostine and dexrazoxane against the toxicity of doxorubicin in spontaneously hypertensive rats. Cancer Chemother Pharmacol 45:329-334, 2000 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 249 EP 253 PG 5 ER PT J AU Schiffer, Ch Schiesser, M Lehr, J Tariverdian, G Glaeser, D Heinz, G Mikuz, G Sergi, C AF Schiffer, Christiane Schiesser, Monika Lehr, Jutta Tariverdian, Gholamali Glaeser, Dieter Heinz, Gabriel Mikuz, Gregor Sergi, Consolato TI Unique Occurrence of Brachmann-de Lange Syndrome in a Fetus whose Mother Presented with a Diffuse Large B-Cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports AB Brachmann-De Lange Syndrome (BDLS, MIM 122470) is a rare multiple congenital anomaly/mental retardation syndrome characterized by a variable phenotype including intrauterine fetal growth retardation, limb reduction and distinctive facial and skull features (low frontal hairline, synophrys, anteverted nostrils, long philtrum, downturned corners of the mouth, micro- and retrognathia, low-set ears and micro-/brachycephaly), as well as a significant psychological developmental delay. A proposed classification system for BDLS include a classic type with characteristic facial and skull changes, a mild type where similar changes may develop with time or may be partially expressed, and a third type including phenocopies, where phenotypic changes are casually related to chromosomal aneuploidies or teratogenic exposures. We report on a 22-week gestation fetus with BDLS, showing intrauterine fetal growth retardation, brachycephaly, micro-/retrognathia and monolateral single bone of the forearm, in a woman harboring diffuse large B-cell lymphoma. Meticulous family history was negative for malformations, syndromes, congenital anomalies or psychiatric disorders. There are very few reports of BDLS at early gestation, but to the best of our knowledge, this is the first case occurring simultaneously with a hematological neoplastic disease of the mother. C1 [Schiffer, Christiane] Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental HealthMannheim, Germany. [Schiesser, Monika] Medical University of Heidelberg, Department of Obstetrics and GynecologyHeidelberg, Germany. [Lehr, Jutta] Medical University of Heidelberg, Department of Obstetrics and GynecologyHeidelberg, Germany. [Tariverdian, Gholamali] Medical University of Heidelberg, Department of Clinical GeneticsHeidelberg, Germany. [Glaeser, Dieter] Center of Human Genetics, Gregor Mendel LaboratoriesNeu-Ulm, Germany. [Heinz, Gabriel] Center of Medical GeneticsOsnabrueck, Germany. [Mikuz, Gregor] Medical University of Innsbruck, Department of Pathology, Muellerstrasse 44, A-6020 Innsbruck, Austria. [Sergi, Consolato] Medical University of Innsbruck, Department of Pathology, Muellerstrasse 44, A-6020 Innsbruck, Austria. RP Sergi, C (reprint author), Medical University of Innsbruck, Department of Pathology, A-6020 Innsbruck, Austria. EM consolato.sergi@i-med.ac.at CR Aitken DA, Ireland M, Berry E, Crossley JA, Macri JN, Burn J, Connor JM: Second-trimester pregnancy associated plasma protein-A levels are reduced in Cornelia de Lange syndrome pregnancies. Prenat Diagn 19: 706-710, 1999 Ayral D, Raudrant D, Charleux JP, Noel B: Duplication of the long arm of chromosome 3, dup 3q, in a newborn infant whose father is carrier of pericentric inversion of chromosome 9. Pediatrie 39: 681-690, 1984 De Die-Smulders C, Theunissen P, Schrander-Stumpel C, Frijns JP: On the variable expression of the Brachmann-de Lange syndrome. Clin Genet 41: 42-45, 1992 Ireland M, Donnai D, Burn J: Brachmann-de Lange syndrome. Delineation of the clinical phenotype. Am J Med Genet 47: 959-964, 1993 Jackson L, Kline AD, Barr MA, Koch S: De Lange syndrome: a clinical review of 310 individuals. Am J Med Genet 47: 940- 946, 1993 Kliewer MA, Kahler SG, Hertzberg BS, Bowie JD: Fetal biometry in the Brachmann-de Lange syndrome. Am J Med Genet 47: 1035-1041, 1993 Krantz ID, McCallum J, DeScipio C, Kaur M, Gillis LA, Yaeger D, Jukofsky L, Wasserman N, Bottani A, Morris CA, Nowaczyk MJ, Toriello H, Bamshad MJ, Carey JC, Rappaport E, Kawauchi S, Lander AD, Calof AL, Li HH, Devoto M, Jackson LG: Cornelia de Lange syndrome is caused by mutations in NIPBL, the human homolog of Drosophila melanogaster Nipped-B. Nat Genet 36: 631-635, 2004 Lakshminarayana P, Nallasivam P: Cornelia de Lange syndrome with ring chromosome 3. J Med Genet 27: 405-406, 1990 Lee W, McNie B, Chaiworapongsa T, Conoscenti G, Kalache K, Vettraino IM, Romero R, Comstock C: Three-dimensional ultrasonographic presentation of micrognathia. J Ultrasound Med 21: 775-781, 2002 Nowaczyk MJ, Mohide P: Brachmann-de Lange Syndrome. McMaster University Prenatal Diagnosis Rounds. Front Fet Heath 2001; 3, www.sickkids.on.ca) Pajkrt E, Weisz B, Firth HV, Chitty LS: Fetal cardiac anomalies and genetic syndromes. Prenat Diagn 24: 1104-1115, 2004 Pilu G, Nicolaides KH: Diagnosis of fetal abnormalities. The 18-23-week scan. Parthenon Publishing, New York, NY, 1999 Ranzini AC, Day-Salvatore D, Farren-Chavez D, McLean DA, Greco R: Prenatal diagnosis of de Lange syndrome. J Ultrasound Med 16: 755-758, 1997 Sekimoto H, Osada H, Kimura H, Kamiyama M, Arai K, Sekiya S: Prenatal findings in Brachmann-de Lange syndrome. Arch Gynecol Obstet 263: 182-184, 2000 Sugita K, Izumi T, Yamaguchi K, Fukuyama Y, Sato A, Kajita A: Cornelia de Lange syndrome associated with a suprasellar germinoma. Brain Dev 8: 541-546, 1986 Tonkin ET, Wang TJ, Lisgo S, Bamshad MJ, Strachan T: NIPBL, encoding a homolog of fungal Scc2-type sister chromatid cohesion proteins and fly Nipped-B, is mutated in Cornelia de Lange syndrome. Nat Genet 36: 636-641, 2004 Urban M, Hartung J: Ultrasonographic and clinical appearance of a 22-week-old fetus with Brachmann-de Lange syndrome. Am J Med Genet 102: 73-75, 2001 Van Allen MI, Filippi G, Siegel-Bartelt J, Yong SL, McGillivray B, Zuker RM, Smith CR, Magee Jf, Ritchie S, Toi A, Reynolds JF: Clinical variability within Brachmann-de Lange syndrome: a proposed classification system. Am J Med Genet 47: 947- 958, 1993 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 255 EP 259 PG 5 ER PT J AU Szomor, Al Saati, T Delsol, G Kereskai, L Szijarto, Zs Losonczy, H AF Szomor, Arpad Al Saati, Talal Delsol, Georges Kereskai, Laszlo Szijarto, Zsuzsanna Losonczy, Hajna TI Primary Bone Marrow T-cell Anaplastic Large Cell Lymphoma with Triple M Gradient SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE anaplastic large cell lymphoma; T-cell receptor gene rearrangement; immunoglobulin heavy chain gene rearrangement; M gradient; bone marrow ID anaplastic large cell lymphoma; T-cell receptor gene rearrangement; immunoglobulin heavy chain gene rearrangement; M gradient; bone marrow AB We present a case of a 60-year-old male patient with primary bone marrow anaplastic large cell lymphoma. He was admitted to the hospital with the symptoms of anemia and fever. There was no evidence of lymphadenopathy or splenomegaly. Immunoelectrophoresis showed the presence of a triple M gradient (double IgM and an IgG), with the IgG and one of the IgM paraproteins functioning as a cryoglobulin. The patient had no hepatitis C virus infection. Bone marrow biopsy showed massive CD30-positive, ALK-negative large lymphoid cell infiltration of T-cell origin with anaplastic morphology. PCR analysis of lymphoid cells separated from the bone marrow demonstrated the presence of a B/T hybrid genotype disorder with no evidence of the t(2;5), nor t(1;2) translocations. The patient entered a period of remission following CHOP chemotherapy. The patient subsequently died of sepsis as a consequence of serious humoral immunodeficiency. C1 [Szomor, Arpad] University of Pecs, I. Department of Internal Medicine, Ifjusag u. 13., H-7624 Pecs, Hungary. [Al Saati, Talal] Central Hospital University Purpan, Laboratory of Anatomic PathologyToulouse, France. [Delsol, Georges] Central Hospital University Purpan, Laboratory of Anatomic PathologyToulouse, France. [Kereskai, Laszlo] University of Pecs, Department of PathologyPecs, Hungary. [Szijarto, Zsuzsanna] University of Pecs, Department of OphthalmologyPecs, Hungary. [Losonczy, Hajna] University of Pecs, I. Department of Internal Medicine, Ifjusag u. 13., H-7624 Pecs, Hungary. RP Szomor, (reprint author), University of Pecs, I. Department of Internal Medicine, H-7624 Pecs, Hungary. EM aszomor@clinics.pote.hu CR Falini B, Pileri S, Zinzani PL, Carbone A, Zagonel V, Wolf- Peters CD, Verhoef G, Manestrina F, Tedeschini G, Paulli M, Lazzarino M, Giardini R, Aiello A, Foss HD, Araujo I, Fizzotti M, Pelicci PG, Flenghi L, Martelli MF, Santucci A: ALK+ lymphoma: clinico-pathological findings and outcome. Blood 93: 2697-2706, 1999 Garcia MJ, Martinez-Delgado B, Granizo JJ, Benitez J, Rivas C: IgH, TCR-gamma, and TCR-beta gene rearrangement in 80 B- and T-cell non-Hodgkin’s lymphomas: study of the associations between proliferation and so-called „aberrant” patterns. Diagn Mol Pathol 10: 69-77, 2001 Greer JP, Kinney MC, Collins RD, Salhany KE, Wolff SN, Hainsworth JD, Flexner JM, Stein RS: Clinical features of 31 patients with Ki-1 anaplastic large-cell lymphoma. J Clin Oncol 9: 539-547, 1991 Lamant L, Meggetto F, Al Saati T, Brugieres L, Paillerets BB, Dastugue N, Bernheim A, Rubie H, Terrier-Lacombe J, Robert A, Brousset P, Rigal F, Schlaifer D, Shiota M, Mori S, Delsol G: High incidence of the t(2;5)(p23;q35, translocation in anaplastic large cell lymphoma and its lack of detection in Hodgkin’s disease. Comparison of cytogenetic analysis, reverse transcriptase- polymerase chain reaction, and P-80 immunostaining. Blood 87: 284-291, 1996 Longo G, Federico M, Pieresca C, Avanzini P, Iannito E, Di Prisco AU, Baldini L, Brugiatelli M, Clo V, Bevini M, Silingardi V: Anaplastic large cell lymphoma, CD30+/Ki-1+). Analysis of 35 cases followed at GISL centres. Eur J Cancer 31A: 1763-1767, 1995 Shulman LN, Frisard N, Antin JH, Wheeler C, Pinkus G, Magauran N, Mauch P, Nobles E, Mashal R, Canellos G, Tung N, Kadin M: Primary Ki-1 anaplastic large-cell lymphoma in adults: clinical characteristics and therapeutic outcome. J Clin Oncol 11: 937-942, 1993 Stein H, Foss HD, Durkop H, Marafioti T, Delsol G, Pulford K, Pileri S, Falini B: CD30+ anaplastic large cell lymphoma: a review of its histopathologic, genetic and clinical features. Blood 96: 3681-3695, 2000 Szomor A, Molnar L, Ivanyi J, Radvanyi G, Nagy Z, Karadi A, Gergely L, Banyai A, Demeter J, Aryan H, Gasztonyi Z, Kiss A, Kollar B, Egyed M, Losonczy H, Kelenyi G, Pajor L: Extranodal involvement in primary systemic anaplastic large cell lymphoma, ALCL, in adults. Ann Hematol 80: B144-145, 2001 Theriault C, Galoin S, Valmary S, Selves J, Lamant L, Roda D, Rigal-Huguet F, Brousset P, Delsol G, Al Saati T: PCR analysis of immunoglobulin heavy chain, IgH, and TcR-gamma chain rearrangements in the diagnosis of lymphoproliferative disorders of a study of 525 cases. Mod Pathol 13: 1269-1279, 2000 Tilly H, Gaulard P, Lepage E, Dumontet C, Diebold J, Plantier I, Berger F, Symann M, Pertella T, Lederlin P, Briere J: Primary anaplastic large-cell lymphoma in adults: clinical presentation, immunophenotype, and outcome. Blood 90: 3727-3734, 1997 Zinzani PL, Bendandi M, Martelli M, Falini B, Sabattini E, Amadori S, Gherlinzoni F, Martelli MF, Mandelli F, Tura S, Pileri SA: Anaplastic large-cell lymphoma: prognostic evaluation of 90 adult patients. J Clin Oncol 14: 955-962, 1996 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2007 VL 13 IS 3 BP 260 EP 262 PG 3 ER PT J AU Tornoczky, T Semjen, D Shimada, H Ambros, MI AF Tornoczky, Tamas Semjen, David Shimada, Hiroyuki Ambros, M Inge TI Pathology of Peripheral Neuroblastic Tumors: Significance of Prominent Nucleoli in Undifferentiated/Poorly Differentiated Neuroblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE LCN; LNN; neuroblastoma; nucleoli; INPC; MYCN; amplification; ganglion cell; maturation; Schwann cell ID LCN; LNN; neuroblastoma; nucleoli; INPC; MYCN; amplification; ganglion cell; maturation; Schwann cell AB The presence of large cells having simultaneously increased cytoplasmic and nuclear volume accompanied by prominent nucleoli; i.e., differentiating neuroblasts and ganglion cells, is well documented in peripheral neuroblastic tumors (pNTs), and considered as one of the signs of tumor maturation and an indication of a better prognosis of the patients. On the other hand, in 2004 it was reported that large-cell neuroblastoma composed of neuroblastic cells with only nuclear enlargement without recognizable cytoplasmic maturation behaved poorly clinically. Here we are proposing a new pNT subtype in the neuroblastoma category, in addition to the undifferentiated, poorly differentiated and differentiating subtypes: that is large nucleolar neuroblastoma (LNN) characterized by large prominent nucleoli and no or very little amount of discernible cytoplasm. LNN, whose neuroblastic cells are often large in size due to nuclear enlargement, includes those tumors previously categorized into the large-cell neuroblastoma group. LNN tumors, regardless of the size of nuclei, seem to behave aggressively with a very poor prognosis of the patients. It is speculated that nucleolar enlargement without cytoplasmic maturation in LNN tumor cells can be a sign of MYCN amplification. C1 [Tornoczky, Tamas] University of Pecs, Department of Pathology, Szigeti ut 12., H-7643 Pecs, Hungary. [Semjen, David] University of Pecs, Department of Pathology, Szigeti ut 12., H-7643 Pecs, Hungary. [Shimada, Hiroyuki] University of Southern California, Keck Medical Center, Department of PathologyLos Angeles, CA, USA. [Ambros, M Inge] St. Anna Kinderkrebsforschung, Children’s Cancer Research InstituteVienna, Austria. RP Tornoczky, T (reprint author), University of Pecs, Department of Pathology, H-7643 Pecs, Hungary. EM ttamas64@hotmail.com CR Shimada H, Ambros IM, Dehner LP, Hata J, Joshi VV, Roald B: Terminology and morphologic criteria of neuroblastic tumors. Recommendations by the International Neuroblastoma Pathology Committee. Cancer 86: 349-363, 1999 Peuchmaur M, d’Amore ES, Joshi VV, Roald B, Dehner LP, Gerbing RB, Stram DO, Lukens JN, Matthay KK. Shimada H: Revision of the International Neuroblastoma Pathology Classification: confirmation of favorable and unfavorable prognostic subsets in ganglioneuroblastoma, nodular. Cancer 98:2274-2281, 2003 Shimada H, Ambros IM, Dehner LP, Hata J, Joshi VV, Roald B, Stram DO, Gerbing RB, Lukens JN, Matthay KK, Castleberry RP: International Neuroblastoma Pathology Classification, the Shimada System). Cancer 86: 364-372, 1999 Shimada H, Umehara S, Monobe Y, Hachitanda Y, Nakagawa A, Goto S, Gerbing RB, Stram DO, Lukens JN, Matthay KK: International neuroblastoma pathology classification for prognostic evaluation of patients with peripheral neuroblastic tumors. A report from the Children’s Cancer Group. Cancer 92:2451- 2461, 2001 Shimada H, Chatten J, Newton WA Jr., Sachs N, Hamoudi AB, Chiba T, Marsden HB, Misugi K: Histopathologic prognostic factors in neuroblastic tumors: definition of subtypes of ganglioneuroblastoma and an age-linked classification of neuroblastomas. J Natl Cancer Inst 73:405-416, 1984 Brodeur GM, Seeger RC, Schwab M, Varmus HE, Bishop JM: Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science 224:1121- 1124, 1984 Seeger RC, Brodeur GM, Sather H, Dalton A, Siegel SE, Wong KY, Hammond D: Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. N Engl J Med 313:1111-1116, 1985 Goto S, Umehara S, Gerbing RB, Stram DO, Brodeur GM, Seeger RC, Lukens JN, Matthay KK, Shimada H: Histopathology, International Neuroblastoma Pathology Classification, and MYCN status in patients with peripheral neuroblastic tumors. A report from the Children’s Cancer Group. Cancer 92: 2699-2708, 2001 Tornoczky T, Kalman E, Kajtar P, Nyari T, Pearson A.D.J, Tweddle D, Board J, Shimada H: Large cell neuroblastoma: a distinct phenotype of neuroblastoma with aggressive clinical behavior. Cancer 100:390-397, 2004 Kobayashi C, Monforte-Munoz HL, Gerbing RB, Stram DO, Matthay KK, Lukens JN, Seeger RC, Shimada H: Enlarged and prominent nucleoli may be indicative of MYCN amplification. Cancer 103:174-180, 2005 Ambros IM, Hata J, Joshi VV, Roald B, Dehner LP, Tuchler H, Potschger U, Shimada H: Morphologic features of neuroblastoma, Schwannian stroma-poor tumors, in clinically favorable and unfavorable groups. Cancer 94:1574-1583, 2002 Boon K, Caron H, Asperen R, Valentijn L, Hermus MC, Sluis P, Roobeek I, Weis I, Voute PA, Schwab M, Versteeg R: N-myc enhances the expression of a large set of genes functioning in ribosome biogenesis and protein synthesis. EMBO J 20: 1383- 1393, 2001 Lamont JM, McManamy CS, Pearson AD, Clifford SC, Ellison DW: Combined histopathological and molecular cytogenetic stratification of medulloblastoma patients. Clin Cancer Res 10: 5482-5493, 2004 Stearns D, Chaudhry A, Abel TW, Burger PC, Dang CV, Eberhart CG: C-myc overexpression causes anaplasia in medulloblastoma. Cancer Res 66: 673-681, 2006 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 269 EP 275 PG 7 ER PT J AU Grant, GS Das, R Cerceo, MCh Rubinstein, SW Latimer, JJ AF Grant, G Stephen Das, Rubina Cerceo, M Christina Rubinstein, S Wendy Latimer, J Jean TI Elevated Levels of Somatic Mutation in a Manifesting BRCA1 Mutation Carrier SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE somatic mutation; hypoxanthine-guanine phosphoribosyl transferase; glycophorin A; BRCA1 gene; inherited breast cancer syndrome; Fanconi anemia; xeroderma pigmentosum ID somatic mutation; hypoxanthine-guanine phosphoribosyl transferase; glycophorin A; BRCA1 gene; inherited breast cancer syndrome; Fanconi anemia; xeroderma pigmentosum AB Homozygous loss of activity at the breast cancerpredisposing genes BRCA1 and BRCA2 (FANCD1) confers increased susceptibility to DNA double strand breaks, but this genotype occurs only in the tumor itself, following loss of heterozygosity at one of these loci. Thus, if these genes play a role in tumor etiology as opposed to tumor progression, they must manifest a heterozygous phenotype at the cellular level. To investigate the potential consequences of somatic heterozygosity for a BRCA1 mutation demonstrably associated with breast carcinogenesis on background somatic mutational burden, we applied the two standard assays of in vivo human somatic mutation to blood samples from a manifesting carrier of the Q1200X mutation in BRCA1 whose tumor was uniquely ascertained through an MRI screening study. The patient had an allele-loss mutation frequency of 19.4 x 10-6 at the autosomal GPA locus in erythrocytes and 17.1 x 10-6 at the X-linked HPRT locus in lymphocytes. Both of these mutation frequencies are significantly higher than expected from age-matched disease-free controls (P < 0.05). Mutation at the HPRT locus was similarly elevated in lymphoblastoid cell lines established from three other BRCA1 mutation carriers with breast cancer. Our patient’s GPA mutation frequency is below the level established for diagnosis of homozygous Fanconi anemia patients, but consistent with data from obligate heterozygotes. The increased HPRT mutation frequency is more reminiscent of data from patients with xeroderma pigmentosum, a disease characterized by UV sensitivity and deficiency in the nucleotide excision pathway of DNA repair. Therefore, this BRCA1-associated breast cancer patient manifests a unique phenotype of increased background mutagenesis that likely contributed to the development of her disease independent of loss of heterozygosity at the susceptibility locus. C1 [Grant, G Stephen] Graduate School of Public Health, Department of Environmental and Occupational HealthPittsburgh, PA, USA. [Das, Rubina] Graduate School of Public Health, Department of Environmental and Occupational HealthPittsburgh, PA, USA. [Cerceo, M Christina] Graduate School of Public Health, Department of Environmental and Occupational HealthPittsburgh, PA, USA. [Rubinstein, S Wendy] Northwestern University Feinberg School of Medicine, Department of MedicineChicago, IL, USA. [Latimer, J Jean] University of Pittsburgh, School of Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences and Magee Womens Research InstitutePittsburgh, PA, USA. RP Grant, GS (reprint author), Graduate School of Public Health, Department of Environmental and Occupational Health, Pittsburgh, USA. EM grantsg@upmc.edu CR D’Andrea AD, Grompe M: The Fanconi anaemia/BRCA pathway. Nat Rev Cancer 3: 23-34, 2003 Howlett NG, Taniguchi T, Olson S, Cox B, Waisfisz Q, De Die- Smulders C, Persky N, Grompe M, Joenje H, Pals G, Ikeda H, Fox EA, D’Andrea AD: Biallelic inactivation of BRCA2 in Fanconi anemia. Science 297: 606-609, 2002 Papadopoulos N, Lindblom A: Molecular basis of HNPCC: mutations of MMR genes. Hum Mutat 10: 89-99, 1997 Livingstone LR, White A, Sprouse J, Livanos E, Jacks T, Tlsty TD: Altered cell cycle arrest and gene amplification potential accompany loss of wild-type p53. Cell 70: 923-935, 1992 Nieuwenhuis B, Van Assen-Bolt AJ, Van Waarde-Verhagen MA, Sijmons RH, Van der Hout AH, Bauch T, Streffer C, Kampinga HH: BRCA1 and BRCA2 heterozygosity and repair of X-rayinduced DNA damage. Int J Radiat Biol 78: 285-295, 2002 Trenz K, Rothfuss A, Schutz P, Speit G: Mutagen sensitivity of peripheral blood from women carrying a BRCA1 or BRCA2 mutation. Mutat Res 500: 89-96, 2002 Mamon H, Dahlberg W, Little JB: Hemizygous fibroblast cell strains established from patients with BRCA1 or BRCA2 mutations demonstrate an increased rate of spontaneous mutations and increased radiosensitivity. Int J Radiat Oncol Biol Phys 57(Suppl 2): S346-S347, 2003 Rothfuss A, Schutz P, Bochum S, Volm T, Eberhardt E, Kreienberg R, Vogel W, Speit G: Induced micronucleus frequencies in peripheral lymphocytes as a screening test for carriers of a BRCA1 mutation in breast cancer families. Cancer Res 60: 390-394, 2000 Grant SG, Jensen RH: Use of hematopoietic cells and markers for the detection and quantitation of human in vivo somatic mutation. In: Immunobiology of Transfusion Medicine., Ed: Garratty G), Marcel Dekker, New York, 1993, pp. 299-323 Grant SG, Bigbee WL, Langlois RG, Jensen RH: Methods for the detection of mutational and segregational events: relevance to the monitoring of survivors of childhood cancer. In: Late Effects of Treatment for Childhood Cancer., Eds: Green DM, and D’Angio GJ), Wiley-Liss, New York, 1992, pp. 133-150 Tompa A, Sapi E: Detection of 6-thioguanine resistance in human peripheral blood lymphocytes, PBL, of industrial workers and lung cancer patients. Mutat Res 210: 345-351, 1989 Cole J, Skopek TR: Somatic mutant frequency, mutation rates and mutational spectra in the human population in vivo. Mutat Res 304: 33-105, 1994 Okada S, Ishii H, Nose H, Okusaka T, Kyogoku A, Yoshimori M, Wakabayashi K: Evidence for increased somatic cell mutations in patients with hepatocellular carcinoma. Carcinogenesis 18: 445-449, 1997 Grant SG: Molecular epidemiology of human cancer: biomarkers of genotoxic exposure and susceptibility. J Environ Pathol Toxicol Oncol 20: 245-261, 2001 Langlois RG, Bigbee WL, Jensen RH: The glycophorin A assay for somatic cell mutations in humans. Prog Clin Biol Res 340C: 47-56, 1990 Swift M, Morrell D, Massey RB, Chase CL: Incidence of cancer in 161 families affected by ataxia-telangiecasia. N Engl J Med 325: 1831-1836, 1991 Kerangueven F, Eisinger F, Noguchi T, Allione F, Wargniez V, Eng C, Padberg G, Theillet C, Jacquemier J, Longy M, Sobol H, Birnbaum D: Loss of heterozygosity in human breast carcinomas in the ataxia telangiectasia, Cowden disease and BRCA1 regions. Oncogene 14: 339-347, 1997 Rubinstein WS, Latimer JJ, Sumkin JH, Huerbin M, Grant SG, Vogel VG: Prospective screening study of 0.5 Tesla dedicated magnetic resonance imaging for the detection of breast cancer in young, high risk women. BMC Women’s Health 6: 10, 2006 Latimer JJ, Rubinstein WS, Johnson JM, Kanbour-Shakir A, Vogel VG, Grant SG: Haploinsufficiency for BRCA1 is associated with normal levels of DNA nucleotide excision repair in breast tissue and blood lymphocytes. BMC Med Genet 6: 26, 2005 O’Neill JP, McGinniss MJ, Berman JK, Sullivan LM, Nicklas JA, Albertini RJ: Refinement of a T-lymphocyte cloning assay to quantify the in vivo thioguanine-resistant mutant frequency in humans. Mutagenesis 2: 87-94, 1987 Grant SG: The GPA in vivo somatic mutation assay. Meth Mol Biol 291: 179-195, 2005 Jensen RH, Bigbee WL, Langlois RG: In vivo somatic mutations in the glycophorin A locus of human erythroid cells. Banbury Rep 28: 149-159, 1987 Manchester DK, Nicklas JA, O’Neill JP, Lippert MJ, Grant SG, Langlois RG, Moore DH 3rd, Jensen RH, Albertini RJ, Bigbee WL: Sensitivity of somatic mutations in human umbilical cord blood to maternal environments. Environ Mol Mutagen 26: 203-212, 1995 Grant SG, Reeger W, Wenger SL: Diagnosis of ataxia telangiectasia with the glycophorin A somatic mutation assay. Genet Testing 1: 261-267, 1998 Finette BA, Sullivan LM, O’Neill JP, Nicklas JA, Vacek PM, Albertini RJ: Determination of hprt mutant frequencies in T-lymphocytes from a healthy pediatric population: statistical comparison between newborn, children and adult mutant frequencies, cloning efficiency and age. Mutat Res 308: 223-231, 1994 Tates AD, van Dam FJ, van Mossel H, Schoemaker H, Thijssen JC, Woldring VM, Zwinderman AH, Natarajan AT: Use of the clonal assay for the measurement of frequencies of HPRT mutants in T-lymphocytes from five control populations. Mutat Res 253: 199-213, 1991 Branda RF, Sullivan LM, O’Neill JP, Falta MT, Nicklas JA, Hirsch B, Vacek PM, Albertini RJ: Measurement of HPRT mutant frequencies in T-lymphocytes from healthy human populations. Mutat Res 285: 267-279, 1993 Jones IM, Moore DH, Thomas CB, Thompson CL, Strout CL, Burkhart-Schultz K: Factors affecting HPRT mutant frequency in T-lymphocytes of smokers and nonsmokers. Cancer Epidemiol Biomarkers Prev 2: 249-260, 1993 Finette BA, Kendall H, Vacek PM: Mutational spectral analysis at the HPRT locus in healthy children. Mutat Res 505: 27-41, 2002 Huttner E, Holzapfel B, Kropf S: Frequency of HPRT mutant lymphocytes in a human control population as determined by the T-cell cloning procedure. Mutat Res 348: 83-91, 1995 Becker R, Nikolova T, Wolff I, Lovell D, Huttner E, Foth H: Frequency of HPRT mutants in humans exposed to vinyl chloride via an environmental accident. Mutat Res 494: 87-96, 2001 Sala-Trepat M, Boyse J, Richard P, Papadopoulo D, Moustacchi E: Frequencies of HPRT- lymphocytes and glycophorin A variants erythrocytes in Fanconi anemia patients, their parents and control donors. Mutat Res 289: 115-126, 1993 Evdokimova VN, McLoughlin RK, Wenger SL, Grant SG: Use of the glycophorin A bone marrow somatic mutation assay for rapid, unambiguous identification of Fanconi anemia homozygotes regardless of GPA genotype. Am J Med Genet 135: 59- 65, 2005 Grant SG, Wenger SL, Rubinstein WS, Latimer JJ, Bigbee WL, Auerbach AD: Elevated levels of somatic mutation in homozygotes and heterozygotes for inactivating mutations in the genes of the FA/BRCA DNA repair pathway [abstract]. Am J Hum Genet 75(Suppl): 94, 2004 Vijayalaxmi, Wunder E, Schroeder TM: Spontaneous 6-thioguanine- resistant lymphocytes in Fanconi anemia patients and their heterozygous parents. Hum Genet 70: 264-270, 1985 Tates AD, Bernini LF, Natarajan AT, Ploem JS, Verwoerd NP, Cole J, Green MHL, Arlett CF, Norris PN: Detection of somatic mutants in man: HPRT mutations in lymphocytes and hemoglobin mutations in erythrocytes. Mutat Res 213: 73-82, 1989 Cole J, Green MHL, Stephens G, Waugh AP, Beare D, Steingrimsdottir H, Brideges BA: HPRT somatic mutation data. Prog Clin Biol Res 340C: 25-35, 1990 Norris PG, Limb GA, Hamblin AS, Lehmann AR, Arlett CF, Cole J, Waugh AP, Hawk JL: Immune function, mutant frequency, and cancer risk in the DNA repair defective genodermatoses xeroderma pigmentosum, Cockayne’s syndrome, and trichothiodystrophy. J Invest Dermatol 94: 94-100, 1990 Vermeulen W, Scott RJ, Rodgers S, Muller HJ, Cole J, Arlett CF, Kleijer WJ, Bootsma D, Hoeijmakers JH, Weeda G: Clinical heterogeneity within xeroderma pigmentosum associated with mutations in the DNA repair and transcription gene ERCC3. Am J Hum Genet 54: 191-200, 1994 Anstey AV, Arlett CF, Cole J, Norris PG, Hamblin AS, Limb GA, Lehmann AR, Wilkinson JD, Turner M: Long-term survival and preservation of natural killer cell activity in a xeroderma pigmentosum patient with spontaneous regression and multiple deposits of malignant melanoma. Br J Dermatol 125: 272-278, 1991 Cole J, Arlett CF, Norris PG, Stephens G, Waugh AP, Beare DM, Green MHL: Elevated hprt mutant frequency in circulating T-lymphocytes of xeroderma pigmentosum patients. Mutat Res 273: 171-178, 1992 Lin YW, Kubota M, Hirota H, Furusho K, Tomiwa K, Ochi J, Kasahara Y, Sasaki H, Ohta S: Somatic cell mutation frequency at the HPRT, T-cell antigen receptor and glycophorin A loci in Cockayne syndrome. Mutat Res 337: 49-55, 1995 Akiyama M, Kyoizumi S, Hirai Y, Kusunoki Y, Iwamoto KS, Nakamura N: Mutation frequency in human blood cells increases with age. Mutat Res 338: 141-149, 1995 Morley AA, Cox S, Holliday R: Human lymphocytes resistant to 6-thioguanine increase with age. Mech Ageing Dev 19: 21-26, 1982 Vrieling H, Tates AD, Natarajan AT, van Zeeland AA: Agerelated accumulation of mutations in human T-lymphocytes. Ann N Y Acad Sci 663: 36-42, 1992 Branda RF, O’Neill JP, Jacobson-Kram D, Albertini RJ: Factors influencing mutation at the hprt locus in T-lymphocytes: studies in normal women and women with benign and malignant breast masses. Environ Mol Mutagen 19: 274-281, 1992 Latimer JJ, Kisin E, Zayas-Rivera B, Kanbour-Shakir A, Kelley J, Johnson R, Grant SG: Increased somatic mutation and reduced DNA repair in breast cancer patients and their tumors [abstract]. Proc Am Assoc Cancer Res 40: 440, 1999 Nicklas JA, Lippert MJ, Hunter TC, O’Neill JP, Albertini RJ: Analysis of human HPRT deletion mutations with X-linked probes and pulsed field gel electrophoresis. Environ Mol Mutagen 18: 270-273, 1991 Papadopoulo D, Guillouf C, Mohrenweiser H, Moustacchi E: Hypomutability in Fanconi anemia cells is associated with increased deletion frequency at the HPRT locus. Proc Natl Acad Sci USA 87: 8383-8387, 1990 Laquerbe A, Sala-Trepat M, Vives C, Escarceller M, Papadopoulo D: Molecular spectra of HPRT deletion mutations in circulating T-lymphocytes in Fanconi anemia patients. Mutat Res 431: 341-350, 1999 Elmore E, Swift M: Growth of cultured cells from patients with Fanconi anemia. J Cell Physiol 87: 229-233, 1975 Weksberg R, Buchwald M, Sargent P, Thompson MW, Siminovitch L: Specific cellular defects in patients with Fanconi anemia. J Cell Physiol 101: 311-323, 1979 Thompson LH: Nucleotide excision repair: its relation to human disease. In: DNA Damage and Repair, Volume 2: DNA Repair in Higher Eukaryotes., Eds: Nickoloff JA and Hoekstra MF), Humana, Totowa, NJ, 1998, pp. 335-393 Hartman A-R, Ford JM: BRCA1 induces DNA damage recognition factors and enhances nucleotide excision repair. Nat Genet 32: 180-184, 2002 Takimoto R, MacLachlan TK, Dicker DT, Niitsu Y, Mori T, el- Deiry WS: BRCA1 transcriptionally regulates damaged DNA binding protein, DDB2, in the DNA repair response following UV-irradiation. Cancer Biol Ther 1: 177-186, 2002, comment 1: 187-188, 2002) Kovacs E, Stucki D, Weber W, Muller H: Impaired DNA-repair synthesis in lymphocytes of breast cancer patients. Eur J Cancer Clin Oncol 22: 863-869, 1986 Xiong P, Bondy ML, Li D, Shen H, Wang LE, Singletary SE, Spitz MR, Wei Q: Sensitivity to benzo[a]pyrene diol-epoxide associated with risk of breast cancer in young women and modulation by glutathione S-transferase polymorphisms: a case-control study. Cancer Res 61: 8465-8469, 2001 Kennedy DO, Agrawal M, Shen J, Terry MB, Zhang FF, Senie RT, Motykiewicz G, Santella RM: DNA repair capacity of lymphoblastoid cell lines from sisters discordant for breast cancer. J Natl Cancer Inst 97: 127-132, 2005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 276 EP 283 PG 8 ER PT J AU Jin, PS Kim, HJ Kim, AM Yang, HK Lee, EH Lee, SH Kim, HW AF Jin, Pil Seon Kim, Hun Ji Kim, A Min Yang, Han-Kwang Lee, Eun Hee Lee, Seung Hye Kim, Ho Woo TI Prognostic Significance of Loss of c-Fos Protein in Gastric Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE stomach neoplasms; immunohistochemistry; survival analysis; proto-oncogene protein c-fos; tumor suppressor protein; tissue array analysis ID stomach neoplasms; immunohistochemistry; survival analysis; proto-oncogene protein c-fos; tumor suppressor protein; tissue array analysis AB c-fos was first identified as a viral oncoprotein, and has been studied in terms of its oncogenic function in tumorigenesis. Many experimental and clinical data indicated that c-fos expression plays a role in the progression of several types of carcinomas. However, some recent studies challenge this view as they indicate that c-fos has tumor suppressor activity. In the present study, we assessed c-fos protein expression in 625 consecutive gastric cancers immunohistochemically, and analyzed its relationship with clinicopathologic factors and survival. We found that a loss of c-fos expression is correlated with a more advanced stage, lymph node metastasis, lymphatic invasion and shorter survival, indicating that c-fos expression in gastric cancer cells is lost during progression and that this loss is associated with a poor prognosis. The above findings suggest that loss of c-fos expression has tumor suppressor activity in gastric cancer and we suspect that this suppressor activity might be related to the pro-apoptotic function of c-fos. C1 [Jin, Pil Seon] Seoul National University College of Medicine, Department of Pathology, 28 Yeongeon-dong, 110-799 Seoul, South Korea. [Kim, Hun Ji] Seoul National University College of Medicine, Cancer Research InstituteSeoul, South Korea. [Kim, A Min] Seoul National University College of Medicine, Department of Pathology, 28 Yeongeon-dong, 110-799 Seoul, South Korea. [Yang, Han-Kwang] Seoul National University College of Medicine, Department of SurgerySeoul, South Korea. [Lee, Eun Hee] Seoul National University Bundang Hospital, Department of PathologySeongnam, South Korea. [Lee, Seung Hye] Seoul National University Bundang Hospital, Department of PathologySeongnam, South Korea. [Kim, Ho Woo] Seoul National University College of Medicine, Department of Pathology, 28 Yeongeon-dong, 110-799 Seoul, South Korea. RP Kim, HW (reprint author), Seoul National University College of Medicine, Department of Pathology, 110-799 Seoul, South Korea. EM woohokim@snu.ac.kr CR Halazonetis TD, Georgopoulos K, Greenberg ME, Leder P: c- Jun dimerizes with itself and with c-Fos, forming complexes of different DNA binding affinities. Cell 55: 917-924, 1988 Schutte J, Viallet J, Nau M, Segal S, Fedorko J, Minna J: jun- B inhibits and c-fos stimulates the transforming and trans-activating activities of c-jun. Cell 59: 987-997, 1989 Bakin AV, Curran T: Role of DNA 5-methylcytosine transferase in cell transformation by fos. Science 283: 387-390, 1999 Grigoriadis AE, Schellander K, Wang ZQ, Wagner EF: Osteoblasts are target cells for transformation in c-fos transgenic mice. J Cell Biol 122: 685-701, 1993 Hu E, Mueller E, Oliviero S, Papaioannou VE, Johnson R, Spiegelman BM: Targeted disruption of the c-fos gene demonstrates c-fos dependent and independent pathways for gene expression stimulated by growth factors or oncogenes. EMBO J 13: 3094-3103, 1994 Fialka I, Schwarz H, Reichmann E, Oft M, Busslinger M, Beug H: The estrogen-dependent c-JunER protein causes a reversible loss of mammary epithelial cell polarity involving a destabilization of adherens junctions. J Cell Biol 132: 1115-1132, 1996 Franchi A, Calzolari A, Zampi G: Immunohistochemical detection of c-fos and c-jun expression in osseous and cartilaginous tumours of the skeleton. Virchows Arch 432: 515-519, 1998 Papachristou DJ, Batistatou A, Sykiotis GP, Varakis I, Papavassiliou AG: Activation of the JNK-AP-1 signal transduction pathway is associated with pathogenesis and progression of human osteosarcomas. Bone 32: 364-371, 2003 Gamberi G, Benassi MS, Bohling T, Ragazzini P, Molendini L, Sollazzo MR, Pompetti F, Merli M, Magagnoli G, Balladelli A, Picci P: C-myc and c-fos in human osteosarcoma: prognostic value of mRNA and protein expression. Oncology 55: 556-563, 1998 Bamberger AM, Milde-Langosch K, Rossing E, Goemann C, Loning T: Expression pattern of the AP-1 family in endometrial cancer: correlations with cell cycle regulators. J Cancer Res Clin Oncol 127: 545–550, 2001 Prusty BK, Das BC: Constitutive activation of transcription factor AP-1 in cervical cancer and suppression of human papillomavirus, HPV, transcription and AP-1 activity in HeLa cells by curcumin. Int J Cancer 113: 951-960, 2005 Yuen MF, Wu PC, Lai VC, Lau JY, Lai CL: Expression of c-Myc, c-Fos and c-Jun in hepatocellular carcinoma. Cancer 91: 106- 112, 2001 Wakita K, Ohyanagi H, Yamamoto K, Tokuhisa T, Saitoh Y: Overexpression of c-Ki-ras and c-fos in human pancreatic carcinomas. Int J Pancreatol 11: 43-47, 1992 Bland KI, Konstadoulakis MM, Vezeridis MP, Wanebo HJ: Oncogene protein co-expression. Value of Ha-ras, c-myc, c-fos, and p53 as prognostic discriminants for breast carcinoma. Ann Surg 221: 706-718, 1995 Mikula M, Gotzmann J, Fischer AN, Wolschek MF, Thallinger C, Schulte-Hermann R, Beug H, Mikulits W: The proto-oncoprotein c-Fos negatively regulates hepatocellular tumorigenesis. Oncogene 22: 6725-6738, 2003 American Joint Committee on Cancer: AJCC cancer staging manual 6th ed. Springer-Verlag, New York, 2002 International Agency for Research on Cancer, IARC): World Health Organization Classification of Tumors; pathology and genetics of tumors of the digestive system. IARC Press, Lyon, 2000 Lee HS, Lee HK, Kim HS, Yang HK, Kim YI, Kim WH: MUC1, MUC2, MUC5AC, and MUC6 expressions in gastric carcinomas: their roles as prognostic indicators. Cancer 92: 1427-1434, 2001 Nevine S, Darwish, Kim MA, Chang MS, Lee HS, Lee BL, Kim YI, Kim WH: Prognostic significance of CD24 expression in gastric carcinoma. Cancer Res Treat 36: 298-302, 2004 Kustikova O, Kramerov D, Grigorian M, Berezin V, Bock E, Lukanidin E, Tulchinsky E: Fra-1 induces morphological transformation and increases in vitro invasiveness and mobility of epithelioid adenocarcinoma cells. Mol Cell Biol 18: 7095- 7105, 1998 Levin WJ, Press MF, Gaynor RB, Sukhatme VP, Boone TC, Reissmann PT, Figlin RA, Holmes EC, Souza LM, Slamon DJ: Expression patterns of immediate early transcription factors in human non-small cell lung cancer. The Lung Cancer Study Group. Oncogene 11: 1261-1269, 1995 Liu G, Takano T, Matsuzuka F, Higashiyama T, Kuma K, Amino N: Screening of specific changes in mRNAs in thyroid tumors by sequence specific differential display: decreased expression of c-fos mRNA in papillary carcinoma. Endocr J 46: 459-466, 1999 Fleischmann A, Jochum W, Eferl R, Witowsky J, Wagner EF: Rhabdomyosarcoma development in mice lacking Trp53 and Fos: tumor suppression by the Fos protooncogene. Cancer Cell 4: 477-482, 2003 Grimm C, Wenzel A, Behrens A, Hafezi F, Wagner EF, Reme CE: AP-1 mediated retinal photoreceptor apoptosis is independent of N-terminal phosphorylation of c-Jun. Cell Death Differ 8: 859-867, 2001 Kalra N, Kumar V: c-Fos is a mediator of the c-myc-induced apoptotic signaling in serum-deprived hepatoma cells via the p38 mitogen-activated protein kinase pathway. J Biol Chem 279: 25313-25319, 2004 Silvers AL, Bachelor MA, Bowden GT: The role of JNK and p38 MAPK activities in UVA-induced signaling pathways leading to AP-1 activation and c-Fos expression. Neoplasia 5: 319-329, 2003 Siegmund D, Mauri D, Peters N, Juo P, Thome M, Reichwein M, Blenis J, Scheurich P, Tschopp J, Wajant H: Fas-associated death domain protein, FADD, and caspase-8 mediate up-regulation of c-Fos by Fas ligand and tumor necrosis factor-related apoptosis-inducing ligand, TRAIL, via a FLICE inhibitory protein, FLIP)-regulated pathway. J Biol Chem 276: 32585- 32590, 2001 Willaime S, Vanhoutte P, Caboche J, Lemaigre-Dubreuil Y, Mariani J, Brugg B: Ceramide-induced apoptosis in cortical neurons is mediated by an increase in p38 phosphorylation and not by the decrease in ERK phosphorylation. Eur J Neurosci 13: 2037-2046, 2001 Kondo T, Sharp FR, Honkaniemi J, Mikawa S, Epstein CJ, Chan PH: DNA fragmentation and prolonged expression of c-fos, cjun, and hsp70 in kainic acid-induced neuronal cell death in transgenic mice overexpressing human CuZn-superoxide dismutase. J Cereb Blood Flow Metab 17: 241-256, 1997 Hasegawa K, Litt L, Espanol MT, Sharp FR, Chan PH: Expression of c-fos and hsp70 mRNA in neonatal rat cerebrocortical slices during NMDA-induced necrosis and apoptosis. Brain Res 785: 262-278, 1998 Meyer-ter-Vehn T, Covacci A, Kist M, Pahl HL: Helicobacter pylori activates mitogen-activated protein kinase cascades and induces expression of the proto-oncogenes c-fos and c-jun. J Biol Chem 275: 16064-16072, 2000 Myllykangas S, Monni O, Nagy B, Rautelin H, Knuutila S: Helicobacter pylori infection activates FOS and stress-response genes and alters expression of genes in gastric cancer-specific loci. Genes Chromosomes Cancer 40: 334-341, 2004 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 284 EP 289 PG 6 ER PT J AU Deligezer, U Dalay, N AF Deligezer, Ugur Dalay, Nejat TI Expression of the TRAIL Receptors in Blood Mononuclear Cells in Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TRAIL receptors; expression; circulating tumor cells; translocation ID TRAIL receptors; expression; circulating tumor cells; translocation AB TRAIL receptors are differentially expressed on restricted subpopulations of normal blood cells. In the present study, we investigated the utility of individual TRAIL receptors in evaluating the presence of circulating tumor cells in blood. Patients with chronic myeloid leukemia (CML) carrying the t(9;22) translocation were compared with patients in whom no translocation was detected, with patients with multiple myeloma and with a group of healthy individuals. TRAIL receptor expression was analyzed by RT-PCR in blood mononuclear cells. Blood mononuclear cells of healthy subjects expressed the TRAIL-R1 and TRAIL-R2 death receptors and the TRAIL-R4 decoy receptor while the other decoy receptor TRAIL-R3 was not detectable. This normal expression pattern was also observed in all cases with multiple myeloma and in almost all patients without translocation (42/43; 97.7%). However, in 24/56 (42.9%) of the translocation-positive patients, the expression pattern was completely different. In this group the TRAIL-R4 receptor alone or in combination with TRAIL-R1 disappeared from blood mononuclear cells, while the TRAIL-R2 was expressed at normal level, indicating that the loss of expression is specific for the TRAIL-R4 and TRAIL-R1. This expression pattern was also confirmed by real-time PCR. The differences between the translocation-positive and -negative groups for the TRAIL-R4 and TRAIL-R1 expression were highly significant (p=0.0001 and p=0.0004, respectively). However, the differential expression pattern did not correlate with the number of leukemic cells. Our results suggest a correlation between the presence of leukemic cells in circulation and the differential expression pattern of TRAIL receptors in blood mononuclear cells. C1 [Deligezer, Ugur] Istanbul Medical Faculty, Istanbul University, Oncology Institute, Capa, 34390 Istanbul, Turkey. [Dalay, Nejat] Istanbul Medical Faculty, Istanbul University, Oncology Institute, Capa, 34390 Istanbul, Turkey. RP Dalay, N (reprint author), Istanbul Medical Faculty, Istanbul University, Oncology Institute, 34390 Istanbul, Turkey. EM ndalay@yahoo.com CR Fiorucci G, Vannucchi S, Chiantore MV, et al: TNF-related apoptosis- inducing ligand, TRAIL, as a pro-apoptotic signal transducer with cancer therapeutic potential. Curr Pharm Des 11:933-944, 2005 Kimberley FC, Screaton GR: Following a TRAIL: Update on a ligand and its five receptors. Cell Res 14:359-372, 2004 Pan G, O’Rourke K, Chinnaiyan AM, et al: The receptor for the cytotoxic ligand TRAIL. Science 276:111-113, 1997 Griffith TS, Chin WA, Jackson GC, et al: Intracellular regulation of TRAIL-induced apoptosis in human melanoma cells. J Immunol 161: 2833-2840, 1998 LeBlanc HN, Ashkenazi A: Apo2L/TRAIL and its death and decoy receptors. Cell Death Differ 10:66-75, 2003 Hasegawa H, Yamada Y, Harasawa H, et al: Restricted expression of tumor necrosis factor-related apoptosis-inducing ligand receptor 4 in human peripheral blood lymphocytes. Cell Immunol 231:1-7, 2004 Lum JJ, Pilon AA, Sanchez-Dardon J, et al: Induction of cell death in human immunodeficiency virus-infected macrophages and resting memory CD4 T cells by TRAIL/Apo2l. J Virol 75:11128-11136, 2001 Daigle I, Simon HU: Alternative functions for TRAIL receptors in eosinophils and neutrophils. Swiss Med Wkly 131:231-237, 2001 Renshaw SA, Parmar JS, Singleton V, et al: Acceleration of human neutrophil apoptosis by TRAIL. J Immunol 170:1027-1033, 2003 Billadeau D, Quam L, Thomas W, et al: Detection and quantitation of malignant cells in the peripheral blood of multiple myeloma patients. Blood 80:1818-1824, 1992 Knauf WU, Pochanke G, Ho A: Detection of circulating monoclonal lymphocytes in multiple myeloma patients by analysis of gene rearrangements: correlation with progressive disease. Leuk Res 7:341-345, 1993 Rawstron AC, Owen RG, Davies FE, et al: Circulating plasma cells in multiple myeloma: characterization and correlation with disease stage. Br J Haematol 97:46-55, 1997 Zojer N, Schuster-Kolbe J, Assmann I, et al: Chromosomal aberrations are shared by malignant plasma cells and a small fraction of circulating CD19+ cells in patients with myeloma and monoclonal gammopathy of undetermined significance. Br J Haematol 117:852- 859, 2002 Deligezer U, Erten N, Akisik EE, et al: Circulating fragmented nucleosomal DNA and caspase-3 mRNA in patients with lymphoma and myeloma. Exp Mol Pathol 80:72-76, 2006 Griffith TS, Wiley SR, Kubin MZ, et al: Monocyte-mediated tumoricidal activity via the tumor necrosis factor-related cytokine, TRAIL. J Exp Med 189:1343-1354, 1999 Inoue H, Shiraki K, Yamanaka T, et al: Functional expression of tumor necrosis factor-related apoptosis-inducing ligand in human colonic adenocarcinoma cells. Lab Invest 82:1111-1119, 2002 Janicke RU, Sprengart ML, Wati MR, et al: Caspase-3 is required for DNA fragmentation and morphological changes associated with apoptosis. J Biol Chem 273:9357-9360, 1998 van Dongen JJ, Macintyre EA, Gabert JA, et al: Standardized RTPCR analysis of fusion gene transcripts from chromosome aberrations in acute leukemia for detection of minimal residual disease. Report of the BIOMED-1 Concerted Action: investigation of minimal residual disease in acute leukemia. Leukemia 13:1901-1928, 1999 Narita M, Takahashi M, Liu A, et al: Leukemia blast-induced T-cell anergy demonstrated by leukemia-derived dendritic cells in acute myelogenous leukemia.Exp Hematol 26:709-719, 2001 MacFarlane M, Kohlhaas SL, Sutcliffe MJ, et al: TRAIL receptorselective mutants signal to apoptosis via TRAIL-R1 in primary lymphoid malignancies. Cancer Res 65:11265-11270, 2005 Kaufmann SH, Steensma DP: On the TRAIL of a new therapy for leukemia. Leukemia 19:2195-2202, 2005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 290 EP 294 PG 5 ER PT J AU Felber, M Sonnemann, J Beck, FJ AF Felber, Matthias Sonnemann, Jurgen Beck, F James TI Inhibition of Novel Protein Kinase C-epsilon Augments TRAIL-induced Cell Death in A549 Lung Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lung cancer; medulloblastoma; NPC 15437; protein kinase C-epsilon; TNF; TRAIL ID lung cancer; medulloblastoma; NPC 15437; protein kinase C-epsilon; TNF; TRAIL AB Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has great potential for cancer treatment since it provokes cell death in most tumor cells while leaving most normal cells unscathed. Some cancers, however, show resistance to TRAIL, indicating that TRAIL alone may be insufficient for cancer therapy. Here we studied whether the apoptotic susceptibility of A549 non-small cell lung cancer cells could be modulated by inhibiting protein kinase C (PKC). We show that an inhibitor with preference for novel PKC isozymes, NPC 15437, significantly augmented TRAIL sensitivity of A549 cells, as judged by assessing cell death and mitochondrial membrane potential. Likewise, NPC 15437 also significantly potentiated the responsiveness of DAOY medulloblastoma cells to TRAIL. In contrast, an inhibitor with preference for conventional PKC isozymes, Go6976, did not augment TRAIL sensitivity of A549 cells. To further specify the PKC isozyme responsible for TRAIL sensitization, we used a peptide inhibitor with selectivity for the novel PKC isozyme epsilon, myr-PKC-epsilon V1-2. The inhibition of PKC-epsilon resulted in a significant amplification of the cytotoxic activity of TRAIL in A549 cells. Altogether, our study provides evidence for a considerable role of PKC-epsilon in the apoptotic responsiveness of A549 lung cancer cells, and possibly other malignancies, to TRAIL. C1 [Felber, Matthias] Ernst Moritz Arndt University, Research Center of Pharmacology and Experimental TherapeuticsGreifswald, Germany. [Sonnemann, Jurgen] Ernst Moritz Arndt University, Research Center of Pharmacology and Experimental TherapeuticsGreifswald, Germany. [Beck, F James] Ernst Moritz Arndt University, Department of Pediatric Oncology/Hematology, Soldmannstr. 15, D-17475 Greifswald, Germany. RP Beck, FJ (reprint author), Ernst Moritz Arndt University, Department of Pediatric Oncology/Hematology, D-17475 Greifswald, Germany. EM beck@uni-greifswald.de CR Baader E, Toloczko A, Fuchs U, et al: Tumor necrosis factorrelated apoptosis-inducing ligand-mediated proliferation of tumor cells with receptor-proximal apoptosis defects. Cancer Res 65: 7888-7895, 2005. Basu A, Lu D, Sun B, et al: Proteolytic activation of protein kinase C-epsilon by caspase-mediated processing and transduction of antiapoptotic signals. J Biol Chem 277: 41850- 41856, 2002. Basu A, Weixel KM: Comparison of protein kinase C activity and isoform expression in cisplatin-sensitive and -resistant ovarian carcinoma cells. Int J Cancer 62: 457-460, 1995. Carney DN: Lung cancer—time to move on from chemotherapy. N Engl J Med 346: 126-128, 2002. Courage C, Budworth J, Gescher A: Comparison of ability of protein kinase C inhibitors to arrest cell growth and to alter cellular protein kinase C localisation. Br J Cancer 71: 697-704, 1995. Ding L, Wang H, Lang W, et al: Protein kinase C-epsilon promotes survival of lung cancer cells by suppressing apoptosis through dysregulation of the mitochondrial caspase pathway. J Biol Chem 277: 35305-35313, 2002. Fan QL, Zou WY, Song LH, et al: Synergistic antitumor activity of TRAIL combined with chemotherapeutic agents in A549 cell lines in vitro and in vivo. Cancer Chemother Pharmacol 55: 189-196, 2005. Fesik SW: Promoting apoptosis as a strategy for cancer drug discovery. Nat Rev Cancer 5: 876-885, 2005. Frese S, Brunner T, Gugger M, et al: Enhancement of Apo2L/TRAIL, tumor necrosis factor-related apoptosis-inducing ligand)-induced apoptosis in non-small cell lung cancer cell lines by chemotherapeutic agents without correlation to the expression level of cellular protease caspase-8 inhibitory protein. J Thorac Cardiovasc Surg 123: 168-174, 2002. Fulda S, Debatin KM: Exploiting death receptor signaling pathways for tumor therapy. Biochim Biophys Acta 1705: 27- 41, 2004. Ganten TM, Koschny R, Sykora J, et al: Preclinical differentiation between apparently safe and potentially hepatotoxic applications of TRAIL either alone or in combination with chemotherapeutic drugs. Clin Cancer Res 12: 2640-2646, 2006. Gillespie S, Zhang XD, Hersey P: Variable expression of protein kinase C epsilon in human melanoma cells regulates sensitivity to TRAIL-induced apoptosis. Mol Cancer Ther 4: 668- 676, 2005. Harper N, Hughes MA, Farrow SN, et al: Protein kinase C modulates TRAIL-induced apoptosis by targeting the apical events of death receptor signaling. J Biol Chem 278: 44338- 44347, 2003. Held J, Schulze-Osthoff K: Potential and caveats of TRAIL in cancer therapy. Drug Resist Updat 4: 243-252, 2001. Hofmann J: Protein kinase C isozymes as potential targets for anticancer therapy. Curr Cancer Drug Targets 4: 125-146, 2004. Jemal A, Siegel R, Ward E, et al: Cancer statistics, 2006. CA Cancer J Clin 56: 106-130, 2006. Jin H, Yang R, Fong S, et al: Apo2 ligand/tumor necrosis factor- related apoptosis-inducing ligand cooperates with chemotherapy to inhibit orthotopic lung tumor growth and improve survival. Cancer Res 64: 4900-4905, 2004. Johnson JA, Gray MO, Chen CH, et al: A protein kinase C translocation inhibitor as an isozyme-selective antagonist of cardiac function. J Biol Chem 271: 24962-24966, 1996. Johnstone RW, Ruefli AA, Lowe SW: Apoptosis: a link between cancer genetics and chemotherapy. Cell 108: 153-164, 2002. Kim CY, Giaccia AJ, Strulovici B, et al: Differential expression of protein kinase C epsilon protein in lung cancer cell lines by ionising radiation. Br J Cancer 66: 844-849, 1992. Laack E, Dickgreber N, Muller T, et al: Randomized phase III study of gemcitabine and vinorelbine versus gemcitabine, vinorelbine, and cisplatin in the treatment of advanced nonsmall- cell lung cancer: from the German and Swiss Lung Cancer Study Group. J Clin Oncol 22: 2348-2356, 2004. LeBlanc HN, Ashkenazi A: Apo2L/TRAIL and its death and decoy receptors. Cell Death Differ 10: 66-75, 2003. Manna SK, Aggarwal BB: All-trans-retinoic acid upregulates TNF receptors and potentiates TNF-induced activation of nuclear factors-kappaB, activated protein-1 and apoptosis in human lung cancer cells. Oncogene 19: 2110-2119, 2000. Martiny-Baron G, Kazanietz MG, Mischak H, et al: Selective inhibition of protein kinase C isozymes by the indolocarbazole Go 6976. J Biol Chem 268: 9194-9197, 1993. McJilton MA, Van Sikes C, Wescott GG, et al: Protein kinase Cepsilon interacts with Bax and promotes survival of human prostate cancer cells. Oncogene 22: 7958-7968, 2003. Mitsiades N, Mitsiades CS, Poulaki V, et al: Intracellular regulation of tumor necrosis factor-related apoptosis-inducing ligand- induced apoptosis in human multiple myeloma cells. Blood 99: 2162-2171, 2002. Monick M, Staber J, Thomas K, et al: Respiratory syncytial virus infection results in activation of multiple protein kinase C isoforms leading to activation of mitogen-activated protein kinase. J Immunol 166: 2681-2687, 2001. Odoux C, Albers A, Amoscato AA, et al: TRAIL, FasL and a blocking anti-DR5 antibody augment paclitaxel-induced apoptosis in human non-small-cell lung cancer. Int J Cancer 97: 458-465, 2002. Okhrimenko H, Lu W, Xiang C, et al: Protein kinase C-epsilon regulates the apoptosis and survival of glioma cells. Cancer Res 65: 7301-7309, 2005. Okhrimenko H, Lu W, Xiang C, et al: Roles of tyrosine phosphorylation and cleavage of protein kinase Cdelta in its protective effect against tumor necrosis factor-related apoptosis inducing ligand-induced apoptosis. J Biol Chem 280: 23643- 23652, 2005. Pardo OE, Wellbrock C, Khanzada UK, et al: FGF-2 protects small cell lung cancer cells from apoptosis through a complex involving PKCepsilon, B-Raf and S6K2. EMBO J 25: 3078- 3088, 2006. Platzbecker U, Ward JL, Deeg HJ: Chelerythrin activates caspase- 8, downregulates FLIP long and short, and overcomes resistance to tumour necrosis factor-related apoptosis-inducing ligand in KG1a cells. Br J Haematol 122: 489-497, 2003. Reed JC: Drug insight: Cancer therapy strategies based on restoration of endogenous cell death mechanisms. Nat Clin Pract Oncol 3:388-398, 2006. Saraiva L, Fresco P, Pinto E, et al: Isoform-selectivity of PKC inhibitors acting at the regulatory and catalytic domain of mammalian PKC-alpha, -betaI, -delta, -eta and -zeta. J Enzyme Inhib Med Chem 18: 475-483, 2003. Sarker M, Ruiz-Ruiz C, Lopez-Rivas A: Activation of protein kinase C inhibits TRAIL-induced caspases activation, mitochondrial events and apoptosis in a human leukemic T cell line. Cell Death Differ 8: 172-181, 2001. Schiller JH, Harrington D, Belani CP, et al: Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 346: 92-98, 2002. Shi RX, Ong CN, Shen HM: Protein kinase C inhibition and xlinked inhibitor of apoptosis protein degradation contribute to the sensitization effect of luteolin on tumor necrosis factorrelated apoptosis-inducing ligand-induced apoptosis in cancer cells. Cancer Res 65: 7815-7823, 2005. Shimomoto H, Hasegawa Y, Nozaki Y, et al: Expression of tumor necrosis factor receptors in human lung cancer cells and normal lung tissues. Am J Respir Cell Mol Biol 13: 271-278, 1995. Shinohara H, Kayagaki N, Yagita H, et al: A protective role of PKCepsilon against TNF-related apoptosis-inducing ligand, TRAIL)-induced apoptosis in glioma cells. Biochem Biophys Res Commun 284: 1162-1167, 2001. Sivaprasad U, Shankar E, Basu A: Downregulation of Bid is associated with PKCepsilon-mediated TRAIL resistance. Cell Death Differ 14: 851-860, 2007. Sonnemann J, Gange J, Kumar KS, et al: Histone deacetylase inhibitors interact synergistically with tumor necrosis factorrelated apoptosis-inducing ligand, TRAIL, to induce apoptosis in carcinoma cell lines. Invest New Drugs 23: 99-109, 2005. Sonnemann J, Gekeler V, Sagrauske A, et al: Down-regulation of protein kinase Ceta potentiates the cytotoxic effects of exogenous tumor necrosis factor-related apoptosis-inducing ligand in PC-3 prostate cancer cells. Mol Cancer Ther 3: 773- 781, 2004. Spierings DC, de Vries EG, Timens W, et al: Expression of TRAIL and TRAIL death receptors in stage III non-small cell lung cancer tumors. Clin Cancer Res 9: 3397-3405, 2003. Sullivan JP, Connor JR, Shearer BG, et al: 2,6-Diamino-N-([1-, 1-oxotridecyl)-2-piperidinyl] methyl)hexanamide, NPC 15437): a novel inhibitor of protein kinase C interacting at the regulatory domain. Mol Pharmacol 41: 38-44, 1992. Tillman DM, Izeradjene K, Szucs KS, et al: Rottlerin sensitizes colon carcinoma cells to tumor necrosis factor-related apoptosis- inducing ligand-induced apoptosis via uncoupling of the mitochondria independent of protein kinase C. Cancer Res 63: 5118-5125, 2003. Trauzold A, Wermann H, Arlt A, et al: CD95 and TRAIL receptor- mediated activation of protein kinase C and NF-kappaB contributes to apoptosis resistance in ductal pancreatic adenocarcinoma cells. Oncogene 20: 4258-4269, 2001. van Valen F, Fulda S, Schafer KL, et al: Selective and nonselective toxicity of TRAIL/Apo2L combined with chemotherapy in human bone tumour cells vs. normal human cells. Int J Cancer 107: 929-940, 2003. Wu D, Thakore CU, Wescott GG, et al: Integrin signaling links protein kinase Cepsilon to the protein kinase B/Akt survival pathway in recurrent prostate cancer cells. Oncogene 23: 8659- 8672, 2004. Zhang J, Liu N, Zhang J, et al: PKCdelta protects human breast tumor MCF-7 cells against tumor necrosis factor-related apoptosis- inducing ligand-mediated apoptosis. J Cell Biochem 96: 522-532, 2005. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 295 EP 301 PG 7 ER PT J AU Hajdu, M Luttun, A Pelacho, B Burns, CT Chase, L Gutierrez-Perez, M Jiang, Y Lenvik, T Vas, V Uher, F Sebestyen, A Verfaillie, C AF Hajdu, Melinda Luttun, Aernout Pelacho, Beatriz Burns, C Terry Chase, Lucas Gutierrez-Perez, Maria Jiang, Yuehua Lenvik, Todd Vas, Virag Uher, Ferenc Sebestyen, Anna Verfaillie, Catherine TI Transcriptional Characterization of the Notch Signaling Pathway in Rodent Multipotent Adult Progenitor Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Notch; HES-1; multipotent adult progenitor cell; stem cell ID Notch; HES-1; multipotent adult progenitor cell; stem cell AB The Notch signaling pathway is a multifunctional, evolutionarily conserved pathway, which plays an important role in development as well as stem cell biology. Multipotent adult progenitor cells (MAPCs) represent a unique stem cell population, which is capable of differentiating into cell types of the ectodermal, mesodermal and endodermal lineages in vitro, and contribute to most somatic cell types in vivo. Our aim was to characterize the gene expression of Notch signaling elements in rodent MAPCs. We show that transcripts for Notch-receptors, ligands, regulatory molecules of the pathway and the Hairy/Enhancer of Split-1 (HES-1) target gene are present in mouse and rat low-Oct4 MAPCs. We found that mouse Notch3 and rat Notch1 transcripts increased when cells were cultured at high density for 48 to 96 hours. HES-1 and HES-related transcription factor-1 (HERP-1), transcriptional targets of Notch-signaling, were both elicited by immobilized Delta1 ligand. In addition, mRNA for Notch1 and Notch3 was also induced by Notch-signaling, suggesting the presence of regulatory feedback loops. Slight differences between mouse and rat derived MAPCs suggest that the exact function, transcriptional regulation and the fine-tuning of the signal may be species specific. Taken together, we characterized the gene expression profile of the Notch pathway in rodent low-Oct4-MAPCs, and showed that the pathway is functional and can be modulated. Our results provide an additional tool and a further basis for a better understanding of stem cell biology. C1 [Hajdu, Melinda] University of Minnesota Medical School, Stem Cell InstituteMinneapolis, MN, USA. [Luttun, Aernout] University of Minnesota Medical School, Stem Cell InstituteMinneapolis, MN, USA. [Pelacho, Beatriz] University of Minnesota Medical School, Stem Cell InstituteMinneapolis, MN, USA. [Burns, C Terry] University of Minnesota Medical School, Stem Cell InstituteMinneapolis, MN, USA. [Chase, Lucas] University of Minnesota Medical School, Stem Cell InstituteMinneapolis, MN, USA. [Gutierrez-Perez, Maria] University of Navarra, Hematology and Cell Therapy, Clinica UniversitariaPamplona, Spain. [Jiang, Yuehua] University of Minnesota Medical School, Stem Cell InstituteMinneapolis, MN, USA. [Lenvik, Todd] University of Minnesota Medical School, Stem Cell InstituteMinneapolis, MN, USA. [Vas, Virag] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Uher, Ferenc] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., H-1085 Budapest, Hungary. [Verfaillie, Catherine] University of Minnesota Medical School, Stem Cell InstituteMinneapolis, MN, USA. RP Hajdu, M (reprint author), University of Minnesota Medical School, Stem Cell Institute, Minneapolis, USA. EM melindahajdu@freemail.hu CR Artavanis-Tsakonas S, Rand MD, Lake RJ: Notch signaling: cell fate control and signal integration in development. Science 284: 770-776, 1999. Mumm JS, Kopan R: Notch Signaling: From the Outside In. Dev Biol 228: 151-165, 2000. Jarriault S, Brou C, Logeat F, Schroeter EH, Kopan R, Israel A: Signalling downstream of mammalian Notch. Nature 377: 355-358, 1995. Iso T, Kedes L, Hamamori Y: HES and HERP Families: Multiple Effectors of the Notch Signaling Pathway. J Cellular Physiol 194: 237-255, 2003. Fischer A, Gessler M: Delta-Notch – and then? Protein interactions and proposed modes of repression by Hes and Hey bHLH factors. Nucleic Acids Res 35: 4583-4596, 2007. Liu WH, Lai MZ: Deltex Regulates T-Cell Activation by Targeted Degradation of Active MEKK1. Mol Cell Biol 25: 1367- 1378, 2005. Kojika A, Griffin JD: Notch receptors and hematopoiesis. Exp Hematol 29: 1041-1052, 2001. Anderson AC, Kitchens EA, Chan SW, St. Hill C, Jan YN, Zhong W, Robey EA: The Notch Regulator Numb Links the Notch and TCR Signaling Pathways. J Immunol 174: 890-897, 2005. Hicks C, Johnston SH, diSibio G, Collazo A, Vogt TF, Weinmaster G: Fringe differentially modulates Jagged1 and Delta1 signaling through Notch1 and Notch2. Nature Cell Biol 2: 515- 520, 2000. Milner LA, Bigas A: Notch as a Mediator of Cell Fate Determination in Hematopoiesis: Evidence and Speculation. Blood 93: 2431-2448, 1999. Gaiano N, Fishell G: The Role of Notch in Promoting Glial and Neural Stem Cell Fates. Annu Rev Neurosci 25: 471-490, 2002. Chiba S: Concise Review: Notch Signaling in Stem Cell Systems. Stem Cells 24: 2437-2447, 2006. Karanu FN, Yuefei L, Gallacher L, Sakano S, Bhatia M: Differential response of primitive human CD34- and CD34+ hematopoietic cells to the Notch ligand Jagged-1. Leukemia 17: 1366-1374, 2003. Walsh J, Andrews PW: Expression of Wnt and Notch pathway genes in a pluripotent human embryonal carcinoma cell line and embryonic stem cells. APMIS 111: 197-211, 2003. Noggle SA, Weiler D, Condie BG: Notch Signaling Is Inactive but Inducible in Human Embryonic Stem Cells. Stem Cells 24: 1646-1653, 2006. Rho JY, Yu K, Han JS, Chae JI, Koo DB, Yoon HS, Moon SY, Lee KK, Han YM: Transcriptional profiling of the developmentally important signalling pathways in human embryonic stem cells. Human Reproduction 21: 405-412, 2006. Tokunaga A, Kohyama J, Yoshida T, Nakao K, Sawamoto K, Okano H: Mapping spatio-temporal activation of Notch signaling during neurogenesis and gliogenesis in the developing mouse brain. J Neurochem 90: 142-154, 2004. Androutsellis-Theotokis A, Leker RR, Soldner F, Hoeppner DJ, Ravin R, Poser SW, Rueger MA, Bae SK, Kittappa R, McKay RDG: Notch signalling regulates stem cell numbers in vitro and in vivo. Nature 442: 823-826, 2006. Hitoshi S, Alexson T, Tropepe V, Donoviel D, Elia AJ, Nye JS, Conlon RA, Mak TW, Bernstein A, van der Kooy D: Notch pathway molecules are essential for the maintenance, but not the generation, of mammalian neural stem cells. Genes Dev 16: 846-858, 2002. Fan X, Matsui W, Khaki L, Stearns D, Chun J, Li YM, Eberhart CG: Notch Pathway Inhibition Depletes Stem-like Cells and Blocks Engraftment in Embryonal Brain Tumors. Cancer Res 66: 7445-7452, 2006. Duncan AW, Rattis FM, DiMascio LN, Congdon KL, Pazianos G, Zhao C, Yoon K, Cook JM, Willert K, Gaiano N, Reya T: Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance. Nature Immunology 6: 314-322, 2005. Mungamuri SK, Yang XH, Thor AD, Somasundaram K: Survival Signaling by Notch1: Mammalian Target or Rapamycin, mTOR)-Dependent Inhibition of p53. Cancer Res 66: 4715- 4724, 2006. Nagao M, Sugimori M, Nakafuku M: Crosstalk between Notch and growth factor/cytokine signaling pathways in neural stem cells. Mol Cell Biol 27: 3982-3994, 2007. Varnum-Finney B, Xu L, Brashem-Stein C, Nourigat C, Flowers D, Bakkour S, Pear WS, Bernstein ID: Pluripotent, cytokinedependent, hematopoietic stem cells are immortalized by constitutive Notch1 signaling. Nature Medicine 6: 1278-1281, 2000. Jones P, May G, Healy L, Brown J, Hoyne G, Delassus S, Enver T: Stromal Expression of Jagged 1 Promotes Colony Formation by Fetal Hematopoietic Progenitor Cells. Blood 92: 1505-1511, 1998. Karanu FN, Murdoch B, Miyabayashi T, Ohno M, Koremoto M, Gallacher L, Wu D, Itoh A, Sakano S, Bhatia M: Human homologues of Delta-1 and Delta-4 function as mitogenic regulators of primitive human hematopoietic cells. Blood 97: 1960-1967, 2000. Walker L, Lynch M, Silverman S, Fraser J, Boulter J, Weinmaster G, Gasson JC: The Notch/Jagged Pathway Inhibits Proliferation of Human Hematopoietic Progenitors In Vitro. Stem Cells 17: 162-171, 1999. Lanner F, Sohl M, Farnebo F: Functional arterial and venous fate is determined by graded VEGF signaling and notch status during embryonic stem cell differentiation. Arterioscler Thromb Vasc Biol 27: 487-493, 2007. Gruen L, Grabel L: Concise review: scientific and ethical roadblocks to human embryonic stem cell therapy. Stem Cells 24: 2162-2169, 2006. Dezawa M, Kanno H, Hoshino M, Cho H, Matsumoto N, Itokazu Y, Tajima Y, Yamada H, Sawada H, Ishikawa H, Mimura T, Kitada M, Suzuki Y, Ide C: Specific induction of neuronal cells from bone marrow stromal cells and application for autologous transplantation. J Clin Invest 113: 1701-1710, 2004. Reyes M, Lund T, Lenvik T, Aguiar D, Koodie L, Verfaillie CM: Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells. Blood 98: 2615-2625, 2001. Jiang Y, Jahargirdar BN, Reinhardt RL, Schwartz RE, Keene, CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, Du J, Aldrich S, Lisberg A, Low WC, Largaespada DA, Verfaillie CM: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418: 41-49, 2002. Jiang Y, Vaessen B, Lenvik T, Blackstad M, Reyes M, Verfaillie CM: Multipotent progenitor cells can be isolated from postnatal murine bone marrow, muscle, and brain. Exp Hematol 30: 896-904, 2002. Schwartz RE, Reyes M, Koodie L, Jiang Y, Blackstad M, Lund T, Lenvik T, Johnson S, Hu WS, Verfaillie CM: Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells. J Clin Invest 109: 1291-1302, 2002. Jiang Y, Henderson D, Blackstad M, Chen A, Miller RF, Verfaillie CM: Neuroectodermal differentiation from mouse multipotent adult progenitor cells. Proc Natl Acad Sci USA 100 Suppl 1: 11854-11860, 2003. Breyer A, Estharabadi N, Oki M, Ulloa F, Nelson-Holte M, Lien L, Jian Y: Multipotent adult progenitor cell isolation and culture procedures. Exp Hematol 34: 1596-1601, 2006. Ulloa-Montoya F, Kidder BL, Pauwelyn KA, Chase LG, Luttun A, Crabbe A, Geraerts M, Sharov AA, Piao Y, Ko MS, Hu WS, Verfaillie CM: Comparative transcriptome analysis of embryonic and adult stem cells with extended and limited differentiation capacity. Genome Biol 6: R163, 2007. Anjos-Afonso F, Bonnet D: Nonhematopoietic/endothelial SSEA-1+ cells define the most primitive progenitors in the adult murine bone marrow mesenchymal compartment. Blood 193: 1298-306, 2007. Aranguren XL, Luttun A, Clavel C, Moreno C, Abizanda G, Barajas MA, Pelacho B, Uriz M, Arana M, Echavarri A, Soriano M, Andreu EJ, Merino J, Garcia-Verdugo JM, Verfaillie CM, Prosper F: In vitro and in vivo arterial differentiation of human multipotent adult progenitor cells. Blood 109: 2634- 2642, 2007. Carlson ME, Conboy IM: Regulating the Notch pathway in embryonic, adult and old stem cells. Current Opinion Pharmacol 7: 1-7, 2007. Ramalho-Santos M, Yoon S, Matsuzaki Y, Mulligan RC, Melton DA: "Stemness": Transcriptional Profiling of Embryonic and Adult Stem Cells. Science 298: 597-600, 2002. Neves H, Weerkamp F, Gomes AC, Naber BAE, Gameiro P, Becker JD, Lucio P, Clode N, van Dongen JJM, Staal FJT, Parreira L: Effects of Delta1 and Jagged1 on Early Human Hematopoiesis: Correlation with Expression of Notch Signaling- Related Genes in CD34+ Cells. Stem Cells 24: 1328- 1337, 2006. Zavadil J, Cermak L, Soto-Nieves N, Bottinger EP: Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO J 23: 1155- 1165, 2004. Curry CL, Reed LL, Nickoloff BJ, Miele L, Foreman KE: Notch-independent regulation of Hes-1 expression by c-Jun Nterminal kinase signaling in human endothelial cells. Lab Invest 86: 842-852, 2006. Stockhausen MT, Sjolund J, Axelson H: Regulation of the Notch target gene Hes-1 by TGFalpha induced Ras/MAPK signaling in human neuroblastoma cells. Exp Cell Res 310: 218-228, 2005. Yoshiura S, Ohtsuka T, Takenaka Y, Nagahara H, Yoshikawa K, Kageyama R: Ultradian oscillations of Stat, Smad, and Hes1 expression in response to serum. Proc Natl Acad Sci 104: 11292-11297, 2007. Yu X, Alder JK, Chun JH, Friedman AD, Heimfeld S, Cheng L, Civin CI: HES1 inhibits cycling of hematopoietic progenitor cells via DNA binding. Stem Cells 24: 876-888, 2006. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 302 EP 310 PG 9 ER PT J AU Varga, G Kiss, J Varkonyi, J Vas, V Farkas, P Paloczi, K Uher, F AF Varga, Gergely Kiss, Judit Varkonyi, Judit Vas, Virag Farkas, Peter Paloczi, Katalin Uher, Ferenc TI Inappropriate Notch Activity and Limited Mesenchymal Stem Cell Plasticity in the Bone Marrow of Patients with Myelodysplastic Syndromes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cobblestone area-forming cells; Jagged-1; myelodysplastic syndromes; Notch signaling; stem cell plasticity ID cobblestone area-forming cells; Jagged-1; myelodysplastic syndromes; Notch signaling; stem cell plasticity AB Myelodysplastic syndromes (MDSs) are a heterogeneous group of hematological disorders characterized by ineffective hematopoiesis, enhanced bone marrow apoptosis and frequent progression to acute myeloid leukemia. Several recent studies suggested that, besides the abnormal development of stem cells, microenvironmental alterations are also present in the MDS bone marrow. In this study, we have examined the relative frequencies of stem and progenitor cell subsets of MDS and normal hematopoietic cells growing on stromal cell layers established from MDS patients and from normal donors. When hematopoietic cells from MDS patients were co-cultured with normal stromal cells, the frequency of either early or late cobblestone area-forming cells (CAFC) was significantly lower compared to the corresponding normal control values in 4 out of 8 patients. In the opposite situation, when normal hematopoietic cells were incubated on MDS stromal cells, the CAFC frequencies were decreased in 5 out of 6 patients, compared to normal stromal layer-containing control cultures. Moreover, a soluble Notch ligand (Jagged-1 protein) was an inhibitor of day-35-42 CAFC when normal hematopoietic cells were cultured with normal or MDS stromal cells, but was unable to inhibit MDS stem and early progenitor cell growth (day-35-42 CAFC) on pre-established stromal layers. These findings suggest that in early hematopoietic cells isolated from MDS patients the Notch signal transduction pathway is disrupted. Furthermore, there was a marked reduction in the plasticity of mesenchymal stem cells of MDS patients compared with those of normal marrow donors, in neurogenic and adipogenic differentiation ability and hematopoiesis supporting capacity in vitro. These results are consistent with the hypothesis that when alterations are present in the myelodysplastic stroma environment along with intrinsic changes in a hematopoietic stem/progenitor cell clone, both factors might equally contribute to the abnormal hematopoiesis in MDS. C1 [Varga, Gergely] Semmelweis University, 3rd Department of Internal MedicineBudapest, Hungary. [Kiss, Judit] National Medical Center, Department of Hematology and Stem Cell Transplantation, Dioszegi ut 64., H-1113 Budapest, Hungary. [Varkonyi, Judit] Semmelweis University, 3rd Department of Internal MedicineBudapest, Hungary. [Vas, Virag] National Medical Center, Department of Hematology and Stem Cell Transplantation, Dioszegi ut 64., H-1113 Budapest, Hungary. [Farkas, Peter] Semmelweis University, 3rd Department of Internal MedicineBudapest, Hungary. [Paloczi, Katalin] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Uher, Ferenc] National Medical Center, Department of Hematology and Stem Cell Transplantation, Dioszegi ut 64., H-1113 Budapest, Hungary. RP Uher, F (reprint author), National Medical Center, Department of Hematology and Stem Cell Transplantation, H-1113 Budapest, Hungary. EM uher@kkk.org.hu CR Aizawa S, Nakano M, Iwase O, et al: Bone marrow stroma from refractory anemia of myelodysplastic syndrome is defective in its ability to support normal CD34-positive cell proliferation and differentiation in vitro. Leuk Res 23: 239-246, 1999 Alvi S, Shaher A, Shetty V, et al: Successful establishment of long-term bone marrow cultures in 103 patients with myelodysplastic syndromes. Leuk Res 25: 941-954, 2001 Artavanis-Tsakonas S, Rand MD, Lake RJ: Notch signaling: cell fate control and signal integration in development. Science 284: 770-776, 1999 Baldron V, Ruiz-Hidalgo MJ, Nueda ML, et al: Dlk acts as a negative regulator of Notch1 activation through interactions with specific EGF-like repeats. Exp Cell Res 303: 343-359, 2005 Borojevic R, Roela RA, Rodarte RS, et al: Bone marrow stroma in childhood myelodysplastic syndrome: composition, ability to sustain hematopoiesis in vitro, and altered gene expression. Leuk Res 28: 831-844, 2004 Breems DA, Blokland EAW, Neben S, Ploemacher RE: Frequency analysis of human primitive haematopoietic stem cell subsets using a cobblestone area forming cell assay. Leukemia 8: 1095-1104, 1994 Coutinho LH, Geary CG, Chang J, et al: Functional studies of bone marrow haematopoietic and stromal cells in the myelodysplastic syndrome, MDS). Br J Haematol 75: 16-25, 1990 Flores-Figueroa E, Arana-Trejo RM, Gutierrez-Espindola G, et al: Mesenchymal stem cells in myelodysplastic syndromes: phenotypic and cytogenetic characterization. Leuk Res 29: 215-224, 2005 Flores-Figueroa E, Gutierrez-Espindola G, Montesinos JJ, et al: In vitro characterization of hematopoietic microenvironment cells from patients with myelodysplastic syndrome. Leuk Res 26: 677-686, 2002 Hofmann W-K, de Vos S, Komor M, et al: Characterization of gene expression of CD34+ cells from normal and myelodysplastic bone marrow. Blood 100: 3553-3560, 2002 Jones P, May G, Healy L, et al: Stromal expression of Jagged 1 promotes colony formation by fetal hematopoietic progenitor cells. Blood 92: 1505-1511, 1998 Karanu FN, Murdoch B, Gallacher L, et al: The Notch ligand Jagged-1 represents a novel growth factor of human hematopoietic stem cells. J Exp Med 192: 1365-1372, 2000 Lai EC: Notch signaling: control of cell communication and cell fate. Development 131: 965-973, 2004 Langer F, Stickel J, Tessema M, et al: Overexpression of deltalike, Dlk, in a subset of myelodysplastic syndrome bone marrow trephines. Leuk Res 28: 1081-1083, 2004 Liesveld JL, Jordan CT, Philips GL: The hematopoietic stem cell in myelodysplasia. Stem Cells 22: 590-599, 2004 Miyazato A, Ueno S, Ohmine K, et al: Identification of myelodysplastic syndrome-specific genes by DNA microarray analysis with purified hematopoietic stem cell fraction. Blood 98: 422-427, 2001 Murata-Ohsawa M, Tohda S, Nara N: Cellular analysis of growth suppression induced by the Notch ligands, Delta-1 and Jagged-1 in two myeloid leukemia cell lines. Int J Mol Med 14: 223-226, 2004 Ohishi K, Katayama N, Shiku H, et al: Notch signalling in hematopoiesis. Semin Cell Dev Biol 14: 143-150, 2003 Pittenger MF, Mackay AM, Beck SC, et al: Multilineage potential of adult human mesenchymal stem cells. Science 284: 143- 147, 1999 Radtke F, Wilson A, Mancini SJC, MacDonald HR: Notch regulation of lymphocyte development and function. Nat Immunol 5: 247-253, 2004 Shetty V, Hussaini S, Alvi S, et al: Excessive apoptosis, increased phagocytosis, nuclear inclusion bodies and cylindrical confronting cisternae in bone marrow biopsies of myelodysplastic syndrome patients. Br J Haematol 116: 817-825, 2002 Steensma DP, Tefferi A: The myelodysplastic syndrome(s): a perspective and review highlighting current controversies. Leuk Res 27: 95-120, 2003 Tauro S, Hepburn MD, Bowen DT, Pippard MJ: Assessment of stromal function, and its potential contribution to deregulation of hematopoiesis in the myelodysplastic syndromes. Haematologica 86: 1038-1045, 2001 Tauro S, Hepburn MD, Peddie CM, et al: Functional disturbance of marrow stromal microenvironment in the myelodysplastic syndromes. Leukemia 16: 785-790, 2002 Tennant GB, Walsh V, Tauran LN, et al: Abnormalities of adherent layers grown from bone marrow of patients with myelodysplasia. Br J Haematol 111: 853-862, 2000 Tohda A, Nara N: Expression of Notch1 and Jagged1 proteins in acute myeloid leukemia cells. Leuk Lymphoma 42: 467-472, 2001 Tohda S, Sakano S, Ohsawa M, et al: A novel cell line derived from de novo acute myeloblastic leukaemia with trilineage myelodysplasia which proliferates in response to Notch ligand, Delta-1 protein. Br J Haematol 117: 467-472, 2002 Vardiman JW, Harris NL, Brunning RD: The World Health Organization, WHO, classification of the myeloid neoplasms. Blood 100: 2292-2302, 2002 Varnum-Finney B, Xu L, Brashem-Stein C, et al: Pluripotent, cytokine-dependent, hematopoietic stem cells are immortalized by constitutive Notch1 signaling. Nat Med 6: 1278-1281, 2000 Vas V, Szilagyi L, Paloczi K, Uher F: Soluble Jagged-1 is able to inhibit the function of its multivalent form to induce hematopoietic stem cell self-renewal in a surrogate in vitro assay. J Leuk Biol 75: 714-720, 2004 Weng AP, Aster JC: Multiple niches for Notch in cancer: context is everything. Curr Opin Gen Dev 14: 48-54, 2004 Wetzler M, Kurzrock R, Estrov Z, et al: Cytokine expression in adherent layers from patients with myelodysplastic syndrome and acute myelogenous leukemia. Leuk Res 19: 23-34, 1995 Woodbury D, Schwarz EJ, Prockop DJ, Black IB: Adult human bone marrow stromal cells differentiate into neurons. J Neurosci Res 61: 364-370, 2000 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 311 EP 319 PG 9 ER PT J AU Yuan, A Liu, J Liu, Y Cui, G AF Yuan, Aping Liu, Jinzhong Liu, Yiqing Cui, Guanglin TI Chromogranin A-Positive Tumor Cells in Human Esophageal Squamous Cell Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE esophagus; squamous cell carcinoma; neuroendocrine; differentiation ID esophagus; squamous cell carcinoma; neuroendocrine; differentiation AB Gastrointestinal cancers have frequently shown neuroendocrine (NE) differentiation, but whether NE differentiation occurs in esophageal squamous cell carcinoma (ESCC) remains unclear. In this study, tissue sections obtained from 43 patients with ESCC from a high-incidence area of Northern China were used for the assessing of NE differentiation by immunohistochemistry using antibody against chromogranin A (CGA). In addition, the malignant characteristics and proliferation capacity of CGA-positive cells were also examined by immunohistochemistry. The clinicopathological significance of these CGA-positive tumor cells in ESCC was assessed. Of 43 ESCC samples, CGAimmunoreactive tumor cells were detected in 10 cases (23.26%). However, the CGA-positive tumor cells were scattered at a very low number among non-immunoreactive tumor cells and were rarely constituted a major part of cancer cell nests. Only 4.65% (2/43) cases showed a high density (>10 cells but <1% of total tumor cell mass) of CGA-positive tumor cells. P53 immunoreactivity was frequently shown, while Ki67 was hard to detect in these CGApositive cells. In addition, no relationship between CGA positivity rate and clinicopathological parameters was found. Thus, we concluded that lowdensity CGA-positive tumor cells can be detected in ESCC, supporting the notion that heterogeneous NE differentiation also exists in tumors that lack neuroendocrine cells in their normal epithelial counterparts. C1 [Yuan, Aping] the Second Affiliated Hospital of Zhengzhou University, Department of MedicineZhengzhou, Henan, China. [Liu, Jinzhong] the Fourth Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, Henan, China. [Liu, Yiqing] the Second Affiliated Hospital of Zhengzhou University, Department of MedicineZhengzhou, Henan, China. [Cui, Guanglin] the Second Affiliated Hospital of Zhengzhou University, Department of MedicineZhengzhou, Henan, China. RP Cui, G (reprint author), the Second Affiliated Hospital of Zhengzhou University, Department of Medicine, Zhengzhou, China. EM guanglin.cui@fagmed.uit.no CR Barrett P, Hobbs RC, Coates PJ, et al: Endocrine cells of the human gastrointestinal tract have no proliferative capacity. Histochem J 27:482-486, 1995 Blumenfeld W, Chandhoke DK, Sagerman P, et al: Neuroendocrine differentiation in gastric adenocarcinomas. An immunohistochemical study. Arch Pathol Lab Med 120:478-481, 1996 Bosman FT: Neuroendocrine cells in non-endocrine tumors: what does it mean? Verh Dtsch Ges Pathol 81:62-72, 1997 Bosman FT, Louwerens JW: Neuroendocrine, APUD, cells in ovarian teratomas and testicular teratocarcinomas. Cell Biol Int Rep 5:464, 1981 Chang KT, Huang CY, Tsai CM, et al: Role of IL-6 in neuroendocrine differentiation and chemosensitivity of non-small cell lung cancer. Am J Physiol Lung Cell Mol Physiol 289:L438-445, 2005 Cho KJ, Jang JJ, Lee SS, et al: Basaloid squamous carcinoma of the oesophagus: a distinct neoplasm with multipotential differentiation. Histopathology 36:331-340, 2000 Cui G, Koh TJ, Chen D, et al: Overexpression of glycine-extended gastrin inhibits parietal cell loss and atrophy in the mouse stomach. Cancer Res 64:8160-8166, 2004 de Bruine AP, Dinjens WN, van der Linden EP, et al: Extracellular matrix components induce endocrine differentiation in vitro in NCI-H716 cells. Am J Pathol 142:773-782, 1993 Hamilton K, Chiappori A, Olson S, et al: Prevalence and prognostic significance of neuroendocrine cells in esophageal adenocarcinoma. Mod Pathol 13:475-481, 2000 Hayashi H, Ooi A, Nakanishi I: Biological and clinicopathological significance of endocrine differentiation of gastric adenocarcinoma evaluated by double immunohistochemical labeling for chromogranin A and bromodeoxyuridine. Jpn J Clin Oncol 20:339-348, 1990 Jaskiewicz K, Louw J, Anichkov N: Barrett's oesophagus: mucin composition, neuroendocrine cells, p53 protein, cellular proliferation and differentiation. Anticancer Res 14:1907-1912, 1994 Li MX, Cheng SJ: Carcinogenesis of esophageal cancer in Linxian, China. Chin Med J, Engl, 97:311-313, 1984 Li Z, Shimada Y, Uchida S, et al: TGF-alpha as well as VEGF, PDECGF and bFGF contribute to angiogenesis of esophageal squamous cell carcinoma. Int J Oncol 17:453-460, 2000 Linder S, Myrvold K, Falkmer UG, et al: Neuroendocrine cells in pancreatic duct adenocarcinoma: an immunohistochemical study. J Exp Clin Cancer Res 25:213-221, 2006 Louwerens JK, Schaberg A, Bosman FT: Neuroendocrine cells in cystic mucinous tumours of the ovary. Histopathology 7:389-398, 1983 Lu SH, Zhou Y, Que HP, et al: Peptidergic innervation of human esophageal and cardiac carcinoma. World J Gastroenterol 9:399- 403, 2003 Ohashi K, Nemoto T, Nakamura K, et al: Increased expression of matrix metalloproteinase 7 and 9 and membrane type 1-matrix metalloproteinase in esophageal squamous cell carcinomas. Cancer 88:2201-2209, 2000 Ooi A, Hayashi H, Katsuda S, et al: Gastric carcinoma cells with endocrine differentiation show no evidence of proliferation. Hum Pathol 23:736-741, 1992 Ooi A, Mai M, Ogino T, et al: Endocrine differentiation of gastric adenocarcinoma. The prevalence as evaluated by immunoreactive chromogranin A and its biologic significance. Cancer 62:1096- 1104, 1988 Parkin DM, Bray FI, Devesa SS: Cancer burden in the year 2000. The global picture. Eur J Cancer 37 Suppl 8):S4-S66, 2001 Rengifo-Cam W, Singh P: Role of progastrins and gastrins and their receptors in GI and pancreatic cancers: targets for treatment. Curr Pharm Des 10:2345-2358, 2004 Seifert P, Spitznas M: Tumours may be innervated. Virchows Arch 438:228, 2001 Syversen U, Halvorsen T, Marvik R, et al: Neuroendocrine differentiation in colorectal carcinomas. Eur J Gastroenterol Hepatol 7:667-674, 1995 Szumilo J, Swatek J, Chroscicki A, et al: Colonic adenocarcinoma with numerous paneth and endocrine cells. Pol J Pathol 56:89-92, 2005 Tajima Y, Nakajima T, Sugano I, et al: Hepatocellular carcinoma containing endocrine cells. An autopsy report of triple cancer involving the liver, kidney and thyroid. Acta Pathol Jpn 42:904- 910, 1992 Tezel E, Nagasaka T, Tezel G, et al: Characterization of scattered neuroendocrine cells in ductal carcinoma of the pancreas. Pancreas 25:136-141, 2002 Untergasser G, Plas E, Pfister G, et al: Interferon-gamma induces neuroendocrine-like differentiation of human prostate basal-epithelial cells. Prostate 64:419-429, 2005 Waldum HL, Brenna E: Non-proliferative capacity of endocrine cells of the human gastro-intestinal tract. Histochem J 28:397-398, 1996 Wang JH, Dhillon AP, Sankey EA, et al: 'Neuroendocrine' differentiation in primary neoplasms of the liver. J Pathol 163:61-67, 1991 Wang KL, Yang Q, Cleary KR, et al: The significance of neuroendocrine differentiation in adenocarcinoma of the esophagus and esophagogastric junction after preoperative chemoradiation. Cancer 107:1467-1474, 2006 Wang LS, Chow KC, Wu CW: Expression and up-regulation of interleukin- 6 in oesophageal carcinoma cells by n-sodium butyrate. Br J Cancer 80:1617-1622, 1999 Zhao M, Laissue JA, Zimmermann A: "Neuroendocrine" differentiation in hepatocellular carcinomas, HCCs): immunohistochemical reactivity is related to distinct tumor cell types, but not to tumor grade. Histol Histopathol 8:617-626, 1993 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 321 EP 325 PG 5 ER PT J AU Chang, JH Yoo, ChB Kim, WS Lee, LB Kim, HW AF Chang, Jin Hee Yoo, Chul Byong Kim, Whe Sun Lee, Lan Byung Kim, Ho Woo TI Significance of PML and p53 Protein as Molecular Prognostic Markers of Gallbladder Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gallbladder carcinoma; prognosis; PML protein; p53 protein ID gallbladder carcinoma; prognosis; PML protein; p53 protein AB Molecular markers for cancers are not only useful for cancer detection and prognostic prediction, but may also serve as potential therapeutic targets. In order to identify reliable molecular markers for prognostic prediction in gallbladder carcinoma (GBC), we evaluated the immunohistochemical expression of 15 proteins, namely p53, p27, p16, RB, Smad4, PTEN, FHIT, GSTP1, MGMT, E-cadherin, nm23, CD44, TIMP3, S100A4, and promyelocytic leukemia (PML) in 138 cases of GBC using the tissue microarray method. The prognostic significance was analyzed for each protein. Overexpression of p53 and S100A4, and loss of p27, p16, RB, Smad4, FHIT, E-cadherin and PML expression were associated with poor survival. In particular, PML and p53 showed considerable potential as independent prognostic markers. Patients with normal PML and p53 expression displayed favorable outcomes, compared to those showing abnormal expression of either or both proteins (49% vs. 23% in a 5-year survival rate; 60 months vs. 11 months in median survival, respectively; P=0.009). Thus, PML and p53 are potential candidates for development as clinically applicable molecular prognostic markers of GBC, and may be effective therapeutic targets for the disease in the future. C1 [Chang, Jin Hee] National Cancer Center, Research Institute and HospitalSeoul, South Korea. [Yoo, Chul Byong] National Cancer Center, Research Institute and HospitalSeoul, South Korea. [Kim, Whe Sun] Seoul National University, Department of SurgerySeoul, South Korea. [Lee, Lan Byung] Seoul National University, Department of AnatomySeoul, South Korea. [Kim, Ho Woo] Seoul National University, Cancer Research Institute, 28 Yeongeon-dong, Jongno-gu, 110-799 Seoul, South Korea. RP Kim, HW (reprint author), Seoul National University, Cancer Research Institute, 110-799 Seoul, South Korea. EM woohokim@snu.ac.kr CR Ajiki T, Fujimori T, Onoyama H, Yamamoto M, Kitazawa S, Maeda S, Saitoh Y: K-ras gene mutation in gall bladder carcinomas and dysplasia. Gut 38:426-429, 1996 Ajiki T, Onoyama H, Yamamoto M, Asaka K, Fujimori T, Maeda S, Saitoh Y: p53 protein expression and prognosis in gallbladder carcinoma and premalignant lesions. Hepatogastroenterology 43:521-526, 1996 Albores-Saavedra J. Scoazec JC, Wittekind C, Sripa B, Menck HR, Soehendra N, Sriram PVJ, ed): Carcinoma of the gallbladder and extrahepatic bile ducts. In: Hamilton SR, Aaltonen LA, ed, WHO Classification of Tumors; Pathology and Genetics of Tumors of the Digestive System. IARC Press, Lyon, 2000, pp 203-217 Alkushi A, Lim P, Coldman A, Huntsman D, Miller D, Gilks CB: Interpretation of p53 immunoreactivity in endometrial carcinoma: establishing a clinically relevant cut-off level. Int J Gynecol Pathol 23:129-137, 2004 Chang HJ, Kim SW, Kim YT, Kim WH: Loss of heterozygosity in dysplasia and carcinoma of the gallbladder. Mod Pathol 12:763-769, 1999 Chang HJ, Jee CD, Kim WH: Mutation and altered expression of beta-catenin during gallbladder carcinogenesis. Am J Surg Pathol 26:758-766, 2002 Cubertafond P, Gainant A, Cucchiaro G: Surgical treatment of 724 carcinomas of the gallbladder. Results of the French Surgical Association Survey. Ann Surg 219:275-280, 1994 da Rocha AO, Coutinho LM, Scholl JG, Leboutte LD: The value of p53 protein expression in gallbladder carcinoma: analysis of 60 cases. Hepatogastroenterology 51:1310-1314, 2004 El-Deiry WS: The role of p53 in chemosensitivity and radiosensitivity. Oncogene 22:7486-7495, 2003 Fleming ID, Cooper JS, Henson DE, Hutter RVP, Kennedy BJ, Murphy GP, O’Sullivan B, Sobin LH, Yarbro JW, ed): AJCC cancer staging manual. 5th ed. JB Lippincott, Philadelphia, 1997 Galizia G, Ferraraccio F, Lieto E, Orditura M, Castellano P, Imperatore V, Romano C, Vollaro M, Agostini B, Pignatelli C, De Vita F: Prognostic value of p27, p53, and vascular endothelial growth factor in Dukes A and B colon cancer patients undergoing potentially curative surgery. Dis Colon Rectum 47:1904-1914, 2004 Gurrieri C, Capodieci P, Bernardi R, Scaglioni PP, Nafa K, Rush LJ, Verbel DA, Cordon-Cardo C, Pandolfi PP: Loss of the tumor suppressor PML in human cancers of multiple histologic origins. J Natl Cancer Inst 96:269-279, 2004 House MG, Wistuba II, Argani P, Guo M, Schulick RD, Hruban RH, Herman JG, Maitra A: Progression of gene hypermethylation in gallstone disease leading to gallbladder cancer. Ann Surg Oncol 10:882-889, 2003 Hui AM, Li X, Shi YZ, Torzilli G, Takayama T, Makuuchi M: p27(KIP1, expression in normal epithelia, precancerous lesions, and carcinomas of the gallbladder: association with cancer progression and prognosis. Hepatology 31:1068-1072, 2000 Hui AM, Li X, Shi YZ, Takayama T, Torzilli G, Makuuchi M: Cyclin D1 overexpression is a critical event in gallbladder carcinogenesis and independently predicts decreased survival for patients with gallbladder carcinoma. Clin Cancer Res 6:4272- 4277, 2000 Ito H, Matros E, Brooks DC, Osteen RT, Zinner MJ, Swanson RS, Ashley SW, Whang EE: Treatment outcomes associated with surgery for gallbladder cancer: a 20-year experience. J Gastrointest Surg 8:183-190, 2004 Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, Feuer EJ, Thun MJ Cancer statistics. CA Cancer J Clin 55:10- 30, 2005 Kawamoto T, Shoda J, Irimura T, Miyahara N, Furukawa M, Ueda T, Asano T, Kano M, Koike N, Fukao K, Tanaka N, Todoroki T: Expression of MUC1 mucins in the subserosal layer correlates with postsurgical prognosis of pathological tumor stage 2 carcinoma of the gallbladder. Clin Cancer Res 7:1333-1342, 2001 Kim YW, Huh SH, Park YK, Yoon TY, Lee SM, Hong SH: Expression of the c-erb-B2 and p53 protein in gallbladder carcinomas. Oncol Rep 8:1127-1132, 2001 Kohya N, Miyazaki K, Matsukura S, Yakushiji H, Kitajima Y, Kitahara K, Fukuhara M, Nakabeppu Y, Sekiguchi M: Deficient expression of O(6)-methylguanine-DNA methyltransferase combined with mismatch-repair proteins hMLH1 and hMSH2 is related to poor prognosis in human biliary tract carcinoma. Ann Surg Oncol 9:371-379, 2002 Kwon SY, Chang HJ: Clinicopathological study of unsuspected carcinoma of gallbladder. J Korean Med Science 12: 519-522, 1997 Lee HS, Lee HK, Kim HS, Yang HK, Kim WH: Tumour suppressor gene expression correlates with gastric cancer prognosis. J Pathol 200:39-46, 2003 Levy AD, Murakata LA, Rohrmann CA: Gallbladder carcinoma: radiologic-pathologic correlation. Radiographics 21:295-314, 2001 Li X, Hui AM, Shi YZ, Takayama T, Makuuchi M: Reduced p21(WAF1/CIP1, expression is an early event in gallbladder carcinogenesis and is of prognostic significance for patients with carcinomas of the gallbladder. Hum Pathol 32:771-777, 2001 Misra S, Chaturvedi A, Goel MM, Mehrotra R, Sharma ID, Srivastava AN, Misra NC: Overexpression of p53 protein in gallbladder carcinoma in North India. Eur J Surg Oncol 26:164- 167, 2000 Nakamura T, Ajiki T, Murao S, Kamigaki T, Maeda S, Ku Y, Kuroda Y: Prognostic significance of S100A4 expression in gallbladder cancer. Int J Oncol 20:937-941, 2002 Oshikiri T, Hida Y, Miyamoto M, Hashida H, Katoh K, Suzuoki M, Nakakubo Y, Hiraoka K, Shinohara T, Itoh T, Kondo S, Katoh H: RCAS1 as a tumour progression marker: an independent negative prognostic factor in gallbladder cancer. Br J Cancer 85:1922-1927, 2001 Rego EM, Wang ZG, Peruzzi D, He LZ, Cordon-Cardo C, Pandolfi PP: Role of promyelocytic leukemia, PML, protein in tumor suppression. J Exp Med 193:521-529, 2001 Rosen DG, Huang X, Deavers MT, Malpica A, Silva EG, Liu J: Validation of tissue microarray technology in ovarian carcinoma. Mod Pathol 17:790-797, 2004 Shi YZ, Hui AM, Li X, Takayama T, Makuuchi M: Overexpression of retinoblastoma protein predicts decreased survival and correlates with loss of p16INK4 protein in gallbladder carcinomas. Clin Cancer Res 6:4096-4100, 2000 Soussi T, Caron de Fromentel C, May P: Structural aspects of the p53 protein in relation to gene evolution. Oncogene 5:945- 952, 1990 Takahashi T, Shivapurkar N, Riquelme E, Shigematsu H, Reddy J, Suzuki M, Miyajima K, Zhou X, Bekele BN, Gazdar AF, Wistuba II: Aberrant promoter hypermethylation of multiple genes in gallbladder carcinoma and chronic cholecystitis. Clin Cancer Res 10:6126-6133, 2004 Torhorst J, Bucher C, Kononen J, Haas P, Zuber M, Kochli OR, Mross F, Dieterich H, Moch H, Mihatsch M, Kallioniemi OP, Sauter G: Tissue microarrays for rapid linking of molecular changes to clinical endpoints. Am J Pathol 159:2249-2256, 2001 Varga M, Obrist P, Schneeberger S, Muhlmann G, Felgel- Farnholz C, Fong D, Zitt M, Brunhuber T, Schafer G, Gastl G, Spizzo G: Overexpression of epithelial cell adhesion molecule antigen in gallbladder carcinoma is an independent marker for poor survival. Clin Cancer Res 10:3131-3136, 2004 Wistuba II, Sugio K, Hung J, Kishimoto Y, Virmani AK, Roa I, Albores-Saavedra J, Gazdar AF: Allele-specific mutations involved in the pathogenesis of endemic gallbladder carcinoma in Chile. Cancer Res 55:2511-2515, 1995 Wistuba II, Tang M, Maitra A, Alvarez H, Troncoso P, Pimentel F, Gazdar AF: Genome-wide allelotyping analysis reveals multiple sites of allelic loss in gallbladder carcinoma. Cancer Res 61:3795-3800, 2001 Wistuba II, Ashfaq R, Maitra A, Alvarez H, Riquelme E, Gazdar AF: Fragile histidine triad gene abnormalities in the pathogenesis of gallbladder carcinoma. Am J Pathol 160:2073-2079, 2002 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 326 EP 335 PG 10 ER PT J AU Croci, S Landuzzi, L Nicoletti, G Palladini, A Antognoli, A De Giovanni, C Nanni, P Lollini, PL AF Croci, Stefania Landuzzi, Lorena Nicoletti, Giordano Palladini, Arianna Antognoli, Agnese De Giovanni, Carla Nanni, Patrizia Lollini, Pier-Luigi TI Expression of Connective Tissue Growth Factor (CTGF/CCN2) in a Mouse Model of Rhabdomyosarcomagenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE rhabdomyosarcoma; mouse model; CCN proteins; CCN2/CTGF ID rhabdomyosarcoma; mouse model; CCN proteins; CCN2/CTGF AB Connective tissue growth factor (CTGF/CCN2) is a cysteine-rich matricellular protein that belongs to the CCN (CYR61, CTGF, NOV) protein family. It is highly expressed by human rhabdomyosarcoma cells and sustains their survival. In this study we investigated CCN2 expression in a mouse model of spontaneous rhabdomyosarcomagenesis that combines HER-2/neu oncogene activation and p53 oncosuppressor gene inactivation (BALB-p53neu mice). Murine rhabdomyosarcoma cells showed a 4-26 fold increase in CCN2 mRNA expression regarding to normal thigh muscle. Moreover, they expressed CCN2 protein at levels comparable to human rhabdomyosarcoma cells. Therefore BALBp53neu mice might be useful for the evaluation of the role played by CCN2 in rhabdomyosarcoma in vivo. C1 [Croci, Stefania] University of Bologna, Department of Experimental Pathology, Cancer Research Section, Viale Filopanti 22, I-40126 Bologna, Italy. [Landuzzi, Lorena] Istituti Ortopedici Rizzoli, Laboratorio di Ricerca OncologicaBologna, Italy. [Nicoletti, Giordano] Istituti Ortopedici Rizzoli, Laboratorio di Ricerca OncologicaBologna, Italy. [Palladini, Arianna] University of Bologna, Department of Experimental Pathology, Cancer Research Section, Viale Filopanti 22, I-40126 Bologna, Italy. [Antognoli, Agnese] University of Bologna, Department of Experimental Pathology, Cancer Research Section, Viale Filopanti 22, I-40126 Bologna, Italy. [De Giovanni, Carla] University of Bologna, Department of Experimental Pathology, Cancer Research Section, Viale Filopanti 22, I-40126 Bologna, Italy. [Nanni, Patrizia] University of Bologna, Department of Experimental Pathology, Cancer Research Section, Viale Filopanti 22, I-40126 Bologna, Italy. [Lollini, Pier-Luigi] University of Bologna, Department of Experimental Pathology, Cancer Research Section, Viale Filopanti 22, I-40126 Bologna, Italy. RP Lollini, PL (reprint author), University of Bologna, Department of Experimental Pathology, Cancer Research Section, I-40126 Bologna, Italy. EM pierluigi.lollini@unibo.it CR Aikawa T, Gunn J, Spong SM et al: Connective tissue growth factor-specific antibody attenuates tumor growth, metastasis, and angiogenesis in an orthotopic mouse model of pancreatic cancer. Mol Cancer Ther 5: 1108-1116, 2006 Brigstock DR: The CCN family: a new stimulus package. J Endocrinol 178: 169-175, 2003 Croci S, Landuzzi L, Astolfi A et al: Inhibition of connective tissue growth factor, CTGF/CCN2, expression decreases the survival and myogenic differentiation of human rhabdomyosarcoma cells. Cancer Res 64: 1730-1736, 2004 Dornhofer N, Spong S, Bennewith K et al: Connective tissue growth factor-specific monoclonal antibody therapy inhibits pancreatic tumor growth and metastasis. Cancer Res 66: 5816- 5827, 2006 Lin BR, Chang CC, Che TF et al: Connective tissue growth factor inhibits metastasis and acts as an independent prognostic marker in colorectal cancer. Gastroenterology 128: 9-23, 2005 Nanni P, Nicoletti G, De Giovanni C et al: Development of rhabdomyosarcoma in HER-2/neu transgenic p53 mutant mice. Cancer Res 63: 2728-2732, 2003 Palladini A, Astolfi A, Croci S et al: Endothelin-3 production by human rhabdomyosarcoma: a possible new marker with a paracrine role. Eur J Cancer 42: 680-687, 2006 Perbal B: CCN proteins: multifunctional signalling regulators. Lancet 363: 62-64, 2004 Planque N, Perbal B: A structural approach to the role of CCN, CYR61/CTGF/NOV, proteins in tumourigenesis. Cancer Cell Int 3: 15, 2003 Shimo T, Kubota S, Yoshioka N et al: Pathogenic role of connective tissue growth factor, CTGF/CCN2, in osteolytic metastasis of breast cancer. J Bone Miner Res 21: 1045-1059, 2006 Yang DH, Kim HS, Wilson EM et al: Identification of glycosylated 38-kDa connective tissue growth factor, IGFBP-related protein 2, and proteolytic fragments in human biological fluids, and up-regulation of IGFBP-rP2 expression by TGF-beta in Hs578T human breast cancer cells. J Clin Endocrinol Metab 83: 2593-2596, 1998 Yang F, Tuxhorn JA, Ressler SJ et al: Stromal expression of connective tissue growth factor promotes angiogenesis and prostate cancer tumorigenesis. Cancer Res 65: 8887-8895, 2005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 336 EP 339 PG 4 ER PT J AU Remenar, Szamel, I Budai, B Vincze, B Gaudi, I Gundy, S Kasler, M AF Remenar, Eva Szamel, Iren Budai, Barna Vincze, Borbala Gaudi, Istvan Gundy, Sarolta Kasler, Miklos TI Increase of Hypophyseal Hormone Levels in Male Head and Neck Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE head and neck cancer; sex steroids; hypophysis hormones; alcoholic liver disease ID head and neck cancer; sex steroids; hypophysis hormones; alcoholic liver disease AB Head and neck squamous cell carcinoma (HNSCC) develops in at least 80% of cases in men with a history of smoking and heavy alcohol consumption, still it is only diagnosed in a small proportion of alcoholics. Endocrine milieu is an important factor in carcinogenesis and prognosis of several cancer types. The aim of our study was to investigate sex steroid and hypophyseal hormone status of male HNSCC patients in comparison to healthy volunteers and to patients with alcoholic liver disease, to determine possible hormonal alterations characteristic of cancer. Liver function (GGT level), and serum levels of gonadotropic hormones (FSH, LH, prolactin), sex steroids (estradiol, progesterone, testosterone) and sex hormone-binding globulin (SHBG) were compared in 130 male HNSCC patients, 54 patients with alcoholic liver disease but no known cancer, and 56 healthy controls. We found abnormal values of liver function in both HNSCC patients and alcoholics compared to healthy controls, suggesting the presence of alcoholic liver disease in the former group as well. On the other hand, a significant elevation in the level of DHEA, FSH and LH was observed in cancer patients exclusively. As a conclusion, abnormal alterations in sex steroid hormone levels can frequently be found in HNSCC patients, which may be caused in part by the alcoholic liver damage accompanying the disease. The significant increase in FSH and LH serum levels, observed only in the cancer patients, indicates that these hormones may play a role in the development and/or progression of HNSCC. C1 [Remenar, Eva] National Institute of Oncology, Department of Head and Neck Surgery, Rath Gy. u. 7-9., H-1122 Budapest, Hungary. [Szamel, Iren] National Institute of Oncology, Department of Chemotherapy and ClinicopharmacologyBudapest, Hungary. [Budai, Barna] National Institute of Oncology, Central Clinical LaboratoryBudapest, Hungary. [Vincze, Borbala] National Institute of Oncology, Department of BiochemistryBudapest, Hungary. [Gaudi, Istvan] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Gundy, Sarolta] National Institute of Oncology, National Cancer RegistryBudapest, Hungary. [Kasler, Miklos] National Institute of Oncology, Department of Head and Neck Surgery, Rath Gy. u. 7-9., H-1122 Budapest, Hungary. RP Remenar, (reprint author), National Institute of Oncology, Department of Head and Neck Surgery, H-1122 Budapest, Hungary. EM reva@oncol.hu CR Tsugane S, Fahey MT, Sasaki S, Baba S: Alcohol consumption and all-cause and cancer mortality among middle-aged Japanese men: seven-year follow-up of the JPHC study cohort I. Japan Public Health Center. Am J Epidemiol 150: 1201-1207, 1999 Bray I, Brennan P, Boffetta P: Projections of alcohol- and tobacco-related cancer mortality in Central Europe. Int J Cancer 87: 122-128, 2000 Boffetta P, Ye W, Adami HO, Mucci LA, Nyren O: Risk of cancers of the lung, head and neck in patients hospitalized for alcoholism in Sweden. Br J Cancer 85: 678-682, 2001 World Health Organization: The tobacco atlas. Geneva; World Health Organization, 2002 Zeka A, Gore R, Kriebel D: Effects of alcohol and tobacco on aerodigestive cancer risks: a meta-regression analysis. Cancer Causes Control 14: 897-906, 2003 World Health Organization: Global status report on alcohol. Geneva; World Health Organization, 2004 Ezzati M, Henley SJ, Lopez AD, Thun MJ: Role of smoking in global and regional cancer epidemiology: Current patterns and data needs. Int J Cancer 116: 963-971, 2005 Boffetta P, Hashibe M: Alcohol and cancer. Lancet Oncol 7: 149-156, 2006 Bannister P, Losowsky MS: Sex hormones and chronic liver disease. J Hepatol 6: 258-262, 1988 Wang YJ, Wu JC, Lee SD, Tsai YT, Lo KJ: Gonadal dysfunction and changes in sex hormones in postnecrotic cirrhotic men: a matched study with alcoholic cirrhotic men. Hepatogastroenterology 38: 531-534, 1991 Martinez-Riera A, Santolaria-Fernandez F, Gonzalez Reimers E, Milena A, Gomez-Sirvent JL, Rodriguez-Moreno F, Gonzalez- Martin I, Rodriguez-Rodriguez E: Alcoholic hypogonadism: hormonal response to clomiphene. Alcohol 12: 581-587, 1995 Zietz B, Lock G, Plach B, Drobnik W, Grossmann J, Scholmerich J, Straub RH: Dysfunction of the hypothalamicpituitary- glandular axes and relation to Child-Pugh classification in male patients with alcoholic and virus-related cirrhosis. Eur J Gastroenterol Hepatol 15: 495-501, 2003 Patel JD, Bach PB, Kris MG: Lung cancer in US women. A contemporary epidemic. JAMA 291: 1763-1768, 2004 Yoo HJ, Sepkovic DW, Bradlow HL, Yu GP, Sirilian HV, Schantz SP: Estrogen metabolism as a risk factor for head and neck cancer. Otolaryngol Head Neck Surg 124: 241-247, 2001 Kawai H, Ishii A, Washiya K, Konno T, Kon H, Yamaya C, Ono I, Minamiya Y, Ogawa J: Estrogen receptor a and b are prognostic factors in non-small cell lung cancer. Clin Cancer Res 11: 5084-5089, 2005 Chan JA, Meyerhardt JA, Chan AT, Giovanucci EL, Colditz GA, Fuchs CS: Hormone replacement therapy and survival after colorectal cancer diagnosis. J Clin Oncol 24: 5680-5686, 2006 Giannitrapani L, Soresi M, La Spada E, Cervello M, D’Alessandro N, Montalto G: Sex hormones and risk of liver tumor. Ann N Y Acad Sci 1089: 228-236, 2006 Poynard T, Aubert A, Bedossa P, Abella A, Naveau S, Paraf F, Chaput JC: A simple biological index for detection of alcoholic liver disease in drinkers. Gastroenterology 100: 1397-1402, 1991 Yokoyama A, Muramatsu T, Ohmori T, Yokoyama T, Okuyama K, Takahashi H, Hasegawa Y, Higuchi S, Maruyama K, Shirakura K, Ishii H: Alcohol-related cancers and aldehyde dehydrogenase- 2 in Japanese alcoholics. Carcinogenesis 19: 1383-1387, 1998 Makimoto K, Higuchi S: Alcohol consumption as a major risk factor for the rise in liver cancer mortality rates in Japanese men. Int J Epidemiol 28: 30-34, 1999 Duvoux C, Delacroix I, Richardet J-P, Roudot-Thoraval F, Metreau JM, Faqniez PL, Dhumeaux D, Cherqui D: Increased incidence of oropharyngeal squamous cell carcinomas after liver transplantation for alcoholic cirrhosis. Transplantation 67: 418-421, 1999 Kopp MS, Rethelyi JC: Where psychology meets physiology: chronic stress and premature mortality - the Central-Eastern European health paradox. Brain Res Bull 62: 351-367, 2004 Lukits J, Remenar E, Raso E, Ladanyi A, Kasler M, Timar J: Molecular identification, expression and prognostic role of estrogen- and progesterone receptors in head and neck cancer. Int J Oncol 30:155-160, 2007 Chen SS, Chen KK, Lin AT, Chang YH, Wu HH, Chang LS: The correlation between pretreatment serum hormone levels and treatment outcome for patients with prostatic cancer and bony metastases. BJU Int 89: 710-713, 2002 Bhatavdekar JM, Patel DD, Vora HH, Balar DB: Circulating markers and growth factors as prognosticators in men with advanced tongue cancer. Tumour Biol 14: 55-58, 1993 Bhatavdekar JM, Patel DD, Chikhlikar PR, Mehta RH, Vora HH, Karelia NH, Ghosh N, Shah NG, Suthar TP, Neema JP, et al: Levels of circulating peptide and steroid hormones in men with lung cancer. Neoplasma 41: 101-103, 1994 Bhatavdekar JM, Patel DD, Vora HH, Shah NG, Chikhlikar PR, Ghosh N: Prolactin as a local growth promoter in patients with locally advanced tongue cancer: GCRI experience. Head Neck 22: 257-264, 2000 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 341 EP 344 PG 4 ER PT J AU Kojima, M Nakamura, N Shimizu, K Nishikawa, M Matsumoto, M Higuchi, K Yamane, N Tsukamoto, N Tamaki, Y Inagaki, H AF Kojima, Masaru Nakamura, Naoya Shimizu, Kazuhiko Nishikawa, Masahumi Matsumoto, Morio Higuchi, Keiko Yamane, Nobuo Tsukamoto, Norihumi Tamaki, Yoshio Inagaki, Hiroshi TI Histopathological Variation of Primary Mucosa-associated Lymphoid Tissue Lymphoma of the Oral Cavity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mucosa-associated lymphoid tissue lymphoma; oral cavity; follicular colonization; immunohistochemistry ID mucosa-associated lymphoid tissue lymphoma; oral cavity; follicular colonization; immunohistochemistry AB Primary mucosa-associated lymphoid tissue (MALT) type lymphoma arising in the oral cavity is rare. We examined histopathologic, immunohistological and genotypic findings of seven cases of intraoral MALT lymphoma using formalin-fixed paraffin-embedded tissues. Histologically, two variants have been delineated. (i) In four cases of minor salivary gland type, the lymphoid follicles were surrounded by centrocyte-like (CCL) cells with occasional follicular colonization. The CCL cells invaded the residual salivary gland duct resulting in a lymphoepithelial lesion. CCL cells frequently showed plasmacytic differentiation. (ii) In three cases of follicular growth type, the lesion was characterized by follicular growth pattern resulting from prominent follicular colonization. CCL cells showed minimal plasma cell differentiation. There was no residual epithelial component detected even by cytokeratin immunostaining. There were no Epstein-Barr virus-encoded small RNA-positive cells detected by in situ hybridization. API2-MALT1 fusion transcript does not appear to be associated with either histological variant of primary intraoral MALT lymphoma. C1 [Kojima, Masaru] Gunma Cancer Center Hospital, Department of Pathology and Clinical Laboratories, 617-1, Takabayashinishi-cho, 373-8550 Ohta, Japan. [Nakamura, Naoya] Tokai University School of Medicine, Department of PathologyIsehara, Japan. [Shimizu, Kazuhiko] Ashikaga Red Cross Hospital, Department of Pathology and Clinical LaboratoriesAshikaga, Japan. [Nishikawa, Masahumi] National Tochigi Hospital, Department of Pathology and Clinical LaboratoriesUtsunomiya, Japan. [Matsumoto, Morio] National Nishigunma Hospital, Department of HematologyShibukawa, Japan. [Higuchi, Keiko] Gunma Cancer Center Hospital, Department of RadiologyOhta, Japan. [Yamane, Nobuo] Ashikaga Red Cross Hospital, Department of Oral SurgeryAshikaga, Japan. [Tsukamoto, Norihumi] Gunma University, Department of Medicine and Clinical ScienceGunma, Japan. [Tamaki, Yoshio] Gunma Cancer Center Hospital, Department of RadiologyOhta, Japan. [Inagaki, Hiroshi] Nagoya City University Graduate School of Medical Science, Department of PathologyNagoya, Japan. RP Kojima, M (reprint author), Gunma Cancer Center Hospital, Department of Pathology and Clinical Laboratories, 373-8550 Ohta, Japan. EM mkojima@gunma-cc.jp CR Epstein JB, Epstein JD, Le ND, Gorsky M: Characterization of oral and paraoral malignant lymphoma: a population-based review of 361 cases. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 92:519-525, 2001 Takahashi H, Fujita S, Okabe H, Tsuda N, Tezuka F: Immunophenotypic analysis of extranodal non-Hodgkin’s lymphomas in oral cavity. Pathol Res Pract 189:300-311, 1993 Solomides CC, Miller AS, Christman RA, Talwar J, Simpkins H. Lymphomas of the oral cavity: Histology, immunologic type, and incidence of Epstein-Barr virus infection. Hum Pathol 33:153-157, 2002 Odell EW, Lombardi T, Shirlaw PJ, White CAM: Minor salivary gland hyalinization and amyloidosis in low-grade lymphoma of MALT. J Oral Pathol Med 27:229-232, 1998 Sakabe H, Bamba M, Nomura K, Kitamura S, Segawa H, Yasui H, Inoue T, Taniwaki M, Fujiyama Y, Bamba T: MALT lymphoma at the base of the tongue developing without any background of immunodeficiency or autoimmune disease. Leuk Lymphoma 44:875-878, 2003 Honda K, Kusama H, Takagi S, Sekine S, Noguchi M, Chiba H: Diagnosis of intra-oral MALT lymphoma using seminested polymerase chain reaction. Br J Oral Maxillofac Surg 42:28- 32, 2004 Sakuma H, Okabe M, Yokoi M, Eimoto T, Inagaki H: Spontaneous regression of intraoral mucosa-associated lymphoid tissue lymphoma: molecular study of a case. Pathol Intern 56:331-335, 2006 Kojima M, Sugihara S, Iijima M, Ono T, Yoshizumi T, Masawa N: Marginal zone B-cell lymphoma of minor salivary gland representing tumor-forming amyloidosis of the oral cavity. A case report. J Oral Pathol Med 35:314-316, 2006 Inagaki H, Chan JKC, Ng JWM, Okabe M, Yoshino T, Okamoto M, Ogawa H, Matsushita H, Yokose T, Matsuno Y, Nakamura N, Nagasaka T, Ueda R, Eimoto T, Nakamura S: Primary thymic extranodal marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue type exhibits distinctive clinicopathological and molecular features. Am J Pathol 160:1435-1443, 2002 Isaacson PG, Norton AJ: Extranodal lymphomas. Churchill Livingstone, Edinburgh, 1994 Chan ALC, Chan JKC. Haematolymphoid tumors. In: Pathology and genetics of tumours of head and neck., Eds: Barnes L, Evenson JW, Reichart P, Sidransky D), IARCPress, Lyon, 2005, pp 199-205 Isaacson PG, Wotherspoon AC, Diss T, Pan L: Follicular colonization of B-cell lymphoma of mucosa-associated lymphoid tissue. Am J Surg Pathol 15:819-828, 1991 Kojima M, Yamanaka S, Yoshida T, Shimizu K, Murayama K, Ohno Y, Itoh H, Motoori T, Masawa N, Nakamura S: Histological variety of floral variant of follicular lymphoma. A report of 13 Japanese cases. APMIS 114:626-632, 2006 Lai R, Weiss LM, Chang KL, Arber DA: Frequency of CD43 expression in non-Hodgkin lymphoma. A survey of 742 cases and further characterization of rare CD43+ follicular lymphoma. Am J Clin Pathol 111:488-494, 1999 Wang T, Lasota J, Hanau CA, Miettinen M: Bcl-2 oncoprotein is widespread in lymphoid tissue and lymphoma but its differential expression in benign versus malignant follicles and monocytoid B-cell proliferation is of diagnostic value. APMIS 103: 655-662, 1995 Isaacson PG, Du M-Q: Gastrointestinal lymphoma: where morphology meets molecular biology. J Pathol 205:255-274, 2005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 345 EP 349 PG 5 ER PT J AU Magyar, Z Berkes, E Csapo, Zs Papp, Z AF Magyar, Zoltan Berkes, Eniko Csapo, Zsolt Papp, Zoltan TI Effect of Hormone Replacement Therapy on Postmenopausal Endometrial Bleeding SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE climacterium; postmenopausal hormone therapy; menopause; irregular bleeding ID climacterium; postmenopausal hormone therapy; menopause; irregular bleeding AB The aim of the study was to determine the effect of postmenopausal hormone replacement therapy (HRT) (treatment using estrogen only and sequential and continuous combined estrogen-progestogen treatment) on endometrial bleeding and histological changes of the endometrium. In a six-year period (2000-2005), 5893 patients were given care and the incidence of postmenopausal uterine bleeding was detected in groups of patients having and not having received hormonal treatment at the Menopause Outpatient Unit of the authors’ department. In the case of bleeding, fractioned abrasion was performed and the samples were analyzed histologically. Among the postmenopausal patients who had not been given hormonal treatment, the incidence of bleeding episodes was significantly higher as among those having received hormonal treatment. In the samples, findings of proliferative endometrium occurred significantly more often in case of non-treated patients and those treated with sequential combined hormone therapy compared to patients receiving continuous combined hormone therapy. Although it was statistically not significant, hyperplasia simplex and complex together showed a tendency of reduced incidence in patients medicated by continuous combined treatment. These findings suggest that continuous combined hormonal treatment started at the right time (even before the menopause) may reduce the chances of the development of hyperplasia. A significantly higher incidence of hyperplasia was noted in patients using estrogen treatment only. It is possible that unopposed estrogen treatment further engraves an already diagnosed endometrial hyperplasia. In the group having received hormonal treatment, no complex hyperplasia accompanied by atypia occurred, only hyperplasia simplex was diagnosed in these cases. As a result of continuous reliance on combined preparations, the endometrium had become atrophied, therefore the chance of hyperplasia-related changes and of bleeding as a side effect decreased significantly. According to the authors’ experience, hormonal treatment does not pose a risk to the development of endometrial carcinoma; on the contrary, continuous combined preparations appear to reduce the risk of hyperplasia and, indirectly, the chances of the development of adenocarcinoma. C1 [Magyar, Zoltan] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Baross u. 27, H-1088 Budapest, Hungary. [Berkes, Eniko] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Baross u. 27, H-1088 Budapest, Hungary. [Csapo, Zsolt] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Baross u. 27, H-1088 Budapest, Hungary. [Papp, Zoltan] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Baross u. 27, H-1088 Budapest, Hungary. RP Magyar, Z (reprint author), Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, H-1088 Budapest, Hungary. EM magyar@noi1.sote.hu CR Anastasiadis PG, Koutlaki NG, Skaphida PG et al: Endometrial polypus: prevalence, detection, and malignant potential in women with abnormal uterine bleeding. Eur J Gynaecol Oncol 21: 180-183, 2000. Astrup K, Olivarius N: Frequency of spontaneously occurring postmenopausal bleeding in the general population. Acta Obstet Gynecol Scand 83: 203-205, 2004. Bachmann LM, Riet G, Clark TJ et al: Probability analysis for diagnosis of endometrial hyperplasia and cancer in postmenopausal bleeding: an approach for a rational diagnostic workup. Acta Obstet Gynecol Scand 82: 1-6, 2003. Bernard JP, Rizk E, Camatte S et al: Saline contrast sonohysterography in the preoperative assessment of benign intrauterine disorders. Ultrasound Obstet Gynecol 17: 145-149, 2001. Burger HG, Dudley EC, Hopper JL et al: The endocrinology of the menopausal transition: a cross-sectional study of a population- based sample. J Clin Endocrinol Metab 80: 3537-3545, 1995. Cerin A, Heldaas K, Moeller B: Adverse endometrial effects of long-cycle estrogen and progestagen replacement therapy. The Scandinavian Long-Cycle Study Group. New Engl J Med 334: 668-669, 1996. Clark TJ, Barton PM, Coomarasamy A et al: Investigating postmenopausal bleeding for endometrial cancer: cost-effectiveness of initial diagnostic strategies. Br J Obstet Gynecol 113: 502-510, 2006. Dahmoun M, Odmark IS, Risberg B et al: Apoptosis, proliferation, and sex steroid receptors in postmenopausal endometrium before and during HRT. Maturitas 49: 114-123, 2004. Dijkhuizen FP, Mol BW, Brolmann HA et al: The accuracy of endometrial sampling in the diagnosis of patients with endometrial carcinoma and hyperplasia: a meta-analysis. Cancer 89: 1765-1772, 2000. Epstein E, Valentin L: Managing women with post-menopausal bleeding. Best Pract Res Clin Obstet Gynaecol 18: 125-143, 2004. Erkkola R, Kumento U, Lehmuskoski S et al: No increased risk of endometrial hyperplasia with fixed long-cycle oestrogenprogestogen therapy after five years. J Br Menopause Soc 10: 9-13, 2004. Farquhar CM, Lethaby A, Sowter M et al: An evaluation of risk factors for endometrial hyperplasia in premenopausal women with abnormal menstrual bleeding. Am J Obstet Gynecol 181: 525-529, 1999. Ferenczy A: Pathophysiology of endometrial bleeding. Maturitas 45: 1-14, 2003. Ferenczy A, Gelfand MM: Endometrial histology and bleeding patterns in postmenopausal women taking sequential, combined E2 and dydrogesterone. Maturitas 26: 219-226, 1997. Goldschmit R, Katz Z, Blickstein I et al: The accuracy of endometrial Pipelle sampling with and without sonographic measurement of endometrial thickness. Obstet Gynecol 82: 727-730, 1993. Goldstein SR, Zeltser I, Horan CK et al: Ultrasonographybased triage for perimenopausal patients with abnormal uterine bleeding. Am J Obstet Gynecol 177: 102-108, 1997. Grady D, Ernster VL: Hormone replacement therapy and endometrial cancer: are current regimens safe? J Natl Cancer Inst 89: 1088-1089, 1997. Grimes DA: Diagnostic dilation and curettage: a reappraisal. Am J Obstet Gynecol 142: 1-6, 1982. Gusberg SB: Precursors of corpus carcinoma. Estrogens and adenomatous hyperplasia. Am J Obstet Gynecol 54: 905-907, 1947. Hickey M, Fraser IS: The structure of endometrial microvessels. Hum Reprod 3: 57-66, 2000. Hickey M, Higham J, Sullivan M et al: Endometrial bleeding in hormone replacement therapy users: preliminary findings regarding the role of matrix metalloproteinase 9, MMP-9, and tissue inhibitors of MMPs. Fertil Steril 75: 288-296, 2001. Hickey M, Pillai G, Higham JM et al: Changes in endometrial blood vessels in the endometrium of women with hormone replacement therapy-related irregular bleeding. Hum Reprod 18: 1100-1106, 2003. Holst T, Lang E, Winkler U et al: Bleeding patterns in peri and postmenopausal women taking a continuous combined regimen of estradiol with norethisterone acetate or a conventional sequential regimen of conjugated equine estrogens with medrogestone. Maturitas 43: 265-275, 2002. Hulley S, Grady D, Bush T et al: Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women: Heart and Estrogen/Progestin Replacement Study, HERS). JAMA 280: 605-613, 1998. Kimura T, Kamiura S, Yamamoto T et al: Abnormal uterine bleeding and prognosis of endometrial cancer. Int J Gynecol Obstet 85: 145-150, 2004. Klein NA, Soules MR: Endocrine changes of the perimenopause. Clin Obstet Gynecol 41: 912-920, 1998. March CM: Bleeding problems and treatment. Clin Obstet Gynecol 41: 928-939, 1998. Mirkin S, Navarro F, Archer DF: Hormone therapy and endometrial angiogenesis. Climact 6: 273-277, 2003. Mishell DR Jr, Kaunitz AM: Devices for endometrial sampling. A comparison. J Reprod Med 43: 180-184, 1998. Mossa B, Imperato F, Marziani R et al: Hormonal replacement therapy and evaluation of intrauterine pathology in postmenopausal women: a ten-year study. Eur J Gynaecol Oncol 24: 507-512, 2003. Oehler MK, MacKenzie I, Kehoe S et al: Assessment of abnormal bleeding in menopausal women: an update. J Br Menopause Soc 9: 117-121, 2003. Omodei U, Ferrazzia E, Ruggeri C et al: Endometrial thickness and histological abnormalities in women on hormonal replacement therapy: a transvaginal ultrasound/hysteroscopic study. Ultrasound Obstet Gynecol 15: 317-320, 2000. Papp Z, ed): A szuleszet-nogyogyaszat tankonyve. Semmelweis Kiado, Budapest, 2002. Perrone G, DeAngelis C, Critelli C et al: Hysteroscopic findings in postmenopausal abnormal uterine bleeding: a comparison between HRT users and non-users. Maturitas 43: 251-255, 2002. Pike MC, Peters RK, Cozen W: Estrogen-progestin replacement therapy and endometrial cancer. J Natl Cancer Inst 89: 1110- 1116, 1997. Reslova T, Tosner J, Resl M et al: Endometrial polypus. A clinical study of 245 cases. Arch Gynecol Obstet 262: 133-139, 1999. Samsioe G.: Hormone replacement therapy: aspects of bleeding problems and compliance. Int J Fertil Menopausal Stud 41: 11- 15, 1996. Schmidt T, Nawroth F, Breidenbach M et al: Differential indication for histological evaluation of endometrial fluid in postmenopause. Maturitas 50: 177-181, 2005. Seltzer VL, Benjamin F, Deutsch S: Perimenopausal bleeding patterns and pathological findings. J Am Med Womens Assoc 45: 132-134, 1990. Shapiro S, Kelly JP, Rosenberg L et al: Risk of localized and widespread endometrial cancer in relation to recent and discontinued use of conjugated estrogens. N Engl J Med 313: 969- 972, 1985. Sivridis E, Giatromanolaki A: Proliferative activity in postmenopausal endometrium: the lurking potential for giving rise to an endometrial adenocarcinoma. J Clin Pathol 57: 840-844, 2004. Toon VG, Patrick N: Endometrial safety of hormone replacement therapy: review of literature. Maturitas 42: 93-104, 2002. Valle RF: Office hysteroscopy. Clin Obstet Gynecol 42: 276- 289, 1999. Van de Weijer PHM, Scholten P, van der Mooren MJ: Bleeding patterns and endometrial histology in postmenopausal women taken low dose, sequential combined E2 and dydrogesterone. Climact 2: 1-9, 1999. Weber AM, Belinson JL, Piedmonte MR: Risk factors for endometrial hyperplasia and cancer among women with abnormal bleeding. Obstet Gynecol 93: 594-598, 1999. Wildemeersch D, Schacht E, Wildemeersch P: Performance and acceptability of intrauterine release of levonorgestrel with a miniature delivery system for hormonal substitution therapy, contraception and treatment in peri- and postmenopausal women. Maturitas 44: 237-245, 2003. Woodruff JD, Pickar JH: Incidence of endometrial hyperplasia in postmenopausal women taken conjugated estrogens, Premarin, with medroxyprogesterone acetate versus conjugated estrogens alone. Am J Obstet Gynecol 170: 1213-1223, 1994. Wren BG, Brown L: Compliance with hormonal replacement therapy. Maturitas 13: 17-21, 1991. Writing Group for the PEPI Trial. Effects of hormonal replacement therapy on endometrial histology in postmenopausal women. The Postmenopausal Estrogen-Progestin Interventions, PEPI, Trial. JAMA 275: 370-3751996. Zerbe MJ, Zhang J, Bristow RE et al: Retrograde seeding of malignant cells during hysteroscopy in presumed early endometrial cancer. Gynecol Oncol 79: 55-58, 2000. Zupi E, Sbracia M, Marconi D et al: TNFalpha expression in hyperplastic endometrium. Am J Reprod Immunol 44: 153- 159, 2000. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 351 EP 359 PG 9 ER PT J AU Dehaghani, SA Ghiam, FA Hosseini, M Mansouri, S Ghaderi, A AF Dehaghani, Samsami Alamtaj Ghiam, Fotouhi Alireza Hosseini, Marjan Mansouri, Sareh Ghaderi, Abbas TI Factors Influencing Serum Concentration of CA125 and CA15-3 in Iranian Healthy Postmenopausal Women SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast health; menopause; cancer ID breast health; menopause; cancer AB Screening for breast and ovarian cancers are required due to the late stage at diagnosis and poor survival. Serum CA125 and CA15-3 are important cancerdetecting agents in patients with ovarian and breast cancers, respectively. Elevation of CA125 and CA15-3 level correlates with malignant and non-malignant conditions. Moreover, a series of individual characteristics affect the serum level of these markers. The objective of the present study was to evaluate CA125 and CA15-3 levels in cancer-free postmenopausal women to investigate the impacts of patient parameters on the serum level of these markers. 203 subjects were studied prospectively. Serum CA125 and CA15-3 assessment was done subsequent to the direct interview. The associations between marker levels and presenting features were examined. CA125 and CA15-3 levels were elevated in 35 (17.2%) and 12 (5.9%) of persons, respectively. A higher CA125 level was associated with advanced age (p = 0.046), while a lower level was correlated with hormone replacement therapy (HRT) and having smoking habits (p = 0.000 and p = 0.01, respectively). CA15-3 level was remarkably lower amongst oral contraceptive (OCP) users (p = 0.03). Serum marker levels were not significantly related to menarche age, age at menopause, height, weight, BMI and parity. Serum CA125 is imperative indicator for malignancies of the ovary; however, personal and medical factors influence its serum level. A fair interpretation of results must be due to an accurate attention to the individual characteristics. C1 [Dehaghani, Samsami Alamtaj] Shiraz University of Medical Sciences, Department of Obstetrics and GynecologyShiraz, Iran. [Ghiam, Fotouhi Alireza] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Hosseini, Marjan] Shiraz University of Medical Sciences, Department of Obstetrics and GynecologyShiraz, Iran. [Mansouri, Sareh] Shiraz University of Medical Sciences, Department of Obstetrics and GynecologyShiraz, Iran. [Ghaderi, Abbas] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. RP Ghaderi, A (reprint author), Shiraz University of Medical Sciences, Institute for Cancer Research, Shiraz, Iran. EM ghaderia@sums.ac.ir CR Taechakraichana N, Jaisamrarn U, Panyakhamlerd K, Chaikittisilpa S, Limpaphayom KK: Climacteric: concept, consequence and care. J Med Assoc Thai 85:S1-15, 2002. Rymer J, Morris EP: Extracts from “Clinical evidence”: Menopausal symptoms. BMJ 321: 1516-1519, 2000. van Asselt KM, Kok HS, Putter H, Wijmenga C, Peeters PH, van der Schouw YT, Grobbee DE, te Velde ER, Mosselman S, Pearson PL: Linkage analysis of extremely discordant and concordant sibling pairs identifies quantitative trait loci influencing variation in human menopausal age. Am J Hum Genet 74: 444-453, 2004. Jamjan L, Jerayingmongkol P: Self-image of people in their fifties. Nurs Health Sci 4:A4, 2002. Pike MC, Pearce CL, Wu AH: Prevention of cancers of the breast, endometrium and ovary. Oncogene 23: 6379-6391, 2004. Jacobs IJ, Menon U: Progress and challenges in screening for early detection of ovarian cancer. Mol Cell Proteomics 3: 355- 366, 2004. Crump C, McIntosh MW, Urban N, Anderson G, Karlan BY: Ovarian cancer tumor marker behavior in asymptomatic healthy women: implications for screening. Cancer Epidemiol Biomarkers Prev 9: 1107-1111, 2000. Public Health Service. Healthy people 2010: National health promotion and disease prevention objectives – full report with commentary. Washington, DC: U.S. Department of Health and Human Services, 2000. Maeda T, Inoue M, Koshiba S, Yabuki T, Aoki M, Nunokawa E, et al: Solution structure of the SEA domain from the murine homologue of ovarian cancer antigen CA125, MUC16). J Biol Chem 279: 13174-13182, 2004. Bast RC Jr: Status of tumor markers in ovarian cancer screening. J Clin Oncol 21: 200-205, 2003. Zidan J, Hussein O, Basher W, Zohar S: Serum CA125: a tumor marker for monitoring response to treatment and follow-up in patients with non-Hodgkin’s lymphoma. Oncologist 9: 417- 421, 2004. Taylor-Papadimitriou J, Burchell JM, Plunkett T, Graham R, Correa I, Miles D, Smith M: MUC1 and the immunobiology of cancer. J Mammary Gland Biol Neoplasia 7: 209-221, 2002. Clinton SR, Beason KL, Bryant S, Johnson JT, Jackson M, Wilson C, Holifield K, Vincent C, Hall M: A comparative study of four serological tumor markers for the detection of breast cancer. Biomed Sci Instrum 39: 408-414, 2003. Seregni E, Botti C, Bajetta E, Ferrari L, Martinetti A, Nerini- Molteni S, Bombardieri E: Hormonal regulation of MUC1 expression. Int J Biol Markers 14: 29-35, 1999. O’ Rourke J, Mahon SM: A comprehensive look at the early detection of ovarian cancer. Clin J Oncol Nurs 7: 41-47, 2003. Kayaba H: Tumor markers: essential diagnostic tools for radiologists. Nippon Igaku Hoshasen Gakkai Zasshi 63: 133-139, 2003. Lu KH, Patterson AP, Wang L, Marquez RT, Atkinson EN, Baggerly KA, et al: Selection of potential markers for epithelial ovarian cancer with gene expression arrays and recursive descent partition analysis. Clin Cancer Res 10: 3291-3300, 2004. Green BB, Taplin SH: Breast cancer screening controversies. J Am Board Fam Pract 16: 233-241, 2003. Pauler DK, Menon U, McIntosh M, Symecko HL, Skates SJ, Jacobs IJ: Factors influencing serum CA125II levels in healthy postmenopausal women. Cancer Epidemiol Biomarkers Prev 10: 489-493, 2001. Lopez LA, Del Villar V, Ulla M, Fernandez F, Fernandez LA, Santos I, Rabadan L, Gutierrez M: Prevalence of abnormal levels of serum tumour markers in elderly people. Age Ageing 25: 45-50, 1996. Takami M, Sakamoto H, Ohtani K, Takami T, Satoh K: An evaluation of CA125 levels in 291 normal postmenopausal and 20 endometrial adenocarcinoma-bearing women before and after surgery. Cancer Lett 121: 69-72, 1997. Hornstein MD, Goodman HM, Thomas PP, Knapp RC, Harlow BL: Use of a second-generation CA125 assay in gynecologic patients. Gynecol Obstet Invest 42: 196-200, 1996. Cengiz B, Atabekoglu C, Cetinkaya E, Cengiz SD: Effect of hormone replacement therapy on serum levels of tumor markers in healthy postmenopausal women. Maturitas 46: 301-306, 2003. Menzin AW, Kobrin S, Pollak E, Goodman DB, Rubin SC: The effect of renal function on serum levels of CA125.Gynecol Oncol 58: 375-377, 1995. Brekelmans CT: Risk factors and risk reduction of breast and ovarian cancer. Curr Opin Obstet Gynecol 15: 63-68, 2003. Grover S, Quinn MA, Weideman P, Koh H: Factors influencing serum CA 125 levels in normal women. Obstet Gynecol 79: 511-514, 1992. Johnson KC, Hu J, Mao Y: Passive and active smoking and breast cancer risk in Canada. Cancer Causes Control 11: 211- 221, 2000. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 360 EP 364 PG 5 ER PT J AU Xin, B Yokoyama, Y Shigeto, T Mizunuma, H AF Xin, Bing Yokoyama, Yoshihito Shigeto, Tatsuhiko Mizunuma, Hideki TI Anti-Tumor Effect of Non-Steroidal Anti-Inflammatory Drugs on Human Ovarian Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE non-steroidal anti-inflammatory drugs; selective COX-2 inhibitor; non-specific COX inhibitor; meloxicam; ovarian cancer ID non-steroidal anti-inflammatory drugs; selective COX-2 inhibitor; non-specific COX inhibitor; meloxicam; ovarian cancer AB Many reports have demonstrated that non-steroidal anti-inflammatory drugs (NSAIDs) suppress malignant transformation and tumor growth, and some NSAIDs are expected to be new anti-cancer agents. In this study, we examined the anti-tumor effects of the non-specific cyclooxygenase (COX) inhibitors aspirin and piroxicam, and the selective COX-2 inhibitor meloxicam on xenotransplanted ovarian cancer. Tumor growth and survival were compared in female nu/nu mice, xenografted with subcutaneous OVCAR-3 tumors or with intraperitoneal DISS tumors and treated with aspirin (200 ppm in diet, everyday), piroxicam (150 ppm in diet, everyday) or meloxicam (162 ppm in diet, everyday). Al, of the agents tested significantly suppressed the growth of OVCAR-3 tumors xenotransplanted subcutaneously as compared to the control. There was a significant difference in inhibition of OVCAR-3 tumor growth between meloxicam and aspirin treatment. Meloxicam and piroxicam treatment significantly prolonged survival of mice with malignant ascites derived from DISS cells as compared to control and aspirin treatment. Mice treated with meloxicam survived significantly longer than those treated with piroxicam. There was no significant difference in survival between control and aspirin treatment. Necropsy revealed that one of the 6 cancer-bearing mice treated with piroxicam suffered from stomach perforation. These results indicate that a selective COX-2 inhibitor produces greater anti-tumor effect against ovarian cancer than a nonselective COX inhibitor and that meloxicam may have a potential of leading to a novel therapeutic strategy against ovarian cancer. C1 [Xin, Bing] Hirosaki University School of Medicine, Department of Obstetrics and Gynecology, 5-Zaifu-cho, 036-8562 Hirosaki, Japan. [Yokoyama, Yoshihito] Hirosaki University School of Medicine, Department of Obstetrics and Gynecology, 5-Zaifu-cho, 036-8562 Hirosaki, Japan. [Shigeto, Tatsuhiko] Hirosaki University School of Medicine, Department of Obstetrics and Gynecology, 5-Zaifu-cho, 036-8562 Hirosaki, Japan. [Mizunuma, Hideki] Hirosaki University School of Medicine, Department of Obstetrics and Gynecology, 5-Zaifu-cho, 036-8562 Hirosaki, Japan. RP Yokoyama, Y (reprint author), Hirosaki University School of Medicine, Department of Obstetrics and Gynecology, 036-8562 Hirosaki, Japan. EM yokoyama@cc.hirosaki-u.ac.jp CR Aas AT, Tonnessen TI, Brun A, Salford LG: Growth inhibition of rat glioma cells in vitro and in vivo by aspirin. J Neurooncol 24: 171-180, 1995. Akhmedkhanov A, Toniolo P, Zeleniuch-Jacquotte A, et al: Aspirin and epithelial ovarian cancer. Prev Med 33: 682-687, 2001. Baron JA, Sandlers RS: Non-steroidal anti-inflammatory drugs and cancer prevention. Annu Rev Med 51: 511-523, 2000. Blanco FJ, Guitian R, Moreno J, et al: Effect of anti-inflammatory drugs on COX-1 and COX-2 activity in human articular chondrocytes. J Rheumatol 26: 1366-1373, 1999. Cramer DW, Harlow BL, Titus-Ernstoff L, et al: Over-thecounter analgesics and risk of ovarian cancer. Lancet 351: 104- 107, 1998. Craven PA, DeRubertis FR: Effect of aspirin on 1,2-dimethylhydrazine- induced colonic carcinogenesis. Carcinogenesis 13: 541-546, 1992. Denkert C, Kobel M, Pest S, et al: Expression of cyclooxygenase 2 is an independent prognostic factor in human ovarian carcinoma. Am J Pathol 160: 893-903, 2002. di Palma A, Matarese G, Leone V, et al: Aspirin reduces the outcome of anticancer therapy in Meth A-bearing mice through activation of AKT-glycogen synthase kinase signaling. Mol Cancer Ther 5: 1318-1324, 2006. Din FV, Dunlop MG, Stark LA: Evidence for colorectal cancer cell specificity of aspirin effects on NFB signalling and apoptosis. Br J Cancer 91: 381-388, 2004. Drake JG, Becker JL: Aspirin-induced inhibition of ovarian tumor cell growth. J Obstet Gynecol 100: 677-682, 2002. Duperron C, Castonguay A: Chemoprevention efficacies of aspirin and sulindac against lung tumorigenesis in A/J mice. Carcinogenesis 18: 1001-1006, 1997. Ferrandina G, Lauriola L, Zannoni GF, et al: Increased cyclooxygenase-2, COX-2, expression is associated with chemotherapy resistance and outcome in ovarian cancer patients. Ann Oncol 13: 1205-1211, 2002. Greenlee RT, Murray T, Bolden S, Wingo PA: Cancer statistics. Ca Cancer J Clin 50: 7-33, 2000. Goldman AP, Williams CS, Sheng H, et al: Meloxicam inhibits the growth of colorectal cancer cells. Carcinogenesis 19: 2195- 2199, 1998. Henry D, Lim LLY, Rodriguez LS, et al: Variability in risk of gastrointestinal complications with individual non-steroidal anti-inflammatory drugs: results of a collaborative meta-analysis. Br Med J 312: 1563-1566, 1996. Kern MA, Schoneweis MM, Sahli D, et al: Cyclooxygenase-2 inhibitors suppress the growth of human hepatocellular carcinoma implant in nude mice. Carcinogenesis 25: 1193-1199, 2004. Laudanno OM, Cesolari JA: In vivo selectivity of nonsteroidal anti-inflammatory drugs and gastrointestinal ulcer in rats. Dig Dis Sci 45: 1359-1365, 2000. Li H, Kramer P, Steele V, et al: Effect of duration of treatment with piroxicam on azoxymethane-induced colon cancer, aberrant crypt foci, apoptosis and cell proliferation in rats. Cancer Lett 147: 187-19323, 1998. Liu CH, Chang SH, Narko K, et al: Overexpression of cyclooxygenase-2 is sufficient to induce tumorigenesis in transgenic mice. J Biol Chem 276: 18563-18569, 2000. Mohammed SI, Bennett PF, Craig BA, et at: Effects of the cyclooxygenase inhibitor, piroxicam, on tumor response, apoptosis, and angiogenesis in a canine model of human invasive urinary bladder cancer. Cancer Res 62: 356-358, 2002. Moysich KB, Mettlin C, Piver MS, et al: Regular use of analgesic drugs and ovarian cancer risk. Cancer Epidem Biomarkers Prev 10: 903-906, 2001. Piazza GA, Alberts DS, Hixson LJ, et al: Sulindac sulfone inhibits azoxymethane-induced colon carcinogenesis inn rats without reduced prostaglandin levels. Cancer Res 57: 2909- 2915, 1997. Pollard M, Luckert PH: Prevention and treatment of primary intestinal tumors in rats by piroxicam. Cancer Res 49: 6471- 6473, 1989. Prescott SM, White RL: Intimate connection between APC and prostaglandin H synthase-2. Cell 87: 783-786, 1996. Reddy BS, Rao CV, Rivenson A, Kelloff G: Inhibitory effect of aspirin on azoxymethane-induced colon carcinogenesis in F344 rats. Carcinogenesis 14: 1493-1497, 1993. Ricchi P, Pignata S, Di Popolo A, et al: Effect of aspirin treatment on cell proliferation and differentiation of colon adenocarcinoma Caco-2 cells. Int J Cancer 73: 880-884, 1997. Rozic JG, Chakraborty C, Lala PK: Cyclooxygenase inhibitors retard murine mammary tumor progression by reducing tumor cell migration, invasiveness and angiogenesis. Int J Cancer 93: 497-506, 2001. Sakamoto A, Yokoyama Y, Umemoto M, et al: Clinical implication of expression of cyclooxygenase-2 and peroxisome proliferator activated-receptor g in epithelial ovarian tumours. Br J Cancer 91: 633-638, 2004. Sheng H, Shao J, Kirkland SC, et al: Inhibition of human colon cancer cell growth by selective inhibition of cyclooxygenase-2. J Clin Invest 99: 2254-2259, 1997. Shibata MA, Hirose M, Masuda A, et al: Modification of BHA forestomach carcinogenesis in rats: inhibition by diethylmaleate or indomethacin and enhancement by a retinoid. Carcinogenesis 14: 1265-1269, 1993. Shigemasa K, Tian X, Gu L, et al: Expression of cyclooxygenase- 2 and its relationship to p53 accumulation in ovarian adenocarcinomas. Int J Oncol 22: 99-105, 2003. Smalley W, DuBois RN: Colorectal cancer and non steroidal anti-inflammatory drugs. Adv Pharmacol 39: 1-20, 1997. Smith WL, Garavito RM, DeWitt DL: Prostaglandin endoperoxide H synthases, cyclooxygenase)-1 and -2. J Biol Chem 271: 33157-33160, 1996. Taketo MM: Cyclooxygenase-2 inhibitors in tumorigenesis, Part I). J Natl Cancer Inst 90: 1529-1536, 1998. Tsubouchi Y, Sano H, Kawahito Y, et al: Meloxicam inhibits the growth of non-small cell lung cancer. Anticancer Res 20: 2867- 2872, 2000. Tsujii M, Kawano S, Tsuji S, et al: Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell 93: 705-716, 1998. van Rees BP, Saukkonen K, Ristimaki A, et al: Cyclooxygenase- 2 expression during carcinogenesis in the human stomach. J Pathol 196: 171-179, 2002. Yao M, Kargman S, Lam EC, et al: Inhibition of cyclooxygenase- 2 by rofecoxib attenuates the growth and metastatic potential of colorectal carcinoma in mice. Cancer Res 63: 586-592, 2003. Yokoyama Y, Sakamoto T, Sato S, Saito Y: Evaluation of cytoreductive surgery with pelvic and paraaortic lymphadenectomy and intermittent cisplatin-based combination chemotherapy for improvement of long-term survival in ovarian cancer. Eur J Gynaecol Oncol 20: 361-366, 1999. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 365 EP 369 PG 5 ER PT J AU Pecina-Slaus, N Niku Eva-Martic, T Beros, V Davor, T AF Pecina-Slaus, Nives Niku Eva-Martic, Tamara Beros, Vili Davor, Tomas TI Genetic Alterations of E-cadherin and Beta-Catenin in Germinoma and Teratoma: Report of Two Central Nervous System Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE germinoma; teratoma; E-cadherin gene (CDH1); beta-catenin gene (CTNNB1); Wnt signaling pathway ID germinoma; teratoma; E-cadherin gene (CDH1); beta-catenin gene (CTNNB1); Wnt signaling pathway AB The genetic basis as well as mechanisms of development of germ cell tumors of the CNS are still unexplained. In the present article changes of Ecadherin (CDH1) and beta-catenin (CTNNB1) genes in two CNS germ cell tumors are reported. Both gene products are components of adherens junctions, but are also involved in the Wnt signaling pathway. A case of germinoma of the central nervous system and a case of spinal channel teratoma were tested for loss of heterozygosity (LOH) of E-cadherin gene by PCR amplification of tetranucleotide polymorphism (D16S752). Changes of beta-catenin were tested by heteroduplex method. Both germ cell tumors analyzed demonstrated LOH of the CDH1 gene. Analysis of exon 3 of the CTNNB1 gene showed additional band in the germinoma, suggesting that this sample harbors mutation in the beta-catenin gene. Immunostaining showed that LOHs in our samples were accompanied with the absence of E-cadherin protein. We also investigated E-cadherin expression in four other germinomas, of which three were negative and one was mildly positive. Our findings may contribute to better understanding of the genetic profile of germ cell tumors. C1 [Pecina-Slaus, Nives] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Laboratory of Neurooncology, alata 3,, HR-10000 Zagreb, Croatia. [Niku Eva-Martic, Tamara] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Laboratory of Neurooncology, alata 3,, HR-10000 Zagreb, Croatia. [Beros, Vili] University Hospital Sisters of Charity, Department of NeurosurgeryZagreb, Croatia. [Davor, Tomas] University Hospital Sisters of Charity, Ljudevit Jurak Department of PathologyZagreb, Croatia. RP Pecina-Slaus, N (reprint author), School of Medicine University of Zagreb, Croatian Institute for Brain Research, Laboratory of Neurooncology, HR-10000 Zagreb, Croatia. EM nina@mef.hr CR Adamah DJ, Gokhale PJ, Eastwood DJ, Goepel J, Walsh JR, Moore HD, Andrews PW: Dysfunction of the mitotic:meiotic switch as a potential cause of neoplastic conversion of primordial germ cells. Int J Androl 29:219-227, 2006 Biegel JA: Cytogenetics and molecular genetics of childhood brain tumors. Neuro-oncol 1:139-151, 1999 Bussey KJ, Lawce HJ, Olson SB, Arthur DC, Kalousek DK, Krailo M, Giller R, Heifetz S, Womer R, Magenis RE: Chromosome abnormalities of eighty-one pediatric germ cell tumors: sex-, age-, site-, and histopathology-related differences – a Children’s Cancer Group study. Genes Chromosomes Cancer 25:134-146, 1999 Ellison DW, Onilude OE, Lindsey JC, Lusher ME, Weston CL, Taylor RE, Pearson AD, Clifford SC: Beta-catenin status predicts a favorable outcome in childhood medulloblastoma: the United Kingdom Children’s Cancer Study Group Brain Tumour Committee. J Clin Oncol 23:7951-7957, 2005 Fritsch MK, Schneider DT, Schuster AE, Murdoch FE, Perlman EJ: Activation of Wnt/beta-catenin signaling in distinct histologic subtypes of human germ cell tumors. Pediatr Dev Pathol 9:115-131, 2006 He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B, Kinzler KW: Identification of c-MYC as a target of the APC pathway. Science 281:1509-1512, 1998 Hejazi N, Witzmann A: Spinal intramedullary teratoma with exophytic components: report of two cases and review of the literature. Neurosurg Rev 26:113-116, 2003 Honecker F, Kersemaekers AM, Molier M, Van Weeren PC, Stoop H, De Krijger RR, Wolffenbuttel KP, Oosterhuis W, Bokemeyer C, Looijenga LH: Involvement of E-cadherin and betacatenin in germ cell tumours and in normal male fetal germ cell development. J Pathol 204:167-174, 2004 Hulsken J, Birchmeier W, Behrens J: E-cadherin and APC compete for the interaction with beta-catenin and the cytoskeleton. J Cell Biol 127:2061-2069, 1994 Koch A, Waha A, Tonn JC, Sorensen N, Berthold F, Wolter M, Reifenberger J, Hartmann W, Friedl W, Reifenberger G, Wiestler OD, Pietsch T: Somatic mutations of WNT/wingless signaling pathway components in primitive neuroectodermal tumors. Int J Cancer 3:445-449, 2001 Lie DC, Colamarino SA, Song HJ, Desire L, Mira H, Consiglio A, Lein ES, Jessberger S, Lansford H, Dearie AR, Gage FH: Wnt signalling regulates adult hippocampal neurogenesis. Nature 437:1370-1375, 2005 Mishima K, Kato Y, Kaneko MK, Nakazawa Y, Kunita A, Fujita N, Tsuruo T, Nishikawa R, Hirose T, Matsutani M: Podoplanin expression in primary central nervous system germ cell tumors: a useful histological marker for the diagnosis of germinoma. Acta Neuropathol 111:563-568, 2006 Orford K, Crockett C, Jensen JP, Weissman AM, Byers SW: Serine phosphorylation-regulated ubiquitination and degradation of beta-catenin. J Biol Chem 272: 24735-24738, 1997 Ozawa M, Kelmer R: Molecular organization of the uvomorulin- catenin complex. J Cell Biol 116:989–996, 1992 Patapoutian A, Reichardt LF: Roles of wnt proteins in neural development maintenance. Curr Opin Neurobiol 10:392-399, 2000 Pecina-Slaus N, Zigmund M, Kusec V, Nikuseva Martic T, Cacic M, Slaus M: E-cadherin and β-catenin expression patterns in malignant melanoma assessed by image analysis. J Cutan Pathol 34:239-246, 2007 Pecina-Slaus N: Tumor suppresor gene E-cadherin and its role in normal and malignant cells. Cancer Cell Int E3, 17, 2003, http://www.cancerci.com/content/3/1/17) Peifer M, Polakis P: Wnt signaling in oncogenesis and embryogenesis - a look outside the nucleus. Science 287:1606- 1609, 2000 Rosenblum MK, Matsutani M, Van Meir EG: CNS germ cell tumours. In: World Health Organisation Classification of Tumours: Pathology and Genetics of Tumours of the Nervous System., Eds: Kleihues P and Cavenee WK), IARC Press, Lyon, 2000, pp 207-214 Sano K: Pathogenesis of intracranial germ cell tumors reconsidered. J Neurosurg 90: 258-264, 1999 Santagata S, Hornick JL, Ligon KL: Comparative analysis of germ cell transcription factors in CNS germinoma reveals diagnostic utility of NANOG. Am J Surg Pathol 30:1613-1618, 2006 Scotting PJ: Are cranial germ cell tumours really tumours of germ cells? Neuropathol Appl Neurobiol 32:569-574, 2006 Yokota N, Nishizawa S, Ohta S, Date H, Sugimura H, Namba H, Maekawa M: Role of wnt pathway in medulloblastoma oncogenesis. Int J Cancer 101:198-201, 2002 Yu X, Malenka RC: Beta-catenin is critical for dendritic morphogenesis. Nature Neurosci 6:1169-1177, 2003 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 370 EP 374 PG 5 ER PT J AU Solis-Coria, A Vargas-Gonzalez, R Sotelo-Avila, C AF Solis-Coria, Araceli Vargas-Gonzalez, Roberto Sotelo-Avila, Cirilo TI Rhabdomyomatous Mesenchymal Hamartoma Presenting as a Skin Tag in the Sternoclavicular Area SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE rhabdomyomatous mesenchymal hamartoma; striated muscle hamartoma ID rhabdomyomatous mesenchymal hamartoma; striated muscle hamartoma AB Rhabdomyomatous mesenchymal hamartoma (RMH) is a rare congenital lesion of the dermis and subdermis. It has been described predominantly in newborns, with 30 cases reported in the English literature. Typically, it appears as a skin tag, papule, nodule or a mass involving the face or sternal notch. A 28-day-old girl presented with a 1.4 x 0.8 cm soft skin tag in the right sternoclavicular area. Physical examination revealed no congenital anomalies. A shaved biopsy showed that the core of the lesion contained striated muscle fibers mixed with hair follicles and sebaceous and eccrine glands. Thin epidermis lined the outside of the tag. We report a patient with a RMH in a site not previously reported and discuss the differential diagnosis. C1 [Solis-Coria, Araceli] Clinica de Dermatologia y Cirugia Estetica de PueblaPuebla, Mexico. [Vargas-Gonzalez, Roberto] Hospital Para el Nino Poblano, Laboratorio de Inmunopatologia de Puebla and Department of Pathology, Km 1.5 Carretera Federal Puebla, 72190 Atlixco, Puebla, Mexico. [Sotelo-Avila, Cirilo] Cardinal Glennon Children’s Medical Center, Department of PathologySt. Louis, MO, USA. RP Vargas-Gonzalez, R (reprint author), Hospital Para el Nino Poblano, Laboratorio de Inmunopatologia de Puebla and Department of Pathology, 72190 Atlixco, Puebla, Mexico. EM soncoy@msn.com CR Hendrick SJ, Sanchez RL, Blackwell SJ, Raimer SS: Striated muscle hamartoma: Description of two cases. Pediatr Dermatol 3:153-157, 1986 Read RW, Burnstine M, Rowland JM, Zamir E, Rao NA: Rhabdomyomatous mesenchymal hamartoma of the eyelid. Report of a case and literature review. Ophthalmol 108:798-803, 2001 Rosenberg AS, Kirk J, Morgan MB: Rhabdomyomatous mesenchymal hamartoma: an unusual dermal entity with a report of two cases and review of the literature. J Cutan Pathol 29: 238- 243, 2002 Mills AE: Rhabdomyomatous mesenchymal hamartoma of skin. Am J Dermatopathol 11:58-63, 1989 Elgart GW, Patterson JW: Congenital midline hamartoma: case report with histochemical and immunohistochemical findings. Pediatr Dermatol 7:199-201, 1990 Shan EE, Garen PD, Pai GS, Levkoff AH, Hagerty RC, Maize JC: Multiple rhabdomyomatous mesenchymal hamartomas of skin. Am J Dermatopathol 12:485-491, 1990 Sanchez RL, Raimer SS: Clinical and histologic features of striated muscle hamartoma: possible relationship to Delleman´s syndrome, case report). J Cutan Pathol 21:40-46, 1994 Jansen T, Romiti R, Altmeyer P: Accessory tragus: Report of two cases and review of the literature. Pediatr Dermatol 17:391-394, 2000 Bergevin MA, Sheft Stan, Myer III C, Mc Adams A: Congenital midline cervical cleft. Pediatr Pathol 9: 731-739, 1989 Whitaker SR, Sprinkle M, Chov SM: Nasal glioma. Arch Otolaryngol 107:550-554, 1981. Sotelo AC, Bale PM: Subdermal fibrous hamartoma of infancy: pathology of 40 cases and differential diagnosis. Pediatr Pathol 14:39-52, 1994 Di Sant’ Agnese PA, Knowles DM: Extracardiac rhabdomyoma: a clinico-pathologic study and review of the literature. Cancer 46:780-789, 1980 Mehregan AH, Tavafoghi V, Ghandochi A: Nevus lipomatosus cutaneous superficialis. J Cutan Pathol 2:307-313, 1975 Markel SF, Enzinger FM: Neuromuscular hamartoma – a benign “triton tumor” composed of mature neural and striated muscle elements. Cancer 49:140-144, 1982 Murphy GF: Dermatopathology.1st ed, W.B. Saunders Company, 1995, pp 191-192 Scrivener Y, Petiau P, Rodier-Bruant C, Cribier B, Heid E, Grosshans E: Perianal striated muscle hamartoma associated with hemangioma. Pediatr Dermatol 15:274-276, 1998 Nakanishi H, Hashimoto I, Takiwaki H, Urano Y, Arase S: Striated muscle hamartoma of the nostril. J Dermatol 22:504-507, 1995 Hayes M, van der Westhuizen N: Congenital rhabdomyomatous mesenchymal hamartoma, letter). Am J Dermatopathol 14:64- 65, 1992 Katsumata M, Keong CH, Satoh T: Rhabdomyomatous mesenchymal hamartoma of skin. J Dermatol 17:384-387, 1990 Grilli R, Escalonilla P, Soriano ML, Farina C, Renedo G, Martin L, Requena L: The so-called striated muscle hamartoma is a hamartoma of cutaneous adnexa and mesenchyme, but not of striated muscle, letter). Acta Dermatol Venereol 78: 390, 1998 Ferguson JW, Hutchison HT, Rouse BM: Ocular cerebral and cutaneous malformations: confirmation of an association. Clin Gen 25:464-469, 1984 Miura T, Inoue K: A case of a congenital skin mass in the nostril with a lipoma of the corpus callosum. Jpn J Plast Reconstr Surg 33:365-369, 1990 Chen SH, Driscoll MS, Sanchez RL, Raimer SS: A flesh-colored papule on the neck of a child. Pediatr Dermatol 16:65-67, 1999 Ashfaq R, Timmons CF: Rhabdomyomatous mesenchymal hamartoma of skin. Pediatr Pathol 12:731-735, 1992 Farris PE, Manning S, Vuitch F: Rhabdomyomatous mesenchymal hamartoma. Am J Dermatopathol 16:73-75, 1994 Chang CP, Chen GS: Rhabdomyomatous mesenchymal hamartoma: a plaque-type variant in an adult. Kaohsiung J Med Sci 21:185-188, 2005 Takeyama J, Hayashi T, Sanada T, Shimanuki Y, Saito M, Shirane R: Rhabdomyomatous mesenchymal hamartoma associated with nasofrontal meningocele and dermoid cyst. J Cutan Pathol 32:310-313, 2005 Ortak T, Orbay H, Unlu E, Uysal C, Uraloglu M, Sensoz OM: Rhabdomyomatous mesenchymal hamartoma. J Craniofac Surg 16:1135-1137, 2005 Magro G, Di Benedetto A, Sanges G, , Scalisi F, Alaggio R: Rhabdomyomatous mesenchymal hamartoma of oral cavity: an unusual location for such a rare lesion. Virchows Arch 446:346-347, 2005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 375 EP 378 PG 4 ER PT J AU Cantu De Leon, D Perez Montiel, D Chanona Vilchis, J AF Cantu De Leon, David Perez Montiel, Delia Chanona Vilchis, Jose TI Unusual Case of Subcutaneous Angiosarcoma Metastatic to the Ovary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE ovary; metastatic sarcoma; angiosarcoma ID ovary; metastatic sarcoma; angiosarcoma AB The ovaries are common sites for metastatic disease, however, the most frequent ones are carcinomas. Metastatic sarcomas are very rare in ovary and most of them arise from genital tract. We present the case of a 33-year-old woman with subcutaneous angiosarcoma who had metastatic disease to the ovary resulting in acute abdominal pain. Discussion of the case and a review of the literature are presented. C1 [Cantu De Leon, David] Instituto Nacional de Cancerologia, Department of Surgical Oncology, Av San Fernando #22 Col Seccion XVI, 14080 Mexico City, Mexico. [Perez Montiel, Delia] Instituto Nacional de Cancerologia, Department of PathologyMexico City, Mexico. [Chanona Vilchis, Jose] Instituto Nacional de Cancerologia, Department of PathologyMexico City, Mexico. RP Cantu De Leon, D (reprint author), Instituto Nacional de Cancerologia, Department of Surgical Oncology, 14080 Mexico City, Mexico. EM dcantu@itesm.mx CR Moore RG, Chung M, Granai CO, et al: Incidence of metastasis to the ovaries from nongenital tract primary tumors. Gynecol Oncol 93:87-89, 2004. Ulbright TM, Roth LM, Stehman FB: Secondary ovarian neoplasia. A clinicopathologic study of 35 cases. Cancer 53:1164- 1174, 1984. Young RH, Scully RE: Sarcomas metastatic to the ovary: A report of 21 cases. Int J Gynecol Pathol 9:931-952, 1990. Mazur MT, Hsueh S, Gersell DJ: Metastases to the female genital tract. Analysis of 325 cases. Cancer 53:1978-1984, 1984. Fujiwara K, Ohishi Y, Koike H, et al: Clinical implications of metastases to the ovary. Gynecol Oncol 59:124-128, 1995. Blamey S, McDermott F, Pihl E, et al: Ovarian involvement in adenocarcinoma of the colon and rectum. Surg Gynecol Obstet 153:42-44, 1981. Demopoulos RI, Tuger L, Dubin N: Secondary ovarian carcinoma: a clinical and pathological evaluation. Int J Gynecol Pathol 6:166-175, 1987. Pillay K, Essa AS, Chetty R: Borderline serous cystadenocarcinoma with coexistent angiosarcoma: an unusual form of ovarian carcinoma. Int J Surg Pathol 9:317-321, 2001. Platt JS, Rogers SJ, Flynn CA, et al. Primary angiosarcoma of the ovary: A case report and review of the literature. Gynecol Oncol 73: 443-446, 1999. Scully R. Young RH, Clement PB: Tumors of the ovary, maldeveloped gonads, fallopian tube and broad ligament. Armed Forces Institute of Pathology, 3rd series, 1998. Petru E, Pickel H, Heydarfadai M, et al: Nongenital cancers metastatic to the ovary. Gynecol Oncol 44:83-86, 1992. Yada-Hashimoto N, Yamamoto T, Kamiura S, et al: Metastatic ovarian tumors: A review of 64 cases. Gynecol Oncol 89:314- 317, 2003. Shiromizu K, Kawana T, Sugase M, et al: Experience with the treatment of metastatic ovarian carcinoma. Arch Gynecol Obstet 243:111-114, 1988. Soloway I, Latour J, Young M: Krukenberg tumors of the ovary. Obstet Gynecol 8:636-638, 1956. Eltabbakh GH, Belison JL, Biscotti CU: Osteosarcoma metastatic to the ovary: A case report and review of the literature. Int J Gynecol Pathol 16:76-78, 1997. Webb M, Decker D, Mussey E: Cancer metastatic to the ovary. Obstet Gynecol 45:391-396, 1975. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 379 EP 381 PG 3 ER PT J AU Szynglarewicz, B Matkowski, R Smorag, Z Forgacz, J Pudelko, M Kornafel, J AF Szynglarewicz, Bartlomiej Matkowski, Rafal Smorag, Zbigniew Forgacz, Jozef Pudelko, Marek Kornafel, Jan TI Hepatitis C Virus Infection and Locally Advanced Splenic Marginal Zone Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE hepatitis C; splenic neoplasm; marginal zone lymphoma ID hepatitis C; splenic neoplasm; marginal zone lymphoma AB Splenic marginal zone lymphoma (SMZL) is a rare malignant B-cell neoplasm, usually with an indolent clinical course and favorable prognosis. Treatment options include chemotherapy, surgery, radiation and immunotherapy. In some recent studies an increased incidence of hepatitis C virus (HCV) infection in patients with SMZL was reported and its possible role in lymphomagenesis was emphasized. A 66-year-old woman with twelve-year history of HCV infection was admitted due to locally advanced abdominal tumor involving the spleen and the left part of the diaphragm. Transaminase serum levels were not elevated. Neither peripheral lymphadenopathy nor bone marrow pathology was found. Absolute blood lymphocyte, erythrocyte and platelet counts were normal. A splenectomy with partial diaphragm resection in one block was performed. Recovery was uneventful. Pathologic examination with immunohistochemistry revealed SMZL and confirmed a neoplastic infiltration of the resected diaphragm. Following surgery, chemotherapy (CHOP regimen) and immunotherapy (anti-CD20 antibody) were given. At the last follow-up 15 months after surgery, the patient was free of any symptoms of lymphoma. Surgical resection of even locally advanced SMZL with involvement of adjacent tissues can be performed as a diagnostic and therapeutic procedure. Splenectomy is especially indicated in symptomatic patients without other sites of the disease. HCV infection may result in increased risk of SMZL due to the induction of B-cell lymphoproliferation. Because of possible lymphoma regression following anti-viral therapy, a systematic screening for HCV in patients with SMZL seems to be valuable and helpful for treatment planning. C1 [Szynglarewicz, Bartlomiej] Lower Silesian Oncology Center, Department of Oncological Surgery, Plac Hirszfelda 12, 53-413 Wroclaw, Poland. [Matkowski, Rafal] Wroclaw Medical University, Department of OncologyWroclaw, Poland. [Smorag, Zbigniew] Lower Silesian Oncology Center, Department of Oncological Surgery, Plac Hirszfelda 12, 53-413 Wroclaw, Poland. [Forgacz, Jozef] Lower Silesian Oncology Center, Department of Oncological Surgery, Plac Hirszfelda 12, 53-413 Wroclaw, Poland. [Pudelko, Marek] Lower Silesian Oncology Center, Department of Oncological Surgery, Plac Hirszfelda 12, 53-413 Wroclaw, Poland. [Kornafel, Jan] Wroclaw Medical University, Department of OncologyWroclaw, Poland. RP Szynglarewicz, B (reprint author), Lower Silesian Oncology Center, Department of Oncological Surgery, 53-413 Wroclaw, Poland. EM szynglarewicz.b@dco.com.pl CR Arcaini L, Paulli M, Boveri E, Vallisa D, Bernuzzi P, Orlandi E, Incardona P, Brusamolino E, Passamonti F, Burcheri S, Schena C, Pascutto C, Cavanna L, Margini U, Lazzarino M: Splenic and nodal marginal zone lymphomas are indolent disorders at high hepatitis C virus seroprevalence with distinct presenting features but similar morphologic and phenotypic profiles. Cancer 100: 107-115, 2004 Carbonari M, Caprini E, Tedesco T, Mazzetta F, Tocco V, Casato M, Russo G, Fiorilli M: Hepatitis C virus drives the unconstrained monoclonal expansion of VH1-69-expressing memory B cells in type II cryoglobulinemia: a model of infection-driven lymphomagenesis. J Immunol 174: 6532-6539, 2005 Hermine O, Lefrere F, Bronowicki JP, Mariette X, Jondeau K, Eclache-Sandreau V, Delmas B, Valensi F, Cacoub P, Brechot C, Varet B, Troussard X: Regression of splenic lymphoma with villous lymphocytes after treatment of hepatitis C virus infection. N Engl J Med 347: 89-94, 2002 Iannitto E, Ambrosetti A, Ammatuna E, Colosio M., Florena AM, Tripodo C, Minardi V, Calvaruso G, Mitra ME, Pizzolo G, Menestrina F, Franco V: Splenic marginal zone lymphoma with or without villous lymphocytes. Hematologic findings and outcomes in a series of 57 patients. Cancer 101: 2050-2057, 2004 Kelaidi C, Rollot F, Park S, Tulliez M, Christoforov B, Calmus Y, Podevin P, Bouscary D, Sogni P, Blanche P, Dreyfus F: Response to antiviral treatment in hepatitis C virus-associated marginal zone lymphomas. Leukemia 18: 1711-1716, 2004 Marasca R, Vaccari P, Luppi M, Zucchini P, Castelli I, Barozzi P, Cuoghi A, Torelli G: Immunoglobulin gene mutations and frequent use of VH1-69 and VH4-34 segments in hepatitis C virus-positive and hepatitis C virus-negative nodal marginal zone B-cell lymphoma. Am J Pathol 159: 253-261, 2001 Oscier D, Owen R, Johnson S: Splenic marginal zone lymphoma. Blood Rev 19: 39-51, 2005 Pitini V, Arrigo C, Righi M, Scaffidi M, Sturniolo G: Systematic screening for HCV infection should be performed in patients with splenic marginal zone lymphoma. Br J Haematol 124: 252-253, 2004 Svoboda J, Andreadis C, Downs LH, Miller Jr WT, Tsai DE, Schuster SJ: Regression of advanced non-splenic marginal zone lymphoma after treatment of hepatitis C virus infection. Leuk Lymphoma 46: 1365-1368, 2005 Weng WK, Levy S: Hepatitis C virus, HCV, and lymphomagenesis. Leuk Lymphoma 44: 1113-1120, 2003 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2007 VL 13 IS 4 BP 382 EP 384 PG 3 ER PT J AU Kopper, L AF Kopper, Laszlo TI Lapatinib: A Sword with Two Edges SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Lapatinib; Targeted therapy; EGFR-inhibition; HER2-inhibition; Trastuzumab resistance; Breast cancer ID Lapatinib; Targeted therapy; EGFR-inhibition; HER2-inhibition; Trastuzumab resistance; Breast cancer AB Lapatinib is an oral dual tyrosine kinase inhibitor targeting EGFR1 and EGFR2 (HER2). Phase I trials have shown that lapatinib is well tolerated, with mild diarrhea and skin rush as common adverse effects, and low cardiotoxicity. Phase II and III trials provided evidences on clinical effectiveness in advanced or metastatic breast cancer and potential against brain metastases. Lapatinib is active in combination with trastuzumab and in trastuzumab-resistant patients, moreover it has synergistic action with capecitabine. Several clinical trials are in progress to explore the effectiveness of lapatinib in other combinations and against several tumor types. C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM kopper@korb1.sote.hu CR Bilancia D, Rosati G, Dinota A et al, 2007, Lapatinib in breast cancer. Ann Oncol 18(suppl 6):vi26–vi30 Harari PM, Allen GW, Bonner JA, 2007, Biology of interactions: antiepidermal growth factor receptor agents. J Clin Oncol 25:4057–4065 Wood ER, Truesdale AT, Mc Donald OB et al, 2004, A unique structure for epidermal growth factor receptor bound to GW572016, lapatinib): relationships among protein conformation, inhibitor off-rate, and receptor activity in tumor cells. Cancer Res 64:6652–6659 Xia W, Liu LH, Ho P, Spector NL, 2004, Truncated ErbB2 receptor, p95ErbB2, is regulated by heregulin through heterodimer formation with ErbB3 yet remains sensitive to the dual EGFR/ErbB2 kinase inhibitor GW572016. Oncogene 23:646–653 Rusnak DW, Lackey K, Affleck K et al, 2001, The effects of the novel, reversible epidermal growth factor receptor/ErbB-2 tyrosine kinase inhibitor, GW2016, on the growth of human normal and tumorderived cell lines in vitro and in vivo. Mol Cancer Ther 1:85–94 Chu I, Blackwell K, Chen S, Slingerland J, 2005, The dual ErbB1/ ErbB2 inhibitor, lapatinib, GW572016), cooperates with tamoxifen to inhibit both cell proliferation- and estrogen-dependent gene expression in antiestrogen-resistant breast cancer. Cancer Res 65:18–25 Burris HA, Hurwitz HI, Dees C et al, 2005, Phase I safety, pharmacokinetics, and clinical activity study of lapatinib, GW572016), a reversible dual inhibitor of epidermal growth factor receptor tyrosine kinases in heavily pretreated patients with metastatic carcinomas. J Clin Oncol 23:5305–5313 Blackwell KL, Burstein H, Pegram M, et al, 2005, Determining relevant biomarkers from tissue and serum that may predict response to single agent lapatinib in trastuzumab refractory metastatic breast cancer. Proc Am Soc Clin Oncol 23:(abstr 3004) Chu QSC, Schwartz G, de Bono J et al, 2007, Phase I and pharmacokinetic study of lapatinib in combination with capecitabine in patients with advanced malignancies. J Clin Oncol 25:3753–3758 Siegel-Lakhai WS, Beijnen JH, Vervenne WL et al, 2007, Phase I pharmacokinetic study of the safety and tolerability of lapatinib, GW572016, in combination with oxaliplatin/fluorouracil/ leucovorin, FOLFOX4, in patients with solid tumors. Clin Cancer Res 13:4495–4502 Midgley RS, Kerr DJ, Flaherty KT et al, 2007, A phase I and pharmacokinetic study of lapatinib in combination with infusional 5- fluorouracil, leucovorin and irinotecan. Ann Oncol 18:2025–2029 Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL, 1987, Human breast cancer: correlation of relapse and survival with amplification of the HER2/neu oncogene. Science 235:177–182 Magnifico A, Albano L, Campaner S et al, 2007, Protein kinase C determines HER2 fate in breast carcinoma cells with HER2 protein overexpression without gene amplification. Cancer Res 67:5308–5317 Vogel CL, Cobleigh MA, Tripathy D et al, 2002, Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J Clin Oncol 20:719–726 Hegde PS, Rusnak D, Bertiaux M, Alligood K, Strum J, Gagnon R, Gilmer TM, 2007, Delineation of molecular mechanisms of sensitivity to lapitinib in breast cancer cell lines using global gene expression profiles. Mol Cancer Ther 6:1629–1640 Moy B, Kirkpatrick P, Kar S, Goss P, 2007, Lapatinib. Nature Rev Drug Discovery 6:431–432 Xia W, Gerard CM, Liu L, Baudson NM, Ory TL, Spector NL, 2005, Combining lapatinib, GW572016), a small molecule inhibitor of ErbB1 and ErbB2 tyrosine kinases, with therapeutic anti-ErbB2 antibodies enhances apoptosis of ErbB2-overexpressing breast cancer cells. Oncogene 24:6213–6221 Spector N, Liu L, Gerard C et al, 2005, Combining lapatinib, GW572016, with anti-erbB2 antibodies elicits synergistic apoptotic effects in erbB2 overexpressing breast cancer cells. J Clin Oncol 23, 16S, pt1), abstract 557 Konecny GE, Pegram MD, Venkatesan N et al, 2006, Activity of the dual kinase inhibitor lapatinib, GW572016, against HER-2 overexpressing and trastuzumab-treated breast cancer cells. Cancer Res 66:1630–1639 Storniolo AM, Burris HA, Overmoyer B et al, 2005, A phase I, open-label study of the safety, tolerability and pharmacokinetics of lapatinib, GW572016, in combination with trastuzumab. Breast Cancer Res Treat 2005:94(suppl 1), abstract 1073 Palmieri D, Gril BM, Herring J et al, 2007, Lapatinib prevents the metastatic colonization of EGFR+ and HER2+ breast cancer cells in the brain. Breast Cancer Res Treat suppl 1, abstract 210 Geyer CE, Forster J, Lindquist D et al, 2006, Lapatinib plus capecitabine for HER2-positive advanced breast cancer. New Engl J Med 355:2733–2743 Lin NU, Carey LA, Liu MC et al, 2006, Phase II trial of lapatinib for brain metastases in patients with HER2+ breast cancer. J Clin Oncol 24:18S, abstract 503 Perez EA et al, 2006, Cardiac safety experience in 3127 patients treated with lapatinib. Ann Oncol 17(Suppl 9):A1420 Cameron D, Casey M, Press M et al, 2008, A phase III randomized comparison of lapatinib plus capecitabine versus capecitabine alone in women with advanced breast cancer that has progressed on trastuzumab: updated efficacy and biomarker analyses. Breast Cancer Res Treat, in press), DOI 10.1007/ s10549-007-9885-0 Tuma RS, 2007, Lapatinib moves forward in inflammatory and early HER2-positive breast cancer trials. J Natl Cancer Inst 99:348–349 Spector NL, Blackwell K, Hurley J et al, 2006, EGF103009, a phase II trial of lapatinib monotherapy in patients with relapsed/ refractory inflammatory breast cancer, IBC): clinical activity and biologic predictors of response. J Clin Oncol ASCO Annual Meeing Procs Part I 24:18S, abstract 502 Cristofanilli M, Boussen H, Baselga J et al, 2006, A phase II combination study of lapatinib and paclitaxel as a therapy in patients with newly diagnosed inflammatory breast cancer, IBC). Breast Cancer Res Treat 100(suppl 1):S5, abstract 1 Nahta R, Yuan LXH, Zhang B, Kobayashi R, Esteva FJ, 2005, Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer Res 65:1118–1125 Nahta R, Yuan LXH, Du Y, Esteva FJ, 2007, Lapatinib induces apoptosis in trastuzumab-resistant breast cancer cells: effects on insulin-like growth factor I signaling. Mol Cancer Ther 6:667–674 Xia W, Bacus S, Hegde P et al, 2006, A model of acquired autoresistance to a potent ErbB2 tyrosine kinase inhibitor and a therapeutic strategy to prevent its onset in breast cancer. Proc Natl Acad Sci USA 103:7795–7800 DiGiovanna MP, Chakraborty A, 2006, Combinations of HER2, estrogen receptor, ER, and IGF-I receptor, IGF1R, inhibitors induce apoptosis in breast cancer cells: dramatic effects of HER2 inhibitors on non-overexpressing cells. Proc Am Assoc Cancer Res 47:1226 Witters LM, Scherle PA, Friedman SM et al, 2007, Inhibition of breast cancer growth with the combination of lapatinib and an ADAM protease inhibitor. Breast Cancer Res Treat 106(suppl 1), abstract 1117 Piccart-Gebhart MJ, Perez EA, Baselga J, 2007, ALTTO, Adjuvant Lapatinib and/or Trastuzumab Treatment Optimisation, study, BIG 2-06/N063D/EGF106708): a phase III study for HER2-overexpressing breast cancer, BC). Breast 16(suppl 1): S46, abstract P118 Frassoldati A, Guarneri V, Cagossi K et al, 2007, LET-LOB: preoperative letrozole plus lapatinib or placebo in hormonereceptor positive HER2 negative operable breast cancer. Preliminary report of activity and cardiac tolerability. Breast Cancer Res Treat, suppl 1), abstract 2087 McHugh LA, Kriajevska M, Mellon JK, Griffiths TR, 2007, Combined treatment of bladder cancer cell lines with lapatinib and varying chemotherapy regimens—evidence of schedule-dependent synergy. Urology 69:390–394 Agulnik M, Ezra WE, Cohen RB et al, 2007, Phase II study of lapatinib in recurrent or metastatic epidermal growth factor receptor and/or erbB2 expressing adenoid cystic carcinoma and non-adenoid cystic carcinoma malignant tumors of the salivary glands. J Clin Oncol 25:3978–3984 Widakowich C, de Azambuja E, Gil T et al, 2007, Molecular targeted therapies in breast cancer: where are we now? Internat J Biochem Cell Biol 39:1375–1387 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 1 EP 8 DI 10.1007/s12253-008-9018-z PG 8 ER PT J AU Kozakowski, N Soleiman, A Pammer, J AF Kozakowski, Nicolas Soleiman, Afschin Pammer, Johannes TI BMI-1 Expression is Inversely Correlated with the Grading of Renal Clear Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BMI-1; Renal cell carcinoma; Differentiation; Stem cell ID BMI-1; Renal cell carcinoma; Differentiation; Stem cell AB BMI-1 regulates cell proliferation and differentiation, is involved in stem cell maintenance and can act as an oncogene. We investigated BMI-1 expression in healthy normal kidney and in 77 renal tumours by immunohistochemistry, and correlated it with tumour differentiation. BMI-1 could regularly be demonstrated in distal tubules and in Bowman’s capsule, whereas it was mostly lacking in proximal tubules, indicating that it may rather be a differentiation marker of different renal cell populations than a stem cell marker. In contrast to previous studies demonstrating a correlation between BMI-1 expression and malignancy, we showed that its expression was inversely correlated with the differentiation grade of clear cell carcinoma. Furthermore, despite their different biologies, BMI-1 was strongly expressed in both papillary carcinomas and oncocytomas. Thus, in renal clear cell carcinomas BMI-1 is rather a differentiation marker lost in carcinomas with high malignancy than an oncogene involved in tumour progression. C1 [Kozakowski, Nicolas] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1097 Vienna, Austria. [Soleiman, Afschin] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1097 Vienna, Austria. [Pammer, Johannes] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1097 Vienna, Austria. RP Pammer, J (reprint author), Medical University of Vienna, Department of Pathology, 1097 Vienna, Austria. EM johannes.pammer@meduniwien.ac.at CR Jacobs JJ, Kieboom K, Marino S et al, 1999, The oncogene and Polycomb-group gene BMI-1 regulates cell proliferation and senescence through the ink4a locus. Nature 397:164–168 Jacobs JJ, Scheijen B, Voncken JW et al, 1999, BMI-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Mycinduced apoptosis via INK4a/ARF. Genes Dev 13:2678–2690 Kim JH, Yoon S, Kim CN et al, 2004, The BMI-1 oncoprotein is overexpressed in human colorectal cancer and correlates with the reduced p16INK4a/p14ARF proteins. Cancer Lett 203:217–224 Liu S, Dontu G, Mantle ID et al, 2006, Hedgehog signaling and BMI-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res 66:6063–6071 Raaphorst F, 2003, Self-renewal of hematopoietic and leukemic stem cells: a central role for the Polycomb-group gene BMI-1. Trends Immunol 24:522–524 Muller J, Gaunt S, Lawrence PA, 1995, Function of the Polycomb protein is conserved in mice and flies. Development 121:2847– 2852 Lessard J, Schumacher A, Thorsteinsdottir U et al, 1999, Functional antagonism of the Polycomb-Group genes eed and BMI1 in hemopoietic cell proliferation. Genes Dev 13:2691–2703 Van Kemenade FJ, Raaphorst FM, Blokzijl T et al, 2001, Coexpression of BMI-1 and EZH2 polycomb-group proteins is associated with cycling cells and degree of malignancy in B-cell non-Hodgkin lymphoma. Blood 7:3896–3901 Leung C, Lingbeek M, Shakhova O et al, 2004, BMI1 is essential for cerebellar development and is overexpressed in human medulloblastomas. Nature 428:337–341 Pirrotta V, 1997, PcG complexes and chromatin silencing. Curr Opin Genet Dev 2:249–258 Saurin AJ, Shiels C, Williamson J et al, 1998, The human Polycomb group complex associates with pericentromeric heterochromatin to form a novel nuclear domain. J Cell Biol 142:887– 898 Bea S, Tort F, Pinyol M et al, 2001, BMI-1 gene amplification and overexpression in hematological malignancies occur mainly in mantle cell lymphomas. Cancer Res 61:2409–2412 Kim JH, Yoon S, Jeong SH et al, 2004, Overexpression of BMI-1 oncoprotein correlates with axillary lymph node metastases in invasive ductal breast cancer. Breast 13:383–388 Breuer RH, Snijders P, Sutedja GT et al, 2005, Expression of the p16(INK4a, gene product, methylation of the p16(INK4a, promoter region and expression of the polycomb-group gene BMI-1 in squamous cell lung carcinoma and premalignant endobronchial lesions. Lung Cancer 48(3):299–306 Van Leenders GJ, Dukers D, Hessels D et al, 2007, Polycombgroup oncogenes EZH2, BMI1, and RING1 are overexpressed in prostate cancer with adverse pathologic and clinical features. Eur Urol 52(2):455–63 Van Galen JC, Muris J, Oudejans JJ et al, 2007, Expression of the polycomb-group gene BMI1 is related to an unfavourable prognosis in primary nodal DLBCL. J Clin Pathol 60:167–172 Cohen HT, Macgovern FJ, 2005, Renal-cell carcinoma. N Engl J Med 353:2477–2490 Martel CL, Lara PN, 2003, Renal cell carcinoma: current status and future directions. Crit Rev Oncol Hematol 45:177–190 Reya T, Morrison S, Clarke MF et al, 2001, Stem cells, cancer, and cancer stem cells. Nature 414:105–111 Valk-Lingbeek ME, Bruggeman S, Van Lohuizen M, 2004, Stem cells and cancer: the Polycomb connection. Cell 118:409–418 Al-Awqati Q, Oliver J, 2002, Stem cells in the kidney. Kidney Int 61:387–395 Oliver JA, Maarouf O, Cheema FH et al, 2004, The renal papilla is a niche for adult kidney stem cells. J Clin Invest 114:795–804 Gupta S, Verfaillie C, Chmielewski D et al, 2006, Isolation and characterization of kidney-derived stem cells. J Am Soc Nephrol 17(11):3028–40 Fuhrman SA, Lasky L, Limas C, 1982, Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol 6:655–663 Sika-Paotonu D, Bethwaite P, McCredie MRE et al, 2006, Nucleolar grade but not Fuhrman grade is applicable to papillary renal cell carcinoma. Am J Surg Pathol 30:1091–1096 Delahunt B, Sika-Paotonu D, Bethwaite PB et al, 2007, Fuhrman grading is not appropriate for chromophobe renal cell carcinoma. Am J Surg Pathol 31:957–960 Lessard J, Sauvageau G, 2003, BMI-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature 423:255–260 Collins AT, Maitland NJ, 2006, Prostate cancer stem cells. Eur J Cancer 42:1213–1218 Sanchez-Beato M, Sanchez E, Gonzalez-Carrero J et al, 2006, Variability in the expression of polycomb proteins in different normal and tumoral tissues. A pilot study using tissue microarrays. Mod Pathol 19:684–694 Bussolati B, Bruno S, Grange C et al, 2005, Isolation of renal progenitor cells from adult human kidney. Am J Pathol 166:545–555 Sagrinati C, Netti G, Mazzinghi B et al, 2006, Isolation and characterization of multipotent progenitor cells from the Bowman's capsule of adult human kidneys. J Am Soc Nephrol 17: 2443– 2456 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 9 EP 13 DI 10.1007/s12253-008-9006-3 PG 5 ER PT J AU Szabo, E Korpos, Batmunkh, E Lotz, G Holczbauer, Kovalszky, I Deak, F Kiss, I Schaff, Zs Kiss, A AF Szabo, Erzsebet Korpos, Eva Batmunkh, Enkhjargal Lotz, Gabor Holczbauer, Agnes Kovalszky, Ilona Deak, Ferenc Kiss, Ibolya Schaff, Zsuzsa Kiss, Andras TI Expression of Matrilin-2 in Liver Cirrhosis and Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Matrilin-2; Basement membrane; Extracellular matrix; Hepatocellular carcinoma; Cirrhosis ID Matrilin-2; Basement membrane; Extracellular matrix; Hepatocellular carcinoma; Cirrhosis AB The recently described matrilin protein family is part of the extracellular matrix, their pathophysiological role as well as distribution in liver diseases, however, have not yet been studied. Considering that matrilins have been found to play role in cell growth and tissue remodeling, their possible involvement in carcinogenesis has been raised. The main objective of this study was to investigate the changes in matrilin-2 expression which is one of the main components of basement membranes. Thirty-five cases of surgically resected hepatocellular carcinomas, 35 corresponding surrounding liver tissues and 10 normal liver samples were used for the study. In 15 of 35 cases the tumor developed on the basis of cirrhosis. Matrilin-2 protein expression was detected in normal liver around bile ducts, portal blood vessels, while sinusoids were negative by immunohistochemistry. Cirrhotic surrounding tissue showed intensive matrilin-2 staining along the sinusoids. Tumorous neovasculature was found strongly positive by immunohistochemistry. No differences, however, were detected by morphometry regarding the amount of protein expression based on the grade of hepatocellular carcinomas. Real-time RT-PCR did not show significant differences in matrilin-2 mRNA expression between normal, cirrhotic and tumor samples. This suggests posttranslational modification of matrilin-2 manifesting in altered distribution in liver fibrosis. Our data indicate that matrilin-2 is a novel basement membrane component in the liver, which is synthetized during sinusoidal "capillarization" in cirrhosis and in hepatocellular carcinoma. This is the first report to describe the expression and distribution of matrilin-2 in human normal and cirrhotic liver as well as in hepatocellular carcinoma. C1 [Szabo, Erzsebet] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Korpos, Eva] University of Szeged, Department of Medical ChemistrySzeged, Hungary. [Batmunkh, Enkhjargal] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Holczbauer, Agnes] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Deak, Ferenc] University of Szeged, Department of Medical ChemistrySzeged, Hungary. [Kiss, Ibolya] University of Szeged, Department of Medical ChemistrySzeged, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM schaff@korb2.sote.hu CR Monto A, Wright TL, 2001, The epidemiology and prevention of hepatocellular carcinoma. Semin Oncol 28:441–449 Wu XZ, Chen D, Xie GR, 2006, Extracellular matrix remodeling in hepatocellular carcinoma: effects of soil on seed? Med Hypotheses 66:1115–1120 Damsky CH, Werb Z, 1992, Signal transduction by integrin receptors for extracellular matrix: cooperative processing of extracellular information. Curr Opin Cell Biol 4:772–781 Assoian RK, Marcantonio EE, 1997, The extracellular matrix as a cell cycle control element in atherosclerosis and restenosis. J Clin Invest 100:15–18 Ingber DE, Folkman J, 1989, How does extracellular matrix control capillary morphogenesis? Cell 58:803–805 Juliano RL, Haskill S, 1993, Signal transduction from the extracellular matrix. J Cell Biol 120:577–585 Droz D, Patly N, Paraf F, Chretien Y, Gogusev J, 1994, Composition of extracellular matrix and distribution of cell adhesion molecules in renal cell tumors. Lab Invest 71:710–718 Deak F, Piecha D, Bachrati C, Paulsson M, Kiss I, 1997, Primary structure and expression of matrilin-2, the closest relative of cartilage matrix protein within the von Willebrand factor type Alike module superfamily. J Biol Chem 272:9268–9274 Deak F, Wagener R, Kiss I, Paulsson M, 1999, The matrilins: a novel family of oligomeric extracellular matrix proteins. Matrix Biol 18:55–64 Belluoccio D, Trueb B, 1997, Matrilin-3 from chicken cartilage. FEBS Lett 415:212–216 Wagener R, Ehlen HW, Ko YP, Kobbe B, Mann HH, Sengle G, Paulsson M, 2005, The matrilins—adaptor proteins in the extracellular matrix. FEBS Lett 79:3323–3329 Paulsson M, Heinegard D, 1979, Matrix proteins bound to associatively prepared proteoglycans from bovine cartilage. Biochem J 183:539–545 Paulsson M, Heinegard D, 1981, Purification and structural characterization of a cartilage matrix protein. Biochem J 197:367–375 Piecha D, Muratoglu S, Morgelin M, Hauser N, Studer D, Kiss I, Paulsson M, Deak F, 1999, Matrilin-2, a large, oligomeric matrix protein, is expressed by a great variety of cells and forms fibrillar networks. J Biol Chem 274:13353–13361 Klatt AR, Paulsson M, Wagener R, 2002, Expression of matrilins during maturation of mouse skeletal tissues. Matrix Biol 21:289– 296 Piecha D, Hartmann K, Kobbe B, Haase I, Mauch C, Krieg T, Paulsson M, 2002, Expression of matrilin-2 in human skin. J Invest Dermatol 119:38–43 Segat D, Frie C, Nitsche PD, Klatt AR, Piecha D, Korpos E, Deak F, Wagener R, Paulssson M, Smyth N, 2000, Expression of matrilin-1, -2 and -3 in developing mouse limbs and heart. Matrix Biol 19:649–655 Szabo E, Lodi C, Korpos E, Batmunkh E, Rottenberger Z, Deak F, Kiss I, Tokes AM, Lotz G, Laszlo V, Kiss A, Schaff Z, Nagy P, 2007, Expression of matrilin-2 in oval cells during rat liver regeneration. Matrix Biol 26:554–560 Ishak K, Baptista A, Bianchi L, Callea F, De Groote J, Gudat F, Denk H, Desmet V, Korb G, MacSween RN, 1995, Histological grading and staging of chronic hepatitis. J Hepatol 22:696–699 Korpos E, Molnar A, Papp P, Kiss I, Orosz L, Deak F, 2005, Expression pattern of matrilins and other extracellular matrix proteins characterize distinct stages of cell differentiation during antler development. Matrix Biol 24:124–135 Merz H, Malisius R, Mannweiler S, Zhou R, Hartmann W, Orscheschek K, Moubayed P, Feller AC, 1995, ImmunoMax. A maximized immunohistochemical method for the retrieval and enhancement of hidden antigens. Lab Invest 73:149–156 Mannhalter C, Koizar D, Mitterbauer G, 2000, Evalution of RNA isolation methods and reference genes for RT-PCR analyses of rare target RNA. Clin Lab Med 38:171–177 Pfaffl MW, Horgan GW, Dempfle L, 2002, Relative expression software tool, REST, for group-wise comparison and statistical analysis of relative expression results in real-time PCR. Nucleic Acids Res 1:30–36 Sharma MK, Watson MA, Lyman M, Perry A, Aldape KD, Deak F, Gutmann DH, 2006, Matrilin-2 expression distinguishes clinically relevant subsets of pilocytic astrocytoma. Neurology 66:127–130 Kin M, Torimura T, Ueno T, Inuzuka S, Tanikawa K, 1994, Sinusoidal capillarization in small hepatocellular carcinoma. Pathology International 44:771–778 Mates L, Korpos E, Deak F, Liu Z, Beier DR, Aszodi A, Kiss I, 2002, Comparative analysis of the mouse and human genes, Matn2 and MATN2, for matrilin-2, a filament-forming protein widely distributed in extracellular matrices. Matrix Biol 21:163–174 Shiojiri N, Sugiyama Y, 2004, Immunolocalization of extracellular matrix components and integrins during mouse liver development. Hepatology 40:346–355 Tatrai P, Dudas J, Batmunkh E, Mathe M, Zalatnai A, Schaff Z, Ramadori G, Kovalszky I, 2006, Agrin, a novel basement membrane component in human and rat liver, accumulates in cirrhosis and hepatocellular carcinoma. Lab Invest 86:1149– 1160 Batmunkh E, Tatrai P, Szabo E, Lodi Cs, Holczbauer A, Paska Cs, Kupcsulik P, Kiss A, Schaff Zs, Kovalszky I, 2007, Comparison of the expression of agrin, a basement membrane heparan sulfate proteoglycan, in cholangiocarcinoma and hepatocellular carcinoma. Hum Pathol 38:1508–1515 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 15 EP 22 DI 10.1007/s12253-008-9005-4 PG 8 ER PT J AU Appel, LMM Edelweiss, IM Fleck, J Rivero, FL Rivoire, AW Monego, IH dos Reis, R AF Appel, L M Marcia Edelweiss, I Maria Fleck, James Rivero, F Luis Rivoire, A Waldemar Monego, I Heleusa dos Reis, Ricardo TI p53 and BCL-2 as Prognostic Markers in Endometrial Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial cancer; p53; BCL-2; Prognosis ID Endometrial cancer; p53; BCL-2; Prognosis AB The objective of this study was to verify the frequency of p53 and BCL-2 immunohistochemical expression in patients with endometrial carcinoma and to correlate it with histological factors (histological type, tumor grade, depth of myometrial invasion, lymph node involvement and surgical staging) and survival. Forty-eight patients with endometrial carcinoma who were submitted to primary surgical treatment were assessed. p53 and BCL-2 immunohistochemical expression was determined using paraffin blocks containing the tumor area. p53 and BCL-2 expression was detected in 39.6% and 58.3% of the tumors, respectively. No significant difference was found regarding the frequency of p53 expression when analyzing histological type (33.3% in endometrioid tumors, 58.3% in non-endometrioid tumors; p=0.176), depth of myometrial invasion (p=0.632) and surgical staging (I—11.1%, II—66.7%, III—57.1%; p=0.061). p53 expression was significantly more frequent in undifferentiated tumors (p=0.007) and in those showing lymph node involvement (p=0.030). Univariate analysis showed a positive association with death (RR, 3.358; CI, 1.386–8.134; p=0.005) and short-term survival. The present study did not reveal any correlation between BCL-2 expression and histopathologic markers or survival. In conclusion, this study showed that p53 expression is directly correlated with undifferentiated tumors, lymph-node involvement and risk of death. On the other hand, BCL-2 expression was not correlated with any known histological factors. C1 [Appel, L M Marcia] Medical Post-Graduate Program, School of Medicine, Federal University of Rio Grande do Sul, Hospital de Clinicas de Porto Alegre, Department of Gynecology and Obstetrics, Rua Comendador Caminha, 128/902, CEP 90430-030 Porto Alegre, Rio Grande do Sul, Brazil. [Edelweiss, I Maria] Santa Casa Hospital Complex, Santa Rita Hospital, Department of PathologyPorto Alegre, Rio Grande do Sul, Brazil. [Fleck, James] Medical Post-Graduate Program, School of Medicine, Federal University of Rio Grande do Sul, Hospital de Clinicas de Porto Alegre, Department of Clinical OncologyPorto Alegre, Rio Grande do Sul, Brazil. [Rivero, F Luis] Santa Casa Hospital Complex, Santa Rita Hospital, Department of PathologyPorto Alegre, Rio Grande do Sul, Brazil. [Rivoire, A Waldemar] Medical Post-Graduate Program, School of Medicine, Federal University of Rio Grande do Sul, Hospital de Clinicas de Porto Alegre, Department of Gynecology and Obstetrics, Rua Comendador Caminha, 128/902, CEP 90430-030 Porto Alegre, Rio Grande do Sul, Brazil. [Monego, I Heleusa] Medical Post-Graduate Program, School of Medicine, Federal University of Rio Grande do Sul, Hospital de Clinicas de Porto Alegre, Department of Gynecology and Obstetrics, Rua Comendador Caminha, 128/902, CEP 90430-030 Porto Alegre, Rio Grande do Sul, Brazil. [dos Reis, Ricardo] Medical Post-Graduate Program, School of Medicine, Federal University of Rio Grande do Sul, Hospital de Clinicas de Porto Alegre, Department of Gynecology and Obstetrics, Rua Comendador Caminha, 128/902, CEP 90430-030 Porto Alegre, Rio Grande do Sul, Brazil. RP Appel, LMM (reprint author), Medical Post-Graduate Program, School of Medicine, Federal University of Rio Grande do Sul, Hospital de Clinicas de Porto Alegre, Department of Gynecology and Obstetrics, CEP 90430-030 Porto Alegre, Brazil. EM marciaappel@terra.com.br CR Instituto Nacional do Cancer., 2006, Estimativas da Incidencia e mortalidade por cancer no Brasil. 2003. http://www.inca.gov.br. Accessed May 2 Hacker NF, 2000, Uterine cancer. In: Berek JS and Hacker NF, eds, Practical gynecologic oncology, 3rd ed. Lippincott Williams & Wilkins, Philadelphia, pp 407–456 Axtell LM, Myers MH, 1978, Contrasts in survival of black and white cancer patients. J Natl Cancer Inst 60:1209–1215 Greenlee RT, Murray T, Bolden S, Wingo PA, 2000, Cancer statistics, 2000. CA Cancer J Clin 50:7–33 Mariani A, Sebo TJ, Katzmann JA, Keeney GL, Roche PC, Lesnick TG, Podratz KC, 2000, Pretreatment assessment of prognostic indicators in endometrial cancer. Am J Obstet Gynecol 182:1535–1544 Ohkouchi T, Sakuragi N, Watari H, Nomura E, Todo Y, Yamada H, Fujimoto S, 2002, Prognostic significance of Bcl-2, p53 overexpression, and lymph node metastasis in surgically staged endometrial carcinoma. Am J Obstet Gynecol 187:353–359 Geisler JP, Wiemann MD, Zhou Z, Miller GA, Geisler HE, 1996, p53 as a prognostic indicator in endometrial cancer. Gynecol Oncol 61:245–248 Geisler JP, Geisler HE, Wiemann MD, Zhou Z, Miller GA, Crabtree W, 1999, p53 expression as a prognostic indicator of 5- year survival in endometrial cancer. Gynecol Oncol 74:468–471 Geisler JP, Geisler HE, Wiemann MC, Zhou Z, Miller GA, Crabtree W, 1998, Lack of bcl-2 persistence: an independent prognostic indicator of poor prognosis in endometrial carcinoma. Gynecol Oncol 71:305–307 Sakuragi N, Ohkouchi T, Hareyama H, Ikeda K, Watari H, Fujimoto T, Kuwabara M, Yamamoto R, Sagawa T, Fujino T, Fujimoto S, 1998, Bcl-2 expression and prognosis of patients with endometrial carcinoma. Int J Cancer 79:153–158 Nishioka H, Hiasa Y, Hayashi I, Kitahori Y, Konishi N, Sugimura M, 1993, Immunohistochemical detection of p53 oncoprotein in human oral squamous cell carcinomas and leukoplasias: comparison with proliferating cell nuclear antigen staining and correlation with clinicopathological findings. Oncology 50:426–429 Kounelis S, Kapranos N, Kouri E, Coppola D, Papadaki H, Jones M, 2000, Immunohistochemical profile of endometrial adenocarcinoma: a study of 61 cases and review of the literature. Mod Pathol 13:379–388 Kohler MF, Carney P, Dodge R, Soper J, Clarke-Pearson DL, Marks JR, Berchuck A, 1996, p53 overexpression in advancedstage endometrial adenocarcinoma. Am J Obstet Gynecol 175:1246–1252 Morsi HM, Leers MP, Radespiel-Troger M, Bjorklund V, Kabarity HE, Nap M, Jager W, 2000, Apoptosis, bcl-2 expression, and proliferation in benign and malignant endometrial epithelium: an approach using multiparameter flow cytometry. Gynecol Oncol 77:11–17 Marone M, Ferrandina G, Macchia G, Mozzetti S, 2000, Bcl-2, Bax, Bcl-xl and Bcl-xs expression in neoplastic and normal endometrium. Oncology 58:161–168 Coppola D, Fu L, Nicosia SV, Kounelis S, Jones M, 1998, Prognostic significance of p53, bcl-2, vimentin, and S100 proteinpositive Langerhans cells in endometrial carcinoma. Human Pathol 29:455–462 Yamauchi N, Sakamoto A, Uozaki H, Iihara K, Machinami R, 1996, Immunohistochemical analysis of endometrial adenocarcinoma for bcl-2 and p53 in relation to expression of sex steroid receptor and proliferative activity. Int J Gynecol Pathol 15:202–208 Taskin M, Lallas TA, Barber HR, Shevchuk MM, 1997, bcl-2 and p53 endometrial adenocarcinoma. Mod Pathol 10:728–734 Saegusa M, Okayasu I, 1997, Bcl-2 is closely correlated with favorable prognostic factors and inversely associated with p53 protein accumulation in endometrial carcinomas: immunohistochemical and polymerase chain reaction/loss of heterozygosity findings. J Cancer Res Clin Oncol 123:429–434 College of American Pathologists, 2006, http://www.cap.org. Home>reference and Publications> Cancer Protocols and Checklists. Accessed 22 May Giatromanolaki A, Sivridis E, Koukourakis MI, Harris AL, Gatter KC, 1998, Bcl-2 and p53 expression in stage I endometrial carcinoma. Anticancer Res 18:3689–3694 van Slooten HJ, van der Vijver MJ, van der Velde CJH, van Dierendonck JH, 1998, Loss of Bcl-2 in invasive breast cancer is associated with high rates of cell death, but also with increased proliferative activity. Br J Cancer 77:789–796 Bonneefoy-Berard N, Aouacheria A, Verschelde C, Quemeneur L, Marcais A, Marvel J, 2004, Control of proliferation by Bcl-2 family members. Biochim Biophys Acta 1644:159–168 Boyd J, 1996, Molecular biology in the clinicopathologic assessment of endometrial carcinoma subtypes—editorial. Gynecol Oncol 61:163–165 Ambros RA, Sherman ME, Zahm CM, Bitterman P, Kurman RJ, 1995, Endometrial intraepithelial carcinoma: a distinctive lesion specifically associated with tumors displaying serous differentiation. Hum Pathol 26:1260–1267 Sherman ME, Bur ME, Kurman RJ, 1995, p53 in endometrial cancer and its putative precursors: evidence for diverse pathways of tumorigenesis. Hum Pathol 26:1268–1274 Spiegel GW, 1995, Endometrial carcinoma in situ in postmenopausal women. Am J Surg Pathol 19:417–432 Burton JL, Wells M, 1998, Recent advances in the histopathology and molecular pathology of carcinoma of the endometrium— review. Histopathology 33:297–303 Sakuragi N, Hirai A, Tada M, Yamada H, Yamamoto R, Fujimoto S, et al, 2001, Dominant-negative mutation of p53 tumor suppressor gene in endometrial carcinoma. Gynecol Oncol 83:485–490 Kohler MF, Kerns BJ, Humprey PA, Marks JR, Bast RC, Berchuck A, 1993, Mutation and overexpression of p53 in early-stage epithelial ovarian cancer. Obstet Gynecol 81:643–650 Soong R, Knowles S, Williams KE, Hammond IG, Wysocki SJ, Iacopetta BJ, 1996, Overexpression of p53 protein is an independent prognostic indicator in human endometrial carcinoma. Br J Cancer 74:562–567 Oreskovic S, Babic D, Kalafatic D, Barisic D, Beketic-Oreskovic LA, 2004, Significance of immunohistochemical determination of steroid receptors, cell proliferation factor Ki-67 and protein p53 in endometrial carcinoma. Gynecol Oncol 93:34–40 Tashiro H, Isacson C, Levine R, Kurman RJ, Cho KR, Hedrick L, 1997, p53 gene mutations are common in uterine serous carcinoma and occur early in their pathogenesis. Am J Pathol 150:177–185 Miyashita T, Krajewski S, Krajewska M, Wang HG, Lin HK, Liebermann DA, Hoffman B, Reed JC, 1994, Tumor suppressor p53 is a regulator of bcl-2 and bax gene expression in vitro and in vivo. Oncogene 9:1799–1805 Bonfitto VL, de Angelo Andrade LA, 2003, p53, estrogen and progesterone receptors in diagnostic curettage for endometrial adenocarcinoma and their correlation with morphological data and disease stage at hysterectomy. Sao Paulo Med J 121:163–166 Kohler MF, Berchuck A, Davidoff AM, Humphrey PA, Dodge RK, Iglehart JD, Soper JT, Clarke-Pearson DL, Bast RC Jr, Marks JR, 1992, Overexpression and mutation of p53 in endometrial carcinoma. Cancer Res 52:1622–1627 Hamel NW, Sebo TJ, Wilson TO, Keeney GL, Roche PC, Suman VJ, Hu TC, Podratz KC, 1996, Prognostic value of p53 and proliferation cell nuclear antigen expression in endometrial carcinoma. Gynecol Oncol 62:192–198 Ito K, Watanabe K, Nasim S, Sasano H, Sato S, Yajima A, Silverberg SG, Garrett CT, 1994, Prognostic significance of p53 overexpression in endometrial cancer. Cancer Res 54:4667–4670 Derossi DR, Ito K, Couto Filho J, Bacchi CE, 2003, Avaliacao da expressao da proteina bcl-2 no carcinoma de mama: estudo em puncao aspirativa por agulha fina; correlacao com grau histologico em especimes cirurgicos correspondentes. J Bras Patol Med Lab 39:229–235 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 23 EP 30 DI 10.1007/s12253-008-9000-9 PG 8 ER PT J AU Herszenyi, L Sipos, F Galamb, O Solymosi, N Hritz, I Miheller, P Berczi, L Molnar, B Tulassay, Zs AF Herszenyi, Laszlo Sipos, Ferenc Galamb, Orsolya Solymosi, Norbert Hritz, Istvan Miheller, Pal Berczi, Lajos Molnar, Bela Tulassay, Zsolt TI Matrix Metalloproteinase-9 Expression in the Normal Mucosa–Adenoma–Dysplasia–Adenocarcinoma Sequence of the Colon SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adenoma; Colorectal cancer; Dysplasia; Matrix metalloproteinase 9; Polymerase chain reaction ID Adenoma; Colorectal cancer; Dysplasia; Matrix metalloproteinase 9; Polymerase chain reaction AB It has been proposed that matrix metalloproteinases (MMPs) play a role in tumor invasion. We determined protein expression of matrix metalloproteinase-9 (MMP-9) in colorectal cancer (CRC), corresponding normal mucosa and colorectal adenomas. For confirmation of immunohistochemical results MMP-9 TaqMan RT-PCR analysis was performed. Expression of MMP-9 was determined on paraffin embedded biopsy sections by immunohistochemistry in 31 CRC patients (from cancer tissue and corresponding normal mucosa) and in 30 patients with adenoma (nine adenomas with high grade of dysplasia). MMP-9 immunostaining was determined semiquantitatively. For Taqman RT-PCR analyses normal mucosa (n=5), adenoma without (n=6) and with high grade dysplasia (n=7) and CRC (n=10) were investigated. Statistical analysis with ANOVA, LSD test and correlation analysis were performed. P value of <0.05 was considered significant. The MMP-9 expression in CRC was significantly higher compared to adenomas or the normal mucosa (P=0.001). Significantly higher expression of MMP-9 has been observed in adenomas with high grade dysplasia compared to other adenomas or normal colon (P<0.001). Diffuse strong MMP-9 expression was present in tumor as well as in stromal cells. In adenoma samples, dysplastic epithelial cells showed moderate intensive cytoplasmic MMP-9 expression, with a clear-cut differentiation between dysplastic and nondysplastic areas. Staining intensity correlated with the grade of CRC. We demonstrate a significantly higher expression of MMP-9 in adenoma with high grade dysplasia—CRC sequence as compared to normal tissue. The over-expression of MMP-9 strongly suggests its association with colorectal carcinogenesis. C1 [Herszenyi, Laszlo] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, 1088 Budapest, Hungary. [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, 1088 Budapest, Hungary. [Galamb, Orsolya] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. [Solymosi, Norbert] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, 1088 Budapest, Hungary. [Hritz, Istvan] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, 1088 Budapest, Hungary. [Miheller, Pal] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, 1088 Budapest, Hungary. [Berczi, Lajos] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, 1088 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, 1088 Budapest, Hungary. RP Herszenyi, L (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM hersz@bel2.sote.hu CR Berger AC, Sigurdson ER, LeVoyer T, et al, 2005, Colon cancer survival is associated with decreasing ratio of metastatic to examined lymph nodes. J Clin Oncol 23:8706–8712 Liang H, Wang XN, Wang BG, et al, 2006, Prognostic factors of young patients with colon cancer after surgery. World J Gastroenterol 12:1458–1462 Fuszek P, Horvath HC, Speer G, et al, 2006, Location and age at onset of colorectal cancer in Hungarian patients between 1993 and 2004. The high number of advanced cases supports the need for a colorectal cancer screening program in Hungary. Anticancer Res 26:531–537 Liotta LA, Stetler-Stevenson WG, 1991, Tumor invasion and metastasis: an imbalance of positive and negative regulation. Cancer Res 51:S5054–S5059 Dano K, Behrendt N, Hoyer-Hansen G, et al, 2005, Plasminogen activations and cancer. Thromb Haemost 93:676–681 Herszenyi L, Plebani M, Carraro P, et al, 1997, Impaired fibrinolysis and increased protease levels in gastric and duodenal mucosa of patients with active duodenal ulcer. Am J Gastroenterol 92:843–847 Plebani M, Herszenyi L, Cardin R, et al, 1995, Cysteine and serine proteases in gastric cancer. Cancer 76:367–375 Plebani M, Herszenyi L, Carraro P, et al, 1997, Urokinase-type plasminogen activator receptor in gastric cancer: tissue expression and prognostic role. Clin Exp Metastasis 15:418–425 Herszenyi L, Plebani M, Carraro P, et al, 1999, The role of cysteine and serine proteases in colorectal cancer. Cancer 86:1135–1142 Farinati F, Herszenyi L, Plebani M, et al, 1996, Increased levels of cathepsin B and L, urokinase-type plasminogen activator and inhibitor type-1 as an early event in gastric carcinogenesis. Carcinogenesis 17:2581–2587 Johnson LL, Dyer R, Hupe DJ, 1998, Matrix metalloproteinases. Curr Opin Chem Biol 2:466–471 Stamenkovic I, 2003, Extracellular matrix remodelling: the role of matrix metalloproteinases. J Pathol 200:448–464 Mysliwiec AG, Ornstein DL, 2002, Matrix metalloproteinases in colorectal cancer. Clin Colorectal Cancer 1:208–219 Sato H, Okada Y, SeikiM(1997)Membrane-type metalloproteinases, MT-MMPs, in cell invasion. Thromb Haemost 78:497–500 Nelson AR, Fingleton B, Rothenberg ML, et al, 2000, Matrix metalloproteinases: biologic activity and clinical implications. J Clin Oncol 18:1135–1149 Sier CF, Kubben FJ, Ganesh S, et al, 1996, Tissue levels of matrix metalloproteinase MMP-2 and MMP-9 are related to the overall survival of patients with gastric carcinomas. Br J Cancer 74: 413–417 Matsuyama Y, Takao S, Aikou T, 2002, Comparison of matrix metalloproteinases expression between primary tumors with or without liver metastasis in pancreatic and colorectal carcinomas. J Surg Oncol 80:105–110 Herszenyi L, Hritz I, Pregun I, et al, 2007, Alterations of glutathione S-transferase and matrix metalloproteinase-9 expressions are early events in the esophageal carcinogenesis. World J Gastroenterol 13:676–682 Saito K, Takeha S, Shiba K, et al, 2000, Clinicopathologic significance of urokinase receptor- and MMP-9 positive stromal cells in human colorectal cancer: functional multiplicity of matrix degradation on hematogeneous metastasis. Int J Cancer 86:24–29 Collins HM, Morris TM, Watson SA, 2001, Spectrum of matrix metalloproteinase expression in primary metastatic colon cancer: relationship to the tissue inhibitors of metalloproteinases and membrane-type-1-matrix metalloproteinase. Br J Cancer 84:1664– 1670 Roeb E, Dietrich CG, Winograd R, et al, 2001, Activity and cellular origin of gelatinases in patients with colon and rectal carcinoma: differential activity of matrix metalloproteinase-9. Cancer 92:2680–2691 Waas ET, Lomme RM, deGroot J, et al, 2002, Tissue levels of active matrix metalloproteinase-2 and -9 in colorectal cancer. Br J Cancer 86:1876–1883 Murray D, Morrin M, McDonnel S, 2004, Increased invasion and expression of MMP-9 in human colorectal cell lines by a CD44- dependent mechanism. Anticancer Res 24:489–494 Kim TD, Song KS, Li G, et al, 2006, Activity and expression of urokinase-type plasminogen activator and matrix metalloproteinases in human colorectal cancer. BMC Cancer 6:211. DOI 10.1186/1471- 2407/6/211 Thiefin G, Dupont A, Guillou PJ, et al, 2007, Beneficial influence of microsatellite instability on gelatinase-tissue inhibitors of metalloproteinase balance in colorectal cancer. Anticancer Res 27:583–588 Sardinha TC, Nogueras JJ, Xiong H, et al, 2000, Membrane-type 1 matrix metalloproteinase mRNA expression in colorectal cancer. Dis Colon Rectum 43:389–395 Papadopoulou S, Scorilas A, Arnogianaki N, et al, 2001, Expression of gelatinase-A, MMP-2, in human colon cancer and normal colon mucosa. Tumour Biol 22:383–389 Leeman MF, McKay JA, Murray GI, 2002, Matrix metalloproteinase 13 activity is associated with poor prognosis in colorectal cancer. J Clin Pathol 55:758–762 Pesta M, Holubec L, Topolcan O, et al, 2005, Quantitative estimation of matrix metalloproteinase 2 and 7, MMP-2, MMP-7, and tissue inhibitors of matrix metalloproteinases 1 and 2, TIMP- 1, TIMP-2, in colorectal carcinoma tissue samples. Anticancer Res 25:3387–3391 Zeng ZS, Huang Y, Cohen AM, et al, 1996, Prediction of colorectal cancer relapse and survival via tissue RNA levels of matrix metalloproteinase-9. J Clin Oncol 14:3133–3140 Baker EA, Bergin FG, Leaper DJ, 2000, Matrix metalloproteinases, their tissue inhibitors and colorectal cancer staging. Br J Surg 87:1215–1221 Curran S, Dundas SR, Buxton J, et al, 2004)Matrixmetalloproteinase/ tissue inhibitors of matrix metalloproteinase phenotype identifies poor prognosis colorectal cancers. Cancer Res 10:8229–8234 Illemann M, Bird N, Majeed A, et al, 2006, MMP-9 is differentially expressed in primary human colorectal adenocarcinomas and their metastases. Mol Cancer Res 4:293–302 Kirman I, Jain S, Cekic V, et al, 2006, Altered plasma matrix metalloproteinase-9/tissue inhibitor of matrix, corrected, metalloproteinase- 1 concentration during the early postoperative period in patients with colorectal cancer. Surg Endosc 20:482–486 Islekel H, Oktay G, Terzi C, et al, 2007, Matrix metalloproteinase- 9, -3 and tissue inhibitor of matrix metalloproteinase-1 in colorectal cancer: relationship to clinicopathological variables. Cell Biochem Funct 25:433–441 Dukes CE, 1932, The classification of cancer of the rectum. J Pathol 35:323–332 Turnbull RB, Kyle K, Watson FR, et al, 1967, Cancer of the colon: the influence of the no-touch isolation on the survival rates. Ann Surg 166:420–427 Sipos F, Galamb O, Herszenyi L, et al, 2007, Elevated insulin-like growth factor 1 receptor, hepatocyte growth factor receptor and telomerase expression in mild ulcerative colitis. Scand J Gastroenterol 23:1–10 Bernhard EJ, Gruber SB, Muschel RJ, 1994, Direct evidence linking expression of matrix metalloproteinase 9, 92kDa gelatinase/ collagenase, to the metastatic phenotype in transformed rat embryo cells. Proc Natl Acad Sci USA 91:4293–4297 Legrand C, Gilles C, Zahm JM, et al, 1999, Airway epithelial cell migration dynamics. MMP-9 role in cell-extracellular matrix remodelling. J Cell Biol 146:517–529 Tang Y, Nakada MT, Kesavan P, et al, 2005, Extracellular matrix metalloproteinase inducer stimulates tumor angioegenesis by elevating vascular endothelial cell growth factor and matrix metalloproteinases. Cancer Res 65:3193–3199 Mook OR, Frederiks WM, Van Noorden CJF, 2004, The role of gelatinases in colorectal cancer progression and metastasis. Biochim Biophys Acta 1705:69–89 Zeng ZS, Guillem JG, 1996, Colocalisation of matrix metalloproteinase- 9-mRNA and protein in human colorectal stromal cells. Br J Cancer 74:1161–1167 Lubbe WJ, Zhou ZY, Fu W, et al, 2006, Tumor epithelial cell matrix metalloproteinase 9 is a target for antimetastatic therapy in colorectal cancer. Clin Cancer Res 12:1876–1882 Gao Q, Meijer MJ, Kubben FJ, et al, 2005, Expression of matrix metalloproteinases-2 and -9 in intestinal tissue of patients with inflammatory bowel diseases. Dig Liver Dis 37:584–592 McKaig BC, McWilliams D, Watson SA, et al, 2003, Expression and regulation of tissue inhibitor of metalloproteinase-1 and matrix metalloproteinases by intestinal myofibroblasts in inflammatory bowel disease. Am J Pathol 162:1355–1360 Nosho K, Yoshida M, Yamamoto H, et al, 2005, Association of Ets-related transcriptional E1AF expression with overexpression of matrix metalloproteinases, COX-2 and iNOS in the early stage of colorectal carcinogenesis. Carcinogenesis 26:892–899 Rath T, Roderfeld M, Graf J, et al, 2006, Enhanced expression of MMP-7 and MMP 13 in inflammatory bowel disease: a precancerous potential? Inflamm Bowel Dis 12:1025–1035 Harada K, Hiraoka S, Kato J, et al, 2007, Genetic and epigenetic alterations of Ras signalling pathway in colorectal neoplasia: analysis based on tumour clinicopathological features. Br J Cancer 97:1425–1431 Hashimoto K, Shimizu Y, Suehiro Y, et al, 2008, Hypermethylation status of APC inversely correlates with the presence of submucosal invasion in laterally spreading colorectal tumors.Mol Carcinog 47:1–8 Model F, Osborn N, Ahlquist D, et al, 2007, Identification and validation of colorectal neoplasia-specific methylation markers for accurate classification of disease. Mol Cancer Res 5:153–163 Holten-Andersen MN, Christensen IJ, Nielsen HJ, et al, 2002, Total levels of tissue inhibitor of metalloproteinases 1 in plasma yield high diagnostic sensitivity and specificity in patients with colon cancer. Clin Cancer Res 8:156–164 Pesta M, Topolcan O, Holubec L Jr, et al, 2007, Clinicopathological assessment and quantitative estimation of the matrix metalloproteinases MMP-2 and MMP-7 and the inhibitors TIMP-1 and TIMP-2 in colorectal carcinoma tissue samples. Anticancer Res 27:1863–1867 Frederiksen C, Lykke J, Christensen IJ, et al, 2007, Tissue inhibitor of metalloproteinase-1 levels in plasma from tumour arteries and veins of patients with rectal cancer. Scand J Clin Lab Invest 67:545–552 Ogata Y, Matono K, Sasatomi T, et al, 2005, The MMP-9 expression determined the efficacy of postoperative adjuvant chemotherapy using oral fluoropyrimidines in stage II or III colorectal cancer. Cancer Chemother Pharmacol 57:577–583 Sorensen NM, Bystrom P, Christensen IJ, et al, 2007, TIMP-1 is significantly associated with objective response and survival in metastatic colorectal cancer patients receiving combination of irinotecan, 5-fluorouracil, and folinic acid. Clin Cancer Res 13:4117–4122 Coussens LM, Fingleton B, Matrisian LM, 2002, Matrix metalloproteinase inhibitors and cancer-Trials and tribulations. Science 295:2387–2392 Vihinen P, Ala-Aho R, Kahari VM(2005)Matrix metalloproteinases as therapeutic targets in cancer. Curr Cancer Drug Targets 5:203–220 Burg-Roderfeld M, Roderfeld M, Wagner S, et al, 2007, MMP- 9-hemopexin domain hampers adhesion and migration of colorectal cancer cells. Int J Oncol 30:985–992 Pavlaki M, Zucker S, 2003, Matrix metalloproteinase inhibitors, MMPIs): the beginning of phase I or the termination of phase III clinical trials. Cancer Metastasis Rev 22:177–203 van Smarle S, van Vliet A, Sollie F, et al, 2005, Safety, tolerability and pharmacokinetics of oral S-3304, a novel matrix metalloproteinase inhibitor, in single and multiple dose escalation studies in healthy volunteers. Int J Pharmacol Ther 43:282–293 Chiappori AA, Eckhardt SG, Bukowski R, et al, 2007, A phase I pharmacokinetic and pharmacodynamic study of s-3304, a novel matrix metalloproteinase inhibitor, in patients with advanced and refractory solid tumors. Clin Cancer Res 13:2091–2099 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 31 EP 37 DI 10.1007/s12253-008-9004-5 PG 7 ER PT J AU Hiraishi, Y Wada, T Nakatani, K Tojyo, I Matsumoto, T Kiga, N Negoro, K Fujita, Sh AF Hiraishi, Yukihiro Wada, Takeshi Nakatani, Ken Tojyo, Itaru Matsumoto, Takashi Kiga, Norifumi Negoro, Kenji Fujita, Shigeyuki TI EGFR Inhibitor Enhances Cisplatin Sensitivity of Oral Squamous Cell Carcinoma Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Epidermal growth factor receptor (EGFR); EGFR inhibitor; Oral squamous cell carcinoma (OSCC); Cisplatin-resistant OSCC cell line; Cisplatin sensitivity and resistance ID Epidermal growth factor receptor (EGFR); EGFR inhibitor; Oral squamous cell carcinoma (OSCC); Cisplatin-resistant OSCC cell line; Cisplatin sensitivity and resistance AB Epidermal growth factor receptor (EGFR) is involved in multiple aspects of cancer cell biology. EGFR has already been identified as an important target for cancer therapy, with various kinds of EGFR inhibitors currently used in treatment of several human cancers. Recently, EGFR and its downstream signaling pathways were identified as being associated with cisplatin sensitivity. In addition, EGFR inhibitors have shown significant promise for patients who failed cisplatin-based therapy. In this study, we investigated whether treatment with an EGFR inhibitor improves cisplatin sensitivity in oral squamous cell carcinoma (OSCC) cell lines. The effects of a combination of AG1478, a specific EGFR tyrosine kinase inhibitor, with cisplatin were evaluated in cultured OSCC cell lines and cisplatin-resistant sublines. Higher expression of EGFR and p-EGFR was found in the two cisplatin-resistant cell lines compared with the corresponding parental cell lines. In addition, augmented inhibition of OSCC cell growth by the combination of AG1478 with cisplatin was found in both cell lines. These results suggest that the combination of an EGFR inhibitor and cisplatin may be useful as a rational strategy for the treatment of patients with oral cancer with acquired cisplatin resistance. C1 [Hiraishi, Yukihiro] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 Kimiidera, 641-8509 Wakayama, Japan. [Wada, Takeshi] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 Kimiidera, 641-8509 Wakayama, Japan. [Nakatani, Ken] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 Kimiidera, 641-8509 Wakayama, Japan. [Tojyo, Itaru] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 Kimiidera, 641-8509 Wakayama, Japan. [Matsumoto, Takashi] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 Kimiidera, 641-8509 Wakayama, Japan. [Kiga, Norifumi] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 Kimiidera, 641-8509 Wakayama, Japan. [Negoro, Kenji] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 Kimiidera, 641-8509 Wakayama, Japan. [Fujita, Shigeyuki] Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 811-1 Kimiidera, 641-8509 Wakayama, Japan. RP Hiraishi, Y (reprint author), Wakayama Medical University, Department of Oral and Maxillofacial Surgery, 641-8509 Wakayama, Japan. EM hiraishi@wakayama-med.ac.jp CR Salomon DS, Brandt R, Ciardiello F et al, 1995, Epidermal growth factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol 19:183–232 Hiraishi Y, Wada T, Nakatani K et al, 2006, Immunohistochemical expression of EGFR and p-EGFR in oral squamous cell carcinomas. Pathol Oncol Res 12:87–91 Huang SM, Harari PM, 1999, Epidermal growth factor receptor inhibition in cancer therapy: Biology, rationale and preliminary clinical results. Invest New Drugs 17:259–269 Prenzel N, Fischer OM, Streit S et al, 2001, The epidermal growth factor receptor family as a central element for cellular signal transduction and diversification. Endocr Relat Cancer 8:11– 31 Gibbs JB, 2000, Anticancer drug targets: growth factors and growth factor signaling. J Clin Invest 105:9–13 Mendelsohn J, 2001, The epidermal growth factor receptor as a target of cancer therapy. Endocr Relat Cancer 8:3–9 Herbst RS, Hong WK, 2002, IMC-C225, anti-epidermal growth factor receptor monoclonal antibody for treatment of head and neck cancer. Semin Oncol 29:18–30 Fukuoka M, Yano S, Giaccone G et al, 2003, Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer. J Clin Oncol 21:2237–2246 Mendelsohn J, Baselga J, 2003, Status of epidermal growth factor receptor antagonists in the biology and treatment of cancer. J Clin Oncol 21:2787–2799 SaltzL B, Meropol NJ, Loehrer PJ et al, 2004, Phase II trial of cetuximab in patients with refractory colorectal cancer that express the epidermal growth factor receptor. J Clin Oncol 22:1201–1208 Christen RD, Hom DK, Porter DC et al, 1990, Epidermal growth factor regulates the in vitrosensitivity of human ovarian carcinoma cells to cisplatin. J Clin Invest 86:1632–1640 Aboud-Pirak E, Hurwitz E, Pirak ME et al, 1988, Efficacy of antibodies to epidermal growth factor receptor against KB carcinoma in vitro and in nude mice. J Natl Cancer Inst 80:1605–1611 Dai Q, Ling YH, Lia M et al, 2005, Enhanced sensitivity to the HER1/epidermal growth factor receptor tyrosine kinase inhibitor erlotinib hydrochloride in chemotherapy resistant tumor cell lines. Clin Cancer Res 11:1572–1578 Benhar M, Engelberg D, Levitzki A, 2002, Cisplatin-induced activation of the EGF receptor. Oncogene 21:8723–8731 Bourhis J, 2005, New approaches to enhance chemotherapy in SCCHN. Ann Oncol 16:20–24 Chan JK, Pham H, You XJ et al, 2005, Suppression of ovarian cancer cell tumorigenicity and evasion of Cisplatin resistance using a truncated epidermal growth factor receptor in a rat model. Cancer Res 65:3243–3248 Nakamura M, Nakatani K, Uzawa K et al, 2005, Establishment and characterization of a cisplatin-resistant oral squamous cell carcinoma cell line, H-1R. Oncol Rep 14:1281–1286 Nakatani K, Nakamura M, Uzawa K et al, 2005, Establishment and gene analysis of a cisplatin-resistant cell line, Sa-3R, derived from oral squamous cell carcinoma. Oncol Rep 13:709–714 Downward J, Parker P, Waterfield MD, 1984, Autophosphorylation sites on the epidermal growth factor receptor. Nature 311:483–485 Olayioye MA, Neve RM, Lane HA et al, 2000, The ErbB signaling network: receptor heterodimerization in development and cancer. EMBO J 19:3159–3167 Wang X, Martindale JL, Holbrook NJ, 2000, Requirement for ERK activation in cisplatin-induced apoptosis. J Biol Chem 275:39435–39443 Benhar M, Dalyot I, Engelberg D et al, 2001, Enhanced ROS production in oncogenically transformed cells potentiates c-Jun Nterminal kinase and p38 mitogen-activated protein kinase activation and sensitization to genotoxic stress. Mol Cell Biol 21:6913– 6926 Deschesnes RG, Huot J, Valerie K et al, 2001, Involvement of p38 in apoptosis-associated membrane blebbing and nuclear condensation. Mol Biol Cell 12:1569–1582 Chen Z, Zhang X, Li M et al, 2004, Simultaneously targeting epidermal growth factor receptor tyrosine kinase and cyclooxygenase- 2, an efficient approach to inhibition of squamous cell carcinoma of the head and neck. Clin Cancer Res 10:5930– 5939 El-Marjou A, Delouvee A, Thiery JP et al, 2000, Involvement of epidermal growth factor receptor in chemically induced mouse bladder tumor progression. Carcinogenesis 21:2211– 2218 Wakeling AE, 2002, Epidermal growth factor receptor tyrosine kinase inhibitors. Curr Opin Pharmacol 2:382–387 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 39 EP 43 DI 10.1007/s12253-008-9020-5 PG 5 ER PT J AU Khademi, B Razmkhah, M Erfani, N Gharagozloo, M Ghaderi, A AF Khademi, Bijan Razmkhah, Mahboobeh Erfani, Nasrollah Gharagozloo, Marjan Ghaderi, Abbas TI SDF-1 and CCR5 Genes Polymorphism in Patients with Head and Neck Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Head and neck cancer; SDF-1; CCR5; Polymorphism; Metastasis ID Head and neck cancer; SDF-1; CCR5; Polymorphism; Metastasis AB The frequency of SDF1–3′A and CCR5Δ32 in patients with head and neck cancer were determined in this study. The frequencies of alleles and genotypes of SDF-1 and CCR5 were assessed by PCR method in 156 patients with malignant head and neck cancer, 125 (80.1%) cases with squamous cell carcinoma and 31 (19.9%) cases with salivary gland tumors and compared with 262 age–sex matched healthy control individuals. SDF-1 genotypes in patients with SCC of head and neck, but not with salivary gland tumors, showed a statistically significant difference compared to the normal group (P<0.005 for SCC and P=0.3 for salivary gland tumors). There were no significant differences in the frequencies of SDF1–3′A allele, CCR5 genotypes and alleles between patients and controls. Based on the present study SDF1–3́A may be associated with the susceptibility of patients to SCC of head and neck cancer. C1 [Khademi, Bijan] Shiraz University of Medical Sciences, Department of ENTShiraz, Iran. [Razmkhah, Mahboobeh] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Erfani, Nasrollah] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Gharagozloo, Marjan] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Ghaderi, Abbas] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. RP Razmkhah, M (reprint author), Shiraz University of Medical Sciences, Institute for Cancer Research, Shiraz, Iran. EM razmkhahm@sums.ac.ir CR Estecio MR, Youssef EM, Rahal P, Fukuyama EE, Gois-Filho JF, Maniglia JV, Goloni-Bertollo EM, Issa JP, Tajara EH, 2006, LHX6 is a sensitive methylation marker in head and neck carcinomas. Oncogene 29: kotet, oldalszam Marcus B, Arenberg D, Lee J, Kleer C, Chepeha DB, Schmalbach CE, Islam M, Paul S, Pan Q, Hanash S, Kuick R, Merajver SD, Teknos TN, 2004, Prognostic factors in oral cavity and oropharyngeal squamous cell carcinoma. Cancer 101:2779–2787 Uchida D, Begum NM, Tomizuka Y, Bando T, Almofti A, Yoshida H, Sato M, 2004, Acquisition of lymph node, but not distant metastatic potentials, by the overexpression of CXCR4 in human oral squamous cell carcinoma. Lab Invest 84:1538–1546 Luker KE, Luker GD, 2005, Functions of CXCL12 and CXCR4 in breast cancer. Cancer Lett 23:1–12 Balkwill F, 2003, Chemokine biology in cancer. Semin Immunol 15:49–55 Bleul CC, Fuhlbrigge RC, Casasnovas JM, Aiuti A, Springer TA, 1996, A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1, SDF-1). J Exp Med 184:1101–1109 Aiuti A, Webb IJ, Bleul C, Springer T, Gutierrez-Ramos JC, 1997, The chemokine SDF-1 is a chemoattractant for human CD34 + hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34 + progenitors to peripheral blood. J Exp Med 185:111–120 Perissinotto E, Cavalloni G, Leone F, Fonsato V, Mitola S, Grignani G, Surrenti N, Sangiolo D, Bussolino F, Piacibello W, Aglietta M, 2005, Involvement of chemokine receptor 4/stromal cell-derived factor 1 system during osteosarcoma tumor progression. Clin Cancer Res 11:490–497 Kang H, Watkins G, Douglas -Jones A, Mansel RE, Jiang WG, 2005, The elevated level of CXCR4 is correlated with nodal metastasis of human breast cancer. Breast 14:360–367 Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegui E, Zlotnik A, 2001, Involvement of chemokine receptors in breast cancer metastasis. Nature 410:50–56 Lee BC, Lee TH, Avraham S, Avraham HK, 2004, Involvement of the chemokine receptor CXCR4 and its ligand stromal cell-derived factor 1alpha in breast cancer cell migration through human brain microvascular endothelial cells. Mol Cancer Res 2:327–338 Samara GJ, Lawrence DM, Chiarelli CJ, Valentino MD, Lyubsky S, Zucker S, Vaday GG, 2004, CXCR4-mediated adhesion and MMP-9 secretion in head and neck squamous cell carcinoma. Cancer Lett 214:231–241 Uchida D, Begum NM, Almofti A, Nakashiro K, Kawamata H, Tateishi Y, Hamakawa H, Yoshida H, Sato M, 2003, Possible role of stromal-cell-derived factor-1/CXCR4 signaling on lymph node metastasis of oral squamous cell carcinoma. Exp Cell Res 290:289–302 Almofti A, Uchida D, Begum NM, Tomizuka Y, Iga H, Yoshida H, Sato M, 2004, The clinicopathological significance of the expression of CXCR4 protein in oral squamous cell carcinoma. Int J Oncol 25:65–71 Hwang JH, Hwang JH, Chung HK, Kim DW, Hwang ES, Suh JM, Kim H, You KH, Kwon OY, Ro HK, Jo DY, Shong M, 2003, CXC chemokine receptor 4 expression and function in human anaplastic thyroid cancer cells. J Clin Endocrinol Metab 88:408–416 Mori T, Doi R, Koizumi M, Toyoda E, Ito D, Kami K, Masui T, Fujimoto K, Tamamura H, Hiramatsu K, Fujii N, Imamura M, 2004, CXCR4 antagonist inhibits stromal cell-derived factor 1- induced migration and invasion of human pancreatic cancer. Molecular Cancer Therapeutics 3:29–37 Sun YX, Schneider A, Jung Y, Wang J, Dai J, Wang J, Cook K, Osman NI, Koh-Paige AJ, Shim H, Pienta KJ, Keller ET, McCauley LK, Taichman RS, 2005, Skeletal localization and neutralization of the SDF-1(CXCL12)/CXCR4 axis blocks prostate cancer metastasis and growth in osseous sites in vivo. J Bone Miner Res 20:318–329 Ottaiano A, di Palma A, Napolitano M, Pisano C, Pignata S, Tatangelo F, Botti G, Acquaviva AM, Castello G, Ascierto PA, Iaffaioli RV, Scala S, 2005, Inhibitory effects of anti-CXCR4 antibodies on human colon cancer cells. Cancer Immunol Immunother 54:781–791 Watanabe MA, de Oliveira Cavassin GG, Orellana MD, Milanezi CM, Voltarelli JC, Kashima S, Covas DT, 2003, SDF-1 gene polymorphisms and syncytia induction in Brazilian HIV-1 infected individuals. Microb Pathog 35:31–34 Winkler C, ModiW, SmithMW, Nelson GW,Wu X, Carrington M, Dean M, Honjo T, Tashiro K, Yabe D, Buchbinder S, Vittinghoff E, Goedert JJ, O'Brien TR, Jacobson LP, Detels R, Donfield S, Willoughby A, Gomperts E, Vlahov D, Phair J, O'Brien SJ, 1998, Genetic restriction of AIDS pathogenesis by an SDF-1 chemokine gene variant. Science 118:681–688 Kimura R, Nishioka T, Ishida T, 2003, The SDF1-G801A polymorphism is not associated with SDF1 gene expression in Epstein–Barr virus-transformed lymphoblastoid cells. Genes Immun 4:356–361 Kawasaki E, Ide A, Abiru N, Kobayashi M, Fukushima T, Kuwahara H, Kita A, Uotani S, Yamasaki H, Eguchi K, 2004, Stromal cell-derived factor-1 chemokine gene variant in patients with type 1 diabetes and autoimmune thyroid disease. Ann N Y Acad Sci 1037:79–83 Dean M, Carington M, O'Brien SJ, 2002, Balanced polymorphism selected by genetic versus infectious human disease. Annu Rev Genomics Hum Genet 3:263–292 de Oliveira Cavassin GG, De Lucca FL, Delgado Andre N, Covas DT, Pelegrinelli Fungaro MH, Voltarelli JC, Watanabe MA, 2004, Molecular investigation of the stromal cell-derived factor-1 chemokine in lymphoid leukemia and lymphoma patients from Brazil. Blood Cells Mol Diseases 33:90–93 Razmkhah M, Talei AR, Doroudchi M, Khalili-Azad T, Ghaderi A, 2005, Stromal cell-derived factor-1, SDF-1, alleles and susceptibility to breast carcinoma. Cancer Lett 225:261–266 Razmkhah M, Doroudchi M, Ghayumi SM, Erfani N, Ghaderi A, 2005, cell-derived factor-1, SDF-1, gene and susceptibility of Iranian patients with lung cancer. Lung Cancer 49:311–315 Robinson SC, Scott KA, Wilson JL, Thompson RG, Proudfoot AE, Balkwill FR, 2003, A chemokine receptor antagonist inhibits experimental breast tumor growth. Cancer Res 63:8360–8365 Huang Y, Paxton WA, Wolinsky SM, 1996, The role of a mutant CCR5 allele in HIV-1 transmission and disease progression. Nat Med 2:1240 Zapico I, Coto E, Rodriguez A, Alvarez C, Torre JC, Alvarez V, 2000, CCR5, chemokine receptor-5, DNA-polymorphism influences the severity of rheumatoid arthritis. Genes Immun 1:288–289 Kalev I, Oselin K, Parlist P, Zilmer M, Rajasalu T, Podar T, Mikelsaar AV, 2003, CC-chemokine receptor CCR5-del32 mutation as a modifying pathogenesis factor in type I diabetes. J Diabetes Its Complicat 17:387–391 Miller SA, Dykes DD, Polesky HF, 1988, Simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16:1215 Voevodin A, Samilchuk E, Dashti S, 1999, Frequencies of SDF-1 chemokine, CCR-5, and CCR-2 chemokine receptor gene alleles conferring resistance to human immunodeficiency virus type 1 and AIDS in Kuwaitis. J Med Virol 58:54–58 Sandford AJ, Zhu S, Bai TR, Fitzgerald JM, Pare PD, 2001, The role of the C-C chemokine receptor-5 Delta 32 polymorphism in asthma and in the production of regulated on activation, normal T cells expressed and secreted. J Allergy Clin Immunol 108:69–73 Eisenhardt A, Frey U, Tack M, Rosskopf D, Lummen G, Rubben H, Siffert W, 2005, Expression analysis and potential functional role of the CXCR4 chemokine receptor in bladder cancer. Eur Urol 47:111–117 Kang H, Watkins G, Parr C, Douglas-Jones A, Mansel RE, Jiang WG, 2005, Stromal cell derived factor-1: its influence on invasiveness and migration of breast cancer cells in vitro, and its association with prognosis and survival in human breast cancer. Breast Cancer Res 7:402–410 Sun YX, Wang J, Shelburne CE, Lopatin DE, Chinnaiyan AM, Rubin MA, Pienta KJ, Taichman RS, 2003, Expression of CXCR4 and CXCL12, SDF-1, in human prostate cancers, PCa, in vivo. J Cell Biochem 89:462–473 Delgado MB, Clark-Lewis I, Loetscher P, Langen H, Thelen M, Baggiolini M, Wolf M, 2001, Rapid inactivation of stromal cellderived factor-1 by cathepsin G associated with lymphocytes. Eur J Immunol 31:699–707 Bonavia R, Bajetto A, Barbero S, Pirani P, Florio T, Schettini G, 2003, Chemokines and their receptors in the CNS: expression of CXCL12/SDF-1 and CXCR4 and their role in astrocyte proliferation. Toxicol Lett 139:181–189 Mohle R, Failenschmid C, Bautz F, Kanz L, 1999, Overexpression of chemokine receptor CXCR4 in B cell chronic lymphocytice leukemia is associated with increased functional response to stromal cell-derived factor-1, SDF-1). Leukemia 13:1954–1959 Bajetto A, Bonavia R, Barbero S, Schettini G, 2002, Characterization of chemokines and their receptors in the central nervous system: physiopathological implications. J Neurochem 82:1311– 1329 Lazarini F, Tham TN, Casanova P, Arenzana-Seisdedos F, Dubois- Dalcq M, 2003, Role of the alpha-chemokine stromal cell-derived factor, SDF-1, in the developing and mature central nervous system. Glia 42:139–148 Porcile C, Bajetto A, Barbieri F, Barbero S, Bonavia R, Biglieri M, Pirani P, Florio T, Schettini G, 2005, Stromal cell derived factor-1 alpha, SDF-1 alpha/CXCL12, stimulates ovarian cancer cell growth through the EGF receptor transactivation. Exp Cell Res 308:241–253 Hong X, Jiang F, Kalkanis SN, Zhang ZG, Zhang XP, de Carvalho AC, 2006, SDF-1 and CXCR4 are upregulated by VEGF and contribute to glioma cell invasion. Cancer Lett 236:39–45 Rabkin CS, Yang Q, Goedert JJ, Nguyen G, Mitsuya H, Sei S, 1999, Chemokine and chemokine receptor gene variants and risk of non-Hodgkin's lymphoma in human immunodeficiency virus- 1-infected individuals. Blood 93:1838–1842 Degerli N, Yilmaz E, Bardakci F, 2005, The delta32 allele distribution of the CCR5 gene and its relationship with certain cancers in a Turkish population. Clin Biochem 38:248–252 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 45 EP 50 DI 10.1007/s12253-008-9007-2 PG 6 ER PT J AU Kamory, E Olasz, J Csuka, O AF Kamory, Eniko Olasz, Judit Csuka, Orsolya TI Somatic APC Inactivation Mechanisms in Sporadic Colorectal Cancer Cases in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Allelic imbalance; APC; Mutation; Promoter hypermethylation; Sporadic colorectal cancer ID Allelic imbalance; APC; Mutation; Promoter hypermethylation; Sporadic colorectal cancer AB The role of germline inactivation of the adenomatosis polyposis coli (APC) gene in hereditary colorectal cancer is well known, being the most important cause of familial adenomatous polyposis (FAP) syndrome. Hereditary cases with germline mutations, however, account only for 5–10% of colorectal cancers. The somatic inactivation of this gene has also been observed in sporadic cases. In order to examine the inactivation mechanisms of the APC gene we screened 70 sporadic colorectal cancer cases (27 rectal, 43 intestinal) of different stages for promoter hypermethylation, allelic imbalance (AI) and somatic mutations. The presence of promoter hypermethylation was observed in 21 cases (30%). Fifteen of the examined tumors (21%) showed AI, and also 15 tumors (21%) carried at least one somatic mutation. Thirteen of the detected alterations were novel variations: seven frameshifts, four missense mutations and two polymorphisms. Biallelic inactivation was found in 15 patients (21%). These results suggest that the inactivation of the APC gene is very common in sporadic colorectal cancer, and the main inactivation mechanism of the APC gene is promoter hypermethylation. Allelic imbalance has the same frequency as mutations, and mutations in the APC gene are more common in the early stages and in tumors located in the rectum. C1 [Kamory, Eniko] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Olasz, Judit] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Csuka, Orsolya] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. RP Kamory, E (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, H-1122 Budapest, Hungary. EM eniko.kamory@gmail.com CR Fearon ER, Vogelstein B, 1990, A genetic model for colorectal tumorigenesis. Cell 61(5):759–767 Yang J, Zhang W, Evans PM et al, 2006, Adenomatous polyposis coli, APC, differently regulates beta-catenin phosphorylation and ubiquitination in colon cancer cells. J Biol Chem 281(26):17751– 17757 Senda T, Iizuka-Kogo A, Onouchi T et al, 2007, Adenomatous polyposis coli, APC, plays multiple roles in the intestinal and colorectal epithelia. Med Mol Morphol 40(2):68–81 Fearnhead NS, Britton MP, Bodmer WF, 2001, The ABC of APC. Hum Mol Genet 10(7):721–733 Lambertz S, Ballhausen WG, 1993, Identification of an alternative 5′ untranslated region of the adenomatous polyposis coli gene. Hum Genet 90(6):650–652 Thliveris A, Samowitz W, Matsunami N, 1994, Demonstration of promoter activity and alternative splicing in the region 5′ to exon 1 of the APC gene. Cancer Res 54(11):2991–2995 Esteller M, Sparks A, Toyota M et al, 2000, Analysis of adenomatous polyposis coli promoter hypermethylation in human cancer. Cancer Res 60(16):4366–4371 Xiong Z, Wu AH, Bender CM et al, 2001, Mismatch repair deficiency and CpG island hypermethylation in sporadic colon adenocarcinomas. Cancer Epidemiol Biomarkers Prev 10(7):799– 803 Jubb AM, Bell SM, Quirke P, 2001, Methylation and colorectal cancer. J Pathol 195(1):111–134 Nagase H, Nakamura Y, 1993, Mutations of the APC, adenomatous polyposis coli, gene. Hum Mutat 2(6):425–434 Varesco L, Gismondi V, James R et al, 1993, Identification of APC gene mutations in Italian adenomatous polyposis coli patients by PCR-SSCP analysis. Am J Hum Genet 52(2):280– 285 Wallis YL, Morton DG, McKeown CM et al, 1999, Molecular analysis of the APC gene in 205 families: extended genotypephenotype correlations in FAP and evidence for the role of APC amino acid changes in colorectal cancer predisposition. J Med Genet 36(1):14–20 Segditsas S, Tomlinson I, 2006, Colorectal cancer and genetic alterations in the Wnt pathway. Oncogene 25(57):7531–7537 Maniatis T, Fritsch EF, Sambrook J, 1982, Molecular cloning, a laboratory manual. Cold Spring Harbor Laboratory, New York, USA Frommer M, McDonald LE, Millar DS et al, 1992, A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci USA 89(5):1827–1831 Eads CA, Lord RV, Wickramasinghe K et al, 2001, Epigenetic patterns in the progression of esophageal adenocarcinoma. Cancer Res 61(8):3410–3418 Tomlinson IP, Lambros MB, Roylance RR, 2002, Loss of heterozygosity analysis: practically and conceptually flawed? Genes Chromosomes Cancer 34(4):349–353 Gyapay G, Morissette J, Vignal A et al, 1994, The 1993–94 Genethon human genetic linkage map. Nat Genet 7(2 Spec No): 246–339 Groden J, Thliveris A, Samowitz Wet al, 1991, Identification and characterization of the familial adenomatous polyposis coli gene. Cell 66(3):589–600 Miyoshi Y, Ando H, Nagase H et al, 1992, Germ-line mutations of the APC gene in 53 familial adenomatous polyposis patients. Proc Natl Acad Sci USA 89(10):4452–4456 Ng PC, Henikoff S, 2003, SIFT: predicting amino acid changes that affect protein function. Nucleic Acids Res 31(13):3812– 3814 Sunyaev S, Ramensky V, Koch I et al, 2001, Prediction of deleterious human alleles. Hum Mol Genet 10(6):591–597 Ferrer-Costa C, Orozco M, de la Cruz X, 2004, Sequence-based prediction of pathological mutations. Proteins 57(4):811–819 Arnold CN, Goel A, Niedzwiecki D et al, 2004, APC promoter hypermethylation contributes to the loss of APC expression in colorectal cancers with allelic loss on 5q. Cancer Biol Ther 3, 10):960–964 Thorstensen L, Lind GE, Lovig T et al, 2005, Genetic and epigenetic changes of components affecting the WNT pathway in colorectal carcinomas stratified by microsatellite instability. Neoplasia 7(2):99–108 Furukawa T, Konishi F, Masubuchi S et al, 2002, Densely methylated MLH1 promoter correlates with decreased mRNA expression in sporadic colorectal cancers. Genes Chromosomes Cancer 35(1):1–10 Prall F, Weirich V, Ostwald C, 2007, Phenotypes of invasion in sporadic colorectal carcinomas related to aberrations of the adenomatous polyposis coli, APC, gene. Histopathology 50, 3):318–330 Lamlum H, Ilyas M, Rowan A et al, 1999, The type of somatic mutation at APC in familial adenomatous polyposis is determined by the site of the germline mutation: a new facet to Knudson's ‘two-hit’ hypothesis. Nat Med 5(9):1071–1075 Laken SJ, Petersen GM, Gruber SB et al, 1997, Familial colorectal cancer in Ashkenazim due to a hypermutable tract in APC. Nat Genet 17(1):79–83 Mihalatos M, Apessos A, Dauwerse H et al, 2005, Rare mutations predisposing to familial adenomatous polyposis in Greek FAP patients. BMC Cancer 5(1):40–47 Chen-Shtoyerman R, Theodor L, Harmati E et al, 2003, Genetic analysis of familial colorectal cancer in Israeli Arabs. Hum Mutat 21(4):446–447 Gavert N, Yaron Y, NaimanT et al, 2002, Molecular analysis of the APC gene in 71 Israeli families: 17 novel mutations. Hum Mutat 19(6):664 Paul P, Letteboer T, Gelbert L et al, 1993, Identical APC exon 15 mutations result in a variable phenotype in familial adenomatous polyposis. Hum Mol Genet 2(7):925–931 Hadjisavvas A, Papasavva T, Loizidou M et al, 2006, Novel germline mutations in the APC gene of Cypriot patients with familial and sporadic adenomatous polyposis. Clin Genet 69(5): 404–409 Dundar M, Caglayan AO, Saatci C et al, 2007, How the I1307K adenomatous polyposis coli gene variant contributes in the assessment of risk of colorectal cancer, but not stomach cancer, in a Turkish population. Cancer Genet Cytogenet 177(2):95–97 Derks S, Postma C, Moerkerk PT et al, 2006, Promoter methylation precedes chromosomal alterations in colorectal cancer development. Cell Oncol 28(5–6):247–527 Goel A, Nagasaka T, Arnold CN et al, 2007, The CpG island methylator phenotype and chromosomal instability are inversely correlated in sporadic colorectal cancer. Gastroenterology 132(1): 127–138 Samowitz WS, Slattery ML, Sweeney C et al, 2007, APC mutations and other genetic and epigenetic changes in colon cancer. Mol Cancer Res 5(2):165–170 De Filippo C, Luceri C, Caderni G et al, 2002, Mutations of the APC gene in human sporadic colorectal cancers. Scand J Gastroenterol 37(9):1048–1053 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 51 EP 56 DI 10.1007/s12253-008-9019-y PG 6 ER PT J AU Kato, K Kawashiri, Sh Tanaka, A Noguchi, N Nakaya, H Hase, T Yamamoto, E AF Kato, Koroku Kawashiri, Shuichi Tanaka, Akira Noguchi, Natsuyo Nakaya, Hiromitsu Hase, Takashi Yamamoto, Etsuhide TI Predictive Value of Measuring p53 Labeling Index at the Invasive Front of Oral Squamous Cell Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral squamous cell carcinoma; Invasive front; p53; Labeling index ID Oral squamous cell carcinoma; Invasive front; p53; Labeling index AB Many studies have revealed the frequency of p53 abnormalities in oral cancer. However, it reports only on the relation between clinicopathological findings and p53 expression, and there is no study to examine the relation to the p53 labeling index (p53-LI). The purposes of this study were to examine the correlation between p53 labeling index (p53-LI) at the invasive front of oral squamous cell carcinomas (OSCC) and clinicopathological findings by immunohistochemical staining, and to evaluate clinical significance of measuring p53-LI at the invasive front of OSCC. Sixty-six biopsy specimens of OSCC were randomly selected. Patient age, gender, primary sites, T category, N category, degree of differentiation and mode of cancer invasion were analyzed. p53 expression did not correlate significantly with the clinical findings. However, significant differences were found between p53-LI and the degree of cell differentiation (p<0.05). The p53-LI of highgrade invasive tumors was significantly larger than that of low-grade invasive tumors (p<0.05). The overall survival rate (OS) among low-scoring p53-LI cases was 75.5% whereas that for high-scoring p53-LI cases was 40.6%. The disease-free survival rate (DFS) among low-scoring p53-LI cases was 39.5% whereas that for high-scoring p53-LI cases was 76.1%. Patients with low-scoring p53-LI had a significantly worse prognosis than those with among highscoring p53-LI (p<0.05). Consequently, the measurement of p53-LI at the invasive front of OSCC is significant as one of the indicators of prognosis. C1 [Kato, Koroku] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Japan. [Kawashiri, Shuichi] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Japan. [Tanaka, Akira] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Japan. [Noguchi, Natsuyo] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Japan. [Nakaya, Hiromitsu] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Japan. [Hase, Takashi] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Japan. [Yamamoto, Etsuhide] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Japan. RP Kato, K (reprint author), Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 920-8640 Kanazawa, Japan. EM k-koroku@oral.m.kanazawa-u.ac.jp CR Attardi LD, Lowem SW, Brugarolas J et al, 1996, Transcriptional activation by p53, but not induction of the p21 gene, is essential for oncogene-mediated apoptosis. EMBO J 15:3693–3701 Bankfalvi A, Piffko J, 2000, Prognostic and predictive factors in oral cancer: the role of the invasive tumour front. J Oral Pathol Med 29:291–298 Beenken SW, Krontiras H, Maddox WA et al, 1999, T1 and T2 squamous cell carcinoma of the oral tongue: prognostic factors and the role of elective lymph node dissection. Head Neck 21:124–130 Bryne M, 1998, Is the invasive front of an oral carcinoma the most important area for prognostication? Oral Dis 4:70–77 Cattaretti G, Rilke F, Andreola S et al, 1988, p53 expression in breast cancer. Int J Cancer 41:178–183 Ceccarelli C, Santini D, Chieco P et al, 2001, Quantitative P21WAF-1/P53 immunohistochemical analysis defines groups of primary invasive breast carcinoma with different prognostic indicators. Int J Cancer 95:128–134 Girod SC, Krueger G, Pape HD, 1993, p53 and Ki 67 expression in preneoplastic and neoplastic lesions of the oral mucosa. Int J Oral Maxillofac Surg 22:285–288 Hartwell L, 1992, Defects in a cell cycle checkpoint may be responsible for the genomic instability of cancer cells. Cell 71:543–546 Holm R, Skomedal H, Helland A et al, 1993, Immunohistochemical analysis of p53 protein overexpression in normal, premalignant, and malignant tissues of the cervix uteri. J Pathol 169:21–26 Iggo R, Gatter K, Bartek J et al, 1990, Increased expression of mutant forms of p53 oncogene in primary lung cancer. Lancet 335:675–679 Isabel BC, Chris J, Meijer LM et al, 2001, P53 immunoexpression in non-malignant oral mucosa adjacent to oral squamous cell carcinoma: potential consequences for clinical management. J Pathol 191:132–137 Jankovic R, Brankovic-Magic M, Radulovic S, 2004, The past and the future of cancer genetics. J Balk Union Oncol 9:347–354 Osaki T, Kimura T, Tatemoto Y et al, 2000, Risk factors of metastasis in oral squamous cell carcinomas. Oncology 58:137– 143 Osborne RJ, Merlo GR, Mitsudomi T et al, 1991, Mutations in the p53 gene in primary human breast cancers. Cancer Res 51:6194– 6198 Piffko J, Bankfalvi A, Tory K et al, 1998, Molecular assessment of p53 abnormalities at the invasive front of oral squamous cell carcinoma. Head Neck 20:8–15 Prochazkova J, Lichnovsky V, Kylarova D et al, 2004, Involvement of p53 and Bcl-2 family proteins in regulating programmed cell death and proliferation in human embryogenesis. Gen Physiol Biophys 23:209–229 Purdie CA, O’Grady J, Piris J et al, 1991, p53 expression in colorectal tumors. Am J Pathol 138:807–813 Shear M, Hawkins DM, Farr HW, 1976, The prediction of lymph node metastases from oral squamous carcinoma. Cancer 37:1901– 1907 Shin DM, Lee JS, Lippman SM et al, 1996, p53 expression: predicting recurrence and second primary tumors in head and neck squamous cell carcinoma. J Natl Cancer Inst 88:519–529 Shintani S, Yoshimata Y, Emilio AR et al, 1995, Overexpression of p53 is an early event in the tumourigenesis of oral squamous cell carcinomas. Anticancer Res 71:1248–1252 Vokes EE, Weichselbaum RR, Lippman SM et al, 1993, Head and neck cancer. N Engl J Med 328:184–194 Warnakulasuriya KA, Johnson NW, 1995, Expression of p53 mutant nuclear phosphoprotein in oral carcinoma and potentially malignant oral lesions. J Oral Pathol Med 24:337–342 Warnakulasuriya S, Jia C, Johnson N et al, 2000, p53 and Pglycoprotein expression are significant prognostic markers in advanced head and neck cancer treated with chemo/radiotherapy. J Pathol 191:33–38 Yamamoto E, Kohama G, Sunagawa H et al, 1983, Mode of invasion: bleomycin sensitivity, and clinical course in squamous cell carcinoma of the oral cavity. Cancer 51:2175–2180 Yamamoto E, Miyakawa A, Kohama G, 1984, Mode of invasion and lymph node metastasis in squamous cell carcinoma of oral cavity. Head Neck 6:939–947 Yamamoto E, Sunakawa H, Kohama G, 1983, Clinical course of diffuse invasive carcinoma of the tongue excised after bleomycin treatment. J Maxillofac Surg 11:269–274 Yamazaki Y, Chiba I, Hirai A et al, 2003, Radioresistance in oral squamous cell carcinoma with p53 DNA contact mutation. Am J Clin Oncol 26:24–29 Yonish-Rouach E, Resnitzky D, Lotem J et al, 1991, Wild-type p53 induces apoptosis of myeloid leukaemic cells that is inhibited by interleukin-6. Nature 352:345–347 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 57 EP 61 DI 10.1007/s12253-008-9022-3 PG 5 ER PT J AU Eros, N Karolyi, Zs Marschalko, M Karpati, S Matolcsy, A AF Eros, Nora Karolyi, Zsuzsanna Marschalko, Marta Karpati, Sarolta Matolcsy, Andras TI Clinical, Histopathological, Immunophenotypic and Molecular Analysis of 60 Patients with Cutaneous T-cell Infiltrates with Follow up of Indeterminate Cases to Identify T-cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cutaneous T-cell lymphoma; Mycosis fungoides; Cutaneous T-cell infiltrate; Histopathology; Immunophenotype; Molecular biology ID Cutaneous T-cell lymphoma; Mycosis fungoides; Cutaneous T-cell infiltrate; Histopathology; Immunophenotype; Molecular biology AB Diagnosis of primary cutaneous T-cell lymphomas, especially of mycosis fungoides could be difficult in early stage due to clinical and histopathological similarity to reactive inflammatory dermatoses. To assess diagnostic value of complex histological, immunophenotypic and Tcell receptor γ gene rearrangement analysis, skin biopsy specimen and peripheral blood samples of 60 patients with suspected cutaneous T-cell lymphoma were analyzed. Our results indicate clear distinction between reactive dermatoses (benign cases, n=31) and cutaneous T-cell lymphomas (lymphoma cases, n=17). As definite diagnosis was not obtained in a smaller group of patients (indeterminate cases, n=12), these patients were followed up. Repeated skin biopsy confirmed mycosis fungoides in 6/12 cases, however in 6/12 patients the diagnosis remained indeterminate. We concluded that careful and complex clinical follow up and repeated histopathological, immunophenotypic and molecular analysis is needed for an appropriate diagnosis in the assessment of early stage mycosis fungoides and uncertain clinical cases. C1 [Eros, Nora] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria str. 41, 1085 Budapest, Hungary. [Karolyi, Zsuzsanna] Semmelweis Hospital, Department of DermatologyMiskolc, Hungary. [Marschalko, Marta] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria str. 41, 1085 Budapest, Hungary. [Karpati, Sarolta] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria str. 41, 1085 Budapest, Hungary. [Matolcsy, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Eros, N (reprint author), Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, 1085 Budapest, Hungary. EM dr.eros@citromail.hu CR Bachelez H, 1999, The clinical use of molecular analysis of clonality in cutaneous lymphocytic infiltrates. Arch Dermatol 135:200–202 Bakels V, Oostveen JW, Putte SC, Meijer CJ, Willemze R, 1997, Immunophenotyping and gene rearrangement analysis provide additional criteria to differentiate between cutaneous T-cell lymphomas and pseudo-T-cell lymphomas. Am J Pathol 150:1941–1949 Burg G, Kempf W, Dummer R, 2001, Diagnostic signs of cutaneous lymphomas. J Eur Acad Dermatol Venereol 15:358–360 Delfau-Larue MH, Laroche L, Wechsler J, Lepage E, Lahet C, Asso-Bonnet M et al, 2000, Diagnostic value of dominant T-cell clones in peripheral blood in 363 patients presenting consecutively with a clinical suspicion of cutaneous lymphoma. Blood 96:2987– 2992 Dippel E, Klemke D, Hummel M, Stein H, Goerdt S, 2001, T-cell clonality of undetermined significance. Blood 98:247–248 Flaig MJ, Schuhmann K, Sander CA, 2000, Impact of molecular analysis in the diagnosis of cutaneous lymphoid infiltrates. Semin Cutan Med Surg 19:87–90 Holm N, Flaig MJ, Yazdi AS, Sander CA, 2002, The value of molecular analysis by PCR in the diagnosis of cutaneous lymphocytic infiltrates. J Cutan Pathol 29:447–452 Li N, Bhawan J, 2001, New insights into the applicability of Tcell receptor g gene rearrangement analysis in cutaneous T-cell lymphoma. J Cutan Pathol 28:412–418 Massone C, Kodama K, Kerl H, Cerroni L, 2005, Histopathologic features of early, patch, lesions of mycosis fungoides: a morphologic study on 745 biopsy specimens from 427 patients. Am J Surg Pathol 29:550–560 Muche MJ, Lukowsky A, Asadullah K, Gellrich S, Sterry W, 1997, Demonstration of frequent occurence of clonal T cells in the peripheral blood of patients with primary cutaneous T-cell lymphoma. Blood 90:1636–1642 Muche MJ, Lukowsky A, Heim J, Friedrich M, Audring H, Sterry W, 1999, Demonstration of frequent occurence of clonal T cells in the peripheral blood but not in the skin of patients with small plaque parapsoriasis. Blood 94:1409–1417 Muche MJ, Sterry W, Gellrich S, Rzany B, Audring H, Lukowsky A, 2003, Peripheral blood T-cell clonality in mycosis fungoides and nonlymphoma controls. Diagn Mol Pathol 12:142–150 Nagy M, Feher K, Laszlo T, Szomor A, Losonczy H, Kelenyi G et al, 1999, Analysis of the T-cell receptor g gene rearrangement in lymphoproliferative disorders using polymerase chain reaction. Orv Hetil 140:2441–2444 Pimpinelli N, Olsen EA, Santucci M, Vonderheid E, Haeffner AC, Stevens S et al, 2005, Defining early mycosis fungoides. J Am Acad Dermatol 53:1053–1063 Posnett DN, Sinha R, Kabak S, Russo C, 1994, Clonal populations of T cells in normal elderly humans: the T cell equivalent to “benign monoclonal gammopathy”. J Exp Med 179:609–618 Santucci M, Biggeri A, Feller AC, Massi D, Burg G, 2000, Efficacy of histologic criteria for diagnosing early mycosis fungoides. An EORTC Cutaneous Lymphoma Study Group investigation. Am J Surg Pathol 24:40–50 Smoller BR, Bishop K, Glusac E, Kim YH, Hendrickson M, 1995, Reassessment of histologic parameters in the diagnosis of mycosis fungoides. Am J Surg Pathol 19:1424–1430 Theodorou I, Delfau-Larue MH, Bigorgne C, Lahet C, Cochet G, Bagot M, 1995, Cutaneous T-cell infiltrates: analysis of T-cell receptor g gene rearrangement by polymerase chain reaction and denaturing gradient gel electrophoresis. Blood 86:305–310 Wood GS, 1998, Using molecular biologic analysis of T-cell receptor gene rearrangements to stage cutaneous T-cell lymphoma. Arch Dermatol 134:221–223 Wood GS, 2001, Analysis of clonality in cutaneous T cell lymphoma and associated diseases. Ann N YAcad Sci 941:26–30 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 63 EP 67 DI 10.1007/s12253-008-9014-3 PG 5 ER PT J AU Nagy, CsA Cserep, Zs Tolnay, E Nagykalnai, T Forster, T AF Nagy, Csaba Andras Cserep, Zsuzsanna Tolnay, Edina Nagykalnai, Tamas Forster, Tamas TI Early Diagnosis of Chemotherapy-induced Cardiomyopathy: a Prospective Tissue Doppler Imaging Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Anthracycline; Tissue Doppler imaging; Cardiotoxicity ID Anthracycline; Tissue Doppler imaging; Cardiotoxicity AB The aim of our study was to compare the applicability of the conventional echocardiography and a novel method, tissue Doppler imaging (TDI) in detection of late or subclinical cardiotoxicity following anthracycline chemotherapy in long-term follow up. Forty women (31 to 65 years) were enrolled, who had not received anthracyclines previously and had normal cardiac function. The control group consisted of 20 healthy persons of similar age range. In addition to standard echocardiographic measurements, each patient underwent specific measurements (Eseptum separation, pulmonary venous flow) as well. Furthermore, the myocardial velocity of numerous segments of the mitral anulus obtained with pulsed wave TDI was also detected over a two-year-long period. Systolic left ventricular function did not change significantly either in the study or in the control group. After one year, diastolic left ventricular function was impaired in 39 patients (97.5%), and 29 (72.5%) of these showed clear changes by means of the traditional E/A ratio and TDI. However, in ten patients (25%) the diastolic dysfunction could only be detected with TDI. At the end of the study diastolic dysfunction was detected in each patient, but in 13 patients (32.5%) the relaxation disorder could be revealed only with TDI. Detectable myocardial damage occurred in the study group as a result of anthracycline therapy. Our results confirmed our assumptions that TDI is a more precise and useful examination method than the traditional ones (E/A ratio or deceleration time) to demonstrate isolated diastolic dysfunction. TDI may become a regularly and more widely used noninvasive method to detect subclinical cardiotoxicity emerging after chemotherapy. C1 [Nagy, Csaba Andras] Uzsoki Municipal Hospital, 1st Department of Internal Medicine and Cardiology, Uzsoki u. 29, H-1145 Budapest, Hungary. [Cserep, Zsuzsanna] Semmelweis UniversityBudapest, Hungary. [Tolnay, Edina] Pest County Institute of Pulmonology, 2nd DepartmentTorokbalint, Hungary. [Nagykalnai, Tamas] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai KozpontBudapest, Hungary. [Forster, Tamas] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. RP Nagy, CsA (reprint author), Uzsoki Municipal Hospital, 1st Department of Internal Medicine and Cardiology, H-1145 Budapest, Hungary. EM nagycsaba@uzsoki.hu CR Alam M, Wardell J, Andersson E, Samad BA, Nordlander R, 2000, Effects of first myocardial infarction on left ventricular systolic and diastolic function with the use of mitral annular velocity determined by pulsed wave Doppler tissue imaging. J Am Soc Echocardiogr 13:343–352 Aranda JM Jr, Weston MW, Puleo JA, Fontanet HL, 1998, Effect of loading conditions on myocardial relaxation velocities determined by Doppler tissue imaging in heart transplant recipients. J Heart Lung Transplant 17:693–697 Billingham ME, 1984, Evaluation of anthracycline cardiotoxicity: predictive ability and functional correlation of endomyocardial biopsy. Cancer Treat Symp 3:71–76 Bu’Lock FA, Mott MG, Oakhill A, Martin RP, 1995, Left ventricular diastolic function after anthracycline chemotherapy in childhood: relation with systolic function, symptoms, and pathophysiology. Br Heart J 73:340–350 Burstein H, Winer E, 2000, Primary care for survivors of breast cancer. N Engl J Med 343:1086–1094 Canal P, Gamelin E, Vassal G, Robert J, 1998, Benefits of pharmacological knowledge in the design and monitoring of cancer chemotherapy. Pathol Oncol Res 4:171–178 Dandel M, Hummel M, Muller J, Wellnhofer E, Meyer R, Solowjowa N, Ewert R, Hetzer R, 2001, Reliability of tissue Doppler wall motion monitoring after heart transplantation for replacement of invasive routine screenings by optimally timed cardiac biopsies and catheterizations. Circulation 104(Suppl 1): I184–I191 Kantrowitz NE, Bristow MR, 1984, Cardiotoxicity of antitumor agents. Prog Cardiovasc Dis 27:195–200 Kapusta L, Thijssen JM, Groot-Loonen J, Antonius T, Mulder J, Daniels O, 2000, Tissue Doppler imaging in detection of myocardial dysfunction in survivors of childhood cancer treated with anthracyclines. Ultrasound Med Biol 26:1099–1108 Kapusta L, Thijssen JM, Groot-Loonen J, van Druten JA, Daniels O, 2001, Discriminative ability of conventional echocardiography and tissue Doppler imaging techniques for the detection of subclinical cardiotoxic effects of treatment with anthracyclines. Ultrasound Med Biol 27:1605–1614 Klein AL, Burstow DJ, Tajik AJ, Zachariah PK, Bailey KR, Seward JB, 1994, Effects of Age on left ventricular dimensions and filling dynamics in 117 normal persons. Mayo Clin Proc 69: 212–224 Krischer JP, Epstein S, Cuthbertson DD, Goorin AM, Epstein ML, Lipshultz SE, 1997, Clinical cardiotoxicity following anthracycline treatment for childhood cancer: the Pediatric Oncology Group experience. J Clin Oncol 15:1544–1552 Krupicka J, Markova J, Pohlreich D, Kozak T, Linkova H, Diehl V, 2002, German Hodgkin's Lymphoma Study Group. Echocardiographic evaluation of acute cardiotoxicity in the treatment of Hodgkin disease according to the German Hodgkin's Lymphoma Study Group. Leuk Lymphoma 43:2325–2329 Lefrak EA, Pitha J, Rosenheim S, Gottlieb JA, 1973, A clinicopathologic analysis of adriamycin cardiotoxicity. Cancer 32:302– 314 Lipshultz SE, Colan SD, Gelber RD, Perez-Atayde AR, Sallan SE, Sanders SP, 1991, Late cardiac effects of doxorubicin therapy for acute lymphoblastic leukemia in childhood. N Eng J Med 324:808– 815 Lipshultz SE, Colan SD, Walsh EP, Sanders SP, Sallan SP, 1990, Ventricular tachycardia and sudden unexplained death in late survivors of childhood malignancy treated with doxorubicin. Pediatr Res 27:145A Lipshultz SE, Lipsitz SR, Mone SM, Goorin AM, Sallan SE, Sanders SP, Orav EJ, Gelber RD, Colan SD, 1995, Female sex and drug dose as risk factors for late cardiotoxic effects of doxorubicin therapy for childhood cancer. N Eng J Med 333: 1359–1360 Nagueh SF, Mikati I, Kopelen HA, Middleton KJ, Quinones MA, Zoghbi WA, 1998, Doppler estimation of left ventricular filling pressure in sinus tachycardia. A new application of tissue doppler imaging. Circulation 98:1644–1650 Nagy AC, Tolnay E, Nagykalnai T, Forster T, 2006, Cardiotoxicity of anthracycline in young breast cancer female patients: the possibility of detection of early cardiotoxicity by TDI. Neoplasma 53:511–517 Nakayama K, Miyatake K, Uematsu M, Tanaka N, Kamakura S, Nakatani S, Yamazaki N, Yamagishi M, 1998, Application of tissue Doppler imaging technique in evaluating early ventricular contraction associated with accessory atrioventricular pathways in Wolff–Parkinson–White syndrome. Am Heart J 135:99–106 Nysom K, Colan SD, Lipshultz SR, 1998, Late cardiotoxicity following anthracycline therapy for childhood cancer. Prog Pediatr Cardiol 8:121–128 Pela G, Bruschi G, Cavatorta A, Manca C, Cabassi A, Borghetti A, 2001, Doppler tissue echocardiography: myocardial wall motion velocities in essential hypertension. Eur J Echocardiogr 2:108– 117 Rakowski H, Appleton C, Chan KL, Dumesnil JG, Honos G, Jue J, Koilpillai C, Lepage S, Martin RP, Mercier LA, O’Kelly B, Prieur T, Sanfilippo A, Sasson Z, Alvarez N, Pruitt R, Thompson C, Tomlinson C, 1996, Canadian consensus recommendation for the measurement and reporting of diastolic dysfunction by echocardiography. J Am Soc Echocardiol 9:736–760 Shimizu Y, Uematsu M, Shimizu H, Nakamura K, Yamagishi M, Miyatake K, 1998, Peak negative myocardial velocity gradient in early diastole as a noninvasive indicator of left ventricular diastolic function: comparison with transmitral flow velocity indices. J Am Coll Cardiol 32:1418–1425 Sogaard P, Hassager C, 2004, Tissue Doppler imaging as a guide to resynchronization therapy in patients with congestive heart failure. Curr Opin Cardiol 19:447–451 Steinberg JS, Cohen AJ, Wasserman AG, Cohen P, Ross AM, 1987, Acute arrhythmogenicity of doxorubicin administration. Cancer 60:1213–1218 Steinherz LJ, Steinherz PG, Tan C, 1995, Cardiac failure and dysrhythmias 6–19 years after anthracycline therapy: a series of 15 patients. Med Pediatr Oncol 24:352–361 Steinherz LJ, Steinherz PG, Tan CT, Heller G, Murphy ML, 1991, Cardiac toxicity 4 to 20 years after completing anthracycline therapy. JAMA 266:1672–1677 Tabata T, Oki T, Yamada H, Abe M, Onose Y, Thomas JD, 2000, Subendocardial motion in hypertrophic cardiomyopathy: assessment from long- and short-axis views by pulsed tissue Doppler imaging. J Am Soc Echocardiogr 13:108–115 Tan C, Tasaka H, Yu KP, Murphy ML, Karnofsky DA, 1967, Daunomycin, an antitumor antibiotic, in the treatment of neoplastic disease: clinical evaluation with special reference to childhood leukemia. Cancer 20:333–353 Tassan-Mangina S, Codorean D, Metivier M, Costa B, Himberlin C, Jouannaud C, Blaise AM, Elaerts J, Nazeyrollas P, 2006, Tissue Doppler imaging and conventional echocardiography after anthracycline treatment in adults: early and late alterations of left ventricular function during a prospective study. Eur J Echocardiogr 7:141–146 Uematsu M, Nakatani S, Yamagishi M, Matsuda H, Miyatake K, 1997, Usefulness of myocardial velocity gradient derived from two-dimensional tissue Doppler imaging as an indicator of regional myocardial contraction independent of translational motion assessed in atrial septal defect. Am J Cardiol 79:237– 241 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 69 EP 77 DI 10.1007/s12253-008-9013-4 PG 9 ER PT J AU Camlica, H Duranyildiz, D Oguz, H Oral, NE Yasasever, V AF Camlica, Hakan Duranyildiz, Derya Oguz, Hilal Oral, Nezih Ethem Yasasever, Vildan TI The Diagnostic Value of Macrophage Migration Inhibitory Factor (MIF) in Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MIF. CEA; CA 19-9; Tumor markers; Gastric cancer; Accuracy ID MIF. CEA; CA 19-9; Tumor markers; Gastric cancer; Accuracy AB The present study was conducted to investigate the sensitivity, specificity, predictive values and accuracy of serum MIF, CEA, CA 19-9 levels and their various combinations in patients with gastric cancer. Study group consists of pathologically verified, gastric cancer (n=63) and apparently healthy controls (n=50). Serum MIF concentrations were determined by enzyme-linked immunosorbent assay (ELISA). Serum values of patients were significantly higher than the controls (p=0.011). Diagnostic sensitivity and specificity, predictive values and accuracies were calculated for each marker and their various combinations. The best results were achieved with the marker combination of MIF–CEA–CA 19-9 and MIF–CEA combination. In our opinion, the combination of the markers MIF–CEA is a valuable diagnostic tool for gastric cancer. C1 [Camlica, Hakan] University of Istanbul, Institute of Oncology, Preventive Oncology, Biostatistics and Epidemiology, Capa, 34390 Istanbul, Turkey. [Duranyildiz, Derya] Istanbul Medical Faculty, Istanbul University, Oncology InstituteIstanbul, Turkey. [Oguz, Hilal] Istanbul Medical Faculty, Istanbul University, Oncology InstituteIstanbul, Turkey. [Oral, Nezih Ethem] Istanbul University, Istanbul Medicine Faculty, Institute of Oncology, Department of Radiation OncologyIstanbul, Turkey. [Yasasever, Vildan] Istanbul Medical Faculty, Istanbul University, Oncology InstituteIstanbul, Turkey. RP Camlica, H (reprint author), University of Istanbul, Institute of Oncology, Preventive Oncology, Biostatistics and Epidemiology, 34390 Istanbul, Turkey. EM hcamlica@istanbul.edu.tr CR Correa P, 1992, Human gastric carcinogenesis: a multistep and multifactorial process. Cancer Res 52:6735–6470 Tahara E, Semba S, Tahara H, 1996, Molecular biological observations in gastric cancer. Semin Oncol 23:307–315 Waldman TA, Heberman RH, 1982, Tumor markers in diagnosis and in monitoring therapy. In: Holland JF, Frei E, eds, Cancer Medicine. Lea & Febiger, Philadelphia, pp 1068–1089 Klavins JV, 1985, Tumor markers: clinical and laboratory studies. Alan R. Liss, New York Babaian RJ, Orlando RP, 1986, Elevated prostatic acid phosphatase: a prognostic factor for stage C adeno carcinoma of prostate. J Urol 136:1035–1037 Wanebo HJ, Rao B, Pinsky CM et al, 1978, Preoperative carcinoembryonic antigen level as a prognostic indicator in colorectal cancer. N Eng J Med 299:448–451 Molina R, Filella X, Mengual P et al, 1990, MCA in patients with breast cancer: correlation with CEA and CA 15.3. Int J Biol Markers 5:14–21 David JR, 1996, Delayed hypersensitivity in vitro: its mediation by cell-free substances formed by lymphoid cell-antigen interaction. Proc Natl AcadSci USA 56:72–77 Bloom BR, Bennett B, 1996, Mechanism of a reaction in vitro associated with delayed-type hypersensitivity. Science 153:80–82 Nishihira J, 2000, Macrophage migration inhibitory factor, MIF): its essential role in the immune system and cell growth. Interferon Cytokine Res 20:751–762 Meyer-Siegler K, Hudon PB, 1996, Enhanced expression of macrophage migration inhibitory factor in prostatic adenocarcinoma metastases. Urology 48:448–452 Del Vecchio MT, Tripodi SA, Arcuri F et al, 2000, Macrophage migration inhibitory factor in prostatic adenocarcinoma: correlation with tumor grading and combination endocrine treatment related changes. Prostate 45:51–57 Kamimura A, Kamachi M, Nishihira J et al, 2000, Intracellular distribution of macrophage migration inhibitory factor predicts the prognosis of patients with adenocarcinoma of the lung. Cancer 89:334–341 Tomiyasu M, Yoshino I, Suemitsu R et al, 2002, Quantification of macrophage inhibitory factor mRNA expression in non-small cell lung cancer tissues and its clinical significance. Clin Cancer Res 8:3755–3760 Shimizu T, Abe R, Nakamura H et al, 1999, High expression of macrophage migration inhibitory factor in human melanoma cells and its role in tumor cell growth and angiogenesis. Biochem Biophys Res Commun 264:751–758 Pyle ME, Korbonits M, Gueorguiev M et al, 2003, Macrophage migration inhibitory factor expression is increased in pituitary adenoma cell nuclei. Endocrinol 176:103–110 Munaut C, Boniver J, Foidart JM et al, 2002, Macrophage migration inhibitory factor, MIF, expression inhuman glioblastomas correlates with vascular endothelial growth, VEGF, expression. Neurophathol Appl Neurobiol 28:452–460 Bando H, Matsumoto G, Bando M et al, 2002, Expression of macrophage migration inhibitory factor in human breast cancer: association with nodal spread. Jpn J Cancer Res 93:389–396 Akbar SM, Abe M, Murakami H et al, 2001, Macrophage migration inhibitory factor in hepatocellular carcinoma and liver cirrhosis: relevance to pathogenesis. Cancer Lett 171:125–132 Legendre H, Decaestecker C, Nagy N et al, 2003, Prognostic values of galectin-3 and the macrophage migration inhibitory factor, MIF, in human colorectal cancers. Mod Pathol 16:491– 504 Gold P, Freedman SO, 1965, Specific carcinoembryonic antigens of the human digestive system. J Exp Med 122:467–481 Yasasever V, Camlica H, Oral EN et al, 1999, Utility of CEA in the Diagnosis of Patients with Cancer. J Tumor Marker Oncol 14, 1):33–38 Duffy MJ, 2006, Serum tumor markers in breast cancer: are they of clinical value? Clin Chem 52:345–351 Koprowski H, Steplewski Z, Mitchell K et al, 1979, Colorectal carcinoma antigens detected by hybridoma antibodies. Somatic Cell Genet 5:957–972 Bossuyt PM, Reitsma JB, Bruns DE et al, 2004, Towards complete and accurate Reporting of studies of diagnostic accuracy: the STARD initiative. Fam Pract 21:4–10 Campbell MJ, 1999, Medical statistics: a commonsense approach. John Wiley & Sons Inc, Chichester Solberg HE, 1986, Textbook of clinical chemistry. W.B. Saunders Company, Lexington, pp 356–409 Shkolnik T, Livni E, Reshef R et al, 1987, Comparison of two lymphokines, macrophage migration inhibition, leukocyte adherence inhibition factors, and carcinoembryonic antigen, in colorectal cancer and colonic premalignant lesions. Am J Gastroenterol 82:1275–1278 Shkolnik T, Livni E, Reshef R et al, 1987, The macrophage migration inhibition, MIF, assay as a marker of colorectal cancer. Studies in patients with colorectal cancer, noncolonic neoplasms, and conditions predisposing to colorectal cancer. Dis Colon Rectum 30:101–105 He XX, Yang J, Ding YWet al, 2006, Increased epithelial and serum expression of migration inhibitory factor, MIF, in gastric cancer: potential role of MIF in gastric carcinogenesis. Gut 55:797–802 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 79 EP 83 DI 10.1007/s12253-008-9002-7 PG 5 ER PT J AU Nemes, AJ Redl, P Boda, R Kiss, Cs Marton, JI AF Nemes, A Judit Redl, Pal Boda, Robert Kiss, Csongor Marton, J Ildiko TI Oral Cancer Report from Northeastern Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral cancer; Squamous cell carcinoma; Survival; Northeastern Hungary ID Oral cancer; Squamous cell carcinoma; Survival; Northeastern Hungary AB In Hungary oral and pharyngeal cancers have been reported the fourth most common malignancy in males and the sixth for both sexes. The aim of the present study was to characterize oral squamous cell carcinoma (OSCC) patients in Northeastern Hungary. 119 randomly selected patients with OSCC were included in the study. Epidemiological data, clinicopathological parameters and the risk factors were registered. The most common sites of OSCC were the floor of the mouth (27.7%), the lip (26.9%) and the tongue (22.7%). The majority of the patients was diagnosed with early stage (I–II) lesions and moderately differentiated tumors. The 5-year overall survival rate was 38.7%. There was a significant correlation between survival and tumor size, lymph node involvement and clinical stage. At the time of diagnosis 65.5% of the patients were smokers. Smoking significantly correlated with younger age, male gender, advanced clinical stages and alcohol consumption. 75.5% of the patients consumed alcohol, 41.1% of them exceeding the conventional amount regularly. Drinking habit significantly correlated with younger age, male gender and tumor site i.e. gingiva, retromolar region, tongue. The dental status was acceptable only in 12.6% of the cases. There was a significant correlation between dental status and age, smoking and drinking habits. Clinical stage has the most significant impact on survival and the most important high-risk habits in Northeastern Hungary are smoking and alcohol consumption. Therefore, early detection and treatment, cessation of tobacco and alcohol abuse, and a regular dental care may improve patients’ survival in the region. C1 [Nemes, A Judit] University of Debrecen, Faculty of Dentistry, Nagyerdei krt. 98., 4012 Debrecen, Hungary. [Redl, Pal] University of Debrecen, Faculty of Dentistry, Nagyerdei krt. 98., 4012 Debrecen, Hungary. [Boda, Robert] University of Debrecen, Faculty of Dentistry, Nagyerdei krt. 98., 4012 Debrecen, Hungary. [Kiss, Csongor] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, Nagyerdei krt. 98., 4012 Debrecen, Hungary. [Marton, J Ildiko] University of Debrecen, Faculty of Dentistry, Nagyerdei krt. 98., 4012 Debrecen, Hungary. RP Nemes, AJ (reprint author), University of Debrecen, Faculty of Dentistry, 4012 Debrecen, Hungary. EM nemesj@dote.hu CR Shibuya K, Mathers CD, Boschi-Pinto C et al, 2002, Global and regional estimates of cancer mortality and incidence by site: II. results for the global burden of disease 2000. BMC Cancer 2:37 Otto S, 2003, Cancer epidemiology in Hungary and the Bela Johan National Program for the Decade of Health. Path Oncol Res 9:126–130 La Vecchia C, Lucchini F, Negri E, Levi F, 2004, Trends in cancer mortality in Europe. Oral Oncol 40:433–439 Annertz K, Anderson H, Biorklund A et al, 2002, Incidence and survival of squamous cell carcinoma of the tongue in Scandinavia, with special reference to young adults. Int J Cancer 101:95–99 Schantz SP, Yu GP, 2002, Head and neck cancer incidence trends in young Americans, 1973-1997, with special analysis for tongue cancer. Arch Otolaryngol Head Neck Surg 128:268–274 Coleman MP, Gatta G, Verdecchia A et al, 2003, EUROCARE-3 summary: cancer survival in Europe at the end of the 20th century. Ann Oncol 14:128–149 Neville BW, Damm DD, Allen CM, Soquot JE, 1995, Oral and maxillofacial pathology. W.B. Saunders, Philadelphia, pp 295 World Health Organisation, 1988, International classification of diseases for oncology. WHO, Geneva Sobin LH, Wittekind CH, 1997, Head and neck tumours. In: International Union Against Cancer, Union Internationale Contre Cancer,, eds, TNM classification of malignant tumours. Wiley- Liss, New York, pp 20–24 Bray F, Sankila R, Ferlay J, Parkin DM, 2002, Estimates of cancer incidence and mortality in Europe in 1995. Eur J Cancer 38:99–166 Busquets JM, Garcia HA, Trinidad-Pinedo J, Naez A, 2003, Clinicopathologic characteristics of head and neck squamous cell carcinoma in Puerto Ricans. P R Health Sci J 22:259–264 Chandu A, Adams G, Smith ACH, 2005, Factors affecting survival in patients with oral cancer: an Australian perspective. Int J Oral Maxillofac Surg 34:514–520 Charabi B, Torring H, Kirkegaard J, Hansen HS, 2000, Oral cancer—results of treatment in the Copenhagen University Hospital. Acta Otolaryngol Suppl 543:246–247 Gervasio OL, Dutra RA, Tartaglia SM et al, 2001, Oral squamous cell carcinoma: a retrospective study of 740 cases in a Brazilian population. Braz Dent J 12:57–61 Rodrigues VC, Moss SM, Tuomainen H, 1998, Oral Cancer in the UK: to screen or not to screen. Oral Oncol 34:454–465 Silverman S, 2001, Demographics and occurrence of oral and pharyngeal cancers. The outcomes, the trends, the challenge. JADA 132:7S–11S Chen YK, Huang HC, Lin LM, Lin CC, 1999, Primary oral squamous cell carcinoma: an analysis of 703 cases in southern Taiwan. Oral Oncol 35:173–179 Charabi S, Balle V, Charabi B et al, 1997, Squamous cell carcinoma of the oral cavity: the results of the surgical and nonsurgical therapeutic modalities in a consecutive series of 156 patients treated in Copenhagen county. Acta Otolaryngol Suppl 529:226–228 Crissman JD, Liu WY, Gluckman JL, Cummings G, 1984, Prognostic value of histologic parameters in squamous cell carcinoma of the oropharynx. Cancer 54:2995–3001 Bundgaard T, Bentzen SM, Wildt J et al, 1996, Histopathologic, stereologic, epidemiologic, and clinical parameters in the prognostic evaluation of squamous cell carcinoma of the oral cavity. Head Neck 18:142–152 Oliver AJ, Helfrick JF, Gard D, 1996, Primary oral squamous cell carcinoma: a review of 92 cases. Oral Maxillofac Surg 54:949–954 LeiteICG, KoifmanS1998Survival analysis in a sample of oral cancer patients at a reference hospital in Rio de Janeiro, BrazilOral Oncol 34:347–352 International Agency for Research on Cancer, 1990, Cancer: causes, occurrence and control. IARC Scientific Publications, No. 100 Lyon Andre K, Schraub S, Mercier M, Bontemps P, 1995, Role of alcohol and tobacco in the aetiology of head and neck cancer: a case-control study in Doubs region of France. Eur J Cancer Oral Oncol 31B:301–309 Kozponti Statisztikai Hivatal, 2001, A KSH jelenti 2001/2, Central Statistics Office, Hungary reports). KSH, Budapest, pp 18–22 Orszagos Dohanyfustmnetes Egyesulet, Magyar Gallup Intezet, 2004, Dohanyzas a magyar lakossag koreben, Tobacco smoking in the Hungarian population, 1995-2000-2004 http://www.ode.hu/ ode/monitoring2004.htm Cited 19 Jan 2005 Kopp M, Csoboth C, 2001, Onkarosito magatartasformak a magyar nepesseg koreben, Self-destructive behaviour in the Hungarian population). Magy Onkol 45:139–142 Bundgaard T, Bentzen SM, Wildt J, 1994, The prognostic effect of tobacco and alcohol consumption in intra-oral squamous cell carcinoma. Eur J Cancer Oral Oncol 30B:323–328 Gorsky M, Epstein JB, Oakley C et al, 2004, Carcinoma of the tongue: a case series analysis of clinical presentation, risk factors, staging and outcome. Oral Surg Oral Med Oral Pathol Oral Radiol Endo 98:546–552 Lissowska J, Pilarska A, Pilarski P et al, 2003, Smoking, alcohol, diet, dentition and sexual practices in the epidemiology of oral cancer in Poland. Eur J Cancer Prev 12:25–33 Lockhart PB, Norris CMJr, Pulliam C, 1998, Dental factors in the genesis of squamous cell carcinoma of the oral cavity. Oral Oncol 34:133–139 Moreno-Lopez LA, Esparza-Gomez GC, Gonzalez-Navarro A et al, 2000, Risk of oral cancer associated with tobacco smoking, alcohol consumption and oral hygiene: a case-control study in Madrid, Spain. Oral Oncol 36:170–174 Homann N, Tillonen J, Meurman J et al, 2000, Increased salivary acetaldehyde levels in heavy drinkers and smokers: a microbiological approach to oral cavity cancer. Carcinogenesis 21:57–59 Homann N, Tillonen J, Rintamaki H et al, 2001, Poor dental status increases acetaldehyde production from ethanol in saliva: a possible link to increased oral cancer risk among heavy drinkers. Oral Oncol 37:153–158 de Visscher JGAM, Schaapveld M, Otter R et al, 1998, Epidemiology of cancer of the lip in the Netherlands. Oral Oncol 34:421–426 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 85 EP 92 DI 10.1007/s12253-008-9021-4 PG 8 ER PT J AU Bognar, G Istvan, G Bereczky, B Ondrejka, P AF Bognar, Gabor Istvan, Gabor Bereczky, Biborka Ondrejka, Pal TI Detection of Human Papillomavirus Type 16 in Squamous Cell Carcinoma of the Colon and Its Lymph Node Metastases with PCR and Southern Blot Hybridization SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE Human papillomavirus; HPV; Squamous cell cancer; Colon cancer; PCR; Southern blot hybridization ID Human papillomavirus; HPV; Squamous cell cancer; Colon cancer; PCR; Southern blot hybridization AB The etiological role of human papillomavirus (HPV) in a number of squamous malignant tumors is well known. Squamous cell carcinoma (SCC) of colon is a rare disease with uncertain etiology. Our objective was to detect possible HPV infection in a colon SCC patient. The 94-year-old female patient was operated due to colon tumor causing passage disturbances. Histology confirmed SCC. Tumor tissue and the removed lymph nodes were examined with polymerase chain reaction and Southern blot hybridization techniques. Of HPV types most often occurring in malignant tumors (16, 18) the presence of HPV type 16 could be confirmed in the primary tumor and in four out of the nine surrounding lymph nodes, of which two were metastatic. HPV-16 infection could be detected in an SCC patient in the primary tumor and in surrounding lymph nodes. According to our knowledge, no similar study has been published yet. C1 [Bognar, Gabor] Semmelweis University, 2nd Department of Surgery, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Istvan, Gabor] Semmelweis University, 2nd Department of Surgery, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Bereczky, Biborka] Semmelweis University, 2nd Department of Surgery, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Ondrejka, Pal] Semmelweis University, 2nd Department of Surgery, Kutvolgyi ut 4, 1125 Budapest, Hungary. RP Bognar, G (reprint author), Semmelweis University, 2nd Department of Surgery, 1125 Budapest, Hungary. EM bgabor@kut.sote.hu;bgbgbg@freemail.hu CR McMurray HR, Hguyen D, Westbrook TF, McAnce DJ, 2001, Biology of human papilloma viruses. Int J Exp Pathol 82:15–33 Mantovani F, Banks L, 2001, The human papillomavirus E6 protein and its contribution to malignant progression. Oncogene 20:7677–7685 Nguyen DX, Westbrook TF, McCance DJ, 2002, Human papilloma virus type 16 E7 maintains elevated levels of cdc25A tyrosone phosphatase during deregulation of cell cycle arrest. J Virol 76:619–632 Syrjanen KJ, 2002, HPV infection and oesophageal cancer. J Clin Pathol 55:721–728 Perez LO, Abba MC, Laguens RM, Golijov CD, 2005, Analysis of adenocarcinoma of the colon and rectum: detection of human papillomavirus, HPV, DNA by polymerase chain reaction. Colorectal Dis 7:492–495 Bodaghi S, Yamanegi K, Xiao SY, et al, 2005, Colorectal papillomavirus infection in patients with colorectal cancer. Clin Cancer Res 11:2862–2867 Buyru N, Budak N, Yazici M, Dalay N, 2003, p53 gene mutations are rare in human papillomavirus-associated colon cancer. Oncol Rep 10:2089–2092 Michelassi F, Mishlove LA, Stipa F, Block GE, 1988, Squamous cell carcinoma of the colon: Experience at University of Chicago, review of the literature, report of two cases. Dis Colon Rectum 31:228–235 Sotlar K, Koveker G, Aepinus C, et al, 2001, Human papilloma virus type 16 associated primary squamous cell carcinoma of the rectum. Gastroenterology 120:1046–1048 Audeau A, Han HW, Johnston MJ, et al, 2002, Does human papilloma virus have a role in squamous cell carcinoma of the colon and upper rectum? Eur J Surg Oncol 28:657–660 Bognar G, Imdahl A, Ledniczky G, Ondrejka P, 2006, A HPV infekcio lehetseges prediktiv szerepe a nyelocsorakos betegek neoadjuvans terapiara adott remissziojaban. Magyar Sebeszet 59:97–104 Southern EM, 1975, Detection of specific sequences among DNA fragments separated by gel electrophoresis. J Mol Biol 98:503– 517 Schmidtmann M, 1919, Zur Kenntnis seltener Krebsformen. Virchows Arch B Pathol Anat 226:100–118 Frizelle FA, Hobday KS, Batts KP, Nelson H, 2001, Adenosquamous and squamous carcinoma of the colon and upper rectum. A clinical and histopathological study. Dis Colon Rectum 22:341–346 Cooper HS, 1989, Carcinoma of the colon and rectum. In Norris HT, Ed., Pathology of the colon, small intestine and anus, p 201. Churchill Livingstone, New York Grivalinsky HT, Helwig EG, 1956, Carcinoma of the anorectal junction. Histological considerations. Cancer 9:480–488 Hohm WH, Jackman RJ, 1964, Squamous cell carcinoma of the rectum complicating ulcerative colitis. Mayo Clin Proc 39:249– 251 Goldgraber MB, Humphreys EM, Kirsner JB, Palmer WL, 1960, Carcinoma and ulcerative colitis: clinicopathologic study. Gastroenterology 34:809–838 Michelassi F, Montag AG, Block GE, 1988, Adenosquamous cell carcinoma in ulcerative colitis: Report of a case. Dis Colon Rectum 31:323–326 Weiner MF, Polayes SH, Yidi R, 1962, Squamous carcinoma with schistosomiasis of the colon. Am J Gastroenterol 37:48–54 Pittella JE, Torres AV, 1982, Primary squamous cell carcinoma of the colon and ascending colon: report of a case and review of the literature. Dis Colon Rectum 25:483–487 Williams GT, Blackshaw AJ, Morson BC, 1979, Squamous carcinoma of the colorectum and its genesis. J Pathol 129:139– 147 Yun K, Sherwood MJ, 1992, In situ hybridisation at light and electron microscopic levels: indentification of human papilloma virus nucleic acids. Pathology 24:91–98 Cheng JY, Sheu LF, Meng CL, et al, 1995, Detection of human papilloma virus DNA in colorectal carcinoma by polymerase chain reaction. Gut 37:87–90 Lee YM, Leu SY, Chiang H, et al, 2001, Human papilloma virus type 18 in colorectal cancer. J Microbiol Immunol Infect 34:87–91 Shah KV, Daniel RW, Simons JW, Vogelstein B, 1992, Investigation of colon cancer for human papillomavirus genomic sequences by polymerase chain reaction. J Clin Oncol 51:5–7 Gilbert JM, Mann CV, Scholefield J, Domizio P, 1991, The aetilogy and surgery of carcinoma of the anus, rectum and sigmoid colon in Crohn’s disease. Negative correlation with human papillomavirus type 16, HPV 16). Eur J Surg Oncol 17:507–513 Likaszuk K, Liss J, Wozniak I, et al, 2004, HPV and histopathological status of pelvic lymph node metastases in cervical cancer: a prospective study. J Clin Pathol 57:472–476 Fule T, Csapo Z, Mathe M, et al, 2006, Prognostic significance of high-risk HPV status in advanced cervical cancers and pelvic lymph nodes. Gynecol Oncol 100:570–578 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 93 EP 96 DI 10.1007/s12253-008-9011-6 PG 4 ER PT J AU Ergin, H Yildirim, B Dagdeviren, E Yagci, B Ozen, F Sen, N Duzcan, E AF Ergin, Hacer Yildirim, Basak Dagdeviren, Erol Yagci, Baki Ozen, Fatih Sen, Nilay Duzcan, Ender TI A Prenatally Detected Case of Congenital Hepatoblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatoblastoma; Prenatal diagnosis; Ultrasound ID Hepatoblastoma; Prenatal diagnosis; Ultrasound AB Hepatoblastoma is a rare tumor of childhood and its incidence in the first year of life is about one in a million. Forty-two congenital hepatoblastoma cases were reported so far. Among 42 congenital hepatoblastoma patients, only seven cases have been detected in the prenatal period. Here we report a rare case diagnosed before birth and confirmed by postmortem autopsy. C1 [Ergin, Hacer] Pamukkale University, Faculty of Medicine, Department of PediatricsDenizli, Turkey. [Yildirim, Basak] Pamukkale University, Faculty of Medicine, Department of Obstetrics and GynecologyDenizli, Turkey. [Dagdeviren, Erol] Pamukkale University, Faculty of Medicine, Department of PediatricsDenizli, Turkey. [Yagci, Baki] Pamukkale University, Faculty of Medicine, Department of RadiologyDenizli, Turkey. [Ozen, Fatih] Ege Hospital, Department of PediatricsDenizli, Turkey. [Sen, Nilay] Pamukkale University, Faculty of Medicine, Department of PathologyDenizli, Turkey. [Duzcan, Ender] Pamukkale University, Faculty of Medicine, Department of PathologyDenizli, Turkey. RP Duzcan, E (reprint author), Pamukkale University, Faculty of Medicine, Department of Pathology, Denizli, Turkey. EM eduzcan@pau.edu.tr CR Shih JC, Tsao PN, Huang SF, Yen BL, Lin JH, Lee CN, Hsieh FJ, 2000, Antenatal diagnosis of congenital hepatoblastoma in utero. Ultrasound Obstet Gynecol 16:94–97 Ammann RA, Plaschkes J, Leibundgut K, 1999, Congenital hepatoblastoma: a distinct entity. Med Pediatr Oncol 32:466–468 Woodward PJ, Sohaey R, Kennedy A, Koeller KK, 2005, From the archives of the AFIP: a comprehensive review of fetal tumors with pathologic correlation. Radiographics 25:215–242 Finegold MJ, 1991, Liver tumors. In: Walker WA, Durie PR, Hamilton JR, Walker-Smith JA, Watkins JB, ed, Pediatric gastrointestinal disease: pathophysiology, diagnosis, management. B. C. Decker, Hamilton, 914–926 Lack IF, Neave C, Vawter GF, 1982, Hepatoblastoma: a clinical and pathological study of 54 cases. Am J Surg Pathol 6:693–705 Dehner LP, 1978, Hepatic tumors in the pediatric age group: a distinctive clinicopathologic spectrum. In: Rosenberg HS, Bolande RP, eds, Perspective in pediatric pathology. Year Book, Chicago, 27–68 Roy CC, Silverman A, Alagille D, 1995, Pediatric clinical gastroenterology. Mosby-year Book, St. Louis McNamara A, Levine D, 2005, Intraabdominal fetal echogenic masses: a practical guide to diagnosis and management. Radiographics 25:633–645 Ohishi H, Hirai T, Yamada R, Hirohashi S, Uchida H, Hashimoto H, Jibiki T, Takeuchi Y, 1998, Three-dimensional power Doppler sonography of tumor vascularity. J Ultrasound Med 17:619–622 Miller JH, Greenspan BS, 1985, Integrated imaging of hepatic tumors in childhood. Part II. Benign lesions, congenital, reperative, and inflammatory). Radiology 154:91–100 Foucar E, Williamson RA, Yiu-Chiu V, Varner MW, Kay BR, 1983, Mesenchymal hamartoma of the liver identified by fetal sonography. Am J Roentgenol 140:970–972 Gonen R, Fong K, Chiasson DA, 1989, Prenatal sonographic diagnosis of hepatic hemangioendothelioma with secondary nonimmune hydrops fetalis. Obstet Gynecol 73:485–487 Garmel SH, Crombleholme TM, Semple JP, Bhan I, 1994, Prenatal diagnosis and management of fetal tumors. Semin Perinatol 18:350–365 Weinberg AG, Finegold MJ, 1983, Primary hepatic tumors of childhood. Hum Pathol 14:512–537 Von Schweinitz D, Gluer S, Mildenberger H, 1995, Liver tumors in neonates and very young infants: diagnostic pitfalls and therapeutic problems. Eur J Pediatr Surg 5:72–76 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 97 EP 100 DI 10.1007/s12253-008-9001-8 PG 4 ER PT J AU Horvath, FI Szanto, A Csiki, Z Szodoray, P Zeher, M AF Horvath, Fanny Ildiko Szanto, Antonia Csiki, Zoltan Szodoray, Peter Zeher, Margit TI Intrapulmonary Rheumatoid Nodules in a Patient with Long-Standing Rheumatoid Arthritis Treated with Leflunomide SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intrapulmonary rheumatoid nodules; Leflunomide; Video-associated thoracoscopy; Histological findings ID Intrapulmonary rheumatoid nodules; Leflunomide; Video-associated thoracoscopy; Histological findings AB Rheumatoid nodules are well established manifestations of rheumatoid arthritis but in the lungs they are very rare according to the literature. In our study we present the case of a 34-year-old woman with rheumatoid arthritis and secondary Sjogren’s syndrome who developed multiplex rheumatoid nodules in the lungs 3 years after initiating leflunomide therapy. During leflunomide therapy we did not detect inflammation in the joints. Surprisingly, in November 2005 she started to cough, had low grade fever and low back pain. On the chest X-ray there were multiplex necrobiotic nodules in the lungs. All bacteriological, viral and fungal investigations including tuberculosis, serological tests and cytology were negative. The X-ray, video-associated thoracoscopy and repeated biopsy of the lung followed by histology of the samples proved intrapulmonary rheumatoid nodules, caused by leflunomide. C1 [Horvath, Fanny Ildiko] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. str 22, 4004 Debrecen, Hungary. [Szanto, Antonia] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. str 22, 4004 Debrecen, Hungary. [Csiki, Zoltan] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. str 22, 4004 Debrecen, Hungary. [Szodoray, Peter] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. str 22, 4004 Debrecen, Hungary. [Zeher, Margit] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. str 22, 4004 Debrecen, Hungary. RP Zeher, M (reprint author), University of Debrecen, Medical and Health Center, 3rd Department of Medicine, 4004 Debrecen, Hungary. EM zeher@iiibel.dote.hu CR Scadding JG, 1969, The lungs in rheumatoid arthritis. Proc R Soc Med 62:227–238 Hull S, Mathews JA, 1982, Pulmonary necrobiotic nodules as a presenting feature of rheumatoid arthritis. Ann Rheum Dis 41: 21–24 Jolles H, Moseley PL, Peterson MW, 1989, Nodular pulmonary opacities in patients with rheumatoid arthritis. A diagnostic dilemma. Chest 96:1022–1025 Laloux L, Chevalier X, Maitre B, Lange F, Chanzy MO, Larget- Piet B, Claudepierre P, 1999, Unusual onset of rheumatoid arthritis with diffuse pulmonary nodulosis: a diagnostic problem. J Rheumatol 26:920–922 Teruuchi S, Bando M, Suqiyama Y, Kitamura S, Murayama H, Sohara Y, Hironaka M, Kuriki K, Saito K, 1999, Multiple intrapulmonary rheumatoid nodules. Nihon Kokyuki Gakkai Zasshi 37:829–833 Karadaq F, Polatli M, Senturk T, Kacar F, Sen S, Clidaq O, 2003, Cavitary necrobiotic nodule imitating malignant lung disease in a patient without articular manifestations of rheumatoid arthritis. J Clin Rheumatol 9:246–252 Kitamura A, Matsuno T, Narita M, Shimokata K, Yamashita Y, Mori N, 2004, Rheumatoid arthritis with diffuse pulmonary rheumatoid nodules. Pathol Int 54:798–802 Kobayashi T, Satoh K, Ohkawa M, Satoh A, 2005, Multiple rheumatoid nodules with rapid thin-walled cavity formation producing pneumothorax. J Thorac Imaging 20:47–49 Kerstens PJ, Boerbooms AM, Jeurissen ME, Fast JH, Assmann KJ, van de Putte LB, 1992, Accelerated nodulosis during low dose methotrexate therapy for rheumatoid arthritis. An analysis of ten cases. J Rheumatol 19:867–871 Gotsman I, Goral A, Nusair S, 2001, Secondary spontaneous pneumothorax in a patient with pulmonary rheumatoid nodules during treatment with methotrexate. Rheumatology, Oxford, 40:350–351 Liote H, 2004, Respiratory complications of new treatments for rheumatoid arthritis. Rev Mal Respir 21:1107–1115 Rozin A, Yigla M, Guralnik L, Keidar Z, Vlodavsky E, Rozenbaum M, Nahir AM, Balbir-Gurman A, 2006, Rheumatoid lung nodulosis and osteopathy associated with leflunomide therapy. Clin Rheumatol 25:384–388 Braun MG, Van Rhee R, Becker-Capeller D, 2004, Development and/or increase of rheumatoid nodules in RA patients following leflunomide therapy. Z Rheumatol 63:84–87 Ulusoy H, Bilgici A, Kuru O, Celenk C, 2005, Pulmonary abscess due to leflunomide use in rheumatoid arthritis: a case report. Rheumatol Int 25:139–142 Kamata Y, Nara H, Kamimura T, Haneda K, Iwamoto M, Masuyama J, Okazaki H, Minota S, 2004, Rheumatoid arthritis complicated with acute interstitial pneumonia induced by leflunomide as an adverse reaction. Intern Med 43:1201–1204 Takeishi M, Akiyama Y, Akiba H, Adachi D, Hirano M, Mimura T, 2005, Leflunomide induced acute interstitial pneumonia. J Rheumatol 32:1160–1163 Sakai F, Noma S, Kurihara Y, Yamada H, Azuma A, Kudoh S, Ichikawa Y, 2005, Leflunomide-related lung injury in patients with rheumatoid arthritis: imaging features. Mod Rheumatol 15:173–179 Suissa S, Hudson M, Ernst P, 2006, Leflunomide use and the risk of interstitial lung disease in rheumatoid arthritis. Arthritis Rheum 54:1435–1439 Ochi S, Hariqai M, Mizoquchi F, Iwai H, Haqiyama H, Oka T, Miyasaka N, 2006, Leflunomide-related acute interstitial pneumonia in two patients with rheumatoid arthritis: autopsy findings with a mosaic pattern of acute and organizing diffuse alveolar damage. Mod Rheumatol 16:316–320 Smith CA, Arnett FC, 1991, Diagnosing rheumatoid arthritis: current criteria. Am Fam Physician 44:863–870 Vitali C, Bombardieri S, Jonsson R, Moutsopoulos HM, Alexander EL, Carsons SE, Daniels TE, Fox PC, Fox RI, Kassan SS, Pillemer SR, Talal N, Weisman MH, European Study Group on Classification Criteria for Sjogren’s Syndrome, 2002, Classification criteria for Sjogren’s syndrome: a revised version of the European criteria proposed by the American–European Consensus Group. Ann Rheum Dis 61:554–558 Chen YF, Jobanputra P, Barton P, Jowett S, Bryan S, Clark W, Fry- Smith A, Burls A, 2006, A systematic review of the effectiveness of adalimumab, etanercept and infliximab for the treatment of rheumatoid arthritis in adults and an economic evaluation of their cost-effectiveness. Health Technol Assess 10:1–229 Ostor AJ, Chilvers ER, Somerville MF, Lim AY, Lane SE, Crisp AJ, Scott DG, 2006, Pulmonary complications of infliximab therapy in patients with rheumatoid arthritis. J Rheumatol 33:622–628 Hansen KE, Hildebrand JP, Genovese MC, Cush JJ, Patel S, Cooley DA, Cohen SB, Gangnon RE, Schiff M, 2004, The safety and efficacy of leflunomide in combination with infliximab in rheumatoid arthritis. Arthritis Rheum 51:228–232 Korkmaz C, Us T, Kasifoglu T, Akgun Y, 2006, Anti-cyclic citrullinated peptide, CCP, antibodies in patients with longstanding rheumatoid arthritis and their relationship with extraarticular manifestations. Clin Biochem 39:961–965 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2008 VL 14 IS 1 BP 101 EP 104 DI 10.1007/s12253-008-9003-6 PG 4 ER PT J AU Bianchi, S Vezzosi, V AF Bianchi, Simonetta Vezzosi, Vania TI Microinvasive Carcinoma of the Breast SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Microinvasion; Definition; Diagnosis; Clinical significance; Breast cancer ID Microinvasion; Definition; Diagnosis; Clinical significance; Breast cancer AB The increased rate of early detection of breast cancer due to widespread mammographic screening has led to an increased incidence not only of in situ but also microinvasive carcinoma (MC). MC has been reported to have a favourable prognosis, but specific definitions have varied in the past making the clinical significance of this entity a subject of debate. In fact, although the diagnosis of MC often appears in pathology reports, this term has not been used in a consistent, standardized manner. In addition, the histological diagnosis of MC can be problematical for the pathologist due to a variety of in situ patterns and artefacts that may be misinterpreted as stromal invasion. Definitions and diagnostic criteria of MC are reviewed and discussed. Based on a review of literature, incidence of axillary lymph node involvement, according to different definitions of microinvasion, is reported. C1 [Bianchi, Simonetta] AOU Careggi, Department of Human Pathology and Oncology, Viale Morgagni 85, 50134 Florence, Italy. [Vezzosi, Vania] AOU Careggi, Department of Human Pathology and Oncology, Viale Morgagni 85, 50134 Florence, Italy. RP Bianchi, S (reprint author), AOU Careggi, Department of Human Pathology and Oncology, 50134 Florence, Italy. EM simonetta.bianchi@unifi.it CR Georgian-Smith D, Lawton TJ, 2001, Calcifications of lobular carcinoma in situ of the breast: radiologic–pathologic correlation. AJR 176:1255–1258 Sapino A, Frigerio A, Peterse JL et al, 2000, Mammographically detected in situ lobular carcinoma of the breast. Virchows Arch 436:421–430 Hanby AM, Hughes TA, 2008, In situ and invasive lobular neoplasia of the breast. Histopathology 52:58–66 Tavassoli FA, 1999, Pathology of the breast, 2nd edn. Appleton & Lange, Stanford Boecker W, Parker S, Schulz-Wendtland R et al, 2006, Ductal carcinoma in situ. In: Boecker W, ed, Preneoplasia of the breast. A new conceptual approach to proliferative breast disease. Elsevier GmbH, Munich Nemoto T, Castillo N, Tsukada Y et al, 1998, Lobular carcinoma in situ with microinvasion. J Surg Oncol 67:41–46 Hoda SA, Chiu A, Prasad ML et al, 2000, Are microinvasion and micrometastasis in breast cancer mountains or molehills? Am J Surg 180:305–308 Lagios MD, Wesdahl PR, Margolin FR et al, 1982, Duct carcinoma in situ. Relationship of extent of non invasive disease to the frequency of occult invasion, multicentricity, lymph node metastases, and short-term treatment failures. Cancer 50:1309–1314 Schuh ME, Nemoto T, Penetrante RB et al, 1986, Intraductal carcinoma. Analysis of presentation, pathologic findings, and outcome of disease. Arch Surg 121:1303–1307 Patchefsky AS, Schwartz GF, Finkelstein SD et al, 1989, Heterogeneity of intraductal carcinoma of the breast. Cancer 63:731–741 Wong JH, Kopald KH, Morton DL, 1990, The impact of microinvasion on axillary node metastases and survival in patients with intraductal breast cancer. Arch Surg 125:1298–1301 Silverstein MJ, Waisman JR, Gamagami P et al, 1990, Intraductal carcinoma of the breast, 208 cases): clinical factors influencing treatment choice. Cancer 66:102–108 National Coordinating Group for Breast Cancer Screening Pathology, 1990, Pathology reporting in breast cancer screening. NHSBSP Publications, Sheffield Sloane JP, 1991, Pathology reporting in breast cancer screening. J Clin Pathol 44:710–725 Rosner D, Lane WW, Penetrante R, 1991, Ductal carcinoma in situ with microinvasion. A curable entity using surgery alone without need for adjuvant therapy. Cancer 67:1498–1503 Solin LJ, Fowble BL, Yeh IT et al, 1992, Microinvasive ductal carcinoma of the breast treated with breast-conserving surgery and definitive irradiation. Int J Radiat Oncol Biol Phys 23:961–968 Silver SA, Tavassoli FA, 1998, Mammary ductal carcinoma insitu with microinvasion. Cancer 82:2382–2390 De Mascarel I, MacGrogan G, Mathoulin-Pelissier S et al, 2002, Breast ductal carcinoma in situ with microinvasion. A definition supported by a long-term study of 1248 serially sectioned ductal carcinomas. Cancer 94:2134–2142 Sobin LH, Wittekind CH, eds,, 1997, Breast tumors in TNM classification of malignant tumors, 5th edn. Wiley-Liss, New York National Coordinating Group for Breast Cancer Screening Pathology, 1995, Pathology reporting in breast cancer screening, 2nd edn. NHSBSP Publications, Sheffield Tavassoli FA, Devilee P, eds,, 2003, World health organization classification of tumours. Pathology and genetics of the tumours of the breast and female genital organs. IARC, Lyon Perry N, Broeders M, de Wolf C et al, eds,, 2006, European guidelines for quality assurance in breast cancer screening and diagnosis, 4th edn. European Communities, Luxemburg Rosen PP, 2001, Rosen’s breast pathology, 2nd edn. Lippincott Williams & Wilkins, Philadelphia Damiani S, Ludvikova M, Tomasic G et al, 1999, Myoepithelial cells and basal lamina in poorly differentiated in situ duct carcinoma of the breast. An immunocytochemical study. Virchows Arch 434:227–234 Schnitt SJ, 1998, Microinvasive carcinoma of the breast: a diagnosis in search of a definition. Adv Anat Pathol 5:367–372 Yaziji H, Gown AM, Sneige N, 2000, Detection of stromal invasion in breast cancer: the myoepithelial markers. Adv Anat Pathol 7:100–109 Werling W, Hwang H, Yaziji H et al, 2003, Immunohistochemical distinction of invasive from non-invasive breast lesions. A comparison study of p63 versus calponin and smooth muscle myosin heavy chain. Am J Surg Pathol 27:82–90 Fischer ER, 1997, Pathobiological considerations relating to the treatment of intraductal carcinoma, ductal carcinoma in situ, of the breast. CA Cancer J Clin 47:52–64 Olivotto I, Levine M, 2001, Clinical practice guidelines for the care and treatment of breast cancer: the management of ductal carcinoma in situ, summary of the 2001 update). JAMC 165:912–913 National Coordinating Group for Breast Cancer Screening Pathology, 2005, Pathology reporting of breast disease. NHSBSP Publication No 58, Sheffield. www.rcpath.org Kinne DW, Petrek JA, Osborne MP et al, 1989, Breast carcinoma in situ. Arch Surg 124:33–36 Simpson T, Thirlby RC, Dail DH et al, 1992, Surgical treatment of ductal carcinoma in situ of the breast. 10- to 20-year follow-up. Arch Surg 127:468–472 Silverstein MJ, 1997, Ductal carcinoma in situ with microinvasion. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast, 1st edn. Williams and Wilkins, Baltimore Aktar S, Michaelson RA, Hutter RV et al, 1998, Predictors of axillary lymph node metastases in small, one centimeter or less, T1a, b primary breast cancer. J Clin Oncol 17:120a Jimenez RE, Visscher DW, 1998, Clinicopathologic analysis of microscopically invasive breast carcinoma. Hum Pathol 29:1412– 1419 Le Bouedec G, Penault Llorca F, de Latour M et al, 1999, Carcinoma canalaire microinvasif du sein. J Gynecol Obstet Biol Reprod 28:10–16 Mann GB, Port ER, Rizza C et al, 1999, Six-year follow-up of patients with microinvasion. Ann Surg Oncol 6:591–598 Zavotsky J, Hansen n, Brennan MB et al, 1999, Lymph node metastasis from ductal carcinoma in situ with microinvasion. Cancer 85:2439–2443 Klauber-DeMore N, Tan LK, Liberman L et al, 2000, Sentinel lymph node biopsy: it is indicated in patients with high-risk ductal carcinoma-in-situ and ductal carcinoma-in-situ with microinvasion? Ann Surg Oncol 7:636–642 Padmore RF, Fowble B, Hoffman J et al, 2000, Microinvasive breast carcinoma: clinicopathologic analysis of a single institution experience. Cancer 88:1403–1409 Prasad ML, Osborne MP, Giri DD et al, 2000, Microinvasive carcinoma, T1 mic, of the breast: clinicopathologic profile of 21 cases. Am J Surg Pathol 24:422–428 Cox CE, Nguyen K, Gray RJ et al, 2001, Importance of lymphatic mapping in ductal carcinoma in situ, DCIS): why map DCIS? Am Surg 67:513–521 Wasserberg N, Morgenstern S, Schachter J et al, 2002, Risk factors for lymph node metastases in breast ductal carcinoma in situ with minimal invasive component. Ann Surg 137:1249–1252 Wong SL, Chao C, Edwards MJ et al, 2002, Frequency of sentinel node metastases in patients with favourable breast cancer histologic subtypes. Am J Surg 184:492–498 Intra M, Zurrida S, Maffini F et al, 2003, Sentinel lymph node metastasis in microinvasive breast cancer. Ann Surg 10:1160–1165 Yang M, Moriya T, Oguma M et al, 2003, Microinvasive ductal carcinoma, T1mic, of the breast. The clinicopathological profile and immunohistochemical features of 28 cases. Pathol Int 53:422–428 Buttarelli M, Houvenaeghel G, MartinoMet al, 2004, Prelevement de ganglions sentinelles dans les carcinomes intracanalaires du sein, ± microinvasion). Ann Chir 129:1105–1111 Giard S, Chauvet MP, Houpeau JL et al, 2005, Le ganglion sentinelle sans curage systematique dans le cancer du sein: bilan d’une experience de 1000 interventions. Gynelcol Obstet Fertil 33:213–219 Wilkie C, White L, Dupont D et al, 2005, An update of sentinel lymph node mapping in patients with ductal carcinoma in situ. Am J Surg 190:563–566 Katz A, Gage I, Evans S et al, 2006, Sentinel lymph node positivity of patients with ductal carcinoma in situ or microinvasive breast cancer. Am J Surg 191:761–766 Leidenius M, Salmenkivi K, Von Smitten K et al, 2006, Tumourpositive sentinel node findings in patients with ductal carcinoma in situ. J Surg Oncol 94:380–384 Gray RJ, Mulheron B, Pockaj BA et al, 2007, The optimal management of the axillae of patients with microinvasive breast cancer in the sentinel lymph node era. Am J Surg 194:845–849 Le Bouedec G, de Lapasse C, Mishellany F et al, 2007, Microinvasive ductal carcinoma of the breast. Role of sentinel lymph node biopsy. Gynecol Obstet Fertil 35:317–322 Zavagno G, Belardinelli V, Marconato R et al, 2007, Sentinel lymph node metastasis from mammary ductal carcinoma in situ with microinvasion. Breast 16:146–151 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 105 EP 111 DI 10.1007/s12253-008-9054-8 PG 7 ER PT J AU Lee, HSA Ellis, OI AF Lee, H S Andrew Ellis, O Ian TI The Nottingham Prognostic Index for Invasive Carcinoma of the Breast SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Carcinoma of breast; Histological grade; Prognosis; Nottingham prognostic index ID Carcinoma of breast; Histological grade; Prognosis; Nottingham prognostic index AB A useful prognostic factor in breast cancer has key roles, including identification of a group of patients whose prognosis is so good they do not require further treatment, such as adjuvant systemic therapy, after local surgery, and secondly a group with a poor prognosis for whom additional treatment would be appropriate. To be of clinical use, prognostic factors must show a wide separation in the outcome of the groups identified and select adequate numbers in each group. No single prognostic factor in invasive carcinoma of the breast satisfies all these criteria. However, the Nottingham prognostic index (NPI), which combines nodal status, tumour size and histological grade, does satisfy these criteria. The NPI has been validated by further studies in Nottingham and by studies in several other countries. Predictive factors, such as oestrogen receptor and HER-2 status, predict whether a tumour is likely to respond to a treatment, and are complimentary to prognostic factors. The NPI can be used in combination with predictive factors to select patients for systemic adjuvant treatments. There is the potential to improve the NPI by inclusion of other factors, such as vascular invasion, but any such alterations would require further validation. C1 [Lee, H S Andrew] Nottingham University Hospitals, Department of Histopathology, City Hospital Campus, NG5 1PB Nottingham, UK. [Ellis, O Ian] Nottingham University Hospitals, Department of Histopathology, City Hospital Campus, NG5 1PB Nottingham, UK. RP Lee, HSA (reprint author), Nottingham University Hospitals, Department of Histopathology, NG5 1PB Nottingham, UK. EM andrew.lee@nuh.nhs.uk CR Clark GM, Hilsenbeck SG, Ravdin PM, De Laurentis M, Osborne CK, 1994, Prognostic factors: rationale and methods of analysis and integration. Breast Cancer Res Treat 32:105–112 Blamey RW, 1996, The design and clinical use of the Nottingham Prognostic Index in breast cancer. Breast 5:156–157 Fisher B, Bauer M, Wickerham DL, Redmond CK, Fisher ER, 1983, Relation of number of positive axillary nodes to the prognosis of patients with primary breast cancer. An NSABP update. Cancer 52:1551–1557 Carter CL, Allen C, Henson DE, 1989, Relation of tumor size, lymph node status, and survival in 24,740 breast cancer cases. Cancer 63:181–187 International Union Against Cancer, 2002, Breast tumours. In: Sobin LH, Wittekind C, Eds, TNM classification of malignant tumours. Wiley, Geneva Millis RR, Springall R, Lee AHS, Ryder K, Rytina ERC, Fentiman IS, 2002, Occult axillary lymph node metastases are of no prognostic significance in breast cancer. Br J Cancer 86:396–401 Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19:403–410 Haybittle JL, Blamey RW, Elston CW et al, 1982, A prognostic index in primary breast cancer. Br J Cancer 45:361–366 Balslev I, Axelsson CK, Zedeler K, Rasmussen BB, Carstensen B, Mouridsen HT, 1994, The Nottingham Prognostic Index applied to 9,149 patients from the studies of the Danish Breast Cancer Cooperative Group, DBCG). Breast Cancer Res Treat 32:281–290 Brown JM, Benson EA, Jones M, 1993, Confirmation of a longterm prognostic index for breast cancer. Breast 2:144–147 Sundquist M, Thorstenson S, Brudin L, Nordenskjold B, South East Swedish Breast Cancer Study Group, 1999, Applying the Nottingham Prognostic Index to a Swedish breast cancer population. Breast Cancer Res Treat 53:1–8 Blamey RW, Ellis IO, Pinder SE et al, 2007, Survival of invasive breast cancer according to the Nottingham Prognostic Index in cases diagnosed in 1990–1999. Eur J Cancer 43:1548–1555 van de Vijver MJ, He YD, van't Veer LJ et al, 2002, A geneexpression signature as a predictor of survival in breast cancer. New Engl J Med 347:1999–2009 Abd El-Rehim DM, Ball G, Pinder SE et al, 2005, Highthroughput protein expression analysis using tissue microarray technology of a large well-characterised series identifies biologically distinct classes of breast cancer confirming recent cDNA expression analyses. Int J Cancer 116:340–350 Blamey RW, Mitchell MJ, Ball GR et al, 2007, Prognostic estimation: re-analysis of data from cases diagnosed in 1990–99 by Cox proportional hazards model. Eur J Cancer Suppl 5(3):28 de Mascarel I, Bonichon F, Durand M et al, 1998, Obvious peritumoral emboli: an elusive prognostic factor reappraised. Multivariate analysis of 1320 node-negative breast cancers. Eur J Cancer 34:58–65 Lee AHS, Pinder SE, Macmillan RD et al, 2006, Prognostic value of lymphovascular invasion in women with lymph node negative invasive breast carcinoma. Eur J Cancer 42:357–362 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 113 EP 115 DI 10.1007/s12253-008-9067-3 PG 3 ER PT J AU Cserni, G AF Cserni, Gabor TI Minimal Disease in Sentinel Nodes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Breast cancer; Sentinel lymph node; Micrometastasis; Isolated tumor cells ID Breast cancer; Sentinel lymph node; Micrometastasis; Isolated tumor cells AB Isolated tumor cells and micrometastases represent low-volume or minimal disease in the regional lymph nodes of breast cancer patients as compared to macrometastases. Sentinel lymph node biopsy is a functional selection and removal of the most likely site of regional metastasis, and gives pathologists the opportunity to concentrate detection techniques on a limited number of lymph nodes. Consequently, more lesions belonging in the two mentioned staging categories are discovered in sentinel lymph nodes. Despite some publications contradicting stochastic models of breast cancer, micrometastases seem to reflect a prognosis intermediate between the nodenegative and macrometastatic nodal status, and they also reflect a risk of non-sentinel node involvement slightly higher than that associated with a node-negative status. Data are more contradictory as concerns isolated tumor cells. This minireview summarizes the definitions, their inconsistencies, pathological protocols aiming at the detection of minimal nodal disease, the prognostic impact and non-sentinel node involvement related risk of such nodal lesions, and their therapeutic consequences. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Nyiri ut 38., H6000 Kecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, H6000 Kecskemet, Hungary. EM cserni@freemail.hu CR Saphir O, Amromin GD, 1948, Obscure axillary lymph node metastasis in carcinoma of the breast. Cancer 1:238–241 Lyman GH, Giuliano AE, Somerfield MR et al, 2005, American Society of Clinical Oncology guideline recommendations for sentinel lymph node biopsy in early-stage breast cancer. J Clin Oncol 23:7703–7720 Nemoto T, Vana J, Bedwani RN et al, 1980, Management and survival of female breast cancer: results of a national survey by the American College of Surgeons. Cancer 45:2917–2924 Vinh-Hung V, Burzykowski T, Cserni G et al, 2003, Functional form of the effect of the numbers of axillary nodes on survival in early breast cancer. Int J Oncol 22:697–704 Vinh-Hung V, Verschraegen C, Promish DI et al, 2004, Ratios of involved nodes in early breast cancer. Breast Cancer Res 6:R680–688 Woodward WA, Vinh-Hung V, Ueno NT et al, 2006, Prognostic value of nodal ratios in node-positive breast cancer. J Clin Oncol 24:2910–2916 Huvos AG, Hutter RVP, Berg JW, 1971, Significance of axillary macrometastases and micrometastases in mammary cancer. Ann Surg 173:44–46 Fisher ER, Palekar A, Rockette H et al, 1978, Pathologic findings from the National Surgical Adjuvant Breast Project, protocol No. 4). V. Significance of axillary nodal micro- and macrometastases. Cancer 42:2032–2038 Cserni G, 2008, Sentinel nodes, micrometastases and isolated tumour cells. In: Walker RA, ed, Prognostic and predictive factors in breast cancer, 2nd edn. Informa Healthcare, London, pp 68–87 Dowlatshahi K, Fan M, Snider HC et al, 1997, Lymph node micrometastases from breast carcinoma. Reviewing the dilemma. Cancer 80:1188–1197 Colleoni M, Rotmensz N, Peruzzotti G et al, 2005, Size of breast cancer metastases in axillary lymph nodes: clinical relevance of minimal lymph node involvement. J Clin Oncol 23:1379–1389 Chen SL, Hoehne FM, Giuliano AE, 2007, The prognostic significance of micrometastases in breast cancer: a SEER population-based analysis. Ann Surg Oncol 14:3378–3384 Giuliano AE, Dale PS, Turner RR et al, 1995, Improved axillary staging of breast cancer with sentinel lymphadenectomy. Ann Surg 222:394–401 Cserni G, Amendoeira I, Apostolikas N et al, 2003, Pathological work-up of sentinel lymph nodes in breast cancer. Review of current data to be considered for the formulation of guidelines. Eur J Cancer 39:1654–1667 Weaver DL, 2003, Sentinel lymph nodes and breast carcinoma: which micrometastases are clinically significant? Am J Surg Pathol 27:842–845 Hermanek P, Hutter RVP, Sobin LH et al, 1999, Classification of isolated tumor cells and micrometastasis. Cancer 86:2668–2673 Sobin LH, Wittekind C, eds,, 2002, UICC TNM classification of malignant tumours, 6th edn. Wiley, New York Greene FL, Page DL, Fleming ID et al, eds,, 2002, AJCC cancer staging handbook—TNM classification of malignant tumours, 6th edn. Springer, New York Schwartz GF, Giuliano AE, Veronesi U, 2002, Proceedings of the consensus conference on the role of sentinel lymph node biopsy in carcinoma of the breast, April 19–22, 2001, Philadelphia, Pennsylvania. Cancer 94:2542–2551 Carter BA, Jensen RA, Simpson JF et al, 2000, Benign transport of breast epithelium into axillary lymph nodes after biopsy. Am J Clin Pathol 113:259–265 Bleiweiss IJ, Nagi CS, Jaffer S, 2006, Axillary sentinel lymph nodes can be falsely positive due to iatrogenic displacement and transport of benign epithelial cells in patients with breast carcinoma. J Clin Oncol 24:2013–2018 Cserni G, 2006, Histopathologic examination of the sentinel lymph nodes. Breast J 12(Suppl 2):S152–S156 Cserni G, Amendoeira I, Apostolikas N et al, 2004, Discrepancies in current practice of pathological evaluation of sentinel lymph nodes in breast cancer. Results of a questionnaire based survey by the European Working Group for Breast Screening Pathology. J Clin Pathol 57:695–701 Cserni G, 2004, A model for determining the optimum histology of sentinel lymph nodes in breast cancer. J Clin Pathol 57:467– 471 Weigelt B, Verduijn P, Bosma AJ et al, 2004, Detection of metastases in sentinel lymph nodes of breast cancer patients by multiple mRNA markers. Br J Cancer 90:1531–1537 Blumencranz P, Whitworth PW, Deck K et al, 2007, Sentinel node staging for breast cancer: intraoperative molecular pathology overcomes conventional histologic sampling errors. Am J Surg 194:426–432 Maiorano E, Mazzarol GM, Pruneri G et al, 2003, Ectopic breast tissue as a possible cause of false-positive axillary sentinel lymph node biopsies. Am J Surg Pathol 27:513–518 Orr RK, 1999, The impact of prophylactic axillary node dissection on breast cancer survival—a Bayesian meta-analysis. Ann Surg Oncol 6:109–116 Vinh-Hung V, Verschraegen C, 2004, Breast-conserving surgery with or without radiotherapy: pooled-analysis for risks of ipsilateral breast tumor recurrence and mortality. J Natl Cancer Inst 96:115–121 Clarke M, Collins R, Darby S et al, 2005, Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: an overview of the randomised trials. Lancet 366:2087–2106 Hellman S, 1994, Karnofsky Memorial Lecture. Natural history of small breast cancers. J Clin Oncol 12:2229–2234 Cserni G, 2007, What is a positive sentinel lymph node in a breast cancer patient? A practical approach. Breast 16:152–160 Degnim AC, Griffith KA, Sabel MS et al, 2003, Clinicopathologic features of metastasis in nonsentinel lymph nodes of breast carcinoma patients. Cancer 98:2307–2315 Van Zee KJ, Manasseh DM, Bevilacqua JL et al, 2003, A nomogram for predicting the likelihood of additional nodal metastases in breast cancer patients with a positive sentinel node biopsy. Ann Surg Oncol 10:1140–1151 Hwang RF, Krishnamurthy S, Hunt KK et al, 2003, Clinicopathologic factors predicting involvement of nonsentinel axillary nodes in women with breast cancer. Ann Surg Oncol 10:248–254 Farshid G, Pradhan M, Kollias J et al, 2004, A decision aid for predicting non-sentinel node involvement in women with breast cancer and at least one positive sentinel node. Breast 13:494–501 Barranger E, Coutant C, Flahault A et al, 2005, An axilla scoring system to predict non-sentinel lymph node status in breast cancer patients with sentinel lymph node involvement. Breast Cancer Res Treat 91:113–119 Degnim AC, Reynolds C, Pantvaidya G et al, 2005, Nonsentinel node metastasis in breast cancer patients: assessment of an existing and a new predictive nomogram. Am J Surg 190, 4):543–550 2005 Oct Chagpar AB, Scoggins CR, Martin RC 2nd et al, 2006, Prediction of sentinel lymph node-only disease in women with invasive breast cancer. Am J Surg 192:882–887 Kocsis L, Svebis M, Boross G et al, 2004, Use and limitations of a nomogram predicting the likelihood of non-sentinel node involvement after a positive sentinel node biopsy in breast cancer patients. Am Surg 70:1019–1024 Cserni G, Bianchi S, Vezzosi V et al, 2007, Validation of clinical prediction rules for a low probability of nonsentinel and extensive lymph node involvement in breast cancer patients. Am J Surg 194:288–293 Cserni G, 2007, Comparison of different validation studies on the use of the Memorial-Sloan Kettering Cancer Center nomogram predicting nonsentinel node involvement in sentinel node-positive breast cancer patients. Am J Surg 194:699–700 Alran S, De Rycke Y, Fourchotte V et al, 2007, Validation and limitations of use of a breast cancer nomogram predicting the likelihood of non-sentinel node involvement after positive sentinel node biopsy. Ann Surg Oncol 14:2195–2201 Viale G, Maiorano E, Pruneri G et al, 2005, Predicting the risk for additional axillary metastases in patients with breast carcinoma and positive sentinel lymph node biopsy. Ann Surg 241:319–325 Leidenius MH, Vironen JH, Riihela MS et al, 2005, The prevalence of non-sentinel node metastases in breast cancer patients with sentinel node micrometastases. Eur J Surg Oncol 31:13–18 Houvenaeghel G, Nos C, Mignotte H et al, 2006, Micrometastases in sentinel lymph node in a multicentric study: predictive factors of nonsentinel lymph node involvement—Groupe des Chirurgiens de la Federation des Centres de Lutte Contre le Cancer. J Clin Oncol 24:1814–1822 Cserni G, Bianchi S, Boecker W et al, 2005, Improving the reproducibility of diagnosing micrometastases and isolated tumor cells. Cancer 103:358–367 Cserni G, Sapino A, Decker T, 2006, Discriminating between micrometastases and isolated tumor cells in a regional and institutional setting. Breast 15:347–354 Lebeau A, Cserni G, Dietel M et al, 2007, Pathological examination of sentinel lymph nodes: work-up—interpretation— clinical implications. Breast Care 2:102–108 Turner RR, Weaver DL, Cserni G et al, 2008, Nodal stage classification for breast carcinoma: improving interobserver reproducibility through standardized histologic criteria and image- based training. J Clin Oncol 26:258–263 Querzoli P, Pedriali M, Rinaldi R et al, 2006, Axillary lymph node nanometastases are prognostic factors for disease-free survival and metastatic relapse in breast cancer patients. Clin Cancer Res 12:6696–6701 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 117 EP 121 DI 10.1007/s12253-008-9052-x PG 5 ER PT J AU Egyed, Zs Pentek, Z Jaray, B Kulka, J Svastics, E Kas, J Laszlo, Zs AF Egyed, Zsofia Pentek, Zoltan Jaray, Balazs Kulka, Janina Svastics, Egon Kas, Jozsef Laszlo, Zsolt TI Radial Scar-Significant Diagnostic Challenge SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Mammography; Diagnostic error; Radial scar; Black star - white star; Triple test assessment ID Breast cancer; Mammography; Diagnostic error; Radial scar; Black star - white star; Triple test assessment AB The prevalence of radial scar (RS) is 0.04% in asymptomatic women participating in population screening for breast cancer. It is important to differentiate RS from concomitant malignancies, which occur in 20–30% of patients, or from small stellate carcinomas which give similar radiomorphology. The aim of our study was to evaluate the effectivity of current breast diagnostic methods in distinguishing between real RS, concomitant malignancy and carcinomas imitating RS. Diagnosis of RS was set up in 61 cases by mammography. Forty-four patients underwent surgical excision: histology showed benign or malignant lesions in 28 and 16 cases, respectively. A series of negative results at follow-up proved the benign nature of the lesion in further 11 cases. Six patients were not available for follow-up. Results of mammography, physical examination, ultrasonography and cytology were evaluated and were compared in 39 benign and 16 malignant cases. Results of examinations were reported on the BI-RADS scale ranging from 1 to 5. The mean categorical scores of all diagnostic processes were around the level of borderline lesions: mammography: 3.49, ultrasonography: 3.06, cytology: 2.47 and physical examination: 1.67. The average age of the patients in the benign and malignant groups were the same: 58 years. The two groups did not differ significantly over either distribution of coded mammographical results (p = 0.2092), or the distribution of mammographical parenchyma density patterns (p = 0.4875). However, the malignant and benign groups differed significantly from each other over the distribution of coded ultrasonographic (p = 0.0176) and cytological (p < 0.0001) results. In conclusion, in the preoperative diagnosis of asymptomatic "black-stars", mammography detects the non-palpable lesions, and ultrasonography together with cytology proved better in the analysis, provided FNAB is US guided. Due to the complex diagnostic approach the nature of the "black stars" is known in the majority of cases prior to the surgical biopsy. C1 [Egyed, Zsofia] MaMMa Egeszsegugyi Rt, Kapas u. 29, H-1023 Budapest, Hungary. [Pentek, Zoltan] MaMMa Egeszsegugyi Rt, Kapas u. 29, H-1023 Budapest, Hungary. [Jaray, Balazs] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Svastics, Egon] Budai MAV Hospital, Department of SurgeryBudapest, Hungary. [Kas, Jozsef] Budai MAV Hospital, Department of SurgeryBudapest, Hungary. [Laszlo, Zsolt] MaMMa Egeszsegugyi Rt, Kapas u. 29, H-1023 Budapest, Hungary. RP Egyed, Zs (reprint author), MaMMa Egeszsegugyi Rt, H-1023 Budapest, Hungary. EM egyed.zsofia@axelero.hu CR Wallis MG, Devakumar R, Hosie KB et al, 1993, Complex sclerosing lesions, radial scars, of the breast can be palpable. Clin Radiol 48:319–320 King TA, Scharfenberg JC, Smetherman DH et al, 2000, A better understanding of the term radial scar. Am J Surg 180:428–432 Mokbel K, Price RK, Mostafa A et al, 1999, Radial scar and carcinoma of the breast: microscopic findings in 32 cases. Breast 8:339–342 Tabar L, Dean PB, 1983, Teaching atlas of mammography. Thieme, New York Breast imaging reporting and data system, BI-RADS,, 1995, 2nd ed. Reston VA, American College of Radiology Gram IT, Funkhoser E, Tabar L, 1997, The Tabar classification of mammographic parenchymal patterns. Eur J Radiol 24:131–136 Dessole S, Meloni GB, Capobianco G et al, 2003, Radial scar of the breast: mammographic enigma in pre-and postmenopausal women. Maturitas 34:227–231 Farshid G, Rush G, 2004, Assessment of 142 stellate lesions with imaging features suggestive radial scar discovered during of population-based screening for breast cancer. Am J Surg Pathol 28:1626–1631 Alleva DQ, Smetherman DH, Farr GH Jr et al, 1999, Radiographics 19 Spec No S27–35. discussion S36–37 Sloane JP, Mayers MM, 1993, Carcinoma and atypical hyperplasia in radial scars and complex sclerosing lesions: importance of lesion size and patient age. Histopathology 3:225–231 Sanders ME, Page DL, Simpson JF et al, 2006, Interdependence of radial scar and proliferative disease with respect to invasive breast carcinoma risk in patients with benign breast biopsies. Cancer 106:1453–1461 Kennedy M, Masterson AV, Kerin M et al, 2003, Pathology and clinical relevance of radial scars: a review. J Clin Pathol 56:721 Patterson JA, Scott M, Anderson N et al, 2004, Radial scar, complex sclerosing lesion risk of breast cancer. Analysis of 175 cases in Northern Irelands. Eur J Surg Oncol 30:1065–1068 Jacobs TW, Byme C, Colditz G et al, 1999, Radial scars in benign breast biopsy specimens and the risk of breast cancer. New Engl J Med 340:430 Alvarado-Cabrero I, Tavassoli FA, 2000, Neoplastic and malignant lesions involving or arising in a radial scar: a clinico-pathologic analysis of 17 cases. Breast J 6:96–102 Sewel CW, 2004, Pathology of high-risk breast lesions and ductal carcinoma in situ., review,, 16 refs, Rad Clin North Am 42: 821–830 Denley H, Pinder SE, Tan PH et al, 2000, Metaplastic carcinoma of the breast arising within complex sclerosing lesion: a report of five cases. Histopatholog 36:203–209 Lamb J, McGoogan E, 1994, Fine needle aspiration cytology of breast in invasive carcinoma of tubular type and in radial scar/ complex sclerosing lesions. Cytopathology 5:17–26 Kulka J, Davies JD, Chinyma CN, 1996, Benign and malignant stellate breast lesions: structural differences in 5 microns sections and thick tissue slices. Anticancer Res 16:3965–3970 Kopper L, Timar J, 2002, Gene expression profiles in the diagnosis and prognosis of cancer. Hung Oncol 46:3–9, in Hungarian) Timar J, Jeney A, Kovalszky I et al, 1995, Role of proteoglycans in tumor progression. Pathol Oncol Res 1:85–93 Timar J, Csuka O, Orosz Zs et al, 2001, Molecular pathology of tumor metastasis. Pathol Oncol Res 3:217–230 Chauhan H, Abraham A, Phillips JR et al, 2003, There is more than one kind of myofibroblast: analysis of CD34 expression in benign, in situ, and invasive breast lesions. J Clin Pathol 56:271 Demirkazik FB, Gulsun M, Firat P, 2003, Mammographic features of nonpalpable spiculated lesions. Clin Imaging 27:293–297 Ung OA, Lee WB, Greenberg ML et al, 2001, Complex sclerosing lesion: the lesion is complex, the management is straightforward. ANZ J Surg 71:35–40 Fasih T, Jain M, Shrimankar J et al, 2005, All radial scars/complex sclerosing lesions seen on breast screening mammograms should be excised. Eur J Surg Oncol 31:1125–1128 McBoyle MF, Razek HA, Carter JL et al, 1997, Tubular carcinoma of the breast: an institutional review. Am Surg 63:639–644 Livi L, Paiar F, Meldolesi E et al, 2005, Tubular carcinoma of the breast: outcome and loco-regional recurrence in 307 patients. Eur J Surg Oncol 31:9–12 Daniele S, Imbriaco M, Riccardi A et al, 2003, Mammographic and sonographic features of tubular breast carcinoma 89:417–420 Mitnick JS, Gianutsos R, Pollack AH et al, 1999, Tubular carcinoma of the breast: Sensitivity of diagnostic techniques and correlation with histopathology. Am J Roentgenol 172:319–323 Canade A, Constantini M, Magistrelli A et al, 2002, Radial scar: from conventional imaging to the new techniques. Case reports. Rays 27:319–323 Pediconi F, Occhiati R, Venditti F et al, 2005, Radial scars of the breast: contrast-enhanced magnetic resonance mammography appearance. Breast J 11:23–28 Shetty MK, 2002, Radial scars of the breast: sonographic findings. Ultrasound Q 18:203–207 Chan HHL, Lam PTW, Yuen JHF et al, 2004, Conditions that mimic primary breast carcinoma onmammography and sonography. J Hong Kong Coll Radiol 7:49–55 Cawson JN, 2005, Can sonography be used to help differentiate between radial scars and breast cancers? Breast 14:352–359 Kovacs A, Walker RA, 2003, P-Cadherin as a marker in differential diagnosis of breast lesions. J Clin Pathol 56:139 Baum F, Fischer U, Fuzesi L et al, 2000, The radial scar in contrast media-enhanced MR-mammography. Rofo 172:817–823 Bonzanini M, Gilioli E, Brancato B et al, 1997, Cytologic features of 22 radial scars/ complex sclerosing lesions of the breast, three of which associated with carcinoma, clinical, mammographic, and histologic correlation. Diagn Cytopathol 17:353–362 Cawson JN, Malara F, Kavanagh A et al, 2003, Fourteen-gauge needle core biopsy of mammographically evident radial scars: is excision necessary? Cancer 97:345–351 Gill HK, Ioffe OB, Berg WA, 2003, When is a diagnosis of sclerosing adenosis acceptable at core biopsy? Radiology 228: 50–57 Brenner RJ, Jackman RJ, Parker SH et al, 2002, Percutaneous core needle biopsy of radial scars of the breast: when is excision necessary? Folyoirat 179:1179–1184 Ruiz-Sauri A, Almenar-Medina S, Callaghan RC et al, 1995, A. Radial scar versus tubular carcinoma of the breast. A comparative study with quantitative techniques(morphometry, image-and flow cytometry). Pathol Res Pract 191:547–554 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 123 EP 129 DI 10.1007/s12253-008-9025-0 PG 7 ER PT J AU Kipikasova, L Wolaschka, T Bohus, P Baumohlova, H Bober, J Blazejova, J Mirossay, L Sarissky, M Mirossay, A Cizmarikova, M Potocekova, D Mojzis, J AF Kipikasova, Livia Wolaschka, Tomas Bohus, Peter Baumohlova, Helena Bober, Juraj Blazejova, Jana Mirossay, Ladislav Sarissky, Marek Mirossay, Andrej Cizmarikova, Martina Potocekova, Dana Mojzis, Jan TI Polymorphisms of the XRCC1 and XPD Genes and Breast Cancer Risk: A Case-Control Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer susceptibility; DNA repair; XPD; XRCC1 ID Breast cancer susceptibility; DNA repair; XPD; XRCC1 AB The purpose of this case control study was to evaluate the role of X-ray repair cross-complementing group 1 (XRCC1) and xeroderma pigmentosum group D (XPD) genotypes as genetic indicators of susceptibility to breast cancer (BC). We analysed DNA samples from 114 breast cancer patients and 113 control subjects using polymerase chain reaction–restriction fragment length polymorphism. For the single nucleotide polymorphisms in XRCC1 exon 10 (Arg399Gln, G/A) and XPD exon 23 (Lys751Gln, A/C), no remarkable differences for genotype distribution and allele frequencies were observed between BC group and control group in the study. The genotype frequency for homozygote A/A in XPD exon 6 (Arg156Arg, C/A) were significantly different between BC and control groups (P<0.0001, odds ratio=2.14; 95% confidence interval 1.44–3.17). The data indicate a possible role for XPD (Arg156Arg, C/A) polymorphisms in BC susceptibility. C1 [Kipikasova, Livia] P.J. Safarik University, Faculty of Medicine, Department of Pharmacology, 040 11 Kosice, Slovakia. [Wolaschka, Tomas] P.J. Safarik University, Faculty of Medicine, Department of Pharmacology, 040 11 Kosice, Slovakia. [Bohus, Peter] University Hospital, Department of PathologyKosice, Slovakia. [Baumohlova, Helena] University Hospital, Department of PathologyKosice, Slovakia. [Bober, Juraj] P.J. Safarik University, Faculty of Medicine, 1st Surgery DepartmentKosice, Slovakia. [Blazejova, Jana] P.J. Safarik University, Faculty of Medicine, 1st Surgery DepartmentKosice, Slovakia. [Mirossay, Ladislav] P.J. Safarik University, Faculty of Medicine, Department of Pharmacology, 040 11 Kosice, Slovakia. [Sarissky, Marek] P.J. Safarik University, Faculty of Medicine, Department of Pharmacology, 040 11 Kosice, Slovakia. [Mirossay, Andrej] P.J. Safarik University, Faculty of Medicine, Department of Pharmacology, 040 11 Kosice, Slovakia. [Cizmarikova, Martina] P.J. Safarik University, Faculty of Medicine, Department of Pharmacology, 040 11 Kosice, Slovakia. [Potocekova, Dana] P.J. Safarik University, Faculty of Medicine, Department of InformaticsKosice, Slovakia. [Mojzis, Jan] P.J. Safarik University, Faculty of Medicine, Department of Pharmacology, 040 11 Kosice, Slovakia. RP Mojzis, J (reprint author), P.J. Safarik University, Faculty of Medicine, Department of Pharmacology, 040 11 Kosice, Slovakia. EM jan.mojzis@upjs.sk CR Parkin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics. CA Cancer J Clin 55:74–108 Mohrenweiser HW, Jones IM, 1998, Variation in DNA repair is a factor in cancer susceptibility: a paradigm for the promises and perils of individual and population risk estimation? Mutat Res 400:15–24 Shen M, Jones I, Mohrenweiser H, 1998, Nonconservative amino acid substitutions variants exist at polymorphic frequency in DNA repair genes in healthy humans. Cancer Res 58:604–608 Moullan N, Cox DG, Angele S et al, 2003, Polymorphisms in the DNA repair gene XRCC1, breast cancer risk, and response to radiotherapy. Cancer Epidemiol Biomarkers Prev 12:1168–1174 Duell EJ, Millikan RC, Pittman GS et al, 2001, Polymorphisms in the DNA repair gene XRCC1 and breast cancer. Cancer Epidemiol Biomarkers Prev 10:217–222 Benhamou S, Sarasin A, 2002, ERCC2/XPD gene polymorphisms and cancer risk. Mutagenesis 6:463–469 Stary A, Sarasin A, 2001, ERCC2, Excision repair crosscomplementing rodent repair deficiency, complementation group 2). Atlas Genet Cytogenet Oncol Haematol. http://www.infobiogen. fr/services/chromcancer/Genes/XPDID297.html Miller S, Dykes D, Polesky HF, 1988, A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16:1215 Lunn RM, Langlois RG, Hsieh LL et al, 1999, XRCC1 polymorphism: effect on aflatoxin B1-DNA adducts and glycophorin A variant frequency. Cancer Res 59:2557–2561 Dybdahl M, Vogel U, Frentz G et al, 1999, Polymorphisms in the DNA repair gene XPD: Correlations with risk and age at onset of basal cell carcinoma. Cancer Epidemiol Biomarkers Prev 8:77–81 Savas S, Kim DY, Ahmad MF et al, 2004, Identifying functional genetic variants in DNA repair pathway using protein conservation analysis. Cancer Epidemiol Biomarkers Prev 13:801–807 Butkiewicz D, Rusin M, Enewold L et al, 2001, Genetic polymorphisms in DNA repair genes and risk of lung cancer. Carcinogenesis 22:593–597 Olshan AF, Watson MA, Weissler MC et al, 2002, XRCC1 polymorphisms and head and neck cancer. Cancer Lett 178:181– 186 Stern MC, Umbach DM, van Gils CH et al, 2001, DNA repair gene XRCC1 polymorphisms, smoking, and bladder cancer risk. Cancer Epidemiol Biomarkers Prev 10:125–131 van Gils CH, Bostick RM, Stern MC et al, 2002, Differences in base excision repair capacity may modulate the effect of dietary antioxidant intake on prostate cancer risk: an example of polymorphisms in the XRCC1 gene. Cancer Epidemiol Biomarkers Prev 11:1279–1284 Chacko P, Rajan B, Joseph T et al, 2005, Polymorphisms in DNA repair gene XRCC1 and increased genetic susceptibility to breast cancer. Breast Cancer Res Treat 89:15–21 Smith TR, Miller MS, Lohman K et al, 2003, Polymorphisms of XRCC1 and XRCC3 genes and susceptibility to breast cancer. Cancer Lett 190:183–190 Brewster AM, Jorgensen TJ, Ruczinski I et al, 2006, Polymorphisms of the DNA repair genes XPD, Lys751Gln, and XRCC1, Arg399Gln and Arg194Trp): relationship to breast cancer risk and familial predisposition to breast cancer. Breast Cancer Res Treat 95:73–80 Thyagarajan B, Anderson KE, Folsom AR et al, 2006, No association between XRCC1 and XRCC3 gene polymorphisms and breast cancer risk: Iowa Women’s Health Study. Cancer Detect Prev 30:313–321 Zhang Y, Newcomb PA, Egan KM et al, 2006, Genetic polymorphisms in base-excision repair pathway genes and risk of breast cancer. Cancer Epidemiol Biomarkers Prev 15:353–358 Matullo G, Guarrera S, Carturan S et al, 2001, DNA repair gene polymorphisms, bulky DNA adducts in white blood cells and bladder cancer in a case-control study. Int J Cancer 92:562–567 Winsey SL, Haldar NA, Marsh HP et al, 2000, A variant within the DNA repair gene XRCC3 is associated with the development of melanoma skin cancer. Cancer Res 60:5612–5616 Forsti A, Angelini S, Festa F et al, 2004, Single nucleotide polymorphisms in breast cancer. Oncol Rep 11:917–922 Metsola K, Kataja V, Sillanpaa P et al, 2005, XRCC1 and XPD genetic polymorphisms, smoking and breast cancer risk in a Finnish case-control study. Breast Cancer Res 6:R987–R997 Nexo BA, Vogel U, Olsen A et al, 2003, A specific haplotype of single nucleotide polymorphisms on chromosome 19q13.2-3 encompassing the gene RAI is indicative of post-menopausal breast cancer before age 55. Carcinogenesis 24:899–904 Justenhoven C, Hamann U, Pesch B et al, 2004, ERCC2 genotypes and a corresponding haplotype are linked with breast cancer risk in a German population. Cancer Epidemiol Biomarkers Prev 13:2059–2064 Tang D, Cho S, Rundle A et al, 2002, Polymorphisms in the DNA repair enzyme XPD are associated with increased levels of PAHDNA adducts in a case-control study of breast cancer. Breast Cancer Res Treat 75:159–166 Shi Q, Wang LE, Bondy ML et al, 2004, Reduced DNA repair of benzo(a)pyrene diol epoxide-induced adducts and common XPD polymorphisms in breast cancer patients. Carcinogenesis 25:1695–1700 Dufloth RM, Costa S, Schmitt F et al, 2005, DNA repair gene polymorphisms and susceptibility to familial breast cancer in a group of patients from Campinas, Brazil. Genet Mol Res 4:771– 782 Terry MB, Gammon MD, Zhang FF et al, 2004, Polymorphism in the DNA repair gene XPD, polycyclic aromatic hydrocarbon- DNA adducts, cigarette smoking, and breast cancer risk. Cancer Epidemiol Biomarkers Prev 13:2053–2058 Caggana M, Kilgallen J, Conroy JM et al, 2001, Associations between ERCC2 polymorphisms and gliomas. Cancer Epidemiol Biomarkers Prev 10:355–360 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 131 EP 135 DI 10.1007/s12253-008-9034-z PG 5 ER PT J AU Thielemann, A Kopczynski, Z Filas, V Breborowicz, J Grodecka-Gazdecka, S Baszczuk, A AF Thielemann, Anna Kopczynski, Zygmunt Filas, Violetta Breborowicz, Jan Grodecka-Gazdecka, Sylwia Baszczuk, Aleksandra TI The Determination of VEGF and MVD, among Patients with Primary Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Angiogenesis; Breast cancer; Microvessel density; Neoplasm; Vascular endothelial growth factor ID Angiogenesis; Breast cancer; Microvessel density; Neoplasm; Vascular endothelial growth factor AB The purpose of the study was to ascertain the value of assessment of vascular endothelial growth factor (VEGF) levels and microvessel density, and to search for correlations between them, in women with breast cancer. The assessment considered factors such as the stage of clinical disease advancement—according to International Union Against Cancer, the grade of histological malignancy, status of axillary lymph nodes and the size of the primary tumour. The concentration of VEGF was assessed in the plasma of 103 women with breast cancer, using an immunoenzymatic method (Quantikine test of R&D Systems). Assessment of microvessel density was performed using histopathological immunoperoxidase methods, using an anti-von Willebrand factor antibody (DAKO A/S). A statistically significant relationship was found between rising VEGF levels and microvessel density in women with breast cancer, when compared to values from a control group. A correlation was found between VEGF concentration and microvessel density (MVD) values. Statistically significant differences were found between VEGF levels of patients in stages I, II and III of clinical disease advancement. For MVD, differences were found only between stages I and III. A statistical relationship was also found between VEGF and MVD and tumour size. Similar results were found between VEGF concentrations in women with metastases to the axillary lymph nodes and cytokine levels in women with no metastases. The results of the study suggest that the degree of tumour vascularization and the concentration of VEGF may represent valuable indicators for the assessment of the angiogenic process in women with breast cancer. C1 [Thielemann, Anna] Poznan University of Medical Sciences, Department of Laboratory Medicine, Lakowa St. No 1/2, 61-878 Poznan, Poland. [Kopczynski, Zygmunt] Poznan University of Medical Sciences, Department of Laboratory Medicine, Lakowa St. No 1/2, 61-878 Poznan, Poland. [Filas, Violetta] Poznan University of Medical Sciences, Department of Oncology, Department of Tumour PathologyPoznan, Poland. [Breborowicz, Jan] Poznan University of Medical Sciences, Department of Oncology, Department of Tumour PathologyPoznan, Poland. [Grodecka-Gazdecka, Sylwia] Poznan University of Medical Sciences, Department of Oncology, Department of Oncological SurgeryPoznan, Poland. [Baszczuk, Aleksandra] Poznan University of Medical Sciences, Department of Laboratory Medicine, Lakowa St. No 1/2, 61-878 Poznan, Poland. RP Thielemann, A (reprint author), Poznan University of Medical Sciences, Department of Laboratory Medicine, 61-878 Poznan, Poland. EM anna.thielemann@oncology.am.poznan.pl CR Folkmann J, 2001, Angiogenesis-dependent diseases. Semin Oncol 28:536–542 Ferrara N, 2002, VEGF and the quest for tumour angiogenesis factors. Nat Rev 2:795–803 RisauW(1997, Mechanisms of angiogenesis. Nature 386:671–674 Ferrara N, 2001, Role of vascular endothelial growth factor in regulation of physiological angiogenesis. Am J Physiol Cell Physiol 208:C1358–C1366 Yancopoulos GD, Davis S, Gale NW, et al, 2000, Vascularspecific growth factors and blood vessel formation. Nature 407:242–248 Carmeliet P, Jain RK, 2000, Angiogenesis in cancer and other diseases. Nature 407:249–257 Esteva FJ, Hortobagyi GN, 2004, Prognostic molecular markers in early breast cancer. Breast Cancer Res 6:109–118 Ferrara N, 1999, Molecular and biological properties of vascular endothelial growth factor. J Mol Med 77:527–543 Dvorak HF, 1999, Vascular permeability factor/vascular endothelial growth factor and the significance if microvascular hyperpermeability in angiogenesis. Curr Top Microbiol Immunol 237:97–132 Breier G, 2000, Functions of the VEGF/VEGF receptor system in the vascular system. Semin Thromb Hemost 26(5):553–559 Kraft A, Weindel K, Ochs A, 1999, Vascular endothelial growth factor in the serum and effusions with malignant and nonmalignant disease. Cancer 85:178–187 Ferrara N, 1999, Vascular endothelial growth factor: molecular and biological aspects. Curr Top Microbiol Immunol 237:1–30 Vincenti V, Cassano C, Rocchi M, Persico G, 1996, Assignment of the vascular endothelial growth factor gene to human chromosome 6p21.3. Circulation 93:1493–1499 Ferrara N, Gerger HP, LeCouter J, 2003, The biology of VEGF and its receptors. Nat Med 9:669–676 Boudreeau N, Myers C, 2003, Breast cancer-induced angiogenesis: multiple mechanisms and the role of the microenvironment. Breast Cancer Res 5:140–146 Edel MJ, Harvey JM, Papadimitriou JM, 2000, Comparison of vascularity and angiogenesis in primary invasive mammary carcinomas and in their respective axillary lymph node metastases. Clin Exp Metastasis 18:695–702 Benoy I, Salgado R, Colpaert C, 2002, Serum interleukin 6, plasma VEGF, serum VEGF, and VEGF platelet load in breast cancer patients. Clin Breast Cancer Jan 1(4):311–316 Takahashi Y, Kitadai Y, Bucana CD, Cleary KR, Ellis LM, 1995, Expression of vascular endothelial growth factor and its receptor, KDR correlates with vascularity, metastasis and proliferation of human colon cancer. Cancer Res 55:3964–3968 Yoshikawa T, Tsuburaya A, Kobayashi O et al, 2005, Plasma concentration of VEGF and bFGF in patients with gastric cancer. Cancer Lett 153:7–12 Ferrari G, Scagliotti GV, 1996, Serum and urinary vascular endothelial growth factor levels in non-small cell lung cancer patients. Eur J Cancer 32A:2368–2376 Latil A, Bieche I, Pesche S, Valeri A, Fournier G, 2000, VEGF overexpression in clinically localized prostate tumors and neuropilin- 1 overexpression in metastatic forms. Int J Cancer 89(2):167–171 Brown LF, Berse B, Jackman RW et al, 1993, Increased expression of vascular permeability factor, vascular endothelial growth factor, and its receptors in kidney and bladder carcinomas. Am J Pathol 143:1255–1262 Savropoulus NE, Bouropoulos C, Ioachim IE, 2004, Prognostic significance of angiogenesis in superficial bladder cancer. Int Urol Nephrol 36(2):45–48 Kaio E, Tanaka S, Kitadai Y et al, 2003, Clinical significance of angiogenic factor expression an the deepest invasive site of advanced colorectal carcinoma. Oncology 64:61–73 Seo Y, Baba H, Fakuda T, et al, 2000, High expression of vascular endothelial factor is associated with liver metastasis and poor prognosis with ductal pancreatic adenocarcinoma. Cancer 88:2239–2245 Weidner N, Semple JP, Welch WR et al, 1991, Tumor angiogenesis and metastasis-correlation in invasive breast cancer. N Engl J Med 324:1–8 Weidner N, Folkmann J, Pozza F, 1992, Tumor angiogenesis: a new significant and independent prognostic indicate in early - stage breast carcinoma. J National Cancer Instit 84:875–886 Hansen S, Grabau DA, Sorensen FB, Bak M, Vach W, Rose C, 2000, Vascular grading of angiogenesis: prognostic significance in breast cancer. Br J Cancer 82:339–347 Perrone G, Vincenzi B, Santini D, 2004, Correlation of p53 and bcl-2 expression with VEGF, microvessel density, MVD, and clinicopathological features in colon cancer. Cancer 28:227–234 Stimfl M, Tong D, Fasching B, 2002, Vascular endothelial growth factor splice variants and their prognostic value in breast cancer and ovarian cancer. Clin Cancer Res 8:2253–2259 Nakashima T, Kondoh S, Kitoh H, et al, 2003, Vascular endothelial growth factor-C expression in human gall bladder cancer and its relationship to lymph node metastasis. Int J Mol Med 11:33–39 Granato AM, Nanni O, Falcini F, Folli S, Mosconi G, 2004, Basic fibroblast growth factor and vascular endothelial growth factor serum levels in breast cancer patients and healthy women: useful as diagnostic tools. Breast Cancer Res 6:38–45 Salven P, Perhoniemi V, Tykka H, 1999, Serum VEGF levels in women with a benign breast tumor or breast cancer. Breast Cancer Res Treat 53:161–166 Fuhrmann-Benzakein H, Ma HN, Rubba-Brandt L, 2000, Elevated levels of angiogenic cytokines in the plasma of cancer patients. Int J Cancer 1(85):40–45 Munn RK, Gagliardi A, Stepick-Bieck P, 1996, Serum vascular endothelial growth factor in women with breast cancer. Proc Am Assoc Cancer Res 37:1298–1310 Valkovic T, Dobrila F, Melato M, Sasso F, Rizzardi C, Jonji N, 2002, Correlation between VEGF, angiogenesis and tumor-associated macrophages in invasive ductal breast carcinoma. Virchows Arch 44(6):583–588 Ludovini V, Sidoni L, Pistola L, et al, 2003, Evaluation of the prognostic role of vascular endothelial growth factor and microvessel density in stages I and II breast cancer patients. Breast Cancer Res Treat 81:159–168 Toi M, Kondo S, Suzuki H, 1996, Quantitative analysis of vascular endothelial growth factor in primary breast in primary breast cancer. Cancer 77(6):1101–1106 Wu Y, Saldana L, Chillar RV, 2002, Plasma vascular endothelial growth factor is useful in assessing progression of breast cancer post surgery and during adjuvant treatment. Oncol 20(3):509–516 Heer K, Kumar H, Read JR, Fox JN, 2001, Serum vascular endothelial growth factor in breast cancer. Its relation with cancer type and estrogen receptor status. Clin Cancer Res 7:3491–3494 Lantzsch T, Hefler L, Krause U, 2002, The correlation between immunohistochemically-detected markers of angiogenesis and serum vascular endothelial growth factor in patients with breast cancer. Anticancer Res 22(3):1921–1928 Colozza M, Ludovini V, Gori S et al, 2003, Are vascular endothelial growth factor, VEGF, and microvessel density, MVD, prognostic factors in stage I and II breast cancer patients?. Breast Cancer Res Treat 81:159–168 Holmes MD, Willett WC, 2004, Does diet affect breast cancer risk. Breast Cancer Res 6:170–178 Murray JD, Carlson GW, McLaughlin K, 1997, Tumor angiogenesis as a prognostic factor in laryngeal cancer. Am J Surg 174:523–529 Yokoyama Y, Sato S, Futagami M et al, 2000, Prognostic significance of vascular endothelial growth factor and its receptors in endometrial carcinoma. Gynecol Oncol 77:413–418 Rosselli M, Mineo TC, Basili S, Mariotti S, Martini F et al, 2003, Vascular endothelial growth factor, VEGF-A, plasma levels in non-small cell lung cancer: relationship with coagulation and platelet activation markers. Thromb Haemost 89:177–184 Linderholm B, 2000, Correlation of vascular endothelial growth factor content with recurrences, survival and first relapse size in primary node-positive breast carcinoma after adjuvant treatment. Oncology 18:1423–1431 Toi M, Inada K, Hoshina S, Tominaga T, 1995, Vascular endothelial growth factor and platelet derivated endothelial cell growth factor are frequently coexpressed in highly vascularized human breast cancer. Clin Cancer Res 1:961–964 Li J, Song ST, Jiang ZF et al, 2002, Significance of microvascular density and vascular endothelial growth factor in breast cancer. Anticancer Res 22:1925–1928 MacConmara M, O’Hanlon DM, Kiely MJ, Conolly Y, Jeffer M, Keane FB, 2002, An evaluation of the prognostic significance of vascular endothelial growth factor in node positive primary breast carcinoma. Int J Oncol 209(4):717–727 Medri L, Nanni O, Volpi A, 2000, Tumor microvessel density and prognosis in node-negative breast cancer. Int J Cancer 89(1):74–80 Weidner N, 1995, Current pathologic methods for measuring intratumoral microvessel density within breast carcinoma and other solid tumours. Breast Cancer Res Treat 36–180 Ogawa Y, Chung Y-S, Nakata B et al, 1995, Microvessel quantitation in invasive breast cancer by staining for factor VIIIrelated antigen. Br J Cancer 71:1297–1301 Sledge GW, 2002, Vascular endothelial growth factor in breast cancer. Semin Oncol 29:204–210 Linderholm B, Lindh B, Tavelin B et al, 2000, p53 and vascular endothelial growth factor expression predicts outcome in 833 patients with primary breast carcinoma. Int J Cancer 89, 1):51–62 Adams J, Carder P, Downey S, Forbes MA, 2000, Vascular endothelial growth factor in breast cancer: comparison of plasma, serum and tissue VEGF and microvessel density and effects of tamoxifen. Cancer Res 60:2898–2905 Kopczynski Z, Thielemann A, Grodecka-Gazdecka S, 2004, The value of assessment of vascular endothelial growth factor, VEGF, in clinical status among breast cancer female patients. Ann UMCS Sect DDD 2:109–113 Thielemann A, Kopczynski Z, Grodecka-Gazdecka S, Piekarska B, 2005, The estimation of plasma VEGF concentration in patients with breast carcinoma. Diagn Lab 41:1–10 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 137 EP 144 DI 10.1007/s12253-008-9040-1 PG 8 ER PT J AU Yuan, K Listinsky, MC Singh, KR Listinsky, JJ Siegal, PG AF Yuan, Kun Listinsky, M Catherine Singh, K Raj Listinsky, J Jay Siegal, P Gene TI Cell Surface Associated Alpha-L-Fucose Moieties Modulate Human Breast Cancer Neoplastic Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Fucose; Glycosylation; Matrix metalloproteases; Neoplastic progression ID Breast cancer; Fucose; Glycosylation; Matrix metalloproteases; Neoplastic progression AB Glycosylation drives critical processes important for mammalian cell–cell and cell–matrix interactions. Alpha-L-fucose (α-L-f) is a key monosaccharide component of oligosaccharides that has been found to be overexpressed during tumor progression. Modification of cell surface fucosylation, we hypothesized, alters tumor cell phenotype and function at the end of the neoplastic progression cascade including tumor invasion. Alpha-L-fucosidase (α-L-fase) is a glycosidase that specifically removes (α-L-f) from oligosaccharide sites. We first verified the effectiveness of the α-L-fase to specifically decrease the level of α-L-f on the cell surface of several human breast cancer cell lines and also examined the recovery time for these cells to repopulate their surfaces. To investigate the potential effect of defucosylation on tumor functions, we studied the proliferation, and invasion in vitro of human breast cancer MDA-MB-231 cells as the representative cell model. We further examined several fucose-associated molecules previously shown to be involved in tumor progression, including CD44 and CD15 (Lewis X antigen). We found that α-L-fase pretreatment significantly decreased the invasive capability of breast cancer cells. Deoxyfuconojirimycin (DFJ), a specific α-L-fase inhibitor, reversed this effect. After fucosidase treatment, the level of both CD15 and CD44 were found to be reduced as measured by flow cytometry. α-L-fase treatment, further, did not affect tumor cell proliferation in vitro under identical experimental conditions. Gelatin zymography of conditioned media from tumor cells treated with α-L-fase demonstrated no change in MMP-2 activity while MMP-9 was significantly reduced. In summary, fucose containing glycans were found widely distributed on the cell surface of breast cancer cells and could be effectively removed by α-L-fase treatment. This decreased fucosylation, in turn, was seen to impair the interaction between tumor cells and extracellular matrices, and thus affected key cell functions modulating tumor invasion. Further elucidation of the molecular pathways involved in the inhibition of tumor cell invasion may suggest a rationale for the use of glycobiologic therapeutics to deter tumor progression. C1 [Yuan, Kun] University of Alabama at Birmingham, Department of Pathology, 35233 Birmingham, AL, USA. [Listinsky, M Catherine] University of Alabama at Birmingham, Department of Pathology, 35233 Birmingham, AL, USA. [Singh, K Raj] University of Alabama at Birmingham, Department of Pathology, 35233 Birmingham, AL, USA. [Listinsky, J Jay] University of Alabama at Birmingham, Department of Diagnostic Radiology, 35233 Birmingham, AL, USA. [Siegal, P Gene] University of Alabama at Birmingham, Department of Pathology, 35233 Birmingham, AL, USA. RP Siegal, PG (reprint author), University of Alabama at Birmingham, Department of Pathology, 35233 Birmingham, USA. EM gsiegal@uab.edu CR Dennis JW, Granovsky M, Warren CE, 1999, Glycoprotein glycosylation and cancer progression. Biochim Biophys Acta 1473(1):21–34 Listinsky JJ, Listinsky CM, Alapati V, et al, 2001, Cell surface fucose ablation as a therapeutic strategy for malignant neoplasms. Adv Anat Pathol 8(6):330–337 Mackinnon WB, Delbridge L, Russell P, et al, 1996, Twodimensional proton magnetic resonance spectroscopy for tissue characterization of thyroid neoplasms. World J Surg 20(7):841–847 Mackinnon WB, Russell P, May GL, et al, 1995, Characterization of human ovarian epithelial tumors, ex vivo, by proton magnetic resonance spectroscopy. Int J Gynecol Cancer 5(3):211–221 Fernandez-Rodriguez J, Paez de la Cadena M, Martinez-Zorzono VS, et al, 1997, Fucose levels in sera and in tumours of colorectal adenocarcinoma patients. Cancer Lett 121(2):147–153 Huang W, Li X, 2000, Significance of fucose expression in lung carcinoma and their brain metastases. Zhonghua Bing Li Xue Za Zhi 29(4):259–262 Patel PS, Baxi BR, Adhvaryu SG, et al, 1990, Evaluation of serum sialic acid, heat stable alkaline phosphatase and fucose as markers of breast carcinoma. Anticancer Res 10(4):1071–1074 Isacke CM, Yarwood H, 2002, The hyaluronan receptor, CD44. Int J Biochem Cell Biol 34(7):718–721 Bourguignon LY, 2001, CD44-mediated oncogenic signaling and cytoskeleton activation during mammary tumor progression. J Mammary Gland Biol Neoplasia 6(3):287–297 Stetler-Stevenson WG, 2001, The role of matrix metalloproteinases in tumor invasion, metastasis, and angiogenesis. Surg Oncol Clin N Am 10(2):383–392 Makita M, 1991, [Relationship between the prognosis and the morphology of stromal invasion of the breast cancer]. Nippon Geka Gakkai Zasshi 92(8):1001–1009 van 't Veer LJ Dai H, van de Vijver MJ, et al., 2002, Gene expression profiling predicts clinical outcome of breast cancer. Nature 415(6871):530–536 Nishimura Y, 2003, gem-Diamine 1-N-iminosugars and related iminosugars, candidate of therapeutic agents for tumor metastasis. Curr Top Med Chem 3(5):575–591 Siegal GP Wang MJ, Rinehart CA, et al, 1993, Development of a novel human extracellular matrix for quantitation of the invasiveness of human cells. Cancer Lett 69(2):123–132 Singh RK Rinehart CA, Kim JP, et al, 1996, Tumor cell invasion of basement membrane in vitro is regulated by amino acids. Cancer Invest 14(1):6–18 Schwartz GG Wang MH, Zang M, et al, 1997, 1 α,25- Dihydroxyvitamin D, calcitriol, inhibits the invasiveness of human prostate cancer cells. Cancer Epidemiol Biomarkers Prev 6(9):727–732 Winchester B Barker C, Baines S, et al., 1990, Inhibition of alpha-L-fucosidase by derivatives of deoxyfuconojirimycin and deoxymannojirimycin. Biochem J 265(1):277–282 Orntoft TF, Vestergaard EM, 1999, Clinical aspects of altered glycosylation of glycoproteins in cancer. Electrophoresis 20, 2):362–371 Couldrey C, Green JE, 2000, Metastases: the glycan connection. Breast Cancer Res 2(5):321–323 Laidler P, Litynska A, 1997, Tumor cell N-glycans in metastasis. Acta Biochim Pol 44(2):343–357 Ono M, Hakomori S, 2004, Glycosylation defining cancer cell motility and invasiveness. Glycoconj J 20(1):71–78 Carcel-Trullols J Stanley JS, Saha R, et al, 2006, Characterization of the glycosylation profile of the human breast cancer cell line, MDA-231, and a bone colonizing variant. Int J Oncol 28(5): 1173–1183 Takahashi N Iwahori A, Breitmon TR, et al, 1997, Tunicamycin in combination with retinoic acid synergistically inhibits cell growth while decreasing palmitoylation and enhancing retinoylation of proteins in the human breast cancer cell line MCF-7. Oncol Res 9(10):527–533 Carlberg M Dricu A, Blegen H, et al, 1996, Short exposures to tunicamycin induce apoptosis in SV40-transformed but not in normal human fibroblasts. Carcinogenesis 17(12):2589–2596 Ciolczyk-Wierzbicka D, Amoresano A, Casbarr A, et al, 2004, The structure of the oligosaccharides of N-cadherin from human melanoma cell lines. Glycoconj J 20(7–8):483–492 Becker DJ, Lowe JB, 2003, Fucose: biosynthesis and biological function in mammals. Glycobiology 13(7):41R–53R Kiricuta I, Bojan O, Comes R, et al, 1979, Significance of serum fucose, sialic acid, haptoglobine and phospholipids levels in the evolution and treatment of breast cancer. Arch zeschwulstforsch 49(2):106–112 Natsume A, Wakitani M, Yamane-Ohnuki N, et al, 2005, Fucose removal from complex-type oligosaccharide enhances the antibody- dependent cellular cytotoxicity of single-gene-encoded antibody comprising a single-chain antibody linked the antibody constant region. J Immunol Methods 306(1–2):93–103 Thompson S Cantwell BM, Matta KI, et al., 1992, Parallel changes in the blood levels of abnormally-fucosylated haptoglobin and alpha 1,3 fucosyltransferase in relationship to tumour burden: more evidence for a disturbance of fucose metabolism in cancer. Cancer Lett 65(2):115–121 Ding KF, Zheng S, 2004, Study on relationship of fucosyltransferase gene types in breast cancer with metastasis and prognosis. Zhonghua Wai Ke Za Zhi 42(9):546–550 Hiller KM Mayben JP, Bendt KM et al, 2000, Transfection of alpha(1,3)fucosyltransferase antisense sequences impairs the proliferative and tumorigenic ability of human colon carcinoma cells. Mol Carcinog 27(4):280–288 Liu F Zhang Y, Zhang XY, et al, 2002, Transfection of the nm23- H1 gene into human hepatocarcinoma cell line inhibits the expression of sialyl Lewis X, alpha1,3 fucosyltransferase VII, and metastatic potential. J Cancer Res Clin Oncol 128(4):189–196 Hakomori S, 2002, Glycosylation defining cancer malignancy: new wine in an old bottle. Proc Natl Acad Sci USA 99, 16):10231–10233 Johnson SW, Alhadeff JA, 1991, Mammalian alpha-L-fucosidases. Comp Biochem Physiol B 99(3):479–488 Fernandez-Rodriguez J, Ayude D, de la Cadena MP, et al, 2000, Alpha-L-fucosidase enzyme in the prediction of colorectal cancer patients at high risk of tumor recurrence. Cancer Detect Prev 24, 2):143–149 Hakomori S, 2001, Tumor-associated carbohydrate antigens defining tumor malignancy: basis for development of anti-cancer vaccines. Adv Exp Med Biol 491:369–402 Capurro M Ballore C, Bover L, et al., 1999, Differential lytic and agglutinating activity of the anti-Lewis(x, monoclonal antibody FC-2.15 on human polymorphonuclear neutrophils and MCF-7 breast tumor cells. In vitro and ex vivo studies. Cancer Immunol Immunother 48(2–3):100–108 Gu H, Ni C, Zhan R, 2000, The expression of CD15 mRNA CD44v6 mRNA and nm23H1 mRNA in breast cancer and their clinical significance. Zhonghua Yi Xue Za Zhi 80(11):854–857 Brooks SA, Leathem AJ, 1995, Expression of the CD15 antigen, Lewis x, in breast cancer. Histochem J 27(9):689–693 Madjd Z Parsons T, Watson NF, et al., 2005, High expression of Lewis y/b antigens is associated with decreased survival in lymph node negative breast carcinomas. Breast Cancer Res 7(5):R780–787 Nakagoe T, Fukushima K, Tuji T, et al., 1998, Immunohistochemical expression of ABH/Lewis-related antigens in primary breast carcinomas and metastatic lymph node lesions. Cancer Detect Prev 22(6):499–505 Reitman ML, Trowbridge IS, Kornfeld S, 1980, Mouse lymphoma cell lines resistant to pea lectin are defective in fucose metabolism. J Biol Chem 255(20):9900–9906 Ripka JA, Adamany A, Stanley P, 1986, Two Chinese hamster ovary glycosylation mutants affected in the conversion of GDPmannose to GDP-fucose. Arch Biochem Biophys 249(2):533–545 Dabelsteen E, 2002, ABO blood group antigens in oral mucosa. What is new?. J Oral Pathol Med, 31(2):65–70 Jones JL, Glynn P, Walker RA, 1999, Expression of MMP-2 and MMP-9, their inhibitors, and the activator MT1-MMP in primary breast carcinomas. J Pathol 189(2):161–168 Morini M, Mottolese M, Ferrari N, et al, 2000, The alpha 3 beta 1 integrin is associated with mammary carcinoma cell metastasis, invasion, and gelatinase B, MMP-9, activity. Int J Cancer 87, 3):336–342 Xiong L, Andrews D, Regnier F, 2003, Comparative proteomics of glycoproteins based on lectin selection and isotope coding. J Proteome Res 2(6):618–625 Gasbarri A, Del Prete F, Girnita L, et al, 2003, CD44s adhesive function spontaneous and PMA-inducible CD44 cleavage are regulated at post-translational level in cells of melanocytic lineage. Melanoma Res 13(4):325–337 Catterall JB, Jones LM, Turner GA, 1999, Membrane protein glycosylation and CD44 content in the adhesion of human ovarian cancer cells to hyaluronan. Clin Exp Metastasis 17(7):583–591 Skelton TP Zeng C, Nocks A, et al., 1998, Glycosylation provides both stimulatory and inhibitory effects on cell surface and soluble CD44 binding to hyaluronan. J Cell Biol 140(2):431–446 Bourguignon LY, Zhu D, Zhu H, 1998, CD44 isoform-cytoskeleton interaction in oncogenic signaling and tumor progression. Front Biosci 3:d637–d649 Matsuki H, Yonezawa K, Obata K, et al., 2003, Monoclonal antibodies with defined recognition sequences in the stem region of CD44: detection of differential glycosylation of CD44 between tumor and stromal cells in tissue. Cancer Res, 63(23):8278–8283 Hakomori S, 2004, Glycosynapses: microdomains controlling carbohydrate-dependent cell adhesion and signaling. An Acad Bras Cienc 76(3):553–572 Evans E Leung A, Heinrich V, et al., 2004, Mechanical switching and coupling between two dissociation pathways in a P-selectin adhesion bond. Proc Natl Acad Sci U SA 101(31):11281–11286 Rampal R, Arboleda-Velsquez JF, Nita-Lazar A, et al, 2005, Highly conserved O-fucose sites have distinct effects on Notch1 function. J Biol Chem 280(37):32133–32140 Schachter H, 2005, The search for glycan function: fucosylation of the TGF-beta1 receptor is required for receptor activation. Proc Natl Acad Sci U S A 102(44):15721–15722 Gornik O Dumic J, Flogel M, et al, 2006, Glycoscience—a new frontier in rational drug design. Acta Pharm 56(1):19–30 Kannagi R, 1997, Carbohydrate-mediated cell adhesion involved in hematogenous metastasis of cancer. Glycoconj J, 14(5):577–584 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 145 EP 156 DI 10.1007/s12253-008-9036-x PG 12 ER PT J AU Abdul, M Ramlal, S Hoosein, N AF Abdul, Mansoor Ramlal, Sian Hoosein, Naseema TI Ryanodine Receptor Expression Correlates with Tumor Grade in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ryanodine receptor; Breast cancer; Tumor grade; 4-chloro-m-cresol; MCF-7; MDA-MB-231 ID Ryanodine receptor; Breast cancer; Tumor grade; 4-chloro-m-cresol; MCF-7; MDA-MB-231 AB Ryanodine receptors (RyRs) have been previously implicated in the proliferation of human T-lymphocytes and melanocytes as well as in the motility of astrocytes. We have examined RyR expression in 57 ductal, human breast cancer specimens, by immunohistochemistry. Moderate to high RyR immunostaining was found in 47 (82%) of the specimens. There was a direct correlation between RyR levels and tumor grade (r=0.48, p=0.0002). We have also examined the effect of the RyR agonist 4-chloro-m-cresol on the in-vitro growth of two human breast cancer cell lines, MCF-7 and MDA-MB-231. Treatment with 4-chlorom-cresol inhibited the growth of both breast cancer cell lines, in a dose-dependent manner, with half-maximal inhibition observed at 30 to 50 μg/mL (210–351 μM). Our data suggest that RyR could serve as prognostic indicator and/or as a target for breast cancer treatment. C1 [Abdul, Mansoor] Claflin University, Biology Department, 400 Magnolia Street, 29115 Orangeburg, SC, USA. [Ramlal, Sian] Claflin University, Biology Department, 400 Magnolia Street, 29115 Orangeburg, SC, USA. [Hoosein, Naseema] Claflin University, Biology Department, 400 Magnolia Street, 29115 Orangeburg, SC, USA. RP Hoosein, N (reprint author), Claflin University, Biology Department, 29115 Orangeburg, USA. EM nhoosein@claflin.edu CR Bootman MD, Lipp P, Berridge MJ, 2001, The organization and functions of local Ca(2+, signals. J Cell Sci 114:2213–2222 Mackrill JJ, 1999, Protein–protein interactions in intracellular Ca2+-release channel function. Biochem J 337:345–361 Fill M, Copello JA, 2002, Ryanodine receptor calcium release channels. Physiol Rev 82:893–922 Hakamata Y, Nishimura S, Nakai J et al, 1994, Involvement of the brain type of ryanodine receptor in T-cell proliferation. FEBS Lett 352:206–210 Kang HY, Kim NS, Lee CO et al, 2000, Expression and function of ryanodine receptors in human melanocytes. J Cell Physiol 185:200–206 Matyash M, Matyash V, Nolte C et al, 2002, Requirement of functional ryanodine receptor type 3 for astrocyte migration. FASEB J 16:84–86 Mariot P, Prevarskaya N, Roudbaraki M et al, 2000, Evidence of functional ryanodine receptor involved in apoptosis of prostate cancer, LNCaP, cells. Prostate 43:205–214 Timar J, Kasler M, Katai J et al, 2006, Developments in cancer management by innovative genomics: 2006 report of the National Cancer Consortium. Hungarian Oncology 50:349–359 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 157 EP 160 DI 10.1007/s12253-008-9045-9 PG 4 ER PT J AU Palka, I Kelemen, Gy Ormandi, K Lazar, Gy Nyari, T Thurzo, L Kahan, Zs AF Palka, Istvan Kelemen, Gyongyi Ormandi, Katalin Lazar, Gyorgy Nyari, Tibor Thurzo, Laszlo Kahan, Zsuzsanna TI Tumor Characteristics in Screen-Detected and Symptomatic Breast Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adjuvant therapy; Casting calcifications; Mammographic screening; Mammographic image; Prognosis ID Adjuvant therapy; Casting calcifications; Mammographic screening; Mammographic image; Prognosis AB The natural course of early breast cancer has changed as a result of the introduction of mammographic screening. The present aim was a prospective analysis of screen-detected and symptomatic operable breast cancers in the era of mammographic service screening. The mode of detection (screen-detected, symptomatic or interval cancer), the type of mammographic image and other characteristics (the invasive tumor size, histological tumor type, grade, nodal, hormone receptor and HER2 status and the presence of lymphovascular invasion) of 569 invasive breast cancers were studied. Screen-detected cancers were significantly more frequently of grade I, <10 mm of size and nodenegative (p<0.001, respectively). Symptomatic/interval cancers were significantly more frequently of grade 3, >20 mm of size (p<0.001), and exhibited lymphovascular invasion (p=0.001). Screening-detection of the tumor favored breast-conserving surgery, sentinel lymph node biopsy and the avoidance of chemotherapy (p<0.001). Cancers associated with casting-type calcifications on the mammogram were typically of ductal type (p=0.043), of grade 2–3, estrogen receptor and progesterone receptornegative and HER2-positive (p<0.001). Interval cancers occurred significantly more often at a younger age and remained mammographically occult as compared with other cancers. Mammographic screen-detected cancers demonstrate more favorable prognostic features, and need less extensive treatment than symptomatic or interval cancers. The mammographic appearance of the tumor reflects its biological behavior, and this should be considered in the management optimization. C1 [Palka, Istvan] University of Szeged, Department of PathologySzeged, Hungary. [Kelemen, Gyongyi] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Ormandi, Katalin] University of Szeged, Department of RadiologySzeged, Hungary. [Lazar, Gyorgy] University of Szeged, Department of SurgerySzeged, Hungary. [Nyari, Tibor] University of Szeged, Department of Medical InformaticsSzeged, Hungary. [Thurzo, Laszlo] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. RP Kahan, Zs (reprint author), University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. EM kahan@onko.szote.u-szeged.hu CR Tabar L, Fagerberg G, Duffy SW et al., 1989, The Swedish twocounty trial of mammographic screening for breast cancer: recent results and calculation of benefit. J Epidemiol Community Health 43:107–114 Tabar L, Yeng M-F, Vitak B et al., 2003, Mammography service screening and mortality in breast cancer patients: 20-year followup before and after introduction of screening. Lancet 361:1405– 1410 The Swedish Organised Service Screening Evaluation Group, 2006, Reduction of breast cancer mortality from organised service screening with mammography: 1. Further confirmation with extended data. Cancer Epidemiol Biomarkers Prev 15:45–51 Miller AB, Baines C, To T et al., 1992, Canadian national breast screening study. 1. Breast cancer detection and death rates among women aged 40 to 49 years. CMAJ 147:1459–1476 Kerlikowske K, Grady D, Rubin SM et al., 1995, Efficacy of screening mammography. A meta-analysis. JAMA 273:149–154 Cronin KA, Feuer EJ, Clarke LD, et al., 2006, Impact of adjuvant therapy and mammography on U.S. mortality from 1975 to 2000. Comparison of mortality results from the CISNET breast cancer base case analysis. J Natl Cancer Inst 36:112–121 Moody-Ayers SY, Wells CK, Feinstein AR, 2000, ”Benign” tumors and ”early detection” in mammography-screened patients of a natural cohort with breast cancer. Arch Intern Med 160:1109– 1115 Klemi PJ, Joensuu H, Tikkanen S et al., 1992, Aggressiveness of breast cancers found with and without screening. Br Med J 304:467–469 Klemi PJ, Parvinen I, Pylkkanen L et al., 2003, Significant improvement in breast cancer survival through population-based mammography screening. Breast 12:308–313 Anttinen J, Kautiainen H, Kuopo T, 2006, Role of mammography screening as a predictor of survival in postmenopausal breast cancer patients. Br J Cancer 94:147–151 Joensuu H, Lehtimaki T, Holli K et al., 2004, Risk for distant recurrence of breast cancer detected by mammography screening or other methods. JAMA 292:1064–1073 Shen Y, Yang Y, Inoues LYT et al., 2005, Role of detection method in predicting breast cancer survival: Analysis of randomized screening trials. J Natl Cancer Inst 97:1195–1203 Gill PG, Farshid G, Luke CG et al., 2005, Detection by screening mammography is a powerful independent predictor of survival in women diagnosed with breast cancer. Breast 13:15–22 ErnstMF, Voogd AC, Goebergh JWWet al., 2004, Breast carcinoma diagnosis, treatment, and prognosis before and after the introduction of mass mammographic screening. Cancer 100:1337–1344 Tabar L, Dean PB, Kaufman CS et al., 2000, A new era in the diagnosis of breast cancer. Surg Oncol Clinics NA 9:233–277 Tabar L, Chen TH, Yen AM, et al., 2004, Mammographic tumor features can predict long-term outcomes reliably in women with 1–14-mm invasive breast carcinoma. Cancer 101:1745–1759 Thurfjell E, Thurfjell MG, Lindgren A, 2001, Mammographic finding as predictor of survival in 1–9 mm invasive breast cancers. Worse prognosis for cases presenting as calcifications alone. Breast Cancer Res Treat 67:177–180 Alexander MC, Yankaskas BC, Biesemier KW, 2006, Association of stellate mammographic pattern with survival in small invasive breast tumors. Am J Roentgenol 187:29–73 Peacock C, Given-Wilson RM, Duffy SW, 2004, Mammographic casting-type calcification associated with small screen-detected invasive breast cancers: is this a reliable prognostic indicator? Clin Radiol 59:165–170 Palka I, Ormandi K, Gaal S et al., in press, Casting-type calcifications on the mammogram indicate a higher probability of early relapse and death among operable high-risk breast cancer patients. Acta Oncol Perry NM, 2001, Quality assurance in the diagnosis of breast disease. EUSOMA Working Party. Eur J Cancer 37:159–172 Ravdin PM, Siminoff LA, Davis GJ et al., 2001, Computer program to assist in making decisions about adjuvant therapy for women with early breast cancer. J Clin Oncol 19:980–991 Olivotto IA, Bajdik CD, Ravdin PM, et al., 2005, Populationbased validation of the prognostic model ADJUVANT! for early breast cancer. J Clin Oncol 23:2716–2725 Porter PJ, El-Bastawissi AZ, Mandelson MT et al., 1999, Breast tumor characteristics as predictors of mammographic detection: Comparison of interval- and screen-detected cancers. J Natl Cancer Inst 91:2020–2028 Gilliland FD, Joste N, Stauber PM et al., 2000, Biologic characteristics of interval and screen-detected breast cancers. J Natl Cancer Inst 92:743–749 Crosier M, Scott D, Wilson RG et al., 1999, Differences in Ki67 and c-erbB2 expression between screen-detected and true interval breast cancers. Clin Cancer Res 5:2682–2688 Porter GJ, Evans AJ, Cornford EJ et al., 2007, Influence of mammographic parenchymal pattern in screen-detected and interval invasive breast cancers on pathologic features, mammographic features, and patient survival. Am J Roentgenol 188:676– 683 Porter GJ, Evans AJ, Burrell HC et al., 2007, NHSBSP type 1 interval cancers: a scientifically valid grouping? Clin Radiol 62:262–267 Zunzunegui RG, Chung MA, Oruwari J et al., 2003, Casting-type calcifications with invasion and high-grade ductal carcinoma in situ. Arch Surg 138:537–540 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 161 EP 167 DI 10.1007/s12253-008-9010-7 PG 7 ER PT J AU Pusztai, L AF Pusztai, Lajos TI Preoperative Systemic Chemotherapy and Pathologic Assessment of Response SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Preoperative systemic therapy; Breast cancer ID Preoperative systemic therapy; Breast cancer AB Preoperative systemic (neoadjuvant) chemotherapy is both routine therapeutic modality for locally advanced breast cancer and a translational research model to identify biomarkers that predict treatment response. It is imperative that pathologic response be strongly prognostic in order to optimize the clinical and scientific information that can be gained from neoadjuvant clinical trials. Dichotomization of response as pathologic complete response (pCR) or residual disease (RD) is overly simplistic for these objectives, particularly because residual disease (RD) after neoadjuvant treatment includes a broad range of actual responses from near-pCR to frank resistance. More effective or prolonged neoadjuvant treatments should reduce the extent of RD in many patients, possibly blurring the prognostic distinction between pCR and RD. On the other hand, it should be possible to identify patients with resistant disease in order to develop predictive tests for this adverse outcome. Our research group recently proposed to measure residual cancer burden (RCB) as a continuous variable derived from the primary tumor dimensions, cellularity of the tumor bed, and axillary nodal burden. Each component contributes meaningful pathologic information and can be obtained using routine pathologic materials and methods of interpretation that could easily be implemented in routine diagnostic practice. C1 [Pusztai, Lajos] The University of Texas, M. D. Anderson Cancer Center, Department of Breast Medical Oncology, 77230-1439 Houston, TX, USA. RP Pusztai, L (reprint author), The University of Texas, M. D. Anderson Cancer Center, Department of Breast Medical Oncology, 77230-1439 Houston, USA. EM lpusztai@mdanderson.org CR Rastogi P, Anderson SJ, Bear HD, Geyer CE, Kahlenberg MS, Robidoux A, Margolese RG, Hoehn JL, Vogel VG, Dakhil SR, Tamkus D, King KM, Pajon ER, Wright MJ, Robert J, Paik S, Mamounas EP, Wolmark N, 2008, Preoperative chemotherapy: updates of national surgical adjuvant breast and bowel project protocols B-18 and B-27. J Clin Oncol 26:778–785 Boughey JC, Peintinger F, Meric-Bernstam F, Perry AC, Hunt KK, Babiera GV, Singletary SE, Bedrosian I, Lucci A, Buzdar AU, Pusztai L, Kuerer HM, 2006, Impact of preoperative versus postoperative chemotherapy on the extent and number of surgical procedures in patients treated in randomized clinical trials for breast cancer. Ann Surg 244:464–470 Bonadonna G, Veronesi U, Brambilla C et al, 1990, Primary chemotherapy to avoid mastectomy in tumors with diameters of three centimeters or more. J Natl Cancer Inst 82:1539–1545 Mauri D, Pavlidis N, Ioannidis JP, 2005, Neoadjuvant versus adjuvant systemic treatment in breast cancer: A meta-analysis. J Natl Cancer Inst 97:188–194 Chen AM, Meric-Bernstam F, Hunt KK et al, 2004, Breastconserving therapy after neoadjuvant chemotherapy: The M. D. Anderson Cancer Center experience. J Clin Oncol 22:2303–2312 Fisher B, Bryant J, Wolmark N et al, 1998, Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol 16:2672–2685 Smith IC, Heys SD, Hutcheon AW, Miller ID, Payne S, Gilbert FJ, Ah-See AK, Eremin O, Walker LG, Sarkar TK et al, 2002, Neoadjuvant chemotherapy in breast cancer: significantly enhanced response with docetaxel. J Clin Oncol 20:1456–1466 von Minckwitz G, Kummel S, Vogel P et al, 2008, Neoadjuvant vinorelbine–capecitabine versus docetaxel–doxorubicin–cyclophosphamide in early nonresponsive breast cancer: phase III randomized GeparTrio trial. J Natl Cancer Inst 100:542–551 Wolff AC, Berry D, Carey LA, Colleoni M, Dowsett M, Ellis M, Je G, Mankoff D, Paik S, Pusztai L, Smith ML, Zujewski JA, 2008, Research issues affecting preoperative systemic therapy for operable breast cancer. J Clin Oncol 26:806–813 Carey LA, Metzger R, Dees EC et al, 2005, American Joint Committee on Cancer tumor–node–metastasis stage after neoadjuvant chemotherapy and breast cancer outcome. J Natl Cancer Inst 97:1137–1142 Kuerer HM, Newman LA, Smith TL et al, 1999, Clinical course of breast cancer patients with complete pathologic primary tumor and axillary lymph node response to doxorubicin-based neoadjuvant chemotherapy. J Clin Oncol 17:460–469 Rouzier R, Extra JM, Klijanienko J et al, 2002, Incidence and prognostic significance of complete axillary downstaging after primary chemotherapy in breast cancer patients with T1 to T3 tumors and cytologically proven axillary metastatic lymph nodes. J Clin Oncol 20:1304–1310 Mazouni C, Peintinger F, Kau SW, Andre F, Gonzalez-Angulo AM, Symmans WF, Meric-Bernstam F, Valero V, Hortobagyi GN, Pusztai L, 2007, Residual ductal carcinoma in situ in patients with complete eradication of invasive breast cancer after neoadjuvant chemotherapy does not adversely affect patient outcome. J Clin Oncol 25:2650–2655 Symmans WF, Peintinger F, Hatzis C, Rajan R, Kuerer HM, Valero V, Assad L, Poniecka A, Hennessy TJ, Green MC, Buzdar AU, Singletary SE, Hortobagyi GN, Pusztai L, 2007, Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol 25:4414–4422 Esteva FJ, Hortobagyi GN, 2008, Can early response assessment guide neoadjuvant chemotherapy in early-stage breast cancer? J Natl Cancer Inst 100:521–523 Rouzier R, Pusztai L, Delaloge S, Gonzalez-Angulo AM, Andre F, Hess KR, Buzdar AU, Garbay JR, Spielmann M, Mathieu MC, Symmans WF, Wagner P, Atallah D, Valero V, Berry DA, Hortobagyi GN, 2005, Nomograms to predict pathologic complete response and metastasis-free survival after preoperative chemotherapy for breast cancer. J Clin Oncol 23:8331–8339 Andre F, Mazouni C, Liedtke C, Kau SW, Frye D, Green M, Gonzalez-Angulo AM, Symmans WF, Hortobagyi GN, Pusztai L, 2008, HER2 expression and efficacy of preoperative paclitaxel/ FAC chemotherapy in breast cancer. Breast Cancer Res Treat 108:183–190 Gianni L, Zambetti M, Clark K, Baker J, Cronin M, Wu J, Mariani G, Rodriguez J, Carcangiu M, Watson D, Valagussa P, Rouzier R, Symmans WF, Ross JS, Hortobagyi GN, Pusztai L, Shak S, 2005, Gene expression profiles in paraffin-embedded core biopsy tissue predict response to chemotherapy in women with locally advanced breast cancer. J Clin Oncol 23:7265–7277 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 169 EP 171 DI 10.1007/s12253-008-9070-8 PG 3 ER PT J AU Tot, T Gere, M AF Tot, Tibor Gere, Maria TI Radiological–Pathological Correlation in Diagnosing Breast Carcinoma: The Role of Pathology in the Multimodality Era SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Breast; Breast cancer; Sick lobe hypothesis; Pathology; Radiological–pathological correlation; Large section technique ID Breast; Breast cancer; Sick lobe hypothesis; Pathology; Radiological–pathological correlation; Large section technique AB Breast carcinoma is a lobar disease, as the simultaneously or asynchronously appearing often multiple tumor foci originate from a single sick breast lobe. In its initial phase, the spatial pattern of malignant transformation may be lobar (targeting the entire lobe), segmental (targeting a segment) or terminal (targeting distant terminal ductal-lobular units) within the sick lobe. All these variations are properly characterized by the following parameters: the extent of the disease (the volume of the tissue containing all the actually present malignant structures within the breast), the distribution of the lesions within this tissue (unifocal, multifocal or diffuse, separately for in situ and invasive component), the size of the tumor (corresponding to the largest diameter of the largest invasive focus) and the exact localization of the lesion(s). In addition, intra- and intertumoral heterogeneity have to be noticed, if evident. Combining the results of different imaging modalities (mammography, ultrasound, magnetic resonance imaging) the radiologist may compensate the limitations of individual methods. This multimodality approach leads to more accurate radiological size measurement, more accurate assessment of the distribution of the lesions and disease extent. This represents a challenge for pathologists as the traditional histopathology method based on fragmentation and sampling of macroscopically suspicious lesion(s) is clearly insufficient for modern postoperative radiological–pathological correlation. There is a clear need for more complete examination of the excised tissue and for a three-dimensional reconstruction of the finding, preferably using continuous large tissue slices and two and three-dimensional large-format histological sections. Discordant results may still appear as a consequence of failure in radiological–pathological correlation or related to certain tumor subtypes as invasive lobular carcinoma of diffuse type, low grade in situ lesions or micropapillary ductal in situ carcinoma. C1 [Tot, Tibor] Central Hospital, Department of Pathology and Clinical CytologyFalun, Sweden. [Gere, Maria] Central Hospital, Department of Pathology and Clinical CytologyFalun, Sweden. RP Tot, T (reprint author), Central Hospital, Department of Pathology and Clinical Cytology, Falun, Sweden. EM tibor.tot@ltdalarna.se CR Tot T, 2005, DCIS, cytokeratins and the theory of the sick lobe. Virchows Arch 447:1–8 Tot T, 2007, The theory of the sick lobe and the possible consequences. Int J Surg Pathol 15(4):369–375 Tot T, 2007, The clinical relevance of the distribution of the lesions in 500 consecutive breast cancer cases documented in large-format histological sections. Cancer 110:2551–2560 Going JJ, Mohun TJ, 2006, Human breast duct anatomy, the ‘sick lobe’ hypothesis and intraductal approaches to breast cancer. Breast Cancer Res Treat 97:285–291 Foschini MP, Flaminio F, Miglio R et al, 2007, The impact of large sections on the study of in situ and invasive duct carcinoma of the breast. Hum Pathol 38:1736–1743 Tot T, Tabar L, Dean PB, 2002, Practical breast pathology. Thieme, Stuttgart—New York Tabar L, Tot T, Dean PB, 2005, Breast cancer: the art and science of early detection with mammography: perception, interpretation, histopathologic correlation. Thieme, Stuttgart—New York Tot T, Tabar L, 2006, Radiologic–pathologic correlation of ductal carcinoma in situ of the breast using two- and three-dimensional large histologic sections. Semin Breast Dis 8:144–151 Holland R, Hendricks JH, Vebeek AL et al, 1990, Extent, distribution, and mammographic/histological correlation of breast ductal carcinoma in situ. Lancet 335:519–522 Faverly DRG, Hendricks JHCL, Holland R, 2001, Breast carcinoma of limited extent. Frequency, radiologic–pathologic characteristics, and surgical margin requirements. Cancer 91:647– 659 Van Goethem M, Schelfout K, Dijkmans L et al, 2004, MR mammography in the pre-operative staging of breast cancer in patients with dense breast tissue: comparison with mammography and ultrasound. Eur Radiol 14:809–816 Berg WA, Gutierrez L, NessAiver MS et al, 2004, Diagnostic accuracy of mammography, clinical examination, US, and MRI imaging in preoperative assessment of breast cancer. Radiology 233:830–849 Tabar L, Tot T, Dean PB, 2007, Breast cancer: early detection with mammography. casting type calcifications: sign of a subtype with deceptive features. Thieme, Stuttgart-New York Bosch AM, Kessels AG, Beets GL et al, 2003, Preoperative estimation of the pathological breast tumor size by physical examination, mammography and ultrasound: a prospective study of 105 invasive tumors. Eur J Radiol 48:285–292 Skaane P, Skjorten F, 1999, Ultrasonographic evaluation of invasive lobular carcinoma. Acta Radiol 40:369–375 Moon WK, Noh DY, Im JG, 2002, Multifocal, multicentric, and contralateral breast cancers: bilateral whole-breast US in the preoperative evaluation of patients. Radiology 224:569– 576 Selinko VL, Middleton LP, Dempsey PJ, 2004, Role of sonography in diagnosing and staging invasive lobular carcinoma. J Clin Ultrasound 32:323–332 Winstein SP, Orel SG, Heller R et al, 2001, MR imaging of the breast in patients with invasive lobular carcinomas. Am J Roentgenol 176:399–406 Schelfout K, Van Goethem M, Kersschot E et al, 2004, Preoperative breast MRI in patients with invasive lobular breast cancer. Eur Radiol 14:1209–1216 Neubauer H, Li M, Kuhne-Heid R et al, 2003, High grade and non-high grade ductal carcinoma in situ on dynamic MR mammography: characteristic findings for signal increase and morphological pattern of enhancement. Br J Radiol 76:3–12 Szabo BK, Aspelin P, Kristoffersen Wiberg A et al, 2003, Invasive breast cancer: correlation of dynamic MR features with prognostic factors. Eur Radiol 13:2425–2435 Deurloo EE, Klein Zeggelink WFA, Teerstra HJ et al, 2006, Contrast-enhanced MRI in breast cancer patients eligible for breast-conserving therapy: complementary value for subgroups of patients. Eur Radiol 16:692–701 Tot T, Tabar L, Dean PB, 2000, The pressing need for better mammographic–pathologic correlation of many variations in normal breast anatomy. Virchows Arch 437:338–344 Carlson JW, Birdwell RL, Gombos EC et al, 2007, MRI-directed, wire-localized breast excisions: incidence of malignancy and recommendations for pathologic evaluation. Hum Pathol 32:1754–1759 Luttges J, Kalbfleisch H, Prinz P, 1987, Nipple involvement and multicentricity in breast cancer. A study of whole organ sections. J Cancer Res Clin Oncol 113:481–487 Fochsini MP, Tot T, Eusebi V, 2002, Large section, macrosection, histologic slides. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. 2nd edn. Lippincott, Williams and Wilkins, Philadelphia, pp 249–254 Jackson PA, Merchant W, McCormick CJ et al, 1994, A comparison of large block macrosectioning and conventional techniques in breast pathology. Virchows Arch 425:243–248 Biesemier KW, Alexander CM, 2005, Enhancement of mammographic– pathologic correlation utilizing large format histology for malignant breast diseases. Semin Breast Dis 8:152–162 Mechine-Neuville A, Chenard MP, Gairard B et al, 2000, Large sections in routine breast pathology. A technique adapted to conservative surgery. Ann Pathol 20:275–209 Potter S, Govindarajulu S, Cawthorn SJ et al, 2007, Accuracy of sonographic localization and specimen ultrasound performed by surgeons in impalpable screen-detected breast lesions. Breast 16:425–428 Tot T, 2006, The limited prognostic value of measuring and grading small breast carcinomas: the whole sick lobe versus the details within it. Med Sci Monit 12:170–175 Tot T, 2003, The diffuse type of invasive lobular carcinoma of the breast: morphology and prognosis. Virchows Arch 444:718–724 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 173 EP 178 DI 10.1007/s12253-008-9061-9 PG 6 ER PT J AU Polgar, Cs Kahan, Zs Orosz, Zs Gabor, G Hadijev, J Cserni, G Kulka, J Jani, N Sulyok, Z Lazar, Gy Boross, G Diczhazi, Cs Szabo, Laszlo, Zs Pentek, Z Major, T Fodor, J AF Polgar, Csaba Kahan, Zsuzsanna Orosz, Zsolt Gabor, Gabriella Hadijev, Janaki Cserni, Gabor Kulka, Janina Jani, Nora Sulyok, Zoltan Lazar, Gyorgy Boross, Gabor Diczhazi, Csaba Szabo, Eva Laszlo, Zsolt Pentek, Zoltan Major, Tibor Fodor, Janos TI The Role of Radiotherapy in the Conservative Treatment of Ductal Carcinoma in Situ of the Breast SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Breast-conserving therapy; Ductal carcinoma in situ; Endocrine therapy; Prognostic factors; Radiotherapy ID Breast-conserving therapy; Ductal carcinoma in situ; Endocrine therapy; Prognostic factors; Radiotherapy AB Breast-conserving surgery (BCS) followed by radiotherapy (RT) has become the standard of care for the treatment of early-stage (St. I-II) invasive breast carcinoma. However, controversy exists regarding the value of RT in the conservative treatment of ductal carcinoma in situ (DCIS). In this article we review the role of RT in the management of DCIS. Retrospective and prospective trials and meta-analyses published between 1975 and 2007 in the MEDLINE database, and recent issues of relevant journals/handbooks relating to DCIS, BCS and RT were searchedfor. In retrospective series (10,194 patients) the 10-year rate of local recurrence (LR) with and without RT was reported in the range of 9–28% and 22–54%, respectively. In four large randomised controlled trials (NSABP-B-17, EORTC-10853, UKCCCR, SweDCIS; 4,568 patients) 50 Gy wholebreast RT significantly decreased the 5-year LR rate from 16–22% (annual LR rate: 2.6–5.0%) to 7–10% (annual LR rate: 1.3–1.9%). In a recent meta-analysis of randomised trials the addition of RT to BCS resulted in a 60% risk reduction of both invasive and in situ recurrences. In a multicentre retrospective study, an additional dose of 10 Gy to the tumour bed yielded a further 55% risk reduction compared to RT without boost. To date, no subgroups have been reliably identified that do not benefit from RT after BCS. In the NSABP-B-24 trial, the addition of tamoxifen (TAM) to RT reduced ipsilateral (11.1% vs. 7.7%) and contralateral (4.9% vs. 2.3%) breast events significantly. In contrast, in the UKCCCR study, TAM produced no significant reduction in all breast events. Based on available evidence obtained from retrospective and prospective trials, all patients with DCIS have potential benefit from RT after BCS. Further prospective studies are warranted to identify subgroups of low-risk patients with DCIS for whom RT can be safely omitted. Until long-term results of ongoing studies on outcomes of patients treated with BCS alone (with or without TAM or aromatase inhibitors) are available, RT should be routinely recommended after BCS for all patients except those with contraindication. C1 [Polgar, Csaba] National Institute of Oncology, Center of Radiotherapy, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of OncotherapySzeged, Hungary. [Orosz, Zsolt] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Gabor, Gabriella] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. [Hadijev, Janaki] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of PathologyKecskemet, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Jani, Nora] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Sulyok, Zoltan] National Institute of Oncology, Department of SurgeryBudapest, Hungary. [Lazar, Gyorgy] University of Szeged, Department of SurgerySzeged, Hungary. [Boross, Gabor] Bacs-Kiskun County Hospital, Department of SurgeryKecskemet, Hungary. [Diczhazi, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Szabo, Eva] National Institute of Oncology, Center of RadiologyBudapest, Hungary. [Laszlo, Zsolt] MaMMa Egeszsegugyi RtBudapest, Hungary. [Pentek, Zoltan] MaMMa Egeszsegugyi RtBudapest, Hungary. [Major, Tibor] National Institute of Oncology, Center of Radiotherapy, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Fodor, Janos] National Institute of Oncology, Center of Radiotherapy, Rath Gy. u. 7-9, 1122 Budapest, Hungary. RP Polgar, Cs (reprint author), National Institute of Oncology, Center of Radiotherapy, 1122 Budapest, Hungary. EM polgar@oncol.hu CR Fisher B, Anderson S, Bryant J et al, 2002, Twenty-year followup of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. New Engl J Med 347:1233–1241 Veronesi U, Cascinelli N, Mariani L et al, 2002, Twenty-year follow-up of a randomized study comparing breast-conserving surgery with radical mastectomy for early breast cancer. New Engl J Med 347:1227–1232 Adlard JW, Bundred NJ, 2006, Radiotherapy for ductal carcinoma in situ. Clin Oncol 18:179–184 Schwartz GF, Solin LJ, Olivotto IA et al, 2000, Consensus conference on the treatment of in situ ductal carcinoma of the breast, April 22–25, 1999. Cancer 88:946–954 Fietkau R, 2004, Ductal carcinoma in situ of the breast: Increasing importance of radiotherapy as a part of the therapy approach. Strahlenther Onkol 180:682–689 Leonard GD, Swain SM, 2004, Ductal carcinoma in situ, complexities and challanges. J Natl Cancer Inst 96:906–920 Mokbel K, Cutuli B, 2006, Heterogeneity of ductal carcinoma in situ and its effects on management. Lancet Oncol 7:756–765 Mokbel K, 2005, Contemporary treatment of ductal carcinoma in situ of the breast. Med Sci Monit 11:86–93 Sumner WE, Koniaris LG, Snell SE et al, 2007, Results of 23,810 cases of ductal carcinoma-in-situ. Ann Surg Oncol 14:1638–1643 Arnesson LG, Smeds S, Fagerberg G et al, 1989, Follow-up of two treatment modalities for ductal carcinoma in situ of the breast. Br J Surg 76:672–675 Baird RM, Worth A, Hislop G, 1990, Recurrence after lumpectomy for comedo-type intraductal carcinoma in situ of the female breast. Am J Surg 159:479–481 Blamey RW, Macmillan RD, Rampaul RS et al, 2002, Ductal carcinoma in situ: Experience at Nottingham City Hospital 1973 through 2000. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. Lippincott Williams & Wilkins, Philadelphia, USA, pp 322–328 Carpenter R, Boulter PS, Cooke T et al, 1981, Management of screen detected ductal carcinoma in situ of the female breast. Br J Surg 76:564–567 Lagios MD, 2002, The Lagios experience. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. Lippincott Williams & Wilkins, Philadelphia, USA, pp 303–307 MacAusland SG, Hepel JT, Chong FK et al, 2007, An attempt to independently verify the utility of the Van Nuys Prognostic Index for ductal carcinoma in situ. Cancer 110:2648–2653 Millis RR, Thynne GSJ, 1975, In situ intraductal carcinoma of the breast: a long-term follow-up study. Br J Surg 62:957–962 Ottesen GL, Graversen HP, Blichert-Toft M et al, 2000, Carcinoma in situ of the female breast. Ten-year follow-up results of a nationwide study. Breast Cancer Res Treat 62:197–210 Schwartz GF, 2002, Treatment of subclinical ductal carcinoma in situ of the breast by local excision and surveillance: An updated personal experience. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. Lippincott Williams & Wilkins, Philadelphia, USA, pp 308–321 Wong JS, Kaelin CM, Troyan SL et al, 2006, Prospective study of wide excision alone for ductal carcinoma in situ of the breast. J Clin Oncol 24:1031–1036 Amichetti M, Caffo O, Richetti A et al, 1999, Subclinical ductal carcinoma in situ of the breast: Treatment with conservative surgery and radiotherapy. Tumori 85:488–493 Ben-David MA, Sturtz DE, Griffith KA et al, 2007, Long-term results of conservative surgery and radiotherapy for ductal carcinoma in situ using lung density correction: the University of Michigan experience. Breast J 13:392–400 Fisher ER, Leeming R, Anderson S et al, 1991, Conservative management of intraductal carcinoma, DCIS, of the breast. J Surg Oncol 47:139–147 Gallagher WJ, Koerner FC, Wood WC, 1989, Treatment of intraductal carcinoma with limited surgery: long-term follow-up. J Clin Oncol 7:376–380 Jha MK, Avlonitis VS, Griffith CDM et al, 2001, Aggressive local treatment for screen-detected DCIS results in very low rates of recurrence. Eur J Surg Oncol 27:454–458 Mascarel I, Bonichon F, MacGrogan G et al, 2000, Application of the Van Nuys prognostic index in a retrospective series of 367 ductal carcinomas in situ of the breast examinated by serial macroscopic sectioning: Practical considerations. Breast Cancer Res Treat 61:151–159 Ray GR, Adelson J, Hayhurst E et al, 1993, Ductal carcinoma in situ of the breast: results of treatment by conservative surgery and definitive irradiation. Int J Radiat Oncol Biol Phys 28:105–111 Solin LJ, Fourquet A, Vicini FA et al, 2005, Long-term outcome after breast-conservation treatment with radiation for mammographically detected ductal carcinoma in situ of the breast. Cancer 103:1137–1146 Stotter AT, McNeese M, Oswald MJ et al, 1990, The role of limited surgery with irradiation in primary treatment of ductal in situ breast cancer. Int J Radiat Oncol Biol Phys 18:283–287 Forquet A, Sigal-Zafrani B, Clough KA, 2002, Breast-conserving surgery plus radiation therapy in ductal carcinoma in situ: The Institut Curie experience. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. Lippincott Williams & Wilkins, Philadelphia, USA, pp 367–372 Nakamura S, Woo C, Silberman H et al, 2002, Breast-conserving therapy for ductal carcinoma in situ: a 20-year experience with excision plus radiation therapy. Am J Surg 184:403–409 Jhingran A, Kim JS, Buchholz TA et al, 2002, Age as a predictor of outcome for women with DCIS treated with breast-conserving surgery and radiation: The University of Texas M. D. Anderson Cancer Center experience. Int J Radiat Oncol Biol Phys 54:804–809 Hiramatsu H, Bornstein BA, Recht A et al, 1995, Local recurrence after conservative surgery and radiation therapy for ductal carcinoma in situ. Possible importance of family history. Can J Sci Am 1:55–61 McCormick B, Rosen PP, Kinne D et al, 1991, Duct carcinoma in situ of the breast: An analysis of local control after conservation surgery and radiotherapy. Int J Radiat Oncol Biol Phys 21:289–292 Cataliotti L, Distante V, Orzalesi L et al, 2002, The Florence experience. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. Lippincott Williams & Wilkins, Philadelphia, USA, pp 348–353 Chan KC, Knox WF, Sinha G et al, 2001, Extent of excision margin with required in breast conserving surgery for ductal carcinoma in situ. Cancer 91:9–16 Cutuli B, Cohen-Solal-Le Nir C, De Lafontan B et al, 2001, Ductal carcinoma in situ of the breast results of conservative and radical treatments in 716 patients. Eur J Cancer 37:2365–2372 Dixon JM, 2002, Ductal carcinoma in situ: the Edinburgh experience. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. Lippincott Williams & Wilkins, Philadelphia, USA, pp 343–347 Meijnen P, Oldenburg HSA, Peterse JL et al, 2008, Clinical outcome after selective treatment of patients diagnosed with ductal carcinoma in situ of the breast. Ann Surg Oncol 15:235–243 Omlin A, Amichetti M, Azria D et al, 2006, Boost radiotherapy in young women with ductal carcinoma in situ: a multicentre, retrospective study of the Rare Cancer Network. Lancet Oncol 7:652–656 Park CC, Schnitt SJ, Recht A, 2002, Joint center for radiation therapy experience. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. Lippincott Williams & Wilkins, Philadelphia, USA, pp 373–380 Ringberg A, Idvall I, Ferno M et al, 2000, Ipsilateral local recurrence in relation to therapy and morphological characteristics in patients with ductal carcinoma in situ of the breast. Eur J Surg Oncol 26:444–451 Silverstein MJ, 2002, The university of Southern California/Van Nuys prognostic index. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. Lippincott Williams & Wilkins, Philadelphia, USA, pp 459–473 Silverstein MJ, 2002, The Van Nuys/University of Southern California experience by treatment. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. Lippincott Williams & Wilkins, Philadelphia, USA, pp 337–342 Silverstein MJ, Lagios MD, Groshen S et al, 1999, The influence of margin width on local control of ductal carcinoma in situ of the breast. N Engl J Med 340:1455–1461 Van der Velden APS, Van Vugt R, Van Dijck JAAM et al, 2007, Local recurrences after different treatment strategies for ductal carcinoma in situ of the breast: A population-based study in East Netherlands. Int J Radiat Oncol Biol Phys 69:703–710 Van Zee KJ, Liberman L, Samli B et al, 1999, Long term followup of women with ductal carcinoma in situ treated with breastconserving surgery. Cancer 86:1757–1767 Vargas C, Kestin L, Go N et al, 2005, Factors associated with local recurrence and cause-specific survival in patients with ductal carcinoma in situ of the breast treated with breast-conserving therapy or mastectomy. Int J Radiat Oncol Biol Phys 63:1514–1521 Silverstein MJ, Poller DN, Waisman JR et al, 1995, Prognostic classification of breast ductal carcinoma-in-situ. Lancet 345: 1154–1157 Silverstein MJ, Lagios MD, Craig PH et al, 1996, A prognostic index for ductal carcinoma in situ of the breast. Cancer 77:2267– 2274 Fisher B, Land S, Mamounas E et al, 2001, Prevention of invasive breast cancer in women with ductal carcinoma in situ: An update of the National Surgical Adjuvant Breast and Bowel Project experience. Semin Oncol 28:400–418 Schnitt SJ, Harris JR, Smith BL, 1996, Developing a prognostic index for ductal carcinoma in situ of the breast. Are we there yet? Cancer 77:2189–2192 Bijker N, Meijnen P, Peterse JL et al, 2006, Breast-conserving treatment with or without radiotherapy in ductal carcinoma in situ: Ten-year results of European Organisation for Research and Treatment of Cancer randomized phase III trial 10853-A study by the EORTC Breast Cancer Cooperative Group and EORTC Radiotherapy Group. J Clin Oncol 24:3381–3387 Fisher ER, Dignam J, Tan-Chiu E et al, 1999, Pathologic findings from the National Surgical Adjuvant Breast Project, NSABP, eight-year update of protocol B-17: Intraductal carcinoma. Cancer 86:429–438 Ringberg A, Nordgren H, Thorstensson S et al, 2007, Histopathological risk factors for ipsilateral breast events after breast conserving treatment for ductal carcinoma in situ of the breast – Results from the Swedish randomised trial. Eur J Cancer 43:291–298 Boland GP, Chan KC, Knox WF et al, 2003, Value of the Van Nuys Prognostic Index in prediction of recurrence of ductal carcinoma in situ after breast-conserving surgery. Br J Surg 90:426–432 Boyages J, Delaney G, Taylor R, 1998, Predictors of local recurrence after treatment of ductal carcinoma in situ: a metaanalysis. Cancer 85:616–628 Bijker N, Peterse JL, Duchateau L et al, 2001, Risk factors for recurrence and metastasis after breast-conserving therapy for ductal carcinoma-in-situ: analysis of European organization for research and treatment of cancer trial 10853. J Clin Oncol 19:2263–2271 Emdin SO, Granstrand B, Ringberg A et al, 2006, SweDCIS: Radiotherapy after sector resection for ductal carcinoma in situ of the breast. Results of a randomised trial in a population offered mammography screening. Acta Oncol 45:536–543 Fisher B, Costantino J, Redmond C et al, 1993, Lumpectomy compared with lumpectomy and radiation therapy for the treatment of intraductal breast cancer. New Engl J Med 328:1581–1586 Fisher B, Dignam J, Wolmark N et al, 1999, Tamoxifen in treatment of intraductal breast cancer: National Surgical Adjuvant Breast and Bowel Project B-24 randomised controlled trial. Lancet 353:1993–2000 Houghton J, George WD, Cuzick J et al, 2003, Radiotherapy and tamoxifen in women with completely excised ductal carcinoma in situ of the breast in the UK, Australia, and New Zealand: randomised controlled trial. Lancet 362:95–102 Julien JP, Bijker N, Fentiman IS et al, 2000, Radiotherapy in breast-conserving treatment for ductal carcinoma in situ: First results of the EORTC randomised phase III trial 10853. Lancet 355:528–533 Viani GA, Stefano EJ, Afonso SL et al, 2007, Breast-consreving surgery with or without radiotherapy in women with ductal carcinoma in situ: a meta-analysis of randomized trials. Rad Oncol 2:28, DOI 10.118/178-717X-2-28 Patani N, Cutuli B, Mokbel K, 2008, Current management of DCIS: a review. Breast Cancer Res Treat, in press, DOI 10.1007/ s10549-007-9760-z Orosz Z, Jani N, Kahan Z et al, 2007, Multicentric clinicopathologic analysis of ductal carcnoma in situ. [Abstract] Magyar Onkologia 51:376 Polgar C, Orosz Z, Kahan Z et al, 2007, Combined surgery and radiotherapy for the treatment of breast ductal carcinoma in situ: Scientific evidences and preliminary results of the Hungarian multicentric prospective randomized DCIS trial. [Abstract] Magyar Onkologia 51:384 Bartelink H, Horiot JC, Poortmans PM et al, 2007, Impact of a higher radation dose on local control and survival in breastconserving therapy of early breast cancer: 10-year results of the randomized boost versus no boost EORTC 22881–10882 trial. J Clin Oncol 25:3259–3265 Polgar C, Fodor J, Major T et al, 2001, The role of boost irradiation in the conservative treatment of stage I-II breast cancer. Pathol Oncol Res 7:241–250 Holland R, Faverly DRG, 2002, The local distribution of ductal carcinoma in situ of the breast: whole-organ studies. In: Silverstein MJ, ed, Ductal carcinoma in situ of the breast. Lippincott Williams & Wilkins, Philadelphia, USA, pp 240–248 Polgar C, Strnad V, Major T, 2005, Brachytherapy for partial breast irradiation: The European experience. Semin Radiat Oncol 15:116–122 Vicini FA, Arthur DW, 2005, Breast brachytherapy: North American experience. Semin Radiat Oncol 15:108–115 Polgar C, Fodor J, Major T et al, 2007, Breast-conserving treatment with partial or whole breast irradiation for low-risk invasive breast carcinoma – 5-year results of a randomized trial. Int J Radiat Oncol Biol Phys 69:694–702 Benitez PR, Streeter O, Vicini F et al, 2006, Preliminary results and evaluation of MammoSite balloon brachytherapy for partial breast irradiation for pure ductal carcinoma in situ: a phase II clinical study. Am J Surg 192:427–433 Ragaz J, Coldman A, 1998, Survival impact of adjuvant tamoxifen on competing causes of mortality in breast cancer survivors, with analysis of mortality from contralateral breast cancer, cardiovascular events, endometrial cancer, and thromboembolic epizodes. J Clin Oncol 16:2018–2024 Zhang Z, Yamashita H, Toyama T et al, 2002, Semi-quantitative immunohistochemical analysis of aromatase expression in ductal carcinoma in situ of the breast. Breast Cancer Res Treat 74:47–53 Cuzick J, 2003, Treatment for DCIS – results from clinical trials. Surg Oncol 12:213–219 Ringberg A, Anagnostaki L, Anderson H et al, 2001, Cell biological factors in ductal carcinoma in situ, DCIS, of the breast – relationship to ipsilateral local recurrence and histopathological characteristics. Eur J Cancer 37:1514–1522 Zagouri F, Sergentanis TN, Zografos GC, 2007, Precursors and preinvasive lesions of the breast: the role of molecular prognostic markers in the diagnostic and therapeutic dilemma. World J Surg Oncol 5:57–67 Nofech-Mozes S, Spayne J, Rakovitch E et al, 2005, Prognostic and predictive molecular markers in DCIS: a review. Adv Anat Pathol 12:256–264 Zaugg K, Bodis S, 2000, Is there a role for molecular prognostic factors in the clinical management of ductal carcinoma in situ, DCIS, of the breast? Radiother Oncol 55:85–99 Cornfield DB, Palazzo JP, Schwartz GF et al, 2004, The prognostic significance of multiple morphologic features and biologic markers in ductal carcinoma in situ of the breast: A study of a large cohort of patients treated with surgery alone. Cancer 100:2317–2327 Tang P, Hajdu SI, Lyman GH, 2007, Ductal carcinoma in situ: a review of recent advances. Curr Opin Obstet Gynecol 19: 63–67 Tang P, Wang X, Schiffhauer L et al, 2006, Relationship between nuclear grade of ductal carcinoma in situ and cell origin markers. Ann Clin Lab Sci 36:16–22 Consensus Conference Committee, 1997, Consensus Conference on the Classification of Ductal Carcinoma in Situ, April 25–28, 1997. Cancer 80:1798–1802 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 179 EP 192 DI 10.1007/s12253-008-9044-x PG 14 ER PT J AU Kovacs, Hadjiev, J Lakosi, F Glavak, Cs Antal, G Bogner, P Horvath, Repa, I AF Kovacs, Arpad Hadjiev, Janaki Lakosi, Ferenc Glavak, Csaba Antal, Gergely Bogner, Peter Horvath, Akos Repa, Imre TI Comparison of Photon with Electron Boost in Treatment of Early Stage Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Boost; Breast cancer; Radiotherapy; Photon; Electron ID Boost; Breast cancer; Radiotherapy; Photon; Electron AB In the treatment of early stage breast cancer breast conserving surgery (BCS) followed by whole breast irradiation (WBI) is a standard method. The impact of the tumor bed boost following WBI is well-defined, but there are various delivery methods. In this study the electron and the photon boost techniques were compared. For 78 early stage breast cancer patients both CT based 3D conformal photon boost and electron boost plans were created. For dosimetric comparison coverage index (CI), external volume index (EI) and conformality index (COIN) were studied. Lung volume receiving a dose of 2 Gy was also reviewed. Seventy-eight patients with 156 plans were compared. The mean tumor bed volume was measured as 61.39 cm3 the mean tumor bed-skin distance was 3.13 cm. In the case of CI and COIN significant differences were found in favor of the photon boost. In the comparison of EI no significant difference was detected between the two techniques. The mean lung volume receiving 2 Gy were 42.3 and 168.35 cm3, for photons and electrons respectively. In the adjuvant treatment of early stage breast cancer WBI followed by conformal photon boost showed to be superior to electron boost in focus of the COIN and CI. C1 [Kovacs, Arpad] Kaposi Mor Teaching HospitalKaposvar, Hungary. [Hadjiev, Janaki] Kaposi Mor Teaching HospitalKaposvar, Hungary. [Lakosi, Ferenc] Kaposi Mor Teaching HospitalKaposvar, Hungary. [Glavak, Csaba] Kaposi Mor Teaching HospitalKaposvar, Hungary. [Antal, Gergely] Kaposi Mor Teaching HospitalKaposvar, Hungary. [Bogner, Peter] Kaposi Mor Teaching HospitalKaposvar, Hungary. [Horvath, Akos] University of Debrecen, Medical and Health Science Center, Institute of Nuclear Medicine, Nagyerdei Krt. 98Debrecen, Hungary. [Repa, Imre] Kaposi Mor Teaching HospitalKaposvar, Hungary. RP Horvath, (reprint author), University of Debrecen, Medical and Health Science Center, Institute of Nuclear Medicine, Debrecen, Hungary. EM horvakos@freemail.hu CR Early Breast Cancer Triallists’ Collaborative Group, 1995, Effects of radiotherapy and surgery in early breast cancer: an overview of the randomized trials. N Engl J Med 333:1444–1445 [erratum, N Engl J Med 334:1003, 1996]. Fisher B, Anderson S, Bryant J et al, 2002, Twenty-year followup of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 347:1233–1241 Veronesi U, Cascinelli N, Mariani L et al, 2002, Twenty-year follow-up of a randomized study comparing breast-conserving surgery with radical mastectomy for early breast cancer. N Engl J Med 347:1227–1232 Bartelink H, Horiot JC, Poortmans P et al, 2001, Recurrence rates after treatment of breast cancer with standard radiotherapy with or without additional radiation. N Engl J Med 345:1378–1387 Romestaing P, Lehingue Y, Carrie C et al, 1997, Role of a 10 Gy boost in the conservative treatment of early breast cancer: results of a randomized clinical trial in Lyon, France. J Clin Oncol 15: 963–968 Graham P, Fourquetz A, 2005, Placing the boost in breastconservation radiotherapy: a review of the role, indications and techniques for breast-boost radiotherapy. Clin Oncol 18:210–219 Bedwinek J, 1993, Breast conserving surgery and irradiation: the importance of demarcating the excision cavity with surgical clips. Int J Radiat Oncol Biol Phys 26:675–679 Denham JW, Sillar RW, Clarke D, 1991, Boost dosage to the excision site following conservative surgery for breast cancer: it’s easy to miss! Clin Oncol 3:257–261 Denham JW, Carter ML, 1988, Conservative treatment of breast cancer—where should the booster dose go [letter]? Int J Radiat Oncol Biol Phys 14:399–400 Harrington KJ, Harrison M, Bayle P et al, 1996, Surgical clips in planning the electron boost in breast cancer: a qualitative and quantitative evaluation. Int J Radiat Oncol Biol Phys 34:579– 584 Ringash J, Whelan T, Elliott E et al, 2004, Accuracy of ultrasound in localization of breast boost field. Radiother Oncol 72:61–66 Sedlmayer F, Rahim HBK, Kogelnik HK et al, 1996, Quality assurance in breast cancer brachytherapy: geographic miss in the interstitial boost treatment of the tumor bed. Int J Radiat Oncol Biol Phys 34:1133–1139 American Joint Committee on Cancer, 2002, AJCC cancer staging handbook, sixth edition. Springer, Heidelberg, pp 263–267 International Commission on Radiation Units and Measurement, ICRU,, 1993, ICRU Report No. 50. Prescribing, recording and reporting photon beam therapy. ICRU, Washington, DC Major T, Polgar CS, Fodor J, Somogyi A, Nemeth GY, 2002, Conformality and homogeneity of dose distributions in interstitial implants at idealized target volumes: a comparison between the Paris and dose-point optimized systems. Radiother Oncol 62:103–111 Polgar CS, Fodor J, Major T, Orosz ZS, Nemeth GY, 2001, The role of boost irradiation in the conservative treatment of stage I–II breast cancer. Pathol Oncol Res 7:241–250 Van Limbergen E, 2001, What are the optimal boost methods in relation to boost target depth in the breast? Proceedings of the Consensus Meeting on Breast Cancer: to boost or not to boost and how to do it. GEC-ESTRO, pp. 105–114. Polgar CS, Sulyok Z, Major T et al, 2000, Reexcision and perioperative brachytherapy in the treatment of local relapse after breast conservation: a possible alternative to salvage mastectomy. Hungarian Surgery 53:120–123, in Hungarian) Machtay M, Lanciano R, Hoffman J, Hanks GE, 1994, Inaccuracies in using the lumpectomy scar for planning electron boosts in primary breast carcinoma. Int J Radiat Oncol Biol Phys 30:43–48 Baltas D, Kolotas C, Geramani K et al, 1998, A conformal index, COIN, to evaluate implant quality and dose specification in brachytherapy. Int J Radiat Oncol Biol Phys 40:515–524 Knoos T, Kristensen I, Nilsson P, 1998, Volumetric and dosimetric evaluation of radiation treatment plans: radiation conformity index. Int J Radiat Oncol Biol Phys 42:1169–1176 RegineWF, Ayyangar KM, Komarnicky LT, Bhandare N, Mansfield CM, 1991, Computer-CT planning of the electron boost in definitive breast irradiation. Int J Radiat Oncol Biol Phys 20: 121–125 Smitt MC, Birdwell RL, Goffinet DR, 2001, Breast electron boost planning: comparison of CT and US. Radiology 219:203–206 Hunter MA, McFall TA, Hehr KA, 1996, Breast conserving surgery for primary breast cancer: necessity for surgical clips to define the tumor bed for radiation planning. Radiology 200:281–282 Kovner F, Agay R, Merimsky O, Stadler J, Klausner J, Inbar M, 1999, Clips and scar as the guidelines for breast radiation boost after lumpectomy. Eur J Surg Oncol 25:483–486 Poortmans P, Bartelink H, Horiot J-C, Struikmans H, Van den Bogaert W, Fourquetf A, Jagerg J, Hoogenraadh W, Rodrigusa P, Rodenhuisi CW, Collette L, Pierartj M; On behalf of the EORTC Radiotherapy and Breast Cancer Groups, 2004, The influence of the boost technique on local control in breast conserving treatment in the EORTC ‘boost versus no boost’ randomised trial. Radiother Oncol 72:25–33 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 193 EP 197 DI 10.1007/s12253-008-9015-2 PG 5 ER PT J AU Rieker, JR Hoegel, J Kern, AM Steger, Ch Aulmann, S Mechtersheimer, G Schirmacher, P Blaeker, H AF Rieker, J Ralf Hoegel, Josef Kern, A Michael Steger, Christina Aulmann, Sebastian Mechtersheimer, Gunhild Schirmacher, Peter Blaeker, Hendrik TI A Mathematical Approach Predicting the Number of Events in Different Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Incidence rates; Mutations; Poisson distribution; Probabilities; Tumorigenesis ID Incidence rates; Mutations; Poisson distribution; Probabilities; Tumorigenesis AB Supported by different investigations, multi-step models for tumorigenesis have been proposed for epithelial tumors. The age specific incidence of some cancers shows an exponential rise with increasing patient age. Yet, the onset and the slope of incidence curves varies between tumor types. One simple explanation for this disparity is that the number of mutations required for transformation differs in various tissues. We used a homogeneous Poisson process to estimate the number of events (N) and the intensity or event rate (1) that might be needed for cancer development in various tissues (colon, prostate, oralpharynx, larynx). Estimations were performed, including 95% confidence intervals, for the male and female population. The expected number of events needed was higher in adenocarcinomas (colorectal carcinoma: N≈10 for females and N≈11.0 for males; prostatic cancer: N≈23) than in squamous cell carcinomas (oropharynx: N≈5–6 for females and N≈6 for males; larynx: N≈7 for females and N≈ 8 males). Still, alternative models fixing N to values within the 95% confidence intervals determined, showed good coincidence with epidemiological data. Although the herein applied mathematical model neglects several biologic conditions, especially a presumed acceleration of mutation rates after tumor initiation it offers a plausible theory for the given epidemiologic data and matches with molecular biologic findings in the investigated cancers. C1 [Rieker, J Ralf] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Hoegel, Josef] University of Ulm, Department of Human GeneticsUlm, Germany. [Kern, A Michael] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Steger, Christina] Medical University of Innsbruck, Department of PathologyInnsbruck, Austria. [Aulmann, Sebastian] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Mechtersheimer, Gunhild] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Schirmacher, Peter] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. [Blaeker, Hendrik] University of Heidelberg, Department of Pathology, INF 220/221, 69120 Heidelberg, Germany. RP Blaeker, H (reprint author), University of Heidelberg, Department of Pathology, 69120 Heidelberg, Germany. EM hendrik.blaeker@med.uni-heidelberg.de CR Hasty P, Campisi J, Hoeijmakers J et al, 2003, Aging and genome maintenance: lessons from the mouse? Science 299:1355–1359 Tomlinson I, Sasieni P, Bodmer W, 2002, How many mutations in a cancer? Am J Pathol 160:755–758 Fearon ER, Vogelstein B, 1990, A genetic model for colorectal tumorigenesis. Cell 61:759–767 Fischer JC, Hollomon JH, 1951, A hypothesis for the origin of cancer foci. Cancer 4:916–918 Nordling CO, 1953, A new theory on the cancer-inducing mechanism. Br J Cancer 7:68–72 Armitage P, Doll R, 1954, The age distribution of cancer and a multi-stage theory of carcinogenesis. Br J Cancer 8:1–12 Armitage P, Doll R, 1957, A two-stage theory of carcinogenesis in relation to the age distribution of human cancer. Br J Cancer 11:161–169 Knudson AG, 1971, Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci USA 68:820–823 Knudson AG, 1973, Mutation and human cancer. Adv Cancer Res 17:317–352 Hazelton WD, Clements MS, Moolgavkar SH, 2005, Multistage carcinogenesis and lung cancer mortality in three cohorts. Cancer Epidemiol Biomarkers Prev 14:1171–1181 Breiman L, ed,, 1968, Probability. Wesley, Reading Ries LAG, Eisner MP, Kosary CL et al, eds,, 2005, SEER Cancer Statistics Review, 1975–2002. National Cancer Institute, Bethesda. Accessed at http://seer.cancer.gov/csr/1975_2002/, based on November 2004 SEER data submission, posted to the SEER web site 2005. Cited 15 Jan 2007 Davies H, Bignell GR, Cox C et al, 2002, Mutations of the BRAF gene in human cancer. Nature 417:949–954 Yuen ST, Davies H, Chan TL et al, 2002, Similarity of the phenotypic patterns associated with BRAF and KRAS mutations in colorectal neoplasia. Cancer Res 62:6451–6455 Samuels Y, Velculescu VE, 2004, Oncogenic mutations of PIK3CA in human cancers. Cell Cycle 3:1221–1224 Wang Z, Shen D, Parsons DW, Bardelli A et al, 2004, Mutational analysis of the tyrosine phosphatome in colorectal cancers. Science 304:1164–1166 Loeve F, Boer R, Zauber AG et al, 2004, National Polyp Study data: evidence for regression of adenomas. Int J Cancer 111:633– 639 Nwosu V, Carpten J, Trent JM et al, 2001, Heterogeneity of genetic alterations in prostate cancer: evidence of the complex nature of the disease. Hum Mol Genet 10:2313–2318 Karayi MK, Markham AF, 2004, Molecular biology of prostate cancer. Prostate Cancer Prostatic Dis 7:6–20 KimMM, Califano JA, 2004, Molecular pathology of head-and-neck cancer. Int J Cancer 112:545–553 Pai SI, Wu GS, Ozoren N et al, 1998, Rare loss-of-function mutation of a death receptor gene in head and neck cancer. Cancer Res 58:3513–3518 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 199 EP 204 DI 10.1007/s12253-008-9050-z PG 6 ER PT J AU Belovejdov, V Staribratova, D Dikov, D AF Belovejdov, Vesselin Staribratova, Diana Dikov, Dorian TI Microscopic Peliosis of Pancreatic Islets Associated with Thrombotic Thrombocytopenic Purpura SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Disseminated intravascular coagulation; pancreas; Peliosis; Pancreatic islets; Thrombocytic thrombocytopenic purpura ID Disseminated intravascular coagulation; pancreas; Peliosis; Pancreatic islets; Thrombocytic thrombocytopenic purpura AB Peliosis is a rarely seen histological finding with unexplained fully etiology and pathogenesis. It is presented as cyst-like blood filled cavities. The presence of peliosis in the endocrine part of the pancreas is extremely rarely reported microscopic phenomenon. The authors provide histological, histochemical, immunohistochemical and ultrastructural investigation of microscopic peliosis in the pancreas from an autopsy case with thrombotic trombocytopenic purpura. The findings give ideas for a wide range of pathophysiological and morphogenetic comments of such an unusual morphologic presentation. C1 [Belovejdov, Vesselin] Medical University of Plovdiv, Department of General and Clinical PathologyPlovdiv, Bulgaria. [Staribratova, Diana] Medical University of Plovdiv, Department of General and Clinical PathologyPlovdiv, Bulgaria. [Dikov, Dorian] Centre Hospitalier de Lagny-Marne-La-Vallee, Service d’Anatomie et de Cytologie PathologiquesParis, France. RP Staribratova, D (reprint author), Medical University of Plovdiv, Department of General and Clinical Pathology, Plovdiv, Bulgaria. EM dianastar@abv.bg CR Corpa MVN, Bacchi MM, Bacchi CE et al, 2004, Peliosis hepatis associated with lymphoplasmacytic lymphoma. Arch Pathol Lab Med 128:1283–1285 Kovacs K, Horvath E, Asa S et al, 1986, Microscopic peliosis of pancreatic islets in a woman with MEN-1 syndrome. Arch Pathol Lab Med 110:607–610 Berzigotti A, Magalotti D, Zappoli P et al, 2006, Peliosis hepatic as an early histological finding in idiopathic portal hypertension: a case report. World J Gastroenterol 12(22):3612– 3615 Tsokos M, Erbersdobler A, 2005, Pathology of peliosis. Forensic Sci Int 149, 1):25–33 Zafrani ES, Cazier A, Baudelot AM et al, 1984, Ultrastructural lesions of the liver in human peliosis. A report of 12 cases. Am J Pathol 114(3):349–359 Warfel KA, Ellis GH, 1982, Peliosis of the spleen. Report of a case and review of the literature. Arch Pathol Lab Med 106(2):99– 100 Czapar CA, Weldon-Linne CM, Moore DM et al, 1986, Peliosis hepatic in acquired immunodeficiency syndrome. Arch Pathol Lab Med 110(7):611–613 Gulubova M, 2002, Immunohistochemical localization of collagen Type III and Type IV, laminin, tenascin and α-smooth muscle actin, αSMA, in the human liver in peliosis. Pathol Res Pract 198:803–812 Korner M, Gebbers J-O, 2002, Peliosis of the spleen and haemolytic anemia. Histopathology 41:179–180 Koehler JE, Sanchez MA, Garrido CS et al, 1997, Molecular epidemiology of Bartonellainfections in patients with bacillary angiomatosis–peliosis. The N Eng J Med 337, 26):1876–83 Coire CI, Horvath E, Kovacs K et al, 1997, Cushing’s syndrome from an ectopic pituitary adenoma with peliosis: a histological, immunohistochemical and ultrastructural study and review of the literature. Endocr Pathol 8(1):65–74 Lie JT, 1985, Pulmonary peliosis. Arch Pathol Lab Med 109, 9):878–9 Chetty R, Kennedy M, Ezzat S et al, 2004, Pancreatic endocrine pathology in von Hippel-Lindau disease: an expanding spectrum of lesions. Endocrine Pathology 15(2):141–148 Pernicone PJ, Lie JT, 1987, Isolated peliosis of pancreatic islet cell adenoma. Arch Pathol Lab Med 111:690–691 Ricketts LM, Dlugous M, Luther KB et al, 2007, O-fucosylation is require for ADAMTS 13 secretion. J Biol Chem 282, 23):17014–23 Girma JP, 2006, Biology of von Willebrand factor. Nephrol Ther 2:s143–s148 Fina L, Molgaard HV, Robertson D et al, 1990, Expression of the CD 34 gene in vascular endothelial cells. Blood 75, 12):2417–26 Delas N, Faurel JP, Wechsled B et al, 1982, Association of peliosis and necrotizing vasculitis. Nouv Presse Med 25;11, 37):2787 Galbusera M, Noris M, Remuzzi G, 2006, Thrombotic thrombocytopenic purpura—then and now. Semin Thromb Hemost 32, 2):81–89 Voinchet O, Degott C, Scoazec JY et al, 1988, Peliosis hepatis, nodular regenerative hyperplasia of the liver, and light-chain deposition in a patient with Waldenstrom’s macroglobulinemia. Gastroenterology 95:482–486 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 205 EP 208 DI 10.1007/s12253-008-9048-6 PG 4 ER PT J AU Maka, E Lukats, O Toth, J Fekete, S AF Maka, Erika Lukats, Olga Toth, Jeannette Fekete, Sandor TI Orbital Tumour as Initial Manifestation of Acute Myeloid Leukemia: Granulocytic Sarcoma: Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Orbital granulocytic sarcoma; Acute myeloid leukemia ID Orbital granulocytic sarcoma; Acute myeloid leukemia AB We report orbital involvement as an initial manifestation of acute myeloid leukemia in a 57-year-old woman. The patient presented with painful proptosis and limited ocular motility. Orbital computed tomography revealed bilateral homogeneous masses. Orbital biopsy was performed on the right side; and histopathology disclosed a myelocytic tumour. Despite treatment using irradiation and chemotherapy, the patient died eleven months after presentation. There appear to be only a few previous reports of acute myeloid leukemia cases presenting with orbital involvement, and most cases occurred in children. This very rare condition has a poor survival prognosis, even with radiation treatment and chemotherapy. C1 [Maka, Erika] Semmelweis University, Department of Ophthalmology, Tomo utca 25–29, 1083 Budapest, Hungary. [Lukats, Olga] Semmelweis University, Department of Ophthalmology, Tomo utca 25–29, 1083 Budapest, Hungary. [Toth, Jeannette] Semmelweis University, Department of Ophthalmology, Tomo utca 25–29, 1083 Budapest, Hungary. [Fekete, Sandor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. RP Maka, E (reprint author), Semmelweis University, Department of Ophthalmology, 1083 Budapest, Hungary. EM maker@szem1.sote.hu CR Zimmerman LE, Font RL, 1975, Ophthalmologic manifestations of granulocytic sarcoma, myeloid sarcoma or chloroma). Am J Ophthalmol 80:975–989 Bhattacharjee K, Bhattacharjee H, Das D, Babu K, Mahesh L, Krishnakumar S, Biswas J, 2003, Chloroma of the orbit in a nonleukemic adult: a case report. Orbit 22:293–297 Kincaid MC, Green WR, 1983, Ocular and Orbital Involvement in Leukemia. Surv Ophthalmol 27:211–232 Padillo D, Mencia E, Gutierrez E, Martinez MA, 1999, Orbital granulocytic sarcoma in a myelodysplastic syndrome. Orbit 18: 287–290 Stockl FA, DolmetschAM, SaornilMA, Font RL, BurnierMN, 1997, Orbital granulocytic sarcoma. Br J Ophthalmol 81:1084–1088 Watkins LM, Remulla HD, Rubin PAD, 1997, Orbital granulocytic sarcoma in an elderly patient. Am J Ophthalmol 123:854–856 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 209 EP 211 DI 10.1007/s12253-008-9028-x PG 3 ER PT J AU Leoncini, G Maio, V Puccioni, M Franchi, A De Giorgi, V Ucci, F Santucci, M Massi, D AF Leoncini, Giuseppe Maio, Vincenza Puccioni, Marco Franchi, Alessandro De Giorgi, Vincenzo Ucci, Francesca Santucci, Marco Massi, Daniela TI Orbital Solitary Fibrous Tumor: A Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Solitary fibrous tumor; Orbit ID Solitary fibrous tumor; Orbit AB Solitary fibrous tumor (SFT) is a rare spindle cell neoplasm typically arising in the pleura and involving the orbit as its most common extra-pleural location. We herein describe a well documented case of orbital SFT arising in a 62-year-old woman presenting with progressive swelling of the right upper eyelid and proptosis. The tumor had a benign clinical course, with radical surgical excision followed by regression of the clinical symptoms. We review the clinical, histopathological, and immunohistochemical features of the orbital SFT described so far, with particular emphasis on differential diagnosis with other spindle cell orbital neoplasms. C1 [Leoncini, Giuseppe] AOU Careggi, Department of Human Pathology and Oncology, Viale GB Morgagni 85, 50134 Florence, Italy. [Maio, Vincenza] AOU Careggi, Department of Human Pathology and Oncology, Viale GB Morgagni 85, 50134 Florence, Italy. [Puccioni, Marco] University of Florence, Department of OphthalmologyFlorence, Italy. [Franchi, Alessandro] AOU Careggi, Department of Human Pathology and Oncology, Viale GB Morgagni 85, 50134 Florence, Italy. [De Giorgi, Vincenzo] University of Florence, Department of Dermatological SciencesFlorence, Italy. [Ucci, Francesca] University of Florence, Department of OphthalmologyFlorence, Italy. [Santucci, Marco] AOU Careggi, Department of Human Pathology and Oncology, Viale GB Morgagni 85, 50134 Florence, Italy. [Massi, Daniela] AOU Careggi, Department of Human Pathology and Oncology, Viale GB Morgagni 85, 50134 Florence, Italy. RP Massi, D (reprint author), AOU Careggi, Department of Human Pathology and Oncology, 50134 Florence, Italy. EM Daniela.massi@unifi.it CR De Saint Aubain Somerhausen N, Rubin BP, Fletcher CDM, 1999, Mixoid solitary fibrous tumor: a study of seven cases with emphasis on differential diagnosis. Mod Pathol 12:463–471 Klemper C, Rabin CB, 1931, Primary neoplasm of the pleura; a report of five cases. Arch Pathol 11:385–412 El-Naggar AK, Ro JY, Ayala AG et al, 1989, Localized fibrous tumor of the serosal cavities. Am J Clin Pathol 13:199–209 Kottke-Marchant K, Hart WR, Broughan T, 1989, Localized fibrous tumor, localized fibrous mesothelioma, of the liver. Cancer 64:1096–1102 Witkin GB, Rosai J, 1989, Solitary fibrous tumor of the mediastinum. Am J Surg Pathol 13:547–557 Witkin GB, Rosai J, 1991, Solitary fibrous tumor of the upper respiratory tract. Am J Surg Pathol 15:842–848 Fukunaga M, Nikaido T, 1997, Solitary fibrous tumor of the renal peri-pelvis. Histopathology 30:451–456 Prevot S, Penna C, Imbert JC et al, 1996, Solitary fibrous tumor of the adrenal gland. Mod Pathol 9:1170–1174 Fisher C, Bisceglia M, 1994, Solitary fibrous tumor of the spermatic cord. Br J Urol 74:798–789 Hasegawa T, Hirose T, Seki K et al, 1996, Solitary fibrous tumor of the soft tissue. Am J Clin Pathol 106:325–331 Cowper SE, Kilpatrick T, Proper S et al, 1999, Solitary fibrous tumor of the skin. Am J Dermatopathol 21(3):213–219 Zuckerberg ML, Rosemberg AE, Randolph G et al, 1991, Solitary fibrous tumor of the nasal cavity and paranasal sinuses. Am J Surg Pathol 15:126–130 England DM, Hochholzer L,McCarthyMJ, 1989, Localized benign and malignant fibrous tumors of the pleura: a clinicopathologic review of 223 cases. Am J Surg Pathol 13:640–658 Chan JKC, 1997, Solitary fibrous tumor—everywhere and a diagnosis in vogue. Histopathology 31:568–576 Frederick A Font J, Font RL, 1986, Orbit. In: Spencer WH, ed, Ophthalmic pathology: an atlas and textbook, 3rd edn. Saunders, Philadelphia, pp 2459–2860 Festa S, Lee HJ, Langer P et al, 1999, Solitary fibrous tumor of the orbit: CT and pathologic correlation. Neuroradiology 41:52–54 Scott IU, Tanenbaum M, Rubin D et al, 1996, Solitary fibrous tumor of the lacrimal gland fossa. Ophthalmology 103:1613– 1618 Woo KI, Suh YL, Kim YD, 1999, Solitary fibrous tumor of the lacrimal sac. Ophthal Plast Reconstr Surg 15(6):450–453 Polito E, Tosi M, Toti P et al, 2002, Orbital solitary fibrous tumor with aggressive behaviourThree cases and review of the literature. Graefes Arch Clin Exp Ophthalmol 240(7):570–574 Hayashi S, Kurihara H, Hirato J et al, 2001, Solitary fibrous tumor of the orbit with an extraorbital extension: case report. Neurosurgery 49(5):1241–1245 Carrera M, Prat J, Quintana M, 2001, Malignant solitary fibrous tumor of the orbit: report of a case with 8 years follow-up. Eye 15:102–104 Vallat-Decouvelaere AV, Dry SM, Fletcher CDM, 1998, Atypical and malignant solitary fibrous tumors in extra-thoracic locations. Evidence of their comparability to intra-thoracic tumors. Am J Surg Pathol 22:1501–1511 Hasegawa T, Matsuno Y, Shimoda T et al, 1999, Extrathoracic solitary fibrous tumors: their histological variability and potentially aggressive behaviour. Hum Pathol 30:1464–1473 Enzinger FM, Weiss SW, 1997, Soft tissue tumors, 4th edn. Mosby, USA Moran CA, Suster S, Koss MN, 1992, The spectrum of histologic growth patterns in benign and malignant fibrous tumors of the pleura. Sem Diagn Pathol 9:169–180 Nielsen GP, O’Connell JX, Dickersin GR et al, 1997, Solitary fibrous tumor of soft tissue. A report of 15 cases, including 5 malignant examples with light microscopic, immunohistochemical, and ultrastructural data. Mod Pathol 10:1028–1037 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2008 VL 14 IS 2 BP 213 EP 217 DI 10.1007/s12253-008-9055-7 PG 5 ER PT J AU Deli, T Csernoch, L AF Deli, Tamas Csernoch, Laszlo TI Extracellular ATP and Cancer—An Overview with Special Reference to P2 Purinergic Receptors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Extracellular ATP; P2 purinergic receptors; Cancer; Proliferation; Differentiation; Apoptosis ID Extracellular ATP; P2 purinergic receptors; Cancer; Proliferation; Differentiation; Apoptosis AB Purinergic signal transduction mechanisms have been appreciated as a complex intercellular signalling network that plays an important regulatory role in both short- and long-term processes in practically every living cell. One of the most intriguing aspects of the field is the participation of ATP and other purine nucleotides in the determination of cell fate and the way they direct cells towards proliferation, differentiation or apoptosis, thereby possibly taking part in promoting or preventing malignant transformation. In this review, following a very brief introduction to the historical aspects of purinergic signalling and a concise overview of the structure of and signal transduction pathways coupled to P2 purinergic receptors, the current theories concerning the possible ways how extracellular ATP can alter the function of tumour cells and the effectiveness of anticancer therapies are discussed, including pharmacological, nutritional, vasoactive and ‘anti-antioxidant’ actions of the nucleotide. The effects of ATP on animals inoculated with human tumours and on patients with cancer are looked over next, and then an overview of the literature regarding the expression and presumed functions of P2 purinoceptors on tumour cells in vitro is presented, sorted out according to the relevant special clinical fields. The article is closed by reviewing the latest developments in the diagnostic use of P2 purinergic receptors as tumour markers and prognostic factors, while discussing some of the difficulties and pitfalls of the therapeutic use of ATP analogues. C1 [Deli, Tamas] University of Debrecen, Faculty of Medicine, Department of Physiology, 98 Nagyerdei krt., 4012 Debrecen, Hungary. [Csernoch, Laszlo] University of Debrecen, Faculty of Medicine, Department of Physiology, 98 Nagyerdei krt., 4012 Debrecen, Hungary. RP Csernoch, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Physiology, 4012 Debrecen, Hungary. EM csl@phys.dote.hu CR Drury AN, Szent-Gyorgyi A, 1929, The physiological activity of adenine compounds with special reference to their action upon the mammalian heart. J Physiol 68:213–237 Holton P, 1959, The liberation of adenosine triphosphate on antidromic stimulation of sensory nerves. J Physiol, Lond, 145:494–504 Martinson J, Muren A, 1963, Excitatory and inhibitory effects of vagus stimulation on gastric motility in the cat. Acta Physiol Scand 57:309–316 BurnstockG, 1972, Purinergic nerves. Pharmacol Rev 24:509–581 Burnstock G, 1978, A basis for distinguishing two types of purinergic receptor. In: Straub RW and Bolis L, eds, Cell membrane receptors for drugs and hormones: a multidisciplinary approach. Raven, New York, pp 107–118 Burnstock G, Kennedy C, 1985, Is there a basis for distinguishing two types of P2-purinoceptor? Gen Pharmacol 16:433–440 Lazarowski E, 2006, Regulated release of nucleotides and UDP sugars from astrocytoma cells. Novartis Found Symp 276:73–84 discussion 84–90, 107–12, 275–81 Selzner N, Selzner M, Graf R, Ungethuem U, Fitz JG, Clavien PA, 2004, Water induces autocrine stimulation of tumor cell killing through ATP release and P2 receptor binding. Cell Death Differ Suppl 2:S172–180 Mazurek S, Boschek CB, Eigenbrodt E, 1997, The role of phosphometabolites in cell proliferation, energy metabolism, and tumor therapy. J Bioenerg Biomembranes 29(4):315–330 Zimmermann H, 2001, Ectonucleotidases: some recent developments and a note on nomenclature. Drug Dev Res 52:44–56 Bavaresco L, Bernardi A, Braganhol E, Wink MR, Battastini AM, 2007, Dexamethasone inhibits proliferation and stimulates ecto-5′-nucleotidase/CD73 activity in C6 rat glioma cell line. J Neurooncol 84(1):1–8 Kunzli BM, Berberat PO, Giese T, Csizmadia E, Kaczmarek E, Baker C, Halaceli I, Buchler MW, Friess H, Robson SC, 2007, Upregulation of CD39/NTPDases and P2 receptors in human pancreatic disease. Am J Physiol Gastrointest Liver Physiol 292, 1):G223–G230 Jackson SW, Hoshi T, Wu Y, Sun X, Enjyoji K, Csizmadia E, Sundberg C, Robson SC, 2007, Disordered purinergic signaling inhibits pathological angiogenesis in cd39/Entpd1-null mice. Am J Pathol 171(4):1395–1404 Merighi S, Mirandola P, Varani K, Gessi S, Leung E, Baraldi PG, Tabrizi MA, Borea PA, 2003, A glance at adenosine receptors: novel target for antitumor therapy. Pharmacol Ther 100(1): 31–48 North RA, 2002, Molecular physiology of P2X receptors. Physiol Rev 82(4):1013–1067 Bean BP, 1992, Pharmacology and electrophysiology of ATPactivated ion channels. Trends Pharmacol Sci 13(3):87–90 Egan TM, Samways DS, Li Z, 2006, Biophysics of P2X receptors. Pflugers Arch 452(5):501–512 Thomas SA, Hume RI, 1990, Permeation of both cations and anions through a single class of ATP-activated ion channels in developing chick skeletal muscle. J Gen Physiol 95:569–590 Kim M, Jiang LH, Wilson HL, North RA, Surprenant A, 2001, Proteomic and functional evidence for a P2X7 receptor signalling complex. EMBO J 20(22):6347–6358 Kahlenberg JM, Dubyak GR, 2004, Mechanisms of caspase-1 activation by P2X7 receptor-mediated K+ release. Am J Physiol Cell Physiol 286(5):C1100–C1108 Bulanova E, Budagian V, Orinska Z, Hein M, Petersen F, Thon L, Adam D, Bulfone-Paus S, 2005, Extracellular ATP induces cytokine expression and apoptosis through P2X7 receptor in murine mast cells. J Immunol 174(7):3880–3890 Wang Q, Wang L, Feng YH, Li X, Zeng R, Gorodeski GI, 2004, P2X7 receptor-mediated apoptosis of human cervical epithelial cells. Am J Physiol Cell Physiol 287(5):C1349–C1358 Ferrari D, Chiozzi P, Falzoni S, Dal Susino M, Melchiorri L, Baricordi OR, Di Virgilio F, 1997, Extracellular ATP triggers IL-1 beta release by activating the purinergic P2Z receptor of human macrophages. J Immunol 159(3):1451–1458 Verhoef PA, Estacion M, Schilling W, Dubyak GR, 2003, P2X7 receptor-dependent blebbing and the activation of Rho-effector kinases, caspases, and IL-1 beta release. J Immunol 170, 11):5728–5738 Chiozzi P, Murgia M, Falzoni S, Ferrari D, Di Virgilio F, 1996, Role of the purinergic P2Z receptor in spontaneous cell death in J774 macrophage cultures. Biochem Biophys Res Commun 218, 1):176–181 Ralevic V, Burnstock G, 1998, Receptors for purines and pyrimidines. Pharmacol Rev 50(3):413–492 Burnstock G, 2006, Purinergic signalling. Br J Pharmacol 147, Suppl 1):S172–S181 Burnstock G, 2006, Purinergic signalling—an overview. Novartis Found Symp 276:26–48 discussion 48–57, 275–81 Hollopeter G, Jantzen HM, Vincent D, Li G, England L, Ramakrishnan V, Yang RB, Nurden P, Nurden A, Julius D, Conley PB, 2001, Identification of the platelet ADP receptor targeted by antithrombotic drugs. Nature 409:202–207 Marteau F, Le Poul E, Communi D, Communi D, Labouret C, Savi P, Boeynaems J-M, Suarez Gonzalez N, 2003, Pharmacological characterization of the human P2Y13 receptor. Mol Pharmacol 64:104–112 Szigeti GP, Szappanos H, Deli T, Cseri J, Kovacs L, Csernoch L, 2007, Differentiation-dependent alterations in the extracellular ATP-evoked calcium fluxes of cultured skeletal muscle cells from mice. Pflugers Arch 453(4):509–518 Deli T, Szappanos H, Szigeti GP, Cseri J, Kovacs L, Csernoch L, 2007, Contribution from P2X and P2Y purinoreceptors to ATPevoked changes in intracellular calcium concentration on cultured myotubes. Pflugers Arch 453(4):519–529 Burnstock G, 2006, Pathophysiology and therapeutic potential of purinergic signaling. Pharmacol Rev 58(1):58–86 Rapaport E, 1983, Treatment of human tumor cells with ADP or ATP yields arrest of growth in the S phase of the cell cycle. J Cell Physiol 114(3):279–283 Rapaport E, Fishman RF, Gercel C, 1983, Growth inhibition of human tumor cells in soft-agar cultures by treatment with low levels of adenosine 5′-triphosphate. Cancer Res 43(9):4402– 4406 Rapaport E, 1988, Experimental cancer therapy in mice by adenine nucleotides. Eur J Cancer Clin Oncol 24(9):1491–1497 Wiley JS, Dao-Ung LP, Gu BJ, Sluyter R, Shemon AN, Li C, Taper J, Gallo J, Manoharan A, 2002, A loss-of-function polymorphic mutation in the cytolytic P2X7 receptor gene and chronic lymphocytic leukaemia: a molecular study. Lancet 359, 9312):1114–1119 Zhang XJ, Zheng GG, Ma XT, Yang YH, Li G, Rao Q, Nie K, Wu KF, 2004, Expression of P2X7 in human hematopoietic cell lines and leukemia patients. Leuk Res 28(12):1313–1322 Deli T, Varga N, Adam A, Kenessey I, Raso E, Puskas LG, Tovari J, Fodor J, Feher M, Szigeti GP, Csernoch L, Timar J, 2007, Functional genomics of calcium channels in human melanoma cells. Int J Cancer 121(1):55–65 Feng YH, Li X, Zeng R, Gorodeski GI, 2006, Endogenously expressed truncated P2X7 receptor lacking the C-terminus is preferentially upregulated in epithelial cancer cells and fails to mediate ligand-induced pore formation and apoptosis. Nucleosides Nucleotides Nucleic Acids 25(9–11):1271–1276 Krzeminski P, Misiewicz I, Pomorski P, Kasprzycka-Guttman T, Branska J, 2007, Mitochondrial localization of P2Y1, P2Y2 and P2Y12 receptors in rat astrocytes and glioma C6 cells. Brain Res Bull 71(6):587–592 Rapaport E, Fontaine J, 1989, Anticancer activities of adenine nucleotides in mice are mediated through expansion of erythrocyte ATP pools. Proc Natl Acad Sci USA 86(5):1662– 1666 Rapaport E, Fontaine J, 1989, Generation of extracellular ATP in blood and its mediated inhibition of host weight loss in tumorbearing mice. Biochem Pharmacol 38(23):4261–4266 Ristow M, 2006, Oxidative metabolism in cancer growth. Curr Opin Clin Nutr Metab Care 9(4):339–345 Baba T, Fukui M, Sakata S, Tashima T, Takeshita I, Nakamura T, Inoue T, 1989, Selective enhancement of intratumoural blood flow in malignant gliomas: experimental study in rats by intracarotid administration of adenosine or adenosine triphosphate. Acta Neurochir, Wien, 101(1–2):66–74 Estrela JM, Obrador E, Navarro J, Lasso De la Vega MC, Pellicer JA, 1995, Elimination of Ehrlich tumours by ATPinduced growth inhibition, glutathione depletion and X-rays. Nat Med 1(1):84–88 Obrador E, Navarro J, Mompo J, Asensi M, Pellicer JA, Estrela JM, 1997, Glutathione and the rate of cellular proliferation determine tumour cell sensitivity to tumour necrosis factor in vivo. Biochem J 325(Pt 1):183–189 Shabbir M, Thompson C, Jarmulowiczc M, Mikhailidis D, Burnstock G, 2008, Effect of extracellular ATP on the growth of hormone-refractory prostate cancer in vivo. BJU Int 102, 1):108–112 Shabbir M, Ryten M, Thompson C, Mikhailidis D, Burnstock G, 2008, Purinergic receptor-mediated effects of ATP in high-grade bladder cancer. BJU Int 101(1):106–112 Abraham EH, Salikhova AY, Rapaport E, 2003, ATP in the treatment of advanced cancer. Curr Top Membr 54:415–452 Haskell CM, Wong M, Williams A, Lee LY, 1996, Phase I trial of extracellular adenosine 5′-triphosphate in patients with advanced cancer. Med Pediatr Oncol 27(3):165–173 Haskell CM, Mendoza E, Pisters KM, Fossella FV, Figlin RA, 1998, Phase II study of intravenous adenosine 5′-triphosphate in patients with previously untreated stage IIIB and stage IV nonsmall cell lung cancer. Invest New Drugs 16(1):81–85 Agteresch HJ, Dagnelie PC, van der Gaast A, Stijnen T, Wilson JH, 2000, Randomized clinical trial of adenosine 5′- triphosphate in patients with advanced non-small-cell lung cancer. J Natl Cancer Inst 92(4):321–328 Agteresch HJ, Leij-Halfwerk S, Van Den Berg JW, Hordijk- Luijk CH, Wilson JH, Dagnelie PC, 2000, Effects of ATP infusion on glucose turnover and gluconeogenesis in patients with advanced non-small-cell lung cancer. Clin Sci, Lond, 98, 6):689–695 Leij-Halfwerk S, Agteresch HJ, Sijens PE, Dagnelie PC, 2002, Adenosine triphosphate infusion increases liver energy status in advanced lung cancer patients: an in vivo 31P magnetic resonance spectroscopy study. Hepatology 35(2):421–424 Agteresch HJ, Burgers SA, van der Gaast A, Wilson JH, Dagnelie PC, 2003, Randomized clinical trial of adenosine 5′- triphosphate on tumor growth and survival in advanced lung cancer patients. Anticancer Drugs 14(8):639–644 Hatta Y, Takahashi M, Enomoto Y, Takahashi N, Sawada U, Horie T, 2004, Adenosine triphosphate, ATP, enhances the antitumor effect of etoposide, VP16, in lung cancer cells. Oncol Rep 12:1139–1142 Shabbir M, Thompson CS, Mikhailidis DP, Morgan RJ, Burnstock G, 2004, Adenosine 5-triphosphate, ATP, increases the cytotoxic effect of mitomycin C in the treatment of high-grade bladder cancer. J Urol 171(Suppl 4):260 Schafer R, Sedehizade F, Welte T, Reiser G, 2003, ATP- and UTP-activated P2Y receptors differently regulate proliferation of human lung epithelial tumor cells. Am J Physiol Lung Cell Mol Physiol 285:L376–L385 Shabbir M, Ryten M, Thompson C, Mikhailidis D, Burnstock G, 2008, Characterization of calcium-independent purinergic receptor- mediated apoptosis in hormone-refractory prostate cancer. BJU Int 101(3):352–359 Slater M, Danieletto S, Barden JA, 2005, Expression of the apoptotic calcium channel P2X7 in the glandular epithelium. J Mol Histol 36(3):159–165 Slater M, Danieletto S, Gidley-Baird A, Teh LC, Barden JA, 2004, Early prostate cancer detected using expression of nonfunctional cytolytic P2X7 receptors. Histopathology 44(3):206– 215 Li X, Zhou L, Feng YH, Abdul-Karim FW, Gorodeski GI, 2006, The P2X7 receptor: a novel biomarker of uterine epithelial cancers. Cancer Epidemiol Biomarkers Prev 15(10):1906–1913 Czajkowski R, Lei L, Sabala P, Baranska J, 2002, ADP-evoked phospholipase C stimulation and adenylyl cyclase inhibition in glioma C6 cells occur through two distinct nucleotide receptors, P2Y(1, and P2Y(12). FEBS Lett 513(2–3):179–183 Krzeminski P, Suplat D, Czajkowski R, Pomorski P, Baranska J, 2007, Expression and functional characterization of P2Y1 and P2Y12 nucleotide receptors in long-term serum-deprived glioma C6 cells. FEBS J 274(8):1970–1982 Van Kolen K, Slegers H, 2006, Atypical PKCzeta is involved in RhoA-dependent mitogenic signaling by the P2Y(12, receptor in C6 cells. FEBS J 273(8):1843–1854 Morrone FB, Jacques-Silva MC, Horn AP, Bernardi A, Schwartsmann G, Rodnight R, Lenz G, 2003, Extracellular nucleotides and nucleosides induce proliferation and increase nucleoside transport in human glioma cell lines. J Neurooncol 64, 3):211–218 Cavaliere F, Nestola V, Amadio S, D’Ambrosi N, Angelini DF, Sancesario G, Bernardi G, Volonte C, 2005, The metabotropic P2Y4 receptor participates in the commitment to differentiation and cell death of human neuroblastoma SH-SY5Y cells. Neurobiol Dis 18(1):100–109 Lakshmi S, Joshi PG, 2006, Activation of Src/kinase/phospholipase C/mitogen-activated protein kinase and induction of neurite expression by ATP, independent of nerve growth factor. Neuroscience 141(1):179–189 Raffaghello L, Chiozzi P, Falzoni S, Di Virgilio F, Pistoia V, 2006, The P2X7 receptor sustains the growth of human neuroblastoma cells through a substance P-dependent mechanism. Cancer Res 66(2):907–914 Janssens R, Boeynaems JM, 2001, Effects of extracellular nucleotides and nucleosides on prostate carcinoma cells. Br J Pharmacol 132(2):536–546 Calvert RC, Shabbir M, Thompson CS, Mikhailidis DP, Morgan RJ, Burnstock G, 2004, Immunocytochemical and pharmacological characterisation of P2-purinoceptor-mediated cell growth and death in PC-3 hormone refractory prostate cancer cells. Anticancer Res 24(5A):2853–2859 Chen L, He HY, Li HM, Zheng J, Heng WJ, You JF, Fang WG, 2004, ERK1/2 and p38 pathways are required for P2Y receptormediated prostate cancer invasion. Cancer Lett 215(2):239–247 Katzur AC, Koshimizu T, Tomic M, Schultze-Mosgau A, Ortmann O, Stojilkovic SS, 1999, Expression and responsiveness of P2Y2 receptors in human endometrial cancer cell lines. J Clin Endocrinol Metab 84(11):4085–4091 Schultze-Mosgau A, Katzur AC, Arora KK, Stojilkovic SS, Diedrich K, Ortmann O, 2000, Characterization of calciummobilizing, purinergic P2Y(2, receptors in human ovarian cancer cells. Mol Hum Reprod 6(5):435–442 Choi KC, Tai CJ, Tzeng CR, Auersperg N, Leung PC, 2003, Adenosine triphosphate activates mitogen-activated protein kinase in pre-neoplastic and neoplastic ovarian surface epithelial cells. Biol Reprod 68(1):309–315 Popper LD, Batra S, 1993, Calcium mobilization and cell proliferation activated by extracellular ATP in human ovarian tumour cells. Cell Calcium 14(3):209–218 Vandewalle B, Hornez L, Revillion F, Lefebvre J, 1994, Effect of extracellular ATP on breast tumor cell growth, implication of intracellular calcium. Cancer Lett 85(1):47–54 Dixon CJ, Bowler WB, Fleetwood P, Ginty AF, Gallagher JA, Carron JA, 1997, Extracellular nucleotides stimulate proliferation in MCF-7 breast cancer cells via P2-purinoceptors. Br J Cancer 75(1):34–39 Slater M, Danieletto S, Pooley M, Cheng Teh L, Gidley-Baird A, Barden JA, 2004, Differentiation between cancerous and normal hyperplastic lobules in breast lesions. Breast Cancer Res Treat 83, 1):1–10 Gow IF, Thomson J, Davidson J, Shennan DB, 2005, The effect of a hyposmotic shock and purinergic agonists on K+(Rb+, efflux from cultured human breast cancer cells. Biochim Biophys Acta 1712(1):52–61 Maaser K, Hopfner M, Kap H, Sutter AP, Barthel B, von Lampe B, Zeitz M, Scherubl H, 2002, Extracellular nucleotides inhibit growth of human oesophageal cancer cells via P2Y(2)-receptors. Br J Cancer 86(4):636–644 Wang MX, Ren LM, 2006, Growth inhibitory effect and apoptosis induced by extracellular ATP and adenosine on human gastric carcinoma cells: involvement of intracellular uptake of adenosine. Acta Pharmacol Sin 27(8):1085–1092 Hopfner M, Lemmer K, Jansen A, Hanski C, Riecken EO, Gavish M, Mann B, Buhr H, Glassmeier G, Scherubl H, 1998, Expression of functional P2-purinergic receptors in primary cultures of human colorectal carcinoma cells. Biochem Biophys Res Commun 251(3):811–817 Coutinho-Silva R, Stahl L, Cheung KK, de Campos NE, de Oliveira Souza C, Ojcius DM, Burnstock G, 2005, P2X and P2Y purinergic receptors on human intestinal epithelial carcinoma cells: effects of extracellular nucleotides on apoptosis and cell proliferation. Am J Physiol Gastrointest Liver Physiol 288(5): G1024–G1035 Nylund G, Nordgren S, Delbro DS, 2004, Expression of P2Y2 purinoceptors in MCG 101 murine sarcoma cells, and HT-29 human colon carcinoma cells. Auton Neurosci 112(1–2):69–79 Delbro DS, Nylund G, Nordgren S, 2005, Demonstration of P2Y4 purinergic receptors in the HT-29 human colon cancer cell line. Auton Autacoid Pharmacol 25(4):163–166 Nylund G, Hultman L, Nordgren S, Delbro DS, 2007, P2Y2- and P2Y4 purinergic receptors are over-expressed in human colon cancer. Auton Autacoid Pharmacol 27(2):79–84 PinesA, Perrone L, Bivi N, RomanelloM,DamanteG, Gulisano M, Kelley MR, Quadrifoglio F, Tell G, 2005, Activation of APE1/ Ref-1 is dependent on reactive oxygen species generated after purinergic receptor stimulation by ATP. Nucleic Acids Res 33(14): 4379–4394 Pines A, Bivi N, Vascotto C, Romanello M, D’Ambrosio C, Scaloni A, Damante G, Morisi R, Filetti S, Ferretti E, Quadrifoglio F, Tell G, 2006, Nucleotide receptors stimulation by extracellular ATP controls Hsp90 expression through APE1/ Ref-1 in thyroid cancer cells: a novel tumorigenic pathway. J Cell Physiol 209(1):44–55 SoliniA, Cuccato S, Ferrari D, Santini E, Gulinelli S, CallegariMG, Dardano A, Faviana P, Madec S, Di Virgilio F, Monzani F, 2008, Increased P2X7 receptor expression and function in thyroid papillary cancer: a new potential marker of the disease? Endocrinology 149(1):389–396 Hatta Y, Aizawa S, Itoh T, Baba M, Horie T, 1994, Cytotoxic effect of extracellular ATP on L1210 leukemic cells and normal hemopoietic stem cells. Leuk Res 18(8):637–641 Conigrave AD, van der Weyden L, Holt L, Jiang L, Wilson P, Christopherson RI,MorrisMB, 2000, ExtracellularATP-dependent suppression of proliferation and induction of differentiation of human HL-60 leukemia cells by distinct mechanisms. Biochem Pharmacol 60(11):1585–1591 Adinolfi E, Melchiorri L, Falzoni S, Chiozzi P,Morelli A, Tieghi A, Cuneo A, Castoldi G, Di Virgilio F, Baricordi OR, 2002, P2X7 receptor expression in evolutive and indolent forms of chronic B lymphocytic leukemia. Blood 99(2):706–708 Bernardo AA, Pinto-Silva FE, Persechini PM, Coutinho-Silva R, Meyer-Fernandes JR, de Souza AL, Rumjanek VM, 2006, Effect of extracellular ATP on the human leukaemic cell line K562 and its multidrug counterpart. Mol Cell Biochem 289(1– 2):111–124 Yoon MJ, Lee HJ, Kim JH, Kim DK, 2006, Extracellular ATP induces apoptotic signaling in human monocyte leukemic cells, HL-60 and F-36P. Arch Pharm Res 29(11):1032– 1041 Shemon AN, Sluyter R, Wiley JS, 2007, Rottlerin inhibits P2X, 7, receptor-stimulated phospholipase D activity in chronic lymphocytic leukaemia B-lymphocytes. Immunol Cell Biol 85, 1):68–72 Greig AV, Linge C, Healy V, Lim P, Clayton E, Rustin MH, McGrouther DA, Burnstock G, 2003, Expression of purinergic receptors in non-melanoma skin cancers and their functional roles in A431 cells. J Invest Dermatol 121(2):315–327 White N, Ryten M, Clayton E, Butler P, Burnstock G, 2005, P2Y purinergic receptors regulate the growth of human melanomas. Cancer Lett 224(1):81–91 White N, Butler PE, Burnstock G, 2005, Human melanomas express functional P2X(7, receptors. Cell Tissue Res 321, 3):411–418 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 219 EP 231 DI 10.1007/s12253-008-9071-7 PG 13 ER PT J AU Morgenstern, D Rees, H Sebire, N Shipley, J Anderson, J AF Morgenstern, A. Daniel Rees, Helen Sebire, J. Neil Shipley, Janet Anderson, John TI Rhabdomyosarcoma Subtyping by Immunohistochemical Assessment of Myogenin: Tissue Array Study and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Rhabdomyosarcoma; Myogenin; Tissue array; Diagnosis ID Rhabdomyosarcoma; Myogenin; Tissue array; Diagnosis AB Myogenin immunostaining has been described as a useful marker of the alveolar subtype of rhabdomyosarcoma and as a tool for distinguishing it from the more common embryonal subtype. To add to the growing body of literature describing this phenomenon we analysed myogenin immunohistochemical staining in 152 tumors using a rhabdomyosarcoma tissue array. Results were analysed blinded to histological type by two independent investigators. Samples were excluded if any samples failed to stain with desmin and/or myogenin. Mean percentage of myogenin positive cells was significantly greater for ARMS (n=31; mean percentage positivity 59% (95% confidence intervals ± 7%) than ERMS (n=41, mean percentage positivity 16%, 95% confidence intervals ± 4; P<0.0001). This data is consistent with previously published studies identifying strong nuclear myogenin staining in a high proportion of cells as a marker of alveolar histology. C1 [Morgenstern, A. Daniel] Institute of Child Health, Unit of Molecular Haematology and Cancer Biology, 30 Guilford Street, WC1N 1EH London, UK. [Rees, Helen] Institute of Child Health, Unit of Molecular Haematology and Cancer Biology, 30 Guilford Street, WC1N 1EH London, UK. [Sebire, J. Neil] Great Ormond Street Hospital for Children, Department of HistopathologyLondon, UK. [Shipley, Janet] Intsitute of Cancer Research, Department of Molecular CytogeneticsSutton, Surrey, UK. [Anderson, John] Institute of Child Health, Unit of Molecular Haematology and Cancer Biology, 30 Guilford Street, WC1N 1EH London, UK. RP Anderson, J (reprint author), Institute of Child Health, Unit of Molecular Haematology and Cancer Biology, WC1N 1EH London, UK. EM j.Anderson@ich.ul.ac.uk CR McManus AP, O'Reilly MA, Jones KP, Gusterson BA, Mitchell CD, Pinkerton CR, Shipley JM, 1996, Interphase fluorescence in situ hybridization detection of t(2;13)(q35;q14, in alveolar rhabdomyosarcoma–a diagnostic tool in minimally invasive biopsies. J Pathol 178:410–414 Sapi Z, Antal I, Papai Z, Szendroi M, Mayer A, Jakab K, Pajor L, Bodo M, 2002, Diagnosis of soft tissue tumors by fine-needle aspiration with combined cytopathology and ancillary techniques. Diagn Cytopathol 26:232–242 Anderson J, Gordon T, McManus A, Mapp T, Gould S, Kelsey A, McDowell H, Pinkerton R, Shipley J, Pritchard-Jones K, 2001, Detection of the PAX3-FKHR fusion gene in paediatric rhabdomyosarcoma: a reproducible predictor of outcome? Br J Cancer 85:831–835 Sorensen PH, Lynch JC, Qualman SJ, Tirabosco R, Lim JF, Maurer HM, Bridge JA, Crist WM, Triche TJ, Barr FG, 2002, PAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicators in alveolar rhabdomyosarcoma: a report from the children's oncology group. J Clin Oncol 20:2672–2679 Sebire NJ, Malone M, 2003, Myogenin and MyoD1 expression in paediatric rhabdomyosarcomas. J Clin Pathol 56:412–416 Wachtel M, Runge T, Leuschner I, Stegmaier S, Koscielniak E, Treuner J, Odermatt B, Behnke S, Niggli FK, Schafer BW, 2006, Subtype and prognostic classification of rhabdomyosarcoma by immunohistochemistry. J Clin Oncol 24:816–822 Cessna MH, Zhou H, Perkins SL, Tripp SR, Layfield L, Daines C, Coffin CM, 2001, Are myogenin and myoD1 expression specific for rhabdomyosarcoma? A study of 150 cases, with emphasis on spindle cell mimics. Am J Surg Pathol 25:1150–1157 Wang NP, Marx J, McNutt MA, Rutledge JC, Gown AM, 1995, Expression of myogenic regulatory proteins, myogenin and MyoD1, in small blue round cell tumors of childhood. Am J Pathol 147:1799–1810 Folpe AL, 2002, MyoD1 and myogenin expression in human neoplasia: a review and update. Adv Anat Pathol 9:198–203 Kumar S, Perlman E, Harris CA, Raffeld M, Tsokos M, 2000, Myogenin is a specific marker for rhabdomyosarcoma: an immunohistochemical study in paraffin-embedded tissues. Mod Pathol 13:988–993 Hostein I, Andraud-Fregeville M, Guillou L, Terrier-Lacombe MJ, Deminiere C, Ranchere D, Lussan C, Longavenne E, Bui NB, Delattre O, Coindre JM, 2004, Rhabdomyosarcoma: value of myogenin expression analysis and molecular testing in diagnosing the alveolar subtype: an analysis of 109 paraffin-embedded specimens. Cancer 101:2817–2824 Dias P, Chen B, Dilday B, Palmer H, Hosoi H, Singh S, Wu C, Li X, Thompson J, Parham D, Qualman S, Houghton P, 2000, Strong immunostaining for myogenin in rhabdomyosarcoma is significantly associated with tumors of the alveolar subclass. Am J Pathol 156:399–408 Morotti RA, Nicol KK, Parham DM, Teot LA, Moore J, Hayes J, Meyer W, Qualman SJ, 2006, An immunohistochemical algorithm to facilitate diagnosis and subtyping of rhabdomyosarcoma: the Children's Oncology Group experience. Am J Surg Pathol 30:962–968 Stevens MC, Rey A, Bouvet N, Ellershaw C, Flamant F, Habrand JL, Marsden HB, Martelli H, Sanchez de Toledo J, Spicer RD, Spooner D, Terrier-Lacombe MJ, van Unnik A, Oberlin O, 2005, Treatment of nonmetastatic rhabdomyosarcoma in childhood and adolescence: third study of the International Society of Paediatric Oncology–SIOP Malignant Mesenchymal Tumor 89. J Clin Oncol 23:2618–2628 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 233 EP 238 DI 10.1007/s12253-008-9012-5 PG 6 ER PT J AU Dova, L Golfinopoulos, V Pentheroudakis, G Georgiou, I Pavlidis, N AF Dova, Lefkothea Golfinopoulos, Vassilis Pentheroudakis, George Georgiou, Ioannis Pavlidis, Nicholas TI Systemic Dissemination in Cancer of Unknown Primary is Independent of Mutational Inactivation of the KiSS-1 Metastasis-suppressor Gene SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CUP; KiSS-1 metastasis-suppressor gene; Polymerase chain reaction; KiSS-1; FFPE ID CUP; KiSS-1 metastasis-suppressor gene; Polymerase chain reaction; KiSS-1; FFPE AB Cancer of unknown primary represents a heterogeneous group of malignancies characterised by early systemic dissemination and lack of primary site. KiSS1 is a member of the metastasis-suppressor gene family whose functional role is being investigated in human malignancies. We extracted DNA from 50 paraffin-embedded unknown primary tumors and screened KiSS1 exons III and IV for presence of mutations by means of Single Strand Conformational Polymorphism and direct sequencing. Only one tumor specimen harboured a cytosine to guanine point substitution in base 242 of exon IVa, resulting in a proline to arginine switch at codon 81 of the KiSS1 protein (P81R). The remaining 49 tumors harbored wild-type KiSS1 alleles, indistinguishable from those of peripheral blood lymphocytes of 50 healthy controls. Consequently, the propensity for systemic spread of unknown primary tumors may by due to mutations in genes other than KiSS1 or aberrant epigenetic regulation. C1 [Dova, Lefkothea] Ioannina University Hospital, Hematological LaboratoryIoannina, Greece. [Golfinopoulos, Vassilis] Ioannina University Hospital, Department of Medical OncologyIoannina, Greece. [Pentheroudakis, George] Ioannina University Hospital, Department of Medical OncologyIoannina, Greece. [Georgiou, Ioannis] Ioannina University Hospital, Department of Obstetrics and GynecologyIoannina, Greece. [Pavlidis, Nicholas] Ioannina University Hospital, Department of Medical OncologyIoannina, Greece. RP Pavlidis, N (reprint author), Ioannina University Hospital, Department of Medical Oncology, Ioannina, Greece. EM npavlid@cc.uoi.gr CR Yoshida BA, Sokoloff M, Welch DR, Rinker-Schaeffer CW, 2000, Metastasis-suppressor genes: a review and perspective on an emerging field. J Natl Cancer Inst 92:1717–1730 West A, Vojta PJ, Welch DR, Weissman BE, 1998, Chromosome localization and genomic structure of the KiSS-1 metastasis suppressor gene, KISS1). Genomics 54:145–148 Nash KT, Welch DR, 2006, The KISS1 metastasis suppressor: mechanistic insights and clinical utility. Front Biosci 11:647– 659 Pentheroudakis G, Kostadima L, Dova L, Malamou-Mitsi V, Georgiou I et al, 2006, The missing kiss of life: Analysis of mutation and protein expression of the metastasis-suppressor gene KISS1 in early breast cancer. Ann Oncol 17(Suppl 9):ix59, 101P) Pentheroudakis G, Briasoulis E, Pavlidis N, 2007, Cancer of unknown primary site: missing primary or missing biology? Oncologist 12:418–425 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 239 EP 241 DI 10.1007/s12253-008-9024-1 PG 3 ER PT J AU Mojica, DW Stein, L Hawthorn, L AF Mojica, D Wilfrido Stein, Leighton Hawthorn, Lesleyann TI Universal Reference RNA is Not a Representative Normal Sample for Oligonucleotide Microarray Studies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Reference RNA; Oligonucleotide expression microarray; Experimental bias ID Reference RNA; Oligonucleotide expression microarray; Experimental bias AB Translational research has been defined as the scientific study using human material that will ultimately generate patient specific data. A major caveat in human directed study is the availability of high quality and quantities of patient derived homogeneous cells for analysis. Whereas there exist sources for which tumor tissue and blood samples can be made available, the same cannot be said for normal tissue. The absence of normal control tissue has led to the creation of pooled cell lines and tissues for purchase known as "reference RNA". Although initially created for purposes of standardization, the difficulty associated with acquiring normal tissue has led some investigators to use sources of universal pooled RNA for comparative analysis with clinical tissue specimens. In order to study the effects of using Universal Reference RNA on expression profiling experiments we have evaluated the performance of universal RNA compared to RNA obtained from a purified population of colon epithelial cells in defining a set of altered transcripts in colon cancer. C1 [Mojica, D Wilfrido] State University of New York, University at Buffalo, Department of Pathology, 100 High Street, 14203 Buffalo, NY, USA. [Stein, Leighton] Roswell Park Cancer Institute, Gene Expression Facility, Elm and Carlton Streets, 14263 Buffalo, NY, USA. [Hawthorn, Lesleyann] Roswell Park Cancer Institute, Gene Expression Facility, Elm and Carlton Streets, 14263 Buffalo, NY, USA. RP Mojica, DW (reprint author), State University of New York, University at Buffalo, Department of Pathology, 14203 Buffalo, USA. EM mojica@buffalo.edu CR van’t Veer LJ, Paik S, Hayes DF, 2005, Gene expression profiling of breast cancer: a new tumor marker. J Clin Oncol 23(8):1631– 1635 Cronin M, Ghosh K, Sistare F, Quakenbush J, Vilker V, O’Connell C, 2004, Universal RNA reference materials for gene expression. Clin Chem 50:1464–1471 Cowell JK, Hawthorn L, 2007, The application of microarray technology to the analysis of the cancer genome. Curr Mol Med 7, 1):103–120 Novoradovskaya N, Whitfield ML, Basehore LS et al, 2005, Universal reference RNA as a standard for microarray experiments. BMC Genom 5:20 Schlecht NF, Burk RD, Adrien L et al, 2007, Gene expression profiles in HPV-infected head and neck cancer. J Pathol 213:283– 293 Larsen JE, Pavey SJ, Passmore LH et al, 2007, Expression profiling defines a recurrence signature in lung squamous cell carcinoma. Carcinogenesis 28(3):760–766 Bianchi F, Hu J, Pelosi G et al, 2004, Lung cancers detected by screening with spiral computed tomography have a malignant phenotype when analyzed by cDNA microarray. Clin Cancer Res 10:6023–6028 Warner GC, Reis PP, Jurisica I et al, 2004, Molecular classification of oral cancer by cDNA microarrays identifies overexpressed genes correlated with nodal metastasis. Int J Cancer 110:857–868 Mojica WD, Arshad A, Sharma S et al, 2006, Manual exfoliation plus immunomagnetic bead separation as an initial step toward translational studies. Arch Pathol Lab Med 130(1):74–79 Mojica WD, Stein L, Hawthorn L, 2007, An exfoliation and enrichment strategy results in improved transcriptional profiles when compared to matched formalin fixed samples. BMC Clin Pathol 7:7 Hawthorn L, Stein L, Panzarella J et al, 2006, Characterization of cell-type specific profiles in tissues and isolated cells from squamous cell carcinomas of the lung. Lung Cancer 53(2):129– 142 Irizarry RA, Hobbs B, Collin F et al, 2003, Exploration, normalization, and summaries of high-density oligonucleotide array probe level data. Biostatistics 4:249–264 Alizadeh AA, Eisen MB, Davis RE et al, 2000, Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 403(6769):503–511 Burgess AW, 1998, Growth control mechanisms in normal and transformed intestinal cells. Philos Tran R Soc Lond B Biol Sci 353:903–909 Pinto M, Robine-Leon S, Appay MD, 1983, Enterocyte like differentiation and polarization of the human colon carcinoma cell line Caco-2 in culture. Bio Cell 47:323–330 Allander SV, Illei PB, Chen Y et al, 2002, Expression profiling of synovial sarcoma by cDNA microarrays. Am J Pathol 161, 5):1587–1595 Nielssen TO, West RB, Linn SC et al, 2002, Molecular characterization of soft tissue tumours: a gene expression study. Lancet 359:1301–1307 MacLeod RAF, Dirks WG, Matsui Y et al, 1999, Widespread intraspecies cross-contamination of human tumor cells arising at source. Int J Cancer 83:555–563 Demichelis F, Gruelich H, Macoska JA et al, 2007, SNP panel identification assay, SPIA): a genetic-based assay for the identification of cell lines. Mod Pathol 20(S2):346A, abstract) Celis A, Rasmussen HH, Celis P et al, 1999, Short term culturing of low grade superficial bladder transitional cell carcinoma leads to changes in the expression levels of several proteins involved in key cellular activities. Electrophoresis 20:355–361 Somel M, Khaitovich P, Bahn S et al, 2006, Gene expression becomes heterogeneous with age. Curr Biol 16(10):R359–360 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 243 EP 251 DI 10.1007/s12253-008-9068-2 PG 9 ER PT J AU Reiner-Concin, A Regitnig, P Dinges, PH Hofler, G Lax, S Muller-Holzner, E Obrist, P Rudas, M AF Reiner-Concin, Angelika Regitnig, Peter Dinges, Peter Hans Hofler, Gerald Lax, Sigurd Muller-Holzner, Elisabeth Obrist, Peter Rudas, Margaretha TI Practice of HER-2 Immunohistochemistry in Breast Carcinoma in Austria SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; HER-2; Immunohistochemistry; Quality assurance; Reproducibility ID Breast cancer; HER-2; Immunohistochemistry; Quality assurance; Reproducibility AB Practice and accuracy of immunohistochemistry is known to vary highly. Reliability of HER-2 immunohistochemistry is critical because of its role in patient selection for therapeutical options in breast cancer. Therefore reliability of HER-2 immunohistochemistry in pathology laboratories in Austria was assessed. Ten tissue specimens of invasive ductal breast carcinomas and three cell line samples were tested. Presence/absence of gene amplification was determined by FISH to be used as a gold standard. Laboratories were asked to stain and assess slides using their routine immunohistochemical staining protocol. Overall the study consisted of 311 tests on tissue specimens and 142 on cell lines. In all cases manual scoring was performed. Participation was voluntary and was 94%. Overall sensitivity was 90.5% and specificity 99.2%. Overscoring including true false positive results were found in 6.7% and 6.3% in tissue specimens and cell lines, respectively. False negative determinations were obtained in 1.9% and 2.8% of tissue specimens and cell lines, respectively. HercepTestTM showed slightly higher reliability in comparison with individualized staining methods. By manual scoring inaccurate scoring affected 12.3% of test results and 62% of the laboratories. In conclusion participation rate and accuracy of HER-immunohistochemistry was high all over the country. Manually performed scoring demonstrated some limitations. C1 [Reiner-Concin, Angelika] Donauspital, Pathologisch-Bakteriologisches InstitutVienna, Austria. [Regitnig, Peter] Medical University of Graz, Department of PathologyGraz, Austria. [Dinges, Peter Hans] Hospital Klagenfurt, Department of PathologyKlagenfurt, Austria. [Hofler, Gerald] Medical University of Graz, Department of PathologyGraz, Austria. [Lax, Sigurd] LKH-West Graz, Department of PathologyGraz, Austria. [Muller-Holzner, Elisabeth] Medical University of Innsbruck, Department of Obstetrics and GynecologyInnsbruck, Austria. [Obrist, Peter] Pathohistological LaboratoryLandeck, Austria. [Rudas, Margaretha] Medical University of Vienna, Department of PathologyVienna, Austria. RP Reiner-Concin, A (reprint author), Donauspital, Pathologisch-Bakteriologisches Institut, Vienna, Austria. EM angelika.reiner@wienkav.at CR Wolff AC, Hammond ME, Schwartz JN et al, 2007, American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 131:18– 43 Gennari A, Sormani MP, Pronzato P et al, 2008, HER2 status and efficacy of adjuvant anthracyclines in early breast cancer: a pooled analysis of randomized trials. J Natl Cancer Inst 100:14–20 Yamauchi H, Stearns V, Hayes DF, 2001, When is a tumor marker ready for prime time? A case study of c-erbB-2 as a predictive factor in breast cancer. J Clin Oncol 19:2334–2356 Konecny G, Pauletti G, Pegram M et al, 2003, Quantitative association between HER-2/neu and steroid hormone receptors in hormone receptor-positive primary breast cancer. J Natl Cancer Inst 95:142–153 Menard S, Valagussa P, Pilotti S et al, 2001, Response to cyclophosphamide, methotrexate, and fluorouracil in lymph node-positive breast cancer according to HER2 overexpression and other tumor biologic variables. J Clin Oncol 19:329–335 Press MF, Hung G, Godolphin Wet al, 1994, Sensitivity of HER- 2/neu antibodies in archival tissue samples: potential source of error in immunohistochemical studies of oncogene expression. Cancer Res 54:2771–2777 Wang S, Saboorian MH, Frenkel EP et al, 2001, Assessment of HER-2/neu status in breast cancer. Automated Cellular Imaging System, ACIS)-assisted quantitation of immunohistochemical assay achieves high accuracy in comparison with fluorescence in situ hybridization assay as the standard. Am J Clin Pathol 116:495–503 ItalianNetwork forQuality Assurance of Tumor Biomarkers, INQAT, Group; SIAPEC-IAP, 2005, Interobserver reproducibility of immunohistochemical HER-2/neu assessment in human breast cancer: an update from INQAT round III. Int J Biol Markers 20:189–194 Paik S, Bryant J, Tan-Chiu E et al, 2002, Real-world performance of HER2 testing—National Surgical Adjuvant Breast and Bowel Project experience. J Natl Cancer Inst 94:852–854 Rhodes A, Jasani B, Anderson E et al, 2002, Evaluation of HER- 2/neu immunohistochemical assay sensitivity and scoring on formalin-fixed and paraffin-processed cell lines and breast tumors: a comparative study involving results from laboratories in 21 countries. Am J Clin Pathol 118:408–417 Fleiss JG, 1981, Statistical methods for rates and proportions. Wiley, New York, NY, pp 225–232 Landis JR, Koch GG, 1977, The measurement of observer agreement for categorical data. Biometrics 33:159–174 Bilous M, Dowsett M, Hanna Wet al, 2003, Current perspectives on HER2 testing: a review of national testing guidelines. Mod Pathol 16:173–182 Ellis IO, Bartlett J, Dowsett M et al, 2004, Best Practice No 176: Updated recommendations for HER2 testing in the UK. J Clin Pathol 57:233–237 Bertheau P, Cazals-Hatem D, Meignin Vet al, 1998, Variability of immunohistochemical reactivity on stored paraffin slides. J Clin Pathol 51:370–374 Rhodes A, Borthwick D, Sykes R et al, 2004, The use of cell line standards to reduce HER-2/neu assay variation in multiple European cancer centers and the potential of automated image analysis to provide for more accurate cut points for predicting clinical response to trastuzumab. Am J Clin Pathol 122:51–60 Jacobs TW, Gown AM, Yaziji H et al, 1999, Comparison of fluorescence in situ hybridization and immunohistochemistry for the evaluation of HER-2/neu in breast cancer. J Clin Oncol 17:1974–1982 Lebeau A, Deimling D, Kaltz C et al, 2001, Her-2/neu analysis in archival tissue samples of human breast cancer: comparison of immunohistochemistry and fluorescence in situ hybridization. J Clin Oncol 19:354–63 Tubbs RR, Pettay JD, Roche PC et al, 2001, Discrepancies in clinical laboratory testing of eligibility for trastuzumab therapy: apparent immunohistochemical false-positives do not get the message. J Clin Oncol 19:2714–2721 Hoang MP, Sahin AA, Ordonez NG et al, 2000, HER-2/neu gene amplification compared with HER-2/neu protein overexpression and interobserver reproducibility in invasive breast carcinoma. Am J Clin Pathol 113:852–859 Thomson TA, Hayes MM, Spinelli JJ et al, 2001, HER-2/neu in breast cancer: interobserver variability and performance of immunohistochemistry with 4 antibodies compared with fluorescent in situ hybridization. Mod Pathol 14:1079–1086 Dietel M, Ellis IO, Hofler H et al, 2007, Comparison of automated silver enhanced in situ hybridisation, SISH, and fluorescence ISH, FISH, for the validation of HER2 gene status in breast carcinoma according to the guidelines of the American Society of Clinical Oncology and the College of American Pathologists. Virchows Arch 51:19–25 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 253 EP 259 DI 10.1007/s12253-008-9079-z PG 7 ER PT J AU Caner, V Turk, SN Duzcan, F Satiroglu-Tufan, L Kelten, CE Zencir, S Dodurga, Y Bagci, H Duzcan, ES AF Caner, Vildan Turk, Sen Nilay Duzcan, Fusun Satiroglu-Tufan, Lale Kelten, Canan Esra Zencir, Sevil Dodurga, Yavuz Bagci, Huseyin Duzcan, Ender Suleyman TI No Strong Association Between HER-2/neu Protein Overexpression and Gene Amplification in High-grade Invasive Urothelial Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE FISH; HER-2/neu; Immunohistochemistry; Real-time quantitative PCR; urothelial carcinoma ID FISH; HER-2/neu; Immunohistochemistry; Real-time quantitative PCR; urothelial carcinoma AB The generation of urothelial carcinoma is caused by the accumulation of various molecular changes, as in most malignancies. There are conflicting data about the status of HER-2/neu oncogene in urothelial carcinomas. The aim of this study was to determine the status of HER-2/neu oncogene in high-grade invasive urothelial carcinoma of urinary bladder both in protein and DNA level. We evaluated HER-2/neu protein overexpression by immunohistochemistry (IHC) and gene amplification by fluorescent in situ hybridization (FISH) and real-time quantitative PCR in paraffin-embedded samples of high-grade invasive urothelial carcinoma obtained from 36 patients. Polysomy 17 was also assessed by FISH. Immunohistochemically, HER-2/neu protein overexpression was observed in 22 (61.1%) tumors (ten tumors with score 3+ and 12 with score 2+). Fourteen of 36 tumors (38.9%) were evaluated as negative (score 0 or 1+). Complete concordance between FISH and the PCR was seen in all of the samples scored as 0 and 1+ by IHC. HER-2/neu gene amplification was observed in three of 27 (11.1%) tumors by FISH (nine samples were non-informative) and in eight of 36 (22.2%) tumors by the PCR. The complete concordance between HER2-2/neu protein overexpression and gene amplification was seen only in three of 27 tumors. Polysomy 17 was seen in nine tumors (33.3%). The results indicated that, in contrast to breast cancer, there was no strong association between HER-2/neu overexpression and gene amplification in invasive urothelial carcinomas, and polysomy 17 was higher in tumors showing HER-2/neu overexpression. C1 [Caner, Vildan] Pamukkale University, Medical Faculty, Department of Medical Biology, 20020 Denizli, Turkey. [Turk, Sen Nilay] Pamukkale University, Faculty of Medicine, Department of Pathology, 20020 Denizli, Turkey. [Duzcan, Fusun] Pamukkale University, Medical Faculty, Department of Medical Biology, 20020 Denizli, Turkey. [Satiroglu-Tufan, Lale] Pamukkale University, Medical Faculty, Department of Medical Biology, 20020 Denizli, Turkey. [Kelten, Canan Esra] Pamukkale University, Faculty of Medicine, Department of Pathology, 20020 Denizli, Turkey. [Zencir, Sevil] Pamukkale University, Medical Faculty, Department of Medical Biology, 20020 Denizli, Turkey. [Dodurga, Yavuz] Pamukkale University, Medical Faculty, Department of Medical Biology, 20020 Denizli, Turkey. [Bagci, Huseyin] Pamukkale University, Medical Faculty, Department of Medical Biology, 20020 Denizli, Turkey. [Duzcan, Ender Suleyman] Pamukkale University, Faculty of Medicine, Department of Pathology, 20020 Denizli, Turkey. RP Duzcan, ES (reprint author), Pamukkale University, Faculty of Medicine, Department of Pathology, 20020 Denizli, Turkey. EM eduzcan@yahoo.com CR Latif Z, Watters AD, Dunn I et al, 2004, HER2/neu gene amplification and protein overexpression in G3 pT2 transitional cell carcinoma of the bladder: a role for anti-HER2 therapy? Eur J Cancer 40:56–63 Morrison C, Zanagnolo V, Ramirez N et al, 2006, HER-2 is an independent prognostic factor in endometrial cancer: association with outcome in a large cohort of surgically staged patients. J Clin Oncol 24:2376–2385 Ross JS, Fletcher JA, Linette GP et al, 2003, The Her-2/neu gene and protein in breast cancer 2003: biomarker and target of therapy. Oncologist 8:307–325 Schraml P, Kononen J, Bubendorf L et al, 1999, Tissue microarrays for gene amplification surveys in many different tumor types. Clin Cancer Res 5:1966–1975 Kruger S, Weitsch G, Buttner H et al, 2002, Overexpression of cerbB- 2 oncoprotein in muscle-invasive bladder carcinoma: relationship with gene amplification, clinicopathological parameters and prognostic outcome. Int J Oncol 21:981–987 Underwood M, Barlett J, Reeves J et al, 1995, c-erbB-2 gene amplification: a molecular marker in recurrent bladder tumors? Cancer Res 55:2422–2430 Bartlett J, Mallon E, Cooke T, 2003, The clinical evaluation of HER-2 status: which test to use? J Pathol 199:411–417 Langner C, Gross C, Rehak P et al, 2005, HER2 protein overexpression and gene amplification in upper urinary transitional cell carcinoma: systematic analysis applying tissue microarray technique. Urology 65:176–180 Bast RCJ, Ravdin P, Hayes DF et al, 2001, 2000 update of recommendations for the use of tumor markers in breast and colorectal cancer: clinical practice guidelines of the American Society of Clinical Oncology. J Clin Oncol 19:1865–1878 Lamy PJ, Nanni I, Fina F et al, 2006, Reliability and discriminant validity of HER2 gene amplification and chromosome 17 aneusomy analysis by real-time PCR in primary breast cancer. Int J Biol Markers 21:20–29 Edwards RG, Lara PN, Folkins AK et al, 2002, Does HER2/neu expression provide prognostic information in patients with advanced urothelial carcinoma? Cancer 95:1009–1015 Kruger S, Weitsch G, Buttner H et al, 2002, HER2 overexpression in muscle-invasive urothelial carcinoma of the bladder: prognostic implications. Int J Cancer 102:514–518 Latif Z, Watters AD, Dunn I et al, 2003, HER2/neu overexpression in the development of muscle-invasive transitional cell carcinoma of the bladder. Br J Cancer 89:1305–1309 Liminez RE, Hussain M, Bianco FJ Jr, et al, 2001, HER-2/neu overexpression in muscle-invasive urothelial carcinoma of the bladder: prognostic significance and comparative analysis in primary and metastatic tumors. Clin Cancer Res 7:2440–2447 Simon R, Atefy R, Wagner U et al, 2003, HER-2 and TOP2A coamplification in urinary bladder cancer. Int J Cancer 107:764–772 Sauter G, Moch H, Moore D et al, 1993, Heterogeneity of erbB-2 gene amplification in bladder cancer. Cancer Res 53(Suppl 10): 2199–2203 Gjerdrum LM, Lielpetere I, Rasmussen LM et al, 2001, Laserassisted microdissection of membrane-mounted paraffin sections for polymerase chain reaction analysis: identification of cell populations using immunohistochemistry and in situ hybridization. J Mol Diagn 3:105–110 Persons D, Tubbs R, Cooley LD et al, 2006, HER2 fluorescence in situ hybridization. Arch Pathol Lab Med 130:325–331 Bose S, Mohammed M, Shintaku P et al, 2001, Her-2/neu gene amplification in low to moderately expressing breast cancers: possible role of chromosome 17/Her-2/neu polysomy. Breast J 7:337–344 Wang S, Hossein Saboorian M, Frenkel EP et al, 2002, Aneusomy 17 in breast cancer: its role in HER-2/neu protein expression and implication for clinical assessment of HER-2/neu status. Mod Pathol 15:137–145 Gallucci M, Guadagni F, Marzano R et al, 2005, Status of the p53, p16, RB1, and HER-2 genes and chromosomes 3, 7, 9, and 17 in advanced bladder cancer: correlation with adjacent mucosa and pathological parameters. J Clin Pathol 58:367–371 Ohta JI, Miyoshi Y, Uemura H et al, 2001, Fluorescence in situ hybridization evaluation of c-erbB-2 gene amplification and chromosomal anomalies in bladder cancer. Clin Cancer Res 7: 2463–2467 Hovey RM, Chu L, Balazs M et al, 1998, Genetic alterations in primary bladder cancers and their metastases. Cancer Res 58:3555–3560 Kelly ED, Yoder BJ, Stoler M et al, 2005, The influence of polysomy 17 on HER2 gene and protein expression in adenocarcinoma of the breast. Am J Surg Pathol 29:1221–1227 Miyamoto H, Kubota Y, Noguchi S et al, 2000, c-erbB-2 gene amplification as a prognostic marker in human bladder cancer. Urology 55:679–683 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 261 EP 266 DI 10.1007/s12253-008-9027-y PG 6 ER PT J AU Lyronis, DI Baritaki, S Bizakis, I Krambovitis, E Spandidos, AD AF Lyronis, D Ioannis Baritaki, Stavroula Bizakis, Ioannis Krambovitis, Elias Spandidos, A Demetrios TI K-ras Mutation, HPV Infection and Smoking or Alcohol Abuse Positively Correlate with Esophageal Squamous Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal squamous cell carcinoma; Human Papilloma Virus; B-raf; K-ras; N-ras ID Esophageal squamous cell carcinoma; Human Papilloma Virus; B-raf; K-ras; N-ras AB The Ras/Raf/MEK/ERK (MAPK) signal transduction cascade is an important mediator of a number of cellular fates including growth, survival and apoptosis. The aim of this study was to determine the incidence of B-raf, Kirsten-ras (K-ras) and Neuroblastoma-ras (N-ras) gene mutations in esophageal squamous cell carcinoma (ESCC) in the Greek population. DNA was extracted from 30 ESCC and 32 normal esophageal specimens and screened for V600E B-raf, and K-ras/N-ras codon 12 mutations, by PCR-RFLP based analysis. Among the genes tested, only the heterozygous K-ras mutation was detected in 5 out of the 30 ESCC specimens (16%), whereas no mutation was found in the normal esophageal tissue (P<0.022). The normal samples were screened negative for N-ras and V600E B-raf mutations. The increased risk of esophageal cancer was correlated with tobacco use (OR=3.5, P<0.023) and alcohol abuse (OR=7.22, P<0.001), accompanied with the high incidence of the k-ras codon 12 mutation (22%, OR=1.77 and 21%, OR=1.52), respectively. A similar positive association was seen in human papilloma virus (HPV)-infected patients (OR=5.66, P<0.003). Our overall findings demonstrate that the mutational activation of the K-ras gene, HPV infection and tobacco or alcohol abuse, can be considered independently or in combination as high risk factors for ESCC development. C1 [Lyronis, D Ioannis] University of Crete, Medical School, Department of Virology, 711 10 Heraklion, Crete, Greece. [Baritaki, Stavroula] University of Crete, Medical School, Department of Virology, 711 10 Heraklion, Crete, Greece. [Bizakis, Ioannis] University Hospital of Crete, Department of Otolaryngology, 712 01 Heraklion, Crete, Greece. [Krambovitis, Elias] University of Thessaly, Department of Veterinary MedicineKarditsa, Greece. [Spandidos, A Demetrios] University of Crete, Medical School, Department of Virology, 711 10 Heraklion, Crete, Greece. RP Spandidos, AD (reprint author), University of Crete, Medical School, Department of Virology, 711 10 Heraklion, Greece. EM spandidos@spandidos.gr CR Arber N, Shapira I, Ratan J et al, 2000, Activation of c-K-ras mutations in human gastrointestinal tumors. Gastroenterology 118:1045–1050 Arora S, Mathew R, Mathur M et al, 2001, Alterations in MDM2 in esophageal squamous cell carcinoma: relationship with p53 status. Pathol Oncol Res 7:203–208 Bosetti C, Gallus S, Garavello W et al, 2006, Smoking cessation and the risk of oesophageal cancer: an overview of published studies. Oral Oncol 42:957–964 Fenoglio-Preiser CM, 1999, Gastrointestinal pathology. An atlas and text, 2nd edn. Lippincott-Raven, Philadelphia, pp 102–103 Chetty R, Simelane S, 1999, p53 and cyclin A protein expression in squamous carcinoma of the oesophagus. Pathol Oncol Res 5:193–196 Cohen Y, Xing M, Mambo E et al, 2003, BRAF mutation in papillary thyroid carcinoma. J Natl Cancer Inst 95:625–627 Davies H, Bignell GR, Cox C et al, 2002, Mutations of the BRAF gene in human cancer. Nature 417:949–954 Ellis A, Field JK, Field EA et al, 1994, Tylosis associated with carcinoma of the oesophagus and oral leukoplakia in a large Liverpool family—a review of six generations. Eur J Cancer B Oral Oncol 30:102–112 Enzinger PC, Mayer RJ, 2003, Esophageal cancer. N Engl J Med 349:2241–2252 Fransen K, Klintenas M, Osterstrom A et al, 2004, Mutation analysis of the BRAF, ARAF and RAF-1 genes in human colorectal adenocarcinomas. Carcinogenesis 25:527–533 Garnett MJ, Marais R, 2004, Guilty as charged: B-RAF is a human oncogene. Cancer Cell 6:313–319 Kim IJ, Park JH, Kang HC et al, 2003, Mutational analysis of BRAF and K-ras in gastric cancers: absence of BRAF mutations in gastric cancers. Hum Genet 114:118–120 Lam AK, 2000, Molecular biology of esophageal squamous cell carcinoma. Crit Rev Oncol Hematol 33:71–90 Lee SH, Lee JW, Soung YH et al, 2003, BRAF and KRAS mutations in stomach cancer. Oncogene 22:6942–6945 Lord RV, O'Grady R, Sheehan C et al, 2000, K-ras codon 12 mutations in Barrett’s oesophagus and adenocarcinomas of the oesophagus and oesophagogastric junction. J Gastroenterol Hepatol 15:730–736 Lyronis ID, Baritaki S, Bizakis I et al, 2005, Evaluation of the prevalence of human papillomavirus and Epstein–Barr virus in esophageal squamous cell carcinomas. Int J Biol Markers 20:5–10 Mandard AM, Hainaut P, Hollstein M, 2000, Genetic steps in the development of squamous cell carcinoma of the esophagus. Mutat Res 462:335–342 Mercer KE, Pritchard CA, 2003, Raf proteins and cancer: B-Raf is identified as a mutational target. Biochim Biophys Acta 1653, 1):25–40, Jun 5 Ramachandran S, Ramadas K, Hariharan R et al, 2006, Single nucleotide polymorphisms of DNA repair genes XRC1 and XPD and its molecular mapping in Indian oral cancer. Oral Oncol 42:350–362 Scully C, Field JK, Tanzawa H, 2000, Genetic aberrations in oral or head and neck squamous cell carcinoma 2: chromosomal aberrations. Oral Oncol 36:311–327 Sommerer F, Vieth M, Markwarth A et al, 2004, Mutations of BRAF and KRAS2 in the development of Barrett’s adenocarcinoma. Oncogene 23:554–558 Spandidos DA, Sourvinos G, Tsatsanis C et al, 2002, Normal ras genes: their onco-suppressor and pro-apoptotic functions, review). Int J Oncol 21:237–241 Syrjanen KJ, 2002, HPV infections and oesophageal cancer. J Clin Pathol 55:721–728 Talamini G, Capelli P, Zamboni G et al, 2000, Alcohol, smoking and papillomavirus infection as risk factors for esophageal squamous-cell papilloma and esophageal squamous-cell carcinoma in Italy. Int J Cancer 86:874–878 Tavani A, Bertuzzi M, Talamini R et al, 2003, Coffee and tea intake and risk of oral, pharyngeal and esophageal cancer. Oral Oncol 39:695–700 WangWKP, Schwartz M, Malon G et al, 1994, PCR amplification of 40 year-old paraffin-embedded tumour tissues: comparison of four different DNA extraction and purificationmethods. Int J Oncol 5:0–4 Weber A, Langhanki L, Sommerer F et al, 2003, Mutations of the BRAF gene in squamous cell carcinoma of the head and neck. Oncogene 22:4757–4759 Wu M, Semba S, Oue N et al, 2004, BRAF/K-ras mutation, microsatellite instability, and promoter hypermethylation of hMLH1/MGMT in human gastric carcinomas. Gastric Cancer 7:246–253 Xing M, 2005, BRAF mutation in thyroid cancer. Endocr-Relat Cancer 12:245–262 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 267 EP 273 DI 10.1007/s12253-008-9032-1 PG 7 ER PT J AU Szegedi, I Katona, K Horvath, A Molnar, A Aradi, J Kiss, Cs AF Szegedi, Istvan Katona, Klara Horvath, Andras Molnar, Anna Aradi, Janos Kiss, Csongor TI Bcl-2 Antisense Oligonucleotide Inhibits the Proliferation of Childhood Leukemia/lymphoma Cells of the B-cell Lineage SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BCL-2; Antisense oligonucleotide; Childhood; Leukemia; Lymphoma ID BCL-2; Antisense oligonucleotide; Childhood; Leukemia; Lymphoma AB An 18-mer phosphorothioate bcl-2 atisense oligonucleotide (ASO) inhibited colony formation of three B-cell leukemia/lymphoma cell lines in a dose dependent manner in the range of 0.125–0.5 μmol/l. The srcambled cogener had no detectable effect. A decrease in BCL-2 protein and apoptotic DNA fragmentation was detected in the studied cell lines and primary blast cells of two children with acute lymphoblastic leukemia. Neither BCL-2 protein level, nor DNA integrity was affected by the scrambled control indicating the specific effect ASO. As far as we know, this is the first report on the effects of bcl-2 ASO on childhood leukemia/lymphoma cell samples. C1 [Szegedi, Istvan] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98 Nagyerdei Circle, 4012 Debrecen, Hungary. [Katona, Klara] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Horvath, Andras] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Molnar, Anna] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98 Nagyerdei Circle, 4012 Debrecen, Hungary. [Aradi, Janos] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Kiss, Csongor] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98 Nagyerdei Circle, 4012 Debrecen, Hungary. RP Kiss, Cs (reprint author), Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 4012 Debrecen, Hungary. EM kisscs@dote.hu CR Dean NM, Bennett CF, 2003, Antisense oligonucleotide-based therapeutics for cancer. Oncogene 22:9087–9096 Reed JC, Stein C, Subasinghe C et al, 1990, Antisense-mediated inhibition of BCL2 protooncogene expression and leukemic cell growth and survival:comparison of phosphodiester and phosphrothioate olygonucleotides. Cancer Res 50:6565–6570 Tsujimoto Y, Cossman J, Jaffe E et al, 1985, Involvement of the bcl-2 gene in human follicular lymphoma. Science 228:1440–1443 Reed JC, 1995, Regulation of apoptosis by bcl-2 family proteins and its role in cancer and chemoresistance. Curr Opin Oncol 7:541–546 Adis International Limited, 2007, Oblimersen: Augnerosen, Bcl-2 antisense oligonucleitide- Genta, G3139, GC 3139, Oblimersen sodium. Drugs in R&D 5:321–334 Webb A, Cinnungham D, Cotter F et al, 1997, BCL-2 antisense therapy in patients with non-Hodgkin lymphoma. Lancet 349:1137–1141 Terhorst C, Parham P, Mann DL et al, 1976, Structure of HLA Antigens: amino-acid and carbohydrate compositions and NH2- terminal sequences of four antigen preparations. Proc Natl Acad Sci USA 73:910–914 Cohen JH, Revillard JP, Magaud JP et al, 1987, B-cell maturation stages of Burkitt’s lymphoma cell lines according to Epstein-Barr virus status and type of chromosome translocation. J Natl Cancer Inst 78:235–242 Renne R, Zhong W, Herndier B et al, 1996, Lytic growth of Kaposi’sarcoma-associated herpesvirus, human herpesvirus 8, in culture. Nat Med 2:342–346 Iyer RP, Philips LR, Egan Wet al, 1990, The automated synthesis of sulfur-containing oligodeoxyribonucleotides using 3H-benzodithiol-3- one, 1,1-dioxide as a sulfur-transfer reagent. JOrg Chem 55:4693–4699 Horvath A, Aradi J, 2005, Advantages of sodium perchlorate solution as mobile phase for purification of synthetic oligonucleotides by anion exchange chromatography. Analytical Biochemistry 338:341–343 Benko I, Kovacs P, Szegedi I et al, 2001, Effect of myelopoietic and pleiotropic cytokines on colony formation by blast cells of children with acute lymphoblastic leukemia. Naunyn Schmiedebergs Arch Pharmacol 363:499–508 Zwaan CM, Kaspers GJL, Pieters R et al, 2000, Cellular drug resistance profile in childhood acute leukemia: difference between FAB types and comparison with acute lymphoid leukemia. Blood 96:2879–2886 Kennedy DO, Kijima A,Yano Y et al, 2001, Growth inhibitory effect of green tea extract in Erlich ascites tumor cells involves cytochrome c release and caspase activation. Cancer Lett 166:9- 15. Liu X, Kim CN, Yang J et al, 1996, Induction of apoptotic program in cell free extracts: requirement for dATP and cytochrome c. Cell 86:147–157 Campos L, Sabido O, Rouault JP et al, 1994, Effects of BCL-2 antisense oligonucleotides on in vitro proliferation an survival of normal marrow progenitors and leukemic cells. Blood 84:595– 600 Uchida T, Gao JP, Wang C et al, 2001, Antitumor effect of bcl-2 antisense phosphorothioate oligonucleotides on human renal-cell carcinima cells in vitro an in mice. Mol Urol 5:71–78 Lu P, Chen J, 1999, Apoptosis on human gastric cancer cells induced by bcl-2 intisense oligodeoxynucleotides. Zhonghua Yi Xue Za Zhi 21:253–255 Jahrsdorfer B, Jox R, Muhlenhoff L et al, 2002, Modulation of malignant B cell activation and apoptosis by bcl-2 antisense ODN and immunostimulatory CpG ODN. J Leukoc Biol 72:83–92 Wang FX, Dong ZR, Liu ZL et al, 2003, Effects of bcl-2 antisense oligodeoxynucleotide on proliferation and apoptosis of Raji cells. Zhonghua Xue Ye Xue Za Zhi 24:71–73 Lin Y, Lu L, Chen Z, 2000, The toxic effects of high dose Bcl-2 antisense phosphorothioate oligonucleotides incubation on cell line. ZhonghuaYi Xue Za Zhi 80:694–697 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 275 EP 279 DI 10.1007/s12253-008-9076-2 PG 5 ER PT J AU Csomor, J Kaszas, I Kollar, B Pajor, L Egyhazi, Zs Fekete, S Egyed, M Timar, B AF Csomor, Judit Kaszas, Ilona Kollar, Balazs Pajor, Laszlo Egyhazi, Zsolt Fekete, Sandor Egyed, Miklos Timar, Botond TI Prolonged Survival Using Anti-CD20 Combined Chemotherapy in Primary Prostatic Intravascular Large B-cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DLBCL; Intravascular large B-cell lymphoma; Prostate; Rituximab ID DLBCL; Intravascular large B-cell lymphoma; Prostate; Rituximab AB Here we report a case of a 73-year-old man with primary intravascular large B-cell lymphoma localized to the prostate. Total prostatectomy was performed due to a benign adenoma suggested by ultrasonography. The diagnosis of IVLBL was obtained incidentally from the prostatectomy specimen. Eight months after the initial R-CHOP chemotherapy a relapse was detected in the left inguinal lymph node, where histologic examination revealed common diffuse large B-cell lymphoma with minimal intravascular component. The second complete remission was achieved by R-IEV therapy. Five months later a second relapse occurred and the patient died in the widespread disease and pneumonia. Primary prostate IVLBL is extremely uncommon; to date only four cases have been described. This is a well documented case, where we also confirmed that the initial primary IVLBL and the secondary lymph node involvement are clonally related. Successful treatment depends on early diagnosis of IVLBL, aggressive chemotherapy and the fact that IVLBL should be considered as a generalized disease in spite of negative staging results. C1 [Csomor, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Kaszas, Ilona] Szt. Margit Hospital, Department of PathologyBudapest, Hungary. [Kollar, Balazs] Kaposi Mor Teaching Hospital, Department of OncohematologyKaposvar, Hungary. [Pajor, Laszlo] University of Pecs, Department of PathologyPecs, Hungary. [Egyhazi, Zsolt] Kaposi Mor Teaching Hospital, Department of OncohematologyKaposvar, Hungary. [Fekete, Sandor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Egyed, Miklos] Kaposi Mor Teaching Hospital, Department of OncohematologyKaposvar, Hungary. [Timar, Botond] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. RP Csomor, J (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM csomor@korb1.sote.hu CR Jaffe ES, Harris NL, Stein H, Vardiman JW, Eds,, 2001, World Health Organization classification of tumours. Pathology and genetics of tumours of haematopoietic and lymphoid tissues. IARC, Lyon Ferreri AJ, Campo E, Seymour JF, Willemze R, Ilariucci F, Ambrosetti A et al, 2004, Intravascular lymphoma: clinical presentation, natural history, management and prognostic factors in a series of 38 cases, with special emphasis on the ‘cutaneous variant’. Br J Haematol 127(2):173–183 Bostwick DG, Iczkowski KA, Amin MB, Discigil G, Osborne B, 1998, Malignant lymphoma involving the prostate: report of 62 cases. Cancer 83(4):732–738 Chu PG, Huang Q, Weiss LM, 2005, Incidental and concurrent malignant lymphomas discovered at the time of prostatectomy and prostate biopsy: a study of 29 cases. Am J Surg Pathol 29(5):693– 699 Yin XR, Liu H, Chen HQ, 2005, Intravascular lymphomatosis of the prostate gland. Zhonghua Bing Li Xue Za Zhi 34(3):189–190, Chinese Alfaro J, Espinoza A, Manriquez M, Moyano L, Gonzalez N, Larrondo M, Figueroa G, 2004, Intravascular lymphoma treated with anti CD20 monoclonal antibodies. Report of one case. Rev Med Chil 132(11):1403–1406, Spanish Quintini G, Barbera V, Franco V, Florena AM, Spadola V, Mariani G, 2003, Uncommon presentations of non-Hodgkin’s lymphoma: case 1. Intravascular large B-cell lymphoma: diagnosis on prostate biopsy. J Clin Oncol 21(3):564–565 Chim CS, Loong F, 2002, Intravascular lymphomatosis of the prostate gland. Br J Haematol 119(1):2 Ponzoni M, Arrigoni G, Gould VE, Del Curto B, Maggioni M, Scapinello A et al, 2000, Lack of CD 29, beta1 integrin, and CD 54, ICAM-1, adhesion molecules in intravascular lymphomatosis. Hum Pathol 31(2):220–226 Kishi Y, Kami M, Kusumi E, Mineyama T, Kato D, Hamaki T et al, 2004, Prostatic acid phosphatase: a possible diagnostic marker of intravascular large B-cell lymphoma. Haematologica 89, 5):ECR13 Odawara J, Asada N, Aoki T, Yamakura M, Takeuchi M, Ohuchi T et al, 2007, 18F-Fluorodeoxyglucose positron emission tomography for evaluation of intravascular large B-cell lymphoma. Br J Haematol 136(5):684 BouzaniM, Karmiris T, Rontogianni D, Delimpassi S, Apostolidis J, MpakiriMet al, 2006, Disseminated intravascular B-cell lymphoma: clinicopathological features and outcome of three cases treated with anthracycline-based immunochemotherapy. Oncologist 11(8):923– 928 Ohno T, Sakamoto T, Mizumoto C, Miyoshi T, Ueda M, Takeoka T et al., 2006, Leukemic and meningeal relapse of CD5+ intravascular large B-cell lymphoma with down-modulation of CD20 after rituximab therapy. Int J Hematol 84(1):74–78 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 281 EP 284 DI 10.1007/s12253-008-9051-y PG 4 ER PT J AU Markasz, L Hajas, Gy Kiss, A Lontay, B Rajnavolgyi, Erdodi, F Olah, AF Markasz, Laszlo Hajas, Gyorgy Kiss, Andrea Lontay, Beata Rajnavolgyi, Eva Erdodi, Ferenc Olah, Eva TI Granulocyte Colony Stimulating Factor Increases Drug Resistance of Leukaemic Blast Cells to Daunorubicin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute leukaemia; Daunorubicin; Drug sensitivity; G-CSF; G-CSF-R ID Acute leukaemia; Daunorubicin; Drug sensitivity; G-CSF; G-CSF-R AB Acute leukaemia is known as the most common cancer in childhood. Febrile neutropenia is a common serious side effect of the cytostatic treatment of malignancies. The clinical use of Granulocyte Colony Stimulating Factor (G-CSF) has become widespread to minimize chemotherapy-induced myelosuppression and febrile neutropenia in childhood solid tumors, acute lymphoid leukaemia (ALL) and in several trials with AML. In case of ALL this seems to be reasonable because, due to the absence of G-CSF receptor (G-CSFR) on the surface of normal lymphoid cells, G-CSF does not have any influence on the pathways of proliferation and differentiation of lymphoid lineage cells. It has been suggested, however, that ALL blasts with B or T cell surface antigens as well as biphenotypic leukaemia cells express G-CSFR, and they are able to respond to exogenously added G-CSF with proliferation. In this study we investigated how G-CSF might influence the sensitivity of leukemic cells to daunorubicin induced cell death using MTT assay, flow cytometry and Western blot analysis. After pretreatment of KG-1 leukaemic cells with G-CSF a moderate increase in the resistance of these cells to daunorubicin could be observed. These results draw attention to the risk of G-CSF application as an adjuvant therapy of childhood ALL. In addition, adjuvant treatment of AML patients with G-CSF in order to prevent neutropenia, or its use in priming regimens might result resistance to daunorubicin. C1 [Markasz, Laszlo] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Hajas, Gyorgy] University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Kiss, Andrea] University of Debrecen, Faculty of Medicine, Department of Medical Chemistry, Egyetem ter 1, 4032 Debrecen, Hungary. [Lontay, Beata] University of Debrecen, Faculty of Medicine, Department of Medical Chemistry, Egyetem ter 1, 4032 Debrecen, Hungary. [Rajnavolgyi, Eva] University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Erdodi, Ferenc] University of Debrecen, Faculty of Medicine, Department of Medical Chemistry, Egyetem ter 1, 4032 Debrecen, Hungary. [Olah, Eva] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, Nagyerdei krt. 98, 4032 Debrecen, Hungary. RP Olah, (reprint author), Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 4032 Debrecen, Hungary. EM markaszl@freemail.hu CR Licht JD, Sternberg DW, 2005, The molecular pathology of acute myeloid leukemia. Hematology 2005:137–142 Kager L, Evans WE, 2006, Pharmacogenomics of acute lymphoblastic leukemia. Curr Opin Hematol 13(4):260–265 Woods WG, 2006, Curing childhood acute myeloid leukemia, AML, at the half-way point: promises to keep and miles to go before we sleep. Pediatr Blood Cancer 46(5):565–569 Sasse EC, Sasse AD, Brandalise S et al, 2005, Colony stimulating factors for prevention of myelosupressive therapy induced febrile neutropenia in children with acute lymphoblastic leukaemia. Cochrane Database Syst Rev 3:CD004139 Timmer-Bonte JN, Tjan-Heijnen VC, 2006, Febrile neutropenia: highlighting the role of prophylactic antibiotics and granulocyte colony-stimulating factor during standard dose chemotherapy for solid tumors. Anticancer Drugs 17(8):881–889 Heath JA, Steinherz PG, Altman A et al, 2003, Human granulocyte colony-stimulating factor in children with high-risk acute lymphoblastic leukemia: a Children’s Cancer Group Study. J Clin Oncol 21(8):1612–1617 Lehrnbecher T, Zimmermann M, Reinhardt D et al, 2007, Prophylactic human granulocyte colony-stimulating factor after induction therapy in pediatric acute myeloid leukemia. Blood 109, 3):936–943 Sachs L, 1996, The control of hematopoiesis and leukemia: from basic biology to the clinic. Proc Natl Acad Sci U S A 93, 10):4742–4749 Lopez AF, Williamson DJ, Gamble JR et al, 1986, Recombinant human granulocyte-macrophage colony-stimulating factor stimulates in vitro mature human neutrophil and eosinophil function, surface receptor expression, and survival. J Clin Invest 78, 5):1220–1228 Dempke W, Von Poblozki A, Grothey A et al, 2000, Human hematopoietic growth factors: old lessons and new perspectives. Anticancer Res 20(6D):5155–5164 Frampton JE, Keating GM, 2005, Spotlight on pegfilgrastim in chemotherapy-induced neutropenia. BioDrugs 19(6):405–407 Ravandi F, 2006, Role of cytokines in the treatment of acute leukemias: a review. Leukemia 20(4):563–571 Asano S, 1991, Human granulocyte colony-stimulating factor: its basic aspects and clinical applications. Am J Pediatr Hematol Oncol 13(4):400–413 Nicholson SE, Novak U, Ziegler SF et al, 1995, Distinct regions of the granulocyte colony-stimulating factor receptor are required for tyrosine phosphorylation of the signaling molecules JAK2, Stat3, and p42, p44MAPK. Blood 86(10):3698–3704 Fukunaga R, Seto Y, Mizushima S et al, 1990, Three different mRNAs encoding human granulocyte colony-stimulating factor receptor. Proc Natl Acad Sci U S A 87(22):8702–8706 Glaspy JA, Golde DW, 1992, Granulocyte colony-stimulating factor, G-CSF): preclinical and clinical studies. Semin Oncol 19, 4):386–394 Dombret H, 1996, Granulocytic colony-stimulating factors in the management of patients with acute myeloid leukemia. Hematol Cell Ther 38(3):231–240 Becker PS, 2004, Growth factor priming in therapy of acute myelogenous leukemia. Curr Hematol Rep 3(6):413–418 Hubeek I, Litvinova E, Peters GJ et al, 2004, The effect of G-CSF on the in vitro cytotoxicity of cytarabine and fludarabine in the FLAG combination in pediatric acute myeloid leukemia. Int J Oncol 25(6):1823–1829 Buchner T, Hiddemann W, Wormann B et al, 1997, Hematopoietic growth factors in acute myeloid leukemia: supportive and priming effects. Semin Oncol 24(1):124–131 Nicola NA, Metcalf D, 1985, Binding of 125I-labeled granulocyte colony-stimulating factor to normal murine hemopoietic cells. J Cell Physiol 124(2):313–321 Liu WM, Powles T, Shamash J et al, 2004, Effect of haemopoietic growth factors on cancer cell lines and their role in chemosensitivity. Oncogene 23(4):981–990 Handa A, Kashimura T, Takeuchi S et al, 2000, Expression of functional granulocyte colony-stimulating factor receptors on human B-lymphocytic leukemia cells. Ann Hematol 79(3):127– 131 Kita K, Nishii K, Ohishi K et al, 1993, Frequent gene expression of granulocyte colony-stimulating factor, G-CSF, receptor in CD7 + surface CD3− acute lymphoblastic leukaemia. Leukemia 7, 8):1184–1190 Shimoda K, Okamura S, Harada N et al, 1992, Detection of the granulocyte colony-stimulating factor receptor using biotinylated granulocyte colony-stimulating factor: presence of granulocyte colony-stimulating factor receptor on CD34- positive hematopoietic progenitor cells. Res Exp Med, Berl, 192(4):245–255 Inukai T, Sugita K, Iijima K et al, 1998, Leukemic cells with 11q23 translocations express granulocyte colony-stimulating factor, G-CSF, receptor and their proliferation is stimulated with G-CSF. Leukemia 12(3):382–389 Benko I, Kovacs P, Szegedi I et al, 2001, Effect of myelopoietic and pleiotropic cytokines on colony formation by blast cells of children with acute lymphoblastic leukemia. Naunyn Schmiedebergs Arch Pharmacol 363(5):499–508 Higashigawa M, Kuwabara H, Cao DC et al, 1996, Heterogeneous effects of G-CSF and GM-CSF on cell growth and ara-C cytotoxicity in childhood leukemias which express myeloid markers. Leuk Lymphoma 22(3–4):279–285 de Lau WB, Hurenkamp J, Berendes P et al, 1998, The gene encoding the granulocyte colony-stimulating factor receptor is a target for deregulation in pre-B ALL by the t(1;19)-specific oncoprotein E2A-Pbx1. Oncogene 17(4):503–510 Williams DL, Look AT, Melvin SL et al, 1984, New chromosomal translocations correlate with specific immunophenotypes of childhood acute lymphoblastic leukemia. Cell 36(1):101–109 Matsushita K, Arima N, Ohtsubo H et al, 1997, Granulocytecolony stimulating factor-induced proliferation of primary adult Tcell leukaemia cells. Br J Haematol 96(4):715–723 Oh EJ, Kahng J, Kim Y et al, 2003, Expression of functional markers in acute lymphoblastic leukemia. Leuk Res 27(10):903– 908 Koeffler HP, Golde DW, 1980, Human myeloid leukemia cell lines: a review. Blood 56(3):344–350 Minotti G, Menna P, Salvatorelli E et al, 2004, Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev 56(2):185–229 Ravindranath Y, 2003, Recent advances in pediatric acute lymphoblastic and myeloid leukemia. Curr Opin Oncol 15, 1):23–35 Tallman MS, Gilliland DG, Rowe JM, 2005, Drug therapy for acute myeloid leukemia. Blood 106(4):1154–1163 Hajas G, Zsiros E, Laszlo T et al, 2004, New phenotypic, functional and electrophysiological characteristics of KG-1 cells. Immunol Lett 92(1–2):97–106 Laemmli UK, 1970, Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227(5259):680– 685 Muranyi A, Erdodi F, Ito M et al, 1998, Identification and localization of myosin phosphatase in human platelets. Biochem J 330(Pt 1):225–231 Miyauchi J, Kelleher CA, Yang YC et al, 1987, The effects of three recombinant growth factors, IL-3, GM-CSF, and G-CSF, on the blast cells of acute myeloblastic leukemia maintained in shortterm suspension culture. Blood 70(3):657–663 Delwel R, Salem M, Pellens C et al, 1988, Growth regulation of human acute myeloid leukemia: effects of five recombinant hematopoietic factors in a serum-free culture system. Blood 72, 6):1944–1949 Vellenga E, Ostapovicz D, O’Rourke B et al, 1987, Effects of recombinant IL-3, GM-CSF, and G-CSF on proliferation of leukemic clonogenic cells in short-term and long-term cultures. Leukemia 1(8):584–589 Litvinova E, Gurova K, Chimishkian K et al, 1999, Effects of CSFS and their combinations with chemotherapeutic agents, CH, on leukemic blasts, LB, in children, MTT-assay). Adv Exp Med Biol 457:585–592 van der Kolk DM, de Vries EG, Muller M et al, 2002, The role of drug efflux pumps in acute myeloid leukemia. Leuk Lymphoma 43(4):685–701 Miranda MB, Xu H, Torchia JA et al, 2005, Cytokine-induced myeloid differentiation is dependent on activation of the MEK/ ERK pathway. Leuk Res 29(11):1293–1306 Prieto J, Eklund A, Patarroyo M, 1994, Regulated expression of integrins and other adhesion molecules during differentiation of monocytes into macrophages. Cell Immunol 156(1):191–211 Kishimoto TK, Larson RS, Corbi AL et al, 1989, The leukocyte integrins. Adv Immunol 46:149–182 Larson RS, Springer TA, 1990, Structure and function of leukocyte integrins. Immunol Rev 114:181–217 Inghirami G, Grignani F, Sternas L et al, 1990, Down-regulation of LFA-1 adhesion receptors by C-myc oncogene in human B lymphoblastoid cells. Science 250(4981):682–686 Bashey A, Healy L, Marshall CJ, 1994, Proliferative but not nonproliferative responses to granulocyte colony-stimulating factor are associated with rapid activation of the p21ras/MAP kinase signalling pathway. Blood 83(4):949–957 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 285 EP 292 DI 10.1007/s12253-008-9057-5 PG 8 ER PT J AU Matusan-Ilijas, K Behrem, S Jonjic, N Zarkovic, K Lucin, K AF Matusan-Ilijas, Koviljka Behrem, Senija Jonjic, Nives Zarkovic, Kamelija Lucin, Ksenija TI Osteopontin Expression Correlates with Angiogenesis and Survival in Malignant Astrocytoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; Immunohistochemistry; Osteopontin; Pathological angiogenesis; Prognosis ID Glioma; Immunohistochemistry; Osteopontin; Pathological angiogenesis; Prognosis AB Osteopontin (OPN) is a phosphorylated glycoprotein with diverse functions including angiogenesis, cancer development, invasion and metastasis. The aim of the study was to analyze the expression of OPN in human astrocytomas and to correlate it with angiogenesis and patients’ outcome. Seventy-six human astrocytomas including eight pilocytic astrocytomas (grade I), 10 diffuse astrocytomas (grade II), 8 anaplastic astrocytomas (grade III) and 50 glioblastomas (grade IV) were immunohistochemically stained for OPN protein. The distribution of OPN staining (cytoplasmic and/or interstitial)was assessed and compared tomicrovessel number and patients’ survival. In normal brain tissue some glial and neuronal cells showed weak cytoplasmic staining, while interstitium was negative. Astrocytomas were heterogeneous regarding the OPN expression. High cytoplasmic OPN expression in glioblastomas was associated with poor patients’ survival (p=0.012). Also, we found the association of interstitial OPN expression and angiogenesis (p=0.033), i.e. the number of newly formed blood vessels was higher in tumors showing high interstitial OPN expression. Our results indicate the overexpression of OPN protein in astrocytoma cells and suggest the role of OPN in astrocytoma progression and angiogenesis. C1 [Matusan-Ilijas, Koviljka] Rijeka University School of Medicine, Department of Pathology, Brace Branchetta 20, 51 000 Rijeka, Croatia. [Behrem, Senija] Rijeka University School of Medicine, Department of Pathology, Brace Branchetta 20, 51 000 Rijeka, Croatia. [Jonjic, Nives] Rijeka University School of Medicine, Department of Pathology, Brace Branchetta 20, 51 000 Rijeka, Croatia. [Zarkovic, Kamelija] University Hospital Center Zagreb, Department of Pathology, Kispaticeva 12, 10 000 Zagreb, Croatia. [Lucin, Ksenija] Rijeka University School of Medicine, Department of Pathology, Brace Branchetta 20, 51 000 Rijeka, Croatia. RP Matusan-Ilijas, K (reprint author), Rijeka University School of Medicine, Department of Pathology, 51 000 Rijeka, Croatia. EM kmatusan@yahoo.com CR Uhm JH, Gladson CL, Rao JS, 1999, The role of integrins in the malignant phenotype of gliomas. Front Bioscience 4:188–199 Denhardt D, Guo X, 1993, Osteopontin: a protein with diverse functions. Faseb J 7:1475–1482 Rittling SR, Chambers AF, 2004, Role of osteopontin in tumour progression. Br J Cancer 90:1877–1881 Tuck AB, Arsenault DM, O’Malley FP et al, 1999, Osteopontin induces increased invasiveness and plasminogen activator expression of human mammary epithelial cells. Oncogene 18:4237– 4246 Hirama M, Takahashi F, Takahashi K et al, 2003, Osteopontin overproduced by tumour cells acts as a potent angiogenic factor contributing to tumour growth. Cancer Lett 198:107–117 Lin YH, Huang CJ, Chao JR et al, 2000, Coupling of osteopontin and its cell surface receptor CD44 to the cell survival response elicited by interleukin 3 or granulocyte-macrophage colonystimulating factor. Mol Cell Biol 20:2734–2742 Tuck AB, Hota C, Wilson SM et al, 2003, Osteopontin-induced migration of human mammary epithelial cells involves activation of EGF receptor and multiple signal transduction pathways. Oncogene 22:1198–1205 Denhardt DT, Chambers AF, 1994, Overcoming obstacles to metastasis-defenses against host defenses: osteopontin, OPN, as a shield against attack by cytotoxic host cells. J Cell Biochem 56:48–51 Geissinger E,Weisser C, Fischer P et al, 2002, Autocrine stimulation by osteopontin contributes to antiapoptotic signalling of melanocytes in dermal collagen. Cancer Res. Aug 62:4820–4828 Colla S, Morandi F, Lazzaretti M et al, 2005, Human myeloma cells express the bone regulating gene Runx2/Cbfa1 and produce osteopontin that is involved in angiogenesis in multiple myeloma patients. Leukemia 19:2166–2176 Coppola D, Szabo M, Boulware D et al, 2004, Correlation of osteopontin protein expression and pathological stage across a wide variety of tumour histogenesis. Clin Cancer Res 10:184–190 Matusan K, Dordevic G, Stipic D et al, 2006, Osteopontin expression correlates with prognostic variables and survival in clear cell renal cell carcinoma. J Surg Oncol 94:325–331 Rudland PS, Platt-Higgins A, El-Tanani M et al, 2002, Prognostic significance of the metastasis-associated protein osteopontin in human breast cancer. Cancer Res 62:3417–3427 Shijubo N, Uede T, Kon S et al, 1999, Vascular endothelial growth factor and osteopontin in stage I lung adenocarcinoma. Am J Respir Crit Care Med 160:1269–1273 Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, eds,, 2007, WHO classification of tumours of the central nervous system, fourth edition. IARC Press, Lyon Allred DC, Clark GM, Elledge R et al, 1993, Association of p53 protein expression with tumour cell proliferation rate and clinical outcome in node-negative breast cancer. J Natl Cancer Inst 85:200–206 Ellison JA, Velier JJ, Spera P et al, 1998, Osteopontin and its integrin receptor avb3 are upregulated during formation of the glial scar after focal stroke. Stroke 29:1698–1706 Moon C, Heo S, Ahn M et al, 2004, Immunohistochemical study of osteopontin in spinal cords of rats with clip compression injury. J Vet Med Sci 66:1307–1310 Shin T, Ahn M, Kim H et al, 2005, Temporal expression of osteopontin and CD44 in rat brains with experimental cryolesions. Brain Res 104:95–101 Sinclair C, Mirakhur M, Kirk J et al, 2005, Up-regulation of osteopontin and ab-crystallin in the normal-appearing white matter of multiple sclerosis: an immunohistochemical study utilizing tissue microarrays. Neuropathol Appl Neurobiol 3:292–303 Ding Q, Stewart J Jr, Prince CW et al, 2002, Promotion of malignant astrocytoma cell migration by osteopontin expressed in the normal brain: differences in integrin signalling during cell adhesion to osteopontin and vitronectin. Cancer Res 62:5336–5343 Saitoh Y, Kuratsu J, Takeshima H et al, 1995, Expression of osteopontin in human glioma. Its correlation with the malignancy. Lab Invest 72:55–63 Said HM, Hagemann C, Staab A et al, 2007, Expression patterns of the hypoxia-related genes osteopontin, CA9, erythropoietin, VEGF and HIF-1alpha in human glioma in vitro and in vivo. Radiother Oncol 83:398–405 Jang T, Savarese T, Low HP et al, 2006, Osteopontin expression in intratumoural astrocytes marks tumour progression in gliomas induced by prenatal exposure to N-ethyl-N-nitrosourea. Am J Pathol 168:1676–1685 Tucker MA, Chang PL, Prince CW et al, 1998, TPA-mediated regulation of osteopontin in human malignant glioma cells. Anticancer Res 18:807–812 Colin C, Baeza N, Bartoli C et al, 2006, Identification of genes differentially expressed in glioblastoma versus pilocytic astrocytoma using suppression subtractive hybridization. Oncogene 25:2818–2826 Mariani L, Beaudry C, McDonough WS et al, 2001, Glioma cell motility is associated with reduced transcription of proapoptotic and proliferation genes: a cDNA microarray analysis. J Neuro- Oncol 53:161–176 Senger DR, Ledbetter SR, Claffey KP et al, 1996, Stimulation of endothelial cell migration by vascular permeability factor/vascular endothelial growth factor through cooperative mechanisms involving the avβ3 integrin, osteopontin, and thrombin. Am J Pathol 149:293–305 Scatena M, Almeida M, Chaisson ML et al, 1998, NF-κB mediates avβ3 integrin-induced endothelial cell survival. J Cell Biol 141:1083–1093 Takano S, Tsuboi K, Tomono Y et al, 2000, Tissue factor, osteopontin, avβ3 integrin expression in microvasculature of gliomas associated with vascular endothelial growth factor expression. British J Cancer 82:1967–1973 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 293 EP 298 DI 10.1007/s12253-008-9058-4 PG 6 ER PT J AU Orban, E Szabo, E Lotz, G Kupcsulik, P Paska, Cs Schaff, Zs Kiss, A AF Orban, Erika Szabo, Erzsebet Lotz, Gabor Kupcsulik, Peter Paska, Csilla Schaff, Zsuzsa Kiss, Andras TI Different Expression of Occludin and ZO-1 in Primary and Metastatic Liver Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Colorectal liver metastasis; Occludin; ZO-1 ID Hepatocellular carcinoma; Colorectal liver metastasis; Occludin; ZO-1 AB Tight junction (TJ) components were found to be correlated with carcinogenesis and tumor development. TJs are composed of three main integral membrane proteins; occludin, claudins and JAMs. Alteration of the TJ protein expression may play an important role in the process of cell dissociation, which is among the first steps of tumor invasion and metastasis. Reduced expression of ZO-1 has been reported to be associated with invasion of several tumors. The aim of the present study was to detect differences between occludin and ZO-1 expression in normal liver samples, HCCs and colorectal liver metastases. Expression of occludin and ZO-1 was analysed in 25 surgically removed human hepatocellular carcinomas (HCC) and 25 human colorectal liver metastases. Gene expression levels were measured by real-time RT PCR, protein expression was determined by immunohistochemistry, comparing tumors with the surrounding nontumorous parenchyma and with seven normal liver samples. Occludin and ZO-1 mRNAs showed significant downregulation in HCCs in comparison with normal liver and were also downregulated in the metastases when compared with normal liver. Occludin and ZO-1 proteins were weakly expressed on hepatocytes in normal liver, while strong expression was found on bile canaliculi. In HCCs occludin and ZO-1 did not show immunopositivity on tumor cells, while colorectal metastatic tumors revealed high levels of these molecules. HCCs and metastases are characterized by markedly different protein expression pattern of occludin and ZO-1, which phenomenon might be attributed to the different histogenesis of these tumors. C1 [Orban, Erika] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Szabo, Erzsebet] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Kupcsulik, Peter] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Paska, Csilla] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM schaff@korb2.sote.hu CR Jiang WG, 1998, Cell adhesion molecules in the formation of liver metastasis. J Hepatobiliary Pancreat Surg 5:375–382 Kaihara T, Kusaka T, Nishi M et al., 2003, Dedifferentiation and decreased expression of adhesion molecules, E-cadherin and ZO-1, in colorectal cancer are closely related to liver metastasis. J Exp Clin Cancer Res 22:117–123 Sakisaka S, Kawaguchi T, Taniguchi E et al., 2001, Alterations in tight junctions differ between primary biliary cirrhosis and primary sclerosing cholangitis. Hepatology 33:1460–1468 Kojima T, Yamamoto T, Murata M et al., 2003, Regulation of the blood–biliary barrier: interaction between gap and tight junctions in hepatocytes. Med Electron Microsc 36:157–164 Haynes MD, Martin TA, Jenkins SA et al., 2005, Tight junctions and bladder cancer, review). Int J Mol Med 16:3–9 Cereijido M, Shoshani L, Contreras RG, 2000, Molecular physiology and pathophysiology of tight junctions. I. Biogenesis of tight junctions and epithelial polarity. Am J Physiol Gastrointest Liver Physiol 279:G477–482 Fanning AS, Mitic LL, Anderson JM, 1999, Transmembrane proteins in the tight junction barrier. J Am SocNephrol 10:1337–1345 Saitou M, Furuse M, Sasaki H et al., 2000, Complex phenotype of mice lacking occludin, a component of tight junction strands. Mol Biol Cell 11:4131–4142 Gonzalez-Mariscal L, Betanzos A, Nava P et al., 2003, Tight junction proteins. Prog Biophys Mol Biol 81:1–44 Itoh M, Nagafuchi A, Yonemura S et al., 1993, The 220-kD protein colocalizing with cadherins in non-epithelial cells is identical to ZO-1, a tight junction-associated protein in epithelial cells: cDNA cloning and immunoelectron microscopy. J Cell Biol 121:491–502 Itoh M, Furuse M, Morita K et al., 1999, Direct binding of three tight junction-associated MAGUKs, ZO-1, ZO-2, and ZO-3, with the COOH termini of claudins. J Cell Biol 147:1351–1363 Ebnet K, Schulz CU, Meyer Zu Brickwedde MK et al., 2000, Junctional adhesion molecule interacts with the PDZ domaincontaining proteins AF-6 and ZO-1. J Biol Chem 275:27979– 27988 Fanning AS, Jameson BJ, Jesaitis LA et al., 1998, The tight junction protein ZO-1 establishes a link between the transmembrane protein occludin and the actin cytoskeleton. J Biol Chem 273:29745–29753 Wittchen ES, Haskins J, Stevenson BR, 1999, Protein interactions at the tight junction. Actin has multiple binding partners, and ZO-1 forms independent complexes with ZO-2 and ZO-3. J Biol Chem 274:35179–35185 Fanning AS, Anderson JM, 1999, PDZ domains: fundamental building blocks in the organization of protein complexes at the plasma membrane. J Clin Invest 103:767–772 Lodi C, Szabo E, Holczbauer A et al., 2006, Claudin-4 differentiates biliary tract cancers from hepatocellular carcinomas. Mod Pathol 19:460–469 Pfaffl MW, Horgan GW, Dempfle L, 2002, Relative expression software tool, REST, for group-wise comparison and statistical analysis of relative expression results in real-time PCR. Nucleic Acids Res 30:e36 Tan X, Tamori Y, Egami H et al., 2004, Analysis of invasionmetastasis mechanism in pancreatic cancer: involvement of tight junction transmembrane protein occludin and MEK/ERK signal transduction pathway in cancer cell dissociation. Oncol Rep 11:993–998 Schneeberger EE, Lynch RD, 2004, The tight junction: a multifunctional complex. Am J Physiol Cell Physiol 286: C1213–1228 Saitou M, Fujimoto K, Doi Y et al., 1998, Occludin-deficient embryonic stem cells can differentiate into polarized epithelial cells bearing tight junctions. J Cell Biol 141:397– 408 Smalley KS, Brafford P, Haass NK et al., 2005, Up-regulated expression of zonula occludens protein-1 in human melanoma associates with N-cadherin and contributes to invasion and adhesion. Am J Pathol 166:1541–1554 Mauro L, Bartucci M, Morelli C et al., 2001, IGF-I receptorinduced cell–cell adhesion of MCF-7 breast cancer cells requires the expression of junction protein ZO-1. J Biol Chem 276:39892– 39897 Palmer HG, Gonzalez-Sancho JM, Espada J et al., 2001, Vitamin D(3, promotes the differentiation of colon carcinoma cells by the induction of E-cadherin and the inhibition of beta-catenin signaling. J Cell Biol 154:369–387 Brabletz T, Jung A, Reu S et al., 2001, Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci U S A 98:10356–10361 Busch C, Hanssen TA, Wagener C et al., 2002, Down-regulation of CEACAM1 in human prostate cancer: correlation with loss of cell polarity, increased proliferation rate, and Gleason grade 3 to 4 transition. Hum Pathol 33:290–298 Kimura Y, Shiozaki H, Hirao M et al., 1997, Expression of occludin, tight-junction-associated protein, in human digestive tract. Am J Pathol 151:45–54 Soini Y, 2005, Expression of claudins 1, 2, 3, 4, 5 and 7 in various types of tumours. Histopathology. 46:551–60 Cheung ST, Leung KL, Ip YC et al., 2005, Claudin-10 expression level is associated with recurrence of primary hepatocellular carcinoma. Clin Cancer Res 11:551–556 Ishikawa Y, Akishima-Fukasawa Y, Ito K et al., 2008, Histopathologic determinants of regional lymph node metastasis in early colorectal cancer. Cancer 112:924–933 Kominsky SL, Tyler B, Sosnowski J et al., 2007, Clostridium perfringens enterotoxin as a novel-targeted therapeutic for brain metastasis. Cancer Res 67:7977–7982 Sawada N, Murata M, Kikuchi K et al., 2003, Tight junctions and human diseases. Med Electron Microsc 36:147–156 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 299 EP 306 DI 10.1007/s12253-008-9031-2 PG 8 ER PT J AU Romics, I Banfi, G Szekely, E Krenacs, T Szende, B AF Romics, Imre Banfi, Gergely Szekely, Eszter Krenacs, Tibor Szende, Bela TI Expression of p21waf1/cip1, p27kip1, p63 and Androgen Receptor in Low and High Gleason Score Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE High Gleason score; Loss of p27kip1; Prostate adenocarcinoma ID High Gleason score; Loss of p27kip1; Prostate adenocarcinoma AB The aim of this study was to investigate the expression of p21waf1/cip1, p27kip1, p63 and androgen receptor proteins in relation to serum prostate specific antigen levels in low and high Gleason score prostate cancers. Biopsies of patients suffering from prostate adenocarcinoma of low (3+3 to 3+4) and high (5+4 to 5+5) Gleason scores (13 cases each group) were immunostained for positive regulators of cell cycle control (p21waf1/cip1 and p27kip1), and essential markers of normal prostate gland ontogeny (p63) and growth (androgen receptor) to find differentially expressed markers of malignant progression. Serum prostate specific antigen levels were also monitored at the time of biopsy and following anti-androgen therapy. All cases except one in each group were androgen receptor positive. P63 and p21waf1/cip1 proteins detected in normal basal cell nuclei were lost in all but one studied tumors respectively. P27kip1 protein, however, was detected in all low Gleason score prostate cancers, but it was found in only 7/13 high score cases. Prostate specific antigen levels, either pre- or post-treatment, did not show strict correlation with the p27kip1 results. The low to high grade dedifferentiation of prostate adenocarcinoma is accompanied with the downregulation of p27kip1 protein, which may be an important molecular sign of the lost cell cycle control. C1 [Romics, Imre] Semmelweis University, Department of UrologyBudapest, Hungary. [Banfi, Gergely] Semmelweis University, Department of UrologyBudapest, Hungary. [Szekely, Eszter] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Krenacs, Tibor] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, Ulloi u. 26, 1085 Budapest, Hungary. [Szende, Bela] Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, Ulloi u. 26, 1085 Budapest, Hungary. RP Szende, B (reprint author), Hungarian Academy of Sciences and Semmelweis University, Research Group of Molecular Pathology, 1085 Budapest, Hungary. EM bszende@korb1.sote.hu CR Isaacs JT, Lundmo PI, Berges R, Martikainen P, Kyprianou N, English HF, 1992, Androgen regulation of programmed death of normal and malignant prostatic cells. J Androl 13:457–464 Eder IE, Culig Z, Putz T, Nessler-Menardi C, Bartsch G, Klocker H, 2001, Molecular biology of the androgen receptor: from molecular understanding to the clinic. Eur Urol 40:241–251 Stamey TA, Warrington JA, Caldwell MC et al, 2001, Molecular genetic profiling of Gleason grade 4/5 prostate cancers compared to benign prostatic hyperplasia. J Urol 166:2171–2177 Porkka KP, Visakorpi T, 2001, Detection of differentially expressed genes in prostate cancer by combining suppression subtractive hybridization and cDNA library assay. J Pathol 193:73–79 Meehan KL, Holland JW, Dawkins HJ, 2002, Proteomic analysis of normal and malignant prostate tissue to identify novel proteins lost in cancer. Prostate 50:54–63 Baretton GB, Klenk U, Diebold J, Schmeller N, Lohrs U, 1999, Proliferation- and apoptosis-associated factors in advanced prostatic carcinomas before and after androgen deprivation therapy: prognostic significance of p21/WAF1/CIP1 expression. Br J Cancer 80:546–555 Eder IE, Bektic J, Haag P, Bartsch G, Klocker H, 2004, Genes differentially expressed in prostate cancer. BJU Int 93:1151–1155 Szende B, Romics I, Minik K, Szabo J, Torda I, Lovasz S, Toth L, Bely M, Kerenyi T, Bartok K, Vegh A, 2001, Repeated biopsies in evaluation of therapeutic effects in prostate carcinoma. Prostate 49:93–100 Waltregny D, Leav I, Signoretti S, Soung P, Lin D, Merk F, Adams JY, Bhattecharya N, Cirenei N, Loda M, 2001, Androgendriven prostate epithelial cell proliferation and differentiation in vivo involve the regulation of p27. Mol Endocrinol 15:765–782 Koffer L, Roshong S, Park IK, Cesen-Cummings K, Thopson DR, Dwyer-Nield LD, Rice P, Mamay C, Malkinson AM, Ruch RJ, 2000, Growth inhibition in G1 and altered expression of cyclin D1 and p27kip1 after forced connexin expression in lung and liver carcinoma cells. J Cell Biochem 79:347–354 Zhang Y-W, Morite I, Ikeda M, Ma K-W, Murota S, 2001, Connexin43 suppresses proliferation of osteosarcoma U2OS cells through post-transcriptional regulation of p27. Oncogene 20:4138–4149 Gorbe A, Becker DL, Dux L, Stelkovics E, Krenacs L, Bagdi E, Krenacs T, 2005, Transient upregulation of connenxin43 gap junctions and synchronized cell cycle control precede myoblast fusion in regenerating skeletal muscle in vivo. Histochem Cell Biol 123:573–583 Lawlor MA, Rotwein P, 2000, Coordinate control of muscle cell survival by distinct insulin-like growth factor activated signaling pathways. J Cell Biol 151:1131–1140 Ostrovsky O, Bengal E, 2003, The mitogen-activated protein kinase cascade promotes myoblast cell survival by stabilizing the cyclin-dependent kinase inhibitor, p21WAF1 protein. J Biol Chem 278:21221–21231 Fizazi K, Martizez LA, Sikes CR, Johnston DA, Stephens LC, McDonnell TJ, Logothetis CJ, Trapman J, Pisters LL, Ordonez NG, Troncoso P, Navone NM, 2002, The association of p21, WAF- 1/CIP1, with progression to androgen-independent prostate cancer. Clin Cancer Res 8:775–781 Signoretti S, Waltregny D, Dilks J, Isaac B, Lin D, Garraway L, Yang A, Montironi R, McKeon F, Loda M, 2000, P63 is a prostate basal cell marker and is required for prostate development. Am J Pathol 157:1769–1775 Ronison IB, 2003, Tumor cell senescence in cancer treatment. Cancer Res 63:2705–2715 Di Como CJ, Urist MJ, Babayan I, Drobnjak M, Hedvat CV, Teruya-Feldstein J, Pohar K, Hoos A, Cordon-Cardo C, 2002, P63 expression profiles in human normal and tumor tissues. Clin Can Res 8:494–501 Cordon-Cardo G, Koff A, Drobnjak M, Caodieci P, Osman I, Millard SS, 1998, Distinct altered patterns of 27kip1 gene expression in benign prostate hyperplasia and prostatic carcinoma. J Natl Cancer Inst 90:1284–1291 Guo Y, Sklar GN, Borkowski A, Kyprianou N, 1997, Loss of the cyclin dependent kinase inhibitor p27(Kip1, protein in human prostate cancers correlates with tumor grade. Clin Cancer Res 3:2269–2274 Kuczyk M, Machtens S, Hradil K et al, 1999, Predictive value of p27Kip1 protein expression for the recurrence-free and longterm survival of prostate cancer patients. Br J Cancer 81:1052– 1058 Doganavsargil B, Simsir A, Boyacioglu H, Cal C, Hekimgil M, 2006, A comparison of p21 and p27 immunoexpression in benign glands, prostatic intraepithelial neoplasia and prostate adenocarcinoma. BJU Int 97:644–648 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 307 EP 311 DI 10.1007/s12253-008-9042-z PG 5 ER PT J AU Mark, Zs Bajzik, G Nagy, A Bogner, P Repa, I Strausz, J AF Mark, Zsuzsa Bajzik, Gabor Nagy, Andrea Bogner, Peter Repa, Imre Strausz, Janos TI Comparison of Virtual and Fiberoptic Bronchoscopy in the Management of Airway Stenosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bronchoscopy; Computed tomography; Interventional bronchoscopy .Virtual bronchoscopy ID Bronchoscopy; Computed tomography; Interventional bronchoscopy .Virtual bronchoscopy AB Noninvasive imaging methods can be valuable tools for diagnosing thoracic diseases, especially malignancies. The aim of this study was to compare the effectiveness of conventional and virtual bronchoscopy in the follow-up of patients with large airway stenosis. Twenty-three consecutive patients with stenoses of the trachea and/or the main bronchi were enrolled in this prospective observer study. The causes of stenosis included malignant or benign tumours, goiter, and postintubation stenoses. Patients were evaluated before and after treatment (which included mechanical dilation, laser photocoagulation, stent implantation, radiotherapy, chemotherapy, and surgical resection). The mean time between baseline and follow-up endoscopy was 140 days. No significant differences were observed between the estimated and measured data from bronchofibroscopy and virtual bronchoscopy. Exact measurement of stenoses was performed with virtual bronchoscopy. C1 [Mark, Zsuzsa] Pest County Hospital, III.Pulmonology, Munkacsy Mihaly 70, 2045 Torokbalint, Hungary. [Bajzik, Gabor] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. [Nagy, Andrea] Pest County Hospital, III.Pulmonology, Munkacsy Mihaly 70, 2045 Torokbalint, Hungary. [Bogner, Peter] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. [Repa, Imre] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. [Strausz, Janos] National Koranyi Institute of Pulmonology, Piheno u. 1, 1125 Budapest, Hungary. RP Mark, Zs (reprint author), Pest County Hospital, III.Pulmonology, 2045 Torokbalint, Hungary. EM markzs@t-online.hu CR Neumann K, Winterer J, Kimmig M et al, 2000, Real-time interactive virtual endoscopy of the tracheo-bronchial system: influence of CT imaging protocols and observer ability. Eur J Radiol 33:50–54 Hoppe H, Walder B, Sonnenschein M et al, 2002, Multidetector CT virtual bronchoscopy to grade tracheobronchial stenosis. AJR Am J Roentgenol 178:1195–1200 Liewald F, Lang G, Fleiter T et al, 1998, Comparison of virtual and fiberoptic bronchoscopy. Thorac Cardiovasc Surg 46:361–364 Mark Zs, Bajzik G, Repa I et al, 2001, Virtual bronchoscopy: a new non-invasive diagnostic method in pulmonology. Orv Hetil 142:565–569 Haponik EF, Aquino SL, Vining DJ, 1999, Virtual bronchoscopy. Clin Chest Med 20:201–217 Heyer CM, Nuesslein TG, Jung D et al, 2007, Tracheobronchial anomalies and stenoses: detection with low-dose multidetector CT with virtual tracheobronchoscopy—comparison with flexible tracheabronchoscopy. Radiology 242:542–549 Burke AJ, Vining DJ, McGuirt WF et al, 2000, Evaluation of airway obstruction using virtual endoscopy. Laryngoscope 110:23–29 Colt HG, Crawford SW, Galbraith O III, 2001, Virtual reality bronchoscopy simulation: a revolution in procedural training. Chest 120:1333–1339 McAdams HP, Goodman PC, Kussin P, 1998, Virtual bronchoscopy for directing transbronchial needle aspiration of hilar and mediastinal lymph nodes: a pilot study. AJR Am J Roentgenol 170:1361–1364 Summers RM, 1997, Navigational aids for real-time virtual bronchoscopy. AJR Am J Roentgenol 168:1165–1170 Ferretti G, 1999, Virtual endoscopy: which indications? Rev Mal Respir 3:59–60 Bauer TL, Steiner KV, 2007, Virtual bronchoscopy: clinical applications and limitations. Surg Oncol Clin N Am 16:323– 328 Summers RM, Shaw DJ, Shelhamer JH, 1998, CT virtual bronchoscopy of simulated endobronchial lesions: effect of scanning, reconstruction, and display settings and potential pitfalls. AJR Am J Roentgenol 170:947–950 Koletsis EN, Kalogeropoulu C, Prodromaki E et al, 2007, Tumoral and non-tumoral trachea stenoses: evaluation with three dimensional CT and virtual bronchoscopy. J Cardiothorac Surg 2:18 Amorico MG, Drago A, Vetruccio E et al, 2006, Tracheobronchial stenosis: role of virtual endoscopy in diagnosis and follow-up after therapy. Radiol Med, Torino, 111:1064–1077 Ferretti GR, Kocier M, Calaque O et al, 2003, Follow-up after stent insertion in the tracheobronchial tree: role of helical computed tomography in comparison with fiberoptic bronchoscopy. Eur Radiol 13:1172–1178 Mark Zs, Bajzik G, Nagy A et al, 2004, Virtual bronchoscopy for follow-up investigation of main airway stenoses. Orv Hetil 145:1233–1235 Shitrit D, Valdsislav P, Grubstein A et al, 2005, Accuracy of virtual bronchoscopy for grading tracheobronchial stenosis: correlation with pulmonary function test and fiberoptic bronchoscopy. Chest 128:3545–3550 Finkelstein SE, Schrump DS, Nguyen DM et al, 2003, Comparative evaluation of super high-resolution CT scan and virtual bronchoscopy for the detection of tracheobronchial malignancies. Chest 124:1834–1840 Higgins WE, Ramaswamy K, Swift RD et al, 1998, Virtual bronchoscopy for three-dimensional pulmonary image assessment: state of the art and future needs. Radiographics 18:761–778 Wood BJ, Razavi P, 2002, Virtual endoscopy: a promising new technology. Am Fam Physician 66:107–112 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 313 EP 319 DI 10.1007/s12253-008-9059-3 PG 7 ER PT J AU Hussein, RM Fathi, AN El-Din, MEA Hassan, IH Abdullah, F AL-Hakeem, E Backer, AE AF Hussein, R Mahmoud Fathi, A Nehal El-Din, M Ezz Azza Hassan, I Hewayda Abdullah, Fatemah AL-Hakeem, Eman Backer, Abo Eman TI Alterations of the CD4+, CD8+ T Cell Subsets, Interleukins-1β, IL-10, IL-17, Tumor Necrosis Factor-α and Soluble Intercellular Adhesion Molecule-1 in Rheumatoid Arthritis and Osteoarthritis: Preliminary Observations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Rheumatoid arthritis; Osteoarthritis; Tcells ID Rheumatoid arthritis; Osteoarthritis; Tcells AB Rheumatoid arthritis is a multisystem disease with underlying immune mechanisms. Osteoarthritis is a debilitating, progressive disease of diarthrodial joints associated with the aging process. Although much is known about the pathogenesis of rheumatoid arthritis and osteoarthritis, our understanding of some immunologic changes remains incomplete. This study tries to examine the numeric changes in the T cell subsets and the alterations in the levels of some cytokines and adhesion molecules in these lesions. To accomplish this goal, peripheral blood and synovial fluid samples were obtained from 24 patients with rheumatoid arthritis, 15 patients with osteoarthritis and six healthy controls. The counts of CD4 + and CD8 + T lymphocytes were examined using flow cytometry. The levels of some cytokines (TNF-α, IL1-β, IL-10, and IL-17) and a soluble intercellular adhesion molecule-1 (sICAM-1) were measured in the sera and synovial fluids using enzyme linked immunosorbant assay. We found some variations in the counts of T cell subsets, the levels of cytokines and sICAM-1 adhesion molecule between the healthy controls and the patients with arthritis. High levels of IL-1β, IL-10, IL-17 and TNF-α (in the serum and synovial fluid) were observed in arthritis compared to the healthy controls. In rheumatoid arthritis, a high serum level of sICAM-1 was found compared to its level in the synovial fluid. A high CD4+/CD8+ T cell ratio was found in the blood of the patients with rheumatoid arthritis. In rheumatoid arthritis, the cytokine levels correlated positively with some clinicopathologic features. To conclude, the development of rheumatoid arthritis and osteoarthritis is associated with alteration of the levels of some cytokines. The assessment of these immunologic changes may have potential prognostic roles. C1 [Hussein, R Mahmoud] Assuit University, Assuit University Hospitals, Department of PathologyAssuit, Egypt. [Fathi, A Nehal] Assuit and Ein Shams Universities, Faculty of Medicine, Assuit University Hospitals, Departments of Rheumatology and RehabilitationAssuit, Egypt. [El-Din, M Ezz Azza] Assuit University, Assuit University Hospitals, Department of PathologyAssuit, Egypt. [Hassan, I Hewayda] Assuit University, Assuit University Hospitals, Department of PathologyAssuit, Egypt. [Abdullah, Fatemah] Assuit and Ein Shams Universities, Faculty of Medicine, Assuit University Hospitals, Departments of Rheumatology and RehabilitationAssuit, Egypt. [AL-Hakeem, Eman] Assuit and Ein Shams Universities, Faculty of Medicine, Assuit University Hospitals, Departments of Rheumatology and RehabilitationAssuit, Egypt. [Backer, Abo Eman] Assuit and Ein Shams Universities, Faculty of Medicine, Assuit University Hospitals, Departments of Rheumatology and RehabilitationAssuit, Egypt. RP Hussein, RM (reprint author), Assuit University, Assuit University Hospitals, Department of Pathology, Assuit, Egypt. EM mrcpath17@gmail.com CR Maurer D, Felzmann T, Holter W, Petera P, Smolen J, Knapp W, 1992, Evidence for the presence of activated CD4 T cells with naive phenotype in the peripheral blood of patients with rheumatoid arthritis. Clin Exp Immunol 87:429–434 Frye CA, Yocum DE, Tuan R, Suyana E, Seftor EA, Seftor RE, Khalkhali-Ellis Z, Moore TL, Hendrix MJ, 1996, An in vitro model for studying mechanisms underlying synoviocyte-mediated cartilage invasion in rheumatoid arthritis. Pathol Oncol Res 2:157–166 Khalkhali-Ellis Z, Bulla GA, Schlesinger LS, Kirschmann DA, Moore TL, Hendrix MJ, 1999, C1q-containing immune complexes purified from sera of juvenile rheumatoid arthritis patients mediate IL-8 production by human synoviocytes: role of C1q receptors. J Immunol 163:4612–4620 Choy EH, Connolly DJ, Rapson N, Jeal S, Brown JC, Kingsley GH, Panayi GS, Johnston JM, 2000, Pharmacokinetic, pharmacodynamic and clinical effects of a humanized IgG1 anti-CD4 monoclonal antibody in the peripheral blood and synovial fluid of rheumatoid arthritis patients. Rheumatology, Oxford, 39:1139– 1146 Khalkhali-Ellis Z, Roodman ST, Knutsen AP, Mueller KR, Chauhan B, Moore TL, Hendrix MJ, 1998, Expression of macrophage markers by a population of T cells obtained from synovial fluid of a subgroup of patients with juvenile rheumatoid arthritis. J Rheumatol 25:352–360 Kotake S, Udagawa N, Takahashi N, Matsuzaki K, Itoh K, Ishiyama S, Saito S, Inoue K, Kamatani N, Gillespie MT, Martin TJ, Suda T, 1999, IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis. J Clin Invest 103:1345–1352 Zwerina J, Redlich K, Schett G, Smolen JS, 2005, Pathogenesis of rheumatoid arthritis: targeting cytokines. Ann N Y Acad Sci 1051:716–729 Malemud CJ, 2004, Cytokines as therapeutic targets for osteoarthritis. BioDrugs 18:23–35 Aigner T, Sachse A, Gebhard PM, Roach HI, 2006, Osteoarthritis: pathobiology-targets and ways for therapeutic intervention. Adv Drug Deliv Rev 58:128–149 Zangerle PF, De Groote D, Lopez M, Meuleman RJ, Vrindts Y, Fauchet F, Dehart I, Jadoul M, Radoux D, Franchimont P, 1992, Direct stimulation of cytokines, IL-1 beta, TNF-alpha, IL-6, IL-2, IFN-gamma and GM-CSF, in whole blood: II. Application to rheumatoid arthritis and osteoarthritis. Cytokine 4:568–575 Ezawa K, Yamamura M, Matsui H, Ota Z, Makino H, 1997, Comparative analysis of CD45RA- and CD45RO-positive CD4 + T cells in peripheral blood, synovial fluid, and synovial tissue in patients with rheumatoid arthritis and osteoarthritis. Acta Med Okayama 51:25–31 Petrovic-Rackov L, Pejnovic N, 2005, Clinical significance of IL- 18, IL-15, IL-12 and TNF-alpha measurement in rheumatoid arthritis. Clin Rheumatol 25:448–452 Firestein GS, 2005, Immunologic mechanisms in the pathogenesis of rheumatoid arthritis. J Clin Rheumatol 11(3 Suppl):S39–44 Abramson SB, Amin A, 2002, Blocking the effects of IL-1 in rheumatoid arthritis protects bone and cartilage. Rheumatology, Oxford, 41:972–980 Attur MG, Patel IR, Patel RN, Abramson SB, Amin AR, 1998, Autocrine production of IL-1 beta by human osteoarthritisaffected cartilage and differential regulation of endogenous nitric oxide, IL-6, prostaglandin E2, and IL-8. Proc Assoc Am Physicians 110:65–72 Appel H, Neure L, Kuhne M, Braun J, Rudwaleit M, Sieper J, 2004, An elevated level of IL-10- and TGFbeta-secreting T cells, B cells and macrophages in the synovial membrane of patients with reactive arthritis compared to rheumatoid arthritis. Clin Rheumatol 23:435–440 Verhoef CM, van Roon JA, Vianen ME, Bijlsma JW, Lafeber FP, 2001, Interleukin 10, IL-10), not IL-4 or interferon-gamma production, correlates with progression of joint destruction in rheumatoid arthritis. J Rheumatol 28:1960–1966 Miossec P, 2004, IL-17 in rheumatoid arthritis: a new target for treatment or just another cytokine? Joint Bone Spine 71:87–90 Ryu S, Lee JH, Kim SI, 2006, IL-17 increased the production of vascular endothelial growth factor in rheumatoid arthritis synoviocytes. Clin Rheumatol 25:16–20 Lindsley HB, Smith DD, Davis LS, Koch AE, Lipsky PE, 1992, Regulation of the expression of adhesion molecules by human synoviocytes. Semin Arthritis Rheum 21:330–334 Cush JJ, Rothlein R, Lindsley HB, Mainolfi EA, Lipsky PE, 1993, Increased levels of circulating intercellular adhesion molecule 1 in the sera of patients with rheumatoid arthritis. Arthritis Rheum 36:1098–1102 Gonzalez-Gay MA, Garcia-Unzueta MT, De Matias JM, Gonzalez- Juanatey C, Garcia-Porrua C, Sanchez-Andrade A, Martin J, Llorca J, 2006, Influence of anti-TNF-alpha infliximab therapy on adhesion molecules associated with atherogenesis in patients with rheumatoid arthritis. Clin Exp Rheumatol 24:373–379 Albert DA, Huang G, Dubrow G, Brensinger CM, Berlin JA, Williams HJ, 2004, Criteria for improvement in rheumatoid arthritis: alternatives to the American College of Rheumatology 20. J Rheumatol 31(5):856–866 Hussein MR, Hassan HI, Hofny ER, Elkholy M, Fatehy NA, Abd Elmoniem AE, Ezz El-Din AM, Afifi OA, Rashed HG, 2005, Alterations of mononuclear inflammatory cells, CD4/CD8 + T cells, interleukin 1beta, and tumour necrosis factor alpha in the bronchoalveolar lavage fluid, peripheral blood, and skin of patients with systemic sclerosis. J Clin Pathol 58:178–184 Barrera P, Faure S, Prud’homme JF, Balsa A, Migliorini P, Chimenti D, Radstake TR, van de Putte LB, Pascual-Salcedo D, Westhovens R, Maenaut K, Alves H, Lopes-Vaz A, Stravopoulos C, Spyropoulou M, Fritz P, Bardin T, Charron D, Lepage V, Alibert, Martinez M, Cornelis F, 2001, European genetic study on rheumatoid arthritis: is there a linkage of the interleukin-1, IL-1), IL-10 or IL-4 genes to RA? Clin Exp Rheumatol 19:709–714 Chabaud M, Page G, Miossec P, 2001, Enhancing effect of IL-1, IL-17, and TNF-alpha on macrophage inflammatory protein- 3alpha production in rheumatoid arthritis: regulation by soluble receptors and Th2 cytokines. J Immunol 167:6015–6020 Petrovic-Rackov L, 2006, Evaluation of the degree of clinical rheumatoid arthritis activity based on the concentrations of cytokines TNF-alpha, IL-12, IL-15, and IL-18 in serum and synovial fluid]. Vojnosanit Pregl 63:21–26 Brennan FM, Hayes AL, Ciesielski CJ, Green P, Foxwell BM, Feldmann M, 2002, Evidence that rheumatoid arthritis synovial T cells are similar to cytokine-activated T cells: involvement of phosphatidylinositol 3-kinase and nuclear factor kappaB pathways in tumor necrosis factor alpha production in rheumatoid arthritis. Arthritis Rheum 46:31–41 Fossiez F, Djossou O, Chomarat P, Flores-Romo L, Ait-Yahia S, Maat C, Pin JJ, Garrone P, Garcia E, Saeland S, Blanchard D, Gaillard C, Das Mahapatra B, Rouvier E, Golstein P, Banchereau J, Lebecque S, 1998, T cell interleukin-17 induces stromal cells to produce proinflammatory and hematopoietic cytokines. J Exp Med 183:2593–2603 Chabaud M, Fossiez F, Taupin JL, Miossec P, 1998, Enhancing effect of IL-17 on IL-1-induced IL-6 and leukemia inhibitory factor production by rheumatoid arthritis synoviocytes and its regulation by Th2 cytokines. J Immunol 161:409–414 Khalkhali-Ellis Z, Seftor EA, Nieva DR, Seftor RE, Samaha HA, Bultman L, De Larco JE, Ince A, Moore TL, Hendrix MJ, 1997, Induction of invasive and degradative phenotype in normal synovial fibroblasts exposed to synovial fluid from patients with juvenile rheumatoid arthritis: role of mononuclear cell population. J Rheumatol 24:2451–2460 Cush JJ, Splawski JB, Thomas R, McFarlin JE, Schulze-Koops H, Davis LS, Fujita K, Lipsky PE, 1995, Elevated interleukin-10 levels in patients with rheumatoid arthritis. Arthritis Rheum 38:96–104 Macchioni P, Boiardi L, Casali B, Nicoli D, Farnetti E, Salvarani C, 2000, Intercellular adhesion molecule 1, ICAM-1, gene polymorphisms in Italian patients with rheumatoid arthritis. Clin Exp Rheumatol 18:553–558 Blann AD, Herrick A, Jayson MI, 1995, Altered levels of soluble adhesion molecules in rheumatoid arthritis, vasculitis and systemic sclerosis. Br J Rheumatol 34:814–819 Dolezalova P, Telekesova P, Nemcova D, Hoza J, 2002, Soluble adhesion molecules ICAM-1 and E-selectin in juvenile arthritis: clinical and laboratory correlations. Clin Exp Rheumatol 20: 249–254 Hanyuda M, Kasama T, Isozaki T, Matsunawa MM, Yajima N, Miyaoka H, Uchida H, Kameoka Y, Ide H, Adachi M, 2003, Activated leucocytes express and secrete macrophage inflammatory protein-1alpha upon interaction with synovial fibroblasts of rheumatoid arthritis via a beta2-integrin/ICAM-1 mechanism. Rheumatology, Oxford, 42:1390–1397 Pigott R, Dillon LP, Hemingway IH, Gearing AJ, 1992, Soluble forms of E-selectin, ICAM-1 and VCAM-1 are present in the supernatants of cytokine activated cultured endothelial cells. Biochem Biophys Res Commun 187:584–589 Lindsley HB, Smith DD, Cohick CB, Koch AE, Davis LS, 1993, Proinflammatory cytokines enhance human synoviocyte expression of functional intercellular adhesion molecule-1, ICAM-1). Clin Immunol Immunopathol 68:311–320 Fitzgerald JE, Ricalton NS, Meyer AC, West SG, Kaplan H, Behrendt C, Kotzin BL, 1995, Analysis of clonal CD8 + T cell expansions in normal individuals and patients with rheumatoid arthritis. J Immunol 154:3538–3547 Burmester GR, Stuhlmuller B, Keyszer G, Kinne RW, 1997, Mononuclear phagocytes and rheumatoid synovitis. Mastermind or workhorse in arthritis? Arthritis Rheum 40:5–18 Benito MJ, Veale DJ, FitzGerald O, van den Berg WB, Bresnihan B, 2005, Synovial tissue inflammation in early and late osteoarthritis. Ann Rheum Dis 64:1263–1267 Miltenburg AM, van Laar JM, de Kuiper R, Daha MR, Breedveld FC, 1992, T cells cloned from human rheumatoid synovial membrane functionally represent the Th1 subset. Scand J Immunol 35:603–610 Rittner HL, Zettl A, Jendro MC, Bartz-Bazzanella P, Goronzy JJ, Weyand CM, 1997, Multiple mechanisms support oligoclonal T cell expansion in rheumatoid synovitis. Mol Med 3:452–465 Steiner G, Tohidast-Akrad M, Witzmann G, Vesely M, Studnicka- Benke A, Gal A, Kunaver M, Zenz P, Smolen JS, 1999, Cytokine production by synovial T cells in rheumatoid arthritis. Rheumatology, Oxford, 38:202–213 Franz JK, Kolb SA, Hummel KM, Lahrtz F, Neidhart M, Aicher WK, Pap T, Gay RE, Fontana A, Gay S, 1998, Interleukin-16, produced by synovial fibroblasts, mediates chemoattraction for CD4 + T lymphocytes in rheumatoid arthritis. Eur J Immunol 28:2661–2671 Mamoune A, Durand V, Le Goff P, Pennec YL, Youinou P, Le Corre R, 2000, Abnormal distribution of CD45 isoforms expressed by CD4 + and CD8 + T cells in rheumatoid arthritis. Histol Histopathol 15:587–591 Hatachi S, Iwai Y, Kawano S, Morinobu S, Kobayashi M, Koshiba M, Saura R, Kurosaka M, Honjo T, Kumagai S, 2003, CD4 + PD-1 + T cells accumulate as unique anergic cells in rheumatoid arthritis synovial fluid. J Rheumatol 30:1410–1419 van Amelsfort JM, Jacobs KM, Bijlsma JW, Lafeber FP, Taams LS, 2004, CD4(+)CD25(+, regulatory T cells in rheumatoid arthritis: differences in the presence, phenotype, and function between peripheral blood and synovial fluid. Arthritis Rheum 50:2775–2785 Mottonen M, Heikkinen J, Mustonen L, Isomaki P, Luukkainen R, Lassila O, 2005, CD4 + CD25 + T cells with the phenotypic and functional characteristics of regulatory T cells are enriched in the synovial fluid of patients with rheumatoid arthritis. Clin Exp Immunol 140:360–367 Mallya RK, Mace BE, 1981, The assessment of disease activity in rheumatoid arthritis using a multivariate analysis. Rheumatol Rehabil 20:14–17 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 321 EP 328 DI 10.1007/s12253-008-9016-1 PG 8 ER PT J AU Kobori, L Nagy, P Mathe, Z Hartmann, E Doros, A Paku, S Dezso, K Sapi, Z AF Kobori, Laszlo Nagy, Peter Mathe, Zoltan Hartmann, Erika Doros, Attila Paku, Sandor Dezso, Katalin Sapi, Zoltan TI Malignant Peripheral Nerve Sheath Tumor of the Liver: A Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE Malignant peripheral nerve sheath tumor; Liver; S100; Laminin ID Malignant peripheral nerve sheath tumor; Liver; S100; Laminin AB A large, rapidly growing malignant peripheral nerve sheath tumor (MPNST) of the liver in a young female patient, not associated with von Recklinghausen’s disease, is presented. Diagnosis was based on detailed immunohistochemical and electromicroscopic examination beside the characteristic H&E picture. As far as we know, this is the first reported, unambiguously proven "de novo" MPNST in the liver. Differential diagnostic problems are discussed and a review of the literature is given. C1 [Kobori, Laszlo] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Nagy, Peter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Mathe, Zoltan] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Hartmann, Erika] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Doros, Attila] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Paku, Sandor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Dezso, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. RP Sapi, Z (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM zsapi@freemail.hu CR Schmurun RI, Chibisov VN, 1977, Malignant neurinoma of the liver. Ark Pathol 39:69–71 Young SJ, 1975, Primary malignant neurilemmoma, schwannoma, of the liver in a case of neurofibromatosis. J Pathol 117:151–153 Lederman SM, Martin EC, Laffey KT, Lefkowitch JH, 1987, Hepatic neurofibromatosis, malignant schwannoma, and angiosarcoma in von Recklinghausen’s disease. Gastroenterology 92:234–239 Scheithauer BW, Woodruff JM, Erlandson RA, 1999, Primary malignant tumors of peripheral nerve In Atlas of tumor pathology: Tumors of the Peripheral nervous system, Third Series, Fascicle 24 Washington, DC, Armed Forsis Institute of Pathology, pp 303– 310 Weinberg AG, Finegold MJ, 1983, Primary hepatic tumors in childhood. Hum Pathol 14:512–537 Kiani B, Ferrell LD, Qualman S, Frankel WL, 2006, Immunohistochemical analysis of embryonal sarcoma of the liver. Appl Immunohistochem Mol Morphol 14:193–197 Walker NI, Horn MJ, Strong RW, Lynch SV, Cohen J, Ong TH, Harris OD, 1992, Undifferentiated, embryonal, sarcoma of the liver. Cancer 69:52–59 Fiel MI, Schwarz M, Min AD, Sung MW, Thung SN, 1996, Malignant schwannoma of the liver in a patient without neurofibromatosis. Arch Pathol Lab Med 120:1145–1147 Morikawa Y, Ishihara Y, Matsuura N, 1995, Malignant schwannoma of the liver. Dig Dis Sci 40:1279–1282 Yoshida M, Nakashima Y, Tanaka A, Mori K, Yamaoka Y, 1994, Benign schwannoma of the liver: a case report. Arch Jpn Chir 63:208–214 Hytiroglou P, Linton P, Klion F, Schwartz M, Miller C, Thung SN, 1993, Benign schwannoma of the liver. Arch Pathol Lab Med 117:216–218 Heffron TG, Coventry S, Bedendo F, Baker A, 1993, Resection of primary schwannoma of the liver not associated with neurofibromatosis. Arch Surg 128:1396–1398 Tuder RM, Moraes CF, 1984, Primary semimalignant schwannoma of the liver: light and electron microscopic studies. Pathol Res Pract 178:345–348 Woodruff JM, Selig AM, Crowley K, Allen PW, 1994, Schwannoma, neurilemmoma, with malignant transformation. Am J Surg Pathol 18:882–895 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 329 EP 332 DI 10.1007/s12253-008-9078-0 PG 4 ER PT J AU Hardisson, D Regojo, MR Marino-Enriquez, A Martinez-Garcia, M AF Hardisson, David Regojo, M Rita Marino-Enriquez, Adrian Martinez-Garcia, Mayte TI Signet-Ring Stromal Tumor of the Ovary: Report of a Case and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE Calretinin; Inhibin; Ovary; Sex cord-stromal tumor; Signet-ring stromal tumor ID Calretinin; Inhibin; Ovary; Sex cord-stromal tumor; Signet-ring stromal tumor AB Signet-ring stromal tumor of the ovary is extremely rare, with only ten cases reported in the literature. We report on a case of signet-ring stromal tumor of the left ovary in a 54-year-old woman who presented with abdominal discomfort. Histologically, the tumor was composed of an admixture of spindle and round cells which contained a large cytoplasmic vacuole which displaced the nucleus, creating a signet-ring appearance. Numerous cells showed intracytoplasmic hyaline globules. Immunohistochemically, the tumor cells showed positivity for vimentin, actin, inhibin, and calretinin, thus confirming the ovarian stromal origin of the neoplasm. The patient remains free of disease one year and 9 months after surgery. Signet-ring tumor of the ovary is a rare variant of benign ovarian stromal neoplasm and should be distinguished from metastatic mucin-secreting signet-ring adenocarcinoma. C1 [Hardisson, David] Autonomous University of Madrid, University Hospital La Paz, Department of Pathology, Paseo de la Castellana, 261, 28046 Madrid, Spain. [Regojo, M Rita] Autonomous University of Madrid, University Hospital La Paz, Department of Pathology, Paseo de la Castellana, 261, 28046 Madrid, Spain. [Marino-Enriquez, Adrian] Autonomous University of Madrid, University Hospital La Paz, Department of Pathology, Paseo de la Castellana, 261, 28046 Madrid, Spain. [Martinez-Garcia, Mayte] Autonomous University of Madrid, University Hospital La Paz, Department of Obstetrics and GynecologyMadrid, Spain. RP Hardisson, D (reprint author), Autonomous University of Madrid, University Hospital La Paz, Department of Pathology, 28046 Madrid, Spain. EM dhardisson.hulp@salud.madrid.org CR Ramzy I, 1976, Signet-ring stromal tumor of ovary. Histochemical, light, and electron microscopic study. Cancer 38:166–172 Suarez A, Palacios J, Burgos E et al, 1993, Signet-ring stromal tumor of the ovary: a histochemical, immunohistochemical and ultrastructural study. Virchows Arch A Pathol Anat 422:333–366 Dickersin GR, Young RH, Scully RE, 1995, Signet-ring stromal and related tumors of the ovary. Ultrastruct Pathol 19:401–419 Cashell AW, Jerome WG, Flores E, 2000, Signet ring stromal tumor of the ovary occurring in conjunction with Brenner tumor. Gynecol Oncol 77:323–326 Su RM, Chang KC, Chou CY, 2003, Signet-ring stromal tumor of the ovary: a case report. Int J Gynecol Cancer 13:90–93 Vang R, Bague S, Tavassoli FA et al, 2003, Signet-ring stromal tumor of the ovary: clinicopathologic analysis and comparison with Krukenberg tumor. Int J Gynecol Pathol 23:45–51 Deavers MT, Malpica A, Liu J et al, 2003, Ovarian sex cordstromal tumors: an immunohistochemical study including comparison of calretinin and inhibin. Mod Pathol 16:584–590 McCluggage WG, Maxwell P, 2001, Immunohistochemical staining for calretinin is useful in the diagnosis of ovarian sex cordstromal tumors. Histopathology 38:403–408 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 333 EP 336 DI 10.1007/s12253-008-9080-6 PG 4 ER PT J AU Kojima, M Shimizu, K Sameshima, Sh Saruki, N Nakamura, N AF Kojima, Masaru Shimizu, Kazuhiko Sameshima, Shinichi Saruki, Nobuhiro Nakamura, Naoya TI Focal Lymphoid Hyperplasia of the Terminal Ileum Presenting Mantle Zone Hyperplasia with Clear Cytoplasm. A Report of Three Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Focal lymphoid hyperplasia; Terminal ileum; Mantle zone hyperplasia; MALT lymphoma; Immunohistochemistry ID Focal lymphoid hyperplasia; Terminal ileum; Mantle zone hyperplasia; MALT lymphoma; Immunohistochemistry AB We report three unusual cases of focal lymphoid hyperplasia of the ileocecal valve. The gross specimens showed thickening of the ileocecal valve. Low power magnification showed a dense lymphoid infiltrate in the mucosa and submucosa. This condition was characterized by reactive lymphoid follicles with large reactive germinal centers surrounded by a pale cuff of mantle zone lymphocytes presenting a marginal zone distribution pattern. These cells had intermediate- to- medium-sized round or slightly indented nuclei and a broad rim of clear cytoplasm. However, immunohistochemical study demonstrated that both the mantle zone lymphocytes and the pale cuff of the lymphoid cells were CD20+, sIgM+, sIgD+, CD5−, CD10−, CD23−, CD43−, Bcl-2+,Bcl-6−, CyclinD1−. The polytypic nature of these cells was demonstrated by immunohistochemistry and polymerase chain reaction. This unusual mantle cell hyperplasia with clear cytoplasm associated with focal lymphoid hyperplasia in middle-aged and elderly patients should be differentiated from the extranodal marginal zone B-cell lymphoma of mucosa associated lymphoid tissue type or mantle cell lymphoma showing a marginal zone distribution pattern. To avoid overdiagnosis and overtreatment, it is suggested that immunophenotypic and genotypic studies might be required, and careful attention should be paid to the morphologic examination. C1 [Kojima, Masaru] Gunma Cancer Center Hospital, Department of Pathology and Clinical Laboratories, 617-1, Takabayashinishi-cho, 373-8550 Ohta, Japan. [Shimizu, Kazuhiko] Ashikaga Red Cross Hospital, Department of Pathology and Clinical LaboratoriesAshikaga, Japan. [Sameshima, Shinichi] Gunma Cancer Center Hospital, Department of SurgeryOhta, Japan. [Saruki, Nobuhiro] Gunma Cancer Center Hospital, Department of AnesthesiologyOhta, Japan. [Nakamura, Naoya] Tokai University School of Medicine, Department of PathologyIsehara, Japan. RP Kojima, M (reprint author), Gunma Cancer Center Hospital, Department of Pathology and Clinical Laboratories, 373-8550 Ohta, Japan. EM mkojima@gunma-cc.jp CR Isaacson PG, Norton AJ, 1994, Extranodal lymphomas. Churchill Livingstone, Edinburgh Riddell RH, Petras RE, Williams GT et al, 2003, Tumor of the intestine., Atlas of tumor pathology, 3rd series, fascicle 32). Armed Forces Institute of Pathology, Bethesda, MD Rubin A, Isaacson PG, 1990, Florid reactive lymphoid hyperplasia of the terminal ileum in adults: a condition bearing a close resemblance to low-grade malignant lymphoma. Histopathology 17:19–26 Wan JH, Trainor KJ, Brisco MJ et al, 1990, Monoclonality in B cell lymphoma detected in paraffin wax embedded sections using the polymerase chain reaction. J Clin Pathol 43:888–890 Hunt JP, Chan JA, Samoszuk M et al, 2001, Hyperplasia of mantle/marginal zone B cells with clear cytoplasm in peripheral lymph nodes. A clinicopathologic study of 35 cases. Am J Clin Pathol 116:550–559 Weisenburger DD, Duggan MJ, Perry DA et al, 1991, Non- Hodgkin’s lymphoma of mantle zone origin. Pathol Annu 26I:139–158 Wang T, Lasota J, Hanau CA et al, 1995, Bcl-2 oncoprotein is widespread in lymphoid tissue and lymphoma but its differential expression in benign versus malignant follicles and monocytoid B-cell proliferation is of diagnostic value. APMIS 103:655– 662 Lai R, Weiss LM, Chang KL et al, 1999, Frequency of CD43 expression in non-Hodgkin lymphoma. A survey of 742 cases and further characterization of rare CD43+ follicular lymphomas. Am J Clin Pathol 111:488–494 Swerdlow SH, Zukerberg LR, Yang W-I et al, 1996, The morphologic spectrum of non-Hodgkin’s lymphomas with BCL1/cyclinD1 gene rearrangements. Am J Surg Pathol 20:627– 640 Anagnostopoulos I, Foss HD, Hummel M et al, 2001, Extranodal mantle cell lymphoma mimicking marginal zone cell lymphoma. Histopathology 39:561–565 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 337 EP 340 DI 10.1007/s12253-008-9035-y PG 4 ER PT J AU Farkas, A Istok, R Szekely, E Glasz, T Kulka, J AF Farkas, Andrea Istok, Roland Szekely, Eszter Glasz, Tibor Kulka, Janina TI Pigmented Papillary Carcinoma: A Rare Tumor of the Male Breast SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast carcinoma; Male breast; Melanin pigment; Papillary carcinoma; Pigmented papillary carcinoma ID Breast carcinoma; Male breast; Melanin pigment; Papillary carcinoma; Pigmented papillary carcinoma AB Primary melanin pigment containing tumors of the breast are rare. We report a pigmented papillary carcinoma of a 60-year-old male patient who presented a firm mass 1.7 cm in diameter with an ill defined border on ultrasonography behind the mamilla. To the best of our knowledge this is the third case report of this type of tumor in male breast. C1 [Farkas, Andrea] St Imre Hospital, Department of Pathology, 12–16 Tetenyi ut, 1115 Budapest, Hungary. [Istok, Roland] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Szekely, Eszter] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Glasz, Tibor] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Farkas, A (reprint author), St Imre Hospital, Department of Pathology, 1115 Budapest, Hungary. EM drfarkasandrea@gmail.com CR Romanelli R, Toncini C, 1986, Pigmented papillary carcinoma of the male breast. Tumori 72:105–108 Saitoh K, Saga K, Okazaki M et al, 1998, Pigmented primary carcinoma of the breast: a clinical mimic of malignant melanoma. Br J Dermatol 139:287–290 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2008 VL 14 IS 3 BP 341 EP 343 DI 10.1007/s12253-008-9075-3 PG 3 ER PT J AU Mammas, NI Sourvinos, G Giannoudis, A Spandidos, AD AF Mammas, N Ioannis Sourvinos, George Giannoudis, Athena Spandidos, A Demetrios TI Human Papilloma Virus (HPV) and Host Cellular Interactions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Human papilloma virus; HPV; E6; E7; Host cellular proteins; DNA integration ID Human papilloma virus; HPV; E6; E7; Host cellular proteins; DNA integration AB Viral-induced carcinogenesis has been attributed to the ability of viral oncoproteins to target and interact with the host cellular proteins. It is generally accepted that Human papilloma virus (HPV) E6 and E7 function as the dominant oncoproteins of ‘high-risk’ HPVs by altering the function of critical cellular proteins. Initially it was shown that HPV E6 enhances the degradation of p53, while HPV E7 inactivates the function of the retinoblastoma tumor suppressor protein Rb. However, recent studies during the last decade have identified a number of additional host cellular targets of both HPV E6 and E7 that may also play an important role in malignant cellular transformation. In this review we present the interactions of HPV E6 and E7 with the host cellular target proteins. We also present the role of DNA integration in the malignant transformation of the epithelial cell. C1 [Mammas, N Ioannis] University of Crete, Medical School, Department of Virology, 71100 Heraklion, Crete, Greece. [Sourvinos, George] University of Crete, Medical School, Department of Virology, 71100 Heraklion, Crete, Greece. [Giannoudis, Athena] University of Liverpool, Faculty of MedicineLiverpool, UK. [Spandidos, A Demetrios] University of Crete, Medical School, Department of Virology, 71100 Heraklion, Crete, Greece. RP Spandidos, AD (reprint author), University of Crete, Medical School, Department of Virology, 71100 Heraklion, Greece. EM spandidos@spandidos.gr CR zur Hausen H, 2002, Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer 2:342–350 zur Hausen H, 1996, Papillomavirus infections—a major cause of human cancers. Biochim Biophys Acta 1288:F55–F78 Steben M, Duarte-Franco E, 2007, Human papillomavirus infection: epidemiology and pathophysiology. Gynecol Oncol 107:S2–S5 Fehrmann F, Laimins LA, 2003, Human papillomaviruses: targeting differentiating epithelial cells for malignant transformation. Oncogene 22:5201–5207 Huibregtse JM, Scheffner M, Howley PM, 1991, A cellular protein mediates association of p53 with the E6 oncoprotein of human papillomavirus types 16 or 18. EMBO J 10:4129– 4135 Huibregtse JM, Scheffner M, Howley PM, 1993, Localization of the E6-AP regions that direct human papillomavirus E6 binding, association with p53, and ubiquitination of associated proteins. Mol Cell Biol 13:4918–4927 Kuballa P, Matentzoglu K, Scheffner M, 2007, The role of the ubiquitin ligase E6-AP in human papillomavirus E6-mediated degradation of PDZ domain-containing proteins. J Biol Chem 282:65–71 Handa K, Yugawa T, Narisawa-Saito M et al, 2007, E6APdependent degradation of DLG4/PSD95 by high-risk human papillomavirus type 18 E6 protein. J Virol 81:1379–1389 Scheffner M, Huibregtse JM, Vierstra RD, Howley PM, 1993, The HPV-16 E6 and E6-AP complex functions as a ubiquitinprotein ligase in the ubiquitination of p 53. Cell 75:495–505 Dalal S, Gao Q, Androphy EJ, Band V, 1996, Mutational analysis of human papillomavirus type 16 E6 demonstrates that p53 degradation is necessary for immortalization of mammary epithelial cells. J Virol 70:683–688 Kiyono T, Foster SA, Koop JI et al, 1998, Both Rb/p16INK4a inactivation and telomerase activity are required to immortalize human epithelial cells. Nature 396:84–88 Liu Y, Chen JJ, Gao Q et al, 1999, Multiple functions of human papillomavirus type 16 E6 contribute to the immortalization of mammary epithelial cells. J Virol 73:7297–7307 Chen JJ, Reid CE, Band V, Androphy EJ, 1995, Interaction of papillomavirus E6 oncoproteins with a putative calcium-binding protein. Science 269:529–531 Sherman L, Itzhaki H, Jackman A et al, 2002, Inhibition of serum- and calcium-induced terminal differentiation of human keratinocytes by HPV 16 E6: study of the association with p53 degradation, inhibition of p53 transactivation, and binding to E6BP. Virology 292:309–320 Thomas M, Banks L, 1998, Inhibition of Bak-induced apoptosis by HPV-18 E6. Oncogene 17:2943–2954 Thomas M, Banks L, 1999, Human papillomavirus, HPV, E6 interactions with Bak are conserved amongst E6 proteins from high and low risk HPV types. J Gen Virol 80:1513–1517 Nagata S, 1997, Apoptosis by death factor. Cell 88:355–365 Adams JM, Cory S, 2007, The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene 26:1324–1337 Turner CE, 2000, Paxillin interactions. J Cell Sci 113:4139– 4140 Tong X, Howley PM, 1997, The bovine papillomavirus E6 oncoprotein interacts with paxillin and disrupts the actin cytoskeleton. Proc Natl Acad Sci USA 94:4412–4417 Vande Pol SB, Brown MC, Turner CE, 1998, Association of Bovine Papillomavirus Type 1 E6 oncoprotein with the focal adhesion protein paxillin through a conserved protein interaction motif. Oncogene 16:43–52 Kiyono T, Hiraiwa A, Fujita M et al, 1997, Binding of high-risk human papillomavirus E6 oncoproteins to the human homologue of the drosophila discs large tumor suppressor protein. Proc Natl Acad Sci USA 94:11612–11616 Lee SS, Weiss RS, Javier RT, 1997, Binding of human virus oncoproteins to hDlg/SAP97, a mammalian homolog of the Drosophila discs large tumor suppressor protein. Proc Natl Acad Sci USA 94:6670–6675 Nakagawa S, Huibregtse JM, 2000, Human scribble, Vartul, is targeted for ubiquitin-mediated degradation by the high-risk papillomavirus E6 proteins and the E6AP ubiquitin-protein ligase. Mol Cell Biol 20:8244–8253 Liu Y, Herny GD, Hegde RS, Bleja JD, 2007, Solution structure of the hDlg/SAP97PDZ2 domain and its mechanism of interaction with HPV-18 papillomavirus E6 protein. Biochemistry 46:10864–10874 Glaunsinger BA, Lee SS, Thomas M et al, 2000, Interactions of the PDZ-protein MAGI-1 with adenovirus E4-ORF1 and highrisk papillomavirus E6 oncoproteins. Oncogene 19:5270–5280 Thomas M, Laura R, Hepner K et al, 2002, Oncogenic human papillomavirus E6 proteins target the MAGI-2 and MAGI-3 proteins for degradation. Oncogene 21:5088–5096 Zhang Y, Dasgupta J, Ma RZ et al, 2007, Structures of a human papillomavirus, HPV, E6 polypeptide bound to MAGUK proteins: mechanisms of targeting tumor suppressors by a highrisk HPV oncoprotein. J Virol 81:3618–3626 Thomas M, Glaunsinger B, Pim D et al, 2001, HPV E6 and MAGUK protein interactions: determination of the molecular basis for specific protein recognition and degradation. Oncogene 20:5431–5439 Ronco L, Karpova A, Vidal M, Howley PM, 1998, Human papillomavirus 16 E6 oncoprotein binds to interferon regulatory factor-3 and inhibits its transcriptional activity. Genes Dev 12:2061–2072 Lee SH, Kim JW, Lee HW et al, 2003, Interferon regulatory factor-1, IRF-1, is a mediator for interferon-gamma induced attenuation of telomerase activity and human telomerase reverse transcriptase, hTERT, expression. Oncogene 22:381–391 Kuhne C, Banks L, 1998, E3-ubiquitin ligase/E6-AP links multicopy maintenance protein 7 to the ubiquitination pathway by a novel motif, the L2G box. J Biol Chem 273:34302–34309 Zimmermann H, Degenkolbe R, Bernard HU, O’Connor MJ, 1999, The human papillomavirus type 16 E6 oncoprotein can down-regulate p53 activity by targeting the transcriptional coactivator CBP/p300. J Virol 73:6209–6219 Janknecht R, Hunter T, 1996, Transcription. A growing coactivator network. Nature 383:22–23 Giles RH, Peters DJ, Breuning MH, 1998, Conjunction dysfunction: CBP/p300 in human disease. Trends Genet 14:178–183 Gu W, Shi XL, Roeder RG, 1997, Synergistic activation of transcription by CBP and p53. Nature 387:819–823 Avantaggiati ML, Ogryzko V, Gardner K et al, 1997, Recruitment of p300/CBP in p53-dependent signal pathways. Cell 89:1175–1184 Bernat A, Avvakumov N, Mymryk JS, Banks L, 2003, Interaction between the HPV E7 oncoprotein and the transcriptional coactivator p300. Oncogene 22:7871–7881 Klingelhutz AJ, Foster SA, McDougall JK, 1996, Telomerase activation by the E6 gene product of human papillomavirus type 16. Nature 380:79–82 James MA, Lee JH, Klingelhutz AZ, 2006, HPV 16-E6 associated hTERT promoter acetylation is E6AP dependent, increased in later passage cells and enhanced by loss of p300. Int J Cancer 119:1878–1885 Zhang H, Jin Y, Chen X et al, 2007, Papillomavirus type 16 E6/ E7 and human telomerase reverse transcriptase in esophageal cell immortalization and early transformation. Cancer Lett 245:184– 194 Greider CW, 1998, Telomerase activity, cell proliferation, and cancer. Proc Natl Acad Sci USA 5:90–92 Holt SE, Shay JW, Wright WE, 1996, Refining the telomere– telomerase hypothesis of aging and cancer. Nat Biotechnol 4:836–839 Altschuler DL, Ribeiro-Neto F, 1998, Mitogenic and oncogenic properties of the small G protein Rap1b. Proc Natl Acad Sci USA 95:7475–7479 Gao Q, Singh L, Kumar A et al, 2001, Human papillomavirus type 16 E6-induced degradation of E6TP1 correlates with its ability to immortalize human mammary epithelial cells. Virology 75:4459–4466 Gao Q, Srinivasan S, Boyer S et al, 1999, The E6 oncoproteins of high-risk papillomaviruses bind to a novel putative GAP protein, E6TP1, and target it for degradation. Mol Cell Biol 19:733–744 Singh L, Gao Q, Kumar A et al, 2003, The high-risk human papillomavirus type 16 E6 counters the GAP function of E6TP1 toward small Rap G proteins. J Virol 77:1614–1620 Lee C, Wooldridge TR, Laimins LA, 2007, Analysis of the roles of E6 binding to E6TP1 and nuclear localization in the human papillomavirus type 31 life cycle. Virology 358:201– 210 Gao Q, Kumar A, Singh L et al, 2002, Human papillomavirus E6-induced degradation of E6TP1 is mediated by E6AP ubiquitin ligase. Cancer Res 62:3315–3321 Gao Q, Kumar A, Srinivasan S et al, 2000, PKN binds and phosphorylates human papillomavirus E6 oncoprotein. J Biol Chem 275:14824–14830 Degenhardt YY, Silverstein J, 2001, Gps2, a protein partner for human papillomavirus E6 proteins. J Virol 75:151–160 Gross-Mesilaty S, Reinstein E, Bercovich B et al, 1998, Basal and human papillomavirus E6 oncoprotein-induced degradation of Myc proteins by the ubiquitin pathway. Proc Natl Acad Sci USA 95:8058–8063 Wang YW, Chang HS, Lin CH, Yu WC, 2007, HPV-18 E7 conjugates to c-Myc and mediates its transcriptional activity. Int J Biochem Cell Biol 39:401–412 Liu X, Disbrow GL, Yuan H et al, 2007, Myc and human papillomavirus type 16 E7 genes cooperate to immortalize human keratinocytes. J Virol 81:12689–12695 McMurray HR, McCance DJ, 2003, Human papiloomavirus type 16 E6 activates TERT gene transcription through induction of c-Myc and release of USF-mediated repression. J Virol 77:9852–9861 Li S, Labrecque S, Gauzzi MC et al, 1999, The human papilloma virus, HPV)-18 E6 oncoprotein physically associates with Tyk2 and impairs Jak-STAT activation by interferon-a. Oncogene 18:5727–5737 Lee SS, Glaunsinger B, Mantovani F et al, 2000, Multi-PDZ domain protein MUPP1 is a cellular target for both adenovirus E4-ORF1 and high-risk papillomavirus type 18 E6 oncoproteins. J Virol 74:9680–9693 Tsutsui T, Kumakura S, Yamamoto A et al, 2002, Association of p16(INK4a, and pRb inactivation with immortalization of human cells. Carcinogenesis 23:2111–2117 Mammas IN, Zafiropoulos A, Sifakis S et al, 2005, Human papillomavirus, HPV, typing in relation to ras oncogene mRNA expression in HPV-associated human squamous cervical neoplasia. Int J Biol Markers 20:257–263 Schreiber K, Cannon RE, Karrison RE et al, 2004, Strong synergy between mutant ras and HPV16 E6/E7 in the development of primary tumors. Oncogene 23:3972–3979 Felsani A, Mileo AM, Paggi MG, 2006, Retinoblastoma family proteins as key targets of the small DNA virus oncoproteins. Oncogene 25:5277–5285 Lavia P, Jansen-Durr P, 1999, E2F target genes and cell-cycle checkpoint control. Bioessays 21:221–230 Caldeira S, de Villiers EM, Tommasino M, 2000, Human papillomavirus E7 proteins stimulate proliferation independently of their ability to associate with retinoblastoma protein. Oncogene 19:821–826 Dyson N, Howley PM, Munger K, Harlow E, 1989, The human papillomavirus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product. Science 243:934–937 Gonzalez SL, Stremlau M, He X et al, 2001, Degradation of the retinoblastoma tumor suppressor by the human papillomavirus type 16 E7 oncoprotein is important for functional inactivation and is separable from proteasomal degradation of E7. J Virol 75:7583–7591 Heck DV, Yee CL, Howley PM, Munger K, 1992, Efficiency of binding to the retinoblastoma protein correlates with the transforming capacity of the E7 oncoproteins of the human papillomaviruses. Proc Natl Acad Sci USA 89:4442–4446 Chellappan S, Kraus VB, Kroger B et al, 1992, Adenovirus E1A, simian virus 40 tumor antigen, and human papillomavirus E7 protein share the capacity to disrupt the interaction between the transcription factor E2F and the retinoblastoma gene product. Proc Natl Acad Sci USA 89:4549–4553 Edmonds C, Vousden KH, 1989, A point mutational analysis of human papillomavirus type 16 E7 protein. J Virol 63:2650–2656 Strati K, Lambert PF, 2007, Role of Rb-dependent and Rbindependent functions of papillomavirus E7 oncogene in head and neck cancer. Cancer Res 67:11585–11593 Zerfass K, Levy LM, Cremonesi C et al, 1995, Cell cycledependent disruption of E2F-p107 complexes by human papillomavirus type 16 E7. J Gen Virol 76:1815–1820 Davies R, Hicks R, Crook T et al, 1993, Human papillomavirus type 16 E7 associates with a histone H1 kinase and with p107 through sequences necessary for transformation. J Virol 67:2521–2528 Roman A, 2006, The human papillomavirus E7 protein shines a spotlight on the pRB family member, p130. Cell Cycle 5:567– 568 Zhang B, Chen W, Roman A, 2006, The E7 proteins of low- and high-risk human papillomaviruses share the ability to target the pRB family member p130 for degradation. Proc Natl Acad Sci USA 103:437–442 Grana X, Garriga J, Mayol X, 1998, Role of the retinoblastoma protein family, pRB, p107 and p130 in the negative control of cell growth. Oncogene 17:3365–3383 Stubdal H, Zalvide J, Campbell KS et al, 1997, Inactivation of pRB-related proteins p130 and p107 mediated by the J domain of simian virus 40 large T antigen. Mol Cell Biol 17:4979–4990 Bruce JL, Hurford RK, Classon J et al, 2000, Requirements for cell cycle arrest by p16ink4a. Mol Cell 6:737–742 Dyson N, Dembski M, Fattaey A et al, 1993, Analysis of p107- associated proteins: p107 associates with a form of E2F that differs from pRB-associated E2F-1. J Virol 67:7641–7647 Arroyo M, Bagchi S, Raychaudhuri P, 1993, Association of the human papillomavirus type 16 E7 protein with the S-phasespecific E2F–cyclin A complex. Mol Cell Biol 13:6537–6546 Tommasino M, Adamczewski JP, Carlotti F et al, 1993, HPV16 E7 protein associates with the protein kinase p33CDK2 and cyclin A. Oncogene 8:195–202 McIntyre MC, Ruesch MN, Laimins LA, 1996, Human papillomavirus E7 oncoproteins bind a single form of cyclin E in a complex with cdk2 and p107. Virology 215:73–82 Vogt B, Zerfass-Thome K, Schulze A et al, 1999, Regulation of cyclin E gene expression by the human papillomavirus type 16 E7 oncoprotein. J Gen Virol 80:2103–2113 Balsitis S, Dick F, Dyson N, Lambert PF, 2006, Critical roles for non-pRB targets of human papillomavirus type 16 E7 in cervical carcinogenesis. Cancer Res 66:9393–9400 Zerfass K, Schulze A, Spitkovsky D et al, 1995, Sequential activation of cyclin E and cyclin A gene expression by human papillomavirus type 16 E7 through sequences necessary for transformation. J Virol 69:6389–6399 He W, Staples D, Smith C, Fisher C, 2003, Direct activation of cyclin-dependent kinase 2 by human papillomavirus E7. J Virol 77:10566–10574 Duensing S, Duensing A, Lee DC et al, 2004, Cyclin-dependent kinase inhibitor indirubin-3¢-oxime selectively inhibits human papillomavirus type 16 E7-induced numerical centrosome anomalies. Oncogene 23:8206–8215 Massimi P, Pim D, Storey A, Banks L, 1996, HPV-16 E7 and adenovirus E1a complex formation with TATA box binding protein is enhanced by casein kinase II phosphorylation. Oncogene 12:2325–2330 Maldonado E, Cabrejos ME, Banks L, Allende JE, 2002, Human papillomavirus-16 E7 protein inhibits the DNA interaction of the TATA binding transcription factor. J Cell Biochem 85:663–669 Berezutskaya E, Bagchi S, 1997, The human papillomavirus E7 oncoprotein functionally interacts with the S4 subunit of the 26 S proteasome. J Biol Chem 272:30135–30140 Coux O, Tanaka K, Goldberg AL, 1996, Structure and functions of the 20S and 26S proteasomes. Annu Rev Biochem 65:801–847 Nead MA, Baglia LA, Antimore MJ et al, 1998, Rb binds c-Jun and activates transcription. EMBO J 17:2342–2352 Nishitani J, Nishinaka T, Cheng CH et al, 1999, Recruitment of the retinoblastoma protein to c-Jun enhances transcription activity mediated through the AP-1 binding site. J Biol Chem 274:5454–5461 Schilling B, De-Medine T, Syken J et al, 1998, A novel human DnaJ protein, hTid-1, a homolog of the Drosophila tumor suppressor protein Tid56, can interact with the human papillomavirus type 16 E7 oncoprotein. Virology 247:74–85 Zwerschke W, Mazurek S, Massimi P et al, 1999, Modulation of type M2 pyruvate kinase activity by the human papillomavirus type 16 E7 oncoprotein. Proc Natl Acad Sci USA 96:1291–1296 Mazurek S, Zwerschke W, Jansen-Durr P, Eigenbrodt E, 2001, Metabolic cooperation between different oncogenes during cell transformation: interaction between activated ras and HPV-16 E7. Oncogene 20:6891–6898 Barnard P, McMillan NA, 1999, The human papillomavirus E7 oncoprotein abrogates signaling mediated by interferon-alpha. Virology 259:305–313 Jones DL, Alani RM, Munger K, 1997, The human papillomavirus E7 oncoprotein can uncouple cellular differentiation and proliferation in human keratinocytes by abrogating p21Cip1- mediated inhibition of cdk2. Genes Dev 11:2101–2111 Funk JO,Waga S, Harry J et al, 1997, Inhibition of CDKactivity and PCNA-dependent DNA replication by p21 is blocked by interaction with the HPV-16 E7 oncoprotein. Genes Dev 11:2090–2100 Zehbe I, Ratsch A, Alumni-Fabbroni M et al, 1999, Overriding of cyclin-dependent kinase inhibitors by high and low risk human papillomavirus types: evidence for an in vivo role in cervical lesions. Oncogene 18:2201–2211 Charette ST, McCance DJ, 2007, The E7 protein from human papillomavirus type 16 enhances keratinocyte migration in an Akt-dependent manner. Oncogene 26:7386–7390 Westbrook TF, Nguyen DX, Trash BR, McCance DJ, 2002, E7 abolishes raf-induced arrest via mislocalization of p21(Cip1). Mol Cell Biol 22:7041–7052 Um SJ, Rhyu JW, Kim EJ et al, 2002, Abrogation of IRF-1 response by high-risk HPV E7 protein in vivo. Cancer Lett 179:205–212 Alazawi W, Pett M, Arch B et al, 2002, Changes in cervical keratinocyte gene expression associated with integration of human papillomavirus 16. Cancer Res 62:6959–6965 Klaes R, Woerner SM, Ridder R et al, 1999, Detection of highrisk cervical intraepithelial neoplasia and cervical cancer by amplification of transcripts derived from integrated papillomavirus oncogenes. Cancer Res 59:6132–6136 Giannoudis A, van Duin M, Snijder PJF, Herrington CS, 2001, Variation in the E2-binding domain of HPV 16 is associated with high-grade squamous intraepithelial lesions of the cervix. Br J Cancer 84:1058–1063 Pett M, Coleman N, 2007, Integration of high-risk human papillomavirus: a key event in cervical carcinogenesis? J Pathol 212:356–367 Jeon S, Lambert PF, 1995, Integration of human papillomavirus type 16 DNA into the human genome leads to increased stability of E6 and E7 mRNAs: implications for cervical carcinogenesis. Proc Natl Acad Sci USA 92:1654–1658 Durst M, Croce CM, Gissmann L et al, 1987, Papillomavirus sequences integrate near cellular oncogenes in some cervical carcinomas. Proc Natl Acad Sci USA 84:1070–1074 Ferber MJ, Thorland EC, Brink AA et al, 2003, Preferential integration of human papillomavirus type 18 near the c-myc locus in cervical carcinoma. Oncogene 22:7233–7242 Peter M, Rosty C, Couturier J et al, 2006, MYC activation associated with the integration of HPV DNA at the MYC locus in genital tumors. Oncogene 25:5985–5993 Ferber MJ, Montoya DP, Yu C et al, 2003, Integrations of the hepatitis B virus, HBV, and human papillomavirus, HPV, into the human telomerase reverse transcriptase, hTERT, gene in liver and cervical cancers. Oncogene 22:3813–3820 Ziegert C, Wentzensen N, Vinokurova S et al, 2003, A comprehensive analysis of HPV integration loci in anogenital lesions combining transcript and genome-based amplification techniques. Oncogene 22:3977–3398 Wentzensen N, Vinokurova S, von Knebel DM, 2004, Systematic review of genomic integration sites of human papillomavirus genomes in epithelial dysplasia and invasive cancer of the female lower genital tract. Cancer Res 64:3878–3884 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 345 EP 354 DI 10.1007/s12253-008-9056-6 PG 10 ER PT J AU Gasperov, MN Sabol, I Matovina, M Spaventi, Grce, M AF Gasperov, Milutin Nina Sabol, Ivan Matovina, Mihaela Spaventi, Sime Grce, Magdalena TI Detection and Typing of Human Papillomaviruses Combining Different Methods: Polymerase Chain Reaction, Restriction Fragment Length Polymorphism, Line Probe Assay and Sequencing SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human papillomavirus; Consensus polymerase chain reaction; Restriction fragment length polymorphism; Line probe assay; Sequencing; Type-specific polymerase chain reaction ID Human papillomavirus; Consensus polymerase chain reaction; Restriction fragment length polymorphism; Line probe assay; Sequencing; Type-specific polymerase chain reaction AB The identification of the etiological factor of many cervical precancerous lesions and cervical cancer, the human papillomavirus (HPV) is widely used. In this study, we evaluated the consensus and type-specific (TS) polymerase chain reaction (PCR), restriction fragment length polymorphism (RFLP), line probe assay (LiPA, Innogenetics) and sequencing to determine the HPV types in cervical specimens. Out of 690 High-grade Squamous Intraepithelial Lesion (HSIL) samples, 86.7% were HPV positive and 13.3% HPV negative by consensus primers (MY09/MY11, L1C1/L1C2-1/L1C2-2 and/or GP5/6) directed PCR. Out of 598 HPV positive samples, 85.3%were typed by TS-PCR being HPV 6/11, 16, 18, 31 and/ or 33, while 14.7% remained untyped. Themost prevalent HPV type in the study group was HPV 16, identified in 35.5% cases, while HPV 31 was the second most frequent HPV type with a prevalence of 10.5%. They were followed by HPV types 6/11, 33 and 18 with a prevalence of 7.4%, 6.2% and 4.9%, respectively. Multiple HPV infections with two or more HPV types (6/11, 16, 18, 31 and/or 33) were found in 9.4% cases. A subset of 88 samples was further typed by RFLP and LiPA to determine the rare HPV types in HSIL samples. The most frequent low abundant HPV types in single infections in decreasing order were HPV 53, 58, 66, 56 and 52, while HPV 51 was the most frequent low abundant HPV type found in multiple HPV infections. Multiple HPV infections were mostly found by LiPA in 27.3% cases versus 14.8% cases found by RFLP. The perfect agreement between RFLP and LiPA assay pair was observed only for HPV types 16, 18, 34 and 59 (kappa value of 1). For other HPV types, the inter-assay agreement ranged from very good to no agreement indicating that neither assay is perfect. Sequencing was performed for 33 samples in cases where both RFLP and LiPA were inconclusive. Sequencingwas shown to be a very goodmethod in case of single HPV infection but not applicable in case of multiple HPV infections. Both RFLP and LiPA are good assays for epidemiological studies, although RFLP being cumbersome and time-consuming is less applicable than LiPA. Careful consideration has to be made before the implementation of either HPV typing methods in clinical laboratories. C1 [Gasperov, Milutin Nina] Rudjer Boskovic Institute, Department of Molecular MedicineZagreb, Croatia. [Sabol, Ivan] Rudjer Boskovic Institute, Department of Molecular MedicineZagreb, Croatia. [Matovina, Mihaela] Rudjer Boskovic Institute, Department of Molecular MedicineZagreb, Croatia. [Spaventi, Sime] Croatian Academy of Science and ArtsZagreb, Croatia. [Grce, Magdalena] Rudjer Boskovic Institute, Department of Molecular MedicineZagreb, Croatia. RP Grce, M (reprint author), Rudjer Boskovic Institute, Department of Molecular Medicine, Zagreb, Croatia. EM grce@irb.hr CR zur Hausen H, 2002, Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer 2:342–350 Munoz N, Castellsague X, de Gonzalez AB et al, 2006, Chapter 1: HPV in the etiology of human cancer. Vaccine 24:S1–S10 Gillison ML, Lowy DR, 2004, A causal role for human papillomavirus in head and neck cancer. Lancet 363:1488–1489 de Villiers EM, Fauquet C, Broker TR et al, 2004, Classification of papillomaviruses. Virology 324:17–27 Walboomers JM, Jacobs MV, Manos MM et al, 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189:12–19 Munoz N, Bosch FX, de Sanjose S et al, 2003, Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med 348:518–527 Munoz N, Bosch FX, Castellsague X et al, 2004, Against which human papillomavirus types shall we vaccinate and screen? The international perspective. Int J Cancer 111:278–285 Grce M, Husnjak K, Bozikov J et al, 2001, Evaluation of genital human papillomavirus infections by polymerase chain reaction among Croatian women. Anticancer Res 21:579–584 Bosch FX, Lorincz A, Munoz N et al, 2002, The causal relation between human papillomavirus and cervical cancer. J Clin Pathol 55:244–265 Iftner T, Villa LL, 2003, Chapter 12: Human papillomavirus technologies. J Natl Cancer Inst Monogr 31:80–88 van den Brule AJ, Meijer CJ, Bakels V et al, 1990, Rapid detection of human papillomavirus in cervical scrapes by combined general primer-mediated and type-specific polymerase chain reaction. J Clin Microbiol 28:2739–2743 Soler C, Allibert P, Chardonnet Yet al, 1991, Detection of human papillomavirus types 6, 11, 16 and 18 in mucosal and cutaneous lesions by the multiplex polymerase chain reaction. J Virol Methods 35:143–157 Gravitt PE, Peyton CL, Apple RJ et al, 1998, Genotyping of 27 human papillomavirus types by using L1 consensus PCR products by a single-hybridization, reverse line blot detection method. J Clin Microbiol 36:3020–3027 Kleter B, van Doorn LJ, Schrauwen L et al, 1999, Development and clinical evaluation of a highly sensitive PCR-reverse hybridization line probe assay for detection and identification of anogenital human papillomavirus. J Clin Microbiol 37:2508–2517 van den Brule AJ, Pol R, Fransen-Daalmeijer N et al, 2000, GP5 +/6+PCR followed by reverse line blot analysis enables rapid and high-throughput identification of human papillomavirus genotypes. J Clin Microbiol 40:779–787 Ovanin-Rakic A, Pajtler M, Stankovic T et al, 2003, [Classification of cervical cytological smears “Zagreb 2002”.Modification of the classification “Zagreb 1990” and “NCI Bethesda 2001”]. Gynaecol Perinatol 12:148–153, In Croatian) Milutin-Gasperov N, Sabol I, Halec G et al, 2007, Retrospective study of the prevalence of high-risk human papillomaviruses among Croatian women. Coll Antropol Suppl 2:89–96 Maniatis T, Frisch EF, Sambrook J, eds,, 1989, Molecular cloning: a laboratory manual. Cold Spring Harbor Laboratory, Cold Spring Harbor, New York Grce M, Husnjak K, Magdic L et al, 1997, Detection and typing of human papillomaviruses by polymerase chain reaction in cervical scrapes of Croatian women with abnormal cytology. Eur J Epidemiol 13:645–651 Husnjak K, Grce M, Magdic L et al, 2000, Comparison of five different polymerase chain reaction methods for detection of human papillomavirus in cervical cell specimens. J Virol Methods 88:125–134 Bell DA, Taylor JA, Paulson DF et al, 1993, Genetic risk and carcinogen exposure: a common inherited defect of the carcinogenmetabolism gene glutathione S-transferase M1, GSTM1, that increases susceptibility to bladder cancer. J Natl Cancer Inst 85:1159–1164 Ting Y, Manos MM, 1990, Detection and typing of genital human papillomaviruses. In: Innis MA, Gelfand DH, Sninsky JJ, White TJ, eds, PCR protocols: a guide to methods and applications. Academic, San Diego, p 356 Meyer T, Arndt R, Stockfleth E et al, 1995, Strategy for typing human papillomaviruses by RFLP analysis of PCR products and subsequent hybridization with a generic probe. Biotechniques 19:632–639 Bernard HU, Chan SY, Manos MM et al, 1994, Identification and assessment of known and novel human papillomaviruses by polymerase chain reaction amplification, restriction fragment length polymorphisms, nucleotide sequence, and phylogenetic algorithms. J Infect Dis 170:1077–1085 Quint WG, Scholte G, van Doorn LJ et al, 2001, Comparative analysis of human papillomavirus infections in cervical scrapes and biopsy specimens by general SPF(10, PCR and HPV genotyping. J Pathol 194:51–58 Altschul SF, Gish W, Miller W et al, 1990, Basic local alignment search tool. J Mol Biol 215:403–410 Clifford GM, Smith JS, Aguado T et al, 2003, Comparison of HPV type distribution in high-grade cervical lesions and cervical cancer: a meta-analysis. Br J Cancer 89:101–105 Smith JS, Lindsay L, Hoots B et al, 2007, Human papillomavirus type distribution in invasive cervical cancer and high-grade cervical lesions: a meta-analysis update. Int J Cancer 121:621– 632 Perrons C, Kleter B, Jelley R et al, 2002, Detection and genotyping of human papillomavirus DNA by SPF10 and MY09/11 primers in cervical cells taken from women attending a colposcopy clinic. J Med Virol 67:246–252 van Doorn LJ, Quint W, Kleter B et al, 2002, Genotyping of human papillomavirus in liquid cytology cervical specimens by the PGMY line blot assay and the SPF(10, line probe assay. J Clin Microbiol 40:979–983 Gillio-Tos A, De Marco L, Ghisetti V et al, 2006, Human papillomavirus typing with GP5+/6+polymerase chain reaction reverse line blotting and with commercial type-specific PCR kits. J Clin Virol 36:126–132 Sabol I, Salakova M, Smahelova J et al, 2008, Evaluation of different techniques for the identification of human papillomavirus types of low prevalence. J Clin Microbiol 46:1606–1613, DOI 10.1128/JCM.02328-07 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 355 EP 363 DI 10.1007/s12253-008-9084-2 PG 9 ER PT J AU Tao, XH Shen, Jg Pan, Wl Dong, Ye Meng, Q Honn, VK Jin, R AF Tao, Xiao-Hua Shen, Jian-gen Pan, Wei-li Dong, Yu-e Meng, Qun Honn, V Kenneth Jin, Rongxian TI Significance of SHP-1 and SHP-2 Expression in Human Papillomavirus Infected Condyloma acuminatum and Cervical Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; Condyloma acuminatum; Human papillomavirus; Protein tyrosine phosphatase ID Cervical cancer; Condyloma acuminatum; Human papillomavirus; Protein tyrosine phosphatase AB Human papillomaviruses (HPVs) are a group of DNA viruses that infect the skin and mucous membranes. Type HPV6/11 is closely related to Condyloma acuminatum, while HPV16/18 is the principal cause of cervical cancer. In this study, we examined the expression of protein tyrosine phosphatases SHP-1 and SHP-2 in Condyloma acuminatum, cervical cancer and the relationship between SHP-1/SHP2 expression and HPV infection. Forty Condyloma acuminatum cases, 20 cervical cancer cases and 20 normal human foreskins were examined for HPV infection by in situ hybridization and the expression of SHP-1 and SHP-2 were examined by immunohistochemistry. Results demonstrated that positive expression rates of HPV6/11, HPV16/18, and HPV31/33 were 98%, 10%, and 7.5% in Condyloma acuminatum, 10%, 85%, and 25% in cervical cancer. Only one normal foreskin demonstrated positive staining for HPV16/18. Positive expression rates of SHP-1 and SHP-2 were 80% and 85% in Condyloma acuminatum, 85% and 90% in cervical cancer. The SHP-1 and SHP-2 expressions were mainly distributed in the prickle layer of Condyloma acuminatum and were diffusely distributed in cervical cancer cells. Only 35% and 30% of foreskins demonstrated weak staining in the basal layer cells. There were statistically significant correlations among the infection of HPV and the expression of SHP-1 and SHP-2 in both Condyloma acuminatum and cervical cancer (P<0.05). SHP-1 expression has a positive correlation with SHP-2 expression. Our results demonstrate putative roles of SHP-1 and SHP-2 in the progression of both Condyloma acuminatum and cervical cancer after HPV infection. C1 [Tao, Xiao-Hua] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of PathologyHangzhou, China. [Shen, Jian-gen] Medical College of Zhejiang University, Research Institute of ImmunityHangzhou, China. [Pan, Wei-li] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of PathologyHangzhou, China. [Dong, Yu-e] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of PathologyHangzhou, China. [Meng, Qun] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of PathologyHangzhou, China. [Honn, V Kenneth] Wayne State University, Department of Pathology, 430 Chemistry Building, 48202 Detroit, MI, USA. [Jin, Rongxian] Wayne State University, Department of Pathology, 430 Chemistry Building, 48202 Detroit, MI, USA. RP Jin, R (reprint author), Wayne State University, Department of Pathology, 48202 Detroit, USA. EM rongxianj@wayne.edu CR zur Hausen H, 1996, Roots and perspectives of contemporary papillomavirus research. J Cancer Res Clin Oncol 122:3–13 Dupin N, 2004, Genital warts. Clin Dermatol 22:481–486 Leung AK et al, 2005, Genital infection with human papillomavirus in adolescents. Adv Ther 22:187–197 Moscicki AB, 2005, Impact of HPV infection in adolescent populations. J Adolesc Health 37:S3–S9 Bryan JT, 2007, Developing an HPV vaccine to prevent cervical cancer and genital warts. Vaccine 25:3001–3006 Walboomers JM et al, 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189:12–19 Denu JM et al, 1996, Form and function in protein dephosphorylation. Cell 87:361–364 Hunter T, 2000, Signaling—2000 and beyond. Cell 100:113–127 Tonks NK, Neel BG, 1996, From form to function: signaling by protein tyrosine phosphatases. Cell 87:365–368 Yu WM et al, 2003, Catalytic-dependent and -independent roles of SHP-2 tyrosine phosphatase in interleukin-3 signaling. Oncogene 22:5995–6004 Adachi M et al, 1996, Mammalian SH2-containing protein tyrosine phosphatases. Cell 85:15 Su MWet al, 2001, Targeting Src homology 2 domain-containing tyrosine phosphatase, SHP-1, into lipid rafts inhibits CD3- induced T cell activation. J Immunol 166:3975–3982 Huang Q et al, 2002, The novel role of the C-terminal region of SHP-2. Involvement of Gab1 and SHP-2 phosphatase activity in Elk-1 activation. J Biol Chem 277:29330–29341 Ahmad S et al, 1993, A widely expressed human protein-tyrosine phosphatase containing src homology 2 domains. Proc Natl Acad Sci U S A 90:2197–2201 Feng GS et al, 1993, SH2-containing phosphotyrosine phosphatase as a target of protein-tyrosine kinases. Science 259:1607–1611 Vogel Wet al, 1993, Activation of a phosphotyrosine phosphatase by tyrosine phosphorylation. Science 259:1611–1614 Klingmuller U et al, 1995, Specific recruitment of SH-PTP1 to the erythropoietin receptor causes inactivation of JAK2 and termination of proliferative signals. Cell 80:729–738 Hsu HC et al, 2001, Mutation of the hematopoietic cell phosphatase, Hcph, gene is associated with resistance to gamma-irradiationinduced apoptosis in Src homology protein tyrosine phosphatase, SHP)-1-deficient “motheaten” mutant mice. J Immunol 166:772– 780 Ke Y et al, 2007, Deletion of Shp2 in the brain leads to defective proliferation and differentiation in neural stem cells and early postnatal lethality. Mol Cell Biol 27:6706–6717 Voena C et al, 2007, The tyrosine phosphatase Shp2 interacts with NPM-ALK and regulates anaplastic lymphoma cell growth and migration. Cancer Res 67:4278–4286 Xu R, 2007, Shp2, a novel oncogenic tyrosine phosphatase and potential therapeutic target for human leukemia. Cell Res 17:295–297 Gillison ML et al, 2008, Distinct risk factor profiles for human papillomavirus type 16-positive and human papillomavirus type 16-negative head and neck cancers. J Natl Cancer Inst 100:407– 420 McDonald JS et al, 1996, nm23-H1 expression in squamous cell carcinoma of the head and neck. Pathol Oncol Res 2:34–36 Skerlev M et al, 2006, PL9 The HPV-genital, and oral?, infections—how are we standing now? Oral Dis 12(Suppl 1):2 Richter ON et al, 2003, Bowenoid papulosis of the vulvaimmunotherapeutical approach with topical imiquimod. Arch Gynecol Obstet 268:333–336 Wigbels B et al, 2001, Imiquimod: a new treatment possibility in bowenoid papulosis? Hautarzt 52:128–131 Wang KL, 2007, Human papillomavirus and vaccination in cervical cancer. Taiwan J Obstet Gynecol 46:352–362 Yoneta A et al, 2000, Development of squamous cell carcinoma by two high-risk human papillomaviruses, HPVs), a novel HPV- 67 and HPV-31 from bowenoid papulosis. Br J Dermatol 143:604–608 Wu C et al, 2003, The function of the protein tyrosine phosphatase SHP-1 in cancer. Gene 306:1–12 Zapata PD et al, 2002, Autocrine regulation of human prostate carcinoma cell proliferation by somatostatin through the modulation of the SH2 domain containing protein tyrosine phosphatase, SHP)-1. J Clin Endocrinol Metab 87:915–926 Zapata PD et al, 2004, Phosphotyrosine phosphatase SHP-1, somatostatin and prostate cancer. Actas Urol Esp 28:269–285 Massa PT et al, 2002, Critical role for protein tyrosine phosphatase SHP-1 in controlling infection of central nervous system glia and demyelination by Theiler’s murine encephalomyelitis virus. J Virol 76:8335–8346 Manes S et al, 1999, Concerted activity of tyrosine phosphatase SHP-2 and focal adhesion kinase in regulation of cell motility. Mol Cell Biol 19:3125–3135 Yu DH et al, 1998, Protein-tyrosine phosphatase Shp-2 regulates cell spreading, migration, and focal adhesion. J Biol Chem 273:21125–21131 Coyne CB et al, 2007, Poliovirus entry into human brain microvascular cells requires receptor-induced activation of SHP- 2. EMBO J 26:4016–4028 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 365 EP 371 DI 10.1007/s12253-008-9065-5 PG 7 ER PT J AU Liu, Y Xu, LM Zhong, HH Heng, JW Wu, QB AF Liu, Yan Xu, Lin Mei Zhong, Hao Hao Heng, Jie Wan Wu, Quan Bing TI EGFR Mutations are More Frequent in Well-Differentiated than in Poor-Differentiated Lung Adenocarcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chemotherapy; Epidermal growth factor receptor; Lung adenocarcinoma; Polymerase chain reaction; Sequence analysis ID Chemotherapy; Epidermal growth factor receptor; Lung adenocarcinoma; Polymerase chain reaction; Sequence analysis AB Somatic mutations in epidermal growth factor receptor (EGFR) tyrosine kinase domain, particularly deletions in exon 19 and point mutation in exon 21, are associated with clinical outcome in patients with lung adenocarcinoma, suggesting that EGFR mutation would have an important role in clinical decision making. DNA was extracted from the excised specimens of 60 lung adenocarcinoma patients with phenol-chloroform and ethanol precipitation. Exon 19 and 21 were amplified by PCR, and direct sequenced from both sense and antisense directions. EGFR somatic mutations were present in 13 of 60 patients (21.67%), including seven cases of in-frame deletion in exon 19 around codon 746 and six cases of amino acid substitution in exon 21. Exon 21 mutation is more frequent in adenocarcinomas with bronchi-alveolar component than exon 19 deletions. Mutations were more prevalent in well-differentiated adenocarcinomas (9/27, 33.33%) than in moderate to poor-differentiated adenocarcinomas (4/33, 12.12%) (P<0.05). Adenocarcinomas with bronchi-alveolar components had higher mutation frequency (8/22,36. 36%) than those without bronchi-alveolar components (5/38, 13.16%) (P<0.05). In this study, female patients had more mutation rate than male patients. This trend was also observed in the patients with pathologic stage I–II compared with stage III–IV, but neither of them was statistically significant. Patients with cisplatin-based adjuvant chemotherapy had no significantly prolonged survival compared with single radical resection. But patients with EGFR mutation had relative longer survival. In conclusion, our study suggest that EGFR mutations may be a valuable prognostic factor for disease free survival of surgically treated lung adenocarcinoma patients independently from adjuvant chemotherapy. C1 [Liu, Yan] Health Science Center, Peking University, Department of Pathology, 100191 Beijing, China. [Xu, Lin Mei] Tianjin Chest Hospital, Department of Pathology, 300051 Tianjin, China. [Zhong, Hao Hao] Health Science Center, Peking University, Department of Pathology, 100191 Beijing, China. [Heng, Jie Wan] Health Science Center, Peking University, Department of Pathology, 100191 Beijing, China. [Wu, Quan Bing] Health Science Center, Peking University, Department of Pathology, 100191 Beijing, China. RP Liu, Y (reprint author), Health Science Center, Peking University, Department of Pathology, 100191 Beijing, China. EM laylaly@126.com CR Paez JG, Janne PA, Lee JC et al, 2004, EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304:1497–1500 Lynch TJ, Bell DW, Sordella R et al, 2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small cell lung cancer to gefitinib. N Engl JMed 350:2129–2139 Mitsudomi T, Kosaka T, Endoh H et al, 2005, Mutations of the epidermal growth factor receptor gene predict prolonged survival after gefitinib treatment in patients with non-small-cell lung cancer with postoperative recurrence. J Clin Oncol 23:2513–2520 Qin BM, Chen X, Zhu JD et al, 2005, Identification of EGFR kinase domain mutations among lung cancer patients in China: implication of targeted cancer therapy. Cell Res 15:212–217 Yang SH, Mechanic LE, Yang P et al, 2005, Mutations in tyrosine kinase domain of the epidermal growth factor receptor in nonsmall- cell lung cancer. Clin Cancer Res 11:2106–2110 Rosell R, Ichinose Y, TaronMet al, 2005, Mutations in the tyrosine kinase domain of the EGFR gene associated with gefitinib response in non-small cell lung cancer. Lung Cancer 50:25–33 Sonobe M, Manabe T, Wada H et al, 2005, Mutations in the EGFR gene are linked to smoking-independent, lung adenocarcinoma. Br J Cancer 93(3):355–363 Heneda H, Sasaki H, Shimizu S et al, 2006, Epidermal growth factor receptor gene mutation defines distinct subsets among small adenocarcinomas of the lung. Lung Cancer 52:47–52 Chan SK, Gullick WJ, Hill ME, 2006, Mutations of the epidermal growth factor receptor in non-small cell lung cancer—search and destroy. Eup J Cancer 42:17–23 Bardelli A, Parsons DW, Sillima N et al, 2003, Mutational analysis of the tyrosine kinome in colorectal cancers. Science 300:949–57 Tokumo M, Toyooka S, Kiura K et al, 2005, The relationship between epidermal growth factor receptor mutations and clinicopathologic features in non-small cell lung cancers. Clin Cancer Res 11:1167–1173 Kim KS, Jeong JY, Kim YC et al, 2005, Predictors of the response to gefitinib in refractory non-small cell lung cancer. Clin Cancer Res 11:2244–2251 Jackman DM, Yeap BY, Sequist LV et al, 2006, Exon 19 deletion mutations of epidermal growth factor receptor are associated with prolonged survival in non-small cell lung cancer patients treated with gefitinib or erlobinib. Clin Cancer Res 12:3908–3914 Pan ZK, Zhang L, Zhang X et al, 2005, Epidermal growth factor receptor mutation in Chinese patients with non-small cell lung cancer. Chinese Journal of Cancer 24:919–923 Zhou CC, Zhao YM, Tang L et al, 2005, Epidermal growth factor receptor mutations in Chinese patients with non-small cell lung cancer. Tumor 25:458–461 Jiang B, Zhu GS, Liu F et al, 2005, Mutational analysis of EGFR in Chinese patients with NSCLC. Academic Journal of Shanghai Second Medical University 25:1148–1150 Yoshida Y, Shibata T, Kokubu A et al, 2005, Mutations of the epidermal growth factor receptor gene in atypical adenomatous hyperplasia and bronchioloalveolar carcinoma of the lung. Lung Cancer 50:1–8 Marchetti A, Martella C, Felicioni L et al, 2005, EGFR mutations in non-small cell lung cancer: analysis of a large series of cases and development of a rapid and sensitive method for diagnositic screening with potential implications on pharmacologic treatment. J Clin Oncol 23:857–865 Miller VA, Kris MG, Shah N et al, 2004, Bronchoioloalveolar pathologic subtype and smoking history predict sensitivity to gefitinib in advanced non-small cell lung cancer. J Clin Oncol 22:1103–1109 Haneda H, Sasaki H, Shimizu S et al, 2006, Epidermal growth factor receptor gene mutation defines distinct subsets among small adenocarcinomas of the lung. Lung Cancer 52:47–52 Scagliotti GV, Fossati R, Torri Vet al, 2003, Randomized study of adjuvant chemotherapy for completely resected stage I, II or IIIA non-small-cell lung cancer. J Natl Cancer Inst 95:1453–1461 George S, Schell MJ, Detterbeck FC et al, 1998, Adjuvant chemotherapy for resected non-small cell carcinoma of the lung: why we still don't know. Oncologist 3:35–44 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 373 EP 379 DI 10.1007/s12253-008-9113-1 PG 7 ER PT J AU Menegazzo, M Bagatella, P Marson, P Donadel, C De Silvestro, G Corsini, A AF Menegazzo, Marinella Bagatella, Paola Marson, Piero Donadel, Carla De Silvestro, Giustina Corsini, Augusto TI Reduced Mobilisation of Hematopoietic Stem Cells After Hepatic Resection for Malignant Liver Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hematopoietic stem cell; Liver resection; Cancer; CD34+ mobilisation ID Hematopoietic stem cell; Liver resection; Cancer; CD34+ mobilisation AB Recent studies have demonstrated that hematopoietic stem cells (HSCs) can mobilize following liver resection, thus contributing to the repair of hepatic damage. Aim of this study has been to determine whether the nature of the hepatic lesion (benign vs. malignant disease) can give rise to a different degree of mobilisation of HSCs. Two groups of patients were selected: the first included seven patients undergoing hepatic resection (five major and two minor) for a benign liver disease (focal nodular hyperplasia, hemangioma cavernosa, angioma, biliary adenofibroma) and the second included seven patients undergoing hepatic resection (five major and two minor) for a malignant (either primary or secondary) liver disease. White blood cell count and CD34+ (percentage and total number) at time T0 (basal value before surgery) and at time T1 (value on the sixth–eighth day after surgery) have been evaluated by standard methods. In the group undergoing hepatic resection for a benign liver disease, a significant increase of CD34+ cells, both in percentage (0.082±0.043 vs. 0.048±0,026, p=0.041) and in absolute number (8.14±5.95 vs. 3.26±2.63, p=0.018) have been documented, as opposed to the group of patients affected with a malignant liver disease, where no significant variation has been observed (CD34+ %: 0.044±0.033 vs. 0.041±0.031, p: n.s.; CD34+ total number: 3.52±2.56 vs. 2.27±2.01, p=n.s.) These results show a different bone marrow response to the surgical liver resection depending on the nature of the lesion, thus emphasizing a reduced mobilisation of HSCs in the malignant diseases. Since it has been documented that the type of the hepatic lesion can induce a different regenerative response, it has to be explained how the neoplastic lesions can negatively influence the mobilization of HSCs. It can be hypothesized that a variety of humoral factors, including stromal cellderived factor, matrix metalloproteinases, hepatocyte growth factor and interleukin-8 can influence the process of mobilization of HSCs after liver resection surgery. These substances are also involved in the mechanisms of development and metastasising of many tumours. It is probably in this context that a reason may be found for the different mobilisation of hematopoietic stem cells, depending on the nature of the hepatic lesion treated, that was encountered in this study. C1 [Menegazzo, Marinella] Azienda Ospedale Universita, Division of Surgery, 35128 Padova, Italy. [Bagatella, Paola] Azienda Ospedale Universita di Padova, Blood Transfusion Service, via Giustiniani 2Padova, Italy. [Marson, Piero] Azienda Ospedale Universita di Padova, Blood Transfusion Service, via Giustiniani 2Padova, Italy. [Donadel, Carla] Azienda Ospedale Universita di Padova, Blood Transfusion Service, via Giustiniani 2Padova, Italy. [De Silvestro, Giustina] Azienda Ospedale Universita di Padova, Blood Transfusion Service, via Giustiniani 2Padova, Italy. [Corsini, Augusto] Azienda Ospedale Universita, Division of Surgery, 35128 Padova, Italy. RP Marson, P (reprint author), Azienda Ospedale Universita di Padova, Blood Transfusion Service, Padova, Italy. EM piero.marson@sanita.padova.it CR Holtzer H, 1978, Cell lineages, stem cells and the “quantal” cell cycle. In: Lord BI, Potten CS, Cole RJ, eds, Stem cells and tissue homeostasis. Cambridge University Press, Cambridge, pp 1–28 Potten CS, Loeffler M, 1990, Stem cells: attributes, cycles, spiral pittfalls and uncertainties. Lesson for and from the crypt. Development 110:1101–1120 Petersen BE, Bowen WC, Patrene KD et al, 1999, Bone marrow as a potential source of hepatic oval cells. Science 284:1168–1170 Ferrari G, Cusella De Angelis G et al, 1998, Muscle regeneration by bone marrow-derived myogenic progenitors. Science 279: 1528–1530 Li Y, Chen J, Wang L, Lu M et al, 2001, Treatment of stroke in rat with intracarotid administration of marrow stromal cells. Neurology 56:1666–1672 Jackson KA, Majka SM, Wang H et al, 2001, Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells. J Clin Invest 107:1395–1402 Bjornson CR, Rietze RL, Reynolds BA et al, 1999, Turning brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo. Science 283:534–537 Brazelton TR, Rossi FM, Keshet GI et al, 2000, From marrow to brain: expression of neuronal phenotypes in adult mice. Science 290:1775–1779 Krause DS, Theise ND, Collector MI et al, 2001, Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell 105:369–377 Anderson DJ, Gage FH, Weissman IL, 2001, Can stem cell cross lineage boundaries?. Nat Med 7:393–395 Sell S, 2001, Heterogeneity and plasticity of hepatocyte lineage cells. Hepatology 33:738–50. Vessey CJ, de la Hall P, 2001, Hepatic stem cells: a review. Pathology 33:130–141 Grompe M, 2003, The role of bone marrow stem cells in liver regeneration. Semin Liver Dis 23:363–371 Grompe M, 2004, The importance of knowing your identity: sources of confusion in stem cell biology. Hepatology 39:35–37 Dalakas E, Newsome PN, Harrison DJ et al, 2005, Hematopoietic stem cell trafficking in liver injury. FASEB J 19:1225–1231 De Silvestro G, Vicarioto M, Donadel C et al, 2004, Mobilization of peripheral blood hematopoietic stem cells following liver resection surgery. Hepatogastroenterol 51:805–810 Globerson A, 1999, Hematopoietic stem cells and aging. Exp Geront 34:137–146 Krieg A, Schulte am Esch J, Schmelzle M et al, 2006, Stem cell factor levels do increase in patients subsequent to hepatectomy with the extent of parenchimal loss. Transplant Proc 38: 3556–3558 Kollet O, Shivtiel S, Chen YQ et al, 2003, HGF, SDF-1 and MMP-9 are involved in stress-induced human CD34+ stem cell recruitment to the liver. J Clin Invest 112:160–169 Ceradini DJ, Kulkarni AR, Callaghan MJ et al, 2004, Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 10:858–864 Visse R, Nagase H, 2003, Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structures, function and biochemistry. Circ Res 92:827–839 Heissing B, Hattori K, Dias S et al, 2002, Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell 109:625–637 Watanabe Y, Haruyama T, Akaike T, 2003, Liver-derived matrix metalloproteinase-9, gelatinase B, recruits progenitor cells frombone marrow into the blood circulation. Biol Pharm Bull 26:564–568. Haruyama T, Ajioka I, Akaike T et al, 2000, Regulation and significance of hepatocyte-derived matrix metalloproteinases in liver remodeling. Biochem Biophys Res Commun 272:681–686. Fibbe WE, Pruijt JF, Velders GA et al, 1999, Biology of IL-8- induced stem cell mobilization. Ann NY.Acad Sci 872:71–882. Csete M, 2005, Oxygen in the cultivation of stem cells. Ann NY Acad Sci 1049:1–8 Mori T, Doi R, Koizumi M et al, 2004, CXCR4 antagonist inhibits stromal cell-derived factor 1-induced migration and invasion of human pancreatic cancer. Mol Cancer Ther 3:29–37 Zeelenberg IS, Ruuls-Van Stalle L, Roos E, 2003, The chemokine receptor CXCR4 is required for outgrowth of colon carcinoma micrometastases. Cancer Res 63:3833–389 Fong S, Itahana Y, Sumida T et al, 2003, Id-1 as a molecular target in therapy for breast cancer cell invasion and metastases. Proc Natl Acad Sci U S A 100:13543–13548 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 381 EP 385 DI 10.1007/s12253-008-9091-3 PG 5 ER PT J AU Ueki, T Ohashi, K Jinta, M Okuyama, Y Hiruma, K Akiyama, H Sakamaki, H AF Ueki, Toshimitsu Ohashi, Kazuteru Jinta, Minako Okuyama, Yoshiki Hiruma, Kiyoshi Akiyama, Hideki Sakamaki, Hisashi TI Delayed Hematological Recovery Following Autologous Transplantation Utilizing Peripheral Blood Stem Cells Harvested After Treatment with Arsenic Trioxide SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Arsenic trioxide; Acute promyelocytic leukemia; Delayed hematological recovery; Autologous PBSCT ID Arsenic trioxide; Acute promyelocytic leukemia; Delayed hematological recovery; Autologous PBSCT AB We describe herein two cases of delayed hematological recovery (DHR) following autologous peripheral blood stem cell transplantation (auto-PBSCT) using cells harvested during second molecular remission after treatment with arsenic trioxide (As2O3). Current therapeutic strategies with As2O3 plus auto-PBSCT might be hampered by potential mechanisms of DHR. Our observations highlight the need for study of the real effects of As2O3 on the kinetics of normal hematopoietic engraftment following auto-PBSCT. C1 [Ueki, Toshimitsu] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Ohashi, Kazuteru] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Jinta, Minako] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Okuyama, Yoshiki] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Diseases Center, Transfusion Medicine and Cell Therapy DivisionTokyo, Japan. [Hiruma, Kiyoshi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Diseases Center, Transfusion Medicine and Cell Therapy DivisionTokyo, Japan. [Akiyama, Hideki] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Sakamaki, Hisashi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. RP Ohashi, K (reprint author), Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 113-8677 Tokyo, Japan. EM k.ohashi@cick.jp CR Soignet SL, Maslak P, Wang Z-G, et al, 1998, Complete remission after treatment of acute promyelocytic leukemia with arsenic trioxide. N Engl J Med 339:1341–1348 Niu C, Yan H, Sun HP, et al, 1999, Studies on treatment of acute promyelocytic leukemia with arsenic trioxide: remission induction, follow-up, and molecular monitoring in 11 newly diagnosed and 47 relapsed acute promyelocytic leukemia patients. Blood 94:3315–3324 Douer D, Hu W, Giralt S, et al, 2003, Arsenic trioxide, trisenox, therapy for acute promyelocytic leukemia in the setting of hematopoietic stem cell transplantation. Oncologist 8:132–140 de Botton S, Fawaz A, Chevret S, et al, 2005, Autologous and allogeneic stem-cell transplantation as salvage treatment of acute promyelocytic leukemia initially treated with all-trans-retinoic acid: a retrospective analysis of the European Acute Promyelocytic Leukemia Group. J Clin Oncol 23:120–126 Ohno R, Asou N, Japan Adult Leukemia Study Group, 2004, The recent JALSG study for newly diagnosed patients with acute promyelocytic leukemia, APL). Ann Hematol 83 Suppl 1:77–78 Mehta J, Powles R, Singhal S, et al, 1996, Melphalan-total body irradiation and autologous bone marrow transplantation for adult acute leukemia beyond first remission. Bone Marrow Transplant 18:119–123 Mikoshiba M, Ohashi K, Takei N, et al, 2002, Successful unrelated bone marrow transplantation after arsenic trioxide treatment in a patient with relapsed acute promyelocytic leukemia. Int J Hematol 75:104–106 Beguin Y, Baudoux E, Sautois B, et al, 1998, Hematopoietic recovery in cancer patients after transplantation of autologous peripheral blood CD34+ cells or unmanipulated peripheral blood stem and progenitor cells. Transfusion 38:199–208 Lanza F, Campioni D, Moretti S, et al, 2001, CD34(+, cell subsets and long-term culture colony-forming cells evaluated on both autologous and normal bone marrow stroma predict longterm hematopoietic engraftment in patients undergoing autologous peripheral blood stem cell transplantation. Exp Hematol 29:1484– 1493 Douer D, Tallman MS, 2005, Arsenic trioxide: new clinical experience with an old medication in hematologic malignancies. J Clin Oncol 23:2396–2410 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 387 EP 390 DI 10.1007/s12253-008-9049-5 PG 4 ER PT J AU Kovacs, TG Barany, O Schlick, B Csoka, M Gado, J Ponyi, A Muller, J Nemeth, J Hauser, P Erdelyi, JD AF Kovacs, T Gabor Barany, Olga Schlick, Barbara Csoka, Monika Gado, Judit Ponyi, Andrea Muller, Judit Nemeth, Julia Hauser, Peter Erdelyi, J Daniel TI Late Immune Recovery in Children Treated for Malignant Diseases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Humoral immunity; Cellular immunity; Late effects; Malignant diseases; Children ID Humoral immunity; Cellular immunity; Late effects; Malignant diseases; Children AB In this study we analyzed the recovery of the immune system in children after completion of the therapy. We analysed 88 children (51 boys, 37 girls, mean age at diagnosis: 7.8 years) receiving chemotherapy for malignant diseases (43 acute lymphoblastic leukemia, 15 lymphoma, 20 bone tumor, ten other solid tumors). Serum immunoglobulin levels (Ig), natural killer activity (NK), antibodydependent cellular cytotoxicity (ADCC) and T and B cell proliferation were determined 1 year after cessation of therapy. The mean levels of Ig were in the normal range at a mean of 13 months after chemotherapy (IgG: 11.2±3.3, IgA: 1.6±0.9, IgM: 1.0±0.5 g/l), however in the leukemic patients serum IgG was below the lower limit of the normal range in 3/43 (7.0%) cases, serum IgA was low in 5/43 (11.6%) and serum IgM was decreased in 4/43 (9.3%) cases. In the solid tumor patients IgG values were within the normal range and only 2–2/45 children had lower values for IgA and IgM (4.4%). NK activity decreased in 7/43 (16.3%) leukemic patients, and in 3/45 (6.7%) solid tumor patients, ADCC decreased in 8/43 (18.6%) and 3/45 (6.7%), respectively (p<0.001). B-cell blastic transformation was decreased in 3/43 (7%) leukemic patients and in 4/45 (8.9%) solid tumor patients. At the same time T-cell blastic transformation was altered in 5/43 (11.6%) and in 4/45 (8.9%) cases, respectively. Leukemic patients had significantly more infections during the first year after chemotherapy than solid tumor patients (1.60±1.18 vs 0.96±1.14; p=0.011). No significant correlations could be found between the investigated immune parameters and the number and severity of infections. It is concluded, that cytotoxic therapy can lead to long-term depression of the immune system, first of all in leukemic patients. C1 [Kovacs, T Gabor] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Barany, Olga] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Schlick, Barbara] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Csoka, Monika] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Gado, Judit] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Ponyi, Andrea] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Muller, Judit] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Nemeth, Julia] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Hauser, Peter] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Erdelyi, J Daniel] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Kovacs, TG (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM kovi@gyer2.sote.hu CR Smith MA, Ries LAG, 2002, Childhood cancer: Incidence, survival and mortality. In: Pizzo PA, Poplack DG, eds, Principles and practice of pediatric oncology. Lippincott Williams and Wilkins, Philadelphia, pp 1–12 Schrappe M, 2004, Evolution of BFM trials for childhood ALL. Ann Hematol 83(suppl 1):S121–S123 Pinkerton R, 2005, Continuing challenges in childhood non- Hodgkin lymphoma. Br J Haematol 130:480–488 Smith M, Hare ML, 2004, An overview of progress in childhood cancer survival. J Pediatr Oncol Nursing 21:160–164 Ajiki W, Tsukuma H, Oshima A, 2004, Survival rates of childhood cancer patients in Osaka, Japan. Jap J Clin Oncol 34:50–54 Lando A, Holm K, Nysom K et al, 2001, Thyroid function in survivors of childhood acute lymphoblastic leukaemia: the significance of prophylactic cranial irradiation. Clin Endocrinol 55:21–25 Schwartz CL, 1999, Long-term survivors of childhood cancer: the late effects of therapy. Oncologist 4:45–54 Arguelles B, Barrios V, Pozo J et al, 2000, Modifications of growth velocity and the insulin-like growth factor system in children with acute lymphoblastic leukemia: a longitudinal study. J Clin End Met 85:4087–4092 Larson DL, Kroll S, Jaffe N et al, 1990, Long-term effects of radiotherapy in childhood and adolescence. Am J Surg 160:348– 351 Cicognani A, Pasini A, Pession A et al, 2003, Gonadal function and pubertal development after treatment of a childhood malignancy. J Ped End 16(Suppl 2):321–326 Harris J, Sengar D, Stewart T et al, 1976, The effect of immunosuppressive chemotherapy on immune function in patients with malignant disease. Cancer 37:1058–1069 Hektoen L, Corper HJ, 1921, The effect of mustard gas, dichclorethylsulphid, on antibody formation. J Inf Dis 28:279– 285 Calabresi P, Chabner BA, 1991, Antineoplastic agents. In: Brunton LL, Lazo JS, Parker KL, eds, Goodman and Gilman’s: the pharmacological basis of therapeutics. McGraw-Hill, Singapore, pp 1209–1263 Alanko S, Pelliniemi T, Salmi T, 1994, Recovery of blood lymphocytes and serum immunoglobulins after treatment of solid tumors in children. Ped Hematol Oncol 11:33–45 Mustafa M, Buchanan G, Winick N et al, 1998, Immune recovery in children with malignancy after cessation of chemotherapy. J Ped Hematol Oncol 20:451–457 Komada Y, Zhang SL, Zhou YW et al, 1992, Cellular immunosuppression in children with acute lymphoblastic leukemia: effect of consolidation chemotherapy. Cancer Immunol Immunther 35:271–276 Craenendonk E, van Gennip AH, Abeling NGM et al, 1984, Numerical changes in the various peripheral white blood cells in children as a result of antineoplastic therapy. Acta Haematol 72:315–325 Mackall C, Fleisher T, Brown M et al, 1995, Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy. N Engl J Med 332:143–149 Moritz B, Eder J, Meister B et al, 2001, Intact T-cell regenerative capacity in childhood acute lymphoblastic leukemia after remission induction therapy. Med Ped Oncol 36:283–289 Smith S, Schiffman G, Karayalcin G et al, 1995, Immunodeficiency in long-term surviors of acute lymphoblastic leukemia treated with Berlin-Frankfurt-Munster therapy. J Ped 127:68–75 Reinhardt D, Houliara K, Pekrun A et al, 2003, Impact of conventional chemotherapy on levels of antibodies against vaccine-preventable diseases in children treated for cancer. Scand J Inf Dis 35:851–857 Eckschlager T, Jira M, Strejcek J, 1991, Cell-mediated immunity in children with Hodgkin s and non-Hodgkin s lymphomas. Ped Hematol Oncol 8:251–256 Gale RP, Butturini A, 1991, Maintenance chemotherapy and cure of childhood acute lymphoblastic leukemia. Lancet 338:1315– 1318 Alanko S, Salmi TT, Pelliniemi TT, 1995, Recovery of natural killer cells after chemotherapy for childhood acute lymphoblastic leukemia and solid tumors. Med Ped Oncol 24:373–378 Alanko S, Pelliniemi T, Salmi TT, 1992, Recovery of blood Blymphocytes and serum immunoglobulins after treatment for childhood acute lymphoblastic leukemia. Cancer 69:1481–1486 Yamada S, Komiyama A, 1991, Decrease in number of CD16, Leu 11)+CD45RA, 2H4)+ cells and defective production of natural killer cytotoxic factor in childhood acute lymphoblastic leukemia. Leukemia Res 15:785–790 Jackson SK, Parton J, Shortland G et al, 1990, Serum immunoglobulins to endotoxin core glycolipid: acute leukaemia and other cancers. Arch Dis Child 7:771–773 Yabuhara A, Kawai H, 1990, A recycling defect as a characteristic of natural killer cells in childhood acute lymphoblastic leukemia. Ped Res 28:572–578 Bahceci E, Epperson D, Douek DC et al, 2003, Early reconstitution of the T-cell repertoire after non-myeloablative peripheral blood stem cell transplantation is from post-thymic T-cell expansion and is unaffected by graft-versus-host disease or mixed chimaerism. Br J Haematol 122:934–943 Lynch BA, Vasef MA, Comito M et al, 2003, Effect of in vivo lymphocyte-depleting strategies on development of lymphoproliferative disorders in children post allogeneic bone marrow transplantation. Bone Marrow Transplant 32:527–533 Kalwak K, Moson I, Cwian J et al, 2003, A prospective analysis of immune recovery in children following allogeneic transplantation of T-cell-depleted or non-T-cell-depleted hematopoietic cells from HLA-disparate family donors. Transplant Proc 35:1551– 1555 Kalwak K, Gorczynska E, Toporski J et al, 2002, Immune reconstitution after haematopoietic cell transplantation in children: immunophenotype analysis with regard to factors affecting the speed of recovery. Br J Haematol 118:74–89 Auletta JJ, Fisher VL, 2001, Immune reconstitution in pediatric stem-cell transplantation. Front Bioscience 6:G23–G32 Locatelli F, Maccario R, Comoli P et al, 1996, Hematopoietic and immune recovery after transplantation of cord blood progenitor cells in children. Bone Marrow Transplant 18:1095–1101 Oppemheim JJ, 1975, Use of lymphocyte transformation to assess clinical disorders. In: Vyas GN, Stites DP, Brecher G, eds, Laboratory diagnosis of immunologic disorders. Grune and Stratton, New York, p 87 Benczur M, 1981, In vitro assay for detection of natural killer, NK, cells. In: Erdei A, Fesus L, Rajnavolgyi E, eds, Immunological methods. Springer, Budapest, p 122 Coligan JE, 1991, Current protocols in immunology. NIH, John Wiley Interscience, Boston, pp 181–183 Kristinsson VH, Kristinsson JR, Jonmundsson GK et al, 2001, Immunoglobulin class and subclass concentrations after treatment of childhood leukemia. Ped Hematol Oncol 18:167–172 2001 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 391 EP 397 DI 10.1007/s12253-008-9073-5 PG 7 ER PT J AU Ghaffari, HS Obeidi, N Dehghan, M Alimoghaddam, K Gharehbaghian, A Ghavamzadeh, A AF Ghaffari, H Seyed Obeidi, Narghes Dehghan, Mehdi Alimoghaddam, Kamran Gharehbaghian, Ahmad Ghavamzadeh, Ardashir TI Monitoring of Cytomegalovirus Reactivation in Bone Marrow Transplant Recipients by Real-time PCR SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cytomegalovirus; Cyclosporine; pp65 Antigenemia; Real-time PCR; TaqMan ID Cytomegalovirus; Cyclosporine; pp65 Antigenemia; Real-time PCR; TaqMan AB Cytomegalovirus (CMV) has been recognized as the most important viral pathogen in persons undergoing bone marrow transplantation (BMT). The aim was to develop a quantitative PCR assay to quantify CMV DNA in peripheral blood leukocytes (PBLs) of bone marrow transplantation (BMT) patients. An in-house real-time PCR assay based on TaqMan technology was developed to monitor the quantity of CMV DNA in PBLs of the BMT recipients. Sequential blood samples (415 specimens) were collected from 43 patients as weekly intervals until day 100 after transplantation. The CMV DNA was quantified in parallel with the pp65 antigenemia assay in PBL samples. Viral reactivation occurred in 51% and 41.8% of the recipients as detected by RQ-PCR and antigenemia assays respectively. There was a significant correlation between both assays (P<0.0001); however, the RQ-PCR was more sensitive than the antigenemia. CMV DNA was detected by the RQ-PCR by a median of 14 days earlier than the antigenemia. Preemptive therapy was implemented in the antigenemia positive cases. The administration of ganciclovir led to a rapid decrease in the viral load. After preemptive therapy, the antigenemia achieved a negative result earlier than the RQ-PCR assay (a median of 17.5 days). An increase of viral load in both quantitative assays and of cyclosporine serum level were identified as the most significant risk factors for CMV reactivation. The quantitative CMV PCR might be a useful tool for monitoring the CMV reactivation and guiding the efficacy of the CMV preemptive therapy in BMT recipients. C1 [Ghaffari, H Seyed] Tehran University of Medical Sciences, Research Center, Shariati Hospital, Hematology, Oncology and Bone Marrow Transplantation, 11411 Tehran, Iran. [Obeidi, Narghes] Iranian Blood Transfusion Organization Research CenterTehran, Iran. [Dehghan, Mehdi] Tehran University of Medical Sciences, Research Center, Shariati Hospital, Hematology, Oncology and Bone Marrow Transplantation, 11411 Tehran, Iran. [Alimoghaddam, Kamran] Tehran University of Medical Sciences, Research Center, Shariati Hospital, Hematology, Oncology and Bone Marrow Transplantation, 11411 Tehran, Iran. [Gharehbaghian, Ahmad] Iranian Blood Transfusion Organization Research CenterTehran, Iran. [Ghavamzadeh, Ardashir] Tehran University of Medical Sciences, Research Center, Shariati Hospital, Hematology, Oncology and Bone Marrow Transplantation, 11411 Tehran, Iran. RP Ghaffari, HS (reprint author), Tehran University of Medical Sciences, Research Center, Shariati Hospital, Hematology, Oncology and Bone Marrow Transplantation, 11411 Tehran, Iran. EM shghaffari2000@yahoo.com CR Boeckh M, Boivin G, 1998, Quantitation of cytomegalovirus: methodologic aspects and clinical applications. Clin Microbiol Rev 11:533–554 Salzberger B, Bowden RA, Hackman R et al, 1997, Neutropenia in allogeneic marrow transplant recipients receiving ganciclovirfor prevention of CMV disease: risk factors and outcome. Blood 90: 2502–2508 Li CR, Greenberg PD, Gilbert MJ et al, 1994, Recovery of HLArestricted cytomegalovirus, CMV)-specific T-cell responses after allogeneic bone marrow transplant: correlation with CMV disease and effect of ganciclovir prophylaxis. Blood 83:1971–1979 Goodrich JM, Bowden RA, Fisher L et al, 1993, Ganciclovir prophylaxis to prevent cytomegalovirus disease after allogeneic marrow transplant. Ann Intern Med 118:173–178 Winston DJ, Ho WG, Bartoni K et al, 1999, Ganciclovir prophylaxis of cytomegalovirus infection and disease in allogeneic bone marrow transplant recipients. Results of a placebocontrolled, double-blind trial. Ann Intern Med 118:179–184 Goodrich JM, Mori M, Gleaves CA et al, 1991, Early treatment with ganciclovir to prevent cytomegalovirus disease after allogeneic bone marrow transplantation. N Engl J Med 325:1601–1607 Schmidt GM, Horak DA, Niland JC et al, 1991, A randomized controlled trial of prophylactic ganciclovir for cytomegalovirus pulmonary infection in recipients of allogeneic bone marrow transplants. N Engl J Med 324:1005–1011 Aitken CW, Barrett-Muir C, Millar K et al, 1999, Use of molecular assays in diagnosis and monitoring of cytomegalovirus disease following renal transplantation. J Clin Microbiol 37:2804– 2807 Griscelli F, Barrois M, Chauvin S et al, 2001, Quantification of human cytomegalovirus DNA in bone marrow transplant recipients by real-time PCR. J Clin Microbiol 39:4362–4369 Gleaves CA, Smith TH, Shuster EA et al, 1984, Rapid detection of cytomegalovirus in MRC-5 cells inoculated with urine specimens by using low-speed centrifugation and monoclonal antibody to an early antigen. J Clin Microbiol 19:917–919 Boeckh M, Gooley TA, Myerson D et al, 1996, Cytomegalovirus pp65 antigenemia-guided early treatment with ganciclovir at engraftment after allogeneic marrow transplantation: a randomized double-blind study. Blood 10:4063–4071 Hebart H, Gamer D, Loeffer J et al, 1998, Evaluation of Murex CMV DNA hybrid capture assay for the detection and quantification of cytomegalovirus infection in patients following allogeneic stem cell transplantation. J Clin Microbiol 36:1333–1337 Einsele H, Steidle M, Vallbracht A et al, 1991, Early occurrence of human cytomegalovirus infection after bone marrow transplantation as demonstrated by the polymerase chain reaction technique. Blood 77:1104–1110 Gault E, Miche Y, Dehe’e A et al, 2001, Quantification of human cytomegalovirus DNA by real-time PCR. J Clin Microbiol 39: 772–775 Machida U, Kami M, Fukui T et al, 2000, Real-time automated PCR for early diagnosis and monitoring of cytomegalovirus infection after bone marrow transplantation. J Clin Microbiol 38: 2536–2542 Mori T, Okamoto S, Matsuoka S et al, 2000, Risk-adapted preemptive therapy for cytomegalovirus disease in patients undergoing allogeneic bone marrow transplantation. Bone Marrow Transplant 25:765–769 Nitsche A, Steuer N, Schmidt CA et al, 2000, Detection of human cytomegalovirus DNA by real-time quantitative PCR. J Clin Microbiol 38:2734–2737 Razonable RR, Brown RA, Espy MJ et al, 2001, Comparative quantification of cytomegalovirus, CMV, DNA in solid-organ transplant recipients with CMV infection by using two highthroughput automated systems. J Clin Microbiol 39:4472–4476 Schaade L, Kockelkorn P, Ritter K et al, 2000, Detection of cytomegalovirus DNA in human specimens by lightCycler PCR. J Clin Microbiol 38:4006–4009 Tanaka N, Kimura H, Iida K et al, 2000, Quantitative analysis of cytomegalovirus load using a real-time PCR assay. J Med Virol 60:455–462 Yun Z, Lewensohn-Fuchs I, Ljungman P et al, 2000, Real-time monitoring of cytomegalovirus infections after stem cell transplantation using the TaqMan polymerase chain reaction assays. Transplantation 69:1733–1736 Yakushiji K, Gondo H, Kamezaki K et al, 2002, Monitoring of cytomegalovirus reactivation after allogeneic stem cell transplantation: comparison of an antigenemia assay and quantitative real-time polymerase chain reaction. Bone Marrow Transplant 29:599–606 Leruez-Ville M, Ouachee M, Delarue R et al, 2003, Monitoring cytomegalovirus infection in adult and pediatric bone marrow transplant recipients by a real-time PCR assay performed with blood plasma. J Clin Microbiol 41:2040–2046 Tanabe K, Tokumoto T, Ishikawa N et al, 1997, Comparative study of cytomegalovirus, CMV, antigenemia assay, polymerase chain reaction, serology, and shell vial assay in the early diagnosis and monitoring of CMV infection after renal transplantation. Transplantation 64:1721–1725 Seropian S, Ferguson D, Salloum E et al, 1998, Lack of reactivity to CMV pp65 antigenemia testing in a patient with CMV disease following allogeneic bone marrow transplant. Bone Marrow Transplant 22:507–509 Solano C, Munoz I, Gutierrez A et al, 2001, Qualitative plasma PCR Assay, AMPLICOR CMV Test, versus pp65 antigenemia assay for monitoring cytomegalovirus viremia and guiding preemptive ganciclovir therapy in allogeneic stem cell transplantation. J Clin Microbiol 39:3938–3941 Ljungman P, Plotkin S, 1995, Workshop on CMV disease— definitions, clinical severity scores and new syndromes. Scand J Infect Dis Suppl 99:87–89 Przepiorka D, Weisdorf D, Martin P et al, 1994, Consensus conference on Acute GVHD grading. Bone Marrow Transplant 15:825–828 Jabs DA, Forman M, Enger C et al, 1999, Comparison of cytomegalovirus loads in plasma and leukocytes of patients with cytomegalovirus retinitis. J Clin Microbiol 37:1431–1435 Pellegrin I, Garrigue I, Ekouevi D et al, 2000, New molecular assays to predict occurrence of cytomegalovirus disease in renal transplant recipients. J Infect Dis 182:36–42 Schafer P, Tenschert W, Cremaschi L, et al, 2001, Area under the viraemia curve versus absolute viral load: utility for predicting symptomatic cytomegalovirus infections in kidney transplant patients. J Med Virol 65:85–89 Boeckh M, Hawkins G, Myerson D et al, 1997, Plasma PCR for cytomegalovirus DNA after allogeneic marrow transplantation: comparison with PCR using peripheral blood leukocytes, pp65 antigenemia and viral culture. Transplantation 64:108–113 Ksouri H, Eljed H, Greco A et al, 2007, Analysis of cytomegalovirus, CMV, viremia using the pp65 antigenemia assay, the amplicor CMV test, and a semi- quantitative polymerase chain reaction test after allogenic marrow transplantation. Transpl Infect Dis 9:16–21 Garrigue I, Boucher S, Couzi L et al, 2006, Whole blood realtime quantitative PCR for cytomegalovirus infection follow-up in transplant recipients. J Clin Virol 36:72–75 Mhiri L, Kaabi B, Houimel M, Arrouji Z, Slim A, 2007, Comparison of pp65 antigenemia, quantitative PCR and DNA hybrid capture for detection of cytomegalovirus in transplant recipients and AIDS patients. J Virol Methods 143:23–28 Schvoerer E, Henriot S, Zachary P et al, 2005, Monitoring low cytomegalovirus viremia in transplanted patients by a real-time PCR on plasma. J Med Virol 76:76–81 Martin-Davila P, Fortun J, Gutierrez C et al, 2005, Analysis of a quantitative PCR assay for CMV infection in liver transplant recipients: an intent to find the optimal cut-off value. J Clin Virol 33: 138–144 Gouarin S, Vabret A, Gault E et al, 2004, Quantitative analysis of HCMV DNA load in whole blood of renal transplant patients using real-time PCR assay. J Clin Virol 29:194–201 Li H, Dummer S, Estes WR, et al, 2003, Measurement of human cytomegalovirus loads by quantitative real-time PCR for monitoring clinical intervention in transplant recipients. J Clin Microbiol 41:187–191 Mengelle C, Sandres-Saun’e K, Pasquier C et al, 2003, Automated extraction and quantification of human cytomegalovirus DNA in whole blood by real-time PCR assay. J Clin Microbiol 41:3840–3845 Guiver M, Fox AJ, Mutton K, Mogulkoc N, Egan J, 2001, Evaluation of CMV viral load using TaqMan CMV quantitative PCR and comparison with CMV antigenemia in heart and lung transplant recipients. Transplant 71:1609–1615 Drew WL, 2007, Laboratory diagnosis of cytomegalovirus infection and disease in immunocompromised patients. Curr Opin Infect Dis 20:408–411 Lengerke C, Ljubicic T, Meisner C et al, 2006, Evaluation of the COBAS Amplicor HCMV Monitor for early detection and monitoring of human cytomegalovirus infection after allogenic stem cell transplantation. Bone Marrow Transplant 38:53–60 Allice T, Enrietto M, Pittaluga F et al, 2006, Quantitation of cytomegalovirus DNA by real-time polymerase chain reaction in peripheral blood specimens of patients with solid organ transplants: comparison with end-point PCR and pp65 antigen test. J Med Virol 78:915–922 Pumannova M, Roubalova K, Vitek A, Sajdova J, 2006, Comparison of quantitative competitive polymerase chain reactionenzyme- linked immunosorbent assay with LightCycler-based polymerase chain reaction for measuring cytomegalovirus DNA in patients after hematopoietic stem cell transplantation. Diagn Microbiol Infect Dis 54:115–120 Mori T, Mori S, Kanda Y et al, 2004, Clinical significance of cytomegalovirus, CMV, antigenemia in the prediction and diagnosis of CMV gastrointestinal disease after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 33:431–434 Weinberg A, Hodge TN, Li S et al, 2000, Comparison of PCR, antigenemia assay, and rapid blood culture for detection and prevention of cytomegalovirus disease after lung transplantation. J Clin Microbiol 38(2):768–772 Ikewaki J, Ohtsuka E, Kawano R et al, 2003, Real-time PCR assay compared to nested PCR and antigenemia assays for detecting cytomegalovirus reactivation in adult T-cell leukemialymphoma patients. J Clin Microbiol 41:4382–4387 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 399 EP 409 DI 10.1007/s12253-008-9030-3 PG 11 ER PT J AU Illes, Simon, Zs Toth, E Rosta, A Miltenyi, Zs Molnar, Zs AF Illes, Arpad Simon, Zsofia Toth, Erika Rosta, Andras Miltenyi, Zsofia Molnar, Zsuzsa TI Nodular Lymphocyte Predominant Hodgkin Lymphoma (NLPHL)—Clinicopathological Features Based on the Data of Two Hungarian Lymphoma Centres SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Nodular lymphocyte predominant Hodgkin lymphoma; Classical Hodgkin lymphoma; Differential diagnosis; Immunohistochemistry; Treatment; Prognosis ID Nodular lymphocyte predominant Hodgkin lymphoma; Classical Hodgkin lymphoma; Differential diagnosis; Immunohistochemistry; Treatment; Prognosis AB Clinicopathological features of nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) differ from those of the classical Hodgkin lymphoma (cHL). Our aim was to examine clinical presentation, therapeutic and survival results of NLPHL patients in Hungary based on the data of two centres, and incidentally we analyzed the clinicopathological characteristics and differential diagnostic difficulties of this rare entity. We analyzed the clinical features, treatment and survival data of 536 Hodgkin lymphoma patients who had been diagnosed and primarily treated in our institutes between 1995 and 2004. Mean follow-up time was 82.7 (3–144) months of the total 536 HL patients. Sixteen (3%) of the patients were diagnosed with NLPHL, 93% of them presented with early-stage disease. None of the patients showed extranodal or splenic involvement or bulky disease. One patient received chemotherapy alone, six received only involved field radiotherapy while six underwent combined modality treatment. We applied watch and wait strategy in three cases. Overall response rate was 100% (93.75% complete). Two NLPHL cases transformed to non-Hodgkin’s lymphoma. In contrast to the classical HL, the 10-year prognosticated overall survival rate was 100 vs. 82%, the event free survival was: 75% vs. 70%. In NLPHL group there were no late or multiple relapses and none of them died. Conclusions: NLPHL is a rare disease, thus these are limited experiences with its diagnosis and treatment. Since the disease has an excellent outcomeit is very important to prefer less toxic or local therapies to reach long term survival similar to that of the normal population. C1 [Illes, Arpad] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, 4032 Debrecen, Hungary. [Simon, Zsofia] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, 4032 Debrecen, Hungary. [Toth, Erika] National Institute of OncologyBudapest, Hungary. [Rosta, Andras] National Institute of OncologyBudapest, Hungary. [Miltenyi, Zsofia] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, 4032 Debrecen, Hungary. [Molnar, Zsuzsa] National Institute of OncologyBudapest, Hungary. RP Simon, Zs (reprint author), University of Debrecen, Medical and Health Center, 3rd Department of Medicine, 4032 Debrecen, Hungary. EM zsocogo@gmail.com CR Jackson H, 1937, Classification and prognosis of Hodgkin’s disease and allied disorders. Surg Gynecol Obstet 64:465 Jackson H, Parker F, 1944, Hodgkin’s disease. I. General considerations. N Eng J Med 230:35–44 Lukes RJ, Butler JJ, 1966, The pathology and nomenclature of Hodgkin’s disease. Cancer Res 26:1063–1083 Lukes RJ, Butler JJ, Hicks EB, 1966, Natural history of Hodgkin’s disease as related to its pathologic picture. Cancer 19:317 Tsai HK, Mauch PM, 2007, Nodular lymphocyte-predominant Hodgkin lymphoma. Semin Rad Oncol 17:184–189 Harris NL, Jaffe ES, Stein H, et al, 1994, A revised European– American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood 84:1361–1362 Harris NL, Jaffe ES, Diebold J et al, 1999, World Health Organization Classification of neoplastic diseases of hematopoetic and lymphoid tissues: Report of the Clinical Advisory Commitee Meeting—Airlie House, Virginia, November 1997. J Clin Oncol 17:3835–3849 Nogova L, Rudiger T, Engert A, 2006, Biology, clinical course and management of nodular lymphocyte-predominant Hodgkin lymphoma. Hematology Am Soc Hematol Educ Program 2006:266–272 Diehl V, Sextro M, Franklin J et al, 1999, Clinical presentation, course and prognostic factors in lymphocyte-predominant Hodgkin’s disease and lymphocyte-rich classical Hodgkin’s disease: report from the European Task Force on Lymphoma Project on Lymphocyte-Predominant Hodgkin’s Disease. J Clin Oncol 17:776–783 Hicks J, Flaitz C, 2002, Progressive transformation of germinal centers: review of histopathologic and clinical features. Int J Ped Otorhinolar 65:195–202 Boudova L, Torlakovic E, Delabie J et al, 2003, Nodular lymphocyte-predominant Hodgkin lymphoma with nodules resembling T-cell/histiocyte-rich B-cell lymphoma: differential diagnosis between nodular lymphocyte-predominant Hodgkin lymphoma and T-cell/histiocyte-rich B-cell lymphoma. Blood 102(10):3753–3758 Lister TA, Crowther D, Sutcliffe SB, et al, 1989, Report of a committee convened to discuss the evolution and staging of patients with Hodgkin’s disease: Cotswolds meeting. J Clin Oncol 7:1630–1636 Tubiana M, Henry-Amar M, Carde P, et al, 1989, Toward comprehensive management tailored to prognostic factors of patients with clinical stages I and II in Hodgkin’s disease. The EORTC Lymphoma Group controlled clinical trials: 1964–1987. Blood 73(1):47–56 Hasenclever D, Diehl V, 1998, A prognostic score for advanced Hodgkin’s disease. N Eng J Med 339:1506–1514 Varga Gy et al, 1998, Hodgkin’s disease. Magy Belorv Arch 2, suppl.):80–81 Illes A, Udvardy M, Esik O, et al, 2004, Hodgkin’s disease, HL). In: Lehoczky D, ed, Hematologiai betegsegek kezelesenek modszertana. Documed, Budapest, pp 133–140 Illes A, Keresztes K, Miltenyi Zs et al, 2004, Epidemiologic and treatment data on Hodgkin’s disease in Hungary—report of the Hodgkin’s Disease Work Group. Hematol-Transzfuziol 37:153–163 Jaffe E, Harris NL, Stein H et al, 2001, Pathology and genetics of tumors of haematopoetic and lymphoid tissues. IARC, Lyon, France pp 240–243 Rahemtullah A, Reichard KK, Dorn ME et al, 2007, Doublepositive CD4+CD8+ T-cell populations in nodular lymphocyte predominant Hodgkin lymphoma: further charachterization and relationship to other entities. Haematologica Suppl 5:2 Harris NL, 1998, The many faces of Hodgkin’s disease around the world: what have we learned from its pathology? Ann Oncol 9, 5):S45–S56 Pajor L, 2002, Hodgkin lymphomas: current presentation of pathogenesis and patomorphology. Orv Hetil 143(13):651–661 Wang SA, Olson N, Zukerberg L et al, 2006, Splenic marginal zone lymphoma with micronodular T-cell rich B-cell lymphoma. Am J Surg Pathol 30:128–132 Dogan A, Burke JS, Goteri G et al, 2003, Micronodular T-cell/ histiocyte-rich large B-cell lymphoma of the spleen. Histology, immunophenotype and differential diagnosis. Am J Surg Pathol 27:903–911 Fekete S, Matolcsy A, 1998, A Hodgkin-kor es a non-Hodgkin lymphoma hatarteruletei. In: Paloczi K, Kelenyi G, eds, Non- Hodgkin lymphoma. Springer, Budapest, pp 284–289 Fan Z, Natkunam Y, Bair E et al, 2003, Characterization of variant patterns of nodular lymphocyte predominant Hodgkinlymphoma with immunohistologic and clinical correlation. Am J Surg Pathol 27(10):1346–1356 Nogova L, Reineke T, Josting A, et al, 2005, Lymphocyte predominant and classical Hodgkin’s lymphoma-comparison of outcomes. Eur J Hematol Suppl 66:106–110 Diehl V, Engert A, Re D, 2007, New Strategies for the treatment of advanced-stage Hodgkin’s lymphoma. Hematol Oncol Clin N Am 21:897–914 Cosset JM, Henry-Amar M, Meerwaldt JH, 1991, Long-term toxicity of early stages of Hodgkin’s disease therapy: the EORTC experience. EORTC Lymphoma Cooperative Group. Ann Oncol 2, Suppl.2):77–82 Hutchings M, Loft A, HansenM(2006, Different histopathological subtypes of Hodgkin lymphoma show significantly different levels of FDG uptake. Hematol Oncol 24:146–150 Wirth A, Yuen K, Barton M, 2005, Long-term outcome after radiotherapy alone for lymphocyte-predominant Hodgkin lymphoma: a retrospective multicenter study of the Australian Radiation Oncology Lymphoma Group. Cancer 104:1221–12229 Nogova L, Reineke T, Eich HT, 2005, Extended field radiotherapy, combined modality treatment or involved field radiotherapy for patients with stage IA lymphocyte-predominant Hodgkin’s lymphoma: a retrospective analysis from German Hodgkin Study Group, GHSG). Ann Oncol 16:1683–1687 Wilder RB, Schlembach PJ, Jones D, 2002, European Organization for Research and Treatment of Cancer and Groupe d’Etude des Lymphomes de l’Adulte very favorable and favorable, lymphocyte-predominant Hodgkin disease. Cancer 94:1731–1738 Savage KJ, Hoskins P, Klasa R et al, 2007, ABVD chemotherapy is essential for optimal treatment of limited stage nodular lymphocyte predominant Hodgkin lymphoma. Hematology Suppl 5:27 Muller RP, Eich HT, 2007, Dose and field size, the GSHG experience. Hematology Suppl 5:18 Ekstrand BC, Lucas JB, Horwitz SM, et al, 2003, Rituximab in lymphocyte-predominant Hodgkin disease: results of a phase 2 trial. Blood 101:4285–4289 Schulz H, Rehwald U, Morschhauser F et al, 2008, Rituximab in relapsed lymphocyte-predominant Hodgkin lymphoma: long-term results of a phase-II trial by German Hodgkin Lymphoma Study Group, GHSG). Blood 1:109–111 Younes A, Fayad LY, Goy A, et al, 2006, Results of rituximab plus ABVD in 65 newly diagnosed patients with classical Hodgkin lymphoma: improvement of event free survival, EFS, in all international prognostic score, IPS, groups [abstract]. Blood 108:2742 Younes A, Fayad LY, Goy A et al, 2007, Phase-II study of rituximab plus ABVD for the treatment of newly diagnosed patients with advanced stage classical Hodgkin lymphoma, HL). Hematology Suppl 5:33 Ambinder RF, Jones RJ, Matsui W, 2007, Hodgkin’s lymphoma, HL): evidence for a cancer stem cell and therapeutic implications. Hematology Suppl 5:12 Wickert RS, Weisenburger DD, Tierens A et al, 1995, Clonal relationship between lymphocytic predominance Hodgkin’s disease and concurrent or subsequent large-cell lymphoma of B lineage. Blood 86:2312–2320 Greiner TC, Gascoyne RD, Anderson ME et al, 1996, Nodular lymphocyte-predominant Hodgkin’s disease associated with largecell lymphoma: analysis of Ig gene rearrangements by V-J polymerase chain reaction. Blood 88:657–666 Mathas S, 2007, The pathogenesis of classical Hodgkin’s lymphoma: a model for B-cell plasticity. Hematol Oncol Clin N Am 21:787–804 Mittal BB, Nalesnik M, Composite, 1986, lymphoma, Hodgkin’s and non-Hodgkin’s, of the spleen in a previously untreated patient. Acta Hematol 76:29–32 Sundeen JT, Cossman J, Jaffe ES, 1988, Lymphocyte predominant Hodgkin’s disease nodular subtype with coexistent ‘large cell lymphoma’. Histological progression or composite malignancy? Am J Surg Pathol 12:599–606 Huang JZ, Weisenburger DD, Vose JM, et al, 2004, Diffuse large B-cell lymphoma arising in nodular lymphocyte predominant Hodgkin lymphoma: a report of 21 cases from the Nebraska Lymphoma Study Group. Leuk Lymph 45:1551–1557 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 411 EP 421 DI 10.1007/s12253-008-9043-y PG 11 ER PT J AU Nyari, AT Kajtar, P Bartyik, K Thurzo, L McNally, R Parker, L AF Nyari, Andras Tibor Kajtar, Pal Bartyik, Katalin Thurzo, Laszlo McNally, Richard Parker, Louise TI Seasonal Variation of Childhood Acute Lymphoblastic Leukaemia is Different Between Girls and Boys SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Childhood acute lymphoblastic leukaemia; Gender specific seasonal effect; Male predominance; Simple harmonic regression ID Childhood acute lymphoblastic leukaemia; Gender specific seasonal effect; Male predominance; Simple harmonic regression AB The aim of this study was to investigate seasonal trends in the incidence of acute lymphoblastic leukaemia (ALL) around the times of birth and diagnosis in children aged 0–4 years and also to examine gender specific effects. Children born in South Hungary during 1981–1997 were analysed. Registrations of first malignancies for children, diagnosed under age 5 years before the end of 2002 were obtained from the Hungarian Paediatric Oncology Group providing a representative sample of Hungarian children over a 17 year period of time. Data were available on the corresponding numbers of births for each month of the study period were obtained. Statistical analyses were performed using logistic regression with harmonic components. The study analysed 121 cases of children, aged under 5 years, who were diagnosed with ALL. We found no seasonal effect related to date of diagnosis. However, there was seasonal variability for ALL related to date of birth. Maximal rates were seen in children born in February and August in the simple harmonic regression model for all children diagnosed with ALL. Analysis by gender found evidence of seasonality related to month of birth with peaks in February and August in boys, but different seasonal effects were seen for girls (peak in November, nadir in May). Our study provides some evidence that male specific immune responses to infections around the time of birth could explain the male predominance in the incidence of ALL. C1 [Nyari, Andras Tibor] University of Szeged, Department of Medical Informatics, Koranyi fasor 9, 6720 Szeged, Hungary. [Kajtar, Pal] University of Pecs, Department of PediatricsPecs, Hungary. [Bartyik, Katalin] University of Szeged, Department of PediatricsSzeged, Hungary. [Thurzo, Laszlo] University of Szeged, Department of OncotherapySzeged, Hungary. [McNally, Richard] Newcastle University, Paediatric and Lifecourse Epidemiology Research GroupNewcastle upon Tyne, UK. [Parker, Louise] Dalhousie University, IWK Health Centre, Department of PediatricsHalifax, Canada. RP Nyari, AT (reprint author), University of Szeged, Department of Medical Informatics, 6720 Szeged, Hungary. EM nyari@dmi.u-szeged.hu CR Alexander FE, Boyle P, Carli PM et al, 1998, Spatial temporal patterns in childhood leukaemia: further evidence for an infectious origin. EUROCLUS project. Brit J Cancer 77:812–817 Kinlen LJ, 1989, Infective cause of childhood leukaemia. Lancet 1(8634):378–379 Knox EG, Gilman EA, 1997, Hazard proximities of childhood cancers in Great Britain from 1953–80. J Epidemiol Comm Health 51:151–159 McNally RJQ, Eden TOB, 2004, An infectious aetiology for childhood leukaemia: a review of the evidence. Brit J Haematol 127:243–263 Thorne R, Hunt LP, Mott MG, 1998, Seasonality in the diagnosis of childhood acute lymphoblastic leukaemia. Brit J Cancer 77:678 Nyari TA, Kajtar P, Bartyik K, Thurzo L, Parker L, 2006, Childhood acute lymphoblastic leukaemia in relation to population mixing around the time of birth in South Hungary. Pediatric Blood Cancer 47:944–948 Jakab Zs, Balogh E, Kiss Cs, Olah E, 2002, Epidemiologic studies in a population-based childhood cancer registry in Northeast Hungary. Medical Pediatric Oncol 38:338–344 Central Demographic Agency 1982–1998 Demographic yearbook Central Demographic Agency. Budapest Stolwijk AM, Straatman H, Zielhuis GA, 1999, Studying seasonality by using sine and cosine functions in regression analysis. J Epidemiol Community Health 53:235–238 Kajtar P, Fazekasne KM, Mehes K, 2003, Month of birth in childhood acute lymphoblastic leukemia. Orv Hetil 144:1869– 1871 Westerbeek RM, Blair V, Eden OB et al, 1998, Seasonal variation in the onset of childhood leukemia and lymphoma. Brit J Cancer 78:119–124 Badrinath P, Day NE, Stockton D, 1997, Seasonality in the diagnosis of acute lymphocytic leukaemia. Brit J Cancer 75:1711–1713 Ross JA, Severson RK, Swensen AR et al, 1999, Seasonal variations in the diagnosis of childhood cancer in the United States. Brit J Cancer 81:549–553 Karimi M, Yarmohammadi H, 2003, Seasonal variations in the onset of childhood leukemia/lymphoma: April 1996 to March 2000, Shiraz, Iran. Hematol Oncol 21:51–55 Sorensen HT, Olsen JH, Rothman KJ, 2001, Seasonal variation in month of birth and diagnosis of early childhood acute lymphoblastic leukemia. JAMA 285:168–169 Feltbower RG, Pearce MS, Dickinson HO et al, 2001, Seasonality of birth for cancer in Northern England, UK. Paediatric Perinatal Epidemiol 15:338–345 Higgins CD, dos-Santos-Silva I, Stiller CA, Swerdlow AJ, 2001, Season of birth and diagnosis of children with leukaemia: an analysis of over 15 000 UK cases occurring from 1953–95. Brit J Cancer 84:406–412 Lopman BA, Reacher M et al, 2003, A summertime peak of “winter vomiting disease": surveillance of noroviruses in England and Wales, 1995 to 2002. BMC Public Health 3:13 Green MS, 1992, The male predominance in the incidence of infectious diseases in children: a postulated explanation for disparities in the literature. Int J Epidemiol 21:381– 386 Schlegel RJ, Bellatrti JA, 1969, Increased susceptibility of males to infection. Lancet ii:826–827 Peltola H, 1982, Observations on the seasonal variation of the most common acute pediatric diseases in the Helsinki area, Finland). J Comm Health 7:159–170 Dorak MT, Oguz FS, Yalman N et al, 2002, A male-specific increase in the HLA-DRB4, DR53, frequency in high-risk and relapsed childhood ALL. Leuk Res 26:651–656 Kinlen LJ, 1995, Epidemiological evidence for an infective basis in childhood leukaemia. Brit J Cancer 71:1–5 Greaves MF, 1997, Aetiology of acute leukaemia. Lancet 349:344–349 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 423 EP 428 DI 10.1007/s12253-008-9017-0 PG 6 ER PT J AU Varkonyi, J Szakaly, D Janoskuti, L Hosszufalusi, N Panczel, P Karadi, I Schoket, B AF Varkonyi, Judit Szakaly, Dora Janoskuti, Livia Hosszufalusi, Nora Panczel, Pal Karadi, Istvan Schoket, Bernadette TI Glutathione S-Transferase Enzyme Polymorphisms in a Hungarian Myelodysplasia Study Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Myelodysplastic syndrome; GSTM1; GSTT1; GSTP1; Genetic polymorphism ID Myelodysplastic syndrome; GSTM1; GSTT1; GSTP1; Genetic polymorphism AB GSTM1, GSTT1 and GSTP1 Ile105Val that are members of the GST gene family encode for Phase II drug/xenobiotic metabolizing enzymes, primarily with detoxifying function, and are polymorphic in humans. GSTM1 and GSTT1 homozygous deletion genotypes do not express the enzymes. It has been hypothesised that individuals with homozygous deletion of the GSTM1 and/or GSTT1 gene may have lower detoxification capacity towards genotoxic agents therefore those individuals may be at increased risk of myelodysplastic syndrome which is a preleukemic condition. Genetic polymorphism of GSTM1, GSTT1 and GSTP1 Ile105Val was investigated in a case–control study in a Hungarian patient population comprising 86 patients with myelodysplastic syndrome and 99 hospital-based controls. There were no statistically significant differences between cases and controls for the GSTM1, GSTT1 and GSTP1 Ile105Val genotype frequencies for any of the three genes separately and in various combinations. This suggests that these genetic polymorphisms may not be strong risk factors, if any, for myelodysplastic syndrome. C1 [Varkonyi, Judit] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Szakaly, Dora] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Janoskuti, Livia] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Hosszufalusi, Nora] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Panczel, Pal] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Karadi, Istvan] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Schoket, Bernadette] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Kornyezet-egeszsegugyi IntezetBudapest, Hungary. RP Varkonyi, J (reprint author), Semmelweis University, 3rd Department of Internal Medicine, 1125 Budapest, Hungary. EM varkjud@kut.sote.hu CR Farrow A, Jacobs A, West RR, 1989, Myelodysplasia, chemical exposure and other environmental factors. Leukemia 3:33–35 Kim SY, Choi JK, Cho YH, Chung EJ, Paek D, Chung HW, 2004, Chromosomal aberrations in workers exposed to low levels of benzene: association with genetic polymorphisms. Pharmacogenetics 14:453–463 Mannervik B, Danielson H, 1988, Glutathione transferases— structure and catalytic activity. CRC Crit Rev Biochem 23:283– 337 Vineis P, Malats N, Lang M, d’Errico A, Caporaso N, Cuzick J, Boffetta P, eds,, 1999, Metabolic polymorphisms and susceptibility to cancer. International Agency for Research on Cancer, Lyon, pp 51–62 Lin DX, Tang YM, Peng Q, Lu SX, Ambrosone CB, Kadlubar FF, 1998, Susceptibility to esophageal cancer and genetic polymorphisms in glutathione S-transferases T1, P1 and M1 and cytochrome P450 2E1. Cancer Epidemiol Biomark Prev 7:1013– 1018 Geisler S, Olshan A, 2001, GSTM1, GSTT1 and risk of head and neck cancer: a mini-HuGE review. Am J Epidemiol 154:95–105 Watson MA, Stewart PK, Smith GB, Massey TE, Bell DA, 1998, Human glutathione S-transferase P1 polymorphisms, relationship to lung tissue enzyme activity and population frequency distribution. Carcinogenesis 19:275–280 Srivastava SK, Singhal SS, Hu X, Awasthi YC, Zimniak P, Singh SV, 1999, Differential catalytic efficiency of allelic variants of human glutathione S-transferase α in catalyzing the glutathione conjugation of thiothepa. Arch Biochem Biophys 366:89–94 Lecomte T, Landi B, Beaune Ph, Puig PL, Lirot MA, 2006, Glutathione S-transferase P1 polymorphism, Ile 105 Val, predicts cumulative neuropathy in patients receiving oxiplatin-based chemotherapy. Clin Cancer Res 12:3050–3056 Chen H, Sandler DP, Taylor JA, Shore DL, Liu E, Bloomfield CD, Bell DA, 1996, Increased risk for myelodysplastic syndromes in individuals with glutathione transferase theta 1, GSTT1, gene defect. Lancet 347:295–297 Ozawa S, Schoket B, McDaniel LP, Tang Y-M, Ambrosone CB, Kostic Sz, Vincze I, Adlubar FF, 1999, Analyses of bronchial bulky DNA adduct levels and CYP2C9, GSTP1 and NQO1 genotypes in a Hungarian study population with pulmonary diseases. Carcinogenesis 20:991–995 Landi S, 2000, Mammalian class theta GST and different susceptibility to carcinogens: a review. Mutat Res 463:247–283 Cotton SC, Sharp L, Little J, Brockton N, 2000, Gluthation Stransferase polymorphisms and colorectal cancer: a HuGE review. Am J Epidemiol 151:7–32 Garte S, Gaspari L, Alexandrie AK, Ambrosone C, Autrup H, Autrup JL, Baranova H, Bathum L, Benhamou S, Boffetta P, Bouchardy C, Breskvar K, Brockmoller J, Cascorbi I, Clapper ML, Coutelle C, Daly A, Dell’Omo M, Dolzan V, Dresler CM, Fryer A, Haugen A, Hein DW, Hildesheim A, Hirvonen A, Hsieh LL, Ingelman-Sundberg M, Kalina I, Kang D, Kihara M, Kiyohara C, Kremers P, Lazarus P, Le Marchand L, Lechner MC, van Lieshout EM, London S, Manni JJ, Maugard CM, Morita S, Nazar-Stewart V, Noda K, Oda Y, Parl FF, Pastorelli R, Persson I, Peters WH, Rannug A, Rebbeck T, Risch A, Roelandt L, Romkes M, Ryberg D, Salagovic J, Schoket B, Seidegard J, Shields PG, Sim E, Sinnet D, Strange RC, Stucker I, Sugimura H, To-Figueras J, Vineis P, Yu MC, Taioli E, 2001, Metabolic gene polymorphism frequencies in control populations. Cancer Epidemiol Biomark Prev 10:1239–1248 Atoyebi W, Kusec R, Fidler C, Peto TEA, Boultwood J, Wainscoat JS, 1997, Glutathione S-transferase gene deletions in myelodysplasia. Lancet 349:1450–1451 Sasai Y, Horiike S, Misawa S, Kaneko H, Kobayashi M, Fujii H, Kashima K, Taniwaki M, 1999, Genotype of glutathione Stransferase and other genetic configurations in myelodysplasia. Leuk Res 23:975–981 Ozbek U, Aydin-Sayitoglu M, Hatirnaz O, Erensoy N, 2005, Role of CYP1A1, CYP2E1, GSTT1 and GSTM1 genes in the susceptibility to acute leukemias. Haematologica S2: Abstr 0585 Tsabouri S, Georgiou I, Alamanso I, Bourantas KL, 2000, Increased prevalence of GSTM1 null genotype in MDS patients. Acta Haematol 104:169–173 Arruda VR, Lima CS, Grignoli CR, deMelo MB, Lorand-Metze I, Alberto FL, Saad ST, Costa FF, 2001, Increased risk for acute myeloid leukaemia in individuals with glutathion S-transferase mu-1, GSTM1, and theta 1, GSTT1, gene defects. Eur J Haematol 66:383–388 Davies SM, Robinson LL, Buckley JD, Radolff GA, Ross JA, Perentesis JP, 2000, Gluthation S-transferase polymorphisms in children with myeloid leukemia: a Children’s Cancer Group study. Cancer Epidemiol Biomark Prev 9:563–566 Haase D, Schulz TG, Heutelbeck A, Fonatsch C, Harder L, Griesinger F, Hallier E, Trumper L, 2003, Polymorphisms of metabolizing enzymes, NAT2, GSTT1, GSTM1 and GSTP1, in de novo and therapy-induced MDS and AML. 7th International Symposium onMyelodysplastic Syndromes. Leuk Res 27(1):S1–S2 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 429 EP 433 DI 10.1007/s12253-008-9008-1 PG 5 ER PT J AU Murillo-Ortiz, B Perez-Luque, E Malacara, MJ Daza-Benitez, L Hernandez-Gonzalez, M Benitez-Bribiesca, L AF Murillo-Ortiz, Blanca Perez-Luque, Elva Malacara, Manuel Juan Daza-Benitez, Leonel Hernandez-Gonzalez, Martha Benitez-Bribiesca, Luis TI Expression of Estrogen Receptor Alpha and Beta in Breast Cancers of Pre- and Post-menopausal Women SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Estrogen receptor; Estradiol; Breast cancer ID Estrogen receptor; Estradiol; Breast cancer AB Expression of estrogen receptors (ER) is clinically relevant in designing therapeutic strategies. The relative importance of the two types of estrogen receptors (ER-alpha and ER-beta) in human breast cancers in preand post-menopausal women has not been properly defined. To determine the possible association between the expression of estrogen receptor and serum estradiol levels in pre- and post-menopausal women with breast cancer. 44 patients with invasive ductal carcinoma of the breast were studied and a breast tissue biopsy was taken. ER-alpha and ER-beta were detected by immunocytochemistry. Serum levels of estradiol and estrone were measured by radioimmunoassay and FSH was measured using IRMA. We studied 21 pre- and 23 post-menopausal women with breast carcinoma. Examining the number of cases with tumors positive for ER, we found no differences in the frequency of ER-alpha between pre- and post-menopausal women, but ER-beta decreased marginally after menopause (p<0.051). In cases with tumors positive for ER, the proportion of cells positive for ER-alpha was similar postmenopausally (53.95%) and pre-menopausally (57.21%), but for ER-beta the number of positive cells decreased significantly after menopause (p<0.051). In pre-menopausal women there was a correlation between serum estradiol levels and ER-beta; in post-menopausal women there was a correlation between serum FSH levels and ER-alpha. These results indicate that estradiol levels in women with mammary carcinoma are related to ER-beta expression in the breast tumor tissue. C1 [Murillo-Ortiz, Blanca] Unidad de Investigacion en Epidemiologia Clinica, Unidad Medica de Alta Especialidad No. 1 Bajio, Instituto Mexicano del Seguro Social, UMAE No. 1 Bajio, IMSS, B. Lopez Mateos Esq. Insurgentes S/N, Colonia: Los Paraisos, CP 37320 Leon, Guanajuato, Mexico. [Perez-Luque, Elva] Universidad de Guanajuato, Instituto de Investigaciones MedicasLeon, Guanajuato, Mexico. [Malacara, Manuel Juan] Universidad de Guanajuato, Instituto de Investigaciones MedicasLeon, Guanajuato, Mexico. [Daza-Benitez, Leonel] Unidad de Investigacion en Epidemiologia Clinica, Unidad Medica de Alta Especialidad No. 1 Bajio, Instituto Mexicano del Seguro Social, UMAE No. 1 Bajio, IMSS, B. Lopez Mateos Esq. Insurgentes S/N, Colonia: Los Paraisos, CP 37320 Leon, Guanajuato, Mexico. [Hernandez-Gonzalez, Martha] Unidad de Investigacion en Epidemiologia Clinica, Unidad Medica de Alta Especialidad No. 1 Bajio, Instituto Mexicano del Seguro Social, UMAE No. 1 Bajio, IMSS, B. Lopez Mateos Esq. Insurgentes S/N, Colonia: Los Paraisos, CP 37320 Leon, Guanajuato, Mexico. [Benitez-Bribiesca, Luis] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades OncologicasMexico City, DF, Mexico. RP Murillo-Ortiz, B (reprint author), Unidad de Investigacion en Epidemiologia Clinica, Unidad Medica de Alta Especialidad No. 1 Bajio, Instituto Mexicano del Seguro Social, CP 37320 Leon, Mexico. EM bomo907@hotmail.com;blanca.murillo@imss.gob.mx CR Deroo BJ, Korach KS, 2006, Estrogen receptors and human disease. J Clin Invest 116:561–570 Greene G, Gilna P, Waterfield M, Baker A, Hart Y, Shine J, 1986, Sequence and expression of human estrogen receptor complementary DNA. Science 231:1150–1154 Mosselman S, Polman J, Dijkema R, 1996, ERb: Identification and characterization of a novel human estrogen receptor. FEBS Lett 392:49–53 Moore J, Mckee D, Slentz-Kesler K, Moore L, Jones S, Horne E et al, 1998, Cloning and characterization of human estrogen receptor β isoforms. Biochem Byophis Res Commun; 247:75–78 Gustafsson JA, 2000, An update on estrogen receptors. Semin Perinatol 24:66–69 Hall JL, McDonnell P, 1999, The estrogen receptor b isoform, ERb, of the human estrogen receptor modulates ERa transcriptional activity and it a key regulator of the cellular response to estrogens and antiestrogens. Endocrinology 140:5566–5578 Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S et al, 1997, Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors, alpha, and, beta). Endocrinology 138:863–870 Speirs V, Skliris GP, Burdall SE, Carder PJ, 2002, Distinct expression patterns of ERα and ERβ in normal human mammary gland. J Clin Pathol 55:371–374 Murillo-Ortiz B, Perez-Luque E, Malacara JM, 2005, Mammographic density and the relationship with the PvuII and XbaI genotypes estrogen receptors alpha, ERa, polymorphisms. Ginecol Obstet Mex 73:229–233 Simpson ER, Mahendroot MS, Means GD et al, 1994, Aromatase cytocrome P450, the enzyme reponsible for estrogen biosynthesis. Endocrinol Rev 15:342–355 Meza-Munoz DE, Fajardo ME, Perez-Luque EL, Malacara JM, 2006, Factors associated with estrogen receptors-alpha, ERalpha, and -beta, ER-beta, and progesterone receptor abundance in obese and non obese pre- and post-menopausal women. Steroids 71(6):498–503 Bardin A, Boulle N, Lazennec G, Vignon F, Pujol P, 2004, Loss of ERb expression as a common step in estrogen-dependent tumor progression. Endocrine-Related Cancer 11:537–551 Lazennec G, Bresson D, Lucas A, Chauveau C, Vignon F, 2001, ERb inhibits proliferation and invasion of breast cancer cells. Endocrinology 142:4120–4130 Roger P, Sahla ME, Makela S, Gustafsson JA, Baldet P, Rochefort H, 2001, Decreased expression of estrogen receptor beta protein in proliferative preinvasive mammary tumors. Cancer Res 61:2537–2541 Qiu C, Shan L, Yu M, Snyderwine EG, 2005, Steroid hormone receptor expression and proliferation in rat mammary gland carcinomas induced by 2-amino-1-methyl-6-phenylimidazo [4,5- b]pyridine. Carcinogenesis 26(4):763–769 Harvey JM, Clark GM, Osborne CK, Allred CD, 1999, Estrogen receptor status by immunohistochemistry is superior to the ligandbinding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol 17(5):1474–1481 Jarvinen T, Pelto-Huikko M, Holli K, Isola J, 2000, Estrogen receptor b is coexpressed with ERa and associated with nodal status, grade, and proliferation rate in breast cancer. Am J Pathol 156:29–35 Mann S, Laucirica R, Carlson N, Younes PS, Ali N, Younes A, Li Y, Younes M, 2001, Estrogen receptor beta expression in invasive breast cancer. Human Pathol 32:113–118 Omoto Y, Inoue S, Ogawa S, Toyama T, Yamashita H, Muramatsu M, Kobayashi S, Iwase H, 2001, Clinical value of wild-type estrogen receptor beta expression in breast cancer. Cancer Lett 163:207–212 Speirs V, Parkes A, Kerin MJ, Walton DS, Carleton PJ, Fox JN, Atkin SL, 1999, Coexpression of estrogen receptor a and b: poor prognostic factors in human breast cancer? Cancer Res 59:525– 528 Speirs V, Malone C, Walton DS, Kerin MJ, Atkin SL, 1999, Increased expression of estrogen receptor b mRNA expression in tamoxifen-resistant breast cancer patients. Cancer Res 59:5421– 5424 Palmieri C, Cheng GJ, Saji S, Zelada-Hedman M, Warri A, Weihua Z, Van Noorden S, Wahlstrom T, Coombes RC, Warner M, Gustafsson JA, 2002, Estrogen receptor beta in breast cancer. Endocr-Relat Cancer 9:1–13 Jarzabek K, Koda M, Kozlowski L, Mittre H, Sulkowski S, Kottler M-L, Wolczynski S, 2005, Distinct mRNA, protein expression patterns and distribution of oestrogen receptors a and b in human primary breast cancer: correlation with proliferation marker Ki-67 and clinicopathological factors. Eur J Cancer 41:2924–2934 Zhao C, Lam WFE, Sunters A, Enmark E, Tamburo De Bella M, Coombes RC, Gustafsson JA, Dahlman-Wright K, 2003, Expression of estrogen receptor b isoforms in normal breast epithelial cells and breast cancer: regulation by methylation. Oncogene 22:7600–7606 Shoker BS, Jarvis C, Clarke RB, Anderson E, Hewlett J, Davies MP et al, 1999, Estrogen receptor-positive proliferating cells in the normal and precancerous breast. Am J Pathol 155:1811–1815 Bartow SA, 1998, Use of the autopsy to study ontogeny and expression of the estrogen receptor gene in human breast. J Mammary Gland Biol Neoplasia 3:37–48 Speirs V, Skliris GP, Burdall SE, Carder PJ, 2002, Distinct expression patterns of ER alpha and ER beta in normal human mammary gland. J Clin Pathol 55:371–374 Hall P, Ploner A, Bjohle J, Huang F, Lin C-Y, Liu ET, Miller LD, Nordgren H, Pawitan Y, Shaw P, Skoog L, Smeds J, Wedren S, Ohd J, Bergh J, 2006, Hormone-replacement therapy influences gene expression profiles and is associated with breast-cancer prognosis: a cohort study. BMC Medicine 4:16 Daling JR, Malone KE, Doody DR, Voigt LF, Bernstein L, Marchbanks PA, Coates RJ, Norman SA, Weiss LK, Ursin G, Burkman RT, Deapen D, Folger SG, McDonald JA, Simon MS, Strom BL, Spirtas R, 2003, Association of regimens of hormone replacement therapy to prognostic factors among women diagnosed with breast cancer aged 50–64 years. Cancer Epidemiol Biomarkers Prev 12(11 Pt 1):1175–1181 Sacchini V, Zurrida S, Andreoni G, Luini A, Galimberti V, Veronesi P, Intra M, Viale G, Veronesi U, 2002, Pathologic and biological prognostic factors of breast cancers in short- and longterm hormone replacement therapy users. Ann Surg Oncol 9:266– 271 Cheek J, Lacy J, Toth-Fejel S, Morris K, Calhoun K, Pommier RF, 2002, The impact of hormone replacement therapy on the detection and stage of breast cancer. Arch Surg 137:1015–1021 Collaborative Group on Hormonal Factors in Breast Cancer, 1997, Breast cancer and hormone replacement therapy: collaborative reanalysis of data from 51 epidemiological studies of 52,705 women with breast cancer and 108,411 women without breast cancer. Lancet 350:1047–1059 Brinton LA, Hoover R, Fraumeni JF, 1986, Menopausal oestrogens and breast cancer risk: an expanded case-control study. Br J Cancer 4(5):825–832 Squitieri R, Tartter PI, Ahmed S, Brower ST, Theise ND, 1994, Carcinoma of the breast in postmenopausal hormone user and nonuser control groups. J Am Coll Surg 178(2):167–170 Magnusson C, Holmberg L, Norden T, Lindgren A, Persson I, 1996, Prognostic characteristics in breast cancers after hormone replacement therapy. Breast Cancer Res Treat 38, 3):325–334 Bonnier P, Bessenay F, Sasco AJ, Beedassy B, Lejeune C, Romain S, Charpin C, Piana L, Martin PM, 1998, Impact of menopausal hormone-replacement therapy on clinical and laboratory characteristics of breast cancer. Int JCancer 79(3):278–282 Holli K, Isola J, Cuzick J, 1998, Low biologic aggressiveness in breast cancer in women using hormone replacement therapy. J Clin Oncol 16(9):3115–3120 O’Connor IF, Shembekar MV, Shousha S, 1998, Breast carcinoma developing in patients on hormone replacement therapy: a histological and immunohistological study. J Clin Pathol 51, 12):935–938 Carter AC, Sedransk N, Kelley RM, Ansfield FJ, Ravdin RG, Talley RW, Potter NR, 1977, Diethylsilbestrol: recommended dosages for different categories of breast cancer patients Report of the Cooperative Breast Cancer Group. JAMA 237:2079–2085 Cappelletti V, Celio L, Bajetta E, Allevi A, Longarini R, Miodini P, Villa R, Fabbri A, Mariani L, Giovanazzi R, Galante E, Greco M, Daidone GM, 2004, Prospective evaluation of estrogen receptor-b in predicting response to neoadjuvant antiestrogen therapy in elderly breast cancer patients.. Endocr Relat Cancer 11, 4):761–770, Dec Nonni A, Zagouri F, Sergentanis TN, Lazaris AC, Patsouris ES, Zografos GC, 2007, Immunohistochemical expression of estrogen receptors alpha and beta in lobular neoplasia. Virchows Arch 451, 5):893–897, Nov. Epub 2007 Oct 9 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 435 EP 442 DI 10.1007/s12253-008-9088-y PG 8 ER PT J AU Bujas, T Pavic, I Lenicek, T Mijic, A Kruslin, B Davor, T AF Bujas, Tatjana Pavic, Ivana Lenicek, Tanja Mijic, August Kruslin, Bozo Davor, Tomas TI Peritumoral Retraction Clefting Correlates with Advanced Stage Squamous Cell Carcinoma of the Esophagus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophagus; Metastases; Peritumoral clefting; Squamous cell carcinoma; Tumor stage ID Esophagus; Metastases; Peritumoral clefting; Squamous cell carcinoma; Tumor stage AB The present study was designated to analyze correlation between the presence and extent of peritumoral retraction clefting and various clinicopathologic features in esophageal squamous cell carcinoma (ESCC), and to possibly establish the significance of this phenomenon in ESCC. Fifty-four consecutive patients with advanced ESCC were included in the study. The presence of peritumoral retraction clefting was classified on the basis of the proportion of tumor nests exhibiting this phenomenon. Tumors with clefts that affected up to 25% of tumor nests were classified as group I; with clefts that affected >25% to 50% of tumor nests as group II; with clefts that affected >50% to 75% of tumor nests as group III; and tumors with clefts that affected more than 75% of tumor nests were classified as group IV. Statistical analysis showed a correlation between presence and extent of peritumoral clefting and lymph node metastasis. T3 tumors and tumors with lymph node metastasis had significantly more pronounced peritumoral clefting compared with T2 tumors and tumors without lymph node metastasis. The presence of peritumoral clefting was not associated with the number of affected lymph nodes. There was no correlation between the presence and extent of peritumoral clefting with patient age and sex, and tumor location, diameter and grade. The association of peritumoral retraction clefting in ESCC with local invasiveness and lymph node metastasis indicated that peritumoral clefting could be a simple and useful morphological feature of tumor aggressiveness and may contribute to the pathological and clinical assessment of patients with ESCC. C1 [Bujas, Tatjana] Karlovac General Hospital, Department of Pathology, Andrije Stampara 3, HR-47000 Karlovac, Croatia. [Pavic, Ivana] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska 29, HR-10000 Zagreb, Croatia. [Lenicek, Tanja] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska 29, HR-10000 Zagreb, Croatia. [Mijic, August] Sestre Milosrdnice University Hospital, University Department of Surgery, Vinogradska 29, HR-10000 Zagreb, Croatia. [Kruslin, Bozo] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska 29, HR-10000 Zagreb, Croatia. [Davor, Tomas] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska 29, HR-10000 Zagreb, Croatia. RP Davor, T (reprint author), University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, HR-10000 Zagreb, Croatia. EM dtomas@kbsm.hr CR Hashibe M, Boffetta P, Janout Vet al, 2007, Esophageal cancer in Central and Eastern Europe: tobacco and alcohol. Int J Cancer 120:1518–1522 Strnad M, ed,, 2007, National Cancer Registry. Cancer incidence in Croatia 2005. Bulletin No.30, Croatian National Institute of Public Health, Zagreb Hamilton SR, Aaltonen LA, eds,, 2001, WHO classification of tumours. Pathology and genetics of tumours of the digestive system. IARC, Lyon Sleeman JP, 2000, The lymph node as a bridgehead in the metastatic dissemination of tumors. Recent Results Cancer Res 157:55–81 Clarijs R, Ruiter DJ, de Waal RM, 2001, Lymphangiogenesis in malignant tumours: does it occur? J Pathol 193:143–146 Tomas D, Kruslin B, 2004, The potential value of, myo, fibroblastic stromal reaction in the diagnosis of prostatic adenocarcinoma. Prostate 61:324–331 Cheng MF, Tzao C, Tsai WC et al, 2006, Expression of EMMPRIN and matriptase in esophageal squamous cell carcinoma: correlation with clinicopathological parameters. Dis Esophagus 19:482–486 Kruslin B, Tomas D, Rogatsch H et al, 2003, Periacinar retraction clefting in the prostatic needle core biopsies: an important diagnostic criterion or a simple artifact? Virchows Arch 443:524–527 Bell KA, Smith Sehdev AE, Kurman RJ, 2001, Refined diagnostic criteria for implants associated with ovarian atypical proliferative serous tumors, borderline, and micropapillary serous carcinomas. Am J Surg Pathol 25:419–423 Acs G, Dumoff KL, Solin LJ et al, 2007, Extensive retraction artifact correlates with lymphatic invasion and nodal metastasis and predicts poor outcome in early stage breast carcinoma. Am J Surg Pathol 31:129–140 Irie J, Manucha V, Ioffe OB et al, 2007, Artefact as the pathologist’s friend: peritumoral retraction in in situ and infiltrating duct carcinoma of the breast. Int J Surg Pathol 15:53–59 Kruslin B, Tomas D, Rogatsch H et al, 2005, Correlation of periacinar retraction clefting in needle core biopsies and corresponding prostatectomy specimens of patients with prostatic adenocarcinoma. Int J Surg Pathol 13:67–72 Ulamec M, Tomas D, Ensinger C et al, 2007, Periacinar retraction clefting in proliferative prostatic atrophy and prostatic adenocarcinoma. J Clin Pathol 60:1098–1101 Sobin LH, Wittekind CH, eds,, 2002, TNM classification of malignant tumors, 6th ed. Wiley-Liss, New York Moore RD, Stevenson J, Schoenberg MD, 1960, The response of connective tissue associated with tumors of the skin. Am J Clin Pathol 34:125–130 Halpert B, Sheehan EA, Schmalhorst WR et al, 1963, Carcinoma of the prostate: a survey of 5000 autopsies. Cancer 16:736–742 Halpert B, Schmalhorst WR, Scott RJ, 1966, Carcinoma of the prostate in patients 70 to 79 years old. Cancer 19:695–698 Mckenney JK, Gomez JA, Desai S et al, 2001, Morphologic expressions of urothelial carcinoma in situ: a detailed evaluation of its histologic patterns with emphasis on carcinoma in situ with microinvasion. Am J Surg Pathol 25:356–362 Kruslin B, Tomas D, Cviko A et al, 2006, Periacinar clefting and p63 immunostaining in prostatic intraepithelial neoplasia and prostatic carcinoma. Pathol Oncol Res 12:205–209 Tomas D, Ulamec M, Hudolin T et al, 2006, Myofibroblastic stromal reaction and expression of tenascin-C and laminin in prostate adenocarcinoma. Prostate Cancer Prostatic Dis 9:414–419 Ishibashi S, Ohashi Y, Suzuki T et al, 2006, Tumor-associated tissue eosinophilia in human esophageal squamous cell carcinoma. Anticancer Res 26:1419–1424 Guo SJ, Lin DM, Li J et al, 2007, Tumor-associated macrophages and CD3-zeta expression of tumor-infiltrating lymphocytes in human esophageal squamous-cell carcinoma. Dis Esophagus 20:107–116 Nakanishi Y, Ochiai A, Kato H et al, 2001, Clinicopathological significance of tumor nest configuration in patients with esophageal squamous cell carcinoma. Cancer 91:1114–1120 Nakayama Y, Matsumoto K, Nagato M et al, 2007, Significance of lymphangiogenesis as assessed by immunohistochemistry for podoplanin in patients with esophageal carcinoma. Anticancer Res 27:619–625 Vallbohmer D, Lenz HJ, 2006, Predictive and prognostic molecular markers in outcome of esophageal cancer. Dis Esophagus 19:425–432 Ingber DE, 2002, Cancer as a disease of epithelial–mesenchymal interactions and extracellular matrix regulation. Differentiation 70:547–560 Wever DO, Mareel M, 2003, Role of tissue tumor stroma in cancer cell invasion. J Pathol 200:429–447 Tomakidi P, Stark HJ, Herold-Mende C et al, 2003, Discriminating expression of differentiation markers evolves in transplants of benign and malignant human skin keratinocytes through stromal interactions. J Pathol 200:298–307 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 443 EP 447 DI 10.1007/s12253-008-9038-8 PG 5 ER PT J AU Yuan, A Liu, J Liu, Y Bjornsen, T Varro, A Cui, G AF Yuan, Aping Liu, Jinzhong Liu, Yiqing Bjornsen, Tone Varro, Andrea Cui, Guanglin TI Immunohistochemical Examination of Gastrin, Gastrin Precursors, and Gastrin/CCK-2 Receptor in Human Esophageal Squamous Cell Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophagus; Squamous cell carcinoma; Gastrin; Immunohistochemistry ID Esophagus; Squamous cell carcinoma; Gastrin; Immunohistochemistry AB A promoting effect of gastrin on stimulating Barrett’s oesophagus proliferation has been demonstrated, but whether it plays a regulating role for esophageal squamous cell carcinoma (ESCC) to date has not been fully investigated. The aim of this study is to examine the expressions of gastrin, gastrin precursors and gastrin/CCK-2 receptor in ESCC. Tissue specimen sections from 38 patients with ESSC obtained from a high incidence area of north China were assessed using immunohistochemistry for amidated gastrin, gastrin precursors (progastrin and glycine-extended gastrin) and gastrin/CCK-2 receptors. Their clinical histopathological significance was also analyzed. Of 38 ESCC, the immunoreactivities of gastrin, glycineextended gastrin and progastrin were observed in 13.2% (5/38), 7.9% (3/38) and 23.68% (9/38) cases. The expression of progastrin was obviously higher than other gastrins, though not significantly (P>0.05). In positive cases for gastrin or glycine-extended gastrin, the scores of positive tumor cell numbers were at a lower density (<10/abundantdistributed field). However, the scores of progastrin positive tumor cell density in five of nine positive cases were over 10/abundant-distributed field. The immunoreactivity of gastrin/CCK-2 receptor was also observed in 15.8% (6/38) ESCC cases. There was not significant correlation regarding immunohistochemical results with known histomorphological parameters i.e. gender, tumor location and TNM stages. Based on our current results, ESCC tumor cells could be a possible cellular source of gastrin precursors, which has been postulated to play a role in regulating the growth in some human tumor cells. C1 [Yuan, Aping] Faculty of Medicine, University of Tromso, Institute of Clinical Medicine, Laboratory of Gastroenterology, 9037 Tromso, Norway. [Liu, Jinzhong] the Fourth Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, Henan, China. [Liu, Yiqing] the Second Affiliated Hospital of Zhengzhou University, Department of MedicineZhengzhou, Henan, China. [Bjornsen, Tone] University Hospital of North, Department of PathologyTromso, Norway. [Varro, Andrea] University of Liverpool, Physiology LaboratoryLiverpool, UK. [Cui, Guanglin] Faculty of Medicine, University of Tromso, Institute of Clinical Medicine, Laboratory of Gastroenterology, 9037 Tromso, Norway. RP Cui, G (reprint author), Faculty of Medicine, University of Tromso, Institute of Clinical Medicine, Laboratory of Gastroenterology, 9037 Tromso, Norway. EM guanglin.cui@fagmed.uit.no CR Parkin DM, Bray FI, Devesa SS, 2001, Cancer burden in the year 2000. The global picture. Eur J Cancer 37(Suppl 8):S4–S66 Montesano R, Hollstein M, Hainaut P, 1996, Genetic alterations in esophageal cancer and their relevance to etiology and pathogenesis: a review. Int J Cancer 69:225–235 Zhi H, Wang L, Zhang J et al, 2006, Significance of COX-2 expression in human esophageal squamous cell carcinoma. Carcinogenesis 27:1214–1221 Imsumran A, Adachi Y, Yamamoto H et al, 2007, Insulin-like growth factor-I receptor as a marker for prognosis and a therapeutic target in human esophageal squamous cell carcinoma. Carcinogenesis 28:947–956 Sutter AP, Hopfner M, Huether A et al, 2006, Targeting the epidermal growth factor receptor by erlotinib, Tarceva, for the treatment of esophageal cancer. Int J Cancer 118:1814–1822 Fang MZ, Liu C, Song Y et al, 2004, Over-expression of gastrinreleasing peptide in human esophageal squamous cell carcinomas. Carcinogenesis 25:865–871 Xie JJ, Xu LY, Zhang HH et al, 2005, Role of fascin in the proliferation and invasiveness of esophageal carcinoma cells. Biochem Biophys Res Commun 337:355–362 Dockray GJ, Varro A, Dimaline R et al, 2001, The gastrins: their production and biological activities. Annu Rev Physiol 63:119–139 Ferrand A, Wang TC, 2006, Gastrin and cancer: a review. Cancer Lett 238:15–29 Aly A, Shulkes A, Baldwin GS, 2004, Gastrins, cholecystokinins and gastrointestinal cancer. Biochim Biophys Acta 1704:1–10 Takhar AS, Eremin O, Watson SA, 2004, The role of gastrin in colorectal carcinogenesis. Surgeon 2:251–257 Singh P, Dai B, Wu H et al, 2000, Role of autocrine and endocrine gastrin-like peptides in colonic carcinogenesis. Curr Opin Gastroenterol 16:68–77 Caplin M, Savage K, Khan K et al, 2000, Expression and processing of gastrin in pancreatic adenocarcinoma. Br J Surg 87:1035–1040 Rengifo-Cam W, Singh P, 2004, Role of progastrins and gastrins and their receptors in GI and pancreatic cancers: targets for treatment. Curr Pharm Des 10:2345–2358 Goetze JP, Nielsen FC, Burcharth F et al, 2000, Closing the gastrin loop in pancreatic carcinoma: coexpression of gastrin and its receptor in solid human pancreatic adenocarcinoma. Cancer 88:2487–2494 Caplin M, Khan K, Savage K et al, 1999, Expression and processing of gastrin in hepatocellular carcinoma, fibrolamellar carcinoma and cholangiocarcinoma. J Hepatol 30:519–526 Moody TW, 2006, Peptide hormones and lung cancer. Panminerva Med 48:19–26 Schaer JC, Reubi JC, 1999, High gastrin and cholecystokinin, CCK, gene expression in human neuronal, renal, and myogenic stem cell tumors: comparison with CCK-A and CCK-B receptor contents. J Clin Endocrinol Metab 84:233–239 Negre F, Fagot-Revurat P, Bouisson M et al, 1996, Autocrine stimulation of AR4-2J rat pancreatic tumor cell growth by glycine-extended gastrin. Int J Cancer 66:653–658 Hoosein NM, Kiener PA, Curry RC et al, 1990, Evidence for autocrine growth stimulation of cultured colon tumor cells by a gastrin/cholecystokinin-like peptide. Exp Cell Res 186:15–21 Harris JC, Clarke PA, Awan A et al, 2004, An antiapoptotic role for gastrin and the gastrin/CCK-2 receptor in Barrett’s esophagus. Cancer Res 64:1915–1919 Baldwin GS, 1995, The role of gastrin and cholecystokinin in normal and neoplastic gastrointestinal growth. J Gastroenterol Hepatol 10:215–232 Seva C, Dickinson CJ, Yamada T, 1994, Growth-promoting effects of glycine-extended progastrin. Science 265:410–412 Henwood M, Clarke PA, Smith AM et al, 2001, Expression of gastrin in developing gastric adenocarcinoma. Br J Surg 88:564– 568 Kelly A, Hollande F, Shulkes A et al, 1998, Expression of progastrin-derived peptides and gastrin receptors in a panel of gastrointestinal carcinoma cell lines. J Gastroenterol Hepatol 13:208–214 Koh TJ, Dockray GJ, Varro A et al, 1999, Overexpression of glycine-extended gastrin in transgenic mice results in increased colonic proliferation. J Clin Invest 103:1119–1126 Ottewell PD, Watson AJ, Wang TC et al, 2003, Progastrin stimulates murine colonic epithelial mitosis after DNA damage. Gastroenterology 124:1348–1357 Singh P, Velasco M, Given R et al, 2000, Progastrin expression predisposes mice to colon carcinomas and adenomas in response to a chemical carcinogen. Gastroenterology 119:162–171 Cui G, Koh TJ, Chen D et al, 2004, Overexpression of glycineextended gastrin inhibits parietal cell loss and atrophy in the mouse stomach. Cancer Res 64:8160–8166 Moore TC, Jepeal LI, Boylan MO et al, 2004, Gastrin stimulates receptor-mediated proliferation of human esophageal adenocarcinoma cells. Regul Pept 120:195–203 Van Nieuwenhove Y, De Backer T, Chen D et al, 1998, Gastrin stimulates epithelial cell proliferation in the oesophagus of rats. Virchows Arch 432:371–375 Karaki Y, Shimazaki K, Okamoto M et al, 1996, Effect of endogenous hypergastrinemia on carcinogenesis in the rat esophagus. Surg Today 26:5–11 Haigh CR, Attwood SE, Thompson DG et al, 2003, Gastrin induces proliferation in Barrett’s metaplasia through activation of the CCK2 receptor. Gastroenterology 124:615–625 Abdalla SI, Lao-Sirieix P, Novelli MR et al, 2004, Gastrininduced cyclooxygenase-2 expression in Barrett’s carcinogenesis. Clin Cancer Res 10:4784–4792 Munoz NDN, 1996, Esophageal cancer. In: Schottenfeld D, Fraumeni JF Jr, eds, Cancer epidemiology and prevention. Oxford University Press, New York, pp 681–706 Li MX, Cheng SJ, 1984, Carcinogenesis of esophageal cancer in Linxian, China. Chin Med J, Engl, 97:311–316 Sobin LH, Wittekind CE, 2002, UICC TNM classification of malignant tumors, 6th edn. Wiley, New York Varro A, Dockray GJ, Bate GW et al, 1997, Gastrin biosynthesis in the antrum of patients with pernicious anemia. Gastroenterology 112:733–741 Koh TJ, Field JK, Varro A et al, 2004, Glycine-extended gastrin promotes the growth of lung cancer. Cancer Res 64:196–201 Tezel E, Nagasaka T, Tezel G et al, 2002, Characterization of scattered neuroendocrine cells in ductal carcinoma of the pancreas. Pancreas 25:136–141 Ogunwobi OO, Beales IL, 2006, Glycine-extended gastrin stimulates proliferation and inhibits apoptosis in colon cancer cells via cyclooxygenase-independent pathways. Regul Pept 134:1–8 Harris JC, Gilliam AD, McKenzie AJ et al, 2004, The biological and therapeutic importance of gastrin gene expression in pancreatic adenocarcinomas. Cancer Res 64:5624–5631 Stepan VM, Sawada M, Todisco A et al, 1999, Glycine-extended gastrin exerts growth-promoting effects on human colon cancer cells. Mol Med 5:147–159 Iwase K, Evers BM, Hellmich MR et al, 1997, Regulation of growth of human gastric cancer by gastrin and glycine-extended progastrin. Gastroenterology 113:782–790 Wang TC, Dockray GJ, 1999, Lessons from genetically engineered animal models. I. Physiological studies with gastrin in transgenic mice. Am J Physiol 277:G6–G11 Savage K, Waller HA, Stubbs M et al, 2006, Targeting of cholecystokinin B/gastrin receptor in colonic, pancreatic and hepatocellular carcinoma cell lines. Int J Oncol 29:1429–1435 Van Nieuwenhove Y, Chen D, Willems G, 2001, Postprandial cell proliferation in the esophageal epithelium of rats. Regul Pept 97:131–137 Poorkhalkali N, Helander HF, 2003, Cell kinetics of the oesophageal epithelium in the rat: effects of hypergastrinaemia. Scand J Gastroenterol 38:14–19 Singh P, Owlia A, Espeijo R et al, 1995, Novel gastrin receptors mediate mitogenic effects of gastrin and processing intermediates of gastrin on Swiss 3T3 fibroblasts. Absence of detectable cholecystokinin, CCK)-A and CCK-B receptors. J Biol Chem 270:8429–8438 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 449 EP 455 DI 10.1007/s12253-008-9047-7 PG 7 ER PT J AU Ersoz, S Sert, H Yandi, M Erem, C Mungan, S Ersoz, OH Cobanoglu, U Hacihasanoglu, A AF Ersoz, Safak Sert, Hikmet Yandi, Mustafa Erem, Cihangir Mungan, Sevdegul Ersoz, Onder Halil Cobanoglu, Umit Hacihasanoglu, Arif TI The Significance Of Galectin-3 Expression in the Immunocytochemical Evaluation of Thyroid Fine Needle Aspiration Cytology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thyroid neoplasms; Galectin-3 ID Thyroid neoplasms; Galectin-3 AB The aim of this study is to evaluate the significance of immunohistochemical expression of Galectin-3 in the differential diagnosis of benign and malignant thyroid nodules. We studied the fine needle aspiration specimens of 38 patients who had evaluated for nodular goiter and undergone a thyroid surgery between 2004–2005. Slides had been stained immunocytochemically with Galectin-3. The cytoplasmic staining of Galectin-3 was analyzed. Three cases of five follicular carcinomas had positive staining for Galectin-3, while two had not. Two cases with follicular adenomas were negative for Galectin-3. Five cases of six papillary carcinomas had positive staining for Galectin-3, while one case (the case with a papillary microcarcinoma) had not. The single cases with medullary and anaplastic carcinomas were negative for Galectin-3. None of the cases with a benign thyroid pathology had positive staining for Galectin-3. Galectin-3 immunocytochemical staining, had a sensitivity of 61.5%, specificity of 100%, positive predictive value of 100%, and negative predictive value of 83.3% for thyroid malignancies. For the evaluation of follicular neoplasm, Galectin-3 immunocytochemical staining had a sensitivity of 60%, specificity of 100%, positive predictive value of 100%, and negative predictive value of 50%. Galectin-3 expression in thyrocytes is a strong indicator of a malignant proliferative lesion especially for papillary and to an extent in follicular thyroid neoplasms. Galectin-3 could be used as a supplementary marker for cytological diagnosis. C1 [Ersoz, Safak] Karadeniz Technical University, Faculty of Medicine, Department of Pathology, 61080 Trabzon, Turkey. [Sert, Hikmet] Karadeniz Technical University, Faculty of Medicine, Department of General Surgery, 61080 Trabzon, Turkey. [Yandi, Mustafa] Karadeniz Technical University, Faculty of Medicine, Department of General Surgery, 61080 Trabzon, Turkey. [Erem, Cihangir] Karadeniz Technical University, Faculty of Medicine, Department of Endocrinology, 61080 Trabzon, Turkey. [Mungan, Sevdegul] Karadeniz Technical University, Faculty of Medicine, Department of Pathology, 61080 Trabzon, Turkey. [Ersoz, Onder Halil] Karadeniz Technical University, Faculty of Medicine, Department of Endocrinology, 61080 Trabzon, Turkey. [Cobanoglu, Umit] Karadeniz Technical University, Faculty of Medicine, Department of Pathology, 61080 Trabzon, Turkey. [Hacihasanoglu, Arif] Karadeniz Technical University, Faculty of Medicine, Department of Endocrinology, 61080 Trabzon, Turkey. RP Ersoz, S (reprint author), Karadeniz Technical University, Faculty of Medicine, Department of Pathology, 61080 Trabzon, Turkey. EM ersozs@yahoo.com CR Gasbarri A, Martegani MP, Del Prete F, Lucante T, Natali PG, Bartolazzi A, 1999, Galectin-3 and CD44v6 isoforms in the preoperative evaluation of thyroid nodules. J Clin Oncol 17:3494– 3502 Nunez C, Mendelsohn G, 1989, Fine-needle aspiration and needle biopsy of the thyroid gland. Pathol Annu 24:161–198 Gharib H, 1994, Fine-needle aspiration biopsy of thyroid nodules: advantages, limitations and effect. Mayo Clin Proc 69:44–49 Segura MEA, Magalhaes AV, 2005, Galectin-3 immunostaining in the thyroid neoplasms. J Bras Patol Med Lab 41:341–346 Caraway NP, Sneige N, Saman NA, 1993, Diagnostic pitfalls in thyroid fine-needle aspiration: a review of 394 cases. Diagn Cytopathol 9:345–350 Chiarotti L, Berlingieri MT, De Rosa P et al, 1992, Increased expression of the negative growth factor, galactoside-binding protein gene, in transformed thyroid cells and in human thyroid carcinomas. Oncogene 71:2507–2511 Inohara H, Akahani S, Koths K, Raz A, 1996, Interactions between galectin-3 and Mac-2-binding protein mediate cell–cell adhesion. Cancer Res 56:4530–4534 Inohara H, Raz A, 1995, Functional evidence that cell surface galectin-3 mediates homotypic cell adhesion. Cancer Res 55: 3267–3271 Bartolazzi A, Gasbarri A, Papotti M et al, 2001, Application of an immunodiagnostic method for improving preoperative diagnosis of nodular thyroid lesions. Lancet 357:1644–1650 Inohara H, Honjo Y, Yoshii T et al, 1999, Expression of galectin-3 in fine-needle aspirates as a diagnostic marker differentiating benign from malignant thyroid neoplasms. Cancer 85:2475–2484 Cvejic D, Savin S, Paunovic I, Tatic S, Havelka M, Sinadinovic J, 1998, Immunohistochemical localization of galectin-3 in malignant and benign human thyroid tissue. Anticancer Res 18:2637–2641 Schlinkert RT, van Heerden JA, Goelnerr JR et al, 1997, Factors that predict malignant thyroid lesions when fine-needle aspiration is “suspicious for follicular neoplasm”. Mayo Clin Proc 72:913–916 Tuttle RM, Lemar H, Burch HB, 1998, Clinical features associated with an increased risk of thyroid malignancy in patients follicular neoplasia by fine-needle aspiration. Thyroid 8:377–383 Xu XC, El-Naggar AK, Lotan R, 1995, Differential expression of galectin-1 and galectin-3 in thyroid tumors. Potential diagnostic implications. Am J Pathol 147:815–822 Kim MJ, Kim HJ, Hong SJ, Shong YK, Gong G, 2006, Diagnostic utility of galectin-3 in aspirates of thyroid follicular lesions. Acta Cytol 50:28–34 Mehrotra P, Okpokam A, Bouhaidar R et al, 2004, Galectin-3 does not reliably distinguish benign from malignant thyroid neoplasms. Histopathology 45:493–500 Kovacs RB, Foldes J, Winkler G, Bodo M, Sapi Z, 2003, The investigation of galectin-3 in diseases of the thyroid gland. Eur J Endocrinol 149:449–453 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 457 EP 460 DI 10.1007/s12253-008-9041-0 PG 4 ER PT J AU Kurita, H Kamata, T Koike, T Kobayashi, H Kurashina, K AF Kurita, Hiroshi Kamata, Takahiro Koike, Takeshi Kobayashi, Hiroichi Kurashina, Kenji TI Intraoperative Tissue Staining of Invaded Oral Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intraoperative; Surgery; Tissue staining; Invasion; Surgical margin ID Intraoperative; Surgery; Tissue staining; Invasion; Surgical margin AB The purpose of this study was to assess the ability of intraoperative tissue staining with consecutive application of 0.4% indigo carmine and 0.5% Congo red to demonstrate the extent and border of oral carcinoma invasion. Seventeen patients were included in the study. Once the oral tumor was resected, a vertical section of surgical specimen was taken from the central part of the tumor. The extent and border of the invaded carcinoma were assessed on digital microscopic examination with tissue staining. The results of assessments were compared with corresponding results of conventional histopathological analysis with HE staining, which is considered the gold standard. Tissue staining produced a brown-black stain on normal muscle, connective, and salivary tissues but not tumor and epithelial tissues. It clearly demonstrated the extent and border of tumor invasion in 13 of 17 patients (76.5%); however, detection of remnant vital tumor cells in scar tissue after neoadjuvant chemotherapy, and distinction between the tumor and adipose tissue scattered in the muscle tissue was difficult. The results of this study showed that intraoperative tissue staining was a possible method in demonstrating the extent and border of carcinoma deeply invaded in the soft tissue and selecting the site for additional frozen section analysis, although the method needed some refinement. C1 [Kurita, Hiroshi] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. [Kamata, Takahiro] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. [Koike, Takeshi] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. [Kobayashi, Hiroichi] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. [Kurashina, Kenji] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. RP Kurita, H (reprint author), Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 390-8621 Matsumoto, Japan. EM hkurita@shinshu-u.ac.jp CR Lee JG, 1974, Detection of residual carcinoma of the oral cavity, oropharynx: a study of surgical margins. Trans Am Acad Ophthalmol Otolaryngol 78:49–53 Byers RM, Bland KI, Borlase B, Luna M, 1978, The prognostic and therapeutic value of frozen section determinations in the surgical treatment of squamous carcinoma of the head and neck. Am J Surg 136:525–528 Ribeiro NFF, Godden DRP, Wilson GE, Butterworth DM, Woodwards RTM, 2003, Do frozen sections help achieve adequate surgical margins in the resection of oral carcinoma? Int J Oral Maxillofac Surg 32:152–158 Gooris PJJ, Vermey B, de Visscher JGAM, Roodenburg JLN, 2003, Frozen section examination of the margins for resection of squamous cell carcinoma of the lower lip. J Oral Maxillofac Surg 61:890–894 Kurita H, Uehara S, Funamoto S, Nakatsuka A, Kobayashi H, Kurashina K, 2006, Intraoperative digital microscopic assessment of the deep surgical margins in oral carcinoma surgery: a preliminary report. Am J Surg 191:84–88 Jung M, Kiesslich R, 1999, Chromoendoscopy and intravital staining techniques. Baillieres Best Pract Res Clin Gastroenterol 13:11–19 Fennerty MB, 1999, Tissue staining, chromoscopy, of the gastrointestinal tract. Can J Gastroenterol 13:423–429 Tatsua M, Okuda S, Tamura H, Taniguchi H, 1982, Endoscopic diagnosis of early gastric cancer by the endoscopic Congo redmethylene blue test. Cancer 50:2956–2960 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 461 EP 465 DI 10.1007/s12253-008-9074-4 PG 5 ER PT J AU Mejia, C Ruiz-Azuara, L AF Mejia, Carmen Ruiz-Azuara, Lena TI Casiopeinas IIgly and IIIia Induce Apoptosis in Medulloblastoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cell survival; Cisplatin; MTT method; Mitochondria; Actin; Copper ID Cell survival; Cisplatin; MTT method; Mitochondria; Actin; Copper AB The medulloblastoma is a tumor of the central nervous system that is expressed in childhood. Casiopeinas® are a family of molecules with an active copper 2+ core and an amino acid sequence that seem give them tumoral specificity. The mechanism of action is poorly understood; however, it has been reported that some metals such as copper and some of their complexes are toxic due to their high potential to participate in redox reactions which could cause apoptosis in medulloblastoma cells. Cell survival was measured by the MTT method and apoptosis was identified by the presence of condensed nuclei, disruption of the mitochondrial transmembrane potential, and cytoskeleton disorder. In all cases medulloblastoma cells treated with Casiopeinas showed more apoptotic features than untreated cells. Casiopeinas IIgly and IIIia promise to be important compounds for the treatment of medulloblastoma, mainly by their ability to induce apoptosis. C1 [Mejia, Carmen] UNAM-Campus Juriquilla, Instituto de Neurobiologia, Departamento de Neurobiologia Molecular y Celular, Boulevard Universitario 3001, C.P. 76230 Queretaro, Mexico. [Ruiz-Azuara, Lena] UNAM, Facultad de Quimica, Departamento de Quimica InorganicaMexico City, Mexico. RP Mejia, C (reprint author), UNAM-Campus Juriquilla, Instituto de Neurobiologia, Departamento de Neurobiologia Molecular y Celular, C.P. 76230 Queretaro, Mexico. EM maria.c.mejia@uv.es CR Allen JC, Bloom J, Ertel I et al, 1986, Brain tumors in children: current cooperative and institutional chemotherapy trials in newly diagnosed and recurrent disease. Semin Oncol 13:110–122 Bloom HJ, 1982, Medulloblastoma in children. Increasing survival rates and further prospects. Int J Radiat Oncol Biol Phys 8:2023–2027 Charras GT, Yarrow JC, Horton MA et al, 2005, Non-equilibration of hydrostatic pressure in blebbing cells. Nature 435:365–369 Coleman ML, Sahai EA, Yeo M et al, 2001, Membrane blebbing during apoptosis results from caspase-mediated activation of ROCK I. Nat Cell Biol 3:339–345 Cotter TG, Lennon SV, Glynn JM et al, 1992, Microfilamentdisrupting agents prevent the formation of apoptotic bodies in tumor cells undergoing apoptosis. Cancer Res 52:997–1005, Erratum in: Cancer Res 52:3512, 1992 Davis S, Weiss MJ, Wong JR et al, 1985, Mitochondria and plasma membrane potentials cause unusual accumulation and retention of rhodamine 123 by human breast adenocarcinomaderived MCF-7 cells. J Biol Chem 260:13844–13850 Decaudin D, Marzo I, Brenner C et al, 1998, Mitochondria in chemotherapy-induced apoptosis: a prospective novel target of cancer therapy. Int J Oncol 12:141–152 Ferrer G, Ruiz-Ramirez L, Radi R, 1997, Ternary copper complexes and manganese, III, tetrakis, 4-benzoic acid)(porphyrin, catalyze peroxynitrite-dependent nitration of aromatics. J Chemical Res Toxicol 10:1338–1344 Fuentes-Noriega I, Ruiz-Ramirez L, Tovar Tovar A et al, 2002, Development and validation of a liquid chromatographic method for Casiopeina IIIi in rat plasma. J Chromatogr B Analyt Technol Biomed Life Sci 772:115–121 Koya K, Li Y, Wang H et al, 1996, MKT-077, a novel rhodacyanine dye in clinical trials, exhibits anticarcinoma activity in preclinical studies based on selective mitochondrial accumulation. Cancer Res 56:538–543 Marin-Hernandez A, Gracia-Mora I, Ruiz-Ramirez L et al, 2003, Toxic effects of copper-based antineoplastic drugs, Casiopeinas, on mitochondrial functions. Biochem Pharmacol 65:1979–1989 Moss DK, Betin VM, Malesinski SD et al, 2006, A novel role for microtubules in apoptotic chromatin dynamics and cellular fragmentation. J Cell Sci 119:2362–2374 Nicotera P, Melino G, 2004, Regulation of the apoptosis-necrosis switch. Oncogene 23:2757–2765 Ruiz-Azuara L, 1994, Procedimiento para la obtencion de complejos metalicos como agentes anticancerigenos. Tipo I. Patente de invencion en tramite, SECOFI 18801. P. I., 1990). Patente, Enero 26, no. 172967 Siro S, Minoru T, Kazuo U et al, 1998, Requirement of caspase-3, -like, protease-mediated hydrogen peroxide production for apoptosis induced by various anticancer drugs. J Biol Chem 273:26900–26907 Skehan P, Storeng R, Scudiero D et al, 1990, New colorimetric cytotoxicity assay for anticancer-drug screening. J Natl Cancer Inst 82:1107–1112 Titulo de Marca, 1992, Casiopeina. Reg. 407543 SECOFI Trejo-Solis C, Palencia G, Zuniga S et al, 2005, Cas IIgly induces apoptosis in glioma C6 cells in vitro and in vivo through caspasedependent and caspase-independent mechanisms. Neoplasia 7:563–574 Van GM, Festjens N, Van LG et al, 2003, Mitochondrial intermembrane proteins in cell death. Biochem Biophys Res Commun 304:487–497 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 467 EP 472 DI 10.1007/s12253-008-9060-x PG 6 ER PT J AU Bely, M Apathy, AF Bely, Miklos Apathy, Agnes TI Recurrent Pancreatic Arteritis and Vasculogenic Relapsing Pancreatitis in Rheumatoid Arthritis – A Retrospective Clinicopathologic and Immunohistochemical Study of 161 Autopsy Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Rheumatoid arthritis; Vasculitis; Pancreatitis ID Rheumatoid arthritis; Vasculitis; Pancreatitis AB The aim of this study was to determine: the prevalence, and histological characteristics of vasculitis in the pancreas, and to follow the formal pathogenesis of multifocal pancreatitis due to arteritis and/or arteriolitis (multifocal vasculogenic pancreatitis). A randomized autopsy population of 161 in-patients with rheumatoid arthritis (RA) was studied. Systemic vasculitis (SV) complicated RA in 36 (22.36%) of 161 cases; tissue samples of pancreas were available for histologic evaluation in 28 patients. Pancreatitis and vasculitis were characterized histologically and immunohistochemically. Vasculogenic, multifocal pancreatitis was not recognized clinically. Vasculitis of the pancreatic arterioles and small arteries (branches of splenic artery, upper and lower gastroduodenal arteries) can lead to local ischaemia and to regressive changes in the pancreas. This vasculogenic process is more or less widespread and multifocal, depending on the number of involved vessels and is followed by reactive inflammation, depending on the stages of the pathological process. Because of the recurrent nature of vasculitis with time these regressive changes accumulate within the pancreas and may contribute to an unexpected circulatory failure and sudden death of the patient. Vasculogenic microinfarcts in the pancreas may be clinically characterized by unexplained recurrent abdominal symptoms and spontaneous remissions which insidiously may lead to metabolic failure resistant to therapy. C1 [Bely, Miklos] St John's Hospital, Department of Pathology, Frankel Leo u. 17-19, H-1027 Budapest, Hungary. [Apathy, Agnes] National Institute for Rheumatology and PhysiotherapyBudapest, Hungary. RP Bely, M (reprint author), St John's Hospital, Department of Pathology, H-1027 Budapest, Hungary. EM dr.bely.miklos@t-online.hu CR de Prado NI, de la Calle CMA, de Prado NJM, Sanchez GF, Medranda MA, 2005, Vascular complications of pancreatitis. Rev Clin Esp 205:326–332 Mendelson RM, Anderson J, Marshall M, Ramsay D, 2005, Vascular complications of pancreatitis. ANZ J Surg 75:1073– 1079 de Araujo TS, Kirsner RS, 2001, Vasculitis: wounds 13:99–110 Tada M, Naruse S, Arai A et al, 2004, An autopsy case of systemic vasculitis associated with hepatitis C virus-related mixed cryoglobulinemia presenting severe peripheral neuropathy. Rinsho Shinkeigaku 44:686–690 Pagnoux C, Mahr A, Cohen P, Guillevin L, 2005, Presentation and outcome of gastrointestinal involvement in systemic necrotizing vasculitides: analysis of 62 patients with polyarteritis nodosa, microscopic polyangiitis, Wegener granulomatosis, Churg–Strauss syndrome, or rheumatoid arthritis-associated vasculitis. Medicine, Baltimore, 84:115–128 Christl SU, Borchard F, Keller R et al, 2004, Pancreatic tail tumor as an unusual first manifestation of Wegener’s disease. Z Gastroenterol 42:513–516 Swol-Ben J, Bruns CJ, Muller-Ladner U et al, 2004, Leukoencephalopathy and chronic pancreatitis as concomitant manifestations of systemic lupus erythematosus related to anticardiolipin antibodies. Rheumatol Int 24:177–181 Iwasa S, Katoh R, 2005, Autopsy case of microscopic polyangiitis with crescentic glomerulonephritis and necrotizing pancreatitis. Pathol Int 55:520–523 Haraguchi K, Gunji K, Ito Y, Yokomori N et al, 2005, Extensive pancreatic necrosis in microscopic polyangiitis. Clin Exp Nephrol 9:326–331 Hidaka T, Suzuki K, Kawakami M et al, 1992, An autopsy case of malignant rheumatoid arthritis, MRA, which was difficult to distinguish from polyarteritis nodosa, PN). Ryumachi 32:318–326 O’Neil KM, Jones DM, Lawson JM, 1992, Wegener’s granulomatosis masquerading as pancreatic carcinoma. Dig Dis Sci 37:702–704 Romhanyi G, 1971, Selective differentiation between amyloid and connective tissue structures based on the collagen specific topo-optical staining reaction with congo red. Virchows Arch 354:209–222 Romhanyi G, 1979, Selektive Darstellung sowie methodologische Moglichkeiten der Analyse ultrastruktureller Unterschiede von Amyloidablagerungen. Zbl Allg Pathol Pathol Anat 123:9–16 Wright JR, Calkins E, Humphrey RL, 1977, Potassium permanganate reaction in amyloidosis. Lab Invest 36:274–281 Bely M, Apathy A, 1999, Systemic secondary, AA, amyloidosis in rheumatoid arthritis. In: Kyle RA, Gertz MA, eds, Amyloid and amyloidosis 1998. USA Parthenon Publishing, New York– London, pp 408–410 Bely M, 2006, Histochemical differential diagnosis and polarization optical analysis of amyloid and amyloidosis. The Scientific World Journal 6: 154–168, in memoriam of Professor G Romhanyi, September 15, 1905 to August 29, 1991; http://www. thescientificworld.com/SCIENTIFICWORLDJOURNAL/toc/ TSWJ_ArticleLanding.asp?ArticleId=1899 http://www.abstracts2- view.com/eular/) Cruickshank B, 1954, The arteritis of rheumatoid arthritis. Ann Rheum Dis 13:136–146 Sinclair RJG, Cruickshank B, 1956, A clinical and pathological study of sixteen cases of rheumatoid arthritis with extensive visceral involvement, ‘Rheumatoid disease’). Q J Med 25:313– 332 Cruickshank B, 1958, Heart lesions in rheumatoid disease. J Pathol Bacteriol 76:223–240 Lebowitz WB, 1963, The heart in rheumatoid arthritis, Rheumatoid disease).A clinical and pathological study of sixty-two cases. Ann Intern Med 58:102–123 Sokoloff L, 1964, Cardiac involvement in rheumatoid arthritis and allied disorders: current concepts. Mod Concepts Cardiovasc Dis 33:847–850 Karten I, 1969, Arteritis, myocardial infarction, and rheumatoid arthritis. JAMA 210:1717–1720 Gardner DL, 1972, Causes of death. In: The pathology of rheumatoid arthritis London Edward Arnold, pp 183–197 Davis RF, Engleman EG, 1974, Incidence of myocardial infarction in patients with rheumatoid arthritis. Arthitis Rheum 17:527–533 Eulderink F, 1976, Doodsoorzak: rheumatoide arthritis. Ned T Geneesk 120:357–363 Albada-Kuipers VGA, Bruijn JA, Westedt M-L, Breedveld FC, Eulderink F, 1986, Coronary arteritis complicating rheumatoid arthritis. Ann Rheum Dis 45:963–965 Boers M, Croonen AM, Dijkmans BA, Breedveld FC, Eulderink F, Cats A, Weening JJ, 1987, Renal finding in rheumatoid arthritis: clinical aspect of 132 necropsies. Ann Rheum Dis 46:658–663 Suzuki A, Ohosone Y, Obana M, Mita S, Matsuoka Y, Irimajiri S, Fukuda J, 1994, Cause of death in 81 autopsied patients with rh reumatoid arthritis. J Rheumatol 21:33–36 Bely M, Apathy A, 2002, Systemic vasculitis in rheumatoid arthritis: the frequency, severity and stages of vasculitis in various organs – a retrospective clinicopathologic study of 161 autopsy patients, Abstract). Ann Rheum Dis 61(Suppl 1):394. http://www. abstracts2view.com/eular/ Bely M, Apathy A, 2002, Histologic characteristics of systemic vasculitis in rheumatoid arthritis: the types of vasculitis and size of blood vessels involved by vasculitis – a retrospective clinicopathologic study of 161 autopsy patients, Abstract). Ann Rheum Dis 61(Suppl 1):394–395. http://www.abstracts2view. com/eular/ Bely M, Apathy A, 2006, Szovodmenyek es tarsult megbetegedesek rheumatoid arthritisben – a 234 elhunyt beteg patologiai es klinikai adatainak retrospektiv elemzese alapjan. Orvosi Hetilap 147:1063–1076 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 473 EP 480 DI 10.1007/s12253-008-9026-z PG 8 ER PT J AU Kullmann, T Barta, I Csiszer, E Antus, B Horvath, I AF Kullmann, Tamas Barta, Imre Csiszer, Eszter Antus, Balazs Horvath, Ildiko TI Differential Cytokine Pattern in the Exhaled Breath of Patients with Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Proteomics; Exhaled breath condensate; Microarray. Biomarker ID Lung cancer; Proteomics; Exhaled breath condensate; Microarray. Biomarker AB Tumour cells may alter the protein pattern of biological samples resulting in specific differences that may aid diagnosis and treatment. In this pilot study we tested the cytokine pattern of exhaled breath condensate of patients with lung cancer. Breath condensates collected from 50 smoking patients with lung cancer and 25 smokers without clinical or radiological sign of a pulmonary tumour but having co-morbidities with similar severity as those with lung cancer were pooled for antibody microarray analysis testing 120 cytokines in parallel. Every cytokine on the array gave a signal in both groups. Nine cytokines including eotaxin, FGFs, IL-10 and MIP-3 were present with more than two-fold difference between the two groups. Large number of cytokines is present in the exhaled breath. Further analysis of specific differences associated with lung cancer may have clinical importance. C1 [Kullmann, Tamas] National Koranyi Institute for TB and Pulmonology, Department of PathophysiologyBudapest, Hungary. [Barta, Imre] National Koranyi Institute for TB and Pulmonology, Department of PathophysiologyBudapest, Hungary. [Csiszer, Eszter] National Koranyi Institute for TB and Pulmonology, Department of PulmonologyBudapest, Hungary. [Antus, Balazs] National Koranyi Institute for TB and Pulmonology, Department of PathophysiologyBudapest, Hungary. [Horvath, Ildiko] Semmelweis University, Institute of Morphology and Physiology, Ulloi street 78/a, 1082 Budapest, Hungary. RP Horvath, I (reprint author), Semmelweis University, Institute of Morphology and Physiology, 1082 Budapest, Hungary. EM hildiko@elet2.sote.hu CR Hultschig C, Kreutzberger J, Seitz H et al, 2006, Recent advances of protein microarrays. Curr Opin Chem Biol 10:4–10 Zhong L, Coe SP, Stromberg AJ et al, 2006, Profiling tumorassociated antibodies for early detection of non-small cell lung cancer. J Thoracic Oncol 1:513–519 Horvath I, Hunt J, Barnes PJ, on behalf of the ERS/ATS TaskForce, 2005, Exhaled breath condensate: methodological recommendations and unresolved questions. Eur Respir J 26:523– 548 Matsunaga K, Yanagisawa S, Ichikawa T et al, 2006, Airway cytokine expression measured by means of protein microarray in exhaled breath condensate: correlation with physiologic properties in asthmatic patients. J Allergy Clin Immunol 118:84–90 Czebe K, Barta I, Antus B et al, 2008, Influence of condensing equipment and temperature on exhaled breath condensate pH, total protein and leukotriene concentrations. Respir Med 102: 720–725, Epub 2008 Jan 31 Ko FW, Lau CY, Leung TF et al, 2006, Exhaled breath condensate levels of eotaxin and macrophage-derived chemokine in stable adult asthma patients. Clin Exp Allergy 36:44–51 Leung TF, Wong GW, Ko FWet al, 2004, Increased macrophagederived chemokine in exhaled breath condensate and plasma from children with asthma. Clin Exp Allergy 34:786–791 Sack U, Scheibe R, Wotze lM et al, 2006, Multiplex analysis of cytokines in exhaled breath condensate. Cytometry A 69:169–172 Dimberg J, Hugander A, Wagsater D, 2006, Protein expression of the chemokine, CCL28, in human colorectal cancer. Int J Oncol. 28:315–319 Kwabi-Addo B, Ozen M, Ittmann M, 2004, The role of fibroblast growth factors and their receptors in prostate cancer. Endocr Relat Cancer 11:709–724 Szilasi M, Dolinay T, Nemes Z et al, 2006, Pathology of chronic obstructive pulmonary disease. Pathol Oncol Res 12:52–60 Phillips M, Cataneo RN, Cummin AR et al, 2003, Detection of lung cancer with volatile markers in the breath. Chest 123:2115– 2123 Machado RF, Laskowski D, Deffenderfer O et al, 2005, Detection of lung cancer by sensor array analyses of exhaled breath. Am J Respir Crit Care Med 171:1286–1291 Elek G, Lapis K, 2006, A path or a new road in laboratory diagnostics? Biological mass spectrometry: facts and perspectives. Pathol Oncol Res 12:178–183 Laszlo K, Jozsef T, 2005, Genomics of lung cancer may change diagnosis, prognosis and therapy. Pathol Oncol Res 11:5–10 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 481 EP 483 DI 10.1007/s12253-008-9046-8 PG 3 ER PT J AU Zaka, Z Fodor, J Udvarhelyi, N Orosz, Zs Kasler, M AF Zaka, Zoltan Fodor, Janos Udvarhelyi, Nora Orosz, Zsolt Kasler, Miklos TI Subcutaneous Calcification as a Delayed Complication of Radiotherapy: A Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Ichthyosis; Hodgkin’s disease; Radiotherapy; Subcutaneous calcification ID Ichthyosis; Hodgkin’s disease; Radiotherapy; Subcutaneous calcification AB Subcutaneous calcification following radiotherapy is a very rare late complication. Here we report a case of radiotherapy for Hodgkin’s disease in a patient with ichthyosis. Our review of the literature revealed that subcutaneous calcifications occurred in previously normal skin following irradiation. In our case the calcification developed in abnormal skin after telecobalt radiotherapy for mediastinal Hodgkin’s disease. Ichthyosis is a systemic cutaneous disease and may increase the risk of late radiation complications. C1 [Zaka, Zoltan] National Institute of Oncology, Center of Radiotherapy, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Fodor, Janos] National Institute of Oncology, Center of Radiotherapy, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Udvarhelyi, Nora] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Orosz, Zsolt] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Kasler, Miklos] National Institute of Oncology, Department of SurgeryBudapest, Hungary. RP Zaka, Z (reprint author), National Institute of Oncology, Center of Radiotherapy, 1122 Budapest, Hungary. EM zaka@oncol.hu CR Chon BH, Loeffler JS, 2002, The effect of nonmalignant systemic disease on tolerance to radiation therapy. Oncologist 2:136–143 Cowie F, Jones R, 1999, Subcutaneous calcification as a late effect of orthovoltage chest wall irradiation. Clin Oncol 11:196–197 Steinert M, Gottlober P, Gall H, Peter RU, 2001, Subcutaneous calcification after radiotherapy. Hautarzt 52:518–521 Amin R, Hamilton-Wood C, Silver D, 2002, Subcutaneous calcification following chest wall and breast irradiation: a late complication. Br J Radiol 75:279–282 Carl UM, Hartman KA, 2002, Heterotopic calcification as a late radiation effect: report of 15 cases. Br J Radiol 75:460–463 Lewis VJ, Holt PJ, 2004, Sucutaneous calcification following high-dose radiotherapy. Br J Dermatol 150:1049–1050 Morris MM, Powell SN, 1997, Irradiation in the setting of collagen vascular disease: acute and late complications. J Clin Oncol 15:2728–2735 Gatti RA, 2001, The inherited basis of human radiosensitivity. Acta Oncol 40:702–711 McCuaig C, Marcoux D, Rasmussen JE, Werner MM, Gentner NE, 1993, Trichothiodystrophy associated with photosensitivity, gonadal failure, and striking osteosclerosis. J Am Acad Dermatol 28:820–826 Sator PG, Schmidt JB, Honigsmann H, 2003, Comparison of epidermal hydration and skin surface lipids in healthy individuals and in patients with atopic dermatitis. J Am Acad Dermatol 48:352–358 DiGiovanna JJ, Robinson-Bostom L, 2003, Ichthyosis: etiology, diagnosis, and treatment. Am J Clin Dermatol 4:81–95 Fleckman P, Brumbaugh S, 2002, Absence of the granular layer and keratohyalin define a morphologically distinct subset of individuals with ichthyosis vulgaris. Exp Dermatol 11:327– 336 Lykkesfeldt G, Bennett P, Lykkesfeldt AE, Micic S, Rorth M, Skakkebaek NE, Svenstrup B, 1991, Testis cancer. Ichthyosis constitutes a significant risk factor. Cancer 67:730–734 Elbaum DJ, Kurz G, MacDuff M, 1995, Increased incidence of cutaneous carcinomas in patients with congenital ichthyosis. J Am Acad Dermatol 33:884–886 Kato N, Yasukawa K, Kimura K, Yoshida K, 2000, Anaplastic large-cell lymphoma associated with acquired ichthyosis. J Am Acad Dermatol 42:914–920 Kim KH, Kim JS, Piao YJ, Kim YC, Shur KB, Lee JH, Park JK, 2002, Keratitis, ichthyosis and deafness syndrome with development of multiple hair follicle tumours. Br J Dermatol 147:139–143 Krasagakis K, Ioannidou DJ, Stephanidou M, Manios A, Panayiotides JG, Tosca AD, 2003, Early development of multiple epithelial neoplasms in Netherton syndrome. Dermatology 207:182–184 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 485 EP 488 DI 10.1007/s12253-008-9062-8 PG 4 ER PT J AU Kojima, M Kitamoto, Y Shimizu, K Matsuda, H Masawa, N AF Kojima, Masaru Kitamoto, Yoshizumi Shimizu, Kazuhiko Matsuda, Hazuki Masawa, Nobuhide TI Tonsillar Lesions of Infectious Mononucleosis Resembling MALT Type Lymphoma. A Report of Two Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Infectious mononucleosis; Epstein–Barr virus; Tonsil; MALT type lymphoma; Immunohistochemistry ID Infectious mononucleosis; Epstein–Barr virus; Tonsil; MALT type lymphoma; Immunohistochemistry AB Infectious mononucleosis (IM) is an acute lymphoproliferative disorder that typically occurs in young patients and is usually caused by Epstein–Barr virus. We report here, two cases of tonsillar lesion of IM resembling marginal zone B-cell lymphoma mucosa-associated lymphoid tissue (MALT) type. The patients consisted of an 18-year-old Japanese woman and a 36-year-old Japanese man. Both patients presented with tonsillar mass. Histologically, in one case, the tonsil showed diffuse proliferation of medium-sized lymphocytes. The medium-sized lymphocytes had round or slightly indented nuclei with a small solitary nucleoli and abundant clear cytoplasm and somewhat resembled monocytoid B-cells. In the remaining one case, the lymphoid follicles had hyperplastic germinal centers with ill-defined borders surrounded by a sheet-like proliferation of polymorphous infiltration showing a marginal zone distribution pattern. On high-power field, the interfollicular area was diffusely infiltrated by a polymorphous infiltrate of medium-sized lymphocytes with angulated nuclei somewhat resembling centrocyte-like cells, immunoblasts, plasma cells, plasmacytoid cells and histiocytes with or without epithelioid cell feature. However, there were no CD43+ B-cells in either lesion. Moreover, the polytypic nature of the B-cells was demonstrated by immunohistochemistry or polymerase chain reaction. Although MALT type lymphoma rarely affected young adults, notably, a number of cases have been reported in the tonsil. The present two cases indicated that acute IM should be added to the differential diagnosis for MALT type lymphoma in young adults. C1 [Kojima, Masaru] Gunma Cancer Center Hospital, Department of Pathology and Clinical Laboratories, 617-1, Takabayashinishi-cho, 373-8550 Ohta, Japan. [Kitamoto, Yoshizumi] Gunma Cancer Center Hospital, Department of RadiologyOhta, Japan. [Shimizu, Kazuhiko] Ashikaga Red Cross Hospital, Department of Pathology and Clinical LaboratoriesAshikaga, Japan. [Matsuda, Hazuki] Dokkyo Medical University School of Medicine, Department of Diagnostic and Anatomic PathologyMibu, Japan. [Masawa, Nobuhide] Dokkyo Medical University School of Medicine, Department of Diagnostic and Anatomic PathologyMibu, Japan. RP Kojima, M (reprint author), Gunma Cancer Center Hospital, Department of Pathology and Clinical Laboratories, 373-8550 Ohta, Japan. EM mkojima@gunma-cc.jp CR Symmers WStC, 1992, Infectious mononucleosis. In: Henry K, Symmers WStC, eds, Thymus, lymph node, spleen and lymphatics. Systemic pathology, 3rd edn, vol7. Churchill Livingstone, Edinburgh, pp 432–437 Henle W, Henle GE, Horwiltz CA, 1974, Epstein–Barr virus specific diagnostic tests in infectious mononucleosis. Human Pathol 5:551–565 Childs CC, Parham DM, Berard CW, 1987, Infectious mononucleosis. The spectrum of morphologic changes simulating lymphoma in lymph node and tonsils. Am J Surg Pathol 11:122–132 Strickler JG, Fedeli F, Horwitz CA et al, 1993, Infectious mononucleosis in lymphoid tissue-histopathology, in situ hybridization and differential diagnosis. Arch Pathol Lab Med 117: 269–278 Wan JH, Trainor KJ, Brisco MJ et al, 1990, Monoclonality in B cell lymphoma detected in paraffin wax embedded sections using the polymerase chain reaction. J Clin Pathol 43:888–890 Isaacson PG, Spencer J, 1987, Malignant lymphoma of mucosaassociated lymphoid tissue. Histopathology 11:445–462 Feller AC, Diebold J, 2003, Histopathology of nodal and extranodal non-Hodgkin's lymphomas. Springer, Berlin Wang T, Lasota J, Hanau CA et al, 1995, Bcl-2 oncoprotein is widespread in lymphoid tissue and lymphoma but its differential expression in benign versus malignant follicles and monocytoid B-cell proliferation is of diagnostic value. APMIS 103:655–662 Patsouris E, Noel H, Lennert K, 1990, Lymphoplasmacytic/ lymphoplasmacytoid immunocytoma with a high content of epithelioid cells. Histologic and immunohistochemical findings. Am J Surg Pathol 14:660–670 Kojima M, Nakamura S, Shimizu K et al, 2001, Marginal zone B cell lymphomas of Waldeyer’s ring. A report of two tonsillectomy cases resembling histomorphologic features of inflammatory lesions. Pathol Res Pract 197:781–784 Taddesse-Heath L, Pittaluga S, Sorbara L et al, 2003, Nodal marginal zone B-cell lymphoma in young adults. Am J Surg Pathol 27:522–531 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 489 EP 492 DI 10.1007/s12253-008-9033-0 PG 4 ER PT J AU Sagnak, L Topaloglu, H Gucuk, O Unsal, H Ersoy, H AF Sagnak, Levent Topaloglu, Hikmet Gucuk, Osman Unsal, Han Ersoy, Hamit TI Skip Metastase on the Left Neck Lymph Nodes of the Prostatic Adenocarcinoma with Neuroendocrine Differentiation and Accompanying Thyroid Micropapillary Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate neuroendocrine adenocarcinoma; Skip metastase; Thyroid papillary carcinoma ID Prostate neuroendocrine adenocarcinoma; Skip metastase; Thyroid papillary carcinoma AB We discuss here the thyroid micropapillary carcinoma that was detected incidentally when investigating the primary focus of the left neck multiple lymph node metastases occurring 8 months later in a patient of ours, whose pathological examination of radical prostatectomy and bilateral inguinal lymph node dissection was reported to be pT3N0 and whole body scanning for metastases, was negative. C1 [Sagnak, Levent] Ministry of Health, Yildirim Beyazit Training Hospital, 3rd Urology Clinic, Angoraevleri, Altinbel Villalari, No: 38/A, 06800 Ankara, Turkey. [Topaloglu, Hikmet] Ministry of Health, Yildirim Beyazit Training Hospital, 3rd Urology Clinic, Angoraevleri, Altinbel Villalari, No: 38/A, 06800 Ankara, Turkey. [Gucuk, Osman] Ministry of Health, Yildirim Beyazit Training Hospital, 3rd Urology Clinic, Angoraevleri, Altinbel Villalari, No: 38/A, 06800 Ankara, Turkey. [Unsal, Han] Diskapi Yildirim Beyazit Education and Research Hospital, Department of PathologyAnkara, Turkey. [Ersoy, Hamit] Ministry of Health, Yildirim Beyazit Training Hospital, 3rd Urology Clinic, Angoraevleri, Altinbel Villalari, No: 38/A, 06800 Ankara, Turkey. RP Sagnak, L (reprint author), Ministry of Health, Yildirim Beyazit Training Hospital, 3rd Urology Clinic, 06800 Ankara, Turkey. EM leventsagnak@gmail.com CR Greenlee RT, Murray T, Bolden S, Wingo PA, 2000, Cancer statistics,2000. CA Cancer J Clin 50:7–33 Epstein JI, 2002, Pathology of prostatic neoplasia. In: Walsh PC, Retik AB, Vaughan ED, Wein AJ, eds, Campbell’s Urology, 8th edn, vol.4, chp 86. WB Saunders, Philadelphia, pp 3025–3037 Saitoh H, Yoshida K, Uchijima Y, Kobayashi N, Suwata J, Kamata S, 1990, Two different lymph node metastatic patterns of a prostatic cancer. Cancer 65(8):1843–1846 Wang HJ, Chiang PH, Peng JP, Yu TJ, 2004, Presentation of prostate carcinoma with cervical lymphadenopathy: report of three cases Med J 27(11):840–844 Copeland B, Clark JM, Sura A, Kilpatrick SE, Shockley W, Meredith S, 2001, Prostate carcinoma metastatic to the cervical lymph nodes: report of two cases and review of the literature. Am J Otolaryngol 22(6):420–423 Ozgur A, Ilker Y, Turkeri LN, 2003, Cervical lymph node enlargement on the right side as the initial manifestation of metastatic prostate cancer. Arch Esp Urol 56(7):859–861 Carleton J, van der Riet P, Dahm P, 2005, Metastatic prostate cancer presenting as an asymptomatic neck mass. Prostate Cancer Prostatic Dis 8(3):293–295 di Sant’Agnese PA, 1992, Neuroendocrine differantiation in human prostatic carcinoma. Human Pathol 23(3):287–296 di Sant’Agnese PA, 2000, Divergent neuroendocrine differentiation in prostatic carcinoma. Semin Diagn Pathol 17(2):149–161 Fernandes RC, Matsushita MM, Manuad T, Nascimento Saldiva PH, 2001, Prostate carcinoma with neuroendocrine differentiation: case report and literature review. Rev Hosp Clin Fac Med Sao Paulo 56(5):153–158 Bostwick DG, 1997, Neoplasms of the prostate. In: Bostwick DG, ed, Urologic Surgical Pathology. Mosby-Year Book, St.Louis di Sant’Agnese PA, Cockett AT, 1996, Neuroendocrine differentiation in prostatic malignancy. Cancer 78(2):357–361 Ishida E, Nakamura M, Shimada K, Matsuyoshi S, Tada K, Okajima E, Fujimoto k, Konishi N, 2004, Autopsy case of prostate cancer with multiple endocrine neoplasia 2A. Pathol Int 54(12):918–923 Goulet-Salmon B, Berthe E, Franc S, Chanel S, Galateau-Salle F, Kottler M, Mahoudeau J, Reznik Y, 2004, Prostatic neuroendocrine tumor in multiple endocrine neoplasia type 2b. Endocrinol Invest 27, 6):570–573 Bayram F, Soyuer I, Atmaca H, Demirci D, Gokce C, Canoz O, Unluhizarci K, Kelestimur F, 2004, Prostatic adenocarcinoma metastasis in the thyroid gland. Endocrine Journal 51(4):445–448 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 493 EP 495 DI 10.1007/s12253-008-9029-9 PG 3 ER PT J AU Pina-Oviedo, S Del Valle, L Padilla-Longoria, R Mendoza-Ramon, H Ortiz-Hidalgo, C AF Pina-Oviedo, Sergio Del Valle, Luis Padilla-Longoria, Rafael Mendoza-Ramon, Hilda Ortiz-Hidalgo, Carlos TI Primary Adamantinoma of the Rib. Unusual Presentation for a Bone Neoplasm of Uncertain Origin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adamantinoma; Bone tumor; Bone epithelial neoplasm; Immunohistochemistry ID Adamantinoma; Bone tumor; Bone epithelial neoplasm; Immunohistochemistry AB Adamantinomas are rare, low-grade malignant intra-osseous tumors composed of epithelial and mesenchymal elements, which show a marked predilection for the tibia and fibula of young adult male patients. Although cases of adamantinoma located to the axial skeleton have been reported either as recurrent or metastatic disease, only two cases of primary adamantinoma located to the thoracic wall have been previously described. In this study we present the clinical, radiological and histopathological features of a 24-year-old male with a slow growing, solid-cystic, painful mass, located to the right 11th rib, which was morphological and immunohistochemically diagnosed as a primary classic adamantinoma. Radiological studies showed a multiloculated lesion with a solid component. The patient underwent a whole surgical resection of the lesion. Histologically, multiple foci of epithelial cells with basaloid and squamous components were found intermixed within a fibrous stromal tissue. Immunohistochemical analysis demonstrated expression of cytokeratins, EMA, vimentin and other epithelial markers. Primary affection of the rib is an unusual feature of classic adamantinomas. C1 [Pina-Oviedo, Sergio] Universidad Panamericana, School of Medicine, Laboratory of Tissue and Cell BiologyMexico City, Mexico. [Del Valle, Luis] Temple University, School of Medicine, Department of NeurosciencePhiladelphia, PA, USA. [Padilla-Longoria, Rafael] The American British Cowdray Medical Center, Department of Surgical OncologyMexico City, Mexico. [Mendoza-Ramon, Hilda] The American British Cowdray Medical Center, Department of Pathology, Calle Sur 132, No. 116. Colonia Las Americas, C.P. 01120 Mexico City, Mexico. [Ortiz-Hidalgo, Carlos] Universidad Panamericana, School of Medicine, Laboratory of Tissue and Cell BiologyMexico City, Mexico. RP Ortiz-Hidalgo, C (reprint author), Universidad Panamericana, School of Medicine, Laboratory of Tissue and Cell Biology, Mexico City, Mexico. EM cortiz@abchospital.com CR Hogendoorn PCW, Hashimoto H, 2002, Adamantinoma. In: Fletcher CDM, Unni KK, Mertens F. eds. World Health Organization classification of tumours. Pathology and genetics of tumours of soft tissue and bone. IARC Press, Lyon, France pp. 332–334 Kahn LB, 2003, Adamantinoma, osteofibrous dysplasia and differentiated adamantinoma. Skelet Radiol 32:45–58 Maier C, 1900, Ein primares myelogenes platten epithelkarcinom der ulna. Beitr Klin Chir 26:553–556 Fischer B, 1913, Uber ein primares adamantinoma der tibia. Frankfurt Z Path 12:422–441 Bohndorf K, Nidecker A, Mathias K et al, 1992, The radiological findings in adamantinoma of the long tubular bones. Rofo 157:239–244 Putnam A, Yandow S, Coffin CM, 2003, Classic adamantinoma with osteofibrous dysplasia-like foci and secondary aneurismal bone cyst. Pediatr Dev Pathol 6:173–178 Weiss SW, Dorfman HD, 1977, Adamantinoma of long bone. An analysis of nine new cases with emphasis on metastasizing lesions and fibrous dysplasia-like changes. Hum Pathol 8:141–153 Plump D, Haponik EF, Katz RS et al, 1986, Primary adamantinoma of rib: thoracic manifestations of a rare bone tumor. South Med J 79:352–355 Beppu H, Yamaguchi H, Yoshimura N et al, 1994, Adamantinoma of the rib metastasizing to the liver. Int Med 33:441–445 Czerniak B, Rojas-Corona RR, Dorfman HD, 1989, Morphologic diversity of long bone adamantinoma. The concept of differentiated, regressing, adamantinoma and its relationship to ostefibrous dysplasia. Cancer 64:2319–2334 Maki M, Athanasou N, 2004, Osteofibrous dysplasia and adamantinoma: correlation of proto-oncogene product and matrix protein expression. Hum Pathol 35:69–74 Bridge JA, Dembinski A, DeBoer J et al, 1994, Clonal chromosomal abnormalities in osteofibrous dysplasia. Implications for histopathogenesis and its relationship with adamantinoma. Cancer 73:1746–1752 Kanamori M, Antonescu CR, Scott M et al, 2001, Extra copies of chromosomes 7, 8, 12, 19 and 21 are recurrent in adamantinomas. J Mol Diagn 3:16–21 Hazelbag HM, Fleuren JG, van den Broek LJ et al, 1993, Adamantinoma of the long bones: keratin subclass immunoreactivity pattern with reference to its histogenesis. Am J Surg Pathol 17:1225– 1233 Altini M, Coleman H, Doglioni C et al, 2000, Calretinin expression in ameloblastomas. Histopathology 37:27–32 Bovee JV, van der Broek LJ, de Boer WI et al, 1998, Expression of growth factors and their receptors in adamantinoma of long bones and the implication for its histogenesis. J Pathol 184: 24–30 Qureshi AA, Shott S, Mallin BA et al, 2000, Current trends in the management of adamantinoma of long bones. An international study. J Bone Joint Surg Am 82-A:1122–1131 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2008 VL 14 IS 4 BP 497 EP 502 DI 10.1007/s12253-008-9009-0 PG 6 ER PT J AU Sereno, M De castro, J Belda-Iniesta, C Garcia-Cabezas, AM Cejas, P Casado, E Barriuso, J Feliu, J Larrauri, J AF Sereno, Maria De castro, Javier Belda-Iniesta, Cristobal Garcia-Cabezas, Angel Miguel Cejas, Paloma Casado, Enrique Barriuso, Jorge Feliu, Jaime Larrauri, Javier TI EPO-R Expression Patterns in Resected Gastric Adenocarcinoma Followed by Adjuvant Chemoradiation Treatment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Epo-R; adjuvant chemoradiation ID Gastric cancer; Epo-R; adjuvant chemoradiation AB The primary aim was to determine whether Epo-R immunohistochemical expression is related to disease free survival (DFS) in specimens of GC from patients who underwent adjuvant chemoradiation. Specimens of gastric adenocarcinomas obtained from 44 patients who had undergone curative gastrectomy and adjuvant treatment were investigated immunohistochemically expression of Epo-R. Three patterns for Epo-R staining were defined: Pattern A (secretory cells-like staining), Pattern B (parietallike staining) and Pattern C (chief-like staining). Median DFS was 38 months (CI 95%: 33–43) and 15 months (IC 95%: 3–27) in the pattern B and C, respectively, but it was not reached in the pattern A (p=0.06). Our findings suggest that there may be a relationship between Epo-R expression and DFS in the patients with GC resected. C1 [Sereno, Maria] Infanta Sofia Hospital, Medical Oncology Division, San Sebastian de los ReyesMadrid, Spain. [De castro, Javier] University Hospital La Paz, Medical Oncology DivisionMadrid, Spain. [Belda-Iniesta, Cristobal] University Hospital La Paz, Medical Oncology DivisionMadrid, Spain. [Garcia-Cabezas, Angel Miguel] University Hospital La Paz, Medical Oncology DivisionMadrid, Spain. [Cejas, Paloma] University Hospital La Paz, Medical Oncology DivisionMadrid, Spain. [Casado, Enrique] Infanta Sofia Hospital, Medical Oncology Division, San Sebastian de los ReyesMadrid, Spain. [Barriuso, Jorge] University Hospital La Paz, Medical Oncology DivisionMadrid, Spain. [Feliu, Jaime] University Hospital La Paz, Medical Oncology DivisionMadrid, Spain. [Larrauri, Javier] University Hospital La Paz, Medical Oncology DivisionMadrid, Spain. RP Sereno, M (reprint author), Infanta Sofia Hospital, Medical Oncology Division, Madrid, Spain. EM mariasereno@yahoo.es CR Tilbrook PA, Klinken SP, 1999, Erythropoeitin and erythropoeitin receptor. Growth Factors 17:25–35 Ebert BL, Bunn HF, 1999, Regulation of the erythropoeitin gene. Blood 94:1864–77 Wojchowski DM, Gregory RC, Miller CP, Pandit AK, Pircher TJ, 1999, Signal transduction in the erythropoeitin receptor system. Exp Cell Res 253:143–156 Masuda S, Okano M, Yamagishi K, Nagao M, Ueda M, Sasaki R, 1994, A novel site of erythropoeitin production. Oxygen-dependent production in cultured rat astrocytes. J Biol Chem 269:19488–19493 Acs G, Acs P, Beckwith SM, 2001, Erythropoeitin and erythropoetin receptor in breast carcinoma. Cancer Res 61:3561–3565 Pallard C, Gouileux F, Charon M, Groner B, Gisselbrecht S, Dusanter- Fourt I, 1995, Interleukin-3, eryhropoeitin and prolactin activate a STAT5-like factor in lymphoid cells. J Biol Chem 270:15942–15945 Carroll MP, Spivak JL, McMahon M, Weich N, Rapp UR, May WS, 1991, Erythropoeitin induces Raf-1 activation and Raf-1 is required for erythropoetin-mediated proliferation. J Biol Chem 266:14964–14969 Acs G, Zhang PJ, McGarth CM, 2003, Hypoxia-inducible erythropoietin signalling in squamous dysplasia and squamous cell carcinoma of the uterine cervix and its potential role in cervical carcinogenesis and tumour progression. Am J Pathol 162:1789–1806 Mohyeldin A, Lu H, Dalgard C, Stephen Y, Noam C, Acs G, Verma A, 2005, Erythropoetin signaling promotes invasiveness of human head and neck squamous cell carcinoma. Neoplasia 7:537–543 Henke M, Laszig R, Rube C, Schafer U, Haase KD, Schilcher B, Mose S, Beer KT, Burger U, Dougerty C, 2003, Erythropoetin to treat head and neck cancer patients with anemia undergoing radiotherapy: randomized, double blind, placebo controlled trial. Lancet 362:1255–1260 MacDonald J, Smalley S, Benedetti J, Hundahl S, Estes N, Stemmermann G, Haller D, Ajani J, Gunderson L, Jessup M, Materson J, 2001, Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Eng J Med 345:725–730 Lauren P, 1965, The two histological main types of gastric carcinoma. Diffuse and so-called intestinal type carcinoma. Acta Pathol 64:31–40 Carneiro F, Seixas M, Sobrinho-Simoes M, 1987, New elements for an update classification of the carcinoma of the stomach. Pathol Res Pract 182:308–325 Japanese Research Society of Gastric Cancer, 1995, Japanese classification of gastric cancer. Tokio: Kanehara & company 8:199–201 Kim MJ, Bogic L, Cheung CY, Brace RA, 2001, Placental expression of erithropoetin mRNA protein and receptor in sheep. Placenta 22:484–489 Siren AL, Knerlich F, Poser W, Gleiter CH, Bruck W, Ehrenreich H, 2001, Erythropoetin receptor in human ischemic/hypoxic brain. Acta Neuropathol 101:271–276 Hanby AM, 2005, The pathology of breast cancer and the role of the histopathology laboratory. Clin Oncol, R Coll Radiol, 17:234– 239 Barrett C, Magee H, O'toole D, Daly S, Jeffers M, 2006, Amplification of the HER-2 gene in breast cancers testing 2+ weak positive by HercepTestTM immunohistochemistry: false positive or false negative IHC? J Clin Pathol 250:15–19 Nakamura E, Sugihara H, Bamba M, Hattori T, 2005, Dynamic alteration of the E-cadherin/catenin complex during cell differentiation and invasion of undifferentiated-type gastric carcinomas. J Pathol 349:58–61 Ribatti D, Marzullo A, Nico E, Crivellato E, Ria R, 2003, Erythropoeitin as an angiogenic factor in gastric carcinoma. Histopathology 42:246–50 Okada A, Kinoshita Y, Maekawa T, 1996, Erytropoeitin stimulates proliferation of rat cultured gastric mucosal cells. Digestion 57:328–332 Shao Y, Yang SB, Wang MW, Wu BY, You WD, Li H, 2004, Gene expression profile of human adenocarcinoma by cDNA microarray and clustering. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 21:110−115 Lugli A, Spichtin H, Maurer R, Mirlacher M, Kiefer J, Huusko P, Azorsa D, Terracciano L, Sauter G, Kallioniemi OP, Mousses S, Tornillo L, 2005, Erb-B2 expression across 138 human tumor types in a tissue microarray: high levels of expression in gastrointestinal cancers. Clin Cancer Res 11:6450–6458 Gomes LI, Esteves GH, Carvalho AF, Cristo EB, Hirata R Jr, Martins WK, Marques SM, Camargo LP, Brentani H, Pelosof A, Zitron C, Sallum RA, Montagnini A, Soares FA, Neves EJ, Reis LF, 2005, Expression profile of malignant and nonmalignant lesions of esophagus and stomach: differential activity of functional modules related to inflammation and lipid metabolism. Cancer Res 65:7127–7136 Chen J, Rocken C, Klein-Hitpass L, Gotze T, Leodolter A, Malfertheiner P, Ebert MP, 2004, Microarray analysis of gene expression in metastatic gastric cancer cells after incubation with the methylation inhibitor 5-aza-2'-deoxycytidine. Clin Exp Metastasis 21:389–397 Leung SY, Yuen ST, Chu KM, Mathy JA, Li R, Chan AS, Law S, Wong J, Chen X, So S, 2004, Expression profiling identifies chemokine, C-C motif, ligand 18 as an independent prognostic indicator in gastric cancer. Gastroenterology 127:457–469 Tay ST, Leong SH, Yu K, Aggarwal A, Tan SY, Lee CH, Wong K, Visvanathan J, Lim D, Wong WK, Soo KC, Kon OL, Tan P, 2003, A combined comparative genomic hybridization and expression microarray analysis of gastric cancer reveals novel molecular subtypes. Cancer Res 63:3309–3316 Chen X, Leung SY, Yuen ST, Chu KM, Ji J, Li R, Chan AS, Law S, Troyanskaya OG, Wong J, So S, Botstein D, Brown PO, 2003, Variation in gene expression patterns in human gastric cancers. Mol Biol Cell 14:3208–3215 Takeshita A, Shinjo K, Naito K, Ohnishi K, Higuchi M, Ohno R, 2002, Erythropoietin receptor in myelodysplastic syndrome and leukemia. Leuk Lymphoma 43:261–264 Selzer E, Wacheck V, Kodyium R, Schlagbauer-Wadi H, Schlegel W, Pehamberger H, Jansen B, 2000, Erythropoeitin receptor expression in human melanoma cells. Melanoma Res 10:421–426 Belda Iniesta C, De Castro Carpeno J, Casado Saenz E, Feliu Batlle J, Bernabeu F, Alves J, Cejas P, Sereno M, Perona R, Gonzalez Baron M, 2006, Erythropoietin receptor expression in bladder cancer. ASCO 2006 Annual Meeting Proceedings, Post- Meeting Edition, 24:4584 Eccles TG, Patel A, Verma A, Nicholson D, Lukes Y, Tuttle RM, Francis GL, 2003, Erythropoietin and the erythropoietin receptor are expressed by papillary thyroid carcinoma from children and adolescents. Expression of erythropoietin receptor might be a favorable prognostic indicator. Ann Clin Lab Sci 33:411–422 Takeshita A, Shinjo K, Higuchi M, Miyawaki S, Takemoto Y, Kishimoto Y, Saito K, Takuchi H, Kuriyama K, Kimura Y, Asou N, Takahashi M, Hotta T, Kanamaru A, Ueda R, Ohno R, 2003, Quantitative expression of erythropoietin receptor, EPO-R, on acute leukaemia cells: relationships between the amount of EPO-R and CD phenotypes, in vitro proliferative response, the amount of other cytokine receptors and clinical prognosis. Japan Adult Leukaemia Study Group. Br J Haematol 108:55–63 Acs G, Chen M, Xu X, Acs P, Verma A, Koch CJ, 2004, Autocrine erythropoietin signaling inhibits hypoxia-induced apoptosis in human breast carcinoma cells. Cancer Lett 214:243–251 Hirst DG, 1991, What is the importance, ???, of anemia in radiotherapy? The value of animal studies. Radiother Oncol 20:29–33 Harrisson LB, Chadha M, Hill RJ, Hu K, Sasha D, 2002, Impact of hypoxia and anemia on radiation therapy outcomes. Oncologist 7:492–508 Varlotto J, Stevenson MA, 2005, Anemia, tumor hypoxemia, and the cancer patient. Int J Radiat Oncol Biol Phys 63:25–36 Winter SC, Shah KA, Campo L, Turley H, Leek R, Corbridge RJ, Cox GJ, Harris A, 2005, Relation of erythropoietin and erythropoietin receptor expression to hypoxia and anemia in head and neck squamous cell carcinoma. Clin Cancer Res 11:7614– 7620 Hardee ME, Arcasoy MO, Blackwell KL, Kirkpatrick JP, Dewhirst MW, 2006, Erythropoietin biology in cancer. Clin Cancer Res 12:332–9 Yasuda Y, Masuda S, Chikuma M, Inoue K, Nagao M, Sasaki R, 1998, Estrogen-dependent production of erytrhopoetin in uterus and its implication in uterine angiogenesis. J Biol Chem 273:25381–25387 Elliott S, Busse L, Bass MB, Lu H, Sarosi I, Sinclair AM, Spahr C, Um M, Van G, Begley CG, 2006, Anti-Epo receptor antibodies do not predict Epo receptor expression. Blood 107:1892–1895 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 1 EP 10 DI 10.1007/s12253-008-9118-9 PG 10 ER PT J AU Barbanis, S Ioannou, M Kouvaras, E Karasavvidou, F Nakou, M Papamichali, R Koukoulis, G AF Barbanis, Sotirios Ioannou, Maria Kouvaras, Evangelos Karasavvidou, Foteini Nakou, Marianna Papamichali, Roidoula Koukoulis, George TI INCENP (Inner Centromere Protein) is Overexpressed in High Grade Non-Hodgkin B-cell Lymphomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Inner centromere protein (INCENP); MIB-1; Lymphoma; Aneuploidy; Immunohistochemistry ID Inner centromere protein (INCENP); MIB-1; Lymphoma; Aneuploidy; Immunohistochemistry AB Inner centromere protein (INCENP) is a member of the Chromosomal Passenger Complex (CPC), which is a four member protein complex essential for proper completion of mitosis and cell division (cytokinesis). Inappropriate chromosomal segregation and cytokinesis due to deregulated expression of chromosome passenger proteins may lead to aneuploidy and cancer including lymphomas. According to our knowledge this is the first study investigating immunohistochemical expression of INCENP in lymphoma cases and cancer tissues in general. Our purpose was to characterize the expression of INCENP in cases of non-Hodgkin B-cell lymphomas, to compare the immunoreactivity between low and high grades and to evaluate the correlation between INCENP and MIB-1 labeling indices. We examined INCENP and MIB-1 immunoreactivity in paraffin sections of 55 samples of non-Hodgkin B-cell lymphomas, obtained from 55 patients, 31 men and 24 women. Thirty were of high grade and 25 were of low grade. Our results showed significantly higher nuclear immunohistochemical expression of INCENP in high grade B-cell lymphomas versus low grade ones. Also INCENP expression was significantly correlated with MIB-1 labeling index. Taken together our results point to a possible association between increased INCENP immunostaining and B-cell lymphoma aggressiveness and also stress the need for further investigating the expression of INCENP and other mitotic regulatory proteins in lymphomas and other malignant neoplasms. C1 [Barbanis, Sotirios] University of Thessaly, Medical School, Department of Pathology, 411 10 Larissa, Thessaly, Greece. [Ioannou, Maria] University of Thessaly, Medical School, Department of Pathology, 411 10 Larissa, Thessaly, Greece. [Kouvaras, Evangelos] University of Thessaly, Medical School, Department of Pathology, 411 10 Larissa, Thessaly, Greece. [Karasavvidou, Foteini] University of Thessaly, Medical School, Department of Pathology, 411 10 Larissa, Thessaly, Greece. [Nakou, Marianna] University of Thessaly, Medical School, Department of Pathology, 411 10 Larissa, Thessaly, Greece. [Papamichali, Roidoula] University of Thessaly, Medical School, Department of Pathology, 411 10 Larissa, Thessaly, Greece. [Koukoulis, George] University of Thessaly, Medical School, Department of Pathology, 411 10 Larissa, Thessaly, Greece. RP Barbanis, S (reprint author), University of Thessaly, Medical School, Department of Pathology, 411 10 Larissa, Greece. EM sbarbanis@yahoo.gr CR Ruchaud S, Carmena M, Earnshaw WC, 2007, Chromosomal passengers: conducting cell division. Nat Rev Mol Cell Biol 8:798–812 Vader G, Medema RH, Lens SM, 2006, The chromosomal passenger complex: guiding Aurora-B through mitosis. J Cell Biol 173:833–837 Klein UR, Nigg EA, Gruneberg U, 2006, Centromere targeting of the chromosomal passenger complex requires a ternary subcomplex of Borealin, Survivin, and the N-terminal domain of INCENP. Mol Biol Cell 17:2547–2558 Cooke CA, Heck MM, Earnshaw WC, 1987, The inner centromere protein, INCENP, antigens: movement from inner centromere to midbody during mitosis. J Cell Biol 105:2053–2067 Sessa F, Mapelli M, Ciferri C et al, 2005, Mechanism of Aurora B activation by INCENP and inhibition by hesperadin. Mol Cell 18:379–391 Carmena M, Earnshaw WC, 2006, INCENP at the kinase crossroads. Nat Cell Biol 8:110–111 Adams RR, Eckley DM, Vagnarelli P et al, 2001, Human INCENP colocalizes with the Aurora-B/AIRK2 kinase on chromosomes and is overexpressed in tumour cells. Chromosoma 110:65–74 Cutts SM, Fowler KJ, Kile BT et al, 1999, Defective chromosome segregation, microtubule bundling and nuclear bridging in inner centromere protein gene, Incenp)-disrupted mice. Hum Mol Genet 8:1145–1155 Nguyen HG, Ravid K, 2006, Tetraploidy/aneuploidy and stem cells in cancer promotion: the role of chromosome passenger proteins. J Cell Physiol 208:12–22 Greaves S, 2001, A roar for INCENP. Nat Cell Biol 3:E14 Wheatley SP, Carvalho A, Vagnarelli P, Earnshaw WC, 2001, INCENP is required for proper targeting of survivin to the centromeres and the anaphase spindle during mitosis. Curr Biol 11:886–890 Gerlach U, Kayser G, Walch A et al, 2006, Centrosome-, chromosomal-passenger- and cell-cycle-associated mRNAs are differentially regulated in the development of sporadic colorectal cancer. J Pathol 208:462–472 Jaffe ES, Harris NL, Stein H, Vardiman JW, 2001, Pathology and genetics of tumours of haematopoietic and lymphoid tissues. World Health Organization Classification of Tumours. IARC, Lyon Abramoff MD, Magelhaes PJ, Ram SJ, 2004, Image processing with imageJ. Biophoton Int 11:36–42 Massague J, 2004, G1 cell-cycle control and cancer. Nature 432:298–306 Perez de Castro I, de Carcer G, Malumbres M, 2007, A census of mitotic cancer genes: new insights into tumor cell biology and cancer therapy. Carcinogenesis 28:899–912 Storchova Z, Pellman D, 2004, From polyploidy to aneuploidy, genome instability and cancer. Nat Rev Mol Cell Biol 5:45–54 Altieri DC, 2003, Survivin in apoptosis control and cell cycle regulation in cancer. Prog Cell Cycle Res 5:447–452 Mainou-Fowler T, Overman LM, Dignum H et al, 2008, A new subtype-specific monoclonal antibody for IAP-survivin identifies high-risk patients with diffuse large B-cell lymphoma and improves the prognostic value of bcl-2. Int J Oncol 32:59–68 Cong XL, Han ZC, 2004, Survivin and leukaemia. Int J Hematol 80:232–238 Kajiwara Y, Yamasaki F, Hama S et al, 2003, Expression of survivin in astrocytic tumors: Correlation with malignant grade and prognosis. Cancer 97:1077–1083 Li F, 2005, Role of survivin and its splice variants in tumorigenesis. Br J Cancer 92:212–216 Araki K, Nozaki K, Ueba T, Tatsuka M, Hashimoto N, 2004, High expression of Aurora-B/Aurora and Ipll-like midbodyassociated protein, AIM-1, in astrocytomas. J Neurooncol 67: 53–64 Sorrentino R, Libertini S, Pallante Pl et al, 2005, Aurora B overexpression associates with the thyroid carcinoma undifferentiated phenotype and is required for thyroid carcinoma cell proliferation. J Clin Endocrinol Metab 90:928–935 Sistayanarain A, Tsuneyama K, Zheng H et al, 2006, Expression of Aurora-B kinase and phosphorylated histone H3 in hepatocellular carcinoma. Anticancer Res 26:585–593 Chieffi P, Cozzolino L, Kisslinger A et al, 2006, Aurora B expression directly correlates with prostate cancer malignancy and influence prostate cell proliferation. Prostate 66:326–333 Chang JL, Chen TH, Wang CF et al, 2006, Borealin/Dasra B is a cell cycle-regulated chromosomal passenger protein and its nuclear accumulation is linked to poor prognosis for human gastric cancer. Exp Cell Res 312:962–973 Miller TP, Grogan TM, Dahlberg S et al, 1994, Prognostic significance of the Ki-67-assosiated proliferative antigen in aggressive non-Hodgkin’s lymphomas: a prospective Southwest Oncology Group trial. Blood 83:1460–1466 Petit B, Chaury MP, Le Clorennec C et al, 2005, Indolent lymphoplasmacytic and marginal zone B-cell lymphomas: absence of both IRF4 and Ki-67 expression identifies a better prognostic subgroup. Haematologica 90:200–206 Lehman NL, Tibshirani R, Hsu JY et al, 2007, Oncogenic regulators and substrates of the anaphase promoting complex/ cyclosome are frequently overexpressed in malignant tumors. Am J Pathol 170:1793–1805 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 11 EP 17 DI 10.1007/s12253-008-9094-0 PG 7 ER PT J AU Bica, GC de Moura da Silva, LL Toscani, VN da Cruz, BMI Sa, G Graudenz, SM Zettler, GC AF Bica, Giuliano Claudia de Moura da Silva, Leiria Leonardo Toscani, Vieira Nadima da Cruz, Beatrice Manica Ivana Sa, Gustavo Graudenz, Silveira Marcia Zettler, Galleano Claudio TI MnSOD Gene Polymorphism Association with Steroid-Dependent Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MnSOD polymorphism; Male breast cancer; Female breast cancer; Prostate cancer; Steroid metabolism; Oxidative stress ID MnSOD polymorphism; Male breast cancer; Female breast cancer; Prostate cancer; Steroid metabolism; Oxidative stress AB Oxidative stress enhances carcinogenesis due to DNA damage. Manganese superoxide dismutase (MnSOD) Val16Ala polymorphism has been recently associated with breast and prostate cancer. The role of oxidative stress in male breast cancer is poorly investigated due to the low prevalence of this neoplasia. We studied the relationship between prostate cancer (PC), male (MBC) and female breast cancer (FBC) and this polymorphism in a case–control study. Human genetic polymorphism Val16Ala of MnSOD was obtained from blood and paraffin-embedded tumor samples. The polymorphism was determined in 11 cases of MBC, 51 cases of PC, 89 cases of FBC and 372 age-adjusted healthy controls by polymerase chain reaction–restriction fragment length polymorphism techniques using restriction enzyme Hae III. Chi-square or Fisher test were used to compare the MnSOD frequency distribution. The observed genotypic frequencies of all samples were AA=9.6% (n=50), VV=25.4% (n=133) and AV=64% (n=340), all at Hardy–Weinberg equilibrium. Breast and prostate cancer risk was elevated in male and female patients with the Ala/Ala genotype compared to controls (p=0.006, odds ratio=2.5, 95% confidence interval 1.393–4.541). Even though the frequency of the Ala allele was low (9.6%) in the studied population, these data support the hypothesis that MnSOD and oxidative stress play a significant role in breast cancer risk both in males and females and also brings new information on the role of this polymorphism in prostate cancer. This is the first study which provides some evidence that genetic polymorphism in the MnSOD gene may be associated with an increased risk of male breast cancer. Studies with a larger sample size are needed to confirm the findings. C1 [Bica, Giuliano Claudia] Fundacao Faculdade Federal de Ciencias Medicas de Porto Alegre, Rua Sarmento Leite 245, 90050-170 Porto Alegre, RS, Brazil. [de Moura da Silva, Leiria Leonardo] Fundacao Faculdade Federal de Ciencias Medicas de Porto Alegre, Rua Sarmento Leite 245, 90050-170 Porto Alegre, RS, Brazil. [Toscani, Vieira Nadima] Fundacao Faculdade Federal de Ciencias Medicas de Porto Alegre, Rua Sarmento Leite 245, 90050-170 Porto Alegre, RS, Brazil. [da Cruz, Beatrice Manica Ivana] Universidade Federal de Santa MariaSanta Maria, RS, Brazil. [Sa, Gustavo] Pontificia Universidade Catolica do Rio Grande do SulPorto Alegre, RS, Brazil. [Graudenz, Silveira Marcia] Fundacao Faculdade Federal de Ciencias Medicas de Porto Alegre, Rua Sarmento Leite 245, 90050-170 Porto Alegre, RS, Brazil. [Zettler, Galleano Claudio] Fundacao Faculdade Federal de Ciencias Medicas de Porto Alegre, Rua Sarmento Leite 245, 90050-170 Porto Alegre, RS, Brazil. RP Bica, GC (reprint author), Fundacao Faculdade Federal de Ciencias Medicas de Porto Alegre, 90050-170 Porto Alegre, Brazil. EM bicaclaudia@hotmail.com CR Martindale JL, Holbrook NJ, 2002, Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol 192:1–15 Suh YA, Arnold RS, Lassegue B et al, 1999, Cell transformation by the superoxide-generating oxidase Mox1. Nature 401:79–82 Arbiser JL, Petros J, Klafter R et al, 2002, Reactive oxygen generated by Nox1 triggers the angiogenic switch. Proc Natl Acad Sci U S A 99:715–720 Schwartz JL, Antoniades DZ, Zhao S, 1993, Molecular and biochemical reprogramming of oncogenesis through the activity of prooxidants and antioxidants. Ann NY Acad Sci 686: 262–278 Ross RK, Pike MC, Coetzee GA et al, 1998, Androgen metabolism and prostate cancer: establishing a model of genetic susceptibility. Cancer Res 58:4497–4504 Sutton A, Khoury H, Prip-Buus C, Cepanec C et al, 2003, The Ala16Val genetic dimorphism modulates the import of human manganese superoxide dismutase into rat liver mitochondria. Pharmacogenetics 13:145–157 Mitrunen K, Sillanpaa P, Kataja V et al, 2001, Association between manganese superoxide dismutase, MnSOD, gene polymorphism and breast cancer risk. Carcinogenesis 22:827–829 Millikan RC, Player J, de Cotret AR et al, 2004, Manganese superoxide dismutase Ala-9Val polymorphism and risk of breast cancer in a population-based case–control study of African Americans and whites. Breast Cancer Res 6:R264–274 Taufer M, Peres A, de Andrade VM et al, 2005, Is the Val16Ala manganese superoxide dismutase polymorphism associated with the aging process. J Gerontol A Biol Sci Med Sci 60:432–438 Woodson K, Tangrea JA, Lehman TA et al, 2003, Manganese superoxide dismutase, MnSOD, polymorphism, alpha-tocopherol supplementation and prostate cancer risk in the alpha-tocopherol, beta-carotene cancer prevention study, Finland). Cancer Causes Control 14:513–518 Wang LI, Neuberg D, Christiani DC, 2004, Asbestos exposure, manganese superoxide dismutase, MnSOD, genotype, and lung cancer risk. J Occup Environ Med 46:556–564 Stoehlmacher J, Ingles SA, Park DJ et al, 2002, The -9Ala/-9Val polymorphism in the mitochondrial targeting sequence of the manganese superoxide dismutase gene, MnSOD, is associated with age among Hispanics with colorectal carcinoma. Oncol Rep 9:235–238 Hung RJ, Boffetta P, Brennan P et al, 2004, Genetic polymorphisms of MPO, COMT, MnSOD, NQO1, interactions with environmental exposures and bladder cancer risk. Carcinogenesis 25:973–978 Cai Q, Shu XO, Wen Wet al, 2004, Genetic polymorphism in the manganese superoxide dismutase gene, antioxidant intake, and breast cancer risk: results from the Shanghai Breast Cancer Study. Breast Cancer Res 6:R647–R655 Mitrunen K, Hirvonen A, 2003, Molecular epidemiology of sporadic breast cancer. The role of polymorphic genes involved in oestrogen biosynthesis and metabolism. Mutat Res 544: 9–41 Donegan WL, 1991, Cancer of the breast in men. CA Cancer J Clin 41:339–354 Fentiman IS, Fourquet A, Hortobagyi GN, 2006, Male breast cancer. Lancet 367:595–604 Da Cruz IB, Oliveira G, Taufer M et al, 2003, Angiotensin Iconverting enzyme gene polymorphism in two ethnic groups living in Brazil's southern region: association with age. J Gerontol A Biol Sci Med Sci 58:M851–M856 Peres A, Bauer M, da Cruz IB et al, 2003, Immunophenotyping and T-cell proliferative capacity in a healthy aged population. Biogerontology 4:289–296 Little J, Bradley L, Bray M et al, 2002, Reporting, appraising and integrating data on genotype prevalence and gene-disease associations. Am J Epidem 156:300–310 Li Y, Huang TT, Carlson EJ et al, 1995, Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nat Genet 11:376–381 Mobley JA, Brueggemeier RW, 2004, Estrogen receptor-mediated regulation of oxidative stress and DNA damage in breast cancer. Carcinogenesis 25:3–9 Yager JD, Liehr JG, 1996, Molecular mechanisms of estrogen carcinogenesis. Ann Rev Pharmacol Toxicol 36:203–232 Cavalieri E, Frenkel K, Liehr JG et al, 2000, Estrogens as endogenous genotoxic agents–DNA adducts and mutations. J Natl Cancer Inst Monogr 27:75–93 Wani G, Milo GE, D’Ambrosio SM, 1998, Enhanced expression of the 8-oxo-7,8-dihydrodeoxyguanosine triphosphatase gene in human breast tumor cells. Cancer Lett 125:123–130 Han X, Liehr JG, 1994, 8-Hydroxylation of guanine bases in kidney and liver DNA of hamsters treated with estradiol: role of free radicals in estrogen-induced carcinogenesis. Cancer Res 54: 5515–5517 Musarrat J, Arezina-Wilson J, Wani AA, 1996, Prognostic and aetiological relevance of 8-hydroxyguanosine in human breast carcinogenesis. Eur J Cancer 32A:1209–1214 Egan KM, Thompson PA, Titus-Ernstoff L et al, 2003, MnSOD polymorphism and breast cancer in a population-based case– control study. Cancer Lett 199:27–33 Bhat HK, Calaf G, Hei TK et al, 2003, Critical role of oxidative stress in estrogen-induced carcinogenesis. Proc Natl Acad Sci U S A 100:3913–3918 Patel MM, Bhat HK, 2004, Differential oxidant potential of carcinogenic and weakly carcinogenic estrogens: involvement of metabolic activation and cytochrome P450. J Biochem Mol Toxicol 18:37–42 Hong CC, Tang BK, Rao V et al, 2004, Cytochrome P450 1A2, CYP1A2, activity, mammographic density, and oxidative stress: a cross-sectional study. Breast Cancer Res 6:R338–R351 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 19 EP 24 DI 10.1007/s12253-008-9064-6 PG 6 ER PT J AU Ioannou, GM Stathakis, E Lazaris, CA Papathomas, Th Tsiambas, E Koukoulis, KG AF Ioannou, G Maria Stathakis, Efstathios Lazaris, C Andreas Papathomas, Thomas Tsiambas, Evangelos Koukoulis, K George TI Immunohistochemical Evaluation of 95 Bone Marrow Reactive Plasmacytoses SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD56; Cyclin D1; Immunohistology; Bone marrow biopsies; Reactive plasmacytosis; Plasma cell myeloma ID CD56; Cyclin D1; Immunohistology; Bone marrow biopsies; Reactive plasmacytosis; Plasma cell myeloma AB We histologically and immunohistochemically studied 95 bone marrow (BM) reactive plasmacytoses. Ten biopsies from plasma cell myeloma (PCM) patients served as a control group. In addition, we studied 10 monoclonal gammopathy of undetermined significance (MGUS) cases. Histologically, plasmacytosis varied between 5% and 25% with an interstitial pattern of plasma cell (PC) distribution being characteristically displayed. Immunohistochemically, we did not find any CD56/NCAM nor cyclin D1 expression in all biopsies (95 of 95, 100%), not even a weak, doubtful one; PCs were all polyclonal and CD138 positive. On the contrary, myeloma-associated PCs showed monoclonality for .- or 1- light chain and strong CD56/NCAM immunoreactivity (8 of 10, 80%); four of them were cyclin D1 positive. Osteoblasts exhibited similar CD56/NCAM expression in both groups. Our data confirm the diagnostic utility of CD56/NCAM in the phenotypic characterization of polyclonal plasma cells, suggesting an important role of this particular immunomarker in the BM trephine study of polyclonal versus neoplastic plasmacytic infiltrations. C1 [Ioannou, G Maria] University of Thessaly, Medical School, Department of PathologyLarissa, Greece. [Stathakis, Efstathios] University of Thessaly, Medical School, Department of PathologyLarissa, Greece. [Lazaris, C Andreas] University of Thessaly, Medical School, Department of PathologyLarissa, Greece. [Papathomas, Thomas] University of Thessaly, Medical School, Department of PathologyLarissa, Greece. [Tsiambas, Evangelos] University of Thessaly, Medical School, Department of PathologyLarissa, Greece. [Koukoulis, K George] University of Thessaly, Medical School, Department of PathologyLarissa, Greece. RP Papathomas, Th (reprint author), University of Thessaly, Medical School, Department of Pathology, Larissa, Greece. EM thomaspapathomas@yahoo.gr CR Bain BJ, Clark DM, Lampert IA, Wilkins BS, 2001, Bone marrow pathology. Blackwell Science, Oxford Hyun BK, Kwa D, Gabaldon H, Ashton JK, 1976, Reactive plasmacytic lesions of the bone marrow. Am J Clin Pathol 65:921–928 Wei A, Juneja S, 2003, Bone marrow immunohistology of plasma cell neoplasms. J Clin Pathol 56:406–411 Grogan TM, Van Camp B, Kyle RA, Muller-Hermelink HK, Harris NL, 2001, Plasma cell neoplasms. In: Jaffe ES, Harris NL, Stein H, Vardiman JW, eds, World Health Organization classification of tumours. Pathology and genetics of tumours of haematopoietic and lymphoid tissues. IARC Press, Lyon, pp 142–156 Dick FR, 2002, Chronic lymphoproliferative disorders, immunoproliferative disorders, and malignant lymphoma. In: McClatchey KD, Weinberg RW, Reter RE, Foley T, eds, Clinical laboratory medicine. Lippincott Williams & Wilkins, Philadelphia, pp 923–963 Ely SA, Knowles DM, 2002, Expression of CD56/neural cell adhesion molecule correlates with the presence of lytic bone lesions in multiple myeloma and distinguishes myeloma from monoclonal gammopathy of undetermined significance and lymphomas with plasmacytoid differentiation. Am J Pathol 160: 1293–1299 Pellat-Deceunynck C, Barille S, Jego G et al, 1998, The absence of CD56, NCAM, on malignant plasma cells is a hallmark of plasma cell leukemia and of a special subset of multiple myeloma. Leukemia 12:1977–1982 Rawstron A, Barrans S, Blythe D et al, 1999, Distribution of myeloma plasma cells in peripheral blood and bone marrow correlates with CD56 expression. Br J Haematol 104:138–143 Pellat-Deceunynck C, Barille S, Puthier D et al, 1995, Adhesion molecules on human myeloma cells: significant changes in expression related to malignancy, tumor spreading, and immortalization. Cancer Res 55:3647–3653 Dahl IM, Rasmussen T, Kauric G, Husebekk A, 2002, Differential expression of CD56 and CD44 in the evolution of the extramedullary myeloma. Br J Haematol 116:273–277 Sahara N, Takeshita A, Shigeno K et al, 2002, Clinicopathological and prognostic characteristics of CD56-negative multiple myeloma. Br J Haematol 117:882–885 Hedvat CV, Comenzo RL, Teruya-Feldstein J et al, 2003, Insights into extramedullary tumour cell growth revealed by expression profiling of human plasmacytomas and multiple myeloma. Br J Haematol 122:728–744 Kremer M, Ott G, Nathrath M et al, 2005, Primary extramedullary plasmacytoma and multiple myeloma: phenotypic differences revealed by immunohistochemical analysis. J Pathol 205:92–101 Sahara N, Takeshita A, 2004, Prognostic significance of surface markers expressed in multiple myeloma: CD56 and other antigens. Leuk Lymphoma 45:61–65 Van Camp B, Durie BG, Spier C et al, 1990, Plasma cells in multiple myeloma express a natural killer cell-associated antigen: CD56, NKH-1; Leu-19). Blood 76:377–382 Martin P, Santon A, Bellas C, 2004, Neural cell adhesion molecule expression in plasma cells in bone marrow biopsies and aspirates allows discrimination between multiple myeloma, monoclonal gammopathy of uncertain significance and polyclonal plasmacytosis. Histopathology 44:375–380 Dunphy CH, Nies MK, Gabriel DA, 2007, Correlation of plasma cell percentages by CD138 immunohistochemistry, cyclin D1 status, and CD56 expression with clinical parameters and overall survival in plasma cell myeloma. Appl Immunohistochem Mol Morphol 15:248–254 Fend F, Kremer M, 2007, Diagnosis and classification of malignant lymphoma and related entities in the bone marrow trephine biopsy. Pathobiology 74:133–143 Kremer M, Quintanilla-Martinez L, Nahrig J, Von Schilling C, Fend F, 2005, Immunohistochemistry in bone marrow pathology: a useful adjunct for morphologic diagnosis. Virchows Arch 447:920–937 Sukpanichnant S, Cousar JB, Leelasiri A, Graber SE, Greer JP, Collins RD, 1994, Diagnostic criteria and histologic grading in multiple myeloma: histologic and immunohistologic analysis of 176 cases with clinical correlation. Hum Pathol 25:308–318 Cotelingam JD, 2003, Bone marrow biopsy: interpretive guidelines for the surgical pathologist. Adv Anat Pathol 10:8–26 Brunning RD, 2004, Bone Marrow. In: Rosai J, Houston M, eds, Rosai and Ackerman’s surgical pathology. Mosby, St. Louis, pp 2099–2108 Bataille R, Jego G, Robillard N et al, 2006, The phenotype of normal, reactive and malignant plasma cells. Identification of “many and multiple myelomas” and of new targets for myeloma therapy. Haematologica 91:1234–1240 Sezer O, Heider U, Zavrski I, Possinger K, 2001, Differentiation of monoclonal gammopathy of undetermined significance and multiple myeloma using flow cytometric characteristics of plasma cells. Haematologica 86:837–843 Naresh KN, Lampert I, Hasserjian R et al, 2006, Optimal processing of bone marrow trephine biopsy: the Hammersmith Protocol. J Clin Pathol 59:903–911 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 25 EP 29 DI 10.1007/s12253-008-9069-1 PG 5 ER PT J AU Qiao, L Wang, Y Pang, R Wang, J Dai, Y Ma, J Gu, Q Li, Z Zhang, Y Zou, B Lan, YH Wong, CYB AF Qiao, Liang Wang, Yan Pang, Roberta Wang, Jide Dai, Yun Ma, Juan Gu, Qing Li, Zesong Zhang, Yusheng Zou, Bing Lan, Y H Wong, C Y Benjamin TI Oncogene Functions of FHL2 Are Independent from NF-κBIα in Gastrointestinal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE FHL2; NF-κB; NF-κBIα; Interaction; Gastric cancer; Colon cancer ID FHL2; NF-κB; NF-κBIα; Interaction; Gastric cancer; Colon cancer AB Four and a half of LIM-only protein 2 (FHL2) is an adaptor protein that can interact with many transcription factors and thus plays a variety of biological functions. Previous studies by our group have demonstrated that suppression of FHL2 was capable of inducing tumor cell differentiation, and inhibiting the growth of experimental gastric and colon cancers. Therefore, FHL2 appears to function as an oncogene. In order to further explore the mechanisms of how FHL2 is involved in tumorigenesis, we attempted to test whether FHL2 has any direct association with nuclear factor (NF-κB), the most important transcription factor involved in apoptosis, inflammation, and carcinogenesis. Using an Yeast Two Hybrid (Y2H) screening system, we have shown that FHL2 may have an interaction with NF-κBIα, the coding gene for IκBα which is the most potent endogenous inhibitor for NF-κB activation. However, subsequent studies using co-immunoprecipitation and colocalization failed to confirm the Y2H finding. Downregulation of FHL2 by FHL2-siRNA down-regulated the expression of NF-κB p65. We therefore concluded that under the physiological condition, FHL2 may activate NF-κB pathway, even though such an activation may not be mediated by a direct binding of FHL2 to NF-κB inhibitor protein IκB. C1 [Qiao, Liang] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Wang, Yan] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Pang, Roberta] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Wang, Jide] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Dai, Yun] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Ma, Juan] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Gu, Qing] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Li, Zesong] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Zhang, Yusheng] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Zou, Bing] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Lan, Y H] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. [Wong, C Y Benjamin] Queen Mary Hospital, University of Hong Kong, Department of MedicineHong Kong, Hong Kong. RP Wong, CYB (reprint author), Queen Mary Hospital, University of Hong Kong, Department of Medicine, Hong Kong, Hong Kong. EM bcywong@hku.hk CR Chan KK, Tsui SK, Lee SM, Luk SC, Liew CC, Fung KP, Waye MM, Lee CY, 1998, Molecular cloning and characterization of FHL2, a novel LIM domain protein preferentially expressed in human heart. Gene 210:345–350 Genini M, Schwalbe P, Scholl FA, Remppis A, Mattei MG, Schafer BW, 1997, Subtractive cloning and characterization of DRAL, a novel LIM-domain protein down-regulated in rhabdomyosarcoma. DNA Cell Biol 16:433–442 Scholl FA, McLoughlin P, Ehler E, de Giovanni C, Schafer BW, 2000, DRAL is a p53-responsive gene whose four and a half LIM domain protein product induces apoptosis. J Cell Biol 151:495–506 Kinoshita M, Nakagawa T, Shimizu A, Katsuoka Y, 2005, Differently regulated androgen receptor transcriptional complex in prostate cancer compared with normal prostate. Int J Urol 12:390–397 Gabriel B, Mildenberger S, Weisser CW, Metzger E, Gitsch G, Schule R, Muller JM, 2004, Focal adhesion kinase interacts with the transcriptional coactivator FHL2 and both are overexpressed in epithelial ovarian cancer. Anticancer Res 24:921–927 Wang J, Yang Y, Xia HH, Gu Q, Lin MC, Jiang B, Peng Y, Li G, An X, Zhang Y, Zhuang Z, Zhang Z, Kung HF, Wong BC, 2007, Suppression of FHL2 expression induces cell differentiation and inhibits gastric and colon carcinogenesis. Gastroenterology 132:1066–1076 Wei Y, Renard CA, Labalette C,Wu Y, Levy L, Neuveut C, Prieur X, Flajolet M, Prigent S, Buendia MA, 2003, Identification of the LIM protein FHL2 as a coactivator of b-catenin. J Biol Chem 278:5188– 5194 Yan J, Zhu J, Zhong H, Lu Q, Huang C, Ye Q, 2003, BRCA1 interacts with FHL2 and enhances FHL2 transactivation function. FEBS Lett 553:183–189 Martin BT, Kleiber K,Wixler V, Raab M, Zimmer B, Kaufmann M, Strebhardt K, 2007, FHL2 regulates cell cycle-dependent and doxorubicin-induced p21Cip1/Waf1 expression in breast cancer cells. Cell Cycle 6:1779–1788 Muller JM, Isele U, Metzger E, Rempel A, Moser M, Pscherer A, Breyer T, Holubarsch C, Buettner R, Schule R, 2000, FHL2, a novel tissue-specific coactivator of the androgen receptor. EMBO J 19:359–369 Fimia GM, De Cesare D, Sassone-Corsi P, 2000, A family of LIM-only transcriptional coactivators: tissue-specific expression and selective activation of CREB and CREM. Mol Cell Biol 20:8613–8622 Samson T, Smyth N, Janetzky S, Wendler O, Muller JM, Schule R, von der Mark H, von der Mark K, Wixler V, 2004, The LIM-only proteins FHL2 and FHL3 interact with a- and b-subunits of the muscle a7b1 integrin receptor. J Biol Chem 279:28641–28652 Tanahashi H, Tabira T, 2000, Alzheimer’s disease-associated presenilin 2 interacts with DRAL, an LIM-domain protein. Hum Mol Genet 9:2281–2289 Purcell NH, Darwis D, Bueno OF, Muller JM, Schule R, Molkentin JD, 2004, Extracellular signal-regulated kinase 2 interacts with and is negatively regulated by the LIM-only protein FHL2 in cardiomyocytes. Mol Cell Biol 24:1081–1095 Morlon A, Sassone-Corsi P, 2003, The LIM-only protein FHL2 is a serum-inducible transcriptional coactivator of AP-1. Proc Natl Acad Sci USA 100:3977–3982 Chen D, Xu W, Bales E, Colmenares C, Conacci-Sorrell M, Ishii S, Stavnezer E, Campisi J, Fisher DE, Ben-Ze’ev A, Medrano EE, 2003, SKI activates Wnt/b-catenin signaling in human melanoma. Cancer Res 63:6626–6634 Labalette C, Renard CA, Neuveut C, Buendia MA, Wei Y, 2004, Interaction and functional cooperation between the LIM protein FHL2, CBP/p300, and b-catenin. Mol Cell Biol 24:10689–10702 Yang Y, Hou H, Haller EM, Nicosia SV, Bai W, 2005, Suppression of FOXO1 activity by FHL2 through SIRT1- mediated deacetylation. EMBO J 24:1021–1032 Nair SS, Guo Z, Mueller JM, Koochekpour S, Qiu Y, Tekmal RR, Schule R, Kung HJ, Kumar R, Vadlamudi RK, 2007, Proline-, glutamic acid-, and leucine-rich protein-1/modulator of nongenomic activity of estrogen receptor enhances androgen receptor functions through LIM-only coactivator, four-and-a-half LIM-only protein 2. Mol Endocrinol 21:613–624 Stilo R, Leonardi A, Formisano L, Di Jeso B, Vito P, Liguoro D, 2002, TUCAN/CARDINAL and DRAL participate in a common pathway for modulation of NF-kB activation. FEBS Lett 521:165–169 Kleiber K, Strebhardt K, Martin BT, 2007, The biological relevance of FHL2 in tumour cells and its role as a putative cancer target. Anticancer Res 27:55–61 Bai S, Kitaura H, Zhao H, Chen J, Muller JM, Schule R, 2005, FHL2 inhibits the activated osteoclast in a TRAF6-dependent manner. J Clin Invest 115:2742–2751 James P, Halladay J, Craig EA, 1996, Genomic libraries and a host strain designed for highly efficient two-hybrid selection in yeast. Genetics 144:1425–1436 Ding GR, Honda N, Nakahara T, Tian F, Yoshida M, Hirose H, Miyakoshi J, 2003, Radiosensitization by inhibition of IkB-a phosphorylation in human glioma cells. Radiat Res 160:232–237 Lightcap ES, McCormack TA, Pien CS, Chau V, Adams J, Elliott PJ, 2000, Proteasome inhibition measurements: clinical application. Clin Chem 46:673–683 Gao J, Pfeifer D, He LJ, Qiao F, Zhang Z, Arbman G, Wang ZL, Jia CR, Carstensen J, Sun XF, 2007, Association of NFKBIA polymorphism with colorectal cancer risk and prognosis in Swedish and Chinese populations. Scand J Gastroenterol 42:345– 350 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 31 EP 36 DI 10.1007/s12253-008-9085-1 PG 6 ER PT J AU Dehaghani, SA Rad, RN Fattahi, JM Khadang, B Kashef, AM Sarraf, Z Ghaderi, A AF Dehaghani, Samsami Alamtaj Rad, Rahimi Neda Fattahi, Javad Mohammad Khadang, Baharak Kashef, Amin Mohammad Sarraf, Zahra Ghaderi, Abbas TI Investigation of Soluble HER2 and Transforming Growth Factor Beta-1 Serum Levels in Gestational Trophoblastic Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HER2; TGF-β1; Gestational trophoblastic disease ID HER2; TGF-β1; Gestational trophoblastic disease AB HER2/neu and TGF-β1 are over-expressed in various types of malignancies. It appears that they play an important role in the biologic behavior of tumors and have prognostic value. Gestational tropoblastic diseases (GTDs) comprise of a heterogeneous group characterized by abnormally proliferating trophoblastic tissues, ranging from benign to malignant. The objective of this study was to measure and compare the serum levels of s-HER2 and TGF-β between patients with GTDs and pregnant and nonpregnant controls. Serum levels of s-HER2 and TGF-β1 were determined by ELISA method in 95 GTD patients (55 complete moles, 32 persistent moles, and 8 choriocarcinoma), 30 normal pregnant controls, and 22 normal nonpregnant controls. Mean serum level of s-HER2 did not differ significantly between patients and controls. TGF-β1 serum level was significantly higher in GTD patients (20.29±10.68 pg/ml with 95% confidence interval (CI) of 18.10–22.48 pg/ml) compared with pregnant controls (10.26±11.84 pg/ml with 95% CI of 5.75–14.76 pg/ml) and non-pregnant controls (7.27±9.61 pg/ml with 95% CI of 3.01–11.53 pg/ml) (P<0.001). Our findings suggest that TGF-β1 serum levels in GTD patients may represent a potential prognostic marker. Further investigations with larger sample size and more frequent sampling are required to elucidate this issue. C1 [Dehaghani, Samsami Alamtaj] Shiraz University of Medical Sciences, Hafez hospital, Department of Obstetric and Gynecology, 71345-1798 Shiraz, Iran. [Rad, Rahimi Neda] Shiraz University of Medical Sciences, Hafez hospital, Department of Obstetric and Gynecology, 71345-1798 Shiraz, Iran. [Fattahi, Javad Mohammad] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Khadang, Baharak] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Kashef, Amin Mohammad] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Sarraf, Zahra] Shiraz University of Medical Sciences, Hafez hospital, Department of Obstetric and Gynecology, 71345-1798 Shiraz, Iran. [Ghaderi, Abbas] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. RP Dehaghani, SA (reprint author), Shiraz University of Medical Sciences, Hafez hospital, Department of Obstetric and Gynecology, 71345-1798 Shiraz, Iran. EM samsamia@sums.ac.ir CR Genest DR, Berkowitz RS, Fisher RA, Newlands ES, Fehr M, 2003, Gestational trophoblastic disease. In: Tavassoli FA, Devilee P, eds, World Health Organization Classification of Tumors. Pathology and Genetics. Tumors of the Breast and Female Genital Organs. IARC, Lyon, pp 250–254 Sivanesaratnam V, 2003, Management of gestational trophoblastic disease in developing countries. Best Pract Res Clin Obstet Gynaecol 17:925–42 Hung MC, Lau YK, 1999, Basic science of HER2/neu: a review. Semin Oncol 26:51–59 Hung MC, Schechter AL, Chevray PY, Stern DF, Weinberg RA, 1986, Molecular cloning of the neu gene: absence of gross structural alteration in oncogenic alleles. Proc Natl Acad Sci U S A 83:261–264 Worthylake R, Opresko LK, Wiley HS, 1999, ErbB-2 amplification inhibits down-regulation and induces constitutive activation of both ErbB-2 and epidermal growth factor receptors. J Biol Chem 274:8865–8874 Nunes RA, Harris LN, 2002, The HER2 extracellular domain as a prognostic and predictive factor in breast cancer. Clin Breast Cancer 3:125–135 Tsuda H, Hirohashi S, Shimosato Y, Tanaka Y, Hirota T, Tsugane S, Shiraishi M, Toyoshima K, Yamamoto T, Terada M, 1990, Immunohistochemical study on overexpression of c-erbB-2 protein in human breast cancer: its correlation with gene amplification and long-term survival of patients. Jpn J Cancer Res 81:327–332 Cirisano FD, Karlan BY, 1996, The role of the HER2/neu oncogene in gynecologic cancers. J Soc Gynecol Investig 3:99–105 Blobe GC, Schiemann WP, Lodish HF, 2000, Role of transforming growth factor beta in human disease. N Engl J Med 342:1350–1358 Romagnani S, 2006, Regulation of the T cell response. Clin Exp Allergy 36:1357–1366 Desruisseau S, Palmari J, Giusti C, Romain S, Martin PM, Berthois Y, 2006, Determination of TGFbeta1 protein level in human primary breast cancers and its relationship with survival. Br J Cancer 94:239–246 Lin Y, Kikuchi S, Obata Y, Yagyu K, Tokyo Research Group on Prevention of Gastric Cancer, 2006, Serum levels of transforming growth factor beta1 are significantly correlated with venous invasion in patients with gastric cancer. J Gastroenterol Hepatol 21:432–437 Ivanovic V, Melman A, Davis-Joseph B, Valcic M, Geliebter J, 1995, Elevated plasma levels of TGF-b 1 in patients with invasive prostate cancer. Nat Med 1:282–284 Shim KS, Kim KH, Han WS, Park EB, 1999, Elevated serum levels of transforming growth factor-b1 in patients with colorectal carcinoma: its association with tumor progression and its significant decrease after curative surgical resection. Cancer 85:554–561 Tuncer ZS, Vegh GL, Fulop V, Genest DR, Mok SC, Berkowitz RS, 2000, Expression of epidermal growth factor receptor-related family products in gestational trophoblastic diseases and normal placenta and its relationship with development of postmolar tumor. Gynecol Oncol 77:389–393 Yang X, Zhang Z, Jia C, Li J, Yin L, Jiang S, 2002, The relationship between expression of c-ras, c-erbB-2, nm23, and p53 gene products and development of trophoblastic tumor and their predictive significance for the malignant transformation of complete hydatidiform mole. Gynecol Oncol 85:438–444 Pang ZJ, Xing FQ, 2003, Expression of transforming growth factor-beta and insulin-like growth factor in molar and placental tissues. Arch Gynecol Obstet 269:1–4 Hegab HM, Schindler AE, Rizk M, Ramadan M, 2003, Evaluation of tumour markers in molar pregnancy. Arch Gynecol Obstet 268:151–154 Kohorn EI, 2000, Measurement of CA-125 in trophoblastic disease. Gynecol Oncol 78:39–42 Hojo M, Morimoto T, Maluccio M, Asano T, Morimoto K, Lagman M, Shimbo T, Suthanthiran M, 1999, Cyclosporine induces cancer progression by a cell-autonomous mechanism. Nature 397:530–534 Massague J, 1990, The transforming growth factor-b family. Annu Rev Cell Biol 6:597–641 Norgaard P, Hougaard S, Poulsen HS, Spang-Thomsen M, 1995, Transforming growth factor b and cancer. Cancer Treat Rev 21:367–403 Saito H, Tsuj itani S, Oka S, Kondo A, Ikeguchi M, Maeta M, Kaibara N, 2000, An elevated serum level of TGF-β1 significantly correlated with lymph node metastasis and poor prognosis in patients with gastric carcinoma. Anticancer Res 20:4489–4493 Fukuchi M, Miyazaki T, Fukai Y, Nakajima M, Sohda M, Masuda N, Manda R, Tsukada K, Kato H, Kuwano H, 2004, Plasma level of transforming growth factor beta1 measured from the azygos vein predicts prognosis in patients with esophageal cancer. Clin Cancer Res 10:2738–2741 Glick AB, Kulkarni AB, Tennenbaum T, Hennings H, Flanders KC, O'Reilly M, 1993, Loss of expression of transforming growth factor beta in skin and skin tumors is associated with hyperproliferation and a high risk for malignant conversion. Proc Natl Acad Sci U S A 90:6076–6080 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 37 EP 40 DI 10.1007/s12253-008-9115-z PG 4 ER PT J AU Bori, R Cserni, G AF Bori, Rita Cserni, Gabor TI Basal Phenotype in Breast Carcinoma Occurring in Women Aged 35 or Younger SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Basal-like phenotype; BRCA1 mutation; Breast cancer in young women ID Basal-like phenotype; BRCA1 mutation; Breast cancer in young women AB Breast cancer in the young is considered a special clinical presentation of the disease. Sixty-nine breast cancer cases diagnosed at or before the age of 35 were analyzed for common morphological and immunophenotypical features of basal-like carcinomas. Sixteen carcinomas displayed the immunophenotypical characteristics (estrogen receptor and HER2 negativity and positivity for at least one of the following basal markers: cytokeratin 5 or 14, epidermal growth factor receptor, p63) of basal-like carcinomas, and most of them demonstrated characteristic histological features (pushing borders, lymphocytic peritumoral infiltrate, central hypocellular zone or necrosis, high mitotic rate) too. These tumors were more likely to be high-molecular-weight cytokeratin: 34betaE12 and p53 positive by immunohistochemistry. The presence of a basal-like phenotype can be important as concerns systemic treatment issues and could theoretically be associated with a higher rate of BRCA1 mutations in the young, because of the overlap of BRCA1 mutation associated breast carcinomas and the basal-like phenotype. C1 [Bori, Rita] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, 6000 Kecskemet, Hungary. [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, 6000 Kecskemet, Hungary. RP Bori, R (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, 6000 Kecskemet, Hungary. EM boririta@hotmail.com CR Fernandopulle SM, Cher-Siangang P, Tan PH, 2006, Breast carcinoma in women 35 years and younger: a pathological study. Pathology 38:219–222 Figueiredo JC, Ennis M, Knight JA et al, 2007, Influence of young age at diagnosis and family history of breast or ovarian cancer on breast cancer outcomes in a population-based cohort study. Breast Cancer Res Treat 105:69–80 Armes JE, Egan AJ, Southey MC et al, 1998, The histologic phenotypes of breast carcinoma occurring before age 40 years in women with and without BRCA1 or BRCA2 germline mutations: a population-based study. Cancer 83:2335–2345 Bennett IC, Freitas R Jr, Fentiman IS, 1991, Diagnosis of breast cancer in young women. Aust N Z J Surg 61:284–289 Tai P, Cserni G, Van de Steene J et al, 2005, Modeling the effect of age in T1-T2 breast cancer using the SEER database. BMC Cancer 5:130., DOI 10.1186/1471-2407-5-130 Trecate G, Vergnaghi D, Manoukian S et al, 2006, MRI in the early detection of breast cancer in women with high genetic risk. Tumori 92:517–523 Golshan M, Miron A, Nixon AJ, 2006, The prevalence of germline BRCA1 and BRCA2 mutations in young women with breast cancer undergoing breast-conservation therapy. Am J Surg 192:58–62 Lubinski J, Gorski B, Huzarski T et al, 2006, BRCA1-positive breast cancers in young women from Poland. Breast Cancer Res Treat 99:71–76 Loizidou M, Marcou Y, Anastasiadou V et al, 2007, Contribution of BRCA1 and BRCA2 germline mutations to the incidence of early-onset breast cancer in Cyprus. Clin Genet 71:165–170 Foulkes WD, Brunet JS, Stefansson IM et al, 2004, The prognostic implication of the basal-like, cyclin E high/p27 low/ p53+/glomeruloid-microvascular-proliferation+, phenotype of BRCA1-related breast cancer. Cancer Res 64:830–835 Foulkes WD, Metcalfe K, Sun P et al, 2004, Estrogen receptor status in BRCA1- and BRCA2-related breast cancer: the influence of age, grade, and histological type. Clin Cancer Res 10:2029– 2034 Laakso M, Loman N, Borg A et al, 2005, Cytokeratin 5/14- positive breast cancer: true basal phenotype confined to BRCA1 tumors. Mod Pathol 18:1321–1328 Lakhani SR, Reis-Filho JS, Fulford L et al, 2005, Prediction of BRCA1 status in patients with breast cancer using estrogen receptor and basal phenotype. Clin Cancer Res 11:5175–5180 Lidereau R, Eisinger F, Champeme MH et al, 2000, Major improvement in the efficacy of BRCA1 mutation screening using morphoclinical features of breast cancer. Cancer Res 60:1206–1210 Foulkes WD, Stefansson IM, Chappuis PO et al, 2003, Germline BRCA1 mutations and a basal epithelial phenotype in breast cancer. J Natl Cancer Inst 95:1482–1485 Fulford LG, Easton DF, Reis-Filho JS et al, 2006, Specific morphological features predictive for the basal phenotype in grade 3 invasive ductal carcinoma of breast. Histopathology 49:22–34 Sorlie T, Perou CM, Tibshirani R et al, 2001, Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98:10869–10874 Sorlie T, Tibshirani R, Parker J et al, 2003, Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A 100:8418–8423 Nielsen TO, Hsu FD, Jensen K et al, 2004, Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res 10:5367–5374 Bauer KR, Brown M, Cress RD et al, 2007, Descriptive analysis of estrogen receptor, ER)-negative, progesterone receptor, PR)- negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer Registry. Cancer 109:1721–1728 Rakha EA, El-Sayed ME, Green AR et al, 2007, Prognostic markers in triple-negative breast cancer. Cancer 109:25–32 Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19:403–410 Ellis IO, Pinder SE, Bobrow L et al, 2005, Pathology reporting of breast disease. NHS Publication No 58. Sheffield: NHS Cancer Screening Programmes and the Royal College of Pathologists. Accessed at 12 July 2007. http://www.cancerscreening.nhs.uk/ breastscreen/publications/nhsbsp58-low-resolution.pdf DakoCytomation. HercepTestTM, 8th ed. Accessed at 12 July 2007. http://dist.dako.com/prod_downloadpackageinsert.pdf? objectid=105073005 Takei H, Iino Y, Horiguchi J et al, 1997, Low and high molecular weight cytokeratins in invasive breast carcinoma. Oncol Rep 4:33–38 Ridolfi RL, Rosen PP, Port A et al, 1977, Medullary carcinoma of the breast: a clinicopathologic study with 10 year follow-up. Cancer 40:1365–1385 Tot T, 2000, The cytokeratin profile of medullary carcinoma of the breast. Histopathology 37:175–181 Jacquemier J, Padovani L, Rabayrol L et al, 2005, Typical medullary breast carcinomas have a basal/myoepithelial phenotype. J Pathol 207:260–268 Reis-Filho JS, Milanezi F, Steele D et al, 2006, Metaplastic breast carcinomas are basal-like tumours. Histopathology 49:10–21 Tsuda H, Takarabe T, Hasegawa F et al, 2000, Large, central acellular zones indicating myoepithelial tumor differentiation in high-grade invasive ductal carcinomas as markers of predisposition to lung and brain metastases. Am J Surg Pathol 24:197–202 Bratthauer GL, Miettinen M, Tavassoli FA, 2003, Cytokeratin immunoreactivity in lobular intraepithelial neoplasia. J Histochem Cytochem 51:1527–1531 van ’t Veer LJ, Dai H, van de Vijver MJ et al, 2002, Gene expression profiling predicts clinical outcome of breast cancer. Nature 415(6871):530–536 Banerjee S, Reis-Filho JS, Ashley S et al, 2006, Basal-like breast carcinomas: clinical outcome and response to chemotherapy. J Clin Pathol 59:729–735 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 41 EP 45 DI 10.1007/s12253-008-9090-4 PG 5 ER PT J AU Csete, B Lengyel, Zs Kadar, Zs Battyani, Z AF Csete, Bela Lengyel, Zsuzsanna Kadar, Zsolt Battyani, Zita TI Poly(Adenosine Diphosphate-Ribose) Polymerase-1 Expression in Cutaneous Malignant Melanomas as a New Molecular Marker of Aggressive Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cutaneous malignant melanoma; Immunohistochemistry; Poly(ADP-ribose) polymerase-1 ID Cutaneous malignant melanoma; Immunohistochemistry; Poly(ADP-ribose) polymerase-1 AB Poly(adenosine diphosphate-ribose) polymerases (PARPs) are a family of enzymes, which catalyses poly (ADP-ribosyl)ation of DNA-binding proteins and directly involved in genomic stability, DNA repair, and apoptosis. In this study, we evaluated the immunomorphology of PARP-1 in melanoma and its prognostic importance. We studied PARP-1 expression by immunohistochemistry in a selected series of 54 primary cutaneous malignant melanoma (CMM). The findings of the present study suggest that the neoplastic progression toward the invasive (both horizontal and vertical) growth phase of CMM cells is characterized by the loss of cleavage of PARP-1, probably signaling an imbalance of the apoptotic process in these cells and leading to further gain to aggression. Over-expression of full-length PARP-1 was correlated with recurrence and/or progression of the disease and so act as a promising new biological marker of CMM. Our study represents the evidence of a direct correlation between the PARP-1-mediated apoptotic process and the biologic behavior of CMM. C1 [Csete, Bela] University of Pecs, Department of Dermatology, Venereology and Oncodermatology, 20. Kodaly Z. u.Pecs, Hungary. [Lengyel, Zsuzsanna] University of Pecs, Department of Dermatology, Venereology and Oncodermatology, 20. Kodaly Z. u.Pecs, Hungary. [Kadar, Zsolt] University of Pecs, Department of Dermatology, Venereology and Oncodermatology, 20. Kodaly Z. u.Pecs, Hungary. [Battyani, Zita] University of Pecs, Department of Dermatology, Venereology and Oncodermatology, 20. Kodaly Z. u.Pecs, Hungary. RP Csete, B (reprint author), University of Pecs, Department of Dermatology, Venereology and Oncodermatology, Pecs, Hungary. EM benig@freemail.hu CR Ame JC, Spenkehauer C, de Murcia G, 2004, The PARP superfamily. Bioessays 26:882–893 Wang X, Ohnishi K, Takahasi A et al, 1998, Poly(ADP-ribosyl, ation is required for p53-dependent signal transduction induced by radiation. Oncogene 17:2819–2825 Muller WA, Erlanger M, 1994, Malignant melanoma in life insurance—thickness or anatomic layer? Versicherungsmedizin 46:193–195 Keilholz U, Martus P, Punt CJ et al, 2002, Prognostic factors for survival and factors associated with long-term remission in patients with advanced melanoma receiving cytokine-based treatments: second analysis of a randomised EORTC Melanoma Group trial comparing interferon-alpha2a, IFNalpha, and interleukin 2, IL-2, with or without cisplatin. Eur J Cancer 38:1501–1511 Jansen C, 1995, Effect of sunlight on the skin—what have we learned? Nord Med 110:85–87 Vodenicharov MD, Ghodgaonkar MM, Halappanavar SS et al, 2005, Mechanism of early biphasic activation of poly(ADPribose, polymerase-1 in response to ultraviolet B radiation. J Cell Sci 118:589–599 Staibano S, Pepe S, Muzio L et al, 2005, Poly(adenosine diphosphate-ribose, polymerase-1 expression in malignant melanomas from photoexposed areas of the head and neck region. Human Pathol 36:724–731 Balch CM, Buzaid AC, Atkins MB et al, 2001, Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 19:3635–3648 Masutani M, Nakagama H, Sugimura T, 2005, Poly(ADP-ribosyl, ation in relation to cancer and autoimmune disease. Cell Mol Life Sci 62:769–783 van den Oord JJ, Vandeghinste N, De Ley M et al, 1994, Bcl-2 expression in human melanocytes and melanocytic tumors. Am J Pathol 145:294–300 Boise LH, Gonzalez-Garcia M, Postema CE et al, 1993, Bcl-x, a bcl-2-related gene that functions as a dominant regulator of apoptotic cell death. Cell 74:597–608 Steller H, 1995, Mechanisms and genes of cellular suicide. Science 267:1445–1449 Tang L, Tron VA, Reed JC et al, 1998, Expression of apoptosis regulators in cutaneous malignant melanoma. Clin Cancer Res 4:1865–1871 Helmbach H, Kern MA, Rossmann E et al, 2002, Drug resistance towards etoposide and cisplatin in human melanoma cells is associated with drug-dependent apoptosis deficiency. J Invest Dermatol 118:923–932 Feleszko W, Mlynarczuk I, Olszewska D et al, 2002, Lovastatin potentiates antitumor activity of doxorubicin in murine melanoma via an apoptosis-dependent mechanism. Int J Cancer 100:111–118 Vaculova A, Hofmanova J, Soucek K et al, 2002, Tumor necrosis factor-alpha induces apoptosis associated with poly(ADP-ribose, polymerase cleavage in HT-29 colon cancer cells. Anticancer Res 22:1635–1639 Shyong EQ, Lu Y, Lazinsky A et al, 2002, Effects of the isoflavone 4V,5,7-trihydroxyisoflavone, genistein, on psoralen plus ultraviolet A radiation, PUVA)-induced photodamage. Carcinogenesis 23:317–321 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 47 EP 53 DI 10.1007/s12253-008-9086-0 PG 7 ER PT J AU Krenacs, T Zsakovics, I Diczhazi, Cs Ficsor, L Varga, SV Molnar, B AF Krenacs, Tibor Zsakovics, Ivett Diczhazi, Csaba Ficsor, Levente Varga, S V Molnar, Bela TI The Potential of Digital Microscopy in Breast Pathology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Digital microscopy; Breast pathology; Teleconsultation; Proficiency testing; Biomarker quantitation ID Digital microscopy; Breast pathology; Teleconsultation; Proficiency testing; Biomarker quantitation AB The rapidly evolving field of digital microscopy supports the efficient exploitation of inherent information from stained glass slides to offer widespread utilization in breast histopathology. Digital image signals can be accurately measured, integrated into databases and shared through computer networks. Therefore, digital microscopy can boost telepathology-consultation, gradual- and postgradual teaching, proficiency testing, intra- and interlaboratory validation of biomarker screening interpretation, and automated image analysis of biomarker expression for both diagnostics and research applications. This is a brief highlight of the potential of digital microscopy in breast pathology applications. C1 [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Zsakovics, Ivett] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Diczhazi, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Ficsor, Levente] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Varga, S V] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. RP Krenacs, T (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM krenacst@gmail.com CR Anderson WF, Luo S, Chatterjee N, Rosenberg PS, Matsuno RK, Goodman MT, Hernandez BY, Reichman M, Dolled-Filhart MP, O’Regan RM, Garcia-Closas M, Perou CM, Jatoi I, Cartun RW, Sherman ME, 2008, Human epidermal growth factor receptor-2 and estrogen receptor expression, a demonstration project using the residual tissue repository of the Surveillance, Epidemiology, and End Results, SEER, program. Breast Cancer Res Treat, Feb 7., DOI 10.1007/s10549-008-9918-3 Bloom K, Harrington D, 2004, Enhanced Accuracy of HER-2/neu immunohistochemical scoring using digital microscopy. Am J Clin Pathol 121:619–630 Borgen E, Naume B, Nesland JM, Nowels KW, Pavlak N, Ravkin I, Goldbard S, 2001, Use of automated microscopy for the detection of disseminated tumor cells in bone marrow samples. Cytometry 46:215–221 Cserneky M, Szende B, Fonyad L, Krenacst T, 2008, Telepathology in Hungary. In: Kumar S, ed, Telepathology. Springer, Berlin, in press) Drev P, Grazio SF, Bracko M, 2008, Tissue microarrays for routine diagnostic assessment of HER-2 status in breast carcinoma. Appl Immunohistochem Mol Morphol 16:179–184 Jan 25 [Epub ahead of print] Kayser K, Molnar B, Weinstein RS, 2006, Virtual slides technology. In: Virtual microscopy: fundamentals, applications, perspectives of electronic tissue-based diagnosis. VSV, Berlin, pp 103–123 Krenacs T, Ficsor L, Varga VS, Angeli V, Molnar B, 2008, Digital microscopy for boosting database integration and analysis in TMA studies. In: Simon R, ed, Methods in molecular biology. Humana, Totowa, NJ, in press) Kumar RK, Velan GM, Korell SO, Kandara M, Dee FR, Wakefield D, 2004, Virtual microscopy for learning and assessment in pathology. J Pathol 204:613–618 Lundin M, Lundin J, Helin H, Isola J, 2004, A digital atlas of breast histopathology: an application of web based virtual microscopy. J Clin Pathol 57:1288–1291 Schrader T, Hufnagl P, Schlake W, Dietel M, 2005, Study of efficiency of teleconsultation: the Telepathology Consultation Service of the Professional Assoziation of German Pathologists for the screening program of breast carcinoma, in German). Verh Dtsch Ges Pathol 89:211–218 Skaland I, Ovestad I, Janssen EA, Klos J, Kjellevold KH, Helliesen T, Baak JP, 2008, Digital image analysis improves the quality of subjective HER-2 expression scoring in breast cancer. Appl Immunohistochem Mol Morphol 16(2):185–190 Teodorovic I, Isabelle M, Carbone A, Passioukov A, Lejeune S, Jamine D, Therasse P, Gloghini A, Dinjens WN, Lam KH, Oomen MH, Spatz A, Ratcliffe C, Knox K, Mager R, Kerr D, Pezzella F, van Damme B, van de Vijver M, van Boven H, Morente MM, Alonso S, Kerjaschki D, Pammer J, Lopez-Guerrero JA, Llombart Bosch A, van Veen EB, Oosterhuis JW, Riegman PH, 2006, TuBaFrost 6: virtual microscopy in virtual tumour banking. Eur J Cancer. 42:3110–3116 Witzig TE, Bossy B, Kimlinger T, Roche PC, Ingle JN, Grant C, Donohue J, Suman VJ, Harrington D, Torre-Bueno J, Bauer KD, 2002, Detection of circulating cytokeratin-positive cells in the blood of breast cancer patients using immunomagnetic enrichment and digital microscopy. Clin Cancer Res 8:1085– 1091 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 55 EP 58 DI 10.1007/s12253-008-9087-z PG 4 ER PT J AU Kulka, J Szasz, MA Nemeth, Zs Madaras, L Schaff, Zs Molnar, AI Tokes, AM AF Kulka, Janina Szasz, Marcell Attila Nemeth, Zsuzsanna Madaras, Lilla Schaff, Zsuzsa Molnar, Arthur Istvan Tokes, Anna-Maria TI Expression of Tight Junction Protein Claudin-4 in Basal-Like Breast Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast carcinoma; Basal-like; Tissue microarray; Immunohistochemistry; Claudin-4 ID Breast carcinoma; Basal-like; Tissue microarray; Immunohistochemistry; Claudin-4 AB Claudins (CLDN) are tight junction proteins which contribute to the paracellular transport and ionic permeability of various epithelia. In recent years they came into focus for their suggested role in carcinogenesis and possible role in cancer therapy. According to our previous studies, in breast tissue CLDN4 is also related to the level of cellular differentiation. Thirty-eight estrogen (ER) and progesterone receptor (PgR) negative, HER2/neu negative, but cytokeratin 5/6 positive basal-like—mainly grade 3—breast carcinomas were compared with twenty-one grade 1, twenty-five grade 2 and twenty grade 3 non-basal-like invasive breast carcinomas, in respect to their CLDN4 expression. The immunohistochemical reactions were evaluated both semiquantitatively and by morphometrical analysis. Statistically significant difference (p=0.001) was observable regarding CLDN4 expression in the basal-like group as compared to grade 1 and 2 cancers. Further, CLDN4 expression was significantly higher (p=0.017) in the basal-like compared with the non-basal-like grade 3 carcinomas. Our results suggest that basal-like carcinomas are a subset of breast cancer with high level of CLDN4 protein expression. The finding is in accordance with our former observation that CLDN4 is indeed related to cellular differentiation. This observation may be seen as a further proof that basal-like carcinomas represent a separable group amongst grade 3 breast carcinomas. C1 [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Szasz, Marcell Attila] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Nemeth, Zsuzsanna] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Madaras, Lilla] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Molnar, Arthur Istvan] Semmelweis University, 1st Department of Surgery, 78 Ulloi ut, 1082 Budapest, Hungary. [Tokes, Anna-Maria] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. RP Kulka, J (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM kj@korb2.sote.hu CR Perou CM, Sorlie T, Eisen MB et al, 2000, Molecular portraits of human breast tumours. Nature 406:747–752 Fulford LG, Reis-Filho JS, Ryder K et al, 2007, Basal-like grade III invasive ductal carcinoma of the breast: patterns of metastasis and long-term survival. Breast Cancer Res 9:R4,, DOI 10.1186/bcr1636 Banerjee S, Reis-Filho JS, Ashley S et al, 2006, Basal-like breast carcinomas: clinical outcome and response to chemotherapy. J Clin Pathol 59:729–735 Hu Z, Fan C, Oh DS et al, 2006, The molecular portraits of breast tumors are conserved across microarray platforms. BMC Genomics 7:96 Sorlie T, 2007, Molecular classification of breast tumors: toward improved diagnostics and treatments.MethodsMol Biol 360:91–114 Da Silva L, Clarke C, Lakhani SR, 2007, Demystifying basal-like breast carcinomas. J Clin Pathol 60:1328–1332 Bryan BB, Schnitt SJ, Collins LC, 2006, Ductal carcinoma in situ with basal-like phenotype: a possible precursor to invasive basallike breast cancer. Mod Path 19:617–621 Buerger H, Otterbach F, Simon R et al, 1999, Different genetic pathways in the evolution of invasive breast cancer are associated with distinct morphological subtypes. J Path 189:521–526 Kondoh M, 2006, Claudin as a novel target for drug delivery system. Yakugaku Zasshi 126:711–721 Tokes AM, Kulka J, Paku S et al, 2005, The expression of five different claudins in invasive breast carcinomas: comparison of pT1pN1 and pT1pN0 tumors. Pathol Res Pract 201:537–544 Tokes AM, Kulka J, Paku S et al, 2005, Claudin-1, -3 and -4 proteins and mRNA expression in benign and malignant breast lesions: a research study. Breast Cancer Res 7:R296–R305 Furuse M, Fujita K, Hiiragi T et al, 1998, Claudin-1 and -2: novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol 141:1539–1550 Sobel G, Paska C, Szabo I et al, 2005, Increased expression of claudins in cervical squamous intraepithelial neoplasia and invasive carcinoma. Human Pathol 36:162–169 Sobel G, Nemeth J, Kiss A et al, 2006, Claudin 1 differentiates endometrioid and serous papillary endometrial adenocarcinoma. Gynecol Oncol 103:591–598 Michl P, Barth C, Buchholz M et al, 2003, Claudin-4 expression decreases invasiveness and metastatic potential of pancreatic cancer. Cancer Res 63:6265–6271 Michl P, Buchholz M, Rolke M et al, 2001, Claudin-4: a new target for pancreatic cancer treatment using Clostridium perfringens enterotoxin. Gastroenterology 121:678–684 Garcia S, Dales JP, Charafe-Jauffret E et al, 2007, Poor prognosis in breast carcinomas correlates with increased expression of targetable CD146 and c-Met and with proteomic basal-like phenotype. Human Pathol 38:830–841 Livasy CA, Karaca G, Nanda R et al, 2006, Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Path 19:264–271 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 59 EP 64 DI 10.1007/s12253-008-9089-x PG 6 ER PT J AU Josselin, N Libouban, H Dib, M Ifrah, N Legrand, E Basle, FM Audran, M Chappard, D AF Josselin, Nicolas Libouban, Helene Dib, Mamoun Ifrah, Norbert Legrand, Erick Basle, Felix Michel Audran, Maurice Chappard, Daniel TI Quantification of Dendritic Cells and Osteoclasts in the Bone Marrow of Patients with Monoclonal Gammopathy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE B-cell lymphoma; Bone histomorphometry; Bone resorption; Dendritic cells; Monoclonal gammopathy; Myeloma; Osteoclast ID B-cell lymphoma; Bone histomorphometry; Bone resorption; Dendritic cells; Monoclonal gammopathy; Myeloma; Osteoclast AB The purpose of this study was to find histological clues for reliable differentiation between monoclonal gammopathy of undetermined significance (MGUS) and myeloma when clinical parameters are controversial. Differential appearance of dendritic cells and osteoclasts, two cell types developing from the monocytic lineage upon distinct cytokine activation profile, might be a useful approach. Bone and bone-marrow biopsies performed in 105 patients were studied using histomorphometry after identification of osteoclasts (by histochemical identification of tartrate resistant acid phosphatase) and dendritic cells (by immunohistochemical detection of the S-100 protein). Patients were classified by the World Health Organization criteria but histopathological criteria were more adapted to identify MGUS (53 cases), myeloma (46), B-cell lymphoma (six) since six myeloma were not correctly classified. Histomorphometry was compared to 15 control cases. The number of marrow dendritic cell was significantly increased with B-cell lymphoma >MGUS >myeloma > controls. Dendritic cell were often mixed with lymphoma cells. Myeloma had increased bone resorption with a high osteoclast number and moderate increase in dendritic cells. B-cell lymphomas had a considerable increase in dendritic cell but presented mononucleated osteoclasts. These findings can help in the classification of MGUS in the early stages of the disease and could help to propose preventive treatments. C1 [Josselin, Nicolas] U922-LHEA, INSERM, Faculte de Medecine, 49045 Angers, France. [Libouban, Helene] U922-LHEA, INSERM, Faculte de Medecine, 49045 Angers, France. [Dib, Mamoun] CHU d’Angers, Service d’hematologie and UPRES EA 3863, 49933 Angers, France. [Ifrah, Norbert] CHU d’Angers, Service d’hematologie and UPRES EA 3863, 49933 Angers, France. [Legrand, Erick] U922-LHEA, INSERM, Faculte de Medecine, 49045 Angers, France. [Basle, Felix Michel] U922-LHEA, INSERM, Faculte de Medecine, 49045 Angers, France. [Audran, Maurice] U922-LHEA, INSERM, Faculte de Medecine, 49045 Angers, France. [Chappard, Daniel] U922-LHEA, INSERM, Faculte de Medecine, 49045 Angers, France. RP Chappard, D (reprint author), U922-LHEA, INSERM, Faculte de Medecine, 49045 Angers, France. EM daniel.chappard@univ-angers.fr CR Kyle RA, Therneau TM, Rajkumar SV et al, 2004, Long-term follow-up of 241 patients with monoclonal gammopathy of undetermined significance: the original Mayo Clinic series 25 years later. Mayo Clin Proc 79:859–866 Blouin S, Basle MF, Chappard D, 2008, Interactions between microenvironment and cancer cells in two animal models of bone metastasis. Br J Cancer 98:809–815 Bataille R, Chappard D, Basle M, 1995, Excessive bone resorption in human plasmacytomas: direct induction by tumour cells in vivo. Br J Haematol 90:721–724 Colla S, Zhan F, Xiong W et al, 2007, The oxidative stress response regulates DKK1 expression through the JNK signaling cascade in multiple myeloma plasma cells. Blood 109:4470–4477 Tian E, Zhan F, Walker R et al, 2003, The role of the Wntsignaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N Engl J Med 349:2483–2494 Aubin JE, Bonnelye E, 2000, Osteoprotegerin and its ligand: a new paradigm for regulation of osteoclastogenesis and bone resorption. Osteoporos Int 11:905–913 Banchereau J, Briere F, Caux C et al, 2000, Immunobiology of dendritic cells. Annu Rev Immunol 18:767–811 Dubois B, Massacrier C, Vanbervliet B et al, 1998, Critical role of IL-12 in dendritic cell-induced differentiation of naive B lymphocytes. J Immunol 161:2223–2231 Dubois B, Bridon JM, Fayette J et al, 1999, Dendritic cells directly modulate B cell growth and differentiation. J Leukoc Biol 66:224–230 Rettig MB, Ma HJ, Vescio RA et al, 1997, Kaposi’s sarcomaassociated herpesvirus infection of bone marrow dendritic cells from multiple myeloma patients. Science 276:1851–1854 Olsen SJ, Tarte K, Sherman W et al, 1998, Evidence against KSHV infection in the pathogenesis of multiple myeloma. Virus Res 57:197–202 Miyamoto T, Ohneda O, Arai F et al, 2001, Bifurcation of osteoclasts and dendritic cells from common progenitors. Blood 98:2544–2554 Hsu HC, Lee YM, Yang CF et al, 2001, Detection of Kaposi sarcoma-associated herpesvirus in bone marrow biopsy samples from patients with multiple myeloma. Cancer 91:1409–1413 Brown RD, Pope B, Murray A et al, 2001, Dendritic cells from patients with myeloma are numerically normal but functionally defective as they fail to up-regulate CD80, B7–1, expression after huCD40LT stimulation because of inhibition by transforming growth factor-beta1 and interleukin-10. Blood 98:2992–2998 Ratta M, Fagnoni F, Curti A et al, 2002, Dendritic cells are functionally defective in multiple myeloma: the role of interleukin- 6. Blood 100:230–237 Bataille R, Chappard D, Basle M, 1996, Quantifiable excess of bone resorption in monoclonal gammopathy is an early symptom of malignancy: a prospective study of 87 bone biopsies. Blood 87:4762–4769 Rossi JF, Chappard D, Marcelli C et al, 1990, Microosteoclast resorption as a characteristic feature of B-cell malignancies other than multiple myeloma. Brit J Haematol 76:469–475 Coindre JM, Reiffers J, Goussot JF et al, 1984, Histomorphometric analysis of sclerotic bone from idiopathic myeloid metaplasia, nine cases). J Pathol 144:163–169 Schmidt A, Blanchet O, Dib M et al, 2007, Bone changes in myelofibrosis with myeloid metaplasia: A histomorphometric and microcomputed tomographic study. Eur J Haematol 78:500–509 Beebe K, 2000, Alcohol/xylene: the unlikely fixative /dehydrant/ clearant. J Histotechnol 23:45–50 Chappard D, Palle S, Alexandre C et al, 1986, Simultaneous identification of calcified cartilage bone and osteoid tissue on plastic sections. New polychrome procedures specially adapted to image analysor systems. J Histotechnol 9:95–97 Parfitt AM, Drezner MK, Glorieux FG et al, 1987, Bone histomorphometry: standardization of nomenclature, symbols, and units. J Bone Miner Res 2:595–610, Report of the ASBMR Histomorphometry Nomenclature Committee Chappard D, Alexandre C, Riffat G, 1983, Histochemical identification of osteoclasts. Review of current methods and reappraisal of a simple procedure for routine diagnosis on undecalcified human iliac bone biopsies. Basic Appl Histochem 27:75–85 Chappard D, 1990, Osteoclast count on human bone biopsies: Why and How? In: Takahashi HE, ed, Bone morphometry. Nishimura-Smith–Gordon, Niigata, pp 248–255 van Hensbergen Y, Luykx-de Bakker SA, Heideman DA et al, 2001, Rapid stereology based quantitative immunohistochemistry of dendritic cells in lymph nodes: a methodological study. Anal Cell Pathol 22:143–149 Hsieh HL, Schafer BW, Sasaki N et al, 2003, Expression analysis of S100 proteins and RAGE in human tumors using tissue microarrays. Biochem Biophys Res Commun 307:375–381 Grogan TM, van Kamp B, Kyle RA et al, 2001, Mature B-cell neoplasms. In: Jaffe ES, Harris NL, Stein H, JW V, eds, World Health Organization Classification of Tumours Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues. IARC, Lyon, pp 142–148 Meunier P, Courpron P, 1976, Iliac trabecular bone volume in 236 controls—representativeness of iliac samples. In: Jaworski ZFG, klosevych S, eds, Proceeding of the first worhshop on bone histomorphometry. Univ. of Ottawa, Ottawa, pp 100–105 Bataille R, Chappard D, Klein B, 1992, Mechanisms of bone lesions in multiple myeloma. Hematol Oncol Clin North Am 6:285–295 Chappard D, Rossi JF, Bataille R et al, 1991, Osteoclast cytomorphometry demonstrates an abnormal population in B-cell malignancies but not in multiple myeloma. Calcif Tissue Int 48:13–17 Chappard D, Bataille R, Alexandre C et al, 1986, Monoclonal gammopathy of undetermined significance or multiple myeloma? Histomorphometric analysis of bone changes in 46 patients. J Exp Clin Cancer Res 5:239–248 Barretina J, Junca J, Llano A et al, 2003, CXCR4 and SDF-1 expression in B-cell chronic lymphocytic leukemia and stage of the disease. Ann Hematol 82:500–505 Marcelli C, Chappard D, Rossi JF et al, 1988, Histologic evidence of an abnormal bone remodeling in B cell malignancies other than multiple myeloma. Cancer 62:1163–1170 Tsuge K, Takeda H, Kawada S et al, 2005, Characterization of dendritic cells in differentiated thyroid cancer. J Pathol 205:565–576 Okuyama T, Maehara Y, Kakeji Yet al, 1998, Interrelation between tumor-associated cell surface glycoprotein and host immune response in gastric carcinoma patients. Cancer 82:1468–1475 Ocqueteau M, Orfao A, Almeida J et al, 1998, Immunophenotypic characterization of plasma cells from monoclonal gammopathy of undetermined significance patients. Implications for the differential diagnosis between MGUS and multiple myeloma. Am J Pathol 152:1655–1665 Goldman SA, Baker E, Weyant RJ et al, 1998, Peritumoral CD1apositive dendritic cells are associated with improved survival in patients with tongue carcinoma. Arch Otolaryngol Head Neck Surg 124:641–646 Bethwaite PB, Holloway LJ, Thornton A et al, 1996, Infiltration by immunocompetent cells in early stage invasive carcinoma of the uterine cervix: a prognostic study. Pathology 28:321–327 Eisenthal A, Polyvkin N, Bramante-Schreiber L et al, 2001, Expression of dendritic cells in ovarian tumors correlates with clinical outcome in patients with ovarian cancer. Hum Pathol 32:803–807 Frassanito MA, Cusmai A, Dammacco F, 2001, Deregulated cytokine network and defective Th1 immune response in multiple myeloma. Clin Exp Immunol 125:190–197 Wu L Dakic A, 2004, Development of dendritic cell system. Cell Mol Immunol 1:112–118 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 65 EP 72 DI 10.1007/s12253-008-9092-2 PG 8 ER PT J AU Hussein, RAM Abdel-Magid, MW Saleh, R Nada, E AF Hussein, Rezk A Mahmoud Abdel-Magid, M Wafaa Saleh, Ramadan Nada, Essam TI Phenotypical Characteristics of the Immune Cells in Allergic Contact Dermatitis, Atopic Dermatitis and Pityriasis Rosea SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Immunity; Immune cells; Dermatitis ID Immunity; Immune cells; Dermatitis AB Allergic contact dermatitis (ACD) is a cellmediated, delayed type IV immunologic reaction. Atopic dermatitis (AD) is a chronic inflammatory skin disease that results from a complex interaction between immunologic, genetic, and environmental factors. Pityriasis rosea (PR) is a self-limited eruption of unknown etiology. Immune cell infiltrate is a constant feature in the inflammatory skin diseases. Here, we performed phenotypical characterization of the immune cells in ACD, AD and PR (ten cases each). We performed immunohistochemical stains for B cells (CD20), T cells (CD3), histiocytes (CD68) and T cells with cytotoxic activity (granzyme-B). The data were compared with findings in 20 specimens of normal skin. The results were scored as mean values of positively stained immune cells. Immunohistochemistry showed significantly high counts of immune cells in lesional skin (ACD, AD and PR) compared to the normal one (p<0.05). In the lesional skin, the immune cells were composed predominantly of CD3+ T lymphocytes and CD68+ cells (histiocytes). Some of the CD3+ cells were granzyme B+. The counts of some immune cells (CD3+ and CD68+) were high in ACD compared to AD and PR. The counts of CD20+ and granzyme B+ cells were high in PR compared to ACD and AD. However, these differences did not reach the level of statistical significance. The present data describe the profile of the immune cell infiltrate in AD, ACD and PR. The cellmediated immunity seems to have critical role in the development of these lesions. C1 [Hussein, Rezk A Mahmoud] Assuit University, Assuit University Hospitals, Department of PathologyAssuit, Egypt. [Abdel-Magid, M Wafaa] Sohag University, Sohag University Hospitals, Department of DermatologySohag, Egypt. [Saleh, Ramadan] Sohag University, Sohag University Hospitals, Department of DermatologySohag, Egypt. [Nada, Essam] Sohag University, Sohag University Hospitals, Department of DermatologySohag, Egypt. RP Hussein, RAM (reprint author), Assuit University, Assuit University Hospitals, Department of Pathology, Assuit, Egypt. EM mrcpath17@gmail.com CR McCleskey PE, Swerlick RA, 2001, Clinical review: thioureas and allergic contact dermatitis. Cutis 68:387–396 Felter SP, Robinson MK, Basketter DA, Gerberick GF, 2002, A review of the scientific basis for uncertainty factors for use in quantitative risk assessment for the induction of allergic contact dermatitis. Contact Dermatitis 47:257–266 Brasch J, Burgard J, Sterry W, 1992, Common pathogenetic pathways in allergic and irritant contact dermatitis. J Invest Dermatol 98:166–170 Hunger RE, Yawalkar N, Braathen LR, Brand CU, 2001, CD1apositive dendritic cells transport the antigen DNCB intracellularly from the skin to the regional lymph nodes in the induction phase of allergic contact dermatitis. Arch Dermatol Res 293:420–426 Yawalkar N, Schmid S, Braathen LR, Pichler WJ, 2001, Perforin and granzyme B may contribute to skin inflammation in atopic dermatitis and psoriasis. Br J Dermatol 144:1133–1139 Deguchi M, Aiba S, Ohtani H, Nagura H, Tagami H, 2002, Comparison of the distribution and numbers of antigen-presenting cells among T-lymphocyte-mediated dermatoses: CD1a+, factor XIIIa+, and CD68+ cells in eczematous dermatitis, psoriasis, lichen planus and graft-versus-host disease. Arch Dermatol Res 294:297–302 Kwong CW, Sang WK, 2000, Re: One-year review of pityriasis rosea at the National Skin Centre, Singapore. Ann Acad Med Singapore 29:548 Cavanaugh RM Jr, 1983, Pityriasis rosea in children. A review. Clin Pediatr, Phila, 22:200–203 Chuh AA, Chan HH, Zawar V, 2004, Is human herpesvirus 7 the causative agent of pityriasis rosea?-A critical review. Int J Dermatol 43:870–785 Reinhold U, Kukel S, Goeden B, Neumann U, Wehrmann W, Kreysel HW, 1991, Interleukin-4 promotes the expansion of skininfiltrating lymphocytes from atopic dermatitis in vitro. J Invest Dermatol 96:370–375 Jung K, Linse F, Pals ST, Heller R, Moths C, Neumann C, 1997, Adhesion molecules in atopic dermatitis: patch tests elicited by house dust mite. Contact Dermatitis 37:163–172 Baker BS, Lambert S, Powles AV, Valdimarsson H, Fry L, 1988, Epidermal DR+ T6-dendritic cells in inflammatory skin diseases. Acta Derm Venereol 68:209–217 Bos JD, Hagenaars C, Das PK, Krieg SR, Voorn WJ, Kapsenberg ML, 1989, Predominance of “memory” T cells, CD4+, CDw29+, over “naive” T cells, CD4+, CD45R+, in both normal and diseased human skin. Arch Dermatol Res 281:24–30 Hussein MR, Ahmed RA, 2005, Analysis of the mononuclear inflammatory cell infiltrate in the non-tumorigenic, pre-tumorigenic and tumorigenic keratinocytic hyperproliferative lesions of the skin. Cancer Biol Ther 4:819–821 Hussein MR, Elsers DA, Fadel SA, Omar AE, 2006, Immunohistological characterisation of tumour infiltrating lymphocytes in melanocytic skin lesions. J Clin Pathol 59:316–324 Hussein MR, Ahmed RA, 2005, Analysis of the mononuclear inflammatory cell infiltrate in the cirrhotic, dysplastic nodules and hepatocellular carcinomas in patients with chronic hepatitis C infection. Cancer Biol Ther 4:1075–1078 Hussein MR, Abou-Deif ES, Bedaiwy MA, Said TM, Mustafa MG, Nada E, Ezat A, Agarwal A, 2005, Phenotypic characterization of the immune and mast cell infiltrates in the human testis shows normal and abnormal spermatogenesis. Fertil Steril 83:1447–1453 Gerber BO, Zanni MP, Uguccioni M, Loetscher M, Mackay CR, Pichler WJ, Yawalkar N, Baggiolini M, Moser B, 1997, Functional expression of the eotaxin receptor CCR3 in T lymphocytes co-localizing with eosinophils. Curr Biol 7:836–843 Amerio P, Frezzolini A, Feliciani C, Verdolini R, Teofoli P, De Pita O, Puddu P, 2003, Eotaxins and CCR3 receptor in inflammatory and allergic skin diseases: therapeutical implications. Curr Drug Targets Inflamm Allergy 2:81–94 Ying S, Barata LT, Meng Q, Grant JA, Barkans J, Durham SR, Kay AB, 1998, High-affinity immunoglobulin E receptor, Fc epsilon RI)-bearing eosinophils, mast cells, macrophages and Langerhans’ cells in allergen-induced late-phase cutaneous reactions in atopic subjects. Immunology 93:281–288 Ying S, Meng Q, Barata LT, Kay AB, 2001, Macrophage inflammatory protein-1alpha and C-C chemokine receptor-1 in allergen-induced skin late-phase reactions: relationship to macrophages, neutrophils, basophils, eosinophils and T lymphocytes. Clin Exp Allergy 31:1724–1731 Howe K, Foresman P, Griffin T, Johnson W, 1996, Pityriasis rubra pilaris with acantholysis. J Cutan Pathol 23:270–274 Reinhold U, Kukel S, Goeden B, Neumann U, Kreysel HW, 1991, Functional characterization of skin-infiltrating lymphocytes in atopic dermatitis. Clin Exp Immunol 86:444–448 Lugovic L, Lipozenocic J, Jakic-Razumovic J, 2001, Atopic dermatitis: immunophenotyping of inflammatory cells in skin lesions. Int J Dermatol 40:489–494 Nordlind K, Liden S, 1995, Gamma/delta T cells and human skin reactivity to heavy metals. Arch Dermatol Res 287:137–141 Kiekens RC, Thepen T, Oosting AJ, Bihari IC, Van De Winkel JG, Bruijnzeel-Koomen CA, Knol EF, 2001, Heterogeneity within tissue-specific macrophage and dendritic cell populations during cutaneous inflammation in atopic dermatitis. Br J Dermatol 145:957–965 Cantani A, 2001, Pathogenesis of atopic dermatitis, AD, and the role of allergic factors. Eur Rev Med Pharmacol Sci 5:95–117 Bjerke JR, 2002, The skin as an immunological organ. Tidsskr Nor Laegeforen 122:793–796 Yawalkar N, Hunger RE, Buri C, Schmid S, Egli F, Brand CU, Mueller C, Pichler WJ, Braathen LR, 2001, A comparative study of the expression of cytotoxic proteins in allergic contact dermatitis and psoriasis: spongiotic skin lesions in allergic contact dermatitis are highly infiltrated by T cells expressing perforin and granzyme B. Am J Pathol 158:803–808 Caproni M, Torchia D, Antiga E, Terranova M, Volpi W, Del Bianco E, D’Agata A, Fabbri P, 2007, The comparative effects of tacrolimus and hydrocortisone in adult atopic dermatitis: an immunohistochemical study. Br J Dermatol 156:312–319 Hussein MR, 2005, Tumour-infiltrating lymphocytes and melanoma tumorigenesis: an insight. Br J Dermatol 153:18–21 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 73 EP 79 DI 10.1007/s12253-008-9103-3 PG 7 ER PT J AU Bedaiwy, AM Hussein, RAEM Biscotti, Ch Falcone, T AF Bedaiwy, Ali Mohamed Hussein, Rezk Abd-Elwahed Mahmoud Biscotti, Charles Falcone, Tommaso TI Pelvic Endometriosis is Rarely Associated with Ovarian Borderline Tumours, Cytologic and Architectural Atypia: A Clinicopathologic Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometriosis; Cytologic atypia; Pattern atypia ID Endometriosis; Cytologic atypia; Pattern atypia AB Endometriotic foci, especially ovarian ones, with epithelial cytologic atypia may be precursors of cancer. This study presents an overview of the atypical cytological and histopathological findings associated with endometriosis. Six cases of endometriosis, with atypical histological and cytological changes, were obtained from the archives of the Department of Pathology at Cleveland Clinic Foundation between year 2000 and 2003. The size of the base from which these cases were drawn was 2000 cases of endometriosis. The age range of the patients was from 29 to 52 years. The clinical presentations included infertility (three cases), pelvic pain (three cases), adenexal and pelvic masses (four cases). Stage IV endometriosis with extensive pelvic involvement was found in two patients. Intraoperatively, the endometriotic lesions involved the ovaries (all cases); Cul de sac (four cases); urinary bladder (two cases); sigmoid colon, hemidiaphragms, and uterine vessels (one case each). The endometriotic lesions were associated with uterine leiomyomas (two patients) and adenocarcinoma of the vagina (one patient). Histologically, in addition to endometrial type glands and stroma, usually found in endometriosis, we observed both cytologic and pattern atypism involving the epithelium in all cases. The features of cytologic atypia included nuclear stratification, hyperchromatism, and pleomorphism. The features of pattern atypia were complex glandular pattern, papillary formations and psammoma bodies. In two cases, these features were sufficient for diagnosis of borderline Mullerian seromucinous tumours. One patient had recurred with metastatic adenocarcinoma of the vault. She died later from disseminated metastatic disease. There is a rare association between pelvic endometriosis and borderline ovarian tumours (three cases), cytologic and pattern atypia (two cases); mesothelial hyperplasia, endosalpingiosis (two cases), and metastasis (one case). Cytologic and pattern atypia can develop in the endometriotic foci and therefore, these lesions should be thoroughly scrutinized for presence of these changes. Our findings recommend surgical excision of these foci rather than their simple cauterization. C1 [Bedaiwy, Ali Mohamed] The Assuit University Hospitals and The Cleveland Clinic Foundation, Department of Obstetrics and GynaecologyCleveland, OH, USA. [Hussein, Rezk Abd-Elwahed Mahmoud] King Khalid University, Assir Central Hospital, Department of PathologyAbha, Saudi Arabia. [Biscotti, Charles] The Cleveland Clinic Foundation, Department of PathologyCleveland, OH, USA. [Falcone, Tommaso] The Cleveland Clinic Foundation, Department of Obstetrics and GynaecologyCleveland, OH, USA. RP Hussein, RAEM (reprint author), King Khalid University, Assir Central Hospital, Department of Pathology, Abha, Saudi Arabia. EM mrcpath17@gmail.com CR Akahane T, Sekizawa A, Purwosunu Y et al, 2007, The role of p53 mutation in the carcinomas arising from endometriosis. Int J Gynecol Pathol 26:345–351 Ali-Fehmi R, Khalifeh I, Bandyopadhyay S et al, 2006, Patterns of loss of heterozygosity at 10q23.3 and microsatellite instability in endometriosis, atypical endometriosis, and ovarian carcinoma arising in association with endometriosis. Int J Gynecol Pathol 25:223–229 Bese T, Simsek Y, Bese N et al, 2003, Extensive pelvic endometriosis with malignant change in tamoxifen-treated postmenopausal women. Int J Gynecol Cancer 13:376–380 Clement PB, 2007, The pathology of endometriosis: a survey of the many faces of a common disease emphasizing diagnostic pitfalls and unusual and newly appreciated aspects. Adv Anat Pathol 14:241–260 Edghill EL, Bingham C, Ellard S et al, 2006, Mutations in hepatocyte nuclear factor-1beta and their related phenotypes. J Med Genet 43:84–90 Friedrich M, Villena-Heinsen C, Mink D et al, 1999, Carcinosarcoma, endometrial intraepithelial carcinoma and endometriosis after tamoxifen therapy in breast cancer. Eur J Obstet Gynecol Reprod Biol 82:85–87 Groisman GM, Meir A, 2003, CD10 is helpful in detecting occult or inconspicuous endometrial stromal cells in cases of presumptive endometriosis. Arch Pathol Lab Med 127:1003–1006 Guo L, Liu T, Lang J, 2001, The malignant potential of ovarian atypical endometriosis. Zhonghua Bing Li Xue Za Zhi 30:169– 172 Higashiguchi A, Yamada T, Susumu N et al, 2007, Specific expression of hepatocyte nuclear factor-1beta in the ovarian clear cell adenocarcinoma and its application to cytological diagnosis. Cancer Sci 98:387–391 Hutton RL, Dalton SR, 2007, Primary peritoneal serous borderline tumors. Arch Pathol Lab Med 131:138–144 Jelovsek JE, Winans C, Brainard J et al, 2004, Endometriosis of the liver containing Mullerian adenosarcoma: case report. Am J Obstet Gynecol 191:1725–1727 Kato N, Sasou S, Motoyama T, 2006, Expression of hepatocyte nuclear factor-1beta, HNF-1beta, in clear cell tumors and endometriosis of the ovary. Mod Pathol 19:83–89 Korner M, Burckhardt E, Mazzucchelli L, 2006, Higher frequency of chromosomal aberrations in ovarian endometriosis compared to extragonadal endometriosis: A possible link to endometrioid adenocarcinoma. Mod Pathol 19:1615–1623 Lee KR, Nucci MR, 2003, Ovarian mucinous and mixed epithelial carcinomas of Mullerian, endocervical-like, type: a clinicopathologic analysis of four cases of an uncommon variant associated with endometriosis. Int J Gynecol Pathol 22:42–51 Leiserowitz GS, Gumbs JL, Oi R et al, 2003, Endometriosisrelated malignancies. Int J Gynecol Cancer 13:466–471 Leng JH, Lang JH, Zhao XY et al, 2006, Visual and histologic analysis of laparoscopic diagnosis of endometriosis. Zhonghua Fu Chan Ke Za Zhi 41:111–113 Madej P, Plewka A, Madej JA et al, 2006, Nucleolar organizer regions, NORs, in adenomyosis. Pathol Res Pract 202:433–437 Marchino GL, Gennarelli G, Enria R et al, 2005, Diagnosis of pelvic endometriosis with use of macroscopic versus histologic findings. Fertil Steril 84:12–15 McCluggage WG, Bryson C, Lamki H et al, 2000, Benign, borderline, and malignant endometrioid neoplasia arising in endometriosis in association with tamoxifen therapy. Int J Gynecol Pathol 19:276–279 Mhawech P, Kinkel K, Vlastos G et al, 2002, Ovarian carcinomas in endometriosis: an immunohistochemical and comparative genomic hybridization study. Int J Gynecol Pathol 21:401–406 Minkowitz G, 1996, Psammoma bodies in endometriosis: clinical, cytological, and physiopathological implications. Diagn Cytopathol 14:331–333 Mok SC, Kwong J, Welch WR et al, 2007, Etiology and pathogenesis of epithelial ovarian cancer. Dis Markers 23:367– 376 Obata K, Hoshiai H, 2000, Common genetic changes between endometriosis and ovarian cancer. Gynecol Obstet Invest 50, Suppl 1):39–43 Oral E, Ilvan S, Tustas E et al, 2003, Prevalence of endometriosis in malignant epithelial ovary tumours. Eur J Obstet Gynecol Reprod Biol 109:97–101 Parker RL, Dadmanesh F, Young RH et al, 2004, Polypoid endometriosis: a clinicopathologic analysis of 24 cases and a review of the literature. Am J Surg Pathol 28:285–297 Qian J, Shi Y, Chen X, 2000, Clinical analysis of 25 cases of malignant transformation of endometriosis of the ovary. Zhonghua Fu Chan Ke Za Zhi 35:667–669 Gruppo Italiano per lo Studio dell’Endometriosi, 2001, Relationship between stage, site and morphological characteristics of pelvic endometriosis and pain. Hum Reprod 16:2668–2671 Ria R, Loverro G, Vacca A et al, 2002, Angiogenesis extent and expression of matrix metalloproteinase-2 and -9 agree with progression of ovarian endometriomas. Eur J Clin Invest 32:199–206 Santeusanio G, Ventura L, Partenzi A et al, 1999, Omental endosalpingiosis with endometrial-type stroma in a woman with extensive hemorrhagic pelvic endometriosis. Am J Clin Pathol 111:248–251 Sekizawa A, Amemiya S, Otsuka J et al, 2004, Malignant transformation of endometriosis: application of laser microdissection for analysis of genetic alterations according to pathological changes. Med Electron Microsc 37:97–100 Selvaggi SM, 2002, Pelvic endometriosis diagnosed on touch imprint cytology. Diagn Cytopathol 27:379–381 Spaczynski M, Kedzia W, 2001, Ovarian cancer and endometriosis. Ginekol Pol 72:268–272 Staats PN, Clement PB, Young RH, 2007, Primary endometrioid adenocarcinoma of the vagina: a clinicopathologic study of 18 cases. Am J Surg Pathol 31:1490–1501 Stratton P, Winkel CA, Sinaii N et al, 2002, Location, color, size, depth, and volume may predict endometriosis in lesions resected at surgery. Fertil Steril 78:743–749 Swiersz LM, 2002, Role of endometriosis in cancer and tumor development. Ann N Y Acad Sci 955:281–292 discussion 293– 285, 396–406 Thomas EJ, Campbell IG, 2000, Evidence that endometriosis behaves in a malignant manner. Gynecol Obstet Invest 50(Suppl 1): 2–10 Thomas EJ, Campbell IG, 2000, Molecular genetic defects in endometriosis. Gynecol Obstet Invest 50(Suppl 1):44–50 Ulrich U, Richter O, Wardelmann E et al, 2003, Endometriosis and malignoma. Zentralbl Gynakol 125:239–242 Uzan C, Cortez A, Dufournet C et al, 2005, Endometrium from women with and without endometriosis, and peritoneal, ovarian and bowel endometriosis, show different c-kit protein expression. J Reprod Immunol 65:55–63 Valenzuela P, Ramos P, Redondo S et al, 2007, Endometrioid adenocarcinoma of the ovary and endometriosis. Eur J Obstet Gynecol Reprod Biol 134:83–86 Walter AJ, Hentz JG, Magtibay PM et al, 2001, Endometriosis: correlation between histologic and visual findings at laparoscopy. Am J Obstet Gynecol 184:1407–1411 discussion 1411–1403 Walter I, Handler J, Reifinger M et al, 2001, Association of endometriosis in horses with differentiation of periglandular myofibroblasts and changes of extracellular matrix proteins. Reproduction 121:581–586 Yamamoto S, Tsuda H, Aida S et al, 2007, Immunohistochemical detection of hepatocyte nuclear factor 1beta in ovarian and endometrial clear-cell adenocarcinomas and nonneoplastic endometrium. Hum Pathol 38:1074–1080 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 81 EP 88 DI 10.1007/s12253-008-9072-6 PG 8 ER PT J AU Eric, A Juranic, Z Milovanovic, Z Markovic, I Inic, M Stanojevic-Bakic, N Vojinovic-Golubovic, V AF Eric, Aleksandra Juranic, Zorica Milovanovic, Zorka Markovic, Ivan Inic, Momcilo Stanojevic-Bakic, Nevenka Vojinovic-Golubovic, Vesna TI Effects of Humoral Immunity and Calreticulin Overexpression on Postoperative Course in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Calreticulin; Humoral immunity; Surgery ID Breast cancer; Calreticulin; Humoral immunity; Surgery AB The aim was to investigate whether the humoral immunity and overexpression of calreticulin in tumor tissue determined before surgery, correlate with incidence of metastases in breast cancer patients within two years after operation. Before operation, their humoral immunity and overexpression of caleticulin and Her-2/neu in tumor tissue were analyzed by immunohystochemistry. In 23 patients with metastases in regionally lymph nodes, seven had Her-2/neu overexpression. Among those seven patients, three developed distant metastasis (two women one year and in one woman two years after surgery) and all of them showed the presence of stromal IgG immunoreactivity and overexpression of calreticulin in their tumors tissue. Preliminary data showed that serum IgG immunoreactivity to tumor stroma in combination with overexpression of calreticulin in tumor cells correlate with postoperative appearance of metastases, particularly in the group of patients with Her-2/neu overexpressed tumors and metastases in axillary lymph nodes. C1 [Eric, Aleksandra] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. [Juranic, Zorica] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. [Milovanovic, Zorka] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. [Markovic, Ivan] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. [Inic, Momcilo] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. [Stanojevic-Bakic, Nevenka] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. [Vojinovic-Golubovic, Vesna] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. RP Eric, A (reprint author), Institute of Oncology and Radiology of Serbia, 11 000 Belgrade, Serbia. EM eric.aleksandra@ncrc.ac.yu CR Scanlan MJ, Gout I, Gordon CM, Williamson B, Stockert E, Gure AO 2001 Humoral immunity to human breast cancer: antigen definition and quantitative analysis of mRNA expression. Cancer Immun 1:4 Hayashi E, Kuramitsu Y, Okada F, Fujimoto M, Zhang X, Kobayashi M, Iizuka N, Ueyama Y, Nakamura K 2005 Proteomic profiling for cancer progression: Differential display analysis for the expression of intracellular proteins between regressive and progressive cancer cell lines. Proteomics 5(4):1024–1032 Dissemond J, Busch M, Kothen T, Mors J, Weimann TK, Lindeke A, Goos M, Wagner SN 2004 Differential downregulation of endoplasmic reticulum-residing chaperones calnexin and calreticulin in human metastatic melanoma. Cancer Lett 203(2):225–231 Sipione S, Ewen C, Shostak I, Michalak M, Bleackley RC 2005 Impaired cytolytic activity in calreticulin-deficient CTLs. J Immunol 174(6):3212–3219 Fraser SA, Karimi R, Michalak M, Hudig D 2000 Perforin lytic activity is controlled by calreticulin. J Immunol 164(8):4150–4155 Colleoni M, Rotmensz N, Peruzzotti G, Maisonneuve P, Mazzarol G, Pruneri G 2005 Size of breast cancer metastases in axillary lymph nodes: clinical relevance of minimal lymph node involvement. J Clin Oncol 23(7):1379–1389 Ross JS, Fletcher JA 1998 The HER-2/neu Oncogene in breast cancer: Prognostic factor, predictive factor, and target for therapy. Oncologist 3(4):237–252 Barbera-Guillem E, Nyhus JK, Wolford CC, Friece CR, Sampsel JW 2002 Vascular endothelial growth factor secretion by tumorinfiltrating macrophages essentially supports tumor angiogenesis and IgG immune complexes potentate the process. Cancer Res 62:7042–7049 Shushakova N, Eden G, Dangers M, Zwirner J, Menne J, Gueler F, Luft FC, Haller H, Dumler I 2005 The urokinase/urokinase receptor system mediates the IgG immune complex-induced inflammation in lung. J Immunol 175(6):4060–4068 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 89 EP 90 DI 10.1007/s12253-008-9112-2 PG 2 ER PT J AU Yu, X Xu, X Han, B Zhou, R AF Yu, Xiaoling Xu, Xiaohui Han, Baojian Zhou, Rongxiang TI Inhibitor of DNA Binding-1 Overexpression in Prostate Cancer: Relevance to Tumor Differentiation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Id1; Protein expression; mRNA quantitation; Differentiation ID Prostate cancer; Id1; Protein expression; mRNA quantitation; Differentiation AB Inhibitor of DNA binding-1 (Id1) is a dominantnegative regulator of basic helix–loop–helix transcription factor, which control malignant cell behaviors in several types of carcinomas. This study aimed to find the relationship between Id1 expression and some clinical parameters. Paraffin-embedded tissue specimens from two normal human prostates, 12 benign prostatic hyperplasia (BPH), 43 prostate cancers(PCa) were detected by immunofluorescence assay. Prostatectomy samples from 11 BPH and 28 PCa were used for real time RT-PCR. The relationship between Id1 staining and several patient’s clinical parameters, including Gleason grade, PSA, clinical stage, and size of tumor, was further analyzed. Significant up-regulated Id1 protein was shown in prostate cancer specimens, while only weak expression in some BPH samples (5/12). Analyzed by image software, the mean proportion of Id1 positive staining remarkably increased with the increasing of Gleason grade in prostate cancer specimens (r=0.9967, P<0.01). Id1 expression was not significantly associated with PSA, TNM stage or tumor size. Furthermore, the average mRNA of prostate cancer was 3.09 times of BPH. This study confirms that Id1 protein and mRNA are over expressed in prostate cancer tissues. Overexpression of Id1 protein correlates with tumor tissue differentiation. We propose that Id1 over expression can be used in the analysis of the progression of prostate cancer. C1 [Yu, Xiaoling] Medical College of Qingdao University, Department of PathophysiologyQingdao, China. [Xu, Xiaohui] Medical College of Qingdao University, Department of PathophysiologyQingdao, China. [Han, Baojian] Medical College of Qingdao University, Department of Urinary SurgeryQingdao, China. [Zhou, Rongxiang] Medical College of Qingdao University, Department of Urinary SurgeryQingdao, China. RP Yu, X (reprint author), Medical College of Qingdao University, Department of Pathophysiology, Qingdao, China. EM xiaoling_yu2004@163.com CR Jemal A, Siegel R, Ward E et al, 2007, Cancer statistics, 2007. CA Cancer J Clin 57:43–66 Hsing AW, Tsao L, Devesa SS, 2000, International trends and patterns of prostate cancer incidence and mortality. Int Cancer 85:60–67,, review) Sim HG, Cheng CW, 2005, Changing demography of prostate cancer in Asia. Eur J Cancer 41:834–845 Gu F, 2000, Epidemiological survey of benign prostatic hyperplasia and prostate cancer in China. Chin Med J, Engl, 113:299–302 Hsing AW, Devesa SS, Jin F et al, 1998, Rising incidence of prostate cancer in Shanghai. Cancer Epidemiol Biomarkers Prev 7:83–84, letter) Hasskarl J, Munger K, 2002, Id proteins—tumor marker or oncogenes? Cancer Biology and Therapy 1:91–96, review) Perk J, Iavarone A, Benezra R, 2005, Id family of helix–loop– helix proteins in cancer. Nat Rev Cancer 5:603–604 Han S, Guo C, Hong L et al, 2004, Expression and significance of Id1 helix–loop–helix protein overexpression in gastric cancer. Cancer Lett 216:63–71 Matsuda Y, Yamagiwa S, Takamura M et al, 2005, Overexpressed Id-1 is associated with a high risk of hepatocellular carcinoma development in patients with cirrhosis without transcriptional repression of p16. Cancer 104:1037–1044 Straume O, Akslen LA, 2005, Strong expression of ID1 protein is associated with decreased survival, increased expression of ephrin-A1/EPHA2, and reduced thrombospondin-1 in malignant melanoma. Br J Cancer 93:933–938 Benezra R, Davis RL, Lockshon D et al, 1990, The protein Id: a negative regulator of helix–loop–helix DNA binding proteins. Cell 61:49–59 Alani RM, Hasskarl J, Grace M et al, 1999, Immortalization of primary human keratinocytes by the helix–loop–helix protein,Id- 1. Proc Natl Acad Sci USA 96:9637–9641 Lyden D, Young AZ, Zagzag D et al, 1999, Id1 and Id3 are required for neurogenesis, angiogenesis and vascularization of tumour xenografts. Nature 401:670–677 Ruzinova MB, Benezra R, 2003, Id proteins in development, cell cycle and cancer. Trends Cell Biol 13:410–418 Wong YC, Wang X, Ling MT, 2004, Id-1 expression and cell survival. Apoptosis 9:279–289,, review) Jang KS, Han HX, Paik SS et al, 2006, Id-1 overexpression in invasive ductal carcinoma cells is significantly associated with intratumoral microvessel density in ER-negative/node-positive breast cancer. Cancer Lett 244:203–210 Chetcuti A, Marqan S, Mann S et al, 2001, Identification of differentially expressed genes in organ-confined prostate cancer by gene expression array. Prostate 47:132–140 Ouyang XS, Wang X, Lee DT et al, 2002, Overexpression of ID-1 in prostate cancer. J Urol 167:2598–2602 Coppe JP, Itahana Y, Moore DH et al, 2004, Id-1 and Id-2 proteins as molecular markers for human prostate cancer progression. Clin Cancer Res 10:2044–2051 Yu DS, Hsieh DS, Chang SY, 2006, Increasing expression of GST-pi MIF, and ID1 genes in chemoresistant prostate cancer cells. Arch Androl 52:275–281 Lin JC, Chang SY, Hsieh DS et al, 2005, Modulation of mitogenactivated protein kinase cascades by differentiation-1 protein: acquired drug resistance of hormone independent prostate cancer cells. J Urol 174:2022–2026 Ouyang XS, Wang X, Ling MT et al, 2002, Id-1 stimulates serum independent prostate cancer cell proliferation through inactivation of p16(INK4a)/pRB pathway. Carcinogenesis 23: 721–725 Asirvatham AJ, Schmidt MA, Chaudhary J, 2006, Non-redundant inhibitor of differentiation, Id, gene expression and function in human prostate epithelial cells. Prostate 66:921–935 Perk J, Gil-Bazo I, Chin Y et al, 2006, Reassessment of id1 protein expression in human mammary, prostate, and bladder cancers using a monospecific rabbit monoclonal anti-id1 antibody. Cancer Res 66:10870–10877 Schindl M, Schoppmann SF, Strobel T et al, 2003, Level of Id-1 protein expression correlates with poor differentiation, enhanced malignant potential and more aggressive clinical behavior of epithelial ovarian tumors. Clin Cancer Res 9:779–785 Lin CQ, Singh J, Murata K et al, 2000, A role for Id-1 in the aggressive phenotype and steroid hormone response of human breast cancer cells. Cancer Res 60:1332–1340 Ling MT, Wang X, Lee DT et al, 2004, Id1 expression induces androgen-independent prostate cancer cell growth through activation of epidermal growth factor receptor, EGF-R). Carcinogenesis 25:517–525 Ling MT, Wanq X, Ouyanq XS et al, 2002, Activation of MAPK signaling pathway is essential for Id1 induced serum independent prostate cancer cell growth. Oncogene 21:8498–8505 Ling MT, Wang X, Ouyang XS et al, 2003, Id1 expression promotes cell survival through activation of NF-kB signaling pathway in prostate cancer cells. Oncogene 22:4498–4508 Fong S, Itahana Y, Sumida T et al, 2003, Id-1 as a molecular target in therapy for breast cancer cell invasion and metastasis. Proc Natl Acad Sci USA 100:13543–13548 Fong S, Debs RJ, Desprez PY, 2004, Id genes and proteins as promising targets in cancer therapy. Trends Mol Med 10:387–392, review) Tsuchiya T, Okaji Y, Tsuno NH et al, 2005, Targeting Id1 and Id3 inhibits peritoneal metastasis of gastric cancer. Cancer Sci 96:784–790 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 91 EP 96 DI 10.1007/s12253-008-9096-y PG 6 ER PT J AU Mannweiler, S Pummer, K Auprich, M Galle, G Mehes, G Ratschek, M Tsybrovskyy, O Moinfar, F AF Mannweiler, Sebastian Pummer, Karl Auprich, Marco Galle, Gunter Mehes, Gabor Ratschek, Manfred Tsybrovskyy, Oleksiy Moinfar, Farid TI Diagnostic Yield of Touch Imprint Cytology of Prostate Core Needle Biopsies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Biopsy; Diagnosis; Imprint cytology; Prostate; Cancer ID Biopsy; Diagnosis; Imprint cytology; Prostate; Cancer AB Touch imprint cytology may provide additional information to core needle biopsy interpretation according to previous reports. The aim of this study was to investigate the diagnostic yield of this method in the diagnosis of prostate carcinoma. For this purpose, 452 transrectal prostate needle biopsies were evaluated from 56 patients. All patients were clinically suspicious of having prostate carcinoma. Two touch imprints were prepared from each fresh biopsy cylinder. Results of the standard histology and of the touch imprint evaluation were compared. Histologically negative biopsy cylinders were further evaluated for prostate carcinoma by fine step serial sectioning. The standard histological examination showed adenocarcinoma in 27 patients. Touch imprint cytology revealed atypical cells suspicious of carcinoma in 38 patients. This group included all 27 patients with positive standard histology and further 11 patients with initially negative core biopsy. Following serial sectioning, in three out of these 11 samples, histological evidence of a carcinoma could be proven. Fine step serial sectioning of all 29 core biopsies negative for carcinoma by standard histological examination, 26 patients remained negative. All three core biopsies initially negative by standard histology but positive after serial sectioning had cytology findings suspicious of carcinoma. We conclude, that in problematic cases the additional use of touch imprint cytology and serial sectioning of prostate core needle biopsies significantly improve the diagnostic accuracy. C1 [Mannweiler, Sebastian] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. [Pummer, Karl] Medical University of Graz, Department of UrologyGraz, Austria. [Auprich, Marco] Medical University of Graz, Department of UrologyGraz, Austria. [Galle, Gunter] Medical University of Graz, Department of UrologyGraz, Austria. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Ratschek, Manfred] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. [Tsybrovskyy, Oleksiy] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. [Moinfar, Farid] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. RP Mannweiler, S (reprint author), Medical University of Graz, Department of Pathology, A-8036 Graz, Austria. EM Sebastian.Mannweiler@meduni-graz.at CR Hodge KK, McNeal JE, Terris MK et al, 1989, Random systematic versus directed ultrasound guided transrectal core biopsies of the prostate. J Urol 142:71–74 Fleshner NE, O, Sullivan M, Fair WR, 1997, Prevalence and predictors of a positive repeat transrectal ultrasound guided needle biopsy of the prostate. J Urol 158:505–508 Jacobs TW, Silverman JF, Schroeder B, Raza S, Baum JK, Schnitt SJ, 1999, Accuracy of touch imprint cytology of image-directed breast core needle biopsies. Acta Cytol 43:169–174 Gentry JF, 1986, Pelvic lymph node metastases in prostatic carcinoma. The value of touch imprint cytology. Am J Surg Pathol 10:718–727 Chieco P, Bertaccini A, Giovannini C, Stecca BA, Martorana G, 2001, Telomerase activity in touch-imprint cell preparation from fresh prostate needle biopsy specimens. Eur Urol 40:666–672 Lo J, Billie-Jo K, Amling CHL, Robertson CN, Layfield LJ, 1996, Correlation of DNA ploidy and histologic diagnosis from prostate core-needle biopsies: is DNA ploidy more sensitive than histology for the diagnosis of carcinoma in small specimens? J Surg Oncol 63:41–45 Eble JN, Sauter G, Epstein JI, Sesterhenn IA, 2004, Tumours of the urinary system and male genital organs. World Health Organization classification of tumours. IARC, Lyon Willems JS, Lowhagen T, 1981, Transrectal fine-needle aspiration biopsy for cytologic diagnosis and grading of prostatic carcinoma. Prostate 2:381–395 Epstein NA, 1976, Prostatic biopsy. A morphologic correlation of aspiration cytology with needle biopsy histology. Cancer 38:2078–2087 Leistenschneider W, Nagel R, 1984, Atlas of Prostatic Cytology. Springer-Verlag, Berlin Heidelberg, New York, Tokyo Zincke H, Campbell JT, Utz DC, Farrow GM, Anderson MJ Jr., 1973, Confidence in the negative transrectal needle biopsy. Surg Gynecol Obstet 136:78–80 Rabbani F, Stroumbakis N, Kava BR, Cookson MS, Fair WR, 1998, Incidence and clinical significance of false-negative sextant prostate biopsies. J Urol 159:1247–1250 Eskew LA, Bare RL, McCullough DL, 1997, Systematic 5 region biopsy is superior to sextant method for diagnosing carcinoma of the prostate. J Urol 157:199–202 Durkan GC, Greene DR, 2000, Diagnostic dilemmas in detection of prostate cancer in patients undergoing transrectal ultrasoundguided needle biopsy of the prostate. Pros Canc Pros Dis 3:13–20 Kao J, Upton M, Zhang P, Rosen S, 2002, Individual prostate biopsy core embedding facilitates maximal representation. J Urol 168:496–499 Cserni G, 2004, A model for determining the optimum histology of sentinel lymph nodes in breast cancer. J Clin Pathol 57:467–471 Cserni G, Amendoeira I, Apostolikas N, Bellocq JP, Bianchi S, Bussolati G et al, 2003, Pathological work-up of sentinel lymph nodes in breast cancer. Review of current data to be considered for the formulation of guidelines. Eur J Cancer 39:1654–1667 Lane RB, Lane CG, Mangold KA, Johnson MH, Allsbrook WC, 1998, Needle biopsies of the prostate. What constitutes adequate histologic sampling? Arch Pathol Lab Med 122:833–835 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 97 EP 101 DI 10.1007/s12253-008-9114-0 PG 5 ER PT J AU Cserni, T O’ Donnel, A Paran, S Puri, P AF Cserni, Tamas O’ Donnel, Annemarie Paran, Sri Puri, Prem TI Correlation of Enteric NADPH-d Positive Cell Counts with the Duration of Incubation Period in NADPH-d Histochemistry SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Positive cell counts; Incubation time; NADPH-d histochemistry ID Positive cell counts; Incubation time; NADPH-d histochemistry AB Nicotinamide adenine dinucleotide phosphatediaphorase (NADPH-d) staining can be used in the enteric nervous system to determine nitrergic neuronal counts, critical in motility disorders such as intestinal neuronal dysplasia and hypoganglionosis. The reported incubation periods of specimens with NADPH-d staining solution has varied from 2 to 24 h. The aim of this study is to investigate the impact of the incubation period on the overall NADPHd positive cell counts in porcine rectal submucosal plexus. The submucosal plexus of rectal specimens from 12-weekold pigs (n=5) were studied. Conventional frozen sections were used to identify nitrergic neurons while whole-mount preparations were used to quantify the effect of prolonged duration of incubation on positively identified ganglion cells with NADPH-d histochemistry. The same submucosal ganglia on the conventional sections, and a minimum of 12 ganglia per whole-mount preparation specimen were photographed sequentially at 2, 6, and 24 h and used to count the number of nitrergic cells per ganglion. The same staining solution was used throughout the experiment. Results were analysed using a one-way ANOVA test. Prolonged incubation with the staining solution revealed new NADPH-d positive cells in the ganglia on the conventional sections. The total number of neurons counted in the 12 adjacent ganglia in the whole-mount specimens was 180±55, the mean neuronal cell per ganglion was 15±8 after 2 h of incubation. This increased to 357±17, and to 29±12 after 6 h (p<0.05). A further increase was observed of 515±19 and 43±17 after 24 h (p<0.05). When the photomicrographs were retrospectively analysed, not even the outline of the neuronal cells that stained with prolonged incubation was evident at the earlier time points. NADPH-d positive cell counts increase in proportion to the duration of incubation in NADPH-d histochemistry. Comparative studies attempting to quantify nitrergic cell counts in dysmotility disorders must take into account the variability in NADPH-d positive cell count associated with prolonged incubation in NADPH-d histochemistry. C1 [Cserni, Tamas] University College Dublin, Our Lady’s Children’s Hospital, Children’s Research CentreDublin, Ireland. [O’ Donnel, Annemarie] University College Dublin, Our Lady’s Children’s Hospital, Children’s Research CentreDublin, Ireland. [Paran, Sri] University College Dublin, Our Lady’s Children’s Hospital, Children’s Research CentreDublin, Ireland. [Puri, Prem] University College Dublin, Our Lady’s Children’s Hospital, Children’s Research CentreDublin, Ireland. RP Cserni, T (reprint author), University College Dublin, Our Lady’s Children’s Hospital, Children’s Research Centre, Dublin, Ireland. EM tcserni@yahoo.com CR Bult H, Boeckxstaens GE, Pelckmans PA, Jordaens FH, Van Maercke YM, Herman AG, 1990, Nitric oxide as an inhibitory nonadrenergic non-cholinergic neurotransmitter. Nature 345:346–347 Boeckxstaens GE, Pelckmans PA, Bult H, De Man JG, Herman AG, Van Maercke YM, 1990, Non-adrenergic non-cholinergic relaxation mediated by nitric oxide in the canine ileocolonic junction. Eur J Pharmacol 190:239–246 Stark ME, Bauer AJ, Sarr MG, Szurszewski JH, 1993, Nitric oxide mediates inhibitory nerve input in human and canine jejunum. Gastroenterology 104:398–409 Burleigh DE, 1992, Ng-nitro-L-arginine reduces nonadrenergic, noncholinergic relaxations of human gut. Gastroenterology 102:679–683 Hanani M, Louzon V, Udassin R, Freund HR, Karmeli F, Rachmilewitz D, 1995, Nitric oxide-containing nerves in bowel segments of patients with Hirschsprung's disease. EJ Pediatr Surg 30:818–822 Vanderwinden JM, Mailleux P, Schiffmann SN, Vanderhaeghen JJ, De Laet MH, 1992, Nitric oxide synthase activity in infantile hypertrophic pyloric stenosis. N Engl J Med 327:511–515 Hirakawa H, Kobayashi H, O'Briain DS, Puri P, 1995, Absence of NADPH-diaphorase activity in internal anal sphincter, IAS, achalasia. J Pediatr Gastroenterol Nutr 20:54–58 Dawson TM, Bredt DS, Fotuhi M, Hwang PM, Snyder SH, 1991, Nitric oxide synthase and neuronal NADPH diaphorase are identical in brain and peripheral tissues. Proc Natl Acad Sci U S A 88:7797–7801 Puri P, 2000, Newer neuronal markers. In: Holschneider AM, Puri P, eds, Hirschsprung disease and allied disorders. Harwood, Amsterdam, p 275 Holschneider AM, Puri P, 2000, Intestinal neuronal dysplasia in markers. In: Holschneider AM, Puri P, eds, Hirschsprung disease and allied disorders. Harwood, Amsterdam, p 150 Wester T, O'Briain DS, Puri P, 1999, Notable postnatal alterations in the myenteric plexus of normal human bowel. Gut 44:666–674 Montedonico S, Sri Paran T, Pirker M, Rolle U, Puri P, 2006, Developmental changes in submucosal nitrergic neurons in the porcine distal colon. J Pediatr Surg 41:1029–1035 Scherer-Singler U, Vincent SR, Kimura H, McGeer EG, 1983, Demonstration of a unique population of neurons with NADPHdiaphorase histochemistry. J Neurosci Methods 9:229–234 Timmermans JP, Barbiers M, Scheuermann DW, Stach W, Adriaensen D, Mayer B, De Groodt-Lasseel MH, 1994, Distribution pattern, neurochemical features and projections of nitrergic neurons in the pig small intestine. Ann Anat 176:515– 525 Kuo H, Grant S, Muth N, Hengemihle J, Ingram DK, 1994, The correlation between neuron counts and optical density of NADPH-diaphorase histochemistry in the rat striatum: a quantitative study. Brain Res 660:57–65 Koike S, Hisa Y, Uno T, Murakami Y, Tamada Y, Ibata Y, 1998, Nitric oxide synthase and NADPH-diaphorase in neurons of the rat, dog and guinea pig nodose ganglia. Acta Otolaryngol Suppl 539:110–112 Pitkanen A, Amaral DG, 1991, Distribution of reduced nicotinamide adenine dinucleotide phosphate diaphorase, NADPH-d, cells and fibers in the monkey amygdaloid complex. J Comp Neurol 313:326–348 Wilhelm M, Batori Z, Pasztor I, Gabriel R, 1998, NADPHdiaphorase positive myenteric neurons in the ileum of guinea-pig, rat, rabbit and cat: a comparative study. Eur J Morphol 36:143– 152 Arbones ML, Ribera J, Agullo L, Baltrons MA, Casanovas A, Riveros-Moreno V, Garcia A, 1996, Characteristics of nitric oxide synthase type I of rat cerebellar astrocytes. Glia 18:224–232 Balemba OB, Mortensen K, Semuguruka WD, Hay-Schmidt A, Johansen MV, Dantzer V, 2002, Neuronal nitric oxide synthase activity is increased during granulomatous inflammation in the colon and caecum of pigs infected with Schistosoma japonicum. Auton Neurosci 99:1–12 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 103 EP 107 DI 10.1007/s12253-008-9081-5 PG 5 ER PT J AU Chuangui, Ch Sun, J Liu, G Chen, J AF Chuangui, Chen Sun, Jinjin Liu, Geng Chen, Jianqiu TI Effect of Small Interference RNA Targeting HIF-1α Mediated by rAAV Combined L-Ascorbate on Pancreatic Tumors in Athymic Mice SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Recombinant adeno-associated virus (rAAV); Hypoxia inducible factor (HIF); Small interference RNA (siRNA); L-Ascorbate ID Recombinant adeno-associated virus (rAAV); Hypoxia inducible factor (HIF); Small interference RNA (siRNA); L-Ascorbate AB To study the effect of recombinant adenoassociated virus (rAAV) vector bearing small inference RNA (siRNA) targeting hypoxia inducible factor 1α (HIF-1α) combined L-ascorbate on pancreatic tumors in athymic mice primarily. A cassette encoding siRNA targeting HIF-1α mediated by rAAV was constructed, giving rAAV-siHIF. In vitro, rAAV-hrGFP, rAAV-siHIF and L-ascorbate which were used alone or in combination were delivered to exponentially growing MiaPaCa2 cells. Then, we examined the expression of HIF-1α mRNA and protein, the secretion of VEGF in MiaPaCa2 cells under hypoxic condition with Real-time PCR, Western Blot, ELISA, respectively. In vivo, MiaPaCa2 cells were inoculated subcutaneously on the back of nude mice. Nude mice with xenograft tumor were randomly divided into equal groups and were injected with rAAV-hrGFP or rAAV-siHIF or were fed with L-ascorbate. Then, we measured the size of tumor every 3 days and drew a tumor growth curve. After 30 days, all mice were sacrificed and the tumors were dissected. At last, we examined the expression of HIF-1α, VEGF and CD34 by immunohistochemistry and counted micro-vessel density (MVD). In vitro, we found that rAAV-siHIF could inhibit the expression of HIF-1α mRNA and protein in MiaPaCa2 human pancreatic cancer cells but L-ascorbate could only restrain the expression of HIF-1α protein. Moreover, rAAVsiHIF and L-ascorbate could all inhibit the secretion of vascular VEGF. In vivo, we found that rAAV-siHIF could inhibit the growth of nude mice xenograft tumor and the expression of HIF-1α and VEGF and MVD while Lascorbate can only inhibit the growth of xenograft tumor in the early and middle stage. These results suggest that rAAVsiHIF and L-ascorbate can inhibit the growth of nude mice xenograft tumor and HIF-1α could be a target of pancreatic cancer genetic and pharmacological therapy. C1 [Chuangui, Chen] The Second Hospital of TianJin Medical University, Department of Surgery, 300211 Tianjin, China. [Sun, Jinjin] The Second Hospital of TianJin Medical University, Department of Surgery, 300211 Tianjin, China. [Liu, Geng] The Second Hospital of TianJin Medical University, Department of Surgery, 300211 Tianjin, China. [Chen, Jianqiu] The Second Hospital of TianJin Medical University, Department of Surgery, 300211 Tianjin, China. RP Chen, J (reprint author), The Second Hospital of TianJin Medical University, Department of Surgery, 300211 Tianjin, China. EM chenchen20071011@yahoo.com.cn CR Yin T, Wang C, Liu T et al, 2007, Expression of snail in pancreatic cancer promotes metastasis and chemoresistance. J Surg Res 141:196–203 Brizel DM, Scully SP, Harrelson JM et al, 1996, Tumor oxygenation predicts for the likelihood of distant metastasis in human soft tissue sarcoma. Cancer Res 56:941–943 Semenza GL, 2003, Targeting HIF-1 for cancer therapy. Nature Rev 3:721–732 Mazure NM, Brahimi-Horn MC, Berta MA et al, 2004, HIF-1: master and commander of the hypoxic world. A pharmacological approach to its regulation by siRNAs. Biochem Pharmacol 68:971–980 Frelin C, Ladoux A, D’angelo G, 2000, Vascular endothelial growth factors and angiogenesis. Ann Endocrinol 61:70–74 Forsythe JA, Jiang BH, Iyer NV et al, 1996, Activation of vascular endothelial growth factor gene transcription by hypoxiainducible factor 1. Mol Cell Biol 16:4604–4613 Sivridis E, Giatromanolaki A, Gatter KC et al, 2002, Association of hypoxia-inducible factors 1alpha and 2alpha with activated angiogenic pathways and prognosis in patients with endometrial carcinoma. Cancer 95:1055–1063 Yihai C, 1999, Therapeutic potentials of angiostatin in the treatment of cancer. Haematologica 84:643–650 Zhong H, De Marzo AM, Laughner E et al, 1999, Overexpression of hypoxia-inducible factor-1a in common human cancers and their metastases. Cancer Res 59:5830–5835 Masson N, Willam C, Maxwell PH et al, 2001, Independent function of two destruction domains in hypoxia-inducible factoralpha chains activated by prolyl hydroxylation. EMBO J 20:5197– 5206 Knowles HJ, Raval RR, Harris AL et al, 2003, Effect of ascorbate on the activity of hypoxia-inducible factor in cancer cells. Cancer Res 63:1764–1768 Tomar RS, Matta H, Chaudhary PM, 2003, Use of adenoassociated viral vector for delivery of small interfering RNA. Oncogene 22:5712–5715 Stilwell JL, Samulski RJ, 2003, Adeno-associated virus vectors for therapeutic gene transfer. Biotechniques 34:148–150 Zamore PD, 2001, RNA interference: listening to the sound of silence. Nat Struct Boil 8:746–751 Wu ZJ,Wu XB, Cao H et al, 2001, A novel method for purification of recombinant adeno-associated virus vectors in large-scale. Chin Sci Bull 31:423–430 Weidner N, Semple JP, Welch WR et al, 1991, Tumor angiogenesis andmetastasis: correlation in invasive breast carcinoma. New Eng J Med 324:1–8 Vaupel P, Mayer A, 2007, Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev 26:225–239 Semenza GL, 2000, HIF-1: mediator of physiological and pathophysiological responses to hypoxia. J Appl Physiol 88:1474–1480 Wang F, Li SS, Segersvard R, Strommer L et al, 2007, Hypoxia inducible factor-1 mediates effects of insulin on pancreatic cancer cells and disturbs host energy homeostasis. Am J Pathol 170:469– 477 Lu H, Forbes RA, Verma A, 2002, Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg effect in carcinogenesis. J Bio Chem 26:23111–23115 Buchler P, Reber HA, Buchler M et al, 2003, Hypoxia-inducible factor 1alpha regulates vascular endothelial growth factor expression in human pancreatic cancer. Pancreas 26:55–64 Andre T, Chastre E, Kotelevets L et al, 1998, Tumoral angiogenesis: physiopathology, prognostic value and therapeutic perspectives. Rev Med Interne 19:904–913 Folkman J, 2002, Role of angiogenesis in tumor growth and metastasis. Semin Oncol 29:15–18 Yang ZF, Poon RT, To J et al, 2004, The potential role of hypoxia inducible factor 1 alpha in tumor progression after hypoxia and chemotherapy in hepatocellular carcinoma. Cancer Res 64:5496– 5503 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 109 EP 114 DI 10.1007/s12253-008-9063-7 PG 6 ER PT J AU Perse, M Cerar, A Injac, R Strukelj, B AF Perse, Martina Cerar, Anton Injac, Rade Strukelj, Borut TI N-methylnitrosourea Induced Breast Cancer in Rat, the Histopathology of the Resulting Tumours and its Drawbacks as a Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Animal model; Breast cancer; Histopathology; N-methylnitrosourea; Rats ID Animal model; Breast cancer; Histopathology; N-methylnitrosourea; Rats AB Several animal models of breast cancer have been developed to study various aspects of breast cancer biology. Substantial evidence suggests that the N-methylnitrosourea (MNU) animal model mimics human breast cancer in many respects. It has therefore been used extensively to evaluate preventive and therapeutic agents for human breast cancer. Chemically induced rodent models are also suitable for studying malignant progression. Recently, Liska et al. [7] established two protocols of MNU administration depending on the animal’s age and number of applications of carcinogen, with the aimof investigating the advanced stages of mammary gland tumours. We used the same protocol as Liska but have obtained substantially different results. These results are presented and discussed in the frame of suggested key drawbacks of the MNU induced breast cancer rat model, as a contribution to the debate about the suitability of that model for evaluating preventive and therapeutic agents. C1 [Perse, Martina] University of Ljubljana, Faculty of Medicine, Institute of Pathology, Korytkova 2, 1105 Ljubljana, Slovenia. [Cerar, Anton] University of Ljubljana, Faculty of Medicine, Institute of Pathology, Korytkova 2, 1105 Ljubljana, Slovenia. [Injac, Rade] University of Ljubljana, Faculty of Pharmacy, Institute of Pharmaceutical Biology, Askerceva 7, 1000 Ljubljana, Slovenia. [Strukelj, Borut] University of Ljubljana, Faculty of Pharmacy, Institute of Pharmaceutical Biology, Askerceva 7, 1000 Ljubljana, Slovenia. RP Perse, M (reprint author), University of Ljubljana, Faculty of Medicine, Institute of Pathology, 1105 Ljubljana, Slovenia. EM martina.perse@mf.uni-lj.si CR Parkin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Clarke R, 1996, Animal models of breast cancer: their diversity and role in biomedical research. Breast Cancer Res Treat 39:1–6 Russo J, Russo IH, 1996, Experimentally induced mammary tumors in rats. Breast Cancer Res Treat 39:7–20 McCormick DL, Adamowski CB, Fiks A et al, 1981, Lifetime dose-response relationships for mammary tumor induction by a single administration of N-methyl-N-nitrosourea. Cancer Res 41:1690–1694 Isaacs JT, 1986, Genetic control of resistance to chemically induced mammary adenocarcinogenesis in the rat. Cancer Res 46:3958–3963 Thompson HJ, Adlakha H, 1991, Dose-responsive induction of mammary gland carcinomas by the intraperitoneal injection of 1- methyl-1-nitrosourea. Cancer Res 51:3411–3415 Liska J, Galbavy S, Macejova D et al, 2000, Histopathology of mammary tumours in female rats treated with 1-methyl-1- nitrosourea. Endocr Regul 34:91–96 Thompson HJ, Adlakha H, Singh M, 1992, Effect of carcinogen dose and age at administration on induction of mammary carcinogenesis by 1-methyl-1-nitrosourea. Carcinogenesis 13:1535–1539 Thompson HJ, McGinley JN, Rothhammer K et al, 1995, Rapid induction of mammary intraductal proliferations, ductal carcinoma in situ and carcinomas by the injection of sexually immature female rats with 1-methyl-1-nitrosourea. Carcinogenesis 16:2407–2411 Russo J, Russo IH, 2000, Atlas and histologic classification of tumors of the rat mammary gland. Journal of Mammary Gland Biology and Neoplasia 5:187–200 Chan MM, Lu X, Merchant FM et al, 2005, Gene expression profiling of NMU-induced rat mammary tumors: cross species comparison with human breast cancer. Carcinogenesis 26:1343– 1353 Badawi AF, Eldeen MB, Liu Y et al, 2004, Inhibition of rat mammary gland carcinogenesis by simultaneous targeting of cyclooxygenase-2 and peroxisome proliferator-activated receptor gamma. Cancer Res 64:1181–1189 Kubatka P, Ahlers I, Ahlersova E et al, 2003, Chemoprevention of mammary carcinogenesis in female rats by rofecoxib. Cancer Lett 202:131–136 Mehta RG, 2000, Experimental basis for the prevention of breast cancer. Eur J Cancer 36:1275–1282 Roomi MW, Roomi NW, Ivanov V et al, 2005, Modulation of Nmethyl- N-nitrosourea induced mammary tumors in Sprague– Dawley rats by combination of lysine, proline, arginine, ascorbic acid and green tea extract. Breast Cancer Res 7:R291–R295 Rivera ES, Andrade N, Martin G et al, 1994, Induction of mammary tumors in rat by intraperitoneal injection of NMU: histopathology and estral cycle influence. Cancer Lett 86:223–228 Gullino PM, Pettigrew HM, Grantham FH, 1975, N-nitrosomethylurea as mammary gland carcinogen in rats. J Natl Cancer Inst 54:401–414 Loscher W, Mevissen M, Haussler B, 1997, Seasonal influence on 7, 12-dimethylbenz[a]anthracene-induced mammary carcinogenesis in Sprague–Dawley rats under controlled laboratory conditions. Pharmacol Toxicol 81:265–270 Sumova A, Bendova Z, Sladek M et al, 2004, Seasonal molecular timekeeping within the rat circadian clock. Physiol Res 53 Suppl 1:S167–S176 De Jonage-Canonico MB, Lenoir V, Martin A et al, 2003, Long term inhibition by estradiol or progesterone of melatonin secretion after administration of a mammary carcinogen, the dimethyl benz, a)anthracene, in Sprague–Dawley female rat; inhibitory effect of Melatonin on mammary carcinogenesis. Breast Cancer Res Treat 79:365–377 Halberg F, 1964, Grundlagenforschung zur Atiologie des Karzinoms. Arztl Fortb 2:67–77 Kubatka P,Ahlersova E, Ahlers I et al, 2002)Variability of mammary carcinogenesis induction in female Sprague–Dawley andWistar:Han rats: the effect of season and age. Physiol Res 51:633–640 Lu J, Jiang C, Mitrenga T et al, 1998, Pathogenic characterization of 1-methyl-1-nitrosourea-induced mammary carcinomas in the rat. Carcinogenesis 19:223–227 Reznik G, Ward JM, 1981, Morphology of neoplastic lesions in the clitoral and prepucial gland of the F334 rat. J Cancer Res Clin Oncol 101:249–263 Eustis SL, Haseman JK, Mackenzie WF et al, 1995, Toxicity and carcinogenicity of 2, 3-dibromo-1-propanol in F344/N rats and B6C3F1 mice. Fundam Appl Toxicol 26:41–50 Ito O, Okamoto T, Fujimoto N et al, 1994, Inhibition of mammary tumours by pretreatment with 17 beta-estradiol in F344 rats induced with N-methyl-N-nitrosourea. Jpn J Cancer Res 85:279–283 Workman P, Balmain A, Hickman JA et al, 1988, UKCCCR guidelines for the welfare of animals in experimental neoplasia. Lab Anim 22:195–201 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 115 EP 121 DI 10.1007/s12253-008-9117-x PG 7 ER PT J AU Anagnostou, V Tiniakos, D Chorti, M Kiagia, M Tourkantonis, I Alamara, Ch Syrigos, NK AF Anagnostou, K. Vasiliki Tiniakos, G. Diana Chorti, Maria Kiagia, Maria Tourkantonis, Ioannis Alamara, Christina Syrigos, Nik Konstantinos TI Right Sited Renal Cell Carcinoma Metastasizing to the Contralateral Ovary: Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Renal cell carcinoma; Ovarian metastasis ID Renal cell carcinoma; Ovarian metastasis AB Ovarian metastases from renal cell carcinoma are rare, with only 22 cases reported in the literature. We report a case of a 45-year-old woman, who developed left ovarian and right adrenal metastases 3 months after diagnosis of clear cell renal cell carcinoma and review the literature. This is the fourth reported case of right renal cell carcinoma metastasizing to the left ovary. The patient is alive 4 years after resection of the ovarian tumor, treated with sunitinib. We conclude that, although rare, metastatic renal cell carcinoma should be included in the differential diagnosis of ovarian tumors with clear cell histology. C1 [Anagnostou, K. Vasiliki] University of Athens, 3rd Department of Medicine, Oncology Unit, Building Z, Sotiria General Hospital, Mesogion 152, 115 27 Athens, Greece. [Tiniakos, G. Diana] University of Athens, School of Medicine, Laboratory of Histology & EmbryologyAthens, Greece. [Chorti, Maria] Sismanoglion Hospital, Department of PathologyAthens, Greece. [Kiagia, Maria] University of Athens, 3rd Department of Medicine, Oncology Unit, Building Z, Sotiria General Hospital, Mesogion 152, 115 27 Athens, Greece. [Tourkantonis, Ioannis] University of Athens, 3rd Department of Medicine, Oncology Unit, Building Z, Sotiria General Hospital, Mesogion 152, 115 27 Athens, Greece. [Alamara, Christina] University of Athens, 3rd Department of Medicine, Oncology Unit, Building Z, Sotiria General Hospital, Mesogion 152, 115 27 Athens, Greece. [Syrigos, Nik Konstantinos] University of Athens, 3rd Department of Medicine, Oncology Unit, Building Z, Sotiria General Hospital, Mesogion 152, 115 27 Athens, Greece. RP Anagnostou, V (reprint author), University of Athens, 3rd Department of Medicine, Oncology Unit, 115 27 Athens, Greece. EM valsamo.anangostou@yale.edu CR Lineham M, Zbar B, Bates S et al, 2005, Cancer principals and practice of oncology: cancer of the kidney and ureter, 7th edn. Lippincott, New York, pp 1139–1151 Scully RE, Young RH, Clement PB, 1998, Atlas of tumor pathology: tumors of the ovary, maldeveloped gonads, fallopian tube, and broad ligament. Armed Forces Institute of Pathology, Washington, D.C., pp 141–151, 153, 160, 352, 353 Insabato L, De Rosa G, Franco R et al, 2003, Ovarian metastasis from renal cell carcinoma: a report of three cases. Int J Surg Pathol 11:309–312 Johansson H, 1960, Clinical aspects of metastatic ovarian cancer of extragenital origin. Acta Obstet Gynecol Scand 39:681–697 Kato Y, Numata A, Wada N et al, 2006, A case of metastatic renal cell carcinoma to the ovary. Hinyokika Kiyo 52:923–927 Liu FS, Ho ES,Lu F,Chang CY, 1992, Solitary metastasis of renal cell carcinoma to the ovaries: a case report. Zhonghua Yi Xue Za Zhi, Taipei, 50:165–168 Madersbacher S, Ponholzer A, Franz K et al, 2007, Synchronous bilateral renal cell cancer with a single ovarian metastasis and a fibromuscular dysplasia of the renal artery. Aktuelle Urol 38:52–54 Shinojima T, Nakajima Y, Kiguchi H, 2001, Renal cell carcinoma metastatic to the ovary: a case report. Nippon Hinyokika Gakkai Zasshi 92:694–697 Spencer JR, Eriksen B, Garnett JE, 1993, Metastatic renal tumor presenting as ovarian clear cell carcinoma. Urology 41:582–584 Stadiem ML, 1937, An ovarian hypernephroma. Am J Surg 37:312–318 Stefani P, Selli C, Nicita G et al, 1981, Angiographic aspects of renal cell carcinoma metastatic to the female genitalia. Cardiovasc Interv Radiol 4:183–186 Valappil SV, Toon PG, Anandaram PS, 2004, Ovarian metastasis from primary renal cell carcinoma: report of a case and review of literature. Gynecol Oncol 94:846–849 Young RH, Hart WR, 1992, Renal Cell Carcinoma metastatic to the ovary: a report of three cases emphasizing possible confusion with ovarian clear cell ademocarcinoma. Int J Gynec Pathol 11:96–104 Baker PM, Oliva E, 2004, Immunohistochemistry as a tool in the differential diagnosis of ovarian tumors: an update. Int J Gynecol Pathol 24:39–55 Nolan LP, Heatley MK, 2001, The value of immunohistochemistry in distinguishing between clear cell carcinoma of the kidney and the ovary. Int J Gynecol Pathol 20:155–159 Skinnider BF, Folpe AL, Hennigar RA, Lim SD et al, 2005, Distribution of cytokeratins and vimentin in adult renal neoplasms and normal renal tissue. Potential utility of a cytokeratin antibody panel in the differential diagnosis of renal tumors. Am J Surg Pathol 29:747–754 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 123 EP 127 DI 10.1007/s12253-008-9039-7 PG 5 ER PT J AU Burkadze, G Turashvili, G AF Burkadze, George Turashvili, Gulisa TI A Case of Osteoclast-like Giant Cell Tumor of the Pancreas Associated with Borderline Mucinous Cystic Neoplasm SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteoclast-like giant cell tumor; Mucinous cystic neoplasm; Immunohistochemistry; Pancreas ID Osteoclast-like giant cell tumor; Mucinous cystic neoplasm; Immunohistochemistry; Pancreas AB A 34-year-old, previously healthy female presented with severe acute upper quadrant abdominal pain and an 11-cm cystic mass in the tail of the pancreas. The patient underwent distal pancreatectomy with total gross excision of the mass. Grossly, the mass consisted of a multiloculated cystic lesion measuring 11.7 cm in its greatest dimension. An irregular solid lobulation at the lateral aspect of the cyst was visible, measuring 3 cm in the largest dimension. Histologically, there were two distinct components: a mucinous, neoplastic epithelial cyst with few foci of moderate atypia, and nodular spindle cell areas containing multinucleated tumor giant cells. Immunohistochemically, the multinucleated giant cells were positive for vimentin, CD68 and CD45, and negative for cytokeratin and epithelial membrane antigen (EMA). The spindle cells of hypercellular stroma were stained for vimentin, but not for EMA or carcinoembryonic antigen (CEA). Neuronspecific enolase (NSE), S100 and Ki-67 showed no reactivity. The histological diagnosis "osteoclast-like giant cell tumor of the pancreas associated with borderline mucinous cystic neoplasm" was made. The patient recovered and is free of disease 4 years after the diagnosis. C1 [Burkadze, George] N. Kipshidze Central University Clinic, Department of Pathology, Vazha-Pshavela Avenue, 0160 Tbilisi, Georgia. [Turashvili, Gulisa] N. Kipshidze Central University Clinic, Department of Pathology, Vazha-Pshavela Avenue, 0160 Tbilisi, Georgia. RP Burkadze, G (reprint author), N. Kipshidze Central University Clinic, Department of Pathology, 0160 Tbilisi, Georgia. EM gburkadze@hotmail.com CR Alguacil-Garcia A, Weiland L, 1977, The histologic spectrum, prognosis, and histogenesis of the sarcomatoid carcinoma of the pancreas. Cancer 39:1181–1189 Batsakis J, Ordonez N, Sevidal P, Baker JR, 1988, Osteoclasttype giant cell neoplasms of the parotid gland. J Laryngol Otol 102:901–904 Dizon M, Multhaulpt H, Paskin D, Warhol MJ, 1996, Osteoclastic giant cell tumor of the pancreas: an immunohistochemical study. Arch Pathol Lab Med 20:306–309 Dworak O, Wittekind C, Koerfgen H, Gall F, 1993, Osteoclastic giant cell tumor of the pancreas. An immunohistochemical study and review of the literature. Pathol Res Pract 198:228–231 Gatteschi B, Saccomanno S, Bartoli F, Salvi S, Liu G, Pugliese V, 1995, Mixed pleomorphic osteoclast-like tumor of the pancreas. Light microscopical, immunohistochemical, and molecular biological studies. Int J Pancreatol 18:169–175 Goldberg R, Michelassi F, Montag A, 1991, Osteoclast-like giant cell tumor of the pancreas: immunophenotypic similarity to giant cell tumor of bone. Hum Pathol 22:618–22 Heller K, Manyak M, Tabbara S, Kerr L, 1998, Malignant osteoclast-like giant cell tumor of the kidney. Urology 51:495–498 Klimstra D, Rosai J, 1993, Critical commentary to “Osteoclastic giant cell tumor of the pancreas”. Pathol Res Pract 198:232–233 Kloppel G, 1984, Pancreatic, Non-endocrine Tumours, in Pancreatic Pathology. Churchill Livingstone, London Leighton CC, Shum DT, 2001, Osteoclastic giant cell tumor of the pancreas. Am J Clin Oncol 24:77–80 Machado MAC, Herman P, Montagnini AL, Jukemura J, Leite KRM, Machado MCC, 2001, Benign variant of osteoclast-type giant cell tumor of the pancreas: importance of the lack of epithelial differentiation. Pancreas 22:105–107 Mentes A, Yuce G, 1993, Osteoclast-type giant cell tumor of the pancreas associated with mucinous cystadenoma. Eur J Surg Oncol 19:84–86 Molberg K, Heffes C, Delgado R, Albores-Saavedra J, 1998, Undifferentiated carcinoma with osteoclast-like giant cells of the pancreas and periampullary region. Cancer 82:1279–1287 Oehler U, Jurs M, Kloppel G, Helpap B, 1997, Osteoclast-like giant cell tumor of the pancreas presenting as a pseudocyst-like lesion. Virchows Arch 431:215–218 Rosai J, 1968, Carcinoma of pancreas simulating giant cell tumor of bone: electron-microscopic evidence of its acinar cell origin. Cancer 22:333–334 Shabb N, Tawil A, Mufarrij A, Obeid S, Halabi J, 1997, Mammary carcinoma with osteoclast-like giant cells cytologically mimicking benign breast disease. A case report. Acta Cytol 41:1284–1288 Shiozawa M, Imada T, Ishiwa N, Rino Y, Hasuo K, Takanashi Y, Nakatani Y, Inayama Y, 2002, Osteoclast-like giant cell tumor of the pancreas. Int J Clin Oncol 7:376–380 Sun A, Ohtsuki Y, Liang S, Sonobe H, Iwata J, FurihataM, Takeuchi T, Qiu Y, Chen B, Watanabe R, Ohmori K, 1998, Osteoclast-like giant cell tumor of the pancreas with metastases to gallbladder and lymph nodes. A case report. Pathol Res Pract 194:587–594 Weidner N, Cote R, Suster S, Weiss L, 2003, Modern Surgical Pathology. Saunders, Philadelphia NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 129 EP 131 DI 10.1007/s12253-008-9053-9 PG 3 ER PT J AU Sterlacci, W Veits, L Moser, P Steiner, HJ Ruscher, S Jamnig, H Mikuz, G AF Sterlacci, William Veits, Lothar Moser, Patrizia Steiner, Hans-Jorg Ruscher, Sighard Jamnig, Herbert Mikuz, Gregor TI Idiopathic Systemic Amyloidosis Primarily Affecting the Lungs with Fatal Pulmonary Haemorrhage due to Vascular Involvement SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Amyloidosis; Haemorrhage; Pulmonary ID Amyloidosis; Haemorrhage; Pulmonary AB A patient who presented with dyspnea and suspected interstitial pulmonary fibrosis suffered a fatal pulmonary haemorrhage with no feasible cause for bleeding. Autopsy revealed abundant amyloid deposits in both lungs with a diffuse alveolar septal distribution pattern. Amyloid was also found in the cardiac interstitium and in many vessel walls. Considering the affected organs and the histological characteristics, the deposits were regarded as light chain-type. Amyloidosis, which is generally an uncommon disease, very rarely affects the lung predominantly. Haemorrhagic diathesis is a known complication in amyloidosis patients, although fatal haemorrhage is rare and has not yet been reported solely of pulmonary origin. This report describes an uncommon case of idiopathic systemic amyloidosis mainly manifesting in the lungs. The diagnosis was established after fatal pulmonary haemorrhage caused by vessel impairment due to additional vascular amyloid deposits. C1 [Sterlacci, William] Medical University of Innsbruck, Department of Pathology, Mullerstrasse 44, 6020 Innsbruck, Austria. [Veits, Lothar] Medical University of Innsbruck, Department of Pathology, Mullerstrasse 44, 6020 Innsbruck, Austria. [Moser, Patrizia] Medical University of Innsbruck, Department of Pathology, Mullerstrasse 44, 6020 Innsbruck, Austria. [Steiner, Hans-Jorg] Medical University of Innsbruck, Department of Pathology, Mullerstrasse 44, 6020 Innsbruck, Austria. [Ruscher, Sighard] Hospital Natters, Department for Pneumonology, In der Stille 20, 6161 Natters, Austria. [Jamnig, Herbert] Hospital Natters, Department for Pneumonology, In der Stille 20, 6161 Natters, Austria. [Mikuz, Gregor] Medical University of Innsbruck, Department of Pathology, Mullerstrasse 44, 6020 Innsbruck, Austria. RP Sterlacci, W (reprint author), Medical University of Innsbruck, Department of Pathology, 6020 Innsbruck, Austria. EM william.sterlacci@i-med.ac.at CR Lachmann HJ, Hawkins PN, 2006, Amyloidosis and the lung. Chron Respir Dis 3:203–14 Sanchorawala V, 2006, Light-chain, AL, amyloidosis: diagnosis and treatment. Clin J Am Soc Nephrol 1:1331–1341 Howard ME, Ireton J, Daniels F, Langton D, et al., 2001, Pulmonary presentations of amyloidosis. Respirology 6:61–64 Greipp PR, Kyle RA, Bowie EJ, 1981, Factor-X deficiency in amyloidosis: a critical review. Am J Hematol 11:443–450 Yood RA, Skinner M, Rubinow A et al, 1983, Bleeding manifestations in 100 patients with amyloidosis. JAMA 249: 1322–1324 Mumford AD, O'Donnell J, Gillmore JD et al, 2000, Bleeding symptoms and coagulation abnormalities in 337 patients with ALamyloidosis. Br J Haematol 110:454–460 Wright JR, Calkins E, Humphrey RL, 1977, Potassium permanganate reaction in amyloidosis. A histologic method to assist in differentiating forms of this disease. Lab Invest 6:274–281 Browning MJ, Banks RA, Tribe CR et al, 1985, Ten years’ experience of an amyloid clinic–a clinicopathological survey. Q J Med 54:213–227 Gillmore JD, Hawkins PN, 1999, Amyloidosis and the respiratory tract. Thorax 54:444–451 Berk JL, Keane J, Seldin DC et al, 2003, Persistent pleural effusions in primary systemic amyloidosis: etiology and prognosis. Chest 124:969–977 Lee HM, Naor J, DeAngelis D et al, 2000, Primary localized conjunctival amyloidosis presenting with recurrence of subconjunctival hemorrhage. Am J Ophthalmol 129:245–247 Rapoport M, Yona R, Kaufman S et al, 1994, Unusual bleeding manifestations of amyloidosis in patients with multiple myeloma. Clin Lab Haematol 16:349–353 Alwitry A, Brackenbury ET, Beggs FD et al, 2001, Vascular amyloidosis causing spontaneous mediastinal haemorrhage with haemothorax. Eur J Cardiothorac Surg 20:871–873 Shaheen NA, Salman SD, Nassar VH, 1975, Fatal bronchopulmonary hemorrhage due to unrecognized amyloidosis. Arch Otolaryngol 101:259–261 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 133 EP 136 DI 10.1007/s12253-008-9066-4 PG 4 ER PT J AU Rekhi, B Kumar, R Menon, S Medhi, S Desai, BS AF Rekhi, Bharat Kumar, Rajiv Menon, Santosh Medhi, Seema Desai, B Sangeeta TI Calvarial Metastasis of a Renal Cell Carcinoma, Mimicking a Primary Alveolar Soft Part Sarcoma, in a Young Girl—a Rare Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Calvarial; Skull metastases; Renal cell carcinoma; Alveolar soft part sarcoma; CD10 staining ID Calvarial; Skull metastases; Renal cell carcinoma; Alveolar soft part sarcoma; CD10 staining AB Renal cell carcinoma (RCC) is characterized by an unpredictable clinical behavior. It has a tendency for early metastasis, which, at times is the initial presentation and therein poses a diagnostic challenge. We present a rare case of a disseminated RCC in a 15-year-old girl, who primarily presented with an occipital soft tissue mass. Computed tomography (CT) of the head revealed a soft tissue mass in the scalp, eroding the occipital bone and extending intracranially. Biopsy examination showed overlapping features of an alveolar soft part sarcoma (ASPS) and a RCC. Immunohistochemistry (IHC) showed diffuse positivity for CD10 and focal positivity for vimentin. Cytokeratin (CK) and epithelial membrane antigen (EMA) were negative. The patient was recommended a clinical ‘work-up’ to rule out a possible primary in the kidneys. Her CT scan abdomen unraveled a large, lobulated, heterogeneous cystic mass, involving the middle and upper pole of the left kidney. Diagnosis of a metastatic RCC was ascertained. The present case represents a rare manifestation of a RCC metastasizing at an unusual location i.e. calvarium in the youngest patient known, so far and masquerading a primary ASPS. It also highlights the value of clinico-patho-radiological correlation, including CD10 as a useful IHC marker in diagnosing a RCC in young patients, especially when histopathological features overlap with ASPS. C1 [Rekhi, Bharat] Tata Memorial Hospital, Department of Pathology, Dr Ernest Borges Road, Parel, 400012 Mumbai, India. [Kumar, Rajiv] Tata Memorial Hospital, Department of Pathology, Dr Ernest Borges Road, Parel, 400012 Mumbai, India. [Menon, Santosh] Tata Memorial Hospital, Department of Pathology, Dr Ernest Borges Road, Parel, 400012 Mumbai, India. [Medhi, Seema] Tata Memorial Hospital, Department of RadiodiagnosisMumbai, India. [Desai, B Sangeeta] Tata Memorial Hospital, Department of Pathology, Dr Ernest Borges Road, Parel, 400012 Mumbai, India. RP Rekhi, B (reprint author), Tata Memorial Hospital, Department of Pathology, 400012 Mumbai, India. EM rekhi.bharat@gmail.com CR Pantuck AJ, Zisman A, Belldegrun AS, 2001, The changing natural history of renal cell carcinoma. J Urol 166:1611– 1623 Bruder E, Passera O, Harms D, Leuschner I, Ladanyi M, Argani P, Eble JN, Struckmann K, Schraml P, Moch H, 2004, Morphologic and molecular characterization of renal cell carcinoma in children and young adults. Am J Surg Pathol 28:1117–1132 Renshaw AA, Granter SR, Fletcher JA, Kozakewich HP, Corless CL, Perez-Atayde AR, 1999, Renal cell carcinomas in children and young adults: increased incidence of papillary architecture and unique subtypes. Am J Surg Pathol 23:795–802 Sunita, Kapila K, Singhal RM, Verma K, 1991, Fine needle aspiration diagnosis of an unusual presentation of metastatic renal cell carcinoma, letter). Acta Cytol 35:260–261 Wahner-Roedler L, Sebo T, 1997, Renal cell carcinoma; diagnosis based on metastatic manifestations, case report). Mayo Clin Proc 72:935–941 Koutnouyan HA, Rumore GJ, Kahn JM, 1998, Skull metastasis from renal cell carcinoma. Case report and literature review. Ann Otol Rhinol Laryngol 107:598–602 Gaetani P, Di Ieva A, Colombo P, Tancioni F, Aimar E, Debernardi A, Baena RR, 2005, Calvarial metastases as clinical presentation of renal cell carcinoma: report of two cases and review of the literature. Clin Neurol Neurosurg 107:329–333 De Vos C, Gerard JM, Decat M, Gersdorff M, 2005, Metastatic renal cell carcinoma to the temporal bone: case report. B-ENT 1:43–46 Yeh HC, Yang SF, Ke HL, Lee KS, Huang CH, Wu WJ, 2007, Renal cell carcinoma presenting with skull metastasis: a case report and literature review. Kaohsiung J Med Sci 23:475–479 Forbes GS, McLeod RA, Hattery RR, 1977, Radiographic manifestations of bone metastases from renal carcinoma. AJR Am J Roentgenol 129:61–66 Avery AK, Beckstead J, Renshaw AA, Corless CL, 2000, Use of antibodies to RCC and CD10 in the differential diagnosis of renal neoplasms. Am J Surg Pathol 24(2):203–210 Argani P, Antonescu CR, Illei PB et al, 2001, Primary renal neoplasms with the ASPL-TFE3 gene fusion of alveolar soft part sarcoma: a distinctive tumor entity previously included among renal cell carcinomas of children and adolescents. Am J Pathol 159:179–192 Rackley R, Novick A, Klein E, Bukowski R, McLain D, Goldfarb D, 1994, The impact of adjuvant nephrectomy on multi-modality treatment of metastatic renal cell carcinoma. J Urol 152:1399– 1403 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 137 EP 141 DI 10.1007/s12253-008-9097-x PG 5 ER PT J AU Hortobagyi, T Alhakim, A Biedrzycki, O Djurovic, V Rawal, J Al-Sarraj, S AF Hortobagyi, Tibor Alhakim, Ali Biedrzycki, Olaf Djurovic, Vesna Rawal, Jeewan Al-Sarraj, Safa TI Cysticercosis of the Fourth Ventricle Causing Sudden Death: A Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cysticercosis; Neurocysticercosis; Sudden death; Taenia solium ID Cysticercosis; Neurocysticercosis; Sudden death; Taenia solium AB A 15 years old girl of African origin was admitted with a history of headaches and a generalised tonic seizure. Her clinical examination including fundoscopy was normal. She claimed she had been assaulted. Within a few hours of her admission she was found dead in her bed during the ward round. Cardiopulmonary resuscitation was unsuccessful. At post-mortem, the major organs showed no pathological changes and neck dissection showed no abnormality. Neuropathological examination after formalin fixation revealed a cystic lesion in the fourth ventricle, ependymitis and acute hydrocephalus. Histology showed parts of the parasite Taenia solium and the diagnosis was neurocysticercosis. This case highlights the need for forensic and general pathologists as well as forensic medical examiners and paediatricians to be aware of neurocysticercosis as a possible cause of sudden death in the presence of normal clinical findings and negative autopsy, especially in patients from Asian, African or South American countries. As cysticercosis is the commonest cause of seizures in the developing world, neurocysticercosis needs to be considered as a cause of sudden and unexpected death in any patient with a history of headaches and/or seizures. C1 [Hortobagyi, Tibor] King’s College Hospital, Clinical Neuropathology, Denmark Hill, SE5 9RS London, UK. [Alhakim, Ali] East Surrey Hospital, Department of HistopathologySurrey, UK. [Biedrzycki, Olaf] Royal London Hospital, Institute of PathologyLondon, UK. [Djurovic, Vesna] Forensic Pathology ServicesLondon, UK. [Rawal, Jeewan] BHR NHS Trust, Oldchurch HospitalRomford, Essex, UK. [Al-Sarraj, Safa] King’s College Hospital, Clinical Neuropathology, Denmark Hill, SE5 9RS London, UK. RP Al-Sarraj, S (reprint author), King’s College Hospital, Clinical Neuropathology, SE5 9RS London, UK. EM safa.al-sarraj@kch.nhs.uk CR Esberg G, Reske-Nielsen E, 1988, Sudden death from cerebral cysticercosis. Scand J Infect Dis 20:679–684 Bent Hamida M, Moulonguet A, Romano P, Gray F, 1993, Confrontation at the Salpetriere hospital. May 1991 Cephalagia developing in depressive background and sudden death in a 26 year old woman. Rev Neurol, Paris, 149:362–366 Ndhlovu CE, 1997, An uncommon presentation of cysticercosis. Cent Afr J Med 43:207–209 DeGiorgio CM, Houston I, Oviedo S, Sorvillo F, 2002, Deaths associated with cysticercosis. Report of three cases and review of the literature. Neurosurg Focus 12:e2 Verma SK, Agarwal BB, Agarwal G, 1998, Sudden death in neurocysticercosis by trauma. Forensic Sci. Int. 95:23–26 Rao KR, Lessing D, 2003, Neurocysticercosis in West London. Arch Dis Child 88:471 DeGiorgio CM, Medina MT, Duron R, Zee C, Escueta SP, 2004, Neurocysticercosis. Epilepsy Curr 4:107–111 Del Brutto OH, 2005, Neurocysticercosis. Semin Neurol 25:243–251 Rosenfeld E, 2003, Neurocysticercosis. Update Pediatr Infect Dis J 22:181–182 Oeberst JL, Barnard JJ, Bigio EH, Prahlow JA, 2002, Neurocysticercosis. Am J Forensic Med. Pathol, 2002, 23:31–35 Rajshekhar V, Joshi DD, Doanh NQ, van De N, Xiaonong Z, 2003, Taenia solium taeniosis/cysticercosis in Asia: epidemiology, impact and issues. Acta Trop 87:53–60 White AC Jr, 1997, Neurocysticercosis: a major cause of neurological disease worldwide. Clin. Infect. Dis. 24:101–115 Gubbay AD, Brophy BP, Henley S, Sage M, 1998, Neurocysticerosis. J Clin Neurosci 5:203–207 Pitella JEM, 1997, Neurocysticercosis. Brain Pathol 7:681–693 Deb KP, Carpio A, Sander JWAS, 2000, Neurocysticercosis and epilepsy in developing countries. J. Neurol. Neurosurg. Psychiatry 68:137–143 Goodyear M, Voyvodic F, Brophy B, Sage M, 1997, Spinal cysticercosis. J. Clin. Neurosci 4:25–27 De Souza Queiroz L, Filho AP, Callegaro D, De Faria LL, 1975, Intramedullary cysticercosis. Case report, literature review and comments on pathogenesis. J Neurol Sci 26:61–70 Adhisivam B, 2004, Starry sky: multiple neurocysticercosis. Arch Dis Child Educ Pract 89:ep75 Biedrzycki O, Hortobagyi T, Alhakim A, Hunt N, Djurovic V, Al-Sarraj S, 2006, Sudden deaths caused by intraventricular cysts, neuropathol. Appl. Neurobiol 32:240 [Abstract] Jarquin-Valdivia AA, Rich AT, Yarbrough JL, Thompson RC, 2005, Intraventricular colloid cyst, hydrocephalus and neurogenic stunned myocardium. Clin. Neurol. Neurosurg. 107:361–365 Garcia HH, Evans CA, Nash TE, Takayanagui OM, White AC Jr, Botero D, RajshekharV, Tsang VC, Schantz PM, Allan JC, Flisser A, Correa D, Sarti E, Friedland JS,Martinez SM, Gonzalez AE, Gilman RH, Del Brutto OH, 2002, Current consensus guidelines for treatment of neurocysticercosis. Clin Microbiol Rev 15:747–756 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 143 EP 146 DI 10.1007/s12253-008-9098-9 PG 4 ER PT J AU Suranyi, A Bito, T Vajda, Gy Kaiser, L Gaspar, G Katona, M Szabo, J Pal, A AF Suranyi, Andrea Bito, Tamas Vajda, Gyorgy Kaiser, Laszlo Gaspar, Gabor Katona, Marta Szabo, Janos Pal, Attila TI Unusual Clinical History of a Male Infant with Edwards Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Dysgenesis of corpus callosum; Edwards syndrome; Oesophageal atresia; Renal agenesis; Ultrasound ID Dysgenesis of corpus callosum; Edwards syndrome; Oesophageal atresia; Renal agenesis; Ultrasound AB Edwards syndrome (trisomy of chromosome 18) is generally characterized by the disorders of central nervous system, as well as the musculoskeletal and genitourinary systems. In majority of the cases with trisomy 18 the following malformations can be found: ventricular septal defect, horseshoe kidneys, oesophageal atresia, omphalocele, facial clefts, diaphragmatic hernias and genital hypoplasia. We report a male patient with Edwards syndrome. The boy had a partial agenesis of corpus callosum, oesophageal atresia with tracheo-oesophageal fistula, renal agenesis, ventricular septal defect, Dandy-Walker cyst and low-set malformed ears. The first three features are unique based on previous literature reports on trisomy 18. This report allows a further delineation of the trisomy 18 syndrome. C1 [Suranyi, Andrea] University of Szeged, Department of Obstetrics and Gynaecology, Semmelweis u. 1, 6725 Szeged, Hungary. [Bito, Tamas] University of Szeged, Department of Obstetrics and Gynaecology, Semmelweis u. 1, 6725 Szeged, Hungary. [Vajda, Gyorgy] University of Szeged, Department of Obstetrics and Gynaecology, Semmelweis u. 1, 6725 Szeged, Hungary. [Kaiser, Laszlo] University of Szeged, Department of Obstetrics and Gynaecology, Semmelweis u. 1, 6725 Szeged, Hungary. [Gaspar, Gabor] University of Szeged, Department of Obstetrics and Gynaecology, Semmelweis u. 1, 6725 Szeged, Hungary. [Katona, Marta] University of Szeged, Department of Obstetrics and Gynaecology, Semmelweis u. 1, 6725 Szeged, Hungary. [Szabo, Janos] University of Szeged, Department of Obstetrics and Gynaecology, Semmelweis u. 1, 6725 Szeged, Hungary. [Pal, Attila] University of Szeged, Department of Obstetrics and Gynaecology, Semmelweis u. 1, 6725 Szeged, Hungary. RP Pal, A (reprint author), University of Szeged, Department of Obstetrics and Gynaecology, 6725 Szeged, Hungary. EM PALATTILA@obgyn.szote.u-szeged.hu CR Inbar D, Halpern GJ, Weitz R et al, 1997, Agenesis of the corpus callosum in a mother and son. Am J Med Genet 69:152–154 Lynn RB, Buchanan DC, Fenichel GM et al, 1980, Agenesis of the corpus callosum. Arch Neurol 37:444–445 Naiman J, Fraser FC, 1955, Agenesis of the corpus callosum; a report of two cases in siblings. AMA Arch Neurol Psychiatry 74:182–185 Naritomi K, Chinen Y, Asato Y, 1997, Agenesis of corpus callosum in three sibs. Jpn J Hum Genet 42:539–541 Pineda M, Gonzalez A, Fabregues I et al, 1984, Familial agenesis of the corpus callosum with hypothermia and apneic spells. Neuropediatrics 15:63–67 Pirola B, Bortotto L, Giglio S et al, 1998, Agenesis of the corpus callosum with Probst bundles owing to haploinsufficiency for a gene in an 8 cM region of 6q25. J Med Genet 35:1031–1033 Shapira Y, Cohen T, 1973, Agenesis of the corpus callosum in two sisters. J Med Genet 10:266–269 Tepper R, Zalel Y, Gaon E et al, 1996, Antenatal ultrasonographic findings differentiating complete from partial agenesis of the corpus callosum. Am J Obstet Gynecol 174:877–878 Lockwood CJ, Ghidini A, Aggarwal R et al, 1988, Antenatal diagnosis of partial agenesis of the corpus callosum: a benign cause of ventriculomegaly. Am J Obstet Gynecol 159:184–186 Aribandi M, 2004, Corpus Callosum, Agenesis. In: eMedicine Specialties/Radiology/PEDIATRICS. http://www.emedicine.com/ radio/topic193.htm. Cited 1 Oct 2007 Koubek SL, 2005, Agenesis of the Corpus Callosum. In: image the weekly source for radiology professionals vol. 18, no. 7. http:// www.rt-image.com. Cited 2 Oct 2007 Naional Centre for Healthcare Audit and Inspection, 2004, National Database of Birth Defects and Developmental Disabilities. http://www.oszmk.hu. Cited 2 Oct 2007 Dobyns WB, 1996, Absence makes the search grow longer. Am J Hum Genet 58:7–16 Jonas RE, Kimonis VE, Morales A, 1997, Possible new autosomal recessive syndrome of partial agenesis of the corpus callosum, pontine hypoplasia, focal white matter changes, hypotonia, mental retardation, and minor anomalies. Am J Med Genet 73:184–188 Velisavljev-Filipovic G, 2005, Agenesis of the corpus callosum in a premature infant associated with Langdon–Down syndrome. Med Pregl 58:587–591 Ellison D, Love S, Chimelli L, eds,, 1998, Neuropathology. A reference text of CNS pathology. Mosby Publishing Pilu G, 1999, Agenesis of the corpus callosum. In: Articles/ Central Nervous System/Agenesis of the corpus callosum. http:// www.thefetus.net/page.php?id=73. Cited 5 Oct 2007 Moriyama E, Nishida A, Sonobe H, 2007, Interhemispheric multiloculated ependymal cyst with dysgenesis of the corpus callosum: a case in a preterm fetus. Childs Nerv Syst 23:807–813 Franco I, Kogan S, Fisher J et al, 1993, Genitourinary malformations associated with agenesis of the corpus callosum. J Urol 149: 1119–1121 Gonzalez-Zamora JF, Villegas-Alvarez F, 2005, Esophageal atresia and chromosomal abnormalities in a Mexican children population: descriptive analysis. Cir Pediatr 18:196–199 Sugito K, Koshinaga T, Hoshino M et al, 2006, Study of 24 cases with congenital esophageal atresia: what are the risk factors? Pediatr Int 48:616–621 Saing H, Mya GH, Cheng W, 1998, The involvement of two or more systems and the severity of associated anomalies significantly influence mortality in esophageal atresia. J Pediatr Surg 33:1596–1598 Engum SA, Grosfeld JL, West KW et al, 1995, Analysis of morbidity and mortality in 227 cases of esophageal atresia and/or tracheoesophageal fistula over two decades. Arch Surg 130:502– 508, discussion 508–509 Ein SH, Shandling B, Wesson D et al, 1989, Esophageal atresia with distal tracheoesophageal fistula: associated anomalies and prognosis in the 1980s. J Pediatr Surg 24:1055–1059 Niedrist D, Riegel M, Achermann J et al, 2006, Survival with trisomy 18—data from Switzerland. Am J Med Genet 140: 952–959 Kamnasaran D, 2005, Agenesis of the corpus callosum: lessons from humans and mice. Clin Invest Med 28:267–282 Curnes JT, Laster DW, Koubek TD et al, 1986, MRI of corpus callosal syndromes. AJNR Am J Neuroradiol 7:617–622 Utsunomiya H, Ogasawara T, Hayashi T et al, 1997, Dysgenesis of the corpus callosum and associated telencephalic anomalies: MRI. Neuroradiology 39:302–310 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2009 VL 15 IS 1 BP 147 EP 152 DI 10.1007/s12253-008-9023-2 PG 6 ER PT J AU Zhou, Chj Zhang, Qh Zhang, Tg Sun, Shz Li, H Wang, Y Liu, Zy AF Zhou, Cheng-jun Zhang, Qing-hui Zhang, Ting-guo Sun, Shan-zhen Li, Hong Wang, Yan Liu, Zhi-yan TI Expression of ER, Ki-67 and CylinD1 in the Pre-cancerous Breast of Chinese Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pre-cancerous lesions; Breast; ER; Ki-67; CyclinD1 ID Pre-cancerous lesions; Breast; ER; Ki-67; CyclinD1 AB To investigate the expression and association of ER, Ki-67 and cyclinD1 in usual ductal hyperplasia(UDH), atypical ductal hyperplasia (ADH) and ductal carcinoma in situ(DCIS) in the breast. The study included 56 cases of precancerous lesions which were surgically excised at Qi Lu Hospital of Shangdong University. Immunohistochemistry was used to determine the expression of ER, Ki-67 and cyclinD1 and double-labelling immunofluorescence technique was used to observe the coexpression of ER and Ki-67. The expression and distribution of ER-positive cells were significantly different in UDH, ADH and DCIS. The ER-positive cells were much more in UDH than in normal TDLUs (terminal duct lobular units). The distribution of ER-positive cells interspersed amid ER-negative cells within UDH. However , the ER positive cells showed marked increases in ADH and low grade nuclear DCIS (P<0.05), distributing in almost all constituent cells. The expression of ki-67 and cyclinD1 were significantly different between UDH and DCIS (P<0.05) , and a positive correlation was found between expression of Ki-67 and morphological classification of pre-cancerous lesions (r=0.3522, P<0.05) as well as cyclinD1 (r=0.3901, P<0.05). Double-labelling immunofluorescence showed that there was no coexpression of ER and Ki-67 in normal breast tissue. The coexpression of the two markers was found in ADH and increased in DCIS. Overexpression of ER, Ki-67 and cyclinD1 significantly accompanies the transition of normal cells and UDH to ADH and DCIS. The coexpression of ER and ki-67 may present the early change in carcinogenesis of breast cancer. C1 [Zhou, Cheng-jun] The Second Hospital of Shandong University, 250033 Jinan, China. [Zhang, Qing-hui] Shandong University, Qilu Hospital, Department of Pathology, 250012 Jinan, China. [Zhang, Ting-guo] Shandong University, Qilu Hospital, Department of Pathology, 250012 Jinan, China. [Sun, Shan-zhen] Shandong University, Stomatology Hospital, 250012 Jinan, China. [Li, Hong] Shandong University, Qilu Hospital, Department of Pathology, 250012 Jinan, China. [Wang, Yan] Shandong University, Qilu Hospital, Department of Pathology, 250012 Jinan, China. [Liu, Zhi-yan] Shandong University, Qilu Hospital, Department of Pathology, 250012 Jinan, China. RP Zhang, Qh (reprint author), Shandong University, Qilu Hospital, Department of Pathology, 250012 Jinan, China. EM chengjunzhou@eyou.com CR Yager JD, Davidson NE, 2006, Estrogen carcinogenesis in breast cancer. N Engl J Med 354:270–282 Shaaban AM, Sloane JP, West CR et al, 2002, Breast cancer risk in usual ductal hyperplasia is defined by estrogen receptor- and Ki-67 expression. Am J Pathol 160(2):597–604 Muss HB, Bunn JY, Crocker A et al, 2007, Cyclin D-1, interleukin-6, HER-2/neu, transforming growth factor receptor-II and prediction of relapsein women with early stage, hormone receptor-positive breast cancer treated with tamoxifen. Breast J 13, 4):337–345 Devilea TEF, 2003, WHO: pathology and genetics, tumor of the breast and female genital organs. IARC, Lyon, pp 63–73 Krishnamurthy S, Sneige N, 2002, Molecular and biologic markers of premalignant lesions of human breast. Adv Anat Pathol 3:185–197 Shoker BS, Jarvis C, Sibson DR et al, 1999, Estrogen receptor expression in the normal precancerous breast. J Pathol 188:237–244 Allegra JC, Lippman ME, Green L et al, 1979, Estrogen receptor values in patients with benign breast disease. Cancer 44:228–231 Peterson OW, Hoyer PE, Van Deurs B, 1986, Frequency and distribution of estrogen receptor-positive cells in normal non lactating human breast tissue. J Natl Cancer Inst 77:343–349 Ricketts D, Turnbull L, Tyall G et al, 1991, Oestrogen and progesterone receptors in the normal female breast. Cancer Res 51:1817–1822 Mohsin SK, Hilsenbeck SG, Allred DC, 2000, Estrogen receptor and growth control in premalignant breast disease. Mod Pathol 13:28A Giri DD, Dundas SAC, Nottingham JF, Underwood JCE, 1989, Oestrogen receptors in benign epithelial lesions and introductal carcinoma of the breast: an immunohistological study. Histopathology 15:575–584 Baqai T, Shousha S, 2003, Oestrogen receptor negativity as a marker for high-grade ductal carcinoma in situ of the breast. Histopathology 42:440–447 Fuqua SA, Wiltschke C, Zhang ZX et al, 2000, A hypersensitive estrogen receptor-alpha mutation in premalignant breast lesions. Cancer Res 60:4026–4029 Clarke RB, Howell A, Potten CB, Anderson E, 1997, Dissociation between steroid receptor expression and cell proliferation in the human breast. Cancer Res 57:4987–4991 Ciocca DR, Fanelli MA, 1997, Estrogen receptor and cell proliferation in breast cancer. TEM 8:313–321 Russo IH, Russo J, 1998, Role of hormones in mammary cancer initiation and progression. J Mammary Gland Biol Neoplasia 3:49–64 Shoker BS, Javis C, Clarke RB et al, 1999, Estrogen receptorpositive proliferating cells in the normal and precancerous breast. Am J Pathol 155:1811–1815 Mussawar L, Michael PAD, Shoker BS et al, 2001, Subgroups of non-atypical hyperplasia of breast defined by proliferation of oestrogen receptor-positive cells. J Pathol 193:333–338 Kontos M, Nikolopoulou M, Trafalis DT et al, 2005, The effect of an estrone D-lactam steroid ester derivative on breast cancer cells and its predicted binding interactions with the ligand binding domain of estrogen receptor-alpha. Oncol Res 16:129–142 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 153 EP 158 DI 10.1007/s12253-008-9100-6 PG 6 ER PT J AU Egyed, Zs Jaray, B Kulka, J Pentek, Z AF Egyed, Zsofia Jaray, Balazs Kulka, Janina Pentek, Zoltan TI Triple Test Score for the Evaluation of Invasive Ductal and Lobular Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lobular; Ductal; Breast cancer; Sum-score diagnosis; Triple test; Symptomless ID Lobular; Ductal; Breast cancer; Sum-score diagnosis; Triple test; Symptomless AB The aim of our study was to compare the preoperative sum score diagnostics of invasive ductal and lobular cancers using three or four diagnostic methods. The novelty of this study is the examination of this phenomenon based on sum score, no such papers can be found in the literature. Ductal cancers have higher score values indicating easier diagnostics, but the difference in distribution of the scores was significant ( p=0.0086) only in case of the triple-test. The score values give appropriate opportunity to create their order of diagnostic power which was the same by both histologic types and in their subgroups with low sum-score: the strongest was cytology, followed by mammography, ultrasound and physical examination. No significant difference was found between the two histologic group in their mammographic appearances—stellate, circumscribed, assymmetric distortion or microcalcification (p=0.0694). In low score subgroup besides the occult forms, structural distortion and indeterminate microcalcifications overweighed the stellate and circumscribed lesions typical for the whole groups. In symptomless cases of both histologic groups only one strongly malignant diagnostic test result warrants the right diagnosis. Summarizing the score distribution of the results in case of four diagnostic tools the higher scores—indicating malignancy—were more frequent in the ductal group compared to the lobular ones. Extra attention has to be paid to rare radiomorphologic appearances and to the most deterministic examination, namely cytology. C1 [Egyed, Zsofia] MaMMa Egeszsegugyi Rt, Kapas u. 29, 1023 Budapest, Hungary. [Jaray, Balazs] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Pentek, Zoltan] MaMMa Egeszsegugyi Rt, Kapas u. 29, 1023 Budapest, Hungary. RP Egyed, Zs (reprint author), MaMMa Egeszsegugyi Rt, 1023 Budapest, Hungary. EM egyed.zsofia@t-online.hu CR Tot T, 2003, The diffuse type of invasive lobular carcinoma of the breast: morphology and prognosis. Virchows Arch 43:718–724 Martinez V, Azzopardi JG, 1979, Invasive lobular carcinoma of the breast: incidence and variants. Histopathology 493:467–488 Li CI, Anderson BO, 2003, Trends in incidence rates of invasive lobular and ductal breast carcinoma. JAMA 19(289):1421–1424 Adler OB, Engel A, 1990, Invasive lobular carcinoma. Mammographic pattern. Rofo 152:460–462 Ashikari R, Huvos AG, Urban JA, 1973, Infiltrating lobular carcinoma of the breast. Cancer 31:110–116 Broet P, de la Rochefordiere A, Scholl SM et al, 1995, Contralateral breast cancer:annual incidence and risk parameters. J Clin Oncol 13:1578–1583 Silverstein MJ, Lewinsky BS, Waisman Gierson ED et al, 1994, Infiltrating lobular carcinoma. Is it different from infiltrating duct carcinoma. Cancer 73:1673–1677 Biglia N, Mariani L, Sgro L et al, 2007, Increased incidence of lobular breast cancer in women treated with hormone replacement therapy: implications of diagnosis, surgical and medical treatment. Endocr Relat Cancer 14:549–567 Toikkanen S, Pylkannen L, Joensu H, 1997, Invasive lobular carcinoma of the breast has better short-term and long-term survival than invasive ductal carcinoma. Br J Cancer 78:1234–1240 Hermansen C, Poulsen H, Jensen J et al, 1984, Palpable breast tumours: “triple diagnosis” and operative strategy. Acta Chir Scand 150:625–628 Azzarelli A, Guzzon A, Pilotti S et al, 1983, Accuracy of breast cancer diagnosis by physical and radiologic and cytologic combined examinations. Tumori 69:137–141 Irwing L, Macaskill P, Houssami N, 2002, Evidence relevant to the investigation of breast symptoms:the triple test. Breast 11:215–220 Morris AM, Flowers CR, Morris KT et al, 2003, Comparing the cost-effectiveness of the triple test score to traditional methods for evaluating palpable breast masses. Med Care 41:962–971 Morris KT, Pommier RF, Morris A et al, 2001, Usefulness of triple test score for palpable breast masses. Arch Surg 136:1008– 1012 Vetto JT, Pommier RF, Schmidt WA et al, 1995, Use of “triple test” for palpable breast lesion yields high diagnostic accuracy and cost savings. Am J Surg 169:519–522 Houssami N, Irwing L, Simpson J et al, 2003, Sydney breast imaging accuracy study: comparative sensitivity and specificity of mammography and sonography in young women with symptoms. AJR 180:935–940 Morris KT, Vetto JT, Petty JK et al, 2002, A new score for the evaluation of palpable breast masses in women under age 40. Am J Surg 184:346–347 Houssami N, Irwing L, Loy C, 2002, Accuracy of combines breast imaging in young women. Breast 11:36–40 Kuhl CK, Schrading S, Weigel S et al, 2005, The EVA trial: evaluation of the efficacy of diagnostic methods, mammography, ultrasound, MRI, in the secondary and tertiary prevention of familiar breast cancer. Preliminary results after the first half of the study period. Rofo 177:818–827 Egyed Z, Jaray B, Pentek Z, 2006, Invasive lobular breast cancer: pitfall for the radiologist. Orv Hetil 5:219–226 American College of Radiology, 1995, Breast imaging reporting and data system, BI-RADS), 2nd edn. American College of Radiology, Reston Vincze I, Varbanova M, 1993, Non-parametric mathematical statistics. Theory, and practices, 1st edn. Akad. Kiado, Budapest, in Hungarian) Kemeny S, Deak A, Lakne Komka K et al, 2004, Statistical analysis using Statistica, 1st edn. Muegyetemi Kiado, Budapest, in Hungarian) Sartre-Garu X, Jouve M, Asselain B et al, 1994, Infiltrating lobular carcinoma of the breast Clinicopathologic analysis of 975 cases with reference to data on conservative therapy and metastatic patterns. Cancer 73:1673–1677 Newstead GM, Baute PB, Toth HK, 1992, Invasive lobular and ductal carcinoma: mammographic findings and stage at diagnosis. Radiology 184(3):623–627 Christopher IL, Anderson BO, Porter P et al, 2000, Changing incidence rate of invasive lobular breast carcinoma among older women. Cancer 88:2561–2569 Krecke KN, Gisvold JJ, 1993, Invasive lobular carcinoma of the breast: mammographic findings and extent of disease in 184 patients. AJR 88:957–960 Bazzocchi M, Facecchia I, Zulani C et al, 2000, Diagnostic imaging of lobular carcinoma of the breast: mammographic, ultrasonographic and MR findings. Radiol Med 100:436– 443 Foote FW Jr, Stewart FW, 1946, A histologic classification of carcinoma of the breast. Surgery 19:74–99 Langmar Z, Orosz Z, 1999, Clinico-pathology of lobular breast cancer. Orv Hetil 140(20):1099–1102 Sastre-Garau X, Jouve M, 1996, Infiltrating lobular carcinoma of the breast. Cancer 1:113–120 Silverstein MJ, Lewinsky BS, 1994, Infiltrating lobular carcinoma. Cancer 73:673–77 Steinberg JL, Trudeau ML, Ryder DE et al, 1996, Combined fine-needle aspiration, physical examination and mammography in the diagnosis of palpable breast massses: their relation to outcome for women with primary breast cancer. C J Surg 39: 302–311 Morris A, Pommier RF, Schmidt WA et al, 1998, Accurate evaluation of palpable breast masses by the triple test score. Arch Surg 133:390–394 Mansoor I, Zahrani I, 2002, Analysis of inconclusive breast FNA by triple test. J Pak Med Assoc. 52:25–29 Fenton JJ, Barton MB, Geiger AM et al, 2005, Screening clinical breast examination:how often does it miss lethal breast cancer. J Natl Cancer Inst Monogr 35:67–71 Gill G, Luke C, Roder D, 2006, Prognostic importance of palpability as a feature of screen-detected breast cancers. JMed Screen 13:98–101 Knight JA, Libstug AR, Moravan Vet al, 1998, An assessment of the influence of clinical breast examination reports on the interpretation of mammograms in a breast screening program. Breast Cancer Res Treat 48:65–71 Feigin KN, Keating DM, Telford PM et al, 2006, Clinical breast examination in a comprehensive breast cancer screening program: contribution and cost. Radiology 240:650–655 Bartella L, Liberman L, Morris EA et al, 2006, Nonpalpable mammographically occult invasive breast cancers detected by MRI. AJR 186:865–870 Mathieu I, Mazy S, Willemart B et al, 2005, Inconclusive triple diagnosis in breast cancer imaging: is there a place for scintimammography. J Nucl Med 46:1574–1581 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 159 EP 166 DI 10.1007/s12253-008-9083-3 PG 8 ER PT J AU Mannweiler, S Amersdorfer, P Trajanoski, S Terrett, AJ King, D Mehes, G AF Mannweiler, Sebastian Amersdorfer, Peter Trajanoski, Slave Terrett, A Jonathan King, David Mehes, Gabor TI Heterogeneity of Prostate-Specific Membrane Antigen (PSMA) Expression in Prostate Carcinoma with Distant Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Antibody; Immunohistochemistry; Therapy ID Prostate cancer; Antibody; Immunohistochemistry; Therapy AB Prostate-specific membrane antigen (PSMA) is a transmembrane protein that is overexpressed in advanced stage prostate adenocarcinomas. As a novel target for in vivo prognostic and therapeutic approaches, the distribution pattern of PSMA in primary and metastatic tumors is of significant interest. In this study we addressed the cellular distribution and heterogeneity of PSMA expression. Paraffinembedded sections of 51 patients with primary prostate carcinoma and distant metastases were evaluated. Immunohistochemistry was used to determine the cellular localization, staining intensity and positive cell fraction which were related to tumor type and growth pattern. We demonstrated differences in the intracellular localization of the PSMA immunostaining which seem to be related to the tumor differentiation pattern. A significant number of the primary tumors (7/51) and metastases (6/51) presented with highly heterogeneous PSMA expression and in further 2 primary, and 8 metastatic tumors the staining was in the negative range (<10% positive tumor cells). A direct correlation between histological parameters and PSMA expression could not be demonstrated. Our findings clearly support the feasibility but also direct to potential failures of PSMA-targeted in vivo diagnostic and therapeutic approaches in prostate cancer patients with distant metastasis. C1 [Mannweiler, Sebastian] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, 8036 Graz, Austria. [Amersdorfer, Peter] Oridis-Biomed GMBH, Stiftingtalgasse 3-5, 8010 Graz, Austria. [Trajanoski, Slave] Graz University of Technology, Christian Doppler Laboratory for Genomics and Bioinformatics, Petersgasse 14, 8010 Graz, Austria. [Terrett, A Jonathan] Medarex Inc., 521 Cottonwood Drive, 95035 Milpitas, CA, USA. [King, David] Medarex Inc., 521 Cottonwood Drive, 95035 Milpitas, CA, USA. [Mehes, Gabor] Oridis-Biomed GMBH, Stiftingtalgasse 3-5, 8010 Graz, Austria. RP Mannweiler, S (reprint author), Medical University of Graz, Department of Pathology, 8036 Graz, Austria. EM sebastian.mannweiler@meduni-graz.at CR Chang SS, 2004, Overview of prostate-specific membrane antigen. Rev Urol 6:S13–S16 Pinto JT, Suffoletto BP, Berzin TM et al, 1996, Prostate-specific membrane antigen: a novel folate hydrolase in human prostatic carcinoma cells. Clin Cancer Res 2:1445–1451 Bostwick DG, Pacelli A, Blute M, Roche P, Murphy GP, 1998, Prostate specific membrane antigen expression in prostatic intraepithelial neoplasia and adenocarcinoma: a study of 184 cases. Cancer 82:2256–2261 Perner S, Hofer MD, Kim R, Shah RB, Li H, Moller P, Hautmann RE, Gschwendt JE, Kuefer R, Rubin MA, 2007, Prostate-specific membrane antigen expression as a predictor of prostate cancer progression. Hum Pathol 38:696–701 Horoszewicz JS, Kawinski E, Murphy GP, 1987, Monoclonal antibodies to a new antigenic marker in epithelial cells and serum of prostatic cancer patients. Anticancer Res 7:927–936 Moreno JG, Croce CM, Fischer R et al, 1992, Detection of hematogenous micrometastases in patients with prostate cancer. Cancer Res 52:6110–6112 Silver DA, Pellicer I, Fair WL et al, 1997, Prostate-specific membrane antigen expression in normal and malignant human tissue. Clin Cancer Res 3:81–85 Chang S, O’Keefe DS, Bacich DJ, Reuter VE, Heston WD Gaudin PB, 1999, Prostate-specific membrane antigen is produced in tumor-associated neovasculature. Clin Cancer Res 5:2674–2681 Wright GL, Grob BM, Haley C, Grossman K, Newhall K, Petrylak D, Troyer J, Konchuba A, Schellhammer PF, Moriarty R, 1996, Upregulation of prostate-specific membrane antigen after androgen-deprivation therapy. Urology 48:326–334 Gleason DF, 1977, Histologic grading and clinical staging of prostatic carcinoma. In: Tannebaum M, ed, Urologic pathology: the prostate. Lea & Febiger, Philadelphia, pp 171–197 Lopez-Beltran A, Mikuz G, Luque RJ, Mazzucchelli R, Montironi R, 2006, Current practice of Gleason grading of prostate carcinoma. Virchows Arch 448(2):111–118 Douglas G. Altman: Practical Statistics for Medical Research Chapman & Hall/CRC 1 edition, November 22, 1990) Bander NH, 2006, Technology insight: monoclonal antibody imaging of prostate cancer. Nat Clin Pract Urol 3(4):216–225 Sodee DB, Sodee AE, Bakale G, 2007, Synergistic value of single-photon emission computed tomography/computed tomography fusion to radioimmunoscintigraphic imaging of prostate cancer. Semin Nucl Med 37(1):17–28 Elsasser-Beile U, Wolf P, Gierschner D, Buhler P, Schultze- Seemann W, Wetterauer U, 2006, A new generation of monoclonal and recombinant antibodies against cell-adherent prostate specific membrane antigen for diagnostic and therapeutic targeting of prostate cancer. Prostate 66(13):1359–1370 Ross JS, Sheehan CHE, Fisher HAG, Kaufman RP, Kaur P, Gray K, Webb I, Gray GS, Mosher R, Kallakury BVS, 2003, Correlation of primary tumor prostate-specific membrane antigen expression with disease recurrence in prostate cancer. Clin Cancer Res 9:6357–6362 Zhigang Z, Wenly S, 2004, Prostate stem cell antigen, PSCA, expression in human prostate cancer tissue and its potential role in prostate carcinogenesis and progression of prostate cancer. W J Surg Oncol 2:1–13 Buhler P, Wolf P, Gierschner D, Schabel I, Katzenwadel A, Schultze-Seemann W, Wetterauer U, Tacke M, Swamy M, Schamel WW, Elsasser-Beile U, 2008, A bispecific diabody directed against prostate-specific membrane antigen and CD33 induces T-cell mediated lysis of prostate cancer cells. Can Immunol Immunother 57(1):43–52 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 167 EP 172 DI 10.1007/s12253-008-9104-2 PG 6 ER PT J AU Giaginis, TC Vgenopoulou, S Tsourouflis, SG Politi, NE Kouraklis, PG Theocharis, ES AF Giaginis, T Constantinos Vgenopoulou, Stephanie Tsourouflis, S Gerasimos Politi, N Ekaterini Kouraklis, P Gregorios Theocharis, E Stamatios TI Expression and Clinical Significance of Focal Adhesion Kinase in the Two Distinct Histological Types, Intestinal and Diffuse, of Human Gastric Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Clinicopathological parameters; Diffuse; Focal adhesion kinase; Gastric cancer; Immunohistochemistry; Intestinal; Patients’ survival ID Clinicopathological parameters; Diffuse; Focal adhesion kinase; Gastric cancer; Immunohistochemistry; Intestinal; Patients’ survival AB Focal adhesion kinase (FAK), a non-receptor tyrosine kinase protein, acts as an early modulator of integrin signaling cascade, regulating basic cellular functions. In transformed cells, unopposed FAK signaling has been considered to promote tumor growth, progression and metastasis. The aim of this study was to assess the clinical significance of FAK expression in the two distinct histological types of human gastric neoplasia. FAK expression was assessed immunohistochemically in tumoral samples of 66 gastric adenocarcinoma cases, 30 intestinal and 36 diffuse type, and was statistically analyzed in relation to various clinicopathological characteristics, tumor proliferative capacity and patients’ survival. In intestinal type carcinomas, enhanced FAK expression was significantly associated with increased tumor proliferative capacity (P=0.012). In diffuse type carcinomas, FAK staining intensity was significantly correlated with tumor size (P=0.026) and disease stage (P=0.024), presenting also a borderline association with nodal status (P=0.053). In diffuse type carcinomas, enhanced FAK expression was significantly associated with longer overall survival times (log-rank test, P=0.014), being also identified as an independent prognostic factor in multivariate analysis (Cox regression, P=0.016). In contrast, patients with intestinal type tumors and enhanced FAK expression were characterized by shorter overall survival times, without though reaching statistical significance (log-rank test, P=0.092). The current data support evidence that FAK protein may be considered as a diagnostic and prognostic marker in gastric neoplasia. Further studies conducted on larger clinical samples and highlighting on the distinct impact of the two histological types are warranted to delineate the clinical significance of FAK protein in gastric neoplasia. C1 [Giaginis, T Constantinos] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias street, Goudi, GR11527 Athens, Greece. [Vgenopoulou, Stephanie] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias street, Goudi, GR11527 Athens, Greece. [Tsourouflis, S Gerasimos] University of Athens, Medical School, Second Department of Propedeutic SurgeryAthens, Greece. [Politi, N Ekaterini] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias street, Goudi, GR11527 Athens, Greece. [Kouraklis, P Gregorios] University of Athens, Medical School, Second Department of Propedeutic SurgeryAthens, Greece. [Theocharis, E Stamatios] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias street, Goudi, GR11527 Athens, Greece. RP Theocharis, ES (reprint author), University of Athens, Medical School, Department of Forensic Medicine and Toxicology, GR11527 Athens, Greece. EM theocharis@ath.forthnet.gr CR Lipfert L, Haimovich B, Schaller MD, Cobb BS, Parsons JT, Brugge JS, 1992, Integrin-dependent phosphorylation and activation of the protein tyrosine kinase pp125FAK in platelets. J Cell Biol 119:905–912 Zachary I, Sinnett-Smith J, Rozengurt E, 1992, Bombesin, vasopressin, and endothelin stimulation of tyrosine phosphorylation in Swiss 3T3 cells. Identification of a novel tyrosine kinase as a major substrate. J Biol Chem 267:19031–19034 Ilic D, Furuta Y, Kanazawa S, Takeda N, Sobue K, Nakatsuji N, Nomura S, Fujimoto J, Okada M, Yamamoto T, 1995, Reduced cell motility and enhanced focal adhesion contact formation in cells from FAK-deficient mice. Nature 377:539–544 Cary LA, Chang JF, Guan JL, 1991, Stimulation of cell migration by overexpression of focal adhesion kinase and its association with Src and Fyn. J Cell Sci 109:1787–1794 Li S, Kim M, Hu YL, Jalali S, Schlaepfer DD, Hunter T, Chien S, Shyy JY, 1997, Fluid shear stress activation of focal adhesion kinase. Linking to mitogen-activated protein kinases. J Biol Chem 272:30455–30462 Levy P, Robin H, Kornprobst M, Capeau J, Cherqui G, 1989, Enterocytic differentiation of human Caco-2 cell line correlates with alterations in integrin signaling. J Cell Physiol 177:618–627 Xu LH, Owens LV, Sturge GC, Yang X, Liu ET, Craven RJ, Cance WG, 1996, Attenuation of the expression of the focal adhesion kinase induces apoptosis in tumor cells. Cell Growth Differ 7:413–418 Chatzizacharias NA, Kouraklis G, Theocharis S, 2008, Clinical significance of FAK expression in human neoplasia. Histol Histopathol 23:629–650 Chatzizacharias NA, Kouraklis G, Theocharis S, 2007, Focal adhesion kinase: a promising target for anticancer therapy. Exp Opin ther Targets 11:1315–1328 Chatzizacharias NA, Kouraklis G, Theocharis S, 2008, Disruption of FAK signaling: A side mechanism in cytotoxicity. Toxicology 245:1–10 Iravani S, Mao W, Fu L, Karl R, Yeatman T, Jove R, Coppola D, 1998, Elevated c-Src protein expression is an early event in colonic neoplasia. Lab Invest 78:365–371 Owens LV, Xu L, Craven RJ, Dent GA, Weiner TM, Koengerg L, Liu ET, Cance WG, 1995, Overexpression of the focal adhesion kinase, p125FAK, in invasive human tumors. Cancer Res 55:2752–2755 Theocharis SE, Kouraklis GP, Kakisis JD, Kanelli HG, Apostolakou FE, Karatzas GM, Koutselinis AS, 2003, Focal adhesion kinase expression is not a prognostic predictor in colon adenocarcinoma patients. Eur J Surg Oncol 29:571–574 Furuyama K, Ryuchiro D, Tomohiko M, Toyoda E, Ito D, Kami K, Koizumi M, Kida A, Kawaguchi Y, Fujimoto K, 2006, Clinical significance of focal adhesion kinase in resectable pancreatic cancer. World J Surg 30:219–226 Fuji T, Koshikawa K, Nomoto S, Okochi O, Kaneko T, Inoue S, Yatabe Y, Takeda S, Nakao A, 2004, Focal adhesion kinase is overexpressed in hepatocellular carcinoma and can be served as an independent prognostic factor. J Hepatol 41:104–111 Itoh S, Maeda T, Shimada M, Aishima S, Shirabe K, Tanaka S, Maehara Y, 2004, Role of expression of focal adhesion kinase in progression of hepatocellular carcinoma. Clin Cancer Res 10:2812–2817 Gabriel B, zur Hausen A, Stickeler E, Dietz C, Gitsch G, Fischer DC, Boulda J, Tempfer C, Hasenburg A, 2006, Weak expression of focal adhesion kinase, pp125FAK, in patients with cervical cancer is associated with poor disease outcome. Clin Cancer Res 12:2476–2483 Imaizumi M, Nishimura M Takeuchi S, Murase M, Hamaguchi M, 1997, Role of tyrosine specific phosphorylation of cellular proteins, especially EGF receptor and p125FAK in human lung cancer cells. Lung Cancer 17:69–84 He QY, Cheung YH, Leung SY, Yuen ST, Chu KM, Chiu JF, 2004, Diverse proteomic alteration in gastric adenocarcinoma. Proteomics 4:3276–3287 Forman D, Barley VJ, 2006, Gastric cancer: global pattern of the disease and an overview of environmental risk factors. Best Practice Res Clin Gastroenterol 20:633–649 Hohenberger P, Gretschel S, 2003, Gastric cancer. Lancet 362:305–315 Tani T, Von Koskull H, Virtanen I, 1996, Focal adhesion kinase pp 125FAK is associated with both intercellular junctions and matrix adhesion sites in vivo. Histochem Cell Biol 105:17–25 Su JM, Gui L, Zhou YP, Zha XL, 2002, Expression of focal adhesion kinase and alpha5 and beta1 integrins in carcinomas and its clinical significance. World J Gastroenterol 8:613–618 Vauhkonen M, Vauhkonen H, Sipponen P, 2006, Pathology and molecular biology of gastric cancer. Best Pract Res Clin Gastroenterol 20:651–674 Tahara E, 2004, Genetic pathways of two types of gastric cancer. IARC Sci Publ 157:327–349 Cervantes A, Rodriguez Braun E, Perez Fidalgo A, Chirivella Gonzalez I, 2007, Molecular biology of gastric cancer. Clin Transl Oncol 9:208–215 Lauren P, 1965, The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. Act Pathol Microbiol Scand 64:31–49 Sobin LH, Wittekind C, 1997, TNM Classification of Malignant Tumors, 5th edn. Wiley-Liss, New York Giaginis C, Davides D, Zarros A, Noussia O, Zizi-Serbetzoglou A, Kouraklis G, Theocharis S, 2008, Clinical significance of tumor-associated antigen RCAS1 expression in human pancreatic ductal adenocarcinoma. Dig Dis Sci 53:1728–1734 Cance WG, Harris JE, Iacocca MV, Roche E, Yang X, Chang J, Simkins S, Xu L, 2000, Immunohistochemical analyses of focal adhesion kinase expression in benign and malignant human breast and colon tissues: correlation with preinvasive and invasive phenotypes. Clin Cancer Res 6:2417–2423 Miyaki T, Kato H, nakajima M, Sohda M, Fukai Y, Masuda N, Manda N, Fukuchi M, Tsukada K, Kuwano H, 2003, FAK overexpression is correlated with tumor invasiveness and lymph node metastasis in oesophageal squamous cell carcinoma. Br J Cancer 89:140–145 Lark AL, Livasy CA, Dressler L, Moore DT, Millikan RC, Geradts J, Iacocca M, Cowan D, Little D, Craven RJ, Cance W, 2005, High focal adhesion kinase expression in invasive breast carcinomas is associated with an aggressive phenotype. Mod Pathol 18:1289–1294 Cox BD, Natarajan M, Stettner MR, Gladson CL, 2006, New concepts regarding focal adhesion kinase promotion of cell migration and proliferation. J Cell Biochem 99:35–52 Aguirre Ghiso JA, 2002, Inhibition of FAK signaling activated by urokinase receptor induces dormancy in human carcinoma cells in vivo. Oncogene 21:2513–2524 Ohta R, Yamashita Y, Taketomi A, Kitagawa D, Kuroda Y, Itoh S, Aishima S, Maehara Y, 2006, Reduced expression of focal adhesion kinase in intrahepatic cholangiocarcinoma is associated with poor tumor differentiation. Oncology 71:417–422 Carelli S, Zadra G, Vaira V, Falleni M, Bottiglieri L, Nosotti M, Di Giulio AM, Gorio A, Bosari S, 2006, Up-regulation of focal adhesion kinase in non-small cell lung cancer. Lung Cancer 53:263–271 Wang D, Grammer JR, Cobbs CS, Stewart JE Jr, Liu Z, Rhoden R, Hecker TP, Ding Q, Gladson CL, 2000, p125 Focal adhesion kinase promotes malignant astrocytoma cell proliferation in vitro. J Cell Sci 113:4221–4230 Gabarra-Niecko V, Schaller MD, Dunty JM, 2003, FAK regulates biological processes important for the pathogenesis of cancer. Cancer Metastasis Rev 22:359–374 Sood AK, Coffin JE, Schneider GB, Fletcher MS, DeYoung BR, Gruman LM, Gershenson DM, Schaller MD, Hendrix MJ, 2004, Biological significance of focal adhesion kinase in ovarian cancer: role in migration and invasion. Am J Pathol 165:1087–1095 Yu HG, Tong SL, Ding YM, Fang XM, Zhang XF, Liu ZJ, Zhou YH, Liu QS, Luo HS, Yu JP, 2006, Enhanced expression of cholecystokinin-2 receptor promotes the progression of colon cancer through activation of focal adhesion kinase. Int J cancer 119:2724–2732 Schmitz KJ, Grabellus F, Callies R, Otterbach F, Wohlschlaeger J, Levkau B, Kimmig R, Schmid KW, Baba HA, 2005, High expression of focal adhesion kinase, p125FAK, in node-negative breast cancer is related to overexpression of HER-2/neu and activated Akt kinase but does not predict outcome. Breast Cancer Res 7:R194–203 Canel M, Aecade P, Rodrigo JP, Cabanillas R, Herrero A, Suarez C, Chiara MD, 2006, Overexpression of focal adhesion kinase in head and neck squamous cell carcinoma is independent of fak gene copy number. Clin Cancer Res 12:3272–3279 Zheng H, Takahashi H, Murai Y, Cui Z, Nomoto K, Miwa S, Tsuneyama K, Takano Y, 2007, Pathobiological characteristics of intestinal and diffuse-type gastric carcinoma in Japan: an immunostaining study on the tissue microarray. J Clin Pathol 60:273–277 Viste A, Eide GE, Halvorsen K, Maartmann-Moe H, Soreide O, 1986, The prognostic value of Lauren's histopathological classification system and ABO blood groups in patients with stomach carcinoma. Eur J Surg Oncol 12:135–141 Xu LH, Yang X, Bradham CA, Brenner DA, Baldwin AS Jr, Craven RJ, Cance WG, 2000, The focal adhesion kinase suppresses transformation-associated, anchorage-independent apoptosis in human breast cancer cells. Involvement of death receptor-related signaling pathways. J Biol Chem 275:30597– 30604 Guan GX, Jian HX, Lei DY, Lu HS, Zhang XF, 2006, Construction of retroviral vector of p125FAK specific ribozyme genes and its effects on BGC-823 cells. World J Gastroenterol 12:686–690 Scanlon KJ, 2004, Anti-genes: siRNA, ribozymes and antisense. Curr Pharm Biotechnol 5:415–420 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 173 EP 181 DI 10.1007/s12253-008-9120-2 PG 9 ER PT J AU Hritz, I Gyorffy, H Molnar, B Lakatos, G Sipos, F Pregun, I Juhasz, M Pronai, L Schaff, Zs Tulassay, Zs Herszenyi, L AF Hritz, Istvan Gyorffy, Hajnalka Molnar, Bela Lakatos, Gabor Sipos, Ferenc Pregun, Istvan Juhasz, Mark Pronai, Laszlo Schaff, Zsuzsa Tulassay, Zsolt Herszenyi, Laszlo TI Increased p53 Expression in the Malignant Transformation of Barrett’s Esophagus is Accompanied by an Upward Shift of the Proliferative Compartment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Barrett’s esophagus; Reflux esophagitis; Dysplasia; Adenocarcinoma; Cell proliferation; p53 expression ID Barrett’s esophagus; Reflux esophagitis; Dysplasia; Adenocarcinoma; Cell proliferation; p53 expression AB Neoplastic progression in Barrett’s esophagus (BE) occurs by a multistep process associated with early molecular and morphological changes. This study evaluated cell proliferation and p53 expression and their correlation in the development and progression of esophageal adenocarcinoma. PCNA and p53 expressions were analyzed in biopsy samples by immunohistochemistry including patients with reflux esophagitis, BE, BE with concomitant esophagitis, Barrett’s dysplasia, esophageal adenocarcinoma and a control group without any histological changes. Progressive increase in cell proliferation and p53 expression was found in the sequence of malignant transformation of the esophageal mucosa. While cell proliferation was significantly lower in the control group compared with all other groups, there was no increase in p53 expression of esophageal tissues that were negative for dysplasia. Dysplastic BE tissues revealed significantly higher cell proliferation and p53 expression levels compared to BE, reflux esophagitis or BE with concomitant esophagitis. Both, cell proliferation and p53 expression were significantly higher in adenocarcinoma compared to BE or Barrett’s dysplasia. Interestingly, while just BE with concomitant esophagitis showed significantly higher p53 expression levels than BE, both, BE with concomitant esophagitis and reflux esophagitis revealed significantly higher cell proliferation levels compared to BE. Alterations of cell proliferation and p53 expression showed a strong correlation. Simultaneous activation of cell proliferation and p53 expression strongly suggest their association with esophageal epithelial tumor genesis and particularly, their specific role in the biology of esophageal adenocarcinoma. Quantification of these parameters in BE is thought to be useful to identify patients at higher risk for progression to adenocarcinoma. C1 [Hritz, Istvan] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Gyorffy, Hajnalka] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Lakatos, Gabor] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Pregun, Istvan] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Juhasz, Mark] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Pronai, Laszlo] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Herszenyi, Laszlo] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. RP Hritz, I (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM hritz.istvan@freemail.hu CR Bosetti C, Levi F, Ferlay J et al, 2008, Trends in oesophageal cancer incidence and mortality in Europe. Int J Cancer 122, 5):1118–1129 Wu X, Chen VW, Andrews PA et al, 2007, Incidence of esophageal and gastric cancers among Hispanics, non-Hispanic whites and non-Hispanic blacks in the United States: subsite and histology differences. Cancer Causes Control 18(6):585–593 Flejou JF, 2005, Barrett’s oesophagus: from metaplasia to dysplasia and cancer. Gut 54(Suppl 1):i6–i12 Spechler SJ, Goyal RK, 1986, Barrett’s esophagus. N Engl J Med 315(6):362–371 Westhoff B, Brotze S, Weston A et al, 2005, The frequency of Barrett’s esophagus in high-risk patients with chronic GERD. Gastrointest Endosc 61(2):226–231 Jankowski JA, Harrison RF, Perry I et al, 2000, Barrett’s metaplasia. Lancet 356(9247):2079–2085 Shaheen NJ, 2005, Advances in Barrett’s esophagus and esophageal adenocarcinoma. Gastroenterology 128(6):1554–1566 Srivastava A, Hornick JL, Li X et al, 2007, Extent of low-grade dysplasia is a risk factor for the development of esophageal adenocarcinoma in Barrett’s esophagus. Am J Gastroenterol 102, 3):483–493 Schlansky B, Dimarino AJ Jr, Loren D et al, 2006, A survey of oesophageal cancer: pathology, stage and clinical presentation. Aliment Pharmacol Ther 23(5):587–593 Skacel M, Petras RE, Gramlich TL et al, 2000, The diagnosis of low-grade dysplasia in Barrett’s esophagus and its implications for disease progression. Am J Gastroenterol 95(12):3383–3387 McManus DT, Olaru A, Meltzer SJ, 2004, Biomarkers of esophageal adenocarcinoma and Barrett’s esophagus. Cancer Res 64(5):1561–1569 Younes M, Ertan A, Lechago LV et al, 1997, p53 protein accumulation is a specific marker of malignant potential in Barrett’s metaplasia. Dig Dis Sci 42(4):697–701 Hong MK, Laskin WB, Herman BE et al, 1995, Expansion of the Ki-67 proliferative compartment correlates with degree of dysplasia in Barrett’s esophagus. Cancer 75(2):423–429 Prelich G, Tan CK, Kostura M et al, 1987, Functional identity of proliferating cell nuclear antigen and a DNA polymerase auxiliary protein. Nature 326(6112):517–520 Reid BJ, Sanchez CA, Blount PL et al, 1993, Barrett’s esophagus: cell cycle abnormalities in advancing stages of neoplastic progression. Gastroenterology 105(1):119–129 el-Deiry WS, Harper JW, O’Connor PM et al, 1994, WAF1/CIP1 is induced in p53-mediated G1 arrest and apoptosis. Cancer Res 54(5):1169–1174 Ramel S, Reid BJ, Sanchez CA et al, 1992, Evaluation of p53 protein expression in Barrett’s esophagus by two-parameter flow cytometry. Gastroenterology 102(4 Pt 1):1220–1228 Wang DY, Xiang YY, Tanaka M et al, 1994, High prevalence of p53 protein overexpression in patients with esophageal cancer in Linxian, China and its relationship to progression and prognosis. Cancer 74(12):3089–3096 Slade N, Moll UM, 2003, Mutational analysis of p53 in human tumors: immunocytochemistry. Methods Mol Biol 234:231–243 Bian YS, Osterheld MC, Bosman FT et al, 2001, p53 gene mutation and protein accumulation during neoplastic progression in Barrett’s esophagus. Mod Pathol 14(5):397–403 Geboes K, Van Eyken P et al, 2000, The diagnosis of dysplasia and malignancy in Barrett’s oesophagus. Histopathology 37, 2):99–107 Polkowski W, van Lanschot JJ, ten Kate FJ et al, 1995, The value of p53 and Ki67 as markers for tumour progression in the Barrett’s dysplasia–carcinoma sequence. Surg Oncol 4(3):163– 171 Herszenyi L, Hritz I, Pregun I et al, 2007, Alterations of glutathione S-transferase and matrix metalloproteinase-9 expressions are early events in esophageal carcinogenesis. World J Gastroenterol 13(5):676–682 Hritz I, Kuester D, Vieth M et al, 2006, Secretory leukocyte protease inhibitor expression in various types of gastritis: a specific role of Helicobacter pylori infection. Eur J Gastroenterol Hepatol 18(3):277–282 Hritz I, Herszenyi L, Molnar B et al, 2005, Proton pump inhibitor co-therapy normalizes the increased cell turnover of the gastric mucosa both in NSAID and selective COX-2 users. Int J Immunpathol Pharmacol 18(1):75–84 Hritz I, Herszenyi L, Molnar B et al, 2005, Long-term omeprazole and esomeprazole treatment does not significantly increase gastric epithelial cell proliferation and epithelial growth factor receptor expression and has no effect on apoptosis and p53 expression. World J Gastroenterol 11(30):4721–4726 Fitzgerald RC et al, 2006, Molecular basis of Barrett’s oesophagus and oesophageal adenocarcinoma. Gut 55(12):1810–1820 Reid BJ, Weinstein WM, Lewin KJ et al, 1988, Endoscopic biopsy can detect high-grade dysplasia or early adenocarcinoma in Barrett’s esophagus without grossly recognizable neoplastic lesions. Gastroenterology 94(1):81–90 Theisen J, Nigro JJ, DeMeester TR et al, 2004, Chronology of the Barrett’s metaplasia–dysplasia–carcinoma sequence. Dis Esophagus 17(1):67–70 Kleeff J, Friess H, Liao Q et al, 2002, Immunohistochemical presentation in non-malignant and malignant Barrett’s epithelium. Dis Esophagus 15(1):10–15 Biasco G et al, 2004, Cell proliferation and differentiation in familial adenomatous polyposis, FAP). Hum Pathol 35, 12):1573 Scalmati A, Roncucci L, Ghidini G et al, 1990, Epithelial cell kinetics in the remaining colorectal mucosa after surgery for cancer of the large bowel. Cancer Res 50(24):7937–7941 Bechi P, Balzi M, Becciolini A et al, 1996, Helicobacter pylori and cell proliferation of the gastric mucosa: possible implications for gastric carcinogenesis. Am J Gastroenterol 91(2):271–276 Peters FT, Ganesh S, Kuipers EJ et al, 1998, Epithelial cell proliferative activity of Barrett’s esophagus: methodology and correlation with traditional cancer risk markers. Dig Dis Sci 43, 7):1501–1506 Rioux-Leclercq N, Turlin B, Sutherland F et al, 1999, Analysis of Ki-67, p53 and Bcl-2 expression in the dysplasia–carcinoma sequence of Barrett’s esophagus. Oncol Rep 6(4):877–882 Kim R, Clarke MR, Melhem MF et al, 1997, Expression of p53, PCNA, and CerbB-2 in Barrett’s metaplasia and adenocarcinoma. Dig Dis Sci 42(12):2453–2462 Kataoka H, Joh T, Kasugai K et al, 1997, Mitogenic properties of human gastric juice. Dig Dis Sci 42(8):1747–1754 Lagergren J, Bergstrom R, Lindgren A et al, 1999, Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N Engl J Med 340(11):825–831 Shaheen N, Ransohoff DF et al, 2002, Gastroesophageal reflux, Barrett esophagus, and esophageal cancer: scientific review. JAMA 287(15):1972–1981 Peters FT, Ganesh S, Kuipers EJ et al, 2000, Effect of elimination of acid reflux on epithelial cell proliferative activity of Barrett esophagus. Scand J Gastroenterol 35(12):1238–1244 Ireland AP, Clark GW, DeMeester TR, 1997, Barrett’s esophagus. The significance of p53 in clinical practice. Ann Surg 225(1): 17–30 Flejou JF, Potet F, Muzeau F et al, 1993, Overexpression of p53 protein in Barrett’s syndrome with malignant transformation. J Clin Pathol 46(4):330–333 Schneider PM, Casson AG, Levin B et al, 1996, Mutations of p53 in Barrett’s esophagus and Barrett’s cancer: a prospective study of 98 cases. J Thorac Cardiovasc Surg 111(2):323–331 Hornick JL, Mino-Kenudson M, Lauwers GY et al, 2008, Buried Barrett’s epithelium following photodynamic therapy shows reduced crypt proliferation and absence of DNA content abnormalities. Am J Gastroenterol 103(1):38–47 Younes M, Lebovitz RM, Lechago LV et al, 1993, p53 protein accumulation in Barrett’s metaplasia, dysplasia, and carcinoma: a follow-up study. Gastroenterology 105(6):1637–1642 Sun WH, Sun YL, Fang RN et al, 2005, Expression of cyclooxygenase-2 and matrix metalloproteinase-9 in gastric carcinoma and its correlation with angiogenesis. Jpn J Clin Oncol 35(12):707–713 Takeuchi T, Hisanaga M, Nagao M et al, 2004, The membraneanchored matrix metalloproteinase, MMP, regulator RECK in combination with MMP-9 serves as an informative prognostic indicator for colorectal cancer. Clin Cancer Res 10(16):5572–5579 Samantaray S, Sharma R, Chattopadhyaya TK et al, 2004, Increased expression of MMP-2 and MMP-9 in esophageal squamous cell carcinoma. J Cancer Res Clin Oncol 130(1):37–44 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 183 EP 192 DI 10.1007/s12253-008-9095-z PG 10 ER PT J AU Szarvas, T Jager, T Droste, F Becker, M Kovalszky, I Romics, I Ergun, S Rubben, H AF Szarvas, Tibor Jager, Tobias Droste, Falk Becker, Markus Kovalszky, Ilona Romics, Imre Ergun, Suleyman Rubben, Herbert TI Serum Levels of Angiogenic Factors and their Prognostic Relevance in Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; Serum; Angiopoietin; Tie2; VEGF; Prognosis ID Bladder cancer; Serum; Angiopoietin; Tie2; VEGF; Prognosis AB Angiogenesis plays a critical role in tumor growth. VEGF, angiopoietins (Ang-1, Ang-2) and their tyrosine kinase receptor Tie2 are major regulators of angiogenesis. The aim of this study was to evaluate the prognostic value of the serum levels of these factors in bladder cancer. We analyzed the serum samples of 117 bladder cancer patients and 64 healthy volunteers by enzyme linked immunosorbent assay (ELISA) for Ang-1, Ang-2, VEGF and the extracellular domain of Tie2. The statistical evaluation of the obtained data was performed via Kaplan–Meier log-rank test, univariate Cox analyses as well as Cox proportional hazards regression model. Serum Ang-1 levels of bladder cancer patients were significantly higher (p<0.001), while soluble Ang-2 and Tie2 levels were significantly lower (p=0.016 and p=0.001 respectively) in patients than those in controls. Cox univariate analysis revealed high sTie2 serum level as a risk factor for metastasis and as a borderline significant risk factor for disease related death (p=0.022 and p=0.081 respectively). These correlations were independent from tumor stage and grade in a Cox multivariate model (p=0.016 and p=0.069). These data indicate that the serum levels of analyzed angiogenic factors do change characteristically in bladder cancer. The soluble extracellular serum level of Tie2 may provide a stage and grade independent diagnostic tool to select a high risk group of bladder cancer patients. C1 [Szarvas, Tibor] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Jager, Tobias] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Droste, Falk] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Becker, Markus] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Romics, Imre] Semmelweis University, Department of UrologyBudapest, Hungary. [Ergun, Suleyman] University Hospital of Essen, Institute of AnatomyEssen, Germany. [Rubben, Herbert] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. RP Szarvas, T (reprint author), University of Duisburg-Essen, Department of Urology, 45147 Essen, Germany. EM sztibusz@gmail.com CR Weidner N, Semple JP, Welch WR, Folkman J, 1991, Tumor angiogenesisand metastasis correlation in invasive breast carcinoma. N Engl J Med 324:1–8 Davis S, Aldrich TH, Jones PF, Acheson A, Compton DL, Jain V, Ryan TE, Bruno J, Radziejewski C, Maisonpierre PC, Yancopoulos GD, 1996, Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expression cloning. Cell 87:1161–1169 Papapetropoulos A, Garcia-Cardena G, Dengler TJ, Maisonpierre PC, Yancopoulos GD, SessaWC, 1999, Direct actions of angiopoietin-1 on human endothelium: evidence for network stabilization, cell survival, and interaction with other angiogenic growth factors. Lab Invest 79:213–223 Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C, Compton D, McClain J, Aldrich TH, Papadopoulos N, Daly TJ, Davis S, Sato TN, Yancopoulos GD, 1997, Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 277:55–60 Holash J, Maisonpierre PC, Compton D, Boland P, Alexander CR, Zagzag D, Yancopoulos GD, Wiegand SJ, 1999, Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science 284:1994–1998 Volm M, Koomagi R, Mattern J, 1997, Prognostic value of vascular endothelial growth factor and its receptor flt-1 in squamous cell lung cancer. Int J Cancer 74:64–68 Inoue K, Ozeki Y, Suganuma T, Sugiura Y, Tanaka S, 1997, Vascular endothelial growth factor expression in primary esophageal squamous cell carcinoma. Cancer 79:206–213 Maeda K, Chung Y, Takatsuka S, Ogawa Y, Sawada T, Yamashita Y et al, 1995, Tumor angiogenesis as a predictor of recurrence in gastric carcinoma. J Clin Oncol 13:477–481 Dosquet C, Coudert MC, Lepage E, Cabane J, Richard F, 1997, Are angiogenic factors, cytokines, and soluble adhesion molecules prognostic factors in patients with renal cell carcinoma? Clin Cancer Res 3:2451–2458 Tas F, Duranyildiz D, Oguz H, Camlica H, Yasasever V, Topuz E, 2006, Circulating serum levels of angiogenic factors and vascular endothelial growth factor receptors 1 and 2 in melanoma patients. Melanoma Res 16:405–411 Bernardini S, Fauconnet S, Chabannes E, Henry PC, Adessi G, Bittard H, 2001, Serum levels of vascular endothelial growth factor as a prognostic factor in bladder cancer. J Urol 166:1275–1279 Sfiligoi C, de Luca A, Cascone I, Sorbello V, Fuso L, Ponzone R, Biglia N, Audero E, Arisio R, Bussolino F, Sismondi P, De BortoliM, 2003, Angiopoietin-2 expression in breast cancer correlates with lymph node invasion and short survival. Int J Cancer 103:466–474 Wurmbach JH, Hammerer P, Sevinc S, Huland H, Ergun S, 2000, The expression of angiopoietins and their receptor Tie-2 in human prostate carcinoma. Anticancer Res 20:5217–5220 Wong MP, Chan SY, Fu KH, Leung SY, Cheung N, Yuen ST, Chung LP, 2000, The angiopoietins, tie2 and vascular endothelial growth factor are differentially expressed in the transformation of normal lung to non-small cell lung carcinomas. Lung Cancer 29:11–22 Hata K, Nakayama K, Fujiwaki R, Katabuchi H, Okamura H, Miyazaki K, 2004, Expression of the angopoietin-1, angopoietin- 2, Tie2, and vascular endothelial growth factor gene in epithelial ovarian cancer. Gynecol Oncol 93:215–222 Szarvas T, Jager T, Totsch M, vom Dorp F, Kempkensteffen C, Kovalszky I, Romics I, Ergun S, Rubben H, 2008, Angiogenic switch of angiopietins-Tie2-system and its prognostic value in bladder cancer. Clin Cancer Res, in press) Liersch R, Schliemann C, Bieker R, Hintelmann H, Buechner T, Berdel WE, Mesters RM, 2008, Expression of VEGF-C and its receptor VEGFR-3 in the bone marrow of patients with acute myeloid leukaemia. Leuk Res 32:954–961 Park JH, Park KJ, Kim YS, Sheen SS, Lee KS, Lee HN, Oh YJ, Hwang SC, 2007, Serum angiopoietin-2 as a clinical marker for lung cancer. Chest 132:200–206 Lewis CE, De Palma M, Naldini L, 2007, Tie2-expressing monocytes and tumor angiogenesis: regulation by hypoxia and angiopoietin-2. Cancer Res 67:8429–8432 Reusch P, Barleon B, Weindel K, Martiny-Baron G, Godde A, Siemeister G, Marme D, 2001, Identification of a soluble form of the angiopoietin receptor TIE-2 released from endothelial cells and present in human blood. Angiogenesis 4:123–131 Eble JN, Epstein JI, Sesterhenn I, World Health Organization classification of tumors, 2004, Pathology and genetics of tumors of the urinary system and male genital organs. IARCC, Lyon, pp 89–158 Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC, Davis S, Sato TN, Yancopoulos GD, 1996, Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis. Cell 87:1171–1180 Holash J, Wiegand SJ, Yancopoulos GD, 1999, New model of tumor angiogenesis: dynamic balance between vessel regression and growth mediated by angiopoietins and VEGF. Oncogene 18:5356–5362 Crew JP, Fuggle S, Bicknell R, Cranston DW, de Benedetti A, Harris AL, 2000, Eukaryotic initiation factor-4E in superficial and muscle invasive bladder cancer and its correlation with vascular endothelial growth factor expression and tumour progression. Br J Cancer 82:161–166 O'Brien T, Cranston D, Fuggle S, Bicknell R, Harris AL, 1995, Different angiogenic pathways characterize superficial and invasive bladder cancer. Cancer Res 55:510–513 Quentin T, Schlott T, Korabiowska M, Kathei N, Zoller G, Glaser F, Kunze E, 2004, Alteration of the vascular endothelial growth factor and angiopoietins-1 and -2 pathways in transitional cell carcinomas of the urinary bladder associated with tumor progression. Anticancer Res 24:2745–2756 Oliveira-Ferrer L, Tilki D, Ziegeler G, Hauschild J, Loges S, Irmak S, Kilic E, Huland H, Friedrich M, Ergun S, 2004, Dual role of carcinoembryonic antigen-related cell adhesion molecule 1 in angiogenesis and invasion of human urinary bladder cancer. Cancer Res 64:8932–8938 Beecken WD, Engl T, Hofmann J, Jonas D, Blaheta R, 2005, Clinical relevance of serum angiogenic activity in patients with transitional cell carcinoma of the bladder. J Cell Mol Med 9:655– 661 Tait CR, Jones PF, 2004, Angiopoietins in tumours: the angiogenic switch. J Pathol 204:1–10 Oka N, Yamamoto Y, Takahashi M, Nishitani M, Kanayama HO, Kagawa S, 2005, Expression of angiopoietin-1 and -2, and its clinical significance in human bladder cancer. BJU Int 95:660– 663 Schliemann C, Bieker R, Thoennissen N, Gerss J, Liersch R, Kessler T, Buchner T, Berdel WE, Mesters RM, 2007, Circulating angiopoietin-2 is a strong prognostic factor in acute myeloid leukemia. Leukemia 21:1901–1906 Park JH, Park KJ, Kim YS, Sheen SS, Lee KS, Lee HN, Oh YJ, Hwang SC, 2007, Serum angiopoietin-2 as a clinical marker for lung cancer. Chest 132:200–206 Lewis CE, De Palma M, Naldini L, 2007, Tie2-expressing monocytes and tumor angiogenesis: regulation by hypoxia and angiopoietin-2. Cancer Res 67:8429–8432 Reusch P, Barleon B, Weindel K, Martiny-Baron G, Godde A, Siemeister G, Marme D, 2001, Identification of a soluble form of the angiopoietin receptor TIE-2 released from endothelial cells and present in human blood. Angiogenesis 4:123–131 Harris AL, Reusch P, Barleon B, Hang C, Dobbs N, Marme D, 2001, Soluble Tie2 and Flt1 extracellular domains in serum of patients with renal cancer and response to antiangiogenic therapy. Clin Cancer Res 7:1992–1997 Findley CM, Cudmore MJ, Ahmed A, Kontos CD, 2007, VEGF induces Tie2 shedding via a phosphoinositide 3-kinase/Akt dependent pathway to modulate Tie2 signaling. Arterioscler Thromb Vasc Biol 27:2619–2626 Lin P, Polverini P, Dewhirst M, Shan S, Rao PS, Peters K, 1997, Inhibition of tumor angiogenesis using a soluble receptor establishes a role for Tie2 in pathologic vascular growth. J Clin Invest 100:2072–2078 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 193 EP 201 DI 10.1007/s12253-008-9107-z PG 9 ER PT J AU Toy, H Yavas, O Eren, OO Genc, M Yavas, C AF Toy, Hatice Yavas, Ozlem Eren, Onder Orhan Genc, Mine Yavas, Cagdas TI Correliation Between Osteopontin Protein Expression and Histological Grade of Astrocytomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteopontin; Brain tumor; Grade; Astrocytomas ID Osteopontin; Brain tumor; Grade; Astrocytomas AB Osteopontin is a ligand for the integrin proteins, which are cell surface receptors that mediate the physical and functional interactions between a cell and the extracellular matrix. The expression of osteopontin is reportedly increased in a number of transformed cell lines and tumor tissues. Furthermore, increased expression of osteopontin results in some infiltrative features of tumors. The aim of the study is to demonstrate that expression of osteopontin in human astrocytomas correlates with histological tumor grade. The expression of osteopontin in human astrocytomas was determined with immunohistochemistry. Median osteopontin expression levels were 1%, 7.5%, 60%, and 50% in grade I, II, III, and IV tumors, respectively. Osteopontin staining was significantly higher in high grade (grade III–IV) than low grade (grade I–II) tumors. These findings indicate that osteopontin immunoreactivity in human astrocytomas may correlate with the grade of a tumor. C1 [Toy, Hatice] Selcuk University, Medicine Faculty, Department of Pathology, 42080 Konya, Meram, Turkey. [Yavas, Ozlem] Selcuk University, Medical OncologyKonya, Turkey. [Eren, Onder Orhan] Selcuk University, Medical OncologyKonya, Turkey. [Genc, Mine] Selcuk University, Radiation OncologyKonya, Turkey. [Yavas, Cagdas] Hacettepe University, Faculty of Medicine, Department of Radiation OncologyAnkara, Turkey. RP Toy, H (reprint author), Selcuk University, Medicine Faculty, Department of Pathology, 42080 Konya, Turkey. EM 11hatice@gmail.com CR Denhardt DT, Noda M, 1998, Osteopontin expression and function: role in bone modelling. J Cell Biochem Supply, 30– 31):92–102 Sodek J, Ganss B, McKee MD, 2000, Osteopontin. Crit Rev Oral Biol Med 11(3):279–303 Ritting SR, Chambers AF, 2004, Role of osteopontin in tumour progression. Br Journal Cancer 90:1877–1881 Weber GF, Ashkar S, Glimcher MJ, Cantor H, 1996, Receptorligand interaction between CD44 and osteopontin, Eta-1). Science 26, 271(5248):509–12 Senger DR, Wirth DF, Hynes RO, 1979, Transformed mammalian cells secrete specific proteins and phosphoproteins. Cell 16:885–893 Brown Brown LF, Papadopoulos-Sergiou A, Berse B, Manseau EJ, Tognazzi K, Perruzzi CA, Dvorak HF, Senger DR, 1994, Osteopontin expression and distribution in human carcinomas. Am J Pathol 145(3):610–23 Furger KA, Menon RK, Tuck AB, Bramwell VH, Chambers AF, 2001, The functional and clinical roles of osteopontin in cancer and metastasis. Curr Mol Med 1(5):621–32 Tucker MA, Chang PL, Prince CW, Gillespie GY, Mapstone TB, 1998, TPA-mediated regulation of osteopontin in human malignant glioma cells. Anticancer Res 18(2A):807–12 Chambers AF, Wilson SM, Kerkvliet N, O, Malley FP, Harris JF, Casson AG, 1996, Osteopontin expression in lung cancer. Lung Cancer 15(3):311–23 Coppola D, Szabo M, Boulware D, Muraca P, Alsarraj M, Chambers AF, Yeatman TJ, 2004, Correlation of osteopontin protein expression and pathological stage across a wide variety of tumor histologies. Clin Cancer Res 10(1 Pt 1):184–90 Rudland PS, Platt-Higgins A, El-Tanani M, De Silva Rudland S, Barraclough R, Winstanley JH, Howitt R, West CR, 2002, Prognostic significance of the metastasis-associated protein osteopontin in human breast cancer. Cancer Res 62(12):3417–27 Anita Bellail AC, Hunter SB, Brat DJ, Tan C, Van Meir EG, 2004, Microregional extracellular matrix heterogeneity in brain modulates glioma cell invasion. Int J Biochem Cell Biol 36, 6):1046–69 Roland Goldbrunner RH, Bernstein JJ, Tonn JC, 1998, ECMmediated glioma cell invasion. Microsc Res Tech 43(3):250–7 Saitoh Y, Kuratsu J, Takeshima H, Yamamoto S, Ushio Y, 1995, Expression of osteopontin in human glioma. Its correlation with the malignancy.. Lab Invest 72(1):55–63 Jang T, Savarese T, Low HP, Kim S, Vogel H, Lapointe D, Duong T, Litofsky NS, Weimann JM, Ross AH, Recht L, 2006, Osteopontin expression in intratumoral astrocytes marks tumor progression in gliomas induced by prenatal exposure to N-ethyl- N-nitrosourea. Am J Pathol 168(5):1676–85 Colin C, Baeza N, Bartoli C, Fina F, Eudes N, Nanni I, Martin PM, Ouafik L, Figarella-Branger D, 2005, Identification of genes differentially expressed in glioblastoma versus pilocytic astrocytoma using Suppression Subtractive Hybridization. Oncogene 12:1–9 Ding Q, Stewart J Jr, Prince CW, Chang PL, Trikha M, Han X, Grammer JR, Gladson CL, 2002, Promotion of malignant astrocytoma cell migration by osteopontin expressed in the normal brain: differences in integrin signaling during cell adhesion to osteopontin versus vitronectin. Cancer Res 62(18):5336–43 Said HM, Hagemann C, Staab A, Stojic J, Kuhnel S, Vince GH, Flentje M, Roosen K, Vordermark D, 2007, Expression patterns of the hypoxia-related genes osteopontin, CA9, erythropoietin, VEGF and HIF-1alpha in human glioma in vitro and in vivo. Radiother Oncol 83(3):398–405 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 203 EP 207 DI 10.1007/s12253-008-9130-0 PG 5 ER PT J AU Szendroi, M Antal, I Arato, G AF Szendroi, Miklos Antal, Imre Arato, Gabriella TI Adamantinoma of Long Bones: A Long-term Follow-up Study of 11 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adamantinoma; Long bones; Histology; Surgery ID Adamantinoma; Long bones; Histology; Surgery AB The aim of this study was to evaluate the clinicopathological features and prognostic significances of 11 histologically proven adamantinoma cases based on an average 12,7 year long follow-up. The male: female ratio was 8:3, aged between 4 and 80 years (mean 29,3 years). The initial diagnosis at referral was other than adamantinoma in six patients (fibrous dysplasia, carcinoma metastasis, osteofibrous dysplasia, bone cyst, nonossifying fibroma), referring to the differential diagnostic problems. All tumors were localized to the mid part of tibia. By histological evaluation, basaloid pattern on a background of fibrotic stroma dominated in six patients, while spindle and squamous features were less frequently seen. All adamantinomas were positive for cytokeratins often in coexpression with vimentin. No correlation was experienced between histology and clinical outcome. Intralesional curettage (2 pts) was followed by recurrence of the tumor. Wide resection was performed in eight patients with reconstruction using intercalary fibula autografts in seven patients. Reconstruction-related complications occurred in two third of the cases, all of them could however be controlled by repeated surgery. Six recurrences occurred in four patients, two of these recurrences occurred 20 and 16 years after initial surgery. One patient died 9 years after recognition of the tumor of pulmonary metastases. Adamantinoma of the long bones is a low grade malignant tumor, which clinical outcome is difficult to predict based on histology or surgical stage of the tumor. Wide surgical margin, e.g. resection the tumor reduces the rate of recurrence. This study underlines that recurrences do occur even decades after recognition the tumor, therefore a life-long follow-up of the patient is necessary. C1 [Szendroi, Miklos] Semmelweis University, Department of Orthopedics, 27 Karolina Rd, 1113 Budapest, Hungary. [Antal, Imre] Semmelweis University, Department of Orthopedics, 27 Karolina Rd, 1113 Budapest, Hungary. [Arato, Gabriella] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Szendroi, M (reprint author), Semmelweis University, Department of Orthopedics, 1113 Budapest, Hungary. EM szenmik@orto.sote.hu CR Fischer B, 1913, Uber ein primares Adamantinoma der Tibia. Virchows Arch 12-B:422–441 Dahlin DC, Unni KK, 1986, Bone tumors. General aspects and data on 8,542 cases. 4ed Charles C. Thomas, Spingfield Schajowicz F, 1981, Tumors and tumorlike lesions of bone and joints. Springer, New York, Heidelberg, Berlin Moon MF, 1994, Adamantinoma of the appendicular skeleton in children. Int Orthop 18:379–388 Campanacci M, 1990, Adamantinoma of the long bones. Bone and Soft Tissue Tumors, 629–638, Springer, New York Czerniak B, Rojas-Corona RR, Dorfman HD, 1989, Morphologic diversity of long bone adamantinoma. The concept of differentiated, regressing, adamantinoma and its relationship to osteofibrous dysplasia. Cancer 64:2319–2334 Huvos AG, Marcove RC, 1975, Adamantinoma of long bones. A clinicopathological study of fourteen cases with vascular origin suggested. J Bone and Joint Surg 57-A:148–154 Hazelbag HM, Taminiau AHM, Fleuren GJ et al, 1994, Adamantinoma of the long bones. A clinicopathological study of thirty-two patients with emphasis on histological subtype, precursor lesion, and biological behaviour. J Bone and Joint Surg 76-A:1482–1499 Qureshi AA, Shott S, Mallin BA et al, 2000, Current trends in the management of adamantinoma of long bones. An international study. J Bone Joint Surg 82-A:1122–1131 Keeney GL, Unni KK, Beabout JW et al, 1989, Adamantionoma of long bones. A clinicopathologic study of 85 cases. Cancer 64:730–737 Moon NF, Mori H, 1986, Adamantionoma of the appendicular skeleton - updated. Clin Orthop 204:215–237 Ulmar B, Delling G, Werner M et al, 2006, Classical and atypical location of adamantinomas - presentation of two cases. Onkologie 29:276–278 Papagelopoulos PJ, Mavrogenis A, Sim FH et al, 2007, Clinicopathological features, diagnosis and treatment of adamantinoma of the long bones. Orthopaedics 30(3):211–215 Huntington TW, 1905, Case of bone transference. Use of a segment of fibula to supply a defect in the tibia. Ann Surg 41:249–251 Hicks JD, 1954, Synovial sarcoma of the tibia. J Pathol and Bacteriol 67:151–161 Mori H, Yamamoto S, Hiramatsu K et al, 1984, Adamantinoma of the tibia. Ultrastructural and immunohistological study with reference to histogenesis. Clin Orthop 190:299–310 Pieterse AS, Smith PS, McClure J, 1982, Adamantinoma of long bones: clinical, pathological and ultrastructural features. J Clin Pathol 35:780–786 Hazelbag HM, Wessels JW, Mollevangers P et al, 1997, Cytogenetic analysis of adamantinoma of long bones: further indications for a common histogenesis with osteofibrous dysplasia. Cancer Genet and Cytogenet 97:5–11 Bovee JV, van den Brock LJ, de Boer WI et al, 1998, Expression of growth factors and their receptors in adamantionoma of long bones and the implications for its histogenesis. J Pathol 184:24–30 Dorfman HD, Czerniak B, 1997, Bone tumors. Mosby, St. Louis Povysil C, Matejovsky Z, 1981, Ultrastructure of adamantinoma of long bones. Virchows Archiv Pathol Anat 393:233–244 Kahn LB, 2003, Adamantinoma, osteofibrous dysplasia and differentiated adamantinoma. Skeletal Radiol 32:245–258 Kuruvilla G, Steiner GC, 1998, Osteofibrous dysplasia - like adamantinoma of bone: a report of five cases with immunohistochemical and ultrastructural studies. Hum Pathol 29:809–814 Maki M, Athanasou N, 2004, Osteofibrous dysplasia and adamantionoma: correlation of proto-oncogene product and matrix protein expression. Hum Pathol 35:69–74 Hatori M, Watanabe M, Hosaka M et al, 2006, A classic adamantinoma arising from osteofibrous dysplasia - like adamantionoma in the lower leg: a case report and review of the literature. Tohoku J Exp Med 209:53–59 Jundt G, Remberger K, Roessner A et al, 1995, Adamantionoma of long bones. A histopathological and immunohistochemical study of 23 cases. Pathol Res Pract 191:112–120 Szendroi M, Renyi-Vamos A, Marschalko P et al, 1994, Csoves csontok adamantinomainak viselkedese hosszu tavu utankovetes alapjan. Magy Traum Ortop 37:37–44 Bely M, Gal D, 1982, A case of adamantinoma localized to the tibia. Magyar Traumat 25:75 Bohndorf K, Nidecker A, Mathias K et al, 1992, Radiologische Befunde beim Adamantinom der langen Rohrenknochen. Fortschr Roentgenstr 157:239–244 Van der Woude HJ, Hazelbag HM, Bloem JL, Taminiau AH, Hogendoorn PC, 2004, MRI of adamantinoma of long bones in correlation with histopathology. Am J Roentgenol 183:1737–1744 Baker PL, Dockerty MB, Conventry MB, 1954, Adamantinoma, so-called, of the long bones. Preview of the literature and a report of three new cases. J Bone and Joint Surg 36-A:704–720 Gebhardt MC, Lord FC, Rosenberg AE et al, 1987, The treatment of adamantinoma of the tibia by wide resection and allograft bone transplantation. J Bone and Joint Surg 69-A:1177–1188 Deijkers RL, Bloem RM, Hogendoorn PC, Verlaan JJ, Kroon HM, Taminiau AH, 2002, Hemicortical allograft reconstruction after resection of low-grade malignant bone tumours. J Bone Joint Surg 84-B:1009–1114 Ortiz-Cruz E, Gebhardt MC, Jennings LC et al, 1997, The results of transplantation of intercalary allografts after resection of tumors. J Bone and Joint Surg 79-A:97–106 Rock MG, Beabout JW, Unni KK et al, 1983, Adamantinoma. Orthopaedics 6:472–477 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 209 EP 216 DI 10.1007/s12253-008-9125-x PG 8 ER PT J AU Zhao, E Xu, J Yin, X Sun, Y Shi, J Li, X AF Zhao, Eryang Xu, Jiankai Yin, Xiaodong Sun, Yu Shi, Jinna Li, Xia TI Detection of Deregulated Pathways to Lymphatic Metastasis in Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Deregulated pathway; Lymphatic metastasis; Oral squamous cell carcinomas ID Deregulated pathway; Lymphatic metastasis; Oral squamous cell carcinomas AB Oral squamous cell carcinoma (OSCC) is a common malignancy, in which lymph node metastasis is a major determinant of outcome. The pathway deregulation resulting from a large number of somatic genetic alterations in the development of the tumor, plays an important role in lymphatic metastasis process. To detect the deregulated pathways to lymphatic metastasis in OSCC, we performed pathway-oriented analysis using gene expression profile from 16 samples without lymphatic metastasis and 27 samples with lymphatic metastasis. We identified seven significantly (p<0.05) deregulated pathways: the erythropoietin (EPO) Signaling Pathway, Signaling Pathway from G-Protein Families, Cytokine–cytokine receptor interaction, the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway, Ribosome, Colorectal cancer, B cell receptor signaling pathway. The biological relevance of these pathways to OSCC is the focus of ongoing studies, as well as complex interactions and crosstalk between them. These pathways might provide additional clues about factors that regulate the course for OSCC patients and might offer new opportunities for therapeutic intervention. C1 [Zhao, Eryang] Harbin Medical University, The Stomatological CollegeHarbin, China. [Xu, Jiankai] Harbin Medical University, The Stomatological CollegeHarbin, China. [Yin, Xiaodong] Harbin Medical University, The Stomatological CollegeHarbin, China. [Sun, Yu] Harbin Medical University, The Stomatological CollegeHarbin, China. [Shi, Jinna] Harbin Medical University, The Stomatological CollegeHarbin, China. [Li, Xia] Harbin Medical University, The Stomatological CollegeHarbin, China. RP Li, X (reprint author), Harbin Medical University, The Stomatological College, Harbin, China. EM lixia@hrbmu.edu.cn CR Kalnins IK, Leonard AG, Sako K et al, 1977, Correlation between prognosis and degree of lymph node involvement in carcinoma of the oral cavity. Am J Surg 134:450–454 Greenberg JS, Fowler R, Gomez J et al, 2003, Extent of extracapsular spread: a critical prognosticator in oral tongue cancer. Cancer 97:1464–1470 Ramaswamy S, Ross KN, Lander ES, Golub TR, 2003, A molecular signature of metastasis in primary solid tumors. Nat Genet 33:49–54 Hunter K, Welch DR, Liu ET, 2003, Genetic background is an important determinant of metastatic potential. Nat Genet 34:23–24 author reply 25 Dahlquist KD, Salomonis N, Vranizan K et al, 2002, GenMAPP, a new tool for viewing and analyzing microarray data on biological pathways. Nat Genet 31:19–20 Toruner GA, Ulger C, Alkan M et al, 2004, Association between gene expression profile and tumor invasion in oral squamous cell carcinoma. Cancer Genet Cytogenet 154:27–35 O’Donnell RK, Kupferman M, Wei SJ et al, 2005, Gene expression signature predicts lymphatic metastasis in squamous cell carcinoma of the oral cavity. Oncogene 24:1244–1251 Irizarry RA, Bolstad BM, Collin F et al, 2003, Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res 31:e15 Mukherjee S, Roberts SJ, van der Laan MJ, 2005, Data-adaptive test statistics for microarray data. Bioinformatics 21(Suppl 2): ii108–114 Vaquerizas JM, Conde L, Yankilevich P et al, 2005, GEPAS, an experiment-oriented pipeline for the analysis of microarray gene expression data. Nucleic Acids Res 33:W616–620 Kanehisa M, Goto S, Kawashima S et al, 2004, The KEGG resource for deciphering the genome. Nucleic Acids Res 32: D277–280 Al-Shahrour F, Minguez P, Tarraga J et al, 2007, FatiGO+: a functional profiling tool for genomic data. Integration of functional annotation, regulatory motifs and interaction data with microarray experiments. Nucleic Acids Res 35:W91–96 Winter SC, Shah KA, Campo L et al, 2005, Relation of erythropoietin and erythropoietin receptor expression to hypoxia and anemia in head and neck squamous cell carcinoma. Clin Cancer Res 11:7614–7620 Mohyeldin A, Lu H, Dalgard C et al, 2005, Erythropoietin signaling promotes invasiveness of human head and neck squamous cell carcinoma. Neoplasia 7:537–543 Lai SY, Childs EE, Xi S et al, 2005, Erythropoietin-mediated activation of JAK-STAT signaling contributes to cellular invasion in head and neck squamous cell carcinoma. Oncogene 24:4442–4449 Dunlop EA, Percy MJ, Boland MP et al, 2006, Induction of signalling in non-erythroid cells by pharmacological levels of erythropoietin. Neurodegener Dis 3:94–100 Myklebust JH, Josefsen D, Blomhoff HK et al, 1999, Activation of the cAMP signaling pathway increases apoptosis in human Bprecursor cells and is associated with downregulation of Mcl-1 expression. J Cell Physiol 180:71–80 Srivastava RK, Srivastava AR, Cho-Chung YS, Longo DL, 1999, Synergistic effects of retinoic acid and 8-Cl-cAMP on apoptosis require caspase-3 activation in human ovarian cancer cells. Oncogene 18:1755–1763 Yan L, Herrmann V, Hofer JK, Insel PA, 2000, beta-adrenergic receptor/cAMP-mediated signaling and apoptosis of S49 lymphoma cells. Am J Physiol Cell Physiol 279:C1665–1674 Yang X, Lee FY Sr., Wand GS, 1997, Increased expression of Gs, alpha, enhances activation of the adenylyl cyclase signal transduction cascade. Mol Endocrinol 11:1053–1061 Collins MT, Sarlis NJ, Merino MJ et al, 2003, Thyroid carcinoma in the McCune–Albright syndrome: contributory role of activating Gs alpha mutations. J Clin Endocrinol Metab 88:4413–4417 Landis CA, Masters SB, Spada A et al, 1989, GTPase inhibiting mutations activate the alpha chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature 340:692–696 Levy L, Hill CS, 2006, Alterations in components of the TGFbeta superfamily signaling pathways in human cancer. Cytokine Growth Factor Rev 17:41–58 Roman C, Saha D, Beauchamp R, 2001, TGF-beta and colorectal carcinogenesis. Microsc Res Tech 52:450–457 Cao X, Eu KW, Kumarasinghe MP et al, 2006, Mapping of hereditary mixed polyposis syndrome, HMPS, to chromosome 10q23 by genomewide high-density single nucleotide polymorphism, SNP, scan and identification of BMPR1A loss of function. J Med Genet 43:e13 Heinrich PC, Behrmann I, Muller-Newen G et al, 1998, Interleukin-6-type cytokine signalling through the gp130/Jak/ STAT pathway. Biochem J 334(Pt 2):297–314 Tomida M, Yoshida U, Mogi C et al, 2001, Leukaemia inhibitory factor and interleukin 6 inhibit secretion of prolactin and growth hormone by rat pituitary MtT/SM cells. Cytokine 14:202–207 Acs G, Zhang PJ, Rebbeck TR et al, 2002, Immunohistochemical expression of erythropoietin and erythropoietin receptor in breast carcinoma. Cancer 95:969–981 Kisseleva T, Bhattacharya S, Braunstein J, Schindler CW, 2002, Signaling through the JAK/STAT pathway, recent advances and future challenges. Gene 285:1–24 Chiao PJ, Shin DM, Sacks PG et al, 1992, Elevated expression of the ribosomal protein S2 gene in human tumors. Mol Carcinog 5:219–231 Chen Y, Choong LY, Lin Q et al, 2007, Differential expression of novel tyrosine kinase substrates during breast cancer development. Mol Cell Proteomics 6:2072–2087 Li F, Cao Y, Townsend CM Jr., Ko TC, 2005, TGF-beta signaling in colon cancer cells. World J Surg 29:306–311 Clohisy JC, Roy BC, Biondo C et al, 2003, Direct inhibition of NF-kappa B blocks bone erosion associated with inflammatory arthritis. J Immunol 171:5547–5553 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 217 EP 223 DI 10.1007/s12253-008-9102-4 PG 7 ER PT J AU Toth, J Egervari, K Klekner, Bognar, L Szanto, J Nemes, Z Szollosi, Z AF Toth, Judit Egervari, Kristof Klekner, Almos Bognar, Laszlo Szanto, Janos Nemes, Zoltan Szollosi, Zoltan TI Analysis of EGFR Gene Amplification, Protein Over-expression and Tyrosine Kinase Domain Mutation in Recurrent Glioblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma multiforme; EGFR; Kinase inhibitor treatment ID Glioblastoma multiforme; EGFR; Kinase inhibitor treatment AB Gefitinib and erlotinib are both selective EGFR tyrosine kinase inhibitors (EGFR-TKIs) that have produced responses in a small subgroup of lung cancer patients. The strongest evidence for a role of EGFR in the biology of glioblastoma stems from clinical trials in which 15–20% of recurrent glioblastoma patients experienced significant tumour regression in response to these small-molecule EGFR kinase inhibitors. We examined the protein-kinase domain of the EGFR gene, EGFR protein expression and EGFR gene amplification in 20 cases of recurrent GBMs. EGFR protein over-expression was found in 65% of cases. EGFR protein over-expression was associated with EGFR gene amplification in 35% of cases, and with high polysomy in 15% of cases. No mutations were found in the TK domain of the EGFR gene. Our results confirm that mutations in the kinase domain are absent in recurrent GBM, and this might be a preponderant factor in the lack of major clinical responses of TKIs in GBM, recent studies have suggested that responsiveness to EGFR kinase inhibitors was strongly associated with coexpression of EGFRvIII and PTEN. Further prospective validation of EGFRvIII and PTEN as predictors of the clinical response to EGFR kinase inhibitors in recurrent GBM is strongly anticipated. C1 [Toth, Judit] University of Debrecen, Department of Oncology, Nagyerdei krt.98, 4012 Debrecen, Hungary. [Egervari, Kristof] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt.98, 4012 Debrecen, Hungary. [Klekner, Almos] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt.98, 4012 Debrecen, Hungary. [Bognar, Laszlo] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt.98, 4012 Debrecen, Hungary. [Szanto, Janos] University of Debrecen, Department of Oncology, Nagyerdei krt.98, 4012 Debrecen, Hungary. [Nemes, Zoltan] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt.98, 4012 Debrecen, Hungary. [Szollosi, Zoltan] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt.98, 4012 Debrecen, Hungary. RP Szollosi, Z (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, 4012 Debrecen, Hungary. EM szollosi@dote.hu CR Bruner JM, 1994, Neuropathology of malignant gliomas. Semin Oncol 21(2):126–138 Gilbert MR, Loghin M, 2005, The treatment of malignant gliomas. Curr Treat Options Neurol 7(4):293–303 Rich JN, Hans C, Jones B et al., 2005, Gene expression profiling and genetic markers in glioblastoma survival. Cancer Res. 65(10): 4051–4058 Nicholson RI, Gee JM, Harper ME, 2001, EGFR and cancer prognosis. Eur J Cancer 37(suppl 4):S9–S15 Lynch TJ, Bell DW, SordellaR et al., 2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350:2129–2139 Paez JG, Janne PA, Lee JC et al., 2004, EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304:1497–1500 Nathoo N, Goldlust S, Vogelbaum MA, 2004, Epidermal growth factor receptor antagonists: novel therapy for the treatment of high-grade gliomas. Neurosurgery 54:1480–1488 Barber TD, Vogelstein B, Kinzler KW et al., 2004, Somatic mutations of EGFR in colorectal cancers and glioblastomas. N Engl J Med 351:2883–2883 Kris MG, Natale RB, Herbst RS et al., 2003, Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial. JAMA 290:2149–2158 Fukuoka M, Yano S, Giaccone G et al., 2003, Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer, The IDEAL 1 Trial). J Clin Oncol 21:2237–2246 Mellinghoff IK, Wang MY, Vivanco I et al., 2005, Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med 353:2012–2024 Rich JN, Reardon DA, Peery T et al., 2004, Phase II trial of gefitinib in recurrent glioblastoma. J Clin Oncol 22:133– 142 Li B, Chang CM, Yuan M et al., 2003, Resistance to small molecule inhibitors of epidermal growth factor receptor in malignant gliomas. Cancer Res 63:7443–7450 Rich JN, Rasheed BK, Yan H, 2004, EGFR mutations and sensitivity to gefitinib. N Engl J Med 351:1260–1261 Aldape KD, Ballman K, Furth A et al., 2004, Immunohistochemical detection of EGFRvIII in high malignancy grade astrocytomas and evaluation of prognostic significance. J Neuropathol Exp Neurol 63:700–707 De Pas T, Pelosi P, de Braud F et al., 2004, Modulation of epidermal growth factor receptor status by chemotherapy in patients with locally advanced non-small-cell lung cancer is rare. J Clin Oncol 22:4966–4970 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 225 EP 229 DI 10.1007/s12253-008-9082-4 PG 5 ER PT J AU Fregnani, RE Sobral, ML Alves, AF Soares, AF Kowalski, PL Coletta, DR AF Fregnani, Rodrigues Eduardo Sobral, M Lays Alves, Abreu Fabio Soares, Augusto Fernando Kowalski, Paulo Luis Coletta, D Ricardo TI Presence of Myofibroblasts and Expression of Matrix Metalloproteinase-2 (MMP-2) in Ameloblastomas Correlate with Rupture of the Osseous Cortical SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ameloblastoma; Matrix metalloproteinase-2; Myofibroblast; Prognosis; Urokinase plasminogen activator ID Ameloblastoma; Matrix metalloproteinase-2; Myofibroblast; Prognosis; Urokinase plasminogen activator AB Myofibroblasts are frequent in the stroma of neoplasm and by the expression of proteinases they can influence tumor infiltration and progression. In the present study, presence of myofibroblasts and expression of matrix metalloproteinase-2 (MMP-2) and urokinase plasminogen activator (uPA) were examined in intra-osseous solid multicystic ameloblastomas to determine their roles in the clinicopathological features of the tumors. Fifty seven ameloblastomas were analyzed immunohistochemically with antibodies against the isoform α of the smooth muscle actin (α-SMA), a specific marker of myofibroblasts, MMP-2 and uPA. Myofibroblasts were found in the stroma, in close contact with neoplastic cell islands, of ~58% (n=33) of the ameloblastomas. MMP-2 and uPA were found in the cytoplasm of both neoplastic and stromal cells. A significant correlation between presence of myofibroblasts and MMP-2 expression was observed. Abundant presence of myofibroblast in the stroma of the tumors and expression of MMP-2 in the neoplastic or stromal cells were significantly correlated with rupture of the osseous cortical, which has been considered an important prognostic marker of ameloblastoma aggressiveness. Ours results suggest that abundant presence of myofibroblasts and expression of MMP-2 in solid ameloblastomas may be associated with a more aggressive infiltrative behavior. C1 [Fregnani, Rodrigues Eduardo] A. C. Camargo Cancer Hospital, Department of Stomatology, R. Professor Antonio Prudente, 211 Sao Paulo-SP-Liberdade-CEP, 01509-010 Sao Paulo, Brazil. [Sobral, M Lays] State University of Campinas, School of Dentistry, Department of Oral DiagnosisPiracicaba, Sao Paulo, Brazil. [Alves, Abreu Fabio] A. C. Camargo Cancer Hospital, Department of Stomatology, R. Professor Antonio Prudente, 211 Sao Paulo-SP-Liberdade-CEP, 01509-010 Sao Paulo, Brazil. [Soares, Augusto Fernando] A. C. Camargo Cancer Hospital, Department of PathologySao Paulo, Brazil. [Kowalski, Paulo Luis] A. C. Camargo Cancer Hospital, Department of Head and Neck Surgery and OtorhinolaryngologySao Paulo, Brazil. [Coletta, D Ricardo] State University of Campinas, School of Dentistry, Department of Oral DiagnosisPiracicaba, Sao Paulo, Brazil. RP Fregnani, RE (reprint author), A. C. Camargo Cancer Hospital, Department of Stomatology, 01509-010 Sao Paulo, Brazil. EM erfreg@terra.com.br;eduardofregnani@me.com CR Desmouliere A, Chaponnier C, Gabbiani G, 2005, Tissue repair, contraction, and the myofibroblast. Wound Repair Regen 13:7–12 Desmouliere A, Guyot C, Gabbiani G, 2004, The stroma reaction myofibroblast: a key player in the control of tumor cell behavior. Int J Dev Biol 48:509–517 Kuroda N, Shimasaki N,Miyazaki E et al., 2006, The distribution of myofibroblasts and CD34-positive stromal cells in normal renal pelvis and ureter and their cancers. Histol Histopathol 21:1303–1307 Powell DW, Adegboyega PA, Di Mari JF et al., 2005, Epithelial cells and their neighbors I. Role of intestinal myofibroblasts in development, repair, and cancer. Am J Physiol Gastrointest Liver Physiol 289:G2–G7 Darby IA, Hewitson TD, 2007, Fibroblast differentiation in wound healing and fibrosis. Int Rev Cytol 257:143–179 Bitu CC, Sobral LM, Kellermann MG et al., 2006, Heterogeneous presence of myofibroblasts in hereditary gingival fibromatosis. J Clin Periodontol 33:393–400 Gabbiani G, 2003, The myofibroblast in wound healing and fibrocontractive diseases. J Pathol 200:500–503 Kunz-Schughart LA, Knuechel R, 2002, Tumor-associated fibroblasts, part II): Functional impact on tumor tissue. Histol Histopathol 17:623–637 Kunz-Schughart LA, Knuechel R, 2002, Tumor-associated fibroblasts, part I): Active stromal participants in tumor development and progression? Histol Histopathol 17:599–621 Scotton CJ, Chambers RC, 2007, Molecular targets in pulmonary fibrosis: the myofibroblast in focus. Chest 132:1311–1321 Mukaratirwa S, Koninkx JF, Gruys E et al., 2005, Mutual paracrine effects of colorectal tumour cells and stromal cells: modulation of tumour and stromal cell differentiation and extracellular matrix component production in culture. Int J Exp Pathol 86:219–229 Offersen BV, Nielsen BS, Hoyer-Hansen G et al., 2003, The myofibroblast is the predominant plasminogen activator inhibitor- 1-expressing cell type in human breast carcinomas. Am J Pathol 163:1887–1899 Orimo A, Weinberg RA, 2006, Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle 5:1597–1601 Helary C, Ovtracht L, Coulomb B et al., 2006, Dense fibrillar collagen matrices: a model to study myofibroblast behaviour during wound healing. Biomaterials 27:4443–4452 Soini Y, Satta J, Maatta M et al., 2001, Expression of MMP2, MMP9, MT1-MMP, TIMP1, and TIMP2 mRNA in valvular lesions of the heart. J Pathol 194:225–231 Pender SL,MacDonald TT, 2004, Matrix metalloproteinases and the gut - new roles for old enzymes. Curr Opin Pharmacol 4:546–550 Hatane T, Yoshida E, Kawano J et al., 1998, Prostaglandin I2 analog enhances the expression of urokinase-type plasminogen activator and wound healing in cultured human fibroblast. Biochim Biophys Acta 1403:189–198 Dubuisson L, Monvoisin A, Nielsen BS et al., 2000, Expression and cellular localization of the urokinase-type plasminogen activator and its receptor in human hepatocellular carcinoma. J Pathol 190:190–195 Monvoisin A, Neaud V, De Ledinghen V et al., 1999, Direct evidence that hepatocyte growth factor-induced invasion of hepatocellular carcinoma cells is mediated by urokinase. J Hepatol 30:511–518 Plekhanova OS, Stepanova VV, Ratner EI, 2006, Urokinase plasminogen activator in injured adventitia increases the number of myofibroblasts and augments early proliferation. J Vasc Res 43:437–446 Reichart PA, Philipsen HP, Sonner S, 1995, Ameloblastoma: biological profile of 3677 cases. Eur J Cancer B Oral Oncol 31B:86–99 Kumamoto H, Ooya K, 2006, Immunohistochemical detection of MT1-MMP, RECK, and EMMPRIN in ameloblastic tumors. J Oral Pathol Med 35:345–351 Kumamoto H, Ooya K, 2007, Immunohistochemical detection of uPA, uPAR, PAI-1, and maspin in ameloblastic tumors. J Oral Pathol Med 36:488–494 Kumamoto H, Yamauchi K, Yoshida M et al., 2003, Immunohistochemical detection of matrix metalloproteinases, MMPs, and tissue inhibitors of metalloproteinases, TIMPs, in ameloblastomas. J Oral Pathol Med 32:114–120 Pinheiro JJ, Freitas VM, Moretti AI et al, 2004, Local invasiveness of ameloblastoma. Role played by matrix metalloproteinases and proliferative activity. Histopathology 45:65–72 Suh KS, Crutchley JM, Koochek A et al., 2007, Reciprocal modifications of CLIC4 in tumor epithelium and stroma mark malignant progression of multiple human cancers. Clin Cancer Res 13:121–131 Kellermann MG, Sobral LM, da Silva SD, Zecchin KG, Graner E, Lopes MA, Kowalski LP, Coletta RD, 2008, Mutual paracrine effects of oral squamous cell carcinoma cells and normal oral fibroblasts: induction of fibroblast to myofibroblast transdifferentiation and modulation of tumor cell proliferation. Oral Oncol 4:509–517 Mahadevan D, Von Hoff DD, 2007, Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol Cancer Ther 6:1186–1197 Casey TM, Eneman J, Crocker A et al, 2007, Cancer associated fibroblasts stimulated by transforming growth factor beta1, TGF-beta1, increase invasion rate of tumor cells: a population study. Breast Cancer Res Treat Aug 3., DOI 10.1007/ s10549-007-9684-7 Kellermann MG, Sobral LM, da Silva SD et al., 2007, Myofibroblasts in the stroma of oral squamous cell carcinoma are associated with poor prognosis. Histopathology 51:849–853 Lewis MP, Lygoe KA, Nystrom ML et al., 2004, Tumour-derived TGF-beta1 modulates myofibroblast differentiation and promotes HGF/SF-dependent invasion of squamous carcinoma cells. Br J Cancer 90:822–832 Chauhan H, Abraham A, Phillips JR et al., 2003, There is more than one kind of myofibroblast: analysis of CD34 expression in benign, in situ, and invasive breast lesions. J Clin Pathol 56:271–276 Shimasaki N, Kuroda N, Miyazaki E et al., 2006, The distribution pattern of myofibroblasts in the stroma of human bladder carcinoma depends on their invasiveness. Histol Histopathol 21:349–353 Takata T, Miyauchi M, Ogawa I et al., 2000, Immunoexpression of transforming growth factor beta in desmoplastic ameloblastoma. Virchows Arch 436:319–323 Smith SM, Bartov SA, 1986, Ameloblastoma with myofibroblasts: first report. J Oral Pathol 15:284–286 Rothouse LS, Majack RA, Fay JT, 1980, An ameloblastoma with myofibroblasts and intracellular septate junctions. Cancer 45:2858–2863 Lombardi T, Morgan PR, 1995, Immunohistochemical characterisation of odontogenic cysts with mesenchymal and myofilament markers. J Oral Pathol Med 24:170–176 Vered M, Shohat I, Buchner A et al., 2005, Myofibroblasts in stroma of odontogenic cysts and tumors can contribute to variations in the biological behavior of lesions. Oral Oncol 41:1028–1033 Carlson ER, Marx RE, 2006, The ameloblastoma: primary, curative surgical management. J Oral Maxillofac Surg 64:484–494 Ueno S, Mushimoto K, Shirasu R, 1989, Prognostic evaluation of ameloblastoma based on histologic and radiographic typing. J Oral Maxillofac Surg 47:11–15 Sampson DE, Pogrel MA, 1999, Management of mandibular ameloblastoma: the clinical basis for a treatment algorithm. J Oral Maxillofac Surg 57:1074–1077; discussion 78–9 McKaig BC, McWilliams D, Watson SA et al., 2003, Expression and regulation of tissue inhibitor of metalloproteinase-1 and matrix metalloproteinases by intestinal myofibroblasts in inflammatory bowel disease. Am J Pathol 162:1355–1360 Czochra P, Klopcic B, Meyer E et al., 2006, Liver fibrosis induced by hepatic overexpression of PDGF-B in transgenic mice. J Hepatol 45:419–428 Seomun Y, Kim J, Lee EH et al., 2001, Overexpression of matrix metalloproteinase-2 mediates phenotypic transformation of lens epithelial cells. Biochem J 358:41–48 Porter KE, Turner NA, O’Regan DJ et al., 2004, Tumor necrosis factor alpha induces human atrial myofibroblast proliferation, invasion and MMP-9 secretion: inhibition by simvastatin. Cardiovasc Res 64:507–515 Deng X, He G, Levine A et al., 2008, Adenovirus-mediated expression of TIMP-1 and TIMP-2 in bone inhibits osteolytic degradation by human prostate cancer. Int J Cancer 122:209–218 Dass K, Ahmad A, Azmi AS et al., 2008, Evolving role of uPA/ uPAR system in human cancers. Cancer Treat Rev 34:122–136 Fisher JL,Mackie PS, Howard ML et al., 2001, The expression of the urokinase plasminogen activator system in metastatic murine osteosarcoma: an in vivo mouse model. Clin Cancer Res 7:1654–1660 Stillfried GE, Saunders DN, Ranson M, 2007, Plasminogen binding and activation at the breast cancer cell surface: the integral role of urokinase activity. Breast Cancer Res 9:R14 Nielsen BS, Rank F, Illemann M et al., 2007, Stromal cells associated with early invasive foci in human mammary ductal carcinoma in situ coexpress urokinase and urokinase receptor. Int J Cancer 120:2086–2095 Ji F, Chen YL, Jin EY et al., 2005, Relationship between matrix metalloproteinase-2 mRNA expression and clinicopathological and urokinase-type plasminogen activator system parameters and prognosis in human gastric cancer. World J Gastroenterol 11:3222–3226 Bernstein AM, Twining SS, Warejcka DJ et al., 2007, Urokinase receptor cleavage: a crucial step in fibroblast-to-myofibroblast differentiation. Mol Biol Cell 18:2716–2727 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 231 EP 240 DI 10.1007/s12253-008-9110-4 PG 10 ER PT J AU El-Sarha, IA Magour, MG Zaki, MS El-Sammak, YM AF El-Sarha, I Ashgan Magour, M Gehan Zaki, M Sameh El-Sammak, Y Mohamed TI Serum sFas and Tumor Tissue FasL Negatively Correlated with Survival in Egyptian Patients Suffering from Breast Ductal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE sFas; FasL; Apoptosis; DNA ploidy; S-phase fraction ID sFas; FasL; Apoptosis; DNA ploidy; S-phase fraction AB Fas (CD95-APO-1), a member of tumor necrosis factor receptor super-family, exists in two forms, transmembrane and soluble (sFas). It had been suggested that circulating sFas levels and/or tissue FasL may reflect the severity of invasive breast ductal carcinoma. Few studies showed that neither DNA-index nor ploidy is an independent prognostic indicator, and there is no correlation with clinical outcome. The S-phase fraction (SPF) has been shown to be useful prognostic factor in both node-negative and node-positive tumors. The present work was done to find a correlation between sFas, tissue FasL, ploidy and SPF with prognostic factors and survival of breast ductal carcinoma patients. The present study included two groups; a patients group comprised 30 patients with breast ductal carcinoma and a control group that comprised 15 patients with benign breast swellings. Serum sFas was measured using commercially available ELISA kit and tissue FasL expression was studied using avidin–biotine immunohistochemical staining technique. Cell cycle studies were performed using flow cytometry. Serum sFas was significantly higher in breast ductal carcinoma group than in the benign breast swelling control group. A significant negative correlation between serum sFas and overall survival was found. Tissue FasL expression was directly correlated with distant metastasis and poor overall survival. A significant direct correlation was found between moderate and high SPF with worse pathologic parameters. Serum sFas level, tissue FasL immuno-expression and S-phase fraction are independent prognostic factors in breast ductal carcinoma cases. C1 [El-Sarha, I Ashgan] Alexandria University, Medical Research Institute Teaching Hospital, Department of Chemical PathologyAlexandria, Egypt. [Magour, M Gehan] Alexandria University, Medical Research Institute Teaching Hospital, Department of Chemical Pathology, 165 El Horreya StreetAlexandria, Egypt. [Zaki, M Sameh] Alexandria University, Medical Research Institute, Department of Radiation SciencesAlexandria, Egypt. [El-Sammak, Y Mohamed] Alexandria University, Medical Research Institute Teaching Hospital, Department of Chemical Pathology, 165 El Horreya StreetAlexandria, Egypt. RP El-Sammak, YM (reprint author), Alexandria University, Medical Research Institute Teaching Hospital, Department of Chemical Pathology, Alexandria, Egypt. EM myelsammak@doctors.org.uk CR Reed JC, 2000, Mechanisms of apoptosis. Am J Pathol 156:1415–1430 Saas P, Walker PR, Hahne M et al, 1997, Fas ligand expression by astrocytoma in vivo: maintaining immune privilege in the brain? J Clin Invest 99:1173–1178 Schneioer P, Bodmer JL, Holler N et al, 1997, Characterization of Fas, Apo-1, CD95)-Fas ligand interaction. J Biol Chem 272:18827–18833 Griffith TS, Ferguson TA, 1997, The role of FasL induced apoptosis in immune privilege. Immunol Today 18:2240–2244 Muschen M, Moers C, Warskulat U, Even J, Niederacher D, Beckmann MW, 2000, CD95 ligand expression as a mechanism of immune escape in breast cancer. Immunology 99(1):69–77 Sheen-Chen SM, Chen HS, Eng HL, Chen WJ, 2003, Circulating soluble Fas in patients with breast cancer. World J Surg 27(1):10– 13, Jan Suda T, Takahashi T, Golstein P, Nagata S, 1993, Molecular cloning and expression of the Fas ligand: a novel member of the tumor necrosis factor family. Cell 75:1169–1186 Keane MM, Ettenberg SA, Lowrey GA, Russell EK, Lipkowitz S, 1996, Fas expression and function in normal and malignant breast cell lines. Cancer Res 56:4791–4798 Harrnring C, Reimer T, Jeschke U, Makovitzky J, Kruger K, Gerber B, Kabelitz D, Friese K, 2000, Expression of the apoptosis-inducing ligands FasL and TRAIL in malignant and benign human breast tumors. Histochem Cell Biol 113:189–194 Mullauer L, Mosberger I, Grusch M, Rudas M, Chott A, 2000, Fas ligand is expressed in normal breast epithelial cells and is frequently upregulated in breast cancer. J Pathol 190:20–30 Mann B, Gratchev A, Bohm C et al, 1999, FasL is more frequently expressed in liver metastases of colorectal cancer than in matched primary carcinomas. Br J Cancer 79:1262–1269 Munakata S, Enomoto T, Tsujimoto M et al, 2000, Expression of Fas ligand and other apoptosis-related genes and their prognostic significance in epithelial ovarian neoplasms. Br J Cancer 82:1446–1452 Nagashima H, Mori M, Sadanaga N et al, 2001, Expression of Fas ligand in gastric carcinoma relates to lymph node metastasis. Int J Oncol 18:1157–1162 Munakata S, Watanabe O, Ohashi K, Morino H, 2005, Expression of Fas ligand and bcl-2 in cervical carcinoma and their prognostic significance. Am J Clin Pathol 123(6):879–885 Gatierrez LS, Eliza M, Niven-Fairchild T, Naftolin F, Mor G, 1999, The Fas/Fas-ligand system: a mechanism for immune evasion in human breast carcinomas. Breast Cancer Res Treat 54, 3):245–253 Robert I, 2002, New concepts for the study of anticancer drug resistance [French]. Bull. Cancer 89:17–22 Hortobagyi GN, 2001, Adjuvant systemic therapy for early breast cancer: progress and controversies. Clin Cancer Res 7:1839–1842 Ross JS, Linette GP, Stec J, Ross MS, Anwar S, Boguniewicz A, 2003, DNA ploidy and cell cycle analysis in breast cancer. Am J Clin Pathol 120(Suppl):S72–S84,, ISSN: 0002-9173) Chavez-Uribe EM, Vinuela JE, Cameselle-Teijeiro J, Forteza J, Punal JA, Otero IL, Puente-Dominguez JL, 2002, DNA ploidy and cytonuclear area of peritumoral and paratumoral samples of mastectomy specimens: a useful prognostic marker? Eur J Surg 168(1):37–41 Chassevent A, Jourdan ML, Romain S et al, 2001, S-phase fraction and DNA ploidy in 633 T1T2 breast cancers: a standardized flow cytometric study. Clin Cancer Res 7(4):909– 917,, ISSN: 1078-0432) Greene FL, Page DL, Fleming ID et al, 2002, Breast IN: American joint committee on cancer, AJCC cancer staging manual, 6th edn. Springer, New York, p 171 Burtis CA, Ashwood ER, Bruns DE, 2006, Teitz text book of clinical chemistry and molecular diagnostics, 4th edn. Elsevier Saunders, St Louis, pp 870–871, 797–803, 582–590, 546–549, 604–611, 1892–1904 & 1698 Bouillon R, Coopman SW, Degroote DEH, Eliard PH, 1990, Immunoradiometric assay of parathyrin polyclonal and monoclonal region specific antibodies. Clin Chem 36, 2):271–276 Chen J, Zhou T, Liu C, Shapiro JP, Brauuer MJ, Kieler MC, 1994, Protection from Fas-mediated apoptosis by a soluble form of the Fas molecule. Science 263:1759–1762 Chamness GC, McGuire WL, 1979, Methods for analyzing steroid receptors in human breast cancer. In: McGuire WL, ed, Breast cancer. Advances in research and treatment. Plenum, New York, p 149 Bebenek M, Dus D, Kozlak J, 2006, Fas and Fas ligand as prognostic factors in human breast carcinoma. Med Sci Monit 12, 11):CR457–CR461 Almasri NM, Al Hamad M, 2005, Immunohistochemical evaluation of human epidermal growth factor receptor 2 and estrogen and progesterone receptors in breast carcinoma in Jordan. Breast Cancer Res 7:R598–R604 Feichter GE, Muller A, Kaufmann M et al, 1988, Correlation of DNA flow cytometric results and other prognostic factors in primary breast cancer. Int J Cancer 41:823–828 Feichter GE, Kaufmann M, Muller A et al, 1989, DNA index and cell cycle analysis of primary breast cancer and synchronous axillary lymph node metastases. Breast Cancer Res Treat 13:17–22 Konrey GG, Pegram MD, Beryt M et al, 1999, Therapeutic advantage of chemotherapy drugs in combination with herceptin against human breast cancer cells with HER-2/neu overexpression. Breast Cancer Res Treat 57:114 Kev TJ, Verkasalo PK, Banks E, 2001, Epidemiology in breast cancer. Lancet Oncol 2(3):133–140 Reeves GK, Beral V, Green J, Gathan T, Bull D, 2006, Hormonal therapy of menopause and breast-cancer risk by histological type: a cohort study and meta-analysis. Lancet Oncol 7(11):910–918 Jager JJ, Jansen RL, Arendo JW, 2002, Clinical relevance of apoptotic markers in breast cancer is not yet clear. Apoptosis 7:361–365 Zhang L, Levi E, Majumder P, Yu Y, Aboukameel A, Du J, 2007, Transactivator of transcription-tagged cell cycle and apoptosis regulatory protein-1 peptides suppress the growth of human breast cancer cells in vitro and in vivo. Mol Cancer Ther 6:1661–1672 Motyl T, Gajkowska B, Zarzy J, Gajewska M, Lamparska P, 2006, Apoptosis and autophagy in mammary gland remodeling and breast cancer chemotherapy. J Physiol Pharmacol 7:17–32 Sheen-Chen SM, Chen HS, Eng HL, Chenw J, 2003, Circulating soluble Fas in patients with breast cancer. World J Surg 27:10–13 Djerbi M, Screpanti Y, Catrini A et al, 1999, The inhibitor of death receptor signaling. FLICE inhibitory protein defines a new class of tumor progression factors. J Exp Med 190:1025–1031 Nemesansky E, Tasnadi K, Juhasz P, 1971, Diagnostic value of alkaline phosphatase isoenzyme in cancer patients. Cancer 27:1388–1397 Kao P, Cmklee GG, Taylor R, Heath H, 1990, Parathyroid hormone related peptide in plasma of patients with hypercalcaemia and malignant lesions. Mayo Clin Proc 65:1399– 1407 Koomagi R, Volm M, 1999, Expression of Fas, CD95/APO-1, and Fas ligand in lung cancer, its prognostic and predictive relevance. Int J Cancer 84:239–243 Nagao M, Nakajima Y, Hisanaga M et al, 1999, The alteration of Fas receptor and ligand system in hepatocellular carcinoma: how do hepatoma cells escape from the host immune surveillance in vivo? Hepatology 30:413–421 Shibakita M, Tachibana M, Dahr DK et al, 1999, Prognostic significance of Fas and Fas ligand expression in human esophageal cancer. Clin Cancer Res 5:2464–2469 Ohno S, Tachibana M, Shibakita M et al, 2000, Prognostic significance of Fas and Fas ligand system-associated apoptosis in gastric cancer. Ann Surg Oncol 7:750–757 Mottolese M, Buglioni S, Bracalenti C et al, 2000, Prognostic relevance of altered Fas, CD95)-system in human breast cancer. Int J Cancer 89:127–132 Bebenek M, Dus D, Kozlak J, 2007, Fas/Fas-ligand expressions in primary breast cancer are significant predictors of its skeletal spread. Anticancer Res 27(1A):215–218 Sjostrom J, Blomqvist C, Boguslawski K et al, 2002, The predictive value of bcl-2, bcl-xL, bag-1, Fas, and FasL for chemotherapy response in advanced breast cancer. Clin Cancer Res 8:811–816 Kute TE, Quadri Y, Muss H et al, 1995, Flow cytometry in nodepositive breast cancer: cancer and leukemia group B protocol 8869. Cytometry 22:297–306 Bryant J, Fisher B, Gunduz N et al, 1998, S-phase fraction combined with other patient and tumor characteristics for the prognosis of node-negative, estrogen-receptor positive breast cancer. Breast Cancer Res Treat 51:239–253 Toikkanen S, Pylkkanen L, Joensuu H, 1997, Invasive lobular carcinoma of the breast has better short- and long-term survival then invasive ductal carcinoma. Br J Cancer 76:1234–1240 Remvikos Y, Magdelenat H, Dutrillaux B, 1995, Genetic evolution of breast cancers. III. Age-dependent variations in the correlations between biological indicators of prognosis. Breast Cancer Res Treat 34:25–33 Leonardi E, Cristofori A, Caffo O, Dalla Palma P, 1997, Cytometric DNA analysis and prognostic biomarkers in breast carcinoma. Expression of P53 product in the different ploidy classes. Anal Cell Pathol 15:31–45 Dettmar P, Harbeck N, Thomssen C et al, 1997, Prognostic impact of proliferation-associated factors MIB1, Ki67, and sphase fraction in node-negative breast cancer. Br J Cancer 75:1525–1533 Rudolph P, Alm P, Heidebrech H et al, 1999, Immunologic proliferation marker Ki-S2 as prognostic indicator for lymph node-negative breast cancer. Natl Cancer Inst, Bethesda, 91:271– 278 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 241 EP 250 DI 10.1007/s12253-008-9109-x PG 10 ER PT J AU Kohut, E Hajdu, M Gergely, P Gopcsa, L Kilian, K Paloczi, K Kopper, L Sebestyen, A AF Kohut, Eszter Hajdu, Melinda Gergely, Peter Gopcsa, Laszlo Kilian, Katalin Paloczi, Katalin Kopper, Laszlo Sebestyen, Anna TI Expression of TGFβ1 and Its Signaling Components by Peripheral Lymphocytes in Systemic Lupus Erythematosus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Autoimmune; Signaling; Smad; SLE; TGFβ ID Autoimmune; Signaling; Smad; SLE; TGFβ AB Transforming growth factor β1 (TGFβ1) is an important immunosuppressive cytokine. Defects in its production by lymphocytes and the failure of TGFβ1 to regulate immunological functions have been described in SLE. Expression of TGFβ1 and the related signaling pathway was studied in the peripheral lymphocytes of SLE patients. The total plasma TGFβ1 level in active and inactive SLE patients compared to healthy controls was also measured. TGFβ1 and all downstream signaling elements were expressed in normal cells. However, in more than 50% of SLE patients the isolated Tcell population showed no TGFβ1mRNA expression and at least one member of the TGFβ1 pathway was also missing (TGFβ-RI, Smad2 and Smad3) in more than half of the patients. Total plasma TGFβ1 level was increased in both active and inactive SLE groups compared to normal controls (p< 0.05). These data raise questions about the availability of TGFβ1 signaling in lymphocytes in SLE patients, however, the elevated total plasma TGFβ1 level suggests that the failure of TGFβ1 effects is not the consequence of low level of this cytokine in SLE. C1 [Kohut, Eszter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Hajdu, Melinda] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Gergely, Peter] Semmelweis University, Department of Laboratory MedicineBudapest, Hungary. [Gopcsa, Laszlo] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Kilian, Katalin] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Paloczi, Katalin] National Medical Center, Department of Hematology and Stem Cell TransplantationBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. RP Sebestyen, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM anna@korb1.sote.hu CR Greidinger E, 2001, Apoptosis in lupus pathogenesis. Front Biosci 6:1302–1325 Mageed RA, Prud’homme GJ, 2003, Immunopathology and the gene therapy of lupus. Gene Therapy 10:861–874 Manson JJ, Isenberg DA, 2003, The pathogenesis of systemic lupus erythematosus. Neth J Med 61:343–346 Mok CC, Lau CS, 2003, Pathogenesis of systemic lupus erythematosus. J Clin Pathol 56:481–490 Derynck R, Zhang Ying E, 2003, Smad-dependent and Smadindependent pathways in TGFb family signaling. Nature 425:577– 584 Jackson M, Ahmad Y, Bruce IN, Coupes B, Brenchley P, 2006, Activation of transforming growth factor- β1 and early atherosclerosis in systemic lupus erythematosus. Arthritis Research and Therapy 8:R81 Horwitz DA, Gray D, Ohtsuka K, 1999, Role of NK cells and TGFb in the regulation of T-cell-dependent antibody production in health and autoimmune disease. Microbes Infect 1:1305–1311 Lebman DA, Edmiston JS, 1998, The role of TGFb in growth, differentiation and maturation of B lymphocytes. Microbes Infect 1:1297–304 Schuster N, Krieglstein K, 2002, Mechanisms of TGFb-mediated apoptosis. Cell Tissue Res 307:1–14 Totth A, Sebestyen A, Barna G, Nagy K, Gondor A, Bocsi J, Mihalik R, Petak I, Houghton J, Kopper L, 2001, TGFb induces caspase-dependent but death-receptor independent apoptosis in lymphoid cells. Anticancer Res 21:1207-1212 Sakaguchi S, Sakaguchi N, Takahashi T, 2001, Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol Rev 182:18 Zheng SG, Wang JH, Gray JD, Soucier H, Horwitz DA, 2004, Natural and induced CD4+CD25+ cells educate CD4+CD25- cells to develop suppressive activity: the role of IL-2, TGFb and IL-10. J Immunol 172:5213–5221 Horwitz DA, Gray D, Ohtsuka K, Hirokawa M, Takahashi T, 1997, The immunoregulatory effects of NK cells: the role of TGFb and implications for autoimmunity. Immunology Today 18:538–542 Derynck R, Akhurst RJ, Balmain A, 2001, TGFb signaling in tumor suppression and cancer progression. Nature Genetics 29:117–129 Massague J, Gomis RR, 2006, The logic of TGFb signaling. FEBS Letters 580:2811–2820 Ohtsuka K, Gray D, Stimmler MM, Toro B, Horwitz DA, 1998, Decreased production of TGFb by Lymphocytes from Patients with Systemic Lupus Erythematosus. J Immunol 160:2539–2545 Linker-Israeli M, Quismorio FP Jr, Horwitz DA, 1990, CD8+ lymphocytes from patients with systemic lupus erythematosus sustain rather than suppress spontaneous polyclonal IgG production and synergize with CD4+ cells to support autoantibody synthesis. Arthritis Rheum 33:1216–1225 Liu MF, Wang CR, Fung LL, Wu CR, 2004, Decreased CD4 +CD25+ T cells in Peripheral Blood of Patients with Systemic Lupus Erythematosus. Scand J Immunol 59:198–202 Miyara M, Amoura Z, Parizot C, Badoual C, Dorgham K, Trad S, 2005, Global natural regulatory T cell depletion in active systemic lupus erythematosus. J Immunol 175:8392–8400 Tan EM, Cohen AS, Fries JF, Masi AT, McShane DJ, Rothfield NF, Schaller JG, Talal N, Winchester RJ, 1982, The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 25:1271 Bombardier CD, Gladman D, Urowitz MB, Caron D, Chang CH, 1992, Derivation of the SLEDAI, a disease activity index for lupus patients. The Committee on Prognosis Studies in SLE. Arthritis Rheum 35:630–640 Lu L, Cheng H, Sung P, Yeh J, Shiue Y, Chen A, 2004, Singlenucleotide polymorphisms of transforming growth factor-β1 gene in Taiwanese patients with systemic lupus erythematosus. J Microbiol Immunol Infect 37:145–152 Schotte H, Willeke P, Rust S, Assmann G, Domschke W, Gaubitz M, Schluter B, 2003, The transforming growth factor-β1 gene polymorphism, G915C, in not associated with systemic lupus erythematosus. Lupus 12:86–92 Higashi T, Kyo S, Inoue M, Tani H, Saijoh K, 2006, Novel functional single nucleotide polymorphisms in the latent transforming growth factor-β1 binding protein-1 L promoter: effect on latent latent transforming growth factor-β1 binding protein-1 L expression level and possible prognostic significance in ovarian cancer. J Mol Diagn 8:342–350 McKnight AJ, Savage DA, Patterson CC, Sadlier D, Maxwell AP, 2007, Resequencing of genes for transforming growth factor β1 type 1 and 2 receptors and association analysis of variants with diabetic nephropathy. BMC Medical Genetics 8:5 Robak E, Wozniacka A, Sysa-Jedrzejowska A, Stepien H, Robak T, 2001, Serum levels of angiogenic cytokines in systemic lupus erythematosus and their correlation with disease activity. Eur Cytokine Netw 12:445–452 Ruiz-Arguelles GJ, Ruiz-Arguelles A, Lobato-Mendizabal E, Diaz-Gomez F, Pacheco-Pantoja E, Denkard C, Alarcon-Segovia D, 1991, Disturbances in the tissue plasminogen activator/ plasminogen inhibitor, TPA/PAI, system in systemic lupus erythematosus. Am J Hematol 37:9 Cuchacovich R, Singh R, Cuellar M, Espinoza LR, 2001, Distinct pattern of gene expression by DNA microarray in peripheral blood mononuclear cells of rheumatoid arthritis and psoriatc arthritis patients treated with TNF-antagonists or methotrexate. Arthritis Rheumatism 44(suppl):2949 Drynda S, Kuhne C, Kekow J, 2002, Soluble TNF receptor treatment does not affect raised TGFb levels in rheumatoid arthritis. Ann Rheum Dis 61:667–669 Sjowall C, Ernerudh J, Bengtsson AA, Sturfelt G, Skogh T, 2004, Reduced anti- TNFα autoantibody levels coincide with flare in systemic lupus erythematosus. J Autoimmun 22:315– 323 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 251 EP 256 DI 10.1007/s12253-008-9119-8 PG 6 ER PT J AU Bagheri, K Alimoghadam, K Pourfathollah, AA Hassan, MZ Hajati, J Moazzeni, MS AF Bagheri, Kambiz Alimoghadam, Kamran Pourfathollah, Akbar Ali Hassan, Muhammad Zuhair Hajati, Jamshid Moazzeni, Mohammad Seyyed TI The Efficient Generation of Immunocompetent Dendritic Cells from Leukemic Blasts in Acute Myeloid Leukemia: A Local Experience SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Dendritic cell; Differentiation; Immunotherapy; Iranian AML patients; Leukemia ID Dendritic cell; Differentiation; Immunotherapy; Iranian AML patients; Leukemia AB Dendritic cells (DCs) are the most important antigen presenting cells with potentially useful applications in cancer immunotherapy. Leukemic cells of patients with acute myeloid leukemia (AML) could be differentiated to DC-like cells possessing the ability of stimulating antileukemic immune response. Despite obvious progress in DC-based immunotherapy, some discrepancies were reported in differentiation potential of AML blasts from all patients toward DC like cells. The present study, as a local experience, was set up to generate DCs from AML blasts of various subtypes. Leukemic Blasts from 16 Iranian AML patients were differentiated into functional DCs by culturing in the presence of rhGM-CSF, rhIL-4 and TNFalpha for 8 days. The morphology, expression of key surface molecules and allostimulatory activity of resultant DCs were compared with primary blasts and cultured but cytokine untreated control groups. The pattern of angiotensin-converting enzyme (ACE) expression was used to approve the leukemic origin of generated DCs. Neoexpression or upregulation of DC-associated markers were occurred during culturing period in cytokine treated cells compared with primary blasts and cultured but cytokine untreated control groups: CD1a (63.22% vs. 3.22% and 11.79%), CD83 (41.27% vs. 0.11% and 0.70%), CD40 (15.17% vs. 0.00% and 0.04%), CD80 (49.96 vs. 0.02% and 0.32%), CD86 (56.49% vs. 0.50% and 5.71%) and HLA-DR (52.52% vs. 14.32% and 2.49%) respectively. The potency of generated DCs to induce allogeneic T cell proliferation increased significantly compared to pre and post culture control groups (27,533.4±2,548.3, 8,820.4±1,639.4 and 3,200.35±976 respectively). The expression pattern of ACE in AML-DCs, blast cells and DCs derived from normal monocytes (7.93%, 1.28% and 74.97% respectively) confirmed the leukemic origin of DCs. Our data confirmed the generation of sufficient AML-derived cells with the properties of DCs in all cases. This potency of AML blasts, offers a useful route for active immunotherapy of AML patients. C1 [Bagheri, Kambiz] Tarbiat Modares University, Faculty of Medical Sciences, Department of ImmunologyTehran, Iran. [Alimoghadam, Kamran] Tehran University of Medical Sciences, Research Center, Shariati Hospital, Hematology, Oncology and Bone Marrow TransplantationTehran, Iran. [Pourfathollah, Akbar Ali] Tarbiat Modares University, Faculty of Medical Sciences, Department of ImmunologyTehran, Iran. [Hassan, Muhammad Zuhair] Tarbiat Modares University, Faculty of Medical Sciences, Department of ImmunologyTehran, Iran. [Hajati, Jamshid] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. [Moazzeni, Mohammad Seyyed] Tarbiat Modares University, Faculty of Medical Sciences, Department of ImmunologyTehran, Iran. RP Moazzeni, MS (reprint author), Tarbiat Modares University, Faculty of Medical Sciences, Department of Immunology, Tehran, Iran. EM Moazzeni@modares.ac.ir;smoazzeni@gmail.com CR Wang ZY, Chen Z, 2008, Acute promyelocytic leukemia: from highly fatal to highly curable. Blood 111:2505–2515 Mato AR, Morgans A, Luger SM, 2008, Novel strategies for relapsed and refractory acute myeloid leukemia. Curr Opin Hematol 15:108–114 Capra M, Vilella L, Pereira WV, Coser VM, Fernandes MS, Schilling MA, Almeida D, Gross M, Leite M, Hellwig T, Natchigal G, Zelmanowicz A, Paskulin G, Neumann J, Silla L, 2007, Estimated number of cases, regional distribution and survival of patients diagnosed with acute myeloid leukemia between 1996 and 2000 in Rio Grande do Sul, Brazil. Leuk Lymphoma 48:2381–2386 Pulte D, Gondos A, Brenner H, 2008, Improvements in survival of adults diagnosed with acute myeloblastic leukemia in the early 21st century. Haematologica 93:594–600 Yanada M, Garcia-Manero G, Borthakur G, Ravandi F, Kantarjian H, Estey E, 2008, Relapse and death during first remission in acute myeloid leukemia. Haematologica 93:633–634 Kim HR, Shin JH, Lee JN, Lee EY, 2007, Clinical significance of quantitation of WT1 gene expression for minimal residual disease monitoring of acute myelogenous leukemia. Korean J Lab Med 27:305–312 Ho MM, Hogge DE, Ling V, 2008, MDR1 and BCRP1 expression in leukemic progenitors correlates with chemotherapy response in acute myeloid leukemia. Exp Hematol 36:433–442 Hussein K, Jahagirdar B, Gupta P, Burns L, Larsen K, Weisdorf D, 2008, Day 14 bone marrow biopsy in predicting complete remission and survival in acute myeloid leukemia. Am J Hematol 83(6):446–450 Ommen HB, Nyvold CG, Braendstrup K, Andersen BL, Ommen IB, Hasle H, Hokland P, Ostergaard M, 2008, Relapse prediction in acute myeloid leukaemia patients in complete remission using WT1 as a molecular marker: development of a mathematical model to predict time from molecular to clinical relapse and define optimal sampling intervals. Br J Haematol 141(6):782–791 Aversa F, 2008, Haploidentical haematopoietic stem cell transplantation for acute leukaemia in adults: experience in Europe and the United States. Bone Marrow Transplant 41:473–481 Hamadani M, Awan FT, Copelan EA, 2008, Hematopoietic stem cell transplantation in adults with acute myeloid leukemia. Biol Blood Marrow Transplant 14:556–567 Fefer A, Sullivan KM, Weiden P, Buckner CD, Schoch G, Storb R, Thomas ED, 1987, Graft versus leukemia effect in man: the relapse rate of acute leukemia is lower after allogeneic than after syngeneic marrow transplantation. Prog Clin Biol Res 244:401– 408 Huang XJ, Wang Y, Liu DH, Xu LP, Chen H, Chen YH, Han W, Shi HX, Liu KY, 2008, Modified donor lymphocyte infusion, DLI, for the prophylaxis of leukemia relapse after hematopoietic stem cell transplantation in patients with advanced leukemia— feasibility and safety study. J Clin Immunol 28:390–397 Slavin S, Ackerstein A, Naparstek E, Or R, Weiss L, 1990, The graft-versus-leukemia, GVL, phenomenon: is GVL separable from GVHD? Bone Marrow Transplant 6:155–161 Ge X, Brown J, Sykes M, Boussiotis VA, 2008, CD134- allodepletion allows selective elimination of alloreactive human T cells without loss of virus-specific and leukemia-specific effectors. Biol Blood Marrow Transplant 14:518–530 Hartwig UF, Nonn M, Khan S, Link I, Huber C, Herr W, 2008, Depletion of alloreactive donor T lymphocytes by CD95-mediated activation-induced cell death retains antileukemic, antiviral, and immunoregulatory T cell immunity. Biol Blood Marrow Transplant 14:99–109 Buggins AG, Milojkovic D, Arno MJ, Lea NC, Mufti GJ, Thomas NS, Hirst WJ, 2001, Microenvironment produced by acute myeloid leukemia cells prevents T cell activation and proliferation by inhibition of NF-kappaB, c-Myc, and pRb pathways. J Immunol 167:6021–6030 Narita M, Takahashi M, Liu A, Nikkuni K, Furukawa T, Toba K, Koyama S, Takai K, Sanada M, Aizawa Y, 2001, Leukemia blastinduced T-cell anergy demonstrated by leukemia-derived dendritic cells in acute myelogenous leukemia. Exp Hematol 29:709–719 Orleans-Lindsay JK, Barber LD, Prentice HG, Lowdell MW, 2001, Acute myeloid leukaemia cells secrete a soluble factor that inhibits T and NK cell proliferation but not cytolytic function— implications for the adoptive immunotherapy of leukaemia. Clin Exp Immunol 126:403–411 Duluc D, Delneste Y, Tan F, Moles MP, Grimaud L, Lenoir J, Preisser L, Anegon I, Catala L, Ifrah N, Descamps P, Gamelin E, Gascan H, Hebbar M, Jeannin P, 2007, Tumor-associated leukemia inhibitory factor and IL-6 skew monocyte differentiation into tumor-associated macrophage-like cells. Blood 110:4319– 4330 Hirano N, Takahashi T, Ohtake S, Hirashima K, Emi N, Saito K, Hirano M, Shinohara K, Takeuchi M, Taketazu F, Tsunoda S, Ogura M, Omine M, Saito T, Yazaki Y, Ueda R, Hirai H, 1996, Expression of costimulatory molecules in human leukemias. Leukemia 10:1168–1176 Costello RT, Mallet F, Sainty D, Maraninchi D, Gastaut JA, Olive D, 1998, Regulation of CD80/B7-1 and CD86/B7-2 molecule expression in human primary acute myeloid leukemia and their role in allogenic immune recognition. Eur J Immunol 28:90–103 Bruserud O, 1999, Acute myelogenous leukemia blasts as accessory cells during T lymphocyte activation: possible implications for future therapeutic strategies. Leukemia 13:1175–1187 Adams S, O'Neill DW, Bhardwaj N, 2005, Recent advances in dendritic cell biology. J Clin Immunol 25:87–98 Greiner J, Dohner H, Schmitt M, 2006, Cancer vaccines for patients with acute myeloid leukemia—definition of leukemiaassociated antigens and current clinical protocols targeting these antigens. Haematologica 91:1653–1661 Heinzinger M, Waller CF, von den Berg A, Rosenstiel A, Lange W, 1999, Generation of dendritic cells from patients with chronic myelogenous leukemia. Ann Hematol 78:181–186 Eisendle K, Lang A, Eibl B, Nachbaur D, Glassl H, Fiegl M, Thaler J, Gastl G, 2003, Phenotypic and functional deficiencies of leukaemic dendritic cells from patients with chronic myeloid leukaemia. Br J Haematol 120:63–73 Takahashi T, Tanaka Y, Nieda M, Azuma T, Chiba S, Juji T, Shibata Y, Hirai H, 2003, Dendritic cell vaccination for patients with chronic myelogenous leukemia. Leuk Res 27:795–802 Tong XM, Jin J, Qian WB, Meng HT, Xue YQ, 2005, Biological features of dendritic cells derived from chronic myeloid leukemia cells in vitro. Zhejiang Da Xue Xue Bao Yi Xue Ban 34:348–352, 357 Zhang YF, Wu CY, Zhang LS, Chai Y, 2006, In vitro cytokinesinduced differentiation in mononuclear cell derived dendritic cells from chronic myeloid leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 14:137–141 Kohler T, Plettig R, Wetzstein W, Schmitz M, Ritter M, Mohr B, Schaekel U, Ehninger G, Bornhauser M, 2000, Cytokine-driven differentiation of blasts from patients with acute myelogenous and lymphoblastic leukemia into dendritic cells. Stem Cells 18:139–147 Oehler L, Berer A, Kollars M, Keil F, Konig M, Waclavicek M, Haas O, Knapp W, Lechner K, Geissler K, 2000, Culture requirements for induction of dendritic cell differentiation in acute myeloid leukemia. Ann Hematol 79:355–362 Claxton D, Choudhury A, 2001, Potential for therapy with AMLderived dendritic cells. Leukemia 15:668–669 Yan KH, You SG, Bian SG, Ma GJ, Ge W, Ma S, Liu SH, Zhao CH, 2003, Dendritic cells, DC, induced from acute myeloid leukemia, AML, cells with cytokine cocktails. Zhonghua Xue Ye Xue Za Zhi 24:365–368 Houtenbos I, Westers TM, Ossenkoppele GJ, van de Loosdrecht AA, 2006, Leukemia-derived dendritic cells: towards clinical vaccination protocols in acute myeloid leukemia. Haematologica 91:348–355 Robinson SP, English N, Jaju R, Kearney L, Knight SC, Reid CD, 1998, The in-vitro generation of dendritic cells from blast cells in acute leukaemia. Br J Haematol 103:763–771 Choudhury BA, Liang JC, Thomas EK, Flores-Romo L, Xie QS, Agusala K, Sutaria S, Sinha I, Champlin RE, Claxton DF, 1999, Dendritic cells derived in vitro from acute myelogenous leukemia cells stimulate autologous, antileukemic T-cell responses. Blood 93:780–786 Brouwer RE, van der Hoorn M, Kluin-Nelemans HC, van Zelderen-Bhola S, Willemze R, Falkenburg JH, 2000, The generation of dendritic-like cells with increased allostimulatory function from acute myeloid leukemia cells of various FAB subclasses. Hum Immunol 61:565–574 Harrison BD, Adams JA, Briggs M, Brereton ML, Yin JA, 2001, Stimulation of autologous proliferative and cytotoxic T-cell responses by “leukemic dendritic cells” derived from blast cells in acute myeloid leukemia. Blood 97:2764–2771 Roddie PH, Horton Y, Turner ML, 2002, Primary acute myeloid leukaemia blasts resistant to cytokine-induced differentiation to dendritic-like leukaemia cells can be forced to differentiate by the addition of bryostatin-1. Leukemia 16:84–93 Almand B, Resser JR, Lindman B, Nadaf S, Clark JI, Kwon ED, Carbone DP, Gabrilovich DI, 2000, Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res 6:1755–1766 Bello-Fernandez C, Stasakova J, Renner A, Carballido-Perrig N, Koening M, Waclavicek M, Madjic O, Oehler L, Haas O, Carballido JM, Buschle M, Knapp W, 2003, Retrovirus-mediated IL-7 expression in leukemic dendritic cells generated from primary acute myelogenous leukemias enhances their functional properties. Blood 101:2184–2190 Orsini E, Guarini A, Chiaretti S, Mauro FR, Foa R, 2003, The circulating dendritic cell compartment in patients with chronic lymphocytic leukemia is severely defective and unable to stimulate an effective T-cell response. Cancer Res 63:4497–4506 Van der Reijden HJ, van Rhenen DJ, Lansdorp PM, van’t Veer MB, Langenhuijsen MM, Engelfriet CP, von dem Borne AE, 1983, A comparison of surface marker analysis and FAB classification in acute myeloid leukemia. Blood 61:443–448 Neame PB, Soamboonsrup P, Browman GP, Meyer RM, Benger A, Wilson WE, Walker IR, Saeed N, McBride JA, 1986, Classifying acute leukemia by immunophenotyping: a combined FAB-immunologic classification of AML. Blood 68:1355–1362 Danilov SM, Sadovnikova E, Scharenborg N, Balyasnikova IV, Svinareva DA, Semikina EL, Parovichnikova EN, Savchenko VG, Adema GJ, 2003, Angiotensin-converting enzyme, CD143, is abundantly expressed by dendritic cells and discriminates human monocyte-derived dendritic cells from acute myeloid leukemiaderived dendritic cells. Exp Hematol 31:1301–1309 Conti L, Cardone M, Varano B, Puddu P, Belardelli F, Gessani S, 2008, Role of the cytokine environment and cytokine receptor expression on the generation of functionally distinct dendritic cells from human monocytes. Eur J Immunol 38:750–762 Hassan HT, Zander A, 1996, Stem cell factor as a survival and growth factor in human normal and malignant hematopoiesis. Acta Haematol 95:257–262 Caux C, Saeland S, Favre C, Duvert V, Mannoni P, Banchereau J, 1990, Tumor necrosis factor-alpha strongly potentiates interleukin- 3 and granulocyte-macrophage colony-stimulating factor-induced proliferation of human CD34+ hematopoietic progenitor cells. Blood 75:2292–2298 Mackensen A, Herbst B, Chen JL, Kohler G, Noppen C, Herr W, Spagnoli GC, Cerundolo V, Lindemann A, 2000, Phase I study in melanoma patients of a vaccine with peptide-pulsed dendritic cells generated in vitro from CD34(+, hematopoietic progenitor cells. Int J Cancer 86:385–392 Brunner C, Seiderer J, Schlamp A, Bidlingmaier M, Eigler A, Haimerl W, Lehr HA, Krieg AM, Hartmann G, Endres S, 2000, Enhanced dendritic cell maturation by TNF-alpha or cytidinephosphate- guanosine DNA drives T cell activation in vitro and therapeutic anti-tumor immune responses in vivo. J Immunol 165:6278–6286 Morel Y, Truneh A, Sweet RW, Olive D, Costello RT, 2001, The TNF superfamily members LIGHT and CD154, CD40 ligand, costimulate induction of dendritic cell maturation and elicit specific CTL activity. J Immunol 167:2479–2486 Cignetti A, Bryant E, Allione B, Vitale A, Foa R, Cheever MA, 1999, CD34(+, acute myeloid and lymphoid leukemic blasts can be induced to differentiate into dendritic cells. Blood 94:2048– 2055 Bagheri K, Delirezh N, Moazzeni SM, 2008, PPD extract induces the maturation of human monocyte-derived dendritic cells. Immunopharmacol Immunotoxicol 30:91–104 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 257 EP 267 DI 10.1007/s12253-008-9105-1 PG 11 ER PT J AU Kovacs, Hadjiev, J Lakosi, F Antal, G Vandulek, Cs Somogyine Ezer, Bogner, P Horvath, Repa, I AF Kovacs, Arpad Hadjiev, Janaki Lakosi, Ferenc Antal, Gergely Vandulek, Csaba Somogyine Ezer, Eva Bogner, Peter Horvath, Agnes Repa, Imre TI Dynamic MR Based Analysis of Tumor Movement in Upper and Mid Lobe Localized Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Dynamic MR; Tumor motion ID Lung cancer; Dynamic MR; Tumor motion AB Background and purpose: Tumor motion is a very important factor in the radiotherapy of lung cancer. Uncertainty resulting from tumor movement must be considered in 3D therapy planning especially in case of IMRT or stereotactic therapy. The aim of our dynamic MR based study was to detect tumor movements in upper and mid lobe lung tumors. Patient and methods: Twenty-four patients with newly diagnosed stage II-IV lung cancer were enrolled into the study. According to tumor localization in the right S1–S3 segments 9, in the right S4–S6 segments 2, in the left S1–S3 segments 9 and in the left S4–S6 segments 4 lesions were detected. In normal treatment position individual dynamic MR examinations were performed in axial, sagittal and coronal planes (100 slices/30 sec). For tumor motion analysis E-RAD PAC's software was used. Results: Movements of the tumor under normal breathing conditions were registered in the three main directions. The mean antero-posterior deviation was 0,109 cm (range: 0,063 cm–0,204 cm), the mean mediolateral deviation was 0,114 cm (range: 0,06 cm– 0,244 cm). The greatest deviation was measured in cranio–caudal direction (mean: 0,27 cm, range: 0,079 cm– 0,815 cm). The mean direction independent deviation was 0,18 cm (range: 0,09 cm– 0,48 cm). Conclusion: Dynamic MR is a sensitive and well tolerated method for tumor motion monitoring in high precision 3D therapy planning of lung cancer patients. Our results demonstrate that tumors located in the upper and mid lobes have moderate breath synchronous movements. The greatest deviation occur in cranio–caudal direction. C1 [Kovacs, Arpad] Kaposi Mor Teaching Hospital, Guba S. u 40, H-7400 Kaposvar, Hungary. [Hadjiev, Janaki] Kaposi Mor Teaching Hospital, Guba S. u 40, H-7400 Kaposvar, Hungary. [Lakosi, Ferenc] Kaposi Mor Teaching Hospital, Guba S. u 40, H-7400 Kaposvar, Hungary. [Antal, Gergely] Kaposi Mor Teaching Hospital, Guba S. u 40, H-7400 Kaposvar, Hungary. [Vandulek, Csaba] Kaposi Mor Teaching Hospital, Guba S. u 40, H-7400 Kaposvar, Hungary. [Somogyine Ezer, Eva] Kaposi Mor Teaching Hospital, Tallian Gy. u. 20–32, H-7400 Kaposvar, Hungary. [Bogner, Peter] Kaposi Mor Teaching Hospital, Guba S. u 40, H-7400 Kaposvar, Hungary. [Horvath, Agnes] University of Debrecen, Medical and Health Science Center, Institute of Nuclear Medicine, Nagyerdei krt 98, H-4012 Debrecen, Hungary. [Repa, Imre] Kaposi Mor Teaching Hospital, Guba S. u 40, H-7400 Kaposvar, Hungary. RP Horvath, (reprint author), University of Debrecen, Medical and Health Science Center, Institute of Nuclear Medicine, H-4012 Debrecen, Hungary. EM horvakos@freemail.hu CR Bollmann A, Blankenburg T, Haerting J, Kuss O, Schutte W, Dunst J, Neef H, 2004, on behalf of the HALLUCA Study group. Survival of patients in clinical stages I-IIIb of non-small-cell lung cancer treated with radiation therapy alone. Strahlenther Onkol 8:488–496 Lagerwaard FJ, Senan S, van Meerbeck JP et al, 2002, Has 3D conformal radiotherapy improved the local control in stage I nonsmall cell lung cancer? Radiother Oncol 63:151–157 Kohz P, Stabler A, Beinert T et al, 1995, Reproducibility of quantitative controlled CT. Radiol 197:539–542 Weiss E, Hess CF, 2003, The impact of gross tumor volume, GTV, and clinical target volume, CTV, definition on the total accuracy in radiotherapy. Strahlenther Onkol 1:21–30 Armstrong J, 1998, Target definition for three dimensional radiation therapy of lung cancer. BR J Radiol 846:539–542 Ekberg L, Holmberg O, Wittgren L, Bjelkengren G, Landberg T, 1998, What margins should be added to the clinical target volume in radiotherapy treatment planning for lung cancer? Radiother Oncol 48(1):71–77 Jul Shimizu S, Shirato H, Kagei K et al, 2000, Impact of respiratory movement on the computed tomographic images of small lung tumors in three-dimensional, 3D, radiotherapy. Int J Radiat Oncol Biol Phys 46:1127–1133 Stevens C, Munden R, Forester K et al, 2001, Respiratory-driven lung tumor motion in depend of tumor size, tumor location, and pulmonary function. Int J Radiat Oncol Biol Phys 51:62–68 Gauthier AP, Verbanck S, Estenne M et al, 1994, Threedimensional reconstruction of the in vivo human diaphragm shape at different lung volumes. J Appl Physiol 76:495–506 Gierada DS, Curtin JJ, Erickson SJ et al, 1995, Diaphragmatic motion: fast gradient-recalled-echo MR imaging in healthy subjects. Radiology 194:879–884 Napadow VJ, Mai V, Bankier A et al, 2001, Determination of regional pulmonary parenchymal strain during normal respiration using spin inversion tagged magnetization MRI. J Magn Reson Imaging 13:467–474 International Commission on Radiation Units and Measurements.: ICRU Report 62. Prescribing, recording, and reporting photon beam therapy, Supplement to ICRU Report 50, Bethesda, MD, 1999, by Wambersie A, Landberg T Kubo HD, Hill BC, 1996, Respiration gated radiotherapy treatment: a technical study. Phys Med Biol 41:83–91 Lagerwaard FJ, van Sornsen de Koste JR, Lagerwaard FJ et al, 2001, Multiple “slow” CT scans for incorporating lung tumor mobility in radiotherapy planning. Int J Radiat Oncol Biol Phys 51:932–937 Seppenwoolde Y, Shirato H, Kitamura K et al, 2002, Precise and real-time measurement of 3D tumor motion in lung due to breathing and heartbeat, measured during radiotherapy. Int J Radiat Oncol Biol Phys 53:822–834 de Boer HC, van Sornsen de Koste JR, Senan S et al, 2001, Analysis and reduction of 3D systematic and random setup errors during the simulation and treatment of lung cancer patients with CT-based external beam radiotherapy dose planning. Int J Radiat Oncol Biol Phy 49:857–868 Hof H, Herfarth KK, Munter M et al, 2003, Stereotactic singledose radiotherapy of stage I non-small-cell lung cancer, NSCLC). Int J Radiat Oncol Biol Phys 56:335–341 Noel G, Sarrazin T, Mirabel X, Prevost B, 1997, Use of real-time system of portal imaging in the daily monitroing of patients treated by radiotherapy for thoracic cancer. Cancer Radiother 1:249–257 Kovacs A, Hadjiev J, Lakosi F, Vallyon M, Cselik Z, Bogner P, Horvath A, Repa I, 2007, Thermoplastic patient fixation: influence on chest wall and target motion during radiotherapy of lung cancer. Strahlenther Onkol 183(5):271–278 May Giraud P, De Rycke Y, Dubray B et al, 2001, Conformal radiotherapy, CRT, planning for lung cancer: Analysis of intrathoracic organ motion during extreme phases of breathing. Int J Radiat Oncol Biol Phys 51:1081–1092 van Sornsen de Koste JR, Lagerwaard FJ, Nijssen-Visser MR et al, 2003, Tumor location cannot predict the mobility of lung tumors: A 3D analysis of data generated from multiple CT scans. Int J Radiat Oncol Biol Phys 56:348–354 Shinichiro M, Masahiro E, Shuhei K, Tomoyasu Y, Susumu K, Masayuki B, 2007, Four-dimensional measurement of lung tumor displacement using 256-multi-slice CT-scanner. Lung Cancer 56, 1):59–67 April Plathow C, Ley S, Fink C, Puderbach M, Hosch W, Schmahl A, Debus J, Kauczor H, 2004, Analysis of intratrochacic tumor mobility during whole breathing cycle by dynamic MRI. Int J Radiation Oncology Biol Phys 59(4):952–959 De NeveW, Derycke S, De GersemW, Vkaet L, DeWagner C, 1998, Portal imaging in conformal radiotherapy of lung cancer. In:Mornex F, Van Houtte P, eds, Treatment optimization for lung cancer from classical to innovative procedures. IASLC International Workshop. 24–27 June 1998; Annecy, France). Elsevier, Amsterdam, pp 109–113 Verhey LJ, 1995, Immobilizing and positioning patients for radiotherapy. Semin Radiat Oncol 5:100–114 Kubo HD, Len PM, Minohara S, Mostafavi H, 2000, Breathing synchronized radiotherapy program at the University of California Davis Center Cancer. Med Phys 27:346–353 Hanley J, Debois MM, Mah D et al, 1999, Deep inspiration breathhold technique for lung tumors: The potential value of target immobilization and reduced lung density in dose escalation. Int J Radiat Oncol Biol Phys 45:603–611 Rosenzweig K, Hanley J, Mah D et al, 2000, The deep inspiration breath-hold technique in the treatment of inoperable nonsmall-cell lung cancer. Int J Radiat Oncol Biol Phys 48:81–87 Wong JW, Sharpe MB, Jaffray DA et al, 1999, The use of active breathing control, ABC, to reduce margin for breathing motion. Int J Radiat Oncol Biol Phys 44(4):911–919 Mah D, Hanley J, Rosenzweig K, Yorke E, Braban L, Ling C, Leibel A, Mageras G, 2000, Technical aspects of the deep inspiration breath-hold technique in the treatment of thoracic cancer. Int J Radiat Oncol Biol Phys 48:1175–1185 Plathow C, Hof H, Kuhn S, Puderbach M, Ley S, Biederer J, Claussen CD, Huber PE, Schaefer J, Tuengerthal S, Kauczor HU, 2006, Therapy monitoring using dynamic MRI: analysis of lung motion and intrathoracic tumor mobility before and after radiotherapy. Eur Radiol 16(9):1942–1950 Sep, Epub 2006 Apr 21 Plathow C, Ley S, Fink C et al, 2004, Evaluation of chest motion and volumetry during the breathing cycle by dynamic MRI in healthy subjects: comparison with pulmonary function tests. Invest Radiol 39:202–209 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 269 EP 277 DI 10.1007/s12253-008-9101-5 PG 9 ER PT J AU Nagy, B Szendroi, A Romics, I AF Nagy, Balint Szendroi, Attila Romics, Imre TI Overexpression of CD24, c-myc and Phospholipase 2A in Prostate Cancer Tissue Samples Obtained by Needle Biopsy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Gene expression; CD24; c-myc; Phospholipase 2A ID Prostate cancer; Gene expression; CD24; c-myc; Phospholipase 2A AB Altered CD24, c-myc and phospholipase 2a expression was reported in different cancers. Our aim was to measure the expression of these genes in prostate cancer tissues, and compare it to non-cancerous samples. Prostate tissue samples were collected by needle biopsy from 20 prostate cancer (PCA) and 11 benign prostate hyperplasic (BPH) patients. RNA was isolated; cDNA synthetized, CD24, c-myc and phospholipase 2A (PL2A) expressions were determined by quantitative real-time PCR method. The expression of β-globin gene was measured for normalization of the gene expression results. Serum prostate specific antigen (PSA) levels were determined by microparticle enzyme immunoassay (MEIA) method. PSA levels were significantly different between the PCA and BPH groups, 252.37±308.33 ng/ml vs. 3.5±2.14 ng/ml (p=0.001), respectively. CD24 expression was 988.86±3041 ng/μl in prostate tumor and 4.00±4.25 ng/μl in the BPH group (p=0.035). The c-myc expression was 88.32±11.93 ng/μl in the prostate tumor and 17.08±21.75 ng/μl in the BPH group (p=0.02), and the PL2A 31.36±67.02 ng/μl was in PCA and 5.56±14.08 ng/μl in BPH (p=0.025). Gleason’s scores showed correlation with c-myc (p=0.019) and PSA (p=0.033) levels. Overexpression of PL2A, CD24 and c-myc was observed in prostate cancer samples using quantitative real-time PCR method. C1 [Nagy, Balint] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Baross 27, 1088 Budapest, Hungary. [Szendroi, Attila] Semmelweis University, Department of UrologyBudapest, Hungary. [Romics, Imre] Semmelweis University, Department of UrologyBudapest, Hungary. RP Nagy, B (reprint author), Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, 1088 Budapest, Hungary. EM nabal@noi1.sote.hu CR Muller H, Brenner H, 2006, Urine markers as possible tools for prostate cancer screening: review of performance characteristics and practicality. Clin Chem 52:562–573 Kopper L, Timar J, 2005, Genomics of prostate cancer: is there anything to “translate”. Pathol Oncol Res 11:197–203 Maitland NJ, Collins A, 2005, A tumour stem cell hypothesis for the origins of prostate cancer. BJU Int 96:1219–1223 Hughes C, Murphy A, Martin C et al, 2005, Molecular pathology of prostate cancer. J Clin Pathol 58:673–684 Quinn DI, Henshall SM, Sutherland LR, 2005, Molecular markers of prostate cancer outcome. Eur J Cancer 41:858–887 Glinsky VG, Glinskii BA, Stephenson JA et al, 2004, Gene expression profiling predicts clinical outcome of prostate cancer. J Clin Invest 113:913–923 Prowatke I, Devens F, Benner A et al, 2006, Expression analysis of imbalanced genes in prostate carcinoma using tissue microarrays. Br J Cancer 96:82–88 Nupponen NN, Kakkola L, Koivisto P et al, 1998, Genetic alterations in hormone refractory recurrent prostate carcinomas. Am J Pathol 153:141–148 Kristiansen G, Denkert C, Schluns K et al, 2002, CD24 is expressed in ovarian cancer and is a new independent prognostic marker of patients survival. Am J Pathol 161:1215–1221 Kristiansen G, Schluns K, Yongwei Y et al, 2003, CD24 is an independent prognostic marker of survival in non-small cell lung cancer patients. Br J Cancer 88:231–236 Kristiansen G, Winzer KJ, Mayordomo E et al, 2003, CD24 expression is a new prognostic marker in breast cancer. Clin Cancer Res 9:4906–4913 Kristiansen G, Pilarsky C, Pervan J et al, 2004, CD24 expression is a significant predictor of PSA relapse and poor prognosis in low grade or organ confined prostate cancer. Prostate 58:183–192 Ponzielli R, Katz S, Barsyte-Lovejoy D et al, 2005, Cancer therapeutics: targeting the dark side of Myc. Eur J Cancer 41:2485–2501 Devi RG, Oldenkamp RJ, London AC et al, 2002, Inhibition of human chorionic gonadotropin β-subunit modulates the mitogenic effect of c-myc in human prostate cancer cells. Prostate 53:200–210 Hirabayashi T, Shimuzu T, 2000, Localization and regulation of cytosolic phospholipase A2. Biochim Biophys Acta 1488:124–138 Dong Q, Patel M, Scott KF et al, 2006, Oncogenic action of phospholipase A2 in prostate cancer. Cancer Letters 240:9–16 Wikmam H, Seppanen JK, Sarhadi VK et al, 2004, Caveolins as tumor markers in lung cancer detected by combined use of cDNA and tissue microarrays. J Pathol 203:584–593 Kettunen E, Nicholson AG, Nagy B et al, 2005, L1CAM, INP10, P-cadherin, tPA and ITGB4 overexpression in malignant pleural mesothelioma primary tumors revealed by combined use of cDNA and tissue microarray. Carcinogenesis 26:17–25 Schostak M, Krause H, Miller K et al, 2006, Quantitative realtime RT-PCR of CD24 mRNA in the detection of prostate cancer. BMC Urol 119:1396–1402 Nagy B, Lundan T, Larramendy ML et al, 2003, Abnormal expression of apoptosis-related genes in haematological malignancies: overexpression of MYC is poor prognostic sign in mantle cell lymphoma. Br J Haematol 120:434–441 Ohl F, Jung M, Xu C et al, 2005, Gene expression studies in prostate cancer tissue: with reference gene should be selected for normalization? J Mol Med 83:1014–1024 Calvo A, Gonzalez-Moreno O et al, 2005, Prostate cancer and the genomic revolution: advances using microarray analyses. Mut Res 576:66–79 Raife TJ, Lager DJ, Kemp JD et al, 1994, Expression of CD24, BA-1, predicts monocytic lineage in acute myeloid leukemia. Am J Clin Pathol 101:296–299 Jackson D, Waibel R, Weber E et al, 1992, CD24, a signaltransducing molecule expressed on human B cells, is a major surface antigen on small cell lung carcinomas. Cancer Res 52:5264–5270 Huang LR, Hsu HC, 1995, Cloning and expression of CD24 gene in human hepatocellular carcinoma: a potential early tumor marker gene correlates with p53 mutation and tumor differentiation. Cancer Res 55:4717–4721 Pirrucello SJ, Lang MS, 1990, Differential expression of CD24- related epitopes in mycosis fungiodes/Sezary syndrome: a potential marker for circulating Sezary cells. Blood 76:895–905 Jenkins RB, Qian J, Lieber MM et al, 1997, Detection of c-myc oncogene amplification and chromosomal anomalies in metastatic prostatic carcinoma by fluorescence in situ hybridization. Cancer Res 57:524–531 Bernard D, Pourtuer-Manzanedo A, Gil J et al, 2003, Myc confers androgen-independent prostate cancer cell growth. J Clin Invest 112:1724–1731 Shachaf CM, Kopelman AM, Arvanitis C et al, 2004, MYC inactivation uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer. Nature 431:1112–1117 Arnold I, Watt FM, 2001, c-Myc activation in transgenic mouse epidermis results in mobilization of stem cells and differentiation of their progeny. Curr Biol 11:558–568 Liu YA, Roudier PM, True DL, 2004, Heterogeneity in primary and metastatic prostate cancer as defined by cell surface CD profile. Am J Pathol 165:1543–1556 Hughes-Fulford M, Tjandrawinata RR, Li C-F et al, 2005, Arachidonic acid, an omega-6 fatty acid, induces cytoplasmic phospholipase A2 in prostate carcinoma cells. Carcinogenesis 26:1520–1526 Kallajoki M, Alanen KA, Nevalainen M et al, 1998, Group II phospholipase A2 in human male reproductive organs and genital tumors. Prostate 35:263–272 Shimuzu M, Matsumoto Y, Kurosawa T et al, 2008, Release of arachidonic acid induced by tumor necrosis factor-α in the presence of caspase inhibition: evidence for cytosolic phospholipase A2α- independent pathway. Biochem Pharmacol, DOI 10.1016/j. bcp.2007.11.020 Cummings BS, 2007, Phospholipase A2 as targets for anticancer drugs. Biochem Pharmacol 74:949–959 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 279 EP 283 DI 10.1007/s12253-008-9077-1 PG 5 ER PT J AU Kojima, M Nakamura, N Shimizu, K Segawa, A Kaba, S Masawa, N AF Kojima, Masaru Nakamura, Naoya Shimizu, Kazuhiko Segawa, Atsuki Kaba, Sadayuki Masawa, Nobuhide TI MALT Type Lymphoma Demonstrating Prominent Plasma Cell Differentiation Resembling Fibrous Variant of Hashimoto’s Thyroiditis: a Three Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MALT type lymphoma; Thyroid gland; Fibrous variant; Hashimoto’s thyroiditis; Immunohistochemistry ID MALT type lymphoma; Thyroid gland; Fibrous variant; Hashimoto’s thyroiditis; Immunohistochemistry AB Fibrous variant of Hashimoto’s thyroiditis (HT) is characterized by marked fibrous replacement of one third or more of the thyroid parenchyma. We present here three cases of mucosa associated lymphoid tissue (MALT) type lymphoma demonstrating prominent plasma cell differentiation resembling fibrous variant of HT. Histologically, thyroid structures were disturbed by a diffuse and focally nodular infiltration by mature plasma cells and cells with plasma cell differentiation against a background of prominent hyalinosis. In addition, scattered centrocyte-like (CCL) cells and lymphoepithelial lesions were observed in all three lesions. A portion of the resected specimens in all three cases exhibited HT. However, immunohistochemical study demonstrated that the plasma cells and CCL-cells of these three lesions had monotypic intracytoplasmic kappa light chain. Moreover, these three lesions demonstrated a clonal band on polymerase chain reaction assay for the immunoglobulin heavy chain gene. To avoid underdiagnosis, we emphasize that careful attention should be paid to these immunological features as well as to morphological findings. C1 [Kojima, Masaru] Gunma Cancer Center Hospital, Department of Pathology and Clinical Laboratories, 617–1, Takabayashinishi-cho, 373-8550 Ohta, Japan. [Nakamura, Naoya] Tokai University School of Medicine, Department of PathologyIsehara, Japan. [Shimizu, Kazuhiko] Ashikaga Red Cross Hospital, Department of Pathology and Clinical LaboratoriesAshikaga, Japan. [Segawa, Atsuki] Dokkyo Medical University School of Medicine, Department of Diagnostic and Anatomic PathologyMibu, Japan. [Kaba, Sadayuki] Gunma University, Faculty of Medicine, Department of Laboratory ScienceMaebashi, Japan. [Masawa, Nobuhide] Dokkyo Medical University School of Medicine, Department of Diagnostic and Anatomic PathologyMibu, Japan. RP Kojima, M (reprint author), Gunma Cancer Center Hospital, Department of Pathology and Clinical Laboratories, 373-8550 Ohta, Japan. EM mkojima@gunma-cc.jp CR Hyjek E, Isaacson PG, 1988, Primary B cell lymphoma of the thyroid and its relationship to Hashimoto’s thyroiditis. Hum Pathol 19:1315–1326 Isaacson PG, Norton AJ, 1994, Extranodal lymphomas. Churchill Livingstone, Edinburgh Katz SM, Vickery AL Jr, 1974, The fibrous variant of Hashimoto’s thyroiditis. Hum Pathol 2:161–170 Harach HR, Williams ED, 1983, Fibrous thyroiditis. An immunopathological study. Histopathology 7:739–751 Derringer GA, Thompson LDR, Frommelt RA et al, 2000, Malignant lymphoma of the thyroid gland. A clinicopathologic study of 108 cases. Am J Surg Pathol 24:623–639 Wan JH, Trainor KJ, Brisco MJ et al, 1990, Monoclonality in B cell lymphoma detected in paraffin wax embedded sections using the polymerase chain reaction. J Clin Pathol 43:888–890 Lennert K, Feller AC, 1992, Histopathology of non-Hodgkin’s lymphomas, Based on the updated Kiel classification). Springer, Berlin Hussong JW, Perkins SL, Schnitzer B et al, 1999, Extramedullary plasmacytoma. A form of marginal zone cell lymphoma. Am J Clin Pathol 111:111–115 Kovacs CS, Mant MJ, Nguyen GK et al, 1994, Plasma cell lesions of the thyroid: report of a case of solitary plasmacytoma and a review of the literature. Thyroid 4:65–71 Coffin CM, Watterson J, Priest JC et al, 1995, Extrapulmonary inflammatory myofibroblastic tumor, inflammatory pseudotumor). A clinicopathologic and immunohistochemical study of 84 cases. Am J Surg Pathol 19:859–872 Coffin CM, Hornick JL, Fletcher CDM, 2007, Inflammatory myofibroblastic tumor. Comparison of clinicopathologic, histologic, immunohistochemical features and ALK expression in atypical and aggressive cases. Am J Surg Pathol 31:509–520 Kriegel L, Guetgemann I, Zhou H, 2007, Plasma cell granuloma of the thyroid gland mimicking carcinoma: A case report and review of the literature. Pathol Res Pract 203:813–817 Deniz K, Patrioglu, Oketen T, 2008, Plasma cell granuloma of the thyroid. Case report. APMIS 117:167–172 Kremer M, Ott G, Nathrath M et al, 2005, Primary extramedullary plasmacytoma and multiple myeloma: Phenotypic differences revealed by immunohistochemical analysis. J Pathol 205: 92–101 Feller AC, Diebold J, 2003, Histopathology of nodal and extranodal non-Hodgkin's lymphomas. Springer, Berlin Yatabe Y, Suzuki R, Matsuno Y et al, 2001, Morphological spectrum of cyclin D-1 positive mantle cell lymphoma: study of 168 cases. Pathology International 51:747–761 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 285 EP 289 DI 10.1007/s12253-008-9108-y PG 5 ER PT J AU Saw, S Thomas, N Gleeson, JM Bodi, I Connor, S Hortobagyi, T AF Saw, Sonia Thomas, Nick Gleeson, J Michael Bodi, Istvan Connor, Steve Hortobagyi, Tibor TI Giant Cell Tumour and Central Giant Cell Reparative Granuloma of the Skull: do These Represent Ends of a Spectrum? A Case Report and Literature Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Giant cell tumour; Giant cell reparative granuloma; Histology; Skull bone ID Giant cell tumour; Giant cell reparative granuloma; Histology; Skull bone AB Giant cell tumour (GCT) of bone is an uncommon primary bone neoplasm typically occurring at the epiphyses of long bones in young adults. They are osteolytic neoplasms with approximate local recurrence rates of 25%, and 2% of patients develop pulmonary metastases. These tumours appear very rarely in the skull, with those few reported cases arising predominantly in the sphenoid and occasionally the temporal bones. They demonstrate benign histological features, but are locally aggressive and surgical excision is the treatment of choice. It is widely believed that giant cell tumours should be distinguished from other giant cell lesions, importantly central giant cell reparative granulomata (CGCG) which are thought to have a lower recurrence rate and for which no cases of malignant transformation or metastases have been reported. Investigators have noted that giant cell lesions in the skull bones may be unique and that GCT and CGCG may be part of a spectrum of a single disease process. We present a case of a giant cell tumour of the temporal bone which illustrates and re-emphasises this concept and review the literature on these lesions. C1 [Saw, Sonia] King’s College Hospital, Clinical Neuropathology, De Crespigny Park, SE5 8AF London, UK. [Thomas, Nick] King’s College Hospital, Department of NeurosurgeryLondon, UK. [Gleeson, J Michael] Guy’s Hospital, Department of Otolaryngology and Skull Base SurgeryLondon, UK. [Bodi, Istvan] King’s College Hospital, Clinical Neuropathology, De Crespigny Park, SE5 8AF London, UK. [Connor, Steve] King’s College Hospital, Department of NeuroradiologyLondon, UK. [Hortobagyi, Tibor] King’s College Hospital, Clinical Neuropathology, De Crespigny Park, SE5 8AF London, UK. RP Hortobagyi, T (reprint author), King’s College Hospital, Clinical Neuropathology, SE5 8AF London, UK. EM tibor.hortobagyi@iop.kcl.ac.uk CR Reid R, Banerjee S, Sciot R, 2002, Giant cell tumour. In: Fletcher C, Unni K, Mertens F, eds, Pathology and genetics of tumours of soft tissue and bone, WHO classification of tumours. IARC, Lyon, pp 310–312 Bertoni F, Unni K, Beabout J et al, 1992, Giant cell tumour of the skull. Cancer 70(5):1124–1132 Hirschl S, Katz A, 1974, Giant cell reparative granuloma outside the jaw bone. Hum Pathol 5(2):171–181 Williams J, Thorell W, Treves J et al, 2000, Giant cell reparative granuloma of the petrous temporal bone: a case report and literature review. Skull Base Surg 10(2):89–93 Ung F, Li K, Keith D et al, 1998, Giant cell reparative granuloma of the temporal bone: Case report and review of the literature. Otolaryngol Head Neck Surg 118:525–529 Auclair P, Cuenin P, Kratochvil F et al, 1988, A clinical and histomorphologic comparison of the central giant cell granuloma and the giant cell tumour. Oral Surg 66:197–208 Whitaker S, Waldron C, 1993, Central giant cell lesions of the jaws. a clinical, radiologic and histopathologic study. Oral Surg Oral Med Oral Pathol 75(2):199–208 Jaffe H, 1953, Giant-cell reparative granuloma, traumatic bone cyst, and fibrous, fibro-osseous, dysplasia of the jawbones. Oral Surg 6:159–175 Kashiwagi N, Hirabuki N, Andou K et al, 2006, MRI and CT findings of the giant cell tumours of the skull; five cases and a review of the literature. Eur J Radiol 58:435–443 Elder J, Berry C, Gonzalez-Gomez I et al, 2007, Giant cell tumour of the skull in paediatric patients. J Neurosurg 107(Supp 1 Paed):69–74 Lee M-Y, Lee E-J, 2006, Giant cell tumour of the petrous temporal bone with direct invasion into the middle ear. J Craniofac Surg 17:797–800 Wulling M, Engels C, Jesse N et al, 2001, The nature of giant cell tumour of bone. J Cancer Res Clin Onc 127(8):467–474 Nemoto Y, Inoue Y, Tashiro T et al, 1995, Central giant cell granuloma of the temporal bone. Am J Neuroradiol 16:982– 985 Dickson B, Li S-Q, Wunder J et al, 2008, Giant cell tumour of bone express p63. Mod Pathol 21:369–375 Leonard J, Gokden M, Kyriakos M et al, 2001, Malignant giantcell tumour of the parietal bone: case report and review of the literature. Neurosurgery 48(2):424–429 Schwartz H, Eskew J, Butler M, 2002, Clonality studies in giant cell tumour of bone. J Orthop Res 20(2):387–390 Antal I, Sapi Z, Szendroi M, 1999, The prognostic significance of DNA cytophotometry and proliferation index, Ki-67, in giant cell tumours of bone. Int Orthop 23(6):315–319 Antal I, Sapi Z, Szendroi M, 2000, Malignant transformation of giant cell tumour in the distal radius. Value of the DNA cytophotometry Orthopade 29(7):677–683. German Rousseau M, Handra-Luca A, Lazennec J et al, 2004, Metachronous multicentric giant-cell tumour of the bone in the lower limb. case report and Ki-67 immunohistochemistry study. Virchows Arch 445(1):79–82 Fukunaga M, Nikaido T, Shimoda T et al, 1992, A flow cytometric analysis of giant cell tumours of bone including two cases with malignant transformation. Cancer 70(7):1886– 1894 Sara A, Ayala A, el-Naggar A et al, 1990, Giant cell tumour of bone. a clinicopathologic and DNA flow cytometric analysis. Cancer 66(10):2186–2190 Sulh MA, Greco MA, Jiang T et al, 1996, Proliferation index and vascular density of giant cell tumours of bone: are they prognostic markers? Cancer 77(10):2044–2051 Dahlin D, Unni K, 1986, Bone Tumours—General aspects and data on 8542 cases, 4th edn. Thomas, Springfield Bertoni F, Bacchini P, Staals E, 2003, Malignancy in giant cell tumour of bone. Cancer 97(10):2520–2529 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 291 EP 295 DI 10.1007/s12253-008-9124-y PG 5 ER PT J AU Koseoglu, DR Ozkan, N Filiz, ON Kayaoglu, HA Aydin, M Culha, NE Ersoy, FO AF Koseoglu, Dogan Resit Ozkan, Namik Filiz, Onuk Nurper Kayaoglu, Huseyin Ayhan Aydin, Mehtap Culha, N Emre Ersoy, F Omer TI Intranodal Palisaded Myofibroblastoma; a Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Myofibroblastoma; Intranodal palisaded myofibroblastoma; Amianthoid fiber ID Myofibroblastoma; Intranodal palisaded myofibroblastoma; Amianthoid fiber AB Intranodal palisaded myofibroblastoma (IPM) also called as intranodal hemorrhagic spindle cell tumor with amianthoid fibers is a distinctive and rare mesenchymal neoplasm of lymph nodes. This entity generally misdiagnosed as intranodal Kaposi’s sarcoma or schwannoma in past. In contrast to Kaposi’s sarcoma, it behaves in a benign fashion and does not need any further therapy except total surgical resection of the mass. This neoplasm has a great predilection for the inguinal region. The lesion presents typically as a unilateral, painless, solitary mass. To our knowledge, approximately 53 cases of IPM have been reported in the English-language literature. We present a 43-year-old-male patient with IPM and discuss histological, immunohistochemical features and pathogenesis of this rare benign neoplasm. C1 [Koseoglu, Dogan Resit] Gaziosmanpasa University, School of Medicine, Department of Pathology, 60030 Tokat, Turkey. [Ozkan, Namik] Gaziosmanpasa University, School of Medicine, Department of General SurgeryTokat, Turkey. [Filiz, Onuk Nurper] Gaziosmanpasa University, School of Medicine, Department of Pathology, 60030 Tokat, Turkey. [Kayaoglu, Huseyin Ayhan] Gaziosmanpasa University, School of Medicine, Department of General SurgeryTokat, Turkey. [Aydin, Mehtap] Ankara Ataturk Training and Research Hospital, 1st Pathology ClinicAnkara, Turkey. [Culha, N Emre] State Hospital of Sincan, Laboratory of PathologyAnkara, Turkey. [Ersoy, F Omer] Gaziosmanpasa University, School of Medicine, Department of General SurgeryTokat, Turkey. RP Koseoglu, DR (reprint author), Gaziosmanpasa University, School of Medicine, Department of Pathology, 60030 Tokat, Turkey. EM residdogan@hotmail.com CR Weiss SW, Gnepp DR, Bratthauer GL, 1989, Palisaded myofibribroblastoma. A benign mesenchymal tumor of lymph node. Am J Surg Pathol 13:341–346 Suster S, Rosai J, 1989, Intranodal hemorrhagic spindle-cell tumor with “amianthoid” fibers. Report of six cases of a distinctive mesenchymal neoplasm neoplasm of the inguinal region that simulates Kaposi’s sarcoma. Am J Surg Pathol 13:347–357 Lee JY-Y, Abell E, Shevichik GJ, 1989, Solitary spindle cell tumor with myoid differentiation of the lymph node. Arch Pathol Lab Med 113:547–550 Alguacil-Garcia A, 1992, Intranodal myofibroblastoma in a submandibular lymph node. Am J Clin Pathol 97:69–72 Fletcher CDM, Stirling RW, 1990, Intranodal myofibroblastoma presenting in the submandibular region: evidence of a broader clinical and histologic spectrum. Histopathology 16:287–294 Michal M, Chlumska A, Povysilova V, 1992, Intranodal “amianthoid” myofibroblastoma: report of six cases:immunohistochemical and electron microscopical study. Pathol Res Pract 188:199–204 Creager AJ, Garwacki CP, 1999, Recurrent intranodal palisaded myofibroblastoma with metaplastic bone formation. Arch Pathol Lab Med 123:433–436 Bigotti G, Coli A, Mottolese M, di Filippo F, 1991, Selective location of palisaded myofibroblastoma with amianthoid fibres. J Clin Pathol 44:761–764 Skalova A, Michal M, Chlumska A, LeivoI, 1992, Collagen composition and ultrastructure of the so-called amianthoid fibres in palisaded myofibroblastoma. Ultrastructural and immunohistochemical study. J Pathol 167:335–340 Kleist B, Poetsch M, Schmoll J, 2003, Intranodal palisaded myofibroblastoma with overexpression of cyclin D1. Arch Pathol Lab Med 127:1040–1043 Lioe TF, Allen DC, Bell JC, 1994, A case of multicentric intranodal palisaded myofibroblastoma. Histopathology 24:173–175 Barbareschi M, Mariscotti C, Ferrero S, Pignatiello U, 1990, Intranodal haemorrhagic spindle cell tumor: a benign Kaposi like nodal tumor. Histopathology 17:93–96 Tanda F, Massarelli G, Cossu A, Bosincu L, Cossu S, Ibba M, 1993, Primary spindle cell tumor of lymph node with “amianthoid” fibers: a histological, immunohistochemical and ultrastructural study. Ultrastruct Pathol 17:195–205 Eyden BP, Harris M, Greywoode GIN, Christensen L, Banerjee SS, 1996, Intranodal myofibroblastoma: report of a case. Ultrastruct Pathol 20:79–88 Rossi A, Bulgarini A, Rondanelli E, Incensanti R, 1995, Intranodal palisaded myofibroblastoma: report of three new cases. Tumori 81:464–468 Burns MK, Headington JT, Rasmussen JE, 1991, Palisaded myofibroblastoma simulating chronic primary lymphadenopathic Kaposi’s sarcoma. J Am Acad Dermatol 25:566–568 Onsurbe PM, Heffernan JAJ, Hidalgo GG, 2002, Fine needle aspiration cytology of intranodal myofibroblastoma: a case report. Acta Cytol 46:1143–1147 Ciralik H, Ezberci F, Bulbuloglu E, Aydin A, 2005, Intranodal palisaded myofibroblastoma and differential diagnosis: a case report. Chin Med J 118(20):1758–1760 Perez-Mies B, Campos AI, 2008, Intranodal palisaded myofibroblastoma. Arch Pathol Lab Med 132(8):1224–1225 Cobanoglu U, Siviloglu C, Ersoz S, Ozoran Y, 2006, Intranodal palisaded myofibroblastoma with metaplastic bone formation. Saudi Med J 27(8):1249–1250 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 297 EP 300 DI 10.1007/s12253-008-9122-0 PG 4 ER PT J AU Yano, H Ohe, N Shinoda, J Yoshimura, Shi Iwama, T AF Yano, Hirohito Ohe, Naoyuki Shinoda, Jun Yoshimura, Shin-ichi Iwama, Toru TI Immunohistochemical Study Concerning the Origin of Neurocytoma—A Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Extraventricle; Musashi 1; Neural stem cell; Neurocytoma; Origin; Subependymal ID Extraventricle; Musashi 1; Neural stem cell; Neurocytoma; Origin; Subependymal AB A 26-year-old woman presented with rapid tumor growth in her left frontal lobe during 9 years of observation. Operative findings revealed a massive tumor connected to gelatinous, transparent membranous tissue (MT), which extended from the paraventricular zone and continued into the lateral ventricle. Histological diagnosis was atypical neurocytoma. Immunohistochemical analyses revealed that the tumor was strongly positive for not only neural markers but also a glial marker, while the MT was positive for a neural marker. The Ki-67/MIB-1 labeling index was 9.1% in the tumor body and 0% in the MT. Musashi 1, a marker of neural stem cells, was strongly positive in both the tumor body and the MT. We speculate that the tumor growth was due to a rapid decline of the Musashi 1-positive cells to glial differentiation. These cells may be candidates for the origin of the tumor. C1 [Yano, Hirohito] Gifu University Graduate School of Medicine, Department of Neurosurgery, 1-1 Yanagido, 501-1194 Gifu, Japan. [Ohe, Naoyuki] Gifu University Graduate School of Medicine, Department of Neurosurgery, 1-1 Yanagido, 501-1194 Gifu, Japan. [Shinoda, Jun] Kizawa Memorial Hospital, Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain DysfunctionMinokamo, Japan. [Yoshimura, Shin-ichi] Gifu University Graduate School of Medicine, Department of Neurosurgery, 1-1 Yanagido, 501-1194 Gifu, Japan. [Iwama, Toru] Gifu University Graduate School of Medicine, Department of Neurosurgery, 1-1 Yanagido, 501-1194 Gifu, Japan. RP Yano, H (reprint author), Gifu University Graduate School of Medicine, Department of Neurosurgery, 501-1194 Gifu, Japan. EM hirohito@gifu-u.ac.jp CR Elek G, Slowik F, Eross L, Toth S et al, 1999, Central neurocytoma with malignant course. Neuronal and glial differentiation and craniospinal dissemination. Pathol Oncol Res 5:155– 159 Kim BJ, Kim SS, Kim YI et al, 2004, Forskolin promotes astroglial differentiation of human central neurocytoma cells. Exp Mol Med 29:52–56 Kubota T, Hayashi M, Kawano H et al, 1991, Central neurocytoma: immunohistochemical and ultrastructural study. Acta Neuropathol, Berl, 81:418–427 Valduesza JM, Westphal M, Vortmeyer A et al, 1996, Central neurocytoma: clinical, immunohistologic, and biologic findings of a human neuroglial progenitor tumor. Surg Neurol 45:49–56 von Deimling A, Janzer R, Kleihues P et al, 1990, Patterns of differentiation in central neurocytoma. An immunohistochemical study of eleven biopsies. Acta Neuropathol, Berl, 79:473–479 von Deimling A, Kleihues P, Saremaslani P et al, 1991, Histogenesis and differentiation potential of central neurocytomas. Lab Invest 64:585–591 Westphal M, Stavrou D, Nausch H et al, 1994, Human neurocytoma cells in culture show characteristics of astroglial differentiation. J Neurosci Res 38:698–704 You H, Kim YI, Im SY et al, 2005, Immunohistochemical study of central neurocytoma, subependymoma, and subependymal giant cell astrocytoma. J Neurooncol 74:1–8 Kanemura Y, Mori K, Sakakibara S et al, 2001, Musashi 1, an evolutionarily conserved neural RNA-binding protein, is a versatile marker of human glioma cells in determining their cellular origin, malignancy, and proliferative activity. Differentiation 68:141–152 Toda M, Iizuka Y, Yu W et al, 2001, Expression of the neural RNA-binding protein Musashi1 in human gliomas. Glia 34:1–7 Uchida K, Mukai M, Okano H et al, 2004, Possible oncogenicity of subventricular zone neural stem cells: case report. Neurosurgery 55:977–978 Lee MK, Rebhun LI, Frankfurter A, 1990, Posttranslational modification of class III β-tubulin. Proc Natl Acad Sci USA 87:7195–7199 Menezes JR, Luskin MB, 1994, Expression of neuron-specific tubulin defines a novel population in the proliferative layers of the developing telencephalon. J Neurosci 14:5399–5416 Mullen RJ, Buck CR, Smith AM, 1992, NeuN, a neuronal specific nuclear protein in vertebrates. Development 116:201–211 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2009 VL 15 IS 2 BP 301 EP 305 DI 10.1007/s12253-008-9106-0 PG 5 ER PT J AU Keszthelyi, A Majoros, A Nyirady, P Mayer, P Bach, D Romics, I AF Keszthelyi, Attila Majoros, Attila Nyirady, Peter Mayer, Peter Bach, Dietmar Romics, Imre TI Voiding Symptoms and Urodynamic Findings in Patients with Modified Ileal Neobladde SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; Radical cystectomy; Reddy neobladder; Urinary incontinence; Urodynamics ID Bladder cancer; Radical cystectomy; Reddy neobladder; Urinary incontinence; Urodynamics AB The aim of our study was to find the cause of urinary incontinence and voiding dysfunction in patients undergoing radical cystectomy and orthotopic bladder replacement with modified ileal neobladder (Reddy). Twenty-eight incontinent patients (operated on between 1988 and 2004) were involved in our examination. Based on the complaints of the patients, continence status was evaluated and divided into two groups: group I: partially incontinent (only night-time incontinence) n=11 (39.3%) and group II: totally incontinent (night-time and daytime incontinence) n=17 (60.7%). Detailed urodynamic examination (enterocystometry and urethral pressure profile) in addition to involuntary neobladder contractions and capacity detection were carried out on all patients. Furthermore resting pressure and maximal voluntary contraction ability of the sphincter were determined and statistically analyzed in both groups. Significant difference was noticed in resting pressure and maximal voluntary contraction ability of the sphincter among the partially incontinent and totally incontinent patients. Frequency, intensity and duration of involuntary neobladder contractions also showed significant differences between the two groups. Incontinence of neobladder depends not only on the destruction of resting and contraction capability of the urethral sphincter, but also on the presence or absence of involuntary contractions in the wall of the neobladder and decreased capacity of the neobladder. C1 [Keszthelyi, Attila] Semmelweis UniversityBudapest, Hungary. [Majoros, Attila] Semmelweis UniversityBudapest, Hungary. [Nyirady, Peter] Semmelweis UniversityBudapest, Hungary. [Mayer, Peter] Semmelweis UniversityBudapest, Hungary. [Bach, Dietmar] Semmelweis UniversityBudapest, Hungary. [Romics, Imre] Semmelweis UniversityBudapest, Hungary. RP Keszthelyi, A (reprint author), Semmelweis University, Budapest, Hungary. EM attilakeszthelyi@hotmail.com CR Couvelaire R, 1952, Le reservoir ileal de substitution apres la cystectomia total chez l’homme. J d’Urol 57:408–411 Lilien OM, Camey M, 1984, 25-year experience with replacement of the human bladder, Camey procedure). J Urol 132: 886–889 Hautmann RE, Egghart G, Frohnenberg D, Miller K, 1988, The ileal neobladder. J Urol 136:27–31 Reddy PK, Lange PH, Fraley EE, 1991, Total bladder replacement using detubularised sigmoid colon: technique and results. J Urol 145:51–55 Studer UE, Ackermann D, Casanova GA, Zingg EJ, 1989, Three years experience with ileal low pressure bladder substitute. BJ Urol 63:43–46 Reddy PK, 1987, Non stomal continent reservoir: use of detubularized ileal segment for bladder replacement. World J Urol 5:190–193 Arai Y, Taki Y, Kawase N, Terachi T, Kakehi Y, Okada T, 1999, Orthotopic ileal neobladder in male patients: functional outcames of 66 cases. Int J Urol 6:388–392 Hobisch A, Kadir T, Kinzl J, 2001, Life after cystectomy and orthotopic neobladder versus ileal conduit urinary diversion. Seminars Urol Oncol 19:18–23 Kulkarni JN, Pramesh CS, Rathi S, Pantvaidya GH, 2002, Longterm results of orthotopic neobladder reconstruction after radicals cystectomy. BJ Urol 91:485–488 Skolarikos A, Deliveliotis C, Alargof E, Ferakis N, Protogerou V, Dimopoulos C, 2004, Modified ileal neobladder for continent urinary diversion: functional results after 9 years of experience. Urol 171:2298–2301 Laguna MP, Brenninkmeier M, Bellon JA, Marrero R, Wijkstra H, Rosette J, Isorna S, 2005, Long term functional and urodynamic results of 50 patients receiving a modified sigmoid neobladder created with a short distal segment. J Urol 174:963–967 Yadav SS, Sadadukhi C, Sharma KK, Yadav RG, Mathur R, 2006, Sigmoid orthotopic neobladder after radical cystectomy for bladder tumour: an Indian experience. BJU Inter 99:403–406 Jensen JB, Lundbeck F, Jensen KME, 2006, Comlications and neobladder function of the Hautmann orthotopic ileal neobladder. BJU Inter 98:1289–1294 Giannantoni A, Mearini E, Di Stasi SM, Mearini L, Bini V, Pizzirusso G, 2004, Assesment of bladder and urethral spichteric function before and after radical retropubic prostatectomy. J Urol 171:1563–1566 Hammerer P, Huland H, 1997, Urodynamic evaluation of changes in urinary control after radical retropubic prostatectomy. J Urol 157:233–236 Majoros A, Bach D, Keszthelyi A, Hamvas A, Romics I, 2006, Urinary incontinence and voiding dysfunction after radical retropubic prostatectomy, prospective urodynamic study). Neuroul Urodyn 25:2–7 Gerharz E, Eingartner K, Dopatka T, Kohl U, Basler H, Riedmiller H, 1997, Quality of life after cystectomy and urinary diversion: result of a retrospective interdisciplinar study. J Urol 158:778–785 Mansson A, Caruso A, Capovilla E, 2000, Quality of life after radical cystectomy and orthotopic bladder substitution. A comparison between Italian and Swedish men. BJU Int 85:26–31 Protogerou V, 2004, Modified S-pouch neobladder vs ileal conduit and a matched control population: a quality of life survey. BJ Urol 94:350–354 Peremenis P, Burkhard F, Kessler T, Gramann T, Studer UE, 2004, Ileal orthotopic bladder substitute combined with an afferent tubular segment: long-term upper urinary tract changes and voiding pattern. Eur Urol 46:604–609 Arata R, Saika T, Tsushima T, Abarzua F, Nasu Y, Kumon H, 2007, Orthotopic ileal neobladder versus sigmoid neobladder: a quality of life, QOL, survey. Acta Med Okayama 61:229–234 Gerharz EW, Mansson A, Hunt S, Skinner EC, Mansson W, 2005, Quality of life after cystectomy and urinary diversion: an evidence based analysis. J Urol 174:1729–1736 Kikuchi E, Horiguchi Y, Nakashima J, Ohigashi T, Oya M, Nakagawa K, Miyajima A, Murai M, 2006, Assessment of long – term quality of life using the FACT-BL questionnaire in patients with an ileal conduit, continent reservoir or orthotopic neobladder. Jpn J Clin Oncol 36:712–716 Saika T, Arata R, Tsushima T, Nasu Y, Suyama B, Takeda K, Ebara S, Manabe D, Kobayashi T, Tanimoto R, Kumon H, 2007, Health-related quality of life after radical cystectomy for bladder cancer in elderly patients with an ileal conduit, ureterocutaneostomy, or orthotopic urinary reservoir: a comperative questionnaire survey. Acta Med Okoyama 61:199–203 Schrier B, Laguna MP, van der Pal F, Isorna S, Witjes JA, 2005, Comparison of orthotopic sigmoid and ileal neobladders: continence and urodynamic parameters. Eur Urol 47:679–685 Sevin G, Soyupek S, Armagan A, Hoscan MB, Oksay T, 2004, Ileal orthotopic noebladder, modified Hautmann, via a shorter detubularized ileal segment: experience and results. BJU International 94:355–359 El Bahnasawy MS, Osman Y, Gomha MA, Shaaban AA, Ashamallah A, Ghoneim MA, 2000, Nocturnal enuresis in men with an orthotopic ileal reservoir: urodynamic evaluation. J Urol 164:10–13 Burckhard FC, Kessler TM, Springer J, Studer UE, 2006, Early and late urodynamic assessment of ileal orthotopic bladder substitutes combined with an afferent tubular segment. J Urol 175:2155–2161 El-Bahnasawy MS, Gomba MA, Shaaban AA, 2006, Urethral pressure profile following orthotopic neobladder: differences between nerve sparing and standard radical cystectomy techniques. J Urol 175:1759–1763 Thuroff JW, Mattiasson A, Andersen JT, Hedlund H, Hinman F, Hohenfellner M, Mansson W, Mundy AR, Rowland RG, Steven K, 1996, The standardization of terminology and assessment of functional characteristics of intestinal urinary reservoirs. BJ Urol 78:516–523 Steers WD, 20000, Voiding dysfunction in the orthotopic neobladder. World J Urol 18:330–337 Jakobsen H, Steven K, Stingsby B, Klarskov P, Hald T, 1987, Pathogenesis of noctural urinary incontinence after ileocaecal bladder replacement: continuous measurement of urethral closing pressure during sleep. BJ Urol 59:148–152 Porru D, Usai E, 1994, Orthotopic ileal bladder substitute after radical cystectomy: urodynamic features. Neurourol Urodyn 13:255–260 Tanago EA, 1978, The anatomy and physiology of micturition. Clin Obset Gynecol 5:3–6 Skinner DG, 1987, Editoral comment. J Urol 138:1154 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 307 EP 313 DI 10.1007/s12253-008-9099-8 PG 7 ER PT J AU Lakosi, F Antal, G Vandulek, Cs Kovacs, Garamvolgyi, R Petnehazy, O Bajzik, G Hadjiev, J Repa, I Bogner, P AF Lakosi, Ferenc Antal, Gergely Vandulek, Csaba Kovacs, Arpad Garamvolgyi, Rita Petnehazy, Ors Bajzik, Gabor Hadjiev, Janaki Repa, Imre Bogner, Peter TI Technical Feasibility of Transperineal MR-Guided Prostate Interventions in a Low-Field Open MRI Unit: Canine Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Interventional; Brachytherapy; MR-guidance ID Prostate cancer; Interventional; Brachytherapy; MR-guidance AB Magnetic resonance imaging (MRI) provides superior visualization of the prostate, its substructure, surrounding tissues, and, most important, focal lesions or cancer. The purpose of our canine study was to demonstrate the feasibility of a low-field (0.35 T) transperineal system that enables precise MR image guidance of prostate interventions. The canines were placed in the right lateral decubitus position. Template reconstruction, trajectory planning, contouring were based on T2-weighted FSE images. For image guidance and target confirmation, fast gradient spoiled-echo (FSPGR) sequence was used. MR compatible coaxial needles were manually inserted through the perineum to the base of the prostate. After satisfactory position was confirmed, brachytherapy catheters were placed through the coaxial needles. The mean deviation of the needle displacements was 2.9 mm with a median value of 2.7 mm. 97% of the errors were less than 4.0 mm. The needle placement accuracy was modelled by the Rayleigh distribution with a sigma value of 2.3 mm. Visual confirmation of needle placements was demonstrated on pathology tissue slices. The time needed for each step was: anaesthesia - 15 min, setup and positioning - 15 min, initial imaging - 15 min, template registration, projection - 15 min, contouring, trajectory planning, insertion of 12 needles - 60 min Based on our canine experiences our method seems to be a promising approach for performing feasible, accurate, reliable and high-quality prostate MR guidance within a reasonable time span. C1 [Lakosi, Ferenc] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, H-7400 Kaposvar, Hungary. [Antal, Gergely] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, H-7400 Kaposvar, Hungary. [Vandulek, Csaba] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, H-7400 Kaposvar, Hungary. [Kovacs, Arpad] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, H-7400 Kaposvar, Hungary. [Garamvolgyi, Rita] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, H-7400 Kaposvar, Hungary. [Petnehazy, Ors] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, H-7400 Kaposvar, Hungary. [Bajzik, Gabor] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, H-7400 Kaposvar, Hungary. [Hadjiev, Janaki] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, H-7400 Kaposvar, Hungary. [Repa, Imre] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, H-7400 Kaposvar, Hungary. [Bogner, Peter] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, H-7400 Kaposvar, Hungary. RP Bogner, P (reprint author), Kaposi Mor Teaching Hospital, H-7400 Kaposvar, Hungary. EM bogner.peter@sic.hu CR Otto S, Kasler M, 2005, A hazai es nemzetkozi daganatos halalozasi es megbetegedesi mutatok alakulasa. Magyar Onkologia 49:99–107 Jereczek-Fossa BA, Orecchia R, 2007, Evidence-based radiation oncology: definitive, adjuvant and salvage radiotherapy for nonmetastatic prostate cancer. Radiother Oncol 84:197–215 Morton G, 2005, The emerging role of high-dose-rate brachytherapy for prostate cancer. Clin Oncol 17:219–227 Presti JC Jr, 2000, Prostate cancer: assessment of risk using digital rectal examination, tumor grade, prostate-specific antigen, and systematic biopsy. Radiol Clin North Am 38:49–58 Futterer J, 2007, MR imaging in the local staging of prostate cancer. Eur J Radiol 63:328–334 Morgan VA, Kyriazi S, Ashley SE et al, 2007, Evaluation of the potential of diffusion-weighted imaging in prostate cancer detection. Acta Radiol 48:695–703 Alonzi R, Padhani AR, Allen C, 2007, Dynamic contrast enhanced MRI in prostate cancer. Eur J Radiol 63:335–350 Mueller-Lisse UG, Scherr MK, 2007, Proton MR spectroscopy of the prostate. Eur J Radiol 63:351–360 Pouliot J, Kim Y, Lessard E et al, 2004, Inverse planning for HDR prostate brachytherapy used to boost dominant intraprostatic lesions defined by magnetic resonance spectroscopy imaging. Int J Radiat Oncol Biol Phys 59:1196–1207 Van Lin EN, Futterer JJ, Heymink SW et al, 2006, IMRT boost dose planning on dominant intraprostatic lesions: gold markerbased three-dimensional fusion of CT with dynamic contrastenhanced and 1H-spectroscopic MRI. Int J Radiat Oncol Biol Phys 65:291–303 Beyersdorff D, Winkel A, Hamm B et al, 2005, MR imagingguided prostate biopsy with a closed MR unit at 1.5 T: initial results. Radiology 234:576–581 D’Amico AV, Cormack R, Tempany CM et al, 1998, Real-time magnetic resonance image-guided interstitial brachytherapy in the treatment of select patients with clinically localized prostate cancer. Int J Radiat Oncol Biol Phys 42:507–515 Fischer GS, Di Maio SP, Haker SJ et al, 2006, A system for MRguided prostate interventions. Proceedings of 1st IEEE/RASDEMBS International Conference on Biomedical Robotics and Biomechatronics February 2006, 68–73 Di Maio SP, Pieper S, Chinzei K, 2007, Robot-assisted needle placement in open MRI: system architecture, integration and validation. Comput Aided Surg 12:15–24 Fischer GS, DiMaio SP, Iordachita II et al, 2007, Robotic assistant for transperineal prostate interventions in 3 T closed MRI. Med Image Comput Comput Assist Interv Int Conf Med Image Comput Comput Assist Interv 10:425–433 Engelhard K, Hollenbach HP, Kiefer B et al, 2006, Prostate biopsy in the supine position in a standard 1.5-T scanner under real time MR-imaging control using a MR-compatible endorectal biopsy device. Eur Radiol 16:1237–1243 Menard C, Susil RC, Choyke P et al, 2004, MRI-guided HDR prostate brachytherapy in standard 1.5 T scanner. Int J Radiat Oncol Biol Phys 59:1414–1423 Susil RC, Camphausen K, Choyke P et al, 2004, System for prostate brachytherapy and biopsy in a standard 1.5 T MRI scanner. Magn Reson Med 52:683–687 Zangos S, Eichler K, Engelmann K et al, 2005, MR-guided transgluteal biopsies with an open low-field system in patients with clinically suspected prostate cancer: technique and preliminary results. Eur Radiol 15:174–182 Zangos S, Hercog C, Eichler K, 2007, MR-compatible assistance system for punction in a high-field system: device and feasibility of transgluteal biopsies of the prostate gland. Eur Radiol 17:1118– 1124 Potter R, Haie-Meder C, Van Limbergen E et al, 2005, Recommendations from gynaecological, GYN, GEC ESTRO working group, II): concepts and terms in 3D image-based treatment planning in cervix cancer brachytherapy—3D dose volume parameters and aspects of 3D image-based anatomy, radiation physics, radiobiology. Radiother Oncol 78:67–77 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 315 EP 322 DI 10.1007/s12253-008-9111-3 PG 8 ER PT J AU Cai, L Threadgill, DM Wang, Y Li, M AF Cai, Li Threadgill, D Michael Wang, Yalan Li, Ming TI Effect of Poly (ADP-ribose) Polymerase-1 Inhibition on the Proliferation of Murine Colon Carcinoma CT26 Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PARP-1; NF-κB; Proliferation; Colon carcinoma; In vitro ID PARP-1; NF-κB; Proliferation; Colon carcinoma; In vitro AB To investigate effect of poly (ADP-ribose) polymerase inhibition on the proliferation of CT26 cells in vitro and the mechanism of this effect. CT26 cells were treated with a range of concentrations of 5-Aminoisoquinolin-1-one (PARP inhibitor) in vitro. MTT assays and flow cytometry were used to determine the proliferation and cell cycle distribution, respectively, of the cells. The expression of PARP-1 was investigated by Western blot. The binding of Nuclear Factor-κB to DNA was detected by electrophoretic mobility shift assay. The proliferation of CT26 cells was significantly inhibited by 5-AIQ induced PARP inhibition in a dose-dependent manner. Proliferation was inhibited by 17.5% at 100 μM 5-AIQ, by 27.6% at 500 μM 5-AIQ and by 39.9% at 1000 μM (P<0.05). After treatment with 5-AIQ, the proportion of cells in G0/Gl phases increased and the proportion of cells in S phase decreased. The expression of PARP-1 was attenuated in 5-AIQ-treated CT26 cells (P<0.05) and the binding of NF-κB to DNA binding was similarly diminished (P<0.05). These results suggest that PARP inhibition reduced the proliferation of CT26 cells in vitro and influences the cell cycle. This inhibition is mediated by inhibiting PARP-1, which then diminishes the activity of NF-κB. C1 [Cai, Li] Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. [Threadgill, D Michael] University of Bath, Department of Pharmacy & Pharmacology, BA2 7AY Claverton Down Bath, UK. [Wang, Yalan] Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. [Li, Ming] Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. RP Wang, Y (reprint author), Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. EM wangyalan074@126.com CR Cuzzocrea S, Mazzon E, Di Paola R et al, 2004, 5-Aminoisoquinolinone reduces colon injury by experimental colitis. Naunyn Schmiedebergs Arch Pharmacol 370:464–473 Mota-Filipe H, Sepodes B, McDonald MC et al, 2002, The novel PARP inhibitor 5-aminoisoquinolinone reduces the liver injury caused by ischemia and reperfusion in the rat. Med Sci Monit 8: BR444–453 Cuzzocrea S, McDonald MC, Mazzon E et al, 2002, Effects of 5- aminoisoquinolin-one, a water-soluble, potent inhibitor of the activity of poly, ADP-ribose, polymerase, in a rodent model of lung injury. Biochem Pharmacol 63:293–304 Nosho K, Yamamoto H, Mikami M et al, 2006, Overexpression of poly(ADP-ribose, polymerase-1, PARP-1, in the early stage of colorectal carcinogenesis. Eur J Cancer 42:2374–81 Lockett KL, Hall MC, Xu J, 2004, The ADPRT V762A genetic variant contributes to prostate cancer susceptibility and deficient enzyme function. Cancer Res 64:6344–6348 Albert JM, Cao C, Kim KW et al, 2007, Inhibition of poly(ADPribose, polymerase enhances cell death and improves tumor growth delay in irradiated lung cancer models. Clin. Cancer Res. 13:3033–3042 Kang YH, Yi MJ, Kim MJ et al, 2004, caspase-independent cell death by arsenic trioxide in human cervical cancer cells: reactive oxygen species-mediated poly(ADP-ribose, polymerase-1 activation signals apoptosis-inducing factor release from mitochondria. Cancer Res. 64:8960–8967 Hao LX, Wang YL, Li YY, 2006, Correlation of PARP expression with P-selectin and ICAM-1 expression in colorectal carcinoma. Basic Med Sci Clinics 26:882–887 Woon ECY, Threadgill MD, 2005, Poly(ADP-ribose)polymerase inhibition–Where now? Current Med Chem 12:2373–2392 Rajesh M, Mukhopadhyay P, Godlewski G et al, 2006, Poly, ADP-ribose)polymerase inhibition decreases angiogenesis. Biochem Biophys Res Commun 350:1056–1062 McDonald MC, Mota-Filipe H, Wright JA et al, 2000, Effects of 5-aminoisoquinolinone, a water-soluble, potent inhibitor of the activity of poly, ADP-ribose, polymerase on the organ injury and dysfunction caused by haemorrhagic shock. Br J Pharmacol 130:843–850 Zor T, Selinger Z, 1996, Linearization of the Bradford protein assay increases its sensitivity: theoretical and experimental studies. Anal Biochem 236:302–308 Ding HL, Li F, Xu MT et al, 2002, The effect of inhibiting nuclear factor-kappa B on the diabetic nephropathy. Chinese J Intern Med 41:605–609 Lu JP, Sun H, Cao C et al, 2004, InfIuence of nuclear factor˜κB decoy transfection on nuclear factor˜κB activity and vascular endothelial growth factor/urokinase-type plasminogen activator/ intercellular cell adhesion molecule-1 level of SKOV3 cells. Chinese J Obstet Gvneco 39:533–537 Genovese T, Mazzon E, Muia C et al, 2005, Inhibitors of poly, ADP-ribose, polymerase modulate signal transduction pathways and secondary damage in experimental spinal cord trauma. J Pharmacol Exp Ther 312:449–457 Hao LX, Wang YL, Cai L et al, 2007, Inhibitory effect of 5- aminoisoquinolinone on PARP activity in colon carcinoma cell line HT-29. Chinese J Cancer 26:566–571 Nakajima H, Nagaso H, Kakui N et al, 2004, Critical role of the automodification of poly(ADP-ribose, polymerase-1 in nuclear factor-κB-dependent gene expression in primary cultured mouse glial cells. J Biol Chem 279:42774–42786 Yu LL, Yu HG, Yu JP et al, 2004, Nuclear factor-κB regulates cyclooxygenase-2 expression and cell proliferation in human colorectal carcinoma tissue. Eksp Onkol 26:40–47 Greten FR, Eckmann L, Greten TF et al, 2004, IKKbeta links inflammation and tumorigenesis in a mouse model of colitisassociated cancer. Cel1 118:285–296 Kitamura T, Sekimata M, Kikuchi SI et al, 2005, Involvement of poly(ADP-ribose)polymerase 1 in ERBB2 expression in rheumatoid synovial cells. Am J Physiol Cell Physsiol 289:C82–C83 Wu B, Shi D, Yang GK et al, 2004, Expression of nuclear factor-κB, C-myc and ICAM -1 in human colorectal cancer and its significance. Chinese J Clin Oncol Rehabilitation 11:102–111 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 323 EP 328 DI 10.1007/s12253-008-9116-y PG 6 ER PT J AU Takacs, T Paszt, A Szentpali, K Ormandi, K Lazar, M Palka, I Kahan, Zs Lazar, Gy AF Takacs, Tibor Paszt, Attila Szentpali, Karoly Ormandi, Katalin Lazar, Mate Palka, Istvan Kahan, Zsuzsanna Lazar, Gyorgy TI Importance of Sentinel Lymph Node Biopsy in Surgical Therapy of in situ Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Ductal in situ breast cancer; Microinvasion; Isotope localization; Sentinel lymph node biopsy; Lymph node metastasis ID Ductal in situ breast cancer; Microinvasion; Isotope localization; Sentinel lymph node biopsy; Lymph node metastasis AB The aim of this retrospective study was to determine the rate of sentinel lymph node (SLN) positivity in patients with a final diagnosis of ductal in situ cancer (DCIS) of the breast. Between October 2002 and January 2007, 57 patients with DCIS underwent wide excision after radio-guided lesion localization; 53 of them (53/57, 93%) had participated in simultaneous SLN mapping. SLNs were analysed by 250-micron step-sectioning with haematoxylin and eosin staining and immunohistochemical evaluation. The histologic investigation verified pure breast DCIS in 44 cases (44/57, 77.2%), DCIS with microinvasion in eight cases (8/57, 14%) and lobular in situ breast cancer in five cases (5/57, 8.8%). SLNs were identified in 49 cases (49/53, 92.5%) and removed in 48 cases (48/53, 90.6%), i.e. an average of 1.6 SLNs per patient. In four patients (4/53, 7.6%), the SLN biopsy was unsuccessful because of the failure of the radiocolloid substance to migrate. In these cases, axillary sampling was performed. In one case (1/53, 1.9%), only a parasternal SLN was detected; this was not removed. Histologic analysis of the SLNs and the axillary lymph nodes with haematoxylin and eosin or cytokeratin immunohistochemistry did not prove the presence of metastases. The international data and our present results suggest that routine SLN biopsy is not to be recommended in pure DCIS cases. If the final histology verifies an invasive or microinvasive tumour, or if mastectomy is to be performed, SLN mapping is suggested. C1 [Takacs, Tibor] University of Szeged, Department of Surgery, Pecsi u. 4, H-6720 Szeged, Hungary. [Paszt, Attila] University of Szeged, Department of Surgery, Pecsi u. 4, H-6720 Szeged, Hungary. [Szentpali, Karoly] University of Szeged, Department of Surgery, Pecsi u. 4, H-6720 Szeged, Hungary. [Ormandi, Katalin] University of Szeged, Department of RadiologySzeged, Hungary. [Lazar, Mate] University of Szeged, Department of RadiologySzeged, Hungary. [Palka, Istvan] University of Szeged, Department of PathologySzeged, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of OncotherapySzeged, Hungary. [Lazar, Gyorgy] University of Szeged, Department of Surgery, Pecsi u. 4, H-6720 Szeged, Hungary. RP Lazar, Gy (reprint author), University of Szeged, Department of Surgery, H-6720 Szeged, Hungary. EM lg@surg.szote.u-szeged.hu CR Cody HS 3rd, Klauber-DeMore N, Borgen PI, Van Zee KJ, 2001, Is it really duct carcinoma in situ? Ann surg oncol 8:617–619 Klauber-DeMore N, Tan LK, Liberman L, Kaptain S, Fey J, Borgen P, Heerdt A, Montgomery L, Paglia M, Petrek JA, Cody HS, Van Zee KJ, 2000, Sentinel lymph node biopsy: is it indicated in patiens with high-risk ductal carcinoma-in-situ and ductal carcinoma-in-situ with microinvasion? Ann surg oncol 7:636–642 Mittendorf EA, Arciero CA, Gutchell V, Hooke J, Shriver CD, 2005, Core biopsy diagnosis of ductal carcinoma in situ: an indication for sentinel lymph node biopsy. Curr Surg 62:253–257 Moran CJ, Kell MR, Kerin MJ, 2005, The role of sentinel lymph node biopsy in ductal carcinoma in situ. Eur j surg oncol 31:1105–1111 Zavagno G, Carcoforo P, Marconato R, Franchini Z, Scalco G, Burelli P, Pietrarota P, Lise M, Mencarelli R, Capitanio G, Ballarin A, Pierobon ME, Marconato G, Nitti D, 2005, Role of axillary sentinel lymph node biopsy in patient with pure ductal carcinoma in situ of the breast. BMC Cancer 5:28–34 Renyi-Vamos F, Peley G, Bidlek M, Sinkovics I, Szabo E, Keresztes S, Farkas E, Orosz Z, Koves I, 2003, Nem tapinthato emloelvaltozasok radioizotopos jeloleses eltavolitasa egyideju orszemnyirokcsomo-biopsziaval. Magy Seb 56:9–15 Varga L, Bende S, Botos A, Liptay PW, Papp L, Matai E, Kerekes M, Minik K, Forster G, 2004, Az emlosebeszet valtozasa, ot ev, 1998–2002, retrospektiv elemzese alapjan. Magy Seb 57:13–18 Intra M, Zurrida S, Maffini F, Sonzogni A, Trifiri G, Gennari R, Arnone P, Bassani G, Opazo A, Paganelli G, Viale G, Veronesi U, 2003, Sentinel lymph node metastasis in microinvasive breast cancer. Ann Surg Oncol 10:1160–1165 Stuart K, Boyages J, Brennan M, Ung O, 2005, Ductal carcinoma in situ—management update. Aust Fam Physician 34:949–954 Broekhuizen LN, Wijsman JH, Peterse JL, Rutgers EJ, 2006, The incidence and significance of micrometastases in lymph nodes of patients with ductal carcinoma in situ and T1a carcinoma of the breast. Eur J Surg Oncol 32:502–506 Kelly TA, Kim JA, Patrick R, Grundfest S, Crowe JP, 2003, Axillary lymph node metastases in patient with a final diagnosis of ductal carcinoma in situ. Am J Surg 186:368–370 Veronesi P, Intra M, Vento AR, Naninato P, Caldarella P, Paganelli G, Viale G, 2005, Sentinel lymph node biopsy for localised ductal carcinoma in situ? The Breast 14:520–522 Torok K, Peley G, Matrai Z, Bidlek M, Szabo E, Sinkovics I, Polgar C, Farkas E, Orosz Z, Koves I, 2006, The role of sentinel lymph node biopsy for staging patients with ductal carcinoma in situ of the breast. Magy Seb 59:173–178 Cserni G, Bianchi S, Boecker W, Decker T, Lacerda M, Rank F, Wells CA, 2005, Improving the reproductibility of diagnosing micrometastases an isolated tumor cells. Cancer 103:358– 367 El-Tamer M, Chun J, Gill M, Bassi D, Lee S, Hibshoosh H, Mansukhani M, 2005, Incidence and clinical significance of lymph node metastasis detected by cytokeratin immunhistochemical staining in ductal carcinoma in situ. Ann Surg Oncol 12:254–259 Dupont EL, McCann C, Cox CE, 1999, Ductal carcinoma in situ of the breast cancer control 6(3):264–271 Schwartz GF, Solin LJ, Olivotto IA, Ernster VL, Pressman P, Consensus Conference Committee, 2000, Consensus conference on the treatment of in situ ductal carcinoma of the breast April 22– 25. 1999. Cancer 88:946–954 Intra M, Veronesi P, Mazzarol G, Galimberti V, Luini A, Sacchini V, Trifiro G, Gentilini O, Pruneri G, Naninato P, Torres F, Paganelli G, Viale G, Veronesi U, 2003, Axillary sentinel lymph node biopsy in patients with pure ductal carcinoma in situ of the breast. Archives of Surgery 138:309–313 Leidenius M, Salmenkivi K, von Smitten K, Heikkila P, 2006, Tumour-positive sentinel node findings in patients with ductal carcinoma in situ. J Surg Oncol 94:380–384 Mabry H, Giuliano AE, Silverstein MJ, 2006, What is the value of axillary dissection or sentinel node biopsy in patients with ductal carcinoma ins situ? Am J Surg 192:455–457 Katz A, Gage I, Evans S, Shaffer M, Fleury T, Smith F, Flax R, Drogula C, Petrucci P, Magnant C, 2006, Sentinel lymph node positivity of patients with ductal carcinoma in situ or microinvasive breast cancer. Am J Surg 191:761–766 Wilkie C, White L, Dupont E, Cantor A, Cox CE, 2005, An update of sentinel lymph node mapping in patients with ductal carcinoma in situ. Am J Surg 190:563–566 Farkas EA, Stolier AJ, Teng SC, Bolton JS, Fuhrman GM, 2004, An argument against routine sentinel node mapping for DCIS. Am Surg 70:13–18 Camp R, Feezor R, Kasraeian A, Cendan J, Schell S, Wilkinson E, Copeland E, Lind S, 2005, Sentinel lymph node biopsy for ductal carcinoma in situ: an evolving approach at the University of Florida. Breast J 11:394–397 Zavagno G, Belardinelli V, Marconato R, Carcoforo P, Franchini Z, Scalco G, Burelli P, Pietrarota P, Mencarelli R, Marconato G, Nitti D, 2007, Sentinel lymph node metastasis from mammary ductal carcinoma in situ with microinvasion. The Breast 16:146–151 Cserni G, Ambrozay E, Serenyi P, Bori R, Lorincz M, Lorand K, 2005, A nem operativ patologiai emlodiagnosztika eredmenyei. Magy Radiol 79:178–183 Zujewski J, Eng-Wong J, 2005, Sentinel lymph node biopsy in the management of ductal carcinoma in situ. Clin Breast Cancer 6:216–222 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 329 EP 333 DI 10.1007/s12253-008-9123-z PG 5 ER PT J AU Reti, A Barna, G Pap, Adleff, V Komlosi, LV Jeney, A Kralovanszky, J Budai, B AF Reti, Andrea Barna, Gabor Pap, Eva Adleff, Vilmos Komlosi, L Viktor Jeney, Andras Kralovanszky, Judit Budai, Barna TI Enhancement of 5-Fluorouracil Efficacy on High COX-2 Expressing HCA-7 Cells by Low Dose Indomethacin and NS-398 but not on Low COX-2 Expressing HT-29 Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE COX-2; PGE2; HT-29 and HCA-7 cells; 5-fluorouracil; Indomethacin; NS-398; Cell cycle phase ID COX-2; PGE2; HT-29 and HCA-7 cells; 5-fluorouracil; Indomethacin; NS-398; Cell cycle phase AB The antiproliferative effect of 5-fluorouracil (5-FU) in the presence of low dose non-steroidal antiinflammatory drugs (NSAIDs) on high cyclooxygenase-2 (COX-2)-expressing HCA-7 and low COX-2-expressing HT-29 colon carcinoma cell lines was investigated. Pharmacogenetic parameters were studied to characterize the 5-FU sensitivity of the two cell lines. Thymidylate synthase (TS) and methylenetetrahydrofolate reductase (MTHFR) polymorphisms were determined by PCR analysis. Cell proliferation was measured by SRB assay, cell cycle distribution and apoptosis by FACS analysis. Cyclooxygenase expression was detected by Western blot and also by fluorescence microscopy. Prostaglandin E2 (PGE2) levels were investigated with ELISA kit. The HT-29 cell line was found to be homozygous for TS 2R and 1494ins6 and T homozygous for MTHFR 677 polymorphisms predicting high 5-FU sensitivity (IC50: 10 μM). TS 3R homozygosity, TS 1496del6 and MTHFR 677CT heterozygosity may explain the modest 5-FU sensitivity (IC50: 1.1 mM) of the HCA-7 cell line. Indomethacin and NS-398 (10 μM and 1.77 μM, respectively) reduced the PGE2 level in HCA-7 cells (>90%). Low concentrations of NSAIDs without antiproliferative potency increased the S-phase arrest and enhanced the cytotoxic action of 5-FU only in HCA-7 cells after 48-hours treatment. The presented data suggested that the enhancement of 5-FU cytotoxicity by indomethacin or NS-398 applied in low dose is related to the potency of NSAIDs to modulate the cell-cycle distribution and the apoptosis; however, it seems that this effect might be dependent on cell phenotype, namely on the COX-2 expression. C1 [Reti, Andrea] National Institute of OncologyBudapest, Hungary. [Barna, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Pap, Eva] National Institute of OncologyBudapest, Hungary. [Adleff, Vilmos] National Institute of OncologyBudapest, Hungary. [Komlosi, L Viktor] National Institute of OncologyBudapest, Hungary. [Jeney, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kralovanszky, Judit] National Institute of OncologyBudapest, Hungary. [Budai, Barna] National Institute of OncologyBudapest, Hungary. RP Kralovanszky, J (reprint author), National Institute of Oncology, Budapest, Hungary. EM kralo@oncol.hu CR Prescott SM, Fitzpatrick FA, 2000, Cyclooxygenase-2 and carcinogenesis. Biochim Biophys Acta 1470:69–78 Kawai N, Tsujii M, Tsuji S, 2002, Cyclooxygenases and colon cancer. Prostaglandins Other Lipid Mediat 68–69:187–196 Zha S, Yegnasubramanian V, Nelson WG et al, 2004, Cyclooxygenases in cancer: progress and perspective. Cancer Lett 215: 1–20 Rishikesh MK, Sadhana SS, 2003, Prostaglandins and cyclooxygenase: Their probable role in cancer. Indian J Pharmacol 35:3–12 Weckbecker G, 1991, Biochemical pharmacology and analysis of fluoropyrimidines alone and in combination with modulators. Pharmac Ther 50:367–424 Petak I, Tillman DM, Houghton JA, 2000, p53 dependence of Fas induction and acute apoptosis in response to 5-fluorouracilleucovorin in human colon carcinoma cell lines. Clin Cancer Res 6:4432–4441 Adeyemo D, Imtiaz F, Toffa S et al, 2001, Antioxidants enhance the susceptibility of colon carcinoma cells to 5-fluorouracil by augmenting the induction of the bax protein. Cancer Lett 164: 77–84 Ponthan F, Wickstrom M, Gleissman H et al, 2007, Celecoxib prevents neuroblastoma tumor development and potentiates the effect of chemotherapeutic drugs in vitro and in vivo. Clin Cancer Res 13:1036–1044 Mazhar D, Ang R, Waxman J, 2006, COX inhibitors and breast cancer. Br J Cancer 94:346–350 Mercadante S, 2001, The use of anti-inflammatory drugs in cancer pain. Cancer Treat Rev 27:51–61 Minter HA, Eveson JW, Huntley S et al, 2003, The cyclooxygenase 2-selective inhibitor NS398 inhibits proliferation of oral carcinoma cell lines by mechanisms dependent and independent of reduced prostaglandin E2 synthesis. Clin Cancer Res 9:1885–1897 Ou YC, Yang CR, Cheng CL et al, 2007, Indomethacin induces apoptosis in 786-O renal cell carcinoma cells by activating mitogenactivated protein kinases and AKT. Eur J Pharmacol 563:49–60 Zhang M, Abe Y, Matsushima T et al, 2005, Selective cyclooxygenase 2 inhibitor NS-398 induces apoptosis in myeloma cells via a Bcl-2 independent pathway. Leuk Lymphoma 46:425–433 Aggarwal S, Taneja N, Lin L et al, 2000, Indomethacin-induced apoptosis in esophageal adenocarcinoma cells involves upregulation of Bax and translocation of mitochondrial cytochrome C independent of COX-2 expression. Neoplasia 2:346–356 Shao J, Sheng H, Inoue H et al, 2000, Regulation of constitutive cyclooxygenase-2 expression in colon carcinoma cells. J Biol Chem 275:33951–33956 Hitre E, Budai B, Adleff V et al, 2005, Influence of thymidylate synthase gene polymorphisms on the survival of colorectal cancer patients receiving adjuvant 5-fluorouracil. Pharmacogenet Genomics 15:723–730 Frosst P, Blom HJ, Milos R et al, 1995, A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet 10:111–113 Katona C, Timar F, Jeney A et al, 1999, Modulation of 5- fluorouracil by 5-ethyl-2’-deoxyuridine on cell lines expressing different dihydropyrimidine dehydrogenase activities. Anticancer Drugs 10:561–567 Stubbs VE, Schratl P, Hartnell A et al, 2002, Indomethacin causes prostaglandin D(2)-like and eotaxin-like selective responses in eosinophils and basophils. J Biol Chem 277:26012–26020 Barnett J, Chow J, Ives D, 1994, Purification, characterization and selective inhibition of human prostaglandin G/H synthase 1 and 2 expressed in the baculovirus system. Biochim Biophys Acta 1209:130–139 Sanchez-Alcazar JA, Bradbury DA, Pang L, Knox AJ, 2003, Cyclooxygenase, COX, inhibitors induce apoptosis in non-small cell lung cancer through cyclooxygenase independent pathways. Lung Cancer 40:33–44 Han JH, Roh MS, Park CH et al, 2004, Selective COX-2 inhibitor, NS-398, inhibits the replicative senescence of cultured dermal fibroblasts. Mech Ageing Dev 125:359–366 Kralovanszky J, Katona C, Jeney A et al, 1999, 5-ethyl-2’- deoxyuridine, a modulator of both antitumour action and pharmacokinetics of 5-fluorouracil. J Cancer Res Clin Oncol 125:675–684 Beck A, Etienne MC, Cheradame S et al, 1994, A role for dihydropyrimidine dehydrogenase and thymidylate synthase in tumor sensitivity to fluorouracil. Eur J Cancer 30A:1517–1522 Etienne MC, Ilc K, Formento JL et al, 2004, Thymidylate synthase and methylenetetrahydrofolate reductase gene polymorphisms: relationships with 5-fluorouracil sensitivity. Br J Cancer 90:526–534 Duffy CP, Elliott CJ, O’Connor RA et al, 1998, Enhancement of chemotherapeutic drug toxicity to human tumour cells in vitro by a subset of non-steroidal anti-inflammatory drugs, NSAIDs). Eur J Cancer 34:1250–1259 Bennett A, Gaffen JD, Melhuish PB, Stamford IF, 1987, Studies on the mechanism by which indomethacin increases the anticancer effect of methotrexate. Br J Pharmacol 91:229–235 Ogino M, Hanazono M, 1999, Indomethacin preferentially augments 5-fluorouracil cytotoxicity in Colon 26 tumors by increasing the intracellular inflow of 5-fluorouracil. Int J Clin Oncol 4:22–25 Ogino M, Minoura S, 2001, Indomethacin increases the cytotoxicity of cis-platinum and 5-fluorouracil in the human uterine cervical cancer cell lines SKG-2 and HKUS by increasing the intracellular uptake of the agents. Int J Clin Oncol 6:84–89 Totzke G, Schulze-Osthoff K, Janicke U, 2003, Cyclooxygenase- 2, COX-2, inhibitors sensitize tumor cells specifically to death receptor-induced appoptosis independently of COX-2 inhibition. Oncogene 22:8021–8030 O’Callaghan G, Kelly J, Shanahan F, Houston A, 2008, Prostaglandin E2 stimulates Fas ligand expression via the EP1 receptor in colon cancer cells. Br J Cancer 99(3):502–512 Yasumaru M, Tsuji S, Tsujii M et al, 2003, Inhibition of angiotensin II activity enhanced the antitumor effect of cyclooxygenase- 2 inhibitors via insulin-like growth factor I receptor pathway. Cancer Res 63(20):6726–6734 Touhey S, O’Connor R, Plunkett S et al, 2002, Structure-activity relationship of indomethacin analogues for MRP-1, COX-1 and COX-2 inhibition identification of novel chemotherapeutic drug resistance modulators. Eur J Cancer 38:1661–1670 Kobayashi S, Okada S, Yoshida H, Fujimura S, 1997, Indomethacin enhances the cytotoxicity of VCR and ADR in human pulmonary adenocarcinoma cells. Tohoku J Exp Med 181:361–370 Eichele K, Ramer R, Hinz B, 2008, Decisive role of cyclooxygenase- 2 and lipocalin-type prostaglandin D synthase in chemotherapeutics-induced apoptosis of human cervical carcinoma cells. Oncogene 27:3032–3044 Mizutani Y, Kamoi K, Ukimura O et al, 2002, Synergistic cytotoxicity and apoptosis of JTE-522, a selective cyclooxygenase-2 inhibitor, and 5-fluorouracil against bladder cancer. J Urol 168: 2650–2654 Smakman N, Schaap N, Snijckers CMJT et al, 2005, NS-398, a selective cyclooxygenase-2 inhibitor, reduces experimental bladder carcinoma outgrowth by inhibiting tumor cell proliferation. Urology 66:434–440 Rodrigues NR, Rowan A, Smith ME et al, 1990, p53 mutations in colorectal cancer. Proc Natl Acad Sci USA 87:7555–7559 Liu Y, Bodmer WF, 2006, Analysis of P53 mutations and their expression in 56 colorectal cancer cell lines. Proc Natl Acad Sci USA 103:976–981 Yip-Schneider MT, Sweeney CJ, Jung SH et al, 2001, Cell cycle effects of nonsteroidal anti-inflammatory drugs and enhanced growth inhibition in combination with gemcitabine in pancreatic carcinoma cells. J Pharmacol Exp Ther 298:976–985 Meade EA, Smith WL, DeWitt DL, 1993, Differential inhibition of prostaglandin endoperoxide synthase, cyclooxygenase, isozymes by aspirin and other non-steroidal anti-inflammatory drugs. J Biol Chem 268:6610–6614 Lee SH, Lee MY, Han HJ, 2008, Short-period hypoxia increases mouse embryonic stem cell proliferation through cooperation of arachidonic acid and PI3K/Akt signalling pathways. Cell Prolif 41:230–247 ZhangH,YeY, Bai Z,Wang S, 2008, The COX-2 selective inhibitorindependent COX-2 effect on colon carcinoma cells is associated with the Delta1/Notch1 pathway. Dig Dis Sci 53:2195–2203 Oguri T, Bessho Y, Achiwa H et al, 2007, MRP8/ABCC11 directly confers resistance to 5-fluorouracil. Mol Cancer Ther 6:122–127 Chen ZS, Guo Y, Belinsky MG et al, 2005, Transport of bile acids, sulfated steroids, estradiol 17-beta-D-glucuronide, and leukotriene C4 by human multidrug resistance protein 8, ABCC11). Mol Pharmacol 67:545–557 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 335 EP 344 DI 10.1007/s12253-008-9126-9 PG 10 ER PT J AU Aktas, S Kargi, A Olgun, N Diniz, G Erbay, A Vergin, C AF Aktas, Safiye Kargi, Aydanur Olgun, Nur Diniz, Gulden Erbay, Ayse Vergin, Canan TI Prognostic Significance of Cell Proliferation and Apoptosis-Regulating Proteins in Epstein-Barr Virus Positive and Negative Pediatric Non-Hodgkin’s Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pediatric non-Hodgkin’s lymphoma; Epstein Barr virus; Apoptosis; Proliferation; Prognosis ID Pediatric non-Hodgkin’s lymphoma; Epstein Barr virus; Apoptosis; Proliferation; Prognosis AB Apoptosis-related proteins and proliferation activity and their relationship with Epstein-Barr Virus (EBV) are contemporary issues in pediatric non-Hodgkin’s lymphoma (pNHL). In this study prognostic or pathogenetic role of EBV latent infection, proliferating activity, and apoptosisregulating proteins in pNHL were explored. EBV-EBER, lmp-1, ki-67, bcl-2, survivin, bax, fas, c-myc, p53 and apoptotic index by TUNEL method were explored in 70 pNHL cases and evaluated statistically. Of the 70 cases evaluated, 24 were female and 46 were male. Seven cases were stage I/II and 63 cases were stage III/IV. The mean age was 7.16±3.72(1–15). EBV was positive in (25.7%) cases. Overall survival was 82%, while event free survival was 75%. Bax was expressed in 40% of the cases, while the expression of bcl-2,was 50%, survivin 42.9%, p53 8.6%, fas 18.6% and c-myc in 45.7%. Mean apoptotic index was 131.29±96.69 per 5,000 cells. Mean proliferation index was 55.97% (12–92%). Fas positivity was high in EBV positive cases (p=0.0001). EBV positivity was not related with prognosis. Apoptotic index was found to be an independent prognostic factor (p=0.017). Our results suggest that apoptosis-regulating proteins have a role in the pathogenesis of pNHL. EBV was correlated with apoptotic index and fas, bcl-2. No correlation was observed with proliferation index and studied factors. High apoptotic index was related with good prognosis. C1 [Aktas, Safiye] Dokuz Eylul University, Faculty of Medicine, Department of Pathology, Bornova, 125/7 sokak Brickent A-3, 35150 Izmir, Turkey. [Kargi, Aydanur] Dokuz Eylul University, Faculty of Medicine, Department of Pathology, Bornova, 125/7 sokak Brickent A-3, 35150 Izmir, Turkey. [Olgun, Nur] Dokuz Eylul University, Institute of OncologyIzmir, Turkey. [Diniz, Gulden] Dr Behcet Uz Children’s Research Hospital, Specialist in PathologyIzmir, Turkey. [Erbay, Ayse] Dr Behcet Uz Children’s Research Hospital, Specialist in Pediatric OncologyIzmir, Turkey. [Vergin, Canan] Dr Behcet Uz Children’s Research Hospital, Pediatric HematologyIzmir, Turkey. RP Aktas, S (reprint author), Dokuz Eylul University, Faculty of Medicine, Department of Pathology, 35150 Izmir, Turkey. EM safiyeaktas@yahoo.com CR Harris NL, Jaffe ES, Diebold J, Flandrin G, Muller-Hermelink HK, Vardiman J, Lister TA, Bloomfield CD, 2000, The World Health Organization classification of neoplasms of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting—Airlie House, Virginia, November, 1997. Hematol J 1(1):53–66 Naresh KN, Agarwal B, Nathwani BN, Diebold J, McLennan KA, Muller-Hermelink KH, Armitage JO, Weisenburger DD, 2004, Use of the World Health Organization, WHO, classification of non-Hodgkin's lymphoma in Mumbai, India: a review of 200 consecutive cases by a panel of five expert hematopathologists. Leuk Lymphoma 45(8):1569–1577 Cairo MS, Sposto R, Perkins SL, Meadows AT, Hoover-Regan ML, Anderson JR, Siegel SE, Lones MA, Tedeschi-Blok N, Kadin ME, Kjeldsberg CR, Wilson JF, Sanger W, Morris E, Krailo MD, Finlay JL, 2003, Burkitt's and Burkitt-like lymphoma in children and adolescents: a review of the Children's Cancer Group experience. Br J Haematol 120(4):660–670 Campe H, Jaeger G, Abou-Ajram C, Nitschko H, Griebel M, Montoya C, Klare B, Koszinowski U, 2003, Serial detection of Epstein-Barr virus DNA in sera and peripheral blood leukocyte samples of pediatric renal allograft recipients with persistent mononucleosis-like symptoms defines patients at risk to develop post-transplant lymphoproliferative disease. Pediatr Transplant 7, 1):46–52 Straathof KCM, Bollard CM, Rooney CM, Heshlop HE, 2003, Immunotherapy for Epstein-Barr-Virus associated cancers in children. The Oncologist 8:83–98 Preciado MV, Fallo A, Chabay P, Calcagno L, De Matteo E, 2002, Epstein Barr virus-associated lymphoma in HIV-infected children. Pathol Res Pract 198(5):327–332 Cabalo E, Wilkinson R, Lu NT, Shimizu S, Melish M, McClain KL, Jenson HB, Shiramizu B, 2002, Molecular analysis and pathology of a second pediatric HIV-associated Burkitt lymphoma. Pediatr Pathol Mol Med 21(6):525–530 Rao CR, Gutierrez MI, Bhatia K, Fend F, Franklin J, Appaji L, Gallo G, O'Conor G, Lalitha N, Magrath I, 2000, Association of Burkitt's lymphoma with the Epstein-Barr virus in two developing countries. Leuk Lymphoma 39(3–4):329–337 Araujo I, Foss HD, Bittencourt A, Hummel M, Demel G, Mendonca N, Herbst H, Stein H, 1996, Expression of Epstein- Barr virus-gene products in Burkitt's lymphoma in Northeast Brazil. Blood 87(12):5279–5286 Chabay PA, De Matteo EN, Aversa L, Maglio S, Grinstein S, Preciado MV, 2002, Assessment of Epstein-Barr virus association with pediatric non-Hodgkin lymphoma in immunocompetent and in immunocompromised patients in Argentina. Arch Pathol Lab Med 126(3):331–335 Tacyildiz N, Cavdar AO, Ertem U, Oksal A, Kutluay L, Uluoglu O, Lin JC, 1998, Unusually high frequency of a 69-bp deletion within the carboxy terminus of the LMP-1 oncogene of Epstein-Barr virus detected in Burkitt's lymphoma of Turkish children. Leukemia 12, 11):1796–1805 Ertem U, Duru F, Pamir A, Tacyildiz N, Dagdemir A, Akcayoz A, Uluoglu O, Tezic T, 1996, Burkitt's lymphoma in 63 Turkish children diagnosed over a 10 year period. Pediatr Hematol Oncol 13(2):123–134 Nakamura N, Nakamine H, Tamaru J, Nakamura S, Yoshino T, Ohshima K, Abe M, 2002, The distinction between Burkitt lymphoma and diffuse large B-Cell lymphoma with c-myc rearrangement. Mod Pathol 15(7):771–776 Wennborg AD, Altiok E, Moore JP, Ernberg I, Klein G, 1991, Differential c-myc protein expression in Burkitt's lymphomas and EBV-transformed lymphoblastoid lines. Eur J Cancer 27, 12):1643–1645 Klumb CE, Hassan R, Zalcberg IR, Resende LM, Carrico MK, Maia RC, 2004, p53 protein expression does not correlate with EBV status in childhood B non-Hodgkin lymphomas. Pediatr Blood Cancer 43(2):115–119 Hirose Y, Fukushima T, Masaki Y, Shimoyama K, Karasawa H, Ogawa N,Wano Y, 2004, Epstein-Barr virus-associated composite lymphoma composed of peripheral T-cell lymphoma and an anaplastic variant of a diffuse large B-cell type of non-Hodgkin's lymphoma and strongly expressing p53 protein. Int J Hematol 79, 3):260–265 Camilleri-Broet S, Camparo P, Mokhtari K, Hoang-Xuan KH, Martin A, Arborio M, Hauw JJ, Raphael M, 2000, Overexpression of BCL-2, BCL-X, and BAX in primary central nervous system lymphomas that occur in immunosuppressed patients. Mod Pathol 13(2):158–165 Gutierrez MI, Cherney B, Hussain A, Mostowski H, Tosato G, Magrath I, Bhatia K, 1999, Bax is frequently compromised in Burkitt's lymphomas with irreversible resistance to Fas-induced apoptosis. Cancer Res 59(3):696–703 Faqing T, Zhi H, Liqun Y, Min T, Huanhua G, Xiyun D, Ya C, 2005, Epstein-Barr virus LMP1 initiates cell proliferation and apoptosis inhibition via regulating expression of Survivin in nasopharyngeal carcinoma. Exp Oncol 27(2):96–101 Martinez A, Bellosillo B, Bosch F, Ferrer A, Marce S, Villamor N, Ott G, Montserrat E, Campo E, Colomer D, 2004, Nuclear survivin expression in mantle cell lymphoma is associated with cell proliferation and survival. Am J Pathol 164(2):501–510 Nakayama K, Kamihira S, 2002, Survivin an important determinant for prognosis in adult T-cell leukemia: a novel biomarker in practical hemato-oncology. Leuk Lymphoma 43(12):2249–2255 Idenoue S, Hirohashi Y, Torigoe T, Sato Y, Tamura Y, Hariu H, Yamamoto M, Kurotaki T, Tsuruma T, Asanuma H, Kanaseki T, Ikeda H, Kashiwagi K, Okazaki M, Sasaki K, Sato T, Ohmura T, Hata F, Yamaguchi K, Hirata K, Sato N, 2005, A potent immunogenic general cancer vaccine that targets survivin, an inhibitor of apoptosis proteins. Clin Cancer Res 11(4):1474–1482 Frost M, Newell J, Lones MA, Tripp SR, Cairo MS, Perkins SL, 2004, Comparative immunohistochemical analysis of pediatric Burkitt lymphoma and diffuse large B-cell lymphoma. Am J Clin Pathol 121(3):384–392 Sugimoto M, Tahara H, Ide T, Furuichi Y, 2004, Steps involved in immortalization and tumorigenesis in human B-lymphoblastoid cell lines transformed by Epstein-Barr virus. Cancer Res 64, 10):3361–3364 Melo N, Matutes E, Cordone I, Morilla R, Catovsky D, 1992, Expression of Ki-67 nuclear antigen in B and T cell lymphoproliferative disorders. J Clin Pathol 45:660–663 Oyarzo MP, Drakos E, Atwell C, Amin HM, Medeiros LJ, Rassidakis GZ, 2006, Intrinsic apoptotic pathway in anaplastic large cell lymphoma. Hum Pathol 37(7):874–882 ZhaoWL, DaneshpouyME, Mounier N, Briere J, Leboeuf C, Plassa LF, Turpin E, Cayuela JM, Ameisen JC, Gisselbrecht C, Janin A, 2004, Prognostic significance of bcl-xL gene expression and apoptotic cell counts in follicular lymphoma. Blood 103(2):695–697 Rassidakis GZ, Jones D, Lai R, Ramalingam P, Sarris AH, McDonnell TJ, Medeiros LJ, 2003, BCL-2 family proteins in peripheral T-cell lymphomas: correlation with tumour apoptosis and proliferation. J Pathol. 200(2):240–248 Rassidakis GZ, Sarris AH, Herling M, Ford RJ, Cabanillas F, McDonnell TJ, Medeiros LJ, 2001, Differential expression of BCL-2 family proteins in ALK-positive and ALK-negative anaplastic large cell lymphoma of T/null-cell lineage. Am J Pathol 159(2):527–535 Garcia-Sanz R, Montero MV, Diaz MG et al, 1998, Detection of single and associated lesions of the Bcl-1, Bcl-2, Bcl-6, c-myc, p53 and p16 genes in B-cell non-Hodgkin's lymphomas: value of molecular analysis for a better assignment of the histologic subtype. Haematologica 83(3):209–216 Pagnano KB, Vassallo J, Lorand-Metze I, Costa FF, Saad ST, 2001, p53, Mdm2, and c-Myc overexpression is associated with a poor prognosis in aggressive non-Hodgkin's lymphomas. Am J Hematol 67(2):84–92 Bairey O, Zimra Y, Shaklai M, Okon E, Rabizadeh E, 1999, Bcl-2, Bcl-X, Bax, and Bak expression in short- and long-lived patients with diffuse large B-cell lymphomas. Clin Cancer Res 5, 10):2860–2866 Aktas S, Kargi A, Olgun N, Diniz G, Erbay A, Vergin C, 2007, Prognostic significance of cell proliferation and apoptosis-regulating proteins in Epstein-Barr virus positive and negative pediatric Hodgkin lymphoma. Lymphat Res Biol 5(3):175–182 Mancao C, Hammerschmidt W, 2007, Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival. Blood 110(10):3715–21 Nov 15 Anderton E, Yee J, Smith P, Crook T, White RE, Allday MJ, 2008, Two Epstein-Barr virus, EBV, oncoproteins cooperate to repress expression of the proapoptotic tumour-suppressor Bim: clues to the pathogenesis of Burkitt's lymphoma. Oncogene 27, 4):421–433 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 345 EP 350 DI 10.1007/s12253-008-9127-8 PG 6 ER PT J AU Liu, Y Guo, F Dai, M Wang, D Tong, Y Huang, J Hu, J Li, G AF Liu, Yan Guo, Fengjie Dai, Miao Wang, Di Tong, Yongqing Huang, Jian Hu, Jinyue Li, Guancheng TI Gammaaminobutyric Acid A Receptor Alpha 3 Subunit is Overexpressed in Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer; GABRA3; Immunohistochemistry; In silico; RT-PCR ID Cancer; GABRA3; Immunohistochemistry; In silico; RT-PCR AB The identification of tumor-associated antigens, which are specifically expressed in cancer tissues, is very important for immunotherapy of lung cancer. We have combined the in silico screening and experimental verifying to identify genes that are differently expressed in cancers compared with their corresponding normal tissues. Using these methods, we have identified that GABRA3 gene was overexpressed in lung cancer and rarely expressed in other cancers. Furthermore, GABRA3 protein expression was significantly higher in the lower grade of lung cancer. It may compose functional GABA-gated channel with other subunits. This study demonstrated GABRA3 could be a potential biomarker for diagnosis of lung cancer, and GABAA receptors may play an important role in cancer differentiation. C1 [Liu, Yan] Central South University, Xiangya Medical School, Cancer Research Institute, 410078 Changsha, Hunan Province, China. [Guo, Fengjie] Central South University, Xiangya Medical School, Cancer Research Institute, 410078 Changsha, Hunan Province, China. [Dai, Miao] Affiliated Hospital of Weifang Medical University, 261031 Weifang, Shandong Province, China. [Wang, Di] Central South University, Xiangya Hospital, 410078 Changsha, Hunan Province, China. [Tong, Yongqing] Central South University, Xiangya Medical School, Cancer Research Institute, 410078 Changsha, Hunan Province, China. [Huang, Jian] Central South University, Xiangya Medical School, Cancer Research Institute, 410078 Changsha, Hunan Province, China. [Hu, Jinyue] Central South University, Xiangya Medical School, Cancer Research Institute, 410078 Changsha, Hunan Province, China. [Li, Guancheng] Central South University, Xiangya Medical School, Cancer Research Institute, 410078 Changsha, Hunan Province, China. RP Li, G (reprint author), Central South University, Xiangya Medical School, Cancer Research Institute, 410078 Changsha, China. EM libsun@163.com CR Toronto, Canada, Canadian Cancer Society/National Cancer Institute of Canada, 2006, Canadian Cancer Statistics Carbone DP, 1997, The biology of lung cancer. Semin Oncol 24, 4):388–401 Chen Y, Miller C, Mosher R et al, 2003, Identification of cervical cancer markers by cDNA and tissue microarrays. Cancer Res 63, 8):1927–1935 Takahashi M, Yang XJ, Sugimura J et al, 2003, Molecular subclassification of kidney tumors and the discovery of new diagnostic markers. Oncogene 22(43):6810–6818 Reis EM, Ojopi EP, Alberto FL et al, 2005, Large-scale transcriptome analyses reveal new genetic marker candidates of head, neck, and thyroid cancer. Cancer Res 65(5):1693–1699 Vinals C, Gaulis S, Coche T, 2001, Using in silico transcriptomics to search for tumor-associated antigens for immunotherapy. Vaccine 19(17–19):2607–2614 Jin S,Wanjun Y, Ie-Ming S et al, 2006, Identification of BCOX1, a novel gene overexpressed in breast cancer. Biochimica Biophysica Acta 1760(1):62–69 Rieger MA, Ebner R, Bell DR et al, 2004, Identification of a novel mammary-restricted cytochrome P450, CYP4Z1, with overexpression in breast carcinoma. Cancer Res 64(7):2357–2364 Schmitt AO, Specht T, Beckmann G et al, 1999, Exhaustive mining of EST libraries for genes differentially expressed in normal and tumour tissues. Nucleic Acids Res 27(21):4251–4260 Imyanitov EN, Togo AV, Hanson KP, 2004, Searching for cancerassociated gene polymorphisms: promises and obstacles. Cancer Lett 204(1):3–14 Abdel A, Vincent N, Audrey B et al, 2006, Bioinformatic screening of human ESTs for differentially expressed genes in normal and tumor tissues. BMC Genomics 7:94 Neelands TR, Zhang J, Macdonald RL, 1999, GABAA receptors expressed in undifferentiated human teratocarcinoma NT2 cells differ from those expressed by differentiated NT2-N cells. J Neurosci 19(16):7057–7065 Rasola A, Galietta LJ, Gruenert DC et al, 1994, Volume-sensitive chloride currents in four epithelial cell lines are not directly correlated to the expression of the MDR-1 gene. J Biol Chem 269, 2):1432–1436 Chou CY, Shen MR, Wu SN, 1995, Volume-sensitive chloride channels associated with human cervical carcinogenesis. Cancer Res 55(24):6077–6083 Chen LX, Zhu LY, Jacob TJ et al, 2007, Roles of volumeactivated Cl- currents and regulatory volume decrease in the cell cycle and proliferation in nasopharyngeal carcinoma cells. Cell Prolif 40(2):253–267 Shen MR, Droogmans G, Eggermont J et al, 2000, Differential expression of volume-regulated anion channels during cell cycle progression of human cervical cancer cells. J Physiol 529:385– 394 Mao J, Wang L, Fan A et al, 2007, Blockage of volume-activated chloride channels inhibits migration of nasopharyngeal carcinoma cells. Cell Physiol Biochem 19(5–6):249–258 Sarah KJ, Randy SH, 2005, The gamma-aminobutyric acid A receptor π subunit is overexpressed in pancreatic adenocarcinomas. JOP 6(2):136–142 Akio T, Masayo H, Hidetoshi E et al, 2007, γ-aminobutyric acid, GABA, stimulates pancreatic cancer growth through overexpressing GABAA receptor π subunit. Cancer Res 67(20): 9704–9712 Symmans WF, Fiterman DJ, Anderson SK et al, 2005, A singlegene biomarker identifies breast cancers associated with immature cell type and short duration of prior breastfeeding. Endocr Relat Cancer 12(4):1059–1069 Giammarco F, Luca M, Shannon G, 2005, γ-aminobutyric acid inhibits cholangiocarcinoma growth by cyclic AMP-dependent regulation of the protein kinase A/extracellular signal-regulated kinase 1/2 pathway. Cancer Res 65(24):11437–1146 Joseph J, Niggemann B, Zaenker K et al, 2002, The neurotransmitter g-aminobutyric acid is an inhibitory regulator for the migration of SW480 colon carcinoma cells. Cancer Res 62, 22):6467–6479 Azuma H, Inamoto T, Sakamoto T et al, 2003, γ-Aminobutyric acid as a promoting factor of cancer metastasis; induction of matrix metalloproteinase production is potentially its underlying mechanism. Cancer Res 63(23):8090–8096 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 351 EP 358 DI 10.1007/s12253-008-9128-7 PG 8 ER PT J AU Akkiprik, M Sonmez, O Gulluoglu, MB Caglar, BH Kaya, H Demirkalem, P Abacioglu, U Sengoz, M Sav, A Ozer, A AF Akkiprik, Mustafa Sonmez, Ozgur Gulluoglu, M Bahadir Caglar, B Hale Kaya, Handan Demirkalem, Pakize Abacioglu, Ufuk Sengoz, Meric Sav, Aydin Ozer, Ayse TI Analysis of p53 Gene Polymorphisms and Protein Over-expression in Patients with Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p53 polymorphisms; Haplotype; Breast cancer ID p53 polymorphisms; Haplotype; Breast cancer AB p53 polymorphic variants play an important role in the determination of tumor phenotype and characteristics in breast cancer. In this study, we examined three common polymorphisms in p53 gene and their haplotype combinations to assess their potential association with inherited predisposition to breast cancer development, in relations with the protein over-expression and patients’ demographic data. A total of 99 patients with breast cancer and 107 agematched healthy controls were included in the study. Genotypes were determined using PCR-RFLP and DNA sequencing techniques. Evaluation of p53 protein overexpression was also examined by immunohistochemistry. Among three polymorphisms, increased codon 72 Pro allele frequency (p=0.0067) and the presence of Pro allele were found to be significantly associated with breast cancer (p=0.013). A significant risk was also found in subjects with combinations of specific haplotypes and genotypes. Most of breast cancer women especially younger than 50 years carry at least one p53 polymorphism (p=0.001). There was no any association between these three p53 polymorphisms and the protein over-expression, separately or in interaction, with breast cancer. In conclusion, presence of proline allele at codon 72 alone, and its special combinations with other two polymorphisms appear to be a significant risk factor for breast cancer. Determination of well-known p53 polymorphisms might be a good predictor for breast cancer development especially in women younger than 50 years. C1 [Akkiprik, Mustafa] Marmara University, School of Medicine, Department of Medical Biology, Tibbiye Cad, No: 49, Haydarpasa, 34668 Istanbul, Turkey. [Sonmez, Ozgur] Marmara University, School of Medicine, Department of Medical Biology, Tibbiye Cad, No: 49, Haydarpasa, 34668 Istanbul, Turkey. [Gulluoglu, M Bahadir] Marmara University, School of Medicine, Department of General SurgeryIstanbul, Turkey. [Caglar, B Hale] Marmara University, School of Medicine, Department of Radiation OncologyIstanbul, Turkey. [Kaya, Handan] Marmara University, School of Medicine, Department of PathologyIstanbul, Turkey. [Demirkalem, Pakize] Marmara University, School of Medicine, Department of General SurgeryIstanbul, Turkey. [Abacioglu, Ufuk] Marmara University, School of Medicine, Department of Radiation OncologyIstanbul, Turkey. [Sengoz, Meric] Marmara University, School of Medicine, Department of Radiation OncologyIstanbul, Turkey. [Sav, Aydin] Marmara University, School of Medicine, Department of PathologyIstanbul, Turkey. [Ozer, Ayse] Marmara University, School of Medicine, Department of Medical Biology, Tibbiye Cad, No: 49, Haydarpasa, 34668 Istanbul, Turkey. RP Akkiprik, M (reprint author), Marmara University, School of Medicine, Department of Medical Biology, 34668 Istanbul, Turkey. EM makkiprik@marmara.edu.tr CR Dunning AM, Healey CS, Pharoah PD et al, 1999, A systematic review of genetic polymorphisms and breast cancer risk. Cancer Epidemiol Biomarkers Prev 8:843–854 Levine AJ, Momand J, Finlay CA, 1991, The p53 tumour supressor gene. Nature 351:453–456 Nigro JM, Baker SJ, Preisinger AC et al, 1989, Mutations in the p53 gene occur in diverse human tumour types. Nature 342:705–708 Olivier M, Langerod A, Carrieri P et al, 2006, The clinical value of somatic TP53 gene mutations in 1,794 patients with breast cancer. Clin Cancer Res 12:1157–1167 Olivier M, Eeles R, Hollstein M et al, 2002, The IARC TP53 Database: new online mutation analysis and recommendations to users. Hum Mutat 19:607–614 Weston A, Pan CF, Ksieski HB et al, 1997, p53 haplotype determination in breast cancer. Cancer Epidemiol Biomarkers Prev 6:105–112 Wang-Gohrke S, Rebbeck TR, Besenfelder W et al, 1998, p53 germline polymorphisms are associated with an increased risk for breast cancer in German women. Anticancer Res 18:2095– 2099 Papadakis EN, Dokianakis DN, Spandidos DA, 2000, p53 codon 72 polymorphisms as a risk factor in the development of breast cancer. Mol Cell Bio Res Comm 3:389–392 Kalemi TG, Lambropoulos AF, Gueorguiev M et al, 2005, The association of p53 mutations and p53 codon 72, Her 2 codon 655 and MTHFR C677T polymorphisms with breast cancer in Northern Greece. Cancer Lett 222:57–65 Sjalander A, Birgander R, Hallmans G et al, 1996, p53 polymorphisms and haplotypes in breast cancer. Carcinogenesis 17:1313–1316 Dumont P, Leu JIJ, Pietra ACD et al, 2003, The codon 72 polymorphic variants of p53 have markedly different apoptotic potential. Nat Genet 33:357–365 Marin MC, Jost CA, Brooks LA et al, 2000, A common polymorphism acts as an intragenic modifier of mutant p53 behaviour. Nat Genet 25:47–54 Gemignani F, Moreno V, Landi S et al, 2004, A TP53 polymorphism is associated with increased risk of colorectal cancer and with reduced levels of TP53 mRNA. Oncogene 23:1954–1956 Peller S, Kopilova Y, Slutzki S et al, 1995, A novel polymorphism in intron 6 of the human p53 gene: a possible association with cancer predisposition and susceptibility. DNA Cell Biol 14:983–990 Weston A, Wolff MS, Morabia A, 1998, True extended haplotypes of p53: indicators of breast cancer risk. Cancer Genet Cytogenet 102:153–154 Sjalander A, Birgander R, Kivela A et al, 1995, p53 polymorphisms and haplotypes in different ethnic groups. Hum Hered 45:144–149 Wu X, Zhao H, Amos CI et al, 2002, p53 genotypes and haplotypes associated with lung cancer susceptibility and ethnicity. J Natl Cancer Inst 94:681–690 Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer: experience from a long study with long term follow-up. Histopathology 19:403–410 Singletary SE, Allred C, Ashley P et al, 2002, Revision of the American Joint Committee on Cancer Staging System for breast cancer. J Clin Oncol 20:3628–3636 Soong R, Iacopetta BJ, 1997, A rapid and nonisotopic method for the screening and sequencing of p53 gene mutations in formalin-fixed, paraffin-embedded tumors. Mod Pathol 10:252– 258 John SW, Weitzner G, Rozen R et al, 1991, A rapid procedure for extracting genomic DNA from leukocytes. Nucleic Acid Res 19:408 Rockefeller University software database. [Updated 2006 May 11; cited 2007 June 15] Available from: http://linkage. rockefeller.edu/ ott/eh.html Khadang B, Fattahi MJ, Talei A et al, 2007, Polymorphism of TP53 codon 72 showed no association with breast cancer in Iranian women. Cancer Genet Cytogenet 173:38–42 Franekova M, Zubor P, Stanclova A et al, 2007, Association of p53 polymorphisms with breast cancer: a case-control study in Slovak population. Neoplasma 54:155–161 Buyru N, Tigli H, Dalay N, 2003, p53 codon 72 polymorphism in breast cancer. Oncol Rep 10:711–714 Langerod A, Bukholm IRK, Bregard A et al, 2002, The TP53 Codon 72 polymorphism may affect the function of TP53 mutation in breast carcinomas but not in colorectal carcinomas. Cancer Epidemiol Biomarkers Prev 11:1684–1688 Suspitsin EN, Buslov KG, Grigoriev MY et al, 2003, Evidence against involvement of p53 polymorphism in breast cancer predisposition. Int J Cancer 103:431–433 Wang-Gohrke S, Becher H, Kreienberg R et al, 2002, Intron 3 16 bp duplication polymorphism of p53 is associated with an increased risk or breast cancer by the age of 50 years. Pharmacogenetics 12:269–272 Wegman P, Stal O, Askmalm MS et al, 2006, p53 polymorphic variants at codon 72 and the outcome of therapy in randomized breast cancer patients. Pharmacogenet Genomics 16:347–351 Tan XL, Popanda O, Ambrosone CB et al, 2006, Association between TP53 and p21 genetic polymorphisms and acute side effects of radiotherapy in breast cancer patients. Breast Cancer Res Treat 97:255–262 Costa S, Pinto D, Pereira D et al, 2008, Importance of TP53 codon 72 and intron 3 duplication 16 bp polymorphisms in prediction of susceptibility on breast cancer. BMC Cancer 8:32 Bartley AN, Ross DW, 2002, Validation of p53 immunohistochemistry as a prognostic factor in breast cancer in clinical practice. Arch Pathol Lab Med 126:456–458 Rolland P, Spendlove I, Madjid Z et al, 2007, The p53 positive Bcl-2 negative phenotype is an independent marker of prognosis in breast cancer. Int J Cancer 120:1311–1317 Yamashita H, Toyama T, Nishio M et al, 2006, p53 protein accumulation predicts resistance to endocrine therapy and decreased post-relapse survival in metastatic breast cancer. Breast Cancer Res 8:R48 Roos MA, de Bock GH, de Vries J et al, 2007, p53 overexpression is a predictor of local recurrence after treatment for both in situ and invasive ductal carcinoma of the breast. J Surg Res 140:109–114 Kai K, Nishimura R, Arima N et al, 2006, p53 expression status is a significant molecular marker in predicting the time to endocrine therapy failure in recurrent breast cancer: a cohort study. Int J Clin Oncol 11:426–433 Gammon MD, Hibshoosh H, Terry MB et al, 1999, Cigarette smoking and other risk factors in relation to p53 expression in breast cancer among young women. Cancer Epidemiol Biomarkers Prev 8:255–263 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 359 EP 368 DI 10.1007/s12253-008-9129-6 PG 10 ER PT J AU Han, Zd Bi, Xch Qin, Wj He, Hch Dai, Qsh Zou, J Ye, Yk Liang, Yx Zeng, Gh Chen, Zn Zhong, Wd AF Han, Zhao-dong Bi, Xue-cheng Qin, Wei-jun He, Hui-chan Dai, Qi-shan Zou, Jun Ye, Yong-kang Liang, Yu-xiang Zeng, Guo-hua Chen, Zhi-nan Zhong, Wei-de TI CD147 Expression Indicates Unfavourable Prognosis in Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; CD147; Clinical pathology; Diagnosis; Prognosis ID Prostate cancer; CD147; Clinical pathology; Diagnosis; Prognosis AB Extracellular matrix metalloproteinase inducer (EMMPRIN, also named as CD147) is a multifunctional membrane glycoprotein over-expressed in many kinds of human solid tumors. It has been demonstrated to be involved in tumor invasion and angiogenesis. The aim of this study was to analyze the clinicopathological characteristics of the expression of CD147 in human prostate cancer (PCa), and to evaluate its clinical significance in the histologic classification and prognosis of PCa. CD147 protein expression in paraffin-embedded specimens gathered from 62 cases of PCa and 30 cases of benign prostatic hyperplasia (BPH) were detected by the method of immunohistochemistry. The association of CD147 protein expression with the clinicopathological characteristics and with the prognosis of PCa was subsequently assessed. CD147 expression were positively expressed in 51/62 (82.3%) of PCa and 4/30 (13.3%) of BPH cases, respectively. The positive expression rate of CD147 in PCa tissues was significantly higher than that in BPH. The positive expression of CD147 was dramatically associated with TNM grade (p<0.001), the depth of the prostatic wall invasion (p=0.008), GLEASON Score (p=0.001) and Histologic grade (p=0.001). The patients with CD147 expression were associated with a poor prognosis of PCa (p=0.01) and the survival rate of the patients with a strong positive expression of CD147 was the lowest (p=0.01). The results suggest that the expression of CD147 may be an important feature of PCa and the detection of its expression may benefit us in the prediction of the prognosis of PCa. C1 [Han, Zhao-dong] Guangzhou Medical College, First Affiliated Hospital, #1 Pan Fu Road, 510180 Guangzhou, China. [Bi, Xue-cheng] Southern Medical University, #1 Pan Fu Road, 510180 Guangzhou, China. [Qin, Wei-jun] Fourth Military Medical University, Department of Cell Biology & Cell Engineering Research CentreXi’an, China. [He, Hui-chan] Guangzhou Medical College, First Affiliated Hospital, #1 Pan Fu Road, 510180 Guangzhou, China. [Dai, Qi-shan] Guangzhou Medical College, First Affiliated Hospital, #1 Pan Fu Road, 510180 Guangzhou, China. [Zou, Jun] Guangzhou Medical College, First Affiliated Hospital, #1 Pan Fu Road, 510180 Guangzhou, China. [Ye, Yong-kang] Guangzhou Medical College, First Affiliated Hospital, #1 Pan Fu Road, 510180 Guangzhou, China. [Liang, Yu-xiang] Guangzhou Medical College, First Affiliated Hospital, #1 Pan Fu Road, 510180 Guangzhou, China. [Zeng, Guo-hua] Guangzhou Medical College, First Affiliated HospitalGuangzhou, China. [Chen, Zhi-nan] Fourth Military Medical University, Department of Cell Biology & Cell Engineering Research CentreXi’an, China. [Zhong, Wei-de] Guangzhou Medical College, First Affiliated Hospital, #1 Pan Fu Road, 510180 Guangzhou, China. RP Zhong, Wd (reprint author), Guangzhou Medical College, First Affiliated Hospital, 510180 Guangzhou, China. EM wdezhong@21cn.com CR Schrgder FH, Wildhagen MF, ERSPC, 2001, Screening for prostate cancer: evidence and perspectives. BJU Int 88:811–817 Holmberg L, Bill-Axelson A, Helgesen F, 2002, A randomized trial comparing radical prostatectomy with watchful waiting in early prostate cancer. New Eng J Med 347:781–789 Hochreiter WW, 2008, The issue of prostate cancer evaluation in men with elevated prostate-specific antigen and chronic prostatitis. Andrologia 40:130–133 Loeb S, Catalona WJ, 2008, What to do with an abnormal PSA test. Oncologist 13:299–305 Kehinde EO, Maghrebi MA, Anim JT, 2008, The importance of determining the aggressiveness of prostate cancer using serum and tissue molecular markers. Can J Urol 15:3967–3974 Wang L, Wu G, Yu L, Yuan J, Fang F, Zhai Z et al, 2006, Inhibition of CD147 expression reduces tumor cell invasion in human prostate cancer cell line via rna interference. Cancer Biology & Therapy 5:608–614 Gabison EE, Mourah S, Steinfels E et al, 2005, Differential expression of extracellular matrix metalloproteinase inducer, CD147, in normal and ulcerated corneas. Am. J. Pathol 166:209–219 Li Y, Shang P, Qian A et al, 2003, Inhibitory effects of antisense RNA of HAb18G/CD147 on invasion of hepatocellular carcinoma cells in vitro. World J Gastroente 9:2174–2177 Sidhu SS, Mengistab AT, Tauscher AN, LaVail J, Basbaum C, 2004, The microvesicle as a vehicle for EMMPRIN in tumorstromal interactions. Oncogene 23:956–963 Millimaggi D, Mari M, D’Ascenzo S, Carosa E, Jannini EA, Zucker S et al, 2007, Tumor vesicle-associated CD147 modulates the angiogenic capability of endothelial cells. Neoplasia 9:349– 357 Nabeshima K, Iwasaki H, Koga K, Hojo H, Suzumiya J, Kikuchi M, 2006, Emmprin, basigin/CD147): matrix metalloproteinase modulator and multifunctional cell recognition molecule that plays a critical role in cancer progression. Pathol Int. 56:359–367 London CA, Sekhon HS, Arora V, Stein DA, Iversen PL, Devi GR, 2003, A novel antisense inhibitor of MMP-9 attenuates angiogenesis, human prostate cancer cell invasion and tumorigenicity. Cancer Gene Ther 10:823–832 Mi Z, Oliver T, Guo H, Gao C, Kuo PC, 2007, Thrombin-cleaved COOH, -, terminal osteopontin peptide binds with cyclophilin C to CD147 in murine breast cancer cells. Cancer Res 67:4088–4097 Kakehi Y, 2003, Watchful waiting as a treatment option for localized prostate cancer in the PSA era. Jpn J Clin oncol. 33:1–5 Li QQ, Wang WJ, Xu JD, Cao XX, Chen Q, Yang JM et al, 2007, Up-regulation of CD147 and matrix metalloproteinase-2, -9 induced by P-glycoprotein substrates in multidrug resistant breast cancer cells. Cancer Sci 98:1767–1774 Riethdorf S, Reimers N, Assmann V, Kornfeld JW, Terracciano L, Sauter G et al, 2006, High incidence of EMMPRIN expression in human tumors. Int J Cancer 119:1800–1810 Biswas C, Zhang Y, DeCastro R, Guo H, Nakamura T, Kataoka H et al, 1995, The human tumor cell-derived collagenase stimulatory factor, renamed EMMPRIN, is a member of the immunoglobulin superfamily. Cancer Res 55:434–439 Caudroy S, Polette M, Nawrocki-Raby B, Cao J, Toole BP, Zucker S et al, 2002, EMMPRIN-mediated MMP regulation in tumor and endothelial cells. Clin Exp Metastasis 19:697–702 Tang Y, Nakada MT, Kesavan P, McCabe F, Millar H, Rafferty P et al, 2005, Extracellular matrix metalloproteinase inducer stimulates tumor angiogenesis by elevating vascular endothelial cell growth factor and matrix metalloproteinases. Cancer Res 65:3193–3199 Biswas C, 1982, Tumor cell stimulation of collagenase production by fibroblasts. Biochem Biophys Res Commun 109:1026–1034 Cozzi PJ, Wang J, Delprado W, Perkins AC, Allen BJ, Russell PJ et al, 2005, MUC1, MUC2, MUC4, MUC5AC and MUC6 expression in the progression of prostate cancer. Clin Exp Metastasis 22:565–573 Sun J, Hemler ME, 2001, Regulation of MMP-1 and MMP- 2production through CD147/extracellular matrix metalloproteinase inducer interactions. Cancer research 61:2276–2281 Marieb EA, Zoltan-Jones A, Li R, 2004, Emmprin promotes anchorage-independent growth in human mammary carcinoma cells by stimulating hyaluronan production. Cancer Research. 64:1229–1232 Li HG, Xie DR, Shen XM, 2005, Clinicopathological significance of expression of paxillin, syndecan-l and EMMPRIN in hepatocellular carcinoma. World J Gastroenterol 11:1445–1451 Lanyi M, Antonescu CR, 2002, The precrystalline cytoplasmic granules of alveolar soft part sarcoma contain monocarboxylate transporter 1 and CD147. Am J Pathol 160:1215–1221 Seitz C, Djavan B, 2006, Biological markers of prostate cancer. Ann Urol, Paris). 40:329–335 Zucker S, Hymowitz M, Rollo EE et al, 2001, Tumorigenic potential of extracellular matrix metalloproteinase inducer. Am J Pathol 158:1921–1928 Tang Y, Kesavan P, Marian T, 2004, Tumor-stroma interaction: positive feedback regulation of extracellular matrix metalloproteinase inducer, EMMPRIN, expressio and matrix metalloproteinase- dependent generation of soluble EMMPRIN. Mol Cancer Res 2:73–80 Kanekura T, Chen X, Kanzaki T, 2002, Basigin, CD147, is expressed on melanoma cells and induces tumor cell invasion by stimulating production of matrix metalloproteinases by fibroblasts. Int J Cancer 99:520–528 Jia L, Wang H, Qu S, Miao X, Zhang J, 2008, CD147 regulates vascular endothelial growth factor-expression, tumorigenicity, and chemosensitivity to curcumin in hepatocellular carcinoma. IUBMB Life 60:57–63 Tang Y, Nakada MT, Rafferty P, McCabe FL, Millar H, Cunningham M et al, 2006, Regulation of vascular endothelial growth factor expression by EMMPRIN via the PI3K-Akt signaling pathway. Mol Cancer Res 4:371–377 Madigan MC, Kingsley EA, Cozzi PJ, Delprado WJ, Russell PJ, Li Y, 2008, The role of extracellular matrix metalloproteinase inducer protein in prostate cancer progression. Cancer Immunol Immunother 57(9):1367–1379, DOI 10.1007/s00262-008-0473-x NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 369 EP 374 DI 10.1007/s12253-008-9131-z PG 6 ER PT J AU Miltenyi, Zs Toth, J Gonda, A Tar, I Remenyik, Illes, AF Miltenyi, Zsofia Toth, Judit Gonda, Andrea Tar, Ildiko Remenyik, Eva Illes, Arpad TI Successful Immunomodulatory Therapy in Castleman Disease with Paraneoplastic Pemphigus Vulgaris SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Castleman disease; Immunmodulatory; PET/CT ID Castleman disease; Immunmodulatory; PET/CT AB Castleman disease is a rare lymphoproliferative disorder. The clinical signs and symptoms of the disease are primarily mediated by cytokines, especially interleukin-6. We presented the case of a young female. In May 2004, a 30-year-old otherwise healthy looking woman presented with oral ulcerations resistant to topical and systemic antibiotic and antimycotic treatment. Bullous mucosal lichen or pemphigus vulgaris were suspected. Histological examination and direct and indirect immunofluorescence confirmed the diagnosis of pemphigus. Search for neoplasm revealed a retroperitoneal Castleman tumour sized 15×6×5 cm in the abdominal MRI. The tumour was a bleeder, so the removal was partial. Histological examination showed hyalin hypervascular Castleman disease. Considering her young, fertile age and the multicentric Castleman disease, non-cytostatic immunomodulatory therapy was started including steroid, cyclosporine-A and thalidomide treatment. The control abdominal CT showed a small residual tumour on the bladder. The residual tumour was removed in repeated surgery. At this time the histological examination showed transient type tumour between plasma cell and vascular variant. Currently, i.e. 4 years after the onset of the disease. 18FDG PET/CT examination showed low metabolic active mass in the right iliacal region, but our patient had no symptoms or complaints. She is on 200 mg thalidomide a day and no tumour progression can be seen. Castleman disease can be successfully treated with non-cytostatic immunomodulatory therapy. C1 [Miltenyi, Zsofia] University of Debrecen, Medical and Health Center, 3rd Department of MedicineDebrecen, Hungary. [Toth, Judit] University of Debrecen, Department of OncologyDebrecen, Hungary. [Gonda, Andrea] University of Debrecen, Department of OncologyDebrecen, Hungary. [Tar, Ildiko] University of Debrecen, Faculty of Dentistry, Department of PeriodontologyDebrecen, Hungary. [Remenyik, Eva] University of Debrecen, Department of DermatologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Medical and Health Center, 3rd Department of MedicineDebrecen, Hungary. RP Miltenyi, Zs (reprint author), University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Debrecen, Hungary. EM mil03@freemail.hu CR Castleman B, Thowne VW, 1954, Case records of the Massachuetts General Hospital weekly clinicopathological exercises: Case 40011. N Eng J Med 250:26–30 van den Berge M, Pauwels P, Jakimowicz JJ et al, 2002, Hyaline vascular Castleman’s disease: a case report and brief review of the literature. Neth J Med 60(11):444–447, Dec Casper C, 2005, The aetiology and management of Castleman disease at 50 years: translating pathophysiology to patient care. Br J Haematol 129:3–17 Oksenhendler E, Carcelain G, Aoki Y et al, 2000, High levels of human herpesvirus 8 viral load, human interleukin-6, interleukin-10, and C reactive protein correlate with exacerbation of multicentric Castleman disease in HIV-infected patients. Blood 96:2069–2073 Waterston A, Bower M, 2004, Fifty years of multicentric Castleman’s disease. Acta Oncologica 43:698–704 Harris NL, Swerdlow S, Campo E et al, 2008, The World Health Organization, WHO, classification of lymphoid neoplasms: what’s new? Ann Oncol 19(4):119 Anhalt GJ, Kim SC, Stanley JR et al, 1990, Paraneoplastic pemphigus. An autoimmune mucocutaneus disease associated with neoplasia. N Engl J Med 323:1729–1735 Anhalt GJ, 2004, Paraneoplastic pemphigus. J Investig Dermatol Symp Proc 9(1):29–33 Dham A, Peterson BA, 2007, Castleman disease. Curr Opin Hematol 14:354–359 Chorzelski T, Hashimoto T, Maciejewska B et al, 1999, Paraneoplastic pemphigus associated with Castleman tumor, myasthenia gravis, and bronchiolitis obliterans. J Am Acad Dermatol 41, 3):3939–3400 Nishimoto N, Kanakura Y, Aozasa K et al, 2005, Humanized antiinterleukin- 6 receptor antibody treatment of multicentric Castleman disease. Blood 106:2627–2632 Starkey C, Joste N, Lee FC, 2006, Near-total resolution of multicentric Castleman disease by prolonged treatment with thalidomid. Am J Haematol 81:303–304, letter) Lee FC, Merchant SH, 2003, Alleviation of systemic manifestations of multicentric Castleman’s disease by thalidomide. Am J Hematol 73:48–53 Nikolskaia OV, Nousari CH, Anhalt GJ, 2003, Paraneoplastic pemphigus in association with Castleman’s disease. Br J Dermatol 149:1143–1151 Cunha PR, de Oliveira JR, Salles MJ et al, 2004, Pemphigus vulgaris with involvement of the cervix treated using thalidomide therapy. Int J Dermatol 43(9):682–684 Hess G, Wagner V, Kreft A et al, 2006, Effects of bortezomib on pro-inflammatory cytokine levels and transfusion dependency in a patient with multicentric Castleman disease. Brit J Haematol 134:544–549 Dieval C, Bonnet F, Mauclere S et al, 2007, Multicentric Castleman disease: Use of HHV8 viral load monitoring and positron emission tomography during follow-up. Leuk Lymphoma 48(9):1881–1883 Nakamura Y, Tokuyama O, Muso A et al, 2002, Asymptomatic pelvic Castleman disease in an infertile woman: case report. Arch Gynecol Obstet 269:156–158 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 375 EP 381 DI 10.1007/s12253-008-9133-x PG 7 ER PT J AU Varkonyi, J Bajzik, E Fazakas, Sipka, S Karadi, I AF Varkonyi, Judit Bajzik, Edina Fazakas, Adam Sipka, Sandor Karadi, Istvan TI Short or Long Survival in Multiple Myeloma. A Simple Method for Determining the Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prognostic factors; Multiple myeloma; AMWBC score ID Prognostic factors; Multiple myeloma; AMWBC score AB Finding prognostic factors inmultiple myeloma is a real challenge. Several attempts might be found in the literature for that purpose but the results are not satisfactory. Therefore, in the current retorpective study authors analyzed the potential prognostic value of some laboratory data in 104 patients with multiple myeloma. They found the albumin and M-component ratio being lower than 1 and the initial WBC<4,5×109/l correlated strongly with poor prognosis. In addition the low albumin level was not related to albuminuria and that the low WBC was not linked to bone marrow infiltration rate or to antineutrophil antibody formation. On the basis of their experiences authors created an AMWBC score containing A/M and WBC at diagnosis found to be in good correlation to prognosis. Further studies involving more patients are needed to verify the real prognostic value of AMWBC score in multiple myeloma treated with new targeted therapies. C1 [Varkonyi, Judit] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4., 1125 Budapest, Hungary. [Bajzik, Edina] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4., 1125 Budapest, Hungary. [Fazakas, Adam] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4., 1125 Budapest, Hungary. [Sipka, Sandor] University of Debrecen, Medical and Health Center, 3rd Department of MedicineDebrecen, Hungary. [Karadi, Istvan] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4., 1125 Budapest, Hungary. RP Varkonyi, J (reprint author), Semmelweis University, 3rd Department of Internal Medicine, 1125 Budapest, Hungary. EM varkjud@kut.sote.hu CR Attal M, Harorusseau JL, Stoppa AM et al, 1996, A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. NEJM 335:9–7 Kyle RA, 1983, Long term survival in multiple myeloma. NEJM 308:314–316 Durie BGM, Salmon SE, 1975, A clinical staging system for multiple myeloma. Correlation of measured myeloma cell mass with presenting clinical features, response to treatment and survival. Cancer 38:842–854 Greipp PR, Witzig TE, Gonchoroff NJ, 1985, Immunofluorescent plasma cell labeling indices, Li, using a monoclonal antibody, Bu-1). Am J Hematol 20:289 Sipos A, Csortos C, Sipka S et al, 1998, The antigen/receptor specificity of antigegranulocyte antibodies in patients with SLE. Immunol Letters 19:329–334 Ershler WB, Keller ET, 2000, Age-associated increased interleukin- 6 gene expression, late-life diseases, and frailty. Annu Rev Med 51:245–270 Gautier M, Robertson J, Hars V, 1997, Prognostic factors in survival for patients with multiple myeloma from a single institution over 30 years. Blood 90:/10S1/A 4089 Fassa ABT, Spencer T, Sawyer J et al, 2002, Both hypodiploidy and deletion of chromosome 13 independently confer poor prognosis in multiple myeloma. Brit J Haematol 118:1041–1047 Jacobson JL, Hussein MA, Barlogie B et al, 2003, A new staging system for multiple myeloma patients based on the Southwest Oncology Group, SWOG, experience. Brit J Haematol 122:441–450 Blade J, Rozman C, Cervantes F et al, 1989, A new prognostic system for multiple myeloma based on easily available parameters. Brit J Haematol 72:507–511 Symeonidis A, Kouraklis-Symeonidis A, Grouzi E et al, 2002, Determination of Plasma cell screening potential as an index of maturity myelomatous cell and a strong prognostic factor. Leukemia Lymphoma 43:1605–1612 Kyle RA, GertzMA,Witzig TE et al, 2003, Review of 1,027 patients with newly diagnosed multiple myeloma. Mayo Clin Proc 78:21–33 Greipp PR, Miguel JS, Durie BGM et al, 2005, International staging system for multiple myeloma. JCO 23:3412–3420 Mori M, Abe T, Takei T et al, 1992, The significance of myelodysplasia in untreated patients with multiple myeloma. Rhinosho-Keutsueki 33:662–665 Coppelstone JA, Mufti GJ, Hamblin TJ et al, 1986, Immunological abnormalities in myelodysplastic syndromes. Brit J Haematol 63:149–159 Hus I, Dmoszynska A,Manko J et al, 2004, An evaluation of factors predicting long-term response to thalidomide in 234 patients with relapsed or resistant multiple myeloma. Brit J Cancer 91:1873–1879 Anagnostopoulos A, Zervas K, Zomas A et al, 2003, The international prognostic index, IPI, is predictive for survival in refractory or relapsed myeloma patients treated with thalidomide– based regimens. Blood 937A:3492 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 383 EP 387 DI 10.1007/s12253-008-9134-9 PG 5 ER PT J AU Syamala, SV Syamala, V Sreedharan, H Raveendran, BP Kuttan, R Ankathil, R AF Syamala, S Volga Syamala, Vani Sreedharan, Hariharan Raveendran, B Praveenkumar Kuttan, Ratheesan Ankathil, Ravindran TI Contribution of XPD (Lys751Gln) and XRCC1 (Arg399Gln) Polymorphisms in Familial and Sporadic Breast Cancer Predisposition and Survival: An Indian Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Familial breast cancer; Low penetrance genes; Single nucleotide polymorphisms; Survival; XRCC1; XPD ID Familial breast cancer; Low penetrance genes; Single nucleotide polymorphisms; Survival; XRCC1; XPD AB The etiology of a significant proportion of familial breast cancers is still poorly understood, with known high penetrance gene mutations accounting for only a small proportion of the cases. The increased risk of breast cancer for the majority of women with a family history likely reflects shared minor low penetrant genetic factors. In the present case-control study undertaken to examine the influence of DNA damage repair gene polymorphisms in familial and sporadic breast cancer susceptibility, 219 Sporadic and 140 familial breast cancer patients and 367 controls were genotyped using PCRRFLP. Odds Ratios (ORs) and 95% Confidence Intervals (95%CIs) were calculated by unconditional logistic regression adjusted to age. Variant genotypes XRCC1 Arg/Gln or Gln/Gln and XPD Lys/Gln or Gln/Gln increased both familial and sporadic breast cancer susceptibility. However, when the intra group risk was compared, the risk due to the XPD polymorphic genotypes Lys/Gln or Gln/Gln was significantly lower among familial breast cancer patients compared to sporadic breast cancer patients [OR=0.61; 95%CI=0.39–0.94; p value=0.024) whereas the risk implied by XRCC1 variant genotype was not significantly different between the familial and nonfamilial groups of breast cancer patients [OR=0.97; 95%CI=0.63–1.49; p value=0.882]. Both these variant genotypes were not associated with the disease characteristics or survival of either familial or sporadic breast cancer patients. This study represents an addition to previous published work on GSTs from the same study population and substantiates the hypothesis that the impact of the low penetrance gene polymorphisms differ by family history of the disease. C1 [Syamala, S Volga] Regional Cancer Centre, Division of Cancer Research, 695 011 Trivandrum, Kerala, India. [Syamala, Vani] Regional Cancer Centre, Division of Cancer Research, 695 011 Trivandrum, Kerala, India. [Sreedharan, Hariharan] Regional Cancer Centre, Division of Cancer Research, 695 011 Trivandrum, Kerala, India. [Raveendran, B Praveenkumar] Regional Cancer Centre, Division of Cancer Research, 695 011 Trivandrum, Kerala, India. [Kuttan, Ratheesan] Regional Cancer Centre, Division of Radiation Oncology, 695011 Thiruvananthapuram, Kerala, India. [Ankathil, Ravindran] Regional Cancer Centre, Division of Cancer Research, 695 011 Trivandrum, Kerala, India. RP Ankathil, R (reprint author), Regional Cancer Centre, Division of Cancer Research, 695 011 Trivandrum, India. EM rankathil@gmail.com CR Dapic V, Carvalho MA, Monteiro AN, 2005, Breast cancer susceptibility and the DNA damage response. Cancer Control 12:127–136 Helzlsouer KJ, Harris EL, Parshad R, 1996, DNA repair proficiency: potential susceptiblity factor for breast cancer. J Natl Cancer Inst 88:754–755 Jyothish B, Ankathil R, Chandini R et al, 1998, DNA repair proficiency: a potential marker for identification of high risk members in breast cancer families. Cancer Lett 124:9–13 Matullo G, Palli D, Peluso M et al, 2001, XRCC1, XRCC3, XPD gene polymorphisms, smoking and, 32)P-DNA adducts in a sample of healthy subjects. Carcinogenesis 22:1437–1445 Clarkson SG, Wood RD, 2005, Polymorphisms in the human XPD, ERCC2, gene, DNA repair capacity and cancer susceptibility: an appraisal. DNA Repair, Amst, 4:1068–1074 Pachkowski BF, Winkel S, Kubota Y et al, 2006, XRCC1 genotype and breast cancer: functional studies and epidemiologic data show interactions between XRCC1 codon 280 His and smoking. Cancer Res 66:2860–2868 Figueiredo JC, Knight JA, Briollais L et al, 2004, Polymorphisms XRCC1-R399Q and XRCC3-T241M and the risk of breast cancer at the Ontario site of the Breast Cancer Family Registry. Cancer Epidemiol Biomarkers Prev 13:583–591 Costa S, Pinto D, Pereira D et al, 2007, DNA repair polymorphisms might contribute differentially on familial and sporadic breast cancer susceptibility: a study on a Portuguese population. Breast Cancer Res Treat 103:209–217 Dufloth RM, Costa S, Schmitt F et al, 2005, DNA repair gene polymorphisms and susceptibility to familial breast cancer in a group of patients from Campinas, Brazil. Genet Mol Res 4:771–782 Brewster AM, Jorgensen TJ, Ruczinski I et al, 2006, Polymorphisms of the DNA repair genes XPD, Lys751Gln, and XRCC1, Arg399Gln and Arg194Trp): relationship to breast cancer risk and familial predisposition to breast cancer. Breast Cancer Res Treat 95:73–80 IARC, 1989, IARC Monographs on the evaluation of carcinogenic risks to humans, Vol: 43,diesel and gasoline engine exhausts and some nitroarenes. Lyon 41–185. Bohr VA, 1995, DNA repair fine structure and its relations to genomic instability. Carcinogenesis 16:2885–2892 Jiricny J, Nystrom-Lahti M, 2000, Mismatch repair defects in cancer. Curr Opin Genet Dev 10:157–161 Izumi T, Wiederhold LR, Roy G et al, 2003, Mammalian DNA base excision repair proteins: their interactions and role in repair of oxidative DNA damage. Toxicology 193:43–65 Callebaut I, Mornon JP, 1997, From BRCA1 to RAP1: a widespread BRCT module closely associated with DNA repair. FEBS Lett 400:25–30 Hung RJ, Hall J, Brennan P et al, 2005, Genetic polymorphisms in the base excision repair pathway and cancer risk: a HuGE review. Am J Epidemiol 162:925–942 Friedberg EC, 2001, How nucleotide excision repair protects against cancer. Nat Rev Cancer 1:22–33 Coin F, Marinoni JC, Egly JM, 1998, Mutations in XPD helicase prevent its interaction and regulation by p44, another subunit of TFIIH, resulting in Xeroderma pigmentosum, XP, and trichothiodystrophy, TTD, phenotypes. Pathol Biol, Paris, 46:679–680 Seker H, Butkiewicz D, Bowman ED et al, 2001, Functional significance of XPD polymorphic variants: attenuated apoptosis in human lymphoblastoid cells with the XPD 312 Asp/Asp genotype. Cancer Res 61:7430–7434 Qiao Y, Spitz MR, Shen H et al, 2002, Modulation of repair of ultraviolet damage in the host-cell reactivation assay by polymorphic XPC and XPD/ERCC2 genotypes. Carcinogenesis 23:295–299 Au WW, Salama SA, Sierra-Torres CH, 2003, Functional characterization of polymorphisms in DNA repair genes using cytogenetic challenge assays. Environ Health Perspect 111:1843–1850 Lunn RM, Helzlsouer KJ, Parshad R et al, 2000, XPD polymorphisms: effects on DNA repair proficiency. Carcinogenesis 21:551–555 Spitz MR, Wu X, Wang Y et al, 2001, Modulation of nucleotide excision repair capacity by XPD polymorphisms in lung cancer patients. Cancer Res 61:1354–1357 Lunn RM, Langlois RG, Hsieh LL et al, 1999, XRCC1 polymorphisms: effects on aflatoxin B1-DNA adducts and glycophorin A variant frequency. Cancer Res 59:2557–2561 Abdel-Rahman SZ, El Zein RA, 2000, The 399Gln polymorphism in the DNA repair gene XRCC1 modulates the genotoxic response induced in human lymphocytes by the tobacco-specific nitrosamine NNK. Cancer Lett 159:63–71 Duell EJ, Wiencke JK, Cheng TJ et al, 2000, Polymorphisms in the DNA repair genes XRCC1 and ERCC2 and biomarkers of DNA damage in human blood mononuclear cells. Carcinogenesis 21:965–971 Hu JJ, Smith TR, Miller MS et al, 2001, Amino acid substitution variants of APE1 and XRCC1 genes associated with ionizing radiation sensitivity. Carcinogenesis 22:917–922 Duell EJ, Millikan RC, Pittman GS et al, 2001, Polymorphisms in the DNA repair gene XRCC1 and breast cancer. Cancer Epidemiol Biomarkers Prev 10:217–222 Kim SU, Park SK, Yoo KY et al, 2002, XRCC1 genetic polymorphism and breast cancer risk. Pharmacogenetics 12:335–338 Shu XO, Cai Q, Gao YT et al, 2003, A population-based casecontrol study of the Arg399Gln polymorphism in DNA repair gene XRCC1 and risk of breast cancer. Cancer Epidemiol Biomarkers Prev 12:1462–1467 Smith TR, Miller MS, Lohman K et al, 2003, Polymorphisms of XRCC1 and XRCC3 genes and susceptibility to breast cancer. Cancer Lett 190:183–190 Justenhoven C, Hamann U, Pesch B et al, 2004, ERCC2 genotypes and a corresponding haplotype are linked with breast cancer risk in a German population. Cancer Epidemiol Biomarkers Prev 13:2059–2064 Han J, Hankinson SE, De V et al, 2003, A prospective study of XRCC1 haplotypes and their interaction with plasma carotenoids on breast cancer risk. Cancer Res 63:8536–8541 Moullan N, Cox DG, Angele S et al, 2003, Polymorphisms in the DNA repair gene XRCC1, breast cancer risk, and response to radiotherapy. Cancer Epidemiol Biomarkers Prev 12:1168–1174 Forsti A, Angelini S, Festa F et al, 2004, Single nucleotide polymorphisms in breast cancer. Oncol Rep 11:917–922 Deligezer U, Dalay N, 2004, Association of the XRCC1 gene polymorphisms with cancer risk in Turkish breast cancer patients. Exp Mol Med 36:572–575 Metsola K, Kataja V, Sillanpaa P et al, 2005, XRCC1 and XPD genetic polymorphisms, smoking and breast cancer risk in a Finnish case-control study. Breast Cancer Res 7:R987–R997 Shen J, Gammon MD, Terry MB et al, 2005, Polymorphisms in XRCC1 modify the association between polycyclic aromatic hydrocarbon-DNA adducts, cigarette smoking, dietary antioxidants, and breast cancer risk. Cancer Epidemiol Biomarkers Prev 14:336–342 Bu D, Tomlinson G, Lewis CM et al, 2006, An intronic polymorphism associated with increased XRCC1 expression, reduced apoptosis and familial breast cancer. Breast Cancer Res Treat 99:257–265 Zhang Y, Newcomb PA, Egan KM et al, 2006, Genetic polymorphisms in base-excision repair pathway genes and risk of breast cancer. Cancer Epidemiol Biomarkers Prev 15:353–358 Chacko P, Rajan B, Joseph T et al, 2005, Polymorphisms in DNA repair gene XRCC1 and increased genetic susceptibility to breast cancer. Breast Cancer Res Treat 89:15–21 Patel AV, Calle EE, Pavluck AL et al, 2005, A prospective study of XRCC1, X-ray cross-complementing group 1, polymorphisms and breast cancer risk. Breast Cancer Res 7:R1168–R1173 Shi Q, Wang LE, Bondy ML et al, 2004, Reduced DNA repair of benzo[a]pyrene diol epoxide-induced adducts and common XPD polymorphisms in breast cancer patients. Carcinogenesis 25:1695–1700 Kuschel B, Chenevix-Trench G, Spurdle AB et al, 2005, Common polymorphisms in ERCC2, Xeroderma pigmentosum D, are not associated with breast cancer risk. Cancer Epidemiol Biomarkers Prev 14:1828–1831 Mechanic LE, Millikan RC, Player J et al, 2006, Polymorphisms in nucleotide excision repair genes, smoking and breast cancer in African Americans and whites: a population-based case-control study. Carcinogenesis 27:1377–1385 Terry MB, Gammon MD, Zhang FF et al, 2004, Polymorphism in the DNA repair gene XPD, polycyclic aromatic hydrocarbon- DNA adducts, cigarette smoking, and breast cancer risk. Cancer Epidemiol Biomarkers Prev 13:2053–2058 Kuschel B, Auranen A, McBride S et al, 2002, Variants in DNA double-strand break repair genes and breast cancer susceptibility. Hum Mol Genet 11:1399–1407 Syamala VS, Sreeja L, Syamala V et al., 2008, Influence of germline polymorphisms of GSTT1, GSTM1, and GSTP1 in familial versus sporadic breast cancer susceptibility and survival. Fam Cancer 7:213–220 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 389 EP 397 DI 10.1007/s12253-008-9135-8 PG 9 ER PT J AU Koornstra, JJ de Jong, S Boersma-van Eck, W Zwart, Ny Hollema, H de Vries, GEE Kleibeuker, HJ AF Koornstra, J Jan de Jong, Steven Boersma-van Eck, Wietske Zwart, Nynke Hollema, Harry de Vries, G E Elisabeth Kleibeuker, H Jan TI Fas Ligand Expression in Lynch Syndrome-Associated Colorectal Tumours SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Apoptosis; Colorectal cancer; Counterattack; Fas ligand; HNPCC; Lynch syndrome ID Apoptosis; Colorectal cancer; Counterattack; Fas ligand; HNPCC; Lynch syndrome AB Fas Ligand (FasL) expression by cancer cells may contribute to tumour immune escape via the Fas counterattack against tumour-infiltrating lymphocytes (TILs). Whether this plays a role in colorectal carcinogenesis in Lynch syndrome was examined studying FasL expression, tumour cell apoptosis and number of TILs in colorectal neoplasms from Lynch syndrome patients (50 adenomas, 20 carcinomas) compared with sporadic cases (69 adenomas, 52 carcinomas). FasL expression was observed in 94% of Lynch syndrome adenomas and in all carcinomas. FasL expression patterns and apoptotic indices were similar in Lynch syndrome-associated neoplasms and sporadic cases. The number of TILs was higher in Lynch syndrome neoplasms than in sporadic cases. There were no correlations between FasL expression and tumour cell apoptosis or number of TILs in Lynch syndrome-associated neoplasms. So, FasL expression is an early event in Lynch syndrome and sporadic colorectal carcinogenesis, but not related to TIL number. Taken together, our data do not support a role for the Fas counterattack in colorectal carcinogenesis in Lynch syndrome. C1 [Koornstra, J Jan] University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, 9700 Groningen, RB, The Netherlands. [de Jong, Steven] University of Groningen, University Medical Center Groningen, Department of Medical OncologyGroningen, RB, The Netherlands. [Boersma-van Eck, Wietske] University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, 9700 Groningen, RB, The Netherlands. [Zwart, Nynke] University of Groningen, University Medical Center Groningen, Department of Medical OncologyGroningen, RB, The Netherlands. [Hollema, Harry] University of Groningen, University Medical Center Groningen, Department of PathologyGroningen, RB, The Netherlands. [de Vries, G E Elisabeth] University of Groningen, University Medical Center Groningen, Department of Medical OncologyGroningen, RB, The Netherlands. [Kleibeuker, H Jan] University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, 9700 Groningen, RB, The Netherlands. RP Koornstra, JJ (reprint author), University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, 9700 Groningen, The Netherlands. EM j.j.koornstra@int.umcg.nl CR Hendriks YM, de Jong AE, Morreau H et al, 2006, Diagnostic approach and management of Lynch syndrome, hereditary nonpolyposis colorectal carcinoma): a guide for clinicians. CA Cancer J Clin 56:213–225 Boland CR, Thibodeau SN, Hamilton SR et al, 1998, A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58:5248–5257 De Vos tot Nederveen Cappel WH, Nagengast FM, Griffioen G et al, 2002, Surveillance for hereditary nonpolyposis colorectal cancer: a long-term study on 114 families. Dis Colon Rectum 45:1588–1894 Vasen HF, Nagengast FM, Khan PM, 1995, Interval cancers in hereditary nonpolyposis colorectal cancer, Lynch syndrome). Lancet 345:1183–1184 Ponz de Leon M, Della CG, Benatti P et al, 1998, Frequency and type of colorectal tumors in asymptomatic high-risk individuals in families with hereditary nonpolyposis colorectal cancer. Cancer Epidemiol Biomarkers Prev 7:639–641 Rijcken FE, Hollema H, Kleibeuker JH, 2002, Proximal adenomas in hereditary non-polyposis colorectal cancer are prone to rapid malignant transformation. Gut 50:382–386 Koornstra JJ, de Jong S, Hollema H et al, 2003, Changes in apoptosis during the development of colorectal cancer: a systematic review of the literature. Crit Rev Oncol Hematol 45:37–53 Woerner SM, Kloor M, von Knebel Doeberitz M et al, 2006, Microsatellite instability in the development of DNA mismatch repair deficient tumors. Cancer Biomark 2:69–86 Moller P, Koretz K, Leithauser F et al, 1994, Expression of APO- 1, CD95), a member of the NGF/TNF receptor superfamily, in normal and neoplastic colon epithelium. Int J Cancer 57:371–377 Belluco C, Esposito G, Bertorelle R et al, 2002, Fas ligand is upregulated during the colorectal adenoma-carcinoma sequence. Eur J Surg Oncol 28:120–125 Bennett MW, O’Connell J, Houston A et al, 2001, Fas ligand upregulation is an early event in colonic carcinogenesis. J Clin Pathol 54:598–604 Shimoyama M, Kanda T, Liu L et al, 2001, Expression of Fas ligand is an early event in colorectal carcinogenesis. J Surg Oncol 76:63–68 Younes M, Schwartz MR, Finnie D et al, 1999, Overexpression of Fas ligand, FasL, during malignant transformation in the large bowel and in Barrett’s metaplasia of the esophagus. Hum Pathol 30:1309–1313 O’Connell J, Bennett MW, O’Sullivan GC et al, 1998, Fas ligand expression in primary colon adenocarcinomas: evidence that the Fas counterattack is a prevalent mechanism of immune evasion in human colon cancer. J Pathol 186:240–246 Michael-Robinson JM, Pandeya N, Cummings MC et al, 2003, Fas ligand and tumour counter-attack in colorectal cancer stratified according to microsatellite instability status. J Pathol 201:46–54 van Geelen CM, Westra JL, de Vries EG et al, 2006, Prognostic significance of tumor necrosis factor-related apoptosis-inducing ligand and its receptors in adjuvantly treated stage III colon cancer patients. J Clin Oncol 24:4998–5004 Koornstra JJ, Rijcken FE, de Jong S et al, 2004, Assessment of apoptosis by M30 immunoreactivity and the correlation with morphological criteria in normal colorectal mucosa, adenomas and carcinomas. Histopathology 44:9–17 Jass JR, Sobin LH, Watanabe H, 1990, The World Health Organization’s histologic classification of gastrointestinal tumors. A commentary on the second edition, Cancer 66:2162–2167 Caulin C, Salvesen GS, Oshima RG, 1997, Caspase cleavage of keratin 18 and reorganization of intermediate filaments during epithelial cell apoptosis. J Cell Biol 138:1379–1394 Leers MP, Kolgen W, Bjorklund V et al, 1999, Immunocytochemical detection and mapping of a cytokeratin 18 neo-epitope exposed during early apoptosis. J Pathol 187:567–572 Dolcetti R, Viel A, Doglioni C et al, 1999, High prevalence of activated intraepithelial cytotoxic T lymphocytes and increased neoplastic cell apoptosis in colorectal carcinomas with microsatellite instability. Am J Pathol 154:1805–1813 Smyrk TC, Watson P, Kaul K et al, 2001, Tumor-infiltrating lymphocytes are a marker for microsatellite instability in colorectal carcinoma. Cancer 91:2417–2422 Deng G, Bell I, Crawley S et al, 2004, BRAF mutation is frequently present in sporadic colorectal cancer with methylated hMLH1, but not in hereditary nonpolyposis colorectal cancer. Clin Cancer Res 10:191–195 Johnson V, Volikos E, Halford SE et al, 2005, Exon 3 beta-catenin mutations are specifically associated with colorectal carcinomas in hereditary non-polyposis colorectal cancer syndrome. Gut 54:264–267 Losi L, Fante R, Di Gregorio C et al, 1995, Biologic characterization of hereditary non-polyposis colorectal cancer. Nuclear ploidy, AgNOR count, microvessel distribution, oncogene expression, and grade-related parameters. Am J Clin Pathol 103:265–270 Yagi OK, Akiyama Y, Nomizu T et al, 1998, Proapoptotic gene BAX is frequently mutated in hereditary nonpolyposis colorectal cancers but not in adenomas. Gastroenterology 114:268–274 Jass JR, 2004, HNPCC and sporadic MSI-H colorectal cancer: a review of the morphological similarities and differences. Fam Cancer 3:93–100 Young J, Simms LA, Biden KG et al, 2001, Features of colorectal cancers with high-level microsatellite instability occurring in familial and sporadic settings: parallel pathways of tumorigenesis. Am J Pathol 159:2107–2116 Heijink DM, Kleibeuker JH, Jalving M et al, 2007, Independent induction of caspase-8 and cFLIP expression during colorectal carcinogenesis in sporadic and HNPCC adenomas and carcinomas. Cell Oncol 29:409–419 Rijcken FE, Koornstra JJ, van der Sluis T et al, 2007, Early carcinogenic events in HNPCC adenomas: differences with sporadic adenomas. Dig Dis Sci [Epub ahead of print] Sinicrope FA, Roddey G, Lemoine M et al, 1998, Loss of p21WAF1/Cip1 protein expression accompanies progression of sporadic colorectal neoplasms but not hereditary nonpolyposis colorectal cancers. Clin Cancer Res 4:1251–1261 Sinicrope FA, Lemoine M, Xi L et al, 1999, Reduced expression of cyclooxygenase 2 proteins in hereditary nonpolyposis colorectal cancers relative to sporadic cancers. Gastroenterology 117:350–358 Houston AM, Michael-Robinson JM, Walsh MD et al, 2008, The “Fas counterattack” is not an active mode of tumor immune evasion in colorectal cancer with high-level microsatellite instability. Hum Pathol 39:243–250 Michael-Robinson JM, Biemer-Huttmann A, Purdie DM et al, 2001, Tumour infiltrating lymphocytes and apoptosis are independent features in colorectal cancer stratified according to microsatellite instability status. Gut 48:360–366 Jackson PA, Green MA, Marks CG et al, 1996, Lymphocyte subset infiltration patterns and HLA antigen status in colorectal carcinomas and adenomas. Gut 38:85–89 Rubio CA, Jacobsson B, Castanos-Velez E, 1999, Cytotoxic intraepithelial lymphocytes in colorectal polyps and carcinomas. Anticancer Res 19(4B):3221–3227 Warabi M, Kitagawa M, Hirokawa K, 2000, Loss of MHC class II expression is associated with a decrease of tumor-infiltrating T cells and an increase of metastatic potential of colorectal cancer: immunohistological and histopathological analyses as compared with normal colonic mucosa and adenomas. Pathol Res Pract 196:807–815 Guidoboni M, Gafa R, Viel A et al, 2001, Microsatellite instability and high content of activated cytotoxic lymphocytes identify colon cancer patients with a favorable prognosis. Am J Pathol 159:297–304 Naito Y, Saito K, Shiiba K et al, 1998, CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res 58:3491–3494 Ropponen KM, Eskelinen MJ, Lipponen PK et al, 1997, Prognostic value of tumour-infiltrating lymphocytes, TILs, in colorectal cancer. J Pathol 182:318–324 Dhein J, Walczak H, Baumler C et al, 1995, Autocrine T-cell suicide mediated by APO-1/(Fas/CD95). Nature 373:438–441 Houston A, Waldron-Lynch FD, Bennett MW et al, 2003, Fas ligand expressed in colon cancer is not associated with increased apoptosis of tumor cells in vivo. Int J Cancer 107:209–214 Houston A, Bennett MW, O’Sullivan GC et al, 2003, Fas ligand mediates immune privilege and not inflammation in human colon cancer, irrespective of TGF-beta expression. Br J Cancer 89:1345–1351 Okada K, Komuta K, Hashimoto S et al, 2000, Frequency of apoptosis of tumor-infiltrating lymphocytes induced by fas counterattack in human colorectal carcinoma and its correlation with prognosis. Clin Cancer Res 6:3560–3564 Walker JA, Quirke P, 2001, Viewing apoptosis through a TUNEL. J Pathol 195:275–276 O’Connell J, Houston A, Bennett MW, O’Sullivan GC, Shanahan F, 2001, Immune privilege or inflammation? Insights into the Fas ligand enigma. Nat Med 7:271–274 Restifo NP, 2000, Not so Fas: Re-evaluating the mechanisms of immune privilege and tumor escape. Nat Med 6:493–495 Strater J, Moller P, 2003, CD95, Fas/APO-1)/CD95L in the gastrointestinal tract: fictions and facts. Virchows Arch 442:218–225 Huber V, Fais S, Iero M et al, 2005, Human colorectal cancer cells induce T-cell death through release of proapoptotic microvesicles: role in immune escape. Gastroenterology 128:1796–1804 O’Connell J, O’Sullivan GC, Collins JK et al, 1996, The Fas counterattack: Fas-mediated T cell killing by colon cancer cells expressing Fas ligand. J Exp Med 184:1075–1082 Ryan AE, Shanahan F, O’Connell J et al, 2005, Addressing the “Fas counterattack” controversy: blocking fas ligand expression suppresses tumor immune evasion of colon cancer in vivo. Cancer Res 65:9817–9823 Favre-Felix N, Fromentin A, Hammann A et al, 2000, Cutting edge: the tumor counterattack hypothesis revisited: colon cancer cells do not induce T cell apoptosis via the Fas, CD95, APO-1, pathway. J Immunol 5023–5027 Arai H, Gordon D, Nabel EG et al, 1997, Gene transfer of Fas ligand induces tumor regression in vivo. Proc Natl Acad Sci USA 94:13862–13867 Jekle A, Obst R, Lang F et al, 2000, CD95/CD95 ligand-mediated counterattack does not block T cell cytotoxicity. Biochem Biophys Res Commun 272:395–399 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 399 EP 406 DI 10.1007/s12253-008-9136-7 PG 8 ER PT J AU Berstein, ML Pozharisski, MK Imyanitov, NE Maximova, AN Kovalevskij, YA AF Berstein, M Lev Pozharisski, M Kazimir Imyanitov, N Evgeny Maximova, A Natalya Kovalevskij, Yu Anatoly TI Aromatase, CYP1B1 and Fatty Acid Synthase Expression in Breast Tumors of BRCA1 Mutation Carriers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; BRCA1; Hormonal-metabolic factors; Aromatase; Catechol estrogens; Lipogenesis ID Breast cancer; BRCA1; Hormonal-metabolic factors; Aromatase; Catechol estrogens; Lipogenesis AB Numerous experimental evidence suggest that BRCA1-associated breast carcinomas may have distinct endocrine and metabolic features, however these peculiarities are poorly evaluated in clinical settings. Here we comparatively analyzed for the first time aromatase, estrogen 4-hydroxylase (CYP1B1) and fatty acid synthase immunohistochemical expression in breast tumors obtained from 12 BRCA1 mutations carriers and 22 non-carriers. Aromatase expression was higher in mutation carriers than in sporadic cases (p=0.04), which confirms the earlier results obtained in cell lines with down-regulated wild-type BRCA1 and corroborates the usage of aromatase inhibitors in such patients. No differences between study groups were found in the expression of CYP1B1 and fatty acid synthase, which does not, however, mitigate the need of further search for manifestations of the excessive genotoxic effects of estrogens and for increased lipogenesis in BRCA1 mutations carriers. C1 [Berstein, M Lev] NN Petrov Institute of Oncology, Pesochny, 197758 Saint-Petersburg, Russian Federation. [Pozharisski, M Kazimir] Scientific Center of Radiology and Surgical TechnologiesSaint-Petersburg, Russian Federation. [Imyanitov, N Evgeny] NN Petrov Institute of Oncology, Pesochny, 197758 Saint-Petersburg, Russian Federation. [Maximova, A Natalya] Scientific Center of Radiology and Surgical TechnologiesSaint-Petersburg, Russian Federation. [Kovalevskij, Yu Anatoly] NN Petrov Institute of Oncology, Pesochny, 197758 Saint-Petersburg, Russian Federation. RP Berstein, ML (reprint author), NN Petrov Institute of Oncology, 197758 Saint-Petersburg, Russian Federation. EM levmb@endocrin.spb.ru CR Welcsh PL, Owens KN, King MC, 2000, Insights into the functions of BRCA1 and BRCA2. Trends Genet 16:69–74 Eeles R, Kadouri L, 1999, BRCA1/2 carriers and endocrine risk modifiers. Endocr Relat Cancer 6:521–528 Hilakivi-Clarke L, 2000, Estrogens, BRCA1, and breast cancer. Cancer Res 60:4993–5001 Rosen EM, Fan S, Isaacs C, 2005, BRCA1 in hormonal carcinogenesis: basic and clinical research. Endocr Relat Cancer 12(3):533–548 Hu Y, Ghosh S, Amleh A, Yue W, Lu Y, Katz A et al, 2005, Modulation of aromatase expression by BRCA1: a possible link to tissue-specific tumor suppression. Oncogene 24(56):8343–8348 Lu M, Chen D, Lin Z, Reierstad S, Trauernicht AM, Boyer TG et al, 2006, BRCA1 negatively regulates the cancer-associated aromatase promoters I.3 and II in breast adipose fibroblasts and malignant epithelial cells. J Clin Endocrinol Metab 91(11):4514–4519 Foulkes WD, Metcalfe K, Sun P, Hanna WM, Lynch HT, Ghadirian P et al, 2004, Estrogen receptor status in BRCA1- and BRCA2-related breast cancer: the influence of age, grade, and histological grade. Clin Cancer Res 10(6):2029–2034 Hosey AM, Gorski JJ, Murray MM, Quinn JE, Chung WY, Stewart GE et al, 2007, Molecular basis for estrogen receptor alpha deficiency in BRCA1-linked breast cancer. J Natl Cancer Inst 99(22):1683–1694 Bar Sade-Bruchim RB, Khalil T, Quenneville L, Foulkes W, Chong G, Aloyz RS, 2007, Determining the effect of estrogen metabolites on BRCA1-allelic imbalance in normal breast cells heterozygous for BRCA1 mutations. Proceedings of American Assoc. Cancer Res Annual Meeting, Los Angeles, CA, USA, Abstr 3475) Cao L, Xu X, Cao LL, Wang RH, Coumoul X, Kim SS et al, 2007, Absence of full-length Brca1 sensitizes mice to oxidative stress and carcinogenesis. Carcinogenesis 28(7):1401–1407 Berstein LM, 2008, Endocrinology of wild and mutant BRCA1 gene and types of hormonal carcinogenesis. Future Oncol 4, 1):23–39 Shukla V, Coumoul X, Cao L, Wang RH, Xiao C, Xu X et al, 2006, Absence of the full-length breast cancer-associated gene-1 leads to increased expression of insulin-like growth factor signaling axis members. Cancer Res 66(14):7151–7157 Moreau K, Dizin E, Ray H, Luquain C, Lefai E, Foufelle F et al, 2006, BRCA1 affects lipid synthesis through its interaction with acetyl-CoA carboxylase. J Biol Chem 281(6):3172–3181 Sokolenko AP, Mitiushkina NV, Buslov KG, Bit-Sava EM, Iyevleva AG, Chekmariova EV et al, 2006, High frequency of BRCA1 5382insC mutation in Russian breast cancer patients. Eur J Cancer 42(10):1380–1384 Adlercreutz H, Gorbach SL, Goldin BR, Woods MN, Dwyer JT, Hamalainen E, 1994, Estrogen metabolism and excretion in Oriental and Caucasian women. J Natl Cancer Inst 86(14):1076–1082 Cavalieri E, Chakravarti D, Guttenplan J, Hart E, Ingle J, Jankowiak R et al, 2006, Catechol estrogen quinones as initiators of breast and other human cancers: implications for biomarkers of susceptibility and cancer prevention. Biochim Biophys Acta 1766(1):63–78 Menendez JA, Lupu R, 2007, Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat Rev Cancer 7, 10):763–777 Kuhajda FP, 2006, Fatty acid synthase and cancer: new application of an old pathway. Cancer Res 66(12):5977–5980 Sasano H, Anderson TJ, Silverberg SG, Santen RJ, Conway M, Edwards DP et al, 2005, The validation of new aromatase monoclonal antibodies for immunohistochemistry—a correlation with biochemical activities in 46 cases of breast cancer. J Steroid Biochem Mol Biol 95(1–5):35–39 Marchetti P, Di Rocco CZ, Ricevuto E, Bisegna R, Cianci G, Calista F et al, 2004, Reducing breast cancer incidence in familial breast cancer: overlooking the present panorama. Ann Oncol 15(Suppl 1):I27–I34 Noruzinia M, Coupier I, Pujol P, 2005, Is BRCA1/BRCA2- related breast carcinogenesis estrogen dependent? Cancer 104, 8):1567–1574 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 407 EP 409 DI 10.1007/s12253-008-9137-6 PG 3 ER PT J AU Wu, Y Liu, XM Wang, XJ Zhang, Y Liang, XQ Cao, EH AF Wu, Yan Liu, Xiao-Min Wang, Xiao-Juan Zhang, Yang Liang, Xiao-Qiu Cao, En-Hua TI PIG11 is Involved in Hepatocellular Carcinogenesis and Its Over-expression Promotes Hepg2 Cell Apoptosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PIG11; HCC; Apoptosis; Tumour suppressor gene ID PIG11; HCC; Apoptosis; Tumour suppressor gene AB PIG11 (p53-induced gene 11) is a p53 target gene and candidate tumour suppressor gene. In this study, the expression of PIG11 protein was detected in human hepatocellular carcinoma (HCC) and normal liver tissues with an immunohistochemical method. Compared with expression in human normal liver tissues, the expression of PIG11 protein was significantly down-regulated in human HCC tissues. In addition, a recombinant pLXSNPIG11 retroviral vector was constructed and transfected into HepG2 cells (human hepatocellular carcinoma cell line) and the role of PIG11 in apoptosis was analyzed. The percentage (18.60%) of apoptotic cells transfected with pLXSN-PIG11 was higher than that in cells transfected with pLXSN only (6.03%) or the vehicle control (3.81%) (P<0.01). DNA gel electrophoresis showed a clear DNA ladder in pLXSN-PIG11-infected HepG2 cells. Our results suggested that the PIG11 gene is involved in carcinogenesis and development of hepatocarcinoma. Therefore, PIG11 is considered to be a new candidate liver tumour suppressor gene, and may play an important role in tumour suppression through promotion of cell apoptosis. C1 [Wu, Yan] University of South China, Department of Pathology, 421001 Hengyang, Hunan, China. [Liu, Xiao-Min] University of South China, Department of Pathology, 421001 Hengyang, Hunan, China. [Wang, Xiao-Juan] University of South China, Department of Pathology, 421001 Hengyang, Hunan, China. [Zhang, Yang] University of South China, Department of Pathology, 421001 Hengyang, Hunan, China. [Liang, Xiao-Qiu] University of South China, Department of Pathology, 421001 Hengyang, Hunan, China. [Cao, En-Hua] Chinese Academy of Science, Institute of Biophysics, 15 Datun Road, Chaoyang District, 100101 Beijing, China. RP Liang, XQ (reprint author), University of South China, Department of Pathology, 421001 Hengyang, China. EM liangxiaoqiu505@hotmail.com CR Polyak K, Xia Y, Zweier JL et al, 1997, A model for p53-induced apoptosis. Nature 389:300–305 Zhu J, Jiang J, Zhou W et al, 1999, Differential regulation of cellular target genes by p53 devoid of the PXXP motifs with impaired apoptotic activity. Oncogene 18:2149–2155 Woo SH, Park IC, Park MJ et al, 2002, Arsenic trioxide induces apoptosis through a reactive oxygen spcies-dependent pathway and loss of mitochondrial menbrane potential in Hele cells. Int J Oncol 21:57–63 Ramachandran C, Rodriguez S, Ramachandran R et al, 2005, Expression profiles of apoptotic genes induced by curcumin in human breast cancer and mammary epithelial cell lines. Anticancer Res 25(5):3293–3302 Chiba T, Yokosuka O, Fukai K et al, 2004, Cell growth inhibition and gene expression induced by the histone deacetylase inhibitor, trichostatin A, on human hepatoma cells. Oncology 66(6):481– 491 Liang XQ, Cao EH, Zhang Y et al, 2003, p53-induced gene 11, PIG11, involved in arsenic trioxide induced apoptosis in human gastric cancer MGC-803 cells. Oncology Reports 10(5):1265– 1269 Xiong X, Li H, Cao EH, 2007, PIG11 protein binds to DNA in sequence-independent manner in vitro. BBRC 358:29–34 Liang XQ, Cao EH, Zhang Y et al, 2004, A p53 target gene, PIG11, contributes to chemosensitivity of cells to arsenic trioxide. FEBS Letters 569(1–3):94–98 Ricketts SL, Carter JC, Coleman WB, 2003, Identification of three 11p11.2 candidate liver tumor suppressors through analysis of known human genes. Mol Carcinog 36(2):90–99 Xu L, Hui AY, Albanis E et al, 2005, Human hepatic stellate cell lines, LX-1 and LX-2: new tools for analysis of hepatic fibrosis. Gut 54:142–151 Chiba T, Yokosuka O, Fukai K et al, 2004, Cell growth inhibition and gene expression induced by the histone deacetylase inhibitor, trichostatin A, on human hepatoma cells. Oncology 66:481–491 Song H, Xu Y, 2007, Gain of function of p53 cancer mutants in disrupting critical DNA damage response pathways. Cell Cycle 6, 13):1570–1573 Leder A, McMenamin J, Zhou F et al, 2008, Genome-wide SNP analysis of Tg.AC transgenic mice reveals an oncogenic collaboration between v-Ha-ras and Ink4a, which is absent in p53 deficiency. Oncogene 27(17):2456–2465 Feng W, Xiao J, Zhang Z et al, 2007, Senescence and apoptosis in carcinogenesis of cervical squamous carcinoma. Mod Pathol 20, 9):961–966 Liu L, Qing S, Chen H et al, 2000, An experimental study on arsenic trioxide-selectively induced human hepatocarcinoma cell lines apoptosis and its related genes. Zhonghua Gan Zang Bing Za Zhi 8(6):367–371, Chinese) Rivera A, Maxwell SA, 2005, The p53-induced gene-6, proline oxidase, mediates apoptosis through a calcineurin-dependent pathway. J Biol Chem 280:29346–29354 Ding WX, Shen HM, Ong CN et al, 2000, Critical role of reactive oxygen species and mitochondrial permeability transition in microcystin-induced rapid apoptosis in rat hepatocytes. Hepatology 32(3):547–555 Touyz RM, 2005, Reactive oxygen species as mediators of calcium signaling by angiotensin II:implications in vascular physiology and pathophysiology. Antioxid Redox Signal 7(9–10):1302– 1314 KimHJ, Chakravarti N,Oridate N et al, 2006, N-(4-Hydroxyphenyl, retinamide-induced apoptosis triggered by reactive oxygen species is mediated by activation of MAPKs in head and neck squamous carcinoma cells. Oncogene 25(19):2785–2794 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 411 EP 416 DI 10.1007/s12253-008-9138-5 PG 6 ER PT J AU Demir, R Naschberger, L Demir, I Melling, N Dimmler, A Papadopoulus, Th Sturzl, M Klein, P Hohenberger, W AF Demir, Resit Naschberger, Lisa Demir, Ilknur Melling, Nathaniel Dimmler, Arno Papadopoulus, Thomas Sturzl, Michael Klein, Peter Hohenberger, Werner TI Hypoxia Generates a More Invasive Phenotype of Tumour Cells: An In Vivo Experimental Setup Based on the Chorioallantoic Membrane SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chorioallantoic membrane; Hypoxia; Tumour invasion; Tumor progression ID Chorioallantoic membrane; Hypoxia; Tumour invasion; Tumor progression AB Of all processes involved in carcinogenesis, local invasion and the formation of metastases are the clinically most relevant but the scientifically least well understood at their molecular level. Recent experimental progress has identified that tumour hypoxia not only induces tumour angiogenesis, but also modulates the expression of several genes that have been implicated in tumour invasion and metastasis. Here we developed an in vivo model to understand a number of molecular pathways and cellular mechanisms for tumour invasion in hypoxia. For this purpose fertilized chicken eggs were incubated for 10 days in normoxic conditions. Subsequently colon carcinoma cells (SW-480) were placed on the chorioallantoic membrane. During the following 6 days the eggs were incubated either in normoxic conditions or in stepwise decreasing hypoxic conditions. SW-480 colon carcinoma cells did not invade the epithelial layer in normoxic conditions. In contrast an invasion through the epithelial layer in to the mesoderm was already seen after 3 days when incubated in hypoxic conditions. The chorioallantoic membrane assay described in this paper allows investigating tumour invasion and its cellular mechanisms under defined hypoxic conditions. C1 [Demir, Resit] University of Erlangen, Department of Surgery, Krankenhausstrasse 12, D-91054 Erlangen, Germany. [Naschberger, Lisa] University of Erlangen, Department of Experimental SurgeryErlangen, Germany. [Demir, Ilknur] University of Erlangen, Department of BiologyErlangen, Germany. [Melling, Nathaniel] Albertinen-Krankenhaus, Department of SurgeryHamburg, Germany. [Dimmler, Arno] St. Vincentius-Kliniken, Department of PathologyKarlsruhe, Germany. [Papadopoulus, Thomas] Vivantes Humboldt Klinikum, Department of PathologyBerlin, Germany. [Sturzl, Michael] University of Erlangen, Department of Experimental SurgeryErlangen, Germany. [Klein, Peter] University of Erlangen, Department of Surgery, Krankenhausstrasse 12, D-91054 Erlangen, Germany. [Hohenberger, Werner] University of Erlangen, Department of Surgery, Krankenhausstrasse 12, D-91054 Erlangen, Germany. RP Demir, R (reprint author), University of Erlangen, Department of Surgery, D-91054 Erlangen, Germany. EM resit.demir@uk-erlangen.de CR Hart IR, Saini A, 1992, Biology of tumour metastasis. Lancet 339:1453–1457 Woodhouse EC, Chuaqui RF, Liotta LA, 1997, General mechanisms of metastasis. Cancer 80:1529–1537 Christofori G, 2006, New signals from the invasive front. Nature 441:444–450 Kirchner T, Brabletz T, 2000, Patterning and nuclear beta-catenin expression in the colonic adenoma-carcinoma sequence. Analogies with embryonic gastrulation. Am J Pathol 157:1113–1121 Thiery JP, 2002, Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2:442–454 Brizel DM, Scully SP, Harrelson JM, Layfield LJ, Dodge RK, Charles HC, Samulski TV, Prosnitz LR, Dewhirst MW, 1996, Radiation therapy and hyperthermia improve the oxygenation of human soft tissue sarcomas. Cancer Res 56:5347–5350 Sullivan R, Graham CH, 2007, Hypoxia-driven selection of the metastatic phenotype. Cancer Metastasis Rev 26:319–331 Reizis A, Hammel I, Ar A, 2005, Regional and developmental variations of blood vessel morphometry in the chick embryo chorioallantoic membrane. J Exp Biol 208:2483–2488 Wilting J, Christ B, Bokeloh M,Weich HA, 1993, In vivo effects of vascular endothelial growth factor on the chicken chorioallantoic membrane. Cell Tissue Res 274:163–172 Ribatti D, Vacca A, Iurlaro M, Ria R, Roncali L, Dammacco F, 1996, Human recombinant interferon alpha-2a inhibits angiogenesis of chick area vasculosa in shell-less culture. Int J Microcirc Clin Exp 16:165–169 Demir R, Hoper J, 1997, Effect of beta-interferon on vascular density, mitochondrial metabolism and alkaline phosphatase in normoxia and hypoxia. Adv Exp Med Biol 428:439– 447 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 417 EP 422 DI 10.1007/s12253-008-9140-y PG 6 ER PT J AU Sughayer, AM Zakarneh, L Abu-Shakra, R AF Sughayer, A Maher Zakarneh, Lama Abu-Shakra, Raffat TI Collision Metastasis of Breast and Ovarian Adenocarcinoma in Axillary Lymph Nodes: A Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Collision metastasis ID Collision metastasis AB Despite their accepted clinical and genetic association, the incidence of synchronous breast and ovarian carcinoma is rare. Moreover, collision metastasis from both breast and ovarian carcinomas to the same lymph node, to our knowledge has never been reported. Review of the literature revealed eleven cases of metastatic malignant tumors colliding in the same lymph node, none of which had both ovarian and breast carcinoma. Our case was that of a 63 year old woman presenting with a breast lump that was diagnosed as infiltrating ductal carcinoma after a needle biopsy. One month later the patient was found to have malignant ascites, omental carcinomatosis and an ovarian mass. Histology and immunohistochemistry revealed high grade serous papillary adenocarcinoma. When surgery was done to treat the breast tumor some of the axillary lymph nodes revealed metastases from the breast primary, others metastases from the ovarian primary and one had both tumors in a collision phenomenon. Immunohistochemistry was used to confirm this finding. C1 [Sughayer, A Maher] King Hussein Cancer CenterAmman, Jordan. [Zakarneh, Lama] King Hussein Cancer CenterAmman, Jordan. [Abu-Shakra, Raffat] Ministry of National Guard-Health Affairs, King Abdulaziz Medical City, King Abdullah International Medical Research CentreJeddah, Saudi Arabia. RP Sughayer, AM (reprint author), King Hussein Cancer Center, Amman, Jordan. EM msughayer@khcc.jo CR Wade ZK, Shippey JE, Hamon GA et al, 2004, Collision metastasis of prostatic and colonic adenocarcinoma. Arch Pathol Lab Med 128(3):318–320 Mar Mourra N, Parc Y, McNamara D et al, 2005, Lymph node metastases of prostatic adenocarcinoma in the mesorectum in patients with adenocarcinoma or villous tumor of the rectum with collision phenomenon in a single lymph node: report of five cases. Dis Colon Rectum. 48(2):384–389 Feb Morgen AD, 1969, Carcinomas of rectum and prostatemetastasizing to the same lymph nodes. J Pathol 97:143–145 Ergen A, Balbay MD, Irwin M, Torno R, 1995, Collision metastasis of bladder and prostate carcinoma to a single pelvic lymph node. Int Urol Nephrol 27(6):743–745 Gohji K, Nomi M, Kizaki T et al, 1997, “Collision phenomenon” of prostate and bladder cancers in lymph node metastases. Int J Urol 4(2):222–224 Mar Overstreet K, Haghighi P, 2001, Urothelial and prostate carcinoma metastasizing to the same lymph node: a case report and review of the literature. Arch Pathol Lab Med 125(10):1354–1357 Oct Terada T, Satoh Y, Aoki N et al, 1993, The coexistence of cancer cells of different origin within the same lymph nodes. Surg Radiol Anat 15:119–123 Allal AS, Weintraub J, Remadi S, Abele R, 1996, Concurrent interfollicular Hodgkin’s disease and metastatic breast carcinoma in lymph nodes. Pathol Int 46(10):787–790 Oct Pastolero GC, Coire CI, Asa SL, 1996, Concurrent medullary and papillary carcinomas of thyroid with lymph node metastases. A collision phenomenon. Am J Surg Pathol 20(2):245–250 Feb Carbone A, Volpe R, 1983, Kaposi’s sarcoma in lymph nodes concurrent with Hodgkin’s disease. Am J Clin Pathol 80(2):228– 230 Aug Miki Y, Swensen J, Shattuck-Eidens D et al, 1994, A strong candidate for breast and ovarian cancer susceptibility gene BRCA-1. Science 266:66–71 Wooster R, Neuhausen SL, Mangion J et al, 1994, Localization of a breast cancer susceptibility gene, BRCA-2, to chromosome 13q 12–13. Science 265:2088–2090 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 423 EP 427 DI 10.1007/s12253-008-9141-x PG 5 ER PT J AU Ohara, T Kawashiri, Sh Tanaka, A Noguchi, N Kitahara, H Okamune, A Kato, K Hase, T Nakaya, H Yoshizawa, K AF Ohara, Teruhisa Kawashiri, Shuichi Tanaka, Akira Noguchi, Natsuyo Kitahara, Hiroko Okamune, Ayako Kato, Koroku Hase, Takashi Nakaya, Hiromitsu Yoshizawa, Kunio TI Integrin Expression Levels Correlate with Invasion, Metastasis and Prognosis of Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Integrin; Oral squamous cell carcinoma; Invasion; Metastasis; Prognosis ID Integrin; Oral squamous cell carcinoma; Invasion; Metastasis; Prognosis AB The present study evaluated the relationship between alpha 3, alpha 6A, and beta 1 integrin expression in cancer cells at the invasive front of oral squamous cell carcinoma (OSCC) and survival rates, as well as the clinical and pathological characteristics. Sections of 100 specimens of primary OSCC were immunostained to assess alpha 3, alpha 6A, and beta 1 integrin expression in cancer cells at the invasive front. OSCC patients with higher expression levels of alpha 3, alpha 6A, and beta 1 integrin had significantly better prognosis than those with lower expression levels (median survival at low vs. high expression levels: alpha 3, 37.1 months vs. 55.7 months; alpha 6A, 38.3 months vs. 47.9 months; and beta 1, 26.1 months vs. 46.1 months) (P<0.05). In addition, beta 1 integrin expression showed the highest correlation with clinical and pathological characteristics. This study concludes that alpha 3, alpha 6A, and beta 1 integrin expression in cancer cells at the invasive front are related to the mode of invasion and prognosis in OSCC. C1 [Ohara, Teruhisa] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Ishikawa, Japan. [Kawashiri, Shuichi] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Ishikawa, Japan. [Tanaka, Akira] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Ishikawa, Japan. [Noguchi, Natsuyo] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Ishikawa, Japan. [Kitahara, Hiroko] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Ishikawa, Japan. [Okamune, Ayako] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Ishikawa, Japan. [Kato, Koroku] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Ishikawa, Japan. [Hase, Takashi] Noto Hospital, Department of Oral Surgery, 926-8610 Nanao, Ishikawa, Japan. [Nakaya, Hiromitsu] Municipal Tsuruga Hospital, Department of Oral Surgery, 914-8502 Tsuruga, Fukui, Japan. [Yoshizawa, Kunio] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8640 Kanazawa, Ishikawa, Japan. RP Kawashiri, Sh (reprint author), Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 920-8640 Kanazawa, Japan. EM skawa@med.kanazawa-u.ac.jp CR Tamkun JW, DeSimone DW, Fonda D et al, 1986, Structure of integrin, a glycoprotein involved in the transmembrane linkage between fibronectin and actin. Cell 46:271–282 Avur Z, Geiger B, 1981, The removal of extracellular fibronectin from areas of cell-substrate contact. Cell 25:121–132 Heggeness MH, Ash JF, Singer SJ, 1978, Transmembrane linkage of fibronectin to intracellular actin-containing filaments in cultured human fibroblasts. Ann NY Acad Sci 312:414–417 Calderwood DA, 2004, Integrin activation. J Cell Sci 117:657–666 Maschler S, Wirl G, Spring H et al, 2005, Tumor cell invasiveness correlates with changes in integrin expression and localization. Oncogene 24:2032–2041 Jim H, Varner J, 2004, Integrins: roles in cancer development and as treatment targets. Br J of Cancer 90:561–565 Bottger T, Maschek H, Lobo M et al, 1999, Prognostic value of immunohistochemical expression of beta-1 integrin in pancreatic carcinoma. Oncology 56:308–313 Grzesiak J, Bouvet M, 2006, The α2β1 integrin mediates the malignant phenotype on type I collagen in pancreatic cancer cell lines. Br J Cancer 94:1311–1319 Park C, Zhang H, Pallavicini M et al, 2006, β1 integrin inhibitory antibody induces apoptosis of breast cancer cells, inhibits growth, and distinguishes malignant from normal phenotype in three dimensional culture and in vivo. Cancer Res 66(3):1256–1535 Miura S, Nishioka M, Nozawa K et al, 1997, A three-dimensional morphological study on the tumor-stromal interactions in colorectal adenocarcinoma. Jpn J Res Gastroenterol Carcinogenesis 9:343–348 Fujita S, Suzuki H, Hirohashi S, 1992, Inhibition of cell attachment, invasion and metastasis of human carcinoma cells by anti-integrin β1 subunit antibody. Jpn J Cancer Res 83:1317– 1326 Matsuura N, 1994, The role of integrins in cancer metastasis. Exp Med 12:925–930 Brockbank EC, Bridges J, Marshall CJ et al, 2005, Integrin β1 is required for the invasive behavior but not proliferation of squamous cell carcinoma cells in vivo. Br J Cancer 92:102– 112 Hogervorst F, Admiraal LG, Niessen C et al, 1993, Biochemical characterization and tissue distribution of the A and B variants of the integrin alpha 6 subunit. J Cell Bio 121:179–191 Waes C, Kozarsky K, Warren A et al, 1991, The A9 antigen associated with aggressive human squamous carcinoma is structurally and functionally similar to the newly defined integrin α6β4. Cancer Res 51:2395–2402 Kitahara H, Kawashiri S, Kato K et al, 2008, Immunohistochemical expressions of E-cadherin and β-catenin correlate with the invasion, metastasis and prognosis of oral squamous cell carcinoma. Oral Surg 1:28–34 Thomas GJ, Jones J, Speight PM, 1997, Integrins and Oral Cancer. Oral Oncol 33:381–388 Yamamoto E, Kohama G, Sunagawa H et al, 1983, Mode of invasion, bleomycin sensitivity, and clinical course in squamous cell carcinoma of the oral cavity. Cancer 51:2175–2180 Bottger T, Youssef V, Dutkowski P et al, 1999, α1 integrin expression on squamous cell carcinoma of the esophagus: A new prognostic parameter? Oncology 56:308–313 Fujita S, Watanabe M, Kubota T et al, 1995, Alteration of expression in integrin β1-subunit correlates with invasion and metastasis in colorectal cancer. Cancer Lett 91:145–149 Satake J, Suzuki M, Yokoyama J et al, 1995, Integrin distribution in head and neck cancer: association of the α6 subunit with cervical lymph node metastasis. Jpn J Head Neck Cancer 21:1–5 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 429 EP 436 DI 10.1007/s12253-008-9142-9 PG 8 ER PT J AU Laughlin, MK Luo, D Liu, Ch Shaw, G Warrington, HK Qiu, J Yachnis, TA Harrison, KJ AF Laughlin, M Katharine Luo, Defang Liu, Che Shaw, Gerry Warrington, H Kenneth Qiu, Jingxin Yachnis, T Anthony Harrison, K Jeffrey TI Hematopoietic- and Neurologic-Expressed Sequence 1 Expression in the Murine GL261 and High-Grade Human Gliomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE GL261; Tumor; siRNA; Jupiter; AAV; Glioma ID GL261; Tumor; siRNA; Jupiter; AAV; Glioma AB The hematopoietic- and neurologic-expressed sequence 1 (Hn1) gene encodes a highly conserved protein that is expressed in developing and regenerating tissues. In this study, Hn1 expression was evaluated in human and murine malignant gliomas. Hn1 mRNA and protein were detected in the murine GL261 glioma cell line and in GL261 brain tumors in vivo. HN1 is also expressed in human U118MG and U87MG cell lines. Evaluation of human brain tumors using an anti-Hn1 polyclonal antibody detected strong immunoreactivity in high-grade (WHO III and IV) malignant gliomas. The rate of GL261 cell proliferation in vitro was unaltered by Hn1 depletion using an anti-Hn1 siRNA. However, tumors established from Hn1-depleted GL261 cells formed significantly smaller volumes than those established from control-treated cells. These data suggest a role for Hn1 in the biology of malignant brain tumors. C1 [Laughlin, M Katharine] University of Florida, College of Medicine, Department of Pharmacology & Therapeutics, 1600 SW Archer Rd, 32610-0267 Gainesville, FL, USA. [Luo, Defang] University of Florida, College of Medicine, Department of Pharmacology & Therapeutics, 1600 SW Archer Rd, 32610-0267 Gainesville, FL, USA. [Liu, Che] University of Florida, College of Medicine, Department of Pharmacology & Therapeutics, 1600 SW Archer Rd, 32610-0267 Gainesville, FL, USA. [Shaw, Gerry] University of Florida, College of Medicine, Department of Neuroscience, 32610-0267 Gainesville, FL, USA. [Warrington, H Kenneth] University of Florida, College of Medicine, Department of Pediatrics, 32610-0267 Gainesville, FL, USA. [Qiu, Jingxin] University of Florida, College of Medicine, Department of Pathology, 32610-0267 Gainesville, FL, USA. [Yachnis, T Anthony] University of Florida, College of Medicine, Department of Pathology, 32610-0267 Gainesville, FL, USA. [Harrison, K Jeffrey] University of Florida, College of Medicine, Department of Pharmacology & Therapeutics, 1600 SW Archer Rd, 32610-0267 Gainesville, FL, USA. RP Harrison, KJ (reprint author), University of Florida, College of Medicine, Department of Pharmacology & Therapeutics, 32610-0267 Gainesville, USA. EM jharriso@ufl.edu CR Zujovic V, Luo D, Baker HV, Lopez MC, Miller KR, Streit WJ, Harrison JK, 2005, The facial motor nucleus transcriptional program in response to peripheral nerve injury identifies Hn1 as a regeneration-associated gene. J Neurosci Res 82:581–591 Goto T, Hisatomi O, Kotoura M, Tokunaga F, 2006, Induced expression of hematopoietic- and neurologic-expressed sequence 1 in retinal pigment epithelial cells during newt retina regeneration. Exp Eye Res 83:972–980 Lu KH, Patterson AP, Wang L, Marquez RT, Atkinson EN, Baggerly KA, Ramoth LR, Rosen DG, Liu J, Hellstrom I, Smith D, Hartmann L, Fishman D, Berchuck A, Schmandt R, Whitaker R, Gershenson DM, Mills GB, Bast RC, Jr., 2004, Selection of potential markers for epithelial ovarian cancer with gene expression arrays and recursive descent partition analysis. Clin Cancer Res 10:3291–3300 Tang W, Lai YH, Han XD, Wong PM, Peters LL, Chui DH, 1997, Murine Hn1 on chromosome 11 is expressed in hemopoietic and brain tissues. Mamm Genome 8:695–696 Zhou G, Wang J, Zhang Y, Zhong C, Ni J, Wang L, Guo J, Zhang K, Yu L, Zhao S, 2004, Cloning, expression and subcellular localization of HN1 and HN1L genes, as well as characterization of their orthologs, defining an evolutionarily conserved gene family. Gene 331:115–123 Zimmerman HM, Arnold H, 1941, Experimental brain tumors. I. Tumors produced with methylcholanthrene. Cancer Res 919–924 Ausman JI, Shapiro WR, Rall DP, 1970, Studies on the chemotherapy of experimental brain tumors: development of an experimental model. Cancer Res 30:2394–2400 Szatmari T, Lumniczky K, Desaknai S, Trajcevski S, Hidvegi EJ, Hamada H, Safrany G, 2006, Detailed characterization of the mouse glioma 261 tumor model for experimental glioblastoma therapy. Cancer Sci 97:546–553 San-Galli F, Vrignaud P, Robert J, Coindre JM, Cohadon F, 1989, Assessment of the experimental model of transplanted C6 glioblastoma in Wistar rats. J Neurooncol 7:299–304 Weiner NE, Pyles RB, Chalk CL, Balko MG, Miller MA, Dyer CA, Warnick RE, Parysek LM, 1999, A syngeneic mouse glioma model for study of glioblastoma therapy. J Neuropathol Exp Neurol 58:54–60 Ishii N, Tada M, Hamou MF, Janzer RC, Meagher-Villemure K, Wiestler OD, Tribolet N, Van Meir EG, 1999, Cells with TP53 mutations in low grade astrocytic tumors evolve clonally to malignancy and are an unfavorable prognostic factor. Oncogene 18:5870–5878 Bos JL, 1989, ras oncogenes in human cancer: a review. Cancer Res 49:4682–4689 Sidransky D, Mikkelsen T, Schwechheimer K, Rosenblum ML, Cavanee W, Vogelstein B, 1992, Clonal expansion of p53 mutant cells is associated with brain tumour progression. Nature 355:846–847 Church GM, Gilbert W, 1984, Genomic sequencing. Proc Natl Acad Sci USA 81:1991–1995 Harris J, Ayyub C, Shaw G, 1991, A molecular dissection of the carboxyterminal tails of the major neurofilament subunits NF-M and NF-H. J Neurosci Res 30:47–62 McCarty DM, Monahan PE, Samulski RJ, 2001, Self-complementary recombinant adeno-associated virus, scAAV, vectors promote efficient transduction independently of DNA synthesis. Gene Ther 8:1248–1254 Zolotukhin S, Potter M, Zolotukhin I, Sakai Y, Loiler S, Fraites TJ, Jr., Chiodo VA, Phillipsberg T, Muzyczka N, Hauswirth WW, Flotte TR, Byrne BJ, Snyder RO, 2002, Production and purification of serotype 1, 2, and 5 recombinant adeno-associated viral vectors. Methods 28:158–167 Harrison JK, Luo D, Streit WJ, 2003, In situ hybridization analysis of chemokines and chemokine receptors in the central nervous system. Methods 29:312–318 Qiu J, Ai L, Ramachandran C, Yao B, Gopalakrishnan S, Fields CR, Delmas AL, Dyer LM, Melnick SJ, Yachnis AT, Schwartz PH, Fine HA, Brown KD, Robertson KD, 2008, Invasion suppressor cystatin E/M, CST6): high-level cell type-specific expression in normal brain and epigenetic silencing in gliomas. Lab Invest 88:910–925 Liu C, Luo D, Streit WJ, Harrison JK, 2008, CX3CL1 and CX3CR1 in the GL261 murine model of glioma: CX3CR1 deficiency does not impact tumor growth or infiltration of microglia and lymphocytes. J Neuroimmunol 198:98–105 Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P, 2007, The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114:97–109 Pescarmona GP, Scalerandi M, Delsanto PP, Condat CA, 1999, Non-linear model of cancer growth and metastasis: a limiting nutrient as a major determinant of tumor shape and diffusion. Med Hypotheses 53:497–503 Seiki M, 2003, Membrane-type 1 matrix metalloproteinase: a key enzyme for tumor invasion. Cancer Lett 194:1–11 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 437 EP 444 DI 10.1007/s12253-008-9147-4 PG 8 ER PT J AU Papay, J Sapi, Z Egri, G Gyulai, M Szende, B Losonczy, Gy Timar, J Moldvay, J AF Papay, Judit Sapi, Zoltan Egri, Gabor Gyulai, Marton Szende, Bela Losonczy, Gyorgy Timar, Jozsef Moldvay, Judit TI Platinum-Based Chemotherapy in Lung Cancer Affects the Expression of Certain Biomarkers Including ERCC1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chemotherapy; Immunohistochemistry; Lung cancer; Molecular biology; Neoadjuvant treatment ID Chemotherapy; Immunohistochemistry; Lung cancer; Molecular biology; Neoadjuvant treatment AB Chemotherapies are widely used in the treatment of lung cancer. However, little is known about their effect in the expression of different tissue markers. Seventeen lung cancer tissue blocks obtained by bronchoscopic biopsies together with their corresponding surgical biopsies after neoadjuvant chemotherapy were studied. They included 9 adenocarcinomas (ADC) and 8 squamous cell carcinomas (SCC). Immunohistochemistry was performed on formalinfixed, paraffin-embedded tissues to study the expression of Ki-67, p53, Bcl-2, Bax, Fas-ligand and ERCC1 (excision repair cross-complementation group 1). Out of 17 NSCLC 6 expressed proapoptotic markers and 4 expressed antiapoptotic markers, while in 7 cases the apoptotic markers did not show detectable changes after neoadjuvant chemotherapy. Six of 17 bronchoscopic NSCLC cases expressed increased level of Ki-67 after neoadjuvant treatment. Eight bronchoscopic NSCLC tissues (6 SCC, 2 ADC) expressed ERCC1. All but one ADC became ERCC1 negative after neoadjuvant therapy. There was no newly expressed ERCC1 positive case in the surgical biopsy group. Platinum-based neoadjuvant chemotherapy had no effect on the apoptotic activity of 17 patients’ tumor specimen, however, 6 of 17 bronchoscopic NSCLC cases expressed increased level of Ki-67 after neoadjuvant treatment, in 3 cases the level of Ki-67 became decreased, while 8 cases had no detectable change of proliferation activity. The results of the present study suggest that platinum-based chemotherapy probably induces a selection of tumor cells with more aggressive phenotype, and also affects the expression of tissue marker (ERCC1) that could have predictive value. C1 [Papay, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Egri, Gabor] Bajcsy Hospital, Department of SurgeryBudapest, Hungary. [Gyulai, Marton] County Hospital of PulmonologyTorokbalint, Hungary. [Szende, Bela] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Losonczy, Gyorgy] Semmelweis University, Department of Pulmonology, Diosarok u. 1/c., H-1125 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Moldvay, Judit] Semmelweis University, Department of Pulmonology, Diosarok u. 1/c., H-1125 Budapest, Hungary. RP Moldvay, J (reprint author), Semmelweis University, Department of Pulmonology, H-1125 Budapest, Hungary. EM drmoldvay@hotmail.com CR U.S. Cancer Statistics Working Group. United States Cancer Statistics: 2002–2004 Incidence and Mortality Web-based Report Version. Atlanta, GA): Department of Health and Human Services, Centers for Disease Control and Prevention, and National Cancer Institute; 2007 Noble J, Ellis PM, Mackay JA et al, 2006, Second-line or subsequent systemic therapy for recurrent or progressive nonsmall cell lung cancer: a systematic review and practice guideline. J Thorac Oncol 9:1042–58 Junker K, Langner K, Klinke F et al, 2001, Grading of tumor regression in non-small cell lung cancer: morphology and prognosis. Chest. 120(5):1584–91 Morero JL, Poleri C, Martin C et al, 2007, Influence of apoptosis and cell cycle regulator proteins on chemotherapy response and survival in stage IIIA/IIIB NSCLC patients. J Thorac Oncol. 4:293–8 Ikuta K, Takemura K, Kihara M et al, 2005, Defects in apoptotic signal transduction in cisplatin-resistant non-small cell lung cancer cells. Oncol Rep. 13(6):1229–34 Filipits M, Pirker R, Dunant A et al, 2007, Cell cycle regulators and outcome of adjuvant cisplatin-based chemotherapy in completely resected non-small-cell lung cancer: the International Adjuvant Lung Cancer Trial Biologic Program. J Clin Oncol. 25, 19):2735–40 Olaussen KA, Dunant A, Fouret P et al, 2006, DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N Engl J Med 355:983–91 Rigas JR, Kelly K, 2007, Current treatment paradigms for locally advanced non-small cell lung cancer. J Thorac Oncol. 2(Suppl 2): S77–85 Gilligan D, Nicolson M, Smith I et al, 2007, Preoperative chemotherapy in patients with resectable non-small cell lung cancer: results of the MRC LU22/NVALT 2/EORTC 08012 multicentre randomised trial and update of systematic review. Lancet. 369(9577):1929–37 de Marinis F, Tedesco B, Treggiari S et al, 2007, Role of induction chemotherapy in resectable N2 non-small cell lung cancer. J Thorac Oncol. 5(Suppl 5):S31–4 Abratt RP, Lee JS, Han JY et al, 2006, Phase II trial of gemcitabine-carboplatin-paclitaxel as neoadjuvant chemotherapy for operable non-small cell lung cancer. J Thorac Oncol. 2:135–40 Stinchcombe TE, Socinski MA, 2007, The role of induction therapy for resectable non-small cell lung cancer. Drugs. 67, 3):321–32 De Pauw R, van Meerbeeck JP, 2007, Neoadjuvant chemotherapy in the treatment of nonsmall-cell lung cancer. Curr Opin Oncol. 19, 2):92–7 Meert AP, Martin B, Verdebout JM et al, 2004, Correlation of different markers, p53, EGF-R, c-erbB-2, Ki-67, expression in the diagnostic biopsies and the corresponding resected tumors in nonsmall cell lung cancer. Lung Cancer. 44(3):295–301 Lang DS, Droemann D, Schultz H et al, 2007, A novel human ex vivo model for the analysis of molecular events during lung cancer chemotherapy. Respir Res. 14;8:43 Fujii T, Toyooka S, Ichimura K et al, 2008, ERCC1 protein expression predicts the response of cisplatin-based neoadjuvant chemotherapy in non-small-cell lung cancer. Lung Cancer 59, 3):377–84 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 445 EP 450 DI 10.1007/s12253-009-9155-z PG 6 ER PT J AU Younes, AS Csire, M Kapusinszky, B Szomor, K Takacs, M Berencsi, Gy AF Younes, Ali Saleh Csire, Marta Kapusinszky, Beatrix Szomor, Katalin Takacs, Maria Berencsi, Gyorgy TI Heterogeneous Pathways of Maternal-fetal Transmission of Human Viruses (Review) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Syncytiotrophoblast; Fetal endothelium; Virus transcytosis; Maternal-fetal transport ID Syncytiotrophoblast; Fetal endothelium; Virus transcytosis; Maternal-fetal transport AB Several viruses can pass the maternal-fetal barrier, and cause diseases of the fetus or the newborn. Recently, however, it became obvious, that viruses may invade fetal cells and organs through different routes without acute consequences. Spermatozoa, seminal fluid and lymphocytes in the sperm may transfer viruses into the human zygotes. Viruses were shown to be integrated into human chromosomes and transferred into fetal tissues. The regular maternal-fetal transport of maternal cells has also been discovered. This transport might implicate that lymphotropic viruses can be released into the fetal organs following cellular invasion. It has been shown that many viruses may replicate in human trophoblasts and syncytiotrophoblast cells thus passing the barrier of the maternalfetal interface. The transport of viral immunocomplexes had also been suggested, and the possibility has been put forward that even anti-idiotypes mimicking viral epitopes might be transferred by natural mechanisms into the fetal plasma, in spite of the selective mechanisms of apical to basolateral transcytosis in syncytiotrophoblast and basolateral to apical transcytosis in fetal capillary endothelium. The mechanisms of maternal-fetal transcytosis seem to be different of those observed in differentiated cells and tissue cultures. Membrane fusion and lipid rafts of high cholesterol content are probably the main requirements of fetal transcytosis. The long term presence of viruses in fetal tissues and their interactions with the fetal immune system might result in post partum consequences as far as increased risk of the development of malignancies and chronic pathologic conditions are discussed. C1 [Younes, Ali Saleh] National Center for Epidemiology, Division of Virology, Gyali Street 2–6, 1097 Budapest, Hungary. [Csire, Marta] National Center for Epidemiology, Division of Virology, Gyali Street 2–6, 1097 Budapest, Hungary. [Kapusinszky, Beatrix] National Center for Epidemiology, Division of Virology, Gyali Street 2–6, 1097 Budapest, Hungary. [Szomor, Katalin] National Center for Epidemiology, Division of Virology, Gyali Street 2–6, 1097 Budapest, Hungary. [Takacs, Maria] National Center for Epidemiology, Division of Virology, Gyali Street 2–6, 1097 Budapest, Hungary. [Berencsi, Gyorgy] National Center for Epidemiology, Division of Virology, Gyali Street 2–6, 1097 Budapest, Hungary. RP Berencsi, Gy (reprint author), National Center for Epidemiology, Division of Virology, 1097 Budapest, Hungary. EM berencsi.gyorgy@oek.antsz.hu CR Aikhionbare FO, Kumaresan K, Shamsa F, Bond VC, 2006, HLA-G DNA sequence variants and risk of perinatal HIV-1 transmission. AIDS Res Ther 23:28–36 Aparicio O, Razquin N, Zaratiegui M et al, 2006, Adenovirus virus-associated RNA is processed to functional interfering RNAs involved in virus production. J Virol 80:1376–1384 Armbruster-Moraes E, Ishimoto LM, Leao E, Zugaib M, 1992, Detection of human papillomavirus deoxyribonucleic acid sequences in amniotic fluid during different periods of pregnancy. Am J Obstet Gynecol 166:35–40 Armbruster-Moraes E, Ishimoto LM, Leao E, Zugaib M, 1994, Presence of human papillomavirus DNA in amniotic fluids of pregnant women with cervical lesions. Gynecol Oncol 54:152– 158 Ashshi AM, Cooper RJ, Klapper PE et al, 2000, Detection of human herpes virus 6 DNA in fetal hydrops. Lancet 355:1519– 1520 Avgil M, Ornoy A, 2006, Herpes simplex virus and Epstein-Barr virus infections in pregnancy: consequences of neonatal or intrauterine infection. Reprod Toxicol 21:436–445 Aynaud O, Poveda JD, Huynh B et al, 2002, Frequency of herpes simplex virus, cytomegalovirus and human papillomavirus DNA in semen. Int J STD AIDS 13:547–550 Bacsi A, Aranyosi J, Beck Z et al, 1999, Placental macrophage contact potentiates the complete replicative cycle of human cytomegalovirus in syncytiotrophoblast cells: role of interleukin- 8 and transforming growth factor-beta1. J Interferon Cytokine Res 19:1153–1160 Bacsi A, Csoma E, Beck Z et al, 2001, Induction of HIV-1 replication in latently infected syncytiotrophoblast cells by contact with placental macrophages: role of interleukin-6 and tumor necrosis factor-alpha. J Interferon Cytokine Res 21:1079–1088 Bagasra O, Patel D, Bobroski L et al, 1971, Uptake of heterologous genome by mammalian spermatozoa and its transfer to ova through fertilization. Proc Natl Acad Sci USA 68:353–357 Bagasra O, Patel D, Bobroski L et al, 2005, Localization of human herpesvirus type 8 in human sperms by in situ PCR. J Mol Histol 36:401–412 Baillargeon J, Piper J, Leach CT, 2000, Epidemiology of human herpesvirus 6, HHV-6, infection in pregnant and nonpregnant women. J Clin Virol 16:149–157 Bandi S, Akkina R.: Human embryonic stem cell, hES, derived dendritic cells are functionally normal and are susceptible to HIV-1 infection. AIDS Res Ther. 23;5:1–12, 2008. Bandobashi K, Daibata M, Kamioka M et al, 1997, Human herpesvirus 6, HHV-6)-positive Burkitt's lymphoma: establishment of a novel cell line infected with HHV-6. Blood 90:1200– 1207 Barel MT, Ressing M, Pizzato N et al, 2003, Human cytomegalovirus-encoded US2 differentially affects surface expression of MHC class I locus products and targets membrane-bound, but not soluble HLA-G1 for degradation. J Immunol 171:6757–6765 Bartlett JS, Wilcher R, Samulski RJ, 2000, Infectious entry pathway of adeno-associated virus and adeno-associated virus vectors. J Virol 74:2777–2785 Beer AE, Kwak JY, Ruiz JE, 1994, The biological basis of passage of fetal cellular material into the maternal circulation: a review. Ann N Y Acad Sci 731:1–35 Bezold G, Politch JA, Kiviat NB et al, 2006, Evidence of shared Epstein-Barr viral isolates between sexual partners, and low level EBV in genital secretions. J Med Virol 78:1204–1209 Bezold G, Politch JA, Kiviat NB et al, 2007, Prevalence of sexually transmissible pathogens in semen from asymptomatic male infertility patients with and without leukocytospermia. Fertil Steril 87:1087–1097 Bhat P, Anderson DA, 2007, Hepatitis B virus translocates across a trophoblastic barrier. J Virol 81:7200–7207 Bhoopat L, Khunamornpong S, Sirivatanapa P et al, 2005, Chorioamnionitis is associated with placental transmission of human immunodeficiency virus-1 genotype E in the early gestational period. Mod Pathol 18:1357–1364 Boggess KA, Moss K, Madianos P et al, 2005, Fetal immune response to oral pathogens and risk of preterm birth. Am J Obstet Gynecol 193:1121–1126 Bright NA, Ockleford CD, Anwar M, 1994, Ontogeny and distribution of Fc gamma receptors in the human placenta. Transport or immune surveillance? J Anat 184:297–308 Brudek T, Christensen T, Hansen HJ et al, 2004, Simultaneous presence of endogenous retrovirus and herpes virus antigens has profound effect on cell-mediated immune responses: implications for multiple sclerosis. AIDS Res Hum Retroviruses 20:415–423 Burguete T, Rabreau M, Fontanges-Darriet M et al, 1999, Evidence for infection of the human embryo with adenoassociated virus in pregnancy. Hum Reprod 14:2396–2401 Cantalupo P, Doering A, Sullivan CS et al, 2005, Complete nucleotide sequence of polyomavirus SA12. J Virol 79:13094– 13104 Cason J, Rice P, Best JM, 1998, Transmission of cervical cancerassociated human papilloma viruses from mother to child. Intervirol 41:213–218 Cerboni C, Achour A, Warnmark A et al, 2006, Spontaneous mutations in the human CMV HLA class I homologue UL18 affect its binding to the inhibitory receptor LIR-1/ILT2/CD85j. Eur J Immunol 36:732–741 Chan PK, Tam WH, Yeo Wet al, 2001, High carriage rate of TT virus in the cervices of pregnant women. Clin Infect Dis 32:1376–1377 Christensen T, 2005, Association of human endogenous retroviruses with multiple sclerosis and possible interactions with herpes viruses. Rev Med Virol 15:179–211 Consigli RA, Haynes JI Jr, Chang D et al, 1992, Early events of polyoma infection: adsorption, penetration and nuclear transport. Trans Kans Acad Sci 95:62–69 Cross JC, Lam S, Yagel S, Werb Z, 1999, Defective induction of the transcription factor interferon-stimulated gene factor-3 and interferon alpha insensitivity in human trophoblast cells. Biol Reprod 60:312–321 Csata S, Kulcsar G, 1991, Virus-host studies in human seminal and mouse testicular cells. Acta Chir Hung 32:83–90 Csire M, Mikala G, Peto M et al, 2007, Detection of four lymphotropic herpesviruses in Hungarian patients with multiple myeloma and lymphoma. FEMS Immunol Med Microbiol 49:62– 67 Csoma E, Bacsi A, Liu X et al, 2002, Human herpesvirus 6 variant a infects human term syncytiotrophoblasts in vitro and induces replication of human immunodeficiency virus type 1 in dually infected cells. J Med Virol 67:67–87 Cupers P, Veithen A, Kiss A et al, 1994, Clathrin polymerization is not required for bulk-phase endocytosis in rat fetal fibroblasts. J Cell Biol 127:725–735 Cuss AK, Avery DT, Cannons JL et al, 2006, Expansion of functionally immature transitional B cells is associated with human-immunodeficient states characterized by impaired humoral immunity. J Immunol 176:1506–1516 Czegledy J, 2001, Sexual and non-sexual transmission of human papillomavirus. Acta Microbiol Immunol Hung 48:511–517 Daibata M, Taguchi T, Nemoto Y et al, 1999, Inheritance of chromosomally integrated human herpesvirus 6 DNA. Blood 94:1545–1549 de Haan TR, Beersma MF, Claas EC et al, 2007, Parvovirus B19 infection in pregnancy studied by maternal viral load and immune responses. Fetal Diagn Ther 22:55–62 de-The G, Geser A, Day NE et al, 1978, Epidemiological evidence for causal relationship between Epstein-Barr virus and Burkitt's lymphoma from Ugandan prospective study. Nature 274:756–761 De-The G, 1979, The epidemiology of Burkitt's lymphoma: evidence for a causal association with Epstein-Barr virus. Epidemiol Rev 1:32–54 Derrien M, Pizzato N, Dolcini G et al, 2004, Human immunodeficiency virus 1 downregulates cell surface expression of the non-classical major histocompatibility class I molecule HLA-G1. J Gen Virol 85:1945–1954 Devergne O, Coulomb-L'Hermine A, Capel F et al, 2001, Expression of Epstein-Barr virus-induced gene 3, an interleukin- 12 p40-related molecule, throughout human pregnancy: involvement of syncytiotrophoblasts and extravillous trophoblasts. Am J Pathol 159:1763–1776 Distefano AL, Alonso A, Martin F, Pardon F, 2004, Human cytomegalovirus: detection of congenital and perinatal infection in Argentina. BMC Pediatr 4:11–18 Drobni P, Naslund J, Evander M, 2004, Lactoferrin inhibits human papillomavirus binding and uptake in vitro. Antiviral Res 64:63–68 Dye JF, Vause S, Johnston T et al, 2004, Characterization of cationic amino acid transporters and expression of endothelial nitric oxide synthase in human placental microvascular endothelial cells. FASEB J 18:125–127 El Borai N, LeFevre C, Inoue M et al, 1998, Presence of HSV-1 DNA in semen and menstrual blood. J Reprod Immunol 41:137– 147 Elustondo PA, Hannigan GE, Caniggia I, MacPhee DJ, 2006, Integrin-linked kinase, ILK, is highly expressed in first trimester human chorionic villi and regulates migration of a human cytotrophoblast-derived cell line. Biol Reprod 74:959–968 Enbom M, Strand A, Falk KI, Linde A, 2001, Detection of Epstein-Barr virus, but not human herpesvirus 8, DNA in cervical secretions from Swedish women by real-time polymerase chain reaction. Sex Transm Dis 28:300–306 Ewers H, Smith AE, Sbalzarini IF et al, 2005, Single-particle tracking of murine polyoma virus-like particles on live cells and artificial membranes. Proc Natl Acad Sci USA 102:15110–15115 Fahim MS, Brawner TA, 1980, Treatment of genital herpes simplex virus in male patients. Arch Androl 4:79–85 Feire AL, Koss H, Compton T, 2004, Cellular integrins function as entry receptors for human cytomegalovirus via highly conserved disintegrin-like domain. Proc Natl Acad Sci US 101:15470–15475 Ferry H, Crockford TL, Leung JC, Cornall RJ, 2006, Signals from a self-antigen induce positive selection in early B cell ontogeny but are tolerogenic in adults. J Immunol 176:7402–7411 Fine PE, Adelstein AM, Snowman J et al, 1985, Long term effects of exposure to viral infections in utero. Br Med J, Clin Res Ed, 290:509–511 Fisher S, Genbacev O, Maidji E, 2000, Pereira, L: human cytomegalovirus infection of placental cyncytiotrophoblasts in vitro and in utero: implications for transmission and pathogenesis. J Virol 74:6808–6820 Fjaertoft G, Hakansson L, Foucard T et al, 2000, CD64, Fcgamma receptor I, cell surface expression on maturing neutrophils from preterm and term newborn infants. Acta Paediatr 94:295–302 Foraker AB, Ray A, Da Silva TC et al, 2007, Dynamin 2 regulates riboflavin endocytosis in human placental trophoblasts. Mol Pharmacol 72:553–562 Forster-Waldl E, Sadeghi K, Tamandl D et al, 2005, Monocyte toll-like receptor 4 expression and LPS-induced cytokine production increase during gestational aging. Pediatr Res 58:121–124 Fule T, Mathe M, Suba Z et al, 2006, The presence of human papillomavirus 16 in neural structures and vascular endothelial cells. Virology 348:289–296 Gazit E, Sherf M, Balbin E et al, 2007, HLA-G expression is induced in Epstein-Barr virus-transformed B-cell lines by culture conditions. Hum Immunol 68:463–468 Geser A, de The G, Lenoir G et al, 1982, Final case reporting from the Ugandan prospective study of the relationship between EBV and Burkitt's lymphoma. Int J Cancer 29:397–400 Gibson CS, MacLennan AH, Goldwater PN et al, 2006, South Australian cerebral palsy research group: neurotropic viruses and cerebral palsy: population based case-control study. BMJ 332:76– 80 Gilbert J, Benjamin T, 2004, Uptake pathway of polyomavirus via ganglioside GD1a. J Virol 78:12259–12267 Gotherstrom C, Johnsson AM, Mattsson J et al, 2005, Identification of maternal hematopoietic cells in a 2nd-trimester fetus. Fetal Diagn Ther 20:355–358 Gouarin S, Gault E, Vabret A et al, 2002, Real-time PCR quantification of human cytomegalovirus DNA in amniotic fluid samples of mothers with primary infection. J Clin Microbiol 40:1767–1772 Grey F, Meyers H, White EA, 2007, A human cytomegalovirusencoded microRNA regulates expression of multiple viral genes involved in replication. PLoS Pathog 3:e163 Gu J, Xie Z, Gao Z et al, 2007, H5N1 infection of the respiratory tract and beyond: a molecular pathology study. Lancet 370:1137– 1145 Gupta P, Singh MK, Rinaldo C et al, 1996, Detection of Kaposi's sarcoma herpesvirus DNA in semen of homosexual men with Kaposi's sarcoma. AIDS 10:1596–1598 Hall CB, Caserta MT, Schnabel KC et al, 2004, Congenital infections with human herpesvirus 6, HHV6, and human herpesvirus 7, HHV7). J Pediatr 145:432–435 Hamkar R, Jalilvand S, Abdolbaghi MH et al, 2006, Inadvertent rubella vaccination of pregnant women: evaluation of possible transplacental infection with rubella vaccine. Vaccine 24:3558– 3563 Hanna J, Wald O, Goldman-Wohl D et al, 2003, CXCL12 expression by invasive trophoblasts induces the specific migration of CD16- human natural killer cells. Blood 102:569–577 Hansen GH, Niels-Christiansen LL, Immerdal L et al, 1999, Transcytosis of immunoglobulin A in the mouse enterocyte occurs through glycolipid raft- and rab17-containing compartments. Gastroenterology 116:610–622 Hiroshi C, Ito K, Niikura H, 2006, Clinical manifestations and outcomes of parvovirus B19 infection during pregnancy in Japan. Tohoku J Exp Med 209:277–283 Howard MR, Whitby D, Bahadur G et al, 1997, Detection of human herpesvirus 8 DNA in semen from HIV-infected individuals but not healthy semen donors. AIDS 11:F15–F19 Hsiao FC, Lin M, Tai A et al, 2006, Cutting edge: Epstein-Barr virus transactivates the HERV-K18 superantigen by docking to the human complement receptor 2, CD21, on primary B cells. J Immunol 177:2056–2060 Hsu CS, Chou SY, Liang SJ et al, 2005, Effect of glutamine on cell adhesion molecule expression and leukocyte transmigration in endothelial cells stimulated by preeclamptic plasma. Nutrition 21:1134–1140 Huang YQ, Li JJ, Poiesz BJ et al, 1997, Detection of the herpesvirus-like DNA sequences in matched specimens of semen and blood from patients with AIDS-related Kaposi's sarcoma by polymerase chain reaction in situ hybridization. Am J Pathol 150:147–153 Hunt JS, Petroff MG, McIntire RH, Ober C, 2005, HLA-G and immune tolerance in pregnancy. The FASEB J 19:681–693 Ismail NA, Kampan N, Mahdy ZA et al, 2006, Dengue in pregnancy. Southeast Asian J Trop Med Public Health 37:681–683 Jakobovits A, 2006, [Intrauterine infections] review in Hungarian. Orv Hetil. 147:1723–1730 Jaleel MA, Tsai AC, Sarkar S et al, 2004, Stromal cell-derived factor-1, SDF-1, signalling regulates human placental trophoblast cell survival. Mol Hum Reprod 10:901–909 Jun Y, Kim E, Jin M et al, 2000, Human cytomegalovirus gene products US3 and US6 down-regulate trophoblast class I MHC molecules. J Immunol 164:805–811 Kail M, Hollinshead M, Ansorge W et al, 1991, The cytoplasmic domain of alphavirus E2 glycoprotein contains a short linear recognition signal required for viral budding. EMBO J 10:2343– 2351 Kaleeba JA, Berger EA, 2006, Kaposi's sarcoma-associated herpesvirus fusion-entry receptor: cystine transporter xCT. Science 311:1921–1924 Kapranos N, Petrakou E, Anastasiadou C, Kotronias D, 2003, Detection of herpes simplex virus, cytomegalovirus, and Epstein- Barr virus in the semen of men attending an infertility clinic. Fertil Steril 3:1566–1570 79 Suppl Kaufmann B, Simpson AA, Rossmann MG, 2004, The structure of human parvovirus B19. Proc Natl Acad Sci USA 101:11628– 11633 Keay S, Merigan TC, Rasmussen L, 1989, Identification of cell surface receptors for the 86-kilodalton glycoprotein of human cytomegalovirus. Proc Natl Acad Sci USA 86:10100–10103 Keay S, Baldwin B, 1991, Anti-idiotype antibodies that mimic gp86 of human cytomegalovirus inhibit viral fusion but not attachment. J Virol 65:5124–5128 Keay S, Baldwin B, 1992, The human fibroblast receptor for gp86 of human cytomegalovirus is a phosphorylated glycoprotein. J Virol 66:4834–4838 Kerr JR, 2005, Pathogenesis of parvovirus B19 infection: host gene variability, and possible means and effects of virus persistence. J Vet Med B Infect Dis Vet Public Health 52:335–339 Khosrotehrani K, Johnson KL, Cha DH et al, 2004, Transfer of fetal cells with multilineage potential to maternal tissue. JAMA 292:75–80 Kinzler ER, Compton T, 2005, Characterization of human cytomegalovirus glycoprotein-induced cell-cell fusion. J Virol 79:7827–7837 Klein E, Kis LL, Takahara M, 2006, Pathogenesis of Epstein- Barr virus, EBV)-carrying lymphomas. Acta Microbiol Immunol Acad Sci Hung 53:441–457 Koizumi N, Mizuguchi H, Kondoh M et al, 2004, Efficient gene transfer into human trophoblast cells with adenovirus vector containing chimeric type 5 and 35 fiber protein. Biol Pharm Bull 27:2046–2048 Koizumi N, Mizuguchi H, Kondoh M et al, 2006, Efficient gene transfer into differentiated human trophoblast cells with adenovirus vector containing RGD motif in the fiber protein. Biol Pharm Bull 29:1297–1299 Kotronias D, Kapranos N, 1998, Detection of herpes simplex virus DNA in human spermatozoa by in situ hybridization technique. In Vivo 12:391–394 Kudo Y, Boyd C, 2004, RNA interference-induced reduction in CD98 expression suppresses cell fusion during syncytialization of human placental BeWo cells. FEBS Lett 577:473–477 Kwun HJ, Han HJ, LeeWJ et al, 2002, Transactivation of the human endogenous retrovirus K long terminal repeat by herpes simplex virus type 1 immediate early protein 0. Virus Res 86:93–100 Lafon M, Prehaud C, Megret F et al, 2005, Modulation of HLAG expression in human neural cells after neurotropic viral infections. J Virol 79:15226–15237 Lagaye S, Derrien M, Menu E et al, 2001, European Network for the Study of In Utero Transmission of HIV-1. Cell-to-cell contact results in a selective translocation of maternal human immunodeficiency virus type 1 quasispecies across a trophoblastic barrier by both transcytosis and infection. J Virol 75:4780–4791 Lajoie J, Hargrove J, Zijenah LS et al, 2006, Genetic variants in nonclassical major histocompatibility complex class I human leukocyte antigen, HLA)-E and HLA-G molecules are associated with susceptibility to heterosexual acquisition of HIV-1. J Infect Dis 193:298–301 Lambot N, Lybaert P, Boom A et al, 2006, Evidence for a clathrin-mediated recycling of albumin in human term placenta. Biol Reprod 75:90–97 Lampinen TM, Kulasingam S, Min J et al, 2000, Detection of Kaposi's sarcoma-associated herpesvirus in oral and genital secretions of Zimbabwean women. J Infect Dis 181:1785–1790 Lee HK, Lee HH, Park YM et al, 1991, Regulation of human B cell proliferation and differentiation by seminal plasma. Clin Exp Immunol 85:174–179 Lee WJ, Kwun HJ, Kim HS, Jang KL, 2003, Activation of the human endogenous retrovirus W long terminal repeat by herpes simplex virus type 1 immediate early protein 1. Mol Cells 15: 75–80 Lee J, Sugden B, 2007, A membrane leucine heptad contributes to trafficking, signaling, and transformation by latent membrane protein 1. J Virol 81:9121–9130 Liebl D, Difato F, Hornikova L et al, 2006, Mouse polyomavirus enters early endosomes, requires their acidic pH for productive infection, and meets transferrin cargo in Rab11-positive endosomes. J Virol 80:4610–4622 Lim CJ, Norouziyan F, Shen WC, 2007, Accumulation of transferrin in Caco-2 cells: a possible mechanism of intestinal transferrin absorption. J Control Release 122:393–398 Linton EA, Rodriguez-Linares B, Rashid-Doubell F et al, 2003, Caveolae and caveolin-1 in human term villous trophoblast. Placenta 24:45–57 Lyden TW, Anderson CL, Robinson JM, 2002, The endothelium but not the syncytiotrophoblast of human placenta expresses caveolae. Placenta 23:640–652 Luppi M, Torelli G, 1996, The new lymphotropic herpesviruses, HHV-6, HHV-7, HHV-8, and hepatitis C virus, HCV, in human lymphoproliferative diseases: an overview. Haematologica 81:265–281 Luppi M, Barozzi P, Bosco R et al, 2006, Human herpesvirus 6 latency characterized by high viral load: chromosomal integration in many, but not all, cells. J Infect Dis 194:1020–1021 Maidji E, McDonagh S, Genbacev O et al, 2006, Maternal antibodies enhance or prevent cytomegalovirus infection in the placenta by neonatal Fc receptor-mediated transcytosis. Am J Pathol 168:1210–1226 Maidji E, Genbacev O, Chang HT, Pereira L, 2007, Developmental regulation of human cytomegalovirus receptors in cytotrophoblasts correlates with distinct replication sites in the placenta. J Virol 81:4701–4712 Marazuela M, Martin-Belmonte F, Garcia-Lopez MA et al, 2004, Expression and distribution of MAL2, an essential element of the machinery for basolateral-to-apical transcytosis, in human thyroid epithelial cells. Endocrinology 145:1011–1016 MartinettiM, Pacati I, CucciaMet al, 2006, B: Hierarchy of babylinked immunogenetic risk factors in the vertical transmission of hepatitis C virus. Int J Immunopathol Pharmacol 19:369–378 Martinez I, Gardiner AS, Board KF et al, 2008, Human papillomavirus type 16 reduces the expression of microRNA- 218 in cervical carcinoma cells. Oncogene 27:2575–2582 McCormick JN, Wojtacha D, Edmond E et al, 1988, Do polyclonal rheumatoid factors carry an ‘internal image’ of cytomegalovirus, Epstein-Barr virus and nuclear antigens? Scand J Rheumatol Suppl 75:109–116 McClure LV, Sullivan CS, 2008, Kaposi's sarcoma herpes virus taps into a host microRNA regulatory network. Cell Host Microbe 3:1–3 McLean LK, Chehab FF, Goldberg JD, 1995, Detection of viral deoxyribonucleic acid in the amniotic fluid of low-risk pregnancies by polymerase chain reaction. Am J Obstet Gynecol 173:1282–1286 McShane MP, Longnecker R, 2004, Cell-surface expression of a mutated Epstein-Barr virus glycoprotein B allows fusion independent of other viral proteins. Proc Natl Acad Sci USA 101:17474–17479 Meffre E, Salmon JE, 2007, Autoantibody selection and production in early human life. J Clin Invest 117:598–601 Megret F, Prehaud C, Lafage M et al, 2007, Modulation of HLA-G and HLA-E expression in human neuronal cells after rabies virus or herpes virus simplex type 1 infections. Hum Immunol 68:294–302 Mellembakken JR, Aukrust P, Olafsen MK et al, 2002, Activation of leukocytes during the uteroplacental passage in preeclampsia. Hypertension 39:155–160 Mi S, Lee X, Li X et al, 2000, Syncytin is a captive retroviral envelope protein involved in human placental morphogenesis. Nature 403:785–789 Minarovits J, Hu LF, Imai S et al, 1994, Clonality, expression and methylation patterns of the Epstein-Barr virus genomes in lethal midline granulomas classified as peripheral angiocentric T cell lymphomas. J Gen Virol 75:77–84 Mishima T, Kurasawa G, Ishikawa G et al, 2007, Endothelial expression of Fc gamma receptor IIb in the full-term human placenta. Placenta 28:170–174 Montgomery JD, Jacobson LP, Dhir R, Jenkins FJ, 2005, Localization of human herpesvirus type 8 in human sperms by in situ PCR. J Mol Histol 36:401–412 Montgomery JD, Jacobson LP, Dhir R, Jenkins FJ, 2006, Detection of human herpesvirus 8, HHV-8, in normal prostates. Prostate 66:1302–1310 Mori K, Nishimura M, Tsurudome M et al, 2004, The functional interaction between CD98 and CD147 in regulation of virusinduced cell fusion and osteoclast formation. Med Microbiol Immunol, Berl, 193:155–162 Motsch N, Pfuhl T, Mrazek J et al, 2007, Epstein-Barr Virusencoded latent membrane protein 1, LMP1, induces the expression of the cellular microRNA miR-146a. RNA Biol 4:131–137 Munakata Y, Saito-Ito T, Kumura-Ishii K et al, 2005, Ku80 autoantigen as a cellular coreceptor for human parvovirus B19 infection. Blood 106:3449–3456 Munakata Y, Kato I, Saito T et al, 2006, Human parvovirus B19 infection of monocytic cell line U937 and antibody-dependent enhancement. Virology 345:251–257 Nellaker C, Yao Y, Jones-Brando L et al, 2006, Transactivation of elements in the human endogenous retrovirus W family by viral infection. Retrovirology 3:44–55 Niller HH, Wolf H, Minarovits J, 2007, Epstein-Barr Virus. In: Minarovits J, Gonczol E, Valyi-Nagy T, eds, Latency strategies of herpesviruses. Springer Science, + Business Media, LLC, New York, pp 154–191 Niller HH, Wolf H, Minarovits J, 2008, Regulation and dysregulation of Epstein-Barr virus latency: implications for the development of autoimmune diseases. Autoimmunity, Review, 41:298–328 Nilsson BO, Jin M, Andersson AC et al, 1999, Expression of envelope proteins of endogeneous C-type retrovirus on the surface of mouse and human oocytes at fertilization. Virus Genes 18:115–120 Norja P, Hokynar K, Aaltonen LM et al, 2006, Bioportfolio: lifelong persistence of variant and prototypic erythrovirus DNA genomes in human tissue. Proc Natl Acad Sci USA 103:7450– 7453 Norkin LC, Kuksin D, 2005, The caveolae-mediated sv40 entry pathway bypasses the golgi complex en route to the endoplasmic reticulum. Virol J 2:38–44 Nuckel H, Rebmann V, Durig J et al, 2005, HLA-G expression is associated with an unfavorable outcome and immunodeficiency in chronic lymphocytic leukemia. Blood 105:1694–1698 Odum N, Ledbetter JA, Martin P et al, 1991, Homotypic aggregation of human cell lines by HLA class II-, class Ia- and HLA-G-specific monoclonal antibodies. Eur J Immunol 21:2121–2131 Ohashi M, Yoshikawa T, Ihira M et al, 2002, Reactivation of human herpesvirus 6 and 7 in pregnant women. J Med Virol 67:354–358 Okamoto Y, 1991, Maternal determinants of neonatal immune response: effect of anti-idiotype in the neonate. Adv Exp Med Biol 310:215–222 Otsuki K, Yoda A, Saito H et al, 1999, Amniotic fluid lactoferrin in intrauterine infection. Placenta 20:175–179 Ouellet DL, Plante I, Landry P et al, 2008, Identification of functional microRNAs released through asymmetrical processing of HIV-1 TAR element. Nucleic Acids Res 36:2353–2365 Ordog K, Szendroi A, Szarka K et al, 2003, Perinatal and intrafamily transmission of hepatitis B virus in three generations of a low-prevalence population. J Med Virol 70:194–204 Pangault C, Le Tulzo Y, Minjolle S et al, 2004, HLA-G expression in Guillain-Barre syndrome is associated with primary infection with cytomegalovirus. Viral Immunol 17:123–125 Paradela A, Bravo SB, Henriquez M et al, 2005, Proteomic analysis of apical microvillous membranes of syncytiotrophoblast cells reveals a high degree of similarity with lipid rafts. J Proteome Res 4:2435–2441 Parry S, Holder J, Halterman MW et al, 1998, Transduction of human trophoblastic cells by replication-deficient recombinant viral vectors. Promoting cellular differentiation affects virus entry. Am J Pathol 152:1521–1529 Parry S, Zhang J, Koi H et al, 2006, Transcytosis of human immunodeficiency virus 1 across the placenta is enhanced by treatment with tumour necrosis factor alpha. J Gen Virol 87:2269–2278 Pass RF, 2004, HHV6 and HHV7: persistence and vertical transmission, Editorial). J Pediatrics 145:432–434 Pedersen SM, Oster B, Bundgaard B, Hollsberg P, 2006, Induction of cell-cell fusion from without by human herpesvirus 6B. J Virol 80:9916–9920 Pellett PE, Spira TJ, Bagasra O et al, 1999, Multicenter comparison of PCR assays for detection of human herpesvirus 8 DNA in semen. J Clin Microbiol 37:1298–1301 Petit T, Dommergues M, Socie G et al, 1997, Detection of maternal cells in human fetal blood during the third trimester of pregnancy using allele-specific PCR amplification. Br J Haematol 98:767–771 Pizzato N, Derrien M, Lenfant F, 2004, The short cytoplasmic tail of HLA-G determines its resistance to HIV-1 Nef-mediated cell surface downregulation. Hum Immunol 65:1389–1396 Pizzato M, Helander A, Popova E et al, 2007, Dynamin 2 is required for the enhancement of HIV-1 infectivity by Nef. Proc Natl Acad Sci USA 104:6812–6817 Predescu SA, Predescu DN, Palade GE, 2001, Endothelial transcytotic machinery involves supramolecular protein-lipid complexes. Mol Biol Cell 12:1019–1033 QuerbesW, O'Hara BA, Williams G, Atwood WJ, 2006, Invasion of host cells by JC virus identifies a novel role for caveolae in endosomal sorting of noncaveolar ligands. J Virol 80:9402–9413 Qiu J, Terasaki PI, Miller J et al, 2006, Soluble HLA-G expression and renal graft acceptance. Am J Transplant 6:2152–2156 Rashid-Doubell F, Tannetta D, RedmanCWet al, 2007, Caveolin-1 and lipid rafts in confluent BeWo trophoblasts: evidence for Rock-1 association with caveolin-1. Placenta 28:139–151 Rastogi D, Wang C, Mao X et al, 2007, Antigen-specific immune responses to influenza vaccine in utero. J Clin Invest 117:1637–1646 Rhoades CJ, Williams MA, Kelsey SM, Newland AC, 2000, Monocyte-macrophage system as targets for immunomodulation by intravenous immunoglobulin. Blood Rev 14:14–30 Rice PS, Cason J, Best JM, Banatvala JE, 1999, High risk genital papillomavirus infections are spread vertically. Rev Med Virol 9:15–21 Rice PS, Mant C, Cason J et al, 2000, High prevalence of human papillomavirus type 16 infection among children. J Med Virol 61:70–75 Rogo KO, Nyansera PN, 1989, Congenital condylomata acuminata with meconium staining of amniotic fluid and fetal hydrocephalus: case report. East Afr Med J 66:411–413 Ros C, Gerber M, Kempf C, 2006, Conformational changes in the VP1-unique region of native human parvovirus B19 lead to exposure of internal sequences that play a role in virus neutralization and infectivity. J Virol 80:12017–1224 Roussev RG, Ng SC, Coulam CB, 2007, Natural killer cell functional activity suppression by intravenous immunoglobulin, intralipid and soluble human leukocyte antigen-G. Am J Reprod Immunol 57:262–269 Rouzioux C, Costagliola D, Burgard M et al, 1995, Estimated timing of mother-to-child human immunodeficiency virus type 1, HIV-1, transmission by use of a Markov model: the HIV Infection in Newborns French Collaborative Study Group. Am J Epidemiol 142:1330–1337 Ruprecht K, Obojes K, Wengel V et al, 2006, Regulation of human endogenous retrovirus W protein expression by herpes simplex virus type 1: implications for multiple sclerosis. J Neurovirol 12:65–71 Ryckman BJ, Jarvis MA, Drummond DD et al, 2006, Human cytomegalovirus entry into epithelial and endothelial cells depends on genes UL128 to UL150 and occurs by endocytosis and low-pH fusion. J Virol 80:710–722 Sadagopan S, Sharma-Walia N, Veettil MVet al, 2007, Kaposi's sarcoma-associated herpesvirus induces sustained NF-kappaB activation during de novo infection of primary human dermal microvascular endothelial cells that is essential for viral gene expression. J Virol 81:3949–3968 Sashihara J, Tanaka-Taya K, Tanaka S et al, 2002, High incidence of human herpesvirus 6 infection with a high viral load in cord blood stem cell transplant recipients. Blood 100:2005–2011 Sathish N, Abraham P, Peedicayil A et al, 2004, HPV DNA in plasma of patients with cervical carcinoma. J Clin Virol 31:204–209 Schroder J, 1974, Passage of leukocytes from mother to fetus. Scand J Immunol 3:369–373 Schust DJ, Hill AB, Ploegh HL, 1996, Herpes simplex virus blocks intracellular transport of HLA-G in placentally derived human cells. J Immunol 157:3375–3380 Schust DJ, Tortorella D, Seebach J et al, 1998, Trophoblast class I major histocompatibility complex, MHC, products are resistant to rapid degradation imposed by the human cytomegalovirus, HCMV, gene products US2 and US11. J Exp Med 188:497–503 Schust DJ, Tortorella D, Ploegh HL, 1999, HLA-G and HLA-C at the feto-maternal interface: lessons learned from pathogenic viruses. Semin Cancer Biol 9:37–46 Sherman JK, Menna JH, 1986, Cryosurvival of herpes simplex virus-2 during cryopreservation of human spermatozoa. Cryobiology 23:383–385 Sieczkarski SB, Whittaker GR et al, 2002, Dissecting virus entry via endocytosis. J Gen Virol 83:1535–1545 Simister NE, 2003, Placental transport of immunoglobulin G. Vaccine 21:3365–3369 Sinclair AJ, Farrell PJ, 1995, Host cell requirements for efficient infection of quiescent primary B lymphocytes by Epstein-Barr virus. J Virol 69:5461–5468 Smith EM, Ritchie JM, Yankowitz J et al, 2004, Human papillomavirus prevalence and types in newborns and parents: concordance and modes of transmission. Sex Transm Dis 31:57–62 Smith JL, Campos SK, Ozbun MA, 2007, Human papillomavirus type 31 uses a caveolin 1- and dynamin 2-mediated entry pathway for infection of human keratinocytes. J Virol 81:9922–9931 Straub I, 1996, Experiences of the introduction of country-wide programme for the prevention of hepatitis B virus infection of newborns., in Hungarian). Epinfo 3:225–232 Strauss JH, Wang KS, Schmaljohn AL et al, 1994, Host-cell receptors for Sindbis virus. Arch Virol Suppl 9:473–484 Summerford C, 1998, Samulski RJ: Membrane-associated heparan sulfate proteoglycan is a receptor for adeno-associated virus type 2 virions. J. Virol. 72:1438–1445 Summerford C, Bartlett JS, Samulski RJ, 1999, aVb5 integrin: a co-receptor for adeno-associated virus type 2 infection. Nat Med 5:78–82 Sverdlov M, Shajahan AN, Minshall RD, 2007, Tyrosine phosphorylation-dependence of caveolae-mediated endocytosis. J Cell Molec Med 11:1239–1250 Tabata T, McDonagh S, Kawakatsu H, Pereira L, 2007, Cytotrophoblasts infected with a pathogenic human cytomegalovirus strain dysregulate cell-matrix and cell-cell adhesion molecules: a quantitative analysis. Placenta 28:527–537 Tagawa A, Mezzacasa A, Hayer A et al, 2005, Assembly and trafficking of caveolar domains in the cell: caveolae as stable, cargo-triggered, vesicular transporters. J Cell Biol 170:769–779 Takizawa T, Anderson CL, Robinson JM, 2005, A novel Fc gamma R-defined, IgG-containing organelle in placental endothelium. J Immunol 175:2331–2339 Tarantal AF, Lee CI, Ekert JE et al, 2001, Lentiviral vector gene transfer into fetal rhesus monkeys, Macaca mulatta): lungtargeting approaches. Mol Ther 4:614–621 Taylor MM, Chohan B, Lavreys L et al, 2004, Shedding of human herpesvirus 8 in oral and genital secretions from HIV-1-seropositive and -seronegative Kenyan women. J Infect Dis 190:484–488 Thomas R, Macsween KF, McAulay K et al, 2006, Detection of human herpesvirus 8, HHV-8, in normal prostates. Prostate 66:1302–1310 Thomas R, Macsween KF, McAulay K et al, 2006, Evidence of shared Epstein-Barr viral isolates between sexual partners, and low level EBV in genital secretions. J Med Virol 78:1204–1209 Tong MI, Thursby M, Rakela J et al, 1981, Studies, on the maternal-infant transmission of the viruses which cause acute hepatitis. Gastroenterol 80:999–1004 Toth FD, Aboagye-Mathiesen G, Szabo J et al, 1995, Bidirectional enhancing activities between human T cell leukemialymphoma virus type I and human cytomegalovirus in human term syncytiotrophoblast cells cultured in vitro. AIDS Res Hum Retrov 11:1495–1507 Toth FD, Aboagye-Mathiesen G, Nemes J et al, 1997, Epstein- Barr virus permissively infects human syncytiotrophoblasts in vitro and induces replication of human T cell leukemialymphoma virus type I in dually infected cells. Virology 229:400–414 Tousseyn T, Jorissen E, Reiss K, Hartmann D, 2006,, Make, stick and cut loose-disintegrin metalloproteases in development and disease. Birth Defects Res C Embryo Today 78:24–46 van Ijzendoorn SC, Tuvim MJ, Weimbs T et al, 2002, Direct interaction between Rab3b and the polymeric immunoglobulin receptor controls ligand-stimulated transcytosis in epithelial cells. Dev Cell 2:219–228 Veettil MV, Sharma-Walia N, Sadagopan S et al, 2006, RhoAGTPase facilitates entry of Kaposi's sarcoma-associated herpesvirus, KSHV/HHV-8, into adherent target cells in a Src-dependent manner. J Virol 80:11432–11446 Vidricaire G, Imbeault M, Tremblay MJ, 2004, Endocytic host cell machinery plays a dominant role in intracellular trafficking of incoming human immunodeficiency virus type 1 in human placental trophoblasts. J Virol 78:11904–11915 Vidricaire G, Tremblay MJ, 2005, Rab5 and Rab7, but not ARF6, govern the early events of HIV-1 infection in polarized human placental cells. J Immunol 175:6517–6530 Vidricaire G, Tremblay MJ, 2007, A clathrin, caveolae, and dynamin–independent endocytic pathway requiring free membrane cholesterol drives HIV-1 internalization and infection in polarized trophoblastic cells. J Mol Biol 368:1267–1283 Ward KN, 2005, Human herpesviruses-6 and -7 infections. Curr Opin Infect Dis 18:247–252 Ward KN, Leong HN, Nacheva EP et al, 2006, Human herpesvirus 6 chromosomal integration in immunocompetent patients results in high levels of viral DNA in blood, sera, and hair follicles. J Clin Microbiol 44:1571–1574 Ward KN, 2005, The natural history and laboratory diagnosis of human herpesviruses-6 and -7 infections in the immunocompetent. J Clin Virol 32:183–193 Ward KN, Leong HN, Thiruchelvam AD et al, 2007, Human herpesvirus 6 DNA levels in cerebrospinal fluid due to primary infection differ from those due to chromosomal viral integration and have implications for diagnosis of encephalitis. J Clin Microbiol 45:1298–1304 Warrington RJ, Wong SK, Ramdahin S, Rutherford WJ, 1995, Normal human cord blood B cells can produce high affinity IgG antibodies to dsDNA that are recognized by cord blood-derived anti-idiotypic antibodies. Scand J Immunol 42:397–406 Wegner CC, Jordan JA, 2004, Human parvovirus B19 VP2 empty capsids bind to human villous trophoblast cells in vitro via the globoside receptor. Infect Dis Obstet Gynecol 12:69–78 Wei JN, Wang SP, Shuang JY, 2005, Study on the relationship between fetomaternal cellular traffic and hepatitis B virus intrauterine infection, in Chinese). Zhonghua Liu Xing Bing Xue Za Zhi 26:240–244 Weigel-Kelley KA, Yoder C, Srivastava A, 2001, Recombinant human parvovirus B19 vectors: erythrocyte P antigen is necessary but not sufficient for successful transduction of human hematopoietic cells. J Virol 75:4110–4116 Wolff H, Politch JA, Martinez A et al, 1990, Leukocytospermia is associated with poor semen quality. Fertil Steril 53:528–536 Wolff H, Panhans A, Stolz W, Meurer M, 1993, Adherence of Escherichia coli to sperm: a mannose mediated phenomenon leading to agglutination of sperm and E. coli. Fertil Steril 60:154–158 Wolff H, 1995, The biologic significance of white blood cells in semen. Fertil Steril 63:1143–1157 Wu X, Li DJ, Yuan MM, Zhu Y, Wang MY, 2004, The expression of CXCR4/CXCL12 in first–trimester human trophoblast cells. Biol Reprod 70:1877–1885 Wang X-S, Zhang J-W, 2008, The microRNAs involved in human myeloid differentiation and myelogenous/myeloblastic leukemia. J Cell Mol Med 12:1445–1455 Yang YS, Ho HN, Chen HF et al, 1995, Cytomegalovirus infection and viral shedding in the genital tract of infertile couples. J Med Virol 45:179–182 Yang Y, Zou L, LiM, Zhao Y, 2006, CXCL12/CXCR4 expression in trophoblasts takes part in materno-fetal immune tolerance and vascular remodeling. J Huazhong Univ Sci Technolog Med Sci 26:466–468 You H, Liu Y, Agrawal N et al, 2007, Multiple human papillomavirus types replicate in 3A Trophoblasts. Placenta 29:30–38 Younes SA, Csire M, Palyi B et al, 2007, Endotoxins do not influence transplacental transmission of lymphotropic human herpesviruses and human papillomaviruses into amniotic fluid taken from healthy mothers before parturition in Hungary. Acta Microbiol Immunol Hung 54:279–303 Yuen MF, Fung SK, Tanaka Y et al, 2004, Longitudinal study of hepatitis activity and viral replication before and after HBeAg seroconversion in chronic hepatitis B patients infected with genotype B and C. J Clin Microbiol 42:5036–5040 Zacchi P, Stenmark H, Parton RG et al, 1998, Rab17 regulates membrane trafficking through apical recycling endosomes in polarized epithelial cells. J Cell Biol 140:1039–1053 Zachar V, Spire B, Hirsch I et al, 1991, Human transformed trophoblast-derived cells lacking CD4 receptor exhibit restricted permissiveness for human immunodeficiency virus type 1. J Virol 65:2102–2107 Zanghellini F, Boppana SB, Emery VC et al, 1999, Asymptomatic primary cytomegalovirus infection: virologic and immunologic features. J Infect Dis 180:702–707 Zerial M, McBride H, 2001, Rab proteins as membrane organizers. Nat Rev Mol Cell Biol 2:107–117 Zerr DM, Meier AS, Selke SS et al, 2005, A population–based study of primary human herpesvirus 6 infection. N Engl J Med 352:753–755 Zhao JJ, Hua YJ, Sun DG et al, 2006, Genome-wide microRNA profiling in human fetal nervous tissues by oligonucleotide microarray. Childs Nerv Syst 22:1419–1425 Ziyaeyan M, Alborzi A, Abbasian A et al, 2007, Detection of HCMV DNA in placenta, amniotic fluid and fetuses of seropositive women by nested PCR. Eur J Pediatr 166:723–726 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 451 EP 465 DI 10.1007/s12253-009-9166-9 PG 15 ER PT J AU Kabukcuoglu, F Kabukcuoglu, Y Tanik, C Sakiz, D Karsidag, S AF Kabukcuoglu, Fevziye Kabukcuoglu, Yavuz Tanik, Canan Sakiz, Damlanur Karsidag, Semra TI Breast Carcinoma Metastasis in Recurrent Myxoid Liposarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Beta-catenin; Breast cancer; E-cadherin; Liposarcoma; Tumor to tumor metastasis ID Beta-catenin; Breast cancer; E-cadherin; Liposarcoma; Tumor to tumor metastasis AB Tumor to tumor metastasis is a rare, but well recognized entity, most commonly involving a carcinoma metastasis to a benign or low grade mesenchymal tumor. A case of breast carcinoma metastasis in a recurrent myxoid liposarcoma is presented in this study. A 52-year-old female patient with a history of breast carcinoma (70% invasive lobular carcinoma and 30% invasive ductal carcinoma) presented with a mass in the right lumbar region. The excised mass was diagnosed as myxoid liposarcoma. The tumor recurred twice and was reexcised. Microscopic examination of the second recurrence revealed multiple foci of breast carcinoma metastases in myxoid liposarcoma. Immunohistochemical study showed staining for CK19, GCDFP-15, estrogen and progesterone in metastases. Both breast carcinoma metastasis and myxoid liposarcoma were immunoreactive for E-cadherin and beta-catenin. To our knowledge, this is the first reported case of breast carcinoma metastasis in myxoid liposarcoma, and the first occurrence of metastasis in a liposarcoma. C1 [Kabukcuoglu, Fevziye] Sisli Etfal Training and Research Hospital, Department of Pathology, M. Ismail Hakki Sk. No:10/1 Uskudar, 34672 Istanbul, Turkey. [Kabukcuoglu, Yavuz] Taksim Training and Research Hospital, Department of Orthopedics and TraumatologyIstanbul, Turkey. [Tanik, Canan] Sisli Etfal Training and Research Hospital, Department of Pathology, M. Ismail Hakki Sk. No:10/1 Uskudar, 34672 Istanbul, Turkey. [Sakiz, Damlanur] Sisli Etfal Training and Research Hospital, Department of Pathology, M. Ismail Hakki Sk. No:10/1 Uskudar, 34672 Istanbul, Turkey. [Karsidag, Semra] Sisli Etfal Training and Research Hospital, Department of Plastic Surgery and ReconstructionIstanbul, Turkey. RP Kabukcuoglu, F (reprint author), Sisli Etfal Training and Research Hospital, Department of Pathology, 34672 Istanbul, Turkey. EM fkabukcuoglu@hotmail.com CR Campbell LV, Gilbert E, Chamberlain CR et al, 1968, Metastases of cancer to cancer. Cancer 22:635–643 Gore I, Barr R, 1957, Metastasis of cancer to cancer. Arch Pathol 66:293–298 Grignon DJ, Ro JY, Ayala AG, 1989, Malignant melanoma with metastasis to adenocarcinoma of the prostate. Cancer 63:196–198 Cohn ML, Elliot DD, El-Naggar AK, 2004, Metastatic acinic cell carcinoma in neurofibroma mistaken for carcinosarcoma. Head Neck 27:76–80 Perry LJ, Lewis JC, Ball RY, 1996, Adenocarcinoma of the breast metastatic to benign ovarian fibroma. Gynecol Oncol 62:408–410 Uner A, Tiras MB, Kilic D et al, 2003, Uterine lipoleiomyoma containing metastatic breast carcinoma: a case with two unusual pathologies. Eur J Obstet Gynecol Reprod Biol 106:76–78 Fadare O, Parkash V, Fiedler PN, Mayerson AB, Asiyanbola B, 2005, Tumor-to-tumor metastasis to a thyroid follicular adenoma as the initial presentation of a colonic adenocarcinoma. Pathol Int 55:574–579 Bhargava P, McGrail KM, Manz HJ, 1999, Lung carcinoma presenting as metastasis to intracranial meningioma: case report and review of the literature. Am J Clin Oncol 22:199–202 Wong A, Koszyca B, Blumbergs PC et al, 1999, Malignant melanoma metastatic to meningioma. Pathology 31:162–165 Hamazaki S, Nakashima H, Matsumoto K et al, 2001, Metastasis of renal cell carcinoma to central nervous system hemangioblastoma in two patients with von Hippel-Lindau disease. Pathol Int 51:948–953 Baloch ZW, LiVolsi VA, 1999, Tumor-to-tumor metastasis to follicular variant of papillary carcinoma of thyroid. Arch Pathol Lab Med 123:703–706 Petraki C, Vaslamatzis M, Argyrakos T et al, 2003, Tumor to tumor metastasis: report of two cases and a review of the literature. Int Surg Pathol 11:127–135 Woodhouse EC, Chuaqui RF, Liotta LA, 1997, General mechanisms of metastasis. Cancer 80:1529–1537 Thompson EW, Newgreen DF, 2005, Carcinoma invasion and metastasis: a role for epithelial–mesenchymal transition. Cancer Res 65:5991–5995 Sugarbaker EV, 1979, Some characteristics of metastasis in man. Am J Pathol 97:623–632 Chambers PW, Davis RL, Blanding JD, 1980, Metastases to primary intracranial meningiomas and neurilemmomas. Arch Pathol Lab Med 104:350–354 Schmitt HP, 1984, Metastases of malignant neoplasms to intracranial tumors: the “tumor-in-a-tumor” phenomenon. Virchows Arch A Pathol Anat Histopathol 405:155–160 Aghi M, Kiehl TR, Brisman JL, 2005, Breast adenocarcinoma metastatic to epidural cervical spine meningioma: case report and review of the literature. J Neurooncol 75:149–155 Watanabe T, Fujisawa H, Hasegawa M, 2002, Metastasis of breast cancer to intracranial meningioma. Am J Clin Oncol 25:414–417 Nakajima S, Doi R, Toyoda E, 2004, N-cadherin expression and epithelial-mesenchymal transition in pancreatic carcinoma. Clin Cancer Res 10:4125–4133 Sakamoto A, Oda Y, Adachi T, 2002, Beta-catenin accumulation and gene mutation in exon 3 in dedifferentiated liposarcoma and malignant fibrous histiocytoma. Arch Pathol Lab Med 126:1071– 1078 Azoulay S, Adem C, Pelletier FL, 2005, Skin metastases from unknown origin: role of immunohistochemistry in the evaluation of cutaneous metastases of carcinoma of unknown origin. J Cutan Pathol 32:561–566 Weiss SW, Goldblum JR, 2001, Liposarcoma. In: Enzinger FM, Weiss SW, eds, Soft tissue tumors. 4th edn. Mosby, St Louis ten Heuvel SE, Hoeckstra HJ, Ginkel RJ, 2007, Clinicopathologic prognostic factors in myxoid liposarcoma: a retrospective study of 49 patients with long-term follow-up. Ann Surg Oncol 14:222– 229 Smith TA, Easly KA, Goldblum JR, 1996, Myxoid/ round cell liposarcoma of the extremities. A clinicopathologic study of 29 cases with particular attention to extent of a round cell liposarcoma. Am J Surg Pathol 20:171–180 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 467 EP 471 DI 10.1007/s12253-008-9121-1 PG 5 ER PT J AU Yuan, W Chen, Z Wu, Sh Ge, J Chang, Sh Wang, X Chen, J Chen, Z AF Yuan, Weijie Chen, Zhikang Wu, Shaobin Ge, Jie Chang, Shi Wang, Xianwei Chen, Jingxiang Chen, Zihua TI Expression of EphA2 and E-cadherin in Gastric Cancer: Correlated with Tumor Progression and Lymphogenous Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EphA2; E-cadherin; Gastric cancer; Immunohistochemistry ID EphA2; E-cadherin; Gastric cancer; Immunohistochemistry AB In this study, gastric cancer progression was correlated with the over-expression of erythropoietin-producing hepatocellular (Eph)A2 receptor and down-expression of epithelial cadherin (E-cadherin). Immunohistochemistry of EphA2 and E-cadherin were performed on these tumor samples from 165 primary lesions of gastric cancer. The results showed that expression of EphA2 was obviously increased in gastric cancer tissues (P<0.01), which was positively correlated with the depth of cancer invasion, tumor-node-metastasis (TNM) stage and lymph node metastasis (P<0.05). Meanwhile, the expression of E-cadherin was significantly reduced (P<0.01), which was negatively correlated with the depth of cancer invasion, grade of tumor differentiation, TNM stage and lymph node metas tasis (P<0.05). The correlation between EphA2 and Ecadherin expression was negative (r=−0.198, P=0.011). In conclusion, either the over-expression of EphA2 or the down-expression of E-cadherin is correlated with cancer progression and lymphogenous metastasis in gastric cancer, suggesting that both of them may play an important role in tumor progression and metastasis. C1 [Yuan, Weijie] Central South University, Xiangya Hospital, Department of General Surgery, Xiangya Road, 410008 Changsha, Hunan Province, China. [Chen, Zhikang] Central South University, Xiangya Hospital, Department of General Surgery, Xiangya Road, 410008 Changsha, Hunan Province, China. [Wu, Shaobin] Central South University, Xiangya Hospital, Department of General Surgery, Xiangya Road, 410008 Changsha, Hunan Province, China. [Ge, Jie] Central South University, Xiangya Hospital, Department of General Surgery, Xiangya Road, 410008 Changsha, Hunan Province, China. [Chang, Shi] Central South University, Xiangya Hospital, Department of General Surgery, Xiangya Road, 410008 Changsha, Hunan Province, China. [Wang, Xianwei] Central South University, Xiangya Hospital, Department of General Surgery, Xiangya Road, 410008 Changsha, Hunan Province, China. [Chen, Jingxiang] Central South University, Xiangya Hospital, Department of General Surgery, Xiangya Road, 410008 Changsha, Hunan Province, China. [Chen, Zihua] Central South University, Xiangya Hospital, Department of General Surgery, Xiangya Road, 410008 Changsha, Hunan Province, China. RP Chen, Z (reprint author), Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, China. EM chenzihua1@hotmail.com CR Catalano V, Labianca R, Beretta GD et al, 2005, Gastric cancer. Crit Rev Oncol Hematol 54:209–241 Dicken BJ, Bigam DL, Cass C et al, 2005, Gastric adenocarcinoma: review and considerations for future directions. Ann Surg 241:27–39 Sulman EP, Tang XX, Allen C et al, 1997, ECK, a human EPHrelated gene, maps to 1p36.1, a common region of alteration in human cancers. Genomics 40:371–374 Walker-Daniels J, Hess AR, Hendrix MJC et al, 2003, Differential regulation of EphA2 in normal and malignant cells. Am J Pathol 162:1037–1042 Miyazaki T, Kato H, Fukuchi M et al, 2003, EphA2 overexpression correlates with poor prognosis in esophageal squamous cell carcinoma. Int J Cancer 103:657–663 Zeng G, Hu Z, Kinch MS et al, 2003, High-level expression of EphA2 receptor tyrosine kinase in prostatic intraepithelial neoplasia. Am J Pathol 163:2271–2276 Kinch MS, Moore MB, Harpole DH et al, 2003, Predictive value of the EphA2 receptor tyrosine kinase in lung cancer recurrence and survival. Clin Cancer Res 9:613–618 Han L, Dong Z, Qiao Y et al, 2005, The clinical significance of EphA2 and Ephrin A-1 in epithelial ovarian carcinomas. Gynecol Oncol 99:278–286 Abraham S, Knapp DW, Cheng L et al, 2006, Expression of EphA2 and Ephrin A-1 in carcinoma of the urinary bladder. Clin Cancer Res 12:353–360 Bussemakers MJ, van Bokhoven A, Mees SG et al, 1993, Molecular cloning and characterization of the human E-cadherin cDNA. Mol Biol Rep 17:123–128 Takeichi M, 1993, Cadherins in cancer: implications for invasion and metastasis. Curr Opin Cell Biol 5:806–811 Chen H, Paradies NE, Fedor-Chaiken M et al, 1997, E-cadherin mediates adhesion and suppresses cell motility via distinct mechanisms. J Cell Sci 110:345–356 Hazan RB, Qiao R, Keren R et al, 2004, Cadherin switch in tumor progression. Ann NY Acad Sci 1014:155–163 Zantek ND, Azimi M, Fedor-Chaiken M et al, 1999, E-cadherin regulates the function of the EphA2 receptor tyrosine kinase. Cell Growth Differ 10:629–638 Thaker PH, Deavers M, Celestino J et al, 2004, EphA2 expression is associated with aggressive features in ovarian carcinoma. Clin Cancer Res 10:5145–5150 Lin YG, Han LY, Kamat AA et al, 2007, EphA2 overexpression is associated with angiogenesis in ovarian cancer. Cancer 109:332–340 Shiozaki H, Tahara H, Oka H et al, 1991, Expression of immunoreactive E-cadherin adhesion molecule in human cancer. Am J Pathol 139:17–23 Jawhari A, Jordan S, Poole S et al, 1997, Abnormal immunoreactivity of the E-cadherin-catenin complex in gastric carcinoma: relationship with patient survival. Gastroenterology 112:46–54 Zhou Y, Ran J, Tang C, 2007, Effect of celecoxib on E-cadherin, VEGF, Microvessel density and apoptosis in gastric cancer. Cancer Biol Ther 6:269–275 Sobin LH, Wittekind CH, eds,, 1997, TNM Classification of Malignant Tumors, 5th edn. Wiley, New York Ogawa K, Pasqualini R, Lindberg RA et al, 2000, The ephrin-A1 ligand and its receptor, EphA2, are expressed during tumor neovascularization. Oncogene 19:6043–6052 Zelinski DP, Zantek ND, Stewart JC et al, 2001, EphA2 overexpression causes tumorigenesis of mammary epithelial cells. Cancer Res 61:2301–2306 Fang WB, Brantley-Sieders DM, Parker MA et al, 2005, A kinase-dependent role for EphA2 receptor in promoting tumor growth and metastasis. Oncogene 24:7859–7868 Lu C, Shahzad MM, Wang H et al, 2008, EphA2 overexpression promotes ovarian cancer growth. Cancer Biol Ther 7:1–6 Zantek ND, Walker-Daniels J, Stewart J et al, 2001, MCF-10ANeoST: a new cell system for studying cell-ECM and cell-cell interactions in breast cancer. Clin Cancer Res 7:3640–3648 Kinch MS, Carles-Kinch K, 2003, Overexpression and functional alterations of the EphA2 tyrosine kinase in cancer. Clin Exp Metastasis 20:59–68 Kikawa KD, Vidale DR, Van Etten RL et al, 2002, Regulation of the EphA2 kinase by the low molecular weight tyrosine phosphatase induces transformation. J Biol Chem 277:39274– 39279 Parri M, Buricchi F, Taddei ML et al, 2005, EphrinA1 repulsive response is regulated by an EphA2 tyrosine phosphatase. J Biol Chem 280:34008–34018 Chen HC, Chu RY, Hsu PN et al, 2003, Loss of E-cadherin expression correlates with poor differentiation and invasion into adjacent organs in gastric adenocarcinomas. Cancer Lett 201:97– 106 Lee KH, Shin SJ, Kim KO et al, 2006, Relationship between E-cadherin, matrix metalloproteinase-7 gene expression and clinicopathological features in gastric carcinoma. Oncol Rep 16:823–830 Guilford PJ, Hopkins JB, Grady WM et al, 1999, E-cadherin germline mutations define an inherited cancer syndrome dominated by diffuse gastric cancer. Hum Mutat 14:249–255 Tamura G, Yin J, Wang S et al, 2000, E-Cadherin gene promoter hypermethylation in primary human gastric carcinomas. J Natl Cancer Inst 92:569–573 Koizume S, Tachibana K, Sekiya T et al, 2002, Heterogeneity in the modification and involvement of chromatin components of the CpG island of the silenced human CDH1 gene in cancer cells. Nucleic Acids Res 30:4770–4780 Catimel B, Layton M, Church N et al, 2006, In situ phosphorylation of immobilized receptors on biosensor surfaces: application to E-cadherin/beta-catenin interactions. Anal Biochem 357:277–288 Noe V, Fingleton B, Jacobs K et al, 2001, Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci 114:111–118 Rios-Doria J, Day KC, Kuefer R et al, 2003, The role of calpain in the proteolytic cleavage of E-cadherin in prostate and mammary epithelial cells. J Biol Chem 278:1372–1379 Saito T, Masuda N, Miyazaki T et al, 2004, Expression of EphA2 and E-cadherin in colorectal cancer: correlation with cancer metastasis. Oncol Rep 11:605–611 Orsulic S, Kemler R, 2000, Expression of Eph receptors and ephrins is differentially regulated by E-cadherin. J Cell Sci 113:1793–1802 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 473 EP 478 DI 10.1007/s12253-008-9132-y PG 6 ER PT J AU Kiss, LA Botos, E AF Kiss, L Anna Botos, Erzsebet TI Ocadaic Acid Retains Caveolae in Multicaveolar Clusters SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Caveola; Alternative endocytosis; Caveosomes; Ocadaic acid ID Caveola; Alternative endocytosis; Caveosomes; Ocadaic acid AB Caveola-mediated endocytosis exists parallel to other forms of endocytosis. Being ligand-triggered, caveolar endocytosis provides a more selective and highly regulated way for uptake of specified substances. Internalized caveolae accumulate in intermediate organelles called caveosomes. It is still debated whether caveosomes are independent organelles or the downstream caveosomes interact with the classical endocytotic compartments. In our work caveola internalization was stimulated with a serine/threonine phosphatase (PP1 and PP2A) inhibitor (ocadaic acid—OA). To find out whether caveolar clusters are really independent organelles or they are still connected to the cell surface we used an electron dense surface marker, ruthenium red (Ru red). Since we were especially interested in the fate of caveolar clusters, the cells were treated with OA for longer time. Stimulating caveola-mediated endocytosis, OA treatment resulted in a significant increase in the number of caveolar cluster. Most of these clusters were found Ru red positive indicating that they were still conneted to the cell surface. Our double labeling experiments on ultrathin frozen sections clearly showed that in OA-treated cells caveolae are not transported to late endosomes instead they are accumulted in large multicaveolar clusters. We think that PP2A can be one of the key components to regulate the fusion of various endocytotic compartments and/or the trafficking along the microtubules. C1 [Kiss, L Anna] Semmelweis University, 1st Department of Anatomy, Tuzolto u. 58, 1094 Budapest, Hungary. [Botos, Erzsebet] Semmelweis University, 1st Department of Anatomy, Tuzolto u. 58, 1094 Budapest, Hungary. RP Kiss, LA (reprint author), Semmelweis University, 1st Department of Anatomy, 1094 Budapest, Hungary. EM KissA@ana2.sote.hu CR Sargiacomo M, Sudol M, Tang A, Lisanti MP, 1993, Signal transducing molecules and GP1- linked proteins form a caveolinrich insoluble complex in MDCK cells. J Cell Biol 122:789–807 Murata M, Peranen J, Schreiner R, Wieland F, Kurchalia TV, Simons K, 1995, VIP 21/caveolin is a cholesterol-binding protein. Proc Natl Acad Sci USA 92:10339–10343 Harder T, Simons K, 1997, Caveolae, DIGs and the dynamics of sphigolipid-cholesterol microdomains. Curr Opin Cell Biol 9:534–542 Smart EJ, Graf GA, Mc Nives MA, Engelman JA, Scherer PE, Okamoto T, Lisanti MP, 1999, Caveolins, liquid-ordered domains, and signal transduction. Mol Cell Biol 19:7289–7304 Scherer PE, Lewis RY, Volonte D, Engelman JA, Galbiati F, Couet J, Kohtz DS, van Donselaar E, Peters P, Lisanti MP, 1997, Cell-type and tissue-specific expression of caveolin-2. Caveolins 1 and 2 co-localize and form a stable hetero-oligomeric complex in vivo. J Biol Chem 272:29337–29346 Mora R, Bouilha VL, Marmorstein A, Scherer PE, Brown D, Lisanti MP, Rodriguez-Boulan VL, 1999, Caveolin-2 localizes to the Golgi complex but redistributes to plasma membrane, caveolae and rafts when co-expressed with caveolin-1. J Biol Chem 274:25708–25717 Song KS, Scherer PE, Tang Z, Okamoto T, Li S, Chafel M, Chu C, Kohtz DS, Lisanti MP, 1996, Expression of caveolin-3 in skeletal, cardiac, and smooth muscle cells. Caveolin-3 is a component of the sarcolemma and co-fractionates with dystrophin and dystrophinassociated glycoproteins. J Biol Chem 271:15160–15165 Nishyama K, Trapp BB, Ikezu T, Ranshoff RM, Tomita T, Iwatsubo T, Kanazawa I, Hsiao KK, Lisanti MP, Okamoto T, 1999, Caveolin-3 upregulaton activates B-secterase-mediated cleavage of amiloid precursore protein in Alzheimer’s disease. J Neurosci 19:6538–6548 Schwab W, Galbiati F, Volonte D, Hempel U, Wenzel KW, Funk RHW, Lisanti MP, Kasper M, 1999, Characterization of caveolins from cartilage: expression of caveolin-1, -2 and -3 in chondrocytes and in alginate cell culture of the rat tibia. Histochem Cell Biol 112:41–49 Rothberg KG, Heuser JE, Donzell WC, Ying YS, Glenney JR, Anderson RG, 1992, Caveolin, a protein component of caveolae membrane coats. Cell 68:673–682 Anderson RG, Kamen BA, Rothberg KG, Lacey SW, 1992, Potocytosis: sequestration and transport of small molecules by caveolae. Science 255:410–411 Schnitzer JE, Oh P, Pinney E, Allard J, 1994, Filipin-sensitive caveolae-mediated transport in endothelium: reduced transcytosis, scavenger endocytosis and cappilary permeability of select macromolecules. J Cell Biol 127:1217–1232 Parton RG, Joggerst B, Simons K, 1994, Regulated internalization of caveolae. J Cell Biol 127:1199–1215 Benlimame N, Le PU, Nabi IR, 1998, Localization of autocrine motility factor receptor to caveolae and clathrin—independent internalization of its ligand to smooth endoplasmic reticulum. Mol Biol Cell 9:1773–1786 Weiling D, Hope JC, Chaplin P, Collins RA, Taylor G, Howard CJ, 1999, Involvement of caveolae in the uptake of respiratory syntitial virus antigen by dendritic cells. J Leukoc Biol 66:50–58 Shin JS, Gao Z, Abraham SN, 2000, Involvement of cellular caveolae in bacterial entry into mast cells. Science 289:785–788 Puri V, Watanabe R, Singh RD, Dominguez M, Brown JC, Wheatley CL, Marks DL, Pagano RE, 2001, Clathrin-dependent and independent internalization of plasma membrane sphingolipids initiates two Golgi targeting pathways. J Cell Biol 154:535–547 Pelkmans L, Kartenbeck J, Helenius A, 2001, Caveolae endocytosis of simian virus 40 reveals a new two-step reticular transport pathway to the ER. Nat Cell Biol 3:473–483 Richterova Z, Liebe D, Horak M, Palkova Z, Stokrova J, Hozak P, Korb J, Forstova J, 2001, Caveolae are involved in the traffiching of mouse polyoma virus virion and artificial VP1 pseudocapsids toward cell nuclei. J Virol 75:0880–10891 Campbell SM, Crowe SM, Mark J, 2001, Lipid rafts and HIV-1 from viral entry to assembly of progeny virion. J Clin Virol 22:217–227 Pelkmans L, Helenius A, 2002, Endocytosis via caveolae. Traffic 3:311–320 Pietiainen V, Marjomaki V, Upla P, Pelkmans L, Helenius A, Hyypia T, 2004, Echovirus 1 endocytosis into caveosomes requires lipid rafts, dynamin and signaling events. Mol Biol Cell 15:4911–4925 Glenney JR Jr, 1989, Tyrosine phosphorylation of a 22 kDa protein is correlated with transformation by Rous sarcoma virus. J Biol Chem 264:20163–20166 Li S, Seitz R, Lisanti MP, 1996, Phophorylation of caveolin by Src tyrosine kinases. The α-isoform of caveolin is selectively phosphorylated by vSrc in vivo. J Biol Chem 271:3863–3868 Tiruppathi C, Song W, Bergenfeldt M, Sass P, Malik AB, 1997, Gp60 activation mediates albumin transcytosis in endothelial cells by a tyrosine kinase-dependent pathway. J Biol Chem 272:25968– 25975 Lee H, Park DS, Wang XB, Scherer PE, Schwartz PE, Lisanti MP, 2002, Src-induced phosphorylation of caveolin-2 on tyrosin 19. J Biol Chem 277:4556–34567 Kiss AL, Botos E, Turi A, Mullner N, 2004, Ocadaic acid treatment causes phosphorylation of caveolin-2 and induces internalization of caveolae in rat peritoneal macrophages. Micron 35:707–715 Thomsen P, Roepstooff K, Stahlhut M, van Deurs B, 2002, Caveolae are highly immobile plasma membrane microdomains which are not involved in constitutive endocytic trafficking. Mol Biol Cell 13:238–250 Sharma DK, Choudhury A, Singh RD, Wheatley CL, Marks DL, Pagano RE, 2003, Glycosphingolipids internalized via caveolarelated endocytosis rapidly merge with the clathrin pathway in early endosomes and form microdomains for recycling. J Biol Chem 278:7564–7572 Botos E, Turi A, Mullner N, Kovalszky I, Tatrai P, Kiss AL, 2007, Regulatory role of kinases and phospatases on the internalization of caveolae in HepG2 cells. Micron 38:313–320 Fernandez JJ, Candenas ML, Suoto ML, Trujillo MM, Norte M, 2002, Ocadaic acid, useful tool for studying cellular processes. Current Medicinal Chem 9:229–262 Cohen PT, 2002, Protein phosphatase I.-targated in many directions. J Cell Sci 115:241–256 Nicols BJ, 2002, A distinct class of endosome mediates clathrin independent endocytosis to the Golgi complex. Nat Cell Biol 4:374–378 Weibel ER, Kiseler GS, Scherle WF, 1996, Practical stereoloical method for morphometric cytology. J Cell Biol 30:23–38 Richards AA, Stang E, Peppehok R, Parton RG, 2002, Inhibitors of COP-mediated transport and Cholera toxin action inhibit simian virus 40 infection. Mol Biol Cell 13:1750–1764 Kartenbeck J, Stukenbrok H, Helenius A, 1989, Endocytosis of simian virus 40 into endoplasmic reticulum. J Cell Biol 109:2721–2729 Parton RG, Richard AA, 2003, Lipid rafts and caveolae as portals for endocytosis: new insights and common mechanisms. Traffic 4:724–738 Botos E, Klumperman J, Oorschot V, Igyarto B, Magyar A, Olah M, Kiss AL, 2008, Caveolin-1 is transported to multivesicular bodies after albumin-induced endocytosis of caveolae in HepG2 cells. J Cell Mol Med 12:1–9 Molloy SS, Thomas L, Kamibayashi C, Mumby MC, Thomas G, 1998, Regulation of endosome sorting by a specific PP2A isoform. J Cell Biol 142:1399–1411 Price NE, Munby MC, 1999, Brain protein serine/theorine phosphateses. Curr Opin Neurobiol 9:336–342 Sontag E, Nunbhahdi-Craig V, Lee G, Brandt R, Kamibayaski C, Kuret J, White CLIII, Mumby MC, Bloom GS, 1999, Molecular interactions among protein phosphatase 2A, tau and microtubules. J Biol Chem 274:25490–25498 Varlamov O, Kalinina E, Che F-Y, Fricker LD, 2000, Protein phosphatase 2A binds to the cytoplasmic tail of carboxypeptidase D and regulates post-trans-Golgi network trafficking. J Cell Sci 114:311–322 Schapiro F, Soe TT, Mallet G, Maxfield F, 2004, Role of cytoplasmic domain serines in intracellular trafficking of furin. Mol Biol Cell 15:1882–2894 Rundell K, Parakati R, 2001, The role of the SV40ST antigen in cell growth promotion and transformation. Semin Cancer Biol 11:5–13 Mumby DI, Machleidt T, Ying YS, Anderson RG, Bloom GS, 2002, Dual control of caveolae membrane traffic by microtubules and actin cytoskleton. J Cell Sci 115:4327–4339 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 479 EP 486 DI 10.1007/s12253-008-9139-4 PG 8 ER PT J AU Chen, Y Lu, Y Lu, Ch Zhang, L AF Chen, Yongshun Lu, You Lu, Changli Zhang, Lei TI Beclin-1 Expression is a Predictor of Clinical Outcome in Patients with Esophageal Squamous Cell Carcinoma and Correlated to Hypoxia-Inducible Factor (HIF)-1α Expression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal squamous cell carcinoma; Beclin-1; HIF-1α; Immunohistochemistry; Clinicopathological factors; Survival ID Esophageal squamous cell carcinoma; Beclin-1; HIF-1α; Immunohistochemistry; Clinicopathological factors; Survival AB In the present study, we examined the relationship between Beclin-1 expression and HIF-1α expression in esophageal squamous cell carcinoma(ESCC). There was a loss of Beclin-1 protein expression in 33% of ESCCs. Beclin-1 expression significantly correlated with depth of invasion, lymph node metastasis and clinical stage. Among the 54 patients, The survival rate of the Beclin-1-positive group was better than that of the Beclin-1-negative group. Twenty-five of the 54 (46%) tumor specimens showed high levels of HIF-1α immunoreactivity. Beclin-1 expression was associated with HIF-1α expression. The survival rate of patients with Beclin-1-positive and HIF-1α-low tumors was significantly higher than that of the other groups. These results suggest that Beclin-1 and HIF-1α expression are important determinants of survival in ESCCs. C1 [Chen, Yongshun] Sichuan University, West China Hospital, Division of Thoracic Oncology, 37, Guoxue Lane, 610041 Chengdu, China. [Lu, You] Sichuan University, West China Hospital, Division of Thoracic Oncology, 37, Guoxue Lane, 610041 Chengdu, China. [Lu, Changli] Sichuan University, West China Hospital, Department of PathologyChengdu, China. [Zhang, Lei] Sichuan University, West China Hospital, Department of NephrologyChengdu, China. RP Lu, Y (reprint author), Sichuan University, West China Hospital, Division of Thoracic Oncology, 610041 Chengdu, China. EM radyoulu@hotmail.com CR Guimaraes CA, Linden R, 2004, Programmed cell death. Apoptosis and alternative deathstyles. Eur J Biochem 271:1638–1650 Edinger AL, Thompson CB, 2004, Death by design: apoptosis, necrosis and autophagy. Curr Opin Cell Biol 16:663–669 Baehrecke EH, 2005, Autophagy: dual roles in life and death? Nat Rev Mol Cell Biol 6:505–510 Levine B, 2007, Autophagy and cancer. Nat Cell Biol 446:745– 747 Liang XH, Kleeman LK, Jiang HH et al, 1998, Protection against fatal Sindbis virus encephalitis by beclin 1, a novel Bcl-2 interacting protein. J Virol 72:8586–8596 Aita VM, Liang XH, Murty VVet al, 1999, Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics 59:59–65 Yue Z, Jin S, Yang C et al, 2003, Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci USA 100:15077–15082 Liang XH, Jackson S, Seaman M et al, 1999, Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 402:672–676 Ahn CH, Jeong EG, Lee JW et al, 2007, Expression of beclin-1, an autophagy-related protein, in gastric and colorectal cancers. APMIS 115:1344–1349 Koukourakis MI, Giatromanolaki A, Sivridis E et al, 2002, Hypoxia-inducible factor, HIF1α and HIF2α), angiogenesis and chemoradiotherapy outcome of squamous cell head-and-neck cancer. Int J Radiat Oncol Biol Phys 53:1192–1202 Katsuta M, Miyashita M, Makino H et al, 2005, Correlation of hypoxia inducible factor-1α with lymphatic metastasis via vascular endothelial growth factor-C in human esophageal cancer. Exp Mol Pathol 78:123–130 Kimura S, Kitadai Y, Tanaka S et al, 2004, Expression of hypoxia-inducible factor, HIF)-1 alpha is associated with vascular endothelial growth factor expression and tumour angiogenesis in human oesophageal squamous cell carcinoma. Eur J Cancer 40:1904–1912 Enatsu S, Iwasaki A, Shirakusa T et al, 2006, Expression of hypoxia-inducible factor-1 alpha and its prognostic significance in small-sized adenocarcinomas of the lung. Eur J Cardiothorac Surg 29:891–895 Azad MB, Chen Y, Henson ES et al, 2008, Hypoxia induces autophagic cell death in apoptosis-competent cells through a mechanism involving BNIP3. Autophagy 4:195–204 Zhang H, Bosch-Marce M, Shimoda LA et al, 2008, Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J Biol Chem 283:10892–10903 Semenza GL, 2008, Mitochondrial autophagy: life and breath of the cell. Autophagy 4:534–536 Sobin LH, Fleming ID, 1997, TNM classification of malignant tumors, fifth Edition, Union Internationale Contre le Cancer and the American Joint Committee on Cancer. Cancer 80:1803– 1804 Gozuacik D, Kimchi A, 2004, Autophagy as a cell death and tumor suppression mechanism. Oncogene 23:2891–2906 Wang ZH, Xu L, Duan ZL et al, 2007, Beclin 1-mediated macroautophagy involves regulation of caspase-9 expression in cervical cancer HeLa cells. Gynecol Oncol 107:107–113 Miracco C, Cosci E, Oliveri G et al, 2007, Protein and mRNA expression of autophagy gene Beclin 1 in human brain tumours. Int J Oncol 30:429–436 Furuya D, Tsuji N, Yagihashi A et al, 2005, Beclin 1 augmented cis-diamminedichloroplatinum induced apoptosis via enhancing caspase-9 activity. Exp Cell Res 307:26–40 Marx J, 2006, Autophagy: is it cancer’s friend or foe? Science 312:1160–1161 Daniel F, Legrand A, Pessayre D et al, 2006, Partial Beclin 1 silencing aggravates doxorubicin- and Fas-induced apoptosis in HepG2 cells. World J Gastroenterol 12:2895–2900 Koneri K, Goi T, Hirono Yet al, 2007, Beclin 1 gene inhibits tumor growth in colon cancer cell lines. Anticancer-Res 27:1453–1457 Amaravadi RK, Yu D, Lum JJ et al, 2007, Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest 117:326–336 Zhong H, De Marzo AM, Laughner E et al, 1999, Overexpression of hypoxia-inducible factor 1α in common human cancers and their metastases. Cancer Res 59:5830–5835 Birner P, Schindl M, Obermair A et al, 2000, Overexpression of hypoxia-inducible factor 1α is a marker for an unfavorable prognosis in early-stage invasive cervical cancer. Cancer Res 60:4693–4696 Iyer NV, Kotch LE, Agani F et al, 1998, Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1α. Genes Dev 12:149–162 Brahimi-Horn MC, Pouyssegur J, 2007, Harnessing the hypoxiainducible factor in cancer and ischemic disease. Biochem Pharmacol 73:450–457 Bohensky J, Shapiro IM, Leshinsky S et al, 2007, HIF regulation of chondrocyte apoptosis: induction of the autophagic pathway. Autophagy 3:207–214 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 487 EP 493 DI 10.1007/s12253-008-9143-8 PG 7 ER PT J AU Zhang, ZB Cai, L Zheng, ShG Xiong, Y Dong, JH AF Zhang, Zhi-Bo Cai, Lei Zheng, Shu-Guo Xiong, Yan Dong, Jia-Hong TI Overexpression of Caveolin-1 in Hepatocellular Carcinoma with Metastasis and Worse Prognosis: Correlation with Vascular Endothelial Growth Factor, Microvessel Density and Unpaired Artery SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Caveolin-1; Vascular endothelial growth factor; Angiogenesis; Metastasis; Prognosis; Hepatocellular carcinoma; Microvessel density; Unpaired artery ID Caveolin-1; Vascular endothelial growth factor; Angiogenesis; Metastasis; Prognosis; Hepatocellular carcinoma; Microvessel density; Unpaired artery AB Caveolin-1 is the major structural protein in caveolae, implicated in oncogenesis and angiogenesis. The connections between caveolin-1 and progression and angiogenesis of hepatocellular carcinoma (HCC) is still not clear. Thus we investigated the relationship of caveolin-1 expression, vascular endothelial growth factor (VEGF) expression, microvessel density (MVD), and unpaired artery (UA) with the clinicopathologic features of patients with HCC. Formalin-fixed, paraffin-embedded tissue sections of HCC from 75 patients who had undergone an initial hepatectomy were stained immunohistochemically with specific antibodies against caveolin-1, VEGF, CD34 and α-SMA. The levels of caveolin-1, VEGF, MVD and UA were correlated with the clinicopathologic variables, and tissue sections were also analyzed by dual-label immunofluorescence. We found that increased expression of caveolin-1 was associated with metastasis and with a worse prognosis of HCC. Caveolin-1 expression correlates positively with VEGF, MVD and UA. These results suggest that caveolin-1 may play an important role in the progression of HCC and angiogenesis. C1 [Zhang, Zhi-Bo] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, 400038 Chongqing, China. [Cai, Lei] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, 400038 Chongqing, China. [Zheng, Shu-Guo] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, 400038 Chongqing, China. [Xiong, Yan] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, 400038 Chongqing, China. [Dong, Jia-Hong] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, 400038 Chongqing, China. RP Dong, JH (reprint author), Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, 400038 Chongqing, China. EM Jhdong301@163.com CR Okamoto T, Schlegel A, Scherer PE, Lisanti MP, 1998, Caveolins, a family of scaffolding proteins for organizing “preassembled signaling complexes” at the plasma membrane. J Biol Chem 273:5419–5422 Wikman H, Kettunen E, Seppanen JK, Karjalainen A, Hollmen J, Anttila S, Knuutila S, 2002, Identification of differentially expressed genes in pulmonary adenocarcinoma by using cDNA array. Oncogene 21:5804–5813 Wiechen K, Sers C, Agoulnik A, Arlt K, Dietel M, Schlag PM, Schneider U, 2001, Down-regulation of caveolin-1, a candidate tumor suppressor gene, in sarcomas. Am J Pathol 158:833–839 Kato K, Hida Y, Miyamoto M, Hashida H, Shinohara T, Itoh T, Okushiba S, Kondo S, Katoh H, 2002, Overexpression of caveolin-1 in esophageal squamous cell carcinoma correlates with lymph node metastasis and pathologic stage. Cancer 94:929–933 Suzuoki M, Miyamoto M, Kato K, Hiraoka K, Oshikiri T, Nakakubo Y, Fukunaga A, Shichinohe T, Shinohara T, Itoh T, Kondo S, Katoh H, 2002, Impact of caveolin-1 expression on prognosis of pancreatic ductal adenocarcinoma. Br J Cancer 87:1140–1144 Rajjayabun PH, Garg S, Durkan GC, Charlton R, Robinson MC, Mellon JK, 2001, Caveolin-1 expression is associated with highgrade bladder cancer. Urology 58:811–814 Dewever J, Frerart F, Bouzin C, Baudelet C, Ansiaux R, Sonveaux P, Gallez B, Dessy C, Feron O, 2007, Caveolin-1 is critical for the maturation of tumor blood vessels through the regulation of both endothelial tube formation and mural cell recruitment. Am J Pathol 171:1619–1628 Joo HJ, Oh DK, Kim YS, Lee KB, Kim SJ, 2004, Increased expression of caveolin-1 and microvessel density correlates with metastasis and poor prognosis in clear cell renal cell carcinoma. BJU Int 93:291–296 Anders RA, Yerian LM, Tretiakova M, Davison JM, Quigg RJ, Domer PH, Hoberg J, Hart J, 2003, cDNA microarray analysis of macroregenerative and dysplastic nodules in end-stage hepatitis C virus-induced cirrhosis. Am J Pathol 162:991–1000 Hirasawa Y, Arai M, Imazeki F, Tada M, Mikata R, Fukai K, Miyazaki M, Ochiai T, Saisho H, Yokosuka O, 2006, Methylation status of genes upregulated by demethylating agent 5-aza-2′- deoxycytidine in hepatocellular carcinoma. Oncology 71:77–85 Zhou H, Jia L, Wang S, Wang H, Chu H, Hu Y, Cao J, Zhang J, 2006, Divergent expression and roles for caveolin-1 in mouse hepatocarcinoma cell lines with varying invasive ability. Biochem Biophys Res Commun 345:486–494 Choi HN, Kim KR, Park HS, Jang KY, Kang MJ, Lee DG, Kim YK, Cho BH, Cha EJ, Moon WS, 2007, Expression of caveolin in hepatocellular carcinoma: association with unpaired artery formation and radiologic findings. Korean J Hepatol 13:396–408 Park SS, Kim JE, Kim YA, Kim YC, Kim SW, 2005, Caveolin-1 is down-regulated and inversely correlated with HER2 and EGFR expression status in invasive ductal carcinoma of the breast. Histopathology 47:625–630 Tanigawa N, Lu C, Mitsui T, Miura S, 1997, Quantitation of sinusoid-like vessels in hepatocellular carcinoma: its clinical and prognostic significance. Hepatology 26:1216–1223 Glenney JR Jr, 1989, Tyrosine phosphorylation of a 22-kDa protein is correlated with transformation by Rous sarcoma virus. J Biol Chem 264:20163–20166 Williams TM, Medina F, Badano I, Hazan RB, Hutchinson J, Muller WJ, Chopra NG, Scherer PE, Pestell RG, Lisanti MP, 2004, Caveolin-1 gene disruption promotes mammary tumorigenesis and dramatically enhances lung metastasis in vivo. J Biol Chem 279:51630–51646 Yang G, Addai J, Wheeler TM, Frolov A, Miles BJ, Kadmon D, Thompson TC, 2007, Correlative evidence that prostate cancer cell-derived caveolin-1 mediates angiogenesis. Hum Pathol 38:1688–1695 Barresi V, Cerasoli S, Paioli G, Vitarelli E, Giuffre G, Guiducci G, Tuccari G, Barresi G, 2006, Caveolin-1 in meningiomas: expression and clinico-pathological correlations. Acta Neuropathol 112:617–626 Bailey KM, Liu J, 2008, Caveolin-1 up-regulation during epithelial to mesenchymal transition is mediated by focal adhesion kinase. J Biol Chem 283:13914–13724 Barresi V, Cerasoli S, Tuccari G, 2008, Correlative evidence that tumor cell-derived caveolin-1 mediates angiogenesis in meningiomas. Neuropathology 28:472–478 Yerian LM, Anders RA, Tretiakova M, Hart J, 2004, Caveolin and thrombospondin expression during hepatocellular carcinogenesis. Am J Surg Pathol 28:357–364 Mitsuhashi N, Shimizu H, Ohtsuka M, Wakabayashi Y, Ito H, Kimura F, Yoshidome H, Kato A, Nukui Y, Miyazaki M, 2003, Angiopoietines and Tie-2 expression in angiogenesis and proliferation of human hepatocellular carcinoma. Hepatology 37:1105–1113 Morinaga S, Imada T, Shimizu A, Akaike M, Sugimasa Y, Takemiya S, Takanashi Y, 2001, Angiogenesis in hepatocellular carcinoma as evaluated by alpha smooth muscle actin immunohistochemistry. Hepatogastroenterology 48:224–228 Liu J, Wang XB, Park DS, Lisanti MP, 2002, Caveolin-1 expression enhances endothelial capillary tubule formation. J Biol Chem 277:10661–10668 Griffoni C, Spisni E, Santi S, Riccio M, Guarnieri T, Tomasi V, 2000, Knockdown of caveolin-1 by antisense oligonucleotides impairs angiogenesis in vitro and in vivo. Biochem Biophys Res Commun 276:756–761 Podar K, Shringarpure R, Tai YT, Simoncini M, Sattler M, Ishitsuka K, Richardson PG, Hideshima T, Chauhan D, Anderson KC, 2004, Caveolin-1 is required for vascular endothelial growth factor-triggered multiple myeloma cell migration and is targeted by bortezomib. Cancer Res 64:7500–7506 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 495 EP 502 DI 10.1007/s12253-008-9144-7 PG 8 ER PT J AU Stojsic, Z Brasanac, D Stojiljkovic, M Babic, D Randjelovic, T Terzic, T AF Stojsic, Zorica Brasanac, Dimitrije Stojiljkovic, Miodrag Babic, Darko Randjelovic, Tomislav Terzic, Tatjana TI Composite Carcinoma of the Stomach Associated with Sarcoid-Like Granulomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Composite exocrine-endocrine carcinoma; Gastric carcinoma; Immunohistochemistry; Sarcoid-like granuloma; Stomach ID Composite exocrine-endocrine carcinoma; Gastric carcinoma; Immunohistochemistry; Sarcoid-like granuloma; Stomach AB Composite glandular/exocrine-endocrine carcinoma of the gastrointestinal tract is a special tumor type composed of common adenocarcinoma and the neuroendocrine component comprising at least one-third of the whole tumor area. These tumors are rare in the stomach and mostly published as case reports. We describe a further case of a 36-year-old man being unique in that it was associated with extensive formation of sarcoid-like granulomas. Tumor consisted of, predominantly poorly differentiated, intestinal-type adenocarcinoma and poorly differentiated neuroendocrine, small cell carcinoma. The adenocarcinomatous and neuroendocrine areas were separated, but closely juxtaposed with focal areas showing gradual transition from one to another. Perigastric lymph node metastases corresponded either to neuroendocrine or adenocarcinomatous component. On immunohistochemistry, the exocrine part was positive for cytokeratin 7, whereas superficial well-differentiated parts showed positivity with cytokeratin 20 as well. The neuroendocrine component was negative with those two types of cytokeratin. Both adenocarcinomatous and neuroendocrine tumor portions showed carcinoembryonic antigen (CEA) immunoexpression. Neuroendocrine markers (chromogranin A, synaptophysin and neuron-specific enolase) were diffusely positive in the neuroendocrine component, and found only in the scattered cells within the neoplastic glands of the adenocarcinoma. Entire gastric mucosa and all perigastric lymph nodes were extensively affected by noncaseating, sarcoidlike granulomas. The absence of any clinical manifestations combined with the negative results of chest radiograph and laboratory test for the serum angiotensin converting enzyme argued against the possibility of systemic sarcoidosis. C1 [Stojsic, Zorica] University of Belgrade, Faculty of Medicine, Institute for Pathology, Dr Subotica 1/II, 11000 Belgrade, Serbia, Serbia. [Brasanac, Dimitrije] University of Belgrade, Faculty of Medicine, Institute for Pathology, Dr Subotica 1/II, 11000 Belgrade, Serbia, Serbia. [Stojiljkovic, Miodrag] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia, Serbia. [Babic, Darko] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia, Serbia. [Randjelovic, Tomislav] Clinical Centre, Clinical Department of SurgeryBelgrade, Serbia, Serbia. [Terzic, Tatjana] University of Belgrade, Faculty of Medicine, Institute for Pathology, Dr Subotica 1/II, 11000 Belgrade, Serbia, Serbia. RP Stojsic, Z (reprint author), University of Belgrade, Faculty of Medicine, Institute for Pathology, 11000 Belgrade, Serbia. EM zstojsic@med.bg.ac.yu CR Volante M, Rindi G, Papotti M, 2006, The grey zone between pure, neuro, endocrine and non-(neuro)endocrine tumours: a comment on concepts and classification of mixed exocrineendocrine neoplasms. Virchows Arch 449:499–506 Volante M, Righi L, Asioli S et al, 2007, Goblet cell carcinoids and other mixed neuroendocrine/nonneuroendocrine neoplasms. Virchows Arch 451(Suppl 1):S61–S69 Ooi A, Mai M, Ogino T et al, 1988, Endocrine differentiation of gastric adenocarcinoma. The prevalence as evaluated by immunoreactive chromogranin A and its biologic significance. Cancer 62:1096–1104 Lewin K, 1987, Carcinoid tumors and the mixed, composite, glandular-endocrine cell carcinomas. Am J Surg Pathol 11, Suppl 1):71–86 Yang GCH, Rotterdam H, 1991, Mixed, composite, glandularendocrine cell carcinoma of the stomach. Report of a case and review of literature. Am J Surg Pathol 15:592–598 Lewin KJ, Appelman HD, 1996, Tumors of the esophagus and stomach. In: Rosai J, Sobin LH, eds, Atlas of Tumor Pathology 3rd series, Fascicle 18. Armed Forces Institute of Pathology. Washington DC, pp 351–352 Capella C, La Rosa S, Uccella S et al, 2000, Mixed endocrineexocrine tumors of the gastrointestinal tract. Semin Diagn Pathol 17:91–103 Capella C, Solcia E, Sobin LH et al, 2000, Endocrine tumours of the stomach. In: Hamilton SR, Aaltonen LA, eds, World Health Organization Classification of Tumours, Pathology and Genetics of Tumours of the Digestive System. IARC, Lyon, pp 53–57 Matsui K, Jin XM, Kitagawa M et al, 1998, Clinicopathologic features of neuroendocrine carcinomas of the stomach. Appraisal of small cell and large cell variants. Arch Pathol Lab Med 122:1010–1017 Jiang SX, Mikami T, Umezawa A et al, 2006, Gastric large cell neuroendocrine carcinomas: a distinct clinicopathologic entity. Am J Surg Pathol 30:945–953 Fenoglio-Preiser C, Munoz N, Carneiro F et al, 2000, Gastric carcinoma. In: Hamilton SR, Aaltonen LA, eds, World Health Organization Classification of Tumours, Pathology and Genetics of Tumours of the Digestive System. IARC, Lyon, pp 37–68 Remmele W, Leder LD, 1974, Blut und blutbereitende Organe. In: Doerr W, ed, Organpathologie, Band I. Georg Thieme Verlag, Stuttgart, pp 2-1–2-58 Shigematsu H, Kurita A, Omura Y et al, 1999, Gastric cancer with sarcoid reactions in the regional lymph nodes, the stomach wall, and the splenic parenchyma: report of a case. Surg Today 29:549, 552 Furlan D, Cerutti R, Genasetti A et al, 2003, Microallelotyping defines the monoclonal or the polyclonal origin of mixed and collision endocrine-exocrine tumors of the gut. Lab Invest 83:963–971 Jain D, Eslami-Varzaneh F, Takano AM et al, 2005, Composite glandular and endocrine tumors of the stomach with pancreatic acinar differentiation. Am J Surg Pathol 29:1524–1529 Rayhan N, Sano T, Qian ZR et al, 2005, Histological and immunohistochemical study of composite neuroendocrine-exocrine carcinomas of the stomach. J Med Invest 52:191–202 Fujiyoshi Y, Kuhara H, Eimoto T, 2005, Composite glandular- endocrine cell carcinoma of the stomach. Report of two cases with goblet cell carcinoid component. Pathol Res Pract 200:823–829 Lee EJ, Park SM, Maeng L et al, 2005, Composite glandularendocrine cell carcinomas of the stomach: clinicopathologic and methylation study. APMIS 113:569–576 Nugent SL, Cunningham SC, Alexiev BA et al, 2007, Composite signet-ring cell/neuroendocrine carcinoma of the stomach with a metastatic neuroendocrine carcinoma component: a better prognosis entity. Diagn Pathol 2:43 Ronellenfitsch U, Strobel P, Schwarzbach MHM et al, 2007, A composite adenoendocrine carcinoma of the stomach arising from a neuroendocrine tumor. J Gastrointest Surg 11:1573–1575 Fukunaga M, 2002, Gastric carcinoma resembling pancreatic mixed acinar-endocrine carcinoma. Hum Pathol 33:569–573 Brouland JP, Manivet P, Brocheriou-Spelle I et al, 2001, Histological, immunohistochemical, ultrastructural and biochemical study of human gastric composite tumor: expression of the serotonin-2B receptor by the neuroendocrine component. Endocr Pathol 12:77–86 Ali MH, Davidson A, Azzopardi JG, 1984, Composite gastric carcinoid and adenocarcinoma. Histopathology 8:529–536 Caruso RA, Heyman MF, Rigoli L et al, 1998, Composite early carcinoma, ordinary adenocarcinoma, carcinoid, microglandular- goblet cell carcinoid, neuroendocrine mucinous carcinoma, of the stomach. Histopathology 32:568–578 Adhikari D, Conte C, Eskreis D et al, 2002, Combined adenocarcinoma and carcinoid tumor in atrophic gastritis. Ann Clin Lab Sci 32:422–427 Morishita Y, Tanaka T, Kato K et al, 1991, Gastric collision tumor, carcinoid and adenocarcinoma, with gastritis cystica profunda. Arch Pathol Lab Med 115:1006–1010 Nagaoka S, Toyoshima H, Bandoh T et al, 1996, Composite carcinoid-adenocarcinoma tumor of the stomach: report of a case. Surg Today 26:184–188 Kim KM, Kim MJ, Cho BK et al, 2002, Genetic evidence for the multi-step progression of mixed glandular-neuroendocrine gastric carcinomas. Virchows Arch 440:85–93 Kloppel G, Rindi G, Anlauf M et al, 2007, Site-specific biology and pathology of gastroenteropancreatic neuroendocrine tumors. Virchows Arch 451(Suppl 1):S9–S27 Kashiwabara K, Toyonaga M, Yamaguchi Y et al, 2001, Sarcoid reaction in primary tumor of bronchogenic large cell carcinoma accompanied with massive necrosis. Internal Med 40:127–130 Day DW et al, 2003, Inflammatory disorders of the small intestine. In: Morson and Dawson’s Gastrointestinal Pathology. Oxford, Blackwell, pp 272–323 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 503 EP 510 DI 10.1007/s12253-008-9145-6 PG 8 ER PT J AU Tsiambas, E Manaios, L Papanikolopoulos, C Rigopoulos, ND Tsounis, D Karameris, A Soultati, A Koliopoulou, A Kravvaritis, Ch Sergentanis, Th Patsouris, E Dourakis, S AF Tsiambas, Evangelos Manaios, Loukas Papanikolopoulos, Costas Rigopoulos, N Dimitrios Tsounis, Dimitrios Karameris, Andreas Soultati, Aspasia Koliopoulou, Antigoni Kravvaritis, Christos Sergentanis, Theodoros Patsouris, Efstratios Dourakis, Spyridon TI Chromogenic In Situ Hybridization Analysis of Epidermal Growth Factor Receptor Gene/Chromosome 7 Numerical Aberrations in Hepatocellular Carcinoma Based on Tissue Microarrays SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chromogenic in situ hybridization; Epidermal growth factor receptor; Hepatocellular carcinoma; Tissue microarrays ID Chromogenic in situ hybridization; Epidermal growth factor receptor; Hepatocellular carcinoma; Tissue microarrays AB Although Epidermal Growth Factor Receptor (EGFR) overexpression is observed frequently in hepatocellular carcinomas (HCC), specific gene deregulation mechanisms remain unknown. Our aim was to investigate the prognostic significance of the combined protein and gene/chromosome 7 numerical alterations. Using tissue microarray technology, thirty-five (n=35) paraffin embedded histologically confirmed HCCs were cored and reembedded in a paraffin block. Immunohistochemistry was performed for the determination of EGFR protein levels and evaluated by the performance of digital image analysis. Chromogenic in situ hybridization was also performed based on the use of EGFR gene and chromosome 7 centromeric probes, respectively. EGFR overexpression was observed in 26/35 (74.2%) cases and was correlated to the grade of the tumors and also to the history of the patients (p=0.013, p=0.036, respectively). Numerical alterations regarding gene and chromosome 7 were identified in 4/35 (11.4%) and 12/35 (43.2%) cases associated to the grade of the tumors (p=0.019, p=0.001, respectively) and to the survival rate of the patients (p=0.037, p=0.001, respectively). EGFR overall expression was also correlated to the gene copies (p=0.020). EGFR gene numerical alterations –although rare– and also chromosome 7 aneuploidy maybe affect prognosis in HCC patients. To our knowledge this is the first chromogenic in situ hybridization analysis based on tissue microarrays in hepatocellular carcinoma. C1 [Tsiambas, Evangelos] 417 Veterans Administration Hospital (Ν.Ι.Μ.Τ.S.), Department of Pathology, Section of Molecular Pathology, Ag Paraskevi Attiki, Patriarchou Grigoriou E 17, 15341 Athens, Greece. [Manaios, Loukas] Bioclinic, Dept of SurgeryAthens, Greece. [Papanikolopoulos, Costas] 401 GA Hospital, Dept of Internal MedicineAthens, Greece. [Rigopoulos, N Dimitrios] 401 GA Hospital, Dept of Internal MedicineAthens, Greece. [Tsounis, Dimitrios] 251 AF Hospital, Dept of GastroenterologyAthens, Greece. [Karameris, Andreas] 417 Veterans Administration Hospital (Ν.Ι.Μ.Τ.S.), Department of PathologyAthens, Greece. [Soultati, Aspasia] University of Athens, Hippokration H, Medical School, Dept of GastroenterologyAthens, Greece. [Koliopoulou, Antigoni] Evangelismos Hospital, Dept of Thoracic SurgeryAthens, Greece. [Kravvaritis, Christos] 417 Veterans Administration Hospital (Ν.Ι.Μ.Τ.S.), Department of PathologyAthens, Greece. [Sergentanis, Theodoros] University of Athens, Medical School, Department of PathologyAthens, Greece. [Patsouris, Efstratios] University of Athens, Medical School, Department of PathologyAthens, Greece. [Dourakis, Spyridon] University of Athens, Hippokration H, Medical School, Dept of GastroenterologyAthens, Greece. RP Tsiambas, E (reprint author), 417 Veterans Administration Hospital (Ν.Ι.Μ.Τ.S.), Department of Pathology, Section of Molecular Pathology, 15341 Athens, Greece. EM tsiambasecyto@yahoo.gr CR El Serag H, Davila JA, Petersen NJ, McGlynn KA, 2003, The continuing increase in the incidence of hepatocellular carcinoma in the United States: an update. Ann Intern Med 139:817–23 Llovet JM, Burroughs A, Bruix J, 2003, Hepatocellular carcinoma. Lancet 362:1907–17 Krause DS, Van Etten RA, 2005, Tyrosine kinases as targets for cancer therapy. N Engl J Med 353:172–187 Molina J, Adjei A, 2006, The Ras/Raf/MAPK Pathway. J Thorac Oncol l:17–9 Marquez A, Wu R, Zhao J, Tao J, Shi Z, 2004, Evaluation of epidermal growth factor receptor, EGFR, by chromogenic in situ hybridization, CISH, and immunohistochemistry, IHC, in archival gliomas using bright-field microscopy. Diagn Mol Pathol 13:1–8 Harari PM, 2004, Epidermal growth factor receptor inhibition strategies in oncology. Endocrine-Related Cancer 11:689–708 Arteaga C, 2001, The epidermal growth factor receptor: from mutant oncogene in nonhuman cancers to therapeutic target in human neoplasia. J Clin Oncol 19:S1832–40 Wiedmann MW, Casa K, 2005, Molecular targeted therapy for gastrointestinal cancer. Curr Cancer Drug Targets 5:171–93 Lynch TJ, Bell DW, Sordella R et al., 2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350:2129–39 Paez JG, Janne PA, Lee JC et al., 2005, EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304:1497–500 Huang SM, Armstrong EA, Benavente S, Chinnaiyan P, Harari PM, 2004, Dual-agent molecular targeting of the epidermal growth factor receptor, EGFR): combining anti-EGFR antibody with tyrosine kinase inhibitor. Cancer Res 64:5355–62 Zhu AX, Stuart K, Blaszkowsky LS et al., 2007, Phase 2 study of cetuximab in patients with advanced hepatocellular carcinoma. Cancer 110(3):581–9 Zhao YN, Cao J, Wu FX et al., 2004, Expression and significance of EGF m-RNA and EGFR m-RNA in hepatocellular carcinoma. Ai Zheng 23:762–6 Foster J, Black J, LeVea C et al., 2007, COX-2 expression in hepatocellular carcinoma is an initiation event; while EGF receptor expression with downstream pathway activation is a prognostic predictor of survival. Ann Surg Oncol 14(2):752–8 Breuhahn K, Longerich T, Schirmacher P, 2006, Dysregulation of growth factor signaling in human hepatocellular carcinoma. Oncogene 25(27):3787–0 Villanueva A, Newell P, Chiang DY, Friedman SL, Llovet JM, 2007, Genomics and signaling pathways in hepatocellular carcinoma. Semin Liver Dis 27(1):55–76 Szabo E, Paska C, Kaposi Novak P, Schaff Z, Kiss A, 2004, Similarities and differences in hepatitis B and C virus induced hepatocarcinogenesis. Pathol Oncol Res 10(1):5–11 Walker GJ, Hayward NK, Falvey S, Cooksley WG, 1991, Loss of somatic heterozygosity in hepatocellular carcinoma. Cancer Res 51(16):4367–70 Staib F, Hussain SP, Hofseth LJ, Wang XW, Harris CC, 2003, TP53 and liver carcinogenesis. Hum Mutat 21(3):201–16 Reeves ME, DeMatteo RP, 2000, Genes and viruses in hepatobiliary neoplasia. Semin Surg Oncol 19(2):84–93 Lee S, Park U, Lee YI, 2001, Hepatitis C virus core protein transactivates insulin-like growth factor II gene transcription through acting concurrently on Egr1 and Sp1 sites. Virology 283(2):167–77 Friedrich B, Wollersheim M, Brandenburg B, Foerste R, Will H, Hildt E, 2005, Induction of anti-proliferative mechanisms in hepatitis B virus producing cells. J Hepatol 43(4):696–703 El-Bassiouni A, Nosseir M, Zoheiry M, El-Ahwany E, Ghali A, El-Bassiouni N, 2006, Immunohistochemical expression of CD95, Fas), c-myc and epidermal growth factor receptor in hepatitis C virus infection, cirrhotic liver disease and hepatocellular carcinoma. APMIS 114(6):420–7 Lee SC, Lim SG, Soo R et al., 2007, Lack of somatic mutations in EGFR tyrosine kinase domain in hepatocellular and nasopharyngeal carcinoma. Pharmacogenet Genomics 16(1):73–4 Su MC, Lien HC, Jeng YM, 2005, Absence of epidermal growth factor receptor exon 18–21 mutation in hepatocellular carcinoma. Cancer Lett 224(1):117–21 Bekaii-Saab T, Williams N, Plass C, Calero MV, Eng C, 2006, A novel mutation in the tyrosine kinase domain of ERBB2 in hepatocellular carcinoma. BMC Cancer 6:278–85 Philip PA, Mahoney MR, Allmer C et al., 2005, Phase II study of Erlotinib, OSI-774, in patients with advanced hepatocellular cancer. J Clin Oncol 23:6657–63 Hopfner M, Sutter AP, Huether A, Schuppan D, Zeitz M, Scherubl H, 2004, Targeting the epidermal growth factor receptor by gefitinib for treatment of hepatocellular carcinoma. J Hepatol 41, 6):1008–16 Huether A, Hopfner M, Baradari V, Schuppan D, Scherubl H, 2005, EGFR blockade by cetuximab alone or as combination therapy fot growth control of hepatocellular cancer. Biochem Pharmacol 70(11):1568–78 Ramalingam S, Belani CP, 2007, Recent advances in targeted therapy for non-small cell lung cancer. Expert Opin Ther Targets 11(2):245–57 Buckley AF, Burgart LJ, Kakar S, 2006, Epidermal growth factor receptor expression and gene copy number in fibrolamellar hepatocellular carcinoma. Hum Pathol 37(4):410–4 Wilkens L, Bredt M, Flemming A et al., 2002, Detection of chromosomal aberrations in well-differentiated hepatocellular carcinoma by bright-field in situ hybridization. Mod Pathol 15(4):470–5 Moroni M, Veronese S, Benvenuti S, 2005, Gene copy number for epidermal growth factor receptor, EGFR, and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. Lancet Oncol 6(5):279–86 Moon WS, Chang KJ, Majumdar AP, Tarnawski AS, 2004, Reduced expression of epidermal growth factor receptor-related protein in hepatocellular carcinoma: implications for cancer growth. Digestion 69(4):219–24 Miyaki M, Sato C, Sakai K et al., 2000, Malignant transformation and EGFR activation of immortalized mouse liver epithelial cells caused by HBV enhancer-X from a human hepatocellular carcinoma. Int J Cancer 85(4):518–22 Bloom K, Harrington D, 2004, Enhanced accuracy and reliability of HER-2/neu immunohistochemical scoring using digital microscopy. Am J Clin Pathol 121:620–30 Lehr HA, Jacobs TW, Yaziji H, Schnitt SJ, Gown AM, 2001, Quantitative evaluation of HER-2/neu status in breast cancer by fluorescence in situ hybridization and by immunohistochemistry with image analysis. Am J Clin Pathol 115:814–22 Tsiambas E, Stavrakis I, Lazaris AC, Karameris A, Patsouris E, 2007, Evaluation of epidermal growth factor receptor gene and chromosome 7 alterations in squamous cell carcinoma of the larynx, using chromogenic in situ hybridization on tissue microarrays. J Laryngol Otol 121:563–570 Τsiambas E, Manaios L, Papanikolopoulos K et al., 2008, Epidermal growth factor receptor, vascular endothelial growth factor and hypoxia inducible factor 1a simultaneous deregulation in hepatocellular carcinoma based on tissue microarrays and quantitative protein analysis. J Hepatol Suppl 2 48:S206 Ramanathan RK, Belani CP, Singh DA, 2006, Phase II study of lapatinib, a dual inhibitor of epidermal growth factor receptor, EGFR, tyrosine kinase 1 and 2, Her2/Neu, in patients, pts, with advanced biliary tree cancer, BTC, or hepatocellular cancer, HCC). A California Consortium, CCC-P, Trial. J Clin Oncol 24:401 Liu L, Cao Y, Chen Ch et al., 2008)Sorafenib blocks the RAF/ MEK/ERK pathway, inhibits tumor angiogenesis and induces tumour cell apoptosis in hepatocellular carcinoma model PLC/ PRF/5. Cancer Res 66(24):11851–11858 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 511 EP 520 DI 10.1007/s12253-008-9146-5 PG 10 ER PT J AU Milinovic, D Kalafatic, D Babic, D Oreskovic, BL Grsic, LH Oreskovic, S AF Milinovic, Darko Kalafatic, Drzislav Babic, Damir Oreskovic, Beketic Lidija Grsic, Lovric Helena Oreskovic, Slavko TI Minimally Invasive Therapy of Cervical Intraepithelial Neoplasia for Fertility Preservation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CIN III; Conization; Fertility preservation ID CIN III; Conization; Fertility preservation AB The aim of this study was to determine the extension of cervical intraepithelial neoplasia grade III (CIN III) into endocervical canal and depth of endocervical crypts involvement by CIN with the regard to patients’ age and parity. Correlation between the area of CIN involvement and the extension into endocervical canal was estimated. A total of 218 cervical cone specimens with histologically proven CIN III were included in this study. Extension of CIN into the endocervical canal, depth of involved crypts and ectocervical area affected by CIN were histologically analyzed. The average endocervical crypt involvement was at 1.2 mm of depth. The excision of >4 mm (1.2 mm × 3S.D.) in depth removes >99% of CIN. With the cone length of 15 mm (nulliparous patients) and 18 mm (multiparous patients), no endocervical cone margins were affected with CIN. Since the cone length is the most important determining factor for fertility preservation, the measurement of cervical cone could be essential for future pregnancies. C1 [Milinovic, Darko] General Hospital Gospic, Department of Obstetrics and GynecologyGospic, Croatia. [Kalafatic, Drzislav] University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Obstetrics and GynecologyZagreb, Croatia. [Babic, Damir] University of Zagreb, School of Medicine, Department of PathologyZagreb, Croatia. [Oreskovic, Beketic Lidija] University Hospital for Tumors, Department of OncologyZagreb, Croatia. [Grsic, Lovric Helena] University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Obstetrics and GynecologyZagreb, Croatia. [Oreskovic, Slavko] University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Obstetrics and GynecologyZagreb, Croatia. RP Kalafatic, D (reprint author), University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Obstetrics and Gynecology, Zagreb, Croatia. EM drzislav.kalafatic@zg.t-com.hr CR Bosch FX, Manos MM, Munoz N, 1995, Prevalence of human papillomavirus in cervical cancer: a worldwide perspective. International biological study on cervical cancer, IBSCC, Study group. J Natl Cancer Inst 87:796–802 Formso S, Hansen MH, Jacobsen BK, Oian P, 1996, Pregnancy outcome after laser surgery for cervical intraepithelial neoplasia. Acta Obstet Gynecol Scand 75:139–143 Kurman RJ, Solomon D, 1994, The Bethesda System for reporting cervical/vaginal cytologic diagnoses. Springer-Verlag, New York, NY, p 30–78 Nyirjesy I, Billingsley FS, Forman MR, 1998, Evaluation of atypical and low-grade cervical cytology in private practice. Obstet Gynecol 92:601–607 Ostor AG, 1993, Natural history of cervical intraepithelial neoplasia: a critical review. Int J Gynecol Pathol 12:106 Bjerre B, Eliasson G, Linell F, 1976, Conization as only treatment of carcinoma in situ of the uterine cervix. Am J Obstet Gynecol 125:143–152 Reich O, Pickel H, Lahousen M, 2001, Cervical intraepithelial neoplasia III: long term outcome after cold-knife conization with clear margins. Obstet Gynecol 97:428–430 DiSaia PJ, Creasman WT, 2001, Preinvasive disease of the cervix. In DiSaia PJ, Creasman WT, eds, Clinical gynecologic oncology. Mosby, Inc., St. Louis, p 1–33 Anderson MC, Hartley RB, 1980, Cervical crypt involvement by intraepithelial neoplasia. Obstet Gynecol 55:546–550 Coppleson M, Atkinson KH, Dalrymple JC, 1992, Cervical squamous and glandular intraepithelial neoplasia: clinical features and review of management. Coppleson M, ed, Gynecological Oncology Vol. 1, Churcill Livingstone, Edinburgh, p 549 Boonstra H, Aalders JG, Koudstaal J, Oosterhuis JW, Janssens J, 1990, Minimum extension and appropriate topographic position of tissue destruction for treatment of cervical intraepithelial neoplasia. Obstet Gynecol 75:227–231 Dabic MM, Hlupic L, Babic D, Jukic S, Seiwerth S, 2004, Comparison of polymerase chain reaction and catalyzed signal amplification in situ hybridizationmethods for human papillomavirus detection in paraffin-embedded cervical preneoplastic and neoplastic lesions. Arch Med Res 35:511–516 Perlman SE, Lubianca JN, Kahn JA, 2003, Characteristics of a group of adolescents undergoing Loop Electrical Excision Procedure, LEEP). J Pediatr Adolesc Gynecol 16:15–20 Anderson MC, Hartley RB, 1980, Cervical crypt involvement by intraepithelial neoplasia. Obstet Gynecol 55:546–550 Ljubojevic N, Babic S, Audy-Jurkovic S, Ovanin-Rakic A, Jukic S, Babic D, Grubisic G, Radakovic B, Ljubojevic-Grgec D, 2001, Improved national Croatian diagnostic and therapeutic guidelines for premalignant lesions of the uterine cervix with some costbenefit aspects. Coll Antropol Dec 25:467–474 Abdul-Karim FW, Fu YS, Reagan JW, Wentz WB, 1982, Morphometric study of intraepithelial neoplasia of the uterine cervix. Obstet Gynecol 60:210–214 Ludmir J, Sehdev HM, 2000, Anatomy and physiology of the uterine cervix. Clin Obstet Gynecol 43:433–439 Berdichevsky L, Karmin R, Chuang L, 2004, Treatment of highgrade squamous intraepithelial lesions: a 2-versus 3-step approach. Am J Obstet Gynecol 190:1424–1426 Mathevet P, Chemali E, Roy M, Dargent D, 2003, Long-term outcome of a randomized study comparing three techniques of conization: cold knife, laser, and LEEP. Eur J Obstet Gynecol Reprod Biol 106:214–218 Kleinberg MJ, Straughn JM Jr, Stringer JS, Partridge EE, 2003, A cost-effectiveness analysis of management strategies for cervical intraepithelial neoplasia grades 2 and 3. Am J Obstet Gynecol 188:1186–1188 Lin H, Chang HY, Huang CC, Changchien CC, 2004, Prediction of disease persistence after conization for microinvasive cervical carcinoma and cervical intraepithelial neoplasia grade 3. Int J Gynecol Cancer 14:311–316 Szurkus DC, Harrison TA, 2003, Loop excision for high-grade squamous intraepithelial lesion on cytology: Correlation with colposcopic and histologic findings. Am J Obstet Gynecol 188:1180–1182 Orbo A, Arnesen T, Arnes M, Straume B, 2004, Resection margins in conization as prognostic marker for relapse in highgrade dysplasia of the uterine cervix in northern Norway: a retrospective long-term follow-up material. Gynecol Oncol 93:479–483. Paraskevaidis E, Lolis ED, Koliopoulos G, Alamanos Y, Fotiou S, Kitchener HC, 2000, Cervical intraepithelial neoplasia outcomes after large loop excision with clear margins. Obstet Gynecol 95:828–831 Nuovo J, Melnikow J, Willan AR, Chan BK, 2000, Treatment outcomes for squamous intraepithelial lesions. Int J Gynaecol Obstet 68:25–33 Ljubojevic N, Babic S, Audy-Jurkovic S, et al, 1998, Loop excision of the transformation zone, LETZ, as an outpatient method of management for women with cervical intraepithelial neoplasia: our experience. Coll Antropol 22:533–543 Cecchini S, Visioli CB, Zappa M, Ciatto S., 2002, Recurrence after treatment by loop electrosurgical excision procedure, LEEP, of high-grade cervical intraepithelial neoplasia. Tumori 88:478–480 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 521 EP 525 DI 10.1007/s12253-009-9148-y PG 5 ER PT J AU Bori, R Sejben, I Svebis, M Vajda, K Marko, L Pajkos, G Cserni, G AF Bori, Rita Sejben, Istvan Svebis, Mihaly Vajda, Kornel Marko, Laszlo Pajkos, Gabor Cserni, Gabor TI Heterogeneity of pT3 Colorectal Carcinomas According to the Depth of Invasion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal carcinoma; TNM; Depth of invasion; Metastasis; Venous invasion ID Colorectal carcinoma; TNM; Depth of invasion; Metastasis; Venous invasion AB Colorectal carcinomas (CRCs) infiltrating through the muscularis propria layer without infiltration of adjacent structures, organs or the serosa—i.e. the pT3 tumors, compose the largest subset of large intestinal carcinomas treated by surgical resection. They are heterogeneous in terms of prognosis. CRCs treated by surgery in a period of 69 months were prospectively classified as pT3a tumors (invading to a maximum of 5 mm beyond the muscularis propria) and pT3b tumors (invading deeper). Their nodal status, incidence of vascular invasion and the presence or absence of distant metastases were analyzed in relation to the depth of invasion. Of the 593 CRCs primarily treated by surgery 429 were pT3 tumors. CRCs categorized as pT3a had significantly lower rates of nodal involvement (44% vs 75%), massive nodal involvement (pN2) (9% vs 39%), venous invasion (17% vs 30%) and distant metastasis (11% vs 28%) than pT3b tumors. Significant differences in these prognostic variables in pT3a and pT3b cancers were observed both for carcinomas of the colon and those of the rectum. Such differences were not obvious in further 66 ypT3 cases of rectal carcinoma receiving neoadjuvant treatment before surgery. Tumors in the pT3a category are associated with a better prognostic profile than pT3b tumors. This subdivision might be useful in both prognostication and treatment planning. C1 [Bori, Rita] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., 6000 Kecskemet, Hungary. [Sejben, Istvan] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., 6000 Kecskemet, Hungary. [Svebis, Mihaly] Bacs-Kiskun County Hospital, Department of SurgeryKecskemet, Hungary. [Vajda, Kornel] Kiskunfelegyhaza City Hospital, Department of SurgeryKiskunfelegyhaza, Hungary. [Marko, Laszlo] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. [Pajkos, Gabor] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38., 6000 Kecskemet, Hungary. RP Bori, R (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, 6000 Kecskemet, Hungary. EM boririta@hotmail.com CR Cserni G, 2003, Nodal staging of colorectal carcinomas and sentinel nodes. J Clin Pathol 56:327–335 Sobin LH, Wittekind C, eds,, 2002, UICC TNM classification of malignant tumours, 6th edition. JohnWiley and Sons Inc., New York Greene FL, Page DL, Fleming ID, et al, eds,, 2002, AJCC Cancer Staging Handbook—TNM Classification of Malignant Tumors, 6th edition. Springer Verlag, New York Heinzerling JH, Anthony T, Livingston EH, et al,, 2007, Predictors of distant metastasis and mortality in patients with stage II colorectal cancer. Am Surg 73:230–238 Ricciardi R, Madoff RD, Rothenberger DA, et al,, 2006, Population-based analyses of lymph node metastases in colorectal cancer. Clin Gastroenterol Hepatol 4:1522–1527 Abdulkader M, Abdulla K, Rakha E, et al,, 2006, Routine elastic staining assists detection of vascular invasion in colorectal cancer. Histopathology 49:487–492 Wong SK, Jalaludin BB, Henderson CJ, 2008, Direct tumor invasion in colon cancer: correlation with tumor spread and survival. Dis Colon Rectum 51:1331–1338 Adachi Y, Inomata M, Kakisako K, 1999, Histopathologic characteristics of colorectal cancer with liver metastasis. Dis Colon Rectum 42:1053–1056 Cserni G, Vajda K, Tarjan M, et al,, 1999, Nodal staging of colorectal carcinomas from quantitative and qualitative aspects. Can lymphatic mapping help staging? Pathol Oncol Res 5:291– 296 Wittekind C, Greene FL, Henson DE, eds,, 2003, TNM Supplement: a Commentary on Uniform Use. 3rd edn. Wiley-Liss, New York Compton CC, Greene FL, 2004, The staging of colorectal cancer: 2004 and beyond. CA Cancer J Clin 54:295–308 Lowry R, 2008, VassarStats. http://faculty.vassar.edu/lowry/Vassar Stats.html Cited 11 Jul 2008 Cawthorn SJ, Parums DV, Gibbs NM, et al,, 1990, Extent of mesorectal spread and involvement of lateral resection margin as prognostic factors after surgery for rectal cancer. Lancet 335, 8697):1055–1059 Willett CG, Badizadegan K, Ancukiewicz M, et al,, 1999, Prognostic factors in stage T3N0 rectal cancer: do all patients require postoperative pelvic irradiation and chemotherapy? Dis Colon Rectum 42:167–173 Merkel S, Mansmann U, Siassi M, 2001, The prognostic inhomogeneity of pT3 rectal carcinomas. Int J Colorectal Dis 16:298–304 Harewood GC, Kumar KS, Clain JE, 2004, Clinical implications of quantification of mesorectal tumor invasion by endoscopic ultrasound: All T3 rectal cancers are not equal. J Gastroenterol Hepatol 19:750–755 Scott N, Thorne C, Jayne D, 2004, Lymph node retrieval after neoadjuvant radiotherapy for rectal adenocarcinoma. J Clin Pathol 57:335–336 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 527 EP 532 DI 10.1007/s12253-009-9149-x PG 6 ER PT J AU Nemeth, Zs Szasz, MA Somoracz, Tatrai, P Nemeth, J Gyorffy, H Szijarto, A Kupcsulik, P Kiss, A Schaff, Zs AF Nemeth, Zsuzsanna Szasz, Marcell Attila Somoracz, Aron Tatrai, Peter Nemeth, Julia Gyorffy, Hajnalka Szijarto, Attila Kupcsulik, Peter Kiss, Andras Schaff, Zsuzsa TI Zonula Occludens-1, Occludin, and E-cadherin Protein Expression in Biliary Tract Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adherens junction; Biliary tract cancer; Tight junction ID Adherens junction; Biliary tract cancer; Tight junction AB The incidence of cholangiocarcinomas originating from intrahepatic and extrahepatic bile ducts, as well as of gallbladder carcinoma is increasing worldwide. The malignant transformation of biliary epithelia involves profound alterations of proteins in the intercellular junctions, among others zonula occludens-1 (ZO-1), occludin, and E-cadherin. Each plays important role in the maintenance of epithelial cell polarity and regulation of cell growth and differentiation. Our aim was to investigate ZO-1, occludin, and E-cadherin immunohistochemical reactions in tissue microarray blocks containing 57 normal and 62 neoplastic biliary tract samples. We demonstrated that the tight junction components ZO-1, occludin, and E-cadherin are downregulated in carcinomas arising from various compartments of the biliary tract (normal intrahepatic and extrahepatic bile ducts, gallbladder) as compared with their normal sites of origin. These results were confirmed by discriminant analysis yielding clear separation of the three normal sample groups from carcinomas in the corresponding locations. C1 [Nemeth, Zsuzsanna] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Szasz, Marcell Attila] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Somoracz, Aron] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Tatrai, Peter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Nemeth, Julia] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Gyorffy, Hajnalka] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Szijarto, Attila] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Kupcsulik, Peter] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM schaff@korb2.sote.hu CR Kakar S, Ferrell LD, 2007, Tumors of the liver, gallbladder and biliary tree. In: Fletcher CDM, ed, Diagnostic histopathology of tumors, 3rd edn. Elsevier Limited, Philadelphia, pp 417–462 Chuang SC, Lee KT, Tsai KB, et al, 2004, Immunohistochemical study of DPC4 and p53 proteins in gallbladder and bile duct cancers. World J Surg 28:995–1000 Bajor J, Bero T, GaramszegiM, et al, 2001, Common bile duct tumor in a young woman with ulcerative colitis. Orv Hetil 142:1231–1234 Hong SM, Kim MJ, Pi DY, et al, 2005, Analysis of extrahepatic bile duct carcinomas according to the New American Joint Committee on Cancer staging system focused on tumor classification problems in 222 patients. Cancer 104:802–810 Obama K, Ura K, Li M, et al, 2005, Genome-wide analysis of gene expression in human intrahepatic cholangiocarcinoma. Hepatology 41:1339–1348 Lewis JT, Talwalkar JA, Rosen CB, et al, 2007, Prevalence and risk factors for gallbladder neoplasia in patients with primary sclerosing cholangitis: evidence for a metaplasia-dysplasia-carcinoma sequence. Am J Surg Pathol 31:907–913 Kobayashi S, Ohnuma N, Yoshida H, et al, 2006, Preferable operative age of choledochal dilation types to prevent patients with pancreaticobiliary maljunction from developing biliary tract carcinogenesis. Surgery 139:33–38 Gonzalez-Mariscal L, Lechuga S, Garay E, 2007, Role of tight junctions in cell proliferation and cancer. Prog Histochem Cytochem 42:1–57 Hartsock A, Nelson WJ, 2008, Adherens and tight junctions: structure, function and connections to the actin cytoskeleton. Biochim Biophys Acta 1778(3):660–669 Schneeberger EE, Lynch RD, 2004, The tight junction: a multifunctional complex. Am J Physiol Cell Physiol 286: C1213–C1228 Rodriguez-Boulan E, Nelson WJ, 1989, Morphogenesis of the polarized epithelial cell phenotype. Science 245:718–725 Miyoshi J, Takai Y, 2005, Molecular perspective on tight-junction assembly and epithelial polarity. Adv Drug Deliv Rev 57:815–855 Gumbiner B, 1987, Structure, biochemistry, and assembly of epithelial tight junctions. Am J Physiol 253:C749–C758 Gumbiner BM, 1993, Breaking through the tight junction barrier. J Cell Biol 123:1631–1633 Anderson JM, Van Itallie CM, 1995, Tight junctions and the molecular basis for regulation of paracellular permeability. Am J Physiol 269:G467–G475 Ivanov AI, Nusrat A, Parkos CA, 2005, Endocytosis of the apical junctional complex: mechanisms and possible roles in regulation of epithelial barriers. Bioessays 27:356–365 Tsukita S, Furuse M, Itoh M, 1999, Structural and signalling molecules come together at tight junctions. Curr Opin Cell Biol 11:628–633 Balda MS, Matter K, 1998, Tight junctions. J Cell Sci 111(Pt 5):541–547 Cowin P, Rowlands TM, Hatsell SJ, 2005, Cadherins and catenins in breast cancer. Curr Opin Cell Biol 17:499–508 Berx G, van Roy F, 2001, The E-cadherin/catenin complex: an important gatekeeper in breast cancer tumorigenesis and malignant progression. Breast Cancer Res 3:289–293 Guzman G, Chejfec G, 2007, Tumors of the digestive system. In: Damjanov I, Fan F, ed, Cancer grading manual. Springer, New York, pp 43–44 Umeda K, Ikenouchi J, Katahira-Tayama S, et al, 2006, ZO-1 and ZO-2 independently determine where claudins are polymerized in tight-junction strand formation. Cell 126:741–754 Hugh TJ, Dillon SA, Taylor BA, et al, 1999, Cadherin-catenin expression in primary colorectal cancer: a survival analysis. Br J Cancer 80:1046–1051 Truant SC, Gouyer VP, Leteurtre EA, et al, 2008, E-Cadherin and beta-Catenin mRNA Levels Throughout Colon Cancer Progression. J Surg Res 2008 Feb 11 [Epub ahead of print] Laurila JJ, Karttunen T, Koivukangas V, et al, 2007, Tight junction proteins in gallbladder epithelium: different expression in acute acalculous and calculous cholecystitis. J Histochem Cytochem 55:567–573 Kleeff J, Shi X, Bode HP, et al, 2001, Altered expression and localization of the tight junction protein ZO-1 in primary and metastatic pancreatic cancer. Pancreas 23:259–265 Medici D, Hay ED, Goodenough DA, 2006, Cooperation between snail and LEF-1 transcription factors is essential for TGF-beta1- induced epithelial-mesenchymal transition. Mol Biol Cell 17:1871– 1879 Kurrey NK, K A, Bapat SA, 2005, Snail and Slug are major determinants of ovarian cancer invasiveness at the transcription level. Gynecol Onco l97:155–165 Ikenouchi J, Matsuda M, Furuse M, et al, 2003, Regulation of tight junctions during the epithelium-mesenchyme transition: direct repression of the gene expression of claudins/occludin by Snail. J Cell Sci 116:1959–1967 Okada K, Shimura T, Asakawa K, et al, 2007, Fascin expression is correlated with tumor progression of extrahepatic bile duct cancer. Hepatogastroenterology 54:17–21 Farazi PA, Zeisberg M, Glickman J, et al, 2006, Chronic bile duct injury associated with fibrotic matrix microenvironment provokes cholangiocarcinoma in p53-deficient mice. Cancer Res 66:6622– 6627 Hirata K, Ajiki T, Okazaki T, et al, 2006, Frequent occurrence of abnormal E-cadherin/beta-catenin protein expression in advanced gallbladder cancers and its association with decreased apoptosis. Oncology 71:102–110 Settakorn J, Kaewpila N, Burns GF, et al, 2005, FAT, E-cadherin, beta catenin, HER 2/neu, Ki67 immuno-expression, and histological grade in intrahepatic cholangiocarcinoma. J Clin Pathol 58:1249–1254 Park S, Kim SW, Lee BL, et al, 2006, Expression of E-cadherin and beta-catenin in the adenoma-carcinoma sequence of ampulla of Vater cancer. Hepatogastroenterology 53:28–32 Orban E, Szabo E, Lotz G, et al, 2008, Different expression of Occludin and ZO-1 in primary and metastatic liver tumors. Pathol Oncol Res 14:299–306 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2009 VL 15 IS 3 BP 533 EP 539 DI 10.1007/s12253-009-9150-4 PG 7 ER PT J AU Ellsworth, ER Hooke, AJ Love, B Ellsworth, LD Shriver, DC AF Ellsworth, E Rachel Hooke, A Jeffrey Love, Brad Ellsworth, L Darrell Shriver, D Craig TI Molecular Changes in Primary Breast Tumors and the Nottingham Histologic Score SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Allelic imbalance; Grade; Mitosis; Nuclear atypia; Tubule formation ID Allelic imbalance; Grade; Mitosis; Nuclear atypia; Tubule formation AB Pathological grade is routinely used to stratify breast cancer patients into favorable and less favorable outcome groups. Mechanisms by which genomic changes in breast tumors specifically contribute to the underlying components of tumor grade – tubule formation, nuclear pleomorphism, and mitoses — are unknown. This study examined 26 chromosomal regions known to be altered in breast cancer in 256 invasive breast carcinomas. Differences in overall levels and patterns of allelic imbalance (AI) at each chromosomal region were compared for tumors with favorable (=1) and unfavorable (=3) scores for tubule formation, nuclear pleomorphism and mitotic count. Levels of AI were significantly different between samples with high and low scores for tubule formation (P<0.001), nuclear pleomorphism (P<0.001) and mitotic count (P<0.05). Significantly higher levels of AI were detected at regions 11q23 and 13q12 for tumors with reduced tubule formation, chromosomes 9p21, 11q23, 13q14, 17p13 and 17q12 for those with high levels of nuclear atypia, and chromosomes 1p36, 11q23, and 13q14 for those with high mitotic counts. Region 16q11-q22 showed significantly more AI events in samples with low nuclear atypia. Patterns of genetic changes associated with poorly-differentiated breast tumors were recapitulated by the individual components of the Nottingham Histologic Score. While frequent alteration of 11q23 is common for reduced tubule formation, high nuclear atypia and high mitotic counts, suggesting that this is an early genetic change in the development of poorly-differentiated breast tumors, alterations at the other seven loci associated with poorly-differentiated tumors may specifically influence cell structure, nuclear morphology and cellular proliferation. C1 [Ellsworth, E Rachel] Henry M. Jackson Foundation for the Advancement of Military Medicine, Clinical Breast Care Project, 620 Seventh Street, 15963 Windber, PA, USA. [Hooke, A Jeffrey] Walter Reed Army Medical Center, Clinical Breast Care ProjectWashington, DC, USA. [Love, Brad] InvitrogenCarlsbad, CA, USA. [Ellsworth, L Darrell] Windber Research Institute, Clinical Breast Care ProjectWindber, PA, USA. [Shriver, D Craig] Walter Reed Army Medical Center, Clinical Breast Care ProjectWashington, DC, USA. RP Ellsworth, ER (reprint author), Henry M. Jackson Foundation for the Advancement of Military Medicine, Clinical Breast Care Project, 15963 Windber, USA. EM r.ellsworth@wriwindber.org CR Bloom HJ, Richardson WW, 1957, Histological grading and prognosis in breast cancer. Br J Cancer 11:359–377 Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow up. Histopathology 19:403–410 Henson DE, Ries L, Freedman LS, Carriaga M, 1991, Relationship among outcome, stage of disease, and histologic grade for 22, 616 cases of breast cancer. Cancer 68:2142–2149 Robbins P, Pinder S, de Klerk N et al, 1995, Histological grading of breast carcinomas: a study of interobserver agreement. Hum Pathol 26:873–879 Sorlie T, Perou CM, Tibshirania R et al, 2001, Gene Expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA 98:10869–10874 Sorlie T, Tibshirania R, Parker J et al, 2003, Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci USA 100:8418–8423 Paik S, Shak S, Tang G et al, 2004, A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 351:2817–2826 Ramaswamy S, Ross KN, Lander ES, Golub TR, 2003, A molecular signature of metastasis in primary solid tumors. Nat Genet 33:49–54 van't Veer LJ, Dai H, vande Vijver MJ et al, 2002, Gene expression profiling predicts clincial outcome of breast cancer. Nature 415:530–536 Sotiriou C, Wirapati P, Loi S et al, 2006, Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst 98:262– 272 Ellsworth RE, Hooke JA, Love B et al, 2008, Correlation of levels and patterns of genomic instability with histological grading of invasive breast tumors. Breast Cancer Res Treat 107:259–265 Roylance R, Gorman P, Harris W et al, 1999, Comparative genomic hybridization of breast tumors stratified by histological grade reveals new insights into the biological progression of breast cancer. Cancer Res 59:1433–1436 Roylance R, Gorman P, Hanby A, Tomlinson I, 2002, Allelic imbalance analysis of chromosome 16q shows that grade I and grade III invasive ductal breast cancers follow different genetic pathways. J Pathol 196:32–36 Ellsworth DL, Shriver CD, Ellsworth RE, Deyarmin B, Somiari RI, 2003, Laser capture microdissection of paraffin-embedded tissues. Biotechniques 34:42–46 Ellsworth RE, Ellsworth DL, Lubert SM et al, 2003, Highthroughput loss of heterozygosity mapping in 26 commonly deleted regions in breast cancer. Cancer Epidemiol Biomarkers Prev 12:915–919 Medintz IL, Lee C-CR, Wong WW et al, 2000, Loss of heterozygosity assay for molecular detection of cancer using energy-transfer primers and capillary array electrophoresis. Genome Res 10:1211–1218 Ellsworth RE, Ellsworth DL, Neatrour DM et al, 2005, Allelic imbalance in primary breast carcinomas and metastatic tumors of the axillary lymph nodes. Mol Cancer Res 3:71–77 Fitzgibbons PL, Page DL, Weaver D et al, 2000, Prognostic factors in breast cancer. College of American Pathologists Consensus Statement 1999. Arch Pathol Lab Med 124:966–978 Fechter A, Buettel I, Kuehnel E, Savelyeva L, Schwab M, 2007, Common fragile site RFA11G and rare fragile site FRA11B at 11q23.3 encompass distinct genomic regions. Genes Chromosomes Cancer 46:98–106 Srivastava N, Gochhait S, Gupta P, Bamezai RNK, 2008, Copy number alterations of the H2AFX gene in sporadic breast cancer patients. Genes Chromosomes Cancer 180:121–128 Bagchi A, Papazoglu C, Wu Y et al, 2007, CHD5 is a tumor suppressor at human 1p36. Cell 128:459–475 Ellsworth RE, Ellsworth DL, Love B et al, 2007, Correlation of levels and patterns of genomic instability with histological grading of DCIS. Ann Surg Onc 14:3070–3077 Bieche I, Lidreau R, 2000, Loss of heterozygosity at 13q14 correlates with RB1 gene underexpression in human breast cancer. Mol Carcinog 29:151–158 Buerger H, Mommers EC, Littmann R et al, 2001, Ductal invasive G2 and G3 carcinomas of the breast are the end stages of at least two different lines of genetic evolution. J Pathol 194: 165–170 Cleton-Jansen AM, Buerger H, ter Haar NT et al, 2004, Different mechanisms of chromosome 16 loss of heterozygosity in well— versus poorly differentiated ductal breast cancer. Genes Chromosomes Cancer 41:109–116 van Wezel T, Lombaerts M, van Roon EH et al, 2005, Expression analysis of candidate breast tumor suppressor genes on chromosome 16q. Breast Cancer Res 7:R998–R1004 Blegen H, Will JS, Ghadimi BM et al, 2003, DNA amplifications and aneuploidy, high proliferative activity and impaired cell cycle control characterize breast carcinomas with poor prognosis. Anal Cell Pathol 25:103–114 Seitz S, Poppe K, Fischer J et al, 2001, Detailed deletion mapping in sporadic breast cancer at chromosomal region17p13 distal to the TP53 gene: association with clinicopathological parameters. J Pathol 194:318–326 Winqvist R, Hampton GM, Mannermaa A et al, 1995, Loss of heterozygosity for chromosome 11 in primary human breast tumors is associated with poor survival after metastasis. Cancer Res 55:2660–2664 Tsuda H, Hirohashi S, 1994, Association among p53 gene mutation, nuclear accumulation of the p53 protein and aggressive phenotypes in breast cancer. Int J Cancer 57:498–503 Yang Q, Mori I, Sakurai T et al, 2001, Correlation between nuclear grade and biological prognostic variables in invasive breast cancer. Breast Cancer 8:105–110 Hanby AM, Kelsell DP, Potts HWet al, 2007, Association between loss of heterozygosity of BRCA1 and BRCA2 and morphological attributes of sporadic breast cancer. Int J Cancer 88:204–208 Patel KJ, Yu VP, Lee H et al, 1998, Involvement of Brca2 in DNA repair. Mol Cell 1:347–357 Han X, Saito H, Miki Y, Nakanishi A, 2008, A CRM1-mediated nuclear export signal governs cytoplasmic localization of BRCA2 and is essential for centrosomal localization of BRCA2. Oncogene 27:2969–2977 Rank F, Dombernowsky P, Jespersen NC, Pedersen BV, Keiding N, 1987, Histologic malignancy grading of invasive ductal breast carcinoma. A regression analysis of prognostic factors in low-risk carcinomas from a multicenter trial. Cancer 60:1299–1305 Tsuda H, Akiyama F, Kurosumi M, Sakamoto G, Watanabe T, 1998, Establishment of histological criteria for high-risk nodenegative breast carcinoma for a multi-institutional randomized clinical trial of adjuvant therapy. Japan National Surgical Adjuvant Study of Breast Cancer, NSAS-BC, Pathology Section. Jpn J Clin Oncol 28:486–491 Komaki K, Sano N, Tangoku A, 2006, Problems in histological grading of malignancy and its clinical significance in patients with operable breast cancer. Breast Cancer 13:249–253 Berx G, Cleton-Jansen AM, Nollet F et al, 1995, E-cadherin is a tumour/invasion suppressor gene mutated in human lobular breast cancers. EMBO 14:6107–6115 Simpson PT, Reis-Filho JS, Lambros MB et al, 2008, Molecular profiling pleomorphic lobular carcinomas of the breast: evidence for a common molecular genetic pathway with classic lobular carcinomas. J Pathol 215:231–244 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 541 EP 547 DI 10.1007/s12253-009-9151-3 PG 7 ER PT J AU Salinas, RN Lopes, TC Palma, VP Oshima, TC Bueno, V AF Salinas, R Natalia Lopes, T Camila Palma, V Patricia Oshima, T Celina Bueno, Valquiria TI Lung Tumor Development in the Presence of Sphingosine 1-phosphate Agonist FTY720 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Urethane; FTY720; Apoptosis; Proliferation; CD4 + Tcell ID Lung cancer; Urethane; FTY720; Apoptosis; Proliferation; CD4 + Tcell AB Urethane is a chemical carcinogen which causes lung tumorigenesis in mice with similarities to human adenocarcinoma (AC). The sphingosine 1-phosphate agonist FTY720 administered to mice in doses above 5 mg/kg/day has been able to prevent hepatocellular carcinoma and bladder cancer. We used BALB/c mice in urethane-induced lung cancer model to investigate the effects of a lower dose of FTY720 (1 mg/kg/day). The benefits of FTY720 were associated with the time point of the compound administration. FTY720 30 Group presented lower incidence and smaller area of lung nodules, decreased PCNA and increased Caspase-3 expressions. The findings in FTY720 0 Group (nodule multiplicity and area, PCNA expression) were similar to Urethane Group suggesting that the administration of the compound at early time point did not affect lung tumor development. FTY720 90 Group presented the biggest nodule area which was associated with increased PCNA and decreased Caspase-3 expressions. FTY720 (30 days and 90 days) administration decreased CD4+splenocytes and blood lymphocytes which caused opposite effects in lung tumor development - impairment and improvement respectively. In conclusion, FTY720 in low dose did not provide lung tumor inhibition in mice but its administration 30 days after the chemical carcinogen (Urethane) injection was associated with impaired tumor development. C1 [Salinas, R Natalia] Federal University of Sao Paulo, Immunology Division, Rua Botucatu 862, 4° andar, CEP 04023-900 Sao Paulo, SP, Brazil. [Lopes, T Camila] Federal University of Sao Paulo, Immunology Division, Rua Botucatu 862, 4° andar, CEP 04023-900 Sao Paulo, SP, Brazil. [Palma, V Patricia] USP Ribeirao Preto Medical School, Laboratory of Flow CytometryRibeirao Preto, SP, Brazil. [Oshima, T Celina] Sao Paulo University School of Medicine, Department of PathologySao Paulo, SP, Brazil. [Bueno, Valquiria] Federal University of Sao Paulo, Immunology Division, Rua Botucatu 862, 4° andar, CEP 04023-900 Sao Paulo, SP, Brazil. RP Bueno, V (reprint author), Federal University of Sao Paulo, Immunology Division, CEP 04023-900 Sao Paulo, Brazil. EM valquiria@nefro.epm.br;valquiriabueno@hotmail.com CR Malkinson AM, Beer DS, 1983, Major effect on susceptibility to urethane-induced pulmonary adenoma by a single gene in BALB/ cBy mice. J Natl Cancer Inst 70:931–936 Malkinson AM, 1998, Molecular comparison of human and mouse pulmonary adenocarcinomas. Exp Lung Res 24:541–555 Manson RJ, Kalina M, Nielsen LDet al, 2000, Surfactant protein C expression in urethane-induced murine pulmonary tumors. Am J Pathol 156:175–182 Wang M, Devereux TR, Vikis HGet al, 2004, Pol ioto is a candidate for the mouse pulmonary adenoma resistance 2 locus, a major modifier of chemically induced lung neoplasia. Cancer Res 64:1924–1931 O'Donnell EP, Zerbe LK, Dwyer-Nield LDet al, 2006, Quantitative analysis of early chemically-induced pulmonary lesions in mice of varying susceptibilities to lung tumorigenesis. Cancer Lett 241:197–202 Karasaki H, Obata M, Ogawa K, Lee GH, 1997, Roles of the Pas1 and Par2 genes in determination of the unique, intermediate susceptibility of BALB/cByJ mice to urethane-induction of lung carcinogenesis: differential effects on tumor multiplicity, size and Kras2 mutations. Oncogene 15:1833–1840 Stathopoulos GT, Sherrill TP, Cheng D-Set al, 2007, Epithelial NF-κB activation promotes urethane-induced lung carcinogenesis. Proc Natl Acad Sci U S A 104:18514–18519 LaMontagne K, Littlewood-Evans A, Schnell Cet al, 2006, Antagonism of sphigosine-1-phosphate receptors by FTY720 inhibits angiogenesis and tumor vascularization. Cancer Res 66:221–231 Azuma H, Takahara S, Ichimaru Net al, 2002, Marked Prevention of Tumor Growth and Metastasis by a Novel Immunosuppressive Agent, FTY720, in Mouse Breast Cancer Models. Cancer Res 62:1410–1419 Azuma H, Takahara S, Horie Set al, 2003, Induction of apoptosis in human bladder cancer cells in vitro and in vivo caused by FTY720 treatment. J Urol 169:2372–2377 Schmid G, Guba M, Papyan Aet al, 2005, FTY720 inhibits tumor growth and angiogenesis. Transplant Proc 37:110–111 Chua CW, Lee DT, Ling MTet al, 2005, FTY720, a fungus metabolite, inhibits in vivo growth of androgen-independent prostate cancer. Int J Cancer 117:1039–1048 Lee TK, Man K, Ho JWet al, 2005, FTY720: a promising agent for treatment of metastaic hepatocellular carcinoma. Clin Cancer Res 11:8458–8466 Silva LBL, Ribeiro DA, Cury PMet al, 2008, FTY720 treatment in experimentally urethane-induced lung tumors. J Exp Ther Oncol 7:9–15 Monobe Y, Manabe T, 1995, Morphological changes and proliferative activity of alveolar epithelium in mouse lungs treated with urethane. Virchows Arch 425:583–588 Cha SW, Lee HJ, Cho MHet al, 2001, Role of corticosterone in ethyl carbamate-induced immunosuppression in female BALB/c mice. Toxicol Lett 119:173–181 Yokota K, Hattori A, Kunz HWet al, 1989, Experimental analysis of the effects of cyclosporine on the induction of epithelial tumors. Transplant Proc 21:3211–3214 Caracia RL, Coltera MD, Gown AM, 1989, Analysis of proliferative embedded tissues: comparison with flow cytometric analysis. Am J Pathol 134:733–739 Hall PA, Levison DA, Woods AL, Yu CWet al, 1990, Proliferating cell nuclear antigen, PCNA, immunolocalization in paraffin sections: an index of cell proliferation with evidence of deregulated expression in some neoplasm. J Pathol 162:285–294 Hung JJ, Wang CY, Huang MHet al, 2007, Prognostic factors in resected stage I non-small cell lung cancer with a diameter of 3 cm or less: visceral pleural invasion did not influence overall and disease-free survival. J Thorac Cardiovasc Surg 134:638–643 Radovic S, Babic M, Doric Met al, 2007, Non-small cell lung carcinoma: cyclin D1, bcl-2, p53, Ki-67 and HER-2 proteins expression in resected tumors. Bosn J Basic Med Sci 7:205–211 Wang JD, Takahara S, Nonomura Net al, 1999, Early induction of apoptosis in androgen-independent prostate cancer cell line by FTY720 requires caspase-3 activation. Prostate 40:50–55 Malkinson AM, 2005, Role of inflammation in mouse lung tumorigenesis: a review. Exp Lung Res 31:57–82 Matsuoka Y, Nagahara Y, Ikekita M, Shinomiya T, 2003, A novel immunosuppressive agent FTY720 induced Akt dephosphorylation in leukemia cells. Br J Pharmacol 138:1303–1312 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 549 EP 554 DI 10.1007/s12253-009-9152-2 PG 6 ER PT J AU Ren, T Jiang, B Xing, X Dong, B Peng, L Meng, L Xu, H Shou, Ch AF Ren, Tingting Jiang, Beihai Xing, Xiaofang Dong, Bin Peng, Lirong Meng, Lin Xu, Huiyu Shou, Chengchao TI Prognostic Significance of Phosphatase of Regenerating Liver-3 Expression in Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PRL-3; Expression; Ovarian cancer; Immunohistochemistry; prognosis ID PRL-3; Expression; Ovarian cancer; Immunohistochemistry; prognosis AB Phosphatase of regenerating liver-3 (PRL-3) is overexpressed in several human cancers and associated with tumor progression, invasion and metastasis. However, the correlation between PRL-3 expression and clinical outcome in ovarian cancer has not been studied. In the present study, we investigated the expression of PRL-3 in 119 ovarian cancers and 30 normal ovarian tissues by immunohistochemistry with an anti-PRL-3 mouse monoclonal antibody 3B6, and analyzed its relationship with clinicopathologic factors and survival. The results demonstrated that PRL-3 expression was significantly higher in ovarian cancers compared to normal ovarian tissues (P<0.001). PRL-3 expression is not correlated with patient age, menstruation, tumor size, histological type, residual tumor, or other clinical findings. The patients with PRL-3-positive tumors had a significant poor prognosis than those with PRL-3-negative tumors. Univariate analysis identified PRL-3 expression as a poor outcome predictor (HR 1.925, 95% CI, 1.046–3.544, P=0.035). Multivariate analysis indicated that PRL-3 expression was an independent prognostic factor of borderline significance (HR 1.695, 95% CI, 0.914–3.145, P=0.094). Our results suggest that PRL-3 may serve as a valuable marker for diagnosis of ovarian cancer and as a potential independent prognostic factor for ovarian cancer. C1 [Ren, Tingting] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, 52 Fucheng Road, 100142 Beijing, China. [Jiang, Beihai] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, 52 Fucheng Road, 100142 Beijing, China. [Xing, Xiaofang] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, 52 Fucheng Road, 100142 Beijing, China. [Dong, Bin] Beijing Cancer Hospital & Institute, Core facilityBeijing, China. [Peng, Lirong] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, 52 Fucheng Road, 100142 Beijing, China. [Meng, Lin] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, 52 Fucheng Road, 100142 Beijing, China. [Xu, Huiyu] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, 52 Fucheng Road, 100142 Beijing, China. [Shou, Chengchao] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, 52 Fucheng Road, 100142 Beijing, China. RP Shou, Ch (reprint author), Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, 100142 Beijing, China. EM scc@bjcancer.org CR Cannistra SA, 2004, Cancer of the ovary. N Engl J Med 351:2519–2529 Zurawski VR Jr, Orjaseter H, Andersen A et al, 1988, Elevated serum CA 125 levels prior to diagnosis of ovarian neoplasia: relevance for early detection of ovarian cancer. Int J Cancer 42:677–680 Mastropaolo W, Fernandez Z, Miller EL, 1986, Pronounced increases in the concentration of an ovarian tumor marker, CA- 125, in serum of a healthy subject during menstruation. Clin Chem 32:2110–2111 Takahashi K, Shibukawa T, Moriyama M, 1986, Clinical usefulness and false-positive results of CA 125 as a tumor marker of ovarian cancer-a study on 674 patients. Jpn J Surg 16:305–310 Lyon M, Ducruet A, Wipf P, et al, 2002, Dual-specificity phosphates as targets for antineoplastic agents. Nat Rev Drug Discov 1:961–976 Zeng Q, Hong W, Tan YH, 1998, Mouse PRL-2 and PRL-3, two potentially prenylated protein tyrosine phosphatases homologous to PRL-1. Biochem Biophys Res Commun 244:421–427 Diamond RH, Cressman DE, Laz TM, et al, 1994, PRL-1, a unique nuclear protein tyrosine phosphatase, affects cell growth. Mol Cell Biol 14:3752–3762 Werner SR, Lee PA, DeCamp MW, et al, 2003, Enhanced cell cycle progression and down regulation of p21(Cip1/Waf1, by PRL tyrosine phosphatases. Cancer Lett 202:201–211 Polato F, Codegoni A, Fruscio R, et al, 2005, PRL-3 phosphatase is implicated in ovarian cancer growth. Clin Cancer Res 11:6835– 6839 Zeng Q, Dong JM, Guo K, et al, 2003, PRL-3 and PRL-1 promote cell migration, invasion, and metastasis. Cancer Res 63:2716–2722 Wu X, Zeng H, Zhang X, 2004, Phosphatase of regenerating liver-3 promotes motility and metastasis of mouse melanoma cells. Am J Pathol 23:23–27 Guo K, Li J, Tang JP, et al, 2004, Catalytic domain of PRL-3 plays an essential role in tumor metastasis: formation of PRL-3 tumors inside the blood vessels. Cancer Biol Ther 3:945–951 Li J, Guo K, Koh VW, et al, 2005, Generation of PRL-3- and PRL-1-specific monoclonal antibodies as potential diagnostic markers for cancer metastases. Clin Cancer Res 11:2195–2204 Guo K, Li J, Tang JP, et al, 2008, Monoclonal antibodies target intracellular PRL phosphatases to inhibit cancer metastases in mice. Cancer Biol Ther 7 Peng L, Li Y, Meng L, et al, 2004, Preparation and characterization of monoclonal antibody against protein tyrosine phosphatase PRL-3. Hybrid Hybridomics 23:23–27 Peng L, Ning J, Meng L, et al, 2004b, The association of the expression level of protein tyrosine phosphatase PRL-3 protein with liver metastasis and prognosis of patients with colorectal cancer. J Cancer Res Clin Oncol 130:521–526 Wang L, Peng L, Dong B, et al, 2006, Overexpression of phosphatase of regenerating liver-3 in breast cancer: association with a poor clinical outcome. Ann Oncol 17:1517–1522 Wang Y, Li ZF, He J, et al, 2007, Expression of the human phosphatases of regenerating liver, PRLs, in colonic adenocarcinoma and its correlation with lymph node metastasis. Int J Colorectal Dis 22:1179–1184 Miskad UA, Semba S, Kato H, Yokozaki H, 2004, Expression of PRL-3 phosphatase in human gastric carcinomas: close correlation with invasion and metastasis. Pathobiology 71:176–184 Miskad UA, Semba S, Kato H, et al, 2007, High PRL-3 expression in human gastric cancer is a marker of metastasis and grades of malignancies: an in situ hybridization study. Virchows Arch 450:303–310 Stephens B, Han H, Gokhale V, et al, 2005, PRL phosphatases as potential molecular targets in cancer. Mol Cancer Ther 4:1653– 1661 Han H, Bearss DJ, Browne LW, et al, 2002, Identification of differentially expressed genes in pancreatic cancer cells using cDNA microarray. Cancer Res 62:2890–2896 Stephens B, Han H, Hostetter G, et al, 2008, Small interfering RNA-mediated knockdown of PRL phosphatases results in altered Akt phosphorylation and reduced clonogenicity of pancreatic cancer cells. Mol Cancer Ther 7:202–210 Kato H, Semba S, Miskad UA, et al, 2004, High expression of PRL-3 promotes cancer cell motility and liver metastasis in human colorectal cancer: a predictive molecular marker of metachronous liver and lung metastases. Clin Cancer Res 10:7318–7328 Radke I, Gotte M, Kersting C, et al, 2006, Expression and prognostic impact of the protein tyrosine phosphatases PRL-1, PRL-2, and PRL-3 in breast cancer. Br J Cancer 95:347–354 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 555 EP 560 DI 10.1007/s12253-009-9153-1 PG 6 ER PT J AU Bekasi, S Zalatnai, A AF Bekasi, Sandor Zalatnai, Attila TI Overexpression of Glucocorticoid Receptor in Human Pancreatic Cancer and in Xenografts. An Immunohistochemical Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic cancer; Glucocorticoid receptor; Immunohistochemistry; Xenograft ID Pancreatic cancer; Glucocorticoid receptor; Immunohistochemistry; Xenograft AB Glucocorticoid receptor overexpression has been reported in a variety of human solid tumors, but much less in known about its presence in pancreatic cancer. Only one report is available in the literature, back to 1994, since that no peculiar attention has been paid to this issue. Immunohistochemical analysis of paraffin-embedded tissue sections was performed in human normal pancreata and well differentiated pancreatic adenocarcinomas (monoclonal primary antibody, ABCAM, Cambridge, UK). As positive control invasive ductal adenocarcinoma of the breast was used. In the normal non-tumorous pancreas a strong positivity was detected in all acinar cells, typically in the cytoplasm. Nuclear staining was not visible. The distribution of the positive reaction was homogenous. The ductal pancreatic carcinoma cells also displayed a strong positivity. The location of the immune reaction was mainly cytoplasmic but in some tumors a strong nuclear reaction was also noticed. In some slides acini remained also positive in the close vicinity of the tumor. Although the positivity of the ductal tumor cells was a constant finding in our samples, surprisingly, the liver metastasis was completely negative. Strong glucocorticoid receptor expression was also found in xenografted human pancreatic cancer showing a diffuse, mainly cytoplasmic positivity. Our studies have shown that the human pancreatic carcinomas do overexpress a strong glucocorticoid receptor positivity, but its significance is not clear. However, this finding might have a clinical relevance. C1 [Bekasi, Sandor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. RP Zalatnai, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM zalatnai@korb1.sote.hu CR Wright AP, Zilliacus J, McEwan IJ et al, 1993, Structure and function of the glucocorticoid receptor. J Steroid Biochem Mol Biol 47:11–19 Schwartzman RA, Cidlowski JA, 1994, Glucocorticoid-induced apoptosis of lymphoid cells. Int Arch Allergy Immunol 105:347–354 McEwan IJ, Wright AP, Gustafsson JA, 1997, Mechanism of gene expression by the glucocorticoid receptor: role of protein-protein interactions. Bioessays 19:153–160 Drouin J, Sun YL, Tremblay S et al, 1992, Homodimer formation is rate-limiting for high affinity DNA binding by glucocorticoid receptor. Mol Endocrinol 6:1299–1309 Goulding NJ, 2004, The molecular complexity of glucocorticoid actions in inflammation—a four-ring circus. Curr Opin Pharmacol 4:629–636 Wintermantel TM, Berger S, Greiner EF et al, 2005, Evaluation of steroid receptor function by gene targeting in mice. J Steroid Biochem Mol Biol 93:107–112 Jenkins BD, Pullen CB, Darimont BD, 2001, Novel glucocorticoid receptor coactivator effector mechanisms. Trends Endocrinol Metab 12:122–126 Bhakoo HS, Milholland RJ, Lopez R et al, 1981, High incidence and characterization of glucocorticoid receptors in human malignant melanoma. J Natl Cancer Inst 66:21–25 Chaudhuri PK, Walker MJ, Beattie CW et al, 1981, Distribution of steroid hormone receptors in human soft tissue sarcomas. Surgery 90:149–153 Plager JE, Karakousis CP, 1985, Increased cytosol glucocorticoid receptor content of leiomyosarcoma. Cancer Treat Rep 69:907– 908 P’eng FK, Lui WY, Chang TJ et al, 1988, Glucocorticoid receptors in hepatocellular carcinoma and adjacent liver tissue. Cancer 62:2134–2138 Lui WY, P’eng FK, Chang TJ et al, 1993, Analysis of glucocorticoid receptors in human hepatocellular carcinoma and HepG2 cells. Hepatology 18:1167–1174 Wu CW, Wang SR, Chang TJ et al, 1989, Content of glucocorticoid receptor and arginase in gastric cancer and normal gastric mucosal tissues. Cancer 64:2552–2556 Yeh DC, Cheng SB, Yu CC et al, 2006, Role of glucocorticoid receptor in serosa-involved gastric carcinoma after gastrectomy. J Gastroint Surg 10:706–711 Theocharis S, Kouraklis G, Margeli A et al, 2003, Glucocorticoid receptor, GR, immunohistochemical expression is correlated with cell cycle-related molecules in human colon cancer. Dig Dis Sci 48:1745–1750 Mohler JL, Chen Y, Hamil K et al, 1996, Androgen and glucocorticoid receptors in the stroma and epithelium of prostatic hyperplasia and carcinoma. Clin Cancer Res 2:889–895 Yemelyanov A, Czwornog J, Chebotaev D et al, 2007, Tumor suppressor activity of glucocorticoid receptor in the prostate. Oncogene 26:1885–1896 Sommer P, Le Rouzic P, Gillinham H et al, 2007, Glucocorticoid receptor overexpression exerts an antisurvival effect on human small cell lung cancer cells. Oncogene 26:7111–7121 Lu YS, Lien HC, Yeh PY et al, 2006, Glucocorticoid receptor expression in advanced non-small cell lung cancer: clinicopathological correlation and in vitro effect of glucocorticoid on cell growth and chemosensitivity. Lung Cancer 53:303–310 Lien HC, Lu YS, Cheng AL et al, 2006, Differential expression of glucocorticoid receptor in human breast tissues and related neoplasms. J Pathol 209:317–327 Plese JP, Martins VR, Lopes MT et al, 1985, Steroid receptors in meningiomas. Arq Neuropsiquiatr 43:365–371 Zhao X, Teng LZ, Wang ZG et al, 2007, Expression and significance of glucocorticoid receptor alpha in meningiomas. J Clin Neurosci 14:359–363 Woenckhaus J, Franke FE, Hackethal A et al, 2006, Glucocorticosteroid receptors in ovarian carcinomas. Oncol Rep 15:1137–1140 Lee C, Kao H, Lin H et al, 1998, Estrogen receptors and glucocorticoid receptors in human well-differentiated thyroid cancer. Int J Mol Med 2:229–233 Okazaki SI, Yamakawa M, Maeda K et al, 2006, Expression of glucocorticoid receptors in non-neoplastic lymphoid follicles and B cell type malignant lymphomas. J Clin Pathol 59:410–416 Norman J, Franz M, Schiro R et al, 1994, Functional glucocorticoid receptor modulates pancreatic carcinoma growth through an autocrine loop. J Surg Res 57:33–38 Hofmann J, Kaiser U, Maasberg M et al, 1995, Glucocorticoid receptors and growth inhibitory effects of dexamethasone in human lung cancer cell lines. Eur J Cancer 31A:2053–2058 Osman AM, Jansen PW, Smets LA et al, 1985, Glucocorticoid receptors and cell cycle progression in human melanoma cell lines. J Cell Physiol 125:306–312 Reyes-Moreno C, Koutsilieris M, 1997, Glucocorticoid receptor function possibly modulates cell-cell interactions in osteoblastic metastases on rat skeleton. Clin Exp Metastasis 15:205–217 Braunschweiger PG, Ting HL, Schiffer LM, 1983, Correlation between glucocorticoid receptor content and the antiproliferative effect of dexamethasone in experimental solid tumors. Cancer Res 43:4757–4761 Chebotaev D, Yemelyanov A, Budunova I, 2007, The mechanisms of tumor suppressor effect of glucocorticoid receptor in skin. Mol Carcinog 46:732–740 Guo WX, Antakly T, Cadotte M et al, 1996, Expression and cytokine regulation of glucocorticoid receptors in Kaposi’s sarcoma. Am J Pathol 148:1999–2008 Pang D, Kocherginsky M, Krausz T et al, 2006, Dexamethasone decreases xenograft response to Paclitaxel through inhibition of tumor cell apoptosis. Cancer Biol Ther 5:933–940 Zhang C, Kolb A, Buchler P et al, 2006, Corticosteroid cotreatment induces resistance to chemotherapy in surgical resections, xenografts and established cell lines of pancreatic cancer. BMC Cancer 6:61 Herr I, Ucur E, Herzer K et al, 2003, Glucocorticoid cotreatment induces apoptosis resistance toward cancer therapy in carcinomas. Cancer Res 63:3112–3120 Zhang C, Kolb A, Mattern J et al, 2006, Dexamethasone desensitizes hepatocellular and colorectal tumours toward cytostatic therapy. Cancer Lett 242:104–111 Zhang C, Beckermann B, Kallifatidis G et al, 2006, Corticosteroids induce chemotherapy resistance in the majority of tumour cells from bone, brain, breast, cervix, melanoma and neuroblastoma. Int J Oncol 29:1295–1301 Zhang C, Marme A, Wenger T et al, 2006, Glucocorticoidmediated inhibition of chemotherapy in ovarian carcinomas. Int J Oncol 28:551–558 Zhang C, Wenger T, Mattern J et al, 2007, Clinical and mechanistic aspects of glucocorticoid-induced chemotherapy resistance in the majority of solid tumors. Cancer Biol Ther 6:278–287 Kaiser A, Stier U, Riecken EO et al, 1996, Glucocorticoid receptor concentration modulates glucocorticoid-regulated gene expression in rat pancreatic AR42J cells. Digestion 57:149–160 Logsdon CD, Moessner J, Williams JA et al, 1985, Glucocorticoids increase amylase mRNA levels, secretory organelles, and secretion in pancreatic acinar AR42J cells. J Cell Biol 100:1200– 1208 Evers BM, Thompson EB, Townsend CM Jr et al, 1993, Cortivazol increases glucocorticoid receptor expression and inhibits growth of hamster pancreatic cancer, H2T, in vivo. Pancreas 8:7–14 Eum WS, Li MZ, Sin GS et al, 2003, Dexamethasone-induced differentiation of pancreatic AR42J cell involves p21(waf1/cip1, and MAP kinase pathway. Exp Mol Med 35:379–384 Benz C, Hollander C, Miller B, 1986, Endocrine-responsive pancreatic carcinoma: steroid binding and cytotoxicity studies in human tumor cell lines. Cancer Res 46:2276–2281 Egberts JH, Schniewind B, Patzold M et al, 2008, Dexamethasone reduces tumor recurrence and metastasis after pancreatic tumor resection in SCID mice. Cancer Biol Ther 7:1044–1050 Kim KP, Kim NH, Kim JC et al, 2006, Diagnostic criteria for autoimmune chronic pancreatitis revisited. World J Gastroenterol 12:2487–2496 Sutton R, 2005, Autoimmune pancreatitis—also a Western disease. Gut 54:581–583 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 561 EP 566 DI 10.1007/s12253-009-9154-0 PG 6 ER PT J AU Blazsek, J Dobo Nagy, Cs Blazsek, I Varga, R Vecsei, B Fejerdy, P Varga, G AF Blazsek, Jozsef Dobo Nagy, Csaba Blazsek, Istvan Varga, Rita Vecsei, Balint Fejerdy, Pal Varga, Gabor TI Aminobisphosphonate Stimulates Bone Regeneration and Enforces Consolidation of Titanium Implant into a New Rat Caudal Vertebrae Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE New implant osseointegration model; Osteo-neogenesis; Aminobisphosphonates (Zometa®); Bone regeneration; Micro computer tomography (micro-CT); Biomechanical measurement ID New implant osseointegration model; Osteo-neogenesis; Aminobisphosphonates (Zometa®); Bone regeneration; Micro computer tomography (micro-CT); Biomechanical measurement AB Bisphosphonates are widely used as therapeutic agents in bone disorders including cancer metastasis due to their osteoclast inhibitory effect. Recent data shows that bisphosphonates may also induce bone-building by stimulating osteoblast activity. Clinical observations, however, have revealed that bisphosphonates may cause necrosis in the oral cavity which questions their usefulness in bone regeneration during the consolidation of inorganic implants. Here we report the investigation of bone neogenesis following chronic amine bisphosphonate (Zometa®) treatment in a novel experimental model, using the rat tail vertebra as a support. This method involves (1) implantation of titan screw into the tail vertebrae, (2) systemic bisphosphonate treatment and (3) quantitative biophysical measurements which mirrors consolidation of implant, i.e. strength of fixation and changes in newly formed bone architecture using micro Computer Tomograph (micro-CT). The degree of fixation of titan implants (osseointegration) increased by 36% on the effect of Zometa and the structure of newly formed bone became robust. The mass of new bone increased 3.1-fold at 6 weeks of regeneration, as compared to controls. Thus, Zometa®, a potent aminobisphosphonate used in therapy of cancer metastases, osteoporosis and bone marrow transplantation, significantly increased bone neogenesis and enforced osseointegration of titan implants as measured quantitatively in the rat tail vertebra. Our data support the usefulness of aminobisphosphonates in the rehabilitation of bone loss as well as in improvement osseointegration of implants. We emphasise that this novel method may open up new possibilities for screening the effects of local and systemic treatments. C1 [Blazsek, Jozsef] Semmelweis University, Department of Oral Biology, Nagyvarad ter 4, 1089 Budapest, Hungary. [Dobo Nagy, Csaba] Semmelweis University, Department of ProsthodonticsBudapest, Hungary. [Blazsek, Istvan] Paul-Brousse Hospital, ICIG and Universite de Paris-Sud, INSERM U602 and U972Paris, France. [Varga, Rita] Full-Tech CompanyBudapest, Hungary. [Vecsei, Balint] Semmelweis University, Department of ProsthodonticsBudapest, Hungary. [Fejerdy, Pal] Semmelweis University, Department of ProsthodonticsBudapest, Hungary. [Varga, Gabor] Semmelweis University, Department of Oral BiologyBudapest, Hungary. RP Blazsek, J (reprint author), Semmelweis University, Department of Oral Biology, 1089 Budapest, Hungary. EM blajozs@net.sote.hu CR Ducy P, Schinke T, Karsenty G, 2000, The osteoblast: a sophisticated fibroblast under central surveillance. Science 289:1501–1504 Teitelbaum SL, 2000, Bone resorption by osteoclasts. Science 289:1504–1508 Skillington J, Choy L, Derynck R, 2002, Bone morphogenetic protein and retinoic acid signalling cooperate to induce osteoblast differentiation of preadipocytes. J Cell Biol 159:135–146 Ichida F, Nishimura R, Hata K, Matsubara T, Ikeda F, Hisada K, Yatani H, Cao X, Komori T, Yamaguchi A, Yoneda T, 2004, Reciprocal roles of Msx2 in regulation of osteoblast and adipocyte differentiation. J Biol Chem 279:34015–34022 Hauge EM, Ovesel D, Eriksen EF, Mosekilde L, Melsen F, 2001, Cancellous bone remodelling occurs in specialized compartments lined by cells expressing osteoblastic markers. J Bone Miner Res 16:1575–1582 Eriksen EF, Eghbali-Fatourechi GZ, Khosla S, 2007, Remodeling and vascular spaces in bone. J Bone Miner Res 22:1–6 Kim JB, Leucht P, Luppen CA, Park YJ, Beggs HE, Damsky CH, Helms JA, 2007, Reconciling the roles of FAK in osteoblast differentiation, osteoclast remodeling, and bone regeneration. Bone 41:39–51 Zaidi M, 2007, Skeletal remodeling in health and disease. Nature Med 13:791–798 Guise TA, Yin JJ, Taylor SD, Kumagai Y, Dallas M, Boyce BF, Yoneda T, Mundy GR, 1996, Evidence for a causal role of parathyroid hormone-related protein in the pathogenesis of human breast cancer-mediated osteolysis. J Clin Invest 98:1544–1549 Rodan GA, Martin TJ, 2000, Therapeutic approaches to bone diseases. Science 289:1508–1514 Michaelson MD, Smith MR, 2005, Bisphosphonates for treatment and prevention of bone metastases. J Clin Oncol 23:8219–8223 Boissier S, Ferreras M, Peyruchaud O, 2000, Bisphosphonates inhibit breast and prostate carcinoma cell invasion, an early event in the formation of bone metastases. Cancer Res 60:2949–2954 Zhao Z, Wang Z, Ge C, Krebsbach P, Franceschi RT, 2007, Healing cranial defects with AdRunx2-transduced marrow stromal cells. J Dent Res 86:1207–1211 Laino G, d’Aquino R, Graziano A, Lanza V, Carinci F, Naro F, Pirozzi G, Papaccio G, 2005, A new population of human adult dental pulp stem cells: a useful source of living autologous fibrous bone tissue, LAB). J Bone Miner Res 20:1394–1402 Ohazama A, Modino SA, Miletich I, Sharpe PT, 2004, Stem-cellbased tissue engineering of murine teeth. J Dent Res 83:518–522 Branemark PI, Zarb GA, Albrektsson T, 1985, Tissue-integrated prostheses In: Osseointegration in Clinical Dentistry. Quintessence, Chicago, pp 211–232 Lundborg G, Besjakov J, Branemark PI, 2007, Osseointegrated wrist-joint prosthesis: a 15-year follow-up with focus on bony fixation. Scand J Plast Reconstr Surg Hand Surg 41:130–137 Evans EA, Calderwood DA, 2007, Forces and bond dynamics in cell adhesion. Science 316:1148–1153 Wang DA, Varghese S, Sharma B, Strehin I, Fermanian S, Gorham J, Fairbrother H, Cascio B, Elisseeff JH, 2007, Multifunctional chonroitin sulphate for cartilage tissuebiomaterial integration. Nature Materials 6:385–392 Luckman SP, Hughes DE, Coxon FP, Russell RGG, Rogers MJ, 1998, Nitrogen-containing bisphosphonates inhibit the mevalonate pathway and prevent post-translational prenylation of GTP-binding proteins, including Ras. J Bone Mineral Res 13:581–589 Fisher JE, Rogers MJ, Halasy JM, Luckman SP, Hughes DE, Masarachia PJ, Wesolowski G, Reszka AA, 1999, Alendronate mechanism of action: Geranylgeraniol, an intermediate in the mevalonate pathway, prevents inhibition of osteoclast formation, bone resorption, and kinase activation in vitro. Proc Nat Acad Sci USA 96:133–138 Van Beek E, Lowik C, Van Der Pluijm G, Papapoulos S, 1999, The role of geranylgeranylation in bone resorption and its suppression by bisphosphonates in fetal bone explants in vitro: a clue to the mechanism of action of nitrogen-containing bisphosphonates. J Bone Mineral Res 14:722–729 Lee YP, Schwarz EM, Davies M, 2002, Use of zoledronate to treat osteoblastic versus osteolytic lesions in a severe-combinedimmunodeficient mouse model. Cancer Res 62:5564–5570 Merigo E, Manfredi M, Meleti M, Corradi D, Vescovi P, 2005, Jaw bone necrosis without previous dental extractions associated with the use of bisphosphonates, pamidronate and zoledronate). J Oral Pathol 34:613–614 Farrugia MC, Summerlin DJ, Krowiak E, Huntley T, Freeman S, Borrowdale R, Tomich C, 2006, Osteonecrosis of the mandible or maxilla associated with the use of new generation bisphosphonates. Laryngoscope 116:115–120 Khosla S, Burr D, Cauley J, Dempster DW, Ebeling PR, Felsenberg D, Gagel RF, Gilsanz V, Guise T, Koka S, McCauley LK, McGowan J, McKee MD, Mohla S, Pendrys DG, Raisz LG, Ruggiero SL, Shafer DM, Shum L, Silverman SL, Van Poznak CH,Watts N,Woo SB, Shane E, 2007, Bisphosphonate-associated osteonecrosis of the jaw: report of a task force of the American Society for Bone and Mineral Research. J Bone Miner Res 22:1479–1491 Reinholz GG, Getz B, Pederson L, Sanders ES, Subramaniam M, Ingle JN, Spelsberg TC, 2000, Bisphosphonates directly regulate cell proliferation, differentiation, and gene expression in human osteoblasts. Cancer Res 60:6001–6007 Fromigue O, Body JJ, 2002, Bisphosphonates influence the proliferation and maturation of normal human osteoblasts. J Endocrinol Invest 25:539–546 von Knoch F, Jaquiery C, Kowalsky M, Schaeren S, Alabre C, Martin I, Rubash HE, Shanbhag AS, 2005, Effects of bisphosphonates on proliferation and osteoblast differentiation of human bone marrow stromal cells. Biomaterials 34:6941–6949 Tauchmanova L, Ricci P, Serio B, Lombardi G, Colao A, Rotoli B, Selleri C, 2005, Short-term Zoledronic acid treatment increases bone mineral density and marrow clonogenic fibroblast progenitors after allogeneic stem cell transplantation. J Clin Endocrinol Metab 90:627–634 Ysander M, Branemark R, Olmarker K, Myers RR, 2001, Intramedullary osseointegration: development of a rodent model and study of histology and neuropeptide changes around titanium implants. J Rehabilit Res Dev 38:183–190 Naresh KN, Lampert I, Haserjian R, Lykiis D, Elerfiel K, Horncastle D, Smith N, Murray-Brown W, Stamp GW, 2006, Optimal processing of bone marrow trephine biopsy: the Hammersmith protocol. J Clin Pathol 59:903–911 Kaufman MH, 1992, The atlas of mouse development. Academic, Harcourt Brace & Company, London Saulacic N, Iizuka T, Martin MS, Garcia AG, 2008, Alveolar distraction osteogenesis: a systematic review. Int J Oral Maxillofac Surg 37:1–7 Shubayev VI, Branemark R, Steinauer J, Myers RR, 2004, Titanium implants induce expression of matrix metalloproteases in bone during osseointegration. J Rehab Res Dev 41:757–766 Sato M, Grasser W, Endo N, Akins R, Simmons H, Thompson DD, Golub E, Rodan GA, 1991, Bisphosphonate action. Alendronate localization in rat bone and effects on osteoclast ultrastructure. J Clin Invest 88:2095–2105 Coxon FP, Helfrich MH, Van’t Hof R, 2000, Protein geranylgeranylation is required for osteoclast formation, function, and survival: Inhibition by bisphosphonates and GGTI-298. J Bone Miner Res 15:1467–1476 Amital H, Applbaum YH, Vasiliev L, Rubinow A, 2004, Hypertrophic pulmonary osteoarthropathy: control of pain and symptoms with pamidronate. Clin Rheumatol 23:330–332 Chapurlat RD, Hugueny P, Delmas PD, Meunier PJ, 2004, Treatment of fibrous dysplasia of bone with intravenous pamidronate: long-term effectiveness and evaluation of predictors of response to treatment. Bone 35:235–242 Amant N, McDonald M, Godfrey C, Bilston L, Little D, 2007, Optimal timing of a single dose of zoledronic acid to increase strength in rat fracture repair. J Bone Miner Res 22:867–876 Hashimoto T, Shigetomi M, Ohno T, Matsunaga T, Muramatsu K, Tanaka H, Sugiyama T, Taguchi T, 2007, Sequential treatment with intermittent low-dose human parathyroid hormone, 1–34, and bisphosphonate enhances large-size skeletal reconstruction by vascularized bone transplantation. Calcif Tissue Int 81(3):232–239 Mundy GR, Yoneda T, Hiraga T, 2001, Preclinical studies with zoledronic acid and other bisphosphonates: Impact on the bone microenvironment. Semin Oncol 28(6):35–44 Blazsek I, Chagraoui J, Peault B, 2000, Ontogenic emergence of the hematon, a morphogenic stromal unit that supports multipotential hemopoietic progenitors in mouse bone marrow. Blood 96:3763–3771 Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, Martin RP, Schipani E, Divieti P, Bringhurst FR, Milner LA, Kronenberg HM, Scadden D, 2003, Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425:841–846 Blazsek I, Goldschmidt E, Machover D, Misset JL, Benavides M, Comisso M, Ceresi E, Canon C, Labat ML, Mathe G, 1986, Excess of lympho-reticular cell complexes in the bone marrow linked to T cell mediated dysmyelopoiesis. Biomed Pharmacother 40:28–32 Epling-Burnette PK, Painter JS, Rollison DE, Ku E, Vendron D, Widen R, Boulware D, Zou JX, Bai F, List AF, 2007, Prevalence and clinical association of clonal T-cell expansion in Myelodysplastic Syndrome. Leukemia 21:659–667 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 567 EP 577 DI 10.1007/s12253-009-9156-y PG 11 ER PT J AU Pap, Zs Pavai, Z Denes, L Kovalszky, I Jung, J AF Pap, Zsuzsanna Pavai, Zoltan Denes, Lorand Kovalszky, Ilona Jung, Janos TI An Immunohistochemical Study of Colon Adenomas and Carcinomas: E-cadherin, Syndecan-1, Ets-1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon adenoma; Colon carcinoma; E-cadherin; Ets-1; Immunohistochemistry; Syndecan-1 ID Colon adenoma; Colon carcinoma; E-cadherin; Ets-1; Immunohistochemistry; Syndecan-1 AB It is thought that dysregulation of E-cadherin, syndecan-1 (CD138) and Ets-1 is involved in carcinoma development. E-cadherin is an important epithelial cell adhesion molecule; syndecan-1 (CD138) is a regulatory proteoglycan in both cell-cell and cell-matrix adhesion and Ets-1 is a proto-oncogene and transcription factor, which takes part in extracellular matrix remodeling. Our goal was to study the changes in the expression of these molecules during colon carcinoma development and progression. We tested 117 colon adenomas and 149 de novo and ex adenoma carcinomas of the colon, using the Ultravision Polymer system. The positive reaction rate was 100% for E-cadherin, 98.3% for syndecan-1 and 22.4% for Ets-1 in adenomas, while in carcinomas it was 88.5%, 62.4% and 56.3% respectively. We found decreasing expression of E-cadherin and syndecan-1 throughout colon carcinoma progression and an opposite regulation for the Ets-1 protein. Decrease in expression of syndecan-1 is more pronounced in carcinomas compared to E-cadherin. De novo carcinomas have lower E-cadherin and syndecan-1 expression, and higher Ets-1 expression compared to ex adenoma carcinomas. These findings support the hypothesis that there are differences in the carcinogenesis of these tumors. C1 [Pap, Zsuzsanna] University of Medicine and Pharmacy Targu-Mures, Department of Anatomy and EmbryologyTargu-Mures, Romania. [Pavai, Zoltan] University of Medicine and Pharmacy Targu-Mures, Department of Anatomy and EmbryologyTargu-Mures, Romania. [Denes, Lorand] University of Medicine and Pharmacy Targu-Mures, Department of Anatomy and EmbryologyTargu-Mures, Romania. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Jung, Janos] University of Medicine and Pharmacy Targu-Mures, Department of PathologyTargu-Mures, Romania. RP Pap, Zs (reprint author), University of Medicine and Pharmacy Targu-Mures, Department of Anatomy and Embryology, Targu-Mures, Romania. EM papzsuzsa@yahoo.com CR Valean S, Mircea PA, Oprea L, 2006, Trends of mortality rates from gastric cancer and colorectal cancer in Romania, 1955–2003. J Gastrointestin Liver Dis 15(2):111–115 Hamilton RS, Aaltonen AL, 2000, Pathology and genetics, toumors of the digestive system. IARC Press, Lyon Pecina-Slaus N, 2003, Tumor suppressor gene E-cadherin and its role in normal and malignant cells. Cancer Cell Int 3:17 Day MR, Hao X, Mohammad I, Daszak P, Talbot IC, Forbes A, 1999, Changes in the expression of syndecan-1 in the colorectal adenoma-carcinoma sequence. Virchows Arch 434:121–125 Beauvais DM, Rapraeger CA, 2004, Syndecans in tumor cell adhesion and signaling. RB&E 2:3 http://www.rbej.com/content/2/1/3 Nakayama T, Masahiro I, Akira O, Shinji N, Ichiro S, 2001, Expression of the ets-1 Proto-Oncogene in human colorectal carcinoma. Mod Pathol 14(5):415–422 Arun S, Watson KD, 2005, Ets transcription factors and their emerging roles in human cancer. Eur J Cancer 41:2462–2478 Nosho K, Yoshiday M, Yamamoto M, Taniguchi H, Adachi Y, Mikami M, Hinodal Y, Imai K, 2005, Association of Ets-related transcriptional factor E1AF expression with overexpression of matrix metalloproteinases, COX-2 and iNOS in the early stage of colorectal carcinogenesis. Carcinogenesis 26(5):892–899 Behrens P, Mathiak M, Mangold E, Kirdorf S, Wellmann A, Fogt F, Rothe M, Florin A, Wernert N, 2003, Stromal expression of invasion-promoting matrix-degrading proteases MMP-1 and -9 and the Ets-1 transcription factor in HNPCC carcinomas and sporadic colorectal cancers. Int J Cancer 107:183–188 Khoursheed MA, Mathew TC, Makar RR, Louis S, Asfar SK, Al- Sayer HM, Dashti HM, Al-Bader A, 2003, Expression of E-cadherin in human colorectal cancer. Surg J R Coll Surg Edinb Irel 86–91 Elzagheid A, Algars A, Bendardaf R, Lamlum H, Ristamaki R, Collan Y, Syrjanen K, Pyrhonen S, 2006, E-cadherin expression pattern in primary colorectal carcinomas and their metastases reflects disease outcome. World J Gastroenterol 12(27):4304– 4309 Fujiya M, Watari J, Ashida T, Honda M, Tanabe H, Fujiki T, Saitoh Y, Kohgo Y, 2001, Reduced expression of syndecan-1 affects metastatic potential and clinical outcome in patients with colorectal cancer. Jpn. J Cancer Res 192:1074–1081 Lundin M, Nordling S, Lundin J, Isola J, Wiksten JP, Haglund C, 2005, Epithelial syndecan-1 expression is associated with stage and grade in colorectal cancer. Oncology 68:306–313 Rosai J, 2004, Rosai and Ackerman`s surgical pathology, 9th edn. Mosby, London Geller SA, Dhall D, Alsabeh R, 2008, Application of immunohistochemistry to liver and gastrointestinal neoplasms: liver, stomach, colon, and pancreas. Arch Pathol Lab Med 132:490– 499 Wlodarczyk J, Bethke B, Mueller E, Stolte M, Mueller J, 2001, A comparative study of E-cadherin and stromelysin-3 expression in de novo and ex adenoma carcinoma of the colorectum. Virchows Arch 439:756–761 Batistatou A, Charalabopoulos AK, Scopa CD, Nakanishi Y, Kappas A, Hirohashi S, Agnantis NJ, Charalabopoulos K, 2006, Expression patterns of dysadherin and E-cadherin in lymph node metastases of colorectal carcinoma. Virchows Arch 448:763–767 Peretti T, Waisberg J, Mader AM, Matos L, Costa R, Conceicao G, Lopes A, Nader H, Pinhal M, 2008, Heparanase-2, syndecan- 1, and extracellular matrix remodeling in colorectal carcinoma. Eur J Gastroenterol & Hepatol 20:756–765 Gottardi JC, Wong E, Gumbiner MB, 2001, E-Cadherin suppresses cellular transformation by inhibiting β-catenin signaling in an adhesion-independent manner. J Cell Biol 153(5):1049–1059 Qinglang L, Pyong WP, Wilson LC, Parks CW, 2002, Matrilysin shedding of syndecan-1 regulates Chemokine mobilization and transepithelial efflux of neutrophils in acute lung injury. Cell 111:635–646 Fitzgerald LM, Wang Z, Park WP, Murphy G, Bernfield M, 2000, Shedding of syndecan-1 and -4 ectodomains is regulated by multiple signaling pathways and mediated by a TIMP-3-sensitive metalloproteinase. J. Cell Biol 148(4):811–824 Hashimoto Y, Skacel M, Adams JC, 2008, Association of loss of epithelial syndecan-1 with stage and local metastasis of colorectal adenocarcinomas: an immunohistochemical study of clinically annotated tumors. BMC Cancer 8(185):1–7 Kwak JM, Min BW, Lee JH, Choi JS, Lee SI, Park SS, Kim J, Um JW, Kim SH, Moon HY, 2007, The prognostic significance of Ecadherin and liver intestine-cadherin expression in colorectal cancer. Dis Colon Rectum 50:1873–1880 Dilek F, Topak N, Aktepe F, Sahin O, Turel KS, Sahin DA, Dilek ON, 2008, E-cadherin, β-catenin adhesion complex and relation to matrilysin expression in pT3 rectosigmoid cancers. Pathol Res Practice 204(11):809–815 Tetsuji T, Koji M, Tsuyoshi H, 2006, Colorectal cancer: genetics of development and metastasis. J Gastroenterology 41:185–192 Ortega P, Moran A, de Juan C, Frias C, Hernandez S, Lopez-Asenjo J-A, Sanchez-Pernaute A, Torres A, Iniesta P, Benito M, 2008, DifferentialWnt pathway gene expression and E-cadherin truncation in sporadic colorectal cancers with and without microsatellite instability. Clin Cancer Res 14(4):995–1001 Jurgen D, 2003, The biology of the Ets1 proto-oncogene. Mol Cancer 2:29 http://www.molecular-cancer.com/content/2/1/29 McGuire KJ, Qinglang L, Parks CW, 2003, Matrilysin, Matrix Metalloproteinase-7, mediates E-Cadherin ectodomain shedding in injured lung epithelium. Am J Pathol 162(6):1831–1843 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 579 EP 587 DI 10.1007/s12253-009-9157-x PG 9 ER PT J AU Zhao, ZSh Wang, YY Ye, ZY Tao, HQ AF Zhao, Zhong-Sheng Wang, Yuan-Yu Ye, Zai-Yuan Tao, Hou-Quan TI Prognostic Value of Tumor-Related Molecular Expression in Gastric Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Basic fibroblast growth factor; Matrix metalloproteinase-9; Urokinase plasminogen activator; Prognosis; Molecular markers ID Gastric cancer; Basic fibroblast growth factor; Matrix metalloproteinase-9; Urokinase plasminogen activator; Prognosis; Molecular markers AB In order to identify reliable molecular markers for prognostic prediction in gastric carcinoma, we evaluated the expression of six molecular markers, namely bFGF, IGF-2, HGF, MMP-9, integrin β3 and uPA in gastric cancer. There was a significant correlation between the expression of these markers and the depth of tumor invasion, vessel invasion, lymph node and distant metastasis, TNM stage and microvessel density. The average survival time and 5-year survival rate of patients with positive expression of molecular markers was higher than those with negative expression. Multivariate analysis showed that abnormal expression of bFGF, MMP-9 and uPA, as well as depth of invasion, lymph node and distant metastasis and TNM stage were independently related to poor prognosis of gastric cancer. MMP-9, bFGF and uPA are potential candidates for development as clinically applicable molecular prognostic markers for gastric carcinoma, and may be effective therapeutic targets for the disease in the future. C1 [Zhao, Zhong-Sheng] Zhejiang Provincial People’s Hospital, Department of Pathology, 158 shangtang road, 310014 Hangzhou, China. [Wang, Yuan-Yu] Zhejiang Provincial People’s Hospital, Department of General SurgeryHangzhou, China. [Ye, Zai-Yuan] Zhejiang Provincial People’s Hospital, Department of General SurgeryHangzhou, China. [Tao, Hou-Quan] Zhejiang Provincial People’s Hospital, Department of General SurgeryHangzhou, China. RP Zhao, ZSh (reprint author), Zhejiang Provincial People’s Hospital, Department of Pathology, 310014 Hangzhou, China. EM lywyy1979@hotmail.com CR Inoue M, Tsugane S, 2005, Epidemiology of gastric cancer in Japan. Postgrad Med J 81:419–424 Wenhua Z, Fanghai H, 2008, Surgical therapy of gastric cancer in china. Journal of Practical oncology 23:91–93 Egeblad M, Werb Z et al, 2002, New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2:161– 174 Moon WS, Park HS, Yu KH et al, 2006, Expression of angiopoietin 1, 2 and their common receptor Tie2 in human gastric carcinoma: in implication for angiogenesis. Journal of Korean Medical Science 21:272–278 Niedergethmann M, Hildenbrand R, Wostbrock B et al, 2002, High expression of vascular endothelial growth factor predicts early recurrence and poor prognosis after curative resection for ductal adenocarcinoma of the pancreas. Pancreas 25:122–129 Kolev Y, Uetake H, Iida S et al, 2007, Prognostic significance of VEGF expression in correlation with COX-2, microvessel density, and clinicopathological characteristics in human gastric carcinoma. Annals of Surgical Oncology 14:2738–2747 Mizokami K, Kakeji Y, Oda S et al, 2006, Clinicopathologic significance of hypoxia-inducible factor 1 alpha overexpression in gastric carcinomas. Journal of Surgical Oncology 94:149–154 Weidner N, Semple JP, Welch WR, Folkman J, 1991, Tumor angiogenesis and metastasis-correlation in invasive breast carcinoma. N Engl J Med 324:1–8 Hundahl SA, Phillips JL, Menck HR, 2000, The National Cancer Data Base Report on poor survival of U.S. gastric carcinoma patients treated with gastrectomy: fifth edition American Joint Committee on cancer staging, proximal disease, and the ‘different disease’ hypothesis. Cancer 88:921–932 Park M, Park H, Kim WH, Cho H, Lee JH, 2005, Presence of autocrine hepatocyte growth factor-Met signaling and its role in proliferation and migration of SNU-484 gastric cancer cell line. Exp Mol Med 37:213–219 Niki M, Iso Zaki H, Toyoda M et al, 1999, Serum human hepatocyte growth factor is elevated in patients with metastatic gastric carcinoma. Hepatogastroenterology 46:568–573 Yi HK, Hwang PH, Yang DH, Kang CW, Lee DY, 2001, Expression of the insulin-like growth factors, IGFs, and the IGFbinding proteins, IGFBPs, in human gastric cancer cells. Eur J Cancer 37:2257–2263 Niu JX, Zhang WJ, Ye LY, 2007, The role of adhesion molecules, alpha v beta 3, alpha v beta 5 and their ligands in the tumor cell and endothelial cell adhesion. Eur J Cancer Prev 16:517–527 Vonlaufen A, Wiedle G, Borisch B, 2001, Integrin alpha(v, beta, 3, expression in colon carcinoma correlates with survival. Mod Pathol 14:1126–32 Zhang S, Li L, Lin JY, Lin H, 2003, Imbalance between expression of matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 in invasiveness and metastasis of human gastric carcinoma. World J Gastroenterol 9:899–904 Kaneko T, Konno H, Baba M, 2003, Urokinase-type plasminogen activator expression correlates with tumor angiogenesis and poor outcome in gastric cancer. Cancer Sci 94(1):43–49 Yang SF, Hsieh YS, Lin CL, 2005, Increased plasma levels of urokinase plasminogen activator and matrix metalloproteinase-9 in nonsmall cell lung cancer patients. Clin Chim Acta 354:91–99 Gao ZL, Zhang C, Du GY, Lu ZJ, 2007, Clinical significance of changes in tumor markers, extracellular matrix, MMP-9 and VEGF in patients with gastric carcinoma. Hepatogastroenterology 54:1591–1595 Zhao J, Liu XY, Zhang QY, Jiang W, 2005, Plasma level and prognostic significance of VEGF, bFGF and MMP-9 in patients with advanced non-small-cell lung cancer. Zhonghua Zhong Liu Za Zhi 27:676–679 Ramos-Desimone N, Haha-Dantona E, Siply J et al, 1999, Activation of matrix metalloproteinases 9 Via a converging plasmin/stromelysin-1 cascade enhances tumor cell invasion. J Biol Chem 274:13066–13076 Kim MH, 2003, Flavonoids inhibit VEGF/bFGF-induced angiogenesis in vitro by inhibiting the matrix-degrading proteases. J Cell Biochem 89:529–538 Lee SR, Guo SZ, Scannevin RH, 2007, Induction of matrix metalloproteinase, cytokines and chemokines in rat cortical astrocytes exposed to plasminogen activators. Neurosci Lett 417:1–5 Zhao Y, Lyons CE Jr, Xiao A, 2008, Urokinase directly activates matrix metalloproteinases-9: a potential role in glioblastoma invasion. Biochem Biophys Res Commun 369:1215–1220 Sapienza P, di Marzo L, Borrelli V, 2004, Basic fibroblast growth factor mediates carotid plaque instability through metalloproteinase-2 and -9 expression. Eur J Vasc Endovasc Surg 28:89–97 Rao JS, Gondi C, Chetty C, 2005, Inhibition of invasion, angiogenesis, tumor growth, and metastasis by adenovirusmediated transfer of antisense uPAR and MMP-9 in non-small cell lung cancer cells. Mol Cancer Ther 4:1399–1408 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 589 EP 596 DI 10.1007/s12253-009-9158-9 PG 8 ER PT J AU Fodor, A Molnar, ZsM Krenacs, L Bagdi, E Csomor, J Matolcsy, A Demeter, J AF Fodor, Aniko Molnar, Zsolt Miklos Krenacs, Laszlo Bagdi, Eniko Csomor, Judit Matolcsy, Andras Demeter, Judit TI Autoimmune Hemolytic Anemia as a Risk Factor of Poor Outcome in Patients with Splenic Marginal Zone Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Autoimmune hemolytic anemia; Splenic marginal zone lymphoma; Prognosis; Outcome ID Autoimmune hemolytic anemia; Splenic marginal zone lymphoma; Prognosis; Outcome AB Splenic marginal zone lymphoma is a rare disease, accounting for 1% of all lymphomas. We reviewed our single center experience of 13 patients with splenic marginal zone lymphoma (SMZL). Based on the prognostic model developed by Intergruppo Italiano Linfomi, 31% (4/13) of our patients had good, 38% (5/13) had intermediate and 31% (4/13) had a poor prognosis. The presence of two out of three prognostic factors (anemia, elevated LDH, low serum albumin) assignes the patient into the high risk category. In patients with anemia and an elevated LDH due to hemolysis, the outcome seems to be especially poor. Three out of 13 (23%) cases were complicated by autoimmune hemolytic anemia. All patients with autoimmune hemolytic anaemia (AIHA) died 7–28 months after the diagnosis. The mean follow-up time of those nine patients who are still alive is longer than 5 years (36–100 months). Patients with AIHA had significantly (p<0.001) worse survival than those without AIHA. The main finding of our study is that the presence of AIHA is an adverse prognostic factor in SMZL. C1 [Fodor, Aniko] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor utca 2/A, H-1083 Budapest, Hungary. [Molnar, Zsolt Miklos] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Krenacs, Laszlo] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Bagdi, Eniko] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Csomor, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Matolcsy, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Demeter, Judit] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor utca 2/A, H-1083 Budapest, Hungary. RP Demeter, J (reprint author), Semmelweis University, 1st Department of Internal Medicine, H-1083 Budapest, Hungary. EM demjud@bel1.sote.hu CR Dogan A, 2005, Modern histological classification of low grade B-cell lymphomas. Best Pract Res Clin Haematol 18:11–26 Schmid C, Kirkham N, Diss T, Isaacson PG, 1992, Splenic marginal zone cell lymphoma. Am J Surg Pathol 16:455–466 Melo JV, Robinson DS, Gregory C, Catovsky D, 1987, Splenic B cell lymphoma with “villous” lymphocytes in the peripheral blood: a disorder distinct from hairy cell leukemia. Leukemia 1:294–298 Isaacson PG, Matutes E, Burke M, Catovsky D, 1994, The histopathology of splenic lymphoma with villous lymphocytes. Blood 84:3828–3834 Matutes E, Oscier D, Montalban C et al, 2008, Splenic marginal zone lymphoma proposals for a revision of diagnostic, staging and therapeutic criteria. Leukemia 22:487–495 Arcaini L, Lazzarino M, Colombo N et al, 2006, Splenic marginal zone lymphoma: a prognostic model for clinical use. Blood 107:4643–4649 Thieblemont C, Felman P, Berger F et al, 2002, Treatment of splenic marginal zone B-cell lymphoma: an analysis of 81 patients. Clin Lymphoma 3:41–47 Thieblemont C, Felman P, Callet-Bauchu E et al, 2003, Splenic marginal-zone lymphoma: a distinct clinical and pathological entity. Lancet Oncol 4:95–103 Chacon JI, Mollejo M, Munoz E et al, 2002, Splenic marginal zone lymphoma: clinical characteristics and prognostic factors in a series of 60 patients. Blood 100:1648–1654 Parry-Jones N, Matutes E, Gruszka-Westwood AM et al, 2003, Prognostic features of splenic lymphoma with villous lymphocytes: a report on 129 patients. Br J Haematol 120:759–764 Camacho FI, Mollejo M, Mateo MS et al, 2001, Progression to large B-cell lymphoma in splenic marginal zone lymphoma: a description of a series of 12 cases. Am J Surg Pathol 25:1268– 1276 Dungarwalla M, Appiah-Cubi S, Kulkarni S et al, 2008, Highgrade transformation in splenic marginal zone lymphoma with circulating villous lymphocytes: the site of transformation influences response to therapy and prognosis. Br J Haematol 143:71–74 Epub 2008 Jul 30 Cualing H, Steele P, Zellner D, 2000, Blastic transformation of splenic marginal zone B-cell lymphoma. Arch Pathol Lab Med. 124:748–752 Gale R, Smith OP, Wood M, Mehta AB, 1992, Splenic lymphoma with villous lymphocytes complicated by autoimmune haemolytic anaemia. Lancet 340:1106 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 597 EP 603 DI 10.1007/s12253-009-9159-8 PG 7 ER PT J AU Kuncova, K Janda, A Kasal, P Zamecnik, J AF Kuncova, Klara Janda, Ales Kasal, Pavel Zamecnik, Josef TI Immunohistochemical Prognostic Markers in Intracranial Ependymomas: Systematic Review and Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ependymoma; Immunohistochemistry; Ki-67; Meta-analysis; MIB-1; prognosis ID Ependymoma; Immunohistochemistry; Ki-67; Meta-analysis; MIB-1; prognosis AB Distinction between grade II ependymomas and anaplastic ependymomas based on histopathological examination solely is problematic and, therefore, the management of intracranial ependymomas remains controversial. The aim of this study was to conduct a systematic review (SR) and meta-analysis (MA) of data published on immunohistochemical prognostic markers (IPM) in intracranial ependymomas (IE), and to establish an evidencebased perspective on their clinical value. Following the extensive search based on a strictly defined group of key words, 30 studies reporting results on IPM in IE were identified. Due to a pronounced inter-study heterogeneity, only 14 publications fulfilled the criteria for inclusion into SR. From the total of 67 immunohistochemical markers, 18 were found to correlate with prognosis. However, owing to inadequate data publishing, MA could be performed only with data on proliferation marker MIB-1 (Ki-67) from 5 publications, including 337 patients: The pooled hazard ratio for overall survival was 3.16 (95% confidence interval=1.96–5.09; p<0.001) implicating that patients suffering from tumors with higher immunohistochemical expression of MIB-1 had a significantly worse outcome. Marked inter-study heterogeneity and incomplete data publishing in primary studies significantly limited extent of the SR, and the possibility of performing MA. Although the prognostic impact of MIB-1 immunoexpression in IE could be confirmed, there remains lack of further reliable IPM that could be used in routine diagnosis. We encourage to search for new, useful markers, as well as to standardize lab-techniques and data interpretation algorithms across laboratories in order to increase data compatibility. C1 [Kuncova, Klara] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of PathologyPrague, Czech Republic. [Janda, Ales] Charles University, 2nd Medical School and University Hospital Motol, Department of Medical InformaticsPrague, Czech Republic. [Kasal, Pavel] Charles University, 2nd Medical School and University Hospital Motol, Department of Medical InformaticsPrague, Czech Republic. [Zamecnik, Josef] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of PathologyPrague, Czech Republic. RP Zamecnik, J (reprint author), Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Prague, Czech Republic. EM josef.zamecnik@lfmotol.cuni.cz CR McLendon RE, Wiestler OD, Kros JM, Korshunov A, Ng HK, 2007, Ependymoma. In: Louis DN, Wiestler OD, Cavanee WK, eds, Classification of the Tumours of the Central Nervous System. Lyon, International Agency for Research on Cancer, IARC) Foreman NK, Love S, Thorne R, 1996, Intracranial ependymomas: analysis of prognostic factors in a population-based series. Pediatr Neurosurg 24:119–25 Pollack IF, Gerszten PC, Martinez AJ, Lo KH, Shultz B, Albright AL, Janosky J, Deutsch M, 1995, Intracranial ependymomas of childhood: long-term outcome and prognostic factors. Neurosurgery 37:655–66 Bouffet E, Perilongo G, Canete A, Massimino M, 1998, Intracranial ependymomas in children: a critical review of prognostic factors and a plea for cooperation. Med Pediatr Oncol 30: 319–29; discussion 329–31 Robertson PL, Zeltzer PM, Boyett JM et al, 1998, Survival and prognostic factors following radiation therapy and chemotherapy for ependymomas in children: a report of the Children’s Cancer Group. J Neurosurg 88:695–703 Zamecnik J, Snuderl M, Eckschlager T et al, 2003, Pediatric intracranial ependymomas: prognostic relevance of histological, immunohistochemical, and flow cytometric factors. Mod Pathol 16:980–91 Kurt E, Zheng PP, Hop WC et al, 2006, Identification of relevant prognostic histopathologic features in 69 intracranial ependymomas, excluding myxopapillary ependymomas and subependymomas. Cancer 106:388–95 Bennetto L, Foreman N, Harding B et al, 1998, Ki-67 immunolabelling index is a prognostic indicator in childhood posterior fossa ependymomas. Neuropathol Appl Neurobiol 24:434–40 Khan KS, ter Riet G, Glanville J, Sowden AJ, Kleijnen J, 2001, Undertaking Systematic Reviews of Research on Effectiveness: CRD Guidelines for Those Carrying Out or Commissioning Reviews. CRD Report 4, NHS Centre for Reviews and Dissemination, University of York, York Parmar MK, Torri V, Stewart L, 1998, Extracting summary statistics to perform meta-analyses of the published literature for survival endpoints. Stat Med 17:2815–34 Machin D, Cheung YB, Parmar MK, 2006, Survival Analysis. A Practical Approach, 2nd edn. John Wiley, Chichester Riley RD, Abrams KR, Sutton AJ et al, 2003, Reporting of prognostic markers: current problems and development of guidelines for evidence-based practice in the future. Br J Cancer 88:1191–8 Riley RD, Heney D, Jones DR et al, 2004, A systematic review of molecular and biological tumor markers in neuroblastoma. Clin Cancer Res 10:4–12 Sutton AJ, Abrams KR, Jones DR, Sheldon TA, Song F, 2000, Methods for Meta-analysis in Medical Research. John Wiley and Sons, Ltd, Chichester Rawlings CE, Giangaspero F, Burger PC, Bullard DE, 1988, Ependymomas: a clinicopathologic study. Surg Neurol 29:271–81 Schroder R, Ploner C, Ernestus RI, 1993, The growth potential of ependymomas with varying grades of malignancy measured by the Ki-67 labelling index and mitotic index. Neurosurg Rev 16:145–50 Takeuchi H, Kubota T, Sato K, Llena JF, Hirano A, 2002, Epithelial differentiation and proliferative potential in spinal ependymomas. J Neurooncol 58:13–9 DerSimonian R, Laird N, 1986, Meta-analysis in clinical trials. Control Clin Trials 7:177–88 Tabori U, Ma J, Carter M et al, 2006, Human telomere reverse transcriptase expression predicts progression and survival in pediatric intracranial ependymoma. J Clin Oncol 24:1522–8 Verstegen MJ, Leenstra DT, Ijlst-Keizers H, Bosch DA, 2002, Proliferation- and apoptosis-related proteins in intracranial ependymomas: an immunohistochemical analysis. J Neurooncol 56:21–8 Wolfsberger S, Fischer I, Hoftberger R et al, 2004, Ki-67 immunolabeling index is an accurate predictor of outcome in patients with intracranial ependymoma. Am J Surg Pathol 28:914–20 Zamecnik J, Chanova M, Tichy M, Kodet R, 2004, Distribution of the extracellular matrix glycoproteins in ependymomas—an immunohistochemical study with follow-up analysis. Neoplasma 51:214–22 Shuangshoti S, Rushing EJ, Mena H, Olsen C, Sandberg GD, 2005, Supratentorial extraventricular ependymal neoplasms: a clinicopathologic study of 32 patients. Cancer 103:2598–605 Schiffer D, Cavalla P, Migheli A, Giordana MT, Chiado-Piat L, 1996, Bcl-2 distribution in neuroepithelial tumors: an immunohistochemical study. J Neurooncol 27:101–9 Schiffer D, Chio A, Giordana MT, Pezzulo T, Vigliani MC, 1993, Proliferating cell nuclear antigen expression in brain tumors, and its prognostic role in ependymomas: an immunohistochemical study. Acta Neuropathol, Berl, 85:495–502 Rezai AR, Woo HH, Lee M, Cohen H, Zagzag D, Epstein FJ, 1996, Disseminated ependymomas of the central nervous system. J Neurosurg 85:618–24 Ritter AM, Hess KR, McLendon RE, Langford LA, 1998, Ependymomas: MIB-1 proliferation index and survival. J Neurooncol 40:51–7 Roma AA, Prayson RA, 2006, Expression of cyclo-oxygenase-2 in ependymal tumors. Neuropathology 26:422–8 Prayson RA, 1998, Cyclin D1 and MIB-1 immunohistochemistry in ependymomas: a study of 41 cases. Am J Clin Pathol 110:629–34 Prayson RA, 1999, Clinicopathologic study of 61 patients with ependymoma including MIB-1 immunohistochemistry. Ann Diagn Pathol 3:11–8 Preusser M, Wolfsberger S, Czech T, Slavc I, Budka H, Hainfellner JA, 2005, Survivin expression in intracranial ependymomas and its correlation with tumor cell proliferation and patient outcome. Am J Clin Pathol 124:543–9 Preusser M, Wolfsberger S, Haberler C et al, 2005, Vascularization and expression of hypoxia-related tissue factors in intracranial ependymoma and their impact on patient survival. Acta Neuropathol, Berl, 109:211–6 Nagashima T, Hoshino T, Cho KG, Edwards MS, Hudgins RJ, Davis RL, 1988, The proliferative potential of human ependymomas measured by in situ bromodeoxyuridine labeling. Cancer 61:2433–8 Korshunov A, Sycheva R, Timirgaz V, Golanov A, 1999, Prognostic value of immunoexpression of the chemoresistancerelated proteins in ependymomas: an analysis of 76 cases. J Neurooncol 45:219–27 Korshunov A, Timirgaz V, Golanov A, 1999, Prognostic value of aberrant p53 immunoexpression for the recurrence of ependymoma: An analysis of 76 cases. Neuropathology 19:380–385 Korshunov A, Golanov A, Timirgaz V, 2001, p14ARF protein, FL-132, immunoreactivity in intracranial ependymomas and its prognostic significance: an analysis of 103 cases. Acta Neuropathol, Berl, 102:271–7 Korshunov A, Golanov A, Timirgaz V, 2002, Immunohistochemical markers for prognosis of ependymal neoplasms. J Neurooncol 58:255–70 Ho DM, Hsu CY, Wong TT, Chiang H, 2001, A clinicopathologic study of 81 patients with ependymomas and proposal of diagnostic criteria for anaplastic ependymoma. J Neurooncol 54:77–85 Guyotat J, Champier J, Jouvet A et al, 2001, Differential expression of somatostatin receptors in ependymoma: implications for diagnosis. Int J Cancer 95:144–51 Gilbertson RJ, Bentley L, Hernan R et al, 2002, ERBB receptor signaling promotes ependymoma cell proliferation and represents a potential novel therapeutic target for this disease. Clin Cancer Res 8:3054–64 Figarella-Branger D, Civatte M, Bouvier-Labit C et al, 2000, Prognostic factors in intracranial ependymomas in children. J Neurosurg 93:605–13 Figarella-Branger D, Gambarelli D, Dollo C et al., 1991, Infratentorial ependymomas of childhood. Correlation between histological features, immunohistological phenotype, silver nucleolar organizer region staining values and post-operative survival in 16 cases. Acta Neuropathol, Berl, 82: 208–16 Cruz-Sanchez FF, Garcia-Bachs M, Rossi ML et al, 1992, Epithelial differentiation in gliomas, meningiomas and choroid plexus papillomas. Virchows Arch B Cell Pathol Incl Mol Pathol 62:25–34 Asai A, Hoshino T, Edwards MS, Davis RL, 1992, Predicting the recurrence of ependymomas from the bromodeoxyuridine labeling index. Childs Nerv Syst 8:273–8 Athanasiou A, Perunovic B, Quilty RD, Gorgoulis VG, Kittas C, Love S, 2003, Expression of mos in ependymal gliomas. Am J Clin Pathol 120:699–705 Egger M, Davey Smith G, Altman D, 2001, Systematic Reviews in Health Care: Meta-Analysis in Context. BMJ, London Rickert CH, Paulus W, 2005, Prognosis-related histomorphological and immunohistochemical markers in central nervous system tumors of childhood and adolescence. Acta Neuropathol, Berl, 109:69–92 Altman DG, Lyman GH, 1998, Methodological challenges in the evaluation of prognostic factors in breast cancer. Breast Cancer Res Treat 52:289–303 Hutchon DJ, 2001, Publishing raw data and real time statistical analysis on e-journals. Bmj 322(7285):530 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 605 EP 614 DI 10.1007/s12253-009-9160-2 PG 10 ER PT J AU Hashimoto, Y Tsukamoto, N Nakahashi, H Yokohama, A Saitoh, T Handa, H Matsushima, T Murakami, H Nojima, Y Karasawa, M AF Hashimoto, Yoko Tsukamoto, Norifumi Nakahashi, Hirotaka Yokohama, Akihiko Saitoh, Takayuki Handa, Hiroshi Matsushima, Takafumi Murakami, Hirokazu Nojima, Yoshihisa Karasawa, Masamitsu TI Hairy Cell Leukemia-Related Disorders Consistently Show Low CD27 Expression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hairy cell leukemia; HCL-Japanese variant; HBLD; CD27; IgVH; DRB1*04 ID Hairy cell leukemia; HCL-Japanese variant; HBLD; CD27; IgVH; DRB1*04 AB In Japan, typical hairy cell leukemia (HCL) is rare, and HCL-Japanese variant (HCL-JV) is more common. Hairy B-cell lymphoproliferative disorder (HBLD) is another unusual disorder of polyclonal B-lymphocytosis of hairy cell appearance. In the present study, we analyzed the clinical features of 3 patients with HCL, 3 with HCL-JV, and 3 with HBLD. All HBLD patients had the DRB1*04 allele. As compared with other B-cell lymphoproliferative disorders, CD27 expression on B cells was significantly lower in all patients, ranging from 0.3% to 23.4%. Our results suggest that low CD27 expression may be a distinct feature of these HCL-related disorders. C1 [Hashimoto, Yoko] Gunma University, Graduate School of Medicine, Department of Medicine and Clinical ScienceMaebashi, Gunma, Japan. [Tsukamoto, Norifumi] Gunma University, Graduate School of Medicine, Department of Medicine and Clinical ScienceMaebashi, Gunma, Japan. [Nakahashi, Hirotaka] Gunma University, Graduate School of Medicine, Department of Medicine and Clinical ScienceMaebashi, Gunma, Japan. [Yokohama, Akihiko] Gunma University, Graduate School of Medicine, Department of Medicine and Clinical ScienceMaebashi, Gunma, Japan. [Saitoh, Takayuki] Gunma University, Graduate School of Medicine, Department of Medicine and Clinical ScienceMaebashi, Gunma, Japan. [Handa, Hiroshi] Gunma University, Faculty of Medicine, School of Health ScienceMaebashi, Gunma, Japan. [Matsushima, Takafumi] Gunma University, Graduate School of Medicine, Department of Medicine and Clinical ScienceMaebashi, Gunma, Japan. [Murakami, Hirokazu] Gunma University, Faculty of Medicine, School of Health ScienceMaebashi, Gunma, Japan. [Nojima, Yoshihisa] Gunma University, Graduate School of Medicine, Department of Medicine and Clinical ScienceMaebashi, Gunma, Japan. [Karasawa, Masamitsu] Gunma University, Faculty of Medicine, Blood Transfusion Service, University Hospital, 3-39-15 Showa-machi, 371-8511 Maebashi, Gunma, Japan. RP Karasawa, M (reprint author), Gunma University, Faculty of Medicine, Blood Transfusion Service, University Hospital, 371-8511 Maebashi, Japan. EM karasawa@showa.gunma-u.ac.jp CR Jaffe ES, Harris NL, Stein H, Vardiman JW, 2001, World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues. IARC, Lyon Tiacci E, Liso A, Piris M, Falini B, 2006, Evolving concepts in the pathogenesis of hairy-cell leukaemia. Nat Rev Cancer 6:437– 448 Matutes E, Wotherspoon A, Catovsky D, 2003, The variant form of hairy-cell leukaemia. Best Pract Res Clin Haematol 16:41–56 Machii T, Tokumine Y, Inoue R, Kitani T, 1993, Predominance of distinct subtype of hairy cell leukemia in Japan. Leukemia 7:181–186 Matsue K, Nishi H, Onozawa S, Itoh M, Tsukada K, Yamaguchi M, Nakao S, Kashimura M, 1996, Polyclonal B cell lymphoproliferative disease with hairy cell morphology: A case report and clonal studies. Am J Hematol 51:141–146 Machii T, Yamaguchi M, Inoue R, Tokumine Y, Kuratsune H, Nagai H, Fukuda S, Furuyama K, Yamada O, Yahata Y, Kitani T, 1997, Polyclonal B-cell lymphocytosis with features resembling hairy cell leukemia-Japanese variant. Blood 89:2008–2014 Machii T, Yamaguchi M, Inoue R, Tokumine Y, Kuratsune H, Nagai H, Fukuda S, Furuyama K, Yamada O, Yawata Y, Kitani T, 1997, Identity between hairy B-cell lymphoproliferative disorder and persistent polyclonal B lymphocytosis? Blood 90:2110–2113 Kanbayashi H, Nagata K, Tanaka T, Matsuda S, Sakuma H, Maruyama Y,Machii T, 1998, Polyclonal B-cell lymphocytosis with clinical and hematological features resembling hairy cell leukemia. Rinsho Ketsueki 39:493–498 in Japanese, with English abstruct Yagi Y, Sakabe H, Kakinoki R, Yoshikawa K, Inoue T, Fujiyama Y, Machii T, 2004, A hairy B cell lymphoproliferative disorder resembling hairy cell leukemia. Rinsho Ketsueki 45:312–315 in Japanese, with English abstract Agematsu K, Hokibara S, Nagumo H, Komiyama A, 2000, CD27: a memory B-cell marker. Immunol Today 21:204–206 van Oers MH, Pals ST, Evers LM, van der Schoot CE, Koopman G, Bonfrer JM, Hintzen RQ, von dem Borne AE, van Lier RA, 1993, Expression and release of CD27 in human B-cell malignancies. Blood 82:3430–3436 Dong HY, Shahsafaei A, Dorfman DM, 2002, CD148 and CD27 are expressed in B cell lymphomas derived from both memory and naive B cells. Leuk Lymphoma 43:1855–1858 Guikema JEJ, Hovenga S, Vellenga E, Conradie JJ, Abdulahad WH, Bekkema R, Smit JW, Zhan F, Shaughnessy J Jr, Bos NA, 2003, CD27 is heterogeneously expressed in multiple myeloma: low CD27 expression in patients with high-risk disease. Br J Haematol 121:36–43 Katayama Y, Sakai A, Oue N, Asaoku H, Otsuki T, Shiomomura T, Masuda R, Hino N, Takimoto Y, Imanaka F, Yasui W, Kimura A, 2003, A possible role for the loss of CD27–70 interaction in myelomagenesis. Br J Haematol 120:223–234 Forconi F, Raspadori D, Lenoci M, Luria F, 2005, Absence of surface CD27 distinguishes hairy cell leukemia from other leukemic B-cell malignancies. Haematologica 90:266–268 Nakahashi H, Hashimoto Y, Yamane A, Irisawa H, Yokohama A, Saitoh T, Handa H, Matsushima T, Tsukamoto N, Karasawa M, Murakami H, Nojima Y, 2007, Polyclonal B-cell lymphocytosis with hairy cell appearance: hairy B-cell lymphoproliferative disorder. Rinsho Ketsueki 48:647–651 in Japanese, with English abstruct Koiso H, Tsukamoto N, Miyawaki S, Shinonome S, Nojima Y, Karasawa M, 2002, Quantitative analysis of Cyclin D1 and CD23 expression in mantle cell lymphoma and B-chronic lymphocytic leukemia. Leuk Res 26:809–815 van Dongen JJM, Langerak AW, Bruggemann M, Evans PAS, Hummel M, Lavender FL, Delabesse E, Davi F, Schuuring E, Garcia-Sanz R, van Krieken JHJM, Droese J, Gonzalez D, Bastard C, White HE, Spaargaren M, Gonzalez M, Parreira A, Smith JL, Morgan GJ, Kneba M, Macintyre EA, 2003, Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: Report of the BIOMED-2 concerted action BMH4-CT98–3936. Leukemia 17:2257–2317 Koiso H, Yamane A, Mitsui T, Matsushima T, Tsukamoto N, Murakami H, Nojima Y, Karasawa M, 2006, Distinctive immunoglobulin VH gene usage in Japanese patients with chronic lymphocytic leukemia. Leuk Res 30:272–276 Goldon DS, Jones BM, Browning SW, Spira TJ, Lawrence DN, 1982, Persistent polyclonal lymphocytosis of B lymphocytes. N Eng J Med 307:232–236 Troussard X, Flandrin G, 1996, Chronic B-cell lymphocytosis with binucleated lymphocytes, LWBL): a review of 38 cases. Leuk Lymphoma 20:275–279 Hashimoto M, Kinoshita T, Yamasaki M, Tanaka H, Imanishi T, Ihara H, Ichikawa Y, Fukunishi T, 1994, Gene frequencies and haplotypic associations within the HLA region in 916 unrelated Japanese individuals. Tissue Antigens 44:166–173 Maloum K, Magnac C, Azgui Z, Cau C, Charlotte F, Binet JL, Merle-Beral H, Diqhiero G, 1998, VH gene expression in hairy cell leukemia. Br J Haematol 101:171–178 Vanhentenrijk V, Tierens A, Wlodarska I, Verhoef G, Wolf-Peeters CD, 2004, VH gene analysis of hairy cell leukemia reveals a homogeneous mutation status and suggests its marginal zone Bcell origin. Leukemia 18:1729–1732 Ranheim EA, Cantwell MJ, Kipps TJ, 1995, Expression of CD27 and its ligand, CD70, on chronic lymphocytic leukemia B cells. Blood 85:3556–3565 Ehrhardt GR, Hsu JT, Gartland L, Leu C, Zhang S, Davis RS, Cooper MD, 2005, Expression of the immunoregulatory molecule FcRH4 defines a distinctive tissue-based population of memory B cells. J Exp Med 202:783–791 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 615 EP 621 DI 10.1007/s12253-009-9161-1 PG 7 ER PT J AU Ren, T Xu, L Jiao, Sh Wang, Y Cai, Y Liang, Y Zhou, Y Zhou, H Wen, Z AF Ren, Tao Xu, Lin Jiao, Shuxian Wang, Yanying Cai, Yingyun Liang, Yongjie Zhou, Ya Zhou, Hong Wen, Zhenke TI TLR9 Signaling Promotes Tumor Progression of Human Lung Cancer Cell In Vivo SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CpG oligonucleotide; Lung cancer; TLR9; Tumor progression; Tumor immunity ID CpG oligonucleotide; Lung cancer; TLR9; Tumor progression; Tumor immunity AB Toll like receptor 9 (TLR9) was identified mainly in cells of the immune system, and CpG oligonucleotides (CpG ODN), which induces signaling through TLR9, are currently under investigation as adjuvants in clinical therapies against cancer. However, accumulating data suggested that functional TLR9 was also expressed in tumor cells and the effects of TLR9 signaling on the progression of tumor cells remain undefined. Our previous study demonstrated that the TLR9 signaling could significantly enhance the metastatic potential of human lung cancer cells in vitro. Here we carefully evaluated the direct effect of TLR9 signaling on tumor progression of human lung cancer cells in vitro and in vivo. We observed that TLR9 agonist CpG ODN could robustly enhance the tumor progression of 95D cells which expressed high level of TLR9 in nude mice. Furthermore, the CpG ODN could effectively induce the proliferation and IL-10 secretion of 95D cells in vitro. Finally, we demonstrated that CpG ODN could significantly elevate the tumor progression of TLR9 modifying 95C cells in vitro and in vivo, which could be dramatically abrogated by the inhibitory CpG ODN. Our findings indicated that the TLR9 signaling could promote the tumor progression of human tumor cells, which might provide novel insight into the implications for CpG based anti-tumor therapies. C1 [Ren, Tao] Tongji University, East Hospital, Department of Respiratory MedicineShanghai, China. [Xu, Lin] Zunyi Medical College, Department of ImmunologyGuizhou, China. [Jiao, Shuxian] Qingdao Blood Center, Department of Central Laboratory, 266071 Qingdao, Shandong Province, China. [Wang, Yanying] Fudan University, Zhongshan Hospital, Department of GerontologyShanghai, China. [Cai, Yingyun] Fudan University, Zhongshan Hospital, Department of GerontologyShanghai, China. [Liang, Yongjie] Tongji University, East Hospital, Department of Respiratory MedicineShanghai, China. [Zhou, Ya] Zunyi Medical College, Department of Medical PhysicsGuizhou, China. [Zhou, Hong] Qingdao Blood Center, Department of Central Laboratory, 266071 Qingdao, Shandong Province, China. [Wen, Zhenke] Qingdao Blood Center, Department of Central Laboratory, 266071 Qingdao, Shandong Province, China. RP Wen, Z (reprint author), Qingdao Blood Center, Department of Central Laboratory, 266071 Qingdao, China. EM wenzk@126.com CR Werling D, Jungi TW, 2003, TOLL-like receptors linking innate and adaptive immune response. Vet Immunol Immunopathol 91:1–12 Janeway CA Jr, Medzhitov R, 2002, Innate immune recognition. Annu Rev Immunol 20:197–216 Aliprantis AO, Yang RB, Mark MR et al, 1999, Cell activation and apoptosis by bacterial lipoproteins through toll-like receptor- 2. Science 285:736–739 Alexopoulou L, Holt AC, Medzhitov R et al, 2001, Recognition of double-stranded RNA and activation of NF-kappaB by Toll like receptor 3. Nature 413:732–738 Poltorak A, He X, Smirnova I et al, 1998, Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 282:2085–2088 Hemmi H, Takeuchi O, Kawai T et al, 2000, A Toll-like receptor recognizes bacterial DNA. Nature 408:740–745 Muzio M, Natoli G, Saccani S et al, 1998, The human toll signaling pathway: divergence of nuclear factor kappaB and JNK/ SAPK activation upstream of tumor necrosis factor receptorassociated factor 6, TRAF6). J Exp Med 187:2097–2101 Vollmer J, Weeratna R, Payette P et al, 2004, Characterization of three CpG oligodeoxynucleotide classes with distinct immunostimulatory activities. Eur J Immunol 34:251–262 Kraft AR, Krux F, Schimmer S et al, 2007, CpG Oligodeoxynucleotides allow for effective adoptive T-cell therapy in chronic retroviral infection. Blood 109:2982–2984 Chaudhry UI, Kingham TP, Plitas G et al, 2006, Combined stimulation with interleukin-18 and CpG induces murine natural killer dendritic cells to produce IFN-gamma and inhibit tumor growth. Cancer Res 66:497–504 Roman M, Martin-Orozco E, Goodman JS et al, 1997, Immunostimulatory DNA sequences function as T helper-1-promoting adjuvants. Nat Med 3:849–854 O’Keeffe M, Grumont RJ, Hochrein H et al, 2005, Distinct roles for the NF-kappaB1 and c-Rel transcription factors in the differentiation and survival of plasmacytoid and conventional dendritic cells activated by TLR-9 signals. Blood 106:3457–3464 Steinman RM, Dhodapkar M, 2001, Active immunization against cancer with dendritic cells: the near future. Int J Cancer 94:459– 473 Sparwasser T, Vabulas RM, Villmow B et al, 2000, Bacterial CpG-DNA activates dendritic cells in vivo: T helper cellindependent cytotoxic T cell responses to soluble proteins. Eur J Immunol 30:3591–3597 Heckelsmiller K, Beck S, Rall K et al, 2002, Combined dendritic cell-and CpG oligonucleotide-based immune therapy cures large murine tumors that resist chemotherapy. Eur J Immunol 32:3235– 3245 Okamoto M, Sato M, 2003, Toll-like receptor signaling in anticancer immunity. J Med Invest 50:9–24 Wooldridge JE, Weiner GJ, 2003, CpG DNA and cancer immunotherapy: orchestrating the antitumor immune response. Curr Opin Oncol 15:440–445 Belvin MP, Anderson KV, 1996, A conserved signaling pathway: the Drosophila toll-dorsal pathway. Annu Rev Cell Dev Biol 12:393–416 Coussens LM, Werb Z, 2002, Inflammation and cancer. Nature 420:860–867 Coussens LM, Werb Z, 2001, Inflammatory cells and cancer: think different!. J Exp Med1 93:F23–26 Ren T, Wen ZK, Liu ZM et al, 2007, Functional expression of TLR9 is associated to the metastatic potential of human lung cancer cell: functional active role of TLR9 on tumor metastasis. Cancer Biol Ther 6:1704–1709 Lubin I, Segall H, Marcus H et al, 1994, Engraftment of human peripheral blood lymphocytes in normal strains of mice. Blood 83:2368–2381 Tsushima F, Tanaka K, Otsuki N et al, 2006, Predominant expression of B7-H1 and its immunoregulatory roles in oral squamous cell carcinoma. Oral Oncol 42:268–274 Wang CC, Zeng Q, Hwang LA et al, 2006, Mouse lymphomas caused by an intron-splicing donor site deletion of the FasL gene. Biochem Biophys Res Commun 349:50–58 Gazzinelli RT, Denkers EY, 2006, Protozoan encounters with Toll-like receptor signalling pathways: implications for host parasitism. Nat Rev Immunol 6:895–906 Creagh EM, O’Neill LA, 2006, TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol 27:352–357 Droemann D, Albrecht D, Gerdes J et al, 2005, Human lung cancer cells express functionally active Toll-like receptor 9. Respir Res 6:1 Jarrousse V, Quereux G, Marques-Briand S et al, 2006, Toll-like receptors 2, 4 and 9 expression in cutaneous T-cell lymphoma, mycosis fungoides and Sezary syndrome). Eur J Dermatol 16:636–641 Huang B, Zhao J, Unkeless JC et al, 2008, TLR signaling by tumor and immune cells: a double-edged sword. Oncogene 27:218–224 Merrell MA, Ilvesaro JM, Lehtonen N et al, 2006, Toll-like receptor 9 agonists promote cellular invasion by increasing matrix metalloproteinase activity. Mol Cancer Res 4:437–447 Kelly MG, Alvero AB, Chen R et al, 2006, TLR-4 signaling promotes tumor growth and paclitaxel chemoresistance in ovarian cancer. Cancer Res 66:3859–3868 Melief CJ, 2008, Cancer immunotherapy by dendritic cells. Immunity 29:372–383 Bermudez-Morales VH, Gutierrez LX, Alcocer-Gonzalez JM et al, 2008, Correlation Between IL-10 Gene Expression and HPV Infection in Cervical Cancer: A Mechanism for Immune Response Escape. Cancer Invest 16:1 [Epub ahead of print] Decker T, Schneller F, Sparwasser T et al, 2000, Immunostimulatory CpG-oligonucleotides cause proliferation, cytokine production, and an immunogenic phenotype in chronic lymphocytic leukemia B cells. Blood 95:999–1006 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 623 EP 630 DI 10.1007/s12253-009-9162-0 PG 8 ER PT J AU Szabo, VG Kunstar, A Acsady, Gy AF Szabo, Viktor Gabor Kunstar, Aliz Acsady, Gyorgy TI Methylentetrahydrofolate Reductase and Nitric Oxide Synthase Polymorphism in Patients with Atherosclerosis and Diabetes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Nitric oxide synthase; Methylentetrahydrofolate reductase; Polymorphism; Atherosclerosis; Cardiovascular Risk ID Nitric oxide synthase; Methylentetrahydrofolate reductase; Polymorphism; Atherosclerosis; Cardiovascular Risk AB The development of the atherosclerosis is based on multifactorial causes. In addition to the traditional risk factors, gene polymorphisms can play a role in the disease. Therefore in this study we investigated whether the eNOS and MTHFR gene polymorphisms is associated with myocardial infarction and stroke in patients with or without diabetes. We have identified polymorphisms in the NOS 3 gene and one of these polymorphisms, Glu298→Asp, was found to be a major risk factor for carotid artery disease and myocardial infarction. Our results indicate that the MTHFR G677T allele is significantly associated with MI. MTHFR 677 G/T genotyping may be of clinical importance as a prognostic and therapeutic marker, although further studies are needed to substantiate this hypothesis. C1 [Szabo, Viktor Gabor] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor str. 68, H-1122 Budapest, Hungary. [Kunstar, Aliz] National Institute of OncologyBudapest, Hungary. [Acsady, Gyorgy] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor str. 68, H-1122 Budapest, Hungary. RP Szabo, VG (reprint author), Semmelweis University, Department of Cardiovascular Surgery, H-1122 Budapest, Hungary. EM szabogvdr@gmail.com CR Ross R, 1993, The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature 362:801–809 Boerwinkle E, Elsworth DL, Hallman DM, Biddinger A, 1996, Genetic analysis of atherosclerosis: a research paradigm for the common disease. Hum Mol Genet 5:1405–1410 Pallaud C, Sass C, Zannad F, Siest G, Visvikis S, 2001, APOC3, CETP, fibrinogen, and MTHFR are genetic determinants of carotid intima-media thickness in healthy men, the Stanislas cohort). Clin Genet 59:316–324 Jerrard-Dunne P, Markus HS, Steckel DA, Buehler A, von Kegler S, Sitzer M, 2003, Early carotid atherosclerosis and family history of vascular disease: specific effects on arterial sites have implications for genetic studies. Arterioscler Thromb Vasc Biol 23:302–306 Davignon J, Ganz P, 2004, Role of endothelial dysfunction in atherosclerosis. Circulation 109:27–32 Colombo MG, Paradossi U, Andreassi MG, Botto N, Manfredi S, Masetti S, Biagini A, Clerico A, 2003, Endothelial nitric oxide synthase gene polymorphisms and risk of coronary artery disease. Clin Chem 49:389–395 Jeerooburkhan N, Jones LC, Bujac S, Cooper JA, Miller GJ, Vallance P, Humphries SE, Hingorani AD, 2001, Genetic and environmental determinants of plasma nitrogen oxides and risk of ischemic heart disease. Hypertension 38:1054–1061 Marroni AS,Metzger IF, Souza-Costa DC, Nagassaki S, Sandrim VC, Correa RX, Rios-Santos F, Tanus-Santos JE, 2005, Consistent interethnic differences in the distribution of clinically relevant endothelial nitric oxide synthase genetic polymorphisms. Nitric Oxide 12:177–182 Tanus-Santos JE, Desai M, Flockhart DA, 2001, Effects of ethnicity on the distribution of clinically relevant endothelial nitric oxide variants. Pharmacogenetics 11:719–725 Casas Juan P, Bautista Leonelo E, Humphries Steve E, Hingorani Aroon D, 2004, Endothelial Nitric Oxide Synthase Genotype and Ischemic Heart Disease. Circulation 09:1359–1365 Mazza A, Giugliano D, Motti C, Cortese C, Andreotti F, Marra G, Nulli A, 2000, Glycemia, MTHFR genotype and low homocysteine in uncomplicated type 2 diabetic patients. Atherosclerosis 149:223–224 Selhub J, Jacques PF, Bostom AG, Agostino RB D, Wilson PW, Belanger AJ, Leary DH O, Wolf PA, Schaefer EJ, Rosenberg IH, 1995, Association between plasma homocysteine concentrations and extracranial carotid-artery stenosis. N Engl J Med 332:286–291 Refsum H, Ueland PM, Nygard O, Vollset SE, 1998, Homocysteine and cardiovascular disease. Ann Rev Med 49:31–62 Ueland PM, Refsum H, Shirley AA, 2000, Beresford and Stein Emil Vollset. The controversy over homocysteine and cardiovascular risk1,2. American J Clin Nutr 72:324–332 Refsum H, Ueland PM, Nygard O, Vollset SE, 1998, Homocysteine and cardiovascular disease. Ann Rev Med 49:31–62 Bova I, Chapman J, Sylantiev C, Korczyn AD, Bornstein NM, 1999, The A677V methylenetetrahydrofolate reductase gene polymorphism and carotid atherosclerosis. Stroke 30:2180–2182 Pallaud C, Sass C, Zannad F, Siest G, Visvikis S, 2001, APOC3, CETP, fibrinogen, and MTHFR are genetic determinants of carotid intima-media thickness in healthy men, the Stanislas cohort). Clin Genet 59:316–324 Passaro A, Vanini A, Calzoni F, Alberti L, Zamboni PF, Fellin R, Solini A, 2001, Plasma homocysteine, methylenetetrahydrofolate reductase mutation and carotid damage in elderly healthy women. Atherosclerosis 157:175–180 Inamoto N, Katsuya T, Kokubo Y, Mannami T, Asai T, Baba S, Ogata J, Tomoike H, Ogihara T, 2003, Association of methylenetetrahydrofolate reductase gene polymorphism with carotid atherosclerosis depending on smoking status in a Japanese general population. Stroke 34:1628–1633 Zuliani G, Volpato S, Mecocci P, Cherubini A, 2000, The A677V MTHFR allele is not associated with carotid atherosclerosis in octogenarians. Stroke 31:990–991 Mazza A, Motti C, Nulli A, Marra G, Gnasso A, Pastore A, Federici G, Cortese C, 2000, Lack of association between carotid intima-media thickness and methylenetetrahydrofolate reductase gene polymorphism or serum homocysteine in non-insulindependent diabetes mellitus. Metabolism 1949:718–723 Rossi GP, Cesari M, ZanchettaM, Colonna S, Maiolino G, Pedon L, Cavallin M, Maiolino P, Pessina AC, 2003, The T-786C endothelial nitric oxide synthase genotype is a novel risk factor for coronary artery disease in Caucasian patients of the GENICA study. J Am Coll Cardiol 41:930–937 Alvarez R, Gonzalez P, Batalla A, Reguero JR, Iglesias-Cubero G, Hevia S, Cortina A, Merino E, Gonzalez I, Alvarez V, Coto E, 2001, Association between the NOS3, -786T/C, and the ACE, I/D, DNA genotypes and early coronary artery disease. Nitric Oxide 5:343–348 Cai H, Wilcken DE, Wang XL, 1999, The Glu-298—> Asp, 894G —> T, mutation at exon 7 of the endothelial nitric oxide synthase gene and coronary artery disease. J Mol Med 77:511–514 Hibi K, Ishigami T, Tamura K, Mizushima S, Nyui N, Fujita T, Ochiai H, Kosuge M, Watanabe Y, Yoshii Y, Kihara M, Kimura K, Ishii M, Umemura S, 1998, Endothelial nitric oxide synthase gene polymorphism and acute myocardial infarction. Hypertension 32:521–526 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 631 EP 637 DI 10.1007/s12253-009-9163-z PG 7 ER PT J AU Baranyay, F AF Baranyay, Ferenc TI Histochemical Contributions to the Binding Mechanism of Complement (CR1, CR2) Receptors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CR1; CR2-reversible tissue adherence of E(A) C3b; E(A)C3d ID CR1; CR2-reversible tissue adherence of E(A) C3b; E(A)C3d AB Complement receptors (CR1, CR2, CR3), and their ligands (C3b, C3d, iC3b) are essentially involved in germinal center development and in binding, trapping, and retaining immunocomplexes. Methods studying complement receptor (CR1/CR2)-ligand (C3b/C3d) interactions mostly involve coating of sheep erythrocytes (E), sheep erythrocyte-antisheep erythrocyte antibody (EA complexes) and whole human (h) or mouse (m) sera as a source of complement, EACh/m complexes, as reagents. The observation of Dukor et al. (1970), that EACm complexes in native cryostat sections bind selectively and very strongly to the B lymphocyte regions of lymphoid organs allowed the topo-histochemical analysis of receptor (CR1/CR2)—ligand (C3b/C3d) interactions in such an immunologically important area as the germinal centers. The main finding of this study is, that periodic acid pretreatment of unfixed cryostat tonsil sections-oxidizing vicinal glycol groups of polysaccharide chains into dialdehydes-completely abolished the binding of all EAC/EC complexes to germinal center area. It may suggest the involvement of receptor carbohydrate in C3 receptor/ligand binding. In addition to, the subsequent sodium borohydride reduction-converting aldehydes (produced by periodic acid oxidation) into primary alcohols-restored selectively the binding of all applied EAC/EC complexes to follicular centers. These in vitro topo-histochemical studies give a strong hint for the participation of—OH groups of sugar residues in CR1/CR2 ligand (C3b/C3d) binding. C1 [Baranyay, Ferenc] Medical University of Pecs, Dorottya Kanizsai Hospital, Szekeres J. u. 2-8, 8801 Nagykanizsa, Hungary. RP Baranyay, F (reprint author), Medical University of Pecs, Dorottya Kanizsai Hospital, 8801 Nagykanizsa, Hungary. EM fbaranyay@hotmail.com CR Bara J, Decaens C, Loridon-Rosa B, Oriol R, 1992, Immunohistological characterization of mucin epitopes by pre-treatment of gastro-intestinal sections with periodic acid. J Immunol Methods 149:105–113 Bianco C, Patrick R, Nussenzweig V, 1970, A population of lymphocytes bearing a membrane receptor for antigen-antibodycomplement complexes. J Exp Med 132:702–720 Birmingham DJ, Irshaid F, Gavit KF, Nagaraja HN, Yu CY, Rovin BH, Hebert LA, 2007, A polymorphism in the type one complement receptor, CR1, involves an additional cysteine within the C3b/C4b binding domain that inhibits ligand binding. Mol Immunol 44:3510–3516 Clamp JR, Hough L, 1965, The periodate oxidation of amino acids with reference studies to glycoproteins. Biochem J 94:17–24 Dierich MP, Scheiner O, Mussel HH, Hamman KP, Schopf RE, Schutz Z, 1982, Characterisation of complement receptors. Molecular Immunology 19:1201–1204 Dukor P, Bianco C, Nussenzweig V, 1970, Tissue localization of lymphocytes bearing a membrane receptor for antigen -antibodycomplement complexes. Proc Natl Acad SAci USA 67:991–997 Fearon DT, 1979, Regulation of the amplification C3 convertase of human complement by an inhibitory protein isolated from human erythrocyte membrane. Proc Natl Acad Sci USA 76:5867– 5871 FischerMB, Goerg S, Shen L, Prodeus AP, Goodnow CC, Kelsoe G, Carroll MC, 1998, Dependence of germinal center B cells on expression of CD21/CD35 for survival. Science 280:582–585 Fischer MB, Ma M, Hsu NC, Carroll MC, 1998, Local synthesis of C3 within the splenic lymphoid compartment can reconstitute the impaired immune response in C3-deficient mice. J Immunol 160:2619–2625 Guthridge JM, Rakstang JK, Young KA, Hinshelwood J, Aslam M, Robertson AG, GipsonMG SMR, Moore WT, Meagher M, Karp D, Lambris JD, Perkins SJ, Holers VM, 2001, Structural studies in solution of the recombinant N-terminal pair of short consensus / complement repeat domains of complement receptor type 2, CR2/ CD21, and interactions with its ligand C3dg. Biochemistry 20:5931–5941 Huber H, Polley MJ, Linscott WD, Fudenberg HH, Muller- Eberhardt HJ, 1968, Human monocytes: distinct receptor sites for the third component of complement and for immunoglobulin G. Science 162:1281–1285 Hsu SM, YS H, Li PJ, Monheit J, Ree HJ, Sheibani K, Winberg CD, 1986, L-H variants of Reed-Sternberg cellsexpress sialylated Leu M1 antigens. Am J Path 122:199–203 Klickstein LB, Wong WW, Smith JA, Weis JH, Wilson JG, Fearon DT, 1987, Human C3b/C4b receptor, CR1). Demonstration of long homologous repeating domains that are composed of the short consensus repeats characteristics of C3/C4 binding proteins. J Exp Med 165:1095–1112 Krych-Goldberg M, Atkinson JP, 2001, Structure-function relationships of complement receptor type 1. Immunol Rev 180:112–122 Lambris JD, Scheiner O, Schulz TF, Alsenz J, Dierich MP, 1983, Coupling of C3b to erythrocytes by disulfide bond formation of EC3b for hemolytic and complement receptor assays. J Immunol Methods 65:277–283 Lay WH, Nussenzweig W, 1968, Receptors for complement of leukocytes. J Exp Med 128:991–1007 Lillie RD, Pizzolato P, 1972, Histochemical use of borohydrides as aldehyde blocking reagents. Stain Techn 47:13–16 Lublin DM, Griffith RC, Atkinson JP, 1986, Influence of glycosylation on allelic and cell specific Mr variation receptor processing and ligand binding of the human C3b/C4b receptor. J Biol Chem 261:5736–5744 Malaprade L, 1934, Etude de l'action de polyacools sur l'acide periodique et les periodates alcalins. Bull Soc Chim Franc 5:833– 852 McManus JFA, 1946, Histological demonstration of mucin after periodic acid. Nature 158:202 Molina H, Perkins SJ, Guthridge J, Gorka J, Kinoshita T, Holers M, 1995, Characterisation of a complement receptor 2, CR2, CD21, ligand binding site for C3. An initial model of ligand interaction with two linked short consensus repeat moduls. J Immunol. 154:5426–5435 Muller-Eberhard HJ, 1988, Molecular organisation and function of the complement system. Ann Rev Biochem 57:321–347 Novogrodsky A, Katchalski E, 1972, Membrane site modified on induction of the transformation of lymphocytes by periodate. Proc Natl Acad Sci USA 69:3207–3210 O'Brien RL, Parker JW, 1976, Oxidation-induced lymphocyte transformation. Cell 7:13–20 Okuda T, Sato S, Tachibanam T, 1984, The effect of divalent cations on the rosette formation via C3 receptors. Tohoku Exp Med 143:221–224 Reynes M, Aubert JP, Cohen JH, Audpuin J, Tricottet V, Diebold J, Kazatchkine MD, 1985, Human follicular dendritic cells express CR1, CR2 and CR3 complement receptor antigens. J Immunol 135:2687–2694 Ross GD, Medof E, 1985, Membrane complement receptors specific for bound fragments of C3. Adv Immunol 37:217–267 Stein HE, Grosskopf E, Lennert K, 1980, Demonstration of C3 receptors of frozen sections of normal lymphoid tissue and malignant lymphomas using vartious EAC complexes Blut. Ann Hematol 41:101–118 Szakonyi G, Guthridge JM, Li D, Young K, Holer M, Chen XS, 2001, Structure of complement receptor 2 in complex with its C3d ligand. Science 292:1725–1728 Turner RH, Liener IE, 1975, The use of glutaraldehyde treated erythrocytes for assaying the agglutination activity of lectins. Anal Biochem 68:651–653 Zatz MM, Goldstein AL, Blumenfeld OO, White A, 1972, Regulation of normal and leulaemic lymphocyte transformation and recirculation by sodium periodate oxidation and sodium borohydride reduction. Nature New Biology 240:252–255 Zwirner J, Felber E, Schmidt P, Riethmuller G, Feucht HE, 1989, Complement activation in human germinal centres. Immunology 66:270–77 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 639 EP 644 DI 10.1007/s12253-009-9164-y PG 6 ER PT J AU Roberts, SS Mendonca-Torres, CM Jensen, K Francis, LG Vasko, V AF Roberts, S Stephen Mendonca-Torres, Cecilia Maria Jensen, Kirk Francis, L Gary Vasko, Vasyl TI GABA Receptor Expression in Benign and Malignant Thyroid Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer; GABA receptors; Gene expression; Immunohistochemistry; Thyroid ID Cancer; GABA receptors; Gene expression; Immunohistochemistry; Thyroid AB Neurotransmitter systems have recently been shown to be involved in multiple malignancies including breast, colon and prostate cancers. The role of neurotransmitters and neurotrophic factors has not yet been examined in thyroid cancer. To determine the possible involvement of neurotransmitter systems in thyroid carcinogenesis we characterized the patterns of gammaaminobutyric acid (GABA) receptor expression in normal thyroid and thyroid tumors. We examined the expression patterns of the GABAergic system in 70 human thyroid tumor samples (13 follicular adenomas, 14 follicular carcinomas, 43 papillary carcinomas) and adjacent normal thyroid by immunohistochemistry. GABAergic system mRNA expression in thyroid cancer cell lines derived from primary (FTC133) and metastatic tumors (FTC236 and FTC238) was examined by real time PCR. Overall, GABA receptor expression is increased in tumors compared to normal thyroid tissue. Expression of GABAA receptor β2 was detected in the vasculature of normal thyroid and thyroid tumors but not in thyroid cancer cells. GABAA α2 was detected in metastatic-derived but not in primary-tumor derived cell lines. Expression levels of GABAB R2 and GABA receptor associated protein (GABARAP) are increased in adenomas and thyroid cancer suggesting their role in early stages of thyroid tumorigenesis. This study represents the first demonstration of GABA receptor expression in human thyroid tissue and suggests that the GABAergic system is involved in thyroid carcinogenesis. C1 [Roberts, S Stephen] Uniformed Services University of the Health Sciences, Department of Pediatrics, 4301 Jones Bridge Road, 20814 Bethesda, MD, USA. [Mendonca-Torres, Cecilia Maria] Uniformed Services University of the Health Sciences, Department of Pediatrics, 4301 Jones Bridge Road, 20814 Bethesda, MD, USA. [Jensen, Kirk] Uniformed Services University of the Health Sciences, Department of Pediatrics, 4301 Jones Bridge Road, 20814 Bethesda, MD, USA. [Francis, L Gary] Uniformed Services University of the Health Sciences, Department of Pediatrics, 4301 Jones Bridge Road, 20814 Bethesda, MD, USA. [Vasko, Vasyl] Uniformed Services University of the Health Sciences, Department of Pediatrics, 4301 Jones Bridge Road, 20814 Bethesda, MD, USA. RP Roberts, SS (reprint author), Uniformed Services University of the Health Sciences, Department of Pediatrics, 20814 Bethesda, USA. EM sroberts@usuhs.mil CR Ries LAG, Krapcho M, Stinchcomb DG, Howlader N, Horner MJ, Mariotto A, Miller BA, Feuer EJ, Altekruse SF, Lewis DR, Clegg L, Eisner MP, Reichman M, Edwards BK, 2008, SEER Cancer Statistics Review, 1975–2005. via http://seer.cancer.gov/ csr/1975_2005/ Cited 04 December 2008 DeGroot LJ, Kaplan EL, McCormick M, Straus FH, 1990, Natural history, treatment, and course of papillary thyroid carcinoma. J Clin Endocrinol Metab 71:414–424 Ringel MD, Ladenson PW, 2004, Controversies in the follow-up and management of well-differentiated thyroid cancer. Endocr Relat Cancer 11:97–116 Vasko V, Bauer AJ, Tuttle RM, Francis GL, 2007, Papillary and follicular thyroid cancers in children. Endocr Dev 10:140–172 Hundahl SA, Fleming ID, Fremgen AM, Menck HR, 1998, A national cancer data base report on 53,856 cases of thyroid carcinoma treated in the U.S., 1985–1995 [see comments]. Cancer 83:2638–2648 Ain KB, Egorin MJ, DeSimone PA, 2000, Treatment of anaplastic thyroid carcinoma with paclitaxel: phase 2 trial using ninety-sixhour infusion. Collaborative anaplastic thyroid cancer health intervention trials, CATCHIT, group. Thyroid 10:587–594 Entschladen F, Palm D, Lang K, TLt D, Zaenker KS, 2006, Neoneurogenesis: tumors may initiate their own innervation by the release of neurotrophic factors in analogy to lymphangiogenesis and neoangiogenesis. Med Hypotheses 67:33–35 Palm D, Entschladen F, 2007, Neoneurogenesis and the neuroneoplastic synapse. Prog Exp Tumor Res 39:91–98 Lang K, TLt D, Zaenker KS, Entschladen F, 2006, Inhibitors for metastasis development. Recent Patents Anticancer Drug Discov 1:69–80 Macdonald RL, Olsen RW, 1994, GABAA receptor channels. Annu Rev Neurosci 17:569–602 Akinci MK, Schofield PR, 1999, Widespread expression of GABA(A, receptor subunits in peripheral tissues. Neurosci Res 35:145–153 Ong J, Kerr DI, 1990, GABA-receptors in peripheral tissues. Life Sci 46:1489–1501 Wiens SC, Trudeau VL, 2006, Thyroid hormone and gammaaminobutyric acid, GABA, interactions in neuroendocrine systems. Comp Biochem Physiol A Mol Integr Physiol 144:332–344 Gebauer H, 1981, GABA transport in the rat thyroid. Naunyn Schmiedebergs Arch Pharmacol 317:61–66 Gebauer H, Pabst MA, 1981, Autoradiographic localization of 3H-GABA uptake in the thyroid gland of the rat. Cell Tissue Res 220:873–879 Martin JV, Williams DB, Fitzgerald RM, Im HK, Vonvoigtlander PF, 1996, Thyroid hormonal modulation of the binding and activity of the GABAA receptor complex of brain. Neuroscience 73:705–713 Mason GA, Walker CH, Prange AJ Jr, Bondy SC, 1987, GABA uptake is inhibited by thyroid hormones: implications for depression. Psychoneuroendocrinology 12:53–59 Ahren B, 1989, GABA inhibits thyroid hormone secretion in the mouse. Thyroidology 1:105–108 Jiang Y, Harlocker SL, Molesh DA, Dillon DC, Stolk JA, Houghton RL et al, 2002, Discovery of differentially expressed genes in human breast cancer using subtracted cDNA libraries and cDNA microarrays. Oncogene 21:2270–2282 Kleinrok Z, Matuszek M, Jesipowicz J, Matuszek B, Opolski A, Radzikowski C, 1998, GABA content and GAD activity in colon tumors taken from patients with colon cancer or from xenografted human colon cancer cells growing as s.c. tumors in athymic nu/nu mice. J Physiol Pharmacol 49:303–310 Matuszek M, Jesipowicz M, Kleinrok Z, 2001, GABA content and GAD activity in gastric cancer. Med Sci Monit 7:377–381 Mazurkiewicz M, Opolski A, Wietrzyk J, Radzikowski C, Kleinrok Z, 1999, GABA level and GAD activity in human and mouse normal and neoplastic mammary gland. J Exp Clin Cancer Res 18:247–253 Nicholson-Guthrie CS, Guthrie GD, Sutton GP, Baenziger JC, 2001, Urine GABA levels in ovarian cancer patients: elevated GABA in malignancy. Cancer Lett 162:27–30 Opolski A,MazurkiewiczM,Wietrzyk J, Kleinrok Z, Radzikowski C, 2000, The role of GABA-ergic system in human mammary gland pathology and in growth of transplantable murine mammary cancer. J Exp Clin Cancer Res 19:383–390 Vasko V, Espinosa AV, Scouten W, He H, Auer H, Liyanarachchi S et al, 2007, Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion. Proc Natl Acad Sci USA 104:2803–2808 Hemelaar J, Lelyveld VS, Kessler BM, Ploegh HL, 2003, A single protease, Apg4B, is specific for the autophagy-related ubiquitin-like proteins GATE-16, MAP1-LC3, GABARAP, and Apg8L. J Biol Chem 278:51841–51850 Sou YS, Tanida I, Komatsu M, Ueno T, Kominami E, 2006, Phosphatidylserine in addition to phosphatidylethanolamine is an in vitro target of the mammalian Atg8 modifiers, LC3, GABARAP, and GATE-16. J Biol Chem 281:3017–3024 Tanida I, Ueno T, Kominami E, 2004, LC3 conjugation system in mammalian autophagy. Int J Biochem Cell Biol 36:2503–2518 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 645 EP 650 DI 10.1007/s12253-009-9165-x PG 6 ER PT J AU Chen, HJ Mok, ST Chen, ZH Guo, AL Zhang, XCh Su, J Wu, YL AF Chen, Hua-Jun Mok, S Tony Chen, Zhi-Hong Guo, Ai-Lin Zhang, Xu-Chao Su, Jian Wu, Yi-Long TI Clinicopathologic and Molecular Features of Epidermal Growth Factor Receptor T790M Mutation and c-MET Amplification in Tyrosine Kinase Inhibitor-resistant Chinese Non-small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE c-MET; Epidermal growth factor receptor; Non-small cell lung cancer ; Resistance; T790M ID c-MET; Epidermal growth factor receptor; Non-small cell lung cancer ; Resistance; T790M AB To investigate the clinicopathologic and molecular features of the T790M mutation and c-MET amplification in a cohort of Chinese non-small cell lung cancer (NSCLC) patients resistant to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). EGFR TKI-resistant NSCLC patients (n=29) and corresponding tumor specimens, and 53 samples of postoperative TKI-naive NSCLC patients were collected. EGFR exon 19, 20, and 21 mutations were analyzed. And c-MET gene copy number was determined. The EGFR T790M mutation in exon 20 was not detected in the population of 53 TKI-naive patients, but found in 48.3% (14/29) of the enrolled TKI-resistant patients. c-MET was amplified in 3.8% (2/53) of the TKI-naive NSCLC patients and highly amplified in 17.2% (5/29) of the cohort. Most of T790M mutations were frequently associated with non-smoker, adenocarcinoma and EGFR activating mutations. Three male patients with T790M mutation occurred with wild-type EGFR, and were resistant to the treatments following TKI resistance. Features of c-MET amplification in TKI-naive patients were indistinguishable from TKI-resistant patients. In the group of wild-type EGFR, patients with T790M mutation had median progression free survival (PFS) and overall survival (OS) as 9.6 months and 12.6 months, respectively; whereas the median PFS and OS of c-MET amplified patients was 4.1 months and 8.0 months, respectively. These results suggest that EGFR T790M mutation and c-MET amplification can occur in TKI-resistant NSCLC with wild-type EGFR, and these genetic defects might be related to different survival outcome. c-MET amplification in TKI-naive or -resistant patients might share similarities in clinicopathologic features. C1 [Chen, Hua-Jun] Sun Yat-Sen University, Cancer CenterGuangzhou, China. [Mok, S Tony] Chinese University of Hong Kong, Department of Clinical Oncology, Shatin, New TerritoriesHong Kong, Hong Kong. [Chen, Zhi-Hong] Guangdong General Hospital, Guangdong Lung Cancer Institute, 106 Zhongshan Er Road, 510080 Guangzhou, Guangdong, China. [Guo, Ai-Lin] Guangdong Academy of Medical Sciences, Department of BiochipGuangzhou, China. [Zhang, Xu-Chao] Guangdong Academy of Medical Sciences, Department of BiochipGuangzhou, China. [Su, Jian] Guangdong General Hospital, Guangdong Lung Cancer Institute, 106 Zhongshan Er Road, 510080 Guangzhou, Guangdong, China. [Wu, Yi-Long] Guangdong General Hospital, Guangdong Lung Cancer Institute, 106 Zhongshan Er Road, 510080 Guangzhou, Guangdong, China. RP Wu, YL (reprint author), Guangdong General Hospital, Guangdong Lung Cancer Institute, 510080 Guangzhou, China. EM syylwu@live.cn CR Pao W, Miller VA, Politi KA et al, 2005, Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med 2:e73 Pao W, Balak MN, Riely GJ et al, 2006, Molecular analysis of NSCLC patients with acquired resistance to gefitinib or erlotinib. J Clin Oncol 24(Suppl):383s abstract 7078 Kwak EL, Sordella R, Bell DW et al, 2005, Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proc Natl Acad Sci USA 102:7665–7670 Kobayashi S, Boggon TJ, Dayaram T et al, 2005, EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med 352:786–792 Yun CH, Mengwasser KE, Toms AV et al, 2008, The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci USA 105:2070–2075 Engelman JA, Zejnullahu K, Mitsudomi T et al, 2007, MET amplification leads to gefitinib resistance in lung cancer by activating ErbB3 signaling. Science 316:1039–1043 Ogino A, Kitao H, Hirano S et al, 2007, Emergence of epidermal growth factor receptor T790M mutation during chronic exposure to gefitinib in a non small cell lung cancer cell line. Cancer Res 67:7807–7814 Engelman JA, Zejnullahu K, Mitsudomi Tet al, 2006, Allelic dilution obscures detection of a biologically significant resistance mutation in EGFR-amplified lung cancer. J Clin Invest 116:2695–2706 Wu YL, Zhong WZ, Li LY et al, 2007, Epidermal growth factor receptor mutations and their correlation with gefitinib therapy in patients with non-small cell lung cancer: a meta-analysis based on updated individual patient data from six medical centers in mainland China. J Thorac Oncol 2(5):430–439 Beasley MB, Brambilla E, Travis WD, 2005, The 2004 World Health Organization classification of lung tumors. Semin Roentgenol 40:90–97 Ling G, Persson A, Berne B et al, 2001, Persistent p53 mutations in single cells from normal human skin. Am J Pathol 159:1247–1253 Dietmaier W, Hartmann A, Wallinger S et al, 1999, Multiple mutation analyses in single tumor cells with improved whole genome amplification. Am J Pathol 154:83–95 Bae NC, Chae MH, Lee MH et al, 2007, EGFR, ERBB2, and KRAS mutations in Korean non-small cell lung cancer patients. Cancer Genet Cytogenet 173:107–113 Pallis AG, Voutsina A, Kalikaki A et al, 2007, ‘Classical’ but not ‘other’ mutations of EGFR kinase domain are associated with clinical outcome in gefitinib-treated patients with non-small cell lung cancer. Br J Cancer 97:1560–1566 Kimura H, Fujiwara Y, Sone T et al, 2006, High sensitivity detection of epidermal growth factor receptor mutations in the pleural effusion of non-small cell lung cancer patients. Cancer Sci 97:642–648 Kimura H, Suminoe M, Kasahara K et al, 2007, Evaluation of epidermal growth factor receptor mutation status in serum DNA as a predictor of response to gefitinib, IRESSA). Br J Cancer 97:778–784 Beau-Faller M, Ruppert AM, Voegeli AC et al, 2008, MET gene copy number in non-small cell lung cancer: molecular analysis in a targeted tyrosine kinase inhibitor naive cohort. J Thorac Oncol 3:331–339 Therasse P, Arbuck SG, Eisenhauer EA et al, 2000, New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst 92(3):205–216 Balak MN, Gong Y, Riely GJ et al, 2006, Novel D761Y and common secondary T790M mutations in epidermal growth factor receptor-mutant lung adenocarcinomas with acquired resistance to kinase inhibitors. Clin Cancer Res 12:6494–6501 Kosaka T, Yatabe Y, Endoh H et al, 2006, Analysis of epidermal growth factor receptor gene mutation in patients with non small cell lung cancer and acquired resistance to gefitinib. Clin Cancer Res 12:5764–5769 Newton CR, Graham A, Heptinstall LE et al, 1989, Analysis of any point mutation in DNA. The amplification refractory mutation system, ARMS). Nucleic Acids Res 17:2503–2516 Whitcombe D, Theaker J, Guy SP et al, 1999, Detection of PCR products using self-probing amplicons and fluorescence. Nat Biotechnol 17:804–807 Bean J, Brennan C, Shih JY et al, 2007, MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci USA 104:20932–20937 Inukai M, Toyooka S, Ito S et al, 2006, Presence of epidermal growth factor receptor gene T790M mutation as a minor clone in non-small cell lung cancer. Cancer Res 66:7854–7858 Shih JY, Gow CH, Yang PC, 2005, EGFR mutation conferring primary resistance to gefitinib in non-small-cell lung cancer. N Engl J Med 353(2):207–208 Toyooka S, Kiura K, Mitsudomi T, 2005, EGFR mutation and response of lung cancer to gefitinib. N Engl J Med 352(20):2136 author reply 2136 Bell DW, Gore I, Okimoto RA et al, 2005, Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR. Nat Genet 37(12):1315–1316 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 651 EP 658 DI 10.1007/s12253-009-9167-8 PG 8 ER PT J AU Carlos-Bregni, R Vidaurre, CE Netto, CA Leon, EJ Almeida, PO AF Carlos-Bregni, Roman Vidaurre, C Elisa Netto, Carolina Ana Leon, E Jorge Almeida, P Oslei TI Primary Intraosseous Adenoid Cystic Carcinoma of the Mandible: Histopathological and Immunohistochemical Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Adenoid cystic carcinoma; Intraosseous; Mandible; Immunohistochemistry ID Adenoid cystic carcinoma; Intraosseous; Mandible; Immunohistochemistry AB Primary intraosseous salivary gland tumors of the mandible are rare, with mucopidermoid carcinoma being the most frequent, followed by adenoid cystic carcinoma (ACC). We present a case of a central ACC involving the mandible of a 46-year-old man. He presented an indurated swelling on the vestibular aspect of the left mandibular body and ipsilateral paraesthesia of the lower lip. A panoramic radiography revealed a large radiolucent area, with irregular margins, involving the body and ramus of the left mandible, and CT scan confirmed that the lesion was confined within the mandibular bone. The histopathological features were of an ACC. CT scan also revealed multiple nodular lesions in both lungs suggestive of metastases. The patient was surgically treated by hemimandibulectomy. The patient is well with no evidences of recurrences in the mandible. The present case shows that the clinical and immunohistochemical profile of primary intraosseous ACC is similar to what is found in ACC involving the salivary glands. C1 [Carlos-Bregni, Roman] Centro Clinico de Cabeza y Cuello, 6 ave. 7-39 zona 10, ed. Las Brisas of. 501,, 01010 Ciudad de Guatemala, Guatemala. [Vidaurre, C Elisa] Centro Clinico de Cabeza y Cuello, 6 ave. 7-39 zona 10, ed. Las Brisas of. 501,, 01010 Ciudad de Guatemala, Guatemala. [Netto, Carolina Ana] Centro Clinico de Cabeza y Cuello, 6 ave. 7-39 zona 10, ed. Las Brisas of. 501,, 01010 Ciudad de Guatemala, Guatemala. [Leon, E Jorge] State University of Campinas, School of Dentistry, Department of Oral Diagnosis, Av. Limeira 901, Caixa Postal 52, 13414-903 Piracicaba, Brazil. [Almeida, P Oslei] State University of Campinas, School of Dentistry, Department of Oral Diagnosis, Av. Limeira 901, Caixa Postal 52, 13414-903 Piracicaba, Brazil. RP Leon, EJ (reprint author), State University of Campinas, School of Dentistry, Department of Oral Diagnosis, 13414-903 Piracicaba, Brazil. EM jorgeesquiche@yahoo.com.br CR Bumstead WD, 1955, Cylindroma of the mandible. Oral Surg 8:546 Brookstone MS, Huvos AG, 1992, Central salivary gland tumors of the maxilla and mandible: a clinicopathologic study of 11 cases with an analysis of the literature. J Oral Maxillofac Surg 50:229– 236 Favia G, Maiorano E, Orsini G et al, 2000, Central, intraosseous, adenoid cystic carcinoma of the mandible: report of a case with periapical involvement. J Endod 12:760–763 Martinez-Madrigal F, Pineda-Daboin K, Casiraghi O et al, 2000, Salivary gland tumors of the mandible. Ann Diagn Pathol 4:347– 353 Capodiferro S, Scully C, Macaita MG et al, 2005, Bilateral intraosseous adenoid cystic carcinoma of the mandible: report of a case with lung metastases at first clinical presentation. Oral Dis 11:109–12 Al-Sukhun J, Lindqvist C, Hietanen J et al, 2006, Central adenoid cystic carcinoma of the mandible: case report and literature review of 16 cases. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 101:304–308 Garcia de Marcos JA, Calderon-Polanco J, Poblet E et al, 2008, Primary adenoid cystic carcinoma of the mandible: case report and review of the literature. J Oral Maxillofac Surg 66:2609–2615 Mahomed F, Altini M, Meer S, et al, 2009, Central adenoid cystic carcinoma of the mandible with odontogenic features: Report of a case. Head Neck. Feb 2. [Epub ahead of print] Li Y, Li LJ, Huang J et al, 2008, Central malignant salivary gland tumors of the jaw: retrospective clinical analysis of 22 cases. J Oral Maxillofac Surg 66:2247–2253 Ojha J, Bhattacharyya I, Islam MN et al, 2007, Intraosseous pleomorphic adenoma of the mandible: report of a case and review of the literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 104:21–26 Chen JC, Gnepp DR, Bedrossian CW, 1988, Adenoid cystic carcinoma of the salivary glands: an immunohistochemical analysis. Oral Surg Oral Med Oral Pathol 65:316–326 Araujo VC, Loducca SV, Sousa SO et al, 2001, The cribriform features of adenoid cystic carcinoma and polymorphous low-grade adenocarcinoma: cytokeratin and integrin expression. Ann Diagn Pathol 5:330–334 Foschini MP, Gaiba A, Cocchi R et al, 2005, p63 expression in salivary gland tumors: role of DeltaNp73L in neoplastic transformation. Int J Surg Pathol 13:329–335 Ito K, Ito T, Tsukuda M et al, 1993, An immunohistochemical study of adenoid cystic carcinoma of the external auditory canal. Eur Arch Otorhinolaryngol 250:240–246 Bergman R, Lichtig C, Moscona RA et al, 1991, A comparative immunohistochemical study of adenoid cystic carcinoma of the skin and salivary glands. Am J Dermatopathol 13:162–168 Moran CA, Suster S, Koss MN, 1994, Primary adenoid cystic carcinoma of the lung. A clinicopathologic and immunohistochemical study of 16 cases. Cancer 73:1390–1397 Luo XL, Sun MY, Lu CT et al, 2006, The role of Schwann cell differentiation in perineural invasion of adenoid cystic and mucoepidermoid carcinoma of the salivary glands. Int J Oral Maxillofac Surg 35:733–739 Daa T, Kaku N, Kashima K et al, 2005, Expression of betacatenin, E-cadherin and cyclin D1 in adenoid cystic carcinoma of the salivary gland. J Exp Clin Cancer Res 24:83–87 Edwards PC, Bhuiya T, Kelsch RD, 2003, C-kit expression in the salivary gland neoplasms adenoid cystic carcinoma, polymorphous low-grade adenocarcinoma, and monomorphic adenoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 95:586–593 Carlinfante G, Lazzaretti M, Ferrari S et al, 2005, P53, bcl-2 and Ki-67 expression in adenoid cystic carcinoma of the palate. A clinico-pathologic study of 21 cases with long-term follow-up. Pathol Res Pract 200:791–799 Vargas PA, Cheng Y, Barrett AWet al, 2008, Expression of Mcm- 2, Ki-67 and geminin in benign and malignant salivary gland tumours. J Oral Pathol Med 37:309–318 Woo VL, Bhuiya T, Kelsch R, 2006, Assessment of CD43 expression in adenoid cystic carcinomas, polymorphous low-grade adenocarcinomas, and monomorphic adenomas. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 102:495–500 Soares AB, Ponchio L, Juliano PB et al, 2007, Lymphatic vascular density and lymphangiogenesis during tumour progression of carcinoma ex pleomorphic adenoma. J Clin Pathol 60:995–1000 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 659 EP 664 DI 10.1007/s12253-009-9168-7 PG 6 ER PT J AU Kadota, K Haba, R Kushida, Y Katsuki, N Hayashi, T Miyai, Y Bando, K Taoka, R Kakehi, Y AF Kadota, Kyuichi Haba, Reiji Kushida, Yoshio Katsuki, Naomi Hayashi, Toshitetsu Miyai, Yumi Bando, Kenji Taoka, Rikiya Kakehi, Yoshiyuki TI Adult Extrarenal Wilms’ Tumor Mimicking Mixed Epithelial and Stromal Tumor in the Retroperitoneum: A Case Report with Immunohistochemical Study and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Adult; Extrarenal; Retroperitoneum; Mixed epithelial and stromal tumor; Wilms’ tumor ID Adult; Extrarenal; Retroperitoneum; Mixed epithelial and stromal tumor; Wilms’ tumor AB We report an extremely rare case of adult extrarenal Wilms’ tumor (WT) in a 52-year-old woman who presented with fever and abdominal distension. Computed tomography revealed a well-defined mass lesion measuring 15.0 cm in the right retroperitoneum and that was in contact with the right kidney. The mass and kidney were surgically removed. Grossly, the mass was well-defined, measuring 16.3×11.0×9.8 cm, and appearing grayish-white in color. The border between the mass and the kidney was welldefined. Histologically, the tumor showed a triphasic pattern consisting of stromal, epithelial and blastemal components. The stromal component was predominant in the tumor and consisted both of spindle cells and smooth muscle cells. The epithelial component showed a mature glandular structure. Immunohistochemically, the stromal component was positive for vimentin, smooth muscle actin and desmin. The blastemal component was positive for vimentin, while the epithelial component was positive for cytokeratin (CK) 18, CK7 and vimentin. WT-1 was negative in the all three components, and the Ki-67 proliferation index was low. The postoperative histopathological diagnosis indicated extrarenal WT arising in the retroperitoneum. Although not treated by either chemotherapy or radiation therapy, she was free from disease recurrence for 30 months after surgery. To the best of our knowledge, this report is only the fourth case of adult extrarenal WT arising in the retroperitoneum. Furthermore, the present case showed predominant smooth muscle differentiation and a mature glandular structure, mimicking a mixed epithelial and stromal tumor. C1 [Kadota, Kyuichi] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Haba, Reiji] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Kushida, Yoshio] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Katsuki, Naomi] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Hayashi, Toshitetsu] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Miyai, Yumi] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Bando, Kenji] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Taoka, Rikiya] Kagawa University, Faculty of Medicine, Department of UrologyKagawa, Japan. [Kakehi, Yoshiyuki] Kagawa University, Faculty of Medicine, Department of UrologyKagawa, Japan. RP Kadota, K (reprint author), Kagawa University, University Hospital, Department of Diagnostic Pathology, 761-0793 Kagawa, Japan. EM qichi@med.kagawa-u.ac.jp CR Terenziani M, Spreafico F, Collini P et al, 2004, Adult Wilms’ tumor: A monoinstitutional experience and a review of the literature. Cancer 101:289–293 Muc RS, Grayson W, Grobbelaar JJ, 2001, Adult extrarenal Wilms tumor occurring in the uterus. Arch Pathol LabMed 125:1081–1083 Isaac MA, Vijayalakshmi S, Madhu CS et al, 2000, Pure cystic WT of the ovary with a review of extrarenal Wilms’ tumors. Hum Pathol 31:761–764 Koretz MJ, Wang S, Klein FA et al, 1987, Extrarenal adult Wilms’ tumor. Cancer 60:2484–2488 Fukutomi Y, Shibuya C, Yamamoto S et al, 1988, Extrarenal Wilms’ tumor in the adult patient. A case report and review of the world literature. Am J Clin Pathol 90:618–622 Ratnam GV, Abu-Eshy S, Morad N et al, 2006, Adult extrarenal Wilms’ tumour: A case report and review of literature. West Afr J Med 25:75–78 Gillis AJ, Oosterhuis JW, Schipper ME et al, 1994, Origin and biology of a testicular Wilms’ tumor. Genes Chromosomes Cancer 11:126–135 Adsay NV, Eble JN, Srigley JR et al, 2000, Mixed epithelial and stromal tumor of the kidney. Am J Surg Pathol 24:958–970 Ghanem MA, Van der Kwast TH, Den Hollander JC et al, 2000, Expression and prognostic value of Wilms’ tumor 1 and early growth response 1 proteins in WT. Clin Cancer Res 6:4265–4271 Vasei M, Moch H, Mousavi A et al, 2008, Immunohistochemical profiling of Wilms tumor: a tissue microarray study. Appl Immunohistochem Mol Morphol 16:128–134 Roberts DJ, Haber D, Sklar J et al, 1993, Extrarenal Wilms’ tumors. A study of their relationship with classical renal Wilms’ tumor using expression of WT-1 as a molecular marker. Lab Invest 68:528–536 Reinhard H, Aliani S, Ruebe C et al, 2004, Wilms’ tumor in adults: Results of the Society of Pediatric Oncology, SIOP, 93– 01/Society for Pediatric Oncology and Hematology, GPOH, Study. J Clin Oncol 22:4500–4506 Coppes MJ, Wilson PC, Weitzman S, 1991, Extrarenal Wilms’ tumor: staging, treatment, and prognosis. J Clin Oncol 9:167–174 Ghanem MA, Van der Kwast TH, Sudaryo MK et al, 2004, MIB-1, KI-67, proliferation index and cyclin-dependent kinase inhibitor p27(Kip1, protein expression in nephroblastoma. Clin Cancer Res 10:591–597 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 665 EP 669 DI 10.1007/s12253-009-9169-6 PG 5 ER PT J AU Chua, CT Yao, P Akther, J Young, L Bao, Sh Samaraweera, U Yan, DT Morris, LD AF Chua, C Terence Yao, Peng Akther, Javed Young, Lawrence Bao, Shisan Samaraweera, Ushma Yan, D Tristan Morris, L David TI Differential Expression of Ki-67 and Sex Steroid Hormone Receptors Between Genders in Peritoneal Mesothelioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mesothelioma; Ki-67 antigen; Estrogen receptor; Progesterone receptor; Androgen receptor; Cytoreductive surgery ID Mesothelioma; Ki-67 antigen; Estrogen receptor; Progesterone receptor; Androgen receptor; Cytoreductive surgery AB Gender influence on survival in mesothelioma has been observed in several large clinical series. However, this gender effect has not been investigated. Female patients often have less aggressive tumors and survive longer. However, few studies in the literature have explained the molecular basis of this finding. Understanding this difference at a molecular level may offer the hope of improving survival via hormonal manipulation.We investigate the expression of Ki-67 and sex steroid receptors; estrogen receptors (ER), progesterone receptors (PR) and androgen receptors (AR) to elucidate any pathognomonic difference that characterize this gender difference. Positive expression of markers was observed in 95% (Ki-67), 80% (ER), 100% (PR) and 65% (AR) of patients. Expression of markers between gender showed a higher Ki-67 in males (M=1.3%, F=0.6%), higher estrogen receptor in females (M=0.6%, F=1.7%) and higher progesterone receptor in females (M=1.0%, F=1.4%). Twenty patients were treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in our peritonectomy unit. Paraffin sections of the tumor specimens were retrieved for immunohistochemical analysis. The immunostaining was performed using monoclonal mouse anti-human antibodies on an autostainer (Autostainer Plus; Dako, Inc.). The intensity of the stains were quantified using the Image-Pro Plus (IPP) 4.5 (Media Cybernetics, Silver Spring, MD). For the first time, we demonstrate the presence of sex steroid receptors in peritoneal mesothelioma. Once the exact functional effects of these receptors are understood, the use of established therapeutic options that are clinically available to target the sex steroid pathway may become a reality. C1 [Chua, C Terence] St George Hospital, NSW 2217 Kogarah, Sydney, Australia. [Yao, Peng] St George Hospital, NSW 2217 Kogarah, Sydney, Australia. [Akther, Javed] St George Hospital, NSW 2217 Kogarah, Sydney, Australia. [Young, Lawrence] St George Hospital, NSW 2217 Kogarah, Sydney, Australia. [Bao, Shisan] University of Sydney, School of Medical Sciences and Bosch Institute, Discipline of Pathology, NSW 2006 Sydney, Sydney, Australia. [Samaraweera, Ushma] Prince of Wales Hospital, Department of Pathology, NSW 2031 Randwick, Sydney, Australia. [Yan, D Tristan] St George Hospital, NSW 2217 Kogarah, Sydney, Australia. [Morris, L David] St George Hospital, NSW 2217 Kogarah, Sydney, Australia. RP Morris, LD (reprint author), St George Hospital, NSW 2217 Kogarah, Australia. EM david.morris@unsw.edu.au CR Boffetta P, 2007, Epidemiology of peritoneal mesothelioma: a review. Ann. Oncol. 18:985–90 RobinsonBW, Lake RA(2005, Advances in malignantmesothelioma. N Engl J Med. 353:1591–603 Sugarbaker PH, Welch LS, Mohamed F, Glehen OA, 2003, Review of peritoneal mesothelioma at theWashington Cancer Institute. Surg Oncol Clin N Am. 12:605–621 Acherman YI, Welch LS, Bromley CM, Sugarbaker PH, 2003, Clinical presentation of peritoneal mesothelioma. Tumori 89(3):269–73 Castagneto B, Botta M, Aitini E, Spigno F, Degiovanni D, Alabiso O et al, 2008, Phase II study of pemetrexed in combination with carboplatin in patients with malignant pleural mesothelioma, MPM). Ann. Oncol. 19(2):370–3 Vogelzang NJ, Rusthoven JJ, Symanowski J, Denham C, Kaukel E, Ruffie P et al, 2003, Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma. J Clin Oncol 21(14):2636–44 Janne PA, Wozniak AJ, Belani CP, Keohan M-L, Ross HJ, Polikoff JA et al, 2005, Open-label study of pemetrexed alone or in combination with cisplatin for the treatment of patients with peritoneal mesothelioma: outcomes of an expanded access program. Clin Lung Cancer 7(1):40–6 Yan TD, Welch L, Black D, Sugarbaker PH, 2007, A systematic review on the efficacy of cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for diffuse malignancy peritoneal mesothelioma. Ann. Oncol. 18(5):827–34 Johansson L, Linden CJ, 1996, Aspects of histopathologic subtype as a prognostic factor in 85 pleural mesotheliomas. Chest 109(1):109–14 Mineo TC, Ambrogi V, Pompeo E, Baldi A, Stella F, Aurea P et al, 2008, The value of occult disease in resection margin and lymph node after extrapleural pneumonectomy formalignant mesothelioma. Ann Thorac Surg 85(5):1740–6 Edwards JG, Stewart DJ, Martin-Ucar A, Muller S, Richards C, Waller DA, 2006, The pattern of lymph node involvement influences outcome after extrapleural pneumonectomy for malignant mesothelioma. J Thorac Cardiovasc Surg 131(5):981–7 Yan TD, Yoo D, Sugarbaker PH, 2006, Significance of lymph node metastasis in patients with diffuse malignant peritoneal mesothelioma. Eur. J. Surg. Oncol. 32(9):948–53 Curran D, Sahmoud T, Therasse P, van Meerbeeck J, Postmus PE, Giaccone G, 1998, Prognostic factors in patients with pleural mesothelioma: the European Organization for Research and Treatment of Cancer experience. J Clin Oncol 16(1):145–52 Adams VI, Unni KK, Muhm JR, Jett JR, Ilstrup DM, Bernatz PE, 1986, Diffuse malignant mesothelioma of pleura. Diagnosis and survival in 92 cases. Cancer 58(7):1540–51 Metintas M, Metintas S, Ucgun I, Gibbs AR, Harmanci E, Alatas F et al, 2001, Prognostic factors in diffuse malignant pleural mesothelioma: effects of pretreatment clinical and laboratory characteristics. Respir. Med. 95(10):829–35 Yan TD, Popa E, Brun EA, Cerruto CA, Sugarbaker PH, 2006, Sex difference in diffuse malignant peritoneal mesothelioma. Br. J. Surg. 93(12):1536–42 Antman KH, Blum RH, Greenberger JS, Flowerdew G, Skarin AT, Canellos GP, 1980, Multimodality therapy for malignant mesothelioma based on a study of natural history. Am. J. Med. 68, 3):356–62 Flores RM, Pass HI, Seshan VE, Dycoco J, Zakowski M, Carbone M et al, 2008, Extrapleural pneumonectomy versus pleurectomy/ decortication in the surgical management of malignant pleural mesothelioma: results in 663 patients. Journal of Thoracic & Cardiovascular Surgery 135(3):620–6 Neumann V, Rutten A, Scharmach M, Muller KM, Fischer M, 2004, Factors influencing long-term survival in mesothelioma patients–results of the German mesothelioma register. International Archives of Occupational & Environmental Health 77(3):191–9 Rusch VW, Venkatraman ES, 1999, Important prognostic factors in patients with malignant pleural mesothelioma, managed surgically. Ann. Thorac. Surg. 68(5):1799–804 Carson-Jurica MA, Schrader WT, O’Malley BW, 1990, Steroid receptor family: structure and functions. Endocr. Rev. 11:201–20 McKenna NJ, Lanz RB, O’Malley BW, 1999, Nuclear receptor coregulators: cellular and molecular biology. Endocr. Rev. 20:321–44 Sugarbaker PH, 1995, Peritonectomy procedures. Ann. Surg. 221:29–42 Garcia-Carbonero R, Paz-Ares L, 2006, Systemic chemotherapy in the management of malignant peritoneal mesothelioma. Eur. J. Surg. Oncol. 32(6):676–81 Thomas Scholzen JG, 2000, The Ki-67 protein: From the known and the unknown. J Cell Physiol 182(3):311–22 Smolle J, Hoffmann-Wellenhof R, Soyer H-P, Stettner H, Kerl H, 1989, Nuclear Size and Shape Parameters Correlate with Proliferative Activity in Cutaneous Melanocytic Tumors. J Invest Dermatol 93(1):178–82 Gao X, Loggie BW, Nawaz Z, 2002, The roles of sex steroid receptor coregulators in cancer. Molecular Cancer. 1:7 Schiff R, Saw F, 2002, The importance of the estrogen receptor in breast cancer. In: Pasqualini JR, ed, Breast cancer: prognosis, treatment, and prevention. Marcel Dekker, New York, NY, pp 149–86 Rowan BG, O’Malley BW, 2000, Progesterone receptor coactivators. Steroids 65(10–11):545–9 Heemers HV, Tindall DJ, 2007, Androgen receptor, AR, coregulators: a diversity of functions converging on and regulating the AR transcriptional complex. Endocr. Rev. 28(7):778–808 Ryan PD, Goss PE, 2008, The emerging role of the insulin-like growth factor pathway as a therapeutic target in cancer. Oncologist 13(1):16–24 Barnetson RJ, Burnett RA, Downie I, Harper CM, Roberts F, 2006, Immunohistochemical analysis of peritoneal mesothelioma and primary and secondary serous carcinoma of the peritoneum: antibodies to estrogen and progesterone receptors are useful. Am. J. Clin. Pathol. 125(1):67–76 Horita K, Inase N, Miyake S, Formby B, Toyoda H, Yoshizawa Y, 2001, Progesterone induces apoptosis in malignant mesothelioma cells. Anticancer Res. 21(6A):3871–4 Sawh RN, Malpica A, Deavers MT, Liu J, Silva EG, 2003, Benign cystic mesothelioma of the peritoneum: a clinicopathologic study of 17 cases and immunohistochemical analysis of estrogen and progesterone receptor status. Hum. Pathol. 34(4):369–74 Fisher B, Costantino J, Redmond C, Poisson R, Bowman D, Couture J et al, 1989, A randomized clinical trial evaluating tamoxifen in the treatment of patients with node-negative breast cancer who have estrogen-receptor-positive tumors. N Engl J Med 320(8):479–84 Elledge RM, Allred DC, 2004, Clinical aspects of estrogen and progesterone receptors. In: Harris JR, Lippman ME, Morrow M, eds, Diseases of the Breast, 3rd edn. Lippincott Williams and Wilkins, Philadelphia, PA, pp 603–17 Cui X, Schiff R, Arpino G, Osborne CK, Lee AV, 2005, Biology of progesterone receptor loss in breast cancer and its implications for endocrine therapy. J Clin Oncol 23(30):7721–35 Smith CL, O'Malley BW, 2004, Coregulator Function: A Key to Understanding Tissue Specificity of Selective Receptor Modulators. Endocr. Rev. 25(1):45–71 d'Ancona FCH, Debruyne FMJ, 2005, Endocrine approaches in the therapy of prostate carcinoma. Hum Reprod Update 11, 3):309–17 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 671 EP 678 DI 10.1007/s12253-009-9170-0 PG 8 ER PT J AU Lu, M Ma, J Xue, W Cheng, Ch Wang, Y Zhao, Y Ke, Q Liu, H Liu, Y Li, P Cui, X He, S Shen, A AF Lu, Mudan Ma, Jianbo Xue, Wenqun Cheng, Chun Wang, You Zhao, Yueming Ke, Qing Liu, Haiou Liu, Yonghua Li, Peng Cui, Xiaopeng He, Song Shen, Aiguo TI The Expression and Prognosis of FOXO3a and Skp2 in Human Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human hepatocellular carcinoma; FOXO protein FOXO3a; S-phase kinase protein (Skp2); Huh7; Immunohistochemistry (IHC); Prognosis ID Human hepatocellular carcinoma; FOXO protein FOXO3a; S-phase kinase protein (Skp2); Huh7; Immunohistochemistry (IHC); Prognosis AB The forkhead box proteins (FOXO proteins) comprise a large family of functionally diverse transcription factors involved in cellular proliferation, transformation, differentiation and longevity. Recently, ubiquitination and proteasome degradation of FOXO3a have been reported. In this study, we investigated the role of FOXO3a and Skp2 in human hepatocellular carcinoma progression. Immunohistochemical analysis was performed on formalin-fixed paraffin sections of 91 specimens. Furthermore in vitro, western-blot analysis and protein stabilization studies were used to study the relationship between FOXO3a and Skp2. We found that the expression of FOXO3a was negatively related with Skp2 expression (r=−0.583; p<0.05) and FOXO3a expression correlated significantly with histological grade (p=0.000), cirrhosis (p=0.015), and tumor size (p=0.043) while Skp2 expression correlated significantly with histological grade (p=0.000) and tumor size (p=0.005). Kaplan-Meier analysis revealed that survival curves of low versus high expressers of FOXO3a and Skp2 showed a highly significant separation in HCC (p<0.01). Our results suggested that FOXO3a and Skp2 may be considered to be important prognosis in human hepatocellular carcinoma. In vitro studies suggested that the degradation of FOXO3a may dependent on the expression of Skp2 in the proliferated Huh7 cells. C1 [Lu, Mudan] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, China. [Ma, Jianbo] Affiliated Hospital of Nantong University, Department of Surgery, 226001 Nantong, China. [Xue, Wenqun] Wuxi Maternal and Child Health Hospital, 214002 Wuxi, China. [Cheng, Chun] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, China. [Wang, You] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, China. [Zhao, Yueming] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, China. [Ke, Qing] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, China. [Liu, Haiou] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, China. [Liu, Yonghua] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, China. [Li, Peng] Affiliated Hospital of Nantong University, Department of Surgery, 226001 Nantong, China. [Cui, Xiaopeng] Affiliated Hospital of Nantong University, Department of Surgery, 226001 Nantong, China. [He, Song] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, China. [Shen, Aiguo] Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, 19 Qi-xiu Road, 226001 Nantong, Jiangsu, China. RP Shen, A (reprint author), Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, 226001 Nantong, China. EM Shen_aiguo@yahoo.com CR Anderson MJ, Viars CS, Czekay S, Cavenee WK, Arden KC, 1998, Cloning and characterization of three human forkhead genes that comprise an FKHR-like gene subfamily. Genomics 47:187–199 Aoki M, Jiang H, Vogt PK, 2004, Proteasomal degradation of the FoxO1 transcriptional regulator in cells transformed by the P3k and Akt oncoproteins. Proc Natl Acad Sci U S A 101:13613– 13617 Borkhardt A, Repp R, Haas OA, Leis T, Harbott J, Kreuder J, Hammermann J et al, 1997, Cloning and characterization of AFX, the gene that fuses to MLL in acute leukemias with a t(X;11)(q13;q23). Oncogene 14:195–202 Bosch FX, Ribes J, Borras J, 1999, Epidemiology of primary liver cancer. Semin Liver Dis 19:271–285 Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, Anderson MJ et al, 1999, Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96:857–868 Chakravarthy MV, Abraha TW, Schwartz RJ, Fiorotto ML, Booth FW, 2000, Insulin-like growth factor-I extends in vitro replicative life span of skeletal muscle satellite cells by enhancing G1/S cell cycle progression via the activation of phosphatidylinositol 3'- kinase/Akt signaling pathway. J Biol Chem 275:35942–35952 el-Serag HB, 2001, vi. Epidemiology of hepatocellular carcinoma. Clin Liver Dis 5:87–107 Galili N, Davis RJ, Fredericks WJ, Mukhopadhyay S, Rauscher FJ 3rd, Emanuel BS, Rovera G et al, 1993, Fusion of a fork head domain gene to PAX3 in the solid tumour alveolar rhabdomyosarcoma. Nat Genet 5:230–235 Gilley J, Coffer PJ, Ham J, 2003, FOXO transcription factors directly activate bim gene expression and promote apoptosis in sympathetic neurons. J Cell Biol 162:613–622 Greer EL, Brunet A, 2005, FOXO transcription factors at the interface between longevity and tumor suppression. Oncogene 24:7410–7425 Hromas R, Costa R, 1995, The hepatocyte nuclear factor-3/ forkhead transcription regulatory family in development, inflammation, and neoplasia. Crit Rev Oncol Hematol 20:129–140 HuMC, Lee DF, XiaW, Golfman LS, Ou-Yang F, Yang JY, Zou Yet al, 2004, IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a. Cell 117:225–237 Huang H, Regan KM, Wang F, Wang D, Smith DI, van Deursen JM, Tindall DJ, 2005, Skp2 inhibits FOXO1 in tumor suppression through ubiquitin-mediated degradation. Proc Natl Acad Sci U S A 102:1649–1654 Jacobs FM, van der Heide LP, Wijchers PJ, Burbach JP, Hoekman MF, Smidt MP, 2003, FoxO6, a novel member of the FoxO class of transcription factors with distinct shuttling dynamics. J Biol Chem 278:35959–35967 Kaufmann E, Knochel W, 1996, Five years on the wings of fork head. Mech Dev 57:3–20 Lai E, Clark KL, Burley SK, Darnell JE Jr, 1993, Hepatocyte nuclear factor 3/fork head or "winged helix" proteins: a family of transcription factors of diverse biologic function. Proc Natl Acad Sci U S A 90:10421–104233 Matsuzaki H, Daitoku H, Hatta M, Tanaka K, Fukamizu A, 2003, Insulin-induced phosphorylation of FKHR, Foxo1, targets to proteasomal degradation. Proc Natl Acad Sci U S A 100:11285– 11290 Medema RH, Kops GJ, Bos JL, Burgering BM, 2000, AFX-like Forkhead transcription factors mediate cell-cycle regulation by Ras and PKB through p27kip1. Nature 404:782–787 Modur V, Nagarajan R, Evers BM, Milbrandt J, 2002, FOXO proteins regulate tumor necrosis factor-related apoptosis inducing ligand expression. Implications for PTEN mutation in prostate cancer. J Biol Chem. 277:47928–47937 Parkin DM, Bray F, Ferlay J, Pisani P, 2001, Estimating the world cancer burden: Globocan 2000. Int J Cancer 94:153–156 Plas DR, Thompson CB, 2003, Akt activation promotes degradation of tuberin and FOXO3a via the proteasome. J Biol Chem 278:12361–12366 Potente M, Urbich C, Sasaki K, Hofmann WK, Heeschen C, Aicher A, Kollipara R et al, 2005, Involvement of Foxo transcription factors in angiogenesis and postnatal neovascularization. J Clin Invest 115:2382–2392 Ramaswamy S, Nakamura N, Sansal I, Bergeron L, Sellers WR, 2002, A novel mechanismof gene regulation and tumor suppression by the transcription factor FKHR. Cancer Cell 2:81–91 Scheffner M, Werness BA, Huibregtse JM, Levine AJ, Howley PM, 1990, The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of p53. Cell 63:1129–1136 Xu X, Yamamoto H, Sakon M, Yasui M, Ngan CY, Fukunaga H, Morita T et al, 2003, Overexpression of CDC25A phosphatase is associated with hypergrowth activity and poor prognosis of human hepatocellular carcinomas. Clin Cancer Res 9:1764–1772 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 679 EP 687 DI 10.1007/s12253-009-9171-z PG 9 ER PT J AU Nagy, Zs Turcsik, V Blasko, Gy AF Nagy, Zsuzsanna Turcsik, Vera Blasko, Gyorgy TI The Effect of LMWH (Nadroparin) on Tumor Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE LMWH. Nadroparin; Tumor progression; Survival ID LMWH. Nadroparin; Tumor progression; Survival AB Recent clinical studies on patients with malignancies, who were treated with UHF and LMWHs raised the possibility, that these agents may possess an inhibitory effect on tumor progression. Further studies supported that this effect is independent from the anticoagulant and antithrombotic action. In this retrospective study oncological patients with an increased risk for thromboembolism were choosen, who received prophylactic treatment with an LMWH (nadroparin) at least for 6 months. Comparing with the control group, in some subgroups (T3 and T4, as well as M1) the LMWH-treated patients showed a significantly increased survival. C1 [Nagy, Zsuzsanna] St. Imre Hospital, Department Clinical Oncology, Tetenyi ut 12-15, 1116 Budapest, Hungary. [Turcsik, Vera] Governmental Health Center, Department Anaesthesiology and Intensive CareBudapest, Hungary. [Blasko, Gyorgy] University of Debrecen, Department of Pharmacology and PharmacotherapyDebrecen, Hungary. RP Nagy, Zs (reprint author), St. Imre Hospital, Department Clinical Oncology, 1116 Budapest, Hungary. EM zsulacz@chello.hu CR von Delius S, Ayvaz M, Wagenpfeil S et al, 2007, Effect of lowmolecular- weight heparin on survival in patients with advanced pancreatic adenocarcinoma. Thromb Haemost 98:434–439 Lazo-Lagner A, Goss GD, Spaans JN et al, 2007, The effect of low-molecular-weight heparin on cancer survival. A systematic review and meta-analysis of randomized trials. J Thrombosis Haemostasis 5:729–737 Kakkar AK, 2005, Low-molecular-weight heparin and survival in patients with malignant disease. Cancer Control 12:22–30 Akl EA, VanDoormaal FF, Barba M et al, 2008, Parenteral anticoagulation may prolong the survival of patients with limited small cell lung cancer: a Cochrane systematic review. J Exp Clin Cancer Res 27:4 Kakkar AK, Levine MN, Kadziola Z, 2004, Low molecular weight heparin, therapy with dalteparin, and survival in advanced cancer: the fragmin advanced malignancy outcome study, FAMOUS). J Clin Oncol 22:1944–1948 Tovari J, Bereczky B, Gilly R et al, 2004, Effect of heparin treatment on the metastatization of melanoma in a preclinical model, in Hungarian). Magy Onkol 48:235–241 De Lorenzo F, Dotsenko O, Scully MF et al, 2006, The Role of Anticoagulation in Cancer Patients: Facts and Figures. Anti- Cancer Agents Med Chem 6:579–587 Klerk CPW, Smorenburg SM, Otten HM et al, 2005, The Effect of Low Molecular Weight Heparin on Survival in Patients With Advanced Malignancy. J Clin Oncol 23:2130–2135 Li JP, 2008, Heparin, heparan sulfate and heparanase in cancer: remedy for metastasis? Anti-Cancer Agents Med Chem 8:64–76 Falanga A, Marchetti M, 2007, Heparin in tumor progression and metastatic dissemination. Semin Thromb Haemost 33:688– 694 Castelli R, Porro F, Tarsia P, 2004, The heparins and cancer: review of clinical trials and biological properties. Vasc Med 9:205–213 Vlodavsky I, Ilan N, Nadir Yet al, 2007, Heparanase, heparin and the coagulation system in cancer progression. Thromb Res 120:112–120 Casu B, Vlodavsky I, Sanderson RD, 2009, Pathophysiol Haemost Thromb 2007-08: 36, 195–203,, DOI 10.1159/000175157 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 689 EP 692 DI 10.1007/s12253-009-9204-7 PG 4 ER PT J AU Ding, W Ju, Sh Jiang, Sh Zhu, L Wang, Y Wang, H AF Ding, Weifeng Ju, Shaoqing Jiang, Shengyang Zhu, Li Wang, Yueguo Wang, Huimin TI Reduced APRIL Expression Induces Cellular Senescence via a HSPG-Dependent Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE A proliferation-inducing ligand; Cellular senescence; Short hairpin RNA; Heparan sulfate proteoglycans ID A proliferation-inducing ligand; Cellular senescence; Short hairpin RNA; Heparan sulfate proteoglycans AB APRIL, a member of the TNF superfamily, can induce cell proliferation and is overexpressed in most tumor tissues or cells. Nevertheless, it is still unknown about the effect of decreased levels of APRIL expression on tumor cells. In this study, we analyzed APRIL and HSPG expression in the colon carcinoma cell line, SW480 by Western blot and RT-PCR. And the up-regulation of APRIL and HSPG expression was found in SW480. We also observed that knockdown of APRIL levels in SW480, prominently reversed cell proliferation and partially resulted in senescence phenotypes. Furthermore, cellular senescence due to a decreased level of APRIL expression was associated with engagement of HSPG. Thus, our results suggest that low levels of APRIL play an essential role in cellular senescence via a HSPG-dependent signaling pathway in SW480. C1 [Ding, Weifeng] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, No.20, Road XisiNantong, China. [Ju, Shaoqing] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, No.20, Road XisiNantong, China. [Jiang, Shengyang] Nantong University, Department of Public HealthNantong, China. [Zhu, Li] Nantong University, Jiangsu Province Key Laboratory of NeuroregenerationNantong, China. [Wang, Yueguo] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, No.20, Road XisiNantong, China. [Wang, Huimin] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, No.20, Road XisiNantong, China. RP Wang, H (reprint author), Affiliated Hospital of Nantong University, Department of Laboratory Medicine, Nantong, China. EM ntfyjyk@pub.nt.jsinfo.net CR Bazzoni F, Beutler B, 1996, The tumor necrosis factor ligand and receptor families. N Engl J Med 334:1717–1725 Gaur U, Aggarwal BB, 2003, Regulation of proliferation, survival and apoptosis by members of the TNF superfamily. Biochem Pharmacol 66:1403–1408 Hahne M, Kataoka T, Schroter M, Hofmann K, Irmler M, Bodmer JL, Schneider P et al, 1998, APRIL, a new ligand of the tumor necrosis factor family, stimulates tumor cell growth. J Exp Med 188:1185–1190 Mackay F, Schneider P, Rennert P, Browning J, 2003, BAFF AND APRIL: a tutorial on B cell survival. Annu Rev Immunol 21:231–264 Roschke V, Sosnovtseva S, Ward CD, Hong JS, Smith R, Albert V, Stohl Wet al, 2002, BLyS and APRIL form biologically active heterotrimers that are expressed in patients with systemic immune-based rheumatic diseases. J Immunol 169:4314–4321 Nishio M, Endo T, Tsukada N, Ohata J, Kitada S, Reed JC, Zvaifler NJ, Kipps TJ, 2005, Nurselike cells express BAFF and APRIL, which can promote survival of chronic lymphocytic leukemia cells via a paracrine pathway distinct from that of SDF- 1alpha. Blood 106:1012–1020 Mongini PK, Inman JK, Han H, Fattah RJ, Abramson SB, Attur M, 2006, APRIL and BAFF promote increased viability of replicating human B2 cells via mechanism involving cyclooxygenase 2. J Immunol 176:6736–6751 Tangye SG, Bryant VL, Cuss AK, Good KL, 2006, BAFF, APRIL and human B cell disorders. Semin Immunol 18:305–317 Endo T, Nishio M, Enzler T, Cottam HB, Fukuda T, James DF, Karin M, Kipps TJ, 2006, BAFF and APRIL support chronic lymphocytic leukemia B-cell survival through activation of the canonical NF-kappaB pathway. Blood 109:703–710 Craxton A, Draves KE, Gruppi A, Clark EA, 2005, BAFF regulates B cell survival by downregulating the BH3-only family member Bim via the ERK pathway. J Exp Med 202:1363–1374 Dillon SR, Gross JA, Ansell SM, Novak AJ, 2006, An APRIL to remember: novel TNF ligands as therapeutic targets. Nat Rev Drug Discov 5:235–246 Deshayes F, Lapree G, Portier A, Richard Y, Pencalet P, Mahieu- Caputo D et al, 2004, Abnormal production of the TNF-homologue APRIL increases the proliferation of human malignant glioblastoma cell lines via a specific receptor. Oncogene 23:3005–3012 Rennert P, Schneider P, Cachero TG, Thompson J, Trabach L, Hertig S, Holler N et al, 2000, A soluble form of B cell maturation antigen, a receptor for the tumor necrosis factor family member APRIL, inhibits tumor cell growth. J Expt Med 192:1677–1684 Marsters SA, Yan M, Pitti RM, Haas PE, Dixit VM, Ashkenazi A, 2000, Interaction of the TNF homologues BLyS and APRIL with the TNF receptor homologues BCMA and TACI. Curr Biol 10:785–788 Wu Y, Bressette D, Carrell JA, Kaufman T, Feng P, Taylor K et al, 2000, Tumor necrosis factor, TNF, receptor superfamily member TACI is a high affinity receptor for TNF family members APRIL and BLyS. J Biol Chem 275:35478–35485 Thompson JS, Bixler SA, Qian F, Vora K, Scott ML, Cachero TG et al, 2001, BAFF-R, a newly identified TNF receptor that specifically interacts with BAFF. Science 293:2108–2111 Hendriks J, Planelles L, de Jong-Odding J, Hardenberg G, Pals ST, Hahne M, Spaargaren M, Medema JP, 2005, Heparan sulfate proteoglycan binding promotes APRIL-induced tumor cell proliferation. Cell Death Differ 12:637–648 Ingold K, Zumsteg A, Tardivel A, Huard B, Steiner QG, Cachero TG, Qiang F et al, 2005, Identification of proteoglycans as the APRIL-specific binding partners. J Exp Med 201:1375–1383 Lopez-Fraga M, Fernandez R, Albar JP, Hahne M, 2001, Biologically active APRIL is secreted following intracellular processing in the Golgi apparatus by furin convertase. EMBO Rep 2:945–951 Wallweber HJ, Compaan DM, Starovasnik MA, Hymowitz SG, 2004, The crystal structure of a proliferation-inducing ligand, APRIL. J Mol Biol 343:283–290 Varfolomeev E, Kischkel F, Martin F, Seshasayee D, Wang H, Lawrence D, Olsson C et al, 2004, APRIL-deficient mice have normal immune system development. Mol Cell Biol 24:997–1006 Roth W, Wagenknecht B, Klumpp A, Naumann U, Hahne M, Tschopp J, Weller M, 2001, APRIL, a new member of the tumor necrosis factor family, modulates death ligand—induced apoptosis. Cell Death Differ 8:403–410 Ben-Porath I, Weinberg RA, 2005, The signals and pathways activating cellular senescence. Int J Biochem Cell Biol 37:961–976 Chiu A, Xu W, He B, Dillon SR, Gross JA, Sievers E et al, 2007, Hodgkin lymphoma cells express TACI and BCMA receptors and generate survival and proliferation signals in response to BAFF and APRIL. Blood 109:729–739 Schwaller J, Schneider P, Mhawech-Fauceglia P, McKee T, Myit S, Matthes T et al, 2007, Neutrophil-derived APRIL concentrated in tumor lesions by proteoglycans correlates with human B cell lymphoma aggressiveness. Blood 109:331–338 Wagner M, Hampel B, Bernhard D, Hala M, Zwerschke W, Jansen- Durr P, 2001, Replicative senescence of human endothelial cells in vitro involves G1 arrest, polyploidization and senescence-associated apoptosis. Exp Gerontol 36:1327–1347 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 693 EP 701 DI 10.1007/s12253-009-9172-y PG 9 ER PT J AU Xu, P Yu, Sh Jiang, R Kang, Ch Wang, G Jiang, H Pu, P AF Xu, Peng Yu, Shizhu Jiang, Rongcai Kang, Chunsheng Wang, Guangxiu Jiang, Hao Pu, Peiyu TI Differential Expression of Notch Family Members in Astrocytomas and Medulloblastomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Astrocytomas; Differential expression; Medulloblastomas; Notch family members ID Astrocytomas; Differential expression; Medulloblastomas; Notch family members AB Notch signaling pathway plays an integral role in determining cell fates in development. Growing evidence demonstrates that Notch signaling pathway has versatile effects in tumorigenesis depending on the tumor type, grade and stage. Notch signaling pathway is deregulated in some brain tumors. To examine the differential expression of Notch family members (Notch1, 2, 3, 4) in human astrocytomas and medulloblastomas, and to evaluate their roles in the development of both tumor types. Immunohistochemical staining and Western blot analysis were used to detect Notch1, 2, 3, 4 expression in tissue microarray and freshly resected tissue samples of normal brain, astrocytomas and medulloblastomas. Notch family members were not expressed or barely detectable in normal brain tissues. Notch1, 3, 4 were highly expressed but Notch2 was not expressed in astrocytomas. The percentage of immunopositive tumor cells and level of Notch1 expression was increased with tumor grade. In addition, overexpression of Notch2 was detected in medulloblastomas in contrast to low or no expression of Notch1, 3, 4. Differential expression of Notch1, 2, 3, 4 is detected in astrocytomas and medulloblastomas, that may be related to their different roles playing in the development of brain tumors. C1 [Xu, Peng] Tianjin Medical University General Hospital, Department of Neurosurgery, 154 Anshan Road, 300052 Tianjin, China. [Yu, Shizhu] Tianjin Neurological Institute, Laboratory of NeuropathologyTianjin, China. [Jiang, Rongcai] Tianjin Medical University General Hospital, Department of Neurosurgery, 154 Anshan Road, 300052 Tianjin, China. [Kang, Chunsheng] Tianjin Medical University General Hospital, Department of Neurosurgery, 154 Anshan Road, 300052 Tianjin, China. [Wang, Guangxiu] Tianjin Medical University General Hospital, Department of Neurosurgery, 154 Anshan Road, 300052 Tianjin, China. [Jiang, Hao] Henry Ford Hospital, Department of Neurology, 48202 Detroit, MI, USA. [Pu, Peiyu] Tianjin Medical University General Hospital, Department of Neurosurgery, 154 Anshan Road, 300052 Tianjin, China. RP Pu, P (reprint author), Tianjin Medical University General Hospital, Department of Neurosurgery, 300052 Tianjin, China. EM pupeiyu33@hotmail.com CR Mizutani K, Yoon K, Dang L et al, 2007, Differential Notch signalling distinguishes neural stem cells from intermediate progenitors. Nature 449:351–355 Fiuza UM, Arias AM, 2007, Cell and molecular biology of Notch. J Endocrinol 194:459–474 Yochem J, Weston K, Greenwald I, 1988, The Caenorhabditis elegans lin-12 gene encodes a transmembrane protein with overall similarity to Drosophila Notch. Nature 335:547–550 Lubman OY, Korolev SV, Kopan R, 2004, Anchoring notch genetics and biochemistry; structural analysis of the ankyrin domain sheds light on existing data. Mol Cell 13:619–626 Fleming RJ, 1998, Structural conservation of Notch receptors and ligands. Semin Cell Dev Biol 9:599–607 Leong KG, Karsan A, 2006, Recent insights into the role of Notch signaling in tumorigenesis. Blood 107:2223–2233 Iso T, Kedes L, Hamamori Y, 2003, HES and HERP Families: multiple effectors of the Notch signaling pathway. J Cell Physiol 194:237–255 Lasky J, Wu H, 2005, Notch signaling, brain development, and human disease. Pediatr Res 57:104R–109R Miele L, 2006, Notch signaling. Clin Cancer Res 12:1074–1079 Kovall RA, 2008, More complicated than it looks: assembly of Notch pathway transcription complexes. Oncogene 27:5099–5109 Hansson EM, Lendahl U, Chapman G, 2004, Notch signaling in development and disease. Semin Cancer Biol 14:320–328 Artavanis-Tsakonas S, Rand MD, Lake RJ, 1999, Notch signaling: cell fate control and signal integration in development. Science 284:770–776 Weng AP, Nam Y, Wolfe MS et al, 2003, Growth suppression of pre-T acute lymphoblastic leukemia cells by inhibition of notch signaling. Mol Cell Biol. 23:655–664 Weijzen S, Rizzo P, Braid M et al, 2002, Activation of Notch-1 signaling maintains the neoplastic phenotype in human Rastransformed cells. Nat Med 8:979–986 Zagouras P, Stifani S, Blaumueller CM et al, 1995, Alterations in Notch signaling in neoplastic lesions of the human cervix. Proc Natl Acad Sci U S A 92:6414–6418 Nicolas M, Wolfer A, Raj K et al, 2003, Notch1 functions as a tumor suppressor in mouse skin. Nat Genet 33:416–421 Dotto GP, 2008, Notch tumor suppressor function. Oncogene 27:5115–5123 Qiu MZ, Pu PY, Kang CS et al, 2008, The inhibitory effect of siRNA targeting Notch1 on the U251 glioblastoma cell growth in vivo. Chin J Microsurg 31:214–215 Fan X, Mikolaenko I, Elhassan I et al, 2004, Notchl and notch2 have opposite effects on embryonal brain tumor growth. Cancer Res 64:7787–7793 Miyamoto Y, Maitra A, Ghosh B et al, 2003, Notch mediates TGF alpha-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell 3:565–576 Nickoloff BJ, Osborne BA, Miele L, 2003, Notch signaling as a therapeutic target in cancer: a new approach to the development of cell fate modifying agents. Oncogene 22:6598–6608 Callahan R, Egan SE, 2004, Notch signaling in mammary development and oncogenesis. J Mammary Gland Biol Neoplasia 9:145–163 KunnimalaiyaanM, Chen H, 2007, Tumor suppressor role of Notch- 1 signaling in neuroendocrine tumors. Oncologist 12:535–542 Cuevas IC, Slocum AL, Jun P et al, 2005, Meningioma transcript profiles reveal deregulated Notch signaling pathway. Cancer Res 65:5070–5075 Purow BW, Haque RM, Noel MW et al, 2005, Expression of Notch-1 and its ligands, Delta-like-1 and Jagged-1, is critical for glioma cell survival and proliferation. Cancer Res 65:2353–2363 Kanamori M, Kawaguchi T, Nigro JM et al, 2007, Contribution of Notch signaling activation to human glioblastoma multiforme. J Neurosurg 106:417–427 Shih AH, Holland EC, 2006, Notch signaling enhances nestin expression in gliomas. Neoplasia 8:1072–1082 Dang L, Fan X, Chaudhry A et al, 2006, Notch3 signaling initiates choroid plexus tumor formation. Oncogene 25:487–491 Radtke F, Raj K, 2003, The role of Notch in tumorigenesis: oncogene or tumour suppressor? Nat Rev Cancer 3:756–767 Breunig JJ, Silbereis J, Vaccarino FM et al, 2007, Notch regulates cell fate and dendrite morphology of newborn neurons in the postnatal dentate gyrus. Proc Natl Acad Sci U S A 104:20558–20563 Tokunaga A, Kohyama J, Yoshida T et al, 2004, Mapping spatio-temporal activation of Notch signaling during neurogenesis and gliogenesis in the developing mouse brain. J Neurochem 90:142–154 Taylor MK, Yeager K, Morrison SJ, 2007, Physiological Notch signaling promotes gliogenesis in the developing peripheral and central nervous systems. Development 134:2435–2447 Solecki DJ, Liu XL, Tomoda T et al, 2001, Activated Notch2 signaling inhibits differentiation of cerebellar granule neuron precursors by maintaining proliferation. Neuron 31:557–568 Irvin DK, Zurcher SD, Nguyen T et al, 2001, Expression patterns of Notch1, Notch2, and Notch3 suggest multiple functional roles for the Notch-DSL signaling system during brain development. J Comp Neurol 436:167–181 Bellavia D, Checquolo S, Campese AF et al, 2008, Notch3: from subtle structural differences to functional diversity. Oncogene 27:5092–5098 Katoh M, 2007, Networking of WNT, FGF, Notch, BMP, and Hedgehog signaling pathways during carcinogenesis. Stem Cell Rev 3:30–38 McElhinny AS, Li JL, Wu L, 2008, Mastermind-like transcriptional co-activators: emerging roles in regulating cross talk among multiple signaling pathways. Oncogene 27:5138–5147 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 703 EP 710 DI 10.1007/s12253-009-9173-x PG 8 ER PT J AU Gole, B Duran Alonso, BM Dolenc, V Lah, T AF Gole, Boris Duran Alonso, Beatriz Maria Dolenc, Vincenc Lah, Tamara TI Post-Translational Regulation of Cathepsin B, but not of Other Cysteine Cathepsins, Contributes to Increased Glioblastoma Cell Invasiveness In Vitro SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Brain tumours; Cysteine cathepsins; Cystatins; Glioma; Invasion; Proteolysis; Stefins ID Brain tumours; Cysteine cathepsins; Cystatins; Glioma; Invasion; Proteolysis; Stefins AB Cells that migrate away from a central tumour into brain tissue are responsible for inefficient glioblastoma treatment. This migratory behaviour depends partially on lysosomal cysteine cathepsins. Reportedly, the expression of cathepsins B, L and S gradually increases in the progression from benign astrocytoma to the malignant glioblastoma, although their specific roles in glioma progression have not been revealed. The aim of this study was to clarify their specific contribution to glioblastoma cell invasion. The differences between the matrix invading cells and non-invading core cells from spheroids derived from glioblastoma cell culture and from glioblastoma patients’ biopsies, and embedded in type I collagen, have been studied at the mRNA, protein and cathepsin activity levels. Analyses of the two types of cells showed that the three cathepsins were up-regulated post-translationally, their specific activities increasing in the invading cells. The cystatin levels were also differentially altered, resulting in higher ratio of cathepsins B and L to stefin B in the invading cells. However, using specific synthetic inhibitors and silencing strategies revealed that only cathepsin B activity was involved in the invasion of glioblastoma cells, confirming previous notion of cathepsin B as tumour invasiveness biomarker. Our data support the concept of specific roles of cysteine cathepsins in cancer progression. Finally the study points out on the complexity of protease regulation and the need to include functional proteomics in the systems biology approaches to understand the processes associated with glioma invasion and progression. C1 [Gole, Boris] National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Vecna pot 111, SI-1000 Ljubljana, Slovenia. [Duran Alonso, Beatriz Maria] National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Vecna pot 111, SI-1000 Ljubljana, Slovenia. [Dolenc, Vincenc] University Clinical Centre, Department of NeurosurgeryLjubljana, Slovenia. [Lah, Tamara] National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Vecna pot 111, SI-1000 Ljubljana, Slovenia. RP Gole, B (reprint author), National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, SI-1000 Ljubljana, Slovenia. EM boris.gole@nib.si CR Ohgaki H, Kleihues P, 2005, Epidemiology and etiology of gliomas. Acta Neuropathol 109(1):93–108 Sathornsumetee S, Rich NJ, 2006, New treatment strategies for malignant gliomas. Expert Rev Anticancer Ther 6(7):1087–1104 Rao JS, 2003, Molecular mechanisms of glioma invasiveness: the role of proteases. Nat Rev Cancer 3(7):489–501 Demuth T, Berens M, 2004, Molecular mechanisms of glioma cell migration and invasion. J Neurooncol 70(2):217–228 Wang W, Goswami S, Sahai E et al, 2005, Tumour cells caught in the act of invading: their strategy for enhanced cell motility. Trends Cell Biology 15(3):138–145 Sahai E, 2005, Mechanisms of cancer cell invasion. Curr Opin Genet Dev 15(1):87–96 Mariani L, Beaudry C, McDonough WS et al, 2001, Glioma cell motility is associated with reduced transcription of proapoptotic and proliferation genes: a cDNA microarray analysis. J Neurooncol 53(2):161–176 Demuth T, Rennert JL, Hoelzinger DB et al, 2008, Glioma cells on the run- the migratory transcriptome of 10 human glioma cell lines. BMC Genomics 9:54 Berens ME, Rief MD, Loo MA, Giese A, 1994, The role of extracellular matrix in human astrocytoma migration and proliferation studied in a microliter scale assay. Clin Exp Metastasis 12, 6):405–415 Hoelzinger DB, Nakada M, Demuth T et al, 2008, Autotaxin: a secereted autocrine/paracrine factor that promotes glioma invasion. J Neurooncol 86(3):297–309 Wolf K, Friedl P, 2005, Functional imaging of pericellular proteolysis in cancer cell invasion. Biochemie 87(3–4):315–320 Tu C, Ortega–Cava CF, Chen G et al, 2008, Lysosomal cathepsins B participates in the podosome-mediated extracellular matrix degradation and invasion via secreted lysosomes in the v-Src fibroblasts. Cancer Res 86(22):9147–9156 Lopez–Otin C, Matrisian LM, 2007, Emerging roles of proteases in tumour suppression. Nature Rev Cancer 7(10):800–808 Levicar N, Nuttall RL, Lah TT, 2003, Proteases in brain tumour progression. Acta Neurochir, Wien, 145(9):825–838 Lah TT, Duran Alonso MB, Van Noorden CJ, 2006, Antiprotease therapy in cancer: hot or not? Expert Opin Biol Ther 6(3):257– 279 Rempel SA, Rosenblum ML, Mikkelsen T et al, 1994, CathepsinB expression and localization in glioma progression and invasion. Cancer Res 54(23):6027–6031 Lah TT, Strojnik T, Levicar N et al, 2000, Clinical and experimental studies of cysteine cathepsins and their inhibitors in human brain tumors. Int J Biol Markers 15(1):90–93 Strojnik T, Kos J, Zidanik B, Lah TT, 1999, Cathepsin B immunohistochemical staining in tumour and endothelial cells is a new prognostic factor for survival in patients with brain tumours. Clin Cancer Res 5(3):559–567 Strojnik T, Kavalar R, Trinkaus M, Lah TT, 2005, Cathepsin L in glioma progression: comparison with cathepsin B. Cancer Detect Prev 29(5):448–455 Sivarapathi M, Yamamoto M, Nicolson GL et al, 1996, Expression and immunohistochemical localization of cathepsin L during progression of human gliomas. Clin Exp Metastasis 14, 1):27–34 Flannery T, Gibson D, Mirakhur M et al, 2003, The clinical significance of cathepsin S expression in human astrocytomas. Am J Pathol 163(1):175–182 Flannery T, McQuaid S, McGoohan C et al, 2006, Cathepsin S expression: An independent prognostic factor in glioblastoma tumours-A pilot study. Int J Cancer 119(4):854–860 Kos J, Lah TT, 2006, Cystatins in cancer. In: Zerovnik E, Kopitar-Jerala N, eds, Human Stefins and Cystatins. Nova Science Publishers Inc, New York Abrahamson M, 1994, Cystatins. Methods Enzymol 244:685–700 Lignelid H, Collins VP, Jacobsson B, 1997, Cystatin C and transthyretin expression in normal and neoplastic tissues of the human brain and pituitary. Acta Neuropathol 93(5):494–500 Nakabayashi H, Hara M, Shimuzu K, 2005, Clinicopathologic significance of cystatin C expression in gliomas. Hum Pathol 36, 9):1008–1015 Konduri SD, Yanamandra N, Siddique K et al, 2002, Modulation of cystatin C expression impairs the invasive and tumorigenic potential of human glioblastoma cells. Oncogene 21(57):8705–8712 Bervar A, Zajc I, Sever B et al, 2003, Invasiveness of transformed human breast epithelial cell lines is related to cathepsin B and inhibited by cysteine proteinase inhibitors. Biol Chem 384(3):447–455 Zajc I, Hreljac I, Lah T, 2006, Cathepsin L affects apoptosis of glioblastoma cells: a potential implication in the design of cancer therapeutics. Anticancer Res 26(5A):3357-64 Hegedus B, Marga F, Jakab K et al, 2006, The Interplay of Cell- Cell and Cell-Matrix Interactions in the Invasive Properties of Brain Tumors. Biophys J 91(7):2708–2716 Corcoran A, De Ridder LI, Del Duca D et al, 2003, Evolution of the brain tumour spheroid model: transcending current model limitations. Acta Neurochir, Wien, 145(9):819–824 Rubenstein BM, Kaufman LJ, 2008, The role of extracellular matrix in glioma invasion:a cellular potts model approach. Biophys J 95(12):5661–5680 Gondi CS, Kandhukuri N, Kondraganti S et al, 2006, RNA interference–mediated simultaneous down-regulation of urokinase-type plasminogen activator receptor and cathepsin B induces caspase-mediated apoptosis in SNB 19 human glioma cells. Mol Cancer Ther 5(12):3197–3208 Gocheva V, Zeng W, Ke D et al, 2006, Distinct role for cysteine cathepsin genes in multistage tumourigenesis. Genes Dev 20, 5):543–556 Reinheckel T, Gocheva V, Peters C, Joyce JA, 2008, Roles of cysteine proteases in tumour progression: Analysis of cysteine cathepsins knockout mice in cancer models. In: Edwards D, Hoyer- Hansen G, Blasi F, Sloane BF, eds, The Cancer Degradome. Springer Science+Business Media, New York Lakka C, Gondi CS, Yanamandra N et al, 2004, Inhibition of cathepsin B and MMP-9 gene expression in glioblastoma cell line via RNA interference reduces tumour cell invasion, tumour growth and angiogenesis. Oncogene 23(27):4681– 4689 Premzl A, Zavasnik-Bergant V, Turk V, Kos J, 2003, Intracellular and extracellular cathepsin B facilitate invasion of MCF-10A neoT cells through reconstituted extracellular matrix in vitro. Exp Cell Res 283(2):206–214 Klose A, Wilbrand-Hennes A, Zigrino P et al, 2006, Contact of high-invasive, but not low-invasive, melanoma cells to native collagen I induces the release of mature cathepsin B. Int J Cancer 118(11):2735–2743 Podgorski I, Linebough BE, Sameni M et al, 2005, Bone microenvironment modulates expression and activity of cathepsin B in prostate cancer. Neoplasia 7(3):207–223 Sameni M, Dosescu J, Sloane BF, 2001, Imaging proteolysis by living human glioma cells. Biol Chem 382(5):785–788 Zhu DM, Uckun FM, 2000, Z-Phe-Gly-NHO-Bz, an inhibitor of cysteine cathepsins, induces apoptosis in human cancer cells. Clin Cancer Res 6(5):2064–2069 Felbor U, Kesseler B, Mothes W et al, 2002, Neuronal loss and brain atrophy in mice lacking cathepsins B and L. Proc Natl Acad Sci USA 99(12):7883–7888 Levicar N, Dewey RA, Daley E et al, 2003, Selective supression of cathepsin L by antisense cDNA impairs human brain tumor cell invasion in vitro and promotes apoptosis. Cancer Gene Ther 10, 2):141–151 Castino R, Pace D, Demoz M et al, 2002, Lysosomal proteases as potential targets for the induction of apoptotic cell death in human neuroblastomas. Int J Cancer 97(6):775–779 Bellail AC, Hunter SB, Brat DJ et al, 2004, Microregional extracellular matrix heterogeneity in brain modulates glioma cell invasion. Int J Biochem Cell Biol 36(6):1046–1069 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 711 EP 723 DI 10.1007/s12253-009-9175-8 PG 13 ER PT J AU Bianchi, S Caini, S Cattani, GM Vezzosi, V Biancalani, M Palli, D AF Bianchi, Simonetta Caini, Saverio Cattani, Grazia Maria Vezzosi, Vania Biancalani, Mauro Palli, Domenico TI Diagnostic Concordance in Reporting Breast Needle Core Biopsies using the B Classification—A Panel in Italy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast; Non palpable lesions; Needle core biopsy; Reporting; Diagnostic concordance ID Breast; Non palpable lesions; Needle core biopsy; Reporting; Diagnostic concordance AB The widespread implementation of mammography screening has resulted in an increased frequency of needle core biopsies (NCB). The aim of this study was that of evaluating the diagnostic reproducibility on breast NCB, according to the B-classification, among several pathologists from different Italian regions. Fifty single slides of NCBs performed for non palpable breast lesions were selected to evaluate the diagnostic reproducibility, according to the B classification, among 31 pathologists from different Italian areas, involved in the pathologic diagnosis of screen-detected breast lesions. According to the study majority diagnosis (MD), 21 cases were classified as B2 (benign lesion), 23 B3 (lesion of uncertain malignant potential) and 6 B5 (malignant lesion). Overall, individual kappa coefficients in comparison to MD were good (mean 0.61, range 0.31–0.88). The level of inter-observer agreement, however, appeared lower in differentiating the two intermediate categories B2 and B3, thus potentially leading to over-treatment (false-positives: 26%) or under-treatment (false-negatives: 17%) of individual patients. Specific sub-types of B3 need an improvement of the diagnostic definition. A multidisciplinary approach and consultation with expert colleagues are recommended. C1 [Bianchi, Simonetta] AOU Careggi, Department of Human Pathology and Oncology, AOU Careggi, Viale Morgagni, 50134 Florence, Italy. [Caini, Saverio] Cancer Research and Prevention Institute (ISPO), Molecular and Nutritional Epidemiology UnitFlorence, Italy. [Cattani, Grazia Maria] Bellaria Hospital, Pathologic Anatomy UnitBologna, Italy. [Vezzosi, Vania] AOU Careggi, Department of Human Pathology and Oncology, AOU Careggi, Viale Morgagni, 50134 Florence, Italy. [Biancalani, Mauro] Empoli Hospital, Pathologic Anatomy UnitFlorence, Italy. [Palli, Domenico] Cancer Research and Prevention Institute (ISPO), Molecular and Nutritional Epidemiology UnitFlorence, Italy. RP Bianchi, S (reprint author), AOU Careggi, Department of Human Pathology and Oncology, 50134 Florence, Italy. EM simonetta.bianchi@unifi.it CR El-Sayed ME, Rakka EA, Reed J et al, 2008, Audit of performance of needle core biopsy diagnoses of screen detected breast lesions. Eur J Cancer 44:2580–2586 NHSBSP Breast Screening Programme, 2001, Guidelines for Non-Operative Diagnostic Procedures and Reporting in Breast Cancer Screening. Sheffield: NHSBSP Pub. No.50 European Commission Working Group on Breast Screening Pathology, 2006, Quality assurance guidelines for pathology. In: Perry N, Broeders M, de Wolf C, Tornberg S, Holland R, von Karsa L, eds, European guidelines for quality assurance in breast cancer screening and diagnosis. Volume 6, 4th edn. Office for Official Publications of the European Communities, Luxemburg, pp 219–312 Pinder SE, Reis-Filho JS, 2007, Non-operative breast pathology. J Clin Pathol 60:1297–1299 Verkooijen HM, 2008, Needle core biopsy for screen detected breast lesions: time to raise the bar. Eur J. Cancer 44:2540–2541 Kumaroswamy V, Liston J, Shaaban AM, 2008, Vacuum assisted stereotactic guided mammotome biopsies in the management of screen detected microcalcifications: experience of a large breast screening centre. J Clin Pathol 61:766–769 Lazarus E, Mainiero MB, Schepps B et al, 2006, BI-RADS lexicon for US and mammography: interobserver variability and positive predictive value. Radiology 239:385–391 Schnitt SJ, Vincent-Salomon A, 2003, Columnar cell lesions of the breast. Adv Anat Pathol 10:113–124 Cohen J, 1960, A coefficient of agreement for nominal scales. Educational and Psychological Measurement 20:37–46 Landis JR, Koch GG, 1977, The measurement of observer agreement for categorical data. Biometrics 33:59–174 Bonk U, Gohla G, Sauer U et al, 2005, Grosse Ubereinstimmung Eergebnisse des ersten Mammographie-Screening-Projekts in Bremen aus histopathologischer Sicht. Deutsche Arzteblatt online, http://www.aerzteblatt.de/aufsaetze/0501) Kluttig A, Trocchi P, Heinig A et al, 2007, Reliability and validity of needle biopsy evaluation of breast-abnormalities using the B-categorization—design and objectives of the Diagnosis Optimisation Study, DIOS). BMC Cancer 7:100– 108 Collins LC, Connolly JL, Page DL et al, 2007, Diagnostic agreement in the evaluation of image-guided breast core needle biopsies. Am J Surg Pathol 28:126–131 Houssami N, Ciatto S, Bilous M et al, 2007, Borderline breast core needle histology: predictive values for malignancy in lesions of uncertain malignant potential, B3). Br J Cancer 96:1253–1257 Verkooijen HM, Peterse JL, Schipper MEI et al, 2003, Interobserver variability between general and expert pathologists during the histopathological assessment of large-core needle and open biopsies of non-palpable breast lesions. Eur J Cancer 39:2187– 2191 Lee AD, Denley HE, Pinder SE et al, 2003, Excision biopsy findings of patients with breast needle core biopsies reported as suspicious of malignancy, B4, or lesion of uncertain malignant potential, B3). Histopathology 42:331–336 Dillon MF, McDermott EW, Hill AD et al, 2007, Predictive value of breast lesions of “Uncertain Malignant Potential” and “Suspicious for malignancy” determined by needle core biopsy. Ann Surg Oncol 14:704–711 Tan PH, Ho BCS, Selvarajan S et al, 2005, Pathological diagnosis of columnar cell lesions of the breast: are there issues of reproducibility? J Clin Pathol 58:705–709 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 725 EP 732 DI 10.1007/s12253-009-9176-7 PG 8 ER PT J AU Coufal, O Pavlik, T Fabian, P Bori, R Boross, G Sejben, I Maraz, R Koca, J Krejci, E Horakova, I Foltinova, V Vrtelova, P Chrenko, V Tekle, EW Rajtar, M Svebis, M Fait, V Cserni, G AF Coufal, Oldrich Pavlik, Tomas Fabian, Pavel Bori, Rita Boross, Gabor Sejben, Istvan Maraz, Robert Koca, Jaroslav Krejci, Eva Horakova, Iva Foltinova, Vendula Vrtelova, Pavlina Chrenko, Vojtech Tekle, Eliza Wolde Rajtar, Maria Svebis, Mihaly Fait, Vuk Cserni, Gabor TI Predicting Non-Sentinel Lymph Node Status After Positive Sentinel Biopsy in Breast Cancer: What Model Performs the Best in a Czech Population? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Lymphatic metastasis; Nomogram; Prediction; Sentinel lymph node biopsy; Tumor cells; Isolated ID Breast cancer; Lymphatic metastasis; Nomogram; Prediction; Sentinel lymph node biopsy; Tumor cells; Isolated AB Several models have previously been proposed to predict the probability of non-sentinel lymph node (NSLN) metastases after a positive sentinel lymph node (SLN) biopsy in breast cancer. The aim of this study was to assess the accuracy of two previously published nomograms (MSKCC, Stanford) and to develop an alternative model with the best predictive accuracy in a Czech population. In the basic population of 330 SLN-positive patients from the Czech Republic, the accuracy of the MSKCC and the Stanford nomogramswas tested by the area under the receiver operating characteristics curve (AUC). A new model (MOU nomogram) was proposed according to the results of multivariate analysis of relevant clinicopathologic variables. The new model was validated in an independent test population from Hungary (383 patients). In the basic population, six of 27 patients with isolated tumor cells (ITC) in the SLN harbored additional NSLN metastases. The AUCs of the MSKCC and Stanford nomograms were 0.68 and 0.66, respectively; for the MOU nomogram it reached 0.76. In the test population, the AUC of the MOU nomogram was similar to that of the basic population (0.74). The presence of only ITC in SLN does not preclude further nodal involvement. Additional variables are beneficial when considering the probability of NSLN metastases. In the basic population, the previously published nomograms (MSKCC and Stanford) showed only limited accuracy. The developed MOU nomogram proved more suitable for the basic population, such as for another independent population from a mid-European country. C1 [Coufal, Oldrich] Masaryk Memorial Cancer Institute, Department of Surgical Oncology, Zluty kopec 7, 65653 Brno, Czech Republic. [Pavlik, Tomas] Masaryk University, Institute of Biostatistics and Analyses, Kamenice 126/3, 62500 Brno, Czech Republic. [Fabian, Pavel] Masaryk Memorial Cancer Institute, Department of Pathology, Zluty kopec 7, 65653 Brno, Czech Republic. [Bori, Rita] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38Kecskemet, Hungary. [Boross, Gabor] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38Kecskemet, Hungary. [Sejben, Istvan] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38Kecskemet, Hungary. [Maraz, Robert] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38Kecskemet, Hungary. [Koca, Jaroslav] ADDS&DSC International s.r.o, Jana Uhra 10, 60200 Brno, Czech Republic. [Krejci, Eva] Masaryk Memorial Cancer Institute, Department of Pathology, Zluty kopec 7, 65653 Brno, Czech Republic. [Horakova, Iva] Masaryk Memorial Cancer Institute, Department of Pathology, Zluty kopec 7, 65653 Brno, Czech Republic. [Foltinova, Vendula] Masaryk Memorial Cancer Institute, Department of Surgical Oncology, Zluty kopec 7, 65653 Brno, Czech Republic. [Vrtelova, Pavlina] Masaryk Memorial Cancer Institute, Department of Surgical Oncology, Zluty kopec 7, 65653 Brno, Czech Republic. [Chrenko, Vojtech] Masaryk Memorial Cancer Institute, Department of Surgical Oncology, Zluty kopec 7, 65653 Brno, Czech Republic. [Tekle, Eliza Wolde] Bacs-Kiskun County Hospital, Department of Oncoradiology, Nyiri ut 38Kecskemet, Hungary. [Rajtar, Maria] Bacs-Kiskun County Hospital, Department of Oncoradiology, Nyiri ut 38Kecskemet, Hungary. [Svebis, Mihaly] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38Kecskemet, Hungary. [Fait, Vuk] Masaryk Memorial Cancer Institute, Department of Surgical Oncology, Zluty kopec 7, 65653 Brno, Czech Republic. [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38Kecskemet, Hungary. RP Coufal, O (reprint author), Masaryk Memorial Cancer Institute, Department of Surgical Oncology, 65653 Brno, Czech Republic. EM oldrich.coufal@gmail.com CR Park J, Fey JV, Naik AM, Borgen PI, Van Zee KJ, Cody HS 3rd, 2007, A declining rate of completion axillary dissection in sentinel lymph node-positive breast cancer patients is associated with the use of a multivariate nomogram. Ann Surg 245:462–468 Hwang RF, Gonzalez-Angulo AM, Yi M et al, 2007, Low locoregional failure rates in selected breast cancer patients with tumor-positive sentinel lymph nodes who do not undergo completion axillary dissection. Cancer 10:723–730 Van Zee KJ, Manasseh DM, Bevilacqua JL et al, 2003, A nomogram for predicting the likelihood of additional nodal metastases in breast cancer patient with a positive sentinel node biopsy. Ann Surg Oncol 10:1140–1151 Degnim AC, Reynolds C, Pantvaidya G et al, 2005, Nonsentinel node metastasis in breast cancer patients: assessment of an existing and a new predictive nomogram. Am J Surg 190:543–550 Cripe MH, Beran LC, Liang WC, Sickle-Santanello BJ, 2006, The likelihood of additional nodal disease following a positive sentinel lymph node biopsy in breast cancer patients: validation of a nomogram. Am J Surg 192:484–487 Smidt ML, Kuster DM, van der Wilt GJ, Thunnissen FB, Van Zee KJ, Strobbe LJ, 2005, Can the memorial Sloan-Kettering cancer center nomogram predict the likelihood of nonsentinel lymph node metastases in breast cancer patients in the Netherlands? Ann Surg Oncol 12:1066–1072 Soni NK, Carmalt HL, Gillett DJ, Spillane AJ, 2005, Evaluation of a breast cancer nomogram for prediction of non-sentinel lymph node positivity. Eur J Surg Oncol 31:958–964 Lambert LA, Ayers GD, Hwang RF et al, 2006, Validation of a breast cancer nomogram for predicting nonsentinel lymph node metastases after a positive sentinel node biopsy. Ann Surg Oncol 13:310–320 Ponzone R, Maggiorotto F, Mariani L et al, 2007, Comparison of two models for the prediction of nonsentinel node metastases in breast cancer. Am J Surg 193:686–692 Kocsis L, Svebis M, Boross G et al, 2004, Use and limitations of a nomogram predicting the likelihood of non-sentinel node involvement after a positive sentinel node biopsy in breast cancer patients. Am Surg 70:1019–1024 Zgajnar J, Perhavec A, Hocevar M et al, 2007, Low performance of the MSKCC nomogram in preoperatively ultrasonically negative axillary lymph node in breast cancer patients. J Surg Oncol 96:547–553 Alran S, De Rycke Y, Fourchotte V et al, 2007, Validation and limitations of use of a breast cancer nomogram predicting the likelihood of non-sentinel node involvement after positive sentinel node biopsy. Ann Surg Oncol 14:2195–2201 Pal A, Provenzano E, Duffy SW, Pinder SE, Purushotham AD, 2008, A model for predicting non-sentinel lymph node metastatic disease when the sentinel lymph node is positive. Br J Surg 95:302–309 Klar M, Jochmann A, Foeldi M et al, 2008, The MSKCC nomogram for prediction the likelihood of non-sentinel node involvement in a German breast cancer population. Breast Cancer Res Treat 112:523–531 Poirier E, Sideris L, Dube P, Drolet P, Meterissian SH, 2008, Analysis of clinical applicability of the breast cancer nomogram for positive sentinel lymph node: the canadian experience. Ann Surg Oncol 15:2562–2567 Kohrt HE, Olshen RA, Bermas HR et al, 2008, New models and online calculator for predicting non-sentinel lymph node status in sentinel lymph node positive breast cancer patients. BMC Cancer 8:66 Lyman GH, Giuliano AE, Somerfield MR et al, 2005, American society of clinical oncology guideline recommendations for sentinel lymph node biopsy in early-stage breast cancer. J Clin Oncol 23:7703–7720 Sobin LH, Wittekind Ch, eds,, 2002, TNM Classification of Malignant Tumors, 6th Edn. Wiley & Sons, New Jersey Houvenaeghel G, Nos C, Mignotte H et al, 2006, Micrometastases in sentinel lymph node in a multicentric study: predictive factors of nonsentinel lymph node involvement–Groupe des Chirurgiens de la Federation des Centres de Lutte Contre le Cancer. J Clin Oncol 24:1814–1822 Cserni G, Rajtar M, Boross G et al, 2002, Comparison of vital dye-guided lymphatic mapping and dye plus gamma probe-guided sentinel node biopsy in breast cancer. World J Surg 26:592–597 Cserni G, 2002, Complete sectioning of axillary sentinel nodes in patients with breast cancer. Analysis of two different step sectioning and immunohistochemistry protocols in 246 patients. J Clin Pathol 55:926–931 Memorial Sloan-Kettering Cancer Center. Breast cancer nomograms: additional nodal metastases. http://www.mskcc.org/mskcc/ html/15938.cfm. Citied 24 Feb 2009 Kohrt HE, Olshen RA, Jeffrey SS. Non-sentinel lymph node metastasis calculator. Available: https://www3-hrpdcc.stanford. edu/nsln-calculator/. Citied 24 Feb 2009 Hanley JA, McNeil BJ, 1982, The meaning and use of the area under the Receiver Operating Characteristic, ROC, curve. Radiology 143:29–36 Cserni G, Bianchi S, Vezzosi V et al, 2008, Sentinel lymph node biopsy in staging small, up to 15 mm, breast carcinomas. Results from a European multi-institutional study. Pathol Oncol Res 14:117–121 Turner RR, Weaver DL, Cserni G et al, 2008, Nodal stage classification for breast carcinoma: improving interobserver reproducibility through standardized histologic criteria and image-based training. J Clin Oncol 26:258–263 de Mascarel I, MacGrogan G, Debled M, Brouste V, Mauriac L, 2008, Distinction between isolated tumor cells and micrometastases in breast cancer: is it reliable and useful? Cancer 112:1672–1678 Cserni G, Bianchi S, Vezzosi V et al, 2008, Variations in sentinel node isolated tumour cells/micrometastasis and non-sentinel node involvement rates according to different interpretations of the TNM definitions. Eur J Cancer 44:2185–2191 Cserni G, 2007, Comparison of different validation studies on the use of the Memorial-Sloan Kettering cancer center nomogram predicting nonsentinel node involvement in sentinel node-positive breast cancer patients. Am J Surg 194:699–700 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2009 VL 15 IS 4 BP 733 EP 740 DI 10.1007/s12253-009-9177-6 PG 8 ER PT J AU Aktas, S Celebiler, CA Zadeoglulari, Z Diniz, G Kargi, A Olgun, N AF Aktas, Safiye Celebiler, Cavusoglu Aydan Zadeoglulari, Zeynep Diniz, Gulden Kargi, Aydanur Olgun, Nur TI Expression and Methylation Pattern of p16 in Neuroblastoma Tumorigenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Neuroblastoma; p16; Methylation ID Neuroblastoma; p16; Methylation AB Understanding migration, population and differentiation of primordial neural crest cells will help in evolving biology of neuroblastoma. P16 is a tumour suppressor gene contributing in cell cycle arrest as cyclin dependent kinase inhibitor. Methylation is an important mechanism for silencing tumor suppressor genes. The aim of this study was to evaluate the role of p16 and its methylation pattern in neuroblastoma tumorigenesis. This study included 23 cases (11 male; 12 female) and 31 samples from archival paraffin embedded tissues. P16 was studied in 5 samples of normal adrenal medullar tissue, 5 samples of adrenal tissue including blastic rests, 5 samples of neuroblastoma in situ tissue and in 8 samples of neuroblastoma tissues primary and after chemotherapy in each group. The adrenal gland tissues were obtained from paediatric autopsy cases. Expression of p16 was searched by immunohistochemistry. Methylation specific PCR was used to detect the methylation rate of p16. The age range of autopsy cases was between 20 weeks of foetal age and 36 months of infant age. The mean age of neuroblastoma cases was 45 months. P16 expression was positive in normal adrenal tissues, in one of 5 samples of adrenal blastic rest tissue and in all of samples of after chemotherapy; while no expression was observed in neuroblastoma and neuroblastoma in situ tissues. P16 methylation was observed in samples of neuroblastoma in situ and primary neuroblastoma tissues. Our results suggest that p16 and its methylation seems to play role in neuroblastoma tumorigenesis and in the migration, population and differentiation of primordial neural crest cells. Inhibitors of DNA methylation may provide a useful tool for restoring p16 activity in neuroblastoma treatment. C1 [Aktas, Safiye] Dokuz Eylul University, Institute of Oncology, 125/7 sokak Brickent A-3-3Bornova, Izmir, Turkey. [Celebiler, Cavusoglu Aydan] Dokuz Eylul University, Institute of Oncology, 125/7 sokak Brickent A-3-3Bornova, Izmir, Turkey. [Zadeoglulari, Zeynep] Dokuz Eylul University, Institute of Oncology, 125/7 sokak Brickent A-3-3Bornova, Izmir, Turkey. [Diniz, Gulden] Dokuz Eylul University, Institute of Oncology, 125/7 sokak Brickent A-3-3Bornova, Izmir, Turkey. [Kargi, Aydanur] Dokuz Eylul University, Institute of Oncology, 125/7 sokak Brickent A-3-3Bornova, Izmir, Turkey. [Olgun, Nur] Dokuz Eylul University, Institute of Oncology, 125/7 sokak Brickent A-3-3Bornova, Izmir, Turkey. RP Aktas, S (reprint author), Dokuz Eylul University, Institute of Oncology, Bornova, Turkey. EM safiyeaktas@yahoo.com CR Michalowski MB, de Fraipont F, Plantaz D, Michelland S, Combaret V, Favrot MC, 2008, Methylation of tumor-suppressor genes in neuroblastoma: the RASSF1A gene is almost always methylated in primary tumors. Pediatr Blood Cancer 50(1):29–32 Bassi CL, Martelli L, Cipolotti R, Scrideli CA, Defavery R, Tone LG, 2004, Lack of evidence for mutations or deletions in the CDKN2A/p16 and CDKN2B/p15 genes of Brazilian neuroblastoma patients. Braz J Med Biol Res 37(11):1683–1687 Takita J, Hayashi Y, Nakajima T, Adachi J, Tanaka T, Yamaguchi N, Ogawa Y, Hanada R, Yamamoto K, Yokota J, 1998, The p16, CDKN2A, gene is involved in the growth of neuroblastoma cells and its expression is associated with prognosis of neuroblastoma patients. Oncogene 17(24):3137–3143 Omura-Minamisawa M, Diccianni MB, Chang RC, Batova A, Bridgeman LJ, Schiff J, Cohn SL, London WB, Yu AL, 2001, p16/p14(ARF, cell cycle regulatory pathways in primary neuroblastoma: p16 expression is associated with advanced stage disease. Clin Cancer Res 7(11):3481–3490 Beausejour CM, Krtolica A, Galimi F, Narita M, Lowe SW, Yaswen P, Campisi J, 2003, Reversal of human cellular senescence: roles of the p53 and p16 pathways. EMBO J 22, 16):4212–4222 Ohtani N, Yamakoshi K, Takahashi A, Hara E, 2004, The p16INK4a-RB pathway: molecular link between cellular senescence and tumor suppression. J Med Invest 51(3–4):146–153 Ebinger M, Senf L, Wachowski O, Scheurlen W, 2004, Promoter methylation pattern of caspase-8, P16INK4A, MGMT, TIMP-3, and E-cadherin in medulloblastoma. Pathol Oncol Res 10(1): 17–21 Ivanova TA, Golovina DA, Zavalishina LE, Volgareva GM, Katargin AN, Andreeva YY, Frank GA, Kisseljov FL, 2007, Kisseljova NP.Up-regulation of expression and lack of 5′ CpG island hypermethylation of p16 INK4a in HPV-positive cervical carcinomas. BMC Cancer 7:47 Attri J, Srinivasan R, Majumdar S, Radotra BD, Wig J, 2005, Alterations of tumor suppressor gene p16INK4a in pancreatic ductal carcinoma. BMC Gastroenterol 5:22 Liu Y, Lan Q, Siegfried JM, Luketich JD, Keohavong P, 2006, Aberrant promoter methylation of p16 and MGMT genes in lung tumors from smoking and never-smoking lung cancer patients. Neoplasia 8(1):46–51 Conrad RM, Askin FB, Dehner LP, 2001, Adrenal glands In: Dehner LP, Stocker JT, eds, Pediatric pathology, 2nd edn, vol 2. Lippincott Williams and Wilkins, pp 1040–1093 Aktas S, Kargi A, Olgun N, Diniz G, Erbay A, Vergin C, 2007, Prognostic significance of cell proliferation and apoptosisregulating proteins in Epstein-Barr virus positive and negative pediatric Hodgkin lymphoma. Lymphat Res Biol 5(3):175–182 Holst CR, Nuovo GJ, Esteller M, Chew K, Baylin SB, Herman JG, Tlsty TD, 2003, Methylation of p16(INK4a, promoters occurs in vivo in histologically normal human mammary epithelia. Cancer Res 63(7):1596–1601 De Preter K, Vandesompele J, Heimann P, Yigit N, Beckman S, Schramm A, Eggert A, Stallings RL, Benoit Y, Renard M, De Paepe A, Laureys G, Pahlman S, Speleman F, 2006, Human fetal neuroblast and neuroblastoma transcriptome analysis confirms neuroblast origin and highlights neuroblastoma candidate genes. Genome Biol 7(9):R84 Strieder V, Lutz W, 2003, E2F proteins regulate MYCN expression in neuroblastomas. J Biol Chem 278(5):2983–2989 Gebauer S, Yu AL, Omura-Minamisawa M, Batova A, Diccianni MB, 2004, Expression profiles and clinical relationships of ID2, CDKN1B, and CDKN2A in primary neuroblastoma. Genes Chromosomes Cancer 41(4):297–308 Bassi CL, Martelli L, Cipolotti R, Scrideli CA, Defavery R, Tone LG, 2004, Lack of evidence for mutations or deletions in the CDKN2A/p16 and CDKN2B/p15 genes of Brazilian neuroblastoma patients. Braz J Med Biol Res 37(11):1683–1687 Schumacher A, Kapranov P, Kaminsky Z, Flanagan J, Assadzadeh A, Yau P, Virtanen C, Winegarden N, Cheng J, Gingeras T, Petronis A, 2006, Microarray-based DNA methylation profiling: technology and applications. Nucleic Acids Res 34(2):528–542 Clement G, Benhattar J, 2005, A methylation sensitive dot blot assay, MS-DBA, for the quantitative analysis of DNA methylation in clinical samples. J Clin Pathol 58(2):155–158 Nuovo GJ, Plaia TW, Belinsky SA, Baylin SB, Herman JG, 1999, In situ detection of the hypermethylation-induced inactivation of the p16 gene as an early event in oncogenesis. Proc Natl Acad Sci U S A 96(22):12754–12759 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 1 EP 6 DI 10.1007/s12253-009-9178-5 PG 6 ER PT J AU Noel, JCh Fayt, I Buxant, F AF Noel, Jean-Christophe Fayt, Isabelle Buxant, Frederic TI Proliferating Activity in Paget Disease of the Nipple SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Paget disease; Nipple; Breast; Proliferation; Ki-67; Double stain immunohistochemistry ID Paget disease; Nipple; Breast; Proliferation; Ki-67; Double stain immunohistochemistry AB Paget disease of the nipple is a rare disease characterized by the presence of malignant glandular cells within the squamous epithelium of the nipple. The most common hypothesis to explain the development of Paget disease is an intraepithelial epidermotropic migration of malignant epithelial cells originating from an underlying intraductal carcinoma. If the immunohistochemical properties of the Paget cells in the nipple have been extensively studied, their proliferating characteristics remain paradoxically poorly studied. In the present study we have investigated the proliferating activity of Paget cells in the nipple by using double stain immunohistochemistry with both Ki67 (a protein which is expressed in all active parts of the cell cycle) and cytokeratin 7 (a highly sensitive marker of Paget cells). Ten cases of Paget disease and in their associated intraductal carcinomas (n=10) and/or invasive carcinomas (n=4) were tested. The mean Ki67 index was in Paget disease (26%±10), in intraductal carcinomas (23%±8) and/or in invasive carcinomas (20%±8) (p>0,05). This is the first report to convincingly demonstrate by specific double stain immunohistochemistry that Paget disease and underlying intraductal carcinomas share a close proliferating activity. C1 [Noel, Jean-Christophe] Free University of Brussels (ULB), Erasme Hospital, Departments of Pathology, 808 route de Lennik, 1070 Brussels, Belgium. [Fayt, Isabelle] Free University of Brussels (ULB), Erasme Hospital, Departments of Pathology, 808 route de Lennik, 1070 Brussels, Belgium. [Buxant, Frederic] Free University of Brussels (ULB), Erasme Hospital, Departments of GynecologyBrussels, Belgium. RP Noel, JCh (reprint author), Free University of Brussels (ULB), Erasme Hospital, Departments of Pathology, 1070 Brussels, Belgium. EM Jean-Christophe.Noel@erasme.ulb.ac.be CR Eusebi V, Mai KT, Taranger-Charpin A, 2003, Tumours of the nipple. In: Tavassoli FA, Devilee P, eds, World Health Organization Classification of Tumours. Pathology & Genetics. Tumours of the Breast and Female Genital Organs. IARC, Lyon, pp 104– 106 Ellis IO, Elston CW, Poller DN, 1998, Paget’s disease of the nipple. In: Elston CW, Ellis IO, eds, The breast. Churchill Linvingstone, Edinburgh, pp 276–281 Rosen PP, 2001, Paget’s disease of the nipple. In: Rosen PP, ed, Rosen’s breast pathology. Lippincott Williams & Wilkins, Philadelphia, pp 565–579 Cohen I, Guarner J, DeRose PB, 1993, Mammary Paget’s disease and associated carcinoma. An immunohistochemical study. Arch Pathol Lab Med 117:291–294 Fu W, Mittel VK, Young SC, 2001, Paget disease of the breast: analysis of 41 patients. Am J Clin Oncol 24:397–400 Kothari AS, Beechey-Newman N, Hamed H et al, 2002, Paget disease of the nipple: a multifocal manifestation of higher-risk disease. Cancer 95:1–7 Schelfhout VRJ, Coene ED, Delaey B et al, 2000, Pathogenesis of Paget’s disease: epidermal heregulin-alpha motility factor and the HER receptor family. J Natl Cancer Inst 92:622–628 De Potter CR, Eeckhout I, Schelfhout AM et al, 1994, Keratinocyte induced chemotaxis in the pathogenesis of Paget’s disease of the breast. Histopathology 24:349–356 Marucci G, Betts C, Golouh R et al, 2002, Toker cells are probably precursors of Paget cell carcinoma: a morphological and ultrastructural description. Virchows Arch 441:117–123 Lammie GA, Barnes DM, Millis RR, Gullick WJ, 1989, An immunohistochemical study of the presence of c-erb-2 protein in Paget’s disease of the nipple. Histopathology 15:505–514 Keatings L, Sinclair J, Wright C et al, 1990, C-erb-2 oncoprotein expression in mammary and extrammary Paget’s disease. Histopathology 17:243–247 Ellis PE, Wong Te Fong LF, MPhil R et al, 2002, The role of p53 and Ki67 in Paget’s disease of the vulva and the breast. Gynecologic Oncology 86:150–156 Endl E, Gerdes J, 2000, The Ki-67 protein: fascinating forms of unknown function. Exp Cell Res 257:231–237 Noel JC, Fayt I, Fernandez-Aguilar S et al, 2006, Proliferating activity in columnar cells lesions of the breast. Virchows Arch 449:617–621 Heenen M, Thiriar S, Noel JC et al, 1998, Ki-67 immunostaining of normal human epidermis: comparison with 3H-thymidine labelling and PCNA immunostaining. Dermatology 197:123– 126 Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer. The value of histological grade in breast: experience from a large study with long-term follow-up. Histopathology 19:403–410 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 7 EP 10 DI 10.1007/s12253-009-9179-4 PG 4 ER PT J AU Sipos, F Muzes, Gy Galamb, O Spisak, S Krenacs, T Toth, K Tulassay, Zs Molnar, B AF Sipos, Ferenc Muzes, Gyorgyi Galamb, Orsolya Spisak, Sandor Krenacs, Tibor Toth, Kinga Tulassay, Zsolt Molnar, Bela TI The Possible Role of Isolated Lymphoid Follicles in Colonic Mucosal Repair SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Isolated lymphoid follicle; Colon; Mucosal repair; Epithelial stem cell; Myofibroblast; Follicular dendritic cell; Bone marrow; Mesenchymal-epithelial transition ID Isolated lymphoid follicle; Colon; Mucosal repair; Epithelial stem cell; Myofibroblast; Follicular dendritic cell; Bone marrow; Mesenchymal-epithelial transition AB The continuous reformation and rapid repair of the colonic mucosa is essential for avoiding the aggregation of pernicious mutations induced by bacterial, toxic, or mitogenic factors. Gut-associated lymphoid tissue is supposed to play a central role in the organization of the repair mechanisms. In inflammatory conditions, the number, the diameter and the density of isolated lymphoid follicles (ILFs) are increasing. They are involved not just in immune surveillance, but their presence is also indispensable in normal mucosal regeneration of the colon. The relation of ILFs to the components of mucosal renewal such as bone marrow derived stem cells, follicular dendritic cells, subepithelial myofibroblasts or crypt formation has not been directly studied, and data about their putative organizer role are scattered in scientific literature. Whether they act as a regenerative pool containing stem cells in case of mucosal damage, or they are responsible only for the optimal cytokine milieu for the differentiation of immigrating stem cells is a question under debate. Our aim is to review the relation of ILFs to the different elements of colonic mucosal repair. C1 [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Muzes, Gyorgyi] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Galamb, Orsolya] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Spisak, Sandor] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Toth, Kinga] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. RP Sipos, F (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM sipos.ferenc@freemail.hu CR Nascimbeni R, Di Fabio F, Di Betta E et al, 2005, Morphology of colorectal lymphoid aggregates in cancer, diverticular and inflammatory bowel diseases. Mod Pathol 18:681–685 Dieu-Nosjean MC, Antoine M, Danel C et al, 2008, Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol 26:4410–4417 Shia J, Black D, Hummer AJ et al, 2008, Routinely assessed morphological features correlate with microsatellite instability status in endometrial cancer. Hum Pathol 39:116–125 Wada Y, Nakashima O, Kutami R et al, 1998, Clinicopathological study on hepatocellular carcinoma with lymphocytic infiltration. Hepatology 27:407–414 Saxena SK, Thompson JS, Sharp JG, 1997, Role of organized intestinal lymphoid aggregates in intestinal regeneration. J Invest Surg 10:97–103 Heel KA, McCauley RD, Papadimitriou JM et al, 1997, Peyer’s patches. J Gastroenterol Hepatol 12:122–136 Brandtzaeg P, Kiyono H, Pabst R et al, 2008, Terminology: nomenclature of mucosa-associated lymphoid tissue. Mucosal Immunology 1:31–37 Warger T, Schild H, Rechtsteiner G, 2007, Initiation of adaptive immune responses by transcutaneous immunization. Immunol Lett 109:13–20 Millonig G, Schwentner C, Mueller P et al, 2001, The vascularassociated lymphoid tissue: a new site of local immunity. Curr Opin Lipidol 12:547–553 Neutra MR, Mantis NJ, Kraehenbuhl JP, 2001, Collaboration of epithelial cells with organized mucosal lymphoid tissues. Nat Immunol 2:1004–1009 O’Brien LM, Fitzpatrick E, Baird AW et al, 2008, Eosinophilnerve interactions and neuronal plasticity in rat gut associated lymphoid tissue, GALT, in response to enteric parasitism. J Neuroimmunol 197:1–9 Neutra MR, Frey A, Kraehenbuhl JP, 1996, Epithelial M cells: gateways for mucosal infection and immunization. Cell 86:345– 348 Niedergang F, Kraehenbuhl JP, 2000, Much ado about M cells. Trends Cell Biol 10:137–141 Azzali G, 2003, Structure, lymphatic vascularization and lymphocyte migration in mucosa-associated lymphoid tisse. Immunol Rev 195:178–189 Baird AW, Cuthbert AW, 1987, Neuronal involvement in type 1 hypersensitivity reactions in gut epithelia. Br J Pharmacol 92:647– 655 O’Malley KE, Sloan T, Joyce P et al, 1993, Type I hypersensitivity reactions in intestinal mucosae from rats infected with Fasciola hepatica. Parasite Immunol 15:449–453 Vulchanova L, Casey MA, Crabb GW et al, 2007, Anatomical evidence for enteric neuroimmune interactions in Peyer’s patches. J Neuroimmunol 185:64–74 Chiocchetti R, Mazzuoli G, Albanese V et al, 2008, Anatomical evidence for ileal Peyer’s patches innervation by enteric nervous system: a potential route for prion neuroinvasion? Cell Tissue Res 332:185–194 Freemont AJ, Ford WL, 1985, Functional and morphological changes in post-capillary venules in relation to lymphocytic infiltration into BCG-induced granulomata in rat skin. J Pathol 147:1–12 Cavender DE, Haskard DO, Joseph B et al, 1986, Interleukin 1 increases the binding of human B and T lymphocytes to endothelial cell monolayers. J Immunol 136:203–207 Perry ME, Mustafa Y, Brown KA, 1992, The microvasculature of the human palatine tonsil and its role in the homing of lymphocytes. Adv Otorhinolaryngol 47:11–15 Kvietys PR, Sandig M, 2001, Neutrophil diapedesis: paracellular or transcellular? News Physiol Sci 16:15–19 Witmer AN, Dai J, Weich HA et al, 2002, Expression of vascular endothelial growth factor receptors 1, 2, and 3 in quiescent endothelia. J Histochem Cytochem 50:767–777 Rafii S, Lyden D, 2003, Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med 9:702–712 Takahashi T, Kalka C, Masuda H et al, 1999, Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nat Med 5:434–438 MacDermott RP, Sanderson IR, Reinecker HC, 1998, The central role of chemokines, chemotactic cytokines, in the immunopathogenesis of ulcerative colitis and Crohn’s disease. Inflamm Bowel Dis 4:54–67 Spyridonidis A, Schmitt-Graff A, Tomann T et al, 2004, Epithelial tissue chimerism after human hematopoietic cell transplantation is a real phenomenon. Am J Pathol 164:1147–1155 Korbling M, Katz RL, Khanna A et al, 2002, Hepatocytes and epithelial cells of donor origin in recipients of peripheral-blood stem cells. N Engl J Med 346:738–746 Gould VE, Bloom KJ, Franke WW et al, 1995, Increased numbers of cytokeratin-positive interstitial reticulum cells, CIRC, in reactive, inflammatory and neoplastic lymphadenopathies: hyperplasia or induced expression? Virchows Arch 425:617–629 Sipos F, Molnar B, Zagoni T et al, 2005, Growth in epithelial cell proliferation and apoptosis correlates specifically to the inflammation activity of inflammatory bowel diseases: ulcerative colitis shows specific p53- and EGFR expression alterations. Dis Colon Rectum 48:775–786 Sipos F, Galamb O, Herszenyi L et al, 2008, Elevated insulin-like growth factor 1 receptor, hepatocyte growth factor receptor and telomerase protein expression in mild ulcerative colitis. Scand J Gastroenterol 43:289–298 van Nierop K, de Groot C, 2002, Human follicular dendritic cells: function, origin and development. Semin Immunol 14:251–257 Park CS, Choi YS, 2005, How do follicular dendritic cells interact intimately with B cells in the germinal centre? Immunology 114:2–10 Lindhout E, Koopman G, Pals ST et al, 1997, Triple check for antigen specificity of B cells during germinal centre reactions. Immunol Today 18:573–577 Kapasi ZF, Qin D, Kerr WG et al, 1998, Follicular dendritic cell, FDC, precursors in primary lymphoid tissues. J Immunol 160:1078–1084 Lindhout E, de Groot C, 1995, Follicular dendritic cells and apoptosis: life and death in the germinal centre. Histochem J 27:167–183 Humphrey JH, Grennan D, Sundaram V, 1984, The origin of follicular dendritic cells in the mouse and the mechanism of trapping of immune complexes on them. Eur J Immunol 14:859– 864 Munoz-Fernandez R, Blanco FJ, Frecha C et al, 2006, Follicular dendritic cells are related to bone marrow stromal cell progenitors and to myofibroblasts. J Immunol 177:280–289 Powell DW, Mifflin RC, Valentich JD et al, 1999, Myofibroblasts. II. Intestinal subepithelial myofibroblasts. Am J Physiol 277: C183–201 Joyce NC, Haire MF, Palade GE, 1987, Morphologic and biochemical evidence for a contractile cell network within the rat intestinal mucosa. Gastroenterology 92:68–81 Andoh A, Bamba S, Fujiyama Yet al, 2005, Colonic subepithelial myofibroblasts in mucosal inflammation and repair: contribution of bone marrow-derived stem cells to the gut regenerative response. J Gastroenterol 40:1089–1099 Wallace JL, Granger DN, 1996, The cellular and molecular basis of gastric mucosal defense. FASEB J 10:731–740 Podolsky DK, 1997, Healing the epithelium: solving the problem from two sides. J Gastroenterol 32:122–126 Brittan M, Chance V, Elia G et al, 2005, A regenerative role for bone marrow following experimental colitis: contribution to neovasculogenesis and myofibroblasts. Gastroenterology 128:1984–1995 Fritsch C, Simon-Assmann P, Kedinger M et al, 1997, Cytokines modulate fibroblast phenotype and epithelial-stroma interactions in rat intestine. Gastroenterology 112:826–838 Guo L, Kuroda N, Nakayama H et al, 2006, Cytokeratin-positive subserosal myofibroblasts in gastroduodenal ulcer; another type of myofibroblasts. Histol Histopathol 21:697–704 Hamauzu T, Kuroda N, Guo L et al, 2006, CAM5.2-positive subserosal myofibroblasts in appendicitis. Med Mol Morphol 39:209–213 Yen TH, Wright NA, 2006, The gastrointestinal tract stem cell niche. Stem Cell Rev 2:203–212 Alison MR, Islam S, 2009, Attributes of adult stem cells. J Pathol 217:144–160 Stenson WF, 2008, Toll-like receptors and intestinal epithelial repair. Current Opinion in Gastroenterology 24:103–107 Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F et al, 2004, Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 118:229–241 Li M, Carpio D, Zheng Yet al, 2001, An essential role of the NFkappa B/toll-like receptor pathway in induction of inflammatory and tissue-repair gene expression by necrotic cells. J Immunol 166:7128–7135 Beg AA, 2002, Endogenous ligands of toll-like receptors: implications for regulating inflammatory and immune responses. Trends Immunol 23:509–512 Taylor K, Trowbridge J, Rudisill J et al, 2004, Hyaluronan fragments stimulate endothelial recognition of injury through TLR4. J Biol Chem 279:17079–17084 Majors A, Austin R, de la Motte C et al, 2003, Endoplasmic reticulum stress induces hyaluronan deposition and leukocyte adhesion. J Biol Chem 278:47223–47231 Onishi S, Miyata H, Inamoto T et al, 2007, Immunohistochemical study on the delayed progression of epithelial apoptosis in follicleassociated epithelium of rat Peyer’s patch. J Vet Med Sci 69:1123–1129 Renes IB, Verburg M, Bulsing NP et al, 2002, Protection of the Peyer’s patch-associated crypt and villus epithelium against methotrexate-induced damage is based on its distinct regulation of proliferation. J Pathol 198:60–68 Cadigan KM, Nusse R, 1997, Wnt signaling: a common theme in animal development. Genes Dev 11:3286–3305 DasGupta R, Fuchs E, 1999, Multiple roles for activated LEF/ TCF transcription complexes during hair follicle development and differentiation. Development 126:4557–4568 Korinek V, Barker N, Moerer P et al, 1998, Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat Genet 19:379–383 Haegebarth A, Clevers H, 2009, Wnt signaling, lgr5, and stem cells in the intestine and skin. Am J Pathol 174:715–721 Ouji Y, Yoshikawa M, Shiroi A et al, 2006, Wnt-10b secreted from lymphocytes promotes differentiation of skin epithelial cells. Biochem Biophys Res Commun 342:1063–1069 Malhotra S, Kincade PW, 2009, Wnt-related molecules and signaling pathway equilibrium in hematopoiesis. Cell Stem Cell 4:27–36 Thiery JP, 2003, Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol 15:740–746 Rubio D, Garcia S, De la Cueva T et al, 2008, Human mesenchymal stem cell transformation is associated with a mesenchymal-epithelial transition. Exp Cell Res 314:691–698 Prindull G, Zipori D, 2004, Environmental guidance of normal and tumor cell plasticity: epithelial mesenchymal transitions as a paradigm. Blood 103:2892–2899 Kang Y, Massague J, 2004, Epithelial-mesenchymal transitions: twist in development and metastasis. Cell 118:277–279 Spees JL, Olson SD, Ylostalo J et al, 2003, Differentiation, cell fusion, and nuclear fusion during ex vivo repair of epithelium by human adult stem cells from bone marrow stroma. Proc Natl Acad Sci USA 100:2397–2402 Van Arnam JS, Herzog E, Grove J et al, 2005, Engraftment of bone marrow-derived epithelial cells. Stem Cell Rev 1:21–27 Krause DS, 2005, Engraftment of bone marrow-derived epithelial cells. Ann N Y Acad Sci 1044:117–124 Hardeman EC, Chiu CP, Minty A et al, 1986, The pattern of actin expression in human fibroblast x mouse muscle heterokaryons suggests that human muscle regulatory factors are produced. Cell 47:123–130 Sheng W, Wang G, La Pierre DP et al, 2006, Versican mediates mesenchymal-epithelial transition. Mol Biol Cell 17:2009–2020 Hirose J, Kawashima H, Yoshie O et al, 2001, Versican interacts with chemokines and modulates cellular responses. J Biol Chem 276:5228–5234 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 11 EP 18 DI 10.1007/s12253-009-9181-x PG 8 ER PT J AU Nemeth, J Nemeth, Zs Tatrai, P Peter, I Somoracz, Szasz, MA Kiss, A Schaff, Zs AF Nemeth, Julia Nemeth, Zsuzsanna Tatrai, Peter Peter, Ilona Somoracz, Aron Szasz, Marcell Attila Kiss, Andras Schaff, Zsuzsa TI High Expression of Claudin-1 Protein in Papillary Thyroid Tumor and its Regional Lymph Node Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Claudin-1; Lymph node metastasis; Papillary thyroid carcinoma; Tumor marker ID Claudin-1; Lymph node metastasis; Papillary thyroid carcinoma; Tumor marker AB Claudins, known as major contributors in the formation of the tight junction, are differentially expressed in malignant tumors as compared to the corresponding healthy tissues. Therefore, they are thought to play a role in carcinogenesis and tumor progression. Altered expression of claudin-1 has been reported in several tumor types including endometrial, papillary renal cell and colonic carcinoma, and increased claudin-1 mRNA levels have been observed in papillary thyroid carcinoma (PTC). In this study, we aimed at determining the pattern of claudin-1 expression in various types of thyroid lesions at the protein level and investigating the immunolocalization of β-catenin reported to regulate claudin-1 expression. Samples included 19 PTCs, ten cases of corresponding regional lymph node metastasis, eight papillary microcarcinomas (PMC), 17 follicular thyroid carcinomas (FTC) and 19 follicular adenomas (FA). All cases were evaluated by quantitative immunohistochemistry. Conspicuous claudin-1 immunostaining was detected in the majority of PTC/PMC primary tumors and lymph node metastases (19/27 and 9/10, respectively). On the other hand, we found weak or no claudin-1 expression in any of the FA and FTC cases or peritumoral non-malignant thyroid tissues. Our data prove that high claudin-1 protein expression is specific for PTC and its regional lymph node metastases, while we failed to verify that claudin-1 is regulated by β-catenin in thyroid tumors. Based on these results, claudin-1 may be a useful tumor marker for PTC. C1 [Nemeth, Julia] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Nemeth, Zsuzsanna] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Tatrai, Peter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Peter, Ilona] National Institute of Oncology, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. [Somoracz, Aron] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Szasz, Marcell Attila] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM schaff@korb2.sote.hu CR Pacini F, Schlumberger M, Dralle H et al, 2006, European consensus for the management of patients with differentiated thyroid carcinoma of the follicular epithelium. Eur J Endocrinol 154:787–803 McNicol A, 2007, Pathology of thyroid tumours. Surgery 25:458–462 Kondo T, Ezzat S, Asa SL, 2006, Pathogenetic mechanisms in thyroid follicular-cell neoplasia. Nat Rev Cancer 6:292–306 DeLellis RA, Lloyd RV, Heitz PU, Eng C, eds,, 2004, World Health Organization classification of tumours. Pathology and genetics of tumours of endocrine organs. Thyroid and parathyroid tumours. IARC, Lyon, pp 51–123 Patricia SM, 2008, Thyroid epithelial tumours. Diagn Histopathol 14:236–246 Arif S, Blanes A, Diaz-Cano SJ, 2002, Hashimoto’s thyroiditis shares features with early papillary thyroid carcinoma. Histopathol 41:357–362 Di Pasquale M, Rothstein JL, Palazzo JP, 2001, Pathologic features of Hashimoto’s-associated papillary thyroid carcinomas. Hum Pathol 32:24–30 Repplinger D, Bargren A, Zhang YW et al, 2008, Is Hashimoto’s thyroiditis a risk factor for papillary thyroid cancer? J Surg Res 150:49–52 Gonzalez-Mariscal L, Betanzos A, Nava P et al, 2003, Tight junction proteins. Prog Biophys Mol Biol 81:1–44 Chiba H, Osanai M, Murata M et al, 2008, Transmembrane proteins of tight junctions. Biochim Biophys Acta 1778:588–600 Furuse M, Fujita K, Hiiragi T et al, 1998, Claudin-1 and -2: novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol 141:1539–1550 Hewitt KJ, Agarwal R, Morin PJ, 2006, The claudin gene family: expression in normal and neoplastic tissues. BMC Cancer 6:186 Soini Y, 2005, Expression of claudins 1, 2, 3, 4, 5 and 7 in various types of tumours. Histopathol 46:551–560 14 Swisshelm K, Macek R, Kubbies M, 2005, Role of claudins in tumorigenesis. Adv Drug Deliv Rev 57:919–928 Review 15. Sobel G, Nemeth J, Kiss A et al, 2006, Claudin 1 differentiates endometrioid and serous papillary endometrial adenocarcinoma. Gynecol Oncol 103:591–598 16. Fritzsche FR, Oelrich B, Johannsen M et al, 2008, Claudin-1 protein expression is a prognostic marker of patient survival in renal cell carcinomas. Clin Cancer Res 14:7035–7042 17. Resnick MB, Konkin T, Routhier J et al, 2005, Claudin-1 is a strong prognostic indicator in stage II colonic cancer: a tissue microarray study. Mod Pathol 18:511–518 18. Borka K, Kaliszky P, Szabo E et al, 2007, Claudin expression in pancreatic endocrine tumors as compared with ductal adenocarcinomas. Virchows Arch 450:549–557 19. Tokes AM, Kulka J, Paku S et al, 2005, Claudin-1, -3 and -4 proteins and mRNA expression in benign and malignant breast lesions: a research study. Breast Cancer Res 7:R296–R305 20. Sobel G, Paska C, Szabo I et al, 2005, Increased expression of claudins in cervical squamous intraepithelial neoplasia and invasive carcinoma. Hum Pathol 36:162–169 21. Krause G, Winkler L, Mueller SL et al, 2008, Structure and function of claudins. Biochim Biophys Acta 1778:631–645 22. Kominsky SL, 2006, Claudins: emerging targets for cancer therapy. Expert Rev Mol Med 8:1–11 23. Morin PJ, 2005, Claudin proteins in human cancer: promising new targets for diagnosis and therapy. Cancer Res 65:9603–9606 24. Tzelepi VN, Tsamandas AC, Vlotinou HD et al, 2008, Tight junctions in thyroid carcinogenesis: diverse expression of claudin- 1, claudin-4, claudin-7 and occludin in thyroid neoplasms. Mod Pathol 21:22–30 25. Hucz J, Kowalska M, Jarzab M et al, 2006, Gene expression of metalloproteinase 11, claudin 1 and selected adhesion related genes in papillary thyroid cancer. Endokrynol Pol 57(SupplA):18– 25 26. Fluge O, Bruland O, Akslen LA et al, 2006, Gene expression in poorly differentiated papillary thyroid carcinomas. Thyroid 16:161–175 27. Miwa N, Furuse M, Tsukita S et al, 2001, Involvement of claudin- 1 in the β-catenin/Tcf signaling pathway and its frequent pregulation in human colorectal cancers. Oncol Res 12:469–476 28. Halasz J, Holczbauer A, Cs P, Kovacs M, Benyo G, Verebely T, Zs S, Kiss A, 2006, Claudin-1 and claudin-2 differentiate fetal and embryonal components in human hepatoblastoma. Human Pathol 37:555–561 29. Liu YY, Morreau H, Kievit J et al, 2008, Combined immunostaining with galectin-3, fibronectin-1, CITED-1, Hector Battifora mesothelial-1, cytokeratin-19, peroxisome proliferator-activated receptor-{gamma}, and sodium/iodide symporter antibodies for the differential diagnosis of non-medullary thyroid carcinoma. Eur J Endocrinol 158:375–384 30. Dhawan P, Singh AB, Deane NG et al, 2005, Claudin-1 regulates cellular transformation and metastatic behavior in colon cancer. J Clin Invest 115:1765–1776 31. Wu YL, Zhang S, Wang GR et al, 2008, Expression transformation of claudin-1 in the process of gastric adenocarcinoma invasion. World J Gastroenterol 14:4943–4948 32. Kondo J, Sato F, Kusumi T et al, 2008, Claudin-1 expression is induced by tumor necrosis factor-alpha in human pancreatic cancer cells. Int J Mol Med 22:645–649 33. Chao YC, Pan SH, Yang SC et al, 2009, Claudin-1 is a metastasis suppressor and correlates with clinical outcome in lung adenocarcinoma. Am J Respir Crit Care Med 179:123–33 34. Aldred MA, Huang Y, Liyanarachchi S et al, 2004, Papillary and follicular thyroid carcinomas show distinctly different microarray expression profiles and can be distinguished by a minimum of five genes. J Clin Oncol 22:3531–3539 35. Firestein R, Bass AJ, Kim SY et al, 2008, CDK8 is a colorectal cancer oncogene that regulates beta-catenin activity. Nature 455:547–551 36. Elzagheid A, Buhmeida A, Korkeila E et al, 2008, Nuclear betacatenin expression as a prognostic factor in advanced colorectal carcinoma. World J Gastroenterol 14:3866–3871 37. Ishigaki K, Namba H, Nakashima M et al, 2002, Aberrant localization of beta-catenin correlates with overexpression of its target gene in human papillary thyroid cancer. J Clin Endocrinol Metab 87:3433–40 38. Wang Z, Qiu S, Eltorky MA et al, 2007, Histopathologic and immunohistochemical characterization of a primary papillary thyroid carcinoma in the lateral cervical lymph node. Exp Mol Pathol 82:91–4 39. Nikiforov YE, 2008, Thyroid carcinoma: molecular pathways and therapeutic targets. Mod Pathol 21(Suppl 2):S37–43 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 19 EP 27 DI 10.1007/s12253-009-9182-9 PG 9 ER PT J AU Szendroi, A Dinya, E Kardos, M Szasz, MA Nemeth, Zs Ats, K Kiss, J Antal, I Romics, I Szendroi, M AF Szendroi, Attila Dinya, Elek Kardos, Magdolna Szasz, Marcel Attila Nemeth, Zsuzsanna Ats, Katalin Kiss, Janos Antal, Imre Romics, Imre Szendroi, Miklos TI Prognostic Factors and Survival of Renal Clear Cell Carcinoma Patients with Bone Metastases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bone metastases; Fuhrman grade; Prognostic factors; Renal cell cancer; Surgical treatment; Survival analysis ID Bone metastases; Fuhrman grade; Prognostic factors; Renal cell cancer; Surgical treatment; Survival analysis AB In our retrospective study the pathological and clinical factors, influencing the survival of 65 renal clear cell carcinoma patients operated for bone metastasis between 1990 and 2008 were examined. Based on Kaplan-Meier curves age, gender, clinical symptoms, pathological fracture, progression to the soft tissues, localization and size of the metastasis, whether the occurrence of multiplex metastases is multiorganic or only located to the skeletal system and the stage and grade of primary renal cancer did not influence the survival. The survival significantly improved if the bone metastases were solitary, low Fuhrman grade, late onset; and radical surgery was performed. Based on Cox regression analysis, survival after bone surgery was influenced by the multiplicity and grade of metastasis and by the radicality of the surgery, whereas survival after nephrectomy was significantly influenced by onset time and grade of metastasis. When the solitary metastasis was radically removed, 75.0% of the patients survived the first, and 35.5% the fifth postoperative year. If the metastasis was multiple or the surgery was not radical, no patient survived the fifth year. This is the first report on the prognostic significance of the Fuhrman grade of bone metastasis of renal cell cancer. While the Fuhrman grade of the primary tumour did not influence the survival, the lower grade of metastasis was associated with a significant longer survival. Therefore in cases of solitary, operable, late onset metastases with low Fuhrman grade radical removal is recommended, since this way in 35.5% of cases 5 year survival can be expected. C1 [Szendroi, Attila] Semmelweis University, Department of Urology, Ulloi ut 78/b, H-1082 Budapest, Hungary. [Dinya, Elek] EGIS Pharmaceuticals PLC, Medical DepartmentBudapest, Hungary. [Kardos, Magdolna] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Szasz, Marcel Attila] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Nemeth, Zsuzsanna] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Ats, Katalin] National Institute for Rheumatology and PhysiotherapyBudapest, Hungary. [Kiss, Janos] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Antal, Imre] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Romics, Imre] Semmelweis University, Department of Urology, Ulloi ut 78/b, H-1082 Budapest, Hungary. [Szendroi, Miklos] Semmelweis University, Department of OrthopedicsBudapest, Hungary. RP Szendroi, A (reprint author), Semmelweis University, Department of Urology, H-1082 Budapest, Hungary. EM aszendroi@gmail.com CR Jemal A, Siegel R, Ward E et al, 2008, Cancer statistics, 2008. CA Cancer J Clin 58(2):71–96 Kozlowski JM, 1994, Management of distant solitary recurrence in the patient with renal cancer. Urol Clin North Am 21, 4):601–624 Park WH, Eisen T, 2007, Prognostic factors in renal cell cancer. BJU Int 99(5):1277–1281 Toyoda Y, Shinohara N, Harabayashi T et al, 2007, Survival and prognostic classification of patients with metastatic renal cell carcinoma of bone. Eur Urol 52(1):163–169 Barney JD, Churchill EJ, 1939, Adenocarcinoma of the kidney with metastasis to the lung: cured by nephrectomy and lobectomy. J Urol 42:269–276 Fuchs B, Trousdale RT, Rock MG, 2005, Solitary bony metastasis from renal cell carcinoma. Clin Orth Rel Res 431:187–192 Szendroi M, Sarvary A, 2001, Surgery of bone metastases. In: Besznyak I, ed, Diagnosis and surgery of organ metastases, 1st edn. Akademiai Kiado, Budapest Han R, Pantuck AJ, Bui MHT et al, 2003, Number of metastatic sites rather than location dictates overall survival of patients with node negative metastatic renal cell carcinoma. Urology 61:314–319 Lin PP, Mirza AN, Lewis VO et al, 2007, Patient survival after surgery for osseous metastases from renal cell carcinoma. J Bone Joint Surg 89:1794–1801 Althausen P, Althausen A, Jennings LC et al, 1997, Prognostic factors and surgical treatment of osseous metastases secondary to renal cell carcinoma cancer. 80:1103–1109 Kollender Y, Bickels J, Price WM et al, 2000, Metastatic renal cell carcinoma of bone: indications and technique of surgical intervention. J Urol 164:1505–1508 Durr HR, Maier M, Pfahler M et al, 1999, Surgical treatment of osseous metastases in patinets with renal cell carcinoma. Clin Orthop Relat Res 367:283–290 Swanson DA, Orovan WL, Johnson DE et al, 1981, Osseous metastasis secondary to renal cell carcinoma. Urology 18:556–561 Ficarra V, Righetti R, Martignoni G et al, 2001, Prognostic value of renal cell carcinoma nuclear grading: multivariate analysis of 333 cases. Urol Int 67(2):130–4 Minervini A, Lilas L, Minervini R et al, 2002, Prognostic value of nuclear grading in patients with intracapsular, pT1-pT2, renal cell carcinoma. Long-term analysis in 213 patients. Cancer 94, 10):2590–2595 Lang H, Lindner V, de Fromont M et al, 2005, Multicenter determination of optimal interobserver agreement using the Fuhrman grading system for renal cell carcinoma: assessment of 241 patients with > 15-year follow-up. Cancer 103(3):625–9 Ficarra V, Martignoni G, Maffei N et al, 2005, Original and reviewed nuclear grading according to the Fuhrman system: a multivariate analysis of 388 patients with conventional renal cell carcinoma. Cancer 103(1):68–75 Poel HG, Roukema JA, Horenblas S et al, 1999, Metastasectomy in renal cell carcinoma: a multicenter retrospective analysis. Eur Urol 35:197–203 Onishi T, Ohishi Y, Iizuka N et al, 1995, Study on the pathological grade in the primary and distant metastatic lesions of renal cell carcinoma. Hinyokika Kiyo 41(12):959–63 Baloch KG, Grimer RJ, Carter SR et al, 2000, Radical Surgery for the solitary bony metastasis from renal cell carcinoma. J Bone Joint Surg, Br, 82:62–67 Adiga GU, Dutcher JP, Larkin M, 2004, Characterisation of bone metastases in patients with renal cell cancer. BJU Int 93:1237–1240 Jung ST, Ghert MA, Harrelson JM et al, 2003, Treatment of osseous metastases in patients with renal cell carcinoma. Clin Orthop Rel Res 409:223–231 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 29 EP 38 DI 10.1007/s12253-009-9184-7 PG 10 ER PT J AU Li, Z Tu, J Yu, Zl Wu, Yd Xu, Cm Zhang, Sht AF Li, Zhang Tu, Jun Yu, Zhong-lin Wu, Yong-dong Xu, Cai-min Zhang, Shu-tian TI Effects of the Inhibition of Cyclooxygenase-2 on Human Esophageal Cancer Cells: Inhibition of Cell Proliferation and Induction of Apoptosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal squamous cell carcinoma (ESCC); Cyclooxygenase-2 (COX-2); Acidum acetil salicylicum (aspirin); Cell proliferation; Apoptosis; Synergistic effect ID Esophageal squamous cell carcinoma (ESCC); Cyclooxygenase-2 (COX-2); Acidum acetil salicylicum (aspirin); Cell proliferation; Apoptosis; Synergistic effect AB Cyclooxygenase-2 (COX-2) has been shown to be upregulated in a variety of tumors so that COX-2 may be a potential target in the treatment of cancer. In order to further explore the mechanism, we used RNA interference to study effects of the inhibition of COX-2 on esophageal squamous cell carcinoma (ESCC) lines. Western blot analysis demonstrated that COX-2 expression was significantly reduced in ESCC cells treated with the COX-2-specific siRNA. Furthermore, the COX-2 siRNA treatment inhibited cell proliferation and induced apoptosis in ESCC cells. In addition, the combination treatment of COX-2 siRNA and acidum acetil salicylicum (aspirin) has a synergistic effect. Therefore, this combination has potential as an anticancer therapy for the treatment of ESCC. C1 [Li, Zhang] Capital University of Medical Sciences, Beijing Friendship Hospital, Department of Digestive Diseases, 100050 Beijing, China. [Tu, Jun] University of Texas at San Antonio, Department of Biology, 78249 San Antonio, TX, USA. [Yu, Zhong-lin] Capital University of Medical Sciences, Beijing Friendship Hospital, Department of Digestive Diseases, 100050 Beijing, China. [Wu, Yong-dong] Capital University of Medical Sciences, Beijing Friendship Hospital, Department of Digestive Diseases, 100050 Beijing, China. [Xu, Cai-min] Chinese Academy of Medical Sciences, Department of Biochemistry and Molecular Biology, 100005 Beijing, China. [Zhang, Shu-tian] Capital University of Medical Sciences, Beijing Friendship Hospital, Department of Digestive Diseases, 100050 Beijing, China. RP Zhang, Sht (reprint author), Capital University of Medical Sciences, Beijing Friendship Hospital, Department of Digestive Diseases, 100050 Beijing, China. EM niuziyue1@hotmail.com CR Roth JA, Putnam JB, Rich TA et al, 1997, Cancer of the esophagus. In: Devita VT, Hellman S Jr, Rosenberg SA, eds, Cancer: principles and practice of the oncology. Lippincott-Raven, Philadephia, pp 980–1020 Buskens CJ, Van Rees BP, Sivula A et al, 2002, Prognostic significance of elevated cyclooxygensase-2 expression in patient with adenocarcinoma of the esophagus. Gastroenterology 122:1800–1807 Bakhle YS, 2001, COX-2 and cancer: a new approach to an old problem. Br J Pharmacol 134:1137–1150 Meric JB, Rottey S, Olaussen K et al, 2006, Cyclooxygenase-2 as a target for anticancer drug development. Crit Rev Oncol Hematol 59:51–64 Fearon ER, Volgelstein B, 1990, A genetic model for colorectal tumorigenesis. Cell 61:759–767 Wang LS, Chow KC, Wu YC, 2002, Effects of platelet activating factor, butyrate and interleukin-6 on cyclooxygenase-2 expression in human esophageal cancer cells. Scand J Gastroenterol 37:467– 475 Kase S, Osaki M, Honjo S et al, 2004, Expression of cyclooxygenase-1 and cyclooxygenase-2 in human esophageal mucosa, dysplasia and carcinoma. Pathobiology 71:84–92 Hashimoto N, Inayama M, Fujishima M et al, 2007, Clinicopathologic significance of expression of cyclooxygenase-2 in human esophageal squamous cell carcinoma. Hepatogastroenterology 54:758–760 Bantounas I, Phylactou LA, Uney JB, 2004, RNA interference and the use of small interfering RNA to study gene function in mammalian systems. J Mol Endocrinol 33:545–557 Strillacci A, Griffoni C, Spisni E et al, 2006, RNA interference as a key to knockdown overexpressed cyclooxygenase-2 gene in tumour cells. Br J Cancer 94:1300–1310 Charames GS, Bapat B, 2006, Cyclooxygenase-2 knockdown by RNA interference in colon cancer. Int J Oncol 28:543–549 Abdelrahim M, Safe S, Baker C et al, 2006, RNAi and cancer: Implication and applications. Journal of RNAi and Gene Silencing 2:136–145 Husain SS, Szabo IL, Tamawski AS, 2002, NSAID inhibition of GI cancer growth: clinical implications and molecular mechanisms of action. Am J Gastroentero l 97:542–553 Williams CS, Mann M, DuBois RN, 1999, The role of cyclooxygenases in inflammation, cancer, and development. Oncogene 18:7908–7916 Sumimoto H, 2005, Use of RNA interference technology for cancer specific gene silencing. Ann Cancer Res Therap 13:23–25 Dalby B, Cates S, Harris A et al, 2004, Advanced transfection with Lipofectamine 2000 reagent: primary neurons, siRNA, and high-throughput applications. Methods 33:95–103 Liu X, Li P, Zhang ST, You H et al, 2008, COX-2 mRNA expression in esophageal squmous cell carcinoma, ESCC, and effect by NSAID. Dis Esophagus 21:9–14 Zimmermann KC, Sarbia M, Weber AA, Borchard F, Gabbert HE, Schror K, 1999, Cyclooxygenase-2 expression in human esophageal carcinoma. Cancer Res 59:198–204 Li M, Lotan R, Levin B et al, 2000, Aspirin induction of apoptosis in esophageal cancer: a potential for chemoprevention. Epidemiol Biomarkers Prev 9:545–549 Tsujii M, DuBois RN, 1995, Alterations in cellular adhesion and apoptosis in epithelial cells overexpressing prostaglandin endoperoxide synthase 2. Cell 83:493–501 Zhi H, Wang L, Zhang J et al, 2006, Significance of COX-2 expression in human esophageal squamous cell carcinoma. Carcinogenesis 27:1214–1221 Sheng H, Shao J, Morrow JD et al, 1998, Modulation of apoptosis and Bcl-2 expression by prostaglandin E2 in human colon cancer cells. Cancer Res 56:362–366 Brown JR, DuBois RN, 2005, COX-2: a molecular target for colorectal cancer prevention. J Clin Oncol 23:2840–2855 Smith ML, Hawcroft G, Hull MA, 2000, The effect of nonsteroidal anti-inflammatory drugs on human colorectal cancer cells: evidence of different mechanisms of action. Eur J Cancer 36:664–674 Li P, Zhang ST, Yu ZL et al, 2009, Effects of cyclooxygenase-2 non-selective and selective inhibitors on proliferation inhibition and apoptosis induction of esophageal squamous carcinoma cells. Dis Esophagus 22:21–31 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 39 EP 45 DI 10.1007/s12253-009-9185-6 PG 7 ER PT J AU Zhang, Ch Tu, Z Du, Sh Wang, Y Wang, Q AF Zhang, Che Tu, Ziliang Du, Shiming Wang, Yong Wang, Qibin TI Expression of Matrix Metalloproteinase 2 and Extracellular Matrix Metalloproteinase Inducer are Unfavorable Postoperative Prognostic Factors in Intrahepatic Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intrahepatic cholangiocarcinoma; MMP-2; EMMPRIN; Clinicopathology; Prognosis ID Intrahepatic cholangiocarcinoma; MMP-2; EMMPRIN; Clinicopathology; Prognosis AB Many investigators have indicated that overexpression and amplification of matrix metalloproteinase 2 (MMP-2) and extracellular matrix metalloproteinase inducer (EMMPRIN) are independent prognostic factors for primary tumors. We studied expression of them in tissues from intrahepatic cholangiocarcinoma (IHCCA) and normal bile ducts, and discussed the occurrence and development of IHCCA. Another goal was to explore possible association of MMP-2 and EMMPRIN with clinicopathologic parameters and prognosis of IHCCA. MMP-2 and EMMPRIN expression in 106 cases of IHCCA tissues and 15 cases of normal bile ducts were examined by immunohistochemical staining. Then, the association of MMP-2 and EMMPRIN expression with clinicopathologic parameters and patients’ prognosis was analyzed. The positive expression levels of MMP-2 and EMMPRIN associated significantly with various clinicopathologic risk factors, such as poor histologic differentiation (p=0.03, 0.02), higher TNM stages (p=0.02, 0.01) and decreased tumor-specific survival. In particular, the tumor-specific survival rate of the patients with MMP-2+/ EMMPRIN+expression was the lowest (p<0.01). Using Cox regression analysis of the 89 patients, the conjoined expressions of MMP-2-/EMMPRIN-, MMP-2+/EMMPRIN+, histologic differentiation, and the clinical TNM stages of tumorous tissues were independent prognostic indicators of IHCCA (p<0.01, p<0.01, p=0.02, p=0.01 and p=0.01, respectively). MMP-2 and EMMPRIN expression in primary tumor predicts an unfavorable prognosis in IHCCA, suggesting a crucial role of the two markers in progression of human IHCCA. C1 [Zhang, Che] Yunyang Medical College, Taihe Hospital, 442000 Shiyan, Hubei, China. [Tu, Ziliang] Yunyang Medical College, Taihe Hospital, 442000 Shiyan, Hubei, China. [Du, Shiming] Yunyang Medical College, Taihe Hospital, 442000 Shiyan, Hubei, China. [Wang, Yong] Yunyang Medical College, Taihe Hospital, 442000 Shiyan, Hubei, China. [Wang, Qibin] Yunyang Medical College, Taihe Hospital, 442000 Shiyan, Hubei, China. RP Tu, Z (reprint author), Yunyang Medical College, Taihe Hospital, 442000 Shiyan, China. EM tzl6212@sina.com CR Itatsu K, Zen Y, Yamaguchi J, 2008, Expression of matrix metalloproteinase 7 is an unfavorable postoperative prognostic factor in cholangiocarcinoma of the perihilar, hilar, and extrahepatic bile ducts. Hum Pathol 39:710–719 Itatsu K, Zen Y, Ohira S, 2007, Immunohistochemical analysis of the progression of flat and papillary preneoplastic lesions in intrahepatic cholangiocarcinogenesis in hepatolithiasis. Liver Int 27:1174–1184 Park BK, Paik YH, Park JY et al, 2006, The clinicopathologic significance of the expression of vascular endothelial growth factor-C in intrahepatic cholangiocarcinoma. Am J Clin Oncol 29:138–142 Fu XH, Tang ZH, Zong M, Yang GS, Yao XP, Wu MC, 2004, Clinicopathologic features, diagnosis and surgical treatment of intrahepatic cholangiocarcinoma in 104 patients. Hepatobiliary Pancreat Dis Int 3:279–283 Malhi H, Gores GJ, 2006, Cholangiocarcinoma: modern advances in understanding a deadly old disease. J Hepatol 45:856–867 Nanashima A, Sumida Y, Abo T, Oikawa M, Murakami G, Takeshita H, Fukuoka H, Hidaka S, Nagayasu T, Sakamoto I, Sawai T, 2008, Relationship between pattern of tumor enhancement and clinicopathologic characteristics in intrahepatic cholangiocarcinoma. J Surg Oncol 98:535–539 Nabeshima K, Suzumiya J, Nagano M, Ohshima K, Toole BP, Tamura K et al, 2004, Emmprin, a cell surface inducer of matrix metalloproteinases, MMPs), is expressed in T-cell lymphomas. J Pathol 202:341–351 Davidson B, Goldberg I, Berner A, Kristensen GB, Reich R, 2003, EMMPRIN, extracellular matrix metalloproteinase inducer, is a novel marker of poor outcome in serous ovarian carcinoma. Clin Exp Metastasis 20:161–169 Tsai W-C, Chao Y-C, Sheu L-F, Lin Y-F, Nieh S, Chen A, Yu C-P, Jin J-S, 2007, EMMPRIN and fascin overexpression associated with clinicopathologic parameters of pancreatobiliary adenocarcinoma in Chinese people. APMIS 115:929–938 Riethdorf S, Reimers N, Assmann V, Kornfeld JW, Terracciano L, Sauter G et al, 2006, High incidence of EMMPRIN expression in human tumors. Int J Cancer 119:1800–1810 Guo H, Li R, Zucker S, Toole BP, 2000, EMMPRIN, CD147), an inducer of matrix metalloproteinase synthesis, also binds interstitial collagenase to the tumor cell surface. Cancer Res 60:888–891 Nabeshima K, Iwasaki H, Koga K, Hojo H, Suzumiya J, Kikuchi M, 2006, Emmprin, basigin/CD147): matrix metalloproteinase modulator and multifunctional cell recognition molecule that plays a critical role in cancer progression. Pathol Int 56:359–367 Cheng M-F, Tzao W, Tsai W-C, Lee W-H, Chen A, Chiang H, Sheu L-F, Jin J-S, 2006, Expression of EMMPRIN and matriptase in esophageal squamous cell carcinoma: correlation with clinicopathological parameters. Dis Esophagus 19:482–486 Cozzi PJ, Wang J, Delprado W, Perkins AC, Allen BJ, Russell PJ, Li Y, 2005, MUC1, MUC2, MUC4, MUC5AC and MUC6 expression in the progression of prostate cancer. Clin Exp Metastasis 22:565–573 Tang Y, Kesavan P, Marian T, 2004, Tumor-stroma interaction: positive feedback regulation of extracellular matrix metalloproteinase inducer, EMMPRIN, expressio and matrix metalloproteinase-dependent generation of soluble EMMPRIN molecular. Cancer Res 2:73–80 Romani AA, Soliani P, Desenzani S, Borghetti AF, Crafa P, 2006, The associated expression of Maspin and Bax proteins as a potential prognostic factor in intrahepatic cholangiocarcinoma. BMC Cancer 6:255 Li Q, Wang J-M, Liu C, Xiao B-L, Lu J-X, Zou S-Q, 2008, Correlation of aPKC-ι and E-cadherin expression with invasion and prognosis of cholangiocarcinoma. Hepatobiliary Pancreat Dis Int 7:70–75 Reimers N, Zafrakas K, Assmann V, Egen C, Riethdorf L, Riethdorf S et al, 2004, Expression of extracellular matrix metalloproteases inducer on micrometastatic and primary mammary carcinoma cells. Clin Cancer Res 10:3422–3428 Noguchi Y, Sato T, Hiratam M, Hara T, Ohama K, Ito A, 2003, Identification and characterization of extracellular matrix metalloproteinase inducer in human endometrium during the menstrual cycle in vivo and in vitro. J Clin Endocrinol Metab 88:6063–6072 Li W, Alfaidy N, Challis JRG, 2006, Expression of extracellular matrix metalloproteinase inducer in human placenta and fetal membranes at term labor. J Clin Endocrinol Metab 89:2897–2904 Haseneen NA, Vaday GG, Zucker S, Foda HD, 2003, Mechanical stretch induces MMP-2 release and activation in lung endothelium: role of EMMPRIN. Am J Physiol Lung Cell Mol Physiol 284: L541–L547 Jin J-S, Hsieh D-S, Lin Y-F, Wang J-Y, Sheu L-F, Lee W-H, 2006, Increasing expression of extracellular matrix metalloprotease inducer in renal cell carcinoma: tissue microarray analysis of immunostaining score with clinicopathological parameters. Int J Urol 13:573–580 Li QQ, Wang WJ, Xu JD, Cao XX, Chen Q, Yang JM, 2007, Upregulation of CD147 and matrix metalloproteinase-2, -9 induced by P-glycoprotein substrates in multidrug resistant breast cancer cells. Cancer Sci 98:1767–1774 Riethdorf S, Reimers N, Assmann V, Kornfeld JW, Terracciano L, Sauter G, 2006, High incidence of EMMPRIN expression in human tumors. Int J Cancer 119:1800–1810 Tang Y, Nakada MT, Kesavan P, McCabe F, Millar H, Rafferty P, 2005, Extracellular matrix metalloproteinase inducer stimulates tumor angiogenesis by elevating vascular endothelial cell growth factor and matrix metalloproteinases. Cancer Res 65:3193–3199 Ishibashi Y, Matsumoto T, Niwa M, Suzuki Y, Omura N, Hanyu N et al, 2004, CD147 and matrix metalloproteinase-2 protein expression as significant prognostic factors in esophageal squamous cell carcinoma. Cancer 101:1994–2000 Davidson B, Givant-Horwitz V, Lazarovici P, Risberg B, Nesland JM, Trope CG, Schaefer E, Reich R, 2003, Matrix metalloproteinases, MMP), EMMPRIN, extracellular matrix metalloproteinase inducer, and mitogen-activated protein kinases, MAPK): co-expression in metastatic serous ovarian carcinoma. Clin Exp Metastas 20:621–631 Sillanpaa S, Anttila M, Suhonen K, Hamalainen K, Turpeenniemi- Hujanen T, Puistola U et al, 2007, Prognostic significance of extracellular matrix metalloproteinase inducer and matrix metalloproteinase 2 in epithelial ovarian cancer. Tumour Biol 28:280–289 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 47 EP 53 DI 10.1007/s12253-009-9186-5 PG 7 ER PT J AU Guldur, EM Kibar, Y Deniz, H Bakir, K AF Guldur, Emin Muhammet Kibar, Yasemin Deniz, Hale Bakir, Kemal TI Comparison of Osteopontin, β-catenin and hnRNP B1 Expression in Lung Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteopontin; Beta-catenin; Heterogeneous nuclear ribonucleoprotein B1; Lung cancer ID Osteopontin; Beta-catenin; Heterogeneous nuclear ribonucleoprotein B1; Lung cancer AB This study was performed to compare osteopontin (OPN), β-catenin and heterogeneous nuclear ribonucleoprotein B1 (hnRNP B1) immunreactivities in small cell lung carcinomas (SCLC) and non-small cell lung carcinomas (NSCLC). Correlation of these three antibodies with grade and clinicopathologic stage of the tumor in NSCLC was also investigated. Twenty-nine SCLC, 6 large cell carcinoma, 36 adenocarcinoma and 30 squamous cell carcinoma (SCC), totally 101 cases, were included in this study. OPN, β-catenin and hnRNP B1 expressions were immunohistochemically evaluated. OPN positivity was 6.9% in SCLC and 67% in NSCLC. When NSCLC types were individually considered, OPN positivity was 66.7% in large cell carcinoma, 80% in SCC and 55.6% in adenocarcinomas. β-catenin positivity was observed in 48.6% of NSCLC and none of SCLC cases. These results were statistically significant (p<0.05). Neither grade nor stage of NSCLC was correlated with osteopontin, β-catenin or hnRNP B1 immunreactivity. We observed that OPN and β-catenin are useful in differentiating SCLC from NSCLC. This may be helpful in small lung biopsies where morphology is obscured by crush artifacts. C1 [Guldur, Emin Muhammet] Harran University, Faculty of Medicine, Department of Pathology, 63100 Sanliurfa, Turkey. [Kibar, Yasemin] Sehitkamil Hospital, Department of PathologyGaziantep, Turkey. [Deniz, Hale] Gaziantep Children Hospital, Department of Pathology, SehitkamilGaziantep, Turkey. [Bakir, Kemal] Gaziantep University, Faculty of Medicine, Department of PathologyGaziantep, Turkey. RP Deniz, H (reprint author), Gaziantep Children Hospital, Department of Pathology, Gaziantep, Turkey. EM haleden2000@yahoo.com CR Coppola D, Szabo M, Boulware D, Muraca P, Alsarraj M, Chambers AF, Yeatman TJ, 2004, Correlation of osteopontin protein expression and pathological stage across a wide variety of tumor histologies. Clin Cancer Res 10:184–190 Donati V, Boldrini L, Dell’Omodarme M, Prati MC, Faviana P, Camacci T, Lucchi M, Mussi A, Santoro M, Basolo F, Fontanini G, 2005, Osteopontin expression and prognostic significance in non-small cell lung cancer. Clin Cancer Res 11:6459–6465 Nozawa N, Hashimoto S, Nakashima Y, Matsuo Y, Koga T, Sugio K, Niho Y, Harada M, Sueishi K et al, 2006, Immunohistochemical α and β-catenin and E-cadherin expression and their clinocopathological significance in human lung adenocarcinoma. Pathol Res Pract 202:639–650 Prinen RT, Hirvikoski P, Johansson RT, Hollmen S, Kosma VM, 2001, Reduced expression of α-catenin, β-catenin is associated with high cell proliferative activity and poor differentiation in non-small cell lung cancer. J Clin Pathol 54:391–395 Sueoka E, Sueoka N, Goto Y, Matsuyama S, Nishimura H, Sato M, Fujimura S, Chiba H, Fujiki H, 2001, Heterogeneous nuclear ribonucleoprotein B1 as early cancer biomarker for occult cancer of human lungs and bronchial dysplasia. Cancer Res 61:1896– 1902 Zhou J, Mulshine JL, Unsworth EJ, Scott FM, Avis IM, Vos MD, Treston AM, 1996, Purification and characterization of a protein that permits early detection of lung cancer. Identification of heterogeneous nuclear ribonucleoprotein-A2/B1 as the antigen for monoclonal antibody 703D4. J Biol Chem 271:10760–10766 Zhang J, Takahashi K, Takahashi F, Shimizu K, Ohshita F, Kameda Y, Maeda K, Nishio K, Fukuchi Y, 2001, Differential osteopontin expression in lung cancer. Cancer Lett 171:215–222 Hu Z, Lin D, Yuan J, Xiao T, Zhang H, Sun W, Han N, Ma Y, Di X, Gao M, Ma J, Zhang J, Cheng S, Gao Y, 2005, Overexpression of osteopontin is associated with more aggressive phenotypes in human non-small cell lung cancer. Clin Cancer Res 11:4646–4652 Kase S, Sugio K, Yamazaki K, Okamoto T, Yano T, Sugimachi K, 2000, Expression of E-cadherin and β-Catenin in human nonsmall cell lung cancer and the clinical significance. Clin Cancer Res 6:4789–4796 Wu S, Sato M, Endo C, Sakurada A, Dong B, Aikawa H, Chen Y, Okada Y, Matsumura Y, Sueoka E, Kondo T, 2003, HnRNP B1 protein may be a possible prognostic factor in squamous cell carcinoma of the lung. Lung Cancer 41:179–186 Zech VF, Dlaska M, Tzankov A, Hilbe W, 2006, Prognostic and diagnostic relevance of hnRNP A2/B1, hnRNP B1 and S100 in non- small cell lung cancer. Cancer Detect Prev 30:395–402 Schneider S, Yochim J, Brabender J, Uchida K, Danenberg KD, Metzger R, Schneider PM, Salonga D, Holscher AH, Danenberg PV, 2004, Osteopontin but not osteonectin messenger RNA expression is a prognostic marker in curatively resected non-small cell lung cancer. Clin Cancer Res 10:1588–1596 Choi YS, Shim YM, Kim SH, Son DS, Lee HS, Kim GY, Han J, Kim J, 2003, Prognostic significance of E-cadherin and β-catenin in resected stage 1 non-small cell lung cancer. Eur J Cardiothorac Surg 24:441–449 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 55 EP 59 DI 10.1007/s12253-009-9187-4 PG 5 ER PT J AU Ragab, MS Samaka, MR Shams, MT AF Ragab, M Seham Samaka, M Rehab Shams, M Tahany TI HER2/neu Expression: A Predictor for Differentiation and Survival in Children With Wilms Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Her2/neu; Oncoprotein survival; Wilms tumor; Differentiation ID Her2/neu; Oncoprotein survival; Wilms tumor; Differentiation AB Wilms tumor is a mixed embroynal neoplasm of the kidney . HER2 is an onco-protein. Its over-expression could be implicated in the development of many tumors. The clinico-demographic and pathological data of 28 Wilms tumor patients were , reviewed. The tissue samples were examined by light Microscopy then immunohistochemical staining for HER2/neu expression. Additional 28 normal surrounding renal tissue specimens were included. There was significant differences between HER2/neu positive and HER2/neu negative Wilms tumors in relation to stage, histological phase and epithelial differentiation (P>0.05 for all). The overall survival advantage was noticed if Wilms tumor was at early stages (I and II) (Log-rank=13.23 and P>0.001), homologous epithelial differentiation (Log-rank=6.01 and P=0.04), as well as HER2/neu positive tumors (Log-rank=6.14 and P=0.013). A statistical significant trend toward a longer recurrence free survival was, noticed if Wilms tumor was at early stages (Log-rank=21.22, P>0.0000) and if HER2/neu positive (Log-rank=8.53, P=0.004). HER2/neu expression in Wilms tumor could be a marker for epithelial and homologous differentiation and its expression could be a good predictor for overall survival and longer recurrence free survival. C1 [Ragab, M Seham] Minoufiya University, Pediatric Department, Hematology and Oncology UnitMinoufiya, Egypt. [Samaka, M Rehab] Minoufiya University, Faculty of Medicine, The Department of PathologyMinoufiya, Egypt. [Shams, M Tahany] Suez Canal Universities, Faculty of Medicine, The Department of PathologySuez Canal, Egypt. RP Ragab, MS (reprint author), Minoufiya University, Pediatric Department, Hematology and Oncology Unit, Minoufiya, Egypt. EM seham172001@yahoo.com CR Jaffe N, Huff V, 2004, Neoplasms of the kidney. In: Behrman R, Liegman R, Jenson H, eds, Nelson Textbook of Pediactrics. 16th Edition. W.B. Saunder .Ch. 49. p 1711–1714 Lanzkowsky P, 2005, Wilms tumor :manual of pediatric hematology and oncology. 4th ed. Elsevier Academic Press. 19:548–560 Mokhtar N, Gouda I, Adel I, 2007, Cancer Pathology Registry 2003–2004 and Time Trend Analysis. 3:24–39 Ghanem M, Van Der Kwast T, Den Hollander J et al, 2001, Expression and prognostic value of epidermal growth factor receptor, transforming growth factor- alpha, and c-erb-B-2 in nephroblastoma. Cancer 92:3120–3129 Cendron M, Wajsman Z, 2006, http://WWW.e Medicine .com , Inc, Wilms Tumor excerpt. Last Updated: June 29 Egeler R, Wolff J, Anderson R, Coppes M, 1999, Long-term complications and post-treatment follow-up of patients with Wilms’ tumor. Semin Urol Oncol 17(1):55–61 Salem M, Kinoshita Y, Tajiri T et al, 2006, Association between the HER2 expression and histological differentiation in Wilms tumor. Pediatr Surg Int 22:891–896 Potti A, Forseen S, Koka V et al, 2004, Determination of HER-2/ neu overexpression and clinical predictors of survival in a cohort of 347 patients with primary malignant brain tumors. Cancer Investig 22(4):537–544 Meert A, Martin B, Paesmans M et al, 2003, The role of HER2/ neu expression on the survival of patients with lung cancer: a systemic review of the literature. Br J Cancer 89:959–965 van de Vijver M, 2001, Assessment of the need and appropriate method for testing for the human epidermal growth factor receptor-2(HER2). Eur J Cancer 37:11–17 Perlman E, Grosfeld J, Togashi K, Boccon-Gibod L, 2004, Nephroblastoma. In: Eble N, Sauter G, Epistein J, eds, Pathology and Genetics of tumors of the urinary system and male genital organs, Ch 1. IARC, Lyon, France, pp 48–52 Bozcuk H, Gumus A, Ozbilim G et al, 2005, Cluster analysis of P-Glycoprotein, c-erb-B2 and P53 in relation to tumor histology strongly indicates prognosis in patients with operable non small cell lung cancer. Med Sci Monit 11(6):11–20 http//www.cancer.gov,2007 Wilms Tumor and Other Childhood Kidney Tumors, PDQ®): Treatment Fernandes E, Parham D, Ribiro R, Douglas E, 1988, Teratoid Wilms tumor, an important variant of nephroblastoma. J Pediatr Surg 23:1131–1134 Jimenez R, Wallis T, Tabasczka P, Visscher D, 2000, Determination of HER2/neu status in breast carcinoma: comparative analysis of immunohistochemistry and fluorescent in situ hybridization. Mod Path 13:37–45 Dawson B, Trapp R, 2001, Basic and clinical biostatistics :large medical books. Oxford, London. Boston, pp 270–275 Rivera M, Haber D, 2005, Wilms tumor connecting tumorigenesis and organ development in the kidney. Nat Rev Cancer 5:699–713 Matth L, Bianchi F, Prato I et al, 2001, Renal cell cultures for the study of growth factor interactions underlying kidney organogenesis. Invitro Cell Dev Biol Anim 37:251–258 Taub M, Wang Y, Szczesny T, 1990, Epidermal growth factor or transforming growth factor α is required for kidney tubulogenesis in material cultures in serum-free medium. Proc Natl Acad Sci USA 87:4002–4006 Livasy C, Catrina Reading F, Moore D et al, 2006, EGFR expression and HER2/neu overexpression/amplification in endometrial carcinosarcoma. Gynecol Oncol 100:101–106 Nuciforo P, Pellegrini C, Fasani R et al, 2003, Molecular and Immunohistochemical Analysis of HER2/neu Oncogene in Synovial Sarcoma. Human Pathol 34(7):639–645 Green D, Children’s Oncology Group, 2007, Phase III multimodality therapy based on histology, stage, age, and tumor size in children with Wilms’ tumor, clear cell sarcoma of the kidney, or rhabdoid tumors of the kidney, COG-Q9401, Clinical trial, Completed. [PDQ Clinical Trial] Mitchell C, Jones P, Kelsey A et al, 2000, The treatment of Wilms’ tumour: results of the United Kingdom Children’s cancer study group, UKCCSG, second Wilms’ tumour study. Br J Cancer 83(5):602–608 Ritchey M, Shamberger R, Haase G et al, 2001, Surgical complications after primary nephrectomy for Wilms’ tumor: report from the ntional Wilms’ tumor study group. J Am Coll Surg 192(1):63–68 Yokoi A, McCrudden K, Huang J et al, 2003, Human epidermal growth factor receptor signaling contributes to tumor growth via angiogenesis in her2/neu-expressing experimental Wilms’ tumor. J Peditr Surg 38:1569–1573 Akslen L, Varhaug J, 1995, Oncoprotiens and tumor progression in papillary thyroid. Cancer 76:1643–1654 Sonia L, Ezzat S, Zheng L et al, 1998, Cytoplasmic staining of erbB-2not mRNA levels correlates with differentiation in human thyroid neoplesia. Clinical Endocrinol 49:629–637 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 61 EP 67 DI 10.1007/s12253-009-9188-3 PG 7 ER PT J AU Milicevic, MN Trbojevic-Stankovic, BJ Drachenberg, BC Milicevic, AF Milicevic, M Novica Trbojevic-Stankovic, B Jasna Drachenberg, B Cinthia Milicevic, Zivana TI Stereologic Analysis of Tissue Compartments of Gunshot-injured and Blunt-injured Spleen SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Blunt-injured spleen; Gunshot-injured spleen; Human; Morphometry; Stereology; Splenic tissue compartments ID Blunt-injured spleen; Gunshot-injured spleen; Human; Morphometry; Stereology; Splenic tissue compartments AB The spleen is composed of several tissue compartments and the respective histoquantitative data are essential for complete understanding of immune or pathological processes in this organ. The aim of our study was to determine and compare the stereologic parameters of all tissue compartments of the gunshotinjured and blunt-injured human spleen. The modelbased stereology with point-counting method was utilized to study the volume densities of red pulp, perifollicular zone, marginal zone, white pulp (follicles and periarteriolar lymphoid sheath), and connective tissue. The areal numerical density (the number of follicles per mm2 of tissue section), the numerical density (the number of follicles per mm3 of tissue) of lymphoid follicles and the mean follicle diameter were also determined. Our study provides stereological parameters for all tissue compartments of the human spleen. No morphometric differences were registered between tissue compartments of the blunt-injured and gunshot-injured spleen. As the gunshot-injured spleen was taken as presumably unstimulated in immunological regard, our results suggest that both gunshot-injured and blunt-injured organs may be used as models of the normal human spleen. C1 [Milicevic, M Novica] Faculty of Medicine, Institute of Histology and Embryology, Visegradska 26, 11000 Belgrade, Serbia. [Trbojevic-Stankovic, B Jasna] Clinical Centre, Department of HemodialysisBelgrade, Serbia. [Drachenberg, B Cinthia] University of Maryland, School of Medicine, Department of PathologyBaltimore, MD, USA. [Milicevic, Zivana] Faculty of Medicine, Institute of Histology and Embryology, Visegradska 26, 11000 Belgrade, Serbia. RP Milicevic, (reprint author), Faculty of Medicine, Institute of Histology and Embryology, 11000 Belgrade, Serbia. EM emilicez@sezampro.yu CR Adkins B, Charyulu V, Sun QL, Lobo D, Lopez DM, 2000, Early block in maturation is associated with thymic involution in mammary tumor-bearing mice. J Immunol 164:5635–5640 Benedict CA, De Trez C, Schneider K, Ha S, Patterson G, Ware CF, 2006, Specific remodeling of splenic architecture by cytomegalovirus. PLoS Pathogy 2:e16., DOI 10.1371/journal. ppat.0020016 Drachenberg CB, Papadimitriou JC, 1996, Splenic pathology in different forms of traumatic injury. Am J Clin Pathol 106:695 Farhi DC, Ashfaq R, 1996, Splenic pathology after traumatic injury. Am J Clin Pathol 105:474–478 Ghosh P, Komschlies KL, Cippitelli M et al, 1995, Gradual loss of T-helper 1 populations in spleen of mice during progressive tumor growth. J Natl Cancer Inst 87:1478–1483 Gunia S, Albrecht K, May M, Stosiek P, 2005, The white pulp in the setting of the septic spleen caused by different bacteria: a comparative morphometric study. APMIS 113:675–682 Hollowood K, Goodlad JR, 1998, Germinal centre cell kinetics. J Pathol 185:229–233 Kappler JW, Hoffmann M, 1973, Regulation of the immune response. III. Kinetic differences between thymus- and bone marrow-derived lymphocytes in the proliferative response to heterologous erythrocytes. J Exp Med 137:1325–1337 Khanna KM, McNamara JT, Lefrancois L, 2007, In situ imaging of the endogenous CD8 T cell response to infection. Science 318:116–120 Leemans R, Manson W, Snijder JA et al, 1999, Immune response capacity after human splenic autotransplantation: restoration of response to individual pneumococcal vaccine subtypes. Ann Surg 229:279–285 Mebius RE, Kraal G, 2005, Structure and function of the spleen. Nat Rev Immunol 5:606–616 Milicevic NM, Cuschieri A, Xuereb A, Milicevic Z, 1996, Stereologic study of splenic tissue compartments from traumatically injured and cancer patients. Gen Diagn Pathol 142:41–44 Milicevic Z, Cuschieri A, Xuereb A, Milicevic NM, 1996, Stereological study of tissue compartments of the human spleen. Histol Histopathol 11:833–836 Robinette CD, Fraumeni JF Jr, 1977, Splenectomy and subsequent mortality in veterans of the 1939–45 war. Lancet 310:127– 129 Segura JA, Barbero LG, Marquez J, 1997, Early tumor effect on splenic Th lymphocytes in mice. FEBS Lett 414:1–6 Stamou KM, Toutouzas KG, Kekis PB et al, 2006, Prospective study of the incidence and risk factors of postsplenectomy thrombosis of the portal, mesenteric, and splenic veins. Arch Surg 141:663–669 Sun QL, Charyulu V, Lobo D, Lopez DM, 2002, Role of thymic stromal cell dysfunction in the thymic involution of mammary tumor-bearing mice. Anticancer Res 22:91–96 Weibel ER, 1979, Practical methods for biological morphometry, vol 1. Academic, New York NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 69 EP 73 DI 10.1007/s12253-009-9189-2 PG 5 ER PT J AU Martic, NT Pecina-Slaus, N Kusec, V Kokotovic, T Musinovic, H Davor, T Zeljko, M AF Martic, Nikuseva Tamara Pecina-Slaus, Nives Kusec, Vesna Kokotovic, Tomislav Musinovic, Hana Davor, Tomas Zeljko, Martina TI Changes of AXIN-1 and Beta-Catenin in Neuroepithelial Brain Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Axin; Beta-catenin; Image analysis; Loss of heterozygosity; Neuroepithelial brain tumors; Wnt signaling ID Axin; Beta-catenin; Image analysis; Loss of heterozygosity; Neuroepithelial brain tumors; Wnt signaling AB In the present study changes of components of Wnt signaling pathway—axin (AXIN1) and beta-catenin (CTNNB1) in a sample of 72 neuroepithelial brain tumors were investigated. AXIN-1 gene was tested by PCR/loss of heterozygosity (LOH). Immunostaining and image analysis revealed the quantity and localization of relevant proteins. Polymorphic marker for AXIN-1, showed LOH in 11.1% of tumors. LOH was distributed to 6.3% of glioblastomas, one was found in neuroepithelial dysembrioplastic tumor and one in medulloblastoma. Down regulation of axin expression and up regulation of beta-catenin were detected in the analyzed tumors. Axin was observed in the cytoplasm in 68.8% of samples, in 28.1% in both the cytoplasm and nucleus and 3.1% had no expression. Beta-catenin was observed mainly in the nucleus and cytoplasm (59.4%). Expression in 34.4% of samples was in the cytoplasm and 6.3% showed no expression. Comparison of mean values of relative increase of axin and beta-catenin showed that they are significantly reversely proportional (P=0.014). Relative quantity of beta-catenin in patients with gross deletion of AXIN1 was significantly higher in comparison to patients without LOH (P=0.040). Our results demonstrate that changes of key components of the Wnt signaling play a role in neuroepithelial brain tumors. C1 [Martic, Nikuseva Tamara] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Laboratory of Neurooncology, Salata 12, 10000 Zagreb, Croatia. [Pecina-Slaus, Nives] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Laboratory of Neurooncology, Salata 12, 10000 Zagreb, Croatia. [Kusec, Vesna] Clinical Hospital Centre Zagreb, Clinical Institute of Laboratory Diagnosis, Kispaticeva 12, 10000 Zagreb, Croatia. [Kokotovic, Tomislav] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Laboratory of Neurooncology, Salata 12, 10000 Zagreb, Croatia. [Musinovic, Hana] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Laboratory of Neurooncology, Salata 12, 10000 Zagreb, Croatia. [Davor, Tomas] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska 29, 10000 Zagreb, Croatia. [Zeljko, Martina] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Laboratory of Neurooncology, Salata 12, 10000 Zagreb, Croatia. RP Pecina-Slaus, N (reprint author), School of Medicine University of Zagreb, Croatian Institute for Brain Research, Laboratory of Neurooncology, 10000 Zagreb, Croatia. EM nina@mef.hr CR Louis DN, Ohgaki H, Wiestler OD et al, 2007, The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114:97–109 Strother DR, Pollack IF, Gisher PG et al, 2002, Tumors of the central nervous system. In: Pizzo PA, Poplack DG, eds, Principles and practice of pediatric oncology. Wiliam, Philadelphia, pp 751–824 Gordon MD, Nusse R, 2006, Wnt signalling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem 281:22429–22433 Caricasole A, Bakker A, Copani A et al, 2005, Two sides of the same coin: Wnt signaling in neurodegeneration and neurooncology. Biosci Rep 25:309–327 Luo W, Lin SC, 2004, Axin: a master Scaffold for multiple signaling pathways. Neurosignals 13:99–113 Kikuchi A, 1999, Modulation of Wnt signaling by Axin and Axil. Cytokine Growth Factor Rev 10:255–265 Polakis P, 2007, The many ways of Wnt in cancer. Curr Opin Genet Dev 17:45–51 Patapoutian A, Reichardt LF, 2000, Roles of wnt proteins in neural development maintenance. Curr Opin Neurobiol 10:392–399 Lie DC, Colamarino SA, Song HJ et al, 2005, Wnt signalling regulates adult hippocampal neurogenesis. Nature 437:1370–1375 Yu X, Malenka RC, 2003, Beta-catenin is critical for dendritic morphogenesis. Nature Neurosci 6:1169–1177 Brakeman JS, Gu SH, Wang XB et al, 1999, Neuronal localization of the Adenomatous polyposis coli tumor suppressor protein. Neuroscience 91:661–672 Pecina-Slaus N, Zigmund M, Kusec Vet al, 2007, E-cadherin and beta-catenin expression patterns in malignant melanoma assessed by image analysis. J Cutan Pathol 34:239–246 Padden M, Leech S, Craig B et al, 2007, Differences in expression of junctional adhesion molecule—a and beta-catenin in multiple sclerosis brain tissue: increasing evidence for the role of tight junctions pathology. Acta Neuropathol 113:177–186 Cong F, Varmus H, 2004, Nuclear-cytoplasmic shuttling of Axin regulates subcellular localization of beta-catenin. Proc Natl Acad Sci USA 101:2882–2887 Anderson CB, Neufeld KL, White RL, 2002, Subcellular distribution of Wnt pathway proteins in normal and neoplastic colon. Proc Natl Acad Sci USA 99:8683–8688 Dahmen RP, Koch A, Denkhaus D et al, 2001, Deletions of AXIN1, a component of the WNT/wingless pathway, in sporadic medulloblastomas. Cancer Res 61:7039–7043 Yokota N, Nishizawa S, Ohta S et al, 2002, Role of wnt pathway in medulloblastoma oncogenesis. Int J Cancer 101:198–201 Giangaspero F, Wellek S, Masuoka J et al, 2006, Stratification of medulloblastoma on the basis of histopathological grading. Acta Neuropathol 112:5–12 Ellison DW, Onilude OE, Lindsey JC et al, 2005, United Kingdom children’s cancer study group brain tumour committee. Beta-catenin status predicts a favorable outcome in childhood medulloblastoma: the United Kingdom children’s cancer study group brain tumour committee. J Clin Oncol 23:7951–7957 Nikuseva Martic T, Beros V, Pecina-Slaus N et al, 2007, Genetic changes of CDH1, APC and CTNNB1 found in human brain tumors. Pathol Res Pract 203:779–787 Pecina-Slaus N, Nikuseva-Martic T, Beros V et al, 2007, Genetic alterations of E-cadherin and Beta-Catenin in germinoma and teratoma: report of two central nervous system cases. Pathol Oncol Res 13:370–374 Pecina-Slaus N, Nikuseva Martic T, Tomas D et al, 2008, Meningiomas exhibit loss of heterozygosity of the APC gene. J Neurooncol 87:63–70 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 75 EP 79 DI 10.1007/s12253-009-9190-9 PG 5 ER PT J AU Huang, LW Seow, KM Lee, ChCh Lin, YH Pan, HSh Chen, HJ AF Huang, Lee-Wen Seow, Kok-Min Lee, Chin-Cheng Lin, Yu-Hung Pan, Hun-Shan Chen, Heng-Ju TI Decreased p21 Expression in HPV-18 Positive Cervical Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human papillomavirus (HPV); p21; p53; p27; Cervical carcinoma ID Human papillomavirus (HPV); p21; p53; p27; Cervical carcinoma AB The aim of this study was to investigate the relationship between different human papillomaviruse (HPV) genotypes and the expression of p53, p21 and p27 in cervical carcinomas. A total of 103 cases of cervical carcinomas were assayed for expression of p53, p21 and p27 by immunohistochemistry. HPV typing was carried out by two polymerase chain reaction-based methods. Overall, HPV prevalence was 97.1% among the cervical carcinomas in this study. HPV-16 was detected in 66% of the tumors, HPV-18 in 7.8%, HPV-16/18 in 7.8% and other HPV types in 15.5%. The expression of p53 and p27 was not related to HPV genotype. However, in the HPV-18 positive cervical carcinomas, expression of p21 was significantly decreased or completely absent (P=0.019). Our results indicated that down-regulation of p21 was strongly associated with HPV-18 positive cervical carcinomas. The significantly lower expression of p21 protein in HPV-18 positive samples compared to HPV-18 negative cervical carcinomas supports the hypothesis that inactivation and degradation of p21 proteins by HPV-18 E7 may play an important role in the carcinogenesis of HPV-18 positive cervical neoplasia. C1 [Huang, Lee-Wen] Shin Kong Wu Ho-Su Memorial Hospital, Department of Obstetrics and Gynecology, 95 Wen Chang Road, Shih-Lin District, 111 Taipei, Taiwan, Republic of China. [Seow, Kok-Min] Shin Kong Wu Ho-Su Memorial Hospital, Department of Obstetrics and Gynecology, 95 Wen Chang Road, Shih-Lin District, 111 Taipei, Taiwan, Republic of China. [Lee, Chin-Cheng] Taipei Medical University, College of MedicineTaipei, Taiwan, Republic of China. [Lin, Yu-Hung] Shin Kong Wu Ho-Su Memorial Hospital, Department of Obstetrics and Gynecology, 95 Wen Chang Road, Shih-Lin District, 111 Taipei, Taiwan, Republic of China. [Pan, Hun-Shan] Shin Kong Wu Ho-Su Memorial Hospital, Department of Obstetrics and Gynecology, 95 Wen Chang Road, Shih-Lin District, 111 Taipei, Taiwan, Republic of China. [Chen, Heng-Ju] Shin Kong Wu Ho-Su Memorial Hospital, Department of Obstetrics and Gynecology, 95 Wen Chang Road, Shih-Lin District, 111 Taipei, Taiwan, Republic of China. RP Huang, LW (reprint author), Shin Kong Wu Ho-Su Memorial Hospital, Department of Obstetrics and Gynecology, 111 Taipei, Taiwan, Republic of China. EM m002057@ms.skh.org.tw CR Walboomers JMM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, Snijders PJF, Peto J, Meijer CJLM, Munoz N, 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189:12–19 Hawley-Nelson P, Vousden KH, Hubbert NL, Lowy DR, Schiller JT, 1989, HPV16 E6 and E7 proteins cooperate to immortalize human foreskin keratinocytes. EMBO J 8:3905–3910 Galloway DA, McDougall JK, 1996, The disruption of cell cycle checkpoints by papillomavirus oncoproteins contributes to anogenital neoplasia. Semin Cancer Biol 7:309–315 zur Hausen H, 2000, Papillomaviruses causing cancer: evasion from host-cell control in early events in carcinogenesis. J Natl Cancer Inst 92:690–698 Chen YQ, Cipriano SC, Arenkiel JM, Miller FR, 1995, Tumor suppression by p21WAF1. Cancer Res 55:4536–4539 Costa MJ, Hansen CL, Walls JE, Scudder SA, 1999, Immunohistochemical markers of cell cycle control applied to ovarian and primary peritoneal surface epithelial neoplasms: p21WAF1/CIP1 predicts survival and good response to platinin-based chemotherapy. Hum Pathol 30:640–647 Ropponen KM, Kellokoski JK, Lipponen PK, Pietilainen T, Eskelinen MJ, Alhava EM, Kosma VM, 1999, p21/WAF1 expression in human colorectal carcinoma: association with p53, transcription factor AP-2 and prognosis. Br J Cancer 81:133–140 Ogawa M, Maeda K, Onoda N, Chung YS, Sowa M, 1997, Loss of p21 WAF1/CIP1 expression correlates with disease progression in gastric carcinoma. Br J Cancer 75:1617–1620 Nakayama K, Ishida N, Shirane M, Inomata A, Inoue T, Shishido N, Horii I, Loh DY, Nakayama KI, 1996, Mice lacking p27Kip1 display increased body size, multiple organ hyperplasia, retinal dysplasia, and pituitary tumors. Cell 85:707–720 Troncone G, Vetrani A, Rosa GD, Gerbasio D, Palombini L, 1999, Cyclin dependent kinase inhibitor p27Kip1 expression in normal and neoplastic cervical epithelium. J Clin Pathol 52:880–887 Kim YT, Choi EK, Cho NH, Ko JH, Yang WI, Kim JW, Lee SH, 2000, Expression of cyclin E and p27KIP1 in cervical carcinoma. Cancer Lett 153:41–50 Levine AJ, 1997, p53, the cellular gatekeeper for growth and division. Cell 88:323–331 El-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM, Lin D, Mercer WE, Kinzler KW, Vogelstein B, 1993, WAF1, a potential mediator of p53 tumor suppressor. Cell 75:817–825 Hollstein M, Sidransky D, Vogelstein B, Harris CC, 1991, p53 mutations in human cancers. Science 253:49–53 Woodman CB, Collins S, Rollason TP, Winter H, Bailey A, Yates M et al, 2003, Human papillomavirus type 18 and rapidly progressing cervical intraepithelial neoplasia. Lancet 361:40–43 Liaw KL, Glass AG, Manos MM, Greer CE, Scott DR, Sherman M et al, 1999, Detection of human papillomavirus DNA in cytologically normal women and subsequent cervical squamous intraepithelial lesions. J Natl Cancer Inst 91:954–960 Londesborough P, Ho L, Terry G, Cuzick J, Wheeler C, Singer A, 1996, Human papillomavirus genotype as a predictor of persistence and development of high-grade lesions in women with minor cervical abnormalities. Int J Cancer 69:364–368 Huang LW, Chao SL, Hwang JL, Chou YY, 2002, Down-regulation of p27 is associated with malignant transformation and aggressive phenotype of cervical neoplasms. Gynecol Oncol 85:524–528 Huang LW, Chou YY, Chao SL, Chen TJ, Lee TT, 2001, p53 and p21 expression in precancerous lesions and carcinomas of the uterine cervix: overexpression of p53 predicts poor disease outcome. Gynecol Oncol. 83:348–354 Scheffner M, Werness BA, Huibregtse JM, Levine AJ, Howley PM, 1990, The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of p53. Cell 63:1129–1136 Funk JO, Waga S, Harry JB, Espling E, Stillman B, Galloway DA, 1997, Inhibition of CDK activity and PCNA-dependent DNA replication by p21 is blocked by interaction with the HPV-16 E7 oncoprotein. Genes Dev 11:2090–2100 Zerfass-Thome K, Zwerschke W, Mannhardt B, Tindle R, Botz JW, Jansen-Durr P, 1996, Inactivation of the cdk inhibitor p27KIP1 by the human papillomavirus type 16 E7 oncoprotein. Oncogene. 13:2323–2330 Huang LW, Chao SL, Chen PH, Chou HP, 2004, Multiple HPV genotypes in cervical carcinomas: improved DNA detection and typing in archival tissues. J Clin Virol 29:271–276 Lorincz AT, Reid R, Jenson AB, Greenberg MD, Lancaster W, Kurman RJ, 1992, Human papillomavirus infection of the cervix: relative risk associations of 15 common anogenital types. Obstet Gynecol 79:328–337 Lie AK, Skarsvag S, Skomedal H, Haugen OA, Holm R, 1999, Expression of p53, MDM2, and p21 proteins in high-grade cervical intraepithelial neoplasia and relationship to human papillomavirus infection. Int J Gynecol Pathol 18:5–11 Cavuslu S, Goodlad J, Hobbs C, Connor AM, Raju KS, Best JM, Cason J, 1997, Relationship between human papillomavirus infection and overexpression of p53 protein in cervical carcinomas and lymph node metastases. J Med Virol 53:111–117 Michieli P, Chedid M, Lin D, Pierce JH, Mercer WE, Givol D, 1994, Induction of WAF1/CIP1 by a p53-independent Pathway. Cancer Res 54:3391–3395 Parker SB, Eichele G, Zhang P, Rawls A, Sands AT, Bradley A, Olson EN, Harper JW, Elledge SJ, 1995, p53-independent expression of p21Cip1 in muscle and other terminally differentiating cells. Science 267:1024–1027 Jones DL, Alani RM, Munger K, 1997, The human papillomavirus E7 oncoprotein can uncouple cellular differentiation and proliferation in human keratinocytes by abrogating p21Cip1- mediated inhibition of cdk2. Genes Dev 11:2101–2111 Rangarajan A, Talora C, Okuyama R, Nicolas M, Mammucari C, Oh H, Aster JC, Krishna S, Metzger D, Chambon P, Miele L, Aguet M, Radtke F, Dotto GP, 2001, Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. EMBO J. 20:3427–3436 Werness BA, Wang HQ, Chance J, Goldstein DJ, 1997, p53- independent expression of p21waf1/cip1 in preinvasive and invasive squamous neoplasms of the uterine cervix. Mod Pathol 10:578–584 Nadal A, Jares P, Cazorla M, Fernandez PL, Sanjuan X, Hernandez L, Pinyol M, AldeaM,Mallofre C,Muntane J, Traserra J, Campo E, Cardesa A, 1997, p21WAF1/Cip1 expression is associated with cell differentiation but not with p53 mutations in squamous cell carcinomas of the larynx. J Pathol 183:156–163 Burger RA, Monk BJ, Kurosaki T, Anton-Culver H, Vasilev SA, Berman ML, Wilczynski SP, 1996, Human papillomavirus type 18: association with poor prognosis in early stage cervical cancer. J Natl Cancer Inst 88:1361–1368 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 81 EP 86 DI 10.1007/s12253-009-9191-8 PG 6 ER PT J AU Wong, H Yau, Th Chan, P Ng, OLI Chan, G Hui, P Law, LW Lo, MC Hedley, JA Epstein, JR AF Wong, Hilda Yau, Thomas Chan, Pierre Ng, O L Irene Chan, Gavin Hui, Peter Law, L W Lo, M C Hedley, J Antony Epstein, J Richard TI PPI-Delayed Diagnosis of Gastrinoma: Oncologic Victim of Pharmacologic Success SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Neuroendocrine carcinoma; Pancreatic tumor; Zollinger–Ellison syndrome; Gastrin; Omeprazole ID Neuroendocrine carcinoma; Pancreatic tumor; Zollinger–Ellison syndrome; Gastrin; Omeprazole AB Functional neuroendocrine tumors are often lowgrade malignant neoplasms that can be cured by surgery if detected early, and such detection may in turn be accelerated by the recognition of neuropeptide hypersecretion syndromes. Uniquely, however, relief of peptic symptoms induced by hypergastrinemia is now available from acid-suppressive drugs such as proton-pump inhibitors (PPIs). Here we describe a clinical case in which time to diagnosis from the onset of peptic symptoms was delayed more than 10 years, in part reflecting symptom masking by continuous prescription of the PPI omeprazole.We propose diagnostic criteria for this under-recognized new clinical syndrome, and recommend that physicians routinely measure serum gastrin levels in persistent cases of PPI-dependent dyspepsia unassociated with H. pylori. C1 [Wong, Hilda] Queen Mary Hospital, Department of Medicine, Division of Hematology/Oncology, Professorial Block, 102 Pokfulam RoadHong Kong, Hong Kong. [Yau, Thomas] Queen Mary Hospital, Department of Medicine, Division of Hematology/Oncology, Professorial Block, 102 Pokfulam RoadHong Kong, Hong Kong. [Chan, Pierre] Queen Mary Hospital, Department of Medicine, Division of GastroenterologyHong Kong, Hong Kong. [Ng, O L Irene] The University of Hong Kong, Queen Mary Hospital, Department of PathologyHong Kong, Hong Kong. [Chan, Gavin] The University of Hong Kong, Queen Mary Hospital, Department of PathologyHong Kong, Hong Kong. [Hui, Peter] The University of Hong Kong, Queen Mary Hospital, Department of RadiologyHong Kong, Hong Kong. [Law, L W] The University of Hong Kong, Queen Mary Hospital, Department of SurgeryHong Kong, Hong Kong. [Lo, M C] The University of Hong Kong, Queen Mary Hospital, Department of SurgeryHong Kong, Hong Kong. [Hedley, J Antony] The University of Hong Kong, Department of Community MedicineHong Kong, Hong Kong. [Epstein, J Richard] Queen Mary Hospital, Department of Medicine, Division of Hematology/Oncology, Professorial Block, 102 Pokfulam RoadHong Kong, Hong Kong. RP Epstein, JR (reprint author), Queen Mary Hospital, Department of Medicine, Division of Hematology/Oncology, Hong Kong, Hong Kong. EM repstein@hku.hk;repstein@stvincents.com.au CR Ellison EC, Sparks J, 2003, Zollinger–Ellison syndrome in the era of effective acid suppression: are we unknowingly growing tumors? Am J Surg 186:245–8 Corleto VD, Annibale B, Gibril F et al, 2001, Does the widespread use of proton pump inhibitors mask, complicate and/ or delay the diagnosis of Zollinger–Ellison syndrome? Aliment Pharmacol Ther 15:1555–61 Roy PK, Venzon DJ, Shojamanesh H et al, 2000, Zollinger– Ellison syndrome. Clinical presentation in 261 patients. Medicine, Baltimore, 79:379–411 Ellison EH, Wilson SD, 1964, The Zollinger–Ellison syndrome: re-appraisal and evaluation of 260 registered cases. Ann Surg 160:512–30 Zollinger RM, Ellison EC, O’Dorisio TM et al, 1984, Thirty years’ experience with gastrinoma. World J Surg 8:427–35 Jensen RT, Gardner JD, Raufman JP et al, 1983, Zollinger– Ellison syndrome: current concepts and management. Ann Intern Med 98:59–75 DeVita VT, Lawrence TS, Rosenberg SA, 2008, Cancer: principles of oncology. 8th Ed. Lippincott Williams & Welkins. Blonski WC, Katzka DA, Lichtenstein GR et al, 2005, Idiopathic gastric acid hypersecretion presenting as a diarrheal disorder and mimicking both Zollinger–Ellison syndrome and Crohn’s disease. Eur J Gastroenterol Hepatol 17:441–4 Roy PK, Venzon DJ, Feigenbaum KM et al, 2001, Gastric secretion in Zollinger–Ellison syndrome. Correlation with clinical expression, tumor extent and role in diagnosis—-a prospective NIH study of 235 patients and a review of 984 cases in the literature. Medicine, Baltimore, 80:189–222 Maton PN, 1993, Use of octreotide acetate for control of symptoms in patients with islet cell tumors. World J Surg 17:504–10 Fukui T, Nishio A, Okazaki K et al, 2006, Gastric mucosal hyperplasia via upregulation of gastrin induced by persistent activation of gastric innate immunity in major histocompatibility complex class II deficient mice. Gut 55:607–15 Metz DC, Comer GM, Soffer E et al, 2006, Three-year oral pantoprazole administration is effective for patients with Zollinger– Ellison syndrome and other hypersecretory conditions. Aliment Pharmacol Ther 23:437–44 Metz DC, Sostek MB, Ruszniewski P et al, 2007, Effects of esomeprazole on acid output in patients with Zollinger–Ellison syndrome or idiopathic gastric acid hypersecretion. Am J Gastroenterol 102:2648–54 Banasch M, Schmitz F, 2007, Diagnosis and treatment of gastrinoma in the era of proton pump inhibitors. Wien Klin Wochenschr 119:573–8 Lodish MB, Powell AC, Abu-Asab M et al, 2008, Insulinoma and gastrinoma syndromes from a single intrapancreatic neuroendocrine tumor. J Clin Endocrinol Metab 93:1123–8 Gurevich L, Kazantseva I, Isakov VA et al, 2003, The analysis of immunophenotype of gastrin-producing tumors of the pancreas and gastrointestinal tract. Cancer 98:1967–76 Bamba T, Kosugi S, Kanda T et al, 2007, Multiple carcinoids in the duodenum, pancreas and stomach accompanied with type A gastritis: a case report. World J Gastroenterol 13:2247–9 Ardill JE, Erikkson B, 2003, The importance of the measurement of circulating markers in patients with neuroendocrine tumours of the pancreas and gut. Endocr Relat Cancer 10:459–62 Gibril F, Reynolds JC, Doppman JL et al, 1996, Somatostatin receptor scintigraphy: its sensitivity compared with that of other imaging methods in detecting primary and metastatic gastrinomas A prospective study. Ann Intern Med 125:26–34 Zimmer T, Stolzel U, Bader M et al, 1996, Endoscopic ultrasonography and somatostatin receptor scintigraphy in the preoperative localisation of insulinomas and gastrinomas. Gut 39:562–8 Alexander HR, Fraker DL, Norton JA et al, 1998, Prospective study of somatostatin receptor scintigraphy and its effect on operative outcome in patients with Zollinger–Ellison syndrome. Ann Surg 228:228–38 Thompson NW, Eckhauser FE, Vinik AI et al, 1984, Cystic neuroendocrine neoplasms of the pancreas and liver. Ann Surg 199:158–64 van Santen S, van Soest EJ, Busch OR et al, 2007, Awoman with shock as first sign of Zollinger–Ellison syndrome. Ned Tijdschr Geneeskd 151:478–83 Tartaglia A, Vezzadini C, Bianchini S et al, 2005, Gastrinoma of the stomach: a case report. Int J Gastrointest Cancer 35:211–6 Norton JA, Fraker DL, Alexander HR et al, 1999, Surgery to cure the Zollinger–Ellison syndrome. N Engl J Med 341:635–44 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 87 EP 91 DI 10.1007/s12253-009-9192-7 PG 5 ER PT J AU Vitanovics, D Balint, K Hanzely, Z Banczerowski, P Afra, D AF Vitanovics, Dusan Balint, Katalin Hanzely, Zoltan Banczerowski, Peter Afra, Denes TI Ependymoma in Adults: Surgery, Reoperation and Radiotherapy for Survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adult ependymoma; Radiotherapy; Reoperation; Surgery ID Adult ependymoma; Radiotherapy; Reoperation; Surgery AB Purpose: to retrospectively determine the longterm outcome of adult intracranial ependymoma patients treated with surgery, reoperation, and postoperative radiation therapy. Material and Methods: 61 patients were treated at our institution between 1980 and 2004. Forty patients had World Health Organization (WHO) Grade II ependymoma, and 21 patients had Grade III ependymoma. The median age was 34 years. The majority of patients were female (59%), and 35 had gross total resections (60%). Eighteen patients were reoperated, 15 only once but 2 twice and one six times. Survival times following reoperation was mostly short but some of them reached more than 5 or 10 years. Postoperative radiation therapy was delivered to 31 patients postoperative (55.4%) and to 5 after reoperation, a median total dose of 54 Gy. Results: The median follow-up of surviving patients was 10.6 years. The 5-year and 10-year diseasefree survival rates for all patients were 50% and 32.9% respectively. The 5-year and 10-year overall survival rates for all patients were 57.1% and 39.4%, respectively. A statistically significant effect on prognosis was observed with WHO tumour grade as well as with MIB-1 labelling index. Subtotal resection predicted a worse overall survival, but this failed to reach statistical significance. No statistically significant effect on prognosis was observed with tumour location and radiation therapy. Conclusion: In our experience the use of radiotherapy in adult, intracranial WHO Grade II ependymoma patients had no significant effect on prognosis. Radical surgery and eventual reoperation seems to be more favorable. C1 [Vitanovics, Dusan] National Institute of Neurosurgery, Amerikai ut 57, 1145 Budapest, Hungary. [Balint, Katalin] National Institute of Neurosurgery, Amerikai ut 57, 1145 Budapest, Hungary. [Hanzely, Zoltan] National Institute of Neurosurgery, Amerikai ut 57, 1145 Budapest, Hungary. [Banczerowski, Peter] National Institute of Neurosurgery, Amerikai ut 57, 1145 Budapest, Hungary. [Afra, Denes] National Institute of Neurosurgery, Amerikai ut 57, 1145 Budapest, Hungary. RP Afra, D (reprint author), National Institute of Neurosurgery, 1145 Budapest, Hungary. EM afraden@hotmail.com CR Reni M, Gatta G, Mazza E, Vecht Ch, 2007, Ependymoma. Crit Rev Oncol/Hematol 63:81–89 Burger PSB, 1994, Tumors of the central nervous system: vol. fascicle 10. D.C. Armed Forces Institute of Pathology, Washington D.C Chin HW, Hazel JJ, Kim TH, Freeman C, Maruyama Y, 1984, Intracranial ependymomas and ependymoblastomas. Strahlentherapie 160:191–194 Dohrmann GJ, Farwell JR, Flannery JT, 1976, Ependymomas and ependymoblastomas in children. J Neurosurg 45:273–283 Louis DN, Ohgaki H, Wiestler OD, Canevee WK, 2007, WHO classification of tumours of the central nervous system. International Agency for Research on Cancer, Lyon Schiffer D, Chio A, Giordana MT, Migheli A, Palma L, Pollo B, Soffietti R, Triboli A, 1991, Histologic prognostic factors in ependymoma. Childs Nerv Syst 7:177–182 Metellus A, Barrie M, Figarella-Branger D, Chinot O, Giorgi R, Gouvernet J, Jouvet A, Guyotat J, 2007, Multicentric French study on adult intracranial ependymomas: prognostic factors analysis and therapeutic considerations from a cohort of 152 patients. Brain 130:1338–1349 Afra D, Muller W, Slowik F, Wilcke O, Budka H, Turoczy L, 1983, Supratentorial lobar ependymomas: reports on the grading and survival periods in 80 cases, including 46 recurrences. Acta Neurochir [Wien] 69:243–251 McLaughlin MP, Marcus RB Jr, Buatti JM, McCollough WM, Mickle JP, Kedar A, Maria BL, Lillion RR, 1998, Ependymoma: results, prognostic factors and treatment recommendations. Int J Radiat Oncol Biol Phys 40:845–850 Gerszten PC, Pollack IF, Martinez AJ, Lo KH, Janosky J, Albright AL, 1996, Intracranial ependymomas of childhood. Lack of correlation of histopathology and clinical outcome. Pathol Res Pract 192:515–522 Figarella-Branger D, Civatte M, Bouvier-Labit C, Gouvernet J, Gambarelli D, Gentet JC, Lena G, Choux M, Pellissier JF, 2000, Prognostic factors in intracranial ependymomas in children. J Neurosurg 93:605–611 Lyons MK, Kelly PJ, 1991, Posterior fossa ependymomas: report of 30 cases and review of the literature. Neurosurgery 28:659– 664 Marks JE, Adler SJ, 1982, A comparative study of ependymomas by site of origin. Int J Radiat Oncol Biol Phys 8:37–43 Choux M, Pellissier JF, 2000, Prognostic factors in intracranial ependymomas in children. J Neurosurg 93:605–613 Korshunov A, Golanov A, Sycheva R, Timirgaz V, 2007, The histologic grade is a main prognostic factor for patients with intracranial ependymomas treated in the microneurosurgical era: an analysis of 258 patients. Cancer 100:1230–1237 Timmermann B, Kortmann RD, Kuhl J, Willich N, Bamberg M, 2002, Die interdisziplinare Therapie von Ependymomen im Kindesalter. Strahlenther Onkol. 178:469–479 Ross GW, Rubinstein LJ, 1989, Lack of histopathological correlation of malignant ependymomas with postoperative survival. J Neurosurg 70:31–36 Preusser M, Wolfsberger S, Czech T, Budka I, Slave H, Hainfellner JA, 2005, Survivin expression in intracranial ependymomas and its correlation with tumor cell proliferation and patient outcome. Am J Clin Pathol. 124:543–549 Prayson RA, 1999, Clinicopathologic study of 61 patients with ependymoma including MIB-1 immunohistochemistry. Ann Diagn Pathol 3:11–18 Wolfsberger S, Fischer I, Hoftberger R, Birner P, Slavc I, Dieckmann K, Czech T, Budka H, Hainfellner J, 2004, Ki-67 immunolabeling index is an accurate predictor of outcome in patients with intracranial ependymoma. Am J Surg Pathol 28:914–920 Figarella-Branger D, Gambarelli D, Dollo C, Devictor B, Perez- Castillo AM, Genitori L, Lena G, 1991, Infratentorial ependymomas of childhood. Correlation between histological features, immunohistological phenotype, silver nucleolar organizer region staining values and post-operative survival in 16 cases. Acta Neuropathol [Berl] 82:208–216 Shuangshoti S, Rushing EJ, Mena H, Olsen C, Sandberg GD, 2005, Supratentorial extraventricular ependymal neoplasms: a clinicopathological study of 32 patients. Cancer 103:2598–2605 Onguru O, Kurt B, Gunhan O, Soylemezoglu F, 2006, Cyclooxigenase-2, cox-2, expression and angiogenesis in intracranial ependymomas. Clin Neuropath 25:06–20 Teo Ch, Nakaji P, Symons P, Tobias V, Cohn R, Smee R, 2003, Ependymoma. Childs Nerv Syst 19:270–285 Kaplan E, Meier P, 1958, Nonparametric estimation from incomplete observation. J Am Stat Assoc 53:457–481 Cox DR, 1972, Regression models and life-tables. J R Stat Soc B34:187–220 Casentini L, Gullotta F, Mohrer U, 1981, Clinical and morphological investigations on ependymomas and their tissue cultures. Neurochirurgia 24:51–58 Guyotat J, Signorelli F, Desme S, Frappaz D, Madarassy G, Montange MF, 2002, Intracranial ependymomas in adult patients: analyses of prognostic factors. J Neurooncol 60:255–268 Vanuytsel LJ, Bessell EM, Ashley SE, Bloom HJ, Brada M, 1992, Intracranial ependymoma: long-term results of a policy of surgery and radiotherapy. Int J Radiat Oncol Biol Phys 23:313– 319 Merchant TR, Jenkins JJ, Burger PC, Sanford RA, Sherwood SH, Jones-Wallace D, Heideman RL, Thompson SJ, Helton KJ, Kun LK, 2002, Influence of tumor grade on time to progression after irradiation for localized ependymoma in children. J Radiation Oncology Biol Phys 53:52–57 Rosenblum M, 1998, Ependymal tumors: a review of their diagnostic surgical pathology. Pediatr Neurosurg 28:160–165 Schwartz TH, Kim S, Glick RS, Bagiella E, Balmaceda C, Fetell MR, Stein BH, Sisti MB, Bruce JN, 1999, Supratentorial ependymomas in adult patients. Neurosurgery 44:721–731 Mork SJ, Loken AC, 1977, Ependymoma:a follow-up study of 101 cases. Cancer 40:907–915 Nazar GB, Hoffman HJ, Becker LE, Jenkin D, Humphreys RP, Hendrick EB, 1990, Infratentorial ependymomas in childhood: prognostic factors and treatment. J Neurosurg 72:408–417 Schroder R, Ploner C, Ernestus RI, 1993, The growth potential of ependymomas with varying grades of malignancy measured by the Ki-67 labelling index and mitotic index. Neurosurg Rev 16:145–150 Schulz MD CCW, Zinninger GF, Tefft M, 1968, Radiotherapy of intracranial neoplasms. With a special section on radiotherapeutic management of central nervous system tumors in children. In: Krayenbuhl HMP, Sweet WH, eds, Progress In Neurological Surgery, 2nd edn. Karger S, Basel New York, pp 319–370 Leibel SA, Sheline GE, 1987, Radiation therapy for neoplasms of the brain. J Neurosurg 66:1–22 Donahue B, Steinfeld A, 1998, Intracranial ependymoma in the adult parient:Succesful treatment with surgery and radiotherapy. J Neuro-Oncology 37:131–133 Salazar OM, Rubin P, Bassano D, Marcial VA, 1975, Improved survival of patients with intracranial ependymomas by irradiation: dose selection and field extension. Cancer 35:1563–1573 Paulino AC, Wen BC, 2000, The significance of radiotherapy treatment duration in intracranial ependymoma. Int J Radiat Oncol Biol Phys 47:585–589 Schuller P, Schafer U, Micke O, Willich N, 1999, Radiotherapy for intracranial and spinal ependymomas. A retrospective analysis. Strahlenther Onkol 175:105–111 Shaw EG, Evans RG, Scheithauer BW, Ilstrup DM, Earle JD, 1987, Postoperative radiotherapy of intracranial ependymoma in pediatric and adult patients. Int J Radiat Oncol Biol Phys 13:1457–1462 Wagner W, Schuller P, Willich N, Schober O, Palkovic S, Morgenroth C, Bartenstein P, Prott FJ, Niewohner U, 1995, Intraoperative radiotherapy [IORT] in malignant brain tumors. Strahlenther Onkol 171:154–164 Verstegen MJ, Bosch DA, Troost D, 1997, Treatment of ependymomas. Clinical and non-clinical factors influencing prognosis: a review. Br J Neurosurg 11:542–553 Ikezaki K, Matsushima T, Inoue T, Yokoyama N, Kaneko Y, Fukui M, 1993, Correlation of microanatomical localization with postoperative survival in posterior fossa ependymomas. Neurosurgery 32:38–44 Schild SE, Nisi K, Scheithauer BW, Wong WW, Lyons MK, Schomberg PJ et al, 1947, The results of radiotherapy for ependymomas: the Mayo Clinic experience. Int J Radiat Oncol Biol Phys 42:958 Pollack IF, Gerszten PC, Martinez AJ, Lo KH, Shultz B, Albright AL et al, 1995, Intracranial ependymomas of childhood: long-term outcome and prognostic factors. Neurosurgery 37:655–666 West CR, Bruse DA, Duffner PK, 1985, Ependymomas: factors in clinical and diagnostic staging. Cancer 56:1812–1816 Healey EA, Barnes PD, Kupsky WJ, Scott RM, Sallan SE, Black PM, Tarbell NJ, 1991, The prognostic significance of postoperative residual tumor in ependymoma. Neurosurgery 28:666– 671 Hukin J, Epstein F, Lefton D, Allen J, 1998, Treatment of intracranial ependymoma by surgery alone. Pediat Neurosurg 29:40–45 Robertson PI, Zeltzer PM, Boyett JM, Rorke LB, Allen JC, Geyer JR, Stanly P, Hao Li, Albright AL, McGuire-Cullen P, Finley JL, Stevensjr KL, Milstein JL, Packer RJ, Wisoff J, and the ChildrenCancer Group, 1998, Survival and prognostic factors following radiation therapy and chemotherapy for ependymomas in Chidren’s Cancer Group. J Neurosurg 88:695–703 Palma J, Celli P, Mariottini A, Zalaffi A, Schettini G, 2000, The importance of surgery in supratentorial ependymomas Long-term survival in a series of 23 cases. Child Nerv Syst 16:170–175 Kovalic JJ, Flaris N, Grigsby PW, Pirkowski M, Simpson JR, Roth KA, 1993, Intracranial ependymoma long term outcome, patterns of failure. J Neurooncol 15:125–131 Schroder R, Stutzer H, Klug N, 1996, Prognostic relevance of localization and grading in intracranial ependymomas of childhood. Childs Nerv Syst 12:522–526 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 93 EP 99 DI 10.1007/s12253-009-9194-5 PG 7 ER PT J AU Hazzaa, MS Elashry, MO Afifi, KI AF Hazzaa, M Sahar Elashry, M Osama Afifi, K Ibtesam TI Clusterin as a Diagnostic and Prognostic Marker for Transitional Cell Carcinoma of the Bladder SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; Tumor marker; Clusterin; Over-expression; Prognosis ID Bladder cancer; Tumor marker; Clusterin; Over-expression; Prognosis AB We investigated the feasibility of profiling and measuring the concentration of clusterin in urine and serum for individuals with transitional cell carcinoma (TCC) of the bladder and comparing it with nontumor controls. In addition, we analyzed the correlation of expression of clusterin in specimens of TCC to various clinicopathologic parameters and prognosis of bladder cancer. Blood and urine samples were used from 68 patients with TCC of the bladder and from 61 patients with benign urological diseases. Enzyme-linked immunosorbent assays (ELISA) were performed for clusterin from serum and urine. Quantitation of clusterin mRNA was carried out in 68 bladder tumor specimens from radical cystectomy or transurethral resection and 26 normal bladder specimens from BPH patients by using RT-PCR method. Correlation for the expression of clusterin mRNA with clinicopathologic parameters was analyzed. Serum and urine clusterin was significantly higher in individuals with bladder cancer than control (p=0.001). Sensitivity and specificity of serum and urine clusterin as a tumor marker for TCC of the bladder was found to be 80%, 91%, 87.1% and 96.7% respectively. Clusterin expression was significantly higher in TCC specimens than normal tissue specimens (P<0.001). Expression of clusterin was significantly higher in patients with invasive TCC of the bladder than that in patients with superficial TCC and control (P<0.001). Overexpression of clusterin mRNA was significantly associated with tumor recurrence and overall survival (p<0.001). The recurrence-free survival time of patients with overexpression of clusterin was significantly shorter than that of patients with weak expression of clusterin (9.8 months vs. 35.2 months). Clusterin may be considered as a potential diagnostic and prognostic biomarker for bladder cancer using urine, serum and/or molecular biology techniques. C1 [Hazzaa, M Sahar] Tanta University, Faculty of Medicine, Department of PathologyTanta, Egypt. [Elashry, M Osama] Tanta University, Urology DepartmentTanta, Egypt. [Afifi, K Ibtesam] Tanta University, Microbiology DepartmentTanta, Egypt. RP Elashry, MO (reprint author), Tanta University, Urology Department, Tanta, Egypt. EM oselashry@yahoo.com CR Irani J, Heidenreich A, Mottet N et al, 2008, What is new in bladder cancer diagnosis and management? Eur Urol supplements 7:484–493 Haitel A, Posch B, El-Baz M et al, 2001, Bilharzial related organ confined muscle-invasive bladder cancer: prognostic value of apoptosis markers, proliferation markers, p53, e-cadherin, epidermal growth factor receptor and c-erbB-2. J Urol 165:1481–1487 Millan-Rodriguez F, Chechile-Toniolo G, Salvador-Bayarri J et al, 2000, Primary superficial bladder cancer risk groups according to progression, mortality and recurrence. J Urol. 164:680–684 Cajulis RS, Haines GK 3rd, Frias-Hidvegi D et al, 1995, Cytology, flow cytometry, image analysis, and interphase cytogenetics by fluorescence in situ hybridization in the diagnosis of transitional cell carcinoma in bladder washes: a comparative study. Diagn. Cytopathol. 13(3):214–224 Schmitz-Drager BJ, Fradet Y, Grossman HB, 2008, Bladder cancer markers in patient management: the current perspective. World J Urol 26:1–3 Blaschuk O, Burdzy K, Fritz IB, 1983, Purification and characterization of a cell-aggregating factor, clusterin), the major glycoprotein in ram rete testis fluid. J Biol Chem. 258:7714–7720 Jenne DE, Tschopp J, 1992, Clusterin: the intriguing guises of a widely expressed glycoprotein. Trends Biochem Sci. 17:154–159 Jones SE, Jomary C, 2002, Clusterin. Int J Biochem Cell Biol. 34:427–431 Shannan B, Seifert M, Leskov K et al, 2006, Challenge and promise: roles for clusterin in pathogenesis, progression and therapy of cancer. Cell Death and Differentiation J. 13:12–19 Parczyk K, Pilarsky C, Rachel U et al, 1994, Gp80, Clusterin: TRPM-2, mRNA levels is enhanced in human renal clear cell carcinomas. J Cancer Res Clin Oncol 120:186–188 Steinberg J, Oyasu R, Lang S et al, 1997, Intracellular levels of SGP-2, Clusterin, correlate with tumor grade in prostate cancer. Clin Cancer Res 3:1707–1711 Redondo M, Villar E, Torres-Munoz J et al, 2000, Overexpression of Clusterin in human breast carcinoma. Am J Pathol 157: 393–399 Miyake H, Gleave M, Kamidono S et al, 2002, Overexpression of clusterin in transitional cell carcinoma of the bladder is related to disease progression and recurrence. Urology 50:150–154 Kruger S, Mahnken A, Kausch I et al, 2006, Value of clusterin immunoreactivity as a predictive factor in muscle-invasive urothelial bladder carcinoma. Urology 67(1):105–109 Epstein JI, Amin MB, Reuter VR et al, 1998, The World Health Organization/International Society of Urological Pathology consensus classification of urothelial, transitional cell, neoplasms of the urinary bladder. Am J Surg Pathol 22:1435–1448 Park J, Park J, Ju S et al, 2003, Clusterin mRNA expression in apoptotic and activated rat thymocytes. Cell Research 13(1):49–58 Petrie A, Sabin C, 2005, Medical Statistics at a Glance. 2nd ed., Blackwell Publishing Trougakso IP, So A, Jansen B et al, 2004, Silencing expression of the clusterin/apolipoprotein J gene in human cancer cells using small interfering RNA induces spontaneous apoptosis, reduced growth ability, and cell sensitization to genotoxic and oxidative stress. Cancer Res. 64:1834–1842 Gleave M, Miyake H, 2005, Use of antisense oligonucleotides targeting the cytoprotective gene, clusterin, to enhance androgenand chemo-sensitivity in prostate cancer. World J. Urol. 23:38–46 Wong P, Ulyanova T, Organisciak DT et al, 2001, Expression of multiple forms of clusterin during light-induced retinal degeneration. Curr Eye Res 23:157–165 Leskov KS, Klokov DY, Li J et al, 2003, Synthesis and functional analyses of nuclear clusterin, a cell death protein. J. Biol. Chem. 278:11590–11600 Pucci S, Bonanno E, Pichiorri F et al, 2004, Modulation of different clusterin isoforms in human colon tumorigenesis. Oncogene 23:2298–2304 Chen X, Halberg RB, Ehrhardt WM et al, 2003, Clusterin as a biomarker in murine and human intestinal neoplasia. Proc. Natl. Acad. Sci. USA 100:9530–9535 Saffer H, Wahed A, Rassidakis GZ et al, 2002, Clusterin expression in malignant lymphomas. Mod. Pathol. 15:1221–1223 Stejskal D, Fiala RR, 2006, Evaluation of serum and urine clusterin as a potential tumor marker for urinary bladder cancer. Neoplasma. 53(4):343–346 Miyake H, Hara I, Gleave ME et al, 2004, Protection of androgen dependent human prostate cancer cells from oxidative stressinduced DNA damage by overexpression of clusterin and its modulation by androgen. Prostate 61:318–323 Zellweger T, Kiyama S, Chi K et al, 2003, Overexpression of the cytoprotective protein clusterin decreases radiosensitivity in the human LNCaP prostate tumour model. BJU Int. 92:463–469 Scaltriti M, Brausi M, Amorosi A, 2004, Clusterin, SGP-2, APOJ, expression is downregulated in low- and high-grade human prostate cancer. Int. J. Cancer 108:23–30 July LV, Beraldi E, So A et al, 2004, Nucleotide-based therapies targeting clusterin chemosensitized human lung adenocarcinoma cells both in vitro and in vivo. Mol. Cancer Ther. 3:223–232 Zhang LY, Ying WT, Mao YS, 2003, Loss of clusterin both in serum and tissue correlates with the tumorigenesis of oesophageal squamous cell carcinoma via proteomic approaches. World J Gastroenterol 9:650–654 Xie MJ, Motoo Y, Su SB et al, 2002, Expression of clusterin in human pancreatic cancer. Pancreas 25:234–238 Miyake H, Hara I, Kamidono S et al, 2001, Synergistic chemosensitization and inhibition of tumor growth and metastasis by the antisense oligodeoxynucleotide targeting clusterin gene in a human bladder cancer model. Clin Cancer Res 7:4245–4252 Miyake H, Nelson C, Rennie PS et al, 2000, Acquisition of chemoresistant phenotype by overexpression of the antiapoptotic gene, testosterone-repressed prostate message-2, in prostate cancer xenograft models. Cancer Res 60:2547–2554 Miyake H, Chi KN, Gleave ME, 2000, Antisense TRPM-2 oligodeoxynucleotides chemosensitize human androgenindependent PC-3 prostate cancer cells both in vitro and in vivo. Clin Cancer Res 6:1655–1663 Miyake H, Hara I, Kamidono S et al, 2001, Novel therapeutic strategy for advanced prostate cancer using antisense oligodeoxynucleotides targeting antiapoptotic genes up-regulated after androgen withdrawal to delay androgen-independent progression and enhance chemosensitivity. Int J Urol 8:337–349 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 101 EP 109 DI 10.1007/s12253-009-9196-3 PG 9 ER PT J AU Zhang, LZ Mei, J Qian, ZK Cai, XS Jiang, Y Huang, WD AF Zhang, Li-Zhi Mei, Jiong Qian, Zhi-Kang Cai, Xuan-Song Jiang, Yao Huang, Wei-Da TI The Role of VE-cadherin in Osteosarcoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE VE-cadherin; Osteosarcoma cell; Vasculogenic mimicry; siRNA ID VE-cadherin; Osteosarcoma cell; Vasculogenic mimicry; siRNA AB Osteosarcoma cells can generate vasculogeniclike, patterned networks to obtain nutrients and oxygen, which mimic some function of endotheial-like cells and facilitate tumor malignant progress. These cells also express vascular endothelial-cadherin (VE-cadherin), which is generally accepted as a strictly endothelial-specific transmembrane protein. However, its role is still relatively obscure in osteosarcoma cells. So we inhibit the VE-cadherin gene expression with siRNA in osteosarcoma cells (MG63), and culture those cells in three-dimensional medium, containing Type I collagen or Matrigel, to observe the role of VE-cadherin. Western blotting analysis show that sequence-specific siRNA can significantly decrease the expression of VE-cadherin in MG63 cell. After knockdown of VE-cadherin, osteosacoma cells cann’t induced angiogenic sprout and form osteosarcoma-generated, endotheiallike networks. Our data indicate that VE-cadherin may be a positive and specific regulator not only in angiogenesis, but also in vasculogenic mimicry of osteosarcoma cells. And it can be considered as a new prospective option in the combining treatment of aggressive tumor with highly vascularity, including osteosarcoma. C1 [Zhang, Li-Zhi] Shanghai Jiaotong University, Shanghai Sixth People’s Hospital, Department of Orthopedic Surgery, No. 600 Yishan Road, 200233 Shanghai, China. [Mei, Jiong] Tongji Hospital Affiliated to Tongji University, Department of Orthopedic Surgery, 200065 Shanghai, China. [Qian, Zhi-Kang] Fudan University, Department of Biochemistry, 200433 Shanghai, China. [Cai, Xuan-Song] Tongji Hospital Affiliated to Tongji University, Department of Orthopedic Surgery, 200065 Shanghai, China. [Jiang, Yao] Shanghai Jiaotong University, Shanghai Sixth People’s Hospital, Department of Orthopedic Surgery, No. 600 Yishan Road, 200233 Shanghai, China. [Huang, Wei-Da] Fudan University, Department of Biochemistry, 200433 Shanghai, China. RP Jiang, Y (reprint author), Shanghai Jiaotong University, Shanghai Sixth People’s Hospital, Department of Orthopedic Surgery, 200233 Shanghai, China. EM jiangyao2005@163.com CR Maniotis AJ, Folberg R, Hess A et al, 1999, Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 155:739–752 Lissitzky JC, Parriaux D, Ristorcelli E et al, 2009, Cyclic AMP signaling as a mediator of vasculogenic mimicry in aggressive human melanoma cells in vitro. Cancer Res 69:802– 809 Stevens AP, Spangler B, Wallner S et al, 2009, Direct and tumor microenvironment mediated influences of 5′-deoxy-5′-(methylthio, adenosine on tumor progression of malignant melanoma. J Cell Biochem 106:210–219 Su F, Li B, Wang J, 2009, Molecular regulation of vasculogenic mimicry in human uveal melanoma cells: role of helix-loop-helix Id2, inhibitor of DNA binding 2). Graefes Arch Clin Exp Ophthalmol 247:411–419 Hendrix MJ, Seftor EA, Meltzer PS, 2001, Expression and functional significance of VE-cadherin in aggressive human melanoma cells: role in vasculogenic mimicry. Proc Natl Acad Sci U S A 98:8018–8023 Gavard J, 2009, Breaking the VE-cadherin bonds. FEBS Lett 583:1–6 Vestweber D, Winderlich M, Cagna G et al, 2009, Cell adhesion dynamics at endothelial junctions: VE-cadherin as a major player. Trends Cell Biol 19:8–15 Vestweber D, 2008, VE-cadherin: the major endothelial adhesion molecule controlling cellular junctions and blood vessel formation. Arterioscler Thromb Vasc Biol 28:223–232 Dome B, Hendrix MJ, Paku S, 2007, Alternative vascularization mechanisms in cancer: Pathology and therapeutic implications. Am J Pathol 170:1–15 Monzani E, La Porta CA, 2008, Targeting cancer stem cells to modulate alternative vascularization mechanisms. Stem Cell Rev 4:51–56 Shen R, Ye Y, Chen L, Yan Q, 2008, Precancerous stem cells can serve as tumor vasculogenic progenitors. PLoS ONE 3:e1652 Nikolova-Krstevski V, Bhasin M, Otu HH, 2008, Gene expression analysis of embryonic stem cells expressing VE-cadherin, CD144)during endothelial differentiation. BMC Genomics 9:240 Dejana E, Orsenigo F, Lampugnani MG, 2008, The role of adherens junctions and VE-cadherin in the control of vascular permeability. J Cell Sci 121:2115–2122 Wallez Y, Vilgrain I, Huber P, 2006, Angiogenesis: the VE-cadherin switch. Trends Cardiovasc Med 16:55–59 Taddei A, Giampietro C, Conti A et al, 2008, Endothelial adherens junctions control tight junctions by VE-cadherinmediated upregulation of claudin-5. Nat Cell Biol 10:923–934 Taveau JC, Dubois M, Le Bihan O et al, 2008, Structure of artificial and natural VE-cadherin-based adherens junctions. Biochem Soc Trans 36:189–193 Boda-Heggemann J, Regnier-Vigouroux A, Franke WW, 2009, Beyond vessels: occurrence and regional clustering of vascular endothelial(VE-)cadherin-containing junctions in non-endothelial cells. Cell Tissue Res 335:49–65 Hess AR, Seftor EA, Gruman LM et al, 2006, VE-cadherin regulates EphA2 in aggressive melanoma cells through a novel signaling pathway: implications for vasculogenic mimicry. Cancer Biol Ther 5:228–233 Pezzolo A, Parodi F, Corrias MV et al, 2007, Tumor origin of endothelial cells in human neuroblastoma. J Clin Oncol 25:376–383 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 111 EP 117 DI 10.1007/s12253-009-9198-1 PG 7 ER PT J AU Warth, A Muley, Th Herpel, E Pfannschmidt, J Hoffmann, H Dienemann, H Schirmacher, P Schnabel, AP AF Warth, Arne Muley, Thomas Herpel, Esther Pfannschmidt, Joachim Hoffmann, Hans Dienemann, Hendrik Schirmacher, Peter Schnabel, A Philipp TI A Histochemical Approach to the Diagnosis of Visceral Pleural Infiltration by Non-small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Visceral pleural infiltration (VPI); Staging; Pathology; Elastic layers; Lung cancer; NSCLC; Pleura; Infiltration; Elastic layer; Staining ID Visceral pleural infiltration (VPI); Staging; Pathology; Elastic layers; Lung cancer; NSCLC; Pleura; Infiltration; Elastic layer; Staining AB Introduction: Although invasion of the visceral pleura (VPI) by non-small cell lung cancer (NSCLC) is a TNM-relevant diagnostic criterion and is known to affect the patients´ prognoses, until recently there were no standardized or internationally accepted guidelines. This resulted in a diagnostic ambiguity leading to different tumor staging systems and to hardly comparable patient collectives in research studies world wide. The major problem in this issue is to exactly define what constitutes for the diagnosis of VPI with respect to anatomical landmarks. Methods: In order to address this problem we investigated the pleural infiltration depth of 173 NSCLC specimens without lymph node metastases and proven tumor-related death using elastic stains and a scoring system referring to prominent pleural elastic layers, the lamina elastica externa and interna, as anatomical landmarks. Results: Performing comparative Kaplan-Meier survival analyses for each patient collective we could not find any significant difference in the patients´ survival. This indicates that a differential evaluation of the tumor infiltration depth according to the elastic layers is not practicable. Conclusions: Our findings support the consequent application of the recently proposed, pragmatic approach of the international staging committee for lung cancer (IASLC) to define an internationally accepted and standardized staging system for VPI. C1 [Warth, Arne] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. [Muley, Thomas] Thoraxklinik Heidelberg, Translational Research UnitHeidelberg, Germany. [Herpel, Esther] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. [Pfannschmidt, Joachim] Thoraxklinik Heidelberg, Department of Thoracic SurgeryHeidelberg, Germany. [Hoffmann, Hans] Thoraxklinik Heidelberg, Department of Thoracic SurgeryHeidelberg, Germany. [Dienemann, Hendrik] Thoraxklinik Heidelberg, Department of Thoracic SurgeryHeidelberg, Germany. [Schirmacher, Peter] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. [Schnabel, A Philipp] University of Heidelberg, Department of Pathology, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany. RP Warth, A (reprint author), University of Heidelberg, Department of Pathology, 69120 Heidelberg, Germany. EM arne.warth@med.uni-heidelberg.de CR World Health Organization classification of tumors, 2004, Pathology and genetics. Tumours of the lung, pleura, thymus and heart. IARC, Lyon Osaki T, Nagashima A, Yoshimatsu T et al, 2004, Visceral pleural involvement in nonsmall cell lung cancer: prognostic significance. Ann Thorac Surg 77:1769–1773 Khan OA, Fitzgerald JJ, Field ML et al, 2004, Histological determinants of survival in completely resected T1–2N1M0 nonsmall cell cancer of the lung. Ann Thorac Surg 77:1173–1178 Gallagher B, Urbanski SJ, 1990, The significance of pleural elastica invasion by lung carcinomas. Hum Pathol 21:512–517 Ichinose Y, Yano T, Asoh H et al, 1995, Prognostic factors obtained by a pathologic examination in completely resected nonsmall- cell lung cancer. An analysis in each pathologic stage. J Thorac Cardiovasc Surg 110:601–605 Manac’h D, Riquet M, Medioni J et al, 2001, Visceral pleura invasion by non-small cell lung cancer: an underrated bad prognostic factor. Ann Thorac Surg 71:1088–1093 Shimizu K, Yoshida J, Nagai K et al, 2005, Visceral pleural invasion is an invasive and aggressive indicator of non-small cell lung cancer. J Thorac Cardiovasc Surg 130:160–165 Shimizu K, Yoshida J, Nagai K et al, 2004, Visceral pleural invasion classification in non-small cell lung cancer: a proposal on the basis of outcome assessment. J Thorac Cardiovasc Surg 127:1574–1578 Butnor KJ, Vollmer RT, Blaszyk H et al, 2007, Interobserver agreement on what constitutes visceral pleural invasion by nonsmall cell lung carcinoma: an internet-based assessment of international current practices. Am J Clin Pathol 128:638–647 Butnor KJ, Cooper K, 2005, Visceral pleural invasion in lung cancer: recognizing histologic parameters that impact staging and prognosis. Adv Anat Pathol 12:1–6 Japan Lung Cancer Society, 1999, General rule for clinical and pathological record of lung cancer, 5th edn. Kanehara, Tokyo Antony VB, Sahn SA, Mossman B et al, 1992, NHLBI workshop summaries. Pleural cell biology in health and disease. Am Rev Respir Dis 145:1236–1239 Bunker ML, Raab SS, Landreneau RJ et al, 1999, The diagnosis and significance of visceral pleural invasion in lung carcinoma. Histologic predictors and the role of elastic stains. Am J Clin Pathol 112:777–783 Ichinose Y, Hara N, Ohta M et al, 1993, Is T factor of the TNM staging system a predominant prognostic factor in pathologic stage I non-small-cell lung cancer ? A multivariate prognostic factor analysis of 151 patients. J Thorac Cardiovasc Surg 106:90– 94 Kang JH, Kim KD, Chung KY, 2003, Prognostic value of visceral pleura invasion in non-small cell lung cancer. Eur J Cardiothorac Surg 23:865–869 Hammar S, 1994, Common neoplasms. In: Dail DH, Hammar S, eds, Pulmonary pathology. Springer-Verlag, New York, p 1138 Travis WD, Brambilla E, Rami-Porta R et al, 2008, Visceral pleural invasion: pathologic criteria and use of elastic stains: proposal for the 7th edition of the TNM classification for lung cancer. J Thorac Oncol 3:1384–1390 Shim HS, Park IK, Lee CY et al, 2009, Prognostic significance of visceral pleural invasion in the forthcoming, seventh, edition of TNM classification for lung cancer. Lung Cancer, DOI 10.1016/j. lungcan.2008.11.008 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 119 EP 123 DI 10.1007/s12253-009-9201-x PG 5 ER PT J AU Strenger, V Urban, Ch AF Strenger, Volker Urban, Christian TI Chromosomal Integration of the HHV-6 Genome as a Possible Cause of Persistent HHV-6 Detection in a Patient with Langerhans Cell Histiocytosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Strenger, Volker] Medical University of Graz, Department of Paediatrics and Adolescent Medicine, Auenbruggerplatz 30, 8036 Graz, Austria. [Urban, Christian] Medical University of Graz, Department of Paediatrics and Adolescent Medicine, Auenbruggerplatz 30, 8036 Graz, Austria. RP Strenger, V (reprint author), Medical University of Graz, Department of Paediatrics and Adolescent Medicine, 8036 Graz, Austria. EM volker.strenger@medunigraz.at CR Csire M, Mikala G, Jako J et al, 2007, Persistent long-term human herpesvirus 6, HHV-6, infection in a patient with langerhans cell histiocytosis. Pathol Oncol Res 13:157–160 Boutolleau D, Agut H, Gautheret-Dejean A, 2006, Human herpesvirus 6 genome integration: a possible cause of misdiagnosis of active viral infection? J Infect Dis 194:1019–1020 author reply 1021–1013 Caserta MT, Hall CB, Schnabel K, Lofthus G, McDermott MP, 2007, Human herpesvirus, HHV)-6 and HHV-7 infections in pregnant women. J Infect Dis 196:1296–1303 Daibata M, Taguchi T, Nemoto Y, Taguchi H, Miyoshi I, 1999, Inheritance of chromosomally integrated human herpesvirus 6 DNA. Blood 94:1545–1549 Hubacek P, Maalouf J, Zajickova M et al, 2007, Failure of multiple antivirals to affect high HHV-6 DNAaemia resulting from viral chromosomal integration in case of severe aplastic anaemia. Haematologica 92:e98–e100 Luppi M, Barozzi P, Morris CM, Merelli E, Torelli G, 1998, Integration of human herpesvirus 6 genome in human chromosomes. Lancet 352:1707–1708 Luppi M, Marasca R, Barozzi P et al, 1993, Three cases of human herpesvirus-6 latent infection: integration of viral genome in peripheral blood mononuclear cell DNA. J Med Virol 40:44–52 Tanaka-Taya K, Sashihara J, Kurahashi H et al, 2004, Human herpesvirus 6, HHV-6, is transmitted from parent to child in an integrated form and characterization of cases with chromosomally integrated HHV-6 DNA. J Med Virol 73:465–473 Torelli G, Barozzi P, Marasca R et al, 1995, Targeted integration of human herpesvirus 6 in the p arm of chromosome 17 of human peripheral blood mononuclear cells in vivo. J Med Virol 46:178– 188 Ward KN, Leong HN, Nacheva EP et al, 2006, Human herpesvirus 6 chromosomal integration in immunocompetent patients results in high levels of viral DNA in blood, sera, and hair follicles. J Clin Microbiol 44:1571–1574 Ward KN, Leong HN, Thiruchelvam AD, Atkinson CE, Clark DA, 2007, Human herpesvirus 6 DNA levels in cerebrospinal fluid due to primary infection differ from those due to chromosomal viral integration and have implications for diagnosis of encephalitis. J Clin Microbiol 45:1298–1304 Ward KN, Thiruchelvam AD, Couto-Parada X, 2005, Unexpected occasional persistence of high levels of HHV-6 DNA in sera: detection of variants A and B. J Med Virol 76:563–570 Leong HN, Tuke PW, Tedder RS et al, 2007, The prevalence of chromosomally integrated human herpesvirus 6 genomes in the blood of UK blood donors. J Med Virol 79:45–51 Hall CB, Caserta MT, Schnabel K et al, 2008, Chromosomal integration of human herpesvirus 6 is the major mode of congenital human herpesvirus 6 infection. Pediatrics 122:513– 520 Hubacek P, Muzikova K, Hrdlickova A et al, 2009, Prevalence of HHV-6 integrated chromosomally among children treated for acute lymphoblastic or myeloid leukemia in the Czech Republic. J Med Virol 81:258–263 Clark DA, Nacheva EP, Leong HN et al, 2006, Transmission of integrated human herpesvirus 6 through stem cell transplantation: implications for laboratory diagnosis. J Infect Dis 193:912– 916 Kamble RT, Clark DA, Leong HN, Heslop HE, Brenner MK, Carrum G, 2007, Transmission of integrated human herpesvirus-6 in allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 40:563–566 Clark DA, Tsao EH, Leong HN, Ward KN, Nacheva EP, Griffiths PD, 2006, Reply to Boutolleau et al. and Luppi et al. J Infect Dis 194:1019–1020 author reply 1021–1013 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 125 EP 126 DI 10.1007/s12253-009-9197-2 PG 2 ER PT J AU Yu, L Yang, JSh AF Yu, Lu Yang, Jing Shou TI A Case of Primary Histiocytic Sarcoma Arising from Thyroid Gland SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Histiocytic sarcoma; Thyroid gland; Differential diagnosis; Gene rearrangements; CD163; Immunohistochemistry ID Histiocytic sarcoma; Thyroid gland; Differential diagnosis; Gene rearrangements; CD163; Immunohistochemistry AB Histiocytic sarcoma (HS) is an extremely rare true histiocytic malignancy. We report a case of HS arising from thyroid gland in a 69 year-old man. Following subtotal thyroidectomy, a histopathologic, immunohistologic, and genotypic examination revealed HS. This tumor was composed of large spindle or round epithelioid cells with abundant eosinophilic cytoplasm. The neoplastic cells were positive for macrophage-associated antigen CD68, CD163, and lysozymes, as well as CD45, HLA- DR, DP, DQ, and S100, most consistent with a diagnosis of HS. The BIOMED-2 multiplex PCR analysis showed polyclonal Band T-cell populations. To our knowledge, this is the first report of a rare entity HS involving thyroid gland using a comprehensive immunophenotyping panel including CD163 as well as molecular studies to establish the true histiocytic nature of these lesions. C1 [Yu, Lu] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, No. 17 Chang Le Xi Road, 710032 Xi’an, Shaanxi, China. [Yang, Jing Shou] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, No. 17 Chang Le Xi Road, 710032 Xi’an, Shaanxi, China. RP Yang, JSh (reprint author), Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. EM yangsj@fmmu.edu.cn CR Jaffe ES, 2001, Pathology and genetics of tumours of haematopoietic and lymphoid tissues, Vol. 3. IARC Press, Lyon, France Alexiev BA, Sailey CJ, McClure SA, Ord RA, Zhao X, Papadimitriou JC, 2007, Primary histiocytic sarcoma arising in the head and neck with predominant spindle cell component. Diagn Pathol 2:7 De Vos FY, Gerding MN, Arends JW, Wegman JJ, 2008, Histiocytic sarcoma localised in the thyroid: a case report. Ann Hematol 87:681–682 Weiss LM, Grogan TM, Muller-Hermelink HK, 2001, Histiocytic sarcoma. In: Jaffe ES, Harris NL, Stein H, Vardiman JW, eds, WHO classification of tumors: pathology and genetics of tumors of haematopoietic and lymphoid tissues. IARC Press, Lyon, France, pp 278–279 Nguyen TT, Schwartz EJ, West RB, Warnke RA, Arber DA, Natkunam Y, 2005, Expression of CD163, hemoglobin scavenger receptor, in normal tissues, lymphomas, carcinomas, and sarcomas is largely restricted to the monocyte/macrophage lineage. Am J Surg Pathol 29:617–624 Vos JA, Abbondanzo SL, Barekman CL, Andriko JW, Miettinen M, Aguilera NS, 2005, Histiocytic sarcoma: a study of five cases including the histiocyte marker CD163. Mod Pathol 18:693–704 Krafft AE, Taubenberger JK, Sheng ZM, Bijwaard KE, Abbondanzo SL, Aguilera NS, Lichy JH, 1999, Enhanced sensitivity with a novel TCRgamma PCR assay for clonality studies in 569 formalin-fixed, paraffin-embedded, FFPE, cases. Mol Diagn 4:119–133 Pileri SA, Grogan TM, Harris NL, Banks P, Campo E, Chan JK, Favera RD, Delsol G, De Wolf-Peeters C, Falini B, Gascoyne RD, Gaulard P, Gatter KC, Isaacson PG, Jaffe ES, Kluin P, Knowles DM, Mason DY, Mori S, Muller-Hermelink HK, Piris MA, Ralfkiaer E, Stein H, Su IJ, Warnke RA, Weiss LM, 2002, Tumours of histiocytes and accessory dendritic cells: an immunohistochemical approach to classification from the International Lymphoma Study Group based on 61 cases. Histopathology 41:1– 29 Sun W, Nordberg ML, Fowler MR, 2003, Histiocytic sarcoma involving the central nervous system: clinical, immunohistochemical, and molecular genetic studies of a case with review of the literature. Am J Surg Pathol 27:258–265 Boisseau-Garsaud AM, Vergier B, Beylot-Barry M, Nastasel- Menini F, Dubus P, de Mascarel A, Eghbali H, Beylot C, 1996, Histiocytic sarcoma that mimics benign histiocytosis. J Cutan Pathol 23:275–282 Cao M, Eshoa C, Schultz C, Black J, Zu Y, Chang CC, 2007, Primary central nervous system histiocytic sarcoma with relapse to mediastinum: a case report and review of the literature. Arch Pathol Lab Med 131:301–305 Copie-Bergman C, Wotherspoon AC, Norton AJ, Diss TC, Isaacson PG, 1998, True histiocytic lymphoma: a morphologic, immunohistochemical, and molecular genetic study of 13 cases. Am J Surg Pathol 22:1386–1392 Kamel OW, Gocke CD, Kell DL, Cleary ML, Warnke RA, 1995, True histiocytic lymphoma: a study of 12 cases based on current definition. Leuk Lymphoma 18:81–86 Ohyashiki JH, Ohyashiki K, Toyama K, Kimura N, Minowada J, Kinniburgh AJ, Sandberg AA, 1989, T-cell receptor gene rearrangement and its expression in human myeloid leukemia cell lines. Cancer Genet Cytogenet 37:193–200 Bouabdallah R, Abena P, Chetaille B, Aurran-Schleinitz T, Sainty D, Dubus P, Arnoulet C, Coso D, Xerri L, Gastaut JA, 2001, True histiocytic lymphoma following B-acute lymphoblastic leukaemia: case report with evidence for a common clonal origin in both neoplasms. Br J Haematol 113:1047–1050 Feldman AL, Minniti C, Santi M, Downing JR, Raffeld M, Jaffe ES, 2004, Histiocytic sarcoma after acute lymphoblastic leukaemia: a common clonal origin. Lancet Oncol 5:248–250 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 127 EP 132 DI 10.1007/s12253-009-9193-6 PG 6 ER PT J AU Meng, W Zhou, Y Zhang, H Jiang, L Wang, Z Li, X Zhou, H Chen, Q Zeng, X AF Meng, Wenxia Zhou, Yu Zhang, Hengyu Jiang, Lu Wang, Zhi Li, Xiaoying Zhou, Hongmei Chen, Qianming Zeng, Xin TI Nasal-type NK/T-cell Lymphoma with Palatal Ulcer as the Earliest Clinical Manifestation: A Case Report with Literature Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Epstein-Barr virus; Granzyme B; Immunohistochemistry; NK/T-cell lymphoma; Ulcer ID Epstein-Barr virus; Granzyme B; Immunohistochemistry; NK/T-cell lymphoma; Ulcer AB Extranodal nasal natural killer (NK)/T-cell lymphoma is a very rare kind of lymphoma, Oral cavity involvement of extranodal natural killer/T-cell lymphoma, nasal type is extremely rare, and its clinicopathologic features are also poorly understood. Recently, we experienced an unusual case of Epstein-Barr virus-associated, extranodal NK/T-cell type with a unhealed palatal ulcer as the earliest clinical feature. It is a challenge for oral medicine specialists to make the early diagnosis for this special type of tumor. C1 [Meng, Wenxia] Sichuan University, West China College of Stomatology, State Key Laboratory of Oral Diseases, 14 RenMinNanLu, Section 3, Chengdu, 610041 Sichuan, China. [Zhou, Yu] Sichuan University, West China College of Stomatology, State Key Laboratory of Oral Diseases, 14 RenMinNanLu, Section 3, Chengdu, 610041 Sichuan, China. [Zhang, Hengyu] Sichuan University, West China Hospital, Department of CardiologySichuan, China. [Jiang, Lu] Sichuan University, West China College of Stomatology, State Key Laboratory of Oral Diseases, 14 RenMinNanLu, Section 3, Chengdu, 610041 Sichuan, China. [Wang, Zhi] Sichuan University, West China College of Stomatology, State Key Laboratory of Oral Diseases, 14 RenMinNanLu, Section 3, Chengdu, 610041 Sichuan, China. [Li, Xiaoying] Sichuan University, West China College of Stomatology, Department of Oral MedicineSichuan, China. [Zhou, Hongmei] Sichuan University, West China College of Stomatology, State Key Laboratory of Oral Diseases, 14 RenMinNanLu, Section 3, Chengdu, 610041 Sichuan, China. [Chen, Qianming] Sichuan University, West China College of Stomatology, State Key Laboratory of Oral Diseases, 14 RenMinNanLu, Section 3, Chengdu, 610041 Sichuan, China. [Zeng, Xin] Sichuan University, West China College of Stomatology, State Key Laboratory of Oral Diseases, 14 RenMinNanLu, Section 3, Chengdu, 610041 Sichuan, China. RP Zeng, X (reprint author), Sichuan University, West China College of Stomatology, State Key Laboratory of Oral Diseases, 610041 Sichuan, China. EM zengxin22@163.com CR Harris NL, Jaffe ES, Diebold J et al, 1999, World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting-Airlie House, Virginia November 1997. J Clin Oncol 17:3835–3849 Ng SB, Lai KW, Murugaya S et al, 2004, Nasal-type extranodal natural killer/Tcell lymphomas: a clinicopathologic and genotypic study of 42 cases in Singapore. Mod Pathol 17:1097–1107 Jaffe ES, Harris NL, Stein H et al, 2001, WHO classification of tumors: pathology and genetics of tumors of haematopoietic and lymphoid tissues. IARC, Lyon Rostami S, Sun NCJ, Baker J, 2006, A 30-year-old man presenting with nausea, vomiting, and abdominal pain. Arch Pathol Lab Med 130:e84–86 Zhang YC, Zhao S, Yu JB, 2008, Gastric involvement of extranodal NK/T-cell lymphoma, nasal type: a report of 3 cases with literature review. Int J Surg Pathol 16:450–4 Mraz-Gernhard S, Natkunam Y, Hoppe R, 2001, Natural killer/ natural killer-like T-cell lymphoma, CD56+, presenting in the skin: an increasingly recognized entity with an aggressive course. J Clin Oncol 19:2179–2188 Yang C, Ma J, Yang X, 2008, Natural killer/T-cell nasal-type lymphoma: unusual primary spinal tumor. Spine 33:E929–32 Momose A, Mizuno H, Kajihara S, 2006, EBV-associated nasaltype NK/T-cell lymphoma of the nasal cavity/paranasal sinus in a renal allograft recipient. Nephrol Dial Transplant 21:1413–1416 Lanier LL, Chang C, Philips JH et al, 1991, Molecular and functional analysis of human natural killer cell-associated neural cell adhesion molecule, N-CAM/CD56). J Immunol 146:4421–4426 Went P, Agostinelli C, Gallamini A et al, 2006, Marker expression in peripheral T-cell lymphoma: a proposed clinicalpathologic prognostic score. J Clin Oncol 24:2472–2479 Kim SJ, Kim SJ, Kim BS, Choi CWet al, 2007, Ki-67 expression is predictive of prognosis in patients with stage I/II extranodal NK/T-cell lymphoma, nasal type. Ann Oncol 18:1382–1387 Tao J, Wasik MA, 2001, Epstein-Barr virus associated polymorphic lymphoproliferative disorders occurring in nontransplant settings. Lab Invest 81:429–37 Hoshida Y, Li T, Dong Z et al, 2001, Lymphoproliferative disorders in renal transplant patients in Japan. Int J Cancer 91:869–875 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 133 EP 137 DI 10.1007/s12253-009-9183-8 PG 5 ER PT J AU Dundr, P Pesl, M Povysil, C Bauerova, L Soukup, V AF Dundr, Pavel Pesl, Michael Povysil, Ctibor Bauerova, Lenka Soukup, Viktor TI Primary Large Cell Neuroendocrine Carcinoma of the Kidney SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Carcinoma; Kidney; Large cell; Neuroendocrine; Renal tumors ID Carcinoma; Kidney; Large cell; Neuroendocrine; Renal tumors AB We report a case of a 56-year-old male with a primary large cell neuroendocrine renal carcinoma. Grossly, the left kidney was enlarged by a solid tumor that measured 145×125×100 mm. Histologically, the tumor consisted of large cells with a moderate to abundant amount of eosinophilic cytoplasm. The nuclei were irregular, some of them with finely or coarsely granular chromatin, others with vesicular chromatin and prominent nucleoli. The tumor cells showed multiple mitotic figures (up to 32 mitoses/10 HPF). In some areas, the tumor cells were arranged in solid sheets; however, the predominant pattern was solid-alveolar, trabecular and cribriform. Large areas of tumor necrosis were found. Immunohistochemically, the tumor cells were positive for synaptophysin, CD56 and CD57. Cytokeratin AE1/AE3, vimentin and CD10 were positive only focally. Chromogranin showed weak cytoplasmic positivity in rare tumor cells. Cytokeratin CAM5.2, cytokeratin 34βE12, BerEP 4, EMA, TTF-1, cytokeratin 7, cytokeratin 20, calretinin, serotonin, somatostatin, gastrin, calcitonin, glukagon and insulin were negative. Primary large cell neuroendocrine carcinoma of the kidney is a rare tumor. To the best of our knowledge, only 3 cases of a tumor of this type have been reported to date. C1 [Dundr, Pavel] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 128 00 Prague, Czech Republic. [Pesl, Michael] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Urology, Ke Karlovu 6, 128 00 Prague, Czech Republic. [Povysil, Ctibor] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 128 00 Prague, Czech Republic. [Bauerova, Lenka] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 128 00 Prague, Czech Republic. [Soukup, Viktor] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Urology, Ke Karlovu 6, 128 00 Prague, Czech Republic. RP Dundr, P (reprint author), Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, 128 00 Prague, Czech Republic. EM pdundr@seznam.cz CR Lane BR, Chery F, Jour G et al, 2007, Renal neuroendocrine tumours: a clinicopathological study. BJU Int 100:1030–1035 Mazzucchelli R, Morichetti D, Lopez-Beltran A et al, 2009, Neuroendocrine tumours of the urinary system and male genital organs: clinical significance. BJU Int. Feb 27. [Epub ahead of print] Lagace R, Tremblay M, 1968, Non-chromaffin paraganglioma of the kidney with distant metastases. Can Med Assoc J 99:1095–1098 Sellaturay SV, Arya M, Banisadr S et al, 2006, Primary intrarenal neuroblastoma: A rare, aggressive tumour of childhood mimicking Wilms’ tumour. J Pediatr Urol 2:522–524 Guillou L, Duvoisin B, Chobaz C et al, 1993, Combined smallcell and transitional cell carcinoma of the renal pelvis. A light microscopic, immunohistochemical, and ultrastructural study of a case with literature review. Arch Pathol Lab Med 117:239–243 La Rosa S, Bernasconi B, Micello D et al, 2008, Primary Small Cell Neuroendocrine Carcinoma of the Kidney: Morphological, Immunohistochemical, Ultrastructural, and Cytogenetic Study of a Case and Review of the Literature. Endocr Pathol 20:24–34 Moukassa D, Leroy X, Bouchind’homme B et al, 2000, Primary large cell neuroendocrine carcinoma of the kidney: morphologic and immunohistochemical features of two cases. Ann Pathol 20:357–360 Parada DD, Pena KGB, 2008, Chromophobe renal cell carcinoma with neuroendocrine differentiation. APMIS 116:859–865 Kuroda N, Hes O, Miyazaki E et al, 2006, Frequent expression of neuroendocrine markers in mucinous tubular and spindle cell carcinoma of the kidney. Histol Histopathol 21:7–10 Kuroda N, Nakamura S, Miyazaki E et al, 2004, Low-grade tubular-mucinous renal neoplasm with neuroendocrine differentiation: a histological, immunohistochemical and ultrastructural study. Pathol Int 54:201–207 Armah HB, Parwani AV, 2007, Primary carcinoid tumor arising within mature teratoma of the kidney: report of rare entity and review of the literature. Diagn Pathol 2:15 Boix Orri R, Mora Durban MJ, Sanchez Macias J et al, 2008, Neuroendocrine tumor in a horseshoe kidney: relative risk of association of two related entities. Arch Esp Urol 61:819–822 Romero FR, Rais-Bahrami S, Permpongkosol S et al, 2006, Primary carcinoid tumors of the kidney. J Urol 176:2359–2366 Hansel DE, Epstein JI, Berbescu E et al, 2007, Renal carcinoid tumor: a clinicopathologic study of 21 cases. Am J Surg Pathol 31:1539–1544 Chung CH, Park CY, 2006, Small cell carcinoma of the kidney. Korean J Intern Med 21:191–193 Capella C, Eusebi V, Rosai J, 1984, Primary oat cell carcinoma of the kidney. Am J Surg Pathol 8:855–861 Gonzalez-Lois C, Madero S, Redondo P et al, 2001, Small cell carcinoma of the kidney: a case report and review of the literature. Arch Pathol Lab Med 125:796–798 Kilicarsalan Akkaya B, Mustafa U, Esin O et al, 2003, Primary small cell carcinoma of the kidney. Urol Oncol 21:11–13 Kitamura M, Miyanaga T, Hamada M et al, 1997, Small cell carcinoma of the kidney: case report. Int J Urol 4:422–424 Masuda T, Oikawa H, Yashima A et al, 1998, Renal small cell carcinoma, neuroendocrine carcinoma, without features of transitional cell carcinoma. Pathol Int 48:412–415 Morgan KG, Banerjee SS, Eyden BP, 1996, Primary small cell neuroendocrine carcinoma of the kidney. Ultrastruct Pathol 20:141–144 Tetu B, Ro JY, Ayala AG et al, 1987, Small cell carcinoma of the kidney. Cancer 60:1809–1814 Pollheimer VS, Bodo K, Pollheimer MJ et al, 2007, Merkel cell carcinoma metastasizing to the kidney mimicking primary neuroendocrine renal cancer. APMIS 115:774–777 Ulamec M, Tomas D, Peric-Balja M et al, 2008, Neuroendocrine breast carcinoma metastatic to renal cell carcinoma and ipsilateral adrenal gland. Pathol Res Pract 204:851–855 Ricketts R, Tamboli P, Czerniak B et al, 2008, Tumor-totumor metastasis: report of 2 cases of metastatic carcinoma to angiomyolipoma of the kidney. Arch Pathol Lab Med 132: 1016–1020 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2010 VL 16 IS 1 BP 139 EP 142 DI 10.1007/s12253-009-9180-y PG 4 ER PT J AU Kopper, L AF Kopper, Laszlo TI Panitumumab: An Arrow on Target SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Panitumumab; Colorectal cancer; Anti-EGFR therapy ID Panitumumab; Colorectal cancer; Anti-EGFR therapy AB Several options are available today in the treatment of advanced colorectal cancer: traditional chemotherapeutic regimens, targeted therapies, and their combinations. Panitumumab is a new, fully human anti-EGFR monoclonal antibody, what is well-tolerated, effective as a single agent in chemotherapy refractory patients and in different combinations. The clinical response is restricted to tumors with wild-type RAS, therefore the RAS status should be checked before treatment. C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM kopper@korb1.sote.hu CR Foon KA, Yang XD, Weiner LM et al, 2004, Preclinical and clinical evaluation of ABX-EGF, a fully human anti-epidermal growth factor receptor antibody. Int J Radiat Oncol Biol Phys 58:984–990 Jakobovits A, Amado RG, Yang X et al, 2007, From XenoMouse technology to panitumumab, the first fully human antibody product from transgenic mice. Nat Biotechnol 25:1134–1143 Freeman D, McDorman K, Bussh T et al, 2004, Mono and combination-therapeutic activity of panitumumab, ABX-EGF, on human A431 epidermoid and HT-29 colon carcinoma xenografts: correlation with pharmacodynamic parameters. Proc Am Assoc Cancer Res 27–31 Lopez-Albaitero A, Ferris RL, 2007, Immune activation by epidermal growth factor receptor specific monoclonal antibody therapy for head and neck cancer. Arch Otolaryngeal Head Neck Surg 133:1277–1281 Siena S, Sartore-Bianchi A, Di Nicolantonio F et al, 2009, Biomarkers predicting clinical outcome of epidermal growth factor receptor-targeted therapy in metastatic colorectal cancer. J Natl Cancer Inst 101:1308–1324 Barault L, Veyrie N, Jooste V et al, 2008, Mutations in the RASMAPK, PI(3)K, phosphatidylinositol-3-OH kinase, signaling network correlate with poor survival in a population-based series of colonic cancer. Int J Cancer 122:2255–2259 Artale S, Sartore-Bianchi A, Veronese S et al, 2008, Mutations of KRAS and BRAF in primary and matched metastatic sites of colorectal cancer. J Clin Oncol 26:4217–4219 Andreyev HJ, Norman AR, Cunningham D et al, 2001, Kirsten ras mutation in patients with colorectal cancer: the ‘RASCAL II’ study. Br J Cancer 85:692–696 Van Cutsem E, Peeters M, Siena S et al, 2007, Open-label phase III trial of panitumumab plus best supportive care compared to best supportive care alone in patients with chemotherapy-refractory a metastatic colorectal cancer. J Clin Oncol 25:1658–1664 Amado RG, Wolf M, Peeters M et al, 2008, Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 26:1626–1634 Roth A, Tejpar S, Yan P et al, 2009, Correlation of molecular markers in colon cancer with stage-specific prognosis: results of the translational study on the PETACC 3-EORTC 40093-SAKK 60-00 trial. ASCO, Gastrointestinal cancers Symposium, San Francisco, abstract 288 Moroni M, Veronese S, Benvenuti S et al, 2005, Gene copy number for epidermal growth factor receptor, EGFR, and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. Lancet Oncol 6:279–286 Benvenuti S, Sartore-Bianchi A, Di Nicolantonio F et al, 2007, Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to antiepidermal growth factor receptor antibody therapies. Cancer Res 67:2643–2648 Lievre A, Bachet JB, Le Corre D et al, 2006, KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 66:3992–3995 Monzon FA, Ogino S, Hammond EH et al, 2009, The role of KRAS mutation testing in the management of patients with metastatic colorectal cancer. Arch Pathol Lab Med 133:1600–1606 Di Nicolantonio F, Martini M, Molinari F et al, 2008, Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol 26:5705–5712 Sartore-Bianchi A, Martini M, Molinari F et al, 2009, PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies. Cancer Res 69:1851–1857 Prenen H, De Schutter J, Jacobs B et al, 2009, PIK3CA mutations are not a major determinant of resistance to the epidermal growth factor receptor inhibitor cetuximab in metastatic colorectal cancer. Clin Cancer Res 15:3184–3188 Benvenuti S, Frattini M, Arena S et al, 2008, PIK3CAcancer mutations display gender and tissue specificity patterns. Hum Mutat 29:284–288 Van Cutsem E, Kohne CH, Hitre E et al, 2009, Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med 360:1408–1417 Sartore-Bianchi A, Moroni M, Veronese S et al, 2007, Epidermal growth factor receptor gene copy number and clinical outcome of metastatic colorectal cancer treated with panitumumab. J Clin Oncol 25:3238–3245 Rowinsky EK, Schwartz GH, Gollob JA et al, 2004, Safety, pharmacokinetics, and activity of ABX-EGF, a fully human antiepidermal growth factor receptor monoclonal antibody in patients with metastatic renal cancer. J Clin Oncol 22:3003–3015 Weiner LM, Belldegrun AS, Crawford J et al, 2008, Dose and schedule study of panitumumab monotherapy in patients with advanced solid malignancies. Clin Cancer Res 14:502–508 Lynch TJJ, Kim ES, Eaby B et al, 2007, Epidermal growth factor receptor inhibitor-associated cutaneous toxicities: an evolving paradigm in clinical management. Oncologist 12:610–621 Arends R, Yang B, Schwab G et al, 2005, Flexible dosing schedules of panitumumab in cancer patients. J Clin Oncol 23, abstract 3089 Berlin J, Posex J, Tchekmedyian S et al, 2007, Panitumumab with irinotecan/leucovorin/5-fluorouracil for first-line treatment of metastatic colorectal cancer. Clin Colorectal Cancer 6:427–432 Hecht JR, Patnaik A, Berlin J et al, 2007, Panitumumab monotherapy in patients with previously treated metastatic colorectal cancer. Cancer 110:980–988 Mitchell EP, Hecht JR, Baranda J et al, 2007, Panitumumab activity in metastatic colorectal cancer, mCRC, petients, pts, with low or negative tumor epidermal growth factor receptor, EGFRr, levels: An updated analysis. J Clin Oncol 25, abstract 4082 Doi T, Ohtsu A, Tahara M et al, 2009, Safety and pharmacokinetics of panitumumab in Japanase ptients with advanced solid tumors. Int J Clin Oncol 14:307–314 Kim R. Cetuximab and panitumumab: are they interchangeable? Lancet Oncol, DOI 10.1016/S1470-2045(09)70312-2 Douillard JS, Cassidy J, Tabermero J et al, 2009, Randomized phase 3 study on panitutumab with FOLFOX4 compared to FOLFOX alone as first-line treatment, tx, for metastatic colorectal cancer, mCRC): the PRIME trial. Eur J Cancer Suppl 7:6 Peeters M, Price T, Hotko Y et al, 2009, Randomized phase 3 study of panitumumab with FOLFIRI vs FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancer. Eur J Cancer Suppl 7:2 Hecht JR, Mitchell E, Chidiac T et al, 2009, A randomized phase IIIB trial of chemotherapy, bevacizumab and panitumumab compared with chemotherapy and bevacizumab alone for metastatic colorectal cancer. J Clin Oncol 27:672–680 Tol J,Koopman M, Cats Aet al, 2009, Chemotherapy, bevacizumab, and cetuximab in metastatic colorectal cancer. N Engl J Med 360:563–572 Berlin J, Van Cutsem E, Peeters M et al, 2007, Predictive value of skin toxicity severity for response to panitumumab in patients with metastatic colorectal cancer, mCRC): a pooled analysis of five clinical trials. J Clin Oncol 25(suppl, abstract 4134 Bencardino K, Ronzoni M, Manzoni M et al, 2008, In vivo biological effects of Panitumumab plus chemotherapy in advanced colorectal cancer patients. J Clin Oncol 26, abstract 14576 Hendlisz A, Marechal R, Durbecq V et al, 2008, Modulation and prognostic value of epidermal growth factor receptor expression in circulating tumor cells during chemotherapy in patients with metastatic colon cancer. J Clin Oncol 26, abstract 15038 Freeman DJ, Juan T, Reiner M et al, 2008, Association of KRAS mutational status and clinical outcomes in patients with metastatic colorectal cancer receiving panitumumab alone. Clin Colorectal Cancer 7:184–190 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 143 EP 148 DI 10.1007/s12253-010-9257-7 PG 6 ER PT J AU Werling, K Schaff, Zs Dinya, E Tulassay, Zs AF Werling, Klara Schaff, Zsuzsa Dinya, Elek Tulassay, Zsolt TI Effect of Liver Steatosis on Therapeutic Response in Chronic Hepatitis C Virus Genotype 1 Infected Patients in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatitis C virus; Genotype 1; Steatosis; Therapy ID Hepatitis C virus; Genotype 1; Steatosis; Therapy AB Hepatic steatosis seems a frequent histological alteration seen in chronic hepatitis C virus infected patients. There is still a lot to learn about the exact mechanism of effect of liver steatosis and its influence on the progression of liver diseases. Our study involved 96 chronic hepatitis C genotype 1 infected Hungarian patients who received pegylated interferon and ribavirin treatment for the first time. Degree of steatosis, viral and host factors influencing its development and its effect on the efficiency of antiviral treatment were determined. In 61 (64%) of patients the liver tissue showed varying degree of steatosis, which did not show relationship with level of alcohol consumption (p=0.5792), diabetes mellitus (p=0.5925) or body mass index (p=0.9685) in type 1 chronic hepatitis C patients. Degree of steatosis and virus titer showed strong relationship (OR=2.1). Significant relationship was also found between degree of hepatic steatosis and stage (p=0.0119), as well as between therapeutic response to combined pegylated interferon + ribavirin treatment and steatosis (p=0.0012). Our results demonstrated that steatosis has clinical significance in hepatitis C virus genotype 1 infected patients. C1 [Werling, Klara] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Dinya, Elek] EGIS Pharmaceuticals PLC, Medical DepartmentBudapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. RP Werling, K (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM werling@freemail.hu CR Seeff LB, Buskell-Bales Z, Wright EC, National Heart, Lung and Blood Institute Study Group et al, 1991, Long-term mortality after transfusion-associated non-A,non-B hepatitis. N Engl J Med 327:1906–1911 Poynard T, Bedossa P, Opolon P, 1997, Natural history of liver fibrosis progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, CLINIVIR, and DOSVIRC groups. Lancet 349(9055):825–832 Mihm S, Fayyazi A, Hartmann H et al, 1997, Analysis of histopathological manifestations of chronic hepatitis C virus infection with respect to virus genotype. Hepatology 25:735–739 Czaja AJ, Carpenter HA, Santrach PJ et al, 1998, Host- and disease-specific factors affecting steatosis in chronic hepatitis C. J Hepatol 29:198–206 Hourigan LF, Macdonald GA, Purdie D et al, 1999, Fibrosis in chronic hepatitis C correlates significantly with body mass index and steatosis. Hepatology 29:1215–1219 Rubbia-Brandt L, Quadri R, Abid K et al, 2000, Hepatocyte steatosis is a cytopathic effect of hepatitis C virus genotypes 3. J Hepatol 33:106–115 Adinolfi LE, Gambardella M, Andreana A et al, 2001, Steatosis accelerates the progression of liver damage of chronic hepatitis C patients and correlates with specific HCV genotype and visceral obesity. Hepatology 33:1358–1364 Serfaty L, Andreani T, Giral P et al, 2001, Hepatitis C virus induced hypobetalipoproteinemia: a possible mechanism for steatosis in chronic hepatitis C. J Hepatol 34:428–434 Monto A, Alonzo J, Watson JJ et al, 2002, Steatosis in chronic hepatitis C: relative contribution of obesity, diabetes mellitus, and alcohol. Hepatology 36:729–736 Westin J, Nordlinder H, Lagging M et al, 2002, Steatosis accelerates fibrosis development over time in hepatitis C virus genotype 3 infected patients. J Hepatol 37:837–842 Hui JM, Kench J, Farrell GC et al, 2002, Genotype-specific mechanisms for hepatic steatosis in chronic hepatitis C infection. J Gastroenterol Hepatol 17:873–881 Castera L, Hezode C, Roudot-Thoraval F et al, 2003, Worsening of steatosis is an independent factor of fibrosis progression in untreated patients with chronic hepatitis C and paired liver biopsies. Gut 52:288–292 Poynard T, Ratziu V, McHutchinson J et al, 2003, Effect of treatment with peginterferon or interferon alfa-2b and ribavirin on steatosis in patients infected with hepatitis C. Hepatology 38:75– 85 Asselah T, Boyer N, Guimont MC et al, 2003, Liver fibrosis is not associated with steatosis but with necroinflammation in French patients with chronic hepatitis C. Gut 52:1638–1643 Rubbia-Brandt L, Fabris P, Paganin S et al, 2004, Steatosis affects chronic hepatitis C progression in a genotype-specific way. Gut 53:406–412 Patton HM, Patel K, Behling C et al, 2004, The impact of steatosis on disease progression and early sustained treatment response in chronic hepatitis C patients. J Hepatol 40:484– 490 Asselah T, Rubbia-Brandt L, Marcellin P et al, 2006, Steatosis in chronic hepatitis C: why does it really matter? Gut 55:123–130 Allison ME, Wreghitt T, Palmer CR et al, 1994, Evidence for a link between hepatitis C virus infection and diabetes mellitus in a cirrhotic population. J Hepatol 21:1135–1139 Caronia S, Taylor K, Pagliaro L et al, 1999, Further evidence for an association between non-insulin-dependent diabetes mellitus and chronic hepatitis C virus infection. Hepatology 30:1059–1063 Mason AL, Lau JY, Hoang N et al, 1999, Association of diabetes mellitus and chronic hepatitis C virus infection. Hepatology 29:328–333 Petit JM, Bour JB, Galland-Jos C et al, 2001, Risk factors for diabetes mellitus and early insulin resistance in chronic hepatitis C. J Hepatol 35:279–283 Mehta SH, Brancati FL, Strathdee SA et al, 2003, Hepatitis C infection and incident type 2 diabetes. Hepatology 38:50–56 Hui KQ, Esrailian E, Runyon B et al, 2002, Clinical implication of hepatic steatosis in patients with chronic hepatitis C. Hepatology 34:187A Kumar D, Farell GC, Fung C et al, 2002, Hepatitis C virus genotype 3 is cytopathic to hepatocytes: reversal of hepatic steatosis after sustained therapeutic response. Hepatology 36:1266–1272 Moriya K, Yotsuyanagi H, Shintani Y et al, 1997, Hepatitis C virus core protein induces hepatic steatosis in transgenic mice. J Gen Virol 78:1527–1531 Kumar D, Farrell GC, Fung C et al, 2002, Hepatitis C virus genotype 3 is cytopathic to hepatocytes: reversal of hepatic steatosis after sustained therapeutic response. Hepatology 36:1266–1272 Gervain J, Simon G, Papp I et al, 2001, Determination of the type and subtype of the hepatitis virus C in chronic viral hepatitis patients in Hungary. Orv Hetil 142:1315–1319 Tornai I, Dalmi L, Gervain J et al, 2005, Treatment of patients with chronic hepatitis C, using pegylated interferon alfa-2a and ribavirin — the first experiences in Hungary based upon a multicentric, open, prospective study. LAM 15:807–813 Ishak K, Baptista A, Bianchi L et al, 1995, Histological grading and staging of chronic hepatitis. J Hepatol 22:696–699 Brunt EM, Janney CG, Di Bisceglie AM et al, 1999, Nonalcoholic steatohepatitis: a proposal for grading and staging the histological lesions. Am J Gastroenterol 94:2467–2474 Jarmay K, Karacsony G, Nagy A et al, 2005, Changes in lipid metabolism in chronic hepatitis C. World J Gastroenterol 11:6422–6428 Abid K, Pazienza V, de Gottardi A et al, 2005, An in vitro model of hepatitis C virus genotype 3a-associated triglycerides accumulation. J Hepatol 42:744–751 Barba G, Harper F, Harada T et al, 1997, Hepatitis C core protein shows a cytoplasmic localization and associates to cellular lipid storage droplets. Proc Nat Acad Sci USA 94:1200–1205 Shi ST, Polyak SJ, Tu H et al, 2002, Hepatitis C virus NS5A colocalizes with the core protein on lipid droplets and interacts with apolipoproteins. Virology 292:198–210 Perlemuter G, Sabile A, Letteron P et al, 2002, Hepatitis C core protein inhibits microsomal triglycerid transfer protein activity and very low density lipoprotein secretion: a model of viralrelated steatosis. FASEB J 16:185–194 Fartoux L, Chazouilleres O, Wendum D et al, 2005, Impact of steatosis on progression of fibrosis in patients with mild hepatitis C. Hepatology 41:82–87 Day CP, James OFW, 1998, Steatohepatitis: a tale of two “hits”? Gastroenterology 114:842–845 Lee KS, Buck M, Houglum K et al, 1995, Activation of hepatic stellate cells by TGF alpha and collagen type I is mediated by oxidative stress through c-myb expression. J Clin Invest 96:2461– 2468 Bedossa P, Houglum K, Trautwein C et al, 1994, Stimulation of collagen alpha 1(1, gene expression is associated with lipid peroxidation in hepatocellular injury: a link to tissue fibrosis? Hepatology 19:1262–1271 Parola M, Pinzani M, Casini A et al, 1993, Stimulation of lipid peroxidation or 4-hydroxynonenal treatment increases procollagen alpha 1, 1, gene expression in human liver fat-storing cells. Biochem Biophys Res Commun 194:1044–1050 Farinati F, Cardin R, De Maria N et al, 1995, Iron strorage, lipid peroxidation and glutathion turnover in chronic anti-HCV positive hepatitis. J Hepatol 22:449–456 Paradis V, Mathurin P, Kollinger M et al, 1997, In situ detection of lipid peroxidation in chronic hepatitis C: correlation with pathological features. J Clin Pathol 50:401–406 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 149 EP 157 DI 10.1007/s12253-009-9195-4 PG 9 ER PT J AU Badid, N Baba Ahmed, ZF Merzouk, H Belbraouet, S Mokhtari, N Merzouk, AS Benhabib, R Hamzaoui, D Narce, M AF Badid, Naima Baba Ahmed, Zohra Fatima Merzouk, Hafida Belbraouet, Slimane Mokhtari, Nassima Merzouk, Ahmed Sid Benhabib, Riad Hamzaoui, Djalloul Narce, Michel TI Oxidant/Antioxidant Status, Lipids and Hormonal Profile in Overweight Women with Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Fatty acids; Leptin; Lipids; Lipoproteins; Oxidative stress ID Breast cancer; Fatty acids; Leptin; Lipids; Lipoproteins; Oxidative stress AB This study was carried out to determine the relationships between leptin concentrations, lipid alterations, oxidant/ antioxidant status, in vitro LDL oxidizability and LDL-fatty acid composition in overweight breast cancer patients. Glucose, insulin, leptin, lipids, LDL-cholesteryl ester fatty acids, markers of oxidant status (MDA, Hydroperoxides, carbonyl proteins, conjugated dienes) and markers of antioxidant status (vitamins A, C, E, erythrocyte activities of the enzymes superoxide dismutase, SOD, catalase, glutathione peroxidase,GPx, and glutathione reductase, GR and the serum total antioxidant status, ORAC) were investigated in breast cancer patients and in control women. Our findings showed that insulin, leptin, triglyceride, cholesterol and LDL-C concentrations were increased in patients compared to controls. ORAC and vitamin C and E values were lower while plasma hydroperoxide, carbonyl protein and conjugated diene levels, SOD and GPx activities were higher than in controls. Alterations in LDL-fatty acid composition were associated with their enhanced oxidative susceptibility. There were significant positive correlations between leptin concentrations and LDL-C, hydroperoxides, carbonyl proteins, SOD activity, baseline conjugated diene levels and oxidation rate, and significant negative correlations between leptin and ORAC, lag time and LDL-PUFA in patients. In conclusion, breast cancer is associated with lipid alterations and enhanced oxidative stress linked to high leptin levels in overweight. C1 [Badid, Naima] University of Tlemcen, Faculty of Sciences, Department of Molecular and Cellular BiologyTlemcen, Algeria. [Baba Ahmed, Zohra Fatima] University of Tlemcen, Faculty of Sciences, Department of Molecular and Cellular BiologyTlemcen, Algeria. [Merzouk, Hafida] University of Tlemcen, Faculty of Sciences, Department of Molecular and Cellular BiologyTlemcen, Algeria. [Belbraouet, Slimane] University of Moncton, School of nutritionMoncton, Canada. [Mokhtari, Nassima] University of Tlemcen, Faculty of Sciences, Department of Molecular and Cellular BiologyTlemcen, Algeria. [Merzouk, Ahmed Sid] University of Tlemcen, Faculty of EngineeringTlemcen, Algeria. [Benhabib, Riad] Tlemcen Hospital, Division of Obstetrics and GynecologyTlemcen, Algeria. [Hamzaoui, Djalloul] Surgery Clinic AVICENEMaghnia, Algeria. [Narce, Michel] University of Bourgogne, Faculty of Sciences, INSERM UMR 866,Dijon, France. RP Merzouk, H (reprint author), University of Tlemcen, Faculty of Sciences, Department of Molecular and Cellular Biology, Tlemcen, Algeria. EM hafidamerzouk_2@hotmail.com CR World Health Organization, International Agency for Research on Cancer, 2002, IARC handbooks of cancer prevention: breast cancer screening. IRAC, Layon, pp 1–7 Kaaks R, 1996, Nutrition, hormones, and breast cancer: is insulin the missing link? Cancer Causes Control 7:605–625 Nelson NJ, 2006, Migrant studies aid the search for factors linked to breast cancer risk. J Natl Cancer Inst 98:436–438 Kang DH, 2002, Oxidative stress, DNA damage, and breast cancer. AACN Clin Issues 13:540–549 Malins DC, 1996, Free radicals and breast cancer. Environ Health Perspect 104:1140 Ahn J, Nowell S, McCann SE, Yu J et al, 2006, Associations between catalase phenotype and genotype: modification by epidemiologic factors. Cancer Epidemiol Biomarkers Prev 15: 1217–1222 Saadatian-Elahi M, Norat T, Goudable J, Riboli E, 2004, Biomarkers of dietary fatty acid intake and the risk of breast cancer: a meta-analysis. Int J Cancer 111:584–591 Abu-Bedair FA, El-Gamal BA, Ibrahim NA, El-Aaser AA, 2003, Serum lipids and tissue DNA content in Egyptian female breast cancer patients. Jpn J Clin Oncol 33:278–282 Am F, Branchi A, Sommaviva D, 2000, Serum lipoprotein profile in patients with cancer. A comparison with non-cancer subjects. Int J Clin Lab Res 30:141–145 Merzouk S, Hichami A, Sari A et al, 2004, Impaired oxidant/ antioxidant status and LDL-fatty acid composition are associated with increased susceptibility to peroxidation of LDL in diabetic patients. J Gen Physiol Biophys 23:387–399 Peacock SL, White E, Daling JR, Voigt LF, Malone KE, 1999, Relation between obesity and breast cancer in young women. Am J Epidemiol 149:339–346 Urakawa H, Katsuki A, Sumida Y et al, 2003, Oxidative stress is associated with adiposity and insulin resistance in men. J Clin Endocrinol Metab 88:4673–4676 Schindler TH, Cardenas J, Prior JO et al, 2006, Relationship between increasing body weight, insulin resistance, inflammation, adipocytokine leptin, and coronary circulatory function. J Am Coll Cardiol 47:1188–1195 Maeso Fortuny MC, Brito Diaz B, Cabrera de Leon A, 2006, Leptin, estrogens and cancer. Mini Rev Med Chem 6:897–907 Sulkowska M, Golaszewska J, Wincewicz A, Koda M, Baltaziak M, Sulkowski S, 2006, Leptin—from regulation of fat metabolism to stimulation of breast cancer growth. Pathol Oncol Res 12:69–72 Roe JH, Kuether CA, 1943, The determination of ascorbic acid in whole blood and urine through the 2, 4-dinitrophenylhydrazine derivatives of dehydroascorbic acid. J Biol Chem 147:399–407 Aebi H, 1974, Catalase. In: Bergmeyer HU, ed, Methods of enzymatic analysis, 2nd edn. Verlag Chemie GmbH, Weinheim Paglia DE, Valentine WN, 1967, Studies on the quantitative and qualitative characterizations of erythrocyte glutathione peroxidase. J Lab Clin Med 70:158–169 Goldberg DM, Spooner RJ, 1992, Glutathione reductase. In: Bergmeyer HB, ed, Methods of enzymatic analysis, vol 3, 3rd edn. Verlag Chemie GmbH, Weinheim, pp 258–265 Elstner EF, Youngman RJ, Obwad W, 1983, Superoxide dismutase. In: Bergmeyer HB, ed, Methods of enzymatic analysis, 3rd edn. Verlag Chemie GmbH, Weinheim, vol 3, pp 293–302 Nourooz-Zadeh J, Tajaddini-Sarmadi J, Ling KLE, Wolff PS, 1996, Low-density lipoprotein is the major carrier of lipid hydroperoxides in plasma. Relevance to determination of total plasma lipid hydroperoxide concentrations. Biochem J 313: 781–786 Levine RL, Garland D, Oliver CN et al, 1990, Determination of carbonyl content in oxidatively modified proteins. Methods Enzymol 186:464–478 Lowry OH, Rosebrough NJ, Farr AL, Randall RI, 1951, Protein measurement with folin phenol reagent. J Biol Chem 193:265–275 Esterbauer H, Striegl G, Puhl H, Rotheneder M, 1989, Continuous monitoring of in vitro oxidation of human low density lipoprotein. Free Rad Res Commun 6:67–75 Rose DP, Haffner SM, Baillargeon J, 2007, Adiposity, the metabolic syndrome, and breast cancer in African–American and White American women. Endocr Rev 28:763–777 Tessitore L, Vizio B, Pesola D et al, 2004, Adipocyte expression and circulating levels of leptin increase in both gynaecological and breast cancer patients. Int J Oncol 24:1529–1535 Furberg AS, Veierod MB, Wilsgaard T, Bernstein L, Thune I, 2004, Serum high-density lipoprotein cholesterol, metabolic profile, and breast cancer risk. J Natl Cancer Inst 96:1152–1160 Dhaval SF, 2008, Significance of alterations in plasma lipid profile levels in breast cancer. Integr Cancer Ther 7:33–41 Hasija K, Bagga HK, 2005, Alterations of serum cholesterol and serum lipoprotein in breast cancer of women. Indian J Clin Biochem 20:61–66 Gooden PJ, Boyd NF, Hanna W, Hartwick W, Murray D, Qizilbash A, 1997, Elevated levels of plasma triglycerides are associated with histologically defined premenopausal breast cancer risk. Nutr Cancer 27:284–292 Ambrosone CB, 2000, Oxidants and antioxidants in breast cancer. Antioxid Redox Signal 2:903–917 Rossner P, Terry MB, Gammon MD et al, 2007, Plasma protein carbonyl levels and breast cancer risk. J Cell Mol Med 11:1138–1148 Bougnoux P, Giraudeau B, Couet C, 2006, Diet, cancer, and the lipidome. Cancer Epidemiol Biomarkers Prev 15:416–421 Surapaneni KM, Gopan CS, 2007, Status of lipid peroxidation and antioxidant enzymes in patients with carcinoma of breast. JMSR 15:21–24 Delimaris I, Faviou E, Antonakos G, Stathopoulou E, Zachari A, Dionyssiou-Asteriou A, 2007, Oxidized LDL, serum oxidizability and serum lipid levels in patients with breast or ovarian cancer. Clin Biochem 40:1129–1134 Shannon J, King IB, Moshofsky R et al, 2007, Erythrocyte fatty acids and breast cancer risk: a case-control study in Shanghai, China. Am J Clin Nutr 85:1090–1097 Lu S, Archer MC, 2005, Fatty acid synthase is a potential molecular target for the chemoprevention of breast cancer. Carcinogenesis 26:153–157 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 159 EP 167 DI 10.1007/s12253-009-9199-0 PG 9 ER PT J AU Xu, Y Zhu, M Zhang, Sh Liu, H Li, T Qin, Ch AF Xu, Yijun Zhu, Mingchen Zhang, Shuhong Liu, Hui Li, Tao Qin, Chengyong TI Expression and Prognostic Value of PRL-3 in Human Intrahepatic Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intrahepatic cholangiocarcinoma; PRL-3; Lymph node metastasis; Immunochemistry; Prognosis ID Intrahepatic cholangiocarcinoma; PRL-3; Lymph node metastasis; Immunochemistry; Prognosis AB Phosphatase of regenerating liver (PRL)-3 is involved in the metastasis of various tumors, but the expression of PRL-3 and its possible role in primary intrahepatic cholangiocarcinoma (ICC) has not been reported yet. In this study, we assessed the expression levels of PRL-3 by immunohistochemistry in 102 primary ICC samples, 62 matched lymph node metastases (LNM) and 102 adjacent normal liver tissues. Then we investigated the relationship between PRL-3 expression and clinicopathologic factors. Survival analysis was performed to determine the prognostic significance of PRL-3 expression in ICC. Immunochemistry results suggested PRL-3 expression was negative or weak in non-neoplastic intrahepatic bile ducts of adjacent liver tissue. In primary lesion and LNM high PRL-3 expression was frequently detected. Furthermore, the rate of high PRL-3 expression in LNM was higher than that in primary lesion (80.6% vs. 47.1%, P<0.05). High expression of PRL-3 in primary tumors was significantly associated with TNM (P<0.001), T stage (P<0.001), vascular invasion (P=0.002), and LNM (P<0.001). Survival analysis results with Kaplan-Meier method and Cox proportional hazard model indicated high expression of PRL-3 was correlated with decreased overall survival and was an independent prognostic marker of overall survival. Thus, our results suggested high expression of PRL-3 was correlated with progression and metastasis of ICC and indicated negative prognostic impact. PRL-3 might serve as a novel prognostic marker for patients with ICC. C1 [Xu, Yijun] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. [Zhu, Mingchen] Jiangsu Cancer Hospital, Department of Clinical Laboratory, 42 Baiziting Road, 210009 Nanjing, China. [Zhang, Shuhong] Jinan Central Hospital Affiliated to Shandong University, Department of Gastroenterology, 105 Jiefang Road, 250013 Jinan, China. [Liu, Hui] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. [Li, Tao] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. [Qin, Chengyong] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. RP Qin, Ch (reprint author), Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 250021 Jinan, China. EM qinchengyong2005@yahoo.cn CR Khan SA, Thomas HC, Davidson BR, Taylor-Robinson SD, 2005, Cholangiocarcinoma. Lancet 366:1303–1314 Patel T, 2001, Increasing incidence and mortality of primary intrahepatic cholangiocarcinoma in the United States. Hepatology 33:1353–1357 Shaib Y, EI-Serag HB, 2004, The epidemiology of cholangiocarcinoma. Semin Liver Dis 24:115–125 Taylor-Robinson SD, Toledano MB, Arora S, Keegan TJ, Hargreaves S, Beck A, Khan SA et al, 2001, Increase in mortality rates from intrahepatic cholangiocarcinoma in England and Wales 1968–1998. Gut 48:816–820 Mouzas IA, Dimoulios P, Vlachonikolis IG, Skordilis P, Zoras O, Kouroumalis E, 2002, Increasing incidence of cholangiocarcinoma in Crete 1992–2000. Anticancer Res 22:3637–3641 Okuda K, Nakanuma Y, Miyazaki M, 2002, Cholangiocarcinoma: recent progress. Part 1: epidemiology and etiology. J Gastroenterol Hepatol 17:1049–1055 Okuda K, Nakanuma Y, Miyazaki M, 2002, Cholangiocarcinoma: recent progress. Part 2: molecular pathology and treatment. J Gastroenterol Hepatol 17:1056–1063 Anderson CD, Pinson CW, Berlin J, Chari RS, 2004, Diagnosis and treatment of cholangiocarcinoma. Oncologist 9:43–57 Nakagawa T, Kamiyama T, Kurauchi N, Matsushita M, Nakanishi K, Kamachi H, Kudo T et al, 2005, Number of lymph node metastases is a significant prognostic factor in intrahepatic cholangiocarcinoma. World J Surg 29:728–733 Yamamoto M, Takasaki K, Yoshikawa T, 1999, Lymph node metastasis in intrahepatic cholangiocarcinoma. Jpn J Clin Oncol 29:147–150 Harada K, Masuda S, Hirano M, Nakanuma Y, 2003, Reduced expression of syndecan-1 correlates with histologic dedifferentiation, lymph node metastasis, and poor prognosis in intrahepatic cholangiocarcinoma. Hum Pathol 34:857–863 Shimonishi T, Miyazaki K, Kono N, Sabit H, Tuneyama K, Harada K, Hirabayashi J et al, 2001, Expression of endogenous galectin-1 and galectin-3 in intrahepatic cholangiocarcinoma. Hum Pathol 32:302–310 Alonso A, Sasin J, Bottini N, Friedberg I, Friedberg I, Osterman A, Godzik A et al, 2004, Protein tyrosine phosphatases in the human genome. Cell 117:699–711 Stoker AW, 2005, Protein tyrosine phosphatases and signalling. J Endocrinol 185:19–33 van Huijsduijnen RH, Bombrun A, Swinnen D, 2002, Selecting protein tyrosine phosphatases as drug targets. Drug Discov Today 7:1013–1019 Zeng Q, Hong W, Tan YH, 1998, Mouse PRL-2 and PRL-3, two potentially prenylated protein tyrosine phosphatases homologous to PRL-1. Biochem Biophys Res Commun 244:421–427 Mohn KL, Laz TM, Hsu JC, Melby AE, Bravo R, Taub R, 1991, The immediate-early growth response in regenerating liver and insulin-stimulated H-35 cells: comparison with serum-stimulated 3T3 cells and identification of 41 novel immediate-early genes. Mol Cell Biol 11:381–390 Diamond RH, Cressman DE, Laz TM, Abrams CS, Taub R, 1994, PRL-1, a unique nuclear protein tyrosine phosphatase, affects cell growth. Mol Cel Biol 14:3752–3762 Werner SR, Lee PA, DeCamp MW, Crowell DN, Randall SK, Crowell PL, 2003, Enhanced cell cycle progression and down regulation of p21Cip1/Waf1 by PRL tyrosine phosphatases. Cancer Lett 202:201–211 Matter WF, Estridge T, Zhang C, Belagaje R, Stancato L, Dixon J, Johnson B et al, 2001, Role of PRL-3, a human muscle-specific tyrosine phosphatase, in angiotensin-II signaling. Biochem Biophys Res Commun 283:1061–1068 Zeng Q, Dong JM, Guo K, Li J, Tan HX, Koh V, Pallen CJ et al, 2003, PRL-3 and PRL-1 promote cell migration, invasion, and metastasis. Cancer Res 63:2716–2722 Saha S, Bardelli A, Buckhaults P, Velculescu VE, Rago C, St Croix B, Romans KE et al, 2001, A phosphatase associated with metastasis of colorectal cancer. Science 294:1343–1346 Wu X, Zeng H, Zhang X, Zhao Y, Sha H, Ge X, Zhang M et al, 2004, Phosphatase of regenerating liver-3 promotes motility and metastasis of mouse melanoma cells. Am J Pathol 164:2039– 2054 Kato H, Semba S, Miskad UA, Seo Y, Kasuga M, Yokozaki H, 2004, High expression of PRL-3 promotes cancer cell motility and liver metastasis in human colorectal cancer: a predictive molecular marker of metachronous liver and lung metastases. Clin Cancer Res 10:7318–7328 Qian F, Li YP, Sheng X, Zhang ZC, Song R, Dong W, Cao SX et al, 2007, PRL-3 siRNA inhibits the metastasis of B16-BL6 mouse melanoma cells in vitro and in vivo. Mol Med 13:151–159 Bardelli A, Saha S, Sager JA, Romans KE, Xin B, Markowitz SD, Lengauer C, 2003, PRL-3 expression in metastatic cancers. Clin Cancer Res 9:5607–5615 Miskad UA, Semba S, Kato H, Yokozaki H, 2004, Expression of PRL-3 phosphatase in human gastric carcinomas: close correlation with invasion and metastasis. Pathobiology 71:176–184 Miskad UA, Semba S, Kato H, Matsukawa Y, Kodama Y, Mizuuchi E, Maeda N et al, 2007, High PRL-3 expression in human gastric cancer is a marker of metastasis and grades of malignancies: an in situ hybridization study. Virchows Arch 450:303–310 Wang L, Peng L, Dong B, Kong L, Yan L, Xie Y, Shou C, 2006, Overexpression of phosphatase of regenerating liver-3 in breast cancer: association with a poor clinical outcome. Ann Oncol 17:1517–1522 Polato F, Codegoni A, Fruscio R, Perego P, Mangioni C, Saha S, Bardelli A et al, 2005, PRL-3 phosphatase is implicated in ovarian cancer growth. Clin Cancer Res 11:6835–6839 Ren T, Jiang B, Xing X, Dong B, Peng L, Meng L, Xu H et al, 2009, Prognostic Significance of Phosphatase of Regenerating Liver-3 Expression in Ovarian Cancer. Pathol Oncol Res, Epub ahead of print) Nakanuma Y, Sripa B, Vatanasapt V, Leong ASY, Ponchon T, Ishak KG, 2000, Intrahepatic cholangiocarcinoma. In: Hamilton SR, Aaltonen LA, eds, World Health Organization classification of tumours: pathology and genetics of tumours of the digestive system. IARC Press, Lyon, pp 173–180 Junking M, Wongkham C, Sripa B, Sawanyawisuth K, Araki N, Wongkham S, 2008, Decreased expression of galectin-3 is associated with metastatic potential of liver fluke-associated cholangiocarcinoma. Eur J Cancer 44:619–626 Peng L, Ning J, Meng L, Shou C, 2004, The association of the expression level of protein tyrosine phosphatase PRL-3 protein with liver metastasis and prognosis of patients with colorectal cancer. J Cancer Res Clin Oncol 130:521–526 Nichols JC, Gores GJ, LaRusso NF, Wiesner RH, Nagorney DM, Ritts RE Jr, 1993, Diagnostic role of serum CA19–9 for cholangiocarcinoma in patients with primary sclerosing cholangitis. Mayo Clin Proc 68:874–879 Patel AH, Harnois DM, Klee GG, LaRusso NF, Gores GJ, 2000, The utility of CA19–9 in the diagnoses of cholangiocarcinoma in patients without primary sclerosing cholangitis. Am J Gastroenterol 95:204–207 Paik KY, Jung JC, Heo JS, Choi SH, Choi DW, Kim YI, 2008, What prognostic factors are important for resected intrahepatic cholangiocarcinoma? J Gastroenterol Hepatol 23:766–770 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 169 EP 175 DI 10.1007/s12253-009-9200-y PG 7 ER PT J AU Raspollini, RM Stomaci, N Ringressi, A Franchi, A AF Raspollini, Rosaria Maria Stomaci, Niceta Ringressi, Andrea Franchi, Alessandro TI Primitive Testicular Leiomyosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Testis; Leiomyosarcoma ID Testis; Leiomyosarcoma AB Primary testicular leiomyosarcoma is an uncommon tumor with only few cases described in literature. In young people this rare tumor seems to be related to radiotherapy and anabolic steroids abuse. In older people there are apparently no risk factors. We describe one further case in a 77-years old man with full histological and ultrastructural evaluation. A short term follow-up of one year signals no recurrence of the disease. C1 [Raspollini, Rosaria Maria] AOU Careggi, Department of Human Pathology and Oncology, Viale G.B. Morgagni, 85, 50134 Florence, Italy. [Stomaci, Niceta] University of Florence, Carreggi Hospital, Urology ClinicFlorence, Italy. [Ringressi, Andrea] University of Florence, Carreggi Hospital, Urology ClinicFlorence, Italy. [Franchi, Alessandro] AOU Careggi, Department of Human Pathology and Oncology, Viale G.B. Morgagni, 85, 50134 Florence, Italy. RP Raspollini, RM (reprint author), AOU Careggi, Department of Human Pathology and Oncology, 50134 Florence, Italy. EM mariarosaria.raspollini@unifi.it CR Canales BK, Lukasewycz SJ, Manivel JC, Pryor JL, 2005, Postradiotherapy intratesticular leiomyosarcoma. Urology 66:657. e19–657e.20 Froehner M, Fischer R, Leike S et al, 1999, Intratesticular leiomyosarcoma in a young man after high dose doping with oral-turinabol. Cancer 86:1571–1575 Hachi H, Bougtab A, Amhajji R et al, 2002, A case report of testicular leiomyosarcoma. Med Top Mars 62:531–533 Ali Y, Kehinde EO, Makar R et al, 2002, Leiomyosarcoma complicating chronic inflammation of the testis. Med Principle Pract 11:157–160 Takizawa A, Miura T, Fujinami K et al, 2005, Primary testicular leiomyosarcoma. Int J Urol 12:596–598 Kumar M, Patne SCU, Kumar S, Shukla VK, 2009, Primary highgrade testicular leiomyosarcoma. Indian J Pathol Microbiol 52:91– 93 Eble JN, Sauter G, Epstein JI, Sesterhenn IA, 2004, Tumours of the urinary system and male genital organs. IARC, Lyon Deveci MS, Deveci G, Onguru O, Kilciler M, Celasum B, 2002, Testicular, gonadal stromal, fibroma: case report and review of the literature. Path International 52:326–330 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 177 EP 179 DI 10.1007/s12253-009-9202-9 PG 3 ER PT J AU Mannan, ASRA Rifaat, AA Kahvic, M Kapila, K Mallik, M Grover, KV Bharati, Ch Perry, A AF Mannan, Ala Syed Rifat Abul Rifaat, A Amre Kahvic, Mirza Kapila, Kusum Mallik, Mrinmay Grover, Kumar Vinod Bharati, Chandramouli Perry, Arie TI Proximal-Type Epithelioid Sarcoma in the Groin Presenting as a Diagnostic Dilemma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Epitheliod sarcoma; FISH; INI1; Immunohistochemistry; Malignant rhabdoid tumor ID Epitheliod sarcoma; FISH; INI1; Immunohistochemistry; Malignant rhabdoid tumor AB Epithelioid sarcoma is an uncommon soft-tissue sarcoma typically presenting as a subcutaneous or deep dermal mass in the distal extremities of young adults. Lately, a ‘proximal’ subtype has been described, which occurs in the pelvic and genital areas of somewhat older individuals and tends to behave more aggressively than the conventional subtype. The correct diagnosis of this subtype is essential, since this tumor can be easily mistaken for other malignant tumors that exhibit epithelioid morphology.We report a case of proximal-type epithelioid sarcoma that presented as an inguinal mass in a 47-year-old man. Histologically, the tumor consisted of diffuse sheets of epithelioid cells with scattered rhabdoid morphology. By immunohistochemistry, the neoplastic cells expressed cytokeratin, epithelial membrane antigen, vimentin, CD34, CD99 and showed complete loss of nuclear INI1 protein expression. Fluorescence in situ hybridization was considered borderline for 22q deletion. We present this case to emphasize the importance of diagnosing this uncommon tumor and the role of INI1 immunohistochemistry in establishing the diagnosis. C1 [Mannan, Ala Syed Rifat Abul] Al Jahra Hospital, Department of Pathology, 02153 Jahra, Kuwait. [Rifaat, A Amre] Al Jahra Hospital, Department of Pathology, 02153 Jahra, Kuwait. [Kahvic, Mirza] Al Jahra Hospital, Department of Pathology, 02153 Jahra, Kuwait. [Kapila, Kusum] Kuwait University, Faculty of Medicine, Department of PathologyKuwait City, Kuwait. [Mallik, Mrinmay] Mubarak Al Kabir Hospital, Department of CytopathologyJabriya, Kuwait. [Grover, Kumar Vinod] Al Jahra Hospital, Department of SurgeryJahra, Kuwait. [Bharati, Chandramouli] Al Jahra Hospital, Department of RadiologyJahra, Kuwait. [Perry, Arie] Washington University School of Medicine, Department of Pathology and ImmunologySt. Louis, MO, USA. RP Mannan, ASRA (reprint author), Al Jahra Hospital, Department of Pathology, 02153 Jahra, Kuwait. EM mannanrifat@rediffmail.com CR Guillou L, Wadden C, Coindre JM, Krausz T, Fletcher CD, 1997, “Proximal-type” epithelioid sarcoma: a distinctive aggressive neoplasm showing rhabdoid features. Clinicopathologic, immunohistochemical, and ultrastructural study of a series. Am J Surg Pathol 21:130–146 Roberts CW, Orkin SH, 2004, The SWI/SNF complex-chromatin and cancer. Nat Rev Cancer 4:133–142 Versteege I, Sevenet N, Lange J et al, 1998, Truncating mutation of hSNF5/INI1 in aggressive pediatric cancer. Nature 394:203– 206 Rousseau-Merck MF, Versteege I, Legrand I et al, 1999, hSNF5/ INI1 inactivation is mainly associated with homozygous deletions and mitotic recombinations in rhabdoid tumors. Cancer Res 59:3152–3156 Biegel JA, Kalpana G, Knudsen ES et al, 2002, The role of INI1 and the SWI/SNF complex in the development of rhabdoid tumors: meeting summary from the workshop on childhood atypical teratoid/rhabdoid tumors. Cancer Res 62:323–328 Hoot AC, Russo P, Judkins AR et al, 2004, Immunohistochemical analysis of the hSNF5/INI1 distinguishes renal and extra-renal malignant rhabdoid tumors from other pediatric soft tissue tumors. Am J Sug Pathol 28:1485–1491 Sigauke E, Rakheja D, Maddox DL et al, 2006, Absence of expression of SMARCB1/INI1 in malignant rhabdoid tumors of the central nervous system, kidneys and soft tissue: an immunohistochemical study with implications for diagnosis. Mod Pathol 19:717–725 Perry A, Fuller CE, Judkins AR, Dehner LP, Biegel JA, 2005, INI1 expression is retained in composite rhabdoid tumors, including rhabdoid meningiomas. Mod Pathol 18:951–958 Modena P, Lualdi E, Facchinetti F et al, 2005, SMARCB1/INI1 Tumor suppressor gene is frequently inactivated in epithelioid sarcomas. Cancer Res 65:4012–4019 Hornick JL, Dal Cin P, Fletcher CD, 2009, Loss of INI1 expression is characteristic of both conventional and proximaltype epithelioid sarcoma. Am J Surg Pathol 33:542–550 Chbani L, Guillou L, Terrier P et al, 2009, Epithelioid sarcoma: a clinicopathologic and immunohistochemical study of 106 cases from the French sarcoma group. Am J Clin Pathol 131:22–27 Kohashi K, Izumi T, Oda Y et al, 2009, Infrequent SMARCB1/ INI1 gene alteration in epithelioid sarcoma: a useful tool in distinguishing epithelioid sarcoma from malignant rhabdoid tumor. Hum Pathol 40:349–355 Enzinger FM, 1970, Epithelioid sarcoma: a sarcoma simulating a granuloma or carcinoma. Cancer 26:1029–1041 Hasegawa T, Matsuno Y, Shimoda T, Umeda T, Yokoyama R, Hirohashi S, 2001, Proximal-type epithelioid sarcoma: a clinicopathologic study of 20 cases. Mod Pathol 14:655–663 Daimaru Y, Hashimoto H, Tsuneyoshi M, Enjoji M, 1987, Epithelial profile of epithelioid sarcoma. An immunohistochemical analysis of eight cases. Cancer 59:134–141 Billings SD, Folpe AL, Weiss SW, 2003, Epithelioid sarcoma-like hemangioendothelioma. Am J Surg Pathol 27:48–57 Raoux D, Peoch M, Pedeutour F et al, 2009, Primary epithelioid sarcoma of bone. Report of a unique case, with immunohistochemical and fluorescent in situ hybridization confirmation of INI1 deletion. Am J Surg Pathol 33:954–958 Fanburg-Smith JC, Hengge M, Hengge UR, Smith JS Jr, Miettinen M, 1998, Extrarenal rhabdoid tumors of soft tissue: a clinicopathologic and immunohistochemical study of 18 cases. Ann Diagn Pathol 2:351–362 Oda Y, Tsuneyoshi M, 2006, Extrarenal rhabdoid tumors of soft tissue: clinicopathological and molecular genetic review and distinction from other soft-tissue sarcomas with rhabdoid features. Pathol Int 56:287–295 Rekhi B, Gorad BD, Chinoy RF, 2008, Clinicopathological features with outcomes of a series of conventional and proximaltype epithelioid sarcomas, diagnosed over a period of 10 years at a tertiary cancer hospital in India. Virchows Arch 453:141–153 Chase DR, 1997, Do “rhabdoid features” impart a poorer prognosis to proximal-type epithelioid sarcomas? Commentary. Adv Anat Pathol 5:293–295 Livi L, Shah N, Paiar F, Fisher C, Judson I, Moskovic E et al, 2003, Treatment of epithelioid sarcoma at Royal Marsden Hospital. Sarcoma 7:149–152 Suit HD, Russell WO, Martin RG, 1975, Sarcoma of soft tissue: clinical and histopathologic parameters and response to treatment. Cancer 35:1478–1483 Onol FF, Tanidir Y, Kotiloglu E, Bayramicli M, Turhal S, Turkeri LN, 2006, Proximal type epithelioid sarcoma of the scrotum: a source of diagnostic confusion that needs immediate attention. Eur Urol 49:406–407 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 181 EP 188 DI 10.1007/s12253-009-9203-8 PG 8 ER PT J AU Guertl, B Leuschner, I Guelly, Ch Ebner, B Kronberger, C Hoefler, G AF Guertl, Barbara Leuschner, Ivo Guelly, Christian Ebner, Birgit Kronberger, Cornelia Hoefler, Gerald TI Is Predisposition for Nephroblastoma Linked to Polymorphisms of the WTX Gene? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE WTX; Nephroblastoma; SNP; HRMA; Polymorphism ID WTX; Nephroblastoma; SNP; HRMA; Polymorphism AB Inactivation of Wilms´ tumor X (WTX) gene has been linked to the pathogenesis of a varying percentage of nephroblastomas. In contrast, germline mutations of WTX were identified to cause bone dysplasia, but not to induce the development of nephroblastomas. In our study we investigated whether tumor promotion of nephroblastoma by inactivation of WTX gene is linked to certain single nucleotide polymorphisms (SNPs). Therefore 8 SNPs—distributed over the whole length of the WTX gene—were investigated by high resolution melting curve analysis (HRMA) and sequencing of genomic DNA from nephroblastoma patients (NB) and controls. No difference was detected in the 8 SNPs investigated, which were distributed over the whole length of the gene. Additionally, sequence analysis of the coding part of the WTX gene of the tumor samples revealed no chromosomal aberration. Our study indicates, that inactivation of WTX appears to be a late event in tumorigenesis of nephroblastoma in a subgroup of nephroblastomas. C1 [Guertl, Barbara] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. [Leuschner, Ivo] Christian Albrechts University Kiel, Institute of Pathology, Kiel Pediatric Tumor Registry, Michaelisstr. 11, 24105 Kiel, Germany. [Guelly, Christian] Medical University of Graz, Centre of Medical Research, Stiftingtalstrasse 24, A-8010 Graz, Austria. [Ebner, Birgit] Medical University of Graz, Centre of Medical Research, Stiftingtalstrasse 24, A-8010 Graz, Austria. [Kronberger, Cornelia] General Hospital and Paracelsus Medical University Salzburg, Department of Pathology, Muellner Hauptstrasse 48, A-5020 Salzburg, Austria. [Hoefler, Gerald] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. RP Guertl, B (reprint author), Medical University of Graz, Department of Pathology, A-8036 Graz, Austria. EM barbara.guertl-lackner@klinikum-graz.at CR Breslow NE, Olshan A, Beckwith JB et al, 1993, Epidemiology of Wilms tumor. Med Pediatr Oncol 21:178–181 Rivera MN, Kim WJ, Wells J et al, 2007, An X chromosome gene, WTX, is commonly inactivated in Wilms tumor. Science 315:642– 645 Perotti D, Gamba B, Sardella M et al, 2008, Functional inacitvation of the WTX gene is not a frequent event in Wilms´ tumors. Oncogene 27:4625–4632 Lehnerdt GF, Franz P, Winterhoff S et al, 2008, The GNAS1 T393C polymorphism predicts survival in patients with advanced squamous cell carcinoma of the larynx. Laryngoscope 118:2172– 2176 Guertl B, Leuschner I, Harms D, 2006, Genetic clonality is a feature unifying nephroblastomas regardless of the variety of morphological subtypes. Virch Arch 449:171–174 Scott RH, Stiller CA, Walker L et al, 2006,, 2006, Syndromes and constitutional chromosomal abnormalities associated with Wilms tumour. J Med Genet 43:705–715 Jenkins ZA, vanKogelenberg M, Morgan T et al, 2009, Germline mutations in WTX cause a sclerosing skeletal dysplasia but do not predispose to tumorigenesis. Nat Genet 41:95–100 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 189 EP 191 DI 10.1007/s12253-009-9205-6 PG 3 ER PT J AU Papp, A Cseke, L Farkas, R Pavlovics, G Horvath, G Varga, G Szigeti, A Bellyei, Sz Marton, S Poto, L Kalmar, K Vereczkei, A Pozsgai, E Horvath, PO AF Papp, Andras Cseke, Laszlo Farkas, Robert Pavlovics, Gabor Horvath, Gabor Varga, Gabor Szigeti, Andras Bellyei, Szabolcs Marton, Sandor Poto, Laszlo Kalmar, Katalin Vereczkei, Andras Pozsgai, Eva Horvath, Peter Ors TI Chemo-radiotherapy in Locally Advanced Squamous Cell Oesophageal Cancer—are Upper Third Tumours more Responsive? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Complete response; Locally advanced tumor; Neoadjuvant therapy; Squamous cell oesophageal cancer ID Complete response; Locally advanced tumor; Neoadjuvant therapy; Squamous cell oesophageal cancer AB Before neoadjuvant therapy was widely applied, the prognosis of oesophageal cancer had been considered dependent on the location of the tumor, i.e. upper third cancers had had the worst prognosis. The aim of this retrolective study was to prove the efficiency of the neoadjuvant treatment, and to compare the response of esophageal cancer in different locations. Between January 1998 and September 2005, 102 patients with locally advanced squamous cell oesophageal cancer received preoperative chemo-radiotherapy. In 40 cases the tumor was located in the upper third and in 62 cases in the middle third of the oesophagus. After a four-week-long treatment free period restaging was carried out and patients considered resectable were submitted to surgery. From 40 patients with upper third oesophageal cancer 28 underwent oesophageal resection or pharyngo-laryngectomy. Thiry-five percent a complete histopathological remission was observed. From 62 patients with middle third oesophageal cancer 43 underwent oesophageal resection. Histological examination of the resected specimens documented complete response only in three patients. The median survival and the R0 resection rate were similar in the two groups. Although the resection rate, perioperative morbidity, mortality and the median survival were similar in the two groups, a significantly higher rate of complete response (p<0,05) was observed in patients with upper third oesophageal cancer compared to patients with middle third oesophageal cancer. It seems that upper third oesophageal cancer has superior sensitivity to multimodal treatment therefore our results may support that upper third location is not an unfavorable prognostic factor any more. C1 [Papp, Andras] University of Pecs, Department of Surgery, Ifjusag u.13, H-7624 Pecs, Hungary. [Cseke, Laszlo] University of Pecs, Department of Surgery, Ifjusag u.13, H-7624 Pecs, Hungary. [Farkas, Robert] University of Pecs, Department of Oncology, Edesanyak utja 17, H-7624 Pecs, Hungary. [Pavlovics, Gabor] University of Pecs, Department of Surgery, Ifjusag u.13, H-7624 Pecs, Hungary. [Horvath, Gabor] Tawam Medical Center, The Johns Hopkins Medicine International, Department of Radiation Oncology, Al Ain, 1 Tawam Street, 15258 Abu Dhabi, United Arab Emirates. [Varga, Gabor] University of Pecs, Department of Surgery, Ifjusag u.13, H-7624 Pecs, Hungary. [Szigeti, Andras] University of Pecs, Department of Oncology, Edesanyak utja 17, H-7624 Pecs, Hungary. [Bellyei, Szabolcs] University of Pecs, Department of Oncology, Edesanyak utja 17, H-7624 Pecs, Hungary. [Marton, Sandor] University of Pecs, Medical Faculty, Department of Anesthesia and Intensive Therapy, Ifjusag u.13, H-7624 Pecs, Hungary. [Poto, Laszlo] Pecsi Tudomanyegyetem, Gyogyszereszeti Intezet, Honved utca 1, H-7624 Pecs, Hungary. [Kalmar, Katalin] University of Pecs, Department of Surgery, Ifjusag u.13, H-7624 Pecs, Hungary. [Vereczkei, Andras] University of Pecs, Department of Surgery, Ifjusag u.13, H-7624 Pecs, Hungary. [Pozsgai, Eva] University of Pecs, Department of Oncology, Edesanyak utja 17, H-7624 Pecs, Hungary. [Horvath, Peter Ors] University of Pecs, Department of Surgery, Ifjusag u.13, H-7624 Pecs, Hungary. RP Papp, A (reprint author), University of Pecs, Department of Surgery, H-7624 Pecs, Hungary. EM andras.papp@aok.pte.hu CR Lerut T, Coosemans W, Decker G, De Leyn P, Nafteux Ph, Van Raemdonck D, 2001, Cancer of the esophagus and gastroesophageal junction: potentially curative therapies Surg Oncol 10:113–122 Keighley MRB, 2003, Gastrointestinal cancers in Europe. Aliment Pharmacol Ther 18(Suppl. 3):7–30 Siewert JR, Stein HJ, Feith M, Bruecher BL, Bartels H, Fink U, 2001, Histologic tumor type is an independent prognostic parameter in esophageal cancer: lessons from more than 1,000 consecutive resections at a single center in the Western world. Ann Surg. 234(3):360–367 discussion 368–9 Law S, Kwong DL, Kwok KF, Wong KH, Chu KM, Sham JS, Wong J, 2003, Improvement in treatment results and long-term survival of patients with esophageal cancer: impact of chemoradiation and change in treatment strategy. Ann Surg. 238(3):339– 347 discussion 347–8 P. Sorrentino et al., 1988, Prognostic Significance of Tumor Stage and Lymph Node Involvement in Thoracic Esophageal Cancer. In: J.R. Siewert, A.H.Holscher, ed, Diseases of the esophagus Springer-Verlag Berlin Heidelberg, pp:709–713, Piessen G, Mariette C, Triboulet JP, 2005, Cervical and upperthird thoracic oesophageal carcinoma: a single pathological entity? Ann Chir. 130(2):86–91 Feinstein AR, 1985, Clinical epidemiology. Saunders, Philadelphia Horvath OP, Cseke L, Kalmar K, Varga G, Horvath G, 2001, Larynx-preserving pharyngo-esophagectomy after chemoradiation in the treatment of cancer of the pharyngo-esophageal junction. Ann Thorac Surg. 72(6):2146–2147 Pavlovics G, Cseke L, Papp A, Tizedes G, Tabar BA, Horvath PO, 2006, Esophagus reconstruction with free jejunal transfer. Microsurgery 26(1):73–77 Wong R, Malthaner R, 2003, Combined chemotherapy and radiotherapy, without surgery, compared with radiotherapy alone in localised carcinoma of the esophagus, Cochrane Review), in the Cochrane Library, Issue 1. Oxford, Update Softwer Mariette C, Piessen G, Triboulet JP, 2007, Therapeutic strategies in oesophageal carcinoma: role of surgery and other modalities. Lancet Oncol 8:545–553 El Nakadi I, Van Laethem JL, Houben JJ, Gay F, Closset J, Van Houtte P, Danhier S, Limbosch JM, Lambilliotte JP, Gelin M, 2002, Squamous cell carcinoma of the esophagus: multi-modal therapy in locally advanced disease. World J Surg 26, 1):72–78 Meneu-Diaz JC, Blazquez LA, Vicente E, Nuno J, Quijano Y, Lopez-Hervas P, Devesa M, Fresneda V, 2000, The role of multimodality therapy for resectable esophageal cancer. Am J Surg 179(6):508–513 Bosset JF, Gignoux M, Triboulet JP, Tiret E, Mantion G, Elias D, Lozach P, Ollier JC, Pavy JJ, Mercier M, Sahmoud T, 1997, Chemoradiotherapy followed by surgery compared with surgery alone in squamous-cell cancer of the esophagus. N Engl J Med 337(3):161–167 Visser BC, Venook AP, Patti MG, 2003, Adjuvant and neoadjuvant therapy for esophageal cancer: a critical reappraisal. Surg Oncol 12(1):1–7 Urschel JD, Vasan H, 2003, A meta-analysis of randomized controlled trials that compared neoadjuvant chemoradiation and surgery to surgery alone for resectable esophageal cancer. Am J Surg 185(6):538–543 Kaklamanos IG, Walker GR, Ferry K, Franceschi D, 2003, Neoadjuvant treatment for resectable cancer of the esophagus and the gastroesophageal junction: a meta-analysis of randomized clinical trials. Ann Surg Oncol 10:754–761 Greer SE, Goodney PP, Sutton JE, Birkmeyer JD, 2005, Neoadjuvant chemoradiotherapy for esophageal carcinoma:a metaanalysis. Surgery 137:172–177 Gebski V, Burmeister B, Smithers BM, Foo K, Zalcberg J, Simes J, 2007, Survival benefits from neoadjuvant chemoradiotherapy or chemotherapy in oesophageal carcinoma: a meta-analysis. Lancet Oncol 8:226–234 Urba SG, Orringer MB, Turrisi A et al., 2001, Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol 19:305– 313 Berger AC, Farma J, Scott WJ et al., 2005, Complete response to neoadjuvant chemoradiotherapy in esophageal carcinoma is associated with significantly improved survival. J Clin Oncol 23:4330–4337 Saito R, Suzuki H, Motoyama S, Sasaki S, Okuyama M, Ogawa J, Kitamura M, 2000, A clinical study of surgical treatment of patients with carcinoma of the cervical esophagus extending to the thoracic esophagus. Jpn J Thorac Cardiovasc Surg 48(7):417–423 Peracchia A, Bonavina L, Botturi M, Pagani M, Via A, Saino G, 2001, Current status of surgery for carcinoma of the hypopharynx and cervical esophagus. Dis Esoph 14:95–97 Liebermann-Meffert DM, Luescher U, Neff U, Ruedi TP, Allgower M, 1987, Esophagectomy without thoracotomy: is there a risk of intramediastinal bleeding? A study on blood supply of the esophagus. Ann Surg 206(2):184–192 Stahl M, Stuschke M, Lehmann N, Meyer HJ, Walz MK, Seeber S, Klump B, Budach W, Teichmann R, Schmitt M, Schmitt G, Franke C, Wilke H, 2005, Chemoradiation with and whitout surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 23(10):2310–2317 Bedenne L, Michel P, Bouche O et al., 2002, Randomised phase III trial in locally advanced esophageal cancer: Radiochemotherapy followed by surgery versus radiochemotherapy alone, FFCD 9102). Proc Am Soc Clin Oncol 21:130a abstr 519 Burmeister B, Dickie G, Smithers B, Hodge R, Morton K, 2000, Thirty-four patients with carcinoma of the cervical esophagus treated with chemoradiation therapy. Arch Otolaryngol Head Neck Surg 126:205–208 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 193 EP 200 DI 10.1007/s12253-009-9206-5 PG 8 ER PT J AU Szynglarewicz, B Matkowski, R Halon, A Lacko, A Stepien, M Forgacz, J Pudelko, M Kornafel, J AF Szynglarewicz, Bartlomiej Matkowski, Rafal Halon, Agnieszka Lacko, Aleksandra Stepien, Marcin Forgacz, Jozef Pudelko, Marek Kornafel, Jan TI Association Between Histological Type of Tumour Growth and Patient Survival in T2-T3 Lymph Node-Negative Rectal Cancer Treated with Sphincter-Preserving Total Mesorectal Excision SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Anterior resection; Invasive margin character; Lymphocytic infiltration; Rectal cancer; Total mesorectal excision ID Anterior resection; Invasive margin character; Lymphocytic infiltration; Rectal cancer; Total mesorectal excision AB For rectal cancer patients without nodal metastases the identification of unfavourable factors can be helpful for the better selection for adjuvant therapy and multimodality treatment. The aim of this study was to evaluate the impact of clinico-histological parameters on prognosis in node-negative rectal cancer patients. One hundred and thirty-nine consecutive node negative rectal cancer patients with complete five-year follow-up were studied prospectively. All of them underwent curative anterior resection with total mesorectal excision technique. Seventy-eight patients with tumour penetration beyond the bowel wall received neo-adjuvant short-course radiation (25 Gy) followed by surgery within 1 week and postoperative chemotherapy with 5-fluorouracil and folinic acid in six cycles or adjuvant radiochemotherapy: irradiation (50.4 Gy) combined with chemotherapy (as above). Cancer-specific survival was calculated according to the Kaplan-Meier method. Variables significant in univariate analysis by logrank test (P<0.05) entered the Cox proportional hazard model. Survival was decreased for males, older patients (>60 years) with extraperitoneal, poorly differentiated cancers, tumours with mucinous histology and with the absence of lymphocytic infiltration but with the lack of statistical importance. Prognosis was significantly improved for patients with T2 tumours versus T3 (P<0.01) and with cancers with expanding growth comparing to diffusely infiltrating ones (P<0.01). In multivariate analysis these parameters significantly and independently influenced survival (P<0.01 and P<0.05, respectively). Diffusely infiltrating growth of tumour can reflect the more aggressive cancer behaviour and unfavourable course of disease despite the optimised local control. Apart from the extent of tumour penetration the type of invasive margin can be an additional parameter helpful for the optimal treatment planning and better patient selection for postoperative chemotherapy. C1 [Szynglarewicz, Bartlomiej] Wroclaw Medical University, Lower Silesian Oncology Center, 2nd Department of Surgical Oncology, Plac Hirszfelda 12, 53-413 Wroclaw, Poland. [Matkowski, Rafal] Wroclaw Medical University, Lower Silesian Oncology Center, 2nd Department of Surgical Oncology, Plac Hirszfelda 12, 53-413 Wroclaw, Poland. [Halon, Agnieszka] Wroclaw Medical University, Department of PathologyWroclaw, Poland. [Lacko, Aleksandra] Wroclaw Medical University, Department of OncologyWroclaw, Poland. [Stepien, Marcin] Wroclaw Medical University, Department of OncologyWroclaw, Poland. [Forgacz, Jozef] Wroclaw Medical University, Lower Silesian Oncology Center, 2nd Department of Surgical Oncology, Plac Hirszfelda 12, 53-413 Wroclaw, Poland. [Pudelko, Marek] Wroclaw Medical University, Lower Silesian Oncology Center, 2nd Department of Surgical Oncology, Plac Hirszfelda 12, 53-413 Wroclaw, Poland. [Kornafel, Jan] Wroclaw Medical University, Department of OncologyWroclaw, Poland. RP Matkowski, R (reprint author), Wroclaw Medical University, Lower Silesian Oncology Center, 2nd Department of Surgical Oncology, 53-413 Wroclaw, Poland. EM matkowski.r@dco.com.pl CR Heald RJ, Husband EM, Ryall RDH, 1982, The mesorectum in rectal cancer surgery- the clue to pelvic recurrence? Br J Surg 69:613–616 Dorudi S, Steele RJ, McArdle CS, 2002, Surgery for colorectal cancer. Br Med Bull 64:101–118 Cecil TD, Sexton R, Moran BJ, Heald RJ, 2004, Total mesorectal excision results in low local recurrence rates in lymph nodepositive rectal cancer. Dis Colon Rectum 47:1145–1149 Colquhoun P, Wexner SD, Cohen A, 2003, Adjuvant therapy is valuable in the treatment of rectal cancer despite total mesorectal excision. J Surg Oncol 83:133–139 Merchant NB, Guillem JG, Paty PB et al, 1999, T3N0 rectal cancer: results following sharp mesorectal excision and no adjuvant therapy. J Gastrointest Surg 3:642–647 Price T, Pittman K, Patterson W et al, 2008, Management and survival trends in colorectal cancer. Clin Oncol 20:626–630 Jass JR, Love S, Northover JM, 1987, A new prognostic classification for rectal cancer. Lancet 1:1303–1306 Jass JR, O’Brien MJ, Riddel HR et al, 2007, Recommendations for the reporting of surgically resected specimens of colorectal carcinoma. Virchows Arch 450:1–13 Szynglarewicz B, Matkowski R, Forgacz J et al, 2007, Clinical factors in prediction of prognosis after anterior resection with total mesorectal excision for carcinoma of the rectum. Oncol Rep 17:471–476 Katsumata D, Fukui H, Ono Y et al, 2008, Depth of tumor invasion in locally advanced rectal cancer correlates with patient’s prognosis: the usefulness of elastic stain for its measurement. Surg Today 38:115–122 Bori R, Sejben I, Svebis M et al, 2009, Heterogeneity of pT3 colorectal carcinomas according to the depth of invasion. Pathol Oncol Res, in press). doi 10.1007/s12253-009-9149-x Miyoshi M, Ueno H, Hashiguchi Y et al, 2006, Extent of mesorectal tumor invasion as a prognostic factor after curative surgery for T3 rectal cancer patients. Ann Surg 243:492–498 Yoshida K, Yoshimatsu K, Otani T et al, 2008, The depth of tumor invasion beyond the outer border of the muscularis propria as a prognostic factor for T3 rectal/rectosigmoid cancer. Anticancer Res 28:1773–1778 Szynglarewicz B, Matkowski R, Maciejczyk A et al, 2008, Combined-modality therapy with sphincter-preserving total mesorectal excision for locally advanced rectal cancer: patient’s age and long-term outcome. J Gastrointestin Liver Dis 17:49–52 Compton CC, 2003, Colorectal carcinoma: Diagnostic, Prognostic, and Molecular Features. Mod Pathol 16:376–388 Nagtegaal I, Gaspar C, Marijnen C et al, 2004, Morphological changes in tumour type after radiotherapy are accompanied by changes in gene expression profile but not in clinical behaviour. J Pathol 204:183–292 Sternberg A, Mizrahi A, Amar M, Groisman G, 2006, Detection of venous invasion in surgical specimens of colorectal carcinoma: the efficacy of various types of tissue blocks. J Clin Pathol 59:207–210 Harrison JC, Dean PJ, el-Zeky F et al, 1994, From Dukes to Jass: pathological prognostic indicators in rectal cancer. Hum Pathol 25:498–505 Quirke P, Dixon MF, Clayden AD et al, 1987, Prognostic significance of DNA aneuploidy and cell proliferation in rectal adenocarcinomas. J Pathol 151:285–291 Scott NA, Rainwater LM, Wieand HS et al, 1987, The relative prognostic value of flow cytometric DNA analysis and conventional clinicopathologic criteria in patients with operable rectal carcinoma. Dis Colon Rectum 30:513–520 Secco GB, Fardelli R, Campora E et al, 1990, Prognostic value of Jass’ histological classification in left colon and rectal cancer: a multivariate analysis. Digestion 47:71–80 Ueno H, Jones A, Jass JR, Talbot IC, 2002, Clinicopathological significance of the “keloid-like” collagen and myxoid stroma in advanced rectal cancer. Histopathology 40:327–334 Cianchi F, Messerini L, Palomba A et al, 1997, Character of the invasive margin in colorectal cancer: does it improve prognostic information of Dukes staging? Dis Colon Rectum 40:1170–1175 Jimenez-Anula J, Luque RJ, Gaforio JJ, Delgado M, 2005, Prognostic value of invasive growth pattern in sporadic colorectal cancer. Cir Esp 77:337–342 Shimomura T, Ishiguro S, Konishi H et al, 2004, New indication for endoscopic treatment of colorectal carcinoma with submucosal invasion. J Gastroenterol Hepatol 19:48–55 De Quay N, Cerottini JP, Albe X et al, 1999, Prognosis in Dukes’ B colorectal carcinoma: the Jass classification revisited. Eur J Surg 65:588–592 Ilyas M, Novelli M, Wilkinson K et al, 1997, Tumour recurrence is associated with Jass grouping but not with differences in E-cadherin expression in moderately differentiated Dukes’ B colorectal cancers. J Clin Pathol 50:218–222 Gagliardi G, Stepniewska KA, Hershman MJ et al, 1995, New graderelated prognostic variable for rectal cancer. Br J Surg 82:599–602 Neoptolemos JP, Oates GD, Newbold KM et al, 1995, Cyclin/ proliferation cell nuclear antigen immunohistochemistry does not improve the prognostic power of Dukes’ or Jass’ classifications for colorectal cancer. Br J Surg 82:184–187 LiDestri G, Rinzivillo C, Vasquez E et al, 2001, Evaluation of the prognostic accuracy of Astler-Coller’s and Jass’ classifications of colorectal cancer. Tumori 87:127–129 Fisher ER, Robinsky B, Sass R, Fisher B, 1989, Relative prognostic value of the Dukes and the Jass systems in rectal cancer. Findings from the National Surgical Adjuvant Breast and Bowel Projects, Protocol R-01). Dis Colon Rectum 32:944–949 Deans GT, Heatley M, Anderson N et al, 1994, Jass’ classification revisited. J Am Coll Surg 179:11–17 Dundas SA, Laing RW, O’Cathain A et al, 1988, Feasibility of new prognostic classification for rectal cancer. J Clin Pathol 41:1273–1276 Jass JR, Ajioka Y, Allen JP et al, 1996, Assessment of invasive growth pattern and lymphocytic infiltration in colorectal cancer. Histopathology 28:543–548 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 201 EP 206 DI 10.1007/s12253-009-9207-4 PG 6 ER PT J AU Olasz, L Szalma, J Orsi, E Tornoczky, T Marko, T Nyarady, Z AF Olasz, Lajos Szalma, Jozsef Orsi, Eniko Tornoczky, Tamas Marko, Tamas Nyarady, Zoltan TI Neoadjuvant Chemotherapy: Does It Have Benefits for the Surgeon in the Treatment of Advanced Squamous Cell Cancer of the Oral Cavity? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE OSCC; Neoadjuvant chemotherapy; Surgical margin; Primary tumor regression; Oral cancer ID OSCC; Neoadjuvant chemotherapy; Surgical margin; Primary tumor regression; Oral cancer AB The purpose of this clinicopathological study was to evaluate the effects and efficiency of combined neoadjuvant chemotherapy related to surgical margin. 100 consecutively treated squamous cell cancer patients receiving a combined neoadjuvant therapy were selected (Bleomycin—Vincristin—Methotrexate (BVM) or BVM + Mitolactol or BVM + Cisplatin). After three courses of chemotherapy, the patients were operated on. The largest diameter of the primary tumors was compared before and after chemotherapy. In the surgical specimen, the involvement of surgical margin was assessed. The largest diameter before chemotherapy was: T2 30%; T3 55%; T4A 15%. After chemotherapy, the rest tumor was assessed in the surgical specimen as: no rest 11%; <2 cm 57%; 2–4 cm 28%; 4–6 cm 4%. The no rest and <2 cm (optimal operability) tumor was observed in T2: 94%; in T3: 73%; in the T4A: 0%. Severe side effects (Grade III–IV) were not observed. There was a significant decrease in size (P<0.0001). Of the 100 surgical specimens, 83% had clear-, 9% close- and 8% involved margins. From T4A, there was a 40% (6 patients) involved margin. Based on the significantly better size and operability of primary T2-3, the mild side effects and the high (83%) percentage of clear surgical margins, that is better than other (without preoperative chemotherapy) results, sought the use of chemotherapy is recommended before surgery. Due to the 40% involved margin, we don’t suggest surgery in T4A. C1 [Olasz, Lajos] University of Pecs, Department of Oral and Maxillofacial Surgery, Dischka Gy. u.5., 7621 Pecs, Hungary. [Szalma, Jozsef] University of Pecs, Department of Oral and Maxillofacial Surgery, Dischka Gy. u.5., 7621 Pecs, Hungary. [Orsi, Eniko] University of Pecs, Department of Oral and Maxillofacial Surgery, Dischka Gy. u.5., 7621 Pecs, Hungary. [Tornoczky, Tamas] University of Pecs, Department of Pathology, Szigeti Str. 12, 7624 Pecs, Hungary. [Marko, Tamas] University of Pecs, Department of Informatics, Szigeti Str. 12, 7624 Pecs, Hungary. [Nyarady, Zoltan] University of Pecs, Department of Oral and Maxillofacial Surgery, Dischka Gy. u.5., 7621 Pecs, Hungary. RP Olasz, L (reprint author), University of Pecs, Department of Oral and Maxillofacial Surgery, 7621 Pecs, Hungary. EM lajos.olasz@aok.pte.hu CR Gaudi I, Kasler M, 2002, The course of cancer mortality in Hungary between 1975–2001, English abstract). Magy Onkol 46:291–295 Zs N, Velich N, Bogdan S et al, 2005, The prognostic role of clinical, morphological and molecular markers in oral squamosus cell tumors. Neoplasma 52:95–101 Taylor SG, 1997, Head and neck cancer. Cancer Chemot Biol Response Mod Ann 17 Elsev Science Gibson MK, Forastiere AA, 2004, Multidisciplinary approaches in the management of advanced head and neck tumors: state of art. Curr Opin Oncol 16:220–224 Forastiere AA, 2004, Is there a new role for induction chemotherapy in the treatment of head and neck cancer? J Natl Cancer Inst 96:1647–1649 Gibson MK, Forestiere AA, 2006, Reassassment of the role of induction chemotherapy for head and neck cancer. Lancet Oncol 7:565–574 Bryne M, Koppang HS, Lilleng R et al, 1992, Malignancy grading of the deep invasive margins of oral squamous cell carcinomas has high prognostic value. J Pathol 166:375–381 Spiro RH, Guillamondegui O, Paulin AF et al, 1999, Pattern of invasion and margin assessment in patients with oral tongue cancer. Head Neck 21:408–413 Cooper JS, Pajak TF, Forastiere AA et al, 2004, Radiation Therapy Oncology Group 9501/Intergroup. Postoperative concurrent radiotherapy and chemotherapy for high-risk squamous-cell carcinoma of the head and neck. N Engl J Med 350:1937–1944 Loree TR, Strong EW, 1990, Significance of positive margins in oral cavity squamous carcinoma. Am J Surg 160:410–414 Sutton DN, Brown JS, Rogers SN et al, 2003, The prognostic implications of the surgical margin in oral squamous cell carcinoma. Int J Oral Maxillofac Surg 32:30–34 Beitler JJ, Smith RV, Silver CE et al, 1998, Close or positive margins after surgical resection for the head and neck cancer patient: the addition of brachytherapy improves local control. Int J Radial Oncol Biol Phys 40:313–317 Jacobs JR, Ahmad K, Casiano R et al, 1993, Implications of positive surgical margins. Laryngoscope 103:64–68 Batsakis JG, 1999, Surgical excision margins: a pathologist’s perspective. Adv Anat Pathol 6:140–148 Woolgar JA, Rogers S, West CR et al, 1999, Survival and patterns of recurrence in 200 oral cancer patients treated by radical surgery and neck dissection. Oral Oncol 35:257–265 Licitra L, Grandi C, Guzzo M et al, 2003, Primary chemotherapy in resectable oral cavity squamous cell cancer: a randomized controlled trial. J Clin Oncol 21:327–333 Bettendorf O, Piffko J, Bankfalvi A, 2004, Prognostic and predictive factors in oral squamous cell cancer: important tools for planning individual therapy? Oral Oncol 40:110–119 Beckmann GK, Hoppe F, Pfreundner L et al, 2005, Hyperfractionated accelerated radiotherapy in combination with weekly cisplatin for locally advanced head and neck cancer. Head Neck 27:36–43 van Es RJJ, van Nieuw-Amerongen N, Slootweg PJ et al, 1996, Resection margin as a predictor or recurrence at the primary site for T1 and T2 oral cancers. Arch Otolaryngol Head Neck Surg 122:521–525 Perez CA, Marks J, Powers W, 1977, Preoperative irradiation in head and neck cancer. Semin Oncol 4:387–397 Stevens KR, Moss WT, 1975, Control of localised cancer. Combination of preoperative irradiation and surgery. JAMA 232:1158–1160 Carifi VG, Ohanion ML et al, 1974, Results of high dose radiation and surgery in the treatment of advanced cancer of the head and neck. Am J Surg 128:580–582 Mohr C, Bohndorf W, Carstens J et al, 1994, Preoperative radiochemotherapy and radical surgery in comparison with radical surgery alone. A prospective multicentric randomised DOSAK study of advanced squamous cell carcinoma of the oral cavity and the oropharynx, a 3-year follow up). Int J Oral Maxillofac Surg 23:140–148 Volling P, Schroder M, Muller RP et al, 1994, Induction chemotherapy in primary resectable head and neck tumors: a prospective randomized trial. Int J Oncol 4:909–914 Jakobs C, Makuch R, 1999, Efficacy of adjuvant chemotherapy for patients with resectable head and neck cancer: a subset analysis of the head and neck contracts program. J Clin Oncol 8:838–847 Maipang T, Maipang M, Geater A et al, 1995, Combination chemotherapy as induction therapy for advanced resectable head and neck cancer. J Surg Oncol 59:80–85 Olasz L, Kwashie F, Herczegh P et al, 1996, A comparative study of preoperative B-V-M-M chemotherapy and irradiation in advanced squamous cell cancer of the oral cavity. Neoplasma 43:51–56 Kovacs AF, Turowski B, Ghahremani MT et al, 1999, Intraarterial chemotherapy as neoadjuvant treatment of oral cancer. J Craniomaxillofac Surg 27:302–307 AlSarraf M, Martz K, Herskovic A et al, 1997, Progress report of combined chemoradiotherapy versus radiotherapy alone in patients with esophageal cancer: an intergroup study. J Clin Oncol 15:277–284 Benasso M, Lionetto R, Corvo R et al, 2003, Impact of the treating institution on the survival of patients with head and neck cancer treated with concomitant alternating chemotherapy and radiation. Eur J Cancer 39:1895–1898 Hironaka S, Ohtsu A, Boku N et al, 2003, Nonrandomised comparison between definitive chemoradiotherapy and radical surgery in patients with T 2–3 Nany Mo squamous cell carcinoma of the esophagus. Int J Rad Oncol Biol Phys 57:425–433 Psyrri A, Kwong M, DiStasio S et al, 2004, Cisplatin, fluorouracil, and leucovorin induction chemotherapy followed by concurrent cisplatin chemoradiotherapy for organ preservation and cure in patients with advanced head and neck cancer: longterm follow-up. J Clin Oncol 22:3061–3069 Forastiere AA, Trotti A, 1999, Radiotherapy and concurrent chemotherapy: a strategy that improves locoregional contro and survival in oropharyngeal cancer. J Natl Cancer Inst 91:2056–2066 Basu S, Khanra M, Dash B et al, 1999, The role of neoadjuvant and adjuvant chemotherapy regimens consisting of different combinations of drugs in the treatment of advanced oral cancer. Med Oncol 16:199–203 Olasz L, Szabo I, Horvath A, 1988, A combined treatment for advanced oral cavity cancers. Cancer 62:1267–1274 Price LA, Hill BT, 1986, Impact of primary site Stage III and IV squamous cell carcinomas of the head and neck on 7-year survival figures following initial noncisplatin-containing combination chemotherapy. Resent Results Cancer Res 103:124–134 Athanasiadis J, Taylor S, Vokes EE et al, 1997, Phase II. study of induction and adjuvant chemotherapy for squamous cell carcinoma of the head and neck. A long-term analysis for the Illinois Cancer Center. Rec Res Cancer 79:588–594 Gregoire V, Beauduin M, Humblet Y et al, 1991, A phase I–II trial of induction chemotherapy with carboplatin and fluorouracil in locally advanced head and neck squamous cell carcinoma: report from the UCL-Oncology Group, Belgium. J Clin Oncol 9:1385–1392 Gibson MK, Li Y, Murphy B et al, 2005, Eastern Cooperative Oncology Group. Randomized phase III evaluation of cisplatin plus fluorouracil versus cisplatin plus paclitaxel in advanced head and neck cancer, E1395): an intergroup trial of the Eastern Cooperative Oncology Group. J Clin Oncol 23:3562–3567 Braun OM, Neumeister B, Neuhold N et al, 1989, Histological grading of therapy induced regression in squamous cell carcinomas of the oral cavity. A morphological and immunohistochemical study. Path Res Pract 185:368–372 Sulfaro S, Frustaci S, Volpe R et al, 1989, A pathologic assessment of residue and stromal changes after intra-arterial chemotherapy for head and neck carcinomas. Cancer 64:994– 1001 Suba Z, Szabo G, Barabas J et al, 1992, Histological evaluation of chemotherapeutically induced regression. Reg Cancer Treat 4:313–319 Olasz L, Nemeth A, Nyarady Z et al, 2004, Results and failures with or without cisplatin containing induction chemotherapy in the treatment of squamous cell carcinoma of the head and neck. Cancer Det Prev 28:65–71 Giralt JL, Gonzalez J, del Campo JM et al, 2000, Preoperative induction chemotherapy followed by concurrent chemoradiotherapy in advanced carcinoma of the oral cavity and oropharynx. Cancer 89:939–945 Posner MR, Glisson B, Frenette G et al, 2001, Multicenter phase I–II trial of docetaxel, cisplatin and fluorouracil induction chemotherapy for patients with locally advanced squamous cell cancer of the head and neck. J Clin Oncol 19:1096–1104 Woolgar JA, Rogers SN, Lowe D et al, 2003, Cervical lymph node metastasis in oral cancer: the importance of even microscopic extracapsular spread. Oral Oncol 39:130–137 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 207 EP 212 DI 10.1007/s12253-009-9208-3 PG 6 ER PT J AU Urbanowicz, I Mazur, G Stacherzak-Pawlik, J Bogunia-Kubik, K Wrobel, T Wozniak, M Kuliczkowski, K AF Urbanowicz, Iwona Mazur, Grzegorz Stacherzak-Pawlik, Jolanta Bogunia-Kubik, Katarzyna Wrobel, Tomasz Wozniak, Mieczyslaw Kuliczkowski, Kazimierz TI IFN Gamma Gene Polymorphism May Contribute to the Susceptibility to CLL SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chronic lymphocytic leukemia; Gene polymorphism; INF gamma ID Chronic lymphocytic leukemia; Gene polymorphism; INF gamma AB The pathogenesis of B-cell chronic lymphocytic leukemia (B-CLL) has been linked with the production and activity of certain growth factors. However a significant proportion of CLL patients display immune abnormalities suggestive of aberrant cytokine secretion and/or response. In contrast to B lymphocytes, T cells of B-CLL patients characterise with the increased production of interferongamma (IFN-γ) and this cytokine has been indicated to prevent malignant cells from entering apoptosis including the slowly expanding population of CD5+ B cells that characterizes chronic lymphocytic leukemia. The aim of the present study was to assess whether functionally relevant interferon-gamma gene (IFNG) polymorphism (+847 A/T) contributes to the pathogenesis of B-CLL. In total 110 individuals was investigates, including 61 CLL patients and 50 healthy individuals. The presence of the IFNG AA genotype was found to be associated with susceptibility to CLL (23/61 vs. 7/50, p<0.005, for patients and controls, respectively). This results suggest that individuals rather prone to the lower level of IFN-γ production (associated with the presence of the A allele) appear to be more susceptible to this malignant disease. C1 [Urbanowicz, Iwona] Wroclaw Medical University, Department of Clinical Chemistry, Pasteur 2 Str, 50-367 Wroclaw, Poland. [Mazur, Grzegorz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow TransplantationWroclaw, Poland. [Stacherzak-Pawlik, Jolanta] Wroclaw Medical University, Department of Clinical Chemistry, Pasteur 2 Str, 50-367 Wroclaw, Poland. [Bogunia-Kubik, Katarzyna] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental TherapyWroclaw, Poland. [Wrobel, Tomasz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow TransplantationWroclaw, Poland. [Wozniak, Mieczyslaw] Wroclaw Medical University, Department of Clinical Chemistry, Pasteur 2 Str, 50-367 Wroclaw, Poland. [Kuliczkowski, Kazimierz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow TransplantationWroclaw, Poland. RP Urbanowicz, I (reprint author), Wroclaw Medical University, Department of Clinical Chemistry, 50-367 Wroclaw, Poland. EM IwonaUrb@yahoo.com CR Abbott BL, 2004, Advances in the diagnosis and treatment of chronic lymphocytic leukemia. Clin Adv Hematol Oncol 2:448–454 Zenz T, Mertens D, Dohner H, Stilgenbauer S, 2008, Molecular diagnostics in chronic lymphocytic leukemia—Pathogenetic and clinical implications. Leuk Lymphoma 15:1–10 Mainou-Fowler T, Prentice AG, 1996, Modulation of apoptosis with cytokines in B-cell chronic lymphocytic leukemia. Leuk Lymphoma 21:369–377 Pravica V, Perrey C, Stevens A et al, 2000, A single nucleotide polymorphism in the first intron of the human IFN-gamma gene: absolute correlation with a polymorphic CA microsatellite marker of high IFN-gamma production. Hum Immunol 61:863–866 Pravica V, Asderakis A, Perrey C et al, 1999, In vitro production of IFN-gamma correlates with CA repeat polymorphism in the human IFN-gamma gene. Eur J Immunogenet 26:1–3 Perrey C, Pravica V, Sinnott PJ, Hutchinson IV, 1998, Genotyping for polymorphisms in interferon-gamma, interleukin-10, transforming growth factor-beta 1 and tumour necrosis factoralpha genes: a technical report. Transpl Immunol 6:193–197 Bogunia-Kubik K,Mazur G, Urbanowicz I,Wrobel T, Kuliczkowski K, Wozniak M, Lange A, 2006, Lack of association between the TNF-alpha promoter gene polymorphism and susceptibility to B-cell chronic lymphocytic leukemia. Int J Immunogenet 33:21–24 Kiaii S, Choudhury A, Mozaffari F, Rezvany R, Lundin J, Mellstedt H, Osterborg A, 2006, Signaling molecules and cytokine production in T cells of patients with B-cell chronic lymphocytic leukemia: long-term effects of fludarabine and alemtuzumab treatment. Leuk Lymphoma 47:1229–1238 Kiaii S, Choudhury A, Mozaffari F, Kimby E, Osterborg A, Mellstedt H, 2005, Signaling molecules and cytokine production in T cells of patients with B-cell chronic lymphocytic leukemia, B-CLL): comparison of indolent and progressive disease. Med Oncol 22:291–302 Rossmann ED, Lewin N, Jeddi-Tehrani M, Osterborg A, Mellstedt H, 2002, Intracellular T cell cytokines in patients with B cell chronic lymphocytic leukaemia, B-CLL). Eur J Haematol 68:299– 306 Podhorecka M, Dmoszynska A, Rolinski J, 2004, Intracellular IFN-gamma expression by CD3+/CD8+ cell subset in B-CLL patients correlates with stage of the disease. Eur J Haematol 73:29–35 Totero D, Tazzari PL, Capaia M, Montera MP, Clavio M, Balleari E, Foa R, Gobbi M, 2003, CD40 triggering enhances fludarabineinduced apoptosis of chronic lymphocytic leukemia B-cells through autocrine release of tumor necrosis factor-alpha and interferon-gama and tumor necrosis factor receptor-I-II upregulation. Haematologica 88:148–158 Gallego A, Vargas JA, Castejon R, Citores MJ, Romero Y, Millan I, Durantez A, 2003, Production of intracellular IL-2, TNF-alpha, and IFN-gamma by T cells in B-CLL. Cytometry B Clin Cytom 56:23–29 Warle MC, Farhan A, Metselaar HJ et al, 2003, Are cytokine gene polymorphisms related to in vitro cytokine production profiles? Liver Transpl 9:170–181 Vandenbroeck K, Goris A, 2003, Cytokine gene polymorphisms in multifactorial diseases: gateways to novel targets for immunotherapy? Trends Pharmacol Sci 24:284–289 Zaki M, Douglas R, Patten N, Bachinsky M, Lamb R, Nowell P, Moore J, 2000, Disruption of the IFN-gamma cytokine network in chronic lymphocytic leukemia contributes to resistance of leukemic B cells to apoptosis. Leuk Res 24:611–621 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 213 EP 216 DI 10.1007/s12253-009-9209-2 PG 4 ER PT J AU Chua, CT Yao, P Akther, J Morris, LD AF Chua, C Terence Yao, Peng Akther, Javed Morris, L David TI Impact of Tumor Angiogenesis in Peritoneal Mesothelioma After Radical Cytoreduction and Hyperthermic Intraperitoneal Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mesothelioma; Vascular Endothelial Growth Factor; CD 31 Antigen; Angiogenesis; Cytoreductive surgery ID Mesothelioma; Vascular Endothelial Growth Factor; CD 31 Antigen; Angiogenesis; Cytoreductive surgery AB Peritoneal mesothelioma is one of the peritoneal surface malignancies where long-term survival is a reality after cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). Tumor angiogenesis has been shown to be of prognostic significance on survival in mesothelioma. We investigated the impact of survival of patients with peritoneal mesothelioma following CRS and HIPEC to determine the impact of tumor angiogenesis on survival after this radical surgical treatment. Paraffin sections of 23 patients who were treated with CRS and HIPEC were retrieved for immunohistochemical analysis. The immunostaining was performed using monoclonal mouse anti-human antibodies (VEGF-C and CD31) on an autostainer (Autostainer Plus; Dako, Inc.). The intensity of the stains were quantified using the Image-Pro Plus (IPP) 4.5 (Media Cybernetics, Silver Spring, MD). VEGF expression and microvessel density (MVD) using CD31 staining were studied. The median survival was 94 months with a 3-year survival rate of 51%. There was no impact on patient’s age, sex, peritoneal cancer index, tumor histopathology and survival outcomes between patients with low or high MVD and VEGF expression. After CRS and HIPEC, our results demonstrate that the prognostic significance of tumor angiogenesis is negated, highlighting the potential importance of other co-contributory mechanisms in mesotheliomagenesis and undergoing radial treatment. C1 [Chua, C Terence] St George Hospital, University of New South Wales, Department of Surgery, Kogarah, NSW 2217 Sydney, Australia. [Yao, Peng] St George Hospital, University of New South Wales, Department of Surgery, Kogarah, NSW 2217 Sydney, Australia. [Akther, Javed] St George Hospital, University of New South Wales, Department of Surgery, Kogarah, NSW 2217 Sydney, Australia. [Morris, L David] St George Hospital, University of New South Wales, Department of Surgery, Kogarah, NSW 2217 Sydney, Australia. RP Morris, LD (reprint author), St George Hospital, University of New South Wales, Department of Surgery, NSW 2217 Sydney, Australia. EM david.morris@unsw.edu.au CR Chua TC, Yan TD, Morris DL, 2009, Surgical biology for the clinician: peritoneal mesothelioma: current understanding and management. Can J Surg 52(1):59–64 Musk AW, de Klerk NH, 2004, Epidemiology of malignant mesothelioma in Australia. Lung Cancer 45(Suppl 1):S21–S23 Edwards JG, Swinson DEB, Jones JL, Muller S, Waller DA, O’Byrne KJ, 2003, Tumor necrosis correlates with angiogenesis and is a predictor of poor prognosis in malignant mesothelioma. Chest 124(5):1916–1923 Masood R, Kundra A, Zhu S, Xia G, Scalia P, Smith DL et al, 2003, Malignant mesothelioma growth inhibition by agents that target the VEGF and VEGF-C autocrine loops. Int J Cancer 104, 5):603–610 Edwards JG, Cox G, Andi A, Jones JL, Walker RA, Walker DA et al, 2001, Angiogenesis is an independent prognostic factor in malignant mesothelioma. Br J Cancer 85(6):863–868 Demirag F, Unsal E, Yilmaz A, Caglar A, 2005, Prognostic significance of vascular endothelial growth factor, tumor necrosis, and mitotic activity index in malignant pleural mesothelioma. Chest 128(5):3382–3387 Kothmaier H, Quehenberger F, Halbwedl I, Morbini P, Demirag F, Zeren H et al, 2008, EGFR and PDGFR differentially promote growth in malignant epithelioid mesothelioma of short and long term survivors. Thorax 63(4):345–351 Strizzi L, Catalano A, Vianale G, Orecchia S, Casalini A, Tassi G et al, 2001, Vascular endothelial growth factor is an autocrine growth factor in human malignant mesothelioma. J Pathol 193, 4):468–475 Konig JE, Tolnay E, Wiethege T, Muller KM, 1999, Expression of vascular endothelial growth factor in diffuse malignant pleural mesothelioma. Virchows Arch 435(1):8–12 Ohta Y, Shridhar V, Bright RK, Kalemkerian GP, Du W, Carbone M et al, 1999, VEGF and VEGF type C play an important role in angiogenesis and lymphangiogenesis in human malignant mesothelioma tumours. Br J Cancer 81(1):54–61 Ellis LM, Fidler IJ, 1996, Angiogenesis and metastasis. Eur J Cancer 32A(14):2451–2460 Hicklin DJ, Ellis LM, 2005, Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol 23(5):1011–1027 Stacker SA, Achen MG, Jussila L, Baldwin ME, Alitalo K, 2002, Lymphangiogenesis and cancer metastasis. Nat Rev Cancer 2, 8):573–583 Jassem J, Ramlau R, Santoro A, Schuette W, Chemaissani A, Hong S et al, 2008, Phase III trial of pemetrexed plus best supportive care compared with best supportive care in previously treated patients with advanced malignant pleural mesothelioma. J Clin Oncol 26(10):1698–1704 van Meerbeeck JP, Gaafar R, Manegold C, Van Klaveren RJ, Van Marck EA, Vincent M et al, 2005, Randomized phase III study of cisplatin with or without raltitrexed in patients with malignant pleural mesothelioma: an intergroup study of the European Organisation for Research and Treatment of Cancer Lung Cancer Group and the National Cancer Institute of Canada. J Clin Oncol 23(28):6881–6889 Yan TD, Welch L, Black D, Sugarbaker PH, 2007, A systematic review on the efficacy of cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for diffuse malignancy peritoneal mesothelioma. Ann Oncol 18(5):827–834 Zellos L, Richards WG, Capalbo L, Jaklitsch MT, Chirieac LR, Johnson BE et al, 2009, A phase I study of extrapleural pneumonectomy and intracavitary intraoperative hyperthermic cisplatin with amifostine cytoprotection for malignant pleural mesothelioma. J Thorac Cardiovasc Surg 137(2):453–458 Sugarbaker PH, 1995, Peritonectomy procedures. Ann Surg 221:29–42 Jacquet P, Sugarbaker PH, 1996, Current methodologies for clinical assessment of patients with peritoneal carcinomatosis. J Exp Clin Cancer Res 15:49–58 Yang H, Bocchetta M, Kroczynska B, Elmishad AG, Chen Y, Liu Z et al, 2006, TNF-alpha inhibits asbestos-induced cytotoxicity via a NF-kappaB-dependent pathway, a possible mechanism for asbestos-induced oncogenesis. Proc Natl Acad Sci USA 103, 27):10397–10402 Carbone M, Kratzke RA, Testa JR, 2002, The pathogenesis of mesothelioma. Semin Oncol 29(1):2–17 Ault JG, Cole RW, Jensen CG, Jensen LC, Bachert LA, Rieder CL, 1995, Behavior of crocidolite asbestos during mitosis in living vertebrate lung epithelial cells. Cancer Res 55(4):792–798 Topov J, Kolev K, 1987, Cytology of experimental mesotheliomas induced with crocidolite asbestos. Acta Cytol 31(3):369–373 Craighead JE, Akley NJ, Gould LB, Libbus BL, 1987, Characteristics of tumors and tumor cells cultured from experimental asbestos-induced mesotheliomas in rats. Am J Pathol 129(3):448– 462 Manning CB, Cummins AB, Jung MW, Berlanger I, Timblin CR, Palmer C et al, 2002, A mutant epidermal growth factor receptor targeted to lung epithelium inhibits asbestos-induced proliferation and proto-oncogene expression. Cancer Res 62(15):4169–4175 Zanella CL, Posada J, Tritton TR, Mossman BT, 1996, Asbestos causes stimulation of the extracellular signal-regulated kinase 1 mitogen-activated protein kinase cascade after phosphorylation of the epidermal growth factor receptor. Cancer Res 56(23):5334– 5338 Gazdar AF, Carbone M, 2003, Molecular pathogenesis of malignant mesothelioma and its relationship to simian virus 40. Clin Lung Cancer 5(3):177–181 Aoe K, Hiraki A, Tanaka T, Gemba K-I, Taguchi K, Murakami T et al, 2006, Expression of vascular endothelial growth factor in malignant mesothelioma. Anticancer Res 26(6C):4833–4836 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 217 EP 222 DI 10.1007/s12253-009-9210-9 PG 6 ER PT J AU Kruszyna, Lianeri, M Rydzanicz, M Szyfter, K Jagodzinski, PP AF Kruszyna, Lukasz Lianeri, Margarita Rydzanicz, Malgorzata Szyfter, Krzysztof Jagodzinski, P Pawel TI SDF1-3'A Gene Polymorphism is Associated with Laryngeal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SDF1-3’ G801A; Polymorphism; Laryngeal cancer; CXCR4; PCR-RFLP ID SDF1-3’ G801A; Polymorphism; Laryngeal cancer; CXCR4; PCR-RFLP AB The SDF1-3’ G801A (rs 1801157) polymorphism is associated with increased risk of various types of cancers, including those of the neck and head. Using PCR-RFLPs, we investigated the distribution of SDF1-3′ G801A genotypes in patients with laryngeal cancer (n=118) and controls (n=250) in Poland. We found that patients with SDF1-3’ A/A and G/A genotypes exhibit a 1.863-fold increased risk of laryngeal cancer (95% CI=1.177–2.949, p=0.0086). However, there was no significant increase in risk for the homozygous SDF1-3’ A/A genotype OR=3.235 (95% CI=0.5330−19.633, p=0.3329). We also did not observe a significant association between tumor characteristics and prevalence of alleles or genotypes for the SDF1-3′ G801A polymorphism. Our findings suggest that the SDF1-3'A variant may be associated with an increased risk of laryngeal cancer. C1 [Kruszyna, Lukasz] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. [Lianeri, Margarita] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. [Rydzanicz, Malgorzata] Polish Academy of Sciences, Institute of Human Genetics, Department of Environmental MutagenesisPoznan, Poland. [Szyfter, Krzysztof] Polish Academy of Sciences, Institute of Human Genetics, Department of Environmental MutagenesisPoznan, Poland. [Jagodzinski, P Pawel] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. RP Jagodzinski, PP (reprint author), Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 60-781 Poznan, Poland. EM pjagodzi@am.poznan.pl CR Sadri M, McMahon J, Parker A, 2006, Management of laryngeal dysplasia. Eur Arch Otorhinolaryngol 263:843–852 Sapkota A, Hsu CC, Zaridze D et al, 2008, Dietary risk factors for squamous cell carcinoma of the upper aerodigestive tract in central and eastern Europe. Cancer Causes Control 19:1161–1170 Freedman ND, Abnet CC, Leitzmann MF, Hollenbeck AR et al, 2007, Prospective investigation of the cigarette smokinghead and neck cancer association by sex. Cancer 110:1593– 1601 Szyfter K, Szmeja Z, Szyfter W et al, 1999, Molecular and cellular alterations in tobacco smoke-associated larynx cancer. Mutat Res 445:259–274 Bradford CR, 1999, Predictive factors in head and neck cancer. Hematol Oncol Clin North Am 13:777–785 Moser B, Loetscher P, 2001, Lymphocyte traffic control by chemokines. Nat Immunol 2:123–128 Zlotnik A, Yoshie O, 2000, Chemokines: a new classification system and their role in immunity. Immunity 12:121–127 Campbell JJ, Butcher E, 2000, Chemokines in tissue-specific and microenvironment-specific lymphocyte homing. Curr Opin Immunol 12:336–341 Balkwill F, 2003, Chemokine biology in cancer. Sem Immunol 15:49–55 Zou YR, Kottmann AH, Kuroda M et al, 1998, Function of the chemokine receptor CXCR4 in a heamatopoiesis and in cerebellar development. Nature 393:595–599 Onai N, Zhang Y, Yoneyama H et al, 2008, Impairment of lymphopoiesis and myelopoiesis in mice reconstituted with bone marrow-hematopoietic progenitor cells expressing SDF-1- intrakine. Blood 96:2074–2080 Yasuoka H, Tsujimoto M, Yoshidome K et al, 2008, Cytoplasmic CXCR4 expression in breast cancer: induction by nitric oxide and correlation with lymph node metastasis and poor prognosis. BMC Cancer 8:340 Kim J, Mori T, Chen SL et al, 2006, Chemokine receptor CXCR4 expression in patients with melanoma and colorectal cancer liver metastases and the association with disease outcome. Ann Surg 244:113–120 Marechal R, Demetter P, Nagy N et al, 2009, High expression of CXCR4 may predict poor survival in resected pancreatic adenocarcinoma. Br J Cancer 100:1444–1451 Kajiyama H, Shibata K, Terauchi M et al, 2008, Involvement of SDF-1alpha/CXCR4 axis in the enhanced peritoneal metastasis of epithelial ovarian carcinoma. Int J Cancer 122:91–99 Akashi T, Koizumi K, Tsuneyama K et al, 2008, Chemokine receptor CXCR4 expression and prognosis in patients with metastatic prostate cancer. Cancer Sci 99:539–542 Kijima T, Maulik G, Ma PC et al, 2002, Regulation of cellular proliferation, cytoskeletal function, and signal transduction through CXCR4 and c-Kit in small cell lung cancer cells. Cancer Res 62:6304–6311 Lee HJ, Kim SW, Kim HY et al, 2009, Chemokine receptor CXCR4 expression, function, and clinical implications in gastric cancer. Int J Oncol 34:473–480 Oliveira-Neto HH, Silva ET, Leles CR et al, 2008, Involvement of CXCL12 and CXCR4 in lymph node metastases and development of oral squamous cell carcinomas. Tumour Biol 29:262–271 Taki M, Higashikawa K, Yoneda S et al, 2008, Up-regulation of stromal cell-derived factor-1alpha and its receptor CXCR4 expression accompanied with epithelial-mesenchymal transition in human oral squamous cell carcinoma. Oncol Rep 19:993– 998 Winkler C, Modi W, Smith MWet al, 1998, Genetic restriction of AIDS pathogenesis by an SDF-1 chemokine gene variant. Science 279:389–393 Watanabe MA, de Oliveira Cavassin GG, Orellana MD et al, 2003, SDF-1 gene polymorphisms and syncytia induction in Brazilian HIV-1 infected individuals. Microb Pathog 35:31–34 Kimura R, Nishioka T, Ishida T, 2003, The SDF1–G801A polymorphism is not associated with SDF1 gene expression in Epstein–Barr virus-transformed lymphoblastoid cells. Genes Immun 4:356–361 de Oliveira Cavassin GG, De Lucca FL, Delgado Andre N et al, 2004, Molecular investigation of the stromal cell-derived factor-1 chemokine in lymphoid leukemia and lymphoma patients from Brazil. Blood Cells Mol Diseases 33:90–93 Razmkhah M, Talei AR, Doroudchi M et al, 2004, Stromal cellderived factor-1, SDF-1, alleles and susceptibility to breast carcinoma. Cancer Lett 225:261–266 Razmkhah M, Doroudchi M, Ghayumi SM et al, 2005, Stromal cell-derived factor-1, SDF-1, gene and susceptibility of Iranian patients with lung cancer. Lung Cancer 49:311–315 Teng YH, Liu TH, Tseng HC et al, 2009, Contribution of genetic polymorphisms of stromal cell-derived factor-1 and its receptor, CXCR4, to the susceptibility and clinicopathologic development of oral cancer. Head Neck., DOI 10.1002/hed.21094 Khademi B, Razmkhah M, Erfani N et al, 2008, SDF-1 and CCR5 genes polymorphism in patients with head and neck cancer. Pathol Oncol Res 14:45–50 Vairaktaris E, Vylliotis A, Spyridonodou S et al, 2008, A DNA polymorphism of stromal-derived factor-1 is associated with advanced stages of oral cancer. Anticancer Res 28:271–275 Frazer KA, Ballinger DG, Cox DR et al, 2007, A second generation human haplotype map of over 3.1 million SNPs. Nature 449:851–861 Shirozu M, Nakano T, Inazawa J et al, 1995, Structure and chromosomal localization of the human stromal cell-derived factor 1, SDF1, gene. Genomics 28:495–500 Zlotnik A, 2006, Chemokines and cancer. Int J Cancer 119:2026– 2029 Zlotnik A, 2008, New insights on the role of CXCR4 in cancer metastasis. J Pathol 215:211–213 Bleul CC, Farzan M, Choe H et al, 1996, The lymphocyte chemoattractant SDF-1 is a ligand for LESTR/fusin and blocks HIV-1 entry. Nature 382:829–833 Vlahakis SR, Villasis-Keever A, Gomez T et al, 2002, G protein-coupled chemokine receptors induce both survival and apoptotic signaling pathways. J Immunol 169:5546– 5554 Chang L, Karin M, 2001, Mammalian MAP kinase signalling cascades. Nature 410:37–40 Daly AJ, McIlreavey L, Irwin CR, 2008, Regulation of HGF and SDF-1 expression by oral fibroblasts–implications for invasion of oral cancer. Oral Oncol 44:646–651 Uchida D, Onoue T, Tomizuka Y et al, 2007, Involvement of an autocrine stromal cell derived factor-1/CXCR4 system on the distant metastasis of human oral squamous cell carcinoma. Mol Cancer Res 5:685–694 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 223 EP 227 DI 10.1007/s12253-009-9214-5 PG 5 ER PT J AU Ren, K Zhang, W Shi, Y Gong, J AF Ren, Ke Zhang, Wei Shi, Yujun Gong, Jianping TI Pim-2 Activates API-5 to Inhibit the Apoptosis of Hepatocellular Carcinoma Cells Through NF-κB Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pim-2; NF-κB; API-5; Hepatocellular carcinoma cells; Apoptosis ID Pim-2; NF-κB; API-5; Hepatocellular carcinoma cells; Apoptosis AB Pim-2 is proved to be relevant to the tumorigenesis of hepatocellular carcinoma (HCC), but the mechanism is unclear.We studied the relationship among Pim-2,NF-κBand API-5. In our experiment, expression level of the three factors and phosphorylation level of API-5, aswell as NF-κB activity, were detected in HCC tissues and the nontumorous controls. Then Pim-2 gene was transfected into nontumorous liver cells L02, and Pim-2 SiRNA was transfected into hepatoblastoma cell line HepG2. Parthenolide was added as NF-κB inhibitor. The same detections as above were repeated in the cells, along with the apoptosis analysis. We found the levels of Pim-2, NF-κB and API-5, as well as NF-κB activity, were significantly higher in HCC tissues. Pim-2 level was increased in L02 cells after the transfection of Pim-2 gene, but decreased in HepG2 cells after the transfection of Pim-2 SiRNA. The levels of NF-κB and API-5, as well as NF-κB activity and API-5 phosphorylation level, were in accordance with Pim-2 level, but could be reversed by Parthenolide. Cell apoptosis rates were negatively correlated with API-5 phosphorylation level. Therefore, we infer that Pim-2 could activate API-5 to inhibit the apoptosis of liver cells, and NF-κB is the key regulator. C1 [Ren, Ke] The Second Affiliated Hospital of Chongqing Medical University, Department of Hepatobiliary SurgeryChongqing, China. [Zhang, Wei] The Second Affiliated Hospital of Chongqing Medical University, Department of Hepatobiliary SurgeryChongqing, China. [Shi, Yujun] Sichuan University, West China HospitalChengdu, China. [Gong, Jianping] The Second Affiliated Hospital of Chongqing Medical University, Department of Hepatobiliary SurgeryChongqing, China. RP Gong, J (reprint author), The Second Affiliated Hospital of Chongqing Medical University, Department of Hepatobiliary Surgery, Chongqing, China. EM gongjianping11@126.com CR Breuer ML, Cuypers HT, Berns A, 1989, Evidence for the involvement of pim-2, a new common proviral insertion site, in progression of lymphomas. EMBO J 8(3):743–7488 Ren K, Shi YJ, Gong JP, 2007, The research on the mechanism of Pim-2 suppressing the apoptosis of tumor cell. Prog Physiol Sci 38, 2):136–139 Chen JL, Limnander A, Rothman PB, 2008, Pim-1 and Pim-2 kinases are required for efficient pre-B-cell transformation by v-Abl oncogene. Blood 111(3):1677–1685 Dai H, Li R, Wheeler T et al, 2005, Pim-2 upregulation: biological implications associated with disease progression and perinueral invasion in prostate cancer. Prostate 65(3):276–286 Gong J, Wang J, Ren K et al, 2008, Serine/Threonine Kinase Pim-2 Promotes Liver Tumorigenesis Induction through Mediating Survival and Preventing Apoptosis of Liver Cell. J Surg Res. Apr 22. [Epub ahead of print] Li B, Ye T, Li DH, 2008, Expression of Pim-2 in normal liver and hepatocellular carcinoma tissues. Chinese J Hepatol 16(12):950–951 Hammerman PS, Fox CJ, Cinalli RM et al, 2004, Lymphocyte Transformation by Pim-2 Is Dependent on Nuclear Factor-κB Activation. Cancer Res 64:8341–8348 Aghajanian C, Soignet S, Dizon DS et al, 2002, A phase I trial of the novel proteasome inhibitor PS341 in advanced solid tumor malignancies. Clin Cancer Res 8:2505–2511 White E, 2003, The pims and outs of survival signaling: role for the Pim-2 protein kinase in the suppression of apoptosis by cytokines. Genes Dev 17:1813–1816 Macdonald A, Campbell DG, Toth R et al, 2006, Pim kinases phosphorylate multiple sites on Bad and promote14–3–3 binding and dissociation from Bcl-XL. BMC Cell Biol 7:1186–1471 Peng C, Knebel A, Morrice NA et al, 2007, Pim kinase substrate identification and specificity. J Biochem 141(3):353–62 Wang Y, Lee AT, Ma JZ et al, 2008, Profiling microRNA expression in hepatocellular carcinoma reveals microRNA-224 up-regulation and apoptosis inhibitor-5 as a microRNA-224 specific target. J Biol Chem 283(19):13205–15 Hehner SP, Hofmann TG, Droge W et al, 1999, The antiinflammatory sesquiterpene lactone parthenolide inhibits NF-kappa B by targeting the I kappa B kinase complex. J Immunol 163, 10):5617–23 Li W, Lei P, Yu B et al, 2008, Screening and identification of a novel target specific for hepatoma cell line HepG2 from the FliTrx bacterial peptide library. Acta Biochim Biophys Sin 40, 5):443–51 Guzman ML, Rossi RM, Karnischky L et al, 2005, The sesquiterpene lactone parthenolide induces apoptosis of human acute myelogenous leukemia stem and progenitor cells. Blood 105(11):4163–9 Jiang JX, Mikami K, Venugopal S et al, 2009, Apoptotic body engulfment by hepatic stellate cells promotes their survival by the JAK/STAT and Akt/NF-kappaB-dependent pathways. J Hepatol 51(1):139–48 Hoffmann F, Sass G, Zillies J et al, 2009, A novel technique for selective NF-kappaB inhibition in Kupffer cells - contrary effects in fulminant hepatitis and ischemia/reperfusion. Gut May 25. [Epub ahead of print] Kuboki S, Sakai N, Clarke C et al, 2009, The peptidyl-prolyl isomerase, Pin1, facilitates NF-kappaB binding in hepatocytes and protects against hepatic ischemia/reperfusion injury. J Hepatol May 24. [Epub ahead of print] Pelengaris S, Khan M, Evan G, 2002, c-Myc: more than just a matter of life and death. Nature Reviews. Cancer 2(10):764–776 Hammerman PS, Fox CJ, Cinalli RM et al, 2004, Lymphocyte Transformation by Pim-2 Is Dependent on Nuclear Factor-κB Activation. Cancer Res 64:8341–8348 Zhang Y, Wang Z, Li X et al, 2008, Pim kinase-dependent inhibition of c-Myc degradation. Oncogene 27(35):4809–19 Bachmann M, Moroy T, 2005, The serine/threonine kinase Pim-1. Int J Biochem Cell Biol 37:726–730 Fujii C, Nakamoto Y, Lu P et al, 2005, Aberrant expression of serine/threonine kinase Pim-3 in hepatocellular carcinoma development and its role in the proliferation of human hepatoma cell lines. Int J Cancer 114(2):209–218 Qian K, Wang L, Cywin CL et al, 2009, Hit to lead account of the discovery of a new class of inhibitors of Pim kinases and crystallographic studies revealing an unusual kinase binding mode. J Med Chem 52(7):1814–27 Xia Z, Knaak C, Ma J et al, 2009, Synthesis and evaluation of novel inhibitors of Pim-1 and Pim-2 protein kinases. J Med Chem 52(1):74–86 Tong Y, Stewart KD, Thomas S et al, 2008, Isoxazolo [3, 4-b] quinoline-3, 4, 1H, 9H, -diones as unique, potent and selective inhibitors for Pim-1 and Pim-2 kinases: chemistry, biological activities, and molecular modeling. Bioorg Med Chem Lett 18, 19):5206–5208 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 229 EP 237 DI 10.1007/s12253-009-9215-4 PG 9 ER PT J AU Seow, FH Yip, KW Loh, WH Ithnin, H Por, P Rohaizak, M AF Seow, Fong Heng Yip, Kien Wai Loh, Woon Hui Ithnin, Hairuszah Por, Patricia Rohaizak, Mohammad TI Immunohistochemical Detection of Phospho-Akt, Phospho-BAD, HER2 and Oestrogen Receptors α and β in Malaysian Breast Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Akt; Oestrogen receptor; HER2 ID Breast cancer; Akt; Oestrogen receptor; HER2 AB Activation of Akt signaling pathway has been documented in various human malignancies, including breast carcinoma. The objective of this study is to determine the incidence of Akt phosphorylation in breast tumours and its relationship with expression of ER-α, ER-β, HER2, Ki-67 and phosphorylated Bcl-2 associated death domain (p-BAD). Immunohistochemical staining was performed to detect these molecules on 43 paraffin-embedded breast tumour tissues with commercially available antibodies. Eighteen (41.9%), 3 (7.0%), 23 (53.5%), 35 (81.4%), 21 (48.8%), 29 (67.4%), and 34 (81.0%) of breast tumours were positive for nuclear ER-α, nuclear ER-β, membranous HER2, cytonuclear p-Akt (Thr308), p-Akt (Ser473), p-BAD and Ki-67, respectively. ER-α expression was inversely correlated with HER2 and Ki-67 (P=0.041 and P=0.040, respectively). The p-Akt (Ser473) was correlated with increased level of p-BAD (Ser136) (P=0.012). No relationship of Akt phosphorylation with HER2, ER-α or ER-β was found. The p-Akt (Ser473) immunoreactivity was significantly higher in stage IV than in stage I or II (P=0.036 or P=0.009). The higher Ki-67 and lower ER-α expression showed an association with patient age of <50 years (P=0.004) and with positive nodal status (P=0.033), respectively. Our data suggest that the Akt phosphorylation and inactivation of its downstream target, BAD may play a role in survival of breast cancer cell. This study does not support the simple model of linear HER2/PI3K/Akt pathway in breast cancer. C1 [Seow, Fong Heng] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, 43400 Selangor, Selangor, Malaysia. [Yip, Kien Wai] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, 43400 Selangor, Selangor, Malaysia. [Loh, Woon Hui] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, 43400 Selangor, Selangor, Malaysia. [Ithnin, Hairuszah] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, 43400 Selangor, Selangor, Malaysia. [Por, Patricia] Assunta Hospital, Jalan Templer, 46990 Petaling Jaya, Selangor, Malaysia. [Rohaizak, Mohammad] Hospital Universiti Kebangsaan Malaysia, Faculty of Medicine, Department of Surgery, 56000 Cheras, Malaysia. RP Seow, FH (reprint author), Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, 43400 Selangor, Malaysia. EM shf@medic.upm.edu.my CR Lim GCC, Halimah Y, 2004, Second report of the national cancer registry: cancer incidence in Malaysia 2003. National Cancer Registry, Malaysia Clarke R, Skaar T, Leonessa F et al, 1996, Acquisition of an antiestrogen-resistant phenotype in breast cancer: role of cellular and molecular mechanisms. Cancer Treat Res 87:263–283 Ring A, Dowsett M, 2004, Mechanisms of tamoxifen resistance. Endocr Relat Cancer 11:643–658 Pandolfi PP, 2004, Breast cancer—loss of PTEN predicts resistance to treatment. N Engl J Med 351:2337–2338 Tokunaga E, Kataoka A, Kimura Y et al, 2006, The association between Akt activation and resistance to hormone therapy in metastatic breast cancer. Eur J Cancer 42:629–635 Brognard J, Clark AS, Ni Y et al, 2001, Akt/protein kinase B is constitutively active in non-small cell lung cancer cells and promotes cellular survival and resistance to chemotherapy and radiation. Cancer Res 61:3986–3997 Clark AS, West K, Streicher S et al, 2002, Constitutive and inducible Akt activity promotes resistance to chemotherapy, trastuzumab, or tamoxifen in breast cancer cells. Mol Cancer Ther 1:707–717 Luo Y, Shoemaker AR, Liu X et al, 2005, Potent and selective inhibitors of Akt kinases slow the progress of tumors in vivo. Mol Cancer Ther 4:977–986 Kondapaka SB, Singh SS, Dasmahapatra GP et al, 2003, Perifosine, a novel alkylphospholipid, inhibits protein kinase B activation. Mol Cancer Ther 2:1093–1103 Jones PF, Jakubowicz T, Hemmings BA, 1991, Molecular cloning of a second form of rac protein kinase. Cell Regul 2:1001–1009 Bellacosa A, Kumar CC, Di Cristofano A et al, 2005, Activation of AKT kinases in cancer: implications for therapeutic targeting. Adv Cancer Res 94:29–86 Masure S, Haefner B, Wesselink JJ et al, 1999, Molecular cloning, expression and characterization of the human serine/ threonine kinase Akt-3. Eur J Biochem 265:353–360 Cheng JQ, Godwin AK, Bellacosa A et al, 1992, AKT2, a putative oncogene encoding a member of a subfamily of protein-serine/ threonine kinases, is amplified in human ovarian carcinomas. Proc Natl Acad Sci U S A 89:9267–9271 Alessi DR, James SR, Downes CP et al, 1997, Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha. Curr Biol 7:261–269 Stephens L, Anderson K, Stokoe D et al, 1998, Protein kinase B kinases that mediate phosphatidylinositol 3, 4, 5-trisphosphatedependent activation of protein kinase B. Science 279:710–714 Sarbassov DD, Guertin DA, Ali SM et al, 2005, Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 307:1098–1101 Hresko RC, Mueckler M, 2005, mTOR.RICTOR is the Ser473 kinase for Akt/protein kinase B in 3T3-L1 adipocytes. J Biol Chem 280:40406–40416 Dimmeler S, Fleming I, Fisslthaler B et al, 1999, Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature 399:601–605 Nakanishi K, Sakamoto M, Yasuda J et al, 2002, Critical involvement of the phosphatidylinositol 3-kinase/Akt pathway in anchorage-independent growth and hematogeneous intrahepatic metastasis of liver cancer. Cancer Res 62:2971–2975 Altomare DA, Testa JR, 2005, Perturbations of the AKT signaling pathway in human cancer. Oncogene 24:7455–7464 Wu Y, Mohamed H, Chillar R et al, 2008, Clinical significance of Akt and HER2/neu overexpression in African–American and Latina women with breast cancer. Breast Cancer Res 10:R3 Lazennec G, Bresson D, Lucas A et al, 2001, ER beta inhibits proliferation and invasion of breast cancer cells. Endocrinology 142:4120–4130 Rajnakova A, Goh PM, Chan ST et al, 1997, Expression of differential nitric oxide synthase isoforms in human normal gastric mucosa and gastric cancer tissue. Carcinogenesis 18:1841–1845 Endo K, Terada T, 2000, Protein expression of CD44, standard and variant isoforms, in hepatocellular carcinoma: relationships with tumor grade, clinicopathologic parameters, p53 expression, and patient survival. J Hepatol 32:78–84 Allred DC, Harvey JM, Berardo M et al, 1998, Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11:155–168 Cobleigh MA, Vogel CL, Tripathy D et al, 1999, Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol 17:2639–2648 Scheid MP, Marignani PA, Woodgett JR, 2002, Multiple phosphoinositide 3-kinase-dependent steps in activation of protein kinase B. Mol Cell Biol 22:6247–6260 Nadji M, Gomez-Fernandez C, Ganjei-Azar P et al, 2005, Immunohistochemistry of estrogen and progesterone receptors reconsidered: experience with 5,993 breast cancers. Am J Clin Pathol 123:21–27 Looi LM, Cheah PL, 1998, C-erbB-2 oncoprotein amplification in infiltrating ductal carcinoma of breast relates to high histological grade and loss of oestrogen receptor protein. Malays J Pathol 20:19–23 Pavao M, Traish AM, 2001, Estrogen receptor antibodies: specificity and utility in detection, localization and analyses of estrogen receptor alpha and beta. Steroids 66:1–16 Speirs V, Green CA, Shaaban AM, 2008, Oestrogen receptor beta immunohistochemistry: time to get it right? J Clin Pathol 61:1150–1151 author reply 1151-2 Rhodes A, Jasani B, Balaton AJ et al, 2000, Frequency of oestrogen and progesterone receptor positivity by immunohistochemical analysis in 7016 breast carcinomas: correlation with patient age, assay sensitivity, threshold value, and mammographic screening. J Clin Pathol 53:688–696 Slamon DJ, Godolphin W, Jones LA et al, 1989, Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 244:707–712 Chariyalertsak S, Chariyalertsak S, Cheirsilpa A et al, 1996, Immunohistochemical detection of c-erbB-2 oncoprotein in patients with breast cancer. J Med Assoc Thail 79:715–721 Almasri NM, Al Hamad M, 2005, Immunohistochemical evaluation of human epidermal growth factor receptor 2 and estrogen and progesterone receptors in breast carcinoma in Jordan. Breast Cancer Res 7:R598–R604 Rosa FE, Caldeira JR, Felipes J et al, 2008, Evaluation of estrogen receptor alpha and beta and progesterone receptor expression and correlation with clinicopathologic factors and proliferative marker Ki-67 in breast cancers. Hum Pathol 39:720– 730 Jarzabek K, Koda M, Kozlowski L et al, 2005, Distinct mRNA, protein expression patterns and distribution of oestrogen receptors alpha and beta in human primary breast cancer: correlation with proliferation marker Ki-67 and clinicopathological factors. Eur J Cancer 41:2924–2934 Finlin BS, Gau CL, Murphy GA et al, 2001, RERG is a novel rasrelated, estrogen-regulated and growth-inhibitory gene in breast cancer. J Biol Chem 276:42259–42267 Guo S, Sonenshein GE, 2004, Forkhead box transcription factor FOXO3a regulates estrogen receptor alpha expression and is repressed by the Her-2/neu/phosphatidylinositol 3-kinase/Akt signaling pathway. Mol Cell Biol 24:8681–8690 Fuqua SA, Schiff R, Parra I et al, 2003, Estrogen receptor beta protein in human breast cancer: correlation with clinical tumor parameters. Cancer Res 63:2434–2439 Skliris GP, Munot K, Bell SM et al, 2003, Reduced expression of oestrogen receptor beta in invasive breast cancer and its re-expression using DNA methyl transferase inhibitors in a cell line model. J Pathol 201:213–220 Fox EM, Davis RJ, Shupnik MA, 2008, ERbeta in breast cancer—onlooker, passive player, or active protector? Steroids 73:1039–1051 Al-Bazz YO, Underwood JC, Brown BL et al, 2009, Prognostic significance of Akt, phospho-Akt and BAD expression in primary breast cancer. Eur J Cancer 45:694–704 Bose S, Chandran S, Mirocha JM et al, 2006, The Akt pathway in human breast cancer: a tissue-array-based analysis. Mod Pathol 19:238–245 Ahmad S, Singh N, Glazer RI, 1999, Role of AKT1 in 17betaestradiol- and insulin-like growth factor I, IGF-I)-dependent proliferation and prevention of apoptosis in MCF-7 breast carcinoma cells. Biochem Pharmacol 58:425–430 Tsai EM, Wang SC, Lee JN et al, 2001, Akt activation by estrogen in estrogen receptor-negative breast cancer cells. Cancer Res 61:8390–8392 Kirkegaard T, Witton CJ, McGlynn LM et al, 2005, AKT activation predicts outcome in breast cancer patients treated with tamoxifen. J Pathol 207:139–146 Lenferink AE, Busse D, Flanagan WM et al, 2001, ErbB2/neu kinase modulates cellular p27(Kip1, and cyclin D1 through multiple signaling pathways. Cancer Res 61:6583–6591 Hermanto U, Zong CS, Wang LH, 2001, ErbB2-overexpressing human mammary carcinoma cells display an increased requirement for the phosphatidylinositol 3-kinase signaling pathway in anchorage-independent growth. Oncogene 20:7551–7562 Pianetti S, Arsura M, Romieu-Mourez R et al, 2001, Her-2/neu overexpression induces NF-kappaB via a PI3-kinase/Akt pathway involving calpain-mediated degradation of IkappaB-alpha that can be inhibited by the tumor suppressor PTEN. Oncogene 20:1287– 1299 Zhang G, He B, Weber GF, 2003, Growth factor signaling induces metastasis genes in transformed cells: molecular connection between Akt kinase and osteopontin in breast cancer. Mol Cell Biol 23:6507– 6519 Panigrahi AR, Pinder SE, Chan SY et al, 2004, The role of PTEN and its signalling pathways, including AKT, in breast cancer; an assessment of relationships with other prognostic factors and with outcome. J Pathol 204:93–100 Schmitz KJ, Otterbach F, Callies R et al, 2004, Prognostic relevance of activated Akt kinase in node-negative breast cancer: a clinicopathological study of 99 cases. Mod Pathol 17:15–21 Stal O, Perez-Tenorio G, Akerberg L et al, 2003, Akt kinases in breast cancer and the results of adjuvant therapy. Breast Cancer Res 5:R37–R44 Zhou X, Tan M, Stone Hawthorne Vet al, 2004, Activation of the Akt/mammalian target of rapamycin/4E-BP1 pathway by ErbB2 overexpression predicts tumor progression in breast cancers. Clin Cancer Res 10:6779–6788 Hill MM, Hemmings BA, 2002, Inhibition of protein kinase B/ Akt. implications for cancer therapy. Pharmacol Ther 93:243–251 Zha J, Harada H, Yang E et al, 1996, Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L). Cell 87:619–628 Fernando RI, Wimalasena J, 2004, Estradiol abrogates apoptosis in breast cancer cells through inactivation of BAD: Ras-dependent nongenomic pathways requiring signaling through ERK and Akt. Mol Biol Cell 15:3266–3284 Krasilnikov MA, 2000, Phosphatidylinositol-3 kinase dependent pathways: the role in control of cell growth, survival, and malignant transformation. Biochemistry, Mosc, 65:59–67 Liao Y, Hung MC, 2003, Regulation of the activity of p38 mitogen-activated protein kinase by Akt in cancer and adenoviral protein E1A-mediated sensitization to apoptosis. Mol Cell Biol 23:6836–6848 Yonemori K, Tsuta K, Shimizu C, et al, 2009, Immunohistochemical expression of PTEN and phosphorylated Akt are not correlated with clinical outcome in breast cancer patients treated with trastuzumab-containing neo-adjuvant chemotherapy. Med Oncol 26:344-349 Klauber-DeMore N, 2005, Tumor biology of breast cancer in young women. Breast Dis 23:9–15 Hartley MC, McKinley BP, Rogers EA et al, 2006, Differential expression of prognostic factors and effect on survival in young, < or =40, breast cancer patients: a case-control study. Am Surg 72:1189–1194 discussion 1194-5 HusseinMR, Abd-Elwahed SR, Abdulwahed AR, 2008, Alterations of estrogen receptors, progesterone receptors and c-erbB2 oncogene protein expression in ductal carcinomas of the breast. Cell Biol Int 32:698–707 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 239 EP 248 DI 10.1007/s12253-009-9216-3 PG 10 ER PT J AU Zidar, N Zver, S Jurcic, V AF Zidar, Nina Zver, Samo Jurcic, Vesna TI Extraosseus Plasmacytoma of the Pharynx with Localized Light Chain Deposition. Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Light chain deposition disease; Isolated; Pharynx; Extraosseus plasmacytoma ID Light chain deposition disease; Isolated; Pharynx; Extraosseus plasmacytoma AB Light chain deposition disease (LCDD) is a rare disorder associated with a clonal proliferation of plasma cells, which synthesize abnormal monoclonal immunoglobulin light chains. It is characterized by systemic deposition of light chains in various organs, with the kidneys being most commonly affected. There have been few reports of isolated LCDD, i.e. in the brain, lungs and cervical lymph nodes. We here report on another patient with an isolated form of LCDD, which was limited to the pharyngeal mucosa and was associated with an extraosseus plasmacytoma of the pharynx, expanding the spectrum that has been recognized for LCDD. The patient was treated by local radiotherapy, with an excellent response. A less aggressive clinical course can probably be expected than in the usual form of LCDD, but a longterm follow-up is necessary to establish the clinical significance of this variant of LCDD. C1 [Zidar, Nina] University of Ljubljana, Faculty of Medicine, Institute of Pathology, Korytkova 2, 1000 Ljubljana, Slovenia. [Zver, Samo] University Medical Centre Ljubljana, Department of Haematology, Zaloska 7, 1525 Ljubljana, Slovenia. [Jurcic, Vesna] University of Ljubljana, Faculty of Medicine, Institute of Pathology, Korytkova 2, 1000 Ljubljana, Slovenia. RP Zidar, N (reprint author), University of Ljubljana, Faculty of Medicine, Institute of Pathology, 1000 Ljubljana, Slovenia. EM nina.zidar@mf.uni-lj.si CR Lin J, Markowitz GS, Valeri AM, Kambham N, Sherman WH, Appel GB et al, 2001, Renal monoclonal immunoglobulin deposition disease: the disease spectrum. J Am Soc Nephrol 12:1482–1492 Pozzi C, Locatelli F, 2002, Kidney and liver involvement in monoclonal light chain disorders. Semin Nephrol 22:319–330 Pozzi C, D’Amico M, Fogazzi GB, Curioni S, Ferrario F, Pasquali S et al, 2003, Light chain deposition disease with renal involvement: clinical characteristics and prognostic factors. Am J Kidney Dis 42:1154–1163 McKenna RW, Kyle RA, Kuehl WM, Grogan TM, Harris NL, Coupland RW, 2008, Plasma cell neoplasms. In: Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Vardiman JW, eds, WHO classification of tumours of haemopoietic and lymphoid tissues, 2nd edn. IARC, Lyon, pp 200–213 Popovic M, Tavcar R, Glavac D, Volavsek M, Pirtosek Z, Vizjak A, 2007, Light chain deposition disease restricted to the brain: the first case report. Hum Pathol 38:179–184 Piard F, Yaziji N, Jarry O, Assem M, Martin L, Bernard A et al, 1998, Solitary plasmacytoma of the lung with light chain extracellular deposits: a case report and review of the literature. Histopathol 32:356–361 Rostagno A, Frizzera G, Ylagan L, Kumar A, Ghiso J, Gallo G, 2002, Tumoral non-amyloidotic monoclonal immunoglobulin light chain deposits, ‘aggregoma’): presenting feature of B-cell dyscrasia in three cases with immunohistochemical and biochemical analyses. Br J Haematol 119:62–69 Bellotti V, Merlini G, 1996, Toward understanding the molecular pathogenesis of monoclonal immunoglobulin light-chain deposition. Nephrol Dial Transplant 11:1708–1711 Randall RE, Williamson WC Jr, Mullinax F, Tung NY, Still WJ, 1976, Manifestations of systemic light chain deposition. Am J Med 60:293–299 Buxbaum JN, Genega EM, Lazowski P, Kumar A, Tunick PA, Kronzon I et al, 2000, Infiltrative nonamyloidotic monoclonal immunoglobulin light chain cardiomyopathy: an underappreciated manifestation of plasma cell dyscrasias. Cardiology 93:220–228 Toor AA, Ramdane BA, Joseph J, Thomas M, O’Hara C, Barlogie B et al, 2006, Cardiac nonamyloidotic immunoglobulin deposition disease. Mod Pathol 19:233–237 Rivest C, Turgeon PP, Senecal JL, 1993, Lambda light chain deposition disease presenting as an amyloid-like arthropathy. J Rheumatol 20:880–884 Bhargava P, Rushin JM, Rusnock EJ, Hefter LG, Franks TJ, Sabnis SG et al, 2007, Pulmonary light chain deposition disease: report of five cases and review of the literature. Am J Surg Pathol 31:267–276 Colombat M, Mal H, Copie-Bergman C, Diebold J, Damotte D, Callard P et al, 2008, Primary cystic lung light chain deposition disease: a clinicopathologic entity derived from unmutated B cells with a stereotyped IGHV4-34/IGKV1 receptor. Blood 112:2004– 2012 Khoor A, Myers JL, Tazelaar HD, Kurtin PJ, 2004, Amyloid-like pulmonary nodules, including localized light-chain deposition: clinicopathologic analysis of three cases. Am J Clin Pathol 121:200–204 Weichman K, Dember LM, Prokaeva T, Wright DG, Quillen K, Rosenzweig M et al, 2006, Clinical and molecular characteristics of patients with non-amyloid light chain deposition disorders, and outcome following treatment with high-dose melphalan and autologous stem cell transplantation. Bone Marrow Transplant 38:339–343 Lorenz EC, Gertz MA, Fervenza FC, Dispenzieri A, Lacy MQ, Hayman SR et al, 2008, Long-term outcome of autologous stem cell transplantation in light chain deposition disease. Nephrol Dial Transplant 23:2052–2057 Dores GM, Landgren O, McGlynn KA, Curtis RE, Linet MS, Devesa SS, 2009, Plasmacytoma of bone, extramedullary plasmacytoma, and multiple myeloma: incidence and survival in the United States, 1992–2004. Br J Haematol 144:86–94 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 249 EP 252 DI 10.1007/s12253-009-9218-1 PG 4 ER PT J AU Zhang, LY Jiang, LN Li, FF Li, H Liu, F Gu, Y Song, Y Zhang, F Ye, J Li, Q AF Zhang, Li-Ying Jiang, Li-Na Li, Fan-Fan Li, Hang Liu, Fang Gu, Yu Song, Yue Zhang, Feng Ye, Jing Li, Qing TI Reduced β-catenin Expression is Associated with Good Prognosis in Astrocytoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Astrocytoma; β-catenin; Prognosis; Immunohistochemistry ID Astrocytoma; β-catenin; Prognosis; Immunohistochemistry AB The aim of this study was to evaluate the expression of β-catenin in astrocytoma, and the clinical relevance and prognostic significance of the expression of β-catenin was also analyzed. Immunohistochemistry was performed on 63 resected astrocytoma tumor specimens to detect the expression of β-catenin. The correlation between the results of immuoexpression and the clinicopathologic parameters and patient survival was processed statistically. In 63 samples of astrocytoma, 36 cases were immunoreactive for β-catenin at cytoplasm, ten cases of astrocytoma were immunoreactive at cytomembrane, and four cases of astrocytoma were stained for β-catenin at nucleus. Spearman analysis showed that the distribution of β-catenin was not correlated with the grades of astrocytoma. However, the expression profiles were correlated with the patient’s 2-year survival, but not correlated with the grades, tumor size, sex, age, or tumor location. Patients with low β-catenin expression levels tended to be associated with a better prognosis than those who with high levels (p=0.042). Our results suggest that β-catenin is useful for the prognosis evaluation of astrocytoma. C1 [Zhang, Li-Ying] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. [Jiang, Li-Na] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. [Li, Fan-Fan] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. [Li, Hang] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. [Liu, Fang] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. [Gu, Yu] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. [Song, Yue] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. [Zhang, Feng] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. [Ye, Jing] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. [Li, Qing] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. RP Li, Q (reprint author), Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. EM liqing@fmmu.edu.cn CR Louis DN, 1997, A molecular genetic model of astrocytoma histopathology. Brain Pathol 7:755–764 Baeza N, Masuoka J, Kleihues P et al, 2003, AXIN1 mutations but not deletions in cerebellar medulloblastomas. Oncogene 22:632–636 Collins VP, 1995, Genetic alterations in gliomas. J Neurooncol 24:37–38 Nelson WJ, Nusse R, 2004, Convergence of Wnt, beta-catenin, and cadherin pathways. Science 303:1483–1487 Ilyas M, 2005, Wnt signalling and the mechanistic basis of tumour development. J Pathol 205:130–144 Van Aken E, De Wever O, Correia da Rocha AS et al, 2001, Defective E-cadherin/catenin complexes in human cancer. Virchows Arch 439:725–751 Lustig B, Behrens J, 2003, The Wnt signaling pathway and its role in tumor development. J Cancer Res Clin Oncol 129:199–221 Doucas H, Garcea G, Neal CP et al, 2005, Changes in the Wnt signalling pathway in gastrointestinal cancers and their prognostic significance. Eur J Cancer 41:365–379 Ueta T, Ikeguchi M, Hirooka Y et al, 2002, Beta-catenin and cyclin D1 expression in human hepatocellular carcinoma. Oncol Rep 9:1197–1203 Gerstein AV, Almeida TA, Zhao G et al, 2002, APC/CTNNB1, beta-catenin, pathway alterations in human prostate cancers. Genes Chromosomes Cancer 34:9–16 Ashihara K, Saito T, Mizumoto H et al, 2002, Mutation of betacatenin gene in endometrial cancer but not in associated hyperplasia. Med Electron Microsc 35:9–15 Utsuki S, Sato Y, Oka H et al, 2002, Relationship between the expression of E-, N-cadherins and beta-catenin and tumor grade in astrocytomas. J Neurooncol 57:187–192 Henderson BR, Fagotto F, 2002, The ins and outs of APC and beta-catenin nuclear transport. EMBO Rep 3:834–839 Novak A, Dedhar S, 1999, Signaling through beta-catenin and Lef/Tcf. Cell Mol Life Sci 56:523–537 Akiyama T, 2000, Wnt/beta-catenin signaling. Cytokine Growth Factor Rev 11:273–282 Polakis P, 2000, Wnt signaling and cancer. Genes Dev 14:1837– 1851 Morin PJ, 1999, beta-catenin signaling and cancer. Bioessays 21:1021–1030 Wijnhoven BP, Dinjens WN, Pignatelli M, 2000, E-cadherincatenin cell-cell adhesion complex and human cancer. Br J Surg 87:992–1005 Woo DK, Kim HS, Lee HS et al, 2001, Altered expression and mutation of beta-catenin gene in gastric carcinomas and cell lines. Int J Cancer 95:108–113 Grabsch H, Takeno S, Noguchi T et al, 2001, Different patterns of beta-catenin expression in gastric carcinomas: relationship with clinicopathological parameters and prognostic outcome. Histopathology 39:141–149 Zeng JZ, Hui Z, Rao HL, Zhang M, Hou J, Hui W, Qiu L, 2004, Expression of cytoskeleton proteins α- and β-catenin in gliomas. China Cancer 11:0738–0740 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 253 EP 257 DI 10.1007/s12253-009-9219-0 PG 5 ER PT J AU Li, M Gu, Y Zhang, Z Zhang, Sh Zhang, D Saleem, FA Zhao, X Sun, B AF Li, Man Gu, Yanjun Zhang, Zhiguang Zhang, Shiwu Zhang, Danfang Saleem, F Ali Zhao, Xiulan Sun, Baocun TI Vasculogenic Mimicry: a New Prognostic Sign of Gastric Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric adenocarcinoma; Hypoxia-inducible factor 1α; Microvascular density; Poorly differentiated; Tissue array; Vasculogenic mimicry ID Gastric adenocarcinoma; Hypoxia-inducible factor 1α; Microvascular density; Poorly differentiated; Tissue array; Vasculogenic mimicry AB Vasculogenic mimicry (VM) has been generally recognized as a new pattern of tumor neovascularization. It presents in many human malignancies. Till now, there is no report about VM in gastric adenocarcinoma (GAC). In this study, we collected 173 paraffinembedded human GAC samples, with detailed follow-up and clinicopathologic data. CD31/ periodic acid-Schiff (PAS) double staining, immunohistochemical staining of CK8 & 18 and laminin were performed to validate the existence of VM in GAC. Microvascular density (MVD) and vasulogenic mimicry density (VMD) were counted respectively. VM was observed in 40 of the 173 GAC patients, especially in poorly differentiated GAC (P=0.014). Patients with VM were prone to hematogenous metastasis and distant recurrence compared with patients without VM (P=0.020, 0.029). Higher VMD values was also associated with hematogenous metastasis (P=0.003). Immunohistochemical staining index (SI) of hypoxiainducible factor 1α (HIF-1α), vascular endothelial growth factor (VEGF), matrix metalloproteinase (MMP)-2, and MMP-9 were compared between the VM and non-VM group. The SI of four factors were all higher in the VM group than those of non-VM group (P=0.000, 0.000, 0.004, 0.009, respectively). The Kaplan-Meier survival analysis showed that the VM group has shorter life span compared with non-VM group (P=0.022). Cox proportional hazards model indicated that the presence of VM and TNM stage were independent predictors of poor prognosis (P=0.039 and 0.004) for GAC. In conclusion, VM exists in GAC, especially in poorly differentiated GAC. Additionally, it is an unfavorable prognostic indictor for GAC. Hypoxia may play a role in VM formation in GAC. C1 [Li, Man] Tianjin Medical University, Department of Pathology, 300060 Tianjin, China. [Gu, Yanjun] Tianjin Medical University, Department of Pathology, 300060 Tianjin, China. [Zhang, Zhiguang] The Second Hospital of Tianjin Medical University, Department of Digestive, 300211 Tianjin, China. [Zhang, Shiwu] Tianjin Medical University, Department of Pathology, 300060 Tianjin, China. [Zhang, Danfang] Tianjin Medical University, Department of Pathology, 300060 Tianjin, China. [Saleem, F Ali] Tianjin Medical University, Department of Surgery, 300060 Tianjin, China. [Zhao, Xiulan] Tianjin Medical University, Department of Pathology, 300060 Tianjin, China. [Sun, Baocun] Tianjin Medical University, Department of Pathology, 300060 Tianjin, China. RP Sun, B (reprint author), Tianjin Medical University, Department of Pathology, 300060 Tianjin, China. EM baocunsun@eyou.com CR Maniotis AJ, Folberg R, Hess A et al, 1999, Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 155:739–752 Chang YS, di Tomaso E, McDonald DM et al, 2000, Mosaic blood vessels in tumors: frequency of cancer cells in contact with flowing blood. Proc Natl Acad Sci USA 97:14608–14613 Chen X, Maniotis AJ, Majumdar D et al, 2002, Uveal melanoma cell staining for CD34 and assessment of tumor vascularity. Invest Ophthalmol Vis Sci 43:2533–2539 Folberg R, Mehaffey M, Gardner LM et al, 1997, The microcirculation of choroidal and ciliary body melanomas. Eye 11:227–238 Folberg R, Hendrix MJ, Maniotis AJ, 2000, Vasculogenic mimicry and tumor angiogenesis. Am J Pathol 156:361–381 Sood AK, Seftor EA, FletcherMS et al, 2001)Molecular determinants of ovarian cancer plasticity. Am J Pathol 158:1279–1288 Liu C, Huang H, Donate F et al, 2002, Prostate-specific membrane antigen directed selective thrombotic infarction of tumors. Cancer Res 62:5470–5475 Shirakawa K, Tsuda H, Heike Y et al, 2001, Absence of endothelial cells, central necrosis, and fibrosis are associated with aggressive inflammatory breast cancer. Cancer Res 61:445– 451 Sun B, Zhang S, Zhang D et al, 2006, Vasculogenic mimicry is associated with high tumor grade, invasion and metastasis, and short survival in patients with hepatocellular carcinoma. Oncol Rep 16:693–698 Sun B, Zhang S, Zhao X et al, 2004, Vasculogenic mimicry is associated with poor survival in patients with mesothelial sarcomas and alveolar rhabdomyosarcomas. Int J Oncol 25:1609–1614 van der Schaft DW, Hillen F, Pauwels P et al, 2005, Tumor cell plasticity in Ewing sarcoma, an alternative circulatory system stimulated by hypoxia. Cancer Res 65:11520–11528 Sun B, Qie S, Zhang S et al, 2008, Role and mechanism of vasculogenic mimicry in gastrointestinal stromal tumors. Hum Pathol 39:846–856 Folberg R, Maniotis AJ, 2004, Vasculogenic mimicry. APMIS 112:508–525 Shirakawa K, Wakasugi H, Heike Y et al, 2002, Vasculogenic mimicry and pseudo-comedo formation in breast cancer. Int J Cancer 99:821–828 Pisani P, Parkin DM, Bray F et al, 1999, Estimates of the worldwide mortality from 25 cancers in 1990. Int J Cancer 83:18–29 Tanigawa N, Amaya H, Matsumura M et al, 1996, Extent of tumor vascularization correlates with prognosis and hematogenous metastasis in gastric carcinomas. Cancer Res 56:2671–2676 Zhao HC, Qin R, Chen XX et al, 2006, Microvessel density is a prognostic marker of human gastric cancer. World J Gastroenterol 12:7598–7603 Hasan J, Byers R, Jayson GC, 2002, Intra-tumoural microvessel density in human solid tumours. Br J Cancer 86:1566–1577 Issam Beyrouti M, Beyrouti R, Ben Amar M et al, 2007, Linitis plastica. Presse Med 36:1782–1786 Sood AK, Fletcher MS, Coffin JE et al, 2004, Functional role of matrix metalloproteinases in ovarian tumor cell plasticity. Am J Obstet Gynecol 190:899–909 Hendrix MJ, Seftor EA, Kirschmann DA et al, 2003, Remodeling of the microenvironment by aggressive melanoma tumor cells. Ann N Y Acad Sci 995:151–161 Hess AR, Seftor EA, Seftor RE et al, 2003, Phosphoinositide 3- kinase regulates membrane type 1-matrix metalloproteinase, MMP, and MMP-2 activity during melanoma cell vasculogenic mimicry. Cancer Res 63:4757–4762 Seftor RE, Seftor EA, Koshikawa N et al, 2001, Cooperative interactions of laminin 5 gamma2 chain, matrix metalloproteinase-2, and membrane type-1-matrix / metallo-proteinase are required for mimicry of embryonic vasculogenesis by aggressive melanoma. Cancer Res 61:6322–6327 Sun B, Zhang D, Zhang S et al, 2007, Hypoxia influences vasculogenic mimicry channel formation and tumor invasionrelated protein expression in melanoma. Cancer Lett 249:188– 197 Seftor EA, Meltzer PS, Kirschmann DA et al, 2002, Molecular determinants of human uveal melanoma invasion and metastasis. Clin Exp Metastasis 19:233–246 Hockel M, Vaupel P, 2001, Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 93:266–276 Liao D, Johnson RS, 2007, Hypoxia: a key regulator of angiogenesis in cancer. Cancer Metastasis Rev 26:281–290 Cabuk D, Basaran G, Celikel C et al, 2007, Vascular endothelial growth factor, hypoxia-inducible factor 1a and CD34 expressions in early-stage gastric tumors: relationship with pathological factors and prognostic impact on survival. Oncology 72:111–117 Mizokami K, Kakeji Y, Oda S et al, 2006, Clinicopathologic significance of hypoxia-inducible factor 1a overexpression in gastric carcinomas. J Surg Oncol 94:149–154 Griffiths EA, Pritchard SA, Welch IM et al, 2005, Is the hypoxiainducible factor pathway important in gastric cancer? Eur J Cancer 41:2792–2805 Zhang J, Gao Q, 2008, Morphology and mechanism research of gastric cancer vasculogenic mimicry. Journal of Chongqing Medical University 33:1063–1066 Park SH, Han JK, Kim TK et al, 1999, Unusual gastric tumors: radiologic-pathologic correlation. Radiographics 19:1435–1446 Kunisaki C, Akiyama H, Nomura M et al, 2006, Clinicopathologic properties of poorly-differentiated adenocarcinoma of the stomach: comparison of solid- and non-solid-types. Anticancer Res 26:639–646 Adachi Y, Yasuda K, Inomata M et al, 2000, Pathology and prognosis of gastric carcinoma: well versus poorly differentiated type. Cancer 89:1418–1424 Strosberg JR, Nasir A, Hodul P et al, 2008, Biology and treatment of metastatic gastrointestinal neuroendocrine tumors. Gastrointest Cancer Res 2:113–125 Sood AK, Fletcher MS, Hendrix MJ, 2002, The embryonic-like properties of aggressive human tumor cells. J Soc Gynecol Investig 9:2–9 Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100:57–70 Kim SJ, Rabbani ZN, Dewhirst MW et al, 2005, Expression of HIF-1α CA IX, VEGF, and MMP-9 in surgically resected nonsmall cell lung cancer. Lung Cancer 49:325–335 Malemud CJ, 2006, Matrix metalloproteinases, MMPs, in health and disease: an overview. Front Biosci 11:1696–1701 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 259 EP 266 DI 10.1007/s12253-009-9220-7 PG 8 ER PT J AU Giaginis, C Tsourouflis, G Zizi-Serbetzoglou, A Kouraklis, G Chatzopoulou, E Dimakopoulou, K Theocharis, ES AF Giaginis, Constantinos Tsourouflis, Gerasimos Zizi-Serbetzoglou, Adamantia Kouraklis, Gregorios Chatzopoulou, Elli Dimakopoulou, Konstantina Theocharis, E Stamatios TI Clinical Significance of Ephrin (Eph)-A1, -A2, -A4, -A5 and -A7 Receptors in Pancreatic Ductal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ephrins; Clinical significance; Clinicopathological parameters; Immunohistochemistry; Pancreatic adenocarcinoma; Prognostic marker ID Ephrins; Clinical significance; Clinicopathological parameters; Immunohistochemistry; Pancreatic adenocarcinoma; Prognostic marker AB Ephrin (Eph) receptors have been reported to be frequently overexpressed in a wide variety of cancer types, being associated with tumor growth, invasion, metastasis and angiogenesis. The aim of the present study was to evaluate the clinical significance of Eph-A1, -A2, -A4, -A5 and -A7 expression in pancreatic ductal adenocarcinoma. Eph-A1, -A2, -A4, -A5 and -A7 expression and staining intensity were assessed immunohistochemically in tumoral samples of 67 pancreatic adenocarcinoma patients and were statistically analyzed in relation to clinicopathological characteristics, tumor proliferative capacity and patients’ survival. Eph receptors were abundantly expressed in pancreatic ductal adenocarcinoma cases examined. Eph-A1 staining intensity was significantly associated with tumor size (pT, p=0.008) and tumor histopathological stage (pStage, p=0.012). Eph-A2 expression was significantly associated with patients’ age (p=0.007), while Eph-A4 and Eph-A5 with tumor proliferative capacity (p=0.019 and p=0.011, respectively). Pancreatic adenocarcinoma patients with moderate/intense Eph-A5 or Eph-A7 staining presented significantly shorter survival times compared to those with negative/mild one (log-rank test, p=0.024 and p=0.009, respectively). Multivariate analysis identified Eph-A5 and Eph-A7 staining intensity as independent prognostic factors (p=0.048 and p=0.004, respectively). In conclusion, the present study revealed that Eph receptors were associated with pancreatic cancer characteristics, supporting evidence for their potential clinical application in management and prognosis of pancreatic adenocarcinoma patients. C1 [Giaginis, Constantinos] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias str, 11527 Athens, Greece. [Tsourouflis, Gerasimos] University of Athens, Laikon Hospital, Second Department of Propedeutic Surgery, 17, Agiou Thoma str, 11527 Athens, Greece. [Zizi-Serbetzoglou, Adamantia] Tzaneio General Hospital, Department of Pathology, Zanni & Afentouli str, 18536 Piraeus, Greece. [Kouraklis, Gregorios] University of Athens, Laikon Hospital, Second Department of Propedeutic Surgery, 17, Agiou Thoma str, 11527 Athens, Greece. [Chatzopoulou, Elli] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias str, 11527 Athens, Greece. [Dimakopoulou, Konstantina] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias str, 11527 Athens, Greece. [Theocharis, E Stamatios] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias str, 11527 Athens, Greece. RP Theocharis, ES (reprint author), University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 11527 Athens, Greece. EM theocharis@ath.forthnet.gr CR Zhang J, Hughes SE, 2006, Role of the ephrin and Ephrin receptor tyrosine kinase families in angiogenesis and development of the cardiovascular system. J Pathol 208:453–461 Pasquale EB, 2008, Eph-ephrin bidirectional signaling in physiology and disease. Cell 133:38–52 Nakamoto M, Bergemann AD, 2002, Diverse role for the Eph family of receptor tyrosine kinases in carcinogenesis. Microscopy Res Technique 59:58–67 Surawska H, Ma PC, Salgia R, 2004, The role of ephrins and Eph receptors in cancer. Cytokine Growth Factor Rev 15:419–433 Cheng N, Brantley DM, Chen J, 2002, The ephrins and Eph receptors in angiogenesis. Cytokine Growth Factor Rev 13:75–85 Brandley-Sieders DM, Chen J, 2007, Eph receptor tyrosine kinase in angiogenesis: from development to disease. Angiogenesis 7:17–28 Castano J, Davalos V, Schwartz S Jr, Arango D, 2008, EPH receptors in cancer. Histol Histopathol 23:1011–1023 Ireton RC, Chen J, 2005, EphA2 receptor tyrosine kinase as a promising target for cancer therapeutics. Curr Cancer Drug Targets 5:149–157 Brantley-Sieders D, Schmidt S, Parker M, Chen J, 2004, Eph receptor tyrosine kinases in tumor and tumor microenvironment. Curr Pharm Des 10:3431–342 Heroult M, Schaffner F, Augustin HG, 2004, Eph receptor and ephrin ligand-mediated interactions during angiogenesis and tumor progression. Exp Cell Res 312:642–650 Jemal A, Siegel E, Ward E, Murray T, Xu J, Smigal C, Thun MJ, 2006, Cancer statistics, 2006. CA Cancer J Clin 56:106–130 Sener DB, Jessup JM, Colacchio T, 1999, Pancreatic cancer: a report of treatment and survival trends for 100, 313 patients diagnosed from 1985–95, using national cancer database. J Am Coll Surg 189:1–7 Sarkar FH, Banerjee S, Li Y, 2007, Pancreatic cancer: pathogenesis, prevention and treatment. Toxicol Appl Pharmacol 224:326–336 Hamilton SR, 2000, World health organization classification of tumours. Pathology and genetics tumours of the digestive system. IARC, Lyon Sobin LH, Wittekind C, 1997, TNM classification of malignant tumors, 5th edn. Wiley-Liss, New York Theocharis S, Giaginis C, Chatzopoulou E, Tsourouflis G, Samiou F, Kouraklis G, 2007, Clinical significance of ephrin receptor A1 in pancreatic adenocarcinoma. Virchows Archiv 451:374–374 Giaginis C, Vgenopoulou S, Tsourouflis G, Politi E, Kouraklis G, Theocharis S, 2009, Expression and clinical significance of focal adhesion kinase in the two distinct histological types, intestinal and diffuse, of human gastric adenocarcinoma. Pathol Oncol Res 15:173–181 Giaginis C, Daskalopoulou S, Vgenopoulou S, Sfiniadakis I, Kouraklis G, Theocharis S, 2009, Heat shock protein −27, −60 and −90 expression in gastric cancer: association with clinicopathological variables and patient survival. BMC Gastroenterol 9:14 Giaginis C, Davides D, Zarros A, Noussia O, Zizi-Serbetzoglou A, Kouraklis G, Theocharis S, 2008, Clinical significance of tumor-associated antigen RCAS1 expression in human pancreatic ductal adenocarcinoma. Dig Dis Sci 53:1728–1734 Duxbury MS, Ito H, Zinner MJ, Ashley SW, Whang EE, 2004, EphA2: a determinant of malignant cellular behavior and a potential therapeutic target in pancreatic adenocarcinoma. Oncogene 23:1448–1456 Duxbury MS, Ito H, Zinner MJ, Ashley SW, Whang EE, 2004, Ligation of EphA2 by Ephrin A1-Fc inhibits pancreatic adenocarcinoma cellular invasiveness. Biochem Biophys Res Commun 320:1096–1102 Mudali SV, Fu B, Lakkur SS, Luo M, Embuscado EE, Iacobuzio- Donahue CA, 2006, Patterns of EphA2 protein expression in primary and metastatic pancreatic carcinoma and correlation with genetic status. Clin Exp Metastasis 23:357–365 Abraham S, Knapp DW, Cheng L, Snyder PW, Mittal SK, Bangari DS, Kinch M, Wu L, Dhariwal J, Mohammed SI, 2006, Expression of EphA2 and Ephrin A-1 in carcinoma of the urinary bladder. Clin Cancer Res 12:353–360 Dong Y, Wang J, Sheng Z, Li G, Ma H, Wang X, Zhang R, Lu G, Hu Q, Sugimura H, Zhou X, 2009, Downregulation of EphA1 in colorectal carcinomas correlates with invasion and metastasis. Mod Pathol 22:151–160 Saito T, Masuda N, Miyazaki T, Kanoh K, Suzuki H, Shimura T, Asao T, Kuwano H, 2004, Expression of EphA2 and E-cadherin in colorectal cancer: correlation with cancer metastasis. Oncol Rep 11:605–611 Holm R, Knopp S, Suo Z, Trope C, Nesland JM, 2007, Expression of EphA2 and EphrinA-1 in vulvar carcinomas and its relation to prognosis. J Clin Pathol 60:1086–1091 Hafner C, Becker B, Landthaler M, Vogt T, 2006, Expression profile of Eph receptors and ephrin ligands in human skin and downregulation of EphA1 in nonmelanoma skin cancer. Mod Pathol 19:1369–1377 Li X, Wang Y, Wang Y, Zhen H, Yang H, Fei Z, Zhang J, Liu W, Wang Y, Zhang X, 2007, Expression of EphA2 in human astrocytic tumors: correlation with pathologic grade, proliferation and apoptosis. Tumour Biol 28:165–172 Xu F, ZhongW, Li J, Shanshen Z, Cui J, Nesland JM, Suo Z, 2005, Predictive value of EphA2 and EphrinA-1 expression in oesophageal squamous cell carcinoma. Anticancer Res 25:2943–2950 Han L, Dong Z, Qiao Y, Kristensen GB, Holm R, Nesland JM, Suo Z, 2005, The clinical significance of EphA2 and Ephrin A-1 in epithelial ovarian carcinomas. Gynecol Oncol 99:278–286 Thaker PH, Deavers M, Celestino J, Thornton A, Fletcher MS, Landen CN, Kinch MS, Kiener PA, Sood AK, 2004, EphA2 expression is associated with aggressive features in ovarian carcinoma. Clin Cancer Res 10:5145–5150 Lin YG, Han LY, Kamat AA, Merritt WM, Landen CN, Deavers MT, Fletcher MS, Urbauer DL, Kinch MS, Sood AK, 2007, EphA2 overexpression is associated with angiogenesis in ovarian cancer. Cancer 109:332–340 Herrem CJ, Tatsumi T, Olson KS, Shirai K, Finke JH, Bukowski RM, Zhou M, Richmond AL, Derweesh I, Kinch MS, Storkus WJ, 2005, Expression of EphA2 is prognostic of disease-free interval and overall survival in surgically treated patients with renal cell carcinoma. Clin Cancer Res 11:226–231 Yuan W, Chen Z, Wu S, Ge J, Chang S, Wang X, Chen J, Chen Z, 2009, Expression of EphA2 and E-cadherin in Gastric Cancer: Correlated with Tumor Progression and Lymphogenous Metastasis. Pathol Oncol Res, in press) Zeng G, Hu Z, Kinch MS, Pan CX, Flockhart DA, Kao C, Gardner TA, Zhang S, Li L, Baldridge LA, Koch MO, Ulbright TM, Eble JN, Cheng L, 2003, High-level expression of EphA2 receptor tyrosine kinase in prostatic intraepithelial neoplasia. Am J Pathol 163:2271–2276 Kinch MS, Moore MB, Harpole DH Jr, 2003, Predictive value of the EphA2 receptor tyrosine kinase in lung cancer recurrence and survival. Clin Cancer Res 9:613–618 Shao Z, Zhang WF, Chen XM, 2008, Shang ZJ, 2008, Expression of EphA2 and VEGF in squamous cell carcinoma of the tongue: correlation with the angiogenesis and clinical outcome. Oral Oncol 44:1110–1117 Kamat AA, Coffey D, Merritt WM, Nugent E, Urbauer D, Lin YG, Edwards C, Broaddus R, Coleman RL, Sood AK, 2009, EphA2 overexpression is associated with lack of hormone receptor expression and poor outcome in endometrial cancer. Cancer, in press) Miyazaki T, Kato H, Fukuchi M, Nakajima M, Kuwano H, 2003, EphA2 overexpression correlates with poor prognosis in esophageal squamous cell carcinoma. Int J Cancer 103:657–663 Wang LF, Fokas E, Bieker M, Rose F, Rexin P, Zhu Y, Pagenstecher A, Engenhart-Cabillic R, An HX, 2008, Increased expression of EphA2 correlates with adverse outcome in primary and recurrent glioblastoma multiforme patients. Oncol Rep 19:151–156 Wu D, Suo Z, Kristensen GB, Li S, Troen G, Holm R, Nesland JM, 2004, Prognostic value of EphA2 and EphrinA-1 in squamous cell cervical carcinoma. Gynecol Oncol 94:312–319 Wang LF, Fokas E, Juricko J, You A, Rose F, Pagenstecher A, Engenhart-Cabillic R, An HX, 2008, Increased expression of EphA7 correlates with adverse outcome in primary and recurrent glioblastoma multiforme patients. BMC Cancer 8:79 Iiizumi M, Hosokawa M, Takehara A, Chung S, Nakamura T, Katagiri T, Eguchi H, Ohigashi H, Ishikawa O, Nakamura Y, Nakagawa H, 2006, EphA4 receptor, overexpressed in pancreatic ductal adenocarcinoma, promotes cancer cell growth. Cancer Sci 97:1211–1216 Oki M, Yamamoto H, Taniguchi H, Adachi Y, Imai K, Shinomura Y, 2008, Overexpression of the receptor tyrosine kinase EphA4 in human gastric cancers. World J Gastroenterol 14:5650–5656 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 267 EP 276 DI 10.1007/s12253-009-9221-6 PG 10 ER PT J AU Luo, P Wang, N He, E Eriksson, S Zhou, J Hu, G Zhang, J Skog, S AF Luo, Pengcheng Wang, Naining He, Ellen Eriksson, Staffan Zhou, Ji Hu, Guozhu Zhang, Jie Skog, Sven TI The Proliferation Marker Thymidine Kinase 1 Level is High in Normal Kidney Tubule Cells Compared to other Normal and Malignant Renal Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Kidney; Renal cell carcinoma; Thymidine kinase 1; TK1; Ki-67 ID Kidney; Renal cell carcinoma; Thymidine kinase 1; TK1; Ki-67 AB The activity of the proliferation related enzyme thymidine kinase 1 (TK1) was reported to be 3-fold higher in extracts from normal kidney tissue as compare to renal carcinoma extracts [3]. To verify these unexpected results, determinations of the protein levels of TK1 in normal kidney and in samples from different types of renal cell carcinoma (RCC) were done with immunohistochemistry and Western blot analysis. Two anti-TK1 peptide antibodies reacting with different TK1 epitops were used. TK1 levels were high in tubule cells as compared to glomerulus cells and connective tissue cells, while an intermediary TK1 was observed in renal cell carcinoma (RCC) cells. Western blot analysis demonstrated high levels of TK1 in extract from normal kidney, and lower levels of TK1 in the RCC extracts. The specificity of TK1 staining was demonstrated in competition experiments with excess TK1 antigen. The high TK1 levels in normal kidney tubule cells suggest that they are in a form of activated G1-state. The relatively low TK1 level in RCC, representing TK1 expression in S-phase cells, is in accordance with the low overall proliferation rate of these tumors. These results suggest that cell cycle regulation of TK1 in normal tubule cells differ from that in other type of normal and malignant renal cells. C1 [Luo, Pengcheng] Renmin Hospital of Wuhan University, Department of NephrologyWuhan, China. [Wang, Naining] Karolinska University, Department of Pathology, 141 86 Stockholm, Sweden. [He, Ellen] Karolinska University Hospital, Department of Oncology, 141 86 Stockholm, Sweden. [Eriksson, Staffan] Biomedical Center, Swedish University of Agricultural Science, Department of Anatomy, Physiology & Biochemistry, 751 23 Uppsala, Sweden. [Zhou, Ji] Sino-Swed Molecular Bio-Medicine Research InstituteShenzhen, China. [Hu, Guozhu] Jiangxi People´s Hospital, Department of Central LaboratoryNanchang, China. [Zhang, Jie] Wuhan University, Renmin Hospital, Department of Urology, 238 Ziyang Road, 430060 Wuhan, Hubie Province, China. [Skog, Sven] Karolinska University Hospital, Department of Oncology, 141 86 Stockholm, Sweden. RP Zhang, J (reprint author), Wuhan University, Renmin Hospital, Department of Urology, 430060 Wuhan, China. EM zhangjie888@sina.com CR Rylova SN, Mirzaee S, Albertioni F, Eriksson S, 2007, Expression of deoxynucleoside kinases and 5′-nucleotidases in mouse tissues: implications for mitochondrial toxicity. Biochem Pharmacol 74:169–75 Oudard S, Levalois C, Andrieu JM, Bougaran J, Validire P, Thiounn N, Poupon MF, Fourme E, Chevillard S, 2002, Expression of genes involved in chemoresistance, proliferation and apoptosis in clinical samples of renal cell carcinoma and correlation with clinical outcome. Anticancer Res 22:121–128 Mizutani Y, Wada H, Yoshida O, Fukushima M, Nakao M, Miki T, 2003, Significance of thymidine kinase activity in renal cell carcinoma. J Urology 169:7 06–709 Mizutani Y, Wada H, Yoshida O, Fukushima M, Nonomura M, Nakao M, Miki T, 2003, Significance of thymidylate synthase activity in renal cell carcinoma. Clin Cancer Res 9:1453–1460 O’Neill KL, Zhang F, Li H, Fuja DG, Murray BK, 2007, Thymidine kinase 1-a prognostic and diagnostic indicator in ALL and AML patients. Leukemia 21:560–563 He Q, Fornander T, Johansson H, Johansson U, Hu GZ, Rutqvist LE, Skog S, 2006, Thymidine kinase 1 in serum predicts increased risk of distant or loco-regional recurrence following surgery in patients with early breast cancer. Anticancer Res 26:4753–4759 Vogetseder A, Picard N, Gaspert A, Walch M, Kaissling N, Le Hir M, 2007, The proliferation capacity of the renalproximal tubule involves the bulk of differentiated cells. Am J Physiol Cell Physiol, DOI 10.1152/ajpcell.00227 Vogetseder A, Palan T, Bacic D, Kaissling B, Hir ML, 2007, Proximal tubular epithelial cells are generated by division of differentiated cells in the healthy kidney. Am J Physiol Cell Physiol 292:C807–C813 Tornevik Y, Ullman B, Balzarini J, Wahren B, Eriksson S, 1995, Cytotoxicity of 3′-azido-3′-deoxythymidine correlates with 3′-azidothymidine- 5′-monophosphate, AZTMP, levels, whereas anti-human immunodeficiency virus, HIV, activity correlates with 3′-azidothymidine- 5′-triphosphate, AZTTP, levels in cultured CEM Tlymphoblastoid cells. Biochem Pharmacol 49:829–37 He Q, Mao Y, Wu J, Decker C, Merza M, Wang N, Eriksson S, Castro J, Skog S, 2004, Cytosolic thymidine kinase is a specific histopathologic tumour marker for breast carcinomas. Intern J Oncol 25:945–53 Gasparri F, Wang N, Skog S, Galvani A, Eriksson S, 2009, Thymidine kinase 1 expression define an activated G1 state of the cell cycle as revealed with site specific antibodies and ArrayScan assays. Eur J Cell Biology 88:779–785 Wu C, Yang R, Zhou J, Bao S, Zou L, Zhang P, Mao Y, Wu J, He Q, 2003, Production and characterisation of a novel chicken IgY antibody raised against C-terminal peptide from human thymidine kinase 1. J Immunological Methods 277:157–169 Ozono S, Miyao N, Igarashi T, Marumo K, Nakazawa H, Fukuda M, Tsushima T, Tokuda N, Kawamura J, Murai M, 2004, and Collaboration Group of Japanese Society of Renal Cancer Tumor Doubling Time of Renal Cell Carcinoma Measured by CT. Jpn J Clin Oncol 34:82–85 Letocha H, Eklov S, Gronowitz S, Norlen BJ, Nilsson S, 1996, Deoxythymidine kinase in the staging of prostatic adenocarcinoma. Prostate 29:15–19 Wu JP, Mao YR, Hu LX, Wang N, Wu CJ, He Q, Skog S, 2000, A new cell proliferating marker: Cytosolic thymidine kinase as compared to proliferating cell nuclear antigen in patients with colorectal carcinoma. Anticancer Res 20:4815–4820 Mao Y, Wu JP, Skog S, Eriksson S, Zhao YW, Zhou J, He Q, 2005, Expression of cell proliferating genes in patients with nonsmall cell lungcancer by immunohistochemistry and cDNA profiling. Oncology Report 13:837–46 Sherley JL, Kelly TJ, 1988, Regulation of human thymidine kinase during the cell cycle. J Biol Chem 263:8350–8358 Ke PY, Chang ZF, 2004, Mitotic degradation of human thymidine kinase 1 is dependent on the anaphase-promoting complex/ cyclosome- CDH1-mediated pathway. Mol Cell Biol 24:514–526 Young AN, Amin MB, Moreno CS, Lim SD, Cohen C, Petros JA, Marshall FF, Neish A, 2001, Expression profiling of renal epithelial neoplasms. A method for tumour classification and discovery of diagnostic molecular markers. Amer J Path 158:1639–1651 Ke PY, Kuo YY, Hu CM, Chang ZF, 2005, Control of dTTP pool size by anaphase promoting complex/cyclosome is essential for the maintenance of genetic stability. Genes Dev 19:1920–1933 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 277 EP 283 DI 10.1007/s12253-009-9222-5 PG 7 ER PT J AU Schunemann, PD Grivicich, I Regner, A Leal, FL de Araujo, RD Jotz, PG Fedrigo, AC Simon, D da Rocha, BA AF Schunemann, Pretto Daniel Grivicich, Ivana Regner, Andrea Leal, Freitas Lisiane de Araujo, Romani Daniela Jotz, Pereira Geraldo Fedrigo, Alexandre Carlos Simon, Daniel da Rocha, Brondani Adriana TI Glutamate Promotes Cell Growth by EGFR Signaling on U-87MG Human Glioblastoma Cell Line SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma multiforme; Glutamate; EGFR; Phospho-Akt; Cell proliferation ID Glioblastoma multiforme; Glutamate; EGFR; Phospho-Akt; Cell proliferation AB Accumulating evidences suggest that glutamate plays a key role in the proliferation and invasion of malignant glioblastoma (GBM) tumors. It has been shown that GBM cells release and exploit glutamate for proliferation and invasion through AMPA glutamate receptors. Additionally, amplification of the epidermal growth factor receptor (EGFR) gene occurs in 40–50% of GBM. Since, PI3K/Akt is considered one of the main intracellular pathways involved in EGFR activation, AKT functions could trigger EGFR signaling. Thus, we investigated whether EGFR-phospho-Akt pathway is involved on the glutamate inducing U-87MG human GBM cell line proliferation. For these purpose, we treated the U-87MG cell line with 5 to 200 mM of glutamate and assessed the number of viable cells by trypan blue dye exclusion test. An increase in cell number (50%) was found at 5 mM glutamate, while the addition of DNQX (500 μM), an antagonist of AMPA receptor, inhibited the effect of glutamate on the U87-MG cells proliferation. Also, at 5 mM glutamate we observed an increase on the EGFR and phospho-Akt contents evaluated by immunohistochemistry. Moreover, U-87MG cells treated with glutamate exhibited an increase about 2 times in the EGFR mRNA expression. While, in the presence of the anti-EGFR gefitinib (50 μM) or the PI3K inhibitor wortmannin (5 μM), the U-87MG proliferation was restored to control levels. Together, our data suggest that glutamate signaling mediated by AMPA receptor induces U-87MG human GBM cell line proliferation via EGFR-phospho-Akt pathway. C1 [Schunemann, Pretto Daniel] Universidade Luterana do Brasil, Centro de Pesquisas em Ciencias MedicasCanoas, RS, Brazil. [Grivicich, Ivana] Universidade Luterana do Brasil, Centro de Pesquisas em Ciencias MedicasCanoas, RS, Brazil. [Regner, Andrea] Universidade Luterana do Brasil, Centro de Pesquisas em Ciencias MedicasCanoas, RS, Brazil. [Leal, Freitas Lisiane] Universidade Luterana do Brasil, Centro de Pesquisas em Ciencias MedicasCanoas, RS, Brazil. [de Araujo, Romani Daniela] Universidade Luterana do Brasil, Centro de Pesquisas em Ciencias MedicasCanoas, RS, Brazil. [Jotz, Pereira Geraldo] Universidade Luterana do Brasil, Programa de Pos Graduacao em Diagnostico Genetico e Molecular, Av Farroupilha, 8001, Predio 22, 5° andar, 92245-900 Canoas, RS, Brazil. [Fedrigo, Alexandre Carlos] Universidade Luterana do Brasil, Centro de Pesquisas em Ciencias MedicasCanoas, RS, Brazil. [Simon, Daniel] Universidade Luterana do Brasil, Programa de Pos Graduacao em Diagnostico Genetico e Molecular, Av Farroupilha, 8001, Predio 22, 5° andar, 92245-900 Canoas, RS, Brazil. [da Rocha, Brondani Adriana] Universidade Luterana do Brasil, Centro de Pesquisas em Ciencias MedicasCanoas, RS, Brazil. RP da Rocha, BA (reprint author), Universidade Luterana do Brasil, Centro de Pesquisas em Ciencias Medicas, Canoas, Brazil. EM brondani@terra.com.br CR Gupta T, Sarin R, 2002, Poor-prognosis high-grade gliomas: evolving an evidence-based standard of care. Lancet Oncol 3:557–564 Nieder C, Astner ST, Mehta MP et al, 2008, Improvement, clinical course, and quality of life after palliative radiotherapy for recurrent glioblastoma. Am J Clin Oncol 31:300–305 Hadziahmetovic M, Lo SS, Clarke JW et al, 2008, Palliative treatment of poor prognosis patients with malignant gliomas. Expert Rev Anticancer Ther 8:125–132 Behin A, Hoang-Xuan K, Carpentier AF et al, 2003, Primary brain tumours in adults. Lancet 361:323–331 Gunther W, Skaftnesmo KO, Arnold H et al, 2003, Molecular approaches to brain tumour invasion. Acta Neurochir, Wien, 145:1029–1036 Hoi Sang U, Espiritu OD, Kelley PY et al, 1995, The role of the epidermal growth factor receptor in human gliomas: I. The control of cell growth. J Neurosurg 82:841–846 Chakravarti A, Chakladar A, Delaney MA et al, 2002, The epidermal growth factor receptor pathway mediates resistance to sequential administration of radiation and chemotherapy in primary human glioblastoma cells in a RAS-dependent manner. Cancer Res 62:4307–4315 Krishnan S, Rao RD, James CD et al, 2003, Combination of epidermal growth factor receptor targeted therapy with radiation therapy for malignant gliomas. Front Biosci 8:e1–13 Shinojima N, Tada K, Shiraishi S et al, 2003, Prognostic value of epidermal growth factor receptor in patients with glioblastoma multiforme. Cancer Res 63:6962–6970 Aldape KD, Ballman K, Furth A et al, 2004, Immunohistochemical detection of EGFRvIII in high malignancy grade astrocytomas and evaluation of prognostic significance. J Neuropathol Exp Neurol 63:700–707 Lammering G, Valerie K, Lin PS et al, 2004, Radiation-induced activation of a common variant of EGFR confers enhanced radioresistance. Radiother Oncol 72:267–273 Ohgaki H, Dessen P, Jourde B et al, 2004, Genetic pathways to glioblastoma: a population-based study. Cancer Res 64:6892–6899 Korshunov A, Sycheva R, Golanov A, 2005, The prognostic relevance of molecular alterations in glioblastomas for patients age <50 years. Cancer 104:825–832 Mellinghoff IK, Wang MY, Vivanco I et al, 2005, Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med 353:2012–2024 Arslantas A, Artan S, Oner U et al, 2007, Genomic alterations in low-grade, anaplastic astrocytomas and glioblastomas. Pathol Oncol Res 13:39–46 Brandt B, Meyer-Staeckling S, Schmidt H et al, 2006, Mechanisms of EGFR gene transcription modulation: relationship to cancer risk and therapy response. Clin Cancer Res 12:7252–7260 Sibilia M, Kroismayr R, Lichtenberger BM et al, 2007, The epidermal growth factor receptor: from development to tumorigenesis. Differentiation 75:770–787 Choe G, Horvath S, Cloughesy TF et al, 2003, Analysis of the phosphatidylinositol 3′-kinase signaling pathway in glioblastoma patients in vivo. Cancer Res 63:2742–2746 Ghosh MK, Sharma P, Harbor PC et al, 2005, PI3K-AKT pathway negatively controls EGFR-dependent DNA-binding activity of Stat3 in glioblastoma multiforme cells. Oncogene 24:7290–7300 Labrakakis C, Patt S, Hartmann J et al, 1998, Glutamate receptor activation can trigger electrical activity in human glioma cells. Eur J NeuroSci 10:2153–2162 Sontheimer H, 2008, A role for glutamate in growth and invasion of primary brain tumors. J Neurochem 105:287–295 Mahe C, Bernhard M, Bobirnac I et al, 2004, Functional expression of the serotonin 5-HT7 receptor in human glioblastoma cell lines. Br J Pharmacol 143:404–410 Ye ZC, Sontheimer H, 1999, Glioma cells release excitotoxic concentrations of glutamate. Cancer Res 59:4383–4391 Rzeski W, Turski L, Ikonomidou C, 2001, Glutamate antagonists limit tumor growth. Proc Natl Acad Sci USA 98:6372–6377 Ishiuchi S, Tsuzuki K, Yoshida Yet al, 2002, Blockage of Ca(2+)- permeable AMPA receptors suppresses migration and induces apoptosis in human glioblastoma cells. Nat Med 8:971–978 D’Onofrio M, Arcella A, Bruno V et al, 2003, Pharmacological blockade of mGlu2/3 metabotropic glutamate receptors reduces cell proliferation in cultured human glioma cells. J Neurochem 84:1288–1295 Aronica E, Gorter JA, Ijlst-Keizers H et al, 2003, Expression and functional role of mGluR3 and mGluR5 in human astrocytes and glioma cells: opposite regulation of glutamate transporter proteins. Eur J NeuroSci 17:2106–2118 Arcella A, Carpinelli G, Battaglia G et al, 2005, Pharmacological blockade of group II metabotropic glutamate receptors reduces the growth of glioma cells in vivo. Neuro-oncol 7:236–245 Takano T, Lin JH, Arcuino G et al, 2001, Glutamate release promotes growth of malignant gliomas. Nat Med 7:1010–1015 Lyons SA, Chung WJ, Weaver AK et al, 2007, Autocrine glutamate signaling promotes glioma cell invasion. Cancer Res 67:9463–9471 Ishiuchi S, Yoshida Y, Sugawara K et al, 2007, Ca2+-permeable AMPA receptors regulate growth of human glioblastoma via Akt activation. J Neurosci 27:7987–8001 Dziedzic B, Prevot V, Lomniczi A et al, 2003, Neuron-to-glia signaling mediated by excitatory amino acid receptors regulates ErbB receptor function in astroglial cells of the neuroendocrine brain. J Neurosci 23:915–926 Yoshida Y, Tsuzuki K, Ishiuchi S et al, 2006, Serum-dependence of AMPA receptor-mediated proliferation in glioma cells. Pathol Int 56:262–271 Sigalas E, Regan JD, Kramer PR et al, 2004, Survival of human periodontal ligament cells in media proposed for transport of avulsed teeth. Dent Traumatol 20:21–28 Chomczynski P, Sacchi N, 1987, Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem 162:156–159 Schmidt M, Bachhuber A, Victor A et al, 2003, p53 expression and resistance against paclitaxel in patients with metastatic breast cancer. J Cancer Res Clin Oncol 129:295–302 Knobbe CB, Reifenberger G, 2003, Genetic alterations and aberrant expression of genes related to the phosphatidyl-inositol- 3′-kinase/protein kinase B, Akt, signal transduction pathway in glioblastomas. Brain Pathol 13:507–518 Regner A, Schunemann DP, Grivicich I et al, 2005, Effects of toxic doses of glutamate on Cu-Zn and Mn/superoxide dismutases activities in human glioma cell lines. J Neurooncol 71:9–17 Campbell GL, Bartel R, Freidman HS et al, 1985, Effect of glutamate analogues on brain tumor cell lines. J Neurochem 45:1186–1192 Chen CJ, Liao SL, Kuo JS, 2000, Gliotoxic action of glutamate on cultured astrocytes. J Neurochem 75:1557–1565 Matute C, Alberdi E, Ibarretxe G et al, 2002, Excitotoxicity in glial cells. Eur J Pharmacol 447:239–246 Skog J, Wurdinger T, van Rijn S et al, 2008, Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol 10:1470–1476 Peavy RD, Chang MS, Sanders-Bush E et al, 2001, Metabotropic glutamate receptor 5-induced phosphorylation of extracellular signal-regulated kinase in astrocytes depends on transactivation of the epidermal growth factor receptor. J Neurosci 21:9619–9628 Taylor S, Srinivasan B, Wordinger RJ et al, 2003, Glutamate stimulates neurotrophin expression in cultured Muller cells. Brain Res Mol Brain Res 111:189–197 Prenzel N, Fischer OM, Streit S et al, 2001, The epidermal growth factor receptor family as a central element for cellular signal transduction and diversification. Endocr Relat Cancer 8:11– 31 Yarden Y, Sliwkowski M, 2001, Untangling the ErbB signaling network. Nat Rev Mol Cell Biol 2:27–137 Holbro T, Civenni G, Hynes NE, 2003, The ErbB receptors and their role in cancer progression. Exp Cell Res 284:99–110 Chung WJ, Lyons SA, Nelson GM et al, 2005, Inhibition of cystine uptake disrupts the growth of primary brain tumors. J Neurosci 25:7101–7110 de Groot JF, Liu TJ, Fuller G et al, 2005, The excitatory amino acid transporter-2 induces apoptosis and decreases glioma growth in vitro and in vivo. Cancer Res 65:1934–1940 Voelzke WR, Petty WJ, Lesser GJ, 2008, Targeting the epidermal growth factor receptor in high-grade astrocytomas. Curr Treat Options in Oncol 9:23–31 Shawver LK, Slamon D, Ullrich A, 2002, Smart drugs: tyrosine kinase inhibitors in cancer therapy. Cancer Cell 1:117–123 Viana-Pereira M, Lopes JM, Little S et al, 2008, Analysis of EGFR overexpression, EGFR gene amplification and the EGFRvIII mutation in Portuguese high-grade gliomas. Anticancer Res 28:913–920 Lokker NA, Sullivan CM, Hollenbach SJ et al, 2002, Plateletderived growth factor, PDGF, autocrine signaling regulates survival and mitogenic pathways in glioblastoma cells: evidence that the novel PDGF-C and PDGF-D ligands may play a role in the development of brain tumors. Cancer Res 62:3729–3735 Servidei T, Riccardi A, Sanguinetti M et al, 2006, Increased sensitivity to the platelet-derived growth factor, PDGF, receptor inhibitor STI571 in chemoresistant glioma cells is associated with enhanced PDGF-BB-mediated signaling and STI571-induced Akt inactivation. J Cell Physiol 208:220–228 Wang MY, Lu KV, Zhu S et al, 2006, Mammalian target of rapamycin inhibition promotes response to epidermal growth factor receptor kinase inhibitors in PTEN-deficient and PTENintact glioblastoma cells. Cancer Res 66:7864–7869 Mendelsohn J, Baselga J, 2003, Status of epidermal growth factor receptor antagonists in the biology and treatment of cancer. J Clin Oncol 21:2787–2799 Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100:57–70 Parsa AT, Holland EC, 2004, Cooperative translational control of gene expression by Ras and Akt in cancer. Trends Mol Med 10:607–613 Holland EC, Celestino J, Dai C et al, 2000, Combined activation of Ras and Akt in neural progenitors induces glioblastomas formation in mice. Nat Genet 25:55–57 Sonoda Y, Ozawa T, Aldape KD et al, 2001, Akt pathway activation converts anaplastic astrocytoma to glioblastomas multiforme in a human astrocyte model of glioma. Cancer Res 61:6674–6678 Sanson M, Thillet J, Hoang-Xuan K, 2004, Molecular changes in gliomas. Curr Opin Oncol 16:607–613 Vivanco I, Sawyers CL, 2002, The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer 2:489–501 Santini MT, Rainaldi G, 1999, Three-dimensional spheroid model in tumor biology. Pathobiology 67:148–157 Bates RC, Edwards NS, Yates JD, 2000, Spheroids and cell survival. Crit Rev Oncol Hematol 36:61–74 Ye ZC, Rothstein JD, Sontheimer H, 1999, Compromised glutamate transport in human glioma cells: reductionmislocalization of sodium-dependent glutamate transporters and enhanced activity of cystine-glutamate exchange. J Neurosci 19:10767–10777 Roslin M, Henriksson R, Bergstrom P et al, 2003, Baseline levels of glucose metabolites, glutamate and glycerol in malignant glioma assessed by stereotactic microdialysis. J Neurooncol 61:151–160 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2010 VL 16 IS 2 BP 285 EP 293 DI 10.1007/s12253-009-9223-4 PG 9 ER PT J AU Medinger, M Kleinschmidt, M Mross, K Wehmeyer, B Unger, C Schaefer, HE Weber, R Azemar, M AF Medinger, Michael Kleinschmidt, Manuela Mross, Klaus Wehmeyer, Barbara Unger, Clemens Schaefer, Hans-Eckart Weber, Renate Azemar, Marc TI c-kit (CD117) Expression in Human Tumors and its Prognostic Value: An Immunohistochemical Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE c-kit; Imatinib; Immunohistochemistry; Prognosis; Solid tumors; Tyrosine kinases ID c-kit; Imatinib; Immunohistochemistry; Prognosis; Solid tumors; Tyrosine kinases AB c-kit functions as a tyrosine kinase receptor and represents a target for small molecule kinase inhibitors. The expression pattern for c-kit was studied in different human tumor types to their correlation with prognosis. Paraffinembedded tumor tissues from 282 patients were analyzed immunohistochemically for c-kit expression. Survival and follow-up data were available from 192/282 (68%) patients. c-kit immunopositivity was found in 62/282 (22%) cases. ckit expression was found in 14/83 (17%) colorectal cancers, in 13/62 (21%) breast cancers, in 7/20 sarcomas (35%), in 5/14 (36%) renal cell carcinomas, in 2/12 ovarian cancers (17%) and in 2/12 (17%) hepatocellular carcinomas. We found no significant correlation between c-kit expression and prognosis although a trend to a worse prognosis in patients with c-kit positive tumors could be observed. Expression of c-kit was found in tumor samples with varying intensities and infrequently. C1 [Medinger, Michael] Albert-Ludwigs University Freiburg, Tumor Biology CenterFreiburg im Breisgau, Germany. [Kleinschmidt, Manuela] Albert-Ludwigs University Freiburg, Institute of PathologyFreiburg im Breisgau, Germany. [Mross, Klaus] Albert-Ludwigs University Freiburg, Tumor Biology CenterFreiburg im Breisgau, Germany. [Wehmeyer, Barbara] Novartis AGNurnberg, Germany. [Unger, Clemens] Albert-Ludwigs University Freiburg, Tumor Biology CenterFreiburg im Breisgau, Germany. [Schaefer, Hans-Eckart] Albert-Ludwigs University Freiburg, Institute of PathologyFreiburg im Breisgau, Germany. [Weber, Renate] Internal Medicine, Munchnerstr. 14, 86163 Augsburg, Germany. [Azemar, Marc] Albert-Ludwigs University Freiburg, Tumor Biology CenterFreiburg im Breisgau, Germany. RP Medinger, M (reprint author), Albert-Ludwigs University Freiburg, Tumor Biology Center, Freiburg im Breisgau, Germany. EM MedingerM@uhbs.ch CR Besmer P, Murphy JE, George PC, Qiu FH, Bergold PJ, Lederman L, Snyder HW Jr, Brodeur D, Zuckerman EE, Hardy WD, 1986, A new acute transforming feline retrovirus and relationship of its oncogene v-kit with the protein kinase gene family. Nature 320:415–421 Yarden Y, Kuang WJ, Yang-Feng T, Coussens L, Munemitsu S, Dull TJ, Chen E, Schlessinger J, Francke U, Ullrich A, 1987, Human proto-oncogene c-kit: a new cell surface receptor tyrosine kinase for an unidentified ligand. EMBO J 6:3341–3351 Ashman LK, 1999, The biology of stem cell factor and its receptor C-kit. Int J Biochem Cell Biol 31:1037–1051 Ronnstrand L, 2004, Signal transduction via the stem cell factor/ c-kit. Cell Mol Life Sci 61:2535–2548 Arber DA, Tamayo R, Weiss LM, 1998, Paraffin section detection of the c-kit gene product, CD117, in human tissues: value in the diagnosis of mast cell disorders. Hum Pathol 29:498–504 Lammie A, Drobnjak M, Gerald W, Saad A, Cote R, Cordon-Cardo C, 1994, Expression of c-kit and kit ligand proteins in normal human tissues. J Histochem Cytochem 42:1417–1425 Tsuura Y, Hiraki H, Watanabe K, Igarashi S, Shimamura K, Fukuda T, Suzuki T, Seito T, 1994, Preferential localization of ckit product in tissue mast cells, basal cells of skin, epithelial cells of breast, small cell lung carcinoma and seminoma/dysgerminoma in human: immunohistochemical study on formalin-fixed, paraffin-embedded tissues. Virchows Arch 424:135–141 Natali PG, Nicotra MR, Sures I, Santoro E, Bigotti A, Ullrich A, 1992, Expression of c-kit receptor in normal and transformed human nonlymphoid tissues. Cancer Res 52:6139–6143 Matsuda R, Takahashi T, Nakamura S, Sekido Y, Nishida K, Seto M, Seito T, Sugiura T, Ariyoshi Y, Takahashi T, Ueda R, 1993, Expression of the c-kit protein in human solid tumors and in corresponding fetal and adult normal tissues. Am J Pathol 142:339–346 Hornick JL, Fletcher CD, 2002, Immunohistochemical staining for KIT, CD117, in soft tissue sarcomas is very limited in distribution. Am J Clin Pathol 117:188–193 Chui X, Egami H, Yamashita J, Kurizaki T, Ohmachi H, Yamamoto S, Ogawa M, 1996, Immunohistochemical expression of the c-kit proto-oncogene product in human malignant and nonmalignant breast tissues. Br J Cancer 73:1233–1236 Rubin BP, Singer S, Tsao C, Duensing A, Lux ML, Ruiz R, Hibbard MK, Chen C-J, Xiao S, Tuveson DA, Demetri GD, Fletcher CDM, Fletcher JA, 2001, KIT activation is a ubiquitous feature of gastrointestinal stromal tumors. Cancer Res 61:8118– 8121 Fletcher CD, Berman JJ, Corless C, Gorstein F, Lasota J, Longley BJ, Miettinen M, O’Leary TJ, Remotti H, Rubin BP, Shmookler B, Sobin LH, Weiss SW, 2002, Diagnosis of gastrointestinal stromal tumors: a consensus approach. Hum Pathol 33:459–465 Blay P, Astudillo A, Buesa JM, Campo E, Abad M, Garcia-Garcia J, Miquel R, Marco V, Sierra M, Losa R, Lacave A, Brana A, Balbin M, Freije JM, 2004, Protein kinase C theta is highly expressed in gastrointestinal stromal tumors but not in other mesenchymal neoplasias. Clin Cancer Res 10:4089–4095 Miettinen M, Wang ZF, Lasota J, 2009, DOG1 antibody in the differential diagnosis of gastrointestinal stromal tumors: a study of 1840 cases. Am J Surg Pathol 33:1401–1408 Lippai N, Fule T, Nemeth T, Benedek G, Malyi I, Padi E, Sapi Z, 2008, Keratin-positive gastrointestinal stromal tumor of the stomach mimicking gastric carcinoma: diagnosis confirmed by c-kit mutation analysis. Diagn Mol Pathol 17:241–244 Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Ishiguro S, Kawano K, Hanada M, Kurata A, Takeda M, Muhammad Tunio G, Matsuzawa Y, Kanakura Y, Shinomura Y, Kitamura Y, 1998, Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 279:577–580 Heinrich MC, Griffith DJ, Druker BJ, Wait CL, Ott KA, Zigler AJ, 2000, Inhibition of c-kit receptor tyrosine kinase activity by STI 571, a selective tyrosine kinase inhibitor. Blood 96:925–932 Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, Lydon NB, Kantarjian H, Capdeville R, Ohno-Jones S, Sawyers CL, 2001, Efficacy and safety of a specific inhibitor of the bcr-abl tyrosine kinase in chronic myeloid leukemia. N Engl J Med 344:1031–1037 Kantarjian H, Sawyers C, Hochhaus A et al, 2002, Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N Engl J Med 346:645–652 Hochhaus A, Druker B, Sawyers C, Guilhot F, Schiffer CA, Cortes J, Niederwieser DW, Gambacorti-Passerini C, Stone RM, Goldman J, Fischer T, O’Brien SG, Reiffers JJ, Mone M, Krahnke T, Talpaz M, Kantarjian HM, 2008, Favorable long-term followup results over 6 years for response, survival, and safety with imatinib mesylate therapy in chronic-phase chronic myeloid leukemia after failure of interferon-alpha treatment. Blood 111:1039–1043 Joensuu H, Roberts PJ, Sarlomo-Rikala M, Andersson LC, Tervahartiala P, Tuveson D, Silberman S, Capdeville R, Dimitrijevic S, Druker B, Demetri GD, 2001, Effect of the tyrosine kinase inhibitor STI571 in a patient with a metastatic gastrointestinal stromal tumor. N Engl J Med 344:1052–1056 Demetri GD, von Mehren M, Blanke CD et al, 2002, Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 347:472–480 Heinrich MC, Corless CL, Demetri GD et al, 2003, Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumors. J Clin Oncol 21:4342–4349 Sammarco I, Capurso G, Coppola L, Bonifazi AP, Cassetta S, Fave GD, Carrara A, Grassi GB, Rossi P, Sette C, Geremia R, 2004, Expression of the proto-oncogene c-Kit in normal and tumor tissues from colorectal carcinoma patients. Int J Colorectal Dis 19:545–553 Preto A, Moutinho C, Velho S, Oliveria C, Rebocho AP, Figueiredo J et al, 2007, A subset of colorectal carcinoma expresses c-Kit protein independently of BRAF and/or K-Ras activation. Virchows Arch 450:619–26 Tsuda H, Tani Y, Weisenberger J, Kitada S, Haseqawa T, Murata T et al, 2005, Frequent KIT and epidermal growth factor receptor over-expression in undifferentiated type breast carcinomas with “stem cell-like” features. Cancer Sci 96:333–339 Charpin C, Giusiano S, Charfi S, Secq V, Carpentier S, Andrac L, Lavaut MN, Allasia C, Bonnier P, Garcia S, 2009, Quantitative immunohistochemical expression of c Kit in breast carcinomas is predictive of patients’ outcome. Br J Cancer 7:48–54 Schmandt RE, Broaddus R, Lu KH, Shvartsman H, Thornton A, Malpica A et al, 2003, Expression of c-ABL, c-Kit and plateletderived growth factor receptor-beta in ovarian serous carcinoma and normal ovarian surface epithelium. Cancer 98:758–764 Wei H, Zhao MQ, Dong W, Yang Y, Li JS, 2008, Expression of c-kit protein and mutational status of the c-kit gene in osteosarcoma and their clinicopathological significance. J Int Med Res 36:1008–1014 Simon R, Panussis S, Maurer R, Spichtin H, Glatz K, Tapia C, Mirlacher M, Rufle A, Torhorst J, Sauter G, 2004, KIT, CD117)- positive breast cancers are infrequent and lack KIT gene mutations. Clin Cancer Res 10:178–183 Tsutsui S, Yasuda K, Suzuki K, Takeuchi H, Nishizaki T, Higashi H, Era S, 2006, A loss of c-kit expression is associated with an advanced stage and poor prognosis in breast cancer. Br J Cancer 94:1874–1878 Friederichs J, von Weyhern CW, Rosenberg R, Doll D, Busch R, Lordick F, Siewert JR, Sarbia M, 2009, Immunohistochemical detection of receptor tyrosine kinases c-kit, EGF-R, and PDGF-R in colorectal adenocarcinomas. Langenbecks Arch Surg Mar 12. [Epub ahead of print] Went PT, Dirnhofer S, Bundi M, Mirlacher M, Schraml P, Mangialaio S, Dimitrijevic S, Kononen J, Lugli A, Simon R, Sauter G, 2004, Prevalence of KIT expression in human tumors. J Clin Oncol 22:4514–4522 Becker G, Schmitt-Graeff A, Ertelt V, Blum HE, Allgaier HP, 2007, CD117, c-Kit, expression in human hepatocellular carcinoma. Clin Oncol 19:204–208 Lugli A, Went P, Khanlari B, Nikolova Z, Dirnhofer S, 2004, Rare KIT, CD117, expression in multiple myeloma abrogates the usefulness of imatinib mesylate treatment. Virchows Arch 444:264–268 Rubin BP, Schuetze SM, Eary JF, Norwood TH, Mirza S, Conrad EU, Bruckner JD, 2002, Molecular targeting of platelet-derived growth factor B by imatinib mesylate in a patient with metastatic dermatofibrosarcoma protuberans. J Clin Oncol 20:3586–3591 Piccaluga PP, Malagola M, Rondoni M, Arpinati M, Paolini S, Candoni A, Fanin R, Messa E, Pirrotta MT, Lauria F, Visani G, Alberti D, Rancati F, Vinaccia V, Russo D, Saglio G, Baccarani M, Martinelli G, 2007, Imatinib mesylate in the treatment of newly diagnosed or refractory/resistant c-kit positive acute myeloid leukemia. Results of an Italian multicentre phase II study. Haematologica 92:1721–1722 Coleman RL, Broaddus RR, Bodurka DC, Wolf JK, Burke TW, Kavanagh JJ, Levenback CF, Gershenson DM, 2006, Phase II trial of imatinib mesylate in patients with recurrent platinum-and taxane-resistant epithelial ovarian and primary peritoneal cancers. Gynecol Oncol 101:126–131 Dy GK, Miller AA, Mandrekar SJ, Aubry MC, Langdon RM Jr, Morton RF, Schild SE, Jett JR, Adjei AA, 2005, A phase II trial of imatinib, ST1571, in patients with c-kit expressing relapsed small cell lung cancer: a CALGB and NCCTG study. Ann Oncol 16:1811–1816 Heinrich MC, Joensuu H, Demetri GD, Corless CL, Apperley J, Fletcher JA, Soulieres D, Dirnhofer S, Harlow A, Town A, McKinley A, Supple SG, Seymour J, Di Scala L, van Oosterom A, Herrmann R, Nikolova Z, McArthur AG, 2008, Imatinib Target Exploration Consortium Study B2225. Phase II, open-label study evaluating the activity of imatinib in treating life-threatening malignancies known to be associated with imatinib-sensitive tyrosine kinases. Clin Cancer Res 14:2717–2531 Sihto H, Sarlomo-Rikala M, Tynninen O, Tanner M, Andersson LC, Franssila K, Nupponen NN, Joensuu H, 2005, KIT and platelet-derived growth factor receptor alpha tyrosine kinase gene mutations and KIT amplifications in human solid tumors. J Clin Oncol 23:49–57 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 295 EP 301 DI 10.1007/s12253-010-9247-9 PG 7 ER PT J AU Ladanyi, A Mohos, A Somlai, B Liszkay, G Gilde, K Fejos, Zs Gaudi, I Timar, J AF Ladanyi, Andrea Mohos, Anita Somlai, Beata Liszkay, Gabriella Gilde, Katalin Fejos, Zsuzsanna Gaudi, Istvan Timar, Jozsef TI FOXP3+ Cell Density in Primary Tumor Has No Prognostic Impact in Patients with Cutaneous Malignant Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE FOXP3; Immunohistochemistry; Melanoma; Prognosis; Regulatory T cell ID FOXP3; Immunohistochemistry; Melanoma; Prognosis; Regulatory T cell AB Regulatory T cells (Tregs) have been implicated as inhibitors of antitumor immune reactions. However, data on the relevance of their prevalence at tumor sites in influencing disease outcome are controversial. The aim of our study was to investigate the role in tumor progression and the prognostic impact of the density of lymphocytes expressing FOXP3, a transcription factor expressed predominantly by CD4+CD25+ Tregs, in primary cutaneous melanoma. We examined the infiltration of FOXP3+ cells by immunohistochemistry in tumor samples from 97 patients and evaluated in relation to patient and tumor parameters. The degree of infiltration by FOXP3+ cells did not show correlation with the thickness of melanomas. Moreover, no associations were found with metastasis formation during the 5-year follow-up period, patient survival, or any other clinicopathologic parameters studied. These results suggest that the presence of FOXP3+ lymphocytes in primary tumors is not of prognostic importance in human cutaneous melanoma. C1 [Ladanyi, Andrea] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, 7–9. Rath Gyorgy u, 1122 Budapest, Hungary. [Mohos, Anita] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Somlai, Beata] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Liszkay, Gabriella] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Gilde, Katalin] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Fejos, Zsuzsanna] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Gaudi, Istvan] National Institute of Oncology, National Cancer RegistryBudapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Ladanyi, A (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, 1122 Budapest, Hungary. EM ladanyi@oncol.hu CR Paschen A, Eichmuller S, Schadendorf D, 2004, Identification of tumor antigens and T-cell epitopes, and its clinical application. Cancer Immunol Immunother 53:196–203 Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S, 2000, Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Adv Immunol 74:181–273 Zou W, 2005, Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer 5:263–274 Knutson KL, Disis ML, Salazar LG, 2007, CD4 regulatory T cells in human cancer pathogenesis. Cancer Immunol Immunother 56:271–285 Curiel TJ, Coukos G, Zou L et al, 2004, Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 10:942–949 Hiraoka N, Onozato K, Kosuge T, Hirohashi S, 2006, Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clin Cancer Res 12:5423–5434 Bates GJ, Fox SB, Han C et al, 2006, Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J Clin Oncol 24:5373–5380 Kobayashi N, Hiraoka N, Yamagami W et al, 2007, FOXP3+ regulatory T cells affect the development and progression of hepatocarcinogenesis. Clin Cancer Res 13:902–911 Gao Q, Qiu S-J, Fan J et al, 2007, Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J Clin Oncol 25:2586–2593 Fox SB, Launchbury R, Bates GJ et al, 2007, The number of regulatory T cells in prostate cancer is associated with the androgen receptor and hypoxia-inducible factor, HIF)-2α but not HIF-1α. Prostate 67:623–629 Siddiqui SA, Frigola X, Bonne-Annee S et al, 2007, Tumorinfiltrating Foxp3−CD4+CD25+ T cells predict poor survival in renal cell carcinoma. Clin Cancer Res 13:2075–2081 Badoual C, Hans S, Rodriguez J et al, 2006, Prognostic value of tumor-infiltrating CD4+ T-cell subpopulations in head and neck cancer. Clin Cancer Res 12:465–472 Salama P, Phillips M, Grieu F et al, 2009, Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol 27:186–192 Alvaro T, Lejeune M, Salvado MT et al, 2005, Outcome in Hodgkin’s lymphoma can be predicted from the presence of accompanying cytotoxic and regulatory T cells. Clin Cancer Res 11:1467–1473 Carreras J, Lopez-Guillermo A, Fox BC et al, 2006, High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma. Blood 108:2957–2964 Gjerdrum LM, Woetmann A, Odum N et al, 2007, FOXP3+ regulatory T cells in cutaneous T-cell lymphomas: association with disease stage and survival. Leukemia 21:2512–2518 Lee NR, Song EK, Jang KY et al, 2008, Prognostic impact of tumor infiltrating FOXP3 positive regulatory T cells in diffuse large B-cell lymphoma at diagnosis. Leuk Lymphoma 49:247–256 Mourmouras V, Fimiani M, Rubegni P et al, 2007, Evaluation of tumour-infiltrating CD4+CD25+FOXP3+ regulatory T cells in human cutaneous benign and atypical naevi, melanomas and melanoma metastases. Br J Dermatopathol 157:531–539 De Panfilis G, Campanini N, Santini M et al, 2008, Phase- and stage-related proportions of T cells bearing the transcription factor FOXP3 infiltrate primary melanoma. J Invest Dermatol 128:676– 684 Miracco C, Mourmouras V, Biagioli M et al, 2007, Utility of tumour-infiltrating CD25+FOXP3+ regulatory T cell evaluation in predicting local recurrence in vertical growth phase cutaneous melanoma. Oncol Rep 18:1115–1122 Hillen F, Baeten CIM, van de Winkel A et al, 2008, Leukocyte infiltration and tumor cell plasticity are parameters of aggressiveness in primary cutaneous melanoma. Cancer Immunol Immunother 57:97–106 Ladanyi A, Somlai B, Gilde K et al, 2004, T-cell activation marker expression on tumor-infiltrating lymphocytes as prognostic factor in cutaneous malignant melanoma. Clin Cancer Res 10:521–530 Ladanyi A, Kiss J, Somlai B et al, 2007, Density of DC-LAMP+ mature dendritic cells in combination with activated T lymphocytes infiltrating primary cutaneous melanoma is a strong independent prognostic factor. Cancer Immunol Immunother 56:1459–1469 Balch CM, Buzaid AC, Soong S-J et al, 2001, Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 19:3635–3648 Ebert LM, Tan BS, Browning J et al, 2008, The regulatory T cellassociated transcription factor FOXP3 is expressed by tumor cells. Cancer Res 68:3001–3009 Morgan ME, van Bilsen JH, Bakker AM et al, 2005, Expression of FOXP3 mRNA is not confined to CD4+CD25+ T regulatory cells in humans. Hum Immunol 66:13–20 Roncador G, Brown PJ, Maestre L et al, 2005, Analysis of FOXP3 protein expression in human CD4+CD25+ regulatory T cells at the single-cell level. Eur J Immunol 35:1681–1691 Wang J, Ioan-Facsinay A, van der Voort EI et al, 2007, Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur J Immunol 37:129–138 Fontenot JD, Gavin MA, Rudensky AY, 2003, Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4:330–336 Fontenot JD, Rasmussen JP, Williams LM et al, 2005, Regulatory T cell lineage specification by the forkhead transcription factor Foxp3. Immunity 22:329–341 Hori S, Nomura T, Sakaguchi S, 2003, Control of regulatory T cell development by the transcription factor Foxp3. Science 299:1057–1061 Yagi H, Nomura T, Nakamura K et al, 2004, Crucial role of FOXP3 in the development and function of human CD25+CD4+ regulatory T cells. Int Immunol 16:1643–1656 Walker MR, Kasprowicz DJ, Gersuk VH et al, 2003, Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+CD25− T cells. J Clin Invest 112:1437–1443 Allan SE, Passerini L, Bacchetta R et al, 2005, The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. J Clin Invest 115:3276–3284 Allan SE, Crome SQ, Crellin NK et al, 2007, Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int Immunol 19:345–354 Ziegler SF, 2006, FOXP3: of mice and men. Annu Rev Immunol 24:209–226 Kryczek I, Liu R, Wang G et al, 2009, FOXP3 defines regulatory T cells in human tumor and autoimmune disease. Cancer Res 69:3995–4000 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 303 EP 309 DI 10.1007/s12253-010-9254-x PG 7 ER PT J AU Wincewicz, A Koda, M Sulkowski, S Kanczuga-Koda, L Sulkowska, M AF Wincewicz, Andrzej Koda, Mariusz Sulkowski, Stanislaw Kanczuga-Koda, Luiza Sulkowska, Mariola TI Comparison of Beta-catenin with TGF-beta1, HIF-1alpha and Patients’ Disease-free Survival in Human Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Beta-catenin; TGF-beta1; HIF-1alpha; Patients’ disease free survival; Colorectal cancer ID Beta-catenin; TGF-beta1; HIF-1alpha; Patients’ disease free survival; Colorectal cancer AB Beta-catenin accumulation is suppressed by TGFbeta1 (transforming growth factor beta1) in intestinal epithelium suggesting negative feedback between these two factors. Besides that, beta-catenin interacts with HIF-1alpha (hypoxiainducible factor-1alpha) at the promoter region of HIF-1 target genes. Our study was aimed at comparison of beta-catenin with HIF-1alpha, TGF-beta1, Ki67 and survival of sporadic colorectal cancer patients. Expressions of beta-catenin, TGFbeta1, HIF-1alpha, Ki67 were evaluated in triads of specimens of each primary tumor of 72 sporadic colorectal cancers with immunohistochemistry due to limited availability of tissue material. Disease-free survival was analyzed in case of all 100 beta-catenin stained tumors, in 85 cancers stained for HIF-1 and in 72 neoplasms with TGFbeta1 staining. Beta-catenin, TGF-beta1 and HIF-1alpha accumulated in 72 colorectal cancer cells. Beta-catenin correlated both with HIF-1alpha and TGF-beta1 in all colorectal cancers (p<0.009, r=0.307 and p=0.003, r=0.342, respectively) and in subgroups of different clinico-pathological profile. Beta-catenin failed to correlate with Ki67. In case of beta-catenin, TGF-beta1 and HIF-1alpha, disease-free survival curves failed to show any statistically significant differences between groups of marker negative tumors, cancers with low expression and neoplasms with higher protein expression. Positive correlations between beta-catenin and TGF-beta1 may indicate ineffective attempts of TGF-beta1 to reduce intracellular level of beta-catenin in colorectal cancer. Associations between beta-catenin and HIF-1alpha reflect previously detected interactions between HIF-1alpha with beta-catenin and are confirmative for presence of such reactions in human colorectal cancer. C1 [Wincewicz, Andrzej] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Koda, Mariusz] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Sulkowski, Stanislaw] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Kanczuga-Koda, Luiza] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. [Sulkowska, Mariola] Medical University of Bialystok, Department of General Pathomorphology, Waszyngtona St 13, 15-269 Bialystok, Poland. RP Wincewicz, A (reprint author), Medical University of Bialystok, Department of General Pathomorphology, 15-269 Bialystok, Poland. EM andwinc@gmail.com;ruahpolin@yahoo.com CR Wong NA, Pignatelli M, 2002, Beta-catenin-a linchpin in colorectal carcinogenesis? Am J Pathol 160:389–401 Luchtenborg M, Weijenberg MP, Wark PA, Saritas AM, Roemen GM, van Muijen GN, de Bruine AP, van den Brandt PA, de Goeij AF, 2005, Mutations in APC, CTNNB1 and Kras genes and expression of hMLH1 in sporadic colorectal carcinomas from the Netherlands Cohort Study. BMC Cancer 5:160 Sulkowska M, Wincewicz A, Sulkowski S, Koda M, Kanczuga- Koda L, 2009, Relations of TGF-beta1 with HIF-1alpha, GLUT-1 and longer survival of colorectal cancer patients. Pathology Jan 13:1–7. [Epub ahead of print] Xu J, Attisano L, 2000, Mutations in the tumor suppressors Smad2 and Smad4 inactivate transforming growth factor beta signaling by targeting Smads to the ubiquitin-proteasome pathway. Proc Natl Acad Sci USA 97:4820–4825 Li F, Cao Y, Townsend CM Jr, Ko TC, 2005, TGF-beta signaling in colon cancer cells. World J Surg 29:306–311 Bacman D, Merkel S, Croner R, Papadopoulos T, Brueckl W, Dimmler A, 2007, TGF-beta receptor 2 downregulation in tumour-associated stroma worsens prognosis and high-grade tumours show more tumour-associated macrophages and lower TGF-beta1 expression in colon carcinoma: a retrospective study. BMC Cancer 7:156 Giles RH, Lolkema MP, Snijckers CM, Belderbos M, van der Groep P, Mans DA, van Beest M, van Noort M, Goldschmeding R, van Diest PJ, Clevers H, Voest EE, 2006, Interplay between VHL/HIF1alpha and Wnt/beta-catenin pathways during colorectal tumorigenesis. Oncogene 25:3065–3070 de la Roche M, Worm J, Bienz M, 2008, The function of BCL9 in Wnt/beta-catenin signaling and colorectal cancer cells. BMC Cancer 8:199 Lim JH, Chun YS, Park JW, 2008, Hypoxia-inducible factor- 1alpha obstructs a Wnt signaling pathway by inhibiting the hARD1-mediated activation of beta-catenin. Cancer Res 68:5177–5184 Li LN, Zhang HD, Yuan SJ, Tian ZY, Wang L, Sun ZX, 2007, Artesunate attenuates the growth of human colorectal carcinoma and inhibits hyperactive Wnt/beta-catenin pathway. Int J Cancer 121:1360–1365 Kaidi A, Williams AC, Paraskeva C, 2007, Interaction between beta-catenin and HIF-1 promotes cellular adaptation to hypoxia. Nat Cell Biol 9:210–217 Wincewicz A, Sulkowska M, Koda M, Sulkowski S, 2007, Cumulative expression of HIF-1-alpha, Bax, Bcl-xL and P53 in human colorectal cancer. Pathology 39:334–338 Wincewicz A, Sulkowska M, Koda M, Sulkowski S, 2007, Clinicopathological significance and linkage of the distribution of HIF-1alpha and GLUT-1 in human primary colorectal cancer. Pathol Oncol Res 13:15–20 Wang H, Promkan M, Liu G, Chakrabarty S, 2008, Switch of transforming growth factor beta function from tumor suppression to stimulation in adenomatous polyposis coli, APC, knocked-down human colon carcinoma cells. Cancer Lett 272:253–259 Wang H, Rajan S, Liu G, Chakrabarty S, 2008, Transforming growth factor beta suppresses beta-catenin/Wnt signaling and stimulates an adhesion response in human colon carcinoma cells in a Smad4/DPC4 independent manner. Cancer Lett 264:281–287 Lambie DL, Brown IS, 2008, Multinucleate epithelial change in colorectal hyperplastic polyps: a review of 27 cases. J Clin Pathol 61:611–614 Settakorn J, Kaewpila N, Burns GF, Leong AS, 2005, FAT, Ecadherin, beta catenin, HER 2/neu, Ki67 immuno-expression, and histological grade in intrahepatic cholangiocarcinoma. J Clin Pathol 58:1249–1254 Pirinen RT, Hirvikoski P, Johansson RT, Hollmen S, Kosma VM, 2001, Reduced expression of alpha-catenin, beta-catenin, and gamma-catenin is associated with high cell proliferative activity and poor differentiation in non-small cell lung cancer. J Clin Pathol 54:391–395 Wong SC, Chan AT, Chan JK, Lo YM, 2006, Nuclear betacatenin and Ki-67 expression in choriocarcinoma and its premalignant form. J Clin Pathol 59:387–392 Takeda K, Kinoshita I, Shimizu Y, Ohba Y, Itoh T, Matsuno Y, Shichinohe T, Dosaka-Akita H, 2008, Clinicopathological significance of expression of p-c-Jun, TCF4 and beta-catenin in colorectal tumors. BMC Cancer 8:328 Tien YW, Jeng YM, Hu RH, Chang KJ, Hsu SM, Lee PH, 2004, Intravasation-related metastatic factors in colorectal cancer. Tumour Biol 25:48–55 Pecina-Slaus N, Niku Eva-Martic T, Beros V et al, 2007, Genetic alterations of E-cadherin and beta-catenin in germinoma and teratoma: report of two central nervous system cases. Pathol Oncol Res 13:370–374 Kabukcuoglu F, Kabukcuoglu Y, Tanik C et al, 2008, Breast carcinoma metastasis in recurrent myxoid liposarcoma. Pathol Oncol Res Horst D, Kriegl L, Engel J et al, 2009, CD133 and nuclear betacatenin: the marker combination to detect high risk cases of low stage colorectal cancer. Eur J Cancer Horst D, Reu S, Kriegl L et al, 2009, The intratumoral distribution of nuclear beta-catenin is a prognostic marker in colon cancer. Cancer 115:2063–2070 Bondi J, Bukholm G, Nesland JM, Bakka A, Bukholm IR, 2006, An increase in the number of adhesion proteins with altered expression is associated with an increased risk of cancer death for colon carcinoma patients. Int J Colorectal Dis 21:231–237 Canavese G, Bernardi A, Candelaresi G, Lovadina P, Amerio S, Rossetti V, Rabagliati C, Berardengo E, 2007, Expression of the E-cadherin-catenins complex in sentinel node is related to tumor morphology but not to spread to nonsentinel nodes. Pathol Res Pract 203:517–523 Stewart CJ, Crook ML, Leung YC, Platten M, 2009, Expression of cell cycle regulatory proteins in endometrial adenocarcinoma: variations in conventional tumor areas and in microcystic, elongated and fragmented glands. Mod Pathol, Mar 6. [Epub ahead of print] Nagayama S, Yamada E, Kohno Y, Aoyama T, Fukukawa C, Kubo H, Watanabe G, Katagiri T, Nakamura Y, Sakai Y, Toguchida J, 2009, Inverse correlation of the up-regulation of FZD10 expression and the activation of beta-catenin in synchronous colorectal tumors. Cancer Sci 100:405–412 Mees ST, Mennigen R, Spieker T, Rijcken E, Senninger N, Haier J, Bruewer M, 2009, Expression of tight and adherens junction proteins in ulcerative colitis associated colorectal carcinoma: upregulation of claudin-1, claudin-3, claudin-4, and beta-catenin. Int J Colorectal Dis 24:361–368 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 311 EP 318 DI 10.1007/s12253-009-9217-2 PG 8 ER PT J AU Guo, F Liu, Y Huang, J Li, Y Zhou, G Wang, D Li, Y Wang, J Xie, P Li, G AF Guo, Fengjie Liu, Yan Huang, Jian Li, Yuehui Zhou, Guohua Wang, Di Li, Yalin Wang, Jiajia Xie, Pingli Li, Guancheng TI Identification of Rho GTPase Activating Protein 6 Isoform 1 Variant as a New Molecular Marker in Human Colorectal Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE RhoGAP6 isform 1 variant; Colorectal cancer; Mass spectrometry; Biomarker; Monoclonal antibody ID RhoGAP6 isform 1 variant; Colorectal cancer; Mass spectrometry; Biomarker; Monoclonal antibody AB The early diagnosis of colorectal cancer (CRC) is important because it is one of the most readily curable of all cancers, if detected early. However, the sensitivity of current markers is low. Immunostaining intensity for the monoclonal antibody Hb3 in CRC cell lines and tissues was stronger than in controls. Interestingly, this was associated with a low level of tumor differentiation. We used Hb3-coupled affinity chromatography to search for a corresponding Hb3 antigen as a candidate biomarker for early detection, and identified a Rho GTPase activating protein 6 (RhoGAP6) isoform 1 variant as an Hb3 antigen by mass spectrometry. Using reverse transcription polymerase chain reaction and western blot analysis, we confirmed that the expression levels of this variant were elevated in aberrant cells and tissues. Thus, the RhoGAP6 isoform 1 variant might serve as a biomarker for the development and progression of CRC. C1 [Guo, Fengjie] Central South University, Xiangya Medical School, Cancer Research Institute, 110 Xiangya Road, 410078 Changsha, Hunan Province, China. [Liu, Yan] Central South University, Xiangya Medical School, Cancer Research Institute, 110 Xiangya Road, 410078 Changsha, Hunan Province, China. [Huang, Jian] Central South University, Xiangya Medical School, Cancer Research Institute, 110 Xiangya Road, 410078 Changsha, Hunan Province, China. [Li, Yuehui] Central South University, Xiangya Medical School, Cancer Research Institute, 110 Xiangya Road, 410078 Changsha, Hunan Province, China. [Zhou, Guohua] Central South University, Xiangya Medical School, Cancer Research Institute, 110 Xiangya Road, 410078 Changsha, Hunan Province, China. [Wang, Di] The First Hospital of Changsha, Department of PathologyChangsha, Hunan Province, China. [Li, Yalin] Central South University, Xiangya Medical School, Cancer Research Institute, 110 Xiangya Road, 410078 Changsha, Hunan Province, China. [Wang, Jiajia] Central South University, Xiangya Medical School, Cancer Research Institute, 110 Xiangya Road, 410078 Changsha, Hunan Province, China. [Xie, Pingli] Central South University, Xiangya Medical School, Cancer Research Institute, 110 Xiangya Road, 410078 Changsha, Hunan Province, China. [Li, Guancheng] Central South University, Xiangya Medical School, Cancer Research Institute, 110 Xiangya Road, 410078 Changsha, Hunan Province, China. RP Li, G (reprint author), Central South University, Xiangya Medical School, Cancer Research Institute, 410078 Changsha, China. EM libsun@163.com CR Ferlay J, Autier P, Boniol M et al, 1999, Estimates of the cancer incidence and mortality in Europe in 2006. Ann Oncol 18:581–592 Sung JJ, Lau JY, Goh K, Leung W, 2005, Increasing incidence of colorectal cancer in Asia: implications for screening. Lancet Oncol 6:871–876 Kievit J, 2002, Follow-up of patients with colorectal cancer: numbers needed to test and treat. Eur J Cancer 38:986–999 Cheah PY, 2009, Recent advances in colorectal cancer genetics and diagnostics. Crit Rev Oncol Hematol 69:45–55 Spano JP, Milano G, Vignot S, Khayat D, 2008, Potential predictive markers of response to EGFR-targeted therapies in colorectal cancer. Crit Rev Oncol Hematol 66:21–30 Le Tourneau C, Vidal L, Siu LL, 2008, Progress and challenges in the identification of biomarkers for EGFR and VEGFR targeting anticancer agents. Drug Resist Updat 11:99–109 Spano JP, Lagorce C, Atlan D et al, 2005, Impact of EGFR expression on colorectal cancer patient prognosis and survival. Ann Oncol 16:102–108 Flamini E, Mercatali L, Nanni O et al, 2006, Free DNA and carcinoembryonic antigen serum levels: an important combination for diagnosis of colorectal cancer. Clin Cancer Res 12:6985–6988 Eschrich S, Yang I, Bloom G et al, 2005, Molecular staging for survival prediction of colorectal cancer patients. J Clin Oncol 23:3526–3535 Sun QB, Ho JJ, Kim YS, 1986, Human colonic cancer associated antigens detected by three monoclonal antibodies. Chin Med J 99:63–74 Fu YN, Sun QB, 1988, Effect of monoclonal antibody on human colorectal cancer cells. Aizheng 7:331–334 Wang CL, Guo M, Sun QB, Lin HX, Bi QS, 1989, Targeted therapy for colorectal cancer using monoclonal antibody-ricin A chain. Bull Human Med Univ 14:107–110 Zhang J, Wang CL, Sun QB et al, 1990, Radio immunoimaging study with 131I-labeled anti-colorectal carcinoma monoclonal antibody in nude mouse model. Bull Human Med Univ 15:235–238 Hu JY, Su JZ, Pi ZM, Zhou JG, Sun QB, 1998, Radioimmunoimaging of colorectal cancer using, 99m)Tc labeled monoclonal antibody. World J Gastroenterol 4:303–306 Van Aelst L, D’Souza-Schorey C, 1997, Rho GTPases and signaling networks. Genes Dev 11:2295–2322 Ochocka AM, Grden M, Sakowicz-Burkiewicz M, Szutowicz A, Pawelczyk T, 2008, Regulation of phospholipase C-[delta]1 by ARGHAP6, a GTPase-activating protein for RhoA: possible role for enhanced activity of phospholipase C in hypertension. Intl J Biochem Cell Biol 40:2264–2273 Prakash SK, Paylor R, Lamarche-Vane N et al, 2000, Functional analysis of ARHGAP6, a novel GTPase-activating protein for RhoA. Hum Mol Genet 9:477–488 Schaefer L, Prakash S, Zoghbi HY, 1997, Cloning and characterization of a novel rho-type GTPase-activating protein gene, ARHGAP6, from the critical region for microphthalmia with linear skin defects. Genomics 46:268–277 Moon SY, Zheng Y, 2003, Rho GTPase-activating proteins in cell regulation. Trends Cell Biol 13:13–22 Burridge K, Wennerberg K, 2004, Rho and Rac take center stage. Cell 116:167–179 Durkin ME, Yuan BZ, Zhou X et al, 2007, DLC-1: a Rho GTPase-activating protein and tumour suppressor. J Cell Mol Med 11:1185–1207 Potter JD, 1999, Colorectal cancer: molecules and populations. J Natl Cancer Inst 91:916–932 Liu XF, Zhu SG, Zhang H et al, 2006, The methylation status of the TMS1/ASC gene in cholangiocarcinoma and its clinical significance. Hepatobiliary Pancreat Dis Int 5:449–453 Kim JC, Roh SA, Lee KH et al, 2005, Genetic and pathologic changes associated with lymphovascular invasion of colorectal adenocarcinoma. Clin Exp Metastasis 22:421–428 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 319 EP 326 DI 10.1007/s12253-009-9226-1 PG 8 ER PT J AU Habibollahi, P Jamshidiha, M Daryani, EN Jahanzad, I Ghahremani, HM Ostad, NS AF Habibollahi, Peiman Jamshidiha, Mostafa Daryani, E Nasser Jahanzad, Issa Ghahremani, H Mohammad Ostad, Nasser Seyed TI Correlation Between Inducible Nitric Oxide Synthase and Cyclooxygenase-2 Expression in Human Colorectal Adenocarcinoma: A Cross-Sectional Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Nitric oxide synthase; Cyclooxygenase-2; Adenocarcinoma ID Colorectal cancer; Nitric oxide synthase; Cyclooxygenase-2; Adenocarcinoma AB Cyclooxygenase-2 (COX-2) enzyme is believed to play a role in tumor angiogenesis, differentiation, and apoptosis. The inducible isoform of nitric oxide synthase (iNOS) also has the potential ability to damage DNA and conceivably contribute to tumor formation by a rise in nitric oxide production. Seventeen patients diagnosed with colorectal adenocarcinoma, who underwent surgical resection of the tumor, were enrolled in the study. Two macroscopic tissue samples, one from the tumor and the other from the tumor free surgical margin were collected from every patient as formalin fixed paraffin embedded blocks. Samples were analyzed for iNOS and COX-2 expression by immunohistochemistry and Western blotting. Results were digitized and semi-quantitatively analyzed. Immunohistochemistry revealed a similar pattern of expression for both iNOS and COX-2, as both were detected in tumor and epithelial cells. The mean iNOS and COX-2 levels determined by Western blotting method were significantly higher in tumor than in the tumor-free tissues (Wilcoxon signed-rank test, p<0.001 both for iNOS and COX-2). Patients with lymph node involvement had higher levels of both enzymes in tumors (Mann-Whitney U test, p<0.05). There was correlation between iNOS and COX-2 expression of tumor determined by immunohistochemistry and also by Western blotting (Spearman’s rho test, R=0.53, p=0.03 and R=0.57, p=0.02, respectively). In conclusion, our results point out a relationship between iNOS and COX-2 expression in human colorectal adenocarcinomas and may also suggest a possible link between advanced stages of the disease and higher expression of iNOS and COX-2. C1 [Habibollahi, Peiman] Tehran University of Medical Sciences, Faculty of Pharmacy, Department of Pharmacology & ToxicologyTehran, Iran. [Jamshidiha, Mostafa] Tehran University of Medical Sciences, Faculty of Pharmacy, Department of Pharmacology & ToxicologyTehran, Iran. [Daryani, E Nasser] Tehran University of Medical Sciences, Department of GastroenterologyTehran, Iran. [Jahanzad, Issa] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Ghahremani, H Mohammad] Tehran University of Medical Sciences, Faculty of Pharmacy, Department of Pharmacology & ToxicologyTehran, Iran. [Ostad, Nasser Seyed] Tehran University of Medical Sciences, Faculty of Pharmacy, Department of Pharmacology & ToxicologyTehran, Iran. RP Ostad, NS (reprint author), Tehran University of Medical Sciences, Faculty of Pharmacy, Department of Pharmacology & Toxicology, Tehran, Iran. EM ostadnas@tums.ac.ir CR Jass JR, Whitehall VL, Young J, Leggett BA, 2002, Emerging concepts in colorectal neoplasia. Gastroenterology 123:862–876 Landis SH, Murray T, Bolden S, Wingo PA, 1998, Cancer statistics, 1998. CA Cancer J Clin 48:6–29 Potter JD, 1995, Risk factors for colon neoplasia–epidemiology and biology. Eur J Cancer 31A:1033–1038 Rao CV, Kawamori T, Hamid R, Reddy BS, 1999, Chemoprevention of colonic aberrant crypt foci by an inducible nitric oxide synthase-selective inhibitor. Carcinogenesis 20:641–644 Salvemini D, Misko TP, Masferrer JL, Seibert K, Currie MG, Needleman P, 1993, Nitric oxide activates cyclooxygenase enzymes. Proc Natl Acad Sci U S A 90:7240–7244 Weinberg JB, 2000, Nitric oxide synthase 2 and cyclooxygenase 2 interactions in inflammation. Immunol Res 22:319–341 Rahman MA, Dhar DK, Yamaguchi E et al, 2001, Coexpression of inducible nitric oxide synthase and COX-2 in hepatocellular carcinoma and surrounding liver: possible involvement of COX-2 in the angiogenesis of hepatitis C virus-positive cases. Clin Cancer Res 7:1325–1332 Marrogi AJ, Travis WD, Welsh JA et al, 2000, Nitric oxide synthase, cyclooxygenase 2, and vascular endothelial growth factor in the angiogenesis of non-small cell lung carcinoma. Clin Cancer Res 6:4739–4744 Gallo O, Fabbroni V, Sardi I, Magnelli L, Boddi V, Franchi A, 2002, Correlation between nitric oxide and cyclooxygenase-2 pathways in head and neck squamous cell carcinomas. Biochem Biophys Res Commun 299:517–524 Bing RJ, Miyataka M, Rich KA et al, 2001, Nitric oxide, prostanoids, cyclooxygenase, and angiogenesis in colon and breast cancer. Clin Cancer Res 7:3385–3392 Cianchi F, Cortesini C, Fantappie O et al, 2004, Cyclooxygenase- 2 activation mediates the proangiogenic effect of nitric oxide in colorectal cancer. Clin Cancer Res 10:2694–2704 Ohta T, Takahashi M, Ochiai A, 2006, Increased protein expression of both inducible nitric oxide synthase and cyclooxygenase-2 in human colon cancers. Cancer Lett 239:246–253 Kobzik L, Reid MB, Bredt DS, Stamler JS, 1994, Nitric oxide in skeletal muscle. Nature 372:546–548 Palmer RM, Ferrige AG, Moncada S, 1987, Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature 327:524–526 Jaiswal M, LaRusso NF, Gores GJ, 2001, Nitric oxide in gastrointestinal epithelial cell carcinogenesis: linking inflammation to oncogenesis. Am J Physiol Gastrointest Liver Physiol 281: G626–634 Kim PK, Zamora R, Petrosko P, Billiar TR, 2001, The regulatory role of nitric oxide in apoptosis. Int Immunopharmacol 1:1421– 1441 Cobbs CS, Brenman JE, Aldape KD, Bredt DS, Israel MA, 1995, Expression of nitric oxide synthase in human central nervous system tumors. Cancer Res 55:727–730 Thomsen LL, Lawton FG, Knowles RG, Beesley JE, Riveros- Moreno V, Moncada S, 1994, Nitric oxide synthase activity in human gynecological cancer. Cancer Res 54:1352–1354 Thomsen LL, Miles DW, Happerfield L, Bobrow LG, Knowles RG, Moncada S, 1995, Nitric oxide synthase activity in human breast cancer. Br J Cancer 72:41–44 Chhatwal VJ, Ngoi SS, Chan ST, Chia YW, Moochhala SM, 1994, Aberrant expression of nitric oxide synthase in human polyps, neoplastic colonic mucosa and surrounding peritumoral normal mucosa. Carcinogenesis 15:2081–2085 Moochhala S, Chhatwal VJ, Chan ST, Ngoi SS, Chia YW, Rauff A, 1996, Nitric oxide synthase activity and expression in human colorectal cancer. Carcinogenesis 17:1171–1174 Williams CS, DuBois RN, 1996, Prostaglandin endoperoxide synthase: why two isoforms? Am J Physiol 270:G393–400 de Leval X, Delarge J, Somers F et al, 2000, Recent advances in inducible cyclooxygenase, COX-2, inhibition. Curr Med Chem 7:1041–1062 Prescott SM, Fitzpatrick FA, 2000, Cyclooxygenase-2 and carcinogenesis. Biochim Biophys Acta 1470:M69–78 Xu XM, Sansores-Garcia L, Chen XM, Matijevic-Aleksic N, Du M, Wu KK, 1999, Suppression of inducible cyclooxygenase 2 gene transcription by aspirin and sodium salicylate. Proc Natl Acad Sci U S A 96:5292–5297 Smith T, McCracken J, Shin YK, DeWitt D, 2000, Arachidonic acid and nonsteroidal anti-inflammatory drugs induce conformational changes in the human prostaglandin endoperoxide H2 synthase-2, cyclooxygenase-2). J Biol Chem 275:40407–40415 Dimberg J, Samuelsson A, Hugander A, Soderkvist P, 1999, Differential expression of cyclooxygenase 2 in human colorectal cancer. Gut 45:730–732 Fujita T, Matsui M, Takaku K et al, 1998, Size- and invasiondependent increase in cyclooxygenase 2 levels in human colorectal carcinomas. Cancer Res 58:4823–4826 Kargman SL, O'Neill GP, Vickers PJ, Evans JF, Mancini JA, Jothy S, 1995, Expression of prostaglandin G/H synthase-1 and -2 protein in human colon cancer. Cancer Res 55:2556–2559 Sheehan KM, Sheahan K, O'Donoghue DP et al, 1999, The relationship between cyclooxygenase-2 expression and colorectal cancer. JAMA 282:1254–1257 Zhang H, Sun XF, 2002, Overexpression of cyclooxygenase-2 correlates with advanced stages of colorectal cancer. Am J Gastroenterol 97:1037–1041 Greene FL, American Joint Committee on Cancer, American Cancer Society, 2002, AJCC cancer staging manual. In. New York: Springer-Verlag: 114 Shi SR, Liu C, Balgley BM, Lee C, Taylor CR, 2006, Protein extraction from formalin-fixed, paraffin-embedded tissue sections: quality evaluation by mass spectrometry. J Histochem Cytochem 54:739–743 Gallo O, Schiavone N, Papucci L et al, 2003, Down-regulation of nitric oxide synthase-2 and cyclooxygenase-2 pathways by p53 in squamous cell carcinoma. Am J Pathol 163:723–732 Liu Q, Chan ST, Mahendran R, 2003, Nitric oxide induces cyclooxygenase expression and inhibits cell growth in colon cancer cell lines. Carcinogenesis 24:637–642 Timoshenko AV, Lala PK, Chakraborty C, 2004, PGE2-mediated upregulation of iNOS in murine breast cancer cells through the activation of EP4 receptors. Int J Cancer 108:384–389 Hong SK, Gul YA, Ithnin H, Talib A, Seow HF, 2004, Expression of beta-catenin, COX-2 and iNOS in colorectal cancer: relevance of COX-2 adn iNOS inhibitors for treatment in Malaysia. Asian J Surg 27:10–17 Cianchi F, Cuzzocrea S, Vinci MC, et al., 2009, Heterogeneous expression of cyclooxygenase-2 and inducible nitric oxide synthase within colorectal tumors: Correlation with tumor angiogenesis. Dig Liver Dis. Kojima M, Morisaki T, Tsukahara Y et al, 1999, Nitric oxide synthase expression and nitric oxide production in human colon carcinoma tissue. J Surg Oncol 70:222–229 Yagihashi N, Kasajima H, Sugai S et al, 2000, Increased in situ expression of nitric oxide synthase in human colorectal cancer. Virchows Arch 436:109–114 Takahashi M, Mutoh M, Kawamori T, Sugimura T, Wakabayashi K, 2000, Altered expression of beta-catenin, inducible nitric oxide synthase and cyclooxygenase-2 in azoxymethane-induced rat colon carcinogenesis. Carcinogenesis 21:1319–1327 Cianchi F, Cortesini C, Fantappie O et al, 2003, Inducible nitric oxide synthase expression in human colorectal cancer: correlation with tumor angiogenesis. Am J Pathol 162:793–801 Lagares-Garcia JA, Moore RA, Collier B, Heggere M, Diaz F, Qian F, 2001, Nitric oxide synthase as a marker in colorectal carcinoma. Am Surg 67:709–713 Nozoe T, Yasuda M, Honda M, Inutsuka S, Korenaga D, 2002, Immunohistochemical expression of cytokine induced nitric oxide synthase in colorectal carcinoma. Oncol Rep 9:521–524 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 327 EP 335 DI 10.1007/s12253-009-9239-9 PG 9 ER PT J AU Gisterek, I Matkowski, R Lacko, A Sedlaczek, P Szewczyk, K Biecek, P Halon, A Staszek, U Szelachowska, J Pudelko, M Bebenek, M Harlozinska-Szmyrka, A Kornafel, J AF Gisterek, Iwona Matkowski, Rafal Lacko, Aleksandra Sedlaczek, Pawel Szewczyk, Krzysztof Biecek, Przemyslaw Halon, Agnieszka Staszek, Urszula Szelachowska, Jolanta Pudelko, Marek Bebenek, Marek Harlozinska-Szmyrka, Antonina Kornafel, Jan TI Serum Vascular Endothelial Growth Factors A, C and D in Human Breast Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Vascular endothelial growth factor-A; Vascular endothelial growth factor-C; Vascular endothelial growth factor-D; Serum assay; Breast cancer ID Vascular endothelial growth factor-A; Vascular endothelial growth factor-C; Vascular endothelial growth factor-D; Serum assay; Breast cancer AB Available evidence suggests that vascular endothelial growth factor (VEGF) a potent regulator of vasculogenesis and tumor angiogenesis may be a predictor of recurrence in breast cancer patients. We sought to determine whether VEGF serum levels (VEGF-A, VEGF-C and VEGF-D) in 377 patients with malignant and benign breast tumors differ and whether there is association between vascular growth factors, clinicopathologic features and prognosis. There was no significant difference in investigated circulating angiogenic markers between patients with malignant and non malignant lesions. We found strong correlation between VEGF-A and VEGF-D and between VEGF- C and VEGF-D. Besides serum VEGF-D levels and estrogen receptor (ER) expressions no other correlations between VEGF and clinicopathologic variables were observed. However, elevated VEGF-A and VEGF-C concentrations were associated with increased number of erythrocytes, leukocytes and platelets. In Cox model values of angiogenic serum markers and recognized prognostic markers in breast cancer, VEGF-C turned out as independent prognostic factor. Our study is the first analysis showing correlation between serum concentrations of three angiogenic factors: VEGF-A, VEGF-C, VEGF-D. Associations between angiogenic cytokines and number of blood cells may be due to release of VEGF from platelets and leucocytes. Prognostic role of VEGF is still uncertain, though VEGF-C has a potential to serve as a prognostic marker. C1 [Gisterek, Iwona] Wroclaw Medical University, Department of Oncology, pl. Hirszfelda 12, 53-413 Wroclaw, Poland. [Matkowski, Rafal] Wroclaw Medical University, Department of Oncology, pl. Hirszfelda 12, 53-413 Wroclaw, Poland. [Lacko, Aleksandra] Wroclaw Medical University, Department of Oncology, pl. Hirszfelda 12, 53-413 Wroclaw, Poland. [Sedlaczek, Pawel] Wroclaw Medical University, Department of Clinical ImmunologyWroclaw, Poland. [Szewczyk, Krzysztof] Wroclaw Medical University, Department of Oncology, pl. Hirszfelda 12, 53-413 Wroclaw, Poland. [Biecek, Przemyslaw] University of Warsaw, Faculty of Mathematics, Informatics, and MechanicsWarsaw, Poland. [Halon, Agnieszka] Wroclaw Medical University, Department of PathologyWroclaw, Poland. [Staszek, Urszula] Wroclaw Medical University, Department of Oncology, pl. Hirszfelda 12, 53-413 Wroclaw, Poland. [Szelachowska, Jolanta] Wroclaw Medical University, Department of Oncology, pl. Hirszfelda 12, 53-413 Wroclaw, Poland. [Pudelko, Marek] Lower Silesian Oncology Center, Department of Oncological SurgeryWroclaw, Poland. [Bebenek, Marek] Lower Silesian Oncology Center, Department of Oncological SurgeryWroclaw, Poland. [Harlozinska-Szmyrka, Antonina] Wroclaw Medical University, Department of Clinical ImmunologyWroclaw, Poland. [Kornafel, Jan] Wroclaw Medical University, Department of Oncology, pl. Hirszfelda 12, 53-413 Wroclaw, Poland. RP Matkowski, R (reprint author), Wroclaw Medical University, Department of Oncology, 53-413 Wroclaw, Poland. EM matkowski.r@dco.com.pl CR Kinoshita J, Kitamura K, Kabashima A, Saeki H, Tanaka S, Sugimachi K, 2001, Clinical significance of vascular endothelial growth factor-C, VEGF-C, in breast cancer. Breast Cancer Res Treat 66:159–164 Heer K, Kumar H, Read J, Fox J, Monson J, Kerin M, 2001, Serum vascular endothelial growth factor in breast cancer: Its relation with cancer type and estrogen receptor status. Clin Cancer Res 7:3491–3494 Mitsuhashi A, Suzuka K, Yamazawa K, Matsui H, Seki K, Sekiya S, 2005, Serum vascular endothelial growth factor, VEGF, and VEGF-C levels as a tumor markers in patients with cervical carcinoma. Cancer 103:724–730 Homer J, Anyanwu K, Ell S, Greenman J, Stafford N, 1999, Serum vascular endothelial growth factor in patients with head and neck squamous cell carcinoma. Clin Otolaryngol 24:426–430 Hefler L, Zeillinger R, Grimm C, Sood A, Cheng W, Gadducci A, Tempfer CB, Reinthaller A, 2006, Preoperative serum vascular endothelial growth factor as a prognostic parameter in ovarian cancer. Gynecol Oncol 103:512–517 Gonzalez F, Quesada A, Sevilla I, Baca F, Medina M, Amores J, Diaz JM, Rius-Diaz F, Marques E, Alba E, 2007, Prognostic value of serum angiogenic activity in colorectal cancer patients. J Cell Mol Med 11:120–128 De Vita F, Orditura M, Lieto E, Infusino S, Morgillo F, Martinelli E, Castellano P, Romano C, Ciardiello F, Catalano G, Pignatelli C, Galizia G, 2004, Elevated perioperative serum vascular endothelial growth factor levels in patients with colon carcinoma. Cancer 100:270–278 Wang T, Deng M, Qiu W, Dong W, 2007, Association of serum vascular endothelial growth factor –C and lymphatic vessel density with lymph node metastasis and prognosis of patients with gastric cancer. World J Gastroenterol 13:1794–1798 Tamura M, Ohta Y, 2003, Serum vascular endothelial growth factor –C level in patients with primary nonsmall cell lung carcinoma. Cancer 98:1217–1222 Bando H, Weich H, Brokelmann M, Horiguchi S, Funata N, Ogawa T, Toi M, 2005, Association between intratumoral free and total VEGF, soluble VEGFR-1, VEGFR-2 and prognosis in breast cancer. Br J Cancer 92:553–561 Foekens J, Peters H, Grabenchtchikov N, Look M, Meijer-van Gelder M, Geurts-Moespot A, van der Kwast TH, Sweep CG, Klijn JG, 2001, High tumor levels of vascular endothelial growth factor predict poor response to systemic therapy in breast cancer. Cancer Res 61:5407–541 Granato A, Frassineti G, Giovannini N, Ballardini M, Nanni O, Maltoni R, Amadori D, Volpi A, 2006, Do serum angiogenic growth factors provide additional information to that of conventional markers in monitoring the course of metastatic breast cancer? Tumor Biol 27:302–308 R development Core Team: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. ISBN 3-900051-07-0, 2008, URL http://www.R-project.org Konukoglu D, Turhan M, Celik V, Turna H, 2007, Relation of serum vascular endothelial growth factor as an angiogenesis biomarker with nitric oxide & urokinase-type plasminogen activator in breast cancer patients. Indian J Med Res 125:747–751 Adams J, Carder P, Downey S, Forbes M, MacLennan K, Allgar V, Kaufman S, Hallam S, Bicknell R, Walker JJ, Cairnduff F, Selby PJ, Perren TJ, Lansdown M, Banks RE, 2000, Vascular endothelial growth factor, VEGF, in breast cancer: comparison of plasma, serum, and tissue VEGF and microvessel density and effects of tamoxifen. Cancer Res 60:2898–2905 Mathur S, Mathur R, Gray E, Lane D, Underwood P, Kohler M, Creasman WT, 2005, Serum vascular endothelial growth factor C, VEGF-C, as a specific biomarker for advanced cervical cancer: relationship to insulin-like growth factor II, IGF-II), IGF binding protein 3, IGF-BP3, and VEGF-B. Gynecol Oncol 98:467–483 Tamura M, Oda M, Matsumoto I, Tsunezuka Y, Kawakami K, Ohta Y, Watanabe G, 2004, The combination assai with circulating vascular endothelial growth factor, VEGF)-C, Matrix Metalloproteinase-9, and VEGF for diagnosing lymph node metastasis in patients with non-small cell lung cancer. Ann Surg Oncol 11:928–933 Linderholm B, Tavelin B, Grankvist K, Henriksson R, 1998, Vascular endothelial growth factor is of high prognostic value in node-negative breast carcinoma. J Clin Oncol 16:3121–3128 Ryden L, Stendahl M, Jonsson H, Emdin S, Bengtsson N, Landberg G, 2005, Tumor specific VEGF-A and VEGFR2 in postmenopausal breast cancer patients with long term folllow up: implication of link between VEGF pathway and tamoxifen response. Breast Cancer Res Treat 10:135–143 Ryden L, Jirstrom K, Bendahl P, Ferno M, Nordenskjold B, Stal O, Thorstenson S, Jonsson PE, Landberg G, 2005, Tumor specific expression of vascular endothelial growth factor receptor 2 but not vascular endothelial growth factor or human epidermal growth factor receptor 2 is associated with impaired response to adjuvant tamoxifen in premenopausal breast cancer. J Clin Oncol 23:4695– 4704 Svensson S, Jirstrom K, Ryden L, Roos G, Emdin S, Ostrowski M, Landberg G, 2005, ERK phosphorylation is linked to VEGFR2 expression and Ets-2 pphosphorylation in breast cancer and is assocoated with tamoxifen resistance and small tumors with good prognosis. Oncogene 24:4370–4379 Werther K, Christensen I, Nielsen H, 2002, Determination of vascular endothelial growth factor, VEGF, in circulating blood: significance of VEGF in various leucocytes and platelets. Scand J Clin Lab Invest 62:343–350 Kut C, Gabhann F, Popel A, 2007, Where is VEGF in the body? A meta-analysis of VEGF distribution in cancer. Br J Cancer 97:978–985 Kusumanto Y, Dam W, Hospers G, Meijer C, Mulder N, 2003, Platelets and granulocytes, in particular the neutrophils, form important compartments for circulating vascular endothelial growth factor. Angiogenesis 6:283–287 Yang W, Klos K, Yang Y, Smith T, Shi D, Yu D, 2002, ErbB2 overexpression correlates with increased expression of vascular endothelial growth factors A, C, and D in human breast carcinoma. Cancer 94:2855–2861 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 337 EP 344 DI 10.1007/s12253-009-9211-8 PG 8 ER PT J AU Watari, H Kanuma, T Ohta, Y Hassan, KM Mitamura, T Hosaka, M Minegishi, T Sakuragi, N AF Watari, Hidemichi Kanuma, Tatsuya Ohta, Yoko Hassan, Kamel Mohamed Mitamura, Takashi Hosaka, Masayoshi Minegishi, Takashi Sakuragi, Noriaki TI Clusterin Expression Inversely Correlates with Chemosensitivity and Predicts Poor Survival in Patients with Locally Advanced Cervical Cancer Treated with Cisplatin-Based Neoadjuvant Chemotherapy and Radical Hysterectomy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Clusterin; Neoadjuvant chemotherapy; Cervical cancer; Lymphnode metastasis; Immunohistochemistry ID Clusterin; Neoadjuvant chemotherapy; Cervical cancer; Lymphnode metastasis; Immunohistochemistry AB Overexpression of clusterin, an antiapoptotic molecule, has been reported to induce resistance to chemotherapy in a variety of cancer cell types. The aim of this study was to evaluate the significance of clusterin expression to predict response to platinum-based neoadjuvant chemotherapy and survival of patients with invasive cervical cancer who subsequently underwent radical hysterectomy. Biopsy specimens of invasive cervical cancer before neoadjuvant chemotherapy were obtained from 46 patients who subsequently underwent radical hysterectomy at Hokkaido University Hospital and Gunma University Hospital from 1994 to 2007. The expression of clusterin protein was analyzed by immunohistochemistry. Findings were evaluated in relation to several clinicopathological factors. Survival analyses were performed by the Kaplan-Meier curves and the log-rank test. Independent prognostic factors were determined by multivariate Cox regression analysis. Clusterin protein was mainly present in the cytoplasm of cervical cancer cells. The expression of clusterin protein in cervical cancer tissues before neoadjuvant chemotherapy was significantly related to poor response to chemotherapy among factors analyzed. Univariate analysis on prognostic factors showed that response to chemotherapy (p=0.01), lymph node metastasis (p=0.02), and clusterin expression (p=0.02) were related to survival. Multivariate analysis revealed that lymph node metastasis (p=0.03), and clusterin expression (p=0.03) were independent prognostic factors for survival of cervical cancer patients. We conclude that clusterin expression could be a new molecular marker to predict response to platinumbased chemotherapy and survival of patients with cervical cancer treated with neoadjuvant chemotherapy and radical hysterectomy. C1 [Watari, Hidemichi] Hokkaido University, Graduate School of Medicine, Department of Obstetrics and Gynecology, North 15, West 7, Kita-Ku, 060-8638 Sapporo, Japan. [Kanuma, Tatsuya] Gunma University, Graduate School of Medicine, Department of Obstetrics and Gynecology, 371-8511 Maebashi, Japan. [Ohta, Yoko] Hokkaido University, Graduate School of Medicine, Department of Obstetrics and Gynecology, North 15, West 7, Kita-Ku, 060-8638 Sapporo, Japan. [Hassan, Kamel Mohamed] Hokkaido University, Graduate School of Medicine, Department of Obstetrics and Gynecology, North 15, West 7, Kita-Ku, 060-8638 Sapporo, Japan. [Mitamura, Takashi] Hokkaido University, Graduate School of Medicine, Department of Obstetrics and Gynecology, North 15, West 7, Kita-Ku, 060-8638 Sapporo, Japan. [Hosaka, Masayoshi] Hokkaido University, Graduate School of Medicine, Department of Obstetrics and Gynecology, North 15, West 7, Kita-Ku, 060-8638 Sapporo, Japan. [Minegishi, Takashi] Gunma University, Graduate School of Medicine, Department of Obstetrics and Gynecology, 371-8511 Maebashi, Japan. [Sakuragi, Noriaki] Hokkaido University, Graduate School of Medicine, Department of Obstetrics and Gynecology, North 15, West 7, Kita-Ku, 060-8638 Sapporo, Japan. RP Watari, H (reprint author), Hokkaido University, Graduate School of Medicine, Department of Obstetrics and Gynecology, 060-8638 Sapporo, Japan. EM watarih@med.hokudai.ac.jp CR Panici PB, Greggi S, Scambia G et al, 1998, Long-term survival following neoadjuvant chemotherapy and radical surgery in locally advanced cervical cancer. Eur J Cancer 34:341–346 Redondo M, Villar E, Torres-Munoz J et al, 2002, Overexpression of clusterin in human breast carcinoma. Am J Pathol 157:393–399 Pucci S, Bonanno E, Pichiorri F et al, 2004, Modulation of different clusterin isoforms in human colon tumorigenesis. Oncogene 23:2298–2304 Chung J, Kwak C, Jin RJ et al, 2004, Enhanced chemosensitivity of bladder cancer cells to cisplatin by suppression of clusterin in vitro. Cancer Lett 203:155–161 Zellweger T, Chi K, Miyake H et al, 2002, Enhanced radiosensitivity in prostate cancer by inhibition of the cell survival protein clusterin. Clin Cancer Res 8:3276–3284 Miyake H, Hara S, Arakawa S et al, 2002, Overexpression of clusterin is an independent prognostic factor for nonpapillary renal cell carcinoma. J Urol 167:703–706 Kruger S, Ola V, Fisher D et al, 2007, Prognostic significance of clusterin immunoreactivity in breast cancer. Neoplasma 54:46– 50 Watari H, Ohta Y, Hassan MK et al, 2008, Clusterin expression predicts survival of invasive cervical cancer patients treated with radical hysterectomy and systematic lymphadenectomy. Gynecol Oncol 108:527–532 Konishi I, Nanbu K, Mandai M et al, 1998, Tumor response to neoadjuvant chemotherapy correlates with the expression of Pglycoprotein and PCNA but not GST-pi in the tumor cells of cervical carcinoma. Gynecol Oncol 70:365–371 Faried LS, Faried A, Kanuma T et al, 2006, Predictive and prognostic role of activated mammalian target of rapamycin in cervical cancer treated with cisplatin-based neoadjuvant chemotherapy. Oncol Rep 16:57–63 Saito T, Takehara M, Tanaka R et al, 2004, Correlation between responsiveness of neoadjuvant chemotherapy and apoptosisassociated proteins for cervical adenocarcinoma. Gynecol Oncol 92:284–292 Costa S, Terzano P, Bovicelli A et al, 2004, CD44 isoform 6, CD44v6, is a prognostic indicator of the response to neoadjuvant chemotherapy in cervical carcinoma. Gynecol Oncol 80:67–73 Ferrandina G, Lauriola L, Distefano MG et al, 2002, Increased cyclooxygenase-2 expression is associated with chemotherapy resistance and poor survival in cervical cancer patients. J Clin Oncol 20:973–981 Chung HH, Kim MK, Kim JW et al, 2006, XRCC1 R399Q polymorphism is associated with response to platinum-based neoadjuvant chemotherapy in bulhy cervical cancer. Gynecol Oncol 103:1031–1037 Sowery RD, Hadaschik BA, So AI et al, 2008, Clusterin knockdown using the antisense oligonucleotide OGX-011 resensitizes docetaxel-refractory prostate cancer PC-3 cells to chemotherapy. BJU Int 102:389–397 Miyake H, Eto H, Hara I et al, 2004, Synergistic antitumor activity by combined treatment with gemcitabine and antisense oligodeoxynucleotide targeting clusterin gene in an intravesical administration model against human bladder cancer kotcc-1 cells. J Urol 171:2477–2481 Mizutani K, Matsumoto K, Hasegawa N et al, 2006, Expression of clusterin, XIAP and survivin, and their changes by camptothecin, CPT, treatment in CPT-resistant PC-3 and CPT-sensitive LNCaP cells. Exp Oncol 28:209–251 Lourda M, Trougakos P, Gonos ES, 2006, Development of resistance to chemotherapeutic drugs in human osteosarcoma cell lines largely depends on up-regulation of Clusterin/Apolipoprotein J. Int J Cancer 120:611–622 Park DC, Yeo SG, Shin EY et al, 2006, Custerin confers paclitaxel resistance in cervical cancer. Gynecol Oncol 103:996– 1000 Lahousen M, Haas J, Pockel H et al, 1999, Chemotherapy versus radiotherapy versus observation for high-risk cervical carcinoma after radical hysterectomy: a randomized, prospective multicenter trial. Gynecol Oncol 73:196–201 Saad F, Hotte SJ, North S, et al, 2008, A phase II randomized study evaluating custirsen, OGX-011, in patients with hormone refractory prostate cancer, HRPC, who relapsed on or within 6 months of first-line docetaxel therapy. ASCO Genitourinary cancers symposium; abstract#151 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 345 EP 352 DI 10.1007/s12253-009-9235-0 PG 8 ER PT J AU Qin, F Song, Y Li, Z Zhao, L Zhang, Y Geng, L AF Qin, Fengjin Song, Yao Li, Zijian Zhao, Ling Zhang, Youyi Geng, Li TI S100A8/A9 Induces Apoptosis and Inhibits Metastasis of CasKi Human Cervical Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Apoptosis; Cervical cancer; Metastasis; MMP-2; S100 proteins ID Apoptosis; Cervical cancer; Metastasis; MMP-2; S100 proteins AB S100 proteins, a family of Ca2+-binding proteins, have been linked to several human diseases in recent years. Deregulated expression of S100 proteins, including S100A9 and its partner S100A8, was reported to be associated with neoplastic disorders. In our previous study using serial analysis of gene expression, we identified decreased expressions of S100A9 and S100A8 in human cervical squamous cell carcinoma. To investigate the functions of S100A8 and S100A9 in cervical cancer, we purified recombinant S100A8 and S100A9 proteins and treated CaSki human cervical cancer cells with these proteins. We found that S100A8/A9 induced apoptosis and inhibited migration of CaSki cells; S100A8/A9 also reduced the expression of matrix metalloproteinase (MMP)-2 in CaSki cells. In summary, this study suggests that S100A8 and S100A9 have inhibitory effects on the proliferation of CaSki carcinoma cells by inducing cell apoptosis and on the invasiveness of CaSki cells. C1 [Qin, Fengjin] Peking University Third Hospital, Department of Obstetrics and Gynecology, 100191 Beijing, China. [Song, Yao] Ministry of Education, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Science, Institute of Vascular Medicine, 100191 Beijing, China. [Li, Zijian] Ministry of Education, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Science, Institute of Vascular Medicine, 100191 Beijing, China. [Zhao, Ling] Peking University Third Hospital, Department of Obstetrics and Gynecology, 100191 Beijing, China. [Zhang, Youyi] Ministry of Education, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Science, Institute of Vascular Medicine, 100191 Beijing, China. [Geng, Li] Peking University Third Hospital, Department of Obstetrics and Gynecology, 100191 Beijing, China. RP Geng, L (reprint author), Peking University Third Hospital, Department of Obstetrics and Gynecology, 100191 Beijing, China. EM gengli57@163.com CR Parkin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55(2):74–108 Walboomers JM, Jacobs MV, Manos MM et al, 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189(1):12–19 Hinchliffe SA, van Velzen D, Korporaal H et al, 1995, Transience of cervical HPV infection in sexually active, young women with normal cervicovaginal cytology. Br J Cancer 72(4):943–945 Martin CM, Astbury K, O’Leary JJ, 2006, Molecular profiling of cervical neoplasia. Expert Rev Mol Diagn 6(2):217–229 Polyak K, Riggins GJ, 2001, Gene discovery using the serial analysis of gene expression technique: implications for cancer research. J Clin Oncol 19(11):2948–2958 Kang HW, Geng L, Zhao L et al, 2004, Gene expression profile in human normal cervical squamous epithelium. Chin J Reprod Health 15(5):303–305 Zimmer DB, Cornwall EH, Landar A et al, 1995, The S100 protein family: history, function, and expression. Brain Res Bull 37(4):417–429 Donato R, 2003, Intracellular and extracellular roles of S100 proteins. Microsc Res Tech 60(6):540–551 Heizmann CW, Fritz G, Schafer BW, 2002, S100 proteins: structure, functions and pathology. Front Biosci 7:d1356–d1368 Yui S, Nakatani Y, Mikami M, 2003, Calprotectin, S100A8/ S100A9), an inflammatory protein complex from neutrophils with a broad apoptosis-inducing activity. Biol Pharm Bull 26(6):753– 760 Wang J, Cai Y, Hao Xu et al, 2004, Expression of MRP14 gene is frequently down-regulated in Chinese human esophageal cancer. Cell Res 14(1):46–53 Roesch Ely M, Nees M, Karsai S et al, 2005, Transcript and proteome analysis reveals reduced expression of calgranulins in head and neck squamous cell carcinoma. Eur J Cell Biol 84(2–3): 431–444 Arai K, Teratani T, Kuruto-Niwa R et al, 2004, S100A9 expression in invasive ductal carcinoma of the breast: S100A9 expression in adenocarcinoma is closely associated with poor tumour differentiation. Eur J Cancer 40(8):1179–1187 Arai K, Teratani T, Nozawa R et al, 2001, Immunohistochemical investigation of S100A9 expression in pulmonary adenocarcinoma: S100A9 expression is associated with tumor differentiation. Oncol Rep 8(3):591–596 Ryckman C, Vandal K, Rouleau P et al, 2003, Proinflammatory activities of S100: proteins S100A8, S100A9, and S100A8/A9 induce neutrophil chemotaxis and adhesion. J Immunol 170, 6):3233–3242 Cohen GM, 1997, Caspases: the executioners of apoptosis. Biochem J 326(Pt 1):1–16 Haemmerlin G, Strauli P, 1981, In vitro motility of cells from human epidermoid carcinoma: a study by phase-contrast and reflection-contrast cinematography. Int J Cancer 27(5):603–610 Cramer LP, Mitchison TJ, 1995, Myosin is involved in postmitotic cell spreading. J Cell Biol 131(1):179–189 John A, Tuszynski G, 2001, The role of matrix metalloproteinases in tumor angiogenesis and tumor metastasis. Pathol Oncol Res 7, 1):14–23 Mitra A, Chakrabarti J, Chattopadhyay N et al, 2003, Membraneassociated MMP-2 in human cervical cancer. J Environ Pathol Toxicol Oncol 22(2):93–100 Juuti A, Lundin J, Nordling S et al, 2006, Epithelial MMP-2 expression correlates with worse prognosis in pancreatic cancer. Oncology 71(1-2):61–68 Shim KN, Jung SA, Joo YH et al, 2007, Clinical significance of tissue levels of matrix metalloproteinases and tissue inhibitors of metalloproteinases in gastric cancer. J Gastroenterol 42(2):120– 128 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 353 EP 360 DI 10.1007/s12253-009-9225-2 PG 8 ER PT J AU Al-Attar, A Shehata, M Durrant, L Moseley, P Deen, S Chan, S AF Al-Attar, Ahmad Shehata, Mohamed Durrant, Lindy Moseley, Paul Deen, Suha Chan, Stephen TI T Cell Density and Location Can Influence the Prognosis of Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intra-tumoural T cells; Ovarian cancer; Prognosis; Stromal T cells ID Intra-tumoural T cells; Ovarian cancer; Prognosis; Stromal T cells AB The aims of this study were to examine the significance of CD3+ cells in patients with epithelial ovarian cancer and to determine their influence on the disease in relation to their location within tumours. A 157-core tissue-microarray constructed from primary ovarian cancer patients treated at Nottingham-University-Hospitals (2000–2007) was stained for the T-cell marker CD3. The number of CD3+ cells in direct contact with tumour cells was counted per tumour area. These were considered as "intra-tumoural T-cells (ITTC)". Cores were divided into CD3 'high' or 'low' density tumours. "Stromal T-cells (STC)" were assigned as 'positive' or 'negative'. The study population had a median follow-up time of 36-months (0– 75). The number of ITTC counted in tumour cores ranged between 0 and 184/mm2. 90-tumours-(57%) were found to be in the "low-density" rubric, while 56-(36%) were of a "high-density" T-cell population. STC were found in 118-cores-(75%)-compared to 22-cores-(14%)-negative cores. Higher number of ITTC correlated with lower-grade-(p=0.045), tumour-type-(p=0.034), and longer-median-survivaltimes (57-versus 37-months for high-and low-ITTC densities, respectively, p=0.038). This relationship was reversed when tumours were infiltrated by CD3+ cells in the stroma, predicting worse-survival (Log-rank-test, p=0.028). Combining ITTC with STC produced an interesting pattern where the ITTC-low/STC + ve had the worst prognosis (p=0.003). Infiltration of ovarian cancer by T-cells can influence its prognosis depending on the location of these cells (intratumoural-versus-stromal). The former predicts improved survival, while the latter is probably contributing to tumour progression and, in turn, worse survival. C1 [Al-Attar, Ahmad] Nottingham University, Department of Clinical Oncology, Hospitals, NHS Trust–City Campus, Hucknall Road, NG5 1PB Nottingham, UK. [Shehata, Mohamed] Nottingham University, Department of Clinical Oncology, Hospitals, NHS Trust–City Campus, Hucknall Road, NG5 1PB Nottingham, UK. [Durrant, Lindy] University of Nottingham, Department of Academic Oncology, Hucknall Road, NG5 1PB Nottingham, UK. [Moseley, Paul] Nottingham University, Department of Clinical Oncology, Hospitals, NHS Trust–City Campus, Hucknall Road, NG5 1PB Nottingham, UK. [Deen, Suha] Nottingham University Hospitals, Department of Histopathology, NHS Trust–QMC Campus, Derby Road, NG7 2UH Nottingham, UK. [Chan, Stephen] Nottingham University, Department of Clinical Oncology, Hospitals, NHS Trust–City Campus, Hucknall Road, NG5 1PB Nottingham, UK. RP Chan, S (reprint author), Nottingham University, Department of Clinical Oncology, NG5 1PB Nottingham, UK. EM steve.chan@nuh.nhs.uk CR CRUK, 2009, UK Ovarian Cancer statistics [cited 2009 20/05/ 2009]; Available from: Hussein MR, Hassan HI, 2006, Analysis of the mononuclear inflammatory cell infiltrate in the normal breast, benign proliferative breast disease, in situ and infiltrating ductal breast carcinomas: preliminary observations. J Clin Pathol 59(9):972–977 Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F et al, 2005, Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA 102, 51):18538–18543 Shah CA, Allison KH, Garcia RL, Gray HJ, Goff BA, Swisher EM, 2008, Intratumoral T cells, tumor-associated macrophages, and regulatory T cells: association with p53 mutations, circulating tumor DNA and survival in women with ovarian cancer. Gynecol Oncol 109(2):215–219 Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA,Massobrio M, Regnani G et al, 2003, Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 348(3):203–213 Echchakir H, Vergnon I, Dorothee G, Grunenwald D, Chouaib S, Mami-Chouaib F, 2000, Evidence for in situ expansion of diverse antitumor-specific cytotoxic T lymphocyte clones in a human large cell carcinoma of the lung. Int Immunol 12(4):537–546 Halapi E, Yamamoto Y, Juhlin C, Jeddi-Tehrani M, Grunewald J, Andersson R et al, 1993, Restricted T cell receptor V-beta and J-beta usage in T cells from interleukin-2-cultured lymphocytes of ovarian and renal carcinomas. Cancer Immunol Immunother 36, 3):191–197 Balkwill F, 2009, Tumour necrosis factor and cancer. Nat Rev 9, 5):361–371 Hagemann T, Balkwill F, Lawrence T, 2007, Inflammation and cancer: a double-edged sword. Canc Cell 12(4):300–301 Kulbe H, Thompson R, Wilson JL, Robinson S, Hagemann T, Fatah R et al, 2007, The inflammatory cytokine tumor necrosis factor-alpha generates an autocrine tumor-promoting network in epithelial ovarian cancer cells. Cancer Res 67(2):585–592 Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD, 2002, Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 3(11):991–998 Dougan M, Dranoff G, 2009, The immune response to tumors. Current protocols in immunology. In: Coligan JE et al, eds, Chapter 20:Unit 20 11. Stewart TJ, Abrams SI, 2008, How tumours escape mass destruction. Oncogene. 27(45):5894–5903 Halpern AC, Schuchter LM, 1997, Prognostic models in melanoma. Semin Oncol 24(1 Suppl 4):S2–S7 Marrogi AJ, Munshi A, Merogi AJ, Ohadike Y, El-Habashi A, Marrogi OL et al, 1997, Study of tumor infiltrating lymphocytes and transforming growth factor-beta as prognostic factors in breast carcinoma. Int J Cancer 74(5):492–501 Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H et al, 1998, CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res 58, 16):3491–3494 Nakano O, Sato M, Naito Y, Suzuki K, Orikasa S, Aizawa M et al, 2001, Proliferative activity of intratumoral CD8(+, T-lymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity. Cancer Res 61, 13):5132–5136 Vesalainen S, Lipponen P, Talja M, Syrjanen K, 1994, Histological grade, perineural infiltration, tumour-infiltrating lymphocytes and apoptosis as determinants of long-term prognosis in prostatic adenocarcinoma. Eur J Cancer 30A(12):1797–1803 Raspollini MR, Castiglione F, Rossi Degl'innocenti D, Amunni G, Villanucci A, Garbini F et al, 2005, Tumour-infiltrating gamma/ delta T-lymphocytes are correlated with a brief disease-free interval in advanced ovarian serous carcinoma. Ann Oncol 16, 4):590–596 Dadmarz RD, Ordoubadi A, Mixon A, Thompson CO, Barracchini KC, Hijazi YM et al, 1996, Tumor-infiltrating lymphocytes from human ovarian cancer patients recognize autologous tumor in an MHC class II-restricted fashion. Canc J Sci Am 2(5):263–272 Kooi S, Zhang HZ, Patenia R, Edwards CL, Platsoucas CD, Freedman RS, 1996, HLA class I expression on human ovarian carcinoma cells correlates with T-cell infiltration in vivo and T-cell expansion in vitro in low concentrations of recombinant interleukin-2. Cell Immunol 174(2):116–128 Loercher AE, Nash MA, Kavanagh JJ, Platsoucas CD, Freedman RS, 1999, Identification of an IL-10-producing HLA-DR-negative monocyte subset in the malignant ascites of patients with ovarian carcinoma that inhibits cytokine protein expression and proliferation of autologous T cells. J Immunol. 163(11):6251–6260 Nakashima M, Sonoda K, Watanabe T, 1999, Inhibition of cell growth and induction of apoptotic cell death by the human tumorassociated antigen RCAS1. Nat Med 5(8):938–942 Rabinowich H, Reichert TE, Kashii Y, Gastman BR, Bell MC, Whiteside TL, 1998, Lymphocyte apoptosis induced by Fas ligand- expressing ovarian carcinoma cells. Implications for altered expression of T cell receptor in tumor-associated lymphocytes. J Clin Invest 101(11):2579–2588 Santin AD, Bellone S, Ravaggi A, Pecorelli S, Cannon MJ, Parham GP, 2000, Induction of ovarian tumor-specific CD8+ cytotoxic T lymphocytes by acid-eluted peptide-pulsed autologous dendritic cells. Obstet Gynecol 96(3):422–430 Woo EY, Chu CS, Goletz TJ, Schlienger K, Yeh H, Coukos G et al, 2001, Regulatory CD4(+)CD25(+, T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Res 61(12):4766–4772 Mantovani A, Allavena P, Sica A, Balkwill F, 2008, Cancerrelated inflammation. Nature 454(7203):436–444 Nelson BH, 2008, The impact of T-cell immunity on ovarian cancer outcomes. Immunol Rev 222:101–116 Hayashi K, Yonamine K, Masuko-Hongo K, Iida T, Yamamoto K, Nishioka K et al, 1999, Clonal expansion of T cells that are specific for autologous ovarian tumor among tumor-infiltrating T cells in humans. Gynecol Oncol 74(1):86–92 Negus RP, Stamp GW, Hadley J, Balkwill FR, 1997, Quantitative assessment of the leukocyte infiltrate in ovarian cancer and its relationship to the expression of C-C chemokines. Am J Pathol 150(5):1723–1734 Santin AD, Hermonat PL, Ravaggi A, Bellone S, Roman JJ, Smith CV et al, 2001, Phenotypic and functional analysis of tumor-infiltrating lymphocytes compared with tumor-associated lymphocytes from ascitic fluid and peripheral blood lymphocytes in patients with advanced ovarian cancer. Gynecol Obstet Invest 51(4):254–261 Tomsova M, Melichar B, Sedlakova I, Steiner I, 2008, Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma. Gynecol Oncol 108(2):415–420 Rolland P, Spendlove I, Madjid Z, Rakha EA, Patel P, Ellis IO et al, 2007, The p53 positive Bcl-2 negative phenotype is an independent marker of prognosis in breast cancer. Int J Cancer 120(6):1311–1317 Vicari AP, Treilleux I, Lebecque S, 2004, Regulation of the trafficking of tumour-infiltrating dendritic cells by chemokines. Semin Cancer Biol 14(3):161–169 Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P et al, 2004, Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 10(9):942–949 Bernsen MR, Diepstra JH, van Mil P, Punt CJ, Figdor CG, van Muijen GN et al, 2004, Presence and localization of T-cell subsets in relation to melanocyte differentiation antigen expression and tumour regression as assessed by immunohistochemistry and molecular analysis of microdissected T cells. J Pathol 202(1):70–79 Haas M, Dimmler A, Hohenberger W, Grabenbauer GG, Niedobitek G, Distel LV, 2009, Stromal regulatory T-cells are associated with a favourable prognosis in gastric cancer of the cardia. BMC Gastroenterol. 9:65 Menetrier-Caux C, Gobert M, Caux C, 2009, Differences in tumor regulatory T-cell localization and activation status impact patient outcome. Canc Res Mizukami Y, Kono K, Kawaguchi Y, Akaike H, Kamimura K, Sugai H et al, 2008, Localisation pattern of Foxp3+ regulatory T cells is associated with clinical behaviour in gastric cancer. Br J Cancer 98(1):148–153 Leffers N, Gooden MJ, de Jong RA, Hoogeboom BN, ten Hoor KA, Hollema H et al, 2009, Prognostic significance of tumorinfiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunol Immunother 58, 3):449–459 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 361 EP 370 DI 10.1007/s12253-009-9230-5 PG 10 ER PT J AU Gunia, S May, M Koch, S Dietel, M Erbersdobler, A AF Gunia, Sven May, Matthias Koch, Stefan Dietel, Manfred Erbersdobler, Andreas TI MUC1 Expression in Incidental Prostate Cancer Predicts Staging and Grading on the Subsequent Radical Prostatectomy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Incidental prostate cancer; MUC1 (episialin); Immunohistochemistry; Histopathologic outcome parameters; Radical prostatectomy ID Incidental prostate cancer; MUC1 (episialin); Immunohistochemistry; Histopathologic outcome parameters; Radical prostatectomy AB The behavior of Incidental prostate cancer (IPC) cannot be reliably predicted by means of conventional histomorphology. MUC1 (episialin) expression has been linked to poor outcome in peripheral prostate cancer (PC). We aimed to determine the so far neglected prognostic role of MUC1 expression in IPC which most commonly represents transition zone cancer. Using Tissue microarray (TMA), we assessed the association between MUC1 expression recorded in transurethral resection specimens of the prostate (TURP chips) and histopathologic outcome parameters (Gleason scores and histologic staging) performed on the subsequent radical prostatectomies (RPs) in a study cohort of 54 patients. Due to tissue loss during arraying and sectioning, a total of 44 (81.5%) tumor samples remained available for immunostaining which was dichotomized by two independent clinical pathologists as being absent or present. MUC1 expression was present in 7 (15.9%) of the 44 IPC immunohistochemically investigated with a striking overrepresentation in high stage tumors, and was significantly correlated with histopathologic staging (ρ=0.4; p=0.02) and Gleason scores (ρ=0.3; p=0.03) performed on the corresponding RPs. These data were confirmed by means of the McNemar test (staging: p=0.01; grading: p=0.04). Our findings suggest that MUC1 might become a valuable adjunct to enable individual prognostic ramification prior to radical surgery in prostate cancer histologically detected in TURP chips. This interesting observation clearly awaits validation by larger studies surveying clinical follow-up data. C1 [Gunia, Sven] Charite-University Medicine Teaching Hospital, Department of Pathology, Pieskower Straße 33, 15526 Bad Saarow, Germany. [May, Matthias] Klinikum St. Elisabeth, Department of UrologyStraubing, Germany. [Koch, Stefan] Charite-University Medicine Teaching Hospital, Department of Pathology, Pieskower Straße 33, 15526 Bad Saarow, Germany. [Dietel, Manfred] Charite University Hospital, Institute of Pathology, Campus Charite MitteBerlin, Germany. [Erbersdobler, Andreas] Charite University Hospital, Institute of Pathology, Campus Charite MitteBerlin, Germany. RP Gunia, S (reprint author), Charite-University Medicine Teaching Hospital, Department of Pathology, 15526 Bad Saarow, Germany. EM sven.gunia@helios-kliniken.de CR Van Andel G, Vleeming R, Kurth KH, de Reijke TM, 1995, Incidental carcinoma of the prostate. Semin Surg Oncol 11:36–45 Sheldon CA, Williams RD, Fraley EE, 1980, Incidental carcinoma of the prostate: a review of the literature and critical reappraisal of classification. J Urol 124:626–631 Sobin LH, Wittekind CH, eds,, 2002, TNM classification of malignant tumors. 6th edition. New York: Wiley-Liss, pp 172–175 Melchior S, Hadaschik B, Thuroff S, Thomas C, Gillitzer R, Thuroff J, 2008, Outcome of radical prostatectomy for incidental carcinoma of the prostate. BJU Int Dec 5 [Epub ahead of print] Gum JR, 1992)Mucin genes and the proteins they encode: structure, diversity, and regulation. Am J Respir Cell Mol Biol 7:557–564 Jass JR, Roberton AM, 1994, Colorectal mucin histochemistry in health and disease: a critical review. Pathol Int 44:487–504 Baldus SE, Palmen C, Thiele J, 2007, MUC1, EMA, expressing plasma cells in bone marrow infiltrated by plasma cell myeloma. Histol Histopathol 22:889–893 Yang E, Hu XF, Xing PX, 2007, Advances of MUC1 as a target for breast cancer immunotherapy. Histol Histopathol 22:905–922 Garbar C, Mascaux C, Wespes E, 2008, Expression of MUC1 and sialyl-Tn in benign prostatic glands, high-grade prostate intraepithelial neoplasia and malignant prostatic glands: a preliminary study. Anal Quant Cytol Histol 30:71–77 Burke PA, Gregg JP, Bakhtiar B, Beckett LA, Denardo GL, Albrecht H, De Vere White RW, De Nardo SJ, 2006, Characterization of MUC1 glycoprotein on prostate cancer for selection of targeting molecules. Int J Oncol 29:49–55 Li Y, Cozzi PJ, 2007, MUC1 is a promising therapeutic target for prostate cancer therapy. Curr Cancer Drug Targets 7:259–271 Strawbridge RJ, Nister M, Brismar K, Li C, Lindstrom S, 2008, Influence of MUC1 genetic variation on prostate cancer risk and survival. Eur J Hum Genet 16:1521–1525 Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP, 1998, Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4:844–847 Paul R, Knebel C, van Radenborgh H, Kubler H, Alschibaja M, Gunther M, Hartung R, 2005, Incidental carcinoma of the prostate: can we and should we recommend radical prostatectomy? Urologe A 44:1054–1058 Gunia S,Koch S,May M, Dietel M, ErbersdoblerA(2009, Expression of prostatic acid phosphatase, PSAP, in transurethral resection specimens of the prostate is predictive of histopathologic tumor stage in subsequent radical prostetctomies. Virchows Arch 454:573–579 Morrison C, Merati K,MarshWL Jr, De Lott L, Cohn DE, Young G, Frankel WL, 2007, The mucin expression profile of endometrial carcinoma and correlation with clinical-pathologic parameters. Appl Immunohistochem Mol Morphol 15:426–431 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 371 EP 375 DI 10.1007/s12253-009-9231-4 PG 5 ER PT J AU Smuk, G Illes, Keresztes, K Kereskai, L Marton, B Nagy, Zs Lacza, Pajor, L AF Smuk, Gabor Illes, Arpad Keresztes, Katalin Kereskai, Laszlo Marton, Balazs Nagy, Zsofia Lacza, Agnes Pajor, Laszlo TI Pheno- and Genotypic Features of Epstein-Barr Virus Associated B-Cell Lymphoproliferations in Peripheral T-Cell Lymphomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE B-cell lymphoproliferation; Epstein-Barr virus; T-cell lymphoma ID B-cell lymphoproliferation; Epstein-Barr virus; T-cell lymphoma AB Among the 300 peripheral T-cell lymphomas (PTCL) searched for EBV positive non-resting B-cells by EBER in situ hybridization 12 have been identified with various forms of EBV-driven B-cell proliferation. This could be categorized into three major forms. i. In the first form scattered immature, mononuclear B-cells of immuno-, centroblastic type with CD20+. CD30+ CD45+, LMP1+ phenotype, reactive appearance and polyclonal immunoglobulin heavy chains gene rearrangement (IgH-R) were admixed to the PTCL cells. ii. The second form mimicked diffuse large B-cell lymphoma as homogenous sheets, largely demarcated from the PTCL, of mononuclear, immature B-cell of CD20+, CD30+, CD45+, LMP1+, EBNA-2+ phenotype but with lack of monoclonal IgH-R were present. iii. In the third form scattered Hodgkin-Reed-Sternberg (HRS) type of cells were noticed which exhibited the CD15+/−, CD20−/+, CD30+, CD45−, LMP1+, EBNA-2- phenotype and in 50% showed clonal IgH gene rearrangement in whole tissue DNA extract. The IgH associated transcription factors’ (OCT2, BOB.1/OBF.1, PU.1) expression patterns in these cells corresponded to those of HRS cells in cHL. Based on analysis of 65 PTCLs, we have identified in the positive cases a highly significant increase of EBV+ small, reactive, resting B-cell compartment (75.9 / 100 HPF in PTCL vs. 1.5 / 100 HPF in control lymph nodes) likely to be due to the decreased immune surveillance. This progressive accumulation of EBV+ bystander B-cell population in PTCLs might be the source of various B-cell proliferations, which in any form represent major diagnostic pitfalls and require a careful differential diagnostic procedure. C1 [Smuk, Gabor] University of Pecs, Department of Pathology, 12 Szigeti Str, 7624 Pecs, Hungary. [Illes, Arpad] University of Debrecen, Medical and Health Center, 3rd Department of MedicineDebrecen, Hungary. [Keresztes, Katalin] University of Debrecen, Medical and Health Center, 3rd Department of MedicineDebrecen, Hungary. [Kereskai, Laszlo] University of Pecs, Department of Pathology, 12 Szigeti Str, 7624 Pecs, Hungary. [Marton, Balazs] University of Pecs, Department of Pathology, 12 Szigeti Str, 7624 Pecs, Hungary. [Nagy, Zsofia] University of Pecs, Department of Pathology, 12 Szigeti Str, 7624 Pecs, Hungary. [Lacza, Agnes] University of Pecs, Department of Pathology, 12 Szigeti Str, 7624 Pecs, Hungary. [Pajor, Laszlo] University of Pecs, Department of Pathology, 12 Szigeti Str, 7624 Pecs, Hungary. RP Pajor, L (reprint author), University of Pecs, Department of Pathology, 7624 Pecs, Hungary. EM laszlo.pajor@kk.pte.hu;titkar.pathology@kk.pte.hu CR Barry ST, Jaffe SE, Sorbara L et al, 2003, Peripheral T-cell lymphomas expressing CD30 and CD15. Am J Surg Pathol 27:1513–1521 Quintanilla-Martinez L, Fend F, Rodriguez Moguel L et al, 1999, Peripheral T-cell lymphoma with Reed-Sternberg-like cell of Bcell phenotype and genotype associated with Epstein-Barr virus infection. Am J Surg Pathol 23:1233–1240 Pajor L, Kajtar B, Jakso P et al, 2006, Epstein-Barr virus-induced B-cell proliferation of Hodgkin’s and Reed-Sternberg cell phenoand genotype may develop in peripheral T-cell lymphomas. Histopathology 49:553–557 Ho WYJ, Ho CSF, Chan CLA et al, 1998, Frequent detection of Epstein-Barr virus-infected B cells in peripheral T-cell lymphomas. J Pathol 185:79–85 Niedobitek G, Baumann I, Brabletz T et al, 2000, Hodgkin’s disease and peripheral T-cell lymphoma: composite lymphoma with evidence of Epstein-Barr virus infection. J Pathol 191:394–399 Zettl A, Lee S-S, Rudiger T et al, 2002, Epstein-Barr virusassociated B-cell lymphoproliferative disorders in angioimmunoblastic T-cell lymphoma and peripheral T-cell lymphoma, unspecified. Am J Clin Pathol 117:368–379 Tornoczky T, Kelenyi G, Pajor L et al, 1998, EBER oligonucleotide RNA in situ hybridization in EBV associated neoplasms. Patol Oncol Res 4:201–205 Trainor KJ, Brisco MJ, Wan JH et al, 1991, Gene rearrangement in B- and T-lymphoproliferative disease detected by the polymerase chain reaction. Blood 78:192–196 Pajor L, Matolcsy A, Vass JA et al, 1998, Phenotypic and genotypic analyses of blastic cell population suggest that pure Blymphoblastic leukaemia may arise from myelodysplastic syndrome. Leuk Res 22:13–17 Higgins PTJ, Rijn M, Jones DC et al, 2000, Peripheral T-cell lymphoma complicated by a proliferation of large B cells. Am J Clin Pathol 114:236–247 Reichard KK, Schwartz JE, Higgins PJ et al, 2006, CD10 expression in peripheral T-cell lymphomas complicated by a proliferation of large B-cells. Modern Pathol 19:337–343 Thorley-Lawson AD, Gross A, 2004, Persistence of the Epstein- Barr virus and the origins of associated lymphomas. N Engl J Med 350:1328–1337 Klein E, 2004, Non-proliferative interactions of Epstein-Barr virus and human B lymphocytes. Fiola Biol-Prague 50:131–135 Amon W, Farrell JP, 2005, Reactivation of Epstein-Barr virus from latency. Rev Med Virol 15:149–156 Atayar C, Poppema S, Blokzijl T et al, 2005, Expression of the Tcell transciption factors, GATA-3 and T-bet, in the neoplastic cells of Hodgkin lymphomas. Am J Pathol 166:127–134 McCune CR, Syrbu IS, Vasef AM, 2006, Expression profiling of transcription factors Pax-5, Oct-1, Oct-2, BOB.1, and PU.1 in Hodgkin’s and non-Hodgkin’s lymphomas: a comparative study using high throughput tissue microarrays. Modern Pathol 19:1010–1018 Korbjuhn P, Anagnostopoulos I, Hummel M et al, 1993, Frequent latent Epstein-Barr virus infection of neoplastic T cells and Bystander B cells in human immunodeficiency virus-negative European peripheral pleomorphic T-cell lymphomas. Blood 82:217–223 Suwiwat S, Pradutkanchana J, Ishida T et al, 2007, Quantitative analysis of cell-free Epstein-Barr virus DNA in the plasma of patients with peripheral T-cell and NK-cell lymphomas and peripheral T-cell proliferative diseases. J Clin Virol 40:277–283 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 377 EP 383 DI 10.1007/s12253-009-9233-2 PG 7 ER PT J AU Lu, HCh Chou, FP Yeh, KT Chang, YS Hsu, CN Chang, JG AF Lu, Hsiu-Chin Chou, Fen-Pi Yeh, Kun-Tu Chang, Ya-Sian Hsu, C Nicholas Chang, Jan-Gowth TI Expression of Protein Kinase C Family in Human Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Protein kinase C; Human hepatocellular carcinoma ID Protein kinase C; Human hepatocellular carcinoma AB Protein kinase Cs (PKCs) play important roles in signal transduction, cell regulation, and tumor formation. In the present study, we analyzed the expression of PKCs in human hepatocellular carcinoma (HCC) tissues and explored their roles in the development of HCC. Real-time quantitative PCR and immunohistochemistry showed that PKCβ and PKCθ were down-regulated in HCC tissues. Reduced expression of PKCθ is well correlated with the grade of cancer cells (p=0.009), and the down-regulated expression of PKCβII is associated with HBV infection (p=0.035). Our findings suggest particular roles of the two PKC isoenzymes in the hepatocarcinogenesis of human HCC. C1 [Lu, Hsiu-Chin] Kaohsiung Medical University, College of Medicine, Kaohsiung Medical University Hospital, Institute of Clinical Research, Department of Laboratory MedicineKaohsiung, Taiwan, Republic of China. [Chou, Fen-Pi] Chung Shan Medical University, Department of Medical Laboratory and BiotechnologyTaichung, Taiwan, Republic of China. [Yeh, Kun-Tu] Changhua Christian Hospital, Department of PathologyChanghua, Taiwan, Republic of China. [Chang, Ya-Sian] Kaohsiung Medical University, College of Medicine, Kaohsiung Medical University Hospital, Institute of Clinical Research, Department of Laboratory MedicineKaohsiung, Taiwan, Republic of China. [Hsu, C Nicholas] Kaohsiung Medical University, College of Medicine, Kaohsiung Medical University Hospital, Institute of Clinical Research, Department of Laboratory MedicineKaohsiung, Taiwan, Republic of China. [Chang, Jan-Gowth] Kaohsiung Medical University, College of Medicine, Kaohsiung Medical University Hospital, Institute of Clinical Research, Department of Laboratory MedicineKaohsiung, Taiwan, Republic of China. RP Chang, JG (reprint author), Kaohsiung Medical University, College of Medicine, Kaohsiung Medical University Hospital, Institute of Clinical Research, Department of Laboratory Medicine, Kaohsiung, Taiwan, Republic of China. EM jgchang@ms.kmuh.org.tw CR El-Serag HB, Rudolph KL, 2007, Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterol 132:2557–2576 Marrero CR, Marrero JA, 2007, Viral hepatitis and hepatocellular carcinoma. Arch Med Res 38:612–620 McGlynn KA, London WT, 2005, Epidemiology and natural history of hepatocellular carcinoma. Best Pract Res Clin Gastroenterol 19:3–23 Morgan TR, Mandayam S, Jamal MM, 2004, Alcohol and hepatocellular carcinoma. Gastroenterol 127:S87–96 Seeff LB, Hoofnagle JH, 2006, Epidemiology of hepatocellular carcinoma in areas of low hepatitis B and hepatitis C endemicity. Oncogene 25:3771–3777 Feitelson MA, Sun B, Satiroglu Tufan NL et al, 2002, Genetic mechanisms of hepatocarcinogenesis. Oncogene 21:2593–2604 Laurent-Puig P, Zucman-Rossi J, 2006, Genetics of hepatocellular tumors. Oncogene 25:3778–3786 Thorgeirsson SS, Grisham JW, 2002, Molecular pathogenesis of human hepatocellular carcinoma. Nat Genet 31:339–346 Villanueva A, Newell P, Chiang DY et al, 2007, Genomics and signaling pathways in hepatocellular carcinoma. Semin Liver Dis 27:55–76 Mellor H, Parker PJ, 1998, The extended protein kinase C superfamily. Biochem J 332(Pt 2):281–292 Newton AC, 2001, Protein kinase C: structural and spatial regulation by phosphorylation, cofactors, and macromolecular interactions. Chem Rev 101:2353–2364 Nishizuka Y, 1992, Intracellular signaling by hydrolysis of phospholipids and activation of protein kinase C. Science 258:607–614 Ohno S, Nishizuka Y, 2002, Protein kinase C isotypes and their specific functions: prologue. J Biochem 132:509–511 Besson A, Yong VW, 2000, Involvement of p21(Waf1/Cip1, in protein kinase C alpha-induced cell cycle progression. Mol Cell Biol 20:4580–4590 Griner EM, Kazanietz MG, 2007, Protein kinase C and other diacylglycerol effectors in cancer. Nat Rev Cancer 7:281–294 Gutcher I, Webb PR, Anderson NG, 2003, The isoform-specific regulation of apoptosis by protein kinase C. Cell Mol Life Sci 60:1061–1070 Koivunen J, Aaltonen V, Peltonen J, 2006, Protein kinase C, PKC, family in cancer progression. Cancer Lett 235:1–10 Mandil R, Ashkenazi E, Blass M et al, 2001, Protein kinase Calpha and protein kinase Cdelta play opposite roles in the proliferation and apoptosis of glioma cells. Cancer Res 61:4612– 4619 Ng T, Shima D, Squire A et al, 1999, PKCalpha regulates beta1 integrin-dependent cell motility through association and control of integrin traffic. EMBO J 18:3909–3923 Nishizuka Y, 1995, Protein kinase C and lipid signaling for sustained cellular responses. FASEB J 9:484–496 Scaglione-Sewell B, Abraham C, Bissonnette M et al, 1998, Decreased PKC-alpha expression increases cellular proliferation, decreases differentiation, and enhances the transformed phenotype of CaCo-2 cells. Cancer Res 58:1074–1081 Wetsel WC, Khan WA, Merchenthaler I et al, 1992, Tissue and cellular distribution of the extended family of protein kinase C isoenzymes. J Cell Biol 117:121–133 Fournier DB, Chisamore M, Lurain JR et al, 2001, Protein kinase C alpha expression is inversely related to ER status in endometrial carcinoma: possible role in AP-1-mediated proliferation of ER-negative endometrial cancer. Gynecol Oncol 81:366–372 Koren R, Ben Meir D, Langzam L et al, 2004, Expression of protein kinase C isoenzymes in benign hyperplasia and carcinoma of prostate. Oncol Rep 11:321–326 Koren R, Langzam L, Paz A et al, 2000, Protein kinase C, PKC, isoenzymes immunohistochemistry in lymph node revealing solution-fixed, paraffin-embedded bladder tumors. Appl Immunohistochem Mol Morphol 8:166–171 Langzam L, Koren R, Gal R et al, 2001, Patterns of protein kinase C isoenzyme expression in transitional cell carcinoma of bladder. Relation to degree of malignancy. Am J Clin Pathol 116:377–385 Martinez-Gimeno C, Diaz-Meco MT, Dominguez I et al, 1995, Alterations in levels of different protein kinase C isotypes and their influence on behavior of squamous cell carcinoma of the oral cavity: epsilon PKC, a novel prognostic factor for relapse and survival. Head Neck 17:516–525 Varga A, Czifra G, Tallai B et al, 2004, Tumor grade-dependent alterations in the protein kinase C isoform pattern in urinary bladder carcinomas. Eur Urol 46:462–465 Ainsworth PD, Winstanley JH, Pearson JM et al, 2004, Protein kinase C alpha expression in normal breast, ductal carcinoma in situ and invasive ductal carcinoma. Eur J Cancer 40:2269–2273 Gokmen-Polar Y, Murray NR, Velasco MA et al, 2001, Elevated protein kinase C betaII is an early promotive event in colon carcinogenesis. Cancer Res 61:1375–1381 Kerfoot C, Huang W, Rotenberg SA, 2004, Immunohistochemical analysis of advanced human breast carcinomas reveals downregulation of protein kinase C alpha. J Histochem Cytochem 52:419–422 Neill GW, Ghali LR, Green JL et al, 2003, Loss of protein kinase Calpha expression may enhance the tumorigenic potential of Gli1 in basal cell carcinoma. Cancer Res 63:4692–4697 Tsai JH, Hsieh YS, Kuo SJ et al, 2000, Alteration in the expression of protein kinase C isoforms in human hepatocellular carcinoma. Cancer Lett 161:171–175 Edmondson HA, Steiner PE, 1954, Primary carcinoma of the liver: a study of 100 cases among 48, 900 necropsies. Cancer 7:462–503 Yeh KT, Yang MY, Liu TC et al, 2005, Abnormal expression of period 1, PER1, in endometrial carcinoma. J Pathol 206:111–120 Yuo CY, Lin HH, Chang YS et al, 2008, 5-(N-ethyl-N-isopropyl)- amiloride enhances SMN2 exon 7 inclusion and protein expression in spinal muscular atrophy cells. Ann Neurol 63:26–34 Engers R, Mrzyk S, Springer E et al, 2000, Protein kinase C in human renal cell carcinomas: role in invasion and differential isoenzyme expression. Br J Cancer 82:1063–1069 Kamimura K, Hojo H, Abe M, 2004, Characterization of expression of protein kinase C isozymes in human B-cell lymphoma: relationship between its expression and prognosis. Pathol Int 54:224–230 Weichert W, Gekeler V, Denkert C et al, 2003, Protein kinase C isoform expression in ovarian carcinoma correlates with indicators of poor prognosis. Int J Oncol 23:633–639 Wu TT, Hsieh YH, Wu CC et al, 2007, Overexpression of protein kinase C alpha mRNA in human hepatocellular carcinoma: a potential marker of disease prognosis. Clin Chim Acta 382:54–58 Chang KJ, Lin JK, Lee PH et al, 1996, The altered activity of membrane-bound protein kinase C in human liver cancer. Cancer Lett 105:211–215 Hsieh YH, Wu TT, Tsai JH et al, 2006, PKCalpha expression regulated by Elk-1 and MZF-1 in human HCC cells. Biochem Biophys Res Commun 339:217–225 Wu TT, Hsieh YH, Hsieh YS et al, 2008, Reduction of PKC alpha decreases cell proliferation, migration, and invasion of human malignant hepatocellular carcinoma. J Cell Biochem 103:9–20 Heo KS, Kim DU, Kim L et al, 2008, Activation of PKCbeta(II, and PKCtheta is essential for LDL-induced cell proliferation of human aortic smooth muscle cells via Gi-mediated Erk1/2 activation and Egr-1 upregulation. Biochem Biophys Res Commun 368:126–131 Nagahama K, Ogawa A, Shirane K et al, 2008, Protein kinase C theta plays a fundamental role in different types of chronic colitis. Gastroenterol 134:459–469 Hayashi K, Altman A, 2007, Protein kinase C theta, PKCtheta): a key player in T cell life and death. Pharmacol Res 55:537–544 Sims TN, Soos TJ, Xenias HS et al, 2007, Opposing effects of PKCtheta and WASp on symmetry breaking and relocation of the immunological synapse. Cell 129:773–785 Ou WB, Zhu MJ, Demetri GD et al, 2008, Protein kinase C-theta regulates KIT expression and proliferation in gastrointestinal stromal tumors. Oncogene 27:5624–5634 Belguise K, Sonenshein GE, 2007, PKCtheta promotes c-Reldriven mammary tumorigenesis in mice and humans by repressing estrogen receptor alpha synthesis. J Clin Invest 117:4009–4021 Cen L, Arnoczky KJ, Hsieh FC et al, 2007, Phosphorylation profiles of protein kinases in alveolar and embryonal rhabdomyosarcoma. Mod Pathol 20:936–946 Despouy G, Joiner M, Le Toriellec E et al, 2007, The TCL1 oncoprotein inhibits activation-induced cell death by impairing PKCtheta and ERK pathways. Blood 110:4406–4416 Kim KM, Kang DW, Moon WS et al, 2006, PKCtheta expression in gastrointestinal stromal tumor. Mod Pathol 19:1480–1486 Gao Z, Wang Z, Zhang X et al, 2007, Inactivation of PKCtheta leads to increased susceptibility to obesity and dietary insulin resistance in mice. Am J Physiol Endocrinol Metab 292:E84– 91 Haasch D, Berg C, Clampit JE et al, 2006, PKCtheta is a key player in the development of insulin resistance. Biochem Biophys Res Commun 343:361–368 Garaude J, Kaminski S, Charni S et al, 2008, Impaired anti-leukemic immune response in PKCtheta-deficient mice. Mol Immunol 45:3463–3469 Schaffel R, Morais JC, Biasoli I et al, 2007, PKC-beta II expression has prognostic impact in nodal diffuse large B-cell lymphoma. Mod Pathol 20:326–330 Bazarsky E, Wolfson M, Galron D et al, 1997, Persistent measles virus infection of murine neuroblastoma cells differentially affects the expression of PKC individual isoenzymes. Virus Genes 15:227–234 Langzam L, Koren R, Gal R et al, 2001, Patterns of protein kinase C isoenzyme expression in transitional cell carcinoma of bladder. Am J Clin Pathol 116:377–385 Filmus J, Capurro M, 2004, Glypican-3 and alphafetoprotein as diagnostic tests for hepatocellular carcinoma. Mol Diagn 8:207–212 Lin YC, Lee PH, Yao YT et al, 2007, Alpha-fetoproteinproducing pancreatic acinar cell carcinoma. J Formos Med Assoc 106:669–672 Yamada K, Fujioka Y, Ebihara Y et al, 1994, Alpha-fetoprotein producing undifferentiated carcinoma of the bladder. J Urol 152:958–960 Chen DS, Sung JL, Sheu JC et al, 1984, Serum alpha-fetoprotein in the early stage of human hepatocellular carcinoma. Gastroenterol 86:1404–1409 Sherman M, 2001, Alphafetoprotein: an obituary. J Hepatol 34:603–605 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 385 EP 391 DI 10.1007/s12253-009-9228-z PG 7 ER PT J AU Vuletic, A Konjevic, G Milanovic, D Ruzdijic, S Jurisic, V AF Vuletic, Ana Konjevic, Gordana Milanovic, Desanka Ruzdijic, Sabera Jurisic, Vladimir TI Antiproliferative Effect of 13-cis-Retinoic Acid is Associated with Granulocyte Differentiation and Decrease in Cyclin B1 and Bcl-2 Protein Levels in G0/G1 Arrested HL-60 Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cell cycle; Promyelocytic leukemia; Proliferation; Retinoic acid; Differentiation ID Cell cycle; Promyelocytic leukemia; Proliferation; Retinoic acid; Differentiation AB Retinoic acid (RA), similar to specific growth factors, can induce differentiation of proliferating promyelocytic precursors into terminally differentiated granulocytes, although little is known about effects of its 13-cis isomer on promyelocytic leukemia (PML). In this study we demonstrate that 13-cis-RA has a dose and time-dependant antiproliferative effect on HL-60 PML cell line, that it induces cell accumulation in resting G0/G1 phase of the cell cycle followed by an increase in CD11b granulocyte differentiation antigen expression. The obtained increase in the percentage of HL-60 cells in G0/G1 phase and complementary decrease in S phase of the cell cycle are accompanied by a decrease in the expression of cell cycle regulatory molecule cyclin B1. We also show the induction of interferon regulatory factor-1 (IRF-1) transcription that can, also, to some extent contribute to the antiproliferative effect of 13-cis-RA. Furthermore, down-regulation of Bcl-2 protein expression in 13-cis-RA treated HL-60 cells may contribute to sensitivity to apoptosis of growth arrested HL-60 promyelocytic cells. C1 [Vuletic, Ana] Institute of Oncology and Radiology of SerbiaBelgrade, Serbia. [Konjevic, Gordana] Institute of Oncology and Radiology of SerbiaBelgrade, Serbia. [Milanovic, Desanka] University of Belgrade, Institute for Biological ResearchBelgrade, Serbia. [Ruzdijic, Sabera] University of Belgrade, Institute for Biological ResearchBelgrade, Serbia. [Jurisic, Vladimir] University of Kragujevac, Faculty of MedicineKragujevac, Serbia. RP Vuletic, A (reprint author), Institute of Oncology and Radiology of Serbia, Belgrade, Serbia. EM radovanovica@ncrc.ac.rs CR Blaner WS, Olson JA, 1994, Retinol and retinoic acid metabolism. Retinoids: Biology, Chemistry and Medicine. 2nd ed. Raven Press New York, NY Bruserud O, Gjertsen BT, Huang T-S, 2000, Induction of apoptosisa possible strategy in treatment of adult acute myelogeneous leukemia. Oncologist 5:454–462 Passegue E, Jamieson CH, Ailles LE, Weissman IL, 2003, Normal and leukemic hematopoiesis: are leukemias a stem cell disorder or a reacquisition of stem cell characteristics? PNAS 100:11842–11849 Nelson AM, Gilliland KL, Cong Z, Thiboutot DM, 2006, 13-cis Retinoic acid induces apoptosis and cell cycle arrest in human SEB-1 sebocytes. J Invest Dermatol 126:2178–2189 Alltrucci L, Gronemyer H, 2001, Nuclear receptors in cell life and death. Trends Endocrinology Methabolism 10:460–468 Melnick A, Licht JD, 1999, Deconstructing a disease: RARalpha, its fusion partners, and their roles in the pathogenesis of acute promyelocytic leukemia. Blood 93:3167–3215 Price NC, 1996, Proteins. Academic Press, Labfax, Oxford Ikezoe T, Daar ES, Hisatake J, Taguchi H, Koeffler HP, 2000, HIV-1 protease inhibitors decrease proliferation and induce differentiation of human myelocytic leukemia cells. Blood 96: 3553–3559 Gianni M, Terao M, Fortino I, Calzi M, Viggiano V, Barbui T, Rambaldi A, Garattini E, 1997, Stat1 is induced and activated by all-trans retinoic acid in acute promyelocytic leukemia cells. Blood 89:1001–1012 Lanvers C, Hempel G, Blaschke G, Boos J, 1998, Chemically induced isomerization and differential uptake modulate retinoic acid deposition in HL-60 cells. FASEB 15:1627–1633 Dimberg A, Bahram F, Kalberg I, Larsson L-G, Nilsson F, Oberg K, 2002, Retinoic acid-induced cell cycle arrest of human myeloid cell lines is associated with sequental down-regulation of c-Myc and cyclin E and posttranscriptional up-regulaiton of p27Kip 1. Blood 99:2199–21206 Wang JG, Barsky LW, Davicioni E, Weinberg KI, Triche TJ, Zhang XK, Wu L, 2006, Retinoic acid induces leukemia cell G1 arrest and transition into differentiation by inhibiting cyclindependent kinase-activating kinase binding and phosphorylation of PML/RARalpha. FASEB 20:2142–2144 Liu T-X, Zhang W, Tao J, Zhang R-B, Zhang Q-H, Zhao C-J, Tong J-H, Lanotte M, Waxman S, Chen S-J, Mao M, Hu G-X, Zhu L, Chen Z, 2000, Gene expression underlying retinoic acid induced differentiation of acute promyelocytic leukemia cells. Blood 96:1496–1504 Breitman TR, Collins SJ, Keene BR, 1981, Terminal differentiation of human promyelocytic leukemic cells in primary culture in response to retinoic acid. Blood 57:1000–1004 Grignani F, Testa U, Rogaia D, Ferrucci PF, Samoggia P, Pinto A, Aldinucci D, Gelmetti V, Fagioli M, Alcalay M, Seeler J, Grignani F, Nicoletti I, Peschle C, Pelicci PG, 1996, Effects on differentiation by the promyelocytic leukemia PML/RARalpha protein depend on the fusion of the PML protein dimerization and RARalpha DNA binding domains. EMBO J 15:4949–4958 Shiohara M, Dawson MI, Hobbs PD, Sawai N, Higuchi T, Koike K, Komiyama A, Koeffler HP, 1999, Effects of novel RAR- and RXR-selective retinoids on myeloid leukemic proliferation and differentiation in vitro. Blood 93:2057–2066 Stabellini G, Brugnoli F, Calastrini C, Vizzotto L, Vertemati M, Baroni T, Caramelli E, Marinucci L, Pellati A, Bertagnolo V, 2004, Ornithine decarboxilase, polyamines and CD11b expression in HL-60 cells during differentiation induced by retinoic acid. Biomed Pharmacother 58:401–4066 Li S-W, Tang D, Ahrens KP, She J-X, Braylan RC, Yang L, 2003, All-trans-retinoic acid induces CD52 expression in acute promyelocytic leukemia. Blood 101:1977–1980 Zhu J, Gianni M, Kopf E, Honore N, Chelbi-Alix M, Koken M, Quignon F, Egly CR, de The H, 1999, Retinoic acid induces proteasome-dependent degradation of retinoic acid receptor α, RARα, and oncogenic RARα fusion proteins. PNAS 26:14807– 14812 Hochstrasser M, 1995, Ubiquitin, proteasomes, and the regulation of intracellular protein degradation. Curr Opin Cell Biol 7:215– 222 Muller S, Matunis M, Dejean A, 1998, Conjugation with the ubiquitin-related modifer SUMO-1 regulates the partitioning of PML within the nucleus. EMBOJ 17:61–70 Morosetti R, Park DJ, Chumakov AM, Grillier I, Shiohara M, Gombart AF, Nakamaki T, Weinberg K, Koeffler HP, 1997, A novel, myeloid transcription factor, C/EBP epsilon, is upregulated during granulocytic, but not monocytic, differentiation. Blood 90:2591–2600 Hunter T, Pines J, 1994, Cyclins and cancer. II: Cyclin D and CDK inhibitors come of age Cell 79:573–582 Burger C, Wick M, Muller R, 1994, Lineage Lineage specific regulation of cell cycle gene expression in differentiating myeloid cells. J Cell Sci 107:2047–2054 Morgan MA, Dolp O, Reuter CW, 2001, Cell-cycle-dependent activation of mitogen-ac activated protein kinase kinase, MEK-1/2, in myeloid leukemia cell lines and induction of growth inhibition and apoptosis by inhibitors of RAS signaling. Blood 97:1823–1834 Miranda MB, McGuire TF, Johnson DE, 2002, Importance of MEK1/2 signaling in monocytic and granulocytic differentiation of myeloid cell lines. Leukemia 16:683–692. Yen A, Sturgil R, Varvayanis S, 1997, Increasing c-FMS expression decreases retinoic acid concentration needed to cause cell differentiation and Rb hypophosphorylation. Can Res 57:2020–2028 Andreeff M, Jiang S, Zhang X, Konopleva M, Estrov Z, Snell VE, Xie Z, Okcu MF, Sanchez-Williams G, Dong J, Estey EH, Champlin RC, Kornblau SM, Reed JC, Zhao S, 1999, Expression of Bcl-2 related genes in normal and AML progenitors: changed induced by chemotherapy and retinoic acid. Leukemia 13:1881– 1892 Campos L, Rouault JP, Sabido O, Oriol P, Roubi N, Vasselon C, Archimbaud E, Magaud JP, Guyotat D, 1993, High expression of Bcl-2 in acute promyelocitic leukemia is associated with poor response to chemotherapy. Blood 81:3091–3096 Naumovski L, Cleary ML, 1994, Bcl-2 inhibits apopotsis associated with terminal differentiation of HL-60 myeloid leukemia cells. Blood 83:2261–2267 Hu Z-B, Minden MD, McCulloch EA, 1998, Phosphorylation of Bcl-2 after exposure of human leukemic cells to retinoic acid. Blood 5:1768–1775 Slack JL, YuM(1998, Consitutive expression of the promyelocytic leukemia-associated oncogene PML-RARα in TF1 cells:isoformspecific and retinoid acid-dependent effect of growth, bcl-2 expression, and apoptosis. Blood 91:3347–3356 Matikainen S, Ronni T, Hurme M, Pine R, Julkunen I, 1996, Retinoic acid activates interferon regulatory factor-1 gene expression in myeloid cells. Blood 88:114–123 Harada H, Takahashi E, Itoh S, Harada K, Hori TA, Taniguchi T, 1994, Structure and regulation of the human interferon regulatory factor 1, IRF-1, and IRF-2 genes: implications for a gene network in the interferon system. Mol Cell Biol 14:1500–1509 Gianni M, Terao M, Fortino I, LiCalzi M, Viggiano V, Barbui T, Rambaldi A, Garattini E, 1997, Stat1 is induced and activated by all-trans retinoic acid in acute promyelocytic leukemia cells. Blood 89:1001–1012 Matikainen S, Lehtonen A, Sareneva T, Julkunen I, 1998, Regulation of IRF and STAT gene expression by retinoic acid. Leuk Lymphoma 30:63–71 Percario Z, Giandomenico V, Chiantore MV, Vannucchi S, Hiscott J, Affabris E, 1999, Retinoic acid is able to induce interferon regulatory factor 1 in squamous carcinoma cell line via a STAT-1 independent signaling pathway. Cell Growth Differ 10:263–270 Chow WA, Fang JJ, Yee JK, 2000, The IFN regulatory factor family participates in regulation of Fas ligand gene expression in T cells. J Immunol 164:3512–3518 Flynn PJ, Miller WJ, Weisdorf DJ, Arthur DC, Brunning R, Branda RF, 1983, Retinoic acid treatment of acute promyelocytic leukemia: in vitro and in vivo observations. Blood 62(6):1211– 1217 Graf N, Riesinger P, Reinhard H, 1995, Retinoids in the treatment of acute promyelocytic leukemia. Review of the literature Klin Padiatr 207(2):43–47 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 393 EP 401 DI 10.1007/s12253-009-9241-2 PG 9 ER PT J AU Sun, Chm Huang, Shf Zeng, Jm Liu, Db Xiao, Q Tian, Wj Zhu, Xd Huang, Zg Feng, Wl AF Sun, Cheng-ming Huang, Shi-feng Zeng, Jian-ming Liu, Din-bing Xiao, Qing Tian, Wen-jun Zhu, Xi-dan Huang, Zong-gan Feng, Wen-li TI Per2 Inhibits K562 Leukemia Cell Growth In Vitro and In Vivo Through Cell Cycle Arrest and Apoptosis Induction SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Circadian clock; Per2; Apoptosis; Cell cycle arrest; K562 cell ID Circadian clock; Per2; Apoptosis; Cell cycle arrest; K562 cell AB Per2 regulates other molecular and biochemical processes beyond their established role in the regulation of the mammalian circadian clock, herein we investigated the growth inhibiting potential of Per2 in human K562 leukemia cells and the underlying mechanisms .The results showed that over-expression of Per2 induced not only cell cycle arrest at G2/M phase but also an increase in apoptosis, which was confirmed by characteristic morphological changes, FCM and evident DNA fragmentation. Further experiments confirmed both up-regulation of P53 and down-regulation of CylinB1and C-myc. On the other hand, while P53 was found to be down-regulated. CylinB1 and C-myc were up-regulated. after Per2 knockdown. In leukemia mice, Per2 transfection was shown to suppress cellular proliferation and accelerate apoptosis of K562 cells. Moreover, fewer leukemia cells were found to have infiltrated into the livers and spleens of the mice from the Per2 transfected group as compared with those from the control group. In summary, Per2 displayed a significant anti-tumor effect through cell cycle arrest and apoptosis induction in K562 cells. These data further support the emerging role of the circadian clock in critical aspects of cancer development and thorough research is underway on the mechanism of Per2 in the leukemia. C1 [Sun, Cheng-ming] Ministry of Education, Chongqing Medical University, Key Laboratory of Laboratory Medical Diagnostics, Department of Clinical Hematology, 400016 Chongqing, China. [Huang, Shi-feng] Ministry of Education, Chongqing Medical University, Key Laboratory of Laboratory Medical Diagnostics, Department of Clinical Hematology, 400016 Chongqing, China. [Zeng, Jian-ming] Ministry of Education, Chongqing Medical University, Key Laboratory of Laboratory Medical Diagnostics, Department of Clinical Hematology, 400016 Chongqing, China. [Liu, Din-bing] Ministry of Education, Chongqing Medical University, Key Laboratory of Laboratory Medical Diagnostics, Department of Clinical Hematology, 400016 Chongqing, China. [Xiao, Qing] Chongqing Medical University, Clinic College, Department of Hematology, 400016 Chongqing, China. [Tian, Wen-jun] Ministry of Education, Chongqing Medical University, Key Laboratory of Laboratory Medical Diagnostics, Department of Clinical Hematology, 400016 Chongqing, China. [Zhu, Xi-dan] Ministry of Education, Chongqing Medical University, Key Laboratory of Laboratory Medical Diagnostics, Department of Clinical Hematology, 400016 Chongqing, China. [Huang, Zong-gan] Chongqing Medical University, Clinic College, Department of Hematology, 400016 Chongqing, China. [Feng, Wen-li] Ministry of Education, Chongqing Medical University, Key Laboratory of Laboratory Medical Diagnostics, Department of Clinical Hematology, 400016 Chongqing, China. RP Feng, Wl (reprint author), Ministry of Education, Chongqing Medical University, Key Laboratory of Laboratory Medical Diagnostics, Department of Clinical Hematology, 400016 Chongqing, China. EM fengwlcqmu@sina.com CR Reppert SM, Weaver DR, 2002, Coordination of circadian timing in mammals. Nature 418:935–941 Schibler U, Sassone-Corsi P, 2002, A web of circadian pacemakers. Cell 111:919–922 Lowrey PL, Takahashi JS, 2004, Mammalian circadian biology: Elucidating genome-wide levels of temporal organization. Ann Rev Genomics Hum Genet. 5:407–441 Ko CH, Takahashi JS, 2006, Molecular components of the mammalian circadian clock. Hum Mol Genet 15:271–277 Zheng B, Larkin DW, Albrecht U et al, 1999, The mPer2 gene encodes a functional component of the mammalian circadian clock. Nature 400:169–173 Zheng B, Albrecht U, Kaasik K et al, 2001, Nonredundant roles of the mPer1 and mPer2 genes in the mammalian circadian clock. Cell 105:683–694 Fu L, Lee CC, 2003, The circadian clock: pacemaker and tumour suppressor. Nat Rev Cancer. 3:350–361 Canaple L, Kakizawa T, Laudet V, 2003, The days and nights of cancer cells. Cancer Res 63:7545–7552 Chen ST, Choo KB, 2005, HouM.F., Yeh K.T., Kuo S.J. and Chang J.G, Deregulated expression of the PER1; PER2 and PER3 genes in breast cancers. Carcinogenesis. 26:1241–1246 Gery S, Gombart AF, Yi WS, Koeffler C, Hofmann WK, 2005, and Koeffler H.P, Transcription profiling of C/EBP targetsidentifies Per2 as a gene implicated in myeloid leukemia. Blood. 106:2827–2836 Gery S, Virk RK, Chumakov K, Yu A, 2007, and Koeffler H.P, The clock gene Per2 links the circadian system to the estrogen receptor. Oncogene. 26:7916–7920 Hua H, Wang Z, Halberg F et al, 2006, Circadian gene mPer2 overexpression induces cancer cell apoptosis. Cancer Sci. 97:589–596 MingYu Yang, Yi-Chang Liu, Sheng-Fung Lin et al, 2006, Downregulation of circadian clock genes in chronic myeloid leukemia: alternative methylation pattern of hPER3. Cancer Sci 97:1298–1307 Levi F, Filipski E, Iurisci I, Li XM, Innominato P, 2007, Crosstalks between circadian timing system and cell division cycle determine cancer biology and therapeutics. Cold Spring Harb Symp Quant Biol 72:465–475 Fu L, Pelicano H, Liu J, Lee C, 2002, The circadian gene Period2 plays an important role in tumor suppression and DNA damage response in vivo. Cell 111:41–50 Elledge SJ, 1996, Cell cycle checkpoints: preventing an identity crisis. Science 274:1664–1672 Shah JV, Cleveland DW, 2000, Waiting for anaphase: Mad2 and the spindle assembly checkpoint. Cell 103:997–1000 Fluge O, Bruland O, Akslen LA, Lillehaug JR, Varhaug JE, 2006, Gene expression in poorly differentiated papillary thyroid carcinomas. Thyroid. 16:161–175 Hansel DE, Dhara S, Huang RC, 2005, CDC2/CDK1 expression in esophageal adenocarcinoma and precursor lesions serves as a diagnostic and cancer progression marker and potential novel drug target. Am J Surg Pathol 29:390–399 Ito Y, Takeda T, Sakon M, Monden M, Tsujimoto M, Matsuura N, 2000, Expression and prognostic role of cyclin-dependent kinase 1, cdc2, in hepatocellular carcinoma. Oncology 59:68–74 Matsuo T, Yamaguchi S, Mitsui S, Emi A, Shimoda F, 2003, and Okamura, H, Control mechanism of the circadian clock for timing of cell division in vivo. Science 302:255–259 Granda TG, Liu XH, Smaaland R, Cermakian N, Filipski E, Sassone-Corsi P, Levi F, 2005, Circadian regulation of cell cycle and apoptosis proteins in mouse bone marrow and tumor. FASEB J 19:304–306 Vogelstein B, Lane D, 2000, and Levine A.J, Surfing the p53 network. Nature 408:307–310 Liu G, Chen X, 2006, Regulation of the p53 transcriptional activity. J. Cell. Biochem. 97:448–458 Stark GR, Taylor WR, 2006, Control of the G2/M transition. Mol Biotechnol 32:227–248 Hu R, Aplin AE, 2008, Skp2 regulates G2/M progression in a p53-dependent manner. Mol Biol Cell 19:4602–4610 Blum-Jensen P, Hunter T, 2001, Oncogenic kinase singaling. Nature 411:355–365 Vira M, HenrikssonM, 2006, The Myc oncoprotein as a therapeutic target for human cancer. Semin Cancer Biof. 16:318–330 Wu Ch, Riggelen J, Yetil A, 2007, et a1, Cellular senescence is an important mechanism of tumor regression upon c-Myc inactivation. PNAS. 104:13028–13033 Evan GI, Vousden KH, 2001, Proliferation, cell cycle and apoptosis in cancer. Nature 411:342–348 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 403 EP 411 DI 10.1007/s12253-009-9227-0 PG 9 ER PT J AU Zhong, F Zhang, Sh Shao, Ch Yang, J Wu, X AF Zhong, Fei Zhang, Shineng Shao, Chunkui Yang, Jing Wu, Xiangyuan TI Arsenic Trioxide Inhibits Cholangiocarcinoma Cell Growth and induces Apoptosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Arsenic trioxide; Apoptosis; Cholangiocarcinoma; Mitochondrial pathway; AKT ID Arsenic trioxide; Apoptosis; Cholangiocarcinoma; Mitochondrial pathway; AKT AB Arsenic trioxide (As2O3), an ingredient in many traditional Chinese medicines, has drawn broad attention due to its therapeutic effects on a variety of cancers, including some solid tumors. However, the effects of As2O3 on cholangiocarcinoma have not been reported. In the present study, we demonstrate for the first time that clinically obtainable concentrations of As2O3 inhibit cell growth and induce apoptosis in human cholangiocarcinoma SK-ChA-1 cells. As2O3-induced apoptosis was partially inhibited by caspase inhibitor and accompanied by changes in the expression of Bcl-2 family proteins, decrease of mitochondrial membrane potential (MMP), release of cytochrome C from mitochondria, activation of caspase-3, caspase-9, and cleavage of poly (ADP-ribose) polymerase (PARP). Thus As2O3 induces apoptosis in SK-ChA-1 cells via mitochondria-mediated, caspases-dependent pathways. As2O3 inhibition of Akt phosphorylation may contribute to As2O3-mediated cholangiocarcinoma cell growth inhibition and apoptosis induction. C1 [Zhong, Fei] Sun Yat-Sen University, Third Affiliated Hospital, Department of Medical Oncology, 510630 Guangzhou, 510630, China. [Zhang, Shineng] Sun Yat-Sen University, Second Affiliated Hospital, Department of GastroenterologyGuangzhou, 510120, China. [Shao, Chunkui] Sun Yat-Sen University, Third Affiliated Hospital, Department of PathologyGuangzhou, 510630, China. [Yang, Jing] Sun Yat-Sen University, Third Affiliated Hospital, VIP Health-care centerGuangzhou, 510630, China. [Wu, Xiangyuan] Sun Yat-Sen University, Third Affiliated Hospital, Department of Medical Oncology, 510630 Guangzhou, 510630, China. RP Wu, X (reprint author), Sun Yat-Sen University, Third Affiliated Hospital, Department of Medical Oncology, 510630 Guangzhou, China. EM Wuxyonco@yahoo.com.cn CR Welzel TM, McGlynn KA, Hsing AW et al, 2006, Impact of classification of hilar cholangiocarcinomas, Klatskin tumors, on the incidence of intra- and extrahepatic cholangiocarcinoma in the United States. J Natl Cancer Inst 98:873–875 West J, Wood H, Logan RF et al, 2006, Trends in the incidence of primary liver and biliary tract cancers in England and Wales 1971–2001. Br J Cancer 94:1751–1758 Khan SA, Davidson BR, Goldin R et al, 2002, Guidelines for the diagnosis and treatment of cholangiocarcinoma: consensus document. Gut 51(S6):VI 1–9 Shaib Y, El-Serag HB, 2004, The epidemiology of cholangiocarcinoma. Semin Liver Dis 24:115–125 Anderson CD, Pinson CW, Berlin J et al, 2004, Diagnosis and treatment of cholangiocarcinoma. Oncologist 9:43–57 Zhang TC, Cao EH, Li JF et al, 1999, Induction of apoptosis and inhibition of human gastric cancer MGC-803 cell growth by arsenic trioxide. Eur J Cancer 35:1258–1263 Chou WC, Dang CV, 2005, Acute promyelocytic leukemia: recent advances in therapy and molecular basis of response to arsenic therapies. Curr Opin Hematol 12:1–6 Zhu J, Chen Z, Lallemand-Breitenbach V et al, 1999, How acute promyelocytic leukaemia revived arsenic. Nat Rev Cancer 2:705–713 Shen ZY, Shen WY, Chen MH et al, 2003, Reactive oxygen species and antioxidants in apoptosis of esophageal cancer cells induced by As2O3. Int J Mol Med 11:479–484 Uslu R, Sanli UA, Sezgin C et al, 2000, Arsenic trioxidemediated cytotoxicity and apoptosis in prostate and ovarian carcinoma cell lines. Clin Cancer Res 6:4957–4964 Zhang T, Wang SS, Hong L et al, 2003, Arsenic trioxide induces apoptosis of rat hepatocellular carcinoma cells in vivo. J Exp Clin Cancer Res 22:61–68 Jin HO, Yoon SI, Seo SK et al, 2006, Synergistic induction of apoptosis by sulindac and arsenic trioxide in human lung cancer A549 cells via reactive oxygen species-dependent downregulation of surviving. Biochem Pharmacol 72:1228–1236 Shao QS, Ye ZY, Ling ZQ et al, 2005, Cell cycle arrest and apoptotic cell death in cultured human gastric carcinoma cells mediated by arsenic trioxide. World J Gastroenterol 11:3451–3456 Hoorens A, Van de Casteele M, Kloppel G et al, 1996, Glucose promotes survival of rat pancreatic beta cells by activating synthesis of proteins which suppress a constitutive apoptotic program. J Clin Invest 98:1568–1574 Kachadourian R, Day BJ, 2006, Flavonoid-induced glutathione depletion: potential implications for cancer treatment. Free Radic Biol Med 41:65–76 Lecoeur H, Prevost MC, Gougeon ML, 2001, Oncosis is associated with exposure of phosphatidylserine residues on the outside layer of the plasma membrane: a reconsideration of the specificity of the annexin V/propidium iodide assay. Cytometry 44:65–72 Kroemer G, Galluzzi L, Vandenabeele P et al, 2009, Classification of cell death: recommendations of the nomenclature committee on cell death 2009. Cell Death Differ 16:3–11 Wang J, Ito T, Ukada N et al, 2006, PI3K-AKT pathway mediates growth and survival signals during development of fetal mouse lung. Tissue Cell 37:25–35 Shen ZX, Chen GQ, Ni JH et al, 1997, Use of arsenic trioxide, As2O3, in the treatment of acute promyelocytic leukemia, APL): II. Clinical efficacy and pharmacokinetics in relapsed patients. Blood 89:3354–3360 Gazitt Y, Akay C, 2005, Arsenic trioxide: an anti cancer missile with multiple warheads. Hematology 10:205–213 Reed JC, 2001, Apoptosis-regulating proteins as targets for drug discovery. Trends Mol Med 7:314–319 Desagher S, Martinou JC, 2000, Mitochondria as the central control point of apoptosis. Trends Cell Biol 10:369–377 van Loo G, Saelens X, van Gurp M et al, 2002, The role of mitochondrial factors in apoptosis: a Russian roulette with more than one bullet. Cell Death Differ 9:1031–1042 Thornberry NA, Lazebnik Y, 1998, Caspases: enemies within. Science 281:1312–1316 Antonsson B, 2004, Mitochondria and the Bcl-2 family proteins in apoptosis signaling pathways. Mol Cell Biochem 256– 257:141–155 Susin SA, Lorenzo HK, Zamzami N et al, 1999, Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 397:441–446 McCafferty-Grad J, Bahlis NJ, Krett N et al, 2003, Arsenic trioxide uses caspase-dependent and caspase-independent death pathways in myeloma cells. Mol Cancer Ther 2:1155–1164 Park MT, Kang YH, Park IC et al, 2007, Combination treatment with arsenic trioxide and phytosphingosine enhances apoptotic cell death in arsenic trioxide-resistant cancer cells. Mol Cancer Ther 6:82–92 Mahieux R, Pise-Masison C, Gessain A et al, 2001, Arsenic trioxide induces apoptosis in human T-cell leukemia virus type 1- and type 2-infected cells by a caspase-3-dependent mechanism involving Bcl-2 cleavage. Blood 98:3762–3769 Platanias LC, 2009, Biological responses to arsenic compounds. J Biol Chem 284:18583–18587 Antonsson B, 2004, Mitochondria and the Bcl-2 family proteins in apoptosis signaling pathways. Mol Cell Biochem 256– 257:141–155 Tsujimoto Y, 2002, Bcl-2 family of proteins: life-or-death switch in mitochondria. Biosci Rep 22:47–58 Vivanco I, Sawyers CL, 2002, The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer 2:489–501 Testa JR, Bellacosa A, 2001, AKT plays a central role in tumorigenesis. Proc Natl Acad Sci USA 98:10983–10985 Lawlor MA, Alessi DR, 2001, PKB/Akt: a key mediator of cell proliferation, survival and insulin responses? J Cell Sci 114:2903– 2910 Wu T, Leng J, Han C et al, 2004, The cyclooxygenase-2 inhibitor celecoxib blocks phosphorylation of Akt and induces apoptosis in human cholangiocarcinoma cells. Mol Cancer Ther 3:299–307 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 413 EP 420 DI 10.1007/s12253-009-9234-1 PG 8 ER PT J AU Becsagh, P Varga, K Szakacs, O Kopper, L Orosz, Zs AF Becsagh, Peter Varga, Katalin Szakacs, Orsolya Kopper, Laszlo Orosz, Zsolt TI High Resolution Melting Curve Analysis of DNA Sequence Alterations of Various Sizes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Melting curve; High resolution; DNA; Mutations; GIST ID Melting curve; High resolution; DNA; Mutations; GIST AB In the diagnostic workflow we need to think in algorithms, containing more assays. One of the most important task in the management of cancer patient is to detect nucleic acid sequence changes in clinical specimens. Before using the most expensive method to analyze direct our targets, a screening assay is needed to reduce the number of samples. In the detection of gene-sequence alterations classical screening methods are available, as SSCP, DGGE or TGGE, (Finke Exp Clin Endocrinol Diabetes 104:92–97, 1996; Lessa and Applebaum Mol Ecol 2:119–129, 1993) however these are very time consuming processes. At this time in the molecular lab the real-time PCR equipments are very popular and with the function of melting curve analysis it can be a very convenient, simple and cost-efficient screening method. C1 [Becsagh, Peter] Roche Hungary LtdBudaors, Hungary. [Varga, Katalin] National Institute of OncologyBudapest, Hungary. [Szakacs, Orsolya] MMM LtdBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Orosz, Zsolt] National Institute of OncologyBudapest, Hungary. RP Becsagh, P (reprint author), Roche Hungary Ltd, Budaors, Hungary. EM peter.becsagh@roche.com CR Finke R, 1996, Theoretical basis and application of molecular diagnostics. Exp Clin Endocrinol Diabetes 92(Suppl 4):92–97, Review Lessa EP, Applebaum G, 1993, Screening techniques for detecting allelic variation in DNA sequences. Mol Ecol 2, 2):119–129, Review Wittwer CT, Reed GH, Gundry CN, Vandersteen JG, Pryor RJ, 2003, High resolution genotyping by amplicon melting analysis using LCGreen. Clin Chem 49:853–860 Herrmann MG, Durtschi JD, Bromley LK,Wittwer CT, Voelkerding KV, 2006, DNA melting analysis for mutation scanning and genotyping: a cross platform comparison. Clin Chem 52:494–503 Seipp MT, Durtschi JD, Voelkerding KV, Wittwer CT, 2009, Multiplex amplicon genotyping by high-resolution melting. J Biomol Tech 20:160–164 Wittwer CT, 2009, High-resolution DNA melting analysis: advancements and limitations. Hum Mutat 30:857–859 Crews N, Wittwer CT, Montgomery J et al, 2009, Spatial DNA melting analysis for genotyping and variant scanning. Anal Chem 81:2053–2058 Willmore-Payne C, Holden JA, Wittwer CT, Layfield LJ, 2009, The use of EGFR exon 19 and 21 unlabeled DNA probes to screen for activating mutations in non-small cell lung cancer. J Biomol Tech 19:217–224 Antonescu CR, Sommer G, Sarran L, Tschernyavsky SJ, Riedel E, Woodruff JM, Robson M, Maki R, Brennan MF, Ladanyi M, DeMatteo RP, Besmer P, 2003, Association of KIT exon 9 mutations with nongastric primary site and aggressive behavior: KIT mutation analysis and clinical correlates of 120 gastrointestinal stromal tumors. Clin Cancer Res 9(9):3329–3337 Talaulikar D, Gray JX, Shadbolt B, McNiven M, Dahlstrom JE, 2008, A comparative study of the quality of DNA obtained from fresh frozen and formalin-fixed decalcified paraffin-embedded bone marrow trephine biopsy specimens using two different methods. J Clin Pathol 61(1):119–123, Epub 2007 Jun Erali M, Voelkerding KV, Wittwer CT, 2008, High resolution melting applications for clinical laboratory medicine. Exp Mol Pathol 85:50–58 Erali M, Wittwer CT, 2010, High resolution melting analysis for gene scanning. Methods, article in press., DOI 10.1016/j. ymeth.2010.01.013 Korbie DJ, Mattick JS, 2008, Touchdown PCR for increased specificity and sensitivity in PCR amplification. Nat Protoc 3, 9):1452–1456 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 421 EP 426 DI 10.1007/s12253-010-9274-6 PG 6 ER PT J AU Adly, AM Hussein, RAM AF Adly, A Mohamed Hussein, Rezk Abdelwahed Mahmoud TI Immunohistological Profile of the Ras Homologous B Protein (RhoB) in Human Testes Showing Normal Spermatogenesis, Spermatogenic Arrest and Sertoli Cell Only Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE RhoB; Human testis; Fertility; Infertility; Ras ID RhoB; Human testis; Fertility; Infertility; Ras AB Ras homologous B protein (RhoB) belongs to the Ras homologous subfamily which consists of low molecular weight (21 kDa) GTP-binding proteins. Rho proteins are regulatory molecules associated with various kinases and as such they mediate changes in cell shape, contractility, motility and gene expression. To date, no data are available about the expression pattern of RhoB protein in the human testis showing normal and abnormal spermatogenesis. The present study addresses these issues. Human testicular biopsy specimens were obtained from patients suffering from post-testicular infertility (testis showing normal spermatogenesis, 10 cases) and testicular infertility (testis showing Sertoli cell only syndrome and spermatogenic arrest, 10 patients each). The expression of RhoB was examined using in situ immunofluorescent staining methods. In testes showing normal spermatogenesis, RhoB had a strong expression in the seminiferous epithelium (cytoplasm of Sertoli-cells, spermatogonia and spermatocytes) and in the interstitium (Leydig cells). RhoB expression was weak in the myofibroblasts and absent in the spermatids and sperms. In the testes showing abnormal spermatogenesis, RhoB expression wasmoderate in the seminiferous epithelium (cytoplasm of Sertoli cells, spermatogonia and spermatocytes) and was completely absent in the Leydig cells, myofibroblasts, spermatids and sperms. To the best of our knowledge, this study provides the first morphological indication that RhoB protein is expressed in human testis and its expression undergoes testicular infertility associated changes. These findings suggest the involvement of RhoB in the process of spermatogenesis in human and their possible therapeutic ramifications in testicular infertility are open for further investigations. C1 [Adly, A Mohamed] Sohag University, Faculty of Science, Department of ZoologySohag, Egypt. [Hussein, Rezk Abdelwahed Mahmoud] King Khalid University, Assir Central Hospital, Department of PathologyAbha, Saudi Arabia. RP Hussein, RAM (reprint author), King Khalid University, Assir Central Hospital, Department of Pathology, Abha, Saudi Arabia. EM mrcpath17@gmail.com;mrh17@gawab.com CR Hu E, 2006, Recent patents on Rho signaling pathway as therapeutic target for cardiovascular diseases. Recent Patents Cardiovasc Drug Discov 1:249–263 Berken A, Wittinghofer A, 2008, Structure and function of Rhotype molecular switches in plants. Plant Physiol Biochem 46:380– 393 Nowak JM, Grzanka A, Zuryn A, Stepien A, 2008, The Rho protein family and its role in the cellular cytoskeleton. Postepy Hig Med Dosw, Online, 62:110–117 Modarressi MH, Cheng M, Tarnasky HA, Lamarche-Vane N, de Rooij DG, Ruan Y, van der Hoorn FA, 2004, A novel testicular RhoGAP-domain protein induces apoptosis. Biol Reprod 71:1980–1990 Genth H, Dreger SC, Huelsenbeck J, Just I, 2008, Clostridium difficile toxins: more than mere inhibitors of Rho proteins. Int J Biochem Cell Biol 40:592–597 Nakakuki T, Ito M, Iwasaki H, Kureishi Y, Okamoto R, Moriki N, Kongo M, Kato S, Yamada N, Isaka N, Nakano T, 2005, Rho/ Rho-kinase pathway contributes to C-reactive protein-induced plasminogen activator inhibitor-1 expression in endothelial cells. Arterioscler Thromb Vasc Biol 25:2088–2093 Sequeira L, Dubyk CW, Riesenberger TA, Cooper CR, van Golen KL, 2008, Rho GTPases in PC-3 prostate cancer cell morphology, invasion and tumor cell diapedesis. Clin Exp Metastasis 25:569– 579 Zhao L, Wang H, Li J, Liu Y, Ding Y, 2008, Overexpression of Rho GDP-dissociation inhibitor Alpha is associated with tumor progression and poor prognosis of colorectal cancer. J Proteome Res 7(9):3994–4003 Scott RW, Olson MF, 2007, LIM kinases: function, regulation and association with human disease. J Mol Med 85:555–568 Ramos S, Khademi F, Somesh BP, Rivero F, 2002, Genomic organization and expression profile of the small GTPases of the RhoBTB family in human and mouse. Gene 298:147–157 Liu GY, Gao SZ, Ge CR, Zhang X, 2008, Cloning, nucleotide sequence and tissue expression profile of three novel porcine genes—RHOB, RHOG and PRAF1. Mol Biol, Mosk, 42:59– 62 Kao TJ, Millette CF, 2007, L-type voltage-operated Ca(+2, channels modulate transient Ca(+2, influx triggered by activation of Sertoli cell surface L-selectin. J Cell Biochem 101:1023–1037 Lui WY, Lee WM, Cheng CY, 2003, Sertoli-germ cell adherens junction dynamics in the testis are regulated by RhoB GTPase via the ROCK/LIMK signaling pathway. Biol Reprod 68:2189–2206 Lui WY, Lee WM, Cheng CY, 2003, Rho GTPases and spermatogenesis. Biochim Biophys Acta 1593:121–129 Lui WY, Mruk DD, Cheng CY, 2005, Interactions among IQGAP1, Cdc42, and the cadherin/catenin protein complex regulate Sertoli-germ cell adherens junction dynamics in the testis. J Cell Physiol 202:49–66 Shim H, Lee H, Jeoung D, 2006, Cancer/testis antigen cancerassociated gene, CAGE, promotes motility of cancer cells through activation of focal adhesion kinase, FAK). Biotechnol Lett 28:2057–2063 Kamai T, Arai K, Tsujii T, Honda M, Yoshida K, 2001, Overexpression of RhoA mRNA is associated with advanced stage in testicular germ cell tumour. BJU Int 87:227–231 Hussein MR, Abou-Deif ES, Bedaiwy MA, Said TM, Mustafa MG, Nada E, Ezat A, Agarwal A, 2005, Phenotypic characterization of the immune and mast cell infiltrates in the human testis shows normal and abnormal spermatogenesis. Fertil Steril 83:1447–1453 Hussein MR, Abu-Dief EE, Abou El-Ghait AT, Adly MA, Abdelraheem MH, 2006, Melatonin and roentgen irradiation of the testis. Fertil Steril 86:750–752 Hussein MR, Abu-Dief EE, Abou El-Ghait AT, Adly MA, Abdelraheem MH, 2006, Morphological evaluation of the radioprotective effects of melatonin against X-ray-induced early and acute testis damage in Albino rats: an animal model. Int J Exp Pathol 87:237–250 Hirano K, Matsuura F, Tsukamoto K, Zhang Z, Matsuyama A, Takaishi K, Komuro R, Suehiro T, Yamashita S, Takai Y, Matsuzawa Y, 2000, Decreased expression of a member of the Rho GTPase family, Cdc42Hs, in cells from Tangier disease—the small G protein may play a role in cholesterol efflux. FEBS Lett 484:275–279 Freeman EA, Jani P, Millette CE, 2002, Expression and potential function of Rho family small G proteins in cells of the mammalian seminiferous epithelium. Cell Commun Adhes 9:189–204 Nakamura K, Fujita A, Murata T, Watanabe G, Mori C, Fujita J, Watanabe N, Ishizaki T, Yoshida O, Narumiya S, 1999, Rhophilin, a small GTPase Rho-binding protein, is abundantly expressed in the mouse testis and localized in the principal piece of the sperm tail. FEBS Lett 445:9–13 Takahashi H, Koshimizu U, Miyazaki J, Nakamura T, 2002, Impaired spermatogenic ability of testicular germ cells in mice deficient in the LIM-kinase 2 gene. Dev Biol 241:259–272 Cunto FD, Imarisio S, Camera P, Boitani C, Altruda F, Silengo L, 2002, Essential role of citron kinase in cytokinesis of spermatogenic precursors. J Cell Sci 115:4819–4826 Togawa A, Miyoshi J, Ishizaki H, Tanaka M, Takakura A, Nishioka H, Yoshida H, Doi T, Mizoguchi A, Matsuura N, Niho Y, Nishimune Y, Nishikawa S, Takai Y, 1999, Progressive impairment of kidneys and reproductive organs in mice lacking Rho GDIalpha. Oncogene 18:5373–5380 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 427 EP 433 DI 10.1007/s12253-009-9232-3 PG 7 ER PT J AU Kadota, K Kushida, Y Miyai, Y Katsuki, N Hayashi, T Bando, K Shibuya, Sh Haba, R AF Kadota, Kyuichi Kushida, Yoshio Miyai, Yumi Katsuki, Naomi Hayashi, Toshitetsu Bando, Kenji Shibuya, Shinsuke Haba, Reiji TI Epidermoid Cyst in an Intrapancreatic Accessory Spleen: Three Case Reports and Review of the Literatures SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Epidermoid cyst; Pancreas; Accessory spleen; Carbohydrate antigen 19-9; Immunohistochemistry; Histogenesis ID Epidermoid cyst; Pancreas; Accessory spleen; Carbohydrate antigen 19-9; Immunohistochemistry; Histogenesis AB The development of an epidermoid cyst in an intrapancreatic accessory spleen is an extremely rare lesion, with only 17 cases being reported in the English literature. All such cases were located in the pancreatic tail, some of which showed carbohydrate antigen 19-9 (CA19-9) immunoreactivity in the lining of the epithelium. A few of themindicated an elevation of the serum CA19-9 level. Here we report three cases of an epidermoid cyst in an intrapancreatic accessory spleen. Cases 1 and 2 were 57-year-old and 70-year-old women, while case 3 was a 37-year-old man. All three cases were asymptomatic. Serum CA19-9 levels showed within normal limits (case 1), slightly elevated (case 2), and clearly elevated (case 3). They underwent a distal pancreatectomy with splenectomy (cases 1 and 2) and without splenectomy (case 3). Grossly, the surgical specimen was a welldemarcated, multiple (case 1) or solitary (cases 2 and 3) cystic mass in the pancreatic tail. A high level of fluid CA 19-9 was detected in case 1. Microscopically, the cystic walls were lined with squamous and cuboidal epithelium, which were surrounded by normal splenic tissue and hyalinized fibrous tissue. The lining squamous epithelium was revealed as nonkeratinizing (Cases 1and 2) or keratinizing (Case 3). Immunohistochemically, CA19-9 was positive in the monolayer and surface layer of the cuboidal epithelium, but negative for the keratinizing squamous epithelium. As for the histogenesis, it is suggested that the cystic lining of the epithelium may derive from the pancreatic duct which protrudes into the accessory spleen. C1 [Kadota, Kyuichi] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Kushida, Yoshio] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Miyai, Yumi] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Katsuki, Naomi] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Hayashi, Toshitetsu] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Bando, Kenji] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Shibuya, Shinsuke] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. [Haba, Reiji] Kagawa University, University Hospital, Department of Diagnostic Pathology, 1750-1, Ikenobe, Miki-cho, 761-0793 Kagawa, Japan. RP Kadota, K (reprint author), Kagawa University, University Hospital, Department of Diagnostic Pathology, 761-0793 Kagawa, Japan. EM qichi@med.kagawa-u.ac.jp CR Davidson ED, Campbell WG, Hersh T, 1980, Epidermoid splenic cyst occurring in an intrapancreatic accessory spleen. Dig Dis Sci 25:964–967 Morohoshi T, Hamamoto T, Kunimura T et al, 1991, Epidermoid cyst derived from an accessory spleen in the pancreas. A case report with literature survey. Acta Pathol Jpn 41:916–921 Nakae Y, Hayakawa T, Kondo T et al, 1991, Epidermoid cyst occurring in a pancreatic accessory spleen. J Clin Gastroenterol 13:362–364 Tang X, Tanaka Y, Tsutsumi Y, 1994, Epithelial inclusion cysts in an intrapancreatic accessory spleen. Pathol Int 44:652–654 Furukawa H, Kosuge T, Kanai Y, Mukai K, 1998, Epidermoid cyst in an intrapancreatic accessory spleen: CT and pathologic findings. AJR Am J Roentgenol 171:271 Higaki K, Jimi A, Watanabe J et al, 1998, Epidermoid cyst of the spleen with CA19, 9 or carcinoembryonic antigen productions: report of three cases. Am J Surg Pathol 22:704–708 Tateyama H, Tada T, Murase T et al, 1998, Lymphoepithelial cyst and epidermoid cyst of the accessory spleen in the pancreas. Mod Pathol 11:1171–1177 Sasou S, Nakamura S, Inomata M, 1999, Epithelial splenic cysts in an intrapancreatic accessory spleen and spleen. Pathol Int 49:1078–1083 Choi SK, Ahn SI, Hong KC et al, 2000, A case of epidermoid cyst of the intrapancreatic accessory spleen. J Korean Med Sci 15:589–592 Tsutsumi S, Kojima T, Fukai Y et al, 2000, Epidermoid cyst of an intrapancreatic accessory spleen, a case report. Hepatogastroenterology 47:1462–1464 Horibe Y, Murakami M, Yamao K et al, 2001, Epithelial inclusion cyst, epidermoid cyst, formation with epithelioid cell granuloma in an intrapancreatic accessory spleen. Pathol Int 51:50–54 Sonomura T, Kataoka S, Chikugo T et al, 2002, Epidermoid cyst originating from an intrapancreatic accessory spleen. Abdom Imaging 27:560–562 Kanazawa H, Kamiya J, Nagino M et al, 2004, Epidermoid cyst in an intrapancreatic accessory spleen: a case report. J Hepatobiliary Pancreat Surg 11:61–63 Ru K, Kalra A, Ucci A, 2007, Epidermoid cyst of intrapancreatic accessory spleen. Dig Dis Sci 52:1229–1232 Itano O, Shiraga N, Kouta E et al, 2008, Epidermoid cyst originating from an intrapancreatic accessory spleen. J Hepatobiliary Pancreat Surg 15:436–439 Servais EL, Sarkaria IS, Solomon GJ et al, 2008, Giant epidermoid cyst within an intrapancreatic accessory spleen mimicking a cystic neoplasm of the pancreas: case report and review of the literature. Pancreas 36:98–100 Gleeson FC, Kendrick ML, Chari ST et al, 2008, Epidermoid accessory splenic cyst masquerading as a pancreatic mucinous cystic neoplasm. Endoscopy 40:E141–2 Robbins FG, Yellin AE, Lingua RW et al, 1978, Splenic epidermoid cysts. Ann Surg 187:231–235 Halpert B, Alden ZA, 1964, Accessory spleens in or at the tail of the pancreas. A survey of 2, 700 additional necropsies. Arch Pathol 77:652–654 Burrig KF, 1988, Epithelial, true, splenic cysts. Pathogenesis of the mesothelial and so-called epidermoid cyst of the spleen. Am J Surg Pathol 12:275–81 Elit L, Aylward B, 1989, Splenic cyst carcinoma presenting in pregnancy. Am J Hematol 32:57–60 Schacht V, Dadras SS, Johnson LA et al, 2005, Up-regulation of the lymphatic marker podoplanin, a mucin-type transmembrane glycoprotein, in human squamous cell carcinomas and germ cell tumours. Am J Pathol 166:913–921 Ough YD, Nash HR, Wood DA, 1981, Mesothelial cysts of the spleen with squamous metaplasia. Am J Clin Pathol 76:666–669 Lifschitz-Mercer B, Open M, Kushnir I, Czernobilsky B, 1994, Epidermoid cyst of the spleen: a cytokeratin profile with comparison to other squamous epithelia. Virchows Arch 424:213–216 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 435 EP 442 DI 10.1007/s12253-009-9229-y PG 8 ER PT J AU Svec, A Bury, Y AF Svec, Alexandr Bury, Yvonne TI Haemangioma of the Parathyroid Gland. Does it Really Exist? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Parathyroid gland; Chromogranin A; Haemangioma; VEGF; Tumour-associated vascular proliferations ID Parathyroid gland; Chromogranin A; Haemangioma; VEGF; Tumour-associated vascular proliferations AB We are reporting a case of a capillary haemangiomalike proliferation arising within a parathyroid gland adenoma, associated with primary hyperparathyroidism. The vessel proliferation bearing a close resemblance to a capillary haemangioma consisted of tightly packed capillaries, endothelial buds and occasional small caliber muscle-containing vessels. The observation expands the spectrum of tumourassociated vascular proliferations by adding an exuberant haemangioma-like pattern to its extreme end. These are a heterogeneous group of lesions reportedly induced by aberrant production of angiogenic factors. We investigated expression of VEGF, pKDR, FGF2, HIF1α and HIF2α and only VEGF gave a strong positive reaction in the adenoma cells entrapped in the vascular meshwork. Although this does not constitute a proof that aberrant VEGF production was a causative agent, unexpected supportive evidence for its pathogenic role emerged from a failure to detect chromogranin A. Chromogranin A is a precursor of several regulatory proteins, including vasostatin I, a multilevel suppressor of VEGF. The production of vasostatin I may have been reduced in a chromogranin A-negative adenoma which could lead to a loss of its opposing effect on VEGF–regulated processes. The only two other published cases of haemangioma of the parathyroid gland were reported in patients diagnosed with primary parathyroid hyperplasia with hyperparathyroidism, a pathophysiologic condition similar to our case. Therefore we raise the question whether these tumours could also represent a reactive phenomenon. C1 [Svec, Alexandr] James Cook University Hospital, Department of Cellular Pathology, Marton Road, TS4 3BW Middlesbrough, UK. [Bury, Yvonne] James Cook University Hospital, Department of Cellular Pathology, Marton Road, TS4 3BW Middlesbrough, UK. RP Svec, A (reprint author), James Cook University Hospital, Department of Cellular Pathology, TS4 3BW Middlesbrough, UK. EM alexandr.svec@vfn.cz CR Chan JKC, 2007, Tumours of the thyroid and parathyroid glands. Parathyroid glands. In: Fletcher CDM, ed, Diagnostic histopathology of tumours, 3rd edn. Churchill Livingstone, London Rosai J, 2004, Parathyroid glands. In: Rosai J, ed, Rosai and Ackerman's surgical pathology, 9th edn. Mosby, St Louis DeLellis RA, Williams ED, 2004, Tumours of the thyroid and parathyroid. In: DeLellis RA, Lloyd RV, Heitz PU, Eng Ch, eds, Pathology and genetics of tumours of endocrine organs. IARC, Lyon Garcia de la Torre N, Buley I, Wass JAH et al, 2004, Angiogenesis and lymphangiogenesis in parathyroid proliferative lesions. J Clin Endocrinol Metab 89:2890–2896 Merino M, Chuaqui R, Fernandez P, 1996, Parathyroid haemangioma: a report of two cases. Endocr Pathol 7:319–322 Gaudin P, Rosai J, 1995, Florid vascular proliferation associated with neural and neuroendocrine neoplasms. A diagnostic clue and potential pitfall. Am J Surg Pathol 19:642–652 Baloch ZW, LiVolsi VA, 1998, Intravascular Kaposi's-like spindle cell proliferation of the capsular vessels of follicular-derived thyroid carcinomas. Mod Pathol 11:995–998 Tse LL, Chan I, Chan JK, 2001, Capsular intravascular endothelial hyperplasia: a peculiar form of vasoproliferative lesion associated with thyroid carcinoma. Histopathology 39:463–468 Vodovnik A, 2002, Capsular vascular proliferation associated with thyroid paraganglioma. Histopathology 41:273 Kuhn E, De Anda J, Manoni S et al, 2006, Renal cell carcinoma associated with prominent angioleiomyoma-like proliferation: report of 5 cases and review of the literature. Am J Surg Pathol 30:1372–1381 Carter W, Uy K, Ward M et al, 2000, Parathyroid-induced angiogenesis is VEGF-dependent. Surgery 128:458–464 Lazaris AC, Tseleni-Balafouta S, Papathomas T et al, 2006, Immunohistochemical investigation of angiogenic factors in parathyroid proliferative lesions. Eur J Endocrinol 154:827–833 Martins P, Schmitt F, Almeida H et al, 2008, Evaluation of parathyroid gland angiogenesis in chronic kidney disease associated with secondary hyperparathyroidism. Nephrol Dial Transplant 23:2889–2894 Rashid G, Bernheim J, Green J et al, 2008, Parathyroid hormone stimulates the endothelial expression of vascular endothelial growth factor. Eur J Clin Invest 38:798–803 Viacava P, Bocci G, Fanelli G et al, 2006, Microvessel density in human normal and neoplastic parathyroids. Endocr Pathol 17:175–181 Belloni D, Scabini S, Foglieni C et al, 2007, The vasostatin-I fragment of chromogranin A inhibits VEGF-induced endothelial cell proliferation and migration. FASEB J 21:3052–62 Gould VE, Wiedenmann B, Lee I et al, 1987, Synaptophysin expression in neuroendocrine neoplasms as determined by immunocytochemistry. Am J Pathol 126:243–57 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 443 EP 446 DI 10.1007/s12253-009-9236-z PG 4 ER PT J AU Matsuda, I Imai, Y Hirota, S AF Matsuda, Ikuo Imai, Yukihiro Hirota, Seiichi TI Global Histone Modification Profiles are Well Conserved Between Normal B Lymphocytes and Neoplastic Counterparts SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Diagnostic histopathology; Tumor; Cell type; Global histone modification; Immunohistochemistry; Reactive lymphoid hyperplasia; Germinal center B cell; Mantle zone B cells; Follicular lymphoma; Mantle cell lymphoma ID Diagnostic histopathology; Tumor; Cell type; Global histone modification; Immunohistochemistry; Reactive lymphoid hyperplasia; Germinal center B cell; Mantle zone B cells; Follicular lymphoma; Mantle cell lymphoma AB The identification of cell type is essential in diagnostic tumor histopathology. We hypothesized that some patterns of global histone modification are specific to both particular cell types of non-neoplastic tissues and their neoplastic counterparts. To examine the hypothesis in lymphoid cells, global histone modification patterns of germinal center and mantle zone B cells in reactive lymphoid hyperplasia (RLH) were compared with those of follicular lymphoma (FL) and mantle cell lymphoma (MTL) cells by immunohistochemistry. We revealed that FL cells and MTL cells exhibited the similar histone modification pattern to that of germinal center B lymphocytes and mantle zone B lymphocytes in RLH, respectively. These results indicate that global histone modification profiles specific to non-neoplastic germinal center B lymphocytes and mantle zone B lymphocytes are well conserved in corresponding neoplastic lymphoma cells, and suggest that they will be indicative of tumor cell type at least in B cell lymphoma. C1 [Matsuda, Ikuo] Hyogo College of Medicine, Department of Surgical Pathology, 1-1 Mukogawa-cho, 663-8501 Nishinomiya, Hyogo, Japan. [Imai, Yukihiro] Kobe City Medical Center General Hospital, Department of PathologyKobe, Hyogo, Japan. [Hirota, Seiichi] Hyogo College of Medicine, Department of Surgical Pathology, 1-1 Mukogawa-cho, 663-8501 Nishinomiya, Hyogo, Japan. RP Hirota, S (reprint author), Hyogo College of Medicine, Department of Surgical Pathology, 663-8501 Nishinomiya, Japan. EM hiros@hyo-med.ac.jp CR Jones PA, Baylin SB, 2007, The epigenetics of cancer. Cell 128:683–692 Swerdlow SH, Campo E, Harris NL et al, eds,, 2008, WHO classification of tumours of haematopoietic and lymphoid tissues, 4th edn. WHO, Geneva Seligson DB, Horvath S, Shi T et al, 2005, Global histone modification patterns predict risk of prostate cancer recurrence. Nature 435:1262–1266 Barlesi F, Giaccone G, Gallegos-Ruiz MI et al, 2007, Global histone modifications predict prognosis of resected non-small-cell lung cancer. J Clin Oncol 25:4358–4364 Tzao C, Tung HJ, Jin JS et al, 2009, Prognostic significance of global histone modifications in resected squamous cell carcinoma of the esophagus. Mod Pathol 22:252–260 Krivtsov AV, Armstrong SA, 2007, MLL translocations, histone modifications and leukemia stem-cell development. Nat Rev Cancer 7:823–833 Pal S, Balocchi RA, Byrd JC et al, 2007, Low levels of miR-92b/ 96 induce PRMT5 translation and H3R8/H4R3 methylation in mantle cell lymphoma. EMBO J 26:3558–3569 Seligson DB, Horvath S, McBrian MA et al, 2009, Global levels of histone modifications predict prognosis in different cancers. Am J Pathol 174:1619–1628 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 447 EP 451 DI 10.1007/s12253-009-9224-3 PG 5 ER PT J AU Bardi, E Dobos, Kappelmayer, J Kiss, Cs AF Bardi, Edit Dobos, Eva Kappelmayer, Janos Kiss, Csongor TI Differential Effect of Corticosteroids on Serum Cystatin C in Thrombocytopenic Purpura and Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute lymphoblastic leukemia; Coricosteroid therapy; Cystatin C; Glomerular function; Idiopathic thrombocytopenic purpura; Leukemic cell burden ID Acute lymphoblastic leukemia; Coricosteroid therapy; Cystatin C; Glomerular function; Idiopathic thrombocytopenic purpura; Leukemic cell burden AB The aim of our study was to evaluate the influence of steroid therapy on serum cystatin C (cysC) concentrations in patients with acute lymphoblastic leukemia (ALL) and idiopthias thrombocytopenias purpura (ITP). We studied 17 patients with ITP (girls: boys=5:12, mean age: 7.6 yrs, range between 1 to 17 years) and 18 patients with ALL (girls: boys=6:12, mean age: 6.3 yrs, range between 2 to 17 years). CysC and white blood cell count (WBC) in both group of patients were determined before and after 300 mg/m2 cumulative dose of steroid therapy. Corticosteroids increased the level of cysC in both groups of patients, however significant increase was found only in ITP patients between pre- and posttreatment values (0,96±0,27 mg/L vs. 1,16±0,3 mg/L, p=0,02). Pretreatment cysC concentrations were within the reference range in patients with ITP but not with ALL and were significantly higher in ALL patients, than in ITP patients (1,23±1,12 mg/L vs. 0,96±0,27 mg/L, p=0,02). Pretreatment WBC of ALL patients were significantly higher than of ITP patients (22,58 G/L, min. 3,5 G/L, max. 102,1 G/L vs. 7,46 G/L, min. 4,8 G/L, max. 12,3 G/L, p=0.03). We have found significant correlation between pretreatment cysC and WBC values in ALL patients (p=0.04). Although the concentration of cysC may be slightly and reversibly influenced by corticosteroid treatment, cysC is sensitive to detect early and moderate deteoriantation of GFR in children with cancer. C1 [Bardi, Edit] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-Oncology, 98 Nagyerdei Circle, 4032 Debrecen, Hungary. [Dobos, Eva] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-Oncology, 98 Nagyerdei Circle, 4032 Debrecen, Hungary. [Kappelmayer, Janos] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Kiss, Csongor] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-Oncology, 98 Nagyerdei Circle, 4032 Debrecen, Hungary. RP Bardi, E (reprint author), University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-Oncology, 4032 Debrecen, Hungary. EM editbardi@hotmail.com CR Bardi E, Bobok I, Olah VA, Olah E, Kappelmayer J, Kiss C, 2004, Cystatin C is a suitable marker of glomerular function in children with cancer. Pediatr Nephrol 10:1145–1147 Mussap M, Plebani M, 2004, Biochemistry and clinical role of human cystatin C. Crit Rev Clin Lab Sci 41:467–550 Risch L, Herklotz R, Blumberg A, Huber AR, 2001, Effects of glucocorticoid immunosuppression on serum cystatin c concentrations in renal transplant patients. Clin Chem 47:2055–2059 Demirtas S, Akan O, Can M, Elmali E, Akan H, 2006, Cystatin C can be affected by nonrenal factors: a preliminary study on leukemia. Clin Biochem 39:115–118 Cimerman N, Brguljan PM, Krasovec M, 2000, Serum cystatin C, a potent inhibitor of cysteine proteinases, is elevated in asthmatic patients. Clin Chim Acta 300:83–95 Bokenkamp A, van Wijk JAE, Lentze MJ, 2002, Effect of corticosteroid therapy on serum cystatin C and ß2-microglobulin concentrations. Clin Chem 48:1123–1126 Schrappe M, Reiter A, Zimmermann M et al, 2000, Long-term results of four consecutive trials in childhood ALL performed by the ALL-BFM study group from 1981 to 1995, Berlin-Frankfurt- Munster. Leukemia 14:2205–2222 Jaffe M, 1986, On the precipitation that picric acid produces in normal urine, and on a new reaction of creatinine [in German]. Zietschrift Phisologica Chemica 10:391–400 Counahan R, Chantler C, Ghazali S, Kirkwood B, Rose F, Barrat TM, 1976, Estimation of glomerular filtration rate from plasma creatinine concentration in children. Arch Dis Child 51:875–878 Bardi E, Olah VA, Bartyik K, Endreffy E, Jenei C, Kappelmayer J, Kiss C, 2004, Late effects on renal glomerular and tubular function of childhood cancer survivors. Pediatr Blood Cancer 43:668–673 Bjarnadottir M, Grubb A, Olafsson I, 1995, Promoter mediated dexamethasone-induced increase in cystatin C production in HeLa cels. Scand J Clin Lab Invest 55:617–623 Fricker M, Wiesli P, Brandle M, Schwegler B, Schmid C, 2003, Impact of thyroid dysfunction on serum cystatin c. Kidney Int 63:1944–1947 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 453 EP 456 DI 10.1007/s12253-009-9243-0 PG 4 ER PT J AU Bonavina, L Laface, L Picozzi, S Nencioni, M Siboni, S Bona, D Sironi, A Sorba, F Clemente, C AF Bonavina, Luigi Laface, Letizia Picozzi, Stefano Nencioni, Marco Siboni, Stefano Bona, Davide Sironi, Andrea Sorba, Francesca Clemente, Claudio TI Proposal of a Punch Biopsy Protocol as a Pre-requisite for the Establishment of a Tissue Bank from Resected Esophageal Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal carcinoma; Punch biopsy; Tumor bank; Tissue bank; Surgical oncology ID Esophageal carcinoma; Punch biopsy; Tumor bank; Tissue bank; Surgical oncology AB With the development of tissue banking, a need for homogeneous methods of collection, processing, and storage of tissue has emerged. We describe the implementation of a biological bank in a high-volume, tertiary care University referral center for esophageal cancer surgery.We also propose an original punch biopsy technique of the surgical specimen. The method proved to be simple, reproducible, and not expensive. Unified standards for specimen collection are necessary to improve results of specimen-based diagnostic testing and research in surgical oncology. C1 [Bonavina, Luigi] University of Milano Medical School, Division of General Surgery, IRCCS Policlinico San Donato, Department of Medical and Surgical SciencesMilan, Italy. [Laface, Letizia] University of Milano Medical School, Division of General Surgery, IRCCS Policlinico San Donato, Department of Medical and Surgical SciencesMilan, Italy. [Picozzi, Stefano] University of Milano Medical School, Division of General Surgery, IRCCS Policlinico San Donato, Department of Medical and Surgical SciencesMilan, Italy. [Nencioni, Marco] University of Milano Medical School, Division of General Surgery, IRCCS Policlinico San Donato, Department of Medical and Surgical SciencesMilan, Italy. [Siboni, Stefano] University of Milano Medical School, Division of General Surgery, IRCCS Policlinico San Donato, Department of Medical and Surgical SciencesMilan, Italy. [Bona, Davide] University of Milano Medical School, Division of General Surgery, IRCCS Policlinico San Donato, Department of Medical and Surgical SciencesMilan, Italy. [Sironi, Andrea] University of Milano Medical School, Division of General Surgery, IRCCS Policlinico San Donato, Department of Medical and Surgical SciencesMilan, Italy. [Sorba, Francesca] University of Milano Medical School, Division of General Surgery, IRCCS Policlinico San Donato, Department of Medical and Surgical SciencesMilan, Italy. [Clemente, Claudio] Istituto Clinico S. Ambrogio, Division of PathologyMilan, Italy. RP Bonavina, L (reprint author), University of Milano Medical School, Division of General Surgery, IRCCS Policlinico San Donato, Department of Medical and Surgical Sciences, Milan, Italy. EM luigi.bonavina@unimi.it CR Howe M, 2001, National prostate tumour bank launched in Australia. Lancet Oncol 2:656 Morrin H, Gunningham S, Currie M, Dachs G, Fox S, Robinson B, 2005, The Christchurch tissue bank to support cancer research. NZMJ 118:1–12 Teodorovic I, Therrasse P, Spatz A et al, 2003, Human tissue research: EORTC recommendations on its practical consequences. Eur J Cancer 39:2256–63 Leyland-Jones B, Ambrosone C, Bartlett J, Ellis M, Enos R, Raji A et al, 2008, Recommendations for collection and handling of specimens from group breast cancer clinical trials. J Clin Oncol 26:5638–44 Siewert R, Lordick F, Ott K, Stein H, Weber W, Becker K et al, 2007, Induction therapy in Barrett cancer. Influence on surgical risk and outcome. Ann Surg 246:624–631 Pennathur A, Luketich J, 2008, Resection for esophageal cancer: strategies for optimal management. Ann Thorac Surg 85:S751–756 Kandioler D, Hejna M, Zwrtek R et al, 2007, p53 adapted neoadjuvant therapy for esophageal cancer: pilot study. J Clin Oncol 25:201s, abstract 4535) Ota Y, Tagagi Y, Osaka Y, Shinohara M, Hoshino S, Tsuchida A et al, 2007, Usefulness of serum protein profiling for prediction of preoperative chemoradiosensitivity of esophageal cancer. Oncol Rep 18:653–657 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 457 EP 460 DI 10.1007/s12253-009-9244-z PG 4 ER PT J AU Perez, OL Barbisan, G Ottino, A Pianzola, H Golijow, DC AF Perez, Orlando Luis Barbisan, Gisela Ottino, Anabel Pianzola, Horacio Golijow, Daniel Carlos TI Human Papillomavirus DNA and Oncogene Alterations in Colorectal Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human Papillomavirus; Colorectal carcinoma; Viral integration; Proto-oncogene activation; k-ras; c-myc ID Human Papillomavirus; Colorectal carcinoma; Viral integration; Proto-oncogene activation; k-ras; c-myc AB The aim of the present study is to determine the presence and molecular integrity of high-risk HPV types in colorectal adenocarcinomas and to assess whether viral DNA is related to common proto-oncogene alterations, such as k-ras mutations and c-myc gene amplification, in colorectal cancer. Seventy-five colorectal adenocarcinomas were screened for HPV infection using nested-PCR (MY09/11-GP5+/6+). HPV typing was performed by type-specific PCR for HPV 16 and HPV 18 DNA. Unidentified samples were subsequently sequenced to determine the viral genotype. The physical status of HPV was determined by a nested PCR approach for type-specific E2 sequences. Cmyc amplification was assessed by co-amplification with β-globin as control locus, and mutation in k-ras codons 12 and 13 by ARMS-PCR. Overall, HPV was detected in thirty-three colorectal specimens (44%). HPV 16 was the prevalent type (16/75), followed by HPV 18 (15/75), HPV 31 (1/75) and HPV 66 (1/75). E2 disruption was detected in 56.3% of HPV 16 and in 40% of HPV 18 positive tumors. C-myc amplification was detected in 29.4% of cases, while k-ras mutations in 30.7%. There was no significant trend for HPV infection in tumors harboring either k-ras or c-myc alterations. This study demonstrates HPV DNA and viral integration in colorectal tumors, suggesting a potential role of this virus in colorectal carcinogenesis. There was no concurrence, however, of k-ras and c-myc activation with viral infection. C1 [Perez, Orlando Luis] University of La Plata, Faculty of Veterinary Genetics, IGEVET-CONICET (Institute of Veterinary Genetics-National Council of Scientific and Technical Investigations), 60 and 118 street, PC B1900 La Plata, Buenos Aires, Argentina. [Barbisan, Gisela] University of La Plata, Faculty of Veterinary Genetics, IGEVET-CONICET (Institute of Veterinary Genetics-National Council of Scientific and Technical Investigations), 60 and 118 street, PC B1900 La Plata, Buenos Aires, Argentina. [Ottino, Anabel] Interzonal Hospital of San Martin, Service of Pathology, 1791 1st street, PC B1900 La Plata, Buenos Aires, Argentina. [Pianzola, Horacio] Interzonal Hospital of San Martin, Service of Pathology, 1791 1st street, PC B1900 La Plata, Buenos Aires, Argentina. [Golijow, Daniel Carlos] University of La Plata, Faculty of Veterinary Genetics, IGEVET-CONICET (Institute of Veterinary Genetics-National Council of Scientific and Technical Investigations), 60 and 118 street, PC B1900 La Plata, Buenos Aires, Argentina. RP Perez, OL (reprint author), University of La Plata, Faculty of Veterinary Genetics, IGEVET-CONICET (Institute of Veterinary Genetics-National Council of Scientific and Technical Investigations), PC B1900 La Plata, Argentina. EM perezlo@gmail.com CR Ferlay J, Bray F, Pisani P et al, 2004, GLOBOCAN 2002: cancer incidence, mortality and prevalence worldwide, IARC Cancer- Base N° 5. IARC, Lyon Beart RW, 1995, Colon and rectum. In: Abeloff MD, Armitage JO, Allen SL, Niedehuber JE, eds, Clinical oncology. Churchill Livingstone, New York Gazelle GS, McMahon PM, Scholz FJ, 2001, Screening for colorectal cancer. Radiology 221:273–274 Vainio H, Miller AB, 2003, Primary and secondary prevention in colorectal cancer. Acta Oncol 42:809–815 Bognar G, Istvan G, Bereczky B et al, 2008, Detection of human papillomavirus type 16 in squamous cell carcinoma of the colon and its lymph node metastases with PCS and southern blot hybridization. Pathol Oncol Res 14:93–96 Bodaghi S, Yamanegi K, Xiao SY, 2005, Colorectal papillomavirus infection in patients with colorectal cancer. Clin Cancer Res 11:2862–2867 Yu HG, Shun LB, Luo HS et al, 2002, Deletion of the FHIT gene in human colorectal cancer is independent of high-risk HPV infection. Int J Colorectal Dis 17:396–401 Damin DC, Caetano MB, Rosito MA et al, 2007, Evidence for an association of human papillomavirus infection and colorectal cancer. Eur J Surg Oncol 33:569–574 Shah KV, Daniel RW, Simons JW et al, 1992, Investigation of colon cancers for human papillomavirus genomic sequences by polymerase chain reaction. J Surg Oncol 51:5–7 Shroyer KR, Kim JG, Manos MM et al, 1992, Papillomavirus found in anorectal squamous carcinoma, not in colon adenocarcinoma. Arch Surg 127:741–744 zur Hausen H, 1999, Viruses in human cancers. Eur J Cancer 35:1878–1885 Bosch FX, de Sanjose S, 2007, The epidemiology of human papillomavirus infection and cervical cancer. Dis Markers 23:213–227 Giuliano AR, Tortolero-Luna G, Ferrer E et al, 2008, Epidemiology of human papillomavirus infection in men, cancers other than cervical and benign conditions. Vaccine 26:17–28 Ferber MJ, Montoya DP, Yu C et al, 2003, Integrations of the hepatitis B virus, HBV, and human papillomavirus, HPV, into the human telomerase reverse transcriptase, hTERT, gene in liver and cervical cancers. Oncogene 22:3813–3820 Ferber MJ, Thorland EC, Brink AA et al, 2003, Preferential integration of human papillomavirus type 18 near the c-myc locus in cervical carcinoma. Oncogene 22:7233–7242 Chakrabarti O, Krishna S, 2003, Molecular interactions of ‘high risk’ human papillomaviruses E6 and E7 oncoproteins: implications for tumour progression. J Biosci 28:337–348 Pett MR, Pett MR, Alazawi WO et al, 2004, Acquisition of highlevel chromosomal instability is associated with integration of human papillomavirus type 16 in cervical keratinocytes. Cancer Res 64:1359–1368 Badaracco G, Venuti A, Sedati A et al, 2002, HPV16 and HPV18 in genital tumors: significantly different levels of viral integration and correlation to tumor invasiveness. J Med Virol 67:574–582 Thorland EC, Myers SL, Gostout BS, 2003, Common fragile sites are preferential targets for HPV16 integrations in cervical tumors. Oncogene 22:1225–1237 Ryan KM, Birnie GD, 1996, Myc oncogenes: the enigmatic family. Biochem J 314:713–721 Augenlicht LH, Wadler S, Corner G, 1997, Low-level c-myc amplification in human colonic carcinoma cell lines and tumors: a frequent, p53-independent mutation associated with improved outcome in a randomized multi-institutional trial. Cancer Res 57:1769–1775 Obara K, Yokoyama M, Asano G et al, 2001, Evaluation of myc and chromosome 8 copy number in colorectal cancer using interphase cytogenetics. Int J Oncol 18:233–239 Zaharieva B, Simon R, Ruiz C et al, 2005, High-throughput tissue microarray analysis of CMYC amplification in urinary bladder cancer. Int J Cancer 117:952–956 Cheng JY, Meng CL, Chao CF et al, 1991, Human papillomavirus type-related DNA and c-myc oncogene alterations in colon cancer cell lines. Dis Colon Rectum 34:469–474 Bos JL, 1989, ras oncogenes in human cancer: a review. Cancer Res 49:4682–4689 Vogelstein B, Fearon ER, Hamilton SR et al, 1988, Genetic alterations during colorectal-tumor development. N Engl J Med 319:525–532 Fearon ER, Vogelstein B, 1990, A genetic model for colorectal tumorigenesis. Cell 61:759–767 Buyru N, Tezol A, Dalay N, 2006, Coexistence of K-ras mutations and HPV infection in colon cancer. BMC Cancer 6:115 Miller SA, Dykes DD, Polesky HF, 1988, A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16:1215 Evander M, Edlund K, Boden E et al, 1992, Comparison of onestep and two-step polymerase chain reaction with degenerate general primers in a population-based studies of human papillomavirus infection in young Swedish women. J Clin Microbiol 30:987–992 de Roda Husman AM, Walboomers JM, van den Brule AJ et al, 1995, The use of general primers GP5 and GP6 elongated at their 3′ ends with adjacent highly conserved sequences improves human papillomavirus detection by PCR. J Gen Virol 76:1057– 1062 Lonn U, Lonn S, Nilsson B et al, 1995, Prognostic value of erb- B2 and myc amplification in breast cancer imprints. Cancer 75:2681–2687 Hatzaki A, Razi E, Anagnostopoulou K et al, 2001, A modified mutagenic PCR-RFLP method for K-ras codon 12 and 13 mutations detection in NSCLC patients. Mol Cell Probes 15:243–247 Ilyas M, Straub J, Tomlinson IP et al, 1999, Genetic pathways in colorectal and other cancers. Eur J Cancer 35:1986–2002 Perez LO, Abba MC, Laguens RM et al, 2005, Analysis of adenocarcinoma of the colon and rectum: detection of human papillomavirus, HPV, DNA by polymerase chain reaction. Colorectal Dis 7:492–495 Lee YM, Leu SY, Chiang H et al, 2001, Human papillomavirus type 18 in colorectal cancer. J Microbiol Immunol Infect 34:87– 91 Erisman MD, Rothberg PG, Diehl RE et al, 1985, Deregulation of c-myc gene expression in human colon carcinoma is not accompanied by amplification or rearrangement of the gene. Mol Cell Biol 5:1969–1976 He QJ, Zeng WF, Sham JS et al, 2003, Recurrent genetic alterations in 26 colorectal carcinomas and 21 adenomas from Chinese patients. Cancer Genet Cytogenet 144:112–118 Knosel T, Petersen S, Schwabe H et al, 2002, Incidence of chromosomal imbalances in advanced colorectal carcinomas and their metastases. Virchows Arch 440:187–194 Al-Kuraya K, Novotny H, Bavi P et al, 2007, HER2, TOP2A, CCND1, EGFR and C-MYC oncogene amplification in colorectal cancer. J Clin Pathol 60:768–772 He TC, Sparks AB, Rago C et al, 1998, Identification of c-MYC as a target of the APC pathway. Science 281:1509–1512 Herrick J, Conti C, Teissier S et al, 2005, Genomic organization of amplified MYC genes suggests distinct mechanisms of amplification in tumorigenesis. Cancer Res 65:1174–1179 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2010 VL 16 IS 3 BP 461 EP 468 DI 10.1007/s12253-010-9246-x PG 8 ER PT J AU Fauzee, JSN Pan, J Wang, Yl AF Fauzee, Jasmine Selimah Nilufer Pan, Juan Wang, Ya-lan TI PARP and PARG Inhibitors—New Therapeutic Targets in Cancer Treatment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PARP. PARG; PARP inhibitors; PARG inhibitors ID PARP. PARG; PARP inhibitors; PARG inhibitors AB Today, the number of cancer patients throughout the world is increasing alarmingly and as per the World Health Organisation (WHO) data and statistics the prediction for the year 2020 will be 15 million new cases as compared to only 10 million cases in year 2000 leaving us dumbfounded. A lot of effort has been put in by researchers and scientists over decades to find drugs helpful in the treatment of cancers for the benefit of patients—The latest being the Poly ADP-ribose polymerase (PARP) and the Poly ADP-ribose glycohydrolase (PARG) inhibitors. This review highlights their mechanism of action under the rationale of their use and current development in the field of cancer. C1 [Fauzee, Jasmine Selimah Nilufer] Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. [Pan, Juan] Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. [Wang, Ya-lan] Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. RP Wang, Yl (reprint author), Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. EM wangyalan074@126.com CR D’amours D, Desnoyers S, D’silva I, Poirier GG, 1999, Poly, ADP-ribosyl)ation reactions in the regulation of nuclear functions. J Biochem 342:249–268 Parise RA, Shawaqfeh M, Egorin MJ, Beumer JH, 2008, Liquid chromatography-mass spactometric assay for quantitation in human plasma of ABT-888, an orally available, small molecule inhibitor of poly, ADP-ribose, polymerase. J Chromatogr B Analyt Technol Biomed Life Sci 872(1–2):141– 147 Hochegger H, Dejsuphong D, Fukushima T, Morrison C et al, 2006, Parp-1 protects homologous recombination from interferenve by Ku and Ligase IV in vertebrate cells. EMBO J 25:1305– 1314 Virag L, Szabo C, 2002, The therapeutic potential of poly, ADPribose, polymerase inhibitors. Pharmacol Rev 54:375–429 Meyer RG, Meyer-Ficca ML, Jacobson EL, Jacobson MK, 2003, Human poly, ADP-ribose, glycohydrolase, PARG, gene and the common promoter sequence it shares with inner mitochondrial membrane translocase 23, TIM23). Gene 314:181–190 Koh DW, Lawler AM, Poitras MF et al, 2004, Failure to degrade poly, ADP-ribose, increased sensitivity to cytotoxicity and early embryonic lethality. PNAS 101(51):17699–17704 Cortes U, Tong WM, Coyle DL et al, 2004, Depletion of the 110-Kilodalton isoform of poly, ADP-ribose, glycohydrolase increases sensitivity to genotoxic and endotoxic stress in mice. MCB 24(16):7163–7178 Poitras MF, Koh DW, Yu SWet al, 2007, Spatial and functional relationship between poly, ADP-ribose, polymerase-1 and Poly, ADP-ribose, glycohydrolase in the brain. Neuroscience 148, 1):198–211 Miwa M, Matsutani, 2007, PolyADP-ribosylation and cancer. Cancer Sci 98(10):1528–1535 Blenn C, Althaus FR, Manlanga M, 2006, Poly, ADP-ribose, glycohydrolase silencing protects against H2O2-induced cell death. J Biochem 396:419–429 Ratnam K, Low JA, 2007, Current development of clinical poly, ADP-ribose, inhibitor in oncology. Clin Cancer Res 13, 5):1383–1388 Decker P, Miranda EA, De Murcia G, Muller S, 1999, An improved nonisotopic test to screen a large series of new inhibitor molecules of poly, ADP-ribose, polymerase activity for therapeutic applications. Clin Cancer Res 5:1169–1172 Cuzzocrea S, Paola RD, Mazzon E et al, 2005, PARG activity mediates intestinal injury induced by splanchnic artery occlusion and reperfusion. FASEB J 19:558–566 Plummer R, Jones C, Middleton M et al, 2008, Phase I study of the poly, ADP-ribose, polymerase inhibitor, AG014699, in combination with temozolomide in patients with advanced Solid Tumors. Clin Cancer Res 14(23):7917–7923 Valenzuela MT, Guerrerro R, Nunez MI et al, 2002, PARP-1 modifies the effectiveness of P53 mediated DNA damage response. Oncogene 21(7):1108–1116 Oei SL, Ziegler M, 2000, ATP for the DNA ligation step in base excision repair is generated from poly, ADP-ribose). J Biol Chem 275:23234–23239 McCabe N, Turner NC, Lord CJ et al, 2006, Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly, ADP-ribose, polymerase inhibition. Cancer Res 66(16):8109–8115 Eguchi Y, Shimizu S, Tsujimoto Y, 1997, Intracellular ATP levels determine cell death fate by apoptosis or necrosis. Cancer Res 57:1835–1840 Kumari SR, Mendoza-Alvarez H, Alvarez-Gonzalez R, 1998, Functional interactions of p53 with poly(ADP-ribose, polymerase, PARP, during apoptosis following DNA damage: covalent poly(ADP-ribosyl)ation of p53by exogenous PARP and noncovalent binding of p53 to the Mr 85,000 proteolytic fragment1. Cancer Res 58:5075–5078 Affar EB, Germain M, Winstall E et al, 2001, Caspase-3- mediated processing of poly, ADP-ribose, glycohydrolase during apoptosis. JBC 276(4):2935–2942 Koh DW, Dawson TM, Dawson VL, 2005, Mediation of cell death by Poly, ADP-ribose)-1. Pharmacol Res 52(1):5–14 Bhaskara, et al, 2009, Differrential PARP cleavage: an indication for existence of multiple forms of cell death in human gilomas. Neurology India 57(3) Hung Q, Wu YT, Tan HL, Ong CN, Shen HM, 2009, A novel function of PARP-1 in modulation of autophagy and necrosis under oxidative stress. Cell Death Diff 16:264–277 Lee Y, Shacter E, 1999, Oxidative stress inhibits apoptosis in human lymphoma cells. J Bio Chem 274:19792–19798 Walisser JA, Thies RC, 1999, Poly, ADP-ribose, polymerase inhibition in oxidant stressed endothelial cells prevents oncosis and permits caspase activation and apoptosis. Exp Cell Res 251, 12):401–413 Mathews MT, Berk BC, 2008, PARP-1 inhibition prevents oxidative and nitrosative stress–induced endothelial cell death via transactivation of the VEGF receptor 2. Arterioscler Thromb Vasc Biol 28:711–717 Kovacs K, Toth A, Deres P et al, 2006, Critical role of PI3- kinase/Akt activation in the PARP inhibitor induced heart function recovery during ischemia–reperfusion. Biochem Pharmacol 71(4):441–452 Klaidman L, Morales M, Kem S et al, 2003, Nicotinamide offers multiple protective mechanisms in stroke as a precursor for NAD+, as a PARP inhibitor and by partial restoration of mitochondrial function. Pharmacology 69(3):150–157 Filipovic DM, Meng X, Reeves WB, 1999, Inhibition of PARP prevents oxidant-induced necrosis but not apoptosis in LLC-PK1 cells. Am J Physiol Renal Physiol 277:428–436 Roesner JP, Vagts DA, Iber T et al, 2006, Protective effects of PARP inhibition on liver microcirculation and function after haemorrhagic shock and resuscitation in male rats. Intensive Care Med 32(10):1649–1657 Oku H, Goto W, Tsujimoto M et al, 2003, Effects of Poly(ADPribose, polymerase, PARP, Inhibitor on NMDA-induced retinal injury. Invest Ophthalmol Vis Sci 44: E-Abstract 27 Charron MJ, Bonner-Weir S, 1999, Implicating PARP and NAD+ depletion in type I diabetes. Nat Med 5:269–270 Ha HC, Snyder SH, 1999, Poly, ADP-ribose, polymerase is a mediator of necrotic cell death by ATP depletion. PNAS 96:13978–13982 Hanai S, Kanai M, Ohashi S et al, 2004, Loss of poly, ADPribose, glycohydrolase causes progressive neurodegeneration in Drosophila melanogaster. PNAS 101(1):82–86 Keil C, Grobe T, Oei SL, 2006, MNNG-induced cell death is controlled by interactions between PARP-1, Poly, ADP-ribose, glycohydrolase, and XRCC1. J Biochem 281:34394–34405 Menissier-de Murcia J, Mark M, Wendling O, Wynshaw-Boris A, de Murcia G, 2001, Early embryonic lethality in PARP-1 Atm double-mutant mice suggests a functional synergy in cell proliferation during development. MCB 21(5):1828–1832 Yang YG, Cortes U, Patnaik S, Jasin M, Wang Z, 2004, Ablation of PARP-1 does not interfere with the repair of DNA doublestrand breaks, but compromises the reactivation of stalled replication forks. Oncogene 23:3872–3882 [abstract] Nozaki T, Masutani M, Watanabe M et al, 1999, Syncytiotrophoblastic giant cells in Teratocarcinoma like tumors derived from Parp-disrupted mouse embryonic stem cells. PNAS 96, 23):13345–13350 Saxena A, Saffery R, Wong LH et al, 2002, Centromere proteins Cenpa, Cenpb, and Bub3 interact with poly, ADP-ribose, polymerase-1 protein and are poly, ADP-ribosyl, ated. J Biol Chem 277:26921–26926 Bhatia M, Kirkland JB, Meckling-Gill KA, 1996, Overexpression of poly, ADP-ribose, polymerase promotes cell cycle arrest and inhibits neutrophilic differentiation of NB4 acute promyelocytic leukemia cells. Cell Growth Differ 7(1):91–100 Bhatia M, Kirkland JB, Meckling-Gill KA, 1995, Modulation of poly, ADP-ribose, polymerase during neutrophilic and monocytic differentiation of promyelocytic, NB4, and myelocytic, HL-60, leukaemia cells. J Biochem 308:131–137 Harnacke K, Kruhoffer M, Orntoft TF, Hass R, 2005, Downmodulation of poly(ADP-ribose, polymerase-1, PARP-1, in human TUR leukemia cells restores transcriptional responsiveness for differentiation and cell cycle arrest. Eur J Cell Biol 84, 11):885–896 [abstract] Masutani M, Nozaki T, Watanabe M et al, 2001, Involvement of poly(ADP-ribose, polymerase in trophoblastic cell differentiation during tumorigenesis. Mutant Res 477:111–117 Shiokawa M, Masutani M, Fujihara H et al, 2005, Genetic alteration of poly(ADP-ribose, polymerase-1 in human germ cell tumors. Jpn J Clin Oncol 35(2):97–102 Sevigny MB, Silva JM, Lan WC, Alano CC, Swanson RA, 2003, Expression and activity of poly(ADP-ribose, glycohydrolase in cultured astrocytes, neurons, and C6 glioma cells. Mol Brain Res 117(2):213–220 [abstract] Kraus W, Lis J, 2003, PARP goes transcription. Cell 113, 6):677–683 Pavri R, Lewis B, Kim TK et al, 2005, PARP-1 determines specificity in a retinoid signaling pathway via direct modulation of mediator. Moll Cell 18(1):83–96 [abstract] Idogawa M, Yamada T, Honda K et al, 2005, Poly(ADPribose, polymerase-1 is a component of the oncogenic T-cell factor-4/beta-catenin complex. Gastroenterology 128(7):1919– 1936 [abstract] Soldatenkov VA, Albor A, Patel BKR et al, 1999, Regulation of the human poly(ADP-ribose, polymerase promoter by ETS transcription factor. Oncogene 18(27):3954–3962 Ogino H, Nozaki T, Gunji A et al, 2007, Loss of Parp-1 affects gene expression profile in a genome-wide manner in ES cells and liver cells. BMC Genomics 8:227., DOI 10.1186/1471-2164-8-227 Aldinussi A, Gerlini G, Fossati S et al, 2007, A key role for poly, ADP-ribose)-1 during human dendritic cell maturation. J Immuno 179:305–312 Zaniolo K, Desnoyers S, Leclerc S, Guerin S, 2007, Regulation of poly(ADP-ribose, polymerase-1, PARP-1, gene expression through the post-translational modification of Sp1: a nuclear target protein of PARP-1. BMC Genomics 8:96., DOI 10.1186/ 1471-2199-8-96 Tong WM, Hande MP, Lansdorp PM, Wang ZQ, 2001, DNA strand break-sensing molecule poly(ADP-Ribose, polymerase cooperates with p53 in telomere function, chromosome stability, and tumor suppression. Mol Cell Biol 21(12):4046–4054 Eberhart CG, 2003, Medulloblastoma in mice lacking p53 and PARP. Am J Pathol 162(1):7–10 Oei SL, Griesenbeck J, Schweiger M, Ziegler M, 1998, Regulation of RNA polymerase II-dependent transcription by poly(ADP-ribosyl)ation of transcription factors. JBC 273:31644–31647 Hassa PO, Buerki C, Lombardi C, Imhof R, Hottiger MO, 2003, Transcriptional coactivation of nuclear factor Kappa Bdependent gene expression by p300 is regulated by poly, ADP)-ribose polymerase-1. JBC 278(46):45145–45153 Nie J, Sakamoto S, Song D, Qu Z, Ota K, Taniguchi T, 1998, Interaction of Oct-1 and automodification domain of poly, ADPribose, synthetase. FEBS Lett 424(1–2):27–32 Dear TN, Hainzl T, Follo M, 1997, Identification of interaction partners for the basic-helix-loop-helix protein E47. Oncogene 14:891–898 Butler AJ, Ordhal CP, 1999, Poly, ADP-Ribose, polymerase binds with transcription enhancer factor 1 to MCAT1 elements to regulate muscle-specific transcription. Mol Cell Boil 19(1):296– 306 Taniguchi T, Agemori M, Kameshita I, Nishikimi M, Shizuta Y, 1982, Participation of poly(ADP-ribosyl)ation in the depression of RNA synthesis caused by treatment of mouse lymphoma cells with methylnitrosourea. J BC 257:4027–4030 Shimokawa T, Masutani M, Nagasawa S et al, 1999, Isolation and cloning of rat poly, ADP-ribose, glycohydrolase: presence of a potential nuclear export signal conserved in mammalian orthologs. J Biochem 126:748–755 Erdelyi K, Kiss A, Bankondi E et al, 2005, Gallotannin inhibits the expression of chemokines and inflammatory cytokines in A549 cells. Mol Pharmacol 68:895–904 Frizzell KM, Gamble MJ, Berrocal JG, 2009, Global analysis of transcriptional regulation by poly(ADP-ribose, polymerase-1 and poly(ADP-ribose, glycohydrolase in MCF-7 human breast cancer cells. J Biol Chem 284(49):33926–33938 [abstract] Wang ZQ, Stingl L, Morrison C et al, 1997, PARP is important for genomic stability but dispensable in? Apoptosis. Genes Dev 11:2347–2358 Folkman J, 2002, Role of angiogenesis in tumor growth and metastasis. Semin Oncol 29:15–18 Tentori L, Lacal PM, Muzi A et al, 2007, Poly(ADP-ribose, polymerase, PARP, inhibition or PARP-1 gene deletion reduces angiogenesis. Eur J Cancer 43(14):2124–2133 Pyriochou A, Olah G, Deitch EA, Szabo C, Papapetropoulos A, 2008, Inhibition of angiogenesis by the poly, ADP-ribose, polymerase inhibitor PJ-34. IJMM 22:113–118 Rajesh M, Mukhopadhyay P, Batkai S et al, 2006, Pharmacological inhibition of poly, ADP-ribose, polymerase inhibits angiogenesis. BBRC 350(2):352–357 Rajesh M, Mukhopadhyay P, Godlewski G, 2006, Poly, ADPribose, polymerase inhibition decreases angiogenesis. BBRC 350(4):1056–1062 Berger NA, Adams JW, Sikorski GW et al, 1978, Synthesis of DNA and poly, Adenosine Diphosphate Ribose, in normal and chronic lymphocytic leukemia lymphocytes. J Clin Inves 62:111–118 Wielckens K, Garbrecht M, Kittler M, Hilz H, 1980, ADPribosylation of nuclear proteins in normal lymphocytes and in low- grade malignant non-Hodgkin lymphoma cells. Eur J Biochem 104:279–287 Hirai K, Ueda K, Hayaishi O, 1983, Abberation of poly, adenosine diphosphate ribose, in human colon adenomatous polyps and cancers. Cancer Res 43:3441–3446 Fukushima M, Kuzuya OK, Ikai K, 1981, Poly, ADP-ribose, synthesis in human cervical cancer cell diagnostic cytological usefulness. Cancer Lett 14:227–236 Shiobara M, Miyazaki M, Ito H et al, 2001, Enhanced polyadenosine diphosphate ribosylation in cirrhotic liver and carcinoma tissues in patients with hepatocellular carcinoma. J Gastroenterol Hepatol 16:338–344 Lin L, Li J, Wang YL et al, 2009, Relationship of PARG with colorectal carcinoma PARP, VEGF and b-FGF in colorectal carcinoma. Chin J Cancer Res 21(2):135–141 Leal AP et al, 2009, PARP inhibitors: New partners in the therapy of cancer and inflammatory diseases. Free Radic Biol Med, in press). Doi:10.1016 Lord CJ, McDonald S, Swift S et al, 2008, A high-throughput RNA interference screen for DNA repair determinants of PARP inhibitor sensitivity. DNA Repair, Amst, 7:2010–2019 Turner NC, Lord CJ, Iorns E et al, 2008, A synthetic lethal siRNA screen identifying genes mediating sensitivity to a PARP inhibitor. EMBO J 27(9):1368–1377 Ashworth A, 2008, A synthetic lethal therapeutic approach: poly, ADP, ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair. J Clin Oncol 26, 22):3785–3790 Bryant HE, Schultz N, Thomas HD et al, 2005, Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADPribose, polymerase. Nature 434:913–917 [abstract] Turner N, Tutt A, Ashworth A, 2005, Targeting the DNA repair defect of BRCA tumours. Curr Opin Pharmacol 5, 4):388–393 De Soto JA, Wang X, Tominaga Y et al, 2006, The inhibition and treatment of breast cancer with poly, ADP-ribose, polymerase, PARP-1, inhibitors. Int J Biol Sci 2(4):179–185 De Soto JA, Deng CX, 2006, PARP-1 inhibitors: are they the long-sought genetically specific drugs for BRCA1/2-associated breast cancers? Int J Med Sci 3(4):117–123 Curtin NJ, 2005, PARP inhibitors for cancer therapy. Expert Rev Mol Med 7(4):1–20 [abstract] Liu X, Shi Y, Guan R et al, 2008, Potentiation of temozolomide cytotoxicity by poly(ADP)ribose polymerase inhibitor ABT-888 requires a conversion of single stranded DNA damages to double-stranded DNA breaks. Mol Cancer Res 6(10):1621–1629 Suesse S, Scholz CJ, Buerkle A, Iler LW, 2004, Poly, ADPribose, polymerase, PARP-1, and P53 independently function in regulating double strand break repair in primate cells. Nucleic Acids Res 32:2669–2680 Purnell MR, Whish WJD, 1980, Novel inhibitors of poly, ADPribose, synthetase. J Biochem 185:775–777 Banasik M, Komura H, Shimoyama M, Ueda K, 1992, Specific inhibitors of poly, ADP-ribose, synthetase and mono(ADPribosyl, transferase. J Biol Chem 267:1569–1575 Genovese T, Di Paola R, Catalano P et al, 2004, Treatment with a novel poly(ADP-ribose, glycohydrolase inhibitor reduces development of septic shock-like syndrome induced by zymosan in mice. Crit Care Med 32(6):1365–1374 Masutani M, Shimokawa T, Igarashi M et al, 2002)Inhibition of poly(ADP-ribose, glycohydrolase activity by cyclic peptide antibiotics containing piperazic acid residues. Proceedings of the Japanese academy; Ser B, physical and biological sciences 78, 1):15–17 Formentini L, Arapistas P, Pitelli M et al, 2008, Mono-galloyl glucose derivatives are potent poly(ADP-ribose, glycohydrolase, PARG, inhibitors and partially reduced PARP-1 dependent cell death. BJP 155:1235–1249 Yang X, Lippman ME, 1999, BRCA1 and BRCA2 in breast cancer. Breast Cancer Res Treat 54(1):1–10 De Soto JA, Wang X, Tominaga Y et al, 2006, The inhibition and treatment of breast cancer with poly, ADP-ribose, polymerase, PARP-1, inhibitors. Int J Biol Sci 2(4):179–185 Munozo-Gamez JA, Martin-Olivia D, Aguilar-Quesada R, 2005, PARP inhibition sensitizes p53-deficient breast cancer cells to doxorubicin-induced apoptosis. Biochem J 386:119–125 Rottenberg S, Jaspers JE, Kersbergen A et al, 2008, High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD2281 alone and in combination with platinum drugs. PNAS 105(44):17079–17084 Gopall R, Hong C & Yi S, 2009, Gefitinib as monotherapy in the first-line setting in non-small cell lung Cancer. http://www.ispub. com/journal/the_internet_journal_of_pulmonary_medicine/ volume_11_number_2_5/article/gefitinib-as-monotherapy-in-thefirst- line-setting-in-non-small-cell-lung-cancer.html Albert JM, Cao C, Kim KWet al, 2007, Inhibition of poly(ADPribose, polymerase enhances cell death and improves tumor growth delay in irradiated lung cancer models. Clin Cancer Res 13(10):3033–3042 Kinders RJ, Hollingshead M, Khin S et al, 2008, Preclinical modeling of a phase 0 clinical trial: qualification of a pharmacodynamic assay of poly, ADP-ribose, polymerase in tumor biopsies of mouse xenografts. Clin Cancer Res 14(21):6877–6885 Donawho CK, Luo Y, Penning TD et al, 2007, ABT-888, an orally active poly(ADP-ribose, polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models. Clin Cancer Res 13(9):2728–2737 Calabrese CR, Batey MA, Thomas HD, 2003, Identification of potent nontoxic poly, ADP-ribose, polymerase-1 inhibitors: chemopotentiation and pharmacological studies. Clin Cancer Res 9:2711–2718 Hao LX, Wang YL, Cai L, YY LI, 2007, Inhibitory effect of 5-Aminoisoquinolinone on Parp activity in colon carcinoma cell line HT-29. Chin J Cancer 26(6):566–571 Li M, Threadgill MD, Wang YL, Cai L, Lin X, 2009, A poly, ADP-ribose, polymerase inhibition down-regulates expression of metastasis-related genes in CT26 colon carcinoma cells. Pathobiology 76:108–116 Tentori L, Portarena I, Barbarino M et al, 2003, Inhibition of telomerase increases resistance of melanoma cells to temozolomide, but not to temozolomide combined with poly, ADPribose, polymerase inhibitor. Mol Pharmacol 63:192–202 Tentori L, Leonetti C, Scarsella M et al, 2003, Systemic administration of GPI 15427, a novel poly(ADP-ribose, polymerase-1 inhibitor, increases the antitumor activity of temozolomide against intracranial melanoma, glioma, lymphoma. Clin Cancer Res 9:5370–5379 Tentori L, Leonetti C, Scarsella M et al, 2005, Poly(ADP-ribose, glycohydrolase inhibitor as chemosensitiser of malignant melanoma for temozolomide. Eur J Cancer 41(18):2948–2957 Dungey FA, Caldecott KW, Chalmers AJ, 2009, Enhanced radiosensitisation of human glioma cells by combining inhibition of PARP with inhibition of Hsp90. Mol Cancer Ther 8(8):2243–2254 Fong PC, Boss DS, Yap TA et al, 2009, Inhibition of poly(ADPribose, polymerase in tumors from BRCA mutation carriers. N Engl J Med 361:123–134 Gien LT, Mackay HJ, 2010, The emerging role of PARP inhibitors in the treatment of epithelial ovarian cancer. J Oncol., DOI 10.1155/2010/151750 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 469 EP 478 DI 10.1007/s12253-010-9266-6 PG 10 ER PT J AU Wittschieber, D Kollermann, J Schlomm, Th Sauter, G Erbersdobler, A AF Wittschieber, Daniel Kollermann, Jens Schlomm, Thorsten Sauter, Guido Erbersdobler, Andreas TI Nuclear Grading Versus Gleason Grading in Small Samples Containing Prostate Cancer: A Tissue Microarray Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Gleason grading; Fuhrman nuclear grading; Prognosis; TMA ID Prostate cancer; Gleason grading; Fuhrman nuclear grading; Prognosis; TMA AB In this study we addressed the question whether nuclear grading in very small samples of prostate cancer would provide additional prognostic information as compared to Gleason grading. Therefore, a tissue microarray (TMA) was constructed comprising a total number of 3,261 prostate cancers. Blinded for all clinical and pathological data, the TMA spots (diameter 0.6 mm) containing cancer were graded with two systems: First, for nuclear features according to a modified Fuhrman grading system, and second, by using a simplified Gleason system. The results were compared with tumour stage, tumour grade and follow-up data. Although nuclear grading could easily be performed on the TMA spots, no correlation was found with tumour stage, grade or PSA recurrence after prostatectomy. However, Gleason grading, even when performed on the small TMA spots, provided significant prognostic information. Correlation with Gleason scores determined in the complete prostatectomy specimens showed moderate agreement in low-grade (score≤6) or intermediate (score=7) tumours, but poor agreement with high-grade (score≥8) tumours. In conclusion, the Fuhrman grading of prostate cancer does not appear to be of any prognostic importance so the Gleason grading remains the system of choice, even in tumour specimens smaller than 1 mm. C1 [Wittschieber, Daniel] Charite University Hospital, Institute of Pathology, Chariteplatz 1, 10117 Berlin, Germany. [Kollermann, Jens] University Medical Center, Institute of PathologyHamburg-Eppendorf, Germany. [Schlomm, Thorsten] University Medical Center, Martini-Clinic, Prostate Cancer CenterHamburg-Eppendorf, Germany. [Sauter, Guido] University Medical Center, Institute of PathologyHamburg-Eppendorf, Germany. [Erbersdobler, Andreas] Charite University Hospital, Institute of Pathology, Chariteplatz 1, 10117 Berlin, Germany. RP Wittschieber, D (reprint author), Charite University Hospital, Institute of Pathology, 10117 Berlin, Germany. EM daniel.wittschieber@charite.de CR Gleason DF, 1966, Classification of prostatic carcinomas. Cancer Chemother Rep 50:125–128 Gleason DF, Mellinger GT, 1974, Prediction of prognosis for prostatic adenocarcinoma by combined histological grading and clinical staging. J Urol 111:58–64 Gleason DF, 1992, Histologic grading of prostate cancer: a perspective. Hum Pathol 23:273–279 Epstein JI, Pizov G, Walsh PC, 1993, Correlation of pathologic findings with progression after radical retropubic prostatectomy. Cancer 71:3582–3593 Oesterling JE, Brendler CB, Epstein JI et al, 1987, Correlation of clinical stage, serum prostatic acid phosphatase and preoperative Gleason grade with final pathological stage in 275 patients with clinically localized adenocarcinoma of the prostate. J Urol 138:92–98 Di Silverio F, D’Eramo G, Buscarini M et al, 1996, DNA ploidy, Gleason score, pathological stage and serum PSA levels as predictors of disease-free survival in C-D1 prostatic cancer patients submitted to radical retropubic prostatectomy. Eur Urol 30:316–321 Sogani PC, Israel A, Lieberman PH et al, 1985, Gleason grading of prostate cancer: a predictor of survival. Urology 25:223–227 Bostwick DG, Foster CS, 1999, Predictive factors in prostate cancer: current concepts from the 1999 College of American Pathologists Conference on Solid Tumor Prognostic Factors and the 1999 World Health Organization Second International Consultation on Prostate Cancer. Semin Urol Oncol 17:222–272 Humphrey PA, 2004, Gleason grading and prognostic factors in carcinoma of the prostate. Mod Pathol 17:292–306 Epstein JI, Carmichael M, Partin AWet al, 1993, Is tumor volume an independent predictor of progression following radical prostatectomy? A multivariate analysis of 185 clinical stage B adenocarcinomas of the prostate with 5 years of followup. J Urol 149:1478–1481 Epstein JI, CarmichaelMJ, Pizov G et al, 1993, Influence of capsular penetration on progression following radical prostatectomy: a study of 196 cases with long-term followup. J Urol 150:135–141 Egan AJ, Bostwick DG, 1997, Prediction of extraprostatic extension of prostate cancer based on needle biopsy findings: perineural invasion lacks significance on multivariate analysis. Am J Surg Pathol 21:1496–1500 Sagalowsky AI, Milam H, Reveley LR, 1982, Prediction of lymphatic metastases by Gleason histologic grading in prostatic cancer. J Urol 128:951–952 Gaeta JF, Asirwatham JE,Miller G et al, 1980, Histologic grading of primary prostatic cancer: a new approach to an old problem. J Urol 123:689–693 Bocking A, Kiehn J, Heinzel-Wach M, 1982, Combined histologic grading of prostatic carcinoma. Cancer 50:288–294 Zhou M, Hayasaka S, Taylor JM et al, 2001, Lack of association of prostate carcinoma nuclear grading with prostate specific antigen recurrence after radical prostatectomy. J Urol 166:2193–2197 Khan MA, Walsh PC, Miller MC et al, 2003, Quantitative alterations in nuclear structure predict prostate carcinoma distant metastasis and death in men with biochemical recurrence after radical prostatectomy. Cancer 98:2583–2591 Veltri RW, Miller MC, Isharwal S et al, 2008, Prediction of prostate-specific antigen recurrence in men with long-term followup postprostatectomy using quantitative nuclear morphometry. Cancer Epidemiol Biomark Prev 17:102–110 Fuhrman SA, Lasky LC, Limas C, 1982, Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol 6:655–663 Bubendorf L, Kononen J, Koivisto P et al, 1999, Survey of gene amplifications during prostate cancer progression by highthroughout fluorescence in situ hybridization on tissue microarrays. Cancer Res 59:803–806 Schlomm T, Kirstein P, Iwers L et al, 2007, Clinical significance of epidermal growth factor receptor protein overexpression and gene copy number gains in prostate cancer. Clin Cancer Res 13:6579–6584 Schlomm T, Iwers L, Kirstein P et al, 2008, Clinical significance of p53 alterations in surgically treated prostate cancers. Mod Pathol 21:1371–1378 Erbersdobler A, Isbarn H, Steiner I et al, 2009, Predictive value of prostate-specific antigen expression in prostate cancer: a tissue microarray study. Urology 74:1169–1173 DeMarzo AM, Nelson WG, Isaacs WB et al, 2003, Pathological and molecular aspects of prostate cancer. Lancet 361:955–964 Epstein JI, 2004, Diagnosis and reporting of limited adenocarcinoma of the prostate on needle biopsy. Mod Pathol 17:307–315 Diamond DA, Berry SJ, Umbricht C et al, 1982, Computerized image analysis of nuclear shape as a prognostic factor for prostatic cancer. Prostate 3:321–332 Partin AW, Walsh AC, Pitcock RV et al, 1982, A comparison of nuclear morphometry and Gleason grade as a predictor of prognosis in stage A2 prostate cancer: a critical analysis. J Urol 142:1254–1258 Robutti F, Pilato FP, Betta PG, 1989, A new method of grading malignancy of prostate carcinoma using quantitative microscopic nuclear features. Pathol Res Pract 185:701–703 Fujikawa K, Sasaki M, Arai Y et al, 1997, Prognostic criteria in patients with prostate cancer: Gleason score versus volumeweighted mean nuclear volume. Clin Cancer Res 3:613–618 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 479 EP 484 DI 10.1007/s12253-010-9270-x PG 6 ER PT J AU Anghel, A Narita, D Seclaman, E Popovici, E Anghel, M Tamas, L AF Anghel, Andrei Narita, Diana Seclaman, Edward Popovici, Emilian Anghel, Mariana Tamas, Liviu TI Estrogen Receptor Alpha Polymorphisms and the Risk of Malignancies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute myeloid leukemia; Bladder; Breast; Colorectal; Hepatocellular carcinomas; Susceptibility; Estrogen receptor alpha gene; Single nucleotide polymorphisms ID Acute myeloid leukemia; Bladder; Breast; Colorectal; Hepatocellular carcinomas; Susceptibility; Estrogen receptor alpha gene; Single nucleotide polymorphisms AB Estrogens represent risk factors for endocrinerelated cancers and play also an important role in the development and progression of other malignancies. In order to analyze the associations between estrogen receptor gene alpha polymorphisms and cancers susceptibility, we genotyped six single nucleotide polymorphisms (SNPs) in 163 Caucasian cancer patients—103 breast cancers and 60 other malignancies (colorectal, bladder, hepatocellular carcinoma and acute myeloid leukemia)—and 114 healthy controls using hybridization probes. We performed Armitage` s association trend-test to evaluate the risk. Linkage disequilibrium (LD) was assessed for each pair of markers. The genotypes CC and CT of rs3798577 were significantly associated with the cancers risk (p-trend breast=4×10-5; p-trend cancers=1×10-5); in discrepancy with breast cancer where the C-allele represented the risk allele, for bladder, hepatocellular carcinomas and leukemia, the T allele seems to confer susceptibility. The minor G allele of rs1801132 was protective in our cases (p=1×10-4); for rs2228480, the heterozygous frequency was higher for cancer groups (p=0.03); the SNP pairs rs2228480&rs3798577 and rs2234693&rs9340799 were in low LD; the haplo types T-A of rs2234693&rs9340799 and G-C of rs2228480&rs3798577 showed a trend to be higher represented in breast cancers; T allele of rs2234693 was higher expressed in breast, colon cancers and leukemia; rs2077647 was associated with colon (p=0.008, C-risk allele) and bladder (p=0.01, T-risk allele) cancers. We concluded that ESR1 polymorphisms may have distinct impact in carcinogenesis and further genotyping will establish whether these findings remain significant in larger cohorts. C1 [Anghel, Andrei] University of Medicine and Pharmacy, Biochemistry Department, Eftimie Murgu 2A, 300041 Timisoara, Romania. [Narita, Diana] University of Medicine and Pharmacy, Biochemistry Department, Eftimie Murgu 2A, 300041 Timisoara, Romania. [Seclaman, Edward] University of Medicine and Pharmacy, Biochemistry Department, Eftimie Murgu 2A, 300041 Timisoara, Romania. [Popovici, Emilian] University of Medicine and Pharmacy, Eftimie Murgu 2A, 300041 Timisoara, Romania. [Anghel, Mariana] University of Medicine and Pharmacy, Eftimie Murgu 2A, 300041 Timisoara, Romania. [Tamas, Liviu] University of Medicine and Pharmacy, Biochemistry Department, Eftimie Murgu 2A, 300041 Timisoara, Romania. RP Narita, D (reprint author), University of Medicine and Pharmacy, Biochemistry Department, 300041 Timisoara, Romania. EM diana_narita@yahoo.com;biochim@umft.ro CR Dunning AM, Healey CS, Pharoah PD et al, 1999, A systematic review of genetic polymorphisms and breast cancer risk. Cancer Epidemiol Biomarkers Prev 8:843–854 Pharoah PD, Antoniou AC, Easton DF, Ponder BA, 2008, Polygenes, risk prediction, and targeted prevention of breast cancer. N Engl J Med 358:2796–2803 Ripperger T, Gadzicki MA, Schlegelberger B, 2009, Breast cancer susceptibility: current knowledge and implications for genetic counseling. Eu J Human Genetics 17:722–731 Pharoah PD, Tyrer J, Dunning AM et al, 2007, Association between common variation in 120 candidate genes and breast cancer risk. PLoS Genet 3:e42 Gold B, Kirchhoff T, Stefanov S et al, 2008, Genome-wide association study provides evidence for a breast cancer risk locus at 6q22.33. PNAS 105:4340–4345 Easton DF, Pooley KA, Dunning AM et al, 2007, Genome-wide association study identifies novel breast cancer susceptibility loci. Nature 447:1087–1093 Baynes C, Healey CS, Pooley CA et al, 2007, Common variants in the ATM, BRCA1, BRCA2, CHEK2 and TP53 cancer susceptibility genes are unlike to increase breast cancer risk. Breast Cancer Res 9:R27 Gennari L, Merlotti D, De Paola V et al, 2005, Estrogen receptor gene polymorphisms and the genetics of osteoporosis: a HuGE review. Am J Epidemiol 161:307–320 Deroo BJ, Korach KS, 2006, Estrogen receptors and human disease. J Clin Invest 116:561–570 Breivik RA, Lothe GI, Meling TO et al, 1997, Different genetic pathways to proximal and distal colorectal cancer influenced by sex-related factors. Int J Cancer 74:664–667 Martineti V, Silvestri S, Tonelli F, Brandi ML, 2008, Control of colon cancer development and progression by selected estrogen receptor modulators. Expert Rev Endocrinol Metab 3:503–511 Foley EF, Jazaeri AA, Shupnik MA et al, 2000, Selective loss of estrogen receptor beta in malignant human colon. Cancer Res 60:245–248 Kouzmenko AP, Takeyama K, Kawasaki Y et al, 2008, Liganddependent interaction between estrogen receptor alpha and adenomatous polyposis coli. Genes To Cells 13:723–730 Slattery ML, Sweeney C, Murtaugh M et al, 2005, Associations between ERalpha, ERbeta, and AR genotypes and colon and rectal cancer. Cancer Epidemiol Biomarkers Prev 14:2936–2942 Liu W, Konduri SD, Bansal S et al, 2006, Estrogen receptor-alpha binds p53 tumor suppressor protein directly and represses its function. J Biol Chemistry 281:9837–9840 Teng J, Wang ZY, Jarrard DF, Bjorling DE, 2008, Roles of estrogen receptor α and β in modulating urothelial cell proliferation. Endocr Relat Canc 15:351–364 Sun W, Zhong F, Zhi L et al, 2009, Systematic-omics analysis of HBV-associated liver diseases. Cancer Lett 286:89–95 Deng G, Zhou G, Zhai Y et al, 2004, Association of estrogen receptor Alpha polymorphisms with susceptibility to chronic hepatitis B virus infection. Hepatology 40:318–326 Zhai Y, Zhou G, Deng G et al, 2006, Estrogen receptor alpha polymorphisms associated with susceptibility to hepatocellular carcinoma in hepatitis B virus carriers. Gastroenterology 130:2001–2009 Danel L, Cordier G, Revillard JP, Saez S, 1982, Presence of estrogen binding sites and growth-stimulating effect of estradiol in the human myelogenous cell line HL60. Cancer Res 42:4701– 4705 Hatfill SJ, Brusnicky J, Fester E, 1991, Immunocytochemical identification of nuclear estrogen-receptors in human acute myeloid leukemia. Leuk Res 15:315–320 Skibola CF, Bracci PM, Halperin E et al, 2008, Polymorphisms in the estrogen receptor 1 and vitamin C and matrix metalloproteinase gene families are associated with susceptibility to lymphoma. PLoS 3:e2816 Yuan HY, Chiou JJ, Tseng WH et al, 2006, FASTSNP: an always up-to-date and extendable service for SNP function analysis and prioritization. Nucleic Acids Res 34(Web Server issue):W635–641 Conde L, Vaquerizas JM, Dopazo H, et al, 2006, PupaSuite: finding functional single nucleotide polymorphisms for large-scale genotyping purposes. Nucleic Acids Res 34(Web Server issue): W621-625 Available from http://bioinfo.cipf.es/pupasuite/ Raymond M, Rousset F, 1995, GENEPOP, version 1.2): population genetics software for exact tests and ecumenicism. J. Heredity, 86:248–249. http://genepop.curtin.edu.au/ Gaunt TR, Rodriguez S, Day INM, 2007, Cubic exact solutions for the estimation of pair-wise haplotype frequencies: implications for linkage disequilibrium analyses and a web tool 'CubeX'. BMC Bioinformatics 8:428–437. Available from http://www.oege.org/ software/cubex/ International HapMap Consortium, Frazer KA, Ballinger DG, Cox DR et al, 2007, A second generation human haplotype map of over 3.1 million SNPs. Nature 449:851–861 Nilsson M, Dahlmann I, Jiao H et al, 2007, Impact of estrogen receptor gene polymorphisms and mRNA levels on obesity and lipolysis—a cohort study. BMC Medical Genetics 8:73–85 Zofkova I, Zajickova K, Hill M, 2002, The estrogen receptor alpha gene determines serum androstenedione levels in postmenopausal women. Steroids 67:815–819 Herrington DM, Howard TD, Hawkins GA, 2002, Estrogenreceptor polymorphism and effects of estrogen replacement therapy on high-density lipoprotein cholesterol in women with coronary disease. N Engl J Med 346:967–974 Long JR, Xu H, Zhao LJ et al, 2005, The estrogen receptor α gene is linked and/or associated with age of menarche in different ethnic groups. J Med Genet 42:796–800 Speer G, Cseh K, Winkler G, 2001, Estrogen and vitamin D receptor, VDR, genotypes and the expression of ErbB-2 and EGF receptor in human rectal cancers. Eur J Cancer 37:1463–1468 Notarnicola M, Gristina R, Messa C, 2001, Estrogen receptors and microsatellite instability in colorectal carcinoma patients. Cancer Lett 168:65–70 Kadiyska T, Yakulov T, Kaneva R et al, 2007, Vitamin D and estrogen receptor gene polymorphisms and the risk of colorectal cancer in Bulgaria. Int J Colorectal Dis 22:395–400 Zuppan PJ, 1989, Polymorphisms at the estrogen receptor locus and linkage relationships on chromosome 6q. Cytogenet Cell Genet 51:1116–1120 Yaich L, Dupont WD, Cavener DR, Parl FF, 1992, Analysis of the PvuII restriction fragment-length polymorphism and exon structure of the estrogen receptor gene in breast cancer and peripheral blood. Cancer Res 52:77–83 Jakimiuk AJ, Nowicka M, Bogusiewicz M et al, 2007, Prevalence of estrogen receptor α PvuII and XbaI polymorphism in population of Polish postmenopausal women. Folia Histochem et cytobiol 45:331–338 Boyapati SM, Shu XO, Ruan ZX et al, 2005, Polymorphisms in ER-α gene interact with estrogen receptor status in breast cancer survival. Clin Cancer Res 11:1093–1098 Moret-Onland CN, van Gils CH, Roest M et al, 2005, The estrogen receptor α gene and breast cancer risk. Canc Causes Contr 16:1195–1202 Gonzalez-Zuloeta Ladd AM, Vasquez AA, Rivadeneira F et al, 2008, Estrogen receptor α polymorphisms and postmenopausal breast cancer risk. Breast Cancer Res Treat 107:415–419 Wedren S, Lovmar L, Humphreys K et al, 2004, Estrogen receptor α gene haplotype and postmenopausal breast cancer risk: a case control study. Breast Cancer Res 6:R437–R449 Anderson TI, Heimdal KR, Skrede M et al, 2004, Oestrogen receptor, ESR, polymorphisms and breast cancer susceptibility. Hum Genet 94:665–670 Shin A, Kang D, Nishio H et al, 2003, Estrogen receptor alpha gene polymorphisms and breast cancer risk. Breast Cancer Res Treat 80:127–131 Cai Q, Shu XO, Jin F et al, 2003, Genetic polymorphisms in the estrogen receptor alpha gene and risk of breast cancer: results from the Shanghai breast cancer study. Cancer Epidemiol Biomarkers Prev 12:853–859 Tanaka Y, Sasaki M, Kaneuchi M et al, 2003, Estrogen receptor alpha polymorphisms and renal cell carcinoma-a possible risk. Mol Cell Endocrinol 202:109–116 Tapper W, Hammond V, Gerty S, Ennis S, Simmonds P, Collins A, the Prospective study of Outcomes in Sporadic versus Hereditary breast cancer, POSH, Steering Group and Diana Eccles, 2008, The influence of genetic variation in 30 selected genes on the clinical characteristics of early onset breast cancer. Breast Cancer Res 10:R108–R118 Einarsdottir K, Darabi H, Li Yet al, 2008, ESR1 and EGF genetic variation in relation to breast cancer risk and survival. Breast Cancer Res 10:R15–R24 Hsiao WC, Young KC, Lin SL, Lin PW, 2004, Estrogen receptor–α polymorphism in Taiwanese clinical breast cancer population: a case-control study. Breast Cancer Res 6:180– 186 Iwase H, Kobayashi S, 1997, Alterations and polymorphisms of the estrogen receptor gene in breast cancer. Breast Cancer 4:57– 66 Roodi N, Bailey LR, Kao WYet al, 1995, Estrogen receptor gene analysis in estrogen receptor-positive and receptor-negative primary breast cancer. J Natl Cancer Inst 87:446–451 Vasconcelos A, Medeiros R, Veiga I et al, 2002, Analysis of estrogen receptor polymorphism in codon 325 by PCR-SSCP in breast cancer: association with lymph node metastasis. Breast J 8:226–229 Southey MC, Batten LE, McCredie MR et al, 1998, Estrogen receptor polymorphism at codon 325 and risk of breast cancer in women before age forty. J Nat Cancer Inst 90:532–536 Gallichio L, Berndt S, McSorley MA et al, 2006, Polymorphisms in estrogen-metabolizing and estrogen receptor genes and the risk of developing breast cancer among a cohort of women with benign breast disease. BMC Cancer 6:173–184 Gold B, Kalush F, Bergeron J et al, 2004, Estrogen receptor genotypes and haplotypes associated with breast cancer risk. Cancer Res 64:8891–8900 Wang J, Higuchi R, Modugno F et al, 2007, Estrogen receptor alpha haplotypes and breast cancer risk in older Caucasian women. Breast Cancer Res and Treat 106:273–280 Yang J, Singleton DW, Shaughnessy EA, Khan SA, 2008, The Fdomain of estrogen receptor-alpha inhibits ligand induced receptor dimerization. Mol Cell Endocrinol 295:94–100 Venables JP, 2004, Aberrant and alternative splicing in cancer. Cancer Res 64:7647–7654 Lin S, Coutinho-Mansfield G, Wang D et al, 2008, The splicing factor SC35 has an active role in transcriptional elongation. Nat Struct&Mol Biol 15:819–826 Shi J, Hu Z, Pabon K, Scotto KW, 2008, Caffeine regulates alternative splicing in a subset of cancer-associated genes: a role for SC35. Mol Cell Biol 28:883–895 Long JR, Zhao LJ, Liu PY et al, 2004, Pattern of linkage disequilibrium and haplotype distribution in disease candidate genes. BMC Genetics 5:11–18 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 485 EP 496 DI 10.1007/s12253-010-9263-9 PG 12 ER PT J AU Michailidi, Ch Giaginis, C Stolakis, V Alexandrou, P Klijanienko, J Delladetsima, I Chatzizacharias, N Tsourouflis, G Theocharis, S AF Michailidi, Christina Giaginis, Costas Stolakis, Vassilios Alexandrou, Paraskevi Klijanienko, Jerzy Delladetsima, Ioanna Chatzizacharias, Nicolaos Tsourouflis, Gerasimos Theocharis, Stamatios TI Evaluation of FAK and Src Expression in Human Benign and Malignant Thyroid Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Focal Adhesion Kinase (FAK); Src; Thyroid cancer; immunohistochemistry; Clinicopathological parameters; Diagnosis ID Focal Adhesion Kinase (FAK); Src; Thyroid cancer; immunohistochemistry; Clinicopathological parameters; Diagnosis AB Focal Adhesion Kinase (FAK) and Src have been reported to regulate tumor growth, invasion, metastasis and angiogenesis. The present study aimed to evaluate by immunohistochemistry the clinical significance of FAK and Src expression in 108 patients with benign and malignant thyroid lesions. Total FAK expression provided a distinct discrimination between malignant and benign (p=0.00001), as well as between papillary carcinoma and hyperplastic nodules thyroid lesions (p=0.00005), being also associated with follicular cells’ proliferative capacity (p=0.0003). In malignant thyroid lesions, total FAK expression was associated with tumor size (p=0.0455), and presence of capsular (p=0.0102) and lymphatic (p=0.0173) invasion. Total Src expression was borderline increased in cases of papillary carcinoma compared to hyperplastic nodules (p=0.0993), being also correlated with tumor size (p=0.0169). FAK and Src expression was ascribed to a significant extent to the phosphorylated forms of the enzymes, which provided a better discrimination between malignant and benign thyroid lesions. The current data revealed that FAK and to a lesser extent Src expression could be considered of clinical utility in thyroid neoplasia with potential use as therapeutic targets. C1 [Michailidi, Christina] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 M. Asias str., Goudi, GR11527 Athens, Greece. [Giaginis, Costas] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 M. Asias str., Goudi, GR11527 Athens, Greece. [Stolakis, Vassilios] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 M. Asias str., Goudi, GR11527 Athens, Greece. [Alexandrou, Paraskevi] University of Athens, Medical School, Department of PathologyAthens, Greece. [Klijanienko, Jerzy] Institut Curie, Department of PathologyParis, France. [Delladetsima, Ioanna] University of Athens, Medical School, Department of PathologyAthens, Greece. [Chatzizacharias, Nicolaos] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 M. Asias str., Goudi, GR11527 Athens, Greece. [Tsourouflis, Gerasimos] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 M. Asias str., Goudi, GR11527 Athens, Greece. [Theocharis, Stamatios] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 M. Asias str., Goudi, GR11527 Athens, Greece. RP Theocharis, S (reprint author), University of Athens, Medical School, Department of Forensic Medicine and Toxicology, GR11527 Athens, Greece. EM theocharis@ath.forthnet.gr CR Lipfert L, Haimovich B, Schaller MD et al, 1992, Integrindependent phosphorylation and activation of the protein tyrosine kinase pp 125FAK in platelets. J Cell Biol 119:905–912 Zachary I, Sinnett-Smith J, Rozengurt E, 1992, Bombesin, vasopressin, and endothelin stimulation of tyrosine phosphorylation in Swiss 3T3 cells. Identification of a novel tyrosine kinase as a major substrate. J Biol Chem 267:19031–19034 Ilic D, Furuta Y, Kanazawa S et al, 1995, Reduced cell motility and enhanced focal adhesion contact formation in cells from FAKdeficient mice. Nature 377:539–544 Cary LA, Chang JF, Guan JL, 1991, Stimulation of cell migration by overexpression of focal adhesion kinase and its association with Src and Fyn. J Cell Sci 109:1787–17894 Li S, Kim M, Hu YL et al, 1997, Fluid shear stress activation of focal adhesion kinase. Linking to mitogen-activated protein kinases. J Biol Chem 272:30455–30462 Xing Z, Chen HC, Nowlen JK et al, 1994, Direct interaction of v- Src with the focal adhesion kinase mediated by the Src SH2 domain. Mol Biol Cell 5:413–421 Rahimi N, Hung W, Tremblay E et al, 1998, c-Src kinase activity is required for hepatocyte growth factor-induced motility and anchorage-independent growth of mammary carcinoma cells. J Biol Chem 273:33714–33721 Zachary I, 1997, Focal adhesion kinase. Int J Biochem Cell Biol 29:929–934 Cary LA, Guan JL, 1999, Focal adhesion kinase in integrinmediated signaling. Front Biosci 4:D102–D113 Schlaepfer DD, Hauck CR, Stieg DJ, 1999, Signaling through focal adhesion kinase. Prog Biophys Mol Biol 71:435–478 Cox BD, Natarajan M, Stettner MR et al, 2006, New concepts regarding focal adhesion kinase promotion of cell migration and proliferation. J Cell Biochem 99:35–52 Laird AD, Cherrington JM, 2003, Small molecule tyrosine kinase inhibitors: clinical development of anticancer agents. Expert Opin Investig Drugs 12:51–64 Chatzizacharias NA, Kouraklis GP, Theocharis SE, 2007, Focal adhesion kinase: a promising target for anticancer therapy. Expert Opin Ther Targets 11:1315–1328 Talamonti MS, Roh MS, Curley SA et al, 1993, Increase in activity and level of pp 60c-src in progressive stages of human colorectal cancer. J Clin Invest 91:53–60 Lutz MP, Esser IB, Flossmann-Kast BB et al, 1998, Overexpression and activation of the tyrosine kinase Src in human pancreatic carcinoma. Biochem Biophys Res Commun 243:503–508 Verbeek BS, Vroom TM, Adriaansen-Slot SS et al, 1996, c-Src protein expression is increased in human breast cancer. An immunohistochemical and biochemical analysis. J Pathol 180:383–388 Ito H, Gardner-Thorpe J, Zinner MJ et al, 2003, Inhibition of tyrosine kinase Src suppresses pancreatic cancer invasiveness. Surgery 134:221–226 Myoui A, Nishimura R, Williams PJ et al, 2003, C-SRC tyrosine kinase activity is associated with tumor colonization in bone and lung in an animal model of human breast cancer metastasis. Cancer Res 63:5028–5033 Duxbury MS, Ito H, Zinner MJ et al, 2004, Inhibition of SRC tyrosine kinase impairs inherent and acquired gemcitabine resistance in human pancreatic adenocarcinoma cells. Clin Cancer Res 10:2307–2318 Chatzizacharias NA, Kouraklis GP, Theocharis SE, 2008, Clinical significance of FAK expression in human neoplasia. Histol Histopathol 23:629–650 Gharib H, Papini E, 2007, Thyroid nodules: clinical importance, assessment, and treatment. Endocrinol Metab Clin N Am 36:707–735 Gharib H, Papini E, Paschke R, 2008, Thyroid nodules: a review of current guidelines, practices, and prospects. Eur J Endocrinol 159:493–505 Roberts SS, Mendonca-Torres MC, Jensen K, Francis GL, Vasko V, 2009, GABA receptor expression in benign and malignant thyroid tumors. Pathol Oncol Res., DOI 10.1007/s12253-009-9165-x Ersoz S, Sert H, Yandi M, Erem C, Mungan S, Ersoz HO, Cobanoglu U, Hacihasanoglu A, 2009, The significance of galectin-3 expression in the immunocytochemical evaluation of thyroid fine needle aspiration cytology. Pathol Oncol Res., DOI 10.1007/s12253-008-9041-0 Fisher DA, 2007, Thyroid function and dysfunction in premature infants. Pediatr Endocrinol Rev 4:317–328 Owens LV, Xu L, Dent GA et al, 1996, Focal adhesion kinase as a marker of invasive potential in differentiated human thyroid cancer. Ann Surg Oncol 3:100–105 Kim SJ, Park JW, Yoon JS et al, 2004, Increased expression of focal adhesion kinase in thyroid cancer: immunohistochemical study. J Korean Med Sci 19:710–715 Rothhut B, Ghoneim C, Antonicelli F et al, 2007, Epidermal growth factor stimulates matrix metalloproteinase-9 expression and invasion in human follicular thyroid carcinoma cells through Focal adhesion kinase. Biochimie 89:613–624 Rosai J, 2004, Appendix C. Staging of cancer. In: Houston M, ed, Rosai and Ackerman’s surgical pathology, 9th edn. London, Mosby, pp 2809–2810 Giaginis CT, Vgenopoulou S, Tsourouflis GS et al, 2009, Expression and clinical significance of focal adhesion kinase in the two distinct histological types, intestinal and diffuse, of human gastric adenocarcinoma. Pathol Oncol Res., DOI 10.1007/s12253- 008-9120-2 Giaginis CT, Zarros AC, Papaefthymiou MA et al, 2008, Clinical significance of tumor-associated antigen RCAS1 expression in human pancreatic ductal adenocarcinoma. Dig Dis Sci 53:1728–1734 Giaginis C, Zarros A, Alexandrou P et al, 2009, Evaluation of Coxsackievirus and Adenovirus Receptor, CAR, expression in human benign and malignant thyroid lesions. AMPIS Accepted Finn RS, 2008, Targeting Src in breast. Ann Oncol 19:1379–1386 Han EK, McGonigal T, 2007, Role of focal adhesion kinase in human cancer: a potential target for drug discovery. Anticancer Agents Med Chem 7:681–684 Chatzizacharias NA, Kouraklis G, Theocharis S, 2008, Disruption of FAK signaling: a side mechanism in cytotoxicity. Toxicology 245:1–10 Cao X, You QD, Li ZY et al, 2008, Recent progress of SRC family kinase inhibitors as anticancer agents. Mini Rev Med Chem 8:1053–1063 Miyaki T, Kato H, Nakajima M et al, 2003, FAK overexpression is correlated with tumor invasiveness and lymph node metastasis in oesophageal squamous cell carcinoma. Br J Cancer 89:140–145 Lark AL, Livasy CA, Dressler L et al, 2005, High focal adhesion kinase expression in invasive breast carcinomas is associated with an aggressive phenotype. Mod Pathol 18:1289–1294 Aguirre Ghiso JA, 2002, Inhibition of FAK signaling activated by urokinase receptor induces dormancy in human carcinoma cells in vivo. Oncogene 21:2513–2524 Furuyama K, Ryuchiro D, Tomohiko M et al, 2006, Clinical significance of focal adhesion kinase in resectable pancreatic cancer. World J Surg 30:219–226 Fuji T, Koshikawa K, Nomoto S et al, 2004, Focal adhesion kinase is overexpressed in hepatocellular carcinoma and can be served as an independent prognostic factor. J Hepatol 41:104–111 Ohta R,Yamashita Y, Taketomi A et al, 2006, Reduced expression of focal adhesion kinase in intrahepatic cholangiocarcinoma is associated with poor tumor differentiation. Oncology 71:417–422 Carelli S, Zadra G, Vaira V et al, 2006, Up-regulation of focal adhesion kinase in non-small cell lung cancer. Lung Cancer 53:263–271 Wang D, Grammer JR, Cobbs CS et al, 2000, p125 Focal adhesion kinase promotes malignant astrocytoma cell proliferation in vitro. J Cell Sci 113:4221–4230 Gabarra-Niecko V, Schaller MD, Dunty JM, 2003, FAK regulates biological processes important for the pathogenesis of cancer. Cancer Metastasis Rev 22:359–374 Sood AK, Coffin JE, Schneider GB et al, 2004, Biological significance of focal adhesion kinase in ovarian cancer: role in migration and invasion. Am J Pathol 165:1087–1095 Yu HG, Tong SL, Ding YM et al, 2006, Enhanced expression of cholecystokinin-2 receptor promotes the progression of colon cancer through activation of focal adhesion kinase. Int J Cancer 119:2724–2732 Ito Y, Kawakatsu H, Takeda T et al, 2002, Activation of c-Src is inversely correlated with biological aggressiveness of breast carcinoma. Breast Cancer Res Treat 76:261–267 Ito Y, Kawakatsu H, Takeda T et al, 2001, Activation of c-Src gene product in hepatocellular carcinoma is highly correlated with the indices of early stage phenotype. J Hepatol 35:68–73 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 497 EP 507 DI 10.1007/s12253-010-9269-3 PG 11 ER PT J AU Huang, Q Xia, Z You, Y Pu, P AF Huang, Qiang Xia, Zhibo You, Yongping Pu, Peiyu TI Wild Type p53 Gene Sensitizes Rat C6 Glioma Cells to HSV-TK/ACV Treatment In Vitro and In Vivo SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; wt-p53; HSV-TK/ACV; Suicide gene therapy; Combined gene therapy ID Glioma; wt-p53; HSV-TK/ACV; Suicide gene therapy; Combined gene therapy AB Suicide gene therapy using herpes simplex virusthymidine kinase (HSV-TK)/ganciclovir (GCV), has been extensively tested for the treatment of glioma. Our previous study showed that exogenous wild type p53 (wt-p53) enhanced the anti-tumor effect of HSV-TK/GCV therapy. However, the use of GCV is hindered by its low penetration to the brain and its toxity when used at higher dose. In the present study, we used another pro-drug, acyclovir (ACV), and examined the therapeutic efficacy of HSV-TK/ACV combining with wt-p53 in C6 glioma cells. We observed that wt-p53 combined with HSV-TK/ACV resulted in the super-additive anti-tumor effect in vitro. Exogenous wt-p53 significantly enhanced the sensitivity of TK positive C6 cells to ACV in vitro. Our in vivo experiment demonstrated that the effect of wt-p53 and HSV-TK/ACV combination therapy was better than that of HSV-TK/ACV alone. The survival time of tumor-bearing rats treated with wt-p53 in combination with HSV-TK/ACV was also significantly prolonged than those treated with HSV-TK/ACV alone. These results suggest that wt-p53 can enhance the therapeutic efficacy of HSV-TK/ACV both in vitro and in vivo. These findings are considerably valuable with the respect of using less toxic ACV as prodrug. This novel strategy could provide benefit to HSV-TK/prodrug gene therapy. C1 [Huang, Qiang] Tianjin Medical University General Hospital, Department of Neurosurgery, 154 Anshan Road, 300052 Tianjin, China. [Xia, Zhibo] The First Affiliated Hospital of Sun Yat-sen University, Department of Neurosurgery, 58 Zhongshan Road II, 510080 Guangzhou, Guangdong, China. [You, Yongping] The First Affiliated Hospital of Nanjing Medical University, Department of Neurosurgery, 210029 Nanjing, China. [Pu, Peiyu] Tianjin Medical University General Hospital, Department of Neurosurgery, 154 Anshan Road, 300052 Tianjin, China. RP Pu, P (reprint author), Tianjin Medical University General Hospital, Department of Neurosurgery, 300052 Tianjin, China. EM pupeiyu33@hotmail.com CR Faulds D, Heel RC, 1990, Ganciclovir. A review of its antiviral activity, pharmacokinetic properties and therapeutic efficacy in ytomegalovirus infection. Drugs 39:5997–6331 Elion GB, 1982, Mechanism of action and selectivity of acyclovir. Am J Med 3:7–13 Fyfe JA, Keller PM, Furman PA et al, 1978, Thymidine kinase from herpes simplex virus phosphorylates the new antiviral compound 9-(2-hydroxyethoxymethyl, guanine. J Biol Chem 253:8721–8727 Touraine RL, Vahanian N, Ramsey WJ et al, 1998, Enhancement of the herpes simplex virus thymidine kinase/ganciclovir bystander effect and its antitumor efficacy in vivo by pharmacologic manipulation of gap junctions, Hum. Gene Ther 9:2385–2391 Wagstaff AJ, Faulds D, Goa KL et al, 1994, Aciclovir. A reappraisal of its antiviral activity, pharmacokinetic properties and therapeutic efficacy, Drugs 47:153–205 Brewster ME, Raghavan K, Pop E et al, 1994, Enhanced delivery of ganciclovir to the brain through the use of redox targeting. Antimicrob, Agents Chemother 38:817–823 Balzarini J, Bohman C, De Clercq E et al, 1993, Differential mechanism of cytostatic effect of, E)-5-(2-bromovinyl)-29- deoxyuridine, 9-(1, 3-dihydroxy-2-propoxymethyl, guanine, and other antiherpetic drugs on tumor cells transfected by the thymidine kinase gene of herpes simplex virus type 1 or type 2. J Biol Chem 268:6332–6337 Huang Q, Pu PY, Xia ZB et al, 2007, Exogenous wt-p53 enhances the antitumor effect of HSV-TK/GCV on C6 glioma cells. J Neurooncol 82:239–248 Pu PY, Liu XW, Liu AX et al, 2000, Inhibitory effect of antisense epidermal growth factor receptor RNA on the proliferation of rat C6 glioma cells in vitro and in vivo. J Neurosurg 92:132–139 Rosolen AR, Frascella E, di Francesco C et al, 1998, In vitro and in vivo antitumor effect of retrovirus-mediated herpes simplex thymidine kinase gene-transfer in human medulloblastoma. Gene Ther 5:113–120 Maron A, Gustin T, Le Roux A et al, 1996, Gene therapy of rat C6 glioma using adenovirus-mediated transfer of the HSV-tk gene: long-term follow-up by magnetic resonance imaging. Gene Ther 3:315–322 Fulci G, Chiocca EA, 2007, The status of gene therapy for brain tumors. Expert Opin Biol Ther 7:197–208 Reichman HR, Farrell CL, Del Maestro RF et al, 1986, Effects of steroids and nonsteroid anti-inflammatory agents on vascular permeability in a rat glioma model. J Neurosurg 65:233–237 LeMay DR, Kittaka M, Gordon EM et al, 1998, Intravenous RMP-7 increases delivery of ganciclovir into rat brain tumors and enhances the effects of herpes simplex virus thymidine kinase gene therapy, Hum. Gene Ther 9:989–995 Terao S, Shirakawa T, Goda K et al, 2005, Recombinant interleukin-2 enhanced the antitumor effect of ADV/RSV-HSVtk/ACV therapy in a murine bladder cancer model. Anticancer Res 25:2757–2760 Hayashi K, Lee JB, Maitani Y et al, 2006, The role of a HSV thymidine kinase stimulating substance, scopadulciol, in improving the efficacy of cancer gene therapy. J Gene Med 8:1056–1067 Hayashi K, Hayashi T, Sun HD et al, 2002, Contribution of a combination of ponicidin and acyclovir/ganciclovir to the antitumor efficacy of the herpes simplex virus thymidine kinase gene therapy system. Hum Gene Ther 13:415–423 Beltinger C, Fulda S, Kammertoens T et al, 1999, Herpes simplex virus thymidine kinase/ ganciclovir-induced apoptosis involves ligand-independent death receptor aggregation and activation of caspases. Proc Natl Acad Sci USA 96:8699–8704 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 509 EP 514 DI 10.1007/s12253-009-9240-3 PG 6 ER PT J AU Ren, HZ Wang, JSh Wang, P Pan, Gq Wen, JF Fu, H Shan, Xz AF Ren, Hong-Zheng Wang, Jin-Sheng Wang, Peng Pan, Guo-qing Wen, Ji-Fang Fu, Hua Shan, Xu-zheng TI Increased Expression of Prohibitin and its Relationship with Poor Prognosis in Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prohibitin; Esophageal squamous cell carcinoma; Prognosis; Immunohistochemistry ID Prohibitin; Esophageal squamous cell carcinoma; Prognosis; Immunohistochemistry AB Prohibitin, a potential tumor suppressor, has been shown to be an anti- proliferative protein, a regulator of cell-cycle progression and in apoptosis. Recently, it was found to be over-expressed in breast cancer and gastric cancer, and it has been suggested as a biomarker in those diseases. To clarify the role and the prognostic significance of prohibitin expression in esophageal squamous cell carcinoma (ESCC), we analyzed the expression in ESCC and their corresponding nonneoplastic epithelia tissues by immunohistochemistry(IHC), Western blotting and realtime quantitative reverse transcription polymerase chain reaction(QRT-PCR).The relationship between prohibitin expression and clinicopathological variables was examined by statistical analysis. The findings suggested the upregulation of prohibitin play an important role in the carcinogenesis of ESCC. The over-expression of prihibitin was significantly correlated with the depth of tumor, lymph node metastasis, distant metastasis, lymphatic invasion and vascular invasion of ESCC. These results suggested that prohibitin(+), lymph node metastasis and distant metastasis could be the independent risk factors for worse prognosis in ESCC patients. C1 [Ren, Hong-Zheng] Central South University, Xiangya Basic Medical College, Department of PathologyChangsha, China. [Wang, Jin-Sheng] Changzhi Medical College, Department of PathologyChangzhi, China. [Wang, Peng] Central South University, Xiangya Basic Medical College, Department of PathologyChangsha, China. [Pan, Guo-qing] Central South University, Xiangya Basic Medical College, Department of PathologyChangsha, China. [Wen, Ji-Fang] Central South University, Xiangya Basic Medical College, Department of PathologyChangsha, China. [Fu, Hua] Central South University, Xiangya Basic Medical College, Department of PathologyChangsha, China. [Shan, Xu-zheng] Central South University, Public Health College, Department of Health Statistics and EpidemiologyChangsha, China. RP Wen, JF (reprint author), Central South University, Xiangya Basic Medical College, Department of Pathology, Changsha, China. EM jifangwen@hotmail.com CR Vizcaino AP, Moreno V, Lambert R, Parkin DM, 2002, Time trends incidence of both major histologic types of esophageal carcinomas in selected countries, 1973–1995. Int J Cancer 99, 6):860–868 Stoner GD, Gupta A, 2001, Etiology and chemoprevention of esophageal squamous cell carcinoma. Carcinogenesis 22, 11):1737–1746 Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55(2):74–108 Piper PW, Bringloe D, 2002, Loss of prohibitins, though it shortens the replicative life span of yeast cells undergoing division, does not shorten the chronological life span of G0- arrested cells. Mech Ageing Dev 123(4):287–295 Dell’Orco RT, McClung JK, Jupe ER, Liu XT, 1996, Prohibitin and the senescent phenotype. Exp Gerontol 31(1–2):245–252 Jupe ER, Liu XT, Kiehlbauch JL, McClung JK, Dell’Orco RT, 1996, The 39 untranslated region of prohibitin and cellular immortalization. Exp Cell Res 224(1):128–135 Kang X, Zhang L, Sun J et al, 2008, Prohibitin: a potential biomarker for tissue-based detection of gastric cancer. J Gastroenterol 43(8):618–625 Dell’Orco RT, Jupe ER, Manjeshwar S et al, 1997, Prohibitin: a new biomarker for breast tumors. Breast J 3:85–89 Greene FL, Page DL, Fleming ID et al, 2002, AJCC cancer staging manual, 6th edn. Springer-Verlag, New York Kononen J, Bubendorf L, Kallioniemi A et al, 1998, Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4(7):844–847 Gastl G, Spizzo G, Obrist P, Dunser M, Mikuz G, 2000, Ep-CAM overexpression in breast cancer as a predictor of survival. Lancet 356(9246):1981–1982 Pancione M, Forte N, Sabatino L et al, 2009, Reduced beta-catenin and peroxisome proliferator-activated receptor-gamma expression levels are associated with colorectal cancer metastatic progression: correlation with tumor-associated macrophages, cyclooxygenase 2, and patient outcome. Hum Pathol 40(5):714–725 Allen JW, Richardson JD, Edwards MJ, 1997, Squamous cell carcinoma of the esophagus: a review and update. Surg Oncol 6, 4):193–200 Nijtmans LG, de Jong L, Artal Sanz M et al, 2000, Prohibitins act as a membrane-bound chaperone for the stabilization of mitochondrial proteins. EMBO J 19(11):2444–2451 Coates PJ, Nenutil R, McGregor A et al, 2001, Mammalian PHB proteins respond to mitochondrial stress and decrease during cellular senescence. Exp Cell Res 265(2):262–273 McClung JK, Jupe ER, Liu XT, Dell’Orco RT, 1995, Prohibitin: potential role in senescence, development, and tumor suppression. Exp Gerontol 30(2):99–124 Ikonen E, Fiedler K, Parton RG, Simons K, 1995, Prohibitin, an antiproliferative protein, is localized to mitochondria. FEBS Lett 358(3):273–277 Wang S, Nath N, Adlam M, Chellappan S, 1999, Prohibitin, a potential tumor suppressor, interacts with RB and regulates E2F function. Oncogene 18(23):3501–3510 Wang KJ, Wang RT, Zhang JZ, 2004, Identification of tumor markers using two- dimensional electrophoresis in gastric carcinoma. World J Gastroenterol 10(15):2179–2183 Ummanni R, Junker H, Zimmermann U et al, 2008, Prohibitin identified by proteomic analysis of prostate biopsies distinguishes hyperplasia and cancer. Cancer Lett 266(2): 171–185 Asamoto M, Cohen SM, 1994, Prohibitin gene is overexpressed but not mutated in rat bladder carcinomas and cell lines. Cancer Lett 83(1–2):201–207 Jupe ER, Liu XT, Kiehlbauch JL, McClung JK, Dell’Orco RT, 1996, Prohibitin in breast cancer cell lines: loss of antiproliferative activity is linked to 3′untranslated region mutations. Cell Growth Differ 7(7):871–878 Pickens A, Orringer MB, 2003, Geographical distribution and racial disparsity in esophageal cancer. Ann Thorac Surg 76(4): S1367–1369 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 515 EP 522 DI 10.1007/s12253-009-9242-1 PG 8 ER PT J AU Yamada, Shi Yanamoto, S Kawasaki, G Mizuno, A Nemoto, KT AF Yamada, Shin-ichi Yanamoto, Souichi Kawasaki, Goro Mizuno, Akio Nemoto, K Takayuki TI Overexpression of Cortactin Increases Invasion Potential in Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cortactin; Invasion; Metastasis; RNA interference; Oral squamous cell carcinoma ID Cortactin; Invasion; Metastasis; RNA interference; Oral squamous cell carcinoma AB Cortactin, an F-actin binding protein, stabilizes F-actin networks and promotes actin polymerization by activating the Arp2/3 complex. Overexpression of cortactin has been reported in several human cancers. Cortactin stimulates cell migration, invasion, and experimental metastasis. However, the underlying mechanism is not still understood. In the present study, we therefore evaluated the possibility that cortactin could be appropriate as a molecular target for cancer gene therapy. In 70 primary oral squamous cell carcinomas and 10 normal oral mucosal specimens, cortactin expression was evaluated by immunological analyses, and the correlations of the overexpression of cortactin with clinicopathologic factors were evaluated. Overexpression of cortactin was detected in 32 of 70 oral squamous cell carcinomas; significantly more frequently than in normal oral mucosa. Cortactin overexpression was more frequent in higher grade cancers according to T classification, N classifications, and invasive pattern. Moreover, RNAi-mediated decrease in cortactin expression reduced invasion. Downregulation of cortactin expression increased the expression levels of E-cadherin, β-catenin, and EpCAM. The siRNA of cortactin also reduced PTHrP expression via EGF signaling. These results consistently indicate that the overexpression of cortactin is strongly associated with an aggressive phenotype of oral squamous cell carcinoma. In conclusion, we propose that cortactin could be a potential molecular target of gene therapy by RNAi targeting in oral squamous cell carcinoma. C1 [Yamada, Shin-ichi] Nagasaki University Graduate School of Biomedical Sciences, Department of Regenerative Oral Surgery, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yanamoto, Souichi] Nagasaki University Graduate School of Biomedical Sciences, Department of Regenerative Oral Surgery, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Kawasaki, Goro] Nagasaki University Graduate School of Biomedical Sciences, Department of Regenerative Oral Surgery, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Mizuno, Akio] Nagasaki University Graduate School of Biomedical Sciences, Department of Regenerative Oral Surgery, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Nemoto, K Takayuki] Nagasaki University Graduate School of Biomedical Sciences, Unit of Basic Medical Sciences, Course of Medical and Dental Sciences, Department of Oral Molecular Biology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. RP Yamada, Shi (reprint author), Nagasaki University Graduate School of Biomedical Sciences, Department of Regenerative Oral Surgery, 852-8588 Nagasaki, Japan. EM shinshin@nagasaki-u.ac.jp CR Mao L, Hong WK, Papadimitrakopoulou VA, 2004, Focus on head and neck cancer. Cancer Cell 5:311–316 Hicks WL Jr, North JH Jr, Loree TR et al, 1998, Surgery as a single modality therapy for squamous cell carcinoma of the oral tongue. Am J Otolaryngol 19:24–28 Sessions DG, Lenox J, Spector GJ et al, 2003, Analysis of treatment results for base of tongue cancer. Laryngoscope 113:1252–1261 Gonzalez-Garcia R, Naval-Gias L, Rodriguez-Campo FJ et al, 2008, Contralateral lymph neck node metastasis of squamous cell carcinoma of the oral cavity: a retrospective analytic study in 315 patients. J Oral Maxillofac Surg 66:1390–1398 Ziober BL, Silverman SS Jr, Kramer RH, 2001, Adhesive mechanisms regulating invasion and metastasis in oral cancer. Crit Rev Oral Biol Med 12:499–510 Bankfalvi A, Krassort M, Buchwalow IB et al, 2002, Gains and losses of adhesion molecules, CD44, E-cadherin, and betacatenin, during oral carcinogenesis and tumour progression. J Pathol 198:343–351 Arora S, Kaur J, Sharma C et al, 2005, Stromelysin 3, Ets-1, and vascular endothelial growth factor expression in oral precancerous and cancerous lesions: correlation with microvessel density, progression, and prognosis. Clin Cancer Res 15(11):2272–2284 Yanamoto S, Kawasaki G, Yoshitomi I et al, 2007, Clinicopathologic significance of EpCAM expression in squamous cell carcinoma of the tongue and its possibility as a potential target for tongue cancer gene therapy. Oral Oncol 43:869–877 Lin M, Smith LT, Smiraglia DJ et al, 2006, DNA copy number gains in head and neck squamous cell carcinoma. Oncogene 25:1424–1433 Hui AB, Or YY, Takano H et al, 2005, Array-based comparative genomic hybridization analysis identified cyclin D1 as a target oncogene at 11q13.3 in nasopharyngeal carcinoma. Cancer Res 65:8125–8133 Zaharieva BM, Simon R, Diener PA et al, 2003, High-throughput tissue microarray analysis of 11q13 gene amplification, CCND1, FGF3, FGF4, EMS1, in urinary bladder cancer. J Pathol 201:603– 608 Meredith SD, Levine PA, Burns JA et al, 1995, Chromosome 11q13 amplification in head and neck squamous cell carcinoma. Association with poor prognosis. Arch Otolaryngol Head Neck Surg 121:790–794 Schuuring E, 1995, The involvement of the chromosome 11q13 region in human malignancies: cyclin D1 and EMS1 are two new candidate oncogenes—a review. Gene 159:83–96 Merritt WD, Weissler MC, Turk BF et al, 1990, Oncogene amplification in squamous cell carcinoma of the head and neck. Arch Otolaryngol Head Neck Surg 116:1394–1398 Leonard JH, Kearsley JH, Chenevix-Trench G et al, 1991, Analysis of gene amplification in head-and-neck squamous-cell carcinoma. Int J Cancer 48:511–515 Williams ME, Gaffey MJ, Weiss LM et al, 1993, Chromosome 11Q13 amplification in head and neck squamous cell carcinoma. Arch Otolaryngol Head Neck Surg 119:1238–1243 Callender T, el-Naggar AK, Lee MS et al, 1994, PRAD-1, CCND1)/cyclin D1 oncogene amplification in primary head and neck squamous cell carcinoma. Cancer 74:152–158 Fortin A, Guerry M, Guerry R et al, 1997, Chromosome 11q13 gene amplifications in oral and oropharyngeal carcinomas: no correlation with subclinical lymph node invasion and disease recurrence. Clin Cancer Res 3:1609–1614 Akervall JA, Michalides RJ, Mineta H et al, 1997, Amplification of cyclin D1 in squamous cell carcinoma of the head and neck and the prognostic value of chromosomal abnormalities and cyclin D1 overexpression. Cancer 79:380–389 Alavi S, Namazie A, Calcaterra TC et al, 1999, Clinical application of fluorescence in situ hybridization for chromosome 11q13 analysis in head and neck cancer. Laryngoscope 109:874– 879 Rodrigo JP, Garcia LA, Ramos S et al, 2000, EMS1 gene amplification correlates with poor prognosis in squamous cell carcinomas of the head and neck. Clin Cancer Res 6:3177–3182 Xia J, Chen Q, Li B et al, 2007, Amplifications of TAOS1 and EMS1 genes in oral carcinogenesis: association with clinicopathological features. Oral Oncol 43:508–514 Freier K, Sticht C, Hofele C et al, 2006, Recurrent coamplification of cytoskeleton-associated genes EMS1 and SHANK2 with CCND1 in oral squamous cell carcinoma. Genes Chromosomes Cancer 45:118–125 Muller D, Millon R, Lidereau R et al, 1994, Frequent amplification of 11q13 DNA markers is associated with lymph node involvement in human head and neck squamous cell carcinomas. Eur J Cancer B Oral Oncol 30:113–120 Muller D, Millon R, Velten M et al, 1997, Amplification of 11q13 DNA markers in head and neck squamous cell carcinomas: correlation with clinical outcome. Eur J Cancer 33:2203–2210 Takes RP, Baatenburg de Jong RJ, Schuuring E et al, 1997, Markers for assessment of nodal metastasis in laryngeal carcinoma. Arch Otolaryngol Head Neck Surg 123:412–419 Rodrigo JP, Garcia LA, Ramos S et al, 2000, EMS1 gene amplification correlates with poor prognosis in squamous cell carcinomas of the head and neck. Clin Cancer Res 6:3177– 3182 Schuuring E, Verhoeven E, Mooi WJ et al, 1992, Identification and cloning of two overexpressed genes, U21B31/PRAD1 and EMS1, within the amplified chromosome 11q13 region in human carcinomas. Oncogene 7:355–361 Fantl V, Smith R, Brookes S et al, 1993, Chromosome 11q13 abnormalities in human breast cancer. Cancer Surv 18:77–94 Karlseder J, Zeillinger R, Schneeberger C et al, 1994, Patterns of DNA amplification at band q13 of chromosome 11 in human breast cancer. Genes Chromosomes Cancer 9:42–48 Peters G, Fantl V, Smith R et al, 1995, Chromosome 11q13 markers and D-type cyclins in breast cancer. Breast Cancer Res Treat 33:125–135 Weed SA, Parsons JT, 2001, Cortactin: coupling membrane dynamics to cortical actin assembly. Oncogene 20:6418–6434 Daly RJ, 2004, Cortactin signalling and dynamic actin networks. Biochem J 382:13–25 Xiao-Ping H, Tie-Hua R, Peng L et al, 2006, Cyclin D1 overexpression in esophageal cancer from southern China and its clinical significance. Cancer Lett 231:94–101 Bringuier PP, Tamimi Y, Schuuring E et al, 1996, Expression of cyclin D1 and EMS1 in bladder tumours; relationship with chromosome 11q13 amplification. Oncogene 12:1747–1753 Rothschild BL, Shim AH, Ammer AG et al, 2006, Cortactin overexpression regulates actin-related protein 2/3 complex activity, motility, and invasion in carcinomas with chromosome 11q13 amplification. Cancer Res 66:8017–8025 Luo ML, Shen XM, Zhang Y et al, 2006, Amplification and overexpression of CTTN, EMS1, contribute to the metastasis of esophageal squamous cell carcinoma by promoting cell migration and anoikis resistance. Cancer Res 66:11690–11699 Hsu NY, Yeh KT, Chiang IP et al, 2008, Cortactin overexpression in the esophageal squamous cell carcinoma and its involvement in the carcinogenesis. Dis Esophagus 21:402–408 Strewler GJ, Nissenson RA, 1990, Hypercalcemia in malignancy. West J Med 153:635–640 Strewler GJ, 2000, The physiology of parathyroid hormonerelated protein. N Engl J Med 342:177–185 Yamada T, Tsuda M, Ohba Y et al, 2008, PTHrP promotes malignancy of human oral cancer cell downstream of the EGFR signaling. Biochem Biophys Res Commun 368:575–581 Lua BL, Low BC, 2005, Cortactin phosphorylation as a switch for actin cytoskeletal network and cell dynamics control. FEBS Lett 579:577–585 Patel AS, Schechter GL, Wasilenko WJ et al, 1998, Overexpression of EMS1/cortactin in NIH3T3 fibroblasts causes increased cell motility and invasion in vitro. Oncogene 16:3227– 3232 Li Y, Tondravi M, Liu J et al, 2001, Cortactin potentiates bone metastasis of breast cancer cells. Cancer Res 61:6906–6911 Coopman PJ, Do MT, Thompson EW et al, 1998, Phagocytosis of cross-linked gelatin matrix by human breast carcinoma cells correlates with their invasive capacity. Clin Cancer Res 4:507– 515 Chen WT, 1989, Proteolytic activity of specialized surface protrusions formed at rosette contact sites of transformed cells. J Exp Zool 251:167–185 Wu H, Parsons JT, 1993, Cortactin, an 80/85-kilodalton pp 60src substrate, is a filamentous actin-binding protein enriched in the cell cortex. J Cell Biol 120:1417–1426 Helwani FM, Kovacs EM, Paterson AD et al, 2004, Cortactin is necessary for E-cadherin-mediated contact formation and actin reorganization. J Cell Biol 164:899–910 Kovacs EM, Goodwin M, Ali RG et al, 2002, Cadherin-directed actin assembly: E-cadherin physically associates with the Arp2/3 complex to direct actin assembly in nascent adhesive contacts. Curr Biol 12:379–382 Weed SA, Karginov AV, Schafer DA et al, 2000, Cortactin localization to sites of actin assembly in lamellipodia requires interactions with F-actin and the Arp2/3 complex. J Cell Biol 151:29–40 Weaver AM, Karginov AV, Kinley AW et al, 2001, Cortactin promotes and stabilizes Arp2/3-induced actin filament network formation. Curr Biol 11:370–374 El Sayegh TY, Arora PD, Laschinger CA et al, 2004, Cortactin associates with N-cadherin adhesions and mediates intercellular adhesion strengthening in fibroblasts. J Cell Sci 117:5117–5131 Hoschuetzky H, Aberle H, Kemler R, 1994, Beta-catenin mediates the interaction of the cadherin-catenin complex with epidermal growth factor receptor. J Cell Biol 127:1375–1380 Balzar M, Prins FA, Bakker HA et al, 1994, The structural analysis of adhesions mediated by Ep-CAM. Exp Cell Res 246:108–121 Winter MJ, Nagelkerken B, Mertens AE et al, 2003, Expression of Ep-CAM shifts the state of cadherin-mediated adhesions from strong to weak. Exp Cell Res 285:50–58 Todd R, Wong DT, 1999, Epidermal growth factor receptor, EGFR, biology and human oral cancer. Histol Histopathol 14:491–500 Rubin Grandis J, Melhem MF, Gooding WE et al, 1998, Levels of TGF-alpha and EGFR protein in head and neck squamous cell carcinoma and patient survival. J Natl Cancer Inst 90:824–832 Timpson P, Lynch DK, Schramek D et al, 2005, Cortactin overexpression inhibits ligand-induced down-regulation of the epidermal growth factor receptor. Cancer Res 65:3273–3280 Burtis WJ, Brady TG, Orloff JJ et al, 1990, Immunochemical characterization of circulating parathyroid hormone-related protein in patients with humoral hypercalcemia of cancer. N Engl J Med 322:1106–1112 Liao J, McCauley LK, 2006, Skeletal metastasis: established and emerging roles of parathyroid hormone related protein, PTHrP). Cancer Metastasis Rev 25:559–571 Tuschl T, 2002, Expanding small RNA interference. Nat Biotechnol 20:446–448 Kudo Y, Kitajima S, Ogawa I et al, 2005, Small interfering RNA targeting of S phase kinase-interacting protein 2 inhibits cell growth of oral cancer cells by inhibiting p27 degradation. Mol Cancer Ther 4:471–476 Yanamoto S, Iwamoto T, Kawasaki G et al, 2005, Silencing of the p53R2 gene by RNA interference inhibits growth and enhances 5-fluorouracil sensitivity of oral cancer cells. Cancer Lett 223:67–76 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 523 EP 531 DI 10.1007/s12253-009-9245-y PG 9 ER PT J AU Naidu, R Har, ChY Taib, AMN AF Naidu, Rakesh Har, Cheng Yip Taib, Aishah Mohd Nur TI Genetic Polymorphisms of Paraoxonase 1 (PON1) Gene: Association Between L55M or Q192R with Breast Cancer Risk and Clinico-Pathological Parameters SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Paraoxonase 1; Polymorphism ID Breast cancer; Paraoxonase 1; Polymorphism AB The aim of the present study was to evaluate the association between the paraoxonase 1 (PON1) L55M and Q192R polymorphisms and breast cancer risk as well as clinico-pathological characteristics of the patients. Genotyping of these polymorphisms was performed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method in a hospital-based Malaysian population. Peripheral blood samples were collected from 387 breast cancer patients and 252 normal and healthy women who had no history of any malignancy. The genotype (P=0.023) and allele (P=0.008) frequencies of L55M polymorphism were significantly different between the breast cancer cases and normal individuals. However, the distribution of genotype (P=0.333) and allele (P=0.163) frequencies of Q192R polymorphism showed lack of statistical significance. Women who were MM homozygotes (OR=2.229; 95% CI, 1.219–4.075) and carriers of M allele genotype (OR=1.429; 95% CI, 1.035–1.974) or M allele (OR=1.397; 95% CI, 1.093–1.785) were associated with increased risk of breast cancer. However, women who were heterozygous (OR=0.793; 95% CI, 0.567–1.110) or homozygous (OR=0.746; 95% CI, 0.407–1.370) for R allele or carriers of R allele (OR=0.838; 95%, 0.654–1.074) were not associated with breast cancer risk. The M allele genotype was significantly associated with estrogen receptor negativity (P=0.046) and nodal involvement (P=0.004) but R allele genotype was not associated with any of the clinico-pathological characteristics. In conclusion, our findings suggest that the polymorphic variant of L55M polymorphism could be a useful genetic marker for tumor prognosis and to identify women who might be at greater risk of developing breast cancer in a hospital-based Malaysian population. C1 [Naidu, Rakesh] Monash University, Sunway Campus, School of Medicine and Health Sciences, Jalan Lagoon Selatan, 46150 Bandar Sunway, Selangor Darul Ehsan, Malaysia. [Har, Cheng Yip] Universiti Kebangsaan Malaysia, Faculty of Medicine, Department of Surgery, 50603 Kuala Lumpur, Malaysia. [Taib, Aishah Mohd Nur] Universiti Kebangsaan Malaysia, Faculty of Medicine, Department of Surgery, 50603 Kuala Lumpur, Malaysia. RP Naidu, R (reprint author), Monash University, Sunway Campus, School of Medicine and Health Sciences, 46150 Bandar Sunway, Malaysia. EM kdrakeshna@hotmail.com CR Mackness B, Durrington PN, Mackness MI, 1998, Human serum paraoxonase. Gen Pharmacol 31:329–336 La Du BN, Adkins S, Kuo CL et al, 1993, Studies on human serum paraoxonase/arylesterase. Chem Biol Interact 87:25–34 Mackness MI, Mackness B, Durrington PN et al, 1998, Paraoxonase and coronary heart disease. Curr Opin Lipidol 9:319–324 Teiber JF, Billecke SS, La Du BN et al, 2007, Estrogen esters as substrates for human paraoxonases. Arch Biochem Biophys 461:24–29 Mackness MI, Arrol S, Mackness B et al, 1997, Alloenzymes of paraoxonase and effectiveness of high-density lipoproteins in protecting low-density lipoprotein against lipid peroxidation. Lancet 349:851–852 Humbert R, Adler DA, Disteche CM et al, 1993, The molecular basis of the human serum paraoxonase activity polymorphism. Nat Genet 3:73–76 Brophy VH, Jampsa RL, Clendenning JB et al, 2001, Effects of 5′ regulatory-region polymorphisms on paraoxonase-gene, PON1, expression. Am J Hum Genet 68:1428–1436 Aviram M, Hardak E, Vaya J et al, 2000, Human serum paraoxonases, PON1, Q and R selectively decrease lipid peroxides in human coronary and carotid atherosclerotic lesions: PON1 esterase and peroxidase-like activities. Circulation 101:2510–2517 Akcay MN, Yilmaz I, Polat MF et al, 2003, Serum paraoxonase levels in gastric cancer. Hepatogastroenterology 50:cclxxiii–v Akcay MN, Polat MF, Yilmaz I et al, 2003, Serum paraoxonase levels in pancreatic cancer. Hepatogastroenterology 50:ccxxv–vii Elkiran ET, Mar N, Aygen B et al, 2007, Serum paraoxonase and arylesterase activities in patients with lung cancer in a Turkish population. BMC Cancer 7:48 Gallicchio L, McSorley MA, Newschaffer CJ et al, 2007, Body mass, polymorphisms in obesity-related genes, and the risk of developing breast cancer among women with benign breast disease. Cancer Detect Prev 31:95–101 Stevens VL, Rodriguez C, Pavluck AL et al, 2006, Association of polymorphisms in the paraoxonase 1 gene with breast cancer incidence in the CPS-II Nutrition Cohort. Cancer Epidemiol Biomarkers Prev 15:1226–1228 Antognelli C, Buono CD, Ludovini V et al, 2009, YP17, GSTP1, PON1, and GLO1 gene polymorphisms as risk factors for breast cancer: an Italian case-control study. BMC Cancer 9:115–128 Lee CH, Lee KY, Choe KH et al, 2005, Effects of oxidative DNA damage induced by polycyclic aromatic hydrocarbons and genetic polymorphism of the paraoxonase-1, PON1, gene on lung cancer. J Prev Med Pub Health 38:345–350 Lurie G, Wilkens LR, Thompson PJ et al, 2008, Genetic polymorphisms in the paraoxonase 1 gene and risk of ovarian epithelial carcinoma. Cancer Epidemiol Biomarkers Prev 17:2070–2076 Antognelli C, Mearini L, Talesa VN et al, 2005, Association of CYP17, GSTP1, and PON1 polymorphisms with the risk of prostate cancer. Prostate 63:240–251 Kerridge I, Lincz L, Scorgie F et al, 2002, Association between xenobiotic gene polymorphisms and non-Hodgkin’s lymphoma risk. Br J Haematol 118:477–481 Lincz LF, Kerridge I, Scorgie FE et al, 2004, Xenobiotic gene polymorphisms and susceptibility to multiple myeloma. Haematologica 89:628–639 van der Logt EMJ, Janssen CHJM, van Hooijdonk Z et al, 2006, No association between genetic polymorphisms in NAD(P)H oxidase p22phox and Paraoxonase 1 and colorectal cancer risk. Anticancer Res 25:1465–1470 Kafadar AK, Ergen A, Zeybek U et al, 2006, Paraoxonase 192 gene polymorphism and serum paraoxonase activity in high grade gliomas and meningiomas. Cell Biochem Funct 24:455–460 Lim GCC, Halimah Y, 2004, Second Report of the National Cancer Registry. Cancer Incidence in Malaysia 2003. National Cancer Registry. Kuala Lumpur Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer I. Histopathology 19:403–410 Bloom HI, Richardson WW, 1957, Histological grading and prognosis. Br J Cancer 11:359–377 Breast. AJCC Cancer Staging Manual, 2002, In: American Joint Committee on Cancer. 6th ed. Springer, New York, pp 171–180 Singletary E, Allred C, Ashley P et al, 2002, Revision of the American Joint Committee on Cancer staging system for breast cancer. J Clin Oncol 20:3628–3636 Naidu R, Har YC, Taib NA, 2007, P27 V109G Polymorphism is associated with lymph node metastases but not with increased risk of breast cancer. J Exp Clin Cancer Res 26:133–140 Zama T, Murata M, Matsubara Y et al, 1997, A 192Arg Variant of the human paraoxonase, HUMPONA, gene polymorphism is associated with an increased risk for coronary artery disease in the Japanese. Arterioscler Thromb Vasc Biol 17:3565–3569 Naidu R, Har YC, Taib NA, 2009, Polymorphism of FGFR4 Gly388Arg does not confer an increased risk to breast cancer development. Oncol Res 18(2–3):65–71 Leviev I, Deakin S, James RW, 2001, Decreased stability of the M54 isoform of paraoxonase as a contributary factor to variations in human serum paraoxonase concentrations. J Lipid Res 42:528– 535 Brophy VH, Jarvik GP, Richter RJ et al, 2000, Analysis of paraoxonase, PON1, L55M status requires both genotype and phenotype. Pharmacogenetics 10:453–460 Durrington PN, Mackness B, Mackness MI, 2001, Paraoxonase and atherosclerosis. Arterioscler Thromb Vasc Biol 21:473–480 Ferre N, Camps J, Fernandez-Ballart J et al, 2003, Regulation of serum paraoxonase activity by genetic, nutritional, and lifestyle factors in the general population. Clin Chem 49:1491–1497 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 533 EP 540 DI 10.1007/s12253-010-9267-5 PG 8 ER PT J AU Valcz, G Sipos, F Krenacs, T Molnar, J Patai, V Leiszter, K Toth, K Solymosi, N Galamb, O Molnar, B Tulassay, Zs AF Valcz, Gabor Sipos, Ferenc Krenacs, Tibor Molnar, Jeannette Patai, V Arpad Leiszter, Katalin Toth, Kinga Solymosi, Norbert Galamb, Orsolya Molnar, Bela Tulassay, Zsolt TI Elevated Osteopontin Expression and Proliferative/Apoptotic Ratio in the Colorectal Adenoma–Dysplasia–Carcinoma Sequence SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Proliferative/apoptotic ratio; Adenoma–dysplasia–carcinoma sequence; Gene expression; Tissue microarray ID Proliferative/apoptotic ratio; Adenoma–dysplasia–carcinoma sequence; Gene expression; Tissue microarray AB Colorectal cancer progression is characterized by altered epithelial proliferation and apoptosis and by changed expression of tumor development regulators.Our aims were to determine the proliferative/apoptotic epithelial cell ratio (PAR) in the adenoma–dysplasia–carcinoma sequence (ADCS), and to examine its association with osteopontin (OPN), a previously identified protein product related to cancer development. One mm diameter cores from 13 healthy colons, 13 adenomas and 13 colon carcinoma samples were included into a tissue microarray (TMA) block. TUNEL reaction and Ki-67 immunohistochemistry were applied to determine the PAR. The osteopontin protein was also immunodetected. Stained slides were semiquantitatively evaluated using digital microscope and statistically analyzed with logistic regression and Fisher’s exact test. The PAR continuously increased along the ADCS. It was significantly (p<0.001) higher in cancer epithelium (8.84±7.01) than in adenomas (1.40±0.78) and in normal controls (0.89±0.21) (p<0.001). Also, significant positive correlation was observed between elevated PAR and the expression of osteopontin. Cytoplasmic OPN expression was weak in healthy samples. In contrast, cytoplasmic immunoreaction was moderately intensive in adenomas, while in colon cancer strong, diffuse cytoplasmic immune staining was detected. Increasing PAR and OPN expression along ADCS may help monitoring colorectal cancer progression. The significantly elevated OPN protein levels we found during normal epithelium to carcinoma progression may contribute to the increased fibroblast–myofibroblast transition determining stem cell niche in colorectal cancer. C1 [Valcz, Gabor] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Molnar, Jeannette] National Institute of Food and Nutrition ScienceBudapest, Hungary. [Patai, V Arpad] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Leiszter, Katalin] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Toth, Kinga] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Solymosi, Norbert] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Galamb, Orsolya] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. RP Valcz, G (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM valczg@yahoo.com CR Burgess AW, 1998, Growth control mechanisms in normal and transformed intestinal cells. Philos Trans R Soc Lond B Biol Sci 353:903–909 Walker MR, Patel KK, Stappenbeck TS, 2009, The stem cell niche. J Pathol 217:169–180 Fearon ER, Vogelstein B, 1990, A genetic model for colorectal tumorigenesis. Cell 61:759–767 Vogelstein B, Fearon ER, Hamilton SR et al, 1988, Genetic alterations during colorectal-tumor development. N Engl J Med 319:525–532 Leslie A, Carey FA, Pratt NR et al, 2002, The colorectal adenoma–carcinoma sequence. Br J Surg 89:845–860 Scholzen T, Gerdes J, 2000, The Ki-67 protein: from the known and the unknown. J Cell Physiol 182:311–322 Sipos F, Zagoni T, Molnar B et al, 2002, Changes in the proliferation and apoptosis of colonic epithelial cells in correlation with histologic activity of ulcerative colitis. Orv Hetil 143:2485– 2488 Tuck AB, Chambers AF, 2001, The role of osteopontin in breast cancer: clinical and experimental studies. J Mammary Gland Biol Neoplasia 6:419–429 Galamb O, Sipos F, Spisak S et al, 2009, Potential biomarkers of colorectal adenoma–dysplasia–carcinoma progression: mRNA expression profiling and in situ protein detection on TMAs reveal 15 sequentially upregulated and 2 downregulated genes. Cell Oncol 31:19–29 Wai PY, Kuo PC, 2008, Osteopontin: regulation in tumor metastasis. Cancer Metastasis Rev 27:103–118 Rangaswami H, Bulbule A, Kundu GC, 2006, Osteopontin: role in cell signaling and cancer progression. Trends Cell Biol 16:79–87 Bautista DS et al, 1994, Inhibition of Arg-Gly-Asp, RGD)- mediated cell adhesion to osteopontin by a monoclonal antibody against osteopontin. J Biol Chem 269:23280–23285 Hirama M, Takahashi F, Takahashi K et al, 2003, Osteopontin overproduced by tumor cells acts as a potent angiogenic factor contributing to tumor growth. Cancer Lett 198:107–117 Weber GF, 2008, Molecular mechanisms of metastasis. Cancer Lett 270:181–190 Lin YH, Yang-Yen HF, 2001, The osteopontin-CD44 survival signal involves activation of the phosphatidylinositol 3-kinase/Akt signaling pathway. J Biol Chem 276:46024–46030 Huang W, Carlsen B, Rudkin G et al, 2004, Osteopontin is a negative regular of proliferation and differentiation in MC3T3-E1 pre-osteoblastic cells. Bone 34:799–808 Panda D, Kundu GC, Lee BI et al, 1997, Potential roles of osteopontin and alphaVbeta3 integrin in the development of coronary artery restenosis after angioplasty. Proc Natl Acad Sci USA 94:9308–9313 Elgavish A, Prince C, Chang PL, 1998, Osteopontin stimulates a subpopulation of quiescent human prostate epithelial cells with high proliferative potential to divide in vitro. Prostate 35:83–94 El-Tanani MK, Campbell FC, Kurisetty V et al, 2006, The regulation and role of osteopontin in malignant transformation and cancer. Cytokine Growth Factor Rev 17:463–474 Lenga Y, Koh A, Perera AS et al, 2008, Osteopontin expression is required for myofibroblast differentiation. Circ Res 102:319–327 Saika S, Shirai K, Yamanaka O et al, 2007, Loss of osteopontin perturbs the epithelial–mesenchymal transition in an injured mouse lens epithelium. Lab Invest 87:130–138 Miyazaki K, Okada Y, Yamanaka O et al, 2008, Corneal wound healing in an osteopontin-deficient mouse. Invest Ophthalmol Vis Sci 49:1367–1375 Junaid A, Moon MC, Harding GE et al, 2007, Osteopontin localizes to the nucleus of 293 cells and associates with polo-like kinase-1. Am J Physiol Cell Physiol 292:919–926 Katenkamp D, Berndt R, Perevoshchikov A et al, 1984, Ultrastructural observations of the stroma of human colon carcinomas with special regard to myofibroblasts. Arch Geschwulstforsch 54:301–308 Hawinkels LJ, Verspaget HW, van der Reijden JJ et al, 2009, Active TGF-beta1 correlates with myofibroblasts and malignancy in the colorectal adenoma–carcinoma sequence. Cancer Sci 100:663–670 Weber GF, 2001, The metastasis gene osteopontin: a candidate target for cancer therapy. Biochim Biophys Acta 1552:61–85 Dai J, Li B, Shi J et al, 2010, A humanized anti-osteopontin antibody inhibits breast cancer growth and metastasis in vivo. Cancer Immunol Immunother 59:355–366 Thalmann GN, Sikes RA, Devoll RE et al, 1999, Osteopontin: possible role in prostate cancer progression. Clin Cancer Res 5:2271–2277 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 541 EP 545 DI 10.1007/s12253-010-9260-z PG 5 ER PT J AU Jaberipour, M Habibagahi, M Hosseini, A Habibabad, RS Talei, AAR Ghaderi, A AF Jaberipour, Mansooreh Habibagahi, Mojtaba Hosseini, Ahmad Habibabad, Rezai Saadat Talei, Abd-Al-Rasoul Ghaderi, Abbas TI Increased CTLA-4 and FOXP3 Transcripts in Peripheral Blood Mononuclear Cells of Patients with Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; CTLA-4 and FOXP3 ID Breast cancer; CTLA-4 and FOXP3 AB Generation of Regulatory T cells (Tregs) is known to play a major role in progression and modulation of the immune escape mechanisms in cancer. These cells express Forkhead/winged helix transcription factor (FOXP3) and also Cytotoxic T-lymphocyte antigen-4 (CTLA-4), as a negative regulatory molecule which, is a potential target for immunotherapy. We, therefore, evaluated FOXP3 and CTLA-4 transcripts in the peripheral blood mononuclear cells from 55 women with histologically-confirmed infiltrating ductal carcinoma of the breast. Blood samples from 40 healthy volunteer women without a history of malignancies or autoimmune disorders were also obtained as a control. The abundance of FOXP3 and CTLA-4 gene transcripts was determined by quantitative real-time PCR (qRT-PCR). Compared to healthy individuals, significantly higher amounts of these transcripts were found in the mononuclear cells from breast cancer patients. Also, a significant correlation was found between CTLA-4 and FOXP3 expressions in a group of patients. Among patients with early stage, nonmetastatic or low-grade disease, the relative expression of CTLA-4 was about 10-fold as much as in the control group. These patients also showed a significant increase, more than 10 fold, in mean relative FOXP3 expression. The results of this investigation point to functional activity of Treg cells in early stages of breast cancer, a finding which emphasizes the significance of Tregs as an imminent target for breast cancer immunotherapy. C1 [Jaberipour, Mansooreh] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Habibagahi, Mojtaba] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Hosseini, Ahmad] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Habibabad, Rezai Saadat] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Talei, Abd-Al-Rasoul] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Ghaderi, Abbas] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. RP Habibagahi, M (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Immunology, Shiraz, Iran. EM agahim@sums.ac.ir CR Kaklamanis L, Townsend A, Doussis-Anagnostopoulou IA et al, 1994, Loss of major histocompatibility complex-encoded transporter associated with antigen presentation, TAP, in colorectal cancer. Am J Pathol 145:505–509 Elgert KD, Alleva DG, Mullins DW, 1998, Tumor-induced immune dysfunction: the macrophage connection. J Leukoc Biol 64:275–290 Staveley-O’Carroll K, Sotomayor E, Montgomery J et al, 1998, Induction of antigen-specific T cell anergy: an early event in the course of tumor progression. Proc Natl Acad Sci USA 95:1178–1183 Roncarolo MG, Gregori S, Levings M, 2003, Type 1 T regulatory cells and their relationship with CD4+CD25+ T regulatory cells. Novartis Found Symp 252:115–127 Caras I, Grigorescu A, Stavaru C et al, 2004, Evidence for immune defects in breast and lung cancer patients. Cancer Immunol Immunother 53:1146–1152 Wolf AM, Wolf D, Steurer M et al, 2003, Increase of regulatory T cells in the peripheral blood of cancer patients. Clin Cancer Res 9:606–612 O’Garra A, Vieira P, 2004, Regulatory T cells and mechanisms of immune system control. Nat Med 10:801–805 Sakaguchi S, 2000, Regulatory T cells: key controllers of immunologic self-tolerance. Cell 101:455–458 Sakaguchi S, 2005, Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol 6:345–352 Hori S, Nomura T, Sakaguchi S, 2003, Control of regulatory T cell development by the transcription factor Foxp3. Science 299:1057–1061 Fontenot JD, Rasmussen JP, Williams LM et al, 2005, Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity 22:329–341 Woo EY, Chu CS, Goletz TJ et al, 2001, Regulatory CD4(+, CD25(+, T cells in tumors from patients with early-stage nonsmall cell lung cancer and late-stage ovarian cancer. Cancer Res 61:4766–4772 Liyanage UK, Moore TT, Joo HG et al, 2002, Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J Immunol 169:2756–2761 Viguier M, Lemaitre F, Verola O et al, 2004, Foxp3 expressing CD4+CD25(high, regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells. J Immunol 173:1444–1453 Ormandy LA, Hillemann T, Wedemeyer H et al, 2005, Increased populations of regulatory T cells in peripheral blood of patients with hepatocellular carcinoma. Cancer Res 65:2457–2464 Clarke SL, Betts GJ, Plant A et al, 2006, CD4+CD25+FOXP3+ regulatory T cells suppress anti-tumor immune responses in patients with colorectal cancer. PLoSONE 1:e129 Gallimore A, Godkin A, 2008, Regulatory T cells and tumour immunity—observations in mice and men. Immunology 123:157–163 Strauss L, Bergmann C, Gooding W et al, 2007, The frequency and suppressor function of CD4+CD25highFoxp3+ T cells in the circulation of patients with squamous cell carcinoma of the head and neck. Clin Cancer Res 13:6301–6311 Bi Y, Wei L, Mao HT, Zhang L, Zuo WS, 2008, Expressions of Fas, CTLA-4 and RhoBTB2 genes in breast carcinoma and their relationship with clinicopathological factors. Zhonghua ZhongLiu Za Zhi 30:749–753, Article in Chinease Ohara M, Yamaguchi Y, Matsuura K et al, 2009, Possible involvement of regulatory T cells in tumor onset and progression in primary breast cancer. Cancer Immunol Immunother 58:441–447 Mansfield AS, Heikkila PS, Vaara AT et al, 2009, Simultaneous Foxp3 and IDO expression is associated with sentinel lymph node metastases in breast cancer. BMC Cancer 9:231 Merlo A, Casalini P, Carcangiu ML et al, 2009, FOXP3 expression and overall survival in breast cancer. J Clin Oncol 27:1746–1752 Gupta S, Joshi K, Wig JD, Arora SK, 2007, Intratumoral FOXP3 expression in infiltrating breast carcinoma: its association with clinicopathologic parameters and angiogenesis. Acta Oncol 46:792–797 Pfaffl MW, 2001, A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 29:e45 Shevach EM, McHugh RS, Piccirillo CA et al, 2001, Control of T-cell activation by CD4+ CD25+ suppressor T cells. Immunol Rev 182:58–67 Hilchey SP, Bernstein SH, 2007, Use of CFSE to monitor ex vivo regulatory T-cell suppression of CD4+ and CD8+ T-cell proliferation within unseparated mononuclear cells from malignant and nonmalignant human lymph node biopsies. Immunol Invest 36:629–648 Brusko TM, Hulme MA, Myhr CB et al, 2007, Assessing the in vitro suppressive capacity of regulatory T cells. Immunol Invest 36:607–628 Capriotti E, Vonderheid EC, Thoburn CJ et al, 2008, Expression of T-plastin, FoxP3 and other tumor-associated markers by leukemic Tcells of cutaneous T-cell lymphoma. Leuk Lymphoma 49:1190–1201 Tuve S, Chen BM, Liu Y et al, 2007, Combination of tumor sitelocated CTL-associated antigen-4 blockade and systemic regulatory T-cell depletion induces tumor-destructive immune responses. Cancer Res 67:5929–5939 Matsuura K, Yamaguchi Y, Ueno H et al, 2006, Maturation of dendritic cells and T-cell responses in sentinel lymph nodes from patients with breast carcinoma. Cancer 106:1227–1236 Bates GJ, Fox SB, Han C et al, 2006, Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J Clin Oncol 24:5373–5380 Hodi FS, Mihm MC, Soiffer RJ et al, 2003, Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci USA 100:4712–4717 Phan GQ, Yang JC, Sherry RM et al, 2003, Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci USA 100:8372–8377 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 547 EP 551 DI 10.1007/s12253-010-9256-8 PG 5 ER PT J AU Bircan, A Bircan, S Kapucuoglu, N Songur, N Ozturk, O Akkaya, A AF Bircan, Ahmet Bircan, Sema Kapucuoglu, Nilgun Songur, Necla Ozturk, Onder Akkaya, Ahmet TI Maspin, VEGF and p53 Expression in Small Biopsies of Primary Advanced Lung Cancer and Relationship with Clinicopathologic Parameters SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Maspin; VEGF; p53; Biopsy; Immunohistochemistry ID Lung cancer; Maspin; VEGF; p53; Biopsy; Immunohistochemistry AB Maspin, one of the serine protease inhibitors, has been shown to inhibit tumor progression and metastasis. We aimed to investigate maspin, p53 and VEGF expression in patients with squamous cell carcinoma (SCC), adenocarcinoma (AC) and small cell lung carcinoma (SCLC). The study included 28 SCC, 18 AC, 17 SCLC biopsy samples. We used the streptavidin biotin immunoperoxidase method to test for maspin, p53 and VEGF antibodies. Medical records of these patients were reviewed from archival files. Cytoplasmic maspin expression was detected in 89.3%, 77.8%, 52.9% of SCC, AC and SCLC, respectively. The rate was significantly higher in non-small cell lung cancer (NSCLC) and SCC than SCLC (p=0.013, p=0.021, respectively). The mean percentages of maspin expression were significantly higher in NSCLC, SCC and AC than in SCLC (p=0.0001, p=0.0001, p=0.038, respectively). In ACs, maspin and p53 expressions were correlated, although this was not statistically significant (p=0.053, r=0.464), and maspin positive cases had a significantly higher T status compared to negative cases (p=0.036). In SCC, the stage of disease was positively correlated with p53 (p=0.007, r=0.536) and negatively correlated with VEGF expression (p=0.013, r=−0.498). Multivariate analysis demonstrated that stage of disease was a significant independent prognostic parameter in NSCLC (95% confidence interval: 1.067–3.969; p=0.031). Although maspin expression is higher in SCC and AC, and is related with higher T status in AC, our data did not indicate its prognostic significance. Larger scale studies are needed to reveal the exact role of maspin in lung cancer pathogenesis. C1 [Bircan, Ahmet] Suleyman Demirel University, Faculty of Medicine, Department of Pulmonary Medicine, 32260 Cunur, Isparta, Turkey. [Bircan, Sema] Suleyman Demirel University, Faculty of Medicine, Department of PathologyIsparta, Turkey. [Kapucuoglu, Nilgun] Suleyman Demirel University, Faculty of Medicine, Department of PathologyIsparta, Turkey. [Songur, Necla] Suleyman Demirel University, Faculty of Medicine, Department of Pulmonary Medicine, 32260 Cunur, Isparta, Turkey. [Ozturk, Onder] Suleyman Demirel University, Faculty of Medicine, Department of Pulmonary Medicine, 32260 Cunur, Isparta, Turkey. [Akkaya, Ahmet] Suleyman Demirel University, Faculty of Medicine, Department of Pulmonary Medicine, 32260 Cunur, Isparta, Turkey. RP Bircan, A (reprint author), Suleyman Demirel University, Faculty of Medicine, Department of Pulmonary Medicine, 32260 Cunur, Turkey. EM ahbircan@yahoo.com CR Zou Z, Anisowicz A, Hendrix MJ et al, 1994, Maspin, a serpin with tumor-suppressing activity in human mammary epithelial cells. Science 263:526–529 Chen EI, Yates JR, 2006, Maspin and tumor metastasis. IUBMB Life 58:25–29 Bailey CM, Khalkhali-Ellis Z, Seftor EA et al, 2006, Biological functions of Maspin. J Cell Physiol 209:617–624 Lockett J, Yin S, Li X et al, 2006, Tumor suppressive maspin and epithelial homeostasis. J Cell Biochem 97:651–660 Zhang M, Volpert O, Shi YH et al, 2000, Maspin is an angiogenesis inhibitor. Nat Med 6:196–199 Nakagawa M, Katakura H, Adachi M et al, 2006, Maspin expression and its clinical significance in non-small cell lung cancer. Ann Surg Oncol 13:1517–1523 Woenckhaus M, Bubendorf L, Dalquen P et al, 2007, Nuclear and cytoplasmic maspin expression in primary non-small cell lung cancer. J Clin Pathol 60:483–486 Takanami I, Abiko T, Koizumi S, 2008, Expression of maspin in non-small-cell lung cancer: correlation with clinical features. Clin Lung Cancer 9:361–366 Katakura H, Takenaka K, Nakagawa M et al, 2006, Maspin gene expression is a significant prognostic factor in resected non-small cell lung cancer, NSCLC).Maspin in NSCLC. Lung Cancer 51:323–328 Hirai K, Koizumi K, Haraguchi S et al, 2005, Prognostic significance of the tumor suppressor gene maspin in non-small cell lung cancer. Ann Thorac Surg 79:248–253 Zheng HC, Saito H, Masuda S et al, 2008, Cytoplasmic and nuclear maspin expression in lung carcinomas: an immunohistochemical study using tissue microarrays. App Immunohistochem Mol Morphol 16:459–465 Nakashima M, Ohike N, Nagasaki K et al, 2004, Prognostic significance of the maspin tumor suppressor gene in pulmonary adenocarcinoma. J Cancer Res Clin Oncol 130:475–479 Lonardo F, Li X, Siddiq F et al, 2006, Maspin nuclear localization is linked to favorable morphological features in pulmonary adenocarcinoma. Lung Cancer 51:31–39 Frey A, Soubani AO, Adam AK et al, 2009, Nuclear, compared with combined nuclear and cytoplasmic expression of maspin, is linked in lung adenocarcinoma to reduced VEGF-A levels and in Stage I, improved survival. Histopathology 54:590–597 Yilmaz A, Ernam D, Unsal E et al, 2007, Vascular endothelial growth factor immunostaining correlates with postoperative relapse and survival in non-small cell lung cancer. Arch Med Res 38:764–768 Bremnes RM, Camps C, Sirera R, 2006, Angiogenesis in nonsmall cell lung cancer: the prognostic impact of neoangiogenesis and the cytokines VEGF and bFGF in tumours and blood. Lung Cancer 51:143–158 Boldrini L, Gisfredi S, Ursino S et al, 2005, Interleukin-8 in nonsmall cell lung carcinoma: relation with angiogenic pattern and p53 alterations. Lung Cancer 50:309–317 Zhu CQ, Shih W, Ling CH et al, 2006, Immunohistochemical markers of prognosis in non-small cell lung cancer: a review and proposal for a multiphase approach to marker evaluation. J Clin Pathol 59:790–800 Hollstein M, Sidransky D, Volgestein B et al, 1991, p53 mutation in human cancers. Science 253:49–53 Travis WD, Brambilla E, Muller-Hermelink HK, Harris CC, 2004, World Health Organization classification of tumours. Pathology and genetics of tumours of the lung, pleura, thymus and heart. IARC Press, Lyon Ludovini V, Gregorc V, Pistola L et al, 2004, Vascular endothelial growth factor, p53, Rb, Bcl-2 expression and response to chemotherapy in advanced non-small cell lung cancer. Lung Cancer 46:77–85 Freemantle SJ, Dmitrovsky E, 2002, Clinical link between p53 and angiogenesis in lung cancer. J Clin Oncol 20:883–884 Yuan A, Yu CJ, Luh KT et al, 2002, Aberrant p53 expression correlates with expression of vascular endothelial growth factor mRNA and interleukin-8 mRNA and neoangiogenesis in nonsmall- cell lung cancer. J Clin Oncol 20:900–910 Mukhopadhyay D, Tsiokas L, Sukhatme VP, 1995, Wild-type p53 and v-Src exert opposing influences on human vascular endothelial growth factor gene expression. Cancer Res 57:6161–6165 Zou Z, Gao C, Nagaich AK et al, 2000, p53 regulates the expression of the tumor suppressor gene maspin. J Biol Chem 275:6051–6054 Oken MM, Creech RH, Tormey DC et al, 1982, Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol 5:649–655 Mountain CF, 1997, Revisions in the international system for staging lung cancer. Chest 111:1710–1717 Stahel RA, Ginsberg R, Havemann K et al, 1989, Staging and prognostic factors in small cell lung cancer: a consensus report. Lung Cancer 5:119–126 Smith SL, Watson SG, Ratschiller D et al, 2003, Maspin- the most commonly expressed gene of the 18q21.3 serpin cluster in lung cancer- is strongly expressed in preneoplastic bronchial lesions. Oncogene 22:8677–8687 Steels E, Paesmans M, Berghamans T et al, 2001, Role of p53 as a prognostic factor for survival in lung cancer: a systematic review of the literature with a meta-analysis. Eur Respir J 18:705–719 Laudanski J, Niklinska W, Burzykowski T et al, 2001, Prognostic significance of p53 and bcl-2 abnormalities in operable nonsmall cell lung cancer. Eur Respir J 17:660–666 Tsao MS, Aviel-Ronen S, Ding K et al, 2007, Prognostic and predictive importance of p53 and RAS for adjuvant chemotherapy in non small-cell lung cancer. J Clin Oncol 25:5240–5247 O’Byrne KJ, Koukourakis MI, Giatromanolaki A et al, 2000, Vascular endothelial growth factor, platelet-derived endothelial cell growth factor and angiogenesis in non-small-cell lung cancer. Br J Cancer 82:1427–1432 Fontanini G, Boldrini L, Vignati S et al, 1998, Bcl-2 and p53 regulate vascular endothelial growth factor, VEGF)-mediated angiogenesis in non-small cell lung carcinoma. Eur J Cancer 34:718–723 Yoo J, Yung JH, Lee MA et al, 2007, Immunohistochemical analysis of non-small cell lung cancer: correlation with clinical parameters and prognosis. J Korean Med Sci 22:318–325 Ito H, Oshita F, Kameda Y et al, 2002, Expression of vascular endothelial growth factor and basic fibroblast growth factor in small adenocarcinomas. Oncol Rep 9:119–123 Seto T, Higashiyama M, Funai H et al, 2006, Prognostic value of expression of vascular endothelial growth factor and its flt-1 and KDR receptors in stage I non-small- cell lung cancer. Lung Cancer 53:91–96 Liao M, Wang H, Lin Z et al, 2001, Vascular endothelial growth factor and other biological predictors related to the postoperative survival rate on non-small cell lung cancer. Lung Cancer 33:125– 132 Nakashima T, Huang CL, Liu D et al, 2004, Expression of vascular endothelial growth factor-A and vascular endothelial growth factor-C as prognostic factors for non small cell lung cancer. Med Sci Monit 10:BR157–BR165 Niklinska W, Burzykowski T, Chyczewski L, Niklinski J, 2001, Expression of vascular endothelial growth factor, VEGF, in nonsmall cell lung cancer, NSCLC): association with p53 gene mutation and prognosis. Lung Cancer 34:S59–S64 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 553 EP 561 DI 10.1007/s12253-010-9259-5 PG 9 ER PT J AU Eren, F Akkiprik, M Atug, Sonmez, O Tahan, G Ozdemir, F Hamzaoglu, H Celikel, A Imeryuz, N Avsar, E Ozer, A AF Eren, Fatih Akkiprik, Mustafa Atug, Ozlen Sonmez, Ozgur Tahan, Gulgun Ozdemir, Filiz Hamzaoglu, Over Hulya Celikel, Ataizi Cigdem Imeryuz, Nese Avsar, Erol Ozer, Ayse TI R72P Polymorphism of TP53 in Ulcerative Colitis Patients is Associated with the Incidence of Colectomy, Use of Steroids and the Presence of a Positive Family History SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p53; Codon 72 polymorphism; Colorectal cancer; Ulcerative colitis; Colectomy; Steroid; Family history ID p53; Codon 72 polymorphism; Colorectal cancer; Ulcerative colitis; Colectomy; Steroid; Family history AB P53 tumor suppressor protein is one of the pivotal regulators for genome integrity, cell cycle and apoptosis. The most commonly and extensively studied single nucleotide polymorphism (SNP) of p53 is Arg>Pro substitution on codon 72 (R72P). Although we know that the SNP has unique functional effects on the protein, its clinical significance is not clearly identified yet. Aim of the study was to access the relationship between R72P genotype distribution and clinical variables in patients with ulcerative colitis (UC) and colorectal cancer (CRC). Genomic DNA samples were extracted from 95 UC, 50 CRC, and 219 healthy controls. R72P genotype analysis was carried out with polymerase chain reaction following by restriction enzyme digestion. We observed that Pro allele carriage is a strong risk factor for CRC (OR=3.03; 95%CI=1.91–2.40; p=0.003), but only modest association with UC (OR=1.61; 95%CI=0.98–2.65; p=0.059) (Pro/Pro and Pro/Arg genotypes vs. Arg/Arg genotype). We did not find any correlation between genotype distribution of the polymorphism and clinical parameters of CRC, but in UC, Pro/Pro genotype was significantly related to an inflammatory bowel disease family history (OR=8.0; 95%CI=1.68–38.08, p=0.015), and Arg/Pro genotype was significantly associated with the history of disease-related colectomy (OR=17.77; 95%CI=0.98–323.34, p=0.012) and steroid use (OR=10.14; 95%CI=2.63–39.12, p=0.0002). Our data suggest that R72P variant seems to be associated with high risk for development of CRC but carries low risk for development of UC. R72P genotypes might be a useful predictive marker for surgical and medical treatment of UC. C1 [Eren, Fatih] Marmara University, School of Medicine, Department of Medical Biology, Tibbiye Cad., No: 49, Haydarpasa, 34668 Istanbul, Turkey. [Akkiprik, Mustafa] Marmara University, School of Medicine, Department of Medical Biology, Tibbiye Cad., No: 49, Haydarpasa, 34668 Istanbul, Turkey. [Atug, Ozlen] Marmara University, Gastroenterology InstituteBasibuyuk, Turkey. [Sonmez, Ozgur] Marmara University, School of Medicine, Department of Medical Biology, Tibbiye Cad., No: 49, Haydarpasa, 34668 Istanbul, Turkey. [Tahan, Gulgun] Marmara University, Gastroenterology InstituteBasibuyuk, Turkey. [Ozdemir, Filiz] Marmara University, Gastroenterology InstituteBasibuyuk, Turkey. [Hamzaoglu, Over Hulya] Marmara University, Gastroenterology InstituteBasibuyuk, Turkey. [Celikel, Ataizi Cigdem] Marmara University, School of Medicine, Department of PathologyAltunizade, Turkey. [Imeryuz, Nese] Marmara University, Gastroenterology InstituteBasibuyuk, Turkey. [Avsar, Erol] Marmara University, Gastroenterology InstituteBasibuyuk, Turkey. [Ozer, Ayse] Marmara University, School of Medicine, Department of Medical Biology, Tibbiye Cad., No: 49, Haydarpasa, 34668 Istanbul, Turkey. RP Akkiprik, M (reprint author), Marmara University, School of Medicine, Department of Medical Biology, 34668 Istanbul, Turkey. EM akkiprik@yahoo.com CR Sakamuro D, Sabbatini P, White E et al, 1997, The polyproline region of p53 is required to activate apoptosis but not growth arrest. Oncogene 15:887–898 Walker KK, Levine AJ, 1996, Identification of a novel p53 functional domain that is necessary for efficient growth suppression. Proc Natl Acad Sci U S A 93:15335–15340 Whibley C, Pharoah PD, Hollstein M, 2009, p53 polymorphisms: cancer implications. Nat Rev Cancer 9:95–107 Marin MC, Jost CA, Brooks LA et al, 2000, A common polymorphism acts as an intragenic modifier of mutant p53 behaviour. Nat Genet 25:47–54 Gemignani F,Moreno V, Landi S et al, 2004, ATP53 polymorphism is associated with increased risk of colorectal cancer and with reduced levels of TP53 mRNA. Oncogene 23:1954–1956 Dumont P, Leu JI, Della Pietra AC et al, 2003, The codon 72 polymorphic variants of p53 have markedly different apoptotic potential. Nat Genet 33:357–365 Sullivan A, Syed N, Gasco M et al, 2004, Polymorphism in wildtype p53 modulates response to chemotherapy in vitro and in vivo. Oncogene 23:3328–3337 Xu Y, Yao L, Ouyang T et al, 2005, p53 Codon 72 polymorphism predicts the pathologic response to neoadjuvant chemotherapy in patients with breast cancer. Clin Cancer Res 11:7328–7333 Nelson HH, Wilkojmen M, Marsit CJ et al, 2005, TP53 mutation, allelism and survival in non-small cell lung cancer. Carcinogenesis 26:1770–1773 Ekbom A, Helmick C, Zack M et al, 1990, Ulcerative colitis and colorectal cancer. A population-based study. N Engl J Med 323:1228–1233 Burmer GC, Rabinovitch PS, Haggitt RC et al, 1992, Neoplastic progression in ulcerative colitis: histology, DNA content, and loss of a p53 allele. Gastroenterology 103:1602–1610 Brentnall TA, Crispin DA, Rabinovitch PS et al, 1994, Mutations in the p53 gene: an early marker of neoplastic progression in ulcerative colitis. Gastroenterology 107:369–378 Harpaz N, Peck AL, Yin J et al, 1994, p53 protein expression in ulcerative colitis-associated colorectal dysplasia and carcinoma. Hum Pathol 25:1069–1074 Holzmann K, Klump B, Borchard F et al, 1998, Comparative analysis of histology, DNA content, p53 and Ki-ras mutations in colectomy specimens with long-standing ulcerative colitis. Int J Cancer 76:1–6 Lashner BA, Shapiro BD, Husain A et al, 1999, Evaluation of the usefulness of testing for p53 mutations in colorectal cancer surveillance for ulcerative colitis. Am J Gastroenterol 94:456–462 Lennard-Jones JE, 1989, Classification of inflammatory bowel disease. Scand J Gastroenterol Suppl 170:2–6 Hanauer SB, Sandborn W, 2001, Management of Crohn’s Disease in adults. Am J Gastroenterol 96:635–643 Soong R, Iacopetta BJ, 1997, A rapid and nonisotopic method for the screening and sequencing of p53 gene mutations in formalin fixed, paraffin-embedded tumours. Med Pathol 10:252–258 Zhu ZZ, Cong WM, Liu SF et al, 2005, A p53 polymorphism modifies the risk of hepatocellular carcinoma among non-carriers but not carriers of chronic hepatitis B virus infection. Cancer Lett 229:77–83 Sayhan N, Yazici H, Budak M et al, 2001, P53 codon 72 genotypes in colon cancer. Association with human papillomavirus infection. Res Commun Mol Pathol Pharmacol 109:25–34 Zhu ZZ, Wang AZ, Jia HR et al, 2007, Association of the TP53 codon 72 polymorphism with colorectal cancer in a Chinese population. Jpn J Clin Oncol 37:385–390 Cao Z, Song JH, Park YK et al, 2009, The p53 codon 72 polymorphism and susceptibility to colorectal cancer in Korean patients. Neoplasma 56:114–118 Perez LO, Abba MC, Dulout FN et al, 2006, Evaluation of p53 codon 72 polymorphismin adenocarcinomas of the colon and rectum in La Plata, Argentina. World J Gastroenterol 12:1426–1429 Dakouras A, Nikiteas N, Papadakis E et al, 2008, P53Arg72 homozygosity and its increased incidence in left-sided sporadic colorectal adenocarcinomas, in a Greek-Caucasian population. Anticancer Res 28:1039–1043 Sjalander A, Birgander R, Athlin L et al, 1995, P53 germ line haplotypes associated with increased risk for colorectal cancer. Carcinogenesis 16:1461–1464 Koushik A, Tranah GJ, Ma J et al, 2006, p53 Arg72Pro polymorphism and risk of colorectal adenoma and cancer. Int J Cancer 119:1863–1868 Vietri MT, Riegler G, Ursillo A et al, 2007, p53 codon 72 polymorphism in patients affected with ulcerative colitis. J Gastroenterol 42:456–460 Kaiser MA, Beart RW, 2001, Surgical management of ulcerative colitis. Swiss Med Wkly 131:323–337 Sleisenger MH, Fordtran JS, 1989, Gastrointestinal disease volume 2 pathophysiology diagnosis management, 4th edn. Saunders, Philadelphia Lampinen M, Sangfelt P, Taha Y, Carlson M, 2008, Accumulation, activation, and survival of neutrophils in ulcerative colitis: regulation by locally produced factors in the colon and impact of steroid treatment. Int J Colorectal Dis 23:939–946 Hagiwara C, Tanaka M, Kudo H, 2002, Increase in colorectal epithelial apoptotic cells in patients with ulcerative colitis ultimately requiring surgery. J Gastroenterol Hepatol 17:758–764 Yoshiyama S, Miki C, Okita Y, Araki T, Uchida K, Kusunoki M, 2008, Neutrophil-related immunoinflammatory disturbance in steroid-overdosed ulcerative colitis patients. J Gastroenterol 43:789–797 Ueyama H, Kiyohara T, Sawada N et al, 1998, High Fas ligand expression on lymphocytes in lesions of ulcerative colitis. Gut 43:48–55 Thomas M, Kalita A, Labrecque S, Pim D, Banks L, Matlashwski G, 1999, Two polymorphic variants of wild-type p53 differ biochemically and biologically. Mol Cell Biol 19:1092–1100 Pim D, Banks L, 2004, p53 polymorphic variants at codon 72 exert different effects on cell cycle progression. Int J Cancer 108:196–199 Komarova EA, Krivokrysenko V, Wang K et al, 2005, p53 is a suppressor of inflammatory response in mice. FASEB J 19:1030– 1032 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 563 EP 568 DI 10.1007/s12253-010-9255-9 PG 6 ER PT J AU Bektas, S Bahadir, B Ucan, HB Ozdamar, OS AF Bektas, Sibel Bahadir, Burak Ucan, Hamdi Bulent Ozdamar, Oguz Sukru TI CD24 and Galectin-1 Expressions in Gastric Adenocarcinoma and Clinicopathologic Significance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD24; Clinicopathologic variables; Galectin-1; Gastric adenocarcinoma; Prognosis ID CD24; Clinicopathologic variables; Galectin-1; Gastric adenocarcinoma; Prognosis AB CD24 and galectin-1 expression in gastric adenocarcinoma and their clinicopathologic significance remained largely unknown. We aimed to evaluate expressions and staining intensities of CD24 and galectin-1 in gastric adenocarcinoma and to investigate the interrelation with clinicopathologic parameters including survival. 93 cases with gastric adenocarcinoma were reevaluated histopathologically and immunohistochemistry was performed with antibodies against CD24 and galectin-1. Staining intensities of both markers in tumor cells and staining intensity of galectin-1 in tumor-associated stromal cells were scored semiquantitatively. The relationship between expression and staining intensity of CD24 and galectin-1 and clinicopathologic variables were assessed. CD24 staining intensity was associated with lymphovascular invasion (p=0.007), serosal invasion (p=0.001), stage (p=0.001) and lymph node metastasis (p=0.005). Galectin-1 staining intensity in tumor-associated stromal cells was associated with tumor location (p=0.031), lymphovascular invasion (p=0.001), perineural invasion (p=0.001), serosal invasion (p=0.001), differentiation (p=0.003), stage (p=0.001) and lymph node metastasis (p=0.001). Staining intensity of CD24 (p=0.019) and gal-1 (p=0.018) were associated with patient survival. Staining intensity of CD24 in tumor cells and galectin-1 in tumor-associated stromal cells were related with certain clinicopathologic variables. Our findings suggest that these markers are independent prognostic indicators of poor survival and may serve as useful targets for novel therapies. C1 [Bektas, Sibel] Zonguldak Karaelmas University, School of Medicine, Department of Pathology, 67600 Kozlu, Zonguldak, Turkey. [Bahadir, Burak] Zonguldak Karaelmas University, School of Medicine, Department of Pathology, 67600 Kozlu, Zonguldak, Turkey. [Ucan, Hamdi Bulent] Zonguldak Karaelmas University, School of Medicine, Department of Surgery, 67600 Kozlu, Zonguldak, Turkey. [Ozdamar, Oguz Sukru] Zonguldak Karaelmas University, School of Medicine, Department of Pathology, 67600 Kozlu, Zonguldak, Turkey. RP Bektas, S (reprint author), Zonguldak Karaelmas University, School of Medicine, Department of Pathology, 67600 Kozlu, Turkey. EM sibel_bektas@yahoo.com CR Parkin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Rosai J, 2004, Stomach. In: Rosai J, ed, Rosai and Ackerman’s surgical pathology, 9th edn. Elsevier-Mosby, New York, pp 648– 684 Chou YY, Jeng YM, Lee TT et al, 2007, Cytoplasmic CD24 expression is a novel prognostic factor in diffuse-type gastric adenocarcinoma. Ann Surg Oncol 14:2748–2758 Lim SC, Oh SH, 2005, The role of CD24 in various human epithelial neoplasias. Pathol Res Pract 201:479–486 Baumann P, Cremers N, Kroese F et al, 2005, CD24 expression causes the acquisition of multiple cellular properties associated with tumor growth and metastasis. Cancer Res 65:10783–10793 Li O, Zheng P, Liu Y, 2004, CD24 expression on T cells is required for optimal T cell proliferation in lymphopenic host. J Exp Med 200:1083–1089 Nielsen PJ, Lorenz B, Muller AM et al, 1997, Altered erythrocytes and a leaky block in B-cell development in CD24/ HSA-deficient mice. Blood 89:1058–1067 Aigner S, Sthoeger ZM, Fogel M et al, 1997, CD24, a mucin-type glycoprotein, is a ligand for P-selectin on human tumor cells. Blood 89:3385–3395 Athanassiadou P, Grapsa D, Gonidi M et al, 2009, CD24 expression has a prognostic impact in breast carcinoma. Pathol Res Pract 205:524–533 Kristiansen G, Winzer KJ, Mayordomo E et al, 2003, CD24 expression is a new prognostic marker in breast cancer. Clin Cancer Res 9:4906–4913 Kristiansen G, Denkert C, Schluns K et al, 2002, CD24 is expressed in ovarian cancer and is a new independent prognostic marker of patient survival. Am J Pathol 161:1215–1221 Lee HJ, Kim DI, Kwak C et al, 2008, Expression of CD24 in clear cell renal cell carcinoma and its prognostic significance. Urology 72:603–607 Kristiansen G, Pilarsky C, Pervan J et al, 2004, CD24 expression is a significant predictor of PSA relapse and poor prognosis in low grade or organ confined prostate cancer. Prostate 58:183–192 Sano A, Kato H, Sakurai S et al, 2009, CD24 expression is a novel prognostic factor in esophageal squamous cell carcinoma. Ann Surg Oncol 16:506–514 Kristiansen G, Schluns K, Yongwei Y et al, 2003, CD24 is an independent prognostic marker of survival in nonsmall cell lung cancer patients. Br J Cancer 88:231–236 Georgiadis V, Stewart HJ, Pollard HJ et al, 2007, Lack of galectin-1 results in defects in myoblast fusion and muscle regeneration. Dev Dyn 236:1014–1024 Hsu DK, Liu FT, 2004, Regulation of cellular homeostasis by galectins. Glycoconj J 19:507–515 Hittelet A, Legendre H, Nagy N et al, 2003, Upregulation of galectins-1 and -3 in human colon cancer and their role in regulating cell migration. Int J Cancer 103:370–379 Sakaguchi M, Imaizumi Y, Okano H, 2007, Expression and function of galectin-1 in adult neural stem cells. Cell Mol Life Sci 64:1254–1258 Satelli A, Rao PS, Gupta PK et al, 2008, Varied expression and localization of multiple galectins in different cancer cell lines. Oncol Rep 19:587–594 Thijssen VL, Hulsmans S, Griffioen AW, 2008, The galectin profile of the endothelium: altered expression and localization in activated and tumor endothelial cells. Am J Pathol 172:545–553 Jung EJ, Moon HG, Cho BI et al, 2007, Galectin-1 expression in cancer-associated stromal cells correlates tumor invasiveness and tumor progression in breast cancer. Int J Cancer 120:2331–2338 Clausse N, van den Brule F, Waltregny D et al, 1999, Galectin-1 expression in prostate tumor-associated capillary endothelial cells is increased by prostate carcinoma cells and modulates heterotypic cell–cell adhesion. Angiogenesis 3:317–325 Nagy N, Legendre H, Engels O et al, 2003, Refined prognostic evaluation in colon carcinoma using immunohistochemical galectin fingerprinting. Cancer 97:1849–1858 Kristiansen G, Sammar M, Altevogt P, 2004, Tumour biological aspects of CD24, a mucin-like adhesion molecule. J Mol Histol 35:255–262 Lim SC, 2005, CD24 and human carcinoma: tumor biological aspects. Biomed Pharmacother 59:S351–354 van den Brule F, Califice S, Castronovo V, 2004, Expression of galectins in cancer: a critical review. Glycoconj J 19:537–542 Camby I, Le Mercier M, Lefranc F et al, 2006, Galectin-1: a small protein with major functions. Glycobiology 16:137R–157R Fogel M, Friederichs J, Zeller Yet al, 1999, CD24 is a marker for human breast carcinoma. Cancer Lett 143:87–94 Bircan S, Kapucuoglu N, Baspinar S et al, 2006, CD24 expression in ductal carcinoma in situ and invasive ductal carcinoma of breast: an immunohistochemistry-based pilot study. Pathol Res Pract 202:569–576 Weichert W, Denkert C, Burkhardt M et al, 2005, Cytoplasmic CD24 expression in colorectal cancer independently correlates with shortened patient survival. Clin Cancer Res 11:6574–6581 Sagiv E, Arber N, 2008, The novel oncogene CD24 and its arising role in the carcinogenesis of the GI tract: from research to therapy. Expert Rev Gastroenterol Hepatol 2:125–133 Nagy B, Szendroi A, Romics I, 2009, Overexpression of CD24, c-myc and phospholipase 2A in prostate cancer tissue samples obtained by needle biopsy. Pathol Oncol Res 15:279–283 Huang LR, Hsu HC, 1995, Cloning and expression of CD24 gene in human hepatocellular carcinoma: a potential early tumor marker gene correlates with p53 mutation and tumor differentiation. Cancer Res 55:4717–4721 Kim KH, Choi JS, Kim JM et al, 2009, Enhanced CD24 expression in endometrial carcinoma and its expression pattern in normal and hyperplastic endometrium. Histol Histopathol 24:309–316 Kim JB, Ko E, Han W et al, 2008, CD24 cross-linking induces apoptosis in, and inhibits migration of, MCF-7 breast cancer cells. BMC Cancer 24:118 Agrawal S, Kuvshinoff BW, Khoury T et al, 2007, CD24 expression is an independent prognostic marker in cholangiocarcinoma. J Gastrointest Surg 11:445–451 Chiariotti L, Berlingieri MT, Battaglia C et al, 1995, Expression of galectin-1 in normal human tyroid gland and in differentiated and poorly differentiated tyroid tumors. Int J Cancer 64:171–175 van den Brule F, Califice S, Garnier F et al, 2003, Galectin-1 accumulation in the ovary carcinoma peritumoral stroma is induced by ovary carcinoma cells and affects both cancer cell proliferation and adhesion to laminin-1 and fibronectin. Lab Invest 83:377–386 Shimonishi T, Miyazaki K, Kano N et al, 2001, Expression of endogenous galectin-1 and galectin-3 in intrahepatic cholangiocarcinoma. Hum Pathol 32:302–310 Berberat PO, Friess H, Wang L et al, 2001, Comparative analysis of galectins in primary tumors and tumor metastasis in human pancreatic cancer. J Histochem Cytochem 49:539–549 Cindolo L, Benvenuto G, Salvatore G et al, 1999, Galectin-1 and galectin-3 expression in human bladder transitional-cell carcinoma. Int J Cancer 84:39–43 Cooper DN, Barondes SH, 1990, Evidence for export of a muscle lectin from cytosol to extracellular matrix and for a novel secretory mechanism. J Cell Biol 110:1681–1691 van den Brule F, Waltregny D, Castronovo V, 2001, Increased expression of galectin-1 in carcinoma-associated stroma predicts poor outcome in prostate carcinomapatients. J Pathol 193:80–87 Zhu XL, Liang L, Ding YQ, 2007, Galectin-1 expression in human colorectal carcinoma and its clinical significance. Nan Fang Yi Ke Da Xue Xue Bao 27:1331–1334 Le QT, Shi G, Cao H et al, 2005, Galectin-1: a link between tumor hypoxia and tumor immune privilege. J Clin Oncol 23:8932–8941 Rorive S, Belot N, Decaestecker C et al, 2001, Galectin-1 is highly expressed in human gliomas with relevance for modulation of invasion of tumor astrocytes into the brain parenchyma. Glia 33:241–255 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 569 EP 577 DI 10.1007/s12253-010-9248-8 PG 9 ER PT J AU Mahjour, BS Ghaffarpasand, F Fattahi, JM Ghaderi, A Fotouhi Ghiam, A Karimi, M AF Mahjour, Babak Seyed Ghaffarpasand, Fariborz Fattahi, Javad Mohammad Ghaderi, Abbas Fotouhi Ghiam, Alireza Karimi, Mehran TI Seroprevalence of Human Herpes Simplex, Hepatitis B and Epstein-Barr Viruses in Children with Acute Lymphoblastic Leukemia in Southern Iran SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute Lymphoblastic Leukemia (ALL); Epstein-Barr Virus (EBV); Hepatitis B Virus (HBV); Human Herpes Simplex Virus (HSV); Iran ID Acute Lymphoblastic Leukemia (ALL); Epstein-Barr Virus (EBV); Hepatitis B Virus (HBV); Human Herpes Simplex Virus (HSV); Iran AB To investigate the seroprevalence of Herpes Simplex Viruses (HSV1 and HSV2), Ebstein-Barr Virus (EBV) and Hepatitis B Virus (HBV) in children with acute lymphoblastic leukemia (ALL) in southern Iran. 90 patients with ALL and 90 age-sex matched healthy participants were enrolled in this study. Antibodies (IgG) against HBsAg, HSV1, HSV2, EBV different antigens including Epstein-Barr nuclear antigen-1 (EBNA-1), viral capsid antigen (VCA) and early antigen (EA) were measured by enzyme-linked immunosorbent assay (ELISA). There were 54 (60%) male and 36 (40%) female in both study groups with mean age of 8.47±3.61 and 8.61±2.84 years in case and control group respectively (P=0.812). The prevalence of antibodies against HBsAg (P=0.002), HSV1 (P<0.0001), VCA (P=0.021) and EA (P<0.0001) antigens of EBV were significantly higher in ALL patients. With the results of this study, we could not exclude a connection between these viral infections and later leukemogenesis in childhood ALL, although nor latent infection nor congenital infection cannot be excluded by this method. C1 [Mahjour, Babak Seyed] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Ghaffarpasand, Fariborz] Fasa University of Medical SciencesFasa, Iran. [Fattahi, Javad Mohammad] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Ghaderi, Abbas] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Fotouhi Ghiam, Alireza] McGill University, Douglas Hospital, Mental Health University InstituteMontreal, Quebec, Canada. [Karimi, Mehran] Shiraz University of Medical Sciences, Nemazee Hospital, Hematology Research CenterShiraz, Iran. RP Karimi, M (reprint author), Shiraz University of Medical Sciences, Nemazee Hospital, Hematology Research Center, Shiraz, Iran. EM karimim@sums.ac.ir CR Greenlee RT, Murray T, Bolden S, Wingo PA, 2000, Cancer statistics, 2000. CA Cancer J Clin 50:7–33 Kinlen L, 1988, Evidence for an infective cause of childhood leukemia: comparison of a Scottish new town with nuclear reprocessing sites in Britain. Lancet 2:1323–1327 Kinlen L, Doll R, 2004, Population mixing and childhood leukaemia: fallon and other US clusters. Br J Cancer 91:1–3 Greaves M, 2006, Infection, immune responses and the aetiology of childhood leukaemia. Nat Rev Cancer 6:193–203 Weitzman MD, Ornelles DA, 2005, Inactivating intracellular antiviral responses during adenovirus infection. Oncogene 24:7686–7696 Wilkinson DE, Weller SK, 2006, Herpes simplex virus type I disrupts the ATR-dependent DNA-damage response during lytic infection. J Cell Sci 119:2695–2703 Sonmez M, Bektas O, Yilmaz M, Durmus A, Akdogan E, Topbas M, Erturk M, Ovali E, Omay SB, 2007, The relation of lymphoma and hepatitis B virus/hepatitis C virus infections in the region of east black sea, Turkey. Tumori 93:536–539 Priftakis P, Dalianis T, Carstensen J, Samuelsson U, Lewensohn- Fuchs I, Bogdanovic G, Winiarski J, Gustafsson B, 2003, Human polyomavirus DNA is not detected in Guthrie cards, dried blood spots, from children who developed acute lymphoblastic leukemia. Med Pediatr Oncol 40:219–223 Bogdanovic G, Jernberg AG, Priftakis P, Grillner L, Gustafsson B, 2004, Human herpes virus 6 or Epstein-Barr virus were not detected in Guthrie cards from children who later developed leukaemia. Br J Cancer 91:913–915 Gustafsson B, Jernberg AG, Priftakis P, Bogdanovic G, 2006, No CMV DNA in Guthrie cards from children who later developed ALL. Pediatr Hematol Oncol 23:199–205 Isa A, Priftakis P, Broliden K, Gustafsson B, 2004, Human parvovirus B19 DNA is not detected in Guthrie cards from children who have developed acute lymphoblastic leukemia. Pediatr Blood Cancer 42:357–360 Greaves M, 1999, Molecular genetics, natural history and the demise of childhood leukaemia. Eur J Cancer 35:1941–1953 Wiemels JL, Cazzaniga G, Daniotti M, Eden OB, Addison GM, Masera G, Saha V, Biondi A, Greaves MF, 1999, Prenatal origin of acute lymphoblastic leukaemia in children. Lancet 354:1499– 1503 Kinlen L, Jiang J, Hemminki K, 2002, A case-control study of childhood leukaemia and paternal occupational contact level in rural Sweden. Br J Cancer 86:732–737 Kinlen LJ, Balkwill A, 2001, Infective cause of childhood leukaemia and wartime population mixing in Orkney and Shetland. Lancet 358:155 Petridou E, Revinthi K, Alexander FE, Haidas S, Koliouskas D, Kosmidis H, Piperopoulou F, Tzortzatou F, Trichopoulos D, 1996, Space-time clustering of childhood leukaemia in Greece: evidence supporting a viral aetiology. Br J Cancer 73:1278– 1283 Gilman EA, McNally RJ, Cartwright RA, 1999, Space-time clustering of acute lymphoblastic leukaemia in parts of the U.K., 1984–1993). Eur J Cancer 35:91–96 Gustafsson B, Carstensen J, 1999, Evidence of space-time clustering of childhood acute lymphoblastic leukaemia in Sweden. Br J Cancer 79:655–657 Gustafsson B, Carstensen J, 2000, Space-time clustering of childhood lymphatic leukaemias and non-Hodgkin’s lymphomas in Sweden. Eur J Epidemiol 16:1111–1116 Birch JM, Alexander FE, Blair V, Eden OB, Taylor GM, McNally RJ, 2000, Space-time clustering patterns in childhood leukaemia support a role for infection. Br J Cancer 82:1571–1576 Knox EG, Stewart A, Kneale G, 1980, Childhood leukaemia and mother—foetus infection. Br J Cancer 42:158–161 Gustafsson B, Huang W, Bogdanovic G, Gauffin F, Nordgren A, Talekar G, Ornelles DA, Gooding LR, 2007, Adenovirus DNA is detected at increased frequency in Guthrie cards from children who develop acute lymphoblastic leukaemia. Br J Cancer 97:992– 994 Loutfy SA, 2006, HM, Ibrahim MF, Hafez MM. Seroprevalence of herpes simplex virus types 1 and 2, Epstein-Barr virus, and cytomegalovirus in children with acute lymphoblastic leukemia in Egypt. Saudi Med J 27:1139–1145 Burkitt D, 1958, A sarcoma involving the jaws in African children. Br J Surg 46:218–223 EpsteinMA, Achong BG, Barr YM(1964)Virus particles in cultured lymphoblasts from Burkitt’s lymphoma. Lancet 15:702–703 Costa S, Barrasso R, Terzano P, Zerbini M, Carpi C, Musiani M, 1985, Detection of active Epstein–Barr infection in pregnant women. Eur J Clin Microbiol 4:335–336 Lehtinen M, Koskela P, Ogmundsdottir HM, Bloigu A, Dillner J, Gudnadottir M, Hakulinen T, Kjartansdottir A, Kvarnung M, Pukkala E, Tulinius H, Lehtinen T, 2003, Maternal herpesvirus infections and risk of acute lymphoblastic leukemia in the offspring. Am J Epidemiol 158:207–213 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 579 EP 582 DI 10.1007/s12253-010-9258-6 PG 4 ER PT J AU Kakihana, K Ohashi, K Akiyama, H Sakamaki, H AF Kakihana, Kazuhiko Ohashi, Kazuteru Akiyama, Hideki Sakamaki, Hisashi TI Correlation Between Survival and Number of Mobilized CD34+ Cells in Patients with Multiple Myeloma or Waldenstrom Macroglobulinemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mobilized CD34+ cell number; Multiple myeloma; Waldenstrom macroglobulinemia; Autologous stem cell transplantation ID Mobilized CD34+ cell number; Multiple myeloma; Waldenstrom macroglobulinemia; Autologous stem cell transplantation AB High-dose chemotherapy followed by autologous stem cell transplantation is the established treatment for symptomatic multiple myeloma (MM) or Waldenstrom macroglobulinemia (WM). We retrospectively analyzed the impact of mobilized CD34+ cell number on clinical outcomes in patients with MM or WM who underwent autologous stem cell transplantation in our hospital from 1997 to 2007. A total of 39 patients were identified. All patients received peripheral stem cell support after a conditioning regimen. We defined patients with collection of a large number (≥ 8×106/kg) of CD34+ cells as super mobilizers (SM), and all others as normal mobilizers (NM). Although hematological engraftment was earlier in the SM group, overall survival did not differ significantly between groups (P=0.392). Likewise, no significant differences were seen in progression-free survival (P=0.201) or survival after relapse (P=0.330). In conclusion, our retrospective study could not find any correlation between survival and number of mobilized CD34+ cells, in contrast to previously reported results. C1 [Kakihana, Kazuhiko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22, Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Ohashi, Kazuteru] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22, Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Akiyama, Hideki] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22, Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Sakamaki, Hisashi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22, Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. RP Ohashi, K (reprint author), Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 113-8677 Tokyo, Japan. EM k.ohashi@cick.jp CR Attal M, Harousseau JL, Stoppa AM et al, 1996, A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Francais du Myelome. N Engl J Med 335:91–97 Barlogie B, Jagannath S, Vesole DH et al, 1997, Superiority of tandem autologous transplantation over standard therapy for previously untreated multiple myeloma. Blood 89:789–793 Child JA, Morgan GJ, Davies FE et al, 2003, High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med 348:1875–1883 Fermand JP, Katsahian S, Divine M et al, 2005, High-dose therapy and autologous blood stem-cell transplantation compared with conventional treatment in myeloma patients aged 55 to 65 years: long-term results of a randomized control trial from the Group Myelome-Autogreffe. J Clin Oncol 23:9227–9233 Stockerl-Goldstein KE, Reddy SA, Horning SF et al, 2000, Favorable treatment outcome in non-Hodgkin’s lymphoma patients with “poor” mobilization of peripheral blood progenitor cells. Biol Blood Marrow Transplant 6:506–512 Hiwase DK, Hiwase S, Bailey M et al, 2008, Higher infused lymphocyte dose predicts higher lymphocyte recovery, which in turn, predicts superior overall survival following autologous hematopoietic stem cell transplantation for multiple myeloma. Biol Blood Marrow Transplant 14:116–124 Bolwell BJ, Pohlman B, Rybicki L et al, 2007, Patients mobilizing large numbers of CD34+ cells, ‘super mobilizers’, have improved survival in autologous stem cell transplantation for lymphoid malignancies. Bone Marrow Transplant 40:437–441 Wang S, Nademanee A, Qian D et al, 2007, Peripheral blood hematopoietic stem cell mobilization and collection efficacy is not an independent prognostic factor for autologous stem cell transplantation. Transfusion 47:2207–2216 Galimberti S, Morabito F, Guerrini F et al, 2003, Peripheral blood stem cell contamination evaluated by a highly sensitive molecular method fails to predict outcome of autotransplanted multiple myeloma patients. Br J Haematol 120:405–412 Gordan LN, Sugrue MW, Lynch JW et al, 2003, Poor mobilization of peripheral blood stem cells is a risk factor for worse outcome in lymphoma patients undergoing autologous stem cell transplantation. Leuk Lymphoma 44:815–820 O’Shea D, Giles C, Terpos E et al, 2006, Predictive factors for survival in myeloma patients who undergo autologous stem cell transplantation: a single-centre experience in 211 patients. Bone Marrow Transplant 37:731–737 Krejci M, Buchler T, Hajek R et al, 2005, Prognostic factors for survival after autologous transplantation: a single centre experience in 133 multiple myeloma patients. Bone Marrow Transplant 35:159–164 Wahlin A, Eriksson M, Hultdin M, 2004, Relation between harvest success and outcome after autologous peripheral blood stem cell transplantation in multiple myeloma. Eur J Haematol 73:263–268 Krejci M, Hajek R, Buchler T et al, 2007, Simple variables predict survival after autologous transplantation: a single centre experience in 181 multiple myeloma patients. Neoplasma 54:143–148 Dean R, Masci P, Pohlman B et al, 2005, Dendritic cells in autologous hematopoietic stem cell transplantation for diffuse large B-cell lymphoma: graft content and post transplant recovery predict survival. Bone Marrow Transplant 36:1049–1052 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 583 EP 587 DI 10.1007/s12253-009-9238-x PG 5 ER PT J AU Fang, Y Zhao, L Zang, M Chen, S Yan, F Di, X Duren, A AF Fang, Yujiang Zhao, Lei Zang, Meifu Chen, Songsen Yan, Feng Di, Xu Duren, Alicia TI A Dispensable Role for P450scc in the Overproduction of Aldosterone in Aldosterone-Producing Adenoma and Idiopathic Hyperaldosteronism in Patients with Primary Aldosteronism SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Aldosteronism; Adrenocortical adenomas; Idiopathic hyepraldosteronism; P450scc ID Aldosteronism; Adrenocortical adenomas; Idiopathic hyepraldosteronism; P450scc AB Our previous study suggests that cytochrome P-450 carbon 17α-hydroxylase/17,20-lyase (P450c17α) correlated with the overproduction of aldosterone in aldosterone-producing adenoma (APA) and idiopathic hyperaldosteronism (IHA) in patients with primary aldosteronism. To further investigate if cytochrome P-450 cholesterol side-chain cleavage enzyme (P450scc) contributes to the overproduction of aldosterone in APA and IHA and if its mRNA expression differs in APA and IHA in patients with primary aldosteronism, we studied the expression of P450scc mRNA in APA and idiopathic hyperplastic nodules. Total RNA was extracted from APA of eight patients diagnosed as APA, idiopathic hyperplastic nodules of four patients diagnosed as IHA, seven normal adrenal glands and one normal muscle tissue. P450scc mRNA was examined by Northern blot analysis. No significant difference in P450scc mRNA was found among normal adrenal gland, APA or idiopathic hyperplastic nodules (P>0.05). These results suggest that P450scc contributes little to the overproduction of aldosterone in APA and IHA and cannot be considered as a marker to differentiate between them in patients with primary aldosteronism. C1 [Fang, Yujiang] University of Missouri, Department of Internal Medicine, 65211 Columbia, MO, USA. [Zhao, Lei] Wannan Medical College, Department of Internal Medicine, 241004 Wuhu, China. [Zang, Meifu] Pekin Union Medical College Hospital, Department of Urology, 100073 Beijing, China. [Chen, Songsen] Chinese Academy of Medical Sciences, Department of Biochemistry and Molecular Biology, 100073 Beijing, China. [Yan, Feng] Wannan Medical College, Department of Internal Medicine, 241004 Wuhu, China. [Di, Xu] Chinese Academy of Medical Sciences, Department of Biochemistry and Molecular Biology, 100073 Beijing, China. [Duren, Alicia] University of Missouri, Department of Internal Medicine, 65211 Columbia, MO, USA. RP Fang, Y (reprint author), University of Missouri, Department of Internal Medicine, 65211 Columbia, USA. EM fangy@health.missouri.edu CR Gavras I, 2007, The incidentally discovered adrenal mass. N Engl J Med 356:2005–2006 Dluhy RG, Williams GH, 2004, Aldosterone–villain or bystander? N Engl JMed 351:8–10 Ganguly A, 1998, Primary aldosteronism. N Engl J Med 339:1828–1834 Fang Y, Zang M, Chen S, Di X, 2000, Expression of cytochrome P-450c17α-hydroxynase mRNA in adrenocortical adenomas and hyperplasia from patients with primary aldosteronism and its significance. Basic Med Sci Clin 20:30–33, Chinese Chomczynski P, Sacchi N, 1987, Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem 162:156–159 Chung B, Matteson KJ, Voutilainen R, Mohandas TK, Miller WL, 1986, Human cholesterol side chain cleavage enzyme, P450scc: cDNAcloning, assignment of the gen to chromosone 15, and expression in the placenta. Proc Natl Acad Sci USA 83:8962– 8966 Morohashi L, Fujii-Kuriyama Y, Okada Y, Sogawa K, Hirose T, Inayama S, Omura T, 1984, Molecular cloning and nucleotide sequence of cDNA for mRNAof mitochondrial P450scc of bovine adrenal cortex. Proc Natl Acad Sci USA 81:4647 Morohashi K, Sogawa K, Omura T, Fujii-Luriyama Y, 1987, Gene structure of human cytochrome P450scc, cholesterol desmolase. J Biochem 101:879–887 Sparkea RS, Klisak I, Miller WL, 1991, Regional mapping of grnes encoding hyman steroidogenic enzymes: P450scc to 15q24- q25; and P450c17 to 10q24-q25. DNA Cell Biol 10:359–365 Ogo A, Haji M, Ohashi M, Nawata H, 1991, Expression of adrenocortical adenomas from patients with primary aldosteronism. Mol Cell Endocrinol 76:7–12 Shibata H, Suzuki H, Ogishima T, Ishimura Y, Saruta T, 1993, Significance of steroidogenic enzymes in the pathogenesis of adrenal tumour. Acta Endocrinol 128:235–242 Bassett MH, Mayhew B, Rehman K, White PC, Mantero F, Arnaldi G, Stewart PM, Bujalska I, Rainey WE, 2005, Expression profiles for steroidogenic enzymes in adrenocortical disease. J Clin Endocrinol Metab 90:5446–5455 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 589 EP 592 DI 10.1007/s12253-009-9237-y PG 4 ER PT J AU Frohlich, G Agoston, P Lovey, J Polgar, Cs Major, T AF Frohlich, Georgina Agoston, Peter Lovey, Jozsef Polgar, Csaba Major, Tibor TI The Effect of Needle Number on the Quality of High-dose-rate Prostate Brachytherapy Implants SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Dose-volume analysis; High-dose-rate; Prostate brachytherapy; Number of needles; Organs at risk; Treatment planning ID Dose-volume analysis; High-dose-rate; Prostate brachytherapy; Number of needles; Organs at risk; Treatment planning AB The aim of this study is to evaluate the effect of the number of needles on the quality of dose distributions in high-dose-rate (HDR) prostate implants regarding target coverage, dose homogeneity and dose to organs at risk. Treatment plans of 174 implants were evaluated using cumulative dose-volume histograms. The plans were divided into three groups according to the number of implanted needles: <15: LNG (low number group), 15–17: MNG (medium number group) and >17: HNG (high number group). Treatment planning was based on transrectal ultrasound imaging. Dose-volume parameters for target (V90, V100, V150, V200, D90, Dmin) and quality indices (DNR, DHI, CI, COIN) were calculated. Maximal dose in reference points and high dose volumes were determined for rectum and urethra. Nonparametric analysis of variance and correlation was used with regard to needle numbers. Between the groups differences were found only in the following parameters: Vp was larger when more needles were used with the values of 22.8 cm3, 28.0 cm3 and 30.9 cm3 for the three groups, and more needles were used when the central cross-section of the prostate was larger. V200 in MNG was lower than in LNG (12%, 14%). Dose to rectum was higher in MNG than in LNG (D2: 51%, 47%). Doses to the urethra were higher in HNG than in MNG (D1: 142%, 137%, and D0.1: 128%, 125%). There was no significant difference in other parameters. Different number of needles results significant differences in treatment plans. However, the optimal needle number depends on not only the size of the prostate, but also the individual anatomy of the patient. Based on our results, in most cases the use of 15–17 needles seems to provide a dosimetrically acceptable treatment plan in HDR prostate implants. C1 [Frohlich, Georgina] Semmelweis University, School of PhD Studies, Ulloi ut 26, 1085 Budapest, Hungary. [Agoston, Peter] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. [Lovey, Jozsef] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. [Polgar, Csaba] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. [Major, Tibor] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. RP Frohlich, G (reprint author), Semmelweis University, School of PhD Studies, 1085 Budapest, Hungary. EM gfrohlich@oncol.hu CR Akimoto T, Ito K, Saitoh JI et al, 2005, Acute genitourinary toxicity after high-dose-rate, HDR, brachytherapy combined with hypofractionated external-beam radiation therapy for localized prostate cancer: correlation between the urethral dose in HDR brachytherapy and the severity of acute genitourinary toxicity. Int J Radiat Oncol Biol Phys 63:463–471 Akimoto T, Katoh H, Noda SE et al, 2005, Acute genitourinary toxicity after high-dose-rate, HDR, brachytherapy combined with hypofractionated external-beam radiation therapy for localized prostate cancer: second analysis to determine the correlation between the urethral dose in HDR brachytherapy and the severity of acute genitourinary toxicity. Int J Radiat Oncol Biol Phys 63:472–478 Demanes DJ, Rodriguez RR, Altieri GA, 2000, High dose rate prostate brachytherapy: the California Endocurietherapy, CET, Method. Radiother Oncol 57:289–296 Demanes DJ, Rodriguez RR, Schour LD et al, 2005, High-doserate intensity-modulated brachytherapy with external beam radiotherapy for prostate cancer: California Endocurietherapy’s 10-year results. Int J Radiat Oncol Biol Phys 61:1306–1316 Hsu ICJ, Cabrera AR, Weinberg V et al, 2005, Combined modality treatment with high-dose-rate brachytherapy boost for locally advanced prostate cancer. Brachytherapy 4:202–206 Jo J, Hiratsuka J, Fujii T et al, 2004, High-dose-rate Iridium-192 afterloading therapy combined with external beam radiotherapy for T1c-T3bN0M0 prostate cancer. Urology 64:556–560 Kovacs G, Melchert C, Sommerauer M et al, 2007, Intensity modulated high-dose-rate brachytherapy boost complementary to external beam radiation for intermediate- and high-risk localized prostate cancer patients—How we do it in Lubeck/Germany. Brachytherapy 6:142–148 Mate TP, Gottesman JE, Hatton J et al, 1998, High-dose-rate afterloading 192-Iridium prostate brachytherapy: feasibility report. Int J Radiat Oncol Biol Phys 41:525–533 Martin T, Hey-Koch S, Strassmann G et al, 2000, 3D interstitial HDR brachytherapy combined with 3D external beam radiotherapy and androgen deprivation for prostate cancer. Strahlenther Onkol 176:361–367 Pinkawa M, Fischedick K, Treusacher P et al, 2006, Dose-volume impact in high-dose-rate Iridium-192 brachytherapy as a boost to external beam radiotherapy for localized prostate cancer—a phase II study. Radiother Oncol 78:41–46 Vargas CE, Ghilezan M, Hollander M et al, 2005, A new model using number of needles and androgen deprivation to predict chronic urinary toxicity for high or low dose rate prostate brachytherapy. J Urol 174:882–887 Charra-Brunaud C, Hsu ICJ, Weinberg V et al, 2003, Analysis of interaction between number of implant catheters and dose-volume histograms in prostate high-dose-rate brachytherapy using a computer model. Int J Radiat Oncol Biol Phys 56:586–591 D’Amico AV, Whittington R, Malkowicz B et al, 1998, Biochemical outcome after radical prostatectomy, external beam radiation therapy, or interstitial radiation therapy for clinically localized prostate cancer. JAMA 280:969–974 Baltas D, Kolotas C, Geramani K et al, 1998, A conformal index, COIN, to evaluate implant quality and dose specification in brachytherapy. Int J Radiat Oncol Biol Phys 40:515–524 Hoskin PJ, 2000, High-dose-rate brachytherapy boost treatment in radical radiotherapy for prostate cancer. Radiother Oncol 57:285– 288 Morton GC, 2005, The emerging role of high-dose-rate brachytherapy for prostate cancer. Clin Oncol 17:219–227 Sathya JR, Davis IR, Julian JA et al, 2005, Randomized trial comparing Iridium implant plus external-beam radiation therapy with external-beam radiation therapy alone in node-negative locally advanced cancer of the prostate. J Clin Oncol 23:1192– 1199 Stromberg J, Martinez A, Gonzalez J et al, 1995, Ultrasoundguided high-dose-rate conformal brachytherapy boost in prostate cancer: treatment description and preliminary results of a phase I/II clinical trial. Int J Radiat Oncol Biol Phys 33:161–171 Martinez AA, Pataki I, Edmundson G et al, 2001, Phase II prospective study of the use of conformal highdose-rate brachytherapy as monotherapy for the treatment of favourable stage prostate cancer: a feasibility report. Int J Radiat Oncol Biol Phys 49:61–69 Yoshioka Y, Nose T, Yoshida K et al, 2003, High-dose-rate brachytherapy as monotherapy for localized prostate cancer: a retrospective analysis with special focus on tolerance and chronic toxicity. Int J Radiat Oncol Biol Phys 56:213–220 Edmundson GK, Rizzo NR, Teahan M et al, 1993, Concurrent treatment planning for outpatient high-dose-rate prostate template implants. Int J Radiat Oncol Biol Phys 27:1215–1223 Jacob D, Raben A, Sarkar A et al, 2008, Anatomy-based inverse planning simulated annealing optimization in high-dose-rate prostate brachytherapy: significant dosimetric advantage over other optimization techniques. Int J Radiat Oncol Biol Phys 72:820–827 Kolkman-Deurloo IKK, Deleye XGJ, Jansen PP et al, 2004, Anatomy based inverse planning in HDR prostate brachytherapy. Radiother Oncol 73:73–77 Nickers P, Lenaerts E, Thissen B et al, 2005, Does inverse planning applied to Iridium192 high dose rate prostate brachytherapy improve the optimization of the dose afforded by the Paris system? Radiother Oncol 74:131–136 Yoshioka Y, Nishimura T, Kamata M et al, 2005, Evaluation of anatomy-based dwell position and inverse optimization in highdose-rate brachytherapy of prostate cancer: a dosimetric comparison to a conventional cylindrical dwell position, geometric optimization, and dose-point optimization. Radiother Oncol 75: 311–317 Ash D, Flynn A, Battermann J et al, 2000, ESTRO/EAU/EORTC recommendations on permanent seed implantation for localized prostate cancer. Radiother Oncol 57:315–321 Nag S, Bice W, Dewyngaert K et al, 2000, The American Brachytherapy Society recommendations for permanent prostate brachytherapy postimplant dosimetric analysis. Int J Radiat Oncol Biol Phys 46:221–230 Kovacs G, Potter R, Loch T et al, 2005, GEC/ESTRO-EAU recommendations on temporary brachytherapy using stepping sources for localised prostate cancer. Radiother Oncol 74:37–148 Edmundson GK, Yan D, Martinez AA, 1995, Intraoperative optimization of needle placement and dwell times for conformal prostate brachytherapy. Int J Radiat Oncol Biol Phys 33:1257– 1263 Khoo VS, 2005, Radiotherapeutic techniques for prostate cancer, dose escalation and brachytherapy. Clin Oncol 17:560–571 Lakosi F, Antal G, Vandulek C et al, 2009, Technical feasibility of transperineal MR-guided prostate interventions in a low-field open MRI unit: canine study. Pathol Oncol Res 15(3):315–322 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 593 EP 599 DI 10.1007/s12253-010-9252-z PG 7 ER PT J AU Dundr, P Fischerova, D Povysil, C Berkova, A Bauerova, L Cibula, D AF Dundr, Pavel Fischerova, Daniela Povysil, Ctibor Berkova, Adela Bauerova, Lenka Cibula, David TI Uterine Tumors with Neuroectodermal Differentiation. A Report of 4 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Uterine tumors; PNET; Ewing sarcoma; EWSR1 gene; Neuroectodermal ID Uterine tumors; PNET; Ewing sarcoma; EWSR1 gene; Neuroectodermal AB We report four cases of uterine tumors with neuroectodermal differentiation. One tumor had neuroectodermal component only; in the three other tumors, the neuroectodermal component was admixed with another component, namely rhabdomyosarcoma (1 case), and endometrioid adenocarcinoma (2 cases). Histologically, the neuroectodermal component consisted of small to medium sized cells arranged in diffuse sheets. The tumor cells had round nuclei with stippled to coarsely granular chromatin, mostly with non-prominent nucleoli, and scant eosinophilic or amphophilic cytoplasm. Immunohistochemically, 4/4 tumors showed expression of vimentin, synaptophysin and CD56; 3/4 tumors were CD99 and NSE positive; 2/4 tumors showed focal expression of S-100 protein; and 1/4 tumors had focal dot-like cytoplasmic positivity for cytokeratin AE1/AE3. FLI-1 was negative in all cases. FISH examination was performed and none of the tumors showed rearrangement of EWSR1 gene. Uterine tumors with neuroectodermal differentiation are rare; to the best of our knowledge only 44 cases have been reported in the literature to date, referred to as Ewing sarcoma, peripheral PNET (pPNET), PNET (not otherwise specified) and uterine tumors with neuroendocrine differentiation. C1 [Dundr, Pavel] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 128 00 Prague, Czech Republic. [Fischerova, Daniela] Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Obstetrics and Gynecology, Apolinarska 18, 2 120 00 Prague, Czech Republic. [Povysil, Ctibor] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 128 00 Prague, Czech Republic. [Berkova, Adela] Charles University in Prague and General University Hospital in Prague, First Faculty of Medicine, 1st Department of Medicine—Department of Haematology, U nemocnice 2, 2 128 00 Prague, Czech Republic. [Bauerova, Lenka] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 128 00 Prague, Czech Republic. [Cibula, David] Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Obstetrics and Gynecology, Apolinarska 18, 2 120 00 Prague, Czech Republic. RP Dundr, P (reprint author), Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, 128 00 Prague, Czech Republic. EM pdundr@seznam.cz CR Wick MR, 2000, Immunohistology of neuroendocrine and neuroectodermal tumors. Semin Diagn Pathol 3:194–203 Farley J, O’Boyle JD, Heaton J, Remmenga S, 1997, Extraosseous Ewing sarcoma of the vagina. Obstet Gynecol 96:832–834 Habib K, Finet JF, Plantier F et al, 1992, Rare lesion of the vulva. Arch Anat Cytol Pathol 40:158–159 Kawauchi S, Fukuda T, Miyamoto S et al, 1998, Peripheral primitive neuroectodermal tumor of the ovary confirmed by CD99 immunostaining, karyotypic analysis, and RT-PCR for EWS/FLI- 1 chimeric mRNA. Am J Surg Pathol 22:1417–1422 Delattre O, Zucman J, Melot T et al, 1994, The Ewing family of tumors-a subgroup of small-round-cell tumors defined by specific chimeric transcripts. N Engl J Med 331:294–299 Akbayir O, Gungorduk K, Rafioglu G et al, 2008, Primary primitive neuroectodermal tumor of the uterus: a case report. Arch Gynecol Obstet 277:345–348 Blattner JM, Gable P, Quigley MM, McHale MT, 2007, Primitive neuroectodermal tumor of the uterus. Gynecol Oncol 106:419–422 Celik H, Gurates B, Karaoglu A et al, 2009, Uterine primitive neuroectodermal tumor: a case report. Arch Gynecol Obstet 279:259– 261 Christensen B, Schmidt U, Schindler A, 1994, Primitive neuroectodermal tumor of the uterus—a monophasic differentiated mixed Mullerian tumor? Zentralbl Gynakol 116:577–580 Daya D, Lukka H, Clement PB, 1992, Primitive neuroectodermal tumors of the uterus: a report of four cases. Hum Pathol 23:1120– 1129 Euscher ED, Deavers MT, Lopez-Terrada D et al, 2008, Uterine tumors with neuroectodermal differentiation: a series of 17 cases and review of the literature. Am J Surg Pathol 32:219–228 Fraggetta F, Magro G, Vasquez E, 1997, Primitive neuroectodermal tumour of the uterus with focal cartilaginous differentiation. Histopathology 30:483–485 Fukunaga M, Nomura K, Endo Y, Ushigome S, Aizawa S, 1996, Carcinosarcoma of the uterus with extensive neuroectodermal differentiation. Histopathology 29:565–570 Karseladze AI, Filipova NA, Navarro S, Llombart-Bosch A, 2001, Primitive neuroectodermal tumor of the uterus. A case report. J Reprod Med 46:845–848 Majeed U, Ilyas MA, Uddin N et al, 2009, Primary Ewing’s sarcoma-primitive neuroectodermal tumour of uterus. J Obstet Gynaecol 29:73–74 Mittal S, Sumana G, Gupta M, Gupta B, 2007, Primitive neuroectodermal tumor of the uterus: a case report. Int J Gynecol Cancer 17:524–527 Molyneux AJ, Deen S, Sundaresan V, 1992, Primitive neuroectodermal tumour of the uterus. Histopathology 21:584–585 Ng SB, Sittampalam K, Chuah KL, 2002, Primitive neuroectodermal tumours of the uterus: a case report with cytological correlation and review of the literature. Pathology 34:455–461 Odunsi K, Olatinwo M, Collins Y et al, 2004, Primary primitive neuroectodermal tumor of the uterus: a report of two cases and review of the literature. Gynecol Oncol 92:689–696 Park JY, Lee S, Kang HJ, Kim HS, Park SY, 2007, Primary Ewing’s sarcoma-primitive neuroectodermal tumor of the uterus: a case report and literature review. Gynecol Oncol 106: 427–432 Peres E, Mattoo TK, Poulik J, Warrier I, 2005, Primitive neuroectodermal tumor, PNET, of the uterus in a renal allograft patient: a case report. Pediatr Blood Cancer 44:283–285 Quddus MR, Rashid L, Sung J et al, 2009, Ewing´s sarcoma / Peripheral primitive neuroectodermal tumor, ES/PNET, differentiation in endometrial serous carcinomas. Reprod Sci 16:591– 595 Rose PG, O’Toole RV, Keyhani-Rofagha S, Qualman S, Boutselis JG, 1987, Malignant peripheral primitive neuroectodermal tumor of the uterus. J Surg Oncol 35:165–169 Sinkre P, Albores-Saavedra J, Miller DS, Copeland LJ, Hameed A, 2000, Endometrial endometrioid carcinomas associated with Ewing sarcoma/peripheral primitive neuroectodermal tumor. Int J Gynecol Pathol 19:127–132 Sorensen JB, Schultze HR, Madsen EL, Holund B, 1998, Primitive neuroectodermal tumor, PNET, of the uterine cavity. Eur J Obstet Gynecol Reprod Biol 76:181–184 Taieb S, Cabaret V, Bonodeau F, Leblanc E, Besson P, 1998, MRI of primitive neuroectodermal tumor of the uterus. J Comput Assist Tomogr 22:896–898 Varghese L, Arnesen M, 2006, Boente M, 2006, Primitive neuroectodermal tumor of the uterus: a case report and review of literature. Int J Gynecol Pathol 25:373–377 Venizelos ID, Zafrakas M, Dragoumis K et al, 2004, Primitive neuroectodermal tumor, PNET, of the uterine isthmus. Eur J Gynaecol Oncol 25:384–386 Gu M, Antonescu CR, Guiter G et al, 2000, Cytokereatin immunoreactivity in Ewing´s sarcoma: prevalence in 50 cases confirmed by molecular diagnostic studies. Am J Surg Pathol 24:410–416 Ushigome S, Machinami R, Sorensen PH, 2001, Ewing sarcoma / Primitive neuroectodermal tumor, PNET). In: Fletcher CDM, Unni KK, Mertens F, eds, WHO classification of tumours. Tumours of soft tissue and bones. IARC, Lyon, pp 297–300 Ellison DA, Parham DM, Bridge J, Beckwith JB, 2007, Immunohistochemistry of primary malignant neuroepithelial tumors of the kidney: a potential source of confusion? A study of 30 cases from the National Wilms Tumor Study Pathology Center. Hum Pathol 38:205–211 Tsuji S, Hisaoka M, Morimitsu Y et al, 1998, Peripheral primitive neuroectodermal tumour of the lung: report of two cases. Histopathology 33:369–374 Sato S, Yajima A, Kimura N et al, 1996, Peripheral neuroepithelioma, peripheral primitive neuroectodermal tumor, of the uterine cervix. Tohoku J Exp Med 180:187–195 Ward BS, Hitchcock CL, Keyhani S, 2000, Primitive neuroectodermal tumor of the uterus. A case report. Acta Cytol 44:667–672 Hachitanda Y, Tsuneyoshi M, Enjoji M, Nakagawara A, Ikeda K, 1990, Congenital primitive neuroectodermal tumor with epithelial and glial differentiation: an ultrastructural and immunohistochemical study. Arch Pathol Lab Med 114:101–105 Weidner N, Tjoe J, 1994, Immunohistochemical profile of monoclonal antibody O13: antibody that recognises glycoprotein p30/32MIC2 and is useful in diagnosing Ewing´s sarcoma and peripheral neuroepithelioma. Am J Surg Pathol 18:486–494 Mhawech-Fauceglia P, Herrmann F, Penetrante R et al, 2006, Diagnostic utility of FLI-1 monoclonal antibody and dualcolour, break-apart probe fluorescent in situ, FISH, analysis in Ewing´s sarcoma/primitive neuroectodermal tumour, EWS/ PNET). A comparative study with CD99 and FLI-1 polyclonal antibodies. Histopathology 49:569–575., DOI 10.1111/j.1365- 2559.2006.02535.x Rosi S, Orvieto E, Furlanetto A et al, 2004, Utility of the immunohistochemical detection of FLI-1 expression in round cell and vascular neoplasm using a monoclonal antibody. Mod Pathol 17:547–552., DOI 10.1038/modpathol.3800065 Berkova A, Dundr P, Povysil C, Melcakova S, Tvrdik D, 2008, A comparision of RT-PCR and FISH techniques in molecular diagnosis of Ewing’s sarcoma in paraffin-embedded tissue. Cesk Patol 44:67–70 Gersell DJ, Duncan DA, Fulling KH, 1989, Malignant mixed mullerian tumor of the uterus with neuroectodermal differentiation. Int J Gynecol Pathol 8:169–178 Young RH, Kleinman GM, Swcully RE, 1981, Glioma of the uterus. Report of a case with comments on histogenesis. Am J Surg Pathol 5:695–699 Lumadue JA, Askin FB, Perlman EJ, 1994, MIC2 analysis of small cell carcinoma. Am J Clin Pathol 102:692–694 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 601 EP 608 DI 10.1007/s12253-010-9249-7 PG 8 ER PT J AU Jain, KR Mehta, R Badve, S AF Jain, K Rohit Mehta, Rutika Badve, Sunil TI Conjunctival Squamous Cell Carcinoma Due to Ocular Prostheses: A Case Report and Review of Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Conjunctiva; Squamous cell carcinoma; Prostheses ID Conjunctiva; Squamous cell carcinoma; Prostheses AB Conjunctival squamous cell carcinoma (SCC) is a rare finding in everyday clinical practice, but is the most common malignancy of the ocular surface. The incidence of this malignancy in the United States is 0.03 per 100,000 persons. It is one extreme of a spectrum of lesions encompassed in ocular surface squamous neoplasia which range from dysplasia to carcinoma in situ to invasive SCC. Exposure to ultraviolet radiation B (UVB), human papilloma virus (HPV), and human immunodeficiency virus (HIV) infection of eroded ocular surface are important risk factors predisposing to the development of this malignancy. Herein we report a case of SCC arising in chronic conjunctival irritation due to prolonged prosthetic use following enucleation for traumatic eye injury. C1 [Jain, K Rohit] Indiana University, Department of Pathology, 350 W 11th Street, CPL 4010, 46202 Indianapolis, IN, USA. [Mehta, Rutika] Indiana University, Department of Pathology, 350 W 11th Street, CPL 4010, 46202 Indianapolis, IN, USA. [Badve, Sunil] Indiana University, Department of Pathology, 350 W 11th Street, CPL 4010, 46202 Indianapolis, IN, USA. RP Badve, S (reprint author), Indiana University, Department of Pathology, 46202 Indianapolis, USA. EM sbadve@iupui.edu CR McKelvie PA, Daniell M, McNab A, Loughnan M, Santamaria JD, 2002, Squamous cell carcinoma of the conjunctiva: a series of 26 cases. Br J Ophthalmol 86(2):168–173 Nguyen J, Ivan D, Esmaeli B, 2008, Conjunctival squamous cell carcinoma in the anophthalmic socket. Ophthal Plast Reconstr Surg 24(2):98–101 Newton R, Ziegler J, Ateenyi-Agaba C, Bousarghin L, Casabonne D, Beral V et al, 2002, The epidemiology of conjunctival squamous cell carcinoma in Uganda. Br J Cancer 87(3):301–308 Shields JA, Shields CL, 2007, Conjunctival invasive squamous cell carcinoma. In: Shields JA, Shields CL, eds, Eyelid, conjunctival, and orbital tumors: an atlas and textbook, 2nd ed. Lippincott Williams & Wilkins, New York, pp 292–306 Whittaker KW, Trivedi D, Bridger J, Sandramouli S, 2002, Ocular surface squamous neoplasia: report of an unusual case and review of the literature. Orbit, Amsterdam, Netherlands, 21, 3):209–215 Karcioglu ZA, Issa TM, 1997, Human papilloma virus in neoplastic and non-neoplastic conditions of the external eye. Br J Ophthalmol 81(7):595–598 Kaltreider SA, 2000, The ideal ocular prosthesis: analysis of prosthetic volume. Ophthal Plast Reconstr Surg 16(5):388–392 Kim JH, Lee MJ, Choung HK, Kim NJ, Hwang SW, Sung MS et al, 2008, Conjunctival cytologic features in anophthalmic patients wearing an ocular prosthesis. Ophthal Plast Reconstr Surg 24, 4):290–295 Mehta M, Fay A, 2009, Squamous cell carcinoma of the eyelid and conjunctiva. Int Ophthalmol Clin 49(1):111–121 Chang WJ, Tse DT, Rosa RH, Huang A, Johnson TE, Schiffman J, 2005, Conjunctival cytology features of giant papillary conjunctivitis associated with ocular prostheses. Ophthal Plast Reconstr Surg 21(1):39–45 Diepgen TL, Mahler V, 2002, The epidemiology of skin cancer. Br J Dermatol 146(Suppl 61):1–6 Tunc M, Char DH, Crawford B, Miller T, 1999, Intraepithelial and invasive squamous cell carcinoma of the conjunctiva: analysis of 60 cases. Br J Ophthalmol 83(1):98–103 Tuppurainen K, Raninen A, Kosunen O, Kankkunen JP, Kellokoski J, Syrjanen S et al, 1992, Squamous cell carcinoma of the conjunctiva. Failure to demonstrate HPV DNA by in situ hybridization and polymerase chain reaction. Acta Ophthalmol 70(2):248– 254 Hsu VJ, Agarwal MR, 2009, Re: “Conjunctival squamous cell carcinoma in the anophthalmic socket”. Ophthal Plast Reconstr Surg 25(3):257–258 Chaudhry TA, Memon M, Ahmad K, 2006, Use of artificial eye and conjunctival squamous cell carcinoma. J Postgrad Med 52(3):234–235 Endo T, Hata J, Togashi S, Yanagibayashi S, Nakayama Y, 2006, Conjunctival squamous cell carcinoma of the orbit 40 years after enucleation. Ophthal Plast Reconstr Surg 22(4):299–301 Campanella PC, Goldberg SH, Erlichman K, Abendroth C, 1998, Squamous cell tumors and ocular prostheses. Ophthal Plast Reconstr Surg 14(1):45–49 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 609 EP 612 DI 10.1007/s12253-010-9251-0 PG 4 ER PT J AU Terada, T AF Terada, Tadashi TI Minute Myopericytoma of the Neck: A Case Report with Literature Review and Differential Diagnosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Skin; Myopericytoma; Perivascular myoid cells; Histopathology; Immunohistochemistry ID Skin; Myopericytoma; Perivascular myoid cells; Histopathology; Immunohistochemistry AB Reports of cutaneous myopericytoma (MPC) are very rare. The author herein reports a case of minute MPC of the neck. A 56-year-old woman noticed a painful small tumor in the neck, and consulted to our hospital. Dermatologists’s diagnosis is a hyperplastic lymph node. Excision of the tumor was performed. Grossly, the tumor was a sold white tumor measuring 3×3×3 mm. Microscopically, it consisted of many vascular channels and perivascular cell proliferation encased by a fibrous capsule. The vascular proliferation showed a hemangiopericytoma (HPC)-like pattren such as staghorn-like vessels. Fibrosis was not present. The HPC-like cells had vesicular nuclei and polygonal cytoplasm. No atypia is recognized. The HPC-like cells focally showed vague nodular proliferation around the vessels. Immunohistocheically, the tumor cells were negative for cytokeratin, and positive for vimentin. The vasculatures were positive for factor VIII-related antigen, CD34, and CD31. The HPC-like tumor cells were positive for α-smooth muscle actin and h-caldesmon, but negative for desmin, S100 protein, melanosome, bcl-2, CD99, and KIT. The Ki-67 labeling was 8% and p53 was negative. The pathologic diagnosis was MPC of the neck skin. The patient is now alive without recurrence 4 years after the excision. A review of the literature revealed 73 cases of MPC from 6 papers. MPC is male predominance, and the patients ages ranges from 13 to 87 years with the median of 47 years. The most common location was lower extremities followed in order by upper extremities, head and neck, and trunk. One MPC occurred within the vasculature, and 3 cases of MPC developed in the scar or trauma lesions. The prognosis after excision is good, but a very minority showed local recurrence. A differential diagnosis was also made. C1 [Terada, Tadashi] Shizuoka City Shimizu Hospital, Department of Pathology, Miyakami 1231, Shimizu-Ku, 424-8636 Shizuoka, Japan. RP Terada, T (reprint author), Shizuoka City Shimizu Hospital, Department of Pathology, 424-8636 Shizuoka, Japan. EM piyo0111jp@yahoo.co.jp CR Middeleton LP, Duray PH, Merino MJ, 1998, The histological spectrum of hemangiopericytoma: application of immunohistochemical analysis including proliferative markers to facilitate diagnosis and predict prognosis. Hum Pathol 29:636–640 Croxatto JO, Font RL, 1982, Hemangiopericytoma of the orbid: a clinicopathologic study of 30 cases. Hum Pathol 13:210–218 Menzel T, Del Tos AP, Sapi Z, Kutzner H, 2006, Myopericytoma of the skin and soft tissue: clinicopathologic and immunohistochemical study of 54 cases. Am J Surg Pathol 30:104–113 Dray MS, McCarthy SW, Palmer AA, Bonar SF, Stalley PD, Marjoniemi V, Millar E, Scolyer RA, 2006, Myoipericytoma: a unifying term for a spectrum of tumours that show overlapping features with myofibroma. A review of 14 cases. J Clin Pathol 59:67–73 Folpes AL, Mentzel T, Lehr HA, Fisher C, Balzer BL, Weiss SW, 2005, Perivascular epithelioid cell neoplasms of soft tissue and gynecologic origin: a clinicopathologic study of 26 cases and review of the literature. Am J Surg Pathol 29:1558–1575 Liegl B, Hornick JL, Fletcher CD, 2008, Primary cutaneous PEComa: distinctive clear cell lesions of skin. Am J Surg Pathol 32:608–614 Miettinen M, Paal E, Lasota J, Sobin JH, 2002, Gastrointestinal glomus tumors: a clinicopathologic, immunohistochemical, and molecular genetic study of 32 cases. Am J Surg Pathol 26:301– 311 Folpe AL, Fanburg-Smith JC, Miettinen M, Weiss SW, 2001, Atypical and malignant glomus tumors: analysis of 52 cases, with a proposal for the reclassification of glomus tumors. Am J Surg Pathol 25:1–12 Matsuyama A, Hisaoka M, Hashimoto H, 2007, Angioleiomyoma: a clinicopatholgic and immunohistochemical reappraisal with special reference to the correlation with myopericytoma. Hum Pathol 38:645–651 Granter SR, Badizadegan K, Fletcher CD, 1998, Myofibromatosis in adults, glomangiopericytoma, and myopericytoma: a spectrum of tumors showing perivascular myoid differentiation. Am J Surg Pathol 22:513–525 Thompson LD, Miettinen M, WEnig BM, 2003, Sinonasal hemangiopericytoma: a clinicopathologic and immunophenotypic analysis of 104 cases showing perivascular myoid cell differentiation. Am J Surg Pathol 27:737–749 Lin IH, Kuo FY, Su CY, Lin HC, 2006, Sinonasal-type hemangiopericytoma of the sphenoid sinus. Otolaryngol Head Neck Surg 135:977–979 Rajaram V, Brat DJ, Perry A, 2004, Anaplastic meningioma versus meningeal hemangiopericytoma: immunohistochemical and genetic markers. Hum Pathol 35:1413–1418 Terada T, Kawaguchi M, Furukawa K, Sekido Y, Osamura Y, 2002, Minute mixed ductal-endocrine carcinoma of the pancreas with predominant intraductal growth. Pathol Int 52:740–746 Terada T, Kawaguchi M, 2005, Primary clear cell adenocarcinoma of the peritoneum. Tohoku J Exp Med 271:271–275 Alawi F, Stratton D, Freedman PD, 2001, Solitary fibrous tumor of the oral soft tissue: a clinicopathological and immunohistochemical study of 16 cases. Am J Surg Pathol 35:900–910 Miettinen M, Lasota J, 2006, Gastrointestinal stromal tumor: review on morphology, molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med 130:1466–1478 Miettinen M, Lasota J, 2005, KIT, CD117): a review on expression in normal and neoplastic tissue, and mutations and their clinicopathologic correlation. Appl Immunohistochem Mol Morphol 13:205–220 Terada T, 2009, Gastrointestinal stromal tumor of the uterus: a case report with genetic analyses of c-kit and PDGFRA genes. Int J Gynecol Oncol 28:29–34 Terada T, 2009, Primary extragastrointestinal stromal tumors of the transverse mesocolon without c-kit mutations but with PDGFRA mutations. Med Oncol 26:233–237 Terada T, 2008, Primary multiple extragastrointestinal stromal tumors of the omentum with different mutations of c-kit gene. World J Gastroenterol 14:7256–7259 Michal M, Fanburg-Smith JC, Lasota J, Fetsch JF, Lichky J, Miettinen M, 2006, Minute synovial sarcoma of the hands and feet: a clinicopathologic study of 21 tumors less than 1 cm. Am J Surg Pathol 30:721–726 McMenamin ME, Fletcher CD, 2002, Malignant myopericytoma: expanding the spectrum of tumours with myopericytic differentiation. Histopathology 41:450–460 Scott RS, Blank KL, Proffer LH, Kraus EW, Heim-Hall J, 2006, Perivascular myoma of myopericytoma and myofibromatosis-type arising in a chronic scar. J Cutan Pathol 33:231–235 Laga AC, Tajirian AL, Islam MN, Bhattachayya I, Cohen DM, Plamondon CJ, Robinson-Bostom L, 2008, Myopericytoma: report of two cases associated with trauma. J Cutan Pathol 35:866–870 McMenamin ME, Calonje E, 2002, Intravascular myopericytoma. J Cutan Pathol 29:557–561 Mimami Y, Shiomi T, Manaba T, 2002, Perivascular myoma: case report and immunohistochemical and ultrastructural studies. Pathol Int 52:69–74 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2010 VL 16 IS 4 BP 613 EP 616 DI 10.1007/s12253-010-9253-y PG 4 ER PT J AU Wang, Y Sun, W Guan, Ch Yu, H Pan, MZ AF Wang, Yan Sun, Weijia Guan, Chao Yu, He Pan, M Z TI Distribution of Basement Membrane in Supraglottic Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Supraglottic squamous cell carcinoma; Basement membrane; Type IV collagen; Cervical lymph node metastasis ID Supraglottic squamous cell carcinoma; Basement membrane; Type IV collagen; Cervical lymph node metastasis AB The object of this study was to assess the distribution of basement membrane in supraglottic squamous cell carcinomas. Expression of type IV collagen was detected by immunohistochemistry in resected supraglottic squamous cell carcinomas, and the correlation was examined between expression of type IV collagen and clinicopathological factors and cervical lymph node metastasis of supraglottic squamous cell carcinomas patients. An intact, continuous basement membrane was found in 17 cases (42.5%), while partial or widespread loss of the basement membrane was detected in the other 23 cases (57.5%). Heavily defective basement membrane was much more frequently observed in cases with poor histological differentiation (P<0.05). Cases with BM destruction were more likely to be accompanied by cervical lymph node metastasis (P<0.05). These data suggest that assessing the distribution pattern of basement membrane may be helpful in evaluating the malignancy grading of supraglottic squamous cell carcinomas and the potential occurrence of cervical lymph node metastasis. C1 [Wang, Yan] China Medical University, The 1st Affiliated Hospital, Department of Otolaryngology, 110001 Shenyang, China. [Sun, Weijia] China Medical University, The 4th Affiliated Hospital, Department of Surgery, 110001 Shenyang, China. [Guan, Chao] China Medical University, The 1st Affiliated Hospital, Laryngal Cancer Research Institution, 110001 Shenyang, China. [Yu, He] China Medical University, The 1st Affiliated Hospital, Department of Otolaryngology, 110001 Shenyang, China. [Pan, M Z] China Medical University, The 1st Affiliated Hospital, Department of Otolaryngology, 110001 Shenyang, China. RP Wang, Y (reprint author), China Medical University, The 1st Affiliated Hospital, Department of Otolaryngology, 110001 Shenyang, China. EM wangyan@mail.cmu.edu.cn CR Brown PD, Levy AT, Margulies IM, Liotta LA, Stetler-Stevenson WG, 1990, Independent distribution pattern and celluler processing of the 72 KD type IV collagenase and interstital collagenase in human tumorgenic cell lines. Cancer Res 50:6184–6191 Hasty KA, Pourmotabbed TF, Goldberg GI, Thompson JP, Spinella DG, Stevens RM, Mainardi CL, 1990, Human neutrophil collagenase. A distinct gene product with homology to other matrix metalloproteinases. J Biol chem 265:11421–11424 Collier IE, Wilhelm SM, Eisen AZ, Marmer BL, Grant GA, Seltzer JL, Kronberger A, He CS, Bauer EA, Goldberg GI, 1988, H-ras oncogene-transformed human bronchial epithelial cells, TBE-1, secrete a single metalloprotease capable of degrading basement membrane collagen. J Biol Chem 263:6579–6587 Lunt SJ, Chaudary N, Hill RP, 2009, The tumor microenvironment and metastatic disease. Clin Exp Metastasis 26:19–34 Hay ED, 1982, Cell biology of ertracellular matrix. Plenum, New York Liotta LA, 1984, Tumor invasion and metastases: role of the basement membrane. Am J Pathol 117:339–348 Leblond CP, Inoue S, 1989, Structure, composition and assembly of basement membrane. Am J Anat 185:367–390 Sakr WA, Zarbo RJ, Jacobs JR, Crissman JD, 1987, Distribution of basement membrane in squamous cell carcinoma of the head and neck. Hum pathol 18:1043–1050 Carter RL, Burman JF, Barr L, Gusterson BA, 1985, Immunohistochemical localization of basement membrane type IV Collagen in invasive and metastatic squamous carcinomas of the head and neck. J Pathol 147:159–164 Martinez-Hernandez A, Amenta PS, 1983, The basement membrane in pathology. Lab Invest 48:656–677 Barsky SH, Siegal GP, Jannotta F, Liotta LA, 1983, Loss of basement membrane components by invasive tumors but not by their begin counterparts. Lab Invest 49:140–147 Yamamoto E, Kohama G, Sunakawa H, Iwai M, Hiratsuka H, 1983, Mode of invasion, bleomycin sensitivity and clinical course in squamous cell carcinoma of the oral cavity. Cancer 51:2175–2180 Radisky DC, 2005, Epithelial-mesenchymal transition. J Cell Sci 118:4325–4326 Bienz M, 2005, beta-Catenin a pivot between cell adhesion and Wnt signalling. Curr Biol 15:64–67 Bonitsis N, Batistatou A, Karantima S, Charalabopoulos K, 2006, The role of cadherin/catenin complex in malignant melanoma. Exp Oncol 28:187–193 Andreasen PA, Egelund R, Petersen HH, 2000, The plasminogen activation system in tumor growth, invasion, and metastasis. Cell Mol Life Sci 57:25–40 Osinsky SP, Ganusevich II, Bubnovskaya LN, Valkovskaya NV, Kovelskaya AV, Sergienko TK, Zimina SV, 2005, Hypoxia level and matrix metalloproteinases-2 and–9 activity in Lewis lung carcinoma: correlation with metastasis. Exp Oncol 27:202–205 Rofstad EK, Rasmussen H, Galappathi K, Mathiesen B, Nilsen K, Graff BA, 2002, Hypoxia promotes lymph node metastasis in human melanoma xenografts by up-regulating the urokinase-type plasminogen activator receptor. Cancer Res 62:1847–1853 Visser R, van der Beek JM, Havenith MG, Cleutjens JP, Bosman FT, 1986, Immunocytochemical detection of basement membrane antigens in the histopathological evaluation of laryngeul dysplasia and neoplasia. Histopathology 10:171–180 Repassy G, Czigner J, Ribari O, Lapis K, 1988, The barrier-like role of activated connective tissue against the spread of supraglottic carcinoma. Arch Otorhinolaryngol 245:151–154 Wang Y, Tian J, Ren Z, 1997, The relationship between histologic grading of malignancy and regional lymph node metastasis in squamous cell carcinoma of larynx. J Clin Otorhinolaryngol, China, 11:160–162 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 1 EP 5 DI 10.1007/s12253-010-9250-1 PG 5 ER PT J AU Mojtahedi, Z Khademi, B Hashemi, BS Abtahi, MBS Ghasemi, AM Fattahi, JM Ghaderi, A AF Mojtahedi, Zahra Khademi, Bijan Hashemi, Basir Seyed Abtahi, Mohammad Bagher Seyed Ghasemi, Ali Mohammad Fattahi, Javad Mohammad Ghaderi, Abbas TI Serum Interleukine-6 Concentration, But Not Interleukine-18, is Associated with Head and Neck Squamous Cell Carcinoma Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cytokine; Head and neck neoplasms; Interleukin-18; Interleukin-6; SCC ID Cytokine; Head and neck neoplasms; Interleukin-18; Interleukin-6; SCC AB Inflammation has been linked to various steps in tumorigenesis. Interleukin (IL)-6 and IL-18 are two inflammatory cytokines whose serum concentrations are elevated in several types of cancer, including head and neck squamous cell carcinoma (HNSCC) in some studies. This study was designed to analyze the serum concentrations of these cytokines in Iranian HNSCC patients. Serum IL-6 and IL-18 concentrations were assayed by ELISA commercial kits in 65 untreated patients and 20 healthy volunteers. Serum IL-6 concentration was significantly increased in patients compared to healthy individuals (p<0.000). IL-6 concentration increased as the tumor stage progressed, and a significant difference appeared between stage IV vs. stage I/II/III (p=0.03) disease. Although serum IL-18 concentration was higher in patients than in healthy individuals, the difference was not statistically significant (p=0.06). Moreover, there was no association between serum IL-18 concentration and tumor stage (p=0.47). A significant difference was observed in serum IL-18 concentration according to the gender with higher IL-18 concentration in male patients (p=0.01). In conclusion, serum concentration of IL-6 might correlate with the stage of tumor progression in Iranian HNSCC patients. Further studies with larger numbers of patients are required to exclude the possible minor correlation of serum IL-18 concentration with tumor stage. C1 [Mojtahedi, Zahra] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Khademi, Bijan] Shiraz University of Medical Sciences, Khalili hospital, Department of ENTShiraz, Iran. [Hashemi, Basir Seyed] Shiraz University of Medical Sciences, Khalili hospital, Department of ENTShiraz, Iran. [Abtahi, Mohammad Bagher Seyed] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Ghasemi, Ali Mohammad] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Fattahi, Javad Mohammad] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Ghaderi, Abbas] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. RP Ghaderi, A (reprint author), Shiraz University of Medical Sciences, Institute for Cancer Research, Shiraz, Iran. EM ghaderia@sums.ac.ir CR Hardisson D, 2003, Molecular pathogenesis of head and neck squamous cell carcinoma. Eur Arch Otorhinolaryngol 260:502– 508 Aggarwal BB, Shishodia S, Sandur SK, 2006, Inflammation and cancer: how hot is the link? Biochem Pharmacol 72:1605–1621 Heikkila K, Ebrahim S, Lawlor DA, 2008, Systematic review of the association between circulating interleukin-6, IL-6, and cancer. Eur J Cancer 44:937–945 Nakanishi K, Yoshimoto T, Tsutsui H et al, 2001, Interleukin-18 is a unique cytokine that stimulates both Th1 and Th2 responses depending on its cytokine milieu. Cytokine Growth Factor Rev 12:53–72 Chen Z, Malhotra PS, Thomas GR et al, 1999, Expression of proinflammatory and proangiogenic cytokines in patients with head and neck cancer. Clin Cancer Res 5:1369–1379 Duffy SA, Taylor JM, Terrell JE et al, 2008, Interleukin-6 predicts recurrence and survival among head and neck cancer patients. Cancer 113:750–757 Riedel F, Zaiss I, Herzog D et al, 2005, Serum levels of interleukin-6 in patients with primary head and neck squamous cell carcinoma. Anticancer Res 25:2761–2765 Riedel F, Adam S, Feick P et al, 2004, Expression of IL-18 in patients with head and neck squamous cell carcinoma. Int J Mol Med 13:267–272 Jablonska E, Puzewska W, Grabowska Z et al, 2005, VEGF, IL- 18 and NO production by neutrophils and their serum levels in patients with oral cavity cancer. Cytokine 30:93–99 Jablonska E, Piotrowski L, Grabowska Z, 1997, Serum levels of IL-1b, IL-6, TNF-a, sTNF-RI and CRP in patients with oral cavity cancer. Pathol Oncol Res 3:126–129 Dienz O, Rincon M, 2009, The effects of IL-6 on CD4 T cell responses. Clin Immunol 130:27–33 Vidal-Vanaclocha F, Mendoza L, Telleria N et al, 2006, Clinical and experimental approaches to the pathophysiology of interleukin- 18 in cancer progression. Cancer Metastasis Rev 25:417–434 Samsami Dehaghani A, Shahriary K, Kashef MA et al, 2009, Interleukin-18 gene promoter and serum level in women with ovarian cancer. Mol Biol Rep 36:2393–2397 Bukan N, Sozen S, Coskun U et al, 2003, Serum interleukin-18 and nitric oxide activity in bladder carcinoma. Eur Cytokine Netw 14:163–167 Diakowska D, Markocka-Maczka K, Grabowski K et al, 2006, Serum interleukin-12 and interleukin-18 levels in patients with oesophageal squamous cell carcinoma. Exp Oncol 28:319–322 Jebreel A, Mistry D, Loke D et al, 2007, Investigation of interleukin 10, 12 and 18 levels in patients with head and neck cancer. J Laryngol Otol 121:246–252 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 7 EP 10 DI 10.1007/s12253-010-9261-y PG 4 ER PT J AU Valcz, G Krenacs, T Sipos, F Leiszter, K Toth, K Balogh, Zs Csizmadia, A Muzes, Gy Molnar, B Tulassay, Zs AF Valcz, Gabor Krenacs, Tibor Sipos, Ferenc Leiszter, Katalin Toth, Kinga Balogh, Zsofia Csizmadia, Annamaria Muzes, Gyorgyi Molnar, Bela Tulassay, Zsolt TI The Role of the Bone Marrow Derived Mesenchymal Stem Cells in Colonic Epithelial Regeneration SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mesenchymal stem cells; Mucosal regeneration; Mesenchymal-epithelial transition; Isolated lymphoid aggregates ID Mesenchymal stem cells; Mucosal regeneration; Mesenchymal-epithelial transition; Isolated lymphoid aggregates AB Bone marrow derived mesenchymal stem cells (BM-MSCs) take part in the colonic mucosal regeneration. They are multipotent cells, which can be identified with both negative (i.e. CD13, CD 14, CD45, c-Kit, major histocompatibility complex /MHC class I and II) and positive (i.e. CD54 (ICAM1), CD133, CD146 (MCAM), CD166, Flk-1, Sca-1, Thy-1, stage-specific antigen I /SSEAI and Musashi-1, HLA class I) markers. These cells can repopulate the gastrointestinal mucosa as they may differentiate into stromal- (i.e. myofi-broblast) or epithelial-like (Paneth-, epithel-, goblet or enteroendocrin) cells without proliferation. During the mesenchymal to epithelial transition (MET) stem cells enter the epithelial layer and take up epithelial cell-like properties. Rarely BM-MSCs may retain their stem cell characteristics and are capable of producing progeny. The isolated lymphoid aggregates may serve as a platform from where BM-MSCs migrate to the nearby crypts as mediated by several chemoattractant proteins, which are expressed in injured tissue. The number of BM-MSCs is influenced by the degree of inflammation. In this review we summarize the current information about the role of BM-MSCs in the repair progress of injured colonic epithelium and their potential clinical applications. C1 [Valcz, Gabor] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Leiszter, Katalin] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Toth, Kinga] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Balogh, Zsofia] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Csizmadia, Annamaria] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Muzes, Gyorgyi] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. RP Valcz, G (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM valczg@yahoo.com CR Grove JE, Bruscia E, Krause DS, 2004, Plasticity of bone marrow-derived stem cells. Stem Cells 22:487–500 Potten CS, Loeffler M, 1990, Stem cells: attributes, cycles, spirals, pitfalls and uncertainties. Lessons for and from the crypt. Development 110:1001–1020 Matsumoto T, Okamoto R, Yajima T et al, 2005, Increase of bone marrow-derived secretory lineage epithelial cells during regeneration in the human intestine. Gastroenterology 128:1851–1867 Brittan M, Wright NA, 2004, Stem cell in gastrointestinal structure and neoplastic development. Gut 53:899–910 Tanaka F, Tominaga K, Ochi M et al, 2008, Exogenous administration of mesenchymal stem cells ameliorates dextran sulfate sodium-induced colitis via anti-inflammatory action in damaged tissue in rats. Life Sci 83:771–779 Wei Y, Nie Y, Lai J et al, 2009, Comparison of the population capacity of hematopoietic and mesenchymal stem cells in experimental colitis rat model. Transplantation 88:42–48 De Wever O, Demetter P, Mareel M et al, 2008, Stromal myofibroblasts are drivers of invasive cancer growth. Int J Cancer 123:2229–2238 Brittan M, Hunt T, Jeffery R et al, 2002, Bone marrow derivation of pericryptal myofibroblasts in the mouse and human small intestine and colon. Gut 50:752–757 Barry FP, Murphy JM, 2004, Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol 36:568–584 Spees JL, Olson SD, Ylostalo J et al, 2003, Differentiation, cell fusion, and nuclear fusion during ex vivo repair of epithelium by human adult stem cells from bone marrow stroma. Proc Natl Acad Sci U S A 100:2397–2402 Minguell JJ, Erices A, Conget P, 2001, Mesenchymal stem cells. Exp Biol Med, Maywood, 226:507–520 Patel SA, Sherman L, Munoz J et al, 2008, Immunological properties of mesenchymal stem cells and clinical implications. Arch Immunol Ther Exp, Warsz, 56:1–8 Jiang Y, Jahagirdar BN, Reinhardt RL et al, 2002, Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418:41–49 Kassem M, Kristiansen M, Abdallah BM, 2004, Mesenchymal stem cells: cell biology and potential use in therapy. Basic Clin Pharmacol Toxicol 95:209–214 Matsumoto T, Okamoto R, Yajima T et al, 2005, Increase of bone marrow-derived secretory lineage epithelial cells during regeneration in the human intestine. Gastroenterology 128:1851–1867 Phinney DG, Prockop DJ, 2007, Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair current views. Stem Cells 25:2896– 2902 Pittenger MF, Mackay AM, Beck SC et al, 1999, Multilineage potential of adult human mesenchymal stem cells. Science 284:143–147 Baksh D, Yao R, Tuan RS, 2007, Comparison of proliferative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow. Stem Cells 25:1384–1392 Haasters F, Prall WC, Anz D et al, 2009, Morphological and immunocytochemical characteristics indicate the yield of early progenitors and represent a quality control for human mesenchymal stem cell culturing. J Anat 214:759–767 Mansilla E, Marin GH, Drago H et al, 2006, Bloodstream cells phenotypically identical to human mesenchymal bone marrow stem cells circulate in large amounts under the influence of acute large skin damage: new evidence for their use in regenerative medicine. Transplant Proc 38:967–969 Gargett CE, Schwab KE, Zillwood RM et al, 2009, Isolation and culture of epithelial progenitors and mesenchymal stem cells from human endometrium. Biol Reprod 80:1136–1145 Satija NK, Gurudutta GU, Sharma S et al, 2007, Mesenchymal stem cells: molecular targets for tissue engineering. Stem Cells Dev 16:7–23 Leedham SJ, Brittan M, McDonald SA et al, 2005, Intestinal stem cells. J Cell Mol Med 9:11–24 Chamberlain G, Fox J, Ashton B et al, 2007, Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells 25:2739–2749 Okamoto R, Yajima T, Yamazaki M et al, 2002, Damaged epithelia regenerated by bone marrow-derived cells in the human gastrointestinal tract. Nat Med 8:1011–1017 Li C, Kong Y, Wang H et al, 2009, Homing of bone marrow mesenchymal stem cells mediated by sphingosine 1-phosphate contributes to liver fibrosis. J Hepatol 50:1174–1183 Liu ZJ, Zhuge Y, Velazquez OC, 2009, Trafficking and differentiation of mesenchymal stem cells. J Cell Biochem 106:984–991 Liu N, Chen R, Du H et al, 2009, Expression of IL-10 and TNFalpha in rats with cerebral infarction after transplantation with mesenchymal stem cells. Cell Mol Immunol 6:207–213 Kim YS, Park HJ, Hong MH et al, 2009, TNF-alpha enhances engraftment of mesenchymal stem cells into infarcted myocardium. Front Biosci 14:2845–2856 Yabana T, Arimura Y, Tanaka H et al, 2009, Enhancing epithelial engraftment of rat mesenchymal stem cells restores epithelial barrier integrity. J Pathol 218:350–359 Komori M, Tsuji S, Tsujii M et al, 2005, Involvement of bone marrow-derived cells in healing of experimental colitis in rats. Wound Repair Regen 13:109–118 Emura M, Ochiai A, Horino M et al, 2000, Development of myofibroblasts from human bone marrow mesenchymal stem cells cocultured with human colon carcinoma cells and TGF beta 1. In Vitro Cell Dev Biol Anim 36:77–80 Brenmoehl J, Miller SN, Hofmann C et al, 2009, Transforming growth factor-beta 1 induces intestinal myofibroblast differentiation and modulates their migration. World J Gastroenterol 15:1431–1442 Beck PL, Rosenberg IM, Xavier RJ et al, 2003, Transforming growth factor-beta mediates intestinal healing and susceptibility to injury in vitro and in vivo through epithelial cells. Am J Pathol 162:597–608 Roufosse CA, Direkze NC, Otto WR et al, 2004, Circulating mesenchymal stem cells. Int J Biochem Cell Biol 36:585–597 Andoh A, Bamba S, Fujiyama Yet al, 2005, Colonic subepithelial myofibroblasts in mucosal inflammation and repair: contribution of bone marrow-derived stem cells to the gut regenerative response. J Gastroenterol 40:1089–1099 Brittan M, Chance V, Elia G et al, 2005, A regenerative role for bone marrow following experimental colitis: contribution to neovasculogenesis and myofibroblasts. Gastroenterology 128:1984–1995 Powell DW, Mifflin RC, Valentich JD et al, 1999, Myofibroblasts. I. Paracrine cells important in health and disease. Am J Physiol 277:1–9 Powell DW, Mifflin RC, Valentich JD et al, 1999, Myofibroblasts. II. Intestinal subepithelial myofibroblasts. Am J Physiol 277:183– 201 Baum B, Settleman J, Quinlan MP, 2008, Transitions between epithelial and mesenchymal states in development and disease. Semin Cell Dev Biol 19:294–308 Saxena SK, Thompson JS, Sharp JG, 1997, Role of organized intestinal lymphoid aggregates in intestinal regeneration. J Invest Surg 10:97–103 Chugh AR, Zuba-Surma EK, Dawn B, 2009, Bone marrowderived mesenchymal stems cells and cardiac repair. Minerva Cardioangiol 57:185–202 Sugaya K, Merchant S, 2008, How to approach Alzheimer’s disease therapy using stem cell technologies. J Alzheimers Dis 15:241–254 Le Visage C, Dunham B, Flint P et al, 2004, Coculture of mesenchymal stem cells and respiratory epithelial cells to engineer a human composite respiratory mucosa. Tissue Eng 10:1426–1435 Powell DW, Adegboyega PA, Di Mari JF et al, 2005, Epithelial cells and their neighbors I. Role of intestinal myofibroblasts in development, repair, and cancer. Am J Physiol Gastrointest Liver Physiol 289:2–7 Rubio D, Garcia S, De la Cueva T et al, 2008, Human mesenchymal stem cell transformation is associated with a mesenchymal-epithelial transition. Exp Cell Res 314:691–698 Houghton J, Wang TC, 2005, Helicobacter pylori and gastric cancer: a new paradigm for inflammation-associated epithelial cancers. Gastroenterology 128:1567–1578 Panes J, Salas A, 2009, Mechanisms underlying the beneficial effects of stem cell therapies for inflammatory bowel diseases. Gut 58:898–900 Khalil PN, Weiler V, Nelson PJ et al, 2007, Nonmyeloablative stem cell therapy enhances microcirculation and tissue regeneration in murine inflammatory bowel disease. Gastroenterology 132:944–954 Asari S, Itakura S, Ferreri K et al, 2009, Mesenchymal stem cells suppress B-cell terminal differentiation. Exp Hematol 37:604–615 Ringden O, Uzunel M, Rasmusson I et al, 2006, Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation 81:1390–1397 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 11 EP 16 DI 10.1007/s12253-010-9262-x PG 6 ER PT J AU Kovacs, Lakosi, F Liposits, G Toller, G Hadjiev, J Vandulek, Cs Walter, N Glavak, Cs Antal, G Horvath, Repa, I Bogner, P AF Kovacs, Arpad Lakosi, Ferenc Liposits, Gabor Toller, Gabor Hadjiev, Janaki Vandulek, Csaba Walter, Norbert Glavak, Csaba Antal, Gergely Horvath, Akos Repa, Imre Bogner, Peter TI 3-D Conformal Photon Boost in the Treatment of Early Stage Breast Cancer: Four Year Follow Up Results SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Radiotherapy; 3-D conformal photon boost ID Breast cancer; Radiotherapy; 3-D conformal photon boost AB In the treatment of early stage breast cancer, breast conserving surgery (BCS) followed by whole breast irradiation (WBI) is the standard treatment. The impact of the tumor bed boost following WBI is well-defined, but there are various delivery methods. In this study we demonstrate our 4 year experience with the 3-D conformal boost technique. Between January 2004 and June 2005, 77 early stage (Stage I–II) breast cancer patients were treated in our institute with whole breast irradiation (WBI, 50.4 Gy in 28 fractions) after breast conserving surgery. Following WBI, 3-D conformal photon boost was delivered (10–16 Gy in five to eight fractions) for all patients. The clinical outcome was retrospectively recorded in terms of survival and local control. The side effect profile (fibrosis, fat necrosis and cosmetic outcome) was also recorded and studied. In our patient group the mean follow up time was 46.8 months (median: 52, range: 17–71, SD: 14.4) The 4-year probability of local tumor control was 96% (crude rate: 74/77–96.1%), the 4-year probability of overall survival was 96% (crude rate: 74/77–96.1%) in this patient group. In case of the distant metastasis free survival the probability was 89, 5% (crude rate: 70/77–90, 1%). Probability of disease specific survival was 98% (crude rate: 76/77–98. 7%). Local relapse occurred in three cases (3.9%). In ten cases (12.9%) asymptomatic grade I–II breast fibrosis, in eight cases (10.4%) asymptomatic breast fat necrosis were registered. For 14 patients (18.2%) asymptomatic lung fibrosis was recorded on the control CT scans. In term of the relapse free survival, the close resection margin and the nodal positivity resulted in significant difference in favor of the clear resection margin group and the node negative group. In this study the 3-D conformal photon boost resulted in good local control and side effect profile. The presence of tumor bed clips resulted in significantly lower boost PTV volumes, but no correlation was found between the irradiated boost volume and the breast fibrosis. In the relapse free survival analysis, nodal negativity and clear margin status resulted in significantly better RFS. C1 [Kovacs, Arpad] Kaposi Mor Teaching Hospital, Guba S. u 40, 7400 Kaposvar, Hungary. [Lakosi, Ferenc] Kaposi Mor Teaching Hospital, Guba S. u 40, 7400 Kaposvar, Hungary. [Liposits, Gabor] Kaposi Mor Teaching Hospital, Guba S. u 40, 7400 Kaposvar, Hungary. [Toller, Gabor] Kaposi Mor Teaching Hospital, Guba S. u 40, 7400 Kaposvar, Hungary. [Hadjiev, Janaki] Kaposi Mor Teaching Hospital, Guba S. u 40, 7400 Kaposvar, Hungary. [Vandulek, Csaba] Kaposi Mor Teaching Hospital, Guba S. u 40, 7400 Kaposvar, Hungary. [Walter, Norbert] University of Pecs, Faculty of Health Sciences, Vorosmarty u. 4., H-7621 Pecs, Hungary. [Glavak, Csaba] Kaposi Mor Teaching Hospital, Guba S. u 40, 7400 Kaposvar, Hungary. [Antal, Gergely] Kaposi Mor Teaching Hospital, Guba S. u 40, 7400 Kaposvar, Hungary. [Horvath, Akos] University of DebrecenDebrecen, Hungary. [Repa, Imre] Kaposi Mor Teaching Hospital, Guba S. u 40, 7400 Kaposvar, Hungary. [Bogner, Peter] Kaposi Mor Teaching Hospital, Guba S. u 40, 7400 Kaposvar, Hungary. RP Kovacs, (reprint author), Kaposi Mor Teaching Hospital, 7400 Kaposvar, Hungary. EM kovacs.arpad@sic.hu CR Bartelink H, Horiot JC, Poortmans P et al, 2001, Recurrence rates after treatment of breast cancer with standard radiotherapy with or without additional radiation. N Engl J Med 345:1378–1387 Bates A, Swift C, Kwa W, Moravan V, Aquino-Parsons C, 2007, A computed tomography-based protocol vs conventional clinical mark-up for breast electron boost. Clin Oncol 19:349–355 Bedwinek J, 1993, Breast conserving surgery and irradiation: the importance of demarcating the excision cavity with surgical clips. Int J Radiat Oncol Biol Phys 26:675–679 Denham JW, Sillar RW, Clarke D, 1991, Boost dosage to the excision site following conservative surgery for breast cancer: it’s easy to miss! Clin Oncol 3:257–261 Denham JW, Carter ML, 1988, Conservative treatment of breast cancer-where should the booster dose go [letter]? Int J Radiat Oncol Biol Phys 14:399–400 Early Breast Cancer Triallists’ Collaborative Group, 1995, Effects of radiotherapy and surgery in early breast cancer: an overview of the randomized trials. N Engl J Med 333:1444e1445 [erratum, N Engl J Med 1996;334:1003] Fisher B, Anderson S, Bryant J et al, 2002, Twenty-year followup of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 347:1233–1241 Goldberg H, Proznitz RG, Olson JA et al, 2005, Definition of post-lumpectomy tumor bed for radiotherapy boost field planning: CT versus surgical clips. Int J Radiat Oncol Phys 63:209–213 Graham P, Fourquetz A, 2005, Placing the boost in breastconservation radiotherapy: a review of the role, indications and techniques for breast-boost radiotherapy. Clin Oncol 18:210–219 Harrington KJ, Harrison M, Bayle P et al, 1996, Surgical clips in planning the electron boost in breast cancer: a qualitative and quantitative evaluation. Int J Radiat Oncol Biol Phys 34:579–584 Hunter MA, McFall TA, Hehr KA, 1996, Breast conserving surgery for primary breast cancer: necessity for surgical clips to define the tumor bed for radiation planning. Radiology 200:281– 282 International Commission on Radiation Units and Measurement, ICRU,, 1993, ICRU Report No. 50. Prescribing, recording and reporting photon beam therapy. Washington, DC: ICRU Kovacs A, Hadjiev J, Lakosi F et al, 2008, Comparison of photon with electron boost in treatment of early stage breast cancer. Pathol Oncol Res 14:193–197 Kovner F, Agay R, Merimsky O, Stadler J, Klausner J, Inbar M, 1999, Clips and scar as the guidelines for breast radiation boost after lumpectomy. Eur J Surg Oncol 25:483–486 Machtay M, Lanciano R, Hoffman J, Hanks GE, 1994, Inaccuracies in using the lumpectomy scar for planning electron boosts in primary breast carcinoma. Int J Radiat Oncol Biol Phys 30:43–48 Cs Polgar, Fodor J, Major T, Zs Orosz, Gy Nemeth, 2001, The role of boost irradiation in the conservative treatment of stage I-II breast cancer. Pathol Oncol Res 7(4):241–250 Poortmans P, Bartelink H, Horiot J-C, Struikmans H, Van den Bogaert W, Fourquet A, Jagerg J, Hoogenraadh W, Rodrigusa P, Rodenhuisi CW, Collette L, Pierartj M, On behalf of the EORTC Radiotherapy and Breast Cancer Groups, 2004, The influence of the boost technique on local control in breast-conserving treatment in the EORTC ‘boost versus no boost’ randomized trial. Radiother Oncol 72:25–33 Regine WF, Ayyangar KM, Komarnicky LT, Bhandare N, Mansfield CM, 1991, Computer-CT planning of the electron boost in definitive breast irradiation. Int J Radiat Oncol Biol Phys 20:121–125 Ringash J, Whelan T, Elliott E et al, 2004, Accuracy of ultrasound in localization of breast boost field. Radiother Oncol 72:61–66 Romestaing P, Lehingue Y, Carrie C et al, 1997, Role of a 10 Gy boost in the conservative treatment of early breast cancer: results of a randomized clinical trial in Lyon, France. J Clin Oncol 15:963–968 Uwini SA, Antonini N, Poortmans MP, Boersma L, Hurkmans C, Leer JW, Horiot J, Struikmans H, Bartelink H, 2009, The influence of the use of CT-planning on the irradiated boost volume in breast conserving treatment. Radiother Oncol 93:87–93 Smitt MC, Birdwell RL, Goffinet DR, 2001, Breast electron boost planning: comparison of CT and US. Radiology 219:203–206 Van Limbergen E, 2001, What are the optimal boost methods in relation to boost target depth in the breast? Proceedings of the Consensus Meeting on Breast Cancer: To boost or not to boost and how to do it. GEC-ESTRO, pp 105–114 Veronesi U, Cascinelli N, Mariani L et al, 2002, Twenty-year follow-up of a randomized study comparing breast-conserving surgery with radical mastectomy for early breast cancer. N Engl J Med 347:1227–1232 Vrieling C, Collette L, Fourquet A et al, 1999, The influence of the boost in breast-conserving therapy on cosmetic outcome in the EORTC “boost versus no boost” trial. Int J Radiat Oncol Biol Phys 45:677–685 Vrieling C, Collette L, Fourquet A et al, 2000, The influence of patient, tumor, and treatment factors on the cosmetic results after breastconserving therapy in the EORTC “Boost versus No Boost” Trial. Radiother Oncol 55:219–232 Weed DW, Yan D, Martinez AA, Vicini FA, Wilkinson TJ, Wong J, 2004, The validity of surgical clips as a radiographic surrogate for the lumpectomy cavity in image-guided accelerated partial breast irradiation. Int J Radiat Oncol Biol Phys 60:484–492 Lovey K, Fodor J, Major T et al, 2007, Fat necrosis after partialbreast irradiation with brachytherapy or electron irradiation versus standard whole-breast radiotherapy- 4 year results of a randomized trial. Int J Radiat Oncol Biol Phys 69(3):724–731 Cox JD, Stetz J, Pajak TF, 1995, Toxicity criteria of the Radiation Therapy Oncology Group, RTOG, and the European Organization for Research and Treatment of Cancer, EORTC). Int J Radiat Oncol Biol Phys 31:1341–1346 Harris J, Levine M, Svensson G et al, 1979, Analysis of cosmetic results following primary radiation therapy for stage I and II carcinoma of the breast. Int J Radiat Oncol Biol Phys 5:257–261 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 17 EP 23 DI 10.1007/s12253-010-9264-8 PG 7 ER PT J AU Saucy, F Bachmann, D Peterman, O Sordat, B Sordat, I Dorta, G AF Saucy, Francois Bachmann, Daniel Peterman, Olivier Sordat, Bernard Sordat, Isabelle Dorta, Gian TI The Plasminogen System in Microdissected Colonic Mucosa Distant from an Isolated Adenoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Urokinase-type plasminogen activators; Plasminogen activator inhibitors; Laser microdissection; Tubular adenoma; Colonic stroma; Colonic epithelium ID Urokinase-type plasminogen activators; Plasminogen activator inhibitors; Laser microdissection; Tubular adenoma; Colonic stroma; Colonic epithelium AB In the colon, the urokinase-type plasminogen activator (uPA), its receptor (uPAR), and plasminogen activator inhibitors, PAI-1 and PAI-2, are implicated in the transition from mucosa to adenoma and tumour progression. However, expression in the mucosa adjacent, or distant, to an adenoma has not yet been investigated. Three biopsies from mucosae adjacent (20 cm, ipsilateral) and distant (contralateral) to an isolated tubular adenoma were analysed in 14 patients and 8 controls. Laser microdissection isolated stromal and epithelial crypt components, and quantitative RT-PCR analyses of uPA, uPAR, PAI-1 and PAI-2 mRNA levels were performed. Among controls, no significant differences in the markers were noted. With left colon isolated tubular adenoma, uPA, uPAR, and PAI-2 mRNA levels were significantly increased in the adjacent mucosal stroma compared to epithelial crypt levels (p<0.05). In right colon adenoma, the mRNA levels of these 3 molecular markers were significantly increased only in the adjacent mucosal stromal samples (p<0.05). Isolated tubular adenoma in the colon increases significantly the mRNA levels of 3 proteolysis-associated molecular markers in the stromal, but not in the epithelial, components of adjacent mucosa. These results suggest the presence of regional and dynamic interactions in apparently non-involved mucosae. C1 [Saucy, Francois] University Hospital Lausanne, Division of Gastroenterology and Hepatology, 1011 Lausanne, Switzerland. [Bachmann, Daniel] University Hospital Lausanne, Division of Gastroenterology and Hepatology, 1011 Lausanne, Switzerland. [Peterman, Olivier] Swiss Institute for Experimental Cancer Research, 1066 Epalinges, Switzerland. [Sordat, Bernard] Swiss Institute for Experimental Cancer Research, 1066 Epalinges, Switzerland. [Sordat, Isabelle] Swiss Institute for Experimental Cancer Research, 1066 Epalinges, Switzerland. [Dorta, Gian] University Hospital Lausanne, Division of Gastroenterology and Hepatology, 1011 Lausanne, Switzerland. RP Saucy, F (reprint author), University Hospital Lausanne, Division of Gastroenterology and Hepatology, 1011 Lausanne, Switzerland. EM francois.saucy@chuv.ch CR Parkin DM, Bray F, Ferlay J, Pisani P, 2001, Estimating the world cancer burden: Globocan 2000. Int J Cancer 94(2):153–156 O'Brien MJ, Winawer SJ, Zauber AG, Gottlieb LS, Sternberg SS, Diaz B et al, 1990, The national polyp study. Patient and polyp characteristics associated with high-grade dysplasia in colorectal adenomas. Gastroenterology 98(2):371–379 Vogelstein B, Kinzler KW, 2004, Cancer genes and the pathways they control. Nat Med 10(8):789–799 Gregorieff A, Clevers H, 2005, Wnt signaling in the intestinal epithelium: from endoderm to cancer. Genes Dev 19(8):877–890 Galon J, Fridman WH, Pages F, 2007, The adaptive immunologic microenvironment in colorectal cancer: a novel perspective. Cancer Res 67(5):1883–1886 van den Brink GR, Offerhaus GJ, 2007, The morphogenetic code and colon cancer development. Cancer Cell 11(2):109–117 Dano K, Behrendt N, Hoyer-Hansen G, Johnsen M, Lund LR, Ploug M et al, 2005, Plasminogen activation and cancer. Thromb Haemost 93(4):676–681 Mazzieri R, Blasi F, 2005, The urokinase receptor and the regulation of cell proliferation. Thromb Haemost 93(4):641–646 Yang L, Avila H, Wang H, Trevino J, Gallick GE, Kitadai Y et al, 2006, Plasticity in urokinase-type plasminogen activator receptor, uPAR, display in colon cancer yields metastable subpopulations oscillating in cell surface uPAR density–implications in tumor progression. Cancer Res 66(16):7957–7967 Czekay RP, Aertgeerts K, Curriden SA, Loskutoff DJ, 2003, Plasminogen activator inhibitor-1 detaches cells from extracellular matrices by inactivating integrins. J Cell Biol 160(5):781–791 Duffy MJ, 2005, Predictive markers in breast and other cancers: a review. Clin Chem 51(3):494–503 Duffy MJ, McGowan PM, Gallagher WM, 2008, Cancer invasion and metastasis: changing views. J Pathol 214(3):283–293 Sordat I, Chaubert P, Protiva P, Guillou L, Mazzucchelli L, Saraga E et al, 1997, In situ stromal expression of the urokinase/plasmin system correlates with epithelial dysplasia in colorectal adenomas. Am J Pathol 150(1):283–295 Protiva P, Sordat I, Chaubert P, Saraga E, Tran-Thang C, Sordat B et al, 1998, Alterations in plasminogen activation correlate with epithelial cell dysplasia grading in colorectal adenomas. Br J Cancer 77(2):297–304 Dorta G, Chaubert P, Benhattar J, Saraga E, Protiva P, Blum AL, Sordat B, 1998, PAI-1 is an indicator for adenoma reccurence after polypectomy. Gastroenterology 114:A585 Mueller MM, Fusenig NE, 2004, Friends or foes—bipolar effects of the tumour stroma in cancer. Nat Rev Cancer 4(11):839–849 Jass JR, Sobin LH, Watanabe H, 1990, The World Health Organization's histologic classification of gastrointestinal tumors. A commentary on the second edition. Cancer 66(10):2162–2167 Winawer SJ, Zauber AG, O'Brien MJ, Gottlieb LS, Sternberg SS, Stewart ET et al, 1992, The national polyp study. Design, methods, and characteristics of patients with newly diagnosed polyps. The national polyp study workgroup. Cancer 70(5 Suppl):1236–1245 Lechner S, Muller-Ladner U, Renke B, Scholmerich J, Ruschoff J, Kullmann F, 2003, Gene expression pattern of laser microdissected colonic crypts of adenomas with low grade dysplasia. Gut 52(8):1148–1153 Danjoux M, Guimbaud R, Al Saati T, Meggetto F, Carrere N, Portier G et al, 2006, Contribution of microdissection for the detection of microsatellite instability in colorectal cancer. Hum Pathol 37(3):361–368 Sabates-Bellver J, Van der Flier LG, de Palo M, Cattaneo E, Maake C, Rehrauer H et al, 2007, Transcriptome profile of human colorectal adenomas. Mol Cancer Res 5(12):1263– 1275 Sugiyama Y, Farrow B, Murillo C, Li J, Watanabe H, Sugiyama K et al, 2005, Analysis of differential gene expression patterns in colon cancer and cancer stroma using microdissected tissues. Gastroenterology 128(2):480–486 Tlsty TD, Coussens LM, 2006, Tumor stroma and regulation of cancer development. Annu Rev Pathol 1:119–150 Del Rosso M, Fibbi G, Pucci M, Margheri F, Serrati S, 2008, The plasminogen activation system in inflammation. Front Biosci 13:4667–4686 Sier CF, Verspaget HW, Griffioen G, Verheijen JH, Quax PH, Dooijewaard G et al, 1991, Imbalance of plasminogen activators and their inhibitors in human colorectal neoplasia. Implications of urokinase in colorectal carcinogenesis. Gastroenterology 101, 6):1522–1528 Huang EH, Park JC, Appelman H, Weinberg AD, Banerjee M, Logsdon CD et al, 2006, Induction of inflammatory bowel disease accelerates adenoma formation in Min +/-mice. Surgery 139, 6):782–788 Ploplis VA, Tipton H, Menchen H, Castellino FJ, 2007, A urokinase-type plasminogen activator deficiency diminishes the frequency of intestinal adenomas in ApcMin/+ mice. J Pathol 213, 3):266–274 Balkwill F, Coussens LM, 2004, Cancer: an inflammatory link. Nature 431(7007):405–406 Sidenius N, Blasi F, 2003, The urokinase plasminogen activator system in cancer: recent advances and implication for prognosis and therapy. Cancer Metastasis Rev 22(2-3):205–222 Ishiguro K, Yoshida T, Yagishita H, Numata Y, Okayasu T, 2006, Epithelial and stromal genetic instability contributes to genesis of colorectal adenomas. Gut 55(5):695–702 Croucher DR, Saunders DN, Lobov S, Ranson M, 2008, Revisiting the biological roles of PAI2, SERPINB2, in cancer. Nat Rev Cancer 8(7):535–545 Issa JP, 2008, Colon cancer: it's CIN or CIMP. Clin Cancer Res 14(19):5939–5940 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 25 EP 31 DI 10.1007/s12253-010-9268-4 PG 7 ER PT J AU Lu, S Tian, J Lv, Z Wang, H Bai, X Liu, W Li, J Xu, W AF Lu, Sumei Tian, Jiajun Lv, Zhenghua Wang, Haibo Bai, Xiaohui Liu, Wenwen Li, Jianfeng Xu, Wei TI The Probable Role of Tumor Stem Cells for Lymph Node Metastasis in Supraglottic Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD133; CD44; Lymph node metastasis; Supraglottic carcinoma; Tumor stem cell ID CD133; CD44; Lymph node metastasis; Supraglottic carcinoma; Tumor stem cell AB Tumor stem cells (TSC), which are considered as likely candidates for the origin of cancer, are deduced to be responsible for tumor metastasis theoretically. We therefore investigated whether TSC were associated with lymph node metastasis in supraglottic carcinoma. Immunohistochemistry was performed for CD44, CD133, and LYVE-1 to detect TSC and lymphatic vessel density (LVD) in 66 primary supraglottic carcinoma tissue samples from 30 patients with lymph node metastasis (N+) and 36 patients without (N0). Reverse transcriptase polymerase chain reaction (RT-PCR) and Western blot were used to detect the expression of CD44 and CD133 at mRNA and protein levels in N+ and N0 primary tumors. The LVD was 22.4±10.26 in 30N+ and 6.8±4.09 in 36N0 samples subjected to immunohistochemistry, which was associated with their clinical nodal stages. There were 43.33% CD44-positive and 93.33% CD133-positive samples in 30N+, and 13.89% CD44-positive and 44.44% CD133-positive samples in 36N0 (P<0.05). However, in each positive slide, there were only 5∼10% CD44-positive cells, but 70∼85% CD133-possitive cells. The expressions of CD44 and CD133 of N+ obtained through RT-PCR and Western blot were significantly higher than those of N0. These results suggest that TSC identified through CD44-positive cells in N+ were significantly higher than those in N0, indicating that TSC may be responsible for lymph node metastasis. CD133, whose expression is not restricted to TSC, may be unspecific for TSC identification in hypostatic supraglottic carcinoma. C1 [Lu, Sumei] Provincial Hospital affiliated to Shandong University, Institute of Eye and ENT, 250021 Jinan, China. [Tian, Jiajun] Provincial Hospital affiliated to Shandong University, Department of Otolaryngology-Head and Neck Surgery, 250021 Jinan, China. [Lv, Zhenghua] Provincial Hospital affiliated to Shandong University, Department of Otolaryngology-Head and Neck Surgery, 250021 Jinan, China. [Wang, Haibo] Provincial Hospital affiliated to Shandong University, Institute of Eye and ENT, 250021 Jinan, China. [Bai, Xiaohui] Provincial Hospital affiliated to Shandong University, Institute of Eye and ENT, 250021 Jinan, China. [Liu, Wenwen] Provincial Hospital affiliated to Shandong University, Institute of Eye and ENT, 250021 Jinan, China. [Li, Jianfeng] Provincial Hospital affiliated to Shandong University, Institute of Eye and ENT, 250021 Jinan, China. [Xu, Wei] Provincial Hospital affiliated to Shandong University, Department of Otolaryngology-Head and Neck Surgery, 250021 Jinan, China. RP Xu, W (reprint author), Provincial Hospital affiliated to Shandong University, Department of Otolaryngology-Head and Neck Surgery, 250021 Jinan, China. EM xwhns@yahoo.com.cn CR Kyzas PA, Geleff S, Batistatou A et al, 2005, Evidence for lymphangiogenesis and its prognostic implications in head and neck squamous cell carcinoma. J Pathol 206(2):170–7 Zhao D, Pan J, Li XQ et al, 2008, Intratumoral lymphangiogenesis in oral squamous cell carcinoma and its clinicopathological significance. J Oral Pathol Med 37(10):616–25 Dalerba P, Cho RW, Clarke MF, 2007, Cancer stem cells: models and concepts. Annu Rev Med 58:267–84 Clarke MF, Fuller M, 2006, Stem cells and cancer: two faces of eve. Cell 124:1111–5 Wicha MS, Liu S, Dontu G, 2006, Cancer stem cells: an old idea: a paradigm shift. Cancer Res 66:1883–1890 Pardal R, Clarke MF, Morrison SJ, 2003, Applying the principles of stem-cell biology to cancer. Nat Rev Cancer 3(12):895–902 Prince ME, Ailles LE, 2008, Cancer stem cells in head and neck squamous cell cancer. J Clin Oncol 26(17):2871–5 Ailles L, Prince M, 2009, Cancer stem cells in head and neck squamous cell carcinoma. Methods Mol Biol 568:175–93 Wei XD, Zhou L, Cheng L et al, 2009, In vivo investigation of CD133 as a putative marker of cancer stem cells in Hep-2 cell line. Head and neck 31:94–101 Audet N, Beasley NJ, MacMillan C et al, 2005, Lymphatic vessel density, nodal metastases, and prognosis in patients with head and neck cancer. Arch Otolaryngol Head Neck Surg 131:1065–70 Koskinen WJ, Bono P, Leivo I et al, 2005, Lymphatic vessel density in vocal cord carcinomas assessed with LYVE-1 receptor expression. Radiother Oncol 77:172–5 Gao P, Zhou GY, Yin G et al, 2006, Lymphatic vessel density as a prognostic indicator for patients with stage I cervical carcinoma. Hum Pathol 37:719–25 Mori D, Yamasaki F, Shibaki M et al, 2007, Lateral peritumoral lymphatic vessel invasion can predict lymph node metastasis in esophageal squamous cell carcinoma. Mod Pathol 20:694–700 Maula SM, Luukkaa M, Grenman R et al, 2003, Intratumoral lymphatics are essential for the metastatic spread and prognosis in squamous cell carcinomas of the head and neck region. Cancer Res 63:1920–6 Gao P, Zhou GY, Zhang QH et al, 2008, Clinicopathological significance of peritumoral lymphatic vessel density in gastric carcinoma. Cancer Lett 263:223–230 Wang J, Guo LP, Chen LZ et al, 2007, Identification of cancer stem cell-like side population cells in human nasopharyngeal carcinoma cell line. Cancer Res 67:3716–24 Bianchini C, Ciorba A, Pelucchi S et al, 2008, Head and neck cancer: the possible role of stem cells. Eur Arch Otorhinolaryngol 265:17–20 Harper LJ, Piper K, Common J et al, 2007, Stem cell patterns in cell lines derived from head and neck squamous cell carcinoma. J Oral Pathol Med 36:594–603 Pries R, Witrkopf N, Trenkle T et al, 2008, Potential stem cell marker CD44 is constitutively expressed in permanent cell lines of head and neck cancer. In Vivo 22:89–92 Wright MH, Calcagno AM, Salcido CD et al, 2008, Brca1 breast tumors contain distinct CD44+/CD24−and CD133+ cells with cancer stem cell characteristics. Breast Cancer Res 10:R10 Du L, Wang H, He L, 2008, CD44 is of functional importance for colorectal cancer stem cells. Clin Cancer Res 14(21):6751–60 Zito G, Richiusa P, Bommarito A et al, 2008, In vitro identification and characterization of CD133, pos, cancer stemlike cells in anaplastic thyroid carcinoma cell lines. PLoS One 3: e3544 Choi D, Lee HW, Hur KY et al, 2009, Cancer stem cell markers CD133 and CD24 correlate with invasiveness and differentiation in colorectal adenocarcinoma. World J Gastroenterol 15, 18):2258–64 Griguer CE, Oliva CR, Gobin E et al, 2008, CD133 is a marker of bioenergetic stress in human glioma. PLoS One 3(11):e3655 Richardson GD, Robson CN, Lang SH et al, 2004, CD133, a novel marker for human prostatic epithelial stem cells. J Cell Sci 117:3539–45 Olempska M, Eisenach PA, Ammerpohl O et al, 2007, Detection of tumor stem cell markers in pancreatic carcinoma cell lines. Hepatobiliary Pancreat Dis Int 6:92–7 Immervoll H, Hoem D, Sakariassen PO et al, 2008, Expression of the “stem cell marker” CD133 in pancreas and pancreatic ductal adenocarcinomas. BMC Cancer 8:48 Shmelkov SV, Butler JM, Hooper AT et al, 2008, CD133 expression is not restricted to stem cells, and both CD133+ and CD133−metastatic colon cancer cells initiate tumors. J Clin Invest 118:2111–2120 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 33 EP 38 DI 10.1007/s12253-010-9271-9 PG 6 ER PT J AU Hadarits, F Kisfali, P Mohas, M Maasz, A Sumegi, K Szabo, M Hetyesy, K Valasek, A Janicsek, I Wittmann, I Melegh, B AF Hadarits, Ferenc Kisfali, Peter Mohas, Marton Maasz, Anita Sumegi, Katalin Szabo, Melinda Hetyesy, Katalin Valasek, Andrea Janicsek, Ingrid Wittmann, Istvan Melegh, Bela TI Stepwise Positive Association Between APOA5 Minor Allele Frequencies and Increasing Plasma Triglyceride Quartiles in Random Patients with Hypertriglyceridemia of Unclarified Origin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ApoA5; Hypertriglyceridemia; T-1131C; IVS3+G476A; T1259C; C56G; PCR-RFLP ID ApoA5; Hypertriglyceridemia; T-1131C; IVS3+G476A; T1259C; C56G; PCR-RFLP AB Apolipoprotein A5 (ApoA5) gene and its protein product play a central role in the complex regulation of circulating triglyceride levels in humans. Naturally occurring variants of the apolipoprotein A5 gene have been associated with increased triglyceride levels and have been found to confer risk for cardiovascular diseases. In our study, four polymorphisms, the T-1131C, IVS3+G476A, T1259C, and C56G alleles of APOA5 were analyzed in a total of 436 patients by polymerase chain reaction—restriction fragment length polymorphism methods. The randomly selected patients were classified into four quartile (q) groups based on triglyceride levels (q1: TG<1.31 mmol/l; q2: 1.31–2.90 mmol/l; q3: 2.91–4.85 mmol/l; q4: TG>4.85 mmol/l). We observed significant stepwise increasing association between the four APOA5 minor allele carrier frequencies and plasma triglyceride quartiles: -1131C (q1: 4.44%; q2: 8.95%; q3: 12.9%; q4: 20.6%), IVS3+476A (q1: 4.44%; q2: 5.79%; q3: 11.1%; q4: 19.7%), 1259C (q1: 4.44%; q2: 6.84%; q3: 11.1%; q4: 20.6%) and 56G (q1: 5.64%; q2: 6.31%; q3: 11.16%; q4: 11.9%). The serum total cholesterol and high density lipoprotein-cholesterol levels also showed allele-dependent differences in the quartiles. The findings presented here revealed a special arrangement of APOA5 minor alleles in patients with different serum triglyceride ranges in Hungarians. C1 [Hadarits, Ferenc] Markusovszky County Hospital, Central LaboratorySzombathely, Hungary. [Kisfali, Peter] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Mohas, Marton] University of Pecs, 2nd Department of Medicine and Nephrological CenterPecs, Hungary. [Maasz, Anita] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Sumegi, Katalin] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Szabo, Melinda] Robert Koch Hospital, Department of PulmonologyEdeleny, Hungary. [Hetyesy, Katalin] Aladar Petz County Hospital, Central LaboratoryGyor, Hungary. [Valasek, Andrea] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Janicsek, Ingrid] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Wittmann, Istvan] University of Pecs, 2nd Department of Medicine and Nephrological CenterPecs, Hungary. [Melegh, Bela] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. RP Melegh, B (reprint author), University of Pecs, Department of Medical Genetics and Child Development, Pecs, Hungary. EM bela.melegh@aok.pte.hu CR Charlton M, 2009, Obesity, hyperlipidemia, and metabolic syndrome. Liver Transpl 15:S83–S89 Karadag MK, Akbulut M, 2009, Low HDL levels as the most common metabolic syndrome risk factor in heart failure. Int Heart J 50:571–580 Ewald N, Hardt PD, Kloer HU, 2009, Severe hypertriglyceridemia and pancreatitis: presentation and management. Curr Opin Lipidol Hopkins PN, Heiss G, Ellison RC et al, 2003, Coronary artery disease risk in familial combined hyperlipidemia and familial hypertriglyceridemia: a case-control comparison from the National Heart, Lung, and Blood Institute Family Heart Study. Circulation 108:519–523 St-Pierre J, Lemieux I, Vohl MC et al, 2002, Contribution of abdominal obesity and hypertriglyceridemia to impaired fasting glucose and coronary artery disease. Am J Cardiol 90:15–18 Pennacchio LA, Olivier M, Hubacek JA et al, 2001, An apolipoprotein influencing triglycerides in humans and mice revealed by comparative sequencing. Science 294:169–173 van der Vliet HN, Sammels MG, Leegwater AC et al, 2001, Apolipoprotein A-V: a novel apolipoprotein associated with an early phase of liver regeneration. J Biol Chem 276:44512–44520 Baum L, Tomlinson B, Thomas GN, 2003, APOA5-1131T>C polymorphism is associated with triglyceride levels in Chinese men. Clin Genet 63:377–379 Hubacek JA, Adamkova V, Ceska R et al, 2004, New variants in the apolipoprotein AV gene in individuals with extreme triglyceride levels. Physiol Res 53:225–228 Hubacek JA, Skodova Z, Adamkova V et al, 2004, The influence of APOAV polymorphisms, T-1131>C and S19>W, on plasma triglyceride levels and risk of myocardial infarction. Clin Genet 65:126–130 Pennacchio LA, Olivier M, Hubacek JA et al, 2002, Two independent apolipoprotein A5 haplotypes influence human plasma triglyceride levels. Hum Mol Genet 11:3031–3038 Havasi V, Szolnoki Z, Talian G et al, 2006, Apolipoprotein A5 gene promoter region T-1131C polymorphism associates with elevated circulating triglyceride levels and confers susceptibility for development of ischemic stroke. J Mol Neurosci 29:177–183 Maasz A, Kisfali P, Jaromi L et al, 2008, Apolipoprotein A5 gene IVS3+G476A allelic variant confers susceptibility for development of ischemic stroke. Circ J 72:1065–1070 Maasz A, Kisfali P, Szolnoki Z et al, 2008, Apolipoprotein A5 gene C56G variant confers risk for the development of largevessel associated ischemic stroke. J Neurol 255:649–654 Ruiz-Narvaez EA, Yang Y, Nakanishi Y et al, 2005, APOC3/A5 haplotypes, lipid levels, and risk of myocardial infarction in the Central Valley of Costa Rica. J Lipid Res 46:2605–2613 Szalai C, Keszei M, Duba J et al, 2004, Polymorphism in the promoter region of the apolipoprotein A5 gene is associated with an increased susceptibility for coronary artery disease. Atherosclerosis 173:109–114 Tang Y, Sun P, Guo D et al, 2006, A genetic variant c.553G>T in the apolipoprotein A5 gene is associated with an increased risk of coronary artery disease and altered triglyceride levels in a Chinese population. Atherosclerosis 185:433–437 Kovar J, Adamkova V, 2008, Lipoprotein lipase activity determined in vivo is lower in carriers of apolipoprotein A-V gene variants 19W and -1131C. Physiol Res 57:555–561 Merkel M, Heeren J, 2005, Give me A5 for lipoprotein hydrolysis! J Clin Invest 115:2694–2696 Vaessen SF, Sierts JA, Kuivenhoven JA et al, 2009, Efficient lowering of triglyceride levels in mice by human apoAV protein variants associated with hypertriglyceridemia. Biochem Biophys Res Commun 379:542–546 Wong-Mauldin K, Raussens V, Forte TM et al, 2009, Apolipoprotein A-V N-terminal domain altered lipid interaction properties in vitro explain hypertriglyceridemic phenotype associated with natural truncation mutants. J Biol Chem 284(48):33369–33376 Kisfali P, Mohas M, Maasz A et al, 2009, Haplotype analysis of the apolipoprotein A5 gene in patients with the metabolic syndrome. Nutr Metab Cardiovasc Dis Ko YL, Ko YS, Wu SM et al, 1997, Interaction between obesity and genetic polymorphisms in the apolipoprotein CIII gene and lipoprotein lipase gene on the risk of hypertriglyceridemia in Chinese. Hum Genet 100:327–333 Nakanishi N, Nishina K, Okamoto M et al, 2004, Clustering of components of the metabolic syndrome and risk for development of type 2 diabetes in Japanese male office workers. Diab Res Clin Pract 63:185–194 Alan S, UlgenMS, Ozturk O et al, 2003, Relation between coronary artery disease, risk factors and intima-media thickness of carotid artery, arterial distensibility, and stiffness index. Angiology 54:261–267 Takase B, Matsushima Y, Uehata A et al, 2008, Endothelial dysfunction, carotid artery plaque burden, and conventional exercise-induced myocardial ischemia as predictors of coronary artery disease prognosis. Cardiovasc Ultrasound 6:61 Wald DS, Bestwick JP, Morton G et al, 2009, Combining carotid intima-media thickness with carotid plaque on screening for coronary heart disease. J Med Screen 16:155–159 Eichenbaum-Voline S, Olivier M, Jones EL et al, 2004, Linkage and association between distinct variants of the APOA1/C3/A4/ A5 gene cluster and familial combined hyperlipidemia. Arterioscler Thromb Vasc Biol 24:167–174 Liu ZK, Hu M, Baum L et al, 2009, Associations of polymorphisms in the apolipoprotein A1/C3/A4/A5 gene cluster with familial combined hyperlipidaemia in Hong Kong Chinese. Atherosclerosis 208(2):427–432 Wang J, BanMR, Kennedy BA et al, 2008, APOA5 genetic variants are markers for classic hyperlipoproteinemia phenotypes and hypertriglyceridemia. Nat Clin Pract Cardiovasc Med 5:730–737 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 39 EP 44 DI 10.1007/s12253-010-9273-7 PG 6 ER PT J AU Liu, B Zhang, Y Liao, M Deng, Z Gong, L Jiang, J Lynn, L Wu, K Miao, X AF Liu, Bo Zhang, Yangde Liao, Mingmei Deng, Zhansheng Gong, Liansheng Jiang, Jiarui Lynn, Ling Wu, Kai Miao, Xiongying TI Clinicopathologic and Prognostic Significance of CD24 in Gallbladder Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gallbladder carcinoma; CD24; Clinicopathology; Prognosis ID Gallbladder carcinoma; CD24; Clinicopathology; Prognosis AB CD24, a small cell surface protein, has emerged as a novel oncogene and prognostic factor for poor outcomes in many human cancers. However, the association of CD24 expression pattern in gallbladder carcinoma with patients’ survival has not been reported. To shed light on this problem, we performed an analysis on the relationship between CD24 expression and prognostic parameters in gallbladder carcinoma. CD24 expression was examined immunohistochemically on paraffin-embedded tissue specimens from 207 patients who underwent surgical treatment for gallbladder carcinoma in the period between January 2004 and May 2009. CD24 positive expression was found in 78.7% (163/207) of the tumor samples. It tended to be associated positively with tumor histological grades and pT stages. Kaplan-Meier curves showed that CD24 positive expression was significantly related to decreased overall survival (p<0.01). Multivariate analysis, including CD24 expression, pT stage, tumor grade, and resection margin involvement, showed that CD24 positive expression was an independent prognostic marker in gallbladder carcinoma (p=0.02; relative risk=1.6). Our data demonstrate for the first time that CD24 is an important marker of malignancy and poor prognosis in gallbladder carcinoma. Its detection combined with cancerous staging may increase the ability of investigators to predict the prognosis of patients with gallbladder carcinoma. Furthermore, the CD24 antigen represents an attractive target for specific therapies with monoclonal antibodies in patients with CD24-overexpressing gallbladder carcinoma, so the detection of CD24 may help clinicians select patients likely to benefit from novel molecular therapies. C1 [Liu, Bo] Central South University, The Second Xiangya Hospital, Department of General Surgery, 410011 Changsha, Hunan Province, China. [Zhang, Yangde] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan Province, China. [Liao, Mingmei] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan Province, China. [Deng, Zhansheng] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan Province, China. [Gong, Liansheng] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan Province, China. [Jiang, Jiarui] Central South University, The Second Xiangya Hospital, Department of General Surgery, 410011 Changsha, Hunan Province, China. [Lynn, Ling] Central South University, The Second Xiangya Hospital, Department of Orthopedics, 410011 Changsha, Hunan Province, China. [Wu, Kai] Central South University, The First Xiangya Hospital, Department of Orthopedics, 410011 Changsha, Hunan Province, China. [Miao, Xiongying] Central South University, The Second Xiangya Hospital, Department of General Surgery, 410011 Changsha, Hunan Province, China. RP Miao, X (reprint author), Central South University, The Second Xiangya Hospital, Department of General Surgery, 410011 Changsha, China. EM mxymxy126@126.com CR Roa I, De Aretxabala X, Araya JC et al, 2006, Preneoplastic lesions in gallbladder cancer. J Surg Oncol 93:615–623 Lewis JT, Talwalkar JA, Rosen CB et al, 2003, Prevalence and risk factors for gallbladder neoplasia in patients with primary sclerosing cholangitis: evidence for a metaplasia-dysplasiacarcinoma sequence. Am J Surg Pathol 31:907–913 Misra S, Chaturvedi A, Misra NC et al, 2003, Carcinoma of the gallbladder. Lancet Oncol 4:167–176 Taguchi T, Kiyokawa N, Mimori K et al, 2003, Pre-B cell antigen receptor-mediated signal inhibits CD24-induced apoptosis in human pre-B cells. J Immunol 170:252–260 Jung KC, Park WS, Kim HJ et al, 2004, TCR independent and caspase-independent apoptosis of murine thymocytes by CD24 cross-linking. J Immunol 172:795–802 Kristiansen G, Sammar M, Altevogt P, 2004, Tumor biological aspects of CD24, a mucin-like adhesion molecule. J Mol Histol 35:255–262 Friederichs J, Zeller Y, Hafezi-Moghadam A et al, 2000, The CD24/P-selectin binding pathway initiates lung arrest of human A125 adenocarcinoma cells. Cancer Res 60:6714–6722 Schindelmann S, Windisch J, Grundmann R et al, 2002, Expression profiling of mammary carcinoma cell lines: correlation of in vitro invasiveness with expression of CD24. Tumor Biol 23:139–145 Senner V, Sturm A, Baur I et al, 1999, CD24 promotes invasion of glioma cells in vivo. J Neuropathol Exp Neurol 58:795–802 Kristiansen G, Schluns K, Yonwei Y et al, 2003, CD24 is an independent prognostic marker of survival in non-small cell lung cancer patients. Br J Cancer 88:231–235 Kristiansen G, Winzer KJ, Mayordomo E et al, 2003, CD24 expression is a new prognostic marker in breast cancer. Clin Cancer Res 9:4906–4913 Kristiansen G, Pilarsky C, Pervan J et al, 2004, CD24 expression is a significant predictor of PSA relapse and poor prognosis in low grade or organ confined prostate cancer. Prostate 58:183–192 Su MC, Hsu C, Kao HL et al, 2006, CD24 expression is a prognostic factor in intrahepatic cholangiocarcinoma. Cancer Lett 235:34–39 Weichert W, Denkert C, Burkhardt M et al, 2005, Cytoplasmic CD24 expression in colorectal cancer independently correlates with shortened patient survival. Clin Cancer Res 11:6574–6581 Kristiansen G, Denkert C, Karsten S et al, 2002, CD24 is expressed in ovarian cancer and is a new independent prognostic marker of patient survival. Am J Pathol 161:1215–1221 Winkler A, Zigeuner R, Rehak P et al, 2007, CD24 expression in urothelial carcinoma of the upper urinary tract correlates with tumour progression. Virchows Arch 450:59–64 Fan YZ, Zhang JT, Yang HC et al, 2002, Expression of MMP-2, TIMP-2 protein and the ratio of MMP-2/TIMP-2 in gallbladder carcinoma and their significance.World J Gastroenterol 8:1138–1143 Taner CB, Nagorney DM, Donohue JH, 2004, Surgical treatment of gallbladder cancer. J Gastrointest Surg 8:83–89 Ma HB, Hu HT, Di ZL et al, 2005, Association of cyclin D1, p16 and retinoblastoma protein expressions with prognosis and metastasis of gallbladder carcinoma. World J Gastroenterol 11:744–747 Aigner S, Ramos CL, Hafezi-Moghadam A et al, 1998, CD24 mediates rolling of breast carcinoma cells on P-selectin. FASEB J 12:1241–1251 Lim SC, 2005, CD24 and human carcinoma: tumor biological aspects. Biomed Pharmacother 59:351–354 Zhang Y, Yao F, Yao X et al, 2009, Role of CCL5 in invasion, proliferation and proportion of CD44+/CD24-phenotype of MCF-7 cells and correlation of CCL5 and CCR5 expression with breast cancer progression. Oncol Rep 21:1113–1121 Welsh JB, Zarrinkar PP, Sapinoso LM et al, 2001, Analysis of gene expression profiles in normal and neoplastic ovarian tissue samples identifies candidate molecular markers of epithelial ovarian cancer. Proc Natl Acad Sci USA 98:1176–1181 Sagiv E, Memeo L, Karin A et al, 2006, CD24 is a new oncogene, early at the multistep process of colorectal cancer carcinogenesis. Gastroenterology 131:630–639 Sagiv E, Arber N, 2008, The novel oncogene CD24 and its arising role in the carcinogenesis of the GI tract: from research to therapy. Exp Rev Gastroenterol Hepatol 2:125–1233 Huang LR, Hsu HC, 1995, Cloning and expression of CD24 gene in human hepatocellular carcinoma: a potential early tumor marker gene correlates with p53 mutation and tumor differentiation. Cancer Res 55:4717–4721 Lee JH, Kim SH, Lee ES et al, 2009, CD24 overexpression in cancer development and progression: a meta-analysis. Oncol Rep 22:1149–1156 Jacob J, Bellach J, Grutzmann R et al, 2004, Expression of CD24 in adenocarcinomas of the pancreas correlates with higher tumor grades. Pancreatology 4:454–460 Park JS, Jung WH, Kim JK et al, 2009, Estrogen receptor α, estrogen receptor β, and progesterone receptor as possible prognostic factor in radically resected gallbladder carcinoma. J Surg Res 152:104–110 Varga M, Obrist P, Schneeberger S et al, 2004, Overexpression of epithelial cell adhesion molecule antigen in gallbladder carcinoma is an independent marker for poor survival. Clin Cancer Res 10:3131–3136 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 45 EP 50 DI 10.1007/s12253-010-9278-2 PG 6 ER PT J AU Sabbatini, M Comi, C Chiocchetti, A Piffanelli, V Car, GP Dianzani, U Monaco, F Cannas, M AF Sabbatini, Maurizio Comi, Cristoforo Chiocchetti, Annalisa Piffanelli, Valentina Car, Giorgio Pier Dianzani, Umberto Monaco, Francesco Cannas, Mario TI Signals of Apoptotic Pathways in Several Types of Meningioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Caspases; Caspase-inhibitor; Bax; bcl-2; TUNEL ID Caspases; Caspase-inhibitor; Bax; bcl-2; TUNEL AB Meningiomas are intracranial tumour derived from meningothelial cells, which aggressive behaviour has been frequently associated to cell apoptosis. In this paper activation of several factors involved in apoptosis has been investigated on biopsies of primary, non recurrent meningiomas. Benign (meningotheliomatous, transitional, fibrous, angiomatous), atypical and anaplastic meningiomas were analysed by immunohistochemistry and western blot, to visualize the occurring of different apoptotic pathways and their association with clinical grading. Apoptotic cell have been detected by a double colorimetric staining for TUNEL and caspase-3 active form. Apoptotic signal positive cells have been detected in all type of meningiomas analysed, with exception of meningotheliomatous meningiomas. Differences have been found in the activation of apoptotic pathways between several types of grade I meningiomas and among benign, anaplastic and atypical meningiomas. An intense expression of several apoptotic inhibitor occurred in grade I meningiomas. The correlation among expression of apoptotic and inhibitory factors and cell proliferation index may suggest that in grade I meningiomas apoptosis may be related to mechanisms involved into tumor cells surviving. Instead in grade II and III meningiomas the same correlation seems indicate an high turnover of tumor cells that might be useful as index of cell proliferation and tumor mass growth. C1 [Sabbatini, Maurizio] University of Eastern Piedmont, Department of Clinical and Experimental Medicine, Laboratory of Human Anatomy, Via Solaroli 17, 28100 Novara, Italy. [Comi, Cristoforo] University of Eastern Piedmont, Department of NeurologyNovara, Italy. [Chiocchetti, Annalisa] University of Eastern Piedmont, Department of Medical Science, Laboratory of ImmunologyNovara, Italy. [Piffanelli, Valentina] University of Eastern Piedmont, Department of Clinical and Experimental Medicine, Laboratory of Human Anatomy, Via Solaroli 17, 28100 Novara, Italy. [Car, Giorgio Pier] Ospedale Maggiore della Carita of Novara, Neurosurgery DivisionNovara, Italy. [Dianzani, Umberto] University of Eastern Piedmont, Department of Medical Science, Laboratory of ImmunologyNovara, Italy. [Monaco, Francesco] University of Eastern Piedmont, Department of NeurologyNovara, Italy. [Cannas, Mario] University of Eastern Piedmont, Department of Clinical and Experimental Medicine, Laboratory of Human Anatomy, Via Solaroli 17, 28100 Novara, Italy. RP Sabbatini, M (reprint author), University of Eastern Piedmont, Department of Clinical and Experimental Medicine, Laboratory of Human Anatomy, 28100 Novara, Italy. EM maurizio.sabbatini@med.unipmn.it CR Perry A, Stafford SL, Scheithauer BW et al, 1997, Meningioma grading. An analysis of histologic parameters. Am J Surg Pathol 21:1455–1465 Perry A, Louis DN, Scheithauer BW et al, 2007, World Health Organization classification of tumours of the central nervous system, 4th edn. IARC, Lyon, pp 164–172 Lowe SW, Jacks T, Housman DE et al, 1994, Abrogation of oncogene-associated apoptosis allows transformation of p53- deficient cells. Proc Natl Acad Sci USA 91:2026–2030 Hopkins-Donaldson S, Bodmer J, Balmas Bourloud K et al, 2000, Loss of caspase-8 expression in highly malignant human neuroblastoma cells correlates with resistance to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. Cancer Res 60:4315–4319 Kayaselcuk F, Zorludemir S, Bal N et al, 2004, The expression of survivin and Ki-67 in meningiomas: correlation with grade and clinical outcome. J Neurooncol 67:209–214 Kostantinidou AE, Givalos N, Gakiopoulou H et al, 2007, Caspase-3 immunohistochemical expression is a maker of apoposis, increased grade and early recurrence in intracranial meningiomas. Apoptosis 12:695–705 Patel T, Gores GJ, Kaufmann SH, 1996, The role of proteases during apoptosis. FASEB J 10:587–597 Vermeulen K, Van Bockstaele DR, Berneman ZN, 2005, Apoptosis: mechanisms and relevance in cancer. Ann Hematol 84:627–639 Elkon KB, 1999, Caspases: multifunctional proteases. J Exp Med 12:1725–1727 Schwerk C, Schulze-Osthoff K, 2003, Non-apoptotic functions of caspases in cellular proliferation and differentiation. Biochem Pharmacol 66:1453–1458 Sasaki T, Lopes MB, Hankins GR, Helm GA, 2002, Expression of surviving, an inhibitor of apoptosis protein, in tumors of the nervous system. Acta Neuropathol 104:105–109 Kim R, Emi R, Tanabe K, Murakami S, Uchida Y, Arihiro K, 2006, Regulation and interplay of apoptotic and non-appoptotic cell death. J Pathol 208:319–326 Strasser A, O’Connor L, Dixit VM, 2000, Apoptosis signalling. Annu Rev Biochem 69:217–245 Cowling V, Downward J, 2002, Caspase-6 is the direct activator of caspase-8 in the cytochrome c-induced apoptosis pathway: absolute requirement for removal of caspase-6 prodomain. Cell Death Diff 9:1046–1056 Adams JM, Cory S, 1998, The Bcl-2 protein family: arbiters of cell survival. Science 281:1322–1326 Shimizu S, Narita M, Tsujimoto Y, 1999, Bcl-2 family proteins regulate the release of apoptogenic cytochrome-c by the mitochrondial channel VDAC. Nature 399:483–487 Tang D, Kidd VJ, 1998, Cleavage of DFF-45/ICAD by multiple caspases is essential for function during apoptosis. J Biol Chem 273:28549–28552 Scott FL, Denault JB, Riedl SJ, Shin H, Renatus M, Salvesen GS, 2005, XIAP inhibits caspase-3 and -7 using two binding sites: evolutionarily conserved mechanism of IAPs. EMBO J 24:645–655 Matsumori Y, Northington FJ, Hong SM, 2006, Reduction of caspase-8 and -9 cleavage is associated with increased c-FLIP and increased binding of Apaf-1 and Hsp70 after neonatal hypoxic/ischemic injury in mice overexpressing Hsp70. Stroke 37:507–512 Kostantinidou AE, Pavlopoulos PM, Patsouris E, Kaklamani L, Davaris P, 1998, Expression of apoptotic and proliferation markers in meningiomas. J Pathol 186:325–330 Charriaut-Marianque C, Ben Ari Y, 1995, A cautionary note on the use of TUNEL stain to determine apoptosis. Neuroreport 7:61–64 Grasl-Kraupp B, Ruttkay-Nedecky B, Koudelka H, Bukowska K, Bursch W, Schultze-Hermann R, 1995, In situ detection of fragmented DNA, TUNEL assay, fails to discriminate among apoptosis, necrosis, and autolytic cell death: a cautionary role. Hepathology 21:1465–1468 Moreno RD, Lizama C, Urzua N, Vergara SP, Reyes JG, 2006, Caspase activation throughout the first wave of spermatogenesis in the rat. Cell Tissue Res 325:533–540 Mosnier JF, Perret AG, Brunon J, Boucheron S, 1996, Expression of the bcl-2 oncoprotein in meningiomas. Am J Pathol 106:652–659 Smith DW, Day TA, 1993, Neurochemical identification of fospositive neurons using two-colour immunoperoxidase staining. J Neurosci Methods 47:73–83 Reno F, Sabbatini M, Lombardi F, Stella M, Pezzato C, Magliacani G, Cannas M, 2003, In vitro mechanical compression induces apoptosis and regulates cytokines release in hypertrophic scars. Wound Rep and Reg 11:331–336 Das A, Tan W-L, Smith DR, 2003, Expression of the inhibitor of apoptosis protein survivin in benign meningiomas. Cancer Lett 193:217–223 Jacobs TW, Prioleau JE, Stilman IE, Schnitt SJ, 1996, Loss of tumor marker-immunostaining intensity on stored paraffin slides of breast cancer. J Nat Cancer Inst 88:1054–1059 Takei H, Buckleair W, Powell SZ, 2008, Immunohistochemical expression of apoptosis regulating proteins and sex hormone receptors in meningiomas. Neuropathology 28:62–68 Armitage PA, Schwindack C, Bastin ME, Whittle IR, 2007, Quantitative assessment of intracranial tumor response to dexamethasone using diffusion, perfusion and permeability magnetic resonance imaging. Magn Reson Imaging 25:303–310 Thoren FB, Romero AI, Hellstrand K, 2006, Oxygen radicals induce poly(ADP-ribose, polymerase-dependent cell death in cytotoxic lymphocytes. J Immunol 176:7301–7307 Fulda S, Kufer MU, Meyer E, Van Valen F, Dockhorn- Dworniczak B, Debatin KM, 2001, Sensitization for death receptor- or drug-induced apoptosis by re-expression of caspase-8 through demethylation or gene transfer. Oncogene 20:5865–5877 Borner C, Monney L, 1999, Apoptosis without caspases: an inefficient molecular guillotine? Cell Death Diff 6:497–507 Sordet O, Rebe C, Plenchette S, Zermati Y, Hermine O, Vainchenker W, Garrido C, Solary E, Dubrez-Daloz L, 2002, Specific involvement of caspases in the differentiation of monocytes into macrophages. Blood 100:4446–4453 Banks DP, Plescia J, Altieri DC, Chen J, Rosenberg SH, Zhang H, NG SC, 1999, Survivin does not inhibit caspase-3 activity. Blood 96:4002–4003 Zangemeister-Wittke U, Simon HU, 2004, An IAP in action: the multiple roles of surviving in differentiation, immunity and malignancy. Cell Cycle 3:1121–1123 Fischer U, Janssen K, Schulze-Osthoff K, 2007, Does caspase inhibition promote clonogenic tumor growth? Cell Cycle 6:3048–3053 Al-Khalaf HH, Lach B, Allam A, Hassounah M, Alkhani A, Elkum N, Alrokayan SA, Aboussekhra A, 2008, Expression of survivin and p16INK4a/Cdk6/pRB proteins and induction of apoptosis in response to radiation and cisplatin in meningioma cells. Brain Res 1188:25–34 McLean CA, Jolley D, Cukier E, Giles G, Gonzales MF, 1993, Atypical and malignant meningiomas: importance of micronecrosis as a prognostic indicator. Histopathology 33:64–70 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 51 EP 59 DI 10.1007/s12253-010-9279-1 PG 9 ER PT J AU Legan, M Tevzic, Tolar, A Luzar, B Marolt, FV AF Legan, Mateja Tevzic, Spela Tolar, Ana Luzar, Bostjan Marolt, Ferlan Vera TI Glucose Transporter-1 (GLUT-1) Immunoreactivity in Benign, Premalignant and Malignant Lesions of the Gallbladder SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Diagnosis; Gallbladder adenocarcinoma; GLUT-1; Immunohistochemistry; Prognosis ID Diagnosis; Gallbladder adenocarcinoma; GLUT-1; Immunohistochemistry; Prognosis AB GLUT-1 is a transmembrane glucose transport protein that allows the facilitated transport of glucose into cells, normally expressed in tissues which depend mainly on glucose metabolism. Enhanced expression of GLUT-1 can also be found in a large spectrum of carcinomas. This study aimed to investigate GLUT-1 expression in gallbladder tissue: from normal tissue samples, hyperplasias, lowgrade and high-grade dysplasias to gallbladder carcinomas. In all, 115 archived samples of gallbladder tissue from 68 patients, presented after cholecystectomy, were immunohistochemically stained for GLUT-1. According to the intensity of GLUT-1 immunoreactivity, samples were divided into negative (stained 0–10% of cells stained), positive with weak to moderate (10–50%) and positive with strong (>50%) GLUT-1 expression. The GLUT-1 immunoreactivity of the samples showed a characteristic increase from premalignant lesions to carcinomas. Normal gallbladder tissue samples did not express GLUT-1 (100%). Weak expression was shown only focally in hyperplasias, but to a greater extent with low-grade dysplasias (20%), high-grade dysplasias (40%) and carcinomas (51.8%). Normal gallbladder tissue is GLUT-1 negative. GLUT-1 expression in carcinoma tissue is significantly higher than in dysplastic lesions. Strong GLUT-1 expression indicates 100% specificity for detecting gallbladder carcinomas. Therefore, GLUT-1 is a candidate as a diagnostic as well as a tissue prognostic marker in gallbladder carcinoma patients. C1 [Legan, Mateja] University of Ljubljana, Faculty of Medicine, Institute of Histology and Embryology, Korytkova 2, SI-1000 Ljubljana, Slovenia. [Tevzic, Spela] University of Ljubljana, Faculty of Medicine, Institute of Histology and Embryology, Korytkova 2, SI-1000 Ljubljana, Slovenia. [Tolar, Ana] University of Ljubljana, Faculty of Medicine, Institute of Histology and Embryology, Korytkova 2, SI-1000 Ljubljana, Slovenia. [Luzar, Bostjan] University of Ljubljana, Faculty of Medicine, Institute of PathologyLjubljana, Slovenia. [Marolt, Ferlan Vera] University of Ljubljana, Faculty of Medicine, Institute of PathologyLjubljana, Slovenia. RP Legan, M (reprint author), University of Ljubljana, Faculty of Medicine, Institute of Histology and Embryology, SI-1000 Ljubljana, Slovenia. EM mateja.legan@mf.uni-lj.si CR Dills WL, 1993, Nutritional and physiological consequences of tumor glycolysis. Parasitology 107:S177–S186 Pessin JE, Bell GI, 1992, Mammalian facilitative glucose transporter family: structure and molecular regulation. Ann Rev Physiol 54:911–930 Ebert BL, Firth JD, Ratcliffe PJ, 1995, Hypoxia and mitochondrial inhibitors regulate expression of glucose transporter-1 via distinct cis-acting sequences. J Biol Chem 270:29083–29089 Pardridge WM, Boado RJ, Farrell CR, 1990, Brain-type glucose transporter, GLUT-1, is selectively localized to the blood-brain barrier. J Biol Chem 265:18035–18040 Gould GW, Holman GD, 1993, The glucose transporter family: structure, function and tissue-specific expression. Biochem J 53:4204–4211 Froehner SC, Davies A, Baldwin SA, Lienhard GE, 1998, The blood-nerve barrier is rich in glucose transporter. J Neurocytol 17:173–178 Younes M, Lechago LV, Somoano JR, Mosharaf M, Lechago J, 1996, Wide expression of the human erythrocyte glucose transporter Glut-1 in human cancers. Cancer Res 56:1164–1167 Noguchi Y, Marat D, Saito A et al, 1999, Expression of facilitative glucose transporters in gastric tumors. Hepato Gastroenterol 46:2683–2689 Reisser C, Eichhorn K, Herold-Mende C, Born AL, Bannasch P, 1999, Expression of facilitative glucose transporter proteins during development of squamous cell carcinomas of the head and neck. Int J Cancer 80:194–198 Haber RS, Rathan A, Weiser KR et al, 1998, GLUT1 glucose transporter expression in colorectal carcinoma: a marker for poor prognosis. Cancer 83:34–40 Kalir T, Wang B, Goldfischer M et al, 2002, Immunohistochemical staining of GLUT1 in benign, borderline, and malignant ovarian epithelia. Cancer 94:1078–1082 Ashton-Sager A, Paulino AFG, Afify AM, 2006, GLUT-1 is preferentially expressed in atypical endometrial hyperplasia and endometrial adenocarcinoma. Appl Immunohistochem Mol Morphol 14:187–192 Chandan VS, Faquin WC, Wilbur DC, Khurana KK, 2006, The utility of GLUT-1 immunolocalization in cell blocks: An adjunct to the fine needle aspiration diagnosis of cystic squamous lesions of the head and neck. Cancer 108:124–128 Ito H, Duxbury M, Zinner MJ, Ashley SW, Whang EE, 2004, Glucose transporter-1 gene expression is associated with pancreatic cancer invasiveness and MMP-2 activity. Surgery 136:548–556 Grover-McKay M, Walsh SA, Seftor EA, Thomas PA, Hendrix MJC, 1998, Role for glucose transporter 1 protein in human breast cancer. Pathol Oncol Res 4:115–120 Chandan VS, Faquin WC, Wilbur DC, Khurana KK, 2006, The role of immunolocalization of CD57 and GLUT-1 in cell blocks in fine-needle aspiration diagnosis of papillary thyroid carcinoma. Cancer 108:331–336 Kim YW, Park YK, Yoon T, Lee SM, 2002, Expression of GLUT-1 glucose transporter in gallbladder carcinomas. Hepatogastroenterol 49:907–911 Evans A, Bates V, Troy H et al, 2008, GLUT-1 as a therapeutic target: increased chemoresistance and HIF-1 independent link with cell turnover is revealed through COMPARE analysis and metabolomic studies. Cancer Chemother Pharmacol 61:377–393 Legan M, Luzar B, Marolt VF, 2009, Expression of cyclooxygenase-2, glucose transporter-1 and angiogenesis in gallbladder carcinomas and their impact on prognosis. Scand J Gastroenterol 44:1101–1108 Cooper R, Sarioglu S, Sokmen S et al, 2003, Glucose transporter-1, GLUT-1): a potential marker of prognosis in rectal carcinoma? Brit J Cancer 89:870–876 Kanoh K, Shimura T, Tsutsumi S et al, 2001, Significance of contracted cholecystitis lesions as high risk for gallbladder carcinogenesis. Cancer Lett 169:7–14 Tsuchida A, Itoi T, Aoki T, Koyanagi Y, 2003, Carcinogenetic process in gallbladder mucosa with pancreaticobiliary maljunction. Oncol Rep 10:1693–1699 Roa I, Araya JC, Villaseca M et al, 1996, Preneoplastic lesions and gallbladder cancer: an estimate of the period required for progression. Gastroenterol 111:232–236 Cantuaria G, Fagotti A, Ferrandina G et al, 2001, GLUT-1 expression in ovarian carcinoma: association with survival and response to chemotherapy. Cancer 92:1144–1150 Griffiths EA, Pritchard SA, Welch IM et al, 2005, Is the hypoxiainducible factor pathway important in gastric cancer? Eur J Cancer 41:2792–2805 Airley R, Loncaster J, Davidson S et al, 2001, Glucose transporter Glut-1 expression correlates with tumor hypoxia and predicts metastasis-free survival in advanced carcinoma of the cervix. Clin Cancer Res 7:928–934 Miller JH, Mullin JM, McAvoy E, Kleinzeller A, 1992, Polarity of transport of 2-deoxy-D-glucose and D-glucose by cultured renal epithelia, LLC-PK1). Biochim Biophys Acta 1110:209–217 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 61 EP 66 DI 10.1007/s12253-010-9281-7 PG 6 ER PT J AU Song, HJ Cao, Z Jung, HY Nam, WS Kim, YS Lee, YJ Park, SW AF Song, Hwi Jae Cao, Zhang Jung, Hwan Yoon Nam, Woo Suk Kim, Young Su Lee, Young Jung Park, Sang Won TI Genetic Alterations and Expression Pattern of CEACAM1 in Colorectal Adenomas and Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CEACAM1; Colorectal cancer; Somatic mutation; Expression; Immunohistochemistry ID CEACAM1; Colorectal cancer; Somatic mutation; Expression; Immunohistochemistry AB Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is expressed on epithelial cells throughout the intestinal tract and is a negative regulator of tumor cell growth, suggesting that it may function as a tumor suppressor. In this study, to determine whether the CEACAM1 is involved in colorectal tumorigenesis, we have investigated the genetic alterations, including mutations and allelic loss, of the CEACAM1 gene in 17 colonic adenomas and 123 sporadic colorectal cancers. In addition, the expression pattern of the CEACAM1 protein was examined in 60 colonic adenomas and 123 sporadic colorectal adenocarcinomas. No mutation was found in colonic adenomas, but four somatic missense mutations, L36F, T312I, V398I and A445V, were detected in colorectal cancers. Interestingly, all of the mutations were found in left-side colon cancers of the patients with clinical stage III. In LOH analysis, nine adenomas were informative for at least one of the markers and five (55.6%) showed allelic loss. Thirty-eight cancers were informative at D19S211 and D19S872 markers and 21 (56.3%) showed LOH at these markers. Statistically, the frequency of allelic loss at the CEACAM1 locus was not associated with clinicopathologic parameters (P>0.05). In immunohistochemical analysis, loss of expression of CEACAM1 protein was detected in nine (15.0%) and 30 (24.4%) of 60 colorectal adenomas and 123 colorectal cancers. Statistically, there was no significant relationship between loss of CEACAM1 expression and clinicopathologic parameters, including clinical stage, tumor location, tumor size, lymph node metastasis and 5-year survival (P>0.05). These data suggest that genetic alteration and loss of expression of the CEACAM1 may contribute to the development of colorectal cancers, as an early event. C1 [Song, Hwi Jae] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Seocho-gu, 137-701 Seoul, South Korea. [Cao, Zhang] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Seocho-gu, 137-701 Seoul, South Korea. [Jung, Hwan Yoon] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Seocho-gu, 137-701 Seoul, South Korea. [Nam, Woo Suk] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Seocho-gu, 137-701 Seoul, South Korea. [Kim, Young Su] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Seocho-gu, 137-701 Seoul, South Korea. [Lee, Young Jung] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Seocho-gu, 137-701 Seoul, South Korea. [Park, Sang Won] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Seocho-gu, 137-701 Seoul, South Korea. RP Park, SW (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM wonsang@catholic.ac.kr CR Weitz J, Koch M, Debus J et al, 2005, Colorectal cancer. Lancet 365:153–165 Shin HR, Jung KW, Won YJ et al, 2004, 2002 Annual report of the central cancer registry: based on registered data from 139 hospitals. Cancer Res Treat 36:103–114 Fearon ER, Vogelstein B, 1990, A genetic model for colorectal tumorigenesis. Cell 61:759–767 Thompson JA, Grunert F, Zimmermann W, 1991, Carcinoembryonic antigen gene family: molecular biology and clinical perspectives. J Clin Lab Anal 5:344–366 Tynan K, Olsen A, Trask B et al, 1992, Assembly and analysis of cosmid contigs in the CEA-gene family region of human chromosome 19. Nucleic Acids Res 20:1629–1636 Beauchemin N, Draber P, Dveksler G et al, 1999, Redefined nomenclature for members of the carcinoembryonic antigen family. Exp Cell Res 252:243–249 Watt SM, Teixeira AM, Zhou GQ et al, 2001, Homophilic adhesion of human CEACAM1 involves N-terminal domain interactions: structural analysis of the binding site. Blood 98:1469–1479 Shively JE, 2004, CEACAM1 and hyperplastic polyps: new links in the chain of events leading to colon cancer. Oncogene 23:9303–9305 Kleinerman DI, Troncoso P, Lin SH et al, 1995, Consistent expression of an epithelial cell adhesion molecule, C-CAM, during human prostate development and loss of expression in prostate cancer: implication as a tumor suppressor. Cancer Res 55:1215–1220 Luo W, Wood CG, Earley K et al, 1997, Suppression of tumorigenicity of breast cancer cells by an epithelial cells adhesion molecule, C-CAM1): the adhesion and growth suppression are mediated by different domains. Oncogene 14:1697–1704 Houde C, Roy S, Leung N et al, 2003, The cell adhesion molecule CEACAM1-L is a substrate of caspase-2-mediated cleavage in apoptotic mouse intestinal cells. J Biol Chem 278:16929–16935 Nollau P, Scheller H, Kona-Horstmann M et al, 1997, Expression of CD66a, human C-CAM, and other members of the carcinoembryonic antigen gene family of adhesion molecules in human colorectal adenomas. Cancer Res 57:2354–2357 Zhang L, Zhou W, Velculescu VE et al, 1997, Gene expression profiles in normal and cancer cells. Science 276:1268–1272 Nittka S, Gunther J, Ebisch C et al, 2004, The human tumor suppressor CEACAM1 modulates apoptosis and is implicated in early colorectal tumorigenesis. Oncogene 23:9306–9313 Oliveira-Ferrer L, Tilki D, Ziegeler G et al, 2004, Dual role of carcinoembryonic antigen-related cell adhesion molecule 1 in angiogenesis and invasion of human urinary bladder cancer. Cancer Res 64:8932–8938 Horst AK, Ito WD, Dabelstein J et al, 2006, Carcinoembryonic antigen-related cell adhesion molecule 1 modulates vascular remodeling in vitro and in vivo. J Clin Invest 116:1596–1605 Tilki D, Irmak S, Oliveira-Ferrer L et al, 2006, CEA-related cell adhesion molecule-1 is involved in angiogenic switch in prostate cancer. Oncogene 25:4965–4974 Leung N, Turbide C, Olson M et al, 2006, Deletion of the carcinoembryonic antigen-related cell adhesion molecule 1, Ceacam1, gene contributes to colon tumor progression in a murine model of carcinogenesis. Oncogene 25:5527–5536 Astler VB, Coller FA, 1954, The prognostic significance of direct extension of carcinoma of the colon and rectum. Ann Surg 139:846–851 Kononen J, Bubendorf L, Kallioniemi A et al, 1998, Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4:844–847 Lee JY, Dong SM, Kim SY et al, 1998, A simple, precise and economical microdissection technique for analysis of genomic DNA from archival tissue sections. Virchows Arch 433:305–309 Park WS, Oh RR, Kim YS et al, 2000, Absence of mutations in the kinase domain of the Met gene and frequent expression of Met and HGF/SF protein in primary gastric carcinomas. APMIS 108:195–200 Zalzali H, Naudin C, Bastide P et al, 2008, CEACAM1, a SOX9 direct transcriptional target identified in the colon epithelium. Oncogene 27:7131–7138 Kunath T, Ordonez-Garcia C, Turbide C et al, 1995, Inhibition of colonic tumor cell growth by biliary glycoprotein. Oncogene 11:2373–2383 Leung N, Turbide C, Balachandra B et al, 2008, Intestinal tumor progression is promoted by decreased apoptosis and dysregulated Wnt signaling in Ceacam1-/- mice. Oncogene 27:4943–4953 Bastide P, Darido C, Pannequin J et al, 2007, Sox9 regulates cell proliferation and is required for Paneth cell differentiation in the intestinal epithelium. J Cell Biol 178:635–648 Izzi L, Turbide C, Houde C et al, 1999, cis-Determinants in the cytoplasmic domain of CEACAM1 responsible for its tumor inhibitory function. Oncogene 18:5563–5572 Wikstrom K, Kjellstrom G, Obrink B, 1996, Homophilic intercellular adhesion mediated by C-CAM is due to a domain Idomain 1 reciprocal binding. Exp Cell Res 227:360–366 Fournes B, Farrah J, Olson M et al, 2003, Distinct Rho GTPase activities regulate epithelial cell localization of the adhesion molecule CEACAM1. Involvement of the CEACAM1 transmembrane domain. Mol Cell Biol 23:7291–7304 Phan D, Cheng CJ, Galfione M et al, 2004, Identification of Sp2 as a transcriptional repressor of carcinoembryonic antigen-related cell adhesion molecule 1 in tumorigenesis. Cancer Res 64:3072–3078 Tanaka K, Hinoda Y, Takahashi H et al, 1997, Decreased expression of biliary glycoprotein in hepatocellular carcinomas. Int J Cancer 74:15–19 Huang J, Simpson JF, Glackin C et al, 1998, Expression of biliary glycoprotein, CD66a, in normal and malignant breast epithelial cells. Anticancer Res 18:3203–3212 Busch C, Hanssen TA, Wagener C et al, 2002, Down-regulation of CEACAM1 in human prostate cancer: correlation with loss of cell polarity, increased proliferation rate, and Gleason grade 3 to 4 transition. Hum Pathol 33:290–298 Ebrahimnejad A, Streichert T, Nollau P et al, 2004, CEACAM1 enhances invasion and migration of melanocytic and melanoma cells. Am J Pathol 165:1781–1787 Liu W, Wei W, Winer D et al, 2007, CEACAM1 impedes thyroid cancer growth but promotes invasiveness: a putative mechanism for early metastases. Oncogene 26:2747–2758 Dango S, Sienel W, Schreiber M et al, 2008, Elevated expression of carcinoembryonic antigen-related cell adhesion molecule 1, CEACAM-1, is associated with increased angiogenic potential in non-small-cell lung cancer. Lung Cancer 60:426–433 Zhou CJ, Liu B, Zhu KX et al, 2009, The different expression of carcinoembryonic antigen-related cell adhesion molecule 1, CEACAM1, and possible roles in gastric carcinomas. Pathol Res Pract 205:483–489 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 67 EP 74 DI 10.1007/s12253-010-9282-6 PG 8 ER PT J AU Topic, A Ljujic, M Nikolic, A Petrovic-Stanojevic, N Dopudja-Pantic, V Mitic-Milikic, M Radojkovic, D AF Topic, Aleksandra Ljujic, Mila Nikolic, Aleksandra Petrovic-Stanojevic, Natasa Dopudja-Pantic, Vesna Mitic-Milikic, Marija Radojkovic, Dragica TI Alpha-1-antitrypsin Phenotypes and Neutrophil Elastase Gene Promoter Polymorphisms in Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Alpha-1-antitrypsin; Neutrophil elastase; Polymorphisms; Lung cancer ID Alpha-1-antitrypsin; Neutrophil elastase; Polymorphisms; Lung cancer AB Imbalance between neutrophil elastase and alpha-1-antitrypsin (AAT) leads to emphysema in smokers as well as in patients with inherited alpha-1-antitrypsin deficiency. AAT as a proven inhibitor of apoptosis may play role in lung cancer (LC) progression. The aim was to analyse AAT protein variants and polymorphism in promoter region of the neutrophil elastase gene (ELA2) in patients with primary lung cancer. AAT phenotypisation by isoelectric focusing method and ELA2 gene promoter characterization by DNA sequencing were performed in 66 patients with primary lung cancer. Results showed that the frequency of M1 allele and PiM1 homozygotes in LC patients was significantly higher when compared to the healthy subjects (f=0.6360 and 0.7424 respectively). The most frequent ELA2 promoter region genotypes in LC patients were −903TT and −741GG. There were significantly more patients with intermediate and high ELA2 genotype activity, compared to those with low activity (91% vs. 9%, respectively). In conclusion, we found that PiM1 homozygosity could be associated with the lung cancer, probably due to increased synthesis of this antiapoptotic protein. Non-MM variants of AAT and ELA2 genotypes with predicted intermediate or high activity could also represent a risk factor for aggressive form of lung cancer associated with extrathoracic metastases. C1 [Topic, Aleksandra] Belgrade University, Faculty of Pharmacy, Institute of Medical Biochemistry, Vojvode Stepe 450, 11221 Belgrade, Serbia. [Ljujic, Mila] University of Belgrade, Institute of Molecular Genetics and Genetic EngineeringBelgrade, Serbia. [Nikolic, Aleksandra] University of Belgrade, Institute of Molecular Genetics and Genetic EngineeringBelgrade, Serbia. [Petrovic-Stanojevic, Natasa] Zvezdara University Medical CenterBelgrade, Serbia. [Dopudja-Pantic, Vesna] Zvezdara University Medical CenterBelgrade, Serbia. [Mitic-Milikic, Marija] Clinical Centre of Serbia, Institute for Lung Diseases and TuberculosisBelgrade, Serbia. [Radojkovic, Dragica] University of Belgrade, Institute of Molecular Genetics and Genetic EngineeringBelgrade, Serbia. RP Topic, A (reprint author), Belgrade University, Faculty of Pharmacy, Institute of Medical Biochemistry, 11221 Belgrade, Serbia. EM aleksandra.topic1@gmail.com CR Cancer Research UK, 2005, Commonly diagnosed cancers worldwide. Retrieved on 2008-01-11. http://info.cancerresearchuk. org/cancerstats/world/index.htm Mattson ME, Pollack ES, Cullen JW, 1987, What are the odds that smoking will kill you? Am J Public Health 77:425–431 Bach PB, Kattan MW, Thornquist MD, Kris MG, Tate RC, Barnett MJ, Hsieh LJ, Begg CB, 2003, Variations in lung cancer risk among smokers. J Natl Cancer Inst 95:470–478 Yao H, Rahman I, 2009, Current concepts on the role of inflammation in COPD and lung cancer. Curr Opin Pharmacol 9:375–383 Carp H, Miller F, Hoidal JR, Janoff A, 1982, Potential mechanism of emphysema: alpha 1-proteinase inhibitor recovered from lungs of cigarette smokers contains oxidized methionine and has decreased elastase inhibitory capacity. Proc Natl Acad Sci USA 79:2041–2045 Taggart C, Cervantes-Laurean D, Kim G, McElvaney NG, Wehr N, Moss J, Levine RL, 2000, Oxidation of either methionine 351 or methionine 358 in alpha 1-antitrypsin causes loss of antineutrophil elastase activity. J Biol Chem 75:27258–27265 Walser T, Cui X, Yanagawa J, Lee JM, Heinrich E, Lee G, Sharma S, Dubinett SM, 2008, Smoking and lung cancer: the role of inflammation. Proc Am Thorac Soc 5:811–815 Petrache I, Fijalkowska I, Zhen L, Medler TR, Brown E, Cruz P, Choe KH, Taraseviciene-Stewart L, Scerbavicius R, Shapiro L, Zhang B, Song S, Hicklin D, Voelkel NF, Flotte T, Tuder RM, 2006, A novel antiapoptotic role for α1-antitrypsin in the prevention of pulmonary emphysema. Am J Respir Crit Care Med 173:1222–1228 Zimmer M, Medcalf RL, Fink TM, Mattmann C, Lichter P, Jenne DE, 1992, Three human elastase-like genes coordinately expressed in myelomonocytic lineage are organized as a single genetic locus on 19pter. Proc Natl Acad Sci 89:8215–8223 Takahashi H, Nukiwa T, Basset P, Crystal RG, 1988, Myelomonocytic cell lineage expression of the neutrophil elastase gene. J Biol Chem 263:2543–2547 Kostmann R, 1956, Infantile genetic agranulocytosis. Acta Paediatr 45(suppl 105):1–78 Ancliff PJ, Gale RE, Liesner R, Hann IM, Linch DC, 2001, Mutations in the ELA2 gene encoding neutrophil elastase are present in most patients with sporadic severe congenital neutropenia but only in some patients with the familial form of the disease. Blood 98:2645–2650 Yamashita J, Tashiro K, Yoneda S, Kawahara K, Shirakusa T, 1996, Local increase in polymorphonuclear leukocyte elastase is associated with tumor invasiveness in non-small cell lung cancer. Chest 109:1328–1334 Taniguchi K, Yang P, Jett J, Bass E, Meyer R, Wang Y, Deschamps C, Liu W, 2002, Polymorphisms in the promoter region of the neutrophil elastase gene are associated with lung cancer development. Clin Cancer Res 8:1115–1120 Park JY, Chen L, Lee J, Sellers T, Tockman MS, 2005, Polymorphisms in the promoter region of neutrophil elastase gene and lung cancer risk. Lung Cancer 48:315–321 Topic A, Juranic Z, Jelic S, Magazinovic Golubicic I, 2009, Polymorphism of alpha-1-antitrypsin in haematological malignancies. Genet Mol Biol 32:716–719 Kishimoto Y, Yamada S, Hirayama C, 1990, An association between AAT phenotype and chronic liver disease. Hum Genet 84:132–136 Topic AS, Jelic-Ivanovic ZD, Spasojevic-Kalimanovska VV, Spasic SM, 2006, Association of moderate alpha-1-antitrypsin deficiency with lung cancer in the Serbian population. Arch Med Res 37:866–870 Yang P, Wentzlaff KA, Katzmann JA, Marks RS, Allen MS, Lesnick TG, Lindor NM, Myers JL, Wiegert E, Midthun DE, Thibodeau SN, Krowka MJ, 1999, Alpha1-antitrypsin deficiency allele carriers among LC patients. Cancer Epidemiol Biomarkers Prev 8:461–465 Pott GB, Chan ED, Dinarello CA, Shapiro L, 2009, α-1- Antitrypsin is an endogenous inhibitor of proinflammatory cytokine production in whole blood. J Leukoc Biol 85:886– 895 Hadzic R, Nita I, Tassidis H, Riesbeck K, Wingren AG, Janciauskiene S, 2006, α1-Antitrypsin inhibits Moraxella catarrhalis MID protein-induced tonsillar B cell proliferation and IL-6 release. Immunol Lett 102:141–147 Molano RD, Pileggi A, Song S, Zahr E, Jose SS, Molina J, Fort A, Wasserfall C, Ricordi C, Atkinson MA, Inverardi L, 2008, Prolonged islet allograft survival by alpha-1 antitrypsin: the role of humoral immunity. Transplant Proc 40:455–456 Churg A, Dai J, Zay K, Karsan A, Hendricks R, Yee C, Martin R, MacKenzie R, Xie C, Zhang L, Shapiro S, Wright JL, 2001, Alpha-1-antitrypsin and a broad spectrum metalloprotease inhibitor, RS113456, have similar acute anti-inflammatory effects. Lab Invest 81:1119–1131 Petrache I, Fijalkowska I, Medler TR, Skirball J, Cruz P, Zhen L, Petrache HI, Flotte TR, Tuder RM, 2006, α-1 antitrypsin inhibits caspase-3 activity, preventing lung endothelial cell apoptosis. Am J Pathol 169:1155–1166 Maneckjee R, Minna JD, 1994, Opioids induce while nicotine suppresses apoptosis in human lung cancer cells. Cell Growth Differ 5:1033–1040 Yamashita J, Ogawa M, Abe M, Hayashi N, Kurusu Y, Kawahara K, Shirakusa T, 1997, Tumor neutrophil elastase is closely associated with the direct extension of non-small cell lung cancer into the aorta. Chest 111:885–890 Yang P, Bamlet WR, Sun Z, Ebbert JO, Aubry MC, Taylor WR, Marks RS, Deschamps C, Swensen SJ, Wieben ED, Cunningham JM, Melton LJ, de Andrade M, 2005, α1-antitrypsin and neutrophil elastase imbalance and lung cancer risk. Chest 128:445–452 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 75 EP 80 DI 10.1007/s12253-010-9283-5 PG 6 ER PT J AU Olah, Balogh, E Pajor, L Jakab, Zs AF Olah, Eva Balogh, Erzsebet Pajor, Laszlo Jakab, Zsuzsanna TI Ten-year Experiences on Initial Genetic Examination in Childhood Acute Lymphoblastic Leukaemia in Hungary (1993–2002). Technical Approaches and Clinical Implementation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ALL; Childhood; Chromosome aberration; FISH; Prognostic factor ID ALL; Childhood; Chromosome aberration; FISH; Prognostic factor AB A nationwide study was started in 1993 to provide genetic diagnosis for all newly diagnosed childhood ALL cases in Hungary using cytogenetic examination, DNA-index determination, FISH (aneuploidy, ABL/BCR, TEL/AML1) and molecular genetic tests (ABL/BCR, MLL/AF4, TEL/AML1). Aim of the study was to assess the usefullness of different genetic methods, to study the frequency of various aberrations and their prognostic significance. Results were synthesized for genetic subgrouping of patients. To assess the prognostic value of genetic aberrations overall and event-free survival of genetic subgroups were compared using Kaplan-Meier method. Prognostic role of aberrations was investigated by multivariate analysis (Cox’s regression) as well in comparison with other factors (age, sex, major congenital abnormalities, initial WBC, therapy, immunophenotype). Five hundred eighty-eight ALL cases were diagnosed between 1993–2002. Cytogenetic examination was performed in 537 (91%) (success rate 73%), DNA-index in 265 (45%), FISH in 74 (13%), TEL/AML1 RT-PCR in 219 (37%) cases producing genetic diagnosis in 457 patients (78%). Proportion of subgroups with good prognosis in prae-B-cell ALL was lower than expected: hyperdiploidB 18% (73/400), TEL/AML1+ 9% (36/400). Univariate analysis showed significantly better 5-year EFS in TEL/AML1+ (82%) and hyperdiploidB cases (78%) than in tetraploid (44%) or pseudodiploid (52%) subgroups. By multivariate analysis main negative prognostic factors were: congenital abnormalities, high WBC, delay in therapy, specific translocations. Conclusion: Complementary use of each of genetic methods used is necessary for reliable genetic diagnosis according to the algorythm presented. Specific genetic alterations proved to be of prognostic significance. C1 [Olah, Eva] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98 Nagyerdei krt, 4012 Debrecen, Hungary. [Balogh, Erzsebet] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 98 Nagyerdei krt, 4012 Debrecen, Hungary. [Pajor, Laszlo] University of Pecs, Department of PathologyPecs, Hungary. [Jakab, Zsuzsanna] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. RP Olah, (reprint author), Medical and Health Science Center, University of Debrecen, Department of Pediatrics, 4012 Debrecen, Hungary. EM eolah@dote.hu CR Anastasi J, 1993, in situ hybridization in leukemia. Applications in diagnosis, subclassification, and monitoring the response to therapy. Ann N Y Acad Sci 677:214–224 Andreasson P, Hoglund M, Bekassy AN, Garwicz S, Heldrup J, Mitelman F, Johansson B, 2000, Cytogenetic and FISH studies of a single center consecutive series of 152 childhood acute lymphoblastic leukaemias. Eur J Haematol 65(1):40–51 Armstrong SA, Look AT, 2005, Molecular genetics of acute lymphoblastic leukaemia. J Clin Oncol 23(26):6306–6315 Berger A, Strehl S, Hekele A, Ambros PF, Haas OA, Gadner H, 1994, Interphase cytogenetic study of childhood acute lymphoblastic leukaemia. Med Pediatr Oncol 23(5):413–421 Chessels JM, Bailey C, Richards SM, 1995, Intensification of treatment and survival in all children with lymphoblastic leukaemia: Results of UK Medical Research Council trial UKALL X. Lancet 345:143 Chessels JM, Swansbury GJ, Reeves B, Bailey CC, Richards SM, 1997, Cytogenetics and prognosis in childhood lymphoblastic leukaemia: results of MRC UKALL X. Medical Research Council Working Party in Childhood Leukaemia. Br J Haematol 99(1):3–100 Gray JW, Kallioniemi A, Kallioniemi O, Pallavicini M, Waldman F, Pinkel D, 1992, Molecular cytogenetics: diagnosis and prognostic assessment. Curr Opin Biotechnol 3(6):623–631 Greaves MF, Wiemels J, 2003, Origins of chromosome translocations in childhood leukaemia. Nat Rev Cancer 3(9):639–649 Harbott J, Ritterbach J, Ludwig WD, Bartram CR, Reiter A, Lampert F, 1993, Clinical significance of cytogenetic studies in childhood acute lymphoblastic leukaemia: experience of the BFM trials. Recent Results Cancer Res 131:123–132 Heim S, Mitelman F, 1992, Cytogenetic analysis in the diagnosis of acute leukaemia. Cancer 70(6 Suppl):1701–1709 Huh YO, Andreeff M, 1994, Flow cytometry. Clinical and research applications in haematologic malignancies. Hematol Oncol Clin North Am 8(4):703–723 ISCN, 1995, An international system for human cytogenetic nomenclature. Karger, Basel Johansson B, Mertens F, Mitelman F, 2004, Clinical and biological importance of cytogenetic abnormalities in childhood and adult acute lymphoblastic leukaemia. Ann Med 36(7):492–503 Kersey JH, 1997, Fifty years of studies of the biology and therapy of childhood leukaemia. Blood 90(11):4243–4251 Markasz L, Kis LL, Stuber G, Flaberg E, Otvos R, Eksborg S, Skribek H, Olah E, Szekely L, 2007, Hodgkin-lymhoma-derived cells show high sensitivity to dactinomycin and paclitaxel. Leuk Lymphoma 48(9):1835–1845 Markasz L, Stuber G, Flaberg E, Gustafsson Jernberg A, Eksborg S, Olah E, Skribek H, Szekely L, 2006, Cytotoxic drug sensitivity of Epstein-Barr virus transformed lymphoblastoid B cells. BMC Cancer 6(1):265 Markasz L, Stuber G, Vanherberghen B, Flaberg E, Olah E, Carbone E, Eksborg S, Klein E, Skribek H, Szekely L, 2007, Effect of frequently used chemotherapeutic drugs on the cytotoxic activity of human natural killer cells. Mol Cancer Ther 6:644–654 Markasz L, Hajas G, Kiss A, Lontay B, Rajnavolgyi E, Erdodi F, Olah E, 2007, Granulocyte colony stimulating factor increases drug resistance to daunorubicin and induces proliferation of leukaemic blast cells. Pathol Oncol Res, in press) Mitelman F, 1986, Geographic heterogeneity of chromosome aberrations in haematologic disorders. Cancer Genet Cytogenet 20, 3–4):203–208 Mitelman F, Heim S, 1992, Quantitative acute leukaemia cytogenetics. Genes Chromosomes Cancer 5(1):57–66 Moricke A, Zimmermann M, Reiter A, Gadner H, Odenwald E, Harbott J, Ludwig WD, Riehm H, Schrappe M, 2005, Prognostic impact of age in children and adolescents with acute lymphoblastic leukaemia: data from the trials ALL-BFM 86, 90, and 95. Klin Padiatr 6:310–320 Pinkel D, 1987, Curing children of leukaemia. Cancer 59, 10):1683–1691 Pinkel D, 1989, Species-specific therapy of acute lymphoid leukaemia. Haematol Blood Transfus 32:27–36 Pui CH, Crist WM, 1992, Cytogenetic abnormalities in childhood acute lymphoblastic leukaemia correlates with clinical features and treatment outcome. Leuk Lymphoma 7(4):259–274 Pui CH, Evans WE, 1998, Acute lymphoblastic leukaemia. N Engl J Med 339:605–615 Raimondi SC, 1993, Current status of cytogenetic research in childhood acute lymphoblastic leukaemia. Blood 81(9):2237– 2251 Ritterbach J, Hiddemann W, Beck JD, Schrappe M, Janka-Schaub G, Ludwig WD, Harbott J, Lampert F, 1998, Detection of hyperdiploid karyotypes, >50 chromosomes, in childhood acute lymphoblastic leukaemia, ALL, using fluorescence in situ hybridization, FISH). Leukaemia 12(3):427–433 Rowley JD, 1999, The role of chromosome translocations in leukaemogenesis. Semin Hematol 4(Suppl 7):59–72 Schrappe M, Camitta B, Pui CH, Eden T, Gaynon P, Gustafsson G, Janka-Schaub GE, Kamps W, Masera G, Sallan S, Tsuchida M, Vilmer E, 2000, Long-term results of large prospective trials in childhood acute lymphoblastic leukaemia. Leukaemia 12:2193– 2194 Schrappe M, Reiter A, Zimmermann M, Harbott J, Ludwig WD, Henze G, Gadner H, Odenwald E, Riehm H, 2000, Long-term results of four consecutive trials in childhood ALL performed by the ALL-BFM study group from 1981 to 1995. Berlin-Frankfurt- Munster. Leukaemia 12:2205–2222 Schuler D, 1999, Systemizing childhood cancer care in Hungary: twenty-five years of progress. Med Pediatr Oncol 32(1):68–70 Secker-Walker LM, 1990, Prognostic and biological importance of chromosome findings in acute lymphoblastic leukaemia. Cancer Genet Cytogenet 49(1):1–13 Trueworthy R, Shuster J, Look T, Crist W, Borowitz M, Carroll A, Frankel L, Harris M, Wagner H, Haggard M et al, 1992, Ploidy of lymphoblasts is the strongest predictor of treatment outcome in B-progenitor cell acute lymphoblastic leukaemia of childhood: a Pediatric Oncology Group study. J Clin Oncol 10(4):606–613 Yeoh EJ, Ross ME, Shurtleff SA, Williams WK, Patel D, Mahfouz R, Behm FG, Raimondi SC, Relling MV, Patel A, Cheng C, Campana D, Wilkins D, Zhou X, Li J, Liu H, Pui CH, Evans WE, Naeve C, Wong L, Downing JR, 2002, Classification, subtype discovery, and prediction of outcome in paediatric acute lymphoblastic leukaemia by gene expression profiling. Cancer Cell 2:133–143 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 81 EP 90 DI 10.1007/s12253-010-9286-2 PG 10 ER PT J AU Agrawal, U Mishra, KA Salgia, P Verma, S Mohanty, KN Saxena, S AF Agrawal, Usha Mishra, K Ashwani Salgia, Payal Verma, Saurabh Mohanty, K Nayan Saxena, Sunita TI Role of Tumor Suppressor and Angiogenesis Markers in Prediction of Recurrence of Non Muscle Invasive Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non muscle invasive bladder cancer; Biomarkers; Predictive role; Survival analysis; Hazard ratio ID Non muscle invasive bladder cancer; Biomarkers; Predictive role; Survival analysis; Hazard ratio AB Non muscle invasive bladder cancers recur frequently and identification of biomarkers for predicting recurrence are necessary. The present study evaluated the individual and synergistic effects of tumor suppressor (p53/p21waf1) and angiogenesis [vascular endothelial growth factor (VEGF)/endoglin (CD105)] markers. The study included 90 cases of non muscle invasive bladder cancer. Cell spots were stained with primary antibodies and Flourescein isothiocyanate (FITC). Slides were observed under confocal laser scanning microscope for protein expression. The association between the markers individually and synergistically with recurrence were assessed by a χ2 and Fisher’s Exact test. Survival analysis was performed to predict recurrence and test for significant difference in recurrence free survival probability. Recurrence [overall:39(43.3%) and low grade(LG):26(54.2%)] was significant with p53 and VEGF expression and the profiles p53/VEGF, p53/CD105, VEGF/CD105, p53/p21/CD105, p53/VEGF/CD105 and all four were significantly associated with recurrence in both groups. In the multivariable model the [HR(95%CI),p: overall and LG] profiles p21/VEGF [2.195(1.052-4.582),0.036; 3.425(1.332-8.811),0.011], VEGF/CD105 [2.624(1.274-5.403),0.009 and 3.380(1.348-8.472),0.009], p53/p21/CD105 [2.000(0.993-4.027),0.052 and 2.539(1.047-6.157),0.039], p53/VEGF/CD105 [2.360(1.148-4.849),0.020 and 2.738(1.104-6.788),0.030], p21/VEGF/CD105 [2.611 (1.189-5.731),0.017 and 3.946(1.530-10.182),0.005] and all four [2.382(1.021-5.556),0.045 and 3.572(1.287-9.911),0.014] significantly predicted the recurrence along with significant log rank. In the pTa subset (n=33) the profiles p53/p21, p53/CD105, p21/VEGF, VEGF/CD105, p53/VEGF/CD105, p53/p21/CD105 and p21/VEGF/CD105, significantly predicted hazard for recurrence. The present study emphasizes an underlying association between tumor suppressor (p21waf1) and angiogenesis (VEGF/CD105) biomarkers. In addition combination profiles appeared to indicate an aggressive nature with high propensity for recurrence in LG and pTa tumours. C1 [Agrawal, Usha] Indian Council of Medical Research (ICMR), Institute of Pathology, Safdarjung Hospital Campus, 110029 New Delhi, India. [Mishra, K Ashwani] Indian Council of Medical Research (ICMR), Institute of Pathology, Safdarjung Hospital Campus, 110029 New Delhi, India. [Salgia, Payal] Indian Council of Medical Research (ICMR), Institute of Pathology, Safdarjung Hospital Campus, 110029 New Delhi, India. [Verma, Saurabh] Indian Council of Medical Research (ICMR), Institute of Pathology, Safdarjung Hospital Campus, 110029 New Delhi, India. [Mohanty, K Nayan] Safdarjung Hospital, Department of Urology, 110029 New Delhi, India. [Saxena, Sunita] Indian Council of Medical Research (ICMR), Institute of Pathology, Safdarjung Hospital Campus, 110029 New Delhi, India. RP Saxena, S (reprint author), Indian Council of Medical Research (ICMR), Institute of Pathology, 110029 New Delhi, India. EM sunita_saxena@yahoo.com CR Consolidated Report of Population Based Cancer Registries 2001–2004, Incidence and Distribution of Cancer, 2006, National Cancer Registry Programme. Indian Council of Medical Research, ICMR). Bangalore Mackenzie N, Torti FM, Faysal M, 1981, The natural history of superficial bladder tumors. Proc Am Soc Cancer Res 22:198 Oosterlinck W, Lobel B, Jakse G et al, 2002, European Association of Urology, EAU, working group on oncological urology. Guidelines on bladder cancer. Eur Urol 41:105–112 Mohanty NK, Nayak RL, Vasudeva P et al, 2008, Management of BCG non-responders with fixed dose intravesical gemcitabine in superficial transitional cell carcinoma of urinary bladder. Indian J Urol 24:44–47 Pfister C, Moore L, Allard P et al, 1999, Predictive value of cell cycle markers p53, MDM2, p21, and Ki-67 in superficial bladder tumor recurrence. Clin Cancer Res 5:4079–4084 Cordon-Cardov C, Zhang Z, Dalbagni G et al, 1997, Cooperative effects of p53 and pRb alterations in primary superficial bladder tumors. Cancer Res 57:1217–1221 Esrig D, Elmajian D, Groshen S et al, 1994, Accumulation of nuclear p53 and tumor progression in bladder cancer. N Engl J Med 331:1259–1264 Sarkis AS, Dalbagni G, Cordon-Cardo C et al, 1993, Nuclear overexpression of p53 protein in transitional cell bladder carcinoma: a marker for disease progression. J Natl Cancer Inst 85:53–59 Zeng YX, El-Deiry WS, 1996, Regulation of p21WAF1/CIP1 expression by p53-independent pathways. Oncogene 12:1557–1564 El-Deiry WS, Tokino T, Waldman T et al, 1995, Topological control of p21WAF1/CIP1 expression in normal and neoplastic tissues. Cancer Res 55:2910–2919 Crew JP, O’Brien T, Bradburn M et al, 1997, VEGF is a predictor of relapse and stage progression in superficial bladder cancer. Cancer Res 57:5281–5285 Duff SE, Li C, Garland JM et al, 2003, CD105 is important for angiogenesis: evidence and potential applications. FASEB J 17:984–992 Dangle PP, Wang WP, Mayerson J et al, 2008, Low grade papillary transitional cell carcinoma pelvic recurrence masquerading as high grade invasive carcinoma, ten years after radical cystectomy. World J Surg Oncol 6:103 Mian C, Lodde M, Comploj E et al, 2006, Multiprobe fluorescence in situ hybridisation: prognostic perspectives in superficial bladder cancer. J of Clin Pathol 59:984–987 Lu M,Wikman F, Orntoft TF et al, 2002, Impact of alterations affecting the p53 pathway in bladder cancer on clinical outcome, assessed by conventional and array-based methods. Clin Cancer Res 8:171–179 Datto MB, Li Y, Panus JF et al, 1995, Transforming growth factor beta induces the cyclin-dependent kinase inhibitor p21 through a p53- independent mechanism. Proc Natl Acad Sci USA 92:5545–5549 Stein JP, Ginsberg DA, Grossfeld GD et al, 1998, Effect of p21waf1/cip1 expression on tumour progression in bladder cancer. J Natl Cancer Inst 90:1072–1079 Lipponen P, Aaltomaa S, Eskelinen M et al, 1998, Expression of p21(waf1/cip1, protein in transitional cell bladder tumors and its prognostic value. Eur Urol 34:237–243 Korkolopoulou P, Konstantinidou AE, Thomas-Tsagli E et al, 2000, WAF1/p21 protein expression is an independent prognostic indicator in superficial and invasive bladder cancer. Appl Immunohistochem Mol Morphol 8:285–292 Feliksas J, Peter G, Kushima M et al, 2002, Value of p21 and p53 overexpression in predicting the outcome after systemic chemotherapy of Urothelial cancers. Acta Med Litu 9:93–99 Grossfield GD, Ginsberg DA, Stein JP, 1997, Thrombospondin-1 expression in bladder cancer with p53 alteration, tumor angiogenesis and tumor progression. J Natl Cancer Inst 89:219–227 Saad RS, Liu YL, Nathan G et al, 2004, Endoglin, CD105, and vascular endothelial growth factor as prognostic markers in colorectal cancer. Mod Pathol 17:197–203 Mineo TC, Ambrogi V, Baldi A et al, 2004, Prognostic impact of VEGF, CD31, CD34, and CD105 expression and tumour vessel invasion after radical surgery for IB–IIA non-small cell lung cancer. J Clin Pathol 57:591–597 El-Gohary YM, Silverman JF, Olson PR et al, 2007, Endoglin, CD105, and vascular endothelial growth factor as prognostic markers in prostatic adenocarcinoma. Am J Clin Pathol 127:572–579 Kumar S, Ghella A, Li C et al, 1999, Breast carcinoma: vascular density determined using CD105 antibody correlates with tumor prognosis. Cancer Res 59:856–861 Fonsatti E, Altomonte M, Nicotra MR et al, 2003, Endoglin, CD105): a powerful therapeutic target on tumor-associated angiogenetic blood vessels. Oncogene 22:6557–6563 Ellis LM, Hicklin DJ, 2008, VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer 8:579–591 Shariat SF, Tokunaga H, Zhou J et al, 2004, p53, p21, pRB, and p16 expression predict clinical outcome in cystectomy with bladder cancer. J Clin Oncol 22:1014–1024 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 91 EP 101 DI 10.1007/s12253-010-9287-1 PG 11 ER PT J AU Mammas, NI Sourvinos, G Zaravinos, A Spandidos, AD AF Mammas, N Ioannis Sourvinos, George Zaravinos, Apostolos Spandidos, A Demetrios TI Vaccination against Human Papilloma Virus (HPV): Epidemiological Evidence of HPV in Non-genital Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HPV; Non-genital cancer; Oesophageal; Laryngeal; Oropharyngeal; Lung; Urothelial; Breast; Colon; Vaccination; Childhood ID HPV; Non-genital cancer; Oesophageal; Laryngeal; Oropharyngeal; Lung; Urothelial; Breast; Colon; Vaccination; Childhood AB Recently, the vaccine against human papillomavirus (HPV) was introduced in the national vaccination programmes of several countries worldwide. The established association between HPV and the progression of cervical neoplasia provides evidence of the expected protection of the vaccine against cervical cancer. During the last two decades several studies have also examined the possible involvement of HPV in non-genital cancers and have proposed the presence of HPV in oesophageal, laryngeal, oropharyngeal, lung, urothelial, breast and colon cancers. The possible involvement of HPV in these types of cancer would necessitate the introduction of the vaccine in both boys and girls. However, the role of HPV in the pathogenesis of these types of cancer has yet to be proven. Moreover, the controversial evidence of the possible impact of the vaccination against HPV in the prevention of non-genital cancers needs to be further evaluated. In this review, we present an overview of the existing epidemiological evidence regarding the detection of HPV in non-genital cancers. C1 [Mammas, N Ioannis] University of Crete, Medical School, Department of VirologyHeraklion, Greece. [Sourvinos, George] University of Crete, Medical School, Department of VirologyHeraklion, Greece. [Zaravinos, Apostolos] University of Crete, Medical School, Department of VirologyHeraklion, Greece. [Spandidos, A Demetrios] University of Crete, Medical School, Department of VirologyHeraklion, Greece. RP Spandidos, AD (reprint author), University of Crete, Medical School, Department of Virology, Heraklion, Greece. EM spandidos@spandidos.gr CR zur Hausen H, 2002, Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer 2:342–350 Sanclemente G, Gill DK, 2002, Human papillomavirus molecular biology and pathogenesis. J Eur Acad Dermatol Venereol 16:231–240 Peto J, Gilham C, Deacon J et al, 2004, Cervical HPV infection and neoplasia in a large population-based prospective study: the Manchester cohort. Br J Cancer 91:942–953 Munoz N, Bosch FX, de Sanjose S et al, 2003, Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med 348:518–527 Clifford GM, Smith JS, Plummer M et al, 2003, Human papillomavirus types in invasive cervical cancer worldwide: a meta-analysis. Br J Cancer 88:63–73 Fehrmann F, Laimins LA, 2003, Human papillomaviruses: targeting differentiating epithelial cells for malignant transformation. Oncogene 22:5201–5207 Mammas I, Sourvinos G, Giannoudis A, Spandidos DA, 2008, Human papilloma virus, HPV, and host cellular interactions. Pathol Oncol Res 14:345–354 Zaravinos A, Mammas I, Sourvinos G, Spandidos DA, 2009, Detection methods of human papilloma virus, HPV). Int J Biol Markers 24(4):215–222 Syrjanen KJ, 2002, HPV infections and oesophageal cancer. J Clin Pathol 55:721–728 Sur M, Cooper K, 1998, The role of the human papilloma virus in esophageal cancer. Pathology 30:348–354 Bohn OL, Navarro L, Saldivar J, Sanchez-Sosa S, 2008, Identification of human papillomavirus in esophageal squamous papillomas. World J Gastroenterol 14:7107–7111 van Zeeburg HJ, Snijders PJ, Wu T et al, 2008, Clinical and molecular characteristics of squamous cell carcinomas from Fanconi anemia patients. J Natl Cancer Inst 100:1649–1653 Rai N, Jenkins GJ, McAdam E et al, 2008, Human papillomavirus infection in Barrett’s oesophagus in the UK: an infrequent event. J Clin Virol 43:250–252 Bognar G, Imdahl A, Ledniczky G, Ondrejka P, 2008, Possible role of human papilloma virus infection in response to neoadjuvant therapy in patients with esophageal cancer. Hepatogastroenterology 55:93–97 Koh JS, Lee SS, Baek HJ, Kim YI, 2008, No association of high-risk human papillomavirus with esophageal squamous cell carcinomas among Koreans, as determined by polymerase chain reaction. Dis Esophagus 21:114–117 Lu XM, Monnier-Benoit S, Mo LZ, Xu SY et al, 2008, Human papillomavirus in esophageal squamous cell carcinoma of the high-risk Kazakh ethnic group in Xinjiang, China. Eur J Surg Oncol 34:765–770 Matsha T, Donninger H, Erasmus RT et al, 2007, Expression of p53 and its homolog, p73, in HPV DNA positive oesophageal squamous cell carcinomas. Virology 369:182–190 Shuyama K, Castillo A, Aguayo F et al, 2007, Human papillomavirus in high- and low-risk areas of oesophageal squamous cell carcinoma in China. Br J Cancer 96:1554–1559 Pantelis A, Pantelis D, Ruemmele P et al, 2007, p53 Codon 72 polymorphism, loss of heterozygosity and high-risk human papillomavirus infection in a low-incidence German esophageal squamous cell carcinoma patient cohort. Oncol Rep 17:1243–1248 Far AE, Aghakhani A, Hamkar R et al, 2007, Frequency of human papillomavirus infection in oesophageal squamous cell carcinoma in Iranian patients. Scand J Infect Dis 39:58–62 Qi ZL, Huo X, Xu XJ et al, 2006, Relationship between HPV16/ 18 E6 and 53, 21WAF1, MDM2, Ki67 and cyclin D1 expression in esophageal squamous cell carcinoma: comparative study by using tissue microarray technology. Exp Oncol 28:235–240 Castillo A, Aguayo F, Koriyama C et al, 2006, Human papillomavirus in esophageal squamous cell carcinoma in Colombia and Chile. World J Gastroenterol 12:6188–6192 Souto Damin AP, Guedes Frazzon AP, de Carvalho DD et al, 2006, Detection of human papillomavirus DNA in squamous cell carcinoma of the esophagus by auto-nested PCR. Dis Esophagus 19:64–68 Dreilich M, Bergqvist M, Moberg M et al, 2006, High-risk human papilloma virus, HPV, and survival in patients with esophageal carcinoma: a pilot study. BMC Cancer 6:94 Gao GF, Roth MJ, Wei WQ et al, 2006, No association between HPV infection and the neoplastic progression of esophageal squamous cell carcinoma: result from a cross-sectional study in a high-risk region of China. Int J Cancer 119:1354–1359 Lyronis ID, Baritaki S, Bizakis I et al, 2005, Evaluation of the prevalence of human papillomavirus and Epstein-Barr virus in esophageal squamous cell carcinomas. Int J Biol Markers 20:5–10 White RE, Mungatana C, Mutuma G et al, 2005, Absence of human papillomavirus in esophageal carcinomas from southwestern Kenya. Dis Esophagus 18:28–30 Farhadi M, Tahmasebi Z, Merat S et al, 2005, Human papillomavirus in squamous cell carcinoma of esophagus in a high-risk population. World J Gastroenterol 11:1200–1203 Bahnassy AA, Zekri AR, Abdallah S et al, 2005, Human papillomavirus infection in Egyptian esophageal carcinoma: correlation with p53, p21, mdm2, C-erbB2 and impact on survival. Pathol Int 55:53–62 Katiyar S, Hedau S, Jain N et al, 2005, p53 gene mutation and human papillomavirus, HPV, infection in esophageal carcinoma from three different endemic geographic regions of India. Cancer Lett 218:69–79 Lu XM, Zhang YM, Lin RY et al, 2004, p53 polymorphism in human papillomavirus-associated Kazakh’s esophageal cancer in Xinjiang, China. World J Gastroenterol 10:2775–2778 Acevedo-Nuno E, Gonzalez-Ojeda A, Vazquez-Camacho G et al, 2004, Human papillomavirus DNA and protein in tissue samples of oesophageal cancer, Barrett’s oesophagus and oesophagitis. Anticancer Res 24:1319–1323 de Villiers EM, Gunst K, Stein H, Scherubl H, 2004, Esophageal squamous cell cancer in patients with head and neck cancer: prevalence of human papillomavirus DNA sequences. Int J Cancer 109:253–258 Weston AC, Prolla JC, 2003, Association between esophageal squamous cell carcinoma and human papillomavirus detected by Hybrid Capture II assay. Dis Esophagus 16:224–228 Kamath AM, Wu TT, Heitmiller R et al, 2000, Investigation of the association of esophageal carcinoma with human papillomaviruses. Dis Esophagus 13:122–124 Awerkiew S, Bollschweiler E, Metzger R et al, 2003, Esophageal cancer in Germany is associated with Epstein-Barr-virus but not with papillomaviruses. Med Microbiol Immunol 192:137–140 Zhou XB, Guo M, Quan LP et al, 2003, Detection of human papillomavirus in Chinese esophageal squamous cell carcinoma and its adjacent normal epithelium. World J Gastroenterol 9:1170–1173 Li T, Lu ZM, Guo M et al, 2002, p53 codon 72 polymorphism, C/G, and the risk of human papillomavirus-associated carcinomas in China. Cancer 95:2571–2576 Shen ZY, Hu SP, Lu LC et al, 2002, Detection of human papillomavirus in esophageal carcinoma. J Med Virol 68:412–416 Hasegawa M, Ohoka I, Yamazaki K et al, 2002, Expression of p21/ WAF-1, status of apoptosis and p53 mutation in esophageal squamous cell carcinoma with HPVinfection. Pathol Int 52:442–450 Matsha T, Erasmus R, Kafuko AB et al, 2002, Human papillomavirus associated with oesophageal cancer. J Clin Pathol 55:587–590 Astori G, Merluzzi S, Arzese A et al, 2001, Detection of human papillomavirus DNA and p53 gene mutations in esophageal cancer samples and adjacent normal mucosa. Digestion 64:9–14 Chang F, Syrjanen S, Shen Q et al, 2000, Evaluation of HPV,CMV, HSV and EBV in esophageal squamous cell carcinomas from a high-incidence area of China. Anticancer Res 20:3935–3940 Lambot MA, Haot J, Peny MO et al, 2000, Evaluation of the role of human papillomavirus in oesophageal squamous cell carcinoma in Belgium. Acta Gastroenterol Belg 63:154–156 Talamini G, Capelli P, Zamboni G et al, 2000, Alcohol, smoking and papillomavirus infection as risk factors for esophageal squamous-cell papilloma and esophageal squamous-cell carcinoma in Italy. Int J Cancer 86:874–878 Chang F, Syrjanen S, Shen Q et al, 2000, Human papillomavirus involvement in esophageal carcinogenesis in the high-incidence area of China. A study of 700 cases by screening and typespecific in situ hybridization. Scand J Gastroenterol 35:123–130 de Villiers EM, Lavergne D, Chang F et al, 1999, An interlaboratory study to determine the presence of human papillomavirus DNA in esophageal carcinoma from China. Int J Cancer 81:225–228 Lavergne D, de Villiers EM, 1999, Papillomavirus in esophageal papillomas and carcinomas. Int J Cancer 80:681–684 Takahashi A, Ogoshi S, Ono H, Ishikawa T et al, 1998, Highrisk human papillomavirus infection and overexpression of p53 protein in squamous cell carcinoma of the esophagus from Japan. Dis Esophagus 11:162–167 Khurshid A, Kazuya N, Hanae I, Manabu I, 1998, Infection of human papillomavirus, HPV, and Epstein-Barr virus, EBV, and p53 expression in human esophageal carcinoma. J Pak Med Assoc 48:138–142 Morgan RJ, Perry AC, Newcomb PV et al, 1997, Human papillomavirus and oesophageal squamous cell carcinoma in the UK. Eur J Surg Oncol 23:513–517 Kok TC, Nooter K, Tjong-A-Hung SP et al, 1997, No evidence of known types of human papillomavirus in squamous cell cancer of the oesophagus in a low-risk area. Rotterdam Oesophageal Tumour Study Group. Eur J Cancer 33:1865–1868 He D, Zhang DK, Lam KY et al, 1997, Prevalence of HPV infection in esophageal squamous cell carcinoma in Chinese patients and its relationship to the p53 gene mutation. Int J Cancer 72:959–964 Lam KY, He D, Ma L et al, 1997, Presence of human papillomavirus in esophageal squamous cell carcinomas of Hong Kong Chinese and its relationship with p53 gene mutation. Hum Pathol 28:657–663 Miller BA, Davidson M, Myerson D et al, 1997, Human papillomavirus type 16 DNA in esophageal carcinomas from Alaska Natives. Int J Cancer 71:222 Saegusa M, Hashimura M, Takano Y et al, 1997, Absence of human papillomavirus genomic sequences detected by the polymerase chain reaction in oesophageal and gastric carcinomas in Japan. Mol Pathol 50:101–104 Mizobuchi S, Sakamoto H, Tachimori Y et al, 1997, Absence of human papillomavirus-16 and -18 DNA and Epstein-Barr virus DNA in esophageal squamous cell carcinoma. Jpn J Clin Oncol 27:1–5 Morgan RJ, Perry AC, Newcomb PV et al, 1997, Investigation of oesophageal adenocarcinoma for viral genomic sequences. Eur J Surg Oncol 23:24–29 Turner JR, Shen LH, Crum CP et al, 1997, Low prevalence of human papillomavirus infection in esophageal squamous cell carcinomas from North America: analysis by a highly sensitive and specific polymerase chain reaction-based approach. Hum Pathol 28:174–178 Chang F, Syrjanen S, Wang L et al, 1997, p53 overexpression and human papillomavirus, HPV, infection in oesophageal squamous cell carcinomas derived from a high-incidence area in China. Anticancer Res 17:709–715 Suzuk L, Noffsinger AE, Hui YZ, Fenoglio-Preiser CM, 1996, Detection of human papillomavirus in esophageal squamous cell carcinoma. Cancer 78:704–710 Shibagaki I, Tanaka H, Shimada Y et al, 1995, p53 mutation, murine double minute 2 amplification, and human papillomavirus infection are frequently involved but not associated with each other in esophageal squamous cell carcinoma. Clin Cancer Res 1:769–773 Smits HL, Tjong-A-Hung SP, ter Schegget J et al, 1995, Absence of human papillomavirus DNA from esophageal carcinoma as determined by multiple broad spectrum polymerase chain reactions. J Med Virol 46:213–215 Cooper K, Taylor L, Govind S, 1995, Human papillomavirus DNA in oesophageal carcinomas in South Africa. J Pathol 175:273–277 Chen B, Yin H, Dhurandhar N, 1994,, 1994, Detection of human papillomavirus DNA in esophageal squamous cell carcinomas by the polymerase chain reaction using general consensus primers. Hum Pathol 25:920–923 Si HX, Tsao SW, Poon CS et al, 2005, Physical status of HPV-16 in esophageal squamous cell carcinoma. J Clin Virol 32:19–23 Yao PF, Li GC, Li J et al, 2006, Evidence of human papilloma virus infection and its epidemiology in esophageal squamous cell carcinoma. World J Gastroenterol 12:1352–1355 Sugar J, Vereczkey I, Toth J, 1996, Some etio-pathogenetic factors in laryngeal carcinogenesis. J Environ Pathol Toxicol Oncol 15:195–199 Baumann JL, Cohen S, Evjen AN et al, 2009, Human papillomavirus in early laryngeal carcinoma. Laryngoscope 119:1531–1537 Morshed K, Polz-Dacewicz M, Szymanski M, Polz D, 2008, Short-fragment PCR assay for highly sensitive broad-spectrum detection of human papillomaviruses in laryngeal squamous cell carcinoma and normal mucosa: clinico-pathological evaluation. Eur Arch Otorhinolaryngol 265:S89–S96 Szladek G, Juhasz A, Kardos G et al, 2005, High co-prevalence of genogroup 1 TT virus and human papillomavirus is associated with poor clinical outcome of laryngeal carcinoma. J Clin Pathol 58:402–405 Almadori G, Cadoni G, Cattani P et al, 2001, Human papillomavirus infection and epidermal growth factor receptor expression in primary laryngeal squamous cell carcinoma. Clin Cancer Res 7:3988–3993 Smith EM, Summersgill KF, Allen J et al, 2000, Human papillomavirus and risk of laryngeal cancer. Ann Otol Rhinol Laryngol 109:1069–1076 Lindeberg H, Krogdahl A, 1999, Laryngeal cancer and human papillomavirus: HPV is absent in the majority of laryngeal carcinomas. Cancer Lett 146:9–13 Cerovac Z, Sarcevic B, Kralj Z, Ban J, 1996, Detection of human papillomavirus, HPV, type 6, 16 and 18 in head and neck squamous cell carcinomas by in situ hybridization. Neoplasma 43:185–194 Clayman GL, Stewart MG, Weber RS et al, 1994, Human papillomavirus in laryngeal and hypopharyngeal carcinomas. Relationship to survival. Arch Otolaryngol Head Neck Surg 120:743–748 Wang H, Lin YC, Kang XS et al, 1991, Detection of human papilloma virus, HPV, in laryngeal carcinoma tissue. Chin Med J, Engl, 104:523–525 Reidy PM, Dedo HH, Rabah R et al, 2004, Integration of human papillomavirus type 11 in recurrent respiratory papillomaassociated cancer. Laryngoscope 114:1906–1909 Turazza E, Lapena A, Sprovieri O et al, 1997, Low-risk human papillomavirus types 6 and 11 associated with carcinomas of the genital and upper aero-digestive tract. Acta Obstet Gynecol Scand 76:271–276 Lee LA, Cheng AJ, Fang TJ et al, 2008, High incidence of malignant transformation of laryngeal papilloma in Taiwan. Laryngoscope 118:50–55 Syrjanen S, 2004, HPV infections and tonsillar carcinoma. J Clin Pathol 57:449–455 Nasman A, Attner P, Hammarstedt L et al, 2009, Incidence of human papillomavirus, HPV, positive tonsillar carcinoma in Stockholm, Sweden: an epidemic of viral-induced carcinoma? Int J Cancer 125:362–366 Romanitan M, Nasman A, Ramqvist T et al, 2008, Human papillomavirus frequency in oral and oropharyngeal cancer in Greece. Anticancer Res 28:2077–2080 Hafkamp HC, Manni JJ, Haesevoets A et al, 2008, Marked differences in survival rate between smokers and nonsmokers with HPV 16-associated tonsillar carcinomas. Int J Cancer 122:2656– 2664 Kuo KT, Hsiao CH, Lin CH et al, 2008, The biomarkers of human papillomavirus infection in tonsillar squamous cell carcinoma-molecular basis and predicting favorable outcome. Mod Pathol 21:376–386 Westra WH, Taube JM, Poeta ML et al, 2008, Inverse relationship between human papillomavirus-16 infection and disruptive p53 gene mutations in squamous cell carcinoma of the head and neck. Clin Cancer Res 14:366–369 Charfi L, Jouffroy T, de Cremoux P et al, 2008, Two types of squamous cell carcinoma of the palatine tonsil characterized by distinct etiology, molecular features and outcome. Cancer Lett 260:72–78 Pintos J, Black MJ, Sadeghi N et al, 2008, Human papillomavirus infection and oral cancer: a case-control study in Montreal, Canada. Oral Oncol 44:242–250 Li W, Tran N, Lee SC et al, 2007, New evidence for geographic variation in the role of human papillomavirus in tonsillar carcinogenesis. Pathology 39:217–222 Chien CY, Su CY, Fang FM et al, 2008, Lower prevalence but favorable survival for human papillomavirus-related squamous cell carcinoma of tonsil in Taiwan. Oral Oncol 44:174–179 Mellin Dahlstrand H, Lindquist D, Bjornestal L et al, 2005, P16, INK4a, correlates to human papillomavirus presence, response to radiotherapy and clinical outcome in tonsillar carcinoma. Anticancer Res 25:4375–4383 Begum S, Cao D, Gillison M et al, 2005, Tissue distribution of human papillomavirus 16 DNA integration in patients with tonsillar carcinoma. Clin Cancer Res 11:5694–5699 Hoffmann M, Gottschlich S, Gorogh T et al, 2005, Human papillomaviruses in lymph node neck metastases of head and neck cancers. Acta Otolaryngol 125:415–421 Venuti A, Badaracco G, Rizzo C et al, 2004, Presence of HPV in head and neck tumours: high prevalence in tonsillar localization. J Exp Clin Cancer Res 23:561–566 Azzimonti B, Pagano M, Mondini M et al, 2004, Altered patterns of the interferon-inducible gene IFI16 expression in head and neck squamous cell carcinoma: immunohistochemical study including correlation with retinoblastoma protein, human papillomavirus infection and proliferation index. Histopathology 45:560–572 Li W, Thompson CH, Cossart YE et al, 2004, The expression of key cell cycle markers and presence of human papillomavirus in squamous cell carcinoma of the tonsil. Head Neck 26:1–9 El-Mofty SK, Lu DW, 2003, Prevalence of human papillomavirus type 16 DNA in squamous cell carcinoma of the palatine tonsil, and not the oral cavity, in young patients: a distinct clinicopathologic and molecular disease entity. Am J Surg Pathol 27:1463–1470 Hafkamp HC, Speel EJ, Haesevoets A et al, 2003, A subset of head and neck squamous cell carcinomas exhibits integration of HPV 16/18 DNA and overexpression of p16INK4A and p53 in the absence of mutations in p53 exons 5–8. Int J Cancer 107:394–400 Mellin H, Friesland S, Auer G et al, 2003, Human papillomavirus and DNA ploidy in tonsillar cancer–correlation to prognosis. Anticancer Res 23:2821–2828 Koskinen WJ, Chen RW, Leivo I et al, 2003, Prevalence and physical status of human papillomavirus in squamous cell carcinomas of the head and neck. Int J Cancer 107:401–406 Ryerson AB, Peters ES, Coughlin SS et al, 2008, Burden of potentially human papillomavirus-associated cancers of the oropharynx and oral cavity in the US, 1998–2003. Cancer 113:2901–2909 Dahlstrand H, Nasman A, Romanitan M et al, 2008, Human papillomavirus accounts both for increased incidence and better prognosis in tonsillar cancer. Anticancer Res 28:1133–1138 Kim SH, Koo BS, Kang S et al, 2007, HPV integration begins in the tonsillar crypt and leads to the alteration of p16, EGFR and c-myc during tumor formation. Int J Cancer 120:1418–1425 Fischer CA, Zlobec I, Green E et al, 2010, Is the improved prognosis of p16 positive oropharyngeal squamous cell carcinoma dependent of the treatment modality? Int J Cancer 126:1256–1262 CohenMA, Basha SR, Reichenbach DK et al, 2008, Increased viral load correlates with improved survival in HPV-16-associated tonsil carcinoma patients. Acta Otolaryngol 128:583–589 D’Souza G, Agrawal Y, Halpem J et al, 2009, Oral sexual behaviours associated with prevalent oral human papillomavirus infection. J Infect Dis 199:1263–1269 Mammas IN, Sourvinos G, Spandidos DA, 2009, Human papilloma virus, HPV, infection in children and adolescents. Eur J Pediatr 168:267–273 Chen YC, Chen JH, Richard K et al, 2004, Lung adenocarcinoma and human papillomavirus infection. Cancer 101:1428–1436 Syrjanen KJ, 2002, HPV infections and lung cancer. J Clin Pathol 55:885–891 Yu Y, Yang A, Hu S, Yan H, 2009, Correlation of HPV-16/18 infection of human papillomavirus with lung squamous cell carcinomas in Western China. Oncol Rep 21:1627–1632 Castillo A, Aguayo F, Koriyama C et al, 2006, Human papillomavirus in lung carcinomas among three Latin American countries. Oncol Rep 15:883–888 Wang Y, Wang A, Jiang R et al, 2008, Human papillomavirus type 16 and 18 infection is associated with lung cancer patients from the central part of China. Oncol Rep 20:333–339 Aguayo F, Castillo A, Koriyama C et al, 2007, Human papillomavirus-16 is integrated in lung carcinomas: a study in Chile. Br J Cancer 97:85–91 Giuliani L, Jaxmar T, Casadio C et al, 2007, Detection of oncogenic viruses SV40, BKV, JCV, HCMV, HPV and p53 codon 72 polymorphism in lung carcinoma. Lung Cancer 57:273–281 Park MS, Chang YS, Shin JH et al, 2007, The prevalence of human papillomavirus infection in Korean non-small cell lung cancer patients. Yonsei Med J 48:69–77 Nadji SA, Mokhtari-Azad T, Mahmoodi M et al, 2007, Relationship between lung cancer and human papillomavirus in north of Iran, Mazandaran province. Cancer Lett 248:41–46 Ciotti M, Giuliani L, Ambrogi V et al, 2006, Detection and expression of human papillomavirus oncogenes in non-small cell lung cancer. Oncol Rep 16:183–189 Jain N, Singh V, Hedau S et al, 2005, Infection of human papillomavirus type 18 and p53 codon 72 polymorphism in lung cancer patients from India. Chest 128:3999–4007 Coissard CJ, Besson G, Polette MC et al, 2005, Prevalence of human papillomaviruses in lung carcinomas: a study of 218 cases. Mod Pathol 18:1606–1609 Brouchet L, Valmary S, Dahan M et al, 2005, Detection of oncogenic virus genomes and gene products in lung carcinoma. Br J Cancer 92:743–746 Zafer E, Ergun MA, Alver G et al, 2004, Detection and typing of human papillomavirus in non-small cell lung cancer. Respiration 71:88–90 FeiY,Yang J,HsiehWC et al, 2006)Different human papillomavirus 16/18 infection in Chinese non-small cell lung cancer patients living in Wuhan, China. Jpn J Clin Oncol 36:274–279 Miasko A, NiklinskaW,Niklinski J et al, 2001, Detection of human papillomavirus in non-small cell lung carcinoma by polymerase chain reaction. Folia Histochem Cytobiol 39:127–128 Miyagi J, Kinjo T, Tsuhako K et al, 2001, Extremely high Langerhans cell infiltration contributes to the favourable prognosis of HPV-infected squamous cell carcinoma and adenocarcinoma of the lung. Histopathology 38:355–367 Kaya H, Kotiloglu E, Inanli S et al, 2001, Prevalence of human papillomavirus, HPV, DNA in larynx and lung carcinomas. Pathologica 93:531–534 Cheng YW, Chiou HL, Sheu GT et al, 2001, The association of human papillomavirus 16/18 infection with lung cancer among nonsmoking Taiwanese women. Cancer Res 61:2799–2803 Gorgoulis VG, Zacharatos P, Kotsinas A et al, 1999, Human papilloma virus, HPV, is possibly involved in laryngeal but not in lung carcinogenesis. Hum Pathol 30:274–283 Tsuhako K, Nakazato I, Hirayasu T et al, 1998, Human papillomavirus DNA in adenosquamous carcinoma of the lung. J Clin Pathol 51:741–749 Papadopoulou K, Labropoulou V, Davaris P et al, 1998, Detection of human papillomaviruses in squamous cell carcinomas of the lung. Virchows Arch 433:49–54 Bohlmeyer T, Le TN, Shroyer AL et al, 1998, Detection of human papillomavirus in squamous cell carcinomas of the lung by polymerase chain reaction. Am J Respir Cell Mol Biol 18:265–269 Soini Y, Nuorva K, Kamel D et al, 1996, Presence of human papillomavirus DNA and abnormal p53 protein accumulation in lung carcinoma. Thorax 51:887–893 Thomas P, De Lamballerie X, Garbe L et al, 1995, Detection of human papillomavirus DNA in primary lung carcinoma by nested polymerase chain reaction. Cell Mol Biol 41:1093–1097 Li Q, Hu K, Pan X et al, 1995, Detection of human papillomavirus types 16, 18 DNA related sequences in bronchogenic carcinoma by polymerase chain reaction. Chin Med J, Engl, 108:610–614 Kinoshita I, Dosaka-Akita H, Shindoh M et al, 1995, Human papillomavirus type 18 DNA and E6-E7 mRNA are detected in squamous cell carcinoma and adenocarcinoma of the lung. Br J Cancer 71:344–349 Szabo I, Sepp R, Nakamoto K et al, 1994, Human papillomavirus not found in squamous and large cell lung carcinomas by polymerase chain reaction. Cancer 73:2740–2744 Yousem SA, Ohori NP, Sonmez-Alpan E, 1992, Occurrence of human papillomavirus DNA in primary lung neoplasms. Cancer 69:693–697 Bejui-Thivolet F, Liagre N, Chignol MC et al, 1990, Detection of human papillomavirus DNA in squamous bronchial metaplasia and squamous cell carcinomas of the lung by in situ hybridization using biotinylated probes in paraffin-embedded specimens. Hum Pathol 21:111–116 Cheng YW, Wu MF, Wang J et al, 2007, Human papillomavirus 16/18 E6 oncoprotein is expressed in lung cancer and related with p53 inactivation. Cancer Res 67:10686– 10693 Cheng YW, Wu TC, Chen CY et al, 2008, Human telomerase reverse transcriptase activated by E6 oncoprotein is required for human papillomavirus-16/18-infected lung tumorigenesis. Clin Cancer Res 14:7173–7179 Hsu NY, Cheng YW, Chan IP et al, 2009, Association between expression of human papillomavirus 16/18 E6 oncoprotein and survival in patients with stage I non-small cell lung cancer. Oncol Rep 21:81–87 Nadji SA, Mahmoodi M, Ziaee AA et al, 2007, An increased lung cancer risk associated with codon 72 polymorphism in the TP53 gene and human papillomavirus infection in Mazandaran province, Iran. Lung Cancer 56:145–151 Buyru N, Altinisik J, Isin M, Dalay N, 2008, p53 codon 72 polymorphism and HPV status in lung cancer. Med Sci Monit 14:493–497 Abol-Enein H, 2008, Infection: is it a cause of bladder cancer? Scand J Urol Nephrol Suppl 218:79–84 Lopez-Beltran A, Escudero AL, 1997, Human papillomavirus and bladder cancer. Biomed Pharmacother 51:252–257 Wiwanitkit V, 2005, Urinary bladder carcinoma and human papilloma virus infection, an appraisal of risk. Asian Pac J Cancer Prev 6:217–218 Aggarwal S, Arora VK, Gupta S et al, 2009, Koilocytosis: correlations with high-risk HPV and its comparison on tissue sections and cytology, urothelial carcinoma. Diagn Cytopathol 37:174–177 Badawi H, Ahmed H, Ismail A et al, 2008, Role of human papillomavirus types 16, 18, and 52 in recurrent cystitis and urinary bladder cancer among Egyptian patients. Medscape J Med 10:232 Hodges A, Talley L, Gokden N, 2006, Human Papillomavirus DNA and P16INK4A are not detected in renal tumors with immunohistochemistry and signal-amplified in situ hybridization in paraffin-embedded tissue. Appl Immunohistochem Mol Morphol 14:432–435 Helal Tel A, Fadel MT, El-Sayed NK, 2006, Human papilloma virus and p53 expression in bladder cancer in Egypt: relationship to schistosomiasis and clinicopathologic factors. Pathol Oncol Res 12:173–178 Barghi MR, Hajimohammadmehdiarbab A, Moghaddam SM, Kazemi B, 2005, Correlation between human papillomavirus infection and bladder transitional cell carcinoma. BMC Infect Dis 5:102 Youshya S, Purdie K, Breuer J et al, 2005, Does human papillomavirus play a role in the development of bladder transitional cell carcinoma? A comparison of PCR and immunohistochemical analysis. J Clin Pathol 58:207–210 Khaled HM, Bahnassi AA, Zekri AR et al, 2003, Correlation between p53 mutations and HPV in bilharzial bladder cancer. Urol Oncol 21:334–341 Fioriti D, Pietropaolo V, Dal Forno S et al, 2003, Urothelial bladder carcinoma and viral infections: different association with human polyomaviruses and papillomaviruses. Int J Immunopathol Pharmacol 16:283–288 Soulitzis N, Sourvinos G, Dokianakis DN, Spandidos DA, 2002, p53 codon 72 polymorphism and its association with bladder cancer. Cancer Lett 179:175–183 Khaled HM, Raafat A, Mokhtar N et al, 2001, Human papilloma virus infection and overexpression of p53 protein in bilharzial bladder cancer. Tumori 87:256–261 Westenend PJ, Stoop JA, Hendriks JG, 2001, Human papillomaviruses 6/11, 16/18 and 31/33/51 are not associated with squamous cell carcinoma of the urinary bladder. BJU Int 88:198–201 Sur M, Cooper K, Allard U, 2001, Investigation of human papillomavirus in transitional cell carcinomas of the urinary bladder in South Africa. Pathology 33:17–20 Simoneau M, LaRue H, Fradet Y, 1999, Low frequency of human papillomavirus infection in initial papillary bladder tumors. Urol Res 27:180–184 De Gaetani C, Ferrari G, Righi E et al, 1999, Detection of human papillomavirus DNA in urinary bladder carcinoma by in situ hybridisation. J Clin Pathol 52:103–106 Tekin MI, Tuncer S, Aki FT et al, 1999, Human papillomavirus associated with bladder carcinoma? Analysis by polymerase chain reaction. Int J Urol 6:184–186 Gazzaniga P, Vercillo R, Gradilone A et al, 1998, Prevalence of papillomavirus, Epstein-Barr virus, cytomegalovirus, and herpes simplex virus type 2 in urinary bladder cancer. J Med Virol 55:262–267 Chan KW, Wong KY, Srivastava G, 1997, Prevalence of six types of human papillomavirus in inverted papilloma and papillary transitional cell carcinoma of the bladder: an evaluation by polymerase chain reaction. J Clin Pathol 50:1018–1021 Aynaud O, Tranbaloc P, Orth G, 1998, Lack of evidence for a role of human papillomaviruses in transitional cell carcinoma of the bladder. J Urol 159:86–89 Lu QL, Lalani E-N, Abel P, 1997, Human papillomavirus 16 and 18 infection is absent in urinary bladder carcinomas. Eur Urol 31:428–432 Boucher NR, Scholefield JH, Anderson JB, 1996, The aetiological significance of human papillomavirus in bladder cancer. Br J Urol 78:866–869 Lopez-Beltran A, Escudero AL, Carrasco-Aznar JC, Vicioso- Recio L, 1996, Human papillomavirus infection and transitional cell carcinoma of the bladder. Immunohistochemistry and in situ hybridization. Pathol Res Pract 192:154–159 Mvula M, Iwasaka T, Iguchi A et al, 1996, Do human papillomaviruses have a role in the pathogenesis of bladder carcinoma? J Urol 155:471–474 Tenti P, Zappatore R, Romagnoli S et al, 1996, p53 overexpression and human papillomavirus infection in transitional cell carcinoma of the urinary bladder: correlation with histological parameters. J Pathol 178:65–70 Lopez-Beltran A, Escudero AL, Vicioso L et al, 1996, Human papillomavirus DNA as a factor determining the survival of bladder cancer patients. Br J Cancer 73:124–127 Smetana Z, Keller T, Leventon-Kriss S et al, 1995, Presence of human papilloma virus in transitional cell carcinoma in Jewish population in Israel. Cell Mol Biol 41:1017–1023 Kim KH, Kim YS, 1995, Analysis of p53 tumor suppressor gene mutations and human papillomavirus infection in human bladder cancers. Yonsei Med J 36:322–331 Gopalkrishna V, Srivastava AN, Hedau S et al, 1995, Detection of human papillomavirus DNA sequences in cancer of the urinary bladder by in situ hybridisation and polymerase chain reaction. Genitourin Med 71:231–233 Sano T, Sakurai S, Fukuda T, Nakajima T, 1995, Unsuccessful effort to detect human papillomavirus DNA in urinary bladder cancers by the polymerase chain reaction and in situ hybridization. Pathol Int 45:506–512 Lopez-Beltran A, Munoz E, 1995, Transitional cell carcinoma of the bladder: low incidence of human papillomavirus DNA detected by the polymerase chain reaction and in situ hybridization. Histopathology 26:565–569 Kamel D, Paakko P, Pollanen R et al, 1995, Human papillomavirus DNA and abnormal p53 expression in carcinoma of the urinary bladder. APMIS 103:331–338 LaRue H, Simoneau M, Fradet Y, 1995, Human papillomavirus in transitional cell carcinoma of the urinary bladder. Clin Cancer Res 1:435–440 Noel JC, Thiry L, Verhest A et al, 1994, Transitional cell carcinoma of the bladder: evaluation of the role of human papillomaviruses. Urology 44:671–675 Maloney KE, Wiener JS, Walther PJ, 1994, Oncogenic human papillomaviruses are rarely associated with squamous cell carcinoma of the bladder: evaluation by differential polymerase chain reaction. J Urol 151:360–364 Agliano AM, Gradilone A, Gazzaniga P et al, 1994, High frequency of human papillomavirus detection in urinary bladder cancer. Urol Int 53:125–129 Furihata M, Inoue K, Ohtsuki Y et al, 1993, High-risk human papillomavirus infections and overexpression of p53 protein as prognostic indicators in transitional cell carcinoma of the urinary bladder. Cancer Res 53:4823–4827 Saltzstein DR, Orihuela E, Kocurek JN et al, 1993, Failure of the polymerase chain reaction, PCR, to detect human papilloma virus, HPV, in transitional cell carcinoma of the bladder. Anticancer Res 13:423–425 Anwar K, Naiki H, Nakakuki K, Inuzuka M, 1992, High frequency of human papillomavirus infection in carcinoma of the urinary bladder. Cancer 70:1967–1973 Shibutani YF, Schoenberg MP, Carpiniello VL, Malloy TR, 1992, Human papillomavirus associated with bladder cancer. Urology 40:15–17 Chetsanga C, Malmstrom PU, Gyllensten U et al, 1992, Low incidence of human papillomavirus type 16 DNA in bladder tumor detected by the polymerase chain reaction. Cancer 69:1208–1211 Knowles MA, 1992, Human papillomavirus sequences are not detectable by Southern blotting or general primer-mediated polymerase chain reaction in transitional cell tumours of the bladder. Urol Res 20:297–301 Bryant P, Davies P, Wilson D, 1991, Detection of human papillomavirus DNA in cancer of the urinary bladder by in situ hybridisation. Br J Urol 68:49–52 de Leon DC, Montiel DP, Nemcova J et al, 2009, Human papillomavirus, HPV, in breast tumors: prevalence in a group of Mexican patients. BMC Cancer 9:26 Khan NA, Castillo A, Koriyama C et al, 2008, Human papillomavirus detected in female breast carcinomas in Japan. Br J Cancer 99:408–414 Akil N, Yasmeen A, Kassab A et al, 2008, High-risk human papillomavirus infections in breast cancer in Syrian women and their association with Id-1 expression: a tissue microarray study. Br J Cancer 99:404–407 Choi YL, Cho EY, Kim JH et al, 2007, Detection of human papillomavirus DNA by DNA chip in breast carcinomas of Korean women. Tumour Biol 28:327–332 Mendizabal-Ruiz AP, Morales JA, Ramirez-Jirano LJ et al, 2009, Low frequency of human papillomavirus DNA in breast cancer tissue. Breast Cancer Res Treat 114:189–194 Grenier J, Soria JC, Mathieu MC et al, 2007, Differential immunohistochemical and biological profile of squamous cell carcinoma of the breast. Anticancer Res 27:547–555 Gumus M, Yumuk PF, Salepci T et al, 2006, HPV DNA frequency and subset analysis in human breast cancer patients’ normal and tumoral tissue samples. J Exp Clin Cancer Res 25:515–521 Lindel K, Forster A, Altermatt HJ et al, 2007, Breast cancer and human papillomavirus, HPV, infection: no evidence of a viral etiology in a group of Swiss women. Breast 16:172–177 Kroupis C, Markou A, Vourlidis N et al, 2006, Presence of highrisk human papillomavirus sequences in breast cancer tissues and association with histopathological characteristics. Clin Biochem 39:727–731 Kan CY, Iacopetta BJ, Lawson JS, Whitaker NJ, 2005, Identification of human papillomavirus DNA gene sequences in human breast cancer. Br J Cancer 93:946–948 de Villiers EM, Sandstrom RE, zur Hausen H, Buck CE, 2005, Presence of papillomavirus sequences in condylomatous lesions of the mamillae and in invasive carcinoma of the breast. Breast Cancer Res 7:R1–R11 Tsai JH, Tsai CH, Cheng MH et al, 2005, Association of viral factors with non-familial breast cancer in Taiwan by comparison with non-cancerous, fibroadenoma, and thyroid tumor tissues. J Med Virol 75:276–281 Damin AP, Karam R, Zettler CG et al, 2004, Evidence for an association of human papillomavirus and breast carcinomas. Breast Cancer Res Treat 84:131–137 Liu Y, Klimberg VS, Andrews NR et al, 2001, Human papillomavirus DNA is present in a subset of unselected breast cancers. J Hum Virol 4:329–334 Yu Y, Morimoto T, Sasa M, Okazaki K et al, 2000, Human papillomavirus type 33 DNA in breast cancer in Chinese. Breast Cancer 7:33–36 de Cremoux P, Thioux M, Lebigot I, Sigal-Zafrani B, Salmon R, Sastre-Garau X, Institut Curie Breast Group, 2008, No evidence of human papillomavirus DNA sequences in invasive breast carcinoma. Breast Cancer Res Treat 109:55–58 Wrede D, Luqmani YA, Coombes RC, Vousden KH, 1992, Absence of HPV 16 and 18 DNA in breast cancer. Br J Cancer 65:891–894 Bratthauer GL, Tavassoli FA, O’Leary TJ, 1992, Etiology of breast carcinoma: no apparent role for papillomavirus types 6/11/ 16/18. Pathol Res Pract 188:384–386 Widschwendter A, Brunhuber T, Wiedemair A et al, 2004, Detection of human papillomavirus DNA in breast cancer of patients with cervical cancer history. J Clin Virol 31:292–297 Hennig EM, Di Lonardo A, Venuti A, Holm R et al, 1999, HPV 16 in multiple neoplastic lesions in women with CIN III. J Exp Clin Cancer Res 18:369–377 Damin DC, Caetano MB, Rosito MA et al, 2007, Evidence for an association of human papillomavirus infection and colorectal cancer. Eur J Surg Oncol 33:569–574 Bodaghi S, Yamanegi K, Xiao SY et al, 2005, Colorectal papillomavirus infection in patients with colorectal cancer. Clin Cancer Res 11:2862–2867 LuDW, El-Mofty SK,Wang HL, 2003, Expression of p16, Rb, and p53 proteins in squamous cell carcinomas of the anorectal region harboring human papillomavirus DNA. Mod Pathol 16:692–699 Lee YM, Leu SY, Chiang H et al, 2001, Human papillomavirus type 18 in colorectal cancer. J Microbiol Immunol Infect 34:87–91 McGregor B, Byrne P, Kirgan D et al, 1993, Confirmation of the association of human papillomavirus with human colon cancer. Am J Surg 166:738–740 Shah KV, Daniel RW, Simons JW, Vogelstein B, 1992, Investigation of colon cancers for human papillomavirus genomic sequences by polymerase chain reaction. J Surg Oncol 51:5–7 Kirgan D, Manalo P, McGregor B, 1990, Immunohistochemical demonstration of human papilloma virus antigen in human colon neoplasms. J Surg Res 48:397–402 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 103 EP 119 DI 10.1007/s12253-010-9288-0 PG 17 ER PT J AU Elhefnawy, GN AF Elhefnawy, Galal Nadia TI Contribution of Electron Microscopy to the Final Diagnosis of Renal Biopsies in Egyptian Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Renal biopsy; Glomerulonephritis; Electron microscopy ID Renal biopsy; Glomerulonephritis; Electron microscopy AB There are few publications studying the impact and cost benefit relationship of electron microscopy in the diagnosis of glomerulopathies in routine service. The aim of this study is to assess the contribution of EM to the final diagnosis of renal glomerular diseases in Egyptian patients. Retrospective evaluation of 120 renal biopsy specimens received for primary diagnosis at EM center of Ain Shams university Specialized hospital, Cairo Egypt during 2007in the knowledge of light microscopic, immunofluorescence and electron microscopic findings. It was found that EM was essential for diagnosis in 25% of renal biopsies, corresponding to 100% of hereditary glomerulopathies and 23.5% of other glomerulopathies, It was useful to the dignosis in 41.67% of the cases, confirming the preliminary diagnosis. In 33.33% of cases EM was considered unhelpful in diagnosis. It’s concluded that the importance of EM has not decreased during the last years. New glomerular diseases and variants can be diagnosed only by EM as fibrillary glomerulonephritis and immunotactoid glomerulopathy. Routine evaluation of allograft biopsies should include EM to achieve better recognition of capillary lesions of chronic rejection. EM provides useful diagnostic information in about 66% of native renal biopsies. Kidney biopsy protocols should include EM in all biopsy cases. If electron microscopy cannot be performed routinely on all such biopsies, tissue should be reserved for EM studies. C1 [Elhefnawy, Galal Nadia] Ain Shams Faculty of Medicine, Department of Pathology, 11811 Cairo, Egypt. RP Elhefnawy, GN (reprint author), Ain Shams Faculty of Medicine, Department of Pathology, 11811 Cairo, Egypt. EM n.ga.lal@hotmail.com CR Jennette JC, Olson JL, Schwartz MM, Silva FG, 2007, Primer on the diagnosis of renal disease. In: Jennette JC, Olson JL, Schwartz MM, Silva FG, eds, Heptinstall’s pathology of the kidney, 6th edn. Lippincott Williams & Wilkins publishers, p 108 Tucker JA, 2000, The continuing value of electron microscopy in surgical pathology. Ultrastruct Pathol 24(6):383–389 Tighe JR, Jones NF, 1970, The diagnostic value of routine electron microscopy of renal biopsies. Proc R Soc Med 63, 5):475–477 Ben-Bassat M, Stark H, Robson M, Rosenfeld J, 1974, Value of routine electron microscopy in the differential diagnosis of nephrotic syndrome. Pathol Microbiol 41(1):26–40 Dische FE, Parsons V, 1977, Experience in the diagnosis of glomerulonephritis using combined light microscopical, ultrastructural and immunofluorescence techniques; an analysis of 134 cases. Histopathology 1(5):331–362 Spargo BH, 1975, Practical use of electron microscopy for the diagnosis of glomerular disease. Hum Pathol 6(4):405– 420 Siegel NJ, Spargo BH, Kashgarian M, Hayslett JP, 1973, An evaluation of routine electron microscopy in the examination of renal biopsies. Nephron 10(4):209–215 Collan Y, Klockars M, Heino M, 1978, Revision of light microscopic kidney biopsy diagnosis in glomerular disease. Nephron 20(1):24–31 Skjorten F, Halvorsen S, 1981, A study of the value of resin— embedded semi-thin sections and electron microscopy in the diagnosis of renal biopsies. Acta Pathol Microbiol Scand 4:257– 262 Pearson JM, McWilliam LJ, Coyne JD, Curry A, 1994, Value of electron microscopy in diagnosis of renal disease. J Clin Pathol 47, 2):126–128 Hass M, 1997, A reevaluation of routine electron microscopy in the examination of native renal biopsies. J Am Soc Nephrol 1:70– 76 Wang S, Zhan Y, Zou W, 1998, The evaluation of electron microscopy in the pathological diagnosis of renal biopsies. Zonghua yi xue za zhi 78(10):782–784 Sementilli A, Moura LA, Franco MF, 2004, The role of electron microscopy for the diagnosis of glomerulopathies. Sao Paulo Med J, Vol. No.3 Collan Y, Hirsimaki P, Aho H, Wuorela M, Sundstom J, Tertti R, Metsrinne K, 2005, Ultrastruct Pathol 29(6):461–468 Rivera A, Magliato S, Meleg-Smith S, 2001, Value of electron microscopy in the diagnosis of childhood nephrotic syndrome. Ultrastruct Pathol 313–320 Strife CF, Mc Adams AJ, West CD, 1982, Membranoproliferative glomerulonephritis characterized by focal segmental proliferation. Clin Nephrol 18:9 Zhou XJ, Silva FG, 2007, Membranoproliferative glomerulonephritis. In: Jennette JC, Olson JL, Schwartz M, Silva FG, eds, Heptinstall’s pathology of the kidney, 6th edn. Lippincott Williams & Wilkins publishers, p 277 HerraraGA, 1999, The value of electronmicroscopy in the diagnosis and clinical management of lupus nephritis. Ultrastruct Pathol 23:63 Grande JP, Balow JE, 1998, Renal biopsy in lupus nephritis. Lupus 7(9):611–617 Herrara GA, Isaac J, Turbet-Herrara EA, 2009, Role of electon microscopy in transplant renal pathology. Ultrastruct Pathol 21, 6):481–498 Ivanyi B, Kemeny E, Szederkenyi E, Marofka F, Szenohradszky P, 2001, The role of electron microscopy in the diagnosis of chronic renal allograft rejection. Mod Pathol 3; 14(12):1200– 1208 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 121 EP 125 DI 10.1007/s12253-010-9290-6 PG 5 ER PT J AU Jiang, Y Long, H Wang, W Liu, H Tang, Y Zhang, X AF Jiang, Yong Long, Hu Wang, Weiya Liu, Huawei Tang, Yuan Zhang, Xiuhui TI Clinicopathological Features and Immunoexpression Profiles of Goblet Cell Carcinoid and Typical Carcinoid of the Appendix SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Appendiceal neoplasm; CEA; CgA; CK7; CK20; Immunohistochemistry; Goblet cell carcinoid; Ki67; P63; Typical carcinoid ID Appendiceal neoplasm; CEA; CgA; CK7; CK20; Immunohistochemistry; Goblet cell carcinoid; Ki67; P63; Typical carcinoid AB Goblet cell carcinoid (GCC) of the appendix is currently classified as a neuroendocrine tumor, together with typical carcinoid (TC) of the appendix. However, whether GCC is a variant of TC or a mucin-producing adenocarcinoma is still controversial. To get a better understanding, we investigated the clinicopathological features of 55 Chinese patients (26 GCCs and 29 TCs), and explored the histochemical properties and expression profiles of CK7, CK20, P63, CEA, CgA, NSE, CD56, and Ki67 in 37 out of these 55 patients (18 GCCs and 19 TCs). Our results showed GCC had a male predominance, older age involvement, significantly larger tumor size, and significantly more frequency of mesoappendix infiltration than TC. Alcian blue/PAS stains were positive in all the GCC cases, while negative in all the TC cases. CK7 and CK20 expressions were significantly more frequent in GCC (P=0.03 and 0.00001, respectively). However, P63 expression was detected in none of the GCC cases but in 6 TC cases (P=0.02). Although the expression of CgA was similar, strong expression (3+) was significantly more frequent in TC (P=5.7×10−11). Also, NSE and CD56 expressions were significantly more frequent in TC (P=0.02 and 1.26×10−4, respectively). CEA expression was significantly more frequent in GCC (P=2.4×10−6). Finally, Ki67 index was low in GCC (4.7%), but significantly higher than TC (0.9%) (P=5.4×10−6). Taken together, these distinct features support that GCC differs from TC, a classical neuroendocrine tumor, and harbors an immunophenotype of adenocarcinoma. Therefore, the term "low grade adenocarcinoma with neuroendocrine differentiation" might be more appropriate for GCC. C1 [Jiang, Yong] Sichuan University, West China Hospital, Department of Pathology, No. 37, Guo Xue Xiang, 610041 Chengdu, Sichuan, China. [Long, Hu] Sichuan University, West China College of Stomatology, 610041 Chengdu, Sichuan, China. [Wang, Weiya] Sichuan University, West China Hospital, Department of Pathology, No. 37, Guo Xue Xiang, 610041 Chengdu, Sichuan, China. [Liu, Huawei] Sichuan University, West China School of Medicine, 610041 Chengdu, Sichuan, China. [Tang, Yuan] Sichuan University, West China Hospital, Department of Pathology, No. 37, Guo Xue Xiang, 610041 Chengdu, Sichuan, China. [Zhang, Xiuhui] Sichuan University, West China Hospital, Department of Pathology, No. 37, Guo Xue Xiang, 610041 Chengdu, Sichuan, China. RP Zhang, X (reprint author), Sichuan University, West China Hospital, Department of Pathology, 610041 Chengdu, China. EM xiuhui_zhang@yahoo.com CR Capella C, Solcia E, Sobin LH,Arnold R, 2000, Endocrine tumors of the appendix. World Health Organization Classification of Tumours. Pathology and genetics of tumours of the digestive system. In: Hamilton SR, Aaltonen LA, ed, IARC Press, Lyon, pp 99–101 Pahlavan PS, Kanthan R, 2005, Goblet cell carcinoid of the appendix. World J Surg Oncol 3:36 Kanthan R, Saxena A, Kanthan SC, 2001, Goblet cell carcinoids of the appendix: immunophenotype and ultrastructural study. Arch Pathol Lab Med 125(3):386–90 Park K, Blessing K, Kerr K, Chetty U, Gilmour H, 1990, Goblet cell carcinoid of the appendix. Gut 31:322–324 Deans GT, Spence RA, 1995, Neoplastic lesions of the appendix. Br J Surg 82:299–306 Butler TA, Houshiar A, Lin F, Wilson SE, 1994, Goblet cell carcinoid of the appendix. Am J Surg 168:685–687 Alsaad KO, Serra S, Schmitt A, Perren A, Chetty R, 2007, Cytokeratins 7 and 20 immunoexpression profile in goblet cell and classical carcinoids of appendix. Endocr Pathol 18(1):16–22 van Eeden S, Offerhaus GJ, Hart AA et al, 2007, Goblet cell carcinoid of the appendix: a specific type of carcinoma. Histopathology 51(6):763–73 Ramnani DM, Wistuba II, Behrens C et al, 1999, K-ras and p53 mutations in the pathogenesis of classical and goblet cell carcinoids of the appendix. Cancer 86(1):14–21 Stancu M, Wu TT, Wallace C et al, 2003, Genetic alterations in goblet cell carcinoids of the vermiform appendix and comparison with gastrointestinal carcinoid tumors. Mod Pathol 16(12):1189–98 Subbuswamy SG, Gibbs NM, Ross CF, Morson BC, 1974, Goblet cell carcinoid of the appendix. Cancer 34(2):338–44 Kende AI, Carr NJ, Sobin LH, 2003, Expression of cytokeratins 7 and 20 in carcinomas of the gastrointestinal tract. Histopathology 42(2):137–40 Toumpanakis C, Standish RA, Baishnab E, Winslet MC, Caplin ME, 2007, Goblet cell carcinoid tumors, adenocarcinoid, of the appendix. Dis Colon Rectum 50(3):315–22 Wang BY, Gil J, Kaufman D et al, 2002, P63 in pulmonary epithelium, pulmonary squamous neoplasms, and other pulmonary tumors. Hum Pathol 33(9):921–6 Owens SR, Greenson JK, 2007, Immunohistochemical staining for p63 is useful in the diagnosis of anal squamous cell carcinomas. Am J Surg Pathol 31(2):285–90 Sokmensuer C, Gedikoglu G, Uzunalimoglu B, 2001, Importance of proliferation markers in gastrointestinal carcinoid tumors: a clinicopathologic study. Hepatogastroenterology 48(39):720–3 Li CC, Hirowaka M, Qian ZR, Xu B, Sano T, 2002, Expression of E-cadherin, b-catenin, and Ki-67 in goblet cell carcinoids of the appendix: an immunohistochemical study with clinical correlation. Endocr Pathol 13(1):47–58 McCusker ME, Cote TR, Clegg LX, Sobin LH, 2002, Primary malignant neoplasms of the appendix: a population-based study from the surveillance, epidemiology and end-results program, 1973–1998. Cancer 94(12):3307–12 Konishi T, Watanabe T, Muto T, Kotake K, Nagawa H, 2006, Site distribution of gastrointestinal carcinoids differs between races. Gut 55(7):1051–2 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 127 EP 132 DI 10.1007/s12253-010-9291-5 PG 6 ER PT J AU Warchol, T Kruszyna, Lianeri, M Roszak, A Jagodzinski, PP AF Warchol, Teresa Kruszyna, Lukasz Lianeri, Margarita Roszak, Andrzej Jagodzinski, P Pawel TI Distribution of CCND1 A870G Polymorphism in Patients with Advanced Uterine Cervical Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical carcinoma; CCND1; Polymorphism ID Cervical carcinoma; CCND1; Polymorphism AB We examined the distribution of the CCND1 A870G (rs9344) polymorphic variant in patients with cervical cancer (n=129) and healthy individuals (n=288) in a sample of a Polish cohort. We showed that patients with advanced cervical cancer bearing the CCND1 A/A and A/G genotypes displayed a 1.811-fold increased risk of cervical cancer (95% CI=1.150–2.852, p=0.0098). We also found a significantly higher frequency of the CCND1 870A allele in patients with cancer than in controls, p=0.0116. Our investigation confirmed that the CCND1 870A gene variant may be a genetic risk factor in the incidence of advanced cervical cancer. C1 [Warchol, Teresa] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. [Kruszyna, Lukasz] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. [Lianeri, Margarita] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. [Roszak, Andrzej] Greater Poland Cancer Center, Department of Radiotherapy and Gynecological OncologyPoznan, Poland. [Jagodzinski, P Pawel] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. RP Jagodzinski, PP (reprint author), Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 60-781 Poznan, Poland. EM pjagodzi@am.poznan.pl CR Parkin DM, Bray F, Ferlay J, Pisani P et al, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Parkin DM, Bray F, 2006, Chapter 2: the burden of HPV-related cancers. Vaccine 24(S3):11–25 Munoz N, Bosch FX, de Sanjose S et al, 2003, Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med 348:518–527 Baseman JG, Koutsky LA, 2005, The epidemiology of human papillomavirus infections. J Clin Virol 32(Suppl 1):S16–24 Magnusson PK, Sparen P, Gyllensten UB, 1999, Genetic link to cervical tumours. Nature 400:29–30 Hemminki K, Dong C, Vaittinen P, 1999, Familial risks in cervical cancer: is there a hereditary component? Int J Cancer 82:775–781 Hemminki K, Chen B, 2006, Familial risks for cervical tumors in full and half siblings: etiologic apportioning. Cancer Epidemiol BiomarkersPrev 15:1413–1414 Kim JK, Diehl JA, 2009, Nuclear cyclin D1: an oncogenic driver in human cancer. J Cell Physiol 220:292–296 Hall M, Peters G, 1996, Genetic alterations of cyclins, cyclindependent kinases, and Cdk inhibitors in human cancer. Adv Cancer Res 68:67–108 Wang L, Habuchi T, Mitsumori K et al, 2003, Increased risk of prostate cancer associated with AA genotype of cyclin D1 gene A870G polymorphism. Int J Cancer 103:116–120 Onay UV, Aaltonen K, Briollais L et al, 2008, Combined effect of CCND1 and COMT polymorphisms and increased breast cancer risk. BMC Cancer 8:6 Le Marchand L, Seifried A, Lum-Jones A et al, 2003, Association of the cyclin D1 A870G polymorphism with advanced colorectal cancer. JAMA 290:2843–2848 Rydzanicz M, Golusinski P, Mielcarek-Kuchta D et al, 2006, Cyclin D1 gene, CCND1, polymorphism and the risk of squamous cell carcinoma of the larynx. Eur Arch Otorhinolaryngol 263:43–48 Izzo JG, Malhotra U, Wu TT et al, 2005, Impact of cyclin D1 A870G polymorphism in esophageal adenocarcinoma tumorigenesis. Semin Oncol 32:S11–5 Hou X, Wang S, Zhou Y et al, 2005, Cyclin D1 gene polymorphism and susceptibility to childhood acute lymphoblastic leukemia in a Chinese population. Int J Hematol 82:206–209 Castro FA, Haimila K, Sareneva I et al, 2009, Association of HLA-DRB1, interleukin-6 and cyclin D1 polymorphisms with cervical cancer in the Swedish population–a candidate gene approach. Int J Cancer 125:1851–1858 Thakur N, Hussain S, Kohaar I et al, 2009, Genetic variant of CCND1: association with HPV-mediated cervical cancer in Indian population. Biomarkers 14:219–225 Satinder K, Chander SR, Pushpinder K et al, 2008, Cyclin D1, G870A, polymorphism and risk of cervix cancer: a case control study in north Indian population. Mol Cell Biochem 315:151–157 Betticher DC, Thatcher N, Altermatt HJ et al, 1995, Alternate splicing produces a novel cyclin D1 transcript. Oncogene 11:1005–11011 Lu F, Gladden AB, Diehl JA, 2003, An alternatively spliced cyclin D1 isoform, cyclin D1b, is a nuclear oncogene. Cancer Res 63:7056–7061 Solomon DA, Wang Y, Fox SR et al, 2003, Cyclin D1 splice variants. Differential effects on localization, RB phosphorylation, and cellular transformation. J Biol Chem 278:30339–30347 Jeon YT, Kim JW, Song JH et al, 2005, Cyclin D1 G870A polymorphism and squamous cell carcinoma of the uterine cervix in Korean women. Cancer Lett 223:259–263 Catarino R, Matos A, Pinto D et al, 2005, Increased risk of cervical cancer associated with cyclin D1 gene A870G polymorphism. Cancer Genet Cytogenet 160:49–54 Martin CM, Astbury K, O'Leary JJ, 2006, Molecular profiling of cervical neoplasia. Expert Rev Mol Diagn 6:217–229 Sherr CJ, 1993, Mammalian G1 cyclins. Cell 73:1059–1065 Sherr CJ, Roberts JM, 1999, CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev 13:1501–1512 Jares P, Colomer D, Campo E, 2007, Genetic and molecular pathogenesis of mantle cell lymphoma: perspectives for new targeted therapeutics. Nat Rev Cancer 7:750–762 Jin M, Inoue S, Umemura T et al, 2001, Cyclin D1, p16 and retinoblastoma gene product expression as a predictor for prognosis in non-small cell lung cancer at stages I and II. Lung Cancer 34:207–218 Bartkova J, Lukas J, Muller H et al, 1995, Abnormal patterns of D-type cyclin expression and G1 regulation in human head and neck cancer. Cancer Res 55:949–956 Bartkova J, Lukas J, Muller H et al, 1994, Cyclin D1 protein expression and function in human breast cancer. Int J Cancer 57:353–361 Shamma A, Doki Y, Shiozaki H et al, 2000, Cyclin D1 overexpression in esophageal dysplasia: a possible biomarker for carcinogenesis of esophageal squamous cell carcinoma. Int J Oncol 16:261–266 Al Moustafa AE, Foulkes WD, Wong A et al, 2004, Cyclin D1 is essential for neoplastic transformation induced by both E6/E7 and E6/E7/ErbB-2 cooperation in normal cells. Oncogene 23:5252–5256 Wang L, Habuchi T, Takahashi T et al, 2002, Carcinogenesis 23:257–264 Pabalan N, Bapat B, Sung L et al, 2008, Cyclin D1 Pro241Pro, CCND1-G870A, polymorphism is associated with increased cancer risk in human populations: a meta-analysis. Cancer Epidemiol Biomarkers Prev 17:2773–2781 Huh K, Zhou X, Hayakawa H et al, 2007, Human papillomavirus type 16 E7 oncoprotein associates with the cullin 2 ubiquitin ligase complex, which contributes to degradation of the retinoblastoma tumor suppressor. J Virol 81:9737–9747 Huh KW, DeMasi J, Ogawa H et al, 2005, Association of the human papillomavirus type 16 E7 oncoprotein with the 600-kDa retinoblastoma protein-associated factor, p600. Proc Natl Acad Sci U S A 102:11492–11497 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 133 EP 137 DI 10.1007/s12253-010-9293-3 PG 5 ER PT J AU de Armas, Y Capo, V Gonzalez, I Mederos, L Diaz, R de Waard, HJ Rodriguez, A Garcia, Y Cabanas, R AF de Armas, Yaxsier Capo, Virginia Gonzalez, Ida Mederos, Lilian Diaz, Raul de Waard, H Jacobus Rodriguez, Alberto Garcia, Yarmila Cabanas, Ricardo TI Concomitant Mycobacterium avium Infection and Hodgkin’s Disease in a Lymph Node from an HIV-negative Child SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Mycobacterium avium; Hodgkin’s disease; PCR ID Mycobacterium avium; Hodgkin’s disease; PCR AB We report a case of an immunocompetent child with simultaneously an infection with Mycobacterium avium and Hodgkin’s disease in a cervical lymph node. A positive PCR result for M. avium on a biopsy of the lymph node directed the definitive diagnosis for both etiologies and avoided a possible dissemination of this infection after chemotherapy was started. C1 [de Armas, Yaxsier] Institute of Tropical MedicineHavana City, Cuba. [Capo, Virginia] Institute of Tropical MedicineHavana City, Cuba. [Gonzalez, Ida] Institute of Tropical MedicineHavana City, Cuba. [Mederos, Lilian] Institute of Tropical MedicineHavana City, Cuba. [Diaz, Raul] Institute of Tropical MedicineHavana City, Cuba. [de Waard, H Jacobus] Instituto de BiomedicinaCaracas, Venezuela. [Rodriguez, Alberto] Limonar County HospitalLimonar, Matanzas Province, Cuba. [Garcia, Yarmila] Juan Manuel Marquez HospitalHavana City, Cuba. [Cabanas, Ricardo] Juan Manuel Marquez HospitalHavana City, Cuba. RP de Armas, Y (reprint author), Institute of Tropical Medicine, Havana City, Cuba. EM Yaxsier@ipk.sld.cu CR Jarzembowski JA, Young MB, 2008, Nontuberculous mycobacterial infections. Arch Pathol Lab Med 132:1333–1341 Rosenberg SA, Kaplan HS, 1966, Evidence for an orderly progression in the spread of Hodgkin’s disease. Cancer Res 26:1225–1231 Karakas Z, Agaoglu L, Taravari B et al, 2003, Pulmonary tuberculosis in children with Hodgkin’s lymphoma. Hematol J 4:78–81 Centkowski P, Sawczuk-Chabin J, Prochorec M, Warzocha K, 2005, Hodgkin’slymphoma and tuberculosis coexistence in cervical lymph nodes. Leuk Lymphoma 46:471–475 Telenti A, Marchesi F, Balz M, Bally F, Bottger EC, Bodmer T, 1993, Rapid identification of mycobacteria to the species level by polymerase chain reaction and restriction enzyme analysis. J Clin Microbiol 31:175–178 Cook SM, Bartos RE, Pierson CL, Frank TS, 1994, Detection and characterization of atypical mycobacteria by the polymerase chain reaction. Diagn Mol Pathol 23:53–58 Brousset P, Marchou B, Chittal SM, Delsol G, 1994, Concomitant. Mycobacterium avium complex infection and Epstein-Barr virus associated Hodgkin’s disease in a lymph node from a patient with AIDS. Histopathology 24:586–588 Schulz S, Cabras AD, Kremer M et al, 2005, Species identification of mycobacteria in paraffin-embedded tissues: frequent detection of nontuberculous mycobacteria. Modern Pathol 18:274–282 Thegerstrom J, Romanus V, Friman V, Brudin L, Haeming PD, Olsen B, 2008, Mycobacterium avium lymphadenopathy among children, Sweden. Emerg Infect Dis 14:661–663 Falkinham JO III, 1996, Epidemiology of infection by nontuberculous mycobacteria. Clin Microbiol Rev 9:177–215 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 139 EP 140 DI 10.1007/s12253-010-9275-5 PG 2 ER PT J AU Demeter, J Istenes, I Fodor, A Paksi, M Dombi, P Valasinyoszki, E Csomor, J Matolcsy, A Nagy, GZs AF Demeter, Judit Istenes, Ildiko Fodor, Aniko Paksi, Melinda Dombi, Peter Valasinyoszki, Erika Csomor, Judit Matolcsy, Andras Nagy, G Zsolt TI Efficacy of Romiplostim in the Treatment of Chemotherapy Induced Thrombocytopenia (CIT) in a Patient with Mantle Cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Chemotherapy; Lymphoma; Thrombocytopenia; Romiplostim ID Chemotherapy; Lymphoma; Thrombocytopenia; Romiplostim AB Chemotherapy induced thrombopenia (CIT) is difficult to treat, as previous treatment options, including recombinant human thrombopoietin proved to be of limited efficacy. Here we report a case of a mantle cell lymphoma patient treated with intensive chemotherapy, who belongs to Yehova’s witnesses and therefore did not accept platelet transfusions. At the time of severe thrombocytopenia (zero thrombocytes/ per mikroliter) and gastrointestinal bleeding, on day 13 following the start of hyperCVAD B chemotherapy, romiplostim treatment was given resulting in quick normalisation of the platelet count followed by thrombocytosis. Based on our observation in further studiesmodification of the dose and timing of romiplostim injection in CIT should be considered. C1 [Demeter, Judit] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor utca 2/A, 1083 Budapest, Hungary. [Istenes, Ildiko] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor utca 2/A, 1083 Budapest, Hungary. [Fodor, Aniko] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor utca 2/A, 1083 Budapest, Hungary. [Paksi, Melinda] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor utca 2/A, 1083 Budapest, Hungary. [Dombi, Peter] Szent Borbala Hospital, Department of HematologyTatabanya, Hungary. [Valasinyoszki, Erika] Szent Borbala Hospital, Department of HematologyTatabanya, Hungary. [Csomor, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Matolcsy, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Nagy, G Zsolt] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor utca 2/A, 1083 Budapest, Hungary. RP Demeter, J (reprint author), Semmelweis University, 1st Department of Internal Medicine, 1083 Budapest, Hungary. EM demjud@bel1.sote.hu CR Levy B, Arnason JE, Bussel JB, 2008, The use of secondgeneration thrombopoietic agents for chemotherapy-induced thrombocytopenia. Curr Opin Oncol 20:690–696 Natale R, Charu V, Schutte W et al, 2009, Safety of romiplostim for treatment of chemotherapy-induced thrombocytopenia, CIT, in patients with advanced non-small cell lung cancer, NSCLC). Eur J Cancer Suppl 7:574 Fanale M, Stiff P, Noonan K et al, 2009, Safety of romiplostim for treatment of severe chemotherapy induced thrombocytopenia, CIT, in patients with lymphoma receiving multi-cycle chemotherapy: results from an open-label dose- and schedule-finding study. Eur J Cancer Suppl 7:562 Dalal S, Boddapati M, Lowery MN et al, 2006, Treatment of acute myeloid leukemia in a Jehovah’s Witness. Ann Hematol 85:407– 408 Nash MJ, Cohen H, 2004, Management of Jehovah's Witness patients with haematological problems. Blood Rev 18:211–7 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 141 EP 143 DI 10.1007/s12253-010-9276-4 PG 3 ER PT J AU Rekhi, B Folpe, LA Deshmukh, M Jambhekar, AN AF Rekhi, Bharat Folpe, L Andrew Deshmukh, Mahesh Jambhekar, Ajit Nirmala TI Sclerosing Epithelioid Fibrosarcoma–A Report of Two Cases with Cytogenetic Analysis of FUS Gene Rearrangement by FISH Technique SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sclerosing epithelioid fibrosarcoma; Low-grade fibromyxoid sarcoma; Soft tissue sarcomas; FISH technique; FUS rearrangement ID Sclerosing epithelioid fibrosarcoma; Low-grade fibromyxoid sarcoma; Soft tissue sarcomas; FISH technique; FUS rearrangement AB Sclerosing epithelioid fibrosarcoma (SEF) is a rare soft tissue sarcoma. Recently, a link has been suggested between SEF and low-grade fibromyxoid sarcoma (LGFMS) on the basis of the finding of the characteristic translocation t(7;16) (FUS-CREB3L2) of LGFMS in a small number of studied cases of SEF. The frequency of this translocation in SEF is still unknown. We present 2 cases of SEF with cytogenetic analysis for FUS rearrangement. The tumors occurred in 12 and 58 year old patients, respectively and consisted of a well to partially circumscribed, non-encapsulated mass, comprising monomorphic, polygonal cells arranged in aggregates, cords and single file arrays in a variably sclerotic stroma. The cells exhibited minimal nuclear atypia with moderate amount of clear to eosinophilic cytoplasm and rare mitotic figures. One case also showed bland spindle cell areas with myxoid change, as seen in LGFMS. By immunohistochemistry (IHC), the tumor cells were diffusely positive for vimentin, focally for S-100 in 1 case and negative for cytokeratin (CK), epithelial membrane antigen (EMA), HMB-45, desmin, smooth muscle actin (SMA), H-caldesmon, Myo D-1, CD34 and CD 168. By fluorescent in-situ hybridization (FISH) technique, the case with mixed SEF and LGFMS histology was positive for FUS rearrangement. Our study reinforces the previously reported relationship between SEF and LGFMS, and suggests that SEF may represent a variant of LGFMS in at least some cases, rather than an entirely distinct fibrosarcoma variant. C1 [Rekhi, Bharat] Tata Memorial Hospital, Department of Pathology, Dr E.B. Road, Parel, 400012 Mumbai, Maharashtra, India. [Folpe, L Andrew] Mayo Clinic, Department of Laboratory Medicine and Pathology, 200 First Street SW, 55905 Rochester, MN, USA. [Deshmukh, Mahesh] Tata Memorial Hospital, Department of Pathology, Dr E.B. Road, Parel, 400012 Mumbai, Maharashtra, India. [Jambhekar, Ajit Nirmala] Tata Memorial Hospital, Department of Pathology, Dr E.B. Road, Parel, 400012 Mumbai, Maharashtra, India. RP Rekhi, B (reprint author), Tata Memorial Hospital, Department of Pathology, 400012 Mumbai, India. EM rekhi.bharat@gmail.com CR Meis-Kindblom JM, Kindblom LG, van den Berg E, Molennar WM, 2002, Tumors of soft tissue and bone. Pathology and genetics. In: Fletcher CDM, Unni K, Mertens F, eds, World Health Organization classification of tumors. IARC, Lyon, pp 200–204 Antonescu CR, Rosenblum MK, Pereira P, Nascimento AG, Woodruff JM, 2001, Sclerosing epithelioid fibrosarcoma; a study of 16 cases and confirmation of a clinically distinct tumor. Am J Surg Pathol 25:699–709 Meis-Kindblom JM, Kindblom LG, Enzinger FM, 1995, Sclerosing epithelioid fibrosarcoma: a variant of fibrosarcoma simulating a carcinoma. Am J Pathol 19:979–993 Eyden BP, Manson C, Banerjee SS, Roberts ISD, Harris M, 1998, Sclerosing epithelioid fibrosarcoma: a study of five cases emphasizing diagnostic criteria. Histopathology 33:354–360 Chow LTC, Lui YH, Kumta SM, Allen PW, 2004, Primary sclerosing epithelioid fibrosarcoma of the sacrum: a case report and review of the literature. J Clin Pathol 57:90–94 Smith P, Almeida B, Krajacevic TB, 2008, Sclerosing epithelioid fibrosarcoma as a rare cause of ascites in a young man; a case report. J Med Case reports 2:248 Guillou L, Benhattar J, Gengler C, Gallagher G, Ranchere-Vince D, Collin F, Terrier P, Terrier-Lacombe MJ, Leroux A, Marques B, Aubain Somerhausen Nde S, Keslair F, Pedeutour F, Coindre JM, 2007, Translocation-positive low-grade fibromyxoid sarcoma: clinicopathologic and molecular analysis of a series expanding the morphologic spectrum and suggesting potential relationship to sclerosing epithelioid fibrosarcoma: a study from the French Sarcoma Group. Am J Surg Pathol 31:1387–1402 Medeiros F, Erickson-Johnson MR, Keeney GL, Clayton AC, Nascimento AG, Wang X, Oliveira AM, 2007, Frequency and characterization of HMGA2 and HMGA1 rearrangements in mesenchymal tumors of the lower genital tract. Genes Chromosomes Cancer 46:981–990 Reid R, de Silva MV, Paterson L, Ryan E, Fisher C, 2003, Lowgrade fibromyxoid sarcoma and hyalinizing spindle cell tumor with giant rosettes share a common t(7;16)(q34;p11, translocation. Am J Surg Pathol 27:1229–1236 Folpe AL, Weiss SW, 2003, Ossifying fibromyxoid tumor of soft parts: a clinicopathologic study of 70 cases with emphasis on atypical and malignant variants. Am J Surg Pathol 27:421– 431 Folpe AL, Goldblum JR, Rubin BP, Shehata BM, Liu W, Dei Tos AP, Weiss SW, 2005, Morphologic and immunophenotypic diversity in Ewing family tumors: a study of 66 genetically confirmed cases. Am J Surg Pathol 29:1025–1033 Folpe AL, McKenney JK, Bridge JA, Weiss SW, 2002, Sclerosing rhabdomyosarcoma in adults: report of four cases of a hyalinizing, matrix-rich variant of rhabdomyosarcoma that may be confused with osteosarcoma, chondrosarcoma, or angiosarcoma. Am J Surg Pathol 26:1175–1183 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 145 EP 148 DI 10.1007/s12253-010-9277-3 PG 4 ER PT J AU Patrelli, ST Silini, ME Berretta, R Thai, E Gizzo, S Bacchi Modena, A Nardelli, BG AF Patrelli, Silvio Tito Silini, Maria Enrico Berretta, Roberto Thai, Elena Gizzo, Salvatore Bacchi Modena, Alberto Nardelli, Battista Giovanni TI Squamotransitional Cell Carcinoma of the Vagina: Diagnosis and Clinical Management SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Squamotransitional cell carcinoma; Transitonal cell carcinoma; HPV-DNA; Immunohistochemistry; Vagina; Cervix; Endometrium; Urogenital tract; Urothelial carcinoma; Squamous carcinoma ID Squamotransitional cell carcinoma; Transitonal cell carcinoma; HPV-DNA; Immunohistochemistry; Vagina; Cervix; Endometrium; Urogenital tract; Urothelial carcinoma; Squamous carcinoma AB Primary squamotransitional cell carcinoma (STCC) is rare squamous cell tumor variant resembling transitional cell carcinoma (TCC) of the urinary tract. STCC occurs rarely in the vagina and its clinical and pathological correlates are poorly known. We report a unique case of a 66-year-old Italian woman with STCC of the vagina. A biopsy of the tumor was performed. The tumor qualified as a STCC. Following biopsy, the patient underwent radical hysterectomy (Piver’s III-type) with bilateral salpingo-oophorectomy, upper colpectomy, appendicectomy, peritoneal cytology, and lymphadenectomy. The patient is now healthy without evidence of recurrence at 30 months after surgery. Pathologically, cytoarchitectural characteristics distinguish this histotype (STCC) from conventional squamous cell carcinoma of the genital tract. The cytokeratin staining pattern (CK7 positive and CK20 negative), the p63 expression and the positivity for p16ink4a and high-risk HPV are the main elements of differential diagnosis. We suggest that STCC of the vagina should be treated by radical surgery, possibly followed by adjuvant therapy based on staging results and should receive a long-term follow-up. C1 [Patrelli, Silvio Tito] University of Parma, Ob/Gyn Clinic, Via Gramsci, 14, 43100 Parma, Italy. [Silini, Maria Enrico] University of Parma, Institute of PathologyParma, Italy. [Berretta, Roberto] University of Parma, Ob/Gyn Clinic, Via Gramsci, 14, 43100 Parma, Italy. [Thai, Elena] University of Parma, Institute of PathologyParma, Italy. [Gizzo, Salvatore] University of Parma, Ob/Gyn Clinic, Via Gramsci, 14, 43100 Parma, Italy. [Bacchi Modena, Alberto] University of Parma, Ob/Gyn Clinic, Via Gramsci, 14, 43100 Parma, Italy. [Nardelli, Battista Giovanni] University of Parma, Ob/Gyn Clinic, Via Gramsci, 14, 43100 Parma, Italy. RP Patrelli, ST (reprint author), University of Parma, Ob/Gyn Clinic, 43100 Parma, Italy. EM titosilvio.patrelli@gmail.com CR Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics, 2009. CA Cancer J Clin 59(4):225–249 Peters WA III, Kuman NB, Morley GW, 1985, Carcinoma of the vagina. Cancer 55:892–897 Rubin SC, Young J, Mikuta JJ, 1985, Squamous carcinoma of the vagina: treatment, complications, and long term follow-up. Gynecol Oncol 20:346–353 Benedet JL, Murphy KJ, Fairey RN, Boyes DA, 1983, Primary invasive carcinoma of the vagina. Obstet Gynecol 62:715–719 Rose PG, Stoler MH, Abdul-Karim FW, 1998, Papillary squamotransitional cell carcinoma of the vagina. Int J Gynecol Pathol 17:372–375 Vesoulis Z, Erhardt CA, 2001, Cytologic diagnosis of vaginal papillary squamotransitional cell carcinoma. A case report. Acta Cytol 45:465–469 Gao Z, Bhuiya T, Falkowski O, 2005, Papillary squamotransitional cell carcinoma of the vagina: a case report and review of literature. J Obstet Gynaecol 25:94–96 Kleter B, van Doorn LJ, Schrauwen L, Molijn A, Sastrowijoto S, ter Schegget J, Lindeman J, ter Harmsel B, Burger M, Quint W, 1999, Development and clinical evaluation of a highly sensitive PCR-reverse hybridization line probe assay for detection and identification of anogenital human papillomavirus. J Clin Microbiol 37:2508–2517 Koenig C, Turnicky RP, Kankam CF, Tavassoli FA, 1997, Papillary squamotransitional cell carcinoma of the cervix: a report of 32 cases. Am J Surg Pathol 21:915–921 Randall ME, Andersen WA, Mills SE, Kim JA, 1986, Papillary squamous cell carcinoma of the uterine cervix: a clinicopathologic study of nine cases. Int J Gynecol Pathol 5:1–10 Urist MJ, Di Como CJ, Lu ML, Charytonowicz E, Verbel D, Crum CP, Ince TA, McKeon FD, Cordon-Cardo C, 2002, Loss of p63 expression is associated with tumor progression in bladder cancer. Am J Pathol 161:1199–1206 Lininger RA, Wistuba I, Gazdar A, Koenig C, Tavassoli FA, Albores-Saavedra J, 1998, Human papillomavirus type 16 is detected in transitional cell carcinomas and squamotransitional cell carcinomas of the cervix and endometrium. Cancer 83:521– 527 Robert ME, Fu YS, 1990, Papillary squamous cell carcinoma of the uterine cervix—a review with emphasis on prognostic factors and unusual variants. Semin Diagn Pathol 7:173– 189 Qizilbash AH, 1974, Papillary squamous tumors of the uterine cervix: a clinical and pathologic study of 21 cases. Am J Clin Pathol 61:508–520 Bass PS, Birch B, Smart C, Theaker JM, Wells M, 1994, Lowgrade transitional cell carcinoma of the vagina—an unusual cause of vaginal bleeding. Histopathology 24(6):581–583 Fetissof F, Haillot O, Lanson Y, Arbeille B, Lansac J, 1990, Papillary tumour of the vagina resembling transitional cell carcinoma. Pathol Res Pract 186(3):358–364 Jendresen MB, Kvist E, Glenthoj A, 1997, Papillary transitional cell tumour in the vagina. Scand J Urol Nephrol 31, 1):107–108 Singer G, Hohl MK, Hering F, Anabitarte M, 1998, Transitional cell carcinoma of the vagina with pagetoid spread pattern. Hum Pathol 29(3):299–301 Tardio JC, Salas C, 2001, Vaginal papillary carcinomas with transitional cell differentiation: a morphological variant of squamous cell carcinoma? Histopathology 39(4):436–438 Mondaini N, Giubilei G, Raspollini MR, Crisci A, Orlando V, 2005, Recurrence of vaginal implantation of transitional cell carcinoma of the urinary tract. Gynecol Oncol 97, 2):669–670 Hermanova M, Vitezslav V, Husicka R, Neumanova R, Pacik D, 2008, Multicentric transitional cell carcinoma of the vagina and the ureter. Ann Diagn Pathol 12(5):365–367 Labonte S, Tetu B, Boucher D, Larue H, 2001, Transitional cell carcinoma of the endometrium associated with a benign ovarian Brenner tumor: a case report. Hum Pathol 32:230–232 Ordonez NG, 2000, Transitional cell carcinomas of the ovary and bladder are immunophenotypically different. Histophatology 36:433–438 Ollayos CW, Lichy J, Duncan BW, Ali IS, 1996, Papillary squamous carcinoma of the uterine cervix: report of case with HPV 16 DNA and brief review. Gynecol Oncol 63:388–391 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 149 EP 153 DI 10.1007/s12253-010-9280-8 PG 5 ER PT J AU Ioannidis, O Papaemmanuil, S Kakoutis, E Papadopoulos, G Chatzopoulos, S Kotronis, A Makrantonakis, N AF Ioannidis, Orestis Papaemmanuil, Stylliani Kakoutis, Emmanouil Papadopoulos, George Chatzopoulos, Stavros Kotronis, Anastasios Makrantonakis, Nikolaos TI Fibroepithelioma of Pinkus in Continuity with Nodular Basal Cell Carcinoma: Supporting Evidence of the Malignant Nature of the Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Basal cell carcinoma; Fibroepithelioma of Pinkus; Nodular; Skin cancer; Trichoblastoma ID Basal cell carcinoma; Fibroepithelioma of Pinkus; Nodular; Skin cancer; Trichoblastoma AB Basal cell carcinoma, the most common skin cancer, has several clinical and histopathological variants, with its most common form being nodular basal cell carcinoma. Fibrioepithelioma of Pinkus is considered as an unusual variant of basal cell carcinoma by some authors while others consider it to be a benign analogue of basal cell carcinoma. We present a rare case of fibroepithelioma of Pinkus in continuity with a nodular basal cell carcinoma, a finding that supports the classification of fibroepithelioma of Pinkus as a variant of basal cell carcinoma. C1 [Ioannidis, Orestis] General Regional Hospital ‘George Papanikolaou’, First Surgical Department, Alexandrou Mihailidi 13, 54640 Thessaloniki, Greece. [Papaemmanuil, Stylliani] General Regional Hospital ‘George Papanikolaou’, Department of PathologyThessaloniki, Greece. [Kakoutis, Emmanouil] General Regional Hospital ‘George Papanikolaou’, First Surgical DepartmentThessaloniki, Greece. [Papadopoulos, George] General Regional Hospital ‘George Papanikolaou’, First Surgical DepartmentThessaloniki, Greece. [Chatzopoulos, Stavros] General Regional Hospital ‘George Papanikolaou’, First Surgical DepartmentThessaloniki, Greece. [Kotronis, Anastasios] General Regional Hospital ‘George Papanikolaou’, First Surgical DepartmentThessaloniki, Greece. [Makrantonakis, Nikolaos] General Regional Hospital ‘George Papanikolaou’, First Surgical DepartmentThessaloniki, Greece. RP Ioannidis, O (reprint author), General Regional Hospital ‘George Papanikolaou’, First Surgical Department, 54640 Thessaloniki, Greece. EM telonakos@hotmail.com CR Bath-Hextall F, Bong J, Perkins W et al, 2004, Interventions for basal cell carcinoma of the skin: systematic review. BMJ 329:705 Wong CS, Strange RC, Lear JT, 2003, Basal cell carcinoma. BMJ 327:794–8 Crowson AN, 2006, Basal cell carcinoma: biology, morphology and clinical implications. Mod Pathol 19:S127–47 Brooke RC, 2005, Basal cell carcinoma. Clin Med 5:551–4 Misago N, Suzuki Y, Miura Y et al, 2004, Giant polypoid basal cell carcinoma with features of fibroepithelioma of Pinkus and extensive cornification. Eur J Dermatol 14:272–5 Gellin GA, Bender B, 1966, Giant premalignant fibroepithelioma. Arch Dermatol 94:70–3 Pinkus H, 1953, Premalignant fibroepithelial tumors of skin. Arch Dermatol Syphilol 67:598–615 Zalaudek I, Ferrara G, Broganelli P et al, 2006, Dermoscopy patterns of fibroepithelioma of pinkus. Arch Dermatol 142:1318–22 Bowen AR, LeBoit PE, 2005, Fibroepithelioma of pinkus is a fenestrated trichoblastoma. Am J Dermatopathol 27:149–54 Katona TM, Ravis SM, Perkins SM et al, 2007, Expression of androgen receptor by fibroepithelioma of Pinkus: evidence supporting classification as a basal cell carcinoma variant? Am J Dermatopathol 29:7–12 Stern JB, Haupt HM, Smith RR, 1994, Fibroepithelioma of Pinkus: eccrine duct spread of basal cell carcinoma. Am J Dermatopathol 16:585–7 Schulz T, Hartschuh W, 1997, Merkel cells are absent in basal cell carcinomas but frequently found in trichoblastomas. An immunohistochemical study. J Cutan Pathol 24:14–24 Ackerman AB, Gottlieb GJ, 2005, Fibroepithelial tumor of Pinkus is trichoblastic, basal-cell, carcinoma. Am J Dermatopathol 27:155–9 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 155 EP 157 DI 10.1007/s12253-010-9284-4 PG 3 ER PT J AU Terada, T Moriki, T AF Terada, Tadashi Moriki, Toshiaki TI Monolobar Hepatobiliary Fibropolycystic Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Caroli’s disease; Polycystic liver; Monolobar disease; Persistent ductal plate; Portal thrombi; Intrahepatic bile duct development ID Caroli’s disease; Polycystic liver; Monolobar disease; Persistent ductal plate; Portal thrombi; Intrahepatic bile duct development AB We herein report a case of monolobar hepatobiliary fibropolycystic disease. A 75-year-old woman presented with heartburn. Imaging modalities including US, CT, and MRI revealed marked atrophy and multiple biliary cysts of the hepatic left lobe. The hepatic right lobe was normal. ERCP and bile duct endoscopy revealed anomalous pancreaticobiliary union, choledochal dilation, dilation of left intrahepatic bile ducts, and small choledochal non-invasive adenocarcinoma. Polycystic kidney diseases were absent. The patient underwent pancreatico-duodenectomy and extended hepatic left lobectomy. Grossly, the hepatic left lobe was markedly atrophic, and studded with numerous biliary cysts. The left intrahepatic bile ducts were dilated (Caroli’s disease) and the common bile duct showed type I choledochal dilation. The right hepatic lobe was normal. Histologically, the hepatic left lobe was replaced by fibroelastosis. The intrahepatic bile ducts showed ductal plate malformation such as irregular contours, invaginations, and protrusions. The numerous biliary cysts also showed ductal plate malformation. There were numerous persistent ductal plates and microhamartomas. Many hyalinized destructive biliary cysts and ductal plates were recognized. The liver parenchyma was scant and free of hepatocellular malformations. The portal veins showed old obliterative portal thrombosis. The right hepatic lobe was normal. Immunohistochemically, the biliary cells were positive for cytokeratin 7, 8, 18 and 19, and MUC6 and CD10, but negative for MUC2 and MUC5AC. The biliary cysts, persistent ductal plate, and microhamartomas were positive for fetal apomucin antigen MUC1. C1 [Terada, Tadashi] Shizuoka City Shimizu Hospital, Department of Pathology, Miyakami 1231, 424-8636 Shizuoka, Japan. [Moriki, Toshiaki] Shizuoka City Shimizu Hospital, Department of Pathology, Miyakami 1231, 424-8636 Shizuoka, Japan. RP Terada, T (reprint author), Shizuoka City Shimizu Hospital, Department of Pathology, 424-8636 Shizuoka, Japan. EM piyo0111jp@yahoo.co.jp CR Jorgensen MJ, 1977, The ductal plate malformation. Acta Pathol Microbiol Scand Supple 257:1–87 Desmet VJ, 1992, Congenital disease of intrahepatic bile ducts: variations on the theme “ductal plate malformation”. Hepatology 16:1069–1083 Terada T, Nakanuma Y, Ohta G, 1987, Glandular elements around the intrahepatic bile ducts in man: their morphology and distribution in normal livers. Liver 7:1–8 Terada T, Nakanuma Y, 1995, Detection of apoptosis and expression of apoptosis-related proteins during human intrahepatic bile duct development. Am J Pathol 146:67–74 Terada T, Nakanuma Y, 1993, Development of human intrahepatic peribiliary glands: histological, keratin immunohistochemical and mucus histochemical analyses. Lab Invest 68:261–269 Terada T, Okada Y, Nakanuma Y, 1995, Expression of matrix proteinases during human intrahepatic bile duct development: a possible role in biliary cell migration. Am J Pathol 147:1207–1213 Terada T, Nakanuma Y, 1995, Expression of pancreatic enzymes, α-amylase, trypsinogen and lipase, during human liver development and maturation. Gastroenterology 108:1236–1245 Terada T, Nakanuma Y, 1994, Profiles of expression of carbohydrate chain structures during human intrahepatic bile duct development and maturation: a lectin-histochemical and immunohistochemical study. Hepatology 20:388–397 Terada T, Nakanuma Y, 1994, Expression of tenascin, type IV collagen and laminin during human intrahepatic bile duct development and in intrahepatic cholangiocarcinoma. Histopathology 25: 143–150 Terada T, Ohta T, Nakanuma Y, 1994, Expression of transforming growth factor-α and its receptor during human liver development and maturation. Virchows Archiv 424:669–675 Terada T, Nakanuma Y, Sirica AE, 1998, Immunohistochemical demonstration of MET overexpression in human intrahepatic cholangiocarcinoma and in hepatolithiasis. Hum Pathol 29:175–180 Terada T, Ashida K, Kitamura Y, Matsunaga Y, Takashima K, Kato M, Ohta T, 1998, Expression of E-cadherin, alpha-catenin and beta-catenin during human intrahepatic bile duct development. J Hepatol 28:263–269 Terada T, Kato M, Horie S, Endo K, Kitamura Y, 1998, Expression of pancreatic alpha-amylase protein and messenger RNA on hilar primitive bile ducts and hepatocytes during human fetal liver organogenesis: an immunohistochemical and in situ hybridization study. Liver 18:313–319 Terada T, Ukita Y, Ueyama J, Ohta T, 2000, Protein expression of double-stranded RNA-activated protein kinase, PKR, in intrahepatic bile ducts in normal adult livers, fetal livers, primary biliary cirrhosis, hepatolithiasis and intrahepatic cholangiocarcinoma. Liver 20:450–457 Kato M, Shinozawa T, Kato S, Terada T, 2000, Divergent expression of midkine in the human fetal liver and kidney: an immunohistochemical analysis of developmental changes in hilar primitive bile ducts and hepatocytes. Liver 20:475–481 Kato M, Shinozawa T, Kato S, Terada T, 2004, Immunohistochemical localization of truncated midkine in developing human bile ducts. Histol Histopathol 18:129–134 Sasaki M, Nakanuma Y, Terada T, Kim YS, 1995, Biliary epithelial expression of MUC1, MUC2, MUC3 and MUC5/6 apomucins during intrahepatic bile duct development and maturation. Am J Pathol 147:574–579 Terada T, Nakanuma Y, 1993, Development of human peribiliary capillary plexus: a lectin-histochemical and immunohistochemical study. Hepatology 18:529–536 Terada T, Kitamura Y, Nakanuma Y, 1997, Normal and abnormal development of the intrahepatic biliary system: a review Tohoku. J Exp Med 181:19–32 Summerfield JA, Nagafuchi Y, Sherlock S, Cadafalch J, Scheuer PJ, 1986, Hepatobiliary fibropolycystic disease: a clinical and histological review of 51 patients. J Hepatol 2:141–156 Terada T, Kawaguchi M, Furukawa K, Sekido Y, Osamura Y, 2002, Minute mixed ductal-endocrine carcinoma of the pancreas with predominant intraductal growth. Pathol Int 52:740–746 Terada T, Kawaguchi M, 2005, Primary clear cell adenocarcinoma of the peritoneum. Tohoku J Exp Med 271:271–275 Terada T, Tanigichi M, 2004, Intraductal oncocytic papillary neoplasm of the liver. Pathol Int 54:116–123 Terada T, 2009, Gastrointestinal stromal tumor of the uterus: a case report with genetic analyses of c-kit and PDGFRA genes. Int J Gynecol Oncol 28:29–34 Terada T, 2008, Primary multiple extragastrointestinal stromal tumors of the omentum with different mutations of c-kit gene. World J Gastroenterol 14:7256–7259 Nakanuma Y, Kurumaya H, Ohta G, 1984, Multiple cysts in the hepatic hilum and their pathogenesis: a suggestion of periductal gland origin. Virchows Arch [A] 404:341–350 Wanless IR, Zahradnik J, Heathcote EJ, 1987, Hepatic cysts of periductal gland origin presenting as obstructive jaundice. Gastroenterology 93:89 Terada T, Nakanuma Y, 1990, Pathological observations of intrahepatic peribiliary glands in 1000 consecutive autopsy livers: III. Survey of necroinflammation and cystic dilatation Hepatology 12:1229–1233 Kida T, Nakanuma Y, Terada T, 1992, Cystic dilatation of peribiliary glands in livers with adult polycystic disease and livers with solitary non-parasitic cysts: an autopsy study. Hepatology 16:334–340 Terada T, Minato H, Nakanuma Y, ShinozakiK,Kobayashi S,Matsui O, 1992, Ultrasound visualization of hepatic peribiliary cysts: a comparison with morphology. Am J Gastroenterol 87:1499–1502 Itai Y, Ebihara R, Tohno E, Tsunoda HS, Kurosaki Y, Saida Y, Doy M, 1994, Hepatic peribiliary cysts: multiple tiny cysts within the larger portal tract, hepatic hilum, or both. Radiology 191:107–110 Terada T, Nakanuma Y, 1988, Congenital biliary dilatation in autosomal dominant adult polycystic disease of the liver and kidneys. Arch Pathol Lab Med 112:1113–1116 Terada T, Matsushita H, Tashiro J, Sairenji T, Erigichi M, Osada I, 2003, Cholesterol hepatolithiasis with peribiliary cysts. Pathol Int 53:716–720 Roda E, Sama C, Festi D, Aldini R, Mazzella G, Roda A, Barbara L, 1979, Caroli’s disease: description of a rare clinical case with monolobar localization. Am J Gastroenterol 71:621–626 Boyle MJ, Doyle GD, McNulty JG, 1989, Monolobar Caroli’s disease. Am J Gastroenterol 84:1437–1444 Giovanardi RO, 2003, Monolobar Caroli's disease in an adult: case report. Hepatogastroenterology 50:2185–2187 Mohan LN, Thomas PG, Kilpadi AB, D’cunha S, 1991, Liver atrophy associated with monolobar Caroli’s disease. HPB Surg 4:203–206 Popovsky MA, Costa JC, Doppman JL, 1979, Meyenburg complexes of the liver and bile cysts as a consequence of hepatic ischemia. Hum Pathol 10:425–432 Terada T, Takegoshi T, Doishita K, Nakanuma Y, 1988, Histological study of intrahepatic cavernous transformation in a patient with primary myelofibrosis and portal venous thrombosis. Virchows Arch [A] 412:339–345 Terada T, Ishida F, Nakanuma Y, 1989, Vascular plexus around intrahepatic bile ducts in normal livers and portal hypertension. J Hepatol 8:139–149 Terada T, Hoso M, Nakanuma Y, 1989, Microvasculature in the small portal tracts in idiopathic portal hypertension: a morphological comparison with other hepatic diseases. Virchows Arch [A] 415:61–68 Terada T, Nakanuma Y, Hoso M, Obata H, 1991, Expression of HLA-DR antigen on hepatic vascular endothelial cells in idiopathic portal hypertension. Clin Exp Immunol 84:303–307 Terada T, Nakanuma Y, Obata H, 1991, HLA-DR expression on the microvasculature in portal tracts in idiopathic portal hypertension: immunohistochemical characteristics and relation to portal phlebosclerosis. Arch Pathol Lab Med 115:993–997 Van Eyken P, Sciot R, Callea F, Van der Steen K, Moerman P, Desmet VJ, 1988, The development of the intrahepatic bile ducts in man: a keratin immunohistochemical study. Hepatology 8:1589–1595 Awasthi A, Das A, Srinivasan R, Joshi K, 2004, Morphological and immunohistochemical analysis of ductal plate malformation: correlation with fetal liver. Histopathology 45:260–267 Furukawa T, Kloppel G, Volkan Adsay N et al, 2005, Classification of types of intraductal papillary-mucinous neoplasm of the pancreas: a consensus study. Virchows Arch 447:794–799 Shibahara H, Tamada S, Higashi M, Goto M, Batra SK, Hollingsworth MA, Imai K, Yonezawa S, 2004, MUC4 is a novel prognostic factor of intrahepatic cholangiocarcinoma-mass forming type. Hepatology 39:220–229 Torres VE, Harris PC, Pirson Y, 2007, Autosomal dominant polycystic kidney disease. Lancet 14:1287–1301 Gabow PA, 1993, Autosomal dominant polycystic kidney disease. N Eng J Med 332–342 Grantham JJ, 2008, Clinical practice: autosomal dominant polycystic kidney disease. N Eng J Med 359:1477–1485 Young WT, Thomas GV, Blethyn AJ, Lawrie BW, 1992, Choledochal cysts and congenital anomalies of the pancreaticobiliary junction: the clinical findings, radiology and outcome in nine cases. Br J Radiol 65:33–38 Misra SP, Dwivedi M, 1990, Pancreaticobiliary ductal union. Gut 1144–1149 Noun R, Sayegh R, Tohme-Noun C, Honein K, Smayra T, Aoun N, 2006, Extracystic biliary carcinoma associated with anomalous psancreaticobiliary junction and cysts. J Hepatobiliary Pancreat Surg 13:557–559 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 159 EP 165 DI 10.1007/s12253-010-9285-3 PG 7 ER PT J AU Terada, T AF Terada, Tadashi TI Solitary Plasmacytoma of the Thracic Vertebra Presenting with Sudden Paraplegia and Back Pain: A Pathologic Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Plasmacytoma; Vertebra; Bone; Immunohistology; Histopathology ID Plasmacytoma; Vertebra; Bone; Immunohistology; Histopathology AB Solitary plasmacytoma (SPC) accounts for 5% of plasma cell neoplasm. SPC of the spine is relatively rare, and SPC presenting with sudden paraplegia is very rare. A 53 year-old woman was admitted to our hospital complaining of sudden severe paraplegia and back pain. Emergency MRI revealed a tumor of the thoracic vertebra no. 10 (Th10). Pressure fracture of the Th10 was also recognized. The tumor was not osteolytic and invasion was recognized around the Th10. The tumor directly compressed the spinal cord. An excision of the tumor was performed under the clinical diagnosis of metastatic carcinoma. Pathologtically, the tumor consisted of plasmacytoid atypical cells with hyperchromatic nuclei. Histochemically, the tumor cells showed pyroninophilia. Immunohistochemically, the tumor cells were positive for λ-light chain, but negative for cytokeratin, epithelial mermbrane antigen, vimentin, CD45, CD20, CD45RO, κ-light chain, α-heavy chain, λ-heavy chain, μ-heavy chain, δ-heavy chain, ε-heavy chain, IgA, IgG, IgM, synaptophysin, chromogranin, S100 protein, desmin, α-smooth muscle antigen, myoglobin, and p53 protein. The Ki-67 labeling was 73%. The author diagnosed the tumor as SPC with λ-light chain disease. After the diagnosis, whole body CT and MRI revealed no other tumors. Blood and serum test revealed no significant changes; no M-protein was recognized. However, voided urine test revealed λ-light chain protein. The patient underwent fixation operation of TH10, and received radiation (50 Gray) and chemotherapy. No recurrence or transformation into myeloma occurred at the present time 25 months after the first manifestation. The present study indicated that pathological examination is an only clue to the diagnosis of SPC of the vertebra bone. C1 [Terada, Tadashi] Shizuoka City Shimizu Hospital, Department of Pathology, Miyakami 1231 Shimizu-Ku, 424-8636 Shizuoka, Japan. RP Terada, T (reprint author), Shizuoka City Shimizu Hospital, Department of Pathology, 424-8636 Shizuoka, Japan. EM piyo0111jp@yahoo.co.jp CR Grogan TM, Muller-Hermelink HK, Van Camp B, Harris NL, Kyle RA, 2001, Plasm cell neoplasms. In: Jaffe ES, Harris NL, Stein H, Vardiman JW, eds, World health organization classification of tumours. Pathology and genetics of tumours of haematopoietic and lymphoid tissues. IARC Press, Lyon, pp 142–156 Bacci G, Savini R, Calderoni P, Gnudi S, Minutllo A, Picci P, 1982, Solitary plasmacytoma of the vertebral column: a report of 15 cases. Tumori 30:271–275 Poor MM, Hitchon PW, Riggs CE, 1988, Solitary spinal plasmacytoma: management and outcome. J Spinal Disord 1:295–300 Colak A, Cataltepe O, Ozgen T, Erbengi A, 1989, Spinal cord compression caused by plasmacytoma: a retrospective review of 14 cases. Neurosurg Rev 12:305–308 Frassica DA, Fraqssica FJ, Schray MF, Sim FH, Kyle RA, 1989, Solitary plasmcytoma of bone: mayo clinic experience. Int J Radiat Oncol Biol Phys 16:43–48 McLain RF, Weinstein JN, 1989, Solitary plasmacytoma of the spine: a review of 84 cases. J Spiral Disord 2:69–74 Liebross RH, Ha CS, Cox JD, Weber D, Delasalle K, Alexanian R, 1998, Solitary bone plasmacytoma: outcome and prognostic factors after radiotherapy. Int J Radiat Oncol Biol Phys 41:1063–1067 Baba H, Maezawa Y, Furusawa N, Wada M, Kokuba Y, Imura S, Imamura Y, Yamada Y, 1998, Solitary plasmcytoma of the spine associated with neurological complications. Spinal Cord 36:470– 475 Dave BR, Nanda A, Anandjiwala JV, 2009, Transpedicular percutaneous biopsy of vertebral body lesions: a case series of 71 cases. Spinal Cord 47:384–389 Terada T, Kawaguchi M, Furukawa K, Sekido Y, Osamura RY, 2002, Minute mixed ductal-endocrine carcinoma of the pancreas with predominant intraductal growth. Pathol Int 52:740–746 Terada T, Kawaguchi M, 2005, Primary clear cell adenocarcinoma of the peritoneum. Tohoku J Exp Med 206:271–275 Terada T, Taniguchi M, 2004, Intraductal oncocytic papillary neoplasm of the liver. Pathol Int 54:116–123 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 167 EP 169 DI 10.1007/s12253-010-9292-4 PG 3 ER PT J AU Fukuyama, A Yokoyama, Y Futagami, M Shigeto, T Wada, R Mizunuma, H AF Fukuyama, Asami Yokoyama, Yoshihito Futagami, Masayuki Shigeto, Tatsuhiko Wada, Ryuichi Mizunuma, Hideki TI A Case of Uterine Leiomyoma with Intravenous Leiomyomatosis—Histological Investigation of the Pathological Condition SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE CD-34 antibody. Immunostaining; Intravenous leiomyomatosis; Pathological genesis; Uterine leiomyoma ID CD-34 antibody. Immunostaining; Intravenous leiomyomatosis; Pathological genesis; Uterine leiomyoma AB Intravenous leiomyomatosis (IVL) is generally defined as a histologically benign leiomyoma derived from a uterine myoma or intrauterine venous wall that has grown and extended intravenously. We here report on a single case of IVL, and investigate its pathological genesis. Regarding the part of the myoma extending to the vessel lumen, observations found the myoma to be pushing into the vessel. Immunostaining with CD34 antibody gave an image of the area where the myoma pushed into the vessel, showing CD34-positive vessel endothelium cells folded back into a layer covering the myoma, and continuing to line of the surface of the myoma within the vessel. Early pathological genesis of IVL was clarified for the first time that the tumor did not invade the vessel by breaking the venous wall, but rather advanced by stretching the vascular wall and progressing into the vein like a polyp, covered in endothelium cells. C1 [Fukuyama, Asami] Hirosaki University Graduate School of Medicine, Department of Obstetrics and Gynecology, 5-Zaifu-cho, Hirosaki, 036-8562 Aomori, Japan. [Yokoyama, Yoshihito] Hirosaki University Graduate School of Medicine, Department of Obstetrics and Gynecology, 5-Zaifu-cho, Hirosaki, 036-8562 Aomori, Japan. [Futagami, Masayuki] Hirosaki University Graduate School of Medicine, Department of Obstetrics and Gynecology, 5-Zaifu-cho, Hirosaki, 036-8562 Aomori, Japan. [Shigeto, Tatsuhiko] Hirosaki University Graduate School of Medicine, Department of Obstetrics and Gynecology, 5-Zaifu-cho, Hirosaki, 036-8562 Aomori, Japan. [Wada, Ryuichi] Hirosaki University Graduate School of Medicine, Department of Pathology and Molecular Medicine, 5-Zaifu-cho, Hirosaki, 036-8562 Aomori, Japan. [Mizunuma, Hideki] Hirosaki University Graduate School of Medicine, Department of Obstetrics and Gynecology, 5-Zaifu-cho, Hirosaki, 036-8562 Aomori, Japan. RP Yokoyama, Y (reprint author), Hirosaki University Graduate School of Medicine, Department of Obstetrics and Gynecology, 036-8562 Aomori, Japan. EM yokoyama@cc.hirosaki-u.ac.jp CR Norris HJ, Parmley T, 1975, Mesenchymal tumors of the uterus. Intravenous leiomyomatosis. A clinical and pathologic study of 14 cases. Cancer 36:2146–2178 Timmis AD, Smallpeice C, Davies AC et al, 1980, Intracardiac spread of intravenous leiomyomatosis with successful surgical excision. N Engl J Med 303:1043–1044 Suginami H, Kaura R, Ochi H et al, 1990, Intravenous leiomyomatosis with cardiac extension: successful surgical management and histopathological study. Obstet Gynecol 76:527–529 Lam PM, Lo KW, Yu MY et al, 2004, Intravenous leiomyomatosis: two cases with different routes of tumor extension. J Vasc Surg 39:465–469 Knauer E, 1903, Beitrag zur Anatomie der Uterusmyome. Beitr z Gynak 1:695 Sitzenfrey A, 1911, Ueber Venenmyome des Uterus mit intravaskularem. Ztschr f Geburtsh u Gynak 68:1 Merchant S, Malpica A, Deavers MT et al, 2002, Vessels within vessels in the myometrium. Am J Surg Pathol 26:232–236 Oki A, Yoshikawa H, 2007, Intravenous leiomyomatosis. Obstet Gynecol, Tokyo, 74:663–669, In Japanese Nam MS, Jeon MJ, Kim YT et al, 2003, Pelvic leiomyomatosis with intracaval and intracardiac extension: a case report and review of literature. Gynecol Oncol 89:175–180 Mitsuhashi A, Nagai Y, Sugita M et al, 1999, GnRH agonist for intravenous leiomyomatosis eith cardiac extension. A case report. J Reprod Med 44:883–886 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 171 EP 174 DI 10.1007/s12253-010-9265-7 PG 4 ER PT J AU Stojsic, J Stevic, R Kontic, M Stojsic, Z Drndarevic, N Bunjevacki, V Jekic, B AF Stojsic, Jelena Stevic, Ruza Kontic, Milica Stojsic, Zorica Drndarevic, Neda Bunjevacki, Vera Jekic, Biljana TI Large Cell Lung Carcinoma with Unusual Imaging Feature, Immunophenotype and Genetic Finding SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Large cell lung carcinoma; Immunophenotype; Imaging; Tumor genetic; Diagnosis ID Large cell lung carcinoma; Immunophenotype; Imaging; Tumor genetic; Diagnosis AB We present a case of large cell lung carcinoma in sixty-one year old male with typical lung cancer symptoms but unusual radiological presentation and immunophenotype. Tumor morphological finding related to its radiological finding was suggestive for large cell lymphoma or carcinoma, but its immunophenotype made confusion for pathological diagnosis. No p53 mutations were detected in genetic investigation. Multidisciplinar diagnostic approach to some tumors is useful for their final diagnosis. C1 [Stojsic, Jelena] Clinical Centre of Serbia, Institute for Lung Diseases and TuberculosisBelgrade, Serbia. [Stevic, Ruza] Clinical Centre of Serbia, Institute of RadiologyBelgrade, Serbia. [Kontic, Milica] Clinical Centre of Serbia, Institute for Lung Diseases and TuberculosisBelgrade, Serbia. [Stojsic, Zorica] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia. [Drndarevic, Neda] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia. [Bunjevacki, Vera] University in Belgrade, Medical Faculty, Institute for Human GeneticsBelgrade, Serbia. [Jekic, Biljana] University in Belgrade, Medical Faculty, Institute for Human GeneticsBelgrade, Serbia. RP Stojsic, J (reprint author), Clinical Centre of Serbia, Institute for Lung Diseases and Tuberculosis, Belgrade, Serbia. EM dr.jelenastoj@sezampro.rs CR Byrd RB, Carr DT, Miller WE, Payne WS, Woolen LB, 1968, Radiographic abnormalities in carcinoma of the lung as related to histological cell type. Thorax 24:573–575 Tan D, Zander DS, 2008, Immunohistochemistry for assessment of pulmonary and pleural neoplasms: a review and update. Int J Clin Exp Pathol 1:19–31 Travis WD, Brambilla B, Muller-Hermelink K, Harris CC, 2004, Pathology and genetics of tumors of the lung, pleura, thymus and heart. IARC, Lyon Franklin AW, 2000, Diagnosis of lung cancer. Pathology of invasive and preinvasive neoplasia. Chest 117:80s–89s Greenblatt MS, Bennett WP, Hollstein M, Harris CC, 1994, Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res 54:4855–4878 Gealy R, Zhang L, Siegfried JM, Luketich JD, Keohavong P, 1999, Comparison of mutations in the p53 and K-ras genes in lung carcinomas from smoking and nonsmoking women. Cancer Epidemiol Biomark Prev 8:297–302 Hernandez-Boussard TM, Hainaut P, 1998, A specific spectrum of p53 mutations in lung cancer from smokers: review of mutations compiled in the IARC p53 database. Environ Health Perspect 106:385–391 Gao WM, Mady HH, Yu GY et al, 2003, Comparison of p53 mutations between adenocarcinoma and squamous cell carcinoma of the lung: unique spectra involving G to A transitions and G to T transversions in both histological types. Lung Cancer 40:141– 150 Padua RA, Guinn BA, Al-Sabah AI, Smith M, Taylor C, Pettersson T, Ridge S, Carter G, White D, Oscier D, Chevret S, West R, 1998, RAS, FMS and p53 mutations and poor clinical outcome in myelodysplasias: a 10-year follow-up. Leukemia 12:887–892 Li Z, Wu J, Yang D, Zhang L, 2006, Large cell carcinoma of lung: analysis of CT signs and review of the literature. Chin Ger J Clin Oncol 5(5):309–311 Akata S, Okada S, Maeda J, Park J, Yoshimura M, Saito K, Kakizaki D, Abe K, Kato H, 2007, Computed tomographic findings of large cell neuroendocrine carcinoma of the lung. Clin Imaging 31(6):379–384 Stojsic J, Milenkovic B, Radojicic J, Percinkovski M, 2007, Alveolar adenoma—a rare lung tumor. Srp Arh Celokup Lek 135, 7–8):569–571 [Serbian] Stojsic J, Milenkovic B, Radojicic J, Percinkovski M, 2007, Pulmonary sclerosing hemangioma—case report. Srp Arh Celokup Lek 135(9–10):461–464 [Serbian] Gustafsson BI, Kidd M, Chan A, Malfertheiner MV, Modli IM, 2008, Bronchopulmonary neuroendocrine tumors. Cancer 113:5– 21 Faggiano A, Sabourin J-C, Lumbroso J, Duvillard P, Leboulleux S, Dromain C, Colao A, Schlumberger M, Baudin E, 2007, Pulmonary and extrapulmonary poorly differentiated large cell neuroendocrine carcinomas.Diagnostic and prognostic features. Cancer 110(2):265–274 Maddau C, Confortini M, Bisanzi S, Janni A, Montinaro F, Paci E, Pontenani G, Rulli P, Salani A, Zappa M, Benvenuti A, Carozzi FM, 2006, Prognostic significance of p53 and Ki-67 antigen expression in surgically treated non-small cell lung cancer. Immunocytochemical detection with imprint cytology. Am J Clin Pathol 125:425–431 Bosari S, Viale G, 1995, The clinical significance of p53 aberrations in human tumors. Virchows Arch 427:229–241 Takahashi T, Munakata M, Ohtsuka Y, 2002, Expression and alteration of Ras and P53 proteins in patients with lung carcinoma accompanied by idiopathic pulmonary fibrosis. Cancer 95(3):624– 633 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2011 VL 17 IS 1 BP 175 EP 179 DI 10.1007/s12253-010-9272-8 PG 5 ER PT J AU Zaravinos, A Chatziioannou, M Lambrou, IG Boulalas, I Delakas, D Spandidos, AD AF Zaravinos, Apostolos Chatziioannou, Maria Lambrou, I George Boulalas, Ioannis Delakas, Dimitris Spandidos, A Demetrios TI Implication of RAF and RKIP Genes in Urinary Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE RAF family genes; RKIP; Microarrays; qPCR; Computational analysis; Transitional cell carcinoma of the urinary bladder ID RAF family genes; RKIP; Microarrays; qPCR; Computational analysis; Transitional cell carcinoma of the urinary bladder AB RKIP has been shown to regulate the RAS-RAFMEK-ERK kinase cascade acting as modulator of apoptosis and metastasis in prostate cancer. Our goal was to examine the expression of the RAF (A-RAF, B-RAF and RAF-1) and RKIP genes in urinary bladder cancer. Microarray analysis and qPCR was employed to investigate the expression of RAF and RKIP, in 30 patients with transitional cell carcinoma (TCC) of the urinary bladder vs. the corresponding levels of adjacent normal tissue. Computational analysis was also performed on Gene Expression Omnibus (GEO) datasets, to unravel differences in the expression of RAF or RKIP between tumor and control samples, and between superficial and muscle invasive tumors. Microarray analysis revealed >2-fold expression of BRAF and RKIP in T2, T3, grade III tumors vs. controls. B-RAF over-expression was verified by qPCR in pT1, grade III tumors vs. their normal counterparts (p=0.016). qPCR revealed a significant RKIP reduction in TCC vs. normal tissue (p=0.002 and p<0.001 for T1, grade II and Ta-T1, grade III, respectively); All RAF genes were positively correlated among each other (A-RAF/B-RAF, p=0.003; A-RAF/RAF-1, p<0.001; B-RAF/RAF-1, p=0.050), whereas B-RAF was negatively correlated with RKIP in TCC (p=0.050). Further computational analysis revealed different expression profiles for the genes of interest, among muscle invasive carcinomas, superficial TCCs, cystectomy specimens and normal tissue. The reduced RKIP mRNA levels in TCC and the elevated levels of B-RAF in pT1, grade III tumors vs. normal tissue, corroborate that these genes are involved in the pathogenesis of urinary bladder cancer. C1 [Zaravinos, Apostolos] University of Crete, Medical School, Department of Virology, 71110 Heraklion, Crete, Greece. [Chatziioannou, Maria] University of Crete, Medical School, Department of Virology, 71110 Heraklion, Crete, Greece. [Lambrou, I George] University of Athens, 1st Department of Pediatrics, Goudi, 11527 Athens, Greece. [Boulalas, Ioannis] University of Crete, Medical School, Department of Virology, 71110 Heraklion, Crete, Greece. [Delakas, Dimitris] Department of Urology, VoulaAthens, Greece. [Spandidos, A Demetrios] University of Crete, Medical School, Department of Virology, 71110 Heraklion, Crete, Greece. RP Spandidos, AD (reprint author), University of Crete, Medical School, Department of Virology, 71110 Heraklion, Greece. EM spandidos@spandidos.gr CR Jemal A, Siegel R, Ward E et al, 2008, Cancer statistics. CA Cancer J Clin 58:71–96 Lopez-Beltran A, Montironi R, 2004, Non-invasive urothelial neoplasms: according to the most recent WHO classification. Eur Urol 46:170–176 O’Neill E, Kolch W, 2004, Conferring specificity on the ubiquitous Raf/MEK signalling pathway. Br J Cancer 90:283–288 Avruch J, Khokhlatchev A, Kyriakis JM et al, 2001, Ras activation of the Raf kinase: tyrosine kinase recruitment of the MAP kinase cascade. Recent Prog Horm Res 56:127–155 KolchW(2000, Meaningful relationships: the regulation of the Ras/ Raf/MEK/ERK pathway by protein interactions. Biochem J 351(Pt 2):289–305 Storm SM, Cleveland JL, Rapp UR, 1990, Expression of raf family proto-oncogenes in normal mouse tissues. Oncogene 5:345–351 Barnier JV, Papin C, Eychene A, Lecoq O, Calothy G, 1995, The mouse B-raf gene encodes multiple protein isoforms with tissuespecific expression. J Biol Chem 270:23381–23389 Luckett JC, Huser MB, Giagtzoglou N, Brown JE, Pritchard CA, 2000, Expression of the A-raf proto-oncogene in the normal adult and embryonic mouse. Cell Growth Differ 11:163–171 Simister PC, Banfield MJ, Brady RL, 2002, The crystal structure of PEBP-2, a homologue of the PEBP/RKIP family. Acta Crystallogr D Biol Crystallogr 58:1077–1080 Odabaei G, Chatterjee D, Jazirehi AR, Goodglick L, Yeung K, Bonavida B, 2004, Raf-1 kinase inhibitor protein: structure, function, regulation of cell signaling, and pivotal role in apoptosis. Adv Cancer Res 91:169–200 Yeung K, Seitz T, Li S et al, 1999, Suppression of Raf-1 kinase activity and MAP kinase signalling by RKIP. Nature 401:173–177 Park S, Yeung ML, Beach S, Shields JM, Yeung KC, 2005, RKIP downregulates B-Raf kinase activity in melanoma cancer cells. Oncogene 24:3535–3540 Fu Z, Smith PC, Zhang L et al, 2003, Effects of raf kinase inhibitor protein expression on suppression of prostate cancer metastasis. J Natl Cancer Inst 95:878–889 SchuiererMM, Bataille F, Hagan S, KolchW, Bosserhoff AK, 2004, Reduction in Raf kinase inhibitor protein expression is associated with increased Ras-extracellular signal-regulated kinase signaling in melanoma cell lines. Cancer Res 64:5186–5192 Van Gelder RN, von ZastrowME, Yool A, DementWC, Barchas JD, Eberwine JH, 1990)Amplified synthesized from limited quantities of heterogeneous cDNA. Proc Natl Acad Sci USA 87:1663–1667 Diez Alvarez DR, Dopazo A, 2007, Codelink: an R package for analysis of GE healthcare gene expression bioarrays. Bioinformatics 23:1168–1169 Altman NS, Hua J, 2006, Extending the loop design for two-channel microarray experiments. Genet Res 88:153–163 Sturn A, Quackenbush J, Trajanoski Z, 2002, Genesis: cluster analysis of microarray data. Bioinformatics 18:207–208 Boulalas I, Zaravinos A, Karyotis I, Delakas D, Spandidos DA, 2009, Activation of RAS family genes in urothelial carcinoma. J Urol 181:2312–2319 Dyrskjot L, KruhofferM, Thykjaer Tet al, 2004, Gene expression in the urinary bladder: a common carcinoma in situ gene expression signature exists disregarding histopathological classification. Cancer Res 64:4040–4048 Mengual L, Burset M, Ars E et al, 2009, DNA microarray expression profiling of bladder cancer allows identification of noninvasive diagnostic markers. J Urol 182:741–748 Davies H, Bignell GR, Cox C et al, 2002, Mutations of the BRAF gene in human cancer. Nature 417:949–954 Zlobec I, Kovac M, Erzberger P et al, 2010, Combined analysis of specific KRAS mutation, BRAF and microsatellite instability identifies prognostic subgroups of sporadic and hereditary colorectal cancer. Int J Cancer Feb 16, 2010 Zlobec I, Bihl MP, Schwarb H, Terracciano L, Lugli A, 2009, Clinicopathological and protein characterization of BRAF- and KRAS- mutated colorectal cancer and implications for prognosis. Int J Cancer Nov 11, 2009 Libra M, Malaponte G, Navolanic PM et al, 2005, Analysis of BRAF mutation in primary and metastatic melanoma. Cell Cycle 10:1382–1384 Baldus SE, Schaefer KL, Engers R, Hartleb D, Stoecklein NH, Gabbert HE, 2010, Prevalence and heterogeneity of KRAS, BRAF, and PIK3CA mutations in primary colorectal adenocarcinomas and their corresponding metastases. Clin Cancer Res 16:790–799 Boulalas I, Zaravinos A, Delakas D, Spandidos DA, 2009, Mutational analysis of the BRAF gene in transitional cell carcinoma of the bladder. Int J Biol Markers 24:17–21 Zaravinos A, Kanellou P, Baritaki S, Bonavida B, Spandidos DA, 2009, BRAF and RKIP are significantly decreased in cutaneous squamous cell carcinoma. Cell Cycle 8:1402–1408 Zaravinos A, Bizakis J, Spandidos DA, 2008, RKIP and BRAF aberrations in human nasal polyps and the adjacent turbinate mucosae. Cancer Lett 264:288–298 Gattenlohner S, Etschmann B, Riedmiller H, Muller-Hermelink HK, 2009, Lack of KRAS and BRAF mutation in renal cell carcinoma. Eur Urol 55:1490–1491 Karlou M, Saetta AA, Korkolopoulou P et al, 2009, Activation of extracellular regulated kinases, ERK1/2, predicts poor prognosis in urothelial bladder carcinoma and is not associated with B-Raf gene mutations. Pathology 41:327–334 Mhawech-Fauceglia P, Fischer G, Beck A, Cheney RT, Herrmann FR, 2006, Raf1, Aurora-A/STK15 and E-cadherin biomarkers expression in patients with pTa/pT1 urothelial bladder carcinoma; a retrospective TMA study of 246 patients with long-term follow-up. Eur J Surg Oncol 32:439–444 McPhillips F, Mullen P, MacLeod KG et al, 2006, Raf-1 is the predominant Raf isoform that mediates growth factor-stimulated growth in ovarian cancer cells. Carcinogenesis 27:729–739 Simon R, Richter J, Wagner U et al, 2001, High-throughput tissue microarray analysis of 3p25, RAF1, and 8p12, FGFR1, copy number alterations in urinary bladder cancer. Cancer Res 61:4514– 4519 O’Neill E, Rushworth L, Baccarini M, Kolch W, 2004, Role of the kinase MST2 in suppression of apoptosis by the protooncogene product Raf-1. Science 306:2267–2270 Ehrenreiter K, Piazzolla D, Velamoor Vet al, 2005, Raf-1 regulates Rho signaling and cell migration. J Cell Biol 168:955–964 Wan PT, Garnett MJ, Roe SM, Lee S et al, 2004, Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 116:855–867 Keller ET, Fu Z, Yeung K, Brennan M, 2004, Raf kinase inhibitor protein: a prostate cancer metastasis suppressor gene. Cancer Lett 207:131–137 Fu Z, Kitagawa Y, Shen R et al, 2006, Metastasis suppressor gene Raf kinase inhibitor protein, RKIP, is a novel prognostic marker in prostate cancer. Prostate 66:248–256 Blaveri E, Brewer JL, Roydasgupta R et al, 2005, Bladder cancer stage and outcome by array-based comparative genomic hybridization. Clin Cancer Res 11:7012–7022 Keller ET, Fu Z, Brennan M, 2005, The biology of a prostate cancer metastasis suppressor protein: Raf kinase inhibitor protein. J Cell Biochem 94:273–278 Woods Ignatoski KM, Grewal NK, Markwart SM et al, 2008, Loss of Raf kinase inhibitory protein induces radioresistance in prostate cancer. Int J Radiat Oncol Biol Phys 72:153–160 Li HZ, Wang Y, Gao Y et al, 2008, Effects of raf kinase inhibitor protein expression on metastasis and progression of human epithelial ovarian cancer. Mol Cancer Res 6:917–928 Lou TF, Gray CW, Gray DM, 2003, The reduction of Raf-1 protein by phosphorothioate ODNs and siRNAs targeted to the same two mRNA sequences. Oligonucleotides 13:313–324 Chatterjee D, Bai Y, Wang Z et al, 2004, RKIP sensitizes prostate and breast cancer cells to drug-induced apoptosis. J Biol Chem 279:17515–17523 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 181 EP 190 DI 10.1007/s12253-010-9295-1 PG 10 ER PT J AU Guzman, G Chennuri, R Voros, A Boumendjel, R Locante, A Patel, R Valyi-Nagy, T AF Guzman, Grace Chennuri, Rohini Voros, Andras Boumendjel, Redouane Locante, Alberto Patel, Roshan Valyi-Nagy, Tibor TI Nucleometric Study of Anisonucleosis, Diabetes and Oxidative Damage in Liver Biopsies of Orthotopic Liver Transplant Recipients with Chronic Hepatitis C Virus Infection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Diabetes; Hepatitis C virus (HCV); Hepatocyte anisonucleosis; Image analysis; Nucleometry; Oxidative damage; 8-hydroxy-2′deoxyguanosine (8-OHdG) immunomarker ID Diabetes; Hepatitis C virus (HCV); Hepatocyte anisonucleosis; Image analysis; Nucleometry; Oxidative damage; 8-hydroxy-2′deoxyguanosine (8-OHdG) immunomarker AB Anisonucleosis is defined as a morphological manifestation of nuclear injury characterized by variation in the size of the cell nuclei. It has been described in variety of benign conditions and is most pronounced in dysplasia and malignancy. To better understand the pathogenesis of anisonucleosis in liver diseases, this study focused on hepatocyte anisonucleosis in biopsies of liver transplant recipients who developed recurrent chronic hepatitis C virus (HCV) infection. Post transplant surveillance liver biopsy specimens were evaluated employing light microscopy, immunohistochemistry, digital image analysis, and nucleometry for histopathological analyses, measurement of nuclear size, and quantification of tissue expression of oxidative marker 8-hydroxy-2′deoxyguanosine (8-OHdG). Our aim in this study was to determine whether there were any independent associations between hepatocyte anisonucleosis and various clinicopathological parameters. These features included patient age, body mass index, gender, race, donor age, live versus cadaveric donor status, history of diabetes mellitus, history of tacrolimus and cyclosporine therapy, duration post transplant and parameters of hepatitis activity index, fibrosis index, steatosis, and oxidative tissue damage in formalin fixed paraffin embedded (FFPE) liver biopsies as determined by immunohistochemistry using 8-OHdG, an indicator of hydroxyl radical mediated tissue damage. Our findings suggested that in liver transplant recipients with recurrent chronic HCV infection, hepatocyte anisonucleosis is more pronounced in individuals with diabetes mellitus (p=0.0016), and among those who have heightened hepatic expression of the oxidative damage marker 8-OHdG (p=0.0053). Further studies are necessary to determine whether anisonucleosis is an independent marker for diabetes or oxidative damage. C1 [Guzman, Grace] University of Illinois at Chicago, College of Medicine, Department of Pathology, Room 130, 840 South Wood Street, M/C 847, 60612 Chicago, IL, USA. [Chennuri, Rohini] University of Illinois at Chicago, College of Medicine, Department of Pathology, Room 130, 840 South Wood Street, M/C 847, 60612 Chicago, IL, USA. [Voros, Andras] University of Illinois at Chicago, College of Medicine, Department of Pathology, Room 130, 840 South Wood Street, M/C 847, 60612 Chicago, IL, USA. [Boumendjel, Redouane] University of Illinois at Chicago, College of Medicine, Department of Pathology, Room 130, 840 South Wood Street, M/C 847, 60612 Chicago, IL, USA. [Locante, Alberto] University of Illinois at Chicago, College of Medicine, Department of Pathology, Room 130, 840 South Wood Street, M/C 847, 60612 Chicago, IL, USA. [Patel, Roshan] University of Illinois at Chicago, College of Medicine, Department of Pathology, Room 130, 840 South Wood Street, M/C 847, 60612 Chicago, IL, USA. [Valyi-Nagy, Tibor] University of Illinois at Chicago, College of Medicine, Department of Pathology, Room 130, 840 South Wood Street, M/C 847, 60612 Chicago, IL, USA. RP Guzman, G (reprint author), University of Illinois at Chicago, College of Medicine, Department of Pathology, 60612 Chicago, USA. EM GraceGuz@uic.edu CR Crawford Jm, 2004, The liver and biliary tree. Kumar VFN, Abbas A, eds). Philadelphia, WB Saunders, pp 877–937 Henmi A, Uchida T, Shikata T, 1985, Karyometric analysis of liver cell dysplasia and hepatocellular carcinoma. Evidence against precancerous nature of liver cell dysplasia. Cancer 55:2594–2599 Zeppa P, Zabatta A, Fulciniti F, Vetrani A, Di Benedetto G, De Rosa G, Palombini L, 1988, The role of morphometry in the cytology of well-differentiated hepatocarcinoma and cirrhosis with atypia. Anal Quant Cytol Histol 10:343–348 Kuo SH, Lai MY, Liu YR, Lee YT, Chen DS, Lee CS, Hsu HC, 1994, Nuclear area and DNA content in tumor and nontumor portions of hepatocellular carcinoma. Anal Quant Cytol Histol 16:153–158 Koutselini H, Lazaris AC, Kavantzas N, Kiritsi T, Davaris PS, 2000, Significance of nuclear morphometry as a diagnostic tool in fine-needle aspirates of the liver. Eur J Gastroenterol Hepatol 12:913–921 Wood LJ, Mundo F, Searle J, Powell LW, 1985, Sulindac hepatotoxicity: effects of acute and chronic exposure. Aust NZ J Med 15:397–401 Chadee K, Meerovitch E, 1984, The pathogenesis of experimentally induced amebic liver abscess in the gerbil, Meriones unguiculatus). Am J Pathol 117:71–80 Giri RK, Das BR, 1996, Differential expression of c-jun and c-myc in N-nitroso diethylamine-induced hepatic oncogenesis in AKR mice. Cancer Lett 109:121–127 Wise M, Bialek S, Finelli L, Bell BP, Sorvillo F, 2008, Changing trends in hepatitis C-related mortality in the United States, 1995– 2004. Hepatology 47:1128–1135 El-Serag HB, Hampel H, Javadi F, 2006, The association between diabetes and hepatocellular carcinoma: a systematic review of epidemiologic evidence. Clin Gastroenterol Hepatol 4:369–380 El-Serag HB, Tran T, Everhart JE, 2004, Diabetes increases the risk of chronic liver disease and hepatocellular carcinoma. Gastroenterology 126:460–468 Cui J, Holmes EH, Greene TG, Liu PK, 2000, Oxidative DNA damage precedes DNA fragmentation after experimental stroke in rat brain. FASEB J 14:955–967 Valyi-Nagy T, Dermody TS, 2005, Role of oxidative damage in the pathogenesis of viral infections of the nervous system. Histol Histopathol 20:957–967 Cotler SJ, Kallwitz E, Tencate V, Bhushan A, Berkes J, Benedetti E, Layden-Almer J, Layden TJ, Valyi-Nagy T, Guzman G, 2007, Diabetes and hepatic oxidative damage are associated with hepatitis C progression after liver transplantation. Transplantation 84:587–591 Everhart JE, Wei Y, Eng H, Charlton MR, Persing DH, Wiesner RH, Lake JR, Germer JJ, Zetterman RK, Hoofnagle JH, 1999, Recurrent and new hepatitis C virus infection after liver transplantation. Hepatology 29:1220–1226 Lefkowitch JH, 2009, Recent developments in liver pathology. Hum Pathol 40:445–455 Batts KP, Ludwig J, 1995, Chronic hepatitis. An update on terminology and reporting. Am J Surg Pathol 19:1409–1417 Demetris AJ, Adeyi O, Bellamy CO, Clouston A, Charlotte F, Czaja A, Daskal I, El-Monayeri MS, Fontes P, Fung J, Gridelli B, Guido M, Haga H, Hart J, Honsova E, Hubscher S, Itoh T, Jhala N, Jungmann P, Khettry U, Lassman C, Ligato S, Lunz JG 3rd, Marcos A, Minervini MI, Molne J, Nalesnik M, Nasser I, Neil D, Ochoa E, Pappo O, Randhawa P, Reinholt FP, Ruiz P, Sebagh M, Spada M, Sonzogni A, Tsamandas AC, Wernerson A, Wu T, Yilmaz F, 2006, Liver biopsy interpretation for causes of late liver allograft dysfunction. Hepatology 44:489–501 Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, Cummings OW, Ferrell LD, Liu YC, Torbenson MS, Unalp-Arida A, Yeh M, Mccullough AJ, Sanyal AJ, 2005, Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 41:1313–1321 Anthony PP, Vogel CL, Barker LF, 1973, Liver cell dysplasia: a premalignant condition. J Clin Pathol 26:217–223 Anthony PP, 1976, Precursor lesions for liver cancer in humans. Cancer Res 36:2579–2583 Guzman G, Wu SJ, Kajdacsy-Balla A, Cotler SJ, 2006, Alphamethylacyl- CoA racemase, AMACR/P504S, can distinguish hepatocellular carcinoma and dysplastic hepatocytes from benign nondysplastic hepatocytes. Appl Immunohistochem Mol Morphol 14:411–416 Luo J, Zha S, Gage WR, Dunn TA, Hicks JL, Bennett CJ, Ewing CM, Platz EA, Ferdinandusse S, Wanders RJ, Trent JM, Isaacs WB, De Marzo AM, 2002, Alpha-methylacyl-CoA racemase: a new molecular marker for prostate cancer. Cancer Res 62:2220– 2226 Jiang Z, Wu CL, Woda BA, Iczkowski KA, Chu PG, Tretiakova MS, Young RH, Weiss LM, Blute RD Jr, Brendler CB, Krausz T, Xu JC, Rock KL, Amin MB, Yang XJ, 2004, Alpha-methylacyl- CoA racemase: a multi-institutional study of a new prostate cancer marker. Histopathology 45:218–225 Poynard T, Bedossa P, Opolon P, 1997, Natural history of liver fibrosis progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, CLINIVIR, and DOSVIRC groups. Lancet 349:825–832 Russack V, Vass L, Gupta PK, 1993, Comparison of morphologic features of benign hepatocytes associated with nonmalignant and malignant liver lesions. Acta Cytol 37:153–157 Gong G, Waris G, Tanveer R, Siddiqui A, 2001, Human hepatitis C virus NS5A protein alters intracellular calcium levels, induces oxidative stress, and activates STAT-3 and NF-kappa B. Proc Natl Acad Sci USA 98:9599–9604 Abdalla MY, Ahmad IM, Spitz DR, Schmidt WN, Britigan BE, 2005, Hepatitis C virus-core and non structural proteins lead to different effects on cellular antioxidant defenses. J Med Virol 76:489–497 Evans JL, Maddux BA, Goldfine ID, 2005, The molecular basis for oxidative stress-induced insulin resistance. Antioxid Redox Signal 7:1040–1052 Poli G, 2000, Pathogenesis of liver fibrosis: role of oxidative stress. Mol Aspects Med 21:49–98 Davila JA, Morgan RO, Shaib Y, Mcglynn KA, El-Serag HB, 2005, Diabetes increases the risk of hepatocellular carcinoma in the United States: a population based case control study. Gut 54:533–539 Campbell JS, Johnson MM, Bauer RL, Hudkins KL, Gilbertson DG, Riehle KJ, Yeh MM, Alpers CE, Fausto N, 2007, Targeting stromal cells for the treatment of platelet-derived growth factor C-induced hepatocellular carcinogenesis. Differentiation 75:843–852 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 191 EP 199 DI 10.1007/s12253-010-9296-0 PG 9 ER PT J AU Lee, JH Ryu, TY Cho, ChH Kim, DK AF Lee, Jae-Ho Ryu, Tae-Yung Cho, Chi-Heum Kim, Dae-Kwang TI Different Characteristics of Mitochondrial Microsatellite Instability Between Uterine Leiomyomas and Leiomyosarcomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Genetic instability; Mitochondrial microsatellite instability; Uterine leiomyoma; Uterine leiomyosarcomas ID Genetic instability; Mitochondrial microsatellite instability; Uterine leiomyoma; Uterine leiomyosarcomas AB Uterine leiomyomas are benign tumors of the uterus that arise clonally from smooth muscle cells of the myometrium and are the most common reason for hysterectomies. The aim of this study was to evaluate mitochondrial microsatellite instability (mtMSI) in uterine leiomyomas and leiomyosarcomas to clarify the molecular pathogenetic distinction between these tumors. DNA was isolated from paired normal and tumoral tissues in 50 patients with uterine leiomyomas and 14 patients with leiomyosarcomas. mtMSI was analyzed by using eight microsatellite markers. Our result showed that mitochondrial microsatellite instability was not found in all uterine leiomyomas. However, 3 (21.4%) of 14 patients with leiomyosarcomas had mtMSI and the frequencies of mtMSI in these tumors were significantly different (p<0.01). Distinctive characteristics of mitochondrial genetic instability in uterine leiomyomas and leiomyosarcomas suggested the potential of mtMSI as a marker for differential diagnosis between them. C1 [Lee, Jae-Ho] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeoldaero, Dalseo-Gu, 2800 Daegu, South Korea. [Ryu, Tae-Yung] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeoldaero, Dalseo-Gu, 2800 Daegu, South Korea. [Cho, Chi-Heum] Keimyung University, School of Medicine, Department of Obstetrics and GynecologyDaegu, South Korea. [Kim, Dae-Kwang] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeoldaero, Dalseo-Gu, 2800 Daegu, South Korea. RP Kim, DK (reprint author), Keimyung University, School of Medicine, Department of Anatomy, 2800 Daegu, South Korea. EM dkkimmd@kmu.ac.kr CR Cramer SF, Patel A, 1990, The frequency of uterine leiomyomas. Am J Clin Pathol 94:435–438 Parker WH, 2007, Etiology, symptomatology, and diagnosis of uterine myomas. Fertil Steril 87:725–736 Walker CL, Stewart EA, 2005, Uterine fibroids: the elephant in the room. Science 308:1589–1592 Chen L, Yang B, 2008, Immunohistochemical analysis of p16, p53, and Ki-67 expression in uterine smooth muscle tumors. Int J Gynecol Pathol 27:326–332 Packenham JP, du Manoir S, Schrock E et al, 1997, Analysis of genetic alterations in uterine leiomyomas and leiomyosarcomas by comparative genomic hybridization. Mol Carcinog 19:273–279 Thibodeau S, Bren G, Schaid D, 1993, Microsatellite instability in cancer of the proximal colon. Science 75:1027–1038 Brenmall T, 1995, Microsatellite instability—Shifting concepts in tumorigenesis. Am J Path 147:561–563 Boland CR, Thibodeau SN, Hamilton SR et al, 1998, A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58:5248–5257 Naidoo R, Chetty R, 1998, The application of microsatellites in molecular pathology. Pathol Oncol Res 4:310–315 Quade BJ, Pinto AP, Howard DR et al, 1999, Frequent loss of heterozygosity for chromosome 10 in uterine leiomyosarcoma in contrast to leiomyoma. Am J Pathol 154:945–950 Suwa K, Ohmori M, Miki H, 1999, Microsatellite alterations in various sarcomas in Japanese patients. J Orthop Sci 4:223–230 Amant F, Dorfling CM, Dreyer L et al, 2001, Microsatellite instability in uterine sarcomas. Int J Gynecol Cancer 11:218– 223 Howell N, Kubacka I, Mackey DA, 1996, How rapidly does the humanmitochondrial genome evolve?AmJ HumGenet 59:501–509 Paabo S, 1996, Mutational hot spots in the mitochondrial microcosm. Am J Hum Genet 59:493–496 Khrapko K, Coller HA, Andre PC et al, 1997, Mitochondrial mutational spectra in human cells and tissues. Proc Natl Acad Sci USA 94:13798–13803 Richard SM, Bailliet G, Paez GL et al, 2000, Nuclear and mitochondrial genome instability in human breast cancer. Cancer Res 60:4231–4237 Bianchi NO, Bianchi MS, Richard SM, 2001, Mitochondrial genome instability in human cancers. Mutat Res 488:9–23 Wang Y, Liu VW, Ngan HY et al, 2005, Frequent occurrence of mitochondrial microsatellite instability in the D-loop region of human cancers. Ann N Y Acad Sci 1042:123–129 Wang Y, Liu VW, Tsang PC et al, 2006, Microsatellite instability in mitochondrial genome of common female cancers. Int J Gynecol Cancer 16:259–266 Habano W, Nakamura S, Sugai T, 1998, Microsatellite instability in the mitochondrial DNA of colorectal carcinomas: evidence for mismatch repair systems in mitochondrial genome. Oncogene 17:1931–1937 Lee JH, Choi IJ, Song DK et al, 2010, Genetic instability in the human lymphocyte exposed to hypoxia. Cancer Genet Cytogenet 196:83–88 Jeong CW, Lee JH, Sohn SS et al, 2010)Mitochondrialmicrosatellite instability in gastric cancer and gastric epithelial dysplasia as a precancerous lesion. Cancer Epidemiol 34:323–327 Bell SW, Kempson RL, HendricksonMR, 1994, Problematic uterine smooth muscle neoplasms. A clinicopathologic study of 213 cases. Am J Surg Pathol 18:535–558 Ha TW, Han KH, Son DG et al, 2005, Analysis of loss of heterozygosity in Korean patients with keratoacanthoma. J Korean Med Sci 20:340–343 Fletcher JA, Morton CC, Paelka K et al, 1990, Chromosome aberration in uterine smooth muscle tumors: potential diagnostic relevance of cytogenetic instability. Cancer Res 50:4092–4097 Laxman R, Currie JL, Kurman RJ et al, 1993, Cytogenetic profile of uterine sarcomas. Cancer 71:1283–1288 Sreekantaiah C, Davis JR, Sandberg AA, 1993, Chromosomal abnormalities in leiomyosarcomas. Am J Pathol 142:293–305 Pandis N, Heim S, Bardi G et al, 1991, Chromosome analysis of 96 uterine leiomyomas. Cancer Genet Cytogenet 55:11–18 Heim S, Nilbert M, Vanni R et al, 1988, A specific translocation, t, 12;14)(q14–15;q23–24), characterizes a subgroup of uterine leiomyomas. Cancer Genet Cytogenet 32:13–17 Ozisik YY, Meloni AM, Surti U et al, 1993, Deletion 7q22 in uterine leiomyoma: a cytogenetic review. Cancer Genet Cytogenet 71:1–6 Mittal KR, Chen F, Wei JJ et al, 2009, Molecular and immunohistochemical evidence for the origin of uterine leiomyosarcomas from associated leiomyoma and symplastic leiomyoma-like areas. Mod Pathol 22:1303–1311 French D, Cermele C, Lombardi AM et al, 1998, Microsatellite alterations in uterine leiomyomas. Anticancer Res 18:349–352 Risinger JI, Umar A, Boyer JC et al, 1995, Microsatellite instability in gynecological sarcomas and in hMSH2 mutant uterine sarcoma cell lines defective in mismatch repair activity. Cancer Res 55:5664–5669 Stoneking M, 2000, Hypervariable sites in the mtDNA control region are mutational hotspots. Am J Hum Genet 67:1029–1032 Lievre A, Chapusot C, Bouvier AM et al, 2005, Clinical value of mitochondrial mutations in colorectal cancer. J Clin Oncol 23:3517–3525 Lee HC, Yin PH, Lin JC et al, 2005, Mitochondrial genome instability and mtDNA depletion in human cancers. Ann N Y Acad Sci 104:109–122 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 201 EP 205 DI 10.1007/s12253-010-9297-z PG 5 ER PT J AU Kovacs, Toth, L Glavak, Cs Lakosi, F Hadjiev, J Bajzik, G Vandulek, Cs Repa, I AF Kovacs, Arpad Toth, Lilla Glavak, Csaba Lakosi, Ferenc Hadjiev, Janaki Bajzik, Gabor Vandulek, Csaba Repa, Imre TI Integrating Functional MRI Information into Radiotherapy Planning of CNS Tumors-Early Experiences SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CNS tumor; fMRI; Radiotherapy ID CNS tumor; fMRI; Radiotherapy AB The purpose of our study was to examine the integration of functional MRI (fMRI) information into 3D based planning process of the central nervous system (CNS) malignancies. Between 01.01.2008 and 01.12.2008 four patients with astrocytoma were enrolled to this study. Before the planning process conventional planning CT, postoperative MR and individual functional MRI examinations were delivered. For the functional MRI examination the following four types of stimulus were applied: acoustic, visual, somatosensory and numeral. Three different theoretical planning situations were applied and compared: 3D conformal plan without fMRI information, 3D conformal plan with fMRI information and IMRT plan with fMRI information. For plan comparison DVH analysis, and NTCP model were used. fMRI based OR definition resulted in 4 additional OR’s in the contouring process. As these cases demonstrate, an average of 50% dose reduction was achieved in OR, OR2 and OR3 with IMRT and fMRI based 3D planning, especially in case of midline localization and big tumor extent. IMRT provides additional sparing effect in the optic tract and brainstem, especially for localizations close to the midline. Our results demonstrated that using fMRI information in conventional 3D based treatment planning potentially benefits significant dose reduction in critical organs, with no compromise in PTV coverage. fMRI can be widely used even in low grade cases (long life expectancies, lower acute and late toxicity in radiotherapy) and in cases with high grade astrocytomas or metastases (higher dose to PTV with better risk organ sparing in radiotherapy). C1 [Kovacs, Arpad] Kaposvar University, Department of Radiation Oncology, Guba S Street 40, 7400 Kaposvar, Hungary. [Toth, Lilla] Kaposvar University, Department of Radiation Oncology, Guba S Street 40, 7400 Kaposvar, Hungary. [Glavak, Csaba] Kaposvar University, Department of Radiation Oncology, Guba S Street 40, 7400 Kaposvar, Hungary. [Lakosi, Ferenc] Kaposvar University, Department of Radiation Oncology, Guba S Street 40, 7400 Kaposvar, Hungary. [Hadjiev, Janaki] Kaposvar University, Department of Radiation Oncology, Guba S Street 40, 7400 Kaposvar, Hungary. [Bajzik, Gabor] Kaposvar University, Department of Radiation Oncology, Guba S Street 40, 7400 Kaposvar, Hungary. [Vandulek, Csaba] Kaposvar University, Department of Radiation Oncology, Guba S Street 40, 7400 Kaposvar, Hungary. [Repa, Imre] Kaposvar University, Department of Radiation Oncology, Guba S Street 40, 7400 Kaposvar, Hungary. RP Kovacs, (reprint author), Kaposvar University, Department of Radiation Oncology, 7400 Kaposvar, Hungary. EM kovacs.arpad@sic.hu CR Alvarez R, Liney G, Beavis A, 2006, Repeatability of Functional MRI for Conformal Avoidance Radiotherapy Planning. J Magn Reson Imaging 23:108–114 Aoyama H, Kamada K, Shirato H et al, 2004, Integration of mfunctional brain information into stereotactic irradiation treatment planning using magnetoencephalography and magnetic resonance axonography. Int J Radiat Oncol Biol Phys 58:1177–1183 Bleehen NM, Stenning SP, 1991, A medical research council trial of two radiotherapy doses in the treatment of grades 3 and 4 astrocytoma. Br J Cancer 64:769–774 Burman C, Kutcher GJ, Emami B, Goitein M, 1991, Fitting of tissue tolerance data to analytic function: improving the therapeutic ratio. Int J Radiat Oncol Biol Phys 21:123–135 Chang J, Kowalski A, Hou B, Narayana A, 2008, Feasibility Study of Intensity-Modulated Radiotherapy, IMRT, treatment planning using brain functional MRI. Med Dosim 33(1):42–47 Clark BG et al, 1998, The integral biologically effective dose to predict brain stem toxicity of hypofractionated stereotactic radiotherapy. Int J Radiat Oncol Biol Phys 40:667–675 Emami B et al, 1991, Tolerance of normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys 21:109–122 Flickinger JC, Kondziolka D, Lunsford LD et al, 2000, Development of a model to predict permanent symptomatic postradiosurgery. Int J Radiat Oncol Biol Phys 46:1143–1148 Jemal A, Murray T, Ward E et al, 2005, Cancer statistics, 2005. CA Cancer J Clin 55:10–30 Lee SW, Fraass BA, Marsh LH et al, 1999, Patterns of failure following high-dose 3-D conformal radiotherapy for high-grade astrocytomas: a quantitative dosimetric study. Int J Radiat Oncol Biol Phys 43:79–88 Liu WC, Schulder M, Narra V et al, 2000, Functional magnetic resonance imaging aided radiation treatment planning. Med Phys 27:1563–1572 Marks JE et al, 1980, Cerebral radionecrosis: incidence and risk in relation to dose, time fractionation and volume. Int J Radiat Oncol Biol Phys 7:243–252 Moonen CTW, Bandettini PA, eds,, 1999, Functional MRI1. Springer-Verlag, Berlin Nelson DF,Diener-WestM, Horton J et al, 1988, Combinedmodality approach to treatment of malignant gliomas - Re-evaluation of RTOG 7401/ECOG 1374 with long-term follow-up: A joint study of the radiation therapy oncology group and the eastern cooperative oncology group. NCI Monogr 279–284 Rowinsky EK, 1997, Paclitaxel pharmacology and other tumor types. Semin Oncol 24:S19-1–S19-12 Teh BS, Mai WY, Grant WH et al, 2002, Intensity modulated radiotherapy, IMRT, decreases treatment-related morbidity and potentially enhances tumor control. Cancer Invest 20:437–451 Voges J, Treuer H, Sturm V et al, 1996, Risk analysis of linear radiosurgery. Int J Radiat Oncol Biol Phys 36:1055–1063 Walker MD, Alexander E, Hunt WE et al, 1978, Evaluation of BCNU and/or radiotherapy in the treatment of anaplastic gliomas. J Neurosurg 49:333–343 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 207 EP 217 DI 10.1007/s12253-010-9298-y PG 11 ER PT J AU Boonmars, Th Wu, Z Boonjaruspinyo, S Puapairoj, A Kaewsamut, B Nagano, I Pinlaor, S Yongvanit, P Wonkchalee, O Juasook, A Sudsarn, P Srisawangwong, T AF Boonmars, Thidarut Wu, Zhiliang Boonjaruspinyo, Sirintip Puapairoj, Anucha Kaewsamut, Butsara Nagano, Isao Pinlaor, Somchai Yongvanit, Puangrat Wonkchalee, Orasa Juasook, Amornrat Sudsarn, Pakkayanee Srisawangwong, Tuanchai TI Involvement of c-Ski Oncoprotein in Carcinogenesis of Cholangiocacinoma Induced by Opisthorchis viverrini and N-nitrosodimethylamine SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Opisthorchiasis; Opisthorchis viverrini; c-Ski; TGF-β; CCA; Carcinogenesis ID Opisthorchiasis; Opisthorchis viverrini; c-Ski; TGF-β; CCA; Carcinogenesis AB Opisthorchiasis is the major public health problem in the endemic areas of Thailand and Laos because Opisthorchis viverrini infection causes serious hepatobiliary diseases including CCA. The molecular mechanism of the CCA carcinogenesis induced by the infection remains obscure. To reveal the potential genes and signaling pathways to involve in the carcinogenesis, the present study investigated the expression of c-Ski, an oncogene, and two TGF-β signaling pathway relative genes, TGF-β and Smad4, during the development of CCA induced by O. viverrini infection in hamster model, and in human opisthorchiasis associated CCA. The results showed that the expression of c-Ski gene was greatly up-regulated during the carcinogenesis of CCA in hamster model. The overexpression of c-Ski was confirmed by immunohistological staining result which showed the increased expression of c-Ski protein in cytoplasm of the epithelial lining of hepatic bile ducts. Moreover, the immunohistological staining of the specimens of human opisthorchiasis associated CCA revealed the up-regulated expression of c-Ski and Smad4 proteins in the cytoplasm of the epithelial lining of hepatic bile ducts and stomal fibrosis respectively. The expression of TGF-β and Smad4 were up-regulated, which expression kinetics was time-dependent of CCA development. These results suggest that c-Ski is likely involved in the carcinogenesis of CCA induced by O. viverrini infection through regulating TGF-β signaling pathway. C1 [Boonmars, Thidarut] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Wu, Zhiliang] Gifu University, Graduate School of Medicine, Department of Parasitology, Yanagido1-1, 501-1194 Gifu, Japan. [Boonjaruspinyo, Sirintip] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Puapairoj, Anucha] Khon Kaen University, Faculty of Medicine, Department of Pathology, 40002 Khon Kaen, Thailand. [Kaewsamut, Butsara] Khon Kaen University, Northeast Laboratory Animal Center, 40002 Khon Kaen, Thailand. [Nagano, Isao] Gifu University, Graduate School of Medicine, Department of Parasitology, Yanagido1-1, 501-1194 Gifu, Japan. [Pinlaor, Somchai] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Yongvanit, Puangrat] Khon Kaen University, Faculty of Medicine, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Wonkchalee, Orasa] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Juasook, Amornrat] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Sudsarn, Pakkayanee] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Srisawangwong, Tuanchai] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. RP Boonmars, Th (reprint author), Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. EM bthida@kku.ac.th CR IARC, 1994, Infection with liver flukes, Opisthorchis viverrini, Opisthorchis felineus and Clonorchis sinensis). IARC Monogr Eval Carcinog Risk Chem Hum 61:121–175 Sithithaworn P, Haswell-Elkins M, 2003, Epidemiology of Opisthorchis viverrini. Acta Trop 88:187–194 Sripa B, 2003, Pathobiology of opisthorchiasis: an update. Acta Trop 88:209–220 Thamavit W, Kongkanuntn R, Tiwawech D, Moore MA, 1987, Level of Opisthorchis infestation and carcinogen dose-dependence of cholangiocarcinoma induction in Syrian golden hamsters. Virchows Arch B Cell Pathol Incl Mol Pathol 54:52–58 Thamavit W, Pairojkul C, Tiwawech D et al, 1993, Promotion of cholangiocarcinogenesis in the hamster liver by bile duct ligation after dimethylnitrosamine initiation. Carcinogenesis 14:2415– 2417 Thamavit W, Pairojkul C, Tiwawech D, Shirai T, Ito N, 1994, Lack of promoting effect of proline on bile duct cancer development in dimethylnitrosamine-initiated hamster livers. Teratog Carcinog Mutagen 14:169–174 Li Y, Turck CM, Teumer JK, Stavnezer E, 1986, Unique sequence, ski, in Sloan-Kettering avian retroviruses with properties of a new cell-derived oncogene. J Virol 57:1065–1072 Colmenares C, Stavnezer E, 1989, The ski oncogene induces muscle differentiation in quail embryo cells. Cell 59:293–303 Fumagalli S, Doneda L, Nomura N, Larizza L, 1993, Expression of the c-Ski proto-oncogene in human melanoma cell lines. Melanoma Res 3:23–27 Pelzer T, Lyons GE, Kim S, Moreadith RW, 1996, Cloning and characterization of the murine homolog of the sno proto-oncogene reveals a novel splice variant. Dev Dyn 205:114–125 Atanasoski S, Notterpek L, Lee HY et al, 2004, The protooncogene Ski controls Schwann cell proliferation and myelination. Neuron 43:499–511 Reed JA, Bales E, Xu W, Okan NA, Bandyopadhyay D, Medrano EE, 2001, Cytoplasmic localization of the oncogenic protein Ski in human cutaneous melanomas in vivo: functional implications for transforming growth factor beta signaling. Cancer Res 61:8074–8078 Reed JA, Lin Q, Chen D, Mian IS, Medrano EE, 2005, SKI pathways inducing progression of human melanoma. Cancer Metastasis Rev 24:265–272 Markowitz S, Wang J, Myeroff L et al, 1995, Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science 268:1336–1338 Miyaki M, Iijima T, Konishi M et al, 1999, Higher frequency of Smad4 gene mutation in human colorectal cancer with distant metastasis. Oncogene 18:3098–3103 Namciu S, Lyons GE, Micales BK et al, 1995, Enhanced expression of mouse c-Ski accompanies terminal skeletal muscle differentiation in vivo and in vitro. Dev Dyn 204:291–300 Zhang F, Lundin M, Ristimaki A et al, 2003, Ski-related novel protein N, SnoN), a negative controller of transforming growth factor-beta signaling, is a prognostic marker in estrogen receptorpositive breast carcinomas. Cancer Res 63:5005–5010 Fukuchi M, Nakajima M, Fukai Y et al, 2004, Increased expression of c-Ski as a co-repressor in transforming growth factor-beta signaling correlates with progression of esophageal squamous cell carcinoma. Int J Cancer 108:818–824 Ritter M, Kattmann D, Teichler S et al, 2006, Inhibition of retinoic acid receptor signaling by Ski in acute myeloid leukemia. Leukemia 20:437–443 Bravou V, Antonacopoulou A, Papadaki H et al, 2009, TGF-beta repressors SnoN and Ski are implicated in human colorectal carcinogenesis. Cell Oncol 31:41–51 Shi Y, Massague J, 2003, Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 113:685–700 Kaewkes S, 2003, Taxonomy and biology of liver flukes. Acta Trop 88:177–186 Boonmars T, Srisawangwong T, Srirach P et al, 2007, Apoptosisrelated gene expressions in hamsters re-infected with Opisthorchis viverrini and re-treated with praziquantel. Parasitol Res 102:57–62 Luo K, 2004, Ski and SnoN: negative regulators of TGF-beta signaling. Curr Opin Genet Dev 14:65–70 Akiyoshi S, Inoue H, Hanai J et al, 1999, c-Ski acts as a transcriptional co-repressor in transforming growth factor-beta signaling through interaction with smads. J Biol Chem 274:35269–35277 Prunier C, Pessah M, Ferrand N et al, 2003, The oncoprotein Ski acts as an antagonist of transforming growth factor-beta signaling by suppressing Smad2 phosphorylation. J Biol Chem 278:26249– 26257 Suzuki H, Yagi K, Kondo M et al, 2004, c-Ski inhibits the TGFbeta signaling pathway through stabilization of inactive Smad complexes on Smad- binding elements. Oncogene 23:5068–5076 Zhang B, Halder SK, Zhang S, Datta PK, 2009, Targeting transforming growth factor-beta signaling in liver metastasis of colon cancer. Cancer Lett 277:114–120 Patsenker E, Popov Y, Stickel F et al, 2008, Inhibition of integrin alphavbeta6 on cholangiocytes blocks transforming growth factorbeta activation and retards biliary fibrosis progression. Gastroenterology 135:660–670 Dumont N, Arteaga CL, 2000, Transforming growth factor-β and breast cancer: tumor promoting effects of transforming growth factor-β. Breast Cancer Res 2:125–132 Milani S, Herbst H, Schuppan D, Stein H, Surrenti C, 1991, Transforming growth factors beta 1 and beta 2 are differentially expressed in fibrotic liver disease. Am J Pathol 139:1221–1229 Levy L, Hill CS, 2006, Alterations in components of the TGF-β superfamily signaling pathways in human cancer. Cytokine Growth Factor Rev 17:41–58 Dranoff G, 2004, Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer 4:11–22 Jakowlew SB, 2006, Transforming growth factor-beta in cancer and metastasis. Cancer Metastasis Rev 25:435–457 Laiho M, DeCaprio JA, Ludlow JW, Livingston DM, Massague J, 1990, Growth inhibition by TGF-beta linked to suppression of retinoblastoma protein phosphorylation. Cell 62:175–185 Hannon GJ, Beach D, 1994, p15INK4B is a potential effector of TGF-beta-induced cell cycle arrest. Nature 371:257–261 Iavarone A, Massague J, 1999, E2F and histone deacetylase mediate transforming growth factor beta repression of cdc25A during keratinocyte cell cycle arrest. Mol Cell Biol 19:916–940 Jang CW, Chen CH, Chen CC et al, 2002, TGF-beta induces apoptosis through Smad-mediated expression of DAP-kinase. Nat Cell Biol 4:51–58 Kim BG, Li C, Qiao Wet al, 2006, Smad4 signalling in T cells is required for suppression of gastrointestinal cancer. Nature 441:1015–1019 Kang YK, Kim WH, Jang JJ, 2002, Expression of G1-S modulators, p53, p16, p27, cyclin D1, Rb, and Smad4/Dpc4 in intrahepatic cholangiocarcinoma. Hum Pathol 33:877–883 Liu Y, Wang LF, Zou HF, Song XY, Xu HF, Lin P, Zheng HH, Yu XG, 2006, Expression and location of Smad2, 4 mRNAs during and after liver fibrogenesis of rats. World J Gastroenterol 14;12, 10):1577–1582 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 219 EP 227 DI 10.1007/s12253-010-9300-8 PG 9 ER PT J AU Tatrai, E Hartyanszky, I Laszik, A Acsady, Gy Sotonyi, P Hubay, M AF Tatrai, Eniko Hartyanszky, Istvan Laszik, Andras Acsady, Gyorgy Sotonyi, Peter Hubay, Marta TI The Role of Viral Infections in the Development of Dilated Cardiomyopathy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cardiomyopathy; Endomyocardial biopsy; Myocardium; Polymerase chain reaction; Viruses ID Cardiomyopathy; Endomyocardial biopsy; Myocardium; Polymerase chain reaction; Viruses AB Enteroviruses (EVs) are the most frequent pathogens in myocarditis and in the subsequently developing dilated cardiomyopathy as well. Furthermore, persistence of other viruses might play a pathogenic role in the evolution from myocarditis to dilated cardiomyopathy. Explanted heart of 28 patients, who underwent heart transplantation were screened for EV, AdV3 and HHV6 sequences in order to assess the incidence of cardiac viral infection that may be implicated in the pathogenesis of cardiomyopathy, and estimate viral distribution in the myocardium. Viral sequences were extracted from five different regions of the hearts. Nested PCR was used to amplify conservative regions of AdV3, HHV6 and EVs. Histological examination was performed on routinely processed myocardial samples. AdV3 was verified in one fourth of the patients. ADV3 and HHV6 sequences coexisted in one case with inflammatory cardiomyopathy. Some patients had more than one positive area of their heart. AdV3 positive right ventricular samples were double in amount compared to the left ones. None of the patients had positive result for EV. This is the first occasion to identify AdV3 (a mainly respiratory infective virus) sequence in explanted hearts of cardiomyopathy patients. Though the clinical importance of our results is still unclear, AdV3 could be a new member of the viral group with possible pathogenic effect on the myocardium. Regional distribution of viral sequence location confirmed that the right ventricular wall as a biopsy sampling site might be adequate for endomyocardial biopsy pro diagnostic purposes. C1 [Tatrai, Eniko] Semmelweis University, Department of Forensic & Insurance Medicine, 93 Ulloi str, 1091 Budapest, Hungary. [Hartyanszky, Istvan] Semmelweis University, Department of Cardiovascular Surgery, 1122 Budapest, Hungary. [Laszik, Andras] Semmelweis University, Department of Forensic & Insurance Medicine, 93 Ulloi str, 1091 Budapest, Hungary. [Acsady, Gyorgy] Semmelweis University, Department of Cardiovascular Surgery, 1122 Budapest, Hungary. [Sotonyi, Peter] Semmelweis University, Department of Forensic & Insurance Medicine, 93 Ulloi str, 1091 Budapest, Hungary. [Hubay, Marta] Semmelweis University, Department of Forensic & Insurance Medicine, 93 Ulloi str, 1091 Budapest, Hungary. RP Tatrai, E (reprint author), Semmelweis University, Department of Forensic & Insurance Medicine, 1091 Budapest, Hungary. EM tatraie@freemail.hu CR Goodwin JF, Oakley CM, 1972, The cardiomyopathies. Br Heart J 34(6):545–552 Brandenburg RO, Chazov E, Cherian G et al, 1980, Report of the WHO/ISFC task force on the definition and classification of cardiomyopathies. Br Heart J 44(6):672–673 Richardson P, McKenna W, Bristow M et al, 1996, Report of the 1995 World Health Organization/International Society and Federation of Cardiology Task Force on the Definition and Classification of cardiomyopathies. Circulation 93(5):841–842 Mason JW, 2003, Myocarditis and dilated cardiomyopathy: an inflammatory link. Cardiovasc Res 60(1):5–10 Bowles NE, Ni J, Kearney DL et al, 2003, Detection of viruses in myocardial tissues by polymerase chain reaction: evidence of adenovirus as a common cause of myocarditis in children and adults. J Am Coll Cardiol 42(3):466–472 Basso C, Calabrese F, Corrado D et al, 2001, Postmortem diagnosis in sudden cardiac death victims: macroscopic, microscopic and molecular findings. Cardiovasc Res 50(2):290–300 Francalanci P, Chance JL, Vatta M et al, 2004, Cardiotropic viruses in the myocardium of children with end-stage heart disease. J Heart Lung Transplant 23(9):1046–1052 Woodruff JF, 1980, Viral myocarditis. A review. Am J Pathol 101, 2):425–484 Comar M, D'Agaro P, Campello C et al, 2009, Human herpes virus 6 in archival cardiac tissues from children with idiopathic dilated cardiomyopathy or congenital heart disease. J Clin Pathol 62(1):80–83 Fukae S, Ashizawa N, Morikawa S et al, 2000, A fatal case of fulminant myocarditis with human herpesvirus-6 infection. Intern Med 39(8):632–636 Rohayem J, Dinger J, Fischer R et al, 2001, Fatal myocarditis associated with acute parvovirus B19 and human herpesvirus 6 coinfection. J Clin Microbiol 39(12):4585–4587 Yoshikawa T, Ihira M, Suzuki K et al, 2001, Fatal acute myocarditis in an infant with human herpesvirus 6 infection. J Clin Pathol 54(10):792–795 Savon C, Acosta B, Valdes O et al, 2008, A myocarditis outbreak with fatal cases associated with adenovirus subgenera C among children from Havana City in 2005. J Clin Virol 43(2):152–157 James L, Vernon MO, Jones RC et al, 2007, Outbreak of human adenovirus type 3 infection in a pediatric long-term care facility– Illinois, 2005. Clin Infect Dis 45(4):416–420 Herbert FA, Wilkinson D, Burchak E et al, 1977, Adenovirus type 3 pneumonia causing lung damage in childhood. Can Med Assoc J 116(3):274–276 Dennert R, Crijns HJ, Heymans S, 2008, Acute viral myocarditis. Eur Heart J 29:2073–2082 Caforio AL, 1994, Role of autoimmunity in dilated cardiomyopathy. Br Heart J 72(6 Suppl):S30–34 Martin AB, Webber S, Fricker FJ et al, 1994, Acute myocarditis. Rapid diagnosis by PCR in children. Circulation 90(1):330–339 Bowles NE, Kearney DL, Ni J et al, 1999, The detection of viral genomes by polymerase chain reaction in the myocardium of pediatric patients with advanced HIV disease. J Am Coll Cardiol 34(3):857–865 Pauschinger M, Bowles NE, Fuentes-Garcia FJ et al, 1999, Detection of adenoviral genome in the myocardium of adult patients with idiopathic left ventricular dysfunction. Circulation 99(10):1348–1354 Rey L, Lambert V, Wattre P et al, 2001, Detection of enteroviruses ribonucleic acid sequences in endomyocardial tissue from adult patients with chronic dilated cardiomyopathy by a rapid RT-PCR and hybridization assay. J Med Virol 64, 2):133–140 Donoso Mantke O, Nitsche A, Meyer R et al, 2004, Analysing myocardial tissue from explanted hearts of heart transplant recipients and multi-organ donors for the presence of parvovirus B19 DNA. J Clin Virol 31(1):32–39 Suedkamp M, Lissel C, Eiffert H et al, 1999, Cardiac myocytes of hearts from patients with end-stage dilated cardiomyopathy do not contain Borrelia burgdorferi DNA. Am Heart J 138(2 Pt 1):269–272 Tatrai E, Hartyanszky I, Laszik A et al, 2007, Molecular biological virus identification in dilated cardiomyopathy. Orvosi Hetilap 148(48):2275–2278 Mahrholdt H, Wagner A, Deluigi CC et al, 2006, Presentation, patterns of myocardial damage, and clinical course of viral myocarditis. Circulation 114(15):1581–1590 Hauck AJ, Kearney DL, Edwards WD, 1989, Evaluation of postmortem endomyocardial biopsy specimens from 38 patients with lymphocytic myocarditis: implications for role of sampling error. Mayo Clin Proc 64(10):1235–1245 Kuhl U, Pauschinger M, Noutsias M et al, 2005, High prevalence of viral genomes and multiple viral infections in the myocardium of adults with “idiopathic” left ventricular dysfunction. Circulation 111(7):887–893 Kuhl U, Pauschinger M, Schwimmbeck PL et al, 2003, Interferon-beta treatment eliminates cardiotropic viruses and improves left ventricular function in patients with myocardial persistence of viral genomes and left ventricular dysfunction. Circulation 107(22):2793–2798 Maekawa Y, Ouzounian M, Opavsky MA et al, 2007, Connecting the missing link between dilated cardiomyopathy and viral myocarditis: virus, cytoskeleton, and innate immunity. Circulation 115(1):5–8 Lawson CM, 2000, Evidence for mimicry by viral antigens in animal models of autoimmune disease including myocarditis. Cell Mol Life Sci 57(4):552–560 Heymans S, Pauschinger M, De Palma A et al, 2006, Inhibition of urokinase-type plasminogen activator or matrix metalloproteinases prevents cardiac injury and dysfunction during viral myocarditis. Circulation 114(6):565–573 Caforio AL, Grazzini M, Mann JM et al, 1992, Identification of alpha- and beta-cardiac myosin heavy chain isoforms as major autoantigens in dilated cardiomyopathy. Circulation 85(5):1734– 1742 Caruso A, Rotola A, Comar M et al, 2002, HHV-6 infects human aortic and heart microvascular endothelial cells, increasing their ability to secrete proinflammatory chemokines. J Med Virol 67, 4):528–533 Horvath R, Cerny J, Benedik J et al, 2000, The possible role of human cytomegalovirus, HCMV, in the origin of atherosclerosis. J Clin Virol 16(1):17–24 Ismail A, Khosravi H, Olson H, 1999, The role of infection in atherosclerosis and coronary artery disease: a new therapeutic target. Heart Dis 1(4):233–240 Andreoletti L, Hober D, Decoene C et al, 1996, Detection of enteroviral RNA by polymerase chain reaction in endomyocardial tissue of patients with chronic cardiac diseases. J Med Virol 48, 1):53–59 Petitjean J, Kopecka H, Freymuth F et al, 1992, Detection of enteroviruses in endomyocardial biopsy by molecular approach. J Med Virol 37(1):76–82 de Leeuw N, Melchers WJ, Balk AH et al, 1998, No evidence for persistent enterovirus infection in patients with end-stage idiopathic dilated cardiomyopathy. J Infect Dis 178(1):256–259 Mahon NG, Zal B, Arno G et al, 2001, Absence of viral nucleic acids in early and late dilated cardiomyopathy. Heart 86(6):687– 692 Weiss LM, Liu XF, Chang KL et al, 1992, Detection of enteroviral RNA in idiopathic dilated cardiomyopathy and other human cardiac tissues. J Clin Invest 90(1):156–159 Maron BJ, Towbin JA, Thiene G et al, 2006, Contemporary definitions and classification of the cardiomyopathies: an American Heart Association Scientific Statement from the Council on Clinical Cardiology, Heart Failure and Transplantation Committee; Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interdisciplinary Working Groups; and Council on Epidemiology and Prevention. Circulation 113(14):1807–1816 Osterziel KJ, Scheffold T, Perrot A et al, 2001, Genetics of dilated cardiomyopathy. Z Kardiol 90(7):461–469 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 229 EP 235 DI 10.1007/s12253-010-9302-6 PG 7 ER PT J AU Zhang, B Feng, X Wang, J Xu, X Lin, N Liu, H AF Zhang, Biao Feng, Xuequan Wang, Jinhuan Xu, Xinnu Lin, Na Liu, Hongsheng TI Combined Antitumor Effect of Ad-bFGF-siRNA and Ad-Vpr on the Growth of Xenograft Glioma in Nude Mouse Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE siRNA; bFGF; Vpr; Adenoviral vector ID siRNA; bFGF; Vpr; Adenoviral vector AB Basic fibroblast growth factor (bFGF) has been demonstrated to correlate with glioma grade and clinical outcome and has established its possible usefulness as a target for glioma therapy. Vpr has been described as an antitumor agent and displays a potent antitumor nature. Here, we try to investigate whether a combined treatment with bFGF-siRNA and Vpr gene would have a enhanced effectiveness on glioma in vitro and in vivo.After treatments with only Ad-bFGF-siRNA, only Ad-Vpr, and a combination of both, we assessed the changes in cell proliferation, cell cycle, and apoptosis in vitro by the methods of MTT, PI and FITC-AnnexinV double staining, respevtively. In addition, we also evaluated the combined effect of bFGF-siRNA and Vpr gene therapy on glioma in vivo using xenograft glioma models in nude mice. Combined Ad-bFGF-siRNA and Ad-Vpr treatment was more better successful in inhibiting cell proliferation in comparison with treatments of either Ad-bFGF-siRNA or Ad-Vpr alone. Treatment of Ad-Vpr alone or a treatment of a combination of Ad-bFGF-siRNA and Ad-Vpr induced the G2/M cell cycle arrest and apoptosis; however, combined treatment was more effective than the Ad-Vpr treatment alone. Although each single treatment can slow the growth of xenograft glioma, the combined treatment with AdbFGF-siRNA and Ad-Vpr was better than either the AdbFGF-siRNA or Ad-Vpr treatment alone. Our results suggest that the combination therapy with bFGF-siRNA and Vpr gene can achieve a enhanced activity of antiglioma, supporting the idea that the combination of these two antitumor agents could open new perspectives in glioma therapy. C1 [Zhang, Biao] Tianjin Huan Hu Hospital, Clinical Lab, 300060 Tianjin, China. [Feng, Xuequan] Tianjin Medical University General Hospital, Department of Neurosurgery, 300192 Tianjin, China. [Wang, Jinhuan] Tianjin Medical University General Hospital, Department of Neurosurgery, 122# Qixiangtai Road, Hexi District, 300060 Tianjin, China. [Xu, Xinnu] Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 300192 Tianjin, China. [Lin, Na] Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 300192 Tianjin, China. [Liu, Hongsheng] Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 300192 Tianjin, China. RP Wang, J (reprint author), Tianjin Medical University General Hospital, Department of Neurosurgery, 300060 Tianjin, China. EM wangjinhuanfch@yahoo.com.cn CR Hutterer M, Knyazev P, Abate A, Reschke M, Maier H, Stefanova N, Knyazeva T, Barbieri V, Reindl M, Muigg A, Kostron H, Stockhammer G, Ullrich A, 2008, Axl and growth arrest-specific gene 6 are frequently overexpressed in human gliomas and predict poor prognosis in patients with glioblastoma multiforme. Clin Cancer Res 14(1):130–138 Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO, 2005, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352(10):987–996 Auguste P, Gursel DB, Lemiere S, Reimers D, Cuevas P, Carceller F, Di Santo JP, Bikfalvi A, 2001, Inhibition of fibroblast growth factor/fibroblast growth factor receptor activity in glioma cells impedes tumor growth by both angiogenesis-dependent and -independent mechanisms. Cancer Res 61(4):1717–1726 Ryther RC, Flynt AS, Phillips JA 3rd, Patton JG, 2005, siRNA therapeutics: big potential from small RNAs. Gene Ther 12(1):5– 11 Muthumani K, Hwang DS, Desai BM, Zhang D, Dayes N, Green DR, Weiner DB, 2002, HIV-1 Vpr induces apoptosis through caspase 9 in T cells and peripheral blood mononuclear cells. J Biol Chem 277(40):37820–37831 Muthumani K, Zhang D, Hwang DS, Kudchodkar S, Dayes NS, Desai BM, Malik AS, Yang JS, Chattergoon MA, Maguire HC Jr, Weiner DB, 2002, Adenovirus encoding HIV-1 Vpr activates caspase 9 and induces apoptotic cell death in both p53 positive and negative human tumor cell lines. Oncogene 21(30):4613– 4625 McCray AN, Ugen KE, Heller R, 2007, Enhancement of antimelanoma activity of a plasmid expressing HIV-1 Vpr delivered through in vivo electroporation. Cancer Biol Ther 6(8):1269–1275 McCray AN, Ugen KE, Muthumani K, Kim JJ, Weiner DB, Heller R, 2006, Complete regression of established subcutaneous B16 murine melanoma tumors after delivery of an HIV-1 Vprexpressing plasmid by in vivo electroporation. Mol Ther 14, 5):647–655 Muthumani K, Choo AY, Hwang DS, Ugen KE, Weiner DB, 2004, HIV-1 Vpr: enhancing sensitivity of tumors to apoptosis. Curr Drug Deliv 1(4):335–344 Muthumani K, Lambert VM, Sardesai NY, Kim JJ, Heller R, Weiner DB, Ugen KE, 2009, Analysis of the potential for HIV-1 Vpr as an anti-cancer agent. Curr HIV Res 7(2):144–152 Muthumani K, Lambert VM, Shanmugam M, Thieu KP, Choo AY, Chung JC, Satishchandran A, Kim JJ, Weiner DB, Ugen KE, 2009, Anti-tumor activity mediated by protein and peptide transduction of HIV viral protein R, Vpr). Cancer Biol Ther 8, 2):180–187 Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100(1):57–70 Jordan JP, Hand CM, Markowitz RS, Black P, 1992, Test for chemotherapeutic sensitivity of cerebral gliomas: use of colorimetric MTT assay. J Neurooncol 14(1):19–35 Chen B, Timiryasova TM, Andres ML, Kajioka EH, Dutta-Roy R, Gridley DS, Fodor I, 2000, Evaluation of combined vaccinia virus-mediated antitumor gene therapy with p53, IL-2, and IL-12 in a glioma model. Cancer Gene Ther 7(11):1437–1447 Fukui S, Nawashiro H, Otani N, Ooigawa H, Nomura N, Yano A, Miyazawa T, Ohnuki A, Tsuzuki N, Katoh H, Ishihara S, Shima K, 2003, Nuclear accumulation of basic fibroblast growth factor in human astrocytic tumors. Cancer 97(12):3061– 3067 Baguma-Nibasheka M, Li AW, Murphy PR, 2007, The fibroblast growth factor-2 antisense gene inhibits nuclear accumulation of FGF-2 and delays cell cycle progression in C6 glioma cells. Mol Cell Endocrinol 267(1–2):127–136 Aoki T, Kato S, Fox JC, Okamoto K, Sakata K, Morimatsu M, Shigemori M, 2002, Inhibition of autocrine fibroblast growth factor signaling by the adenovirus-mediated expression of an antisense transgene or a dominant negative receptor in human glioma cells in vitro. Int J Oncol 21(3):629–636 Takahashi JA, Fukumoto M, Kozai Y, Ito N, Oda Y, Kikuchi H, Hatanaka M, 1991, Inhibition of cell growth and tumorigenesis of human glioblastoma cells by a neutralizing antibody against human basic fibroblast growth factor. FEBS Lett 288(1–2):65–71 Marek L, Ware KE, Fritzsche A, Hercule P, Helton WR, Smith JE, McDermott LA, Coldren CD, Nemenoff RA, Merrick DT, Helfrich BA, Bunn PA Jr, Heasley LE, 2009, Fibroblast growth factor, FGF, and FGF receptor-mediated autocrine signaling in non-small-cell lung cancer cells. Mol Pharmacol 75(1):196–207 Wang SJ, Wang JH, Zhang YW, Xu XN, Liu HS, 2008, Effects of small interfering RNA targeting basic fibroblast growth factor on proliferation and apoptosis of glioma cell line U251. Ai Zheng 27, 9):905–909 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 237 EP 242 DI 10.1007/s12253-010-9303-5 PG 6 ER PT J AU Mitrovic, Z Ilic, I Aurer, I Kinda, BS Radman, I Dotlic, S Ajdukovic, R Labar, B AF Mitrovic, Zdravko Ilic, Ivana Aurer, Igor Kinda, Basic Sandra Radman, Ivo Dotlic, Snjezana Ajdukovic, Radmila Labar, Boris TI Prognostic Significance of Survivin and Caspase-3 Immunohistochemical Expression in Patients with Diffuse Large B-cell Lymphoma Treated with Rituximab and CHOP SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-Hodgkin’s lymphoma; Diffuse large B-cell lymphoma; Survivin; Caspase-3; Prognostic factors; Rituximab; Immunohistochemistry ID Non-Hodgkin’s lymphoma; Diffuse large B-cell lymphoma; Survivin; Caspase-3; Prognostic factors; Rituximab; Immunohistochemistry AB Survivin is an inhibitor of apoptosis whose expression may be associated with inferior outcome in patients with diffuse large B-cell lymphoma (DLBCL) treated without rituximab. Caspase-3 is the final caspase of the apoptotic cascade and its pattern of expression may also be related to patients’ outcome. In this study we investigated immunohistochemical expression of survivin and caspase-3 (CPP32) in 57 patients with DLBCL treated with rituximab and CHOP (RCHOP). According to previously published criteria, we separately analyzed correlation of different types of survivin expression with patients’ outcome. Nuclear survivin was expressed in only 26% of cases, cytoplasmic survivin was expressed in 81% of cases while application of immunoreactivity scoring system yielded 58% of survivin positive cases. Caspase-3 was expressed in 77% of cases. There were no significant correlations between any type of survivin expression and response to treatment or survival of the patients. The expression of caspase-3 was also not associated with patients’ outcome. We conclude that survivin and caspase-3 have no significant prognostic significance in patients with DLBCL treated with R-CHOP. C1 [Mitrovic, Zdravko] University Hospital Center Zagreb, Division of Hematology and Department of Medicine, Kispaticeva 12, 10 000 Zagreb, Croatia. [Ilic, Ivana] University Hospital Center Zagreb, Department of PathologyZagreb, Croatia. [Aurer, Igor] University Hospital Center Zagreb, Division of Hematology and Department of Medicine, Kispaticeva 12, 10 000 Zagreb, Croatia. [Kinda, Basic Sandra] University Hospital Center Zagreb, Division of Hematology and Department of MedicineZagreb, Croatia. [Radman, Ivo] University Hospital Center Zagreb, Division of Hematology and Department of MedicineZagreb, Croatia. [Dotlic, Snjezana] University Hospital Center Zagreb, Department of PathologyZagreb, Croatia. [Ajdukovic, Radmila] University Hospital Dubrava, Department of Medicine, Av. Gojka Suska 6, 10 000 Zagreb, Croatia. [Labar, Boris] University Hospital Center Zagreb, Division of Hematology and Department of Medicine, Kispaticeva 12, 10 000 Zagreb, Croatia. RP Mitrovic, Z (reprint author), University Hospital Center Zagreb, Division of Hematology and Department of Medicine, 10 000 Zagreb, Croatia. EM mitrovic@mef.hr CR Feugier P, Van Hoof A, Sebban C et al, 2005, Long-term results of the R-CHOP study in the treatment of elderly patients with diffuse large B-cell lymphoma: a study by the Groupe d’Etude des Lymphomes de l’Adulte. J Clin Oncol 23:4117–4126 Pfreundschuh M, Trumper L, Osterborg A et al, 2006, CHOPlike chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with good-prognosis diffuse large-B cell lymphoma: a randomized controlled trial by the MabThera International Trial, MInT, Group. Lancet Oncol 7:379–391 Cvetkovic RS, Perry CM, 2006, Rituximab—a review of its use in Non-Hodgkin’s lymphoma and chronic lymphocytic leukaemia. Drugs 66:791–820 Guha M, Altieri DC, 2009, Survivin as a global target of intrinsic tumor suppression networks. Cell Cycle 8:2708–2710 Li F, Yang J, Ramnath N et al, 2005, Nuclear or cytoplasmic expression of survivin: what is the significance? Int J Cancer 114:509–512 Watanuki-Miyauchi R, Kojima Y, Tsurumi H et al, 2005, Expression of survivin and of antigen detected by a novel monoclonal antibody, T332, is associated with outcome of diffuse large B-cell lymphoma and its subtypes. Pathol Int 55:324–330 Karabatsou K, Pal P, Dodd S et al, 2006, Expression of survivin, platelet-derived growth factor A, PDGF-A, and PDGF receptor alpha in primary central nervous system lymphoma. J Neurooncol 79:171–179 Lin L, Min Z, Ping Z, 2007, Expression of PLK1 and survivin in diffuse large B-cell lymphoma. Leuk Lymphoma 48:2179–2183 Mainou-Fowler T, Overman LM, Dignum H et al, 2008, A new subtype-specific monoclonal antibody for IAP-survivin identifies high-risk patients with diffuse large B-cell lymphoma and improves the prognostic value of bcl-2. Int J Oncol 32:59–68 Adida C, Haioun C, Gaulard P et al, 2000, Prognostic significance of survivin expression in diffuse large B-cell lymphomas. Blood 96:1921–1925 Aktas S, Kargi A, Olgun N et al, 2009, Prognostic significance of cell proliferation and apoptosis-regulating proteins in Epstein-Barr Virus positive and negative pediatric non-Hodgkin’s lymphoma. Pathol Oncol Res 15:345–350 Paydas S, Seydaoglu G, Ergin M et al, 2009, Prognostic significance of angiogenic/lymphangiogenic, anti-apoptotic, inflammatory and viral factors in 88 cases with diffuse large B cell lymphoma and review of the literature. Leuk Res 33:1627–1635 Hans CP, Weisenburger DD, Greiner TC et al, 2005, Expression of PKC-beta or cyclin D2 predicts for inferior survival in diffuse large B-cell lymphoma. Mod Pathol 18:1377–1384 Krajewski S, Gascoyne RD, Zapeta JM et al, 1997, Immunolocalisation of the ICE/Ced 3-family protease, CPP32, Caspase-3, in non-Hodgkin lymphomas, Chronic lymphocytic leukemias and reactive lymph nodes. Blood 89:3817–3825 Donoghue S, Baden SH, Lauder I et al, 1999, Immunohistochemical localization of caspase-3 correlates with clinical outcome in B-cell diffuse large cell lymphoma. Cancer Res 59:5386–5391 ten Berge RL, Meijer CJ, Dukers DF et al, 2002, Expression levels of apoptosis-related proteins predict clinical outcome in anaplastic large cell lymphoma. Blood 99:4540–4546 Muris JJ, Cillessen SA, Vos Wet al, 2005, Immunohistochemical profiling of caspase signalling pathways predicts clinical response to chemotherapy in primary nodal diffuse large B-cell lymphomas. Blood 105:2916–2923 Cheson BD, Horning SJ, Coiffier B et al, 1999, Report of an international workshop to standardize response criteria for non- Hodgkin’s lymphomas. NCI Sponsored International Working group. J Clin Oncol 17:1244–1253 The International Non-Hodgkin’s Lymphoma Prognostic Factors Project, 1993, A predictive model for aggressive non-hodgkin’s lymphoma. N Engl J Med 329:987–994 Mounier N, Briere J, Gisselbrecht C et al, 2003, Rituximab plus CHOP, R-CHOP, overcomes bcl-2-associated resistance to chemotherapy in elderly patients with diffuse large B-cell lymphoma, DLBCL). Blood 101:4279–4284 Ansell SM, Arendt BK, Grote DM et al, 2004, Inhibition of survivin expression suppresses the growth of aggressive non- Hodgkin’s lymphoma. Leukemia 18:616–623 Tolcher AW, Mita A, Lewis LD et al, 2008, Phase I and pharmacokinetic study of YM155, a small-molecule inhibitor of survivin. J Clin Oncol 26:5198–5203 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 243 EP 247 DI 10.1007/s12253-010-9304-4 PG 5 ER PT J AU Gergely, L Vancsa, A Miltenyi, Zs Simon, Zs Barath, S Illes, AF Gergely, Lajos Vancsa, Andrea Miltenyi, Zsofia Simon, Zsofia Barath, Sandor Illes, Arpad TI Pretreatment T Lymphocyte Numbers Are Contributing to the Prognostic Significance of Absolute Lymphocyte Numbers in B-cell Non-Hodgkins Lymphomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NHL; Lymphocytes; T cells; Prognostic factors ID NHL; Lymphocytes; T cells; Prognostic factors AB Targeted immuno-chemotherapy resulted in greatly improved survival of B cell lymphoma patients. Several prognostic markers are investigated, amongst them the pretreatment absolute lymphocyte numbers. We investigated lymphocyte subpopulations and correlated this data with prognosis of patients. 88 patients (mean age: 56 years, 18–88, median follow up 32 months) with B cell lymphomas were investigated. There were 51 DLBCL, 16 Follicular NHL, 4 MALT, 7 Marginal Zone NHL, 10 Small lymphocytic cases were investigated. Our data showed that overall survival was statistically significant up to the 0.9 G/l absolute lymphocyte numbers as dividers between the subgroups. The CD19+ B cell numbers, or the CD56+/CD3- NK cell numbers did not represent any significant differences between subgroups. However CD3+, CD4+ and CD8+ T cells were differentiating statistically significant subgroups. Pretreatment CD3+ cell number less than 700/ul and CD8+ cell number less than 200/ul were corresponding with significantly inferior overall survival. These could be verified in the bad prognostic IPI group as well. Our data further support the importance of pretreatment lymphocyte numbers and highlight CD3+ and CD8+ lymphocytes to be the key factors in predicting outcome. C1 [Gergely, Lajos] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz zs. Krt. 22, 4032 Debrecen, Hungary. [Vancsa, Andrea] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz zs. Krt. 22, 4032 Debrecen, Hungary. [Miltenyi, Zsofia] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz zs. Krt. 22, 4032 Debrecen, Hungary. [Simon, Zsofia] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz zs. Krt. 22, 4032 Debrecen, Hungary. [Barath, Sandor] University of Debrecen, Medical and Health Center, 3rd Department of MedicineDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz zs. Krt. 22, 4032 Debrecen, Hungary. RP Gergely, L (reprint author), University of Debrecen, Medical and Health Center, 3rd Department of Medicine, 4032 Debrecen, Hungary. EM lgergely@iiibel.dote.hu CR Yamaguchi M, Nakamura N, Suzuki R, Kagami Y, Okamoto M, Ichinohasama R et al, 2008, De novo CD5+ diffuse large B-cell lymphoma: results of a detailed clinicopathological review in 120 patients. Haematologica 93(8):1195–1202 Marini G, Ippoliti G, Ascari E, Casirola G, 1972, The absolute lymphocyte count in Hodgkin’s disease as an aid in survival prognosis. Haematologica 57(12):822–831 Decaudin D, Des Guetz G, Mathiot C, Dumont J, Hubert P, Vincent-Salomon A et al, 2003, Absolute lymphocyte count as a predictive factor for response to monoclonal anti-CD20 antibody therapy. Ann Oncol 14(1):171–172 Siddiqui M, Ristow K, Markovic SN, Witzig TE, Habermann TM, Colgan JP et al, 2006, Absolute lymphocyte count predicts overall survival in follicular lymphomas. Br J Haematol 134, 6):596–601 Czuczman MS, Grillo-Lopez AJ, Alkuzweny B, Weaver R, Larocca A, McLaughlin P, 2006, Prognostic factors for non-Hodgkin’s lymphoma patients treated with chemotherapy may not predict outcome in patients treated with rituximab. Leuk Lymphoma 47, 9):1830–1840 Wahlin BE, Sander B, Christensson B, Kimby E, 2007, CD8+ Tcell content in diagnostic lymph nodes measured by flow cytometry is a predictor of survival in follicular lymphoma. Clin Cancer Res 13(2 Pt 1):388–397 Kaplan E, Meier P, 1958, Nonparametric estimation from incomplete observations. J Am Stat Assoc 53:457–481 Crowley JJ, McCoy J, 1993, Survival trees by goodness of split. J Am Stat Assoc 88:457–467 Cox DR, 1972, Regression models and life tablets. J Roy Stat Soc, Senes to Applied Statistics 34:187–202 Delgado J, Pratt G, Phillips N, Briones J, Fegan C, Nomdedeu J et al, 2009, Beta2-microglobulin is a better predictor of treatmentfree survival in patients with chronic lymphocytic leukaemia if adjusted according to glomerular filtration rate. Br J Haematol 145, 6):801–805 Talaulikar D, Choudhury A, Shadbolt B, Brown M, 2008, Lymphocytopenia as a prognostic marker for diffuse large B cell lymphomas. Leuk Lymphoma 49(5):959–964 Oki Y, Yamamoto K, Kato H, Kuwatsuka Y, Taji H, Kagami Yet al, 2008, Low absolute lymphocyte count is a poor prognostic marker in patients with diffuse large B-cell lymphoma and suggests patients’ survival benefit from rituximab. Eur J Haematol 81(6):448–453 Cox MC, Nofroni I, Ruco L, Amodeo R, Ferrari A, La Verde G et al, 2008, Low absolute lymphocyte count is a poor prognostic factor in diffuse-large-B-cell-lymphoma. Leuk Lymphoma 49, 9):1745–1751 Kim DH, Baek JH, Chae YS, Kim YK, Kim HJ, Park YH et al, 2007, Absolute lymphocyte counts predicts response to chemotherapy and survival in diffuse large B-cell lymphoma. Leukemia 21(10):2227–2230 Seshadri T, Pintilie M, Keating A, Crump M, Kuruvilla J, 2008, The relationship between absolute lymphocyte count with PFS in patients with Hodgkin’s lymphoma undergoing autologous hematopoietic cell transplant. Bone Marrow Transplant 42(1):29–34 De Angulo G, Yuen C, Palla SL, Anderson PM, Zweidler-McKay PA, 2008, Absolute lymphocyte count is a novel prognostic indicator in ALL and AML: implications for risk stratification and future studies. Cancer 112(2):407–415 Yano Y, Ueda Y, Itoh T, Fuji N, Okugawa K, Naito K et al, 2006, A new strategy using autologous dendritic cells and lymphokine-activated killer cells for cancer immunotherapy: efficient maturation of DCs by co-culture with LAK cells in vitro. Oncol Rep 16(1):147–152 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 249 EP 255 DI 10.1007/s12253-010-9306-2 PG 7 ER PT J AU Chen, X Liao, J Lu, Y Duan, X Sun, W AF Chen, Xiong Liao, Jie Lu, YeBin Duan, XiaoHui Sun, Weijia TI Activation of the PI3K/Akt Pathway Mediates Bone Morphogenetic Protein 2-Induced Invasion of Pancreatic Cancer Cells Panc-1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PI3K/Akt; Bone morphogenetic protein-2 (BMP-2); Invasion; Epithelial to mesenchymal transition (EMT) ID PI3K/Akt; Bone morphogenetic protein-2 (BMP-2); Invasion; Epithelial to mesenchymal transition (EMT) AB Bone morphogenetic proteins (BMPs) signaling has an emerging role in pancreatic cancer. However, because of the multiple effects of different BMPs, no final conclusions have been made as to the role of BMPs in pancreatic cancer. In our studies, we have focused on bone morphogenetic protein 2(BMP-2) because it induces an epithelial to mesenchymal transition (EMT) and accelerates invasion in the human pancreatic cancer cell line Panc-1. It has been reported that the phosphatidylinositol 3-kinase (PI3K)/Akt pathway mediates invasion of gastric and colon cancer cells, which is unrevealed in pancreatic cancer cells. The objective of our study was to investigate whether BMP-2 mediated invasion might pass through the PI3K/Akt pathway. Our results show that expression of phosphorylation of Akt was increased by treatment with BMP-2, but not Noggin, a BMP-2 antagonist. Then pretreatment of Panc-1 cells with LY294002, an inhibitor of the PI3K/AKT pathway, significantly inhibited BMP-2-induced EMT and invasiveness. The data suggest that BMP-2 accelerates invasion of panc-1 cells via the PI3K/AKT pathway in panc-1 cells, which gives clues to searching new therapy targets in advanced pancreatic cancer. C1 [Chen, Xiong] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, China. [Liao, Jie] Central South University, XiangYa Hospital, Department of Endocrinology, 410008 Changsha, China. [Lu, YeBin] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, China. [Duan, XiaoHui] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, China. [Sun, Weijia] Central South University, Institute of Pancreatic Diseases, 410008 Changsha, China. RP Sun, W (reprint author), Central South University, Institute of Pancreatic Diseases, 410008 Changsha, China. EM sunweijiays@hotmail.com CR Gansler T, Brawley OW, 2010, Cancer statistics, 2010. CA Cancer J Clin 60(1):1 Li DH, Xie KP, Wolff R, Abbruzzese JL, 2004, Pancreatic cancer. Lancet 363(9414):1049–1057 ten Dijke P, Korchynskyi E, Valdimarsdottir G, Goumans MJ, 2003, Controlling cell fate by bone morphogenetic protein receptors. Mol Cell Endocrinol 211(1–2):105–113 Yamamoto Y, Oelgeschlager M, 2004, Regulation of bone morphogenetic proteins in early embryonic development. Naturwissenschaften 91(11):519–534 Mukhopadhyay A, McGuire T, Peng CY, Kessler JA, 2009, Differential effects of BMP signaling on parvalbumin and somatostatin interneuron differentiation. Development 136(15): 2633–2642 Mukhopadhyay P, Webb CL, Warner DR, Greene RM, Pisano MM, 2008, BMP signaling dynamics in embryonic orofacial tissue. J Cell Physiol 216(3):771–779 Shimizu T, Tanaka T, Iso T, Kawai-Kowase K, Kurabayashi M, 2009, Bone morphogenetic protein-2, BMP-2, and BMP-7 act as instructive factors to specify notch signaling controlling smooth muscle cell phenotype. Circulation 120(18):S1114 Gong SG, Guo C, 2003, Bmp4 gene is expressed at the putative site of fusion in the midfacial region. Differentiation 71(3):228– 236 Gordon KJ, Kirkbride KC, How T, Blobe GC, 2009, Bone morphogenetic proteins induce pancreatic cancer cell invasiveness through a Smad1-dependent mechanism that involves matrix metalloproteinase-2. Carcinogenesis 30(2):238–248 Beck SE, Carethers JM, 2007, BMP suppresses PTEN expression via RAS/ERK signaling. Cancer Biol Ther 6(8):1313–1317 Ma J, Sawai H, Matsuo Y, Ochi N, Yasuda A, Takahashi H, Wakasugi T, Funahashi H, Sato M, Takeyama H, 2008, IGF-1 mediates PTEN suppression and enhances cell invasion and proliferation via activation of the IGF-1/PI3K/Akt signaling pathway in pancreatic cancer cells. J Surg Res Stoll V, Calleja V, Vassaux G, Downward J, Lemoine NR, 2005, Dominant negative inhibitors of signalling through the phosphoinositol 3-kinase pathway for gene therapy of pancreatic cancer. Gut 54(1):109–116 Reichert M, Saur D, Hamacher R, Schmid RM, Schneider G, 2007, Phosphoinositide-3-kinase signaling controls S-phase kinase-associated protein 2 transcription via E2F1 in pancreatic ductal adenocarcinoma cells. Cancer Res 67(9):4149–4156 Chen PH, Yang CR, 2008, Decoy receptor 3 expression in AsPC-1 human pancreatic adenocarcinoma cells via the phosphatidylinositol 3-kinase-, Akt-, and NF-kappa B-dependent pathway. J Immunol 181(12):8441–8449 Liu DP, Zhang Y, Dang CX, Ma QY, Lee W, Chen W, 2007, siRNA directed against TrkA sensitizes human pancreatic cancer cells to apoptosis induced by gemcitabine through an inactivation of PI3K/Akt-dependent pathway. Oncol Rep 18(3):673–677 Yip-Schneider MT, Wiesenauer CA, Schmidt CM, 2003, Inhibition of the phosphatidylinositol 3′-kinase signaling pathway increases the responsiveness of pancreatic carcinoma cells to sulindac. J Gastrointest Surg 7(3):354–363 Kang MH, Kang HN, Kim JL, Kim JS, Oh SC, Yoo YA, 2009, Inhibition of PI3 kinase/Akt pathway is required for BMP2- induced EMT and invasion. Oncol Rep 22(3):525–534 Kang MH, Kim JS, Seo JE, Oh SC, Yoo YA, 2010, BMP2 accelerates the motility and invasiveness of gastric cancer cells via activation of the phosphatidylinositol 3-kinase, PI3K)/Akt pathway. Exp Cell Res 316(1):24–37 Hamada S, Satoh K, Hirota M, Kimura K, Kanno A, Masamune A, Shimosegawa T, 2007, Bone morphogenetic protein 4 induces epithelial-mesenchymal transition through MSX2 induction on pancreatic cancer cell line. J Cell Physiol 213(3):768–774 Amiri A, Noei F, Jeganathan S, Kulkarni G, Pinke DE, Lee JM, 2007, eEF1A2 activates Akt and stimulates Akt-dependent actin remodeling, invasion and migration. Oncogene 26(21):3027–3040 Liu H, Radisky DC, Nelson CM, Zhang H, Fata JE, Roth RA, Bissell MJ, 2006, Mechanism of Akt1 inhibition of breast cancer cell invasion reveals a protumorigenic role for TSC2. Proc Natl Acad Sci USA 103(11):4134–4139 Meng Q, Xia C, Fang J, Rojanasakul Y, Jiang BH, 2006, Role of PI3K and AKT specific isoforms in ovarian cancer cell migration, invasion and proliferation through the p70S6K1 pathway. Cell Signal 18(12):2262–2271 Sugimori K, Matsui K, Motomura H, Tokoro T, Wang JY, Higa S, Kimura T, Kitajima I, 2005, BMP-2 prevents apoptosis of the N1511 chondrocytic cell line through PI3K/Akt-mediated NF-kappa B activation. J Bone Miner Metab 23(6):411–419 Gilmartin B, Fox E, Mcardle E, Daly P, Hopkins A, Harrison M, Kay E, Fitzpatrick J, Dervan P, Mccann A, 2006, EMT in human bladder cancer: the fundamental role of snail transcriptional repression. BJU Int 97:7 Maddipatla S, Khoury T, Gibbs JF, Nasser E, Rutledge P, Iwata K, Black J, Javle M, 2007, Epithelial-mesenchymal transition, EMT, and role of p-Erk in surgically resected pancreatic cancer. Ann Surg Oncol 14(2):4 Roskelley CD, Somasiri AM, Tognon C, Sorensen P, Nielsen J, McNagny K, Huntsman D, Dedhar S, 2004, On the road to EMT: Genes and phenes that regulate tumor subtype-specific breast cancer progression. FASEB J 18(5):A761–A762 Spaderna S, Schmalhofer O, Hlubek F, Berx G, Eger A, Merkel S, Jung A, Kirchner T, Brabletz T, 2006, A transient, EMT-linked loss of basement membranes indicates metastasis and poor survival in colorectal cancer. Gastroenterology 131(3):830–840 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 257 EP 261 DI 10.1007/s12253-010-9307-1 PG 5 ER PT J AU Khoor, A Byrd-Gloster, LA Nicosia, VS AF Khoor, Andras Byrd-Gloster, L Angela Nicosia, V Santo TI Expression of Thyroid Transcription Factor-1 in Malignant Pleural Effusions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TTF-1; Cytology; Pleural effusions; Lung; Adenocarcinoma; Malignant mesothelioma ID TTF-1; Cytology; Pleural effusions; Lung; Adenocarcinoma; Malignant mesothelioma AB Separating adenocarcinoma of the lung from nonpulmonary adenocarcinoma or malignant mesothelioma is difficult, especially in cytology specimens. Consequently, it is important to identify markers that may facilitate this distinction. Thyroid transcription factor-1 (TTF-1) is a homeodomain containing transcription factor expressed selectively in the thyroid, lung, and diencephalon. TTF-1 is also expressed in adenocarcinomas of the lung and is widely used as a pulmonary adenocarcinoma marker in surgical specimens. However, the utility of TTF-1 has rarely been investigated in cytology. In this study, we evaluated the expression of TTF-1 in malignant pleural effusions. The primary tumors included 26 pulmonary adenocarcinomas, 26 non-pulmonary adenocarcinomas (13 breast, 5 ovarian, 2 gastric, 2 prostatic, 1 esophageal, 1 colonic, 1 pancreatic and 1 renal) and 4 malignant mesotheliomas. Immunocytochemistry was performed on sections of cell blocks, using a mouse monoclonal TTF-1 antibody (clone 8G7G3/1) and a biotin-streptavidin detection system. Nuclear immunoreactivity for TTF-1 was detected in 19 pulmonary adenocarcinomas. All nonpulmonary adenocarcinomas and malignant mesotheliomas were negative. These data indicate that TTF-1 maintains its sensitivity (73%) and specificity (100%) in cell block preparations and is useful in separating adenocarcinoma of the lung from non-pulmonary adenocarcinoma and malignant mesothelioma in cytology specimens. C1 [Khoor, Andras] Mayo Clinic, Department of Laboratory Medicine and Pathology, 4500 San Pablo Road, 32224 Jacksonville, FL, USA. [Byrd-Gloster, L Angela] Pathologists’ Laboratory, PCHermitage, TN, USA. [Nicosia, V Santo] University of South Florida, College of Medicine, Department of Pathology and Cell BiologyTampa, FL, USA. RP Khoor, A (reprint author), Mayo Clinic, Department of Laboratory Medicine and Pathology, 32224 Jacksonville, USA. EM khoor.andras@mayo.edu CR Johnston WW, 1985, The malignant pleural effusion. A review of cytopathologic diagnoses of 584 specimens from 472 consecutive patients. Cancer 56:905–909 Monte SA, Ehya H, Lang WR, 1987, Positive effusion cytology as the initial presentation of malignancy. Acta Cytol 31:448–452 Attanoos RL, Gibbs AR, 2003, ‘Pseudomesotheliomatous’ carcinomas of the pleura: a 10-year analysis of cases from the Environmental Lung Disease Research Group Cardiff. Histopathology 43:444–452 Lazzaro D, Price M, de Felice M, Di Lauro R, 1991, The transcription factor TTF-1 is expressed at the onset of thyroid and lung morphogenesis and in restricted regions of the foetal brain. Development 113:1093–1104 Stahlman MT, Gray ME, Whitsett JA, 1996, Expression of thyroid transcription factor-1(TTF-1, in fetal and neonatal human lung. J Histochem Cytochem 44:673–678 Whitsett JA, Glasser SW, 1998, Regulation of surfactant protein gene transcription. Biochim Biophys Acta 1408:303–311 Maeda Y, Dave V, Whitsett JA, 2007, Transcriptional control of lung morphogenesis. Physiol Rev 87:219–244 Katoh R, Kawaoi A, Miyagi E et al, 2000, Thyroid transcription factor-1 in normal, hyperplastic, and neoplastic follicular thyroid cells examined by immunohistochemistry and nonradioactive in situ hybridization. Mod Pathol 13:570–576 Katoh R, Miyagi E, Nakamura N et al, 2000, Expression of thyroid transcription factor-1, TTF-1, in human C cells and medullary thyroid carcinomas. Hum Pathol 31:386–393 Burke LM, Rush WI, Khoor A et al, 1999, Alveolar adenoma: a histochemical, immunohistochemical, and ultrastructural analysis of 17 cases. Hum Pathol 30:158–167 Devouassoux-Shisheboran M, Hayashi T, Linnoila RI et al, 2000, A clinicopathologic study of 100 cases of pulmonary sclerosing hemangioma with immunohistochemical studies: TTF-1 is expressed in both round and surface cells, suggesting an origin from primitive respiratory epithelium. Am J Surg Pathol 24:906–916 Holzinger A, Dingle S, Bejarano PA et al, 1996, Monoclonal antibody to thyroid transcription factor-1: production, characterization, and usefulness in tumor diagnosis. Hybridoma 15:49–53 Khoor A, Whitsett JA, Stahlman MT et al, 1999, Utility of surfactant protein B precursor and thyroid transcription factor 1 in differentiating adenocarcinoma of the lung from malignant mesothelioma. Hum Pathol 30:695–700 Folpe AL, Gown AM, Lamps LWet al, 1999, Thyroid transcription factor-1: immunohistochemical evaluation in pulmonary neuroendocrine tumors. Mod Pathol 12:5–8 Byrd-Gloster AL, Khoor A, Glass LF et al, 2000, Differential expression of thyroid transcription factor 1 in small cell lung carcinoma and Merkel cell tumor. Hum Pathol 31:58–62 Bejarano PA, Baughman RP, Biddinger PWet al, 1996, Surfactant proteins and thyroid transcription factor-1 in pulmonary and breast carcinomas. Mod Pathol 9:445–452 Moldvay J, Jackel M, Bogos K et al, 2004, The role of TTF-1 in differentiating primary and metastatic lung adenocarcinomas. Pathol Oncol Res 10:85–88 Strickland-Marmol LB, Khoor A, Livingston SK, Rojiani A, 2007, Utility of tissue-specific transcription factors thyroid transcription factor 1 and Cdx2 in determining the primary site of metastatic adenocarcinomas to the brain. Arch Pathol Lab Med 131:1686–1690 Afify AM, al-Khafaji BM, 2002, Diagnostic utility of thyroid transcription factor-1 expression in adenocarcinomas presenting in serous fluids. Acta Cytol 46:675–678 Hecht JL, Pinkus JL, Weinstein LJ, Pinkus GS, 2001, The value of thyroid transcription factor-1 in cytologic preparations as a marker for metastatic adenocarcinoma of lung origin. Am J Clin Pathol 116:483–488 Ng WK, Chow JC, Ng PK, 2002, Thyroid transcription factor-1 is highly sensitive and specific in differentiating metastatic pulmonary from extrapulmonary adenocarcinoma in effusion fluid cytology specimens. Cancer 96:43–48 Yang GC, Wan LS, Papellas J, Waisman J, 1998, Compact cell blocks. Use for body fluids, fine needle aspirations and endometrial brush biopsies. Acta Cytol 42:703–706 Armitage P, Berry G, Matthews JNS, 2002, Statistical Methods in Medical Research, Fourth edth edn. Blackwell Science, Oxford Colby TV, 2004, Adenocarcinoma. In: Travis WD, Brambilla E, Muller-Hermelink HK, Harris CC, eds, Pathology and genetics of tumours of the lung, pleura, thymus and heart. IARC, Lyon, pp 35–44 KhoorA,GrayME, HullWMet al, 1993)Developmental expression of SP-A and SP-A mRNA in the proximal and distal respiratory epithelium in the human fetus and newborn. J Histochem Cytochem 41:1311–1319 Khoor A, Gray ME, Singh G, Stahlman MT, 1996, Ontogeny of Clara cell-specific protein and its mRNA: their association with neuroepithelial bodies in human fetal lung and in bronchopulmonary dysplasia. J Histochem Cytochem 44:1429–1438 Khoor A, Stahlman MT, Gray ME, Whitsett JA, 1994, Temporalspatial distribution of SP-B and SP-C proteins and mRNAs in developing respiratory epithelium of human lung. J Histochem Cytochem 42:1187–1199 Khoor A, Whitsett JA, Stahlman MT et al, 1997, Immunoreactivity for thyroid transcription factor-1, TTF-1, correlates with better prognosis in early stage adenocarcinoma of the lung. Am J Respir Crit Care Med 155:A36, Abstract Puglisi F, Barbone F, Damante G et al, 1999, Prognostic value of thyroid transcription factor-1 in primary, resected, non-small cell lung carcinoma. Mod Pathol 12:318–324 Tan D, Li Q, Deeb G et al, 2003, Thyroid transcription factor-1 expression prevalence and its clinical implications in non-small cell lung cancer: a high-throughput tissue microarray and immunohistochemistry study. Hum Pathol 34:597–604 Weir BA, Woo MS, Getz G et al, 2007, Characterizing the cancer genome in lung adenocarcinoma. Nature 450:893–898 Kwei KA, Kim YH, Girard L et al, 2008, Genomic profiling identifies TITF1 as a lineage-specific oncogene amplified in lung cancer. Oncogene 27:3635–3640 Di Loreto C, Puglisi F, Di Lauro V et al, 1998, TTF-1 protein expression in pleural malignant mesotheliomas and adenocarcinomas of the lung. Cancer Lett 124:73–78 Pelosi G, Fraggetta F, Pasini F et al, 2001, Immunoreactivity for thyroid transcription factor-1 in stage I non-small cell carcinomas of the lung. Am J Surg Pathol 25:363–372 Ordonez NG, 2003, The immunohistochemical diagnosis of mesothelioma: a comparative study of epithelioid mesothelioma and lung adenocarcinoma. Am J Surg Pathol 27:1031–1051 Motoi N, Szoke J, Riely GJ et al, 2008, Lung adenocarcinoma: modification of the 2004 WHO mixed subtype to include the major histologic subtype suggests correlations between papillary and micropapillary adenocarcinoma subtypes, EGFR mutations and gene expression analysis. Am J Surg Pathol 32:810–827 Tsuta K, Ishii G, Nitadori J et al, 2006, Comparison of the immunophenotypes of signet-ring cell carcinoma, solid adenocarcinoma with mucin production, and mucinous bronchioloalveolar carcinoma of the lung characterized by the presence of cytoplasmic mucin. J Pathol 209:78–87 Kubba LA, McCluggage WG, Liu J et al, 2008, Thyroid transcription factor-1 expression in ovarian epithelial neoplasms. Mod Pathol 21:485–490 Zhang PJ, Gao HG, Pasha TL et al, 2009, TTF-1 expression in ovarian and uterine epithelial neoplasia and its potential significance, an immunohistochemical assessment with multiple monoclonal antibodies and different secondary detection systems. Int J Gynecol Pathol 28:10–18 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 263 EP 267 DI 10.1007/s12253-010-9308-0 PG 5 ER PT J AU Ashtiani, OZ Hasheminasab, SM Ayati, M Goulian, SB Modarressi, HM AF Ashtiani, Ousati Zahra Hasheminasab, Sayed-Mohammad Ayati, Mohsen Goulian, Sabah Bareto Modarressi, Hossein Mohammad TI Are GSTM1, GSTT1 and CAG Repeat Length of Androgen Receptor Gene Polymorphisms Associated with Risk of Prostate Cancer in Iranian Patients? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; AR gene; CAG repeat; GSTM1; GSTT1 ID Prostate cancer; AR gene; CAG repeat; GSTM1; GSTT1 AB We conducted this study to investigate whether CAG repeat length in androgen receptor gene and GSTM1 and GSTT1 polymorphisms influence prostate cancer risk in Iranian newly diagnosed cancer patients compared to age-matched BPH group and healthy individuals. DNA from 110 pathologically-confirmed prostate cancer patients, 99 age-matched men with Benign Prostatic Hyperplasia (BPH) and 100 healthy individuals were extracted and amplified by polymerase chain reaction (PCR). PCR products were examined by electrophoresis and sequencing. The mean number of CAG repeat in prostate cancer patients was significantly smaller than normal (19.9 vs 22.8; p<0.0001) and BPH groups (19.9 vs 21.9; P<0.0001) The mean difference between normal individuals and BPH group was also significant (21.9 vs. 22.8; P=0.003). Presence of GSTM1 null genotype were significantly higher in cancer and BPH group vs. normal individuals (both P values< 0.0001). there was not seen association between GSTT1 null or positive genotype with cancer risk, but analysis of GSTM1 null and GSTT1 positive in combination was statistically associated with Prostate cancer risk (OR=8.4, 95% CI 1.53–46.73). Our results showed that CAG repeat polymorphism in AR gene may act as a risk modifier and GSTM1 null genotypes also may be contributed to prostate cancer susceptibility in Iranian patients. C1 [Ashtiani, Ousati Zahra] Tehran University of Medical Sciences, Faculty of Medicine, Department of Medical GeneticsTehran, Iran. [Hasheminasab, Sayed-Mohammad] Tehran University of Medical Sciences, Faculty of Medicine, Department of Medical GeneticsTehran, Iran. [Ayati, Mohsen] Tehran University of Medical Sciences, Imam Khomeini General Hospital, Department of Urology TehranTehran, Iran. [Goulian, Sabah Bareto] Biston Ultasound ClinicTehran, Iran. [Modarressi, Hossein Mohammad] Tehran University of Medical Sciences, Faculty of Medicine, Department of Medical GeneticsTehran, Iran. RP Modarressi, HM (reprint author), Tehran University of Medical Sciences, Faculty of Medicine, Department of Medical Genetics, Tehran, Iran. EM modaresi@sina.tums.ac.ir CR Foley R, Hollywood D, Lawler M, 2004, Molecular pathology of prostate cancer: the key to identifying new biomarkers of disease. Endocr Relat Cancer 11(15369449):477–488 Visakorpi T, 2003, The molecular genetics of prostate cancer. Urology 62(14607212):3–10 Mishra D, Thangaraj K, Mandhani A, Kumar A, Mittal R, 2005, Is reduced CAG repeat length in androgen receptor gene associated with risk of prostate cancer in Indian population? Clin Genet 68(15952987):55–60 Hsing AW, Gao YT, Wu G, Wang X, Deng J, Chen YL et al, 2000, Polymorphic CAG and GGN repeat lengths in the androgen receptor gene and prostate cancer risk: a populationbased case-control study in China. Cancer Res 60(11016637):5111– 5116 Linja MJ, Visakorpi T, 2004, Alterations of androgen receptor in prostate cancer. J Steroid Biochem Mol Biol 92(15663988):255– 264 Pinarbasi H, Silig Y, Gurelik M, 2005, Genetic polymorphisms of GSTs and their association with primary brain tumor incidence. Cancer Genet Cytogenet 156(15642394):144–149 Srivastava DSL, Mandhani A, Mittal B, Mittal RD, 2005, Genetic polymorphism of glutathione S-transferase genes, GSTM1, GSTT1 and GSTP1, and susceptibility to prostate cancer in Northern India. BJU Int 95(15638917):170–173 Pandey SN, Jain M, Nigam P, Choudhuri G, Mittal B, 2006, Genetic polymorphisms in GSTM1, GSTT1, GSTP1, GSTM3 and the susceptibility to gallbladder cancer in North India. Biomarkers 11(16760134):250–261 Hayes JD, Strange RC, 2000, Glutathione S-transferase polymorphisms and their biological consequences. Pharmacology 61, 10971201):154–166 Hayes JD, Flanagan JU, Jowsey IR, 2005, Glutathione transferases. Annu Rev Pharmacol Toxicol 45(15822171):51–88 Mallick S, Romana M, Blanchet P, Multigner L, 2007, GSTM1 and GSTT1 polymorphisms and the risk of prostate cancer in a Caribbean population of African descent. Urology 69, 17572208):1165–1169 Mo Z, Gao Y, Cao Y, Gao F, Jian L, 2009, An updating metaanalysis of the GSTM1, GSTT1, and GSTP1 polymorphisms and prostate cancer: a HuGE review. Prostate 69(19143011):662–688 Gsur A, Preyer M, Haidinger G, Zidek T, Madersbacher S, Schatzl G et al, 2002, Polymorphic CAG repeats in the androgen receptor gene, prostate-specific antigen polymorphism and prostate cancer risk. Carcinogenesis 23(12376473):1647–1651 Balic I, Graham ST, Troyer DA, Higgins BA, Pollock BH, Johnson-Pais TL et al, 2002, Androgen receptor length polymorphism associated with prostate cancer risk in Hispanic men. J Urol 168(12394768):2245–2248 Hakimi JM, Schoenberg MP, Rondinelli RH, Piantadosi S, Barrack ER, 1997, Androgen receptor variants with short glutamine or glycine repeats may identify unique subpopulations of men with prostate cancer. Clin Cancer Res 3(9815849):1599– 1608 Katoh T, Yamano Y, Tsuji M, Watanabe M, 2008, Genetic polymorphisms of human cytosol glutathione S-transferases and prostate cancer. Pharmacogenomics 9(18154451):93–9104 Gsur A, Haidinger G, Hinteregger S, Bernhofer G, Schatzl G, Madersbacher S et al, 2001, Polymorphisms of glutathione- S-transferase genes, GSTP1, GSTM1 and GSTT1, and prostatecancer risk. Int J Cancer 95(11307147):152–155 Murata M, Watanabe M, Yamanaka M, Kubota Y, Ito H, Nagao M et al, 2001, Genetic polymorphisms in cytochrome P450, CYP, 1A1, CYP1A2, CYP2E1, glutathione S-transferase, GST, M1 and GSTT1 and susceptibility to prostate cancer in the Japanese population. Cancer Lett 165(11275366):171–177 Aktas D, Hascicek M, Sozen S, Ozen H, Tuncbilek E, 2004, CYP1A1 and GSTM1 polymorphic genotypes in patients with prostate cancer in a Turkish population. Cancer Genet Cytogenet 154(15381379):81–85 Acevedo C, Opazo JL, Huidobro C, Cabezas J, Iturrieta J, Quinones Sepulveda L, 2003, Positive correlation between single or combined genotypes of CYP1A1 and GSTM1 in relation to prostate cancer in Chilean people. Prostate 57, 12949934):111–117 Silig Y, Pinarbasi H, Gunes S, Ayan S, Bagci H, Cetinkaya O, 2006, Polymorphisms of CYP1A1, GSTM1, GSTT1, and prostate cancer risk in Turkish population. Cancer Investig 24, 16466991):41–45 Caceres DD, Iturrieta J, Acevedo C, Huidobro C, Varela N, Quinones L, 2005, Relationship among metabolizing genes, smoking and alcohol used as modifier factors on prostate cancer risk: exploring some gene-gene and gene-environment interactions. Eur J Epidemiol 20(15756908):79–88 Lai M-T, Chen R-H, Tsai F-J, Wan L, Chen W-C, 2005, Glutathione S-transferase M1 gene but not insulin-like growth factor-2 gene or epidermal growth factor gene is associated with prostate cancer. Urol Oncol 23(16018936):225–229 Nock NL, Liu X, Cicek MS, Li L, Macarie F, Rybicki BA et al, 2006, Polymorphisms in polycyclic aromatic hydrocarbon metabolism and conjugation genes, interactions with smoking and prostate cancer risk. Cancer Epidemiol Biomark Prev 15, 16614120):756–761 Mittal RD, Srivastava DSL, Mandhani A, Kumar A, Mittal B, 2004, Polymorphism of GSTM1 and GSTT1 genes in prostate cancer: a study from North India. Indian J Cancer 41, 15472409):115–119 Steinhoff C, Franke KH, Golka K, Thier R, Romer HC, Rotzel C et al, 2000, Glutathione transferase isozyme genotypes in patients with prostate and bladder carcinoma. Arch Toxicol 74, 11131031):521–526 Nakazato H, Suzuki K, Matsui H, Koike H, Okugi H, Ohtake N et al, 2003, Association of genetic polymorphisms of glutathione-Stransferase genes, GSTM1, GSTT1 and GSTP1, with familial prostate cancer risk in a Japanese population. Anticancer Res 23, 12926131):2897–2902 Ntais C, Polycarpou A, Ioannidis JPA, 2005, Association of GSTM1, GSTT1, and GSTP1 gene polymorphisms with the risk of prostate cancer: a meta-analysis. Cancer Epidemiol Biomark Prev 14(15668493):176–181 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 269 EP 275 DI 10.1007/s12253-010-9309-z PG 7 ER PT J AU Chatzizacharias, AN Giaginis, C Gatzidou, E Tsourouflis, G Sfiniadakis, I Alexandrou, P Theocharis, ES AF Chatzizacharias, A Nikolaos Giaginis, Constantinos Gatzidou, Elisavet Tsourouflis, Gerasimos Sfiniadakis, Ioannis Alexandrou, Paraskevi Theocharis, E Stamatios TI Expression and Clinical Significance of FAK and Src Proteins in Human Endometrial Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE FAK; Src; Clinical significance; Immunohistochemistry; Endometrial adenocarcinoma ID FAK; Src; Clinical significance; Immunohistochemistry; Endometrial adenocarcinoma AB Focal Adhesion Kinase (FAK) is a protein tyrosine kinase, localised in the focal adhesions, which, upon activation interacts with Src, another tyrosine kinase, regulating several cellular signalling pathways. Both enzymes have been implicated in malignant transformation and disease progression. The aim of the present study was to evaluate the clinical significance of FAK and Src expression in cases of endometrial adenocarcinoma. The total (t) and the activated, phosphorylated (p) forms of FAK and Src proteins were assessed immunohistochemically in tumour specimens obtained from 43 endometrial adenocarcinoma patients and were statistically analyzed in relation to various clinicopathological parameters and tumour proliferative capacity, reflected by Ki-67 labelling index. t-FAK positivity was significantly correlated with FIGO disease stage (p=0.031), and t-FAK overexpression with patients’ age (p=0.015). No statistically significant correlation was identified between t-FAK staining intensity, t-Src positivity, overexpression or staining intensity and any of the clinicopathological parameters tested. No significant correlation was found between neither the positivity nor the intensity of staining of either p-FAk or p-Src with any of the parameters under study. Nonetheless, important, but non-significant, trends were identified between t-FAK staining intensity, t-Src positivity and overexpression and patients’ survival (log rank, p=0.122, p=0.090 and p=0.057 respectively). Similarly, p-FAK and p-Src staining characteristics seemed to correlate, even though nonsignificantly, with patients’ survival (log rank, p=0.051 and p=0.070 for p-FAK and p-Src expression, respectively; log rank, p=0.134 and p=0.110 for p-FAK and p-Src staining intensity, respectively). These results support an important potential role of FAK-Src signalling in endometrial malignant disease progress and render further research in this field a necessity. C1 [Chatzizacharias, A Nikolaos] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75, Mikras Asias streetm, Goudi, GR11527 Athens, Greece. [Giaginis, Constantinos] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75, Mikras Asias streetm, Goudi, GR11527 Athens, Greece. [Gatzidou, Elisavet] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75, Mikras Asias streetm, Goudi, GR11527 Athens, Greece. [Tsourouflis, Gerasimos] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75, Mikras Asias streetm, Goudi, GR11527 Athens, Greece. [Sfiniadakis, Ioannis] Naval Hospital Athens, Department of PathologyAthens, Greece. [Alexandrou, Paraskevi] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75, Mikras Asias streetm, Goudi, GR11527 Athens, Greece. [Theocharis, E Stamatios] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75, Mikras Asias streetm, Goudi, GR11527 Athens, Greece. RP Theocharis, ES (reprint author), University of Athens, Medical School, Department of Forensic Medicine and Toxicology, GR11527 Athens, Greece. EM theocharis@ath.forthnet.gr CR Guan JL, Shalloway D, 1992, Regulation of focal adhesionassociated protein tyrosine kinase by both cellular adhesion and oncogenic transformation. Nature 358:690–692 Schaller MD, Borgman CA, Cobb BS, Vines RR, Reynolds AB, Parsons JT, 1992, pp 125FAK a structurally distinctive proteintyrosine kinase associated with focal adhesions. Proc Natl Acad Sci USA 89:5192–5196 Hanks SK, CalalbMB, Harper MC, Patel SK, 1992, Focal adhesion protein-tyrosine kinase phosphorylated in response to cell attachment to fibronectin. Proc Natl Acad Sci USA 89:8487–8491 Lipfert L, Haimovich B, Schaller MD, Cobb BS, Parsons JT, Brugge JS, 1992, Integrin-dependent phosphorylation and activation of the protein tyrosine kinase pp 125FAK in platelets. J Cell Biol 119:905–912 Zachary I, Sinnett-Smith J, Bombesin RE, 1992, Vasopressin, and endothelin stimulation of tyrosine phosphorylation in Swiss 3 T3 cells. Identification of a novel tyrosine kinase as a major substrate. J Biol Chem 267:19031–19034 Zachary I, 1997, Focal adhesion kinase. Int J Biochem Cell Biol 29:929–934 Schaller MD, Parsons JT, 1994, Focal adhesion kinase and associated proteins. Curr Opin Cell Biol 6:705–710 Salasznyk RM, Klees RF, Williams WA, Boskey A, Plopper GE, 2007, Focal adhesion kinase signaling pathways regulate the osteogenic differentiation of human mesenchymal stem cells. Exp Cell Res 313:22–37 Cary LA, Guan JL, 1999, Focal adhesion kinase in integrinmediated signaling. Front Biosci 4:D102–113 Xing Z, Chen HC, Nowlen JK, Taylor SJ, Shalloway D, Guan JL, 1994, Direct interaction of v-Src with the focal adhesion kinase mediated by the Src SH2 domain. Mol Biol Cell 5:413–421 Rahimi N, Hung W, Tremblay E, Saulnier R, Elliott B, 1998, c- Src kinase activity is required for hepatocyte growth factorinduced motility and anchorage-independent growth of mammary carcinoma cells. J Biol Chem 273:33714–33721 Hauck CR, Hunter T, Schlaepfer DD, 2001, The v-Src SH3 domain facilitates a cell adhesion-independent association with focal adhesion kinase. J Biol Chem 276:17653–17662 Frame MC, 2002, Src in cancer: deregulation and consequences for cell behaviour. Biochim Biophys Acta 1602:114–130 Hauck SDD, CR SDJ, 1999, Signaling through focal adhesion kinase. Prog Biophys Mol Biol 71:435–478 Cox BD, Natarajan M, Stettner MR, Gladson CL, 2006, New concepts regarding focal adhesion kinase promotion of cell migration and proliferation. J Cell Biochem 99:35–52 MacPhee DJ, Mostachfi H, Han R, Lye SJ, Post M, Caniggia I, 2001, Focal adhesion kinase is a key mediator of human trophoblast development. Lab Invest 81:1469–1483 Furuta Y, Ilic D, Kanazawa S, Takeda N, Yamamoto T, Aizawa S, 1995, Mesodermal defect in late phase of gastrulation by a targeted mutation of focal adhesion kinase, FAK. Oncogene 11:1989–1995 Sorenson CM, Sheibani N, 1999, Focal adhesion kinase, paxillin, and bcl-2: analysis of expression, phosphorylation, and association during morphogenesis. Dev Dyn 215:371–382 Shen T-L, Park AY-J, Alcaraz A et al, 2005, Conditional knockout of focal adhesion kinase in endothelial cells reveals its role in angiogenesis and vascular development in late embryogenesis. J Cell Biol 169:941–952 Talamonti MS, Roh MS, Curley SA, Gallick GE, 1993, Increase in activity and level of pp 60c-src in progressive stages of human colorectal cancer. J Clin Invest 91:53–60 Lutz MP, Esser IB, Flossmann-Kast BB et al, 1998, Overexpression and activation of the tyrosine kinase Src in human pancreatic carcinoma. Biochem Biophys Res Commun 243:503–508 Verbeek BS, Vroom TM, Adriaansen-Slot SS et al, 1996, c-Src protein expression is increased in human breast cancer. An immunohistochemical and biochemical analysis. J Pathol 180:383–388 Ito H, Gardner-Thorpe J, Zinner MJ, Ashley SW, Whang EE, 2003, Inhibition of tyrosine kinase Src suppresses pancreatic cancer invasiveness. Surgery 134:221–226 Myoui A, Nishimura R, Williams PJ et al, 2003, C-SRC tyrosine kinase activity is associated with tumor colonization in bone and lung in an animal model of human breast cancer metastasis. Cancer Res 63:5028–5033 Duxbury MS, Ito H, Zinner MJ, Ashley SW, Whang EE, 2004, Inhibition of SRC tyrosine kinase impairs inherent and acquired gemcitabine resistance in human pancreatic adenocarcinoma cells. Clin Cancer Res 10:2307–2318 Chatzizacharias NA, Kouraklis GP, Theocharis SE, 2008, Clinical significance of FAK expression in human neoplasia. Histol Histopathol 23:629–650 Laird AD, Cherrington JM, 2003, Small molecule tyrosine kinase inhibitors: clinical development of anticancer agents. Expert Opin Investig Drugs 12:51–64 Chatzizacharias NA, Kouraklis GP, Theocharis SE, 2007, Focal adhesion kinase: a promising target for anticancer therapy. Expert Opin Ther Targets 11:1315–1328 Livasy CA, Moore D, Cance WG, Lininger RA, 2004, Focal adhesion kinase overexpression in endometrial neoplasia. Appl Immunohistochem Mol Morphol 12:342–345 Su JM, Gui L, Zhou YP, Zha XL, 2002, Expression of focal adhesion kinase and alpha5 and beta1 integrins in carcinomas and its clinical significance. World J Gastroenterol 8:613–618 Giaginis CT, Vgenopoulou S, Tsourouflis GS, Politi EN, Kouraklis GP, Theocharis SE, 2009, Expression and clinical significance of focal adhesion kinase in the two distinct histological types, intestinal and diffuse, of human gastric adenocarcinoma. Pathol Oncol Res 15:173–181 Chatzizacharias NA, Giaginis C, Diamanto Zizi-Serbetzoglou D et al, 2010, Evaluation of the clinical significance of Focal Adhesion Kinase and Src expression in human pancreatic ductal adenocarcinoma. Pancreas 39:930–936 Macwhinnie N, Monaghan H, 2004, The use of P53, PTEN, and C-erbB-2 to differentiate uterine serous papillary carcinoma from endometrioid endometrial carcinoma. Int J Gynecol Cancer 14:938–946 Sobel G, Nemeth J, Kiss A, Lotz G, Szabo I, Udvarhelyi N, Schaff Z, Paska C, 2006, Claudin 1 differentiates endometrioid and serous papillary endometrial adenocarcinoma. Gynec Oncol 103:591–598 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 277 EP 285 DI 10.1007/s12253-010-9310-6 PG 9 ER PT J AU Ieni, A Barresi, V Grosso, M Speciale, G Rosa, AM Tuccari, G AF Ieni, Antonio Barresi, Valeria Grosso, Maddalena Speciale, Giuseppe Rosa, A Michele Tuccari, Giovanni TI Does Lactoferrin Behave as an Immunohistochemical Oncofetal Marker in Bone and Cartilage Human Neoplasms? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bone tumours; Fetal tissue; Immunohistochemistry; Lactoferrin; Oncofetal antigens ID Bone tumours; Fetal tissue; Immunohistochemistry; Lactoferrin; Oncofetal antigens AB By immunohistochemistry, lactoferrin (LF) has been extensively investigated in human neoplastic tissues; moreover, LF is able to promote bone growth in a murine model. Until now, no systematic studies on human osteocartilagineous fetal samples have been performed in comparison to corresponding neoplastic specimens to verify if LF may represent an oncofetal marker in this field of pathology. By a monoclonal antibody (clone 1A1; Biodesign International; w.d. 1:75) the distribution pattern of LF in bones of 25 human fetal tissues (8–34 gestation weeks), 10 adults (47–82 years) and 30 cartilage as well as 27 bone tumours (9–76 years) was analyzed. LF was encountered in 23/57 cases of osteocartilagineous tumors and namely in 10/10 giant cell tumours, 5/7 osteoid osteomas, 3/3 chondroblastomas, 3/3 chondromyxoid fibromas, 1/1 myeloma, 1/1 adamantinoma. No LF immunoexpression was detected in osteosarcomas, chondrosarcomas, ossifying fibromas, osteochondroma and enchondromas. In embryo-fetal tissues, LF immunoreactivity was localized in mesenchymal cells as well as in chondroblasts at the 8th gestational week and in immature osteocytes and osteoblasts up to the 18th gestation week, with a considerable decrease by the 24th week. No LF expression was found in any bone district since the 30th and up to the 34th week of gestation as well as in corresponding adult samples. Our findings indicate a role for LF as a bone growth regulator in the early phases of the human endochondral ossification, although the hypothesis of LF as oncofetal marker appears questionable in bone tumours. C1 [Ieni, Antonio] Azienda Ospedaliera Universitaria, Department of Human Pathology, Pad. D, Via Consolare Valeria, 98125 Messina, Italy. [Barresi, Valeria] Azienda Ospedaliera Universitaria, Department of Human Pathology, Pad. D, Via Consolare Valeria, 98125 Messina, Italy. [Grosso, Maddalena] Azienda Ospedaliera Universitaria, Department of Human Pathology, Pad. D, Via Consolare Valeria, 98125 Messina, Italy. [Speciale, Giuseppe] Azienda Ospedaliera Universitaria, Department of Human Pathology, Pad. D, Via Consolare Valeria, 98125 Messina, Italy. [Rosa, A Michele] Azienda Ospedaliera Universitaria, Orthopaedics Clinic, Pad. D, Via Consolare Valeria, 98125 Messina, Italy. [Tuccari, Giovanni] Azienda Ospedaliera Universitaria, Department of Human Pathology, Pad. D, Via Consolare Valeria, 98125 Messina, Italy. RP Ieni, A (reprint author), Azienda Ospedaliera Universitaria, Department of Human Pathology, 98125 Messina, Italy. EM calaienco@hotmail.com CR de Lellis RA, Wolfe HJ, 1981, The pathobiology of the human calcitonin, C)-cell: a review. Pathol Annu 16(Pt 2):25–52 Herberman RB, 1977, Immunogenicity of tumor antigens. Biochim Biophys Acta 473(2):93–119 Sarandakou A, Protonotariou E, Rizos D, 2007, Tumor markers in biological fluids associated with pregnancy. Crit Rev Clin Lab Sci 44(2):151–178 Monk M,Holding C, 2001)Human embryonic genes re-expressed in cancer cells. Oncogene 20(56):8085–8091 Mueller-Pillasch F, Lacher U, Wallrapp C et al, 1997, Cloning of a gene highly overexpressed in cancer coding for a novel KHdomain containing protein. Oncogene 14(22):2729–2733 Jiang Z, Lohse CM, Chu PG et al, 2008, Oncofetal protein IMP3: a novel molecular marker that predicts metastasis of papillary and chromophobe renal cell carcinomas. Cancer 112(12):2676– 2682 Ward PP, Mendoza-Meneses M, Mulac-Jericevic B et al, 1999, Restricted spatiotemporal expression of lactoferrin during murine embryonic development. Endocrinology 140:1852–1860 Cornish J, Callon KE, Naot D et al, 2004, Lactoferrin is a potent regulator of bone cell activity and increases bone formation in vivo. Endocrinology 145:366–374 Cornish J, Palmano K, Callon KE et al, 2006, Lactoferrin and bone; structureactivity relationships. Biochem Cell Biol 84:297– 302 Naot D, Grey A, Reid IR et al, 2005, Lactoferrin—a novel bone growth factor. Clin Med Res 3:93–101 Giuffre G, Barresi V, Skliros C et al, 2007, Immunoexpression of lactoferrin in human sporadic renal cell carcinomas. Oncol Rep 17:1021–1026 Baker EN, Baker HM, Kidd RD, 2002, Lactoferrin and transferrin: functional variations on a common structural framework. Biochem Cell Biol 80(1):27–34 Masson PL, Heremans JF, 1971, Lactoferrin in milk from different species. Comp Biochem Physiol B 39(1):119–129 Lonnerdal B, 2009, Nutritional roles of lactoferrin. Curr Opin Clin Nutr Metab Care 12(3):293–297 Pierce A, Legrand D, Mazurier J, 2009, Lactoferrin: a multifunctional protein. Med Sci 25(4):361–369 Rodrigues L, Teixeira J, Schmitt F et al, 2009, Lactoferrin and cancer disease prevention. Crit Rev Food Sci Nutr Mar 49, 3):203–217 Benaissa M, Peyrat JP, Hornez L et al, 2005, Expression and prognostic value of lactoferrin mRNA isoforms in human breast cancer. Int J Cancer 114(2):299–306 Wolf JS, Li D, Taylor RJ, O'Malley BW Jr, 2003, Lactoferrin inhibits growth of malignant tumors of the head and neck. ORL J Otorhinolaryngol Relat Spec 65(5):245–249 Xiao Y, Monitto CL, Minhas KM et al, 2004, Lactoferrin down-regulates G1 cyclin-dependent kinases during growth arrest of head and neck cancer cells. Clin Cancer Res 10(24):8683– 8686 Caselitz J, Jaup T, Seifert G, 1981, Lactoferrin and lysozyme in carcinomas of the parotid gland. A comparative immunocytochemical study with the occurrence in normal and inflamed tissue. Virchows Arch A Pathol Anat Histol 394(1–2):61–73 Barresi G, Tuccari G, 1984, Lactoferrin in benign hypertrophy and carcinomas of the prostatic gland. Virchows Arch A Pathol Anat Histopathol 403:59–66 Charpin C, Lachard A, Pourreau-Schneider N et al, 1985, Localization of lactoferrin and nonspecific cross-reacting antigen in human breast carcinomas. An immunohistochemical study using the avidin-biotin-peroxidase complex method. Cancer 55, 11):2612–2617 Rossiello R, Carriero MV, Giordano GG, 1984, Distribution of ferritin, transferrin and lactoferrin in breast carcinoma tissue. J Clin Pathol 37(1):51–55 Barresi G, Tuccari G, 1987, Iron-binding proteins in thyroid tumours. An immunocytochemical study Pathol Res Pract 182, 3):344–351 Cabaret V, Vilain MO, Delobelle-Deroide A et al, 1992, Immunohistochemical demonstration of ceruloplasmin and lactoferrin in a series of 59 thyroid tumors. Ann Pathol 12(6):347–352 Tuccari G, Barresi G, 1985, Immunohistochemical demonstration of lactoferrin in follicular adenomas and thyroid carcinomas. Virchows Arch A Pathol Anat Histopathol 406:67–74 Tuccari G, Barresi G, Arena F et al, 1989, Immunocytochemical detection of lactoferrin in human gastric carcinomas and adenomas. Arch Pathol Lab Med 113:912–915 Tuccari G, Rizzo A, Crisafulli C et al, 1992, Iron-binding proteins in human colorectal adenomas and carcinomas: an immunocytochemical investigation. Histol Histopathol 7:543–547 Tuccari G, Rossiello R, Barresi G, 1997, Iron binding proteins in gallbladder carcinomas. An immunocytochemical investigation. Histol Histopathol 12:671–676 Tuccari G, Giuffre G, Crisafulli C et al, 1999, Immunohistochemical detection of lactoferrin in human astrocytomas and multiforme glioblastomas. Eur J Histochem 43(4):317–322 Tuccari G, Giuffre G, Scarf R et al, 2005, Immunolocalization of lactoferrin in surgically resected pigmented skin lesions. Eur J Histochem 49(1):33–38 Giuffre G, Arena F, Scarfi R et al, 2006, Lactoferrin immunoexpression in endometrial carcinomas: relationships with sex steroid hormone receptors, ER and PR), proliferation indices, Ki-67 and AgNOR, and survival. Oncol Rep 16(2):257–263 Ieni A, Barresi V, Grosso M et al, 2009, Lactoferrin immunoexpression in human normal and neoplastic bone tissue. J Bone Miner Metab 27:364–371 Ieni A, Barresi V, Grosso M et al, 2009, Immunolocalization of lactoferrin in cartilageforming neoplasms. J Orthop Sci 14:732– 737 Garre C, Bianchi-Scarra G, Sirito M et al, 1992, Lactoferrin binding sites and nuclear localization in K562(S, cells. J Cell Physiol 153(3):477–482 Penco S, Scarfi S, Giovine M et al, 2001, Identification of an import signal for, and the nuclear localization of, human lactoferrin. Biotechnol Appl Biochem 34:151–159 James IE, Dodds RA, Olivera DL et al, 1996, Human osteoclastoma-derived stromal cells: correlation of the ability to form mineralized nodules in vitro with formation of bone in vivo. J Bone Miner Res 11(10):1453–1460 Joyner CJ, Quinn JM, Triffitt JT et al, 1992, Phenotypic characterisation of mononuclear and multinucleated cells of giant cell tumour of bone. Bone Miner 16(1):37–48 Pittenger MF, Mackay AM, Beck SC et al, 1999, Multilineage potential of adult human mesenchymal stem cells. Science 284, 5411):143–147 Karsenty G, 2008, Transcriptional control of skeletogenesis. Annu Rev Genomics Hum Genet 9:183–196 Wulling M, Delling G, Kaiser E, 2003, The origin of the neoplastic stromal cell in giant cell tumor of bone. Hum Pathol 34(10):983–993 Aigner T, 2007, Histogenesis of the skeleton and morphogenesis of cartilage-forming neoplasias. Verh Dtsch Ges Pathol 91:49–56 Yagi M, Suzuki N, Takayama T, 2009, Effects of lactoferrin on the differentiation of pluripotent mesenchymal cells. Cell Biol Int 33(3):283–289 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 287 EP 293 DI 10.1007/s12253-010-9311-5 PG 7 ER PT J AU Bouanene, H Hadj Kacem, H Ben Fatma, L Ben Limem, H Ben Ahmed, S Yakoub, S Miled, A AF Bouanene, Houda Hadj Kacem, Hassen Ben Fatma, Leila Ben Limem, Halima Ben Ahmed, Slim Yakoub, Salwa Miled, Abdelhedi TI Polymorphisms in the MUC16 Gene: Potential Implication in Epithelial Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MUC16; Polymorphism; CA125; Epithelial ovarian cancer ID MUC16; Polymorphism; CA125; Epithelial ovarian cancer AB MUC16 plays an important role in epithelial ovarian cancer. In this paper, we studied the association between two tags SNPs of MUC16 and the risk of epithelial ovarian cancer. We aimed also to test the association between these tags SNPs and elevated level of the protein CA125. We analyzed a collection of 117 cases. Forty-one samples of patients with epithelial ovarian cancer and 76 samples from Tunisian volunteers were genotyped for two synonymous coding tags SNPs of the MUC16 gene (rs1596797, A/C and rs2547065, C/G) using polymerase chain reaction and sequencing. For the rs1596797 SNP, there was no significant difference in genotype distribution, a rare variation observed in only one patient. For the polymorphism rs2547065, mean CA125 levels were 24 and 78 UI/ml in patients with GG and GC genotypes versus 230 UI/ml in patients with CC genotype (P=0.36). Compared to the C/C genotype, the ‘G’ allele (C/G+G/G genotypes) did not significantly modified the risk of developping epithelial ovarian cancer (OR=0.43; 95% CI). As for the polymorphism rs1596797, compared to the C/C genotype, the ‘A’ allele (C/A+A/A genotypes) did not significantly modified the risk of developping epithelial ovarian cancer (OR=881.7; 95% CI). MUC16 gene polymorphisms selected in this study are neither involved in genetic predisposition to epithelial ovarian cancer nor associated with CA125 level. C1 [Bouanene, Houda] CHU Farhat Hached, Laboratory of Biochemistry, 4000 Sousse, Tunisia. [Hadj Kacem, Hassen] University of Sfax, Centre de Biotechnologie de SfaxSfax, Tunisia. [Ben Fatma, Leila] CHU Farhat Hached, Service of OncologySousse, Tunisia. [Ben Limem, Halima] CHU Farhat Hached, Laboratory of Biochemistry, 4000 Sousse, Tunisia. [Ben Ahmed, Slim] CHU Farhat Hached, Service of OncologySousse, Tunisia. [Yakoub, Salwa] Center of Blood TransfusionSousse, Tunisia. [Miled, Abdelhedi] CHU Farhat Hached, Laboratory of Biochemistry, 4000 Sousse, Tunisia. RP Bouanene, H (reprint author), CHU Farhat Hached, Laboratory of Biochemistry, 4000 Sousse, Tunisia. EM bouanenehouda2@yahoo.fr CR Boyd J, 1998, Molecular genetics of hereditary ovarian cancer. Oncology 12:399–406 Risch HA, McLaughlin JR, Cole D et al, 2001, Prevalence and penetrance of germline BRCA1 and BRCA2 mutations in a population series of 649 women with ovarian cancer. Am J Hum Genet 68:700–710 Sekine M, Nagata H, Tsuji S et al, 2001, Localization of a novel susceptibility gene for familial ovarian cancer to chromosome 3p22-25. Hum Mol Genet 10:1421–1429 Sellers TA, Huang Y, Cunningham J et al, 2008, Association of single nucleotide polymorphisms in glycosylation genes with risk of epithelial ovarian cancer. Cancer Epidemiol Biomark Prev 17:397–404 Gubbels JAA, Belisle J, Onda M et al, 2006, Mesothelin-MUC16 binding is a high affinity. N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol Cancer 5:50 Boivin M, Lane D, Piche A et al, 2009, CA125, MUC16, tumor antigen selectivelymodulates the sensitivity of ovarian cancer cells to genotoxic drug-induced apoptosis. Gynecol Oncol 115:407–413 Belisle J, Gubbels JAA, Raphae CA et al, 2007, Peritoneal natural killer cells from epithelial ovarian cancer patients show an altered phenotype and bind to the tumour marker MUC16, CA125). Immunology 122:418–429 Kaneko SJ,Gerasimova T, Smith Tet al, 2003, CA125 andUQCRFS1 FISH studies of ovarian carcinoma. Gynecol Oncol 90:29–36 O’Brien TJ, Beard JB, Underwood LJ et al, 2001, The CA125 gene: an extracellular superstructure dominated by repeat sequences. Tumor Biol 22:348–366 Yin BWT, Lloyd KO, 2001, Molecular cloning of the CA125 ovarian cancer antigen, identification as a new mucin, MUC16. J Biol Chem 276:27371–27375 Porchet N, Aubert JP, 2004, Les genes MUC: mucin or not mucin? That is the question. Med Sci 20:569–574 Gabriel SB, Schaffner SF, Nguyen H et al, 2002, The structure of haplotype blocks in the human genome. Science 296:2225–2229 Carlson CS, Eberle MA, Rieder MJ et al, 2004, Selecting a maximally informative set of single-nucleotide polymorphisms for association analyses using linkage disequilibrium. Am J Hum Genet 74:106–120 Seelenmeyer C, Wegehingel S, Lechner J et al, 2003, The cancer antigen CA125 represents a novel counter receptor for galectin-1. J Cell Sci 116:1305–1318 Rustin GJ, Marples M, Nelstrop AE et al, 2001, Use of CA-125 to define progression of ovarian cancer in patients with persistently elevated levels. J Clin Oncol 19:4054–4057 Hefler LA, Ludwig E, Lebrecht A et al, 2002, Polymorphisms of the interleukin-1 gene cluster and ovarian cancer. J Soc Gynecol Investig 9:386–390 Wong NK, Easton RL, Panico M et al, 2003, Characterization of the oligosaccharides associated with the human ovarian tumor marker CA125. J Biol Chem 278:28619–28634 Ubell ML, Khampang P, Kerschner JE, 2010, Mucin gene polymorphisms in otitis media patients. Laryngoscope 120:132– 138 Korbi S, Descoteaux-Chatti D, 1995, Registre du cancer du centre tunisien, Le cancer dans le centre tunisien, 1er janvier 1987−31 decembre 1993 McLemore MR, Aouizerat B, 2005, Introducing the MUC16 gene: implications for prevention and early detection in epithelial ovarian cancer. Biol Res Nur 6:262–267 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 295 EP 299 DI 10.1007/s12253-010-9314-2 PG 5 ER PT J AU Biro, A Fodor, Z Major, J Tompa, A AF Biro, Anna Fodor, Zoltan Major, Jeno Tompa, Anna TI Immunotoxicity Monitoring of Hospital Staff Occupationally Exposed to Cytostatic Drugs SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD antigens; Cytostatic drug; Immunotoxicity; Lymphocyte phenotype; Occupational exposure; Oxidative burst ID CD antigens; Cytostatic drug; Immunotoxicity; Lymphocyte phenotype; Occupational exposure; Oxidative burst AB The aim of our study was to investigate the immunotoxicity of occupational cytostatic drug exposure, and to assess the possible effect of confounding factors, such as age and smoking. In this human study, the immunotoxic effect of antineoplastic drugs was investigated among 306 nurses working in oncology chemotherapy units. Results were compared to 98 non-exposed women. The immune status of the subjects was characterized by immune phenotyping of peripheral blood lymphocytes by flow cytometry, using monoclonal antibodies against surface antigens (CD3, CD4, CD8, CD19, CD25, CD45, CD56 and CD71). The killing ability of neutrophil leukocytes was assessed by the measurement of reactive oxygen intermediate production. Occupational exposure to antineoplastic drugs caused shifts in the major lymphocyte subpopulations, resulting in a statistically significant increase in the ratio of B cells. Cytostatic drug exposure also manifested itself in a decreased frequency of CD25 positive, activated T lymphocytes, and increased oxidative burst of neutrophil granulocytes, both of which may have a functional impact on the immune system of exposed subjects. In the younger subjects exposure also caused a shift in T cell subpopulations: a reduction in the cytotoxic T cell population lead to an elevated Th/Tc ratio. In the exposed group, smoking increased activation of T lymphocyte subpopulations. In conclusion, we have demonstrated that low dose occupational cytostatic drug exposure is immunotoxic, and age and smoking modify the effect of exposure. C1 [Biro, Anna] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Kemiai Biztonsagi Intezet, Citogenetikai es Molekularis Toxikologiai Osztaly, Nagyvarad ter 2, H-1096 Budapest, Hungary. [Fodor, Zoltan] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Kemiai Biztonsagi Intezet, Citogenetikai es Molekularis Toxikologiai Osztaly, Nagyvarad ter 2, H-1096 Budapest, Hungary. [Major, Jeno] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Kemiai Biztonsagi Intezet, Citogenetikai es Molekularis Toxikologiai Osztaly, Nagyvarad ter 2, H-1096 Budapest, Hungary. [Tompa, Anna] Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Kemiai Biztonsagi Intezet, Citogenetikai es Molekularis Toxikologiai Osztaly, Nagyvarad ter 2, H-1096 Budapest, Hungary. RP Biro, A (reprint author), Fodor Jozsef Orszagos Kozegeszsegugyi Kozpont, Orszagos Kemiai Biztonsagi Intezet, Citogenetikai es Molekularis Toxikologiai Osztaly, H-1096 Budapest, Hungary. EM biro.anna@okbi.antsz.hu CR McDevitt JJ, Lees PS, McDiarmid MA, 1993, Exposure of hospital pharmacists and nurses to antineoplastic agents. J Occup Med 35:57–60 Neal AW, Wadden RA, Chlou WL, 1983, Exposure of hospital workers to airborne antineoplastic agents. Am J Hosp Pharm 40:597–601 Hirst M, Mills DG, Tse S, Levin L, White DF, 1984, Occupational exposure to cyclophosphamide. Lancet 1:186–188 IARC monographs on the evaluation of carcinogenic risks to humans, volume 76, 2000. Some antiviral and antineoplastic drugs, and other pharmaceutical agents, WHO International Agency for Research on Cancer Sorsa M, Hemminki K, Vainio H, 1985, Occupational exposure to anticancer drugs—potential and real hazards. Mutat Res 154:135– 149 Jakab MG, Major J, Tompa A, 2001, Follow-up genotoxicological monitoring of nurses handling antineoplastic drugs. J Toxicol Environ Health 62:307–318 Connor TH, McDiarmid MA, 2006, Preventing occupational exposures to antineoplastic drugs in health care settings. CA Cancer J Clin 56:354–365 Sorsa M, Anderson D, 1996, Monitoring of occupational exposure to cytostatic anticancer agents. Mutat Res 355:253–261 Sabbioni ME, Castiglione M, Hurny C, Siegrist HP, Bacchi M, Bernhard J et al, 1999, Interaction of tamoxifen with concurrent cytotoxic adjuvant treatment affects lymphocytes and lymphocyte subsets counts in breast cancer patients. Support Care Cancer 7:149–153 Tong AW, Seamour B, Lawson JM, Ordonez G, Vukelja S, Hyman W et al, 2000, Cellular immune profile of patients with advanced cancer before and after taxane treatment. Am J Clin Oncol 23:463–472 Zielinski CC, Muller C, Kubista E, Staffen A, Eibl MM, 1990, Effects of adjuvant chemotherapy on specific and non-specific immune mechanisms. Acta Med Austriaca 17:11–14 Spatari G, Fenga C, Minciullo PL, Di Pasquale G, Cacciola A, Ventura-Spagnolo E et al, 2005, Modification of interleukin-15 serum levels in workers exposed to chemotherapeutic agents. Mediators Inflamm 1:60–62 Biro A, Pallinger E, Major J, Jakab MG, Klupp T, Falus A, Tompa A, 2002, Lymphocyte phenotype analysis and chromosome aberration frequency of workers occupationally exposed to styrene, benzene, polycyclic aromatic carbohydrates or mixed solvents. Immunol Lett 81:133–140 Biro A, Pallinger E, Falus A, Tompa A, 2004, Characterization of chemically exposed groups by immunotoxicological methods. Hungarian Oncology 48:137–139 Tompa A, Jakab M, Biro A, Magyar B, Fodor Z, Klupp T, Major J, 2006, Chemical safety and health conditions among Hungarian hospital nurses. Ann N Y Acad Sci 1076:635–648 Mackall CL, 2000, T-cell immunodeficiency following cytotoxic antineoplastic therapy: a review. Stem Cells 18:10–18 Mackall CL, Fleisher TA, Brown MR, Magrath IT, Shad AT, Horowitz ME et al, 1994, Lymphocyte depletion during treatment with intensive chemotherapy for cancer. Blood 84:2221–2228 Smith KA, 1988, Interleukin-2, inception, impact, and implications. Science 240:1169–1176 Takeshita T, Asao H, Ohtani K, Ishii N, Kumaki S, Tanaka N et al, 1992, Cloning of the gamma chain of the human IL-2 receptor. Science 257:379–382 Siegel JP, Sharon M, Smith PL, Leonard WJ, 1987, The IL-2 receptor beta chain, p70): role in mediating signals for LAK, NK, and proliferative activities. Science 238:75–78 Kondo M, Takeshita T, Higuchi M, Nakamura M, Sudo T, Nishikawa S et al, 1994, Functional participation of the IL-2 receptor gamma chain in IL-7 receptor complexes. Science 263:1453–1454 Leonard WJ, Kronke M, Peffer NJ, Depper JM, Greene WC, 1985, Interleukin 2 receptor gene expression in normal human T lymphocytes. Proc Natl Acad Sci USA 82:6281–6285 Lan RY, Selmi C, Gershwin ME, 2008, The regulatory, inflammatory, and T cell programming roles of interleukin-2, IL-2). J Autoimmun 31:7–12 Regeczy N, Gorog G, Paloczi K, 2001, Developing an expert system for immunophenotypical diagnosis in immunodeficiency. Age-related reference values of peripheral blood lymphocyte subpopulations in Hungary. Immunol Lett 77:47–54 Freedman DS, Flanders WD, Barboriak JJ, Malarcher AM, Gates L, 1996, Cigarette smoking and leukocyte subpopulations in men. Ann Epidemiol 6:299–306 Schwartz J, Weiss ST, 1994, Cigarette smoking and peripheral blood leukocyte differentials. Ann Epidemiol 4:236–242 Hughes DA, Haslam PL, Townsend PJ, Turner-Warwick M, 1985, Numerical and functional alterations in circulatory lymphocytes in cigarette smokers. Clin Exp Immunol 61:459–466 Schaberg T, Theilacker C, Nitschke OT, Lode H, 1997, Lymphocyte subsets in peripheral blood and smoking habits. Lung 175:387–394 Zavitz CCJ, Gaschler GJ, Robbins CS, Botelho FM, Cox PG, Stampfli MR, 2008, Impact of cigarette smoke on T and B cell responsiveness. Cell Immunol 253:38–44 Bijl M, Horst G, Limburg PC, Kallenberg CGM, 2001, Effects of smoking on activation markers, Fas expression and apoptosis of peripheral blood lymphocytes. Eur J Clin Invest 31:550–553 Tanigawa T, Araki S, Sata F, Nakata A, Araki T, 1998, Effects of smoking, aromatic amines, and chromates on CD4+ and CD8+ T lymphocytes in male workers. Environ Res, Section A 78:59–63 Tanigawa T, Araki S, Nakata A, Sakurai S, 1998, Increase in the helper inducer, CD4+CD29+, T lymphocytes in smokers. Ind Health 36:78–81 Condliffe AM, Hawkins PT, Stephens LR, Haslett C, Chilvers ER, 1998, Priming of human neutrophil superoxide generation by tumour necrosis factor-α is signalled by enhanced phosphatidylinositol 3, 4, 5-trisphosphate but not inositol 1, 4, 5-trisphosphate accumulation. FEBS Lett 439:147 Smedly LA, Tonnesen MG, Sandhaus RA, Haslett C, Guthrie LA, Johnston RB Jr et al, 1986, Neutrophil-mediated injury to endothelial cells: enhancement by endotoxin and essential role of neutrophil elastase. J Clin Invest 77:1233 Worthen GS, Haslett C, Smedly LA, Rees AJ, Gumbay RS, Henson JE et al, 1986, Lung vascular injury induced by chemotactic factors: enhancement by bacterial endotoxins. Fed Proc 45:7 Lee WL, Downey GP, 2001, Neutrophil activation and acute lung injury. Curr Opin Crit Care 7:1 Shortridge LA, Lemasters GK, Valanis B, Hertzberg V, 1995, Menstrual cycles in nurses handling antineoplastic drugs. Cancer Nurs 18:439–444 Valanis B, Vollmer W, Labuhn K, Glass A, 1997, Occupational exposure to antineoplastic agents and self-reported infertility among nurses and pharmacists. J Occup Environ Med 39:574– 580 Krstev S, Perunicic B, Vidakovic A, 2003, Work practice and some adverse health effects in nurses handling antineoplastic drugs. Med Lav 94:432–439 Schwenk M, Sack U, Esser C, Klein R, 2007, Diagnostic relevance of the determination of lymphocyte subpopulations in environmental medicineInt. J Hyg Environ Health 210:177– 198 Environmental Health Criteria 180: Principles and methods for assessing direct immunotoxicity associated with exposure to chemicals. World Health Organization, Geneva, 1992 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 301 EP 308 DI 10.1007/s12253-010-9317-z PG 8 ER PT J AU Demir, R Peros, G Hohenberger, W AF Demir, Resit Peros, Georgios Hohenberger, Werner TI Definition of the “Drug-Angiogenic-Activity-Index” that Allows the Quantification of the Positive and Negative Angiogenic Active Drugs: A Study Based on the Chorioallantoic Membrane Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Angiogenesis; Chorioallantoic membrane; Drug angiogenic index; Antiangiogenic therapy ID Angiogenesis; Chorioallantoic membrane; Drug angiogenic index; Antiangiogenic therapy AB Since the introduction of the angiogenic therapy by Folkman et al. in the 1970’ies many antiangiogenic drugs were identified. Only few of them are still now in clinical use. Also the Vascular Endothelial Growth Factor (VEGF), the cytokine with the highest angiogenic activity, has been identified. Its antagonist, Bevacizumab, is produced and admitted for the angiogenic therapy in first line for metastatic colorectal cancer. When we look at preclinical studies, they fail of in vivo models that define the "Drug-Angiogenic-Activity-Index" of angiogenic or antiangiogenic drugs. This work proposes a possible standardized procedure to define the "Drug Angiogenic Activity Index" by counting the vascular intersections (VIS) on the Chorioallantoic Membrane after drug application. The equation was defined as follows: {ΔVIS[Drug]−ΔVIS[Control]} / Δ VIS[Control]. For VEGF a Drug-Angiogenic-Activity-Index of 0.92 was found and for Bevacizumab a −1. This means almost that double of the naturally angiogenic activity was achieved by VEGF on the Chorioallantoic membrane. A complete blocking of naturally angiogenic activity was observed after Bevacizumabs application. Establishing the "Drug-Angiogenic-Activity-Index" in the preclinical phase will give us an impact of effectivness for the new constructed antiangiogenic drugs like the impact of effectiveness in the cortisone family. C1 [Demir, Resit] University of Erlangen, Department of Surgery, Krankenhausstrasse 12, 91054 Erlangen, Germany. [Peros, Georgios] University of Erlangen, Department of Surgery, Krankenhausstrasse 12, 91054 Erlangen, Germany. [Hohenberger, Werner] University of Erlangen, Department of Surgery, Krankenhausstrasse 12, 91054 Erlangen, Germany. RP Demir, R (reprint author), University of Erlangen, Department of Surgery, 91054 Erlangen, Germany. EM r.demir@gmx.net CR Folkman J, 1971, Tumor angiogenesis: therapeutic implications. N Engl J Med 285:1182–1186 Folkman J, 1974, Tumor angiogenesis. Adv Cancer Res 19:331–358 Hirschmann R, 1992, The cortisone era: aspects of its impact. Some contributions of the Merck Laboratories. Steroids 57:579–592 Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E, Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F, 2004, Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350:2335–2342 Saltz LB, Lenz HJ, Kindler HL, Hochster HS, Wadler S, Hoff PM, Kemeny NE, Hollywood EM, Gonen M, Quinones M, Morse M, Chen HX, 2007, Randomized phase II trial of cetuximab, bevacizumab, and irinotecan compared with cetuximab and bevacizumab alone in irinotecan-refractory colorectal cancer: the BOND-2 study. J Clin Oncol 25:4557–4561 Ribatti D, Vacca A, Roncali L, Dammacco F, 1996, The chick embryo chorioallantoic membrane as a model for in vivo research on angiogenesis. Int J Dev Biol 40:1189–1197 Wilting J, Christ B, Bokeloh M, 1991, A modified chorioallantoic membrane, CAM, assay for qualitative and quantitative study of growth factors. Studies on the effects of carriers, PBS, angiogenin, and bFGF. Anat Embryol, Berl, 183:259–271 Demir R, Hoper J, 1997, Effect of beta-interferon on vascular density, mitochondrial metabolism and alkaline phosphatase in normoxia and hypoxia. Adv Exp Med Biol 428:439–447 Ribatti D, Gualandris A, Bastaki M, Vacca A, Iurlaro M, Roncali L, Presta M, 1997, New model for the study of angiogenesis and antiangiogenesis in the chick embryo chorioallantoic membrane: the gelatin sponge/chorioallantoic membrane assay. J Vasc Res 34:455–463 Kunzi-Rapp K, Genze F, Kufer R, Reich E, Hautmann RE, Gschwend JE, 2001, Chorioallantoic membrane assay: vascularized 3-dimensional cell culture system for human prostate cancer cells as an animal substitute model. J Urol 166:1502–1507 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 309 EP 313 DI 10.1007/s12253-010-9318-y PG 5 ER PT J AU Lakosi, F Antal, G Vandulek, Cs Kovacs, Toller, LG Rakasz, I Bajzik, G Hadjiev, J Bogner, P Repa, I AF Lakosi, Ferenc Antal, Gergely Vandulek, Csaba Kovacs, Arpad Toller, L Gabor Rakasz, Istvan Bajzik, Gabor Hadjiev, Janaki Bogner, Peter Repa, Imre TI Open MR-Guided High-Dose-Rate (HDR) Prostate Brachytherapy: Feasibility and Initial Experiences Open MR-Guided High-Dose-Rate (HDR) Prostate Brachytherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HDR; Brachytherapy; Image guidance; Open MRI; Prostate cancer ID HDR; Brachytherapy; Image guidance; Open MRI; Prostate cancer AB The aim of our pilot study was to demonstrate the feasibility and dosimetric quality of MR-guided HDR prostate brachytherapy in a low-field 0.35T open MRI scanner and to present our initial clinical experiences. 16 patiets with intermediate- to high-risk localized prostate cancer were treated with 46–60 Gy of external beam radiotherapy preceded and/or followed by an 8 Gy MR-guided HDR boost. For interventions an MR compatible custom-made system, coaxial needles and plastic catheters were used. Template reconstruction, trajectory planning, image guidance, contouring and treatment planning were exclusively based on MR images. For treatment planning, dose-point- and anatomy-based inverse planning optimization was used. Image quality was found to be good to excellent in almost all cases. The mean catheter placement accuracy modeled by Rayleigh distribution was 2.9 mm with a sigma value of 2.3 mm. The mean and standard deviation (SD) of the dosimetric results for the target volume were the following: V100: 94.2±4.3%, V150: 43.9±6.8%, V200: 18.5±5.9%. The mean D0.1, D1 and D1 values for the intraprostatic urethra were 117.6±12.5%, 98.5±19.9% and 122.3±16.4%, respectively. Regarding the rectal wall the mean D0.1, D1 and D2 values were 77.3±7.2%, 64.8±7.5%, and 53.2±9.1%, respectively. The mean maximum dose for the inner rectal surface was 53.5±9.2%. No RTOG Grade 3 or worse acute toxicities were observed. Our method seems to be a promising approach for performing feasible, accurate and high-quality MR-guided HDR prostate brachytherapy. To determine the long term side effects and outcome higher number of patients, additional follow-up is needed. C1 [Lakosi, Ferenc] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, 7400 Kaposvar, Hungary. [Antal, Gergely] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, 7400 Kaposvar, Hungary. [Vandulek, Csaba] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, 7400 Kaposvar, Hungary. [Kovacs, Arpad] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, 7400 Kaposvar, Hungary. [Toller, L Gabor] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, 7400 Kaposvar, Hungary. [Rakasz, Istvan] Kaposi Mor Teaching Hospital, Department of Urology, Tallian Gyula Street 20-32, 7400 Kaposvar, Hungary. [Bajzik, Gabor] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, 7400 Kaposvar, Hungary. [Hadjiev, Janaki] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, 7400 Kaposvar, Hungary. [Bogner, Peter] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, 7400 Kaposvar, Hungary. [Repa, Imre] Kaposi Mor Teaching Hospital, Guba Sandor Street 40, 7400 Kaposvar, Hungary. RP Kovacs, (reprint author), Kaposi Mor Teaching Hospital, 7400 Kaposvar, Hungary. EM kovacs.arpad@sic.hu CR Kasler M, Sz O, 2008, European and Hungarian national tasks in oncology. Magy Onkol 52:21–33 Jereczek-Fossa BA, Orecchia R, 2007, Evidence-based radiation oncology: definitive, adjuvant and salvage radiotherapy for nonmetastatic prostate cancer. Radiother Oncol 84:197–215 Morton G, 2005, The emerging role of high-dose-rate brachytherapy for prostate cancer. Clin Oncol 17:219–227 Hoskin PJ, Motohashi K, Bownes P et al, 2007, High dose rate brachytherapy in combination with external beam radiotherapy in the radical treatment of prostate cancer: initial results of a randomised phase three trial. Radiother Oncol 84:114–120 Guix B, Bartina JM, Henriquez I et al, 2007, Dose escalation by combined treatment 3-D conformal radiotherapy plus HDR brachytherapy as treatment for intermediate-or-high-risk cancer: early toxicity and biochemical outcome of a prospective randomized trial. [Abstract] Radiother Oncol 83(suppl1):82a Presti JC Jr, 2000, Prostate cancer: assessment of risk using digital rectal examination, tumor grade, prostate-specific antigen, and systematic biopsy. Radiol Clin North Am 38:49–58 Aigner F, Mitterberger M, Rehder P et al, 2010, Status of transrectal ultrasound imaging of the prostate. J Endourol 24:685–691 Futterer J, 2007, MR imaging in the local staging of prostate cancer. Eur J Radiol 63:328–334 Morgan VA, Kyriazi S, Ashley SE et al, 2007, Evaluation of the potential of diffusion-weighted imaging in prostate cancer detection. Acta Radiol 48:695–703 Kim Y, Hsu IC, Lessard E et al, 2008, Class solution in inverse planned HDR prostate brachytherapy for dose escalation of DIL defined by combined MRI/MRSI. Radiother Oncol 88:148–155 Van Lin EN, Futterer JJ, Heymink SW et al, 2006, IMRT boost dose planning on dominant intraprostatic lesions: gold markerbased three-dimensional fusion of CT with dynamic contrastenhanced and 1H-spectroscopic MRI. Int J Radiat Oncol Biol Phys 65:291–303 Tempany C, Straus S, Hata N et al, 2008, MR-guided prostate interventions. J Magn Reson Imaging 27:356–367 Cormack RA, Kooy H, Tempany CM et al, 2000, A clinical method for real-time dosimetric guidance of transperineal 125I prostate implants using interventional magnetic resonance imaging. Int J Radiat Oncol Biol Phys 46:207–214 Blumenfeld P, Hata N, DiMaio S et al, 2007, Transperineal prostate biopsy under magnetic resonance image guidance: a needle placement accuracy study. J Magn Reson Imaging 26:688–694 Ares C, Popowski Y, Pampallona S et al, 2009, Hypofractionated boost with high-dose-rate brachytherapy and open magnetic resonance imaging-guided implants for locally aggressive prostate cancer: a sequential dose-escalation pilot study. Int J Radiat Oncol Biol Phys 75:656–663 Menard C, Susil RC, Choyke P et al, 2004, MRI-guided HDR prostate brachytherapy in standard 1.5T scanner. Int J Radiat Oncol Biol Phys 59:1414–1423 Susil RC, Camphausen K, Choyke P et al, 2004, System for prostate brachytherapy and biopsy in a standard 1.5T MRI scanner. Magn Reson Med 52:683–687 Fischer GS, DiMaio SP, Iordachita II et al, 2007, Robotic assistant for transperineal prostate interventions in 3T closed MRI. Med Image Comput Comput Assist Interv Int Conf Med Image Comput Comput Assist Interv 10:425–433 Muntener M, Patriciu A, Petrisor D et al, 2008, Transperineal prostate intervention: robot for fully automated MR imaging–system description and proof of principle in a canine model. Radiology 247:543–549 Hadjiev J, Antal G, Antalffy Z et al, 2004, A novel technique with a flexible applicator for MRI-based brachytherapy of cervical cancer. Eur J Gynaecol Oncol 25:347–350 Lakosi F, Antal G, Vandulek C et al, 2009, Technical feasibility of transperineal MR-guided prostate interventions in a low-field open MRI unit: canine study. Pathol Oncol Res 15:315–322 National Comprehensive Cancer Network: National practice guidelines in oncology-prostate cancer, Version 2, 2008, monograph online). http://www.nccn.org/professionals/physician-gls/PDF/prostate.pdf Kovacs G, Potter R, Loch T, Hammer J et al, 2005, GEC/ESTROEAU recommendations on temporary brachytherapy using stepping sources for localised prostate cancer. Radiother Oncol 74:137–148 Baltas D, Kolotas C, Geramani K et al, 1998, A conformal index, COIN, to evaluate implant quality and dose specification in brachytherapy. Int J Radiat Oncol Biol Phys 40:515–524 Major T, Polgar C, Somogyi A et al, 2000, Evaluation of the dose uniformity for double-plane high dose rate interstitial breast implants with the use of dose reference points and dose nonuniformity ratio. Radiother Oncol 54:213–220 Wu A, Ulin K, Sternick ES, 1988, A dose homogeneity index for evaluating 192Ir interstitial breast implants. Med Phys 15:104– 107 Cox JD, Stetz J, Pajak TF, 1995, Toxicity criteria of the Radiation Therapy Oncology Group, RTOG, and the European Organization for Research and Treatment of Cancer, EORTC). Int J Radiat Oncol Biol Phys 31:1341–1346 Zangos S, Eichler K, Engelmann K et al, 2005, MR-guided transgluteal biopsies with an open low-field system in patients with clinically suspected prostate cancer: technique and preliminary results. Eur Radiol 15:174–182 Potter R, Kirisits C, Fidarova EF et al, 2008, Present status and future of high-precision image guided adaptive brachytherapy for cervix carcinoma. Acta Oncol 47:1325–1336 Citrin D, Ning H, Guion P et al, 2005, Inverse treatment planning based on MRI for HDR prostate brachytherapy. Int J Radiat Oncol Biol Phys 61:1267–1275 Morton GC, Sankreacha R, Halina P et al, 2008, A comparison of anatomy-based inverse planning with simulated annealing and graphical optimization for high-dose-rate prostate brachytherapy. Brachytherapy 7:12–16 Jacob D, Raben A, Sarkar A et al, 2008, Anatomy-based inverse planning simulated annealing optimization in high-dose-rate prostate brachytherapy: significant dosimetric advantage over other optimization techniques. Int J Radiat Oncol Biol Phys 72:820–827 Akimoto T, Katoh H, Kitamoto Y et al, 2006, Anatomy-based inverse optimization in high-dose-rate brachytherapy combined with hypofractionated external beam radiotherapy for localized prostate cancer: comparison of incidence of acute genitourinary toxicity between anatomy-based inverse optimization and geometric optimization. Int J Radiat Oncol Biol Phys 64:1360–1366 Frohlich G, Agoston P, Lovey J et al, 2007, Dosimetric evaluation of interstitial high-dose-rate implants for localised prostate cancer. Magy Onkol 51:31–38 Antal G, Lakosi F, Vandulek CS et al, 2009, MR modelling of the rectal dosimeter probe during MR-guided high-dose-rate, HDR, prostate brachytherapy: feasibility and initial experiences. [Abstract] Radiother Oncol 91(1):38 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 315 EP 324 DI 10.1007/s12253-010-9319-x PG 10 ER PT J AU Szarvas, T Jager, T Becker, M Tschirdewahn, S Niedworok, Ch Kovalszky, I Rubben, H Ergun, S vom Dorp, F AF Szarvas, Tibor Jager, Tobias Becker, Markus Tschirdewahn, Stephan Niedworok, Christian Kovalszky, Ilona Rubben, Herbert Ergun, Suleyman vom Dorp, Frank TI Validation of Circulating MMP-7 Level as an Independent Prognostic Marker of Poor Survival in Urinary Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; MMP-7; Matrilysin; Metastasis; Plasma; Prognosis; Serum ID Bladder cancer; MMP-7; Matrilysin; Metastasis; Plasma; Prognosis; Serum AB Molecular marker analyses aiming a more accurate disease characterization and risk stratification of cancer patients provided several promising marker candidates in the last few years. However, recent reviews underlined the paramount importance of validation, since many of the initially promising results could not be confirmed in independent patient cohorts. If serum or plasma is a more appropriate sample to test for prognostic markers is a matter of debate. We recently found serum MMP-7 levels to correlate with poor patients’ prognosis in urinary bladder cancer. In this study, we examined associations of the MMP-7 plasma levels with clinical follow-up data in an independent cohort of bladder cancer patients to validate our former results and to assess if plasma is also suitable for MMP-7 analysis. Plasma levels of 97 patients and 22 controls were analyzed, using enzyme-linked immunosorbent assay. Associations between MMP-7 plasma concentrations and clinical data were assessed applying both univariate and multivariate analysis. Plasma MMP-7 levels were significantly higher in patients than in controls. Similarly to our former findings in sera, high MMP-7 plasma levels proved to be significant and independent predictors of both overall and disease-specific survival. In addition, we observed a metastasis-specific difference in MMP-7 levels between serum and plasma. In summary, we confirmed the prognostic relevance of circulating MMP-7 levels in an independent cohort of patients and concluded that circulating MMP-7 levels may help to identify bladder cancer patients at high-risk of disease progression who could benefit from an adjuvant chemotherapy or from an extended lymph node dissection. C1 [Szarvas, Tibor] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Jager, Tobias] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Becker, Markus] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Tschirdewahn, Stephan] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Niedworok, Christian] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Rubben, Herbert] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Ergun, Suleyman] University Hospital of Essen, Institute of AnatomyEssen, Germany. [vom Dorp, Frank] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. RP vom Dorp, F (reprint author), University of Duisburg-Essen, Department of Urology, 45147 Essen, Germany. EM frankvomdorp@web.de CR Madersbacher S, Hochreiter W, Burkhard F, Thalmann GN, Danuser H, Markwalder R, Studer UE, 2003, Radical cystectomy for bladder cancer today—a homogeneous series without neoadjuvant therapy. J Clin Oncol 21:690–696 Hautmann RE, Gschwend JE, de Petriconi RC, Kron M, Volkmer BG, 2006, Cystectomy for transitional cell carcinoma of the bladder: results of a surgery only series in the neobladder era. J Urol 176:486–492 vom Dorp F, Borgermann C, Schenck M, Becker M, Rose A, Szarvas T, Rubben H, 2009, Role of lymphadenectomy in patients with invasive urothelial carcinoma of the bladder. Urologe 48:51– 53 Dyrskjot L, Zieger K, Real FX, Malats N, Carrato A, Hurst C, Kotwal S, Knowles M, Malmstrom PU, de la Torre M, Wester K, Allory Y, Vordos D, CaillaultA RF, Hein AM, Jensen JL, Jensen KM, Marcussen N, Orntoft TF, 2007, Gene expression signatures predict outcome in non-muscle-invasive bladder carcinoma: a multicenter validation study. Clin Cancer Res 13:3545–3551 Szarvas T, Becker M, vom Dorp F, Gethmann C, Totsch M, Bankfalvi A, Schmid KW, Romics I, Rubben H, Ergun S, 2010, Matrix metalloproteinase-7 as a marker of metastasis and predictor of poor survival in bladder cancer. Cancer Sci 101:1300–1308 Riley RD, Sauerbrei W, Altman DG, 2009, Prognostic markers in cancer: the evolution of evidence from single studies to metaanalysis, and beyond. Br J Cancer 100:1219–1229 Altman DG, Royston P, 2000, What do we mean by validating a prognostic model? Stat Med 19:453–473 Bleeker SE, Moll HA, Steyerberg EW, Donders AR, Derksen- Lubsen G, Grobbee DE, Moons KG, 2003, External validation is necessary in prediction research: a clinical example. J Clin Epidemiol 56:826–832 Werther K, Christensen IJ, Nielsen HJ, Danish RANX05 Colorectal Cancer Study Group, 2002, Prognostic impact of matched preoperative plasma and serum VEGF in patients with primary colorectal carcinoma. Br J Cancer 86:417–423 Eble JN, Epstein JI, Sesterhenn I, 2004, World Health Organization classification of tumors. Pathology and genetics of tumors of the urinary system and male genital organs. IARCC Press, Lyon, pp 89–158 Jung K, Klotzek S, Stephan C, Mannello F, Lein M, 2008, Impact of blood sampling on the circulating matrix metalloproteinases 1, 2, 3, 7, 8, and 9. Clin Chem 54:772–773 Powell WC, Fingleton B, Wilson CL, Boothby M, Matrisian LM, 1999, The metalloproteinase matrilysin proteolytically generates active soluble Fas ligand and potentiates epithelial cell apoptosis. Curr Biol 9:1441–1447 Noe V, Fingleton B, Jacobs K, Crawford HC, Vermeulen S, Steelant W, Bruyneel E, Matrisian LM, Mareel M, 2001, Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci 114:111–118 Ito TK, Ishii G, Saito S, Yano K, Hoshino A, Suzuki T, Ochiai A, 2009, Degradation of soluble VEGF receptor-1 by MMP-7 allows VEGF access to endothelial cells. Blood 113:2363–2369 Cornelius LA, Nehring LC, Harding E, Bolanowski M, Welgus HG, Kobayashi DK, Pierce RA, Shapiro SD, 1998, Matrix metalloproteinases generate angiostatin: effects on neovascularization. J Immunol 161:6845–6852 Wang WS, Chen PM, Wang HS, Liang WY, Su Y, 2006, Matrix metalloproteinase-7 increases resistance to Fas-mediated apoptosis and is a poor prognostic factor of patients with colorectal carcinoma. Carcinogenesis 27:1113–1120 Koskensalo S, Mrena J, Wiksten JP, Nordling S, Kokkola A, Hagstrom J, Haglund C, 2010, MMP-7 overexpression is an independent prognostic marker in gastric cancer. Tumour Biol 31:149–155 Yamamoto H, Itoh F, Iku S, Adachi Y, Fukushima H, Sasaki S, Mukaiya M, Hirata K, Imai K, 2001, Expression of matrix metalloproteinases and tissue inhibitors of metalloproteinases in human pancreatic adenocarcinomas: clinicopathologic and prognostic significance of matrilysin expression. J Clin Oncol 19:1118–1127 Pajouh MS, Nagle RB, Breathnach R, Finch JS, Brawer MK, Bowden GT, 1991, Expression of metalloproteinase genes in human prostate cancer. J Cancer Res Clin Oncol 117:144–150 Maurel J, Nadal C, Garcia-Albeniz X, Gallego R, Carcereny E, Almendro V, Marmol M, Gallardo E, Auge JM, Longaron R, Martinez-Fernandez A, Molina R, Castells A, Gascon P, 2007,, 2007, Serum matrix metalloproteinase 7 levels identifies poor prognosis advanced colorectal cancer patients. Int J Cancer 121:1066–1071 Martinez-Fernandez A, Garcia-Albeniz X, Pineda E, Visa L, Gallego R, Codony-Servat J, Auge JM, Longaron R, Gascon P, Lacy A, Castells A, Maurel J, 2009, Serum matrilysin levels predict outcome in curatively resected colorectal cancer patients. Ann Surg Oncol 16:1412–1420 Ramankulov A, Lein M, Johannsen M, Schrader M, Miller K, Jung K, 2008, Plasma matrix metalloproteinase-7 as a metastatic marker and survival predictor in patients with renal cell carcinomas. Cancer Sci 99:1188–1194 Szarvas T, Becker M, vom Dorp F, Meschede J, Scherag A, Bankfalvi A, Henning R, Schmid KW, Romics I, Rubben H, Ergun S, 2010, Elevated serum matrix metalloproteinase 7 levels predict poor prognosis after radical prostatectomy Int J Cancer, In press) Szarvas T, Singer BB, Becker M, vom Dorp F, Jager T, Szendroi A, Riesz P, Romics I, Rubben H, Ergun S, 2010, Urinary MMP-7 level is associated with the presence of metastasis in bladder cancer. BJU Int, In press) NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 325 EP 332 DI 10.1007/s12253-010-9320-4 PG 8 ER PT J AU Mao, YL Li, ZW Lou, ChJ Pang, D Zhang, YQ AF Mao, Yin-Ling Li, Zhi-Wei Lou, Chang-Jie Pang, Da Zhang, Yan-Qiao TI PHOSPHO-STAT5 Expression is Associated with Poor Prognosis of Human Colonic Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon carcinoma; Immunohistochemistry; Prognosis; Signal transducer and activator of transcription- 5 (STAT5) ID Colon carcinoma; Immunohistochemistry; Prognosis; Signal transducer and activator of transcription- 5 (STAT5) AB The signal transducer and activator of transcription-5 (STAT5) protein has been shown to play an important role in tumor progression through stimulating cell proliferation and preventing apoptosis. STAT5 activation has been observed in a variety of human tumors and cancer cell lines. However, it is not clear how activated STAT5 is expressed in colon cancer. In this study, we aimed to investigate phospho-STAT5 (activated form of STAT5) expression and its relationship with the clinicopathological factors and overall survival of patients with colonic adenocarcinoma. A total of 121 histological samples were selected for this study. Immunohistochemistry was used to detect the expression of phospho-STAT5. Analysis of the immunohistochemical staining was based on the proportion of stained cells in the field: positive, >15% stained cells, and negative, <15% stained cells. Survival times were analyzed using the Kaplan-Meier method, and the differences between groups were assessed with the log-rank test. A multivariate Cox regression model was used for prognostic power analysis. Expression of phospho-STAT5 was observed in the cytoplasms of colonic adenocarcinoma cells. Univariate analysis showed that phospho-STAT5 immunoreactivity was correlated with the depth of tumor invasion (P-value=0.009), tumornode-metastasis (TNM) stage (P-value=0.048) and shorter overall survival times (P-value=0.026). Lymph node metastasis, distant metastasis and TNM stage were associated with shorter overall survival times (P-value range from 0.003-<0.001). Multivariate analysis showed that only distant metastasis was an independent predictor of overall survival time (P-value=0.016). Our findings first demonstrate that phospho-STAT5 is frequently present and active in colonic adenocarcinoma and related to poor prognosis. C1 [Mao, Yin-Ling] Harbin Medical University, Cancer Hospital, Department of Medical Oncology, No. 150, Haping Road, 150040 Harbin, Heilongjiang Province, China. [Li, Zhi-Wei] Harbin Medical University, Cancer Hospital, Department of Medical Oncology, No. 150, Haping Road, 150040 Harbin, Heilongjiang Province, China. [Lou, Chang-Jie] Harbin Medical University, Cancer Hospital, Department of Medical Oncology, No. 150, Haping Road, 150040 Harbin, Heilongjiang Province, China. [Pang, Da] The Affiliated Tumor Hospital of Harbin Medical University, Department of Surgery, 150040 Harbin, Heilongjiang Province, China. [Zhang, Yan-Qiao] Harbin Medical University, Cancer Hospital, Department of Medical Oncology, No. 150, Haping Road, 150040 Harbin, Heilongjiang Province, China. RP Zhang, YQ (reprint author), Harbin Medical University, Cancer Hospital, Department of Medical Oncology, 150040 Harbin, China. EM yanqiaozhang@126.com CR Lievre A, Bachet JB, Le Corre D et al, 2006, KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 66:3992–3995 Galizia G, Lieto E, Ferraraccio F et al, 2004, Determination of molecular marker expression can predict clinical outcome in colon carcinomas. Clin Cancer Res 10:3490–3499 Muto T, Kotake K, Koyama Y, 2001, Colorectal cancer statistics in Japan: data from JSCCR registration, 1974–1993. Int J Clin Oncol 6:171–176 Newland RC, Dent OF, Lyttle MN et al, 1994, Pathologic determinants of survival associated with colorectal cancer with lymph node metastases. A multivariate analysis of 579 patients. Cancer 73:2076–2082 Guba M, Seeliger H, Kleespies A et al, 2004, Vascular endothelial Kusaba T, Nakayama T, Yamazumi K et al, 2006, Activation of STAT3 is a marker of poor prognosis in human colorectal cancer. Oncol Rep 15:1445–1451 Buettner R, Mora LB, Jove R, 2002, Activated STAT signaling in human tumors provides novel molecular targets for therapeutic intervention. Clin Cancer Res 8:945–954 Calo V, Migliavacca M, Bazan Vet al, 2003, STAT proteins: from normal control of cellular events to tumorigenesis. J Cell Physiol 197:157–168 Darnell JE Jr, 1997, STATs and gene regulation. Science 277:1630–1635 Ramana CV, Chatterjee-Kishore M, Nguyen H et al, 2000, Complex roles of Stat1 in regulating gene expression. Oncogene 19:2619–2627 Corvinus FM, Orth C, Moriggl R et al, 2005, Persistent STAT3 activation in colon cancer is associated with enhanced cell proliferation and tumor growth. Neoplasia 7:545–555 GongW,Wang L, Yao JC et al, 2005, Expression of activated signal transducer and activator of transcription 3 predicts expression of vascular endothelial growth factor in and angiogenic phenotype of human gastric cancer. Clin Cancer Res 11:1386–1393 Wei D, Le X, Zheng L et al, 2003, Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis. Oncogene 22:319–329 Coffer PJ, Koenderman L, de Groot RP, 2000, The role of STATs in myeloid differentiation and leukemia. Oncogene 19:2511–2522 Li H, Zhang Y, Glass A et al, 2005, Activation of signal transducer and activator of transcription-5 in prostate cancer predicts early recurrence. Clin Cancer Res 11:5863–5868 Li H, Ahonen TJ, Alanen K et al, 2004, Activation of signal transducer and activator of transcription 5 in human prostate cancer is associated with high histological grade. Cancer Res 64:4774–4782 Nevalainen MT, Xie J, Torhorst J et al, 2004, Signal transducer and activator of transcription-5 activation and breast cancer prognosis. J Clin Oncol 22:2053–2060 Lee TK, Man K, Poon RT et al, 2006, Signal transducers and activators of transcription 5b activation enhances hepatocellular carcinoma aggressiveness through induction of epithelialmesenchymal transition. Cancer Res 66:9948–9956 Xiong H, Su WY, Liang QC et al, 2009, Inhibition of STAT5 induces G1 cell cycle arrest and reduces tumor cell invasion in human colorectal cancer cells. Lab Invest 89:717–725 O’Connell JB, Maggard MA, Ko CY, 2004, Colon cancer survival rates with the new American Joint Committee on Cancer sixth edition staging. J Natl Cancer Inst 96:1420–1425 Jass JR, Sobin LH, Watanabe H, 1990, The World Health Organization’s histologic classification of gastrointestinal tumors. A commentary on the second edition. Cancer 66:2162–2167 Hsiao JR, Jin YT, Tsai ST et al, 2003, Constitutive activation of STAT3 and STAT5 is present in the majority of nasopharyngeal carcinoma and correlates with better prognosis. Br J Cancer 89:344–349 Ahonen TJ, Xie J, LeBaron MJ et al, 2003, Inhibition of transcription factor Stat5 induces cell death of human prostate cancer cells. J Biol Chem 278:27287–27292 Ren S, Cai HR, Li M et al, 2002, Loss of Stat5a delays mammary cancer progression in a mouse model. Oncogene 21:4335–4339 Cotarla I, Ren S, Zhang Y et al, 2004, Stat5a is tyrosine phosphorylated and nuclear localized in a high proportion of human breast cancers. Int J Cancer 108:665–671 Harir N, Boudot C, Friedbichler K et al, 2008, Oncogenic Kit controls neoplastic mast cell growth through a Stat5/PI3-kinase signaling cascade. Blood 112:2463–2473 Harir N, Pecquet C, Kerenyi M et al, 2007, Constitutive activation of Stat5 promotes its cytoplasmic localization and association with PI3-kinase in myeloid leukemias. Blood 109: 1678–1686 Klampfer L, 2008, The role of signal transducers and activators of transcription in colon cancer. Front Biosci 13:2888–2899 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 333 EP 339 DI 10.1007/s12253-010-9321-3 PG 7 ER PT J AU Ru, GQ Wang, HJ Xu, WJ Zhao, ZSh AF Ru, Guo-Qing Wang, Hui-Ju Xu, Wen-Jun Zhao, Zhong-Sheng TI Upregulation of Twist in Gastric Carcinoma Associated with Tumor Invasion and Poor Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric carcinoma; Metastasis; RT-PCR; Twist expression ID Gastric carcinoma; Metastasis; RT-PCR; Twist expression AB Tumor invasion and metastasis are the most common causes of death in gastric carcinoma. Twist, a transcription factor of the basic helix-loop-helix class, reportedly regulates cancer metastasis and induces epithelial-mesenchymal transition (EMT). We evaluated the expression of Twist and its effect on cell migration in gastric carcinoma (GC). Twist expression was detected by real-time quantitative RT-PCR in gastric tumor tissue, metastatic lymph nodes and normal gastric tissue from 47 gastric carcinoma patients who had undergone gastrectomies with radical lymph node dissections without preoperative treatment. Twist expression was also analyzed immunohistochemically in 436 gastric cancer cases. GC tumor tissue and metastatic lymph nodes was upregulated compared with normal gastric mucosa (P<0.05). Twist protein expression differed significantly among gastric tumor tissue, matched normal gastric mucosa, and lymph nodes (P<0.05) In stages I, II, and III, 5-year survival rates of patients with high Twist expression were significantly lower than in patients with low expression (P<0.05). In stage IV, Twist expression did not correlate with 5-year survival rates (P=0.07). Further multivariate analysis suggested that depth of invasion, lymph-node and distant metastases, TNM stage, and upregulation of TWIST were independent prognostic indicators for GC. Twist expression in gastric cancer is associated significantly with lymph-node and distant metastases, and poor prognosis. Twist may be a useful marker for the development, progression and metastasis of GC. C1 [Ru, Guo-Qing] Wenzhou Medical University, 325035 Wenzhou, Zhejiang, China. [Wang, Hui-Ju] Key Laboratory of Gastroenterology of Zhejiang Province, 310014 Hangzhou, China. [Xu, Wen-Jun] Zhejiang Provincial People’s Hospital, Department of Pathology, 158 Shangtang Road, 310014 Hangzhou, Zhejiang, China. [Zhao, Zhong-Sheng] Zhejiang Provincial People’s Hospital, Department of Pathology, 158 Shangtang Road, 310014 Hangzhou, Zhejiang, China. RP Zhao, ZSh (reprint author), Zhejiang Provincial People’s Hospital, Department of Pathology, 310014 Hangzhou, China. EM zhaozhongsheng50@126.com CR Zhan WH, Han FH, 2008, Surgical therapy of gastric cancer in china. J Pract Oncol 23:91–93 Eccles SA, Welch DR, 2007, Metastasis: recent discoveries and novel treatment strategies. Lancet 369:1742–1757 Gilles C, Thompson EW, 1996, The epithelial-to-mesenchymal transition and metastatic progression in carcinoma. Breast J 2:83– 96 Hay ED, 1995, An overview of epithelio-mesenchymal transformation. Acta Anat 154:8–20 Monk M,Holding C, 2001)Human embryonic genes re-expressed in cancer cells. Oncogene 20:8085–8091 Bates RC, Mercurio AM, 2005, The epithelial-mesenchymal transition, EMT, and colorectal cancer progression. Cancer Biol Ther 4:365–370 Xue C, Plieth D, Venkov C et al, 2003, The gatekeeper effect of epithelial-mesenchymal transition regulates the frequency of breast cancer metastasis. Cancer Res 63:3386–3394 Thiery JP, Sleeman JP, 2006, Complex networks orchestrate epithelia-mesenchymal transitions. Nat Rev Mol Cell Biol 7:131– 142 Thiery JP, 2003, Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol 15:740–746 Thiery JP, 2002, Epithelial-mesenchymal transitions in tumour progression. Nature Rev 2:442–454 Yook JI, Li XY, Ota I et al, 2005, Wnt-dependent regulation of the E-cadherin repressor snail. J Biol Chem 280:11740–11748 Zhou BP, Deng J, Xia W et al, 2004, Dual regulation of Snail by GSK-3beta-mediated phosphorylation in control of epithelialmesenchymal transition. Nature Cell Biol 6:931–940 Cheng GZ, Zhang W, Wang LH, 2008, Regulation of cancer cell survival, migration, and invasion by Twist: AKT2 comes to interplay. Cancer Res 68:957–960 Yang J, Mani SA, Donaher JL et al, 2004, Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117:927–939 Yang J, Mani SA, Weinberg RA, 2006, Exploring a new twist on tumor metastasis. Cancer Res 66:4549–4552 Lee TK, Poon RT, Yuen AP et al, 2006, Twist overexpression correlates with hepatocellular carcinoma metastasis through induction of epithelial-mesenchymal transition. Clin Cancer Res 12:5369–5376 Yang Z, Zhang X, Gang H et al, 2007, Up-regulation of gastric cancer cell invasion by Twist is accompanied by N-cadherin and fibronectin expression. Biochem Biophys Res Comm 358:925– 930 Lee HS, Lee HK, Kim HS et al, 2003, Tumour suppressor gene expression correlates with gastric cancer prognosis. J Pathol 200:39–46 Lee HS, Lee HK, Kim HS et al, 2001, MUC1, MUC2, MUC5AC, and MUC6 expressions in gastric carcinomas: their roles as prognostic indicators. Cancer 92:1427–1434 Zhang D, Salto-Tellez M, Putti TC et al, 2003, Reliability of tissue microarrays in detecting protein expression and gene amplification in breast cancer. Mod Pathol 16:79–84 Lee HS, Cho SB, Lee HE et al, 2007, Protein expression profiling and molecular classification of gastric cancer by the tissue array method. Clin Cancer Res 13:4154–4163 Kolev Y, Uetake H, Iida S et al, 2007, Prognostic significance of VEGF expression in correlation with COX-2, microvessel density and clinicopathological characteristics in human gastric carcinoma. Ann Surg Oncology 14:2738–2747 Mizokami K, Kakeji Y, Oda S et al, 2006, Clinicopathologic significance of hypoxia-inducible factor 1 alpha overexpression in gastric carcinomas. J Surg Oncology 94:149–154 Song LB, Liao WT, Mai HQ et al, 2006, The clinical significance of twist expression in nasopharyngeal carcinoma. Cancer Lett 242:258–265 Kolev Y, Uetake H, Iida S et al, 2007, Prognostic significance of VEGF expression in correlation with COX-2, microvessel density, and clinicopathological characteristics in human gastric carcinoma. Ann Surg Oncol 14:2738–2747 Chou YY, Jeng YM, Lee TT et al, 2007, Cytoplasmic CD24 expression is a novel prognostic factor in diffuse-type gastric adenocarcinoma. Ann Surg Oncol 14:2748–2758 Chen ZF, Behringer RR, 1995, Twist is required in head mesenchyme for cranial neural tube morphogenesis. Genes Dev 9:686–699 Leptin M, 1991, Twist and snail as positive and negative regulators during Drosophila mesoderm development. Genes Dev 5:1568–1576 Maestro R, Dei Tos AP, Hamamori Y et al, 1999, Twist is a potential oncogene that inhibits apoptosis. Genes Dev 13:2207– 2217 Valsesia-Wittmann S, Magdeleine M, Dupasquier S et al, 2004, Oncogenic cooperation between H-Twist and N-Myc overrides failsafe programs in cancer cells. Cancer Cell 6:625–630 Yang J, Mani SA, Donaher JL et al, 2004, Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117:927–939 Rosivatz E, Becker I, Specht K et al, 2002, Differential expression of the epithelial-mesenchymal transition regulators snail, SIP1, and twist in gastric cancer. Am J Pathol 161:1881– 1891 Martin TA, Goyal A, Watkins G et al, 2005, Expression of the transcription factors snail, slug, and Twist and their clinical significance in human breast cancer. Ann Surg Oncol 12:488–496 Hoek K, Rimm DL, Williams KR et al, 2004, Expression profiling reveals novel pathways in the transformation of melanocytes to melanomas. Cancer Res 64:5270–5282 Watanabe O, Imamura H, Shimizu T et al, 2004, Expression of twist and wnt in human breast cancer. Anticancer Res 24:3851– 3856 Zi X, Guo Y, Simoneau AR et al, 2005, Expression of Frzb/ secreted Frizzled-related protein 3, a secreted Wnt antagonist, in human androgen-independent prostate cancer PC-3 cells suppresses tumor growth and cellular invasiveness. Cancer Res 65:9762–9770 Guo Y, Zi X, Koontz Z et al, 2007, Blocking Wnt/LRP5 signaling by a soluble receptor modulates the epithelial to mesenchymal transition and suppressesmet andmetalloproteinases in osteosarcoma Saos-2 cells. J Orthop Res 25:964–971 Bienz M, Clevers H, 2000, Linking colorectal cancer to Wnt signaling. Cell 103:311–320 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 341 EP 347 DI 10.1007/s12253-010-9332-0 PG 7 ER PT J AU Goncalves, RA Carneiro, JVA Martins, I de Faria, ASP Ferreira, AM de Mello, LRE Fogaca, SH Elia, CSC de Souza, SPH AF Goncalves, Rodrigues Andrea Carneiro, Jose Vasconcellos Antonio Martins, Ivanir de Faria, Antonio Silvestre Paulo Ferreira, Aparecida Maria de Mello, Linhares Riello Eduardo Fogaca, Soares Homero Elia, Carvalho Siqueira Celeste de Souza, Siffert Pereira Heitor TI Prognostic Significance of p53 Protein Expression in Early Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Early gastric cancer; p53; Prognosis ID Early gastric cancer; p53; Prognosis AB Mutations of the p53 tumor suppressor gene have been associated with abnormalities in cell cycle regulation, DNA repair and synthesis, apoptosis, and it has been implicated in the prognosis of advanced gastric cancer. The aim of this study was to evaluate the occurrence of p53 gene mutation and its possible prognostic implications in early gastric cancer. In a retrospective study, we studied 80 patients with early gastric cancer treated surgically between 1982 and 2001. Mutation of p53 gene was investigated in surgical gastric specimens by immunohistochemistry, and results were analyzed in relation to gender, age, macroscopic appearance, size and location of tumor, presence of lymph nodes, Lauren’s histological type, degree of differentiation, and the 5-year survival. The expression of p53 was more frequent among the intestinal type (p = 0.003), the differentiated (p = 0.007), and the macroscopically elevated tumors (p = 0.038). Nevertheless, the isolated expression of p53 was not associated with the 5-year survival, or with the frequency of lymph node involvement. The degree of differentiation was detected as an independent factor related to the outcome of patients (0.044). Significantly shorter survival time was found in p53-negative compared with p53-positive patients, when considering the degree of differentiation of tumors, as assessed by Cox regression analysis (0.049). The association of p53 with the intestinal type, the degree of differentiation and morphological characteristics, may reflect the involvement of chronic inflammatory process underlying early gastric cancer. In this population sample, the expression of p53 alone has no prognostic value for early gastric cancer. However, the significant difference in p53 expression between subgroups of degree of differentiation of tumors can influence post-operative outcome of patients and may be related to possible distinct etiopathogenic subtypes. C1 [Goncalves, Rodrigues Andrea] Universidade Federal do Rio de Janeiro (UFRJ), Hospital Universitario Clementino Fraga Filho, Departamento de Clinica Medica, Rodolpho Paulo Rocco 255, Ilha do Fundao, 21941-913 Rio de Janeiro, RJ, Brazil. [Carneiro, Jose Vasconcellos Antonio] Universidade Federal do Rio de Janeiro (UFRJ), Hospital Universitario Clementino Fraga Filho, Departamento de Clinica Medica, Rodolpho Paulo Rocco 255, Ilha do Fundao, 21941-913 Rio de Janeiro, RJ, Brazil. [Martins, Ivanir] Instituto Nacional de Cancer, Divisao de Patologia, 20230-130 Rio de Janeiro, Brazil. [de Faria, Antonio Silvestre Paulo] Instituto Nacional de Cancer, Divisao de Patologia, 20230-130 Rio de Janeiro, Brazil. [Ferreira, Aparecida Maria] Instituto Nacional do Cancer, Secao de Endoscopia Digestiva, 20230-130 Rio de Janeiro, Brazil. [de Mello, Linhares Riello Eduardo] Instituto Nacional do Cancer, e Secao de Cirurgia Oncologica Abdomino-pelvica, 20230-130 Rio de Janeiro, Brazil. [Fogaca, Soares Homero] Universidade Federal do Rio de Janeiro (UFRJ), Hospital Universitario Clementino Fraga Filho, Departamento de Clinica Medica, Rodolpho Paulo Rocco 255, Ilha do Fundao, 21941-913 Rio de Janeiro, RJ, Brazil. [Elia, Carvalho Siqueira Celeste] Universidade Federal do Rio de Janeiro (UFRJ), Hospital Universitario Clementino Fraga Filho, Departamento de Clinica Medica, Rodolpho Paulo Rocco 255, Ilha do Fundao, 21941-913 Rio de Janeiro, RJ, Brazil. [de Souza, Siffert Pereira Heitor] Universidade Federal do Rio de Janeiro (UFRJ), Hospital Universitario Clementino Fraga Filho, Departamento de Clinica Medica, Rodolpho Paulo Rocco 255, Ilha do Fundao, 21941-913 Rio de Janeiro, RJ, Brazil. RP de Souza, SPH (reprint author), Universidade Federal do Rio de Janeiro (UFRJ), Hospital Universitario Clementino Fraga Filho, Departamento de Clinica Medica, 21941-913 Rio de Janeiro, Brazil. EM hsouza@hucff.ufrj.br;heitor.souza@gmail.com CR Botterweck AA, Schouten LJ, Volovics A, Dorant E, van Den Brandt PA, 2000, Trends in incidence of adenocarcinoma of the oesophagus and gastric cardia in ten European countries. Int J Epidemiol 29:645–654 Boussioutas A, Taupin D, 2001, Towards a molecular approach to gastric cancer management. Intern Med J 31:296–303 Shah MA, 2006, Gastric cancer: an update. Curr Oncol Rep 8:183–191 Field K, Michael M, Leong T, 2008, Locally advanced and metastatic gastric cancer: current management and new treatment developments. Drugs 68:299–317 Kaibara N, Sumi K, Yonekawa M, Ohta M, Makino M, Kimura O, Nishidoi H, Koga S, 1990, Does extensive dissection of lymph nodes improve the results of surgical treatment of gastric cancer? Am J Surg 159:218–221 Maehara Y, Kabashima A, Koga T, Tokunaga E, Takeuchi H, Kakeji Y, Sugimachi K, 2000, Vascular invasion and potential for tumor angiogenesis and metastasis in gastric carcinoma. Surgery 128:408–416 Forman D, Burley VJ, 2006, Gastric cancer: global pattern of the disease and an overview of environmental risk factors. Best Pract Res Clin Gastroenterol 20:633–649 Anderson C, Nijagal A, Kim J, 2006, Molecular markers for gastric adenocarcinoma: an update. Mol Diagn Ther 10:345–352 Smith MG, Hold GL, Tahara E, El-Omar EM, 2006, Cellular and molecular aspects of gastric cancer. World J Gastroenterol 12:2979–2990 Prives C, Hall PA, 1999, The p53 pathway. J Pathol 187:112–126 Greenblatt MS, Bennett WP, Hollstein M, Harris CC, 1994, Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res 54:4855–4878 Hainaut P, Hernandez T, Robinson A, Rodriguez-Tome P, Flores T, Hollstein M et al, 1998, IARC database of p53 gene mutations in human tumors and cell lines: updated compilation, revised formats and new visualization tools. Nucleic Acids Res 26:205–213 Kastan MB, Onyekwere O, Sidransky D, Vogelstein B, Craig RW, 1991, Participation of p53 protein in the cellular response to DNA damage. Cancer Res 51:6304–6311 Hollstein M, Sidransky D, Vogelstein B, Harris C, 1991, p53 mutations in human cancers. Science 253:49–53 Lane DP, 1994, The regulation of p53 function: Steiner award lecture. Int J Cancer 57:623–627 Starzynska T, Bromley M, Ghosh A, Stern PL, 1992, Prognostic significance of p53 overexpression in gastric and colorectal carcinoma. Br J Cancer 66:558–562 Kakeji Y, Korenaga D, Tsujitani S, Baba H, Anai H, Maehara Y, Sugimachi K, 1993, Gastric cancer with p53 overexpression has high potential for metastasising to lymph nodes. Br J Cancer 67:589–593 Joypaul BV, Hopwood D, Newman EL, Qureshi S, Grant A, Ogston SA, Lane DP, Cuschieri A, 1994, The prognostic significance of the accumulation of p53 tumour-suppressor gene protein in gastric adenocarcinoma. Br J Cancer 69:943–946 Motojima K, Furui J, Kohara N, Ito T, Kanematsu T, 1994, Expression of p53 protein in gastric carcinomas is not independently prognostic. Surgery 116:890–895 Gabbert HE, Muller W, Schneiders A, Meier S, Hommel G, 1995, The relationship of p53 expression to the prognosis of 418 patients with gastric carcinoma. Cancer 76:720–726 Bataille F, Rummele P, Dietmaier W, Gaag D, Klebl F, Reichle A, Wild P, Hofstadter F, Hartmann A, 2003, Alterations in p53 predict response to preoperative high dose chemotherapy in patients with gastric cancer. Mol Pathol 56:286–92 Xiangming C, Hokita S, Natsugoe S, Tanabe G, Baba M, Takao S, Kuroshima K, Aikou T, 1999, Cooccurrence of reduced expression of alpha-catenin and overexpression of p53 is a predictor of lymph node metastasis in early gastric cancer. Oncology 57:131–137 Lauren P, 1965, The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand 64:31–49 Montesano R, Hollstein M, Hainaut P, 1996, Genetic alterations in esophageal cancer and their relevance to etiology and pathogenesis: a review. Int J Cancer 69:225–235 Soussi T, Beroud C, 2001, Assessing TP53 status in human tumours to evaluate clinical outcome. Nat Rev Cancer 1:233–240 Carson DA, Lois A, 1995, Cancer progression and p53. Lancet 346:1009–1011 Chang F, Syrjanen S, Syrjanen K, 1995, Implications of the p53 tumorsuppressor gene in clinical oncology. J Clin Oncol 13:1009– 1022 Yokozaki H, Kuniyasu H, Kitadai Y, Nishimura K, Todo H, Ayhan A, YasuiW, Ito H, Tahara E, 1992, p53 point mutations in primary human gastric carcinomas. J Cancer Res Clin Oncol 119:67–70 Uchino S, Noguchi M, Ochiai A, Saito T, Kobayashi M, Hirohashi S, 1993, p53 mutation in gastric cancer: a genetic model for carcinogenesis is common to gastric and colorectal cancer. Int J Cancer 54:759–764 Agarwal ML, Taylor WR, Chernov MV, Chernova OB, Stark GR, 1998, The p53 network. J Biol Chem 273:1–4 Hall PA, Lane DP, 1994, p53 in tumor pathology: can we trust immunohistochemistry?-Revisited! J Pathol 72:1–4 Gannon JV, Greaves R, Iggo R, Lane DP, 1990, Activating mutations in p53 produce a common conformational effect. A monoclonal antibody specific for the mutant form. EMBO J 9:1595–1602 Levine AJ, Momand J, Finlay CA, 1991, The p53 tumour suppressor gene. Nature 351:453–456 Maehara Y, Tomoda M, Hasuda S, Kabashima A, Tokunaga E, Kakeji Y, Sugimachi K, 1999, Prognostic value of p53 protein expression for patients with gastric cancer—a multivariate analysis. Br J Cancer 79:1255–1261 Ming SC, 1998, Cellular and molecular pathology of gastric carcinoma and precursor lesions: A critical review. Gastric Cancer 1:31–50 Wang JY, Lin SR, Hsieh JS, Hsu CH, Huang YS, Huang TJ, 2001, Mutations of p53 gene in gastric carcinoma in Taiwan. Anticancer Res 21:513–520 Tahara E, Semba S, Tahara H, 1996, Molecular biological observations in gastric cancer. Semin Oncol 23:307–315 Wu MS, Shun CT, Lee WC, Chen CJ, Wang HP, Lee WJ, Sheu JC, Lin JT, 1998, Overexpression of p53 in different subtypes of intestinal metaplasia and gastric cancer. Br J Cancer 78:971–973 Wu MS, Shun CT, Wu CC, Hsu TY, Lin MT, Chang MC, Wang HP, Lin JT, 2000, Epstein-Barr virus-associated gastric carcinomas: relation to H. pylori infection and genetic alterations. Gastroenterology 118:1031–1038 Correa P, 1992, Human gastric carcinogenesis: a multistep and multifactorial process–First American Cancer Society Award Lecture on Cancer Epidemiology and Prevention. Cancer Res 52:6735–6740 Kodama M, Fujioka T, Murakami K, Okimoto T, Sato R, Watanabe K, Nasu M, 2005, Eradication of Helicobacter pylori reduced the immunohistochemical detection of p53 and MDM2 in gastric mucosa. J Gastroenterol Hepatol 20:941–946 Morgan C, Jenkins GJS, Ashton T, Griffiths AP, Baxter JN, Parry EM, Parry JM, 2003, Detection of p53 mutations in precancerous gastric tissue. Br J Cancer 89:1314–1319 Kodama M, Murakami K, Okimoto T, Sato R, Watanabe K, Fujioka T, 2007, Expression of mutant type-p53 products in H pylori-associated chronic gastritis. World J Gastroenterol 13:1541–1546 Li JH, Shi XZ, Lv S, Liu M, Xu GW, 2005, Effect of helicobacter pylori infection on p53 expression of gastric mucosa and adenocarcinoma with microsatellite instability. World J Gastroenterol 11:4363–4366 Maehara Y, Kakeji Y, Oda S, Baba H, Sugimachi K, 2001, Tumor growth patterns and biological characteristics of early gastric carcinoma. Oncology 61:102–112 Noda H, Maehara Y, Irie K, Kakeji Y, Yonemura T, Sugimachi K, 2001, Growth pattern and expressions of cell cycle regulator proteins p53 and p21 WAF1/CIP1 in early gastric carcinoma. Cancer 92:1828–1835 McLaren R, Kuzu I, Dunnill M, Harris A, Lane D, Gatter KC, 1992, The relationship of p53 immunostaining to survival in carcinoma of the lung. Br J Cancer 66:735–738 Hamilton JP, Meltzer SJ, 2006, A Review of the Genomics of Gastric Cancer. Clin Gastroenterol Hepatol 4:416–425 Yamashita H, Nishio M, Toyama T, Sugiura H, Zhang Z, Kobayashi S, Iwase H, 2004, Coexistence of HER2 overexpression and p53 protein accumulation is a strong prognostic molecular marker in breast cancer. Breast Cancer Res 6:R24–30 Lu C-D, Altieri DC, Tanigawa N, 1998, Expression of A Novel Antiapoptosis Gene, Survivin, Correlated with Tumor Cell Apoptosis and p53 Accumulation in Gastric Carcinomas. Cancer Res 58:1808–1812 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 349 EP 355 DI 10.1007/s12253-010-9333-z PG 7 ER PT J AU Cai, W Sun, Y Wang, W Han, Ch Ouchida, M Xia, W Zhao, X Sun, B AF Cai, Wenjuan Sun, Yan Wang, Wei Han, Chunrong Ouchida, Mamoru Xia, Wenbin Zhao, Xiulan Sun, Baocun TI The Effect of SYT-SSX and Extracellular Signal-Regulated Kinase (ERK) on Cell Proliferation in Synovial Sarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Synovial sarcoma; Cell proliferation; Fusion gene ID Synovial sarcoma; Cell proliferation; Fusion gene AB The character of Synovial sarcoma is the chromosomal translocation t(X; 18)(p11.2;q11.2), which results in the fusion of the SYT gene with a SSX gene. There is little study that could fully elucidate the mechanism of pathogenesis of this fusion transcript. This study is designed to gain more insight into the function of this fusion gene. We evaluated the whole genome expression in SYO-1 cells inhibited as a result of specific small interfering RNA for SYT-SSX. Cell proliferation and apoptosis were analyzed by flow cytometer and MTT. The proteins correlated with proliferation were also detected using western blot. TUNEL and Immunohistochemical stain assessment were also carried out on TMA of SS tissues. The mRNA level reduced over 90% caused by SYT-SSX specific siRNA. Five pathways were employed, that ERK1/2 pathway was differential significantly (p=0.043218). Meanwhile, down-regulation of SYT-SSX fusion gene expression would inhibit the proliferation of SS cell and the survival rate decreased (34.1%), while apoptotic rate increased (10.92%). After transfected with SYT-SSX-specific siRNA it caused a block in G1/G0 phase (31.99%) of SYO-1 cells compared with control cells. The protein level of ERK1/2, p-ERK, and cyclin D1 altered in same trend with expression of SYT-SSX. In TMA stain assessment, SYT-SSX positive group with high ki-67 LI expressed more cyclin D1and CDK4 than the SYT-SSX negative group. High ki-67 LI was detected in cases with p-ERK expression. Meanwhile, cyclin D1 and CDK4 were shown to be more expressed in tumor cells with p-ERK expression. Our results suggest that the fusion gene SYTSSX should be considered to play important role on SS cell growth via ERK pathway. This study may be valuable for understanding the pathogenic role and molecular mechanism of the fusion gene SYT-SSX in synovial sarcoma through the proposed genome-wide approach. Furthermore, the research would open up the possibility of using SYT-SSX and ERK as a therapeutic target. C1 [Cai, Wenjuan] Tianjin Medical University, Department of PathologyTianjin, China. [Sun, Yan] Tianjin Medical University, Department of PathologyTianjin, China. [Wang, Wei] Tianjin Medical University, Department of PathologyTianjin, China. [Han, Chunrong] Tianjin Medical University, Department of PathologyTianjin, China. [Ouchida, Mamoru] Okayama University, Graduate School of Medicine and Dentistry, Department of Molecular GeneticsOkayama, Japan. [Xia, Wenbin] Tianjin Medical University, Department of PathologyTianjin, China. [Zhao, Xiulan] Tianjin Medical University, Department of Pathology, 300060 Tianjin, China. [Sun, Baocun] Tianjin Medical University, Department of PathologyTianjin, China. RP Sun, B (reprint author), Tianjin Medical University, Department of Pathology, Tianjin, China. EM baocunsun@gmail.com CR Lewis JJ, Antonescu CR, Leung DH et al, 2000, Synovial sarcoma: a multivariate analysis of prognostic factors in 112 patients with primary localized tumors of the extremity. J Clin Oncol 18(10):2087–2094 Clark J, Rocques PJ, Crew AJ et al, 1994, Identification of novel genes, SYT and SSX, involved in the t(X;18)(p11.2;q11.2, translocation found in human synovial sarcoma. Nat Genet 7, 4):502–508 dos Santos NR, de Bruijn DR, van Kessel AG, 2001, Molecular mechanisms underlying human synovial sarcoma development. Genes Chromosom Cancer 30(1):1–14 Ladanyi M, 2001, Fusions of the SYT and SSX genes in synovial sarcoma. Oncogene 20(40):5755–5762 Nagai M, Tanaka S, Tsuda M et al, 2001, Analysis of transforming activity of human synovial sarcoma-associated chimeric protein SYT-SSX1 bound to chromatin remodeling factor hBRM/hSNF2 alpha. Proc Natl Acad Sci USA 98, 7):3843–3848 Nilsson G, Skytting B, Xie Y et al, 1999, The SYT-SSX1 variant of synovial sarcoma is associated with a high rate of tumor cell proliferation and poor clinical outcome. Cancer Res 59(13):3180– 3184 Inagaki H, Nagasaka T, Otsuka T, Sugiura E, Nakashima N, Eimoto T, 2000, Association of SYT-SSX fusion types with proliferative activity and prognosis in synovial sarcoma. Mod Pathol 13(5):482–488 Xie Y, Skytting B, Nilsson G et al, 2002, The SYT-SSX1 fusion type of synovial sarcoma is associated with increased expression of cyclin A and D1. A link between t(X;18)(p11.2; q11.2, and the cell cycle machinery. Oncogene 21(37):5791–5796 Xie Y, Tornkvist M, Aalto Y et al, 2003, Gene expression profile by blocking the SYT-SSX fusion gene in synovial sarcoma cells. Identification of XRCC4 as a putative SYT-SSX target gene. Oncogene 22(48):7628–7631 Kawai A, Naito N, Yoshida A et al, 2004, Establishment and characterization of a biphasic synovial sarcoma cell line, SYO-1. Cancer Lett 204(1):105–113 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T), method. Methods 25(4):402–408 Siami K, McCluggage WG, Ordonez NG et al, 2007, Thyroid transcription factor-1 expression in endometrial and endocervical adenocarcinomas. Am J Surg Pathol 31(11):1759–1763 Remmele W, Schicketanz KH, 1993, Immunohistochemical determination of estrogen and progesterone receptor content in human breast cancer. Computer-assisted image analysis, QIC score, vs. subjective grading, IRS). Pathol Res Pract 189(8):862– 866 Xie Y, Skytting B, Nilsson G et al, 2002, SYT-SSX is critical for cyclin D1 expression in synovial sarcoma cells: a gain of function of the t(X;18)(p11.2;q11.2, translocation. Cancer Res 62(13):3861–3867 Ramos JW, 2008, The regulation of extracellular signal-regulated kinase, ERK, in mammalian cells. Int J Biochem Cell Biol 40, 12):2707–2719 Yu Q, Geng Y, Sicinski P, 2001, Specific protection against breast cancers by cyclin D1 ablation. Nature 411(6841):1017–1021 Bose S, Chandran S, Mirocha JM, Bose N, 2006, The Akt pathway in human breast cancer: a tissue-array-based analysis. Mod Pathol 19(2):238–245 Hsu MK, Qiao L, Ho V et al, 2006, Ethanol reduces p38 kinase activation and cyclin D1 protein expression after partial hepatectomy in rats. J Hepatol 44(2):375–382 Ramdass B, Maliekal TT, Lakshmi S et al, 2007, Coexpression of Notch1 and NF-kappaB signaling pathway components in human cervical cancer progression. Gynecol Oncol 104(2):352–361 Liu Y, Xi L, Liao G et al, 2007, Inhibition of PC cell-derived growth factor, PCDGF)/granulin-epithelin precursor, GEP, decreased cell proliferation and invasion through downregulation of cyclin D and CDK4 and inactivation of MMP-2. BMC Cancer 7:22 Watanabe M, Miyajima N, Igarashi M, Endo Y, Watanabe N, Sugano S, 2008, Sodium phenylacetate inhibits the Ras/ MAPK signaling pathway to induce reduction of the c-Raf-1 protein in human and canine breast cancer cells. Breast Cancer Res Treat Recchia AG, Musti AM, Lanzino M et al, 2009, A cross-talk between the androgen receptor and the epidermal growth factor receptor leads to p38MAPK-dependent activation of mTOR and cyclinD1 expression in prostate and lung cancer cells. Int J Biochem Cell Biol 41(3):603–614 Bockstaele L, Coulonval K, Kooken H, Paternot S, Roger PP, 2006, Regulation of CDK4. Cell Div 1:25 Lobenhofer EK, Huper G, Iglehart JD, Marks JR, 2000, Inhibition of mitogen-activated protein kinase and phosphatidylinositol 3-kinase activity in MCF-7 cells prevents estrogen-induced mitogenesis. Cell Growth Differ 11(2):99–110 Squires MS, Nixon PM, Cook SJ, 2002, Cell-cycle arrest by PD184352 requires inhibition of extracellular signal-regulated kinases, ERK, 1/2 but not ERK5/BMK1. Biochem J 366(Pt 2):673–680 Motti ML, De Marco C, Califano D et al, 2007, Loss of p27 expression through RAS–>BRAF–>MAP kinase-dependent pathway in human thyroid carcinomas. Cell cycle 6(22):2817– 2825 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 357 EP 367 DI 10.1007/s12253-010-9334-y PG 11 ER PT J AU Zhao, P Lu, Y Liu, L Zhong, M AF Zhao, Po Lu, Yali Liu, Lin Zhong, Mei TI Aberrant Cytoplasmic Expression of Cyclin B1 Protein and its Correlation with EBV-LMP1, P53 and P16(INK4A) in Classical Hodgkin Lymphoma in China SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lymphoma; Cyclin B1; EBV; P53; p16; Immunohistochemistry ID Lymphoma; Cyclin B1; EBV; P53; p16; Immunohistochemistry AB The relationships between the expression of cyclin B1, EBV-LMP1, P53 and P16(INK4A) in Chinese classical Hodgkin lymphoma are unknown and need exploring. Samples of classical Hodgkin lymphoma from 60 Chinese patients were analyzed for the expression of cyclin B1, EBV-LMP1, P53 and P16(INK4A) proteins by immunohistochemistry. Cyclin B1 protein was overexpressed in 90.0% (54/60) of this group of classical Hodgkin lymphoma, staining mainly and strongly in cytoplasm but also sparsely and weakly in nucleus of the Hodgkin and Reed-Sternberg (HRS) cells. EBV-LMP1, P53 and P16 (INK4A) were overexpressed in 85.0%, 96.7% and 71.7% of Hodgkin lymphoma, respectively. EBV-LMP1, P53 and P16(INK4A) were was noted in the nucleus of HRS cells. Microscopically, cyclin B1 and P53 staining distinguished the HRS cells from the complex background of lymphocytes. Cyclin B1 was positively correlated with EBV-LMP1 (P<0.001) and P53(P<0.001), but was inversely related to P16(INK4A) (P<0.05). It is suggested that overexpression of cyclin B1 could play an important role in the evolution of classical Hodgkin lymphoma, and cyclin B1 may be considered as a potential adjunct marker to identify HRS cells in diagnosis and be served as Hodgkin lymphomaassociated antigen in the near future. C1 [Zhao, Po] Chinese People’s Liberation Army (PLA) General Hospital, Department of Pathology, 28 Fuxing Road, 100853 Beijing, China. [Lu, Yali] Chinese People’s Liberation Army (PLA) General Hospital, Department of Pathology, 28 Fuxing Road, 100853 Beijing, China. [Liu, Lin] Chinese People’s Liberation Army (PLA) General Hospital, Department of Pathology, 28 Fuxing Road, 100853 Beijing, China. [Zhong, Mei] Chinese People’s Liberation Army (PLA) General Hospital, Department of Pathology, 28 Fuxing Road, 100853 Beijing, China. RP Zhao, P (reprint author), Chinese People’s Liberation Army (PLA) General Hospital, Department of Pathology, 100853 Beijing, China. EM zhaopo@301hospital.com.cn CR Stein H, Delsol G, Pileri S, Said J, Mann R, Poppema S, Jaffe ES, Swerdlow SH, 2001, Classical Hodgkin lymphoma. In: Jaffe ES, Harris NL, Stein H, Vardiman JW, eds, World Health Organization classification of tumours—tumours of haematopoeitic and lymphoid tissues, Chapter 8. IARC, Lyon, pp 244– 253 Doree M, Galas S, 1994, The cyclin-dependent protein kinases and the control of cell division. FASEB J 8(14):1114–1121 Yasuda M, Takesue F, Inutsuka S, Honda M, Nozoe T, Korenaga D, 2002, Overexpression of cyclin B1 in gastric cancer and its clinicopathological significance: an immunohistological study. J Cancer Res Clin Oncol 128(8):412–416 Korenaga D, Takesue F, Yasuda M, Honda M, Nozoe T, Inutsuka S, 2002, The relationship between cyclin B1 overexpression and lymph node metastasis in human colorectal cancer. Surgery 131(1 Suppl):S114–120 Takeno S, Noguchi T, Kikuchi R, Uchida Y, Yokoyama S, Muller W, 2002, Prognostic value of cyclin B1 in patients with esophageal squamous cell carcinoma. Cancer 94(11):2874–2881 Hassan KA, El-Naggar AK, Soria JC, Liu D, Hong WK, Mao L, 2001, Clinical significance of cyclin B1 protein expression in squamous cell carcinoma of the tongue. Clin Cancer Res 7, 8):2458–2462 Soria JC, Jang SJ, Khuri FR, Hassan K, Liu D, Hong WK, Mao L, 2000, Overexpression of cyclin B1 in early-stage non-small cell lung cancer and its clinical implication. Cancer Res 60(15):4000– 4004 Jin YH, Park CK, 2002, Expression of cyclin B1 and cdc2 in nodal non-Hodgkin’s lymphoma and its prognostic implications. J Korean Med Sci 17(3):322–327 Bai M, Vlachonikolis J, Agnantis NJ, Tsanou E, Dimou S, Nicolaides C, Stefanaki S, Pavlidis N, Kanavaros P, 2001, Low expression of p27 protein combined with altered p53 and Rb/p16 expression status is associated with increased expression of cyclin A and cyclin B1 in diffuse large B-cell lymphomas. Mod Pathol 14(11):1105–1113 Banerjee SK, Weston AP, Zoubine MN, Campbell DR, Cherian R, 2000, Expression of cdc2 and cyclin B1 in Helicobacter pyloriassociated gastric MALT and MALT lymphoma: relationship to cell death, proliferation, and transformation. Am J Pathol 156, 1):217–225 Kao H, Marto JA, Hoffmann TK, Shabanowitz J, Finkelstein SD, Whiteside TL, Hunt DF, Finn OJ, 2001, Identification of cyclin B1 as a shared human epithelial tumor-associated antigen recognized by T cells. J Exp Med 194(9):1313–1323 Porter LA, Cukier IH, Lee JM, 2003, Nuclear localization of cyclin B1 regulates DNA damage-induced apoptosis. Blood 101, 5):1928–1933 Yu M, Zhan Q, Finn OJ, 2002, Immune recognition of cyclin B1 as a tumor antigen is a result of its overexpression in human tumors that is caused by non-functional p53. Mol Immunol 38, 12–13):981–987 Ersvaer E, Zhang JY, McCormack E, Olsnes A, Anensen N, Tan EM, Gjertsen BT, Bruserud O, 2007, Cyclin B1 is commonly expressed in the cytoplasm of primary human acute myelogenous leukemia cells and serves as a leukemia-associated antigen associated with autoantibody response in a subset of patients. Eur J Haematol 79(3):210–225 Sorensen RB, Andersen RS, Svane IM, Engell-Noerregaard L, Hadrup SR, Balslev E, Andersen MH, thor Straten P, 2009, CD8 T-cell responses against cyclin B1 in breast cancer patients with tumors overexpressing p53. Clin Cancer Res 15, 5):1543-1549 Suzuki H, Graziano DF, McKolanis J, Finn OJ, 2005, T celldependent antibody responses against aberrantly expressed cyclin B1 protein in patients with cancer and premalignant disease. Clin Cancer Res 11(4):1521–1526 Hagting A, Karlsson C, Clute P, Jackman M, Pines J, 1998, MPF localization is controlled by nuclear export. EMBOJ 17(14):4127–4138 Yang J, Bardes ES, Moore JD, Brennan J, Powers MA, Kornbluth S, 1998, Control of cyclin B1 localization through regulated binding of the nuclear export factor CRM1. Genes Dev 12, 14):2131–2143 Liu B, Chen XY, 2004, Effect of EB virus latent membrane protein 1 on mitosis control of nasopharyngeal carcinoma cell line CNE1. Ai Zheng 23(5):512–516 Yue W, Shackelford J, Pagano JS, 2006, cdc2/cyclin B1- dependent phosphorylation of EBNA2 at Ser243 regulates its function in mitosis. J Virol 80(4):2045–2050 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 369 EP 373 DI 10.1007/s12253-010-9335-x PG 5 ER PT J AU Narita, D Seclaman, E Ilina, R Cireap, N Ursoniu, S Anghel, A AF Narita, Diana Seclaman, Edward Ilina, Razvan Cireap, Natalia Ursoniu, Sorin Anghel, Andrei TI ADAM12 and ADAM17 Gene Expression in Laser-capture Microdissected and Non-microdissected Breast Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ADAM12; ADAM17; Gene expression; Laser-capture microdissection; Breast tumors ID ADAM12; ADAM17; Gene expression; Laser-capture microdissection; Breast tumors AB ADAM (a disintegrin and metalloprotease)12 and ADAM17 are multidomain transmembrane proteins involved in ectodomain shedding of cytokines, growth factors and adhesion molecules, with pivotal activities in the tumor microenvironment. The aim of this study was to confirm the up-regulation of ADAM17 and ADAM12 gene splicing variants in breast tumors and to delineate their expression between laser-capture microdissected (LCM) and nonmicrodissected breast tumors. The gene expression was analyzed by quantitative-reverse transcription-PCR in a total sample of 109 breast tumors paired with corresponding nonneoplastic breast tissues. ADAM12 and 17 proteins expression for corresponding tissue samples was confirmed by immunohistochemistry. ADAM12S, 12L and 17 genes were significantly up-regulated in either malign or benign LCM samples when compared to non-tumor controls. For non-LCM samples, it was obtained also an increased expression for ADAM12 and 17 genes in cancers, while in benign tumors only ADAM12 variants were significantly upregulated compared to controls. When benign versus malignant tumors were compared, in LCM samples all investigated genes displayed a higher expression in cancers, whereas in non-LCM, ADAM12 variants were overexpressed in benign samples. The increased expression of ADAM12 protein in the tumor cells and stroma of benign breast diseases was immunohistochemically confirmed. These differences between LCM and non-LCM samples were explained by the contribution of the stroma to the expression of this marker. This study underlines the accuracy conferred by homogenous LCM samples on gene expression profiles and confers further evidence regarding the role of ADAM12 and 17 in the breast tumorigenesis and progression. C1 [Narita, Diana] University of Medicine and Pharmacy, Biochemistry Department, Eftimie Murgu Square No. 2A, 300041 Timisoara, Romania. [Seclaman, Edward] University of Medicine and Pharmacy, Biochemistry Department, Eftimie Murgu Square No. 2A, 300041 Timisoara, Romania. [Ilina, Razvan] University of Medicine and Pharmacy, Department of Surgical OncologyTimisoara, Romania. [Cireap, Natalia] University of Medicine and Pharmacy, Department of Surgical OncologyTimisoara, Romania. [Ursoniu, Sorin] University of Medicine and Pharmacy, Department of Public HealthTimisoara, Romania. [Anghel, Andrei] University of Medicine and Pharmacy, Biochemistry Department, Eftimie Murgu Square No. 2A, 300041 Timisoara, Romania. RP Narita, D (reprint author), University of Medicine and Pharmacy, Biochemistry Department, 300041 Timisoara, Romania. EM diana_narita@yahoo.com CR Blobel CP, 2005, ADAMS: key components in EGFR signalling and development. Nat Rev Cancer 6:32–43 Arribas J, Bech-Serra JJ, Santiago-Josefat B, 2006, ADAMs, cell migration and cancer. Cancer Metastasis Rev 25:57–68 Iba K, Albrechtsen R, Gilpin B, Frohlich C et al, 2000, The cysteine-rich domain of human ADAM 12 supports cell adhesion through syndecans and triggers signaling events that lead to β1 integrin-dependent cell spreading. J Cell Biol 149:1143–1156 Ohtsu H, Dempsey PJ, Eguchi S, 2006, ADAMs as mediators of EGF receptor transactivation by G-protein-coupled receptors. Am J Physiol Cell Physiol 291:C1–C10 Mochizuki A, Okada Y, 2007, ADAMs in cancer cell proliferation and progression. Cancer Sci 98:621–628 Murphy G, 2008, The ADAMs: signalling scissors in the tumor microenvironment. Nature Reviews Cancer 8:929–941 Edwards DR, Handsley MM, Pennington CJ, 2008, The ADAM metalloproteinases. Mol Aspects Med 29:258–289 Duffy MJ, McKiernan E, O’Donovan N, McGowan P, 2009, Role of ADAMs in Cancer Formation and Progression. Clin Cancer Res 15:1140–1144 Jacobsen J, Wewer UM, 2009, Targeting ADAM12 in human disease: head, body or tail? Curr Pharm Des 15:2300–2310 Kveiborg M, Albrechtsen R, Couchman JR, Wewer UM, 2008, Cellular roles of ADAM12 in health and disease. Int J Biochem Cell Biol 40:1685–1702 Wewer UM, Morgelin M, Holck P et al, 2006, ADAM12 is a four-leafed clover. The excised prodomain remains bound to the mature enzyme. J Biol Chem 14:9418–9422 Frohlich C, Albrechtsen R, Dyrskjot L et al, 2006, Molecular profiling of ADAM12 in human bladder cancer. Clin Cancer Res 12:7359–7368 Kveiborg M, Frohlich C, Albrechtsen R et al, 2005, A role for ADAM 12 in breast tumor progression and stromal cell apoptosis. Cancer Res 65:4754–4760 Peduto L, Reuter VE, Schara-Fujisawa A et al, 2006, ADAM12 is highly expressed in carcinoma-associated stroma and is required for mouse prostate tumor progression. Oncogene 25:5462–5466 Sorensen HP, Vives RR, Manetopoulos C et al, 2008, Heparan sulfate regulates ADAM12 through a molecular switch mechanism. J Biol Chem 46:31920–31932 O’Shea C, McKie N, Buggy Yet al, 2003, Expression of ADAM- 9 mRNA and protein in human breast cancer. Int J Cancer 105:754–761 Lendeckel U, Kohl J, Arndt M et al, 2005, Increased expression of ADAM family members in human breast cancer and breast cancer cell lines. J Cancer Res Clin Oncol 131:41–48 Kuefer R, Day KC, Kleer CG et al, 2006, ADAM15 disintegrin is associated with aggressive prostate and breast cancer diseases. Neoplasia 8:319–329 Sjoblom T, Jones S, Wood L et al, 2006, The consensus coding sequences of human breast and colorectal cancers. Science 314:268–274 Borrell-Pages M, Rojo F, Albanell J, Baselga J, Arribas J, 2003, TACE is required for the activation of the EGFR by TGF-alpha in tumors. EMBO J 22:1114–1124 Zheng Y, Saftig P, Hartmann D, Blobel C, 2004, Evaluation of the contribution of different ADAMs to tumor necrosis factor α, TNF α, shedding and of the function of the TNF α ectodomain in ensuring selective stimulated shedding by the TNF α convertase, TACE/ADAM17). J Biol Chem 41:42898–42906 Kawaguchi N, Horiuchi K, Becherer JD, Toyama Y, Besmer P, Blobel CP, 2007, Different ADAMs have distinct influences on Kit ligand processing: phorbol-ester-stimulated ectodomain shedding of Kitl1 by ADAM17 is reduced by ADAM19. J Cell Science 120:943–952 Horiuchi K, Le Gall S, Schulte M, Yamaguchi T et al, 2007, Substrate selectivity of epidermal growth factor-receptor ligand sheddases and their regulation by phorbol esters and calcium influx. Mol Biol Cell 18:176–188 McGowan PM, McKiernan E, Bolster F et al, 2008, ADAM-17 predicts adverse outcome in patients with breast cancer. Ann Oncol 19:1075–1081 Zucker S, Cao J, Chen WT, 2000, Critical appraisal of the use of matrix metalloproteinases inhibitors in cancer treatment. Oncogene 19:6642–6650 Zhou BB, Petyon M, He B et al, 2006, Targeting ADAMmediated ligand cleavage toinhibit HER2 and EGFR pathways in non-small cell lung cancer. Cancer Cell 10:39–50 Liu X, Fridman JS, Wang Q et al, 2006, Selective inhibition of ADAM metalloproteases blocks HER-2 extracellular domain, ECD, cleavage and potentiates the anti-tumor effects of trastuzumab. Cancer Biol Ther 5:648–656 Fridman JS, Caulder E, Hansbury M, 2007, Selective inhibition of ADAM metalloproteases as a novel approach for modulating ErbB pathways in cancer. Clin Cancer Res 13:1892–1902 Emmert-Buck MR, Bonner RF, Smith PD et al, 1996, Lasercapture microdissection. Science 274:998–1001 Fend F, Raffeld M, 2000, Laser capture microdissection in pathology. J Clin Pathol 53:666–672 Okuducu AF, Jansen V, Hahne JC et al, 2003, Influence of histochemical stains on quantitative gene expression analysis after laser-assisted microdissection. Int J Mol Med 11:449–453 Espina V, Wulfkuhle JD, Calvert VS et al, 2006, Laser-capture microdissection. Nature Protocols 2:586–602 De Kok JB, Roelofs RW, Giesendorf BA et al, 2005, Normalization of gene expression measurements in tumor tissues: comparison of 13 endogenous control genes. Lab Invest 85:154– 159 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2$$CT method. Methods 25:402–408 Michel C, Desdouets C, Sacre-Salem B et al, 2003, Liver gene expression profiles of rats treated with clofibric acid. Comparison of whole liver and laser capture microdissected liver. Am J Pathol 6:2191–2199 Erickson HS, Albert PS, Gillespie WJ et al, 2007, Normalization strategies for quantitative RT-PCR in microdissected tissue. Lab Invest 87:951–962 Shi Z, Xu W, Loechel F, Wewer UM, Murphy LJ, 2000, ADAM12, a disintegrin and metalloprotease, interacts with insulin-like growth factor-binding protein-3. J Biol Chem 275:18 574–18 580 Laigaard J, Sorensen T, Frohlich C et al, 2003, ADAM12: a novel first-trimester maternal serum marker for Down syndrome. Prenat Diagn 23:1086–1091 Roy R, Wewer MU, Zurakowski D, Pories ES, Moses MA, 2004, ADAM 12 cleaves extracellular matrix proteins and correlates with cancer status and stage. J Biol Chem 49:51323–51330 Pories S, Zurakowski D, Roy R et al, 2008, Urinary metalloproteinases: noninvasive biomarkers for breast cancer risk assessment. Cancer Epidemiol Biomarkers Prev 17:1034–1042 Le Pabic H, Bonnier D, Wewer UM et al, 2003, ADAM12 in human liver cancers: TGF-β-regulated expression in stellate cells is associated with matrix remodeling. Hepatology 37:1056–1066 Kodama T, Ikeda E, Okada A et al, 2004, ADAM12 is selectively overexpressed in human glioblastomas and is associated with glioblastoma cell proliferation and shedding of heparin-binding epidermal growth factor. Am J Pathol 165:1743–1753 Dyczynska E, Syta E, Sun D, Zolkiewska A, 2008, Breast cancerassociated mutations in metalloprotease disintegrin ADAM12 interfere with the intracellular trafficking and processing of the protein. Int J Cancer 122:2634–2640 Atfi A, Dumont E, Colland F et al, 2007, The disintegrin and metalloproteinase ADAM12 contributes to TGF-β signaling through interaction with the type II receptor. J Cell Biol 178:201–208 Bierie B, Moses HL, 2006, Tumor microenvironment: TGFβ: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer 6:506–520 Niimi H, Pardali K, Vanlandewijck M, Heldin CH, Moustakas A, 2007, Notch signaling is necessary for epithelial growth arrest by TGF-β. J Cell Biol 176:695–707 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 375 EP 385 DI 10.1007/s12253-010-9336-9 PG 11 ER PT J AU Yang, L Bian, Y Huang, Sh Ma, X Zhang, Ch Su, X Chen, ZJ Xie, J Zhang, H AF Yang, Ling Bian, Yuehong Huang, Shuhong Ma, Xiaoli Zhang, Chi Su, Xiulan Chen, Zi-Jiang Xie, Jingwu Zhang, Hongwei TI Identification of Signature Genes for Detecting Hedgehog Pathway Activation in Esophageal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal cancer; Hedgehog; Hedgehoginteracting protein; Platelet derived growth factor receptor alpha; Smoothened; Suppressor of fused ID Esophageal cancer; Hedgehog; Hedgehoginteracting protein; Platelet derived growth factor receptor alpha; Smoothened; Suppressor of fused AB The hedgehog signaling pathway plays an important role in cell growth and differentiation both in normal embryonic development and in tumors. Our previous work shows that hedgehog pathway is frequently activated in esophageal cancers. To further elucidate the role of hedgehog pathway in esophageal cancers we examined the expression of the target genes, hedgehog-interacting protein (HIP) and platelet derived growth factor receptor alpha (PDGFRα) and hedgehog signaling molecules, smoothened (SMO), suppressor of fused (Su(Fu)) in the specimens using in-situ hybridization and RT-PCR. We found that HIP, PDGFRα, SMO and Su(Fu) gene highly expressed in the primary esophageal squamous cell carcinomas but not in normal esophageal tissue. The transcripts of HIP, PDGFRα and SMO were expressed in 13 of 15 esophageal cancers. Su(Fu) expression was missing in 2 esophageal cancers. The results from in-situ hybridization were further confirmed by RT-PCR. Our results revealed a set of genes for detecting hedgehog signaling activation in esophageal cancer. C1 [Yang, Ling] Shandong University, School of Life Sciences, Institute of Developmental Biology, 250100 Jinan, China. [Bian, Yuehong] Shandong University, School of Life Sciences, Institute of Developmental Biology, 250100 Jinan, China. [Huang, Shuhong] Shandong University, School of Life Sciences, Institute of Developmental Biology, 250100 Jinan, China. [Ma, Xiaoli] Shandong University, School of Life Sciences, Institute of Developmental Biology, 250100 Jinan, China. [Zhang, Chi] Shandong University, School of Life Sciences, Institute of Developmental Biology, 250100 Jinan, China. [Su, Xiulan] Inner Mongolia Medical College, 010050 Hohhot, China. [Chen, Zi-Jiang] Provincial Hospital Affiliated to Shandong University, Reproductive Medical Center, 250021 Jinan, China. [Xie, Jingwu] Indiana University School of Medicine, Division of hematology and Oncology, Indiana University Simon Cancer Center, Wells Center for Pediatric Research, Department of Pediatrics, 46202 Indianapolis, IN, USA. [Zhang, Hongwei] Shandong University, School of Life Sciences, Institute of Developmental Biology, 250100 Jinan, China. RP Zhang, H (reprint author), Shandong University, School of Life Sciences, Institute of Developmental Biology, 250100 Jinan, China. EM zhw@sdu.edu.cn CR Xie J, Murone M, Luoh SM, Ryan A, Gu Q, Zhang C, Bonifas JM, Lam CW, Hynes M, Goddard A, Rosenthal A, Epstein EH Jr, de Sauvage FJ, 1998, Activating Smoothened mutations in sporadic basal-cell carcinoma. Nature 391:90–2 Lam CW, Xie J, To KF, Ng HK, Lee KC, Yuen NW, Lim PL, Chan LY, Tong SF, McCormick F, 1999, A frequent activated smoothened mutation in sporadic basal cell carcinomas. Oncogene 18:833–6 Kallassy M, Toftgard R, Ueda M, Nakazawa K, Vorechovsky I, Yamasaki H, Nakazawa H, 1997, Patched, ptch)-associated preferential expression of smoothened, smoh, in human basal cell carcinoma of the skin. Cancer Res 57:4731–5 Oniscu A, James RM, Morris RG, Bader S, Malcomson RD, Harrison DJ, 2004, Expression of Sonic hedgehog pathway genes is altered in colonic neoplasia. J Pathol 203:909–17 Shao J, Zhang L, Gao J, Li Z, Chen Z, 2006, Aberrant expression of PTCH, patched gene, and Smo, smoothened gene, in human pancreatic cancerous tissues and its association with hyperglycemia. Pancreas 33:38–44 Martin ST, Sato N, Dhara S, Chang R, Hustinx SR, Abe T, Maitra A, Goggins M, 2005, Aberrant methylation of the Human Hedgehog interacting protein, HHIP, gene in pancreatic neoplasms. Cancer Biol Ther 4:728–33 Taniguchi H, Yamamoto H, Akutsu N, Nosho K, Adachi Y, Imai K, Shinomura Y, 2007, Transcriptional silencing of hedgehoginteracting protein by CpG hypermethylation and chromatic structure in human gastrointestinal cancer. J Pathol 213:131–9 Tada M, Kanai F, Tanaka Y, Tateishi K, Ohta M, Asaoka Y, Seto M, Muroyama R, Fukai K, Imazeki F, Kawabe T, Yokosuka O, Omata M, 2008, Down-regulation of hedgehog-interacting protein through genetic and epigenetic alterations in human hepatocellular carcinoma. Clin Cancer Res 14:3768–76 Svard J, Heby-Henricson K, Persson-Lek M, Rozell B, Lauth M, Bergstrom A, Ericson J, Toftgard R, Teglund S, 2006, Genetic elimination of Suppressor of fused reveals an essential repressor function in the mammalian Hedgehog signaling pathway. Dev Cell 10:187–97 Svard J, Rozell B, Toftgard R, Teglund S, 2009, Tumor suppressor gene co-operativity in compound Patched1 and suppressor of fused heterozygous mutant mice. Mol Carcinog 48:408–19 Sheng T, Li C, Zhang X, Chi S, He N, Chen K, McCormick F, Gatalica Z, Xie J, 2004, Activation of the hedgehog pathway in advanced prostate cancer. Mol Cancer 3:29 Chi S, Huang S, Li C, Zhang X, He N, Bhutani MS, Jones D, Castro CY, Logrono R, Haque A, Zwischenberger J, Tyring SK, Zhang H, Xie J, 2006, Activation of the hedgehog pathway in a subset of lung cancers. Cancer Lett 244:53–60 Reifenberger J, Wolter M, Knobbe CB, Kohler B, Schonicke A, Scharwachter C, Kumar K, Blaschke B, Ruzicka T, Reifenberger G, 2005, Somatic mutations in the PTCH, SMOH, SUFUH and TP53 genes in sporadic basal cell carcinomas. Br J Dermatol 152:43–51 Taylor MD, Liu L, Raffel C, Hui CC, Mainprize TG, Zhang X, Agatep R, Chiappa S, Gao L, Lowrance A, Hao A, Goldstein AM, Stavrou T, Scherer SW, Dura WT, Wainwright B, Squire JA, Rutka JT, Hogg D, 2002, Mutations in SUFU predispose to medulloblastoma. Nat Genet 31:306–10 Rikova K, Guo A, Zeng Q, Possemato A, Yu J, Haack H, Nardone J, Lee K, Reeves C, Li Y, Hu Y, Tan Z, StokesM, Sullivan L,Mitchell J, Wetzel R, Macneill J, Ren JM, Yuan J, Bakalarski CE, Villen J, Kornhauser JM, Smith B, Li D, Zhou X, Gygi SP, Gu TL, Polakiewicz RD, Rush J, Comb MJ, 2007, Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 131:1190–203 Taja-Chayeb L, Chavez-Blanco A, Martinez-Tlahuel J, Gonzalez- Fierro A, Candelaria M, Chanona-Vilchis J, Robles E, Duenas- Gonzalez A, 2006, Expression of platelet derived growth factor family members and the potential role of imatinib mesylate for cervical cancer. Cancer Cell Int 6:22 Zhang T, Sun HC, Xu Y, Zhang KZ, Wang L, Qin LX, Wu WZ, Liu YK, Ye SL, Tang ZY, 2005, Overexpression of plateletderived growth factor receptor alpha in endothelial cells of hepatocellular carcinoma associated with high metastatic potential. Clin Cancer Res 11:8557–63 Matei D, Emerson RE, Lai YC, Baldridge LA, Rao J, Yiannoutsos C, Donner DD, 2006, Autocrine activation of PDGFRalpha promotes the progression of ovarian cancer. Oncogene 25:2060–9 Jechlinger M, Sommer A, Moriggl R, Seither P, Kraut N, Capodiecci P, Donovan M, Cordon-Cardo C, Beug H, Grunert S, 2006, Autocrine PDGFR signaling promotes mammary cancer metastasis. J Clin Invest 116:1561–70 Lev DC, Kim SJ, Onn A, Stone V, Nam DH, Yazici S, Fidler IJ, Price JE, 2005, Inhibition of platelet-derived growth factor receptor signaling restricts the growth of human breast cancer in the bone of nude mice. Clin Cancer Res 11:306–14 Russell MR, Jamieson WL, Dolloff NG, Fatatis A, 2009, The alpha-receptor for platelet-derived growth factor as a target for antibody-mediated inhibition of skeletal metastases from prostate cancer cells. Oncogene 28:412–21 Wehler TC, Frerichs K, Graf C, Drescher D, Schimanski K, Biesterfeld S, Berger MR, Kanzler S, Junginger T, Galle PR, Moehler M, Gockel I, Schimanski CC, 2008, PDGFRalpha/beta expression correlates with the metastatic behavior of human colorectal cancer: a possible rationale for a molecular targeting strategy. Oncol Rep 19:697–704 Ma X, Sheng T, Zhang Y, Zhang X, He J, Huang S, Chen K, Sultz J, Adegboyega PA, Zhang H, Xie J, 2006, Hedgehog signaling is activated in subsets of esophageal cancers. Int J Cancer 118:139–48 Sarbia M, Becker KF, Hofler H, 2004, Pathology of upper gastrointestinal malignancies. Semin Oncol 31:465–75 Bonifas JM, Pennypacker S, Chuang PT, McMahon AP, Williams M, Rosenthal A, De Sauvage FJ, Epstein EH Jr, 2001, Activation of expression of hedgehog target genes in basal cell carcinomas. J Invest Dermatol 116:739–42 Xie J, Aszterbaum M, Zhang X, Bonifas JM, Zachary C, Epstein E, McCormick F, 2001, A role of PDGFRalpha in basal cell carcinoma proliferation. Proc Natl Acad Sci USA 98:9255–9 Heinrich MC, Corless CL, Demetri GD, Blanke CD, von Mehren M, Joensuu H, McGreevey LS, Chen CJ, Van den Abbeele AD, Druker BJ, Kiese B, Eisenberg B, Roberts PJ, Singer S, Fletcher CD, Silberman S, Dimitrijevic S, Fletcher JA, 2003, Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol 21:4342–9 Barnfield PC, Zhang X, Thanabalasingham V, Yoshida M, Hui CC, 2005, Negative regulation of Gli1 and Gli2 activator function by Suppressor of fused through multiple mechanisms. Differentiation 73:397–405 Karhadkar SS, Bova GS, Abdallah N, Dhara S, Gardner D, Maitra A, Isaacs JT, Berman DM, Beachy PA, 2004, Hedgehog signalling in prostate regeneration, neoplasia and metastasis. Nature 431:707–12 Yang L, Huang SH, Bian YH, Ma XL, Zhang HW, Xie JW, 2010, Indentification of signature genes for detecting hedgehog signaling activation in gastric cancer. Mol Med Rep 3:473–478 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 387 EP 391 DI 10.1007/s12253-010-9337-8 PG 5 ER PT J AU Baliko, Z Sarosi, V Illes, BM Varga, Zs Hegedus, G Molnar, P Szakall, Sz AF Baliko, Zoltan Sarosi, Veronika Illes, Balazs Miklos Varga, Zsuzsanna Hegedus, Geza Molnar, Peter Szakall, Szabolcs TI PET-CT Imaging and Reality SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Chronic necrotizing pulmonary aspergillosis; Immunocompromised state; Metastatising colon neoplasia; PET/CT ID Chronic necrotizing pulmonary aspergillosis; Immunocompromised state; Metastatising colon neoplasia; PET/CT AB The spectrum of human diseases caused by members of the Aspergillus genus is extensive. It ranges from allergic reactions to colonization of preexisting pulmonary cavities to invasion and destruction of lung parenchyma with pyemic spread to brain, skin, and other organs, causing rapid death. The immune status of the host is a crucial factor in determining the phenotype and severity of the disease. In this case report Chronic Necrotizing Pulmonary Aspergillosis (CNPA), a rare, locally- or semi-invasive variant of pulmonary Aspergillosis, mimicking lung metastasis is presented. The 60-year-old male patient had earlier received multiple cycles of systemic chemotherapy due to colorectal carcinoma. Our case report focuses on the benefits and the possible disadvantages of PET-CT imaging in CNPA. C1 [Baliko, Zoltan] Baranya County Hospital, Department of Respiratory Medicine, 2 Rakoczi street, 7623 Pecs, Hungary. [Sarosi, Veronika] Baranya County Hospital, Department of Respiratory Medicine, 2 Rakoczi street, 7623 Pecs, Hungary. [Illes, Balazs Miklos] University of Pecs, I. Department of Internal Medicine, 2 Rakoczi street, 7623 Pecs, Hungary. [Varga, Zsuzsanna] Baranya County Hospital, Department of OncologyPecs, Hungary. [Hegedus, Geza] County Hospital of Baranya, Department of PathologyPecs, Hungary. [Molnar, Peter] Szent Imre Hospital, Department of Diagnostic ImagingBudapest, Hungary. [Szakall, Szabolcs] Szent Imre Hospital, Department of Diagnostic ImagingBudapest, Hungary. RP Illes, BM (reprint author), University of Pecs, I. Department of Internal Medicine, 7623 Pecs, Hungary. EM illes.balazs@yahoo.com CR Soubani AO, Chandrasekar PH, 2002, The clinical spectrum of pulmonary aspergillosis. Chest 121:1988–1999 Gefter W, Weingrad T, Epstein D et al, 1981, “Semi-invasive” pulmonary aspergillosis: a new look at the spectrum of Aspergillus infections of the lung. Radiology 140:313–321 Mandell G, Bennett J, Dolin R, eds,, 1995, Principles and practice of infectious diseases. Churchill—Livingstone Bohacs A, Kocsis J, Somoskovi A, Meszaros Z, Sapi Z, Tamasi L, Bartfai Z, 2004, Chronic necrotizing pulmonary aspergillosis in an immunocompromised patient. Orvosi Hetilap 145(35): 1811–1815 Pass HI et al, 2005, Lung cancer—principles and practice. Lippincott Williams & Wilkins NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 393 EP 395 DI 10.1007/s12253-010-9299-x PG 3 ER PT J AU Liu, X Deng, Y Zhang, X Mukherjee, R Huang, W Zhang, G Wang, H Li, X AF Liu, Xi Deng, Yuan Zhang, Xuebin Mukherjee, Rajarshi Huang, Wei Zhang, Guanjun Wang, Hongyan Li, Xiaofeng TI Interdigitating Dendritic Cell Sarcoma Following Adult Liver Transplantation: Case Report and Literature Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Adult; Differential diagnosis; Epstein-Barr virus; Interdigitating dendritic cell sarcoma; Liver transplantation; Tacrolimus ID Adult; Differential diagnosis; Epstein-Barr virus; Interdigitating dendritic cell sarcoma; Liver transplantation; Tacrolimus AB Interdigitating dendritic cell sarcoma is an extremely rare neoplasm derived from professional antigen presenting cells. We report an unusual case of such a tumor occurring in a 61-year-old woman who had undergone orthotopic liver transplantation for stage IVA2 primary hepatocellular carcinoma with a raised preoperative α-fetoprotein level, followed by tacrolimus-based immunosuppressive therapy. During her subsequent management, the tacrolimus blood levels ranged from 7.9 ng/mL to 16.1 ng/mL. Physical examination revealed bilateral neck and left axillary lymphadenopathy. No evidence of either chronic hepatitis B virus or Epstein-Barr virus could be detected in serum. An excisional biopsy of a right neck lymph node was performed. Microscopically, the normal architecture was diffusely effaced by a proliferation of spindled to ovoid cells arrayed in a fascicular, ill-defined whorled pattern and small lymphocytes were admixed in varying numbers with the tumor cells. Immunohistochemical studies showed that the tumor cells were positive for S100 protein, vimentin and CD68. Based on these findings, the case was diagnosed as an interdigitating dendritic cell sarcoma. The patient unfortunately had no response to 2 cycles of CHOP chemotherapy (cyclophosphamide, doxorubicin, vincristine, and prednisone), and died of wide spread disease 6 months after the original biopsy. We propose that tacrolimus-based immunosuppression was associated with the development of interdigitating dendritic cell sarcoma after liver transplantation in this case. C1 [Liu, Xi] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Pathology, 710061 Xi’an, China. [Deng, Yuan] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Pathology, 710061 Xi’an, China. [Zhang, Xuebin] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Pathology, 710061 Xi’an, China. [Mukherjee, Rajarshi] Royal Liverpool University Hospital, Division of Surgery and OncologyLiverpool, UK. [Huang, Wei] Royal Liverpool University Hospital, Division of Surgery and OncologyLiverpool, UK. [Zhang, Guanjun] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Pathology, 710061 Xi’an, China. [Wang, Hongyan] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Pathology, 710061 Xi’an, China. [Li, Xiaofeng] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Pathology, 710061 Xi’an, China. RP Liu, X (reprint author), Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Pathology, 710061 Xi’an, China. EM liuxizgq@gmail.com CR Swerdlow SH, Campo E, Harris NL et al, 2008, WHO classification of tumours of haematopoietic and lymphoid tissues. IARC, Lyon Inaba K, Inaba M, 1999, Cell biologic properties of dendritic cells. Saishinigaku 54:14–21 Fonseca R, Yamakawa M, Nakamura S et al, 1998, Follicular dendritic cell sarcoma and interdigitating reticulum cell sarcoma: a review. Am J Hematol 59:161–167 Turner RR, Wood GS, Beckstead JH et al, 1984, Histiocytic malignancies: morphologic, immunologic, and enzymatic heterogeneity. Am J Surg Pathol 8:485–500 American Liver Tumor Study Group, 1998, A randomized prospective multi-institutional trial of orthotopic liver transplantation or partial hepatic resection with or without adjuvant chemotherapy for hepatocellular carcinoma. Investigator Booklet and Protocol Feltkamp CA, van Heerde P, Feltkamp-Vroom TM et al, 1981, A malignant tumor arising from interdigitating cells; light microscopical, ultrastructural, immuno-and enzyme-histochemical characteristics. Virchows Arch A Pathol Anat Histol 393:183–192 Nakamura S, Hara K, Suchi T et al, 1988, Interdigitating cell sarcoma: a morphologic, immunohistologic, and enzymehistochemical study. Cancer 61:562–568 Weiss LM, Berry GJ, Dorfman RF et al, 1990, Spindle cell neoplasms of lymph nodes of probable reticulum cell lineage: true reticulum cell sarcoma? Am J Surg Pathol 14:405–414 Miettinen M, Fletcher CD, Lasota J, 1993, True histiocytic lymphoma of small intestine: analysis of two S-100 proteinpositive cases with features of interdigitating reticulum cells sarcoma. Am J Clin Pathol 100:285–292 Nakamura S, Koshikawa T, Kitoh K et al, 1994, Interdigitating cell sarcoma: a morphologic and immunologic study of lymph node lesions in four cases. Pathol Int 44:374–386 Vasef MA, Zaatari GS, Chan WC et al, 1995, Dendritic cell tumors associated with low-grade B-cell malignancies. Report of three doses. Am J Clin Pathol 104:696–701 Liu SM, Huang PH, Liu JM, 1998, Interdigitating reticulum cell tumor of lymph node: a case report and literature review. Pathol Int 48:974–980 Luk IS, Shek TW, Tang VW et al, 1999, Interdigitating dendritic cell tumor of the testis: a novel testicular spindle cell neoplasm. Am J Surg Pathol 23:1141–1148 Gaertner EM, Tsokos M, Derringer GA et al, 2001, Interdigitating dendritic cell sarcoma: a report of four cases and review of the literature. Am J Clin Pathol 115:589–597 Kawachi K, Nakatani Y, Inayama Y et al, 2002, Interdigitating dendritic cell sarcoma of the spleen: report of a case with a review of the literature. Am J Surg Pathol 26:530–537 Olnes MJ, Nicol T, Duncan M et al, 2002, Interdigitating dendritic cell sarcoma: a rare malignancy responsive to ABVD chemotherapy. Leuk Lymphoma 43:817–821 Dillon KM, Hill CM, Cameron CH et al, 2002, Mediastinal mixed dendritic cell sarcoma with hybrid features. J Clin Pathol 55:791–794 Barwell N, Howatson R, Jackson R et al, 2004, Interdigitating dendritic cell sarcoma of salivary gland associated lymphoid tissue not associated with HHV-8 or EBV infection. J Clin Pathol 57:87–89 Pillay K, Solomon R, Daubenton JD et al, 2004, Interdigitating dendritic cell sarcoma: a report of four paediatric cases and review of the literature. Histopathology 44:283–291 Androulaki A, Giaslakiotis K, Lazaris AC, 2005, Interdigitating dendritic cell sarcoma/tumour of the tonsil. Br J Haematol 131:415 Rupar G, Beham-Schmid C, Galle G et al, 2005, Interdigitating dendritic cell sarcoma of urinary bladder mimicking large intravesical calculus. Urology 66:1109 Kanaan H, Al-Maghrabi J, Linjawi A et al, 2006, Interdigitating dendritic cell sarcoma of the duodenum with rapidly fatal course: a case report and review of the literature. Arch Pathol Lab Med 130:205–208 Sharma M, Ahsan F, Ah-See KW et al, 2006, Interdigitating dendritic cell sarcoma of the parotid gland. J Laryngol Otol 120:244–246 Cossu A, Deiana A, Lissia A et al, 2006, Synchronous interdigitating dendritic cell sarcoma and B-cell small lymphocytic lymphoma in a lymph node. Arch Pathol Lab Med 130:544– 547 Jo S, Babb MJ, Hilsinger RL Jr, 2006, Interdigitating dendritic cell sarcoma of cervical lymph nodes. Arch Otolaryngol Head Neck Surg 132:1257–1259 Nayer H, Murphy KM, Hawkins AL et al, 2006, Clonal cytogenetic abnormalities and BCL2 rearrangement in interdigitating dendritic cell sarcoma. Leuk Lymphoma 47:2651–2654 Boldin I, Brix-Grunwald G, Scarpatetti MM et al, 2008, Interdigitating dendritic cell sarcoma of the eyelid with a rapidly fatal course. Arch Ophthalmol 126:738–740 Feldman AL, Arber DA, Pittaluga S et al, 2008, Clonally related follicular lymphomas and histiocytic/dendritic cell sarcomas: evidence for transdifferentiation of the follicular lymphoma clone. Blood 111:5433–5439 Lee JC, Christensen T, O’Hara CJ, 2009, Metastatic interdigitating dendritic cell sarcoma masquerading as a skin primary tumor: a case report and review of the literature. Am J Dermatopathol 31:88–93 Kapucuoglu N, Percinel S, Ventura T et al, 2009, Dendritic cell sarcomas/tumours of the breast: report of two cases. Virchows Arch 454:333–339 Adam Z, Pour L, Vesely K et al, 2009, Interdigitating dendritic cell sarcoma of the leg. Onkologie 32:364–365 Efune G, Sumer BD, Sarode VR et al, 2009, Interdigitating dendritic cell sarcoma of the parotid gland: case report and literature review. Am J Otolaryngol 30:264–268 Chen W, Lau SK, Fong D et al, 2009, High frequency of clonal immunoglobulin receptor gene rearrangements in sporadic histiocytic/ dendritic cell sarcomas. Am J Surg Pathol 33:863–873 Fraser CR, Wang W, Gomez M et al, 2009, Transformation of chronic lymphocytic leukemia/small lymphocytic lymphoma to interdigitating dendritic cell sarcoma: evidence for transdifferentiation of the lymphoma clone. Am J Clin Pathol 132:928–939 Cox KL, Lawrence-Miyasaki LS, Garcia-Kennedy R et al, 1995, An increased incidence of Epstein-Barr Virus infection and lymphoproliferative disorder in young children on FK506 after liver transplantation. Transplantation 59:524–529 Cacciarelli TV, Reyes J, Jaffe R et al, 2001, Primary tacrolimus, FK506, therapy and the long-term risk of post-transplant lymphoproliferative disease in pediatric liver transplant recipients. Pediatr Transplant 5:359–364 Kairouz S, Hashash J, Kabbara W et al, 2007, Dendritic cell neoplasms: an overview. Am J Hematol 82:924–928 Grogg KL, Macon WR, Kurtin PJ et al, 2005, A survey of clusterin and fascin expression in sarcomas and spindle cell neoplasms: strong clusterin immunostaining is highly specific for follicular dendritic cell tumor. Mod Pathol 18:260–266 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 397 EP 402 DI 10.1007/s12253-010-9312-4 PG 6 ER PT J AU Mete, Dogan, Kapran, Y Tihan, D Erbil, Y Ozarmagan, S AF Mete, Ozgur Dogan, Oner Kapran, Yersu Tihan, Deniz Erbil, Yesim Ozarmagan, Selcuk TI Intestinal Langerhans Cell Histiocytosis-like Lesion in an Adult Presented with Diverticulitis: A Reactive or Neoplastic Condition? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Langerhans cell histiocytosis; Langerhans cell hyperplasia; Langerhans cell histiocytosis-like lesion; Gastrointestinal tract; Diverticulitis ID Langerhans cell histiocytosis; Langerhans cell hyperplasia; Langerhans cell histiocytosis-like lesion; Gastrointestinal tract; Diverticulitis AB The involvement of the gut by Langerhans cell histiocytosis (LCH) is very rare in adults; however this is usually observed with a disseminated disease in children. We report a 75-year-old male patient who underwent right hemicolectomy for a complicated intestinal diverticular disease. The surgical specimen revealed LCH-like proliferative lesion associated with diverticulitis. The overall morphological and immunohistochemical findings are indistinguishable from LCH. Systemic scans and subsequently performed bone marrow biopsies were free of disease. Although the HUMARA clonality assay cannot be assessed, the lack of evidence of LCH progression or disease elsewhere in the whole body strongly supported the possibility of an atypical reactive phenomenon probably due to the underlying intestinal diverticular disease. Therefore, it is important to avoid diagnosing such a unifocal Langerhans cell proliferation as LCH in patients with underlying pathologies in the absence of systemic involvement. Therefore, without knowledge of clonal status of a unifocal Langerhans cell proliferation, we recommend using the terminology of LCH-like lesion. C1 [Mete, Ozgur] Istanbul Medical Faculty, Department of Pathology, Temel Bilimler Binasi, CapaIstanbul, Turkey. [Dogan, Oner] Istanbul Medical Faculty, Department of Pathology, Temel Bilimler Binasi, CapaIstanbul, Turkey. [Kapran, Yersu] Istanbul Medical Faculty, Department of Pathology, Temel Bilimler Binasi, CapaIstanbul, Turkey. [Tihan, Deniz] Istanbul Faculty of Medicine, Istanbul University, Department of SurgeryIstanbul, Turkey. [Erbil, Yesim] Istanbul Faculty of Medicine, Istanbul University, Department of SurgeryIstanbul, Turkey. [Ozarmagan, Selcuk] Istanbul Faculty of Medicine, Istanbul University, Department of SurgeryIstanbul, Turkey. RP Mete, (reprint author), Istanbul Medical Faculty, Department of Pathology, Istanbul, Turkey. EM ozgurmete77@gmail.com CR Levy J, Khaskelberg A, Garvin J, 2001, Langerhans cell histiocytosis, LCH, involving the gastrointestinal tract: a clinical-electrogastrographic correlation. J Pediatr Gastroenterol Nutr 33:511–514 Santos-Machado TM, Cristofani LM, Almedia MT, Maluf PT, Costa PA, Pereira MA et al, 1999, Disseminated Langerhans cell histiocytosis and massive protein-losing enteropathy. Braz J of Med and Biol Res 32:1095–1099 Egeler RM, Schipper ME, Heymans HS, 1990, Gastrointestinal involvement in Langerhans cell histiocytosis, Histiocytosis X): a clinical report of three cases. Eur J Pediatr 149:325–329 Patel BJ, Chippindale AJ, Gupta SC, 1991, Case report: small bowel histiocytosis-X. Clin Radiol 44:62–63 Terracciano L, Kocher T, Cathomas G, Bubendorf L, Lehmann FS, 1999, Langerhans cell histiocytosis of the stomach with atypical morphological features. Pathol 49:553–556 Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Vardiman JW, eds,, 2008, WHO classification of tumours of haematopoietic and lymphoid tissues. IARC, Lyon Merad M, Ginhoux F, Collin M, 2008, Origin, homeostasis and function of Langerhans cells and other langerin expressing dendritic cells. Nat Rev Immunol 8:935–947 Chang SY, Kweon MN, 2010, Langerin-expressing dendritic cells in gut-associated lymphoid tissues. Immunol Rev 234:233–346 Lau SK, Chu PG, Weiss LM, 2008, Immunohistochemical expression of Langerin in Langerhans cell histiocytosis and non- Langerhans cell histiocytic disorders. Am J Surg Pathol 32:615– 619 Verdijk P, Dijkman R, Plasmeijer EI, Mulder AA, Zoutman WH, Mieke Mommaas A et al, 2005, A lack of Birbeck granules in Langerhans cells is associated with a naturally occurring point mutation in the human langerin gene. J Invest Dermatol 124:714– 717 Kissenpfennig A, Ait-Yahia S, Clair-Moninot V, Stossel H, Badell E, Bordat Y et al, 2005, Disruption of the langerin ⁄ CD207 gene abolishes Birbeck granules without a marked loss of Langerhans cell function. Mol Cell Biol 25:88–99 Negrin-Dastis S, Butenda D, Dorzee J, Fastrez J, D'Odemont JP, 2007, Complete disappearance of lung abnormalities on highresolution computed tomography: a case of histiocytosis X. Can Respir J 14:235–237 Christie LJ, Evans AT, Bray SE, Smith ME, Kernohan NM, Levison DA et al, 2006, Lesions resembling Langerhans cell histiocytosis in association with other lymphoproliferative disorders: a reactive or neoplastic phenomenon? Hum Pathol 37:32–39 Bhattacharjee P, Glusac EJ, 2007, Langerhans cell hyperplasia in scabies: a mimic of Langerhans cell histiocytosis. J Cutan Pathol 34:716–720 Burch JM, Krol A, Weston WL, 2004, Sarcoptes scabiei infestation misdiagnosed and treated as Langerhans cell histiocytosis. Pediatr Dermatol 21:58–62 Singaram C, Sengupta A, Stevens C, Spechler SJ, Goyal RK, 1991, Localization of calcitonin gene-related peptide in human esophageal Langerhans cells. Gastroenterology 100:560–563 Schneider EN, Smoller BR, Lamps LW, 2004, Histiocytic subpopulations in the gastrointestinal tract: distribution and possible relationship to function. Appl Immunohistochem Mol Morphol 12:356–359 Niess JH, Reinecker HC, 2005, Lamina propria dendritic cells in the physiology of the gastrointestinal tract. Curr Opin Gastroenterol 21:687–691 Selim MA, Shea CR. Langerhans cell histiocytosis. eMedicine Specialities, available at http://www.emedicine.com/derm/topic216. htm, last updated 07.02.2007). Giona F, Caruso R, Testi AM, Moleti ML, Malagnino F, Martelli M et al, 1997, Langerhans cell histiocytosis in adults: a clinical and therapeutic analysis of 11 patients from a single institution. Cancer 80:1786–1791 Choi SW, Bangaru BS, Wu CD, Finlay JL, 2003, Gastrointestinal involvement in disseminated Langerhans cell histiocytosis, LCH, with durable complete response to 2-chlorodeoxyadenosine and high-dose cytarabine. J Pediatr Hematol Oncol 25:503–506 Hait E, Liang M, Degar B, Glickman J, Fox VL, 2006, Gastrointestinal tract involvement in Langerhans cell histiocytosis: case report and literature review. Pediatrics 118:e1593–e1599 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 403 EP 407 DI 10.1007/s12253-010-9313-3 PG 5 ER PT J AU Harangi, M Kovacs, T Rakoczi, Rejto, L Miko, L Toth, L Szucs, G Galuska, L Paragh, Gy AF Harangi, Mariann Kovacs, Tibor Rakoczi, Eva Rejto, Laszlo Miko, Laszlo Toth, Laszlo Szucs, Gabriella Galuska, Laszlo Paragh, Gyorgy TI Malignancy or Inflammation? A Case Report of a Young Man with Fever of Unknown Origin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Fever of unknown origin; Chronic osteomyelitis; Vertebral biopsy; Positron emission tomography; Magnetic resonance imaging ID Fever of unknown origin; Chronic osteomyelitis; Vertebral biopsy; Positron emission tomography; Magnetic resonance imaging AB A case of a young man with fever of unknown origin is presented. This diagnosis can be frustrating for both patients and physicians because the diagnostic workup often involves numerous noninvasive and invasive procedures that sometimes fail to explain the fever. In the presented case some of the imaging diagnostic findings suggested malignant hematological disorder. However, histopathological and microbiological investigation proved vertebral osteomyelitis caused by Staphylococcus haemolyticus. Diagnosis was established by positron emission tomography, magnetic resonance imaging, and culture and histopathological analysis of a spinal biopsy. 3 months of antibiotic therapy was curative. Biopsy and microbiological investigation may be necessary in patients with fever, back pain and evidence of a spinal lesion on imaging, even if neoplastic disease is suspected. C1 [Harangi, Mariann] University of Debrecen, Medical and Health Science Center, First Department of Medicine, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Kovacs, Tibor] University of Debrecen, Medical and Health Science Center, First Department of Medicine, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Rakoczi, Eva] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-OncologyDebrecen, Hungary. [Rejto, Laszlo] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Miko, Laszlo] University of Debrecen, Clinical Center, Department of NeurosurgeryDebrecen, Hungary. [Toth, Laszlo] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Szucs, Gabriella] University of Debrecen, Medical and Health Science Center, Department of RheumatologyDebrecen, Hungary. [Galuska, Laszlo] University of Debrecen, Medical and Health Science Center, Institute of Nuclear MedicineDebrecen, Hungary. [Paragh, Gyorgy] University of Debrecen, Medical and Health Science Center, First Department of Medicine, Nagyerdei krt. 98, 4032 Debrecen, Hungary. RP Paragh, Gy (reprint author), University of Debrecen, Medical and Health Science Center, First Department of Medicine, 4032 Debrecen, Hungary. EM paragh@hotmail.com CR Petersdorf RG, Beeson P, 1961, Fever of unexplained origin: report on 100 cases. Medicine, Baltimore, 40:1–30 Roth AR, Basello GM, 2003, Approach to the adult patient with fever of unknown origin. Am Fam Physician 68:2223–2228 Mourad O, Palda V, Detsky AS, 2003, A comprehensive evidence-based approach to fever of unknown origin. Arch Intern Med 163:545–551 Zimmerli W, 2010, Clinical practice. Vertebral osteomyelitis. N Engl J Med 362:1022–1029 Gomez-Rodriguez N, Ferreio-Seoane JL, Ibanez-Ruan J, Servillano-Castano J, 1998, Spondylodiscitis cause by Streptococcus equisimilis. Br J Rheumatol 37:1030–1032 Kumar A, Sandoe J, Kumar N, 2005, Three cases of vertebral osteomyelitis caused by Streptococcus dysgalactiae subsp. equisimilis. J Med Microbiol 54:1103–1105 Carragee EJ, 1997, Pyogenic vertebral osteomyelitis. J Bone Joint Surg Am 79:874–880 Gemmel F, Dumarey N, Palestro CJ, 2006, Radionuclide imaging of spinal infections. Eur J Nucl Med Mol Imaging 33:1226–1237 Kwee TC, Basu S, Cheng G, Alavi A, 2010, FDG PET/CT in carcinoma of unknown primary. Eur J Nucl Med Mol Imaging 37:635–644 Sammak B, Abd El Bagi M, Al Shahed M et al, 1999, Osteomyelitis: a review of currently used imaging techniques. Eur Radiol 9:894–900 Hsu CY, Yu CW, Wu MZ et al, 2008, Unusual manifestations of vertebral osteomyelitis: intraosseous lesions mimicking metastases. AJNR Am J Neuroradiol 29:1104–1110 Gentry LO, 1990, Antibiotic therapy for osteomyelitis. Infect Dis Clin North Am 4:485–499 Legrand E, Flipo RM, Guggenbuhl P et al, 2001, Management of nontuberculous infectious discitis. treatments used in 110 patients admitted to 12 teaching hospitals in France. Joint Bone Spine 68:504–509 Bhavan KP, Kirmani N, 2009, Hematogenous vertebral osteomyelitis. Mol Med 106:277–282 Roblot F, Besnier JM, Juhel L et al, 2007, Optimal duration of antibiotic therapy in vertebral osteomyelitis. Semin Arthritis Rheum 36:269–277 Yoon SH, Chung SK, Kim KJ et al, 2010, Pyogenic vertebral osteomyelitis: identification of microorganism and laboratory markers used to predict clinical outcome. Eur Spine J 19:575– 582 Hassoun A, Taur Y, Singer C, 2006, Evaluation of thin needle aspiration biopsy in the diagnosis and management of vertebral osteomyelitis, VO). Int J Infect Dis 10:486–487 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 409 EP 413 DI 10.1007/s12253-010-9315-1 PG 5 ER PT J AU Myung, KJ Jeong, BJ Han, D Song, SCh Moon, JH Kim, AY Kim, EJ Byun, Sj Kim, HW Chang, SM AF Myung, Kyung Jae Jeong, Bong Ji Han, Daehee Song, Sung Chi Moon, Jong Hyeon Kim, A Young Kim, Eun Ji Byun, Sun-ju Kim, Ho Woo Chang, Soo Mee TI Well-Differentiated Liposarcoma of the Oesophagus: Clinicopathological, Immunohistochemical and Array CGH Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Oesophagus; Well-differentiated liposarcoma; Array comparative genomic hybridization; Chromosome; CDK4; MDM2 ID Oesophagus; Well-differentiated liposarcoma; Array comparative genomic hybridization; Chromosome; CDK4; MDM2 AB Liposarcoma develops extremely rarely in the oesophagus. Microscopically, it exhibits subtle atypia of H&E-stained features. Accordingly, immunohistochemical features and chromosomal alterations are used for its confirmatory diagnosis. However, cytogenetic analysis has not been performed for oesophageal liposarcoma. We studied chromosomal alterations using array comparative genomic hybridization (CGH), as well as endoscopic, radiological, H&E-stained and immunohistochemical features in the oesophageal well-differentiated liposarcoma of a 67-year-old man. Array CGH analysis revealed the presence of high-level amplifications at chromosomal locations 1p12–1q21.2, 12q13.2–12q15 and 12q21.33–12q23.1. At least 29 genes were highly amplified (log2 ratio >2), among which CDK4 and MDM2 were the most highly amplified (log2 ratio >4) and were accepted as major target genes. Moreover, the amplification of AMDHD1, HAL and LTA4H (log2 ratio=3.153) was a novel finding. This case suggests the presence of a characteristic profile of gene amplification in well-differentiated liposarcoma of the oesophagus. The amplified genes may be of pathogenic importance for primary oesophageal well-differentiated liposarcoma. C1 [Myung, Kyung Jae] Seoul National University College of Medicine, Seoul National University Boramae Hospital, Department of Pathology, 425 Shindaebang-dong, Dongjak-gu, 156-707 Seoul, South Korea. [Jeong, Bong Ji] Seoul National University Boramae Hospital, Department of Internal Medicine, 156-707 Seoul, South Korea. [Han, Daehee] Catholic University Seoul St. Mary’s Hospital, Department of Radiology, 137-701 Seoul, South Korea. [Song, Sung Chi] Seoul National University Boramae Hospital, Department of Radiology, 156-707 Seoul, South Korea. [Moon, Jong Hyeon] Seoul National University Boramae Hospital, Department of Thoracic Surgery, 156-707 Seoul, South Korea. [Kim, A Young] Seoul National University College of Medicine, Seoul National University Boramae Hospital, Department of Pathology, 425 Shindaebang-dong, Dongjak-gu, 156-707 Seoul, South Korea. [Kim, Eun Ji] Seoul National University College of Medicine, Seoul National University Boramae Hospital, Department of Pathology, 425 Shindaebang-dong, Dongjak-gu, 156-707 Seoul, South Korea. [Byun, Sun-ju] Seoul National University College of Medicine, Seoul National University Boramae Hospital, Department of Pathology, 425 Shindaebang-dong, Dongjak-gu, 156-707 Seoul, South Korea. [Kim, Ho Woo] Seoul National University College of Medicine, Department of Pathology, 110-779 Seoul, South Korea. [Chang, Soo Mee] Seoul National University College of Medicine, Seoul National University Boramae Hospital, Department of Pathology, 425 Shindaebang-dong, Dongjak-gu, 156-707 Seoul, South Korea. RP Chang, SM (reprint author), Seoul National University College of Medicine, Seoul National University Boramae Hospital, Department of Pathology, 156-707 Seoul, South Korea. EM meesooch@snu.ac.kr CR Mansour KA, Fritz RC, Jacobs DM, Vellios F, 1983, Pedunculated liposarcoma of the esophagus: a first case report. J Thorac Cardiovasc Surg 86:447–450 Boggi U, Viacava P, Naccarato AG, Giulianotti PC, di Candio G, Battolla L, Mosca F, 1997, Giant pedunculated liposarcomas of the esophagus: literature review and case report. Hepatogastroenterology 44:398–407 Garcia M, Buitrago E, Bejarano PA, Casillas J, 2004, Large esophageal liposarcoma: a case report and review of the literature. Arch Pathol Lab Med 128:922–925 Yang B, Shi PZ, Li X, Xu RJ, 2006, Well-differentiated liposarcoma of esophagus. Chin Med J Engl 119:438–440 Will U, Lorenz P, Urban H, Meyer F, 2007, Curative endoscopic resection of huge pedunculated esophageal liposarcoma. Endoscopy 39:E15–E16 Xu S, Xu Z, Hou Y, Tan Y, 2008, Primary pedunculated giant esophageal liposarcoma: case report. Dysphagia 23:327– 330 Fletcher CDM, Unni K, Mertens F, 2000, World health organization classification of tumours. Pathology and genetics. Tumours of soft tissue and bone. IARC, Lyon, pp 35–46 Dei Tos AP, Doglioni C, Piccinin S et al, 2000, Coordinated expression and amplification of the MDM2, CDK4, and HMGI-C genes in atypical lipomatous tumours. J Pathol 190:531–536 Pedeutour F, Forus A, Coindre JM et al, 1999, Structure of the supernumerary ring and giant rod chromosomes in adipose tissue tumors. Genes Chromosom Cancer 24:30–41 Persson F, Olofsson A, Sjogren H, Chebbo N, Nilsson B, Stenman G, Aman P, 2008, Characterization of the 12q amplicons by highresolution, oligonucleotide array CGH and expression analyses of a novel liposarcoma cell line. Cancer Lett 260:37–47 Fritz B, Schubert F, Wrobel G et al, 2002, Microarray-based copy number and expression profiling in dedifferentiated and pleomorphic liposarcoma. Cancer Res 62:2993–2998 Assie G, Guillaud-Bataille M, Ragazzon B, Bertagna X, Bertherat J, Clauser E, 2010, The pathophysiology, diagnosis and prognosis of adrenocortical tumors revisited by transcriptome analyses. Trends Endocrinol Metab 21:325–334 Aleman G, Ortiz V, Langley E, Tovar AR, Torres N, 2005, Regulation by glucagon of the rat histidase gene promoter in cultured rat hepatocytes and human hepatoblastoma cells. Am J Physiol Endocrinol Metab 289:E172–179 Via M, De Giacomo A, Corvol H et al, 2010, Role of LTA4H and ALOX5AP genes in the risk for asthma in Latinos. Clin Exp Allergy 40:482–589 Ishii K, Zaitsu M, Yonemitsu N, Kan Y, Hamasaki Y, Matsuo M, 2009, 5-5-lipoxygenase pathway promotes cell proliferation in human glioma cell lines. Clin Neuropathol 28:445–452 Kubbutat MH, Jones SN, Vousden KH, 1997, Regulation of p53 stability by Mdm2. Nature 387:299–303 Shangary S, Wang S, 2008, Targeting the MDM2-p53 interaction for cancer therapy. Clin Cancer Res 14:5318–5324 Sherr CJ, 1996, Cancer cell cycles. Science 274:1672–1677 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 415 EP 420 DI 10.1007/s12253-010-9322-2 PG 6 ER PT J AU Albayrak, M Balcik, SO Alan, S Dincer, LS Yuksel, KM Albayrak, A AF Albayrak, Murat Balcik, Sahin Ozlem Alan, Saadet Dincer, Lutfu Suleyman Yuksel, Kurt Meltem Albayrak, Aynur TI A Rare Cause of Fever, Hepatosplenomegaly, and Thrombocytopenia: Hepatosplenic Gamma/Delta T-Cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports C1 [Albayrak, Murat] Diskapi Yildirim Beyazit Education and Research Hospital, Department of Hematology, Umut Mah. Sembol Sok. 26/5 Seyranbaglari, CankayaAnkara, Turkey. [Balcik, Sahin Ozlem] Fatih University, Medical School, Department of Hematology, Dizgi Sokak 9/6 Basinevleri, 06120 Ankara, Turkey. [Alan, Saadet] Dr. Abdurrahman Yurtarslan Ankara Oncology Education and Research Hospital, Department of PathologyAnkara, Turkey. [Dincer, Lutfu Suleyman] Yeditepe University, Medical School, Department of HematologyIstanbul, Turkey. [Yuksel, Kurt Meltem] Dr. Abdurrahman Yurtarslan Ankara Oncology Education and Research Hospital, Department of HematologyAnkara, Turkey. [Albayrak, Aynur] Diskapi Yildirim Beyazit Education and Research Hospital, Department of PathologyAnkara, Turkey. RP Balcik, SO (reprint author), Fatih University, Medical School, Department of Hematology, 06120 Ankara, Turkey. EM drozlembalcik@yahoo.com CR Moleti ML, Testi AM, Giona F et al, 2006, Gamma-delta hepatosplenic T-cell lymphoma. Description of a case with immunophenotypic and molecular follow-up successfully treated with chemotherapy alone. Leuk Lymphoma 47, 2):333–336 Weidman E, 2000, Hepatosplenic T cell Lymphoma. A review on 45 cases since the first report describing the disease as a distinct lymphoma entity in 1990. Leukemia 14:991–997 Belhadj K, Reyes F, Farcet JP et al, 2003, Hepatosplenic gammadelta T-cell lymphoma is a rare clinicopathologic entity with poor outcome: report on a series of 21 patients. Blood 15;102, 13):4261–4269 Copie-Bergman C, Wotherspoon AC, Norton AJ et al, 1998, True histiocytic Lymphoma: a morphologic, immunohistochemical, and molecular genetic study of 13 cases. AmJ Surg Pathol 22:1386–1392 Munir J, Preston G, Polish R, 2004, Case report: a common presentation of a rare disease-hepatosplenic T-cell lymphoma. Hawaii Med J 63(11):341–343 Meulenbeld HJ, Spiering W, Nooijen P et al, 2007, Hepatosplenic gammadelta T-cell lymphoma: a case report. Eur J Intern Med 18, 3):241–243 Jaeger G, Bauer F, Brezinschek R et al, 2008, Hepatosplenic gammadelta T-cell lymphoma successfully treated with a combination of alemtuzumab and cladribine. Ann Oncol 19(5):1025–1026 Gopcsa L, Banyai A, Tamaska J et al, 2002, Hepatosplenic gamma delta T-cell lymphoma with leukemic phase successfully treated with 2-chlorodeoxyadenosine. Haematologia, Budap, 32, 4):519–527 Iannitto E, Barbera V, Quintini G et al, 2002, Hepatosplenic gammadelta T-cell lymphoma: complete response induced by treatment with pentostatin. Br J Haematol 117(4):995–996 Corazzelli G, Capobianco G, Russo F et al, 2005, Pentostatin, 2′- deoxycoformycin, for the treatment of hepatosplenic gammadelta T-cell lymphomas. Haematologica 90(3):ECR14 Grigg AP, 2001, 2′-Deoxycoformycin for hepatosplenic gammadelta T-cell lymphoma. Leuk Lymphoma 42(4):797–799 Mittal S, Milner BJ, Johnston PW et al, 2006, A case of hepatosplenic gamma-delta T-cell lymphoma with a transient response to fludarabine and alemtuzumab. Eur J Haematol 76, 6):531–534 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 421 EP 423 DI 10.1007/s12253-010-9324-0 PG 3 ER PT J AU Dobi, Kelemen, Gy Kaizer, L Weiczner, R Thurzo, L Kahan, Zs AF Dobi, Agnes Kelemen, Gyongyi Kaizer, Laszlo Weiczner, Roland Thurzo, Laszlo Kahan, Zsuzsanna TI Breast Cancer under 40 Years of Age: Increasing Number and Worse Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Age; Breast cancer; Incidence; Predictive factors; Prognostic factors ID Age; Breast cancer; Incidence; Predictive factors; Prognostic factors AB Breast cancer at a relatively young age with a poor prognosis is currently exhibiting an increasing incidence. In a retrospective cohort analysis of early breast cancer cases after surgery from our institutional patient registry, 141 patients aged ≤40 years constituted the younger group, with 300 randomly selected patients aged >40 years as controls. A significant and steady increase was found in the relative number of younger cases during the years 2004–2009. The histological type and grade and the lymph node status of the cancers differed significantly between the two groups, with more aggressive biological behaviour, a more advanced stage and a worse prognosis in the younger group. Half of the cancers in the younger cohort were ER-negative, while two-thirds in the control group were ER-positive. Comparativelymore tumours were PR-positive and HER2-negative in the control group than in the younger group. The rates of triple-negative cases were 25% and 13% in the younger age and the control group, respectively (p=0.026). Significantly higher mastectomy and axillary block dissection rates were observed in the younger age group, and more chemotherapy was administered than in the control group. Our findings demonstrate the significance of breast cancer in cases aged <40 years, and draw attention to the need for appropriate care in these cases. C1 [Dobi, Agnes] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Kelemen, Gyongyi] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Kaizer, Laszlo] University of Szeged, Department of PathologySzeged, Hungary. [Weiczner, Roland] University of Szeged, Department of AnatomySzeged, Hungary. [Thurzo, Laszlo] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. RP Dobi, (reprint author), University of Szeged, Department of Oncotherapy, 6720 Szeged, Hungary. EM dobiagnes@gmail.com CR Agarwal G, Pradeep PV, Aggarwal V et al, 2007, Spectrum of breast cancer in Asian women. World J Surg 31:1031–1040 Brinton LA, Sherman ME, Carreon JD, Anderson WF, 2008, Recent trends in breast cancer among younger women in the United States. J Natl Cancer Inst 100:1643–1648 Bouchardy C, Fioretta G, Verkooijen HM et al, 2007, Recent increase of breast cancer incidence among women under the age of forty. Br J Cancer 96:1743–1746 Fredholm H, Eaker S, Frisell J et al, 2009, Breast cancer in young women: poor survival despite intensive treatment. PLoS One 4:e7695 Chung M, Chang HR, Bland KI, Wanebo HJ, 1996, Younger women with breast carcinoma have a poorer prognosis than older women. Cancer 77:97–103 McAree B, O’Donnell ME, Spence A et al, 2010, Breast cancer in women under 40 years of age: A series of 57 cases from Northern Ireland. Breast., DOI 10.1016/j.breast.2009.12.002 Winchester DP, 1996, Breast cancer in young women. Surg Clin North Am 76:279–287 Maggard MA, O’Connell JB, Lane KE et al, 2003, Do young breast cancer patients have worse outcomes? J Surg Res 113:109–113 Fernandopulle SM, Cher-Siangang P, Tan PH, 2006, Breast carcinoma in women 35 years and younger: a pathological study. Pathology 38:219–222 Colleoni M, Rotmensz N, Robertson C et al, 2002, Very young women, <35 years, with operable breast cancer: features of disease at presentation. Ann Oncol 13:273–279 Anders CK, Hsu DS, Breakwater G et al, 2008, Young age at diagnosis correlates with worse prognosis and defines a subset of breast cancers with shared patterns of gene expression. J Clin Oncol 26:3324–3330 Bori R, Cserni G, 2009, Basal phenotype in breast carcinoma occurring in women aged 35 or younger. Pathol Oncol Res 15:41–45 Cabanes A, Vidal E, Perez-Gomez B et al, 2009, Age-specific breast, uterine and ovarian cancer mortality trends in Spain: Changes from 1980–2006. Cancer Epid 33:169–175 Szekely B, Madaras L, Gy S et al, 2010, A fiatal es idoskori emlorak osszehasonlitasa klinikopathologiai jellemzok alapjan. Magy Onkol 54:19–26 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 425 EP 428 DI 10.1007/s12253-010-9305-3 PG 4 ER PT J AU Wang, XX Fu, L Li, X Wu, X Zhu, Z Fu, L Dong, JT AF Wang, Xin-Xin Fu, Liya Li, Xuan Wu, Xiao Zhu, Zhengmao Fu, Li Dong, Jin-Tang TI Somatic Mutations of the Mixed-Lineage Leukemia 3 (MLL3) Gene in Primary Breast Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MLL3; Mutation; Single nucleotide polymorphism; Expression; Breast cancer ID MLL3; Mutation; Single nucleotide polymorphism; Expression; Breast cancer AB The mixed-lineage leukemia 3 (MLL3) gene, which encodes an important component of a histone H3 lysine 4 methyltransferase complex named the ASC-2- and Mll3-containing complex (ASCOM), has been implicated as a tumor suppressor gene due to its frequent mutations in multiple types of human tumors as well as tumor induction upon targeted inactivation of the gene in mice. The role of MLL3 in breast cancer, however, remains unknown. In this study, we sequenced all 59 exons of MLL3 (14.7 Kb coding sequence) in 38 breast cancers from Chinese women, and found three somatic mutations in two of the cases, including one frameshift mutation (c.2687 ins A) that truncates the majority of the MLL3 protein, and two synonymous mutations. In addition to 24 known single nucleotide polymorphisms (SNPs), 5 novel SNPs were also detected in the 38 women; and interestingly, all the 5 novel SNPs alter amino acid sequences of MLL3 thus could have functional consequences. We also examined the expression of MLL3 mRNA in 30 breast tumors and their matched normal breast tissues. While no associations were found between expression change and clinicopathologic parameters, 40% of the samples showed reduced expression in cancer tissues. These results suggest that mutation of MLL3 plays a role in the development of breast cancer. C1 [Wang, Xin-Xin] Cancer Hospital of Tianjin Medical University, Department of Breast Cancer Pathology and Research Laboratory, Huan Hu Xi Road, 300060 Tianjin, China. [Fu, Liya] Cancer Hospital of Tianjin Medical University, Department of Breast Cancer Pathology and Research Laboratory, Huan Hu Xi Road, 300060 Tianjin, China. [Li, Xuan] Nankai University, College of Life Sciences, Department of Genetics and Cell Biology, 94 Weijin Road, 300071 Tianjin, China. [Wu, Xiao] Nankai University, College of Life Sciences, Department of Genetics and Cell Biology, 94 Weijin Road, 300071 Tianjin, China. [Zhu, Zhengmao] Nankai University, College of Life Sciences, Department of Genetics and Cell Biology, 94 Weijin Road, 300071 Tianjin, China. [Fu, Li] Nankai University, College of Life Sciences, Department of Genetics and Cell Biology, 94 Weijin Road, 300071 Tianjin, China. [Dong, Jin-Tang] Nankai University, College of Life Sciences, Department of Genetics and Cell Biology, 94 Weijin Road, 300071 Tianjin, China. RP Fu, L (reprint author), Cancer Hospital of Tianjin Medical University, Department of Breast Cancer Pathology and Research Laboratory, 300060 Tianjin, China. EM fulijyb@hotmail.com CR Ruault M, Brun ME, Ventura M et al, 2002, MLL3, a new human member of the TRX/MLL gene family, maps to 7q36, a chromosome region frequently deleted in myeloid leukaemia. Gene 284(1–2):73–81 Goo YH, Sohn YC, Kim DH et al, 2003, Activating signal cointegrator 2 belongs to a novel steady-state complex that contains a subset of trithorax group proteins. Mol Cell Biol 23, 1):140–149 Lee S, Lee DK, Dou Y et al, 2006, Coactivator as a target gene specificity determinant for histone H3 lysine 4 methyltransferases. Proc Natl Acad Sci USA 103(42):15392–15397 Cho YW, Hong T, Hong S et al, 2007, PTIP associates with MLL3- and MLL4-containing histone H3 lysine 4 methyltransferase complex. J Biol Chem 282(28):20395–20406 Lee MG, Villa R, Trojer P et al, 2007, Demethylation of H3K27 regulates polycomb recruitment and H2A ubiquitination. Science 318(5849):447–450 Patel SR, Kim D, Levitan I et al, 2007, The BRCT-domain containing protein PTIP links PAX2 to a histone H3, lysine 4 methyltransferase complex. Dev Cell 13(4):580–592 Lee J, Saha PK, Yang QH et al, 2008, Targeted inactivation of MLL3 histone H3-Lys-4 methyltransferase activity in the mouse reveals vital roles for MLL3 in adipogenesis. Proc Natl Acad Sci USA 105(49):19229–19234 Tan YC, Chow VT, 2001, Novel human HALR, MLL3, gene encodes a protein homologous to ALR and to ALL-1 involved in leukemia, and maps to chromosome 7q36 associated with leukemia and developmental defects. Cancer Detect Prev 25, 5):454–469 Ahn CH, Yoo NJ, Lee JW et al, 2007, Absence of MLL3 mutations in colorectal carcinomas of Korean patients. APMIS 115(7):859–860 Vakoc CR, Wen YY, Gibbs RA et al, 2009, Low frequency of MLL3 mutations in colorectal carcinoma. Cancer Genet Cytogenet 189(2):140–141 Sjoblom T, Jones S, Wood LD et al, 2006, The consensus coding sequences of human breast and colorectal cancers. Science 314, 5797):268–274 Wood LD, Parsons DW, Jones S et al, 2007, The genomic landscapes of human breast and colorectal cancers. Science 318, 5853):1108–1113 Balakrishnan A, Bleeker FE, Lamba S et al, 2007, Novel somatic and germline mutations in cancer candidate genes in glioblastoma, melanoma, and pancreatic carcinoma. Cancer Res 67(8):3545–3550 Lee J, Kim DH, Lee S et al, 2009, A tumor suppressive coactivator complex of p53 containing ASC-2 and histone H3- lysine-4 methyltransferase MLL3 or its paralogue MLL4. Proc Natl Acad Sci USA 106(21):8513–8518 Stratton MR, Campbell PJ, Futreal PA, 2009, The cancer genome. Nature 458(7239):719–724 Stephens P, Edkins S, Davies H et al, 2005, A screen of the complete protein kinase gene family identifies diverse patterns of somatic mutations in human breast cancer. Nat Genet 37(6):590– 592 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 429 EP 433 DI 10.1007/s12253-010-9316-0 PG 5 ER PT J AU Khanmohammadi, M Bagheri Garmarudi, A Samani, S Ghasemi, K Ashuri, A AF Khanmohammadi, Mohammadreza Bagheri Garmarudi, Amir Samani, Simin Ghasemi, Keyvan Ashuri, Ahmad TI Application of Linear Discriminant Analysis and Attenuated Total Reflectance Fourier Transform Infrared Microspectroscopy for Diagnosis of Colon Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon cancer; Diagnosis; ATR-FTIR; Chemometrics; LDA ID Colon cancer; Diagnosis; ATR-FTIR; Chemometrics; LDA AB Attenuated Total Reflectance Fourier Transform Infrared (ATR-FTIR) microspectroscopy was applied for detection of colon cancer according to the spectral features of colon tissues. Supervised classificationmodels can be trained to identify the tissue type based on the spectroscopic fingerprint. A total of 78 colon tissues were used in spectroscopy studies. Major spectral differences were observed in 1,740–900 cm−1 spectral region. Several chemometric methods such as analysis of variance (ANOVA), cluster analysis (CA) and linear discriminate analysis (LDA) were applied for classification of IR spectra. Utilizing the chemometric techniques, clear and reproducible differences were observed between the spectra of normal and cancer cases, suggesting that infrared microspectroscopy in conjunction with spectral data processing would be useful for diagnostic classification. Using LDA technique, the spectra were classified into cancer and normal tissue classes with an accuracy of 95.8%. The sensitivity and specificity was 100 and 93.1%, respectively. C1 [Khanmohammadi, Mohammadreza] Imam Khomeini International University, Faculty of Science, Chemistry Department, 34149-1-6818 Qazvin, Iran. [Bagheri Garmarudi, Amir] Imam Khomeini International University, Faculty of Science, Chemistry Department, 34149-1-6818 Qazvin, Iran. [Samani, Simin] Qazvin University of Medical Sciences, Pathology DepartmentQazvin, Iran. [Ghasemi, Keyvan] Imam Khomeini International University, Faculty of Science, Chemistry Department, 34149-1-6818 Qazvin, Iran. [Ashuri, Ahmad] Imam Khomeini International University, Faculty of Science, Chemistry Department, 34149-1-6818 Qazvin, Iran. RP Khanmohammadi, M (reprint author), Imam Khomeini International University, Faculty of Science, Chemistry Department, 34149-1-6818 Qazvin, Iran. EM mrkhanmohammadi@gmail.com CR Cancer Facts & Figures, 2009, A report by American Cancer Society, accessed at: http://www.cancer.org Lipkin M, 1987, Proliferation and differentiation of normal and diseased gastrointestinal cells. In: Johnson LR, ed, Physiology of the gastrointestinal tract, 2nd edn. Raven, New York Wargovich MJ, Chen CD, Jimenez A, Steele VE, Velasco M, Stephens LC et al, 1996, Aberrant crypts as a biomarker for colon cancer: evaluation of potential chemopreventive agents in the rat. Cancer Epidemiol Biomark Prev 5:355–360 Deschner EE, 1980, Cell proliferation as a biological marker in human colorectal neoplasia. In: Winawer S, Schottenfield D, Sherlock P, eds, Colorectal cancer: prevention, epidemiology, and screening. Raven, New York Enomoto T, Kuranami M, Kakita A, 2000, Variations in the expression of platelet-derived endothelial cell growth factor in human colorectal polyps. Surg Today 30:711–717 Zonios GI, Cothren RM, Arendt JT, Van Dam J, Crawford JM et al, 1996, Morphological model of human colon tissue fluorescence. IEEE Trans Biomed Eng 43:437–446 Keller JM, Cable S, el Bouhtoury F, Heusser S, Scotto C, Armbruster L et al, 1993, Peroxisome through cell differentiation and neoplasia. Biol Cell 77:77–88 Sandforth F, Heimpel S, Balzer T, Gutschmidt S, Riecken EO, 1988, Characterization of stereomicroscopically identified preneoplastic lesions during dimethylhydrazine-induced colonic carcinogenesis. Eur J Clin Invest 18:655–662 Potten CS, Kellett M, Roberts SA, Rew DA, Wilson GD, 1992, Measurement of in vivo proliferation in human colorectal mucosa using bromodeoxyuridine. Gut 33:71–78 Griffiths DF, Davies SJ, Williams D, Williams GT, Williams ED, 1988, Demonstration of somatic mutation and colonic crypt clonality by X-linked enzyme histochemistry. Nature 333:461– 463 Potten CS, 1992, The significance of spontaneous and induced apoptosis in the gastrointestinal tract of mice. Cancer Metastasis Rev 11:179–195 Yamashita N, Minamoto T, Onda M, Esumi H, 1994, Increased cell proliferation of azoxymethane-induced aberrant crypt foci of rat colon. Jpn J Cancer Res 85:692–698 Furihata T, Kawamata H, Kubota K, Fujimori T, 2002, Evaluation of the malignant potential of aberrant crypt foci by immunohistochemical staining for beta-catenin in inflammation-induced rat colon carcinogenesis. Int J Mol Med 9:353–358 Sharifi N, Ghaffarzadegan K, Ayatollahi H, Shakeri M, Sadeghian M, Bordbar Azari J, 2009, Evaluation of angiogenesis in colorectal carcinoma by CD34 immunohisto-chemistry method and its correlation with clinicopathologic parameters. Acta Med Iran 47:161–164 Chung W, Kwabi-Addo B, Ittmann M, Jelinek J, Shen L, Yu Y, Issa JPJ, 2008, Identification of novel tumor markers in prostate, colon and breast cancer by unbiased methylation profiling. PLoS ONE 3(4):e2079 Mantsch HH, Chapman D, 1996, Infrared spectroscopy of biomolecules. Wiley, New York Jackson M, Kim K, Tetteh J, Mansfield JR, Dolenko B, Somorjai RL et al, 1998, Cancer diagnosis by infrared spectroscopy: methodological aspects. SPIE 3257:24–34 Afanasyeva NI, Kolyakov SF, Artjushenko SG, Sokolov VV, Frank GA, 1998, Minimally invasive and ex vivo diagnostics of breast cancer tissues by fiber optic evanescent wave Fourier Transform IR, FEW-FTIR, Spectroscopy. SPIE 3250:140–146 Diem M, Boydston-White MS, Chiriboga L, 1999, Infrared spectroscopy of cells and tissues: shining light on to a novel subject. Appl Spectrosc 53:148–161 Franck P, Nabet P, Dousset B, 1998, Applications of infrared spectroscopy to medical biology. Cell Mol Biol 44:273–275 Andrus PG, Strickland RD, 1998, Cancer grading by Fourier transform infrared spectroscopy. Biospectroscopy 4:37–46 Diem M, Chiriboga L, Lasch P, Pacifiico A, 2002, IR spectra and IR spectral maps of individual normal and cancerous cells. Biopolymers 67:349–353 German MJ, Hammiche A, Ragavan N, Tobin MJ, Cooper LJ, Matanhelia SS et al, 2006, IR spectroscopy with multivariate analysis potentially facilitates the segregation of different types of prostate cells. Biophys J 90:3783–3795 Argov S, Ramesh J, Salman A, Sinelnikov I, Goldstein J, Guterman H, Mordechai S, 2002, Diagnostic potential of Fourier-transform infrared microspectroscopy and advanced computational methods in colon cancer patients. J Biomed Opt 7:1–7 Wong PTT, Rigas B, 1990, Infrared spectra of microtome sections of human colon tissues. Appl Spectrosc 44:1715–1718 Chen YJ, Hsieh YW, Cheng YD, Liao CC, 2001, Study on the secondary structure of protein in amide I band from human colon cancer tissue by Fourier-transform infrared spectroscopy. Chang Gung Med J 24:541–546 Lasch P, Haensch W, Lewis EN, Kidder LH, Naumann D, 2002, Characterization of colorectal adenocarcinoma sections by spatially resolved FT-IR microspectroscopy. Appl Spect 56:9–36 Lasch P, Haensch W, Naumann D, Diem M, 2004, Imaging of colorectal adenocarcinoma using FT-IR microspectroscopy and cluster analysis. Biochim Biophys Acta 1688:176–186 Krafft C, Codrich D, Pelizzo G, Sergo V, 2008, Raman and FTIR microscopic imaging of colon tissue: a comparative study. J Biophotonics 1(2):154–169 Conti C, Ferraris P, Giorgini E, Rubini C, Sabbatini S, Tosi G, Anastasopoulou J, Arapantoni P, Boukaki E, Konstadoukakis S, Theophanides T, Valavanis C, 2008, FT-IR microimaging spectroscopy: a comparison between healthy and neoplastic human colon tissues. J Mol Struct 881(1–3):46–51 Petter CH, Heigl N, Rainer M, Bakry R, Pallua J, Bonn GK, Huck CW, 2009, Fourier-transform infrared chemical imaging focus on tumour progression in human tissue. Curr Med Chem 16(3):318– 326 Shao X, Mo J, Zheng W, Huang Z, 2008, Near-infrared fluorescence imaging for colonic cancer detection. Proceddings of SPIE, 6848 Khanmohammadi M, Ansari MA, Bagheri GarmarudiA,Hassanzadeh G, Garoosi G, 2007, Cancer diagnosis by discrimination between normal and malignant human blood samples using attenuated total reflectance—Fourier transform infrared spectroscopy. Cancer Investig 25:397–404 Khanmohammadi M, Nasiri R, Ghasemi K, Samani S, Bagheri Garmarudi A, 2007, Diagnosis of basal cell carcinoma by infrared spectroscopy of whole blood samples applying soft independent modeling class analogy. J Cancer Res Clin Oncol doi 10.1007/ s00432-007-0286-x Brereton RG, 2003, Chemometrics: data analysis for the laboratory and chemical plant. Wiley, London Kaufman L, Rousseeuw PJ, 1990, Finding groups in data. Wiley, New York Waddell RJH, NicDaeid N, Littlejohn D, 2004, Classification of ecstasy tablets using trace metal analysis with the application of chemometric procedures and artificial neural network algorithms. Analyst 129:235–240 Lavine BK, 2000, Clustering and classification of analytical data. In: Meyers RA, ed, Encyclopedia of analytical chemistry. Wiley, New York Pisani P, Parkin DM, Bray F, Ferlay J, 1999, Estimates of the worldwide mortality from 25 cancers in 1990. Int J Cancer 83:18–29 Diem M, 1993, Introduction to modern vibrational spectroscopy. Wiley, New York Krupnik E, Jackson M, Bird RP, Smith ICP, Mantsch HH, 1998, Investigation into the infrared spectroscopic characteristics of normal and malignant colonic epithelium. SPIE 3257:307–310 Parker FS, 1971, Application of infrared spectroscopy in biochemistry, biology, and medicine. Plenum, New York NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2011 VL 17 IS 2 BP 435 EP 441 DI 10.1007/s12253-010-9326-y PG 7 ER PT J AU Vegso, Gy Hajdu, M Sebestyen, A AF Vegso, Gyula Hajdu, Melinda Sebestyen, Anna TI Lymphoproliferative Disorders After Solid Organ Transplantation—Classification, Incidence, Risk Factors, Early Detection and Treatment Options SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Adoptive T-cell therapy; Early detection; Epstein-Barr virus; Immunosuppression; Lymphoma; Posttransplant lymphoproliferative disorders; Rituximab; Risk factors; Solid organ transplantation; Therapy ID Adoptive T-cell therapy; Early detection; Epstein-Barr virus; Immunosuppression; Lymphoma; Posttransplant lymphoproliferative disorders; Rituximab; Risk factors; Solid organ transplantation; Therapy AB Posttransplant lymphoproliferative disorder (PTLD) is a heterogeneous disease group of benign and malignant entities. The new World Health Organisation classification introduced in 2008 distinguishes early lesions, polymorphic, monomorphic and classical Hodgkin lymphoma-type PTLD. Based on the time of appearance, early and late forms can be identified. PTLDs are the second most frequent posttransplantation tumors in adulthood, and the most frequent ones in childhood. The incidence varies with the transplanted organ — from 1%–2% following kidney transplantation to as high as 10% following thoracic organ transplantation — due to different intensities in immunosuppression. Immunocompromised state and Epstein-Barr virus (EBV) infection are the two major risk factors. In Europe and the US approximately 85% of PTLDs are of B-cell origin, and the majority are EBV-associated. Symptoms are often unspecific; extranodal, organ manifestations and central nervous system involvement is common. Early lesions respond well to a decrease in immunosuppression. Malignant entities are treated with rituximab, chemotherapy, radiotherapy and surgical therapy. Adoptive T-cell transfer represents a promising therapeutic approach. The prognosis is favorable in early PTLD, and poor in late PTLD. Five-year survival is 30% for high-grade lymphomas. The prognosis of EBV-negative lymphomas is worse. Lowering the risk of PTLD may be achieved by low dose maintenance immunosuppression, immunosuppressive drugs inhibiting cell proliferation, and special immunotherapy (e.g. interleukin-2 inhibitors). Early detection is especially important for high risk — e.g. EBV-negative — patients, where the appearance of EBV-DNA and the increase in its titer may help. C1 [Vegso, Gyula] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, H-1082 Budapest, Hungary. [Hajdu, Melinda] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Vegso, Gy (reprint author), Semmelweis University, Department of Transplantation and Surgery, H-1082 Budapest, Hungary. EM vegso@trans.sote.hu CR Penn I, 1999, Posttransplant malignancies. Transplant Proc 31:1260–1262 Vasudev B, Hariharan S, 2007, Cancer after transplantation. Curr Opin Nephrol Hypertens 16:523–528 ANZDATA A, Zealand N, 2006, Dialysis and Transplant Registry. ANZDATA Registry, Woodville Kasiske BL, Snyder JJ, Gilbertson DT et al, 2004, Cancer after kidney transplantation in the United States. Am J Transplant 4:905–913 Buell JF, Gross TG, Woodle ES, 2005, Malignancy after transplantation. Transplantation 80:S254–S264 Doak PB, Montgomerie JZ, North JD et al, 1968, Reticulum cell sarcoma after renal homotransplantation and azathioprine and prednisone therapy. Br Med J 4:746–748 Starzl TE, 1968)Discussion ofMurray JE,Wilson RE, Tilney NL et al Five years’ experience in renal transplantation with immunosuppressive drugs: survival, function, complications and the role of lymphocyte depletion by thoracic duct fistula. Ann Surg 168:416– 435 Penn I, Starzl TE, 1972, Malignant tumors arising de novo in immunosuppressed organ transplant recipients. Transplantation 14:407 Penn I, 1975, The incidence of malignancies in transplant recipients. Transplant Proc 7:323–326 Starzl TE, Nalesnik MA, Porter KA et al, 1984, Reversibility of lymphomas and lymphoproliferative lesions developing under cyclosporin-steroid therapy. Lancet 1(8377):583–587 Harris NL, Jaffe ES, Diebold J et al, 2000, The World Health Organization classification of neoplastic diseases of the haematopoietic and lymphoid tissues: Report of the Clinical Advisory Committee Meeting. Airlie House, Virginia, November 1997. Histopathology 36:69–86 Harris NL, Swerdlow SH, Frizzera G et al, 2001, Posttransplant lymphoproliferative disorders. In: Jaffe ES, Harris NL, Stein H, Vardiman JW, eds, World Health Organization classification of tumors. Pathology and genetics of tumors of haematopoietic and lymphoid tissue. IARC Press, Lyon, pp 264–269 Swerdlow SH, Webber SA, Chadburn A et al, 2008, Posttransplant lymphoproliferative disorders, PTLD). In: WHO Classification of Tumours of Haematopoietic and Lymphoid Tissue. IARC Press, Lyon, pp 343–350 Mucha K, Foroncewicz B, Ziarkiewicz-Wroblewska B et al, 2010, Post-transplant lymphoproliferative disorder in view of the new WHO classification: a more rational approach to a protean disease ? Nephrol Dial Transplant 25:2089–2098 Parker A, Bowles K, Bradley JA et al, 2010, Diagnosis of posttransplant lymphoproliferative disorder in solid organ transplant recipients—BCSHand BTS Guidelines. Br JHaematol 149:675–692 Tsao L, Hsi ED, 2007, The clinicopathologic spectrum of posttransplantation lymphoproliferative disorders. Arch Pathol Lab Med 131:1209–1218 Taylor AL, Marcus R, Bradley JA, 2005, Post-transplant lymphoproliferative disorders, PTLD, after solid organ transplantation. Crit Rev Oncol Hematol 56:155–167 Hoshida Y, Li T, Dong Z et al, 2001, Lymphoproliferative disorders in renal transplant patients in Japan. Int J Cancer 91:869–875 Guthery SL, Heubi JE, Bucuvalas JE et al, 2003, Determination of risk factors for Epstein-Barr virus-associated posttransplant lymphoproliferative disorder in pediatric liver transplant recipients using objective case ascertainment. Transplantation 75:987–993 Opelz G, Dohler B, 2004, Lymphomas after solid organ transplantation: a collaborative transplant study report. Am J Transplant 4:222–230 Bakker NA, van Imhoff GW, Verschuuren EA et al, 2007, Presentation and early detection of post-transplant lymphoproliferative disorder after solid organ transplantation. Transpl Int 20:207–218 Shahinian VB, Muirhead N, Jevnikar AM et al, 2003, Epstein- Barr virus seronegativity is a risk factor for late-onset posttranslant lymphoproliferative disorder in adult renal allograft recipients. Transplantation 75:851–856 Bakker NA, van Imhoff GW, Verschuuren EA et al, 2005, HLA antigens and post renal transplant lymphoproliferative disease: HLA-B matching is critical. Transplantation 80:595–599 Walker RC, Paya CV, Marshall WF et al, 1995, Pretransplantation seronegative Epstein-Barr virus statusz is the primary risk factor for posttransplantation lymphoproliferative disorder in adult heart, lung, and other solid organ transplantations. J Heart Lung Transplant 14:214–221 Capello D, Rossi D, Gaidano G, 2005, Post-transplant lymphoproliferative disorders: molecular basis of disease histogenesis and pathogenesis. Hematol Oncol 23:61–67 Capello D, Rossi D, Gaudino G et al, 2003, Simian virus 40 infection in lymphoproliferative disorders. Lancet 36:88–89 Cohen AH, Sweet SC, Mendeloff E et al, 2000, High incidence of posttransplant lymphoproliferative disease in pediatric patients with cystic fibrosis. Am J Respir Crit Care Med 161:1252–1255 Newell KA, Alonso EM, Kelly SM et al, 1997, Association between liver transplantation for Langerhans cell histiocytosis, rejection, and development of posttransplant lymphoproliferative disease in children. J Pediatr 131:98–104 Nalesnik MA, Zeevi A, Randhawa PS et al, 1999, Cytokine mRNA profiles in Epstein-Barr virus-associated post-transplant lymphoproliferative disorders. Clin Transplant 13:39–44 Rossi D, Gaidano G, Gloghini A et al, 2003, Frequent aberrant promoter hypermethylation of O6-methylguanine-DNA methyltransferase and death-associated protein kinase genes in immunodeficiency-related lymphomas. Br J Haematol 123:475– 478 Cesarman E, Chadburn A, Liu YF et al, 1998, BCL-6 gene mutations in posttransplantation lymphoproliferative disorders predict response to therapy and clinical outcome. Blood 92:2294–2302 Djokic M, Le Beau MM, Swinnen LJ et al, 2006, Post-transplant lymphoproliferative disorder subtypes correlate with different recurring chromosomal abnormalities. Genes Chromosom Cancer 45:313–318 Boubenider S, Hiesse C, Groupy C et al, 1997, Incidence and consequences of post-transplantation lymphoproliferative disorders. J Nephrol 10:136–145 Opelz G, Henderson R, 1993, Incidence of non-Hodgkin lymphoma in kidney and heart transplant recipients. Lancet 342(8886–8887):1514–1516 Younes BS, McDiarmid SV, Martin MG et al, 2000, The effect of immunosuppression on posttransplant lymphoproliferative disease in pediatric liver transplant patients. Transplantation 70:94–99 Cao S, Cox KL, Berquist Wet al, 1999, Long-term outcomes in pediatric liver recipients: comparison between cyclospirin A and tacrolimus. Pediatr Transplant 3:22–26 Cockfield SM, Preiksaitis J, Harvey E et al, 1991, Is sequential use of ALG and OKT3 in renal transplants associated with an increased incidence of fulminant posttransplant lymphoproliferative disorder? Transplant Proc 23:1106–1107 Caillard S, Dharnidharka V, Agodoa L et al, 2005, Posttransplant lymphoproliferative disorders after renal transplantation in the United States in era ofmodern immunosuppression. Transplantation 80:1233–1243 Kirk AD, Cherikh WS, Ring M et al, 2007, Dissociation of depletional induction and posttransplant lymphoproliferative disease in kidney recipients treated with alemtuzumab. Am J Transplant 7:2619–2625 Kahan BD, Yakupoglu YK, Schoenberg L et al, 2005, Low incidence of malignancy among sirolimus/cyclosporine-treated renal transplant recipients. Transplantation 80:749–758 Kahan BD, Knight R, Schoenberg L et al, 2003, Ten years of sirolimus therapy for human renal transplantation: the University of Texas at Houston experience. Transplant Proc 35:25S–34S Yakupoglu YK, Buell JF, Woodle S et al, 2006, Individualization of immunosuppressive therapy. III. Sirolimus associated with a reduced incidence of malignancy. Transplant Proc 38:358–361 DiNardo CD, Tsai DE, 2010, Treatment advances in posttransplant lymphoproliferative disease. Curr Opin Hematol 17:368– 374 O’Neill JO, Edwards LB, Taylor DO, 2006, Mycophenolate mofetil and risk of developing malignancy after orthotopic heart transplantation: analysis of the transplant registry of the International Society for Heart and Lung Transplantation. J Heart Lung Transplant 25:1186–1191 Robson R, Cecka JM, Opelz G et al, 2005, Prospective registrybased observational cohort study of the long-term risk of malignancies in renal transplant patients treated with mycophenolate mofetil. Am J Transplant 5:2954–2960 Funch DP, Ko HH, Travasso J et al, 2005, Posttransplant lymphoproliferative disorder among renal transplant patients in relation to the use of mycophenolate mofetil. Transplantation 80:1174–1180 Durrbach A, Pestana JM, Pearson T et al, 2010, A phase III study of belatacept versus cyclosporine in kidney transplants from extended criteria donors, BENEFIT-EXT study). Am J Transplant 10:547–557 Pascual J, 2007, Post-transplant lymphoproliferative disorder— the potential of proliferation signal inhibitors. Nephrol Dial Transplant 22:27–35 Dolcetti R, 2007, B lymphocytes and Epstein-Barr virus: the lesson of post-transplant lymphoproliferative disorders. Autoimmun Rev 7:96–101 Carbone A, Gloghini A, Dotti G, 2008, EBV-associated lymphoproliferative disorders: classification and treatment. Oncologist 13:577–585 Snow AL, Martinez OM, 2007, Epstein-Barr virus: evasive maneuvers in the development of PTLD. Am J Transplant 7:271–277 Weiss LM, Movahed LA, Warnke RA et al, 1989, Detection of Epstein-Barr viral genomes in Reed-Sternberg cells of Hodgkin’s disease. N Engl J Med 320:502–506 Raab-Traub N, 2002, Epstein-Barr virus in the pathogenesis of NPC. Semin Cancer Biol 12:431–441 Dolcetti R, Masucci MG, 2003, Epstein-Barr virus: induction and control of cell transformation. J Cell Physiol 196:207–218 Pattle SB, Farrel PJ, 2006, The role of Epstein-Barr virus in cancer. Expert Opin Biol Ther 6:1193–1205 Capello D, Cerri M, Muti G et al, 2003, Molecular histogenesis of posttransplantation lymphoproliferative disorders. Blood 102:3775–3785 Davis JE, Sherritt MA, Bharadwaj M et al, 2004, Determining virological, serological and immunological parameters of EBV infection in the development of PTLD. Int Immunol 16:983–989 Nelson BP, Nalesnik MA, Bahler DW et al, 2000, Epstein-Barr virus-negative posttransplant lymphoproliferative disorders: a distinct entity ? Am J Surg Pathol 24:375–385 Penn I, Porat G, 1995, Central nervous system lymphomas in organ allograft recipients. Transplantation 59:240–244 Bakker NA, van Imhoff GW, Verschuuren EA et al, 2005, Early onset post-transplant lymphoproliferative disease is associated with allograft localization. Clin Transplant 19:327–334 Paranjothi S, Yusen RD, Kraus MD et al, 2001, Lymphoproliferative disease after lung transplantation: comparison of presentation and outcome of early and late cases. J Heart Lung Transplant 20:1054–1063 Savoie A, Perpete C, Carpentier L et al, 1994, Direct correlation between the load of Epstein-Barr virus-infected lymphocytes in the peripheral blood of pediatric transplant patients and risk of lymphoproliferative disease. Blood 83:2715–2722 Riddler SA, Breinig MC, McKnight JL, 1994, Increased levels of circulating Epstein-Barr virus, EBV)-infected lymphocytes and decreased EBV nuclear antigen antibody responses are associated with the development of posttransplant lymphoproliferative disease in solid-organ transplant recipients. Blood 84:972–984 Humar A, Michaels M, 2006, AST ID Working Group on Infectious Disease Monitoring. American Society of Transplantation recommendations for screening, monitoring and reporting of infectious complications in immunosuppression trials in recipients of organ transplantation. Am J Transplant 6:262–274 Patel S, Zuckerman M, Smith M, 2003, Real-time quantitative PCR of Epstein-Barr virus BZLF1 DNA using the LightCycler. J Virol Meth 109:227–233 Fafi-Kremer S, Brengel-Pesce K, Bargues G et al, 2004, Assessment of automated DNA extraction coupled with real-time PCR for measuring Epstein-Barr virus load in whole blood, peripheral mononuclear cells and plasma. J Clin Virol 30:157–164 Wagner HJ, Wessel M, Jabs W et al, 2001, Patients at risk for development of posttransplant lymphoproliferative disorder: plasma versus peripheral blood mononuclear cells as material for quantification of Epstein-Barr viral load by using real-time quantitative polymerase chain reaction. Transplantation 72:1012–1019 Tsai DE, Douglas L, Andreadis C et al, 2008, EBV PCR in the diagnosis and monitoring of posttransplant lymphoproliferative disorder: results of a two-arm prospective trial. Am J Transplant 8:1016–1024 Jang JY, Kim KM, Lee YJ et al, 2008, Quantitative Epstein- Barr virus viral load monitoring in pediatric liver transplantation. Transplant Proc 40:2546–2548 Omar H, Hagglund H, Gustafsson-Jernberg A et al, 2009, Target monitoring of patients at high risk of post-transplant lymphoproliferative disease by quantitative Epstein-Barr virus polymerase chain reaction. Transpl Infect Dis 11:393–399 Smets F, Latinne D, Bazin H et al, 2002, Ratio between Epstein- Barr viral load and anti-Epstein-Barr virus specific T-cell response as a predictive marker of posttransplant lymphoproliferative disease. Transplantation 73:1603–1610 Bianchi E, Pascual M, Nicod M et al, 2008, Clinical usefulness of FDG-PET/CT scan imaging in the management of posttransplant lymphoproliferative disease. Transplantation 85:707–712 Parker A, Bowles K, Bradley JA et al, 2010, Management of post-transplant lymphoproliferative disorder in adult solid organ transplant recipients—BCSH and BTS Guidelines. Br J Haematol 149:693–705 Paya CV, Fung JJ, Nalesnik MA et al, 1999, Epstein-Barr virusinduced posttransplant lymphoproliferative disorders. ASTS/ ASTP EBV-PTLD Task Force and The Mayo Clinic Organized International Consensus Development Meeting. Transplantation 68:1517–1525 Green M, Michaels MG, Webber SA et al, 1999, The management of Epstein-Barr virus associated post-transplant lymphoproliferative disorders in pediatric solid-organ transplant recipients. Pediatr Transplant 3:271–281 Gottschalk S, Rooney CM, Heslop HE, 2005, Post-transplant lymphoproliferative disorders. Annu Rev Med 56:29–44 Muti G, Cantoni S, Oreste P et al, 2002, Cooperative Study Group on PTLDs. Post-transplant lymphoproliferative disorders: improved outcome after clinico-pathologically tailored treatment. Haematologica 87:67–77 Elstrom RL, Andreadis C, Aqui NA et al, 2006, Treatment of PTLD with rituximab or chemotherapy. Am J Transplant 6:569– 576 Gross TG, Bucuvalas JC, Park JR et al, 2005, Low-dose chemotherapy for Epstein-Barr virus-positive post-transplantation lymphoproliferative disease in children after solid organ transplantation. J Clin Oncol 23:6481–6488 Ohta H, Fukushima N, Ozono K, 2009, Pediatric post-transplant lymphoproliferative disorder after cardiac transplantation. Int J Hematol 90:127–136 Milpied N, Vasseur B, Parquet N et al, 2000, Humanized anti- CD20 monoclonal antibody, Rituximab, in post transplant Blymphoproliferative disorder: a retrospective analysis on 32 patients. Ann Oncol 11:113–116 Ganne V, Siddiqi N, Kamaplath B et al, 2003, Humanized anti- CD20 monoclonal antibody, Rituximab, treatment for posttransplant lymphoproliferative disorder. Clin Transplant 17:417– 422 Trappe R, Hinrichs C, Appel U et al, 2009, Treatment of PTLD with rituximab and CHOP reduces the risk of renal graft impairment after reduction of immunosuppression. Am J Transplant 9:2331–2337 Ganne V, Siddiqi N, Kamplath B et al, 2003, Humanized anti- CD20 monoclonal antibody, Rituximab, treatment for posttransplant lymphoproliferative disorder. Clin Transplant 17:417– 422 Heslop HE, Savoldo B, Rooney CM, 2004, Cellular therapy of Epstein-Barr-virus-associated post-transplant lymphoproliferative disease. Best Pract Res Clin Haematol 17:401–413 Savoldo B, Rooney CM, Quiros-Tejeira RE et al, 2005, Cellular immunity to Epstein-Barr virus in liver transplant recipients treated with rituximab for post-transplant lymphoproliferative disease. Am J Transplant 5:566–572 Kinoshita T, Nagai H, Murate T et al, 1998, CD-20-negative relapse in B-cell lymphoma after treatment with Rituximab. J Clin Oncol 16:3916 Choquet S, Leblond V, Herbrecht R et al, 2006, Efficacy and safety of rituximab in B-cell post-transplantation lymphoproliferative disorders: results of a prospective multicenter phase 2 study. Blood 107:3053–3057 Rooney CM, Smith CA, Ng CY et al, 1995, Use of genemodified virus specific T lymphocytes to control Epstein-Barr virus-related lymphoproliferation. Lancet 345:9–13 Rooney CM, Smith CA, Ng CY et al, 1998, Infusion of cytotoxic T cells for the prevention and treatment of Epstein- Barr virus-induced lymphoma in allogeneic transplant recipients. Blood 92:1549–1555 Feng S, Buell JF, Chari RS et al, 2003, Tumors and transplantation: the 2003 third annual ASTS state-of-the-art winter symposium. Am J Transplant 3:1481–1487 Haque T, Wilkie GM, Taylor C et al, 2002, Treatment of Epstein-Barr-virus-positive post-transplantation lymphoproliferative disease with partly HLA-matched allogeneic cytotoxic T cells. Lancet 360(9331):436–442 Haque T, Wilkie GM, Jones MM et al, 2007, Allogeneic cytotoxic T-cell therapy for EBV-positive posttransplantation lymphoproliferative disease: results of a phase 2 multicenter clinical trial. Blood 110:1123–1131 Zhan X, Brown B, Slobod KS et al, 2003, Inhibition of ex vivoexpanded cytotoxic T-lymphocyte function by high-dose cyclosporine. Transplantation 76:739–740 Brewin J, Mancao C, Straathof K et al, 2009, Generation of EBV-specific cytotoxic T-cells that are resistant to calcineurin inhibitors for the treatment of post-transplant lymphoproliferative disease. Blood 114:4792–4803 Davis CL, Wood BL, Sabath DE et al, 1998, Interferon-alpha treatment of posttransplant lymphoproliferative disorder in recipients of solid organ transplants. Transplantation 66:1770– 1779 Davis JE, Moss DJ, 2004, Treatment options for post-transplant lymphoproliferative disorder and other Epstein-Barr virusassociated malignancies. Tissue Antigens 63:285–292 Haddad E, Paczesny S, Leblond V et al, 2001, Treatment of Blymphoproliferative disorder with a monoclonal anti-interleukin- 6 antibody in 12 patients: a multicenter phase 1–2 clinical trial. Blood 97:1590–1597 Perrine SP, Hermine O, Small T et al, 2007, A phase 1/2 trial of arginine butyrate and ganciclovir in patients with Epstein- Barr virus-associated lymphoid malignancies. Blood 109:2571–2578 Preiksaitis JK, 2004, New developments in the diagnosis and management of posttransplantation lymphoproliferative disorders in solid organ transplant recipients. Clin Infect Dis 39:1016– 1023 Funch DP, Walker AM, Schneider G et al, 2005, Ganciclovir and acyclovir reduce the risk of post-transplant lymphoproliferative disorder in renal transplant recipients. Am J Transplant 5:2894– 2900 Malouf MA, Chhajed PN, Hopkins P et al, 2002, Anti-viral profilaxis reduces the incidence of lymphoproliferative disease in lung transplant recipients. J Heart Lung Transplant 21:547–554 Opelz G, Daniel V, Naujokat C et al, 2007, Effect of cytomegalovirus prophylaxis with immunoglobulin or with antiviral drugs on post-transplant non-Hodgkin lymphoma:a multicentre retrospective analysis. Lancet Oncol 8:212–218 Gu SY, Huang TM, Ruan L et al, 1995, First EBV vaccine trial in humans using recombinant vaccinia virus expressing the major membrane antigen. Dev Biol Stand 84:171–177 Posfay-Barbe KM, Siegrist CA, 2009, Immunization and transplantation—what is new and what is coming? Pediatr Transplant 13:404–410 Nepomuceno RR, Balatoni CE, Natkunam Y et al, 2003, Rapamycin inhibits the interleukin 10 signal transduction pathway and the growth of Epstein Barr virus B-cell lymphomas. Cancer Res 63:4472–4480 Alexandru S, Gonzalez E, Grande C et al, 2009, Monotherapy rapamycin in renal transplant recipients with lymphoma successfully treated with rituximab. Transplant Proc 41:2435–2437 Holmes MV, Caplin B, Atkinson C et al, 2009, Prospective monitoring of Epstein-Barr virus DNA in adult renal transplant recipients during the early posttransplant period: role of mycophenolate mofetil. Transplantation 87:852–856 Vegso G, Sebestyen A, Paku S et al, 2007, Antiproliferative and apoptotic effects of mycophenolic acid in human B-cell non- Hodgkin lymphomas. Leuk Res 31:1003–1008 Takebe N, Cheng X, Fandy TE et al, 2006, IMP dehydrogenase inhibitor mycophenolate mofetil induces caspase-dependent apoptosis and cell cycle inhibition in multiple myeloma cells. Mol Cancer Ther 5:457–466 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 443 EP 454 DI 10.1007/s12253-010-9329-8 PG 12 ER PT J AU Diaz Delgado, M Hernandez Amate, A Pereira Gallardo, S Jaramillo, S Virizuela Echaburu, AJ Gonzalez-Campora, JR AF Diaz Delgado, Mario Hernandez Amate, Alicia Pereira Gallardo, Sofia Jaramillo, Sara Virizuela Echaburu, Antonio Juan Gonzalez-Campora, J Ricardo TI Gastrointestinal Stromal Tumors: Morphological, Immunohistochemical and Molecular Changes Associated with Kinase Inhibitor Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Gastrointestinal stromal tumors; GIST; Imatinib mesylate; sunitinib malate; Primary resistance; Secondary resistance; Morphological changes; Molecular changes ID Gastrointestinal stromal tumors; GIST; Imatinib mesylate; sunitinib malate; Primary resistance; Secondary resistance; Morphological changes; Molecular changes AB Recurrent or metastatic GISTs are currently treated with kinase inhibitors since they achieves disease control in 70–85% of patients but this response depend on KIT and PDGFRA gene mutation status. We review the morfological and molecular findings associated to kinase inhibitors administration in GISTs based on the literature on Medline and authors’ own experience. The initial response to kinase inhibitors (imatinib mesylate, Gleevec, Novartis) usually is partial and depend on the mutational KIT or PDGFRA state. Amongst patients wih KIT mutations, the best results are achived in those harboring exon 11 (85%) and exon 9 (45%) mutations. GISTs harboring PDGFRA gene mutations generally respond favorably except those involving the Asp842Val mutation. In the absence of KIT/PDGFRA gene mutations, partial response or disease stabilization is reported in 23% and 50% of patients, respectively, and disease progression in 19%. Histological examination of tumors displaying an initial response to imatinib reveals a highly-variable reduction in the number of tumor cells, a decline in the proliferative index, myxohyaline or sclerohyaline stroma, and a varying degree of bleeding and edema, necrosis and cystification. 72% of patients with initial good response to imatinib, display metastases or new nodule growth within an existing clinically-quiescent tumor after 12–36 months of treatment. This secondary resistance is characterized by a number of well-defined morphological and molecular changes. Histologically, the new growths display increased mitotic activity, pleomorphism, an epithelioid or mixed phenotype and persistent KIT expression although more rarely, dedifferentiation and loss of KIT expression (Fig. 4), as well as trans-differentiation into a rhabdomyosarcoma or epithelial phenotype has been reported. Molecularly, 46–67% of patients present additional KIT mutations, generally in the kinase domain (exons 13, 14 and 17) but also in the ATP-binding domain (exons 15,16) of the same allele. Secondary PDGFRA mutations are very rare. Secondary mutations have not been observed in GISTs not harboring KIT/PDGFRA mutations, or in tumors displaying an unusual morphology or loss of CD117 expression. A number of studies highlight the presence of different resistance mutations within different new tumor nodules, as well as the simultaneous development of distinct resistant tumor subclones within a single lesion (acquired polyclonal resistance). Secondary mutation in genes other than KIT/PDGFRA has only been reported in BRAF (Val600Glu). C1 [Diaz Delgado, Mario] Hospital de Merida, Servicio de Anatomia PatologicaBadajoz, Spain. [Hernandez Amate, Alicia] Hospital Infanta Cristina, Servicio de Anatomia PatologicaBadajoz, Spain. [Pereira Gallardo, Sofia] Virgen Macarena University Hospital and University of Seville Medical School, Department of Pathology, Avda. Dr. Fedriani s/n, 41009 Seville, Spain. [Jaramillo, Sara] Virgen Macarena University Hospital and University of Seville Medical School, Department of Pathology, Avda. Dr. Fedriani s/n, 41009 Seville, Spain. [Virizuela Echaburu, Antonio Juan] Virgen Macarena University Hospital, Oncology ServiceSeville, Spain. [Gonzalez-Campora, J Ricardo] Virgen Macarena University Hospital and University of Seville Medical School, Department of Pathology, Avda. Dr. Fedriani s/n, 41009 Seville, Spain. RP Gonzalez-Campora, JR (reprint author), Virgen Macarena University Hospital and University of Seville Medical School, Department of Pathology, 41009 Seville, Spain. EM rcampora@us.es CR Miettinen M, Lasota J, 2006, Gastrointestinal Stromal Tumors. Review on morphology, molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med 130:1466–1478 Kindblom LG, Remotti HE, Aldenborg F, Meis-Kindblom JM, 1998, Gastrointestinal pacemaker cell tumor, GIPACT): gastrointestinal stromal tumors show phenotypic characteristics of the interstitial cells of Cajal. Am J Pathol 152:1259–1269 Joensuu H, Roberts PJ, Sarlomo-Rikala M et al, 2001, Effect of the tyrosine kinase inhibitor STI571 in a patient with a metastatic gastrointestinal stromal tumor. N Engl J Med 344:1052–1056 Corless CL, Heinrich MC, 2008, Molecular pathobiology of gastrointestinal stromal sarcomas. Annu Rev Pathol 3:557–86 Dematteo RP, Heinrich MC, El-Rifai WM, Demetri G, 2002, Clinical mamagement of gastrointestinal stromal tumor: before and after STI-571. Hum Pathol 33:466–477 Heinrich MC, Corless CL, Demetri GD et al, 2003, Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol 21:4342–4349 Debiec-Rychter M, Wasag B, Stul M et al, 2004, Gastrointestinal stromal tumours, GISTs, negative for KIT, CD117 antigen, immunoreactivity. J Pathol 202:430–438 Debiec-Rychter M, 2007, Sunitinib -a new approach following failure of imatinib in patients with advanced gastrointestinal stromal tumor-. Nat Clin Pract Oncol 4:342–343 Lasota J, Miettinen M, 2008, Clinical significance of oncogenic KIT and PDGFRA mutations in gastrointestinal stromal tumours. Histopathology 53:245–266 Dematteo RP, Ballman KV, Antonescu CR et al, 2009, Adjuvant imatinib mesylate after resection of localised, primary gastrointestinal stromal tumour: a randomised, double-blind, placebocontrolled trial. Lancet 373:1097–1104 Demetri GD, van Oosterom AT, Garrett CR et al, 2006, Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet 368:1329–1338 Heinrich MC, Maki RG, Corless CL et al, 2008, Primary and secondary kinase genotypes correlate with the biological and clinical activity of sunitinib in imatinib-resistant gastrointestinal stromal tumor. J Clin Oncol 26:5352–5359 Abdulkader I, Cameselle-Teijeiro J, Forteza J, 2006, Pathological changes related to Imatinib treatment in a patient with a metastatic gastrointestinal stromal tumour. Histopathology 46:99–100 Agaram NP, Besmer P, Wong GC et al, 2007, Pathologic and molecular heterogeneity in imatinib-stable or imatinib-responsive gastrointestinal stromal tumors. Clin Cancer Res 13:170–181 Bickenbach K, Wilcox R, Veerapong J et al, 2007, A review of resistance patterns and phenotypic changes in gastrointestinal stromal tumors following imatinib mesylate therapy. J Gastrointest Surg 11:758–766 Antonescu CR, Besmer P, Guo T et al, 2005, Acquired resistance to imatinib in gastrointestinal stromal tumor occurs through secondary gene mutation. Clin Cancer Res 11:4182–4190 Pauwels P, Debiec-Rychter M, Stul M et al, 2005, Changing phenotype of gastrointestinal stromal tumours under imatinib mesylate treatment: a potential diagnostic pitfall. Histopathology 47:41–47 Liegl B, Hornick JL, Antonescu CR et al, 2009, Rhabdomyosarcomatous differentiation in gastrointestinal stromal tumors after tyrosine kinase inhibitor therapy: a novel form of tumor progression. Am J Surg Pathol 33:218–226 Wardelmann E, Schildhaus HU, Merkelbach-Bruse S, Buttner R, 2006, Therapeutic targets in gastrointestinal stromal tumors. Verh Dtsch Ges Pathol 90:73–79 Heinrich MC, Corless CL, Blanke CD et al, 2006, Molecular correlates of imatinib resistance in gastrointestinal stromal tumors. J Clin Oncol 24:4764–4774 Liegl B, Kepten I, Le C, Zhu M et al, 2008, Heterogeneity of kinase inhibitor resistance mechanisms in GIST. J Pathol 216:64–74 Mahadevan D, Cooke L, Riley C et al, 2007, A novel tyrosine kinase switch is a mechanism of imatinib resistance in gastrointestinal stromal tumors. Oncogene 26:3909–3919 Agaram NP, Wong GC, Guo T, Maki RG et al, 2008, Novel V600E BRAF mutations in imatinib-naive and imatinib-resistant gastrointestinal stromal tumors. Genes Chromosom Cancer 47:853–859 Schoffski P, Reichardt P, Blay JY et al, 2010, A phase I-II study of everolimus, RAD001, in combination with imatinib in patients with imatinib-resistant gastrointestinal stromal tumors. Ann Oncol 21:1990–1998 Bauer S, Yu LK, Demetri GD, Fletcher JA, 2006, Heat shock protein 90 inhibition in imatinib-resistant gastrointestinal stromal tumor. Cancer Res 66:9153–9161 Sambol EB, Ambrosini G et al, 2006, Flavopiridol targets c-KIT transcription and induces apoptosis in gastrointestinal stromal tumor cells. Cancer Res 66:5858–5866 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 455 EP 461 DI 10.1007/s12253-011-9362-2 PG 7 ER PT J AU Bene, L Falus, A Baffy, N Fulop, KA AF Bene, Laszlo Falus, Andras Baffy, Noemi Fulop, Kristof Andras TI Cellular and Molecular Mechanisms in the Two Major Forms of Inflammatory Bowel Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Crohn’s disease; Inflammatory bowl disease; Intestinal tract; Ulcerative colitis ID Crohn’s disease; Inflammatory bowl disease; Intestinal tract; Ulcerative colitis AB The factors involved in the pathogenesis of Crohn’s disease and ulcerative colitis, the two major types of inflammatory bowel disease (IBD) are summarized. Intestinal antigens composed of bacterial flora along with antigen presentation and impaired mucosal barrier have an important role in the initiation of IBD. The bacterial community may be modified by the use of antibiotics and probiotics. The dentritic cells recognize the antigens by cell surface Toll like receptor and the cytoplasmic CARD/NOD system. The balance between Th1/Th2/Th17 cell populations being the source of a variety of cytokines regulates the inflammatory mechanisms and the clearance of microbes. The intracellular killing and digestion, including autophagy, are important in the protection against microbes and their toxins. The homing process determines the location and distribution of the immune cells along the gut. All these players are potential targets of pharmacological manipulation of disease status. C1 [Bene, Laszlo] Peterfy Hospital, Department of Internal MedicineBudapest, Hungary. [Falus, Andras] Semmelweis University, Department of Genetics, Cell and ImmunobiologyBudapest, Hungary. [Baffy, Noemi] Semmelweis University, Department of Genetics, Cell and ImmunobiologyBudapest, Hungary. [Fulop, Kristof Andras] Semmelweis University, Department of Genetics, Cell and ImmunobiologyBudapest, Hungary. RP Falus, A (reprint author), Semmelweis University, Department of Genetics, Cell and Immunobiology, Budapest, Hungary. EM faland@dgci.sote.hu CR Tresca AJ, 2009, Differences Between Ulcerative Colitis and Crohn’s Disease. In: About.com: Health. http://ibdcrohns.about. com/od/ulcerativecolitis/a/diffuccd.htm. Cited 16. Jun 2010 Balfour SR, 2001, Induction of mucosal immune responses by bacteria and bacterial components. Curr Opin Gastroenterol 17:555–561 Mai V, Morris JG, 2004, Colonic bacterial flora: changing understandings in the molecular age. J Nutr 134:459–464 Wilson KH, 2002, Natural biota of the human gastrointestinal tract. In: Blaser MJ, Smith PD, Ravdin JI, eds, gastrointestinal tract. Lippincott Williams & Wilkins, Philadelphia Danese S, Sans M, Fiocchi C, 2004, Inflammatory bowel disease: the role of environmental factors. Autoimmun Rev 3:394–400 Cong Y, Weaver CT, Lazenby A et al, 2002, Bacterial-reactive T regulatory cells inhibit pathogenic immune responses to the enteric flora. J Immunol 169:6112–6119 Bai PA, Ouyang Q, 2006, Probiotics and inflammatory bowel disease. Postgrad Med J 82:376–382 Groux H, O'Garra A, Bigler M et al, 1997, A CD4+ T-cell subset inhibits antigen specific T-cell responses and prevents colitis. Nature 389:737–742 Abbas AK, Murphy KM, Sher A, 1996, Functional diversity of helper T lymphocytes. Nature 383:787–793 Boirivant M, Marini M, Di Felice G et al, 1999, Lamina propria T-cells in Crohn’s disease and other gastrointestinal inflammation show defective CD2 pathway-induced apoptosis. Gastroenterology 116:557–565 Bu P, Keshavarzian A, Stone DD et al, 2001, Apoptosis:one of the mechanisms that maintains unresponsivness of the intestinal mucosal immune system. J Immunol 166:6399–6403 Nagler-Anderson C, Bober LA, Robinson ME et al, 1986, Suppression of type II collagen- induced arthritis by intragastric administration of soluble type II. collagen. Proc Natl Acad Sci USA 83:7443–7446 Higgins PJ, Weiner HL, 1988, Suppression of experimental encephalomyelitis by oral administration of myelin basic protein and its fragments. J Immunol 140:440–445 Vrabec TR, Gregerson DS, Dua HS et al, 1992, Inhibition of experimental autoimmun uveoretinitis by oral administration by oral administration of S-antigen and syntetic peptides. Autoimmunity 12:175–184 Neurath MF, Fuss I, Kelsall BL et al, 2004, Experimental granulomatous colitis in mice abrogated by induction of TGF-β mediated oral tolerance. J Exp Med 183:2605–2616 Kraus TA, Toy L, Chan L et al, 2004, Failure to induce oral tolerance in Crohn’s disease and ulcerative colitis patients: posible genetic risk. Ann NY Acad Sci 1029:225–238 Ilian Y, 2004, Oral immune ragulation toward disease-assiciated antigens: results of phase I clinical trials in Crohn’s disease and chronic hepatitis. Ann NY Acad Sci 1029:286–298 Hart AL, Stagg AJ, Kamm MA, 2003, Use of probiotics in the treatment of inflammatory bowel disease. J Clin Gastroenterol 36:111–119 Niess JH, 2008, Role of mucosal dendritic cells in inflammatory bowel disease. World J Gastroenterol 14:5138–5148 Coombes JL, Maloy KJ, 2007, Control of intestinal homeostasis by regulatory T-cells and dendritic cells. Semin Immunol 19:116–126 Iwasaki A, Kelsall BL, 2001, Unique functions of CD11b+, CD8 alpha+, and double-negative Peyer’s patch dendritic cells. J Immunol 166:4884–4890 Hart AL, Lammers K, Brigidi P et al, 2004, Modulation of human dendritic cell phenotype and function by probiotic bacteria. Gut 53:1602–1609 Macpherson AJ, Uhr T, 2004, Induction of protective IgA by intestinal dendritic cells carrying commensal bacteria. Science 303:1662–1665 Mora JR, Bono MR, Manjunath N et al, 2003, Selective imrprinting of gut-homing T cells by Peyer’s patch dendritic cells. Nature 424:88–93 Krajina T, Leithauser F, Moller P et al, 2003, Colonic lamina propria dendritic cells in mice with CD4+ T cell-induced colitis. Eur J Immunol 33:1073–1083 Bell SJ, Rigby R, English N et al, 2001, Migration and maturation of human colonic dendritic cells. J Immunol 166:4958–4967 Rimoldi M, Chieppa M, Salucci V et al, 2005, Intestinal immun homeostasis is regulated by the crosstalk between epithelial cells and dendritic cells. Nat Immunol 6:507–514 Xavier RJ, Podolsky DK, 2007, Unravelling the pathogenesis of inflammatory bowel disease. Nature 448:427–434 Suzuki M, Hisamatsu T, Podolsky DK, 2003, Gamma interferon augments the intracellular pathway for lipopolysaccahride, LPS, recognition in human intestinal epithelial cells through coordinated upregulation of LPS uptake and expression of the intracellular Tolle-like receptor 4-MD-2 complex. Infect Immun 71:3503–3511 Ortega-Cava CF, Ishihara S, Rumi MA et al, 2003, Strategic compertmentalization of toll-like receptor 4 in the mouse gut. J Immunol 170:3977–3985 Abreu MT, Vora P, Faure E et al, 2001, Decreased expression of Toll-like receptor-4 and MD-2 correlates with intestinal epithelial cell protection adainst dysregulated proinflammatory gene expression in response to bacterial lipopolysaccharide. J Immunol 167:1609–1616 Cario E, Podolsky DK, 2000, Differencial alteration in intestinal epithelial cell expression of toll like receptor 3, TLR3, and TLR4 in inflammatory bowel disease. Infect Immun 68:7010–7017 Dubuquoy L, Jansson EA, Deeb S et al, 2003, Impaired expression of peroxisome proliferator-activated receptor gamma in ulcerative colitis. Gastroenterology 124:1265–1276 Frolova L, Drastich P, Rossmann P et al, 2008, Expression of Toll-like receptor 2, TLR2), TLR4, and CD14 in biopsy samples of patients with inflammatory bowel diseases: upregulated expression of TLR2 in terminal ileum of patients with ulcerative colitis. Histochem Cytochem 56:267–274 Lakatos PL, Kiss LS, Palatka K et al, 2011, Serum lipopolysaccharide-binding protein and soluble CD14 are markers of disease activity in patients with Crohn’s disease. Inflamm Bowel Dis 17:767–777 Trinchieri G, 2003, Interleukin-12 and regulation of innate resistance and adaptive immunity. Nat Rev Immunol 3:133–146 Mosmann TR, Cherwinski H, Bond MWet al, 1986, Two types of murine helper T cell clone. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol 136:2348–2357 Hayashi F, Smith KD, Ozinsky A et al, 2001, The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature 410:1099–1103 Papp M, Altorjay I, Norman GL et al, 2007, Seroreactivity to microbial components in Crohn’s disease is associated with ileal involvement, noninflammatory disease behavior and NOD2/ CARD15 genotype, but not with risk for surgery in a Hungarian cohort of IBD patients. Inflamm Bowel Dis 13:984–992 Abreu MT, Taylor KD, Lin YC et al, 2002, Mutations in NOD2 are associated with fibrostenosing disease in patients with Crohn’s disease. Gastroenterology 123:679–688 Szamosi T, Lakatos PL; Hungarian IBD Study Group et al, 2009, The 3′UTR NFKBIA variant is associated with extensive colitis in Hungarian IBD patients. Dig Dis Sci 54:351–359 Hugot JP, Chamaillard M, Zouali H et al, 2001, Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature 411:599–603 Chamaillard M, Philpott D, Girardin SE, 2003, Gene enviroment interaction modulated by allelic heterogenety in inflammatory bowel diseases. Proc Natl Acad Sci USA 100:3455–3460 Hugot J-P, 2006, CARD15/NOD2 mutations in Crohn’s disease. Ann NY Acad Sci 1072:9–18 Abreu MT, Arnold ET, Thomas LS et al, 2002, TLR4 and MD-2 expression is regulated by immune medaited signals in human epithelial cells. J Biol Chem 277:20431–20437 Weigmann B, Nemetz A, Becker C et al, 2004, A critical regulatory role of leucin zipper transcription factor c-Maf in Th1- mediated experimental colitis. J Immunol 173:3446–3455 Elson CO, Cong Y, Iqbal N et al, 2001, Immuno-bacterial homeostasis in gut: new insight into the old enigma. Semin Immunol 13:187–194 Boirivant M, Fuss IJ, Chu A et al, 1998, Oxazolone colitis: a murine model of T helper cell type 2 colitis treatable with antibodies to interleukin 4. J Exp Med 188:1929–1939 Maeda S, Hsu LC, Liu H et al, 2005, Nod2 mutation in Crohn’s disease potentiates NF-kappaB activity and IL-1beta processing. Science 307:734–738 Girardin SE, Boneca IG, Carneiro LA et al, 2003, Nod1 detects a unique muropeptide from gram-negative bacterial peptidoglycan. Science 300:1584–1587 Lala S, Ogura Y, Osborne C et al, 2003, Crohn’s disease and the NOD2 gene: a role for paneth cells. Gastroenterology 125:47–57 Ramasundara M, Leach ST, Lemberg DA, Day AS, 2009, Defensins and inflammation: the role of defensins in inflammatory bowel disease. J Gastroenterol Hepatol 24:202–208 Inohara N, Ogura Y, Fontalba A et al, 2003, Host recognition of bacterial muramyl dipeptide mediated through NOD2. Implications for Crohn’s disease. J Biol Chem 278:5509–5512 Berrebi D, Maudinas R, Hugot JP et al, 2003, Card15 gene overexpression in mononuclear and epithelial cells of the inflammed Crohn’s disease colon. Gut 52:840–846 Hisamatsu T, Suzuki M, Reinecker HC, 2003, CARD15/NOD2 functions as an antibacterial factor in human intestinal epithelial cells. Gastroenterology 24:1001–1009 Brimnes J, Reimann J, Nissen M et al, 2001, Enteric bacterial antigens activate CD4(+, T cells from scid mice with inflammatory bowel disease. Eur J Immunol 31:23–31 Peluso I, Pallone F, Monteleone G, 2006, Interleukin-12 and Th1 immune response in Crohn’s disease: pathogenetic relevance and therapeutic implication. World J Gastroenterol 12:5606–5610 Neurath MF, Weigmann B, Finotto S et al, 2002, The transcription factor T-bet regulates mucosal T cell activation in experimental colitis and Cronh’s disease. J Exp Med 195:1129–1143 Afkarian M, Sedy JR, Yang J et al, 2002, T-bet is a STAT1- induced regulator of IL-12R expression in naive CD4+ T cells. Nat Immunol 3:506–508 Heller F, Fuss IJ, Nieuwenhuis EE et al, 2002, Oxazolone colitis, a Th2 colitis model resembling ulcerative colitis, is mediated by IL-13-producing NK-T cells. Immunity 17:629–638 Pastorelli L, Garg RR, Hoang SB et al, 2010, Epithelial-derived IL-33 and its receptor ST2 are dysregulated in ulcerative colitis and in experimental Th1/Th2 driven enteritis. Proc Natl Acad Sci U S 107:8017–8022 Levy DE, Lee CK, 2002, What does Stat3 do? J Clin Invest 109:1143–1148 Mudter J, Weigmann B, Bartsch B et al, 2005, Activation pattern of signal transducer and activators of transcription, STAT, factors in inflammatory bowel diseases. Am J Gastroenterol 100:64–72 Shull MM, Ormsby I, Kier AB et al, 1992, Targeted disruption of the mouse transforming growth factor-beta 1 gene results in multifocal inflammatory disease. Nature 359:693–699 Babyatsky MW, Rossiter G, Podolsky DK, 1996, Expression of transforming growth factors alpha and beta in colonic mucosa in inflammatory bowel disease. Gastroenterology 110:975–984 Yamaguchi T, Ihara K, Matsumoto T et al, 2001, Inflammatory bowel disease-like colitis in glycogen storage disease type 1b. Inflamm Bowel Dis 7:128–132 Weaver CT, Hatton RD, Mangan PR et al, 2007, IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu Rev Immunol 25:851–852 Kastelein RA, Hunter CA, Cua DJ, 2007, Discovery and biology of IL-23 and IL-27: related but functionally distinct regulators of inflammation. Annu Rev Immunol 25:221–242 Ivanov II, McKenzie BS, Zhou L et al, 2006, The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126:1121–1133 Langrish CL, Chen Y, Blumenschein WM et al, 2005, IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 201:233–240 Zhou L, Lopes JE, Chong MM et al, 2008, TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature 453:236–240 Shen W, Durum SK, 2010, Synergy of IL-23 and Th17 cytokines: new light on inflammatory bowel disease. Neurochem Res 35:940–946 Bettelli E, Oukka M, Kuchroo VK, 2007, TH-17 cells in the circle of immunity and autoimmunity. Nat Immunol 8:345–350 Kinugasa T, Sakaguchi T, Gu X et al, 2000, Claudins regulate the intestinal barrier in response to immune mediators. Gastroenterology 118:1001–1011 Rioux JD, Xavier RJ, Taylor KD et al, 2007, Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat Genet 39:596–604 Ellson CD, Davidson K, Ferguson GJ et al, 2006, Neutrophils from p40phox2/2 mice exhibit severe defects in NADPH oxidase regulation and oxidant-dependent bacterial killing. J Exp Med 203:1927–1937 Ogawa M, Yoshimori T, Suzuki T, Sagara H et al, 2004, Escape of intracellular Shigella from autophagy. Science 307:727–731 Lakatos PL, Szamosi T, Szilvasi A et al, 2008, ATG16L1 and IL23 receptor, IL23R, genes are associated with disease susceptibility in Hungarian CD patients. Dig Liver Dis 40:867–873 Hampe J, Franke A, Rosenstiel P et al, 2007, A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat Genet 39:207– 211 Parkes M, Barrett JC, Prescott NJ et al, 2007, Sequence variants in the autophagy gene IRGM and multiple other replicating loci contribute to Crohn’s disease susceptibility. Nat Genet 39:830–832 Singh SB, Davis AS, Taylor G et al, 2006, IRGM induces autophagy to eliminate intracellular mycobacteria. Science 313:1438–1441 Taylor GA, 2007, IRG proteins: key mediators of interferonregulated host resistance to intracellular pathogens. Cell Microbiol 9:1099–1107 Makitalo L, Kolho KL, Karikoski R et al, 2010, Expression profiles of matrix metalloproteinases and their inhibitors in colonic inflammation related to pediatric inflammatory bowel disease. Scand J Gastroenterol 45:862–871 Salmi M, Andrew DP, Butcher EC et al, 1995, Dual binding capacity of mucosal immunoblasts to mucosal and synovial endothelium in humans: dissection of the molecular mechanisms. J Exp Med 181:137–149 Bernstein CN, Sargent M, Rector E, 2002, Alteration in expression of beta 2 integrins on lamina propria lymphocytes in ulcerative colitis and Crohn’s disease. Clin Immunol 104:67–72 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 463 EP 472 DI 10.1007/s12253-011-9397-4 PG 10 ER PT J AU You, Q Wang, Xsh Fu, Sb Jin, Xm AF You, Qi Wang, Xi-shan Fu, Song-bin Jin, Xiao-ming TI Downregulated Expression of Inhibitor of Growth 4 (ING4) in Advanced Colorectal Cancers: A Non-Randomized Experimental Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal carcinoma; Tumor suppressor gene; ING4; Immunohistochemistry ID Colorectal carcinoma; Tumor suppressor gene; ING4; Immunohistochemistry AB Colorectal cancer has a high cure rate if it can be detected early. Identifying and understanding the genes involved may enable early diagnosis and reduce mortality. The aim of our study was to investigate the correlation between the expression of ING4 and the pathological features in patients with colorectal cancer. We assayed ING4 protein expression levels in tumor samples from 97 patients diagnosed with colorectal cancer between January 2001 and January 2002. The patients received no other treatments except surgery. ING4 protein expression was downregulated in adenoma relative to normal mucosa and further reduced in colorectal cancer tissues. Furthermore, the suppression of ING4 expression was also related to the more advanced Dukes’ stages. We observed that ING4 expression levels in patients with lymphatic metastasis were lower than those without metastasis. Together, our results indicate that ING4 play a role in colorectal carcinoma progression. C1 [You, Qi] The Tumor Hospital of Harbin Medical University, Department of Colorectal Surgery, 150040 Harbin, Heilongjiang Province, China. [Wang, Xi-shan] The Tumor Hospital of Harbin Medical University, Department of Colorectal Surgery, 150040 Harbin, Heilongjiang Province, China. [Fu, Song-bin] The Tumor Hospital of Harbin Medical University, Department of Colorectal Surgery, 150040 Harbin, Heilongjiang Province, China. [Jin, Xiao-ming] The Tumor Hospital of Harbin Medical University, Department of Colorectal Surgery, 150040 Harbin, Heilongjiang Province, China. RP Wang, Xsh (reprint author), The Tumor Hospital of Harbin Medical University, Department of Colorectal Surgery, 150040 Harbin, China. EM wxshan1208@yahoo.com.cn CR United States Cancer Statistics, USCS), 1999–2005 Cancer Incidence and Mortality Data, 2007; http://apps.nccd.cdc.gov/ uscs/ Hawk ET, Levin B, 2005, Colorectal cancer prevention. J Clin Oncol 23:378–391 Cheah PY, 1990, Hypotheses for the etiology of colorectal cancer— an overview. Nutr Cancer 14:5–13 Lichtenstein P et al, 2000, Environmental and heritable factors in the causation of cancer—analyses of cohorts of twins from Sweden, Denmark and Finland. N Eng J Med 343:78–85 Zhien M, 1995, Expression and clinical significance of gene mutation in human colon tumor tissues. Chin J Onc 17:332–334 Kim S, Chin K, Gray JW, Bishop JM, 2004, A screen for genes that suppress loss of contact inhibition: identification of ING4 as a candidate tumor suppressor gene in human cancer. Proc Natl Acad Sci USA 101:16251–16256 Shiseki M et al, 2003, p29ING4 and p28ING5 bind to p53 and p300, and enhance p53 activity. Cancer Res 63:2373–2378 Shen JC et al, 2007, Inhibitor of growth 4 suppresses cell spreading and cell migration by interacting with a novel binding partner, liprin alpha1. Cancer Res 67:2552–2558 Zhang X et al, 2004, ING4 induces G2/M cell cycle arrest and enhances the chemosensitivity to DNA-damage agents in HepG2 cells. FEBS Lett 570:7–12 Li X et al, 2008, ING4 induces cell growth inhibition in human lung adenocarcinoma A549 cells by means of Wnt-1/beta-catenin signaling pathway. Anat Rec 291:593–600 Garkavtsev I et al, 2004, The candidate tumour suppressor protein ING4 regulates brain tumour growth and angiogenesis. Nature 428:328–332 Ozer A, Wu LC, Bruick RK, 2005, The candidate tumor suppressor ING4 represses activation of the hypoxia inducible factor, HIF). Proc Natl Acad Sci USA 102:7481–7486 Kim S, 2005, HuntING4 new tumor suppressors. Cell Cycle 4:516–517 Xu L, Li T, 2007, The effect of ING4 gene in tumor progression. Int J Genetics 30:147–149 Aasland R, Gibson TJ, Stewart AF, 1995, The PHD finger: implications for chromatin-mediated transcriptional regulation. Trends Biochem Sci 20:56–59 Campos EI, ChinMY, KuoWH, Li G, 2004, Biological functions of the ING family tumor suppressors. Cell Mol Life Sci 61:2597–2613 Gunduz M et al, 2005, Frequent deletion and down-regulation of ING4, a candidate tumor suppressor gene at 12p13, in head and neck squamous cell carcinomas. Gene 356:109–117 Zhang X et al, 2005, Nuclear localization signal of ING4 plays a key role in its binding to p53. Biochem Biophys Res Commun 331:1032–1038 Brat DJ, Bellail AC, VanMeir EG, 2005, The role of interleukin 8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro Oncol 7:122–133 Ozer A, Bruick RK, 2005, Regulation of HIF by prolyl hydroxylases: recruitment of the candidate tumor suppressor protein ING4. Cell Cycle 4:1153–1156 Colla S et al, 2007, The new tumor-suppressor gene inhibitor of growth family member 4, ING4, regulates the production of proangiogenic molecules by myeloma cells and suppresses hypoxia-inducible factor-1 alpha, HIF-1 alpha, activity: involvement in myeloma induced angiogenesis. Blood 110:4464–4475 Doyon Y et al, 2006, ING tumor suppressor proteins are critical regulators of chromatin acetylation required for genome expression and perpetuation. Mol Cell 21:51–64 Hassler M et al, 2006, Diversity of cytogenetic and pathohistologic profiles in glioblastoma. Cancer Genet Cytogenet 166:46–55 Liu L, Chang H, Yang JC, 2007, Candidate tumor suppressor gene ING4: research progress. Chin J Mod Drug Appl 1:69–72 Li J, Martinka M, Li G, 2008, Role of ING4 in human melanoma cell migration, invasion and patient survival. Carcinogenesis 29:1373–1379 Yu JK, Yang MQ, Jiang TJ, Zheng S, 2004, The optimal combination of serum tumor markers with bioinformatics in diagnosis of colorectal carcinoma. Zhejiang Da Xue Xue Bao Yi Xue Ban 33(5):407–410 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 473 EP 477 DI 10.1007/s12253-010-9301-7 PG 5 ER PT J AU Li, N Wang, J Shen, Sh Bu, X Tian, X Huang, P AF Li, Nan Wang, Jingmei Shen, Shanshan Bu, Xiaodong Tian, Xiaoqiang Huang, Peilin TI Expression of p53, Ki-67 and c-Myc Proteins is Predictive of the Surgical Molecular Margin in Colorectal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal carcinoma; Surgical molecular margin; p53; Ki-67; c-Myc ID Colorectal carcinoma; Surgical molecular margin; p53; Ki-67; c-Myc AB Surgical resection is the mainstay of treatment for colorectal carcinoma, however, the overall survival is modest due to frequent local recurrence from residual cancer cells after "curative" resection. Therefore, the status of surgical margin (tumor free or positive) has a significant influence on patient’s survival. The difference in molecular profile between mucosa neighboring tumor lesions and remote area (surgical margin) may aid in evaluating resection status. 44 colorectal tumor tissues with corresponding adjacent non-neoplastic mucosa (within 3 cm from tumor tissues), and 110 tumor tissues with corresponding surgical margin mucosa (5 cm from tumor tissues) were randomly collected, fixed in 10% formalin and followed by embedding in paraffin. And the expression of p53, Ki-67 and c-Myc were investigated by tissue microarray (TMA) and immunohistochmistry. The expression of p53, Ki-67 and c-Myc were decreased in both adjacent non-neoplastic mucosa and mucosa of surgical margin, comparing to their expression in corresponding cancer cells. Furthermore, the expression of these proteins in mucosa of remote area (surgical margin) was significantly lower than those adjacent to tumor lesions. The expression of p53, Ki-67 and c-Myc in mucosa can be used as molecular marker for assessing surgical margin status in colorectal carcinoma. C1 [Li, Nan] Affiliated Drum Tower Hospital of Nanjing University Medical School, Department of Pathology, Dingjia Qiao. 87, 210019 Nanjing, China. [Wang, Jingmei] Affiliated Drum Tower Hospital of Nanjing University Medical School, Department of PathologyNanjing, China. [Shen, Shanshan] Affiliated Drum Tower Hospital of Nanjing University Medical School, Department of Pathology, Dingjia Qiao. 87, 210019 Nanjing, China. [Bu, Xiaodong] Affiliated Drum Tower Hospital of Nanjing University Medical School, Department of Pathology, Dingjia Qiao. 87, 210019 Nanjing, China. [Tian, Xiaoqiang] Affiliated Drum Tower Hospital of Nanjing University Medical School, Department of Pathology, Dingjia Qiao. 87, 210019 Nanjing, China. [Huang, Peilin] Affiliated Drum Tower Hospital of Nanjing University Medical School, Department of Pathology, Dingjia Qiao. 87, 210019 Nanjing, China. RP Huang, P (reprint author), Affiliated Drum Tower Hospital of Nanjing University Medical School, Department of Pathology, 210019 Nanjing, China. EM hplwpp@yahoo.cn CR Jemal A, Siegel R, Ward E et al, 2008, Cancer statistics. CA Cancer J Clin 58:71–96 Braakhuis BJ, Bloemena E, Leemans CR et al, 2010, Molecular analysis of surgical margins in head and neck cancer: More than a marginal issue. Oral Oncol 46:485–491 Wood LD, Parsons DW, Jones S et al, 2007, The genomic landscapes of human breast and colorectal cancers. Science 318:1108–1113 Kikuchi Y, Dinjens WN, Bosman FT, 1997, Proliferation and apoptosis in proliferative lesions of the colon and rectum. Virchows Arch 431:111–117 Kononen J, Bubendor L, Kallioniemi A et al, 1998, Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4:844–847 Pacifico MD, Grover R, Richman P et al, 2004, Validation of tissue microarray for the immunohistochemical profiling of melanoma. Melanoma Res 14:39–42 Zlobec I, Holler S, Tornillo L et al, 2009, Combined histomorphologic and immunohistochemical phenotype to predict the presence of vascular invasion in colon cancer. Dis Colon Rectum 52:1114–1121 Kammerer U, Kapp M, Gassel AM et al, 2001, A new rapid immunohistochemical staining technique using the EnVision antibody complex. J Histochem Cytochem 49:623–630 Kamath A, Helie M, Bifulco CB et al, 2009, Lack of immunohistochemical detection of VEGF in prostate carcinoma. Appl Immunohistochem Mol Morphol 17:227–232 Fu CG, Tominaga O, Nagawa H et al, 1998, Role of p53 and p21/WAF1 detection in patient selection for preoperative radiotherapy in rectal cancer patients. Dis Colon Rectum 41:68–74 Bateman AC, Carr NJ, Warren BF, 2005, The retroperitoneal surface in distal caecal and proximal ascending colon carcinoma: the Cinderella surgical margin? J Clin Pathol 58:426– 428 Goldenberg D, Harden S, Masayesva BG et al, 2004, Intraoperative molecular margin analysis in head and neck cancer. Arch Otolaryngol Head Neck Surg 130:39–44 van Houten VM, Leemans CR, Kummer JA et al, 2004, Molecular diagnosis of surgical margins and local recurrence in head and neck cancer patients: a prospective study. Clin Cancer Res 10:3614–3620 Masasyesva BG, Tong BC, Brock MV et al, 2005, Molecular margin analysis predicts local recurrence after sublobar resection of lung cancer. Int J Cancer 113:1022–1025 Yang B, Gao YT, Du Z et al, 2005, Methylation-based molecular margin analysis in hepatocellular carcinoma. Biochem Biophys Res Commun 338:1353–1358 Kane MF, Loda M, Gaida GM et al, 1997, Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic colon tumors and mismatch repair-defective human tumor cell lines. Cancer Res 57:808–811 Murad JC, Ribeiro U Jr, Safatle-Ribeiro AVet al, 2007, Evaluation of molecular markers in hepatic metastasis of colorectal adenocarcinoma. Hepatogastroenterology 54:1029–1033 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 479 EP 487 DI 10.1007/s12253-010-9323-1 PG 9 ER PT J AU Kadivar, M Monabati, A Joulaee, A Hosseini, N AF Kadivar, Maryam Monabati, Ahmad Joulaee, Azadeh Hosseini, Niloufar TI Epstein-Barr Virus and Breast Cancer: Lack of Evidence for an Association in Iranian Women SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Epstein–Barr virus (EBV); Breast carcinoma; Iran; Immunohistochemistry (IHC); Polymerase chain reaction (PCR) ID Epstein–Barr virus (EBV); Breast carcinoma; Iran; Immunohistochemistry (IHC); Polymerase chain reaction (PCR) AB Controversies regarding the role of Epstein-Barr virus (EBV) in breast cancer and lack of published literature in this regard in Iran, prompted us to assess EBV presence in 100 breast carcinoma and 42 control biopsies obtained from Iranian women. Breast carcinoma cases were comprised of 81 invasive ductal carcinoma NOS, 9 invasive lobular carcinoma, 1 apocrine carcinoma, 2 cribriform carcinoma, 2 papillary carcinoma and 5 mucinous carcinoma. Control biopsies consisted of 13 fibroadenoma, 9 benign epithelial proliferation (adenosis and sclerosing adenosis), 9 usual ductal hyperplasia, 4 atypical ductal hyperplasia, 4 non-proliferative fibrocystic changes and 3 normal breast tissue. To identify EBV-infected cells we applied immunohistochemical analysis, using monoclonal antibody against Epstein-Barr virusencoded nuclear antigen 2 (EBNA-2) and latent membrane protein 1 (LMP-1). Further, polymerase chain reaction (PCR) was used to amplify EBV DNA, with primers that cover the EBV encoded RNA (EBER) and BamHIW regions. EBNA-2 and LMP-1 immunohistochemistry were negative in all breast cancer and control specimens. Using PCR, none of the 100 breast cancer samples or the 42 control specimens showed detectable EBV DNA. These results indicate that EBV may not play a significant role in the etiology of breast cancer in Iranian women. C1 [Kadivar, Maryam] Tehran University of Medical Sciences, Hazrat Rasool-e-Akram Hospital, Department of Pathology, Niayesh St, Sattarkhan AveTehran, Iran. [Monabati, Ahmad] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. [Joulaee, Azadeh] Shahid Beheshti University of Medical Sciences, Department of Breast & General surgeryTehran, Iran. [Hosseini, Niloufar] Shahid Beheshti University of Medical SciencesTehran, Iran. RP Kadivar, M (reprint author), Tehran University of Medical Sciences, Hazrat Rasool-e-Akram Hospital, Department of Pathology, Tehran, Iran. EM dmkadivar@gmail.com CR Labrecque LG, Barnes DM, Fentiman IS et al, 1995, Epstein-Barr virus in epithelial cell tumors: a breast cancer study. Cancer Res 55:39–45 Parkin DM, Bray FI, Devesa SS, 2001, Cancer burden in the year 2000. The global picture. Eur J Cancer 37(Suppl 8):54–66 He J-R, Song Er-Wei, Ren Ze-Fang, 2009, Research advancement on relationship between Epstein-Barr virus and breast cancer. Chin J Cancer 28(8):1–6 Fawzy S, Sallam M, Awad NM, 2008, Detection of Epstein-Barr virus in breast carcinoma in Egyptian women. Clin Biochem 41, 7–8):486–492 Joshi D, Quadri M, Gangane N et al, 2009, Association of Epstein-Barr virus infection with breast cancer in rural Indian women. PLoS One 4 4(12):e8180 Kallkan A, Ozdarendeli A, Bulut Y et al, 2005, Investigation of Epstein-Barr virus DNA in formalin-fixed and paraffin-embedded breast cancer tissues. Med Princ Pract 14(4):268–271 Cox B, Richardson A, Graham P et al, 2010, Breast cancer, cytomegalovirus and Epstein-Barr virus: a nested case-control study. Br J Cancer 102(11):1665–1669 Bonnet M, Guinebretiere J, Kremmer E et al, 1999, Detection of EBV DNA by polymerase chain reaction in invasive breast cancers. J Natl Cancer Inst 91:1376–1381 Saito I, Servenius B, Compton T et al, 1989, Detection of EBV DNA by polymerase chain reaction in blood and tissue biopsies from patients with Sjogren’s syndrome. J Exp Med 169:2191– 2198 Horiuchi K, Mishima K, Ohsawa M et al, 1994, Carcinoma of stomach and breast with lymphoid stroma: localisation of Epstein-Barr virus. J Clin Pathol 47:538–540 Luqmani YA, Shousha S, 1995, Presence of Epstein-Barr virus in breast carcinoma. Int J Oncol 6:899–903 Brink AA, van den Brule AJ, van Diest P et al, 2000, Detection of Epstein-Barr virus in invasive breast cancers. J Natl Cancer Inst 92:655–656 Fina F, Romain S, Ouafik L et al, 2001, Frequency and genome load of Epstein-Barr virus in 509 breast cancers from different geographical areas. Br J Cancer 84:783–790 Grinstein S, Preciado MV, Gattuso P et al, 2002, Demonstration of Epstein-Barr virus in carcinomas of various sites. Cancer Res 62:4876–4878 Gaffey MJ, Frierson HF, Mills SE et al, 1993, Medullary carcinoma of breast: identification of lymphocyte subpopulations and their significance. Mod Pathol 6:721–728 Lespagnard L, Cochaux P, Larsimont D et al, 1995, Absence of Epstein-Barr virus in medullary carcinoma of the breast as demonstrated by immunophenotyping, in situ hybridization and polymerase chain reaction. Am J Clin Pathol 103:449–452 Chu JS, Chen CC, Chang KJ, 1998, In situ detection of Epstien-Barr virus in breast cancer. Cancer Lett 124:53–57 Arbach H, Viglasky V, Lefeu F et al, 2006, Epstein-Barr virus, EBV, genome and expression in breast cancer tissue: effect of EBV infection of breast cancer cells on resistance to paclitaxel, Taxol). J Virol 80:845–853 Perkins RS, Sahm K, Marando C et al, 2006, Analysis of Epstein-Barr virus reservoirs in paired blood and breast cancer primary biopsy specimens by real time PCR. Breast Cancer Res 8(6):R70 Glaser SL, Hsu JL, Gulley ML, 2004, Epstein-Barr Virus and Breast Cancer: State of the Evidence for Viral Carcinogenesis. Cancer Epidemiol Biomark Prev 13:688–697 McCall SA, Lichy JH, Bijwaard KE et al, 2001, Epstein-Barr virus detection in ductal carcinoma of the breast. J Natl Cancer Inst 93:148–150 Deshpande CG, Badve S, Kidwai N et al, 2002, Lack of expression of the Epstein-Barr Virus, EBV, gene products, EBERs, EBNA1, LMP1, and LMP2A, in breast cancer cells. Lab Invest 82:1193–1199 Chu PG, Chang KL, Chen YY et al, 2001, No significant association of Epstein-Barr virus infection with invasive breast carcinoma. Am J Pathol 159:571–578 Chang KL, Albujar PF, Chen YY et al, 1993, High prevalence of Epstain-Barr virus in the reed-sternberg cells of Hodgkin’s disease occurring in Peru. Blood 81:496–501 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 489 EP 492 DI 10.1007/s12253-010-9325-z PG 4 ER PT J AU Kim, SN Kang, YJ Jo, JO Kim, YH Oh, RY Kim, YO Jung, HM Ock, SM Cha, HJ AF Kim, Soo Nam Kang, Yun-Jeong Jo, Jin-Ok Kim, Yeol Heung Oh, Rim Young Kim, Young-Ok Jung, Hyung Min Ock, Sun Mee Cha, Hee-Jae TI Elevated Expression of Thymosin β4, Vascular Endothelial Growth Factor (VEGF), and Hypoxia Inducible Factor (HIF)-1α in Early-Stage Cervical Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; Thymosin β4; VEGF; HIF-1α; Angiogenesis ID Cervical cancer; Thymosin β4; VEGF; HIF-1α; Angiogenesis AB Recent studies have shown that thymosin β4 (TB-4) is highly related with tumor metastasis and angiogenesis. In addition, TB-4 induced the expression of VEGF in melanoma cells. We investigated the expression patterns of TB-4 and related angiogenic proteins, VEGF, and HIF-1α, at various stages of cervical cancers and also identified the expression pattern of these proteins in metastatic cervical cancers. Expression patterns of TB-4, VEGF, and HIF-1α were studied with tissue microarray containing 42 samples of cervical cancers. In addition, 15 cervical cancers and metastatic tumors in lymph nodes from patients who have metastatic tumors were also analyzed to confirm the role of TB-4, VEGF, and HIF -1α in cervical cancer metastasis. The expression levels of TB-4, VEGF, and HIF-1α were very weak at early cancer stages (stages 0 to 1A) but significantly increased at stage 1B. The numbers of blood vessels in tumors were also increased at stage 1B. The expression patterns of TB-4, VEGF, and HIF-1α were compared in tumors without lymph node metastasis, primary tumors with lymph node metastasis, and metastatic tumors in lymph nodes. The expression levels of TB-4, VEGF, and HIF-1α in primary tumors with lymph node metastasis and their metastatic tumors in lymph node were less than in tumors without lymph node metastasis. These data suggest that TB-4, VEGF, and HIF-1α triggered angiogensis and tumor invasiveness to surrounding tissues at early stage of cervical carcinoma but have a negative or no effect on the metastatic potential. C1 [Kim, Soo Nam] Kosin University College of Medicine, Department of Obstetrics and GynecologyBusan, South Korea. [Kang, Yun-Jeong] Kosin University College of Medicine, Department of Parasitology and Genetics, 34, Annam-dong, Seo-gu, 602-703 Busan, South Korea. [Jo, Jin-Ok] Kosin University College of Medicine, Department of Parasitology and Genetics, 34, Annam-dong, Seo-gu, 602-703 Busan, South Korea. [Kim, Yeol Heung] Kosin University College of Medicine, Department of Obstetrics and GynecologyBusan, South Korea. [Oh, Rim Young] Kosin University College of Medicine, Department of Obstetrics and GynecologyBusan, South Korea. [Kim, Young-Ok] Kosin University College of Medicine, Department of PathologyBusan, South Korea. [Jung, Hyung Min] Kyung Hee University, College of Medicine, Department of Obstetrics and GynecologySeoul, South Korea. [Ock, Sun Mee] Kosin University College of Medicine, Department of Parasitology and Genetics, 34, Annam-dong, Seo-gu, 602-703 Busan, South Korea. [Cha, Hee-Jae] Kosin University College of Medicine, Department of Parasitology and Genetics, 34, Annam-dong, Seo-gu, 602-703 Busan, South Korea. RP Cha, HJ (reprint author), Kosin University College of Medicine, Department of Parasitology and Genetics, 602-703 Busan, South Korea. EM hcha@kosin.ac.kr CR Low TL, Hu SK, Goldstein AL, 1981, Complete amino acid sequence of bovine thymosin beta 4: a thymic hormone that induces deoxynucleotidy l transferase activity in thymocyte populations. Proc Natl Acad Sci USA 78:1162–1166 Low TL, Goldstein AL, 1985, Thymic hormones: an overview. Methods Enzymol 116:213–219 Safer D, Nachmias VT, 1994, Beta thymosins as actin binding peptides. BioEssays 16:473–479 Yu FX, Lin SC, Morrison-Bogorad M et al, 1993, Thymosin beta 10 and thymosin beta 4 are both actin monomer sequestering proteins. J Biol Chem 268:502–509 Yu FX, Lin SC, Morrison-Bogorad M et al, 1994, Effects of thymosin beta 4 and thymosin beta 10 on actin structures in living cells. Cell Motil Cytoskeleton 27:13–25 Grant DS, Rose W, Yaen C et al, 1999, Thymosin beta 4 enhances endothelial cell differentiation and angiogenesis. Angiogenesis 3:125–135 Philp D, Goldstein AL, Kleinman HK, 2004, Thymosin beta 4 promotes angiogenesis, wound healing and hair follicle development. Mech Ageing Dev 125:113–115 Philp D, St-Surin S, Cha HJ et al, 2007, Thymosin beta 4 induces hair growth via stem cell migration and differentiation. Ann NY Acad Sci 1112:95–103 Iguchi K, Usami Y, Hirano K et al, 1999, Decreased thymosin beta 4 in apoptosis induced by a variety of antitumor drugs. Biochem Pharmacol 57:1105–1111 Niu M, Nachmias VT, 2007, Increased resistance to apoptosis in cells overexpressing thymosin beta four: a role for focal adhesion kinase pp 125FAK. Cell Adhes Commun 7:311–320 Young JD, Lawrence AJ, MacLean AG et al, 1999, Thymosin beta 4 sulfoxide is an anti-inflammatory agent generated by monocytes in the presence of glucocorticoids. Nat Med 5:1424– 1427 Cha HJ, Jeong MJ, Kleinman HK, 2003, Role of thymosin beta 4 in tumor metastasis and angiogenesis. J Natl Cancer Inst 95:1674– 1680 Kobayashi T, Okada F, Fujii N et al, 2002, Thymosin-beta4 regulates motility and metastasis of malignant mouse fibrosarcoma cells. Am J Pathol 160:869–882 Wang WS, Chen PM, Hsiao HL et al, 2004, Overexpression of the thymosin beta-4 gene is associated with increased invasion of SW480 colon carcinoma cells and the distant metastasis of human colorectal carcinoma. Oncogene 23:6666–6671 Huang HC, Hu CH, Tang MC et al, 2007, Thymosin beta 4 triggers an epithelial-mesenchymal transition in colorectal carcinoma by upregulating integrin-linked kinase. Oncogene 26:2781– 2790 Larsson LI, Holck S, 2007, Localization of thymosin beta-4 in tumors. Ann NY Acad Sci 1112:317–325 Larsson LI, Holck S, 2007, Occurrence of thymosin beta 4 in human breast cancer cells and in other cell types of the tumor microenvironment. Hum Pathol 38:114–119 Ji P, Diederichs S, Wang W et al, 2003, MALAT-1, a novel noncoding RNA, and thymosin beta 4 predict metastasis and survival at early-stage non-small cell lung cancer. Oncogene 22:8031–8041 Jia YT, Li ZX, Liu M et al, 2009, Endostar reduces the growth and metastasis by inhibiting angiogenesis and lymphangiogenesis in nude mouse models of human cervical cancer. Zhonghua Zhong Liu Za Zhi 31:254–257 Willmott LJ, Monk BJ, 2009, Cervical cancer therapy: current, future and anti-angiogensis targeted treatment. Expert Rev Anticancer Ther 9:895–903 Clark EA, Golub TR, Lander ES et al, 2000, Genomic analysis of metastasis reveals an essential role for RhoC. Nature 406:532–535 Otto AM, Mqller CSG, Huff T et al, 2002, Chemotherapeutic drugs change actin skeleton organization and the expression of betathymosins in human breast cancer cells. J Cancer Res Clin Oncol 128:247–256 Yamamoto T, Gotoh M, Kitajima M et al, 1993, Thymosin beta-4 expression is correlated with metastatic capacity of colorectal carcinomas. Biochem Biophys Res Commun 193:706– 710 Monk BJ, Willmott LJ, Sumner DA, 2010, Anti-angiogenesis agents in metastatic or recurrent cervical cancer. Gynecol Oncol 116:181–186 Goncharuk IV, Vorobjova LI, Lukyanova NYet al, 2009, Vascular endothelial growth factor exression in uterine cervical cancer: correlation with clinicopathologic characteristics and survival. Exp Oncol 31:179–181 No JH, Jo H, Kim SH et al, 2009, Expression of vascular endothelial growth factor and hypoxia inducible factor-1alpha in cervical neoplasia. Ann NY Acad Sci 1171:105–110 Sosne G, Szliter EA, Barrett R et al, 2002, Thymosin beta 4 promotes corneal wound healing and decreases inflammation in vivo following alkali injury. Exp Eye Res 74:293–299 Sosne G, Qiu P, Christopherson PL et al, 2007, Thymosin beta 4 suppression of corneal NFkappaB: a potential anti-inflammatory pathway. Exp Eye Res 48:663–669 Badamchian M, Fagarasan MO, Danne RL et al, 2003, Thymosin beta 4 reduces lethality and downregulates inflammatorymediators in endotoxin-induced septic shock. Int Immunopharmacol 3:1225–1233 Falk V, Lundgren N, Quarfordt L et al, 1982, Primary surgical treatment of carcinoma stage I of the uterine cervix. Acta Obstet Gynecol Scand 61:481–486 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 493 EP 502 DI 10.1007/s12253-010-9327-x PG 10 ER PT J AU Toth, K Galamb, O Spisak, S Wichmann, B Sipos, F Valcz, G Leiszter, K Molnar, B Tulassay, Zs AF Toth, Kinga Galamb, Orsolya Spisak, Sandor Wichmann, Barnabas Sipos, Ferenc Valcz, Gabor Leiszter, Katalin Molnar, Bela Tulassay, Zsolt TI The Influence of Methylated Septin 9 Gene on RNA and Protein Level in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; mRNA expression; Peripheral blood; Protein expression; Septin 9 ID Colorectal cancer; mRNA expression; Peripheral blood; Protein expression; Septin 9 AB Colorectal cancer is one of the leading death causes in the world. Specificity and sensitivity of the present screening methods are unsuitable and their compliance is too low. Nowadays the most effective method is the colonoscopy, because it gives not only macroscopic diagnosis but therapeutic possibility as well, however the compliance of the patients is very low. Hence development of new diagnostic methods is needed. Altered expression of septin 9 was found in several tumor types including colorectal cancer. The aim of this study was to detect the methylation related mRNA and protein expression changes of septin 9 in colorectal adenomadysplasia-carcinoma sequence and to analyze its reversibility by demethylation treatment. Septin 9 protein expression showed significant difference between normal and colorectal cancer (CRC) samples (p<0,001). According to biopsy microarray results, septin 9 mRNA expression decreased in the progression of colon neoplastic disease (p<0,001). In laser microdissected epithelial cells, septin 9 significantly underexpressed in CRC compared to healthy controls (p<0,001). The expression of septin9_v1 region was higher in the healthy samples, while septin9_v2, v4, v4*, v5 overexpression were detected in cancer epithelial cells compared to normal. The septin 9 mRNA and protein levels of HT29 cells increased after demethylation treatment. The increasing methylation of septin 9 gene during colorectal adenomadysplasia-carcinoma sequence progression is reflected in the decreasing mRNA and protein expression, especially in the epithelium. These changes can be reversed by demethylation agents converting this screening marker gene into therapeutic target. C1 [Toth, Kinga] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Galamb, Orsolya] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Spisak, Sandor] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Wichmann, Barnabas] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Valcz, Gabor] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Leiszter, Katalin] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. RP Toth, K (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM drtothkinga@yahoo.com CR Boyle P, Ferlay J, 2005, Cancer incidence and mortality in Europe, 2004. Ann Oncol 16:481–488 Jemal A, Murray T, Ward E et al, 2005, Cancer statistics, 2005. CA Cancer J Clin 55:10–30 Ebert MP, Model F, Mooney S et al, 2006, Aristaless-like homeobox-4 gene methylation is a potential marker for colorectal adenocarcinomas. Gastroenterology 131:1418–1430 Lofton-Day C, Model F, Devos T et al, 2008, DNA methylation biomarkers for blood-based colorectal cancer screening. Clin Chem 54:414–423 Grutzmann R, Molnar B, Pilarsky C et al, 2008, Sensitive detection of colorectal cancer in peripheral blood by septin 9 DNA methylation assay. PLoS ONE 3:e3759 Devos T, Tetzner R, Model F et al, 2009, Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem 55:1337–1346 Scott M, Hyland PL, McGregor G et al, 2005, Multimodality expression profiling shows SEPT9 to be overexpressed in a wide range of human tumours. Oncogene 24:4588–4700 Galamb O, Gyorffy B, Sipos F, 2008, Inflammation, adenoma and cancer: objective classification of colon biopsy specimens with gene expression signature. Dis Markers 25:1–16 Hall PA, Russell SE, 2004, The pathobiology of the septin gene family. J Pathol 204:489–505 Russell SE, McIlhatton MA, Burrows JF, 2000, Isolation and mapping of a human septin gene to a region on chromosome 17q, commonly deleted in sporadic epithelial ovarian tumors. Cancer Res 60:4729–4734 Kalikin LM, Sims HL, Petty EM, 2000, Genomic and expression analyses of alternatively spliced transcripts of the MLL septin-like fusion gene, MSF, that map to a 17q25 region of loss in breast and ovarian tumors. Genomics 63:165–172 Burrows JF, Chanduloy S, McIlhatton MA et al, 2003, Altered expression of the septin gene, SEPT9, in ovarian neoplasia. J Pathol 201:581–588 Montagna C, LyuMS, Hunter K et al, 2003, The Septin 9, MSF, gene is amplified and overexpressed in mouse mammary gland adenocarcinomas and human breast cancer cell lines. Cancer Res 63:2179–2187 Tatsumi K, Taki T, Taniwaki M et al, 2001, The CDCREL1 gene fused to MLL in de novo acute myeloid leukemia with t(11;22,, q23;q11.2, and its frequent expression in myeloid leukemia cell lines. Genes Chromosom Cancer 30:230–235 Kim HJ, Ki CS, Park Q et al, 2003, MLL/SEPTIN6 chimeric transcript from invins(X;11)(q24;q23q13, in acute monocytic leukemia: report of a case and review of the literature. Genes Chromosom Cancer 38:8–12 Kojima K, Sakai I, Hasegawa A et al, 2004, FLJ10849, a septin family gene, fuses MLL in a novel leukemia cell line CNLBC1 derived from chronic neutrophilic leukemia in transformation with t(4;11)(q21;q23). Leukemia 18:998–1005 Tanaka M, Tanaka T, Matsuzaki S et al, 2003, Rapid and quantitative detection of human septin family Bradeion as a practical diagnostic method of colorectal and urologic cancers. Med Sci Monit 9:61–68 Kim DS, Hubbard SL, Peraud A et al, 2004, Analysis of mammalian septin expression in human malignant brain tumors. Neoplasia 6:168–178 Model F, Osborn N, Ahlquist D et al, 2007, Identification and validation of colorectal neoplasia-specific methylation markers for accurate classification of disease. Mol Cancer Res 5:153–163 Toth K, Galamb O, Spisak S et al, 2009, Free circulating DNA based colorectal cancer screening from peripheral blood: the possibility of the methylated septin 9 gene marker. Orv Hetil 150:969–977 Scott M, McCluggage WG, Hillan KJ et al, 2006, Altered patterns of transcription of the septin 9 gene, SEPT9, in ovarian tumorigenesis. Int J Cancer 118:1325–1329 Montagna C, Lyu MS, Hunter K, 2003, The Septin 9, MSF, gene is amplified and overexpressed in mouse mammary gland adenocarcinomas and human breast cancer cell lines. Cancer Res 63:2179–2187 Gonzalez ME, Peterson EA, Privette LM, 2007, High SEPT9_v1 expression in human breast cancer cells is associated with oncogenic phenotypes. Cancer Res 67:8554–8564 Liu ZJ, Wang G, Cai Y, 2009, Androgen receptor CpG island methylation status in human leukemia cancer cells. Cancer Investig 27:156–162 Yap OW, Bhat G, Liu L et al, 2009, Epigenetic modifications of the Estrogen receptor beta gene in epithelial ovarian cancer cells. Anticancer Res 29:139–144 Deng G, Kakar S, Okudiara K et al, 2009, Unique methylation pattern of oncostatin m receptor gene in cancers of colorectum and other digestive organs. Clin Cancer Res 15:1519–1526 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 503 EP 509 DI 10.1007/s12253-010-9338-7 PG 7 ER PT J AU Halon, A Materna, V Drag-Zalesinska, M Nowak-Markwitz, E Gansukh, T Donizy, P Spaczynski, M Zabel, M Dietel, M Lage, H Surowiak, P AF Halon, Agnieszka Materna, Verena Drag-Zalesinska, Malgorzata Nowak-Markwitz, Ewa Gansukh, Tserenchunt Donizy, Piotr Spaczynski, Marek Zabel, Maciej Dietel, Manfred Lage, Hermann Surowiak, Pawel TI Estrogen Receptor Alpha Expression in Ovarian Cancer Predicts Longer Overall Survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Estrogen receptor alpha; Ovarian cancer; Immunohistochemistry; Cisplatin ID Estrogen receptor alpha; Ovarian cancer; Immunohistochemistry; Cisplatin AB Estrogen as a potential factor of ovarian carcinogenesis, acts via two nuclear receptors, estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ), but the cellular signal pathways involved are not completely clear so far. In this study we have described the expression of ERα, detected by immunocytochemistry in 11 ovarian carcinoma cell lines and by immunohistochemistry in 43 Federation Internationale des Gyneacologistes et Obstetristes stage III ovarian carcinoma specimens prepared before and after treatment with cisplatin-based schemes. For cisplatin resistance is a major obstacle in the treatment of ovarian carcinoma, analysis of cisplatin sensitivity in 11 ovarian carcinoma cell line was also performed. The strong nuclear ERα expression was only shown in the single A2780P cell line. Expression of ERα in tissue specimens did not reveal any correlations between histopathological parameters (histologic type and grading). We demonstrated a significant association with ERα expression in specimens from primary laparotomies (PL) and cause–specific survival. In the cases terminated by death of the patient, overall immunoreactivity score of ERα expression at PL was significantly lower than in surviving patients. In addition, Kaplan-Meier analysis revealed significantly shorter overall survival time and progression-free time in cases with lower immunoreactivity score of ERα expression at PL. Our findings support the hypothesis that aberrant hormone activity, by way of altered receptor expression, might be an important factor in the malignant transformation of ovarian cancer. C1 [Halon, Agnieszka] Wroclaw Medical University, Department of Pathology, ul. Marcinkowskiego 1, 50-367 Wroclaw, Poland. [Materna, Verena] Charite University Hospital, Institute of Pathology, Chariteplatz. 1 20/21, 10117 Berlin, Germany. [Drag-Zalesinska, Malgorzata] Wroclaw Medical University, Department of Human Morphology and Embryology, ul. Chalubinskiego 6a, 50-356 Wroclaw, Poland. [Nowak-Markwitz, Ewa] Poznan University of Medical Sciences, Cathedral of Mother’s and Child’s Health, ul. Polna 33, 60-535 Poznan, Poland. [Gansukh, Tserenchunt] Medical Research Institute of Mongolia, Bayangol duureg, Ard-Ayush Street-1, 210526 Ulan Bator, Mongolia. [Donizy, Piotr] Wroclaw Medical University, Department of Pathology, ul. Marcinkowskiego 1, 50-367 Wroclaw, Poland. [Spaczynski, Marek] Poznan University of Medical Sciences, Cathedral of Mother’s and Child’s Health, ul. Polna 33, 60-535 Poznan, Poland. [Zabel, Maciej] Wroclaw Medical University, Department of Human Morphology and Embryology, ul. Chalubinskiego 6a, 50-356 Wroclaw, Poland. [Dietel, Manfred] Charite University Hospital, Institute of Pathology, Chariteplatz. 1 20/21, 10117 Berlin, Germany. [Lage, Hermann] Charite University Hospital, Institute of Pathology, Chariteplatz. 1 20/21, 10117 Berlin, Germany. [Surowiak, Pawel] Charite University Hospital, Institute of Pathology, Chariteplatz. 1 20/21, 10117 Berlin, Germany. RP Halon, A (reprint author), Wroclaw Medical University, Department of Pathology, 50-367 Wroclaw, Poland. EM ahalon2@gmail.com CR Lazannec G, 2006, Estrogen receptor beta, a possible tumor suppressor involved in ovarian carcinogenesis. Cancer Lett 231:151–157 Pearce ST, Jordan VC, 2004, The biological role of estrogen receptors α and β in cancer. Crit Rev Oncol Hematol 50:3–22 Lindgren PR, Cajander S, Backstrom T et al, 2004, Estrogen and progesterone receptors in ovarian epithelial tumors. Mol Cell Endocrinol 221:97–104 Pujol P, Rey JM, Nirde P et al, 1998, Differential expression of estrogen receptor-alpha and -beta messenger RNAs as a potential marker of ovarian carcinogenesis. Cancer Res 58:5367–5373 Rutheford T, Brown WD, Sapi E et al, 2000, Absence of estrogen receptor-beta expression in metastatic ovarian cancer. Obstet Gynecol 96:417–421 Cunat S, Hoffmann P, Pujol P, 2004, Estrogens and epithelial ovarian cancer. Gynecol Oncol 94:25–32 Hiller SG, Anderson RA, Williams AR et al, 1998, Expression of oestrogen receptor alpha and beta in cultured human ovarian surface epithelial cells. Mol Hum Reprod 4:811–815 Brandenberger AW, Tee MK, Jaffe RB, 1998, Estrogen receptor alpha, ER-alpha, and beta, ER-beta, mRNAs in normal ovary, ovarian serous cystadenocarcinoma and ovarian cancer cell lines: down-regulation of ER-beta in neoplastic tissues. J Clin Endocrinol Metab 83:1025–1028 Kurebayashi J, 2003, Endocrine-resistant breast cancer: underlying mechanisms and strategies for overcoming resistance. Breast Cancer 10:112–119 Harries M, Kaye SB, 2001, Recent advances in the treatment of epithelial ovarian cancer. Expert Opin Investig Drugs 10:1715–1724 McClay EF, Albright KD, Jones JA et al, 1994, Tamoxifen delays the development of resistance to cisplatin in human melanoma and ovarian cancer cell lines. Br J Cancer 70:449–452 Materna V, Liedert B, Homale J et al, 2005, Protection of platinum-DNA adduct formation and reversal of cisplatin resistance by anti-MRP2 hammerhead ribozymes in human cancer cells. Int J Cancer 115:393–402 Materna V, Stege A, Surowiak P et al, 2006, RNA interferencetriggered reversal of ABCC2-dependent cisplatin resistance in human cancer cells. Biochem Biophys Res Commun 348:153–157 Kowalski P, Surowiak P, Lage H, 2005, Reversal of different drug-resistant phenotypes by an autocatalytic multitarget multiribozyme directed against the transcripts of the ABC transporters MDR1/P-gp, MRP2, and BCRP. Mol Ther 11:508–522 Surowiak P, Materna V, Kaplenko I et al, 2006, ABCC2, MRP2, cMOAT, can be localized in the nuclear membrane of ovarian carcinomas and correlates with resistance to cisplatin and clinical outcome. Clin Cancer Res 12:7149–7158 Gyorffy B, Surowiak P, Kiesslich O et al, 2006, Resistance prediction profile for eleven anticancer agents at clinical concentrations based on the gene expression pattern of thirty cell lines. Int J Cancer 118:1699–1712 Sobin LH, Wittekind Ch, eds,, 1997, UICC TNM Classification of malignant tumors. Wiley-Liss, New York Shimizu Y, Kamoi S, Amada S et al, 1998, Toward the development of a universal grading system of ovarian epithelial carcinoma: Testing of a proposed system in a series of 461 patients with uniform treatment and follow-up. Cancer 82:893– 901 Remmele W, Stenger HE, 1987, Recommendation for uniform definition of an immunoreactive score, IRS, for immunohistochemical estrogen receptor detection, ER-ICA, in breast cancer tissue. Pathologe 8:138–140 Fitzgibbons PL, Page DL, Weaver D et al, 2000, Prognostic factors in breast cancer. college of american pathologists consensus statement 1999. Arch Pathol Lab Med 124:966–978 Ogawa Y, Moriya T, Kato Y et al, 2004, Immunohistochemical assessment for estrogen receptor and progesterone receptor status in breast cancer: analysis for a cut-off point as the predictor for endocrine therapy. Breast Cancer 11:267–275 Burges A, Bruning A, Dannenmann C et al, 2010, Prognostic significance of estrogen receptor alpha and beta expression in human serous carcinomas of the ovary. Arch Gynecol Obstet 281:511–517 Lee P, Rosen DG, Zhu C et al, 2005, Expression of progesterone receptor is favorable prognostic marker in ovarian cancer. Gynecol Oncol 96:671–677 Li AJ, Baldwin RL, Karlan BY, 2003, Estrogen and progesterone receptor subtype expression in normal and malignant ovarian epithelial cell cultures. Am J Obstet Gynecol 189:22–27 Hecht JL, Kotsopoulos J, Hankinson SE et al, 2009, Relationship between epidemiological risk and hormone receptor expression in ovarian cancer: results from the Nurses’ Health Study. Cancer Epidemiol Biomark Prev 18:1–14 Sakaguchi H, Fujimoto J, Aoki I et al, 2002, Expression of oestrogen receptor a and b in uterine endometrial and ovarian cancers. Eur J Cancer 38:S74–S75 Chao KC, Wang PH, Yen MS et al, 2005, Role of estrogen and progesterone in the survival of ovarian tumors—a study of the human ovarian adenocarcinoma cell line OC-117-VGH. J Chin Med Assoc 68:360–367 Park SH, Cheung LW, Wong AS et al, 2008, Estrogen regulates Snail and Slug in the down-regulation of E-cadherin and induces metastatic potential of ovarian cancer cells through estrogen receptor α. Mol Endocrinol 22:2085–2098 Therasse P, Arbuck SG, Eisenhauer EA et al, 2000, New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 92:205–216 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 511 EP 518 DI 10.1007/s12253-010-9340-0 PG 8 ER PT J AU Fernandez-Acenero, JM Cordova, Sh Manzarbeitia, F Medina, C AF Fernandez-Acenero, Jesus Maria Cordova, Sharon Manzarbeitia, Felix Medina, Camino TI Immunohistochemical Profile of Urothelial and Small Cell Carcinomas of the Bladder SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Urothelial cell carcinoma; Small cell carcinoma; Neuroendocrine tumors; TTF-1; Immunohistochemistry ID Urothelial cell carcinoma; Small cell carcinoma; Neuroendocrine tumors; TTF-1; Immunohistochemistry AB Small cell carcinoma of the bladder is an uncommon and rather aggressive bladder tumor, representing less than 1% of all vesical tumors. Small cell carcinoma of different organs has been shown to express markers of neuroendocrine differentiation, and also thyroid transcription factor 1 (TTF-1). TTF-1 is a transcription factor and its expression has been shown mainly in pulmonary small cell carcinomas and adenocarcinomas and in thyroid tumors. Although it was initially proposed as a useful marker to delineate the origin of metastatic adenocarcinomas from the lung, its expression is being increasingly reported in tumors from different origins. The goal of this review is to analyse the immunohistochemical profile of small cell carcinoma of the bladder and to compare it to classical urothelial cell carcinomas. With this aim we have reviewed the small cell bladder carcinomas diagnosed in a single tertiary hospital in Madrid (Fundacion Jimenez Diaz) in the last 12 years. We have found 6 pure small cell carcinomas and performed a wide panel of immunohistochemistry, including cytokeratins 7 and 20, enolase, chromogranin, synaptophysin, CD56 and TTF-1 to these tumors and also to 30 high grade urothelial cell carcinomas of usual type. Only one of our small cell carcinoma cases showed positivity for TTF-1, while five expressed CD56 and four neuron-specific enolase. None of our cases expressed cytokeratin 20 or 7. To our surprise we found a case of conventional urothelial cell carcinoma expressing focally TTF-1. These results are in accordance with the current literature, although our rate of TTF-1 expression (16.6%) is on the low end of the spectrum. C1 [Fernandez-Acenero, Jesus Maria] University Hospital Fundacion Jimenez Diaz, Department of PathologyMadrid, Spain. [Cordova, Sharon] University Hospital Fundacion Jimenez Diaz, Department of PathologyMadrid, Spain. [Manzarbeitia, Felix] University Hospital Fundacion Jimenez Diaz, Department of PathologyMadrid, Spain. [Medina, Camino] University Hospital Fundacion Jimenez Diaz, Department of PathologyMadrid, Spain. RP Fernandez-Acenero, JM (reprint author), University Hospital Fundacion Jimenez Diaz, Department of Pathology, Madrid, Spain. EM mgg10167@gmail.com CR Blomjous CE, Vos W, De Voogt HJ, Van der Valk P, 1989, Small cell carcinoma of the urinary bladder. A clinicopathological, morphometric, immunohistochemical and ultrastructural series of 18 cases. Cancer 64:1347–1357 Lopez JL, Angulo JC, Flores N, 1994, Small cell carcinoma of the urinary bladder. A clinicopathological study of six cases. Br J Urol 73:43–49 Agoff SN, Lamps LW, Philip AT, Amin MB, Schmidt RA, True LD, Folpe AL, 2000, Thyroid transcription factor-1 is expressed in extrapulmonary small cell carcinomas but not in other extrapulmonary neuroendocrine tumors. Mod Pathol 13:238–242 Cheng L, Jones TD, McCarthy RP, Eble JN, Wang M, MacLennan GT, Lopez-Beltran A, Yang XJ, Koch MO, Zhang S, Pan CX, Baldridge LA, 2005, Molecular genetic evidence for a common clonal origin of urinary bladder small cell carcinoma and coexisting urothelial carcinoma. Am J Pathol 166:1533–1539 Jones TD, Kernek KM, Yang XJ, Lopez-Beltran A, MacLennan GT, Eble JN, Lin H, Pan CX, Tretiakova M, Baldridge LA, Cheng L, 2005, Thyroid transcription factor 1 expression in small cell carcinoma of the urinary bladder: an immunohistochemical profile of 44 cases. Hum Pathol 36:718–723 Alijo Serrano F, Sanchez-Mora N, Angel Arranz J, Hernandez C, Alvarez-Fernandez E, 2007, Large cell and small cell neuroendocrine bladder carcinoma: immunohistochemical and outcome study in a single institution. Am J Clin Pathol 128:733–739 Mackey JR, Au HJ, Hugh J, Venner P, 1998, Genitourinary small cell carcinoma: determination of clinical and therapeutic factor associated with survival. J Urol 159:1624–1629 Cheng L, Pan CX, Yang XJ, Lopez-Beltran A, MacLennan GT, Lin H, Kuzel TM, Papavero V, Tretiakova M, Nigro K, Koch MO, Eble JN, 2004, Small cell carcinoma of the urinary bladder: a clinicopathologic analysis of 64 patients. Cancer 101:957–962 Abbosh PH, Wang M, Eble JN, Lopez-Beltran A, Maclennan GT, Montironi R, Zheng S, Pan CX, Zhou H, Cheng L, 2008, Hypermethylation of tumor-suppressor gene CpG islands in smallcell carcinoma of the urinary bladder. Mod Pathol 21:355–362 Wang X, Zhang S, MacLennan GT, Eble JN, Lopez-Beltran A, Yang XJ, Pan CX, Zhou H, Montironi R, Cheng L, 2007, Epidermal growth factor receptor protein expression and gene amplification in small cell carcinoma of the urinary bladder. Clin Cancer Res 13:953–957 Shin SJ, DeLellis RA, Ying L, Rosen PP, 2000, Small cell carcinoma of the breast. A clinicopathological and immunohistochemical study of nine patients. Am J Surg Pathol 24:1231–1238 Choi SJ, Kim JM, Han JY, Ahn SI, Kim JS, Kim L, Park IS, Chu YC, 2007, Extrapulmonary small cell carcinoma of the liver: clinicopathological and immunohistochemical findings. Yonsei Med J 48:1066–1071 Ordonez NG, 2000, Value of thyroid transcription factor-1 immunostaining in distinguishing small cell lung carcinomas from other small cell carcinomas. Am J Surg Pathol 24:1217– 1223 Cheuk W, Kwan MY, Suster S, Chan JK, 2001, Immunostaining for thyroid transcription factor 1 and cytokeratin 20 aids the detection of small cell carcinoma from Merkel cell carcinoma, but not pulmonary from extrapulmonary small cell carcinomas. Arch Pathol Lab Med 125:228–231 Siami K, McCluggage WG, Ordonez NG, Euscher ED, Malpica A, Sneige N, Silva EG, Deavers MT, 2007, Thyroid transcription factor-1 expression in endometrial and endocervical adenocarcinomas. Am J Surg Pathol 31:1759–1763 Lee WJ, Kim CH, Chang SE, Lee MW, Choi JH, Moon KC, Koh JK, 2009, Cutaneous metastasis from large-cell neuroendocrine carcinoma of the urinary bladder expressing CK20 and TTF-1. Am J Dermatopathol 31:166–169 Byrd-Gloster AL, Khoor A, Glass LF, Messina JL, Whitsett JA, Livingston SK, Cagle PT, 2000, Differential expression of thyroid transcription factor 1 in small cell lung carcinoma and Merkel cell tumor. Hum Pathol 31:58–62 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 519 EP 523 DI 10.1007/s12253-010-9341-z PG 5 ER PT J AU Fotovati, A Abu-Ali, S Kage, M Shirouzu, K Yamana, H Kuwano, M AF Fotovati, Abbas Abu-Ali, Samah Kage, Masayoshi Shirouzu, Kazuo Yamana, Hideaki Kuwano, Michihiko TI N-myc Downstream-regulated Gene 1 (NDRG1) a Differentiation Marker of Human Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Differentiation; Cancer biomarkers; NDRG1; Cap43; Anticancer differentiation therapy ID Breast cancer; Differentiation; Cancer biomarkers; NDRG1; Cap43; Anticancer differentiation therapy AB N-myc downstream-regulated gene 1 (NDRG1), also called differentiation-related gene-1 (Drg1) and Cap43, is expressed in various normal tissues and suppressed in several malignancies. In this study, whether NDRG1 expression was correlated with differentiation of human breast cancer cells has been investigated. Endogenous expression level of NDRG1 was closely correlated with differentiation status of breast cancer cell lines. Furthermore, sodium butyrate (NaB), an inducer of cellular differentiation, increased the expression of β-casein, a milk-related differentiation marker, together with upregulation of NDRG1 in breast cancer cells. In contrast, inhibition of NDRG1 by its siRNA resulted in reduced accumulation of β-casein. Immunohistochemical analysis showed co-expression of NDRG1 and β-casein or milk fat protein (MFP), another differentiation marker of breast tissue, in the mouse xenograft model of breast cancer. Furthermore, overexpression of NDRG1 expanded the differentiated area in the xenograft model of breast cancer. In human breast cancer, using samples from 45 patients, we also showed close relationship between NDRG1 and β-casein or MFP expression. Altogether, in vitro and in vivo data demonstrated a possible role of NDRG1 in differentiation of breast cancer. We concluded that NDRG1 could be used as a biomarker for differentiation of breast cancer for both diagnostic and therapeutic purposes. C1 [Fotovati, Abbas] Kurume University, Institute of Cancer Innovative Therapy, Laboratory of Molecular SurgeryKurume, Japan. [Abu-Ali, Samah] JOYUP Bio-Medicals, Laboratory of Biomedical Research, Nishi-Ogi-Minami Suginami-kuTokyo, Japan. [Kage, Masayoshi] Kurume University, Institute of Cancer Innovative Therapy, Laboratory of Molecular SurgeryKurume, Japan. [Shirouzu, Kazuo] Kurume University, School of Medicine, Kurume University Hospital, Department of SurgeryKurume, Japan. [Yamana, Hideaki] Kurume University, School of Medicine, Kurume University Hospital, Department of SurgeryKurume, Japan. [Kuwano, Michihiko] Kyushu University, Graduate School of Pharmaceutical Sciences, Department of PharmaceuticsFukuoka, Japan. RP Fotovati, A (reprint author), Kurume University, Institute of Cancer Innovative Therapy, Laboratory of Molecular Surgery, Kurume, Japan. CR Zhou D, Salnikow K, Costa M, 1998, Cap43, a novel gene specifically induced by ni2+ compounds. Cancer Res 58, 10):2182–2189 van Belzen N, Dinjens WN, Diesveld MP, Groen NA, van der Made AC, Nozawa Y, Vlietstra R, Trapman J, Bosman FT, 1997, A novel gene which is up-regulated during colon epithelial cell differentiation and down-regulated in colorectal neoplasms. Lab Invest 77(1):85–92 Shimono A, Okuda T, Kondoh H, 1999, N-myc-dependent repression of ndr1, a gene identified by direct subtraction of whole mouse embryo cdnas between wild type and n-myc mutant. Mech Dev 83(1–2):39–52 Kurdistani SK, Arizti P, Reimer CL, Sugrue MM, Aaronson SA, Lee SW, 1998, Inhibition of tumor cell growth by rtp/rit42 and its responsiveness to p53 and DNA damage. Cancer Res 58, 19):4439–4444 Salnikow K, Kluz T, Costa M, 1999, Role of ca(2+, in the regulation of nickel-inducible cap43 gene expression. Toxicol Appl Pharmacol 160(2):127–132 Masuda K, Ono M, Okamoto M, Morikawa W, Otsubo M, Migita T, Tsuneyoshi M, Okuda H, Shuin T, Naito S, Kuwano M, 2003, Downregulation of cap43 gene by von hippel-lindau tumor suppressor protein in human renal cancer cells. Int J Cancer 105, 6):803–810 Park H, Adams MA, Lachat P, Bosman F, Pang SC, Graham CH, 2000, Hypoxia induces the expression of a 43-kda protein, proxy-1, in normal and malignant cells. Biochem Biophys Res Commun 276(1):321–328 Nishie A, Masuda K, Otsubo M, Migita T, Tsuneyoshi M, Kohno K, Shuin T, Naito S, Ono M, Kuwano M, 2001, High expression of the cap43 gene in infiltrating macrophages of human renal cell carcinomas. Clin Cancer Res 7(7):2145–2151 Okuda H, Hirai S, Takaki Y, Kamada M, Baba M, Sakai N, Kishida T, Kaneko S, Yao M, Ohno S, Shuin T, 1999, Direct interaction of the beta-domain of vhl tumor suppressor protein with the regulatory domain of atypical pkc isotypes. Biochem Biophys Res Commun 263(2):491–497 Lachat P, Shaw P, Gebhard S, van Belzen N, Chaubert P, Bosman FT, 2002, Expression of ndrg1, a differentiation-related gene, in human tissues. Histochem Cell Biol 118(5):399–408 Wakisaka Y, Furuta A, Masuda K, Morikawa W, Kuwano M, Iwaki T, 2003, Cellular distribution of ndrg1 protein in the rat kidney and brain during normal postnatal development. J Histochem Cytochem 51(11):1515–1525 Okuda T, Higashi Y, Kokame K, Tanaka C, Kondoh H, Miyata T, 2004, Ndrg1-deficient mice exhibit a progressive demyelinating disorder of peripheral nerves. Mol Cell Biol 24(9):3949–3956 Hirata K, Masuda K, Morikawa W, He JW, Kuraoka A, Kuwano M, Kawabuchi M, 2004, N-myc downstream-regulated gene 1 expression in injured sciatic nerves. Glia 47(4):325–334 Guan RJ, Ford HL, FuY, LiY, ShawLM, PardeeAB, 2000, Drg-1 as a differentiation-related, putative metastatic suppressor gene in human colon cancer. Cancer Res 60(3):749–755 Bandyopadhyay S, Pai SK, Gross SC, Hirota S, Hosobe S, Miura K, Saito K, Commes T, Hayashi S, Watabe M, Watabe K, 2003, The drg-1 gene suppresses tumor metastasis in prostate cancer. Cancer Res 63(8):1731–1736 Bandyopadhyay S, Pai SK, Hirota S, Hosobe S, Takano Y, Saito K, Piquemal D, Commes T, Watabe M, Gross SC, Wang Y, Ran S, Watabe K, 2004, Role of the putative tumor metastasis suppressor gene drg-1 in breast cancer progression. Oncogene 23(33):5675–5681 Fotovati A, Fujii T, Yamaguchi M, Kage M, Shirouzu K, Oie S, Basaki Y, Ono M, Yamana H, Kuwano M, 2006, 17beta-estradiol induces down-regulation of cap43/ndrg1/drg-1, a putative differentiation-related and metastasis suppressor gene, in human breast cancer cells. Clin Cancer Res 12(10):3010–3018 Ozpolat B, 2009, Resistance to differentiation therapy. In: Mehta K and Siddik ZH, eds, Drug resistance in cancer cells, 1st edn. Springer, New York Leszczyniecka M, Roberts T, Dent P, Grant S, Fisher PB, 2001, Differentiation therapy of human cancer: Basic science and clinical applications. Pharmacol Ther 90(2–3):105–156 Breitman TR, Selonick SE, Collins SJ, 1980, Induction of differentiation of the human promyelocytic leukemia cell line, hl-60, by retinoic acid. Proc Natl Acad Sci U S A 77(5):2936–2940 Huberman E, Callaham MF, 1979, Induction of terminal differentiation in human promyelocytic leukemia cells by tumorpromoting agents. Proc Natl Acad Sci U S A 76(3):1293–1297 Lane AA, Chabner BA, 2009, Histone deacetylase inhibitors in cancer therapy. J Clin Oncol 27(32):5459–5468 Piquemal D, Joulia D, Balaguer P, Basset A, Marti J, Commes T, 1999, Differential expression of the rtp/drg1/ndr1 gene product in proliferating and growth arrested cells. Biochim Biophys Acta 1450(3):364–373 Munshi A, Kurland JF, Nishikawa T, Tanaka T, Hobbs ML, Tucker SL, Ismail S, Stevens C, Meyn RE, 2005, Histone deacetylase inhibitors radiosensitize human melanoma cells by suppressing DNA repair activity. Clin Cancer Res 11(13):4912– 4922 Gore SD, Weng LJ, Zhai S, Figg WD, Donehower RC, Dover GJ, Grever M, Griffin CA, Grochow LB, Rowinsky EK, Zabalena Y, Hawkins AL, Burks K, Miller CB, 2001, Impact of the putative differentiating agent sodium phenylbutyrate on myelodysplastic syndromes and acute myeloid leukemia. Clin Cancer Res 7, 8):2330–2339 Santini V, Gozzini A, Scappini B, Grossi A, Rossi Ferrini P, 2001, Searching for the magic bullet against cancer: the butyrate saga. Leuk Lymphoma 42(3):275–289 Warrell RP Jr, He LZ, Richon V, Calleja E, Pandolfi PP, 1998, Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. J Natl Cancer Inst 90(21):1621–1625 Gozzini A, Rovida E, Dello Sbarba P, Galimberti S, Santini V, 2003, Butyrates, as a single drug, induce histone acetylation and granulocytic maturation: possible selectivity on core binding factor-acute myeloid leukemia blasts. Cancer Res 63(24):8955– 8961 Maruyama Y, OnoM, Kawahara A, Yokoyama T, Basaki Y, KageM, Aoyagi S, Kinoshita H, Kuwano M, 2006, Tumor growth suppression in pancreatic cancer by a putative metastasis suppressor gene cap43/ndrg1/drg-1 through modulation of angiogenesis. Cancer Res 66(12):6233–6242 Russo J, Mailo D, Hu YF, Balogh G, Sheriff F, Russo IH, 2005, Breast differentiation and its implication in cancer prevention. Clin Cancer Res 11(2 Pt 2):931s–936s Russo J, Hu YF, Silva ID, Russo IH, 2001, Cancer risk related to mammary gland structure and development. Microsc Res Tech 52, 2):204–223 CG RJ, Russo IH, 1993, Critical approach to the malignant transformation of human breast epithelial cells. CRC Crit Rev Oncog 4:403–417 Russo J, Gusterson BA, Rogers AE, Russo IH, Wellings SR, van Zwieten MJ, 1990, Comparative study of human and rat mammary tumorigenesis. Lab Invest 62(3):244–278 Kouros-Mehr H, Bechis SK, Slorach EM, Littlepage LE, Egeblad M, Ewald AJ, Pai SY, Ho IC, Werb Z, 2008, Gata-3 links tumor differentiation and dissemination in a luminal breast cancer model. Cancer Cell 13(2):141–152 van Belzen N, Dinjens WN, Eussen BH, Bosman FT, 1998, Expression of differentiation-related genes in colorectal cancer: possible implications for prognosis. Histol Histopathol 13, 4):1233–1242 Visscher DW, Zarbo RJ, Greenawald KA, Crissman JD, 1990, Prognostic significance of morphological parameters and flow cytometric DNA analysis in carcinoma of the breast. Pathol Annu 25(Pt 1):171–210 Page DL, 1991, Prognosis and breast cancer. Recognition of lethal and favorable prognostic types. Am J Surg Pathol 15, 4):334–349 Shah MA, Kortmansky J, Motwani M, Drobnjak M, Gonen M, Yi S, Weyerbacher A, Cordon-Cardo C, Lefkowitz R, Brenner B, O’Reilly E, Saltz L, Tong W, Kelsen DP, Schwartz GK, 2005, A phase i clinical trial of the sequential combination of irinotecan followed by flavopiridol. Clin Cancer Res 11(10):3836–3845 Marcha JA, Boulaiz H, Peran M, Prados JC, Campos J, Gonzalez FJ, Rodriguez-Serrano F, Melguizo C, Velez C, Carrillo E, Hita F, Ortiz R, Martinez-Amat A, Caba O, Ventura C, Aranega A, 2009, Cancer stem cells and differentiation therapy. In: Macrcha JA et al, ed, Therapeutic potential of differentiation in cancer and normal stem cells, 1st edn. Nova Science Publishers Inc, New York NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 525 EP 533 DI 10.1007/s12253-010-9342-y PG 9 ER PT J AU Jankovic Velickovic, L Dolicanin, Z Hattori, T Pesic, I Djordjevic, B Stojanovic, M Stankovic, J Visnic, M Stefanovic, V AF Jankovic Velickovic, Ljubinka Dolicanin, Zana Hattori, Takanori Pesic, Ivana Djordjevic, Biljana Stojanovic, Mariola Stankovic, Jablan Visnic, Milan Stefanovic, Vladisav TI Divergent Squamous Differentiation in Upper Urothelial Carcinoma—Comparative Clinicopathological and Molecular Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Divergent differentiation; Squamous metaplasia; Molecular markers; Proliferative activity; Upper urothelial carcinoma ID Divergent differentiation; Squamous metaplasia; Molecular markers; Proliferative activity; Upper urothelial carcinoma AB Upper urothelial carcinoma (UUC) has a plasticity to demonstrate divergent differentiation with squamous metaplastic elements. There was no previous study exploring profiling of molecular markers in metaplastic squamous upper urothelial carcinoma (SUUC) and conventional upper urothelial carcinoma (CUUC). The aims of this study was to compare expression of the phenotypic characteristics of tumors and molecular markers (p53, p16, cyclin D1, E-cadherin, HER-2, Ki-67, Bcl-2, Bax) in SUUC and CUUC. SUUC was detected in 20% of 44 patients. There was significant difference between SUUC and CUUC in the pathological stage, grade, growth and presence of lymphovasular invasion (p<0.05; 0.05; 0.05; 0.01 respectively). The mean Ki-67 and p53 labeling index was significantly higher in SUUC than in CUUC (p<0.05; 0.05). There was no significant difference in the expression of p16, cyclin D1, E-cadherin, HER-2, Bcl-2 and Bax between SUUC and CUUC. Univariant model showed that SUUC was significantly associated with lymphovascular invasion (p=0.007), Ki-67 activity (p=0.016) and growth (p=0.026). Exploration of UUC with squamous divergent differentiation showed changes in phenotypic characteristics and Ki-67, as well as similar molecular profile with CUUC. C1 [Jankovic Velickovic, Ljubinka] Faculty of Medicine, Institute of PathologyNis, Serbia. [Dolicanin, Zana] Faculty of Medicine, Institute of PathologyNis, Serbia. [Hattori, Takanori] Shiga University of Medical Science, Department of PathologyOhtsu, Japan. [Pesic, Ivana] Faculty of Medicine, Institute of PathophysiologyNis, Serbia. [Djordjevic, Biljana] Faculty of Medicine, Institute of PathologyNis, Serbia. [Stojanovic, Mariola] Public Health Institute, Department of StatisticsNis, Serbia. [Stankovic, Jablan] Faculty of Medicine, Clinic of SurgeryNis, Serbia. [Visnic, Milan] Faculty of Medicine, Clinic of SurgeryNis, Serbia. [Stefanovic, Vladisav] Faculty of Medicine, Institute of Nephrology, Bul. Zorana Djindjica 81, 18000 Nis, Serbia. RP Stefanovic, V (reprint author), Faculty of Medicine, Institute of Nephrology, 18000 Nis, Serbia. EM stefan@ni.ac.rs CR Shanks JH, Iczkowski KA, 2009, Divergent differentiation in urothelial carcinoma and other bladder cancer subtypes with selected mimics. Histopathology 54:885–900 Nigwekar P, Amin BM, 2008, The many faces of urothelial carcinoma. An update with an emphasis on recently described variants. Adv Anat Pathol 15:218–233 Sung TM, Wang M, MacLennan TG, Eble NJ, Tan P-H, Lopez- Beltran A, Montironi R, Harris JJ, Kuhar M, Cheng L, 2007, Histogenesis of sarcomatoid urothelial carcinoma of the urinary bladder: evidence for a common clonal origin with divergent differentiation. J Pathol 211:420–430 Amstrong BA, Wang M, Eble NJ, MacLennan TG, Montironi R, Tan P-H, Lopez-Beltran A, Zhang S, Baldridge AL, Spartz H, Cheng L, 2009, TP53 mutational analysis supports monoclonal origin of biphasic sarcomatoid urothelial carcinoma, carcinosarcoma, of the urinary bladder. Mod Path 22:113–118 Martin EJ, Jenkins JB, Zuk JR, Blandy PJ, Baithun IS, 1989, Clinical importance of squamous metaplasia in invasive transitional cell carcinoma of the bladder. J Clin Pathol 42:250–253 Perez-Montiel D,Wakely EP,HesO, MichalM,Suster S, 2006)Highgrade urothelial carcinoma of the renal pelvis: clinicopathologic study of 108 cases with emphasis on unusual morphologic variants. Mod Path 19:494–503 Young HR, Zukerberg R, 1991, Microcystic transitional cell carcinoma of the urinary bladder. Am J Clin Pathol 96:635–639 Lopez-Beltran A, Sauter G, Gasser T, Hartmann A, Schmitz- Drager BJ, Helpap B, Ayala AG, Tamboli P, Knowles MA, Sidransky D, Cordon-Cardo C, Jones PA, Cairns P, Simon R, Amin MB, Tyezinski JE, 2004, Tumors of the urinary system. Infiltrating urothelial carcinoma. In: Eble JN, Sauter G, Epstein JI, Sesterhenn IA, eds, World Health Organization classification of tumours. Pathology and genetics. Tumours of the urinary system and male genital organs. IARC, Lyon, pp 93–109 Sobin LH, Wittekind C, eds,, 2002, TNM classification of malignant tumors, 6th edn. Wiley-Liss, New York Genega EM, Kapali M, Torres-Quinones M, Huang WC, Knauss JS, Wang LP, Raghunath PN, Kozlowski C, Malkowicz SB, Tomaszewski JE, 2005, Impact of the 1998 World health organization/international society of urological pathology classification system for urothelial neoplasms of the kidney. Mod Pathol 18:11–18 Benedict FW, Lerner PS, Zhou J, Shen X, Tokunaga H, Czerniak B, 1999, Level of retinoblastoma protein expression correlates with p16, MTS-1/INK4A/CDKN2, status in bladder cancer. Oncogene 18:1197–1203 Shariat SF, Tokunaga H, Zhou J, Kim J, Ayala GE, Benedict WF, Lerner SP, 2004, P53, p21, pRB, and p16 expression predict clinical outcome in cystectomy with bladder cancer. J Clin Oncol 22:1014–1024 Genega ME, Porter RC, 2002, Urothelial neoplasms of the kidney and ureter. Am J Clin Pathol 117:S36–S48 Jung I, Messing E, 2000, Molecular mechanisms and pathways in bladder cancer development and progression. Cancer Control 7:325–334 Amin BM, 2009, Histological variants of urothelial carcinoma: diagnostic, therapeutic and prognostic implications. Histological variants of urothelial carcinoma. Mod Path 22:S96–S118 Black CP, Brown AG, Dinney PC, 2009, The impact of variant histology on the outcome of bladder cancer treated with curative intent. Urol Oncol 27:3–7 Lopez-Beltran A, Sauter G, Gasser T et al, 1988, Squamous and glandular differentiation in urothelial bladder carcinomas. Histopathology, histochemistry and immunohistochemical expression of carcinoembryonic anrigen. Histol Histopathol 3:63–68 Lopez-Beltran A, Requena JM, Alvarez-Kindelan J, Quintero A, Blanca A, Montironi R, 2007, Squamous differentiation in primary urothelial carcinoma of the urinary tract as seen by MAC387 immunohistochemistry. J Clin Pathol 60:332–335 Kamijima S, Tobe T, Suyama T, Ueda T, Igarashi T, Ichikawa T, Ito H, 2005, The prognostic value of p53, Ki-67 and matrix metalloproteinases MMP-2 and MMP-9 in transitional cell carcinoma of the renal pelvis and ureter. Int J Urol 12:941–947 Fromont G, Roupret M, Amira N, Sibony M, Vallancien G, Validire P, Cussenot O, 2005, Tissue microarray analysis of the prognostic value of E-cadherin, Ki-67, p53, p27, survivin and MSH2 expression in upper urinary tract transitional cell carcinoma. Eur Urol 48:764–770 El-kott AF, 2007, Flow cytometry and Ki-67 expression in rat’s urinary bladder carcinogenesis treated with Allium Sativum. Cancer Ther 5:185–192 Eltz S, Comperat E, Cussenot O, Roupret M, 2008, Molecular and histological markers in urothelial carcinoma of the upper urinary tract. BJUI 102:532–535 Kunju LP, Lee CT, Montie J, Shah RB, 2005, Utility of cytokeratin 20 and Ki-67 as markers of urothelial dysplasia. Pathol Int 55:248–254 Shamsdin SA, Mehrabani D, Hosseinzadeh M, 2008, Expression of Ki-67, C-erbB2 and EGFR in TCC of the urinary bladder and their correlation with tumor grading. Iranian Red Crescent Med J 10:95–99 Aaltonen V, Koivunen J, Laato M, Peltonen J, 2006, Heterogeneity of cellular proliferation within transitional cell carcinoma: correlation of protein kinase C alpha/betal expression and activity. J Histochem Cytochem 54:795–806 Quintero A, Alvarez-Kindelan J, Luque RJ, Gonzales-Campora R, Requena MJ, Montironi R, Lopez-Beltran A, 2006, Ki-67 MIB1 labelling index and the prognosis of primary TaT1 urothelial cell carcinoma of the bladder. J Clin Pathol 59:83–88 Xue-Ru Wu, 2005, Urothelial tumorigenesis: a tale of divergent pathways. Nat Rev Cancer 5:713–725 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 535 EP 539 DI 10.1007/s12253-010-9343-x PG 5 ER PT J AU Horvath, Zs Torday, L Hitre, E Ganofszky, E Juhos, Czegledi, F Urban, L Polgar, Cs Lang, I Eckhardt, S Kasler, M AF Horvath, Zsolt Torday, Laszlo Hitre, Erika Ganofszky, Erna Juhos, Eva Czegledi, Ferenc Urban, Laszlo Polgar, Csaba Lang, Istvan Eckhardt, Sandor Kasler, Miklos TI Inflammatory Breast Cancer—Comparing the Effectivity of Preoperative Docetaxel-Epirubicine Protocol to Conventional Antracycline-Containing Chemotherapy to Achieve Clinical Benefit and Complete Pathological Response SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Anthracycline; Docetaxel; Inflammatory breast cancer; Pathological complete remission; Primary systemic chemotherapy ID Anthracycline; Docetaxel; Inflammatory breast cancer; Pathological complete remission; Primary systemic chemotherapy AB Our retrospective analysis compared the effectiveness of conventional antracycline-containing protocols (A+) and docetaxel/epirubicine (TE) as primary systemic chemotherapies (PSCT) for inflammatory breast cancer (IBC). Seventy IBC patients received either A+(n=48) or TE (n=22) as PSCT. The objective clinical response and clinical benefit rate of treated patients were 54.3% (A+: 54,2% vs. TE: 54,5%; p=0,28) and 92.8% (A+: 91,7% vs. TE: 95,5%; p=0,57), respectively. The clinical complete response rate (cCR) was 23.2% (A+: 27,1% vs. TE:4,5%; χ2=4,79; p=0,03) with 7.14% (A+: 10,4% vs. TE:0%; χ2=2,47; p=0,12) of pathological complete responses (pCR). The median progression free (PFS)/local progression free (LPFS)/overall survival (OS) was 2.0/5.4/4.0 years, respectively. Patients achieving cCR had a tendency for better survival parameters than patients with less than cCR. Response rates or survival data were not statistically different in the two chemotherapy (CT) treatment groups. The survival was not influenced by the number of CT cycles in either protocols. In this set of patients, the clinical efficacy of the two alternative primary systemic chemotherapies (A+and TE) is equivalent in the treatment of inflammatory breast cancer (IBC), despite of the significant difference in favour of A+noticed in CRs. Six cycles of CT could be enough for patients achieving CR, however sequential pre- and/or postoperative CT with non crossresistant drugs should be considered for non-responders. C1 [Horvath, Zsolt] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy u 7-9, H1122 Budapest, Hungary. [Torday, Laszlo] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H6720 Szeged, Hungary. [Hitre, Erika] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy u 7-9, H1122 Budapest, Hungary. [Ganofszky, Erna] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy u 7-9, H1122 Budapest, Hungary. [Juhos, Eva] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy u 7-9, H1122 Budapest, Hungary. [Czegledi, Ferenc] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy u 7-9, H1122 Budapest, Hungary. [Urban, Laszlo] Petz Aladar County et University Teaching Hospital, Oncopulmonolgic Centre, Vasvary u. 2-4, H9024 Gyor, Hungary. [Polgar, Csaba] National Institute of Oncology, Center of Radiotherapy, Rath Gy u 7-9, H1122 Budapest, Hungary. [Lang, Istvan] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy u 7-9, H1122 Budapest, Hungary. [Eckhardt, Sandor] National Institute of Oncology, Department of Diagnostic Pathology, Rath Gy u 7-9, H1122 Budapest, Hungary. [Kasler, Miklos] National Institute of Oncology, Department of Diagnostic Pathology, Rath Gy u 7-9, H1122 Budapest, Hungary. RP Horvath, Zs (reprint author), National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, H1122 Budapest, Hungary. EM hor.zsolt@oncol.hu;zsodicsom@gmail.com CR Levine PH, Steinhorn SC, Ries LG, Aron JL, 1985, Inflammatory breast cancer: the experience of the surveillance, epidemiology, and end results, SEER, program. J Natl Cancer Inst 74:291–297 Hance KW, Anderson WF, Devesa SS et al, 2005, Trends in inflammatory breast carcinoma incidence and survival: the surveillance, epidemiology, and end results program at the National Cancer Institute. J Natl Cancer Inst 97:966–975 Giordano SH, Hortobagyi GN, 2003, Inflammatory breast cancer: clinical progress and the main problems that must be addressed. Breast Cancer Res 5:284–288 Chang S, Parker SL, Pham T et al, 1998, Inflammatory breast carcinoma incidence and survival: the surveillance, epidemiology, and end results program of the National Cancer Institute, 1975- 1992. Cancer 82:2366–2372 Haagensen CD, 1971, Diseases of the breast. Saunders, Philadelphia Chevallier B, Bastit P, Graic Y et al, 1993, The Centre H. Becquerel studies in inflammatory non metastatic breast cancer. Combined modality approach in 178 patients. Br J Cancer 67:594–601 Jaiyesimi IA, Buzdar AU, Hortobagyi G, 1992, Inflammatory breast cancer: a review. J Clin Oncol 10:1014–1024 Gianni L, Eiermann W, Semiglazov V et al, 2010, Neoadjuvant chemotherapy with trastuzumab followed by adjuvant trastuzumab versus neoadjuvant chemotherapy alone, in patients with HER2-positive locally advanced breast cancer, the NOAH trial): a randomised controlled superiority trial with a parallel HER2- negative cohort. Lancet 375:377–384 Mehta RS, Schubbert T, Kong K, 2008, Trastuzumab in inflammatory breast cancer. Ann Oncol 19:1815–1817 Ward S, Simpson E, Davis S et al, 2007, Taxanes for the adjuvant treatment of early breast cancer: systematic review and economic evaluation. Health Technol Assess 11:1–144 Sinn HP, Schmid H, Junkermann H et al, 1994, Histologic regression of breast cancer after primary, neoadjuvant, chemotherapy. Geburtshilfe Frauenheilkd 54:552–558 von Minckwitz G, Kummel S, Vogel P et al, 2008, Intensified neoadjuvant chemotherapy in early-responding breast cancer: phase III randomized GeparTrio study. J Natl Cancer Inst 100:552–562 von Minckwitz G, Kummel S, Vogel P et al, 2008, Neoadjuvant vinorelbine-capecitabine versus docetaxel-doxorubicincyclophosphamide in early nonresponsive breast cancer: phase III randomized GeparTrio trial. J Natl Cancer Inst 100:542–551 Low JA, Berman AW, Steinberg SM et al, 2004, Long-term follow-up for locally advanced and inflammatory breast cancer patients treated with multimodality therapy. J Clin Oncol 22:4067–4074 de Matteis A, Nuzzo F, D’Aiuto G et al, 2002, Docetaxel plus epidoxorubicin as neoadjuvant treatment in patients with large operable or locally advanced carcinoma of the breast: a singlecenter, phase II study. Cancer 94:895–901 Cristofanilli M, Valero V, Buzdar AU et al, 2007, Inflammatory breast cancer, IBC, and patterns of recurrence: understanding the biology of a unique disease. Cancer 110:1436–1444 Muller C, Caputo A, Schumacher M et al, 2007, Clinical response by palpation during primary systemic therapy with four dosedense cycles doxorubicin and docetaxel in patients with operable breast cancer: further results from a randomised controlled trial. Eur J Cancer 43:1654–1661 Kuerer HM, Newman LA, Smith TL et al, 1999, Clinical course of breast cancer patients with complete pathologic primary tumor and axillary lymph node response to doxorubicin-based neoadjuvant chemotherapy. J Clin Oncol 17:460–469 Ueno NT, Buzdar AU, Singletary SE et al, 1997, Combinedmodality treatment of inflammatory breast carcinoma: twenty years of experience at M. D. Anderson Cancer Center. Cancer Chemother Pharmacol 40:321–329 Harris EE, Schultz D, Bertsch H et al, 2003, Ten-year outcome after combined modality therapy for inflammatory breast cancer. Int J Radiat Oncol Biol Phys 55:1200–1208 Bauer RL, Busch E, Levine E, Edge SB, 1995, Therapy for inflammatory breast cancer: impact of doxorubicin-based therapy. Ann Surg Oncol 2:288–294 Rouzier R, Pusztai L, Delaloge S et al, 2005, Nomograms to predict pathologic complete response and metastasis-free survival after preoperative chemotherapy for breast cancer. J Clin Oncol 23:8331–8339 Abrial SC, Penault-Llorca F, Delva R et al, 2005, High prognostic significance of residual disease after neoadjuvant chemotherapy: a retrospective study in 710 patients with operable breast cancer. Breast Cancer Res Treat 94:255–263 Chaturvedi S, McLaren C, Schofield AC et al, 2005, Patterns of local and distant disease relapse in patients with breast cancer treated with primary chemotherapy: do patients with a complete pathological response differ from those with residual tumour in the breast? Breast Cancer Res Treat 93:151–158 Mazouni C, Kau SW, Frye D et al, 2007, Inclusion of taxanes, particularly weekly paclitaxel, in preoperative chemotherapy improves pathologic complete response rate in estrogen receptorpositive breast cancers. Ann Oncol 18:874–880 Panades M, Olivotto IA, Speers CH et al, 2005, Evolving treatment strategies for inflammatory breast cancer: a population-based survival analysis. J Clin Oncol 23:1941–1950 Baldini E, Gardin G, Giannessi PG et al, 2003, Accelerated versus standard cyclophosphamide, epirubicin and 5-fluorouracil or cyclophosphamide, methotrexate and 5-fluorouracil: a randomized phase III trial in locally advanced breast cancer. Ann Oncol 14:227–232 Barker JL, Montague ED, Peters LJ, 1980, Clinical experience with irradiation of inflammatory carcinoma of the breast with and without elective chemotherapy. Cancer 45:625–629 Cristofanilli M, Buzdar AU, Sneige N et al, 2001, Paclitaxel in the multimodality treatment for inflammatory breast carcinoma. Cancer 92:1775–1782 Smith IC, Heys SD, Hutcheon AW et al, 2002, Neoadjuvant chemotherapy in breast cancer: significantly enhanced response with docetaxel. J Clin Oncol 20:1456–1466 Evans TR, Yellowlees A, Foster E et al, 2005, Phase III randomized trial of doxorubicin and docetaxel versus doxorubicin and cyclophosphamide as primary medical therapy in women with breast cancer: an anglo-celtic cooperative oncology group study. J Clin Oncol 23:2988–2995 Bear HD, Anderson S, Smith RE et al, 2006, Sequential preoperative or postoperative docetaxel added to preoperative doxorubicin plus cyclophosphamide for operable breast cancer: National Surgical Adjuvant Breast and Bowel Project Protocol B- 27. J Clin Oncol 24:2019–2027 GralowJR, Burstein HJ,WoodWet al, 2008, Preoperative therapy in invasive breast cancer: pathologic assessment and systemic therapy issues in operable disease. J Clin Oncol 26:814–819 Wang B, Fu JF, Hong ZW, 2009, Efficacy of neoadjuvant chemotherapy with FEC and TEC regimen on breast cancer. Ai Zheng 28:292–296 Steger GG, Galid A, Gnant M et al, 2007, Pathologic complete response with six compared with three cycles of neoadjuvant epirubicin plus docetaxel and granulocyte colony-stimulating factor in operable breast cancer: results of ABCSG-14. J Clin Oncol 25:2012–2018 Manga GP, Shahi PK, Urena MM et al, 2009, Phase II study of neoadjuvant treatment with doxorubicin, docetaxel, and capecitabine, ATX, in locally advanced or inflammatory breast cancer. Breast Cancer 17:205–211 Damast S, Ho AY, Montgomery L et al, 2009, Locoregional Outcomes of Inflammatory Breast Cancer Patients Treated with Standard Fractionation Radiation and Daily Skin Bolus in the Taxane Era. Int J Radiat Oncol Biol Phys 77:1105–1112 Schmidt M, Bremer E, Hasenclever D et al, 2007, Role of the progesterone receptor for paclitaxel resistance in primary breast cancer. Br J Cancer 96:241–247 Voordeckers M, Vinh-Hung V, Van de Steene J et al, 2004, The lymph node ratio as prognostic factor in node-positive breast cancer. Radiother Oncol 70:225–230 Thomas E, Holmes FA, Smith TL et al, 2004, The use of alternate, non-cross-resistant adjuvant chemotherapy on the basis of pathologic response to a neoadjuvant doxorubicin-based regimen in women with operable breast cancer: long-term results from a prospective randomized trial. J Clin Oncol 22:2294–2302 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 541 EP 550 DI 10.1007/s12253-010-9344-9 PG 10 ER PT J AU Vereczkey, I Serester, O Dobos, J Gallai, M Szakacs, O Szentirmay, Z Toth, E AF Vereczkey, Ildiko Serester, Orsolya Dobos, Judit Gallai, Monika Szakacs, Orsolya Szentirmay, Zoltan Toth, Erika TI Molecular Characterization of 103 Ovarian Serous and Mucinous Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Borderline; Molecular; Mucinous; Ovary; Pathogenesis; Serous ID Borderline; Molecular; Mucinous; Ovary; Pathogenesis; Serous AB The pathogenesis of ovarian carcinomas is heterogeneous, with even the same entities showing great variance. In our study we investigated the mutations of the BRAF, KRAS, and p53 genes in serous and mucinous borderline tumors and in low grade and high grade serous and mucinous tumors. The mutations of BRAF and KRAS genes have been shown in 60% of borderline and low grade (well differentiated) serous and mucinous tumors, but very rarely in high grade (moderately and poorly differentiated) carcinomas. However mutations of p53 are very common in high grade tumors and this indicates a "dualistic" model of ovarian tumorigenesis. A total of 80 serous tumors, including serous borderline, low grade and high grade tumors, and 23 mucinous tumors, including borderline and invasive tumors were analysed for BRAF and KRAS mutations using real time PCR method followed by melting point analysis. P53 mutation was investigated by immunohistochemistry. We assumed mutation of the p53 gene when 100% of tumor cells showed strong nuclear positivity. We observed differences in genetic alterations in the development of the low grade tumors and between low and high grade tumors too. In some bilateral or stage II-III cases we observed differences between the mutation status of the left and right ovarian tumors and between the primary tumor and its implants. In one case in a tumor with micropapillary pattern showing high grade nuclear atypia we could detect mutations in both KRAS and p53 genes. The majority of our mucinous ovarian tumor cases showed a KRAS mutation. We have not found mutations of the BRAF and p53 genes in these cases. We have found as have others, that there is a dualistic pathway of ovarian carcinogenesis. In the majority of cases, low grade epithelial tumors develop in a stepwise manner due to genetic alterations of the members of MAP-kinase pathway; however mutation of the p53 gene is the key event in the development of high grade tumors. C1 [Vereczkey, Ildiko] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gy u 7-9, 1122 Budapest, Hungary. [Serester, Orsolya] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gy u 7-9, 1122 Budapest, Hungary. [Dobos, Judit] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gy u 7-9, 1122 Budapest, Hungary. [Gallai, Monika] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gy u 7-9, 1122 Budapest, Hungary. [Szakacs, Orsolya] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gy u 7-9, 1122 Budapest, Hungary. [Szentirmay, Zoltan] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gy u 7-9, 1122 Budapest, Hungary. [Toth, Erika] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gy u 7-9, 1122 Budapest, Hungary. RP Vereczkey, I (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, 1122 Budapest, Hungary. EM vereczkey.ildiko@oncol.hu CR FIGO. Intenational Federation of Gynecology and Obstetrics, 1971, Classification and staging of malignant tumours in the female pelvis. Acta Obstet Gynecol Scand 50:1–7 Serov SF, Scully RE, Sobin LH, 1973, International histologic Classification of Tumours. No. 9. Histological Typing of Ovarian Tumours. World Health Organization, Geneva Osterbeerg L, Akeson M, Levan K et al, 2006, Genetic alterations of serous borderline tumors of the ovary compared to stage I serous ovarian carcinomas. Cancer Genet Cytogenet 167:103–108 Singer G, Oldt R, Cohen Y et al, 2003, Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst 95:484–486 Singer G, Stohr R, Cope L et al, 2005, Patterns of p53 mutations separate ovarian serous borderline tumors and low —and highgrade carcinomas and provide support for a new model of ovarian carcinogenesis, A mutational analysis with immunohistochemical correlation). Am J Surg Pathol 29(2):218–224 Hsu C-Y, Bristow R, Cha M et al, 2004, Characterization of active mitogen-activated protein kinase in ovarian serous carcinomas. Clin Cancer Res 10:6432–6436 Staebler A, Diebold J, 2007, Molekularpathologie der epithelialen Ovarialneoplasien, Von der Phanotyp-Genotyp-Korrelation zu neuen Ansatzpunkten in Diagnostik und Therapie). Pathologe 28:180–186 Ho C-L, Kurman RJ, Dehari R et al, 2004, Mutations of BRAF and KRAS precede the development of ovarian serous borderline tumors. Cancer Res 64:6915–6918 Shih I-M, Kurman RJ, 2004, Ovarian tumorigenesis, Review). Am J Path 164(5):1511–1518 Kurman RJ, Shih I-M, 2008, Pathogenesis of ovarian cancer. Lessons from morphology and molecular biology and their implications. Int J Gynecol Pathol 27(2):151–160 Seidman JD, Horkayne-Szakaly I, Haiba M et al, 2004, The histologic type and stage distribution of ovarian carcinomas of surface epithelial origin. Int J Gynecol Pathol 23:41–44 Hart WR, 2005, Borderline epithelial tumors of the ovary. Mod Path 18:33–50 Bell DA, 2005, Origins and molecular pathology of ovarian cancer. Mod Pathol 18:S19–S32 Christie M, Oehler MK, 2006, Molecular pathology of epithelial ovarian cancer. J Br Menopause Soc 12(2):57–63 Singer G, Kurman RJ, Chang H-W et al, 2002, Diverse tumorigenic pathways in ovarian serous carcinoma. Am J Pathol 160:1223–1228 Shih I-M, Kurman RJ, 2005, Molecular pathogenesis of ovarian borderline tumors: new insight and old challenges. Clin Cancer Res 11:7273–7279 Sridhar SS, Hedley D, Siu LL, 2005, Raf kinase as a target for anticancer therapeutics. Mol Cancer Ther 4(4):677–685 Pohl G, Ho C-L, Kurman RJ, 2005, Inactivation of the Mitogen- Activared Protein Kinase pathway as a potential target-based therapy in ovarian serous tumors with KRAS or BRAF mutations. Cancer Res 65(5):1994–2000 Sieben NLG, Macropoulos P, Roemen GMJM et al, 2004, In ovarian neoplasms BRAF, but not KRAS, mutations are restricted to low-grade serous tumours. J Pathol 202:336–340 Mayr D, Hirschmann A, Lohrs U et al, 2006, KRAS and BRAF mutations in ovarian tumors: A comprehensive study of invasive carcinomas, borderline tumors and extraovarian implants. Gynecol Oncol 103:883–887 Ueda M, Toji E, Noda S, 2007, Germ line and somatic mutations of BRAF V599E in ovarian carcinoma. Int J Gynecol Cancer Wang L, Arnold K, 2003, Common mutations prevalent in lowgrade ovarian cancers. J Nat Cancer Inst 95(6):417–418 Garrett AP, Lee KP, Colitti CR et al, 2001, K-RAS mutation may be an early event in mucinous ovarian tumorigenesis. Int J Gynecol Pathol 20:244–251 Vang R, Shih I-M, Salani R et al, 2008, Subdividing ovarian and peritoneal serous carcinoma into moderately differentiated and poorly differentiated does not have biologic validity based on molecular genetic and in vitro drug resistance data. Am J Surg Pathol 32(11):1667–1674 Kobel M, Hunstman D, Gilks CB, 2008, Critical molecular abnormalities in high grade serous carcinoma of the ovary. Expert Rev Mol Med 10:e22 Russell SEH, McCluggage WG, 2004, A multistep model for ovarian tumorigenesis: the value of mutation analysis in the KRAS and BRAF genes. L Pathol 203:617–619 Lee Y, Miron A, Drapkin R et al, 2007, A candidate precursor to serous sarcinoma that originates in the distal fallopian tube. J Pathol 211(1):26–35 Goodman MT, Shvetsov YB, 2009, Incidence of ovarian, peritoneal, and fallopian tube carcinomas in the United States, 1995–2004. Cancer Epidemiol Biomarkers Prev 18(1):132–139 Tong G-X, Chiriboga L, Hamele-Bena D et al, 2007, Expression of PAX2 in papillary serous carcinoma of the ovary: immunohistochemical evidence of fallopian tube or secondary Mullerian system origin? Mod Pathol 20:856–863 Roh MH, Kindelberger D, Crum CP, 2009, Serous tubal intraepithelial carcinoma and dominant ovarian mass: clues to serous tumor origin? Am J Surg Pathol 33(3):376–383 Leiato MM, Soslow RA, Baergen RN et al, 2004, Mutation and expression of the TP53 gene in early stage epithelial ovarian carcinoma. Gynecol Oncol 93:301–306 Morice P, Uzan C, Kane A et al, 2009, Prognostic factors of patients with advanced stage serous borderline tumors of the ovary. J Clin Oncol 27:15s, suppl;abstr 5573) Sieben NL, Roemen GMJM, Oosting J, 2006, Clonal analysis favours a monoclonal origin for serous borderline tumours with peritoneal implants. J Pathol 210:405–411 Krishnamurti U, Sasatomi E, Swalsky PA, 2005, Microdissectionbased mutational genotyping of serous borderline tumors of the ovary. Int J Gynecol Pathol 24:56–61 Diebold J, Seemuller F, Lohrs U, 2003, K-RAS mutations in ovarian and extraovarian lesions of serous tumors of borderline malignancy. Lab Invest 83:251–258 Zanotti KM, Hart WR, Kennedy AW, 1999, Allelic imbalance on chromosome 17p13 in borderline, low malignant potential, epithelial ovarian tumors. Int J Gynecol Pathol 18:247–253 Gu J, Roth LM, Younger C et al, 2001, Molecular evidence for the independent origin of extraovarian papillary serous tumors of low malignant potential. J Natl Cancer Inst 93:1147–1152 Lu KH, Bell DA, Welch WR et al, 1998, Evidence for the multifocal origin of bilateral and advanced human serous borderline ovarian tumors. Cancer Res 58:2328–2330 Davies H, Bignell GR, Cox C et al, 2002, Mutations of BRAF gene in human cancer. Nature 417:949–954 Cheng EJ, Kurman RJ, Wang M et al, 2004, Molecular genetic analysis of ovarian serous cystadenomas. Lab Invest 84:778– 784 Burks RT, Sherman ME, Kurman RJ, 1996, Micropapillary serous carcinoma of the ovary: a distinctive low-grade carcinoma related to serous borderline tumors. Am J Surg Pathol 20:1319–1330 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 551 EP 559 DI 10.1007/s12253-010-9345-8 PG 9 ER PT J AU Li, QL Yang, ZL Liu, JQ Miao, XY AF Li, Qing-Long Yang, Zhu-Lin Liu, Jie-Qiong Miao, Xiong-Ying TI Expression of CDX2 and Hepatocyte Antigen in Benign and Malignant Lesions of Gallbladder and Its Correlation with Histopathologic Type and Clinical Outcome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gallbladder cancer; Chronic cholecystitis; CDX2; Hepatocyte antigen (Hep); Hepatocyte paraffin 1; Immunohistochemistry; Clinicopathology ID Gallbladder cancer; Chronic cholecystitis; CDX2; Hepatocyte antigen (Hep); Hepatocyte paraffin 1; Immunohistochemistry; Clinicopathology AB Recent studies have shown that both CDX2 and Hepatocyte antigen (Hep) are detected in different types of cancer and associated with clinical prognosis. However, fever studies have examined gallbladder cancer specimens, and little is known about the clinicopathological significance of both CDX2 and Hep expression in gallbladder adenocarcinomas. In present study, we examined the expression frequencies of CDX2 and Hepatocyte antigen (Hep), and explored their clinicopathologic significances in gallbladder adenocarcinoma. Immunohistochemistry was used to detect and compare the frequencies of CDX2 and Hep expression in 108 samples of gallbladder adenocarcinoma, 46 peri-tumor tissues and 35 chronic cholecystitis. The expression frequencies for CDX2 and Hep were 49/108 (45.4%) and 45/108 (41.7%) in gallbladder carcinoma; 13/46 (28.3%) and 11/46 (23.9) in peri-tumor tissues; 5/35 (14.3%) and 2/35 (5.7%) in chronic cholecystitis. The positive staining of CDX2 or Hep in gallbladder adenocarcinoma was significantly higher than that in peritumoral tissues (both, P<0.05), and chronic cholecystits (both, P<0.01). The expression of CDX2 or Hep was negatively correlated to grade of differentiation, tumor size and lymph node metastasis (P<0.01 or P<0.05). Elevated expression frequency of CDX2 or Hep was associated with increased overall survival (P=0.003 or P=0.002). Multivariate Cox regression analysis showed that CDX2 (P=0.014) or Hep (P=0.026) expression was an independent prognostic predictor in gallbladder adenocarcinoma. CDX2 and Hep might function as important biological markers in the development and prognosis of gallbladder adenocarcinoma. C1 [Li, Qing-Long] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 139 Middle Renmin Road, 410011 Changsha, Hunan, China. [Yang, Zhu-Lin] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 139 Middle Renmin Road, 410011 Changsha, Hunan, China. [Liu, Jie-Qiong] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 139 Middle Renmin Road, 410011 Changsha, Hunan, China. [Miao, Xiong-Ying] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 139 Middle Renmin Road, 410011 Changsha, Hunan, China. RP Yang, ZL (reprint author), Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, China. EM zhulin.yang@yahoo.com CR Jones RS, 1990, Carcinoma of the gallbladder. Surg Clin North Am 70:1419–1428 Carriaga MT, Henson DE, 1995, Liver, gallbladder, extraHepatic bile ducts, and pancreas. Cancer 75:171–190 Suh E, Chen L, Taylor J, Traber PG, 1994, A homeodomain protein related to caudal regulates intestine-specific gene transcription. Mol Cell Biol 14:7340–7351 Silberg DG, Swain GP, Suh ER, Traber PG, 2000, Cdx1 and cdx2 expression during intestinal development. Gastroenterology 119:961–971 Phillips RW, Frierson HF Jr, Moskaluk CA, 2003, Cdx2 as a marker of epithelial intestinal differentiation in the esophagus. Am J Surg Pathol 27:1442–1447 Eda A, Osawa H, Satoh K et al, 2003, Aberrant expression of CDX2 in Barrett’s epithelium and inflammatory esophageal mucosa. J Gastroenterol 38:14–22 Bai YQ, Yamamoto H, Akiyama Yet al, 2002, Ectopic expression of homeodomain protein CDX2 in intestinal metaplasia and carcinomas of the stomach. Cancer Lett 176:47–55 Mutoh H, Hakamata Y, Sato K et al, 2002, Conversion of gastric mucosa to intestinal metaplasia in Cdx2-expressing transgenic mice. Biochem Biophys Res Commun 294:470–479 Kim HS, Lee JS, Freund JN et al, 2006, CDX-2 homeobox gene expression in human gastric carcinoma and precursor lesions. J Gastroenterol Hepatol 21:438–442 Vallbohmer D, DeMeester SR, Peters JH et al, 2006, Cdx-2 expression in squamous and metaplastic columnar epithelia of the esophagus. Dis Esophagus 19:260–266 Nonaka D, Kusamura S, Baratti D, Casali P, Younan R, Deraco M, 2006, CDX-2 expression in pseudomyxoma peritonei: a clinicopathological study of 42 cases. Histopathology 49:381– 387 Ishikawa A, Sasaki M, Ohira S et al, 2004, Aberrant expression of CDX2 is closely related to the intestinal metaplasia and MUC2 expression in intraductal papillary neoplasm of the liver in Hepatolithiasis. Lab Investigation Journal Technical Methods Pathology 84:629–638 Fan Z, Li J, Dong B, Huang X, 2005, Expression of Cdx2 and Hepatocyte antigen in gastric carcinoma: correlation with histologic type and implications for prognosis. Clin Cancer Res 11:6162–6170 Jinawath A, Akiyama Y, Yuasa Y, Pairojkul C, 2006, Expression of phosphorylated ERK1/2 and homeodomain protein CDX2 in cholangiocarcinoma. J Cancer Res Clin Oncol 132:805–810 Sakamoto H, Mutoh H, Ido K, Satoh K, Hayakawa H, Sugano K, 2007, A close relationship between intestinal metaplasia and Cdx2 expression in human gallbladders with cholelithiasis. Hum Pathol 38:66–71 Chang YT, Hsu C, Jeng YM, Chang MC, Wei SC, Wong JM, 2007, Expression of the caudal-type homeodomain transcription factor CDX2 is related to clinical outcome in biliary tract carcinoma. J Gastroenterol Hepatol 22:389–394 Wu XS, Akiyama Y, Igari T et al, 2005, Expression of homeodomain protein CDX2 in gallbladder carcinomas. J Cancer Res Clin Oncol 131:271–278 Chu PG, Ishizawa S,Wu E,Weiss LM(2002, Hepatocyte antigen as a marker of Hepatocellular carcinoma: an immunohistochemical comparison to carcinoembryonic antigen, CD10, and alphafetoprotein. Am J Surg Pathol 26:978–988 Kakar S, Gown AM, Goodman ZD, Ferrell LD, 2007, Best practices in diagnostic immunohistochemistry: Hepatocellular carcinoma versus metastatic neoplasms. Arch Pathol Lab Med 131:1648–1654 Leong AS, Sormunen RT, Tsui WM, Liew CT, 1998, Hep Par 1 and selected antibodies in the immunohistological distinction of Hepatocellular carcinoma from cholangiocarcinoma, combined tumours and metastatic carcinoma. Histopathology 33:318–324 Mosnier JF, Kandel C, Cazals-Hatem D et al, 2009, N-cadherin serves as diagnostic biomarker in intraHepatic and perihilar cholangiocarcinomas. Mod Pathol 22:182–190 Maitra A, Murakata LA, Albores-Saavedra J, 2001, Immunoreactivity for Hepatocyte paraffin 1 antibody in Hepatoid adenocarcinomas of the gastrointestinal tract. Am J Clin Pathol 115:689–694 Lugli A, Tornillo L, Mirlacher M, Bundi M, Sauter G, Terracciano LM, 2004, Hepatocyte paraffin 1 expression in human normal and neoplastic tissues: tissue microarray analysis on 3, 940 tissue samples. Am J Clin Pathol 122:721–727 Fan Z, van de Rijn M, Montgomery K, Rouse RV, 2003, Hep par 1 antibody stain for the differential diagnosis of Hepatocellular carcinoma: 676 tumors tested using tissue microarrays and conventional tissue sections. Mod Pathol 16:137–144 Mac MT, Chung F, Lin F, Hui P, Balzer BL, Wang HL, 2009, Expression of Hepatocyte antigen in small intestinal epithelium and adenocarcinoma. Am J Clin Pathol 132:80–85 Mizoshita T, Tsukamoto T, Nakanishi H et al, 2003, Expression of Cdx2 and the phenotype of advanced gastric cancers: relationship with prognosis. J Cancer Res Clin Oncol 129:727– 734 Seno H, Oshima M, Taniguchi MA et al, 2002, CDX2 expression in the stomach with intestinal metaplasia and intestinal-type cancer: prognostic implications. Int J Oncol 21:769–774 Matsumoto K, Mizoshita T, Tsukamoto T et al, 2004, Cdx2 expression in pancreatic tumors: relationship with prognosis of invasive ductal carcinomas. Oncol Rep 12:1239–1243 Hansel DE, Maitra A, Lin JW et al, 2005, Expression of the caudal-type homeodomain transcription factors CDX 1/2 and outcome in carcinomas of the ampulla of Vater. J Clin Oncol 23:1811–1818 Hong SM, Cho H, Moskaluk CA, Frierson HF Jr, Yu E, Ro JY, 2005, CDX2 and MUC2 protein expression in extraHepatic bile duct carcinoma. Am J Clin Pathol 124:361–370 Kakar S, Muir T, Murphy LM, Lloyd RV, Burgart LJ, 2003, Immunoreactivity of Hep Par 1 in Hepatic and extraHepatic tumors and its correlation with albumin in situ hybridization in Hepatocellular carcinoma. Am J Clin Pathol 119:361–366 Ramos-Vara JA, Miller MA, 2002, Immunohistochemical characterization of canine intestinal epithelial and mesenchymal tumours with a monoclonal antibody to Hepatocyte paraffin 1, Hep Par 1). Histochem J 34:397–401 Hishinuma M, Ohashi KI, Yamauchi N et al, 2006, Hepatocellular oncofetal protein, glypican 3 is a sensitive marker for alphafetoprotein- producing gastric carcinoma. Histopathology 49:479–486 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 561 EP 568 DI 10.1007/s12253-010-9346-7 PG 8 ER PT J AU Iyengar, B Singh, VA AF Iyengar, Bhanu Singh, V Avantika TI Embryonic Vasculogenesis in Nodular Melanomas and Tumour Differentiation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Vasculogenic mimicry; Incorporated microvasculature; Pigmented .Amelanotic; Laminin; Integrin ID Vasculogenic mimicry; Incorporated microvasculature; Pigmented .Amelanotic; Laminin; Integrin AB The relationship of vasculogenic mimicry to pigment in nodular vertical growth phase [VGP] cutaneous melanomas is assessed in this study. 10 nodules each from 27 tumors, 15 pigmented and 12 amelanotic were sampled in proportion to the pigment level. Serial frozen and paraffin sections subjected to HE, Reticulin, PAS to assess the vascular pattern; Dopa Oxidase and Immunopositivity for HMB45, LN5 [laminin 5] & integrin[α5β1], and EM [electron microscopy] to identify Weibel-Palade bodies within endothelial cells. The vascular pattern, pigment and the immunopositivity was mapped to assess the percentage VM [vasculogenic sinusoids] vs INC [incorporated microvasculature]. In pigmented melanomas, INC from preexisting stromal vessels is predominant. Amelanotic melanomas show embryonic vasculogenic mimicry, a selfpropagating system of spaces within the sheets of tumors cells. Both INC and VM co-exist in tumors with both amelanotic and melanotic nodules. In areas with VM, loci of LN5 and α5β1 integrin positive cells appear within the proliferating columns, positivity in these cells suggesting a switch to a more aggressive form. Irregular spaces appear lined by tumor cells, with initial hemopoeitic activity, coalesce and interlink into tubular networks. Spaces lined by tumor cells extend into an intricate network which then connects with the angiogenetic system. The tumor cells lining the vasculogenic spaces are positive for LN5, α5β1 integrin. Statistically, INC is significantly higher in pigmented melanomas, whereas amelanotic melanomas show significantly higher VM. Pigmentation is correlated positively with INC and negatively with VM. INC and VM are negatively correlated with each other. C1 [Iyengar, Bhanu] Iyengar Farm, Pigment Cell Center, Brijwasan Road, PO Kapashera, 110037 New Delhi, India. [Singh, V Avantika] Iyengar Farm, Pigment Cell Center, Brijwasan Road, PO Kapashera, 110037 New Delhi, India. RP Iyengar, B (reprint author), Iyengar Farm, Pigment Cell Center, 110037 New Delhi, India. EM bhanu_i@yahoo.com CR Folkman J, 1995, Clinical applications of research on angiogenesis. Seminars in Medicine of the Beth Israel Hospital, Boston. New Engl J Med 333:1757–1763 Folkman J, 1971, Tumour angiogenesis: therapeutic implications. N Engl J Med 285:1182–1186 Risau W, 1997, Mechanism of angiogenesis. Nature 387:671–674 Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe’er J, Trent JM, Meltzer PS, Hendrix MJC, 1999, Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 13:739–752 Prophet ED, Mills B, Arrington JB, Sobin LH, 1994, Laboratory methods in histotechnology. American registry of pathology, Armed Forces Institute of Pathology, Washington Mikel UV, 1994, Advanced laboratory methods in histology and pathology. American Registry of Pathology, Armed Forces Institute of Pathology, Washington Pearse AGE, 1985, Histochemistry theoretical and applied. Vol II: analytical Technology. Churchill Livingstone, London, pp 611– 674 Liu W, Dowling JP, Murray WK, McArthur GA, Thompson JF, Wolfe R, Kelly JW, 2006, Rate of growth in Melanomas: characteristics and associations of rapidly growing melanomas. Arch Dermatol 142:131–138 Shellman YG, Chapman JT, Fujita M, Norris DA, Maxwell IH, 2000, Expression of activated N-ras in a primary melanoma cell line counteracts growth inhibition by transforming growth factorß. J Invest Dermatol 114:1200–1204 Hanahan D, Folkman J, 1996, Patterns and emerging mechanisms of the angiogenic switch during tumourigenesis. Cell 86:353–364 Papetti M, Herman IM, 2002, Mechanisms of normal and tumorderived angiogenesis. Am J Physiol Cell Physiol 282:C947–C970 Norrby K, 1997, Angiogenesis: new aspects relating to its initiation and control. APMIS 105:417–437 Verheul HM, Voest EE, Schlingemann RO, 2004, Are tumours angiogenesis-dependent? J Pathol 202(1):5–13 Shima DT, Mailhos C, 2000, Vascular development biology: getting nervous. Curr Opin Genet Dev 10:536–542 Dickson BJ, 2002, Molecular mechanisms of axon guidance. Science 298:1959–1964 Bjornson CR, Rietze RL, Reynolds BA, Magli MC, Vescovi AL, 1999, Turning brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo. Science 283(5401):534–537 Oishi K, Uchida MK, 2003, Functional differentiation of neural stem cells into endothelial cells. Nippon Yakurigaku Zasshi 122, Suppl):27P–29P Wurmser AE, Nakashima K, Summers RG, Toni N, D’Amour KA, Lie DC, Gage FH, 2004, Cell fusion-independent differentiation of neural stem cells to the endothelial lineage. Nature 430, 6997):350–356 Huttenbach Y, Prieto VG, Reed JA, 2002, Desmoplastic and spindle cell melanomas express protein markers of the neural crest but not of later committed stages of Schwann cell differentiation. J Cutan Pathol 29(9):562–568 Akimoto S, Nakanishi Y, Sakamoto M, Kanai Y, Hirohashi S, 2004, Laminin 5 beta3 and gamma2 chains are frequently coexpressed in cancer cells. Pathol Int 54(9):688–692 Seftor REB, Seftor EA, Koshikawa N, Meltzer PS, Gardner LMG, Bilban M, Stetler-Stevenson WG, Quaranta V, Hendrix MJC, 2001, Cooperative interactions of Laminin 5γ2 Chain, Matrix Metalloproteinase-2, and Membrane Type-1-Matrix/Metalloproteinase are required for mimicry of embryonic vasculogenesis by aggressive melanoma. Cancer Res 61:6322–6327 Hendrix MJ, Seftor EA, Kirschmann DA, Quaranta V, Seftor RE, 2003, Remodeling of the microenvironment by aggressive melanoma tumor cells. Ann NY Acad Sci 995:151–161 Hendrix MJC, Seftor REB, Seftor EA, Gruman LM, Lee LML, Nickoloff BJ, Miele L, Sheriff DD, Schatteman GC, 2002, Transendothelial function of human metastatic melanoma cells. Role of the microenvironment in cell-fate determination. Cancer Res 62:665–668 Goldfinger LE, Hopkinson SB, deHart GW, Collawn S, Couchman JR, Jones JCR, 1999, The a3 laminin subunit, a6b4 and a3b1 integrin coordinately regulate wound healing in cultured epithelial cells and in the skin. J Cell Sci 112:2615–2629 Trikha M, Timar J, Zacharek A, Nemeth JA, Cai Y, Dome B, Somlai B, Raso E, Ladanyi A, Honn KV, 2002, 2002 Role for beta3 integrins in humanmelanoma growth and survival. Int J Cancer 101(2):156–167 Dome B, Paku S, Somlai B, Timar J, 2002, Vascularization of cutaneous melanoma involves vessel co-option and has clinical significance. J Pathol 197(3):355–362 Timar J, Dome B, Fazekas K, Janovics A, Paku S, 2001, Angiogenesis-dependent diseases and angiogenesis therapy. Pathol Oncol Res 7(2):85–94, Review Koistinen P, Ahonen M, Kahari VM, Heino J, 2004, alphaV integrin promotes in vitro and in vivo survival of cells in metastatic melanoma. Int J Cancer 112(1):61–70 Mitjans F, Meyer T, Fittschen C, Goodman S, Jonczyk A, Marshall JF, Reyes G, Piulats J, 2000, In vivo therapy of malignant melanoma by means of antagonists of alphav integrins. Int J Cancer 87(5):716–723 Pignatelli M, Stamp G, 1995, Integrins in tumour development and spread. Cancer Surv 24:113–127 Ivaska J, Heino J, 2000, Adhesion receptors and cell invasion: mechanisms of integrin guided degradation of extracellular matrix. Cell Mol Life Sci 57:16–24 Mitra A, Chakrabarti J, Chatterjee A, 2003, Binding of alpha5 monoclonal antibody to cell surface α5β1 integrin modulates MMP-2 and MMP-7 activity in B16F10 melanoma cells. J Environ Pathol Toxicol Oncol 22(3):167–178 Zambruno G, Marchisio PC, Melchiori A, Bondanza S, Cancedda R, De Lucca M, 1993, Expression of integrin receptors and their role in adhesion, spreading and migration of normal human melanocytes. J Cell Sci 105:179–190 Dirkx AEM, Egbrink MGA, Castermans K, van der Schaft DWJ, Thijssen VLJL, Dings RPM, Kwee L, Mayo KH, Wagstaff J, Boumater Steege JCA, Griffioen AW, 2006, Anti-angiogenesis therapy can overcome endothelial cell anergy and promote leukocyte-endothelium interactions and infiltration in tumors. FASEB J 20:621–630 Castermans K, Griffioen AW, 2007, Tumor blood vessels, a difficult hurdle for infiltrating leukocytes. Biochim Biophys Acta, Rev Cancer 1776:160–174 Molema G, 2002, Tumor vasculature directed drug targeting: applying new technologies and knowledge to the development of clinically relevant therapies. Pharm Res 19:1251–1258 Griffioen AW, Tromp SC, Hillen HFP, 2002, Angiogenesis modulates the tumour immune response. Int J Expert Pathol 79:363–368 Griffioen AW(2008, Anti-angiogenesis: making the tumor vulnerable to the immune system. Cancer Immunol Immunother 57:1553–1558 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 569 EP 577 DI 10.1007/s12253-010-9350-y PG 9 ER PT J AU Mehdi, JS Ali, A Rizvi, AM AF Mehdi, Jafar Syed Ali, Asgar Rizvi, Alam Moshahid TI Parkin Gene Alterations in Ovarian Carcinoma from Northern Indian Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Loss of heterozygosity; Ovarian cancer; Parkin gene; Reverse transcriptase PCR; Tumor suppressor gene ID Loss of heterozygosity; Ovarian cancer; Parkin gene; Reverse transcriptase PCR; Tumor suppressor gene AB Parkin, a tumor suppressor gene located on chromosome 6q25-27, has been identified as a target for mutation in many human malignancies like breast, ovaries, cervical and lungs etc. After a preliminary report on the loss of heterozygosity and altered Parkin expression in breast and ovarian tumors, we aimed to study loss of heterozygosity in the Parkin gene associated microsatellite markers and its expression in human ovarian cancer patients from Indian population. We examined 102 paired normal and ovarian cancer samples for allelic loss in Parkin gene locus using Parkin gene associated microsatellite markers through loss of heterozygosity and changes in its expression through semiquantitative RT-PCR. Loss of heterozygosity identified common region of loss in Parkin locus with highest frequency for the intragenic marker D6S1599 (53%) whereas, 49 of 102 (48%) specimens showed decreased or no expression of Parkin in ovarian tumors. The study revealed that presence of loss of heterozygosity was significantly higher in both the intragenic markers (D6S1599 and D6S305) as compared with the locus of flanking region (D6S1008) with their p value 0.000001 and 0.00008, respectively. It also revealed that Parkin inactivation is probably a combination of loss of heterozygosity coupled with downregulation of Parkin gene through an alternative means like epigenetic mechanism. These data strongly supports the previous study and argue that Parkin is a tumor suppressor gene whose inactivation may play an important role in ovarian carcinoma. C1 [Mehdi, Jafar Syed] Jamia Millia Islamia, Department of Biosciences, Genome Biology Lab, Maulana Mohammad Ali Jauhar MargNew Delhi, India. [Ali, Asgar] Jamia Millia Islamia, Department of Biosciences, Genome Biology Lab, Maulana Mohammad Ali Jauhar MargNew Delhi, India. [Rizvi, Alam Moshahid] Jamia Millia Islamia, Department of Biosciences, Genome Biology Lab, Maulana Mohammad Ali Jauhar MargNew Delhi, India. RP Rizvi, AM (reprint author), Jamia Millia Islamia, Department of Biosciences, Genome Biology Lab, New Delhi, India. EM rizvijmi@gmail.com CR Landis SH, Murray T, Bolden S, Wingo PA, 1998, Cancer statistics, 1998. Cancer J Clin 48:6–29 Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ, 2008, Cancer statistics, 2008. CA Cancer J Clin 58(2):71–96 Boyle P, Levin B, 2008, International Agency for Research on Cancer, and World Health Organization, “World cancer report 2008.” Distributed by WHO Press, Geneva, Switzerland American Cancer Society. Facts & Figures 2009 GLOBOCAN, 2008, Cancer incidence, mortality and prevalence worldwide. IARC 2008. IARC, Lyon Theile M, Seitz S, Arnold W, Jandrig B, Frege R, Schlag PM, Haensch W, Guski H, Winzer KJ, Barrett JC, Scherneck S, 1996, A defined chromosome 6q fragment, at D6S310, harbors a putative tumor suppressor gene for breast cancer. Oncogene 13:677–685 Bastian BC, LeBoit PE, Hamm H, Brocker EB, Pinkel D, 1998, Chromosomal gains and losses in primary cutaneous melanomas detected by comparative genomic hybridization. Cancer Res 58:2170–2175 Reutzel D, Mende M, Naumann S, Storkel S, Brenner W, Zabel B, Decker J, 2001, Genomic imbalances in 61 renal cancers from the proximal tubulus detected by comparative genomic hybridization. Cytogenet Cell Genet 93:221–227 Jin C, Martins C, Jin Y, Wiegant J, Wennerberg J, Dictor M, Gisselsson D, Strombeck B, Fonseca I, Mitelman F, Tanke HJ, Hoglund M, Mertens F, 2001, Characterization of chromosome aberrations in salivary gland tumors by FISH, including multicolor COBRA-FISH. Genes Chromosome Cancer 30:161–167 Jackson A, Carrara P, Duke V, Sinclair P, Papaioannou M, Harrison CJ, Foroni L, 2000, Deletion of 6q16–q21 in human lymphoid malignancies: a mapping and deletion analysis. Cancer Res 60:2775–2779 Carvalho B, Veen AY, Gartner F, Carneiro F, Seruca R, Buys CHCM, Kok K, 2001, Allelic gains and losses in distinct regions of chromosome 6 in gastric carcinoma. Cancer Genet Cytogenet 131(1):54–59 De Souza AT, Hankins GR, Washington MK, Fine RL, Orton TC, Jirtle RL, 1995, Frequent loss of heterozygosity on 6q at the mannose 6-phosphate/insulin-like growth factor II receptor locus in human hepatocellular tumors. Oncogene 10(9):1725–1729 Merlo A, Gabrielson E, Mabry M, Vollmer R, Baylin SB, Sidransky D, 1994, Homozygous deletion on chromosome 9p and loss of heterozygosity on 9q, 6p, and 6q in primary human small cell lung cancer. Cancer Res 54:2322–2326 Visakorpi T, Kallioniemi AH, Syvanen AC, Hyytinen ER, Karhu R, Tammela T, Isola JJ, Kallioniemi OP, 1995, Genetic changes in primary reccurent prostate cancer by comparative genomic hybridization. Cancer Res 55:342–347 Tahara H, Smith AP, Gaz RD, Cryns VL, Arnold A, 1996, Genomic localization of novel candidate tumor suppressor gene loci in human parathyroid adenomas. Cancer Res 56:599– 605 Sebsebe L, Lea P, Markku S, Mika N, Sirkku S, Kirsti HP, Tom B, 2004, Loss of Heterozygosity at 6q is frequent and concurrent with 3p loss in sporadic and familial capillary hemangioblastomas. J Neuropathol Exp Neurol 63(10):1072– 1079 Inoue M, Marx A, Zettl A, Strobel P, Muller-Hermelink HK, Starostik P, 2002, Chromosome 6 suffers frequent and multiple aberrations in thymoma. Am J Pathol 161(4):1507–1513 Mazurenko NN, Beliakov IS, Bliyev AY, Guo Z, Hu X, Vinokourova SV, Bidzhieva BA, Pavlova LS, Ponten J, Kisseljov FL, 2003, Genetic alterations at chromosome 6 associated with cervical cancer progression. Mol Biol 37(3):404–411 Wan M, Sun T, Vyas R, Zheng J, Granada E, Dubeau L, 1999, Suppression of tumorigenicity in human ovarian cancer cell lines is controlled by a 2 cM fragment in chromosomal region 6q24– q25. Oncogene 18:1545–1551 Sheng ZM, Marchetti A, Buttitta F, Champeme MH, Campani D, Bistocchi M, Lidereau R, Callahan R, 1996, Multiple regions of chromosome 6q affected by loss of heterozygosity in primary human breast carcinomas. Br J Cancer 73:144–147 Colitti CV, Rodabaugh KJ, Welch WR, Berkowitz RS, Mok SC, 1998, A novel 4 cM minimal deletion unit on chromosome 6q25.1-q25.2 associated with high grade invasive epithelial ovarian carcinoma. Oncogene 16:555–559 Tibiletti MG, Bernasconi B, Furlan D, Riva C, Trubia M, Buraggi G, Franchi M, Bolis P, Mariani A, Frigerio L, Capella C, Taramelli R, 1996, Early involvement of 6q in surface epithelial ovarian tumors. Cancer Res 56:4493–4498 Cooke IE, Shelling AN, Le Meuth VG, Charnock ML, Ganesan TS, 1996, Allele loss on chromosome arm 6q and fine mapping of the region at 6q27 in epithelial ovarian cancer. Genes Chromosom Cancer 15:223–233 Tibiletti MG, Trubia M, Ponti E, Sessa L, Acquati F, Furlan D, Bernasconi B, Fichera M, Mihalich A, Ziegler A, Volz A, Facco C, 1998, Physical map of the D6S149–D6S193 region on chromosome 6Q27 and its involvement in benign surface epithelial ovarian tumours. Oncogene 16:1639–1642 Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N, 1998, Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature, Lond, 392:605–608 Cesari R, Martin ES, Calin GA, Pentimalli F, Bichi R, McAdams H, Trapasso F, Drusco A, Shimizu M, Masciullo V, d’Andrilli G, Scambia G, Picchio MC, Alder H, Godwin AK, Croce CM, 2003, Parkin, a gene implicated in autosomal recessive juvenile parkinsonism, is a candidate tumor suppressor gene on chromosome 6q25–q27. Proc Natl Acad Sci USA 100:5956–5961 Denison SR, Callahan G, Becker NA, Phillips LA, Smith DI, 2003, Characterization of FRA6E and its potential role in autosomal recessive juvenile parkinsonism and ovarian cancer. Genes Chromosom Cancer 38:40–52 West A, Periquet M, Lincoln S, Lucking CB, Nicholl D, Bonifati V, Rawal N, Gasser T, Lohmann E, Hardy J, Brice A, Farrer M, 2002, Complex relationship between Parkin mutations and Parkinson disease. Am J Med Genet 114:584–591 Picchio MC, Martin ES, Cesari R, Calin GA, Yendamuri S, Kuroki T, Pentimalli F, Sarti M, Yoder K, Kaiser LR, Fishel R, Croce CM, 2004, Alterations of the tumor suppressor gene Parkin in non-small cell lung cancer. Clin Cancer Res 10:2720– 2724 Mehdi SJ, Alam MS, Batra S, Rizvi MMA, 2010, Allelic loss of 6q25-27, the PARKIN tumor suppressor gene locus, in cervical carcinoma. Medical Oncology, in press)., DOI 10.1007/s/12032- 010-9633-x Denison SR, Wang F, Becker NA, Schule B, Kock N, Phillips LA, Klein C, Smith DI, 2003, Genetic alteration in the common fragile site gene PARK2 in ovarian and other cancers. Oncogene 22:8370–8378 Wang F, Denison S, Lai JP, Philips LA, Montoya D, Kock N, Schule B, Klein C, Shridhar V, Roberts LR, Smith DI, 2004, Parkin gene alterations in hepatocellular carcinoma. Genes Chromosom Cancer 40:85–96 Veeriah S, Taylor BS, Meng S, Fang F, Yilmaz E, Vivanco I, Janakiraman M, Schultz N, Hanrahan AJ, Pao W, Ladanyi M, Sander C, Heguy A, Holland EC, Paty PB, Mischel PS, Liau L, Cloughesy TF, Mellinghoff IK, Solit DB, Chan TA, 2010, Somatic mutations of the Parkinson’s disease-associated gene PARK2 in glioblastoma and other human malignancies. Nat Genet 42(1):77–82 Benedet JL, Bender H, Jones H III, Ngan HYS, Pecorelli S, 2000, FIGO staging classifications and clinical practice guidelines in the management of gynecologic cancers. Int J Gynecol Obstet 70:209–262 Sambrook J, Russell DW, 2001, Molecular cloning: a laboratory manual, 3rd edn. Cold Spring Harbor Lab, Plainview Yuping X, Lili M, Chengbo H, Jinyi L, Lei X, Yan X, 2007, MSI/ LOH and extron expression of the FHIT gene in gastric carcinoma. Front Med China 1(1):99–103 Preacher KJ, 2001, Calculation for the chi-square test: an interactive calculation tool for chi-square tests of goodness of fit and independence [Computer software]. Available from http:// www.quantpsy.org Foulkes WD, Ragoussis J, Stamp GWH, Allan GJ, Trowsdale J, 1993, Frequent loss of heterozygosity on chromosome 6 in human ovarian carcinoma. Br J Cancer 67:551–559 WanMZT, D’Abling G, Zheng I, VeliscuM, Dubeau L, 1994, Three distinct regions of chromosome 6 are targets of loss of heterozygosity in human ovarian carcinomas. J Oncol 5:1043–1048 Agirre X, Roman-Gomez J, Vazquez I, Jimenez-Velasco A, Garate L, Montiel-Duarte C, Artieda P, Cordeu L, Lahortiga I, Calasanz MJ, Heiniger A, Torres A, Minna JD, Prosper F, 2006, Abnormal methylation of the common PARK2 and PACRG promoter is associated with downregulation of gene expression in acute lymphoblastic leukemia and chronic myeloid leukemia. Int J Cancer 118:1945–1953 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 579 EP 586 DI 10.1007/s12253-010-9351-x PG 8 ER PT J AU Cui, Q Jiang, W Guo, J Liu, Ch Li, D Wang, X Zeng, Y AF Cui, Qiu Jiang, Weihao Guo, Jun Liu, Cheng Li, Dingfeng Wang, Xiaohong Zeng, Yanjun TI Relationship Between Hypermethylated MGMT Gene and Osteosarcoma Necrosis Rate After Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; DNA methylation; CpG island; Drug resistance; Neoplasm; O6-Methylguanine-DNA Methyltransferase ID Osteosarcoma; DNA methylation; CpG island; Drug resistance; Neoplasm; O6-Methylguanine-DNA Methyltransferase AB To investigate the relativity of MGMT(O-6-methylguanine-DNA methyltransferase) gene methylation from patients with protein expression and osteosarcoma necrosis rate after chemotherapy. Fifty-one oteosarcoma tissues were collected, Methylation of MGMT gene promoter was detected by methylation-specific PCR method, and protein expression of MGMT was examined by immunohistochemistry procedure, the relationship between methylated MGMT gene expression and patients response to chemotherapy was analyzed. The positive ratio of methylation MGMT gene promoter in 51 patients was 23.5% (12 in 51). Negative percentage of protein expression of MGMT was 27.5% (14 in 51). It seemed that methylation of MGMT gene in osteosarcoma tissues had no evident relationship with the patient’s age, sexuality, and the size and type of neoplasms, etc. The necrosis rates of methylated MGMT of osteosarcoma (tumor grade from I to IV) were 0 (0/51), 3.9% (2/51), 5.9% (3/51), 13.7% (7/51), respectively. In contrast, the necrosis rates of unmethylated MGMT of osteosarcoma (tumor grade from I to IV) were 45.1% (23/51), 25.5% (13/51), 3.9% (2/51), 2.0% (1/51), respectively. It suggest that methylated and unmethylated MGMT gene of osteosarcoma have significant difference in protein expression. The unmethylated MGMT gene has higher positive protein expression (u=−4.92, P<0.001). Methylation of MGMT gene has higher tumor necrosis rate in osteosarcoma patients. Methylation in MGMT promoter may be important for judging the effect of chemotherapy in Osteosarcoma patients. C1 [Cui, Qiu] 307 Hospital of PLA, Department of Orthopedics, PLA 307th Hospital, 100071 Beijing, China. [Jiang, Weihao] 307 Hospital of PLA, Department of Orthopedics, PLA 307th Hospital, 100071 Beijing, China. [Guo, Jun] 307 Hospital of PLA, Department of Orthopedics, PLA 307th Hospital, 100071 Beijing, China. [Liu, Cheng] 307 Hospital of PLA, Department of Orthopedics, PLA 307th Hospital, 100071 Beijing, China. [Li, Dingfeng] 307 Hospital of PLA, Department of Orthopedics, PLA 307th Hospital, 100071 Beijing, China. [Wang, Xiaohong] Beijing University of Technology, Biomechanics and Medical Information Institute, 100022 Beijing, China. [Zeng, Yanjun] Beijing University of Technology, Biomechanics and Medical Information Institute, 100022 Beijing, China. RP Li, D (reprint author), 307 Hospital of PLA, Department of Orthopedics, 100071 Beijing, China. EM 307yygk@sina.com CR Sabharwal A, Middleton MR, 2006, Exploiting the role of O6- methylguanine DNA Methyltransferase, MGMT, in cancer therapy[J]. Curr Opin Pharmacol 6(4):355–363 Kaina B, ChristmannM, Naumann S, Roos WP, 2007, MGMT: key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents[J]. DNA Repair, Amst, 6(8):1079–99 Jacinto FV, Esteller M, 2007, MGMT hypermethylation: a prognostic foe, a predictive friend[J]. DNA Rep 10(1):10–16 Helleday T, Petermann E, Lundin C et al, 2008, DNA repair pathways as targets for cancer therapy[J]. Nat Rev 8(3):193–204 Rosen G, Caparros B, Huvos AG et al, 1982, Preoperative chemotherapy for osteogenic sarcoma: selection of Postoperativeadjuvant chemotherapy based on the response of the primary tumor to preoperative chemotherapy [J]. Cancer 49(6):1221–1230 Lee S, Kim WH, Jung HY et al, 2002, Aberrant CpG island methylation of multiple genes in intrahepatic cholangiocarcinoma [J]. Am J Pathol 161(3):1015–1022 Ho JW, 2006, Potential and cytotoxicity of cis-Platinum complex with anti-tumor activity in combination therapy[J]. Recent Patents on Anti-Cancer Drug Discovery 1:129–134 Sabharwal A, Middleton MR, 2006, Exploiting the role of O6- methylguanine DNA methyltransferase, MGMT, in cancer therapy[ J]. Curr Opin Pharmascol 6(4):355–363 Verbeek B, Southgate TD, Gilham DE, Margison GP, 2008, O6- Methylguanine-DNA methyltransferase inactivation and chemotherapy[ J]. Br Med Bull 85:17–33 Voelter V, Diserens AC, Moulin A et al, 2008, Infrequent promoter methylation of the MGMT gene in liver metastases from uveal melanoma[J]. Int J Cancer 123(5):1215–8 Gal-Yan EN, Saito Y, Egger G et al, 2008, Cancer epigenetics: modifications, screening, and therapy[J]. Annu Rev Med 59:267– 280 Weisenberger DJ, Siegmund KD, Campan M et al, 2006, CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 38:787–793 Kawaguchi K, Oda Y, Saito T et al, 2006, DNA hypermethylation status of multiple genes in soft tissue sarcomas[J]. Mod Pathol 19, 1):106–114 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 587 EP 591 DI 10.1007/s12253-010-9354-7 PG 5 ER PT J AU Beketic-Oreskovic, L Ozretic, P Rabbani, NZ Jackson, LI Sarcevic, B Levanat, S Maric, P Babic, I Vujaskovic, Z AF Beketic-Oreskovic, Lidija Ozretic, Petar Rabbani, N Zahid Jackson, L Isabel Sarcevic, Bozena Levanat, Sonja Maric, Petra Babic, Ivan Vujaskovic, Zeljko TI Prognostic Significance of Carbonic Anhydrase IX (CA-IX), Endoglin (CD105) and 8-hydroxy-2′-deoxyguanosine (8-OHdG) in Breast Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE 8-OHdG; Breast cancer; CA-IX; CD105; Survival analysis ID 8-OHdG; Breast cancer; CA-IX; CD105; Survival analysis AB The aimof this studywas to examine the prognostic significance of carbonic anhydrase IX (CA-IX), an endogenous marker for tumor hypoxia; endoglin (CD105), a proliferation-associated and hypoxia-inducible glycoprotein and 8-hydroxy-2′-deoxyguanosine (8-OHdG), an oxidative DNA lesion, in breast cancer patients. Immunohistochemical expressions of CA-IX, CD105 and 8-OHdG, analyzed on paraffin-embedded tumor tissues from forty female breast cancer patients, were used to assess their prognostic implication on overall survival (OS) and relapse-free survival (RFS). Patients with high CA-IX expression (above cut-off value) had a higher occurrence of relapse (P=0.002). High CA-IX expression was significantly associated with shorter RFS (P<0.001, hazard ratio (HR) 0.21) and shorter OS (P<0.001, HR 0.19). Lymph node negative patients with high CA-IX expression had worse RFS (P=0.031, HR 0.14) and OS (P=0.005, HR 0.05). Patients with grade I&II tumors and high CA-IX expression showed shorter RFS (P=0.028, HR 0.28) and OS (P=0.008, HR 0.20). Worse OS (P=0.046, HR 0.28) was found in subgroup of patients with grade II tumors and high CA-IX expression. Among all three markers, only high CA-IX expression was strong independent prognostic indicator for shorter OS (HR 4.14, 95% CI 1.28–13.35, P=0.018) and shorter RFS (HR 3.99, 95% CI 1.38–11.59, P=0.011). Elevated expression of CA-IX was an independent prognostic factor for decreased RFS and OS and a significant marker for tumor aggressiveness. CD105 had week prognostic value; whereas, 8-OHdG, in this study, did not provide sufficient evidence as a prognostic indicator in breast cancer patients. C1 [Beketic-Oreskovic, Lidija] University Hospital for Tumors, University of Zagreb, School of Medicine, Department of Radiation Oncology, Ilica 197, 10000 Zagreb, Croatia. [Ozretic, Petar] Rudjer Boskovic Institute, Department of Molecular MedicineZagreb, Croatia. [Rabbani, N Zahid] Duke University Medical Center, Department of Radiation OncologyDurham, NC, USA. [Jackson, L Isabel] Duke University Medical Center, Department of Radiation OncologyDurham, NC, USA. [Sarcevic, Bozena] University Hospital for Tumors, University of Zagreb, School of Medicine, Department of PathologyZagreb, Croatia. [Levanat, Sonja] Rudjer Boskovic Institute, Department of Molecular MedicineZagreb, Croatia. [Maric, Petra] University Hospital for Tumors, University of Zagreb, School of Medicine, Department of Radiation Oncology, Ilica 197, 10000 Zagreb, Croatia. [Babic, Ivan] University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Obstetrics and GynecologyZagreb, Croatia. [Vujaskovic, Zeljko] Duke University Medical Center, Department of Radiation OncologyDurham, NC, USA. RP Maric, P (reprint author), University Hospital for Tumors, University of Zagreb, School of Medicine, Department of Radiation Oncology, 10000 Zagreb, Croatia. EM lidija.beketic.oreskovic@zg.t-com.hr CR Jemal A, Siegel R, Ward E et al, 2008, Cancer statistics. CA Cancer J Clin 58:71–96 Fisher B, Bauer M, Wickerham DL et al, 1983, Relation of number of positive axillary nodes to the prognosis of patients with primary breast cancer. An NSABP update. Cancer 52:1551–1557 Davis BW, Gelber RD, Goldhirsch A, 1986, Prognostic significance of tumor grade in clinical trials of adjuvant therapy for breast cancer with axillary lymph node metastasis. Cancer 58:2662–2670 Koscielny S, Tubiana M, Le MG et al, 1984, Breast cancer: relationship between the size of the primary tumour and the probability of metastatic dissemination. Br J Cancer 49:709–715 Clark GM, McGuire WL, 1988, Steroid receptors and other prognostic factors in primary breast cancer. Semin Oncol 15:20– 25 Leek RD, Landers RJ, Harris AL et al, 1999, Necrosis correlates with high vascular density and focal macrophage infiltration in invasive carcinoma of the breast. Br J Cancer 79:991–995 Mueller-Klieser W, Freyer JP, Sutherland RM, 1983, Evidence for a major role of glucose in controlling development of necrosis in EMT6/Ro multicell tumor spheroids. Adv Exp Med Biol 159:487–495 Parliament MB, Franko AJ, Allalunis-Turner MJ et al, 1997, Anomalous patterns of nitroimidazole binding adjacent to necrosis in human glioma xenografts: possible role of decreased oxygen consumption. Br J Cancer 75:311–318 Ramanujan S, Koenig GC, Padera TP et al, 2000, Local imbalance of proangiogenic and antiangiogenic factors: a potential mechanism of focal necrosis and dormancy in tumors. Cancer Res 60:1442–1448 Ruan K, Song G, Ouyang G, 2009, Role of hypoxia in the hallmarks of human cancer. J Cell Biochem 107:1053–1062 Vaupel P, Schlenger K, Knoop C et al, 1991, Oxygenation of human tumors: evaluation of tissue oxygen distribution in breast cancers by computerized O2 tension measurements. Cancer Res 51:3316–3322 Hockel M, Vaupel P, 2001, Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 93:266–276 Harris AL, 2002, Hypoxia–a key regulatory factor in tumour growth. Nat Rev Cancer 2:38–47 Shweiki D, Itin A, Soffer D et al, 1992, Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature 359:843–845 Maxwell PH, Dachs GU, Gleadle JM et al, 1997, Hypoxiainducible factor-1 modulates gene expression in solid tumors and influences both angiogenesis and tumor growth. Proc Natl Acad Sci USA 94:8104–8109 Zhong H, De Marzo AM, Laughner E et al, 1999, Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res 59:5830–5835 Wykoff CC, Beasley NJ, Watson PH et al, 2000, Hypoxiainducible expression of tumor-associated carbonic anhydrases. Cancer Res 60:7075–7083 Chia SK, Wykoff CC, Watson PH et al, 2001, Prognostic significance of a novel hypoxia-regulated marker, carbonic anhydrase IX, in invasive breast carcinoma. J Clin Oncol 19:3660–3668 Span PN, Bussink J, Manders P et al, 2003, Carbonic anhydrase-9 expression levels and prognosis in human breast cancer: association with treatment outcome. Br J Cancer 89:271–276 Hussain SA, Ganesan R, Reynolds G et al, 2007, Hypoxiaregulated carbonic anhydrase IX expression is associated with poor survival in patients with invasive breast cancer. Br J Cancer 96:104–109 Loncaster JA, Harris AL, Davidson SE et al, 2001, Carbonic anhydrase, CA IX, expression, a potential new intrinsic marker of hypoxia: correlations with tumor oxygen measurements and prognosis in locally advanced carcinoma of the cervix. Cancer Res 61:6394–6399 Olive PL, Aquino-Parsons C, MacPhail SH et al, 2001, Carbonic anhydrase 9 as an endogenous marker for hypoxic cells in cervical cancer. Cancer Res 61:8924–8929 Giatromanolaki A, Koukourakis MI, Sivridis E et al, 2001, Expression of hypoxia-inducible carbonic anhydrase-9 relates to angiogenic pathways and independently to poor outcome in nonsmall cell lung cancer. Cancer Res 61:7992–7998 Kim SJ, Rabbani ZN, Vollmer RT et al, 2004, Carbonic anhydrase IX in early-stage non-small cell lung cancer. Clin Cancer Res 10:7925–7933 Kim SJ, Rabbani ZN, Dewhirst MW et al, 2005, Expression of HIF-1alpha, CA IX, VEGF, and MMP-9 in surgically resected non-small cell lung cancer. Lung Cancer 49:325–335 Hui EP, Chan AT, Pezzella F et al, 2002, Coexpression of hypoxia-inducible factors 1alpha and 2alpha, carbonic anhydrase IX, and vascular endothelial growth factor in nasopharyngeal carcinoma and relationship to survival. Clin Cancer Res 8:2595– 2604 Liao SY, Aurelio ON, Jan K et al, 1997, Identification of the MN/ CA9 protein as a reliable diagnostic biomarker of clear cell carcinoma of the kidney. Cancer Res 57:2827–2831 McKiernan JM, Buttyan R, Bander NH et al, 1997, Expression of the tumor-associated gene MN: a potential biomarker for human renal cell carcinoma. Cancer Res 57:2362–2365 Colpaert CG, Vermeulen PB, Fox SB et al, 2003, The presence of a fibrotic focus in invasive breast carcinoma correlates with the expression of carbonic anhydrase IX and is a marker of hypoxia and poor prognosis. Breast Cancer Res Treat 81:137–147 Vleugel MM, Greijer AE, Shvarts A et al, 2005, Differential prognostic impact of hypoxia induced and diffuse HIF-1alpha expression in invasive breast cancer. J Clin Pathol 58:172–177 Kumar P, Wang JM, Bernabeu C, 1996, CD105 and angiogenesis. J Pathol 178:363–366 Fonsatti E, Del Vecchio L, Altomonte M et al, 2001, Endoglin: an accessory component of the TGF-beta-binding receptor-complex with diagnostic, prognostic, and bioimmunotherapeutic potential in human malignancies. J Cell Physiol 188:1–7 Kumar S, Ghellal A, Li C et al, 1999, Breast carcinoma: vascular density determined using CD105 antibody correlates with tumor prognosis. Cancer Res 59:856–861 Dales JP, Garcia S, Bonnier P et al, 2003, CD105 expression is a marker of high metastatic risk and poor outcome in breast carcinomas. Correlations between immunohistochemical analysis and long-term follow-up in a series of 929 patients. Am J Clin Pathol 119:374–380 Marnett LJ, 2000, Oxyradicals and DNA damage. Carcinogenesis 21:361–370 Wu LL, Chiou CC, Chang PY et al, 2004, Urinary 8-OHdG: a marker of oxidative stress to DNA and a risk factor for cancer, atherosclerosis and diabetics. Clin Chim Acta 339:1–9 Nagashima M, Tsuda H, Takenoshita S et al, 1995, 8- hydroxydeoxyguanosine levels in DNA of human breast cancer are not significantly different from those of non-cancerous breast tissues by the HPLC-ECD method. Cancer Lett 90:157–162 Musarrat J, Arezina-Wilson J, Wani AA, 1996, Prognostic and aetiological relevance of 8-hydroxyguanosine in human breast carcinogenesis. Eur J Cancer 32A:1209–1214 Kuo HW, Chou SY, Hu TW et al, 2007, Urinary 8-hydroxy-2′- deoxyguanosine, 8-OHdG, and genetic polymorphisms in breast cancer patients. Mutat Res 631:62–68 Himmetoglu S, Dincer Y, Ersoy YE et al, 2009, DNA oxidation and antioxidant status in breast cancer. J Investig Med 57:720–723 Karihtala P, Winqvist R, Syvaoja JE et al, 2006, Increasing oxidative damage and loss of mismatch repair enzymes during breast carcinogenesis. Eur J Cancer 42:2653–2659 Wang X, Ryter SW, Dai C et al, 2003, Necrotic cell death in response to oxidant stress involves the activation of the apoptogenic caspase-8/bid pathway. J Biol Chem 278:29184– 29191 Liao SY, Darcy KM, Randall LM et al, 2010, Prognostic relevance of carbonic anhydrase-IX in high-risk, early-stage cervical cancer: a Gynecologic Oncology Group study. Gynecol Oncol 116:452–458 Chandel NS, McClintock DS, Feliciano CE et al, 2000, Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1alpha during hypoxia: a mechanism of O2 sensing. J Biol Chem 275:25130–25138 Klimova T, Chandel NS, 2008, Mitochondrial complex III regulates hypoxic activation of HIF. Cell Death Differ 15:660–666 Fitzgibbons PL, Page DL, Weaver D et al, 2000, Prognostic factors in breast cancer. College of American Pathologists Consensus Statement 1999. Arch Pathol Lab Med 124:966–978 Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term followup. Histopathology 19:403–410 Elledge RM, McGuire WL, 1993, Prognostic factors and therapeutic decisions in axillary node-negative breast cancer. Annu Rev Med 44:201–210 Rosen PR, Groshen S, Saigo PE et al, 1989, A long-term followup study of survival in stage I, T1N0M0, and stage II, T1N1M0, breast carcinoma. J Clin Oncol 7:355–366 Honda M, Yamada Y, Tomonaga M et al, 2000, Correlation of urinary 8-hydroxy-2′-deoxyguanosine, 8-OHdG), a biomarker of oxidative DNA damage, and clinical features of hematological disorders: a pilot study. Leuk Res 24:461–468 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 593 EP 603 DI 10.1007/s12253-010-9355-6 PG 11 ER PT J AU Matsumoto, N Umezawa, T Sasaki, T Nakajima, K Kanetsuna, Y Sasaki, H AF Matsumoto, Naoki Umezawa, Takashi Sasaki, Toru Nakajima, Kuninobu Kanetsuna, Yukiko Sasaki, Hiroshi TI Clinical and Prognostic Value of the Presence of Irregular Giant Nuclear Cells in pT1 Ovarian Clear Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intraoperative diagnosis; Nuclear morphometry; Ovarian cancer; Pathology; Touch imprint cytology ID Intraoperative diagnosis; Nuclear morphometry; Ovarian cancer; Pathology; Touch imprint cytology AB In the early stages of epithelial ovarian cancer, histopathological grading is important. However, the grading of ovarian clear cell carcinoma (OCCC) remains controversial. We aimed to identify irregular giant nuclear cells (IGNCs) by a simple method in clinical practice, and to evaluate the prognostic value of IGNCs in pT1 OCCC. Eighty-seven pT1 OCCC patients who underwent initial surgery at Jikei University Kashiwa Hospital, Chiba, Japan, were retrospectively assessed. Paraffin-embedded tissue sections (PTSs) stained with hematoxylin and eosin were reviewed. Giant nuclear cells (GNCs) were defined as cells with a nuclear length of more than twice the median nuclear length. GNCs with irregular nuclear circumferences were defined as IGNCs. Cases where one or more GNCs existed and where IGNCs accounted for >10% of the GNCs were classified as IGNC-positive. We also attempted to identify IGNCs on touch imprint cytology smears (TICSs). Among the 87 cases, 68 were IGNC-negative and 19 were IGNC-positive. The 5-year disease-free and overall survival rates were 88.9% and 90.3% in the total patients, 98.3% and 100% in the IGNC-negative group, and 59.7% and 62.0% in the IGNC-positive group, respectively. These survival rates were significantly lower in the IGNC-positive group than in the IGNC-negative group (adjusted hazard ratio=14, 95% confidence interval=2.7–124 and adjusted hazard ratio=25, 95% confidence interval=2.9–768, respectively). Prognostic differences were not identified for other factors. IGNC identification on 28 available TICSs predicted IGNC identification on PTSs (sensitivity=50.0%, specificity=100%, P=0.007). The presence of IGNCs has clinical and prognostic value for pT1 OCCC. C1 [Matsumoto, Naoki] Jikei University Kashiwa Hospital, Department of Obstetrics and Gynecology, 163-1 Kashiwashita, 277-8567 Kashiwa, Chiba, Japan. [Umezawa, Takashi] Jikei University School of Medicine, Department of PathologyTokyo, Japan. [Sasaki, Toru] Tokyo Medical University, Department of Obstetrics and GynecologyTokyo, Japan. [Nakajima, Kuninobu] Jikei University School of Medicine, Department of Obstetrics and GynecologyTokyo, Japan. [Kanetsuna, Yukiko] Jikei University Kashiwa Hospital, Department of Clinical PathologyKashiwa, Chiba, Japan. [Sasaki, Hiroshi] Jikei University Kashiwa Hospital, Department of Obstetrics and Gynecology, 163-1 Kashiwashita, 277-8567 Kashiwa, Chiba, Japan. RP Matsumoto, N (reprint author), Jikei University Kashiwa Hospital, Department of Obstetrics and Gynecology, 277-8567 Kashiwa, Japan. EM nmazmoto@my.home.ne.jp CR Serov SF, Scully RE, Sobin LH, 1973, Histological typing of ovarian tumors. In: International histological classification of tumors, no.9. Geneva: World Health Organization, p37–42 Chan JK, Teoh D, Hu JM et al, 2008, Do clear cell ovarian carcinomas have poorer prognosis compared to other epithelial cell types? A study of 1411 clear cell ovarian cancers. Gynecol Oncol 109:370–376 Konishi I, 2008, Gynecologic oncology committee report [written in Japanese]. Acta Obstet Gynaecol Jpn 60:1001–1085 Lee KR, Tavassoli FA, Prat J et al, 2003, Surface epithelialstromal tumors. In: Tavassoli FA, Devilee P, eds, WHO classification of tumours. IARC, Lyon, pp 117–145 Liu CQ, Sasaki H, Fahey MT et al, 1999, Prognostic value of nuclear morphometry in patients with TNM stage T1 ovarian clear cell adenocarcinoma. Br J Cancer 79:1736–1741 Young RC, Walton LA, Ellenberg SS et al, 1990, Adjuvant therapy in stage I and stage II epithelial ovarian cancer. Results of two prospective randomized trials. N Engl J Med 322:1021–1027 Trimbos JB, Schueler JA, van der Burg M et al, 1991, Watch and wait after careful surgical treatment and staging in welldifferentiated early ovarian cancer. Cancer 67:597–602 Monga M, Carmichael JA, ShelleyWE et al, 1991, Surgery without adjuvant chemotherapy for early epithelial ovarian carcinoma after comprehensive surgical staging. Gynecol Oncol 43:195–197 Acta Obstet Gynecol Scand, 1971, Classification and staging of malignant tumours in the female pelvis. Acta Obstet Gynecol Scand 50:1–7 Silverberg SG, 2000, Histopathologic grading of ovarian carcinoma: a review and proposal. Int J Gynecol Pathol 19:7–15 Malpica A, 2008, Grading of ovarian cancer: a histotype-specific approach. Int J Gynecol Pathol 27:175–181 Bookman MA, Greer BE, Ozols RF, 2003, Optimal therapy of advanced ovarian cancer: carboplatin and paclitaxel vs. cisplatin and paclitaxel, GOG 158, and an update on GOG0 182-ICON5. Int J Gynecol Cancer 13:735–740 Utsunomiya H, Akahira J, Tanno S et al, 2006, Paclitaxelplatinum combination chemotherapy for advanced or recurrent ovarian clear cell adenocarcinoma: a multicenter trial. Int J Gynecol Cancer 16:52–56 Sirichaisutdhikorn D, Suprasert P, Khunamornpong S, 2009, Clinical outcome of the ovarian clear cell carcinoma compared to other epithelial ovarian cancers when treated with paclitaxel and carboplatin. Asian Pac J Cancer Prev 10:1041–1045 Sugiyama T, Kamura T, Kigawa J et al, 2000, Clinical characteristics of clear cell carcinoma of the ovary: a distinct histologic type with poor prognosis and resistance to platinum-based chemotherapy. Cancer 88:2584–2589 Maggioni A, Benedetti PP, Dell’Anna T et al, 2006, Randomised study of systematic lymphadenectomy in patients with epithelial ovarian cancer macroscopically confined to the pelvis. Br J Cancer 95:699–704 Chan JK, Munro EG, Cheung MK et al, 2007, Association of lymphadenectomy and survival in stage I ovarian cancer patients. Obstet Gynecol 109:12–19 Suzuki S, Kajiyama H, Shibata K et al, 2008, Is there any association between retroperitoneal lymphadenectomy and survival benefit in ovarian clear cell carcinoma patients? Ann Oncol 19:1284–1287 Sakuragi N, Yamada H, Oikawa M et al, 2000, Prognostic significance of lymph node metastasis and clear cell histology in ovarian carcinoma limited to the pelvis, pT1M0 and pT2M0). Gynecol Oncol 79:251–255 Takano M, Kikuchi Y, Yaegashi N et al, 2006, Clear cell carcinoma of the ovary: a retrospective multicentre experience of 254 patients with complete surgical staging. Br J Cancer 94:1369– 1374 Ushijima K, 2007, Management of retroperitoneal lymph nodes in the treatment of ovarian cancer. Int J Clin Oncol 12:181–186 Geomini P, Bremer G, Kruitwagen R et al, 2005, Diagnostic accuracy of frozen section diagnosis of the adnexal mass: a metaanalysis. Gynecol Oncol 96:1–9 Stewart CJ, Brennan BA, Hammond IG et al, 2008, Intraoperative assessment of clear cell carcinoma of the ovary. Int J Gynecol Pathol 27:475–482 Vrdoljak-Mozetic D, Stankovic T, Krasevic M et al, 2006, Intraoperative cytology of clear cell carcinoma of the ovary. Cytopathology 17:390–395 Fuhrman SA, Lasky LC, Limas C, 1982, Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol 6:655–663 Ficarra V, Martignoni G, Maffei N et al, 2005, Original and reviewed nuclear grading according to the Fuhrman system. Cancer 103:68–75 Rioux-Leclercq N, Karakiewicz PI, Trinh QD et al, 2007, Prognostic ability of simplified nuclear grading of renal cell carcinoma. Cancer 109:868–874 Novara G, Martignoni G, Artibani Wet al, 2007, Grading systems in renal cell carcinoma. J Urol 177:430–436 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 605 EP 611 DI 10.1007/s12253-010-9356-5 PG 7 ER PT J AU Chen, CHA Waterboer, T Keleher, A Morrison, B Jindal, Sh McMillan, D Nicol, D Gardiner, AR McMillan, AJN Antonsson, A AF Chen, C-H Alice Waterboer, Tim Keleher, Annie Morrison, Beth Jindal, Shalini McMillan, Denise Nicol, David Gardiner, A Robert McMillan, A J Nigel Antonsson, Annika TI Human Papillomavirus in Benign Prostatic Hyperplasia and Prostatic Adenocarcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Benign prostatic hyperplasia (BPH); Human papillomavirus (HPV); Polymerase chain reaction (PCR); Prostate cancer; Serology ID Benign prostatic hyperplasia (BPH); Human papillomavirus (HPV); Polymerase chain reaction (PCR); Prostate cancer; Serology AB The aim of this study was to determine the prevalence of human papillomavirus (HPV) types in tissue and HPV antibodies in prostatic disease. Prostate tissue samples were collected from 51 patients diagnosed with adenocarcinoma and 11 with benign prostatic hyperplasia (BPH). All tissue samples were confirmed by histology. Plasma samples were available for 52 prostate patients. We investigated HPV DNA prevalence by PCR, and PCR positive samples were HPV type determined by sequencing. Prevalence of antibodies against twenty-seven HPV proteins from fourteen different HPV types was assessed in the plasma samples. The HPV DNA prevalence in the tissue samples was 14% (7/51) for prostate cancer samples and 27% (3/11) for BPHs. HPV-18 was the only type detected in tissue samples (10/62). No significant difference in HPV prevalence between the prostate cancer and BPH samples was found. HPV-positive cells were identified in eight of our thirteen prostate tissue slides (3/3 BPH and 5/10 adenocarcinoma) by in situ hybridisation, and the positive cells were found in epithelial cells and peripheral blood cells. Serology data showed no significant increase in levels of antibodies against any of the HPV-18 proteins tested for in prostatic disease patients. Antibodies against HPV-1, HPV-4, HPV-6 and HPV-11 were significantly higher in the group of males with prostatic disease. Our study did not show an association between prostatic disease and either presence of HPV DNA in samples or previous exposure of high-risk HPV. C1 [Chen, C-H Alice] The University of Queensland, Princess Alexandra Hospital, Diamantina Institute for Cancer, Immunology and Metabolic Medicine, 4102 Brisbane, QLD, Australia. [Waterboer, Tim] German Cancer Research Center (DKFZ), Infection and Cancer Program, 69120 Heidelberg, Germany. [Keleher, Annie] The University of Queensland, Princess Alexandra Hospital, Diamantina Institute for Cancer, Immunology and Metabolic Medicine, 4102 Brisbane, QLD, Australia. [Morrison, Beth] The University of Queensland, Princess Alexandra Hospital, Department of Urology, Central Clinical Division, 4102 Brisbane, QLD, Australia. [Jindal, Shalini] Dame Roma Mitchell Cancer Research Laboratories, 5000 Adelaide, SA, Australia. [McMillan, Denise] The University of Queensland, Princess Alexandra Hospital, Diamantina Institute for Cancer, Immunology and Metabolic Medicine, 4102 Brisbane, QLD, Australia. [Nicol, David] The University of Queensland, Princess Alexandra Hospital, Department of Urology, Central Clinical Division, 4102 Brisbane, QLD, Australia. [Gardiner, A Robert] The University of Queensland, Royal Brisbane Hospital, Department of Surgery, Central Clinical Division, 4029 Brisbane, QLD, Australia. [McMillan, A J Nigel] The University of Queensland, Princess Alexandra Hospital, Diamantina Institute for Cancer, Immunology and Metabolic Medicine, 4102 Brisbane, QLD, Australia. [Antonsson, Annika] The University of Queensland, Princess Alexandra Hospital, Diamantina Institute for Cancer, Immunology and Metabolic Medicine, 4102 Brisbane, QLD, Australia. RP Antonsson, A (reprint author), The University of Queensland, Princess Alexandra Hospital, Diamantina Institute for Cancer, Immunology and Metabolic Medicine, 4102 Brisbane, Australia. EM annika.antonsson@qimr.edu.au CR zur Hausen H, 1996, Papillomavirus infections-a major cause of human cancers. Biochim Biophys Acta 1288:F55–F78 Effert PJ, Frye RA, Neubauer A, Liu ET, Walther PJ, 1992, Human papillomavirus types 16 and 18 are not involved in human prostate carcinogenesis: analysis of archival human prostate cancer specimens by differential polymerase chain reaction. J Urol 147:192–196 Serth J, Panitz F, Paeslack U, Kuczyk MA, Jonas U, 1999, Increased levels of human papillomavirus type 16 DNA in a subset of prostate cancers. Cancer Res 59:823–825 Zambrano A, Kalantari M, Simoneau A, Jensen JL, Villarreal LP, 2002, Detection of human polyomaviruses and papillomaviruses in prostatic tissue reveals the prostate as a habitat for multiple viral infections. Prostate 53:263–276 McNicol PJ, Dodd JG, 1991, High prevalence of human papillomavirus in prostate tissues. J Urol 145:850–853 Andersson S, Rylander E, Larson B, Sigurdardottir S, Backlund I, Sallstrom J, Wilander E, 2003, Types of human papillomavirus revealed in cervical adenocarcinomas after DNA sequencing. Oncol Rep 10:175–179 Castellsague X, Diaz M, de Sanjose S, Munoz N, Herrero R, Franceschi S, Peeling RW, Ashley R, Smith JS, Snijders PJ, Meijer CJ, Bosch FX, 2006, Worldwide human papillomavirus etiology of cervical adenocarcinoma and its cofactors: implications for screening and prevention. J Natl Cancer Inst 98:303–315 Mohan H, Bal A, Punia RP, Bawa AS, 2003, Squamous cell carcinoma of the prostate. Int J Urol 10:114–116 Al-Maghrabi JA, 2007, The role of human papillomavirus infection in prostate cancer. Saudi Med J 28:326–333 Dodd JG, Paraskevas M, McNicol PJ, 1993, Detection of human papillomavirus 16 transcription in human prostate tissue. J Urol 149:400–402 Terris MK, Peehl DM, 1997, Human papillomavirus detection by polymerase chain reaction in benign and malignant prostate tissue is dependent on the primer set utilized. Urology 50:150–156 Ruijter E, van de Kaa C, Miller G, Ruiter D, Debruyne F, Schalken J, 1999, Molecular genetics and epidemiology of prostate carcinoma. Endocr Rev 20:22–45 Adami HO, Kuper H, Andersson SO, Bergstrom R, Dillner J, 2003, Prostate cancer risk and serologic evidence of human papilloma virus infection: a population-based case-control study. Cancer Epidemiol Biomark Prev 12:872–875 Rosenblatt KA, Carter JJ, Iwasaki LM, Galloway DA, Stanford JL, 2003, Serologic evidence of human papillomavirus 16 and 18 infections and risk of prostate cancer. Cancer Epidemiol Biomark Prev 12:763–768 Sutcliffe S, Viscidi RP, Till C, Goodman PJ, Hoque AM, Hsing AW, Thompson IM, Zenilman JM, De Marzo AM, Platz EA, 2010, Human papillomavirus types 16, 18, and 31 serostatus and prostate cancer risk in the Prostate Cancer Prevention Trial. Cancer Epidemiol Biomark Prev 19:614–618 Forslund O, Antonsson A, Nordin P, Stenquist B, Hansson BG, 1999, A broad range of human papillomavirus types detected with a general PCR method suitable for analysis of cutaneous tumours and normal skin. J Gen Virol 80:2437–2443 Waterboer T, Sehr P, Michael KM, Franceschi S, Nieland JD, Joos TO, TemplinMF, PawlitaM(2005)Multiplex human papillomavirus serology based on in situ-purified glutathione s-transferase fusion proteins. Clin Chem 51:1845–1853 Michael KM, Waterboer T, Sehr P, Rother A, Reidel U, Boeing H, Bravo IG, Schlehofer J, Gartner BC, Pawlita M, 2008, Seroprevalence of 34 human papillomavirus types in the German general population. PLoS Pathog 4:e1000091 Heideman DA, Waterboer T, Pawlita M, Delis-van Diemen P, Nindl I, Leijte JA, Bonfrer JM, Horenblas S, Meijer CJ, Snijders PJ, 2007, Human papillomavirus-16 is the predominant type etiologically involved in penile squamous cell carcinoma. J Clin Oncol 25:4550–4556 Chen AC-H, McMillan NAJ, Antonsson A, 2008, Human papillomavirus type spectrum in normal skin of individuals with or without a history of frequent sun exposure. J Gen Virol 89:2891–2897 Heng B, Glenn WK, Ye Y, Tran B, Delprado W, Lutze-Mann L, Whitaker NJ, Lawson JS, 2009, Human papilloma virus is associated with breast cancer. Br J Cancer 101:1345–1350 Kan CY, Iacopetta BJ, Lawson JS, Whitaker NJ, 2005, Identification of human papillomavirus DNA gene sequences in human breast cancer. Br J Cancer 93:946–948 Strickler HD, Goedert JJ, 2001, Sexual behavior and evidence for an infectious cause of prostate cancer. Epidemiol Rev 23:144–151 Huang WY, Hayes R, Pfeiffer R, Viscidi RP, Lee FK, Wang YF, Reding D, Whitby D, Papp JR, Rabkin CS, 2008, Sexually transmissible infections and prostate cancer risk. Cancer Epidemiol Biomark Prev 17:2374–2381 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 613 EP 617 DI 10.1007/s12253-010-9357-4 PG 5 ER PT J AU Vural, Uluoglu, Akyurek, N Oguz, A Karadeniz, C AF Vural, Cigdem Uluoglu, Omer Akyurek, Nalan Oguz, Aynur Karadeniz, Ceyda TI The Evaluation of CD99 Immunoreactivity and EWS/FLI1 Translocation by Fluorescence in situ Hybridization in Central PNETs and Ewing’s Sarcoma Family of Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ewing’s sarcoma family of tumors; Peripheral primitive neuroectodermal tumor; CD99; EWS-FLI1 translocation; Fluorescence in situ hybridization ID Ewing’s sarcoma family of tumors; Peripheral primitive neuroectodermal tumor; CD99; EWS-FLI1 translocation; Fluorescence in situ hybridization AB Ewing’s sarcoma family of tumors (ESFTs) are indicated by malignant, small, round and blue cell tumors of the bone and soft tissue. Gene rearrangements between EWS gene on chromosome 22q12 and members of the ETS gene family are common in and specific to ESFTs. Another defining characteristic of ESFTs is their membranous expression of the CD99. In contrast, such translocations and immunoreactivity are not found in central primitive neuroectodermal tumors (cPNETs). The aim of this study was to investigate the detection of EWS/FLI1 translocations and CD99 immunoreactivity in order to evaluate their clinicopathological features and their roles in the differential diagnosis of these tumors. In this study, we investigated CD99 immunoreactivity using immunohistochemistry and Ewing’s sarcoma / Friend leukaemia virus integration 1 (EWS/FLI1) translocation using the fluorescence in situ hybridization (FISH) method in 23 cases. CD99 expression was detected in 10/11 (90%) ESFT cases and 2/7 cPNET cases. In 18 cases EWS/FLI1 translocation was examined using the FISH method. The EWS/FLI1 translocations were detected in 7/8 (87.5%) ESFTs cases, whereas non of 8 cPNET cases were detected with this translocation. One case could not be classified as either central or peripheral, showed EWS/FLI1 translocation. There was a statistically significant difference in CD99 expression (p=0.0013) and EWS/FLI1 translocation (p=0,002) between cPNETs and ESFTs cases. In conclusion, CD99 expression and EWS/FLI1 translocation are specific and sensitive markers in the diagnosis of ESFTs. However, these were often not found in cases of cPNET. Therefore, in the diagnosis of ESFTs, clinical, radiological, histopathological and immunohistochemical parameters should always be evaluated together. C1 [Vural, Cigdem] Baskent University Faculty of Medicine, Department of PathologyAnkara, Turkey. [Uluoglu, Omer] Gazi University Faculty of Medicine, Department of PathologyAnkara, Turkey. [Akyurek, Nalan] Gazi University Faculty of Medicine, Department of PathologyAnkara, Turkey. [Oguz, Aynur] Gazi University Faculty of Medicine, Department of Pediatric OncologyAnkara, Turkey. [Karadeniz, Ceyda] Gazi University Faculty of Medicine, Department of Pediatric OncologyAnkara, Turkey. RP Vural, (reprint author), Baskent University Faculty of Medicine, Department of Pathology, Ankara, Turkey. EM dr.cvural@gmail.com CR Furuno Y, Nishimura S, Kamiyama H et al, 2008, Intracranial peripheral-type primitive neuroectodermal tumor-case report. Neurol Med Chir, Tokyo, 48(2):72–76 McLendon RE, Judkins AR, Eberhart CG et al, 2007, Central nervous system primitive neuroectodermal tumours. In: Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, eds, WHO classification of tumours of the central nervous system, 4th edn. IARC, Lyon Weiss SW, Goldblum JR, 2008, Ewing’s sarcoma/PNET tumor family and related lesions. In: Schmitt W, Black S, eds, Enzinger & weiss’s soft tissue tumors, 5th edn. Mosby Elsevier, Philadelphia Folpe AL, Goldblum JR, Rubin BP et al, 2005, Morphologic and immunophenotypic diversity in Ewing family tumors: a study of 66 genetically confirmed cases. Am J Surg Pathol 29(8):1025–1033 Kontny U, 2006, Regulation of apoptosis and proliferation in ewing’s sarcoma-oppotunities for targeted therapy. Hematol Oncol 24(1):14–21 O’Sullivan MJ, Perlman EJ, Furman J et al, 2001, Visceral primitive peripheral neuroectodermal tumors: a clinicopathologic and molecular study. Hum Pathol 32(10):1109–1115 Ushigome S, Machinami R, Sorensen PH, 2002, Chapter 14: Ewing sarcoma/primitive neuroectodermal tumor. In: Fletcher CDM, Unni KK, Mertens F, eds, World health organization classification of tumors: pathology& genetics of tumours of soft tissue and bone. IARC Press, Lyon Llombart-Bosch A, Navarro S, 2001, Immunohistochemical detection of EWS and FLI-1 proteins in ewing sarcoma and primitive neuroectodermal tumors: comparative analysis with CD99, MIC-2, expression. Appl Immunohistochem Mol Morphol 9(3):255–260 Mobley BC, Roulston D, Shah GV et al, 2006, Peripheral primitive neuroectodermal tumor/ewing’s sarcoma of the craniospinal vault: case reports and review. Hum Pathol 37(7):845–853 Khoury JD, 2005, Ewing sarcoma family of tumors. Adv Anat Pathol 12(4):212–220 Amiel A, Ohali A, Fejgin M et al, 2003, Molecular cytogenetic parameters in ewing sarcoma. Cancer Genet Cytogenet 140, 2):107–112 Folpe AL, Hill CE, Parham DM et al, 2000, Immunohistochemical detection of FLI-1 protein expression: a study of 132 round cell tumors with emphasis on CD99-positivemimics of ewing’s sarcoma/primitive neuroectodermal tumor. Am J Surg Pathol 24(12):1657–1662 Gardner LJ, Ayala AG, Monforte HL et al, 2004, Ewing sarcoma/ peripheral primitive neuroectodermal tumor adult abdominal tumors with an ewing sarcoma gene rearrangement demonstrated by fluorescence in situ hybridization in paraffin sections. Appl Immunohistochem Mol Morphol 12(2):160–165 Hadfield MG, Quezado MM, Williams RL et al, 2000, Ewing’s family of tumors involving structures related to the central nervous system: a review. Pediatr Dev Pathol 3(3):203–210 Mhawech-Fauceglia P, Hermann F, Penetrante R et al, 2006, Diagnostic utility of FLI-1 monoclonal antibody and dual-colour, break-apart probe fluorescence in situ, FISH, analysis in ewing’s sarcoma/primitive neuroectodermal tumour, EWS/PNET): a comparative study with CD99 and FLI-1 polyclonal antibodies. Histopathology 49(6):569–575 Mazur MA, Gururangan S, Bridge JA et al, 1999, Intracranial ewing sarcoma. Pediatr Blood Cancer 45(6):850–856 Cohn SL, 1999, Diagnosis and classification of the small round cell tumors of childhood. Am J Pathol 155(1):11–15 Devoe K, Weidner N, 2000, Immunohistochemistry of small round-cell tumors. Semin Diagn Pathol 17(3):216–224 Hasegawa SL, Davison JM, Rutten A et al, 1998, Primary cutaneous ewing’s sarcoma: immunophenotypic and molecular cytogenetic evaluation of five cases. Am J Surg Pathol 22(3):310– 318 Sheaff M, McManus A, Scheimberg I et al, 1997, Primitive neuroectodermal tumor of the kidney confirmed by fluorescence in situ hybridization. Am J Surg Pathol 21(4):461–468 Pekala JS, Gururangan S, Provenzale JM et al, 2006, Central nervous system extraosseous ewing sarcoma: radiologic manifestations of this newly defined pathologic entity. Am J Neuroradiol 27(3):580–583 Kazmi SA, Perry A, Pressey JG et al, 2007, Primary ewing sarcoma of the brain a case report and literature review. Diagn Mol Pathol 16(2):108–111 Kampman WA, Kros JM, De Jong THR et al, 2006, Primitive neuroectodermal tumors, PNETs, located in the spinal canal: the relevance of classification as central or peripheral PNET, case report of a primary spinal PNET occurence with a critical literature review. J Neurooncol 77(1):65–72 D’ Antonio A, Caleo A, Garcia JF et al, 2004, Primary peripheral PNET/ewing’s sarcoma of the dura with FISH analysis. Histopathology 45(6):642–656 Ishii N, Hiraga H, Sawamura Y et al, 2001, Alternative EWSFLI1 fusion gene and MIC2 expression in peripheral and central primitive neuroectodermal tumors. Neuropathology 21(1):40–44 De Alava E, Gerald WL, 2000, Molecular biology of the ewing’s sarcoma/primitive neuroectodermal tumor family. J Clin Oncol 18, 1):204–213 Rossi S, Orvieto E, Furlanetto A et al, 2004, Utility of the immunohistochemical detection of FLI-1 expression in round cell and vascular neoplasm using a monoclonal antibody. Mod Pathol 17(5):547–552 Thorner P, Squire J, Chilton-MacNeill S et al, 1996, Is the EWS/ FLI-1 fusion transcript specific for ewing sarcoma and peripheral primitive neuroectodermal tumor?: a report of four cases showing this transcript in a wider range of tumor types. Am J Pathol 148, 4):1125–1138 Park YK, Chi SG, Park HR et al, 1998, Detection of t(11;22)(q24; q12, translocation of ewing’s sarcoma in paraffin embedded tissue by nested reverse transcription- polymerase chain reaction. J Korean Med Sci 13(4):395–399 Ginsberg JP, De Alava E, Ladanyi M et al, 1999, EWS-FLI1 and EWS-ERG gene fusions are associated with similar clinical phenotypes in ewing’s sarcoma. J Clin Oncol 17(6):1809–1814 Qian X, Jin L, Shearer BM et al, 2005, Molecular diagnosis of ewing’s sarcoma/primitive neuroectodermal tumor in formalinfixed paraffin-embedded tissues by RT-PCR and fluorescence in situ hybridization. Diagn Mol Pathol 14(1):23–28 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 619 EP 625 DI 10.1007/s12253-010-9358-3 PG 7 ER PT J AU Lu, L Zhao, G Luu-The, V Ouellet, J Fan, Z Labrie, F Pelletier, G AF Lu, Lu Zhao, Gang Luu-The, Van Ouellet, Johanne Fan, Zhinmin Labrie, Fernand Pelletier, Georges TI Expression of 11β-hydroxysteroid Dehydrogenase Type 1 in Breast Cancer and Adjacent Non-Malignant Tissue. An Immunocytochemical Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE 11β-hydroxysteroid dehydrogenase type 1; Breast cancer; Glucocorticoid receptors; Immunocytochemistry; Cortisol ID 11β-hydroxysteroid dehydrogenase type 1; Breast cancer; Glucocorticoid receptors; Immunocytochemistry; Cortisol AB Intratumoral biosynthesis of hormone steroids is thought to play a role in the pathogenesis and development of human breast cancer. There is evidence that glucocorticoids may inhibit the development and progression of breast cancer. 11β-hydroxysteroid dehydrogenase (11β-HSD) type 1 is the enzyme which converts inactive cortisone to active cortisol. In order to study the expression of 11β-HSD type 1 in breast cancer and non-cancerous breast tissue, we have developed specific antibodies to 11β-HSD type 1 and proceeded to localization of the enzyme in 84 specimens of breast carcinoma and adjacent nonmalignant tissues by immnohistochemistry. The results were correlated with the expression of androgen receptor, estrogen receptor, progesterone receptor, glucocorticoid receptor and CDC47, a cell division marker, as well as the tumor stage, tumor size, nodal status and menopausal status. The expression of 11β-HSD type 1 in 64% of breast cancer specimens appeared significantly lower than that observed in normal adjacent tissues (97% of cases being positive). There was no significant correlation between 11β-HSD type 1 expression and the clinicopathological parameters studied. The decrease in 11β-HSD type 1 expression in breast cancer as compared to that observed in the adjacent normal tissues may play a role in the development and/or progression of the cancer by modifying the intratumoral levels of glucocorticoids. C1 [Lu, Lu] Laval University Hospital Research Center, Molecular Endocrinology and Oncology Research Center, 2705 Laurier blvd, G1V 4G2 Quebec, QC, Canada. [Zhao, Gang] Laval University Hospital Research Center, Molecular Endocrinology and Oncology Research Center, 2705 Laurier blvd, G1V 4G2 Quebec, QC, Canada. [Luu-The, Van] Laval University Hospital Research Center, Molecular Endocrinology and Oncology Research Center, 2705 Laurier blvd, G1V 4G2 Quebec, QC, Canada. [Ouellet, Johanne] Laval University Hospital Research Center, Molecular Endocrinology and Oncology Research Center, 2705 Laurier blvd, G1V 4G2 Quebec, QC, Canada. [Fan, Zhinmin] Jilin University, China-Japan Union Hospital, No74, Xin Min Street, 130021 Changchun, Jilin Province, China. [Labrie, Fernand] Laval University Hospital Research Center, Molecular Endocrinology and Oncology Research Center, 2705 Laurier blvd, G1V 4G2 Quebec, QC, Canada. [Pelletier, Georges] Laval University Hospital Research Center, Molecular Endocrinology and Oncology Research Center, 2705 Laurier blvd, G1V 4G2 Quebec, QC, Canada. RP Pelletier, G (reprint author), Laval University Hospital Research Center, Molecular Endocrinology and Oncology Research Center, G1V 4G2 Quebec, Canada. EM georges.pelletier@crchul.ulaval.ca CR Allegra JC, Lippman ME, Thompson EB, Simon R, Barlock A, Green L, Huff KK, Do HM, Aitken SC, 1979, Distribution, frequency, and quantitative analysis of estrogen, progesterone, androgen, and glucocorticoid receptors in human breast cancer. Cancer Res 39(5):1447–1454 Chapman KE, Kotelvtsev YV, Jamieson PM, Williams LJ, Mullins JJ, Seckl JR, 1997, Tissue-specific modulation of glucocorticoid action by the 11 beta-hydroxysteroid dehydrogenases. Biochem Soc Trans 25(2):583–587 Darcy KM, Shoemaker SF, Lee PP, Ganis BA, Ip MM, 1995, Hydrocortisone and progesterone regulation of the proliferation, morphogenesis, and functional differentiation of normal rat mammary epithelial cells in three dimensional primary culture. J Cell Physiol 163:365–379 Feigelson HS, Teras LR, Diver WR, Tang W, Patel AV, Stevens VL, Calle EE, Thun MJ, Bouzyk M, 2008, Genetic variation in candidate obesity genes ADRB2, ADRB3, GHRL, HSD11B1, IRS1, IRS2 and SHC1 and risk for breast cancer in the Cancer Prevention Study II. Breast Cancer Res 10(4):1–11 Gong H, Jarzynka MJ, Cole TJ, Lee JH, Wada T, Zhang B, Gao J, Song WC, DeFranco DB, Cheng SY, Xie W, 2008, Glucocorticoids antagonize estrogens by glucocorticoid receptor-mediated activation of estrogen sulfotransferase. Cancer Res 68(18):7386– 7393 Goya L, Maiyar AC, Ge Y, Firestone GL, 1993, Glucocorticoids induce a G1/G0 cell cycle arrest of Con8 rat mammary tumor cells that is synchronously reversed by steroid withdrawal or addition of transforming growth factor-alpha. Mol Endocrinol 7:1121–1132 Heuson JC, 1974, Hormones by administration. In: Atkins H, ed, The treatment of breast cancer. University Park Press, Baltimore, pp 140–146 Hundermark S, Buler H, Rudolf M, Weitzel HK, Ragosch V, 1997, Inhibition of 11 beta-hydroxysteroid dehydrogenase activity enhances the antiproliferative effect of glucocorticoids on MCF-7 and ZR-75-1 breast cancer cells. J Endocrinol 155, 1):181–180 Keith BD, 2008, Systematic review of the clinical effect of glucocorticoids on nonhematologic malignancy. BMC Cancer 8:84–103 Kotelevtsev Y, Holmes MC, Burchell A, Houston PM, Schmoll D, Jamieson P, Best R, Brown R, Edwards CR, Seckl JR, Mullins JJ, 1997, 11beta-hydroxysteroid dehydrogenase type 1 knockout mice show attenuated glucocorticoid-inducible responses and resist hyperglycemia on obesity or stress. Proc Natl Acad Sci USA 94:14924–14929 Krozowski Z, 1999, The 11β-hydroxysteroid dehydrogenases: functions and physiological effects. Mol Cell Endocrinol 151:121–127 Lien HC, Lu YS, Cheng AL, Chang WC, Jeng YM, Kuo YH, Huang CS, Chang KJ, Yao YT, 2006, Differential expression of glucocorticoid receptor in human beast tissues and related neoplasms. J Pathol 209:317–327 Lippman M, Bolan G, Hu K, 1976, The effects of glucocorticoids and progesterone on hormone-responsive human breast cancer in long-term tissue culture. Cancer Res 36:4602–4609 Lyons WR, 1958, Hormonal synergism in mammary growth. Proc R Soc Lond B Biol Sci 149:303–325 Osborne CK, Monaco ME, Kahn CR, Hu K, Bonzert D, Lippman ME, 1979, Direct inhibition of growth and antagonism of insulin action by glucocorticoids in human breast cancer cells in culture. Cancer Res 39:2422–2428 Pang D, Kocherginsky M, Drausz T, Kim SY, Conzen SD, 2006, Dexamethasone decreases xenograft response to Paclitaxel through inhibityion of tumor cell apoptosis. Cancer Biol Ther 5(8):941–942 Pasqualini JR, 2004, The selective estrogen enzyme modulators in breast cancer: a review. Biophys Acta 1654(2):123–143 Pelletier G, Luu-The V, Li S, Bujold G, Labrie F, 2007, Localization and glucocorticoid regulation of 11β-hydroxysteroid dehydrogenase type 1 mRNA in the male mouse forebrain. Neuroscience 145:110– 115 Sasano H, Suzuki T, Miki Y, Moriya T, 2008, Intracrinology of estrogens and androgens in breast carcinoma. J Steroid Biocyhem Mol Bill 108(3–5):181–185 Song D, Liu G, Luu-The V, Zhao D, Want I, Zhang H, Xueling G, Li S, Desy L, Labrie F, Pelletier G, 2006, Expression of aromatase and 17β-hydroxysteroid dehydrogease types 1, 7 and 12 in breast cancer. An immunocytochemical study. J Steroid Biochem Mol Biol 101:136–144 Steward PM, Krozowski ZS, 1999, 11 beta-Hydroxysteroid dehydrogenase. Vitam Horm 57:249–324 Teulings FAG, Van Gilse HA, 1977, Demonstration of glucocorticoid receptors in human mammary carcinomas. Horm Res, Basel, 8:107–116 Wang H, Want Y, Rayburn ER, Hill DL, Rinehart JJ, Zhang R, 2007, Dexamethasone as a chemosensitizer for breast cancer chemotherapy: potentiaion of the antitumor activity of adriamycin, modulation of cytokine expression, and pharmacokinetics. Int J Clin Oncol 30(4):947–953 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 627 EP 632 DI 10.1007/s12253-011-9361-3 PG 6 ER PT J AU Stamatelli, A Saetta, AA Bei, Th Kavantzas, N Michalopoulos, VN Patsouris, E Aroni, K AF Stamatelli, Angeliki Saetta, A Angelica Bei, Thaleia Kavantzas, Nicolaos Michalopoulos, V Nicolaos Patsouris, Efstratios Aroni, Kiriaki TI B-Raf Mutations, Microsatellite Instability and p53 Protein Expression in Sporadic Basal Cell Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Basal cell carcinoma; B-Raf; Microsatellite instability; p53; Skin ID Basal cell carcinoma; B-Raf; Microsatellite instability; p53; Skin AB Basal Cell Carcinoma (BCC) is the most common skin malignancy. Genes related to the Ras/Raf signalling pathway have been implicated in the pathogenesis of skin cancer. The objective of this study was to investigate the presence of B-Raf mutations in sporadic BCCs as well as its correlation with the phenotype of microsatellite instability (MSI), the clinicopathological parameters of the tumours and p53 protein expression. 83 BCC specimens were screened for B-Raf mutations, applying polymerase chain reaction, single-stranded conformation polymorphism (PCR-SSCP) and DNA sequencing. MSI status was examined using mononucleotide microsatellite markers and p53 protein expression was demonstrated by immunohistochemical staining. A C to T transition at 1790 nucleotide leading to a silent mutation L597L; and a T to A transversion causing an amino acid change (F610I) have been found. MSI was detected in 5% of the cases and p53 accumulation was present in 37/83 samples studied. Although rare B-Raf alterations have been observed in BCC, none of them harboured the hot-spot mutation T1799A commonly present in melanomas and colon carcinomas. Consequently, no correlation could be determined between B-Raf alterations, MSI status, the clinicopathological features and p53 protein expression. Our results are in favour of a secondary importance for Ras signalling cascade genes in BCC pathogenesis. C1 [Stamatelli, Angeliki] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str, GR-115 27 Athens, Greece. [Saetta, A Angelica] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str, GR-115 27 Athens, Greece. [Bei, Thaleia] NIH, National Institutes of Child Health and Human Development, 20892 Bethesda, MD, USA. [Kavantzas, Nicolaos] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str, GR-115 27 Athens, Greece. [Michalopoulos, V Nicolaos] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str, GR-115 27 Athens, Greece. [Patsouris, Efstratios] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str, GR-115 27 Athens, Greece. [Aroni, Kiriaki] University of Athens, Medical School, Department of Pathology, 75 Mikras Asias Str, GR-115 27 Athens, Greece. RP Stamatelli, A (reprint author), University of Athens, Medical School, Department of Pathology, GR-115 27 Athens, Greece. EM agstam@med.uoa.gr CR Ionescu DN, Arida M, Jukic DM, 2006, Metastatic basal cell carcinoma: four case reports, review of literature, and immunohistochemical evaluation. Arch Pathol Lab Med 130:45–51 Crowson AN, 2006, Basal cell carcinoma: biology, morphology and clinical implications. Mod Pathol 19(suppl 2):S127–S147 Green CL, Khavari PA, 2004, Targets for molecular therapy of skin cancer. Semin Cancer Biol 14:63–69 Reifenberger J, Wolter M, Knobbe CB et al, 2005, Somatic mutations in the PTCH, SMOH, SUFUH and TP53 genes in sporadic basal cell carcinomas. Br J Dermatol 152:43–51 Pons M, Quintanilla M, 2006, Molecular biology of malignant melanoma and other cutaneous tumors. Clin Transl Oncol 8:466–74 Dhomen N, Marais R, 2007, New insight into BRAF mutations in cancer. Curr Opin Genet Dev 17:31–39 Houben R, Becker JC, Kappel A et al, 2004, Constitutive activation of the Ras-Raf signaling pathway in metastatic melanoma is associated with poor prognosis. J Carcinog 26:3–6 Davies H, Bignell GR, Cox C, 2002, Mutations of the BRAF gene in human cancer. Nature 417:949–954 Rajagopalan H, Bardelli A, Lengauer C et al, 2002, Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature 418, 6901):934 Domingo E, Espin E, Armengol M et al, 2004, Activated BRAF targets proximal colon tumors with mismatch repair deficiency and MLH1 inactivation. Genes Chromosom Cancer 39:138–142 Saetta AA, Aroni K, Stamatelli A et al, 2005, Expression of mismatch repair enzymes, hMLH1 and hMSH2 is not associated with microsatellite instability and p53 protein accumulation in basal cell carcinoma. Arch Dermatol Res 297:99–107 Zhou XP, Hoang JM, Li YJ et al, 1998, Determination of the replication error phenotype in human tumours without the requirement for matching normal DNA by analysis of mononucleotide repeat microsatellites. Genes Chromosom Cancer 21:101–107 de la Chapelle A, 1999, Testing tumors for microsatellite instability. Eur J Hum Genet 7:407–408 Roberts PJ, Der CJ, 2007, Targeting the Raf-MEK-ERK mitogenactivated protein kinase cascade for the treatment of cancer. Oncogene 26:3291–3310 Mercer KE, Pritchard CA, 2003, Raf proteins and cancer: B-Raf is identified as a mutational target. Biochim Biophys Acta 1653:25–40 Libra M, Malaponte G, Bevelacqua V et al, 2006, Absence of BRAF gene mutation in non-melanoma skin tumors. Cell Cycle 5:968–970 Ziegler A, Leffell DJ, Kunala S et al, 1993, Mutation hotspots due to sunlight in the p53 gene of nonmelanoma skin cancers. Proc Natl Acad Sci USA 90:4216–4220 Shea CR, McNutt NS, Volkenandt M et al, 1992, Overexpression of p53 protein in basal cell carcinomas of human skin. Am J Pathol 14:25–29 Barbareschi M, Girlando S, Cristofolini P et al, 1992, p53 protein expression in basal cell carcinomas. Histopathology 21:579–581 De Rosa G, Staibano S, Barra E et al, 1993, p53 protein in aggressive and non-aggressive basal cell carcinoma. J Cutan Pathol 20:429–434 Ro YS, Cooper PN, Lee JA et al, 1993, p53 protein expression in benign and malignant skin tumours. Br J Dermatol 128:237–241 Barrett TL, Smith KJ, Hodge JJ et al, 1997, Immunohistochemical nuclear staining for p53, PCNA, and Ki-67 in different histologic variants of basal cell carcinoma. J Am Acad Dermatol 37:430–437 Boonchai W, Walsh M, Cummings M et al, 2000, Expression of p53 in arsenic related and sporadic basal cell carcinoma. Arch Dermatol 136:195–198 Demirkan NC, Colakoglu N, Duzcan E, 2000, Value of p53 protein in biological behavior of basal cell carcinoma and in normal epithelia adjacent to carcinomas. Pathol Oncol Res 6:272–274 Auepemkiate S, Boonyaphiphat P, Thongsuksai P, 2002, p53 expression related to the aggressive infiltrative histopathological feature of basal cell carcinoma. Histopathology 40:568–573 Ansarin H, Daliri M, Soltani-Arabshahi R, 2006, Expression of p53 in aggressive and non-aggressive histologic variants of basal cell carcinoma. Eur J Dermatol 16:543–547 Quinn GA, Healy E, Rehman I et al, 1995, Microsatellite instability in human non-melanoma and melanoma skin cancer. J Invest Dermatol 104:309–312 Wisgerhof HC, Hameetman L, Tensen CP et al, 2009, Azathioprineinduced microsatellite instability is not observed in skin carcinomas of organ transplant recipients. J Invest Dermatol 129:1307–1309 Young LC, Listgarten J, Trotter MJ et al, 2008, Evidence that dysregulated DNA mismatch repair characterizes human nonmelanoma skin cancer. Br J Dermatol 158:59–69 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 633 EP 637 DI 10.1007/s12253-011-9363-1 PG 5 ER PT J AU Deisch, J Raisanen, J Rakheja, D AF Deisch, Jeremy Raisanen, Jack Rakheja, Dinesh TI Immunoexpression of SALL4 in Wilms Tumors and Developing Kidney SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Immunohistochemistry; Kidney development; Nephrogenic rest; SALL4; Wilms tumor ID Immunohistochemistry; Kidney development; Nephrogenic rest; SALL4; Wilms tumor AB SALL4 is a zinc finger transcription factor that plays a role in the maintainence and pluripotency of embryonic stem cell and is important in renal development where SALL4 mutations give rise to renal malformations. Because Wilms tumor recapitulates renal embryogenesis, we hypothesized that Wilms tumor cells may also express SALL4. We performed immunohistochemistry for SALL4 on tissue microarray sections of Wilms tumors, nephrogenic rests, and fetal renal cortices. Half (26 out of 52) of the Wilms tumors showed SALL4 immunoreactivity, ranging from strong and diffuse to focal and weak. Blastemal, epithelial, and combined blastemal and epithelial patterns of immunoreactivity were identified. No cases showed stromal staining. In the fetal kidney, SALL4 expression was restricted to the blastema and primitive epithelium at 15 weeks’ gestation. SALL4 staining was not seen at later gestational ages, in non-neoplastic postnatal kidneys, or in nephrogenic rests. Our study is the first to demonstrate SALL4 immunoreactivity in Wilms tumors and in developing fetal kidney. The absence of SALL4 staining in nephrogenic rests, the presumed precursors of Wilms tumors, is intriguing and suggests that Wilms tumors have a pluripotency quality that may be lacking in nephrogenic rests. C1 [Deisch, Jeremy] University of Texas Southwestern Medical Center, Department of Pathology MC 9073, 5323 Harry Hines Boulevard, 75390 Dallas, TX, USA. [Raisanen, Jack] University of Texas Southwestern Medical Center, Department of Pathology MC 9073, 5323 Harry Hines Boulevard, 75390 Dallas, TX, USA. [Rakheja, Dinesh] University of Texas Southwestern Medical Center, Department of Pathology MC 9073, 5323 Harry Hines Boulevard, 75390 Dallas, TX, USA. RP Rakheja, D (reprint author), University of Texas Southwestern Medical Center, Department of Pathology MC 9073, 75390 Dallas, USA. EM dinesh.rakheja@utsouthwestern.edu CR Pode-Shakked N, Metsuyanim S, Rom-Gross E et al, 2009, Developmental tumourigenesis: NCAM as a putative marker for the malignant renal stem/progenitor cell population. J Cell Mol Med 13:1792–1808 Yang J, Chai L, Fowles TC et al, 2008, Genome-wide analysis reveals Sall4 to be a major regulator of pluripotency in murineembryonic stem cells. Proc Natl Acad Sci USA 105:19756–19761 Zhang J, TamWL, Tong GQ et al, 2006, Sall4 modulates embryonic stem cell pluripotency and early embryonic development by the transcriptional regulation of Pou5f1. Nat Cell Biol 8:1114–1123 Kuhnlein RP, Frommer G, Friedrich M et al, 1994, Spalt encodes an evolutionarily conserved zinc finger protein of novel structure which provides homeotic gene function in the head and tail region of the Drosophila embryo. EMBO J 13:168–179 Kohlhase J, Taschner PE, Burfeind P et al, 1999, Molecular analysis of SALL1 mutations in Townes-Brocks syndrome. Am J Hum Genet 64:435–445 Kohlhase J, Wischermann A, Reichenbach H, Froster U, Engel W, 1998, Mutations in the SALL1 putative transcription factor gene cause Townes-Brocks syndrome. Nat Genet 18:81–83 Metsuyanim S, Harari-Steinberg O, Buzhor E et al, 2009, Expression of stem cell markers in the human fetal kidney. PLoS ONE 4:e6709 Ma Y, Singer DB, Gozman A et al, 2001, Hsal 1 is related to kidney and gonad development and is expressed in Wilms tumor. Pediatr Nephrol 16:701–709 Metsuyanim S, Pode-Shakked N, Schmidt-Ott KM et al, 2008, Accumulation of malignant renal stem cells is associated with epigenetic changes in normal renal progenitor genes. Stem Cells 26:1808–1817 Chai L, Yang J, Di C et al, 2006, Transcriptional activation of the SALL1 by the human SIX1 homeodomain during kidney development. J Biol Chem 281:18918–18926 Yang J, Chai L, Gao C et al, 2008, SALL4 is a key regulator of survival and apoptosis in human leukemic cells. Blood 112:805– 813 Al-Baradie R, Yamada K, St Hilaire C et al, 2002, Duane radial ray syndrome, Okihiro syndrome, maps to 20q13 and results from mutations in SALL4, a new member of the SAL family. Am J Hum Genet 71:1195–1199 Borozdin W, Boehm D, Leipoldt M et al, 2004, SALL4 deletions are a common cause of Okihiro and acro-renal-ocular syndromes and confirm haploinsufficiency as the pathogenic mechanism. J Med Genet 41:e113 Borozdin W, Wright MJ, Hennekam RC et al, 2004, Novel mutations in the gene SALL4 provide further evidence for acrorenal- ocular and Okihiro syndromes being allelic entities, and extend the phenotypic spectrum. J Med Genet 41:e102 Kohlhase J, Chitayat D, Kotzot D et al, 2005, SALL4 mutations in Okihiro syndrome, Duane-radial ray syndrome), acro-renalocular syndrome, and related disorders. Hum Mutat 26:176–183 Kohlhase J, Heinrich M, Schubert L et al, 2002, Okihiro syndrome is caused by SALL4 mutations. Hum Mol Genet 11:2979–2987 Kohlhase J, Schubert L, Liebers M et al, 2003, Mutations at the SALL4 locus on chromosome 20 result in a range of clinically overlapping phenotypes, including Okihiro syndrome, Holt-Oram syndrome, acro-renal-ocular syndrome, and patients previously reported to represent thalidomide embryopathy. J Med Genet 40:473–478 Miertus J, Borozdin W, Frecer Vet al, 2006, A SALL4 zinc finger missense mutation predicted to result in increased DNA binding affinity is associated with cranial midline defects and mild features of Okihiro syndrome. Hum Genet 119:154–161 Terhal P, Rosler B, Kohlhase J, 2006, A family with features overlapping Okihiro syndrome, hemifacial microsomia and isolated Duane anomaly caused by a novel SALL4 mutation. Am J Med Genet A 140:222–226 Okihiro MM, Tasaki T, Nakano KK, Bennett BK, 1977, Duane syndrome and congenital upper-limb anomalies. A familial occurrence. Arch Neurol 34:174–179 Paradisi I, Arias S, 2007, IVIC syndrome is caused by a c.2607delA mutation in the SALL4 locus. Am J Med Genet A 143:326–332 Lu J, Jeong HW, Kong N et al, 2009, Stem cell factor SALL4 represses the transcriptions of PTEN and SALL1 through an epigenetic repressor complex. PLoS ONE 4:e5577 Sakaki-Yumoto M, Kobayashi C, Sato A et al, 2006, The murine homolog of SALL4, a causative gene in Okihiro syndrome, is essential for embryonic stem cell proliferation, and cooperates with Sall1 in anorectal, heart, brain and kidney development. Development 133:3005–3013 Cao D, Guo S, Allan RW, Molberg KH, Peng Y, 2009, SALL4 is a novel sensitive and specific marker of ovarian primitive germ cell tumors and is particularly useful in distinguishing yolk sac tumor from clear cell carcinoma. Am J Surg Pathol 33:894–904 Cao D, Humphrey PA, Allan RW, 2009, SALL4 is a novel sensitive and specific marker for metastatic germ cell tumors, with particular utility in detection of metastatic yolk sac tumors. Cancer 115:2640–2651 Cao D, Li J, Guo CC, Allan RW, Humphrey PA, 2009, SALL4 is a novel diagnostic marker for testicular germ cell tumors. Am J Surg Pathol 33:1065–1077 Liu A, Cheng L, Du J et al, 2010, Diagnostic utility of novel stem cell markers SALL4, OCT4, NANOG, SOX2, UTF1, and TCL1 in primary mediastinal germ cell tumors. Am J Surg Pathol 34:697–706 Mei K, Liu A, Allan RWet al, 2009, Diagnostic utility of SALL4 in primary germ cell tumors of the central nervous system: a study of 77 cases. Mod Pathol 22:1628–1636 Wang F, Liu A, Peng Y et al, 2009, Diagnostic utility of SALL4 in extragonadal yolk sac tumors: an immunohistochemical study of 59 cases with comparison to placental-like alkaline phosphatase, alpha-fetoprotein, and glypican-3. Am J Surg Pathol 33:1529–1539 Ushiku T, Shinozaki A, Shibahara J et al, 2010, SALL4 represents fetal gut differentiation of gastric cancer, and is diagnostically useful in distinguishing hepatoid gastric carcinoma from hepatocellular carcinoma. Am J Surg Pathol 34:533–540 Shuai X, Zhou D, Shen T et al, 2009, Overexpression of the novel oncogene SALL4 and activation of the Wnt/beta-catenin pathway in myelodysplastic syndromes. Cancer Genet Cytogenet 194:119–124 Cui W, Kong NR, Ma Y, Amin HM, Lai R, Chai L, 2006, Differential expression of the novel oncogene, SALL4, in lymphoma, plasma cell myeloma, and acute lymphoblastic leukemia. Mod Pathol 19:1585–1592 Ma Y, Cui W, Yang J et al, 2006, SALL4, a novel oncogene, is constitutively expressed in human acute myeloid leukemia, AML, and induces AML in transgenic mice. Blood 108:2726–2735 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 639 EP 644 DI 10.1007/s12253-011-9364-0 PG 6 ER PT J AU Mise, J Dembitz, V Banfic, H Visnjic, D AF Mise, Josko Dembitz, Vilma Banfic, Hrvoje Visnjic, Dora TI Combined Inhibition of PI3K and mTOR Exerts Synergistic Antiproliferative Effect, but Diminishes Differentiative Properties of Rapamycin in Acute Myeloid Leukemia Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ATRA; Leukemia; PI3K; PMA; Rapamycin ID ATRA; Leukemia; PI3K; PMA; Rapamycin AB A novel strategy has been suggested to enhance rapamycin-based cancer therapy through combining mammalian target of rapamycin (mTOR)-inhibitors with an inhibitor of the phosphatydilinositol 3-kinase PI3K/Akt or mitogen-activated protein kinase/extracellular signalregulated kinase (MAPK/ERK) pathway. However, recent study demonstrated the potentiating effect of rapamycin on all-trans-retinoic acid (ATRA)-mediated differentiation of acute myelogenous leukemia (AML) cells, prompting us to investigate the effects of longitudinal inhibition of PI3K/Akt/mTOR signaling pathway on both proliferation and differentiative capacity of AML. In NB4, HL-60, U937 and K562 cell lines, rapamycin exerted minimal antiproliferative effects, and combining PI3K inhibitor LY 294002 and rapamycin inhibited proliferation more than LY 294002 alone. Rapamycin potentiated differentiation of ATRA-treated NB4 cells, but the combination of rapamycin and LY 294002 inhibited the expression of CD11b in both ATRA- and phorbol myristate acetate (PMA)-stimulated cells more than PI3K inhibitor alone. These results demonstrate that, although the combination of PI3K inhibitor and rapamycin is more effective in inhibiting proliferation of AML, the concomitant inhibition of PI3K and mTOR by LY 294002 and rapamycin has more inhibitory effects on ATRA-mediated differentiation than the presence of PI3K-inhibitor alone, and diminishes positive effects of rapamycin on leukemia cell differentiation. C1 [Mise, Josko] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Salata 3, 10 000 Zagreb, Croatia. [Dembitz, Vilma] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Salata 3, 10 000 Zagreb, Croatia. [Banfic, Hrvoje] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Salata 3, 10 000 Zagreb, Croatia. [Visnjic, Dora] School of Medicine University of Zagreb, Croatian Institute for Brain Research, Salata 3, 10 000 Zagreb, Croatia. RP Visnjic, D (reprint author), School of Medicine University of Zagreb, Croatian Institute for Brain Research, 10 000 Zagreb, Croatia. EM visnjic@mef.hr CR Xu Q, Simpson SE, Scialla TJ et al, 2003, Survival of acute myeloid leukemia cells requires PI3 kinase activation. Blood 102:972–980 Follo MY, Mongiorgi S, Bosi C et al, 2007, The Akt/mammalian target of rapamycin signal transduction pathway is activated in high-risk myelodysplastic syndromes and influences cell survival and proliferation. Cancer Res 67:4287–4294 LoPiccolo J, Blumenthal GM, Bernstein WB et al, 2008, Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist Updat 11:32–50 Papa V, Tazzari PL, Chiarini F et al, 2008, Proapoptotic activity and chemosensitizing effect of the novel Akt inhibitor perifosine in acute myelogenous leukemia cells. Leukemia 22:147–160 TabelliniG, Tazzari PL, Bortul R et al, 2004, Novel 2′-substituted, 3′- deoxy-phosphatidyl-myo-inositol analogues reduce drug resistance in human leukaemia cell lines with an activated phosphoinositide 3-kinase/Akt pathway. Br J Haematol 126:574–582 Richardson P, Wolf JL, Jakubowiak A et al, 2009, Perifosine in combination with bortezomib and dexamethasone extends progression-free survival and overall survival in relapsed/refractory multiple myeloma patients previously treated with bortezombib: updated phase I/II Trial Results. Blood, ASH Ann Meeting Abstracts). 114:1869 Altman JK, Platanias LC, 2008, Exploiting the mammalian target of rapamycin pathway in hematologic malignancies. Curr Opin Hematol 15:88–94 Bhaskar PT, Hay N, 2007, The two TORCs and Akt. Dev Cell 12:487–502 Recher C, Beyne-Rauzy O, Demur C et al, 2005, Antileukemic activity of rapamycin in acute myeloid leukemia. Blood 105:2527–2534 Nishioka C, Ikezoe T, Yang J et al, 2008, Blockade of mTOR signaling potentiates the ability of histone deacetylase inhibitor to induce growth arrest and differentiation of acute myelogenous leukemia cells. Leukemia 22:2159–2168 Rizzieri DA, Feldman E, Dipersio JF et al, 2008, A phase 2 clinical trial of deforolimus, AP23573, MK-8669), a novel mammalian target of rapamycin inhibitor, in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res 14:2756–2762 Yee KW, Zeng Z, Konopleva M et al, 2006, Phase I/II study of the mammalian target of rapamycin inhibitor everolimus, RAD001, in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res 12:5165–5173 Kharas MG, Janes MR, Scarfone VM et al, 2008, Ablation of PI3K blocks BCR-ABL leukemogenesis in mice, and a dual PI3K/ mTOR inhibitor prevents expansion of human BCR-ABL + leukemia cells. J Clin Invest 118:3038–3050 Sun SY, Rosenberg LM, Wang X et al, 2005, Activation of Akt and eIF4E survival pathways by rapamycin-mediated mammalian target of rapamycin inhibition. Cancer Res 65:7052–7058 Tamburini J, Chapuis N, Bardet V et al, 2008, Mammalian target of rapamycin, mTOR, inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways. Blood 111:379–382 Carracedo A, Ma L, Teruya-Feldstein J et al, 2008, Inhibition of mTORC1 leads to MAPK pathway activation through a PI3Kdependent feedback loop in human cancer. J Clin Invest 118:3065–3074 Kinkade CW, Castillo-Martin M, Puzio-Kuter A et al, 2008, Targeting AKT/mTOR and ERK MAPK signaling inhibits hormone-refractory prostate cancer in a preclinical mouse model. J Clin Invest 118:3051–3064 Wang ZY, Chen Z, 2008, Acute promyelocytic leukemia: from highly fatal to highly curable. Blood 111:2505–2515 Petrie K, Zelent A, Waxman S, 2009, Differentiation therapy of acute myeloid leukemia: past, present and future. Curr Opin Hematol 16:84–91 Matkovic K, Brugnoli F, Bertagnoli V et al, 2006, The role of the nuclear Akt activation and Akt inhibitors in all-trans-retinoic aciddifferentiated HL-60 cells. Leukemia 20:941–951 Nishioka C, Ikezoe T, Yang J et al, 2009, Inhibition of mammalian target of rapamycin signaling potentiates the effects of all-trans retinoic acid to induce growth arrest and differentiation of human acute myelogenous leukemia cells. Int J Cancer 125:1710–1720 Matkovic K, Lukinovic-Skudar V, Banfic H et al, 2009, The activity of extracellular signal-regulated kinase is required during G2/M phase before metaphase-anaphase transition in synchronized leukemia cell lines. Int J Hematol 89:159–166 Breslin EM, White PC, Shore AM et al, 2005, LY294002 and rapamycin co-operate to inhibit T-cell proliferation. Br J Pharmacol 144:791–800 Ikezoe T, Nishioka C, Bandobashi K et al, 2007, Longitudinal inhibition of PI3K/Akt/mTOR signaling by LY294002 and rapamycin induces growth arrest of adult T-cell leukemia cells. Leuk Res 31:673–682 Scholl S, Bondeva T, Liu Y et al, 2008, Additive effects of PI3- kinase and MAPK activities on NB4 cell granulocyte differentiation: potential role of phosphatidylinositol 3-kinase gamma. J Cancer Res Clin Oncol 134:861–872 Yen A, Roberson MS, Varvayanis S et al, 1998, Retinoic acid induced mitogen-activated protein, MAP)/extracellular signalregulated kinase, ERK, kinase-dependent MAP kinase activation needed to elicit HL-60 cell differentiation and growth arrest. Cancer Res 58:3163–3172 Das D, Pintucci G, Stern A, 2000, MAPK-dependent expression of p21WAF and p27kip1 in PMA-induced differentiation of HL-60 cells. FEBS Lett 472:50–52 Miranda MB, McGuire TF, Johnson DE, 2002, Importance of MEK-1/-2 signaling in monocytic and granulocytic differentiation of myeloid cell lines. Leukemia 16:683–692 Crabbe T, Welham MJ, Ward SG, 2007, The PI3K inhibitor arsenal: choose your weapon! Trends Biochem Sci 32:450–456 Chiarini F, Fala F, Tazzari PL et al, 2009, Dual inhibition of class IA phosphatidylinositol 3-kinase and mammalian target of rapamycin as a new therapeutic option for T-cell acute lymphoblastic leukemia. Cancer Res 69:3520–3528 Park S, Chapuis N, Bardet V et al, 2008, PI-103, a dual inhibitor of Class IA phosphatidylinositide 3-kinase and mTOR, has antileukemic activity in AML. Leukemia 22:1698–1706 Bertagnolo V, Neri LM, Marchisio M et al, 1999, Phosphoinositide 3-kinase activity is essential for all-trans-retinoic acid-induced granulocytic differentiation of HL-60 cells. Cancer Res 59:542–546 Lal L, Li Y, Smith J et al, 2005, Activation of the p70 S6 kinase by all-trans-retinoic acid in acute promyelocytic leukemia cells. Blood 105:1669–1677 Hay N, 2005, The Akt-mTOR tango and its relevance to cancer. Cancer Cell 8:179–183 Park SJ, Kang SY, Kim NS et al, 2002, Phosphatidylinositol 3-kinase regulates PMA-induced differentiation and superoxide production in HL-60 cells. Immunopharmacol Immunotoxicol 24:211–226 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 645 EP 656 DI 10.1007/s12253-011-9365-z PG 12 ER PT J AU Fang, Z Xiong, Y Li, J Liu, L Li, M Zhang, W Shi, L Wan, J AF Fang, Zhengyu Xiong, Yi Li, Jiana Liu, Li Li, Manhui Zhang, Wei Shi, Lei Wan, Jun TI Detection of APC Gene Deletions in Colorectal Malignancies Using Quantitative PCR in a Chinese Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; APC; Copy number variation; Gene expression ID Colorectal cancer; APC; Copy number variation; Gene expression AB The adenomatous polyposis coli (APC) gene has been shown to be involved in genetic instability and to be downregluated in several human carcinomas. The chromosome locus of APC, 5q21-22, is frequently deleted in colorectal cancers (CRCs). The functional impact of such regions needs to be extensively investigated in large amount of clinical samples. Case-matched tissues of CRC and adjacent normal epithelium (n=134) were included in this study. Quantitative PCR was carried out to examine the copy number as well as mRNA expression of APC gene in colorectal malignancies. Our results showed that copy number deletions of APC were present in a relatively high percentage of colorectal cancer samples (26.1%, 35 out of 134). There was a positive correlation between copy number decrease of APC and tumor progression in CRCs. Furthermore, copy number loss of APC was correlated with decreased mRNA expression. However, mRNA levels of APC were also impaired in CRC samples with unaltered copy numbers, indicating that sporadic CRCs exhibit different mechanisms of APC regulation. C1 [Fang, Zhengyu] Shenzhen-PKU-HKUST Medical Center, Biomedical Research InstituteShenzhen, Guangdong Province, China. [Xiong, Yi] Shenzhen-PKU-HKUST Medical Center, Biomedical Research InstituteShenzhen, Guangdong Province, China. [Li, Jiana] Shenzhen-PKU-HKUST Medical Center, Biomedical Research InstituteShenzhen, Guangdong Province, China. [Liu, Li] Shenzhen-PKU-HKUST Medical Center, Biomedical Research InstituteShenzhen, Guangdong Province, China. [Li, Manhui] Shenzhen-PKU-HKUST Medical Center, Biomedical Research InstituteShenzhen, Guangdong Province, China. [Zhang, Wei] Ji-Nan University, JNU-HKUST Joint LabGuangzhou, Guangdong, China. [Shi, Lei] The Hong Kong University of Science and Technology, Division of Life Science, Section of Biochemistry and Cell BiologyKowloon, Hong Kong. [Wan, Jun] Shenzhen-PKU-HKUST Medical Center, Biomedical Research InstituteShenzhen, Guangdong Province, China. RP Wan, J (reprint author), Shenzhen-PKU-HKUST Medical Center, Biomedical Research Institute, Shenzhen, China. EM wanj@ust.hk CR Munemitsu S, Souza B, Muller O et al, 1994, The apc gene product associates with microtubules in vivo and promotes their assembly in vitro. Cancer Res 54:3676–3681 Henderson BR, 2000, Nuclear-cytoplasmic shuttling of apc regulates beta-catenin subcellular localization and turnover. Nat Cell Biol 2:653–660 Neufeld KL, Nix DA, Bogerd H et al, 2000, Adenomatous polyposis coli protein contains two nuclear export signals and shuttles between the nucleus and cytoplasm. Proc Natl Acad Sci USA 97:12085–12090 Wong MH, Hermiston ML, Syder AJ et al, 1996, Forced expression of the tumor suppressor adenomatosis polyposis coli protein induces disordered cell migration in the intestinal epithelium. Proc Natl Acad Sci USA 93:9588–9593 Narayan S, Roy D, 2003, Role of apc and DNA mismatch repair genes in the development of colorectal cancers. Mol Cancer 2:41 Smith KJ, Levy DB, Maupin P et al, 1994, Wild-type but not mutant apc associates with the microtubule cytoskeleton. Cancer Res 54:3672–3675 Barth AI, Pollack AL, Altschuler Y et al, 1997, Nh2-terminal deletion of beta-catenin results in stable colocalization of mutant beta-catenin with adenomatous polyposis coli protein and altered mdck cell adhesion. J Cell Biol 136:693–706 Goss KH, Groden J, 2000, Biology of the adenomatous polyposis coli tumor suppressor. J Clin Oncol 18:1967–1979 Fearnhead NS, Britton MP, Bodmer WF, 2001, The abc of apc. Hum Mol Genet 10:721–733 Gumbiner BM, 1997, Carcinogenesis: a balance between betacatenin and apc. Curr Biol 7:R443–R446 McCartney BM, Nathke IS, 2008, Cell regulation by the apc protein apc as master regulator of epithelia. Curr Opin Cell Biol 20:186–193 Huang J, Zheng S, Jin S, 1996, apc mutation analysis in sporadic colorectal cancer. Zhonghua Zhong Liu Za Zhi 18:415–418 Ruiz-Ponte C, Vega A, Carracedo A et al, 2001, Mutation analysis of the adenomatous polyposis coli, apc, gene in northwest spanish patients with familial adenomatous polyposis, fap, and sporadic colorectal cancer. Hum Mutat 18:355 Lichtenstein P, Holm NV, Verkasalo PK et al, 2000, Environmental and heritable factors in the causation of cancer—analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med 343:78–85 Chung DC, 2000, The genetic basis of colorectal cancer: Insights into critical pathways of tumorigenesis. Gastroenterology 119:854–865 Camps J, Armengol G, del Rey J et al, 2006, Genome-wide differences between microsatellite stable and unstable colorectal tumors. Carcinogenesis 27:419–428 Knosel T, Petersen S, Schwabe H et al, 2002, Incidence of chromosomal imbalances in advanced colorectal carcinomas and their metastases. Virchows Arch 440:187–194 Ueno K, Kumagai T, Kijima T et al, 1998, Cloning and tissue expression of cdnas from chromosome 5q21-22 which is frequently deleted in advanced lung cancer. Hum Genet 102:63– 68 Redon R, Ishikawa S, Fitch KR et al, 2006, Global variation in copy number in the human genome. Nature 444:444–454 Oreffo VI, Robinson S, You M et al, 1998, Decreased expression of the adenomatous polyposis coli, apc, and mutated in colorectal cancer, mcc, genes in mouse lung neoplasia. Mol Carcinog 21:37–49 Bodmer WF, Cottrell S, Frischauf AM et al, 1989, Genetic analysis of colorectal cancer. Princess Takamatsu Symp 20:49–59 Luo L, Shen GQ, Stiffler KA et al, 2006, Loss of heterozygosity in human aberrant crypt foci, acf), a putative precursor of colon cancer. Carcinogenesis 27:1153–1159 Dermitzakis ET, Stranger BE, 2006, Genetic variation in human gene expression. Mamm Genome 17:503–508 Reymond A, Henrichsen CN, Harewood L et al, 2007, Side effects of genome structural changes. Curr Opin Genet Dev 17:381–386 Jass JR, Barker M, Fraser L et al, 2003, Apc mutation and tumour budding in colorectal cancer. J Clin Pathol 56:69–73 Ostwald C, Linnebacher M, Weirich V et al, 2009, Chromosomally and microsatellite stable colorectal carcinomas without the cpg island methylator phenotype in a molecular classification. Int J Oncol 35:321–327 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 657 EP 661 DI 10.1007/s12253-010-9359-2 PG 5 ER PT J AU Cserni, G Francz, M Kalman, E Kelemen, Gy Komjathy, CsD Kovacs, I Kulka, J Sarkadi, L Udvarhelyi, N Vass, L Voros, A AF Cserni, Gabor Francz, Monika Kalman, Endre Kelemen, Gyongyi Komjathy, Csaba Detre Kovacs, Ilona Kulka, Janina Sarkadi, Laszlo Udvarhelyi, Nora Vass, Laszlo Voros, Andras TI Estrogen Receptor Negative and Progesterone Receptor Positive Breast Carcinomas—How Frequent are they? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Estrogen receptor; Progesterone receptor; Immunohistochemistry; Estrogen receptor-negative progesterone receptor-positive breast cancer ID Breast cancer; Estrogen receptor; Progesterone receptor; Immunohistochemistry; Estrogen receptor-negative progesterone receptor-positive breast cancer AB Estrogen receptor (ER) testing has become an important part of breast cancer reporting as the ER status is a predictor of hormonal treatment efficacy. Progesteron receptors (PR) are often tested in parallel, and the best response to hormonal manipulations can be expected in tumors positive for both receptors. The existence of breast cancers with an ER negative and PR positive phenotype is controversial. A series of cases with this phenotype were reevaluated to clarify the existence and the frequency of this entity. A total of 205/6587 (3.1%; range of the rate per department: 0.3–7.1%.) cases reported to have the ER-negative and PR-positive status by immunohistochemistry were collected from 9 Hungarian departments. After careful reevaluation of the tumor slides and control tissues with a 1% cut-off for positivity and restaining of the questionable cases, all but 1 of the reevaluable 182 cases changed their original phenotype. Most cases converted to dual positives (n=124) or dual negatives (n=31) or unassessable / questionable. ER-negative and PR-positive breast cancers are very rare if existing. Such a phenotype should prompt reassessment. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, 6000 Kecskemet, Hungary. [Francz, Monika] Josa Andras Teaching Hospital, Department of Pathology, Szent Istvan u.68, 4400 Nyiregyhaza, Hungary. [Kalman, Endre] University of Pecs, Department of Pathology, Szigeti ut 12, 7624 Pecs, Hungary. [Kelemen, Gyongyi] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Komjathy, Csaba Detre] Kenezy Teaching Hospital, Department of Pathology, Bartok Bela u. 2-26, 4043 Debrecen, Hungary. [Kovacs, Ilona] Kenezy Teaching Hospital, Department of Pathology, Bartok Bela u. 2-26, 4043 Debrecen, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Sarkadi, Laszlo] Kenezy Teaching Hospital, Department of Pathology, Bartok Bela u. 2-26, 4043 Debrecen, Hungary. [Udvarhelyi, Nora] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy u.7-9, 1122 Budapest, Hungary. [Vass, Laszlo] Flor Ferenc University Hospital of Pest County, Department. of Pathology/Cytopathology, Semmelweis ter 1, 2143 Kistarcsa, Hungary. [Voros, Andras] University of Szeged, Department of Pathology, Allomas u. 2, 6720 Szeged, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, 6000 Kecskemet, Hungary. EM cserni@freemail.hu CR Moshin SK, Weiss H, Havinghurst T et al, 2004, Progesterone receptor by immunohistochemistry and clinical outcome in breast cancer: a validation study. Mod Pathol 17:1545–1554 Viale G, Regan MM, Maiorano E et al, 2008, Chemoendocrine compared with endocrine adjuvant therapies for node-negative breast cancer: predictive value of centrally reviewed expression of estrogen and progesterone receptors—International Breast Cancer Study Group. J Clin Oncol 26:1404–1410 Nadji M, Gomez-Ferenandez C, Ganjei-Azar P et al, 2005, Immunohistochemistry of estrogen and progesterone receptors reconsidered: experience with 5, 993 breast cancers. Am J Clin Pathol 123:21–27 De Maeyer L, Van Limbergen E, De Nys K et al, 2008, Does estrogen receptor-negative/progesterone receptor-positive breast carcinoma exist? J Clin Oncol 26:335–340 Goldhirsch A, Glick JH, Gelber RD et al, 2005, Meeting highlights: International expert consensus on the primary therapy of early breast cancer 2005. Ann Oncol 16:1569–1583 Rhodes A, Jasani B, 2009, The oestrogen receptor-negative/ progesterone receptor-positive breast tumour: a biological entity or a technical artefact? J Clin Pathol 62:95–96 Rakha EA, El-Sayed ME, Green AR et al, 2007, Biological and clinical characteristics of breast cancer with single hormone receptor-positive phenotype. J Clin Oncol 25:4772–4778 Hammond MEH, Hayes DF, Dowsett M et al, 2010, American Society of Clinical Oncology / College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol 28:2784–2795 Viale G, Regan MM, Maiorano E et al, 2007, Prognostic and predictive value of centrally reviewed expression of estrogen and progesterone receptors in a randomized trial comparing letrozole and tamoxifen adjuvant therapy for postmenopausal early breast cancer: BIG 1-98. J Clin Oncol 25:3846–3652 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 663 EP 668 DI 10.1007/s12253-011-9366-y PG 6 ER PT J AU Golmoghaddam, H Pezeshki, MA Ghaderi, A Doroudchi, M AF Golmoghaddam, Hossein Pezeshki, Mohammad Abdul Ghaderi, Abbas Doroudchi, Mehrnoosh TI CD1a and CD1d Genes Polymorphisms in Breast, Colorectal and Lung Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD1; Allele; Breast cancer; Lung cancer; Colorectal cancer ID CD1; Allele; Breast cancer; Lung cancer; Colorectal cancer AB CD1 molecules might contribute to anti-tumor immune response by presentation of tumor-derived lipid and glycolipid antigens to T cells and NKT cells. Polymorphisms in CD1 genes have been suggested to modify ligand binding of CD1 molecules and thereby change the antigen presenting ability of these molecules. The aim of this study was to investigate the exon 2 polymorphisms of CD1a and CD1d in several high incident cancers in Iran. For this purpose, 201 female breast cancer patients and 207 healthy women, 64 lung cancer patients and 95 healthy individuals and 109 patients with colorectal cancer and 109 healthy controls were recruited to this study. Using PCR-SSP method, no significant correlation was found in genotype and allele frequencies of CD1a between all three studied groups and their control counterparts. Moreover, a dominant frequency of CD1d 01 (A) allele was observed in the majority of studied individuals. No significant association between the CD1 polymorphisms and prognostic factors in breast, lung and colorectal cancers was detected. Our results highlight the conserved nature of CD1 genes and may point to the immuoregulatory functions of CD1 molecules in cancer that can be exerted through fine tuning of NK, T and NKT cells. C1 [Golmoghaddam, Hossein] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Pezeshki, Mohammad Abdul] Shiraz University of Medical Sciences, Institute for Cancer ResearchShiraz, Iran. [Ghaderi, Abbas] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Doroudchi, Mehrnoosh] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. RP Doroudchi, M (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Immunology, Shiraz, Iran. EM mdoroudchi@gmail.com CR Mori L, De Libero G, 2008, Presentation of lipid antigens to T cells. Immunol Lett 117:1–8 Salio M, Silk JD, Cerundolo V, 2010, Recent advances in processing and presentation of CD1 bound lipid antigens. Curr Opin Immunol 22:81–8 De Libero G, Mori L, 2010, How the immune system detects lipid antigens. Prog Lipid Res 49:120–7 De Libero G, Mori L, 2005, Recognition of lipid antigens by T cells. Nat Rev Immunol 5:485–96 Coventry B, Heinzel S, 2004, CD1a in human cancers: a new role for an old molecule. Trends Immunol 25:242–8 Porcelli SA, Modlin RL, 1999, The CD1 system: antigenpresenting molecules for T cell recognition of lipids and glycolipids. Annu Rev Immunol 17:297–329 Zeng Z, Castano AR, Segelke BWet al, 1997, Crystal structure of mouse CD1: An MHC-like fold with a large hydrophobic binding groove. Science 277:339–45 Kasmar A, Van Rhijn I, Moody DB, 2009, The evolved functions of CD1 during infection. Curr Opin Immunol 21:397–403 Swann J, Crowe NY, Hayakawa Y et al, 2004, Regulation of antitumour immunity by CD1d-restricted NKT cells. Immunol Cell Biol 82:323–31 Metelitsa LS, Weinberg KI, Emanuel PD, Seeger RC, 2003, Expression of CD1d by myelomonocytic leukemias provides a target for cytotoxic NKT cells. Leukemia 17:1068–77 La Rocca G, Anzalone R, Corrao S et al, 2008, CD1a downregulation in primary invasive ductal breast carcinoma may predict regional lymph node invasion and patient outcome. Histopathology 52:203–12 Poindexter NJ, Sahin A, Hunt KK, Grimm EA, 2004, Analysis of dendritic cells in tumor-free and tumor-containing sentinel lymph nodes from patients with breast cancer. Breast Cancer Res 6: R408–15 Coventry BJ, Morton J, 2003, CD1a-positive infiltrating-dendritic cell density and 5-year survival from human breast cancer. Br J Cancer 89:533–8 Iwamoto M, Shinohara H, Miyamoto A et al, 2003, Prognostic value of tumor-infiltrating dendritic cells expressing CD83 in human breast carcinomas. Int J Cancer 104:92–7 Cappello F, Rappa F, Anzalone R et al, 2005, CD1a expression by Barrett’s metaplasia of gastric type may help to predict its evolution towards cancer. Br J Cancer 92:888–90 McMullen TP, Lai R, Dabbagh L et al, 2010, Survival in rectal cancer is predicted by T cell infiltration of tumour-associated lymphoid nodules. Clin Exp Immunol 161:81–8 Gulubova M, Manolova I, Ananiev J et al, 2010, Role of TGF-beta1, its receptor TGFbetaRII, and Smad proteins in the progression of colorectal cancer. Int J Colorectal Dis 25:591–9 Sandel MH, Dadabayev AR, Menon AG et al, 2005, Prognostic value of tumor-infiltrating dendritic cells in colorectal cancer: role of maturation status and intratumoral localization. Clin Cancer Res 11:2576–82 Baleeiro RB, Anselmo LB, Soares FA et al, 2008, High frequency of immature dendritic cells and altered in situ production of interleukin 4 and tumor necrosis factor-alpha in lung cancer. Cancer Immunol Immunother 57:1335–45 Bergeron A, El-Hage F, Kambouchner M et al, 2006, Characterisation of dendritic cell subsets in lung cancer microenvironments. Eur Respir J 28:1170–7 Iorio E, Mezzanzanica D, Alberti P et al, 2005, Alterations of choline phospholipid metabolism in ovarian tumor progression. Cancer Res 65:9369–76 Han M, Hannick LI, DiBrino M, Robinson MA, 1999, Polymorphism of human CD1 genes. Tissue Antigens 54:122–7 Mirones I, Oteo M, Parra-Cuadrado JF, Martinez-Naves E, 2000, Identification of two novel human CD1E alleles. Tissue Antigens 56:159–61 Tamouza R, Sghiri R, Ramasawmy R et al, 2002, Two novel CD1 E alleles identified in black African individuals. Tissue Antigens 59:417–20 Gan LH, Pan YQ, Xu DP et al, 2010, Polymorphism of human CD1a, CD1d, and CD1e in exon 2 in Chinese Han and She ethnic populations. Tissue Antigens 75:691–5 Tourne S, Maitre B, Collmann A et al, 2008, Cutting edge: a naturally occurring mutation in CD1e impairs lipid antigen presentation. J Immunol 180:3642–6 Caporale CM, Papola F, Fioroni MA et al, 2006, Susceptibility to Guillain-Barre syndrome is associated to polymorphisms of CD1 genes. J Neuroimmunol 177:112–8 De Angelis MV, Notturno F, Caporale CM et al, 2007, Polymorphisms of CD1 genes in chronic dysimmune neuropathies. J Neuroimmunol 186:161–3 Jones DC, Gelder CM, Ahmad T et al, 2001, CD1 genotyping of patients with Mycobacterium malmoense pulmonary disease. Tissue Antigens 58:19–23 Miller SA, Dykes DD, Polesky HF, 1988, A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16:1215 Brossay L, Kronenberg M, 1999, Highly conserved antigenpresenting function of CD1d molecules. Immunogenetics 50:146– 51 Balk SP, Bleicher PA, Terhorst C, 1991, Isolation and expression of cDNA encoding the murine homologues of CD1. J Immunol 146:768–74 Hughes AL, 1991, Evolutionary origin and diversification of the mammalian CD1 antigen genes. Mol Biol Evol 8:185–201 Hughes AL, Yeager M, Ten Elshof AE, Chorney MJ, 1999, A new taxonomy of mammalian MHC class I molecules. Immunol Today 20:22–6 Dufour FD, Silveira PA, Baxter AG, 2008, Interactions between B-lymphocytes and type 1 NKT cells in autoimmune diabetes. J Immunotoxicol 5:249–57 Yang JQ, Singh AK, Wilson MT, Satoh M et al, 2003, Immunoregulatory role of CD1d in the hydrocarbon oil-induced model of lupus nephritis. J Immunol 171:2142–2153 Gumperz JE, 2006, The ins and outs of CD1 molecules: bringing lipids under immunological surveillance. Traffic 7:2– 13 Brigl M, Bry L, Kent SC et al, 2003, Mechanism of CD1drestricted natural killer T cell activation during microbial infection. Nat Immunol 4:1230–7 Hegde S, Chen X, Keaton JM et al, 2007, NKT cells direct monocytes into a DC differentiation pathway. J Leukoc Biol 81:1224–35 Cheng L, Ueno A, Cho S et al, 2007, Efficient activation of Valpha14 invariant NKT cells by foreign lipid antigen is associated with concurrent dendritic cell-specific self recognition. J Immunol 178:2755–62 Cheroutre H, Holcombe HR, Tangri S et al, 1995, Antigenpresenting function of the TL antigen and mouse CD1 molecules. Immunol Rev 147:31–52 Vincent MS, Leslie DS, Gumperz JE et al, 2002, CD1-dependent dendritic cell instruction. Nat Immunol 3:1163–8 Laskarin G, Redzovic A, Rubesa Z et al, 2008, Decidual natural killer cell tuning by autologous dendritic cells. Am J Reprod Immunol 59:433–45 Yue SC, Shaulov A, Wang R et al, 2005, CD1d ligation on human monocytes directly signals rapid NF-kappaB activation and production of bioactive IL-12. Proc Natl Acad Sci USA 102:11811–6 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 669 EP 675 DI 10.1007/s12253-011-9367-x PG 7 ER PT J AU Pimentel-Nunes, P Afonso, L Lopes, P Roncon-Albuquerque, R Goncalves, N Henrique, R Moreira-Dias, L Leite-Moreira, FA Dinis-Ribeiro, M AF Pimentel-Nunes, Pedro Afonso, Luis Lopes, Paula Roncon-Albuquerque, Roberto Goncalves, Nadia Henrique, Rui Moreira-Dias, Luis Leite-Moreira, F Adelino Dinis-Ribeiro, Mario TI Increased Expression of Toll-like Receptors (TLR) 2, 4 and 5 in Gastric Dysplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric pathology; Dysplasia; Innate immunity receptors; TLRs ID Gastric pathology; Dysplasia; Innate immunity receptors; TLRs AB TLRs are important innate immunity receptors. Even though TLR2, 4 and 5 appear to be important for Helicobacter pylori (HP) recognition, their role in the evolution of gastritis to more advanced lesions is still unknown. To compare the expression of TLR2, 4 and 5 in normal gastric mucosa, HP+ gastritis, intestinal metaplasia, dysplasia and adenocarcinoma. Immunohistochemistry for TLR2, 4 and 5 was performed with anti-TLR2-TLR4-TLR5 antibodies in 117 histological samples of normal gastric mucosa (n=22), HP+ gastritis (n=20), intestinal metaplasia (n=33), dysplasia (mucosectomy specimens, n=20) and intestinal type adenocarcinoma (surgery specimens,n=22); quantification of expression was performed independently by two pathologists taking into account the percentage of positive epithelial cells and the degree of expression (zero to three score). A statistically significant trend for progressive increase of TLRs expression from normal mucosa to gastric dysplasia was found (mean expression: normal mucosa 0.1; gastritis 1.0; metaplasia 2.2; dysplasia 2.8, p<0.01). All dysplasia samples presented more than 90% positive epithelial cells with strong expression (2.8;95%CI2.7–3). There was less TLRs expression in carcinomas (TLR2:1.0; TLR4:2.0 and TLR5:1.2, p<0.05) when compared with dysplasia, with TLR4 being more expressed than TLR2 and 5 in these lesions (p=0.03). A score of all markers’ expression of eight leads to a low (4%) false positive rate in patients with precancerous conditions. Progression of gastric lesions associated with gastric carcinogenesis is associated with increased TLRs expression. Gastric dysplasia presents a high level of TLRs expression, suggesting that these receptors may play a role in adenocarcinoma development. C1 [Pimentel-Nunes, Pedro] University of Porto, Cardiovascular Research & Development Unit, Al. Prof. Hernani Monteiro, 4200-319 Porto, Portugal. [Afonso, Luis] Portuguese Oncology Institute, Department of PathologyPorto, Portugal. [Lopes, Paula] Portuguese Oncology Institute, Department of PathologyPorto, Portugal. [Roncon-Albuquerque, Roberto] University of Porto, Cardiovascular Research & Development Unit, Al. Prof. Hernani Monteiro, 4200-319 Porto, Portugal. [Goncalves, Nadia] University of Porto, Cardiovascular Research & Development Unit, Al. Prof. Hernani Monteiro, 4200-319 Porto, Portugal. [Henrique, Rui] Portuguese Oncology Institute, Department of PathologyPorto, Portugal. [Moreira-Dias, Luis] Portuguese Oncology Institute, Gastroenterology DepartmentPorto, Portugal. [Leite-Moreira, F Adelino] University of Porto, Cardiovascular Research & Development Unit, Al. Prof. Hernani Monteiro, 4200-319 Porto, Portugal. [Dinis-Ribeiro, Mario] Portuguese Oncology Institute, Gastroenterology DepartmentPorto, Portugal. RP Pimentel-Nunes, P (reprint author), University of Porto, Cardiovascular Research & Development Unit, 4200-319 Porto, Portugal. EM pedronunesml@gmail.com CR Akira S, Hemmi H, 2003, Recognition of pathogen-associated molecular patterns by TLR family. Immunol Lett 85:85–95 Takeda K, Kaisho T, Akira S, 2003, Toll-like receptors. Annu Rev Immunol 21:335–376 Akira S, Takeda K, 2004, Toll-like receptor signalling. Nat Rev Immunol 4:499–511., DOI 10.1038/nri1391 Kawai T, Akira S, 2006, TLR signaling. Cell Death Differ 13:816–825., DOI 10.1038/sj.cdd.4401850 Rock FL, Hardiman G, Timans JC, Kastelein RA, Bazan JF, 1998, A family of human receptors structurally related to Drosophila Toll. Proc Natl Acad Sci USA 95:588–593 Medzhitov R, Janeway C Jr, 2000, Innate immunity. N Engl J Med 343:338–344 O’Mahony DS, Pham U, Iyer R, Hawn TR, Liles WC, 2008, Differential constitutive and cytokine-modulated expression of human Toll-like receptors in primary neutrophils, monocytes, and macrophages. Int J Med Sci 5:1–8 Kaisho T, Akira S, 2004, Pleiotropic function of Toll-like receptors. Microbes Infect 6:1388–1394., DOI 10.1016/j. micinf.2004.08.019 Clyne M, Drumm B, 1993, Adherence of Helicobacter pylori to primary human gastrointestinal cells. Infect Immun 61:4051– 4057 Ernst PB, Gold BD, 2000, The disease spectrum of Helicobacter pylori: the immunopathogenesis of gastroduodenal ulcer and gastric cancer. Annu Rev Microbiol 54:615–640 Fuchs CS, Mayer RJ, 1995, Gastric carcinoma. N Engl J Med 333:32–41 Houghton J, Wang TC, 2006, Tumors of the stomach. In: Feldman M, Friedman LS, Brandt LJ, eds, Sleisenger and Fordtran’s gastrointestinal and liver disease, 8 edn. Saunders Elsevier, pp 1139–1170 Kuipers EJ, Perez-Perez GI, Meuwissen SG, Blaser MJ, 1995, Helicobacter pylori and atrophic gastritis: importance of the cagA status. J Natl Cancer Inst 87:1777–1780 Correa P, 1992, Human gastric carcinogenesis: a multistep and multifactorial process—First American Cancer Society Award Lecture on Cancer Epidemiology and Prevention. Cancer Res 52:6735–6740 Correa P, 1995, Helicobacter pylori and gastric carcinogenesis. Am J Surg Pathol 19(Suppl 1):S37–S43 de Vries AC, Kuipers EJ, Rauws EA, 2009, Helicobacter pylori eradication and gastric cancer: when is the horse out of the barn? Am J Gastroenterol 104:1342–1345., DOI 10.1038/ajg.2008.15 Rokkas T, Pistiolas D, Sechopoulos P, Robotis I, Margantinis G, 2007, The long-term impact of Helicobacter pylori eradication on gastric histology: a systematic review and meta-analysis. Helicobacter 12(Suppl 2):32–38., DOI 10.1111/j.1523-5378.2007.00563.x Uemura N, Okamoto S, Yamamoto S, Matsumura N, Yamaguchi S, Yamakido M et al, 2001, Helicobacter pylori infection and the development of gastric cancer. N Engl J Med 345:784–789., DOI 10.1056/NEJMoa001999 Pimentel-Nunes P, Soares JB, Roncon-Albuquerque R Jr, Dinis- Ribeiro M, Leite-Moreira AF, 2010, Toll-like receptors as therapeutic targets in gastrointestinal diseases. Expert Opin Ther Targets 14:347–368., DOI 10.1517/14728221003642027 Rad R, Ballhorn W, Voland P, Eisenacher K, Mages J, Rad L et al, 2009, Extracellular and intracellular pattern recognition receptors cooperate in the recognition of Helicobacter pylori. Gastroenterology 136:2247–2257., DOI 10.1053/j.gastro.2009.02.066 Rad R, Brenner L, Krug A, Voland P, Mages J, Lang R et al, 2007, Toll-like receptor-dependent activation of antigen-presenting cells affects adaptive immunity to Helicobacter pylori. Gastroenterology 133:150–163.e3., DOI 10.1053/j.gastro.2007.04.071 Basak C, Pathak SK, Bhattacharyya A, Pathak S, Basu J, Kundu M, 2005, The secreted peptidyl prolyl cis, trans-isomerase HP0175 of Helicobacter pylori induces apoptosis of gastric epithelial cells in a TLR4- and apoptosis signal-regulating kinase 1-dependent manner. J Immunol 174:5672–5680 Ishihara S, Rumi MA, Kadowaki Y, Ortega-Cava CF, Yuki T, Yoshino N et al, 2004, Essential role of MD-2 in TLR4-dependent signaling during Helicobacter pylori-associated gastritis. J Immunol 173:1406–1416 Kawahara T, Kuwano Y, Teshima-Kondo S, Kawai T, Nikawa T, Kishi K et al, 2001, Toll-like receptor 4 regulates gastric pit cell responses to Helicobacter pylori infection. JMed Investig 48:190–197 Smith MF Jr, Mitchell A, Li G, Ding S, Fitzmaurice AM, Ryan K et al, 2003, Toll-like receptor, TLR, 2 and TLR5, but not TLR4, are required for Helicobacter pylori-induced NF-kappa B activation and chemokine expression by epithelial cells. J Biol Chem 278:32552–32560., DOI 10.1074/jbc.M305536200 Torok AM, Bouton AH, Goldberg JB, 2005, Helicobacter pylori induces interleukin-8 secretion by Toll-like receptor 2- and Tolllike receptor 5-dependent and -independent pathways. Infect Immun 73:1523–1531., DOI 10.1128/IAI.73.3.1523-1531.2005 Lee SK, Stack A, Katzowitsch E, Aizawa SI, Suerbaum S, Josenhans C, 2003, Helicobacter pylori flagellins have very low intrinsic activity to stimulate human gastric epithelial cells via TLR5. Microbes Infect 5:1345–1356 Andersen-Nissen E, Smith KD, Strobe KL, Barrett SL, Cookson BT, Logan SM et al, 2005, Evasion of Toll-like receptor 5 by flagellated bacteria. Proc Natl Acad Sci USA 102:9247–9252., DOI 10.1073/pnas.0502040102 Gewirtz AT, Yu Y, Krishna US, Israel DA, Lyons SL, Peek RM Jr, 2004, Helicobacter pylori flagellin evades toll-like receptor 5- mediated innate immunity. J Infect Dis 189:1914–1920., DOI 10.1086/386289 Schmausser B, Andrulis M, Endrich S, Lee SK, Josenhans C, Muller-Hermelink HK et al, 2004, Expression and subcellular distribution of toll-like receptors TLR4, TLR5 and TLR9 on the gastric epithelium in Helicobacter pylori infection. Clin Exp Immunol 136:521–526., DOI 10.1111/j.1365-2249.2004.02464.x Schmausser B, Andrulis M, Endrich S, Muller-Hermelink HK, Eck M, 2005, Toll-like receptors TLR4, TLR5 and TLR9 on gastric carcinoma cells: an implication for interaction with Helicobacter pylori. Int J Med Microbiol 295:179–185 Hopkins PA, Sriskandan S, 2005, Mammalian Toll-like receptors: to immunity and beyond. Clin Exp Immunol 140:395–407., DOI 10.1111/j.1365-2249.2005.02801.x Liew FY, Xu D, Brint EK, O’Neill LA, 2005, Negative regulation of toll-like receptor-mediated immune responses. Nat Rev Immunol 5:446–458., DOI 10.1038/nri1630 Ortega-Cava CF, Ishihara S, Rumi MA, Kawashima K, Ishimura N, Kazumori H et al, 2003, Strategic compartmentalization of Toll-like receptor 4 in the mouse gut. J Immunol 170:3977–3985 French SW, Oliva J, French BA, Li J, Bardag-Gorce F Alcohol, nutrition and liver cancer: role of Toll-like receptor signaling. World J Gastroenterol 16:1344–1348 Fukata M, Abreu MT, 2008, Role of Toll-like receptors in gastrointestinal malignancies. Oncogene 27:234–243., DOI 10.1038/ sj.onc.1210908 Achyut BR, Ghoshal UC, Moorchung N, Mittal B, 2007, Association of Toll-like receptor-4, Asp299Gly and Thr399Ileu, gene polymorphisms with gastritis and precancerous lesions. Hum Immunol 68:901–907., DOI 10.1016/j.humimm.2007.10.006 Hishida A, Matsuo K, Goto Y, Mitsuda Y, Hiraki A, Naito M et al, 2009, Toll-like receptor 4 +3725 G/C polymorphism, Helicobacter pylori seropositivity, and the risk of gastric atrophy and gastric cancer in Japanese. Helicobacter 14:47–53., DOI 10.1111/ j.1523-5378.2009.00659.x Tahara T, Arisawa T, Wang F, Shibata T, Nakamura M, Sakata M et al, 2007, Toll-like receptor 2–196 to 174del polymorphism influences the susceptibility of Japanese people to gastric cancer. Cancer Sci 98:1790–1794., DOI 10.1111/j.1349-7006.2007.00590.x Tahara T, Arisawa T, Wang F, Shibata T, Nakamura M, Sakata M et al, 2008, Toll-like receptor 2, TLR, -196 to 174del polymorphism in gastro-duodenal diseases in Japanese population. Dig Dis Sci 53:919–924., DOI 10.1007/s10620-007-9950-x la Trejo-de OA, Torres J, Perez-Rodriguez M, Camorlinga-Ponce M, Luna LF, Abdo-Francis JM et al, 2008, TLR4 single-nucleotide polymorphisms alter mucosal cytokine and chemokine patterns in Mexican patients with Helicobacter pylori-associated gastroduodenal diseases. Clin Immunol 129:333–340., DOI 10.1016/j.clim.2008.07.009 Chang YJ, Wu MS, Lin JT, Chen CC, 2005, Helicobacter pylori- Induced invasion and angiogenesis of gastric cells is mediated by cyclooxygenase-2 induction through TLR2/TLR9 and promoter regulation. J Immunol 175:8242–8252 Chang YJ, Wu MS, Lin JT, Sheu BS, Muta T, Inoue H et al, 2004, Induction of cyclooxygenase-2 overexpression in human gastric epithelial cells by Helicobacter pylori involves TLR2/TLR9 and c- Src-dependent nuclear factor-kappaB activation. Mol Pharmacol 66:1465–1477., DOI 10.1124/mol.104.005199 Chochi K, Ichikura T, Kinoshita M, Majima T, Shinomiya N, Tsujimoto H et al, 2008, Helicobacter pylori augments growth of gastric cancers via the lipopolysaccharide-toll-like receptor 4 pathway whereas its lipopolysaccharide attenuates antitumor activities of human mononuclear cells. Clin Cancer Res 14:2909–2917., DOI 10.1158/1078-0432.CCR-07-4467 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 677 EP 683 DI 10.1007/s12253-011-9368-9 PG 7 ER PT J AU Gao, J Zhang, J Long, Y Tian, Y Lu, X AF Gao, Jinbo Zhang, Jinghui Long, Yaoping Tian, Yuan Lu, Xiaoming TI Expression of Tankyrase 1 in Gastric Cancer and Its Correlation with Telomerase Activity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tankyrase 1; Gastric cancer; Telomerase; Telomere ID Tankyrase 1; Gastric cancer; Telomerase; Telomere AB Tankyrase 1, which enhances telomerase access to telomeres, plays an important role in telomere maintenance. The aim of this study was to determine the expression and potential role of tankyrase 1 in gastric cancer development and progression. We examined the expression of tankyrase 1 by RT-PCR and Western blotting, and assessed telomerase activity by TRAP-ELISA method in gastric cancer and adjacent normal tissues. We found that tankyrase 1 expression was significantly up-regulated in gastric cancer tissues compared to normal corresponding tissues. Tankyrase 1 over-expression by gastric cancerous tissue was significantly associated with tumor histology differentiation and tumor stage. Moreover, tankyrase 1 expression was significantly correlation with telomerase activity. Our results indicate that tankyrase 1 overexpression may play an important role in gastric cancer development and progression. Tankyrase 1 may be used as a biomarker of gastric cancer and may serve as a target for cancer therapy. C1 [Gao, Jinbo] Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Department of Surgery, 1277 Jiefang Avenue, 430022 Wuhan, Hubei, China. [Zhang, Jinghui] Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Department of Surgery, 1277 Jiefang Avenue, 430022 Wuhan, Hubei, China. [Long, Yaoping] Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Department of Surgery, 1277 Jiefang Avenue, 430022 Wuhan, Hubei, China. [Tian, Yuan] Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Department of Surgery, 1277 Jiefang Avenue, 430022 Wuhan, Hubei, China. [Lu, Xiaoming] Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Department of Surgery, 1277 Jiefang Avenue, 430022 Wuhan, Hubei, China. RP Gao, J (reprint author), Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Department of Surgery, 430022 Wuhan, China. EM drgaojinbo@gmail.com CR Zakian VA, 1995, Telomeres: beginning to understand the end. Science 270:1601–1607 Blackburn EH, 1991, Structure and function of telomeres. Nature 350:569–573 Levy MZ, Allsopp RC, Futcher AB, Greider CW, Harley CB, 1992, Telomere end-replication problem and cell aging. J Mol Biol 225:951–960 Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100:57–70 Osterhage JL, Friedman KL, 2009, Chromosome end maintenance by telomerase. J Biol Chem 284:16061–16065 De Boeck G, Forsyth RG, Praet M, Hogendoorn PC, 2009, Telomere-associated proteins:cross-talk between telomere maintenance and telomere-lengthening mechanisms. J Pathol 217:327–344 Feng J, Funk WD, Wang SS et al, 1995, The RNA component of human telomerase. Science 269:1236–1241 Nakamura TM, Morin GB, Chapman KB et al, 1997, Telomerase catalytic subunit homologs from fission yeast and human. Science 277:955–959 Kim NW, Piatuszek MA, Pronse KR et al, 1994, Specific association of human telomerase activity with immortal cells and cancer. Science 266:2011–2015 Shay JW, Bacchetti S, 1997, A survey of telomerase activity in human cancer. Eur J Cancer 33:787–791 Artandi SE, DePinho RA, 2010, Telomeres and telomerase in cancer. Carcinogenesis 31:9–18 Smith S, Giriat I, Schmitt A, de Lange T, 1998, Tankyrase, a poly, ADP-ribose, polymerase at human telomeres. Science 282:1484– 1487 Cook BD, Dynek JN, Chang W, Shostak G, Smith S, 2002, Role for the related poly(ADP-Ribose, polymerases tankyrase 1 and 2 at human telomeres. Mol Cell Biol 22:332–342 Smith S, de Lange T, 2000, Tankyrase promotes telomere elongation in human cells. Curr Biol 10:1299–1302 Xu D, Zheng C, Bergenbrant S et al, 2001, Telomerase activity in plasma cell dyscrasias. Br J Cancer 84:621–625 Klapper W, Krams M, Qian W, Janssen D, Parwaresch R, 2003, Telomerase activity in B-cell non-Hodgkin lymphomas is regulated by hTERT transcription and correlated with telomerebinding protein expression but uncoupled from proliferation. Br J Cancer 89:713–719 Gelmini S, Poggesi M, Distante V et al, 2004, Tankyrase, a positive regulator of telomere elongation, is over expressed in human breast cancer. Cancer Lett 216:81–87 Gelmini S, Poggesi M, Pinzani P et al, 2006, Distribution of Tankyrase-1 mRNA expression in colon cancer and its prospective correlation with progression stage. Oncol Rep 16:1261–1266 Gelmini S, Quattrone S, Malentacchi F et al, 2007, Tankyrase-1 mRNA expression in bladder cancer and paired urine sediment: preliminary experience. Clin Chem Lab Med 45:862–866 Sobin LH, Wittekind C, 2002, UICC: TNM classification of malignant tumors, 6th edn. Wiley, New York Matsutani N, Yokozaki H, Tahara E et al, 2001, Expression of telomeric repeat binding factor 1 and 2 and TRF1-interacting nuclear protein 2 in human gastric carcinomas. Int J Oncol 19:507–512 Yamada M, Tsuji N, Nakamura M et al, 2002, Down-regulation of TRF1, TRF2 and TIN2 genes is important to maintain telomeric DNA for gastric cancers. Anticancer Res 22:3303–3307 Canudas S, Houghtaling BR, Kim JY et al, 2007, Protein requirements for sister telomere association in human cells. EMBO J 26:4867–4878 Dynek JN, Smith S, 2004, Resolution of sister telomere association is required for progression through mitosis. Science 304:97–100 Yeh TY, Sbodio JI, Tsun ZY, Luo B, Chi NW, 2007, Insulinstimulated exocytosis of GLUT4 is enhanced by IRAP and its partner tankyrase. Biochem J 402:279–290 Tahara H, Kunivasu H, Yokozaki H et al, 1995, Telomerase activity in preneoplastic and neoplastic gastric and colorectal lesions. Clin Cancer Res 1:1245–1251 Yoo J, Park SY, Kang SJ, Kim BK, Shim SI, Kang CS, 2003, Expression of telomerase activity, human telomerase RNA, and telomerase reverse transcriptase in gastric adenocarcinomas. Mol Pathol 16:700–707 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 685 EP 690 DI 10.1007/s12253-011-9369-8 PG 6 ER PT J AU Rijavec, M Silar, M Triller, N Kern, I Cegovnik, U Kosnik, M Korosec, P AF Rijavec, Matija Silar, Mira Triller, Nadja Kern, Izidor Cegovnik, Urska Kosnik, Mitja Korosec, Peter TI Expressions of Topoisomerase IIα and BCRP in Metastatic Cells are Associated with Overall Survival in Small Cell Lung Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Small cell lung cancer; Metastatic cells; Topoisomerase IIα; Breast cancer resistance protein (BCRP); mRNA expression levels ID Small cell lung cancer; Metastatic cells; Topoisomerase IIα; Breast cancer resistance protein (BCRP); mRNA expression levels AB The aim of this study was to investigate the mRNA expression levels of multidrug resistanceassociated proteins in chemo-naive metastatic lung cancer cells and to determine the correlation with response to chemotherapy and overall survival. Metastatic cells were obtained by transbronchial fine needle aspiration biopsy of enlarged mediastinal lymph nodes in 14 patients with small cell lung cancer (SCLC) and 7 patients with non-small cell lung cancer (NSCLC). After cytological confirmation of lung cancer type, total RNA was extracted from biopsy samples and reverse transcribed to cDNA, and real-time PCR for the genes of interest [P-glycoprotein (P-gp), multidrug resistance protein 1 (MRP1), breast cancer resistance protein (BCRP), lung resistance protein (LRP) and topoisomerase IIα (TOPIIα)], was performed. We observed significantly decreased expression of BCRP and significantly increased expression of TOPIIα in metastatic SCLC cells compared to NSCLC. Furthermore, in SCLC high topoisomerase IIα and low BCRP expression levels positively correlated with longer overall survival. Our results showed higher expression levels of BCRP as well as lower levels of topoisomerase IIα in chemonaive metastatic cells in NSCLC than in SCLC. These results correlate with previous observations that metastatic SCLC cells at the beginning of chemotherapy are potentially more sensitive to chemotherapeutic agents while in metastatic NSCLC cells resistance is usually inherent. We also showed that altered levels of topoisomerase IIα and BCRP in SCLC are important factors that contribute to resistance to chemotherapeutics that interfere with the enzyme and/or DNA and are highly associated with overall survival. C1 [Rijavec, Matija] University Clinic of Respiratory and Allergic Diseases, Golnik 36, 4204 Golnik, Slovenia. [Silar, Mira] University Clinic of Respiratory and Allergic Diseases, Golnik 36, 4204 Golnik, Slovenia. [Triller, Nadja] University Clinic of Respiratory and Allergic Diseases, Golnik 36, 4204 Golnik, Slovenia. [Kern, Izidor] University Clinic of Respiratory and Allergic Diseases, Golnik 36, 4204 Golnik, Slovenia. [Cegovnik, Urska] University Clinic of Respiratory and Allergic Diseases, Golnik 36, 4204 Golnik, Slovenia. [Kosnik, Mitja] University Clinic of Respiratory and Allergic Diseases, Golnik 36, 4204 Golnik, Slovenia. [Korosec, Peter] University Clinic of Respiratory and Allergic Diseases, Golnik 36, 4204 Golnik, Slovenia. RP Rijavec, M (reprint author), University Clinic of Respiratory and Allergic Diseases, 4204 Golnik, Slovenia. EM matija.rijavec@klinika-golnik.si CR National Comprehensive Cancer Network, Clinical Practice Guidelines in Oncology: Small cell lung cancer. Available at: http://www.nccn.org/professionals/physician_gls/PDF/sclc.pdf. Accessed June 3, 2009 National Comprehensive Cancer Network, Clinical Practice Guidelines in Oncology: Non-Small cell lung cancer. Available at: http://www.nccn.org/professionals/physician_gls/PDF/nscl.pdf. Accessed June 3, 2009 Scagliotti GV, Novello S, Selvaggi G, 1999, Multidrug resistance in non-small-cell lung cancer. Ann Oncol 10:S83–S86 Smith EF, Postmus PE, 1995, Chemotherapy of small cell lung cancer. In: Carney DN, ed, lung cancer. The Bath Press, Avon, pp 156–172 Ihde DC, Minna JD, 1991, Non-small cell lung cancer. Part I: biology, diagnosis, and staging. Curr Probl Cancer 15:61–104 Spiro SG, Porter JC, 2002, Lung cancer-where are we today? Current advances in staging and nonsurgical treatment. Am J Respir Crit Care Med 166:1166–1196 Ikuta K, Takemura K, Sasaki K, Kihara M, Nishimura M, Ueda N, Naito S, Lee E, Shimizu E, Yamauchi A, 2005, Expression of multidrug resistance proteins and accumulation of cisplatin in human non-small cell lung cancer cells. Biol Pharm Bull 28:707–712 Chhatriwala H, Jafri N, Salgia R, 2006, A review of topoisomerase inhibition in lung cancer. Cancer Biol Ther 5:1600–1607 Endicott JA, Ling V, 1989, The biochemistry of P-glycoproteinmediated multidrug resistance. Ann Rev Biochem 58:137–171 Grant CE, Valdimarsson G, Hipfner DR, Almquist KC, Cole SPC, Deeley RG, 1994, Overexpression of multidrug resistanceassociated protein, MRP, increases resistance to natural product drugs. Cancer Res 54:357–361 Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK, Ross DD, 1998, A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci USA 95:15665– 15670 Klein I, Sarkadi B, Varadi A, 1999, An inventory of the human ABC proteins. Biochim Biophys Acta 1461:237–262 Triller N, Korosec P, Kern I, Kosnik M, Debeljak A, 2006, Multidrug resistance in small cell lung cancer: expression of P-glycoprotein, multidrug resistance protein 1 and lung resistance protein in chemo-naive patients and in relapsed disease. Lung Cancer 54:235–240 Berger W, Elbling L, Micksche M, 2000, Expression of the major vault protein LRP in human non-small-cell lung cancer cells: activation by short-term exposure to antineoplastic drugs. Int J Cancer 88:293–300 Larsen AK, Escargueil AE, Skladanowski A, 2000, Resistance mechanisms associated with altered intracellular distribution of anticancer agents. Pharmacol Ther 85:217–229 Osheroff N, Corbett AH, Robinson MJ, 1994, Mechanism of action of topoisomerase II-targeted antineoplastic drugs. Adv Pharmacol 29:105–126 Barrett-Lee PJ, 2005, Growth factor signalling in clinical breast cancer and its impact on response to conventional therapies: a review of chemotherapy. Endocr Relat Cancer 12: S125–S133 Guinee DG Jr, Holden JA, Benfield JR, Woodward ML, Przygodzki RM, Fishback NF, Koss MN, Travis WD, 1996, Comparison of DNA topoisomerase II alpha expression in small cell and nonsmall cell carcinoma of the lung. In search of a mechanism of chemotherapeutic response. Cancer 78:729–735 Pusztai L, Gianni L, 2004, Technology insight: emerging techniques to predict response to preoperative chemotherapy in breast cancer. Nat Clin Pract Oncol 1:44–50 Goldman B, 2003, Multidrug resistance: can new drugs help chemotherapy score against cancer? J Natl Cancer Inst 95:255–257 Leonard GD, Fojo T, Bates SE, 2003, The role of ABC transporters in clinical practice. Oncologist 8:411–424 Kawabata S, Oka M, Soda H, Shiozawa K, Nakatomi K, Tsurutani J, Nakamura Y, Doi S, Kitazaki T, Sugahara K, Yamada Y, Kamihira S, Kohno S, 2003, Expression and functional analyses of breast cancer resistance protein in lung cancer. Clin Cancer Res 9:3052–3057 Kitazono M, Sumizawa T, Takebayashi Y, Chen ZS, Furukawa T, Nagayama S, Tani A, Takao S, Aikou T, Akiyama S, 1999, Multidrug resistance and the lung resistance-related protein in human colon carcinoma SW-620 cells. J Natl Cancer Inst 91:1647–1653 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 691 EP 696 DI 10.1007/s12253-011-9370-2 PG 6 ER PT J AU Duzcan, F Duzcan, ES Sen, S Yorukoglu, K Caner, V Sen Turk, N Cetin, OG Kelten, C Tuna, B Sarsik, B Tepeli, E AF Duzcan, Fusun Duzcan, Ender Suleyman Sen, Sait Yorukoglu, Kutsal Caner, Vildan Sen Turk, Nilay Cetin, Ozan Gokhan Kelten, Canan Tuna, Burcin Sarsik, Banu Tepeli, Emre TI Expression and Amplification of Topoisomerase-2α in Type 1 and Type 2 Papillary Renal Cell Carcinomas and Its Correlation with HER2/neu Amplification SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary renal cell carcinoma; Topoisomerase- 2α; HER2/neu; FISH; Immunohistochemistry ID Papillary renal cell carcinoma; Topoisomerase- 2α; HER2/neu; FISH; Immunohistochemistry AB The current study was undertaken to investigate chromosomal and genetical aberrations leading to overexpression of Topoisomerase-2α (TOP2α) and to reveal the possible association of these aberrations with HER2/neu overexpression and gene amplification, and to search for the relationship between TOP2α and HER2/neu status with prognostical biomarkers in papillary renal cell carcinoma (RCC), a group of tumors with diverse molecular, chromosomal and clinical features. Archival cases of papillary RCC obtained from Departments of Pathology of Pamukkale, Ege and Dokuz Eylul Universities were studied in two groups (type 1 and type 2) each containing 20 cases. The level of TOP2α and HER2/neu expression by tumor cells were determined immunohistochemically. A multicolor FISH probe was used to define both amplification of HER2/neu and TOP2α genes, and polysomy 17. The ratio of cells expressing TOP2α in type 1 and type 2 papillary RCC were 24.29% and 6.89%, respectively. The difference was statistically significant comparing the average or median values of groups separately (p=0.002). The expression levels of TOP2α and HER2/neu were also correlated. TOP2α and HER2/neu were co-amplified in both groups. Immunohistochemical expression was not observed in 15 of 23 cases with HER2/neu amplification. The most frequent finding detected by FISH method was polysomy of chromosome 17. We had contradictory results compared with the findings reported in the limited numbers of literature. It shows us that papillary RCC constitute a heterogenous group of tumors with various cytogenetic features and morphological classification of these tumors may not be compatible with their molecular characteristics. C1 [Duzcan, Fusun] Pamukkale University, Medical Faculty, Department of Medical Genetics, Doktorlar Cad. Kat:3, BayramyeriDenizli, Turkey. [Duzcan, Ender Suleyman] Pamukkale University, Faculty of Medicine, Department of PathologyDenizli, Turkey. [Sen, Sait] Ege University, Faculty of Medicine, Department of PathologyIzmir, Turkey. [Yorukoglu, Kutsal] Dokuz Eylul University, Faculty of Medicine, Department of PathologyIzmir, Turkey. [Caner, Vildan] Pamukkale University, Medical Faculty, Department of Medical BiologyDenizli, Turkey. [Sen Turk, Nilay] Pamukkale University, Faculty of Medicine, Department of PathologyDenizli, Turkey. [Cetin, Ozan Gokhan] Pamukkale University, Medical Faculty, Department of Medical Genetics, Doktorlar Cad. Kat:3, BayramyeriDenizli, Turkey. [Kelten, Canan] Pamukkale University, Faculty of Medicine, Department of PathologyDenizli, Turkey. [Tuna, Burcin] Dokuz Eylul University, Faculty of Medicine, Department of PathologyIzmir, Turkey. [Sarsik, Banu] Ege University, Faculty of Medicine, Department of PathologyIzmir, Turkey. [Tepeli, Emre] Pamukkale University, Medical Faculty, Department of Medical Genetics, Doktorlar Cad. Kat:3, BayramyeriDenizli, Turkey. RP Duzcan, F (reprint author), Pamukkale University, Medical Faculty, Department of Medical Genetics, Denizli, Turkey. EM fusunduzcan@yahoo.com;fduzcan@pau.edu.tr CR Eble J, Sauter G, Epstein J, Sesterhenn I, 2004, Tumours of the urinary system and male genital organs. In: Eble J, Sauter G, Epstein J, Sesterhenn I, eds, World health organization classification of tumours. IARC, Lyon, pp 27–29 Delahunt B, Eble JN, 1997, Papillary renal cell carcinoma: a clinicopathologic and immunohistochemical study of 105 tumors. Mod Pathol 10(6):537–544 Murphy WM, Grignon DJ, Perlman EJ, 2004, Tumors of the kidney, bladder and related urinary structures. AFIP atlas of tumor pathology, 4th series. American Registry of Pathology, Washington, pp 123–130 Petersen RO, Sesterhenn IA, Davis CJ, 2009, Urologic pathology, 3rd edn. Lippincott Williams and Wilkins, Philadelphia, pp 54–56 Delahunt B, Eble J, McCredis M, Bethwaite P, Stewart J, Bilous M, 2001, Morphologic typing of papillary renal cell carcinoma: comparison of growth kinetics and patient survival in 66 cases. Hum Pathol 32:590–595 Pignot G, Elie C, Conquy S et al, 2007, Survival analysis of 130 patients with papillary renal cell carcinoma: prognostic utility of type 1 and type 2 subclassification. Urology 69:230–235 Jiang F, Richter J, Schraml P, Bubendorf L, Gasser T, Sauter G, Mihatsch MJ, Moch H, 1998, Chromosomal imbalances in papillary renal cell carcinoma. Am J Pathol 153:1467–1473 Sanders ME, Mick R, Tomaszewski JE, Barr FG, 2002, Unique patterns of allelic imbalance distinguish type 1 from type 2 sporadic papillary renal cell carcinoma. Am J Pathol 161:997–1005 Kovacs G, Fuzesi L, Emanual A, Kung HF, 1991, Cytogenetics of papillary renal cell tumors. Genes Chromosom Cancer 3:249– 255 Glukhova L, Goguel AF, Chudoba I, Angevin E, Pavon C, Terrier-Lacombe MJ, Meddeb M, Escudier B, Bernheim A, 1998, Overpresentation of 7q31 and 17q in renal cell carcinomas. Genes Chromosom Cancer 22:171–178 Hughson MD, Dickman K, Bigler SA, Meloni AM, Sandberg AA, 1998, Clear-cell and papillary carcinoma of the kidney: an analysis of chromosome 3, 7, and 17 abnormalities by microsatellite amplification, cytogenetics, and fluorescence in situ hybridization. Cancer Genet Cytogenet 106:93–104 Hughson MD, Bigler S, Dickman K, Kovacs G, 1999, Renal cell carcinoma of end-stage renal disease: an analysis of chromosome 3, 7, and 17 abnormalities by microsatellite amplification. Mod Pathol 12:301–309 Dekel Y, Frede T, Kugel V, Neumann G, Rassweiler J, Koren R, 2005, Human DNA topoisomerase II-alpha expression in laporospically treated renal cell carcinoma. Oncol Rep 14:271– 274 Cooper GM, Hausman RE, 2006, Genomik DNA’nin replikasyonu, korunmasi ve yeniden duzenlenmesi. In: Sakizli M, Atabey N, eds, Hucre-Molekuler Yaklasim. Translational, 3rd edn. Izmir Tip Kitapevi, Izmir, pp 186–187 Jarvinen TAH, Liu ET, 2003, Topoisomerase IIα gene, TOP2A, amplification and deletion in cancer—more common than anticipated. Cytopathology 14:309–313 Varis A, Wolf M, Monni O, Vakkari ML, Kokkola A, Moskalk C, Frierson H Jr, Powell SM, Knuutila S, Kalioniemi A, El-Rifai W, 2002, Targets of gene amplification and overexpression at 17q in gastric cancer. Cancer Res 62:2625–2629 Wikman H, Kettunen E, Seppanen JK, Karjalainen A, Hollmen J, Anttila S, Knuutila S, 2002, Identification of differentially expressed genes in pulmonary adenocarcinoma by using cDNA array. Oncogene 21:5804–5813 Yang XJ, Tan MH, Kim HL et al, 2005, A molecular classification of papillary renal cell carcinoma. Cancer Res 65:5628–5637 Vang Nielsen K, Muller S, Moller S, Schonau A, Balslev E, Knoop AS, Ejlertsen B, 2010, Aberrations of ERBB2 and TOP2A genes in breast cancer. Mol Oncol 4:161–168 Reese DM, Small EJ, Magrane G, Waldman FM, Chew K, Sudilovsky D, 2001, HER2 protein expression and gene amplification in androgen-independent prostate cancer. Am J Clin Pathol 116:234–239 Press MF, Finn RS, Cameron D, Di Leo A, Geyer CE, Villalobos IE, Santiago A, Guzman R, Gasparyan A, Ma Y, Danenberg K, Martin AM, Williams L, Oliva C, Stein S, Gagnon R, Arbushites M, Koehler MT, 2008, HER-2 gene amplification, HER-2 and epidermal growth factor receptor mRNA and protein expression, and lapatinib efficacy in women with metastatic breast cancer. Clin Cancer Res 14:7861–7870 Caner V, Turk NS, Duzcan F, Tufan NL, Kelten EC, Zencir S, Dodurga Y, Bagci H, Duzcan SE, 2008, No strong association between HER-2/neu protein overexpression and gene amplification in high-grade invasive urothelial carcinomas. Pathol Oncol Res 14:261–266 Jarvinen TA, Tanner M, Barlund M, Borg A, Isola J, 1999, Characterization of topoisomerase II alpha gene amplification and deletion in breast cancer. Genes Chromosom Cancer 26:142–150 Jarvinen TA, Tanner M, Rantanen V, Barlund M, Borg A, Grenman S, Isola J, 2000, Amplification and deletion of topoisomerase IIalpha associate with ErbB-2 amplification and affect sensitivity to topoisomerase II inhibitor doxorubicin in breast cancer. Am J Pathol 156:839–847 Di Leo A, Isola J, 2003, Topoisomerase II alpha as a marker predicting the efficacy of anthracyclines in breast cancer: are we at the end of the beginning? Clin Breast Cancer 4:179–186 Bhargava R, Lal P, Chen B, 2005, HER-2/neu and topoisomerase IIa gene amplification and protein expression in invasive breast carcinomas: chromogenic in situ hybridization and immunohistochemical analyses. Am J Clin Pathol 123:889–895 Wang Q, Zambetti GP, Suttle DP, 1997, Inhibition of DNA topoisomerase II alpha gene expression by the p53 tumor suppressor. Mol Cell Biol 17:389–397 Wu Z, Reed RF, Parker Suttle D, 2003, Nuclear factor-y binding to the topoisomerase IIα promoter is inhibited by both the p53 tumor suppressor and anticancer drugs. Mol Pharmacol 63:359– 367 Baylin SB, Jones PA, 2007, Epigenetic determinants of cancer. In: Allis CD, Jenuwein T, Reinberg D, eds, Epigenetics. Cold Spring Harbor, pp 457–476 Barrandon C, Spiluttini B, Bensaude O, 2008, Non-coding RNAs regulating the transcriptional machinery. Biol Cell 100:83–95 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 697 EP 703 DI 10.1007/s12253-011-9372-0 PG 7 ER PT J AU Chu, T Chen, X Yu, J Xiao, J Fu, Z AF Chu, Tongwei Chen, Xiaoyang Yu, Jie Xiao, Jianwen Fu, Zhou TI Extracellular Matrix Metalloproteinase Inducer is a Negative Prognostic Factor of Pediatric Medulloblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Medulloblastoma; Extracellular matrix metalloproteinase inducer; Immunohistochemistry; Prognosis ID Medulloblastoma; Extracellular matrix metalloproteinase inducer; Immunohistochemistry; Prognosis AB Medulloblastoma (MB) is the most common embryonal CNS tumor of childhood. Its survival rates have significantly improved over the years due to developments in diagnostic techniques and therapeutic strategies. However, it is still an important cause of cancer-related deaths in children. Extracellular matrix metalloproteinase inducer (EMMPRIN) is a member of the immunoglobulin family and a glycoprotein enriched on the surface of many types of tumor cells. Therefore, the aim of this study was to investigate whether the expression patterns of EMMPRIN may predict the clinical prognosis in MB. EMMPRIN expression in a series of 56 MB with various grades and pathological types was analyzed by immunohistochemical staining on paraffin-embedded sections. Then, the correlation of EMMPRIN expression patterns with clinicalpathological features of patients and its prognostic relevance were determined. Immunohistochemistry revealed that the positive expression rate of EMMPRIN in MB (75.0%, 42/56) was significantly higher than that in normal cerebellums (6.7%, 2/30, p<0.001). In addition, EMMPRIN expression in MB was up-regulated in higher metastatic stage (p<0.01), aggressive histopathological type (p<0.005), necrosis (p<0.01), as well as with undifferentiated tumor (p<0.01). Furthermore, over-expression of EMMPRIN correlated significantly with poor prognosis (0.01 T KRAS transversion in patients with constitutional MUTYH biallelic mutations. Cancer Lett 274:266–270 Voz ML, Agten NS, Van de Ven WJ et al, 2000, PLAG1, the main translocation target in pleomorphic adenoma of the salivary glands, is a positive regulator of IGF-II. Cancer Res 60:106–113 Van Dyck F, Declercq J, Braem CV et al, 2007, PLAG1, the prototype of the PLAG gene family: versatility in tumour development. Int J Oncol 30:765–774 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 765 EP 769 DI 10.1007/s12253-011-9384-9 PG 5 ER PT J AU Ngow, AH Wan Khairina, MNW AF Ngow, A Harris Wan Khairina, Mohd Nowalid Wan TI Cardiac Metastasis: A Rare Involvement of Primitive Neuroectodermal Tumour of the Lung SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE PNET; Cardiac Metastasis; Ewing’s Sarcoma; Extraosseous Ewing’s Sarcoma ID PNET; Cardiac Metastasis; Ewing’s Sarcoma; Extraosseous Ewing’s Sarcoma AB A 15 year-old adolescent was referred with 2 month history of worsening of breathlessness and haemoptysis. He also reported constitutional symptoms of fever, poor appetite and weight loss. The chest roentgenogram showed a massive right pleural effusion with apparent cardiomegaly. The cardiac silhouette over the right heart border was obliterated and the mediastinum was widened. Computed tomogram of the thorax showed a bulky heterogeneous mass in the right lung with extension to the heart. Subsequent CT guided lung biopsy revealed Primitive Neuroectodermal tumour (PNET). Here, we illustrate the clinical course of an aggressive pulmonary PNET with lethal cardiac metastasis. C1 [Ngow, A Harris] International Islamic University Malaysia/Hospital Tengku Ampuan Afzan, Jalan Hospital, Kulliyyah of Medicine, 25150 Kuantan, Pahang, Malaysia. [Wan Khairina, Mohd Nowalid Wan] Ministry of Health Malaysia, Hospital Tengku Ampuan Afzan, 25150 Kuantan, Pahang, Malaysia. RP Ngow, AH (reprint author), International Islamic University Malaysia/Hospital Tengku Ampuan Afzan, Jalan Hospital, Kulliyyah of Medicine, 25150 Kuantan, Malaysia. EM harrisngow@gmail.com CR Besirli K, Arslan C, Tuzun H, Oz B, 2000, The primitive Neuroectodermal tumour of the heart. Eur J Cardiothorac Surg 18:619–621 Charney DA, Charney JM, Ghali VS, Teplitz C, 1996, Primitive Neuroectodermal tumour of the myocardium: a case report, review of the literature, immunohistochemical and ultrastructural study. Hum Pathol 27:1365–1369 Hendershot E, 2005, Treatment approaches for metastatic Ewing’s sarcoma: a review of the literature. J Pediatr Oncol Nurs 22:339– 352 Askin FB, Rosai J, Sibley RK, Dehner LP, McAlister WH, 1979, Malignant small cell tumour of the thoracopulmonary region in childhood. Cancer 43:2438–2451 Reynen K, Kockeritz U, Strasser RH, 2004, Metastases to the heart. Ann Oncol 15:375–381 Hawkins DS, Felgenhauer J, Park J, Kreissman S, Thomson B, Douglas J et al, 2002, Peripheral blood stem cell support reduces the toxicity of intensive chemotharpy for children and adolescents with metastatic sarcomas. Cancer 95:1354–1365 Marley EF, Liapis H, Humphry PA, Nadler RB, Siegel CL, Zhu X, Brandt SM, Dehner LP, 1997, Primitive Neuroectodermal tumor of the kidney another enigma: a pathologic, immunohistochemical and molecular diagnostic study. Am J Surg Pathol 21:354–359 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 771 EP 774 DI 10.1007/s12253-010-9328-9 PG 4 ER PT J AU Ioannidis, O Papaemmanouil, S Chatzopoulos, S Paraskevas, G Konstantara, A Kotronis, A Kakoutis, E Makrantonakis, A AF Ioannidis, Orestis Papaemmanouil, Styliani Chatzopoulos, Stavros Paraskevas, George Konstantara, Athina Kotronis, Anastasios Kakoutis, Emmanouil Makrantonakis, Apostolos TI Giant Bilateral Symptomatic Adrenal Myelolipomas Associated with Congenital Adrenal Hyperplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Adrenal gland; Bilateral myelolipomma; Congenital adrenal hyperplasia; 21-hydroxylase deficiency; Giant myelolipoma; Symptomatic myelolipoma ID Adrenal gland; Bilateral myelolipomma; Congenital adrenal hyperplasia; 21-hydroxylase deficiency; Giant myelolipoma; Symptomatic myelolipoma AB Adrenal myelolipoma is an uncommon, benign, biochemically non-functioning and endocrinologically inactive tumor composed of variable amounts of mature adipose tissue and scattered islands of haemopoietic elements, including erythroid, myeloid and lymphoid series, as well as megakaryocytes. Diagnosis of myelolipomas is based on imaging, with ultrasonography, CT and MRI being effective in more than 90% of cases. Differential diagnosis includes other containing fat adrenal masses such as teratoma, lipoma and liposarcoma. The optimal treatment depends on the size and symptoms of the myelolipoma. For incidentally discovered, asymptomatic adrenal myelolipomas smaller than 4 cm surveillance seems to be enough while symptomatic, complicated, hormonally active and larger than 7 cm myelolipomas, should be surgically removed. We present a case of giant bilateral symptomatic adrenal myelolipomas associated with congenital adrenal hyperplasia. A 34 year old female, with congenital adrenal hyperplasia because of 21-hydroxylase deficiency, presented with diffuse abdominal pain and vomiting. Physical examination revealed hirsutism, pronounced virilization and palpable masses both on the right and left abdominal area. The abdominal CT demonstrated bilateral large masses in the anatomical position of the adrenal glands with densities indicating adipose tissue. The differential diagnosis was between myelolipoma and liposarcoma. For diagnostic and also therapeutical reasons, as the masses were large and symptomatic and causing pressure to the surrounding structures, the patient was submitted to laparotomy for bilateral excision. Histopathological examination established the diagnosis of adrenal myelolipoma. C1 [Ioannidis, Orestis] General Regional Hospital ‘George Papanikolaou’, First Surgical Department, Alexandrou Mihailidi 13, 54640 Thessaloniki, Greece. [Papaemmanouil, Styliani] General Regional Hospital ‘George Papanikolaou’, Department of PathologyThessaloniki, Greece. [Chatzopoulos, Stavros] General Regional Hospital ‘George Papanikolaou’, First Surgical Department, Alexandrou Mihailidi 13, 54640 Thessaloniki, Greece. [Paraskevas, George] General Regional Hospital ‘George Papanikolaou’, First Surgical Department, Alexandrou Mihailidi 13, 54640 Thessaloniki, Greece. [Konstantara, Athina] General Regional Hospital ‘George Papanikolaou’, First Surgical Department, Alexandrou Mihailidi 13, 54640 Thessaloniki, Greece. [Kotronis, Anastasios] General Regional Hospital ‘George Papanikolaou’, First Surgical Department, Alexandrou Mihailidi 13, 54640 Thessaloniki, Greece. [Kakoutis, Emmanouil] General Regional Hospital ‘George Papanikolaou’, First Surgical Department, Alexandrou Mihailidi 13, 54640 Thessaloniki, Greece. [Makrantonakis, Apostolos] General Regional Hospital ‘George Papanikolaou’, First Surgical Department, Alexandrou Mihailidi 13, 54640 Thessaloniki, Greece. RP Ioannidis, O (reprint author), General Regional Hospital ‘George Papanikolaou’, First Surgical Department, 54640 Thessaloniki, Greece. EM telonakos@hotmail.com CR Daneshmand S, Quek ML, 2006, Adrenal myelolipoma: diagnosis and management. Urol J 3:71–74 Osborn M, Smith M, Senbanjo T, Crofton M, Robinson S, Rajan P, 2002, Adrenal myelolipoma—clinical, radiological and cytological findings: a case report. Cytopathology 13:242–246 Cyran KM, Kenney PJ, Memel DS, Yacoub I, 1996, Adrenal myelolipoma. AJR Am J Roentgenol 166:395–400 Fujiwara R, Onishi T, Shimada A, Nakai T, Miyabo S, Nakakugi K, Yamamoto M, 1993, Adrenal myelolipoma: comparison of diagnostic imaging and pathological findings. Intern Med 32:166– 170 Talwalkar SS, Shaheen SP 2nd, 2006, Extra-adrenal myelolipoma in the renal hilum: a case report and review of the literature. Arch Pathol Lab Med 130:1049–1052 Lam KY, Lo CY, 2001, Adrenal lipomatous tumours: a 30 year clinicopathological experience at a single institution. J Clin Pathol 54:707–712 Repassy DL, Csata S, Sterlik G, Ivanyi A, 2001, Giant adrenal myelolipoma. Pathol Oncol Res 7:72–73 Nermoen I, Folling I, Vegge K, Larmo A, Nedrebo BG, Husebye ES, Lovas K, 2009, Two adults with adrenal myelolipoma and 21-hydroxylase deficiency. Case Report Med 2009:916891 Kalidindi RS, Hattingh L, 2006, Bilateral giant adrenal myelolipomas. Abdom Imaging 31:125–127 Palmer WE, Gerard-McFarland EL, Chew FS, 1991, Adrenal myelolipoma. AJR Am J Roentgenol 156:724 O’Daniel-Pierce ME, Weeks JA, McGrath PC, 1996, Giant adrenal myelolipoma. South Med J 89:1116–1118 Gould JD, Mitty HA, Pertsemlidis D, Szporn AH, 1987, Adrenal myelolipoma: diagnosis by fine-needle aspiration. AJR Am J Roentgenol 148:921–922 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 775 EP 778 DI 10.1007/s12253-010-9330-2 PG 4 ER PT J AU Tamas, L Sari, E Repassy, G Szabo, P Bagdi, E Krenacs, L Demeter, J AF Tamas, Laszlo Sari, Eszter Repassy, Gabor Szabo, Peter Bagdi, Eniko Krenacs, Laszlo Demeter, Judit TI Spontaneous Remission in Localized Diffuse Large B-cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Diffuse large B-cell lymphoma; Spontaneous remission; Activated B-cell type DLBLC; Root of tongue; Untreated lymphoma ID Diffuse large B-cell lymphoma; Spontaneous remission; Activated B-cell type DLBLC; Root of tongue; Untreated lymphoma AB Diffuse large B-cell lymphoma (DLBCL) is an aggressive neoplastic disease of the lymphatic system, the activated B-cell type of this disease is likely to have a substantially worse prognosis. In this study, we report the favorable outcome of the activated B-cell type of DLBCL, though untreated, 7 years after diagnosis. In 2003, DLBCL localized to the root of tongue was found in the patient complaining of dysphonia and a pharyngeal globus perception but the patient did not agree to get any active hematological treatment. During the following years, the patient did not have any complaints. At the otorhinolaryngological control examination, in 2010, she was complaintfree, had normal laboratory parameters. Moreover a PETCT scan did not reveal metabolic activity relating to malignancy. The extraordinary disease process can be explained by the spontaneous regression of the activated B-cell type DLBCL. Spontaneous regression of oral lymphoma has been published only exceptionally. To our knowledge, no report of spontaneous regression of activated B-cell type DLBLC has been reported. C1 [Tamas, Laszlo] Jahn Ferenc South-Pest Hospital, Department of Oto-Rhino-Laryngology, Koves u. 1., 1204 Budapest, Hungary. [Sari, Eszter] Semmelweis University, 1st Department of Internal MedicineBudapest, Hungary. [Repassy, Gabor] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Szabo, Peter] PET-CT Medical Diagnostic LtdBudapest, Hungary. [Bagdi, Eniko] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Krenacs, Laszlo] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Demeter, Judit] Semmelweis University, 1st Department of Internal MedicineBudapest, Hungary. RP Tamas, L (reprint author), Jahn Ferenc South-Pest Hospital, Department of Oto-Rhino-Laryngology, 1204 Budapest, Hungary. EM tamlaci@freemail.hu CR Alizadeh AA, Eisen MB, Davis RE et al, 2000, Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 403:503–511 Chang CC, McClintock S, Ronald P et al, 2004, Immunohistochemical expression patterns of germinal center and activation Bcell markers correlate with prognosis in diffuse large B-cell lymphoma. Am J Surg Pathol 28:464–470 Hans CP, Weisenburger DD, Greiner TC et al, 2004, Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood 103:275–282 Papac RJ, 1999, Spontaneous regression of cancer. Cancer Treatment Reviews 22:395–423 Sengupta N, MacFie TS, MacDonald TT et al, 2010, Cancer immunoediting and “spontaneous” tumor regression. Path Res and Pract 206:1–8 Fujimoto K, Endo T, Nishio M et al, 2009, Complete remission of splenic marginal zone lymphoma after an acute flare-up of hepatitis B in a hepatitis B virus carrier. Int J Hematol 90:601–604 Drobyski WR, Qazi, 1989, Spontaneous regression in non- Hodgkin’s lymphoma: clinical and pathogenic considerations. Am J Hem 31:138–141 Gattiker HH, Wiltshaw E, Galton DAG, 1980, Spontaneous regression in non-Hodgkin ’s lymphoma. Cancer 45:2627– 2632 Krikorian JG, Carol S, Cooney DP et al, 1980, Spontaneous regression of non-Hodgkin’s Lymphoma: a report of nine cases. Cancer 46:2093–2099 Abe R, Ogawa K, Maruyama Y et al, 2007, Spontaneous regression of diffuse large B-cell lymphoma harbouring epsteinbarr virus: a case report and review of the literature. J Clin Exp Hematopathol 47:23–26 Evans LS, Hancock B, 2003, Non-Hodgkin lymphoma. The Lancet Vol 362:139–146 Daly RM, Healy CM, Toner ME et al, 2008, Spontaneous regression of non-Hodgkin’s lymphoma int he oral cavity after incisional biopsy. Br J Or Maxillofac Surg 46:223–225 Savarrio L, Gibosn J, Dunlop DJ et al, 1999, Spontaneous regression of an anaplastic large cell lymphoma int he oral cavity: first reported case and review of the literature. Or Onc 35:609–613 Koga M, Kusukawa J, Havabuchi N, 2003, Spontaneous regression of extranodal malignant lymphoma occurred int he gingiva. Or Onc 39:323–324 Iqbal J, Greiner TC, Patel K et al, 2009, Gene expression profiling in lymphoma diagnosis and management. Best Pract & Res Clin Haem 22:191–210 Camilleri-Broet S, Criniere E, Broet P et al, 2006, A uniform activated B-cell-like immunophenotpye might explain the poor prognosis of primary central nervous system lymphomas: analysis of 83 cases. Blood 107:190–196 Lu X, Nechushtan H, Ding F et al, 2005, Distinct IL-4-induced gene expression, proliferation, and intracellular signaling in germinal center B-cell like and activated B-cell –like diffuse large-cell lymphomas. Blood 105(7):2924–2932 Barrans SL, Carter I, Owen RG et al, 2002, Germinal center phenotype and bcl-2 expression combined with the International Prognostic Index improves patient risk stratification in diffuse large B-cell lymphoma. Blood 99:1136–1143 Armitage JO, 2008, Is lymphoma occuring int he elderly the same disease? Leuk Lymphoma 49:14–6 Gutierrez A, Mestre F, Perez-Manga G, Rodriguez J, 2010, Diffuse large B-cell lymphoma in the older. Crit Rev Oncol HematolMar 18. [Epub ahead of print], DOI 10.1016/j.critrevonc.2010.02.009 Liu Y-H, Xu FP, Zhuang HG et al, 2008, Clinicopathologic significance of immunphenotypic profiles related to germinal center and activation B-cell differentiation in diffuse large B-cell lymphoma from Chinese patients. Hum Path 39:875–884 Pfreundschuh M, 2010, How I treat elderly patients with diffuse large B-cell lymphoma. Blood 116:5103–10 Miller TP, Unger JM, Spier CM et al, 2004, Effect of adding rituximab to three cycles of CHOP plus involved –field radiotherapy for limited –stage agressive diffuse B-cell lymphoma, SWOG-0014). Blood 104:48a Persky DO, Unger JM, Spier CM et al, 2008, Phase II study of rituximab plus three cycles of CHOP and involved –field radiotherapy for patients with limited-stage agressive B-cell lymphoma: Southwest Oncology Group study 0014. J Clin Oncol 26(14):2258–2263 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2011 VL 17 IS 3 BP 779 EP 784 DI 10.1007/s12253-011-9379-6 PG 6 ER PT J AU Sikalidis, KA Varamini, B AF Sikalidis, K Angelos Varamini, Behzad TI Roles of Hormones and Signaling Molecules in Describing the Relationship Between Obesity and Colon cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Colon cancer; Inflammation; Insulin; Leptin; Obesity ID Colon cancer; Inflammation; Insulin; Leptin; Obesity AB Colon cancer represents a highly prevalent disease in the Western world. While dietary and lifestyle recommendations remain important factors in disease prevention and treatment, epidemiological data have made it clear that obesity and excess body weight remain significant risk factors for the disease. A number of potential direct and indirect relationships exist between obesity and increased risk of colon cancer. Several mechanisms which appear promising and warrant further investigation are discussed here, specifically the modifying role of insulin and insulin-like growth factors, leptin, adipose-tissue induced changes in estrogens and androgens, and inflammatory molecules. A brief review of these hormones and signaling molecules and their action in colon cancer development is described. A thorough integration and understanding of the mechanisms of action these systems exert on colonic epithelia will be important in designing studies and experiments aimed at elucidating disease etiology for prevention and treatment. C1 [Sikalidis, K Angelos] Cornell University, Division of Nutritional Sciences, 214 Savage Hall, 14853 Ithaca, NY, USA. [Varamini, Behzad] University of Pennsylvania, School of Medicine, 19104 Philadelphia, PA, USA. RP Sikalidis, KA (reprint author), Cornell University, Division of Nutritional Sciences, 14853 Ithaca, USA. EM as545@cornell.edu CR World Health Organization official website National Cancer Institute official website Jemal A, Murray T, Ward E et al, 2005, Cancer Statistics, 2005. CA Cancer J Clin 55:10–30 Forte A, De Sanctis R, Leonetti G, Manfredelli S, Urbano V, Bezzi M, 2008, Dietary chemoprevention of colorectal cancer. Ann Ital Chir 79:261–267 Bird CL, Frankl HD, Lee ER, Haile RW, 1998, Obesity, weight gain, large weight changes, and adenomatous polyps of the left colon and rectum. Am J Epidemiol 147:670–680 Bostick RM, Potter JD, Kushi LH et al, 1994, Sugar, meat, and fat intake, and non-dietary risk factors for colon cancer incidence in Iowa women, United States). Cancer Causes Control 5:38–52 Dietz AT, Newcomb PA, Marcus PM, Storer BE, 1995, The association of body size and large bowel cancer risk in Wisconsin, United States, women. Cancer Causes Control 6:30–36 Giovannucci E, Colditz GA, Stampfer MJ, Willett WC, 1996, Physical activity, obesity, and risk of colorectal adenoma in women, United States). Cancer Causes Control 7:253–263 Martinez ME, Giovannucci E, Spiegelman D, Hunter DJ, Willett WC, Colditz GA, 1997, Leisure-time physical activity, body size, and colon cancer in women. Nurses’ Health Study Research Group. J Natl Cancer Inst 89:948–955 Murphy TK, Calle EE, Rodriguez C, Khan HS, Thun MJ, 2000, Body mass index and colon cancer mortality in a large prospective study. Am J Epidemiol 152:847–854 Takahashi M, Fukuda K, Sugimura T, Wakabayashi K, 1998, β-Catenin is frequently mutated and demonstrates altered cellular location in azoxymethane-induced rat colon tumors. Cancer Res 58:42–46 Yamada Y, Yoshimi N,Hirose Yet al, 2000, Frequent β-catenin gene mutations and accumulations of the protein in the putative preneoplastic lesions lackingmacroscopic aberrant crypt foci appearance, in rat colon carcinogenesis. Cancer Res 60:3323–3327 Lee WM, Lu S, Medline A, Archer MC, 2001, Susceptibility of lean and obese Zucker rats to tumorigenesis induced by N-methyl- N-nitrosourea. Cancer Lett 162:155–160 Wargovich MJ, Jimenez A, McKee K et al, 2000, Efficacy of potential chemopreventive agents on rat colon aberrant crypt formation and progression. Carcinogenesis 21:1149–1155 Lanas A, Ferrandez A, 2009, NSAIDs and the colon. Curr Opin Gastroenterol 25:44–49 Yang CS, Lambert JD, Ju J, Lu G, Sang S, 2007, Tea and cancer prevention: molecular mechanisms and human relevance. Toxicol Appl Pharmacol 224:265–273 Holt PR, 2008, New insights into calcium, dairy and colon cancer. World J Gastroenterol 14:4429–4433 Franco A, Sikalidis AK, Solis Herruzo JA, 2005, Colorectal cancer: influence of diet and lifestyle factors. Rev Esp Enferm Dig 97:432–448 Ferguson LR, Philpott M, Karunasinghe N, 2004, Dietary cancer and prevention using antimutagens. Toxicology 198:147–159 Bruce WR, Wolever TMS, Giacca A, 2000, Mechanisms linking diet and colorectal cancer: the possible role of insulin resistance. Nutr Cancer 37:19–26 Giugliano D, Ceriello A, Esposito K, 2008, Are there specific treatments for the metabolic syndrome? Am J Clin Nutr 87:8–11 Komninou D, Ayonote A, Richie JP Jr, Rigas B, 2003, Insulin resistance and its contribution to colon carcinogenesis. Exp Biol Med, Maywood, 228:396–405 Stipanuk MH, 2000, Biochemical and physiological aspects of human nutrition. Saunders, Philadelphia Kopelman PG, 1994, Hormones and obesity. Baillieres Clin Endocrinol Metab 8:549–575 Nelson WG, De Marzo AM, DeWeese TL, Isaacs WB, 2004, The role of inflammation in the pathogenesis of prostate cancer. J Urol 172:S6–11, discussion S11-2 Renehan AG, Frystyk J, Flyvbjerg A, 2006, Obesity and cancer risk: the role of the insulin-IGF axis. Trends Endocrinol Metab 17:328–336 Sigma Aldrich USA official website Giovannucci E, 2003, Nutrition, insulin, insulin-like growth factors and cancer. Horm Metab Res 35:694–704 Pass C, MacRae VE, Ahmed SF, Farquharson C, 2009, Inflammatory cytokines and the GH/IGF-I axis: novel actions on bone growth. Cell Biochem Funct 27:119–127 Rajah R, Valentinis B, Cohen P, 1997, Insulin-like growth factor, IGF)-binding protein-3 induces apoptosis and mediates the effects of transforming growth factor-beta 1 on programmed cell death through a p53- and IGF-independent mechanism. J Biol Chem 272:12181–12188 Rechler MM, 1997, Growth inhibition by insulin-like growth factor, IGF, binding protein-3—what’s IGF got to do with it? Endocrinology 138:2645–2647 Jones JI, Clemmons DR, 1995, Insulin-like growth factors and their binding proteins: biological actions. Endocr Rev 16:3–34 Bjork J, Nilsson J, Hultcrantz R, Johansson C, 1993, Growthregulatory effects of sensory neuropeptides, epidermal growth factor, insulin, and somatostatin on the non-transformed intestinal epithelial cell line IEC-6 and the colon cancer cell line HT 29. Scand J Gastroenterol 28:879–884 Guo YS, Narayan S, Yallampalli C, Singh P, 1992, Characterization of insulin-like growth factor I receptors in human colon cancer. Gastroenterology 102:1101–1108 Koenuma M, Yamori T, Tsuruo T, 1989, Insulin and insulin-like growth factor 1 stimulate proliferation of metastatic variants of colon carcinoma 26. Jpn J Cancer Res 80:51–58 Pollak MN, Perdue JF, Margolese RG, Baer K, Richard M, 1987, Presence of somatomedin receptors on primary human breast and colon carcinomas. Cancer Lett 38:223–230 Watkins LF, Lewis LR, Levine AE, 1990, Characterization of the synergistic effect of insulin and transferrin and the regulation of their receptorsonahumancoloncarcinoma cell line. Int JCancer 45:372–375 Aaronson SA, 1991, Growth factors and cancer. Science 254:1146–1153 Warren RS, Yuan H, Matli MR, Ferrara N, Donner DB, 1996, Induction of vascular endothelial growth factor by insulin-like growth factor 1 in colorectal carcinoma. J Biol Chem 271:29483–29488 Dalgleish AG, O’Byrne K, 2006, Inflammation and cancer: the role of the immune response and angiogenesis. Cancer Treat Res 130:1–38 Poullis A, Foster R, Shetty A, Fagerhol MK, Mendall MA, 2004, Bowel inflammation as measured by fecal calprotectin: a link between lifestyle factors and colorectal cancer risk. Cancer Epidemiol Biomark Prev 13:279–284 Burgering BM, Medema RH, Maassen JA et al, 1991, Insulin stimulation of gene expression mediated by p21ras activation. EMBO J 10:1103–1109 Jhun BH, Meinkoth JL, Leitner JW, Draznin B, Olefsky JM, 1994, Insulin and insulin-like growth factor-I signal transduction requires p21ras. J Biol Chem 269:5699–5704 Gutierrez L, Magee AI, Marshall CJ, Hancock JF, 1989, Posttranslational processing of p21ras is two-step and involves carboxylmethylation and carboxy-terminal proteolysis. EMBO J 8:1093–1098 Goalstone ML, Wall K, Leitner JW et al, 1999, Increased amounts of farnesylated p21Ras in tissues of hyperinsulinaemic animals. Diabetologia 42:310–316 Leitner JW, Kline T, Carel K, Goalstone M, Draznin B, 1997, Hyperinsulinemia potentiates activation of p21Ras by growth factors. Endocrinology 138:2211–2214 Ahima RS, Antwi DA, 2008, Brain regulation of appetite and satiety. Endocrinol Metab Clin North Am 37:811–823 Jequier E, 2002, Leptin signaling, adiposity, and energy balance. Ann NY Acad Sci 967:379–388 Stattin P, Lukanova A, Biessy C et al, 2004, Obesity and colon cancer: does leptin provide a link? Int J Cancer 109:149–152 Jaffe T, Schwartz B, 2008, Leptin promotes motility and invasiveness in human colon cancer cells by activating multiple signal-transduction pathways. Int J Cancer 123:2543–2556 Tran TT, Medline A, Bruce WR, 1996, Insulin promotion of colon tumors in rats. Cancer Epidemiol Biomark Prev 5:1013– 1015 Ahima RS, Flier JS, 2000, Adipose tissue as an endocrine organ. Trends Endocrinol Metab 11:327–332 Zhu Z, Jiang W, Thompson HJ, 2002, An experimental paradigm for studying the cellular and molecular mechanisms of cancer inhibition by energy restriction. Mol Carcinog 35:51–56 Attoub S, Noe V, Pirola L et al, 2000, Leptin promotes invasiveness of kidney and colonic epithelial cells via phosphoinositide 3-kinase-, Rho-, and Rac-dependent signaling pathways. FASEB J 14:2329–2338 Morton NM, Emilsson V, Liu Y-L, Cawthorne MA, 1998, Leptin action in intestinal cells. J Biol Chem 273:26194–26201 Slattery ML, Ballard-Barbash R, Edwards S, Caan BJ, Potter JD, 2003, Body mass index and colon cancer: an evaluation of the modifying effects of estrogen, United States). Cancer Causes Control 14:75–84 Butler AA, Blakesley VA, Poulaki V, Tsokos M, Wood TL, LeRoith D, 1998, Stimulation of tumor growth by recombinant human insulin-like growth factor-I, IGF-I, is dependent on the dose and the level of IGF-I receptor expression. Cancer Res 58:3021–3027 Khandwala HM, McCutcheon IE, Flyvbjerg A, Friend KE, 2000, The effects of insulin-like growth factors on tumorigenesis and neoplastic growth. Endocr Rev 21:215–244 Yavari K, Taghikhani M, Ghannadi Maragheh M, Mesbah-Namin SA, Babaei MH, 2010, Downregulation of IGF-IR expression by RNAi inhibits proliferation and enhances chemosensitization of human colon cancer cells. Int J Colorectal Dis 25:9–16 Polderman KH, Gooren LJ, Asscheman H, Bakker A, Heine RJ, 1994, Induction of insulin resistance by androgens and estrogens. J Clin Endocrinol Metab 79:265–271 Di Domenico M, Castoria G, Bilancio A, Migliaccio A, Auricchio F, 1996, Estradiol activation of human colon carcinoma-derived Caco-2 cell growth. Cancer Res 56:4516–4521 Rosen N, Bolen JB, Schwartz AM, Cohen P, DeSeau V, Israel MA, 1986, Analysis of pp 60c-src protein kinase activity in human tumor cell lines and tissues. J Biol Chem 261:13754–13759 Talamonti MS, Roh MS, Curley SA, Gallick GE, 1993, Increase in activity and level of pp 60c-src in progressive stages of human colorectal cancer. J Clin Invest 91:53–60 Aligayer H, Boyd DD, Heiss MM, Abdalla EK, Curley SA, Gallick GE, 2002, Activation of Src kinase in primary colorectal carcinoma: an indicator of poor clinical prognosis. Cancer 94:344–351 Mathew S, George SP, Wang Y et al, 2008, Potential molecular mechanism for c-Src kinase-mediated regulation of intestinal cell migration. J Biol Chem 283:22709–22722 Gordon D, 1999, Inhibitor of MAP kinase activation blocks colon cancer growth. Gastroenterology 117:523 Park KS, Kim NG, Kim JJ, Kim H, Ahn YH, Choi KY, 1999, Differential regulation of MAP kinase cascade in human colorectal tumorigenesis. Br J Cancer 81:1116–1121 Rasouli N, Kern PA, 2008, Adipocytokines and the metabolic complications of obesity. J Clin Endocrinol Metab 93:S64–S73 Hsing AW, Sakoda LC, Chua S Jr, 2007, Obesity, metabolic syndrome, and prostate cancer. Am J Clin Nutr 86:s843–s857 Greenberg AS, ObinMS, 2006, Obesity and the role of adipose tissue in inflammation and metabolism. Am J Clin Nutr 83:461S–465S Itzkowitz SH, Yio X, 2004, Inflammation and Cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol 287:G7–G17 Trayhurn P, 2005, Endocrine and signalling role of adipose tissue: new perspectives on fat. Acta Physiol Scand 184:285–293 Paruchuri S, Broom O, Dib K, Sjolander A, 2005, The proinflammatory mediator leukotriene D4 induces phosphatidylinositol 3-kinase and Rac-dependent migration of intestinal epithelial cells. J Biol Chem 280:13538–13544 Pahl HL, 1999, Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene 18:6853–6866 Lodish H, Darnell J, Matsudaira P, Berk A, Scott MP, 2004, Molecular cell biology, 5th edn. W. H. Freeman Company Friedenreich CM, Orenstein MR, 2002, Physical activity and cancer prevention: etiologic evidence and biological mechanisms. J Nutr 132:3456S–3464S Furuta Y, Hall ER, Sanduja S, Barkley T Jr, Milas L, 1988, Prostaglandin production by murine tumors as a predictor for therapeutic response to indomethacin. Cancer Res 48:3002–7 Martinez ME, Heddens D, Earnest DL et al, 1999, Physical activity, body mass index, and prostaglandin E2 levels in rectal mucosa. J Natl Cancer Inst 91:950–953 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 785 EP 790 DI 10.1007/s12253-010-9352-9 PG 6 ER PT J AU Dunai, Zs Bauer, IP Mihalik, R AF Dunai, Zsuzsanna Bauer, I Pal Mihalik, Rudolf TI Necroptosis: Biochemical, Physiological and Pathological Aspects SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Necrosis; Necroptosis; Programmed cell death; RIPK1; RIPK3; TNF; Death receptor ID Necrosis; Necroptosis; Programmed cell death; RIPK1; RIPK3; TNF; Death receptor AB Programmed cell death is a key component of tissue homeostasis, normal development and wide variety of diseases. Conventional view refers to programmed cell death form as caspase-mediated apoptosis while necrosis is considered as an accidental and unwanted cell demise, carried out in a non-regulated manner and caused by extreme conditions. However, accumulating evidences indicate that necrotic cell death can also be a regulated process. The term necroptosis has been introduced to describe a cell death receptor-induced, caspase-independent, highly regulated type of programmed cell death process with morphological resemblance of necrosis. Necroptosis recently has been found to contribute to a wide range of pathologic cell death forms including ischemic brain injury, neurodegenerative diseases and viral infection, therefore a better understanding of the necroptotic signaling machinery has clinical relevance. C1 [Dunai, Zsuzsanna] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy street 7-9, H-1122 Budapest, Hungary. [Bauer, I Pal] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Tuzolto street 37-47, H-1094 Budapest, Hungary. [Mihalik, Rudolf] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Rath Gyorgy street 7-9, H-1122 Budapest, Hungary. RP Dunai, Zs (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, H-1122 Budapest, Hungary. EM dunaizs@oncol.hu CR Kerr JF, Wyllie AH, Currie AR, 1972, Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 26(4):239–257 Metzstein MM, Stanfield GM, Horvitz HR, 1998, Genetics of programmed cell death in C. elegans: past, present and future. Trends Genet 14(10):410–416 Kroemer G, El-Deiry WS, Golstein P, Peter ME, Vaux D, Vandenabeele P et al, 2005, Classification of cell death: recommendations of the nomenclature committee on cell death. Cell Death Differ 12(suppl 2):1463–1467 Deas O, Dumont C, MacFarlane M, Rouleau M, Hebib C, Harper F et al, 1998, Caspase-independent cell death induced by anti- CD2 or staurosporine in activated human peripheral T lymphocytes. J Immunol 161(7):3375–3383 Majno G, Joris I, 1995, Apoptosis, oncosis, and necrosis: an overview of cell death. Am J Pathol 146(1):3–15 Lemasters JJ, 1999, V. Necrapoptosis and the mitochondrial permeability transition: shared pathways to necrosis and apoptosis. Am J Physiol 276(1):G1–6 Li M, Beg AA, 2000, Induction of necrotic-like cell death by tumor necrosis factor alpha and caspase inhibitors: novel mechanism for killing virus-infected cells. J Virol 74(16):7470–7477 Borst P, Rottenberg S, 2004, Cancer cell death by programmed necrosis? Drug Resist Updat 7(6):321–324 Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N et al, 2005, Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol 1, 2):112–119 Vercammen D, Brouckaert G, Denecker G, Van De Craen M, Declercq W, Fiers W et al, 1998, Dual signaling of the Fas receptor: initiation of both apoptotic and necrotic cell death pathways. J Exp Med 188(5):919–930 Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S et al, 2000, Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol 1(6):489–495 Kawahara A, Ohsawa Y, Matsumura H, Uchiyama Y, Nagata S, 1998, Caspase-independent cell killing by Fas-associated protein with death domain. J Cell Biol 143(5):1353–1360 Laster SM, Wood JG, Gooding LR, 1988, Tumor necrosis factor can induce both apoptic and necrotic forms of cell lysis. J Immunol 141(8):2629–2634 Schulze-Osthoff K, Krammer PH, Droge W, 1994, Divergent signalling via APO-1/Fas and the TNF receptor, two homologous molecules involved in physiological cell death. EMBO J 13, 19):4587–4596 Hsu H, Huang J, Shu HB, Baichwal V, Goeddel DV, 1996, TNFdependent recruitment of the protein kinase RIP to the TNF receptor-1 signaling complex. Immunity 4(4):387–396 Degterev A, Hitomi J, Germscheid M, Ch’en IL, Korkina O, Teng X et al, 2008, Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol 4(5):313–321 Yuan J, 2009, Neuroprotective strategies targeting apoptotic and necrotic cell death for stroke. Apoptosis 14(4):469–477 Neumar RW, 2000, Molecular mechanisms of ischemic neuronal injury. Ann Emerg Med 36(5):483–506 McCully JD, Wakiyama H, Hsieh YJ, Jones M, Levitsky S, 2004, Differential contribution of necrosis and apoptosis in myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 286, 5 55–5):H1923–H1935 Zhang X, Chen Y, Jenkins LW, Kochanek PM, Clark RSB, 2005, Bench-to-bedside review: apoptosis/programmed cell death triggered by traumatic brain injury. Crit Care 9(1):66–75 Vercammen D, Beyaert R, Denecker G, Goossens V, Van Loo G, Declercq W et al, 1998, Inhibition of caspases increases the sensitivity of L929 cells to necrosis mediated by tumor necrosis factor. J Exp Med 187(9):1477–1485 Chan FKM, Shisler J, Bixby JG, Felices M, Zheng L, Appel M et al, 2003, A role for tumor necrosis factor receptor-2 and receptorinteracting protein in programmed necrosis and antiviral responses. J Biol Chem 278(51):51613–51621 Andera L, 2009, Signaling activated by the death receptors of the TNFR family. Biomed Pap 153(3):173–180 Micheau O, Tschopp J, 2003, Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 114(2):181– 190 Ea CK, Deng L, Xia ZP, Pineda G, Chen ZJ, 2006, Activation of IKK by TNFα requires site-specific ubiquitination of RIP1 and polyubiquitin binding by NEMO. Mol Cell 22(2):245–257 Chen ZJ, 2005, Ubiquitin signalling in the NF-kB pathway. Nat Cell Biol 7(8):758–765 Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G., 2010, Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol 11(10):700–714 Cho Y, Challa S, Moquin D, Genga R, Ray TD, Guildford M et al, 2009, Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell 137(6):1112–1123 Feng S, Yang Y, Mei Y,Ma L, Zhu DE, Hoti N et al, 2007, Cleavage of RIP3 inactivates its caspase-independent apoptosis pathway by removal of kinase domain. Cell Signal 19(10):2056–2067 Jin Z, El-Deiry WS, 2006, Distinct signaling pathways in TRAILversus tumor necrosis factor-induced apoptosis. Mol Cell Biol 26, 21):8136–8148 Harper N, Hughes M, MacFarlane M, Cohen GM, 2003, Fasassociated death domain protein and caspase-8 are not recruited to the tumor necrosis factor receptor I signaling complex during tumor necrosis factor-induced apoptosis. J Biol Chem 278, 28):25534–25541 Morgan MJ, Kim YS, Liu ZG, 2009, Membrane-bound Fas ligand requires RIP1 for efficient activation of caspase-8 within the death-inducing signaling complex. J Immunol 183(5):3278– 3284 Lavrik IN., 2011, Regulation of death receptor-induced apoptosis induced via CD95/Fas and other death receptors. Mol Biol 45, 1):150–155 Gonzalvez F, Ashkenazi A., 2010, New insights into apoptosis signaling by Apo2L/TRAIL. Oncogene 29(34):4752–4765 Sakahira H, Enari M, Nagata S, 1998, Cleavage of CAD inhibitor in CAD activation and DNA degradation during apoptosis. Nature 391(6662):96–99 Luthi AU, Martin SJ, 2007, The CASBAH: a searchable database of caspase substrates. Cell Death Diff 14(4):641–650 Bulat N, Widmann C, 2009, Caspase substrates and neurodegenerative diseases. Brain Res Bull 80(4–5):251–267 Irmler M, Thome M, Hahne M, Schneider P, Hofmann K, Steiner Vet al, 1997, Inhibition of death receptor signals by cellular FLIP. Nature 388(6638):190–195 Oosten ASV, Navis G, Stegeman CA, Joles JA, Klok PA, Kuipers F et al, 2001, Increased expression of cFLIPL in colonic adenocarcinoma. J Pathol 194(1):15–19 Tepper CG, Seldin MF, 1999, Modulation of caspase-8 and FLICE-inhibitory protein expression as a potential mechanism of Epstein-Barr virus tumorigenesis in Burkitt’s lymphoma. Blood 94, 5):1727–1737 Hitomi J, Christofferson DE, Ng A, Yao J, Degterev A, Xavier RJ et al, 2008, Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway. Cell 135(7):1311– 1323 He S, Wang L, Miao L, Wang T, Du F, Zhao L et al, 2009, Receptor interacting protein Kinase-3 determines cellular necrotic response to TNF-α. Cell 137(6):1100–1111 Zhang DW, Shao J, Lin J, Zhang N, Lu BJ, Lin SC et al, 2009, RIP3, an energy metabolism regulator that switches TNF-induced cell death from apoptosis to necrosis. Science 325(5938):332–336 Vanden Berghe T, Declercq W, Vandenabeele P, 2007, NADPH Oxidases: new players in TNF-induced necrotic cell death. Mol Cell 26(6):769–771 Kim YS, Morgan MJ, Choksi S, Liu ZG, 2007, TNF-induced activation of the Nox1 NADPH oxidase and its role in the induction of necrotic cell death. Mol Cell 26(5):675–687 Temkin V, Huang Q, Liu H, Osada H, Pope RM, 2006, Inhibition of ADP/ATP exchange in receptor-interacting protein-mediated necrosis. Mol Cell Biol 26(6):2215–2225 Berghe TV, Vanlangenakker N, Parthoens E, Deckers W, Devos M, Festjens N et al., 2010, Necroptosis, necrosis and secondary necrosis converge on similar cellular disintegration features. Cell Death Differ 17(6):922–930 Yu L, Alva A, Su H, Dutt P, Freundt E, Welsh S et al, 2004, Regulation of an ATG7-beclin 1 program of autophaglic cell death by caspase-8. Science 304(5676):1500–1502 Bonapace L, Bornhauser BC, Schmitz M, Cario G, Ziegler U, Niggli FK et al., 2010, Induction of autophagy-dependent necroptosis is required for childhood acute lymphoblastic leukemia cells to overcome glucocorticoid resistance. J Clin Invest 120, 4):1310–1323 Yoshida H, Kong YY, Yoshida R, Elia AJ, Hakem A, Hakem R et al, 1998, Apaf1 is required for mitochondrial pathways of apoptosis and brain development. Cell 94(6):739–750 Lindsten T, Ross AJ, King A, Zong WX, Rathmell JC, Shiels HA et al, 2000, The combined functions of proapoptotic Bcl-2 family members Bak and Bax are essential for normal development of multiple tissues. Mol Cell 6(6):1389–1399 Lindsten T, Thompson CB, 2006, Cell death in the absence of Bax and Bak. Cell Death Differ 13(8):1272–1276 Laporte C, Kosta A, Klein G, Aubry L, Lam D, Tresse E et al, 2007, A necrotic cell death model in a protist. Cell Death Differ 14(2):266–274 Barkla DH, Gibson PR, 1999, The fate of epithelial cells in the human large intestine. Pathology 31(3):230–238 Emons J, Chagin AS, Hultenby K, Zhivotovsky B, Wit JM, Karperien M et al, 2009, Epiphyseal fusion in the human growth plate does not involve classical apoptosis. Pediatr Res 66(6):654–659 Kumar A, Rothman J, 2007, Cell death: hook, line and linker. Curr Biol 17(8):R286–R289 Abraham MC, Lu Y, Shaham S, 2007, A morphologically conserved nonapoptotic program promotes linker cell death in Caenorhabditis elegans. Dev Cell 12(1):73–86 Yuan J, Kroemer G., 2010, Alternative cell death mechanisms in development and beyond. Genes Dev 24(23):2592–2602 McCall K., 2010, Genetic control of necrosis-another type of programmed cell death. Curr Opin Cell Biol 22(6):882–888 Zong WX, Thompson CB, 2006, Necrotic death as a cell fate. Genes Dev 20(1):1–15 Golstein P, Kroemer G, 2005, Redundant cell death mechanisms as relics and backups. Cell Death Differ 12(suppl 2):1490–1496 Benedict CA, Norris PS, Ware CF, 2002, To kill or be killed: viral evasion of apoptosis. Nat Immunol 3(11):1013–1018 Berglund AC, Sjolund E, Ostlund G, Sonnhammer ELL, 2008, InParanoid 6: eukaryotic ortholog clusters with inparalogs. Nucleic Acids Res 36(suppl 1):D263–D266 Mareninova OA, Sung KF, Hong P, Lugea A, Pandol SJ, Gukovsky I et al, 2006, Cell death in pancreatitis: caspases protect from necrotizing pancreatitis. J Biol Chem 281(6):3370– 3381 West T, Atzeva M, Holtzman DM, 2006, Caspase-3 deficiency during development increases vulnerability to hypoxic-ischemic injury through caspase-3-independent pathways. Neurobiol Dis 22(3):523–537 Stoll G, Jander S, Schroeter M, 2002, Detrimental and beneficial effects of injury-induced inflammation and cytokine expression in the nervous system. Adv Exp Med Biol 513:87–113 Zhang M, Chen L, 2008, Status of cytokines in ischemia reperfusion induced heart injury. Cardiovasc Hematol Disord Drug Targets 8(3):161–172 You Z, Savitz SI, Yang J, Degterev A, Yuan J, Cuny GD et al, 2008, Necrostatin-1 reduces histopathology and improves functional outcome after controlled cortical impact in mice. J Cereb Blood Flow Metab 28(9):1564–1573 Li Y, Yang X, Ma C, Qiao J, Zhang C, 2008, Necroptosis contributes to the NMDA-induced excitotoxicity in rat’s cultured cortical neurons. Neurosci Lett 447(2–3):120–123 Tan S, Schubert D, Maher P, 2001, Oxytosis: a novel form of programmed cell death. Curr Top Med Chem 1(6):497–506 Xu X, Chua CC, Kong J, Kostrzewa RM, Kumaraguru U, Hamdy RC et al, 2007, Necrostatin-1 protects against glutamate-induced glutathione depletion and caspase-independent cell death in HT-22 cells. J Neurochem 103(5):2004–2014 Cregan SP, Dawson VL, Slack RS, 2004, Role of AIF in caspasedependent and caspase-independent cell death. Oncogene 23(16 REV. ISS. 2):2785–2796 Kim S, Dayani L, Rosenberg PA, Li J., 2010, RIP1 kinase mediates arachidonic acid-induced oxidative death of oligodendrocyte precursors. Int J Physiol Pathophysiol Pharmacol 2(2):137–147 Davis CW, Hawkins BJ, Ramasamy S, Irrinki KM, Cameron BA, Islam K et al., 2010, Nitration of the mitochondrial complex I subunit NDUFB8 elicits RIP1- and RIP3-mediated necrosis. Free Radic Biol Med 48(2):306–317 Smith CCT, Davidson SM, Lim SY, Simpkin JC, Hothersall JS, Yellon DM, 2007, Necrostatin: a potentially novel cardioprotective agent? Cardiovasc Drugs Ther 21(4):227–233 Lim SY, Davidson SM, Mocanu MM, Yellon DM, Smith CCT, 2007, The cardioprotective effect of necrostatin requires the cyclophilin-D component of the mitochondrial permeability transition pore. Cardiovasc Drugs Ther 21(6):467–469 Bao L, Li Y, Deng SX, Landry D, Tabas I, 2006, Sitosterolcontaining lipoproteins trigger free sterol-induced caspaseindependent death in ACAT-competent macrophages. J Biol Chem 281(44):33635–33649 Han W, Li L, Qiu S, Lu Q, Pan Q, Gu Y et al, 2007, Shikonin circumvents cancer drug resistance by induction of a necroptotic death. Mol Cancer Ther 6(5):1641–1649 Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100(1):57–70 Hu W, Kavanagh JJ, 2003, Anticancer therapy targeting the apoptotic pathway. Lancet Oncol 4(12):721–729 Li QX, Yu DH, Liu G, Ke N, McKelvy J, Wong-Staal F, 2008, Selective anticancer strategies via intervention of the death pathways relevant to cell transformation. Cell Death Differ 15, 8):1197–1210 Horita H, Frankel AE, Thorburn A, 2008, Acute myeloid leukemia-targeted toxin activates both apoptotic and necroptotic death mechanisms. PLoS ONE 3(12):e3909 Tinel A, Tschopp J, 2004, The PIDDosome, a protein complex implicated in activation of caspase-2 in response to genotoxic stress. Science 304(5672):843–846 Janssens S, Tinel A, Lippens S, Tschopp J, 2005, PIDD mediates NF-kB activation in response to DNA damage. Cell 123(6):1079– 1092 Kim IR, Murakami K, Chen NJ, Saibil SD, Matysiak-Zablocki E, Elford AR et al, 2009, DNA damage- and stress-induced apoptosis occurs independently of PIDD. Apoptosis 14(9):1039– 1049 Tu HC, Ren D, Wang GX, Chen DY, Westergard TD, Kim H et al, 2009, The p53-cathepsin axis cooperates with ROS to activate programmed necrotic death upon DNA damage. Proc Natl Acad Sci USA 106(4):1093–1098 Zong WX, Ditsworth D, Bauer DE, Wang ZQ, Thompson CB, 2004, Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes Dev 18(11):1272–1282 Hanahan D, Weinberg RA., 2011, Hallmarks of cancer: the next generation. Cell 144(5):646–674 Grivennikov SI, Greten FR, Karin M., 2010, Immunity, inflammation, and cancer. Cell 140(6):883–899 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 791 EP 800 DI 10.1007/s12253-011-9433-4 PG 10 ER PT J AU Vicha, A Holzerova, M Krepelova, A Musil, Z Prochazka, P Sumerauer, D Kodet, R Eckschlager, T Jarosova, M AF Vicha, Ales Holzerova, Milena Krepelova, Anna Musil, Zdenek Prochazka, Pavel Sumerauer, David Kodet, Roman Eckschlager, Tomas Jarosova, Marie TI Molecular Cytogenetic Characterization in Four Pediatric Pheochromocytomas and Paragangliomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pheochromocytoma; Paraganglioma; Comparative genomic hybridization; Pediatric; Microarray ID Pheochromocytoma; Paraganglioma; Comparative genomic hybridization; Pediatric; Microarray AB Pheochromocytomas (PCCs) are rare tumors among children and adolescents and therefore are not genetically well characterized. The most frequently observed chromosomal changes in PCC are losses of 1p, 3q and/or 3p, 6q, 17p, 11q, 22q, and gains of 9q and 17q. Aberrations involving chromosome 11 are more common in malignant tumors. Unfortunately information about gene aberrations in childhood PCC’s is limited. We used comparative genomic hybridization (CGH) and array comparative genomic hybridization (aCGH) to screen for copy number changes in four children suffering from pheochromocytoma or paraganglioma. Patients were diagnosed at the age 13 or 14 years. Bilateral pheochromocytoma was associated with von Hippel-Lindau syndrome (VHL). Multiple paraganglioma was associated with a germline mutation in SDHB. We found very good concordance between the results of CGH and aCGH techniques. Losses were observed more frequently than gains. All cases had a loss of chromosome 11 or 11p. Other aberrations were loss of chromosome 3 and 11 in sporadic pheochromocytoma, and loss of 3p and 11p in pheochromocytoma, which carried the VHL mutation. The deletion of chromosome 1p and other changes were observed in paragangliomas. We conclude that both array CGH and CGH analysis identified similar chromosomal regions involved in tumorigenesis of pheochromocytoma and paragangliomas, but we found 3 discrepancies between the methods. We didn’t find any, of the proposed, molecular markers of malignancy in our benign cases and therefore we speculate that molecular cytogenetic examination may be helpful in separating benign and malignant forms in the future. C1 [Vicha, Ales] Charles University, 2nd Medical School and University Hospital Motol, Department of Pediatric Hematology and Oncology, Fakultni nemocnice v Motole, V uvalu 84Prague, Czech Republic. [Holzerova, Milena] Palacky University and University Hospital, Medical School, Department of Hemato-oncologyOlomouc, Czech Republic. [Krepelova, Anna] Charles University and University Hospital Motol, 2nd Medical School, Institute of Biology and Medical GeneticsPrague, Czech Republic. [Musil, Zdenek] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Institute of Biology and Medical GeneticsPrague, Czech Republic. [Prochazka, Pavel] Charles University, 2nd Medical School and University Hospital Motol, Department of Pediatric Hematology and Oncology, Fakultni nemocnice v Motole, V uvalu 84Prague, Czech Republic. [Sumerauer, David] Charles University, 2nd Medical School and University Hospital Motol, Department of Pediatric Hematology and Oncology, Fakultni nemocnice v Motole, V uvalu 84Prague, Czech Republic. [Kodet, Roman] Charles University and University Hospital Motol, 2nd Medical School, Department of Pathology and molecular medicinePrague, Czech Republic. [Eckschlager, Tomas] Charles University, 2nd Medical School and University Hospital Motol, Department of Pediatric Hematology and Oncology, Fakultni nemocnice v Motole, V uvalu 84Prague, Czech Republic. [Jarosova, Marie] Palacky University and University Hospital, Medical School, Department of Hemato-oncologyOlomouc, Czech Republic. RP Vicha, A (reprint author), Charles University, 2nd Medical School and University Hospital Motol, Department of Pediatric Hematology and Oncology, Prague, Czech Republic. EM ales.vicha@lfmotol.cuni.cz CR Havekes B, Romijn JA, Eisenhofer G et al, 2009, Update on pediatric pheochromocytoma. Pediatr Nephrol 24:943–950 Lenders JW, Eisenhofer G, Mannelli M et al, 2005, Phaeochromocytoma. Lancet 366:665–675 Lemeta S, Salmenkivi K, Pylkkanen L et al, 2006, Frequent loss of heterozygosity at 6q in pheochromocytoma. Hum Pathol 37:749–754 Ciftci AO, Tanyel FC, Senocak ME et al, 2001, Pheochromocytoma in children. J Pediatr Surg 36:447–452 Ross JH, 2000, Pheochromocytoma. Special considerations in children. Urol Clin North Am 27:393–402 Caty MG, Coran AG, Geagen M et al, 1990, Current diagnosis and treatment of pheochromocytoma in children. Experience with 22 consecutive tumors in 14 patients. Arch Surg 125:978–981 Eisenhofer G, Bornstein SR, Brouwers FM et al, 2004, Malignant pheochromocytoma: current status and initiatives for future progress. Endocr Relat Cancer 11:423–436 Huang H, Abraham J, Hung E et al, 2008, Treatment of malignant pheochromocytoma/paraganglioma with cyclophosphamide, vincristine, and dacarbazine: recommendation from a 22-year follow-up of 18 patients. Cancer 113:2020–2028 De Krijger RR, Petri BJ, Van Nederveen FH et al, 2006, Frequent genetic changes in childhood pheochromocytomas. Ann NY Acad Sci 1073:166–176 Astuti D, Latif F, Dallol A et al, 2001, Gene mutations in the succinate dehydrogenase subunit SDHB cause susceptibility to familial pheochromocytoma and to familial paraganglioma. Am J Hum Genet 69:49–54 Mannelli M, Simi L, Gagliano MS et al, 2007, Genetics and biology of pheochromocytoma. Exp Clin Endocrinol Diabetes 115:160–165 Neumann HP, Berger DP, Sigmund G et al, 1993, Pheochromocytomas, multiple endocrine neoplasia type 2, and von Hippel- Lindau disease. N Engl J Med 329:1531–1538 Eng C, Crossey PA, Mulligan LM et al, 1995, Mutations in the RET proto-oncogene and the von Hippel-Lindau disease tumour suppressor gene in sporadic and syndromic phaeochromocytomas. J Med Genet 32:934–937 Erlic Z, Neumann HP, 2009, Familial pheochromocytoma. Hormones, Athens, 8:29–38 McNicol AM, Young WF Jr., Kawashma A, Komminoth P, Tischler AS, 2004, Benign phaeochromocytoma. In: DeLellis RA, Lloyd RV, Heitz PU, Eng C, eds). WHO classification of tumours: pathology and genetics of tumours of endocrine organs. Lyon7 International Agency for Research on Cancer, IARC, 151–5 Medeiros LJ, Wolf BC, Balogh K et al, 1985, Adrenal pheochromocytoma: a clinicopathologic review of 60 cases. Hum Pathol 16:580–589 Werbel SS, Ober KP, 1995, Pheochromocytoma. Update on diagnosis, localization, and management. Med Clin North Am 79:131–153 Adjalle R, Plouin PF, Pacak K et al, 2009, Treatment of malignant pheochromocytoma. Horm Metab Res 41:687–696 Dannenberg H, Speel EJ, Zhao J et al, 2000, Losses of chromosomes 1p and 3q are early genetic events in the development of sporadic pheochromocytomas. Am J Pathol 157:353–359 Edstrom E, Mahlamaki E, Nord B et al, 2000, Comparative genomic hybridization reveals frequent losses of chromosomes 1p and 3q in pheochromocytomas and abdominal paragangliomas, suggesting a common genetic etiology. Am J Pathol 156:651– 659 Hering A, Guratowska M, Bucsky P et al, 2006, Characteristic genomic imbalances in pediatric pheochromocytoma. Genes Chromosom Cancer 45:602–607 Lui WO, Chen J, Glasker S et al, 2002, Selective loss of chromosome 11 in pheochromocytomas associated with the VHL syndrome. Oncogene 21:1117–1122 Musil Z, Puchmajerova A, Krepelova A, et al. Paraganglioma in a 13-year-old girl: a novel SDHB gene mutation in the family? Cancer Genet Cytogenet 197:189–92 Bedrnicek J, Vicha A, Jarosova M et al, 2005, Characterization of drug-resistant neuroblastoma cell lines by comparative genomic hybridization. Neoplasma 52:415–419 Hofstra RM, Wu Y, Stulp RP et al, 2000, RET and GDNF gene scanning in Hirschsprung patients using two dual denaturing gel systems. Hum Mutat 15:418–429 Petri BJ, Speel EJ, Korpershoek E et al, 2008, Frequent loss of 17p, but no p53 mutations or protein overexpression in benign and malignant pheochromocytomas. Mod Pathol 21:407–413 van Nederveen FH, Korpershoek E, deLeeuw RJ et al, 2009, Array-comparative genomic hybridization in sporadic benign pheochromocytomas. Endocr Relat Cancer 16:505–513 Khosla S, Patel VM, Hay ID et al, 1991, Loss of heterozygosity suggests multiple genetic alterations in pheochromocytomas and medullary thyroid carcinomas. J Clin Invest 87:1691–1699 Hensen EF, Jordanova ES, van Minderhout IJ et al, 2004, Somatic loss of maternal chromosome 11 causes parent-of-origin-dependent inheritance in SDHD-linked paraganglioma and phaeochromocytoma families. Oncogene 23:4076–4083 Burkhardt B,Moericke A, KlapperWet al, 2008, Pediatric precursor T lymphoblastic leukemia and lymphoblastic lymphoma: differences in the common regions with loss of heterozygosity at chromosome 6q and their prognostic impact. Leuk Lymphoma 49:451–461 Nakano M, Takahashi H, Shiraishi T et al, 2008, Prediction of clinically insignificant prostate cancer by detection of allelic imbalance at 6q, 8p and 13q. Pathol Int 58:415–420 Speel EJ, Richter J, Moch H et al, 1999, Genetic differences in endocrine pancreatic tumor subtypes detected by comparative genomic hybridization. Am J Pathol 155:1787–1794 August C, August K, Schroeder S et al, 2004, CGH and CD 44/ MIB-1 immunohistochemistry are helpful to distinguish metastasized from nonmetastasized sporadic pheochromocytomas. Mod Pathol 17:1119–1128 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 801 EP 808 DI 10.1007/s12253-011-9385-8 PG 8 ER PT J AU Rykala, J Przybylowska, K Majsterek, I Pasz-Walczak, G Sygut, A Dziki, A Kruk-Jeromin, J AF Rykala, Jan Przybylowska, Karolina Majsterek, Ireneusz Pasz-Walczak, Grazyna Sygut, Andrzej Dziki, Adam Kruk-Jeromin, Julia TI Angiogenesis Markers Quantification in Breast Cancer and Their Correlation with Clinicopathological Prognostic Variables SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Angiogenesis; Cancer progression; Multiplex protein assay ID Breast cancer; Angiogenesis; Cancer progression; Multiplex protein assay AB Tumoural angiogenesis is essential for the growth and spread of breast cancer cells. Therefore the aim of this study was to assess the diagnostic performance of angiogenesis markers in tumours and there reflecting levels in serum of breast cancer patients. Angiogenin, Ang2, fibroblast growth factor basic, intercellular adhesion molecule (ICAM)-1, keratinocyte growth factor (KGF), platelet-derived growth factor-BB, and VEGF-A were measured using a FASTQuant angiogenic growth factor multiplex protein assay. We observed that breast cancer tumours exhibited high levels of PDGF-BB, bFGF and VEGF, and extremely high levels of TIMP-1 and Ang-2, whereas in serum we found significantly higher levels of Ang-2, PDGF-BB, bFGF, ICAM-1 and VEGF in patients with breast cancer compared to the benign breast diseases patients. Moreover, some of these angiogenesis markers evaluated in tumour and serum of breast cancer patients exhibited association with standard clinical parameters, ER status as well as MVD of tumours. Angiogenesis markers play important roles in tumour growth, invasion and metastasis. Our results suggest that analysis of angiogenesis markers in tumour and serum of breast cancer patients using multiplex protein assay can improve diagnosis and prognosis in this diseases. C1 [Rykala, Jan] Medical University of Lodz, Barlicki Hospital, Department of Plastic, Reconstructive and Aesthetic Surgery, Kopcinskiego 22, 90–153 Lodz, Poland. [Przybylowska, Karolina] Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, Hallera 1, 90–647 Lodz, Poland. [Majsterek, Ireneusz] Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, Hallera 1, 90–647 Lodz, Poland. [Pasz-Walczak, Grazyna] Medical University of Lodz, Department of Pathology, Paderewskiego 4, 93–509 Lodz, Poland. [Sygut, Andrzej] Medical University of Lodz, Department of General and Colorectal Surgery, Hallera 1, 90–647 Lodz, Poland. [Dziki, Adam] Medical University of Lodz, Department of General and Colorectal Surgery, Hallera 1, 90–647 Lodz, Poland. [Kruk-Jeromin, Julia] Medical University of Lodz, Barlicki Hospital, Department of Plastic, Reconstructive and Aesthetic Surgery, Kopcinskiego 22, 90–153 Lodz, Poland. RP Rykala, J (reprint author), Medical University of Lodz, Barlicki Hospital, Department of Plastic, Reconstructive and Aesthetic Surgery, 90–153 Lodz, Poland. EM janekrykalarykala@gmail.com CR Baeriswyl V, Christofori G, 2009, The angiogenic switch in carcinogenesis. Semin Cancer Biol 19(5):329–337 Fujita Y, Abe R, Shimizu H, 2008, Clinical approaches toward tumor angiogenesis: past, present and future. Curr Pharm Des 14, 36):3820–3834 Poon RT, Fan ST, Wong J, 2001, Clinical implications of circulating angiogenic factors in cancer patients. J Clin Oncol 19, 4):1207–1225 Uzzan B, Nicolas P, Cucherat M, Perret GY, 2004, Microvessel density as a prognostic factor in women with breast cancer: a systematic review of the literature and meta-analysis. Cancer Res 64(9):2941–2945 Ozalp S, Yalcin OT, Acikalin M, Tanir HM, Oner U, Akkoyunlu A, 2003, Microvessel density, MVD, as a prognosticator in endometrial carcinoma. Eur J Gynaecol Oncol 24(3–4):305–308 Bono AV, Celato N, Cova V, Salvadore M, Chinetti S, Novario R, 2002, Microvessel density in prostate carcinoma. Prostate Cancer Prostatic Dis 5(2):123–127 Liang JT, Huang KC, Jeng YM, Lee PH, Lai HS, Hsu HC, 2004, Microvessel density, cyclo-oxygenase 2 expression, K-ras mutation and p53 overexpression in colonic cancer. Br J Surg 91(3):355–361 Szarvas T, Jager T, Droste F, Becker M, Kovalszky I, Romics I, Ergun S, Rubben H, 2009, Serum levels of angiogenic factors and their prognostic relevance in bladder cancer. Pathol Oncol Res 15, 2):193–201 Tas F, Duranyildiz D, Oguz H, Camlica H, Yasasever V, Topuz E, 2006, Circulating serum levels of angiogenic factors and vascular endothelial growth factor receptors 1 and 2 in melanoma patients. Melanoma Res 16(5):405–411 Balacescu O, Neagoe I, Balacescu L, Crisan N, Feciche B, Tudoran O, Coman I, Irimie A, 2008, Angiogenesis serum protein quantification for prostate. Pathology Curr Urol 2:181–187 Balsari A, Maier JA, Colnaghi MI, Menard S, 1999, Correlation between tumor vascularity, vascular endothelial growth factor production by tumor cells, serum vascular endothelial growth factor levels, and serum angiogenic activity in patients with breast carcinoma. Lab Invest 79(7):897–902 Zhao J, Yan F, Ju H, Tang J, Qin J, 2004, Correlation between serum vascular endothelial growth factor and endostatin levels in patients with breast cancer. Cancer Lett 204(1):87–95 Duranyildiz D, Camlica H, Soydinc HO, Derin D, Yasasever V, 2009, Serum levels of angiogenic factors in early breast cancer remain close to normal. Breast 18(1):26–29 Weidner N, Semple JP, Welch ER, Folkman J, 1991, Tumor angiogenesis et metastasis—correlation in invasive breast carcinoma. N Engl J Med 324:1–8 Vermeulen PB, Gasparini G, Fox SB, 1996, Quantification of angiogenesis in solid human tumors: an International Consensus on the methodology and criteria of evaluation. Eur J Cancer 32:2474–2484 Kranz A, Mattfeldt T, Waltenberger J, 1999, Molecular mediators of tumor angiogenesis: enhanced expression and activation of vascular endothelial growth factor receptor KDR in primary breast cancer. Int J Cancer 84(3):293–298 Ghosh S, Sullivan CA, Zerkowski MP, Molinaro AM, Rimm DL, Camp RL, Chung GG, 2008, High levels of vascular endothelial growth factor and its receptors, VEGFR-1, VEGFR-2, neuropilin-1, are associated with worse outcome in breast cancer. Hum Pathol 39(12):1835–1843 Kawai H, Li H, Chun P, Avraham S, Avraham HK, 2002, Direct interaction between BRCA1 and the estrogen receptor regulates vascular endothelial growth factor, VEGF, transcription and secretion in breast cancer cells. Oncogene 21(50):7730–7739 Ali SH, O’Donnell AL, Balu D, Pohl MB, Seyler MJ, Mohamed S, Mousa S, Dandona P, 2000, Estrogen receptor-alpha in the inhibition of cancer growth and angiogenesis. Cancer Res 60, 24):7094–7098 Bos R, van Diest PJ, de Jong JS, van der Groep P, van der Valk P, van derWall E, 2005, Hypoxia-inducible factor-1alpha is associated with angiogenesis, and expression of bFGF, PDGF-BB, and EGFR in invasive breast cancer. Histopathology 46(1):31–36 Guo P, Hu B, Gu W, Xu L, Wang D, Huang HJ, Cavenee WK, Cheng SY, 2003, Platelet-derived growth factor-B enhances glioma angiogenesis by stimulating vascular endothelial growth factor expression in tumor endothelia and by promoting pericyte recruitment. Am J Pathol 162(4):1083–1093 Zhu L, Loo WT, Cheng CW, Chow LW, 2006, Possible predictive markers related to micro-metastasis in breast cancer patients. Oncol Rep 15(5):1217–1223 Schweigerer L, Neufeld G, Friedman J, Abraham JA, Fiddes JC, Gospodarowicz D, 1987, Capillary endothelial cells express basic fibroblast growth factor, a mitogen that promotes their own growth. Nature 325(6101):257–259 Gravdal K, Halvorsen OJ, Haukaas SA, Akslen LA, 2006, Expression of bFGF/FGFR-1 and vascular proliferation related to clinicopathologic features and tumor progress in localized prostate cancer. Virchows Arch 448(1):68–74 Shi YH, Bingle L, Gong LH, Wang YX, Corke KP, Fang WG, 2007, Basic FGF augments hypoxia induced HIF-1-alpha expression and VEGF release in T47D breast cancer cells. Pathology 39(4):396–400 Sartippour MR, Zhang L, Lu M, Wang HJ, Brooks MN, 2005, Nipple fluid basic fibroblast growth factor in patients with breast cancer. Cancer Epidemiol Biomarkers Prev 14(12):2995–2998 Wurtz SO, Schrohl AS, Mouridsen H, Brunner N, 2008, TIMP-1 as a tumor marker in breast cancer–an update. Acta Oncol 47, 4):580–590 Hayakawa T, Yamashita K, Tanzawa K, Uchijima E, Iwata K, 1992, Growth-promoting activity of tissue inhibitor ofmetalloproteinases-1, TIMP-1, for a wide range of cells. A possible new growth factor in serum. FEBS Lett 298(1):29–32 Liu XW, Bernardo MM, Fridman R, Kim HR, 2003, Tissue inhibitor of metalloproteinase-1 protects human breast epithelial cells against intrinsic apoptotic cell death via the focal adhesion kinase/phosphatidylinositol 3-kinase and MAPK signaling pathway. J Biol Chem 278(41):40364–40372 Liu H, Chen B, Lilly B, 2008, Fibroblasts potentiate blood vessel formation partially through secreted factor TIMP-1. Angiogenesis 11(3):223–234 O’Hanlon DM, Fitzsimons H, Lynch J, Tormey S, Malone C, Given HF, 2002, Soluble adhesion molecules, E-selectin, ICAM-1 and VCAM-1, in breast carcinoma. Eur J Cancer 38:2252–2257 Kostler WJ, Tomek S, Brodowicz T et al, 2001, Soluble ICAM-1 in breast cancer: clinical significance and biological implications. Cancer Immunol Immunother 50:483–490 Kawai Y, Kaidoh M, Yokoyama Y, Sano K, Ohhashi T, 2009, Chemokine CCL2 facilitates ICAM-1-mediated interactions of cancer cells and lymphatic endothelial cells in sentinel lymph nodes. Cancer Sci 100(3):419–428 Tsutsui S, Inoue H, Yasuda K, Suzuki K, Takeuchi H, Nishizaki T, Higashi H, Era S, Mori M, 2006, Angiopoietin 2 expression in invasive ductal carcinoma of the breast: its relationship to the VEGF expression and microvessel density. Breast Cancer Res Treat 98(3):261–266 Helfrich I, Edler L, Sucker A, Thomas M, Christian S, Schadendorf D, Augustin HG, 2009, Angiopoietin-2 levels are associated with disease progression in metastatic malignant melanoma. Clin Cancer Res 15(4):1384–1392 Zhou YZ, Fang XQ, Li H, Diao YT, Yang YF, Zhao DL, Wu K, Li HQ, 2007, Role of serum angiopoietin-2 level in screening for esophageal squamous cell cancer and its precursors. Chin Med J, Engl, 120(14):1216–1219 A Randomized Phase III Trial of Paclitaxel With or Without Bevacizumab as First-Line Therapy for Locally Recurrent or Metastatic Breast Cancer: Eastern Cooperative Oncology Group trial E 2100. Abstract 7 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 809 EP 817 DI 10.1007/s12253-011-9387-6 PG 9 ER PT J AU Naghibalhossaini, F Hosseini, MH Mokarram, P Zamani, M AF Naghibalhossaini, Fakhraddin Hosseini, Mahmoodzadeh Hamideh Mokarram, Pooneh Zamani, Mozhdeh TI High Frequency of Genes’ Promoter Methylation, but Lack of BRAF V600E Mutation among Iranian Colorectal Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon cancer; BRAF; KRAS; Methylation ID Colon cancer; BRAF; KRAS; Methylation AB Gene silencing due to DNA hypermethylation is a major mechanism for loss of tumor suppressor genes function in colorectal cancer. Activating V600E mutation in BRAF gene has been linked with widespread methylation of CpG islands in sporadic colorectal cancers. The aim of the present study was to evaluate the methylation status of three cancer-related genes, APC2, p14ARF, and ECAD in colorectal carcinogenesis and their association with the mutational status of BRAF and KRAS among Iranian colorectal cancer patients. DNA from 110 unselected series of sporadic colorectal cancer patients was examined for BRAF V600E mutation by PCR-RFLP. Promoter methylation of genes in tumors was determined by methylation specific PCR. The frequency of APC2, E-CAD, and p14 methylation was 92.6%, 40.4% and 16.7%, respectively. But, no V600E mutation was identified in the BRAF gene in any sample. No association was found in cases showing epigenetic APC, ECAD, and p14 abnormality with the clinicopathological parameters under study. The association between KRAS mutations and the so called methylator phenotype was previously reported. Therefore, we also analyzed the association between the hot spot KRAS gene mutations in codons of 12 and 13 with genes’ promoter hypermethylation in a subset of this group of patients. Out of 86 tumors, KRAS was mutated in 24 (28%) of tumors, the majority occurring in codon 12. KRAS mutations were not associated with genes’ methylation in this tumor series. These findings suggest a distinct molecular pathway for methylation of APC2, p14, and ECAD genes from those previously described for colorectal cancers with BRAF or KRAS mutations. C1 [Naghibalhossaini, Fakhraddin] Shiraz University of Medical Sciences, Department of Biochemistry, Zand StreetShiraz, Fars, Iran. [Hosseini, Mahmoodzadeh Hamideh] Shiraz University of Medical Sciences, Department of Biochemistry, Zand StreetShiraz, Fars, Iran. [Mokarram, Pooneh] Shiraz University of Medical Sciences, Department of Biochemistry, Zand StreetShiraz, Fars, Iran. [Zamani, Mozhdeh] Shiraz University of Medical Sciences, Department of Biochemistry, Zand StreetShiraz, Fars, Iran. RP Naghibalhossaini, F (reprint author), Shiraz University of Medical Sciences, Department of Biochemistry, Shiraz, Iran. EM fakhraddin.naghibalhossaini@mail.mcgill.ca CR Malekzadeh R, Bishehsari F, Mahdavinia M, Ansari R, 2009, Epidemiology and molecular genetics of colorectal cancer in Iran: a review. Arch Iran Med 12:161–169 Fazeli MS, Adel MG, Lebaschi AH, 2007, Colorectal carcinoma: a retrospective, descriptive study of age, gender, subsite, stage, and differentiation in Iran from 1995 to 2001 as observed in Tehran University. Dis Colon Rectum 50:990–995 Jass JR, 2007, Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology 50:113e30 Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP, 1999, CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci USA 96:8681–8686 Marais R, Marshall CJ, 1996, Control of the ERK MAP kinase cascade by Ras and Raf. Cancer Surv 27:101–125 Li WQ, Kawakami K, Ruszkiewicz A, Bennett G, Moore J, Iacopetta B, 2006, BRAF mutations are associated with distinctive clinical, pathological and molecular features of colorectal cancer independently of microsatellite instability status. Mol Cancer 5:2 Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S et al, 2002, Mutations of the BRAF gene in human cancer. Nature 417:949–954 Rajagopalan H, Bardelli A, Lengauer C, Kinzler KW, Vogelstein B, Velculescu VE, 2002, Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature 418:934 Suehiro Y, Wong CW, Chirieac LR, Kondo Y, Shen L, Webb CR et al, 2008, Epigenetic-genetic interactions in the APC/WNT, RAS/RAF, and P53 pathways in colorectal carcinoma. Clin Cancer Res 14:2560–2569 Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA et al, 2006, CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 38:787–793 English DR, Young JP, Simpson JA, Jenkins MA, Southey MC, Walsh MD et al, 2008, Ethnicity and risk for colorectal cancers showing somatic BRAF V600E mutation or CpG island methylator phenotype. Cancer Epidemiol Biomark Prev 17:1774–1780 Nagasaka T, Sasamoto H, Notohara K, Cullings HM, Takeda M, Kimura K et al, 2004, Colorectal cancer with mutation in BRAF, KRAS, and wild-type with respect to both oncogenes showing different patterns of DNA methylation. J Clin Oncol 22:4584–4594 Kominami K, Nagasaka T, Cullings HM, Hoshizima N, Sasamoto H, Young J et al, 2009, Methylation in p14(ARF, is frequently observed in colorectal cancer with low-level microsatellite instability. J Int Med Res 37:1038–1045 Auerkari EI, 2006, Methylation of tumor suppressor genes p16, INK4a), p27(Kip1, and E-cadherin in carcinogenesis. Oral Oncol 42:5–13 Schuebel KE, Chen W, Cope L, Glockner SC, Suzuki H, Yi JM et al, 2007, Comparing the DNA hypermethylome with gene mutations in human colorectal cancer. PLoS Genet 3:1709–1723 Kumar K, Brim H, Giardiello F, Smoot DT, Nouraie M, Lee EL et al, 2009, Distinct BRAF, V600E, and KRAS mutations in high microsatellite instability sporadic colorectal cancer in African Americans. Clin Cancer Res 15:1155–1161 Garinis GA, Menounos PG, Spanakis NE, Papadopoulos K, Karavitis G, Parassi I et al, 2002, Hypermethylation-associated transcriptional silencing of E-cadherin in primary sporadic colorectal carcinomas. J Pathol 198:442–449 Takeichi M, 1995, Morphogenetic roles of classic cadherins. Curr Opin Cell Biol 7:619–627 Zheng S, Chen P, McMillan A, Lafuente A, Lafuente MJ, Ballesta A et al, 2000, Correlations of partial and extensive methylation at the p14(ARF, locus with reduced mRNA expression in colorectal cancer cell lines and clinicopathological features in primary tumors. Carcinogenesis 21:2057–2064 Mokarram P, Naghibalhossaini F, Saberi Firoozi M, Hosseini SV, Izadpanah A, Salahi H et al, 2008, Methylenetetrahydrofolate reductase C677T genotype affects promoter methylation of tumorspecific genes in sporadic colorectal cancer through an interaction with folate/vitamin B12 status.World J Gastroenterol 14:3662–3671 Servomaa K, Kiuru A, Kosma VM, Hirvikoski P, Rytomaa T, 2000, p53 and K-ras gene mutations in carcinoma of the rectum among Finnish women. Mol Pathol 53:24–30 Gazin C, Wajapeyee N, Gobeil S, Virbasius CM, Green MR, 2007, An elaborate pathway required for Ras-mediated epigenetic silencing. Nature 449:1073–1077 Mokarram P, Kumar K, Brim H, Naghibalhossaini F, Saberifiroozi M, Nouraie M et al, 2009, Distinct high-profile methylated genes in colorectal cancer. PLoS ONE 4:e7012 Miranda E, Destro A, Malesci A, Balladore E, Bianchi P, Baryshnikova E et al, 2006, Genetic and epigenetic changes in primary metastatic and nonmetastatic colorectal cancer. Br J Cancer 95:1101–1107 Wheeler JM, Kim HC, Efstathiou JA, Ilyas M, Mortensen NJ, Bodmer WF, 2001, Hypermethylation of the promoter region of the E-cadherin gene, CDH1, in sporadic and ulcerative colitis associated colorectal cancer. Gut 48:367–371 Strathdee G, 2002, Epigenetic versus genetic alterations in the inactivation of E-cadherin. Semin Cancer Biol 12:373–379 van Engeland M, Weijenberg MP, Roemen GM, Brink M, de Bruine AP, Goldbohm RA et al, 2003, Effects of dietary folate and alcohol intake on promoter methylation in sporadic colorectal cancer: the Netherlands cohort study on diet and cancer. Cancer Res 63:3133–3137 Naghibalhossaini F, Mokarram P, Khalili I, Vasei M, Hosseini SV, Ashktorab H et al, 2010, MTHFR C677T and A1298C variant genotypes and the risk of microsatellite instability among Iranian colorectal cancer patients. Cancer Genet Cytogenet 197:142–151 Yuen ST, Davies H, Chan TL, Ho JW, Bignell GR, Cox C et al, 2002, Similarity of the phenotypic patterns associated with BRAF and KRAS mutations in colorectal neoplasia. Cancer Res 62:6451–6455 Ogino S, Kawasaki T, Kirkner GJ, Kraft P, Loda M, Fuchs CS, 2007, Evaluation of markers for CpG island methylator phenotype, CIMP, in colorectal cancer by a large population-based sample. J Mol Diagn 9:305–314 Brim H, Mokarram P, Naghibalhossaini F, Saberi-Firoozi M, Al- Mandhari M, Al-Mawaly K et al, 2008, Impact of BRAF, MLH1 on the incidence of microsatellite instability high colorectal cancer in populations based study. Mol Cancer 7:68 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 819 EP 825 DI 10.1007/s12253-011-9388-5 PG 7 ER PT J AU Patara, M Santos, MME Coudry, dAR Soares, AF Ferreira, OF Rossi, MB AF Patara, Marcelo Santos, Maria Monteiro Erika Coudry, de Almeida Renata Soares, Augusto Fernando Ferreira, Oliveira Fabio Rossi, Mauro Benedito TI Ezrin Expression as a Prognostic Marker in Colorectal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Ezrin; Biological marker; Prognostic; Adenocarcinoma ID Colorectal cancer; Ezrin; Biological marker; Prognostic; Adenocarcinoma AB Ezrin protein acts in the regulation of cytoskeletal and directly influences survival and tumor progression; there is an increase in its expression in metastatic cells and tissues in several types of cancer including colorectal cancer. 250 Patients with colorectal cancer submitted to surgery from 1995 to 2002. Protein expression was carried through by Tissue Micro Array immunohistochemical tests of paraffined neoplasic tissues and associated with clinical variables. Differentiation degree, lymph node invasion, metastasis at diagnosis, and palliative surgery were associated to a higher expression of the protein and survival. Higher expression of the Ezrin correlates with tumor aggressiveness and worse prognosis for colorectal cancer. C1 [Patara, Marcelo] College of Medical Sciences Santa Casa de Sao Paulo, CNPq—PIBIC, Rua Professor Antonio Prudente, 211, 01509–010 Sao Paulo, SP, Brazil. [Santos, Maria Monteiro Erika] Hospital A.C.Camargo, Center of ResearchSao Paulo, Brazil. [Coudry, de Almeida Renata] A. C. Camargo Cancer Hospital, Department of PathologySao Paulo, Brazil. [Soares, Augusto Fernando] A. C. Camargo Cancer Hospital, Department of PathologySao Paulo, Brazil. [Ferreira, Oliveira Fabio] A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery DepartmentSao Paulo, Brazil. [Rossi, Mauro Benedito] A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery DepartmentSao Paulo, Brazil. RP Patara, M (reprint author), College of Medical Sciences Santa Casa de Sao Paulo, CNPq—PIBIC, 01509–010 Sao Paulo, Brazil. EM marcelopatara@gmail.com CR Junqueira LC, Carneiro J, 2005, Molecular Basis of Cytoskeleton and Cell Movement. In: Junqueira LC, Carneiro J, eds, Celular and molecular biology, 8th edn. Guanabara Koogan S.A, Rio de Janeiro Kierzembaum AL, 2006, Epithelium. In: Kierzembaum AL, ed, Histology and cell biology: an introdution to pathology, 2nd edn. Elsevier-Mosby, Philadelphia Van Furden D, Johnson K, Segbert C, Bossinger O, 2004, The C. elegans ezrin-radixin-moesin protein ERM-1 is necessary for apical junction remodeling and tubulogenesis in the intestine. Dev Biol 272:262–76 Elliott BE, Meens JA, SenGupta SK, Louvard D, Arpin M, 2005, The membrane cytoskeletal crosslinker ezrin is required for metastasis of breast carcinoma cells. Breast Cancer Res 7:R365–73 Tsukita S, Yonemura S, Tsukita S, 1997, ERM, ezrin/radixin/ moesin, family: from cytoskeletal to signal transduction. Curr Opin Cell Biol 9:70–5 Ilmonen S, Vaheri A, Asko-Seljavaara S, Carpen O, 2005, Ezrin in primary cutaneous melanoma. Mod Pathol 18:503–10 Crepaldi T, Gautreau A, Comoglio PM, Louvard D, Arpin M, 1997, Ezrin is an effector of hepatocite growth factor-mediated migration and morphogenesis in ephitelial cells. J Cell Biol 138:423–34 Mackay DJ, Esch F, Furthmayr H, Hall A, 1997, Rho- and racdependent assembly of focal adhesion complexes and actin filaments in permeabilized fibroblasts: an essencial role for ezrin/radixin/moesin proteins. J Cell Biol 138:927–38 Mangeat P, Roy C, Martin M, 1999, ERM proteins in cell adhesion and membrane dynamics. Trends Cell Biol 9:187–92 Pujuguet P, Del Maestro L, Gautreau A, Louvard D, Arpin M, 2003, Ezrin regulates E-cadherin-dependent adherens junction assembly through Rac 1 activation. Mol Biol Cell 14:2181–91 Khanna C, Wan X, Bose S, Cassaday R, Olomu O, Mendoza A, Yeung C, Gorlick R, Hewitt SM, Helman LJ, 2004, The membrane-cytoskeletal linker ezrin is necessary for osteosarcoma metastasis. Nat Med 10(2):182–6 Orian-Rousseau V, Chen L, Sleeman JP, Herrlich P, Ponta H, 2002, CD44 is required for two consecutive steps in HGF/c-met signaling. Genes Dev 16:3074–86 Hiscox S, Jiang WG, 1999, Ezrin regulates cell-cell and cellmatrix adhesion, a possible role with E-cadherin/beta-catenin. J Cell Sci 112:3081–90 Tokunou M, Niki T, Saiotoh Y, Imamura H, Sakamoto M, Hirohashi S, 2000, Altered expression of the ERM proteins in lung adenocarcinoma. Lab Invest 80(11):1643–50 Brescher A, Edwards K, Fehon RG, 2002, ERM proteins and merlin: integrators at the cell cortex. Nat Rev Mol Cell Biol 2:586–99 Fievet BT, Gautreau A, Roy C, Del Maestro L, Mangeat P, Louvard D, Arpin M, 2004, Phophoinositide binding and phosphorylation act sequentially in the activation mechanism of ezrin. J Cell Biol 164:653–59 Valdman A, Fang X, Pang ST, Nilsson B, Ekman P, 2005, Ezrin expression in prostate cancer and benign prostatic tissue. Eur Urol 48:852–57 Yeh TS, Tseng JH, Liu NJ, Chen TC, Jan YY, Chen MF, 2005, Significance of cellular distributio of ezrin in pancreatic cystic neoplasms and ductal adenocarcinoma. Arch Surg 140:1184–90 Kobel M, Langhammer T, Huttelmaier S, Schmitt WD, Kriese K, Dittmer J, Strauss HG, Thomssen C, Hauptmann S, 2006, Ezrin expression is related to poor prognosis in FIGO stage I endometrioid carcinomas. Mod Pathol 19:581–7 Ohtani K, Sakamoto H, Rutherford T, Chen Z, Satoh K, Naftolin F, 1999, Ezrin, a membrane-cytoskeletal linking protein, is involved in the process of invasion of endometrial cancer cells. Cancer Lett 147:31–8 Ohtani K, Sakamoto H, Rutherford T, Chen Z, Kikuchi A, Yamamoto T, Satoh K, Naftolin F, 2002, Ezrin, a membranecytoskeletal linking protein, is highly expressed in atypical endometrial hyperplasia and uterine endometrioid adenocarcinoma. Cancer Lett 179:79–86 Musial J, Sporny S, Nowicki A, 2007, Prognostic significance of E-cadherin and ezrin immunohistochemical expression in prostate cancer. Pol J Pathol 58(4):235–43 Yu Y, Khan J, Khanna C, Helman L, Meltzer PS, Merlino G, 2004, Expression profiling identifies cytoskeletal organizer ezrin and the developmental homeoprotein Six-1 as a key metastatic regulators. Nat Med 10:175–81 Makitie T, Summanen P, Tarkkanen A, Kivela T, 1999, Microvascular density in predicting survival of patients with choroidal and ciliary body melanoma. Invest Opphthalmol Vis Sci 40:2471–80 Foss AJE, Alexander RA, Jefferies LW, Hungerford JL, Harris AL, Lightman S, 1996, Microvessel count predicts survival in uveal melanoma. Cancer Res 56:2900–03 Folkman J, 1995, Clinical applications of research on angiogenesis. N Engl J Med 333:1757–1763 Hunter KW, 2004, Ezrin, a key component in tumor metastasis. Trends Mol Med 10:201–204 Sarrio D, Rodriguez-Pinilla SM, Dotor A, Calero F, Hardisson D, Palacios J, 2006, Abnormal ezrin localization is associated with clinicopathological features in invasive breast carcinomas. Breast Cancer Res and Treat 98(1):71–9 Wald FA, Oriolo AS, Casanova ML, Salas PJ, 2005, Intermediate filaments interaract with dormant ezrin in intestinal epithelial Cells. Mol Biol Cell 16:4096–107 Davies ML, Rmali K, Parr C, Hiscox S, Jiang WG, 2006, Loss of cellular distribution of ezrin in human colon cancer. Paper presented at the 2006 Gastrointestinal Cancers Symposium. ASCO-Abstract p 322. Makitie T, Carpen O, Vaheri A, Kivela T, 2001, Ezrin as a prognostic indicator and its relationship to tumor characteristics in uveal malignant melanoma. Invest Ophthalmol Vis Sci 42:2442–2448 Elzagheid A, Korkeila E, Bendardaf R, Buhmeida A, Heikkila S, Vaheri A, Syrjanen K, Pyrhonen S, Carpen O, 2008, Intense cytoplasmic ezrin immunoreactivity predicts poor survival in colorectal cancer. Hum Pathol 39(12):1737–43 Wang HJ, Zhu JS, Zhang Q, Sun Q, Guo H, 2009, High level of ezrin expression in colorectal cancer tissues is closely related to tumor malignancy. World J Gastroenterol 15(16):2016–9 Weng WH, Ahlen J, Astrom K, Lui WO, Larsson C, 2005, Prognostic impact of immunohistochemical expression of ezrin in highly malignant soft tissue sarcomas. Clin Cancer Res 11, 17):6198–204 Bal N, Yildirim S, Nursal TZ, Bolat F, Kayaselcuk F, 2007, Association of ezrin expression in intestinal and diffuse gastric carcinoma with clinicopathological parameters and tumor type. World J Gastroenterol 13(27):3726–9 Lim JW, Kim H, Kim JM, Kim JS, Jung HC, Kim KH, 2004, Cellular stress-related protein expression in Helicobacter pyloriinfected gastric epithelial AGS cells. Int J Biochem Cell Biol 36, 8):1624–34 Curto M, Al McClatchey, 2004, Ezrin. A metastatic determinant? Cancer Cell 5:113–14 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 827 EP 833 DI 10.1007/s12253-011-9389-4 PG 7 ER PT J AU Hata, M Machi, J Mamou, J Yanagihara, TE Saegusa-Beecroft, E Kobayashi, KG Wong, CMC Fung, C Feleppa, JE Sakamoto, K AF Hata, Masaki Machi, Junji Mamou, Jonathan Yanagihara, T Eugene Saegusa-Beecroft, Emi Kobayashi, K Gregory Wong, C M Clifford Fung, Conway Feleppa, J Ernest Sakamoto, Kazuhiro TI Entire-volume Serial Histological Examination for Detection of Micrometastases in Lymph Nodes of Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Lymph nodes; Micrometastases; Serial histological examination ID Colorectal cancer; Lymph nodes; Micrometastases; Serial histological examination AB The purpose of this study was to accurately detect lymph-node micrometastases, i.e., metastatic cancer foci that have a size between 2.0 and 0.2 mm, in nodes excised from colorectal cancer (CRC) patients, and to determine how frequently micrometastases might be missed when standard histological examination procedures are used. A total of 311 lymph nodes were removed and examined from 90 patients with Stage I to IV CRC. The number of slices of histology sections ranged from 6 to 75 per node (average=25.5; SD=11.1), which provided a total of 7,943 slices. Lymph nodes were examined in their entire volume at every 50-μm and 100-μm intervals for nodes smaller and larger than 5 mm respectively. The total number of thin sections examined in each node and the number of thin sections where metastatic foci were present were counted. The number of thin sections with metastatic foci and the total number of slices was determined for each node. In addition, the presence or absence of metastatic foci in the "central" slice was determined. Micrometastases were found in 12/311 (3.9%) of all lymph nodes. In the 12 lymph nodes with micrometastases, the rate of metastatic slices over all slices was 39.4% (range=6.3 to 81.3%; SD=25.8%) In the central slice of each node, micrometastases were present only in 6 of 12 lymph nodes (50%); accordingly, they were not present in the central slice for half the micrometastatic nodes. These 6 nodes represented 1.9% of the 311 nodes and 11.1% of the 54 metastatic nodes. This study suggests that a significant fraction of micrometastases can be missed by traditional singleslice sectioning; half of the micrometastases would have been overlooked in our data set of 311 nodes. C1 [Hata, Masaki] University of Hawaii and Kuakini Medical Center, Department of Surgery, 405 N. Kuakini Street, Suite 601, 96817 Honolulu, HI, USA. [Machi, Junji] University of Hawaii and Kuakini Medical Center, Department of Surgery, 405 N. Kuakini Street, Suite 601, 96817 Honolulu, HI, USA. [Mamou, Jonathan] Riverside Research InstituteNew York, NY, USA. [Yanagihara, T Eugene] University of Hawaii and Kuakini Medical Center, Department of PathologyHonolulu, HI, USA. [Saegusa-Beecroft, Emi] University of Hawaii and Kuakini Medical Center, Department of Surgery, 405 N. Kuakini Street, Suite 601, 96817 Honolulu, HI, USA. [Kobayashi, K Gregory] University of Hawaii and Kuakini Medical Center, Department of PathologyHonolulu, HI, USA. [Wong, C M Clifford] University of Hawaii and Kuakini Medical Center, Department of PathologyHonolulu, HI, USA. [Fung, Conway] University of Hawaii and Kuakini Medical Center, Department of PathologyHonolulu, HI, USA. [Feleppa, J Ernest] Riverside Research InstituteNew York, NY, USA. [Sakamoto, Kazuhiro] Juntendo University School of Medicine, Department of Coloproctological SurgeryTokyo, Japan. RP Hata, M (reprint author), University of Hawaii and Kuakini Medical Center, Department of Surgery, 96817 Honolulu, USA. EM masakihata4999@yahoo.co.jp CR Greene FL, Page DL, Fleming ID et al, 2002, AJCC cancer staging manual. 6th ed. New York Springer;. Meijerink WJ, van der Pas MH, van der Peet DL et al, 2009, New horizons in colorectal cancer surgery. Surg Endosc 23:1–3 Cohen AM, Kelsen D, Saltz L et al, 1998, Adjuvant therapy for colorectal cancer. Curr Prob Cancer 22:5–65 Gunderson LL, Sargent DJ, Tepper JE et al, 2004, Impact of T and N stage and treatment on survival and relapse in adjuvant rectal cancer: a pooled analysis. J Clin Oncol 22:1785–1796 Tan LK, Giri D, Hummer AJ et al, 2008, Occult Axillary Node Metastases in Breast Cancer Are Prognostically Significant: Results in 368 Node-Negative Patients With 20-Year Follow-Up. J Clin Oncol 26:1803–1809 Maehara Y, Oshiro T, Endo K et al, 1996, Clinical significance of occult micrometastases in lymph nodes from patients with early gastric cancer who died of recurrence. Surgery 119:397–402 Cai J, Ikeguchi M, Maeta M et al, 2000, Micrometastasis in lymph nodes and microinvasion of the muscularis propria in primary lesions of submucosal gastric cancer. Surgery 127:32– 39 Nakajo A, Natsugoe S, Ishigami S et al, 2001, Detection and prediction of micrometastasis in the lymph nodes of patients with pN0 gastric cancer. Ann Surg Oncol 8:158–162 Reed J, Rosman M, Verbanac KM et al, 2009, Prognostic Implications of Isolated Tumor Cells and Micrometastases in Sentinel Nodes of Patients with Invasive Breast Cancer: 10-Year Analysis of Patients Enrolled in the Prospective East Carolina University/ Anne Arundel Medical Center Sentinel Node Multicenter Study. J Am Coll Surg 208:333–340 Izbicki JR, Hosch SB, Pichlmeier U et al, 1997, Prognostic value of immunohistochemically identifiable tumor cells in lymph nodes of patients with completely resected esophageal cancer. N Engl J Med 337:1188–1194 Oberg A, Stenling R, Tavelin B et al, 1998, Are lymph node micrometastases of any clinical significance in Dukes stages A and B colorectal cancer? Dis Colon Rectum 41:1244–1249 Choi H-J, Choi Y-Y, Hong S-H, 2002, Incidence and prognostic implications of isolated tumor cells in lymph nodes from patients with Dukes B colorectal carcinoma. Dis Colon Rectum 45:750– 756 Fisher ER, Colangelo L, Wieand S et al, 2003, Lack of influence of cytokeratin-positive mini micrometastases in “negative node” patients with colorectal cancer: findings from the National Surgical Adjuvant Breast and Bowel Projects protocols R-01 and C-01. Dis Colon Rectum 46:1021–1026 Noura S, Yamamoto H, Miyake Y et al, 2002, Immunohistochemical assessment of localization and frequency of micrometastasis in lymph nodes of colorectal cancer. Clin Cancer Res 8:759–767 Palma RT, Waisberg J, Bromberg SH et al, 2003, Micrometastasis in regional lymph nodes of extirpated colorectal carcinoma: immunohistochemical study using anti-cytokeratin antibodies AE1/AE3. Colorectal Dis 5:164–168 Kronberg U, Lo’pez-Kostner F, Soto G et al, 2004, Detection of lymphatic micrometastasis in patients with stages I and II colorectal cancer: impact on five-year survival. Dis Colon Rectum 47:1151–1157 Messerini L, Cianchi F, Cortesini C et al, 2006, Incidence and prognostic significance of occult tumor cells in lymph nodes from patients with stage IIA colorectal carcinoma. Hum Pathol 37:1259–1267 Clarke G, Ryan E, O’Keane JC et al, 2000, The detection of cytokeratins in lymph node of Duke’s B colorectal cancer subjects predicts a poor outcome. Eur J Gastroenterol Hepatol 12:549–552 Shimoyama M, Yamazaki T, Suda T et al, 2003, Prognostic significance of lateral lymph node micrometastases in lower rectal cancer. Dis Colon Rectum 46:333–339 Sasaki M, Watanabe H, Jass JR et al, 1997, Occult lymph node metastases detected by cytokeratin immunohistochemistry predicts recurrence in “node-negative” colorectal cancer. J Gastroenterol 32:758–764 Isaka N, Nozue M, Doy M et al, 1999, Prognostic significance of perirectal lymph node micrometastases in Dukes’ B rectal carcinoma: an immunohistochemical study by CAM5.2. Clin Cancer Res 5:2065–2068 Yasuda K, Adachi Y, Shiraishi N et al, 2001, Pattern of lymph node micrometastasis and prognosis of patients with colorectal cancer. Ann Surg Oncol 8:300–304 Bukholm IRK, Bondi J, Wiik P et al, 2003, Presence of isolated tumour cells in mesenteric lymph nodes predicts poor prognosis in patients with stage II colon cancer. Eur J Surg Oncol 29:862–866 Bilchik AJ, Stojadinovich A, Wainberg Z et al, 2009, Targeted lymph node evaluation in colorectal cancer: A decade of progress! J Surg Oncol 99:273–274 Iddings D, Ahmad A, Elashoff D et al, 2006, The Prognostic Effect of Micrometastases in Previously Staged Lymph Node Negative, N0, Colorectal Carcinoma: A Meta-analysis. Ann Surg Oncol 13:1386–1392 Hermanek P, Hutter RV, Sobin LH et al, 1999, International Union Against Cancer. Classification of isolated tumor cells and micrometastasis. Cancer 86:2668–2673 Ishii K, Kinami S, Funaki K et al, 2008, Detection of sentinel and non-sentinel lymph node micrometastases by complete serial sectioning and immunohistochemical analysis for gastric cancer. J Exp Clin Cancer Res 27:1–7 Feleppa E, Mamou J, Hata M et al, 2009, Ultrasonic detection of metastases in dissected lymph nodes of cancer patients Acoustical Imaging, Vol.30, Ed. Hua Lee, Klewer Academic, NY, in press) Mamou J, Coron A, Hata M et al, 2009, High-frequency quantitative ultrasound imaging of cancerous lymph-nodes. JJAP 48: 07GK08-1: 07GK08-8 Mamou J, Coron A, Hata M et al, 2010, Three-dimensional High-frequency Characterization of Cancerous Lymph Nodes. Ultrasound Med Biol 36:361–375 Pheby D, Levine D, Pitcher R et al, 2004, Lymph-node harvests directly influence the staging of colorectal cancer: evidence of a regional audit. J Clin Pathol 57:43–47 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 835 EP 841 DI 10.1007/s12253-011-9390-y PG 7 ER PT J AU Kandemir, ON Barut, F Gun, DB Keser, HS Karadayi, N Gun, M Ozdamar, OS AF Kandemir, Onak Nilufer Barut, Figen Gun, Dogan Banu Keser, Hallac Sevinc Karadayi, Nimet Gun, Mustafa Ozdamar, Oguz Sukru TI Lymphatic Differentiation in Classic Kaposi’s Sarcoma: Patterns of D2-40 Immunoexpression in the Course of Tumor Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE D2-40; Immunohistochemistry; Kaposi sarcoma; Lymphatic; Podoplanin ID D2-40; Immunohistochemistry; Kaposi sarcoma; Lymphatic; Podoplanin AB The recent development of lymphatic endotheliumspecific immuno-indicators has given rise to research on the histogenesis of Kaposi sarcoma (KS), specifically focusing on its lymphatic root and differentiation. D2-40 is a new lymphatic marker that recognizes podoplanin and is easily applied to formalin-fixed paraffin-embedded human tissues. This study examined D2-40 immunoexpression in 178 classical KS lesions using immunohistochemical methods. D2-40 immunoexpression was also examined in 63 non-KS soft tissue lesions to test the reliability of D2-40 monoclonal antibody in the pathological diagnosis of KS. D2-40 immunoreactivity was detected at all of the KS lesions and in lymphangioma and nonneoplastic lymphatic endothelium. There was no significant relationship between the extent of D2-40 staining and histopathological stage; however, there was a positive correlation between the staining intensity and histopathological stage in KS cases. D2-40 immunoreactivity was detected at all histopathological stages of KS and may be added to the routine immunohistochemical panel used for the differential diagnosis of KS. Widespread D2-40 protein expression is evidence of a lymphatic origin or the differentiation of neoplastic cells in KS, and D2-40 expression increases with tumor progression. C1 [Kandemir, Onak Nilufer] Zonguldak Karaelmas University, School of Medicine, Department of Pathology, 67600 Kozlu, Zonguldak, Turkey. [Barut, Figen] Zonguldak Karaelmas University, School of Medicine, Department of Pathology, 67600 Kozlu, Zonguldak, Turkey. [Gun, Dogan Banu] Zonguldak Karaelmas University, School of Medicine, Department of Pathology, 67600 Kozlu, Zonguldak, Turkey. [Keser, Hallac Sevinc] Lutfi Kirdar Kartal Training and Research Hospital, Department of PathologyIstanbul, Turkey. [Karadayi, Nimet] Lutfi Kirdar Kartal Training and Research Hospital, Department of PathologyIstanbul, Turkey. [Gun, Mustafa] Ataturk State Hospital, Department of PathologyZonguldak, Turkey. [Ozdamar, Oguz Sukru] Zonguldak Karaelmas University, School of Medicine, Department of Pathology, 67600 Kozlu, Zonguldak, Turkey. RP Kandemir, ON (reprint author), Zonguldak Karaelmas University, School of Medicine, Department of Pathology, 67600 Kozlu, Turkey. EM niluferkandemir@yahoo.com CR Weiss SW, Goldblum JR, 2001, Malignant vascular tumours. In: Weiss SW, Goldblum JR, eds, Enzinger and Weiss’s soft tissue tumors, 4th edn. Mosby, St Louis, pp 917–53 Antman K, Chang Y, 2000, Kaposi’s sarcoma. N Engl J Med 342:1027–1038 Pantanowitz L, Grayson W, Simonart T et al, 2009, Pathology of Kaposi’s sarcoma. J HIV Ther 14:41–47 Tapparo JW, Conant MA, Wolfe SF et al, 1993, Kaposi’s sarcoma. Epidemiology, pathogenesis, histology, clinical spectrum, staging criteria and therapy. J Am Acad Dermatol 28:371–395 Ensoli B, Sgadari C, Barillari G et al, 2001, Biology of Kaposi’s sarcoma. Eur J Cancer 37:1251–1269 Douglas JL, Gustin JK, Dezube B et al, 2007, Kaposi’s sarcoma: a model of both malignancy and chronic inflammation. Panminerva Med 49:119–138 Noel JC, Hermans P, Andre J et al, 1996, Herpesvirus-like DNA sequences and Kaposi’s sarcoma: relationship with epidemiology, clinical spectrum, and histologic features. Cancer 77:2132–2136 Viejo-Borbolla A, Schulz TF, 2003, Kaposi’s sarcoma-associated herpesvirus, KSHV/HHV8): key aspects of epidemiology and pathogenesis. AIDS Rev 5:222–229 Boshoff C, Weiss RA, 2001, Epidemiology and pathogenesis of Kaposi’s sarcoma-associated herpesvirus. Philos Trans R Soc Lond B Biol Sci 29:517–534 Beckstead JH, Wood GS, Fletcher V, 1985, Evidence for the origin of Kaposi’s sarcoma from lymphatic endothelium. Am J Pathol 119:294–300 Jones RR, Spaull JS, Spry C et al, 1986, Histogenesis of Kaposi’s sarcoma in patients with and without acquired immune deficiency syndrome, AIDS). J Clin Pathol 39:742–749 Karasek MA, 1994, Origin of spindle-shaped cells in Kaposi’s sarcoma. Lymphology 27:41–44 Facchetti F, Lucini L, Gavazzoni R et al, 1988, Immunomorphological analysis of the role of blood vessel endothelium in the morphogenesis of cutaneous Kaposi’s sarcoma: a study of 57 cases. Histopathology 12:581–593 Sankey EA, More L, Dhillon AP, 1990, QBEnd/10: a new immunostain for the routine diagnosis of Kaposi’s sarcoma. J Pathol 161:267–271 Russell Jones R, Orchard G et al, 1995, Immunostaining for CD31 and CD34 in Kaposi sarcoma. J Clin Pathol 48:1011–1016 Hoerl HD, Goldbloom JR, 1997, Immunoreactivity patterns of CD31and CD68 in 28 cases of Kaposi’s sarcoma: evidence supporting endothelial differentiation in the spindle cell compartment. Applied Immunohistochemistry 5:173–178 Kalof AN, Cooper K, 2009, D2–40 immunohistochemistry–so far! Adv Anat Pathol 16:62–64 Jussila L,Valtola R, Partanen TA et al, 1998, Lymphatic endothelium and Kaposi’s sarcoma spindle cells detected by antibodies against the vascular endothelial growth factor receptor-3. Cancer Res 58:1599– 1604 Weninger W, Partanen TA, Breiteneder-Geleff S et al, 1999, Expression of vascular endothelial growth factor receptor-3 and podoplanin suggests a lymphatic endothelial cell origin of Kaposi’s sarcoma tumor cells. Lab Invest 79:243–251 Kahn HJ, Bailey D, Marks A, 2002, Monoclonal antibody D2-40, a new marker of lymphatic endothelium, reacts with Kaposi"s sarcoma and a subset of angiosarcomas. Mod Pathol 15:434–440 Xu H, Edwards JR, Espinosa O et al, 2004, Expression of a lymphatic endothelial cell marker in benign and malignant vascular tumors. Hum Pathol 35:857–861 Pyakurel P, Pak F, Mwakigonja AR et al, 2006, Lymphatic and vascular origin of Kaposi"s sarcoma spindle cells during tumor development. Int J Cancer 119:1262–1267 Dubina M, Goldenberg G, 2009, Positive staining of tumor-stage Kaposi sarcoma with lymphatic marker D2-40. J Am Acad Dermatol 61:276–280 Ramirez-Amador V, Martinez-Mata G, Gonzalez-Ramirez I et al, 2009, Clinical, histological, and immunohistochemical findings in oral Kaposi"s sarcoma in a series of Mexican AIDS patients. Comparative study. J Oral Pathol Med 38:328–333 Taddeo A, Presicce P, Brambilla L et al, 2008, Circulating endothelial progenitor cells are increased in patients with classic Kaposi"s sarcoma. J Invest Dermatol 128:2125–2128 Ordonez NG, 2006, Podoplanin: a novel diagnostic immunohistochemical marker. Adv Anat Pathol 13:83–88 Arai E, Kuramochi A, Tsuchida T et al, 2006, Usefulness of D2-40 immunohistochemistry for differentiation between Kaposiform hemangioendothelioma and tufted angioma. J Cutan Pathol 33:492– 497 Breiteneder-Geleff S, Soleiman A, Kowalski H et al, 1999, Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries. Podoplanin as a specific marker for lymphatic endothelium. Am J Pathol 154:385–394 Lamovec J, Knuutila S, 2002, Kaposi sarcoma. In: Fletcher CDM, Unni KK, Mertens F, eds, World health organisation classification of tumours. Pathology and genetics of tumours of soft tissue and bone. IARC, Lyon, pp 170–172 FukunagaM(2005, Expression of D2-40 in lymphatic endothelium of normal tissues and in vascular tumors. Histopathology 46:396–402 Marks A, Sutherland DR, Bailey D et al, 1999, Characterization and distribution of an oncofetal antigen, M2A antigen, expressed on testicular germ cell tumors. Br J Cancer 80:569–578 Miettinen M, Lindenmayer AE, Chaubal A, 1994, Endothelial cell markers CD31, CD34, and BNH9 antibody to H- and Yantigens—evaluation of their specificity and sensitivity in the diagnosis of vascular tumors and comparison with von Willebrand factor. Mod Pathol 7:82–90 Nadimi H, Saatee S, Armin A et al, 1988, Expression of endothelial cell markers PAL-E and EN-4 and Ia-antigens in Kaposi"s sarcoma. J Oral Pathol 17:416–420 Cheung L, Rockson SG, 2005, The lymphatic biology of Kaposi’s sarcoma. Lymphat Res Biol 3:25–35 Carroll PA, Brazeau E, Lagunoff M, 2004, Kaposi’s sarcomaassociated herpesvirus infection of blood endothelial cells induces lymphatic differentiation. Virology 328:7–18 Sakakibara S, Tosato G, 2009, Regulation of angiogenesis in malignancies associated with Epstein-Barr virus and Kaposi"s sarcoma-associated herpes virus. Future Microbiol 4:903–917 Komatsu T, Ballestas ME, Barbera AJ et al, 2002, The KSHV latency-associated nuclear antigen: a multifunctional protein. Front Biosci 7:726–730 Wang HW, Trotter MW, Lagos D et al, 2004, Kaposi sarcoma herpesvirus-induced cellular reprogramming contributes to the lymphatic endothelial gene expression in Kaposi sarcoma. Nat Genet 36:687–693 Sturzl M, Brandstetter H, Roth KW, 1992, Kaposi’s sarcoma: a review of gene expression and ultrastructure of KS spindle cells in vivo. AIDS Res Hum Retroviruses 8:1753–1762 Hong YK, Foreman K, Shin JW et al, 2004, Lymphatic reprogramming of blood vascular endothelium by Kaposi sarcoma-associated herpesvirus. Nat Genet 36:683–685 Marchevsky AM, 2008, Application of immunohistochemistry to the diagnosis of malignant mesothelioma. Arch Pathol Lab Med 132:397–401 Yu H, Pinkus GS, Hornick JL, 2007, Diffuse membranous immunoreactivity for podoplanin, D2-40, distinguishes primary and metastatic seminomas from other germ cell tumors and metastatic neoplasms. Am J Clin Pathol 128:767–775 Shibahara J, Kashima T, Kikuchi Y et al, 2006, Podoplanin is expressed in subsets of tumors of the central nervous system. Virchows Arch 448:493–499 Yu H, Gibson JA, Pinkus GS et al, 2007, Podoplanin, D2-40, is a novel marker for follicular dendritic cell tumors. Am J Clin Pathol 128:776–782 Sauter B, Foedinger D, Sterniczky B et al, 1998, Immunoelectron microscopic characterization of human dermal lymphatic microvascular endothelial cells. Differential expression of CD31, CD34, and type IV collagen with lymphatic endothelial cells vs blood capillary endothelial cells in normal human skin, lymphangioma, and hemangioma in situ. J Histochem Cytochem 46:165–176 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 843 EP 851 DI 10.1007/s12253-011-9392-9 PG 9 ER PT J AU Szabo, VG Acsady, Gy AF Szabo, Viktor Gabor Acsady, Gyorgy TI Tumornecrosis-Factor-α 308 GA Polymorphism in Atherosclerotic Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tumornecrosis-factor-α; Polymorphism; Atherosclerosis; Cardiovascular risk ID Tumornecrosis-factor-α; Polymorphism; Atherosclerosis; Cardiovascular risk AB The development of the atherosclerosis is a multifactorial process, where the clinical pattern is determined by environmental and genetic factors. Except for the classical risk factors of atherosclerosis (hypertension, lipid-metabolic disorders, diabetes, smoking) the clinical signs can be influenced by the genetic variants (polymorphisms) of the enzymes, which are responsible for the endothelial cell function and for the thrombotic factors. In our examination our aim was to define the TNF-α 308GA polymorphisms in atherosclerotic diabetic, atherosclerotic non-diabetic and healthy patients. We found correlation of the frequency of myocardial infarction and stroke in atherosclerotic diabetic and atherosclerotic non-diabetic patients. We proved that among patients with mutant TNF-α AA genotype the occurrence of cardiovascular events is significantly higher: Mutant AA homozygous genotype: control group 1, 6%, MI group 10,7%, p<0,005, OR: 8,17 versus Normal GG allele: control group 76,7%, MI group 61,3%. The TNF-α AA genotype can have a clinical importance as a prognostic and therapeutic marker, although further studies are needed to confirm this hypothesis. C1 [Szabo, Viktor Gabor] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor str. 68, 1122 Budapest, Hungary. [Acsady, Gyorgy] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor str. 68, 1122 Budapest, Hungary. RP Szabo, VG (reprint author), Semmelweis University, Department of Cardiovascular Surgery, 1122 Budapest, Hungary. EM szabogvdr@gmail.com CR Yang Z, Ming X, 2006, Recent advances in understanding endothelial dysfunction in atherosclerosis. Clin Med Res 4, 1):53–65 Blanke G, Ridker P, 2001, Inflammatory mechanisms in atherosclerosis: from laboratory evidence to clinical application. Ital Heart J 2(11):796–800 Ablij HC, Meinders AE, 2002, C-reactive protein: history and revival. Eur J Int Med 13:412–422 Ridker P, Rifai N, Pfeffer M, Sacks F, Lepage S, Braunwald E, 2000, Elevation of tumor necrosis factor-alfa, and increased risk of recurrent coronary events after myocardial infarction. Circulation 101:2149–2153 Stenvinkel P, 2001, Endothelial dysfunction and inflammation— is there a link? Nephrol Dial Transplant 16:1968–1971 Shah P, 1998, Role of infalammation and metalloproteinases in plaque disruption and thrombosis. Vasc Med 3:199–206 Geisler T, Bhatt DL, 2004, The role of inflammation in atherosclerosis: current and future strategies of medical treatment. Med Sci Monit 10(12):308–316 Stoll G, Bendzus M, 2006, Inflammation and atherosclerosis: novel insights into plaque formation and destabilization. Stroke 37:1923–1932 Fiotti N, Giansante C, Ponte E, Delbello C, Calabrese S, Zacchi T, Dobrina A, Guarnieri G, 1999, Atherosclerosis and inflammation. Patterns of cytokine regulation in patients with periferial arterial disease. Atherosclerosis 145:51–60 Libby P, Ridker PM, Maseri A, 2002, Inflammation and Atherosclerosis. Circulation 105:1135–1143 Monraats PS, Pires NMM, Schepers A, Agema W, Boesten L, Vries M, Zwinderman A, de Maat M, Doevendans P, Winter R, Tio R, Waltenberger J, Hart L, Frants R, Quax P, Vlijmen B, Havekes L, Laarse A, Wall E, Jukema W, 2005, Tumor necrosis factor- α plays and important role in restenosis development The. FASEB J 19:1998–2004 Dalziel B, Gosby A, Richman R, Bryson J, Caterson I, 2002, Association of the TNF-α, −308 G/A, promoter polymorphism with insulin resistance in obesity. Obes Res 10(5):401–407 Hotamisligil G, 2006, Inflammation and metabolic disorders. Nature 444(14):860–867 Nieszner E, Bardos P, Baranyi E, Preda I, 2005, A diabetes mellitus cardiovascularis szovodmenyei es diagnozisuk. LAM 15, 10):722–729 Vendrell J, Fernandez-Real JM, Gutierrez C, Gutierrez C, Zamora A, Simon I, Bardaji A, Ricart W, Richart C, 2003, A polymorphysm in the promoter of the tumor necrosis facrtor-α gene, −308, is associated with coronary heart disease in type 2 diabetic patients. Atherosclerosis 167:257–264 Fernandez-Real JM, Gutierrez C, Ricart W, Casamitjana R, Fernandez-Castaner M, Vendrell J, Richart C, Soler J, 9, 1997, The TNF-alpha gene Nco I polymorphism influences the relationship among insulin resistance, percent body fat, and increased serum leptin levels. Diabetes 46(9):2096–2098 Welt FGP, Rogers C, 2002, Inflammation and restenosis in the stent era. Arterioscler Thromb Vasc Biol 22:1769–1776 Szabo GV, Kunstar A, 2009, Acsady Gy.: methylentetrahydrofolate reductase and nitric oxid synthase polymorphysm in patients with atherosclerosis and diabetes. Pathol Oncol Res 15:631–637 Ascer E, Bertolami M, Venturinelli M, Buccheri V, Souza J, Nicolau J, Ramires JA, Serrano C, 2004, Atorvastatin reduces proinflammatory markers in hypercholesterolemic patients. Atherosclerosis 177:161–166 Goodwin BL, Pendleton LC, Levy MM, Solomonson LP, Eichler DC, 2007, Tumor necrosis factor- α reduces argininosuccinate synthase expression and nitrix oxide production in aortic endothelial cells. Am J Physiol Heart Circ Physiol 293:1115–1121 Wilson AG, Symons JA, McDowell TL, McDevitt H, 1997, Effects of a polymorphismin the human tumor necrosis factor α promoter on transcriptional activation. Proc Natl Acad Sci USA 94:3195–3199 Herrmann SM, Ricard S, Nicaud V, Arveiler D, Evans A, Ruidavets JB, Luc G, Bara L, Parra HJ, Poirier O, Cambien F, 1988, Polymorphisms of the tumour necrosis factir- α gene, coronary heart diseases and obesity. Eur J Clin Invest 92:14–8 Elkind MS, Cheng J, Boden-Albala B, Rundek T, Thomas J, Chen H, Rabbani LE, Sacco RL, 2002, Tumor necrosis factor receptor levels are associated with carotid atherosclerosis. Stroke 33:31–38 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 853 EP 857 DI 10.1007/s12253-011-9393-8 PG 5 ER PT J AU Liu, Dc Yang, ZL Jiang, S AF Liu, Dong-cai Yang, Zhu-Lin Jiang, Song TI Identification of PEG10 and TSG101 as Carcinogenesis, Progression, and Poor-Prognosis Related Biomarkers for Gallbladder Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gallbladder cancer; Gallbladder polyp; Chronic cholecystitis; PEG10; TSG101 ID Gallbladder cancer; Gallbladder polyp; Chronic cholecystitis; PEG10; TSG101 AB PEG10 is a transcriptional factor while TSG101 is involved in numerous cellular processes, including apoptotic resistance. Overexpression of PEG10 and TSG101 were observed in a variety of human cancers. However, their expression and clinical significance in gallbladder cancer (GBC) have not yet been identified. To understand the tumor biology of GBC at the molecular level, we examined PEG10 and TSG101 expression in 108 adenocarcinomas, 15 gallbladder polyps, 35 chronic cholecystitis tissues, and 46 peritumoral tissues by using immunohistochemistry. Overexpression of PEG10 and TSG101 was detected in gallbladder adenocarcinoma (48.1% and 47.2%, respectively). Conversely, there was less expression detected in the peritumoral tissues (19.6%), adenomatous polyps (13.3%), and gallbladder epithelium with chronic cholecystitis (5.1%) (p<0.01, p<0.05, and p<0.01, respectively). Notably, the benign lesions with positive PEG10 and/or TSG101 expression showed moderately or severely atypical hyperplasia in gallbladder epithelium. The overexpression of PEG10 and TSG101 was significantly associated with differentiation, tumor mass, lymph node metastasis and invasion of adenocarcinoma. Univariate Kaplan-Meier analysis showed that overexpression of PEG10 (p=0.041) and TSG101 (p=0.025) was closely associated with decreased overall survival. Multivariate Cox regression analysis revealed that positive expression of PEG10 (p=0.036) or TSG101 (p=0.022) is a predictor of poor prognosis in gallbladder adenocarcinoma. Our study suggested that overexpression of PEG10 and TSG101 might be closely related to the carcinogenesis, progression, clinical biological behaviors, and prognosis of gallbladder adenocarcinoma. C1 [Liu, Dong-cai] Central South University, Second Xiangya Hospital, Department of Geriatric Surgery, 410011 Changsha, Hunan, China. [Yang, Zhu-Lin] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, Hunan, China. [Jiang, Song] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, Hunan, China. RP Yang, ZL (reprint author), Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, China. EM yangzhulin8@sina.com CR Ono R, Kobayashi S, Wagatsuma H et al, 2001, A retrotransposonderived gene, PEG10, is a novel imprinted gene located on human chromosome 7q21. Genomics 73:232–237 Okabe H, Satoh S, Furukawa Y et al, 2003, Involvement of PEG10 in human hepatocellular carcinogenesis through interaction with SIAH1. Cancer Res 63:3043–3048 Kainz B, Shehata M, Bilban M et al, 2007, Overexpression of the paternally expressed gene 10, PEG10, from the imprinted locus on chromosome 7q21 in high-risk B-cell chronic lymphocytic leukemia. Int J Cancer 121:1984–1993 Tsuji K, Yasui K, Gen Y et al, 2010, PEG10 is a probable target for the amplification at 7q21 detected in hepatocellular carcinoma. Cancer Genet Cytogenet 198:118–125 Chunsong H, Yuling H, Li Wet al, 2006, CXC chemokine ligand 13 and CC chemokine ligand 19 cooperatively render resistance to apoptosis in B cell lineage acute and chronic lymphocytic leukemia CD23 + CD5+ B cells. J Immunol 177:6713–6722 Li CM, Margolin AA, Salas M et al, 2006, PEG10 is a c-MYC target gene in cancer cells. Cancer Res 66:665–672 Ip WK, Lai PB, Wong NL et al, 2007, Identification of PEG10 as a progression related biomarker for hepatocellular carcinoma. Cancer Lett 250:284–91 Jie X, Lang C, Jian Q et al, 2007, Androgen activates PEG10 to promote carcinogenesis in hepatic cancer cells. Oncogene 26:5741– 5751 Lux A, Beil C, Majety M et al, 2005, Human retroviral gag- and gag-pol-like proteins interact with the transforming growth factorbeta receptor activin receptor-like kinase 1. J Biol Chem 280:8482–8493 Ono R, Nakamura K, Inoue K et al, 2006, Deletion of Peg10, an imprinted gene acquired from a retrotransposon, causes early embryonic lethality. Nat Genet 38:101–106 Liu RT, Huang CC, You HL et al, 2002, Overexpression of tumor susceptibility gene TSG101 in human papillary thyroid carcinomas. Oncogene 21:4830–4837 Young TW, Rosen DG, Mei FC et al, 2007, Up-regulation of tumor susceptibility gene 101 conveys poor prognosis through suppression of p21 expression in ovarian cancer. Clin Cancer Res 13:3848–3854 Koon N, Schneider-Stock R, Sarlomo-Rikala M et al, 2004, Molecular targets for tumour progression in gastrointestinal stromal tumours. Gut 53:235–240 Ma XR, Edmund Sim UH, Pauline B et al, 2008, Overexpression of WNT2 and TSG101 genes in colorectal carcinoma. Trop Biomed 25:46–57 Li L, Liao J, Ruland J et al, 2001, A TSG101/MDM2 regulatory loop modulates MDM2 degradation and MDM2/p53 feedback control. Proc Natl Acad Sci USA 98:1619–1624 Li L, Cohen SN, 1996, Tsg101: a novel tumor susceptibility gene isolated by controlled homozygous functional knockout of allelic loci in mammalian cells. Cell 85:319–329 Zhu G, Gilchrist R, Borley N et al, 2004, Reduction of TSG101 protein has a negative impact on tumor cell growth. Int J Cancer 109:541–547 Bashirova AA, Bleiber G, Qi Y et al, 2006, Consistent effects of TSG101 genetic variability on multiple outcomes of exposure to human immunodeficiency virus type 1. J Virol 80:6757–6763 Goldin RD, Roa JC, 2009, Gallbladder cancer: a morphological and molecular update. Histopathology 55:218–229 Gatto M, Bragazzi MC, Semeraro R et al, 2010, Cholangiocarcinoma: update and future perspectives. Dig Liver Dis 42:253–260 Jayaraman S, Jarnagin WR, 2010, Management of gallbladder cancer. Gastroenterol Clin North Am 39:331–342 Maurya SK, Tewari M, Mishra RR, 2010, Genetic abberations in gallbladder cancer. Surg Oncol Sep 28. [Epub ahead of print] Sanada Y, Yoshida K, Ohara M, Tsutani Y, 2007, Expression of orotate phosphoribosyltransferase, OPRT, in hepatobiliary and pancreatic carcinoma. Pathol Oncol Res 13:105–113 Chang HJ, Yoo BC, Kim SW, Lee BL, Kim WH, 2007, Significance of PML and p53 protein as molecular prognostic markers of gallbladder carcinomas. Pathol Oncol Res 13:326–335 Nemeth Z, Szasz AM, Somoracz A, Tatrai P, Nemeth J et al, 2009, Zonula Occludens-1, Occludin, and E-cadherin Protein Expression in Biliary Tract cancers. Pathol Oncol Res 15:533–539 Li Q, Yang Z, 2009, Expression of phospho-ERK1/2 and PI3-K in benign and malignant gallbladder lesions and its clinical and pathological correlations. J Exp& Clin Cancer Res 28:65–72 Li Q, Yang Z, Liu J, et al, 2010). Expression of CDx2 and Hepatocyte antigen in benign and malignant lesions of gallbladder and its correlation with histopathologic typr and clinical outcome. Pathol Oncol Res Dec 28. 2010 [Epub ahead of print] Malik IA, 2004, Gallbladder cancer: current status. Expert Opin Pharmacother 5:1271–1277 Tucek S, Tomasek J, Halamkova J, 2010, Bile duct malignancies. Klin Onkol 23:231–241 Zhao Y, You H, Liu F et al, 2002, Differentially expressed gene profiles between multidrug resistant gastric adenocarcinoma cells and their parental cells. Cancer Lett 185:211–218 Shen H, Pan Y, Han Z et al, 2004, Reversal of multidrug resistance of gastric cancer cells by downregulation of TSG101 with TSG101siRNA. Cancer Biol Ther 3:561–565 Lee CT, Park KH, Yanagisawa K et al, 2004, Combination therapy with conditionally replicating adenovirus and replication defective adenovirus. Cancer Res 64:6660–6665 Lee CT, Lee YJ, Kwon SY et al, 2006, In vivo imaging of adenovirus transduction and enhanced therapeutic efficacy of combination therapy with conditionally replicating adenovirus and adenovirus-p27. Cancer Res 66:372–377 Huang Q, Zhang X, Wang H et al, 2004, A novel conditionally replicative adenovirus vector targeting telomerase-positive tumor cells. Clin Cancer Res 10:1439–1445 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 859 EP 866 DI 10.1007/s12253-011-9394-7 PG 8 ER PT J AU Ruppenthal, DR Nicolini, C Filho, FFA Meurer, R Damin, PA Rohe, A Alexandre, OPC Damin, CD AF Ruppenthal, D Rubia Nicolini, Carmela Filho, Fabiano Ferreira Antonio Meurer, Rosalva Damin, P Andrea Rohe, Adriana Alexandre, O P Claudio Damin, C Daniel TI TWIST1 Promoter Methylation in Primary Colorectal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; TWIST1; Methylation; Prognosis; Colon ID Colorectal cancer; TWIST1; Methylation; Prognosis; Colon AB TWIST1 gene, a transcription factor that belongs to the family of basic helix–loop–helix proteins, has been related to tumor progression and metastasis in different cancers. The aim of our study was to investigate TWIST1 promoter methylation in patients with primary colorectal carcinoma and determine its correlation with prognostic factors and disease outcome. Seventy-three patients with primary colorectal adenocarcinoma were studied. From each patient two tissue samples were collected: one sample of the tumor and one sample of normal colorectal tissue from an area located 15 cm away from the tumor. Samples of colorectal mucosa obtained from 30 individuals without malignant disease were also studied as a control group. All tissues were analyzed through methylation-specific PCR. TWIST1 hypermethylation was detected in colorectal specimens of 46 patients with cancer, but in none of the tissues from the nonmalignant control group (p<0.001). In cancer patients, TWIST1 hypermethylation was found in 38 of 73 tumor samples as compared with 20 of 73 matched samples of non-cancerous colorectal tissue (P=0.001). TWIST1 hypermethylation was not correlated with prognostic predictors for the disease outcome, patients’ overall survival and disease-free survival rates. We concluded that TWIST1 hypermethylation is present in the colon and rectum of most patients with colorectal carcinoma, suggesting this molecular alteration may be involved in the process of colorectal carcinogenesis. C1 [Ruppenthal, D Rubia] Methodist University Center (IPA), Laboratory of Molecular BiologyPorto Alegre, RS, Brazil. [Nicolini, Carmela] Federal University of Health Sciences of Porto Alegre, Laboratory of Molecular BiologyPorto Alegre, RS, Brazil. [Filho, Fabiano Ferreira Antonio] Federal University of Health Sciences of Porto Alegre, Laboratory of Molecular BiologyPorto Alegre, RS, Brazil. [Meurer, Rosalva] Federal University of Health Sciences of Porto Alegre, Laboratory of Molecular BiologyPorto Alegre, RS, Brazil. [Damin, P Andrea] Federal University of Health Sciences of Porto Alegre, Laboratory of Molecular BiologyPorto Alegre, RS, Brazil. [Rohe, Adriana] Federal University of Health Sciences of Porto Alegre, Laboratory of Molecular BiologyPorto Alegre, RS, Brazil. [Alexandre, O P Claudio] Federal University of Health Sciences of Porto Alegre, Laboratory of Molecular BiologyPorto Alegre, RS, Brazil. [Damin, C Daniel] Federal University of Rio Grande do Sul, Department of Surgery, Rua Ramiro Barcelos 2350, 035-903 Porto Alegre, RS, Brazil. RP Damin, CD (reprint author), Federal University of Rio Grande do Sul, Department of Surgery, 035-903 Porto Alegre, Brazil. EM damin@terra.com.br CR Issa JP, 2000, The epigenetics of colorectal cancer. Ann N Y Acad Sci 910:140–153 Lee S, Hwang KS, Lee HJ et al, 2004, Aberrant CpG island hypermethylation of multiple genes in colorectal neoplasia. Lab Invest 84:884–893 Toyota M, Ahuja N, Ohe-Toyota M et al, 1999, CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci U S A 96:8681–8686 Weisenberger DJ, Siegmund KD, Campan M et al, 2006, CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 38:787–793 Teodoridis JM, Hardie C, Brown R, 2008, CpG island methylator phenotype, CIMP, in cancer: causes and implications. Cancer Lett 268:177–186 Fearon ER, Vogelstein B, 1990, A genetic model for colorectal tumorigenesis. Cell 61:759–767 Feinberg AP, Ohlsson R, Henikoff S, 2006, The epigenetic progenitor origin of human cancer. Nat Rev Genet 7:21–33 Fackler MJ, McVeigh M, Evron E et al, 2003, DNA methylation of RASSF1A, HIN-1, RAR-beta, Cyclin D2 and TWIST in situ and invasive lobular breast carcinoma. Int J Cancer 107:970–975 Gort EH, Suijkerbuijk KP, Roothaan SM et al, 2008, Methylation of the TWIST1 promoter, TWIST1 mRNA levels, and immunohistochemical expression of TWIST1 in breast cancer. Cancer Epidemiol Biomarkers Prev 17:3325–3330 Kwok WK, Ling MT, Lee TW et al, 2005, Up-regulation of TWIST in prostate cancer and its implication as a therapeutic target. Cancer Res 65:5153–5162 Fuchtbauer EM, 1995, Expression of M-TWIST during postimplantation development of the mouse. Dev Dyn 204:316–322 Maestro R, Dei Tos AP, Hamamori Y et al, 1999, TWIST is a potential oncogene that inhibits apoptosis. Genes Dev 13:2207–2217 Mironchik Y, Winnard PT Jr, Vesuna F et al, 2005, TWIST1 overexpression induces in vivo angiogenesis and correlates with chromosomal instability in breast cancer. Cancer Res 65:10801–10809 Elias MC, Tozer KR, Silber JR et al, 2005, TWIST1 is expressed in human gliomas and promotes invasion. Neoplasia 7:824–837 Valdes-Mora F, Gomez del Pulgar T, Bandres E et al, 2009, TWIST1 overexpression is associated with nodal invasion and male sex in primary colorectal cancer. Ann Surg Oncol 16:78–87 Kyo S, Sakaguchi J, Ohno S et al, 2006, High TWIST1 expression is involved in infiltrative endometrial cancer and affects patient survival. Hum Pathol 37:431–438 Vesuna F, van Diest P, Chen JH, Raman V, 2008, Twist is a transcriptional repressor of E-cadherin gene expression in breast cancer. Biochem Biophys Res Commun 367:235–241 Rosivatz E, Becker I, Specht K et al, 2002, Differential expression of the epithelial-mesenchymal transition regulators snail, SIP1, and TWIST1 in gastric cancer. Am J Pathol 161:1881–1891 Yan-Qi Z, Xue-Yan G, Shuang H et al, 2007, Expression and significance of TWIST basic helix-loop-helix protein overexpression in gastric cancer. Pathology 39:470–475 Yuen HF, Chan YP, Wong ML et al, 2007, Upregulation of Twist in oesophageal squamous cell carcinoma is associated with neoplastic transformation and distant metastasis. J Clin Pathol 60:510–514 Ohuchida K, Mizumoto K, Ohhashi S et al, 2007, Twist, a novel oncogene, is upregulated in pancreatic cancer: clinical implication of Twist expression in pancreatic juice. Int J Cancer 120:1634–1640 American Joint Committee on Cancer, AJCC,, 1988, Manual for staging of cancer, 3rd edn. Lippincott, Philadelphia Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB, 1996, Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S A 93:9821–9826 Leslie A, Carey FA, Pratt NR, Steele RJ, 2002, The colorectal adenoma-carcinoma sequence. Br J Surg 89:845–860 Peltomaki P, 2001, Deficient DNA mismatch repair: a common etiologic factor for colon cancer. Hum Mol Genet 10:735–740 Baylin SB, Ohm JE, 2006, Epigenetic gene silencing in cancer - a mechanism for early oncogenic pathway addiction? Nat Rev Cancer 6:107–116 Frigola J, Sole X, Paz MF et al, 2005, Differential DNA hypermethylation and hypomethylation signatures in colorectal cancer. Hum Mol Genet 14:319–326 Yang J, Mani SA, Donaher JL et al, 2004, TWIST1, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117:927–939 Yang J, Mani SA, Weinberg RA, 2006, Exploring a new TWIST1 on tumor metastasis. Cancer Res 66:4549–4552 Okada T, Suheiro Y, Ueno K et al, 2010, TWIST1 hypermethylation is observed frequently in colorectal tumors and its overexpression is associated with unfavorable outcomes in patients with colorectal cancer. Genes Chromosomes Cancer 49:452–462 Martin TA, Goyal A, Watkins G, Jiang WG, 2005, Expression of the transcription factors snail, slug, and TWIST1 and their clinical significance in human breast cancer. Ann Surg Oncol 12:488–496 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 867 EP 872 DI 10.1007/s12253-011-9395-6 PG 6 ER PT J AU Cai, Ch Jiang, Fn Liang, Yx He, Hch Han, Zd Dai, Qsh Qin, Gq Chen, Jh Chen, Xb Chen, Yr Zeng, Gh Zhu, Jg Zhong, Wd AF Cai, Chao Jiang, Fu-neng Liang, Yu-xiang He, Hui-chan Han, Zhao-dong Dai, Qi-shan Qin, Guo-qiang Chen, Jia-hong Chen, Xi-bin Chen, Yan-ru Zeng, Guo-hua Zhu, Jian-guo Zhong, Wei-de TI Classical and Alternative Nuclear Factor-κB Pathways: A Comparison among Normal Prostate, Benign Prostate Hyperplasia and Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Immunohistochemistry; Nuclear factor κB; Prognosis; Prostate cancer; Real-time quantitative RT-PCR ID Immunohistochemistry; Nuclear factor κB; Prognosis; Prostate cancer; Real-time quantitative RT-PCR AB Nuclear factor-κB (NF-κB) is controlled by the classical and alternative NF-κB pathways, the role of which in prostate cancer (PCa) is not clearly defined. To provide this missing translational link, we compared the classical and alternative NF-κB pathways in normal prostate, benign prostate hyperplasia (BPH) and PCa. Prostate specimens were divided into three groups: group A, PCa (n=68); group B, BPH (n=60); and group C, normal prostates (n=15). The gene expression levels of NF-κB1 and NF-κB2 were determined by real-time quantitative RT-PCR. Additionally, we analyzed the expression and sub-cellular localization of phosphorylated P50 (p-P50) and phosphorylated P52 (p-P52) proteins by immunohistochemical staining. Furthermore, associations were made between NF-κB pathway proteins and patients’ prognosis. Compared with BPH and normal prostate tissues, the expression of NF-κB1 gene was differentially down-regulated by >1.5-fold, whereas NF-κB2 gene was differentially up-regulated by >2-fold in PCa tissues. The proportion of p-P50 positive patients in group A (26.5%) was significantly lower than in group B (88.3%, p=0.005) and C (100%, p=0.002). The proportion of p-P52 positive patients in group A (42.6%) was significantly higher than in group B (11.7%, p=0.009) and C (6.7%, p=0.008). Comparison of the survival curves in group A according to p-P52 expression showed a significant difference between positive and negative patients. The p-P52 positive patients showed worse prognosis (p=0.019). Our findings suggest for the first time that the classical and alternative NF-κB pathways have an important role in PCa. p-P52 might be a predictor of poor prognosis for PCa. C1 [Cai, Chao] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Jiang, Fu-neng] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Liang, Yu-xiang] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [He, Hui-chan] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Han, Zhao-dong] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Dai, Qi-shan] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Qin, Guo-qiang] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Chen, Jia-hong] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Chen, Xi-bin] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Chen, Yan-ru] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Zeng, Guo-hua] Guangzhou Medical College, First Affiliated Hospital of Guangzhou Medical College, Urology Key Laboratory of Guangdong Province, 510230 Guangzhou, China. [Zhu, Jian-guo] Guizhou Provincial People’s Hospital, Department of Urology, 550002 Guizhou, China. [Zhong, Wei-de] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. RP Zhong, Wd (reprint author), Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. EM zhongwd2009@live.cn CR Hughes C, Murphy A, Martin C et al, 2005, Molecular pathology of PCA. J Clin Path 58:673–684 Taplin M-E, Rajeshkumar B, Halabi S et al, 2003, Androgen receptor mutations in androgen-independent prostate cancer: Cancer and Leukemia Group B Study 9663. J Clin Oncol 21:2673–2678 Beinke S, Ley SC, 2004, Functions of NF-kB1 and NF-kB2 in immune cell biology. Biochem J 382:393–409 Hayden MS, Ghosh S, 2008, Shared principles in NF-kB signaling. Cell 132:344–362 Hayden MS, Ghosh S, 2004, Signaling to NF-kappaB. Genes Dev 18:2195–2224 Bonizzi G, Karin M, 2004, The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol 25:280–288 Li Q, Verma IM, 2002, NF-kappa B regulation in the immune system. Nat Rev Immunol 2:725–734 Meylan E, Dooley AL, Feldser DM, 2009, Requirement for NF-kappaB signalling in a mouse model of lung adenocarcinoma. Nature 462:104–107 Xu Y, Josson S, Fang F, 2009, RelB enhances prostate cancer growth: implications for the role of the nuclear factor-KB alternative pathway in Tumorigeni City. Cancer Res 69:3267– 3271 Lessard L, Begin L, Gleave M et al, 2005, Nuclear localisation of nuclear factor-kappaB transcription factors in prostate cancer: an immunohistochemical study. Br J Cancer 93:1019–1023 Locke JA, Guns ES, Lubik AA, 2008, Androgen levels increase by intratumoral de novo steroidogenesis during progression of castration-resistant prostate cancer. Cancer Res 68:6407–6415 D’Antonio JM, Ma C, Monzon FA et al, 2008, Longitudinal analysis of androgen deprivation of prostate cancer cells identifies pathways to androgen independence. Prostate 68:698–714 Titus MA, Schell MJ, Lih FB et al, 2005, Testosterone and dihydrotestosterone tissue levels in recurrent prostate cancer. Clin Cancer Res 11:4653–4657 Palvimo JJ, Reinikainen P, Ikonen T et al, 1996, Mutual transcriptional interference between RelA and androgen receptor. J Biol Chem 271:24151–24156 Chen CD, Sawyers CL, 2002, NF-kappa B activates prostate-specific antigen expression and is upregulated in androgen-independent prostate cancer. Mol Cell Biol 22:2862–2870 Nadiminty N, Lou W, Sun M, 2010, Aberrant activation of the androgen receptor by NF-kappaB2/p52 in prostate cancer cells. Cancer Res 70:3309–3319 Nadiminty N, Chun JY, Lou Wet al, 2008, NF-κB2/p52 enhances androgen-independent growth of human LNCaP cells via protection from apoptotic cell death and cell cycle arrest induced by androgen-deprivation. Prostate 68:1725–1733 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 873 EP 878 DI 10.1007/s12253-011-9396-5 PG 6 ER PT J AU Rouissi, K Ouerhani, S Hamrita, B Bougatef, K Marrakchi, R Cherif, M Ben Slama, RM Bouzouita, M Chebil, M Ben Ammar Elgaaied, A AF Rouissi, Kamel Ouerhani, Slah Hamrita, Bechr Bougatef, Karim Marrakchi, Raja Cherif, Mohamed Ben Slama, Riadh Mohamed Bouzouita, Mohamed Chebil, Mohamed Ben Ammar Elgaaied, Amel TI Smoking and Polymorphisms in Xenobiotic Metabolism and DNA Repair Genes are Additive Risk Factors Affecting Bladder Cancer in Northern Tunisia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; Xenobiotics; DNA repair; Predisposition ID Bladder cancer; Xenobiotics; DNA repair; Predisposition AB Cancer epidemiology has undergone marked development since the nineteen-fifties. One of the most spectacular and specific contributions was the demonstration of the massive effect of smoking and genetic polymorphisms on the occurrence of bladder cancer. The tobacco carcinogens are metabolized by various xenobiotic metabolizing enzymes, such as the super-families of Nacetyltransferases (NAT) and glutathione S-transferases (GST). DNA repair is essential to an individual’s ability to respond to damage caused by tobacco carcinogens. Alterations in DNA repair genes may affect cancer risk by influencing individual susceptibility to this environmental exposure. Polymorphisms in NAT2, GST and DNA repair genes alter the ability of these enzymes to metabolize carcinogens or to repair alterations caused by this process. We have conducted a case-control study to assess the role of smoking, slow NAT2 variants, GSTM1 and GSTT1 null, and XPC, XPD, XPG nucleotide excision-repair (NER) genotypes in bladder cancer development in North Tunisia. Taken alone, each gene unless NAT2 did not appear to be a factor affecting bladder cancer susceptibility. For the NAT2 slow acetylator genotypes, the NAT2*5/*7 diplotype was found to have a 7-fold increased risk to develop bladder cancer (OR=7.14; 95% CI: 1.30–51.41). However, in tobacco consumers, we have shown that Null GSTM1, Wild GSTT1, Slow NAT2, XPC (CC) and XPG (CC) are genetic risk factors for the disease. When combined together in susceptible individuals compared to protected individuals these risk factors give an elevated OR (OR=61). So, we have shown a strong cumulative effect of tobacco and different combinations of studied genetic risk factors which lead to a great susceptibility to bladder cancer. C1 [Rouissi, Kamel] University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 2092 Tunis, Tunisia. [Ouerhani, Slah] University of Tunis El Manar, Pasteur Institute of Tunis, Laboratory of Molecular and Cellular HaematologyTunis, Tunisia. [Hamrita, Bechr] Charles Nicole Hospital, Department of UrologyTunis, Tunisia. [Bougatef, Karim] University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 2092 Tunis, Tunisia. [Marrakchi, Raja] University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 2092 Tunis, Tunisia. [Cherif, Mohamed] Charles Nicole Hospital, Department of UrologyTunis, Tunisia. [Ben Slama, Riadh Mohamed] Charles Nicole Hospital, Department of UrologyTunis, Tunisia. [Bouzouita, Mohamed] Charles Nicole Hospital, Department of UrologyTunis, Tunisia. [Chebil, Mohamed] Charles Nicole Hospital, Department of UrologyTunis, Tunisia. [Ben Ammar Elgaaied, Amel] University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 2092 Tunis, Tunisia. RP Rouissi, K (reprint author), University of El Tunis Manar I, Faculty of Sciences of Tunis, Laboratory of Genetics, Immunology and Human Pathology, 2092 Tunis, Tunisia. EM rouissik2000@yahoo.fr CR Cohen SM, Johansson SL, 1992, Epidemiology and etiology of bladder cancer. Urol Clin North Am 19:421–428 Raunio H, Husgafvel-Pursiainen K, Anttila S et al, 1995, Diagnosis of polymorphisms in carcinogen-activating and inactivating enzymes and cancer susceptibility: a review. Gene 159:113–121 Kaprio J, 2000, Science, medicine, and the future. Genetic epidemiology. B Med J 320:1257–1259 Ford JG, Li Y, O’SullivanMMet al, 2000, Glutathione S-transferase M1 polymorphism and lung cancer risk in African-Americans. Carcinogenesis 21:1971–1975 Karagas MR, Park S, Warren A et al, 2005, Gender, smoking, glutathione- S-transferase variants and bladder cancer incidence: a population-based study. Cancer Lett 219:63–69 Abdel-Rahman SZ, Anwar WA, Abdel-Ala WE et al, 1998, GSTM1 and GSTT1 genes are potential risk modifiers for bladder cancer. Cancer Detect Prev 22:129–138 Salagovic J, Kalina I, Stubna J et al, 1998, Genetic polymorphism of glutathione S-transferase M1 and T1 as a risk factor in lung and bladder cancers. Neoplasma 45:312–317 Hengstler JG, Kett A, Arand M et al, 1998, Glutathione Stransferase T1 and M1 gene defects in ovarian carcinoma. Cancer Lett 130:43–48 Katoh T, Inatomi H, Kim H et al, 1998, Effects of glutathione Stransferase, GST, M1 and GSTT1 genotypes on urothelial cancer risk. Cancer Lett 132:147–152 Badawi AF, Hirvonen A, Bell DA et al, 1995, Role of aromatic amine acetyltransferases, NAT1 and NAT2, in carcinogen-DNA adducts formation in human urinary bladder. Cancer Res 55:5230–5237 Kloth MT, Gee RL, Messing EM et al, 1994, Expression of N acetyltransferase, NAT, in cultured human uroepithelial cells. Carcinogenesis 15:2781–2787 Blum M, Grant DM, McBride W et al, 1990, Human arylamine N-acetyltransferase genes: isolation, chromosomal localization, and functional expression. DNA Cell Biol 9:193–203 Brockmoller J, Cascorbi I, Kerb R et al, 1996, Combined analysis of inherited polymorphisms in arylamine N-acetyltransferase 2, glutathione S-transferases M1 and T1, microsomal epoxide hydrolase and cytochrome P450 enzymes as modulators of bladder cancer risk. Cancer Res 56:3915–3925 Risch A, Wallace DMA, Bathers S et al, 1995, Slow N-acetylation genotype is a susceptibility factor in occupational and smoking related bladder cancer. Hum Mol Genet 4:231–236 Probst-Hensch NM, Bell DA, Watson MA et al, 2000, Nacetyltransferase 2 phenotype but not NAT1*10 genotype affects aminobiphenylhemoglobin adduct levels. Cancer Epidemiol Biomarkers Prev 9:619–623 Yu MC, Skipper PL, Taghizadeh K et al, 1994, Acetylator phenotype, aminobiphenylhemoglobin adduct levels, and bladder cancer risk in white, black, and Asian men in Los Angeles, California. J Nat Cancer Inst 86:712–716 Airoldi L, Orsi F, Magagnotti C et al, 2002, Determinants of 4- aminobiphenyl-DNA adducts in bladder cancer biopsies. Carcinogenesis 23:861–866 Hao GY, Zhang WD, Chen YH et al, 2004, Relationship between genetic polymorphism of NAT2 and susceptibility to urinary bladder cancer. Zhonghua Zhong Liu Za Zhi 26:283–286 Wu XF, Dzenis YA, 2006, Guided self-assembly diblock copolymer thin films on chemically patterned substrartes. J Chem Phys 125:174–1777 Chen CH, Shun CT, Huang KH et al, 2007, Stopping smoking might reduce tumour recurrence in non-muscle invasive bladder cancer. BJU Int 100:281–286 Garcia-Closas M, Malats N, Real FX et al, 2006, Genetic variation in the nucleotide-excision repair pathway and bladder cancer risk. Cancer Epidemiol Biomarkers Prev 15:536–542 Sanyal S, De Verdier PJ, Steineck G et al, 2007, Polymorphisms in XPD, XPC and the risk of death in patients with urinary bladder neoplasms. Acta Oncol 46:31–41 Zhu Y, Yang H, Chen Q et al, 2008, Modulation of DNA damage/ DNA repair capacity by XPC polymorphisms. DNA Repair 7:141–148 Dworaczek H, Xiao W, 2007, Xeroderma pigmentosum: a glimpse into nucleotide-excision repair, genetic instability, and cancer. Crit Rev Oncog 13:159–177 Shen M, Hung RJ, Brennan P et al, 2003, Polymorphisms of the DNA repair genes XRCC1, XRCC3, XPD, interaction with environmental exposures, and bladder cancer risk in a casecontrol study in northern Italy. Cancer Epidemiol Biomarkers Prev 12:1234–1240 Sellami A, Jlidi R, HsairiMet al, 2000, Registre du cancer du Sud Tunisie 1997. Hopital Habib Bourguiba 2:32–35 Sambrook J, Fritsch EF, Maniatis T, 1989, Molecular cloning: a laboratory manual, 2nd edn. Cold Spring Harbor Laboratory, Cold Spring Harbor Arand M, Muhlbaur R, Hengstler J et al, 1996, A multiplex polymerase chain reaction protocol for the simultaneous analysis of the glutathione S transferase GSTM1 and GSTT1 polymorphisms. Annal Biochem 236:384–386 Hsieh FI, Pu YS, Chern HD et al, 1999, Genetic polymorphisms of N-acetyltransferase 1 and 2 and risk of cigarette smokingrelated bladder cancer. Br J Cancer 81:537–541 Fukino K, Sasaki Y, Hirai S et al, 2008, Effect of Nacetyltransferase 2(NAT2), CYP2E1 and Glutathione-Stransferase, GST, genotypes on the serum Concentrations of isoniazid and metabolites in tuberculosis patients. J Toxicol Sci 33:187–195 Vastis KP, Weber WW, Bell DA et al, 1995, Nomenclature for Nacetyltransferases. Pharmacogenetics 5:1–17 Sanyal S, Festa F, Sakano S et al, 2004, Polymorphisms in DNA repair and metabolic genes in bladder cancer. Carcinogenesis 25:729–734 De Ruyck K, Szaumkessel M, De Rudder I et al, 2007, Polymorphisms in base-excision repair and nucleotide-excision repair genes in relation to lung cancer risk. Mutat Res 631:101–110 O’Gorman TW, Woolson RF, 1993, The effect of category choice on the odds ratio and several measures of association in casecontrol studies. Commun Stat 22:1157–1171 Ouerhani S, Tebourski F, Slama MR et al, 2006, The role of glutathione transferases M1 and T1 in individual susceptibility to bladder cancer in a Tunisian population. Ann Hum Biol 33:529–535 Garte S, Gaspari L, Alexandrie AK et al, 2001, Metabolic gene polymorphism frequencies in control populations. Cancer Epidemiol Biomarkers Prev 10:1239–1248 Kempkes M, Golka K, Reich S et al, 1996, Glutathione transferase GSTM1 and GSTT1 null genotypes as potential risk factors for urothelial cancer of the bladder. Arch Toxicol 71:123–126 Lee SJ, Cho SH, Park SK et al, 2002, Combined effect of glutathione S-transferase M1 and T1 genotypes on bladder cancer risk. Cancer Lett 77:173–179 Kim WJ, Kim H, Kim CH et al, 2002, GSTT1-null genotype is a protective factor against bladder cancer. Urology 60:913–918 Hein DW, 2006, N-acetyltransferase 2 genetic polymorphism: effects of carcinogen and haplotype on urinary bladder cancer risk. Oncogene 25:1649–1658 Khedhaier A, Hassen E, Bouaouina N et al, 2008, Implication of xenobiotic metabolizing enzyme gene, CYP2E1, CYP2C19, CYP2D6, mEH and NAT2, polymorphisms in breast carcinoma. BMC Cancer 28:109–121 Bell DA, Taylor JA, Butler MA et al, 1993, Genotype/phenotype discordance for human arylamine N-acetyltransferase, NAT2, reveals a new slow-acetylator allele common in African- Americans. Carcinogenesis 14:1689–1692 El Desoky ES, Abdel Salam YM, Salama RH et al, 2005, NAT2*5/*5 genotype, 341T>C, is a potential risk factor for schistosomiasis- associated bladder cancer in Egyptians. Ther Drug Monit 27:297–304 Fretland AJ, Doll MA, Leff MA et al, 2001, Functional characterization of nucleotide polymorphisms in the coding region of N-acetyltransferase 1. Pharmacogenetics 11:511–520 Zang Y, Zhao S, Doll MA et al, 2004, The T341C, Ile114Thr, polymorphism of N-acetyltransferase 2 yields slow acetylator phenotype by enhanced protein degradation. Pharmacogenetics 14:717–723 Garcia-Closas M, Malats N, Silverman D et al, 2005, NAT2 slow acetylation, GSTM1 null genotype, and risk of bladder cancer: results from the Spanish Bladder Cancer Study and meta-analyses. Lancet 366:649–659 Lin J, Swan GE, Shields PG et al, 2007, Mutagen sensitivity and genetic variants in nucleotide-excision repair pathway: genotypephenotype correlation. Cancer Epidemiol Biomarkers Prev 16:2065–2071 Fontana L, Bosviel R, Delort L, 2008, DNA repair gene ERCC2, XPC, XRCC1, XRCC3 polymorphisms and associations with bladder cancer risk in a French cohort. Anticancer Res 28:1853– 1856 Rouissi K, Ouerhani S, Oliveira E et al, 2009, Polymorphisms in one-carbon metabolism pathway genes and risk for bladder cancer in a Tunisian population. Cancer Genet Cytogenet 195:43–53 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 879 EP 886 DI 10.1007/s12253-011-9398-3 PG 8 ER PT J AU Bojkova, B Garajova, M Pec, M Kubatka, P Kajo, K Mokan, M Kassayova, M Orendas, P Kiskova, T Ahlersova, E Ahlers, I AF Bojkova, Bianka Garajova, Miroslava Pec, Martin Kubatka, Peter Kajo, Karol Mokan, Marian Kassayova, Monika Orendas, Peter Kiskova, Terezia Ahlersova, Eva Ahlers, Ivan TI Metabolic Effects of Pioglitazone in Chemically-Induced Mammary Carcinogenesis in Rats SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chemoprevention; Mammary carcinogenesis; Metabolism; Pioglitazone; Rat ID Chemoprevention; Mammary carcinogenesis; Metabolism; Pioglitazone; Rat AB In this paper, the effect of peroral antidiabetic pioglitazone, a thiazolidinedione derivate, on selected parameters of carbohydrate and lipid metabolism in Nmethyl-N-nitrosourea-induced mammary carcinogenesis in female Sprague–Dawley rats was evaluated. Pioglitazone was administered in the diet at two concentrations (10 ppm and 100 ppm), the chemoprevention was initiated 12 days before carcinogenesis induction and lasted until the termination of the experiment. The experiment was terminated 17 weeks after carcinogenesis induction, selected organs and tissues were removed and weighed and basic metabolic and hormonal parameters were determined. Pioglitazone increased glycemia (without exceeding normal values) and glycogen concentration in both liver and heart muscle without altering insulinemia and increased triacylglycerol concentration in liver, these changes were more prominent in group with higher dose. Pioglitazone also reduced corticosterone serum concentration and attenuated lipid peroxidation. Pioglitazone and other glitazones may be useful in alleviation of unfavourable metabolic changes in cancer patients. C1 [Bojkova, Bianka] P.J.Safarik University, Faculty of Science, Institute of Biology and Ecology, Moyzesova 11, 041 67 Kosice, Slovakia. [Garajova, Miroslava] P.J.Safarik University, Faculty of Science, Institute of Biology and Ecology, Moyzesova 11, 041 67 Kosice, Slovakia. [Pec, Martin] Comenius University, Jessenius Faculty of Medicine, Department of Medical Biology, Mala Hora 4, 037 54 Martin, Slovakia. [Kubatka, Peter] Comenius University, Jessenius Faculty of Medicine, Department of Medical Biology, Mala Hora 4, 037 54 Martin, Slovakia. [Kajo, Karol] Comenius University, Jessenius Faculty of Medicine, Department of Pathology, Kollarova 2, 036 01 Martin, Slovakia. [Mokan, Marian] Comenius University, Jessenius Faculty of Medicine, Clinic of Internal Medicine I., Kollarova 2, 036 01 Martin, Slovakia. [Kassayova, Monika] P.J.Safarik University, Faculty of Science, Institute of Biology and Ecology, Moyzesova 11, 041 67 Kosice, Slovakia. [Orendas, Peter] P.J.Safarik University, Faculty of Science, Institute of Biology and Ecology, Moyzesova 11, 041 67 Kosice, Slovakia. [Kiskova, Terezia] P.J.Safarik University, Faculty of Science, Institute of Biology and Ecology, Moyzesova 11, 041 67 Kosice, Slovakia. [Ahlersova, Eva] P.J.Safarik University, Faculty of Science, Institute of Biology and Ecology, Moyzesova 11, 041 67 Kosice, Slovakia. [Ahlers, Ivan] P.J.Safarik University, Faculty of Science, Institute of Biology and Ecology, Moyzesova 11, 041 67 Kosice, Slovakia. RP Bojkova, B (reprint author), P.J.Safarik University, Faculty of Science, Institute of Biology and Ecology, 041 67 Kosice, Slovakia. EM bianka.bojkova@upjs.sk CR Wolf I, Rubinek T, 2008, Diabetes mellitus and breast cancer. Front Diabetes 19:97–113 Zhou G, Myers R, Li Y et al, 2001, Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 108:1167–1174 Dowling RJO, Zakikhani M, Fantus IG et al, 2007, Metformin inhibits mammalian target of rapamycin-dependent translation in breast cancer cells. Cancer Res 67:10804–10812 Hatton JL, Yee LD, 2008, Clinical use of PPARγ ligands in cancer. PPAR Research art. no. 159415., DOI 10.1155/2008/159415 Fryer LGD, Parbu-Patel A, Carling C, 2002, The anti-diabetic drugs rosiglitazone and metformin stimulate AMP-activated protein kinase through distinct signaling pathways. J Biol Chem 277:25226–25232 Luo Z, Saha AK, Xiang X et al, 2005, AMPK, the metabolic syndrome and cancer. Trends Pharmacol Sci 26:69–76 Elstner E, Muller C, Koshizuka K et al, 1998, Ligands for peroxisome proliferator-activated receptorγ and retinoic acid receptor inhibit growth and induce apoptosis of human breast cancer cells in vitro and in BNX mice. Proc Natl Acad Sci USA 95:8806–8811 Bonofiglio D, Gabriele S, Aquila S et al, 2009, Peroxisome proliferator-activated receptor gamma activates fas ligand gene promoter inducing apoptosis in human breast cancer cells. Breast Cancer Res Tr 113:423–434 Bonofiglio D, Cione E, Qi H et al, 2009, Combined low doses of PPARγ and RXR ligands trigger an intrinsic apoptotic pathway in human breast cancer cells. Am J Pathol 175:1270–1280 Zhou J, Zhang W, Liang B et al, 2009, PPARγ activation induces autophagy in breast cancer cells. Int J Biochem Cell B 41:2334– 2342 Suh N, Wang Y, Williams CR et al, 1999, A new ligand for the peroxisome proliferator-activated receptor-γ, PPAR-γ), GW7845, inhibits rat mammary carcinogenesis. Cancer Res 59:5671–5673 Badawi AF, Eldeen MB, Liu Y et al, 2004, Inhibition of rat mammary gland carcinogenesis by simultaneous targeting of cyclooxygenase-2 and peroxisome proliferator-activated receptor γ. Cancer Res 64:1181–1189 Kocdor H, Kocdor MA, Canda T et al, 2009, Chemopreventive efficacies of rosiglitazone, fenretinide and their combination against rat mammary carcinogenesis. Clin Trans Oncol 11:243–249 Bojkova B, Garajova M, Kajo K et al, 2010, Pioglitazone in chemically induced mammary carcinogenesis in rats. Eur J Cancer Prev 19:379–384 Koro C, Barrett C, Qizilbash N, 2007, Cancer risks in thiazolidinedione users compared to other anti-diabetic agents. Pharmacoepidem Dr S 16:485–492 Monami M, Lamanna C, Marchionni N et al, 2008, Rosiglitazone and risk of cancer: A meta-analysis of randomized clinical trials. Diabetes Care 31:1455–1460 Yee LD, Williams N, Wen P et al, 2007, Pilot study of rosiglitazone therapy in women with breast cancer: effects of short-term therapy on tumour tissue and serum markers. Clin Cancer Res 13:246–252 Burstein HJ, Demetri GD, Mueller E et al, 2003, Use of the peroxisome proliferator-activated receptor, PPAR, γ ligand troglitazone as treatment for refractory breast cancer: a phase II study. Breast Cancer Res Tr 79:391–397 Reichle A, Bross K, Vogt T et al, 2004, Pioglitazone and rofecoxib combined with angiostatically scheduled trofosfamide in the treatment of far-advanced melanoma and soft tissue sarcoma. Cancer 101:2247–2256 Hau P, Kunz-Schughart L, Bogdahn U et al, 2008, Low-dose chemotherapy in combination with COX-2 inhibitors and PPARgamma agonists in recurrent high-grade gliomas—a phase II study. Oncology 73:21–25 Bartlett GR, 1959, Phosphorus assay in column chromotheraphy. J Biol Chem 234:466–468 Zlatkis A, Zak B, Boyle AJ, 1953, A new method for the direct determination of cholesterol. J Lab Clin Med 41:486–490 Roe JH, Dailey RE, 1966, Determination of glycogen with the anthrone reagent. Anal Biochem 15:245–250 Satoh K, 1978, Serum lipid peroxide in cerebrovascular disorders determined by a new colorimetric method. Clin Chim Acta 90:37–43 Guillemin R, Clayton GW, Smith JD et al, 1958, Measurement of free corticosteroids in rat plasma. Physiological validation of the method. Endocrinology 63:349–355 Lelbach A,Muzes G, Feher J, 2007, Current perspectives of catabolic mediators of cancer cachexia. Med Sci Monitor 13:168–173 Douglas RG, Shaw JHF, 1990, Metabolic effects of cancer. Br J Surg 77:246–254 Knapp ML, Al-Sheibani S, Riches PG et al, 1991, Hormonal factors associated with weight loss in patients with advanced breast cancer. Ann Clin Biochem 28:480–486 Kokoglu E, Karaarslan I, Karaarslan HM et al, 1994, Alterations of serum lipids and lipoproteins in breast cancer. Cancer Lett 82:175–178 Bojkova B, Orendas P, Garajova M et al, 2009, Metformin in chemically-induced mammary carcinogenesis in rats. Neoplasma 56:267–272 West IC, 2000, Radicals and oxidative stress in diabetes. Diabetic Med 17:171–180 Droge W, 2002, Free radicals in the physiological control of cell function. Physiol Rev 82:47–95 Rahimian R, Fakhfouri G, Rasouli MR et al, 2009, Effect of pioglitazone on sciatic nerve/reperfusion injury in rats. Pediatr Neurosurg 45:126–131 Somi MH, Hajipour B, Asl NA et al, 2009, Pioglitazone attenuates ischemia/reperfusion-induced liver injury in rats. Transplant P 41:4105–4109 Schaur RJ, Fellier H, Gleispach H, 1979, Tumour host relations. I. Increased plasma cortisol in tumour-bearing humans compared with patients with benign surgical diseases. J Cancer Res Clin 93:281–285 Feng Z, Marti A, Jehn B et al, 1995, Glucocorticoid and progesterone inhibit involution and programmed cell death in the mouse mammary gland. J Cell Biol 131:1095–1103 Schorr K, Furth PA, 2000, Induction of bcl-x(L, expression in mammary epithelial cells is glucocorticoid-dependent but not signal transducer and activator of transcription 5-dependent. Cancer Res 60:5950–5953 Verheus M, McKay JD, Kaaks R et al, 2008, Common genetic variation in the IGF-1 gene, serum IGF-1 levels and breast density. Breast Cancer Res Tr 112:109–122 Hankinson SE, Willett WC, Colditz GA et al, 1998, Circulating concentrations of insulin-like growth factor-I and risk of breast cancer. Lancet 351:1393–1396 Hankinson SE, Schernhammer ES, 2003, Insulin-like growth factor and breast cancer risk: Evidence from observation studies. Breast Dis 17:27–40 Schernhammer ES, Holly JM, Pollak MN et al, 2005, Circulating levels of insulin-like growth factors, their binding proteins, and breast cancer risk. Cancer Epidem Biomar 14:699–704 Rinaldi S, Peeters PHM, Berrino F et al, 2006, IGF-I, IGFBP-3 and breast cancer risk in women: The European prospective investigation into cancer and nutrition. Endocr Relat Cancer 13:593–605 Rollison DE, Giuliano AR, Risendal BC et al, 2010, Serum insulin-like growth factor, IGF)-1 and IGF binding protein-3 in relation to breast cancer among Hispanic and white, non-Hispanic women in the US Southwest. Breast Cancer Res Tr 121:661–669 Crown AL, Cottle K, Lightman SL et al, 2002, What is the role of the insulin-like growth factor system in the pathophysiology of cancer cachexia, and how is it regulated? Clin Endocrinol 56:723– 733 Costelli P, Muscaritoli M, Bossola M et al, 2006, IGF-1 is downregulated in experimental cancer cachexia. Am J Physiol- Reg I 291:674–683 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 887 EP 892 DI 10.1007/s12253-011-9399-2 PG 6 ER PT J AU Kravchick, S Peled, R Cytron, Sh AF Kravchick, Sergey Peled, Ronit Cytron, Shmuel TI Watchful Waiting and Active Surveillance Approach in Patients with Low Risk Localized Prostatic Cancer: An Experience of Out-Patients Clinic with 12-Year Follow-Up SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate; Cancer; Expectant management; Body mass index; TRUS-biopsy ID Prostate; Cancer; Expectant management; Body mass index; TRUS-biopsy AB In this study we evaluated the safety of expectant approach in the patients with low risk prostate cancer in the reality of community based out-patients clinics. 48 men were enrolled into the study. The inclusion criteria were age ranged from 60 to 75 years and the Epstein criteria for low risk prostate cancer. Patients were managed expectantly while curative treatment was offered when indicated. Initial and final Charlson comorbidity index (CCI) and BMI were assessed for all men. Patients’ median follow-up was 81.1±29.1 years. During this study 41.7% of the patients chose active forms of treatment. Cancer was found in 20.8% (n-10) of our patients. Two first sessions of re-biopsy diagnosed 92% of T1c upgrading. Six men with CCI ≥2 died from concomitant disease and no one died from PCa. Significant correlation was found between BMI and final CCI ≥2 (p-0.001). Expectant approach can be considered as self alternative to active treatment model in selected group of patients with well differentiated PCa, however 20.8% of these patients are still at risk of having aggressive form of cancer. Expectant approach is particular beneficial for the patients with CCI 1–2 and high BMI. C1 [Kravchick, Sergey] Barzialy Medical Center, Urology DepartmentAshkelon, Israel. [Peled, Ronit] Ben-Gurion University of the Negev, Faculty of Health Sciences, Department of Health Systems ManagementBeer-Sheva, Israel. [Cytron, Shmuel] Barzialy Medical Center, Urology DepartmentAshkelon, Israel. RP Kravchick, S (reprint author), Barzialy Medical Center, Urology Department, Ashkelon, Israel. EM cambell@netvision.net.il;sergeyk@barzi.health.gov.il CR Schroder FH, Hugosson J, Roobol MJ et al, 2009, Screening and prostate-cancer mortality in a randomized European study. N Engl J Med 360:1320–1328 Epstein JI, Walsh PC, Carmichael M, Brendler CB, 1994, Pathologic and clinical findings to predict tumor extent of nonpalpable, stage T1c, prostate cancer. JAMA 271(5):368–374 Carter HB, Kettermann A, Warlick C, Metter EJ, Landis P, Walsh PC, Epstein JI, 2007, Expectant management of prostate cancer with curative intent: an update of the Johns Hopkins experience. J Urol 178(6):2359–2364, discussion 2364–5 Barocas DA, Cowan JE, Smith JA Jr, Carroll PR, 2008, What percentage of patients with newly diagnosed carcinoma of the prostate are candidates for surveillance? An analysis of the CaPSURE database. J Urol 180(4):1330–1334 Charlson ME, Pompei P, Ales KL, MacKenzie CR, 1987, A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis 40, 5):373–383 Nash PH, Bruce JE, Indudhara R, Shinohara K, 1996, Transrectal ultrasound guided prostatic nerve blockade eases systematic needle biopsy of the prostate. J Urol 155:607–609 Jeldres C, Suardi N, Walz J, Hutterer GC, Ahyai S, Lattouf JB, Haese A, Graefen M, Erbersdobler A, Heinzer H, Huland H, Karakiewicz PI, 2008, Validation of the contemporary epstein criteria for insignificant prostate cancer in European men. Eur Urol 54(6):1306–1313 Bastian PJ, Mangold LA, Epstein JI, Partin AW(2004, Characteristics of insignificant clinical T1c prostate tumors. A contemporary analysis. Cancer 101(9):2001–2005 Lee MC, Dong F, Stephenson AJ, Jones JS, Magi-Galluzzi C, Klein EA, 2010, The Epstein criteria predict for organconfined but not insignificant disease and a high likelihood of cure at radical prostatectomy. Eur Urol 58(1):90–95, Epub 2009 Oct 23 Klotz L, 2004, Active surveillance with selective delayed intervention: using natural history to guide treatment in good risk prostate cancer. J Urol 172(5 Pt 2):S48–S50, discussion S50-1 Roemeling S, Roobol MJ, Postma R, Gosselaar C, van der Kwast TH, Bangma CH, Schroder FH, 2006, Management and survival of screen-detected prostate cancer patients who might have been suitable for active surveillance. Eur Urol 50(3):475–482, Epub 2006 May 3 Al Otaibi M, Ross P, Fahmy N, Jeyaganth S, Trottier H, Sircar K, Begin LR, Souhami L, Kassouf W, Aprikian A, Tanguay S, 2008, Role of repeated biopsy of the prostate in predicting disease progression in patients with prostate cancer on active surveillance. Cancer 113(2):286–292 Draisma G, Boer R, Otto SJ, van der Cruijsen IW, Damhuis RA, Schroder FH, de Koning HJ, 2003, Lead times and overdetection due to prostate-specific antigen screening: estimates from the European Randomized Study of Screening for Prostate Cancer. J Natl Cancer Inst 95(12):868–878 Steyerberg EW, Roobol MJ, Kattan MW, van der Kwast TH, de Koning HJ, Schroder FH, 2007, Prediction of indolent prostate cancer: validation and updating of a prognostic nomogram. J Urol 177(1):107–112, discussion 112 Albertsen PC, Hanley JA, Fine J, 2005, 20-year outcomes following conservative management of clinically localized prostate cancer. JAMA 293(17):2095–2101 Dall’Era MA, Cooperberg MR, Chan JM, Davies BJ, Albertsen PC, Klotz LH, Warlick CA, Holmberg L, Bailey DE Jr, Wallace ME, Kantoff PW, Carroll PR, 2008, Active surveillance for earlystage prostate cancer: review of the current literature. Cancer 112, 8):1650–1659 Stav K, Leibovici D, Sandbank J, Lindner A, Zisman A, 2008, Saturation prostate biopsy in high risk patients after multiple previous negative biopsies. Urology 71(3):399–403 Djavan B, Waldert M, Zlotta A, Dobronski P, Seitz C, Remzi M, Borkowski A, Schulman C, 2001, Marberger M Safety and morbidity of first and repeat transrectal ultrasound guided prostate needle biopsies: results of a prospective European prostate cancer detection study. J Urol 166(3):856–860 Post PN, Kil PJ, Hendrikx AJ, Janssen-Heijnen ML, Crommelin MA, Coebergh JW, 1999, Comorbidity in patients with prostate cancer and its relevance to treatment choice. BJU Int 84(6):652–656 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 893 EP 897 DI 10.1007/s12253-011-9400-0 PG 5 ER PT J AU Faccion, SR Ferreira, MR Grabois, FM Fonseca, CTh de Oliveira, AJ Maia, CR AF Faccion, Soares Roberta Ferreira, Moreira Regina Grabois, Fornaciari Marilia Fonseca, Carvalho Theresinha de Oliveira, Antonio Jose Maia, Ciuvalschi Raquel TI Lack of Prognostic Significance of Survivin in Pediatric Medulloblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Survivin; Childhood; Medulloblastoma; Prognostic factor; Immunohistochemistry ID Survivin; Childhood; Medulloblastoma; Prognostic factor; Immunohistochemistry AB Medulloblastoma (MDB) is the most common malignant cerebellar tumor in children. Because of the significant rate of mortality and treatment-related morbidity, the identification of prognostic factors could lead to a more accurate selection of patients who can benefit from a less aggressive therapy and improve risk stratification. Survivin is an inhibitor of apoptosis protein (IAP), the expression of which has been associated with worse prognosis in MDB. However, both of its subcellular localizations may contribute to tumor progression, and ultimately, survivin subcellular localization prognostic value depends on tumor type biological features. The goal of this study was to analyze these survivin features in the pediatric MDB tumor samples and its impact on clinical outcome. Survivin expression and subcellular localization were accessed by immunohistochemistry in a series of 41 tumor samples. Kaplan-Meier survival curves were compared using the log-rank test. Survivin expression ranged from completely absent to fully present in a notably higher pattern of nuclear localization than cytoplasmic (19 of 41 versus 4 of 41, respectively).However, survivin expression and subcellular localization were not associated with five-year overall survival or metastasis status at diagnosis, which was the only statistically significant prognostic factor in our series (p=0.008). Taken together, our results suggest that survivin expression should be further studied in large, multicenter series to determine its accurate impact on prognosis and pathobiology of pediatric MDB. C1 [Faccion, Soares Roberta] Instituto Nacional de Cancer (INCA), Programa de Pesquisa em Hemato-Oncologia Molecular, Coordenacao Geral Tecnico-Cientifica, Praca da Cruz Vermelha, 23, 6o andar, Ala B, Centro, 20230–130 Rio de Janeiro, RJ, Brazil. [Ferreira, Moreira Regina] INCA, HCI, Coordenacao Geral de Gestao Assistencial, Servico de Pediatria, 20230-130 Rio de Janeiro, RJ, Brazil. [Grabois, Fornaciari Marilia] INCA, HCI, Coordenacao Geral de Gestao Assistencial, Servico de Pediatria, 20230-130 Rio de Janeiro, RJ, Brazil. [Fonseca, Carvalho Theresinha] Instituto Nacional de Cancer, Divisao de Patologia, 20220-400 Rio de Janeiro, RJ, Brazil. [de Oliveira, Antonio Jose] INCA, HCI, Coordenacao Geral de Gestao Assistencial, Servico de Neurocirurgia, 20230-130 Rio de Janeiro, RJ, Brazil. [Maia, Ciuvalschi Raquel] Instituto Nacional de Cancer (INCA), Programa de Pesquisa em Hemato-Oncologia Molecular, Coordenacao Geral Tecnico-Cientifica, Praca da Cruz Vermelha, 23, 6o andar, Ala B, Centro, 20230–130 Rio de Janeiro, RJ, Brazil. RP Maia, CR (reprint author), Instituto Nacional de Cancer (INCA), Programa de Pesquisa em Hemato-Oncologia Molecular, Coordenacao Geral Tecnico-Cientifica, 20230–130 Rio de Janeiro, Brazil. EM rcmaia@inca.gov.br CR Rossi A, Caracciolo V, Russo G et al, 2008, Medulloblastoma: from molecular pathology to therapy. Clin Cancer Res 14:971– 976 Wlodarski PK, Jozwiak J, 2008, Therapeutic targets for medulloblastoma. Expert Opin Ther Targets 12:449–461 Polkinghorn WR, Tarbell NJ, 2007, Medulloblastoma: tumorigenesis, current clinical paradigm, and efforts to improve risk stratification. Nat Clin Pract Oncol 4:295–304 Crawford JR, MacDonald TJ, Packer RJ, 2007, Medulloblastoma in childhood: new biological advances. Lancet Neurol 6:1073– 1085 Packer RJ, 2008, Childhood brain tumors: accomplishments and ongoing challenges. J Child Neurol 23:1122–1127 Gilbertson RJ, Gajjar A, 2005, Molecular biology of medulloblastoma: will it ever make a difference to clinical management? J Neurooncol 75:273–278 Packer RJ, 2005, Progress and challenges in childhood brain tumors. J Neurooncol 75:239–242 Augello C, Caruso L, Maggioni M et al, 2009, Inhibitors of apoptosis proteins, IAPs, expression and their prognostic significance in hepatocellular carcinoma. BMC Cancer 9:125 Mizutani Y, Nakanishi H, Li YN et al, 2007, Overexpression of XIAP expression in renal cell carcinoma predicts a worse prognosis. Int J Oncol 30:919–925 Schlette EJ, Medeiros LJ, Goy A et al, 2004, Survivin expression predicts poorer prognosis in anaplastic large-cell lymphoma. J Clin Oncol 22:1682–1688 Shibata T, Noguchi T, Takeno S et al, 2008, Disturbed XIAP and XAF1 expression balance is an independent prognostic factor in gastric adenocarcinomas. Ann Surg Oncol 15:3579– 3587 Sung KW, Choi J, Hwang YK et al, 2009, Overexpression of Xlinked inhibitor of apoptosis protein, XIAP, is an independent unfavorable prognostic factor in childhood de novo acute myeloid leukemia. J Korean Med Sci 24:605–613 Katoh M, Wilmotte R, Belkouch MC et al, 2003, Survivin in brain tumors: an attractive target for immunotherapy. J Neurooncol 64:71–76 Pennati M, Folini M, Zaffaroni N, 2007, Targeting survivin in cancer therapy: fulfilled promises and open questions. Carcinogenesis 28:1133–1139 Capalbo G, Rodel C, Stauber RH et al, 2007, The role of survivin for radiation therapy. Prognostic and predictive factor and therapeutic target. Strahlenther Onkol 183:593–599 Ambrosini G, Adida C, Altieri DC, 1997, A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 3:917–921 Tamm I, Wang Y, Sausville E et al, 1998, IAP-family protein survivin inhibits caspase activity and apoptosis induced by Fas, CD95), Bax, caspases, and anticancer drugs. Cancer Res 58:5315–5320 Li F, Ambrosini G, Chu EY et al, 1998, Control of apoptosis and mitotic spindle checkpoint by survivin. Nature 396:580–584 Stauber RH, Mann W, Knauer SK, 2007, Nuclear and cytoplasmic survivin: molecular mechanism, prognostic, and therapeutic potential. Cancer Res 67:5999–6002 Shirai K, Suzuki Y, Oka K et al, 2009, Nuclear survivin expression predicts poorer prognosis in glioblastoma. J Neurooncol 91:353–358 Saito T, Arifin MT, Hama S et al, 2007, Survivin subcellular localization in high-grade astrocytomas: simultaneous expression in both nucleus and cytoplasm is negative prognostic marker. J Neurooncol 82:193–198 Haberler C, Slavc I, Czech T et al, 2006, Histopathological prognostic factors in medulloblastoma: high expression of survivin is related to unfavourable outcome. Eur J Cancer 42:2996–3003 Sasaki T, Lopes MB, Hankins GR et al, 2002, Expression of survivin, an inhibitor of apoptosis protein, in tumors of the nervous system. Acta Neuropathol 104:105–109 Pizem J, Cort A, Zadravec-Zaletel L et al, 2005, Survivin is a negative prognostic marker in medulloblastoma. Neuropathol Appl Neurobiol 31:422–428 Fangusaro JR, Jiang Y, Holloway MP et al, 2005, Survivin, Survivin-2B, and Survivin-deItaEx3 expression in medulloblastoma: biologic markers of tumour morphology and clinical outcome. Br J Cancer 92:359–365 Li XN, Shu Q, Su JM et al, 2007, Differential expression of survivin splice isoforms in medulloblastomas. Neuropathol Appl Neurobiol 33:67–76 Bodey B, Bodey V, Siegel SE, et al., 2004, Survivin expression in childhood medulloblastomas: a possible diagnostic and prognostic marker. In vivo, Athens, Greece, 18: pp 713–718 Packer RJ, Goldwein J, Nicholson HS et al, 1999, Treatment of children with medulloblastomas with reduced-dose craniospinal radiation therapy and adjuvant chemotherapy: a children's cancer group study. J Clin Oncol 17:2127–2136 Louis DN, Ohgaki H, Wiestler OD et al, 2007, The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114:97–109 de Faria GP, de Oliveira JA, de Oliveira JG et al, 2008, Differences in the expression pattern of P-glycoprotein and MRP1 in low-grade and high-grade gliomas. Cancer Invest 26:883–889 Mita AC, Mita MM, Nawrocki ST et al, 2008, Survivin: key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin Cancer Res 14:5000–5005 Honda R, Korner R, Nigg EA, 2003, Exploring the functional interactions between Aurora B, INCENP, and survivin in mitosis. Mol Biol Cell 14:3325–3341 Satoh T, Okamoto I, Miyazaki M et al, 2009, Phase I study of YM155, a novel survivin suppressant, in patients with advanced solid tumors. Clin Cancer Res 15:3872–3880 Tolcher AW, Mita A, Lewis LD et al, 2008, Phase I and pharmacokinetic study of YM155, a small-molecule inhibitor of survivin. J Clin Oncol 26:5198–5203 Fisher PG, Burger PC, Eberhart CG, 2004, Biologic risk stratification of medulloblastoma: the real time is now. J Clin Oncol 22:971–974 Brandes AA, Paris MK, 2004, Review of the prognostic factors in medulloblastoma of children and adults. Crit Rev Oncol Hematol 50:121–128 Sarkar C, Pramanik P, Karak AK et al, 2002, Are childhood and adult medulloblastomas different? A comparative study of clinicopathological features, proliferation index and apoptotic index. J Neurooncol 59:49–61 Korshunov A, Remke M, Werft Wet al, 2010, Adult and pediatric medulloblastomas are genetically distinct and require different algorithms for molecular risk stratification. J Clin Oncol 28:3054– 3060 Grossi F, Spizzo R, Bordo D et al, 2010, Prognostic stratification of stage IIIA pN2 non-small cell lung cancer by hierarchical clustering analysis of tissue microarray immunostaining data: an Alpe Adria Thoracic Oncology Multidisciplinary Group study, ATOM 014). J Thorac Oncol 5:1354–1360 Paydas S, Ergin M, Seydaoglu G et al, 2009, Prognostic [corrected] significance of angiogenic/lymphangiogenic, antiapoptotic, inflammatory and viral factors in 88 cases with diffuse large B cell lymphoma and review of the literature. Leuk Res 33:1627–1635 Lin CK, Chao TK, Yu CP et al, 2009, The expression of six biomarkers in the four most common ovarian cancers: correlation with clinicopathological parameters. Apmis 117:162–175 Dogu GG, Ozkan M, Ozturk F et al, 2010, Triple-negative breast cancer: immunohistochemical correlation with basaloid markers and prognostic value of survivin. Med Oncol, Northwood, London, England, 27:34–39 Mitrovic Z, Ilic I, Aurer I, et al., 2011, Prognostic significance of survivin and caspase-3 immunohistochemical expression in patients with diffuse large B-cell lymphoma treated with rituximab and CHOP. Pathol Oncol Res Ellison D, 2002, Classifying the medulloblastoma: insights from morphology and molecular genetics. Neuropathol Appl Neurobiol 28:257–282 Hoffman WH, Biade S, Zilfou JT et al, 2002, Transcriptional repression of the anti-apoptotic survivin gene by wild type p53. J Biol Chem 277:3247–3257 Mirza A, McGuirk M, Hockenberry TN et al, 2002, Human survivin is negatively regulated by wild-type p53 and participates in p53-dependent apoptotic pathway. Oncogene 21:2613–2622 Li F, Altieri DC, 1999, Transcriptional analysis of human survivin gene expression. Biochem J 344(Pt 2):305–311 Sah NK, Khan Z, Khan GJ et al, 2006, Structural, functional and therapeutic biology of survivin. Cancer Lett 244:164–171 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 899 EP 908 DI 10.1007/s12253-011-9401-z PG 10 ER PT J AU Li, L Wang, YY Zhao, ZSh Ma, J AF Li, Li Wang, Yuan-Yu Zhao, Zhong-Sheng Ma, Jie TI Ezrin is Associated with Gastric Cancer Progression and Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric carcinoma; Ezrin; Progression; Prognosis ID Gastric carcinoma; Ezrin; Progression; Prognosis AB To investigat the clinical significance of Ezrin in the development and progression of gastric cancer. Immunohistochemistry was employed to analyze Ezrin expression in 436 clinicopathologically characterized gastric cancer cases. Ezrin protein levels were up-regulated in gastric cancer lesions compared with adjacent noncancerous tissues. Positive expression of Ezrin correlated with age, size of tumor, location of tumor, depth of invasion, vessel invasion, lymph node and distant metastasis and TNM stage. In stages I, II and III, the 5 year survival rate of patients with a high expression of Ezrin was significantly lower than those in patients with low expression. In stage IV, Ezrin expression did not correlate with the 5 year survival rate. Further multivariate analysis suggested that the depth of invasion, lymph node and distant metastasis, TNM stage, and up-regulation of Ezrin were independent prognostic indicators for the disease. Expression of Ezrin in gastric cancer is significantly associated with lymph node and distant metastasis, and poor prognosis. Ezrin protein could be useful markers to predict tumor progression and prognosis. C1 [Li, Li] Zhejiang Provincial People’s Hospital, Department of Pathology, 310014 Hangzhou, China. [Wang, Yuan-Yu] Zhejiang Provincial People’s Hospital, Gastrointestinal Surgery, 310014 Hangzhou, China. [Zhao, Zhong-Sheng] Zhejiang Provincial People’s Hospital, Department of Pathology, 310014 Hangzhou, China. [Ma, Jie] Zhejiang Provincial People’s Hospital, Department of Pathology, 310014 Hangzhou, China. RP Zhao, ZSh (reprint author), Zhejiang Provincial People’s Hospital, Department of Pathology, 310014 Hangzhou, China. EM zhaozhongsheng50@126.com CR Zhan W-H, Han F-H, 2008, Surgical therapy of gastric cancer in china. J Pract Oncol 23:91–93 Bretscher A, Edwards K, Fehonv RG, 2002, ERM proteins and merlin: integrators at the cell cortex. Nat Rev Mol Cell Biol 3:586–599 Gautreau A, Louvard D, Arpin M, 2002, ERM proteins and NF2 tumor suppressor: the Yin and Yang of cortical actin or ganization and cell growth signaling. Curr Opin Cell Biol 14:104–109 Saotome I, Curto M, McClatchey AI, 2004, Ezrin is essential for epithelial organization and villus morphogenesis in the developing intestine. Dev Cell 6:855–864 Srivastava J, Elliott BE, Louvard D, Arpin M, 2005, Srcdependent ezrin phosphorylation in adhesion-mediated signaling. Mol Biol Cell 16:1481–1490 Wei YC, Li CF, Yu SC, Chou FF, Fang FM, Eng HL, Uen YH, Tian YF, Wu JM, Li SH, Huang WW, Li WM, Huang HY, 2009, Ezrin overexpression in gastrointestinal stromal tumors: an independent adverse prognosticator associated with the nongastric location. Mod Pathol 22(10):1351–1360 Zhai JW, Yang XG, Yang FS, Hu JG, Hua WX, 2010, Expression and clinical significance of Ezrin and E-cadherin in esophageal squamous cell carcinoma. Chin J Cancer 29(3):317–320 Elzagheid A, Korkeila E, Bendardaf R, Buhmeida A, Heikkila S, Vaheri A, Syrjanen K, Pyrhonen S, Carpen O, 2008, Intense cytoplasmic ezrin immunoreactivity predicts poor survival in colorectal cancer. Hum Pathol 39(12):1737–1743 Valdman A, Fang X, Pang ST, Nilsson B, Ekman P, Egevad L, 2005, Ezrin expression in prostate cancer and benign prostatic tissue. Eur Urol 48(5):852–857 Musial J, Sporny S, Nowicki A, 2007, Prognostic significance of E-cadherin and ezrin immunohistochemical expression in prostate cancer. Pol J Pathol 58(4):235–243 Bal N, Yildirim S, Nursal TZ, Bolat F, Kayaselcuk F, 2007, Association of ezrin expression in intestinal and diffuse gastric carcinoma with clinicopathological parameters and tumor type. World J Gastroenterol 13(27):3726–3729 Lee HS, Lee HK, Kim HS et al, 2003, Tumour suppressor gene expression correlates with gastric cancer prognosis. J Pathol 200:39–46 Zhang D, Salto-Tellez M, Putti TC et al, 2003, Reliability of tissue microarrays in detecting protein expression and gene amplification in breast cancer. Mod Pathol 16:79–84 Lee HS, Cho SB, Lee HE et al, 2007, Protein expression profiling and molecular classification of gastric cancer by the tissue array method. Clin Cancer Res 13(14):4154–4163 Song LB, Liao WT, Mai HQ et al, 2006, The clinical significance of twist expression in nasopharyngeal carcinoma. Cancer Lett 242:258–265 Fukuoka J, Fujii T, Shih JH et al, 2004, Chromatin remodeling factors and BRM/BRG1 expression as prognostic indicators in non-small cell lung cancer. Clin Cancer Res 10:4314–4324 Hundahl SA, Phillips JL, Menck HR, 2000, The National Cancer Data Base Report on poor survival of U.S. gastric carcinoma patients treated with gastrectomy: Fifth edition American Joint Committee on cancer staging, proximal disease, and the ‘different disease’ hypothesis. Cancer 88:921–932 Wang HJ, Zhu JS, Zhang Q, Guo H, Dai YH, Xiong XP, 2009, RNAi-mediated silencing of ezrin gene reverses malignant behavior of human gastric cancer cell line SGC-7901. J Dig Dis 10(4):258–264 Meng Y, Lu Z, Yu S, Zhang Q, Ma Y, Chen J, 2010, Ezrin promotes invasion and metastasis of pancreatic cancer cells. J Transl Med 8:61 Chuan YC, Iglesias-Gato D, Fernandez-Perez L, Cedazo-Minguez A, Pang ST, Norstedt G, Pousette A, Flores-Morales A, 2010, Ezrin mediates c-Myc actions in prostate cancer cell invasion. Oncogene 29(10):1531–1542 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 909 EP 915 DI 10.1007/s12253-011-9402-y PG 7 ER PT J AU Cerne, JZ Frkovic-Grazio, S Gersak, K AF Cerne, Jasmina-Ziva Frkovic-Grazio, Snjezana Gersak, Ksenija TI Breast Tumor Characteristics in Hormone Replacement Therapy Users SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Breast tumor characteristics; HRT use; Mammography ID Breast cancer; Breast tumor characteristics; HRT use; Mammography AB The aim of this study was to further elucidate the influence of HRT use, regarding duration, regimen and route of administration, on breast tumor characteristics. We evaluated the associations between HRT use and breast tumor characteristics in 530 postmenopausal women diagnosed with invasive breast cancer. Detailed information on HRT use and mammographic attendance were collected through a postal questionnaire. Adjusted odds ratios and 95% confidence intervals were calculated using logistic regression. Tumors in HRT users were significantly smaller, more often of ductal histologic type and with lower grade and lower mitotic index compared to tumors in nonusers. Tumor characteristics did not vary significantly by HRT duration, regimen and route of administration, except for mitotic index, which was more often of score 2 in long-term users, and of score 3 in short-term users. Higher mammographic surveillance among HRT users did not explain our results. We conclude that tumors in HRT users have a more favorable prognostic profile regardless of duration, regimen and route of administration. These effects seem to be due to the influence of HRT on preexisting tumors causing their greater differentiation rather than earlier detection due to mammographic surveillance. C1 [Cerne, Jasmina-Ziva] University Medical Centre Ljubljana, Department of Obstetrics and Gynecology, Slajmerjeva 3, 1000 Ljubljana, Slovenia. [Frkovic-Grazio, Snjezana] Institute of Oncology, Department of Pathology, Zaloska 2, 1000 Ljubljana, Slovenia. [Gersak, Ksenija] University Medical Centre Ljubljana, Department of Obstetrics and Gynecology, Slajmerjeva 3, 1000 Ljubljana, Slovenia. RP Gersak, K (reprint author), University Medical Centre Ljubljana, Department of Obstetrics and Gynecology, 1000 Ljubljana, Slovenia. EM ksenija.gersak@mf.uni-lj.si CR Rossouw JE, Anderson GL, Prentice RL et al, 2002, Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. JAMA 288:321–333 Collaborative Group on Hormonal Factors in Breast Cancer, 1997, Breast cancer, hormone replacement therapy: collaborative reanalysis of data from 51 epidemiology studies of 52705 women with breast cancer and 108411 women without breast cancer. Lancet 350:1047–1059 Pines A, Sturdee DW, Birkhauser MH et al, 2007, IMS updated recommendations on postmenopausal hormone therapy. Climacteric 10:181–194 Chlebowski RT, Hendrix SL, Langer RD et al, 2003, Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the Women’s Health Initiative Randomized Trial. JAMA 289:3243–3253 Flesch-Janys D, Slanger T, Mutschelknauss E et al, 2008, Risk of different histological types of postmenopausal breast cancer by type and regimen of menopausal hormone therapy. Int J Cancer 123:933–941 Reeves GK, Beral V, Green J et al, 2006, Hormonal therapy for menopause and breast-cancer risk by histological type: a cohort study and meta-analysis. Lancet Oncol 7:910–918 Biglia N, Sgro L, Defabiani E et al, 2005, The influence of hormone replacement therapy on the pathology of breast cancer. Eur J Surg Oncol 31:467–472 Bonnier P, Bessenay F, Sasco AJ et al, 1998, Impact of menopausal hormone replacement therapy on clinical and laboratory characteristics of breast cancer. Int J Cancer 79:278–282 Cheek J, Lacy J, Toth-Fejel S et al, 2002, The impact of hormone replacement therapy on the detection and stage of breast cancer. Arch Surg 137:1015–1019 Holli K, Isola J, Cuzick J, 1998, Low biologic aggressiveness in breast cancer in women using hormone replacement therapy. J Clin Oncol 16:3115–3120 Chen WY, Hankinson SE, Schnitt SJ et al, 2004, Association of hormone replacement therapy to estrogen and progesterone receptor status in invasive breast carcinoma. Cancer 101:1490– 1500 Stahlberg C, Pedersen A, Andersen Z et al, 2004, Breast cancer with different prognostic characteristics developing in Danish women using hormone replacement therapy. Br J Cancer 91:644– 650 Borgquist S, Anagnostaki L, Jirstrom K et al, 2007, Breast tumors following combined hormone replacement therapy express favourable prognostic factors. Int J Cancer 120:2202–2207 Harding C, Knox WF, Faragher EB et al, 1996, Hormone replacement therapy and tumor grade in breast cancer: prospective study in screening unit. BMJ 312:1646–1647 Schuetz F, Diel IJ, Pueschel M et al, 2007, Reduced incidence of distant metastases and lower mortality in 1072 patients with breast cancer with a history of hormone replacement therapy. Am J Obstet Gynecol 196:342.e1–342.e9 Delgado RC, Lubian Lopez DM, 2001, Prognosis of breast cancers detected in women receiving hormone replacement therapy. Maturitas 38:147–156 Rosenberg LU, Granath F, Dickman PW et al, 2008, Menopausal hormone therapy in relation to breast cancer characteristics and prognosis: a cohort study. Breast Cancer Res 10:R78 Schairer C, Gail M, Byrne C et al, 1999, Estrogen replacement therapy and breast cancer survival in a large screening study. J Natl Cancer Inst 91:264–270 Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19:403–410 Tjonneland A, Christensen J, Thomsen BL et al, 2004, Hormone replacement therapy in relation to breast carcinoma incidence rate ratios: a prospective Danish cohort study. Cancer 100:2328–2337 Hwang ES, Chew T, Shiboski S et al, 2005, Risk factors for estrogen-positive disease. Arch Surg 140:58–62 Kumar AS, Cureton E, Shim V et al, 2006, Type and duration of exogenous hormone use affects breast cancer histology. Ann Surg Oncol 14:695–703 Kavanagh AM, Mitchell H, Giles GG, 2000, Hormone replacement therapy and accuracy of mammographic screening. Lancet 355:270–274 Banks E, 2001, Hormone replacement therapy and the sensitivity and specificity of breast cancer screening: a review. J Med Screen 8:29–34 Speroff L, 2008, Postmenopausal hormone therapy and the risk of breast cancer: a contrary thought. Menopause 15:393–400 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 917 EP 923 DI 10.1007/s12253-011-9403-x PG 7 ER PT J AU Varga, T Somogyi, A Barna, G Wichmann, B Nagy, G Racz, K Selmeci, L Firneisz, G AF Varga, Timea Somogyi, Aniko Barna, Gabor Wichmann, Barna Nagy, Geza Racz, Karoly Selmeci, Laszlo Firneisz, Gabor TI Higher Serum DPP-4 Enzyme Activity and Decreased Lymphocyte CD26 Expression in Type 1 Diabetes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Dipeptidyl peptidase-4 (DPP-4); CD26; Type 1 diabetes (T1DM); Islet cell antibody (ICA); Glutamic acid decarboxylase antibody (GAD) ID Dipeptidyl peptidase-4 (DPP-4); CD26; Type 1 diabetes (T1DM); Islet cell antibody (ICA); Glutamic acid decarboxylase antibody (GAD) AB Dipeptidyl peptidase-4 (DPP-4) is involved in the metabolism of peptide hormones, T-cell activation and proliferation. In type 1 diabetes mellitus (T1DM) β-cell destruction involves a number of dysregulated T-cells. Our aim was to assess the serum DPP-4 activity and the lymphocyte membrane bound CD26 expression in patients with type 1 diabetes and healthy controls. Ninety-eight (T1DM: 48, F/M=20/28, mean age: 34.4y; control: 50, F/M=39/11 mean age: 32,4y) individuals were included. Fasting serum DPP-4 enzymatic activity, plasma glucose (FPG), CD26 expression on CD3+, CD4+ and CD8+ lymphocytes, HbA1c and body mass index (BMI) were assessed. ICA and GAD antibodies were assessed in the T1DM group. DPP-4 enzymatic activity was determined by kinetic enzyme assay, ICA and GAD were assessed by ELISA. Determination of the CD26 expression on CD3+, CD4+ and CD8+ lymphocytes was performed by flowcytometric analysis. We found higher serum DPP-4 activity (Mean: T1DM: 30.069 U/L, control: 22.62 U/L, p<0.0001) and decreased CD26 lymphocyte expression on all lymphocyte subpopulations in T1DM. Fasting serum DPP-4 activity was independent from the ICA or GAD status of patients with T1DM. Here we first present that the serum DPP-4 activity is increased and the lymphocyte membrane bound CD26 expression is decreased in type 1 diabetes. Decreased lymphocyte membrane bound CD26 expression in T1DM might be a novel part of the T-lymphocyte regulatory dysfunction observed in type 1 diabetes mellitus. These results might provide some basis for the clinical implication of DPP-4 inhibition in patients with T1DM. C1 [Varga, Timea] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Somogyi, Aniko] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Barna, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Wichmann, Barna] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Nagy, Geza] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Racz, Karoly] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Selmeci, Laszlo] Semmelweis University, Department of Cardiovascular SurgeryBudapest, Hungary. [Firneisz, Gabor] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. RP Varga, T (reprint author), Semmelweis University, 2nd Department of Internal Medicine, Budapest, Hungary. EM timcsus@yahoo.com CR Gorrell MD, Gysbers V, McCaughan GW, 2001, CD26: a multifunctional integral membrane and secreted protein of activated lymphocytes. Scand J Immunol 54:249–264 Morimoto C, Schlossman SF, 1998, The structure and function of CD26 in the T-cell immune response. Immunol Rev 161:55–70 Ohnuma K, Yamochi T, Uchiyama M, Nishibashi K, Iwata S, Morimoto C, 2005, CD26 mediates dissociation of Tollip and IRAK-1 from caveolin-1 and induces upregulation of CD86 on antigen-presenting cells. Mol Cell Biol 25:7743–7757 Ikushima H, Munakata Y, Iwata S, Ohnuma K, Kobayashi S, Morimoto C, 2002, Soluble CD26/dipeptidyl peptidase IV enhances transendothelial migration via its interaction with mannose 6-phosphate/insulin-like growth factor II receptor. Cell Immunol 215:106–110 Ohnuma K, Takahashi N, Yamochi T, Hosono O, Dang NH, Morimoto C, 2008, Role of CD26/dipeptidyl peptidase IV in human T cell activation and function. Front Biosci 13:2299–2310 Ohnuma K, Dang NH, Morimoto C, 2008, Revisiting an old acquaintance: CD26 and its molecular mechanisms in T cell function. Trends Immunol 29(6):295–301 Stone SF, Lee S, Keane NM, Price P, French MA, 2002, Association of increased Hepatitis C Virus, HCV)–Specific IgG and soluble CD26 Dipeptidyl peptidase IV enzyme activity with hepatotoxicity after highly active antiretroviral therapy in human immunodeficiency virus–HCV-coinfected patients. J Infect Dis 186:1498–1502 Kobayashi H, Hosono U, Mimori T, Kawasaki H, Hoang Dang N, Tanaka H, Morimoto C, 2000, Reduction of serum soluble CD26/ dipeptidyl peptidase IV enzyme activity and its correlation with disease activity in systemic lupus erythematosus. J Rheumatol 29, 9):1858–1866 Cordero OJ, Salgado FJ, Meravarela A, Nogueira M, 2001, Serum interleukin-12, interleukin- 15, soluble CD26, and adenosine deaminase in patients with rheumatoid arthritis. Rheumatol Int 21:69–74 Schonermarck U, Csernok E, Trabandt A, Hansen H, Gross WL, 2000, Circulating cytokines and soluble CD23, CD26 and CD30 in ANCA associated vasculitides. Clin Exp Rheumatol 18:457– 463 Firneisz G, Lakatos PL, Szalay F, Hungarian Viral Hepatitis Study Group, 2001, Serum dipeptidyl peptidase IV, DPP IV, CD26, activity in chronic hepatitis C. Scand J Gastroenterol 36:877–880 Lefebvre J, Murphey LJ, Hartert TV, Jiao Shan R, Simmons WH, Brown NJ, 2002, Dipeptidyl peptidase IVactivity in patients with ACE-inhibitor associated angioedema. Hypertension 39:460–464 Stancikova M, Lojda Z, Lukac J, Ruzickova M, 1992, Dipeptidyl peptidase IV in patients with systemic lupus erythematosus. Clin Exp Rheumatol 10:381–385 Blazquez MV, Madueno JA, Gonzalez R, Jurado R, Bachovchin WW, Pena J, Munoz E, 1992, Selective decrease of CD26 expression in T cells from HIV-1-infected individuals. J Immunol 149(9):3073–3077 Jones D, Dang NH, Duvic M, Washington LT, Huh YO, 2001, Absence ofCD26 expression is a useful marker for diagnosis of T-cell lymphoma in peripheral blood. Am J Clin Pathol 115(6):885–892 Firneisz G, Varga T, Lengyel G, Feher J, Ghyczy D, Wichmann B, Selmeci L, Tulassay Zs, Racz K, Somogyi A, 2010, Serum dipeptidyl peptidase-4 activity in insulin resistant patients with non-alcoholic fatty liver disease: a novel liver disease biomarker. PLOS One 5(8):e12226 Varga Tet al, 2008, Higher serum dipeitidyl peptidase-4 activity in type 1 diabetes mellitus than in type 2: a direct comparison. Diabetologia 51(Suppl 1):S239, A589 Selmeci L, Szokodi I, Horvat-Karajz K, 1996, A sensitive microplate-based continuous monitoring, kinetic, assay for serum neutral endopeptidase, EC 3.4.24.11, activity. Clin Chim Acta 244:111–116 Ellis SL, Moser EG, Snell-Bergeon JK, Gutin RS, Rodionova AS, Garg SK, 2010, Effect of sitagliptin on glucose control in patients with type 1 diabetes- a pilot study. Diabetologia 53:S39, A77 Andrieu T, Thibault V, Malet I, Laporte J, Bauvois B, Agut H, Cahour A, 2003, Similar increased serum dipeptidyl peptidase IV activity in chronic hepatitis C and other viral infections. J Clin Virol 27:59–68 Hadjiyanni I, Siminovitch KA, Danska JS, Drucker DJ, 2010, Glucagon-like peptide-1 receptor signalling selectively regulates murine lymphocyte proliferation and maintenance of peripheral regulatory T cells. Diabetologia 53:730–740 Morran MP, Omenn GS, Pietropaolo M, 2008, Immunology and genetics of type 1 diabetes. Mt Sinai J Med 75(4):314–327 Hafler DA, Fox DA, Manning ME, Schlossman SF, Reinherz EL, Weiner HL, 1985, In vivo activated T lymphocytes in the peripheral blood and cerebrospinal fluid of patients with multiple sclerosis. N Engl J Med 312:1405–1411 Ohnuma K, Inoue H, Uchiyama M, Yamochi T, Hosono OD, Nam H, Morimoto C, 2006, T-cell activation via CD26 and caveolin-1 in rheumatoid synovium. Mod Rheumatol 16:3–13 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 925 EP 930 DI 10.1007/s12253-011-9404-9 PG 6 ER PT J AU Xiong, X Ren, HZ Li, MH Mei, JH Wen, JF Zheng, ChL AF Xiong, Xin Ren, Hong-Zheng Li, Min-Hua Mei, Jin-Hong Wen, Ji-Fang Zheng, Chang-Li TI Down-Regulated miRNA-214 Induces a Cell Cycle G1 Arrest in Gastric Cancer Cells by Up-Regulating the PTEN Protein SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Apoptosis; Cell cycle; Gastric cancer cell line; miRNA-214; PTEN ID Apoptosis; Cell cycle; Gastric cancer cell line; miRNA-214; PTEN AB To detect the expression of miRNA-214 in human gastric cancer cell lines of BGC823, MKN45 and SGC7901, and to identify the effect of miRNA-214 on cell cycle and apoptosis of these cells. Expression of miRNA-214 in human normal gastric mucosal cell line GES-1 and human gastric cancer cell lines was detected by real-time reversetranscription polymerase chain reaction. Antisense-miRNA-214 oligonucleotides were transfected transiently into gastric cancer cell lines to down-regulate the expression of miRNA-214. The cell cycle and apoptosis were studied by flow cytometry assay. PTEN, one of the target genes of miRNA-214 was detected by using of immunocytochemistry and Western blotting. MiRNA-214 was overexpressed in gastric cancer cell lines of BGC823, MKN45 and SGC7901 compared with normal gastric mucosal cell line GES-1. Antisense-miRNA-214 oligonucleotides significantly down-regulated the expression of miRNA-214, and increased the portion of G1-phase and decreased the portion of S-phase in BGC823 and MKN45 cells. The immunocytochemistry test and Western blotting analysis showed that the down-regulation of miRNA-214 could significantly up-regulate the expression of PTEN in BGC823 and MKN45 cells. MiRNA-214 is overexpressed in human gastric cancer cell lines of BGC823, MKN45 and SGC7901. The down-regulation of miRNA-214 could induce a G1 cell cycle arrest in them, the up-regulation of PTEN maybe one of the mechanism. C1 [Xiong, Xin] Central South University, Xiangya Basic Medical College, Department of Pathology, 410013 Changsha, Hunan Province, China. [Ren, Hong-Zheng] Central South University, Xiangya Basic Medical College, Department of Pathology, 410013 Changsha, Hunan Province, China. [Li, Min-Hua] the First Affiliated Hospital of Nanchang University, Department of Pathology, 330006 Nanchang, Jiangxi Province, China. [Mei, Jin-Hong] the First Affiliated Hospital of Nanchang University, Department of Pathology, 330006 Nanchang, Jiangxi Province, China. [Wen, Ji-Fang] Central South University, Xiangya Basic Medical College, Department of Pathology, 410013 Changsha, Hunan Province, China. [Zheng, Chang-Li] Central South University, Xiangya Basic Medical College, Department of Pathology, 410013 Changsha, Hunan Province, China. RP Zheng, ChL (reprint author), Central South University, Xiangya Basic Medical College, Department of Pathology, 410013 Changsha, China. EM changlizheng1125@yahoo.com.cn CR Parkin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108., DOI 10.3322/canjclin. 55.2.74 Chen CZ, 2005, MicroRNAs as oncogenes and tumor suppressors. New Engl J Med 353:1768–1771., DOI 10.1056/NEJMp058190 Zhang B, Pan X, CobbGP et al, 2007, MicroRNAs as oncogenes and tumor suppressors. Dev Biol 302:1–12., DOI 10.1016/j. ydbio.2006.08.028 Zhang HW, Li MB, Han Y et al, 2010, Down-Regulation of miR- 27a Might Reverse MultidrugResistance of Esophageal Squamous Cell Carcinoma. Dig Dis Sci 55:2545–2551., DOI 10.1007/s10620- 009-1051-6 Song B, Wang C, Liu J et al, 2010, MicroRNA-141 Regulates Smad Interacting Protein 1, SIP1, and Inhibits Migration and Invasion of Colorectal Cancer Cells. Dig Dis Sci 55:2365–2372., DOI 10.1007/s10620-009-1008-9 Ihara M, Yamamoto H, Kikuchi A, 2005, SUMO-1 modification of PIASy, an E3 ligase, is necessary for PIASy-dependent activation of Tcf-4. Mol Cell Biol 25:3506–3518., DOI 10.1128/ MCB.25.9.3506-3518.2005 Zhang HH, Wang XJ, Li GX et al, 2007, Detection of let-7a microRNA by real-time PCR in gastric carcinoma. World J Gastroenterol 13:2883–2888, PMid:17569129 Petrocca F, Visone R, Onelli MR et al, 2008, E2F1-regulated microRNAs impair TGFbeta-dependent cell-cycle arrest and apoptosis in gastric cancer. Cancer Cell 13:272–286., DOI 10.1016/j.ccr.2008.02.013 Li Z, Zhan W, Wang Z et al, 2006, Inhibition of PRL-3 gene expression in gastric cancer cell line SGC7901 via microRNA suppressed reduces peritoneal metastasis. Biochem Biophys Res Commun 348:229–237., DOI 10.1016/j.bbrc.2006.07.043 Motoyama K, Inoue H, Nakamura Y et al, 2008, Clinical significance of high mobility group A2 in human gastric cancer and its relationship to let-7 microRNA family. Clin Cancer Res 14:2334–2340., DOI 10.1158/1078-0432.CCR-07-4667 Xia L, Zhang D, Du R et al, 2008, miR-15b and miR-16 modulate multidrug resistance by targeting BCL2 in human gastric cancer cells. Int J Cancer 123:372–379., DOI 10.1002/ijc.23501 Volinia S, Calin GA, Liu CG et al, 2006, A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA 103:2257–2261., DOI 10.1073/pnas.0510565103 Flynt AS, Li N, Thatcher EJ et al, 2007, Zebrafishc modulates Hedgehog signaling to specify muscle cell fate. Nat Genet 39:259–263., DOI 10.1038/ng1953 Yang H, Kong W, He L et al, 2008, MicroRNA expression profiling in human ovarian cancer: miR-214 induces cell survival and cisplatin resistance by targeting PTEN. Cancer Res 68:425– 433., DOI 10.1158/0008-5472.CAN-07-2488 Cheng AM, Byrom MW, Shelton J et al, 2005, Antisense inhibition of human miRNAs and indications for an involvement of miRNA in cell growth and apoptosis. Nucleic Acids Res 33:1290–1297., DOI 10.1093/nar/gki200 Griffiths-Jones S, Grocock RJ, van Dongen S et al, 2006, miRBase: MicroRNA sequences, targets and gene nomenclature. Nucleic Acids Res 34:140–144., DOI 10.1093/nar/gkj112 LivakKJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−delta delta C(T), method. Methods 25(4):402–408., DOI 10.1006/meth.2001.1262 Cimmino A, Calin GA, Fabbri M et al, 2005, miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA 102:13944–13949., DOI 10.1073/pnas.0506654102 Kluiver J, Haralambieva E, de Jong D et al, 2006, Lack of BIC and microRNA miR-155 expression in primary cases of Burkitt lymphoma. Genes Chromosomes Cancer 45:147–153., DOI 10.1002/gcc.20273 Eis PS, Tam W, Sun L et al, 2005, Accumulation of miR-155 and BIC RNA in human B cell lymphomas. Proc Natl Acad Sci USA 102:3627–3632., DOI 10.1073/pnas.0500613102 Chan SH, Wu CW, Li AF et al, 2008, miR-21 microRNA expression in human gastric carcinomas and its clinical association. Anticancer Res 28:907–911, PMid:14744438 do Kim N, Chae HS, Oh ST et al, 2007, Expression of viral microRNAs in Epstein-Barr virus-associated gastric carcinoma. J Virol 81:1033–1036., DOI 10.1128/JVI.02271-06 Ueda T, Volinia S, Okumura H et al, 2010, Relation between microRNA expression and progression and prognosis of gastric cancer: a microRNA expression analysis. Lancet Oncol 11:136– 146., DOI 10.1016/S1470-2045(09)70343-2 Papagiannakopoulos T, Shapiro A, Kosik KS, 2008, MicroRNA- 21 targets a network of key tumor-suppressive pathways in glioblastoma cells. Cancer Res 68:8164–8172., DOI 10.1158/0008- 5472.CAN-08-1305 Decembrini S, Bressan D, Vignali R et al, 2009, MicroRNAs couple cell fate and developmental timing in retina. Proc Natl Acad Sci USA 106:21179–21184., DOI 10.1073/pnas.0909167106 Fei J, Lan FF, Guo M et al, 2008, Inhibitory effects of antimiRNA oligonucleotides, AMOs, on A549 cell growth. J Drug Target 16:688–693., DOI 10.1080/10611860802295946 Meng F, Henson R, Lang M et al, 2006, Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines. Gastroenterology 130:2113–2129., DOI 10.1053/j.gastro.2006.02.057 Chan JA, Krichevsky AM, Kosik KS, 2005, MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res 65:6029–6033., DOI 10.1158/0008-5472.CAN-05-0137 Kang YH, Lee HS, Kim WH, 2002, Promoter methylation and silencing of PTEN in gastric carcinoma. Lab Invest 82:285–291, PMid:11896207 Hwang PH, Kim SY, Lee JC et al, 2005, PTEN/MMAC1 enhances the growth inhibition by anticancer drugs with downregulation of IGF-II expression in gastric cancer cells. Exp Mol Med 37:391–398, PMid:16264263 Hang Y, Zheng YC, Cao Y et al, 2005, Suppression of gastric cancer growth by adenovirus-mediated transfer of the PTEN gene. World J Gastroenterol 11:2224–2229, PMid:15818730 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 931 EP 937 DI 10.1007/s12253-011-9406-7 PG 7 ER PT J AU Wu, Q Yang, Z Hu, S Su, T An, Y Zhang, Z Nie, Y Wang, X Shi, Y Fan, D AF Wu, Qiong Yang, Zhiping Hu, Sijun Su, Tao An, Yanxin Zhang, Zhiyong Nie, Yongzhan Wang, Xin Shi, Yongquan Fan, Daiming TI Stem Cell Associated Genes Working with One MiRNA Cluster Have Different Clinic Pathologic Values in Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Clinic pathologic; PTEN; P21; P57; C-Myc; Stemness; Prognosis ID Clinic pathologic; PTEN; P21; P57; C-Myc; Stemness; Prognosis AB Cancer stem cells are nowadays considered to be the origin of cancer. Also, stem cell associated genes are emerging as predictors of cancer malignancy. We investigated the association of several stemness genes (c-Myc, PTEN, p57 and p21) with clinic pathological parameters and survival in stomach cancer by performing immunohistochemistry on paraffin sections of gastric cancer patients who underwent surgical staging with following-up statistics. We discovered that expression of c-Myc was significantly related to distant metastasis, the combined expression of PTEN and p21 correlated positively to overall survival, while p57 was less useful in overall survival prediction in gastric cancer. Additionally, there is a positive correlation between expressions of p57 and p21. In conclusion, our present study indicated that expression of stemness genes (c-Myc, PTEN, p57 and p21) performed different predictive potential in the evaluation of clinical malignancy levels in gastric cancer. C1 [Wu, Qiong] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 127 Changle Western Road, 710032 Xi’an, Shaanxi, China. [Yang, Zhiping] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 127 Changle Western Road, 710032 Xi’an, Shaanxi, China. [Hu, Sijun] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 127 Changle Western Road, 710032 Xi’an, Shaanxi, China. [Su, Tao] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 127 Changle Western Road, 710032 Xi’an, Shaanxi, China. [An, Yanxin] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 127 Changle Western Road, 710032 Xi’an, Shaanxi, China. [Zhang, Zhiyong] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 127 Changle Western Road, 710032 Xi’an, Shaanxi, China. [Nie, Yongzhan] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 127 Changle Western Road, 710032 Xi’an, Shaanxi, China. [Wang, Xin] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 127 Changle Western Road, 710032 Xi’an, Shaanxi, China. [Shi, Yongquan] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 127 Changle Western Road, 710032 Xi’an, Shaanxi, China. [Fan, Daiming] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 127 Changle Western Road, 710032 Xi’an, Shaanxi, China. RP Shi, Y (reprint author), Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. EM shiyquan@fmmu.edu.cn CR Shipitsin M, Polyak K, 2008, The cancer stem cell hypothesis: in search of definitions, markers, and relevance. Lab Invest 88:459– 463 Boman BM, Wicha MS, 2008, Cancer stem cells: a step toward the cure. J Clin Oncol 26:2795–2799 Yamashita T, Forgues M, Wang W et al, 2008, EpCAM and - fetoprotein expression defines novel prognostic subtypes of hepatocellular carcinoma. Cancer Res 68:1451–1461 Piccioli P, Balbi G, Serra M et al, 2009, CTLA-4 +49A>G polymorphism of recipients of HLA-matched sibling allogeneic stem cell transplantation is associated with survival and relapse incidence. Ann Hematol 89:613–618 Reya T, Morrison SJ, Clarke MF et al, 2001, Stem cells, cancer, and cancer stem cells. Nature 414:105–111 Kanatsu-Shinohara M, Takashima S, Shinohara T, 2010, Transmission distortion by loss of p21 or p27 cyclin-dependent kinase inhibitors following competitive spermatogonial transplantation. Proc Natl Acad Sci 107:6210–6215 Fasano CA, Phoenix TN, Kokovay E et al, 2009, Bmi-1 cooperates with Foxg1 to maintain neural stem cell self-renewal in the forebrain. Genes Dev 23:561–574 Georgia S, Soliz R, Li M et al, 2006, p57 and Hes1 coordinate cell cycle exit with self-renewal of pancreatic progenitors. Dev Biol 298:22–31 Gregorian C, Nakashima J, Le Belle J et al, 2009, Pten deletion in adult neural stem/progenitor cells enhances constitutive neurogenesis. J Neurosci 29:1874–1886 Zheng H, Ying H, Yan H et al, 2008, p53 and Pten control neural and glioma stem/progenitor cell renewal and differentiation. Nature 455:1129–1133 Chen H, Qian K, Tang ZP et al, 2010, Bioinformatics and microarray analysis of microRNA expression profiles of murine embryonic stem cells, neural stem cells induced from ESCs and isolated from E8·5 mouse neural tube. Neurol Res 32:603–613 Inomata M, Tagawa H, Guo YM et al, 2008, MicroRNA-17-92 down-regulates expression of distinct targets in different B-cell lymphoma subtypes. Blood 113:396–402 Dews M, Homayouni A, Yu D et al, 2006, Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster. Nat Genet 38:1060–1065 Varlakhanova NV, Cotterman RF, deVries WN et al, 2010, myc maintains embryonic stem cell pluripotency and self-renewal. Differentiation 80:9–19 Wang J, Wang H, Li Z et al, 2008, c-Myc is required for maintenance of glioma cancer stem cells. PLoS ONE 3:e3769 Satyanarayana A, Hilton MB, Kaldis P, 2008, p21 Inhibits Cdk1 in the absence of Cdk2 to maintain the G1/S phase DNA damage checkpoint. Mol Biol Cell 19:65–77 Guo Y-L, Chakraborty S, Rajan SS et al, 2010, Effects of oxidative stress on mouse embryonic stem cell proliferation, apoptosis, senescence, and self-renewal. Stem Cells Dev 19:1321– 1331 Kouraklis G, Katsoulis IE, Theocharis S et al, 2008, Does the expression of Cyclin E, pRb, and p21 correlate with prognosis in gastric adenocarcinoma? Dig Dis Sci 54:1015–1020 Lee K-H, Lee HE, Cho SJ et al, 2008, Immunohistochemical analysis of cell cycle-related molecules in gastric carcinoma: prognostic significance, correlation with clinicopathological parameters, proliferation and apoptosis. Pathobiology 75:364–372 Mackay HJ, Gallinger S, Tsao MS et al, 2010, Prognostic value of microsatellite instability, MSI, and PTEN expression in women with endometrial cancer: Results from studies of the NCIC Clinical Trials Group, NCIC CTG). Eur J Cancer 46:1365–1373 Zheng, 2009, PTEN expression and mutation in colorectal carcinomas. Oncology Reports 22 Capodanno A, Camerini A, Orlandini C et al, 2009, Dysregulated PI3K/Akt/PTEN pathway is a marker of a short disease-free survival in node-negative breast carcinoma. Hum Pathol 40:1408– 1417 Wang C, Yang R, Yue D et al, 2009, Expression of FAK and PTEN in bronchioloalveolar carcinoma and lung adenocarcinoma. Lung 187:104–109 Axanova LS, Chen YQ, McCoy T et al, 2010, 1,25-dihydroxyvitamin D3 and PI3K/AKT inhibitors synergistically inhibit growth and induce senescence in prostate cancer cells. Prostate 70:1658–1671 Ito Y, Takeda T, Sakon M et al, 2001, Expression of p57/Kip2 protein in hepatocellular carcinoma. Oncology 61:221–225 Hsu KW, Hsieh RH, Wu CW et al, 2009, MBP-1 suppresses growth and metastasis of gastric cancer cells through COX-2. Mol Biol Cell 20:5127–5137 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 939 EP 946 DI 10.1007/s12253-011-9407-6 PG 8 ER PT J AU Li, D Cui, Q Liu, Y Wang, X Liu, Ch Liu, Sh Zeng, Y AF Li, Dingfeng Cui, Qiu Liu, Yaosheng Wang, Xiaohong Liu, Cheng Liu, Shubin Zeng, Yanjun TI Chemotherapy Response Analysis for Osteosarcom with Intra-arterial Chemotherapy by Subcutaneous Implantable Delivery System SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; Neoadjuvant chemotherapy; Arterial chemotherapy; Survival rate ID Osteosarcoma; Neoadjuvant chemotherapy; Arterial chemotherapy; Survival rate AB To summarize the experience in intraarterial neoadjuvant chemotherapy for extremity osteosarcoma. Between January 2002 and December 2007,111 patients with stage IIB extremity osteosarcoma received preoperative intraarterial therapy with subcutaneous implantation of chemotherapy pump as well as en bloc resection, and postoperative adjuvant chemotherapy. There were 63 males and 48 females with an average age of 18 (range, 14~39 years). The time from symptom onset to hospitalization varied from several days to 6 months. The induction chemotherapy regimen includes: epirubicin [50~70 mg/m2 by 4-hour intraarterial infusion/day for 3 day] and cisplatin [100~120 mg/m2 by 2-hour intraarterial infusion/day for 3 days] repetitively every 2~3 weeks. Among which 24 cases only received two cycles induction chemotherapy was set to nonstandard chemotherapy group and 87 cases received three to six cycles induction chemotherapy set to standard chemotherapy group. The number of preoperative chemotherapy-cycles of standard chemotherapy group depends on the clinical and radiographic evaluation of chemotherapy efficacy. Median follow-up time was 28(8~48) months. The rate of limb preservation surgery was 89.53% (77/86) in standard chemotherapy group,and was 37.5% (9/24) in nonstandard chemotherapy group. Kaplan-Meier survival analysis showed that the 3-year overall survival rate and disease free survival rate of all the 111 cases were 68.3% and 65.9% respectively. There were significant differences in overall survival rate (38.9%, 80.0%, P=0.000), disease free survival rate (30.1%, 79.5%, P=0.000), distant metastasis rate (66.67%, 16.09%, P=0.0000) and local recurrence rate (58.33%, 13.79%, P=0.0000) between nonstandard chemotherapy group and standard chemotherapy group. Standard intraarterial neo-adjuvant chemotherapy was more effective than nonstandard intraarterial induction chemotherapy to stage IIB extremity osteosarcoma. C1 [Li, Dingfeng] 307 Hospital of PLA, Department of Orthopedics, 100071 Beijing, China. [Cui, Qiu] 307 Hospital of PLA, Department of Orthopedics, 100071 Beijing, China. [Liu, Yaosheng] 307 Hospital of PLA, Department of Orthopedics, 100071 Beijing, China. [Wang, Xiaohong] Beijing University of Technology, Biomechanics and Medical Information Institute, 100022 Beijing, China. [Liu, Cheng] 307 Hospital of PLA, Department of Orthopedics, 100071 Beijing, China. [Liu, Shubin] 307 Hospital of PLA, Department of Orthopedics, 100071 Beijing, China. [Zeng, Yanjun] Beijing University of Technology, Biomechanics and Medical Information Institute, 100022 Beijing, China. RP Li, D (reprint author), 307 Hospital of PLA, Department of Orthopedics, 100071 Beijing, China. EM 307yygk@sina.com CR Rosen G, Marcove RC, Caparros B et al, 1979, Primary osteogenic sarcoma - the rationale for preoperative chemotherapy and delayed surgery. Cancer 43:2163–2192 Bacci G, Longhi A, Versari M et al, 2006, Prognostic factors for osteosarcoma of the extremity treated with neoadjuvant chemotherapy. Fifteen year experience in 789 patients treated at a single institution. Cancer 106:1154–1161 Guo W, Yang RL, Tang XD et al, 2004, Neoadjuvant chemotherapy for osteosarcoma. Natl Med J Chin 84:1186–1190 Ferrari S, Smeland S, Mercuri M, Italian and Scandinavian Sarcoma Groups et al, 2005, Neoadjuvant chemotherapy with high-dose ifosfamide, high-dose methotrexate, Cisplatin, and doxorubicin for patients with localized ostesarcoma of the extremity. A joint study by the Italian and Scandinavian Sarcoma Groups. J Clin Oncol 23:8845–8852 Goorin AM, Schwartzentruber DJ, Devidas M et al, 2003, Presurgical chemotherapy compared with immediate surgery and adjuvant chemotherapy for nonmetastatic osteosarcoma: Pediatric Oncology Group Study POG-8651. J Clin Oncol 21:1574–1580 Bacci G, Ferrari S, Longhi A et al, 2003, Preoperative therapy versus immediate surgery in nonmetastatic osteosarcoma. J Clin Oncol 21:4662–4663 Li D, Cui Q, Fan H et al, 2005, Extendible replacement of the distal femur in the treatment of osteosarcoma in growing individuals. Chin J Reparative Reconstr Surg 19:560–562 Dahlin DC, Coventry MB, 1967, Osteogenic sarcoma. A study of six hundred cases. J Bone Joint Surg Am 49:101–110 Lin F, Dong Y, Tang X et al, 2006, Clinical analysis on efficacy and prognosis of new adjuvant chemotherapy for stageIIB osteosarcoma. Tumor 26:1011–1014 Rao BN, Rodriguez-Galindo C, 2003, Intra-arterial cisplatin in osteosarcoma: same question, different answer. Ann Surg Oncol 10:481–483 Guo wei; Li dasen; Shen danhua et al, 2006, Treatment of multifocal osteosarcoma. Chin J Orthop 26:378–380 Bramwell VH, Burgers MV, Souhami RL et al, 1997, A randomized comparison of two short intensive chemotherapy regimens in children and young adults with osteosarcoma: results in patients with metastases: a study of the European Osteosarcoma Intergroup. Sarcoma 1:155–160 Souhami RL, CraftAW,Van der EijkenJWet al, 1997, Randomised trial of two regimens of chemotherapy in operable osteosarcoma: a study of the European Osteosarcoma Intergroup. Lancet 350(9082):911–917 Stewart DJ, Benjamin RS, Zimmerman S et al, 1983, Clinical pharmacology of intraarterial cis-diamminedichloroplatinum(II). Cancer Res 43:917–920 Jaffe N, Knapp J, Chuang VP et al, 1983, Osteosarcoma: intra-arterial treatment of the primary tumorwith cis-diammine-dichloroplatinumII, CDP). Angiographic, pathologic, and pharmacologic studies. Cancer 51:402–407 Wilkins RM, Cullen JW, Camozzi AB et al, 2005, Improved survival in primary nonmetastatic pediatric osteosarcoma of the extremity. Clin Orthop Relat Res 438:128–136 Hugate RR, Wilkins RM, Kelly CM et al, 2008, Intraarterial chemotherapy for extremity osteosarcoma and MFH in adults. Clin Orthop Relat Res 466:1292–1301 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 947 EP 953 DI 10.1007/s12253-011-9408-5 PG 7 ER PT J AU Stanczak, A Stec, R Bodnar, L Olszewski, W Cichowicz, M Kozlowski, W Szczylik, C Pietrucha, T Wieczorek, M Lamparska-Przybysz, M AF Stanczak, Aleksandra Stec, Rafal Bodnar, Lubomir Olszewski, Wojciech Cichowicz, Marzena Kozlowski, Wojciech Szczylik, Cezary Pietrucha, Tadeusz Wieczorek, Maciej Lamparska-Przybysz, Monika TI Prognostic Significance of Wnt-1, β-catenin and E-cadherin Expression in Advanced Colorectal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE β-catenin; E-cadherin; Wnt-1; Wnt signaling pathway; Advanced colorectal carcinoma; Prognostic factor ID β-catenin; E-cadherin; Wnt-1; Wnt signaling pathway; Advanced colorectal carcinoma; Prognostic factor AB Wnt/β-catenin pathway plays an important role in initiation and progression of colorectal oncogenesis. The aim of this study was to determine expression and localization of E-cadherin, β-catenin and Wnt-1 proteins in colorectal tumors. Expression of β-catenin, E-cadherin and Wnt-1 was determined by immunohistochemistry on advanced colorectal cancers. Abnormal expression of Ecadherin, β-catenin, Wnt-1 was observed. Additionally, we revealed correlations between levels of studied proteins and histoclinical data. In multivariate analysis nuclear β-catenin, higher carcinoembryonic antigen serum level before treatment, female sex and tumor localized in colon or rectum were independent unfavorable prognostic factors. These findings support the hypothesis that Wnt/β-catenin pathway plays an important role in advanced colorectal carcinoma. C1 [Stanczak, Aleksandra] Medical University of Lodz, Department of Medical BiotechnologyLodz, Poland. [Stec, Rafal] Military Institute of Medicine, Oncology DepartmentWarsaw, Poland. [Bodnar, Lubomir] Military Institute of Medicine, Oncology DepartmentWarsaw, Poland. [Olszewski, Wojciech] Medical Center for Postgraduate Education, and Maria Sk³odowska-Curie Memorial Cancer Center and Institute of Oncology, Department of PathologyWarsaw, Poland. [Cichowicz, Marzena] Military Institute of Medicine, Pathomorphology DepartmentWarsaw, Poland. [Kozlowski, Wojciech] Military Institute of Medicine, Pathomorphology DepartmentWarsaw, Poland. [Szczylik, Cezary] Military Institute of Medicine, Oncology DepartmentWarsaw, Poland. [Pietrucha, Tadeusz] Medical University of Lodz, Department of Medical BiotechnologyLodz, Poland. [Wieczorek, Maciej] Celon Pharma Ltd, Innovative Drugs Research & Development Department, 41A Mokra St. Kielpin, 05–092 Lomianki, Poland. [Lamparska-Przybysz, Monika] Celon Pharma Ltd, Innovative Drugs Research & Development Department, 41A Mokra St. Kielpin, 05–092 Lomianki, Poland. RP Stanczak, A (reprint author), Medical University of Lodz, Department of Medical Biotechnology, Lodz, Poland. EM aleksandra.stanczak@celonpharma.com CR Ferlay J, Shin HR, Bray F, Forman D,Mathers C, Parkin DM(2010, GLOBOCAN 2008, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 10. Lyon, France: International Agency for Research on Cancer. http://globocan.iarc.fr. Cited 20 Sept 2010 Sant M, Allemani C, Santaquilani M, Knijn A,Marchesi F, Capocaccia R, 2009, EUROCARE Working Group. EUROCARE-4. Survival of cancer patients diagnosed in 1995–1999. Results and commentary. Eur J Cancer 45:931–91 American Joint Committee on Cancer, 2010, Colon and rectum. In: Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, eds, AJCC Cancer Staging Manual, The Seventh Edition. Springer, New York Compton CC, Fielding LP, Burgart LJ, Conley B, Cooper HS, Hamilton SR et al, 2000, Prognostic factors in colorectal cancer. College of American Pathologists Consensus Statement 1999. Arch Pathol Lab Med 124:979–94 Chen JS, Hsieh PS, Chiang JM, Yeh CY, Tsai WS, Tang R et al, 2010, Clinical outcome of signet ring cell carcinoma and mucinous adenocarcinoma of the colon. Chang Gung Med J 33:51–7 Matsumoto K, Nakayama Y, Inoue Y, Minagawa N, Katsuki T, Shibao K et al, 2007, Lymphatic microvessel density is an independent prognostic factor in colorectal cancer. Dis Colon Rectum 50:308–14 MacDonald BT, Tamai K, He X, 2009, Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell 17:9–26 Tetsu O, McCormick F, 1999, Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 398:422–6 Brabletz T, Jung A, Dag S, Hlubek F, Kirchner T, 1999, beta-catenin regulates the expression of the matrix metalloproteinase-7 in human colorectal cancer. Am J Pathol 155:1033–8 He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT et al, 1998, Identification of c-MYC as a target of the APC pathway. Science 281:1509–12 Zhang T, Otevrel T, Gao Z, Gao Z, Ehrlich SM, Fields JZ et al, 2001, Evidence that APC regulates survivin expression: a possible mechanism contributing to the stem cell origin of colon cancer. Cancer Res 61:8664–7 Gavert N, Ben-Ze’ev A, 2007, beta-Catenin signaling in biological control and cancer. J Cell Biochem 102:820–8 Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN et al, 1992, APC mutations occur early during colorectal tumorigenesis. Nature 359:235–7 Sparks AB, Morin PJ, Vogelstein B, Kinzler KW, 1998, Mutational analysis of the APC/beta-catenin/Tcf pathway in colorectal cancer. Cancer Res 58:1130–4 Morin PJ, Sparks AB, Korinek V, Barker N, Clevers H, Vogelstein B et al, 1997, Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 275:1787–90 Hugh TJ, Dillon SA, O’Dowd G, Getty B, Pignatelli M, Poston GJ et al, 1999, beta-catenin expression in primary and metastatic colorectal carcinoma. Int J Cancer 82:504–11 Hugh TJ, Dillon SA, Taylor BA, Pignatelli M, Poston GJ, Kinsella AR, 1999, Cadherin-catenin expression in primary colorectal cancer: a survival analysis. Br J Cancer 80:1046–51 Han SA, Chun H, Park CM, Kang SJ, Kim SH, Sohn D et al, 2006, Prognostic significance of beta-catenin in colorectal cancer with liver metastasis. Clin Oncol, R Coll Radiol, 18:761–7 Krzakowski M, ed,, 2001, Onkologia kliniczna, 1st edn. Warsaw, Borgis Wydawnictwo Medyczne Delektorskaya VV, Perevoshchikov AG, Golovkov DA, Kushlinskii NE, 2005, Expression of E-cadherin, beta-catenin, and CD-44v6 cell adhesion molecules in primary tumors and metastases of colorectal adenocarcinoma. Bull Exp Biol Med 139:706–10 Choi HN, Kim KR, Lee JH, Park HS, Jang KY, Chung MJ et al, 2009, Serum response factor enhances liver metastasis of colorectal carcinoma via alteration of the E-cadherin/beta-catenin complex. Oncol Rep 21:57–63 Filiz AI, Senol Z, Sucullu I, Kurt Y, Demirbas S, Levhi Akin M, 2010, The survival effect of E-cadherin and catenins in colorectal carcinomas. Colorectal Dis 12:1223–30 El-Bahrawy MA, Poulsom R, Jeffery R, Talbot I, Alison MR, 2001, The expression of E-cadherin and catenins in sporadic colorectal carcinoma. Hum Pathol 32:1216–24 Schuhmacher C, Becker I, Oswald S, Atkinson MJ, Nekarda H, Becker KF et al, 1999, Loss of immunohistochemical E-cadherin expression in colon cancer is not due to structural gene alterations. Virchows Arch 434:489–95 Hahn-Stromberg V, Edvardsson H, Bodin L, Franzen L, 2008, Disturbed expression of E-cadherin, beta-catenin and tight junction proteins in colon carcinoma is unrelated to growth pattern and genetic polymorphisms. APMIS 116:253–62 Huiping C, Sigurgeirsdottir JR, Jonasson JG, Eiriksdottir G, Johannsdottir JT, Egilsson V et al, 1999, Chromosome alterations and E-cadherin gene mutations in human lobular breast cancer. Br J Cancer 81:1103–10 Huiping C, Kristjansdottir S, Jonasson JG, Magnusson J, Egilsson V, Ingvarsson S, 2001, Alterations of E-cadherin and beta-catenin in gastric cancer. BMC Cancer 1:16 Truant SC, Gouyer VP, Leteurtre EA, Zerimech F, Huet GM, Pruvot FR, 2008, E-cadherin and beta-catenin mRNA levels throughout colon cancer progression. J Surg Res 150:212–8 Avizienyte E, Wyke AW, Jones RJ, McLean GW, Westhoff MA, Brunton VG et al, 2002, Src-induced de-regulation of E-cadherin in colon cancer cells requires integrin signalling. Nat Cell Biol 4:632–8 Ikeguchi M, Makino M, Kaibara N, 2001, Clinical significance of E-cadherin-catenin complex expression in metastatic foci of colorectal carcinoma. J Surg Oncol 77:201–7 Miyamoto S, Endoh Y, Hasebe T, Ishii G, Kodama K, Goya M et al, 2004, Nuclear beta-catenin accumulation as a prognostic factor in Dukes’ D human colorectal cancers. Oncol Rep 12:245–51 Elzagheid A, Buhmeida A, Korkeila E, Collan Y, Syrjanen K, Pyrhonen S, 2008, Nuclear beta-catenin expression as a prognostic factor in advanced colorectal carcinoma. World J Gastroenterol 14:3866–71 Vider BZ, Zimber A, Chastre E, Prevot S, Gespach C, Estlein D et al, 1996, Evidence for the involvement of the Wnt 2 gene in human colorectal cancer. Oncogene 12:153–8 Holcombe RF, Marsh JL, Waterman ML, Lin F, Milovanovic T, Truong T, 2002, Expression of Wnt ligands and Frizzled receptors in colonic mucosa and in colon carcinoma. Mol Pathol 55:220–6 Khor TO, Gul YA, Ithnin H, Seow HF, 2006, A comparative study of the expression of Wnt-1, WISP-1, survivin and cyclin-D1 in colorectal carcinoma. Int J Colorectal Dis 21:291–300 Nishisho I, Nakamura Y, Miyoshi Y, Miki Y, Ando H, Horii A et al, 1991, Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science 253:665–669 Nakamura Y, Nishisho I, Kinzler KW, Vogelstein B, Miyoshi Y, Miki Y et al, 1992, Mutations of the APC, adenomatous polyposis coli, gene in FAP, familial polyposis coli, patients and in sporadic colorectal tumors. Tohoku J Exp Med 168:141–7 Varesco L, Gismondi V, James R, Robertson M, Grammatico P, Groden J et al, 1993, Identification of APC gene mutations in Italian adenomatous polyposis coli patients by PCR-SSCP analysis. Am J Hum Genet 52:280–5 Polakis P, Hart M, Rubinfeld B, 1999, Defects in the regulation of beta-catenin in colorectal cancer. Adv Exp Med Biol 470:23–32 Joo M, Shahsafaei A, Odze RD, 2009, Paneth cell differentiation in colonic epithelial neoplasms: evidence for the role of the Apc/ beta-catenin/Tcf pathway. Hum Pathol 40:872–80 Chiang JM, Chou YH, Chen TC, Ng KF, Lin JL, 2002, Nuclear beta-catenin expression is closely related to ulcerative growth of colorectal carcinoma. Br J Cancer 86:1124–9 Kapiteijn E, Liefers GJ, Los LC, Kranenbarg EK, Hermans J, Tollenaar RA et al, 2001, Mechanisms of oncogenesis in colon versus rectal cancer. J Pathol 195:171–8 Aamodt R, Bondi J, Andersen SN, Bukholm G, Bukholm IRK, 2008, Expression of nuclear β-Catenin in rectal versus colonic cancers. Open Clin Cancer J 2:13–17 Wray CM, Ziogas A, Hinojosa MW, Le H, Stamos MJ, Zell JA, 2009, Tumor subsite location within the colon is prognostic for survival after colon cancer diagnosis. Dis Colon Rectum 52:1359–66 Bufill JA, 1990, Colorectal cancer: evidence for distinct genetic categories based on proximal or distal tumor location. Ann Intern Med 113:779–88 Li FY, Lai MD, 2009, Colorectal cancer, one entity or three. J Zhejiang Univ Sci B 10:219–29 Nilbert M, Planck M, Fernebro E, Borg A, Johnson A, 1999, Microsatellite instability is rare in rectal carcinomas and signifies hereditary cancer. Eur J Cancer 35:942–5 Frattini M, Balestra D, Suardi S, Oggionni M, Alberici P, Radice P et al, 2004, Different genetic features associated with colon and rectal carcinogenesis. Clin Cancer Res 10:4015–21 Horst D, Reu S, Kriegl L, Engel J, Kirchner T, Jung A, 2009, The intratumoral distribution of nuclear beta-catenin is a prognostic marker in colon cancer. Cancer 115:2063–70 Mulcahy MF, Benson AB 3rd, 1999, The role of carcinoembryonic antigen monitoring in management of colorectal cancer. Curr Oncol Rep 1:168–72 Bast RC Jr, Ravdin P, Hayes DF, Bates S, Fritsche H Jr, Jessup JM et al, 2001, 2000 update of recommendations for the use of tumor markers in breast and colorectal cancer: clinical practice guidelines of the American Society of Clinical Oncology. J Clin Oncol 19:1865–78 Duffy MJ, van Dalen A, Haglund C, Hansson L, Holinski-Feder E, Klapdor R et al, 2007, Tumour markers in colorectal cancer: European Group on Tumour Markers, EGTM, guidelines for clinical use. Eur J Cancer 43:1348–60 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 955 EP 963 DI 10.1007/s12253-011-9409-4 PG 9 ER PT J AU Chen, ShW Guo, ZM Zhang, Q Yang, AK Li, QL Zhuang, ShM Wang, LP Song, M AF Chen, Shu-Wei Guo, Zhu-Ming Zhang, Quan Yang, An-Kui Li, Qiu-Li Zhuang, Shi-Min Wang, Li-Ping Song, Ming TI Invasion of the Hypoglossal Nerve by Adenoid Cystic Carcinoma of the Tongue: Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Neural invasion; Adenoid cystic carcinoma; Hypoglossal nerve; Tongue; Treatment ID Neural invasion; Adenoid cystic carcinoma; Hypoglossal nerve; Tongue; Treatment AB Adenoid cystic carcinoma (ACC) is a rare but highly aggressive malignancy mainly originating from the salivary glands. ACC is well known for its propensity toward neural invasion (NI). NI is the process of neoplastic invasion in and along nerves. It is a distinct and welldocumented phenomenon in ACC; however, it is an underestimated route of metastatic spread. Multiple distant metastases can be established through NI route, and NI is believed to portend a poor prognosis. Despite increasing recognition of NI in many malignancies, the molecular mechanism behind NI is not well established. We present a unique case of hypoglossal nerve invasion by ACC arising from the minor salivary glands in the tongue of a 34-yearold man. We also review and discuss current theories on the pathogenesis and mechanism of NI. C1 [Chen, Shu-Wei] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. [Guo, Zhu-Ming] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. [Zhang, Quan] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. [Yang, An-Kui] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. [Li, Qiu-Li] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. [Zhuang, Shi-Min] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. [Wang, Li-Ping] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. [Song, Ming] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. RP Song, M (reprint author), Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 510060 Guangzhou, China. EM songming@sysucc.org.cn CR Spiro RH, Huvos AG, Strong EW, 1974, Adenoid cystic carcinoma of salivary origin. A clinicopathologic study of 242 cases. Am J Surg 128:512–520 El-Naggar AK, Huvos AG, 2005, Adenoid cystic carcinoma. In: Barnes L, Eveson JW, Reichart P, Sidransky D, eds, World Health Organization classification of tumors: pathology and genetics of head and neck tumors. IARC, Lyon Barrett AW, Speight PM, 2009, Perineural invasion in adenoid cystic carcinoma of the salivary glands: a valid prognostic indicator? Oral Oncol 45:936–940 Silvester KC, Barnes S, 1990, Adenoid cystic carcinoma of the tongue presenting as a hypoglossal nerve palsy. Br J Oral Maxillofac Surg 28:122–124 Wang D, Li Y, He H, Liu L, Wu L, He Z, 2007, Intraoral minor salivary gland tumors in a Chinese population: a retrospective study on 737 cases. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 104:94–100 van der Wal JE, Snow GB, van der Waal I, 1990, Intraoral adenoid cystic carcinoma. The presence of perineural spread in relation to site, size, local extension, and metastatic spread in 22 cases. Cancer 66:2031–2033 Agarwal JP, Jain S, Gupta T, Tiwari M, Laskar SG, Dinshaw KA et al, 2008, Intraoral adenoid cystic carcinoma: prognostic factors and outcome. Oral Oncol 44:986–993 Hanna E, Vural E, Prokopakis E, Carrau R, Snyderman C, Weissman J, 2007, The sensitivity and specificity of highresolution imaging in evaluating perineural spread of adenoid cystic carcinoma to the skull base. Arch Otolaryngol Head Neck Surg 133:541–545 Bockman DE, Buchler M, Beger HG, 1994, Interaction of pancreatic ductal carcinoma with nerves leads to nerve damage. Gastroenterology 107:219–230 Krasna MJ, Flancbaum L, Cody RP, Shneibaum S, Ben AG, 1988, Vascular and neural invasion in colorectal carcinoma. Incidence and prognostic significance. Cancer 61:1018–1023 Beard CJ, Chen MH, Cote K, Loffredo M, Renshaw AA, Hurwitz M et al, 2004, Perineural invasion is associated with increased relapse after external beam radiotherapy for men with low-risk prostate cancer and may be a marker for occult, high-grade cancer. Int J Radiat Oncol Biol Phys 58:19–24 Goepfert H, Dichtel WJ, Medina JE, Lindberg RD, Luna MD, 1984, Perineural invasion in squamous cell skin carcinoma of the head and neck. Am J Surg 148:542–547 Hong SK, Kwak C, Jeon HG, Lee E, Lee SE, 2005, Do vascular, lymphatic, and perineural invasion have prognostic implications for bladder cancer after radical cystectomy? Urology 65:697–702 Shirai K, Ebata T, Oda K, Nishio H, Nagasaka T, Nimura Y et al, 2008, Perineural invasion is a prognostic factor in intrahepatic cholangiocarcinoma. World J Surg 32:2395–2402 Marchesi F, Piemonti L, Mantovani A, Allavena P, 2010, Molecular mechanisms of perineural invasion, a forgotten pathway of dissemination and metastasis. Cytokine Growth Factor Rev 21:77–82 Ayala GE, Wheeler TM, Shine HD, Schmelz M, Frolov A, Chakraborty S et al, 2001, In vitro dorsal root ganglia and human prostate cell line interaction: redefining perineural invasion in prostate cancer. Prostate 49:213–223 Cornell RJ, Rowley D, Wheeler T, Ali N, Ayala G, 2003, Neuroepithelial interactions in prostate cancer are enhanced in the presence ofprostatic stroma. Urology 61:870–875 Ceyhan GO, Demir IE, Altintas B, Rauch U, Thiel G, Muller MW et al, 2008, Neural invasion in pancreatic cancer: a mutual tropism between neurons and cancer cells. Biochem Biophys Res Commun 374:442–447 Ceyhan GO, Giese NA, Erkan M, Kerscher AG, Wente MN, Giese T et al, 2006, The neurotrophic factor artemin promotes pancreatic cancer invasion. Ann Surg 244:274–281 Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM et al, 2006, Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci 9:917–924 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 965 EP 968 DI 10.1007/s12253-010-9339-6 PG 4 ER PT J AU Yu, L Yang, JSh AF Yu, Lu Yang, Jing Shou TI Kaposiform Hemangioendothelioma of the Spleen in an Adult: An Initial Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Kaposiform hemangioendothelioma; Kasabach–Merritt phenomenon; Vascular tumor; Immunohistochemistry; Spleen ID Kaposiform hemangioendothelioma; Kasabach–Merritt phenomenon; Vascular tumor; Immunohistochemistry; Spleen AB Kaposiform hemangioendothelioma (KHE) is a rare locally aggressive vascular neoplasm characterized by infiltrating nodules and sheets of spindle cells, and unmistakable resemblance to Kaposi's sarcoma. KHE occurs mainly in newborns and infants and presents most commonly in the skin, deep soft tissue, and bone. We report a case of KHE in a 36-year-old female who presented with a spleen mass and underwent splenectomy. Macroscopic examination revealed a large, dark-red, firm mass in the spleen. Histologically, the tumor consisted of irregular, infiltrating nodules of densely packed spindle-shaped tumor cells closely associated with small slit-like and sieve-like blood vessels, which were separated with hyalinized hypocellular fibrous stroma. Immunohistochemically, both spindle and epithelioid cells were positive for CD34, CD31, and vimentin, but negative for EMA, cytokeratin, CD21, CD35, CD1a, and S-100 protein. The well-formed capillaries and mature vessels but not spindle tumor cell showed reactivity for factor VIII- related antigen. Alpha-Smooth muscle actin was detected in pericytes surrounding small round or slit-like capillaries. The final histologic diagnosis was KHE. Followup 6 month after operation revealed no sign of recurrence or metastasis.To the best of our knowledge, this is the first report of KHE arising in the spleen. C1 [Yu, Lu] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, No. 17 Chang Le Xi Road, 710032 Xi’an, Shaanxi, China. [Yang, Jing Shou] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, No. 17 Chang Le Xi Road, 710032 Xi’an, Shaanxi, China. RP Yang, JSh (reprint author), Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. EM yangsj@fmmu.edu.cn CR Tsang WYW, 2002, Kaposiform hemangioendothelioma. World Health Organization Classification of Tumours: pathology and genetics of tumours of the soft tissues and bones. In: Fletcher CDM, Unni KK, Mertens F, eds). IARC, Lyon, pp 163–164 Zukerberg LR, Nickoloff BJ, Weiss SW, 1993, Kaposiform hemangioendothelioma of infancy and childhood. An aggressive neoplasm associated with Kasabach-Merritt syndrome and lymphangiomatosis. Am J Surg Pathol 17:321–328 Lyons LL, North PE, Mac-Moune Lai F, Stoler MH, Folpe AL, Weiss SW, 2004, Kaposiform hemangioendothelioma: a study of 33 cases emphasizing its pathologic, immunophenotypic, and biologic uniqueness from juvenile hemangioma. Am J Surg Pathol 28:559–568 Mac-Moune Lai F, To KF, Choi PC, Leung PC, Kumta SM, Yuen PP, Lam WY, Cheung AN, Allen PW, 2001, Kaposiform hemangioendothelioma: five patients with cutaneous lesion and long follow-up. Mod Pathol 14:1087–1092 Fernandez Y, Bernabeu-Wittel M, Garcia-Morillo JS, 2009, Kaposiform hemangioendothelioma. Eur J Intern Med 20:106–113 Mentzel T, Mazzoleni G, Dei Tos AP, Fletcher CD, 1997, Kaposiform hemangioendothelioma in adults. Clinicopathologic and immunohistochemical analysis of three cases. Am J Clin Pathol 108:450–455 San Miguel FL, Spurbeck W, Budding C, Horton J, 2008, Kaposiform hemangioendothelioma: a rare cause of spontaneous hemothorax in infancy. Review of the literature. J Pediatr Surg 43:e37–e41 Tello MA, Shields G, Gadre SA, Ryan M, 2004, Pathology quiz case 2. Diagnosis: Kaposiform hemangioendothelioma, KHE). Arch Otolaryngol Head Neck Surg 130:991, diag 993–994 Sarkar M, Mulliken JB, Kozakewich HP, Robertson RL, Burrows PE, 1997, Thrombocytopenic coagulopathy, Kasabach-Merritt phenomenon, is associated with Kaposiform hemangioendothelioma and not with common infantile hemangioma. Plast Reconstr Surg 100:1377–1386 Blei F, Karp N, Rofsky N, Rosen R, Greco MA, 1998, Successful multimodal therapy for kaposiform hemangioendothelioma complicated by Kasabach-Merritt phenomenon: case report and review of the literature. Pediatr Hematol Oncol 15:295–305 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 969 EP 972 DI 10.1007/s12253-010-9331-1 PG 4 ER PT J AU Cui, Q Li, D Liu, Ch Guo, J Liu, Sh Liu, Y Zhai, MJ Zeng, Y AF Cui, Qiu Li, Dingfeng Liu, Cheng Guo, Jun Liu, Shubin Liu, Yaosheng Zhai, M J Zeng, Yanjun TI Two Case-Reports of the Limb Salvage Treatment of Osteosarcoma Consolidated with Obvious Pathological Fractures SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Osteosarcoma; Pathological fracture; Limb salvage treatment; Chemotherapy ID Osteosarcoma; Pathological fracture; Limb salvage treatment; Chemotherapy AB Objective: The effect of the limb salvage of the treatment of Osteosarcoma Consolidated with Obvious Pathological Fractures is not very well, the purpose of this paper is to track the efficacy of limb salvage treatment when the patients accepted the artery intervention chemotherapy and enclosed 2 clinical case-reports. Methods: From January 2003 to September 2005, 2 clinical cases which one is a male, 29 years old, was confirmed osteosarcoma on the left distal femur, and the other is a female, 15 years old and has osteosarcoma on the right arm with obvious pathological fracture. After receiving arterial chemotherapy pump embedded, then started chemotherapy after the tumor biopsy, and the next process prosthesis replacement in limb salvage surgery after 5 times chemotherapy, follow on the next 5 times chemotherapy after the surgery. Results: With an average follow-up 70 months, there are no postoperative infection and prosthesis loosening found, also didn’t detected tumor recurrence and metastasis, and the limb function recovered well. Conclusions: With the effective, neoadjuvant chemotherapy and comprehensive treatment, salvage treatment is not the contraindication of the patients with pathological fractures of combined primary osteosarcoma, and the treatment with long-term follow-up effectively. C1 [Cui, Qiu] Academy of Military Medical Sciences, Department of Bone Tumor, No.8 of East Avenue, Fengtai District, 100071 Beijing, China. [Li, Dingfeng] Academy of Military Medical Sciences, Department of Bone Tumor, No.8 of East Avenue, Fengtai District, 100071 Beijing, China. [Liu, Cheng] Academy of Military Medical Sciences, Department of Bone Tumor, No.8 of East Avenue, Fengtai District, 100071 Beijing, China. [Guo, Jun] Academy of Military Medical Sciences, Department of Bone Tumor, No.8 of East Avenue, Fengtai District, 100071 Beijing, China. [Liu, Shubin] Academy of Military Medical Sciences, Department of Bone Tumor, No.8 of East Avenue, Fengtai District, 100071 Beijing, China. [Liu, Yaosheng] Academy of Military Medical Sciences, Department of Bone Tumor, No.8 of East Avenue, Fengtai District, 100071 Beijing, China. [Zhai, M J] Beijing University of Technology, Biomechanics and Medical Information Institute, 100022 Beijing, China. [Zeng, Yanjun] Beijing University of Technology, Biomechanics and Medical Information Institute, 100022 Beijing, China. RP Li, D (reprint author), Academy of Military Medical Sciences, Department of Bone Tumor, 100071 Beijing, China. EM 307yygk@sina.com CR McKenna RJ, Schwinn CP, Soong KY et al, 1966, Sarcomata of osteogenic series: an analysis of 553 cases. J Bone Joint Surg Am 48:1–26 Krugluger J, Gisinger B, Windhager R, Salzer-Kuntschik M,Kotz R, 1993, Fracture in osteosarcoma. J Bone Joint Surg [Br] 75:210 Kim MS, Lee SY, Lee TR et al, 2009, Prognostic nomogram for predicting the 5 year probability of developing metastasis after neo-adjuvant chemotherapy and definitive surgery for AJCC stage II extremity osteosarcoma. Ann Oncol 20:955–960 Bramer JA, van Linge JH, Grimer RJ, Scholten RJ, 2009, Prognostic factors in localized extremity osteosarcoma: A systematic review. Eur J Surg Oncol 35(10):1030–1036 Scully SP, Ghert MA, Zurakowski D, Thompson RC, Gebhardt MC.Pathologic fracture in osteosarcoma : prognostic importance and treatment implications. J Bone Joint Surg Am. 2002;84- A:49-57. Shin KH, Rougraff BT, Simon MA, 1994, Oncologic outcome of primary bone sarcoma of the pelvis. Clin Orthop 304:207–217 Jaffe N, Spears R, Eftekhari F et al, 1987, Impact of chemotherapy on primary tumor and survival. Cancer 59(4):701–709 Adubu A, Sferopoulos NK, Tillman RM et al, 1996, The surgical treatment and outcome of pathological fractures in localised osteosarcoma. J Bone Joint Surg Br 78(5):694–8 Zeifang F, Sabo D, Ewerbeck V, 2000, Pathological fracture in primary malignant bone tumors. Chirurg 71(9):1121–1125 Mayil V, Natarajan RH, Govardhan S et al, 2000, Limb salvage sugery for for pathological fractures in osteosarcoma. Int Orthop 24:170–172 Bacci G, Ferrari S, Longhi A, 2003, Nonmetastatic osteosarcoma of the extremity with pathologic fracture at presentation:local and systemic control by amputation or limb salvage after preoperative chemotherapy. Acta Orthop Scand 74(4):449–54 Ebeid W, Amin S, Abdelmegid A, 2005, Limb Salvage Management of Pathologic Fractures of Primary Malignant Bone Tumors. Cancer Control 12(1):57–61 Rongli Y, Yi G, Wanpeng X et al, 2004, Surgical Treatment of Pathological Fracture Caused by Bone Tumor. Chin J Clin Oncol 31(11):620–623 Huai J, Jian-jun L, Wei X et al, 2008, Neoadjuvant chemotherapy combined with limb salvage surgery for treatment of extremity osteosarcoma with pathological fracture. Journal of Clinical Orthopaedics 11(3):236–239 Yibo W, Li DF, 2002, Experimental and clinical of the caffeine as a cancer enhancer. Chin J Clin Oncol 29(6):450–453 Eilber FC, Rosen G, Eckardt J et al, 2001, Treatment-induced pathologic necrosis:a predictor of local recurrence and survival in patients receiving neoadjuvant therapy for high-grade extremity soft tissue sarcomas. J Clin Oncol 19(13):3203–3209 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2011 VL 17 IS 4 BP 973 EP 979 DI 10.1007/s12253-010-9347-6 PG 7 ER PT J AU Vegso, Gy Gorog, D Fehervari, I Nemes, B Doros, A Langer, MR Kobori, L AF Vegso, Gyula Gorog, Denes Fehervari, Imre Nemes, Balazs Doros, Attila Langer, Miklos Robert Kobori, Laszlo TI Role of Organ Transplantation in the Treatment of Malignancies – Hepatocellular Carcinoma as the Most Common Tumour Treated with Transplantation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Bridging therapy; Bronchioloalveolar carcinoma; Des-gamma-carboxy prothrombin; Extended criteria; Heart sarcomas; Hepatocellular carcinoma; Living donor liver transplantation; Liver transplantation; Milan criteria; Proliferation signal inhibitor ID Bridging therapy; Bronchioloalveolar carcinoma; Des-gamma-carboxy prothrombin; Extended criteria; Heart sarcomas; Hepatocellular carcinoma; Living donor liver transplantation; Liver transplantation; Milan criteria; Proliferation signal inhibitor AB There are only few malignant tumours where organ transplantation is the treatment of choice. Transplantation can be considered individually in certain lung carcinomas, unresectable heart tumours, cholangiocellular carcinoma and Klatskin tumour. It is acceptable in unresectable chemosensitive hepatoblastoma, epitheloid haemangioendothelioma, liver metastasis of neuroendocrine tumours and as the most common indication, the early hepatocellular carcinoma (HCC) in cirrhotic liver. Results of liver transplantation (LT) for HCC according to Milan criteria as a "gold standard" are excellent. Time of LT has a great influence on the results. While patients are on waiting list, locoregional therapies may help prevent tumour progress. Living donor LT is an acceptable treatment of HCC. The greatest experience with this procedure is in Asia. Despite the favourable results, LT as the treatment of HCC is debated and raises several questions: regarding indication and expectable outcome. Milan criteria seem to answer this questions although they are too strict. The number and size of HCC foci per se is not sufficient predictor of eligibility to transplantation and for prognosis. Majority of the prognostic factors can be evaluated only after transplantation with pathological examination of HCC. Aim of the present research is to find prognostic factors that are characteristic of biological behaviour of HCC, which can be detected before LT in order to select patients who have the greatest benefit from LT. Re-definition of eligibility criteria is an actual question; an international consensus based on additional prospective studies is required for the "new" recommendation. C1 [Vegso, Gyula] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, 1082 Budapest, Hungary. [Gorog, Denes] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, 1082 Budapest, Hungary. [Fehervari, Imre] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, 1082 Budapest, Hungary. [Nemes, Balazs] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, 1082 Budapest, Hungary. [Doros, Attila] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, 1082 Budapest, Hungary. [Langer, Miklos Robert] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, 1082 Budapest, Hungary. [Kobori, Laszlo] Semmelweis University, Department of Transplantation and Surgery, Baross u. 23, 1082 Budapest, Hungary. RP Vegso, Gy (reprint author), Semmelweis University, Department of Transplantation and Surgery, 1082 Budapest, Hungary. EM vegso.gyula@med.semmelweis-univ.hu CR Buell JF, Gross TG, Woodle ES, 2005, Malignancy after transplantation. Transplantation 80:S254–S264 Knoll G, Cockfield S, Blydt-Hansen T et al, 2005, Canadian Society of Transplantation consensus guidelines on eligibility for kidney transplantation. CMAJ 173:1181–1184 De Perrot M, Fischer S, Waddel TK et al, 2004, Role of lung transplantation in the treatment of bronchogenic carcinomas for patients with end-stage pulmonary disease. J Clin Oncol 22:4351– 4356 Mathew J, Kratzke RA, 2009, Lung cancer and lung transplantation. J Thorac Oncol 4:753–760 Gowdamarajan A, Michler RE, 2000, Therapy for primary cardiac tumors: is there a role for heart transplantation? Curr Op Cardiol 15:121–126 Shanmugam G, 2006, Primary cardiac sarcoma. Eur J Cardiothorac Surg 29:925–932 Gupta A, 2008, Primary cardiac sarcomas. Expert Rev Cardiovasc Ther 6:1295–1297 O’Grady JG, 2000, Treatment options for other hepatic malignancies. Liver Transpl 6:S23–S29 Ito F, Cho CS, Rikkers LF et al, 2009, Hilar cholangiocarcinoma: current management. Ann Surg 250:210–218 Robles R, Parilla P, Sanchez-Bueno F et al, 2010, Liver transplantation increases R0 resection and survival of patients with a non-disseminated unresectable Klatskin tumour. Cir Esp 87:82–88 Rea DJ, Heimbach JK, Rosen CB et al, 2005, Liver transplantation with neoadjuvant chemoradiation is more effective than resection for hilar cholangiocarcinoma. Ann Surg 242:451–458 Hertl M, Cosimi AB, 2005, Liver transplantation for malignancy. Oncologist 10:269–281 Pankin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Gomaa AI, Khan SA, Toledano MB et al, 2008, Hepatocellular carcinoma: epidemiology, risk factors and pathogenesis. World J Gastroenterol 14:4300–4308 He J, Gu D, Wu X et al, 2005, Major causes of death among men and women in China. N Engl J Med 353:1124–1134 Schutte K, Bornschein J, Malfertheiner P, 2009, Hepatocellular carcinoma—epidemiological trends and risk factor. Dig Dis 27:80–92 Llovet JM, Bru C, Bruix J, 1999, Prognosis of hepatocellular carcinoma: the BCLC staging classification. Semin Liver Dis 19:329–338 Hsu CY, Hsia CY, Huang YH et al, 2010, Selecting an optimal staging system for hepatocellular carcinoma: comparison of 5 currently used prognostic models. Cancer 116:3006–3014 Forner A, Ayuso C, Real MB et al, 2009, Diagnosis and treatment of hepatocellular carcinoma. Med Clin 132:272–286 Dank M, 2009, New possibilities in the management of hepatocellular carcinoma. Lege Artis Med 19:15–21 Llovet JM, Bruix J, 2008, Molecular targeted therapies in hepatocellular carcinoma. Hepatology 48:1312–1327 Tremosini S, Reig M, de Lope CD et al, 2010, Treatment of early hepatocellular carcinoma: towards personalized therapy. Dig Liver Dis 42:S242–S248 Bruix J, Sherman M, 2011, Management of hepatocellular carcinoma: an update. Hepatology 53:1020–1022 El-Serag HB, Marrero JA, Rudolph L et al, 2008, Diagnosis and treatment of hepatocellular carcinoma. Gastroenterology 134:1752–1763 Ishizaki Y, Kawasaki S, 2008, The evolution of liver transplantation for hepatocellular carcinoma, past, present, and future). J Gastroenterol 43:18–26 Ringe B, Pichlmayr R, Wittekind C et al, 1991, Surgical treatment of hepatocellular carcinoma: experience with liver resection and transplantation in 198 patients. World J Surg 15:270–285 Mazzaferro V, Regalia E, Doci R et al, 1996, Liver transplantation for the treatment of small hepatocellular carcinoma in patients with cirrhosis. N Engl J Med 334:693–699 Sharma P, Balan V, Hernandez JL et al, 2004, Liver transplantation for hepatocellular carcinoma: the MELD impact. Liver Transpl 10:36–41 Raia S, Nery JR, Mies S, 1989, Liver transplantation from live donors. Lancet 2:497 Hashikura Y, Makuuchi M, Kawasaki S et al, 1994, Successful living-related partial liver transplantation to an adult patient. Lancet 343:1233–1234 Tanwar S, Khan SA, Grover VPB et al, 2009, Liver transplantation for hepatocellular carcinoma. World J Gastroenterol 15:5511–5516 Chung HY, Chan SC, Lo CM et al, 2010, Strategies for widening liver donor pool. Asian J Surg 33:63–69 de Villa VH, Lo CM, Chen CH, 2003, Ethics and rationale of livingdonor liver transplantation in Asia. Transplantation 15:S2–S5 Taketomi A, Soejima Y, Yoshizumi T et al, 2008, Liver transplantation for hepatocellular carcinoma. J Hepatobiliary Pancreat Surg 15:124–130 Lee SG, Hwang S, Moon DB et al, 2008, Expanded indication criteria of living donor liver transplantation for hepatocellular carcinoma at one large-volume center. Liver Transpl 14:935–945 Vakili K, Pomposelli JJ, Cheah YL et al, 2009, Living donor liver transplantation for hepatocellular carcinoma: increased recurrence but improved survival. Liver Transpl 15:1861–1866 Kaihara S, Kiuchi T, Ueda M et al, 2003, Living-donor liver transplantation for hepatocellular carcinoma. Transplantation 75: S37–S40 Todo S, Furukawa H, Japanese Study Group on Organ Transplantation, 2004, Living donor liver transplantation for adult patients with hepatocellular carcinoma. Experience in Japan. Ann Surg 240:451–461 Fisher RA, Kulik LM, Freise CE et al, 2007, Hepatocellular carcinoma recurrence and death following living and deceased donor liver transplantation. Am J Transplant 7:1601–1608 Silva MF, Wigg AJ, 2010, Current controversies surrounding liver transplantation for hepatocellular carcinoma. J Gastoenterol Hepatol 25:1217–1226 Kaido T, Uemoto S, 2010, Does living donation have adventages over deceased donation in liver transplantation ? J Gastroenterol Hepatol 25:1598–1603 Washburn K, 2010, Model for end stage liver disease and hepatocellular carcinoma: a moving target. Transplant Rev 24:11–17 Rayaie K, Feng S, 2007, Allocation policy for hepatocellular carcinomain the MELD era: room for improvement ? Liver Transpl 13:S36–S43 Lesurtel M, Mullhaupt B, Pestalozzi BC et al, 2006, Transarterial chemoembolization as a bridge to liver transplantation for hepatocellular carcinoma: an evidence-based analysis. Am J Transpl 6:2644–2650 Lopez PM, Villanueva A, Roayaie S et al, 2006, Neoadjuvant therapies for hepatocellular carcinoma before liver transplantation: a critical appraisal. Liver Transpl 12:1747–1754 Northup PG, Argo CK, Shah NL et al, 2009, Locoregional therapy of T1 hepatocellular carcinomas is associated with increased transplant waiting list mortality and offers no additional post-transplant survival. Hepatology 50:306A Llovet JM, Mas X, Aponte JJ et al, 2002, Cost-effectiveness of adjuvant therapy for hepatocellular carcinoma during the waiting list for liver transplantation. Gut 50:123–128 Sandrousi C, Dawson LA, Lee M et al, 2009, Radiotherapy as a bridge to liver transplantation for hepatocellular carcinoma. Transpl Int 23:299–306 Lau WY, Lai EC, Leung TW, 2010, Current role of selective internal irradiation with Yttrium-90 microspheres in the management of hepatocellular carcioma: a systematic review. Int J Radiat Oncol Biol Phys., DOI 10.016/j.ijrobp. 2010.06.010 Llovet JM, Ricci S, Mazzaferro V et al, 2008, Sorafenib in advenced hepatocellular carcinoma. N Engl J Med 359:378–390 Vitale A, Volk ML, Pastorelli D et al, 2010, Use of sorafenib in patients with hepatocellular carcinoma before liver transplantation: a cost-benefit analysis while awaiting data on sorafenib safety. Hepatology 51:165–173 Bhoori S, Sposito C, Germini A et al, 2010, The challanges of liver transplantation for hepatocellular carcinoma on cirrhosis. Transplant Int 23:712–722 Navasa M, Bruix J, 2010, Multifaced perspective of the waiting list for liver transplantation: the value of pharmacokinetic models. Hepatology 51:12–15 Rowe IA, 2010, Sorafenib therapy in patients with hepatocellular carcinoma before liver transplantation. Hepatology 52:1171–1172 Ravaioli M, Grazi GL, Piscaglia F et al, 2008, Liver transplantation for hepatocellular carcinoma: results of down-staging in patients initially outside the Milan selection criteria. Am J Transplant 8:2547–2557 Jang JW, You CR, Kim CW et al, 2010, Benefit of downsizing hepatocellular carcinoma in a liver transplant population. Aliment Pharmacol Ther 31:415–423 Bhoori S, Schiavo M, Russo A et al, 2007, First-line treatment for hepatocellular carcinoma: resection or transplantation ? Transplant Proc 39:2271–2273 Yao FY, 2008, Liver transplantation for hepatocellular carcinoma: beyond the Milan criteria. Am J Transpl 8:1982–1989 Silva M, Moya A, Berenguer M et al, 2008, Expanded criteria for liver transplantation in patients with cirrhosis and hepatocellular carcinoma. Liver Transpl 14:1449–1460 Yao FY, Xiao L, Bass NM et al, 2007, Liver transplantation for hepatocellular carcinoma: validation of the UCSF-expanded criteria based on preoperative imaging. Am J Transplant 7:2587– 2596 Duffy JP, Vardanian A, Benjamin E et al, 2007, Liver transplantation criteria for hepatocellular carcinoma should be expanded: a 22-year experience with 467 patients at UCLA. Ann Surg 246:502–511 Mazzaferro V, Llovet JM, Miceli R et al, 2009, Predicting survival after liver transplantation in patients with hepatocellular carcinoma beyond the Milan criteria: a retrospective, exploratory analysis. Lancet Oncol 10:35–43 Sotiropoulos GC, Molmenti EP, Lang H, 2009, Milan criteria, upto- seven criteria, and the illusion of a rescue package for patients with liver cancer. Lancet Oncol 10:207–208 Toso C, Asthana S, Bigam DL et al, 2009, Reassessing selection criteria prior to liver transplantation for hepatocellular carcinoma utilizing the Scientific Registry of Transplant Recipients database. Hepatology 49:832–838 Ito T, Takada Y, Ueda M et al, 2007, Expansion of selection criteria for patients with hepatocellular carcinoma in living donor liver transplantation. Liver Transpl 13:1637–1644 Hwang S, Lee SG, Belghiti J, 2009, Liver transplantation for HCC: its role: Eastern and Western perspectives. J Hepatobiliary Pancreat Surg., DOI 10.1007/s00534-009-0241-0 Sugawara Y, Tamura S, Makuuchi M, 2007, Living donor liver transplantation for hepatocellular carcinoma: Tokyo University series. Dig Dis 25:310–312 Soejima Y, Taketomi Y, Yoshizumi T et al, 2007, Extended indication for living donor liver transplantation in patients with hepatocellular carcinoma. Transplantation 83:893–899 Todo S, Furukawa H, Tada M, Japanese Liver Transplantation Study Group, 2007, Extending indication: role of living donor liver transplantation for hepatocellular carcinoma. Liver Transpl 13:S48–S54 Kwon CH, Kim DJ, Han YS et al, 2007, HCC in living donor liver transplantation: can we expand the Milan criteria ? Dig Dis 25:313–319 Bruix J, Llovet JM, 2002, Prognostic prediction and treatment strategies in hepatocellular carcinoma. Hepatology 35:519–524 Belghiti J, Durand F, 2011, Criteria for liver transplantation for hepatocellular carcinoma: what is an acceptable outcome ? Liver Int Suppl 1:161–163., DOI 10.1111/j.1478-3231.2010.024113.x Hayashi PH, Trotter JF, Forman L et al, 2004, Impact of transplant diagnosis of hepatocellular carcinoma on cadaveric liver allocation in the era of MELD score. Liver Transpl 1:42–48 Vibert E, Azoulay D, Hoti E et al, 2010, Progression of alphafetoprotein before liver transplantation for hepatocellular carcinoma in cirrhotic patients: a critical factor. Am J Transplant 10:129–137 Shirabe K, Itoh S, Yoshizumi Y et al, 2007, The predictors of microvascular invasion in candidates for liver transplantation with hepatocellular carcinoma–with special reference to the serum levels of des-gamma-carboxy prothrombin. J Surg Oncol 95:253– 240 Fujikawa T, Shiraha H, Yamamoto K, 2009, Significance of desgamma- carboxy prothrombin production in hepatocellular carcinoma. Acta Med Okayama 63:299–304 Shimada M, Yonemura Y, Ijichi H et al, 2005, Living donor liver transplantation for hepatocellular carcinoma: a special reference to a preoperative des-gamma-carboxy prothrombin value. Transplant Proc 37:1177–1179 Lee JW, Paeng JC, Kang KW et al, 2009, Prediction of tumor recurrence by 18F-FDG PET in liver transplantation for hepatocellular carcinoma. J Nucl Med 50:682–687 Toso C, Merani S, Bigam DL et al, 2010, Sirolimus-based immunosuppression is associated with increased survival after liver transplantation for hepatocellular carcinoma. Hepatology 51:1237–1243 Wang Z, Zhou J, Fan J et al, 2008, Effect of rapamycin alone and in combination with sorafenib in an orthotopic model of human hepatocellular carcinoma. Clin Cancer Res 14:5124– 5130 Bhoori S, Toffanin S, Sposito C et al, 2010, Personalized molecular targeted therapy in advenced, recurrent hepatocellular carcinoma after liver transplantation: a proof of principle. J Hepatol 52:771–775 Wang Y, Speeg KV, Washburn WK et al, 2010, Sirolimus plus sorafenib in treating HCC recurrence after liver transplantation: a case report. World J Gastroenterol 16:5518–5522 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 1 EP 10 DI 10.1007/s12253-011-9441-4 PG 10 ER PT J AU Kakihana, K Ohashi, K Hirashima, Y Murata, Y Kobayashi, T Yamashita, T Sakamaki, H Akiyama, H AF Kakihana, Kazuhiko Ohashi, Kazuteru Hirashima, Yuka Murata, Yutaka Kobayashi, Takeshi Yamashita, Takuya Sakamaki, Hisashi Akiyama, Hideki TI Clinical Impact of Pre-transplant Pulmonary Impairment on Survival After Allogeneic Hematopoietic Stem Cell Transplantation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Fatal pulmonary complication; Obstructive ventilatory impairment; Restrictive ventilatory impairment; Reduced intensity conditioning regimen ID Fatal pulmonary complication; Obstructive ventilatory impairment; Restrictive ventilatory impairment; Reduced intensity conditioning regimen AB We retrospectively analyzed the clinical outcomes of patients with pulmonary impairment before undergoing allogeneic hematopoietic stem cell transplantation (HSCT) for the first time. Among 297 evaluable patients who underwent their first HSCT, 23 had restrictive, obstructive or mixed ventilatory impairment (n=9, 13 and 1, respectively). Males predominated among the patients with pulmonary impairment (p=0.037) and received a reduced intensity conditioning (RIC) regimen more frequently, although the difference did not reach statistical significance (p=0.05). Among 23 patients with pulmonary impairment, 9 underwent post-transplant pulmonary function tests and obstructive ventilatory impairment progressed only in 2 patients, both of whom developed bronchiolitis obliterans. Kaplan-Meier estimates of 3-year overall (OS) among patients with and without pulmonary impairment were 57% and 47%, respectively, and those of relapse-free survival (RFS) were 70%, and 68%, respectively, with no significant differences between the groups (OS, p=0.235; RFS, p=0.287). The rates of nonrelapse mortality also did not significantly differ (p=0.835). Our results suggest that allogeneic HSCT is safe for patients with pulmonary impairment. The lower frequency of fatal pulmonary complications after HSCT and the RIC regimen might contribute to favorable survival rates. C1 [Kakihana, Kazuhiko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113–8677 Tokyo, Japan. [Ohashi, Kazuteru] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113–8677 Tokyo, Japan. [Hirashima, Yuka] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Diseases Center, Pharmacy DivisionTokyo, Japan. [Murata, Yutaka] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113–8677 Tokyo, Japan. [Kobayashi, Takeshi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113–8677 Tokyo, Japan. [Yamashita, Takuya] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113–8677 Tokyo, Japan. [Sakamaki, Hisashi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113–8677 Tokyo, Japan. [Akiyama, Hideki] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113–8677 Tokyo, Japan. RP Kakihana, K (reprint author), Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 113–8677 Tokyo, Japan. EM kakihana@cick.jp CR Marras TK, Szalai JP, Chan CK, Lipton JH, Messner HA, Laupacis A, 2002, Pulmonary function abnormalities after allogeneic marrow transplantation: a systematic review and assessment of an existing predictive instrument. Bone Marrow Transplant 30:599–607 Badier M, Guillot C, Delpierre S, Vanuxem P, Blaise D, Maraninchi D, 1993, Pulmonary function changes 100 days and one year after bone marrow transplantation. Bone Marrow Transplant 12:457–461 Gore EM, Lawton CA, Ash RC, Lipchik RJ, 1996, Pulmonary function changes in long-term survivors of bone marrow transplantation. Int J Radiat Oncol Biol Phys 36:67–75 Chiou TJ, Tung SL, Wang WS, Tzeng WF, Yen CC, Fan FS et al, 2002, Pulmonary function changes in long-term survivors of chronic myelogenous leukemia after allogeneic bone marrow transplantation: a Taiwan experience. Cancer Invest 20:880–888 Clark JG, Schwartz DA, Flournoy N, Sullivan KM, Crawford SW, Thomas ED, 1987, Risk factors for airflow obstruction in recipients of bone marrow transplants. Ann Intern Med 107:648–656 El-Khatib M, Bou-Khalil P, Abbas O, Salman A, Jamaleddine G, 2007, Value of pretransplant pulmonary function tests in predicting pulmonary complications after autologous peripheral stem cell transplantation. Lung 185:321–324 Savani BN, Montero A, Srinivasan R, Singh A, Shenoy A, Mielke S et al, 2006, Chronic GVHD and pretransplantation abnormalities in pulmonary function are the main determinants predicting worsening pulmonary function in long-term survivors after stem cell transplantation. Biol Blood Marrow Transplant 12:1261–1269 Chien JW, Martin PJ, Gooley TA, Flowers ME, Heckbert SR, Nichols WG et al, 2003, Airflow obstruction after myeloablative allogeneic hematopoietic stem cell transplantation. Am J Respir Crit Care Med 168:208–214 Chien JW, Madtes DK, Clark JG, 2005, Pulmonary function testing prior to hematopoietic stem cell transplantation. Bone Marrow Transplant 35:429–435 Ghalie R, Szidon JP, Thompson L, Nawas YN, Dolce A, Kaizer H, 1992, Evaluation of pulmonary complications after bone marrow transplantation: the role of pretransplant pulmonary function tests. Bone Marrow Transplant 10:359–365 Lee MY, Chiou TJ, Yang MH, Bai LY, Hsiao LT, Chao TC et al, 2005, Relatively favorable outcomes of post-transplant pulmonary function in patients with chronic myeloid leukemia receiving non-myeloablative allogeneic hematopoietic stem cell transplantation. Eur J Haematol 74:152–157 Goldberg SL, Klumpp TR, Magdalinski AJ, Mangan KF, 1998, Value of the pretransplant evaluation in predicting toxic day-100 mortality among blood stem-cell and bone marrow transplant recipients. J Clin Oncol 16:3796–3802 Sorror ML, Maris MB, Storb R, Baron F, Sandmaier BM, Maloney DG et al, 2005, Hematopoietic cell transplantation, HCT)-specific comorbidity index: a new tool for risk assessment before allogeneic HCT. Blood 106:2912–2919 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 11 EP 16 DI 10.1007/s12253-011-9383-x PG 6 ER PT J AU Liu, X He, Z Li, Ch Huang, G Ding, C Liu, H AF Liu, Xingyan He, Zhiwei Li, Cai-hong Huang, Guoliang Ding, Congcong Liu, Hong TI Correlation Analysis of JAK-STAT Pathway Components on Prognosis of Patients with Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Janus kinases; Signal transducers and activator of transcription; Clinical pathology; Prognosis ID Prostate cancer; Janus kinases; Signal transducers and activator of transcription; Clinical pathology; Prognosis AB Janus kinases (JAK)/signal transducers and activator of transcription (STAT) pathway is activated constitutively in prostate cancer (PCa). Despite previous reports implying a role of this pathway in the development of clinical hormone-refractory PCa, the correlation of pathway members with the clinicopathologic features and prognosis of patients with PCa has not been elucidated. To address this problem, pJAK-1Tyr1022/1023 and pSTAT-3Tyr705 were evaluated by immunostaining in needle biopsies of the prostate from 202 PCa patients treated by definitive therapy (105 cases) or hormonal therapy (97 cases). The correlation of two protein expression with the clinicopathologic features and the prognosis of PCa were subsequently assessed. The expression levels of pJAK-1Tyr1022/1023 and pSTAT-3Tyr705 were both positively correlated with Gleason score and clinical stage of patients with PCa. Their expression was also significantly higher in patients with biochemical (prostate-specific antigen, PSA) failure than that in those with no PSA failure (both P<0.001). In all patients, the recurrence-free survival (RFS) rates were significantly higher in those with low pJAK-1Tyr1022/1023 and pSTAT-3Tyr705 expression than that in those with high expression (both P<0.001). Moreover, for patients treated by definitive or hormonal therapy, the RFS rates in those with lower pJAK-1Tyr1022/1023 (P<0.001 and 0.012, respectively) and pSTAT-3Tyr705 expression (P<0.001 and 0.015, respectively) were significantly higher than in those with higher expression. Cox multivariate analysis showed that the expression levels of pJAK-1Tyr1022/1023 (P=0.002) and pSTAT-3Tyr705 (P=0.005) were prognostic factors for PCa in addition to extraprostatic extension (P=0.026) and Gleason score (P=0.018). The results of pJAK-1Tyr1022/1023 and pSTAT-3Tyr705 immunostainings in needle-biopsy specimens are prognostic factors for PCa. C1 [Liu, Xingyan] Guangdong Medical University, Cancer Research Institute, 523808 Dongguan, Guangdong, China. [He, Zhiwei] Guangdong Medical University, Cancer Research Institute, 523808 Dongguan, Guangdong, China. [Li, Cai-hong] Key Laboratory of Medical Molecular Activity Research, 523808 Dongguan, Guangdong, China. [Huang, Guoliang] Guangdong Medical University, Cancer Research Institute, 523808 Dongguan, Guangdong, China. [Ding, Congcong] Guangdong Medical University, Cancer Research Institute, 523808 Dongguan, Guangdong, China. [Liu, Hong] Guangdong Medical University, 523808 Dongguan, Guangdong, China. RP Liu, H (reprint author), Guangdong Medical University, 523808 Dongguan, China. EM alison0428@126.com CR Kehinde EO, Maghrebi MA, Anim JT, 2008, The importance of determining the aggressiveness of prostate cancer using serum and tissue molecular markers. Can J Urol 15:3967–74 Hochreiter WW, 2008, The issue of prostate cancer evaluation in men with elevated prostate-specific antigen and chronic prostatitis. Andrologia 40:130–3 Loeb S, Catalona WJ, 2008, What to do with an abnormal PSA test. Oncologist 13:299–305 Bolli R, Dawn B, Xuan YT, 2003, Role of the JAK-STAT pathway in protection against myocardial ischemia/reperfusion injury. Trends Cardiovasc Med 13:72–9 Booz GW, Day JN, Baker KM, 2002, Interplay between the cardiac renin angiotensin system and JAK-STAT signaling: role in cardiac hypertrophy, ischemia/reperfusion dysfunction, and heart failure. J Mol Cell Cardiol 34:1443–53 Gross ER, Hsu AK, Gross GJ, 2006, The JAK/STAT pathway is essential for opioid-induced cardioprotection: JAK2 as a mediator of STAT-3, Akt, and GSK-3β. Am J Physiol Heart Circ Physiol 291:827–34 Kandalam U, Clark MA. Angiotensin II activates JAK2/STAT-3 pathway and induces interleukin-6 production in cultured rat brainstem astrocytes, Regul Pept, DOI 10.1016/j.regpep.2009.09.001 Darnell JE Jr, 1997, STATs and gene regulation. Science 277:1630–5 Levy DE, Darnell JE Jr, 2002, Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol 36:51–62 Calo V, Migliavacca M, Bazan V, 2003, STAT proteins: from normal control of cellular events to tumorigenesis. J Cell Physiol 197:157–68 Barton BE, Karras JG, Murphy TF, Barton A, Huang HF, 2004, Signal transducer and activator of transcription 3, STAT3, activation in prostate cancer: Direct STAT3 inhibition induces apoptosis in prostate cancer lines. Mol Cancer Ther 3:11–20 Agarwal C, Tyagi A, Kaur M, 2007, Silibinin inhibits constitutive activation of Stat3, and causes caspase activation and apoptotic death of human prostate carcinoma DU145 cells. Carcinogenesis 28:1463–70 Tam L, McGlynn LM, Traynor P, 2007, Expression levels of the JAK/STAT pathway in the transition from hormone-sensitive to hormone-refractory prostate cancer. Br J Cancer 97:378–83 Schroder FH, Hermanek P, Denis L, 1992, classification of prostate cancer. Prostate Suppl 4:129–38 Gleason DF, Mellinger GT, 1974, Prediction of prognosis for prostatic adenocarcinoma by combined histological grading and clinical staging. J Urol 111:58–64 Wang L, Guojun Wu, Lei Yu, 2006, Inhibition of CD147 expression reduces tumor cell invasion in human prostate cancer cell line via RNA interference. Cancer Biol Ther 5:608–14 Pulukuri S, Patel J, Estes N, 2007, Matrix metalloproteinase-1, MMP-1, expression and biological signiWcance in the progression of prostate cancer, abstract). In: American Association for Cancer Research Annual Meeting: proceedings 14–18 Kakehi Y, 2003, Watchful waiting as a treatment option for localized prostate cancer in the PSA era. Jpn J Clin Oncol 33:1–5 Lacronique V, Boureux A, Valle VD, 1997, A TEL-JAK2 fusion protein with constitutive kinase activity in human leukemia. Science 278:1309–12 Alvarez JV, Greulich H, Sellers WR, 2006, Signal transducer and activator of transcription 3 is required for the oncogenic effects of non–small-cell lung cancer–associated mutations of the epidermal growth factor receptor. Cancer Res 66:3162–8 Xiong H, Zhang ZG, Tian XQ, 2008, Inhibition of JAK1, 2/ STAT3 signaling induces apoptosis, cell cycle arrest, and reduces tumor cell invasion in colorectal cancer cells. Neoplasia 10:287– 97 Ram PT, Horvath CM, Lyengar R, 2000, Stat3-mediated transformation of NIH-3T3 cells by the constitutively active Q205L Gαo protein. Science 287:142–4 Tu Y, Zhong Y, Fu J. Activation of JAK/STAT signal pathway predicts poor prognosis of patients with gliomas. Med Oncol. In press Wang T, Niu G, Kortylewski M, 2004, Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat Med 10:48–54 Cozzi PJ, Wang J, Delprado W et al, 2005, MUC1, MUC2, MUC4, MUC5AC and MUC6 expression in the progression of prostate cancer. Clin Exp Metastasis 22:565–73 Seitz C, Djavan B, 2006, Biological markers of prostate cancer. Ann Urol, Paris, 40:329–35 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 17 EP 23 DI 10.1007/s12253-011-9410-y PG 7 ER PT J AU Kacar, A Arikok, TA Kokenek Unal, DT Onder, E Hucumenoglu, S Alper, M AF Kacar, Ayper Arikok, Turker Ata Kokenek Unal, Dilay Tuba Onder, Evrim Hucumenoglu, Sema Alper, Murat TI Stromal Expression of CD34, α-Smooth Muscle Actin and CD26/DPPIV in Squamous Cell Carcinoma of the Skin: A Comparative Immunohistochemical Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD34; α-SMA; CD26; Squamous cell carcinoma; Tumor stroma ID CD34; α-SMA; CD26; Squamous cell carcinoma; Tumor stroma AB Invasion pathogenesis is one of the most complicated issues in the literature. There are numerous studies concerning the tumor markers implicated in the preinvasiveinvasive tumor sequence. Despite ample studies on the invasion pathogenesis of cutaneous melanomas, there is limited and dispersed work presently available on nonmelanoma skin cancer. The vast knowledge in the literature concerning this issue in squamous cell carcinoma comes mostly from the studies of the oral cavity, esophagus, larynx, and cervix. In this study, we investigated tumor-free neighboring stroma and tumor stroma in squamous cell carcinomas (SCCs) of the skin as well as keratoacanthomas (KAs) with respect to the presence of stromal CD34-positive (CD34+) fibrocytes and α-smooth muscle actin-positive (α-SMA+) myofibroblasts using seborrheic keratosis (SKs) and nontumoral skin samples as controls. We also evaluated the stromal expression pattern of CD26/DPPIV (CD26), a tumor suppressor gene product that also has immunoregulatory properties. Immunohistochemistry was performed on samples of 31 SCC, 8 KA, 15 SK and 10 non-tumoral skin samples. Peri-tumoral stroma from resection margins was also evaluated. We found that CD34 and α-SMA demonstrated significantly different staining between benign and malignant squamous skin lesions consisting of a loss of CD34+ fibrocytes paralleled by a gain of α-SMA+ myofibroblasts in malignant tumor stroma. Additionally, it was shown that CD26 expression was lower in tumor stroma when compared to that of tumor neighboring stroma. However, we concluded that this finding may be attributable to the solar elastosis areas in the peritumoral tissue, which shows diffuse strong positivity for this marker. C1 [Kacar, Ayper] Ankara Children’s Hematology and Oncology Hospital, Pathology DepartmentAnkara, Turkey. [Arikok, Turker Ata] Diskapi Yildirim Beyazit Education and Research Hospital, Department of PathologyAnkara, Turkey. [Kokenek Unal, Dilay Tuba] Diskapi Yildirim Beyazit Education and Research Hospital, Department of PathologyAnkara, Turkey. [Onder, Evrim] Diskapi Yildirim Beyazit Education and Research Hospital, Department of PathologyAnkara, Turkey. [Hucumenoglu, Sema] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Pathology DivisionAnkara, Turkey. [Alper, Murat] Diskapi Yildirim Beyazit Education and Research Hospital, Department of PathologyAnkara, Turkey. RP Kacar, A (reprint author), Ankara Children’s Hematology and Oncology Hospital, Pathology Department, Ankara, Turkey. EM ayperkacar@yahoo.com CR Kojc N, Zidar N, Vodopivec B et al, 2005, Expression of CD 34, alpha-smooth muscle actin, and transforming growth factor beta-1 in squamous intraepithelial lesions and squamous cell carcinoma of larynx and hypopharynx. Hum Pathol 36(1):16–21 Parrott JA, Nilsson E, Mosher R et al, 2001, Stromal epithelial interaction in the progression of the ovarian cancer: influence and source of tumor stromal cells. Mol Cell Endocrinol 175:29–39 Dvorak HF, 1986, Tumors: wounds that do not heal. N Engl J Med 315:1650–1659 Barth PJ, Zu Schweinsberg TS, Ramaswamy A, Moll R, 2004, CD 34 + fibrocytes, alpha-smooth muscle antigen-positive myofibroblasts, and CD 117 expression in the stroma of invasive squamous cell carcinomas of the oral cavity pharynx and larynx. Virchows Arch 444(3):231–234 Barth PJ, Ebrahimsade S, Hellinger A et al, 2002, CD34+ fibrocytes in neoplastic and inflammatory pancreatic lesions. Virchows Arch 440:128–133 Barth PJ, Ebrahimsade S, Ramaswamy A et al, 2002, CD34+ fibrocytes in invasive ductal carcinoma, ductal carcinoma in situ, and benign lesions. Virchows Arch 440:298–303 Barth PJ, Ramaswamy A, Moll R et al, 2002, CD34+ fibrocytes in normal cervical stroma, cervical intraepithelial neoplasia III, and invasive squamous cell carcinoma of the cervix uteri. Virchows Arch 441:564–568 Chauhan H, Abraham A, Philips JR et al, 2003, There is more than one kind ofmyofibroblast: analysis of CD34 expression in benign, in situ, and invasive breast lesions. J Clin Pathol 56:271–276 Kirchmann TT, Pietro VG, Smoller BR, 1994, CD34 staining pattern distinguishes basal cell carcinoma from trichoepithelioma. Arch Dermatol 130:589–592 Ramaswamy A, Moll R, Barth PJ, 2003, CD34+ fibrocytes in tubular carcinomas and radical scars of the breast. Virchows Arch 443:536–540 Havre PA, Abe M, Urasaki Y et al, 2008, The role of CD26/ dipeptidyl peptidase IV in cancer. Front Biosci 13:1634–1645 Lambeir AM, Durinks C, Scharpe S, De Meester I, 2003, Dipeptidyl-peptidase IV from bench to bedside; an update on structural properties, functions, and clinical aspects of the enzyme DPP IV. Crit Rev Clin Lab Sci 40(3):209–294 Lima MA, Gontijo VA, Schmitt FCL, 1998, CD26, dipeptidyl aminopeptidase IV, expression in normal and diseased human thyroid glands. Endocr Pathol 9:43–52 Kajiyama H, Shibata K, Terauchi M et al, 2006, Involvement of DPPIV/CD26 in epithelial morphology and suppressed invasive ability in ovarian carcinoma cells. Ann N YAcad Sci 1086:233–240 Tanaka T, Umeki K, Yamamoto I et al, 1995, CD26, dipeptidyl peptidase IV/DPP IV, as a novel molecular marker for differentiated thyroid carcinoma. Int J Cancer 64:326–331 Wilson MJ, Ruhrland AR, Quast BJ et al, 2000, Dipeptidyl peptidase IV activities are elevated in prostate cancers and adjacent benign hyperplastic glands. J Androl 21:220–226 Kikkawa F, Kajiyama H, Shibata K et al, 2005, Dipeptidyl peptidase in tumor progression. Biochim Biophys Acta 1751:45–51 Wesley UV, Albino AP, Tiwari S et al, 1999, A role for dipeptidyl peptidase IVin suppressing the malignant phenotype of melanocytic cells. J Exp Med 190:311–322 Moehrle MC, Schlagenhauff BE, Klessen C et al, 1995, Aminopeptidase M and dipeptidyl peptidase IV activity in epithelial skin tumours: a histochemical study. J Cutan Pathol 22, 3):241–247 Goscinski MA, Suo ZH, Nesland JM et al, 2008, Dipeptidyl peptidase IV expression in cancer and stromal cells of human esophageal cell carcinomas, adenocarcinomas and squamous cell carcinoma cell lines. APMIS 116:823–831 Tsuzuki S, Ota H, Hayama M et al, 2001, Proliferation of α- smooth muscle actin-containing stromal cells, myofibroblasts, in the lamina propria subjacent to intraepithelial carcinoma of the esophagus. Scand J Gastroenterol 36(1):86–91 Zeisberg EM, Potenta S, Xie L et al, 2007, Discovery of endothelial to mesenchymal transition as a source for carcinomaassociated fibroblasts. Cancer Res 67(21):10123–10128 Ohtani H, 1998, Stromal reaction in cancer tissue: pathophysiologic significance of matrix degrading enzymes in relation to matrix turnover and immune/inflammatory reaction. Pathol Int 48:1–9 Pupa SM, Menard S, Forti S et al, 2002, New insights into the role of extracellular matrix during tumour onset and progression. J Cell Physiol 192:259–267 Kalluri R, Zeisberg M, 2006, Fibroblasts in cancer. Nat Rev Cancer 6:392–401 Gabbiani G, 1998, The cellular derivation and the life span of the myofibroblast. Pathol Res Pract 192:708–711 Ruiter D, Bogenrieder T, Elder D et al, 2002, Melanoma-stroma interactions: structural and functional aspects. Lancet Oncol 3:35– 43 Bermpohl F, Loster K, Reutter W et al, 1998, Rat dipeptidyl peptidase IV, DPP IV, exhibits endopeptidase activity with specificity for denatured fibrillar collagens. FEBS Lett 428, 3):152–156 Ghersi G, Dong H, Goldstein LA et al, 2002, Regulation of fibroblast migration on collagenous matrix by a cell surface peptidase complex. J Biol Chem 277(32):29231–29241 Van den Oord JJ, 1998, Expression of CD26/dipeptidyl-petidase IV in benign and malignant pigment-cell lesions of the skin. Br J Dermatol 138(4):615–621 Bauvois B, 1988, A collagen-binding glycoprotein on the surface of mouse fibroblasts is identified as dipeptidyl peptidase IV. Biochem J 252:723–731 Loster K, Zeilinger K, Schuppan D, 1995, The cysteine rich region of dipeptidyl peptidase IV, CD26, is the collagen binding site. Biochem Biophys Res Commun 217(1):341–348 Piazza GA, Callanan HM, Mowery J et al, 1989, Evidence for a role of dipeptidyl peptidase IV in fibronectin-mediated interactions of hepatocytes with extracellular matrix. Biochem J 262:327–334 Chen W-T, 1992, Membrane proteases: role in tissue remodelling and tumor invasion. Curr Opin Cell Biol 4:802–809 Thielitz A, Vetter RW, Schultze B et al, 2008, Inhibitors of dipeptidyl peptidase IV-like activity mediate antifibrotic effects in normal and keloid derived skin fibroblasts. J Invest Dermatol 128, 4):855–866 Wang XM, Yu DM, McCaughan GWet al, 2006, Extra-enzymatic roles of DPIV and FAP in cell adhesion and migration on collagen and fibronectin. Adv Exp Med Biol 575:213–222 Jost MM, Lamerz J, Tammen H et al, 2009, Dipeptidyl peptidase 4, DPP4, inhibitors represent a novel class of oral antihyperglycemic agents. The complete pharmacological profile of these proteases. Biochem Pharmacol 77(2):228–237 Campbell TB, Hangoc G, Liu Yet al, 2007, Inhibition of CD26 in human cord blood CD34+ cells enhances their engraftment of nonobese diabetic/severe combined immunodeficiency mice. Stem Cells Dev 16(3):347–354 Ghersi G, Zhao Q, Salamone M, Yeh Y, 2006, The protease complex consisting of dipeptidyl peptidase IV and seprase plays a role in the migration and invasion of human endothelial cells in collagenous matrices. Cancer Res 66(9):4652–4661 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 25 EP 31 DI 10.1007/s12253-011-9412-9 PG 7 ER PT J AU Kafousi, M Vrekoussis, Th Tsentelierou, E Pavlakis, K Navrozoglou, I Dousias, V Sanidas, E Tsiftsis, D Georgoulias, V Stathopoulos, NE AF Kafousi, Maria Vrekoussis, Thomas Tsentelierou, Eleftheria Pavlakis, Kitty Navrozoglou, Iordanis Dousias, Vassilios Sanidas, Elias Tsiftsis, Dimitrios Georgoulias, Vassilios Stathopoulos, N Efstathios TI Immunohistochemical Study of the Angiogenetic Network of VEGF, HIF1α, VEGFR-2 and Endothelial Nitric Oxide Synthase (eNOS) in Human Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Angiogenesis; Breast Cancer; eNOS; VEGF ID Angiogenesis; Breast Cancer; eNOS; VEGF AB Background: The role of Nitric Oxide (NO) in angiogenesis has not been fully clarified yet. A dual role for NO, either inductive or inhibitory, has been proposed on the basis of different effects that high or low concentrations of NO may exert on the angiogenic process. Additionally, it has been referred that NO may induce VEGF production, while VEGF may induce NO production via up-regulation of the endothelial nitric oxide synthase (eNOS), the two pathways being reverse. The aim of the current study was to investigate the expression of key molecules involved in these opposite pathways in primary breast cancer. Methods: Representative tumor samples from 242 patients with early-stage breast cancer (invasive ductal breast carcinomas) were investigated for the expression of VEGF, VEGFR-2, HIF1α, iNOS, and eNOS using immunohistochemistry. Results: Endothelial NOS was found in 159 cases, VEGF in 131 cases, HIF-1α in 139 cases, VEGFR2 in 185 cases and inducible NOS (iNOS) in 22 cases. There was a significant correlation between the expression of VEGF and VEGFR-2, eNOS and VEGF, eNOS and VEGFR-2, eNOS and HIF1α. No statistically significant correlation was found between iNOS and the rest of the studied molecules. Conclusions: In breast cancer cases, the major molecules regulating NO and VEGF production can be co-expressed in the individual carcinomas implying a possibility for the relevant pathways to be active; however appropriate functional experiments remain to be conducted to prove such a hypothesis. C1 [Kafousi, Maria] Medical School of the University of Crete, Department of Pathology, Voutes, 70003 Heraklion, Crete, Greece. [Vrekoussis, Thomas] Medical School of the University of Crete, Department of Pathology, Voutes, 70003 Heraklion, Crete, Greece. [Tsentelierou, Eleftheria] Medical School of the University of Crete, Department of Pathology, Voutes, 70003 Heraklion, Crete, Greece. [Pavlakis, Kitty] University of Athens, Medical School, Department of PathologyAthens, Greece. [Navrozoglou, Iordanis] Ioannina University Hospital, Department of Obstetrics and GynecologyIoannina, Epirus, Greece. [Dousias, Vassilios] Ioannina University Hospital, Department of Obstetrics and GynecologyIoannina, Epirus, Greece. [Sanidas, Elias] Medical School, University of Crete, Department of Surgical OncologyHeraklion, Crete, Greece. [Tsiftsis, Dimitrios] Medical School, University of Crete, Department of Surgical OncologyHeraklion, Crete, Greece. [Georgoulias, Vassilios] Medical School, University of Crete, Department of Medical OncologyHeraklion, Crete, Greece. [Stathopoulos, N Efstathios] Medical School of the University of Crete, Department of Pathology, Voutes, 70003 Heraklion, Crete, Greece. RP Stathopoulos, NE (reprint author), Medical School of the University of Crete, Department of Pathology, 70003 Heraklion, Greece. EM stath@med.uoc.gr CR Fox SB, Generali DG, Harris AL, 2007, Breast tumour angiogenesis. Breast Canc Res 9:216 Kimura H, Esumi H, 2003, Reciprocal regulation between nitric oxide and vascular endothelial growth factor in angiogenesis. Acta Biochim Pol 50:49–59 Amano K, Matsubara H, Iba O et al, 2003, Enhancement of ischemia-induced angiogenesis by eNOS overexpression. Hypertension 41:156–62 Fukumura D, Gohongi T, Kadambi A et al, 2001, Predominant role of endothelial nitric oxide synthase in vascular endothelial growth factor-induced angiogenesis and vascular permeability. Proc Natl Acad Sci USA 98:2604–9 Sandau KB, Fandrey J, Brune B, 2001, Accumulation of HIF- 1alpha under the influence of nitric oxide. Blood 97:1009–15 Sandau KB, Faus HG, Brune B, 2000, Induction of hypoxiainducible- factor 1 by nitric oxide is mediated via the PI 3 K pathway. Biochem Biophys Res Commun 278:263–7 Ghiso N, Rohan RM, Amano S, Garland R, Adamis AP, 1999, Suppression of hypoxia-associated vascular endothelial growth factor gene expression by nitric oxide via cGMP. Invest Ophthalmol Vis Sci 40:1033–9 Choi KS, Bae MK, Jeong JW, Moon HE, Kim KW, 2003, Hypoxia-induced angiogenesis during carcinogenesis. J Biochem Mol Biol 36:120–7 TsuzukiY, FukumuraD, Oosthuyse B,Koike C, Carmeliet P, Jain RK, 2000, Vascular endothelial growth factor, VEGF, modulation by targeting hypoxia-inducible factor-1alpha–>hypoxia response element–> VEGF cascade differentially regulates vascular response and growth rate in tumors. Cancer Res 60:6248–52 Chou MT, Wang J, Fujita DJ, 2002, Src kinase becomes preferentially associated with the VEGFR, KDR/Flk-1, following VEGF stimulation of vascular endothelial cells. BMC Biochem 3:32 Duval M, Le Boeuf F, Huot J, Gratton JP, 2007, Src-mediated phosphorylation of Hsp90 in response to vascular endothelial growth factor, VEGF, is required for VEGF receptor-2 signaling to endothelial NO synthase. Mol Biol Cell 18:4659–68 He H, Venema VJ, Gu X, Venema RC, Marrero MB, Caldwell RB, 1999, Vascular endothelial growth factor signals endothelial cell production of nitric oxide and prostacyclin through flk-1/KDR activation of c-Src. J Biol Chem 274:25130–5 Hood JD, Meininger CJ, Ziche M, Granger HJ, 1998, VEGF upregulates ecNOS message, protein, and NO production in human endothelial cells. Am J Physiol 274:H1054–8 Shen BQ, Lee DY, Zioncheck TF, 1999, Vascular endothelial growth factor governs endothelial nitric-oxide synthase expression via a KDR/Flk-1 receptor and a protein kinase C signaling pathway. J Biol Chem 274:33057–63 Beleslin-Cokic BB, Cokic VP, Yu X, Weksler BB, Schechter AN, Noguchi CT, 2004, Erythropoietin and hypoxia stimulate erythropoietin receptor and nitric oxide production by endothelial cells. Blood 104:2073–80 Loibl S, Strank C, von Minckwitz G et al, 2005, Immunohistochemical evaluation of endothelial nitric oxide synthase expression in primary breast cancer. Breast 14:230–5 Mortensen K, Christensen IJ, Nielsen HJ, Hansen U, Larsson LI, 2004, High expression of endothelial cell nitric oxide synthase in peritumoral microvessels predicts increased disease-free survival in colorectal cancer. Cancer Lett 216:109–14 Mortensen K, Holck S, Christensen IJ et al, 1999, Endothelial cell nitric oxide synthase in peritumoral microvessels is a favorable prognostic indicator in premenopausal breast cancer patients. Clin Cancer Res 5:1093–7 Nakamura Y, Yasuoka H, Tsujimoto M et al, 2006, Nitric oxide in breast cancer: induction of vascular endothelial growth factor-C and correlation with metastasis and poor prognosis. Clin Cancer Res 12:1201–7 Kwan ML, Kushi LH, Weltzien E et al, 2009, Epidemiology of breast cancer subtypes in two prospective cohort studies of breast cancer survivors. Breast Cancer Res 11:R31 Kalmanti L, Dampaki K, Dimitriou H, Stiakaki E, Chaniotis V, Stathopoulos E, 2005, A morphometric approach for the evaluation of angiogenesis in acute lymphoblastic leukemia of childhood. Leuk Res 29:673–7 Aebersold DM, Burri P, Beer KT et al, 2001, Expression of hypoxia-inducible factor-1alpha: a novel predictive and prognostic parameter in the radiotherapy of oropharyngeal cancer. Cancer Res 61:2911–16 Nakopoulou L, Stefanaki K, Panayotopoulou E et al, 2002, Expression of the vascular endothelial growth factor receptor-2/ Flk-1 in breast carcinomas: correlation with proliferation. Hum Pathol 33:863–70 Takahashi Y, Tucker SL, Kitadai Y et al, 1997, Vessel counts and expression of vascular endothelial growth factor as prognostic factors in node-negative colon cancer. Arch Surg 132:541–6 Meunier-Carpentier S, Dales JP, Djemli A et al, 2005, Comparison of the prognosis indication of VEGFR-1 and VEGFR-2 and Tie2 receptor expression in breast carcinoma. Int J Oncol 26:977–84 Mylona E, Alexandrou P, Giannopoulou I et al, 2007, The prognostic value of vascular endothelial growth factors, VEGFs)-A and -B and their receptor, VEGFR-1, in invasive breast carcinoma. Gynecol Oncol 104:557–63 Trastour C, Benizri E, Ettore F et al, 2007, HIF-1alpha and CA IX staining in invasive breast carcinomas: prognosis and treatment outcome. Int J Cancer 120:1451–8 Hayes DF, Miller K, Sledge G, 2007, Angiogenesis as targeted breast cancer therapy. Breast 16(Suppl 2):S17–19 Namba T, Koike H, Murakami K et al, 2003, Angiogenesis induced by endothelial nitric oxide synthase gene through vascular endothelial growth factor expression in a rat hindlimb ischemia model. Circulation 108:2250–7 Duenas-Gonzalez A, Isales CM, del Mar Abad-Hernandez M, Gonzalez-Sarmiento R, Sangueza O, Rodriguez-Commes J, 1997, Expression of inducible nitric oxide synthase in breast cancer correlates with metastatic disease. Mod Pathol 10:645–9 Vleugel MM, Greijer AE, Shvarts A et al, 2005, Differential prognostic impact of hypoxia induced and diffuse HIF-1alpha expression in invasive breast cancer. J Clin Pathol 58:172–7 Lundgren K, Holm C, Landberg G, 2007, Hypoxia and breast cancer: prognostic and therapeutic implications. Cell Mol Life Sci 64:3233–47 Lechner M, Lirk P, Rieder J, 2005, Inducible nitric oxide synthase, iNOS, in tumor biology: the two sides of the same coin. Semin Cancer Biol 15:277–89 Loibl S, Buck A, Strank C et al, 2005, The role of early expression of inducible nitric oxide synthase in human breast cancer. Eur J Cancer 41:265–71 Vakkala M, Paakko P, Soini Y, 2000, eNOS expression is associated with the estrogen and progesterone receptor status in invasive breast carcinoma. Int J Oncol 17:667–71 Sumi D, Ignarro LJ, 2003, Estrogen-related receptor alpha 1 up-regulates endothelial nitric oxide synthase expression. Proc Natl Acad Sci USA 100:14451–6 Loibl S, Bratengeier J, Farines Vet al, 2006, Investigations on the inducible and endothelial nitric oxide synthases in human breast cancer cell line MCF-7 - estrogen has an influence on e-NOS, but not on i-NOS. Pathol Res Pract 202:1–7 Gavin KM, Seals DR, Silver AE, Moreau KL, 2009, Vascular endothelial estrogen receptor alpha is modulated by estrogen status and related to endothelial function and endothelial nitric oxide synthase in healthy women. J Clin Endocrinol Metab 94:3513–20 Oishi A, Ohmichi M, Takahashi K et al, 2004, Medroxyprogesterone acetate attenuates estrogen-induced nitric oxide production in human umbilical vein endothelial cells. Biochem Biophys Res Commun 324:193–8 Zerr-Fouineau M, Chataigneau M, Blot C, Schini-Kerth VB, 2007, Progestins overcome inhibition of platelet aggregation by endothelial cells by down-regulating endothelial NO synthase via glucocorticoid receptors. FASEB J 21:265–73 Chi JT, Wang Z, Nuyten DS et al, 2006, Gene expression programs in response to hypoxia: cell type specificity and prognostic significance in human cancers. PLoS Med 3:e47 Milani M, Harris AL, 2008, Targeting tumour hypoxia in breast cancer. Eur J Cancer 44:2766–73 Kurebayashi J, Otsuki T, Moriya T, Sonoo H, 2001, Hypoxia reduces hormone responsiveness of human breast cancer cells. Jpn J Cancer Res 92:1093–101 Yamamoto Y, Ibusuki M, Okumura Y et al, 2008, Hypoxiainducible factor 1alpha is closely linked to an aggressive phenotype in breast cancer. Breast Cancer Res Treat 110:465–75 Laughner E, Taghavi P, Chiles K, Mahon PC, Semenza GL, 2001, HER2, neu, signaling increases the rate of hypoxiainducible factor 1alpha, HIF-1alpha, synthesis: novel mechanism for HIF-1-mediated vascular endothelial growth factor expression. Mol Cell Biol 21:3995–4004 Giatromanolaki A, Koukourakis MI, Simopoulos C et al, 2004, c-erbB-2 related aggressiveness in breast cancer is hypoxia inducible factor-1alpha dependent. Clin Cancer Res 10:7972–7 Traina TA, Rugo HS, Dickler M, 2007, Bevacizumab for advanced breast cancer. Hematol Oncol Clin North Am 21:303– 19 Marty M, Pivot X, 2008, The potential of anti-vascular endothelial growth factor therapy in metastatic breast cancer: Clinical experience with anti-angiogenic agents, focusing on bevacizumab. Eur J Cancer 44:912–20 Patiar S, Harris AL, 2006, Role of hypoxia-inducible factor- 1alpha as a cancer therapy target. Endocr Relat Cancer 13(Suppl 1):S61–75 Melillo G, 2007, Targeting hypoxia cell signaling for cancer therapy. Cancer Metastasis Rev 26:341–52 Coulet F, Nadaud S, Agrapart M, Soubrier F, 2003, Identification of hypoxia-response element in the human endothelial nitric-oxide synthase gene promoter. J Biol Chem 278:46230–40 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 33 EP 41 DI 10.1007/s12253-011-9413-8 PG 9 ER PT J AU Varoczy, L Zilahi, E Gyetvai, Kajtar, B Gergely, L Sipka, S Illes, AF Varoczy, Laszlo Zilahi, Erika Gyetvai, Agnes Kajtar, Bela Gergely, Lajos Sipka, Sandor Illes, Arpad TI Fc-Gamma-Receptor IIIa Polymorphism and Gene Expression Profile Do Not Predict the Prognosis in Diffuse Large B-cell Lymphoma Treated with R-CHOP Protocol SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Diffuse large B-cell lymphoma; Rituximab; Fc-gamma-receptor IIIa polymorphism; Gene expression profile; Treatment response; Survival ID Diffuse large B-cell lymphoma; Rituximab; Fc-gamma-receptor IIIa polymorphism; Gene expression profile; Treatment response; Survival AB The addition of rituximab to conventional chemotherapy has significantly improved the treatment outcome in diffuse large B-cell lymphoma. However, differences in treatment response and survival data can be observed independently from the International Prognostic Index scores. The current study evaluated the impact of Fc-gamma-receptor IIIa polymorphism and gene expression profile on the response of DLBCL patients to R-CHOP therapy as well as on their survival results. Fifty-one patients were involved, thirty-two females, nineteen males, their median age was 53.1 years. The FCGR3A polymorphism at the 158. amino acid position determined with PCR method showed the following results: VV: 12 cases (23.5%), VF: 29 cases (56.8%) and FF: 10 cases (19.6%), respectively. There was no significant difference between the treatment responses of the three groups. The event-free survival data were less favorable in the F-allele carriers than in V/V homozygous patients, but without any significancy, and the overall survival curves were almost the same. As for the gene expression profile, 20 patients’ biopsy specimens showed germinal center and 31 showed non-germinal center characteristics. Treatment results did not differ from each other in the two groups. Both the event-free and the overall survival data were more favorable in the GC group, however the differences were not significant. Our results contest the predictive value of Fcgamma-receptor IIIa polymorphism and gene expression profile in diffuse large B-cell lymphoma. C1 [Varoczy, Laszlo] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. 22., 4032 Debrecen, Hungary. [Zilahi, Erika] University of Debrecen, Medical and Health Science Center, Regional Laboratory for Immunology, Moricz Zs. 22., 4032 Debrecen, Hungary. [Gyetvai, Agnes] University of Debrecen, Medical and Health Science Center, Regional Laboratory for Immunology, Moricz Zs. 22., 4032 Debrecen, Hungary. [Kajtar, Bela] University of Pecs, Department of PathologyPecs, Hungary. [Gergely, Lajos] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. 22., 4032 Debrecen, Hungary. [Sipka, Sandor] University of Debrecen, Medical and Health Science Center, Regional Laboratory for Immunology, Moricz Zs. 22., 4032 Debrecen, Hungary. [Illes, Arpad] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. 22., 4032 Debrecen, Hungary. RP Varoczy, L (reprint author), University of Debrecen, Medical and Health Center, 3rd Department of Medicine, 4032 Debrecen, Hungary. EM laszlo.varoczy@gmail.com CR Coiffier B, Reyes F, Groupe d’Etude des Lymphomes de l’Adulte, 2005, Best treatment of agressive non-Hodgkin’s lymphoma: a French perspective. Oncology, Williston Park, 19:7–15 Nitsu N, 2010, Current treatment strategy of diffuse large B-cell lymphomas. Int J Hematol 92:231–237 Sehn LH, Berry B, Chhanabhai M et al, 2007, The revised International Prognostic Index is a better predictor of outcome than the standard IPI for patients with diffuse large B-cell lymphoma treated with R-CHOP. Blood 109:1857–1861 Lossos IS, Morgensztern D, 2006, Prognostic biomarkers in diffuse large B-cell lymphoma. J Clin Oncol 24:995–1007 Shan D, Ledbetter JA, Press OW, 1998, Apoptosis of malignant human B cells by ligation of CD20 with monoclonal antibodies. Blood 91:1644–1652 Villamor N, Montserrat E, Colomer D, 2003, Mechanism of action and resistance to monoclonal antibody therapy. Sem Oncol 30:424–433 Koene HR, Kleijer M, Algra J et al, 1997, FcgRIIIa-158 V/F polymorphism influences the binding of IgG by natural killer cell FcgRIIIa, idependently of the FcgRIIIa-48 L/R/H phenotype. Blood 1997:1109–1114 Hatjiharissi E, Lian X, Santos DD et al, 2007, Increased natural killer cell expression of CD16, augmented binding and ADCC activity to rituximab among individuals expressing the FcgRIIIa-158 V/Vand V/F polymorphism. Blood 110:2561–2564 Alizadeh AA, Eisen MB, Davis RE et al, 2000, Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 403:503–511 Morgenszter D, Martin MG, Lossos IS, 2007, Gene expression profiling in diffuse large B-cell lymphoma. Leuk Lymph 48:669– 682 Hans CP, Weisenburger DD, Greiner TC et al, 2004, Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood 103:275–282 Cartron G,Watier H, Golay J et al, 2004, Fromthe bench to bedside: ways to improve rituximab efficacy. Blood 104:2635–2642 Cartron G, Dacheux L, Salles G et al, 2002, Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgRIIIa gene. Blood 99:754–758 Treon SP, Hansen M, Branagan AR et al, 2005, Polymorphisms in FcgRIIIa, CD16, receptor expression are associated with clinical response to rituximab in Waldenstrom’s macroglobulinaemia. J Clin Oncol 23:474–481 Farag SS, Flinn IW, Modali R et al, 2004, Fc gamma RIIIa and Fc gamma RIIa polymorphisms do not predict response to rituximab in B-cell chronic lymphocytic leukaemia. Blood 103:1472–1474 Carlotti E, Palumbo GA, Oldani E et al, 2007, Fc gamma RIIIa and Fc gamma RIIa polymorphisms do not predict clinical outcome of follicular non-Hodgkin’s lymphoma patients treated with sequential CHOP and rituximab. Hematologica 92:1127–1130 Kim DW, Jung HD, Kim JG et al, 2006, FCGR3A gene polymorphisms may correlate with response to frontline R-CHOP therapy for diffuse large B-cell lymphoma. Blood 108:2720–2725 Sjo LD, Poulsen CB, Hansen M et al, 2007, Profiling of diffuse large B-cell lymphoma by immunohistochemistry: identification of prognostic subgroups. Eur J Haematol 79:501–507 Nyman H, Adde M, Karjalainen-Lindsberg ML et al, 2007, Prognostic impact of immunohistochemically defined germinal center phenotype in diffuse large B-cell lymphoma patients treated with immunochemotherapy. Blood 109:4930–4935 Fu K, Weisenberger DD, Choi W et al, 2008, Addition of rituximab to standard chemotherapy improves the survival of both the germinal center B-cell-like and non-germinal center B-celllike subtypes of diffuse large B-cell lymphoma. J Clin Oncol 26:4587–4594 Mounier N, Briere J, Gisselbrecht C et al, 2003, Rituximab plus CHOP, R-CHOP, overcomes bcl-2-associated resistance to chemotherapy in elderly patients with diffuse large B-cell lymphoma, DLBCL). Blood 101:4279–4284 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 43 EP 48 DI 10.1007/s12253-011-9414-7 PG 6 ER PT J AU Mahmoodi, M Nahvi, H Mahmoudi, M Kasaian, A Mohagheghi, MA Divsalar, K Nahavandian, B Jafari, A Ansarpour, B Moradi, B Aghamohammadi, A Amirzargar, AA AF Mahmoodi, Majid Nahvi, Hedayat Mahmoudi, Mahdi Kasaian, Amir Mohagheghi, Mohammad-Ali Divsalar, Kouros Nahavandian, Bijan Jafari, Abbas Ansarpour, Bita Moradi, Batoul Aghamohammadi, Asghar Amirzargar, Ali-Akbar TI HLA-DRB1,-DQA1 and -DQB1 Allele and Haplotype Frequencies in Female Patients with Early Onset Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Allele frequency; Breast cancer; Human leukocyte antigen (HLA); HLA-DRB1; HLA-DQA1; HLA-DQB1 ID Allele frequency; Breast cancer; Human leukocyte antigen (HLA); HLA-DRB1; HLA-DQA1; HLA-DQB1 AB Based on the reports, few HLA class II alleles are associated with susceptibility or protection in breast cancer. Here we investigate the association between HLA class II alleles and breast cancer in Iranian women. 100 patients with pathologically proven breast cancer who referred to Cancer Institute were randomly selected and compared with a group of 80 healthy blood donor subjects. The patients were studied in two groups, group 1 includes patients aging 40 years or younger and group 2 include patients aging over 40 years. HLA class II alleles were determined by amplification of DNA followed by HLA-typing using sequence-specific primer (SSP) for each allele. In group 1, the most frequent alleles were HLA-DQA1*0301 (P=0.002, OR=3.3) and HLA-DQB1*0302 (P=0.04, OR=2.8). In group 2, the following alleles increased significantly than those in controls including HLA-DQA1*0301 (P=0.001, OR=3.4) and HLA-DRB1*0301 (P=0.04, OR=2.3). In complete group of patients, the frequency of HLADQA1* 0301 (P=0.001, OR=3.4) and HLA-DRB1*1303 (P=0.02, OR=2.3) increased significantly than those in control group. HLA-DQA1*0505, HLA-DQA1*0101, HLA-DRB1*1301and HLA-DRB1*0101 alleles showed negative association with breast cancer. Our findings suggest that HLA-DQA1*0301 allele is mainly associated with increased risk of breast cancer including early-onset of the disease. HLA-DQA1*0505 and HLA-DRB1*1301 are involved in protection. We conclude that specific alleles of HLA class II influence breast cancer risk. C1 [Mahmoodi, Majid] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Nahvi, Hedayat] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Mahmoudi, Mahdi] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. [Kasaian, Amir] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Mohagheghi, Mohammad-Ali] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Divsalar, Kouros] Kerman University of Medical Sciences, Neuroscience Research CenterKerman, Iran. [Nahavandian, Bijan] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Jafari, Abbas] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Ansarpour, Bita] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. [Moradi, Batoul] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. [Aghamohammadi, Asghar] Tehran University of Medical Scie, Growth and Development Research CenterTehran, Iran. [Amirzargar, Ali-Akbar] Tehran University of Medical Sciences, Faculty of Medicine, Department of ImmunologyTehran, Iran. RP Amirzargar, AA (reprint author), Tehran University of Medical Sciences, Faculty of Medicine, Department of Immunology, Tehran, Iran. EM amirzara@tums.ac.ir CR Parkin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55(2):74–108 Forbes JF, 1997, The incidence of breast cancer: the global burden, public health considerations. Semin Oncol 24(Suppl 1): S1-20–S1-35 de Jong MM, Nolte IM, te Meerman GJ et al, 2002, Genes other than BRCA1 and BRCA2 involved in breast cancer susceptibility. J Med Genet 39(4):225–242 Futreal PA, Liu Q, Shattuck-Eidens D et al, 1994, BRCA1 mutations in primary breast and ovarian carcinomas. Science 7, 266(5182)):120–122 Pistillo MP, Nicolo G, Salvi S et al, 2000, Biochemical analysis of HLA class I subunits expression in breast cancer tissues. Hum Immunol 61:397–407 Salih HR, Nussler V, 2001, Commentary: immune escape versus tumor tolerance: how do tumors evade immune surveillance? Eur J Med Res 6:323–332 Dunn GP, Bruce AT, Ikeda H et al, 2002, Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 3:991–998 Forbes SA, Trowsdale J, 1997, The MHC quarterly report. Immunogenetics 50:152–159 Hosono S, Kawase T, Matsuo K et al, 2010, HLA-A alleles and the risk of cervical squamous cell carcinoma in Japanese women. J Epidemiol 20(4):295–301 Castro FA, Haimila K, Sareneva I et al, 2009, Association of HLA-DRB1, interleukin-6 and cyclin D1 polymorphisms with cervical cancer in the Swedish population–a candidate gene approach. Int J Cancer 125(8):1851–1858 Lee HW, Hahm KB, Lee JS et al, 2009, Association of the human leukocyte antigen class II alleles with chronic atrophic gastritis and gastric carcinoma in Koreans. J Dig Dis 10(4):265–271 Ishigami S, Natsugoe S, Nakajo A et al, 2008, HLA-class I expression in gastric cancer. J Surg Oncol 97(7):605–608 Gamzatova Z, Villabona L, van der Zanden H et al, 2007, Analysis of HLA class I-II haplotype frequency and segregation in a cohort of patients with advanced stage ovarian cancer. Tissue Antigens 70(3):205–213 Gogas H, Kirkwood JM, Falk CS et al, 2010, Correlation of molecular human leukocyte antigen typing and outcome in highrisk melanoma patients receiving adjuvant interferon. Cancer 116, 18):4326–4333 Chaudhuri S, Cariappa A, Tang M et al, 2000, Genetic susceptibility to breast cancer: HLA DQB*03032 and HLA DRB1*11 may represent protective alleles. Proc Natl Acad Sci U S A 97(21):11451–11454 Ghaderi A, Talei A, Gharesi-Fard B et al, 2001, HLA-DRB1 alleles and the susceptibility of Iranian patients with breast cancer. Pathol Oncol Res 7:39–41 Lavado R, Benavides M, Villar E et al, 2005, The HLA-B7 allele confers susceptibility to breast cancer in Spanish women. Immunol Lett 101(2):223–225 Cantu de Leon D, Perez-Montiel D, Villavicencio V et al, 2009, High resolution human leukocyte antigen, HLA, class I and class II allele typing in Mexican mestizo women with sporadic breast cancer: case-control study. BMC Cancer 9:48 Miller SA, Dykes DD, Polesky HE, 1989, A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16:1215–1221 Amirzargar AA, Khosravi F, Dianat SS et al, 2007, Association of HLA class II allele and haplotype frequencies with chronic myelogenous leukemia and age-at-onset of the disease. Pathol Oncol Res 13(1):47–51 Olerup O, Zetterquist H, 1992, HLA-DR typing by PCR amplification with sequence-specific primers, PCR-SSP, in 2 hours: an alternative to serological DR typing in clinical practice including donor-recipient matching in cadaveric transplantation. Tissue Antigens 39:225–235 Amirzargar A, Mytilineos J, Farjadian S et al, 2001, Human leukocyte antigen class II allele frequencies and haplotype association in Iranian normal population. Hum Immunol 62:1234–1238 McPherson K, Steel CM, Dixon JM, 2000, ABC of breast diseases. Breast cancer–epidemiology, risk factors and genetics. BMJ 321(7261):624–628 Ferlay J, Shin HR, Bray F et al, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127:2893–2917 World Cancer Report 2008.WHO, http://www.iarc.fr/en/publications/ pdfsonline/wcr/2008/index.php, 2 November 2010, date last accessed). Anderson BO, Jakesz R, 2008, Breast cancer issues in developing countries: an overview of the Breast Health Global Initiative. World J Surg 32:2578–2585 The Top 10 Causes of Death in 2004, 2008, Fact Sheet of WHO Report, World Health Organization. Harirchi I, Mousavi SM, Mohagheghi MA et al, 2009, Early detection for breast cancer in Iran. Asian Pac J Cancer Prev 10, 5):849–851 Mousavi SM, Gouya MM, Ramazani R et al, 2009, Cancer incidence and mortality in Iran. Ann Oncol 20:556–563 Baccar Harrath A, Yacoubi Loueslati B, Troudi W et al, 2006, HLA class II polymorphism: protective or risk factors to breast cancer in Tunisia? Pathol Oncol Res 12(2):79–81 Ferrera A, Olivo A, Alaez C et al, 1999, HLA-DOA1 and -DOB1 loci in Honduran women with cervical dysplasia and invasive cervical carcinoma and their relationship to human papillomavirus infection. Hum Biol 71(3):367–379 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 49 EP 55 DI 10.1007/s12253-011-9415-6 PG 7 ER PT J AU Li, X Zhang, Q He, W Meng, W Yan, J Zhang, L Zhu, X Liu, T Li, Y Bai, Z AF Li, Xun Zhang, Quanbao He, Wenting Meng, Wenbo Yan, Jun Zhang, Lei Zhu, Xiaoliang Liu, Tao Li, Yumin Bai, Zhongtian TI Low Frequency of PIK3CA Gene Mutations in Hepatocellular Carcinoma in Chinese Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; PIK3CA; Gene mutation; Hepatitis B virus ID Hepatocellular carcinoma; PIK3CA; Gene mutation; Hepatitis B virus AB PI3K/AKT constitutes an important pathway regulating the signaling of multiple biological processes and plays a critical role in carcinogenesis. PIK3CA gene missense mutations have been reported in many human cancer types. The mutation of it in hepatocellular carcinoma cases varies with different races and regions. In this study, we investigated PIK3CA mutation in Chinese hepatocellular carcinoma patients. A total 90 Chinese patients of hepatocellular carcinoma were recruited in this study. Exons 9 and 20 hotspots mutations of PIK3CA gene were detected by PCR-based DNA sequencing. Two point mutations (E542K and D549H) in exon 9 were found in only one patient (1/90; 1.11%), no mutation was found in exon 20 in any cases. 57 patients are associated with HBV infection (57/90; 63.3%), and 8 patients with HCV infection (8/90; 8.9%). The frequency of the PIK3CA mutations in hepatocellular carcinoma seems to be lower in Chinese hepatocellular carcinoma patients. These findings suggest that PI3K mutations may not play a major role in hepatic carcinogenesis in Chinese. HBV infection has close relationship with HCC in Chinese. C1 [Li, Xun] The First Hospital of Lanzhou University, The Second Department of General Surgery, 730000 Lanzhou, China. [Zhang, Quanbao] The First Hospital of Lanzhou University, The Second Department of General Surgery, 730000 Lanzhou, China. [He, Wenting] The First Hospital of Lanzhou University, The Second Department of General Surgery, 730000 Lanzhou, China. [Meng, Wenbo] The First Hospital of Lanzhou University, The Second Department of General Surgery, 730000 Lanzhou, China. [Yan, Jun] The First Hospital of Lanzhou University, The Second Department of General Surgery, 730000 Lanzhou, China. [Zhang, Lei] The First Hospital of Lanzhou University, The Second Department of General Surgery, 730000 Lanzhou, China. [Zhu, Xiaoliang] The First Hospital of Lanzhou University, The Second Department of General Surgery, 730000 Lanzhou, China. [Liu, Tao] Key Laboratory of Digestive System, 730000 Lanzhou, Gansu Province, China. [Li, Yumin] Key Laboratory of Digestive System, 730000 Lanzhou, Gansu Province, China. [Bai, Zhongtian] The First Hospital of Lanzhou University, The Second Department of General Surgery, 730000 Lanzhou, China. RP Li, Y (reprint author), Key Laboratory of Digestive System, 730000 Lanzhou, China. EM liym@lzu.edu.cn CR Parkin DM, Whelan SL, Ferlay J, et al, 2005, Cancer incidence in five continents, Vols. I to VIII, IARC Cancer Base). International Agency for Research on Cancer Zhang JY, Dai M, Wang X et al, 1998, A case-control study of hepatitis B and C virus infection as risk factors for hepatocellular carcinoma in Henan. China Int J Epidemiol 27:574–578 Osaki M, Oshimura M, Ito H, 2004, PI3K-Akt pathway: its functions and altertions in human cancer. Apoptosis 9:667–676 Samuels Y, Wang Z, Bardelli A et al, 2004, High frequency of mutations of the PIK3CA gene in human cancers. Science 23:554 Yamamoto H, Shigematsu H, Nomura M et al, 2008, PIK3CA mutations and copy number gains in human lung cancers. Cancer Res 68:6913–6921 Okudela K, Suzuki M, Kageyama S et al, 2007, PIK3CA mutation and amplification in human lung cancer. Pathol Int 57:664–671 Souglakos J, Philips J,Wang R et al, 2009, Prognostic and predictive value of common mutations for treatment response and survival in patients with metastatic colorectal cancer. Br J Cancer 101:465–472 Sartore-Bianchi A, Martini M, Molinari F et al, 2009, PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targetedmonoclonal antibodies. Cancer Res 69:1851–1857 Phillips WA, Russell SE, Ciavarell ML et al, 2006, Mutation analysis of PIK3CA and PIK3CB in esophageal cancer and Barrett’s esophagus. Int J Cancer 118:2644–2646 Mori R, Ishiguro H, Kimura M et al, 2008, PIK3CA mutation status in Japanese esophageal squamous cell carcinoma. J Surg Res 145:320–326 Kalinsky K, Jacks LM, Heguy A et al, 2009, PIK3CA mutation associates with improved outcome in breast cancer. Clin Cancer Res 15:5049–5059 Stemke-Hale K, Gonzalez-Angulo AM, Lluch A et al, 2008, An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res 68:6084–6091 Velho S, Oliveira C, Ferreira A et al, 2005, The prevalence of PIK3CA mutations in gastric and colon cancer. Eur J Cancer 41:1649–1654 Vivian L, Chi W, Tsun C et al, 2005, Mutations of PIK3CA in gastric adenocarcinoma. BMC Cancer 5:29–34 Kuo KT, Mao TL, Jones S et al, 2009, Frequent activating mutations of PIK3CA in ovarian clear cell carcinoma. Am J Pathol 174:1597–1601 Jehad A, Prashant B, Wael AH et al, 2009, PIK3CA alterations in Middle Eastern ovarian cancers. Mol Cancer 8:51–53 Lee JW, Soung YH, Kim SY et al, 2005, PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas. Oncogene 24:1477–1480 Boyault S, Rickman DS, Reynies A et al, 2007, Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology 45:42–52 Riener M, Bawohl M, Clavien PA et al, 2008, Rare PIK3CA hotspot mutations in carcinomas of the biliary tract. Genes Chromosome Canc 47:363–367 Tanaka Y, Kanai F, Tada M et al, 2006, Absence of PIK3CA hotspot mutations in hepatocellular carcinoma in Japanese patients. Oncogene 25:2950–2952 Llovet JM, Bru C, Bruix J, 1999, Prognosis of hepatocellular carcinoma: the BCLC staging classification. Semin Liver Dis 19:329–338 Kang S, Bader AG, Vogt PK, 2005, Phosphatidylinositol 3-kinase mutations identified in human cancer are oncogenic. Proc Natl Acad Sci USA 102:802–807 Ikenoue T, Kanai F, Hikiba Y et al, 2005, Functional analysis of PIK3CA gene mutations in human colorectal cancer. Cancer Res 65:4562–4567 Liu Z, Roberts TM, 2006, Human tumor mutants in the p110 subunit of PI3K. Cell Cycle 5:675–677 Gymnopoulos M, Elsliger MA, Vogt PK, 2007, Rare cancerspecific mutations in PIK3CA show gain of function. PNAS 104:5569–5574 Mohandas KM, 2004, Hepatitis B associated hepatocellular carcinoma: epidemiology, diagnosis and treatment. Hep B Annual 1:140–152 Russo MW, Jacobson IM, 2002, Hepatocellular cancer: screening, surveillance, and prevention. In: Kelsen DP, Daly JM, Kern SE et al, eds, Gastrointestinal oncology: principles and practices. Lippincott, Williams and Wilkins, Philadelphia, pp 559–568 Sun Z, Ming L, Zhu X et al, 2002, Prevention and control of hepatitis B in China. J Med Virol 67:447–450 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 57 EP 60 DI 10.1007/s12253-011-9416-5 PG 4 ER PT J AU Nikolenyi, A Uhercsak, G Csenki, M Hamar, S Csorgo, E Tanczos, E Thurzo, L Brodowicz, Th Wagnerova, M Kahan, Zs AF Nikolenyi, Aliz Uhercsak, Gabriella Csenki, Melinda Hamar, Sandor Csorgo, Erika Tanczos, Ervin Thurzo, Laszlo Brodowicz, Thomas Wagnerova, Maria Kahan, Zsuzsanna TI Tumour Topoisomerase II Alpha Protein Expression and Outcome After Adjuvant Dose-Dense Anthracycline-Based Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Anthracyclines; Adjuvant chemotherapy; Breast cancer; Dose-dense chemotherapy; Topoisomerase II alpha ID Anthracyclines; Adjuvant chemotherapy; Breast cancer; Dose-dense chemotherapy; Topoisomerase II alpha AB There is a need for the selection of those breast cancers where benefit may be attained from the addition of an anthracycline to the adjuvant chemotherapy. The expression of topoisomerase II alpha (TOP2A) protein in 3 cohorts of breast cancers treated with adjuvant dose-dense anthracycline-based chemotherapy was determined retrospectively. The TOP2A status was analysed in relation with the other standard tumour features and the outcome. TOP2A IHC results were assessable in 106 patients: with a cut-off value of 15%, 48% of the tumours were classified as TOP2A-positive. The expression of TOP2A correlated with that of Ki67 (R=0.532, p<0.001) and a high grade (p=0.04), but did not correlate with the proportion of ER- or PR-positive cells in the tumour. More tumors were TOP2A-negative among the ER- or PR-positive cancers than among the ER/PR-negative cancers (p=0.021 and p=0.002, respectively). After a median follow-up time of 64.5 months, 31 relapses (23.5%) and 23 deaths (17.4%) had occurred in 131 patients. The overall survival was longer in the TOP2A-positive cases than in the TOP2A-negative cases. The recurrence-free survival and the overall survival were significantly more favourable in the ER/PR-negative and TOP2A-positive tumours than in other subgroups. In a Cox proportional hazards model, the grade and TOP2A remained significant determinants in the ER/PR-negative subgroup. TOP2A positivity and grade 3 indicated a decrease in the risk of death with HR=0.211 (95% CI: 0.042–1.05, p=0.056) and HR=0.216 (95% CI: 0.047–0.990, p=0.048), respectively. A higher sensitivity to anthracycline-containing regimens is suggested in ER/PR-negative and TOP2A-positive cancers. C1 [Nikolenyi, Aliz] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Uhercsak, Gabriella] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Csenki, Melinda] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Hamar, Sandor] University of Szeged, Department of PathologySzeged, Hungary. [Csorgo, Erika] University of Szeged, Department of PathologySzeged, Hungary. [Tanczos, Ervin] University of Szeged, Department of Medical InformaticsSzeged, Hungary. [Thurzo, Laszlo] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Brodowicz, Thomas] University of Szeged, Department of Medical InformaticsSzeged, Hungary. [Wagnerova, Maria] Central European Cooperative Oncology GroupVienna, Austria. [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. RP Kahan, Zs (reprint author), University of Szeged, Department of Oncotherapy, 6720 Szeged, Hungary. EM kahan@onko.szote.u-szeged.hu CR Early Breast Cancer Trialists’ Collaborative Group, EBCTCG,, 2005, Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet 365:1687–1717 Slamon D, Eiermann W, Robert N et al, 2005, BCIRG 006: 2nd interim analysis phase II randomized trial comparing doxorubicin and cyclophosphamide followed by docetaxel positive early, AC®TH, with docetaxel, carboplatin and trastuzumab, TCH, in HER2 positive early breast cancer patients: BCIRG 006 study. Breast Cancer Res Treat 94:S5 [abstr. 1] Gianni L, Norton L, Wolmark N, Suter TM, Bonadonna G, Hortobagyi GN, 2009, Role of anthracyclines int he treatment of early breast cancer. J Clin Oncol 27:4798–4808 Gennari A, Sormani MP, Pronzato P, Puntoni M, Colozza M, Pfeffer U, Bruzzi P, 2008, HER2 status and efficacy of adjuvant anthracyclines in early breast cancer: a pooled analysis of randomized trials. J Natl Cancer Inst 100:14–20 Pritchard KI, Shepherd LE, O’Malley FP, Andrulis IL, Tu D, Bramwell VH, National Cancer Institute of Canada Clinical Trials Group, 2006, HER2 and responsiveness of breast cancer to adjuvant chemotherapy. N Engl J Med 354:2103–2111 Bartlett JM, Munro A, Cameron DA, Thomas J, Prescott R, Twelves CJ, 2008, Type 1 receptor tyrosine kinase profiles identify patients with enhanced benefit from anthracyclines in the BR9601 adjuvant breast cancer chemotherapy trial. J Clin Oncol 26:5027–5035 Rouzier R, Perou CM, Symmans WF, Ibrahim N, Cristofanilli M, Anderson K, Hess KR, Stec J, Ayers M, Wagner P, Morandi P, Fan C, Rabiul I, Ross JS, Hortobagyi GN, Pusztai L, 2005, Breast cancer subtypes respond differently to preoperative chemotherapy. Clin Cancer Res 11:5678–5685 Di Leo A, Biganzoli L, Claudion W, Licitra S, Pestrin M, Larsimont D, 2008, Topoisomerase II alpha as a marker predicting anthracyclines’ activity in early breast cancer patients: ready for the primetime? Eur J Cancer 44:2791–2798 Jarvinen TA, Liu ET, 2003, HER-2/neu and topoisomerase II alpha in breast cancer. Breast Cancer Res Treat 78:299–311 Isaacs RJ, Davies SL, Sandri MI, Redwood C, Wells NJ, Hickson ID, 1998, Physiological regulation of eukaryotic topoisomerase II. Biochim Biophys Acta 1400:121–137 Nielsen KV, Ejlertsen B, Moller S, Jorgensen JT, Knoop A, Knudsen H, Mouridsen HT, 2008, The value of TOP2A gene copy number variation as a biomarker in breast cancer: Update of DBCG trial 89D. Acta Oncol 47:725–734 Di Leo A, Gancberg D, Larsimont D, Tanner M, Jarvinen T, Rouas G, Dolci S, Leroy JY, Paesmans M, Isola J, Piccart MJ, 2002, HER-2 amplification and topoisomerase II alpha gene aberrations as predictive markers in node-positive breast cancer patients randomly treated either with an anthracycline-based therapy or with cyclophosphamide, methotrexate, and 5-fluorouracil. Clin Cancer Res 8:1107–1116 Knoop AS, Knudsen H, Balslev E, Rasmussen BB, Overgaard J, Nielsen KV, Schonau A, Gunnarsdottir K, Olsen KE, Mouridsen H, Ejlertsen B, Danish Breast Cancer Cooperative Group, 2005, Retrospective analysis of topoisomerase IIa amplifications and deletions as predictive markers in primary breast cancer patients randomly assigned to cyclophosphamide, methotrexate, and fluorouracil or cyclophosphamide, epirubicin, and fluorouracil: Danish Breast Cancer Cooperative Group. J Clin Oncol 23:7483–7490 O'Malley FP, Chia S, Tu D, Shepherd LE, Levine MN, Bramwell VH, Andrulis IL, Pritchard KI, 2009, Topoisomerase II alpha and responsiveness of breast cancer to adjuvant chemotherapy. J Natl Cancer Inst 101:644–650 Harris LN, Broadwater G, Abu-Khalaf M, Cowan D, Thor AD, Budman D, Cirrincione CT, Berry DA, Winer EP, Hudis CA, Hayes DF, Friedman P, Ellis M, Dressler L, 2009, Topoisomerase II{alpha} amplification does not predict benefit from dose-intense cyclophosphamide, doxorubicin, and fluorouracil therapy in HER2-amplified early breast cancer: results of CALGB 8541/ 150013. J Clin Oncol 27:3430–3436 Arriola E, Rodriguez-Pinilla SM, Lambros MBK, Jones RL, James M, Savage K, Smith IE, Dowsett M, Reis-Filho JS, 2007, Topoisomerase II alpha amplification may predict benefit from adjuvant anthracyclines in HER2 positive early breast cancer. Breast Cancer Res Treat 106:181–189 Schindlbeck C, Mayr D, Olivier C, Rack B, Engelstaedter V, Jueckstock J, Jenderek C, Andergassen U, Jeschke U, Friese K, 2010, Topoisomerase II alpha expression rather than gene amplification predicts responsiveness of adjuvant anthracyclinebased chemotherapy in women with primary breast cancer. J Cancer Res Clin Oncol 136:1029–1037 Tanner M, Isola J, Wiklund T, Erikstein B, Kellokumpu-Lehtinen P, MalmstromP,Wilking N, Nilsson J, Bergh J, 2006, Topoisomerase II alpha gene amplification predicts favorable treatment response to tailored and dose-escalated anthracycline-based adjuvant chemotherapy in HER-2/neu-amplified breast cancer: Scandinavian Breast Group Trial 9401. J Clin Oncol 24:2428–2436 Fritz P, Cabrera CM, Dippon J, Gerteis A, Simon W, Aulitzky WE, van der Kuip H, 2005, c-erbB2 and topoisomerase II alpha protein expression independently predict poor survival in primary human breast cancer: a retrospective study. Breast Cancer Res 7: R374–R384 Durbecq V, Desmed C, Paesmans M, Cardoso F, Di Leo A, Mano M, Rouas G, Leroy JY, Sotiriou C, Piccart M, Larsimont D, 2004, Correlation between topoisomerase-IIalpha gene amplification and protein expression in HER-2 amplified breast cancer. Int J Oncol 25:1473–1479 Mueller RE, Parkes RK, Andrulis I, O'Malley FP, 2004, Amplification of the TOP2A gene does not predict high levels of topoisomerase II alpha protein in human breast tumor samples. Genes Chromosomes Cancer 39:288–297 Callagy G, Pharoah P, Chin SF, Sangan T, Daigo Y, Jackson L, Caldas C, 2005, Identification and validation of prognostic markers in breast cancer with the complementary use of array- CGH and tissue microarrays. J Pathol 205:388–396 Brase JC, Schmidt M, Fischbach T, Sultmann H, Bojar H, Koelbl H, Hellwig B, Rahnenfuhrer J, Hengstler JG, Gehrmann MC, 2010, ERBB2 and TOP2A in breast cancer: a comprehensive analysis of gene amplification, RNA levels, and protein expression and their influence on prognosis and prediction. Clin Cancer Res 16:2391–2401 Konecny GE, Pauletti G, Untch M, Wang HJ, Mobus V, Kuhn W, Thomssen C, Harbeck N, Wang L, Apple S, Janicke F, Slamon DJ, 2010, Association between HER2, TOP2A, and response to anthracycline-based preoperative chemotherapy in high-risk primary breast cancer. Breast Cancer Res Treat 120:481–489 Durbecq V, Paesmans M, Cardoso F, Desmedt C, Di Leo A, Chan S, Friedrichs K, Pinter T, Van Belle S, Murray E, Bodrogi I, Walpole E, Lesperance B, Korec S, Crown J, Simmonds P, Perren TJ, Leroy JY, Rouas G, Sotiriou C, Piccart M, Larsimont D, 2004, Topoisomerase-II alpha expression as a predictive marker in a population of advanced breast cancer patients randomly treated either with single-agent doxorubicin or single-agent docetaxel. Mol Cancer Ther 3:1207–1214 Kahan Z, Uhercsak G, Hajnal-Papp R, Boda K, Thurzo L, 2005, Dose-dense sequential adriamycin-Paclitaxel-cyclophosphamide chemotherapy is well tolerated and safe in high-risk early breast cancer. Oncology 68:446–453 Kahan Z, Spanik S, Wagnerova M, Skacel T, Planko B, Fitzthum E, Lindner E, Soldatenkova V, Zielinski CC, Brodowicz T, 2008, Feasibility of two dose-dense FEC regimens with growth factor support for adjuvant therapy in patients with early breast cancer: results from a randomised study of the Central European Cooperative Oncology Group, CECOG). Breast Cancer Res Treat 112:557–563 Nocito A, Kononen J, Kallioniemi OP, Sauter G, 2001, Tissue microarrays, TMAs, for high-throughput molecular pathology research. Int J Cancer 94:1–5 Park K, Kim J, Lim S, Han S, 2003, Topoisomerase II-alpha, topoII, and HER2 amplification in breast cancers and response to preoperative doxorubicin chemotherapy. Eur J Cancer 395:631– 634 Tubbs R, Barlow WE, Budd GT, Swain E, Porter P, Gown A, Yeh IT, Sledge G, Shapiro C, Ingle J, Haskell C, Albain KS, Livingston R, Hayes DF, 2009, Outcome of patients with earlystage breast cancer treated with doxorubicin-based adjuvant chemotherapy as a function of HER2 and TOP2A status. J Clin Oncol 27:3881–3886 Gunnarsdottir KA, Jensen MB, Zahrieh D, Gelber RD, Knoop A, Bonetti M, Mouridsen H, Ejlertsen B, 2010, CEF is superior to CMF for tumours with TOP2A aberrations: a Subpopulation Treatment Effect Pattern Plot, STEPP, analysis on Danish Breast Cancer Cooperative Group Study 89D. Breast Cancer Res Treat 123:163–169 Hannemann J, Kristel P, van Tinteren H, Bontenbal M, van Hoesel QG, Smit WM, Nooij MA, Voest EE, van der Wall E, Hupperets P, de Vries EG, Rodenhuis S, van de Vijver MJ, 2006, Molecular subtypes of breast cancer and amplification of topoisomerase II alpha: predictive role in dose intensive adjuvant chemotherapy. Br J Cancer 95:1334–1341 Di Leo A, Larsimont D, Gancberg D, Jarvinen T, Beauduin M, Vindevoghel A, Michel J, Focan CH, Ries F, Gobert PH, Closon- Dejardin MT, Dolci S, Rouas G, Paesmans M, Lobelle JP, Isola J, Piccart MJ, 2001, HER-2 and topo-isomerase IIalpha as predictive markers in a population of node-positive breast cancer patients randomly treated with adjuvant CMF or epirubicin plus cyclophosphamide. Ann Oncol 12:1081–1089 Berry DA, Cirrincione C, Henderson IC, Citron ML, Budman DR, Goldstein LJ, Martino S, Perez EA, Muss HB, Norton L, Hudis C, Winer EP, 2006, Estrogen-receptor status and outcomes of modern chemotherapy for patients with node-positive breast cancer. JAMA 295:1658–1667 Rody A, Karn T, Ruckhaberle E, Muller V, Gehrmann M, Solbach C, Ahr A, Gatje R, Holtrich U, Kaufmann M, 2009, Gene expression of topoisomerase II alpha, TOP2A, by microarray analysis is highly prognostic in estrogen receptor, ER, positive breast cancer. Breast Cancer Res Treat 113:4574–66 Jarvinen TA, Kononen J, Pelto-Huikko M, Isola J, 1996, Expression of topoisomerase II alpha is associated with rapid cell proliferation, aneuploidy, and c-erbB2 overexpression in breast cancer. Am J Pathol 148:2073–2082 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 61 EP 68 DI 10.1007/s12253-011-9417-4 PG 8 ER PT J AU Sugunadevi, G Suresh, K Vijayaanand, AM Rajalingam, K Sathiyapriya, J AF Sugunadevi, Govindasamy Suresh, Kathiresan Vijayaanand, Arokia Mariadoss Rajalingam, Kasinathan Sathiyapriya, Jagadeesan TI Anti Genotoxic Effect of Mosinone-A on 7, 12-Dimethyl Benz[a] Anthracene Induced Genotoxicity in Male Golden Syrian Hamsters SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chromosomal aberrations; Comet assay; Genotoxicity; DMBA; Micronuclei; Mosinone-A ID Chromosomal aberrations; Comet assay; Genotoxicity; DMBA; Micronuclei; Mosinone-A AB The present study was aimed to evaluate the antigenotoxic effect of Mosinone-A on 7,12-dimethylbenz [a]anthracene induced genotoxicity. The frequency of micronucleated polychromatic erythrocytes [MnPCEs], chromosomal aberrations [CA], DNA damage (comet assay) as cytogenetic markers and the status of lipid peroxidation byproducts, antioxidants and phase II detoxification agents were used as biochemical markers to assess the antigenotoxic effect of Mosinone-A on DMBA induced genotoxicity. A single intraperitoneal injection of DMBA (30 mg/kg b.wt) to golden Syrian hamsters, resulted in marked elevation in the frequency of MnPCEs, aberrations in the chromosomal structure were found in bone marrow and DNA damage (comet assay) was found in blood cells and altered level of lipid peroxidation, antioxidants, and phase II detoxification agents. Oral pretreatment of Mosinone-A (2 mg/kg b.wt) for 5 days to DMBA treated animals significantly reduced the frequency of MnPCEs, chromosomal abnormalities such as chromosomal break, gap, minute, fragment, DNA damage and reversed the status of biochemical variables. Our results thus demonstrated the antigenotoxic effect of Mosinone-A on DMBA induced genotoxicity in male golden Syrian hamsters. C1 [Sugunadevi, Govindasamy] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608 002 Tamil Nadu, India. [Suresh, Kathiresan] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608 002 Tamil Nadu, India. [Vijayaanand, Arokia Mariadoss] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608 002 Tamil Nadu, India. [Rajalingam, Kasinathan] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608 002 Tamil Nadu, India. [Sathiyapriya, Jagadeesan] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608 002 Tamil Nadu, India. RP Suresh, K (reprint author), Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, 608 002 Tamil Nadu, India. EM suraj_cks@yahoo.co.in CR Britto AP, Ravindran G, 2007, A review of cytogenetics and its automation. J Med Sci 7:1–18 Limoli CL, Kaplan MI, Philips JW et al, 1997, Differential induction of chromosomal instability by DNA strand-breaking agents. Cancer Res 57:4048–4056 Sancar A, 1994, Mechanism of DNA excision repair. Science 266:1954–1956 Tice RR, Agurell E, Anderson D et al, 2000, Single cell gel comet assay: guidelines for in vitro and in vivo genetic toxicology testing. Environ Mol Mutagen 35:206–221 Zimonjic DB, Savkovic N, Andjelkovic M, 1990, Genotoxic agents: the effects, principles and methodology of detection. Naucna Knjiga Beo-grad: 1–395 Schriever-Schwemmer G, Klieshch U, Adler D, 1997, Extruded micronuclei induced by colchicine or acrylamide contain mostly lagging chromosomes identified in paint brush smears by minor and major mouse DNA probes. Mutagenesis 12:201–207 Panjamurthy K, Manoharan S, Menon VP et al, 2008, Protective role of withaferin-A on 7,12-dimethylbenz[a]anthracene induced genotoxicity in bone marrow of Syrian golden hamsters. J Biochem Mol Toxicol 22:251–258 Bonassi S, Znaor A, Cappi M et al, 2007, An increased micronuclei frequency in peripheral blood lymphocytes predicts the risk of cancer in humans. Carcinogenesis 28:625–631 Henderson L, Wolfreys A, Fedyk J et al, 1998, The ability of the comet assay to discriminate between genotoxin and cytotoxins. Mutagenesis 13:89–94 Manoharan S, Panjamurthy K, Vasudevan K et al, 2006, Protective effects of Jasminum grandiflorum Linn on DMBA induced chromosomal aberrations in bone marrow of wistar rats. Int J Pharmacol 2:406–410 Miyata M, Furukawa M, Takahahsi K et al, 2001, Mechanism of 7,12-dimethyl benz[a]anthracene induced immunotoxicity: role of metabolic activation at the target organ. Jpn J Pharmacol 86:302– 309 Bhuvaneswari V, Velmurugan B, Abraham SK et al, 2004, Tomato and garlic by gavage modulate 7,12-dimethyl benz [a]anthracene induced genotoxicity and oxidative stress in mice. Braz J Med Biol Res 37:1029–1034 Ray G, Husain SH, 2002, Oxidant, antioxidants and carcinogenesis. Indian J Exp Biol 40:1213–1232 Manoharan S, Kavitha K, Senthil N et al, 2006, Evaluation of anticarcinogenic effects of Clerodendron inerme on 7,12-dimethylbenz[ a]anthracene induced hamster buccal pouch carcinogenesis. Singapore Med J 47:1038–1043 Suresh K, Manoharan S, Panjamurthy K et al, 2006, Chemopreventive and antilipid peroxidative efficacy of Annona squamosa bark extracts in experimental oral carcinogenesis. Pakistan J Biol Sci 9:2600–2605 Mayer C, Schmezer P, Freese R et al, 2000, Lipid peroxidation status, somatic mutations and micronuclei in peripheral lymphocytes: a case observation on a possible interrelationship. Cancer Lett 152:167–169 Bhuvaneswari V, Abraham SK, Nagini S, 2005, Combinatorial antigenotoxic effects of tomato and garlic through modulation of xenobiotic-metabolizing enzymes during hamster buccal pouch carcinogenesis. Nutrition 21:726–731 Park EJ, Pezzuto JM, 2002, Botanicals in cancer chemoprevention. Cancer Metastasis Rev 21:231–255 Nepka C, Asprodini E, Kouretas D, 1999, Tannins, xenobiotic metabolism and cancer chemoprevention in experimental animals. Eur J Drug Metab Pharmacokinet 24:183–189 Aruna K, Sivaramakrishnan VM, 1992, Anticarcinogenic effects of some Indian plant products. Food Chem Toxicol 30:953–956 Chavan MJ, Wakte PS, Shinde DB, 2010, Analgesic and antiinflammatory activity of caryophyllene oxide from Annona squamosa L.Bark. Phytomedicine 17:149–151 Fujimoto Y, 1988, Squamosin, a new cytotoxic bis-tetrahydrofuran containing acetogenin from Annona squamosa. Chem Pharm Bull 36:4802–4806 Londerhausen M, 1991, Molecular mode of action of annocins. Pestic Sci 33:427–438 Hopp DC, 1997, Novel mono tetra hydro furan ring acetogenins from the bark of Annona squamosa, showing cytotoxic selectivities for the human pancreatic carcinoma cell line PAC-2. J Nat Prod 60:581–586 Maclaughlin JL, Hoop DC, 2004, Selectivity cytotoxic acetogenin compound. United States patent US6242483: 242–42 Schmid W, 1973, Chemical mutagen testing on in vivo somatic mammalian cells. Agents Actions 3:77–85 Kilian DJ, Moreland FM, Benge MC et al, 1977, Collaborative studies to measure inters laboratory variation with the in vivo bone marrow metaphase procedure. In: Kilbey BJ, Legator M, Nichols W, Romel C, eds, Handbook of mutagenicity test procedures. Elsevier, Amsterdam, North-Holland), pp 243–260 Singh NP, 2000, Micro gels for estimation of DNA strand breaks, DNA protein crosslinks and apoptosis. Mutat Res 455:111–127 Yagi K, 1987, Lipid peroxides and human diseases. Chem Phys Lipids 45:337–351 Ohkawa H, Ohishi N, Yagi K, 1979, Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 95:351–358 Kakkar P, Das B, Viswanathan PN, 1984, A modified spectrophotometric assay of superoxide dismutase. Indian J Biochem Biophys 21:130–132 Rotruck T, Pope AL, Ganther HE et al, 1973, Selenium: biochemical role as a component of glutathione peroxidase. Science 179:588–590 Sinha AK, 1972, Colorimetric assay of catalase. Anal Biochem 47:389–394 Beutler E, Kelley BM, 1963, The effect of sodium nitrite on RBC glutathione. Experientia 29:96–97 Habig WH, Pabst MJ, Jakoby WB et al, 1974, Glutathione-Stransferase, the first enzymatic step in mercapturic acid formation. J Biol Chem 249:7130–7139 Carlberg I, Mannervik B, 1985, Glutathione reductase assay. Methods in Enzymol Academic Press New York 113: 484–490 Osaka M, Matsuo S, Koh T et al, 1996, Specific N-ras mutation in bone marrow with in 48 hrs of 7,12-dimethylbenz[a]anthracene treatment in Huggins-sugiyama rat leukemogenesis. Mol Carcinog 16:126–131 Chandra Mohan KVP, Bhuvaneswari V, Abraham SK, 2003, Dose-dependent protection of tomato against 7,12-dimethylbenz [a]anthracene induced genotoxicity and oxidative stress in mice. J Med Food 6:169–173 Pugalendhi P, Manoharan S, Panjamurthy K, Balakrishnan S, Nirmal MR, 2009, Antigenotoxic effect of genistein against 7,12- dimethylbenz[a]anthracene induced genotoxicity in bone marrow cells of female Wistar rats. Pharmacol Rep 61:296–303 Ragers AE, Zeisel SH, Groopman J, 1993, Diet and carcinogenesis. Carcinogenesis 14:2205–2217 Hopp DC, Zeng L, Gu Z et al, 1996, Squamotacin: an annonaceous acetogenin with cytotoxic selectivity for the human prostate tumor cell line, PC/3). J Nat Prod 59:97–99 Zeng L, 1996, Novel mono tetra hydro furan ring acetogenins from the bark of Annona squamosa, showing cytotoxic selectivities for the certain specific human tumor cell line PAC-2. J Nat Prods 59:1035–1042 Celik A, Cavas T, Ergene-Gozukara S, 2003, Cytogenetic monitoring in petrol station attendants: micronucleus test in exfoliated buccal cells. Mutagenesis 18:417–421 Schmid W, 1975, The micronucleus test. Mutat Res 31:9–15 Fairbairn DW, Olive PL, O’Neill KL, 1995, The comet assay: a comprehensive review. Mutat Res 339:37–59 Piperakis SM, Visvardis EE, Tassiou AM, 1999, Comet assay for nuclear DNA damage. Meth Enzymol 300:184–194 Ames BN, Shigenaga MK, Hagen TM, 1993, Oxidants, antioxidants, and the degenerative disease of aging. Proc Natl Acad Sci USA 90:7915–7922 Dreher D, Junod AF, 1996, Role of oxygen free radicals in cancer development. Eur J Canc 32:30–38 Wilkinson J 4th, Clapper ML, 1997, Detoxification enzymes and chemoprevention. Proc Soc Exp Biol Med 216:192–200 Schwartz JL, Shklar G, 1996, Glutathione inhibits experimental oral carcinogenesis, P53 expression and angiogenesis. Nutr cancer 26:229–236 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 69 EP 77 DI 10.1007/s12253-011-9418-3 PG 9 ER PT J AU Shen, GJ Xu, YCh Li, X Dong, JM Jiang, NZ Wang, J Wang, BL AF Shen, Guo Jian Xu, Yang Chao Li, Xin Dong, Jun Ming Jiang, Nong Zi Wang, Jin Wang, Bo Lin TI Dystroglycan is Associated with Tumor Progression and Patient Survival in Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adhesion molecule; α-dystroglycan; β-dystroglycan; Gastric cancer; Prognosis ID Adhesion molecule; α-dystroglycan; β-dystroglycan; Gastric cancer; Prognosis AB Previous reports had indicated that there was a possible correlation of dystroglycan (DG) with biological behavior of cancer cells and cancer patients’ survival. However, the role of DG expression in gastric cancer was rarely studied. In this study, α-DG and β-DG expression were determined by immunohistochemistry in specimens of primary cancer, metastatic lymph node, distal metastatic lesion, and their normal counterpart tissues in 20 gastric cancer patients. Correlations between α-DG and β-DG expression and prognosis were retrospectively analyzed. Our results found that positive expression of α-DG in normal mucosa, paired primary tumor, metastatic lymph node and distal metastatic site was detected in 95%, 70%, 25%, and 5% specimens, individually. Regarding β-DG,it was 70%, 55%, 10%, and 10%, individually. Patients who had lower α-DG expression in tumors than in normal counterparts showed poor survival (p=0.002), whereas such a correlation was not found in the case of β-DG (p=0.079). Difference of α-DG between primary tumor and its normal counterparts was an independent prognostic factor in gastric cancer with distal metastasis. This study showed DG expression was gradually reduced during tumor progression. Different expression of α-DG, but not β-DG, between primary tumor and normal specimen, correlated with patient survival, implicating a potential marker for gastric cancer prognosis. C1 [Shen, Guo Jian] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical Oncology, 310016 Hangzhou, China. [Xu, Yang Chao] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical Oncology, 310016 Hangzhou, China. [Li, Xin] Hangzhou First People’s Hospital, Department of Medical Oncology, 310016 Hangzhou, China. [Dong, Jun Ming] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical Oncology, 310016 Hangzhou, China. [Jiang, Nong Zi] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of PathologyHangzhou, China. [Wang, Jin] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of PathologyHangzhou, China. [Wang, Bo Lin] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical Oncology, 310016 Hangzhou, China. RP Wang, BL (reprint author), Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical Oncology, 310016 Hangzhou, China. EM wanglinbo@medmail.com.cn CR Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60:277–300 Masterson J, O'Dea S, 2007, Posttranslational truncation of Ecadherin and significance for tumour progression. Cells Tissues Organs 185:175–179 Sgambato A, Brancaccio A, 2005, The dystroglycan complex: from biology to cancer. J Cell Physiol 205:163–169 Henry MD, Campbell KP, 1998, A role for dystroglycan in basement membrane assembly. Cell 95:859–870 Ibraghimov-Beskrovnaya O, Ervasti JM, Leveille CJ, Slaughter CA, Sernett SW, Campbell KP, 1992, Primary structure of dystrophin-associated glycoproteins linking dystrophin to the extracellular matrix. Nature 355:696–702 Brennan PA, Jing J, Ethunandan M, Gorecki D, 2004, Dystroglycan complex in cancer. Eur J Surg Oncol 30:589–592 HenryMD, CohenMB, Campbell KP, 2001, Reduced expression of dystroglycan in breast and prostate cancer. Hum Pathol 32:791–795 Sgambato A, Migaldi M, Montanari M et al, 2003, Dystroglycan expression is frequently reduced in human breast and colon cancers and is associated with tumor progression. Am J Pathol 162:849–860 Sgambato A, Tarquini E, Resci F et al, 2006, Aberrant expression of alpha- dystroglycan in cervical and vulvar cancer. Gynecol Oncol 103:397–404 Cross SS, Lippitt J, Mitchell A et al, 2008, Expression of betadystroglycan is reduced or absent in many human carcinomas. Histopathology 53:561–566 Sgambato A, De Paola B, Migaldi M et al, 2007, Dystroglycan expression is reduced during prostate tumorigenesis and is regulated by androgens in prostate cancer cells. J Cell Physiol 213:528–39 Moon YW, Rha SY, Zhang X et al, 2009, Increments of alphadystroglycan expression in liver metastasis correlate with poor survival in gastric cancer. J Surg Oncol 100:459–465 Xu LZ, Yang W, 1996, Immunohistochemistry results assessment. China Oncol 6:229–231 Schwock J, Dhani N, Hedley DW, 2010, Targeting focal adhesion kinase signaling in tumor growth and metastasis. Expert Opin Ther Targets 14:77–94 Sgambato A, Camerini A, Amoroso D et al, 2007, Expression of dystroglycan correlates with tumor grade and predicts survival in renal cell carcinoma. Cancer Biol Ther 6:1840–1846 Losasso C, Di Tommaso F, Sgambato A et al, 2000, Anomalous dystroglycan in carcinoma cell lines. FEBS Lett 484:194–198 Jiang X, Rieder S, Giese NA, Friess H, Michalski CW, Kleeff J, 2009, Reduced alpha-Dystroglycan Expression Correlates with Shortened Patient Survival in Pancreatic Cancer. J Surg Res [Epub ahead of print] NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 79 EP 84 DI 10.1007/s12253-011-9419-2 PG 6 ER PT J AU Lakatos, G Sipos, F Miheller, P Hritz, I Varga, ZsM Juhasz, M Molnar, B Tulassay, Zs Herszenyi, L AF Lakatos, Gabor Sipos, Ferenc Miheller, Pal Hritz, Istvan Varga, Zsofia Maria Juhasz, Mark Molnar, Bela Tulassay, Zsolt Herszenyi, Laszlo TI The Behavior of Matrix Metalloproteinase-9 in Lymphocytic Colitis, Collagenous Colitis and Ulcerative Colitis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Matrix metalloproteinase; Ulcerative colitis; Lymphocytic colitis; Collagenous colitis; Immunohistochemistry; Inflammation ID Matrix metalloproteinase; Ulcerative colitis; Lymphocytic colitis; Collagenous colitis; Immunohistochemistry; Inflammation AB Matrix metalloproteinases play an important role in extracellular matrix remodelling. It has been proposed that matrix metalloproteinase-9 (MMP-9) is involved in epithelial damage in ulcerative colitis (UC). However, to our knowledge, no data are available in terms of MMP-9 expression in microscopic colitis. Determination of mucosal protein expression levels of MMP-9 in lymphocytic colitis (LC), collagenous colitis (CC) and UC. MMP-9 immunohistochemical expressions were analyzed in paraffinembedded tissue samples by immunohistochemistry including patients with LC, CC, UC, active diverticulitis, inactive diverticular disease and healthy control subjects. UC was also subgrouped according to the severity of inflammation. Immunostaining was determined semiquantitatively. Independent colonic biopsies from healthy and severe UC cases were used for gene expression analyses. For further comparison MMP-9 serum antigen levels were also determined in patients with UC and control patients without macroscopic or microscopic changes during colonoscopy. MMP-9 mucosal expression was significantly higher in UC (26.7±19.5%) compared to LC (6.6±9.3%), CC (6.4±7.6%), active diverticulitis (5.33±2.4%), inactive diverticular disease (5.0±2.2%) and controls (6.3±2.6%) (P<0.001). The immunohistochemical expression of MMP-9 in LC and CC was similar as compared to controls. MMP-9 expression was significantly higher in each inflammatory group of UC compared to controls (mild: 11.0±2.8%, moderate: 23.9±3.7%, severe UC: 52.6±3.9% and 6.3±2.6%, respectively, P<0.005). The gene expression microarray data and RT-PCR results demonstrated a significantly higher expression of MMP-9 in severely active UC compared to healthy controls (P<0.001). Significantly higher MMP-9 serum antigen concentrations were observed in UC patients compared with the control group (P<0.05). MMP-9 seems to play no role in the inflammatory process of LC and CC. In contrast, the mucosal up-regulation of MMP-9 correlated with the severity of inflammation in UC. The increased MMP-9 expression could contribute to the severity of mucosal damage in active UC. C1 [Lakatos, Gabor] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Miheller, Pal] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Hritz, Istvan] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Varga, Zsofia Maria] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Juhasz, Mark] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Molnar, Bela] Hungarian Academy of Science, Clinical Gastroenterology Research UnitBudapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Herszenyi, Laszlo] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. RP Herszenyi, L (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM hersz@bel2.sote.hu CR Kirsner JB, 2001, Historical origins of current IBD concepts. World J Gastroenterol 7:175–184 Kucharzik T, Maaser C, Lugering A, Kagnoff M, Mayer L, Targan S, Domschke W, 2006, Recent understanding of IBD pathogenesis: implications for future therapies. Inflamm Bowel Dis 12:1068–1083 Liotta LA, Stetler-Stevenson WG, 1991, Tumor invasion and metastasis: an imbalance of positive and negative regulation. Cancer Res 51:5054–5059 Plebani M, Herszenyi L, Cardin R, Roveroni G, Carraro P, De Paoli M, Rugge M, Nitti D, Naccarato R, Farinati F, 1995, Cysteine and serine proteases in gastric cancer. Cancer 76:367–375 Herszenyi L, Plebani M, Carraro P, De Paoli M, Roveroni R, Cardin R, Tulassay Zs, Naccarato R, Farinati F, 1999, The role of cysteine and serine proteases in colorectal cancer. Cancer 86:1135–1142 Herszenyi L, Farinati F, Cardin R, Istvan G, Molnar DL, Hritz I, De Paoli M, Plebani M, Tulassay Z, 2008, Tumor marker utility and prognostic relevance of cathepsin B, cathepsin L, urokinasetype plasminogen activator, plasminogen activator inhibitor type- 1, CEA and CA 19-9 in colorectal cancer. BMC Cancer 8:194., DOI 10.1186/1471-2407-8-194 Herszenyi L, Plebani M, Carraro P, De Paoli M, Di Mario F, Kusstatscher S, Rugge M, Cardin R, Naccarato R, Farinati F, 1997, Impaired fibrinolysis and increased protease levels in gastric and duodenal mucosa of patients with active duodenal ulcer. Am J Gastroenterol 92:843–847 Herszenyi L, Istvan G, Cardin R, De Paoli M, Plebani M, Tulassay Z, Farinati F, 2008, Serum cathepsin B and plasma urokinase-type plasminogen activator levels in gastrointestinal tract cancers. Eur J Cancer Prev 17:438–445 Baugh MD, Perry MJ, Hollander AP, Davies DR, Cross SS, Lobo AJ, Taylor CJ, Evans GS, 1999, Matrix metalloproteinase levels are elevated in inflammatory bowel disease. Gastroenterology 117:814–822 von Lampe B, Barthel B, Coupland SE, Riecken EO, Rosewicz S, 2000, Differential expression of matrix metalloproteinases and their tissue inhibitors in colon mucosa of patients with inflammatory bowel disease. Gut 47:63–73 Monteleone G, Caruso R, Fina D, Peluso I, Gioia V, Stolfi C, Fantini MC, Caprioli F, Tersigni R, Alessandroni L, MacDonald TT, Pallone F, 2006, Control of matrix metalloproteinase production in human intestinal fibroblasts by interleukin 21. Gut 55:1774–1780 Ravi A, Garg P, Sitaraman SV, 2007, Matrix metalloproteinases in inflammatory bowel disease: Boon or a baine? Inflamm Bowel Dis 13:97–107 Herszenyi L, Hritz I, Pregun I, Sipos F, Juhasz M, Molnar B, Tulassay Z, 2007, Alterations of glutathione S-transferase and matrix metalloproteinase-9 expressions are early events in the esophageal carcinogenesis. World J Gastroenterol 13:676–682 Herszenyi L, Sipos F, Galamb O, Solymosi N, Hritz I, Miheller P, Berczi L, Molnar B, Tulassay Z, 2008, Matrix metalloproteinase- 9 expression in the normal mucosa-adenoma-dysplasia-adenocarcinoma sequence of the colon. Pathol Oncol Res 14(1):31–37 Nelson AR, Fingleton B, Rothenberg ML, Matrisian LM, 2000, Matrix metalloproteinases: biologic activity and clinical implications. J Clin Oncol 18:1135–1149 Kirkegaard T, Hansen A, Bruun E, Brynskov J, 2004, Expression and localisation of matrix metalloproteinases and their natural inhibitors in fistulae of patients with Crohn’s disease. Gut 53:701–709 Bailey CJ, Hembry RM, Alexander A, Irving MH, Grant ME, Shuttleworth CA, 1994, Distribution of the matrix metalloproteinases stromelysin, gelatinases A and B, and collagenase in Crohn’s disease and normal intestine. J Clin Pathol 47:113–116 Stallmach A, Chan CC, Ecker KW, Feifel G, Herbst H, Schuppan D, Zeitz M, 2000, Comparable expression of matrix metalloproteinases 1 and 2 in pouchitis and ulcerative colitis. Gut 47:415–422 Von Lampe B, Barthel B, Coupland SE, Riecken EO, Rosewicz S, 2000, Differential expression of matrix metalloproteinases and their tissue inhibitors in colon mucosa of patients with inflammatory bowel disease. Gut 47:63–73 Nielsen OH, Vainer B, de Muckadell OB, 2004, Microscopic colitis: a missed diagnosis? Lancet 364:2055–2057 Aqel B, Bishop M, Krishna M, Cangemi J, 2003, Collagenous colitis evolving into ulcerative colitis: a case report and review of the literature. Dig Dis Sci 48:2323–2327 Sipos F, Molnar B, Zagoni T, Berczi L, Tulassay Z, 2005, Growth in epithelial cell proliferation and apoptosis correlates specifically to the inflammation activity of inflammatory bowel disease: ulcerative colitis shows specific p53- and EGFR expression alterations. Dis Colon Rectum 48:775–786 Galamb O, Sipos F, Dinya E, Spisak S, Tulassay Z, Molnar B, 2006, mRNA expression, functional profiling and multivariate classification of colon biopsy specimen by cDNA overall glass microarray. World J Gastroenterol 12:6998–7006 Medina C, Radomski MW, 2006, Role of matrix metalloproteinases in intestinal inflammation. J Pharmacol Exp Ther 318, 3):933–938 Pender SL, Tickle SP, Docherty AJ, Howie D, Wathen NC, MacDonald TT, 1997, A major role for matrix metalloproteinases in T cell injury in the gut. J Immunol 158(4):1582–1590 Gordon JN, Pickard KM, Di Sabatino A, Prothero JD, Pender SL, Goggin PM, MacDonald TT, 2008, Matrix metalloproteinase-3 production by gut IgG plasma cells in chronic inflammatory bowel disease. Inflamm Bowel Dis 14(2):195–203 Garg P, Vijay-Kumar M, Wang L, Gewirtz AT, Merlin D, Sitaraman SV, 2009, Matrix metalloproteinase-9-mediated tissue injury overrides the protective effect of matrix metalloproteinase-2 during colitis. Am J Physiol Gastrointest Liver Physiol 296:175– 184 Nyhlin N, Bohr J, Eriksson S, Tysk C, 2006, Systematic review: microscopic colitis. Aliment Pharmacol Ther 23(11):1525–1534 Gao Q, Meijer MJ, Kubben FJ, Sier CF, Kruidenier L, van Duijn W, van den Berg M, van Hogezand RA, Lamers CB, Verspaget HW, 2005, Expression of matrix metalloproteinases-2 and -9 in intestinal tissue of patients with inflammatory bowel diseases. Dig Liver Dis 37:584–592 Baugh MD, Perry MJ, Hollander AP, Davies DR, Cross SS, Lobo AJ, Taylor CJ, Evans GS, 1999, Matrix metalloproteinase levels are elevated in inflammatory bowel disease. Gastroenterology 117:814–822 Manfredi MA, Zurakowski D, Rufo PA, Walker TR, Fox VL, Moses MA, 2008, Increased incidence of urinary matrix metalloproteinases as predictors of disease in pediatric patients with inflammatory bowel disease. Inflamm Bowel Dis 14:1091–1096 Castaneda FE, Walia B, Vijay-Kumar M, Patel NR, Roser S, Kolachala VL, Rojas M, Wang L, Oprea G, Garg P, Gewirtz AT, Roman J, Merlin D, Sitaraman SV, 2005, Targeted deletion of metalloproteinase 9 attenuates experimental colitis in mice: central role of epithelial-derived MMP. Gastroenterology 129:1991–2008 Martinesi M, Treves C, Bonanomi AG, Milla M, Bagnoli S, Zuegel U, Steinmeyer A, Stio M, 2010, Down-regulation of adhesion molecules and matrix metalloproteinases by ZK 156979 in inflammatory bowel diseases. Clin Immunol 136:51–60 Meijer MJ, Mieremet-Ooms MA, van Duijn W, van der Zon AM, Hanemaaijer R, Verheijen JH, van Hogezand RA, Lamers CB, Verspaget HW, 2007, Effect of the tumor necrosis factor-alpha antibody infliximab on the ex vivo mucosal matrix metalloproteinase-proteolytic phenotype in inflammatory bowel disease. Inflamm Bowel Dis 13:200–210 Di Sabatino A, Pender SL, Jackson CL, Prothero JD, Gordon JN, Picariello L, Rovedatti L, Docena G, Monteleone G, Ramtpon DS, Tonelli F, Corazza GR, MacDonald TT, 2007, Functional modulation of Crohn’s disease myofibroblasts by anti-tumor necrosis factor antibodies. Gastroenterology 133:137–149 Wang YD, Yan PY, 2006, Expression of matrix metalloproteinase-1 and tissue inhibitor ofmetalloproteinase-1 in ulcerative colitis.World J Gastroenterol 12:6050–6053 Kobayashi K, Arimura Y, Goto A, Okahara S, Endo T, Shinomura Y, Imai K, 2006, Therapeutic implications of the specific inhibition of causative matrix metalloproteinases in experimental colitis induced by dextran sulphat sodium. J Pathol 209:376–383 Miehlke S, Heymer P, Bethke B, Bastlein E, Meier E, Bartram HP, Wilhelms G, Lehn N, Dorta G, DeLarive J, Tromm A, Bayerdorffer E, Stolte M, 2002, Budesonide treatment for collagenous colitis: a randomized, double-blind, placebo-controlled, multicenter trial. Gastroenterol 123:978–984 Tromm A, Griga T, Mollmann HW, May B, Muller KM, Fisseler- Eckhoff A, 1999, Budesonide for the treatment of collagenous colitis: first results of a pilot trial. Am J Gastroenterol 94:1871–1875 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 85 EP 91 DI 10.1007/s12253-011-9420-9 PG 7 ER PT J AU Cappetta, A Bergamo, F Mescoli, C Lonardi, S Rugge, M Zagonel, V AF Cappetta, Alessandro Bergamo, Francesca Mescoli, Claudia Lonardi, Sara Rugge, Massimo Zagonel, Vittorina TI Hepatoid Adenocarcinoma of the Colon: What Should We Target? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatoid adenocarcinoma; Colon cancer; Hepatocellular carcinoma; Alpha-fetoprotein; FOLFOX; FOLFIRI ID Hepatoid adenocarcinoma; Colon cancer; Hepatocellular carcinoma; Alpha-fetoprotein; FOLFOX; FOLFIRI AB Hepatoid adenocarcinoma is a rare extra hepatic neoplasm that displays morphological and phenotypic features similar to those of hepatocellular carcinoma. We report a case of a 75-year-old woman, presenting with abdominal pain and complaints of weakness and lost of appetite, who was found to have a mass on her right colon. She underwent right hemicolectomy for a pT3N2M0, stage IIIC colon cancer. The tumor phenotype and immunophenotype, as documented by alpha-fetoprotein immunoreaction positivity, were consistent with adenocarcinoma of hepatoid origin. The patient received FOLFOX-4 regimen as adjuvant treatment, relapsed after six cycles, then was switched to FOLFIRI regimen plus Bevacizumab and progressed after only four cycles. She died 1 month later, eight months after the diagnosis. The lack of any clinical benefit despite an aggressive and multimodal therapeutic strategy, raises a question about what should be targeted when we face this rare disease associated with a very poor prognosis. C1 [Cappetta, Alessandro] Veneto Institute of Oncology—IRCCS, Medical Oncology Unit 1, Via Gattamelata 64, 35128 Padova, Italy. [Bergamo, Francesca] Veneto Institute of Oncology—IRCCS, Medical Oncology Unit 1, Via Gattamelata 64, 35128 Padova, Italy. [Mescoli, Claudia] University of Padua, Department of Diagnostic Medicine and Special Therapies, Pathology Unit 2, Via Gabelli 61, 35121 Padova, Italy. [Lonardi, Sara] Veneto Institute of Oncology—IRCCS, Medical Oncology Unit 1, Via Gattamelata 64, 35128 Padova, Italy. [Rugge, Massimo] University of Padua, Department of Diagnostic Medicine and Special Therapies, Pathology Unit 2, Via Gabelli 61, 35121 Padova, Italy. [Zagonel, Vittorina] Veneto Institute of Oncology—IRCCS, Medical Oncology Unit 1, Via Gattamelata 64, 35128 Padova, Italy. RP Cappetta, A (reprint author), Veneto Institute of Oncology—IRCCS, Medical Oncology Unit 1, 35128 Padova, Italy. EM alessandro.cappetta@gmail.com CR Bourreille J, Mateyer P, Sauger F et al, 1970, Existence of alpha fetoprotein during gastric-origin secondary cancer of the liver. Press Med 78:1277–1278 Ishikura H, Fukasawa Y, Ogasawara K et al, 1985, An AFPproducing gastric carcinoma with feature of hepatic differentiation: a case report. Cancer 56:840–848 IshikuraH,Kishimoto T, AndachiHet al, 1997, Gastrintestinal hepatoid adenocarcinoma: venous permeation and mimicry of hepatocellular carcinoma, a report of four cases. Histopathology 31:47–54 Terracciano LM, Glatz K, Mhawech P et al, 2003, Hepatoid adenocarcinoma with liver metastasis mimicking hepatocellular carcinoma: an immunohistochemical and molecular study of eight cases. Am J Surg Pathol 27:1302–1312 Metzgeroth G, Strobel P, Baumbusch T et al, 2010, Hepatoid adenocarcinoma—review of the literature illustrated by a rare case originating in the peritoneal cavity. Onkologie 33(5):263–9 Borgonovo G, Razzetta F, Assalino M et al, 2008, Rectal hepatoid carcinoma with liver metastases in a patient affected by ulcerative colitis. Hepatobiliary Pancreat Dis Int 7(5):539–43 Liu Q, Bannan M, Melamed J, Witkin GB et al, 2007, Two cases of hepatoid adenocarcinoma of the intestine in association with inflammatory bowel disease. Histopathology 51:123–5 Lattes C, Carella R, Faggioli S et al, 2000, Hepatoid adenocarcinoma of the rectum arising in ulcerative colitis: report of a case. Dis Colon rectum 43(1):105–8 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 93 EP 96 DI 10.1007/s12253-011-9424-5 PG 4 ER PT J AU Kallen, EM Nunes Rosado, GF Gonzalez, LA Sanders, EM Cates, MMJ AF Kallen, E Michael Nunes Rosado, G Flavia Gonzalez, L Adriana Sanders, E Melinda Cates, M M Justin TI Occasional Staining for p63 in Malignant Vascular Tumors: A Potential Diagnostic Pitfall SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Angiosarcoma; Hemangioendothelioma; Hemangioma; Kaposi sarcoma; p63 ID Angiosarcoma; Hemangioendothelioma; Hemangioma; Kaposi sarcoma; p63 AB Expression of p63, a putative marker for epithelial or myoepithelial differentiation, has been used to distinguish spindle cell carcinoma from sarcoma. The specificity of p63 for epithelial differentiation has not been thoroughly evaluated however, since p63 expression has been explored in only a handful of mesenchymal tumors. After observing unexpected immunohistochemical staining for p63 in an angiosarcoma of the breast, we evaluated a series of benign and malignant vascular tumors to determine the frequency of such a finding. Nuclear immunoreactivity to p63 was detected, at least focally, in 24% of malignant vascular tumors other than Kaposi sarcoma, which was uniformly negative. Benign vascular tumors were also negative for p63. Although p63 expression in tumors of vascular differentiation is unusual, it may be seen occasionally in some malignant vascular tumors. Thus, p63 is not entirely specific for epithelial differentiation. Since soft tissue angiosarcomas and hemangioendotheliomas sometimes express cytokeratins, the finding of nuclear p63 represents another potential pitfall in the differential diagnosis between poorly-differentiated carcinomas and vascular neoplasms. C1 [Kallen, E Michael] Vanderbilt University Medical Center and Vanderbilt University, School of Medicine, Department of Pathology, 1161 21st Ave. South, Medical Center North C-3322, 37232 Nashville, TN, USA. [Nunes Rosado, G Flavia] Vanderbilt University Medical Center and Vanderbilt University, School of Medicine, Department of Pathology, 1161 21st Ave. South, Medical Center North C-3322, 37232 Nashville, TN, USA. [Gonzalez, L Adriana] Vanderbilt University Medical Center and Vanderbilt University, School of Medicine, Department of Pathology, 1161 21st Ave. South, Medical Center North C-3322, 37232 Nashville, TN, USA. [Sanders, E Melinda] Vanderbilt University Medical Center and Vanderbilt University, School of Medicine, Department of Pathology, 1161 21st Ave. South, Medical Center North C-3322, 37232 Nashville, TN, USA. [Cates, M M Justin] Vanderbilt University Medical Center and Vanderbilt University, School of Medicine, Department of Pathology, 1161 21st Ave. South, Medical Center North C-3322, 37232 Nashville, TN, USA. RP Cates, MMJ (reprint author), Vanderbilt University Medical Center and Vanderbilt University, School of Medicine, Department of Pathology, 37232 Nashville, USA. EM justin.m.cates@vanderbilt.edu CR Dotsch V, Bernassola F, Coutandin D, Candi E, Melino G, 2010, p63 and p73, the ancestors of p53. Cold Spring Harb Perspect Biol 2(9):a004887 Laurikkala J, Mikkola ML, James M, Tummers M, Mills AA, Thesleff I, 2006, p63 regulates multiple signalling pathways required for ectodermal organogenesis and differentiation. Development 133(8):1553–1563 Moll UM, Slade N, 2004, p63 and p73: roles in development and tumor formation. Mol Cancer Res 2(7):371–386 Nekulova M, Holcakova J, Coates P, Vojtesek B, 2011, The role of P63 in cancer, stem cells and cancer stem cells. Cell Mol Biol Lett 16(2):296–327 Truong AB, Kretz M, Ridky TW, Kimmel R, Khavari PA, 2006, p63 regulates proliferation and differentiation of developmentally mature keratinocytes. Genes Dev 20(22):3185–3197 Vanbokhoven H, Melino G, Candi E, Declercq W, 2011, p63, a story of mice and men. J Invest Dermatol, In press)., DOI 10.1038/jid.2011.84 Carter MR, Hornick JL, Lester S, Fletcher CD, 2006, Spindle cell, sarcomatoid, carcinoma of the breast: a clinicopathologic and immunohistochemical analysis of 29 cases. Am J Surg Pathol 30, 3):300–309 Dotto JE, Glusac EJ, 2006, p63 is a useful marker for cutaneous spindle cell squamous cell carcinoma. J Cutan Pathol 33(6):413–417 Koker MM, Kleer CG, 2004, p63 expression in breast cancer: a highly sensitive and specific marker of metaplastic carcinoma. Am J Surg Pathol 28(11):1506–1512 KurianKM, Al-Nafussi A, 2002, Sarcomatoid/metaplastic carcinoma of the breast: a clinicopathological study of 12 cases. Histopathology 40(1):58–64 Morgan MB, Purohit C, Anglin TR, 2008, Immunohistochemical distinction of cutaneous spindle cell carcinoma. Am J Dermatopathol 30(3):228–232 Reis-Filho JS, Schmitt FC, 2002, Taking advantage of basic research: p63 is a reliable myoepithelial and stem cell marker. Adv Anat Pathol 9(5):280–289 Reis-Filho JS, Schmitt FC, 2003, p63 expression in sarcomatoid/ metaplastic carcinomas of the breast. Histopathology 42(1):94–95 Tse GM, Tan PH, Chaiwun B, Putti TC, Lui PC, Tsang AK, Wong FC, Lo AW, 2006, p63 is useful in the diagnosis of mammary metaplastic carcinomas. Pathology 38(1):16–20 Cates JM, Dupont WD, Barnes JW, Edmunds HS, Fasig JH, Olson SJ, Black CC, 2008, Markers of epithelial-mesenchymal transition and epithelial differentiation in sarcomatoid carcinoma: utility in the differential diagnosis with sarcoma. Appl Immunohistochem Mol Morphol 16(3):251–262 Lewis JS, Ritter JH, El-Mofty S, 2005, Alternative epithelialmarkers in sarcomatoid carcinomas of the head and neck, lung, and bladderp63, MOC-31, and TTF-1. Mod Pathol 18(11):1471–1481 Di Como CJ, Urist MJ, Babayan I, Drobnjak M, Hedvat CV, Teruya-Feldstein J, Pohar K, Hoos A, Cordon-Cardo C, 2002, p63 expression profiles in human normal and tumor tissues. Clin Cancer Res 8(2):494–501 Jo VY, Fletcher CDM, 2011, p63 immunohistochemical staining is limited in soft tissue tumors. Mod Pathol 24(1S):15A Chen SY, Takeuchi S, Urabe K, Hayashida S, Kido M, Tomoeda H, Uchi H, Dainichi T, Takahara M, Shibata S, Tu YT, Furue M, Moroi Y, 2008, Overexpression of phosphorylated-ATF2 and STAT3 in cutaneous angiosarcoma and pyogenic granuloma. J Cutan Pathol 35(8):722–730 Terada T, 2011, Angiosarcoma of the oral cavity. Head Neck Pathol 5(1):67–70 Senoo M, Matsumura Y, Habu S, 2002, TAp63γ, p51A, and dNp63α, p73L), two major isoforms of the p63 gene, exert opposite effects on the vascular endothelial growth factor, VEGF, gene expression. Oncogene 21(16):2455–2465 Chu PG, Weiss LM, 2002, Keratin expression in human tissues and neoplasms. Histopathology 40(5):403–439 Meis-Kindblom JM, Kindblom LG, 1998, Angiosarcoma of soft tissue: a study of 80 cases. Am J Surg Pathol 22(6):683– 697 Gill R, O’Donnell RJ, Horvai A, 2009, Utility of immunohistochemistry for endothelial markers in distinguishing epithelioid hemangioendothelioma from carcinoma metastatic to bone. Arch Pathol Lab Med 133(6):967–972 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 97 EP 100 DI 10.1007/s12253-011-9426-3 PG 4 ER PT J AU Corradi, D Alquati, S Bertoni, F Bartoli, V Dei Tos, PA Wenger, D Giannini, C AF Corradi, Domenico Alquati, Sara Bertoni, Franco Bartoli, Veronica Dei Tos, Paolo Angelo Wenger, Doris Giannini, Caterina TI A Small Intraneural Epithelioid Malignant Peripheral Nerve Sheath Tumour of the Median Nerve Simulating a Benign Lesion. Description of a Case and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Malignant peripheral nerve sheath tumour; Epithelioid variant; Surgical margins; Outcome; Immunohistochemistry ID Malignant peripheral nerve sheath tumour; Epithelioid variant; Surgical margins; Outcome; Immunohistochemistry AB The epithelioid variant of malignant peripheral nerve sheath tumours (MPNSTs) is a very rare malignancy. We describe the case of a 30-year-old man complaining of acute pain in his right elbow, mild distal paraesthesias, and some motor deficiencies. He was discovered as having a small fusiform swelling of the median nerve. In view of its very small size, shape, and nonspecific MRI signal, it had initially suggested a benign lesion. The diagnosis of epithelioid MPNST was made only at the histopathological examination. This malignant neoplasm recurred locally fourteen months after surgery. In addition to describe the above very rare case, we have reviewed the literature on epithelioid MPNSTs clearly involving deep major nerve trunks. This case serves as a warning that, even in major nerve trunks, tiny lesions may in reality be early intraneural MPNSTs which, due to their deep location, must be treated adequately with wide margin surgery since the resection margin status represents one of the major parameters influencing the local control of disease and its clinical outcome. C1 [Corradi, Domenico] University of Parma, Institute of Pathology, Via Gramsci 14, 43126 Parma, Italy. [Alquati, Sara] Arcispedale S. Maria Nuova, Department of OncologyReggio Emilia, Italy. [Bertoni, Franco] University of BolognaBologna, Italy. [Bartoli, Veronica] University of Parma, Institute of Pathology, Via Gramsci 14, 43126 Parma, Italy. [Dei Tos, Paolo Angelo] Regional Hospital, Department of PathologyTreviso, Italy. [Wenger, Doris] Mayo Clinic, Department of RadiologyRochester, MN, USA. [Giannini, Caterina] Mayo Clinic, Department of Laboratory Medicine and PathologyRochester, MN, USA. RP Corradi, D (reprint author), University of Parma, Institute of Pathology, 43126 Parma, Italy. EM domenico.corradi@unipr.it CR Scheithauer B, Louis D, Hunter S et al, 2007, Malignant peripheral nerve sheath tumor, MPNST). In: Louis D, Ohgaki H, Wiestler O et al, eds, WHO classification of tumours of the central nervous system. Lyon, IARC Press Weiss S, Goldblum J, 2008, Malignant tumors of the peripheral nerves. In: Weiss S, Goldblum J, eds, Enzinger and Weiss’s Soft Tissue Tumors, 5th edn. Mosby, Philadelphia Anghileri M, Miceli R, Fiore M et al, 2006, Malignant peripheral nerve sheath tumors: prognostic factors and survival in a series of patients treated at a single institution. Cancer 107:1065–1074 Laskin WB, Weiss SW, Bratthauer GL, 1991, Epithelioid variant of malignant peripheral nerve sheath tumor, malignant epithelioid schwannoma). Am J Surg Pathol 15:1136–1145 Lodding P, Kindblom LG, Angervall L, 1986, Epithelioid malignant schwannoma. A study of 14 cases. Virchows Arch A Pathol Anat Histopathol 409:433–451 Suh JS, Abenoza P, Galloway HR et al, 1992, Peripheral, extracranial, nerve tumors: correlation of MR imaging and histologic findings. Radiology 183:341–346 MurpheyMD, SmithWS, Smith SE et al, 1999, Fromthe archives of the AFIP. Imaging of musculoskeletal neurogenic tumors: radiologic-pathologic correlation. Radiographics 19:1253–1280 Ogose A, Hotta T, Morita T et al, 1999, Tumors of peripheral nerves: correlation of symptoms, clinical signs, imaging features, and histologic diagnosis. Skeletal Radiol 28:183–188 Li CS, Huang GS, Wu HD et al, 2008, Differentiation of soft tissue benign and malignant peripheral nerve sheath tumors with magnetic resonance imaging. Clin Imaging 32:121–127 Das Gupta TK, Brasfield RD, 1970, Solitary malignant schwannoma. Ann Surg 171:419–428 Ducatman BS, Scheithauer BW, Piepgras DG et al, 1986, Malignant peripheral nerve sheath tumors. A clinicopathologic study of 120 cases. Cancer 57:2006–2021 Hruban RH, Shiu MH, Senie RT et al, 1990, Malignant peripheral nerve sheath tumors of the buttock and lower extremity. A study of 43 cases. Cancer 66:1253–1265 White HR Jr, 1971, Survival in malignant schwannoma. An 18- year study. Cancer 27:720–729 Wong WW, Hirose T, Scheithauer BW et al, 1998, Malignant peripheral nerve sheath tumor: analysis of treatment outcome. Int J Radiat Oncol Biol Phys 42:351–360 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 101 EP 106 DI 10.1007/s12253-010-9348-5 PG 6 ER PT J AU Papachristou, JD Goodman, M Cieply, K Rao, NMU AF Papachristou, J Dionysios Goodman, Mark Cieply, Kathleen Rao, N M Uma TI Extraskeletal Osteosarcoma of Subcutaneous Soft Tissue with Lymph Node and Skin Metastasis: A Case Report with Fluorescence in Situ Hybridization Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports C1 [Papachristou, J Dionysios] University of Pittsburgh, School of Medicine, Department of PathologyPittsburgh, PA, USA. [Goodman, Mark] University of Pittsburgh, School of Medicine, Department of OrthopedicsPittsburgh, PA, USA. [Cieply, Kathleen] University of Pittsburgh, School of Medicine, Department of PathologyPittsburgh, PA, USA. [Rao, N M Uma] University of Pittsburgh, School of Medicine, Department of PathologyPittsburgh, PA, USA. RP Papachristou, JD (reprint author), University of Pittsburgh, School of Medicine, Department of Pathology, Pittsburgh, USA. EM papachristoudj@med.upatras.gr;dip13@pitt.edu CR Chang EB, Enzinger FM, 1987, Extraskeletal osteosarcoma. Cancer 60:1132–1142 Unni KK, 1998, Osteosarcoma of bone. J Orthop Sci 3:287–294 Banne BL, Evans HL, Ro JYet al, 1990, Extraskeletal osteosarcoma. A clininicopathologic review of 26 cases. Cancer 65:2762–2770 Jensen ML, Schumacher B, Myhren JO et al, 1998, Extraskeletal osteosarcomas: a clinicopathologic study of 25 cases. Am J Surg Pathol 22:588–594 Raymond AK, Ayala AG, Knuutila S, 2002, Conventional osteosarcoma. In: Fletcher CDM, Unni KK, Mertens F, eds, Pathology and genetics of tumors of soft tissue and bone. IARCPress, Lyon Lee JS, Fletsch JF, Wasdhal DA et al, 1995, A review of 40 patients with extraskeletal osteosarcoma. Cancer 76:2253–2259 Ducatman BS, Sheithauer BW, 1984, Malignant peripheral nerve sheath tumors with divergent differentiation. Cancer 54:1049–1057 Hasegawa T, Hirose T, Kudo E et al, 1991, Immunophenotypic heterogeneity in osteosarcomas. Hum Pathol 22:583–590 Fanburg-Smith JC, Bratthauer GL, Miettinen M, 1999, Osteocalcin and osteonectin immunoreactivity in extraskeletal osteosarcoma: a study of 28 cases. Hum Pathol 30:32–38 Rao UNM, Lotze MT, Karakousis CP, 1998, Distant skin and soft tissue metastases from sarcomas. J Surg Onc 69:94–98 Covello SP, Humphreys TR, Jason BL, 2003, A case of extraskeletal osteosarcoma with metastasis to the skin. J Am Dermatol 49:124–127 Riad S, Griffin AM, Liberman B et al, 2004, Lymph node metastasis in soft tissue sarcoma in an extremity. Clin Orthop Relat Res 426:129–134 Liotta LA, Kohn EC, 2001, The microenviroment of the tumorhost interface. Nature 411:375–379 Chambers AF, Groom AC, MacDonald IC, 2002, Dissemination and growth of cancer cells in metastatic sites. Nature Rev Cancer 2:563–572 Papachristou DJ, Batistatou A, Sykiotis GP et al, 2003, Activation of the JNK – AP-1 signal transduction pathways is associated with pathogenesis and progression of human osteosarcomas. Bone 32:364–371 Stock C, Kager L, Fink F-M et al, 2000, Chromosomal regions involved in the pathogenesis of osteosarcomas. Genes Chromosom Cancer 28:329–336 Spillane AJ, Fisher C, 1999, Myxoid liposarcoma-the frequency and the natural history of non-pulmonary soft tissue metastases. Ann Surg Oncol 6:389–394 Cheng EY, Springfield DS, Mankin HJ, 1995, Frequent incidence of extrapulmonary sites of initial metastasis in patients with liposarcoma. Cancer 75:1120–1127 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 107 EP 110 DI 10.1007/s12253-010-9349-4 PG 4 ER PT J AU Chen, ChJ Hsu, HT Lin, MT Pintye, M Chen, JR AF Chen, Chih-Jung Hsu, Hui-Ting Lin, Ming-Tsan Pintye, Mariann Chen, Jim-Ray TI Renal Cell Carcinoma with t(X;17)(p11.2;q25) in a 5-year-old Taiwanese Boy. A Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports C1 [Chen, Chih-Jung] Changhua Christian Hospital, Department of PathologyChanghua, Taiwan, Republic of China. [Hsu, Hui-Ting] Changhua Christian Hospital, Department of PathologyChanghua, Taiwan, Republic of China. [Lin, Ming-Tsan] Changhua Christian Hospital, Department of PediatricChanghua, Taiwan, Republic of China. [Pintye, Mariann] Chang Gung Memorial Hospital at Keelung, Department of Pathology, No.222, Meijin RdKeelung, Taiwan, Republic of China. [Chen, Jim-Ray] Chang Gung Memorial Hospital at Keelung, Department of Pathology, No.222, Meijin RdKeelung, Taiwan, Republic of China. RP Chen, JR (reprint author), Chang Gung Memorial Hospital at Keelung, Department of Pathology, Keelung, Taiwan, Republic of China. EM jimrchen@cgmh.org.tw CR Eble JN, Sauter G, Epstein J et al, 2004, WHO Classification of Tumours. Pathology and genetics of tumours of the urinary system and male genital organs. IARC Press, Lyon, pp 37–38 Argani P, Ladanyi M, 2005, Translocation carcinomas of the kidney. Clin Lab Med 25:363–378 Camparo P, Vasiliu V, Molinie V et al, 2008, Renal translocation carcinomas: clinicopathologic, immunohistochemical, and gene expression profiling analysis of 31 cases with a review of the literature. Am J Surg Pathol 32:656–670 Weterman MA, Wilbrink M, Geurts van Kessel A, 1996, Fusion of the transcription factor TFE3 gene to a novel gene, PRCC, in t, X;1)(p11;q21)-positive papillary renal cell carcinomas. Proc Natl Acad Sci USA 93:15294–15298 Clark J, Lu YJ, Sidhar SK et al, 2007, Fusion of splicing factor genes PSF and NonO, p54nrb, to the TFE3 gene in papillary renal cell carcinoma. Oncogene 15:2233–2239 Argani P, Antonescu A, Illei PB, Lui MY et al, 2001, Primary renal neoplasms with the ASPL-TFE3 gene fusion of alveolar soft part sarcoma: a distinctive tumor entity previously included among renal cell carcinomas of children and adolescents. Am J Pathol 159:179–192 Argani P, Lui MY, Couturier J et al, 2003, A novel CLTC-TFE3 gene fusion in pediatric renal adenocarcinoma with t(X;17)(p11.2; q23). Oncogene 22:5374–5378 Davis IJ, Hsi BL, Arroyo JD et al, 2003, Cloning of an Alpha- TFEB fusion in renal tumors harboring the t(6;11)(p21;q13, chromosome translocation. Proc Natl Acad Sci USA 100:6051– 6056 Argani P, Olgac S, Tickoo SK et al, 2007, Xp11 translocation renal cell carcinoma in adults: expanded clinical, pathologic, and genetic spectrum. Am J Surg Pathol 31:1149–1160 Indolfi P, Terenziani M, Casale F et al, 2003, Renal cell carcinoma in children: a clinicopathologic study. J Clin Oncol 21:530–535 Geller JI, Dome JS, 2004, Local lymph node involvement does not predict poor outcome in pediatric renal cell carcinoma. Cancer 101:1575–1183 Selle B, Furtwangler R, Graf N et al, 2006, Population-based study of renal cell carcinoma in children in Germany, 1980–2005: more frequently localized tumors and underlying disorders compared with adult counterparts. Cancer 107:2906–2914 Ramphal R, Pappo A, Zielenska M et al, 2006, Pediatric renal cell carcinoma: clinical, pathologic, and molecular abnormalities associated with the members of the mit transcription factor family. Am J Clin Pathol 126:349–364 Geller JI, Argani P, Adeniran A et al, 2008, Translocation renal cell carcinoma: lack of negative impact due to lymph node spread. Cancer 112:1607–1616 Goto S, Ikeda K, Nakagawara A et al, 1986, Renal cell carcinoma in Japanese children. J Urol 136:1261–1263 Tsai HL, Chin TW, Chang JW et al, 2006, Renal cell carcinoma in children and young adults. J Chin Med Assoc 69:240–244 Chen CK, Hsueh C, Lai JY et al, 2007, Renal cell carcinoma with t(X;17)(p11.2;q25, in a 6-year-old Taiwanese boy. Virchows Arch 450:215–219 Tomlinson GE, Nisen PD, Timmons CF et al, 1991, Cytogenetics of a renal cell carcinoma in a 17-month-old child. Evidence for Xp11.2 as a recurring breakpoint. Cancer Genet Cytogenet 57:11– 17 Hernandez-Marti MJ, Orellana-Alonso C, Badia-Garrabou L et al, 1995, Renal adenocarcinoma in an 8-year-old child, with a t, X;17)(p11.2;q25). Cancer Genet Cytogenet 83:82–83 Dijkhuizen T, van den Berg E, Wilbrink M et al, 1995, Distinct Xp11.2 breakpoints in two renal cell carcinomas exhibiting X; autosome translocations. Genes Chromosom Cancer 14:43–50 Sidhar SK, Clark J, Gill S et al, 1996, The t(X;1)(p11.2;q21.2, translocation in papillary renal cell carcinoma fuses a novel gene PRCC to the TFE3 transcription factor gene. Hum Mol Genet 5:1333–1338 Bruder E, Passera O, Harms D et al, 2004, Morphologic and molecular characterization of renal cell carcinoma in children and young adults. Am J Surg Pathol 28:1117–1132 Argani P, Lae M, Ballard ET et al, 2006, Translocation carcinomas of the kidney after chemotherapy in childhood. J Clin Oncol 24:1529–1534 Argani P, 2006, The evolving story of renal translocation carcinomas. Am J Clin Pathol 126:332–334 Argani P, Antonescu CR, Couturier J et al, 2002, PRCC-TFE3 renal carcinomas: morphologic, immunohistochemical, ultrastructural, and molecular analysis of an entity associated with the t, X;1)(p11.2;q21). Am J Surg Pathol 26:1553–1566 Argani P, Lae M, Hutchinson B et al, 2005, Renal carcinomas with the t(6;11)(p21;q12): clinicopathologic features and demonstration of the specific alpha-TFEB gene fusion by immunohistochemistry, RT-PCR, and DNA PCR. Am J Surg Pathol 29:230– 240 Jiang Z, Woda BA, Rock KL et al, 2001, P504S: a new molecular marker for the detection of prostate carcinoma. Am J Surg Pathol 25:1397–1404 Paner GP, Srigley JR, Radhakrishnan A et al, 2006, Immunohistochemical analysis of mucinous tubular and spindle cell carcinoma and papillary renal cell carcinoma of the kidney: significant immunophenotypic overlap warrants diagnostic caution. Am J Surg Pathol 30:13–19 Tsuda M, Davis IJ, Argani P et al, 2007, TFE3 fusions activate MET signaling by transcriptional up-regulation, defining another class of tumors as candidates for therapeutic MET inhibition. Cancer Res 67:919–929 Chang KL, Folpe AL, 2001, Diagnostic utility of microphthalmia transcription factor in malignant melanoma and other tumors. Adv Anat Pathol 8:273–275 Hemesath TJ, Steingrimsson E, McGill G et al, 1994, Microphthalmia, a critical factor in melanocyte development, defines a discrete transcription factor family. Genes Dev 8:2770–2780 Mansky KC, Sulzbacher S, Purdom G et al, 2002, The microphthalmia transcription factor and the related helix-loop-helix zipper factors TFE-3 and TFE-C collaborate to activate the tartrate-resistant acid phosphatase promoter. J Leukoc Biol 71:304–310 Lae M, Argani P, Olshen AB et al, 2004, Global gene expression profiles of renal carcinomas with Xp11 translocations, TFE3 gene fusions, suggest a closer relationship to alveolar soft sarcoma than to adult renal cell carcinoma. Mod Pathol 17(S1):163A Lam JS, Shvarts O, Pantuck AJ, 2004, Changing concepts in the surgical management of renal cell carcinoma. Eur Urol 45:692– 705 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2012 VL 18 IS 1 BP 111 EP 116 DI 10.1007/s12253-010-9353-8 PG 6 ER PT J AU Liu, Ch AF Liu, Chibo TI Serum Amyloid A Protein in Clinical Cancer Diagnosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Serum amyloid A; SAA; Cancer diagnosis; Proteomics; Inflammation; SELDI-TOF MS ID Serum amyloid A; SAA; Cancer diagnosis; Proteomics; Inflammation; SELDI-TOF MS AB The serum amyloid A (SAA) protein is an acute phase protein that is synthesized under the regulation of inflammatory cytokines during both acute and chronic inflammation. It is suggested that the SAA increases correlate with many types of carcinogenesis and neoplastic diseases. Th changes in SAA in serum could therefore indicate the progress and malignancy of the disease, as well as the host responses. The present paper reviewed the rationale of using SAA as potential cancer biomarker in clinical diagnosis, including the contribution and involvement of SAA in cancer growth and development. Then we discussed the current applications of SAA in diagnosis and tracing of different types of cancers. Finally the proteomics techniques, especially the SELDI-TOF MS to identify SAA in serum from patients were appreciated as an important manner in clinical diagnosis. C1 [Liu, Chibo] Taizhou Municipal Hospital, Department of Clinical Laboratory, 318000 Taizhou, Zhejiang, China. RP Liu, Ch (reprint author), Taizhou Municipal Hospital, Department of Clinical Laboratory, 318000 Taizhou, China. EM chibo_liu@163.com CR Cecconi D, Palmieri M, Donadelli M, 2011, Proteomics in pancreatic cancer research. Proteomics 11:816–828 van Gorp T, Cadron I, Vergote I, 2011, The utility of proteomics in gynecologic cancers. Curr Opin Obstet Gynecol 23:3–7 Hamrita B, Nasr HB, Chahed K, Chouchane L, 2011, Proteomics approaches: new technologies and clinical applications in breast carcinomas. Gulf J Oncolog 1:36–44 Gemoll T, Roblick UJ, Auer G, Jornvall H, Habermann JK, 2010, SELDI-TOF serum proteomics and colorectal cancer: a current overview. Arch Physiol Biochem 116:188–196 Matta A, Ralhan R, Desouza LV, Siu KW, 2010, Mass spectrometry-based clinical proteomics: head-and-neck cancer biomarkers and drug-targets discovery. Mass Spectrom Rev 29:945–961 Plymoth A, Hainaut P, 2011, Proteomics beyond proteomics: toward clinical applications. Curr Opin Oncol 23:77–82 Sipe J, 1999, Revised nomenclature for serum amyloid A, SAA). Nomenclature Committee of the International Society of Amyloidosis. Part 2. Amyloid 6:67–70 Malle E, de Beer FC, 1996, Human serum amyloid A, SAA, protein: a prominent acute-phase reactant for clinical practice. Eur J Clin Invest 26:427–435 Yamada T, 2006, Serum amyloid A, SAA)–pathogenicity and implication of appearance in plasma. Rinsho Byori 54:509–512 Barcelo JR, Munoz A, MANE JM et al, 2003, Amyloidosis and lung cancer. Clin Lung Canc 4:249–251, discussion 252 Comenzo RL, 2006, Amyloidosis. Curr Treat Options Oncol 7:225–236 Kwon AH, Tsuji K, Yamada H, Okazaki K, Sakaida N, 2007, Amyloidosis of the gallbladder mimicking gallbladder cancer. J Gastroenterol 42:261–264 Al Mofleh IA, 2008, Severe acute pancreatitis: pathogenetic aspects and prognostic factors. World J Gastroenterol 14:675–684 Hilliquin P, 1995, Biological markers in inflammatory rheumatic diseases. Cell Mol Biol, Noisy-le-grand, 41:993–1006 Rau BM, 2007, Predicting severity of acute pancreatitis. Curr Gastroenterol Rep 9:107–115 Rau B, Schilling MK, Beger HG, 2004, Laboratory markers of severe acute pancreatitis. Dig Dis 22:247–257 D’Eril GM, Anesi A, Maggiore M, Leoni V, 2001, Biological variation of serum amyloid A in healthy subjects. Clin Chem 47:1498–1499 Hijmans W, Sipe JD, 1979, Levels of the serum amyloid A protein, SAA, in normal persons of different age groups. Clin Exp Immunol 35:96–100 Biran H, Friedman N, Neumann L, Pras M, Shainkin-Kestenbaum R, 1986, Serum amyloid A, SAA, variations in patients with cancer: correlation with disease activity, stage, primary site, and prognosis. J Clin Pathol 39:794–797 Chan DC, Chen CJ, Chu HC et al, 2007, Evaluation of serum amyloid A as a biomarker for gastric cancer. Ann Surg Oncol 14:84–93 O’Hanlon DM, Lynch J, Cormican M, Given HF, 2002, The acute phase response in breast carcinoma. Anticancer Res 22:1289–1293 Raynes JG, Cooper EH, 1983, Comparison of serum amyloid A protein and C-reactive protein concentrations in cancer and nonmalignant disease. J Clin Pathol 36:798–803 Preciado-Patt L, Levartowsky D, Prass M, Hershkoviz R, Lider O, Fridkin M, 1994, Inhibition of cell adhesion to glycoproteins of the extracellular matrix by peptides corresponding to serum amyloid A. Toward understanding the physiological role of an enigmatic protein. Eur J Biochem 223:35–42 Urieli-Shoval S, Shubinsky G, Linke RP, Fridkin M, Tabi I, Matzner Y, 2002, Adhesion of human platelets to serum amyloid A. Blood 99:1224–1229 Preciado-Patt L, Hershkoviz R, Fridkin M, Lider O, 1996, Serum amyloid A binds specific extracellular matrix glycoproteins and induces the adhesion of resting CD4+ T cells. J Immunol 156:1189–1195 Brown EJ, 1986, The role of extracellular matrix proteins in the control of phagocytosis. J Leukoc Biol 39:579–591 Chen ST, Pan TL, Juan HF, Chen TY, Lin YS, Huang CM, 2008, Breast tumor microenvironment: proteomics highlights the treatments targeting secretome. J Proteome Res 7:1379–1387 Garlanda C, Maina V, Martinez de la Torre Y, Nebuloni M, Locati M, 2008, Inflammatory reaction and implantation: the new entries PTX3 and D6. Placenta 29(Suppl B):129–134 Mas VR, Fisher RA, Archer KJ, Maluf DG, 2009, Proteomics and liver fibrosis: identifying markers of fibrogenesis. Expert Rev Proteomics 6:421–431 Uhlar CM, Whitehead AS, 1999, Serum amyloid A, the major vertebrate acute-phase reactant. Eur J Biochem 265:501–523 Agaram N, Shia J, Klimstra DS et al, 2005, Globular hepatic amyloid: a diagnostic peculiarity that bears clinical significance. Hum Pathol 36:845–849 Alessi P, Leali D, Camozzi M, Cantelmo A, Albini A, Presta M, 2009, Anti-FGF2 approaches as a strategy to compensate resistance to anti-VEGF therapy: long-pentraxin 3 as a novel antiangiogenic FGF2-antagonist. Eur Cytokine Netw 20:225–234 Bareggi SR, Cornelli U, 2010, Clioquinol: review of its mechanisms of action and clinical uses in neurodegenerative disorders. CNS Neurosci Ther Cho WC, 2007, Nasopharyngeal carcinoma: molecular biomarker discovery and progress. Mol Canc 6:1 Cho WC, Yip TT, Yip C et al, 2004, Identification of serum amyloid a protein as a potentially useful biomarker to monitor relapse of nasopharyngeal cancer by serum proteomic profiling. Clin Canc Res 10:43–52 Cho WC, Yip TT, Cheng WW, Au JS, 2010, Serum amyloid A is elevated in the serum of lung cancer patients with poor prognosis. Br J Canc 102:1731–1735 Cocco E, Bellone S, El-Sahwi K et al, 2009, Serum amyloid A, SAA): a novel biomarker for uterine serous papillary cancer. Br J Canc 101:335–341 Cocco E, Bellone S, El-Sahwi K et al, 2010, Serum amyloid A: a novel biomarker for endometrial cancer. Cancer 116:843–851 Cohen AD, Comenzo RL, 2010, Systemic light-chain amyloidosis: advances in diagnosis, prognosis, and therapy. Am Soc Hematol Educ Program 2010:287–294 Comenzo RL, 2007, Current and emerging views and treatments of systemic immunoglobulin light-chain, Al, amyloidosis. Contrib Nephrol 153:195–210 Cremona M, Calabro E, Randi G et al, 2010, Elevated levels of the acute-phase serum amyloid are associated with heightened lung cancer risk. Cancer 116:1326–1335 Dai S, Wang X, Liu L et al, 2007, Discovery and identification of Serum Amyloid A protein elevated in lung cancer serum. Sci China C Life Sci 50:305–311 Junker K, Von Eggeling F, Muller J, Steiner T, Schubert J, 2006, Identification of biomarkers and therapeutic targets for renal cell cancer using ProteinChip technology. Urologe A 45:305–306, 308, 310–12 passim Klock G, Baiersdorfer M, Koch-Brandt C, 2009, Chapter 7: Cell protective functions of secretory Clusterin, sCLU). Adv Canc Res 104:115–138 Le Y, Yang Y, Cui Y et al, 2002, Receptors for chemotactic formyl peptides as pharmacological targets. Int Immunopharmacol 2:1–13 Le L, Chi K, Tyldesley S et al, 2005, Identification of serum amyloid A as a biomarker to distinguish prostate cancer patients with bone lesions. Clin Chem 51:695–707 Liu DH, Wang XM, Zhang LJ et al, 2007, Serum amyloid A protein: a potential biomarker correlated with clinical stage of lung cancer. Biomed Environ Sci 20:33–40 Malle E, Sodin-Semrl S, Kovacevic A, 2009, Serum amyloid A: an acute-phase protein involved in tumour pathogenesis. Cell Mol Life Sci 66:9–26 Marshall FF, 2005, Serum protein profiling by SELDI mass spectrometry: detection of multiple variants of serum amyloid alpha in renal cancer patients. J Urol 173:1919–1920 Maru Y, 2009, Logical structures extracted from metastasis experiments. Canc Sci 100:2006–2013 Meethal SV, Smith MA, Bowen RL, Atwood CS, 2005, The gonadotropin connection in Alzheimer’s disease. Endocrine 26:317–326 Merchant S, Sun J, Korbelik M, 2007, Dying cells program their expedient disposal: serum amyloid P component upregulation in vivo and in vitro induced by photodynamic therapy of cancer. Photochem Photobiol Sci 6:1284–1289 Michaeli A, Finci-Yeheskel Z, Dishon S, Linke RP, Levin M, Urieli-Shoval S, 2008, Serum amyloid A enhances plasminogen activation: implication for a role in colon cancer. Biochem Biophys Res Commun 368:368–373 Moshkovskii SA, Serebryakova MV, Kuteykin-Teplyakov KB et al, 2005, Ovarian cancer marker of 11.7 kDa detected by proteomics is a serum amyloid A1. Proteomics 5:3790–3797 Moshkovskii SA, Vlasova MA, Pyatnitskiy MA et al, 2007, Acute phase serum amyloid A in ovarian cancer as an important component of proteome diagnostic profiling. Proteomics Clin Appl 1:107–117 Okutani D, 2006, The role of long pentraxin 3, a new inflammatory mediator in inflammatory responses. Nihon Rinsho Meneki Gakkai Kaishi 29:107–113 Pierce BL, Neuhouser ML, Wener MH et al, 2009, Correlates of circulating C-reactive protein and serum amyloid A concentrations in breast cancer survivors. Breast Canc Res Treat 114:155–167 Sasazuki S, Inoue M, Sawada N et al, 2010, Plasma levels of Creactive protein and serum amyloid A and gastric cancer in a nested case-control study: Japan Public Health Center-based prospective study. Carcinogenesis 31:712–718 Segal BH, Kwon-Chung J, Walsh TJ et al, 2006, Immunotherapy for fungal infections. Clin Infect Dis 42:507–515 Tolson J, Bogumil R, Brunst E et al, 2004, Serum protein profiling by SELDI mass spectrometry: detection of multiple variants of serum amyloid alpha in renal cancer patients. Lab Invest 84:845–856 Uys A, Rapoport BL, Fickl H, Meyer PW, Anderson R, 2007, Prediction of outcome in cancer patients with febrile neutropenia: comparison of the Multinational Association of Supportive Care in Cancer risk-index score with procalcitonin, C-reactive protein, serum amyloid A, and interleukins-1beta, -6, -8 and −10. Eur J Cancer Care, Engl, 16:475–483 Vermaat JS, van der Tweel I, Mehra N et al, 2010, Two-protein signature of novel serological markers apolipoprotein-A2 and serum amyloid alpha predicts prognosis in patients with metastatic renal cell cancer and improves the currently used prognostic survival models. Ann Oncol 21:1472–1481 Wall JS, Paulus MJ, Gleason S, Gregor J, Solomon A, Kennel SJ, 2006, Micro-imaging of amyloid in mice. Meth Enzymol 412:161–182 Wang CS, Sun CF, 2009, C-reactive protein and malignancy: clinico-pathological association and therapeutic implication. Chang Gung Med J 32:471–482 Wang XD, Lu DH, Song H et al, 2009, Value of combining 64 multi-slice spiral computer tomography and serum amyloid A protein in surgical decision-making in rectal cancer. Zhonghua Wai Ke Za Zhi 47:1693–1697 Wood SL, Rogers M, Cairns DA et al, 2010, Association of serum amyloid A protein and peptide fragments with prognosis in renal cancer. Br J Canc 103:101–111 Yokoi K, Shih LC, Kobayashi R et al, 2005, Serum amyloid A as a tumor marker in sera of nude mice with orthotopic human pancreatic cancer and in plasma of patients with pancreatic cancer. Int J Oncol 27:1361–1369 Hutchens TW, Yip TT, 1993, New desorption strategies for the mass spectrometric analysis of macromolecules. Rapid Commun Mass Spectrom 7:576–580 Zhou M, Veenstra T, 2008, Mass spectrometry: m/z 1983–2008. Biotechniques 44(667–8):670 Clarke CH, Buckley JA, Fung ET, 2005, SELDI-TOF-MS proteomics of breast cancer. Clin Chem Lab Med 43:1314–1320 Engwegen JY, Gast MC, Schellens JH, Beijnen JH, 2006, Clinical proteomics: searching for better tumour markers with SELDI-TOF mass spectrometry. Trends Pharmacol Sci 27:251–259 Fung ET, Enderwick C, 2002, ProteinChip clinical proteomics: computational challenges and solutions. Biotechniques Suppl 34– 8:40–41 Garrisi VM, Abbate I, Quaranta M, Mangia A, Tommasi S, Paradiso A, 2008, SELDI-TOF serum proteomics and breast cancer: which perspective? Expert Rev Proteomics 5:779–785 Kislinger T, Humeny A, Pischetsrieder M, 2004, Analysis of protein glycation products by matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Curr Med Chem 11:2185–2193 Naishiro Y, Suzuki C, Kimura M et al, 2007, Plasma analysis of rheumatoid arthritis by SELDI. Nihon Rinsho Meneki Gakkai Kaishi 30:145–150 Wiltfang J, Lewczuk P, Riederer P et al, 2005, Consensus paper of the WFSBP Task Force on Biological Markers of Dementia: the role of CSF and blood analysis in the early and differential diagnosis of dementia. World J Biol Psychiatr 6:69–84 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 117 EP 121 DI 10.1007/s12253-011-9459-7 PG 5 ER PT J AU Suba, Zs AF Suba, Zsuzsanna TI Interplay Between Insulin Resistance and Estrogen Deficiency as co- Activators in Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Insulin resistance; Estrogen deficiency; Carcinogenesis; Obesity; Type-2 diabetes; Growth factor; menopause; sexual steroid; Cardiovascular disease ID Insulin resistance; Estrogen deficiency; Carcinogenesis; Obesity; Type-2 diabetes; Growth factor; menopause; sexual steroid; Cardiovascular disease AB Both insulin resistance and estrogen deficiency result in complex metabolic disorder based mainly on defective cellular glucose uptake and on an atherogenic serum lipid profile. These alterations may be regarded as high risks for several life-threatening human diseases, such as type-2 diabetes, cardiovascular lesions and malignancies. Insulin resistance and estrogen deficiency are concomitant disorders with mutual interrelationship. Insulin resistance and the compensatory hyperinsulinemia provoke increased androgen synthesis at the expense of decreased estrogen production. Similarly, a moderate or severe decrease in serum estrogen levels enhances the prevalence of insulin resistant states both in men and women. Healthy premenopausal women enjoy the defensive effect of estrogens against metabolic and hormonal disorders. However, even a slight decrease in their circulatory estrogen levels associated with insulin resistance may increase the risk for cancers, particularly in the organs having high estrogen demand (breast, endometrium and ovary). On the other hand, postmenopausal state with profound estrogen deficiency confers high risk for cancers in different organs with either high or moderate estrogen demand. After menopause, hormone replacement therapy improves insulin sensitivity and decreases the enhanced inclination to malignancies in postmenopausal women. Recognition of the thorough interplay between insulin resistance and estrogen deficiency may illuminate many apparently controversial experimental and clinical findings concerning cancer development and therapeutic possibilities. Moreover, their interactions in the initiation and progression of human malignancies may supply new strategies in primary cancer prevention and cancer cure. C1 [Suba, Zsuzsanna] National Institute of Oncology, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. RP Suba, Zs (reprint author), National Institute of Oncology, 1122 Budapest, Hungary. EM subazdr@gmail.com CR Kahn CR, Folli F, 1993, Molecular determinant of insulin action. Hormon Res 39:93–101 Reaven GM, 1988, Banting lecture 1988: Role of insulin resistance in human disease. Diabetes 37:1595–1607 Suba Zs, 2009, Insulin resistance, hyperinsulinemia and cancer risk. Chapter 2, pp 19–40. In: Estrogen versus Cancer. Nova Science Publishers Inc, New York Bjornstrom L, Sjoberg M, 2005, Mechanisms of estrogen receptor signaling: Convergence of genomic and nongenomic actions on target genes. Mol Endocrinol 19:833–842 Reckelhoff JF, 2005, Sex steroids, cardiovascular disease, and hypertension. Hypertension 45:170–174 Suba Zs, 2009, Insulin resistance, estrogen deficiency and cancer risk. Chapter 7. In: Estrogen versus Cancer. Nova Science Publishers Inc. New York Ehrmann DA, Barnes RB, Rosenfield RL, Cavaghan MK, Imperial J, 1999, Prevalence of impaired glucose tolerance and diabetes in women with polycystic ovary syndrome. Diabetes Care 22:141–146 Faustini-Fustini M, Rochira V, Carani C, 1999, Oestrogen deficiency in men: where are we today? Eur J Endocrinol 140:111–129 Schneider JG, Tompkins C, Blumenthal RS, Mora S, 2006, The metabolic syndrome in women. Cardiol Rev 14:286–291 Wu ShI, Chou P, Tsai ShT, 2001, The impact of years since menopause on the development of impaired glucose tolerance. J Clin Epidemiol 54:117–120 Nestler JE, Jakubowicz DJ, DdeVargas AF, Brik C, Quintero N, Medina F, 1998, Insulin stimulates testosterone biosynthesis by human thecal cells from women polycystic ovary syndrome by activating its own receptor and using inositolglycan mediators as the signal transduction system. J Clin Endocrinol Metab 83:2001–2005 DeFronzo RA, Ferrannini E, 1991, Insulin resistance: a multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia and atherosclerotic cardiovascular disease. Diabetes Care 14:173–194 Bloomgarden ZT, 2005, Second World Congress on the Insulin Resistance Syndrome: mediators, pediatric insulin resistance, the polycystic ovary syndrome, and malignancy. Diabetes Care 28:1821–1830 Wrighting Group for the Women’s Health Initiative Investigators, 2002, Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA 288:321– 333 Suba Z, 2007, Gender-related hormonal risk factors for oral cancer. Pathol Oncol Res 13:195–202 Suba Z, 2010, Common soil of smoking-associated and hormone-related cancers: estrogen deficiency. Oncol Rev 4:73– 87 Ragaz J, Wilson K, Frohlich J, Muraca G, Budlovsky J, 2010, Dual estrogen effects on breast cancer: endogenous estrogen stimulates, exogenous estrogen protects. Further investigation of estrogen chemoprevention is warranted. Cancer Res 70(suppl 24):441 Gupta K, Krishnaswamy G, Karnad A, Peiris A, 2002, Insulin: a novel factor in carcinogenesis. Am J Med Sci 323:140–145 Yu H, Rohan Th, 2000, Role of insulin-like growth factor family in cancer development and progression. J Natl Cancer Inst 92:1472–1489 Muti P, Quattrin T, Grant BJ, Krogh V, Micheli A, Schunemann HJ, Ram M, Freudenheim JL, Sieri S, Trevisan M, Berrino F, 2002, Fasting glucose is a risk factor for breast cancer. Cancer Epidemiol Biomark & Prev 11:1361–1368 Balkau B, Kahn H, 2001, HS: Hyperinsulinemia predicts fatal liver cancer but is inversely associated with fatal cancer at some other sites: The Paris Prospective Study. Diabetes Care 24:843– 849 McKeown-Eyssen, 1994, Epidemiology of colorectal cancer revisited: are serum triglycerides and/or plasma glucose associated with risk? Cancer Epidemiol Biomark and Prev 3:687–695 Yeung NG, Husain I, Waterfall N, 2003, Diabetes mellitus and bladder cancer. An epidemiological relationship? Pathol Oncol Res 9:30–31 Barnard RJ, Aronson WJ, Tymchuk CN, Ngo TH, 2002, Prostate cancer: another aspect of the insulin-resistance syndrome? Obes Rev 3:303–308 Ujpal M, Matos O, Bibok Gy, Somogyi A, Szabo Gy, Suba Z, 2004, Diabetes and oral tumors in Hungary. Epidemiological correlations. Diabetes Care 27:770–774 Suba Z, Barabas J, Szabo Gy, Takacs D, Ujpal M, 2005, Increased prevalence of diabetes and obesity in patients with salivary gland tumors. Diabetes Care 28:228 Key TJ, Applyby PN, Reeves GK, 2003, Body mass index, serum sex hormones and breast cancer risk in postmenopausal women. J Natl Cancer Inst 95:1218–1226 Soliman PT, Oh JC, Schmeler KM, Sun CC, Slomovitz BM, Gershenson DM, Burke TW, Lu KH, 2005, Risk factors for young premenopausal women with endometrial cancer. Obstet Gynecol 105:575–580 Cowey S, Hardy RW, 2006, The metabolic syndrome: a highrisk state for cancer? Am J Pathol 169:1505–1522 Beatson TG, 1896, On the treatment of inoperable cases of carcinoma of the mamma. Suggestion for a new treatment with illustrative cases. Lancet 2:104–107 Berrino F, Muti P, Micheli A, Bolelli G, Krogh V, Scijano R et al, 1996, Serum sex hormone levels after menopause and subsequent breast cancer. J Natl Cancer Inst 88:291–296 La Vecchia C, Brinton LA, McTiernan A, 2001, Menopause, hormone replacement therapy and cancer. Maturitas 39:97–115 Feigelson HS, Henderson BE, 1996, Estrogens and breast cancer. Carcinogenesis 17:2279–2284 Colditz GA, Hankinson SE, Hunter DJ, Willett WC, Manson JE, Stampfer MJ et al, 1995, The use of estrogens and progestins and the risk of breast cancer in postmenopausal women. N Engl J Med 332:1589–1593 Diamanti-Kandarakis E, 2004, Hormone replacement therapy and risk of malignancy. Curr Opinion Obstet Gynecol 16:73–78 Key TJ, Pike MC, 1988, The dose-effect relationship between’unopposed’ oestrogens and endometrial mitotic rate: its central role in explaining and predicting endometrial cancer risk. Br J Cancer 57:205–212 Toniolo PG, Levitz M, Zeleniuch-Jacquotte A, Banerjee S, Koenig KL, Shore RE, Strax P, Pasternak BS, 1995, A prospective study of endogenous estrogens and breast cancer in post-menopausal women. J Natl Cancer Inst 86:1076–1082 Henderson BE, Ross RK, Pike MC, 1993, Hormonal chemoprevention of cancer in women. Science 259:633–638 Hulka BS, Liu ET, Lininger RA, 1993, Steroid hormones and risk of breast cancer. Cancer 74:1111–1124 Pike M, Bernstein L, Spicer D, 1993, Exogenous hormones and breast cancer risk. In: Neiderhuber J, ed, Current Therapy in Oncol BC Decker. St. Louis, MO, pp 292–302 Liehr JG, 2000, Is estradiol a genotoxic and mutagenic carcinogen? Endocrine Rev 21:40–54 Greenwald P, Caputo TA, Wolfgang PE, 1977, Endometrial cancer after menopausal use of estrogens. Obstet Gynecol 50:239–243 Rodriguez C, Patel AV, Calle EE, Jacob EJ, 2001, Estrogen replacement therapy and ovarian cancer mortality in a large prospective study of US women. JAMA 285:1460–1465 Negri E, Tzonou A, Beral V et al, 1999, Hormonal therapy for menopause and ovarian cancer in a collaborative re-analysis of European studies. Int J Cancer 80:848–85l Lacey JV, Mink PJ, Lubin JH, Sherman ME, Troisi R, Hartge P, Schatzkin A, Schairer C, 2002, Menopausal hormone replacement therapy and risk of ovarian cancer. JAMA 288:334–341 International Agency for Research on Cancer, 1999, Monographs on the evaluation of carcinogenic risks to humans: hormonal contraception and postmenopausal hormone therapy vol. 72. IARC, Lyon, France Russo J, Fernandez SV, Russo PA, Fernbaugh R, Sheriff FS, Lareef HM, Garber J, Russo IH, 2006, 17-beta-estradiol induces transformation and tumorigenesis in human breast epithelial cells. FASEB J 20:1622–1634 Dickson RB, Gelmann EP, Knabbe C, Jasid K, Bates S, Swain S, 1987, Mechanisms of estrogenic and antiestrogenic regulation of growth of human breast carcinoma. In: Klijn JGM, ed, Hormonal manipulation of cancer: peptides, growth factors and new, anti)steroidal agents. Raven, New York, pp 381–403 Katzenellenbogen BS, 1996, Estrogen receptors: bioactivity and interactions with cell signaling pathways. Biol Reprod 54:287– 293 Ames BN, Shigenaga MK, Gold LS, 1993, DNA lesions, inducible DNA repair and cell division: three key factors in mutagenesis and carcinogenesis. Environ Perspect 101:35–44 Li JJ, Li SA, 1990, Estrogen carcinogenesis in hamster tissues: a critical review. Endocr Rev 11:524–531 Nandi S, 1978, Role of hormones in mammary neoplasia. Cancer Res 38:4046–4049 Clark RB, Howell A, Potten CS, Anderson E, 1997, Dissociation between steroid receptor expression and cell proliferation in human breast. Cancer Res 57:4987–4991 Ashburn SP, Han X, Liehr JG, 1993, Microsomal hydroxylation of 2- and 4-fluorestradiol to catechol metabolites and their conversion to methyl ethers: catechol estrogens as possible mediators of hormonal carcinogenesis. Mol Pharmacol 43:534–541 Ursin G, London S, Stanczyk FZ, Gentzschein E, Paganini, Hill A, Ross RK, Pike MC, 1999, Ureinary 2-hydroxyestrone/16 alphahydroxyestrone ratio and risk of breast cancer in postmenopausal women. J Natl Cancer Inst 91:1067–1072 Salama SA, Kamel M, Awad M, Nasser AH, Al-Hendy A, Botting S, Arrastia C Catecholestrogens induce oxidative stress and malignant transformation in human endometrial glandular cells: protective effect of Catechol-O-methyltransferase Tsutsui T, Barrett JC, 1997, Neoplastic transformation of cultured mammalian cells by estrogens and estrogen-like chemicals. Environ Health Perspect 105(suppl3):619–624 Banerjee SK, Banerjee S, Li SA, Li JJ, 1994, Induction of chromosome aberrations in Syrian hamster renal cortical cells by various estrogens. Mutat Res 311:191–197 Barrett JC, Wong A, McLachlan JA, 1981, Diethylstilbestrol induces neoplastic transformation without measurable gene mutation at two loci. Science 212:1402–1404 Mobley JA, Bhat AS, Brueggemeier RW, 1999, Measurement of oxidative DNA damage by catechol estrogens and analogues in vitro. Chem Res Toxicol 12:270–277 Michos C, Kiortsis DN, Evangelou A, Karkabounas S, 2006, Antioxidant protection during menstrual cycle: the effects of estradiol on ascorbic-dehydroascorbic acid plasma levels and total antioxidant plasma status in eumenorrhoic women during menstrual cycle. Acta Obstet Gynecol Scand 85:960–965 Freedman ND, Lacey JV, Hollenbeck AR, Leitzmann MF, Schatzkin A, Abnet ChC, 2010, The association of menstrual and reproductive factors with upper gastrointestinal tract cancers in the NIH-AARP Cohort. Cancer 116:1572–1581 McGrath M, Michaud DS, De Vivo I, 2006, Hormonal and reproductive factors and the risk of bladder cancer in women. Am J Epidemiol 163:236–244 La Vecchia C, D’Avanzo B, Franceschi S, Negri E, Parazzini F, Decarli A, 1994, Menstrual and reproductive factors and gastriccancer in women. Int J Cancer 59:761–764 Britt K, Ashworth A, Smalley M, 2007, Pregnancy and the risk of breast cancer. Endocrine-Related Cancer 14:907–933 Wolpert BJ, Amr S, Ezzat S, Saleh D, Gouda I, Loay I et al, 2010, Estrogen exposure and bladder cancer risk in Egyptian women. Maturitas Teras LR, Patel AV, Rodriguez C, Thun MJ, Calle EE, 2005, Parity, other reproductive factors, and risk of pancreatic cancer mortality in a large cohort of U.S. women. Cancer Causes and Control 16:1035–40 Bosetti C, Negri E, Franceschi S, Conti E, Levi F, Tomei F, La Vecchia C, 2000, Risk factors for oral and pharyngeal cancer in women: a study from Italy and Switzerland. Br J Cancer 82:204– 207 Suba Zs, 2009, Estrogen deficiency as cancer risk factor. Chapter 5. pp 85–106. In: Estrogen versus Cancer. Nova Science Publishers Inc, New York Olsson H, Bladstrom AM, Ingvar C, 2003, Are smokingassociated cancers prevented or postponed in women using hormone replacement therapy? Obstet Gynecol 102:565–570 Gallus S, Bosetti C, Franceschi S, Levi F, Simonato L, Negri E, La Vecchia C, 2001, Oesophageal cancer in women: tobacco, alcohol, nutritional and hormonal factors. Br J Cancer 85:341–345 Parazzini F, La Vecchia C, Negri E, Franceschi S, Bolis G, 1997, Case-control study of estrogen replacement therapy and risk of cervical cancer. Br Med J 315:85–88 Fernandez E, Gallus S, Bosetti C, Franceschi S, Negri E, La Vecchia C, 2003, Hormone replacement therapy and cancer risk: a systematic analysis from a network of case-control studies. Int J Cancer 105:408–412 Kreuzer M, Gerken M, Heinrich J, Kreienbrock L, Wichmann HE, 2003, Hormonal factors and risk of lung cancer among women? Int J Epidemiol 32:263–271 Anderson GL, Judd HL, Kaunitz AM, Barad DH, Beresford SA, Pettinger M, Liu J, McNeeley SG, Lopez AM, 2003, Women’s Health Initiative Investigators. Effects of estrogen plus progestin on gynaecologic cancers and associated diagnostic procedures: The Women’s Health Initiative randomized trial. JAMA 290:1739–1748 Schapiro S, 2007, False alarm: postmenopausal hormone therapy and ovarian cancer. Climacteric 10:466–470 Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA, Black H, Bonds D et al, 2004, Women’s Health Intitative Steering Committee. Effects of conjugated equine estrogen in postmenopausal women with hysterctomy: the Women’s Health Intitative randomized controlled trial. JAMA 291:1701–1712 LaCroix AZ, Chlebowski RT, Manson JE, Aragaki AK, Johnson KC, Martin L et al, 2011, Health outcomes after stopping conjugated equine estrogens among postmenopausal women with prior hysterectomy. A randomized controlled trial. JAMA 305:1305–1314 Nilsson S, Makela S, Treuter E, Tujague J, Thomsen J, Andersson G, Enmark E, Pettersson K, Warner M, Gustafsson JA, 2001, Mechanisms of estrogen action. Physiol Rev 81:1535–1565 Hall JM, McDonnell DP, 1999, The estrogen receptor β-isoform, ERβ, of the human estrogen receptor modulates ERα transcriptional activity and is a key regulator of the cellular response to estrogens and antiestrogens. Endocrinol 140:5566–5578 Segars J, Driggers P, 2002, Estrogen action and cytoplasmic signaling cascades. Part I: Membrane-associated signaling complexes. Trends Endocrin Metab 13:349–354 Helguero LA, Faulds MH, Gustafsson JA, Haldosen LA, 2005, Estrogen receptors α, ERα, and β, ERβ, differentially regulate proliferation and apoptosis of the normal murine mammary epithelial cell line HC11. Oncogene 24:6605–6616 Suba Zs, 2011, Antiestrogen or estrogen as anticancer drug. Ch. 4. In: Hoffmann AB, ed, Sex Hormones, Development, Regulation and Disorders. Nova Science Publishers Inc, New York Razandi M, Pedram A, Greene GL, Levin ER, 1999, Cell membrane and nuclear estrogen receptors, ERs, originate from a single transcript: studies of ERα and ERβ expressed in Chinese hamster ovary cells. Mol Endocrinol 13:307–319 Driggers PH, Segars JH, 2002, Estrogen action and cytoplasmic signaling pathways. Part II: The role of growth factors and phosphorylation in estrogen signaling. Trends Endocrin Metab 13:422–427 Kato S, Endoh H, Masuhiro Y, Kitamoto T, Uchiyama S, Sasaki H, Masushige S, Gotoh Y, Nishida E, Kawashima H, Metzger D, Chambon P, 1995, Activation of the estrogen receptor though phosphorylation by mitogen-activated protein kinase. Science 270:1491–1494 Lundholt BK, Briand P, Lykkesfeldt AE, 2001, Growth inhibition and growth stimulation by estradiol of estrogen receptor transfected human breast epithelial cell lines involve different pathways. Breast Cancer Res Treat 67:199–214 Massarweh S, Schiff R, 2006, Resistance to endocrine therapy in breast cancer: exploiting estrogen receptor/growth factor crosstalk. Endocrin-Related Cancer Suppl 13(1):S15–24 Marquez-Garban DC, Chen HW, Fishbein MC, Goodglick L, Pietras RJ, 2007, Estrogen receptor signaling pathways in human non-small cell lung cancer. Steroids 72:135–143 Nelson KG, Takahashi T, Bossert NL, Walmer DK, McLachlan JA, 1991, Epidermal growth factor replaces estrogen in the stimulation of female genital-tract growth and differentiation. Proceed Natl Acad Sci USA 88:21–25 Curtis SW, Washburn T, Sewall C, DiAugustine R, Lindzey J, Couse JF, Korach KS, 1996, Physiological coupling of growth factor and steroid receptor signaling pathway: estrogen receptor knockout mice lack estrogen-like response to epidermal growth factor. Proceed Natl Acad Sci USA 93:12626–12630 Barros RPA, Machado UF, Gustafsson JA, 2006, Estrogen receptors: new players in diabetes mellitus. Trends Molecular Med 9:425–431 Choi SB, Jang JS, Park S, 2005, Estrogen and exercise may enhance β-cell function and mass via insulin receptor substrate 2 induction in ovarectomized diabetic rats. Endocrinology 146:4786–4794 Bryzgalova G, Gao H, Ahren B, Zierath JR, Galuska D, Steiler TL et al, 2006, Evidence that oestrogen receptor-alpha plays an important role in the regulation of glucose homeostasis in mice: insulin sensitivity in the liver. Diabetologia 49:588–597 Welch RD, Gorski J, 1999, Regulation of glucose transporters by estradiol in the immature rat uterus. Endocrinology 140:3602–3608 Godsland IF, Walton C, Stevenson JC, 1993, Impact of menopause on metabolism. In: Diamond MP, Naftolin K, eds, Metabolism in the female life cycle. Ares Seerono Symposia, Rome, pp 171–189 Jones ME, Thorburn AW, Britt KL, Hewitt KN, Wreford NG, Proietto J, Oz OK, Leury BJ, Robertson KM, Yao S, Simpson ER, 2000, Aromatase-deficient, ArKO, mice have a phenotype of increased adiposity. Proc Natl Acad Sci USA 97:12735– 12740 Muraki K, Okuya S, Tanizawa Y, 2006, Estrogen receptor alpha regulates insulin sensitivity trough IRS-1 thyrosin phosphorylation in mature 3 T3-L1 adipocytes. Endocr J 53:841–851 Shi J, Simpkins JW, 1997, 17β-Estradiol modulation of glucose transporter 1 expression in blood-brain barrier. Am J Physiol 272:1016–1022 Rosenbaum D, Haber RS, Dunaif A, 1993, Insulin resistance in polycystic ovary syndrome: decreased expression of GLUT-4 glucose transporters in adipocytes. Am J Physiol 264:197–202 Nagira K, Sasaoka T, Wada T, Fukui K, Ikubo M, Hori S, Tsuneki H, Saito S, Kobayashi M, 2006, Altered subcellular distribution of estrogen receptor is implicated in estradiolinduced dual regulation of insulin signaling in 3 T3-L1 adipocytes. Endocrinology 147:1020–1028 Leung KC, Johannson G, Leung GM, Ho KKY, 2004, Estrogen regulation of growth hormone action. Endocrine Rev 25:693– 721 Walsh BW, Schiff I, Rosner B, Greenberg L, Ravinkar V, Sacks FM, 1991, Effects of postmenopausal estrogen replacement on the concentrations and metabolism of plasma lipoproteins. New England J Med 325:1196–1204 Hamden K, Carreau S, Ellouz F, Masmoudi H, El FA, 2007, Protective effect of 17beta-estradiol on oxidative stress and liver dysfunction in aged male rats. J Physiol Biochem 63:195–201 Croston GE, Milan LB, Marschke KB, Reichman M, Briggs MR, 1997, Androgen receptor-mediated antagonism of estrogendependent low density lipoprotein receptor transcription in cultured hepatocytes. Endocrinology 138:3779–3786 Pallottini V, Bulzomi P, Galluzzo P, Martini C, Marino M, 2008, Estrogen regulation of adipose tissue functions: Involvement of estrogen receptor isoforms. Infectious Disorders – Drug Targets 8:52–60 Macotela Y, Boucher J, Tran TT, Kahn CR, 2009, Sex and depot differences in adipocyte insulin sensitivity and glucose. Diabetes 58:803–812 Poehlman ET, Toth MJ, Gardner AW, 1995, Changes in energy balance and body composition at menopause. Ann Intern Med 123:673–675 Suba Z, 2009, Correlations between obesity and risk for salivary gland tumors. Cancer Res J 3:55–68 Calle EE, Kaaks R, 2004, Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer 4:579–591 Baghaei F, Rosmond R, Westberg L, Hellstrand M, Eriksson E, Holm G, Bjorntorp P, 2003, The CYP19 gene and association with androgens and abdominal obesity in premenopausal women. Obesity Res 11:578–585 Bjorntorp P, 1996, The regulation of adipose tissue distribution in humans. Int J Obes Relat Metab Disord 20:291–302 Sartor BM, Dickey RP, 2005, Polycystic ovarian syndrome and the metabolic syndrome. Am J Med 330:336–342 Cara JF, Rosenfield RL, 1988, Insulin-like growth factor I and insulin potentiate luteinizing hormone induced androgen synthesis by rat ovarian thecal-interstitial cells. Endrocrinology 123:733–739 Attia GR, Rainey WE, Carr BR, 2001, Metformin directly inhibits androgen production in human thecal cells. Fertil Steril 76:517–524 Moghetti P, Castello R, Negri C, Tosi F, Spiazzi GG, Brun E, Balducci R, Toscano V, Muggeo M, 1996, Insulin infusion amplifies 17-(alpha)-hydroxy- corticosteroid intermediates response to adrenocorticotropin in hyperandrogenicwomen: apparent relative impairment of 17,20-lyase activity. J Clin Endocrinol Metab 81:881–886 Christakou C, Diamanti-Kandarakis E, 2008, Role of androgen excess on metabolic aberrations and cardiovascular risk in women with polycystic ovary syndrome. Women’s Health 4:583–594 Solomon CG, Hu FB, Dunaif A, Rich-Edwards J, Willett WC, Hunter DJ, Colditz GA, Speizer FE, Manson JE, 2001, Long or highly irregular menstrual cycles as a marker for risk of type-2 diabetes mellitus. JAMA 286:2421–2426 Elting MW, Dorsen TJM, Bezemer PD, Schoemaker J, 2001, Prevalence of diabetes mellitus, hypertension and cardiac complaints in a follow-up study of a Dutch PCOS population. Hum Reprod 16:556–560 Gadducci A, Gargini A, Palla E, Fanucchi A, Genazzani AR, 2005, Polycystic ovary syndrome and gynecological cancers: is there a link? Gynecol Endocrinol 20:200–208 Pierpoint T, McKeigue PM, Isaacs AJ, Wild SH, Jacobs HS, 1998, Mortality of women with polycystic ovary syndrome at long-term follow-up. J Clin Epidemiol 51:581–586 King H, Rewers M, 1993, WHO Ad Hoc Diabetes Reporting Group: Global estimates for prevalence of diabetes mellitus and impaired glucose tolerance in adults. Diabetes Care 21:157–177 Wilson PWE, 1998, Does estrogen reduce glycemic levels? Diabetes Care 21:1585–1586 Hirose K, Toyama T, Iwata H, Takezaki T, Hamajima N, Tajima K, 2003, Insulin, insulin-like growth factor-1 and breast cancer risk in Japanese women. Asian Pac J Cancer Prev 4:239–246 Goodwin PJ, Boyd NF, Hanna W, Hartwick W, Murray D, Qizilbash A, Redwood S, Hood N, DelGiudice ME, Sidlofsky S, McGready D, Wilkinson R, Mahoney L, Connelly P, Page DL, 1997, Elevated levels of plasma triglycerides are associated with histologically defined premenopausal breast cancer risk. Nutr Cancer 27:284–292 Godsland IF, 2005, Oestrogens and insulin secretion. Diabetologia 48:2213–2220 Proudler AJ, Felton CV, Stevenson JC, 1992, Aging and the response of plasma insulin, glucose and C-peptide concentrations to intravenous glucose in postmenopausal women. Clin Sci 83:489–494 Gohlke-Barwolf C, 2000, Coronary artery disease: is menopause a risk factor? Basic Res Cardiol 95:177–183 Rapp K, Schroeder J, Klenk J, Ulmer H, Concin H, Diem G, Oberaigner W, Weiland SK, 2006, Fasting blood glucose and cancer risk in a cohort of more than 140,000 adults in Austria. Diabetologia 49:945–952 Morimoto LM, White E, Chen Z, Chlebowski RT, Hays J, Lopez AM, Manson J, Margolis KL, Muti PC, Stefanick ML, McTiernan A, 2002, Obesity, body size and risk of postmenopausal breast cancer: the Women’s Health Initiative, United States). Cancer Causes Control 13:741–751 McCullough ML, Patel AV, Patel R, Rodriguez C, Feigelson HS, Bandera EV, Gansler T, 2008, Body mass and endometrial cancer risk by hormone replacement therapy and cancer subtype. Cancer Epidemiol Biomark Prev 17:73–79 Takacs D, Koppany F, Mihalyi S, Suba Z, 2011, Decreased oral cancer risk by moderate alcohol consumption in non-smoker postmenopausal women. Oral Oncol 47:537–540 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 123 EP 133 DI 10.1007/s12253-011-9466-8 PG 11 ER PT J AU Koutsaki, M Zaravinos, A Spandidos, AD AF Koutsaki, Maria Zaravinos, Apostolos Spandidos, A Demetrios TI Modern Trends into the Epidemiology and Screening of Ovarian Cancer. Genetic Substrate of the Sporadic Form SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Ovarian cancer; Risk factors; Symptoms; Screening; Oncogenes and onco-suppressor genes ID Ovarian cancer; Risk factors; Symptoms; Screening; Oncogenes and onco-suppressor genes AB Ovarian cancer (OC) is a heterogeneous disease, including a broad spectrum of histological subtypes and demonstrating diverse biological behavior. Epithelialderived ovarian malignant tumours constitute the predominant and most lethal form of the disease. Age, genetic predisposition, gynecological and reproductive factors and environmental factors are the main risk factors that increase the risk for acquiring OC. Vaginal examination, ultrasonography and measurement of blood serum levels of tumour markers, especially CA125 constitute the first-line screening modalities for OC, whereas second-line testing involves more accurate imaging techniques such as color Doppler ultrasound of the lesion or/and a CT scan. Sex steroid hormone pathway genes, cell cycle genes, DNA repair genes, oncogenes and onco-suppressor genes have been associated with a genetic susceptibility to sporadic OC. In the present review we focus on the major oncogenes and onco-suppressor genes in the sporadic form of the disease. Each tumour subtype is associated with a unique molecular signature, as revealed by current genetic and biomarker profiling studies. Different OC pathways emerge early in the process of carcinogenesis, ultimately leading to clinically different tumour types. As mutations acquired early during tumourigenesis will be present in all later stages, largescale gene expression profiling using DNA microarray analysis techniques can help to classify ovarian cancers into clinically relevant subtypes. C1 [Koutsaki, Maria] University of Crete, Medical School, Department of Virology, 71110 Heraklion, Crete, Greece. [Zaravinos, Apostolos] University of Crete, Medical School, Department of Virology, 71110 Heraklion, Crete, Greece. [Spandidos, A Demetrios] University of Crete, Medical School, Department of Virology, 71110 Heraklion, Crete, Greece. RP Spandidos, AD (reprint author), University of Crete, Medical School, Department of Virology, 71110 Heraklion, Greece. EM spandidos@spandidos.gr CR Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics. CA Cancer J Clin 60:277–300 Ferlay J, Parkin DM, Steliarova-Foucher E, 2008, Estimates of cancer incidence and mortality in Europe in. Eur J Cancer 46:765–781 Goodman MT, Howe HL, Tung KH, Hotes J, Miller BA et al, 2003, Incidence of ovarian cancer by race and ethnicity in the United States, 1992–1997. Cancer 97:2676–2685 Coleman MP, Esteve J, Damiecki P, Arslan A, Renard H, 1993, Trends in cancer incidence and mortality. IARC Sci Publ 1–806 Kliewer EV, Smith KR, 1995, Ovarian cancer mortality among immigrants in Australia and Canada. Cancer Epidemiol Biomarkers Prev 4:453–458 Cannistra SA, 2004, Cancer of the ovary. N Engl J Med 351:2519–2529 Titus-Ernstoff L, Perez K, Cramer DW, Harlow BL, Baron JA et al, 2001, Menstrual and reproductive factors in relation to ovarian cancer risk. Br J Cancer 84:714–721 Silverberg SG, 2000, Histopathologic grading of ovarian carcinoma: a review and proposal. Int J Gynecol Pathol 19:7–15 Yancik R, 1993, Ovarian cancer. Age contrasts in incidence, histology, disease stage at diagnosis, and mortality. Cancer 71:517–523 Pecorelli S, Benedet JL, Creasman WT, Shepherd JH, 1999, FIGO staging of gynecologic cancer. 1994–1997 FIGO Committee on Gynecologic Oncology. International Federation of Gynecology and Obstetrics. Int J Gynaecol Obstet 64:5–10 Bristow RE, Tomacruz RS, Armstrong DK, Trimble EL, Montz FJ, 2002, Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a metaanalysis. J Clin Oncol 20:1248–1259 Holschneider CH, Berek JS, 2000, Ovarian cancer: epidemiology, biology, and prognostic factors. Semin Surg Oncol 19:3–10 Engel J, Eckel R, Schubert-Fritschle G, Kerr J, Kuhn W et al, 2002, Moderate progress for ovarian cancer in the last 20 years: prolongation of survival, but no improvement in the cure rate. Eur J Cancer 38:2435–2445 Husseinzadeh N Status of tumor markers in epithelial ovarian cancer has there been any progress? A review. Gynecol Oncol 120:152–157 Boyd J, Rubin SC, 1997, Hereditary ovarian cancer: molecular genetics and clinical implications. Gynecol Oncol 64:196–206 Goldberg JM, Piver MS, Jishi MF, Blumenson L, 1997, Age at onset of ovarian cancer in women with a strong family history of ovarian cancer. Gynecol Oncol 66:3–9 Lynch HT, Casey MJ, Snyder CL, Bewtra C, Lynch JF et al, 2009, Hereditary ovarian carcinoma: heterogeneity, molecular genetics, pathology, and management. Mol Oncol 3:97–137 Easton DF, Ford D, Bishop DT, 1995, Breast and ovarian cancer incidence in BRCA1-mutation carriers. Breast Cancer Linkage Consortium. Am J Hum Genet 56:265–271 Easton DF, Steele L, Fields P, Ormiston W, Averill D et al, 1997, Cancer risks in two large breast cancer families linked to BRCA2 on chromosome 13q12-13. Am J Hum Genet 61:120–128 Ford D, Easton DF, Bishop DT, Narod SA, Goldgar DE, 1994, Risks of cancer in BRCA1-mutation carriers. Breast Cancer Linkage Consortium. Lancet 343:692–695 Narod S, Ford D, Devilee P, Barkardottir RB, Eyfjord J et al, 1995, Genetic heterogeneity of breast-ovarian cancer revisited. Breast Cancer Linkage Consortium. Am J Hum Genet 57:957– 958 Narod SA, Ford D, Devilee P, Barkardottir RB, Lynch HT et al, 1995, An evaluation of genetic heterogeneity in 145 breastovarian cancer families. Breast Cancer Linkage Consortium. Am J Hum Genet 56:254–264 Schubert EL, Lee MK, Mefford HC, Argonza RH, Morrow JE et al, 1997, BRCA2 in American families with four or more cases of breast or ovarian cancer: recurrent and novel mutations, variable expression, penetrance, and the possibility of families whose cancer is not attributable to BRCA1 or BRCA2. Am J Hum Genet 60:1031–1040 Bewtra C, Watson P, Conway T, Read-Hippee C, Lynch HT, 1992, Hereditary ovarian cancer: a clinicopathological study. Int J Gynecol Pathol 11:180–187 Malander S, Rambech E, Kristoffersson U, Halvarsson B, Ridderheim M et al, 2006, The contribution of the hereditary nonpolyposis colorectal cancer syndrome to the development of ovarian cancer. Gynecol Oncol 101:238–243 Rubin SC, Blackwood MA, Bandera C, Behbakht K, Benjamin I et al, 1998, BRCA1, BRCA2, and hereditary nonpolyposis colorectal cancer gene mutations in an unselected ovarian cancer population: relationship to family history and implications for genetic testing. Am J Obstet Gynecol 178:670–677 Lynch HT, Casey MJ, Lynch J, White TE, Godwin AK, 1998, Genetics and ovarian carcinoma. Semin Oncol 25:265–280 Antoniou A, Pharoah PD, Narod S, Risch HA, Eyfjord JE et al, 2003, Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet 72:1117–1130 Franceschi S, La Vecchia C, Booth M, Tzonou A, Negri E et al, 1991, Pooled analysis of 3 European case–control studies of ovarian cancer: II. Age at menarche and at menopause. Int J Cancer 49:57–60 Purdie D, Green A, Bain C, Siskind V, Ward B et al, 1995, Reproductive and other factors and risk of epithelial ovarian cancer: an Australian case–control study. Survey of Women’s Health Study Group. Int J Cancer 62:678–684 Bristow RE, Karlan BY, 1996, Ovulation induction, infertility, and ovarian cancer risk. Fertil Steril 66:499–507 Rossing MA, Daling JR, Weiss NS, Moore DE, Self SG, 1994, Ovarian tumors in a cohort of infertile women. N Engl J Med 331:771–776 Vlahos NF, Economopoulos KP, Creatsas G Fertility drugs and ovarian cancer risk: a critical review of the literature. Ann N Y Acad Sci 1205:214–219 Lacey JV Jr, Mink PJ, Lubin JH, Sherman ME, Troisi R et al, 2002, Menopausal hormone replacement therapy and risk of ovarian cancer. Jama 288:334–341 Schildkraut JM, Schwingl PJ, Bastos E, Evanoff A, Hughes C, 1996, Epithelial ovarian cancer risk among women with polycystic ovary syndrome. Obstet Gynecol 88:554–559 Brinton LA, Gridley G, Persson I, Baron J, Bergqvist A, 1997, Cancer risk after a hospital discharge diagnosis of endometriosis. Am J Obstet Gynecol 176:572–579 Cramer DW, Welch WR, Cassells S, Scully RE, 1983, Mumps, menarche, menopause, and ovarian cancer. Am J Obstet Gynecol 147:1–6 Jelovac D, Armstrong DK Recent progress in the diagnosis and treatment of ovarian cancer. CA Cancer J Clin 61:183–203 Chionh F, Baglietto L, Krishnan K, English DR, MacInnis RJ, et al. Physical activity, body size and composition, and risk of ovarian cancer. Cancer Causes Control 21:2183–2194 Olsen CM, Green AC, Whiteman DC, Sadeghi S, Kolahdooz F et al, 2007, Obesity and the risk of epithelial ovarian cancer: a systematic review and meta-analysis. Eur J Cancer 43:690–709 Jordan SJ, Whiteman DC, Purdie DM, Green AC, Webb PM, 2006, Does smoking increase risk of ovarian cancer? A systematic review. Gynecol Oncol 103:1122–1129 CramerDW, Liberman RF, Titus-Ernstoff L,WelchWR, Greenberg ER et al, 1999, Genital talc exposure and risk of ovarian cancer. Int J Cancer 81:351–356 Cramer DW, Welch WR, Berkowitz RS, Godleski JJ, 2007, Presence of talc in pelvic lymph nodes of a woman with ovarian cancer and long-term genital exposure to cosmetic talc. Obstet Gynecol 110:498–501 Kurian AW, Balise RR, McGuire V, Whittemore AS, 2005, Histologic types of epithelial ovarian cancer: have they different risk factors? Gynecol Oncol 96:520–530 Tung KH, Goodman MT, Wu AH, McDuffie K, Wilkens LR et al, 2003, Reproductive factors and epithelial ovarian cancer risk by histologic type: a multiethnic case–control study. Am J Epidemiol 158:629–638 Webb PM, Purdie DM, Grover S, Jordan S, Dick ML et al, 2004, Symptoms and diagnosis of borderline, early and advanced epithelial ovarian cancer. Gynecol Oncol 92:232–239 Twombly R, 2007, Cancer killer may be “silent” no more. J Natl Cancer Inst 99:1359–1361 Guppy AE, Nathan PD, Rustin GJ, 2005, Epithelial ovarian cancer: a review of current management. Clin Oncol, R Coll Radiol, 17:399–411 Le T, Giede C, Salem S, Lefebvre G, Rosen B et al, 2009, Initial evaluation and referral guidelines for management of pelvic/ovarian masses. J Obstet Gynaecol Can 31:668–680 Badgwell D, Bast RC Jr, 2007, Early detection of ovarian cancer. Dis Markers 23:397–410 Rein BJ, Gupta S, Dada R, Safi J, Michener C, et al, 2011, Potential markers for detection and monitoring of ovarian cancer. J Oncol 475983 O’Brien TJ, Raymond LM, Bannon GA, Ford DH, Hardardottir H et al, 1991, New monoclonal antibodies identify the glycoprotein carrying the CA 125 epitope. Am J Obstet Gynecol 165:1857–1864 Yin BW, Dnistrian A, Lloyd KO, 2002, Ovarian cancer antigen CA125 is encoded by the MUC16 mucin gene. Int J Cancer 98:737–740 Gupta D, Lis CG, 2009, Role of CA125 in predicting ovarian cancer survival—a review of the epidemiological literature. J Ovarian Res 2:13 Rosen DG, Wang L, Atkinson JN, Yu Y, Lu KH et al, 2005, Potential markers that complement expression of CA125 in epithelial ovarian cancer. Gynecol Oncol 99:267–277 Hogdall EV, Christensen L, Kjaer SK, Blaakaer J, Kjaerbye- Thygesen A et al, 2007, CA125 expression pattern, prognosis and correlation with serum CA125 in ovarian tumor patients. From The Danish “MALOVA” Ovarian Cancer Study. Gynecol Oncol 104:508–515 Marth C, Zeimet AG, Bock G, Daxenbichler G, 1992, Modulation of tumour marker CA-125 expression in cultured ovarian carcinoma cells. Eur J Cancer 28A:2002–2006 Helzlsouer KJ, Bush TL, Alberg AJ, Bass KM, Zacur H et al, 1993, Prospective study of serum CA-125 levels as markers of ovarian cancer. Jama 269:1123–1126 Bast RC Jr, Badgwell D, Lu Z, Marquez R, Rosen D et al, 2005, New tumor markers: CA125 and beyond. Int J Gynecol Cancer 15(Suppl 3):274–281 Nakae M, Iwamoto I, Fujino T, Maehata Y, Togami S et al, 2006, Preoperative plasma osteopontin level as a biomarker complementary to carbohydrate antigen 125 in predicting ovarian cancer. J Obstet Gynaecol Res 32:309–314 Rosenthal AN, Menon U, Jacobs IJ, 2006, Screening for ovarian cancer. Clin Obstet Gynecol 49:433–447 Sjovall K, Nilsson B, Einhorn N, 2002, The significance of serum CA 125 elevation in malignant and nonmalignant diseases. Gynecol Oncol 85:175–178 Dorigo O, Berek JS Personalizing CA125 Levels for Ovarian Cancer Screening. Cancer Prev Res, Phila, 4:1356–1359 Skates SJ, Mai P, Horick NK, Piedmonte M, Drescher CW, et al. Large Prospective Study of Ovarian Cancer Screening in High- Risk Women: CA125 Cut-Point Defined by Menopausal Status. Cancer Prev Res, Phila, 4:1401–1408 Moore RG, Brown AK, Miller MC, Skates S, Allard WJ et al, 2008, The use of multiple novel tumor biomarkers for the detection of ovarian carcinoma in patients with a pelvic mass. Gynecol Oncol 108:402–408 Hellstrom I, Hellstrom KE, 2008, SMRP and HE4 as biomarkers for ovarian carcinoma when used alone and in combination with CA125 and/or each other. Adv Exp Med Biol 622:15–21 Hellstrom I, Raycraft J, Hayden-Ledbetter M, Ledbetter JA, Schummer M et al, 2003, The HE4, WFDC2, protein is a biomarker for ovarian carcinoma. Cancer Res 63:3695–3700 Huhtinen K, Suvitie P, Hiissa J, Junnila J, Huvila J et al, 2009, Serum HE4 concentration differentiates malignant ovarian tumours from ovarian endometriotic cysts. Br J Cancer 100:1315– 1319 Moore RG, McMeekin DS, Brown AK, DiSilvestro P, Miller MC et al, 2009, A novel multiple marker bioassay utilizing HE4 and CA125 for the prediction of ovarian cancer in patients with a pelvic mass. Gynecol Oncol 112:40–46 Skates SJ, Menon U, MacDonald N, Rosenthal AN, Oram DH et al, 2003, Calculation of the risk of ovarian cancer from serial CA- 125 values for preclinical detection in postmenopausal women. J Clin Oncol 21:206s–210s Skates S, Troiano R, Knapp RC, 2003, Longitudinal CA125 detection of sporadic papillary serous carcinoma of the peritoneum. Int J Gynecol Cancer 13:693–696 Hawkins RE, Roberts K, Wiltshaw E, Mundy J, Fryatt IJ et al, 1989, The prognostic significance of the half-life of serum CA 125 in patients responding to chemotherapy for epithelial ovarian carcinoma. Br J Obstet Gynaecol 96:1395–1399 Gadducci A, Zola P, Landoni F, Maggino T, Sartori E et al, 1995, Serum half-life of CA 125 during early chemotherapy as an independent prognostic variable for patients with advanced epithelial ovarian cancer: results of a multicentric Italian study. Gynecol Oncol 58:42–47 Jacobs I, Stabile I, Bridges J, Kemsley P, Reynolds C et al, 1988, Multimodal approach to screening for ovarian cancer. Lancet 1:268–271 Lerner JP, Timor-Tritsch IE, Federman A, Abramovich G, 1994, Transvaginal ultrasonographic characterization of ovarian masses with an improved, weighted scoring system. Am J Obstet Gynecol 170:81–85 Fishman DA, Cohen L, Blank SV, Shulman L, Singh D et al, 2005, The role of ultrasound evaluation in the detection of earlystage epithelial ovarian cancer. Am J Obstet Gynecol 192:1214– 1221, discussion 1221–1212 Van Nagell JR Jr, DePriest PD, Ueland FR, DeSimone CP, Cooper AL et al, 2007, Ovarian cancer screening with annual transvaginal sonography: findings of 25,000 women screened. Cancer 109:1887–1896 Valentin L, Skoog L, Epstein E, 2003, Frequency and type of adnexal lesions in autopsy material from postmenopausal women: ultrasound study with histological correlation. Ultrasound Obstet Gynecol 22:284–289 Murta EF, Nomelini RS, 2006, Early diagnosis and predictors of malignancy of adnexal masses. Curr Opin Obstet Gynecol 18:14–19 Braem MG, Schouten LJ, Peeters PH, den Brandt PA, Onland- Moret NC Genetic susceptibility to sporadic ovarian cancer: A systematic review. Biochim Biophys Acta 1816:132–146 Vella N, Aiello M, Russo AE, Scalisi A, Spandidos DA, et al. ‘Genetic profiling’ and ovarian cancer therapy, review). Mol Med Report 4:771–777 Fehrmann RS, Li XY, van der Zee AG, de Jong S, Te Meerman GJ et al, 2007, Profiling studies in ovarian cancer: a review. Oncologist 12:960–966 Spandidos DA, Dokianakis DN, Kallergi G, Aggelakis E, 2000, Molecular basis of gynecological cancer. Ann N Y Acad Sci 900:56–64 Koffa M, Spandidos DA, 1997, Oncogenes and onco-suppressor genes in female genital cancer. Ann N Y Acad Sci 816:347–355 Mammas IN, Zafiropoulos A, Spandidos DA, 2005, Involvement of the ras genes in female genital tract cancer. Int J Oncol 26:1241– 1255 Aunoble B, Sanches R, Didier E, Bignon YJ, 2000, Major oncogenes and tumor suppressor genes involved in epithelial ovarian cancer, review). Int J Oncol 16:567–576 Seidman JD, Kurman RJ, 1996, Subclassification of serous borderline tumors of the ovary into benign and malignant types. A clinicopathologic study of 65 advanced stage cases. Am J Surg Pathol 20:1331–1345 Smith Sehdev AE, Sehdev PS, Kurman RJ, 2003, Noninvasive and invasive micropapillary, low-grade, serous carcinoma of the ovary: a clinicopathologic analysis of 135 cases. Am J Surg Pathol 27:725–736 Shih Ie M, Kurman RJ, 2004, Ovarian tumorigenesis: a proposed model based on morphological and molecular genetic analysis. Am J Pathol 164:1511–1518 Karst AM, Drapkin R, 2010, Ovarian cancer pathogenesis: a model in evolution. J Oncol 932371 Zorn KK, Bonome T, Gangi L, Chandramouli GV, Awtrey CS et al, 2005, Gene expression profiles of serous, endometrioid, and clear cell subtypes of ovarian and endometrial cancer. Clin Cancer Res 11:6422–6430 Bonome T, Lee JY, Park DC, Radonovich M, Pise-Masison C et al, 2005, Expression profiling of serous low malignant potential, low-grade, and high-grade tumors of the ovary. Cancer Res 65:10602–10612 Peyssonnaux C, Eychene A, 2001, The Raf/MEK/ERK pathway: new concepts of activation. Biol Cell 93:53–62 Thomas EJ, Campbell IG, 2000, Molecular genetic defects in endometriosis. Gynecol Obstet Invest 50(Suppl 1):44–50 Singer G, Oldt R 3rd, Cohen Y,Wang BG, Sidransky D et al, 2003, Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst 95:484–486 Obata K, Morland SJ, Watson RH, Hitchcock A, Chenevix- Trench G et al, 1998, Frequent PTEN/MMAC mutations in endometrioid but not serous or mucinous epithelial ovarian tumors. Cancer Res 58:2095–2097 Shahbazian D, Roux PP, Mieulet V, Cohen MS, Raught B et al, 2006, The mTOR/PI3K and MAPK pathways converge on eIF4B to control its phosphorylation and activity. Embo J 25:2781–2791 Janda E, Lehmann K, Killisch I, Jechlinger M, Herzig M et al, 2002, Ras and TGF[beta] cooperatively regulate epithelial cell plasticity and metastasis: dissection of Ras signaling pathways. J Cell Biol 156:299–313 Sabbah M, Emami S, Redeuilh G, Julien S, Prevost G et al, 2008, Molecular signature and therapeutic perspective of the epithelialto- mesenchymal transitions in epithelial cancers. Drug Resist Updat 11:123–151 Skilling JS, Sood A, Niemann T, Lager DJ, Buller RE, 1996, An abundance of p53 null mutations in ovarian carcinoma. Oncogene 13:117–123 Kohler MF, Kerns BJ, Humphrey PA, Marks JR, Bast RC Jr et al, 1993, Mutation and overexpression of p53 in early-stage epithelial ovarian cancer. Obstet Gynecol 81:643–650 Kohler MF, Marks JR, Wiseman RW, Jacobs IJ, Davidoff AM et al, 1993, Spectrum of mutation and frequency of allelic deletion of the p53 gene in ovarian cancer. J Natl Cancer Inst 85:1513– 1519 Lafky JM, Wilken JA, Baron AT, Maihle NJ, 2008, Clinical implications of the ErbB/epidermal growth factor, EGF, receptor family and its ligands in ovarian cancer. Biochim Biophys Acta 1785:232–265 Ross JS, Yang F, Kallakury BV, Sheehan CE, Ambros RA et al, 1999, HER-2/neu oncogene amplification by fluorescence in situ hybridization in epithelial tumors of the ovary. Am J Clin Pathol 111:311–316 Bellacosa A, de Feo D, Godwin AK, Bell DW, Cheng JQ et al, 1995, Molecular alterations of the AKT2 oncogene in ovarian and breast carcinomas. Int J Cancer 64:280–285 Kiaris A, Spandios DA, 1995, Mutations of ras genes in human tumours. Int J Oncol 7:413–429 Zachos G, Spandidos DA, 1997, Expression of ras protooncogenes: regulation and implications in the development of human tumors. Crit Rev Oncol/Hematol 26:65–75 Bell DA, 2005, Origins and molecular pathology of ovarian cancer. Mod Pathol 18(Suppl 2):S19–32 Mayr D, Hirschmann A, Lohrs U, Diebold J, 2006, KRAS and BRAF mutations in ovarian tumors: a comprehensive study of invasive carcinomas, borderline tumors and extraovarian implants. Gynecol Oncol 103:883–887 Teneriello MG, Ebina M, Linnoila RI, Henry M, Nash JD et al, 1993, p53 and Ki-ras gene mutations in epithelial ovarian neoplasms. Cancer Res 53:3103–3108 Varras MN, Sourvinos G, Diakomanolis E, Koumantakis E, Flouris GA et al, 1999, Detection and clinical correlations of ras gene mutations in human ovarian tumors. Oncology 56:89–96 Zachos G, Spandidos DA, 1998, Transcriptional regulation of the c-H-ras1 gene by the P53 protein is implicated in the development of human endometrial and ovarian tumours. Oncogene 16:3013–3017 Boulalas I, Zaravinos A, Karyotis I, Delakas D, Spandidos DA, 2009, Activation of RAS family genes in urothelial carcinoma. J Urol 181:2312–2319 Zaravinos A, Bizakis J, Soufla G, Sourvinos G, Spandidos DA, 2007, Mutations and differential expression of the ras family genes in human nasal polyposis. Int J Oncol 31:1051–1059 Symvoulakis EK, Zaravinos A, Panutsopulos D, Zoras O, Papalambros E et al, 2007, Highly conserved sequence of exon 15 BRAF gene and KRAS codon 12 mutation among Greek patients with colorectal cancer. Int J Biol Markers 22:12–18 Zaravinos A, Kanellou P, Spandidos DAViral DNA detection and RAS mutations in actinic keratosis and nonmelanoma skin cancers. Br J Dermatol 162:325–331 Ratner E, Lu L, Boeke M, Barnett R, Nallur S, et al. A KRASvariant in ovarian cancer acts as a genetic marker of cancer risk. Cancer Res 70:6509–6515 Maihle NJ, Baron AT, Barrette BA, Boardman CH, Christensen TA et al, 2002, EGF/ErbB receptor family in ovarian cancer. Cancer Treat Res 107:247–258 Scambia G, Benedetti Panici P, Ferrandina G, Battaglia F, Distefano M et al, 1994, Significance of epidermal growth factor receptor expression in primary human endometrial cancer. Int J Cancer 56:26–30 Puputti M, Sihto H, Isola J, Butzow R, Joensuu H et al, 2006, Allelic imbalance of HER2 variant in sporadic breast and ovarian cancer. Cancer Genet Cytogenet 167:32–38 Vivanco I, Sawyers CL, 2002, The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer 2:489–501 Nakayama K, Nakayama N, Kurman RJ, Cope L, Pohl G et al, 2006, Sequence mutations and amplification of PIK3CA and AKT2 genes in purified ovarian serous neoplasms. Cancer Biol Ther 5:779–785 Cheng JQ, Godwin AK, Bellacosa A, Taguchi T, Franke TF et al, 1992, AKT2, a putative oncogene encoding a member of a subfamily of protein-serine/threonine kinases, is amplified in human ovarian carcinomas. Proc Natl Acad Sci USA 89:9267– 9271 Arboleda MJ, Lyons JF, Kabbinavar FF, Bray MR, Snow BE et al, 2003, Overexpression of AKT2/protein kinase Bbeta leads to up-regulation of beta1 integrins, increased invasion, and metastasis of human breast and ovarian cancer cells. Cancer Res 63:196–206 Piek JM, Kenemans P, Verheijen RH, 2004, Intraperitoneal serous adenocarcinoma: a critical appraisal of three hypotheses on its cause. Am J Obstet Gynecol 191:718–732 Corney DC, Flesken-Nikitin A, Choi J, Nikitin AY, 2008, Role of p53 and Rb in ovarian cancer. Adv Exp Med Biol 622:99–117 Sigal A, Rotter V, 2000, Oncogenic mutations of the p53 tumor suppressor: the demons of the guardian of the genome. Cancer Res 60:6788–6793 Crijns AP, Duiker EW, de Jong S, Willemse PH, van der Zee AG et al, 2006, Molecular prognostic markers in ovarian cancer: toward patient-tailored therapy. Int J Gynecol Cancer 16(Suppl 1):152–165 Kigawa J, Sato S, Shimada M, Takahashi M, Itamochi H et al, 2001, p53 gene status and chemosensitivity in ovarian cancer. Hum Cell 14:165–171 Langland GT, Yannone SM, Langland RA, Nakao A, Guan Y, et al. Radiosensitivity profiles from a panel of ovarian cancer cell lines exhibiting genetic alterations in p53 and disparate DNAdependent protein kinase activities. Oncol Rep 23:1021–1026 Sakurada A, Hamada H, Fukushige S, Yokoyama T, Yoshinaga K et al, 1999, Adenovirus-mediated delivery of the PTEN gene inhibits cell growth by induction of apoptosis in endometrial cancer. Int J Oncol 15:1069–1074 Levine RL, Cargile CB, Blazes MS, van Rees B, Kurman RJ et al, 1998, PTEN mutations and microsatellite instability in complex atypical hyperplasia, a precursor lesion to uterine endometrioid carcinoma. Cancer Res 58:3254–3258 Mutter GL, Lin MC, Fitzgerald JT, Kum JB, Baak JP et al, 2000, Altered PTEN expression as a diagnostic marker for the earliest endometrial precancers. J Natl Cancer Inst 92:924–930 Sato N, Tsunoda H, Nishida M, Morishita Y, Takimoto Y et al, 2000, Loss of heterozygosity on 10q23.3 and mutation of the tumor suppressor gene PTEN in benign endometrial cyst of the ovary: possible sequence progression from benign endometrial cyst to endometrioid carcinoma and clear cell carcinoma of the ovary. Cancer Res 60:7052–7056 Carser JE, Quinn JE, Michie CO, O’Brien EJ, McCluggage WG, et al. BRCA1 is both a prognostic and predictive biomarker of response to chemotherapy in sporadic epithelial ovarian cancer. Gynecol Oncol Weberpals JI, Tu D, Squire JA, Amin MS, Islam S, et al. Breast cancer 1, BRCA1, protein expression as a prognostic marker in sporadic epithelial ovarian carcinoma: an NCIC CTG OV.16 correlative study. Ann Oncol Zikan M, Pohlreich P, Freitag P, Janousek M, Pavlista D et al, 2008, Inactivation of BRCA1, BRCA2 and p53 genes in sporadic ovarian cancer. Ceska Gynekol 73:298–302 Clark-Knowles KV, O’Brien AM, Weberpals JI, 2010, BRCA1 as a Therapeutic Target in Sporadic Epithelial Ovarian Cancer. J Oncol 891059 Risch HA, Marrett LD, Howe GR, 1994, Parity, contraception, infertility, and the risk of epithelial ovarian cancer. Am J Epidemiol 140:585–597 Hankinson SE, Colditz GA, Hunter DJ,Willett WC, Stampfer MJ et al, 1995, A prospective study of reproductive factors and risk of epithelial ovarian cancer. Cancer 76:284–290 Adami HO, Hsieh CC, Lambe M, Trichopoulos D, Leon D et al, 1994, Parity, age at first childbirth, and risk of ovarian cancer. Lancet 344:1250–1254 Negri E, Franceschi S, Tzonou A, Booth M, La Vecchia C et al, 1991, Pooled analysis of 3 European case–control studies: I. Reproductive factors and risk of epithelial ovarian cancer. Int J Cancer 49:50–56 Rosenblatt KA, Thomas DB, 1993, Lactation and the risk of epithelial ovarian cancer. The WHO Collaborative Study of Neoplasia and Steroid Contraceptives. Int J Epidemiol 22:192–197 Pectasides D, Fountzilas G, Aravantinos G, Kalofonos HP, Efstathiou E et al, 2005, Advanced stage mucinous epithelial ovarian cancer: the Hellenic Cooperative Oncology Group experience. Gynecol Oncol 97:436–441 Ness RB, Grisso JA, Klapper J, Schlesselman JJ, Silberzweig S et al, 2000, Risk of ovarian cancer in relation to estrogen and progestin dose and use characteristics of oral contraceptives. SHARE Study Group. Steroid Hormones and Reproductions. Am J Epidemiol 152:233–241 Gross TP, Schlesselman JJ, 1994, The estimated effect of oral contraceptive use on the cumulative risk of epithelial ovarian cancer. Obstet Gynecol 83:419–424 Cramer DW, Harlow BL, Titus-Ernstoff L, Bohlke K, Welch WR et al, 1998, Over-the-counter analgesics and risk of ovarian cancer. Lancet 351:104–107 Yin L, Grandi N, Raum E, Haug U, Arndt V, et al. Meta-analysis: Circulating vitamin D and ovarian cancer risk. Gynecol Oncol 121:369–375 Hankinson SE, Hunter DJ, Colditz GA, Willett WC, Stampfer MJ et al, 1993, Tubal ligation, hysterectomy, and risk of ovarian cancer. A prospective study. Jama 270:2813–2818 Miracle-McMahill HL, Calle EE, Kosinski AS, Rodriguez C, Wingo PA et al, 1997, Tubal ligation and fatal ovarian cancer in a large prospective cohort study. Am J Epidemiol 145:349–357 Franceschi S, Parazzini F, Negri E, Booth M, La Vecchia C et al, 1991, Pooled analysis of 3 European case–control studies of epithelial ovarian cancer: III. Oral contraceptive use. Int J Cancer 49:61–65 Whittemore AS, Harris R, Itnyre J, 1992, Characteristics relating to ovarian cancer risk: collaborative analysis of 12 US case– control studies. II. Invasive epithelial ovarian cancers in white women. Collaborative Ovarian Cancer Group. Am J Epidemiol 136:1184–1203 Narod SA, Risch H, Moslehi R, Dorum A, Neuhausen S et al, 1998, Oral contraceptives and the risk of hereditary ovarian cancer. Hereditary Ovarian Cancer Clinical Study Group. N Engl J Med 339:424–428 Meijers-Heijboer H, van Geel B, van Putten WL, Henzen-Logmans SC, Seynaeve C et al, 2001, Breast cancer after prophylactic bilateral mastectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 345:159–164 Eisen A, Weber BL, 2001, Prophylactic mastectomy for women with BRCA1 and BRCA2 mutations–facts and controversy. N Engl J Med 345:207–208 Kauff ND, Satagopan JM, Robson ME, Scheuer L, Hensley M et al, 2002, Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 346:1609–1615 Rebbeck TR, Lynch HT, Neuhausen SL, Narod SA, Van’t Veer L et al, 2002, Prophylactic oophorectomy in carriers of BRCA1 or BRCA2 mutations. N Engl J Med 346:1616–1622 Fong PC, Boss DS, Yap TA, Tutt A, Wu P et al, 2009, Inhibition of poly(ADP-ribose, polymerase in tumors from BRCA mutation carriers. N Engl J Med 361:123–134 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 135 EP 148 DI 10.1007/s12253-011-9482-8 PG 14 ER PT J AU Halaszlaki, Cs Takacs, I Butz, H Patocs, A Lakatos, P AF Halaszlaki, Csaba Takacs, Istvan Butz, Henriett Patocs, Attila Lakatos, Peter TI Novel Genetic Mutation in the Background of Carney Complex SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Carney complex; Endocrine overactivity; Genetic study; Regulatory subunit 1A of the protein kinase A; Splice mutation ID Carney complex; Endocrine overactivity; Genetic study; Regulatory subunit 1A of the protein kinase A; Splice mutation AB Carney complex is a rare disease inherited in an autosomal dominant manner. It is mostly caused by inactivating mutations of the subunit of protein kinase A. Carney complex is associated with atrial myxoma, nevi or myxomas of the skin, breast tumor and endocrine overactivity. Primary pigmented nodular adrenocortical disease is the specific endocrine manifestation. The authors present the history of a 53-year-old female patient who had undergone surgery for atrial myxomas, thyroid tumor and breast cancer. She was also operated for an adrenal adenoma causing Cushing’s syndrome. Genetic study revealed a novel mutation in the regulatory subunit of protein kinase A (ivs2-1G>A splice mutation in intron 2). Her heterozygous twins were also genetically screened and one of them carried the same mutation. The authors emphasize that despite the absence of specific treatment for patients with Carney complex, confirmation of the diagnosis by genetic studies is important for the close follow-up of the patient and early identification of novel manifestations. C1 [Halaszlaki, Csaba] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor u. 2/A, 1083 Budapest, Hungary. [Takacs, Istvan] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor u. 2/A, 1083 Budapest, Hungary. [Butz, Henriett] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Patocs, Attila] Hungarian Academy of Sciences, Molecular Medicine Research Group, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Lakatos, Peter] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor u. 2/A, 1083 Budapest, Hungary. RP Halaszlaki, Cs (reprint author), Semmelweis University, 1st Department of Internal Medicine, 1083 Budapest, Hungary. EM drhalaszlaki@gmail.com CR Carney JA, Gordon H, Carpenter PC, Shenoy BV, Go VL, 1985, The complex of myxomas, spotty pigmentation, and endocrine overactivity. Medicine, Baltimore, 64(4):270–283 Stratakis CA, Kirschner LS, Carney JA, 2001, Clinical and molecular features of the Carney complex: diagnostic criteria and recommendations for patient evaluation. J Clin Endocrinol Metab 86(9):4041–4046 Stratakis CA, Kirschner LS, Carney JA, 1998, Carney complex: diagnosis and management of the complex of spotty skin pigmentation, myxomas, endocrine overactivity, and schwannomas. Am J Med Genet 80(2):183–185 Stratakis CA, Horvath A, 1993–2003 Feb 05, Carney Complex. In: Pagon RA BT, Dolan CR, Stephens K, ed, GeneReviews [Internet]. Seattle, WA). http://www.ncbi.nlm.nih.gov/books/ NBK1286/. Last update date:2010 Jun 22. Carney JA, Stratakis CA, 1998, Epithelioid blue nevus and psammomatous melanotic schwannoma: the unusual pigmented skin tumors of the Carney complex. Semin Diagn Pathol 15(3):216–224 Bertherat J, 2006, Carney complex, CNC). Orphanet J Rare Dis 1:21 Libe R, Horvath A, Vezzosi D, Fratticci A, Coste J, Perlemoine K, Ragazzon B, Guillaud-Bataille M, Groussin L, Clauser E, Raffin- Sanson ML, Siegel J, Moran J, Drori-Herishanu L, Faucz FR, Lodish M, Nesterova M, Bertagna X, Bertherat J, Stratakis CA, 2011, Frequent phosphodiesterase 11A gene, PDE11A, defects in patients with Carney complex, CNC, caused by PRKAR1A mutations: PDE11A may contribute to adrenal and testicular tumors in CNC as a modifier of the phenotype. J Clin Endocrinol Metab 96(1):E208–E214 Vezzosi D, Vignaux O, Dupin N, Bertherat J, 2010, Carney complex: clinical and genetic 2010 update. Ann Endocrinol, Paris, 71, 6):486–493 Rothenbuhler A, Stratakis CA, 2010, Clinical and molecular genetics of Carney complex. Best Pract Res Clin Endocrinol Metab 24(3):389–399 Horvath A, Bertherat J, Groussin L, Guillaud-Bataille M, Tsang K, Cazabat L, Libe R, Remmers E, Rene-Corail F, Faucz FR, Clauser E, Calender A, Bertagna X, Carney JA, Stratakis CA, 2010, Mutations and polymorphisms in the gene encoding regulatory subunit type 1-alpha of protein kinase A, PRKAR1A): an update. Hum Mutat 31(4):369–379 Pan L, Peng L, Jean-Gilles J, Zhang X, Wieczorek R, Jain S, Levine V, Osman I, Prieto VG, Lee P, 2010, Novel PRKAR1A gene mutations in Carney Complex. Int J Clin Exp Pathol 3(5):545–548 Storr HL, Metherell LA, Dias R, Savage MO, Rasmussen AK, Clark AJ, Main KM, 2010, Familial isolated primary pigmented nodular adrenocortical disease associated with a novel low penetrance PRKAR1A gene splice site mutation. Horm Res Paediatr 73, 2):115–119 Landry CS, Waguespack SG, Perrier ND, 2009, Surgical management of nonmultiple endocrine neoplasia endocrinopathies: stateof- the-art review. Surg Clin North Am 89(5):1069–1089 Szabo D, Zsippai A, Bendes M, Tombol Z, Szabo PM, Racz K, Igaz P, 2010, Pathogenesis of adrenocortical cancer. Orv Hetil, Hungarian Medical Journal, 151(29):1163–1170, In Hungarian) NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 149 EP 152 DI 10.1007/s12253-012-9502-3 PG 4 ER PT J AU Gurel, D Kargi, A Karaman, I Onen, A Unlu, M AF Gurel, Duygu Kargi, Aydanur Karaman, Ilgin Onen, Ahmet Unlu, Mehtat TI CD10 Expression in Epithelial and Stromal Cells of Non-small Cell Lung Carcinoma (NSCLC): A Clinic and Pathologic Correlation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD10; Carcinoma; Lung; Stromal cells; Epithelial cells ID CD10; Carcinoma; Lung; Stromal cells; Epithelial cells AB CD10 is a zinc dependent metallopeptidase, and its expression in stromal and/or epithelial cells of many carcinomas has been suggested to have prognostic value. This study investigates CD10 expression in epithelial and stromal cells of non small cell lung carcinoma (NSCLC), and evaluates its prognostic value for this tumor and its histologic subtypes. Sixty-six cases of NSCLC [35 cases of nonsquamous cell carcinoma (NSCC) and 31 cases of squamous cell carcinoma (SCC)] were analyzed immunohistochemically for CD10 antibody. Fisher’s exact test and univariate survival analyses were performed. Comparison of clinicopathologic characteristics for NSCLC showed that only stromal CD10 expression had worse prognostic impact, associated with the presence of recurrence (p=0.001), death (p=0.006) and disease positivity (p=0.001). For SCC, CD10 was found to be expressed mainly in the stromal cells, and was associated with a decreased survival (p=0.000) and disease free survival (p=0.000). CD10 expression was restricted to the epithelial cells in NSCC and associated with an increased disease free survival (p=0.036). Stromal CD10 expression apppears to be a worse prognostic factor in NSCLCs. CD10 which is expressed in different cell components of SCC and NSCC appears to have opposing effects on the behaviour of these histologic types. C1 [Gurel, Duygu] Dokuz Eylul University, Faculty of Medicine, Department of PathologyIzmir, Turkey. [Kargi, Aydanur] Dokuz Eylul University, Faculty of Medicine, Department of PathologyIzmir, Turkey. [Karaman, Ilgin] Dokuz Eylul University, Faculty of Medicine, Department of PathologyIzmir, Turkey. [Onen, Ahmet] Dokuz Eylul University, Faculty of Medicine, Department of Thoracic SurgeryIzmir, Turkey. [Unlu, Mehtat] Dokuz Eylul University, Faculty of Medicine, Department of PathologyIzmir, Turkey. RP Gurel, D (reprint author), Dokuz Eylul University, Faculty of Medicine, Department of Pathology, Izmir, Turkey. EM duygu.gurel@deu.edu.tr CR Shipp MA, Vijayaraghavan J, Schmidt EV et al, 1989, Common acute lymphoblastic leukemia antigen, CALLA, is active neutral endopeptidase 24.11, “enkephalinase”): direct evidence by cDNA transfection analysis. Proc Natl Acad Sci USA 86:297–301 Borscheri N, Roessner A, Rocken C, 2001, Canalicular immunostaining of neprilysin, CD10, as a diagnostic marker for hepatocellular carcinomas. Am J Surg Pathol 25:1297–1303 Xiao SY, Wang HL, Hart J et al, 2001, cDNA arrays and immunohistochemistry identification of CD10/CALLA expression in hepatocellular carcinoma. Am J Pathol 159:1415–1421 Xu Y, McKenna RW, Molberg KH et al, 2001, Clinicopathologic analysis of CD10+ and CD10- diffuse large B-cell lymphoma. Identification of a high-risk subset with coexpression of CD10 and bcl-2. Am J Clin Pathol 116(2):183–190 Xu Y, McKenna RW, Kroft SH, 2002, Comparison of multiparameter flow cytometry with cluster analysis and immunohistochemistry for the detection of CD10 in diffuse large B-Cell lymphomas. Mod Pathol 15:413–419 Albrecht M, Gillen S, Wilhelm B et al, 2002, Expression, localization and activity of neutral endopeptidase in cultured cells of benign prostatic hyperplasia and prostate cancer. J Urol 168:336–342 Chu P, Arber DA, 2000, Paraffin-section detection of CD10 in 505 nonhematopoietic neoplasms. Frequent expression in renal cell carcinoma and endometrial stromal sarcoma. Am J Clin Pathol 113:374–382 Moreau I, Duvert V, Caux C et al, 1993, Myofibroblastic stromal cells isolated from human bone marrow induce the proliferation of both early myeloid and B-lymphoid cells. Blood 82:2396–2405 Imai K, Kanzaki H, Fujiwara H et al, 1992, Expression of aminopeptidase N and neutral endopeptidase on the endometrial stromal cells in endometriosis and adenomyosis. Hum Reprod 7:1326–1328 Shipp MA, Tarr GE, Chen CY et al, 1991, CD10/neutral endopeptidase 24.11 hydrolyzes bombesin-like peptides and regulates the growth of small cell carcinomas of the lung. Proc Natl Acad Sci USA 88:10662–10666 Gohring B, Holzhausen HJ, Meye A et al, 1998, Endopeptidase 24.11/CD10 is down-regulated in renal cell cancer. Int J Mol Med 2:409–414 Koiso K, Akaza H, Ohtani M et al, 1994, A new tumor marker for bladder cancer. Int J Urol 1:33–36 Sato Y, Itoh F, Hinoda Y et al, 1996, Expression of CD10/neutral endopeptidase in normal and malignant tissues of the human stomach and colon. J Gastroenterol 31:12–17 Cohen AJ, Bunn PA, FranklinWet al, 1996, Neutral endopeptidase: variable expression in human lung, inactivation in lung cancer, and modulation of peptide-induced calcium flux. Cancer Res 56:831–839 Cohen AJ, Gilman LB, Moore M et al, 1996, Inactivation of neutral endopeptidase in lung cancer. Chest 109:12–13 Tawfic S, Niehans GA, Manivel JC, 2003, The pattern of CD10 expression in selected pathologic entities of the prostate gland. Hum Pathol 34:450–456 Bilalovic N, Sandstad B, Golouh R et al, 2004, CD10 protein expression in tumor and stromal cells of malignant melanoma is associated with tumor progression. Mod Pathol 17:1251–1258 Ogawa H, Iwaya K, Izumi M et al, 2002, Expression of CD10 by stromal cells during colorectal tumor development. Hum Pathol 33:806–811 Huang WB, Zhou XJ, Chen JY et al, 2005, CD10-positive stromal cells in gastric carcinoma: correlation with invasion and metastasis. Jpn J Clin Oncol 35:245–250 Braham H, Trimeche M, Ziadi S et al, 2006, CD10 expression by fusiform stromal cells in nasopharyngeal carcinoma correlates with tumor progression. Virchows Arch 449:220–224 Aiad HA, Hanout HM, 2007, Immunohistochemical expression of CD10 in cutaneous basal and squamous cell carcinomas. J Egypt Natl Canc Inst 19:195–201 Piattelli A, Fioroni M, Iezzi G et al, 2006, CD10 expression in stromal cells of oral cavity squamous cell carcinoma: a clinic and pathologic correlation. Oral Dis 12:301–304 Yada K, Kashima K, Daa T et al, 2004, Expression of CD10 in basal cell carcinoma. Am J Dermatopathol 26:463–471 Iwaya K, Ogawa H, Izumi M et al, 2002, Stromal expression of CD10 in invasive breast carcinoma: a new predictor of clinical outcome. Virchows Arch 440:589–593 Kristiansen G, Schluns K, Yongwei Yet al, 2002, CD10 expression in non-small cell lung cancer. Anal Cell Pathol 24:41–46 Tokuhara T, Adachi M, Hashida H et al, 2001, Neutral endopeptidase/CD10 and aminopeptidase N/CD13 gene expression as a prognostic factor in non-small cell lung cancer. Jpn J Thorac Cardiovasc Surg 49:489–496 Kajiyama H, Shibata K, Terauchi M et al, 2005, Neutral endopeptidase 24.11/CD10 suppresses progressive potential in ovarian carcinoma in vitro and in vivo. Clin Cancer Res 11:1798–1808 Ganju RK, Sunday M, Tsarwhas DG et al, 1994, CD10/NEP in non-small cell lung carcinomas. Relationship to cellular proliferation. J Clin Invest 94:1784–1791 Mountain CF, 1997, Revisions in the international system for staging lung cancer. Chest 111:1710–1717 Johnson AR, Ashton J, Schulz WW et al, 1985, Neutral metalloendopeptidase in human lung tissue and cultured cells. Am Rev Respir Dis 132:564–568 Makretsov NA, Hayes M, Carter BA et al, 2007, Stromal CD10 expression in invasive breast carcinoma correlates with poor prognosis, estrogen receptor negativity, and high grade. Mod Pathol 20:84–89 Basset P, Wolf C, Rouyer N et al, 1994, Stromelysin-3 in stromal tissue as a control factor in breast cancer behavior. Cancer 74:1045–1049 Talvensaari-Mattila A, Paakko P, Hoyhtya M et al, 1998, Matrix metalloproteinase-2 immunoreactive protein: a marker of aggressiveness in breast carcinoma. Cancer 83:1153–1162 Deschamps L, Handra-Luca A, O’Toole D et al, 2006, CD10 expression in pancreatic endocrine tumors: correlation with prognostic factors and survival. Hum Pathol 37:802–808 Abdou AG, 2007, CD10 expression in tumour and stromal cells of bladder carcinoma: an association with bilharziasis. APMIS 115:1206–1218 Sumitamo M, Shen R, Walburg M et al, 2000, Neutral endopeptidase inhibits prostate cancer cell migration by blocking focal adhesion kinase signalling. J Clin Invest 106:1399–1407 Kletsas D, Caselgrandi E, Barbieri D et al, 1998, Neutral endopeptidase-24.11, NEP, activity in human fibroblasts during development and ageing. Mech Ageing Dev 102:15–23 Kondepudi A, Johnson A, 1993, Cytokines increase neutral endopeptidase activity in lung fibroblasts. Am J Respir Cell Mol Biol 8:43–49 Casey ML, Smith JW, Nagai K et al, 1993, Transforming growth factor-beta 1 inhibits enkephalinase, EC 3.4.24.11, gene expression in human endometrial stromal cells and sex skin fibroblasts in culture. J Clin Endocrinol Metab 77:144– 150 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 153 EP 160 DI 10.1007/s12253-011-9421-8 PG 8 ER PT J AU Chen, ChJ Lin, SE Lin, YM Lin, ShH Chen, DR Chen, ChL AF Chen, Chih-Jung Lin, Sey-En Lin, Yueh-Min Lin, Shu-Hui Chen, Dar-Ren Chen, Chi-Long TI Association of Expression of Kruppel-Like Factor 4 and Kruppel-Like Factor 5 with the Clinical Manifestations of Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Immunohistochemistry; KLF4; KLF5; Kruppel-like factor ID Breast cancer; Immunohistochemistry; KLF4; KLF5; Kruppel-like factor AB Kruppel-like factors (KLFs) are import modulators of cell proliferation, differentiation, and transformation and have recently been considered possible prognostic factors in breast cancer. In this study, we investigated the correlation between KLF4 and KLF5 expression and the clinical manifestations of breast cancer by immunohistochemical analysis. We observed increased KLF4 and KLF5 expression in tumor cells (invasive and in situ carcinomas), consistent KLF4 and KLF5 expression in in situ and invasive carcinomas, significant associations between KLF4 expression and tumor grade (p=0.033), size (p=0.035) and stage (p=0.006), and an association between KLF5 expression and tumor grade (p=0.033). Interestingly, we observed a relationship between increasing age and KLF4 expression (p=0.007), with a tendency towards greater expression in tumor cells in patients over 50 years old. Moreover, KLF5 nuclear localization was restricted to non-tumor breast ducts and lobules; however, loss of nuclear expression of KLF5 in in situ and invasive carcinomas was observed. Although the mechanism of the loss of KLF5 nuclear expression is not clear, this phenomenon may imply a possible tumorsuppressor-like role for KLF5 in breast cancer tumorigenesis. The expression of KLF4 and KLF5 in breast cancer patients in Taiwan is similar to that in Western countries, except for the uncertainty surrounding its prognostic significance. Further clarification of the underlying mechanisms of KLF4 and KLF5 expression and their correlations with breast cancer outcomes is necessary. C1 [Chen, Chih-Jung] Taipei Medical University, College of Medicine, Graduate Institute of Clinical Medicine, 250 Wu-Xing Street, 110 Taipei, Taiwan, Republic of China. [Lin, Sey-En] Taipei Medical University, College of Medicine, Graduate Institute of Clinical Medicine, 250 Wu-Xing Street, 110 Taipei, Taiwan, Republic of China. [Lin, Yueh-Min] Changhua Christian Hospital, Department of PathologyChanghua, Taiwan, Republic of China. [Lin, Shu-Hui] Changhua Christian Hospital, Department of PathologyChanghua, Taiwan, Republic of China. [Chen, Dar-Ren] Changhua Christian Hospital, Department of Surgery, Comprehensive Breast Cancer CenterChanghua, Taiwan, Republic of China. [Chen, Chi-Long] Taipei Medical University, College of Medicine, Graduate Institute of Clinical Medicine, 250 Wu-Xing Street, 110 Taipei, Taiwan, Republic of China. RP Chen, ChL (reprint author), Taipei Medical University, College of Medicine, Graduate Institute of Clinical Medicine, 110 Taipei, Taiwan, Republic of China. EM chencl@tmu.edu.tw CR Cheng SH, Tsou MH, Liu MC et al, 2000, Unique features of breast cancer in Taiwan. Breast Cancer Res Treat 63:213–223 Black AR, Black JD, Azizkhan-Clifford J, 2001, Sp1 and Kruppel-like factor family of transcription factors in cell growth regulation and cancer. J Cell Physiol 188:143–160 Dang DT, Pevsner J, Yang VW, 2000, The biology of the mammalian Kruppel-like family of transcription factors. Int J Biochem Cell Biol 32:1103–1121 Turner J, Crossley M, 1999, Mammalian Kruppel-like transcription factors: more than just a pretty finger. Trends Biochem Sci 24:236–240 Clarke ND, Berg JM, 1998, Zinc fingers in Caenorhabditis elegans: finding families and probing pathways. Science 282:2018–2022 Pandya AY, Talley LI, Frost AR et al, 2004, Nuclear localization of KLF4 is associated with an aggressive phenotype in early-stage breast cancer. Clin Cancer Res 10:2709–2719 Tong D, Czerwenka K, Heinze G et al, 2006, Expression of KLF5 is a prognostic factor for disease-free survival and overall survival in patients with breast cancer. Clin Cancer Res 12:2442–2448 Guo H, Lin Y, Zhang H et al, 2007, Tissue factor pathway inhibitor-2 was repressed by CpG hypermethylation through inhibition of KLF6 binding in highly invasive breast cancer cells. BMC Mol Biol 8:110 Wang X, Zheng M, Liu G et al, 2007, Kruppel-like factor 8 induces epithelial to mesenchymal transition and epithelial cell invasion. Cancer Res 67:7184–7193 Ghaleb AM, Nandan MO, Chanchevalap S et al, 2005, Kruppel-like factors 4 and 5: the yin and yang regulators of cellular proliferation. Cell Res 15:92–96 Rowland BD, Bernards R, Peeper DS, 2005, The KLF4 tumor suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene. Nat Cell Biol 7:1074–1082 Rowland BD, Peeper DS, 2006, KLF4, p21 and contextdependent opposing forces in cancer. Nat Rev Cancer 6:11–23 Miller KA, Eklund EA, Peddinghaus ML et al, 2001, Kruppel-like factor 4 regulates laminin alpha 3A expression inmammary epithelial cells. J Biol Chem 276:42863–42868 Foster KW, Ren S, Louro ID et al, 1999, Oncogene expression cloning by retroviral transduction of adenovirus E1A-immortalized rat kidney RK3E cells: transformation of a host with epithelial features by c-MYC and the zinc finger protein GKLF. Cell Growth Differ 10:423–434 Foster KW, Frost AR, McKie-Bell P et al, 2000, Increase of GKLF messenger RNA and protein expression during progression of breast cancer. Cancer Res 60:6488–6495 Chen C, Bhalala HV, Qiao H et al, 2002, A possible tumor suppressor role of the KLF5 transcription factor in human breast cancer. Oncogene 21:6567–6572 Chen C, Zhou Y, Zhou Z et al, 2004, Regulation of KLF5 involves the Sp1 transcription factor in human epithelial cells. Gene 330:133–142 Chen C, Zhou Y, Dong JT, 2006, KLF5: Kruppel-like factor 5, intestinal). In: Atlas of genetics and cytogenetics in oncology and haematology. http://AtlasGeneticsOncology.org/Genes/ KLF5ID41074ch13q21.html. Cited 15 Oct 2006 Rosen PP, 2009, Invasive duct carcinoma: assessment of prognosis, morphologic prognostic markers, and tumor growth rate. In: Rosen PP, ed, Rosen’s breast pathology. Lippincott Williams & Wilkins, Philadelphia, pp 358–404 Chen C, Sun X, Guo P et al, 2005, Human Kruppel-like factor 5 is a target of the E3 ubiquitin ligase WWP1 for proteolysis in epithelial cells. J Biol Chem 280:41553–41561 Graham JD, Yager ML, Hill HD et al, 2005, Altered progesterone receptor isoform expression remodels progestin responsiveness of breast cancer cells. Mol Endocrinol 19:2713–2735 Beckers J, Herrmann F, Rieger S et al, 2005, Identification and validation of novel ERBB2, HER2, NEU, targets including genes involved in angiogenesis. Int J Cancer 114:590–597 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 161 EP 168 DI 10.1007/s12253-011-9422-7 PG 8 ER PT J AU Rasti, M Arabsolghar, R Khatooni, Z Mostafavi-Pour, Z AF Rasti, Mozhgan Arabsolghar, Rita Khatooni, Zahed Mostafavi-Pour, Zoherh TI p53 Binds to Estrogen Receptor 1 Promoter in Human Breast Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Estrogen receptor 1; Methylated promoter; p53; Epigenetic ID Breast cancer; Estrogen receptor 1; Methylated promoter; p53; Epigenetic AB p53 is a tumor suppressor protein that regulates estrogen receptor 1 (ESR1) expression. To investigate the mechanism of ESR1 gene regulation by p53, chromatin immunoprecipitation was applied to assess the binding of p53, DNMT1, HDAC1 and MeCP2 to both silenced ESR1 promoter in MDA-MB-468 cells and active ESR1 promoter in MCF-7 breast cancer cells. The results of chromatin immunoprecipitation experiments showed that p53 protein binds to both unmethylated CpG island of the ESR1 promoter in the ER-positive MCF-7 and the hypermethylated ESR1 promoter in the ER-negative MDA-MB-468 cells. However, repression complex including DNMT1, HDAC1 and MeCP2 is only associated with silenced ESR1 in ER-negative MDA-MB-468 human breast cancer cells. In addition, ectopically expressed wild type p53 failed to reactivate the ESR1 gene in these cells. These results suggest that specific p53 mutations may contribute to loss of estrogen receptor α expression in breast tumors and also support the hypothesis that mutant p53 is likely to impact DNA methylation. C1 [Rasti, Mozhgan] Shiraz University of Medical Sciences, Department of BiochemistryShiraz, Iran. [Arabsolghar, Rita] Shiraz University of Medical Sciences, Department of BiochemistryShiraz, Iran. [Khatooni, Zahed] Shiraz University of Medical Sciences, Department of BiochemistryShiraz, Iran. [Mostafavi-Pour, Zoherh] Shiraz University of Medical Sciences, Department of BiochemistryShiraz, Iran. RP Rasti, M (reprint author), Shiraz University of Medical Sciences, Department of Biochemistry, Shiraz, Iran. EM rasti31@yahoo.com CR Millau JF, Bastien N, Drouin R, 2009, p53 transcriptional activities: a general overview and some thoughts. Mutat Res 681:118–133 Olsson A, Manzl C, Strasser A et al, 2007, How important are post-translational modifications in p53 for selectivity in targetgene transcription and tumour suppression? Cell Death Differ 14:1561–1575 Harms K, Nozell S, Chen X, 2004, The common and distinct target genes of the p53 family transcription factors. Cell Mol Life Sci 61:822–842 Helton ES, Chen X, 2007, p53 modulation of the DNA damage response. J Cell Biochem 100:883–896 Murphy M, Ahn J, Walker KK et al, 1999, Transcriptional repression by wild-type p53 utilizes histone deacetylases, mediated by interaction with mSin3a. Genes Dev 13:2490–2501 Seto E, Usheva A, Zambetti GP et al, 1992, Wild-type p53 binds to the TATA-binding protein and represses transcription. Proc Natl Acad Sci U S A 89:12028–12032 Amaral JD, Xavier JM, Steer CJ et al, 2010, The role of p53 in apoptosis. Discov Med 9:145–152 Olivier M, Eeles R, Hollstein M et al, 2002, The IARC TP53 database: new online mutation analysis and recommendations to users. Hum Mutat 19:607–614 Angeloni SV, Martin MB, Garcia-Morales P et al, 2004, Regulation of estrogen receptor-alpha expression by the tumor suppressor gene p53 in MCF-7 cells. J Endocrinol 180:497–504 Luczak MW, Jagodzinski PP, 2006, The role of DNA methylation in cancer development. Folia Histochem Cytobiol 44:143–154 Miremadi A, Oestergaard MZ, Pharoah PD, et al., 2007, Cancer genetics of epigenetic genes. HumMol Genet 16 Spec No 1:R28-R49. Parrella P, Poeta ML, Gallo AP et al, 2004, Nonrandom distribution of aberrant promoter methylation of cancer-related genes in sporadic breast tumors. Clin Cancer Res 10:5349–5354 Costello JF, Fruhwald MC, Smiraglia DJ et al, 2000, Aberrant CpG-island methylation has non-random and tumour-type-specific patterns. Nat Genet 24:132–138 Sharma D, Blum J, Yang X et al, 2005, Release of methyl CpG binding proteins and histone deacetylase 1 from the Estrogen receptor alpha, ER, promoter upon reactivation in ER-negative human breast cancer cells. Mol Endocrinol 19:1740–1751 Yan L, Nass SJ, Smith D et al, 2003, Specific inhibition of DNMT1 by antisense oligonucleotides induces re-expression of estrogen receptor-alpha, ER, in ER-negative human breast cancer cell lines. Cancer Biol Ther 2:552–556 Yang X, Ferguson AT, Nass SJ et al, 2000, Transcriptional activation of estrogen receptor alpha in human breast cancer cells by histone deacetylase inhibition. Cancer Res 60:6890–6894 Berns EM, Klijn JG, Smid M et al, 1996, TP53 and MYC gene alterations independently predict poor prognosis in breast cancer patients. Genes Chromosomes Cancer 16:170–179 Giacinti L, Claudio PP, Lopez M et al, 2006, Epigenetic information and estrogen receptor alpha expression in breast cancer. Oncologist 11:1–8 Suzuki M, Sunaga N, Shames DS et al, 2004, RNA interferencemediated knockdown of DNA methyltransferase 1 leads to promoter demethylation and gene re-expression in human lung and breast cancer cells. Cancer Res 64:3137–3143 Lapidus RG, Nass SJ, Butash KA et al, 1998, Mapping of ER gene CpG island methylation-specific polymerase chain reaction. Cancer Res 58:2515–2519 Yan L, Yang X, Davidson NE, 2001, Role of DNA methylation and histone acetylation in steroid receptor expression in breast cancer. J Mammary Gland Biol Neoplasia 6:183–192 Vinyals A, Peinado MA, Gonzalez-Garrigues M et al, 1999, Failure of wild-type p53 gene therapy in human cancer cells expressing a mutant p53 protein. Gene Ther 6:22–33 Yang X, Phillips DL, Ferguson AT et al, 2001, Synergistic activation of functional estrogen receptor, ER)-alpha by DNA methyltransferase and histone deacetylase inhibition in human ER-alpha-negative breast cancer cells. Cancer Res 61:7025–7029 EstellerM(2002, CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future. Oncogene 21:5427–5440 Ho JS, Ma W, Mao DYet al, 2005, p53-Dependent transcriptional repression of c-myc is required for G1 cell cycle arrest. Mol Cell Biol 25:7423–7431 Imbriano C, Gurtner A, Cocchiarella F et al, 2005, Direct p53 transcriptional repression: in vivo analysis of CCAAT-containing G2/M promoters. Mol Cell Biol 25:3737–3751 Scoumanne A, Chen X, 2006, The epithelial cell transforming sequence 2, a guanine nucleotide exchange factor for Rho GTPases, is repressed by p53 via protein methyltransferases and is required for G1-S transition. Cancer Res 66:6271–6279 Sengupta S, Shimamoto A, Koshiji M et al, 2005, Tumor suppressor p53 represses transcription of RECQ4 helicase. Oncogene 24:1738–1748 Horvath MM, Wang X, Resnick MA et al, 2007, Divergent evolution of human p53 binding sites: cell cycle versus apoptosis. PLoS Genet 3:e127 Wei CL, Wu Q, Vega VB et al, 2006, A global map of p53 transcription-factor binding sites in the human genome. Cell 124:207–219 Koutsodontis G, Tentes I, Papakosta P et al, 2001, Sp1 plays a critical role in the transcriptional activation of the human cyclindependent kinase inhibitor p21(WAF1/Cip1, gene by the p53 tumor suppressor protein. J Biol Chem 276:29116–29125 Akaogi K, Nakajima Y, Ito I et al, 2009, KLF4 suppresses estrogen-dependent breast cancer growth by inhibiting the transcriptional activity of ERalpha. Oncogene 28:2894–2902 Shirley SH, Rundhaug JE, Tian J et al, 2009, Transcriptional regulation of estrogen receptor-alpha by p53 in human breast cancer cells. Cancer Res 69:3405–3414 Esteve PO, Chin HG, Pradhan S, 2005, Human maintenance DNA, cytosine-5)-methyltransferase and p53 modulate expression of p53-repressed promoters. Proc Natl Acad Sci U S A 102:1000– 1005 Petitjean A, Mathe E, Kato S et al, 2007, Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat 28:622–629 Li SY, Rong M, Iacopetta B, 2006, Germ-line variants in methylgroup metabolism genes and susceptibility to DNA methylation in human breast cancer. Oncol Rep 15:221–225 Oshiro MM, Watts GS, Wozniak RJ et al, 2003, Mutant p53 and aberrant cytosine methylation cooperate to silence gene expression. Oncogene 22:3624–3634 Dhar G, Banerjee S, Dhar K et al, 2008, Gain of oncogenic function of p53 mutants induces invasive phenotypes in human breast cancer cells by silencing CCN5/WISP-2. Cancer Res 68:4580–4587 Goh AM, Coffill CR, Lane DP, 2011, The role of mutant p53 in human cancer. J Pathol 223:116–126 Bertheau P, Espie M, Turpin E et al, 2008, TP53 status and response to chemotherapy in breast cancer. Pathobiology 75:132– 139 Avila MA, Velasco JA, Cansado J et al, 1994, Quercetin mediates the down-regulation of mutant p53 in the human breast cancer cell line MDA-MB468. Cancer Res 54:2424–2428 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 169 EP 175 DI 10.1007/s12253-011-9423-6 PG 7 ER PT J AU Juhasz, E Beres, J Kanizsai, Sz Nagy, K AF Juhasz, Emese Beres, Judit Kanizsai, Szilvia Nagy, Karoly TI The Consequence of a Founder Effect: CCR5-Δ32, CCR2-64I and SDF1-3’A Polymorphism in Vlach Gypsy Population in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CCR5; CCR2; SDF1; HIV-1; Gypsy; Hungary ID CCR5; CCR2; SDF1; HIV-1; Gypsy; Hungary AB Frequencies of genetic polymorphisms of the three most frequent HIV-1 resistance-conferring alleles playing an important role in HIV-1 pathogenesis were analysed in Vlach Gypsy populations living in Hungary, as the largest minority. Mutations in the encoding genes, such as CCR5-Δ32, CCR2-64I and SDF1-3’A are shown to result in protective effects against HIV-1 infection and disease progression. 560 samples collected from Vlach Gypsy individuals living in 6 North-East Hungarian settlements were genotyped by PCR-RFLP method. Overall allele frequencies of CCR5-Δ32, CCR2-64I and SDF1-3’A were found as 0.122, 0.186 and 0.115 respectively. All the observed genotype frequencies were in accordance with Hardy-Weinberg equilibrium . In regions, however, Vlach Gypsies live in majority and in ethnically homogenous communities, a higher CCR5-Δ32 mutations were found, with allele frequencies of 0.148 and 0.140 respectively, which are remarkably higher than those in general Hungarian people, and ten times higher than in regions of North-Western India from where present day Hungarian Gypsies originated in the Middle Ages. In the background of this higher CCR5-Δ32 allele frequency in the population analysed in our study a genetic founder effect could be assumed. Allele frequency of CCR2-64I was found to be among the highest in Europe. SDF1-3’A allele frequency in Vlach Gypsies was significantly lower than in ethnic Hungarians. 63% of the total 560 individuals tested carried at least one of the mutations studied. These results could partially explain the low incidence of HIV/AIDS among Vlach Gypsies in Hungary. C1 [Juhasz, Emese] Semmelweis University, Department of Medical Microbiology, Nagyvarad ter 4, 1089 Budapest, Hungary. [Beres, Judit] National Center for Healthcare Audit and Inspection, Vaci ut 174, 1138 Budapest, Hungary. [Kanizsai, Szilvia] Semmelweis University, Department of Medical Microbiology, Nagyvarad ter 4, 1089 Budapest, Hungary. [Nagy, Karoly] Semmelweis University, Department of Medical Microbiology, Nagyvarad ter 4, 1089 Budapest, Hungary. RP Juhasz, E (reprint author), Semmelweis University, Department of Medical Microbiology, 1089 Budapest, Hungary. EM juheme@net.sote.hu CR Kalaydjieva L, Lochmuller H, Tounev I et al, 2005, Neuromuscular disorders in the Roma, Gypsy, population. Neuromuscul Disord 15:65–71 Almos PZ, Horvath S, Czibula A et al, 2008, H1 tau haplotyperelated genomic variations at 17q21.3 as an Asian heritage of the European Gypsy population. Heredity 101:416–419 Trkola A, 2004, HIV-host interactions: vital to the virus and key to its inhibition. Curr Opin Microbiol 7:555–559 Reiche EM, Bonametti AM, Voltarelli JC et al, 2007, Genetic polymorphisms in the chemokine and chemokine receptors: impact on clinical course and therapy of the human immunodeficiency virus type 1 infection, HIV-1). Curr Med Chem 14:1325–1334 de Silva E, Stumpf MP, 2004, HIV and the CCR5-Delta32 resistance allele. FEMS Microbiol Lett 241:1–12 Jin Q, Agrawal L, Meyer L et al, 2008, CCR5{delta}32 59537-G/A promoter polymorphism is associated with lowtranslational efficiency and the loss of the CCR5{Delta}32 protective effects. J Virol 82:2418–2426 Kaslow RA, Dorak T, Tang JJ, 2005, Influence of host genetic variation on susceptibility to HIV type 1 infection. J Infect Dis 191:S68–S77 Su B, Sun G, Lu D et al, 2000, Distribution of three HIV-1 resistance-conferring polymorphisms, SDF1-3’A, CCR2-641, and CCR5-delta32, in global populations. Eur J Hum Genet 8:975–979 Lucotte G, 2002, Frequencies of 32 base pair deletion of the, delta 32, allele of the CCR5 HIV-1 coreceptor gene in Caucasians: a comparative analysis. Infect Genet Evol 1: 201–205 Winkler C, An P, O'Brien SJ, 2004, Patterns of ethnic diversity among the genes that influence AIDS. Hum Mol Genet 13:R9–R19 Smith MW, Dean M, Carrington M et al, 1997, Contrasting genetic influence of CCR2 and CCR5 variants on HIV-1 infection and disease progression. Hemophilia Growth and Development Study, HGDS), Multicenter AIDS Cohort Study, MACS), Multicenter Hemophilia Cohort Study, MHCS), San Francisco City Cohort, SFCC), ALIVE study. Science 277:959–965 O'Brien SJ, Nelson GW, 2004, Human genes that limit AIDS. Nat Genet 36:565–574 Gharagozloo M, Doroudchi M, Farjadian S et al, 2005, The frequency of CCR5Delta32 and CCR2-64I in southern Iranian normal population. Immunol Lett 96:277–281 Winkler C, Modi W, Smith MWet al, 1998, Genetic restriction of AIDS pathogenesis by an SDF-1 chemokine gene variant. ALIVE study, Hemophilia Growth and Development Study, HGDS), Multicenter AIDS Cohort Study, MACS), Multicenter Hemophilia Cohort Study, MHCS), San Francisco City Cohort, SFCC). Science 279:389–393 Kemeny B, Nagy K, Horvath A, 2000, CCR5 and SDF1 gene polymorphism in HIV-infected and healthy individuals in Hungary. Hung Venerol Arch 4:89–92 Magierowska M, Theodorou I, Debre P et al, 1999, Combined genotypes of CCR5, CCR2, SDF1, and HLA genes can predict the long-term nonprogressor status in human immunodeficiency virus-1-infected individuals. Blood 93:936–941 Apostolakis S, Baritaki S, Krambovitis E et al, 2005, Distribution of HIV/AIDS protective SDF1, CCR5 and CCR2 gene variants within Cretan population. J Clin Virol 34:310–314 Choi BS, Choi JH, Kim SS et al, 2007, CCR2b-64I allelic polymorphisms in advanced HIV-infected Koreans accelerate disease progression. AIDS Res Hum Retroviruses 23:805–811 Szalai C, Czinner A, Csaszar A et al, 1998, Frequency of the HIV-1 resistance CCR5 deletion allele in Hungarian newborns. Eur J Pediatr 157:782 Husain S, Goila R, Shahi S et al, 1998, First report of a healthy Indian heterozygous for delta 32 mutant of HIV-1 co-receptor-CCR5 gene. Gene 207:141–147 Majumder PP, Dey B, 2001, Absence of the HIV-1 protective delta ccr5 allele in most ethnic populations of India. Eur J Hum Genet 9:794–796 Suresh P, Wanchu A, Sachdeva RK et al, 2006, Gene polymorphisms in CCR5, CCR2, CX3CR1, SDF-1 and RANTES in exposed but uninfected partners of HIV-1 infected individuals in North India. J Clin Immunol 26:476–484 Verma R, Gupta RB, Singh K et al, 2007, Distribution of CCR5delta32, CCR2-64I and SDF1-3’A and plasma levels of SDF-1 inHIV-1 seronegativeNorth Indians. J ClinVirol 38:198–203 Kaur G, Singh P, Rapthap CC et al, 2007, Polymorphism in the CCR5 gene promoter and HIV-1 infection in North Indians. Hum Immunol 68:454–461 Sharda S, Gilmour A, Harris V et al, 2008, Chemokin receptor 5, CCR5, deletion polymorphism in North Indian patients with coronary artery disease. Int J Cardiol 124:254–258 Aranka L, Peter M, Jeno K et al, 2009, Genetically determined neuromuscular disorders of some Roma families living in Hungary. Ideggyogy Sz 62:41–47 Herczegfalvi A, Piko H, Karcagi V, 2008, Screening for hereditary neuromuscular disorders with molecular genetics methods in the Roma population of Hungary. Igeggyogy Sz 61:426–430 Szalai C, Csaszar A, Czinner A et al, 1998, High frequency of the CCR5 deletion allele in gypsies living in Hungary. Immunol Lett 63:57–58 Szalai C, Csaszar A, Czinner A et al, 1999, Chemokine receptor CCR2 and CCR5 polymorphisms in children with insulindependent diabetes mellitus. Pediatr Res 46:82–84 Ramana GV, Vasanthi A, Khaja M et al, 2001, Distribution of HIV-1 resistance-conferring polymorphic alleles SDF-1-3’A, CCR2-64I and CCR5-Delta32 in diverse populations of Andhra Pradesh, South India. J Genet 80:37–40 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 177 EP 182 DI 10.1007/s12253-011-9425-4 PG 6 ER PT J AU Wang, Yx Lv, H Li, Zx Li, C Wu, Xy AF Wang, Yue-xiang Lv, Hui Li, Ze-xia Li, Cui Wu, Xiao-ying TI Effect of shRNA Mediated Down-Regulation of Annexin A2 on Biological Behavior of Human Lung Adencarcinoma Cells A549 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Annexin A2; Lung cancer; RNA interference; Proliferation; Invasion ID Annexin A2; Lung cancer; RNA interference; Proliferation; Invasion AB In the previous study, we found that Annexin A2 was significantly up-regulated in lung cancer and could induce related-antigen in lung cancer patients’ serum. To further study the function of Annexin A2, the short hairpin RNA plasmid targeting Annexin A2 was constructed in vitro and transfected into human lung adencarcinoma A549 cells. Knocking down Annexin A2 expression by shRNA, the mRNA level of Annexin A2 was investigated by semi-quantitative RT-PCR. The expression of Annexin A2 protein was examined by Western Blotting and Immuocytochemistry. MTT assay and Transwell chamber model were used to evaluate proliferation and invasion of A549 cells in vitro. The concentration of matrix metalloproteinase-2 (MMP-2) and cathepsin B (CB) in the supernatant was evaluated by ELISA. At 48 h after transfection, the expression of Annexin A2 mRNA and protein was down-regulated significantly, respectively (p<0.05).The proliferation and invasion capability of A549 cells also decreased significantly (p<0.05). The concentration of MMP-2 and CB was down-regulated obviously, respectively (p<0.05). This study implies that Annexin A2 might play an important role in the progression and invasion of human lung cancer cells, and could promote progression of lung cancer by regulating the expression of MMP-2 and CB. C1 [Wang, Yue-xiang] Central South University, Xiangya School of Medicine, Department of Medical Ultrastructure, Xiangya Road #110, 410078 Changsha, Hunan, China. [Lv, Hui] Central South University, Xiangya School of Medicine, Department of Medical Ultrastructure, Xiangya Road #110, 410078 Changsha, Hunan, China. [Li, Ze-xia] Central South University, Xiangya School of Medicine, Department of Medical Ultrastructure, Xiangya Road #110, 410078 Changsha, Hunan, China. [Li, Cui] Central South University, Xiangya Hospital, Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, 410078 Changsha, Hunan, China. [Wu, Xiao-ying] Central South University, Xiangya School of Medicine, Department of Medical Ultrastructure, Xiangya Road #110, 410078 Changsha, Hunan, China. RP Wu, Xy (reprint author), Central South University, Xiangya School of Medicine, Department of Medical Ultrastructure, 410078 Changsha, China. EM WXY14503@Yahoo.com.cn CR Zeuschner D, Stoorvogel W, Gerke V, 2001, Association of annexin 2 with recycling endosomes requires either calcium- or cholesterol-stabilized membrane domains. Eur J Cell Biol 80, 8):499–507 Falsey RR, Marron MT, Gunaherath GM, Shirahatti N, Mahadevan D, Gunatilaka AA, 2006, Actin microfilament aggregation induced by withaferin A is mediated by annexin II. Nat Chem Biol 2:33–38 Mayran N, Parton RG, Gruenberg J, 2003, Annexin II regulates multivesicular endosome biogenesis in the degradation pathway of animal cells. EMBO J 22:3242–3253 Knop M, Aareskjold E, Bode G, Gekre V, 2004, Rab3D and annexin A2 play a role in regulated secretion of vWF, but not tPA, from endothelial cell. EMBO J 23:2982–2992 Yuan Y-W, Sun A-M, Lui Y, Chen H-L, Banerjee AG, 2007, Rna interference of annexin II gene in PC3 cells by using small interference RNA synthesized with in vitro transcription. Chinese Academy of Medical Sciences 22(1):33–37 Gladwin MT, Yao XL, Cowan M, Huang XL, Schneider R, Grant LR, 2000, Retinoic acid reduces p11 protein levels in bronchial epithelial cells by a posttranslational mechanism. Am J Physiol Lung Cell Mol Physiol 279(6):1103–1109 Emoto K, Sawada H, Yamada Y, Fujimoto H, Takahama Y, Ueno M, 2001, Annexin II overexpression is correlated with poor prognosis in human gastric carcinoma. Anticancer Res 21, 2B):1339–1345 Emoto K, Yamada Y, Sawada H, Fujimoto H, Ueno M, Takayama T, 2001, Annexin II over-expression correlates with stromal tenascin-C over-expression: a prognostic marker in colorectal carcinoma. Cancer 92:1419–1426 Ito Y, Arai K, Nozawa R, Yoshida H, Higashiyama T, Takamura Y, 2007, S100A10 expression in thyroid neoplasmsoriginating from the follicular epithelium: contribution to the aggressive characteristic of anaplastic carcinoma. Anticancer Res 27(4C):2679–2683 Domoto T, Miyama Y, Suzuki H, Teratani T, Arai K, Sugiyama T, 2007, Evaluation of S100A10, annexin II and B-FABP expression as markers for renal cell carcinoma. Cancer Sci 98(1):77–82 Esposito I, Penzel R, Chaib-Harrireche M, Barcena U, Bergmann F, Riedl S, 2006, Tenascin C and annexin 2 expression in the process of pancreatic carcinogenesis. J Pathol 208:673–685 Sharma MR, Koltowski L, Ownbey RT, Tuszynski GP, Sharma MC, 2006, Angiogenesis-associated protein annexin II in breast cancer: selective expression in invasive breast cancer and contribution to tumor invasion and progression. Exp Mol Pathol 81(2):146–156 Reeves SA, Chavez-Kappel C, Davis R, Rosenblum M, Israel MA, 1992, Developmental regulation of annexin II, lipocortin 2, in human brain and expression in high grade glioma. Cancer Res 52:6871–6876 Syed SP, Martin AM, Haupt HM, Arenas-Elliot CP, Brooks JJ, 2007, Angiostatin receptor annexin 2 in vascular tumors including angiosarcoma. Hum Pathol 38:508–513 Yang F, Xiao ZQ, Zhang XZ, Li C, Zhang PF, Li MY, 2007, Identification of tumor antigens in human lung squamous carcinoma by serological proteome analysis. J Proteome Res 6, 2):751–758 Wu XY, Xiao ZQ, Chen ZC, Li C, Li JP, Feng XP et al, 2004, Differential analysis of two-dimension gel electrophoresis profiles from the normal-metaplasia-dysplasia-carcinoma tissue of human bronchial epithelium. Pathol Int 54(10):765–773 Li C, Chen ZC, Xiao ZQ, Wu X, Zhan X, Zhang X, 2003, Comparative proteomics analysis of human lung squmous carcinoma. Biochem Biophys Res Commun 309(1):253–260 Yuan YW, Sun AM, Lui Y, Chen LH, Banerjee AG, 2007, RNA interference of Annexin II gene in PC3 cells by using small interference RNA synthesized with in vitro transcription. Chin Med Sci J 22(1):33–37 Choi S, Kobayashi M, Wang J, Habelhah H, Okada F, Hamada J, 2000, Activated leukocyte cell adhesion molecule, ALCAM, and annexin II are involved in the metastatic progression of tumor cells after chemotherapy with adriamycin. Clin Exp Metastasis 18:45–50 Semov A, Moreno MJ, Onichtchenko A, Abulrob A, Ball M, Ekiel I, 2005, Metastasis-associated protein S100A4 induces angiogenesis through interaction with annexin II and accelerated plasmin formation. J Biol Chem 280:20833–20841 Musholt TJ, Hanack J, Brehm C, von Wasielewski R, Musholt PB, 2005, Searching for non-RET molecular alterations in medullary thyroid carcinoma: expression analysis by mRNA differential display. World J Surg 29(4):472–482 Frohlich M, Motte P, Galvin K, Takahashi H, Wands J, Ozturk M, 1990, Enhanced expression of the protein kinase substrate p36 in human hepatocellular carcinoma. J Mol Cell Biol 10(6):3216– 3223 Bae SM, Lee CH, Cho YL, Nam KH, Kim YW, Kim CK et al, 2005, Two-dimensional gel analysis of protein expression profile in squamous cervical cancer patients. Gynecol Oncol 99(1):26–35 Yoon SO, Park SJ, Yun CH, Chung AS, 2003, Roles of matrix metalloproteinases in tumor metastasis and angiogenesis. J Biochem Mol Biol 36:128–137 Westermarck J, Kahari VM, 1999, Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J 13:781–792 Mai J, Finley RL Jr, Waisman DM, Sloane BF, 2000, Human procathepsin B interacts with the annexin II tetramer on the surface of tumor cells. J Biol Chem 275(17):12806–12812 Tummalapalli P, Spomar D, Gondi CS, Olivero WC, Gujrati M, Dinh DH, 2007, RNAi-mediated abrogation of cathepsin B and MMP-9 gene expression in a malignant meningioma cell line leads to decreased tumor growth, invasion and angiogenesis. Int J Onool 31(5):1039 Nakajima M, Welch DR, Belloni PN, Nicolson GL, 1987, Degradation of basement membrane type IV collagen and lung subendothelial matrix by rat adenocarcinoma cell clones of differing metastatic potentials. Cancer Res 47:4869–4876 Basset P, Bellocq JP, Wolf C, Stoll I, Hutin P, 1990, Limacher JM. A novel metalloproteinases gene specifically expressed in stromal cells of breast carcinomas. Nature 348:699–704 Nelson AR, Fingleton B, Rothenberg ML, Matrisian LM, 2000, Matrix metalloproteinases: biologic activity and clinical implications. J Clin Oncol 18:1135–1149 Robinson CM, Stone AM, Shields JD, Huntley S, Paterson IC, Prime SS, 2003, Functional significance of MMP-2 and MMP-9 expression by human malignant oral keratinocyte cell lines. Arch Oral Biol 48(11):779–786 Podgoraki I, Sloane BF, 2003, Cathposin B and its role in cancer progression. Bioehem Symp 70(22):263–276 Rodby S, Sloane BF, Bloin K, 2003, PeficeHular cathepsin B and malignant progression. Cancer Metastasis Rev 22(23):271–286 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 183 EP 190 DI 10.1007/s12253-011-9427-2 PG 8 ER PT J AU Fauzee, JSN Li, Q Wang, Yl Pan, J AF Fauzee, Jasmine Selimah Nilufer Li, Qiaozhuan Wang, Ya-lan Pan, Juan TI Silencing Poly (ADP-Ribose) Glycohydrolase (PARG) Expression Inhibits Growth of Human Colon Cancer Cells In Vitro via PI3K/Akt/NFκ-B Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PARG; PARP; PI3K/Akt pathway; NFκ-B ID PARG; PARP; PI3K/Akt pathway; NFκ-B AB Poly ADP-ribose polymerase (PARP) which is closely related to Poly ADP-ribose glycohydrolase (PARG) has already been thoroughly investigated in both experimental and clinical cancer trials compared to the latter. Nevertheless, in this experiment the importance of PARG expression was highlighted; whereby it is being silenced via lentivirus vector-mediated short hairpin RNA (shRNA). MTT assay showed that there was an inhibition in human Lovo colon cancer cell growth and flow cytometry demonstrated an increase in the population of cells in G0/G1 phase with a decrease in the S phase in transfected Lovo cells. Furthermore, our results suggested that the effect of silencing PARG leads to the inhibition of PARP expression; related to a decrease in the expression of Nuclear Factor Kappa-B (NFκ-B) with an increase in Akt473 phosphorylation; suggesting that the Phosphoinositol 3-kinase (PI3K)/Akt/NFκ-B pathway is important for cellular growth and proliferation. Hence, this study emphasizes and converges on the relevance of silencing PARG which inhibits growth of human colonic cancer cells via PI3K/Akt/NFκ-B pathway; as colon carcinoma remains to be amongst one of the commonest cancers throughout the world with high morbidity and mortality rates. C1 [Fauzee, Jasmine Selimah Nilufer] Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. [Li, Qiaozhuan] Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. [Wang, Ya-lan] Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. [Pan, Juan] Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. RP Wang, Yl (reprint author), Chongqing Medical University, Molecular Medicine and Cancer Research Center, Department of Pathology, 400016 Chongqing, China. EM wangyalan074@126.com CR Blenn C, Althaus FR, Malanga M, 2006, Poly, ADP-ribose, glycohydrolase silencing protects against H2O2-induced cell death. Biochem J 396:419–429 Koh DW, Lawler AM, Poitras MF et al, 2004, Failure to degrade poly, ADP-ribose, increased sensitivity to cytotoxicity and early embryonic lethality. PNAS 101:17699–17704 Poitras MF, Koh DW, Yu SW et al, 2007, Spatial and functional relationship between poly, ADP-ribose, polymerase-1 and Poly, ADP-ribose, glycohydrolase in the brain. Neuroscience 148:198– 211 Meyer RG, Meyer-Ficca ML, Jacobson EL, Jacobson MK, 2003, Human poly, ADP-ribose, glycohydrolase, PARG, gene and the common promoter sequence it shares with inner mitochondrial membrane translocase 23, TIM23). Gene 314:181–190 Fisher AEO, Hochegger H, Takeda S et al, 2007, Poly, ADPribose, polymerase 1 accelerates single-strand break repair in concert with poly, ADP-ribose, glycohydrolase. Mol Cell Biol 27:5597–5605 Cohausz O, Blenn C, Malanga M et al, 2008, The roles of poly, ADP-ribose)-metabolising enzymes in alkylation-induced cell death. Cell Mol Life Sci 65:644–655 Frizzell KM, Gamble MJ, Berrocal JG, 2009, Global analysis of transcriptional regulation by poly, ADP-ribose, polymerase-1 and poly(ADP-ribose, glycohydrolase in MCF-7 human breast cancer cells. J Biol Chem 284:33926–33938 Gagne JP, Moreel X, Gagne P et al, 2009, Proteomic investigation of phosphorylation sites in poly, ADP-ribose, polymerase-1 and poly, ADP-ribose, glycohydrolase. J Proteome Res 8:1014–1029 Palfi A, Toth A, Kulcsar G et al, 2005, The role of akt and mitogen-activated protein kinase systems in the protective effect of poly, ADP-Ribose, polymerase inhibition in langendorff perfused and in isoproterenol-damaged rat hearts. JPET 315:273–282 Tapodi A, Debreceni B, Hanto K et al, 2005, Pivotal role of Akt activation in mitochondrial protection and cell survival by poly, ADP-ribose, polymerase-1 inhibition in oxidative stress. JBC 280:35767–35775 Veres B, Gallyas F Jr, Varbiro G et al, 2003, Decrease of the inflammatory response and induction of the Akt/protein kinase B pathway by poly-(ADP-ribose, polymerase 1 inhibitor in endotoxin-induced septic shock. Biochem Pharmacol 65:1373– 1382 LoPiccolo J, Granville CA, Gills JJ et al, 2007, Targeting Akt in cancer therapy. Anti-cancer drugs 18:861–874 Bader AG, Khang S, Li Z et al, 2005, Oncogenic PI3K deregulates transcription and translation. Nat Rev Cancer 5:921– 929 Li B, Cheung PY, Wang X et al, 2007, Id-1 activation of PI3K/ Akt/NFkB signaling pathway and its significance in promoting survival of esophageal cancer cells. Carcinogenesis 28:2313– 2320 Asano T, Yao Y, Zhu J et al, 2004, The PI 3-kinase/Akt signaling pathway is activated due to aberrant Pten expression and targets transcription factors NF-κB and c-Myc in pancreatic cancer cells. Oncogene 23:8571–8580 Kane LP, Shapiro VS, Stokoe D et al, 1999, Induction of NFkappaB by the Akt/PKB kinase. Curr Biol 9:601–604 Romashkova JA, Makarov SS, 1999, NF-kappaB is a target of AKT in antiapoptotic PDGF signalling. Nature 401:86–90 Virag L, Szabo C, 2002, The therapeutic potential of poly, ADPribose, polymerase inhibitors. Pharmacol Rev 54:375–429 Li M, Threadgill MD, Wang YL, Cai L, Lin X, 2009, A poly, ADP-ribose, polymerase inhibition down-regulates expression of metastasis-related genes in CT26 colon carcinoma cells. Pathobiology 76:108–116 Lin L, Li J, Wang YL et al, 2009, Relationship of PARG with colorectal carcinoma PARP, VEGF and b-FGF in colorectal carcinoma. Chin J Cancer Res 21:135–141 Tulin A, Naumova NM, Menon AK et al, 2006, Drosophilia poly, ADP-Ribose, glycohydrolase mediates chromatin structure and SIR2-dependent silencing. Genetics 172:363–371 Erdelyi K, Bai P, Kovacs I et al, 2009, Dual role of poly(ADPribose, glycohydrolase in the regulation of cell death in oxidatively stresses A549 cells. FASEB 23:3553–3563 Fauzee NJS, Juan P, Wang YL, 2010, PARP and PARG inhibitorsnew therapeutic targets in cancer treatment. Pathol Oncol Res 4:469–478 Karin M, Cao Y, Greten FR et al, 2002, NFκB in cancer: from innocent bystander to major culprit. Nat Rev Cancer 2:301–310 Veres B, Radnai B, Gallyas F et al, 2004, Regulation of kinase cascades and transcripition factors by a poly, ADP-Ribose, polymerase-1 inhibitor, 4-Hydroxyquinazoline, in lipopolysaccharideinduced inflammation in mice. JPET 310:247–255 Kovacs K, Toth A, Deres P et al, 2006, Critical role of PI3-kinase/ Akt activation in the PARP inhibitor induced heart function recovery during ischemia–reperfusion. Biochem Pharmacol 71:441–452 Bar-Shai M, Carmeli E, Reznick AZ, 2005, The role of NF-κB in protein breakdown in immobilization, aging, and exercise from basic processes to promotion of health. Ann NY Acad Sci 1057:431–447 Cai D, Frantz JD, Tawa NE et al, 2004, IKKβ/NF-κB activation causes severe muscle wasting in mice. Cell 119:285–298 Xian L, Guang-jie D, Wang Y et al, 2010, Expressions of Poly, ADP-ribose, Glycohydrolase, PARG, and membrane Type1 Matrix Metalloproteinase, MT1-MMP, in Colorectal Carcinoma. Chin J Cancer Res 22:186–193 Olashaw NE, Kowalik TF, Huang EH et al, 1992, Induction of NF-KB-Like Activity by Platelet-Derived Growth Factor in Mouse Fibroblasts. Mol Biol Cell 3:1131–1139 Bushdid PB, Chen CL, Brantley DM et al, 2001, NF-κB mediates FGF signal regulation of msx-1 expression. Dev Biol 237:107– 115 Hayden MS, West AP, Ghosh S, 2006, NF-κB and the immune response. Oncogene 119:6758–6780 Bai D, Ueno L, Vogt PK, 2009, Akt-mediated regulation of NFκB and the essentialness of NFκB for the oncogenicity of PI3K and Akt. Int J Cancer 125:2863–2870 Guha M, Mackman N, 2002, The phosphatidylinositol 3-kinase- Akt pathway limits lipopolysaccharide activation of signaling pathways and expression of inflammatory mediators in human monocytic cells. JBC 277:32124–32132 Tamizhselvi R, Sun J, Koh YH et al, 2009, Effect of hydrogen sulfide on the phosphatidylinositol 3-kinase-protein kinase B pathway and on caerulein-induced cytokine production in isolated mouse pancreatic acinar cells. JPET 329:1166–1178 Zhou H, Qian J, Li C et al, 2011, Attenuation of cardiac dysfunction by HSPA12B in endotoxin-induced sepsis in mice through a PI3Kdependent mechanism. Cardiovasc Res 89:109–118 Zhao L, Lee JY, Hwang DH, 2008, The phosphatidylinositol 3- kinase/Akt pathway negatively regulates Nod2-mediated NF-kB pathway. Biochempharm 75:1515–1525 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 191 EP 199 DI 10.1007/s12253-011-9428-1 PG 9 ER PT J AU Cao, X Xia, Y Yang, J Jiang, J Chen, L Ni, R Li, L Gu, Z AF Cao, Xiaolei Xia, Yunfei Yang, Junling Jiang, Jinxia Chen, Li Ni, Runzhou Li, Liren Gu, Zhifeng TI Clinical and Biological Significance of Never in Mitosis Gene A-Related Kinase 6 (NEK6) Expression in Hepatic Cell Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human hepatocellular carcinoma (HCC); Nek6; Ki-67; Prognosis ID Human hepatocellular carcinoma (HCC); Nek6; Ki-67; Prognosis AB Nek6 is a cell cycle regulatory gene, which can control cell proliferation and survival. Recent studies suggested that desregulation of Nek6 expression plays a key role in oncogenesis. This study was aimed to investigate the potential roles of Nek6 in hepatocellular carcinoma (HCC) development. Immunohistochemistry and Western blot analysis was performed for Nek6 in 80 hepatocellular carcinoma samples. The data were correlated with clinicopathological features. The univariate and multivariate survival analyses were performed to determine the prognostic significance of Nek6 in HCC. In addition, Nek6 expression vector was used to detect its role in cell cycle control. Nek6 was overexpressed in hepatocellular carcinoma as compared with the adjacent normal tissue. High expression of Nek6 was associated with histological grade and the level of alpha fetal protein, and Nek6 was positively correlated with proliferation marker Ki-67. Univariate analysis showed that Nek6 expression was associated with poor prognosis. Multivariate analysis indicated that Nek6 and Ki-67 protein expression was an independent prognostic marker for HCC. While in vitro, following release from serum starvation of HuH7 HCC cell, the expression of Nek6 was upregulated. Overexpression Nek6 in Huh7 cell could promote the cell cycle. In conclusion, Nek6 is involved in the pathogenesis of hepatocellular carcinoma. It may be a favorable independent poor prognostic parameter for hepatocellular carcinoma. C1 [Cao, Xiaolei] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, Jiangsu, China. [Xia, Yunfei] Affiliated Hospital of Nantong University, Department of Internal Medicine, 20 Xisi Road, 226001 Nantong, Jiangsu, China. [Yang, Junling] Affiliated Hospital of Nantong University, Department of Internal Medicine, 20 Xisi Road, 226001 Nantong, Jiangsu, China. [Jiang, Jinxia] Affiliated Hospital of Nantong University, Department of Internal Medicine, 20 Xisi Road, 226001 Nantong, Jiangsu, China. [Chen, Li] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, Jiangsu, China. [Ni, Runzhou] Affiliated Hospital of Nantong University, Department of Internal Medicine, 20 Xisi Road, 226001 Nantong, Jiangsu, China. [Li, Liren] Affiliated Hospital of Nantong University, Department of Internal Medicine, 20 Xisi Road, 226001 Nantong, Jiangsu, China. [Gu, Zhifeng] Affiliated Hospital of Nantong University, Department of Internal Medicine, 20 Xisi Road, 226001 Nantong, Jiangsu, China. RP Li, L (reprint author), Affiliated Hospital of Nantong University, Department of Internal Medicine, 226001 Nantong, China. EM larry017@163.com CR Llovet JM, Burroughs A, Bruix J, 2003, Hepatocellular carcinoma. Lancet 362:1907–1917 Di Bisceglie AM, 2004, Issues in screening and surveillance for hepatocellular carcinoma. Gastroenterology 127:S104– S107 Parkin DM, Bray F, Ferlay J et al, 2001, Estimating the world cancer burden: GLOBOCAN 2000. Int J Cancer 94:153–156 Curado MP, Edwards B, Shin HR et al, 2007, Cancer incidence in five continents, IARC, Lyon, France, IARC Scientific Publications, No. 160, Vol. IX Sean FA, Katherine AM, Marsha ER, 2009, Hepatocellular carcinoma incidence, mortality, and survival trends in the United States from 1975 to 2005. JCO 27(9):1485–1491 Bosch FX, Ribes J, Diaz M, Cleries R, 2004, Primary liver cancer: worldwide incidence and trends. Gastroenterology 127(5): S6–S16 Lai PBS, Tianyi C, George GC, 2007, Different levels of p53 induced either apoptosis or cell cycle arrest in a doxycyclineregulated hepatocellular carcinoma cell line in vitro. Apoptosis 12, 2):387–393 Siddhartha K, Meifang W, Brian IC, 2008, 2-Methoxyestradiol inhibits hepatocellular carcinoma cell growth by inhibiting Cdc25 and inducing cell cycle arrest and apoptosis. Cancer Chemother Pharmacol 62(5):831–840 Wang H, Pan K, Zhang H et al, 2008, Increased polycomb-group oncogene Bmi-1 expression correlates with poor prognosis in hepatocellular carcinoma. J Cancer Res Clin Oncol 134(5):535– 541 Laura G, Manuela F, Francesca F et al, 2007, Cyclin G1 is a target of miR-122a, a microRNA frequently down-regulated in human hepatocellular carcinoma. Cancer Res 67(13):6092–6099 O’Connell MJ, Krien MJ, Hunter T, 2003, Never say never. The NIMA-related protein kinases in mitotic control. Trends Cell Biol 13(5):221–228 Belham C, Roig J, Caldwell JA et al, 2003, A mitotic cascade of NIMA family kinases Nercc1/Nek9 activates the Nek6 and Nek7 kinases. J Biol Chem 278(37):34897–34909 Manning G, Whyte DB, Martinez R et al, 2002, The protein kinase complement of the human genome. Science 298, 5600):1912–1934 Min Jean Y, Lihua S, David V et al, 2003, The serine/threonine kinase Nek6 is required for cell cycle progression through mitosis. J Biol Chem 278(52):52454–52460 O’Regan L, Fry AM, 2009, The Nek6 and Nek7 protein kinases are required for robust mitotic spindle formation and cytokinesis. Mol Cell Biol 29(14):3975–3990 Joseph R, Marta N, Aaron G et al, 2008, The NIMA-family kinase Nek6 phosphorylates the kinesin Eg5 at a novel site necessary for mitotic spindle formation. Journal of Cell Science 121(pt23):3912–3921 Rounak N, Lihua S, Peter F et al, 2010, Nek6 mediates human cancer cell transformation and is a potential cancer therapeutic target. Mol Cancer Res 8(5):717–728 Ahmed S, Thomas G, Ghoussaini M et al, 2009, Newly discovered breast cancer susceptibility loci on 3p24 and 17q23. 2. Nat Genet 41(5):585–590 Ke H, John ML, Lee NPY et al, 2009, Predicting prognosis in hepatocellular carcinoma after curative surgery with common clinicopathologic parameters. BMC Cancer 9:389 Thorgeirsson SS, Grisham JW, 2002, Molecular pathogenesis of human hepatocellular carcinoma. Nat Genet 31:339–346 Di Bisceglie AM, 2009, Hepatitis B and hepatocellular carcinoma. Hepatology 49(S5):S56–S60 Guillermo C, Ignacio PC, Marcos M, 2007, Targeting cell cycle kinases for cancer therapy. Curr Med Chem 14(9):969–985 Marcos M, Mariano B, 2007, Cell cycle kinases in cancer. Curr Opin Genet Dev 17:60–65 Chen J, Li L, Zhang Y et al, 2006, Interaction of Pin1 with Nek6 and characterization of their expression correlation in Chinese hepatocellular carcinoma patients. Biochem Biophys Res Commun 341(4):1059–1065 Grigioni WF, D’errico A, Bacci F et al, 1989, Primary liver neoplasms: evaluation of proliferative index using MoAb Ki-67. J Pathol 158(1):23–29 Ng IO, Na J, Lai EC et al, 1995, Ki-67 antigen expression in hepatocellular carcinoma using monoclonal antibody MIB1: a comparison with proliferating cell nuclear antigen. Am J Clin Pathol 104(3):313–318 Scholzen T, Gerde J, 2000, The Ki-67 protein: from the known and the unknown. J Cell Physiol 182(3):311–322 Pang R, Yuen J, Yuen MF et al, 2004, PIN1 overexpression and β- catenin gene mutations are distinct oncogenic events in human hepatocellular carcinoma. Oncogene 23(23):4182–4186 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 201 EP 207 DI 10.1007/s12253-011-9429-0 PG 7 ER PT J AU Gumustekin, M Kargi, A Bulut, G Gozukizil, A Ulukus, C Oztop, I Atabey, N AF Gumustekin, Mukaddes Kargi, Aydanur Bulut, Gulay Gozukizil, Aysim Ulukus, Cagnur Oztop, Ilhan Atabey, Nese TI HGF/c-Met Overexpressions, but not Met Mutation, Correlates with Progression of Non-small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE c-Met; HGF; Invasion; Non-small cell lung cancer; RhoA; MMP-2; MMP-9; TIMP-3 ID c-Met; HGF; Invasion; Non-small cell lung cancer; RhoA; MMP-2; MMP-9; TIMP-3 AB Hepatocyte Growth Factor (HGF) and its receptor c-Met are suggested to play an important role in progression of solid organ tumors by mediating cell motility, invasion and metastasis. Overexpression of HGF and c-Met have been shown in non-small-cell lung cancer (NSCLC). However, their role in tumor progression is not clearly defined. The aim of this study is to determine the role of HGF/c-Met pathway and its association with invasion related markers and clinicopathologic parameters in NSCLC. Immunohistochemical analysis was performed on 63 paraffin-embedded NSCLC tumor sections. The expressions of invasion related markers such as Matrix Metalloproteinases (MMPs) 2 and 9, Tissue Inhibitor Metalloproteinase (TIMP) 1 and 3 and RhoA were also examined. Coexpression of HGF/c-Met was significantly associated with lymph node invasion and TIMP-3 and RhoA overexpressions. There were positive correlation between TIMP-3 overexpression and advanced stage and negative correlation between RhoA overexpression and survival. DNA sequencing for Met mutations in both nonkinase and tyrosine kinase (TK) domain was established. A single nucleotide polymorphism (SNP) in sema domain and two SNPs in TK domain of c-Met were found. There was no statistically significant correlation between the presence of c-Met alterations and clinicopathologic parameters except shorter survival time in cases with two SNPs in TK domain. These results suggest that HGF/c-Met might exert their effects in tumor progression in association with RhoA and probably with TIMP-3. The blockade of the HGF/c-Met pathway with RhoA and/or TIMP-3 inhibitors may be an effective therapeutic target for NSCLC treatment. C1 [Gumustekin, Mukaddes] Dokuz Eylul University, Medical School, Department of PharmacologyInciralti, Izmir, Turkey. [Kargi, Aydanur] Dokuz Eylul University, Medical School, Department of PathologyInciralti, Izmir, Turkey. [Bulut, Gulay] Dokuz Eylul University, Medical School, Department of Medical Biology and GeneticsInciralti, Izmir, Turkey. [Gozukizil, Aysim] Dokuz Eylul University, Medical School, Department of Medical Biology and GeneticsInciralti, Izmir, Turkey. [Ulukus, Cagnur] Dokuz Eylul University, Medical School, Department of PathologyInciralti, Izmir, Turkey. [Oztop, Ilhan] Dokuz Eylul University, Medical School, Department of Medical OncologyInciralti, Izmir, Turkey. [Atabey, Nese] Dokuz Eylul University, Medical School, Department of Medical Biology and GeneticsInciralti, Izmir, Turkey. RP Atabey, N (reprint author), Dokuz Eylul University, Medical School, Department of Medical Biology and Genetics, Inciralti, Turkey. EM nese.atabey@deu.edu.tr CR Jemal A, Siegel R, Ward E et al, 2006, Cancer statistics. CA Cancer J Clin 56:106–130 Ramalingam S, Belani C, 2008, Systemic chemotherapy for advanced non-small cell lung cancer: recent advances and future directions. Oncologist 13:5–13 Okamoto I, 2008, Overview of chemoradiation clinical trials for locally advanced non-small cell lung cancer in Japan. Int J Clin Oncol 13:112–116 Ma PC, Jagadeeswaran R, Jagadeesh S et al, 2005, Functional expression and mutations of c-Met and its therapeutic inhibition with SU11274 and small interfering RNA in Non–small cell lung cancer. Cancer Res 65:1479–1488 Jiang WG, Martin TA, Parr C et al, 2005, Hepatocyte growth factor, its receptor, and their potential value in cancer therapies. Crit Rev Oncol Hematol 53:35–69 Nakamura Y, Niki T, Goto A et al, 2007, c-Met activation in lung adenocarcinoma tissues: an immunohistochemical analysis. Cancer Sci 98:1006–1013 Nakamura Y, Matsubara D, Goto A et al, 2008, Constitutive activation of c-Met is correlated with c-Met overexpression and dependent on cell–matrix adhesion in lung adenocarcinoma cell lines. Cancer Sci 99:14–22 Stabile LP, Lyker JS, Land SR et al, 2006, Transgenic mice overexpressing hepatocyte growth factor in the airways show increased susceptibility to lung cancer. Carcinogenesis 27:1547–1555 Pepper MS, Matsumoto K, Nakamura T et al, 1992, Hepatocyte growth factor increases urokinase-type plasminogen activator, u- PA, and u-PA receptor expression in Madin–Darby canine kidney epithelial cells. J Biol Chem 267:20493–20496 Jeffers M, Rong S, Vande Woude GF, 1996, Enhanced tumorigenicity and invasion-metastasis by hepatocyte growth factor/scatter factor-met signalling in human cells concomitant with induction of the urokinase proteolysis network. Mol Cell Biol 16:1115–1125 Lee KH, Hyun MS, Kim JR, 2003, Growth factor-dependent activation of the MAPK pathway in human pancreatic cancer: MEK/ERK and p38 MAP kinase interaction in uP synthesis. Clin Exp Metastasis 20:499–505 Lorenzato A, Olivero M, Patane S et al, 2002, Novel somatic mutations of the MET oncogene in human carcinoma metastases activating cell motility and invasion. Cancer Res 62:7025–7030 Ma PC, Kijima T, Maulik G et al, 2003, c-MET mutational analysis in small cell lung cancer: novel juxtamembrane domain mutations regulating cytoskeletal functions. Cancer Res 63:6272–6281 Ma PC, Tretiakova MS, MacKinnon AC et al, 2008, Expression and mutational analysis of MET in human solid cancers. Genes Chromosomes Cancer 47:1025–1037 Kitajo H, Shibata T, Nagayasu H et al, 2003, Rho regulates the hepatocyte growth factor/scatter factor-stimulated cell motility of human oral squamous cell carcinoma cells. Oncol Rep 10:1351–1356 Ridley AJ, Comoglio PM, Hall A, 1995, Regulation of scatter factor/hepatocyte growth factor responses by Ras, Rac, and Rho in MDCK cells. Mol Cell Biol 5:1110–1122 Mizuno S, Matsumoto K, Li MY, Nakamura T, 2005, HGF reduces advancing lung fibrosis in mice: a potential role for MMP-dependent myofibroblast apoptosis. FASEB J 19:580–582 Ide T, Kitajima Y, Miyoshi A et al, 2006, Tumor-stromal cell interaction under hypoxia increases the invasiveness of pancreatic cancer cells through the hepatocyte growth factor/c-Met pathway. Int J Cancer 119:2750–2759 Wang SW, Pan SL, Peng CYet al, 2007, CHM-1 inhibits hepatocyte growth factor-induced invasion of SK-Hep-1 human hepatocellular carcinoma cells by suppressing matrix metalloproteinase-9 expression. Cancer Lett 257:87–96 Passlick B, Sienel W, Seen-Hibler R et al, 2000, Overexpression of matrix metalloproteinase 2 predicts unfavorable outcome in early-stage non-small cell lung cancer. Clin Cancer Res 6:3944–3948 Sienel W, Hellers J, Morresi-Hauf A et al, 2003, Prognostic impact of matrix metalloproteinase-9 in operable non-small cell lung cancer. Int J Cancer 103:647–651 Suzuki M, Iizasa T, Fujisawa T et al, 1998-99)Expression of matrix metalloproteinases and tissue inhibitor of matrix metalloproteinases in non-small-cell lung cancer. Invasion Metastasis 18:134–141 Thomas P, Khokha R, Shepherd FA et al, 2000, Differential expression of matrix metalloproteinases and their inhibitors in non-small cell lung cancer. J Pathol 190:150–156 Gouyer V, Conti M, Devos P et al, 2005, Tissue inhibitor of metalloproteinase 1 is an independent predictor of prognosis in patients with nonsmall cell lung carcinoma who undergo resection with curative intent. Cancer 103:1676–1684 Simi L, Andreani M, Davini F et al, 2004, Simultaneous measurement of MMP9 and TIMP1 mRNA in human non small cell lung cancers by multiplexreal time RT-PCR. Lung Cancer 45:171–179 Aljada IS, Ramnath N, Donohue K et al, 2004, Upregulation of tissue inhibitor of metalloproteinase-1 protein is associated with progression of human non-small- cell lung cancer. J Clin Oncol 22:3218–3229 Mino N, Takenaka K, Sonobe M et al, 2007, Expression of tissue inhibitor of metalloproteinase-3, TIMP-3, and its prognostic significance in resected non-small cell lung cancer. J Surg Oncol 95:250–257 Sobin LH, Wittekind C, 2002, Lung. In: Sobin LH, Wittekind C, eds, UICC International Union Against Cancer. TNM classification of malignant tumours. 6th edn. Wiley-Liss, New York Gilcrease MZ, Truong L, Brown RW, 1996, Correlation of very late activation integrin and CD44 expression with extrarenal invasion and metastasis of renal cell carcinomas. Hum Pathol 27:1355–1360 Christensen JG, Burrows J, Salgia R, 2005, c-Met as a target for human cancer and characterization of inhibitors for therapeutic intervention. Cancer Lett 225:1–26 Wang D, Sadee W, 2006, Searching for polymorphisms that affect gene expression and mRNA processing: example ABCB1, MDR1). The AAPS Journal 8:E515–E520 Kimchi-Sarfaty C, Oh JM, Kim IW et al, 2007, A "Silent" polymorphism in the MDR1 gene changes substrate specificity. Science 315:525 Li XR, Ji F, Ouyang J et al, 2006, Overexpression of RhoA is associated with poor prognosis in hepatocellular carcinoma. Eur J Surg Oncol 32:1130–1134 Nawrocki-Raby B, Gilles C, Polette M et al, 2003, Upregulation of MMPs by soluble E-cadherin in human lung tumor cells. Int J Cancer 105:790–795 Castagnino P, Soriano JV, Montesano R, Bottaro DP, 1998, Induction of tissue inhibitor of metalloproteinases-3 is a delayed early cellular response to hepatocyte growth factor. Oncogene 17:481–492 Kim SH, Choi HY, Lee J et al, 2007, Elevated activities of MMP- 2 in the non-tumorous lung tissues of curatively resected stage I NSCLC patients are associated with tumor recurrence and a poor survival. J Surg Oncol 95:337–346 Ahonene MA, Baker AH, Kaharl VM, 1998, Adenovirusmediated gene delivery of tissue inhibitor of metalloproteinases- 3 inhibits invasion and induces apoptosis in melanoma cells. Cancer Res 58:2310–2315 Baker AH, George SJ, Zaltsman AB et al, 1999, Inhibition of invasion and induction of apoptotic cell death of cancer cell lines by overexpression of TIMP-3. Br J Cancer 79:1347–1355 Miyazaki T, Kato H, Nakajima M et al, 2004, An immunohistochemical study of TIMP-3 expression in oesophageal squamous cell carcinoma. B J Cancer 91:1556–1560 Yang T, Hawkes SP, 1992, Role of the 21-kDa protein TIMP-3 in oncogenic transformation of cultured embryo fibroblasts. Proc Natl Acad Sci USA 89:10676–10680 Canovas D, Rennie IG, Nichols CE, Sisley K, 2008, Local environmental influences on uveal melanoma. Cancer 112:1787–1794 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 209 EP 218 DI 10.1007/s12253-011-9430-7 PG 10 ER PT J AU Karabon, L Pawlak-Adamska, E Tomkiewicz, A Jedynak, A Kielbinski, M Woszczyk, D Potoczek, S Jonkisz, A Kuliczkowski, K Frydecka, I AF Karabon, Lidia Pawlak-Adamska, Edyta Tomkiewicz, Anna Jedynak, Anna Kielbinski, Marek Woszczyk, Dariusz Potoczek, Stanislaw Jonkisz, Anna Kuliczkowski, Kazimierz Frydecka, Irena TI Variations in Suppressor Molecule CTLA-4 Gene Are Related to Susceptibility to Multiple Myeloma in a Polish Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CTLA-4; Gene polymorphisms; MM ID CTLA-4; Gene polymorphisms; MM AB Various phenotype and functional T-cell abnormalities are observed in multiple myeloma (MM) patients. The aim of this study was to investigate the association between polymorphisms in the gene encoding cytotoxic Tlymphocyte antigen-4 (CTLA-4), a negative regulator of the T-lymphocyte immune response and susceptibility to multiple myeloma in a Polish population. Two hundred MM patients and 380 healthy subjects were genotyped for the following polymorphisms: CTLA-4c.49A>G, CTLA-4g.319C>T, CTLA-4g.*642AT(8_33), CT60 (CTLA-4g.*6230G>A), Jo31 (CTLA-4g.*10223G>T). Our study is the largest and most comprehensive evaluation to date of the association between genetic polymorphisms in the CTLA-4 molecule and multiple myeloma. It was found that CTLA-4c.49A>G[G], CT60[G], and Jo31[G] alleles were more frequently observed in MM patients than in controls (0.50 vs. 0.44, p=0.03, 0.65 vs. 0.58, p=0.04, and 0.63 vs. 0.57, p=0.03, respectively). Moreover, the haplotype CTLA-4c.49A>G[G], CTLA-4g.319C>T[C], CTLA-4g.*642AT(8_33) [8], CT60[G], Jo31[G] including all susceptibility alleles increases the risk of MM about fourfold (OR: 3.79, 95%CI: 2.08–6.89, p=0.00001). These findings indicate that genetic variations in the CTLA-4 gene play role in susceptibility to multiple myeloma and warrant further investigation through replication studies. C1 [Karabon, Lidia] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland. [Pawlak-Adamska, Edyta] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland. [Tomkiewicz, Anna] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland. [Jedynak, Anna] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland. [Kielbinski, Marek] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, Pasteura 1, 50-367 Wroclaw, Poland. [Woszczyk, Dariusz] Regional Hospital in Opole, Department of Hematology, ul. Kosnego 53Opole, Poland. [Potoczek, Stanislaw] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, Pasteura 1, 50-367 Wroclaw, Poland. [Jonkisz, Anna] Medical University, Department of Forensic Medicine, Curie Sklodowskiej 52, 50-369 Wroclaw, Poland. [Kuliczkowski, Kazimierz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, Pasteura 1, 50-367 Wroclaw, Poland. [Frydecka, Irena] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland. RP Karabon, L (reprint author), Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, 53-114 Wroclaw, Poland. EM lkarabon@iitd.pan.wroc.pl CR Mozaffari F, Hansson L, Kiaii S, Ju X, Rossmann ED, Rabbani H, Mellstedt H, Osterborg A, 2004, Signalling molecules and cytokine production in T cells of multiple myeloma-increased abnormalities with advancing stage. Br J Haematol 124:315–324 Bianchi A, Mariani S, Beggiato E, Borrione P, Peola S, Boccadoro M, Pileri A, Massaia M, 1997, Distribution of Tcell signalling molecules in human myeloma. Br J Haematol 97:815–820 Brown RD, Pope B, Yuen E, Gibson J, Joshua DE, 1998, The expression of T cell related costimulatory molecules in multiple myeloma. Leuk Lymphoma 31:379–384 Frassanito MA, Cusmai A, Dammacco F, 2001, Deregulated cytokine network and defective Th1 immune response in multiple myeloma. Clin Exp Immunol 125:190–197 Murakami H, Ogawara H, Hiroshi H, 2004, Th1/Th2 cells in patients with multiple myeloma. Hematology 9:41–45 Hilbert DM, Shen MY, Rapp UR, Rudikoff S, 1995, Tcells induce terminal differentiation of transformed B cells to mature plasma cell tumors. Proc Natl Acad Sci USA 92:649–653 Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM, Thompson CB, Bluestone JA, 1994, CTLA-4 can function as a negative regulator of T cell activation. Immunity 1:405–413 Valk E, Rudd CE, Schneider H, 2008, CTLA-4 trafficking and surface expression. Trends Immunol 29:272–279 Suwalska K, Pawlak E, Karabon L, Tomkiewicz A, Dobosz T, Urbaniak-Kujda D, Kuliczkowski K, Wolowiec D, Jedynak A, Frydecka I, 2008, Association studies of CTLA-4, CD28, and ICOS gene polymorphisms with B-cell chronic lymphocytic leukemia in the Polish population. Hum Immunol 69:193–201 Erfani N, Razmkhah M, Talei AR, Pezeshki AM, Doroudchi M, Monabati A, Ghaderi A, 2006, Cytotoxic T lymphocyte antigen-4 promoter variants in breast cancer. Cancer Genet Cytogenet 165:114–120 Ghaderi A, Yeganeh F, Kalantari T, Talei AR, Pezeshki AM, Doroudchi M, Dehaghani AS, 2004, Cytotoxic T lymphocyte antigen-4 gene in breast cancer. Breast Cancer Res Treat 86:1–7 Piras G, Monne M, Uras A, Palmas A, Murineddu M, Arru L, Bianchi A, Calvisi A, Curreli L, Gaviano E, Lai P, Murgia A, Latte GC, Noli A, Gabbas A, 2005, Genetic analysis of the 2q33 region containing CD28-CTLA4-ICOS genes: association with non- Hodgkin’s lymphoma. Br J Haematol 129:784–790 Su TH, Chang TY, Lee YJ, Chen CK, Liu HF, Chu CC, Lin M, Wang PT, Huang WC, Chen TC, Yang YC, 2007, CTLA-4 gene and susceptibility to human papillomavirus-16-associated cervical squamous cell carcinoma in Taiwanese women. Carcinogenesis 28:1237–1240 Wong YK, Chang KW, Cheng CY, Liu CJ, 2006, Association of CTLA-4 gene polymorphism with oral squamous cell carcinoma. J Oral Pathol Med 35:51–54 Zheng C, Huang D, Liu L, Bjorkholm M, Holm G, Yi Q, Sundblad A, 2001, Cytotoxic T-lymphocyte antigen-4 microsatellite polymorphism is associated with multiple myeloma. Br J Haematol 112:216–218 Anjos S, Nguyen A, Ounissi-Benkalha H, Tessier MC, Polychronakos C, 2002, A common autoimmunity predisposing signal peptide variant of the cytotoxic T-lymphocyte antigen 4 results in inefficient glycosylation of the susceptibility allele. J Biol Chem 277:46478–46486 Wang XB, Zhao X, Giscombe R, Lefvert AK, 2002, A CTLA-4 gene polymorphism at position −318 in the promoter region affects the expression of protein. Genes Immun 3:233–234 Chistiakov DA, Savost'anov KV, Turakulov RI, Efremov IA, Demurov LM, 2006, Genetic analysis and functional evaluation of the C/T(−318, and A/G(−1661, polymorphisms of the CTLA-4 gene in patients affected with Graves’ disease. Clin Immunol 118:233–242 Ligers A, Teleshova N, Masterman T, Huang WX, Hillert J, 2001, CTLA-4 gene expression is influenced by promoter and exon 1 polymorphisms. Genes Immun 2:145–152 Donner H, Braun J, Seidl C, Rau H, Finke R, Ventz M, Walfish PG, Usadel KH, Badenhoop K, 1997, Codon 17 polymorphism of the cytotoxic T lymphocyte antigen 4 gene in Hashimoto’s thyroiditis and Addison’s disease. J Clin Endocrinol Metab 82:4130–4132 Kouki T, Sawai Y, Gardine CA, Fisfalen ME, Alegre ML, DeGroot LJ, 2000, CTLA-4 gene polymorphism at position 49 in exon 1 reduces the inhibitory function of CTLA-4 and contributes to the pathogenesis of Graves’ disease. J Immunol 165:6606–6611 Wang XB, Kakoulidou M, Giscombe R, Qiu Q, Huang D, Pirskanen R, Lefvert AK, 2002, Abnormal expression of CTLA- 4 by T cells from patients with myasthenia gravis: effect of an ATrich gene sequence. J Neuroimmunol 130:224–232 Karabon L, Kosmaczewska A, Bilinska M, Pawlak E, Ciszak L, Jedynak A, Jonkisz A, Noga L, Pokryszko-Dragan A, Koszewicz M, Frydecka I, 2009, The CTLA-4 gene polymorphisms are associated with CTLA-4 protein expression levels in multiple sclerosis patients and with susceptibility to disease. Immunology 128:e787–e796 Daroszewski J, Pawlak E, Karabon L, Frydecka I, Jonkisz A, Slowik M, Bolanowski M, 2009, Soluble CTLA-4 receptor an immunological marker of Graves’ disease and severity of ophthalmopathy is associated with CTLA-4 Jo31 and CT60 gene polymorphisms. Eur J Endocrinol 161:787–793 Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: a report of the International Myeloma Working Group. Br J Haematol 121:749–757, 2003 Greipp PR, San MJ, Durie BG, Crowley JJ, Barlogie B, Blade J, Boccadoro M, Child JA, vet-Loiseau H, Kyle RA, Lahuerta JJ, Ludwig H, Morgan G, Powles R, Shimizu K, Shustik C, Sonneveld P, Tosi P, Turesson I, Westin J, 2005, International staging system for multiple myeloma. J Clin Oncol 23:3412–3420 Ploski R, Wozniak M, Pawlowski R, Monies DM, Branicki W, Kupiec T, Kloosterman A, Dobosz T, Bosch E, Nowak M, Lessig R, Jobling MA, Roewer L, Kayser M, 2002, Homogeneity and distinctiveness of Polish paternal lineages revealed by Y chromosome microsatellite haplotype analysis. Hum Genet 110:592–600 Shi YY, He L, 2005, SHEsis, a powerful software platform for analyses of linkage disequilibrium, haplotype construction, and genetic association at polymorphism loci. Cell Res 15:97–98 Landgren O, Linet MS, McMaster ML, Gridley G, Hemminki K, Goldin LR, 2006, Familial characteristics of autoimmune and hematologic disorders in 8,406 multiple myeloma patients: a population-based case-control study. Int J Cancer 118:3095–3098 Zheng C, Huang D, Liu L, Wu R, Bergenbrant GS, Osterborg A, Bjorkholm M, Holm G, Yi Q, Sundblad A, 2001, Interleukin-10 gene promoter polymorphisms in multiple myeloma. Int J Cancer 95:184–188 Mazur G, Bogunia-Kubik K, Wrobel T, Karabon L, Polak M, Kuliczkowski K, Lange A, 2005, IL-6 and IL-10 promoter gene polymorphisms do not associate with the susceptibility for multiple myeloma. Immunol Lett 96:241–246 Brown EE, Lan Q, Zheng T, Zhang Y, Wang SS, Hoar-Zahm S, Chanock SJ, Rothman N, Baris D, 2007, Common variants in genes that mediate immunity and risk of multiple myeloma. Int J Cancer 120:2715–2722 Lincz LF, Kerridge I, Scorgie FE, Bailey M, Enno A, Spencer A, 2004, Xenobiotic gene polymorphisms and susceptibility to multiple myeloma. Haematologica 89:628–629 Hayden PJ, Tewari P, Morris DW, Staines A, Crowley D, Nieters A, Becker N, de SS, Foretova L, Maynadie M, Cocco PL, Boffetta P, Brennan P, Chanock SJ, Browne PV, Lawler M, 2007, Variation in DNA repair genes XRCC3, XRCC4, XRCC5 and susceptibility to myeloma. Hum Mol Genet 16:3117–3127 Hosgood HD III, Baris D, Zhang Y, Berndt SI, Menashe I, Morton LM, Lee KM, Yeager M, Zahm SH, Chanock S, Zheng T, Lan Q, 2009, Genetic variation in cell cycle and apoptosis related genes and multiple myeloma risk. Leuk Res 33:1609–1614 Mayans S, Lackovic K, Nyholm C, Lindgren P, Ruikka K, Eliasson M, Cilio CM, Holmberg D, 2007, CT60 genotype does not affect CTLA-4 isoform expression despite association to T1D and AITD in northern Sweden. BMC Med Genet 8:3 Gough SC, Walker LS, Sansom DM, 2005, CTLA4 gene polymorphism and autoimmunity. Immunol Rev 204:102–115 Ihara K, Ahmed S, Nakao F, Kinukawa N, Kuromaru R, Matsuura N, Iwata I, Nagafuchi S, Kohno H, Miyako K, Hara T, 2001, Association studies of CTLA-4, CD28, and ICOS gene polymorphisms with type 1 diabetes in the Japanese population. Immunogenetics 53:447–454 Ueda H, Howson JM, Esposito L, Heward J, Snook H, Chamberlain G, Rainbow DB, Hunter KM, Smith AN, Di GG, Herr MH, Dahlman I, Payne F, Smyth D, Lowe C, Twells RC, Howlett S, Healy B, Nutland S, Rance HE, Everett V, Smink LJ, Lam AC, Cordell HJ, Walker NM, Bordin C, Hulme J, Motzo C, Cucca F, Hess JF, Metzker ML, Rogers J, Gregory S, Allahabadia A, Nithiyananthan R, Tuomilehto-Wolf E, Tuomilehto J, Bingley P, Gillespie KM, Undlien DE, Ronningen KS, Guja C, Ionescu- Tirgoviste C, Savage DA, Maxwell AP, Carson DJ, Patterson CC, Franklyn JA, Clayton DG, Peterson LB, Wicker LS, Todd JA, Gough SC, 2003, Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature 423:506–511 Sun T, Zhou Y, Yang M, Hu Z, Tan W, Han X, Shi Y, Yao J, Guo Y, Yu D, Tian T, Zhou X, Shen H, Lin D, 2008, Functional genetic variations in cytotoxic T-lymphocyte antigen 4 and susceptibility to multiple types of cancer. Cancer Res 68:7025–7034 Goldin LR, Pfeiffer RM, Gridley G, Gail MH, Li X, Mellemkjaer L, Olsen JH, Hemminki K, Linet MS, 2004, Familial aggregation of Hodgkin lymphoma and related tumors. Cancer 100:1902–1908 Goldin LR, Pfeiffer RM, Li X, Hemminki K, 2004, Familial risk of lymphoproliferative tumors in families of patients with chronic lymphocytic leukemia: results from the Swedish Family-Cancer Database. Blood 104:1850–1854 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 219 EP 226 DI 10.1007/s12253-011-9431-6 PG 8 ER PT J AU Boonjaraspinyo, S Boonmars, Th Kaewkes, S Laummaunwai, P Pinlaor, S Loilome, W Yongvanit, P Wu, Z Puapairoj, A Bhudhisawasdi, V AF Boonjaraspinyo, Sirintip Boonmars, Thidarut Kaewkes, Sasithorn Laummaunwai, Porntip Pinlaor, Somchai Loilome, Watchalin Yongvanit, Puangrat Wu, Zhiliang Puapairoj, Anucha Bhudhisawasdi, Vajarabhongsa TI Down-Regulated Expression of HSP70 in Correlation with Clinicopathology of Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cholangiocarcinoma; Clinicopathology; Retinoblastoma 1; Cyclin D1; Heat shock protein 70; Heat shock protein 90; Histone deacetylase 6 ID Cholangiocarcinoma; Clinicopathology; Retinoblastoma 1; Cyclin D1; Heat shock protein 70; Heat shock protein 90; Histone deacetylase 6 AB Cholangiocarcinoma is a crucial health problem in northeast Thailand. Although rare, it is a highly fatal disease and the prognosis of CCA patients is very poor. To determine if expression of specific genes is useful for diagnosis and prognosis for CCA. We examined the expression of HSP70, HSP90, RB1, cyclin D1, and HDAC6 in 50 resections of human CCA tissues by quantitative real-time PCR. The expression of HSP70, RB1, and HDAC6 was "dominant down-regulation", while the expression of cyclin D1 and HSP90 was "dominant upregulation". There were no correlations between RB1, cyclin D1, HSP90, and clinicopathological parameters such as status, histology type, histological grading, stage of CCA, and metastasis. A significant association was found between HDAC6 and CCA staging (p=0.000), CCA gross type and HSP70 (p=0.046) as well as RB1 expression (p=0.046). Patients with down-regulation of HSP70 had significantly poorer prognosis than those in the upregulation group (p=0.002). Expression of HSP70 may be useful as a new prognostic marker for CCA. C1 [Boonjaraspinyo, Sirintip] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Boonmars, Thidarut] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Kaewkes, Sasithorn] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Laummaunwai, Porntip] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Pinlaor, Somchai] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Loilome, Watchalin] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Yongvanit, Puangrat] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Wu, Zhiliang] Gifu University, Graduate School of Medicine, Department of Parasitology, 5011194 Gifu, Japan. [Puapairoj, Anucha] Khon Kaen University, Faculty of Medicine, Department of Pathology, 40002 Khon Kaen, Thailand. [Bhudhisawasdi, Vajarabhongsa] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. RP Boonmars, Th (reprint author), Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. EM bthida@kku.ac.th CR International Agency for Research on Cancer, IARC,, 1994, Infection with liver flukes, Opisthorchis viverrini, Opisthorchis felineus and Clonorchis sinensis). IARC Monogr Eval Carcinog Risks Hum 61:121–175 Sithithaworn P, Haswell-Elkins MR, 2003, Epidemiology of Opisthorchis viverrini. Acta Trop 88:187–194 Sripa B, Pairojkul C, 2008, Cholangiocarcinoma: lessons from Thailand. Curr Opin Gastroenterol 24:349–156 Freeman BC, Yamamoto KR, 2002, Disassembly of transcriptional regulatory complexes by molecular chaperones. Science 296:2232–2235 Beere HM, Wolf BB, Cain K et al, 2000, Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome. Nat Cell Biol 2:469–475 Madhavan J, Ganesh A, Kumaramanickavel G, 2008, Retinoblastoma: from disease to discovery. Ophthalmic Res 40:221–226 Das SK, Hashimoto T, Shimizu K et al, 2005, Fucoxanthin induces cell cycle arrest at G0/G1 phase in human colon carcinoma cells through up-regulation of p21WAF1/Cip1 2. Biochim Biophys Acta 1726:328–335 Lundberg AS, Weinberg RA, 1998, Functional inactivation of the retinoblastoma protein requires sequential modification by at least two distinct cyclin-cdk complexes. Mol Cell Biol 18:753–761 Glozak MA, Seto E, 2007, Histone deacetylases and cancer. Oncogene 26:5420–5432 Hideshima T, Bradner JE, Wong J et al, 2005, Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proc Natl Acad Sci U S A 102:8567–8572 DeOliveira ML, Cunningham SC, Cameron JL et al, 2007, Cholangiocarcinoma: thirty-one-year experience with 564 patients at a single institution. Ann Surg 245:755–762 Kondo S, Takada T, Miyazaki M et al, 2008, Guidelines for the management of biliary tract and ampullary carcinomas: surgical treatment. J Hepatobiliary Pancreat Surg 15:41–54 Uttaravichien T, Bhudhisawasdi V, Pairojkul C et al, 1999, Intrahepatic cholangiocarcinoma in Thailand. J Hepatobiliary Pancreat Surg 62:128–135 Green A, Uttaravichien T, Bhudhisawasdi V et al, 1991, Cholangiocarcinoma in north east Thailand: a hospital-based study. Trop Geogr Med 43:193–198 Boonmars T, Wu Z, Boonjaruspinyo S et al, 2009, Alterations of gene expression of RB pathway in Opisthorchis viverrini infectioninduced cholangiocarcinoma. Parasitol Res 105:1273–1281 Liver Cancer Study Group of Japan, 2003, The general rules for the clinical and pathological study of primary liver cancer, 2th ed. Tokyo: Kanehara 15 Nakanuma Y, Sripa B, Batanasapt V et al, 2000, Intrahepatic cholangiocarcinoma. In: Hamilton SR, Aaltonen LA, eds, World Health Organization classification of tumours: pathology and genetics–– tumours of the digestive system. IARC Press, Lyon, pp 173–180 Edge SB, Byrd DR, Compton CC et al, 2009, AJCC cancer staging manual, 7th edn. Springer-Verlag Press, New York Ciocca DR, Calderwood SK, 2005, Heat shock proteins in cancer: diagnostic, prognostic, predictive, and treatment implications. Cell Stress Chaperones 10:86–103 Chuma M, Sakamoto M, Yamazaki K et al, 2003, Expression profiling in multistage hepatocarcinogenesis: identification of HSP70 as a molecular marker of early hepatocellular carcinoma. Hepatology 37:198–207 Lazaris AC, Chatzigianni EB, Panoussopoulos D et al, 1997, Proliferating cell nuclear antigen and heat shock protein 70 immunolocalization in invasive ductal breast cancer not otherwise specified. Breast Cancer Res Treat 43:43–51 Nanbu K, Konishi I, Komatsu T et al, 1996, Expression of heat shock proteins HSP70 and HSP90 in endometrial carcinomas. Correlation with clinicopathology, sex steroid receptor status, and p53 protein expression. Cancer 77:330–338 Abd el All H, Rey A, Duvillard P, 1998, Expression of heat shock protein 70 and c-myc in cervical carcinoma. Anticancer Res 18:1533–1536 Aghdassi A, Phillips P, Dudeja V et al, 2007, Heat shock protein 70 increases tumorigenicity and inhibits apoptosis in pancreatic adenocarcinoma. Cancer Res 67:616–625 Basso AD, Solit DB, Chiosis G et al, 2002, Akt forms an intracellular complex with heat shock protein 90, Hsp90, and Cdc37 and is destabilized by inhibitors of Hsp90 function. J Biol Chem 277:39858–39866 Vanden Berghe T, Kalai M, van Loo G et al, 2003, Disruption of HSP90 function reverts tumor necrosis factor-induced necrosis to apoptosis. J Biol Chem 278:5622–5629 Chen G, Cao P, Goeddel DV, 2002, TNF-induced recruitment and activation of the IKK complex require Cdc37 and Hsp90. Mol Cell 9:401–410 Jameel A, Skilton RA, Campbell TA et al, 1992, Clinical and biological significance of HSP89 alpha in human breast cancer. Int J Cancer 50:409–415 Yano M, Naito Z, Yokoyama M et al, 1999, Expression of Hsp90 and cyclin D1 in human breast cancer. Cancer Lett 137:45–51 Liu X, Ye L,Wang J et al, 1999, Expression of heat shock protein 90 beta in human gastric cancer tissue and SGD7901/VCR of MDRtype gastric cancer cell line. Chin Med J, Engl, 112:1133–1137 Ogata M, Naito Z, Tanaka S et al, 2000, Overexpression and localization of heat shock proteins mRNA in pancreatic carcinoma. J Nippon Med Sch 67:177–185 Cardillo MR, Sale P, Di Silverio F, 2000, Heat shock protein-90, IL-6 and IL-10 in bladder cancer. Anticancer Res 20:4579–4583 Bosco EE, Wang Y, Xu H et al, 2007, The retinoblastoma tumor suppressor modifies the therapeutic response of breast cancer. J Clin Invest 117:218–228 Contu SS, Contu PC, Damin DC et al, 2007, pRB expression in esophageal mucosa of individuals at high risk for squamous cell carcinoma of the esophagus. World J Gastroenterol 13:1728–1731 Chim CS, Wong KY, Loong F et al, 2007, Frequent epigenetic inactivation of Rb1 in addition to p15 and p16 in mantle cell and follicular lymphoma. Hum Pathol 38:1849–1857 Chatterjee SJ, Datar R, Youssefzadeh D et al, 2004, Combined effects of p53, p21, and pRb expression in the progression of bladder transitional cell carcinoma. J Clin Oncol 22:1007–1103 Sgambato A, Migaldi M, Faraglia B et al, 2002, Cyclin D1 expression in papillary superficial bladder cancer: its association with other cell cycle-associated proteins, cell proliferation and clinical outcome. Int J Cancer 97:671–678 Deeb KK, Michalowska AM, Yoon CY et al, 2007, Identification of an integrated SV40 T/t-antigen cancer signature in aggressive human breast, prostate, and lung carcinomas with poor prognosis. Cancer Res 67:8065–8080 Kang YK, Kim WH, Jang JJ, 2002, Expression of G1-S modulators, p53, p16, p27, cyclin D1, Rb, and Smad4/Dpc4 in intrahepatic cholangiocarcinoma. Hum Pathol 33:877–888 Sumrejkanchanakij P, Tamamori-Adachi M, Matsunaga Y et al, 2003, Role of cyclin D1 cytoplasmic sequestration in the survival of postmitotic neurons. Oncogene 22:8723–8730 Kella VK, Constantine R, Parikh NS et al, 2009, Mantle cell lymphoma of the gastrointestinal tract presenting with multiple intussusceptions – case report and review of literature. World J Surg Oncol 7:60 Gillett C, Smith P, Gregory W et al, 1996, Cyclin D1 and prognosis in human breast cancer. Int J Cancer 69:92–99 Azechi H, Nishida N, Fukuda Yet al, 2001, Disruption of the p16/ cyclin D1/retinoblastoma protein pathway in the majority of human hepatocellular carcinomas. Oncology 60:346–354 Mitra AP, Datar RH, Cote RJ, 2006, Molecular pathways in invasive bladder cancer: new insights into mechanisms, progression, and target identification. J Clin Oncol 24:5552–5564 Garcea G, Neal CP, Pattenden CJ et al, 2005, Molecular prognostic markers in pancreatic cancer: a systematic review. Eur J Cancer 41:2213–2236 Zhang Z, Yamashita H, Toyama T et al, 2004, HDAC6 expression is correlated with better survival in breast cancer. Clin Cancer Res 10:6962–6968 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 227 EP 237 DI 10.1007/s12253-011-9432-5 PG 11 ER PT J AU Abdou, GA Asaad, YN Elkased, A Said, H Dawoud, M AF Abdou, Gaber Asmaa Asaad, Youssef Nancy Elkased, Ahmed Said, Hala Dawoud, Marwa TI Adult Pancreatic Neuroblastoma, an Unusual Site and Fatal Outcome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Neuroblastoma; Pancreas; Fatal outcome ID Neuroblastoma; Pancreas; Fatal outcome AB In this report, we describe a classic case of stroma rich neuroblastoma, nodular type in a 22 year old female presented with a pancreatic mass. This rare and unusual presentation elicits several differential diagnostic categories including solid pseudopapillary tumor, pancreatic endocrine tumor, pancreatoblastoma and PNET. In this report, we tried to differentiate between them depending on the histopathological features and using panel of epithelial and neuroendocrine markers. Although of the rarity of pancreatic neuroblastoma as a primary site of origin, however it should be considered in the differential diagnosis of pancreatic masses in children and young adult. Neuropil and ganglionic differentiation are helpful features to recognize neuroblastoma and differentiate them from other small blue cell tumors. The fatal outcome of adult neuroblastoma confirming the independence of age as a prognostic factor in this neoplasm regardless of stage and histology. C1 [Abdou, Gaber Asmaa] Menoufiya University, Faculty of Medicine, Department of PathologyShebein Elkom, Egypt. [Asaad, Youssef Nancy] Menoufiya University, Faculty of Medicine, Department of PathologyShebein Elkom, Egypt. [Elkased, Ahmed] Menofiya University, Faculty of Medicine, Department of SurgeryShebein Elkom, Egypt. [Said, Hala] Menoufiya University, Faculty of Medicine, Department of PathologyShebein Elkom, Egypt. [Dawoud, Marwa] Menoufiya University, Faculty of Medicine, Department of PathologyShebein Elkom, Egypt. RP Abdou, GA (reprint author), Menoufiya University, Faculty of Medicine, Department of Pathology, Shebein Elkom, Egypt. EM Asmaa_elsaidy@yahoo.com CR Howman-Giles R, Shaw PJ, Uren RF, Chung DKV, 2007, Neuroblastoma and other neuroendocrine tumors. Semin Nucl Med 37:286–302 David R, Eftekhari F, Lamki N, Shirkhoda A, Fan S, Kumar P, Singleton EB,Madewell JE, 1989, Themany faces of neuroblastoma. Radiographics 5:859–882 Menegaux F, Olshan AF, Reitnauer PJ, Blatt J, Cohn SL, 2005, Positive association between congenital anomalies and risk of neuroblastoma. Pediatr Blood Canc 45:649–655 Tsuji A, Takayanagi M, Kasugai T, Sugimoto H, Shibahara H, Kawasaki H, 2010, A case of adult neuroblastoma with fatal outcome. Nippon Shokakibyo Gakkai Zasshi 107:1651–1660 Vouriot MA, Marchal AL, Olive D, Benz Lemoine E, Schmitt M, Hoeffel JC, 1985, Neuroblastoma of the tail of the pancreas. Apropos of a case. J Radiol 66:547–549 Hoeffel JC, GalloyMA, Schmitt C, SchmittM(1991, Neuroblastoma of the tail of the pancreas in childhood. Br J Radiol 64:167–168 Kumar HR, Sandoval JA, Lovell MA, Fenton LZ, Bealer JF, 2010, Primary pancreatic neuroblastoma: an unusual tumor in infancy. J Pediatr Surg 45:642–646 Lieber MR, Lack EE, Roberts JRJ, Merino MJ, Patterson K, Restrepo C, Solomon D, Chandra R, Triche TJ, 1987, Solid and papillary epithelial neoplasm of the pancreas. An ultrastructural and immunocytochemical study of six cases. Am J Surg Pathol 11:85–93 Ehehalt F, Saeger HD, Schmidt CM, Grutzmann R, 2009, Neuroendocrine tumors of the pancreas. Oncologist 14:456– 467 Klimstra DS, Wenig BM, Adair CF, Heffess CS, 1995, Pancreatoblastoma. A clinicopathologic study and review of the literature. Am J Surg Pathol 19:1371–1389 Luttges J, Pierre E, Zamboni G, Weh G, Lietz H, Kussmann J, Kloppel G, 1997, Malignant non-epithelial tumors of the pancreas. Pathologe 18:233–237 Bulchmann G, Schuster T, Haas RJ, Joppich I, 2000, Primitive neuroectodermal tumor of the pancreas. An extremely rare tumor. Case report and review of the literature. Klin Padiatr 212:185–188 Movahedi-Lankarani S, Hruban RH, Westra WH, Klimstra DS, 2002, Primitive neuroectodermal tumors of the pancreas: a report of seven cases of a rare neoplasm. Am J Surg Pathol 26:1040– 1047 Perek S, Perek A, Sarman K, Tuzun H, Buyukunal E, 2003, Primitive neuroectodermal tumor of the pancreas. A case report of an extremely rare tumor. Pancreatology 3:352–356 Welsch T, Mechtersheimer G, Aulmann S, Mueller SA, Buechler MW, Schmidt J, Kienle P, 2006, Huge primitive neuroectodermal tumor of the pancreas: report of a case and review of the literature. World J Gastroenterol 12:6070–6073 Christein JD, Kim AW, Jakate S, Deziel DJ, 2002, Central pancreatectomy for a pancreatic ganglioneuroma in a patient with previous neuroblastoma. Pancreatology 2:557–560 Eun YK, So-Young Y, Ji HK, Ki WS, 2008, Pancreatic metastasis in a child suffering with treated stage 4 neuroblastoma. Korean J Radiol 9:84–86 Fishman DS, Manfredi MA, LaBelle JL, Levine JE, Shamberger RC, Rufo PA, 2007, Pancreatitis as the initial manifestation of stage IV neuroblastoma. J Pediatr Gastroenterol Nutr 44:146– 148 Nekrep N, Wang J, Miyatsuka T, German MS, 2008, Signals from the neural crest regulate beta-cell mass in the pancreas. Development 135:2151–2160 Plank JL, Mundell NA, Frist AY, LeGrone AW, Kim T, Musser MA, Walter TJ, Labosky PA, 2011, Influence and timing of arrival of murine neural crest on pancreatic beta cell development and maturation. Dev Biol 349:321–330 Pearse AG, Polak JM, 1971, Neural crest origin of the endocrine polypeptide, APUD, cells of the gastrointestinal tract and pancreas. Gut 12:783–788 Andrew A, Kramer B, Rawdon BB, 1998, The origin of gut and pancreatic neuroendocrine, APUD, cells–the last word? J Pathol 186:117–118 Panovska-Stavridis I, Ivanovski M, Hadzi-Pecova L, Ljatifi A, Trajkov D, Spiroski M, Cevreska L, 2010, A case report of aggressive adult neuroblastoma mimicking acute leukemia with fulminant course and fatal outcome. Prilozi 31:349–359 Chatten J, Shimada H, Sather HN, Wong KY, Siegel SE, Hammond GD, 1988, Prognostic value of histopathology in advanced neuroblastoma: a report from the Children’s Cancer Study Group. Hum Pathol 19:1187–1198 Slavc I, Ellenbogen R, Jung WH, Vawter GF, Kretschmar C, Grier H, Korf BR, 1990, Myc gene amplification and expression in primary human neuroblastoma. Canc Res 50:1459–1463 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 239 EP 243 DI 10.1007/s12253-011-9434-3 PG 5 ER PT J AU Vlachostergios, JP Karasavvidou, F Patrikidou, A Voutsadakis, AI Kakkas, G Moutzouris, G Zintzaras, E Daliani, DD Melekos, DM Papandreou, NCh AF Vlachostergios, J Panagiotis Karasavvidou, Foteini Patrikidou, Anna Voutsadakis, A Ioannis Kakkas, Grigorios Moutzouris, George Zintzaras, Elias Daliani, D Danai Melekos, D Michael Papandreou, N Christos TI p53 and Cyclooxygenase-2 Expression are Directly Associated with Cyclin D1 Expression in Radical Prostatectomy Specimens of Patients with Hormone-Naive Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p53; Cyclooxygenase-2; Cyclin D1; Radical prostatectomy; Prostate cancer; Biochemical failure ID p53; Cyclooxygenase-2; Cyclin D1; Radical prostatectomy; Prostate cancer; Biochemical failure AB Prostate cancer (PCa) is a potentially curable disease when diagnosed in early stages and subsequently treated with radical prostatectomy (RP). However, a significant proportion of patients tend to relapse early, with the emergence of biochemical failure (BF) as an established precursor of progression to metastatic disease. Several candidate molecular markers have been studied in an effort to enhance the accuracy of existing predictive tools regarding the risk of BF after RP. We studied the immunohistochemical expression of p53, cyclooxygenase-2 (COX-2) and cyclin D1 in a cohort of 70 patients that underwent RP for early stage, hormone naive PCa, with the aim of prospectively identifying any possible interrelations as well as correlations with known prognostic parameters such as Gleason score, pathological stage and time to prostate-specific antigen (PSA) relapse. We observed a significant (p=0.003) prognostic role of p53, with high protein expression correlating with shorter time to BF (TTBF) in univariate analysis. Both p53 and COX-2 expression were directly associated with cyclin D1 expression (p=0.055 and p=0.050 respectively). High p53 expression was also found to be an independent prognostic factor (p=0.023). Based on previous data and results provided by this study, p53 expression exerts an independent negative prognostic role in localized prostate cancer and could therefore be evaluated as a useful new molecular marker to be added in the set of known prognostic indicators of the disease. With respect to COX-2 and cyclin D1, further studies are required to elucidate their role in early prediction of PCa relapse after RP. C1 [Vlachostergios, J Panagiotis] University of Thessaly School of Medicine, University Hospital of Larissa, Department of Medical Oncology, 41110 Biopolis, Larissa, Greece. [Karasavvidou, Foteini] University of Thessaly School of Medicine, University Hospital of Larissa, Department of Pathology, 41110 Biopolis, Larissa, Greece. [Patrikidou, Anna] University of Thessaly School of Medicine, University Hospital of Larissa, Department of Medical Oncology, 41110 Biopolis, Larissa, Greece. [Voutsadakis, A Ioannis] University of Thessaly School of Medicine, University Hospital of Larissa, Department of Medical Oncology, 41110 Biopolis, Larissa, Greece. [Kakkas, Grigorios] University of Thessaly School of Medicine, University Hospital of Larissa, Department of Urology, 41110 Biopolis, Larissa, Greece. [Moutzouris, George] University of Thessaly School of Medicine, University Hospital of Larissa, Department of Urology, 41110 Biopolis, Larissa, Greece. [Zintzaras, Elias] University of Thessaly School of Medicine, Department of Biomathematics, 41110 Biopolis, Larissa, Greece. [Daliani, D Danai] University of Thessaly School of Medicine, University Hospital of Larissa, Department of Medical Oncology, 41110 Biopolis, Larissa, Greece. [Melekos, D Michael] University of Thessaly School of Medicine, University Hospital of Larissa, Department of Urology, 41110 Biopolis, Larissa, Greece. [Papandreou, N Christos] University of Thessaly School of Medicine, University Hospital of Larissa, Department of Medical Oncology, 41110 Biopolis, Larissa, Greece. RP Vlachostergios, JP (reprint author), University of Thessaly School of Medicine, University Hospital of Larissa, Department of Medical Oncology, 41110 Biopolis, Greece. EM pvlacho@med.uth.gr CR Bostwick DG, Grignon DJ, Hammond ME et al, 1999, Prognostic factors in prostate cancer. College of American pathologists consensus statement 1999. Arch Pathol Lab Med 124:995–1000 Brooks JD, Bova GS, Ewing CM et al, 1996, An uncertain role for p53 gene alterations in human prostate cancers. Cancer Res 56:3814–3822 Taylor D, KochWM, ZahurakM et al, 1999, Immunohistochemical detection of p53 protein accumulation in head and neck cancer: correlation with p53 gene alterations. Hum Pathol 30:1221–1225 Zha S, Yegnasubramanian V, NelsonWG et al, 2004, Cyclooxygenases in cancer: progress and perspective. Cancer Lett 215:1–20 Pruthi RS, Derksen E, Gaston K, 2003, Cyclooxygenase-2 as a potential target in the prevention and treatment of genitourinary tumors: a review. J Urol 169:2352–2359 Gupta S, Srivastava M, Ahmad N et al, 2000, Over-expression of cyclooxygenase-2 in human prostate adenocarcinoma. Prostate 42:73–78 Yoshimura R, Sano H, Masuda C et al, 2000, Expression of cyclooxygenase-2 in prostate carcinoma. Cancer 89:589–596 Lee LM, Pan CC, Cheng CJ et al, 2001, Expression of cyclooxygenase-2 in prostate adenocarcinoma and benign prostatic hyperplasia. Anticancer Res 21:1291–1294 Uotila P, Valve E, Martikainen P et al, 2001, Increased expression of cyclooxygenase-2 and nitric oxide synthase-2 in human prostate cancer. Urol Res 29:23–28 Zang T, Sun F, LiY(2001, Expression of COX-2 in prostatic cancer and benign prostatic hyperplasia. Zhonghua Wai Ke Za Zhi 39:702–703 Edwards J, Mukherjee R, Munro AF et al, 2004, HER2 and COX2 expression in human prostate cancer. Eur J Cancer 40:50–55 Denkert C, Thoma A, Niesporek S et al, 2007, Overexpression of cyclooxygenase-2 in human prostate carcinoma and prostatic intraepithelial neoplasia-association with increased expression of Polo-like kinase-1. Prostate 67:361–369 Shappell SB, Manning S, Boeglin WE et al, 2001, Alterations in lipoxygenase and cyclooxygenase-2 catalytic activity and mRNA expression in prostate carcinoma. Neoplasia 3:287–303 Bartkova J, Lukas J, Strauss M et al, 1995, Cyclin D1 oncoprotein aberrantly accumulates in malignancies of diverse histogenesis. Oncogene 10:775–778 Chen Y, Martinez LA, LaCava M et al, 1998, Increased cell growth and tumorigenicity in human prostate LNCaP cells by overexpression to cyclin D1. Oncogene 16:1913–1920 Gumbiner LM, Gumerlock PH, Mack PC et al, 1999, Overexpression of cyclin D1 is rare in human prostate carcinoma. Prostate 38:40–45 Shiraishi T, Watanabe M, Muneyuki T et al, 1998, A clinicopathological study of p53, p21, WAF1/CIP1, and cyclin D1 expression in human prostate cancers. Urol Int 61:90–94 Osman I, Yee H, Taneja SS et al, 2004, Neutral endopeptidase protein expression and prognosis in localized prostate cancer. Clin Cancer Res 10:4096–4100 Brehm A, Miska EA, McCance DJ et al, 1998, Retinoblastoma protein recruits histone deacetylase to repress transcription. Nature 391:597–601 Guardavaccaro D, Corrente G, Covone F et al, 2000, Arrest of G, 1)-S progression by the p53-inducible gene PC3 is Rb dependent and relies on the inhibition of cyclin D1 transcription. Mol Cell Biol 20:1797–1815 Rocha S, Martin AM, Meek DW et al, 2003, p53 represses cyclin D1 transcription through down regulation of Bcl-3 and inducing increased association of the p52 NF-kappaB subunit with histone deacetylase 1. Mol Cell Biol 23:4713–4727 Agus DB, Cordon-Cardo C, Fox W et al, 1999, Prostate cancer cell cycle regulators: response to androgen withdrawal and development of androgen independence. J Natl Cancer Inst 91:1869–1876 Narayanan BA, Narayanan NK, Davis L et al, 2006, RNA interference-mediated cyclooxygenase-2 inhibition prevents prostate cancer cell growth and induces differentiation: modulation of neuronal protein synaptophysin, cyclin D1, and androgen receptor. Mol Cancer Ther 5:1117–1125 Subbaramaiah K, Altorki N, Chung WJ et al, 1999, Inhibition of cyclooxygenase-2 gene expression by p53. J Biol Chem 274:10911–10915 Han JA, Kim JI, Ongusaha PP et al, 2002, P53-mediated induction of Cox-2 counteracts p53- or genotoxic stress-induced apoptosis. EMBO J 21:5635–5644 Choi EM, Heo JI, Oh JY et al, 2005, COX-2 regulates p53 activity and inhibits DNA damage-induced apoptosis. Biochem Biophys Res Commun 328:1107–1112 Bauer JJ, Sesterhenn IA, Mostofi KF et al, 1995, p53 nuclear protein expression is an independent prognostic marker in clinically localized prostate cancer patients undergoing radical prostatectomy. Clin Cancer Res 1:1295–1300 Shurbaji MS, Kalbfleisch JH, Thurmond TS, 1995, Immunohistochemical detection of p53 protein as a prognostic indicator in prostate cancer. Hum Pathol 26:106–109 Bauer JJ, Sesterhenn IA, Mostofi FK et al, 1996, Elevated levels of apoptosis regulator proteins p53 and bcl-2 are independent prognostic biomarkers in surgically treated clinically localized prostate cancer. J Urol 156:1511–1516 Moul JW, Bettencourt MC, Sesterhenn IA et al, 1996, Protein expression of p53, bcl-2, and KI-67, MIB-1, as prognostic biomarkers in patients with surgically treated, clinically localized prostate cancer. Surgery 120:159–166 Theodorescu D, Broder SR, Boyd JC et al, 1997, P53, bcl-2 and retinoblastoma proteins as long-term prognostic markers in localized carcinoma of the prostate. J Urol 158:131–137 Brewster SF, Oxley JD, Trivella M et al, 1999, Preoperative p53, bcl-2, CD44 and E-cadherin immunohistochemistry as predictors of biochemical relapse after radical prostatectomy. J Urol 161:1238–1243 Osman I, Drobnjak M, Fazzari M et al, 1999, Inactivation of the p53 pathway in prostate cancer: impact on tumor progression. Clin Cancer Res 5:2082–2088 Leibovich BC, Cheng L, Weaver AL et al, 2000, Outcome prediction with p53 immunostaining after radical prostatectomy in patients with locally advanced prostate cancer. J Urol 163:1756–1760 Deliveliotis C, Skolarikos A, Karayannis A et al, 2003, The prognostic value of p53 and DNA ploidy following radical prostatectomy. World J Urol 21:171–176 Schlomm T, Iwers L, Kirstein P et al, 2008, Clinical significance of p53 alterations in surgically treated prostate cancers. Mod Pathol 21:1371–1378 Oxley JD, Winkler MH, Parry K et al, 2002, P53 and bcl-2 immunohistochemistry in preoperative biopsies as predictors of biochemical recurrence after radical prostatectomy. BJU Int 89:27–32 Revelos K, Petraki C, Gregorakis A et al, 2005, Immunohistochemical expression of Bcl2 is an independent predictor of time-tobiochemical failure in patients with clinically localized prostate cancer following radical prostatectomy. Anticancer Res 25:3123–3133 Yang G, Stapleton AM, Wheeler TM et al, 1996, Clustered p53 immunostaining: a novel pattern associated with prostate cancer progression. Clin Cancer Res 2:399–401 Stapleton AM, Zbell P, Kattan MWet al, 1998, Assessment of the biologic markers p53, Ki-67, and apoptotic index as predictive indicators of prostate carcinoma recurrence after surgery. Cancer 82:168–175 Quinn DI, Henshall SM, Head DR et al, 2000, Prognostic significance of p53 nuclear accumulation in localized prostate cancer treated with radical prostatectomy. Cancer Res 60:1585– 1594 Inoue T, Segawa T, Shiraishi T et al, 2005, Androgen receptor, Ki67, and p53 expression in radical prostatectomy specimens predict treatment failure in Japanese population. Urology 66:332– 337 Wu TT, Hsu YS, Wang JS et al, 2003, The role of p53, bcl-2 and E-cadherin expression in predicting biochemical relapse for organ confined prostate cancer in Taiwan. J Urol 170:78–81 Goto T, Nguyen BP, Nakano M et al, 2008, Utility of Bcl-2, P53, Ki-67, and caveolin-1 immunostaining in the prediction of biochemical failure after radical prostatectomy in a Japanese population. Urology 72:167–171 Nariculam J, Freeman A, Bott S et al, 2009, Utility of tissue microarrays for profiling prognostic biomarkers in clinically localized prostate cancer: the expression of BCL-2, E-cadherin, Ki-67 and p53 as predictors of biochemical failure after radical prostatectomy with nested control for clinical and pathological risk factors. Asian J Androl 11:109–118 Krupski T, Petroni GR, Frierson HF Jr et al, 2000, Microvessel density, p53, retinoblastoma, and chromogranin A immunohistochemistry as predictors of disease-specific survival following radical prostatectomy for carcinoma of the prostate. Urology 55:743–749 Leibovich BC, Cheng L,Weaver AL et al, 2000, Outcome prediction with p53 immunostaining after radical prostatectomy in patients with locally advanced prostate cancer. J Urol 163:1756–1760 Visakorpi T, Kallioniemi OP, Heikkinen A et al, 1992, Small subgroup of aggressive, highly proliferative prostatic carcinomas defined by p53 accumulation. J Natl Cancer Inst 84:883–887 Deliveliotis C, Skolarikos A, Karayannis A et al, 2003, The prognostic value of p53 and DNA ploidy following radical prostatectomy. World J Urol 21:171–176 Kirschenbaum A, Klausner AP, Lee R et al, 2000, Expression of cyclooxygenase-1 and cyclooxygenase-2 in the human prostate. Urology 56:671–676 Zha S, Gage WR, Sauvageot J et al, 2001, Cyclooxygenase-2 is up-regulated in proliferative inflammatory atrophy of the prostate, but not in prostate carcinoma. Cancer Res 61:8617–8623 Wang W, Bergh A, Damber JE, 2005, Cyclooxygenase-2 expression correlates with local chronic inflammation and tumor neovascularization in human prostate cancer. Clin Cancer Res 11:3250–3256 Dassesse T, de Leval X, de Leval L et al, 2006, Activation of the thromboxane A2 pathway in human prostate cancer correlates with tumor Gleason score and pathologic stage. Eur Urol 50:1021–1031 Di Lorenzo G, De Placido S, Autorino R et al, 2005, Expression of biomarkers modulating prostate cancer progression: implications in the treatment of the disease. Prostate Cancer Prostatic Dis 8:54–59 Rubio J, Ramos D, Lopez-Guerrero JA et al, 2005, Immunohistochemical expression of Ki-67 antigen, cox-2 and Bax/Bcl-2 in prostate cancer; prognostic value in biopsies and radical prostatectomy specimens. Eur Urol 48:745–751 Rao DS, Gui D, Koski ME et al, 2006, An inverse relation between COX-2 and E-cadherin expression correlates with aggressive histologic features in prostate cancer. Appl Immunohistochem Mol Morphol 14:375–383 Cohen BL, Gomez P, Omori Y et al, 2006, Cyclooxygenase-2, COX-2, expression is an independent predictor of prostate cancer recurrence. Int J Cancer 119:1082–1087 Bin W, He W, Feng Z, et al, 2009, Prognostic relevance of cyclooxygenase-2, COX-2, expression in Chinese patients with prostate cancer. Acta Histochem, in press)., DOI 10.1016/j. acthis.2009.09.004 Comstock CE, Revelo MP, Buncher CR et al, 2007, Impact of differential cyclin D1 expression and localisation in prostate cancer. Br J Cancer 96:970–979 Kallakury BV, Sheehan CE, Ambros RA et al, 1997, The prognostic significance of p34cdc2 and cyclin D1 protein expression in prostate adenocarcinoma. Cancer 80:753–763 Drobnjak M, Osman I, Scher HI et al, 2000, Overexpression of cyclin D1 is associated with metastatic prostate cancer to bone. Clin Cancer Res 6:1891–1895 Aaltomaa S, Karja V, Lipponen P et al, 2006, Expression of Ki-67, cyclin D1 and apoptosis markers correlated with survival in prostate cancer patients treated by radical prostatectomy. Anticancer Res 26:4873–4878 Gannon PO, Koumakpayi IH, Le Page C et al, 2008, Ebp1 expression in benign and malignant prostate. Cancer Cell Int 8:18–28 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 245 EP 252 DI 10.1007/s12253-011-9435-2 PG 8 ER PT J AU Kandemir, ON Barut, F Bektas, S Ozdamar, OS AF Kandemir, Onak Nilufer Barut, Figen Bektas, Sibel Ozdamar, Oguz Sukru TI Can Lymphatic Vascular Density Be Used in Determining Metastatic Spreading Potential of Tumor in Invasive Ductal Carcinomas? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast carcinoma; D2-40; Lymhangiogenesis; Lymphatic vessel invasion ID Breast carcinoma; D2-40; Lymhangiogenesis; Lymphatic vessel invasion AB Regional lymph node status is the primary parameter determining treatment strategies and prognoses in breast cancer. Lymphatic vessels in primary tumor tissue play a significant role in lymphatic metastasis. The aim of this study was to investigate the correlation of intra- and peritumoral lymphatic microvessel densities (LVD) with prognostic parameters in breast cancer, including lymphatic invasion (LI). Lymphangiogenesis was investigated using D2-40 monoclonal antibody in 69 invasive ductal carcinoma cases who underwent mastectomy and axillary lymph node dissection. Positively stained microvessels were counted at 400× in dense lymphatic vascular foci (hotspots). Tumor LI was established when at least one neoplastic cell cluster was clearly visible inside a D2-40-positive lymph vessel. Relationships were sought between clinicopathological parameters and mean LVD and LI in primary tumor tissue. Peritumoral LVD was markedly higher than intratumoral LVD (p<0.001). No significant relationship was found between intratumoral LVD and clinicopathological parameters (p>0.05). However, significant relationships were detected between peritumoral LVD and LVI [H&E] (p=0.04), number of lymphatic invasion [n/mm2, D2-40] (p=0.001), tumor size (p=0.01), lymph node status (p=0.03), and tumor stage (p=0.04). The immunohistochemical determination of LI and LVD can contribute to the prediction of a tumor’s biological behavior in invasive ductal carcinomas. Peritumoral LVD in primary tumor tissue is closely related to parameters influencing the prognosis of a tumor. C1 [Kandemir, Onak Nilufer] Zonguldak Karaelmas University, Faculty of Medicine, Department of PathologyZonguldak, Turkey. [Barut, Figen] Zonguldak Karaelmas University, Faculty of Medicine, Department of PathologyZonguldak, Turkey. [Bektas, Sibel] Zonguldak Karaelmas University, Faculty of Medicine, Department of PathologyZonguldak, Turkey. [Ozdamar, Oguz Sukru] Zonguldak Karaelmas University, Faculty of Medicine, Department of PathologyZonguldak, Turkey. RP Kandemir, ON (reprint author), Zonguldak Karaelmas University, Faculty of Medicine, Department of Pathology, Zonguldak, Turkey. EM niluferkandemir@yahoo.com CR Ellis LM, Fidler IJ, 1995, Angiogenesis and breast cancer metastasis. Lancet 12:388–390 Kato T, Kameoka S, Kimura T et al, 2003, The combination of angiogenesis and blood vessel invasion as a prognostic indicator in primary breast cancer. Br J Cancer 88:1900–1908 Casadio C, Luini A, Veronesi P et al, 2007, Prognostic role of the extent of peritumoral vascular invasion in operable breast cancer. Ann Oncol 18:1632–1640 Ogiya A, Kahn HJ, Marks A, 2002, A new monoclonal antibody, D2-40, for detection of lymphatic invasion in primary tumors. Lab Invest 82:1255–1257 Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19:403–410 Greene FL, Page DL, Fleming ID et al, 2002, Breast. In: Greene FL, Page DL, Fleming ID, Fritz AG, Balch CM, Haller DG, Morrow M, eds, AJCC cancer staging manual, 6th edn. Springer, New York, pp 223–240 Harvey JM, Clark GM, Osborne K et al, 1999, Estrogen receptor status by immunohistochemistry is superior to the ligandbinding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol 17:1474–1481 Yaziji H, Taylor CR, 2007, Begin at the beginning, with the tissue! The key message underlying the ASCO/CAP Task-force Guideline Recommendations for HER2 testing. Appl Immunohistochem Mol Morphol 15:239–241 Weidner N, 1995, Current pathologic methods for measuring intratumoral microvessel density within breast carcinoma and other solid tumors. Breast Cancer Res Treat 36:169–180 Saad RS, Lindner JL, Liu Y et al, 2009, Lymphatic vessel density as prognostic marker in esophageal adenocarcinoma. Am J Clin Pathol 131:92–98 Vermeulen PB, Gasparini G, Fox SB et al, 1996, Quantification of angiogenesis in solid human tumors: an international consensus on the morphology and criteria of evaluation. Eur J Cancer 32:2474–2484 Saad RS, Kordunsky L, Denning KL et al, 2006, Lymphatic microvessel density as prognostic marker in colorectal carcinoma. Mod Pathol 19:1317–1323 Kawamura L, Carvalho FM, Alves BG et al, 2010, Association between intratumoral lymphatic microvessel density, LMVD, and clinicopathologic features in endometrial cancer: a retrospective cohort study. World J Surg Oncol 14(8):89 De Mascarel I, MacGrogan G, Debled M et al, 2009, D2-40 in breast cancer: should we detect more vascular emboli? Mod Pathol 22:216–222 Agarwal B, Saxena R, Moriyama A et al, 2005, Lymphangiogenesis does not occur in breast cancer. Am J Surg Pathol 29:1449–1455 Colleoni M, Rotmensz N, Maisonneuve P et al, 2007, Prognostic role of the extent of peritumoral vascular invasion in operable breast cancer. Ann Oncol 18(10):1632–1640 Beasley NJ, Prevo R, Banerji S et al, 2002, Intratumoral lymphangiogenesis and lymph node metastasis in head and neck cancer. Cancer Res 62:1315–1320 Yamauchi C, Hasebe T, Iwasaki M et al, 2007, Accurate assessment of lymph vessel tumor emboli in invasive ductal carcinoma of the breast according to tumor areas, and their prognostic significance. Hum Pathol 38:247–259 Nathanson SD, 2003, Insights into the mechanisms of lymph node metastasis. Cancer 98:413–423 Achen MG, Stacker SA, 2008, Molecular control of lymphatic metastasis. Ann N Y Acad Sci 1131:225–234 Shayan R, Achen MG, Stacker SA, 2006, Lymphatic vessels in cancer metastasis: bridging the gaps. Carcinogenesis 27:1729– 1738 Van der Auwera I, Van den Eynden GG, Colpaert CG et al, 2005, Tumor lymphangiogenesis in inflammatory breast carcinoma: a histomorphometric study. Clin Cancer Res 11:7637–7642 Donoghue JF, Lederman FL, Susil BJ et al, 2007, Lymphangiogenesis of normal endometrium and endometrial adenocarcinoma. Hum Reprod 22:1705–1713 Grossklaus DJ, Coffey CS, Shappell SB et al, 2002, Percent of cancer in the biopsy set predicts pathological findings after prostatectomy. J Urol 167:2032–2035 Franchi A, Gallo O, Massi D et al, 2004, Tumor lymphangiogenesis in head and neck squamous cell carcinoma: a morphometric study with clinical correlations. Cancer 101:973–978 Massi D, Puig S, Franchi A et al, 2006, Tumour lymphangiogenesis is a possible predictor of sentinel lymph node status in cutaneous melanoma: a case-control study. J Clin Pathol 59:166– 173 Van der Schaft DW, Pauwels P, Hulsmans S et al, 2007, Absence of lymphangiogenesis in ductal breast cancer at the primary tumor site. Cancer Lett 28:128–136 Schloppmann SF, Birner P, Studer P et al, 2001, Lymphatic microvessel densitiy and lymphovascular invasion assessed by anti-podoplanin immunostaining in human breast cancer. Anticancer Res 21:2351–2355 Van der Auwera I, Cao Y, Tille JC et al, 2006, First international consensus on the methodology of lymphangiogenesis quantification in solid human tumours. Br J Cancer 18:1611–1625 Britto AV, Schenka AA, Moraes-Schenka NG et al, 2009, Immunostaining with D2-40 improves evaluation of lymphovascular invasion, but may not predict sentinel lymph node status in early breast cancer. BMC Cancer 8(9):109 Marinho VF, Metze K, Sanches FS et al, 2008, Lymph vascular invasion in invasive mammary carcinomas identified by the endothelial lymphatic marker D2-40 is associated with other indicators of poor prognosis. BMC Cancer 29(8):64 Ito M, Moriya T, Ishida T et al, 2007, Significance of pathological evaluation for lymphatic vessel invasion in invasive breast cancer. Breast Cancer 14:381–387 El-Gohary YM, Metwally G, Saad RS et al, 2008, Prognostic significance of intratumoral and peritumoral lymphatic density and blood vessel density in invasive breast carcinomas. Am J Clin Pathol 129:578–586 Ejlertsen B, Jensen MB, Rank F et al, 2009, Population-based study of peritumoral lymphovascular invasion and outcome among patients with operable breast cancer. J Natl Cancer Inst 101:729–735 Arnaout-Alkarain A, Kahn HJ, Narod SA et al, 2007, Significance of lymph vessel invasion identified by the endothelial lymphatic marker D2-40 in node negative breast cancer. Mod Pathol 20:183–191 Beasley N, Prevo R, Banerji S et al, 2002, Intratumoral lymphangiogenesis and lymph node metastasis in head and neck cancer. Cancer Res 62:1315–1320 Dadras SS, Bertoncini J, Brown L et al, 2003, Tumor lymphangiogenesis: a novel prognostic indicator for cutaneous melanoma metastasis and survival. Am J Pathol 162:1951–1960 Van den Eynden GG, Van der Auwera I, Van Laere SJ et al, 2006, Induction of lymphangiogenesis in and around axillary lymph node metastases of patients with breast cancer. Br J Cancer 20:1362–1366 Van den Eynden GG, Van der Auwera I, Van Laere SJ et al, 2007, Comparison of molecular determinants of angiogenesis and lymphangiogenesis in lymph node metastases and in primary tumours of patients with breast cancer. J Pathol 213:56–64 Mylona E, Nomikos A, Alexandrou P et al, 2007, Lymphatic and blood vessel morphometry in invasive breast carcinomas: relation with proliferation and VEGF-C and -D proteins expression. Histol Histopathol 22:825–835 Mohammed RA, Ellis IO, Elsheikh S et al, 2009, Lymphatic and angiogenic characteristics in breast cancer: morphometric analysis and prognostic implications. Breast Cancer Res Treat 113:261–273 Witte MH, Jones K, Wilting J et al, 2006, Structure function relationships in the lymphatic system and implications for cancer biology. Cancer Metastasis Rev 6:159–184 Williams CS, Leek RD, Robson AM et al, 2003, Absence of lymphangiogenesis and intratumoural lymph vessels in human metastatic breast cancer. J Pathol 200:195–206 Colleoni M, Rotmensz N, Maisonneuve P et al, 2007, Prognostic role of the extent of peritumoral vascular invasion in operable breast cancer. Ann Oncol 18:1632–1640 Choi WW, Lewis MM, Lawson D et al, 2005, Angiogenic and lymphangiogenic microvessel density in breast carcinoma: correlation with clinicopathologic parameters and VEGF-family gene expression. Mod Pathol 18:143–152 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 253 EP 262 DI 10.1007/s12253-011-9436-1 PG 10 ER PT J AU Wang, Z Yuan, X Jiao, N Zhu, H Zhang, Y Tong, J AF Wang, Zhu Yuan, Xin Jiao, Nanlin Zhu, Hui Zhang, Youwei Tong, Jiandong TI CDH13 and FLBN3 Gene Methylation are Associated with Poor Prognosis in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Methylation; Prognosis; Microdissection ID Colorectal cancer; Methylation; Prognosis; Microdissection AB The aim of this study was to identify potential epigenetic prognostic biomarkers for colorectal cancer (CRC) in the Chinese population. The methylation status of five tumor suppressor genes (CDH13, DLEC1, FBLN3, hMHL1 and RUNX3) was determined using manual microdissection followed by methylation-specific PCR in 85 paired CRC specimens and adjacent normal tissue. The results showed that methylation frequencies in cancerous tissues were 31.8% for CDH13, 37.6% for DLEC1, 38.8% for FBLN3, 22.4% for hMHL1 and 27.1% for RUNX3, all of which were significantly higher than in corresponding normal tissue. Furthermore, CDH13 methylation was associated with poor differentiation (P=0.019) and tended to be predominant in advanced stages (P=0.084); FBLN3 methylation was associated with advanced stages (P=0.027) and lymph node metastasis (P=0.029). Accordingly, the methylation status of CDH13 (P=0.022), FBLN3 (P=0.008), CDH13 and/or FBLN3 (P=0.001) predicted adverse overall survival in CRC, while hMHL1 methylation showed a protective role in survival (P=0.046). Cox proportional hazard models further indicated that CDH13 and/or FBLN3 methylation, but not that of hMHL1, was an independent prognostic factor for CRC. In conclusion, we found CDH13 and FBLN3 gene methylation are potential biomarkers for poor prognosis in CRC. C1 [Wang, Zhu] The second Clinical School of Yangzhou University, Yangzhou No.1 People’s Hospital, Department of Oncology, Number 368, Mid Hanjiang Road, 225009 Yangzhou, China. [Yuan, Xin] The second Clinical School of Yangzhou University, Yangzhou No.1 People’s Hospital, Department of Oncology, Number 368, Mid Hanjiang Road, 225009 Yangzhou, China. [Jiao, Nanlin] Wannan Medical College, Yijishan Hospital, Department of Pathology, 241001 Wuhu, China. [Zhu, Hui] The second Clinical School of Yangzhou University, Yangzhou No.1 People’s Hospital, Department of Oncology, Number 368, Mid Hanjiang Road, 225009 Yangzhou, China. [Zhang, Youwei] The second Clinical School of Yangzhou University, Yangzhou No.1 People’s Hospital, Department of Oncology, Number 368, Mid Hanjiang Road, 225009 Yangzhou, China. [Tong, Jiandong] The second Clinical School of Yangzhou University, Yangzhou No.1 People’s Hospital, Department of Oncology, Number 368, Mid Hanjiang Road, 225009 Yangzhou, China. RP Tong, J (reprint author), The second Clinical School of Yangzhou University, Yangzhou No.1 People’s Hospital, Department of Oncology, 225009 Yangzhou, China. EM tongjd1@yahoo.cn CR Saif MW, Chu E, 2010, Biology of colorectal cancer. Cancer J 16:196–201 Asghar U, Hawkes E, Cunningham D, 2010, Predictive and prognostic biomarkers for targeted therapy in metastatic colorectal cancer. Clin Colorectal Cancer 9:274–281 Lopez J, Percharde M, Coley HM et al, 2009, The context and potential of epigenetics in oncology. Br J Cancer 100:571–577 Lesche R, Eckhardt F, 2007, DNA methylation markers: a versatile diagnostic tool for routine clinical use. Curr Opin Mol Ther 9:222–230 Zhang Y, Song H, Miao Y et al, 2010, Frequent transcriptional inactivation of Kallikrein 10 gene by CpG island hypermethylation in non-small cell lung cancer. Cancer Sci 101:934–940 Ogino S, Nosho K, Kirkner GJ et al, 2009, CpG island methylator phenotype, microsatellite instability, BRAF mutation and clinical outcome in colon cancer. Gut 58:90–96 Kim MS, Lee J, Sidransky D, 2010, DNA methylation markers in colorectal cancer. Cancer Metastasis Rev 29:181–206 Miladi-Abdennadher I, Abdelmaksoud-Damak R, Ayadi L et al, 2010, Hypermethylation of RARβ2 correlates with high COX-2 expression and poor prognosis in patients with colorectal carcinoma. Tumour Biol 31:503–511 Sirivatanauksorn Y, Sirivatanauksorn V, Bhattacharya S et al, 1999, Evolution of genetic abnormalities in hepatocellular carcinomas demonstrated by DNA fingerprinting. J Pathol 189:344–350 Seng TJ, Currey N, Cooper WA et al, 2008, DLEC1 and hMHL1 promoter methylation are associated with poor prognosis in nonsmall cell lung carcinoma. Br J Cancer 99:375–382 Yue W, Dacic S, Sun Q et al, 2007, Frequent inactivation of RAMP2, EFEMP1 and Dutt1 in lung cancer by promoter hypermethylation. Clin Cancer Res 13:4336–4344 Zhang Y, Wang R, Song H et al, 2011, Methylation of multiple genes as a candidate biomarker in non-small cell lung cancer. Cancer Lett 303:21–28 Dietrich D, Lesche R, Tetzner R et al, 2009, Analysis of DNA methylation of multiple genes in microdissected cells from formalin-fixed and paraffin-embedded tissues. J Histochem Cytochem 57:477–489 Yang S, Nan Y, Tian Y et al, 2008, Study of distinct protein profiles for early diagnosis of NSCLC using LCM and SELDITOF- MS. Med Oncol 25:380–386 Xu XL, Yu J, Zhang HY et al, 2004, Methylation profile of the promoter CpG islands of 31 genes that may contribute to colorectal carcinogenesis. World J Gastroenterol 10:3441–3454 Barault L, Charon-Barra C, Jooste Vet al, 2008, Hypermethylator phenotype in sporadic colon cancer: study on a population-based series of 582 cases. Cancer Res 68:8541–8546 Aoyagi H, Iida S, Uetake H et al, 2011, Effect of classification based on combination of mutation and methylation in colorectal cancer prognosis. Oncol Rep 25:789–794 Hibi K, Nakayama H, Kodera Y et al, 2004, CDH13 promoter region is specifically methylated in poorly differentiated colorectal cancer. Br J Cancer 90:1030–1033 Ying J, Poon FF, Yu J et al, 2009, DLEC1 is a functional 3p22.3 tumour suppressor silenced by promoter CpG methylation in colon and gastric cancers. Br J Cancer 100:663–669 Nishio M, Sakakura C, Nagata T et al, 2010, RUNX3 promoter methylation in colorectal cancer: its relationship with microsatellite instability and its suitability as a novel serum tumor marker. Anticancer Res 30:2673–2682 Fujita H, Kato J, Horii J et al, 2007, Decreased expression of hhMHL1 correlates with reduced 5-fluorouracil-mediated apoptosis in colon cancer cells. Oncol Rep 18:1129–1137 de Vega S, Iwamoto T, Yamada Y, 2009, Fibulins: multiple roles in matrix structures and tissue functions. Cell Mol Life Sci 66:1890–1902 Albig AR, Neil JR, Schiemann WP, 2006, Fibulins 3 and 5 antagonize tumor angiogenesis in vivo. Cancer Res 66:2621–2629 Sadr-Nabavi A, Ramser J, Volkmann J et al, 2009, Decreased expression of angiogenesis antagonist EFEMP1 in sporadic breast cancer is caused by aberrant promoter methylation and points to an impact of EFEMP1 as molecular biomarker. Int J Cancer 124:1727–1735 Wang R, Zhang YW, Chen LB, 2010, Aberrant promoter methylation of FBLN-3 gene and clinicopathological significance in non-small cell lung carcinoma. Lung Cancer 69:239–244 Nomoto S, Kanda M, Okamura Y et al, 2010, Epidermal growth factor-containing fibulin-like extracellular matrix protein 1, EFEMP1, a novel tumor-suppressor gene detected in hepatocellular carcinoma using double combination array analysis. Ann Surg Oncol 17:923–932 Brim H, Mokarram P, Naghibalhossaini F et al, 2008, Impact of BRAF, hMHL1 on the incidence of microsatellite instability high colorectal cancer in populations based study. Mol Cancer 7:68 Noda H, Kato Y, Yoshikawa H et al, 2005, Microsatellite instability caused by hhMHL1 promoter methylation increases with tumor progression in right-sided sporadic colorectal cancer. Oncology 69:354–362 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Poynter JN, Siegmund KD, Weisenberger DJ, Colon Cancer Family Registry Investigators et al, 2008, Molecular characterization of MSI-H colorectal cancer by MLHI promoter methylation, immunohistochemistry, and mismatch repair germline mutation screening. Cancer Epidemiol Biomarkers Prev 17:3208–3215 Arai T, Kasahara I, Sawabe M et al, 2010, Role of methylation of the hhMHL1 gene promoter in the development of gastric and colorectal carcinoma in the elderly. Geriatr Gerontol Int 10:S207– S212 Ide T, Kitajima Y, Ohtaka K et al, 2008, Expression of the hhMHL1 gene is a possible predictor for the clinical response to 5-fluorouracil after a surgical resection in colorectal cancer. Oncol Rep 19:1571–1576 Toyota M, Issa JP, 1999, CpG island methylator phenotypes in aging and cancer. Semin Cancer Biol 9:349–357 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 263 EP 270 DI 10.1007/s12253-011-9437-0 PG 8 ER PT J AU Huang, Z Hua, D Hu, Y Cheng, Z Zhou, X Xie, Q Wang, Q Wang, F Du, X Zeng, Y AF Huang, Zhaohui Hua, Dong Hu, Yu Cheng, Zhihong Zhou, Xike Xie, Qigen Wang, Qiongyao Wang, Feng Du, Xiang Zeng, Yanjun TI Quantitation of Plasma Circulating DNA Using Quantitative PCR for the Detection of Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Plasma; Circulating DNA; Quantitative PCR ID Hepatocellular carcinoma; Plasma; Circulating DNA; Quantitative PCR AB Circulating DNA is a potential biomarker for tumor diagnosis and prognosis. This study was aimed to quantify the circulating DNA in plasma from patients with hepatocellular carcinoma (HCC) using quantitative PCR and evaluate its potential clinical value. Blood samples were collected from 72 patients with HCC, 37 with liver cirrhosis or chronic hepatitis and 41 healthy volunteers. Plasma DNA was extracted and quantified by a real-time quantitative PCR method. The diagnostic and prognostic value of plasma DNA analysis for HCC was evaluated. DNA levels in the HCC plasma (median: 173 ng/mL) were significantly higher than those in the healthy controls (9 ng/mL) or control benign patients (46 ng/mL) (P<0.001). The area under the receiver-operation characteristic (ROC) curve (AUC) assessing plasma DNA was 0.949 for healthy controls and 0.874 for control patients. Plasma DNA detection could discriminate HCC from normal controls with 90.2% sensitivity and 90.3% specificity at the cut-off value of 18.2 ng/mL. Combined ROC analyses using plasma DNA and serum AFP revealed an elevated AUC of 0.974 with 95.1% sensitivity and 94.4% specificity in discriminating HCC from normal controls. The plasma DNA levels were positively associated with tumor size (P=0.012), and were significantly elevated in HCC patients with intrahepatic spreading or vascular invasion (P=0.035). The overall survival time of patients with high plasma DNA levels showed a shortened tread when compared with that of patients with low plasma DNA concentrations (P=0.071). Plasma DNA may be a valuable noninvasive tool for the detecting and predicting the metastasis potential of HCC; and the prognostic value of plasma DNA needed further investigation. C1 [Huang, Zhaohui] The Fourth Affiliated Hospital of Suzhou University, Wuxi Oncology Institute, 200 Huihe Road, 214062 Wuxi, Jiangsu Province, China. [Hua, Dong] The Fourth Affiliated Hospital of Suzhou University, Wuxi Oncology Institute, 200 Huihe Road, 214062 Wuxi, Jiangsu Province, China. [Hu, Yu] The Fourth Affiliated Hospital of Suzhou University, Wuxi Oncology Institute, 200 Huihe Road, 214062 Wuxi, Jiangsu Province, China. [Cheng, Zhihong] The Fourth Affiliated Hospital of Suzhou University, Wuxi Oncology Institute, 200 Huihe Road, 214062 Wuxi, Jiangsu Province, China. [Zhou, Xike] The Fourth Affiliated Hospital of Suzhou University, Wuxi Oncology Institute, 200 Huihe Road, 214062 Wuxi, Jiangsu Province, China. [Xie, Qigen] The Fourth Affiliated Hospital of Suzhou University, Wuxi Oncology Institute, 200 Huihe Road, 214062 Wuxi, Jiangsu Province, China. [Wang, Qiongyao] The Fourth Affiliated Hospital of Suzhou University, Wuxi Oncology Institute, 200 Huihe Road, 214062 Wuxi, Jiangsu Province, China. [Wang, Feng] The Fourth Affiliated Hospital of Suzhou University, Wuxi Oncology Institute, 200 Huihe Road, 214062 Wuxi, Jiangsu Province, China. [Du, Xiang] Fudan University Shanghai Cancer Center, Department of Pathology, 270 Dong An Road, 200032 Shanghai, China. [Zeng, Yanjun] Beijing University of Technology, Biomechanics and Medical Information Institute, 100022 Beijing, China. RP Huang, Z (reprint author), The Fourth Affiliated Hospital of Suzhou University, Wuxi Oncology Institute, 214062 Wuxi, China. EM hzhwxsy@126.com CR El-Serag HB, Rudolph KL, 2007, Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 132(7):2557–2576 Farazi PA, DePinho RA, 2006, Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer 6(9):674– 687 Farinati F, Marino D, De Giorgio M et al, 2006, Diagnostic and prognostic role of alpha-fetoprotein in hepatocellular carcinoma: both or neither? Am J Gastroenterol 101(3):524–532 Butt AN, Swaminathan R, 2008, Overview of circulating nucleic acids in plasma/serum. Ann N Y Acad Sci 1137:236–242 Huang ZH, Li LH, Hua D, 2006, Quantitative analysis of plasma circulating DNA at diagnosis and during follow-up of breast cancer patients. Cancer Lett 243(1):64–70 Gal S, Fidler C, Lo YM et al, 2004, Quantitation of circulating DNA in the serum of breast cancer patients by real-time PCR. Br J Cancer 90(6):1211–1215 Sozzi G, Conte D, Mariani L et al, 2001, Analysis of circulating tumor DNA in plasma at diagnosis and during follow-up of lung cancer patients. Cancer Res 61(12):4675–4678 Paci M, Maramotti S, Bellesia E et al, 2009, Circulating plasma DNA as diagnostic biomarker in non-small cell lung cancer. Lung Cancer 64(1):92–97 Kumar S, Guleria R, Singh V et al, 2010, Efficacy of circulating plasma DNA as a diagnostic tool for advanced non-small cell lung cancer and its predictive utility for survival and response to chemotherapy. Lung Cancer 70(2):211–217 Kamat AA, Baldwin M, Urbauer D et al, 2010, Plasma cell-free DNA in ovarian cancer: an independent prognostic biomarker. Cancer 116(8):1918–1925 Kamat AA, Sood AK, Dang D et al, 2006, Quantification of total plasma cell-free DNA in ovarian cancer using real-time PCR. Ann N Y Acad Sci 1075:230–234 Allen D, Butt A, Cahill D et al, 2004, Role of cell-free plasma DNA as a diagnostic marker for prostate cancer. Ann N Y Acad Sci 1022:76–80 Tomita H, Ichikawa D, Ikoma D et al, 2007, Quantification of circulating plasma DNA fragments as tumor markers in patients with esophageal cancer. Anticancer Res 27(4C):2737–2741 Ren N, Qin LX, Tu H et al, 2006, The prognostic value of circulating plasma DNA level and its allelic imbalance on chromosome 8p in patients with hepatocellular carcinoma. J Cancer Res Clin Oncol 132(6):399–407 Sai S, Ichikawa D, Tomita H et al, 2007, Quantification of plasma cell-free DNA in patients with gastric cancer. Anticancer Res 27, 4C):2747–2751 Hauser S, Zahalka T, Ellinger J et al, 2010, Cell-free circulating DNA: diagnostic value in patients with renal cell cancer. Anticancer Res 30(7):2785–2789 Huang ZH, Hua D, Du CH et al, 2007, Quantitation of plasma circulating DNA and its clinical value in the diagnosis and prognosis of breast cancer. Maternal Child Health Care China 22, 15):2095–2097 Leon SA, Shapiro B, Sklaroff DM et al, 1977, Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 37, 3):646–650 Kohler C, Radpour R, Barekati Z et al, 2009, Levels of plasma circulating cell free nuclear and mitochondrial DNA as potential biomarkers for breast tumors. Mol Cancer 8:105 Yoon KA, Park S, Lee SH et al, 2009, Comparison of circulating plasma DNA levels between lung cancer patients and healthy controls. J Mol Diagn 11(3):182–185 Gordian E, Ramachandran K, Reis IM et al, 2010, Serum free circulating DNA is a useful biomarker to distinguish benign versus malignant prostate disease. Cancer Epidemiol Biomarkers Prev 19(8):1984–1991 Ostrow KL, Hoque MO, Loyo M et al, 2010, Molecular analysis of plasma DNA for the early detection of lung cancer by quantitative methylation-specific PCR. Clin Cancer Res 16(13):3463–3472 Lee TH, Montalvo L, Chrebtow V et al, 2001, Quantitation of genomic DNA in plasma and serum samples: higher concentrations of genomic DNA found in serum than in plasma. Transfusion 41(2):276–282 Thijssen MA, Swinkels DW, Ruers TJ et al, 2002, Difference between free circulating plasma and serum DNA in patients with colorectal liver metastases. Anticancer Res 22(1A):421–425 Thierry AR, Mouliere F, Gongora C et al, 2010, Origin and quantification of circulating DNA in mice with human colorectal cancer xenografts. Nucleic Acids Res 38(18):6159–6175 Ren N, Ye QH, Qin LX et al, 2006, Circulating DNA level is negatively associated with the long-term survival of hepatocellular carcinoma patients. World J Gastroenterol 12(24):3911–3914 Galeazzi M, Morozzi G, Piccini M et al, 2003, Dosage and characterization of circulating DNA: present usage and possible applications in systemic autoimmune disorders. Autoimmun Rev 2(1):50–55 Antonatos D, Patsilinakos S, Spanodimos S et al, 2006, Cell-free DNA levels as a prognostic marker in acute myocardial infarction. Ann N Y Acad Sci 1075:278–281 Lippmann ML, Morgan L, Fein A et al, 1982, Plasma and serum concentrations of DNA in pulmonary thromboembolism. Am Rev Respir Dis 125(4):416–419 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 271 EP 276 DI 10.1007/s12253-011-9438-z PG 6 ER PT J AU Hager, M Haufe, H Alinger, B Kolbitsch, Ch AF Hager, Martina Haufe, Heike Alinger, Beate Kolbitsch, Christian TI pS6 Expression in Normal Renal Parenchyma, Primary Renal Cell Carcinomas and their Metastases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Kidney cancer; Metastases; Normal renal parenchyma; pS6; Renal cell carcinoma ID Kidney cancer; Metastases; Normal renal parenchyma; pS6; Renal cell carcinoma AB In cancer therapy novel concepts focus on phosphoinositide-3-kinase/protein kinase B/mammalian target of rapamycin (mTOR) inhibitors. In this context, phosphorylated S6 protein of the 40S ribosomal subunit (pS6) overexpression was previously shown to be associated with sensitivity to inhibitors of mTOR. The present study therefore evaluated pS6 expression in normal renal parenchyma (NRP), primary renal cell carcinomas (PRCC) and their metastases. pS6 and pmTOR expression was immunohistochemically analyzed in a tissue microarray (TMA) from localized primary renal cell carcinoma (lPRCC) (n=35), metastasized primary renal cell carcinoma (mPRCC) (n=45), their metastases (n=45), and NRP (n=45). pS6 expression was stronger in mPRCCs and metastases than in NRP and lPRCCs (p<0.05). In mPRCCs high-grade and high-stage tumors showed higher pS6 levels. pS6 overexpression was more frequently found in metastases (40/45; 88.9%) than in mPRCC (24/45; 53.3%) (p<0.05). Overexpression of pS6 in metastases without concomitant overexpression in their primary tumors was found in 16/45 (35.56%) cases. Patients with pS6 overexpression in mPRCCs but also in metastases showed a tendency to shorter overall survival. pS6 score and pmTOR score correlated positively in NRP and in tumorous tissue (mPRCC and metastases). In conclusion, the present study showed stronger pS6 expression and more frequent overexpression in metastases than in corresponding PRCCs. In approximately one-third of the cases pS6 overexpression was found exclusively in metastases, which is interesting with regard to the association between high pS6 expression and sensitivity to mTOR inhibitor therapy. C1 [Hager, Martina] General Hospital and Paracelsus Medical University Salzburg, Department of Pathology, Muellner Hauptstrasse 48, 5020 Salzburg, Austria. [Haufe, Heike] General Hospital and Paracelsus Medical University Salzburg, Department of Pathology, Muellner Hauptstrasse 48, 5020 Salzburg, Austria. [Alinger, Beate] General Hospital and Paracelsus Medical University Salzburg, Department of Pathology, Muellner Hauptstrasse 48, 5020 Salzburg, Austria. [Kolbitsch, Christian] Innsbruck Medical University (MUI), Department of Anaesthesiology and Intensive Care MedicineInnsbruck, Austria. RP Hager, M (reprint author), General Hospital and Paracelsus Medical University Salzburg, Department of Pathology, 5020 Salzburg, Austria. EM hager.martina@gmx.at CR Dufner A, Thomas G, 1999, Ribosomal S6 kinase signaling and the control of translation. Exp Cell Res 253:100–109 Ferrari S, Bandi HR, Hofsteenge J et al, 1991, Mitogen-activated 70K S6 kinase. Identification of in vitro 40 S ribosomal S6 phosphorylation sites. J Biol Chem 266:22770–22775 Bose S, Chandran S, Mirocha JM et al, 2006, The Akt pathway in human breast cancer: a tissue-array-based analysis. Mod Pathol 19:238–245 Cho D, Signoretti S, Dabora S et al, 2007, Potential histologic and molecular predictors of response to temsirolimus in patients with advanced renal cell carcinoma. Clin Genitourin Canc 5:379– 385 Chow S, Minden MD, Hedley DW, 2006, Constitutive phosphorylation of the S6 ribosomal protein via mTOR and ERK signaling in the peripheral blasts of acute leukemia patients. Exp Hematol 34:1183–1191 Cotler SJ, Hay N, Xie H et al, 2008, Immunohistochemical expression of components of the Akt-mTORC1 pathway is associated with hepatocellular carcinoma in patients with chronic liver disease. Dig Dis Sci 53:844–849 Iwenofu OH, Lackman RD, Staddon AP et al, 2008, Phospho-S6 ribosomal protein: a potential new predictive sarcoma marker for targeted mTOR therapy. Mod Pathol 21:231–237 Pantuck AJ, Seligson DB, Klatte T et al, 2007, Prognostic relevance of the mTOR pathway in renal cell carcinoma: implications for molecular patient selection for targeted therapy. Cancer 109:2257–2267 Robb VA, Karbowniczek M, Klein-Szanto AJ et al, 2007, Activation of the mTOR signaling pathway in renal clear cell carcinoma. J Urol 177:346–352 Trinh XB, Tjalma WA, Vermeulen PB et al, 2009, The VEGF pathway and the AKT/mTOR/p70S6K1 signalling pathway in human epithelial ovarian cancer. Br J Canc 100:971–978 Fresno Vara JA, Casado E, De Castro J et al, 2004, PI3K/Akt signalling pathway and cancer. Canc Treat Rev 30:193–204 Hay N, Sonenberg N, 2004, Upstream and downstream of mTOR. Gene Dev 18:1926–1945 Hudes G, Carducci M, Tomczak P et al, 2007, Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. New Engl J Med 356:2271–2281 Kapoor A, Figlin RA, 2009, Targeted inhibition of mammalian target of rapamycin for the treatment of advanced renal cell carcinoma. Cancer 115:3618–3630 Merseburger AS, Kuczyk MA, 2008, Value of targeted therapies for renal cell cancer. Urologe A 47:1303–1310 Fuhrman SA, Lasky LC, Limas C, 1982, Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol 6:655–663 Eble JN, Sauter G, Epstein JI et al, 2004, WHO classification of tumours. Tumors of the urinary system and male genital organs. IARC, Lyon, pp 23–34 Campbell L, Jasani B, Edwards K et al, 2008, Combined expression of caveolin-1 and an activated AKT/mTOR pathway predicts reduced disease-free survival in clinically confined renal cell carcinoma. Br J Canc 98:931–940 Klatte T, SeligsonDB, La Rochelle J et al, 2009, Molecular signatures of localized clear cell renal cell carcinoma to predict disease-free survival after nephrectomy. Canc Epidemiol Biomarkers Prev 18:894–900 Amato RJ, Jac J, Giessinger S et al, 2009, A phase 2 study with a daily regimen of the oral mTOR inhibitor RAD001, everolimus, in patients with metastatic clear cell renal cell cancer. Cancer 115:2438–2446 Atkins MB, Hidalgo M, Stadler WM et al, 2004, Randomized phase II study of multiple dose levels of CCI-779, a novel mammalian target of rapamycin kinase inhibitor, in patients with advanced refractory renal cell carcinoma. J Clin Oncol 22:909– 918 Motzer RJ, Escudier B, Oudard S et al, 2008, Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372:449– 456 Patel PH, Chadalavada RS, Chaganti RS et al, 2006, Targeting von Hippel-Lindau pathway in renal cell carcinoma. Clin Canc Res 12:7215–7220 Akcakanat A, Sahin A, Shaye AN et al, 2008, Comparison of Akt/mTOR signaling in primary breast tumors and matched distant metastases. Cancer 112:2352–2358 Ljungberg B, Cowan NC, Hanbury DC et al, 2010, EAU guidelines on renal cell carcinoma: the 2010 update. Eur Urol 58:398–406 Youssif TA, Fahmy MA, Koumakpayi IH et al, 2011, The mammalian target of rapamycin pathway is widely activated without PTEN deletion in renal cell carcinoma metastases. Cancer 117:290– 300 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 277 EP 283 DI 10.1007/s12253-011-9439-y PG 7 ER PT J AU Favaro, JW Hetzl, CA Reis, OL Ferreira, U Billis, A Cagnon, HAV AF Favaro, Jose Wagner Hetzl, Cia Amanda Reis, Oliveira Leonardo Ferreira, Ubirajara Billis, Athanase Cagnon, Helena A Valeria TI Periacinar Retraction Clefting in Nonneoplastic and Neoplastic Prostatic Glands: Artifact or Molecular Involvement SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostatic cancer; Histological criteria; Retraction clefting; Dystroglycans; Matrix metalloproteinases; Insulin-like growth factor; Fibroblast growth factor; Vimentin; Laminin; p63 ID Prostatic cancer; Histological criteria; Retraction clefting; Dystroglycans; Matrix metalloproteinases; Insulin-like growth factor; Fibroblast growth factor; Vimentin; Laminin; p63 AB A space between neoplastic acini and prostatic stroma is not rare and studies have interpreted this as an artifact, considering the absence of endothelial cells indicating vascular invasion. Thus, the aims of this work were to characterize and correlate the occurrence and extent of retraction clefting with the reactivities of α and β dystroglycan (αDG, βDG), laminin, matrix metalloproteinase 2 (MMP-2), p63, insulin-like growth factor 1(IGF-1), vimentin, and fibroblast growth factor 2 (FGF-2). The study was based on nonneoplastic and neoplastic prostatic tissues obtained from necropsies and retropubic radical prostatectomies. The results showed that periacinar retraction clefting was significantly more frequent in prostatic carcinoma samples than in normal prostatic acini. Most of the neoplastic acini (72.0%) showed retraction clefting of more than 50% of circumference, which were significantly more frequent in Gleason score 7 and 6. Decreased collagen and reticular and elastic fibers were verified in the stroma around neoplastic acini. Weak and discontinuous αDG, βDG, and laminin immunoreactivities and intensified MMP-2, vimentin, IGF-1 and FGF-2 immunoreactivities were verified in the neoplastic acini; p63 immunoreactivity was negative in all carcinomas. Thus, these findings showed that the lack of epithelial basal cells, DGs, and laminin and increased MMP-2, IGF-1, and FGF-7 could be considered important pathways in periacinar retraction occurrence. This study demonstrated the origin of and the biological mechanisms responsible for periacinar retraction clefting in prostatic carcinoma. C1 [Favaro, Jose Wagner] Univ Estadual Paulista (UNESP), Institute of Biosciences, Department of Anatomy, CP-510, 18618-970 Botucatu, SP, Brazil. [Hetzl, Cia Amanda] UNICAMP - University of Campinas, Institute of Biology, Department of Anatomy, Cellular Biology, Physiology and BiophysicsCampinas, SP, Brazil. [Reis, Oliveira Leonardo] UNICAMP - University of Campinas, School of Medicine, Department of UrologyCampinas, SP, Brazil. [Ferreira, Ubirajara] UNICAMP - University of Campinas, School of Medicine, Department of UrologyCampinas, SP, Brazil. [Billis, Athanase] UNICAMP - University of Campinas, School of Medicine, Department of PathologyCampinas, SP, Brazil. [Cagnon, Helena A Valeria] UNICAMP - University of Campinas, Institute of Biology, Department of Anatomy, Cellular Biology, Physiology and BiophysicsCampinas, SP, Brazil. RP Favaro, JW (reprint author), Univ Estadual Paulista (UNESP), Institute of Biosciences, Department of Anatomy, 18618-970 Botucatu, Brazil. EM wjfavaro@ibb.unesp.br CR Epstein JI, 1995, Diagnostic criteria of limited adenocarcinoma of the prostate on needle biopsy. Hum Pathol 26:223–229 Varma M, Lee MW, Tamboli P, Zarbo RJ, Jimenez RE, Salles PGO, Amin MB, 2002, Morphologic criteria for the diagnosis of prostatic adenocarcinoma in needle biopsy specimens. Arch Pathol Lab Med 126:554–561 Kruslin B, Tomas D, Rogatsch H, Novosel I, Cupic H, Belicza M, Kraus O, Mikuz G, 2003, Periacinar retraction clefting in the prostatic needle core biopsies: an important diagnostic criterion or a simple artifact? Virchows Arch 443:524–527 Baisden BL, Kahane H, Epstein JI, 1999, Perineural invasion, mucinous fibroplasia and glomerulations: diagnostic features on prostate needle biopsy. Am J Surg Pathol 23:918–924 Durham JR, Lee MW, Amin MB, Shulz DS, Stetler-Stevenson WG, 1994, Clefting in adenocarcinoma of the prostate gland. Am J Clin Pathol 102:536 Kruslin B, Novosel I, Reljic A, Cupic H, Spajic B, Belicza M, 2002, Periacinar cleft-like spaces in prostatic needle core biopsies. Acta Clin Croat 41:175 Kruslin B, Tomas D, Cviko A, 2006, Periacinar retraction clefting and p63 immunostaining in prostatic intraepithelial neoplasia and prostatic carcinoma. Pathol Oncol Res 12:205–209 Dean JP, Nelson PS, 2008, Profiling influences of senescent and aged fibroblasts on prostate carcinogenesis. Br J Cancer 98:245– 249 Cornell RJ, Rowley D, Wheller T, 2003, Neuroepithelial interactions in prostate cancer are enhanced in the presence of prostatic stroma. Urology 61:870–875 Sgambato A, Migaldi M, Montanari M, Camerini A, Brancaccio A, Rossi G, Cangiano R, Losasso C, Capelli G, Trentini GP, Cittadini A, 2003, Dystroglycan expression is frequently reduced in human breast and colon cancers and is associated with tumor progression. Am J Pathol 162:849–860 Brennan PA, Jing J, Ethunandan M, Gorecki D, 2004, Dystroglycan complex in cancer. Eur J Surg Oncol 30:589–592 Sgambato A, De Paola B, Migaldi M, Di Salvatore M, Rettino A, Rossi G, Faraglia B, Boninsegna A, Maiorana A, Cittadini A, 2007, Dystroglycan expression is reduced during prostate tumorigenesis and is regulated by androgens in prostate cancer cells. J Cell Physiol 213:528–539 Djavan B, Waldert M, Seitz C, Marberger M, 2001, Insulin-like growth factors and prostate cancer. World J Urol 19:225–233 Reynolds AR, Kyprianou N, 2006, Growth factor signalling in prostatic growth: significance in tumour development and therapeutic targeting. Br J Pharmacol 2:S144–S152 Mostofi FK, Price EBJr, 1973, Tumors of the male genital system, atlas of tumor pathology, second series, fascicle 8. Armed Forces Institute of Pathology, Washington, pp 202–217 Tomas D, Kruslin B, 2004, The potential value of, Myo, fibroblastic stromal reaction in the diagnosis of prostatic adenocarcinoma. Prostate 61:324–331 Halpert B, Sheehan EA, Schmalhorst WR, Scott RJ, 1963, Carcinoma of the prostate: a survey of 5000 autopsies. Cancer 16:736–742 Kruslin B, Tomas D, Rogatsch H, Reljic A, Vucic M, Balicevic D, Belicza M, Mikuz G, 2005, Correlation of periacinar retraction clefting in needle core biopsies and corresponding prostatectomy specimens of patients with prostatic adenocarcinoma. Int J Surg Pathol 13:67–72 Young RH, Srigley JR, Amin MB, Ulbright TM, Cubilla AL, 1998, Atlas of tumor pathology. AFIP, Washington, pp 124–171 Tomas D, Ulamec M, Hudolin T, 2006, Myofibroblastic stromal reaction and expression of tenascin-C and laminin in prostate adenocarcinoma. Prostate Cancer Prostatic Dis 9:414–419 Tomas D, Spajic B,Milosevic M, Demirovic A, Marusic Z, Kruslin B, 2011, Extensive retraction artefact predicts biochemical recurrencefree survival in prostatic carcinoma. Histopathology 58:447–454 Djakiew D, 2000, Deregulated expression of growth factors and their receptors in the development of prostate cancer. Prostate 42:150–160 Stangelberger A, Schally AV, Jl V, Hammann BD, Groot K, Halmos G, Cai RZ, Zarandi M, 2005, Antagonists of growth hormone releasing hormone, GHRH, and of bombesin/gastrin releasing peptide, BN/GRP, suppress the expression of VEGF, bFGF, and receptors of the EGF/HERfam ily in PC-3 and DU-145 human androgen-independent prostate cancers. Prostate 64:303–315 Garrison JB, Kyprianou N, 2004, Novel targeting of apoptosis pathways for prostate cancer therapy. Cur Cancer Drug Targets 4:85–95 Stattin P, Rinaldi S, Biessy C, Stenman Uh, Hallmans G, Kaaks R, 2004, High levels of circulating insulin-like growth factor-I increase prostate cancer risk: a prospective study in a populationbased nonscreened cohort. J Clin Oncol 22:3104–3112 Kwabi-Addo B, Ozen M, Ittmann M, 2004, The role of fibroblast growth factors and their receptors in prostate cancer. Endocr Relat Cancer 11:709–724 Aigner A, Butscheid M, Kunkel P, Krause E, Lamszus K, Wellstein A, Czubayko F, 2001, An FGF-binding protein, FGF-BP, exerts its biological function by parallel paracrine stimulation of tumor cell and endothelial cell proliferation through FGF-2 release. Int J Cancer 92:510–517 Giri D, Ropiquet F, Ittmann M, 1999, Alterations in expression of basic fibroblast growth factor, FGF, 2 and its receptor FGFR-1 in human prostate cancer. Clin Cancer Res 5:1063–1071 Lynch C, Matrisian L, 2002, Matrix metalloproteinases in tumorhost cell communication. Differentiation 70:561–573 Ross J, Kaur P, Sheehan C, Hugh B, Fisher H, Kauffman R, Kallakury B, 2003, Prognostic significance of matrix metalloproteinase 2 and tissue inhibitor of metalloproteinase 2 expression in prostate cancer. Mod Pathol 16:198–205 Martin MD, Matrisian LM, 2007, The other side of MMPs: protective roles in tumor progression. Cancer Metastasis Rev 26:717–724 Mack CF, Knox JD, Powell WC, Nagle RB, Bowden GT, 1993, Functional-role of the metalloproteinase matrilysin in human prostate-cancer. Int J Radiat Oncol Biol Phys 27:217 Gianelli G, Falk-Marzillier J, Schiraldi O, Stetler-Stevenson W, Quaranta V, 1997, Induction of cell migration by matrix metalloproteinase-2 cleavage of laminin-5. Science 277:225–228 Yamada H, Saito F, Fukuta-Ohi H, Zhong D, Hase A, Arai K, Okuyama A, Maekawa R, Shimizu T, Matsumura K, 2001, Processing of β-dystroglycan by matrix metalloproteinase disrupts the link between the extracellular matrix and cell membrane via the dystroglycan complex. Hum Mol Genet 10:1563–1569 Henry MD, Cohen MB, Campbell KP, 2001, Reduced expression of dystroglican in breast and prostate cancer. Hum Pathol 32:791– 795 Gilles C, Thompson EW, 1996, The epithelial to mesenchymal transition and metastatic progression in carcinoma. Breast J 2:83– 96 Thiery JP, 2002, Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2:442–454 Zhao Y, Yan Q, Long X, Chen X, Wang Y, 2008, Vimentin affects the mobility and invasiveness of prostate cancer cells. Cell Biochem Funct 26:571–577 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 285 EP 292 DI 10.1007/s12253-011-9440-5 PG 8 ER PT J AU Jiang, W Wang, Z Li, X Fan, X Duan, Y AF Jiang, Wei Wang, Zhiming Li, Xinying Fan, Xuegong Duan, Yankun TI High-mobility Group Box 1 is Associated with Clinicopathologic Features in Patients with Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma (HCC); Clinicopathology; High-mobility group box 1(HMGB1); Reverse transcription–polymerase chain reaction (RT–PCR); Gene expression ID Hepatocellular carcinoma (HCC); Clinicopathology; High-mobility group box 1(HMGB1); Reverse transcription–polymerase chain reaction (RT–PCR); Gene expression AB High-mobility group box 1(HMGB1) has been associated with many human cancers, but the role of HMGB1 in hepatocellular carcinoma (HCC) remains unclear. The aim of this study is to investigate the expression of HMGB1 in human HCC with regard to its clinical significance. Twelve cases of normal liver tissues, 34 cases of HCC and the corresponding liver tissue just around the tumor (LAT) were collected. Then, all the samples were subjected to clinicopathologic examination, reverse transcription–polymerase chain reaction (RT–PCR), Western-blot (WB) and immunohistochemical analysis for the expression of HMGB1. The relationships between HMGB1 mRNA expression and clinicopathologic parameters were analyzed. RT–PCR demonstrated that the expression of relative HMGB1 mRNA (HMGB1/GAPDH) was 0.854±0.172; the highest in the tissue of HCC, significantly up-regulated compared with that of 0.527±0.155 in LAT and of 0.405±0.087 in normal liver tissues (P<0.001). HMGB1 mRNA overexpression was significantly associated with Edmondson stage, TNM stage, vascular invasion and capsule invasion. Western-blot showed the expression of HMGB1 protein in HCC also as the highest among all the groups. Furthermore this overexpression revealed by immunostaining was predominantly localized in the nuclei of HCC; whereas, none of the stains were seen in normal liver cells and only a trace of it was detected in the cytoplasm of LAT cells. Our results suggested the overexpression of HMGB1 might be an important pathogenetic factor in HCC. The mechanisms of HMGB1 in HCC genesis, development and its possible diagnostic and prognostic roles need to be further explored. C1 [Jiang, Wei] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan, China. [Wang, Zhiming] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan, China. [Li, Xinying] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan, China. [Fan, Xuegong] Central South University, Xiangya Hospital, Department of Infectious Diseases, 410008 Changsha, Hunan, China. [Duan, Yankun] Central South University, Xiangya Hospital, Department of Infectious Diseases, 410008 Changsha, Hunan, China. RP Wang, Z (reprint author), Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, China. EM wzm005@yahoo.com.cn CR Srivatanakul P, Sriplung H, Deerasamee S, 2004, Epidemiology of liver cancer: an overview. Asian Pac J Cancer Prev 5:118–125 Yao DF, Dong ZZ, Yao M, 2007, Specific molecular markers in hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 6:241– 247 El-Serag HB, Rudolph KL, 2007, Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 132:2557–2576 Bosch FX, Ribes J, Cleries R et al, 2005, Epidemiology of hepatocellular carcinoma. Clin Liver Dis 9:191–211 Javaherian K, Liu JF, Wang JC, 1978, Nonhistone proteins HMG1 and HMG2 change the DNA helical structure. Science 199:1345– 1346 Lotze MT, Tracey KJ, 2005, High-mobility group box 1 protein, HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol 5:331–342 Czura CJ, Wang H, Tracey KJ, 2001, Dual roles for HMGB1: DNA binding and cytokine. J Endotoxin Res 7:315–321 Muller S, Scaffidi P, Degryse B et al, 2001, New EMBO members’ review: the double life of HMGB1 chromatin protein: architectural factor and extracellular signal. EMBO J 20:4337– 4340 Kostova N, Zlateva S, Ugrinova I et al, 2010, The expression of HMGB1 protein and its receptor RAGE in human malignant tumors. Mol Cell Biochem 337:251–258 Akaike H, Kono K, Sugai H et al, 2007, Expression of high mobility group box chromosomal protein-1, HMGB-1, in gastric cancer. Anticancer Res 27:449–457 Brezniceanu ML, Volp K, Bosser S et al, 2003, HMGB1 inhibits cell death in yeast and mammalian cells and is abundantly expressed in human breast carcinoma. FASEB J 17:1295–1297 Wu D, Ding Y, Wang S et al, 2008, Increased expression of high mobility group box 1, HMGB1, is associated with progression and poor prognosis in human nasopharyngeal carcinoma. J Pathol 216:167–175 Liu Y, Xie C, Zhang X et al, 2010, Elevated expression of HMGB1 in squamous-cell carcinoma of the head and neck and its clinical significance. Eur J Cancer 46:3007–3015 Schlueter C, Weber H, Meyer B et al, 2005, Angiogenetic signaling through hypoxia: HMGB1: an angiogenetic switch molecule. Am J Pathol 166:1259–1263 Dong Xda E, Ito N, Lotze MT et al, 2007, High mobility group box I, HMGB1, release from tumor cells after treatment: implications for development of targeted chemoimmunotherapy. J Immunother 30:596–606 Kuniyasu H, Oue N, Wakikawa A et al, 2002, Expression of receptors for advanced glycation end-products, RAGE, is closely associated with the invasive and metastatic activity of gastric cancer. J Pathol 196:163–170 Edmondson HA, Steiner PE, 1954, Primary carcinoma of the liver: a study of 100 cases among 48,900 necropsies. Cancer 7:462–503 Greene FL, Page DL, Fleming ID et al, 2002, AJCC cancer staging manual, 6th edn. Springer, New York Thomas JO, Travers AA, 2001, HMG1 and 2, and related ‘architectural’ DNA-binding proteins. Trends Biochem Sci 26:167–174 Ghavami S, Rashedi I, Dattilo BM et al, 2008, S100A8/A9 at low concentration promotes tumor cell growth via RAGE ligation and MAP kinase-dependent pathway. J Leukoc Biol 83:1484–1492 Palumbo R, Galvez BG, Pusterla T et al, 2007, Cells migrating to sites of tissue damage in response to the danger signal HMGB1 require NF-kappaB activation. J Cell Biol 179:33–40 Gardella S, Andrei C, Ferrera D et al, 2002, The nuclear protein HMGB1 is secreted by monocytes via a non-classical, vesiclemediated secretory pathway. EMBO Rep 3:995–1001 Bonaldi T, Talamo F, Scaffidi P et al, 2003, Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. EMBO J 22:5551–5560 Cheng BQ, Jia CQ, Liu CT et al, 2008, Serum high mobility group box chromosomal protein 1 is associated with clinicopathologic features in patients with hepatocellular carcinoma. Dig Liver Dis 40:446–452 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 293 EP 298 DI 10.1007/s12253-011-9442-3 PG 6 ER PT J AU Gaida, MM Bach, TS Gunther, F Baseras, B Tschaharganeh, FD Welsch, Th Felix, K Bergmann, F Hansch, MG Wente, NM AF Gaida, M Matthias Bach, T Sylvia Gunther, Frank Baseras, Billur Tschaharganeh, F Darjus Welsch, Thilo Felix, Klaus Bergmann, Frank Hansch, M Gertrud Wente, N Moritz TI Expression of Galectin-3 in Pancreatic Ductal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic cancer; Galectins; Galectin-3; Expression ID Pancreatic cancer; Galectins; Galectin-3; Expression AB Galectin-3 influences neoangiogenesis, tumor cell adhesion, and tumor-immune-escape mechanisms. Hence, the expression of galectin-3 in pancreatic ductal adenocarcinoma (PDAC) was evaluated. Galectin-3 expression in PDAC cell lines was proven by the presence of intracellular protein and by release into the supernatant. Furthermore, galectin-3 was found in the majority of human tissue samples. Serum concentrations of galectin-3 in PDAC patients did not differ significantly from healthy donors and did not correlate with established tumor markers. In conclusion, galectin-3 is expressed in PDAC tissues suggesting a role in tumor development; however, no relationship between expression and clinical findings could be established. C1 [Gaida, M Matthias] University of Heidelberg, Department of Pathology, 69120 Heidelberg, Germany. [Bach, T Sylvia] University of Heidelberg, Department of Pathology, 69120 Heidelberg, Germany. [Gunther, Frank] University of Heidelberg, Institute of Immunology, 69120 Heidelberg, Germany. [Baseras, Billur] University of Heidelberg, Institute of Immunology, 69120 Heidelberg, Germany. [Tschaharganeh, F Darjus] University of Heidelberg, Department of Pathology, 69120 Heidelberg, Germany. [Welsch, Thilo] University of Heidelberg, Department of Surgery, 69120 Heidelberg, Germany. [Felix, Klaus] University of Heidelberg, Department of Surgery, 69120 Heidelberg, Germany. [Bergmann, Frank] University of Heidelberg, Department of Pathology, 69120 Heidelberg, Germany. [Hansch, M Gertrud] University of Heidelberg, Institute of Immunology, 69120 Heidelberg, Germany. [Wente, N Moritz] University of Heidelberg, Department of Surgery, 69120 Heidelberg, Germany. RP Wente, NM (reprint author), University of Heidelberg, Department of Surgery, 69120 Heidelberg, Germany. EM moritz.wente@aesculap.de CR Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60:277–300 Li D, Xie K, Wolff R, Abbruzzese JL, 2004, Pancreatic cancer. Lancet 363:1049–1057 Warshaw AL, Fernandez del Castillo C, 1992, Pancreatic carcinoma. N Engl J Med 326:455–465 Thijssen VL, Postel R, Brandwijk RJ, Dings RP, Nesmelova I, Satijn S, Verhofstad N, Nakabeppu Y, Baum LG, Bakkers J, Mayo KH, Poirier F, Griffioen AW, 2006, Galectin-1 is essential in tumor angiogenesis and is a target for antiangiogenesis therapy. Proc Natl Acad Sci USA 103:15975–15980 Goetz JG, Joshi B, Lajoie P, Strugnell SS, Scudamore T, Kojic LD, Nabi IR, 2008, Concerted regulation of focal adhesion dynamics by galectin-3 and tyrosine-phosphorylated caveolin-1. J Cell Biol 180:1261–1275 Hittelet A, Legendre H, Nagy N, Bronckart Y, Pector JC, Salmon I, Yeaton P, Gabius HJ, Kiss R, Camby I, 2003, Upregulation of galectins-1 and -3 in human colon cancer and their role in regulating cell migration. Int J Cancer 103:370–379 Rubinstein N, Alvarez M, Zwirner NW, Toscano MA, Ilarregui JM, Bravo A, Mordoh J, Fainboim L, Podhajcer OL, Rabinovich GA, 2004, Targeted inhibition of galectin-1 gene expression in tumor cells results in heightened T cell-mediated rejection; A potential mechanism of tumor-immune privilege. Cancer Cell 5:241–251 Canesin G, Gonzalez-Peramato P, Palou J, Urrutia M, Cordon- Cardo C, Sanchez-Carbayo M, 2010, Galectin-3 expression is associated with bladder cancer progression and clinical outcome. Tumour Biol 31:277–285 Sakaki M, Oka N, Nakanishi R, Yamaguchi K, Fukumori T, Kanayama HO, 2008, Serum level of galectin-3 in human bladder cancer. J Med Invest 55:127–132 Yang RY, Rabinovich GA, Liu FT, 2008, Galectins: structure, function and therapeutic potential. Expert Rev Mol Med 10:e17 Dumic J, Dabelic S, Flogel M, 2006, Galectin-3: an open-ended story. Biochim Biophys Acta 1760:616–635 Liu FT, Hsu DK, 2007, The role of galectin-3 in promotion of the inflammatory response. Drug News Perspect 20:455–460 de Boer RA, Yu L, van Veldhuisen DJ, 2010, Galectin-3 in cardiac remodeling and heart failure. Curr Heart Fail Rep 7:1–8 de Boer RA, Lok DJ, Jaarsma T, van der MP, Voors AA, Hillege HL, van Veldhuisen DJ, 2011, Predictive value of plasma galectin- 3 levels in heart failure with reduced and preserved ejection fraction. Ann Med 43:60–68 Lok DJ, van der MP, de la Porte PW, Lipsic E, Van WJ, Hillege HL, van Veldhuisen DJ, 2010, Prognostic value of galectin-3, a novel marker of fibrosis, in patients with chronic heart failure: data from the DEAL-HF study. Clin Res Cardiol 99:323–328 Cvejic D, Savin S, Petrovic I, Selemetjev S, Paunovic I, Tatic S, Havelka M, 2005, Galectin-3 and proliferating cell nuclear antigen, PCNA, expression in papillary thyroid carcinoma. Exp Oncol 27:210–214 Zaia PL, Oshima CT, de Oliveira LF, ndrade Scherholz PL, Manoukian FN, 2010, Immunoexpression of galectin-3 in colorectal cancer and its relationship with survival. J Gastrointest Cancer. Choi JY, Cho SI, Do NY, Kang CY, Lim SC, 2010, Clinical significance of the expression of galectin-3 and Pim-1 in laryngeal squamous cell carcinoma. J Otolaryngol Head Neck Surg 39:28–34 Sakaki M, Fukumori T, Fukawa T, Elsamman E, Shiirevnyamba A, Nakatsuji H, Kanayama HO, 2010, Clinical significance of Galectin-3 in clear cell renal cell carcinoma. J Med Invest 57:152– 157 Vereecken P, Awada A, Suciu S, Castro G, Morandini R, Litynska A, Lienard D, Ezzedine K, Ghanem G, Heenen M, 2009, Evaluation of the prognostic significance of serum galectin-3 in American Joint Committee on Cancer stage III and stage IV melanoma patients. Melanoma Res 19:316–320 Wang L, Friess H, Zhu Z, Frigeri L, Zimmermann A, Korc M, Berberat PO, Buchler MW, 2000, Galectin-1 and galectin-3 in chronic pancreatitis. Lab Invest 80:1233–1241 Geers C, Moulin P, Gigot JF, Weynand B, Deprez P, Rahier J, Sempoux C, 2006, Solid and pseudopapillary tumor of the pancreas–review and new insights into pathogenesis. Am J Surg Pathol 30:1243–1249 Berberat PO, Friess H, Wang L, Zhu Z, Bley T, Frigeri L, Zimmermann A, Buchler MW, 2001, Comparative analysis of galectins in primary tumors and tumor metastasis in human pancreatic cancer. J Histochem Cytochem 49:539–549 Shimamura T, Sakamoto M, Ino Y, Shimada K, Kosuge T, Sato Y, Tanaka K, Sekihara H, Hirohashi S, 2002, Clinicopathological significance of galectin-3 expression in ductal adenocarcinoma of the pancreas. Clin Cancer Res 8:2570–2575 Hamilton SR, Aaltonen LA, 2000, World Health Organisation classification of tumours. Pathology and genetics of tumours of the digestive system. IARC, Lyon Sobin LH, Gospodarowicz MK, Wittekind C, 2009, TNM classification of malignant tumours. Wiley-Blackwell, Oxford Bergmann F, Breinig M, Hopfner M, Rieker RJ, Fischer L, Kohler C, Esposito I, Kleeff J, Herpel E, Ehemann V, Friess H, Schirmacher P, Kern MA, 2009, Expression pattern and functional relevance of epidermal growth factor receptor and cyclooxygenase-2: novel chemotherapeutic targets in pancreatic endocrine tumors? Am J Gastroenterol 104:171–181 Ceyhan GO, Bergmann F, Kadihasanoglu M, Erkan M, Park W, Hinz U, Giese T, Muller MW, Buchler MW, Giese NA, Friess H, 2007, The neurotrophic factor artemin influences the extent of neural damage and growth in chronic pancreatitis. Gut 56:534– 544 Erkan M, Kleeff J, Esposito I, Giese T, Ketterer K, Buchler MW, Giese NA, Friess H, 2005, Loss of BNIP3 expression is a late event in pancreatic cancer contributing to chemoresistance and worsened prognosis. Oncogene 24:4421–4432 Prieto VG, Mourad-Zeidan AA, Melnikova V, Johnson MM, Lopez A, Diwan AH, Lazar AJ, Shen SS, Zhang PS, Reed JA, Gershenwald JE, Raz A, Bar-Eli M, 2006, Galectin-3 expression is associated with tumor progression and pattern of sun exposure in melanoma. Clin Cancer Res 12:6709–6715 Xu XC, el-Naggar AK, Lotan R, 1995, Differential expression of galectin-1 and galectin-3 in thyroid tumors. Potential diagnostic implications. Am J Pathol 147:815–822 Saussez S, Glinoer D, Chantrain G, Pattou F, Carnaille B, Andre S, Gabius HJ, Laurent G, 2008, Serum galectin-1 and galectin-3 levels in benign and malignant nodular thyroid disease. Thyroid 18:705–712 Koopmann J, Thuluvath PJ, Zahurak ML, Kristiansen TZ, Pandey A, Schulick R, Argani P, Hidalgo M, Iacobelli S, Goggins M, Maitra A, 2004, Mac-2-binding protein is a diagnostic marker for biliary tract carcinoma. Cancer 101:1609–1615 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 299 EP 307 DI 10.1007/s12253-011-9444-1 PG 9 ER PT J AU Rizvi, AM Alam, Sh Mehdi, JS Ali, A Batra, S AF Rizvi, Alam Moshahid Alam, Shabbir Mehdi, Jafar Syed Ali, Asgar Batra, Swaraj TI Allelic Loss of 10q23.3, the PTEN Gene Locus in Cervical Carcinoma from Northern Indian Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Loss of heterozygosity; Squmaous cell carcinoma; Cervical carcinoma; Tumor suppressor gene ID Loss of heterozygosity; Squmaous cell carcinoma; Cervical carcinoma; Tumor suppressor gene AB Cervical cancer is one of the most common malignant diseases affecting women worldwide. Studies on loss of heterozygosity have been made for PTEN gene specific microsatellite markers in malignancies like breast, ovary and lungs and the results have shown a significant association. However the role of this gene is not clearly understood in cervical cancer from Indian population. A total of 135 cervical carcinoma tissues samples were analyzed for loss of heterozygosity. DNA was isolated from the samples and their matched control specimens. Polymerase chain reaction was performed using primer specific for two intragenic markers (D10S198 & D10S192) and one marker (D10S541) in flanking region and further electrophoresed on 8% denaturing polyacrylamide gel. Overall, 31 out of 133 (23%) informative cases showed loss of heterozygosity in at least one locus in the region examined. The percentage of loss of heterozygosity for these markers ranged from 8% (D10S192) to 13% (D10S198). Loss of heterozygosity was more frequently detected in intragenic region (D10S198 & D10S192) than in flanking region, D10S541 (21% versus 9%). These data argue that PTEN is a tumor suppressor gene whose inactivation may play an important role in the carcinoma of uterine cervix. C1 [Rizvi, Alam Moshahid] Jamia Millia Islamia, Department of Biosciences, Genome Biology Lab, Maulana Mohammad Ali Jauhar Marg, 110025 New Delhi, India. [Alam, Shabbir] Jamia Millia Islamia, Department of Biosciences, Genome Biology Lab, Maulana Mohammad Ali Jauhar Marg, 110025 New Delhi, India. [Mehdi, Jafar Syed] Jamia Millia Islamia, Department of Biosciences, Genome Biology Lab, Maulana Mohammad Ali Jauhar Marg, 110025 New Delhi, India. [Ali, Asgar] Jamia Millia Islamia, Department of Biosciences, Genome Biology Lab, Maulana Mohammad Ali Jauhar Marg, 110025 New Delhi, India. [Batra, Swaraj] LNJP/MAMC Campus, Department of Obstetrics & Gynecology, 110002 New Delhi, India. RP Rizvi, AM (reprint author), Jamia Millia Islamia, Department of Biosciences, Genome Biology Lab, 110025 New Delhi, India. EM rizvijmi@gmail.com CR Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100:57–70 Lin JC, Scherer SW, Tougas L, Traverso G, Tsui LC, Andrulis IL, Jothy S, Park M, 1996, Detailed deletion mapping with a refined physical map of 7q31 localizes a putative tumor suppressor gene for breast cancer in the region of MET. Oncogene 13:2001–2008 Knudson AG, 1993, Antioncogenes and human cancer. Proc Natl Acad Sci USA 90:10914–10921 Marshall CJ, 1991, Tumor suppressor genes. Cell 64:313–326 Goddard AD, Solomon E, 1993, Genetic aspects of cancer. In: Harris CC, Hirschorn K, eds, Advances in human genetics. Plenum Press, New York 21: 321–376. Siebert R, Gesk S, Harder S, Plotz S, Matthiesen P, Grote W, Schlegelberger B, Jandrig B, Grasmo-Wendler U, Scherneck S, Rosenwald A, Ott G, 1998, Deletions in the long arm of chromosome 10 in lymphomas with t(14;18): A Pathogenetic Role of the Tumor Suppressor Genes PTEN/MMAC1 and MXI1? Blood 92(11):4487–4489 Steck PA, Lin H, Langford LA, Jasser SA, Koul D, Yung WK, Pershouse MA, 1999, Functional and molecular analyses of 10q deletions in human gliomas. Genes Chromosomes Cancer 24, 2):135–143 Matsuyama H, Pan Y, Yoshihiro S, Kudren D, Naito K, Bergerheim USR, Ekman P, 2003, Clinical significance of chromosome 8p, 10q, and 16q deletions in prostate cancer. Prostate 54(2):103–111 Li YL, Tian Z, Wu DY, Fu BY, Xin Y, 2005, Loss of heterozygosity on 10q23.3 and mutation of tumor suppressor gene PTEN in gastric cancer and precancerous lesions. World J Gastroenterology 11(2):285–288 Sankaranaryanan R, Buduk AM, Rajkumar R, 2001, Effective Screening Programmes for Cervical Cancer in low- and middleincome developing countries. Bull World Health Organ 79:954– 962 Ferlay J. GLOBOCAN 2002. Cancer incidence, mortality and prevalence worldwide. IARC 2004; CancerBase No.5, Version 2.0. IARC Press, Lyon. NATIONAL CANCER REGISTRY PROGRAMME, 2007, Consolidated report of hospital based cancer registries 2001–2003. ICMR, New Delhi American Cancer Society. Facts & Figures 2009. Mitra AB, Murty VVVS, Li RG, Pratap M, Luthra UK, Chaganti RSK, 1994, Allelotype analysis of cervical carcinoma. Cancer Res 54:4481–4487 Jones MH, Koi S, Fujimoto I et al, 1994, Allelotype of uterine cancer by analysis of RFLP and microsatellite polymorphisms: frequent loss of heterozygosity on chromosome arms 3p, 9q, 10q, and 17p. Genes Chromosomes Cancer 9:119–123 Miayani K, Furugen Y, Matsumoto T, Iwabuchi K, Hirose S, Kinoshita K, Fujii H, 2004, Loss of heterozygosity analysis in uterine cervical adenocarcinomas. Gynecol Oncol 94:115–120 Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP, 1998, PTEN is essential for embryonic development and tumour suppression. Nat Genet 19:348–355 Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, Miliaresis C, Rodgers L, McCombie R, Bigner SH, Giovanella BC, Ittmann M, Tycko B, Hibshoosh H, Wigler MH, Parsons R, 1997, PTEN: a putative protein tyrosine phosphatase gene mutated in human brain, breast and prostate cancer. Science 275:1943–1947 Stambolic V, Suzuki A, de la Pompa JL, Brothers GM, Mirtsos C, Sasaki T, Ruland J, 1998, Negative regulation of PKB/Aktdependent cell survival by the tumor suppressor PTEN. Cell 95:29–39 Reid AHM, Attard G, Ambroisine L, Fisher G, Kovacs G, Brewer D, Clark J, Flohr P, Edwards S, Berney DM, Foster CS, Fletcher A, Gerald WL, Moller H, Reuter VE, Scardino PT, Cuzick J, de Bono JS, Cooper CS, 2010, Molecular characterisation of ERG, ETV1 and PTEN gene loci identifies patients at low and high risk of death from prostate cancer. Br J Cancer 102:678–684 Izycka-Swieszewska E, Brzeskwiniewicz M,Wozniak A, Drozynska E, GrajkowskaW, Perek D, Balcerska A, Klepacka T, Limon J, 2010, EGFR, PIK3CA and PTEN gene status and their protein product expression in neuroblastic tumours. Folia Neuropathol 48(4):238– 245 Aneta JJ, Bozena K, Cyprian G, Urszula N, 2010, TP53 mutations in endometrial cancers: relation to PTEN gene defects. Int J Gynecol Cancer 20(2):196–202 Shabbir Alam M, Rizvi MMA, 2011, Cancer of the uterine cervix and PTEN tumor suppressor gene. Lambert Academic Publishing, Germany Kurose K, Zhou XP, Araki T, Eng C, 2000, Biallelic inactivating mutations and an occult germline mutation of PTEN in primary cervical carcinomas. Genes Chromosomes Cancer 29:166–172 Benedet JL, Bender H, Jones H III, Ngan HYS, Pecorelli S, 2000, FIGO staging classifications and clinical practice guidelines in the management of gynecologic cancers. Int J Gynecol Obstet 70:209–262 Sambrook J, Russell DW, 2001, Molecular cloning: a laboratory manual, 3rd edn. Cold Spring Harbor Lab Press, Plainview Yuping X, Lili M, Chengbo H, Jinyi L, Lei X, 2007, MSI/LOH and extron expression of the FHIT gene in gastric carcinoma. Front Med China 1(1):99–103 Preacher KJ, 2001, Calculation for the chi-square test: An interactive calculation tool for chi-square tests of goodness of fit and independence [Computer software]. Available from http://www. quantpsy.org. Hansen MF, Cavenee WK, 1987, Genetics of cancer predeposition. Cancer Res 47:5518–5527 Kim S, Domon-Dell C, Wang Q, Chung DH, Di Cristofano A, Pandolfi PP, Freund JN, Evers BM, 2002, PTEN and TNFalpha regulation of the intestinal-specific Cdx-2 homeobox gene through a PI3K, PKB/Akt, and NF-kappaB-dependent pathway. Gastroenterology 123:1163–1178 McConnachie G, Pass I, Walker SM, Downes CP, 2003, Interfacial kinetic analysis of the tumour suppressor phosphatase, PTEN: evidence for activation by anionic phospholipids. Biochem J 371(3):947–955 Brown KS, Blair D, Reid SD, Nicholson EK, Harnett MM, 2004, FcgammaRIIb-mediated negative regulation of BCR signaling is associated with the recruitment of the MAPKinasephosphatase, Pac-1, and the 3′-inositol phosphatase, PTEN. Cell Signal 16:71– 80 Cho SH, Lee CH, Ahn Y, Kim H, Kim H, Ahn CY, Yang KS, Lee SR, 2004, Redox regulation of PTEN and protein tyrosine phosphatases in H(2)O(2, mediated cell signaling. FEBS Lett 560:7–13 WishartMJ, Dixon JE, 2002, PTEN and myotubularin phosphatases: from 3-phosphoinositide dephosphorylation to disease. Trends Cell Biol 12:579–585 Mills GB, Kohn E, Lu Y, Eder A, Fang X,Wang H, Bast RC, Gray J, Jaffe R, Hortobagyi G, 2003, Linking molecular diagnostics to molecular therapeutics: targeting the PI3K pathway in breast cancer. Semin Oncol 30(5 Suppl 16):93–104 Orchiston EA, Bennett D, Leslie NR, Clarke RG, Winward L, Downes CP, Safrany ST, 2004, PTEN M-CBR3, a versatile and selective regulator of inositol 1,3,4,5,6-pentakisphosphate, Ins, 1,3, 4,5,6, P5). Evidence for Ins, 1,3,4,5,6)P5 as a proliferative signal. J Biol Chem 279:1116–1122 Zhao H, Dupont J, Yakar S, Karas M, LeRoith D, 2004, PTEN inhibits cell proliferation and induces apoptosis by downregulating cell surface IGF-IR expression in prostate cancer cells. Oncogene 23:786–794 Konopka B, Janiec-Jankowska A, Paszko Z, Goluda M, 2004, The coexistence of ERBB2, INT2, and CMYC oncogene amplifications and PTEN gene mutations in endometrial carcinoma. J Cancer Res Clin Oncol 130:114–121 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 309 EP 313 DI 10.1007/s12253-011-9446-z PG 5 ER PT J AU Jia, Y Liu, M Huang, W Wang, Z He, Y Wu, J Ren, Sh Ju, Y Geng, R Li, Z AF Jia, Yitao Liu, Min Huang, Wangang Wang, Zhenbao He, Yutong Wu, Jianhua Ren, Shuguang Ju, Yingchao Geng, Ruichao Li, Zhongxin TI Recombinant Human Endostatin Endostar Inhibits Tumor Growth and Metastasis in a Mouse Xenograft Model of Colon Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Recombinant human endostatin; Colorectal cancer; Metastasis; Angiogenesis; Lymphangiogenesis ID Recombinant human endostatin; Colorectal cancer; Metastasis; Angiogenesis; Lymphangiogenesis AB To investigate the effects of recombinant human endostatin Endostar on metastasis and angiogenesis and lymphangiogenesis of colorectal cancer cells in a mouse xenograft model. Colon cancer cells SW620 were injected subcutaneously into the left hind flank of nude mice to establish mouse xenograft models. The mice were treated with normal saline or Endostar subcutaneously every other day. The growth and lymph node metastasis of tumor cells, angiogenesis and lymphangiogenesis in tumor tissue were detected.Apoptosis and cell cycle distribution were studied by flow cytometry. The expression of VEGF-A, -C, or -D in SW620 cells was determined by immunoblotting assays. Endostar inhibited tumor growth and the rate of lymph node metastasis (P<0.01). The density of blood vessels in or around the tumor area was 12.27±1.21 and 22.25±2.69 per field in Endostar-treated mice and controls (P<0.05), respectively. Endostar also decreased the density of lymphatic vessels in tumor tissues (7.84±0.81 vs. 13.83±1.08, P<0.05). Endostar suppresses angiogenesis and lymphangiogenesis in the lymph nodes with metastases, simultaneously. The expression of VEGF-A, -C and -D in SW620 cells treated with Endostar was substantially lower than that of controls. Endostar inhibited growth and lymph node metastasis of colon cancer cells by inhibiting angiogenesis and lymphangiogenesis in a mouse xenograft model of colon cancer. C1 [Jia, Yitao] Hebei General Hospital, Department of OncologyShijiazhuang, Hebei, China. [Liu, Min] Hebei General Hospital, Geriatrics Key LaboratoryShijiazhuang, Hebei, China. [Huang, Wangang] Hebei Medical University, the Forth Hospital, Second Department of SurgeryShijiazhuang, Hebei, China. [Wang, Zhenbao] Hebei Medical University, the Forth Hospital, Second Department of SurgeryShijiazhuang, Hebei, China. [He, Yutong] Cancer Institute of Hebei ProvinceShijiazhuang, Hebei, China. [Wu, Jianhua] Hebei Medical University, the Forth Hospital, Centre of Animal ExperimentShijiazhuang, Hebei, China. [Ren, Shuguang] Hebei Medical University, the Forth Hospital, Centre of Animal ExperimentShijiazhuang, Hebei, China. [Ju, Yingchao] Hebei Medical University, the Forth Hospital, Centre of Animal ExperimentShijiazhuang, Hebei, China. [Geng, Ruichao] Hebei Medical University, the Forth Hospital, Second Department of SurgeryShijiazhuang, Hebei, China. [Li, Zhongxin] Hebei Medical University, the Forth Hospital, Second Department of SurgeryShijiazhuang, Hebei, China. RP Jia, Y (reprint author), Hebei General Hospital, Department of Oncology, Shijiazhuang, China. EM jiayitao1970@yahoo.cn CR Center MM, Jemal A, Ward E, 2009, International trends in colorectal cancer incidence rates. Cancer Epidemiol Biomarkers Prev 6:1688–1694 Becouarn Y, Rougier P, 1998, Clinical efficacy of oxaliplatin monotherapy: phase II trials in advanced colorectal cancer. Semin Oncol 25(2 Suppl 5):23–31 Ng M, Roy-Chowdhury S, Lum SS, Morgan JW, Wong JH, 2009, The impact of the ratio of positive to total lymph nodes examined and outcome in colorectal cancer. Am Surg 75:873–876 Jemal A, Clegg LX, Ward E, Ries LA, Wu X, Jamison PM, Wingo PA, Howe HL, Anderson RN, Edwards BK, 2004, Annual report to the nation on the status of cancer, 1975–2001, with a special feature regarding survival. Cancer 101:3–27 Folkman J, 2002, Role of angiogenesis in tumor growth and metastasis. Semin Oncol 29(6 Suppl 16):15–18 Byrne AM, Bouchier-Hayes DJ, Harmey JH, 2005, Angiogenic and cell survival functions of vascular endothelial growth factor, VEGF). J Cell Mol Med 9:777–794 Okines A, Puerto OD, Cunningham D, Chau I, Van Cutsem E, Saltz L, Cassidy J, 2009, Surgery with curative-intent in patients treated with first-line chemotherapy plus bevacizumab for metastatic colorectal cancer First BEAT and the randomised phase-III NO16966 trial. Br J Cancer 101:1033–1038 Hsu JY, Wakelee HA, 2009, Monoclonal antibodies targeting vascular endothelial growth factor: current status and future challenges in cancer therapy. BioDrugs 23:289–304 Punt CJ, Tol J, 2009, More is less-combining targeted therapies in metastatic colorectal cancer. Nat Rev Clin Oncol 6:731–733 Duff SE, Jeziorska M, Kumar S, Haboubi N, Sherlock D, O’Dwyer ST, Jayson GC, 2007, Lymphatic vessel density, microvessel density and lymphangiogenic growth factor expression in colorectal cancer. Colorectal Dis 9:793–800 Royston D, Jackson DG, 2009, Mechanisms of lymphatic metastasis in human colorectal adenocarcinoma. J Pathol 217:608–619 Sundlisaeter E, Dicko A, Sakariassen PO, Sondenaa K, Enger PO, Bjerkvig R, 2007, Lymphangiogenesis in colorectal cancer-prognostic and therapeutic aspects. Int J Cancer 121:1401–1409 Onogawa S, Kitadai Y, Tanaka S, Kuwai T, Kimura S, Chayama K, 2004, Expression of VEGF-C and VEGF-D at the invasive edge correlates with lymph node metastasis and prognosis of patients with colorectal carcinoma. Cancer Sci 95:32–39 Duff SE, Li C, Jeziorska M, Kumar S, Saunders MP, Sherlock D, O’Dwyer ST, Jayson GC, 2003, Vascular endothelial growth factors C and D and lymphangiogenesis in gastrointestinal tract malignancy. Br J Cancer 89:426–430 Achen MG, Mann GB, Stacker SA, 2006, Targeting lymphangiogenesis to prevent tumour metastasis. Br J Cancer 94:1355–1360 Wissmann C, Detmar M, 2006, Pathways targeting tumor lymphangiogenesis. Clin Cancer Res 12:6865–6868 Brideau G, Makinen MJ, Elamaa H, Tu H, Nilsson G, Alitalo K, Pihlajaniemi T, Heljasvaara R, 2007, Endostatin overexpression inhibits lymphangiogenesis and lymph node metastasis in mice. Cancer Res 67:11528–11535 Fukumoto S, MorifujiM, Katakura Y, OhishiM, Nakamura S, 2005, Endostatin inhibits lymph node metastasis by a down-regulation of the vascular endothelial growth factor C expression in tumor cells. Clin Exp Metastasis 22:31–38 Zhuo W, Luo C, Wang X, Song X, Fu Y, Luo Y, 2010, Endostatin inhibits tumour lymphangiogenesis and lymphatic metastasis via cell surface nucleolin on lymphangiogenic endothelial cells. J Pathol 222:249–260 Feldman AL, Alexander HR Jr, Bartlett DL, Kranda KC, Miller MS, Costouros NG, Choyke PL, Libutti SK, 2001, A prospective analysis of plasma endostatin levels in colorectal cancer patients with liver metastases. Ann Surg Oncol 8:741–745 Chen W, Fu J, Liu Q, Ruan C, Xiao S, 2003, Retroviral endostatin gene transfer inhibits human colon cancer cell growth in vivo. Chin Med J, Engl, 116:1582–1584 te Velde EA, Reijerkerk A, Brandsma D, Vogten JM, Wu Y, Kranenburg O, Voest EE, Gebbink M, Borel Rinkes IH, 2005, Early endostatin treatment inhibits metastatic seeding of murine colorectal cancer cells in the liver and their adhesion to endothelial cells. Br J Cancer 92:729–735 Yang L, Wang JW, Sun Y, Zhu YZ, Liu XQ, Li WL, 2006, Randomized phase II trial on escalated doses of Rh-endostatin, YH-16, for advanced non-small cell lung cancer. Zhonghua Zhong Liu Za Zhi 28:138–141 Zhang DW, Li HL, Yao Q, Yang WL, Wang HL, Zhai DX, Zhou ZH, 2010, The synergistic effect of recombinant human endostatin, YH-16, combined with oxaliplatin on human colorectal carcinoma. J Int Med Res 38:111–126 La Vecchia C, Bosetti C, Lucchini F, Bertuccio P, Negri E, Boyle P, Levi F, 2010, Cancer mortality in Europe, 2000–2004, and an overview of trends since 1975. Ann Oncol 21:1323–1360 Jung KW, Won YJ, Park S, Kong HJ, Sung J, Shin HR, Park EC, Lee JS, 2009, Cancer statistics in Korea: incidence, mortality and survival in 2005. J Korean Med Sci 24:995–1003 Fujita Y, Abe R, Shimizu H, 2008, Clinical approaches toward tumor angiogenesis: past, present and future. Curr Pharm Des 14:3820–3834 Ferrara N, Kerbel RS, 2005, Angiogenesis as a therapeutic target. Nature 438:967–974 Casanovas O, Hicklin DJ, Bergers G, Hanahan D, 2005, Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 8:299–309 Abdollahi A, Hahnfeldt P, Maercker C, Grone HJ, Debus J, Ansorge W, Folkman J, Hlatky L, Huber PE, 2004, Endostatin’s antiangiogenic signaling network. Mol Cell 13:649– 663 Akbulut H, Altuntas F, Akbulut KG, Ozturk G, Cindoruk M, Unal E, Icli F, 2002, Prognostic role of serum vascular endothelial growth factor, basic fibroblast growth factor and nitric oxide in patients with colorectal carcinoma. Cytokine 20:184–190 Ling Y, Yang Y, Lu N, You QD, Wang S, Gao Y, 2007, Endostar, a novel recombinant human endostatin, exerts antiangiogenic effect via blocking VEGF-induced tyrosine phosphorylation of KDR/Flk-1 of endothelial cells. Biochem Biophys Res Commun 361:79–84 Jenkins DE, Yu SF, Horning YS, Purchio T, Contag PR, 2003, In vivo monitoring of tumor relapse and metastasis using bioluminescent PC-3M-luc-C6 cells in murine models of human prostate cancer. Clin Exp Metastasis 20:745–756 Lee HY, Choi HJ, Park KJ, Shin JS, Kwon HC, Roh MS, Kim C, 2007, Prognostic significance of metastatic lymph node ratio in node-positive colon carcinoma. Ann Surg Oncol 14:1712–1717 Bock F, Onderka J, Dietrich T, Bachmann B, Kruse FE, Paschke M, Zahn G, Cursiefen C, 2007, Role of lymphangiogenesis in cancer. J Clin Oncol 25:4298–4307 Thelen A, Scholz A, Benckert C, von Marschall Z, Schroder M, Wiedenmann B, Neuhaus P, Rosewicz S, Jonas S, 2008, VEGF-D promotes tumor growth and lymphatic spread in a mouse model of hepatocellular carcinoma. Int J Cancer 122:2471–2481 Kopfstein L, Veikkola T, Djonov VG, Baeriswyl V, Schomber T, Strittmatter K, Stacker SA, Achen MG, Alitalo K, Christofori G, 2007, Distinct roles of vascular endothelial growth factor-D in lymphangiogenesis and metastasis. Am J Pathol 170:1348–1361 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 315 EP 323 DI 10.1007/s12253-011-9447-y PG 9 ER PT J AU Bouanene, H Saibi, W Mokni, M Sriha, B Ben Fatma, L Ben Limem, H Ben Ahmed, S Gargouri, A Miled, A AF Bouanene, Houda Saibi, Walid Mokni, Moncef Sriha, Badreddine Ben Fatma, Leila Ben Limem, Halima Ben Ahmed, Slim Gargouri, Ali Miled, Abdelhedi TI Biochemical and Morphological Differences Between CA125 Isolated from Healthy Women and Patients with Epithelial Ovarian Cancer from Tunisian Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CA125; Ovarian cancer; Tumor marker; Biochemical characterization; MUC16 ID CA125; Ovarian cancer; Tumor marker; Biochemical characterization; MUC16 AB Analysis of the structure of CA125 is essential for determining the physiological role of this significant tumor antigen. The objectives of this study were: (1) to identify the characteristics of the CA125 isolated from healthy and patient women with epithelial ovarian cancer; and (2) to determine the ferning structure of this antigen. The cancer-derived CA125 antigen (cCA125) purified by gel filtration and affinity chromatography (Concanavalin A) was run on SDS-PAGE and examined using light microscopy and compared with healthy-derived CA125 antigen (hCA125). Both purified antigen cCA125 and hCA125 showed a high molecular mass (> 2,000 kDa) with high mannose glycans. The ferning patterns related to cCA125 and hCA125 revealed distinct differences in the patterns of arborescence. The ferning morphology of cCA125 antigen was denser than that of hCA125 antigen making an obvious difference between cCA125 and hCA125, with respect to length, branching and distribution of crystals. The current study provides the first evidence for a potential functional link between CA125 and its structure which, in the light of a comparison between cCA125 and hCA125, might proof to be of significant biomedical importance in the future. C1 [Bouanene, Houda] CHU Farhat Hached, Laboratory of Biochemistry, 4000 Sousse, Tunisia. [Saibi, Walid] University of Sfax, Centre de Biotechnologie de Sfax, Laboratory of Biomass Valorization and Protein Production in Eucaryotes, B.P. 1177, 3038 Sfax, Tunisia. [Mokni, Moncef] Farhat Hached Hospital, Department of PathologySousse, Tunisia. [Sriha, Badreddine] Farhat Hached Hospital, Department of PathologySousse, Tunisia. [Ben Fatma, Leila] CHU Farhat Hached, Service of OncologySousse, Tunisia. [Ben Limem, Halima] CHU Farhat Hached, Laboratory of Biochemistry, 4000 Sousse, Tunisia. [Ben Ahmed, Slim] CHU Farhat Hached, Service of OncologySousse, Tunisia. [Gargouri, Ali] University of Sfax, Centre de Biotechnologie de Sfax, Laboratory of Biomass Valorization and Protein Production in Eucaryotes, B.P. 1177, 3038 Sfax, Tunisia. [Miled, Abdelhedi] CHU Farhat Hached, Laboratory of Biochemistry, 4000 Sousse, Tunisia. RP Bouanene, H (reprint author), CHU Farhat Hached, Laboratory of Biochemistry, 4000 Sousse, Tunisia. EM bouanenehouda2@yahoo.fr CR Yin BWT, Lloyd KO, 2001, Molecular cloning of the CA125 ovarian cancer antigen. Identification as a new mucin, MUC16. J Biol Chem 276:27371–27375 JrRC B, Feeney M, Lazarus H et al, 1981, Reactivity of a monoclonal antibody with a human ovarian carcinoma. J Clin Invest 68:1311–1337 Nustad K, Onsrud M, Jansson B et al, 1998, Warren CA 125— epitopes and molecular size. Int J Biol Markers 13:196–199 Hanisch FG, Uhlenbruck G, Dienst C et al, 1985, CA125 and CA 19–9: two cancer-associated sialylsaccharide antigens on a mucus glycoprotein from human milk. Eur J Biochem 149:323–330 Wong NK, Easton RL, Panico M et al, 2003, Characterization of the oligosaccharides associated with the human ovarian tumor marker CA125. J Biol Chem 278:28619–28634 Bouanene H, Miled A, 2010, Conflicting views on the molecular structure of the Cancer Antigen CA125/MUC16. Dis Markers 28:1–10 Spiro RG, 1972, Study of the carbohydrates of glycoproteins. Method Enzymol 28:3–43 Bradford MM, 1976, A rapid and sensitive method for the quantitation of microgram quantities utilizing the principal of protein dye binding. Anal Biochem 72:248–254 Laemmli UK, 1970, Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227:680–685 Bouanene H, Harrabi I, Ferchichi S et al, 2007, Factors predictive of elevated serum CA125 levels in patients with epithelial ovarian cancer. B Cancer 94:E18–E22 Bouanene H, Hadj Kacem H, Ben Fatma L et al, 2010, Polymorphisms in the MUC16 Gene: Potential Implication in Epithelial Ovarian Cancer. Pathol Oncol Res, Epub ahead of print) Lloyd KO, Yin BWT, Kudryashov V, 1997, Isolation and characterization of ovarian cancer antigen CA 125 using a new monoclonal antibody VK-8: identification as a mucin- type molecule. Int J Cancer 71:842–850 Yu YH, Schlossman DM, Harrison CL et al, 1991, Coexpression of different antigenic markers on moieties that bear CA 125 determinants. Cancer Res 51:468–475 Davis HM, JrVR Z, JrRC B et al, 1986, Characterization of the CA 125 antigen associated with human epithelial ovarian carcinomas. Cancer Res 46:6143–6148 Milutinovic B, Jankovic M, 2007, Analysis of the protein and glycan parts of CA125 antigen from human amniotic fluid. Arch Biol Sci 59:97–103 Kobata A, Endo T, 2003, Immobilized lectin columns: useful tools for the fractionation and structural analysis of oligosacchraides. J Chromatogr 597:111–122 Goodell CAR, Belisle JA, Gubbels JAA et al, 2009, Characterization of the tumor marker muc16, ca125, expressed by murine ovarian tumor cell lines and identification of a panel of crossreactive monoclonal antibodies. J Ovar Res 2:8 Jankovic MM, Milutinovic BS, 2008, Glycoforms of CA125 antigen as a possible cancer marker. Cancer Biomark 4:35– 42 Gubbels JAA, Belisle J, Onda M et al, 2006, Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Molecular Cancer 5:50 Kaneko O, Gong L, Zhang J, 2009, A binding domain on mesothelin for CA125/MUC16. J Biol Chem 284:3739–3749 Jankovic MM, Milutinovic BS, 2007, Pregnancy-associated CA125 antigen as mucin: evaluation of ferning morphology. Mol Hum Reprod 13:405–408 Jankovic MM, Tapuskovic BT, 2005, Molecular forms and microheterogeneity of the pregnancy-associated CA125 antigen. Hum Rep 20:2632–2638 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 325 EP 330 DI 10.1007/s12253-011-9448-x PG 6 ER PT J AU Maciejczyk, A Jagoda, E Wysocka, T Matkowski, R Gyorffy, B Lage, H Surowiak, P AF Maciejczyk, Adam Jagoda, Ewa Wysocka, Teresa Matkowski, Rafal Gyorffy, Balazs Lage, Hermann Surowiak, Pawel TI ABCC2 (MRP2, cMOAT) Localized in the Nuclear Envelope of Breast Carcinoma Cells Correlates with Poor Clinical Outcome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ABCC2; Breast cancer; Immunohistochemistry; Prognosis ID ABCC2; Breast cancer; Immunohistochemistry; Prognosis AB Nuclear expression of ABCC2 can be specific for lower differentiated cells and stem cells. The study aimed at examination of ABCC2 expression in breast cancers. The immunohistochemical analyses were performed on 70 samples of breast cancer.We have also studied prognostic value of the ABCC2 mRNA expression using the KM plotter which assessed the effect of 22,277 genes on survival in 1809 breast cancer patients. Immunohistochemical studies demonstrated that ABCC2 expression may be manifested in nuclear envelope of neoplastic cells (ABCC2n) as well as in their cell membrane and cytoplasm (ABCC2c). The univariate and multivariate analyses showed that higher expression of ABCC2n and ABCC2c was typical for cases of a shorter overall survival time. Higher ABBC2n expression was also typical for cases of a shorter disease-free survival and a shorter progression-free time. The KM plotter analysis of the prognostic value of ABCC2 mRNA expression showed that elevated ABCC2 expression was specific for cases of a shorter relapse-free survival only in the estrogen receptor-negative subgroup. The study demonstrated hat breast cancers manifest ABCC2 expression and that it is linked to a less favourable prognosis. Our results suggested that immunohistochemical tests represent a reliable way to detect prognostic value of ABCC2 expression, allowing to demonstrate differences related to subcellular localization of the protein. Cases with nuclear expression of ABCC2 manifested a more aggressive clinical course, which might reflect a less advanced differentiation of neplastic cells, resistance to the applied cytostatic drugs and tamoxifen. C1 [Maciejczyk, Adam] Lower Silesian Oncology Center, pl. Hirszfelda 12, 53-413 Wroclaw, Poland. [Jagoda, Ewa] Wroclaw Medical University, Department of Human Morphology and Embryology, ul. Chalubinskiego 6a, 50-356 Wroclaw, Poland. [Wysocka, Teresa] Wroclaw Medical University, Department of Human Morphology and Embryology, ul. Chalubinskiego 6a, 50-356 Wroclaw, Poland. [Matkowski, Rafal] Wroclaw Medical University, Department of OncologyWroclaw, Poland. [Gyorffy, Balazs] Hungarian Academy of Sciences - Semmelweis University, 1st Dept. of Pediatrics, Research Laboratory for Pediatrics and Nephrology, Bokay u. 53-54, 1083 Budapest, Hungary. [Lage, Hermann] Charite University Hospital, Institute of Pathology, Chariteplatz 1, 10117 Berlin, Germany. [Surowiak, Pawel] Lower Silesian Oncology Center, pl. Hirszfelda 12, 53-413 Wroclaw, Poland. RP Surowiak, P (reprint author), Lower Silesian Oncology Center, 53-413 Wroclaw, Poland. CR Steward BW, Kleihues P, 2003, World cancer report. IARC Press, Lyon Tabar L, Yen MF, Vitak B et al, 2003, Mammography service screening and mortality in breast cancer patients: 20-year followup before and after introduction of screening. Lancet 361:1405– 1410 Gottesman MM, Fojo T, Bates SE, 2002, Multidrug resistance in cancer: role of ATP-dependent transporters. Nature 2:48–58 Lage H, 2003, ABC-transporters: implications on drug resistance from microorganisms to human cancers. Int J Antimicrob Agents 22:188–199 Wada M, Toh S, Taniguchi K et al, 1998, Mutations in the canalicular multispecific organic anion transporter, cMOAT, gene, a novel ABC transporter, in patients with hyperbilirubinemia II/ Dubin-Johnson syndrome. Hum Mol Genet 7:203–207 Taniguchi K, Wada M, Kohno K et al, 1996, A human canalicular multispecific organic anion transporter, cMOAT, gene is overexpressed in cisplatin-resistant human cancer cell lines with decreased drug accumulation. Cancer Res 56:4124–4129 Materna V, Stege A, Surowiak P et al, 2006, RNA interferencetriggered reversal of ABCC2-dependent cisplatin resistance in human cancer cells. Biochem Biophys Res Com 348:153–157 Kowalski P, Surowiak P, Lage H, 2005, Reversal of drug-resistant phenotypes by an autocatalytic multitarget multiribozyme directed against the transcripts of the ABC transporters MDR1/P-gp, MRP2, and BCRP. Mol Ther 11:508–522 Surowiak P, Materna V, Kaplenko I et al, 2006, ABCC2, MRP2, cMOAT, can be localized in the nuclear membrane of ovarian carcinomas and correlates with resistance to cisplatin and clinical outcome. Clin Cancer Res 12(23):7149–7158 Surowiak P, Materna V, Maciejczyk A et al, 2007, Nuclear metallothionein expression correlates with cisplatin resistance of ovarian cancer cells and poor clinical outcome. Virchows Arch 450(3):279–285 Materna V, Surowiak P, Kaplenko I et al, 2007, Taxol-resistanceassociated gene-3, TRAG-3/CSAG2, expression is predictive for clinical outcome in ovarian carcinoma patients. Virchows Arch 450(2):187–194 Sobin LH, Wittekind C, 2002, TNM classification of malignant tumors. AJCC, Wiley-Liss, NewYork Remmele W, Stegner HE, 1987, Recommendation for uniform definition of an immunoreactive score, IRS, for immunohistochemical estrogen receptor detection, ER-ICA, in breast cancer tissue, Article in German). Pathologe 8:138–140 Surowiak P, Materna V, Matkowski R et al, 2005, Relationship between cyclooxygenase 2 and MDR1/P-glycoprotein expressions in invasive breast cancers and their prognostic significance. Breast Cancer Res 7:862–870 http://cubic.bioc.columbia.edu/cgi/var/nair/resonline.pl Gyorffy B, Lanczky A, Eklund AC et al, 2010, An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1809 patients. Breast Cancer Res Treat 123(3):725–731 Statistica PL, 2007,, Statsoft, Poland). Szczuraszek K, Materna V, Halon A et al, 2009, Positive correlation between cyclooxygenase-2 and ABC-transporter expression in non-Hodgkin’s lymphomas. Oncol Rep 22(6):1315– 1323 Dean M, Fojo T, Bates S, 2005, Tumour stem cells and drug resistance. Nat Rev Cancer 5:275–284 Donnenberg VS, Donnenberg AD, 2005, Multiple drug resistance in cancer revisited: the cancer stem cell hypothesis. J Clin Pharmacol 45:872–877 Kiyotani K, Mushiroda T, Imamura CK et al, 2010, Significant effect of polymorphisms in CYP2D6 and ABCC2 on clinical outcomes of adjuvant tamoxifen therapy for breast cancer patients. J Clin Oncol 28(8):1287–1293 Qiu R, Kalhorn TF, Slattery JT, 2004, ABCC2-mediated biliary transport of 4-glutathionylcyclophosphamide and its contribution to elimination of 4-hydroxycyclophosphamide in rat. J Pharmacol Exp Ther 308(3):1204–1212 Rau T, Erney B, Gores R et al, 2006, High-dose methotrexate in pediatric acute lymphoblastic leukemia: impact of ABCC2 polymorphisms on plasma concentrations. Clin Pharmacol Ther 80, 5):468–476 Vlaming ML, van Esch A, Pala Z et al, 2009, Abcc2, Mrp2), Abcc3, Mrp3), and Abcg2, Bcrp1, are the main determinants for rapid elimination of methotrexate and its toxic metabolite 7-hydroxymethotrexate in vivo. Mol Cancer Ther 8(12):3350– 3359 Hulot JS, Villard E, Maguy A et al, 2005, A mutation in the drug transporter gene ABCC2 associated with impaired methotrexate elimination. Pharmacogenet Genomics 15(5):277–285 Hinoshita E, Uchiumi T, Taguchi K et al, 2000, Increased expression of an ATP-binding cassette superfamily transporter, multidrug resistance protein 2, in human colorectal carcinomas. Clin Cancer Res 6(6):2401–2407 Fujita K, Nagashima F, YamamotoWet al, 2008, Association ofATPbinding cassette, sub-family C, number 2, ABCC2, genotype with pharmacokinetics of irinotecan in Japanese patients with metastatic colorectal cancer treated with irinotecan plus infusional 5-fluorouracil/ leucovorin, FOLFIRI). Biol Pharm Bull 31(11):2137–2142 O’Brian CA, Kreso A, Dick JE, 2009, Cancer stem cells in solid tumors: an overview. Semin Radiat Oncol 19(2):71–77 Materna V, Stege A, Surowiak P et al, 2006, RNA interferencetriggered reversal of ABCC2-dependent cisplatin resistance in human cancer cells. Biochem Biophys Res Com 348(1):153–157 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 331 EP 342 DI 10.1007/s12253-011-9449-9 PG 12 ER PT J AU Yan, M Huang, HY Wang, T Wan, Y Cui, ShD Liu, Zz Fan, QX AF Yan, Min Huang, Hong-Yan Wang, Ting Wan, Yi Cui, Shu-De Liu, Zhen-zhen Fan, Qing-Xia TI Dysregulated Expression of Dicer and Drosha in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; microRNAs; Dicer; Drosha ID Breast cancer; microRNAs; Dicer; Drosha AB Large-scale profiling approaches have revealed global down-regulation of microRNAs (miRNAs) in several human cancer types including breast cancer. Altered expression of Dicer and Drosha, two key enzymes in the miRNA maturation, is believed to be one of the most important mechanisms. By using quantitative real-time RTPCR (QT-PCR), we examined the expression of Dicer and Drosha in 49 pairs of matched human breast cancer tissues. Decreased expression was observed in 53.1% (Dicer), 51.9% (Drosha) and 75.5% (Dicer plus Drosha) breast cancer tissues. In conclusion, the decreased expression of Dicer and Drosha may play a role in down-regulation of miRNAs in breast cancer. C1 [Yan, Min] First Affiliated Hospital of Zhengzhou University, Department of Oncology, No. 1, East Jianshe Rd, 450003 Zhengzhou, China. [Huang, Hong-Yan] Affiliated Hospital of Academy of Military Medical Sciences, Department of Breast CancerBeijing, China. [Wang, Ting] Fourth Military Medical University, Xijing Hospital, Department of Vascular and Endocrine SurgeryXi’an, China. [Wan, Yi] Fourth Military Medical University, Department of StatisticsXi’an, China. [Cui, Shu-De] Henan Cancer Hospital, Breast Cancer CenterHenan, China. [Liu, Zhen-zhen] Henan Cancer Hospital, Breast Cancer CenterHenan, China. [Fan, Qing-Xia] First Affiliated Hospital of Zhengzhou University, Department of Oncology, No. 1, East Jianshe Rd, 450003 Zhengzhou, China. RP Fan, QX (reprint author), First Affiliated Hospital of Zhengzhou University, Department of Oncology, 450003 Zhengzhou, China. EM fanqx2011@yeah.net CR Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60(5):277–300 Xu YL, Sun Q, 2010, Headway in resistance to endocrine therapy in breast cancer. J Thorac Dis 2(3):171–177 Lv YG, Yu F, Yao Q, Chen JH, Wang L, 2010, The role of survivin in diagnosis, prognosis and treatment of breast cancer. J Thorac Dis 2(2):100–110 Papademetriou K, Ardavanis A, Kountourakis P, 2010, Neoadjuvant therapy for locally advanced breast cancer: focus on chemotherapy and biological targeted treatments’ armamentarium. J Thorac Dis 2(3):160–170 Erson AE, Petty EM, 2009, miRNAs and cancer: new research developments and potential clinical applications. Cancer Biol Ther 8(24):2317–2322 Yu Z, Baserga R, Chen L, Wang C, Lisanti MP, Pestell RG, 2010, microRNA, cell cycle, and human breast cancer. Am J Pathol 176, 3):1058–1064 Calin GA, Croce CM, 2006, MicroRNA signatures in human cancers. Nat Rev Cancer 6(11):857–866 Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, Menard S, Palazzo JP, Rosenberg A, Musiani P, Volinia S, Nenci I, Calin GA, Querzoli P, Negrini M, Croce CM, 2005, MicroRNA gene expression deregulation in human breast cancer. Cancer Res 65, 16):7065–7070 Blenkiron C, Goldstein LD, Thorne NP, Spiteri I, Chin SF, Dunning MJ, Barbosa-Morais NL, Teschendorff AE, Green AR, Ellis IO, Tavare S, Caldas C, Miska EA, 2007, MicroRNA expression profiling of human breast cancer identifies new markers of tumor subtype. Genome Biol 8(10):R214 Newman MA, Hammond SM, 2010, Emerging paradigms of regulated microRNA processing. Genes Dev 24(11):1086–1092 Veeck J, Esteller M, 2010, Breast cancer epigenetics: from DNA methylation to microRNAs. J Mammary Gland Biol Neoplasia 15, 1):5–17 Sun Q, Wang Y, Zhang Y, Liu F, Cheng X, Hou N, Zhao X, Yang X, 2007, Expression profiling reveals dysregulation of cellular cytoskeletal genes in HBx-induced hepatocarcinogenesis. Cancer Biol Ther 6(5):668–674 Sun Q, Zhang Y, Yang G, Chen X, Zhang Y, Cao G, Wang J, Sun Y, Zhang P, Fan M, Shao N, Yang X, 2008, Transforming growth factor-beta-regulated miR-24 promotes skeletal muscle differentiation. Nucleic Acids Res 36(8):2690–2699 Lewis BP, Burge CB, Bartel DP, 2005, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120(1):15–20 Merritt WM, Lin YG, Han LY, Kamat AA, Spannuth WA, Schmandt R, Urbauer D, Pennacchio LA, Cheng JF, Nick AM, Deavers MT, Mourad-Zeidan A, Wang H, Mueller P, Lenburg ME, Gray JW, Mok S, Birrer MJ, Lopez-Berestein G, Coleman RL, Bar-Eli M, Sood AK, 2008, Dicer, Drosha, and outcomes in patients with ovarian cancer. N Engl J Med 359(25):2641–2650 Sand M, Gambichler T, Skrygan M, Sand D, Scola N, Altmeyer P, Bechara FG, 2010, Expression levels of the microRNA processing enzymes Drosha and dicer in epithelial skin cancer. Cancer Invest 28(6):649–653 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 343 EP 348 DI 10.1007/s12253-011-9450-3 PG 6 ER PT J AU He, Hch Chen, Jh Chen, Xb Qin, Gq Cai, Ch Liang, Yx Han, Zd Dai, Qsh Chen, Yr Zeng, Gh Zhu, Jg Jiang, Fn Zhong, Wd AF He, Hui-chan Chen, Jia-hong Chen, Xi-bin Qin, Guo-qiang Cai, Chao Liang, Yu-xiang Han, Zhao-dong Dai, Qi-shan Chen, Yan-ru Zeng, Guo-hua Zhu, Jian-guo Jiang, Fu-neng Zhong, Wei-de TI Expression of Hedgehog Pathway Components is Associated with Bladder Cancer Progression and Clinical Outcome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hedgehog signaling pathway; Sonic hedgehog; Patched; Gli1; Bladder cancer; Prognosis ID Hedgehog signaling pathway; Sonic hedgehog; Patched; Gli1; Bladder cancer; Prognosis AB Hedgehog (Hh) pathway has been implicated in the tumorigenesis of a large number of human tumors. But its effects on the progression and prognosis of bladder cancer remain poorly understood. The aim of this study was to investigate expression patterns of Hh pathway components in bladder cancer and to elucidate their prognostic values in this tumor. The expression of sonic hedgehog (Shh), its receptor Patched (Ptch1), and downstream transcription factor Gli1 in 118 specimens of bladder cancer and 30 specimens of adjacent normal bladder tissue was determined by immunohistochemistry. Statistical analyses were applied to test the relationship between the expression of these three proteins and clinicopathologic features and prognosis. Immunohistochemical staining results showed the localizations of Shh and Ptch1 proteins to be mainly located in the cytoplasm of bladder cancer cells, whereas Gli1 was mainly localized in the nuclear of tumor cells. Additionally, positive expression of Shh, Ptch1 and Gli1 proteins was correlated with pathological stage (P=0.006, 0.006 and 0.008, respectively), venous invasion (P=0.01, 0.01 and 0.012, respectively) and lymph node metastasis (P=0.009, 0.01 and 0.013, respectively), but not with other factors including age, gender, tumor grade and recurrence of superficial cancer. Moreover, patients with positive expression of Shh, Ptch1 and Gli1 proteins respectively showed poorer disease-free (P=0.002, 0.002 and 0.001, respectively) and overall survival (all P<0.001) than those with negative expression of these three proteins. Univariate and multivariate analysis of prognostic factors in bladder cancer patients indicated that the expression patterns of Shh, Ptch1 and Gli1 proteins were independent unfavorable prognostic factors (all P<0.001). This is the first report describing about the correlation between Hh pathway and the prognosis of bladder cancer. Expression of Shh, Ptch1 and Gli1 proteins was greater in bladder cancers than in the adjacent normal tissues. The examination of their expression is potentially valuable in prognostic evaluation of bladder cancer. C1 [He, Hui-chan] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Chen, Jia-hong] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Chen, Xi-bin] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Qin, Guo-qiang] Southern Medical University, 510515 Guangzhou, China. [Cai, Chao] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Liang, Yu-xiang] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Han, Zhao-dong] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Dai, Qi-shan] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Chen, Yan-ru] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Zeng, Guo-hua] Guangzhou Medical College, First Affiliated Hospital of Guangzhou Medical College, Urology Key Laboratory of Guangdong Province, 510230 Guangzhou, China. [Zhu, Jian-guo] Guizhou Provincial People’s Hospital, Department of Urology, 550002 Guizhou, China. [Jiang, Fu-neng] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. [Zhong, Wei-de] Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. RP Zhong, Wd (reprint author), Guangzhou Medical College, Guangzhou First Municipal People’s Hospital, Department of Urology, 510180 Guangzhou, China. EM wdezhong@21cn.com CR Kramer MW, Escudero DO, Lokeshwar SD et al, 2011, Association of hyaluronic acid family members, HAS1, HAS2, and HYAL-1, with bladder cancer diagnosis and prognosis. Cancer 117:1197–209 Lei Y, Yan S, Ming-De L et al, 2011, Prognostic significance of Aurora-A expression in human bladder cancer. Acta Histochem 113:514–518 Canesin G, Gonzalez-Peramato P, Palou J et al, 2010, Galectin-3 expression is associated with bladder cancer progression and clinical outcome. Tumour Biol 31:277–285 Pasca di Magliano M, Hebrok M, 2003, Hedgehog signalling in cancer formation and maintenance. Nat Rev Cancer 3:903– 911 Kameda C, Tanaka H, Yamasaki A, 2009, The Hedgehog pathway is a possible therapeutic target for patients with estrogen receptornegative breast cancer. Anticancer Res 29:871–879 Lauth M, Toftgrd R, 2007, The Hedgehog pathway as a drug target in cancer therapy. urr Opin Investig. Drugs 8:457–461 Laurendeau I, Ferrer M, Garrido D, 2010, Gene expression profiling of the Hedgehog signaling pathway in human meningiomas. Mol Med 16:262–270 Walter K, Omura N, Hong SM, 2010, Overexpression of smoothened activates the sonic hedgehog signaling pathway in pancreatic cancer-associated fibroblasts. Clin Cancer Res 16:1781–1789 Berman DM, Karhadkar SS, Maitra A, 2003, Widespread requirement for Hedgehog ligand stimulation in growth of digestive tract tumours. Nature 425:846–851 Ma X, Chen K, Huang S, 2005, Frequent activation of the hedgehog pathway in advanced gastric adenocarcinomas. Carcinogenesis 26:1698–705 Thayer SP, di Magliano MP, Heiser PW, 2003, Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 425:851–856 Yanai K, Nagai S, Wada J, 2007, Hedgehog signaling pathway is a possible therapeutic target for gastric cancer. J SurgOncol 95:55–62 Chen G, Goto Y, Sakamoto R et al, 2011, GLI1, a crucial mediator of sonic hedgehog signaling in prostate cancer, functions as a negative modulator for androgen receptor. Biochem Biophys Res Commun 404:809–815 Kelleher FC. Hedgehog signalling and therapeutics in pancreatic cancer. Carcinogenesis. 2010 In press Chen M, HildebrandtMA, Clague J et al, 2010, Genetic variations in the sonic hedgehog pathway affect clinical outcomes in non-muscleinvasive bladder cancer. Cancer Prev Res, Phila, 3:1235–1245 Greene FL, Balch CM, Fleming ID et al, 2002, AJCC cancer staging manual, 6th edn. AJCC Springer-Verlag, New York, pp 335–340 Oue T, Yoneda A, Uehara S et al, 2010, Increased expression of the hedgehog signaling pathway in pediatric solid malignancies. J Pediatr Surg 45:387–392 Brown RS, Wahl RL, 1993, Overexpression of Glut-1 glucose transporter in human breast cancer: an immunohistochemical study. Cancer 72:2979–2985 Ju B, Spitsbergen J, Eden CJ et al, 2009, Co-activation of hedgehog and AKT pathways promote tumorigenesis in zebrafish. Mol Cancer 8:40 Ingham PW, McMahon AP, 2001, Hedgehog signaling in animal development: paradigms and principles. Genes Dev 15:3059–3087 Merchant JL, Saqui-Salces M, El-Zaatari M, 2010, Hedgehog signaling in gastric physiology and cancer. Prog Mol Biol Transl Sci 96:133–156 Morton JP, Mongeau ME, Klimstra DS, 2007, Sonic hedgehog acts at multiple stages during pancreatic tumorigenesis. Proc Natl Acad Sci USA 104:5103–5108 Berman DM, Karhadkar SS, Maitra A, 2003, Widespread requirement for Hedgehog ligand stimulation in growth of digestive tract tumors. Nature 425:846–851 Rush SZ, Abel TW, Valadez JG et al, 2010, Activation of the Hedgehog pathway in pilocytic astrocytomas. Neuro Oncol 12:790–798 Liao X, SiuMK,AuCWet al, 2009)Aberrant activation of hedgehog signaling pathway in ovarian cancers: effect on prognosis, cell invasion and differentiation. Carcinogenesis 30:131–140 Yang Y, Tian X, Xie X et al, 2010, Expression and regulation of hedgehog signaling pathway in pancreatic cancer. Langenbecks Arch Surg 395:515–525 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 349 EP 355 DI 10.1007/s12253-011-9451-2 PG 7 ER PT J AU Lau, CP Wong, YE AF Lau, C Patrick Wong, Y Elaine TI Targeting MET by Tyrosine Kinase Inhibitor Suppresses Growth and Invasion of Nasopharyngeal Carcinoma Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Nasopharyngeal carcinoma; MET; Growth; Invasion; Tyrosine kinase inhibitor ID Nasopharyngeal carcinoma; MET; Growth; Invasion; Tyrosine kinase inhibitor AB Nasopharyngeal carcinoma (NPC) represents a common cancer in endemic areas with high invasive and metastatic potential. It is now known that the HGF-MET signaling pathway plays an important role in mediating the invasive growth of many different types of cancer, including head and neck squamous cell carcinoma. HGF has been shown to stimulate NPC cell growth and invasion in cell line model. The current study aims at demonstrating the effect of MET inhibition by small molecule tyrosine kinase inhibitor PHA665752 on the growth and invasive potential of NPC cell lines. NPC cell lines were used for immunohistochemistry for the MET protein, as well as western blot analysis on MET together with its downstream cascade signaling proteins after treatment with PHA665752. The effect on cell growth, migration and invasion after PHA665752 treatment was also studied. MET inhibition by PHA665752 resulted in highly significant inhibition on NPC cell growth, migration and invasion in vitro. Down-regulation of phospho-MET, phospho-Akt, phospho-MAPK, phospho-STAT3, cyclin D1, β-catenin and PCNA was detected in NPC cells after PHA665752 treatment. MET inhibition with tyrosine kinase inhibitor resulted in suppression of NPC cell growth and invasive potential via down-regulation of a variety of signaling onco-proteins. MET is an important therapeutic target for NPC that warrants further studies and clinical trials. C1 [Lau, C Patrick] Hong Kong Cancer Institute, Sir YK Pao Center for Cancer, Department of Clinical OncologyShatin, Hong Kong SAR, China. [Wong, Y Elaine] Hong Kong Cancer Institute, Sir YK Pao Center for Cancer, Department of Clinical OncologyShatin, Hong Kong SAR, China. RP Lau, CP (reprint author), Hong Kong Cancer Institute, Sir YK Pao Center for Cancer, Department of Clinical Oncology, Shatin, China. EM patricklau75@hotmail.com CR Zur Hausen H, Schulte-Holthausen H, Klein G et al, 1970, EBV DNA in biopsies of Burkitt tumours and anaplastic carcinomas of the nasopharynx. Nature 228:1056–8 Tao Q, Chan ATC, 2007, Nasopharyngeal carcinoma: molecular pathogenesis and therapeutic developments. Expert Rev Mol Med 9:1–24 Lee N, Xia P, Quivey JM et al, 2002, Intensity-modulated radiotherapy in the treatment of nasopharyngeal carcinoma: an update of the UCSF experience. Int J Radiat Oncol Biol Phys 53:12–22 Kam MKM, Teo PML, Chau RMC et al, 2004, Treatment of nasopharyngeal carcinoma with intensity-modulated radiotherapy: The Hong Kong experience. Int J Radiat Oncol Biol Phys 60:1440–50 Baujat B, Audry H, Bourhis J et al, 2006, Chemotherapy in locally advanced nasopharyngeal carcinoma: An individual patient data meta-analysis of eight randomized trials and 1753 patients. Int J Radiat Oncol Biol Phys 64:47–56 Jeffers M, Rong S, Vande Woude GF, 1996, Hepatocyte growth factor/scatter factor-Met signaling in tumorigenicity and invasion/ metastasis. J Mol Med 74:505–13 Birchmeier C, Birchmeier W, Gherardi E et al, 2003, Met, metastasis, motility and more. Nat Rev Mol Cell Bio 4:915–25 Lau PC, Chan AT, 2011, Novel therapeutic target for head and neck squamous cell carcinoma: HGF-MET signaling pathway. Anticancer Drugs 22:665–73 Horikawa T, Sheen T, Takeshita H et al, 2001, Induction of c- Met proto-oncogene by Epstein-Barr Virus Latent Membrane Protein-1 and the correlation with cervical lymph node metastasis of nasopharyngeal carcinoma. Am J Pathol 159:27–33 Qian CN, Guo X, Cao B et al, 2002, Met protein expression level correlates with survival in patients with late-stage nasopharyngeal carcinoma. Cancer Res 62:589–96 Zhou HY, Wan KF, Ip CK et al, 2008, Hepatocyte growth factor enhances proteolysis and invasiveness of human nasopharyngeal cancer cells through activation of PI3K and JNK. FEBS Lett 582:3415–22 Xie LQ, Bian LJ, Li Z et al, 2010, Altered expression of Ecadherin by hepatocyte growth factor and effect on the prognosis of nasopharyngeal carcinoma. Ann Surg Oncol 17:1927–36 Glaser R, Zhang HY, Yao KT et al, 1989, Two epithelial tumor cell lines, HNE-1 and HONE-1, latently infected with Epstein-Barr virus that were derived from nasopharyngeal carcinomas. Proc Natl Acad Sci USA 86:9524–8 Lo AK, Lo KW, Tsao SW et al, 2006, Epstein-Barr virus infection alters cellular signal cascades in human nasopharyngeal epithelial cells. Neoplasia 8:173–80 Paweletz CP, Charboneau L, Bichsel VE et al, 2001, Reverse phase protein microarrays which capture disease progression show activation of pro-survival pathways at the cancer invasion front. Oncogene 20:1981–9 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 357 EP 363 DI 10.1007/s12253-011-9452-1 PG 7 ER PT J AU Ulamec, M Dzombeta, T Cupic, H Lenicek, T Davor, T Kruslin, B AF Ulamec, Monika Dzombeta, Tihana Cupic, Hrvoje Lenicek, Tanja Davor, Tomas Kruslin, Bozo TI Periacinar Retraction Clefting and D2-40 Expression in Prostatic Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE D2 40; Lymphovascular invasion; Prostatic adenocarcinoma; Retraction clefting ID D2 40; Lymphovascular invasion; Prostatic adenocarcinoma; Retraction clefting AB Retraction clefting is known to appear in various types of tumors, but it has only recently been recognized as a specific histological phenomenon. Previously, it was considered merely a laboratory procedure artifact, but lately, there have been some assumptions that peritumoral retractions actually represent lymphatic spaces. In our study, we analyzed neoplastic glands in 52 specimens of prostatic adenocarcinoma. Immunohistochemical analysis was performed using D2-40 antibody, to highlight lymphatic endothelium and thereby differentiate actual lymph vessels or lymphovascular invasion from periacinar retractions. Our results showed that the number of lymph vessels was significantly lower in tumorous tissue compared to adjacent normal prostatic tissue. On the other hand, the number of lymph vessels in tumorous tissue was significantly higher than the number of lymph vessels mimicking periacinar retractions. Overall, the number of lymph vessels mimicking periacinar clefts was particularly low. These results are in accordance with our previous studies, which had shown that periacinar clefting appears due to lack of basal cells and stromal changes around tumorous acini. Also, these results support our hypothesis that retractions do not represent lymph vessels but should be considered a distinct entity, which is proven to be helpful both as diagnostic and predictive factor. C1 [Ulamec, Monika] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska cesta 29Zagreb, Croatia. [Dzombeta, Tihana] University of Zagreb, School of Medicine, Department of PathologyZagreb, Croatia. [Cupic, Hrvoje] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska cesta 29Zagreb, Croatia. [Lenicek, Tanja] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska cesta 29Zagreb, Croatia. [Davor, Tomas] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska cesta 29Zagreb, Croatia. [Kruslin, Bozo] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Vinogradska cesta 29Zagreb, Croatia. RP Kruslin, B (reprint author), University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, Zagreb, Croatia. EM bozo.kruslin@kbcsm.hr CR Halpert B, Schmalhorst WR, 1996, Carcinoma of the prostate in patients 70–79 years old. Cancer 1919:695–698 Halpert B, Sheehan EA, Schmalhorst WR et al, 1963, Carcinoma of the prostate: a survey of 5000 autopsies. Cancer 16:736–742 Kruslin B, Tomas D, Mikuz G, 2011, Periacinar retraction artifact of prostate. Front Biosci 3:226–35 Srigley JR, 2004, Benign mimickers of prostatic adenocarcinoma. Mod Pathol 17:328–48 Herawi M, Parwani AV, Irie J et al, 2005, Small glandular proliferations on needle biopsies: most common benign mimickers of prostatic adenocarcinoma sent in for expert second opinion. Am J Surg Pathol 29:874–80 Ulamec M, Tomas D, Ensinger C et al, 2007, Periacinar retraction clefting in proliferative prostatic atrophy and prostatic carcinoma. J Clin Pathol 60:1098–1101 Kruslin B, Tomas D, Rogatsch H et al, 2003, Periacinar retraction clefting in the prostatic needle core biopsies: an important diagnostic criterion or a simple artifact? Virchows Arch 443:524–7 Kruslin B, Tomas D, Rogatsch H et al, 2005, Correlation of periacinar retraction clefting in needle core biopsies and corresponding prostatectomy specimens of patients with prostatic adenocarcinoma. Int J Surg Pathol 13:67–72 Irie J, Manucha V, Ioffe OB et al, 2007, Artefact as the pathologist’s friend: peritumoral retraction in in situ and infiltrating duct carcinoma of the breast. Int J Surg Pathol 15:53–9 Tomas D, Kruslin B, 2004, The potential value of, myo, fibroblastic stromal reaction in the diagnosis of prostatic adenocarcinoma. Prostate 61:324–331 Tomas D, Ulamec M, Hudolin T et al, 2006, Myofibroblastic stromal reaction and expression of tenascin-C and laminin in prostate cancer. Prostate Cancer Prostatic Dis 9:414–419 Acs G, Dumoff KL, Solin LJ et al, 2007, Extensive retraction artifact correlates with lymphatic invasion and nodal metastasis and predicts poor outcome in early stage breast carcinoma. Am J Surg Pathol 31:129–40 Acs G, Paragh G, Chuang ST et al, 2009, The presence of micropapillary features and retraction artifact in core needle biopsy material predicts lymph node metastasis in breast carcinoma. Am J Surg Pathol 33:202–10 Bujas T, Pavic I, Lenicek T et al, 2008, Peritumoral retraction clefting correlates with advanced stage squamous cell carcinoma of the esophagus. Pathol Oncol Res 14:443–7 Tomas D, Spajic B, Milosevic M et al, 2011, Extensive retraction artifacts predict biochemical recurrence-free survival in prostatic carcinoma. Histopathology 58:447–54 Marks A, Sutherland DR, Bailey D et al, 1999, Characterization and distribution of an oncofetal antigen, M2A antigen, expressed on testicular germ cell tumors. Br J Cancer 80:569–578 Kahn HJ, Bailey D, Marks A, 2002, Monoclonal antibody D2-40, a new marker of lymphatic endothelium, reacts with Kaposi’s and a subset of angiosarcomas. Mod Pathol 15:434–440 Kahn HJ, Marks A, 2002, A new monoclonal antibody, D2-40, for detection of lymphatic invasion in primary tumors. Lab Invest 82:1255–1257 Kruslin B, Tomas D, Cviko A et al, 2006, Periacinar clefting and p63 immunostaining in prostatic intraepithelial neoplasia and prostatic carcinoma. Pathol Oncol Res 12:205–9 Weinstein MH, Signoretti S, Loda M, 2002, Diagnostic utility of immunohistochemical staining for p63, a sensitive marker of prostatic basal cells. Mod Pathol 15:1302–8 Davis LD, Zhang W, Merseburger A et al, 2002, p63 expression profile in normal and malignant prostate epithelial cells. Anticancer Res 22:3819–25 Roma AA, Magi-Galluzzi C, Kral MA et al, 2006, Peritumoral lymphatic invasion is associated with regional lymph node metastases in prostate adenocarcinoma. Mod Pathol 19:392–8 Cheng L, Bishop E, Zhou H et al, 2008, Lymphatic vessel density in radical prostatectomy specimens. Hum Pathol 39:610–5 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 365 EP 370 DI 10.1007/s12253-011-9453-0 PG 6 ER PT J AU Valcz, G Sipos, F Krenacs, T Molnar, J Patai, V Leiszter, K Toth, K Wichmann, B Molnar, B Tulassay, Zs AF Valcz, Gabor Sipos, Ferenc Krenacs, Tibor Molnar, Jeannette Patai, V Arpad Leiszter, Katalin Toth, Kinga Wichmann, Barna Molnar, Bela Tulassay, Zsolt TI Increase of α-SMA+ and CK+ Cells as an Early Sign of Epithelial-Mesenchymal Transition during Colorectal Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Epithelial–myofibroblast transition; Adenoma–carcinoma sequence; Cytokeratin; Alpha–smooth muscle actin ID Epithelial–myofibroblast transition; Adenoma–carcinoma sequence; Cytokeratin; Alpha–smooth muscle actin AB Our aim was to examine cell transition events by detecting the frequency of intrapithelial α-smooth muscle actin (SMA)+/cytokeratin (CK)+ cells during colorectal adenoma–carcinoma sequence, in relation to E-cadherin expression. Our further aim was to determine the proliferative activity of intraepithelial α-SMA+ cells. Histologically healthy, adenoma, and colorectal cancer (CRC) biopsy samples were taken during routine colonoscopy and were included into tissue microarrays (TMAs). Slides immunostained for Ki-67, α-SMA, E-cadherin and pan-cytokeratin were digitalized and analyzed by using a digital microscope software. The proportion of α-SMA+/CK+ cells was significantly higher in CRC samples (3.34±1.01%) compared to healthy (1.94±0.69%) or adenoma (1.62±0.78%) samples (p<0.01). E-cadherin expression negatively correlated with the number of α-SMA+ cells. The majority of intraepithelial α-SMA+ cells were in the proliferative phase. During tumor progression, the appearance of dot-like α-SMA staining in CK positive cells may indicate the initial phase of the epithelial-tomesenchymal transition (EMT). The high proportion of intraepithelial α-SMA+ proliferating cells may refer to their increased plasticity compared to differentiated cells. The negative correlation between E-cadherin and intraepithelial α-SMA expression suggests that EMT is facilitated by a loss of epithelial cell contact. C1 [Valcz, Gabor] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Molnar, Jeannette] National Institute of Food and Nutrition ScienceBudapest, Hungary. [Patai, V Arpad] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Leiszter, Katalin] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Toth, Kinga] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Wichmann, Barna] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Molnar, Bela] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. [Tulassay, Zsolt] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. RP Valcz, G (reprint author), Semmelweis University, 2nd Department of Internal Medicine, Budapest, Hungary. CR Schultheiss G, Diener M, 1998, K+ and Cl- conductances in the distal colon of the rat. Gen Pharmacol 31:337–342 Lu L, Walker WA, 2001, Pathologic and physiologic interactions of bacteria with thegastrointestinal epithelium. Am J Clin Nutr 73:1124–1130 Edmonds CJ, 1984, Absorption and secretion of fluid and electrolytes by the rectum. Scand J Gastroenterol Suppl 93:79–87 Hollier BG, Evans K, Mani SA, 2009, The epithelial-tomesenchymal transition and cancer stem cells: a coalition against cancer therapies. J Mammary Gland Biol Neoplasia 14:29–43 Thiery JP, Acloque H, Huang RYet al, 2009, Epithelial-mesenchymal transitions in development and disease. Cell 139:871–890 Kalluri R,Weinberg RA, 2009, The basics of epithelial-mesenchymal transition. J Clin Invest 119:1420–1428 Kalluri R, 2009, EMT: when epithelial cells decide to become mesenchymal-like cells. J Clin Invest 119:1417–1419 Hugo H, Ackland ML, Blick T et al, 2007, Epithelial–mesenchymal and mesenchymal–epithelial transitions in carcinoma progression. J Cell Physiol 213:374–383 Radisky DC, 2005, Epithelial-mesenchymal transition. J Cell Sci 118:4325–4326 Boyer B, Valles AM, Edme N, 2000, Induction and regulation of epithelial-mesenchymal transitions. Biochem Pharmacol 60:1091–1099 Zavadil J, Bottinger EP, 2005, TGF-β and epithelial-tomesenchymal transitions. Oncogene 24:5764–5774 Lorusso G, Ruegg C, 2008, The tumor microenvironment and its contribution to tumor evolution toward metastasis. Histochem Cell Biol 130:1091–1103 Miyazono K, 2009, Transforming growth factor-beta signaling in epithelial-mesenchymal transition and progression of cancer. Proc Jpn Acad Ser B Phys Biol Sci 85:314–323 Masszi A, Speight P, Charbonney E et al, 2010, Fate-determining mechanisms in epithelial-myofibroblast transition: major inhibitory role for Smad3. J Cell Biol 188:383–399 Masszi A, Fan L, Rosivall L, McCulloch CA et al, 2004, Integrity of cell-cell contacts is a critical regulator of TGF-beta 1-induced epithelial-to-myofibroblast transition: role for beta-catenin. Am J Pathol 165:1955–1967 Reichert M, Muller T, Hunziker W, 2000, The PDZ domains of zonula occludens-1 induce an epithelial to mesenchymal transition of Madin-Darby canine kidney I cells. Evidence for a role of betacatenin/ Tcf/Lef signaling. J Biol Chem 275:9492–9500 Wang J, Zohar R, McCulloch CA, 2006, Multiple roles of alphasmooth muscle actin in mechanotransduction. Exp Cell Res 312:205–214 Li H, Fan X, Houghton J, 2007, Tumor microenvironment: the role of the tumor stroma in cancer. J Cell Biochem 101:805–815 Jass JR, 2007, Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology 50:113–130 Storch KN, Taatjes DJ, Bouffard NA et al, 2007, Alpha smooth muscle actin distribution in cytoplasm and nuclear invaginations of connective tissue fibroblasts. Histochem Cell Biol 127:523–530 Thomas CH, Collier JH, Sfeir CS et al, 2002, Engineering gene expression and protein synthesis by modulation of nuclear shape. Proc Natl Acad Sci USA 99:1972–1977 Wang J, Chen H, Seth A et al, 2003, Mechanical force regulation of myofibroblast differentiation in cardiac fibroblasts. Am J Physiol Heart Circ Physiol 285:1871–1881 Gomez EW, Chen QK, Gjorevski N et al, 2010, Tissue geometry patterns epithelial-mesenchymal transition via intercellular mechanotransduction. J Cell Biochem 110:44–51 Ng YY, Huang TP, Yang WC et al, 1998, Tubular epithelialmyofibroblast transdifferentiation in progressive tubulointerstitial fibrosis in 5/6 nephrectomized rats. Kidney Int 54:864–876 Zhou BP, Deng J, Xia W et al, 2004, Dual regulation of Snail by GSK-3beta-mediated phosphorylation in control of epithelialmesenchymal transition. Nat Cell Biol 6:931–940 Batlle E, Sancho E, Franci C et al, 2000, The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol 2:84–89 Brabletz T, Hlubek F, Spaderna S et al, 2005, Invasion and metastasis in colorectal cancer: epithelial-mesenchymal transition, mesenchymal-epithelial transition, stem cells and beta-catenin. Cells Tissues Organs 179:56–65 Hawinkels LJ, Verspaget HW, van der Reijden JJ et al, 2009, Active TGF-beta1 correlates with myofibroblasts and malignancy in the colorectal adenoma-carcinoma sequence. Cancer Sci 100:663–670 Fan L, Sebe A, Peterfi Z et al, 2007, Cell contact-dependent regulation of epithelial-myofibroblast transition via the rho-rho kinase-phospho-myosin pathway. Mol Biol Cell 18:1083–1097 Masszi A, Di Ciano C, Sirokmany G et al, 2003, Central role for Rho in TGF-beta1-induced alpha-smooth muscle actin expression during epithelial-mesenchymal transition. Am J Physiol Renal Physiol 284:911–924 Pino MS, Kikuchi H, Zeng M et al, 2010, Epithelial to mesenchymal transition is impaired in colon cancer cells with microsatellite instability. Gastroenterology 138:1406–1417 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 371 EP 376 DI 10.1007/s12253-011-9454-z PG 6 ER PT J AU Gong, L Cui, Z Yu, X Wei, Y Peng, J Leng, X AF Gong, Lei Cui, Zhuqingqing Yu, Xin Wei, Yuhua Peng, Jirun Leng, Xisheng TI Hexokinase II in CD133+ and CD133- Hepatoma BEL-7402 Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatoma; CD133; Hexokinase II; BEL- 7402; Therapeutic target ID Hepatoma; CD133; Hexokinase II; BEL- 7402; Therapeutic target AB Hexokinase II is a key enzyme in the glycolytic pathway and CD133+ human hepatoma cells possess cancer stem cell-like properties. The expression and enzyme activity of hexokinase II in CD133+ and CD133- hepatoma cells were examined. CD133 on the surface of the hepatoma BEL-7402 cells was analyzed by flow cytometry and the cells were magnetically sorted into CD133+ and CD133- groups. CD133+ cells comprised 1.04% of the total BEL-7402 cell population. Reverse transcription-polymerase chain reaction (PCR) and quantitative real-time PCR were used to assay the expression of hexokinase II mRNA in these two groups. The level of mRNA in CD133- cells was 4.35 times greater than the level in CD133+ cells. 3,6-biphosphoglucose dehydrogenasecoupled colorimetric method and temperature-sensitive trials were applied to determine the enzyme activity of hexokinase II, which was 1.02 U/g protein in CD133+ cells and 2.47 U/g protein in CD133- cells. Hexokinase II was the major active hexokinase isoform in CD133+ cells, comprising 92.7% of the overall cellular hexokinase activity. The results indicate that hexokinase II is vitally meaningful for CD133+ hepatoma BEL-7402 cells. Hexokinase II represents a new therapeutic target for treating CD133+ hepatoma cells. C1 [Gong, Lei] Peking University People’s Hospital, Department of Hepatobiliary Surgery, No. 11 South Xizhimen Street, Xicheng District, 100044 Beijing, China. [Cui, Zhuqingqing] Peking University People’s Hospital, Department of Hepatobiliary Surgery, No. 11 South Xizhimen Street, Xicheng District, 100044 Beijing, China. [Yu, Xin] Peking University People’s Hospital, Department of Hepatobiliary Surgery, No. 11 South Xizhimen Street, Xicheng District, 100044 Beijing, China. [Wei, Yuhua] Peking University People’s Hospital, Department of Hepatobiliary Surgery, No. 11 South Xizhimen Street, Xicheng District, 100044 Beijing, China. [Peng, Jirun] Peking University People’s Hospital, Department of Hepatobiliary Surgery, No. 11 South Xizhimen Street, Xicheng District, 100044 Beijing, China. [Leng, Xisheng] Peking University People’s Hospital, Department of Hepatobiliary Surgery, No. 11 South Xizhimen Street, Xicheng District, 100044 Beijing, China. RP Leng, X (reprint author), Peking University People’s Hospital, Department of Hepatobiliary Surgery, 100044 Beijing, China. EM lengxs2003@126.com CR Yin AH, Miraglia S, 1997, AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood 90:5002–5012 Shmelkov SV, Clair R, 2005, AC133/CD133/Prominin-1. Int J Biochem Cell Biol 37:715–719 Singh SK, Clarke ID, 2003, Identification of a cancer stem cell in human brain tumors. Cancer Res 63:5821–5828 Singh SK, Hawkins C, 2004, Identification of human brain tumor initiating cells. Nature 432:396–401 Bussolati B, Bruno S, 2005, Isolation of renal progenitor cells from adult human kidney. Am J Pathol 166:545–555 O’Brien CA, Pollett A, 2007, A human colon cancer cell capable of initiating tumor growth in immunodeficient mice. Nature 445:106–110 Ricci-Vitiani L, Lombardi DG, 2007, Identification and expansion of human colon cancer-initiating cells. Nature 445:111–115 Hermann PC, Huber SL, 2007, Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1:313–323 Li C, Heidt DG, 2007, Identification of pancreatic cancer stem cells. Cancer Res 67:1030–1037 Collins AT, Barry PA, 2005, Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 65:10946– 10951 Suetsugu A, Nagaki M, 2006, Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells. Biochem and Biophys Res Comm 351:820–824 Yin S, Li J, 2007, CD133 positive hepatocellular carcinoma cells possess high capacity for tumorigenicity. Int J Cancer 120:1444– 1450 Warburg O, 1956, On the origin of cancer cells. Science 123:309–314 Pedersen PL, Mathupala S, 2002, Mitochondrial bound type II hexokinase: a key player in the growth and survival of many cancers and an ideal prospect for therapeutic intervention. Biochim Biophys Acta 1555:14–20 Wolf A, Agnihotri S, Micallef J et al, 2011, Hexokinase 2 is a key mediator of aerobic glycolysis and promotes tumor growth in human glioblastoma multiforme. J Exp Med 208:313–326 Adams V, KempfW(1995, Determination of hexokinase isoenzyme I and II composition by RT-PCR: increased hexokinase isoenzyme II in human renal cell carcinoma. Biochem Mol Med 54:53–58 Rempel A, Bannasch P, 1994, Differences in expression and intracellular distribution of hexokinase isoenzymes in rat liver cells of different transformation stages. Biochim Biophys Acta 1219:660–668 Nakashima RA, Paggi MG, 1988, Purification and characterization of a bindable form of mitochondrial bound hexokinase from the highly glycolytic AS-30D rat hepatoma cell line. Cancer Res 48:913–919 Shinohara Y, Ichihara J, 1991, Remarkably enhanced expression of the type II hexokinase in rat hepatoma cell line AH130. FEBS Lett 291:55–77 Yasuda S, Arii S, 2004, Hexokinase II and VEGF expression in liver tumors: correlation with hypoxia-inducible factor-1α and its significance. J Hepato 40:117–123 Mandarino LJ, Printz RL, 1995, Regulation of hexokinase II and glycogen synthase mRNA, protein, and activity in human muscle. Am J Physiol Endocrinol Metab 269:E701–E708 Uchida N, BuckDW(2000, Direct isolation of human central nervous system stem cells. Proc Natl Acad Sci USA 97:14720–14725 Kemper K, Grandela C, Medema JP, 2010, Molecular identification and targeting of colorectal cancer stem cells. Oncotarget 1:387–395 Zhang QZ, Shi SH, 2010, A subpopulation of CD133+ cancer stem-like cells characterized in human oral squamous cell carcinoma confer resistance to chemotherapy. Cancer Lett 289:151–160 Ulrike K, Mechthild K, 2010, Cancer stem cells at the crossroads of current cancer therapy failures-Radiation oncology perspective. Semin Cancer Biol 20:116–124 Shinohara Y, 2000, Identification and characterization of hexokinase isozyme predominantly expressed in malignant tumor cells. Yakugaku Zasshi 120:657–666 Golshani-Hebroni SG, Bessman SP, 1997, Hexokinase binding to mitochondria: a basis for proliferative energy metabolism. J Bioenerg Biomembr 29:331–338 Plas DR, Thompson CB, 2002, Cell metabolism in the regulation of programmed cell death. Trends Endocrinol Metab 13:75–78 Pastorino JG, Shulga N, 2003, Mitochondrial binding of hexokinase II inhibits Bax-induced cytochrome c release and apoptosis. J Biol Chem 277:7610–7618 Kim W, Yoon JH, 2007, Apoptosis-inducing antitumor efficacy of hexokinase II inhibitor in hepatocellular carcinoma. Mol Cancer Ther 6:2554–2562 Ihrlund LS, Hernlund E, Khan O et al, 2008, 3-Bromopyruvate as inhibitor of tumour cell energy metabolism and chemopotentiator of platinum drugs. Mol Oncol 2:94–101 Mathupala SP, Ko YH, Pedersen PL, 2010, The pivotal roles of mitochondria in cancer: Warburg and beyond and encouraging prospects for effective therapies. Biochim Biophys Acta 1797:1225–1230 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 377 EP 381 DI 10.1007/s12253-011-9455-y PG 5 ER PT J AU Singh, A Mishra, KA Ylaya, K Hewitt, MS Sharma, ChK Saxena, S AF Singh, Avninder Mishra, Kumar Ashwani Ylaya, Kris Hewitt, M Stephen Sharma, Chand Karam Saxena, Sunita TI Wilms Tumor-1, Claudin-1 and Ezrin Are Useful Immunohistochemical Markers That Help to Distinguish Schwannoma from Fibroblastic Meningioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Biomarkers; Fibroblastic meningioma; Immunohistochemistry; Schwannoma; Tissue microarray ID Biomarkers; Fibroblastic meningioma; Immunohistochemistry; Schwannoma; Tissue microarray AB The aim of this study is to identify immunohistochemical (IHC) markers that can reliably separate schwannoma (SCHW) and fibroblastic meningioma (FM). We selected 106 cases of intracranial SCHW (n=56) and FM (n=50) and constructed a tissue microarray (TMA) of core diameter of 1.0 mm from archival formalin-fixed paraffin-embedded tissue. ATMA-IHC was performed using 14 antibodies. After IHC staining, 98 cores were found suitable for evaluation. The IHC staining was scored as 0–2+ (0, negative; 1+, weak and/or focal 2+ strong and/or diffuse positive). A discriminant analysis (DA) (Wilks’Lambda test) was performed to assess the relative importance of these biomarkers in classifying the two groups FM and SCHW. It showed that WT-1 (Wilks’λ 0.085, p<0.001), EMA (Wilks’λ 0.253, p<0.001), S100 (Wilks’λ 0.487, p<0.001), Claudin-1 (Wilks’ λ 0.57, p<0.001) and Ezrin (Wilks’λ 0.656, p<0.001), SPARC (Wilks’λ 0.751, p<0.01), NP-Y (Wilks’λ, 0.819, p<0.001) and EGFR (Wilks’λ 0.845, p=0.026) were some of the statistically significant markers that discriminated SCHW and FM. For sensitivity and specificity for SCHW the significant markers [Area under the curve (95% CI), p-value] by ROC analysis were WT-1 [0.990(0.000, 1.000), <0.001], S100 [0.880(0.808, 0.951), <0.001] while for diagnosing FM the most sensitive and specific markers were EMA [0.957(0.914, 1.000), <. 001], Claudin-1 [0.857(0.782, 0.932), <0.001] and ezrin [0.792(0.700,0.884),<0.001]. WT-1, Claudin-1 and Ezrin may be potentially useful immunohistochemical adjuncts to EMA and S100 that differentiate SCHW from FM. C1 [Singh, Avninder] National Institute of Pathology (ICMR), Safdarjung Hospital Campus, Room 602, 6th floor, 110029 New Delhi, India. [Mishra, Kumar Ashwani] National Institute of Pathology (ICMR), Safdarjung Hospital Campus, Room 602, 6th floor, 110029 New Delhi, India. [Ylaya, Kris] National Institutes of Health, National Cancer Institute, TARP LabBethesda, MD, USA. [Hewitt, M Stephen] National Institutes of Health, National Cancer Institute, TARP LabBethesda, MD, USA. [Sharma, Chand Karam] Safdarjung Hospital, Department of NeurosurgeryNew Delhi, India. [Saxena, Sunita] National Institute of Pathology (ICMR), Safdarjung Hospital Campus, Room 602, 6th floor, 110029 New Delhi, India. RP Singh, A (reprint author), National Institute of Pathology (ICMR), Safdarjung Hospital Campus, 110029 New Delhi, India. EM dravninder@yahoo.co.in CR Louis DN, Scheithauer BW, Budka H, von Deimling A et al, 2000, Meningiomas. In: Kleihues P, Cavenee WK, eds, Tumours of the nervous system. IARC, Lyon, pp 176–184 Sloof JL, 1984, Pathological anatomical findings in the cerebellopontine angle. Adv Otorhinolaryngol 34:89–953 Weiss SW, Langloss JM, Enzinger FM, 1983, Value of S-100 protein in the diagnosis of soft tissue tumors with particular reference to benign and malignant Schwann cell tumors. Lab Investig 49:299–308 Theaker JM, Gatter KC, Esiri MM, Fleming KA, 1986, Epithelial membrane antigen and cytokeratin expression by meningiomas: an immunohistological study. J Clin Pathol 39:435–439 Meis JM, Ordonez NG, Bruner JM, 1986, Meningiomas. An immunohistochemical study of 50 cases. Arch Pathol Lab Med 110:934–937 Schnitt SJ, Vogel H, 1986, Meningiomas. Diagnostic value of immunoperoxidase staining for epithelial membrane antigen. Am J Surg Pathol 9:640–649 Sloan JP, Ormerod MG, 1981, Distribution of epithelial membrane antigen in normal and neoplastic tissues and its value on diagnosis on diagnostic tumor pathology. Cancer 47:1786–1795 Perentes E, Nakagawa Y, Ross GW et al, 1987, Expression of epithelial membrane antigen in perineurial cells and their derivatives. An immunohistochemical study with multiple markers. Acta Neuropathol 75:160–165 Winek RR, Scheithauer BW, Wick MR, 1989, Meningioma, meningeal hemangiopericytoma, angioblastic meningioma), peripheral hemangiopericytoma, and acoustic schwannoma, 1989, a comparative immunohistochemical study. Am J Surg Pathol 13:251–261 Artlich A, Schmidt D, 1990, Immunohistochemical profile of meningiomas and their histological subtypes. Hum Pathol 21:843– 849 Perry A, Scheithauer BW, Nascimento AG, 1997, The immunophenotypic spectrum of meningeal hemangiopericytoma: a comparison with fibrous meningioma and solitary fibrous tumor of meninges. Am J Surg Pathol 21:1354–1360 Carneiro SS, Scheithauer BW, Nascimento AG, Hirose T, Davis DH, 1996, Solitary fibrous tumor of the meninges: a lesion distinct from fibrous meningioma. Am J Clin Pathol 106:217– 2242 Hahn HP, Bundock EA, Hornick JL, 2006, Immunohistochemical staining for claudin-1 can help distinguish menigiomas from histologic mimics. Am J Clin Pathol 125:203–208 Avninder S, Ylaya K, Hewitt SM, 2008, Tissue microarray: a simple technology that has revolutionized research in pathology. J Postgrad Med 54:158–162 Lusis EA, Chicoine MR, Perry A, 2005, High throughput screening of meningioma biomarkers using a tissue microarray. J Neuro Oncol 73:219–223 Sarwat AM, Al-Salam S, 2009, Expression of HIF-1, galectin, cox-2 and Wilms tumor-1 protein in multiple schwannomas of the conus medullaris. J Neuro Oncol 92:111–115 Lee SB, Huang K, Palmer R, Truong VB, Herzlinger D, Kolquist KA et al, 1999, Wilms’ tumor suppressor WT1 encodes a transcriptional activator of amphiregulin. Cell 98:663–673 Bhattacharjee M, Adesina AM, Goodman C, Powell S, 2003, Claudin-1 expression in meningiomas and schwannomas: possible role in differential diagnosis [abstract]. J Neuropathol Exp Neurol 62:581 Stemmer-Rachamimov AO, Xu L, Gonzalez-Augusti C et al, 1997, Universal absence of merlin, but not other ERM family members in schwannomas. Am J Pathol 151:1649–1654 Bohling T, Turunen O, Jaaskelainen J et al, 1996, Ezrin expression in stromal cells of capillary hemangioblastoma. An immunohistochemical survey of brain tumors. Am J Pathol 148:367–373 Martinez-Glez V, Hernandez-Franco C, Rey JA, 2008, Microarray gene expression profiling in meningiomas and schwannnomas. Curr Med Chem 15:826–833 Welling DB, Lasak JM, Akhmametyeva A, Ghaheri B, Chang LS, 2002, A cDNA microarray analysis of vestibular schwannomas. Otol Neurotol 23:736–748 Fine SW, McClain SA, Maomi L, 2004, Immunohistochemical staining for calretinin is useful for differentiating schwannomas from neurofibromas. Am J Clin Pathol 122:552–559 Perry A, Cai DX, Scheithauer BW, Swanson PE, Lohse CM, Newsham IF et al, 2000, Merlin, DAL-1, and progesterone receptor expression in clinicopathologic subsets of meningioma: a correlative immunohistochemical study of 175 cases. J Neuropathol Exp Neurol 59:872–879 Roser F, Nakamuara M, Bellinzona M, Rosahl SK, Ostertag H, Samii M, 2004, The prognostic value of progesterone receptor status in meningiomas. J Clin Pathol 57:1033–1037 Omulecka A, Papierz W, Nawrocka-Kunecka A, Lewy-Trenda I, 2006, Immunohistochemical expression of progesterone and estrogen receptors in meningiomas. Folia Neuropathol 44:111– 115 Jaiswal S, Agrawal V, Jaiswal AK, Pandey R, Mahapatra AK, 2009, Expression of estrogen and progesterone receptors in vestibular schwannomas and their clinical significance. J Negat Results Biomed 8:9 Dalgorf DM, Roswell C, Bilbao JM, Chen JM, 2008, Immunohistochemical investigation of hormone receptors and vascular growth factor concentration in vestibular schwannomas. Skull Base 18:377–384 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 383 EP 389 DI 10.1007/s12253-011-9456-x PG 7 ER PT J AU Nemeth, BI Tiszlavicz, L AF Nemeth, Balazs Istvan Tiszlavicz, Laszlo TI Biphenotypic Surface Epithelial Cells in the Gastrointestinal Tube with Mixed Epithelial-Myofibroblastic Differentiation: A Paradigm SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Myofibroblast; Actin; Stem cell ID Myofibroblast; Actin; Stem cell AB Epithelial cells and myofibroblasts are wellcharacterized histomorphological elements of tissues. They are distinguished from one another on the basis of topography and of differences in cytokeratin (CK) and α-smooth muscle actin (SMA) expression. Certain epithelial cells exhibit CK / SMA co-expression. This study aimed to define the immunophenotypical characteristics of these biphenotypic cells with respect to cytodifferentiation (broad spectrum of CKs, SMA), cell-cell interaction (E-cadherin, adenomatous polyposis coli - APC, β-catenin), and cell survival (cyclooxygenase-2 - Cox-2). At the routine gastrointestinal pathology service of the Department of Pathology, University of Szeged, tissue samples were identified from instances of cervical inlet patch (n=5), Barrett’s esophagus (n=5), gastritis (n=5), fundic gland polyp (n=2), gastric neoplastic polyp (n=1), inflammatory bowel disease (n=5), and colonic neoplastic polyp (n=3). that contained epithelial cells expressing SMA. These biphenotypic cells were further immunophenotyped. Foregut-derived biphenotypic cells expressed CKs 7 and 20, while hindgut-derived biphenotypic cells expressed only CK 20. Subepithelial myofibroblasts adjacent to biphenotypic epithelium expressed Cox-2, SMA, and β-catenin, as did biphenotypic cells. Myofibroblasts, however, did not express CKs. In neoplastic polyps, APC expression weakened as cytologic atypism increased, while intermingled biphenotypic cells in neoplastic glands overexpressed APC, as did myofibroblasts beneath. CK subspecies expression in biphenotypic cells reflects embryonic development of the gastrointestinal tract. The immunophenotyping analysis addresses bidirectional (via transdifferentiation from epithelia into myofibroblasts or vice versa) formation of biphenotypic cells within damaged epithelium, a phenomenon that must be further analysed. C1 [Nemeth, Balazs Istvan] University of Szeged, Department of Pathology, 6720 Szeged, Hungary. [Tiszlavicz, Laszlo] University of Szeged, Department of Pathology, 2 Allomas Street, 6720 Szeged, Hungary. RP Nemeth, BI (reprint author), University of Szeged, Department of Pathology, 6720 Szeged, Hungary. EM nemeth.istvan.balazs@med.u-szeged.hu CR Adegboyega PA, Mifflin RC, DiMari JF, Saada JI, Powell DW, 2002, Immunohistochemical study of myofibroblasts in normal colonic mucosa, hyperplastic polyps, and adenomatous colorectal polyps. Arch Pathol Lab Med 126:829–836 Bach SP, Renehan AG, Potten CS, 2000, Stem cells: the intestinal stem cell as a paradigm. Carcinogenesis 3:469–476 Behrens J, Jerchow BA, Wurtele M, Grimm J, Asbrand C, Wirtz R, Kuhl M, Wedlich D, Birchmeier W, 1998, Functional interaction of an axin homolog, conductin, with beta-catenin, APC and GSK3beta. Science 280:596–599 Brachvogel B, Moch H, Pausch F, Schlotzer-Schrehardt U, Hofmann C, Hallmann R, von der Mark K, Winkler T, Poschl E, 2005, Perivascular cells expressing annexin A5 define a novel mesenchymal stem cell-like population with the capacity to differentiate into multiple mesenchymal lineages. Development 132:2657–2668 De Vriese S, Tilton RG, Mortier S, Lameire NH, 2006, Myofibroblast transdifferentiation of mesothelial cells is mediated by RAGE and contributes to peritoneal fibrosis in uraemia. Nephrol Dial Transplant 21:2549–2555 Hermiston ML, Wong MH, Gordon JI, 1996, Forced expression of E-cadherin in the mouse intestinal epithelium slows cell migration and provides evidence for nonautonomous regulation of cell fate in a self-renewing system. Genes Develop 10:985–996 Houghton JM, Stoicov C, Nomura S, Rogers AB, Carlson J, Li H, Cai X, Fox JG, Goldenring JR, Wang TC, 2004, Gastric cancer originating from bone marrow–derived cells. Science 306:1568– 1571 Jain MK, Layne MD, Watanabe M, Chin MT, Feinberg MW, Sibinga NE, Hsieh CM, Yet SF, Stemple DL, Lee ME, 1998, In vitro system for differentiating pluripotent neural crest cells into smooth muscle cells. J Biol Chem 273:5993–5996 Joyce NC, Haire MF, Palade GE, 1987, Morphologic and biochemical evidence for a contractile cell network within the rat intestinal mucosa. Gastroenterology 92:68–81 Ng YY, Huang TP, Yang WC, Chen ZP, Yang AH, Mu W, Nikolic-Paterson DJ, Atkins RC, Lan HY, 1998, Tubular epithelial-myofibroblast transdifferentiation in progressive tubulointerstitial fibrosis in 5/6 nephrectomized rats. Kidney Int 54:864–876 Powell DW, Mifflin RC, Valentich JD, Crowe SE, Saada JI, West AB, 1999, Myofibroblasts. I. Paracrine cells important in health and disease. Am J Physiol 277:C1–C19 Powell DW, Mifflin RC, Valentich JD, Crowe SE, Saada JI, West AB, 1999, Myofibroblasts. II. Intestinal subepithelial myofibroblasts. Am J Physiol 277:C183–C201 Powell DW, Adegboyega PA, DiMari JF, Mifflin RC, 2005, Epithelial cells and their neighbors I. Role of intestinal myofibroblasts in development, repair, and cancer. Am J Physiol Gastrointest Liver Physiol 289:G2–G7 Sonoshita M, Takaku K, Oshima M, Sugihara K, Taketo MM, 2002, Cyclooxygenase-2 expression in fibroblasts and endothelial cells of intestinal polyps. Cancer Res 62:6846–6849 Tsujii M, DuBois RN, 1995, Alterations in cellular adhesion and apoptosis in epithelial cells overexpressing prostaglandin endoperoxide synthases-2. Cell 83:493–501 Valentich JD, Popov V, Saada JI, Powell DW, 1997, Phenotypic characterization of an intestinal subepithelial myofibroblast cell line. Am J Physiol 272:C1513–C1524 Vyas SK, Leyland H, Gentry J, Arthur MJ, 1995, Rat hepatic lipocytes synthesize and secrete transin, stromelysin, in early primary culture. Gastroenterology 109:889–898 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 391 EP 396 DI 10.1007/s12253-011-9457-9 PG 6 ER PT J AU Liao, M Tong, P Zhao, J Zhang, Y Li, Z Wang, J Feng, X Hu, M Pan, Y AF Liao, Mingmei Tong, Ping Zhao, Jinfeng Zhang, Yangde Li, Zhehai Wang, Jiwei Feng, Xueping Hu, Man Pan, Yifeng TI Prognostic Value of Matrix Metalloproteinase-1/ Proteinase-Activated Receptor-1 Signaling Axis in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Matrix metalloproteinase-1; Proteinase-activated receptor-1; Prognosis ID Hepatocellular carcinoma; Matrix metalloproteinase-1; Proteinase-activated receptor-1; Prognosis AB Matrix metalloproteinase-1 (MMP-1) is proposed to be involved in both tumor cell invasion and metastasis. MMP-1 proteolytically activates protease activated receptor-1 (PAR-1), which also plays an important role in tumor development and progression. However, it is currently unknown whether MMP-1 activation of PAR-1 has relevance to the progression of hepatocellular carcinoma (HCC). To address this problem, we investigate the clinicopathological and prognostic value of MMP-1/PAR-1 signaling axis in HCC. Immunohistochemistry assay was used to determine the expression of MMP-1 and PAR-1 proteins in normal and HCC tissues. The correlations of MMP-1 and PAR-1 expression with clinicopathological parameters were assessed by Chisquared test. Patient survival and their differences were determined by Kaplan-Meier method and log-rank test. Cox regression was adopted for multivariate analysis of prognostic factors.MMP-1 and PAR-1 immunoreactivities were negative or low in normal liver tissues, but high in HCC tissues. PAR-1 expression was significantly correlated with that of MMP-1 (r=0.896, p<0.0001). The overexpression of MMP-1 and PAR-1 was significantly associated with recurrence, TNM staging and portal vein invasion of HCC. Patients with high MMP-1 and PAR-1 expression had significantly poorer overall survival (OS) (both P<0.001) and disease-free survival (DFS) (both P<0.001) when compared with patients with the low expression of MMP-1 and PAR-1. On multivariate analysis, MMP-1 and PAR-1 expression patterns were found to be independent prognostic factors for OS (both P<0.001) and DFS (both P<0.001). Our results suggest for the first time that the MMP-1/PAR-1 signaling axis might be applied as a novel marker for the prediction of recurrence and metastasis potency and a significant indicator of poor prognosis for patients with HCC. C1 [Liao, Mingmei] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan, China. [Tong, Ping] Central South University, State Key Laboratory of Medical GeneticsChangsha, Hunan, China. [Zhao, Jinfeng] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan, China. [Zhang, Yangde] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan, China. [Li, Zhehai] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan, China. [Wang, Jiwei] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan, China. [Feng, Xueping] Central South University, Xiangya Hospital, 410008 Changsha, Hunan, China. [Hu, Man] Central South University, Xiangya Hospital, 410008 Changsha, Hunan, China. [Pan, Yifeng] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan, China. RP Pan, Y (reprint author), Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, China. EM yifengp@163.com CR Farazi PA, DePinho RA, 2006, Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Canc 6:674–687 Thorgeirsson SS, Grisham JW, 2002, Molecular pathogenesis of human hepatocellular carcinoma. Nat Genet 31:339–346 Maheshwari S, Sarraj A, Kramer J et al, 2007, Oral contraception and the risk of hepatocellular carcinoma. J Hepatol 47:506–513 Fan ST, Mau Lo C, Poon RT et al, 2011, Continuous improvement of survival outcomes of resection of hepatocellular carcinoma: a 20-year experience. Ann Surg 253:745–758 Ko BS, Chang TC, Hsu C et al, 2011, Overexpression of 14-3-3ε predicts tumour metastasis and poor survival in hepatocellular carcinoma. Histopathology 58:705–711 Gerstein ES, Sini L, Ryabov AB et al, 2009, Comparative enzyme immunoassay of matrix metalloproteinases-2, -7, -9 and their tissue inhibitor-2 in tumors and plasma of patients with gastric cancer. Bull Exp Biol Med 148:899–902 Gialeli C, Theocharis AD, Karamanos NK, 2011, Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting. FEBS J 278:16–27 Schropfer A, Kammerer U, Kapp M et al, 2010, Expression pattern of matrix metalloproteinases in human gynecological cancer cell lines. BMC Canc 10:553 Nakopoulou L, Giannopoulou I, Lazaris ACh et al, 2003, The favorable prognostic impact of tissue inhibitor of matrix metalloproteinases-1 protein overexpression in breast cancer cells. APMIS 111:1027–1036 Li Y, Jin X, Kang S et al, 2006, Polymorphisms in the promoter regions of the matrix metalloproteinases-1, -3, -7, and -9 and the risk of epithelial ovarian cancer in China. Gynecol Oncol 101:92–96 Walker RA, Woolley DE, 1999, Immunolocalisation studies of matrix metalloproteinases-1, -2 and -3 in human melanoma. Virchows Arch 435:574–579 Mroczko B, Groblewska M, Lukaszewicz-Zajac M et al, 2009, Pre-treatment serum and plasma levels of matrix metalloproteinase 9, MMP-9, and tissue inhibitor of matrix metalloproteinases 1, TIMP- 1, in gastric cancer patients. Clin Chem Lab Med 47:1133–1139 Hilska M, Roberts PJ, Collan YU et al, 2007, Prognostic significance of matrix metalloproteinases-1, -2, -7 and -13 and tissue inhibitors of metalloproteinases-1, -2, -3 and -4 in colorectal cancer. Int J Canc 121:714–723 Yamashita K, Tanaka Y, Mimori K et al, 2004, Differential expression of MMP and uPA systems and prognostic relevance of their expression in esophageal squamous cell carcinoma. Int J Canc 110:201–207 Okazaki I, Wada N, Nakano M et al, 1997, Difference in gene expression for matrix metalloproteinase-1 between early and advanced hepatocellular carcinomas. Hepatology 25:580– 584 Matsunaga Y, Koda M, Murawaki Y, 2004, Expression of matrix metalloproiteinases, MMPs, and tissue inhibitors of metalloproteinases, TIMPs, in hepatocellular carcinoma tissue, compared with the surrounding non-tumor tissue. Res Commun Mol Pathol Pharmacol 115–116:143–150 Altadill A, Rodriguez M, Gonzalez LO et al, 2009, Liver expression of matrix metalloproteases and their inhibitors in hepatocellular carcinoma. Dig Liver Dis 41:740–748 Nierodzik ML, Karpatkin S, 2006, Thrombin induces tumor growth, metastasis, and angiogenesis: evidence for a thrombinregulated dormant tumor phenotype. Canc Cell 10:355–362 Chambers RC, Dabbagh K, McAnulty RJ et al, 1998, Thrombin stimulates fibroblast procollagen production via proteolytic activation of protease-activated receptor 1. Biochem J 333:121–127 Fujimoto D, Hirono Y, Goi T et al, 2008, Prognostic value of protease-activated receptor-1, PAR-1, and matrix metalloproteinase- 1, MMP-1, in gastric cancer. Anticancer Res 28:847–854 Elste AP, Petersen I, 2010, Expression of proteinase-activated receptor 1–4, PAR 1–4, in human cancer. J Mol Histol 41:89–99 Gao L, Smith RS, Chen LM et al, 2010, Tissue kallikrein promotes prostate cancer cell migration and invasion via a protease-activated receptor-1-dependent signaling pathway. Biol Chem 391:803–812 Hatfield KJ, Reikvam H, Bruserud O, 2010, The crosstalk between the matrix metalloprotease system and the chemokine network in acute myeloid leukemia. Curr Med Chem 17:4448–4461 Grimm M, Lazariotou M, Kircher S et al, 2010, MMP-1 is a, pre-)invasive factor in Barrett-associated esophageal adenocarcinomas and is associated with positive lymph node status. J Transl Med 8:99 Fang WL, Liang WB, He H et al, 2010, Association of matrix metalloproteinases 1, 7, and 9 gene polymorphisms with genetic susceptibility to colorectal carcinoma in a Han Chinese population. DNA Cell Biol 29:657–661 Garcia-Lopez MT, Gutierrez-Rodriguez M, Herranz R, 2010, Thrombin-activated receptors: promising targets for cancer therapy? Curr Med Chem 17:109–128 Boire A, Covic L, Agarwal A et al, 2005, PAR1 is a matrix metalloprotease-1 receptor that promotes invasion and tumorigenesis of breast cancer cells. Cell 120:303–313 Blackburn JS, Liu I, Coon CI et al, 2009, A matrix metalloproteinase-1/protease activated receptor-1 signaling axis promotes melanoma invasion and metastasis. Oncogene 28:4237–4248 Zhang Y, Zhan H, Xu W et al, 2011, Upregulation of matrix metalloproteinase-1 and proteinase-activated receptor-1 promotes the progression of human gliomas. Pathol Res Pract 207:24–29 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 397 EP 403 DI 10.1007/s12253-011-9458-8 PG 7 ER PT J AU Rajalingam, K Sugunadevi, G Arokia Vijayaanand, M Kalaimathi, J Suresh, K AF Rajalingam, Kasinathan Sugunadevi, Govindasamy Arokia Vijayaanand, Mariadoss Kalaimathi, Janakiraman Suresh, Kathiresan TI Anti-Tumour and Anti-Oxidative Potential of Diosgenin against 7, 12-Dimethylbenz(a)anthracene Induced Experimental Oral Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Antioxidants; Lipid peroxidation; Diosgenin; DMBA; Histopathology ID Antioxidants; Lipid peroxidation; Diosgenin; DMBA; Histopathology AB The ultimate aim of the present study was to exploring the chemopreventive efficacy of diosgenin on 7,12-dimethylbenz(a)anthracene (DMBA) induced hamster buccal pouch carcinogenesis. The chemopreventive potential of diosgenin was evaluated by measuring the tumour incidence, tumour volume and tumour burden as well as analyzing the activities of detoxification agents, levels of lipid peroxidation byproducts and antioxidants status by specific colorimetric methods. Oral squamous cell carcinoma (OSCC) was developed in the buccal pouches of male Syrian golden hamsters by painting with 0.5% DMBA in liquid paraffin, thrice a week for 16 weeks. DMBA painted animals were indicating the morphological changes as depicted as hyperplasia, dysplasia and well-developed squamous cell carcinoma. Moreover, antioxidants and lipid peroxidation byproducts levels were drastically altered in DMBA painted hamsters. Oral administration of diosgenin (80 mg/kg bw) to DMBA painted hamsters on alternate days for 16 weeks significantly reduced the formation of oral tumour and normalized the above biochemical abnormalities. We conclude that the diosgenin is probably potent chemopreventive agent due to their antioxidant function in DMBA induced hamster buccal pouch carcinogenesis. C1 [Rajalingam, Kasinathan] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608 002 Tamil Nadu, India. [Sugunadevi, Govindasamy] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608 002 Tamil Nadu, India. [Arokia Vijayaanand, Mariadoss] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608 002 Tamil Nadu, India. [Kalaimathi, Janakiraman] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608 002 Tamil Nadu, India. [Suresh, Kathiresan] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608 002 Tamil Nadu, India. RP Suresh, K (reprint author), Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, 608 002 Tamil Nadu, India. EM suraj_cks@yahoo.co.in CR Kademani D, 2007, Oral cancer. Mayo Clin Proc 82:878–887 Warnakulasuriya S, 2009, Global epidemiology of oral and oropharyngeal cancer. Oral Oncol 45:309–316 Nair V, Bartsch H, Nair J, 2004, Alert for an epidemic of oral cancer due to use of the betel quid substitutes gutkha and pan masala: a review of agents and causative mechanism. Mutagenesis 19:251–262 Raimondi A, Cabrini R, Itoiz ME, 2005, Ploidy analysis of field cancerization and cancer development in the hamster cheek pouch carcinogenesis model. J Oral Pathol Med 34:227– 231 Gimenez-Conti IB, Slaga TJ, 1993, The hamster cheek pouch carcinogenesis. J Cell Biochem 17:83–90 Boring A, Squire TS, Tong T, 1994, Cancer statistics. Calif Cancer J Clin 44:07–26 Gillison ML, 2007, Current topics in the epidemiology of oral cavity and oropharyngeal cancers. Head Neck 29:779–792 Shimada T, 2006, Xenobiotic-metabolising enzymes involved in activation and detoxification of carcinogenic polycyclic aromatic hydrocarbons. Drug Metab Pharmacokinet 21:257– 276 Hecht SS, 2003, Tobacco carcinogens, their biomarker and tobacco-induced cancer. Nat Rev Cancer 3:733–744 Broquist HP, 1991, An experimental, tool to induce glutathione deficiency: elucidation of glutathione and ascorbate in their role as antioxidants. Nutr Rev 50:110–111 Ames BN, ShigenagaMK, Hagen TM(1993, Oxidants, antioxidants and the degenerative diseases of aging. Proc Natl Acad Sci USA 90:7915–7922 Manoharan S, Shreeram S, Nagini S, 1996, Life style can induce lipid peroxidation and compromise of antioxidant defense mechanisms in the erythrocytes of oral cancer patients. Med Sci Res 24:397–400 Li J, Liu X, Guo M et al, 2005, Electrochemical study of breast cancer cells MCF-7 and its application in evaluating the effect of diosgenin. Anal Scis 21:561–564 Raju J, Patlolla JM, Swamy MV et al, 2004, Diosgenin, a steroid saponin of trigonella foenum graecum, fenugreek), inhibits azoxymethane-induced aberrant crypt foci formation in F344 rats and induces apoptosis in HT-29 human colon cancer cells. Cancer Epidemiol Biomark Prev 13:1392–1398 Moalic S, Liagre B, Corbiere C et al, 2001, A plant steroid diosgenin, induces apoptosis, cell cycle arrest and COX activity in osteosarcoma cells. FEBS Lett 501:225–230 Corbiere C, Liagre B, Jerro F et al, 2004, Induction of antiproliferative effect by diosgenin through activation of P53, release of apoptosis-inducing factor(AIF), and modulation of caspase-3 activity in different human cancer cells. Cell Res 14:188–196 Au AL, Kwok CC, Lee AT et al, 2004, Activition of iberiotoxinsensitive, Ca2+-activated K+ channels of porcine isolated left anterior descending coronary artery by diosgenin. Eur J Pharmacol 502:123–133 Ohkawa H, Ohishi N, Yagi K, 1979, Assay for lipid peroxidation in animals tissues by thiobarbituric acid reaction. Anal Biochem 95:351–358 Yagi K, 1978, Lipid peroxides and human disease. Chem Physiol Lip 45:337–351 Kakkar P, Das B, Viswanathan P, 1984, A modified spectrophotometric assay of superoxide dismutase. Ind J Biochem Biophys 21:130– 132 Sinha KA, 1972, Colorimetric assay of catalase. Anal Biochem 47:389–394 Rotruck JT, Pope AL, Ganther HE, 1984, Selenium: biochemical roles as a component of glutathione peroxidase. Science 179:588–590 Beutler E, Kelly BM, 1963, The effect of sodium nitrate on RBC glutathione. Experimentia 19:96–97 Habig WH, Pabst MJ, Jakoby WB, 1974, Glutathione-Stransferase, the first enzymatic step in mercapturic acid formation. J Biol Chem 249:7130–7139 Carlberg I, Mannervik B, 1985, Glutathione reductase. Methods Enzymol. Academic Press New York 113: 484–490 Omaye ST, Turnbull TD, Sauberlish HC, 1979, Selected method for the determination of ascorbic acid in animal’s cells, tissues and fluids. Methods Enzymol, Academic Press New York 62: 03–11 Desai FD, 1984, Vitamin E analysis methods for animal’s tissue. Methods Enzymol Academic Press New York 105: 138–145 Palan PR, Mikhail BS, Basin J, 1973, Plasma levels of antioxidant beta-carotene and tocopherol in uterine cervix dysplasia and cancer. Nutr Cancer 15:13–20 Mann KK, Matulka RA, Hahn ME, 1999, The role of polycyclic aromatic hydrocarbon metabolism in 7,12-dimethylbenz(a)anthracene- induced pre-B lymphocyte apoptosis. Toxicol Appl Pharmacol 161:10–12 Chen YK, Hsue SS, Lin LM, 2003, Immunohistochemical demonstration of p63 in DMBA-induced hamster buccal pouch squamous cell carcinogenesis. Oral Dis 9:235–240 Patel KJ, Desiva HL, Tong DC et al, 2011, Concordance between clinical and histopathological diagnosis of oral mucosal lesions. J Oral Maxillofac Surg 69:125–133 Gottlieb CH, Tomilnson IP, 2005, Mitochondrial tumour suppressors: a genetic and biochemical update. Nat Rev Cancer 5:857–866 Ruzza P, Rosato A, Rossi CR et al, 2009, Glutathione transferases as targets for cancer therapy. Med Chem 9:763–777 Yang Y, Zhou ZI, Ge JP, 2006, Effect of genistein on DMBAinduced oral carcinogenesis in hamster. Carcinogenesis 27:578–583 Kawanishi Y, Hiraku S, 2006, Oxidative and nitrative DNA damage as biomarker for carcinogenesis with special reference of inflammation. Antioxid Redox Signal 8:1047–1058 Burcham PC, 1998, Genotoxic lipid peroxidation products: their DNA damaging properties and role in formation of endogenous DNA adducts. Mutagenesis 13:287–305 Schwartz JL, Shklar G, 1997, Verification in syngeneic hamster of in vitro transformation of hamster oral mucosa by 7, 12-dimethyl benz(a)anthracene. Eur J Cancer 33:431–438 Diplock AT, Rice-Evans AC, Burton RH, 1994, Is there a significant role for lipid peroxidation in the causation of malignancy and for antioxidants in cancer prevention. Cancer Res 54:1952–1956 Chiba I, 2001, Prevention of betel quid chewer’s oral cancer in the Asian pacific area. Asian Pacific J Cancer 2:263–269 Molins DC, 1993, Identification of hydroxyl radical lesions in DNA base structure: biomarkers with a putative link to cancer developments. J Toxical Environ Health 40:247–261 Das UN, 2002, A radical approch to cancer. Med Sci Monit 8:79–92 Oberley TD, 2002, Oxidative damage and cancer. Am J Pathol 160:403–408 Abiaka C, Al-Awadi F, Al-Sayer H et al, 2002, Activities of erythrocyte antioxidant enzymes in cancer patients. J Clin Lab Anal 16:167–171 Gago-Dominquez M, Castelao JE, Pike MC, 2005, Role of lipid peroxidation in the epidemiology and prevention of breast cancer. Cancer Epidemiol Biomark Prev 14:2829–2839 Rice-Evans C, Burdon R, 1993, Free radical—lipid interactions and pathological consequences. Prog Lipid Res 32:71–110 Choi SW, Benzie IFF, Collins AR, 2004, Vitamin C and E: acute interactive effects on biomarkers of antioxidant defence and oxidative stress. Mutt Res 551:109–117 Mates JM, Segura JA, Alonso FT et al, 2008, Intracellular redox status and oxidative: implications of cell proliferation, apoptosis, and carcinogenesis. Arch Toxicol 82:273–299 Zaidi SMKR, Banu N, 2004, Antioxidant potential of vitamin A, E and C in modulating oxidative stress in rat brain. Clin Chim Acta 340:229–233 Shklar G, Schwartz JL, 1996, Vitamin E inhibits experimental carcinogenesis and tumour angiogenesis. Eur J Cancer B Oral Oncol 32:114–119 Zou W, Yue P, Lin N, 2006, Vitamin C inactivates the proteosome inhibitor PS-341 in human cancer cells. Clin Cancer Res 12:273–280 McLellan J, Wolf CR, 1999, Glutathione and glutathionedependant enzyme in cancer drug resistance. Drug Resist Update 2:153–164 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 405 EP 412 DI 10.1007/s12253-011-9460-1 PG 8 ER PT J AU Varga, I Hutoczki, G Szemcsak, DCs Zahuczky, G Toth, J Adamecz, Zs Kenyeres, A Bognar, L Hanzely, Z Klekner, AF Varga, Imre Hutoczki, Gabor Szemcsak, D Csaba Zahuczky, Gabor Toth, Judit Adamecz, Zsolt Kenyeres, Annamaria Bognar, Laszlo Hanzely, Zoltan Klekner, Almos TI Brevican, Neurocan, Tenascin-C and Versican are Mainly Responsible for the Invasiveness of Low-Grade Astrocytoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Astrocytoma; Brain metastasis; Schwannoma; Normal brain; Extracellular matrix; Invasion ID Astrocytoma; Brain metastasis; Schwannoma; Normal brain; Extracellular matrix; Invasion AB The extent of tumor removal determines the effectiveness of postoperative oncotherapy. This is especially true for primary brain tumors, where peritumoral invasion usually makes radical resection impossible. The aim of the study was to determinate the specific expression pattern of invasion related molecules of different intracranial tumors and to identify molecules that are principally responsible for the peritumoral invasiveness of grade II astrocytoma mRNA expression of 26 extracellular matrix (ECM) molecules was determined in tissue samples from grade II astrocytoma, schwannoma, intracerebral metastases of non-small cell lung cancer and normal brain. Immunohistochemical staining for brevican, neurocan, tenascin-C and versican was also performed for each tumor group. Comparing astrocytoma to metastasis, schwannoma and normal brain; and metastasis and schwannoma to normal brain, 22, 17, 20, 21, and 19 molecules, respectively, were found to be significantly overexpressed at the mRNA level. Cluster analysis of mRNA expression showed a specific gene expression pattern for each histological group. Four molecules of 26 were found to be associated to astrocytoma. Immunohistochemical staining confirmed the results of the mRNA analysis at the protein level. Tumors of different origin have a specific invasive phenotype that can evidently determinate on gene expression level. This characteristic expression pattern of the invasion-related molecules might help to screen exact targets for anti-invasion drugs. In case of low-grade astrocytoma. brevican, neurocan, tenascin-C and versican were found to correlate principally with the invasive phenotype of low-grade astrocytoma, thus these molecules can potentially serve as targets for anti-invasion therapy in the future. C1 [Varga, Imre] Kenezy Gyula Hospital, Department of PulmonologyDebrecen, Hungary. [Hutoczki, Gabor] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98., 4032 Debrecen, Hungary. [Szemcsak, D Csaba] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98., 4032 Debrecen, Hungary. [Zahuczky, Gabor] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Toth, Judit] University of Debrecen, Department of OncologyDebrecen, Hungary. [Adamecz, Zsolt] University of Debrecen, Medical and Health Science Center, Institute of Nuclear MedicineDebrecen, Hungary. [Kenyeres, Annamaria] University of Medicine, The Institute of Anatomy, Histology and EmbryologyDebrecen, Hungary. [Bognar, Laszlo] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98., 4032 Debrecen, Hungary. [Hanzely, Zoltan] Orszagos Pszichiatriai es Neurologiai IntezetBudapest, Hungary. [Klekner, Almos] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98., 4032 Debrecen, Hungary. RP Klekner, (reprint author), University of Debrecen, Clinical Center, Department of Neurosurgery, 4032 Debrecen, Hungary. EM aklekner@yahoo.com CR Klekner A, Varga I, Bognar L et al, 2010, Extracellular matrix of cerebral tumors with different invasiveness. Ideggyogy Sz 63(1– 2):38–43, Hu) Varga I, Hutoczki G, Petras M et al, 2010, Expression of invasion-related extracellular matrix molecules in human glioblastoma versus intracerebral lung adenocarcinoma metastasis. Cen Eur Neurosurg 71(4):173–180 Kyoshima K, Akaishi K, Tokushige K et al, 2004, Surgical experience with resection en bloc of intramedullary astrocytomas and ependymomas in the cervical and cervicothoracic region. J Clin Neurosci 11(6):623–628 Ramina R, Coelho Neto M, Fernandes YB et al, 2005, Intrinsic tectal low grade astrocytomas: is surgical removal an alternative treatment? Long-term outcome of eight cases. Arq Neuropsiquiatr 63(1):40–45 Abacioglu U, Caglar H, Atasoy BM et al, 2010, Gamma knife radiosurgery in non small cell lung cancer patients with brain metastases: treatment results and prognostic factors. J BUON 15, 2):274–280 Catinella FP, Kittle CF, Faber LP et al, 1989, Surgical treatment of primary lung cancer and solitary intracranial metastasis. Chest 136(5 Suppl):e30 Okada Y, 2000, Tumor cell-matrix interaction: pericellular matrix degradation and metastasis. Verh Dtsch Ges Pathol 84:33–42 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), method. Methods 25:402–408 Han S, Sidell N, Roman J, 2005, Fibronectin stimulates human lung carcinoma cell proliferation by suppressing p21 gene expression via signals involving Erk and Rho kinase. Canc Lett 219(1):71–81 Hirose J, Kawashima H, Yoshie O et al, 2001, Versican interacts with chemokines and modulates cellular responses. J Biol Chem 276(7):5228–5234 Sainte-Marie G, 1962, A paraffin embedding technique for studies employing immunofluorescence. J Histochem Cytochem 10:250–256 Tuckett F, Morriss-Kay G, 1988, Alcian blue staining of glycosaminoglycans in embryonic material: effect of different fixatives. Histochem J 20:174–182 Kono K, Inoue Y, Nakayama K et al, 2001, The role of diffusionweighted imaging in patients with brain tumors. AJNR Am J Neuroradiol 22(6):1081–1088 Lu S, Ahn D, Johnson G et al, 2004, Diffusion-tensor MR imaging of intracranial neoplasia and associated peritumoral edema: introduction of the tumor infiltration index. Radiology 232(1):221–228 Miklos P, Gabor H, Imre V et al, 2009, Expression pattern of invasion-related molecules in brain tumors of different origin. Magy Onkol 53(3):253–258, Hu) Wang W, Steward CE, Desmond PM, 2009, Diffusion tensor imaging in glioblastoma multiforme and brain metastases: the role of p, q, L, and fractional anisotropy. AJNR Am J Neuroradiol 30, 1):203–208 Lo CK, Yu CH,Ma CC et al, 2010, Surgical management of primary non-small-cell carcinoma of lung with synchronous solitary brain metastasis: local experience. Hong Kong Med J 16(3):186–191 Pfannschmidt J, Dienemann H, 2010, Surgical treatment of oligometastatic non-small cell lung cancer. Lung Canc 69, 3):251–258 Hu B, Kong LL, Matthews RT et al, 2008, The proteoglycan brevican binds to fibronectin after proteolytic cleavage and promotes glioma cell motility. J Biol Chem 283(36):24848– 24859 Viapiano MS, Bi WL, Piepmeier J et al, 2005, Novel tumorspecific isoforms of BEHAB/brevican identified in human malignant gliomas. Canc Res 65(15):6726–6733 Viapiano MS, Hockfield S, Matthews RT, 2008, BEHAB/ brevican requires ADAMTS-mediated proteolytic cleavage to promote glioma invasion. J Neurooncol 88(3):261–272 Viapiano MS, Matthews RT, Hockfield S, 2003, A novel membrane-associated glycovariant of BEHAB/brevican is upregulated during rat brain development and in a rat model of invasive glioma. J Biol Chem 278(35):33239–33247 Nutt CL, Matthews RT, Hockfield S, 2001, Glial tumor invasion: a role for the upregulation and cleavage of BEHAB/brevican. Neuroscientist 7(2):113–122 Gary SC, Hockfield S, 2000, BEHAB/brevican: an extracellular matrix component associated with invasive glioma. Clin Neurosurg 47:72–82 Gary SC, Kelly GM, Hockfield S, 1998, BEHAB/brevican: a brain-specific lectican implicated in gliomas and glial cell motility. Curr Opin Neurobiol 8(5):576–581 Gary SC, Zerillo CA, Chiang VL et al, 2000, cDNA cloning, chromosomal localization, and expression analysis of human BEHAB/brevican, a brain specific proteoglycan regulated during cortical development and in glioma. Gene 256(1–2):139–147 Talts U, Kuhn U, Roos G et al, 2000, Modulation of extracellular matrix adhesiveness by neurocan and identification of its molecular basis. Exp Cell Res 259(2):378–388 Akita K, Toda M, Hosoki Y et al, 2004, Heparan sulphate proteoglycans interact with neurocan and promote neurite outgrowth from cerebellar granule cells. Biochem J 383(Pt 1):129– 138 Rauch U, Feng K, Zhou XH, 2001, Neurocan: a brain chondroitin sulfate proteoglycan. Cell Mol Life Sci 58(12–13):1842–1856 Zinovieva E, Lebrun N, Letourneur F et al, 2008, Lack of association between Tenascin-C gene and spondyloarthritis. Rheumatology, Oxford, 47(11):1655–1658 Meloty-Kapella CV, Degen M, Chiquet-Ehrismann R et al, 2006, Avian tenascin-W: expression in smooth muscle and bone, and effects on calvarial cell spreading and adhesion in vitro. Dev Dyn 235(6):1532–1542 Pajala A, Melkko J, Leppilahti J et al, 2009, Tenascin-C and type I and III collagen expression in total Achilles tendon rupture. Histol Histopathol 24(10):1207–1211 Engel J, 1989, EGF-like domains in extracellular matrix proteins: localized signals for growth and differentiation? FEBS Lett 251, 1–2):1–7 Swindle CS, Tran KT, Johnson TD et al, 2001, Epidermal growth factor, EGF)-like repeats of human tenascin-C as ligands for EGF receptor. J Cell Biol 154(2):459–468 Fischer D, Brown-Ludi M, Schulthess T et al, 1997, Concerted action of tenascin-C domains in cell adhesion, anti-adhesion and promotion of neurite outgrowth. J Cell Sci 110(Pt 13):1513–1522 Hirata E, Arakawa Y, Shirahata M et al, 2009, Endogenous tenascin-C enhances glioblastoma invasion with reactive change of surrounding brain tissue. Canc Sci 100(8):1451–1459 Maris C, Rorive S, Sandras F et al, 2008, Tenascin-C expression relates to clinicopathological features in pilocytic and diffuse astrocytomas. Neuropathol Appl Neurobiol 34(3):316–329 Bicer A, Guclu B, Ozkan A et al, 2010, Expressions of angiogenesis associated matrix metalloproteinases and extracellular matrix proteins in cerebral vascular malformations. J Clin Neurosci 17(2):232–236 Kim CH, Bak KH, Kim YS et al, 2000, Expression of tenascin-C in astrocytic tumors: its relevance to proliferation and angiogenesis. Surg Neurol 54(3):235–240 Leins A, Riva P, Lindstedt R et al, 2003, Expression of tenascin-C in various human brain tumors and its relevance for survival in patients with astrocytoma. Cancer 98(11):2430–2439 Zagzag D, Capo V, 2002, Angiogenesis in the central nervous system: a role for vascular endothelial growth factor/vascular permeability factor and tenascin-C. Common molecular effectors in cerebral neoplastic and non-neoplastic “angiogenic diseases”. Histol Histopathol 17(1):301–321 Mukaratirwa S, Chimonyo M, Obwolo M et al, 2004, Stromal cells and extracellular matrix components in spontaneous canine transmissible venereal tumour at different stages of growth. Histol Histopathol 19(4):1117–1123 Xiang YY, Dong H, Wan Y et al, 2006, Versican G3 domain regulates neurite growth and synaptic transmission of hippocampal neurons by activation of epidermal growth factor receptor. J Biol Chem 281(28):19358–19368 Wu Y, Sheng W, Chen L et al, 2004, Versican V1 isoform induces neuronal differentiation and promotes neurite outgrowth. Mol Biol Cell 15(5):2093–2104 Ricciardelli C, Sakko AJ, Ween MP et al, 2009, The biological role and regulation of versican levels in cancer. Canc Metastasis Rev 28(1–2):233–245 Wu Y, Zhang Y, Cao L et al, 2001, Identification of the motif in versican G3 domain that plays a dominant-negative effect on astrocytoma cell proliferation through inhibiting versican secretion and binding. J Biol Chem 276(17):14178–14186 Zheng PS, Wen J, Ang LC et al, 2004, Versican/PG-M G3 domain promotes tumor growth and angiogenesis. FASEB J 18, 6):754–756 Soltermann A, Tischler V, Arbogast S, 2008, Prognostic significance of epithelial-mesenchymal and mesenchymal-epithelial transition protein expression in non-small cell lung cancer. Clin Canc Res 14(22):7430–7437 Pirinen R, Leinonen T, Bohm J et al, 2005, Versican in nonsmall cell lung cancer: relation to hyaluronan, clinicopathologic factors, and prognosis. Hum Pathol 36(1):44–50 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 413 EP 420 DI 10.1007/s12253-011-9461-0 PG 8 ER PT J AU Kelemen, Gy Varga, Z Lazar, Gy Thurzo, L Kahan, Zs AF Kelemen, Gyongyi Varga, Zoltan Lazar, Gyorgy Thurzo, Laszlo Kahan, Zsuzsanna TI Cosmetic Outcome 1–5 Years After Breast Conservative Surgery, Irradiation and Systemic Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast-conserving therapy; Cosmesis; Fibrosis; Radiotherapy dose; Irradiated volume ID Breast-conserving therapy; Cosmesis; Fibrosis; Radiotherapy dose; Irradiated volume AB The late side-effects of the local therapy of early breast cancer depend on many patient- and therapy-related parameters.We aimed at investigating the factors that influence the cosmetic and functional outcomes among our breast cancer patients after breast-conserving surgery and conformal radiotherapy, with or without adjuvant systemic therapy. A study was made of the association of the cosmetic outcome after a median follow-up time of 2.4 years and the clinical data on 198 patients extracted from a prospectively compiled database. Breast tenderness occurred more frequently among patients ≤50 years old (p<0.05). Long-term side effects were related to radiotherapy-related factors the most, while no effect of the systemic therapy could be detected. The risk of hyperpigmentation, breast edema and breast fibrosis increased by 18%, 23% and 7%, respectively for every 100 cm3 increase in the irradiated breast volume, while that of breast edema and breast fibrosis increased by 21% and 12%, respectively for every 10 cm3 increase in the boost volume. Patients who received a photon boost were significantly more likely to develop breast edema and fibrosis than those who received electrons (p<0.005). Dose inhomogeneity was related to the volume of the irradiated breast (p=0.037). Dyspigmentation developed more often among patients older than 50 years, while smoking favoured both dyspigmentation and teleangiectasia. Breast edema was related to dyspigmentation (p=0.003), fibrosis (p<0.001) and breast asymmetry (p=0.032), whereas none of these abnormalities were associated with teleangiectasia. Body image changes were more frequent at a younger age (p<0.005), while the need to change clothing habits occurred more often at an older age (p<0.05). Radiotherapy-related parameters appear to exert the greatest effect on the overall cosmetic outcome after breast-conserving surgery and postoperative radiotherapy. C1 [Kelemen, Gyongyi] University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. [Varga, Zoltan] University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. [Lazar, Gyorgy] University of Szeged, Department of SurgerySzeged, Hungary. [Thurzo, Laszlo] University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. RP Kahan, Zs (reprint author), University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. EM kahan@onko.szote.u-szeged.hu CR Voogd AC, Nielsen M, Peterse JL, Blichert-Toft M, Bartelink H, Overgaard M et al, 2001, Differences in risk factors for local and distant recurrence after breast-conserving therapy or mastectomy for stage I and II breast cancer: pooled results of two large European randomized trials. J Clin Oncol 19:1688– 1697 Fisher B, Anderson S, Bryant J, Margolese RG, Deutsch M, Fisher ER et al, 2002, Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 347:1233–1241 Veronesi U, Cascinelli N, Mariani L, Greco M, Saccozzi R, Luini A et al, 2002, Twenty-year follow-up of a randomized study comparing breast-conserving surgery with radical mastectomy for early breast cancer. N Engl J Med 347:1227–1232 Offersen BV, Overgaard M, Kroman N, Overgaard J, 2009, Accelerated partial breast irradiation as part of breast conserving therapy of early breast carcinoma: a systematic review. Radiother Oncol 90:1–13 Fyles AW, McCready DR, Manchul LA, Trudeau ME, Merante P, Pintilie M et al, 2004, Tamoxifen with or without breast irradiation in women 50 years of age or older with early breast cancer. N Engl J Med 351:963–970 Fowble B, Fein DA, Hanlon AL, Eisenberg BL, Hoffman JP, Sigurdson ER et al, 1996, The impact of tamoxifen on breast recurrence, cosmesis, complications, and survival in estrogen receptor-positive early-stage breast cancer. Int J Radiat Oncol Biol Phys 35:669–677 Munshi A, Kakkar S, Bhutani R, Jalali R, Budrukkar A, Dinshaw KA, 2009, Factors influencing cosmetic outcome in breast conservation. Clin Oncol, R Coll Radiol, 21:285–293 Azria D, Gourgou S, Sozzi WJ, Zouhair A, Mirimanoff RO, Kramar A et al, 2004, Concomitant use of tamoxifen with radiotherapy enhances subcutaneous breast fibrosis in hypersensitive patients. Br J Cancer 91:1251–1260 Azria D, Betz M, Bourgier C, Sozzi WJ, Ozsahin M, 2010, Identifying patients at risk for late radiation-induced toxicity. Crit Rev Oncol Hematol, Epub ahead of print Azria D, Belkacemi Y, Romieu G, Gourgou S, Gutowski M, Zaman K et al, 2010, Concurrent or sequential adjuvant letrozole and radiotherapy after conservative surgery for early-stage breast cancer, CO-HO-RT): a phase 2 randomised trial. Lancet Oncol 11:258–265 Bartelink H, Horiot JC, Poortmans PM, Struikmans H, Van den Bogaert W, Fourquet A et al, 2007, Impact of a higher radiation dose on local control and survival in breast-conserving therapy of early breast cancer: 10-year results of the randomized boost versus no boost EORTC 22881-10882 trial. J Clin Oncol 25:3259–3265 Wazer DE, Morr J, Erban JK, Schmid CH, Ruthazer R, Schmidt-Ullrich RK, 1997, The effects of postradiation treatment with tamoxifen on local control and cosmetic outcome in the conservatively treated breast. Cancer 80:732–740 Collette S, Collette L, Budiharto T, Horiot JC, Poortmans PM, Struikmans H et al, 2008, Predictors of the risk of fibrosis at 10 years after breast conserving therapy for early breast cancer: a study based on the EORTC Trial 22881-10882 ‘boost versus no boost’. Eur J Cancer 44:2587–2599 Taylor ME, Perez CA, Halverson KJ, Kuske RR, Philpott GW, Garcia DM et al, 1995, Factors influencing cosmetic results after conservation therapy for breast cancer. Int J Radiat Oncol Biol Phys 31:753–764 Johansen J, Overgaard J, Rose C, Engelholm SA, Gadeberg CC, Kjaer M et al, 2002, Cosmetic outcome and breast morbidity in breast-conserving treatment—results from the Danish DBCG- 82TM national randomized trial in breast cancer. Acta Oncol 41:369–380 Curran D, van Dongen JP, Aaronson NK, Kiebert G, Fentiman IS, Mignolet F et al, 1998, Quality of life of early-stage breast cancer patients treated with radical mastectomy or breast-conserving procedures: results of EORTC Trial 10801. The European Organization for Research and Treatment of Cancer, EORTC), Breast Cancer Cooperative Group, BCCG). Eur J Cancer 34:307–314 Cardoso MJ, Cardoso J, Santos AC, Vrieling C, Christie D, Liljegren G et al, 2007, Factors determining esthetic outcome after breast cancer conservative treatment. Breast J 13:140–146 Johansen J, Overgaard J, Overgaard M, 2007, Effect of adjuvant systemic treatment on cosmetic outcome and late normal-tissue reactions after breast conservation. Acta Oncol 46:525–533 Kahan Z, Csenki M, Varga Z, Szil E, Cserhati A, Balogh A et al, 2007, The risk of early and late lung sequelae after conformal radiotherapy in breast cancer patients. Int J Radiat Oncol Biol Phys 68:673–681 Van Limbergen E, Rijnders A, van der Schueren E, Lerut T, Christiaens R, 1989, Cosmetic evaluation of breast conserving treatment for mammary cancer. 2. A quantitative analysis of the influence of radiation dose, fractionation schedules and surgical treatment techniques on cosmetic results. Radiother Oncol 16:253–267 Borger JH, Kemperman H, Smitt HS, Hart A, van Dongen J, Lebesque J et al, 1994, Dose and volume effects on fibrosis after breast conservation therapy. Int J Radiat Oncol Biol Phys 30:1073–1081 Wazer DE, DiPetrillo T, Schmidt-Ullrich R, Weld L, Smith TJ, Marchant DJ et al, 1992, Factors influencing cosmetic outcome and complication risk after conservative surgery and radiotherapy for early-stage breast carcinoma. J Clin Oncol 10:356–363 Pezner RD, Patterson MP, Hill LR, Vora NL, Desai KR, Lipsett JA, 1985, Breast retraction assessment. Multiple variable analysis of factors responsible for cosmetic retraction in patients treated conservatively for stage I or II breast carcinoma. Acta Radiol Oncol 24:327–330 Lilla C, Ambrosone CB, Kropp S, Helmbold I, Schmezer P, von Fournier D et al, 2007, Predictive factors for late normal tissue complications following radiotherapy for breast cancer. Breast Cancer Res Treat 106:143–150 Porta EA, 2002, Pigments in aging: an overview. Ann N Y Acad Sci 959:57–65 Moody AM, Mayles WP, Bliss JM, A’Hern RP, Owen JR, Regan J et al, 1994, The influence of breast size on late radiation effects and association with radiotherapy dose inhomogeneity. Radiother Oncol 33:106–112 Murphy C, Anderson PR, Li T, Bleicher RJ, Sigurdson ER, Goldstein LJ et al, 2010, Impact of the radiation boost on outcomes after breast-conserving surgery and radiation. Int J Radiat Oncol Biol Phys Epub ahead of print Ellenbogen R, Jankauskas S, Collini FJ, 1990, Achieving standardized photographs in aesthetic surgery. Plast Reconstr Surg 86:955–961 Sarin R, Dinshaw KA, Shrivastava SK, Sharma V, Deore SM, 1993, Therapeutic factors influencing the cosmetic outcome and late complications in the conservative management of early breast cancer. Int J Radiat Oncol Biol Phys 27:285–292 Hill-Kayser CE, Chacko D, Hwang WT, Vapiwala N, Solin LJ, 2010, Long-term clinical and cosmetic outcomes after breast conservation treatment for women with early-stage breast carcinoma according to the type of breast boost. Int J Radiat Oncol Biol Phys, Epub ahead of print Harsolia A, Kestin L, Grills I, Wallace M, Jolly S, Jones C et al, 2007, Intensity-modulated radiotherapy results in significant decrease in clinical toxicities compared with conventional wedge-based breast radiotherapy. Int J Radiat Oncol Biol Phys 68:1375–1380 McDonald MW, Godette KD, Whitaker DJ, Davis LW, Johnstone PA, 2010, Three-year outcomes of breast intensity-modulated radiation therapy with simultaneous integrated boost. Int J Radiat Oncol Biol Phys 77:523–530 Abner AL, Recht A, Vicini FA, Silver B, Hayes D, Come S et al, 1991, Cosmetic results after surgery, chemotherapy, and radiation therapy for early breast cancer. Int J Radiat Oncol Biol Phys 21:331–338 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 421 EP 427 DI 10.1007/s12253-011-9462-z PG 7 ER PT J AU Toth, L Andras, Cs Molnar, Cs Tanyi, M Csiki, Z Molnar, P Szanto, J AF Toth, Laszlo Andras, Csilla Molnar, Csaba Tanyi, Miklos Csiki, Zoltan Molnar, Peter Szanto, Janos TI Investigation of β-catenin and E-cadherin Expression in Dukes B2 Stage Colorectal Cancer with Tissue Microarray Method. Is It a Marker of Metastatic Potential in Rectal Cancer? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE β-catenin and E cadherin; Colorectal cancer; TMA; Metastasis ID β-catenin and E cadherin; Colorectal cancer; TMA; Metastasis AB β-catenin and E cadherin are both membraneassociated proteins which are essential regulators and providers of cellular adhesion. In the metastatic cascade of malignant tumours, detachment of tumour cells from each other is a very important step. It has been shown in several tumour types, that reduced expression of these proteins is important. The aim of our study was to clarify the expression profile of these proteins, and correlate the findings with the metastasizing potential of early stage colon and rectal cancers. Formalin fixed and paraffin embedded samples from 79 Dukes B2 stage colorectal cancer were examined using a tissue microarray approach. The expression of β-catenin and E-cadherin proteinswas determined immunohistochemically. Our findings indicated that there is a tendency for metastatic spread in cases when membranous expression of β-catenin is lost (p=0.062). Similarly metastases in negative cases developed more rapidly, than in positive ones (p=0.05). Survival rate was worse in the negative cases. The risk of metastasis in rectal cancer was significantly higher in the β-catenin membranously negative than positive groups (p=0.024) and in case of β-catenin nuclear expression the risk was also higher (p=0.047). Reduced E-cadherin expression also correlated with development of metastatic disease, but this association was statistically not significant. The immunohistochemical analysis of 79 cases shows that in Dukes B2 stage colorectal tumours clarification of β-catenin and E-cadherin expression patterns is reliable for predicting the metastatic potential of early stage rectal cancer and hence the method may have relevant implications in the therapeutic management of these cancers. C1 [Toth, Laszlo] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98., 4032 Debrecen, Hungary. [Andras, Csilla] University of Debrecen, Department of Oncology, Nagyerdei krt. 98., 4032 Debrecen, Hungary. [Molnar, Csaba] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98., 4032 Debrecen, Hungary. [Tanyi, Miklos] University of Debrecen, Department of Surgery and Operative Techniques, Nagyerdei krt. 98., 4032 Debrecen, Hungary. [Csiki, Zoltan] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Nagyerdei krt. 98., 4032 Debrecen, Hungary. [Molnar, Peter] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98., 4032 Debrecen, Hungary. [Szanto, Janos] University of Debrecen, Department of Oncology, Nagyerdei krt. 98., 4032 Debrecen, Hungary. RP Toth, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, 4032 Debrecen, Hungary. EM tothlasz@dote.hu CR Jemal A, Siegel R, Ward E et al, 2008, Cancer statistics, 2008. CA Cancer J Clin 58:71–96 Graziano F, Cascinu S, 2003, Prognostic molecular markers for planning adjuvant chemotherapy trials in Dukes B colorectal cancer patients: how much evidence is enough? Ann Oncol 14:1026–1038 Iqbal S, Stoehlmacher J, Lenz HJ, 2004, Tailored chemotherapy for colorectal cancer: a new approach to therapy. Cancer Invest 22:762–773 Stoehlmacher J, Park DJ, Zhang W et al, 2004, A multivariate analysis of genomic polymorphisms: prediction of clinical outcome to 5-FU/oxaliplatin combination chemotherapy in refractory colorectal cancer. Br J Cancer 91:344–354 Tsanou E, Peschos D, Batistatou A et al, 2008, The E-cadherin adhaesion molecule and colorectal cancer. A global literature approach. Anticancer Res 28:3815–3826 Kikuchi A, 2000, Regulation of β-catenin signalling in the Wnt pathway. Biochem Bioph Res Co 268:243–248 Lugli A, Zlobec I, Minoo P et al, 2007, Prognotic significance of the wnt signalling patway molecules APC, β-catenin and Ecadherin in colorectal cancer—a tissue microarray based analysis. Histopathology 50:453–464 Tetsu O, McCormick F, 1999, β-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 398:422–426 Ikeda S, Kishida S, Yamamoto H et al, 1998, Axin, a negative regulator of the Wnt signalling pathway, forms a complex with GSK-3 beta and β-catenin and promotes GSK3 beta dependent phosphorylation of β-catenin. EMBO J 17:1371–1384 Chen S, Liu J, Li G et al, 2008, Altered distribution of β-catenin and prognostic roles in colorectal carcinogenesis. Scand J Gastroenterol 43:456–464 Han SA, Chun H, Park CM et al, 2006, Prognostic significance of β-catenin in colorectal cancer with liver metastasis. Clin Oncol 18:761–767 Horst D, Reu S, Kriegl L et al, 2009, The intratumoral distribution of nuclear β-catenin is a prognostic marker in colon cancer. Cancer 115:2063–2070 Battifora H, 1986, The multitumor, sausage, tissue block: Novel method for immunhistochemical antibody testing. Lab Invest 55:244–248 Fernebro E, Dictor M, Bendhal P et al, 2002, Evaluation of the tissue microarray technique for immunohistochemical analysis in rectal cancer. Arch Pathol Lab Med 126:702–705 Markowitz SD, Bertagnolli MM, 2009, Molecular basis of colorectal cancer. N Engl J Med 361:2449–60 Buhmeida A, Elzagheid A, Algars A et al, 2008, Expression of the cell-cell adhaesion molecule β-catenin in colorectal carcinomas and their metastasis. APMIS 116:1–9 Hugh TJ, Dillon SA, Taylor BA et al, 1999, Cadherin-catenin expression in primary colorectal cancer: a survival analysis. Br J Cancer 80:1046–1051 Pancione M, Forte N, Sabatino L et al, 2009, Reduced β-catenin and peroxisome proliferator-activated receptor gamma expression levels are associated with colorectal cancer metastatic progression: correlation with tumor associated macrophages, cyclooxigenase 2 and patient outcome. Hum Pathol 40:714–725 Wong SCC, Lo ESF, Chan AKC et al, 2003, Nuclear β-catenin as a potential prognostic and diagnostic marker in patients with colorectal cancer from Hong Kong. J Clin Pathol Mol Pathol 56:347–352 Wong SCC, Lo ESF, Chan AKC et al, 2004, Prognostic and diagnostic significance of β-catenin nuclear immunostaining in colorectal cancer. Clin Cancer Res 10:1401–1408 Tatsuguchi A, Kishida T, Fujimori S et al, 2006, Differential expression of cyclo-oxigenase-2 and nuclear β-catenin in colorectal cancer tissue. Aliment Pharm Therap 24:153–159 Horst D, Kriegl L, Engel J et al, 2009, CD133 and nuclear β- catenin: the marker combination to detect high risk cases of low stage colorectal cancer. Eur J Cancer 45:2034–2040 Nosho K, Yamamoto H, Takahashi T et al, 2007, Genetic and epigenetic profiling in early colorectal tumors and prediction of invasive potential in pT1, early invasive, colorectal cancers. Carcinogenesis 28:1364–1370 Resnick MB, Routhier J, Konkin T et al, 2004, Epidermal growth factor receptor, c-met, β-catenin, and p53 expression as prognostic indicators in stage II colon cancer: a tissue microarray study. Clin Cancer Res 10:3069–3075 Hoos A, Nissan A, Stojadinovic A et al, 2002, Tissue microarray molecular profiling of early, node-negative adenocarcinoma of the rectum: a comprehensive analysis. Clin Cancer Res 8:3841–3849 Kallioniemi OP, Wagner U, Kononen J et al, 2001, Tissue microarray technology for high-throughput molecular profiling of cancer. Hum Mol Genet 10:657–662 Kononen J, Bubendorf L, Kallioniemi A et al, 1998, Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4:844–847 Egervari K, Szollosi Z, Nemes Z, 2007, Tissue microarray technology in breast cancer HER2 diagnostics. Pathol Res Pract 203:169–177 Sapino A, Marchio C, Senetta R et al, 2006, Routine assesment of prognostic factors in breast cancer using a multicore tissue microarray procedure. Virchows Arch 449:288–96 Fernebro E, Bendhal P, Dictor M et al, 2004, Immunohistochemical patterns in rectal cancer: applications of tissue microarray with prognostic correlations. Int J Cancer 111:921–928 Xie D, Sham JST, Zeng WF et al, 2003, Heterogenous expression and assiciation of β-catenin, p16, and c-myc in multistage colorectal tumorigenesis and progression detected by tissue microarray. Int J Cancer 107:896–902 Su Y, Shrubsole MJ, Ness RM et al, 2006, Immunohistochemical expression of Ki-67, Cyclin D1, β-catenin, cyclooxygenase-2 and epidermal growth factor receptor in human colorectal adenoma: a validation study of tissue microarray. Cancer Epidem Biomar 15:1719–1726 Bandapalli OR, Dihlmann S, Helwa R et al, 2009, Transcriptional activation of the β-catenin gene at the invasion front of colorectal liver metastasis. J Pathol 218:370–379 Da Silva L, Parry S, Reid L et al, 2008, Aberrant expression of Ecadherin in lobular carcinomas of the breast. Am J Surg Pathol 32:773–783 Muta H, Noguchi M, Kanai Y et al, 1996, E-cadherin gene mutations in signet ring cell carcinoma of stomach. Jpn J Cancer Res 87:843–848 Kapiteijn E, Liefers GJ, Los LC et al, 2001, Mechanisms of oncogenesis in colon versus rectal cancer. J Pathol 195:171–178 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 429 EP 437 DI 10.1007/s12253-011-9463-y PG 9 ER PT J AU Kaira, K Okumura, T Nakagawa, K Ohde, Y Takahashi, T Murakami, H Naito, T Endo, M Kondo, H Nakajima, T Yamamoto, N AF Kaira, Kyoichi Okumura, Takehiro Nakagawa, Kazuo Ohde, Yasuhisa Takahashi, Toshiaki Murakami, Haruyasu Naito, Tateaki Endo, Masahiro Kondo, Haruhiko Nakajima, Takashi Yamamoto, Nobuyuki TI MUC1 Expression in Pulmonary Metastatic Tumors: A Comparison of Primary Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MUC1; Pulmonary metastatic tumor; NSCLC; 18F-FDG PET; Glut1; Hypoxia ID MUC1; Pulmonary metastatic tumor; NSCLC; 18F-FDG PET; Glut1; Hypoxia AB MUC1 expression has been described as a predictor for tumor progression and worsening of prognosis in various human neoplasms. However, little is known about the role of MUC1 expression in pulmonary metastatic tumors. The aim of this study is to examine the clinicopathological significance of MUC1 expression in pulmonary metastatic tumors (PMT). One hundred forty-seven patients with PMT who underwent 18F-FDG PET before metastasectomy were included in this study. Tumor sections were stained by immunohistochemistry for MUC1, glucose transporter 1 (Glut1), hypoxia-inducible-1α (HIF-1α) and vascular endothelial growth factor (VEGF). 18F-FDG uptake and the expression of these biomarkers were correlated in primary lung cancer. MUC1 expression pattern was classified into high-grade polarized expression (HP), low-grade polarized expression (LP), or depolarized expression (DP) group. Of 147 patients, HP, LP and DP group were 9 (6%), 114 (78%) and 24 (16%), respectively. The expression of Glut1, HIF-1αand VEGF, and 18F-FDG uptake were significantly higher in DP group than HP or LP groups. MUC1 expression with HP and DP pattern was significantly higher in primary lung cancer than in PMT, whereas, MUC1 expression with LP pattern yielded a significantly high positive rate in PMT. LP group was recognized in the majority of patients with pulmonary metastatic adenocarcinoma, especially colon cancer, whereas, HP group was significantly low in pulmonary metastatic adenocarcinoma as compared with primary adenocarcinoma. Polarized MUC1 has a different expression pattern between primary and metastatic tumors with adenocarcinoma, and depolarized MUC1 is closely associated with glucose metabolism and hypoxia. C1 [Kaira, Kyoichi] Shizuoka Cancer Center, Division of Thoracic Oncology, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. [Okumura, Takehiro] Shizuoka Cancer Center, Division of Thoracic Surgery, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. [Nakagawa, Kazuo] Shizuoka Cancer Center, Division of Thoracic Surgery, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. [Ohde, Yasuhisa] Shizuoka Cancer Center, Division of Thoracic Surgery, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. [Takahashi, Toshiaki] Shizuoka Cancer Center, Division of Thoracic Oncology, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. [Murakami, Haruyasu] Shizuoka Cancer Center, Division of Thoracic Oncology, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. [Naito, Tateaki] Shizuoka Cancer Center, Division of Thoracic Oncology, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. [Endo, Masahiro] Shizuoka Cancer Center, Division of Diagnostic Radiology, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. [Kondo, Haruhiko] Shizuoka Cancer Center, Division of Thoracic Surgery, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. [Nakajima, Takashi] Shizuoka Cancer Center, Division of Pathology, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. [Yamamoto, Nobuyuki] Shizuoka Cancer Center, Division of Thoracic Oncology, 1007 Shimonagakubo Nagaizumi-cho Sunto-gun, 411-8777 Shizuoka, Japan. RP Kaira, K (reprint author), Shizuoka Cancer Center, Division of Thoracic Oncology, 411-8777 Shizuoka, Japan. EM kkaira1970@yahoo.co.jp CR Gendler SJ, Lancaster CA, Taylor-Papadimitriou J et al, 1990, Molecular cloning and expression of human tumor-associated polymorphic epithelial mucin. J Biol Chem 265:15286–15293 Schroeder JA, Masril AA, Adriance MC et al, 2004, MUC1 overexpression results in mammary gland tumorigenesis and prolonged alveolar differentiation. Oncogene 23:5739–5747 Nagai S, Takenaka K, Sonobe M et al, 2006, A novel classification of MUC1 expression is correlated with tumor differentiation and postoperative prognosis in non-small cell lung cancer. J Thorac Oncol 1:46–51 Woenckhaus M, Merk J, Stoehr R et al, 2008, Prognostic value of FHIT, CTNNB1, and MUC1 expression in non-small cell lung cancer. Hum Pathol 39:126–136 Utsunomiya T, Yonezawa S, Sakamoto H et al, 1998, Expression of MUC1 andMUC2mucins in gastric carcinomas: its relationship with the prognosis of the patients. Clin Cancer Res 4:2605–2614 Hinoda Y, Ikematsu Y, Horinouchi M et al, 2003, Increased expression of MUC1 in advanced pancreatic cancer. J Gastroenterol 38:1162–1166 Tamada S, GotoM, NomotoMet al, 2002, Expression ofMUC1 and MUC2 mucins in extrahepatic bile duct carcinomas: its relationship with tumor progression and prognosis. Pathol Int 52:713–723 Kawamoto T, Shoda J, Miyahara N et al, 2004, Expression of MUC1 recognized by a monoclonal antibody MY. 1E12 is a useful biomarker for tumor aggressiveness of carcinoma of the gallbladder. Clin Exp Metastasis 21:353–362 Feng H, Ghazizadeh M, Konishi H et al, 2002, Expression of MUC1 and MUC2 mucin gene products in human ovarian carcinomas. Jpn J Clin Oncol 32:525–529 Sivridis E, Giatromanolaki A, Koukourakis MI et al, 2002, Patterns of episialin/MUC1 expression in endometrial carcinomas and prognostic relevance. Histopathology 40:92–100 Baldus SE, Monig SP, Huxel S et al, 2004, MUC1 and nuclear beta-catenin are coexpressed at the invasion front of colorectal carcinomas and are both correlated with tumor prognosis. Clin Cancer Res 10:2790–2796 Kirschenbaum A, Itzkowitz SH, Wang JP et al, 1999, MUC1 expression in prostate carcinoma: correlation with grade and stage. Mol Urol 3:163–168 Broadbent R, Thynne G, McKenzie IFC, 1997, Antibody and T cell responses of patients with adenocarcinoma immunized with mannan-MUC1 fusion protein. J Clin Invest 100:2783–2792 Hirasawa Y, Kohno N, Yokoyama A et al, 2000, Natural antoantobody to MUC1 is a prognostic indicator for non-small cell lung cancer. Am J Respir Crit Care Med 16:589–594 Giatromanolaki A, Koukourakis MI, Sivridis E et al, 2000, Coexpression of MUC1 glycoprotein with multiple angiogenic factors in non-small cell lung cancer suggests coactivation of angiogenic and migration pathways. Clin Cancer Res 6:1917–1921 Situ D, Wang J, Ma Y, et al, 2010, Expression and prognostic relevance of MUC1 in stage IB non-small cell lung cancer. Med Oncol Nov 30 [Epub ahead of print] Kaira K, Okumura T, Ohde Y et al, 2011, Correlation between 18FFDG uptake on PET and molecular biology in metastatic pulmonary tumors. J Nucl Med 52:705–711 Kaira K, Endo M, AbeMet al, 2010, Biologic correlation of 2-[18F]- fluoro-2-deoxy-D-glucose uptake on positron emission tomography in thymic epithelial tumors. J Clin Oncol 28:3746–3753 Mikami Y, Hisatsune A, Tashiro T et al, 2009, Hypoxia enhances MUC1 expression in a lung adenocarcinoma cell line. Biochem Biophys Res Commun 379:1060–1065 Aubert S, Fauquette V, Hemon B et al, 2009, MUC1, a new hypoxia inducible factor target gene, is an actor in clear renal cell carcinoma tumor progression. Cancer Res 69:5707–5715 Kontani K, Taguchi O, Ozaki Yet al, 2003, Dendritic cell vaccine immunotherapy of cancer targeting MUC1 mucins. Int J Mol Med 12:493–502 Kufe DW, 2009, Functional targeting of MUC1 oncogene in human cancers. Cancer Biol Ther 8:1197–1203 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 439 EP 447 DI 10.1007/s12253-011-9465-9 PG 9 ER PT J AU Feher, ZL Pocsay, G Krenacs, L Zvara, Bagdi, E Pocsay, R Lukacs, G Gyory, F Gazdag, A Tarko, E Puskas, GL AF Feher, Z Liliana Pocsay, Gabor Krenacs, Laszlo Zvara, Agnes Bagdi, Eniko Pocsay, Reka Lukacs, Geza Gyory, Ferenc Gazdag, Andrea Tarko, Erzsebet Puskas, G Laszlo TI Amplification of Thymosin Beta 10 and AKAP13 Genes in Metastatic and Aggressive Papillary Thyroid Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thymosin beta 10 (TB10); Tre-2 oncogene; AKAP13; Genomic amplification; Metastasis; Papillary thyroid carcinoma (PTC) ID Thymosin beta 10 (TB10); Tre-2 oncogene; AKAP13; Genomic amplification; Metastasis; Papillary thyroid carcinoma (PTC) AB Papillary thyroid carcinoma (PTC) is the most common well-differentiated thyroid cancer. Although the great majority of the cases exhibit an indolent clinical course, some of them develop local invasion with distant metastasis, and a few cases transform into undifferentiated/anaplastic thyroid carcinoma with a rapidly lethal course. To identify gene copy number alterations predictive of metastatic potential or aggressive transformation, arraybased comparative genomic hybridization (CGH-array) was performed in 43 PTC cases. Formalin-fixed and paraffinembedded samples from primary tumours of 16 cases without metastasis, 14 cases with only regional lymph node metastasis, and 13 cases with distant metastasis, recurrence or extrathyroid extension were analysed. The CGH-array and confirmatory quantitative real-time PCR results identified the deletion of the EIF4EBP3 and TRAK2 gene loci, while amplification of thymosin beta 10 (TB10) and Tre-2 oncogene regions were observed as general markers for PTC. Although there have been several studies implicating TB10 as a specific marker based on gene expression data, our study is the first to report on genomic amplification. Although no significant difference could be detected between the good and bad prognosis cases in the A-kinase anchor protein 13 (AKAP13) gene region, it was discriminative markers for metastasis. Amplification in the AKAP13 region was demonstrated in 42.9% and 15.4% of the cases with local or with distant metastasis, respectively, while no amplification was detected in nonmetastatic cases. AKAP13 and TB10 regions may represent potential new genomic markers for PTC and cancer progression. C1 [Feher, Z Liliana] University of Szeged, Department of Medical Genetics, 6701 Szeged, Hungary. [Pocsay, Gabor] Bekes County Pandy Kalman Hospital, Department of Internal MedicineGyula, Hungary. [Krenacs, Laszlo] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Zvara, Agnes] University of Szeged, Department of Medical Genetics, 6701 Szeged, Hungary. [Bagdi, Eniko] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Pocsay, Reka] Bekes County Pandy Kalman Hospital, Department of Internal MedicineGyula, Hungary. [Lukacs, Geza] University of Debrecen, Medical and Health Science Centre, 1st Department of SurgeryDebrecen, Hungary. [Gyory, Ferenc] University of Debrecen, Medical and Health Science Centre, 1st Department of SurgeryDebrecen, Hungary. [Gazdag, Andrea] Bekes County Pandy Kalman Hospital, Department of Internal MedicineGyula, Hungary. [Tarko, Erzsebet] Borsod-Abauj-Zemplen County Hospital, 2nd Department of Internal MedicineMiskolc, Hungary. [Puskas, G Laszlo] University of Szeged, Department of Medical Genetics, 6701 Szeged, Hungary. RP Puskas, GL (reprint author), University of Szeged, Department of Medical Genetics, 6701 Szeged, Hungary. EM pusi@brc.hu CR Hundahl SA, Fleming ID, Fremgen AM et al, 1998, A National Cancer Data Base report on 53,856 cases of thyroid carcinoma treated in the U.S., 1985–1995. Cancer 83:2638–2646 Samaan NA, Schultz PN, Hickey RC et al, 1992, The results of various modalities of treatment of well differentiated thyroid carcinomas: a retrospective review of 1599 patients. J Clin Endocrinol Metab 75:714–720 Mazzaferri EL, Jhiang SM, 1994, Long-term impact of initial surgical and medical therapy on papillary and follicular thyroid cancer. Am J Med 97:418–428 Gilliland FD, Hunt WC, Morris DM et al, 1997, Prognostic factors for thyroid carcinoma. A population-based study of 15,698 cases from the Surveillance, Epidemiology and End Results, SEER, program 1973–1991. Cancer 79:564–573 Sherman SI, Brierley JD, Sperling M et al, 1998, Prospective multicenter study of thyroid carcinoma treatment: initial analysis of staging and outcome. National Thyroid Cancer Treatment Cooperative Study Registry Group. Cancer 83:1012–1021 Tsang RW, Brierley JD, Simpson WJ et al, 1998, The effects of surgery, radioiodine, and external radiation therapy on the clinical outcome of patients with differentiated thyroid carcinoma. Cancer 82:375–388 Nikiforova MN, Kimura ET, Gandhi M et al, 2003, BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas. J Clin Endocrinol Metab 88:5399–5404 Fagin JA, 2004, Challenging dogma in thyroid cancer molecular genetics-role of RET/PTC and BRAF in tumour initiation. J Clin Endocrinol Metab 89:4264–4266 Xing M, 2005, BRAF mutation in thyroid cancer. Endocr Relat Cancer 12:245–262 Xing M, Westra WH, Tufano RP et al, 2005, BRAF mutation predicts a poorer clinical prognosis for papillary thyroid cancer. J Clin Endocrinol Metab 90:6373–6379 Kondo T, Ezzat S, Asa SL, 2006, Pathogenetic mechanisms in thyroid follicular-cell neoplasia. Nat Rev Cancer 6:292–306 Adeniran AJ, Zhu Z, Gandhi M et al, 2006, Correlation between genetic alterations and microscopic features, clinical manifestations, and prognostic characteristics of thyroid papillary carcinomas. Am J Surg Pathol 30:216–222 Giannini R, Ugolini C, Lupi C et al, 2007, The heterogeneous distribution of BRAF mutation supports the independent clonal origin of distinct tumour foci in multifocal papillary thyroid carcinoma. J Clin Endocrinol Metab 92:3511–3516 Ciampi R, Nikiforov YE, 2007, RET/PTC rearrangements and BRAF mutations in thyroid tumorigenesis. Endocrinology 148:936–941 Suarez HG, 1998, Genetic alterations in human epithelial thyroid tumours. Clin Endocrinol, Oxf, 8:531–546 Garcia-Rostan G, Zhao H, Camp RL et al, 2003, Ras mutations are associated with aggressive tumour phenotypes and poor prognosis in thyroid cancer. J Clin Oncol 1:3226–3235 Ciampi R, Knauf JA, Kerler R et al, 2005, Oncogenic AKAP9- BRAF fusion is a novel mechanism of MAPK pathway activation in thyroid cancer. J Clin Invest 115:94–101 Santoro M, Papotti M, Chiappetta G et al, 2002, RET activation and clinicopathologic features in poorly differentiated thyroid tumours. J Clin Endocrinol Metab 87:370–379 Tallini G, Santoro M, Helie M et al, 1998, RET/PTC oncogene activation defines a subset of papillary thyroid carcinomas lacking evidence of progression to poorly differentiated or undifferentiated tumour phenotypes. Clin Cancer Res 4:287–294 Nikiforov YE, 2002, RET/PTC rearrangement in thyroid tumors. Endocr Pathol 13:3–16 Greco A, Miranda C, Pagliardini S et al, 1997, Chromosome 1 rearrangements involving the genes TPR and NTRK1 produce structurally different thyroid-specific TRK oncogenes. Genes Chromosomes Cancer 19:112–123 Beimfohr C, Klugbauer S, Demidchik EP et al, 1999, NTRK1 rearrangement in papillary thyroid carcinomas of children after the Chernobyl reactor accident. Int J Cancer 80:842–847 Ito T, Seyama T, Mizuno T et al, 1992, Unique association of p53 mutations with undifferentiated but not with differentiated carcinomas of the thyroid gland. Cancer Res 52:1369–1371 Fagin JA, Matsuo K, Karmakar A et al, 1993, High prevalence of mutations of the p53 gene in poorly differentiated human thyroid carcinomas. J Clin Invest 91:179–184 Asakawa H, Kobayashi T, 2002, Multistep carcinogenesis in anaplastic thyroid carcinoma: a case report. Pathology 34:94–97 Garcia-Rostan G, Camp RL, Herrero A et al, 2001, Beta-catenin dysregulation in thyroid neoplasms: down-regulation, aberrant nuclear expression, and CTNNB1 exon 3 mutations are markers for aggressive tumor phenotypes and poor prognosis. Am J Pathol 158:987–996 Huang Y, Prasad M, Lemon WJ et al, 2001, Gene expression in papillary thyroid carcinoma reveals highly consistent profiles. Proc Natl Acad Sci USA 98:15044–15049 Puskas LG, Juhasz F, Zarva A et al, 2005, Gene profiling identifies genes specific for well-differentiated epithelial thyroid tumors. Cell Mol Biol, Noisy-le-grand, 51:177–186 Puskas LG, Farid NR, 2004, Gene expression in thyroid tumors. Cancer Treat Res 122:265–271 Gunn SR, Robetorye RS, Mohammed MS et al, 2007, Comparative genomic hybridization arrays in clinical pathology: progress and challenges. Mol Diagn Ther 11:73–77 Hemmer S, Wasenius VM, Knuutila S et al, 1999, DNA copy number changes in thyroid carcinoma. Am J Pathol 154:1539–1547 Kjellman P, Lagercrantz S, HoogAet al, 2001)Gain of 1q and loss of 9q21.3-q32 are associated with a less favorable prognosis in papillary thyroid carcinoma. Genes Chromosomes Cancer 32:43–49 Rodrigues R, Roque L, Espadinha C et al, 2007, Comparative genomic hybridization, BRAF, RAS, RET, and oligo-array analysis in aneuploid papillary thyroid carcinomas. Oncol Rep 18:917–926 Lee JJ, Au AY, Foukakis T et al, 2008, Array-CGH identifies cyclin D1 and UBCH10 amplicons in anaplastic thyroid carcinoma. Endocr Relat Cancer 15:801–815 Finn S, Smyth P, O’Regan E et al, 2007, Low-level genomic instability is a feature of papillary thyroid carcinoma: an array comparative genomic hybridization study of laser capture microdissected papillary thyroid carcinoma tumors and clonal cell lines. Arch Pathol Lab Med 131:65–73 Wreesmann VB, Sieczka EM, Socci ND et al, 2004, Genomewide profiling of papillary thyroid cancer identifies MUC1 as an independent prognostic marker. Cancer Res 64:3780–3789 Unger K, Malisch E, Thomas G et al, 2008, Array CGH demonstrates characteristic aberration signatures in human papillary thyroid carcinomas governed by RET/PTC. Oncogene 27:4592–4602 Kitamura Y, Shimizu K, Tanaka S et al, 2000, Association of allelic loss on 1q, 4p, 7q, 9p, 9q, and 16q with postoperative death in papillary thyroid carcinoma. Clin Cancer Res 6:1819–1825 Pacini F, Schlumberger M, Harmer C et al, 2005, Post-surgical use of radioiodine 131I in patients with papillary and follicular thyroid cancer and the issue of remnant ablation: a consensus report. Eur J Endocrinol 153:651–659 Pacini F, Schlumberger M, Dralle G et al, 2006, European consensus for the management of patients with differentiated thyroid carcinoma of the follicular epithelium. Eur J Endocrinol 154:767–803 Feher LZ, Balazs M, Kelemen JZ et al, 2006, Improved DOPPCR- based representational whole-genome amplification using quantitative real-time PCR. Diagn Mol Pathol 15:43–48 Telenius H, Carter NP, Bebb CE et al, 1992, Degenerate oligonucleotide-primed PCR: general amplification of target DNA by a single degenerate primer. Genomics 13:718–725 Nagy J, Feher LZ, Sonkodi I et al, 2005, A second field metachronous Merkel cell carcinoma of the lip and the palatine tonsil confirmed by microarray-based comparative genomic hybridisation. Virchows Arch 446:278–286 Yang YH, Dudoit S, Luu P et al, 2002, Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res 30:e15 Pfaffl MW, 2001, A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 29:e45 Onno M, Nakamura T, Mariage-Samson R et al, 1993, Human TRE17 oncogene is generated from a family of homologous polymorphic sequences by single-base changes. DNA Cell Biol 12:107–118 Martinu L, Masuda-Robens JM, Robertson SE et al, 2004, The TBC, Tre-2/Bub2/Cdc16, domain protein TRE17 regulates plasma membrane-endosomal trafficking through activation of Arf6. Mol Cell Biol 24:9752–9762 Masuda-Robens JM, Kutney SN, Qi H et al, 2003, The TRE17 oncogene encodes a component of a novel effector pathway for Rho GTPases Cdc42 and Rac1 and stimulates actin remodeling. Mol Cell Biol 23:2151–2161 Smith MJ, Pozo K, Brickley K et al, 2006, Mapping the GRIF-1 binding domain of the kinesin, KIF5C, substantiates a role for GRIF-1 as an adaptor protein in the anterograde trafficking of cargoes. J Biol Chem 281:27216–27228 Brickley K, Smith MJ, Beck M et al, 2005, GRIF-1 and OIP106, members of a novel gene family of coiled-coil domain proteins: association in vivo and in vitro with kinesin. J Biol Chem 280:14723–14732 Yasuhara T, Okamoto A, Kitagawa T et al, 2005, FGF7-like gene is associated with pericentric inversion of chromosome 9, and FGF7 is involved in the development of ovarian cancer. Int J Oncol 26:1209–1216 Hishikawa Y, Tamaru N, Ejima K et al, 2004, Expression of keratinocyte growth factor and its receptor in human breast cancer: its inhibitory role in the induction of apoptosis possibly through the overexpression of Bcl-2. Arch Histol Cytol 67:455–464 Firth JD, Putnins EE, 2004, Keratinocyte growth factor 1 inhibits wound edge epithelial cell apoptosis in vitro. J Invest Dermatol 122:222–231 Diviani D, Abuin L, Cotecchia S et al, 2004, Anchoring of both PKA and 14-3-3 inhibits the Rho-GEF activity of the AKAP-Lbc signaling complex. EMBO J 23:2811–2820 Shibolet O, Giallourakis C, Rosenberg I et al, 2007, AKAP13, a RhoA GTPase-specific guanine exchange factor, is a novel regulator of TLR2 signaling. J Biol Chem 282:35308–35317 Wirtenberger M, Tchatchou S, Hemminki K et al, 2006, Association of genetic variants in the Rho guanine nucleotide exchange factor AKAP13 with familial breast cancer. Carcinogenesis 27:593–598 Chiappetta G, Pentimalli F, Monaco M et al, 2004, Thymosin beta-10 gene expression as a possible tool in diagnosis of thyroid neoplasias. Oncol Rep 12:239–243 Takano T, Hasegawa Y, Miyauchi A et al, 2002, Quantitative analysis of thymosin beta-10 messenger RNA in thyroid carcinomas. Jpn J Clin Oncol 32:229–232 Hall AK, 1995, Thymosin beta-10 accelerates apoptosis. Cell Mol Biol Res 41:167–180 Maelan AE, Rasmussen TK, Larsson LI, 2007, Localization of thymosin beta10 in breast cancer cells: relationship to actin cytoskeletal remodeling and cell motility. Histochem Cell Biol 127:109–113 Califano D, Monaco C, Santelli G et al, 1998, Thymosin beta-10 gene overexpression correlated with the highly malignant neoplastic phenotype of transformed thyroid cells in vivo and in vitro. Cancer Res 58:823–828 Santelli G, Califano D, Chiappetta G et al, 1999, Thymosin beta-10 gene overexpression is a general event in human carcinogenesis. Am J Pathol 155:799–804 Lee SH, Son MJ, Oh SH et al, 2005, Thymosin {beta}(10, inhibits angiogenesis and tumour growth by interfering with Ras function. Cancer Res 65:137–148 Santelli G, Bartoli PC, Giuliano A et al, 2002, Thymosin beta-10 protein synthesis suppression reduces the growth of human thyroid carcinoma cells in semisolid medium. Thyroid 12:765–772 Huff T, Muller CS, Otto AM et al, 2001, beta-Thymosins, small acidic peptides with multiple functions. Int J Biochem Cell Biol 33:205–220 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 449 EP 458 DI 10.1007/s12253-011-9467-7 PG 10 ER PT J AU El-Gendi, MS Mostafa, FM El-Gendi, MA AF El-Gendi, Mohamed Saba Mostafa, Farouk Mohamed El-Gendi, Mohamed Ahmed TI Stromal Caveolin-1 Expression in Breast Carcinoma. Correlation with Early Tumor Recurrence and Clinical Outcome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast carcinoma; Clinical outcome; Predictive value; Stromal caveolin-1; Tumor recurrence ID Breast carcinoma; Clinical outcome; Predictive value; Stromal caveolin-1; Tumor recurrence AB Caveolin- (cav-1) has been linked to tumor progression and clinical outcome in breast cancer, but its role as a prognostic marker is still unclear. We evaluated stromal and tumor caveolin-1 expression in 91 breast carcinomas, and assessed the association between their expression and clinicopathologic variables as well as patient outcome and early tumor recurrence. Absence of stromal caveolin-1 expression was detected in 18.7% of cases, while 25.3% of cases revealed tumor epithelial caveolin-1 expression. Combined stromal and tumor caveolin-1 immunopositivity was seen in 24.2% of cases. Absence of stromal cav-1 associated with larger tumor size, higher grade, higher nodal stage, higher number of positive nodes, higher TNM stage, positive HER2 status, higher recurrence rate, and shorter mean progression free survival (PFS). Stromal cav-1 status was a significant predictor of PFS in ER+, PR +, and HER2 + tumors. In tamoxifentreated patients, absence of stromal Cav-1 was a significant predictor of poor clinical outcome, suggestive of tamoxifen resistance. Conversely, tumor epithelial and combined caveolin-1 expression, didnot associate with patient outcome. In multivariate analysis, only TNM stage independently associated with survival. Loss of stromal caveolin-1 is a novel breast cancer biomarker that can predict early tumor recurrence, short PFS, and tamoxifen- resistance. Thus, its use as a predictive biomarker, especially in lower grade, lower stage, ER+, PR+, HER2+, and tamoxifen treated patients may allow for early interventions with more aggressive therapies. Thus, stromal marker expression and epithelial-stromal cross talk may be critical for tumor progression and metastasis. C1 [El-Gendi, Mohamed Saba] University of Alexandria, Faculty of Medicine, Department of Pathology, 29 Fawzy Moaaz Street, SmouhaAlexandria, Egypt. [Mostafa, Farouk Mohamed] Alexandria Faculty of Medicine, Departement of Clinical OncologyAlexandria, Egypt. [El-Gendi, Mohamed Ahmed] Alexandria University, Medical Research Institute, Department of Experimental and Clinical SurgeryAlexandria, Egypt. RP El-Gendi, MS (reprint author), University of Alexandria, Faculty of Medicine, Department of Pathology, Alexandria, Egypt. EM sabagendi@yahoo.com CR Liedtke C, Kersting C, Burger H et al, 2007, Caveolin-1 expression in benign and malignant lesions of the breast. World J Surg Oncol 5:110., DOI 10.1186/1477-7819-5-110 Parkin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Witkiewicz AK, Dasgupta A, Sotgia F et al, 2009, An Absence of Stromal Caveolin-1 Expression Predicts Early Tumor Recurrence and Poor Clinical Outcome in Human Breast Cancers. Am J Pathol 174:2023–34 Bhowmick NA, Neilson EG, Moses HL, 2004, Stromal fibroblasts in cancer initiation and progression. Nature 432:332–7 Bissell MJ, Radisky D, 2001, Putting tumors in context. Nat Rev Cancer 1:1–19 Mueller MM, Fusenig NE, 2004, Friends or foes—bipolar effects of the tumor stroma in cancer. Nat Rev Cancer 4:839–49 Kim JB, Stein R, O’Hare MJ, 2005, Tumor-stromal interactions in breast cancer: the role of stroma in tumorigenesis. Tumour Biol 26:173–85 Sloan EK, Ciocca DR, Pouliot N et al, 2009, Stromal cell expression of Caveolin-1 predicts outcome in breast cancer. Am J Pathol 174:2035–43 Olumi AF, Grossfeld GD, Hayward SW, 1999, Carcinomaassociated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res 59:5002–11 Serini G, Gabbiani G, 1999, Mechanisms of myofibroblast activity and phenotypic modulation. Exp Cell Res 250:273–83 Hnasko R, Lisanti MP, 2003, The biology of caveolae: lessons from caveolin knockout mice and implications for human disease. Mol Interv 3:445–64 Koleske AJ, Baltimore D, Lisanti MP, 1995, Reduction of caveolin and caveolae in oncogenically transformed cells. Proc Natl Acad Sci USA 92:1381–5 Fielding CJ, Fielding PE, 2000, Cholesterol and caveolae: structural and functional relationships. Biochim Biophys Acta 1529:210–22 Page DL, Jensen RA, Simpson JF, 1998, Routinely available indicators of prognosis in breast cancer. Breast Cancer Res Treat 51:195–208 Fitzgibbons PL, Page DL, Weaver D et al, 2000, Prognostic factors in breast cancer. College of American Pathologists Consensus Statement 1999. Arch Pathol Lab Med 124, 7):966–78 Ellis IO, Elston CW, 2006, Histologic grade, chapter 19). In: O’Malley FP, Pinder SE, eds, Breast Pathology. Elsevier, Philadelphia, PA, pp 225–33 Savage K, Lambros MB, Robertson D et al, 2007, Caveolin-1 is overexpressed and amplified in a subset of basal-like and metaplastic breast carcinomas: a morphologic, ultrastructural, immunohistochemical, and in situ hybridization analysis. Clin Cancer Res 13:90–101 Allred DC, Harvey JM, Berardo M et al, 1998, Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11:155–68 Ellis IO, Bartlett J, Dowsett M et al, 2004, Best Practice No. 176: Updated recommendation for HER-2 testing in the UK. J Clin Pathol 57:322–7 Williams TM, Medina F, Badano I et al, 2004, Caveolin-1 gene disruption promotes mammary tumorigenesis and dramatically enhances lung metastasis in vivo. Role of cav-1 in cell invasiveness and matrix metalloproteinase, MMP-2/9, secretion. J Biol Chem 279:51630–46 Elsheikh SE, Green AR, Rakha EA et al, 2008, Caveolin-1 and Caveolin- 2 are associated with breast cancer basal-like and triplenegative immunophenotype. Br J Cancer 99:327–34 Li T, Sotgia F, Vuolo MA et al, 2006, Caveolin-1 mutations in human breast cancer: functional association with estrogen receptor alpha-positive status. Am J Pathol 168:1998–2013 Sagara Y, Mimori K, Yoshinaga K et al, 2004, Clinical significance of Caveolin-1, Caveolin-2 and HER-2/neu mRNA expression in human breast cancer. Br J Cancer 91:959–65 Hurlstone AF, Reid G, Reeves JR et al, 1999, Analysis of the Caveolin-1 gene at human chromosome 7q31.1 in primary tumors and tumor-derived cell lines. Oncogene 18:1881–90 Engelman JA, Lee RJ, Karnezis A et al, 1998, Reciprocal regulation of neu tyrosine kinase activity and caveolin-1 protein expression in vitro and in vivo. Implications for human breast cancer. J Biol Chem 273:20448–55 Lee SW, Reimer CL, Oh P et al, 1998, Tumor cell growth inhibition by caveolin re-expression in human breast cancer cells. Oncogene 16:1391–7 Yang G, Truong LD, Timme TL et al, 1998, Elevated expression of caveolin is associated with prostate and breast cancer. Clin Cancer Res 4:1873–80 Galbiati F, Volonte D, Brown AM et al, 2000, Caveolin-1 expression inhibits Wnt/beta-catenin/Lef-1 signaling by recruiting beta-catenin to caveolae membrane domains. J Biol Chem 275:23368–77 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 459 EP 469 DI 10.1007/s12253-011-9469-5 PG 11 ER PT J AU Gao, Ch Pang, L Ren, Ch Ma, T AF Gao, Chengcheng Pang, Liqun Ren, Chengcheng Ma, Tianheng TI Prognostic Value of Raf Kinase Inhibitor Protein in Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal squamous cell carcinoma; Raf kinase inhibitor protein; Prognosis ID Esophageal squamous cell carcinoma; Raf kinase inhibitor protein; Prognosis AB Raf kinase inhibitory protein (RKIP, also PEBP1) is involved in regulation of multiple cellular signaling processes and suppressing metastasis in animal models. Downregulation of RKIP expression has been shown to promote tumor progression in a variety of human cancers. However, its role and clinical significance in resectable esophageal squamous cell carcinoma (ESCC) is still scanty. The purpose of this study was to investigate the prognostic significance of RKIP expression by immunohistochemistry in a group of patients with ESCC treated with surgical resection. RKIP expression in 233 surgically resected ESCC specimens and 49 cases of adjacent normal tissues was detected by using immunohistochemical staining. The clinical and prognostic significance of RKIP expression was statistically analyzed. Kaplan-Meier analysis was used to compare the postoperative survival between groups. Significant downregulation was noted for RKIP protein in ESCCs, compared to adjacent normal tissues (p<0.001). A lower disease-free survival and overall survival of ESCC was found in patients whose tissues had low RKIP expression (both P<0.001). In addition, RKIP expression could stratify the patient survival (disease-free survival / overall survival) in stage II (P=0.01 and 0.02, repectively). The Cox proportionate hazard regression model also established that low expression of RKIP was significantly correlated with increased risk (RR=3.572) of recurrence compared with high RKIP expression (P<0.001). Furthermore, the results of multivariate analysis suggested that RKIP expression (P<0.001) was an independent factor that affected overall survival. These findings suggest that the low expression of RKIP be associated with poor survival in resectable ESCC patients. C1 [Gao, Chengcheng] Nanjing Medical University, Huai’an First People’s Hospital, Department of Gastroenterology, 223300 Huainan, Jiangsu, China. [Pang, Liqun] Nanjing Medical University, Huai’an First People’s Hospital, Department of Gastroenterology, 223300 Huainan, Jiangsu, China. [Ren, Chengcheng] Nanjing Medical University, Huai’an First People’s Hospital, Department of Gastroenterology, 223300 Huainan, Jiangsu, China. [Ma, Tianheng] Nanjing Medical University, Huai’an First People’s Hospital, Department of Gastroenterology, 223300 Huainan, Jiangsu, China. RP Ren, Ch (reprint author), Nanjing Medical University, Huai’an First People’s Hospital, Department of Gastroenterology, 223300 Huainan, China. EM gao.jsha@gmail.com CR Landis SH, Murray T, Bolden S, Wingo PA, 1999, Cancer statistics. CA Cancer J Clin 49:8–31 Yu WW, Guo YM, Zhu M, Cai XW, Zhu ZF, Zhao WX, Fu XL, 2011, Clinicopathological and prognostic significance of EGFR over-expression in esophageal squamous cell carcinoma: a meta-analysis. Hepatogastroenterology 58:426–431 Liu J, Hu Y, Hu W, Xie X, Ela Bella A, Fu J, Rao D., 2011, Expression and prognostic relevance of p21(WAF1, in stage III esophageal squamous cell carcinoma. Dis Esophagus. In press. Keller ET, Fu Z, Brennan M, 2004, The role of Raf kinase inhibitor protein, RKIP, in health and disease. Biochem Pharmacol 68:1049–1053 Keller ET, 2004, Metastasis suppressor genes: a role for raf kinase inhibitor protein, RKIP). Anticancer Drugs 15:663–669 Trakul N, Rosner MR, 2005, Modulation of the MAP kinase signaling cascade by Raf kinase inhibitory protein. Cell Res 15:19–23 Bernier I, Jolles P, 1984, Purification and characterization of a basic 23 kDa cytosolic protein from bovine brain. Biochim Biophys Acta 790:174–181 Khamis ZI, Iczkowski KA, Sang QX., 2011, Metastasis suppressors in human benign prostate, intraepithelial neoplasia, and invasive cancer: their prospects as therapeutic agents. Med Res Rev. In press. Li HZ, Gao Y, Zhao XL, Liu YX, Sun BC, Yang J, Yao Z, 2009, Effects of raf kinase inhibitor protein expression on metastasis and progression of human breast cancer. Mol Cancer Res 7:832–840 Xu YF, Yi Y, Qiu SJ, Gao Q, Li YW, Dai CX, Cai MY, Ju MJ, Zhou J, Zhang BH, Fan J, 2010, PEBP1 downregulation is associated to poor prognosis in HCC related to hepatitis B infection. J Hepatol 53:872–879 Baritaki S, Katsman A, Chatterjee D, Yeung KC, Spandidos DA, Bonavida B, 2007, Regulation of tumor cell sensitivity to TRAILinduced apoptosis by the metastatic suppressor Raf kinase inhibitor protein via Yin Yang 1 inhibition and death receptor 5 up-regulation. J Immunol 179:5441–5453 Rice TW, Blackstone EH, Rusch VW, 2010, 7th edition of the AJCC Cancer Staging Manual: esophagus and esophagogastric junction. Ann Surg Oncol 17:1721–1724 Wang J, Yang YH, Wang AQ, Yao B, Xie G, Feng G, Zhang Y, Cheng ZS, Hui L, Dai TZ, Du XB, Wang D, 2010, Immunohistochemical detection of the Raf kinase inhibitor protein in nonneoplastic gastric tissue and gastric cancer tissue. Med Oncol 27:219–223 Zaravinos A, Chatziioannou M, Lambrou GI, Boulalas I, Delakas D, Spandidos DA, 2011, Implication of RAF and RKIP Genes in Urinary Bladder Cancer. Pathol Oncol Res 17:181–190 Baritaki S, Bonavida B, 2010, Viral infection and cancer: the NFkappaB/ Snail/RKIP loop regulates target cell sensitivity to apoptosis by cytotoxic lymphocytes. Crit Rev Immunol 30:31–46 Wu K, Bonavida B, 2009, The activated NF-kappaB-Snail-RKIP circuitry in cancer regulates both the metastatic cascade and resistance to apoptosis by cytotoxic drugs. Crit Rev Immunol 29:241–254 Fu Z, Smith PC, Zhang L, Rubin MA, Dunn RL, Yao Z, Keller ET, 2003, Effects of raf kinase inhibitor protein expression on suppression of prostate cancer metastasis. J Natl Cancer Inst 95:878–889 Biewenga P, Buist MR, Moerland PD, Loren V, van Themaat E, van Kampen AH, ten Kate FJ, Baas F, 2008, Gene expression in early stage cervical cancer. Gynecol Oncol 108:520–526 Ruan L, Wang GL, Yi H, Chen Y, Tang CE, Zhang PF, Li MY, Li C, Peng F, Li JL, Chen ZC, Xiao ZQ, 2010, Raf kinase inhibitor protein correlates with sensitivity of nasopharyngeal carcinoma to radiotherapy. J Cell Biochem 110:975–981 Al-Mulla F, Hagan S, Al-Ali W et al, 2008, Raf kinase inhibitor protein: mechanism of loss of expression and association with genomic instability. J Clin Pathol 61:524–529 Minoo P, Zlobec I, Baker K et al, 2007, Loss of raf-1 kinase inhibitor protein expression is associated with tumor progression and metastasis in colorectal cancer. Am J Clin Pathol 127:820– 827 Lee HC, Tian B, Sedivy JM, Wands JR, Kim M, 2006, Loss of Raf kinase inhibitor protein promotes cell proliferation and migration of human hepatoma cells. Gastroenterology 131:1208– 1217 Hagan S, Al-Mulla F, Mallon E et al, 2005, Reduction of Raf-1 kinase inhibitor protein expression correlates with breast cancer metastasis. Clin Cancer Res 11:7392–7397 Park S, Yeung ML, Beach S, Shields JM, Yeung KC, 2005, RKIP downregulates B-Raf kinase activity in melanoma cancer cells. Oncogene 24:3535–3540 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 471 EP 477 DI 10.1007/s12253-011-9470-z PG 7 ER PT J AU Kosmaczewska, A Bocko, D Ciszak, L Wlodarska-Polinska, I Kornafel, J Szteblich, A Masternak, A Frydecka, I AF Kosmaczewska, Agata Bocko, Dorota Ciszak, Lidia Wlodarska-Polinska, Iwona Kornafel, Jan Szteblich, Aleksandra Masternak, Anna Frydecka, Irena TI Dysregulated Expression of Both the Costimulatory CD28 and Inhibitory CTLA-4 Molecules in PB T Cells of Advanced Cervical Cancer Patients Suggests Systemic Immunosuppression Related to Disease Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD28; CTLA-4; T cell; Cervical cancer; Progression; Systemic immunosuppression ID CD28; CTLA-4; T cell; Cervical cancer; Progression; Systemic immunosuppression AB Cervical cancer (CC) occurs more frequently in women who are immunosuppressed, suggesting that both local and systemic immune abnormalities may be involved in the evolution of the disease. Costimulatory CD28 and inhibitory CTLA-4 molecules expressed in T cells play a key role in the balanced immune responses. There has been demonstrated a relation between CD28, CTLA-4, and IFN genes in susceptibility to CC, suggesting their importance in CC development. Therefore, we assessed the pattern of CD28 and CTLA-4 expression in T cells from PB of CC patients with advanced CC (stages III and IV according to FIGO) compared to controls. We also examined the ability of PBMCs to secrete IFN-gamma. We found lower frequencies of freshly isolated and ex vivo stimulated CD4+CD28+ and CD8+CD28+ T cells in CC patients than in controls. Loss of CD28 expression was more pronounced in the CD8+ T subset. Markedly increased proportions of CTLA-4+ T cells in CC patients before and after culture compared to controls were also observed. In addition, patients’ T cells exhibited abnormal kinetics of surface CTLA-4 expression, with the peak at 24 h of stimulation, which was in contrast to corresponding normal T cells, revealing maximum CTLA-4 expression at 72 h of stimulation. Of note, markedly higher IFN-gamma concentrations were shown in supernatants of stimulated PBMCs from CC patients. Conclusions: Our report shows the dysregulated CD28 and CTLA-4 expression in PB T cells of CC patients, which may lead to impaired function of these lymphocytes and systemic immunosuppression related to disease progression. C1 [Kosmaczewska, Agata] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, R. Weigla 12, 53-114 Wroclaw, Poland. [Bocko, Dorota] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, R. Weigla 12, 53-114 Wroclaw, Poland. [Ciszak, Lidia] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, R. Weigla 12, 53-114 Wroclaw, Poland. [Wlodarska-Polinska, Iwona] Wroclaw Medical University, Department of Gynecological OncologyWroclaw, Poland. [Kornafel, Jan] Wroclaw Medical University, Department of Gynecological OncologyWroclaw, Poland. [Szteblich, Aleksandra] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, R. Weigla 12, 53-114 Wroclaw, Poland. [Masternak, Anna] Regional Hospital in Opole, Department of HematologyOpole, Poland. [Frydecka, Irena] Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, R. Weigla 12, 53-114 Wroclaw, Poland. RP Kosmaczewska, A (reprint author), Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, 53-114 Wroclaw, Poland. EM kosmacz@iitd.pan.wroc.pl CR Cheng WF, Lee CN, Su YN, Chang MC et al, 2005, Induction of human papillomavirus type 16-specific immunologic responses in a normal and an human papillomavirus-infected populations. Immunology 115:136–149 Rellihan MA, Dooley DP, Burke TW et al, 1990, Rapidly progressing cervical cancer in a patient with human immunodeficiency virus infection. Gynecol Oncol 36:435–438 Ahmed SM, Al-Doujaily H, Reid WMN et al, 2002, The cellular response associated with cervical intraepithelial neoplasia in HIV +and HIV- subjects. Scand J Immunol 56:204–211 Santegoets LA, van Setyrs M, Heijmans-Antonissen C et al, 2008, Reduced local immunity in HIV-related VIN: expression of chemokines and involvement of immunocompetent cells. Int J Cancer 123:616–622 Nasiell K, Roger V, Nasiell M, 1986, Behaviour of mild cervical dysplasia during long term follow-up. Obstet Gynecol 67:665–669 Hilders C, Houbiers J, van Ravenswaay CH et al, 1993, Association between HLA-expression and infiltration of immune cells in cervical carcinoma. Lab Invest 69:651–659 Ivanson EL, Juko-Pecirep I, Gyllensten UB, 2010, Interaction of immunological genes on chromosome 2q33 and IFNG in susceptibility to cervical cancer. Gynec Oncol 116:544–548 Pravica V, Perrey C, Stevens A et al, 2000, A single nucleotide polymorphism in the first intron of the human IFN-gamma gene: absolute correlation with a polymorphic CA microsatellite marker of high IFN-gamma production. Hum Immunol 61:863–866 Guzman VB, Yambartsev A, Goncalves-Primo A et al, 2008, New approach reveals CD28 and IFNG gene interaction in the susceptibility to cervical cancer. Hum Mol Genet 17:1838–1844 Kim CW, Choi SH, Chung EJ, Lee MJ et al, 1999, Alteration of signal transducing molecules and phenotypical characteristics in peripheral blood lymphocytes from gastric carcinoma patients. Pathobiology 67:123–128 Melichar B, Nash MA, Lenzi R, et al. Expression of costimulatory molecules CD80 and CD86 and their receptors CD28, CTLA-4 on malignant ascites CD3+ tumor-infiltrating lymphocytes, TIL, from patients with ovarian and other types of peritoneal carcinomatosis. Clin Exp Immunol 119:19–27 Frydecka I, Kosmaczewska A, Bocko D et al, 2004, Alterations of the expression of T-cell related costimulatory CD28 and down regulatory CD152, CTLA-4, molecules n patients with B-cell chronic lymphocytic leukaemia. Br J Cancer 90:2042–2048 Motta M, Rassenti L, Shelvin BJ et al, 2005, Increased expression of CD152, CTLA-4, by normal T lymphocytes in untreated patients with B-cell chronic lymphocytic leukaemia. Leukemia 19:1788–1793 Labalette M, Leteurtre E, Thumerelle C et al, 1999, Peripheral human CD8+CD28+ T lymphocytes give rise to CD28- progeny, but IL-4 prevents loss of CD28 expression. Int Immunol 11:1327– 1335 Melichar B, Solichova D, Freedman RS, 2006, Neopterin as an indicator of immune activation and prognosis in patients with gynecological malignancies. Int J Gynecol Cancer 16:240–252 Ligers A, Teleshova N, Masterman T et al, 2001, CTLA-4 gene expression is influenced by promoter and exon 1 polymorphisms. Genes Immun 2:145–152 Pawlak E, Karabon L, Wlodarska-Polinska I et al, 2010, Influence of CTLA-4/CD28/ICOS gene polymorphisms on the susceptibility to cervical squamous cell carcinoma and stage of differentiation in the Polish population. Hum Immunol 71:195–200 Su TH, Chang TY, Lee YJ, Chen CK et al, 2007, CTLA-4 gene and susceptibility to human papillomavirus-16-associated cervical cell carcinoma in Taiwanese women. Carcinogenesis 28:1237–1240 Bortwick NJ, Lowdell M, Salmon M, Akbar AN, 2000, Loss of CD28 expression on CD8+ T cells is induced by IL-2 receptor gamma chain signaling cytokines and type I IFN, and increases susceptibility to activation-inducted apoptosis. Int Immunol 12:1005–1013 Bryl E, Vallejo AN, Weyand CM, Goronzy JJ, 2001, Downregulation of CD28 expression by TNF-alpha. J Immunol 167:3231–3238 Chopra V, Dinh TV, Hanningan EV, 1998, Circulating serum levels of cytokines and angiogenic factors in patients with cervical cancer. Cancer Invest 16:152–159 Gaiotti D, Chung J, Iglesias M et al, 2000, Tumor necrosis factoralpha promotes human papillomavirus, HPV, E6/E7 RNA expression and cyclin-dependent kinase activity in HPV-immortalized keratinocytes by a ras-dependent pathway. Mol Carcinog 27:97–109 Azar KK, Tani M, Yasuda H et al, 2004, Increased secretion patterns of interleukin-10 and tumor necrosis factor-alpha in cervical squamous intraepithelial lesions. Hum Pathol 35:1376– 1384 Zijlmans HJ, Fleuren GJ, Baelde HJ et al, 2007, Role of tumor-derived proinflammatory cytokinesGM-CSF, TNF-alpha, and IL-12 in the migration and differentiation of antigen-presenting cells in cervical carcinoma. Cancer 109:556–565 Linsley P, Bradshaw J, Urnes M et al, 1993, CD28 engagement by B7/BB1 induces transient down-regulation of CD28 synthesis and prolonged unresponsiveness to CD28 signalling. J Immunol 150:3161–3169 Higgins GD, Davy M, Roder D et al, 1991, Increased age and mortality associated with cervical carcinomas negative for human papillomavirus RNA. Lancet 338:910–913 Ha SJ, West EE, Araki K et al, 2008, Manipulating both the inhibitory and stimulatory immune system towards the success of therapeutic vaccination against chronic viral infections. Immunol Rev 223:317–333 Nakamoto N, Cho H, Shaked A et al, 2009, Synergistic reversal of intrahepatic HCV-specific CD8 T cell exhaustion by combined PD-1/CTLA-4 blockade. PLoS Pathogens 5:e1000313 Brittenden J, Heys SD, Ross J, Eremin O, 1996, Natural killer cells and cancer. Cancer 77:1226–1243 Cosiski-Marana HR, Santana da Silva J, Moreira de Andrade J, 2000, NK cell activity in the presence of IL-12 is a prognostic assay to neoadjuvant chemotherapy in cervical cancer. Gynecol Oncol 78:318–323 Kern DE, Klarnet JP, Jensen MCV, Greenberg PD, 1986, Requirement for recognition of class II and processed tumor antigen for optimal generation of syngeneic tumor-specific class I-restricted CTL. J Immunol 136:4303–4310 Kosmaczewska A, Frydecka I, Bocko D et al, 2002, Correlation of blood lymphocyte CTLA-4, CD152, induction in Hodgkin’s disease with proliferative activity, interleukin 2 and interferon gamma production. Br J Haematol 118:202–209 Carreno BM, Bennett F, Chau TA et al, 2000, CTLA-4, CD152, can inhibit T cell activation by two different mechanisms depending on its level of cell surface expression. J Immunol 165:1352–1356 Scotto L, Naiyer AJ, Galluzzo S et al, 2004, Overlap between molecular markers expressed by naturally occurring CD4+CD25+ regulatory T cells and CD8+CD28- T suppressor cells. Hum Immunol 65:1297–1306 Sansom DM, Walker LS, 2006, The role of CD28 and cytotoxic T-lymphocyte antigen-4, CTLA-4, in regulatory T-cell biology. Immunol Rev 212:131–148 Karagoz B, Bilgi O, Gumus M et al, 2010, CD8+CD28- cells and CD4+CD25+ regulatory T cells in the peripheral blood of advanced stage lung cancer patients. Med Oncol 27:29–33 Mahic M, Henjum K, Yaqub S et al, 2008, Generation of highly suppressive adaptive CD8(+)CD25(+)FOXP3(+, regulatory T cells by continuous antigen stimulation. Eur J Immunol 38:640–646 Zheng SG, Wang JH, Stohl W et al, 2006, TGF-beta requires CTLA-4 early after T cell activation to induce FoxP3 and generate adaptive CD4+CD25+ regulatory cells. J Immunol 176:3321–3329 Takahashi T, Tagami T, Yamazaki S et al, 2000, Immunologic self-tolerance maintained by CD25+CD4+ regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med 192:303–309 Visser J, Nijman HW, Hoogenboom B-N et al, 2007, Frequencies and role of regulatory T cells in patients with, pre)malignant cervical neoplasia. Clin Exp Immunol 150:199–209 June CH, Ledbetter JA, Linsley PS, Thompson CB, 1990, Role of CD28 receptor in T-cell activation. Immunol Today 11:211–216 Filaci G, Fravega M, Negrini S et al, 2004, Nonantigen specific CD8+ T suppressor lymphocytes originate from CD8+CD28- T cells and inhibit both T-cell proliferation and CTL function. Hum Immunol 65:142–156 Chopra V, Dinh TV, Hannigan EV, 1998, Circulating serum levels of cytokines and angiogenic factors in patients with cervical cancer. Cancer Invest 16:152–159 Diaz-Benitez CE, Navarro-Fuentes KR, Flores-Sosa JA et al, 2009, CD3zeta expression and T cell proliferation are inhibited by TGF-beta1 and IL-10 in cervical cancer patients. J Clin Immunol 29:32–544 Mahic M, Yaqub S, Bryn T et al, 2008, Differentiation of naive CD4+ T cells into CD4+CD25+FOXP3+ regulatory T cells by continuous antigen stimulation. J Leukoc Biol 83:1111–1117 Clerici M, Merola M, Ferrario E et al, 1997, Cytokine production patterns in cervical intraepithelial neoplasia: association with human papillomavirus infection. J Natl Cancer Inst 89:245–250 Tartour E, Gey A, Sastre-Garau E et al, 1998, Prognostic value of intratumoral interferon gamma messenger RNA expression in invasive cervical carcinomas. J Natl Cancer Inst 90:287–294 Santin AD, Ravaggi A, Bellone S et al, 2001, Tumor-infiltrating lymphocytes contain higher numbers of type 1 cytokine expressors and DR+T cells compared with lymphocytes from tumor draining lymph nodes and peripheral blood in patients with cancer of the uterine cervix. Gynecol Oncol 81:424–432 Kusuda T, Shigemasa K, Arihiro K et al, 2005, Relative expression levels of Th1 and Th2 cytokine mRNA are independent prognostic factors in patients with ovarian cancer. Oncol Rep 13:1153–1158 Ubukata H, Motohashi G, Tabuchi T et al, 2010, Evaluations of interferon-gamma/interleukin-4 ratio and neutrophil/lymphocyte ratio as prognostic indicators in gastric cancer patients. J Surg Oncol 102:742–747 Teschendorff AE, Gomez S, Arenas A et al, 2010, Improved prognostic classification of breast cancer defined by antagonistic activation patterns of immune response pathway modules. BMC Cancer 10:604 Tosolini M, Kirilovsky A, Mlecnik B et al, 2011, Clinical impact of different classes of infiltrating T cytotoxic and helper cells, Th1, Th2, Treg, and Th17, in patients with colorectal cancer. Cancer Res 71:1263–1271 De Monte L, Reni M, Tassi E et al, 2011, Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med 208:469–478 Eylar EH, Lefranc C, Baez I et al, 2001, Enhanced interferongamma by CD8+CD28- lymphocytes from HIV+patients. J Clin Immunol 21:135–144 Wang YM, Alexander SI, 2009, CD8 regulatory T cells: what’s old is now new? Immunol Cell Biol 87:192–193 Coussens LM, Werb Z, 2002, Inflammation and cancer. Nature 420:860–867 Ito N, Suzuki Y, Taniguchi Y et al, 2005, Prognostic significance of T helper 1 and 2 and T cytotoxic 1 and 2 cells in patients with non-small cell lung cancer. Anticancer Res 25:2027–2032 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 479 EP 489 DI 10.1007/s12253-011-9471-y PG 11 ER PT J AU Luo, D Huang, H Lu, ML Zhao, GF Chang, J Zheng, MY Wang, Y AF Luo, Deng Huang, Hua Lu, Ming-Liang Zhao, Gong-Fang Chang, Jiang Zheng, Meng-Yao Wang, Yan TI Abnormal Expression of Adhesion Protein Bves is Associated with Gastric Cancer Progression and Poor Survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Bves; Cell adhesion molecules; Metastasis; Prognosis ID Gastric cancer; Bves; Cell adhesion molecules; Metastasis; Prognosis AB Although many molecular and biological studies have shown risk factors for gastric cancer, the available knowledge is still insufficient to unveil the exact mechanism of gastric cancer. To investigate the relationships between Bves expression and the clinicopathologic features of gastric cancer and whether Bves can act as prognostic indicators in gastric cancer. Tissues were obtained from the gastrectomy specimens of 306 human gastric cancer and 78 noncancerous gastric tissue at the Department of Surgery and Pathology, the Second Affiliated Hospital of Kunming Medical University from February 1996 to March 2007. The method of immunohistochemistry was used to investigate the expression of Bves in them. The relationship between Bves expression and the survival times of the patients was retrospectively analysed. Reduced expression of Bves frequently occurred in gastric cancer tissue. Low expression of Bves correlated with histologic differentiation, depth of invasion, regional lymph nodes and distant metastasis, and TNM stages (P<0.05). Bves expression did not correlate with age, gender, location of tumor, size of tumor and histologic type (P>0.05); Further multivariate analysis revealed that lymph node metastasis (P<0.0001), distant metastasis (P<0.0001), surgical treatment (P<0.0001), and the expression of Bves (P<0.0001) were independent prognostic factors in patients with gastric cancer; The Kaplan-Meier plot showed that survival times of patients with low Bves expression was significantly lower than those in patients with high Bves expression. Besides, low Bves expression had a much more significant effect on the survival of those patients with early stage tumors (χ2=131.216,P<0.0001), highlighted by a >51.3% reduction in 3-year survival compared with that of patients with high Bves expression. In late stages, the difference was also significant (χ2=5.818,P=0.016), with a 34.8% reduction in 3-year survival. Reduced expression of Bves in gastric cancer is associated with tumor progression and the patient’s poor survival. This study showed that the studied protein has further provided a basis for the development of potential biomarker for gastric cancer prognosis. C1 [Luo, Deng] the Second Affiliated Hospital of Kunming Medical University, Department of Gastroenterology, No. 1, MaYuan Road, XiShan District, 650101 Kunming, Yunnan Province, China. [Huang, Hua] the Second Affiliated Hospital of Kunming Medical University, Department of Gastroenterology, No. 1, MaYuan Road, XiShan District, 650101 Kunming, Yunnan Province, China. [Lu, Ming-Liang] the Second Affiliated Hospital of Kunming Medical University, Department of Gastroenterology, No. 1, MaYuan Road, XiShan District, 650101 Kunming, Yunnan Province, China. [Zhao, Gong-Fang] the Second Affiliated Hospital of Kunming Medical University, Department of Gastroenterology, No. 1, MaYuan Road, XiShan District, 650101 Kunming, Yunnan Province, China. [Chang, Jiang] the Second Affiliated Hospital of Kunming Medical University, Department of Gastroenterology, No. 1, MaYuan Road, XiShan District, 650101 Kunming, Yunnan Province, China. [Zheng, Meng-Yao] the Second Affiliated Hospital of Kunming Medical University, Department of Gastroenterology, No. 1, MaYuan Road, XiShan District, 650101 Kunming, Yunnan Province, China. [Wang, Yan] the Second Affiliated Hospital of Kunming Medical University, Department of PathologyKunming, Yunnan Province, China. RP Huang, H (reprint author), the Second Affiliated Hospital of Kunming Medical University, Department of Gastroenterology, 650101 Kunming, China. EM 122068863@qq.com CR Ajani JA, Barthel JS, Bekaii-Saab T, Bentrem DJ, D'Amico TA, Das P, Denlinger C, Fuchs CS, Gerdes H, Hayman JA, Hazard L, Hofstetter WL, Ilson DH, Keswani RN, Kleinberg LR, Korn M, Meredith K, Mulcahy MF, Orringer MB, Osarogiagbon RU, Posey JA, Sasson AR, Scott WJ, Shibata S, Strong VE, Washington MK, Willett C, Wood DE, Wright CD, Yang G, 2010, Gastric cancer. J Natl Compr Canc Netw 8(4):378–409 Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics, 2009. CA Cancer J Clin 59(4):225–249., DOI 10.3322/ caac.20006 Ye YW, Dong RZ, Zhou Y, Du CY, Wang CM, Fu H, Shi YQ, 2011, Prognostic analysis of familial gastric cancer in Chinese population. J Surg Oncol 104(1):76–82., DOI 10.1002/jso.21896 Thun M, Jemal A, Desantis C, Blackard B, Ward E, 2009, An overview of the cancer burden for primary care physicians. Prim Care 36(3):439–454., DOI 10.1016/j.pop.2009.04.001 Xishan H, Chen K, Min H, Shufen D, Jifang W, 2011, Cancer survival in Tianjin, China, 1991–1999. IARC Sci Publ 162:69–84 Xiang YB, Jin F, Gao YT, 2011, Cancer survival in Shanghai, China, 1992–1995. IARC Sci Publ 162:55–68 Law SC, Mang OW, 2011, Cancer survival in Hong Kong SAR, China, 1996–2001. IARC Sci Publ 162:33–41 Chen JG, Zhu J, Zhang YH, Lu JH, 2011, Cancer survival in Qidong, China, 1992–2000. IARC Sci Publ 162:43–53 Reese DE, Zavaljevski M, Streiff NL, Bader D, 1999, bves: a novel gene expressed during coronary blood vessel development. Dev Biol 209(1):159–171., DOI 10.1006/dbio.1999.9246 Andree B, Hillemann T, Kessler-Icekson G, Schmitt-John T, Jockusch H, Arnold HH, Brand T, 2000, Isolation and characterization of the novel popeye gene family expressed in skeletal muscle and heart. Dev Biol 223(2):371–382., DOI 10.1006/dbio.2000.9751 DiAngelo JR, Vasavada TK, Cain W, Duncan MK, 2001, Production of monoclonal antibodies against chicken Pop1, BVES). Hybrid Hybridomics 20(5–6):377–381., DOI 10.1089/ 15368590152740789 Vasavada TK, DiAngelo JR, Duncan MK, 2004, Developmental expression of Pop1/Bves. J Histochem Cytochem 52(3):371–377 Ripley AN, Osler ME, Wright CV, Bader D, 2006, Xbves is a regulator of epithelial movement during early Xenopus laevis development. Proc Natl Acad Sci USA 103(3):614–619., DOI 10.1073/pnas.0506095103 Lin S, Zhao D, Bownes M, 2007, Blood vessel/epicardial substance, bves, expression, essential for embryonic development, is down regulated by Grk/EFGR signalling. Int J Dev Biol 51(1):37–44., DOI 10.1387/ijdb.052108sl Knight RF, Bader DM, Backstrom JR, 2003, Membrane topology of Bves/Pop1A, a cell adhesion molecule that displays dynamic changes in cellular distribution during development. J Biol Chem 278(35):32872–32879., DOI 10.1074/jbc.M301961200 Osler ME, Bader DM, 2004, Bves expression during avian embryogenesis. Dev Dyn 229(3):658–667., DOI 10.1002/ dvdy.10490 Gingold-Belfer R, Bergman M, Alcalay Y, Schlesinger H, Aravot D, Berman M, Salman H, Brand T, Kessler-Icekson G, 2011, Popeye domain-containing 1 is down-regulated in failing human hearts. Int J Mol Med 27(1):25–31., DOI 10.3892/ijmm.2010.558 Feng Q, Hawes SE, Stern JE, Wiens L, Lu H, Dong ZM, Jordan CD, Kiviat NB, Vesselle H, 2008, DNA methylation in tumor and matched normal tissues from non-small cell lung cancer patients. Cancer Epidemiol Biomarkers Prev 17(3):645–654., DOI 10.1158/ 1055-9965.EPI-07-2518 Kim M, Jang HR, Haam K, Kang TW, Kim JH, Kim SY, Noh SM, Song KS, Cho JS, Jeong HY, Kim JC, Yoo HS, Kim YS, 2010, Frequent silencing of popeye domain-containing genes, BVES and POPDC3, is associated with promoter hypermethylation in gastric cancer. Carcinogenesis 31(9):1685–1693., DOI 10.1093/carcin/bgq144 Jayagopal A, Yang JL, Haselton FR, Chang MS, 2011, Tight junction-associated signaling pathways modulate cell proliferation in uveal melanoma. Invest Ophthalmol Vis Sci 52(1):588–593., DOI 10.1167/iovs.10-5746 Santiago JM, Sasako M, Osorio J, 2011, [TNM-7th edition 2009, UICC/AJCC, and Japanese Classification 2010 in Gastric Cancer. Towards simplicity and standardisation in the management of gastric cancer]. Cir Esp 89(5):275–281., DOI 10.1016/j. ciresp.2010.10.011 Washington K, 2010, 7th edition of the AJCC cancer staging manual: stomach. Ann Surg Oncol 17(12):3077–3079., DOI 10.1245/s10434-010-1362-z Russ PK, Pino CJ, Williams CS, Bader DM, Haselton FR, Chang MS, 2011, Bves modulates tight junction associated signaling. PLoS One 6(1):e14563., DOI 10.1371/journal.pone.0014563 Russ PK, Kupperman AI, Presley SH, Haselton FR, Chang MS, 2010, Inhibition of RhoA signaling with increased Bves in trabecular meshwork cells. Invest Ophthalmol Vis Sci 51(1):223– 230., DOI 10.1167/iovs.09-3539 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 491 EP 497 DI 10.1007/s12253-011-9472-x PG 7 ER PT J AU Ribeiro, MA Pereira, S Andrade, S Costa, M Lopes, C Aguas, PA Monteiro, PM AF Ribeiro, M Andreia Pereira, Sofia Andrade, Sara Costa, Madalena Lopes, Carlos Aguas, P Artur Monteiro, P Mariana TI Insulin Prevents Leptin Inhibition of RM1 Prostate Cancer Cell Growth SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Obesity; Leptin; Insulin; RM1 cells; Cellular proliferation ID Prostate cancer; Obesity; Leptin; Insulin; RM1 cells; Cellular proliferation AB The association between obesity and cancer is controversial: whereas several epidemiological, clinical and research studies using cancer cell lines have supported that high levels of insulin and leptin could favor prostate cancer development and dissemination, other studies have demonstrated opposite effects or even absence of association. The main goal of this study was to evaluate the in vitro proliferation of murine androgen insensitive prostate carcinoma cells RM1 in the presence of leptin and insulin. After assessing and confirming the presence of leptin and insulin receptors in RM1 cells by immunocytochemistry, cells were cultured in the presence of different concentrations of leptin (0, 25, 50, 100 and 200 ng/mL), insulin (0, 50, 100, 150 and 200 nM) or leptin plus insulin ( 25 ng/ml+50 nM; 50 ng/ml+100 nM; 100 ng/ml+150 nM; 200 ng/ml+200 nM; 25 ng/ml+150 nM; 100 ng/ml+50 nM of leptin plus insulin, respectively). Cell proliferation was evaluated by assessing the percentage of resazurin reduction, a surrogate marker of cell metabolic rate. Leptin significantly decreased the percentage of resazurin reduction in all studied concentrations while there was only a slight or non significant difference in RM1cell proliferation in the presence of insulin or insulin combined with leptin when compared with control. These results show that leptin decreases RM1 prostate cancer cell proliferation at the studied concentrations, while insulin is able to antagonize the leptin inhibition of RM1 prostate cancer cell growth in vitro. The difference in cell growth that is modulated by the various hormonal environments may explain the heterogeneous behavior of prostate cancer in the obese human population. C1 [Ribeiro, M Andreia] University of Porto, Clinical and Experimental Endocrinology, Department of Anatomy, 4099-003 Porto, Portugal. [Pereira, Sofia] University of Porto, Clinical and Experimental Endocrinology, Department of Anatomy, 4099-003 Porto, Portugal. [Andrade, Sara] University of Porto, Clinical and Experimental Endocrinology, Department of Anatomy, 4099-003 Porto, Portugal. [Costa, Madalena] University of Porto, Clinical and Experimental Endocrinology, Department of Anatomy, 4099-003 Porto, Portugal. [Lopes, Carlos] University of Porto, Department of Pathology and Immunology of ICBAS, 4099-003 Porto, Portugal. [Aguas, P Artur] University of Porto, Clinical and Experimental Endocrinology, Department of Anatomy, 4099-003 Porto, Portugal. [Monteiro, P Mariana] University of Porto, Clinical and Experimental Endocrinology, Department of Anatomy, 4099-003 Porto, Portugal. RP Monteiro, PM (reprint author), University of Porto, Clinical and Experimental Endocrinology, Department of Anatomy, 4099-003 Porto, Portugal. EM mpmonteiro@icbas.up.pt CR Kumar RJ, Barqawi AB, Craw ED, 2008, Epidemiology of Prostate Cancer. US Oncology Review 2005 Schaid DJ, 2004, The complex genetic epidemiology of prostate cancer. Hum Mol Genet 13(1):103–121 Mukherjee P, Sotnikov AV, Mangian HJ et al, 1999, Energy intake and prostate tumor growth, angiogenesis, and vascular endothelial growth factor expression. J Natl Cancer Inst 91, 6):512–523 Bosland MC, Oakley-Girvan I, Whittemore AS, 1999, Dietary fat, calories, and prostate cancer risk. J Natl Cancer Inst 91(6):489– 491 Friedenreich CM, Orenstein MR, 2002, Physical activity and cancer prevention: etiologic evidence and biological mechanisms. J Nutr 132(11 Suppl):3456S–3464S Calle EE, Kaaks R, 2004, Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer 4(8):579–591 Crespo CJ, Smit E, Troiano RP et al, 2001, Television watching, energy intake, and obesity in US children: results from the third National Health and Nutrition Examination Survey, 1988–1994. Arch Pediatr Adolesc Med 155(3):360–365 Stubbs CO, Lee AJ, 2004, The obesity epidemic: both energy intake and physical activity contribute. Med J Aust 181(9):489– 491 Mistry T, Digby JE, Desai KM et al, 2007, Obesity and prostate cancer: a role for adipokines. Eur Urol 52(1):46–53 Garg A, 2006, Adipose tissue dysfunction in obesity and lipodystrophy. Clin Cornerstone 8(Suppl 4):S7–S13 Trayhurn P, Beattie JH, 2001, Physiological role of adipose tissue: white adipose tissue as an endocrine and secretory organ. Proc Nutr Soc 60(3):329–339 Kershaw EE, Flier JS, 2004, Adipose tissue as an endocrine organ. J Clin Endocrinol Metab 89(6):2548–2556 Ronti T, Lupattelli G, Mannarino E, 2006, The endocrine function of adipose tissue: an update. Clin Endocrinol, Oxf, 64(4):355–365 Wajchenberg BL, 2000, Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr Rev 21(6):697– 738 Haque WA, Garg A, 2004, Adipocyte biology and adipocytokines. Clin Lab Med 24(1):217–234 Tilg H, Moschen AR, 2006, Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol 6, 10):772–783 Badman MK, Flier JS, 2007, The adipocyte as an active participant in energy balance and metabolism. Gastroenterology 132(6):2103–2115 Zhang Y, Proenca R, Maffei M et al, 1994, Positional cloning of the mouse obese gene and its human homologue. Nature 372, 6505):425–432 Frederich RC, Hamann A, Anderson S et al, 1995, Leptin levels reflect body lipid content in mice: evidence for diet-induced resistance to leptin action. Nat Med 1(12):1311–1314 Boden G, Chen X, Mozzoli M et al, 1996, Effect of fasting on serum leptin in normal human subjects. J Clin Endocrinol Metab 81(9):3419–3423 Bjorbaek C, Uotani S, da Silva B et al, 1997, Divergent signaling capacities of the long and short isoforms of the leptin receptor. J Biol Chem 272(51):32686–32695 Tartaglia LA, 1997, The leptin receptor. J Biol Chem 272, 10):6093–6096 Hoggard N, Hunter L, Duncan JS et al, 1997, Leptin and leptin receptor mRNA and protein expression in the murine fetus and placenta. Proc Natl Acad Sci U S A 94(20):11073–11078 Bailleul B, Akerblom I, Strosberg AD, 1997, The leptin receptor promoter controls expression of a second distinct protein. Nucleic Acids Res 25(14):2752–2758 Somasundar P, Frankenberry KA, Skinner H et al, 2004, Prostate cancer cell proliferation is influenced by leptin. J Surg Res 118, 1):71–82 Banting FG, 1926, An Address on Diabetes and Insulin: Being The Nobel Lecture Delivered at Stockholm on September 15th, 1925. Can Med Assoc J 16(3):221–232 Karam JH, Grodsky GM, Forsham PH, 1968, Insulin secretion in obesity: pseudodiabetes? Am J Clin Nutr 21(12):1445–1454 Kahn BB, Flier JS, 2000, Obesity and insulin resistance. J Clin Invest 106(4):473–481 Cox ME, Gleave ME, Zakikhani M et al, 2009, Insulin receptor expression by human prostate cancers. Prostate 69(1):33–40 Cullen KJ, Yee D, Sly WS et al, 1990, Insulin-like growth factor receptor expression and function in human breast cancer. Cancer Res 50(1):48–53 Frasca F, Pandini G, Sciacca L et al, 2008, The role of insulin receptors and IGF-I receptors in cancer and other diseases. Arch Physiol Biochem 114(1):23–37 Baillargeon J, Rose DP, 2006, Obesity, adipokines, and prostate cancer, review). Int J Oncol 28(3):737–745 Danforth KN, Rodriguez C, Hayes RB et al, 2008, TNF polymorphisms and prostate cancer risk. Prostate 68(4):400–407 Ribeiro AM, Andrade S, Pinho F et al, 2010, Prostate cancer cell proliferation and angiogenesis in different obese mice models. Int J Exp Pathol 91(4):374–386 Baley PA, Yoshida K, Qian W et al, 1995, Progression to androgen insensitivity in a novel in vitro mouse model for prostate cancer. J Steroid Biochem Mol Biol 52(5):403–413 Freedland SJ, Giovannucci E, Platz EA, 2006, Are findings from studies of obesity and prostate cancer really in conflict? Cancer Causes Control 17(1):5–9 Giovannucci E, Rimm EB, Liu Y et al, 2003, Body mass index and risk of prostate cancer in U.S. health professionals. J Natl Cancer Inst 95(16):1240–1244 Malendowicz W, Rucinski M, Macchi C et al, 2006, Leptin and leptin receptors in the prostate and seminal vesicles of the adult rat. Int J Mol Med 18(4):615–618 Hoda MR, Popken G, 2008, Mitogenic and anti-apoptotic actions of adipocyte-derived hormone leptin in prostate cancer cells. BJU Int 102(3):383–388 Deo DD, Rao AP, Bose SS et al, 2008, Differential effects of leptin on the invasive potential of androgen-dependent and - independent prostate carcinoma cells. J Biomed Biotechnol 2008:163902 Hsing AW, Chua S Jr, Gao YT et al, 2001, Prostate cancer risk and serum levels of insulin and leptin: a population-based study. J Natl Cancer Inst 93(10):783–789 Kelly T, Yang W, Chen CS et al, 2008, Global burden of obesity in 2005 and projections to 2030. Int J Obes, Lond, 32(9):1431–1437 Farooqi IS, O’Rahilly S, 2009, Leptin: a pivotal regulator of human energy homeostasis. Am J Clin Nutr 89(3):980S–984S Belkina AC, Denis GV, 2010, Obesity genes and insulin resistance. Curr Opin Endocrinol Diabetes Obes 17(5):472–477 Muoio DM, Newgard CB, 2008, Mechanisms of disease: molecular and metabolic mechanisms of insulin resistance and beta-cell failure in type 2 diabetes. Nat Rev Mol Cell Biol 9, 3):193–205 Wotton CJ, Yeates DG, Goldacre MJ, 2011, Cancer in patients admitted to hospital with diabetes mellitus aged 30 years and over: record linkage studies. Diabetologia 54(3):527–534 Polednak AP, Phillips CE, 2010, Obtaining data on comorbid diabetes among patients in a U.S. population-based tumor registry. J Registry Manag 37(2):57–64 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 499 EP 507 DI 10.1007/s12253-011-9473-9 PG 9 ER PT J AU Hasby, AE AF Hasby, Adel Eiman TI Weapons Ovarian Epithelial Tumors May Use in Immune Escape: An Immunohistochemical Correlational Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE FasL; Survivin; CD3+ T-lymphocytes; Ovarian epithelial tumors; Immune escape ID FasL; Survivin; CD3+ T-lymphocytes; Ovarian epithelial tumors; Immune escape AB Investigate FasL and survivin expression in a series of primary ovarian surface epithelial tumors, correlate their expression with each other, and characterize the presence of CD3+ T-lymphocytes in studied tumors and determine whether their presence correlates with FasL or survivin expression in malignant cases. FasL and survivin expression was assessed in 54 ovarian epithelial tumors. The results were compared between different tumor types and grades. Correlation between both markers’ expression in all studied tumors was done. Either marker’s expression was compared to the mean CD3+ T-lymphocytes per HPF in the studied malignant tumors. Either FasL or survivin expression was significantly higher in malignant versus benign ovarian epithelial tumors (p<0.001 for both) and both markers were strongly correlated to each other (r=0.877 & p<0.001). Malignant tumors show significantly higher mean CD3+ T-lymphocytes than benign and borderline tumors. The mean CD3+ T-lymphocytes decrease significantly on increasing malignant tumor grade (p=0.019) and expression of both FasL and survivin (r=−0.729, -0.582, respectively & p<0.001 for both). The higher expression of FasL and survivin in malignant as compared to benign ovarian tumors suggest that they have a significant role in pathogenesis of ovarian carcinoma. Both markers are strongly correlated to each other and may contribute to immune escape of ovarian carcinoma as their higher expression is associated with decreased number of CD3+T-lymphocytes. C1 [Hasby, Adel Eiman] Tanta University, Faculty of Medicine, Department of PathologyTanta, Ghrbia, Egypt. RP Hasby, AE (reprint author), Tanta University, Faculty of Medicine, Department of Pathology, Tanta, Egypt. EM eimanhasby@yahoo.com CR Ahmad M, Rees RC, Ali SA, 2004, Escape from immunotherapy: possible mechanisms that influence tumor regression/progression. Cancer Immunol Immunother 53:844–854 Ryan AE, Shanahan F, O’Connell J, Houston AM, 2005, Addressing the “Fas counterattack” controversy: blocking fas ligand expression suppresses tumor immune evasion of colon cancer in vivo. Cancer Res 65(21):9817–9823 Suda T, Takahashi T, Golstein P, Nagata S, 1993, Molecular cloning and expression of the Fas ligand, a novel member of the tumor necrosis factor family. Cell 75:1169–1178 Nagata S, Golstein P, 1995, The Fas death factor. Science 267:1449–1456 Griffith TS, Ferguson TA, 1997, The role of FasL-induced apoptosis in immune privilege. Immunol Today 18:240–244 O’Connell J, O’Sullivan GC, Collins JK, Shanahan F, 1996, The Fas counterattack: Fas-mediated T cell killing by colon cancer cells expressing Fas ligand. J Exp Med 184:1075–1082 Gutierrez LS, Eliza M, Niven-Fairchild T, Naftolin F, Mor G, 1999, The Fas/Fas-ligand system: a mechanism for immune evasion in human breast carcinomas. Breast Cancer Res Treat 54:245–253 Restifo NP, 2000, Not so Fas: re-evaluating the mechanisms of immune privilege and tumor escape. Nat Med 6:493–495 O’Connell J, Houston A, Bennett MW, O’Sullivan GC, Shanahan F, 2001, Immune privilege or inflammation? Insights into the Fas ligand enigma. Nat Med 7:271–274 Saikumar P, Dong Z, Mikhailov V, Denton M, Weinberg JM, Venkatachalam MA, 1999, Apoptosis: definition, mechanisms, and relevance to disease. Am J Med 107:489–506 Jaattela M, 1999, Escaping cell death: survival proteins in cancer. Exp Cell Res 248:30–43 Tamm I, Wang Y, Sausville E et al, 1998, IAP-family protein survivin inhibits caspase activity and apoptosis induced by Fas, CD95), Bax, caspases, and anticancer drugs. Cancer Res 58:5315–5320 Conway EM, Pollefeyt S, Steiner-MosonyiMet al, 2002, Deficiency of survivin in transgenic mice exacerbates Fas-induced apoptosis via mitochondrial pathways. Gastroenterology 123:619–631 Ambrosini G, Adida C, Altieri DC, 1997, A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 3:917–921 Yoshida H, Ishiko O, Sumi T, Matsumoto Y, Ogita S, 2001, Survivin, bcl-2 and matrix metalloproteinase-2 enhance progression of clear cell- and serous-type ovarian carcinomas. Int J Oncol 19:537–542 Sui L, Dong Y, Ohno M, Watanabe Y, Sugimoto K, Tokuda M, 2002, Survivin expression and its correlation with cell proliferation and prognosis in epithelial ovarian tumors. Int J Oncol 21:315–320 Takai N, Miyazaki T, Nishida M, Nasu K, Miyakawa I, 2002, Expression of survivin is associated with malignant potential in epithelial ovarian carcinoma. Int J Mol Med 10:211–216 Zaffaroni N, Pennati M, Colella G et al, 2002, Expression of the anti-apoptotic gene survivin correlates with taxol resistance in human ovarian cancer. Cell Mol Life Sci 59:1406–1412 Cohen C, Lohmann CM, Cotsonis G, Lawsin D, Santoianni R, 2003, Survivin expression in ovarian carcinoma: correlation with apoptotic markers and prognosis. Mod Pathol 16:574– 583 Jemal A, Siegel R, Ward E et al, 2008, Cancer statistics, 2008. CA Cancer J Clin 58:71–96 Lee KR, Tavassoli FA, Prat J, Dietel M, 2003, Surface epithelial-stromal tumours. In: Tavassoli FA, Deville P, eds, WHO classification of tumours, pathology and genetics of tumours of the breast and female genital organs. IARC, Lyon, pp 117–145 Munakata S, Enomoto T, Tsujimoto M et al, 2000, Expressions of Fas ligand and other apoptosis-related genes and their prognostic significance in epithelial ovarian neoplasms. Br J Cancer 82:1446–1452 Zhang YL, Li J, Mo HY et al, 2010). Different subsets of tumor infiltrating lymphocytes correlate with NPC progression in different ways. Mol Cancer 10: 9:4 Igney FH, Krammer PH, 2002, Immune escape of tumors: apoptosis resistance and tumor counterattack. J Leukoc Biol 71:907–920 Reed JC, 2001, The Survivin saga goes in vivo. J Clin Investig 108:965–969 Krammer PH, 1997, The tumor strikes back. Cell Death Differ 4:362–364 Igney FH, Behrens CK, Krammer PH, 2000, Tumor counterattack —concept and reality. Eur J Immunol 30:725–731 Walker PR, Saas P, Dietrich PY, 1998, Tumor expression of Fas ligand, CD95L, and the consequences. Curr Opin Immunol 10:564–572 Ma XY, He FX, Wu SF, Lu YP, Ma D, 2004, Expression of survivin in ovarian epithelial carcinoma and its correlation with expression of Fas and FasL. Ai Zheng 23(2):173–176 Arts HJ, de Jong S, Hollema H, Ten Hoor KA, de Vries EG, van der Zee AG, 2005, Fas and Fas ligand in cyst fluids, serum and tumors of patients with benign and, borderline, malignant ovarian tumors. Int J Oncol 26:379–384 Zusman I, Gurevich P, Gurevich E et al, 2001, The immune system, apoptosis and apoptosis-related proteins in human ovarian tumours, a review). Int J Oncol 18:965–972 van Haaften-Day C, Russell P, Davies S et al, 1992, An in vitro study of ovarian atypical proliferating, borderline, serous tumors. Int J Gynecol Cancer 2:41–48 Ben-Hur H, Gurevich P, Huszar M et al, 1999, Apoptosis and apoptosis-related proteins in the epithelium of human ovarian tumors: immunohistochemical and morphometric studies. Eur J Gynaecol Oncol 20(4):249–253 Ferrandina G, Legge F, Martinelli E et al, 2005, Survivin expression in ovarian cancer and its correlation with clinicopathological, surgical and apoptosis-related parameters. Br J Cancer 92:271–277 Asanuma K, Tsuji N, Endoh T, Yagihashi A, Watanabe N, 2004, Survivin enhances Fas ligand expression via up-regulation of specificity protein 1-mediated gene transcription in colon cancer cells. J Immunol 172:3922–3929 Parmiani G, 2005, Tumor-infiltrating T cells–friend or foe of neoplastic cells? N Engl J Med 353:2640–2641 Dieu-Nosjean MC, Antoine M, Danel C et al, 2008, Long-term survival for patients with non-small cell lung cancer with intratumoral lymphoid structures. J Clin Oncol 26:4410–4417 Galon J, Costes A, Sanchez-Cabo F et al, 2006, Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313:1960–1964 Sheu BC, Kuo WH, Chen RJ, Huang SC, Chang KJ, Chow SN, 2008, Clinical significance of tumor-infiltrating lymphocytes in neoplastic progression and lymph node metastasis of human breast cancer. Breast 17:604–610 Zhang L, Conejo-Garcia JR, Katsaros D et al, 2003, Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 348:203–213 Curiel TJ, Coukos G, Zou L et al, 2004, Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 10:942–949 Raspollini MR, Castiglione F, Rossi Degl’innocenti D et al, 2005, Tumour-infiltrating gamma/delta T-lymphocytes are correlated with a brief disease-free interval in advanced ovarian serous carcinoma. Ann Oncol 16:590–596 Sato E, Olson SH, Ahn J et al, 2005, Intraepithelial CD8+ tumor infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA 102:18538–18543 Hamanishi J, Mandai M, Iwasaki M et al, 2007, Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci USA 104:3360–3365 Tomsova M, Melichar B, Sedlakova I, Steiner I, 2008, Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma. Gynecol Oncol 108:415–420 Clarke B, Tinker AV, Lee CH et al, 2009, Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type, and BRCA1 loss. Mod Pathol 22:393–402 Leffers N, Gooden MJ, de Jong RA et al, 2009, Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer. Cancer Immunol Immunother 58:449–459 Stumpf M, Hasenburg A, Riener MO et al, 2009, Intraepithelial CD8-positive T lymphocytes predict survival for patients with serous stage III ovarian carcinomas: relevance of clonal selection of T lymphocytes. Br J Canc 101:1513–1521 Tel-A H, Alla AE, Laban MA, Fahmy RM, 2004, Immunophenotyping of tumor-infiltrating mononuclear cells in ovarian carcinoma. Pathol Oncol Res 10:80–84 Kabawat SE, Bast RC, Welch WR et al, 1983, Expression of major histocompatibility antigens and nature of inflammatory cellular infiltrate in ovarian neoplasms. Int J Cancer 32:547– 554 Kullander S, Rausing A, 1994, Phenotypic characteristics of mononuclear cells in human ovarian tumors – with special reference to cystic and ascitic fluid. Int J Gynecol Cancer 4:298–305 Negus R, Stamp GW, Hadley J, Balkwill, 1997, Quantitative assessment of the leukocyte infiltrate in ovarian cancer and its relationship to the expression of C-C chemokines. Am J Pathol 150:1723–1732 Puccetti L, Manetti R, Parronchi P et al, 2002, Role of low nuclear grading of renal carcinoma cells in the functional profile of tumor-infiltrating T cells. Int J Cancer 98:674–681 Okada K, Komuta K, Hashimoto S, Matsuzaki S, Kanematsu T, Koji T, 2000, Frequency of apoptosis of tumor infiltrating lymphocytes induced by fas counterattack in human colorectal carcinoma and its correlation with prognosis. Clin Cancer Res 6:3560–3564 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 509 EP 518 DI 10.1007/s12253-011-9474-8 PG 10 ER PT J AU Mehes, G Hegyi, K Csonka, T Fazakas, F Kocsis, Zs Radvanyi, G Vadnay, I Bagdi, E Krenacs, L AF Mehes, Gabor Hegyi, Katalin Csonka, Tamas Fazakas, Ferenc Kocsis, Zsolt Radvanyi, Gaspar Vadnay, Istvan Bagdi, Eniko Krenacs, Laszlo TI Primary Uterine NK-Cell Lymphoma, Nasal-Type: A Unique Malignancy of a Prominent Cell Type of the Endometrium SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Aggressive lymphoma; Uterine NK-cells; Angiogenesis; EBV; Dissemination ID Aggressive lymphoma; Uterine NK-cells; Angiogenesis; EBV; Dissemination AB Natural killer (NK) cells host in the human endometrium with dedicated role in reproductive physiology. Interestingly, malignant transformation of these specialized cells has not been presented thus far. Here we report a primary endometrial NK-cell lymphoma of a 48 year-old patient presenting with irregular bleeding. The endometrial curetting showed a dense lymphomatous infiltrate demonstrating highly infiltrative aggressive features with characteristic angiocentric, partially angiodestructive growth pattern and accompanying focal necroses. The lymphoma cells displayed a CD3ε/CD56/TIA-1/granzyme-B-positive and CD5/CD4/CD8/TCRγδ-negative immunophenotype, proved to be positive for Epstein-Barr virus by EBER in situ hybridization, and revealed no clonal T-cell receptor gene rearrangement. The diagnosis of uterine extranodal NK-cell lymphoma, nasal-type was made. Clinically, the disease was limited to the uterus at diagnosis, but progressed rapidly, and the patient died within 5 months due disseminated lymphoma, irrespective of intensive chemotherapy. Genuine NK-cell lymphomas occurring in the uterus as primary site seem to be rare making the therapeutic decisions extremely complicated. C1 [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Hegyi, Katalin] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Csonka, Tamas] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Fazakas, Ferenc] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Kocsis, Zsolt] Kazincbarcika City Hospital, Department of PathologyKazincbarcika, Hungary. [Radvanyi, Gaspar] Semmelweis Hospital, Department of HematologyMiskolc, Hungary. [Vadnay, Istvan] County Hospital of Borsod-Abauj-Zemplen, Department of PathologyMiskolc, Hungary. [Bagdi, Eniko] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Krenacs, Laszlo] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. RP Mehes, G (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, 4032 Debrecen, Hungary. EM gabor.mehes@dote.hu CR Chan JKC, Quintanilla-Martinez L, Ferry JA, Peh SC, 2008, Extranodal NK/T-cell lymphoma, nasal type, in: WHO classification of tumours of haemopoietic and lymphoid tissues. IARC, Lyon, pp 285–288 Kim TM, Heo DS, 2009, Extranodal NK/T-cell lymphoma, nasal type: new staging system and treatment strategies. Cancer Sci 100, 12):2242–2248 Au YW, Weisenburger DD, Intragumtornchai T, Nakamura S, Kim WS, SNG I, Vose J, Armitage JO, Liang R, 2009, Clinical differences between nasal and extranasal natural killer/T-cell lymphoma: a study of 136 cases from the International Peripheral T-cell Lymphoma Project. Blood 113(17):3931–3937 Ohshima K, Liu Q, Koga T, Suzumiya J, Kikuchi M, 2002, Classification of cell lineage and anatomical site and prognosis of extranodal T-cell lymphoma—natural killer cell, cytotoxic Tlymphocyte and non NK/CTL types. Virchows Arch 440, 4):425–435 Huang Y, DeReynies A, De Laval L, Ghazi B, Martin-Garcia N, Travert M, Bosq J, Briere J, Petit B, Thomas E, Coppo P, Marafioti T, Emile JF, Delfau-Laure MH, Schmitt C, Gaulard P, 2010, Gene expression profiling identifies emerging oncogenic pathways operating in extranodal NK/T-cell lymphoma, nasaltype. Blood 115(6):1226–1237 Kohrt H, Advani R, 2009, Extranodal natural killer/T-cell lymphoma: current concepts in biology and treatment. Leuk Lymphoma 50(11):1773–1784 Briese J, Noack F, Harland A, Horny HP, 2006, Primary extranodal NK/T cell lymphoma, nasal type, of the endometrium: report of an unusual case diagnosed at autopsy. Gynecol Obstet Invest 61(3):164–166 Mhawech P, Medeiros LJ, Bueso-Ramos C, Coffey DM, Gei AF, Shahab I, 2000, Natural killer-cell lymphoma involving the gynecologic tract. Arch Pathol Lab Med 124(10):1510–1513 Nakamura S, Kato M, Ichimura K, Yatabe Y, Kagami Y, Suzuki R, Taji H, Kondo E, Asakura S, Kojima M, Murakami S, Yamao K, Tsuzuki T, Adachi GK, Miwa A, Yoshidai T, 2001, Peripheral T/naural killer-cell lymphoma involving the female genital tract: a clinicopathologic study of 5 cases. Int J Hematol 73(1):108–114 Kim TM, Park YH, Lee SY, Kim JH, Kim DW, Im SA, Kim TY, Kim CW, Heo DS, Bang YJ, Chang KH, Kim NK, 2005, Local tumor invasiveness is more predictive of survival than International Prognostic Index in stage I(E)/II(E, extranodal NK/T-cell lymphoma, nasal type. Blood 106(12):3785–3790 Manaster I, Mandelboim O, 2010, The unique properties of uterine NK-cells. Am J Reprod Immunol 63(6):434–444 Kalkunte S, Chichester CO, Gotsch F, Sentman CL, Romero R, Sharma S, 2008, Evolution of non-cytotoxic uterine natural killer cells. Am J Reprod Immunol 59(5):425–432 van den Heuvel M, Peralta C, Bashar S, Taylor S, Horrocks J, Cray BA, 2005, Trafficking of peripheral blood CD56(bright, cells to the decidualizing uterus—new tricks for old dogmas. J Reprod Immunol 67(1–2):21–34 van den Heuvel M, Chantakru S, Xuemei X, Evans SS, Tekpetey F, Mote PA, Clarke CL, Croy BA, 2005, Trafficking of circulating pro-NK-cells to the decidualizing uterus: regulatory mechanisms in the mouse and human. Immunol Invest 34(3):273–293 Quenby S, Nik H, Innes B, Lash G, Turner M, Drury J, Blumer J, 2009, Uterine natural killer cells and angiogenesis in recurrent reproductive failure. Hum Reprod 24(1):45–54 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 519 EP 522 DI 10.1007/s12253-011-9360-4 PG 4 ER PT J AU Giordano, G D’Adda, T Bottarelli, L Lombardi, M Brigati, F Berretta, R Merisio, C AF Giordano, Giovanna D’Adda, Tiziana Bottarelli, Lorena Lombardi, Mariano Brigati, Francesca Berretta, Roberto Merisio, Carla TI Two Cases of Low-Grade Endometriod Carcinoma Associated with Undifferentiated Carcinoma of the Uterus (Dedifferentiated Carcinoma): A Molecular Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Endometrial undifferentiated carcinoma; Dedifferentiated carcinoma; Microsatellite Instability; Loss of heterozygosity ID Endometrial undifferentiated carcinoma; Dedifferentiated carcinoma; Microsatellite Instability; Loss of heterozygosity AB Dedifferentiated carcinoma (DC) is an uterine neoplasm containing both low-grade endometrioid carcinoma (LGEC) and undifferentiated carcinoma (UC). DC is an aggressive tumour even when the UC component represents only 20% of the entire neoplasm. In this paper, two cases DCs at different stages of development, in 61- and 83-yearold women respectively were reported. In addition, in these uterine malignancies microsatellite instability (MSI) and loss of heterozygosity (LOH) were investigated in order to explain its aggressive behavior, in both components. Case #1 presented metastases at diagnosis, while case #2 was at a lower stage. LGEC component was invasive in case #1 and intramucous in case #2. In both cases, UC components were characterized by a high degree of instability, in accordance of its aggressive behaviour and its architectural heterogeneity. Further studies with more numerous cases are mandatory to confirm these data. C1 [Giordano, Giovanna] University of Parma, Institute of Pathology, Viale A. Gramsci, 14, 43100 Parma, Italy. [D’Adda, Tiziana] University of Parma, Institute of Pathology, Viale A. Gramsci, 14, 43100 Parma, Italy. [Bottarelli, Lorena] University of Parma, Institute of Pathology, Viale A. Gramsci, 14, 43100 Parma, Italy. [Lombardi, Mariano] University of Parma, Institute of Pathology, Viale A. Gramsci, 14, 43100 Parma, Italy. [Brigati, Francesca] University of Parma, Institute of Pathology, Viale A. Gramsci, 14, 43100 Parma, Italy. [Berretta, Roberto] University of Parma, Ob/Gyn ClinicParma, Italy. [Merisio, Carla] University of Parma, Ob/Gyn ClinicParma, Italy. RP Giordano, G (reprint author), University of Parma, Institute of Pathology, 43100 Parma, Italy. EM giovanna.giordano@unipr.it CR Silverberg SG, Kurman RJ, Nogales F et al, 2003, Epithelial tumors and related lesions. In: Tavassoli FA, Devilee P, eds, Tumors of the breast and female genital organs: World Health Organization classification of tumours. IARC, Lyon, p 227 Altraulsi B, Malpica A, Deavers MT, Bodurka DC, Broddus R, Silva EG, 2005, Undifferentiated carcinoma of the endometrium. Am J Surg Pathol 29:1316–1321 Silva EG, Deavers MT, Malpica A, 2007, Undifferentiated carcinoma of the endometrium: a review. Pathology 39:134–138 Silva EG, Deavers MT, Bodurka DC et al, 2006, Association of Low-Grade Endometrioid carcinoma of the uterus and ovary with carcinoms: a new type of dedifferentiated carcinoma? Int J Gynecol Pathol 25:52–58 Boland CR, Thibodeau SN, Hamilton SR et al, 1998, A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial pre disposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58:5248–5257 Umar A, Boland CR, Terdiman JP et al, 2004, Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer, Lynch syndrome, and microsatellite instability. J Natl Cancer Inst 96:261–268 D'Adda T, Bottarelli L, Azzoni C et al, 2005, Malignancy-associated X chromosome allelic losses in foregut endocrine neoplasms: further evidence from lung tumors. Mod Pathol 18:795–805 Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics. CA Cancer J Clin 59:225–249 Bokhman JV, 1983, Two pathogenetic types of endometrial carcinoma. Gynecol Oncol 15:10–17 Slomovitz BM, Burke TW, Eifel PJ et al, 2003, Uterine papillary serous carcinoma, UPSC): a single institution review of 129 cases. Gynecol Oncol 91:463–469 Sherman ME, Bur ME, Kurman RJ, 1995, p53 in endometrial cancer and its putative precursors: evidence for diverse pathways of tumorigenesis. Hum Pathol 26:1268–1274 Tashiro H, Blazes MS, Wu R et al, 1997, Mutations in PTEN are frequent in endometrial carcinoma but rare in other common gynecological malignancies. Cancer Res 57:3935–3940 Risinger JI, Hayes AK, Berchuck A, Barrett JC, 1997, PTEN/ MMAC1mutations in endometrial cancers. Cancer Res 57:4736–4738 Duggan BD, Felix JC, Muderspach LI, Tourgeman D, Zheng J, Shibata D, 1994, Microsatellite instability in sporadic endometrial carcinoma. J Natl Cancer Inst 86:1216–1221 Mutter GL,Wada H, FaquinWC, Enomoto T, 1999, K-ras mutations appear in the premalignant phase of both microsatellite stable and unstable endometrial carcinogenesis. Mol Pathol 52:257–262 Lagarda H, Catasus L, Arguelles R, Matias-Guiu X, Prat J, 2001, K-ras mutations in endometrial carcinomas with microsatellite instability. J Pathol 193:193–199 Fukuchi T, Sakamoto M, Tsuda H, Maruyama K, Nozawa S, Hirohashi S, 1998, Beta-catenin mutation in carcinoma of the uterine endometrium. Cancer Res 58:3526–3528 Mirabelli-Primdahl L, Gryfe R, Kim H et al, 1999, Beta-catenin mutations are specific for colorectal carcinomas with microsatellite instability but occur in endometrial carcinomas irrespective of mutator pathway. Cancer Res 59:3346–3351 Risinger JI, Berchuck A, Kohler MF, Watson P, Lynch HT, Boyd J, 1993, Genetic instability of microsatellites in endometrial carcinoma. Cancer Res 53:5100–5103 Mutter GL, Boynton KA, Faquin WC, Ruiz RE, Jovanovic AS, 1996, Allelotype mapping of unstable microsatellites establishes direct lineage continuity between endometrial precancers and cancer. Cancer Res 56:4483–4486 Weissenbach J, Gyapay G, Dib C et al, 1992, A secondgeneration linkage map of the human genome. Nature 359:794– 801 Peltomaki P, Lothe RA, Aaltonen LA et al, 1993, Microsatellite instability is associated with tumors that characterize the hereditary non-polyposis colorectal carcinoma syndrome. Cancer Res 53:5853–5855 Ionov Y, Peinado MA, Malkhosyan S et al, 1993, Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature 363:558–561 Wirtz HC, Muller W, Noguchi T et al, 1998, Prognostic value and clinicopathological profile of microsatellite instability in gastric cancer. Clin Cancer Res 4:1749–1754 Duggan BD, Felix JC, Muderspach LI et al, 1994, Microsatellite instability in sporadic endometrial carcinoma. J Natl Cancer Inst 86:1216–1221 Wagner BJ, Presnell SC, 2009, Loss of heterozygosity in basic concepts of molecular pathology. Springer US 2:97–107 Black D, Soslow RA, Levine DA et al, 2006, Clinicopathologic significance of defective DNA mismatch repair in endometrial carcinoma. J Clin Oncol 24(11):1745–1753 Shia J, Black D, Hummer AJ et al, 2008, Routinely assessed morphological features correlate with microsatellite instability status in endometrial cancer. Hum Pathol 39(1):116–125 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 523 EP 528 DI 10.1007/s12253-011-9386-7 PG 6 ER PT J AU Dundr, P Fischerova, D Povysil, C Tvrdik, D Cibula, D AF Dundr, Pavel Fischerova, Daniela Povysil, Ctibor Tvrdik, Daniel Cibula, David TI Primary Synovial Sarcoma of the Uterus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Female genital tract; Immunohistochemistry; RT-PCR; Synovial sarcoma; Uterus ID Female genital tract; Immunohistochemistry; RT-PCR; Synovial sarcoma; Uterus AB We report a case of a 52-year-old female with synovial sarcoma of the uterine corpus. Grossly, the partly polypoid tumor involved the endometrium with invasion into the inner half of the myometrium. Histologically, the tumor showed biphasic structure with the predominance of poorly differentiated small to medium sized round to oval cells. These cells showed high nuclear to cytoplasmic ratio and were arranged in diffuse sheets. Other component consisted of larger epitheloid cells with ample eosinophilic cytoplasm arranged in irregular nests. These cells were only present in a small amount. Immunohistochemically, the tumor cells in both components showed the expression of EMA, S-100 protein, CD99, and NSE. RT-PCR analysis showed the presence of SYT-SSX1 fusion transcript. At present, the patient shows no signs of tumor relapse 56 months after the diagnosis. To the best of our knowledge, this is the first report of synovial sarcoma arising in uterus. C1 [Dundr, Pavel] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Fischerova, Daniela] Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Obstetrics and GynecologyPrague, Czech Republic. [Povysil, Ctibor] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Tvrdik, Daniel] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Cibula, David] Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Obstetrics and GynecologyPrague, Czech Republic. RP Dundr, P (reprint author), Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, 12800 Prague, Czech Republic. EM pdundr@seznam.cz CR Weiss SW, Goldblum J, 2008, Malignant soft tissue tumors of uncertain type. In: Weiss SW, Goldblum JR, eds, Enzinger and Weiss’s soft tissue tumors, 5th edition). Mosby Elsevier, St. Louis, pp 1161–220 Al-DarajiW, Lasota J, Foss R, MiettinenM(2009, Synovial sarcoma involving the head: analysis of 36 cases with predilection to the parotid and temporal regions. Am J Surg Pathol 33:1494–1503 Tvrdik D, Svatosova J, Dundr P, Povysil C, 2005, Molecular Diagnosis of Synovial Sarcoma: Detection of SYT-SSX1/2 Fusion Transcripts by RT-PCR in Paraffin-Embedded Tissue. Med Sci Monit 11:MT1–7 Drozenova J, Povysil C, Tvrdik D, Babjuk M, Hanus T, 2008, Primary synovial sarcoma of the kidney. Cesk Patol 44:20–22 Makhlouf HR, Ahrens W, Agarwal B et al, 2008, Synovial sarcoma of the stomach: a clinicopathologic, immunohistochemical, and molecular genetic study of 10 cases. Am J Surg Pathol 32:275–281 Roberts CA, Seemayer TA, Neff JR, Alonso A, Nelson M, Bridge JA, 1996, Translocation, X;18, in primary synovial sarcoma of the lung. Cancer Genet Cytogenet 88:49–52 Wang ZH,Wang XC, Xue M, 2010, Clinicopathologic analysis of 4 cases of primary renal synovial sarcoma. Chin J Cancer 29:212–216 Ambani DS, White B, Kaplan AL, Alberto A, 2006, A case of monophasic synovial sarcoma presenting as a vulvar mass. Gynecol Oncol 100:433–436 Holloway CL, Russell AH, Muto M, Albert M, Viswanathan AN, 2007, Synovial cell sarcoma of the vulva: multimodality treatment incorporating preoperative external-beam radiation, hemivulvectomy, flap reconstruction, interstitial brachytherapy, and chemotherapy. Gynecol Oncol 104:253–256 Mitsuhashi A, Nagai Y, Suzuka K et al, 2007, Primary synovial sarcoma in fallopian tube: case report and literature review. Int J Gynecol Pathol 26:34–37 Nielsen GP, Shaw PA, Rosenberg AE, Dickersin GR, Young RH, Scully RE, 1996, Synovial sarcoma of the vulva: a report of two cases. Mod Pathol 9:970–974 Pelosi G, Luzzatto F, Landoni F et al, 2007, Poorly differentiated synovial sarcoma of the vagina: first reported case with immunohistochemical, molecular and ultrastructural data. Histopathology 50:808–810 Smith CJ, Ferrier AJ, Russell P, Danieletto S, 2005, Primary synovial sarcoma of the ovary: first reported case. Pathology 37:385–387 Naka N, Takenaka S, Araki N et al, 2010, Synovial sarcoma is a stem cell malignancy. Stem Cells 28:1119–1131 Povysil C, 1984, Synovial sarcoma with squamous metaplasia. Ultrastruct Pathol 7:207–213 Kosemehmetoglu K, Vrana JA, Folpe AL, 2009, TLE1 expression is not specific for synovial sarcoma: a whole study of 163 soft tissue and bone neoplasms. Mod Pathol 22:872–878 Krskova L, Sumerauer D, Stejskalova E, Kodet R, 2007, A novel variant of SYT-SSX1 fusion gene in a case of spindle cell synovial sarcoma. Diagn Mol Pathol 16:179–183 Turc-Carel C, Dal Cin P, Limon J et al, 1987, Involvement of chromosome X in primary cytogenetic change in human neoplasia: nonrandom translocation in synovial sarcoma. Proc Natl Acad Sci USA 84:1981–1985 Guillou L, Benhattar J, Bonichon F et al, 2004, Histologic grade, but not SYT-SSX fusion type, is an important prognostic factor in patients with synovial sarcoma: a multicenter, retrospective analysis. J Clin Oncol 22:4040–4050 ten Heuvel SE, Hoekstra HJ, Bastiaannet E, Suurmeijer AJ, 2009, The classic prognostic factors tumor stage, tumor size, and tumor grade are the strongest predictors of outcome in synovial sarcoma: no role for SSX fusion type or ezrin expression. Appl Immunohistochem Mol Morphol 17:189–195 Michal M, Fanburg-Smith JC, Lasota J, Fetsch JF, Lichy J, Miettinen M, 2006, Minute synovial sarcomas of the hands and feet: a clinicopathologic study of 21 tumors less than 1 cm. Am J Surg Pathol 30:721–726 Singer S, Baldini EH, Demetri GD, Fletcher JA, Corson JM, 1996, Synovial sarcoma: prognostic significance of tumor size, margin of resection, andmitotic activity for survival. J Clin Oncol 14:1201–1208 Spillane AJ, A’Hern R, Judson IR, Fisher C, Thomas JM, 2000, Synovial sarcoma: a clinicopathologic, staging, and prognostic assessment. J Clin Oncol 18:3794–3803 Palmerini E, Staals EL, Alberghini M et al, 2009, Synovial sarcoma: retrospective analysis of 250 patients treated at a single institution. Cancer 115:2988–2998 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 529 EP 533 DI 10.1007/s12253-011-9391-x PG 5 ER PT J AU Toth, B Katona, M Harsing, J Szepesi, Karpati, S AF Toth, Bela Katona, Maria Harsing, Judit Szepesi, Agota Karpati, Sarolta TI Indeterminate Cell Histiocytosis in a Pediatric Patient: Successful Treatment with Thalidomide SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Indeterminate cell histiocytosis; Thalidomide; Pediatric patient; Immunohistochemistry; Electronmicroscopy; Birbeck granules ID Indeterminate cell histiocytosis; Thalidomide; Pediatric patient; Immunohistochemistry; Electronmicroscopy; Birbeck granules AB The 15-year-old male patient presented several 2–6 mm large livid reddish-yellowish, shiny, compact papules on the head, trunk and extremities, which had developed within the last 4 months. Histology showed normal epidermis with dense dermal infiltrate of histiocytes accompanied by few eosinophils, Touton or foamy giant cells. The histiocytes were S100 positive, CD1a negative and did not contain Birbeck granules ultrastructurally. Chest X ray, EEG, skull MRI did not show pathology. Opthalmology, neurology, oto-rhino-laryngology did not reveal alterations. Based upon the clinical symptoms and the histopathology, the diagnosis of indeterminate cell histiocytosis was confirmed. Cryotherapy and cauterization did not stop the progression of the disease, however, under thalidomide treatment no new symptoms developed and the lesions healed with pigmentation. C1 [Toth, Bela] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria utca 41, 1085 Budapest, Hungary. [Katona, Maria] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria utca 41, 1085 Budapest, Hungary. [Harsing, Judit] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria utca 41, 1085 Budapest, Hungary. [Szepesi, Agota] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Karpati, Sarolta] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria utca 41, 1085 Budapest, Hungary. RP Toth, B (reprint author), Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, 1085 Budapest, Hungary. EM tothbela@bor.sote.hu CR Gianotti F, Caputo R, 1985, Histiocytic syndromes: a review. J Am Acad Dermatol 13:383–404 Bolognia JL, Jorizzo JL, Rapini RP, eds,, 2008, Dermatology. Mosby Elsevier, London Edinburgh New York Philadelphia St Louis Sidney Toronto Ringel E, Moschella S, 1985, Primary histiocytic dermatoses. Arch Dermatol 121:1531–1542 Groopman J, Golde D, 1981, The histiocytic disorders: a pathophysiologic analysis. Ann Intern Med 94:95–107 Coldiron BM, Santa-Cruz D, Freeman RG et al, 1988, Benign non-X histiocytosis: a unique case bridging several of the non-X histiocytic syndromes. J Am Acad Dermatol 18:1282–1289 Manente L, Cotellessa C, Schmitt I et al, 1997, Indeterminate cell histiocytosis: a rare histiocytic disorder. Am J Dermatopathol 19:276–283 Rodriguez-Jurado R, Vidaurri-de la Cruz H, Duran-Mckinster C et al, 2003, Indeterminate cell histiocytosis. Clinical and pathologic study in a pediatric patient. Arch Pathol Lab Med 127(6):748–751 Burgdorf WHC, Plewig G, Wolff HH, Landthaler M, eds,, 2009, Braun-Falco’s dermatology. Springer, Heidelberg Breatnach AS, 1963, A new concept of the relation between the Langerhans cell and the melanocyte. J Invest Dermatol 40:279– 281 Wood GS, Hu CH, Beckstead JH et al, 1985, The indeterminate cell proliferative disorder: report of a case manifesting as an unusual cuteaneous histiocytosis. J Dermatol Surg Oncol 11:1111 Ratzinger G, Burgdorf WH, Metze D et al, 2005, Indeterminate cell histiocytosis: fact or fiction? J Cutan Pathol 32(8):552–560 Kolde G, Brocker EB, 1986, Multiple skin tumors of indeterminate cells in an adult. J Am Acad Dermatol 15:591 Berti E, Gianotti R, Alessi E, 1988, Unusual cutaneous histiocytosis expressing an intermediate immunophenotype between Langerhans cells and dermal macrophages. Arch Dermatol 124:1250–1253 Sidoroff A, Zelger B, Steiner H et al, 1996, Indeterminate cell histiocytosis—a clinicopathological entity with features of both X- and non-X histiocytosis. Br J Dermatol 134:525 Calatayud M, Guell JL, Gris O et al, 2001, Ocular involvement in a case of systemic indeterminate cell hitiocytosis: case report. Cornea 20:769 Zelger B, Burgdorf WHC, 2001, The cutaneous histiocytoses. Adv Dermatol 17:77 Segal GH, Mesa MV, Fishleder AJ et al, 1992, Precursor Langerhans cell histiocytosis. An unusual histiocytic proliferation in a patient with persistent non-Hodgkin lymphoma and terminal acute monocytic leukemia. Cancer 70:547–553 Vener C, Soligo D, Berti E et al, 2007, Indeterminate cell histiocytosis in association with later occurrence of acute myeloblastic leukaemia. Br J Dermatol 156(6):1357–1361 Broekaert SM, Metzler G, Burgdorf W et al, 2007, Multisystem Langerhans cell histiocytosis: successful treatment with thalidomide. Am J Clin Dermatol 8(5):311–314 Wang CH, Chen GS, 2004, Indeterminate cell histiocytosis: a case report. J Med Sci 20:24–30 Malhomme de la Roche H, Lai-Cheong JE, Calonje E et al, 2008, Indeterminate cell histiocytosis responding to total skin electron beam therapy. Br J Dermatol 158(4):838–840 Eichholz A, Merchant S, Gaya AM, 2010, Anti-angiogenesis therapies: their potential in cancer management. Onco Targets Ther 24(3):69–82 Ventura F, Pereira T, da Luz Duarte M et al, 2010, Indeterminate cell histiocytosis in association with acute myeloid leukemia. Dermatol Res Pract 2010:569345. Epub 2010 Jun 21 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 535 EP 538 DI 10.1007/s12253-011-9405-8 PG 4 ER PT J AU Mahta, A Qu, Y Nastic, D Sundstrom, M Kim, YR Saria, M Santagata, S Kesari, S AF Mahta, Ali Qu, Yan Nastic, Denis Sundstrom, Maria Kim, Y Ryan Saria, Marlon Santagata, Sandro Kesari, Santosh TI Relapsing Tumefactive Lesion in an Adult with Medulloblastoma Previously Treated with Chemoradiotherapy and Stem Cell Transplant SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Medulloblastoma; Tumefactive; Stem cell transplant; Radiation necrosis ID Medulloblastoma; Tumefactive; Stem cell transplant; Radiation necrosis AB Herein, we present an adult case of medulloblastoma who received chemotherapy, radiation therapy and stem cell transplantation, and underwent multiple surgical resections for what were thought to be recurrences; however pathology confirmed a diagnosis of relapsing tumefactive lesions. This phenomenon seems to be a consequence of stem cell transplantation rather than a simple radiation treatment effect. C1 [Mahta, Ali] University of California, Moores Cancer Center, Department of Neurosciences, 3855 Health Sciences Drive, Suite 3336, La Jolla, 92093-0819 San Diego, CA, USA. [Qu, Yan] University of California, Moores Cancer Center, Department of Neurosciences, 3855 Health Sciences Drive, Suite 3336, La Jolla, 92093-0819 San Diego, CA, USA. [Nastic, Denis] Umea University Medical SchoolUmea, Sweden. [Sundstrom, Maria] Umea University Medical SchoolUmea, Sweden. [Kim, Y Ryan] University of California, Moores Cancer Center, Department of Neurosciences, 3855 Health Sciences Drive, Suite 3336, La Jolla, 92093-0819 San Diego, CA, USA. [Saria, Marlon] University of California, Moores Cancer Center, Department of Neurosciences, 3855 Health Sciences Drive, Suite 3336, La Jolla, 92093-0819 San Diego, CA, USA. [Santagata, Sandro] Brigham and Women’s Hospital, Department of Pathology, Division of Neuropathology, 75 Francis Street, 02115 Boston, MA, USA. [Kesari, Santosh] University of California, Moores Cancer Center, Department of Neurosciences, 3855 Health Sciences Drive, Suite 3336, La Jolla, 92093-0819 San Diego, CA, USA. RP Kesari, S (reprint author), University of California, Moores Cancer Center, Department of Neurosciences, 92093-0819 San Diego, USA. EM skesari@ucsd.edu CR Schultheiss TE, Kun LE, Ang KK, Stephens LC, 1995, Radiation response of the central nervous system. Int J Radiat Oncol Biol Phys 31(5):1093–1112 Armstrong CL, Hunter JV, Ledakis GE et al, 2002, Late cognitive and radiographic changes related to radiotherapy: initial prospective findings. Neurology 59(1):40–48 Ricci PE, Karis JP, Heiserman JE, Fram EK, Bice AN, Drayer BP, 1998, Differentiating recurrent tumor from radiation necrosis: time for re-evaluation of positron emission tomography? AJNR Am J Neuroradiol 19(3):407–413 Sorensen J, Savitcheva II, Engler H, Langstrom B, 2000, 3. Utility of PET and 11 C-Methionine in the Paediatric Brain Tumors. Clin Positron Imaging 3(4):157 Chao ST, Suh JH, Raja S, Lee SY, Barnett G, 2001, The sensitivity and specificity of FDG PET in distinguishing recurrent brain tumor from radionecrosis in patients treated with stereotactic radiosurgery. Int J Cancer 96(3):191–197 Giannopoulou C, Fragaki C, 2006, Positron emission tomography in evaluating the response to treatment of brain tumors, lymphomas and breast cancer. Hell J Nucl Med 9(2):117–125 Fouladi M, Chintagumpala M, Laningham FH et al, 2004, White matter lesions detected by magnetic resonance imaging after radiotherapy and high-dose chemotherapy in children with medulloblastoma or primitive neuroectodermal tumor. J Clin Oncol 22(22):4551–4560 Asato R, Akiyama Y, Ito M et al, 1992, Nuclear magnetic resonance abnormalities of the cerebral white matter in children with acute lymphoblastic leukemia and malignant lymphoma during and after central nervous system prophylactic treatment with intrathecal methotrexate. Cancer 70(7):1997–2004 Paakko E, Vainionpaa L, Lanning M, Laitinen J, Pyhtinen J, 1992, White matter changes in children treated for acute lymphoblastic leukemia. Cancer 70(11):2728–2733 Hertzberg H, Huk WJ, Ueberall MA et al, 1997, CNS late effects after ALL therapy in childhood. Part I: Neuroradiological findings in long-term survivors of childhood ALL–an evaluation of the interferences between morphology and neuropsychological performance. The German Late Effects Working Group. Med Pediatr Oncol 28(6):387–400 Khong PL, Kwong DL, Chan GC et al, 2003, Diffusion-tensor imaging for the detection and quantification of treatment-induced white matter injury in children with medulloblastoma: a pilot study. AJNR Am J Neuroradiol 24(4):734–740 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 539 EP 543 DI 10.1007/s12253-011-9464-x PG 5 ER PT J AU Tornoczky, T Bogner, B Krausz, Th Ottoffy, G Szuhai, K AF Tornoczky, Tamas Bogner, Barna Krausz, Thomas Ottoffy, Gabor Szuhai, Karoly TI Angiomatoid Fibrous Histiocytoma: Pleomorphic Variant Associated with Multiplication of EWSR1-CREB1 Fusion Gene SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE AFH; Angiomatoid fibrous histiocytoma; Pleomorphic; Multiplication; EWSR1-CREB1 fusion gene ID AFH; Angiomatoid fibrous histiocytoma; Pleomorphic; Multiplication; EWSR1-CREB1 fusion gene AB Angiomatoid fibrous histiocytoma (AFH) is a rare soft tissue tumor which exceptionally occurs in visceral organs or bones. Histologically this is a bland, monomorphic tumor and only occasionally shows pleomorphism. Vast majority of the soft tissue cases share the same translocation and the resulting EWSR1-CREB1 gene fusion as background pathogenetic alteration. Here we report a 10-year-old boy with subcutaneous tumor of the right shoulder. Histological, immunohistochemical and FISH analyses of the case revealed pleomorphic phenotype, characteristic immunophenotype and multiplication of the EWSR1-CREB1 fusion gene in the nuclei of the tumor cells. The possible explanation of the fusion gene multiplication, its relation to the morphology and the clinical outcome are discussed in the context of the published literature. C1 [Tornoczky, Tamas] University of Pecs, Department of Pathology, Rakoczi ut 2, 7623 Pecs, Hungary. [Bogner, Barna] Balassa Janos County Hospital, Department of PathologySzekszard, Hungary. [Krausz, Thomas] University of Illinois at Chicago, College of Medicine, Department of PathologyChicago, IL, USA. [Ottoffy, Gabor] University of Pecs, Department of PediatricsPecs, Hungary. [Szuhai, Karoly] Leiden University, Medical Center, Department of Molecular Cell BiologyLeiden, The Netherlands. RP Tornoczky, T (reprint author), University of Pecs, Department of Pathology, 7623 Pecs, Hungary. EM ttamas64@hotmail.com CR Enzinger FM, 1979, Angiomatoid malignant fibrous histiocytoma: a distinct fibrohistiocytic tumor of children and young adults simulating a vascular neoplasm. Cancer 44(6):2147–2157 Fletcher CDM, 1991, Angiomatoid “malignant fibrous histiocytoma”: an immunohistochemical study indicative of myoid differentiation. Hum Pathol 22(6):563–568 Fanburg-Smith JC, Miettinen M, 1999, Angiomatoid “malignant” fibrous histiocytoma: a clinicopathologic study of 158 cases and further exploration of the myoid phenotype. Hum Pathol 30, 11):1336–1343 Tanas MR, Rubin BP, Montgomery EA et al, 2010, Utility of FISH in the diagnosis of angiomatoid fibrous histiocytoma: a series of 18 cases. Mod Pathol 23:93–97 Waters BL, Panagopoulos I, Allen EF, 2000, Genetic characterization of angiomatoid fibrous histiocytoma identifies fusion of the FUS and ATF genes induced by a chromosomal translocation involving bands 12q13 and 16p11. Cancer Genet Cytogenet 121:109–116 Hallor KH, Micci F, Meis-Kindblom JM et al, 2007, Fusion genes in angiomatoid fibrous histiocytoma. Cancer Lett 251, 1):158–163 Antonescu CR, Dal Cin P, Nafa K et al, 2007, EWSR1-CREB1 is the predominant gene fusion in angiomatoid fibrous histiocytoma. Genes Chromosomes Cancer 46(12):1051–1060 Rossi S, Szuhai K, Ijszenga M et al, 2007, EWSR1-CREB1 and EWSR1-ATF1 fusion genes in angiomatoid fibrous histiocytoma. Clin Cancer Res 13(24):7322–7328 Costa MJ, Weiss SW, 1990, Angiomatoid malignant fibrous histiocytoma. A follow-up study of 108 cases with evaluation of possible histologic predictors of outcome. Am J Surg Pathol 14, 12):1126–1132 Weinreb I, Rubin BP, Goldblum JR, 2008, Pleomorphic angiomatoid fibrous histiocytoma: a case confirmed by fluorescence in situ hybridization analysis for EWSR1 rearrangement. J Cutan Pathol 35, 9):855–860 Matsumura T, Yamaguchi T, Tochigi N et al, 2010, Angiomatoid fibrous histiocytoma including cases with pleomorphic features analysed by fluorescence in situ hybridization. J Clin Pathol 63, 2):124–128 Moura RD, Wang X, Lonzo ML et al, 2011, Reticular angiomatoid “malignant” fibrous histiocytoma-a case report with cytogenetics and molecular genetic analyses. Hum Pathol 42, 9):1359–1363 Mangham DC, Williams A, Lalam RK et al, 2010, Angiomatoid fibrous histiocytoma of bone: a calcifying sclerosing variant mimicking osteosarcoma. Am J Surg Pathol 34:279–285 Ren L, Guo SP, Zhou XG et al, 2009, Angiomatoid fibrous histiocytoma. First report of primary pulmonary origin. Am J Surg Pathol 33:1570–1574 Dunham C, Hussong J, Seiff M et al, 2008, Primary intracerebral angiomatoid fibrous histiocytoma. Report of a case with a t(12;22,, q13;q12, causing type 1 fusion of the EWS and ATF-1 genes. Am J Surg Pathol 32:478–484 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2012 VL 18 IS 2 BP 545 EP 548 DI 10.1007/s12253-011-9468-6 PG 4 ER PT J AU Fan, Ch Yu, J Liu, Y Xu, H Wang, E AF Fan, Chuifeng Yu, Juanhan Liu, Yang Xu, Hongtao Wang, Enhua TI Increased NDRG1 Expression is Associated with Advanced T Stages and Poor Vascularization in Non-small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE IHC; MVD; NDRG1; NSCLC; T stage ID IHC; MVD; NDRG1; NSCLC; T stage AB N-myc downstream regulated gene 1 (NDRG1) is a member of the N-myc downstream regulated gene family which belongs to the alpha/beta hydrolase superfamily. Earlier studies have shown its association with inhibition of tumor metastasis. However, its function in malignant tumors is not fully enunciated. Recently there was increasing evidence that NDRG1 is involved in stress responses. In the current study, we examined the expression of NDRG1 and its correlation with clinicopathological factors and microvessel density (MVD) in non-small cell lung cancer (NSCLC) using immunohistochemistry (IHC). NDRG1 expression in NSCLC (71/115, 61.7%) was higher than that in normal lung tissues (32/115, 27.8%) (p<0.05). NDRG1 expression in NSCLC cells was found in cytoplasm (63/115, 54.8%), nuclear (24/115, 20.9%) and cell membrane (13/115, 11.3%). NDRG1 expression in NSCLC with advanced T stages (T2-4) (63/84, 75.0%) was significantly higher than that with T1 stage (8/31, 25.8%) (P<0.05). No other clinicopathological factors including lymph node metastasis were found to be associated with NDRG1 expression (p>0.05). Moreover increased NDRG1 expression was associated with lower MVD in NSCLC (P<0.05). MVD in adenocarcinoma (33.4±8.4/HP) was significantly higher than that in squamous cell carcinoma (SCC) (19.3±8.1/HP) (P<0.05). No other clinicopathological factors were associated with MVD in NSCLC (p>0.05). The present findings indicate an increase of NDRG1 expression with the progress of tumour extent which may be due to unbalanced tumor oxygenation on account of poor vascularization in NSCLC. C1 [Fan, Chuifeng] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110001 Shenyang, China. [Yu, Juanhan] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110001 Shenyang, China. [Liu, Yang] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110001 Shenyang, China. [Xu, Hongtao] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110001 Shenyang, China. [Wang, Enhua] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110001 Shenyang, China. RP Fan, Ch (reprint author), China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110001 Shenyang, China. EM fanchuifeng@yeah.net CR Lachat P, Shaw P, Gebhard S et al, 2002, Expression of NDRG1, a differentiation-related gene, in human tissues. Histochem Cell Biol 118:399–408 Taketomi Y, Sugiki T, Saito T et al, 2003, Identification of NDRG1 as an early inducible gene during in vitro maturation of cultured mast cells. Biochem Biophys Res Commun 306:339–346 Wakisaka Y, Furuta A, Masuda K et al, 2003, Cellular distribution of NDRG1 protein in the rat kidney and brain during normal postnatal development. J Histochem Cytochem 51:1515–1525 Rutherford MN, Bayly GR, Matthews BP et al, 2001, The leukemogenic transcription factor E2a-Pbx1 induces expression of the putative N-myc and p53 target gene NDRG1 in Ba/F3 cells. Leukemia 15:362–370 Angst E, Sibold S, Tiffon C et al, 2006, Cellular differentiation determines the expression of the hypoxia-inducible protein NDRG1 in pancreatic cancer. Br J Cancer 95:307–313 Echaniz-Laguna A, Degos B, Bonnet C et al, 2007, NDRG1- linked Charcot-Marie-Tooth disease, CMT4D, with central nervous system involvement. Neuromuscul Disord 17:163–168 Zhou D, Salnikow K, Costa M, 1998, Cap43, a novel gene specially induced by Ni2+ compounds. Cancer Res 58:2182–2189 Salnikow K, Blagosklonny MV, Ryan H et al, 2000, Carcinogenic nickel induces genes involved with hypoxic stress. Cancer Res 60:38–41 Hakan C, 2004, Hypoxia upregulates the expression of the NDRG1 gene leading to its overexpression in various human cancers. BMC Genet 5:27 Joseph TC, Hung-Ming W, Ku-Wei C et al, 2005, Identification of differentially expressed genes in oral squamous cell carcinoma, OSCC): Overexpression of NPM, CDK1 and NDRG1 and underexpression of CHES1. Int J Cancer 114:942–949 Sonja S, Vincent R, Adrian K et al, 2007, Hypoxia increases cytoplasmic expression of NDRG1, but is insufficient for its membrane localization in human hepatocellular carcinoma. FEBS Lett 581:989–994 Mei-Sze C, Hongbo S, Siu TC et al, 2007, Overexpression of NDRG1 is an indicator of poor prognosis in hepatocellular carcinoma. Mod Pathol 20:76–83 Sobin DHWC, 2002, International Union Against Cancer, UICC): TNM Classification of Malignant Tumors, 6. Wiley-Liss, New York Travis WDBE, Muller-Hermelink HK, Harris CC, 2004, World health organization classification of tumors: pathology and genetics of tumors of the lung, pleura, thymus and heart. IARC, Lyon Weidner N, Semple JP, Welch WR et al, 1991, Tumor angiogenesis and metastasis-correlation in invasive breast carcinoma. N Engl J Med 324:1–8 Bandyopadhyay S, Pai SK, Gross SC et al, 2003, The Drg-1 gene suppresses tumor metastasis in prostate cancer. Cancer Res 63:1731–1736 Bandyopadhyay S, Pai SK, Hirota S et al, 2004, Role of the putative tumor metastasis suppressor gene Drg-1 in breast cancer progression. Oncogene 23:5675–5681 Ulrix W, Swinnen JV, Heyns W et al, 1999, The differentiationrelated gene 1, Drg1, is markedly upregulated by androgens in LNCaP prostatic adenocarcinoma cells. FEBS Lett 455:23–26 Kokame K, Kato H, Miyata T, 1996, Homocysteine-respondent genes in vascular endothelial cells identified by differential display analysis. GRP78/BiP and novel genes. J Biol Chem 271:29659–29665 Shin N, Kimio U, Naotake T et al, 2008, Cap43/NDRG1/Drg-1 is a molecular target for angiogenesis and a prognostic indicator in cervical adenocarcinoma. Cancer Letters 264:36–43 Maruyama Y, Ono M, Kawahara A et al, 2006, Tumor growth suppression in pancreatic cancer by a putative metastasis suppressor gene Cap43/NDRG1/Drg-1 through modulation of angiogenesis. Cancer Res 66:6233–6242 Guan RJ, Ford HL, Fu Y et al, 2000, Drg-1 as a differentiationrelated, putative metastatic suppressor gene in human colon cancer. Cancer Res 60:749–755 Macchiarini P, Fontanini G, Hardin MJ et al, 1992, Relation of neovascularisation to metastasis of non-small cell lung cancer. Lancet 340:145–146 Fontanini G, Bigini D, Vignati S et al, 1995, Microvessel count predicts metastatic disease and survival in non-small cell lung cancer. J Pathol 177:57–63 Maeda K, Chung YS, Takatsuka S et al, 1995, Tumor angiogenesis as a predictor of recurrence in gastric carcinoma. J Clin Oncol 13:477–481 Yamazaki K, Abe S, Takekawa H et al, 1994, Tumor angiogenesis in human lung adenocarcinoma. Cancer 74:2245–2250 Anne E, Sebastian K, Valentin G et al, 2000, Heterogeneity of angiogenesis and blood vessel maturation in human tumors: implications for antiangiogenic tumor therapies. Cancer Res 60:1388–1393 Fumihiro T, Hiroki O, Kazumasa T et al, 2003, Glomeruloid microvascular proliferation is superior to intratumoral microvessel density as a prognostic marker in non-small cell lung cancer. Cancer Res 63:6791–6794 Guo J, Higashi K, Ueda Y et al, 2006, Microvessel density: correlation with 18F-FDG uptake and prognostic impact in lung adenocarcinomas. J Nucl Med 47:419–425 Trivella M, Pezzella F, Pastorino U et al, 2007, Microvessel density as a prognostic factor in non-small-cell lung carcinoma: a meta-analysis of individual patient data. Lancet Oncol 8:488–499 Sardari NP, Stessels F, Pezzella F et al, 2003, Growth index is independent of microvessel density in non-small cell lung carcinomas. Hum Pathol 34:959–960 Zheng Y, Wang LS, Xia L et al, 2009, NDRG1 is down-regulated in the early apoptotic event induced by camptothecin analogs: the potential role in proteolytic activation of PKC delta and apoptosis. Proteomics 9:2064–2075 Kurdistani SK, Arizti P, Reimer CL et al, 1998, Inhibition of tumor cell growth by RTP/rit42 and its responsiveness to p53 and DNA damage. Cancer Res 58:4439–4444 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 549 EP 556 DI 10.1007/s12253-010-9294-2 PG 8 ER PT J AU Kadar, K Wolf, K Tabori, J Karadi, I Varkonyi, J AF Kadar, Katalin Wolf, Krisztina Tabori, Judit Karadi, Istvan Varkonyi, Judit TI The Albumin and Monoclonal Protein Ratio as Prognostic Marker for Multiple Myeloma in the Era of Novel Agents SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Multiple myeloma; Prognostic factors; Albumin; M-protein; A/M ratio ID Multiple myeloma; Prognostic factors; Albumin; M-protein; A/M ratio AB Multiple myeloma (MM) is a heterogeneous disease group regarding prognosis, clinical course, and response to therapeutic interventions. Numerous prognostic factors have been identified however there was no consensus about the best prognostic indicators or the proper staging systems. In a previous study the A/M ratio containing albumin (A) and monoclonal component (M) emerged as reliable predictor of survival duration in patients treated with conventional chemotherapy. In the current retrospective study authors evaluated the prognostic role of this fraction in the era of novel agents. They assessed the A/M ratio prior treatment in 56 newly diagnosed MM patients from the aspect of the survival time. According to the results the A/M being <1 at the diagnosis indicated significantly poorer prognosis both at the 2 years (p00,01) and at the 5 years (p00,07) survival endpoints. These results proved that A/M ratio remained valuable marker for predicting prognosis in patients treated with proteosome inhibitor and antiangiogenic therapy as well. Authors recommend therefore applying this A/M ratio in further studies for the better pre-treatment stratification. C1 [Kadar, Katalin] Semmelweis University, Kutvolgyi Clinical Centre, Kutvolgyi ut 4., 1125 Budapest, Hungary. [Wolf, Krisztina] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4., 1125 Budapest, Hungary. [Tabori, Judit] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4., 1125 Budapest, Hungary. [Karadi, Istvan] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4., 1125 Budapest, Hungary. [Varkonyi, Judit] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4., 1125 Budapest, Hungary. RP Kadar, K (reprint author), Semmelweis University, Kutvolgyi Clinical Centre, 1125 Budapest, Hungary. EM kadarka@kut.sote.hu CR Rajkumar SV, 2009, Multiple myeloma. Curr Probl Cancer 1:7–64 Kyle RA, 1983, Long-term survival in multiple myeloma. N Engl J Med 308:314–316 Bataille R, Durie BG, Grenier J, 1983, Serum beta-2 microglobulin and survival duration in multiple myeloma: a simple reliable marker for staging. Br J Haematol 55:439–447 Bataille R, Boccadoro M, Klein et al, 1992, C-reactive protein and beta-2 microglobulin produce a simple and powerful myeloma staging system. Blood 80:733–737 Eleutherakis-Papaiakovou V, Bamias A, Gika D et al, 2007, Renal failure in multiple myeloma: incidence, correlations, and prognostic significance. Leuk Lymph 48:337–341 Fonseca R, Barlogie B, Bataille R et al, 2004, Genetics and cytogenetics of multiple myeloma: a workshop report. Cancer Res 64:1546–1558 Greipp PR, Witzig TE, Gonchoroff NJ et al, 1987, Immunofluorescence labelling indices in myeloma and related monoclonal gammopathies. Mayo Clin Proc 62:969–977 San Miguel JF, Fonseca R, Greipp PR, 2004, Prognostic factors and classification for multiple myeloma: contribution to clinical management. In: Malpas JS, Bergsagel DE, Kyle RA, Anderson KC, eds, Myeloma: Biology and management, 3rd edn. Saunders, Elsevier Inc, Oxford, pp 189–199 Witzig TE, Gertz MA, Lust JA et al, 1996, Peripheral blood monoclonal plasma cells as a predictor of survival in patients with multiple myeloma. Blood 88:1780–1787 Blade J, Rozman C, Cervantes F et al, 1989, A new prognostic system for multiple myeloma based on easily available parameters. Br J Haematol 59:113–118 Cavo M, Galieni P, Zuffa E et al, 1989, Prognostic variables and clinical staging in multiple myeloma. Blood 74:1778–1780 Durie BG, Salmon SE, 1975, A clinical staging system for multiple myeloma. Correlation of measured myeloma cell mass with presenting clinical features, response to treatment, and survival. Cancer 36:842–854 Greipp PR, Katzman JA, O’Fallon WM et al, 1988, Value of beta- 2 microglobulin and plasma cell labelling indices as prognostic factors in patients with newly diagnosed myeloma. Blood 72:219– 223 Greipp PR, San Miguel J, Durie BG et al, 2005, International staging system for multiple myeloma. J Clin Oncol 23:3412–3420 Jacobson JL, Hussein MA, Barlogie B et al, 2003, Southwest Oncology Group. A new staging system for multiple myeloma patients based on the Southwest Oncology Group, SWOG, experience. Br J Haematol 122:441–450 Kumar SK, Mihael JR, Buadi FK et al, 2009, Management of newly diagnosed symptomatic multiple myeloma: updated Mayo Stratification of Myeloma and Risk-Adapted Therapy, mSMART, consensus guidelines. Mayo Clin Proc 84(12):1095–1110 Medical Research Working Party on Leukemia in Adults, 1980, Prognostic features on the third MRC myelomatosis trial. Br J Cancer 42:831–840 Merlini G, Waldesnstrom JG, Jayakar SD, 1980, A new improved clinical staging system for multiple myeloma based on analysis of 123 treated patients. Blood 55:1011–1019 San Miguel JF, Garcia Sanz R, Gonzalez M et al, 1995, A new staging system for multiple myeloma based on the number of Sphase plasma cells. Blood 85:448–455 Varkonyi J, Bajzik E, Fazakas A et al, 2009, Short or long survival in multiple myeloma. A simple method for determine the prognosis. Pathol Oncol Res 15:383–387 Kaplan EL, Meier P, 1958, Nonparametric estimation from incomplete observations. J Am Stat Assoc 53:457–481 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 557 EP 561 DI 10.1007/s12253-012-9506-z PG 5 ER PT J AU Lichao, S Liang, P Chunguang, G Fang, L Zhihua, Y Yuliang, R AF Lichao, Sun Liang, Peng Chunguang, Guo Fang, Lv Zhihua, Yang Yuliang, Ran TI Overexpression of PTGIS Could Predict Liver Metastasis and is Correlated with Poor Prognosis in Colon Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PTGIS; Colon cancer; Liver metastasis; Prognosis ID PTGIS; Colon cancer; Liver metastasis; Prognosis AB The purpose of this study was to evaluate the predictive ability of PTGIS for liver metastasis. Protein expression of PTGIS was analyzed on tissue microarray consisting of 117 CRC cases with liver metastasis (M1) and 104 cases of CRC without liver metastasis at least 5 years after resection of primary CRC (M0) by immunohistochemistry. Expression of PTGIS in 147 of 221 of primary lesions exhibited positive staining. Moreover, the PTGIS expression was significantly higher in CRC-M1 than CRC-M0 group. More importantly, the 87% (20/23) heterochronous metastatic cases showed positive staining for PTGIS. Collecting the primary and liver metastatic tumor samples from the same colon cancer patients, we tested the expression of PTGIS and revealed that the expression level of PTGIS in the hepatic metastases was noticeably higher than in the matched primary colon cancer tissues from the same patient in 9 out of 16 cases examined. Logistic regression analysis indicated that the expression of PTGIS and lymph node involvement were risk factors in colon cancer liver metastasis independent of the other variables. In leave-one-out validation model, the combination of PTGIS and lymph node involvement yielded the 89.7% satisfactory sensitivity and 83% specificity for detection of hepatic metastasis. Kaplan–Meier survival analysis revealed a correlation between higher PTGIS expression levels and shorter overall survival times. In conclusion, our results suggest that PTGIS combined with lymph node involvement may be used as accurate predictors of liver metastasis in colorectal cancer. C1 [Lichao, Sun] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, No.17 Panjiayuan Nanli, Chaoyang District, 100021 Beijing, China. [Liang, Peng] China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, 100029 Beijing, China. [Chunguang, Guo] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), Department of abdominal surgical oncology, 100021 Beijing, China. [Fang, Lv] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, No.17 Panjiayuan Nanli, Chaoyang District, 100021 Beijing, China. [Zhihua, Yang] Chinese Academy of Medical Sciences, Department of Biochemistry and Molecular Biology, No.17 Panjiayuan Nanli, Chaoyang District, 100021 Beijing, China. [Yuliang, Ran] Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, No.17 Panjiayuan Nanli, Chaoyang District, 100021 Beijing, China. RP Yuliang, R (reprint author), Chinese Academy of Medical Sciences, Peking Union Medical College, Cancer Institute (Hospital), State Key Laboratory of Molecular Oncology, 100021 Beijing, China. EM ran_yuliang@yahoo.com.cn CR Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60:277–300 McMillan DC, McArdle CS, 2007, Epidemiology of colorectal liver metastases. Surg Oncol 16:3–5 Forman BM, Chen J, Evans RM, 1997, Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors α and δ. Proc Natl Acad Sci U S A 94:4312–4317 Park BH, Vogelstein B, Kinzler KW, 2001, Genetic disruption of PPARdelta decreases the tumorigenicity of human colon cancer cells. Proc Natl Acad Sci U S A 98:2598–2603 Lim H, Gupta RA, Ma W-g et al, 1999, Cyclo-oxygenase-2- derived prostacyclin mediates embryo implantation in the mouse via PPARδ. Genes Dev 13:1561–1574 Cutler NS, Graves-Deal R, LaFleur BJ et al, 2003, Stromal production of prostacyclin confers an antiapoptotic effect to colonic epithelial cells. Cancer Res 63:1748–1751 Frigola J, Munoz M, Clark SJ, Moreno V, Capella G, Peinado MA, 2005, Hypermethylation of the prostacyclin synthase, PTGIS, promoter is a frequent event in colorectal cancer and associated with aneuploidy. Oncogene 24:7320–7326 Lambert LA, Colacchio TA, Barth RJ, 2000, Interval hepatic resection of colorectal metastases improves patient selection*. Curr Surg 57:504 Kan T, Shimada Y, Sato F et al, 2004, Prediction of lymph node metastasis with use of artificial neural networks based on gene expression profiles in esophageal squamous cell carcinoma. Ann Surg Oncol 11:1070–1078 Hu H, Sun L, Guo C et al, 2009, Tumor cell-microenvironment interaction models coupled with clinical validation reveal CCL2 and SNCG as two predictors of colorectal cancer hepatic metastasis. Clin Cancer Res 15:5485–5493 Wang D, DuBois RN, 2007, Measurement of eicosanoids in cancer tissues. Methods Enzymol 433:27–50 He T-C, Chan TA, Vogelstein B, Kinzler KW, 1999, PPAR’ is an APC-regulated target of nonsteroidal anti-inflammatory drugs. Cell 99:335–345 Munemitsu S, Albert I, Souza B, Rubinfeld B, Polakis P, 1995, Regulation of intracellular beta-catenin levels by the adenomatous polyposis coli, APC, tumor-suppressor protein. Proc Natl Acad Sci 92:3046–3050 Galaup A, Cazes A, Le Jan S et al, 2006, Angiopoietin-like 4 prevents metastasis through inhibition of vascular permeability and tumor cell motility and invasiveness. Proc Natl Acad Sci 103:18721–18726 Vane J, Corin RE, 2003, Prostacyclin: a vascular mediator. Eur J Vasc Endovasc Surg 26:571–578 Buchanan FG, Chang W, Sheng H, Shao J, Morrow JD, DuBois RN, 2004, Up-regulation of the enzymes involved in prostacyclin synthesis via Ras induces vascular endothelial growth factor. Gastroenterology 127:1391–1400 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 563 EP 569 DI 10.1007/s12253-011-9478-4 PG 7 ER PT J AU Xu, Y Liu, Z Guo, K AF Xu, Yuanhong Liu, Zhe Guo, Kejian TI The Effect of JDP2 and ATF2 on the Epithelial-mesenchymal Transition of Human Pancreatic Cancer Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic cancer; Epithelial-mesenchymal transition; Jun dimerization protein 2; Activator protein-1 ID Pancreatic cancer; Epithelial-mesenchymal transition; Jun dimerization protein 2; Activator protein-1 AB Pancreatic cancer is a common malignancy with a bleak outcome due to the early occurrence of micrometastases and poor prognosis. The epithelial-mesenchymal transition (EMT) is considered to be related to the invasion and metastasis of a variety of malignant tumors. Currently, there is no research regarding the relationship of pancreatic cancer EMT with Jun dimerization protein 2 (JDP2), an inhibitor of the activator protein-1 (AP-1) family, and activating transcription factor-2 (ATF2), an AP-1-family member. In this study, we used western blot analysis and immunofluorescence to detect the protein expression of the epithelial marker E-cadherin and the mesenchymal marker vimentin in the pancreatic cancer cell line BxPC3, which was transfected with JDP2 and induced by Collagen I. Compared with the negative control, the E-cadherin and vimentin expression levels did not change significantly, whereas E-cadherin expression decreased and vimentin expression increased in the control group transfected with empty plasmid, suggesting that JDP2 inhibits the EMT induced by Collagen I. Additionally, we verified that compared with the negative control, the morphology of the Capan2 cell line induced by TGF-β1 after transfection with ATF2 was significantly changed, as was the mRNA expression of E-cadherin, whereas the mRNA expression of vimentin, Snail, and ATF2 was significantly increased. Cell invasiveness was also significantly increased (P<0.01), suggesting that ATF2, together with TGF-β1, induced EMT in the Capan2 cell line. The members of the AP-1 family are closely related to EMT and that JDP2, as an AP-1-family inhibitor, inhibits EMT, which could lead to a new direction in molecular-targeted therapy for pancreatic cancer. C1 [Xu, Yuanhong] First Hospital of China Medical University, Department of Pancreatic Gastroenterologic Surgery, No. 92, Nanjing Rd, 110001 Shenyang, China. [Liu, Zhe] First Hospital of China Medical University, Department of Pancreatic Gastroenterologic Surgery, No. 92, Nanjing Rd, 110001 Shenyang, China. [Guo, Kejian] First Hospital of China Medical University, Department of Pancreatic Gastroenterologic Surgery, No. 92, Nanjing Rd, 110001 Shenyang, China. RP Xu, Y (reprint author), First Hospital of China Medical University, Department of Pancreatic Gastroenterologic Surgery, 110001 Shenyang, China. EM xuyhsy@yeah.net CR Jemal A, Siegel R, Ward E, Murray T, Xu J, Smigal C, Thun MJ, Cancer statistics, 2006, CA Cancer J Clin 56:106–30 Aronheim A, Zandi E, Hennemann H, Elledge SJ, Karin M, 1997, Isolation of an AP-1 repressor by a novel method for detecting protein-protein interactions. Mol Cell Biol 17:3094–102 Heinrich R, Livne E, Ben-Izhak O, Aronheim A, 2004, The c-Jun dimerization protein 2 inhibits cell transformation and acts as a tumor suppressor gene. J Biol Chem 279:5708–15 Landschulz WH, Johnson PF, McKnight SL, 1988, The leucine zipper: a hypothetical structure common to a new class of DNA binding proteins. Science 240:1759–64 Glover JN, Harrison SC, 1995, Crystal structure of the heterodimeric bZIP transcription factor c-Fos-c-Jun bound to DNA. Nature 373:257–61 Bakin AV, Rinehart C, Tomlinson AK, Arteaga CL, 2002, p38 mitogen-activated protein kinase is required for TGFbetamediated fibroblastic transdifferentiation and cell migration. J Cell Sci 115:3193–206 Cheng GZ, Chan J, Wang Q, Zhang W, Sun CD, Wang LH, 2007, Twist transcriptionally up-regulates AKT2 in breast cancer cells leading to increased migration, invasion, and resistance to paclitaxel. Cancer Res 67:1979–87 Huang TC, Kar S, Javle M, 2010, Personalized therapy for pancreatic cancer: Myth or reality in 2010? J Gastrointest Oncol 1:24–33 Kotowski A, Ma WW, 2011, Emerging therapies in pancreas cancer. J Gastrointest Oncol 2:93–103 Xiao DK, He JX, 2010, Epithelial mesenchymal transition and lung cancer. J Thorac Dis 2:154–9 Shintani Y, Hollingsworth MA, Wheelock MJ, Johnson KR, 2006, Collagen I promotes metastasis in pancreatic cancer by activating c-Jun NH(2)-terminal kinase 1 and up-regulating Ncadherin expression. Cancer Res 66:11745–53 Thuault S, Valcourt U, Petersen M, Manfioletti G, Heldin CH, Moustakas A, 2006, Transforming growth factor-beta employs HMGA2 to elicit epithelial-mesenchymal transition. J Cell Biol 174:175–83 Thuault S, Tan EJ, Peinado H, Cano A, Heldin CH, Moustakas A, 2008, HMGA2 and Smads co-regulate SNAIL1 expression during induction of epithelial-to-mesenchymal transition. J Biol Chem 283:33437–46 Davies M, Robinson M, Smith E, Huntley S, Prime S, Paterson I, 2005, Induction of an epithelial to mesenchymal transition in human immortal and malignant keratinocytes by TGF-beta1 involves MAPK, Smad and AP-1 signalling pathways. J Cell Biochem 95:918–31 Nishioka R, Itoh S, Gui T, Gai Z, Oikawa K, Kawai M, Tani M, Yamaue H, Muragaki Y, 2010, SNAIL induces epithelial-tomesenchymal transition in a human pancreatic cancer cell line, BxPC3, and promotes distant metastasis and invasiveness in vivo. Exp Mol Pathol 89:149–57 Torrisani J, Bournet B, Cordelier P, 2008, Buscail L [New molecular targets in pancreatic cancer].[Article in French]. Bull Cancer 95:503–12 Mackenzie RP, McCollum AD, 2009, Novel agents for the treatment of adenocarcinoma of the pancreas. Expert Rev Anticancer Ther 9:1473–85 Furukawa T, 2009, Molecular pathology of pancreatic cancer: implications for molecular targeting therapy. Clin Gastroenterol Hepatol 7:S35–9 Jin C, Ugai H, Song J, Murata T, Nili F, Sun K, Horikoshi M, Yokoyama KK, 2001, Identification of mouse Jun dimerization protein 2 as a novel repressor of ATF-2. FEBS Lett 489:34–41 Jin C, Li H, Murata T, Sun K, Horikoshi M, Chiu R, Yokoyama KK, 2002, JDP2, a repressor of AP-1, recruits a histone deacetylase 3 complex to inhibit the retinoic acid-induced differentiation of F9 cells. Mol Cell Biol 22:4815–26 Jin C, Kato K, Chimura T, Yamasaki T, Nakade K, Murata T, Li H, Pan J, Zhao M, Sun K, Chiu R, Ito T, Nagata K, Horikoshi M, Yokoyama KK, 2006, Regulation of histone acetylation and nucleosome assembly by transcription factor JDP2. Nat Struct Mol Biol 13:331–8 Yuanhong X, Feng X, Qingchang L, Jianpeng F, Zhe L, Kejian G, 2010, Downregulation of AP-1 repressor JDP2 is associated with tumor metastasis and poor prognosis in patients with pancreatic carcinoma. Int J Biol Markers 25:136–40 Benbrook DM, Jones NC, 1990, Heterodimer formation between CREB and JUN proteins. Oncogene 5:295–302 van Dam H, Castellazzi M, 2001, Distinct roles of Jun: Fos and Jun: ATF dimers in oncogenesis. Oncogene 20:2453–64 Berger AJ, Kluger HM, Li N, Kielhorn E, Halaban R, Ronai Z, Rimm DL, 2003, Subcellular localization of activating transcription factor 2 in melanoma specimens predicts patient survival. Cancer Res 63:8103–7 Papassava P, Gorgoulis VG, Papaevangeliou D, Vlahopoulos S, van Dam H, Zoumpourlis V, 2004, Overexpression of activating transcription factor-2 is required for tumor growth and progression in mouse skin tumors. Cancer Res 64:8573–84 Mei Y, Yuan Z, Song B, Li D, Ma C, Hu C, Ching YP, Li M, 2008, Activating transcription factor 3 up-regulated by c-Jun NH, 2)-terminal kinase/c-Jun contributes to apoptosis induced by potassium deprivation in cerebellar granule neurons. Neuroscience 151:771–9 Lee SH, Bahn JH, Whitlock NC, Baek SJ, 2010, Activating transcription factor 2, ATF2, controls tolfenamic acid-induced ATF3 expression via MAP kinase pathways. Oncogene 29:5182–92 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 571 EP 577 DI 10.1007/s12253-011-9476-6 PG 7 ER PT J AU Racz, G Csenki, Zs Kovacs, R Hegyi, Baska, F Sujbert, L Zsakovics, I Kis, R Gustafson, R Urbanyi, B Szende, B AF Racz, Gergely Csenki, Zsolt Kovacs, Robert Hegyi, Arpad Baska, Ferenc Sujbert, Laszlo Zsakovics, Ivett Kis, Renata Gustafson, Ryan Urbanyi, Bela Szende, Bela TI Subacute Toxicity Assessment of Water Disinfection Byproducts on Zebrafish SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE 4-ethylbenzaldehyde; 2; 4-difluoroaniline; Zebrafish; Water disinfection byproducts ID 4-ethylbenzaldehyde; 2; 4-difluoroaniline; Zebrafish; Water disinfection byproducts AB Disinfection of raw water is essential to the production of drinking water. However, by-products of disinfection may exert toxic effects. The potential toxic effects of two of these compounds, 4-ethylbenzaldehyde (EBA) and 2,4-difluoroaniline (DFA) were investigated using the zebrafish (Danio rerio) model. The two compounds, dissolved, were introduced in duplicate aquariums containing zebrafish in two different concentrations based on LC50 values. The aquarium water containing EBA or DFA was changed every 96 h throughout the 3 months of treatment. Behavior of the fish in each replicate was inspected twice daily. In course of treatment with both concentrations, fish exposed to DFA displayed behavior associated with visible anxiety, while EBA treated were lethargic and did not evade capture. Application of both concentrations of each component into the aquarium water resulted in dystrophic lesions in the liver, kidney and skin of the fish while preneoplastic lesions and tumors were not observed. C1 [Racz, Gergely] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26,, 1085 Budapest, Hungary. [Csenki, Zsolt] Szent Istvan University, Faculty of Agricultural and Environmental Sciences, Department of Fish CultureGodollo, Hungary. [Kovacs, Robert] Szent Istvan University, Faculty of Agricultural and Environmental Sciences, Department of Fish CultureGodollo, Hungary. [Hegyi, Arpad] Szent Istvan University, Faculty of Agricultural and Environmental Sciences, Department of Fish CultureGodollo, Hungary. [Baska, Ferenc] Szent Istvan University, Faculty of Veterinary Medicine, Department of Pathology and Forensic Veterinary Medicine, 1078 Budapest, Hungary. [Sujbert, Laszlo] Semmelweis University, Department of Public Health, Nagyvarad ter 4, 1089 Budapest, Hungary. [Zsakovics, Ivett] 3D HISTECH Kft, Konkoly-Thege ut 29-33, 1121 Budapest, Hungary. [Kis, Renata] 3D HISTECH Kft, Konkoly-Thege ut 29-33, 1121 Budapest, Hungary. [Gustafson, Ryan] University of North Carolina at Chapel Hill, Gillings School of Global Public Health, Department of Environmental Sciences & EngineeringChapel Hill, NC, USA. [Urbanyi, Bela] Szent Istvan University, Faculty of Agricultural and Environmental Sciences, Department of Fish CultureGodollo, Hungary. [Szende, Bela] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26,, 1085 Budapest, Hungary. RP Szende, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM bszende@korb1.sote.hu CR Nieuwenhuijsen MJ, Toledano MB, Elliott P, 2000, Uptake of chlorination disinfection by-products; a review and a discussion of its implications for exposure assessment in epidemiological studies. J Expo Anal Environ Epidemiol 10:586–599 Cognet L, Courtois Y, Mallevialle J, 1986, Mutagenic activity of disinfection by-products. Environ Health Perspect 69:165–175 Woodruff NWet al, 2001, Human cell mutagenicity of chlorinated and unchlorinated water and the disinfection byproduct 3-chloro-4-, dichloromethyl)-5-hydroxy-2(5H)-furanone, MX). Mutat Res 495:157–168 Morris RD et al, 1992, Chlorination, chlorination by-products, and cancer: a meta-analysis. Am J Public Health 82:955–963 Bove GE Jr, Rogerson PA, Vena JE, 2007, Case control study of the geographic variability of exposure to disinfectant byproducts and risk for rectal cancer. Int J Health Geogr 6:18 Villanueva CM et al, 2007, Bladder cancer and exposure to water disinfection by-products through ingestion, bathing, showering, and swimming in pools. Am J Epidemiol 165:148–156 Sujbert L et al, 2006, Genotoxic potential of by-products in drinking water in relation to water disinfection: survey of preozonated and post-chlorinated drinking water by Ames-test. Toxicology 219:106–112 Racz G et al, 2004, Rapid communication: water disinfection byproducts enhanced apoptotic activity in human lymphocytes. J Toxicol Environ Health A 67:1315–1319 Krasner SW et al, 2006, Occurrence of a new generation of disinfection byproducts. Environ Sci Technol 40:7175–7185 Berghmans S et al, 2005, Making waves in cancer research: new models in the zebrafish. Biotechniques 39:227–237 Simon K, Lapis K, 1984, Carcinogenesis studies on guppies. Natl Canc Inst Monogr 65:71–81 Amatruda JF et al, 2002, Zebrafish as a cancer model system. Canc Cell 1:229–231 OECD, OECD 203, 1992, OECD Guidelines for testing of chemicals, 1992. Effects on biotic systems. Method 203. Fish, Acute Toxicity Test. Adopted July 17, 1992 Mulrane L et al, 2008, Automated image analysis in histopathology: a valuable tool in medical diagnostics. Expert Rev Mol Diagn 8:707– 725 Krenacs T et al, 2010, Digital microscopy for boosting database integration and analysis in TMA studies.MethMol Biol 664:163–175 Blaser RE, Chadwick L, McGinnis GC, 2010, Behavioral measures of anxiety in zebrafish, Danio rerio). Behav Brain Res 208:56–62 Rook JJ, 1974, Formation of haloforms during chlorination of natural waters. Wat Treat Exam 2:234–243 von Gunten U et al, 2001, By-products formation during drinking water disinfection: a tool to assess disinfection efficiency? Water Res 35:2095–2099 Cantor KP, 1982, Epidemiological evidence of carcinogenicity of chlorinated organics in drinking water. Environ Health Perspect 46:187–195 Plewa MJ et al, 2002, Mammalian cell cytotoxicity and genotoxicity analysis of drinking water disinfection by-products. Environ Mol Mutagen 40:134–142 Lele Z, Krone PH, 1996, The zebrafish as a model system in developmental, toxicological and transgenic research. Biotechnol Adv 14:57–72 Lammer E et al, 2009, Development of a flow-through system for the fish embryo toxicity test, FET, with the zebrafish, Danio rerio). Toxicol In Vitro 23:1436–1442 Peterson SM, Freeman JL, 2009, Cancer cytogenetics in the zebrafish. Zebrafish 6:355–360 Mione MC, Trede NS, 2010, The zebrafish as a model for cancer. Dis Model Mech 3:517–523 Sullivan C, Kim CH, 2008, Zebrafish as a model for infectious disease and immune function. Fish Shellfish Immunol 25:341– 350 Khudoley VV, 1984, Use of aquarium fish, Danio rerio and Poecilia reticulata, as test species for evaluation of nitrosamine carcinogenicity. Natl Canc Inst Monogr 65:65–70 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 579 EP 584 DI 10.1007/s12253-011-9479-3 PG 6 ER PT J AU Chiang, Y Zhou, X Wang, Z Song, Y Liu, Z Zhao, F Zhu, J Xu, H AF Chiang, Yeunpo Zhou, Xin Wang, Zhenning Song, Yongxi Liu, Zhuangkai Zhao, Fang Zhu, Jinliang Xu, Huimian TI Expression Levels of MicroRNA-192 and -215 in Gastric Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Clinicopathological characteristics; Gastric cancer; microRNA; miR-192; miR-215 ID Clinicopathological characteristics; Gastric cancer; microRNA; miR-192; miR-215 AB MicroRNAs (miRNAs) are small, non-coding RNAs of endogenous origin. Accumulating studies have shown aberrant miRNA expression plays an important role in many tumor types. miR-192 and -215, which have the same "seed region", have not been comprehensively investigated using a large number of cases in gastric cancer. The total RNA was extracted from 118 gastric cancer tissues and three gastric cancer cell lines as well as matched non-tumor adjacent tissues (NATs). After polyadenylation and reverse transcription, expression levels of miR-192 and -215 were determined by real-time PCR and calculation using the 2-ΔΔCT method for evaluation of the association between miR-192, and -215 expression levels and clinicopathological characteristics. There were no significant differences in miR-192 and -215 expression levels between gastric cancer tissues and non-tumor counterparts (both p>0.05, paired ttest). Interestingly, miR-192 and -215 were down-regulated in MGC-803 cells, BGC-823 cells and SGC-7901 cells (all p<0.01, paired t-test). Also, the down-regulation of miR-192 and -215 was demonstrated to be associated with increased tumor sizes (both p00.003, Mann–Whitney U test) and advanced Borrmann type tumors (p00.015 and p00.044, respectively, Kruskal-Wallis H test). Moreover, the expression of miR-192 was significantly lower in the pT4 stage of gastric cancer than in pT1, pT2 and pT3 stages (p00.026). Furthermore, there was a strong correlation between miR-192 and -215 in gastric cancer tissues (p<0.001, Pearson regressions). miR-192 and -215 might be related to the proliferation and invasion of gastric cancer. Potentially, they could become important biomarkers. C1 [Chiang, Yeunpo] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, 110001 Shenyang, China. [Zhou, Xin] ShengJing Hospital of China Medical University, Department of Gynecology and ObstetricsShenyang, China. [Wang, Zhenning] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, 110001 Shenyang, China. [Song, Yongxi] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, 110001 Shenyang, China. [Liu, Zhuangkai] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, 110001 Shenyang, China. [Zhao, Fang] ShengJing Hospital of China Medical University, Department of Gynecology and ObstetricsShenyang, China. [Zhu, Jinliang] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, 110001 Shenyang, China. [Xu, Huimian] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, 110001 Shenyang, China. RP Wang, Z (reprint author), China Medical University, Affiliated First Hospital, Department of Surgical Oncology, 110001 Shenyang, China. EM josieon826@yahoo.com.cn CR Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281–297 Calin GA, Croce CM, 2006, MicroRNA-cancer connection: the beginning of a new tale. Cancer Res 66:7390–7394 Esquela-Kerscher A, Slack FJ, 2006, Oncomirs—microRNAs with a role in cancer. Nat Rev Cancer 6:259–269 Hwang HW, Mendell JT, 2006, MicroRNAs in cell proliferation, cell death, and tumorigenesis. Br J Cancer 94:776–780 Cho WC, 2007, OncomiRs: the discovery and progress of micro- RNAs in cancers. Mol Canc 6:60 Calin GA, Sevignani C, Dumitru CD et al, 2004, Human micro- RNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A 101:2999– 3004 Bandi N, Vassella E, 2011, miR-34a and miR-15a/16 are coregulated in non-small cell lung cancer and control cell cycle progression in a synergistic and Rb-dependent manner. Mol Canc 10:55 Feber A, Xi L, Pennathur A et al, 2011, MicroRNA prognostic signature for nodal metastases and survival in esophageal adenocarcinoma. Ann Thorac Surg 91:1523–1530 Guo J, Miao Y, Xiao B et al, 2009, Differential expression of microRNA species in human gastric cancer versus non-tumorous tissues. J Gastroenterol Hepatol 24:652–657 Park JK, Henry JC, Jiang J et al, 2011, miR-132 and miR-212 are increased in pancreatic cancer and target the retinoblastoma tumor suppressor. Biochem Biophys Res Commun 406:518– 523 Chiang Y, Song Y, Wang Z et al, 2011, Aberrant expression of miR-203 and its clinical significance in gastric and colorectal cancers. J Gastrointest Surg 15:63–67 Marchini S, Cavalieri D, Fruscio R et al, 2011, Association between miR-200c and the survival of patients with stage I epithelial ovarian cancer: a retrospective study of two independent tumour tissue collections. Lancet Oncol 12:273–285 Tsukamoto Y, Nakada C, Noguchi T et al, 2010, MicroRNA-375 is downregulated in gastric carcinomas and regulates cell survival by targeting PDK1 and 14-3-3zeta. Cancer Res 70:2339–2349 Chen Y, Song Y, Wang Z, Yue Z, Xu H, Xing C, 2010, Altered expression of miR-148a and miR-152 in gastrointestinal cancers and its clinical significance. J Gastrointest Surg 14:1170– 1179 Song YX, Yue ZY, Wang ZN et al, 2011, MicroRNA-148b is frequently down-regulated in gastric cancer and acts as a tumor suppressor by inhibiting cell proliferation. Mol Canc 10:1 Xiong X, Ren HZ, Li MH, Mei JH, Wen JF, Zheng CL, 2011, Down-regulated miRNA-214 induces a cell cycle G1 arrest in gastric cancer cells by up-regulating the PTEN protein. Pathol Oncol Res 17:931–937 Shi R, Chiang VL, 2005, Facile means for quantifying microRNA expression by real-time PCR. Biotechniques 39:519–525 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative pcr and the 2, delta delta c, T), method. Methods 25:402–408 Du Y, Xu Y, Ding L et al, 2009, Down-regulation of miR-141 in gastric cancer and its involvement in cell growth. J Gastroenterol 44:556–561 Shah AA, Leidinger P, Blin N, Meese E, 2010, miRNA: small molecules as potential novel biomarkers in cancer. Curr Med Chem 17:4427–4432 Yanaihara N, Caplen N, Bowman E et al, 2006, Unique micro- RNA molecular profiles in lung cancer diagnosis and prognosis. Canc Cell 9:189–198 Mathe EA, Nguyen GH, Bowman ED et al, 2009, MicroRNA expression in squamous cell carcinoma and adenocarcinoma of the esophagus: associations with survival. Clin Cancer Res 15:6192– 6200 Gui J, Tian Y, Wen X et al, 2011, Serum microRNA characterization identifies miR-885-5p as a potential marker for detecting liver pathologies. Clin Sci, Lond, 120:183–193 Braun CJ, Zhang X, Savelyeva I et al, 2008, p53-responsive micrornas 192 and 215 are capable of inducing cell cycle arrest. Cancer Res 68:10094–10104 Georges SA, Biery MC, Kim SY et al, 2008, Coordinated regulation of cell cycle transcripts by p53-Inducible microRNAs, miR- 192 and miR-215. Cancer Res 68:10105–10112 Davidson LA,Wang N, Shah MS, Lupton JR, Ivanov I, Chapkin RS, 2009, n-3 polyunsaturated fatty acids modulate carcinogen-directed non-coding microRNA signatures in rat colon. Carcinogenesis 30:2077–2084 Baffa R, Fassan M, Volinia S et al, 2009, MicroRNA expression profiling of human metastatic cancers identifies cancer gene targets. J Pathol 219:214–221 Jin Z, Selaru FM, Cheng Y et al, 2011, MicroRNA-192 and -215 are upregulated in human gastric cancer in vivo and suppress ALCAM expression in vitro. Oncogene 30:1577–1585 Volinia S, Calin GA, Liu CG et al, 2006, A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 103:2257–2261 Lee EJ, Gusev Y, Jiang J et al, 2007, Expression profiling identifies microRNA signature in pancreatic cancer. Int J Cancer 120:1046– 1054 Quach H, Barreiro LB, Laval G et al, 2009, Signatures of purifying and local positive selection in human miRNAs. Am J Hum Genet 84:316–327 Kim DY, Joo JK, Park YK, Ryu SY, Kim YJ, Kim SK, 2007, Predictors of long-term survival in node-positive gastric carcinoma patients with curative resection. Langenbeck Arch Surg 392:131–134 Shiraishi N, Inomata M, Osawa N, Yasuda K, Adachi Y, Kitano S, 2000, Early and late recurrence after gastrectomy for gastric carcinoma. Cancer 89:255–261 Kim JP, Kim YW, Yang HK, Noh DY, 1994, Significant prognostic factors by multivariate analysis of 3926 gastric cancer patients. World J Surg 18:872–878 Pichiorri F, Suh SS, Rocci A et al, 2010, Downregulation of p53- inducible microRNAs 192, 194, and 215 impairs the p53/MDM2 autoregulatory loop in multiple myeloma development. Canc Cell 18:367–381 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 585 EP 591 DI 10.1007/s12253-011-9480-x PG 7 ER PT J AU Tokes, AM Szasz, MA Juhasz, Schaff, Zs Harsanyi, L Molnar, AI Baranyai, Zs Besznyak, I Zarand, A Salamon, F Kulka, J AF Tokes, Anna-Maria Szasz, Marcell Attila Juhasz, Eva Schaff, Zsuzsa Harsanyi, Laszlo Molnar, Arthur Istvan Baranyai, Zsolt Besznyak, Istvan Zarand, Attila Salamon, Ferenc Kulka, Janina TI Expression of Tight Junction Molecules in Breast Carcinomas Analysed by Array PCR and Immunohistochemistry SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast carcinoma; Tight junction; Claudin; Array; Immunohistochemistry ID Breast carcinoma; Tight junction; Claudin; Array; Immunohistochemistry AB In the past few decades an enormous amount of data became known to clarify the molecular composition and architecture of tight junctions (TJs). Despite the efforts, the expression and function of several TJ genes and proteins in breast carcinoma are still not known and some of the data are contradictory. The expression of forty-four TJ associated genes was examined at mRNA level in eighteen invasive ductal breast carcinoma samples and corresponding normal breast tissues by using low density array PCR. Expressions of claudins (CLDNs) 5, 10, 16, 17, and 18, and ZO-1, ZO-2 were evaluated by immunohistochemistry as well. Using immunohistochemical phenotype as a surrogate for the genetic subtype, 11 luminal A, 3 luminal B, 3 triple negative and one HER2+ cases were included. Ten genes were significantly downregulated in tumors compared with normal breast tissues (CLDNs 5, 10, 16, 18, 19, CTNNAL1, JAM-B, ZO-1, ZO-2 and PARD3), whereas one gene (CLDN17) was significantly up-regulated in tumors when compared with normal breast. At protein level CLDNs 5, 10, 16, 18, ZO-1 and ZO-2 were downregulated in tumors as compared with normal breast tissue. CLDN17 showed variable expression in tumor tissues in comparison to normal breast. In the single HER2+ tumor when compared with the other subtypes CLDNs 5, 16, 17, 18, CTNNAL1, JAM-B, ZO-1, ZO-2 and PARD3 genes were found to be upregulated. We found altered TJ genes and proteins whose expression has not yet been associated with breast carcinoma. Our findings show a tendency of TJ genes and proteins to be downregulated in breast cancer. Further studies are necessary to examine whether the downregulation of the above mentioned TJ associated genes and proteins may contribute to the malignant progression of invasive ductal breast carcinomas. C1 [Tokes, Anna-Maria] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Szasz, Marcell Attila] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Juhasz, Eva] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Harsanyi, Laszlo] Semmelweis University, 1st Department of Surgery, Ulloi ut 78, 1091 Budapest, Hungary. [Molnar, Arthur Istvan] Semmelweis University, 1st Department of Surgery, Ulloi ut 78, 1091 Budapest, Hungary. [Baranyai, Zsolt] Fovarosi Onkormanyzat Uzsoki utcai Oktato Korhaz, Sebeszeti-Ersebeszeti Osztaly, Uzsoki u. 29, 1145 Budapest, Hungary. [Besznyak, Istvan] Fovarosi Onkormanyzat Uzsoki utcai Oktato Korhaz, Sebeszeti-Ersebeszeti Osztaly, Uzsoki u. 29, 1145 Budapest, Hungary. [Zarand, Attila] Fovarosi Onkormanyzat Uzsoki utcai Oktato Korhaz, Sebeszeti-Ersebeszeti Osztaly, Uzsoki u. 29, 1145 Budapest, Hungary. [Salamon, Ferenc] Fovarosi Uzsoki utcai Korhaz, Pathologia, Uzsoki u. 29, 1145 Budapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. RP Tokes, AM (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM tokesa1972@yahoo.co.uk CR Brennan K, Offiah G, McSherry EA et al, 2010, Tight junctions: a barrier to the initiation and progression of breast cancer? J Biomed Biotechnol., DOI 10.1155/2010/460607 Gonzalez-Mariscal L, Betanzos A, Nava P et al, 2003, Tight junction proteins. Progr Biophys Mol Biol 81:1–44 Gonzalez-Mariscal L, Lechuga S, Garay E, 2007, Role of tight junctions in cell proliferation and cancer. Progr HistochemCytochem 42:1–57 Lelievre SA, 2010, Tissue polarity-dependent control of mammary epithelial homeostasis and cancer development: an epigenetic perspective. J Mammary Gland Biol Neoplasia 15:49–63 Alroy J, 1979, Tight junctions adjacent to tumor stromal interface in human invasive transitional cell carcinomas. Virchows Arch 30:289–296 Martinez-Palomo A, 1970, Ultrastructural modifications of intercellular junctions between tumor cells. In Vitro 6:15–20 Martinez-Palomo A, 1970, Ultrastructural modifications of intercellular junctions in some epithelial tumors. Lab Investig 22:605–614 Hough CD, Sherman-Baust CA, Pizer ES et al, 2000, Large-scale serial analysis of gene expression reveals genes differentially expressed in ovarian cancer. Cancer Res 60:6281–6287 Hoevel T, Macek R, Mundigl O et al, 2002, Expression and targeting of the tight junction protein CLDN1 in CLDN1- negative human breast tumor cells. J Cell Physiol 191:60–68 Hoevel T, Macek R, Swisshelm K et al, 2004, Reexpression of the TJ protein CLDN1 induces apoptosis in breast tumor spheroids. Int J Cancer 108:374–383 Kominsky SL, Argani P, Korz D et al, 2003, Loss of the tight junction protein claudin-7 correlates with histological grade in both ductal carcinoma in situ and invasive ductal carcinoma of the breast. Oncogene 22:2021–2033 Kramer F, White K, Kubbies M et al, 2000, Genomic organization of claudin-1 and its assessment in hereditary and sporadic breast cancer. Human Genet 107:249–256 Sobel G, Paska C, Szabo I et al, 2005, Increased expression of claudins in cervical squamous intraepithelial neoplasia and invasive carcinoma. Human Pathol 36:162–169 Sobel G, Nemeth J, Kiss A et al, 2006, Claudin 1 differentiates endometrioid and serous papillary endometrial adenocarcinoma. Gynecol Oncol 103:591–598 Blanchard AA, Skliris GP, Watson PH et al, 2009, Claudins 1, 3, and 4 protein expression in ER negative breast cancer correlates with markers of the basal phenotype. Virchows Arch 454:647–656 Erin N, Wang N, Xin P et al, 2009, Altered gene expression in breast cancer liver metastases. Int J Cancer 124:1503–1516 Hewitt KJ, Agarwal R, Morin PJ, 2006, The claudin gene family: expression in normal and neoplastic tissues. BMC Cancer 6:186 Kim TH, Huh JH, Lee S et al, 2008, Down-regulation of claudin-2 in breast carcinomas is associated with advanced disease. Histopathology 53:48–55 Kulka J, Szasz AM, Nemeth Z et al, 2005, Expression of tight junction protein claudin-4 in basal-like breast carcinomas. Pathol Oncol Res 15:59–64 Lanigan F, McKiernan E, Brennan DJ et al, 2009, Increased claudin-4 expression is associated with poor prognosis and high tumour grade in breast cancer. Int J Cancer 124:2088–2097 Osanai M, Murata M, Nishikiori N et al, 2007, Occludin-mediated premature senescence is a fail-safe mechanism against tumorigenesis in breast carcinoma cells. Cancer Sci 98:1027–1034 Tokes AM, Kulka J, Paku S et al, 2005, Claudin-1, −3 and −4 proteins and mRNA expression in benign and malignant breast lesions: a research study. Breast Cancer Res 7:296–305 Martin TA, Harrison GM, Watkins G et al, 2008, Claudin-16 reduces the aggressive behavior of human breast cancer cells. J Cell Biochem 105:41–52 Naik MU, Naik TU, Suckow AT et al, 2008, Attenuation of junctional adhesion molecule-A is a contributing factor for breast cancer cell invasion. Cancer Res 68:2194–2203 Naik UP, Naik MU, 2008, Putting the brakes on cancer cell migration: JAM-A restrains integrin activation. Cell Adh Migr 2:249–251 Morohashi S, Kusumi T, Sato F et al, 2007, Decreased expression of claudin-1 correlates with recurrence status in breast cancer. Int J Mol Med 20:139–143 Creighton CJ, Li X, Landis M et al, 2009, Residual breast cancers after conventional therapy display mesenchymal as well as tumorinitiating features. PNAS 106:13820–13825 Herschkowitz JI, Simin K, Weigman VJ et al, 2007, Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol 8:R76 Prat A, Perou CM, 2009, Mammary development meets cancer genomics. Nat Med 15:842–844 Prat A, Perou CM, 2011, Deconstructing the molecular portraits of breast cancer. Mol Oncol 5:5–2331 Perou CM, Sorlie T, Eisen MB et al, 2000, Molecular portraits of human breast tumours. Nature 406:747–752 Escudero-Esparza A, Jiang WG, Martin TA, 2011, The Claudin family and its role in cancer and metastasis. Front Biosci 1:1069–1083 Turksen K, Troy TC, 2011, Junctions gone bad: claudins and loss of the barrier in cancer. Biochim Biophys Acta 1:73–79 Dahiya N, Becker KG, Wood WH et al, 2011, Claudin-7 is frequently overexpressed in ovarian cancer and promotes invasion. PLoS One 6:e22119 Amasheh S, Meiri N, Gitter AH et al, 2002, Claudin-2 expression induces cation-selective channels in tight junctions of epithelial cells. J Cell Sci 115:4969–4976 Amasheh S, Schmidt T, Mahn M et al, 2005, Contribution of claudin-5 to barrier properties in tight junctions of epithelial cells. Cell Tissue Res 321:89–96 Nguyen DA, Neville MC, 1998, Tight junction regulation in the mammary gland. J Mammary Gland Biol Neoplasia 3:233–246 Schulzke JD, Fromm M, 2009, Tight junctions: molecular structure meets function. Ann NYAcad Sci 1165:1–6 Tsukita S, Furuse M, 2000, The structure and function of claudins, cell adhesion molecules at tight junctions. Ann NY Acad Sci 915:129–135 Turunen M, Talvensaari-Mattila A, Soini Y et al, 2009, Claudin-5 overexpression correlates with aggressive behavior in serous ovarian adenocarcinoma. Anticancer Res 29:5185–5189 Szasz A, Tokes A, Micsinai MJ et al, 2011, Prognostic significance of claudin expression changes in breast cancer with regional lymph node metastasis. Clin Exp Metastasis 28:55–63 Nemeth Z, Szasz AM, Tatrai P et al, 2009, Claudin-1, −2, −3, −4, −7, −8, and −10 protein expression in biliary tract cancers. J Histochem Cytochem 57:113–121 Sanada Y, Oue N, Mitani Y et al, 2006, Down-regulation of the claudin-18 gene, identified through serial analysis of gene expression data analysis, in gastric cancer with an intestinal phenotype. J Pathol 208:633–642 Hou J, Renigunta A, Gomes AS et al, 2009, Claudin-16 and claudin-19 interaction is required for their assembly into tight junctions and for renal reabsorption of magnesium. PNAS 106:15350– 15355 Zen K, Yasui K, Gen Y et al, 2009, Defective expression of polarity protein PAR-3 gene, PARD3, in esophageal squamous cell carcinoma. Oncogene 28:2910–2918 Ding L, Ellis MJ, Li S et al, 2010, Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature 464:999– 1005 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 593 EP 606 DI 10.1007/s12253-011-9481-9 PG 14 ER PT J AU Arvai, K Nagy, K Barti-Juhasz, H Petak, I Krenacs, T Micsik, T Vegso, Gy Perner, F Szende, B AF Arvai, Kristof Nagy, Katalin Barti-Juhasz, Helga Petak, Istvan Krenacs, Tibor Micsik, Tamas Vegso, Gyula Perner, Ferenc Szende, Bela TI Molecular Profiling of Parathyroid Hyperplasia, Adenoma and Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Parathyroid lesions; Apoptosis; Gene expression profiling; Tissue micro array ID Parathyroid lesions; Apoptosis; Gene expression profiling; Tissue micro array AB The objective of the study was to examine proliferation and apoptosis associated gene expression in the whole sequence parathyroid lesions to reveal specific features of carcinoma. This study was based on surgically removed parathyroid tissues, gene expression analysis was performed both at gene and protein level. First, mRNA isolation was performed from deep-frozen tissue samples, and further apoptosis pathway-specific cDNA macroarray analysis was carried out. The results were validated with real-time PCR. Subsequently, protein expression was analyzed with immunhistochemistry on TissueMicro Array multi-blocks derived from several paraffinembedded samples. cDNA macroarrays revealed elevated expression of both pro-apoptotic (FAS receptor, TRAIL ligand, CASPASE8, and −4) and anti-apoptotic (cIAP1, APOLLON) genes in benign proliferative lesions compared to that in normal gland. TMA studies showed overexpression of KI67, P53, SURVIVIN and APOLLON protein and failure of expression of P27, BCL2, BAX, CHROMOGRANIN-A, SYNAPTOPHYSIN, CYCLIND1, FLIP, TRAIL, CK8, CK18, CK19 in parathyroid carcinoma was detected. These alterations in gene expression of the investigated products could be used in differentiation between beningn and malignant proliferative processes of the parathyroid gland. Authors conclude that a series of alterations in gene expression such as overexpression of APOLLON, P53, KI67 and suppression of P27, BCL2, BAX lead to uncontrolled cell proliferation, but still not leading to increased apoptotic activity in parathyroid carcinoma. C1 [Arvai, Kristof] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Nagy, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Barti-Juhasz, Helga] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Petak, Istvan] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Micsik, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Vegso, Gyula] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Perner, Ferenc] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Szende, Bela] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. RP Szende, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM bszende@korb1.sote.hu CR Fernandez-Ranvier GG, Khanafshar E, Jensen K et al, 2007, Parathyroid carcinoma, atypical parathyroid adenoma, or parathyromatosis? Cancer 110(2):255–264 Adami S, Marcocci C, Gatti D. Epidemiology of primary hyperparathyroidism in Europe. J Bone Miner Res 17 Suppl 2:N18-23 Marx SJ, 2000, Hyperparathyroid and hypoparathyroid disorders. N Engl J Med 343:1863–1875 Fernandez-RanvierGG, Khanafshar E, TachaD et al, 2009, Defining a molecular phenotype for benign and malignant parathyroid tumors. Cancer 115(2):334–344 Stojadinovic A, Hoos A, Nissan A et al, 2003, Parathyroid neoplasms: clinical, histopathological, and tissue microarray-based molecular analysis. Hum Pathol 34(1):54–64 Szende B, Farid P, Vegso G et al, 2004, Apoptosis and P53, Bcl-2 and Bax gene expression in parathyroid glands of patients with hyperparathyroidism. Pathol Oncol Res 10(2): 98–103 Vargas MP, Vargas HI, Kleiner DE et al, 1997, The role of prognostic markers, MiB-1, RB, and bcl-2, in the diagnosis of parathyroid tumors. Mod Pathol 10(1):12–17 Hadar T, Shvero J, Yaniv E et al, 2005, Expression of p53, Ki-67 and Bcl-2 in parathyroid adenoma and residual normal tissue. Pathol Oncol Res 11(1):45–49 Szende B, Arvai K, Petak I et al, 2006, Changes in gene expression in the course of proliferative processes in the parathyroid gland. Magy Onkol 50(2):137–140 Solcia E, Kloppel G, Sobin LH, 2000, Histological typing of endocrine tumours; international histological classification of tumours. WHO Ed. 2. Springer; p.48–55 Shane E, Bilezikian JP, 1982, Parathyroid carcinoma: a review of 62 patients. Endocr Rev 3(2):218–226 Favia G, Lumachi F, Polistina F et al, 1998, Parathyroid carcinoma: sixteen new cases and suggestions for correct management. World J Surg 22(12):1225–1230 Hundahl SA, Fleming ID, Fremgen AM et al, 1999, Two hundred eighty-six cases of parathyroid carcinoma treated in the U.S. between 1985–1995: a National Cancer Data Base Report. Cancer 1;86, 3):538–44 Busaidy NL, Jimenez C, Habra MA et al, 2004, Parathyroid carcinoma: a 22-year experience. Head Neck 26(8):716–726 Haven CJ, Howell VM, Eilers PH et al, 2004, Gene expression of parathyroid tumors: molecular subclassification and identification of the potential malignant phenotype. Cancer Res 64(20):7405–7411 Woodard GE, Lin L, Zhang JH et al, 2005, Parafibromin, product of the hyperparathyroidism-jaw tumor syndrome gene HRPT2, regulates cyclin D1/PRAD1 expression. Oncogene 10;24(7):1272–6 Cetani F, Pardi E, Ambrogini E et al, 2007, Different somatic alterations of the HRPT2 gene in a patient with recurrent sporadic primary hyperparathyroidism carrying an HRPT2 germline mutation. Endocr Relat Cancer 14(2):493–499 Iacobone M, Masi G, Barzon L et al, 2009, Hyperparathyroidism-jaw tumor syndrome: a report of three large kindred. Langenbecks Arch Surg 394(5):817–825 Yang Y-J, Han J-W, Youn H-D et al, 2010, The tumor suppressor, parafibromin, mediates histone H3 K9 methylation for cyclin D1 repression. Nucleic Acids Res 38:382–390 Masi G, Barzon L, Iacobone M et al, 2008, Clinical, genetic, and histopathologic investigation of CDC73-related familial hyperparathyroidism. Endocr Relat Cancer 15(4):1115–1126 Shih RY, Fackler S, Maturo S et al, 2009, Parathyroid carcinoma in multiple endocrine neoplasia type 1 with a classic germline mutation. Endocr Pract 15(6):567–572 Cetani F, Ambrogini E, Viacava P et al, 2007, Should parafibromin staining replace HRTP2 gene analysis as an additional tool for histologic diagnosis of parathyroid carcinoma? Eur J Endocrinol 156(5):547–554 Howell VM, Gill A, Clarkson A et al, 2009, Accuracy of combined protein gene product 9.5 and parafibromin markers for immunohistochemical diagnosis of parathyroid carcinoma. J Clin Endocrinol Metab 94(2):434–441 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 607 EP 614 DI 10.1007/s12253-011-9483-7 PG 8 ER PT J AU Helpap, B Kristiansen, G Beer, M Kollermann, J Oehler, U Pogrebniak, A Fellbaum, Ch AF Helpap, Burkhard Kristiansen, Glen Beer, Michaela Kollermann, Jens Oehler, Ulrich Pogrebniak, Alexsei Fellbaum, Christian TI Improving the Reproducibility of the Gleason Scores in Small Foci of Prostate Cancer - Suggestion of Diagnostic Criteria for Glandular Fusion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate carcinoma; Glandular fusion; Interobserver reproducibility ID Prostate carcinoma; Glandular fusion; Interobserver reproducibility AB High upgrading rates of Gleason score 6 to 7 carcinomas between biopsy and radical prostatectomy specimens may be produced by change of fused glands of pattern 3 to pattern 4. Therefore, inter-observer reproducibility of fused and non-fused glands in biopsy specimens was analysed. Images of H&E stained slides of glands of carcinomas with Gleason score 6 and 7 (3+4) with and without glandular fusions with different lens magnification were analysed by 4 specialized genitourinary pathologists and 3 non-specialized pathologists. The definition of glandular fusion was a complete lack of any stromal fibres between a minimum of two glands and only one line of nuclei within the area of fusion. Overall agreement and interobserver reproducibility of fused versus non-fused glands of non- and uro-pathologically specialized pathologists were lower in lens magnification of 50× in contrast to 200×. The inter-observer reproducibility of fused glands by specialized observer was higher than that of nonspecialized pathologists. The results support the importance of strict but practicable criteria for the diagnosis of fused tumor glands in order to decrease the interobserver variability of Gleason scores, particularly in nonspecialised pathologists. C1 [Helpap, Burkhard] Hegau-Bodensee Hospital of Singen, Department of Pathology, Virchow Str. 10, 78224 Singen, Germany. [Kristiansen, Glen] University of Bonn, Institute of PathologyBonn, Germany. [Beer, Michaela] University of Zurich, Institute of Surgical PathologyZurich, Switzerland. [Kollermann, Jens] HSK- Hospital, Institute of PathologyWiesbaden, Germany. [Oehler, Ulrich] Hegau-Bodensee Hospital of Singen, Department of Pathology, Virchow Str. 10, 78224 Singen, Germany. [Pogrebniak, Alexsei] Hegau-Bodensee Hospital of Singen, Department of Pathology, Virchow Str. 10, 78224 Singen, Germany. [Fellbaum, Christian] Hegau-Bodensee Hospital of Singen, Department of Pathology, Virchow Str. 10, 78224 Singen, Germany. RP Helpap, B (reprint author), Hegau-Bodensee Hospital of Singen, Department of Pathology, 78224 Singen, Germany. EM burkhard.helpap@hbh-kliniken.de CR Epstein JI, Allsbrook WC, Amin MB, 2005, Egevad L and the ISUP Grading Committee The 2005 international society of urological pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol 29:1227– 1242 Egevad L, 2008, Recent trends in Gleason grading of prostate cancer II Prognosis, reproducibility and reporting. Anal Quant Cytol Histol 30:254–260 Helpap B, 2006, Egevad L The significance of modified Gleason grading of prostatic carcinoma in biopsy and radical prostatectomy specimens. Virchows Arch 449:622–627 Veloso SG, Lima MF, Salles PG, Berenstein CK, Scalon JD, Bambirra EA, 2007, Interobserver agreement of Gleason score and modified Gleason score in needle biopsy and in surgical specimen of prostate cancer. Int Braz J Urol 33:639–646 Zareba P, Zhang J, Yilmaz A, Trykov K, 2009, The impact of the 2005 international society of urological pathology, ISUP, consensus on Gleason grading in contemporary practice. Histopathology 55:384–391 Helpap B, Egevad L, 2009, Clinical insignificance of prostate cancer. Are there morphological findings? Urologe 48:170–174 Gleason DE, 1966, Classification of prostatic carcinomas. Cancer Chemother Rep 50:125–128 Gleason DE, Mellinger GT, 1974, Prediction of prognosis for prostatic adenocarcinoma by combined histological grading and clinical staging. J Urol 111:58–64 Gleason DF, 1977, Histological grading and clinical staging of prostatic carcinoma. In: Tannenbaum M, ed, Urologic pathology: the prostate Philadelphia, Lea& Feibiger, 171–198 Gleason DF, 1992, Histologic grading of prostatic cancer. A perspective. Hum Pathol 23:273–279 Mellinger GT, Gleason D, Bailar J, 1967, The histology and prognosis of prostatic cancer. J Urol 97:331–337 Mellinger GT, 1977, Prognosis of prostatic cancinoma. Recent Results Canc Res 61–72 Bostwick DG, 1994, Gleason grading of prostatic needle biopsies: correlation with grade in 316 matched prostatectomies. Am J Surg Pathol 18:796–803 Bostwick DG, 1994, Grading prostate cancer. Am J Clin Pathol 102/4, Suppl 1):38–56 Mostofi FK, Sesterhenn IA, Davies CJ, 2002, Histological typing of prostate tumours. In: WHO international histological classification of tumours. Second edition. Springer, Berlin, pp 13–16 Epstein JI, Algaba F, Allsbrook WC et al, 2004, Acinar adenocarcinoma in Tumours of the prostate. In: Eble JN, Sauter G, Epstein JI, Sesterhenn IA, eds, WHO classification of tumours of urinary system and male genital organs. IARC, Lyon, pp 180–181 Bonkhoff H, 2005, Gleason grading. Diagnostische Kriterien und klinische Bedeutung. Pathologe 26:422–432 Epstein JI, Chan DW, Sokoll LJ et al, 1998, Nonpalpable stage T1c prostate cancer: prediction of insignificant disease using free/ total prostate specific antigen levels and needle biopsy findings. J Urol 160:2407–2411 Epstein JI, Sanderson H, Carter HB, 2005, Scharfstein DO Utility of saturation biopsy to predict insignificant cancer at radical prostatectomy. Urology 66:356–360 Rodriguez-Urrego PA, Cromin AM, Al-Ahmadie HA et al, 2011, Interobserver and intraobserver reproducibility in digital and routine microscopic assessment of prostate needle biopsies. Hum Patho 42:68–74 Brennan RL, Prediger DJ, 1981, Coefficient kappa: some uses, misuses, and alternatives. Educ Psychol Meas 41:687–699 Randolph JJ, 2005, Free-marginal multirater kappa: an alternative to Fleiss’ fixed-marginal multirater kappa. Paper presented at the Joensuu University Learning and Instruction Symposium 2005, Joensuu, Finland, October 14–15th,, ERIC Document Reproduction Service No. ED490661) Allsbrook WC Jr, Mangold KA, Yang X, Epstein JI, 1999, The Gleason grading system. An review. J Urol Pathol 10:141–157 Allsbrook WC Jr, Mangold KA, Johnson MH et al, 2001, Interobserver reproducibility of Gleason grading of prostatic carcinoma: general pathologist. Hum Pathol 32:81–88 Melia J, Moseley R, Griffiths DFR et al, 2006, A UK-based investigation of inter- and intra-observer reproducibility of Gleason grading of prostatic biopsies. Histopathology 48:644– 654 Griffiths DFR, Melia J, McWilliam LJ et al, 2006, A study of Gleason score interpretation in different groups of UK pathologists; techniques for improving reproducibility. Histopathology 48:655–662 Mikami Y, Manabe T, Epstein JI et al, 2003, Accuracy of Gleason grading by practicing pathologists and impact of education on improving agreement. Human Pathol 34:658–665 Allsbrook WC Jr, Mangold KA, Johnson MH et al, 2001, Interobserver reproducibility of Gleason grading of prostatic carcinoma: urologic pathologists. Hum Pathol 32:74–80 Wittschieber D, Kollermann J, Schlomm T et al., 2010, Nuclear grading versus Gleason grading in small samples containing prostate cancer: a tissue microarray study. Pathol Oncol Res 16:479– 484 Burchardt M, Engers R, Muller M et al, 2008, Interobserver reproducibility by Gleason grading: evaluation using prostate cancer tissue microarrays. J Cancer Res Clin Oncol 134:1071– 1078 Epstein JI, 2010, An update of Gleason grading system. J Urol 183:433–440 Delahunt B, Lambs DS, Srigley JR et al, 2010, Gleason scoring: a comparison of classical and modified, international society of urological pathology, criteria using nadir PSA as a clinical end point. Pathology 42:339–343 Helpap B, Egevad L, 2008, Influence of the modified Gleason grading on pT stage and Gleason score of the prostate carcinoma after radical prostatectomy. Anal Quant Cytol Histol 30:1–7 Helpap B, Egevad L, 2009, Modified Gleason grading. An updated review. Histol Histopathol 24:661–666 Helpap B, Oehler U, 2012, Prostatic carcinoma. The significance of second opinion of histology. Pathologe accepted in press Lau WK, Blute Ml, Bostwick DG et al, 2001, Prognostic factors for survival of patients with pathological Gleason score 7 prostate cancer: differences in outcome between primary Gleason grades 3 and 4. J Urol 166:1692–1 Helpap B, Kollermann J, 2012, Combined histoarchitectural and cytological biopsy grading improves grading accuracy in low grade prostate cancer. Int J Urol submitted Egevad L, Algaba F, Berney D et al, 2011, Interactive digital slides with heat maps: a novel method to improve the reproducibility of Gleason grading. Virchows Arch 459:175–182 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 615 EP 621 DI 10.1007/s12253-011-9484-6 PG 7 ER PT J AU Jarai, T Maasz, G Burian, A Bona, A Jambor, E Gerlinger, I Mark, L AF Jarai, Tamas Maasz, Gabor Burian, Andras Bona, Agnes Jambor, Eva Gerlinger, Imre Mark, Laszlo TI Mass Spectrometry-Based Salivary Proteomics for the Discovery of Head and Neck Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Biomarker discovery; MALDI TOF MS; Saliva; Tumor ID Biomarker discovery; MALDI TOF MS; Saliva; Tumor AB The 5-year survival rates for cases of head and neck squamous cell carcinoma (HNSCC) are only some 60%, mainly because 20%–40% of the patients develop a local relapse in the same or an adjacent anatomic region, even when the surgical margins are histologically tumour-free. Tumours are often discovered in an advanced stage because of the lack of specific symptoms and the diagnostic difficulties. The more advanced the stage of the tumour, the more invasive the diagnostic and treatment interventions needed. An early molecular diagnosis is therefore of vital importance in order to increase the survival rate. The aimof this study was to develop an efficient rapid and sensitive mass spectrometric method for the detection of differentially expressed proteins as tumourspecific biomarkers in saliva from HNSCC patients. Whole saliva samples were collected from patients with HNSCC and from healthy subjects. The proteins were profiled by using SDS PAGE, MALDI TOF/TOF mass spectrometry and the Mascot database search engine. Several potential tumour markers were identified, including annexin A1, beta- and gamma-actin, cytokeratin 4 and 13, zinc finger proteins and P53 pathway proteins. All of these proteins play a proven role in tumour genesis, and have not been detected previously in saliva. Salivary proteomics is a non-invasive specific method for cancer diagnosis and follow-up treatment. It provides facilities for the readily reproducible and reliable detection of tumours in early stages. C1 [Jarai, Tamas] PTE KK, Ful-Orr-Gegeszeti es Fej-Nyaksebeszeti KlinikaPecs, Hungary. [Maasz, Gabor] University of Pecs, Institute of Biochemistry and Medical Chemistry, Szigeti str. 12, 7624 Pecs, Hungary. [Burian, Andras] PTE KK, Ful-Orr-Gegeszeti es Fej-Nyaksebeszeti KlinikaPecs, Hungary. [Bona, Agnes] University of Pecs, Institute of Biochemistry and Medical Chemistry, Szigeti str. 12, 7624 Pecs, Hungary. [Jambor, Eva] University of Pecs, Institute of Biochemistry and Medical Chemistry, Szigeti str. 12, 7624 Pecs, Hungary. [Gerlinger, Imre] PTE KK, Ful-Orr-Gegeszeti es Fej-Nyaksebeszeti KlinikaPecs, Hungary. [Mark, Laszlo] University of Pecs, Institute of Biochemistry and Medical Chemistry, Szigeti str. 12, 7624 Pecs, Hungary. RP Mark, L (reprint author), University of Pecs, Institute of Biochemistry and Medical Chemistry, 7624 Pecs, Hungary. EM laszlo.mark@aok.pte.hu CR Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Argiris A, Karamouzis MV, Raben D et al, 2008, Head and neck cancer. Lancet 371:1695–1709 Mackenzie J, Ah-See K, Thakker N, Sloan P, Maran AG, Birch J et al, 2000, Increasing incidence of oral cancer amongst young persons: what is the aetiology? Oral Oncol 36:387–389 Blot WJ, McLaughlin JK, Winn DM, Austin DF, Greenberg RS, Preston-Martin S et al, 1988, Smoking and drinking in relation to oral and pharyngeal cancer. Cancer Res 48:3282–3287 Ide R, Mizoue T, Fujino Y, Hoshiyama Y, Sakata K, Tamakoshi A et al, 2008, Cigarette smoking, alcohol drinking, and oral and pharyngeal cancer mortality in Japan. Oral Dis 14:314–319 Decker J, Goldstein JC, 1982, Risk factors in head and neck cancer. N Engl J Med 306:1151–1155 Hashibe M, Brennan P, Benhamou S, Castellsague X, Chen C, Curado MP et al, 2007, Alcohol drinking in never users of tobacco, cigarette smoking in never drinkers, and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. J Natl Cancer Inst 99:777–789 D’Souza G, Kreimer AR, Viscidi R, Pawlita M, Fakhry C, Koch WM et al, 2007, Case–control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 356:1944–1956 Wu L et al, 2000, Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J Natl Cancer Inst 92:709–720 Gillison ML, Koch WM, Shah KV, 1999, Human papillomavirus in head and neck squamous cell carcinoma: are some head and neck cancers a sexually transmitted disease? Curr Opin Oncol 11:191–199 Gillison ML, Lowy DR, 2004, A causal role for human papillomavirus in head and neck cancer. Lancet 363:1488–1489 KreimerAR, Alberg AJ, Daniel R,Gravitt PE,Viscidi R, Garrett ES et al, 2004, Oral human papillomavirus infection in adults is associated with sexual behavior and HIV serostatus. J Infect Dis 189:686–698 Kreimer AR, Clifford GM, Boyle P, Franceschi S, 2005, Human papillomavirus types in head and neck squamous cell carcinomas worldwide: a systematic review. Cancer Epidemiol Biomarkers Prev 14:467–475 Mork J, Lie AK, Glattre E, Hallmans G, Jellum E, Koskela P et al, 2001, Human papillomavirus infection as a risk factor for squamouscell carcinoma of the head and neck. N Engl J Med 344:1125–1131 Mendenhall WM, Logan HL, 2009, Human papillomavirus and head and neck cancer. Am J Clin Oncol 32:535–539 Fliss MS, Usadel H, Caballero OL et al, 2000, Facile detection of mitochondrial DNA mutations in tumors and bodily fluids. Science 287:2017–2019 Sidransky D, 1997, Nucleic acid-based methods for the detection of cancer. Science 278:1054–1059 Rosas SL, Koch W, da Costa Carvalho MG et al, 2001, Promoter hypermethylation patterns of p16, O6-methylguanine- DNAmethyltransferase, and death-associated protein kinase in tumors and saliva of head and neck cancer patients. Cancer Res 61:939–942 Righini CA, de Fraipont F, Timsit JF et al, 2007, Tumorspecific methylation in saliva: a promising biomarker for early detection of head and neck cancer recurrence. Clin Cancer Res 13:1179–1185 Franzmann EJ, Reategui EP, Pedroso F et al, 2007, Soluble CD44 is a potential marker for the early detection of head and neck cancer. Cancer Epidemiol Biomarkers Prev 16:1348–1355 Nagler R, Bahar G, Shpitzer T et al, 2006, Concomit antanalysis of salivary tumormarkers–a new diagnostic tool for oral cancer. Clin Cancer Res 12:3979–3984 Tavassoli M, Brunel N, Maher R et al, 1998, p53 antibodies in the saliva of patients with squamous cell carcinoma of the oral cavity. Int J Cancer 78:390–391 Xie H, Onsongo G, Popko J et al, 2008, Proteomics analysis of cells inwhole saliva from oral cancer patients via value-added three-dimensional peptide fractionation and tandem mass spectrometry. Mol Cell Proteomics 7:486–498 Karpova MA, Moshkovskii SA, Toropygin IY, Archakov AI, 2010, Cancer specific MALDI-TOF profiles of blood serum and plasma: biological meaning and perspectives. J Proteomics 73:537–551 Lee KD, Lee HS, Jeon CH, 2011, Body fluid biomarkers for early detection of head and neck squamos cell carcinomas. Anticancer Res 31:1161–116 Baker H, Patel V, Molinolo AA, Shillitoe EJ, Ensley JF, Yoo GH et al, 2005, Proteome-wide analysis of head and neck squamous cell carinomas using laser-capture microdissecton and tandem mass spectrometry. Oral Oncol 41:183–199 El-Naggar AK, Mao L, Staekel G et al, 2001, Genetic heterogeneity in saliva from patients with oral squamous carcinomas: implications in molecular diagnosis and screening. J Mol Diagn 3:164–170 Hu S, Arellano M, Boontheung P,Wang J, Zhou H, Jiang J, Elashoff D, Wei R, Loo JA, Wong DT, 2008, Salivary proteomics for oral cancer biomarker discovery. Clin Cancer Res 14:6246–6252 Aizenbud D, Peri-Front Y, Nagler RM, 2008, Salivary analysis and antioxidants in cleft lip and palate children. Arch Oral Biol 53:517–522 Navazesh M, 1993, Methods for collecting saliva. Ann N YAcad Sci 694:72–77 Laemmli UK, 1970, Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227:680–685 Gaal V, Mark L, Kiss P, Kustos I, Tamas A, Kocsis B, Lubics A, Nemeth V, Nemeth A, Lujber L, Pytel J, Toth G, Reglodi D, 2008, Investigation of the effects of PACAP on the composition of tear and endolymph proteins. J Mol Neurosci 36:321–329 Baak JP, Path FR, Hermsen MA, Meijer G, Schmidt J, Janssen EA, 2003, Genomics and proteomics in cancer. Eur J Cancer 39:1199– 1215 Dowling P,Wormald R, Meleady P, Henry M, Curran A, Clynes M, 2008, Analysis of the saliva proteome from patients with head and nack squamos cell carcinoma reveals differenes in abundance levels of proteins associated with tumour progression and metastasis. J Proteomics 71:168–175 Mahfouz ME, Rodrigo JP, Takes RP, Elsheikh MN, Rinaldo A, Brakenhoff RH, Ferlito A, 2010, Current potential and limitations of molecular diagnostic methods in head and neck cancer. Eur Arch Otorhinolaryngol 267:851–860 Mydlarz WK, Hennessey PT, Califano JA, 2010, Advances and perspectives in the molecular diagnosis of head and neck cancer. Expert Opin Med Diag 4:53–65 Xia SH, Hu LP, Hu H, Ying WT, Xu X, Cai Y, Han YL, Chen BS, Wei F, Qian XH, Cai YY, Shen Y, Wu M, Wang MR, 2002, Three isoforms of annexin I are preferentially expressed in normal esophageal epithelia but down-regulated in esophageal squamous cell carcinomas. Oncogene 21:6641–6648 Schaaij-Visser TB, Bremmer JF, Braakhuis BJ, Heck AJ, Slijper M, van der Waal I, Brakenhoff RH, 2010, Evaluation of cornulin, keratin 4, keratin 13 expression and grade of dysplasia for predicting malignant progression of oral leukoplakia. Oral Oncol 46:123– 127 Matsumoto I, Yamamoto M, 2005, Differential expression of the keratin-4, -13,-14, -17 and transglutaminase 3 genes during the development of oral squamous cell carcinoma from leukoplakia. Oral Oncol 41:607–613 Pedrero JMG, Fernandez MP, Morgan RO, Zapatero AH, Gonzalez MV, Nieto CS, Rodrigo JP, 2004, Annexin A1 down-regulation in head and neck cancer is associated with epithelial differentiation status. Am J Pathol 164:73–79 Raynal P, Pollard HB, 1994, Annexins: the problem of assessing the biological role for a gene family of multifunctional calciumand- phosospholipid-binding proteins. Biochim Biophys Acta 1197:63–64 Ohkura S, Kondoh N, Hada A, Arai M, Yamazaki Y, Sindoh M, Takahashi M, Mussunoor S, Murray GI, 2008, The role of annexins in tumour development and progression. J Pathol 216:131–140 Lin CY, Jeng YM, Chou HY, Hsu HC, Yuan RH, Chiang CP et al, 2008, Nuclear localization of annexin A1 is a prognostic factor in oral squamous cell carcinoma. J Surg Oncol 97:544–550 Lo WY, Tsai MH, Tsai Y, Hua CH, Tsai FJ, Huang SY, Tsai CH, Lai CC, 2007, Identification of overexpressed proteins in oral squamous cell carcinoma, OSCC, patients by clinical proteomic analysis. Clin Chim Acta 376:101–107 Makridakis M, Vlahou A, 2010, Secretome proteomics for discovery of cancer biomarkers. J Proteomics 73:2291–2305 Schaaij-Visser T, Brakenhoff RH, Leemans CR, Heck AJR, Slijper M, 2010, Protein biomarker discovery for head and neck cancer. J Proteomics 73:1790–1803 Schaaij-Visser TB, Graveland AP, Gauci S, Braakhuis BJ, Buijze M, Heck AJ et al, 2009, Differential proteomics identifies protein biomarkers that predict local relapse of head and neck squamous cell carcinomas. Clin Cancer Res 15:7666–7675 Wadsworth JD, Somers K, Stack BC, Cazare L, Malik G, Adam BL, Wright GL, Semmes JO, 2004, Identification of patients with head and neck cancer using serum protein profiles. Arch Otolaryngol Head Neck Surg 130:98–104 Wu W, Tang X, Hu W, Lotan R, Hong WK, Mao L, 2002, Identification and validation of metastasis-associated proteins in head and neck cancer cell lines by two-dimensional electrophoresis and mass spectrometry. Clin Exp Metastasis 19:319–326 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 623 EP 628 DI 10.1007/s12253-011-9486-4 PG 6 ER PT J AU Cui, Q Li, D Zhang, J Wang, X Liu, Sh Wang, L Zhang, P Zhou, J Liu, Ch Jiang, W Zeng, Y AF Cui, Qiu Li, Dingfeng Zhang, Jing Wang, Xiaohong Liu, Shubin Wang, Lei Zhang, Ping Zhou, Ju Liu, Cheng Jiang, Weihao Zeng, Yanjun TI The Significance of Preoperative Chemotherapy in Evaluation of Recurrent Soft Tissue Liposarcoma Necrosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Implantable intra-arterial induction chemotherapy; Liposarcoma; Cisplatin; Doxorubicin; Caffeine ID Implantable intra-arterial induction chemotherapy; Liposarcoma; Cisplatin; Doxorubicin; Caffeine AB To investigate the effect of preoperative induction chemotherapy on treatment of recurrent liposarcoma. 21 patients with recurrent liposarcoma received the treatment of preoperative intra-arterial chemotherapy and surgical resection. Intra-arterial chemotherapy was given by subcutaneous implantable drug delivery system with infusion of cisplatin and doxorubicin followed by caffeine. After treatment, patients were followed up for 39 months. The liposarcoma changes in CT imaging were observed in 18 cases and there were 15 cases with medium or severe pathological changes caused by chemotherapy. At the end of the postoperative follow-up of 39 months, liposarcoma reoccurred locally in 2 cases; pulmonary metastasis occurred in 1 case and death in 3 cases. Preoperative intra-arterial chemotherapy is effective for highly malignant tumors such as recurrent liposarcoma and the judgment of prognosis is based on the postoperative pathological changes of such tumor. C1 [Cui, Qiu] 307th Hospital of PLA, Department of Bone Tumor, 100071 Beijing, China. [Li, Dingfeng] 307th Hospital of PLA, Department of Bone Tumor, 100071 Beijing, China. [Zhang, Jing] Beijing University of Technology, Biomedical Engineering Center, 100022 Beijing, China. [Wang, Xiaohong] Beijing University of Technology, Biomedical Engineering Center, 100022 Beijing, China. [Liu, Shubin] 307th Hospital of PLA, Department of Bone Tumor, 100071 Beijing, China. [Wang, Lei] 307th Hospital of PLA, Department of Bone Tumor, 100071 Beijing, China. [Zhang, Ping] 307th Hospital of PLA, Department of Bone Tumor, 100071 Beijing, China. [Zhou, Ju] 307th Hospital of PLA, Department of Bone Tumor, 100071 Beijing, China. [Liu, Cheng] 307th Hospital of PLA, Department of Bone Tumor, 100071 Beijing, China. [Jiang, Weihao] 307th Hospital of PLA, Department of Bone Tumor, 100071 Beijing, China. [Zeng, Yanjun] Beijing University of Technology, Biomedical Engineering Center, 100022 Beijing, China. RP Li, D (reprint author), 307th Hospital of PLA, Department of Bone Tumor, 100071 Beijing, China. EM 307yygk@sina.com CR Enzinger FM, Weiss SW, 1988, Soft tissue tumors. Mosby St. Louis 346–382 Mentzel T, Fletcher CD, 1995, Lipomatous tumours of soft tissues: an update. Virchows Arch 427:353–363 Oliveira AM, Nascimento AG, 2001, Pleomorphic liposarcoma. Semin Diagn Pathol 18:274–285 Gebhard S, Coindre JM, Michels JJ, Terrier P, Bertrand G, Trassard M, Taylor S, Chateau MC, Marques B, Picot V, Guillou L, 2002, Pleomorphic liposarcoma: clinicopathologic, immunohistochemical, and follow-up analysis of 63 cases: a study from the French Federation of Cancer Centers Sarcoma Group. Am J Surg Pathol 26:601–616 Zhang RM, Liu YW, 2003, The chemotherapy of soft tissue sarcoma. Chin J Bone Tumor Bone Dis 2(3):181–185 Therasse P, Arbuck SG, Eisenhauer EA et al, 2000, New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst 92(3):205–216 Rosen G, Caparros B, Huvos AG et al, 1982, Preoperative chemotherapy for osteogenic sarcoma: selection of Postoperativeadjuvant chemotherapy based on the response of the primary tumor to preoperative chemotherapy. J Cancer 49(6):1221–1230 DeLaney TF, Spiro IJ, Suit HD et al, 2003, Neoadjuvant chemotherapy and radiotherapy for large extremity soft tissue sarcomas. Int J Radiat Oncol Biol Phys 56:1117–1127 Said Basly M, Khouni H, Dridi M, 2009, Retroperitoneal liposarcoma. Tunis Med 87(8):552–554 Chung PW, Deheshi BM, Ferguson PC, 2009, Radiosensitivity translates into excellent local control in extremity myxoid liposarcoma: a comparison with other soft tissue sarcomas. Cancer 115(14):3254–3261 Shaerf DA, Mann B, Alorjani M et al, 2011, High-grade intraarticular liposarcoma of the knee. Skeletal Radiol 40(3):363–365 Illuminati G, Ceccanei G, Pacile MA et al, 2010, Surgical outcomes for liposarcoma of the lower limbs with synchronous pulmonary metastases. J Surg Oncol 102(7):827–831 Han HH, Choi KH, Kim DS et al, 2010, Retroperitoneal giant liposarcoma. Korean J Urol 51(8):579–582 Ozaki R, Hamada K, Emori M et al, 2010, Limb salvage operation using intraoperative extracorporeal autogenous irradiated bone and tendon graft for myxoid liposarcoma on dorsum of foot. Foot, Edinb, 20(2–3):90–95 Endo M, 2010, Musculoskeletal tumor. III. Chemotherapy of liposarcoma– the current status of chemotherapy and new findings in clinical trials of novel drugs. Gan to kagaku ryoho. Cancer & chemotherapy 37(3):434–438 Casali PG, Blay JY, 2010, ESMO/CONTICANET/EUROBONET Consensus Panel of experts. Soft tissue sarcomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 21(Suppl 5):v198–v203 Picci P, 2000, Adjuvant chemotherapy for extremity soft-tissue sarcomas in adults. Curr Oncol Rep 2:502–507 Patel SR, Burgess MA, Plager C, Papadopoulos NE, Linke KA, Benjamin RS, 1994, Myxoid liposarcoma. Experience with chemotherapy. Cancer 74(4):1265–1269 Mankin HJ, Hornicek FJ, 2005, Diagnosis, classification, and management of soft tissue sarcomas. Cancer Control 12(1):5–21 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 629 EP 633 DI 10.1007/s12253-011-9487-3 PG 5 ER PT J AU Guo, X Xiong, L Zou, L Zhao, J AF Guo, Xiaodong Xiong, Lu Zou, Lin Zhao, Jingmin TI Upregulation of Bone Morphogenetic Protein 4 is Associated with Poor Prognosis in Patients with Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Bone morphogenetic protein 4; Prognosis ID Hepatocellular carcinoma; Bone morphogenetic protein 4; Prognosis AB Bone morphogenetic protein (BMP) 4 plays a crucial role in tumor invasion and metastasis of various human cancers. However, little is known about the correlation of BMP4 expression with clinical aggressiveness and prognosis in hepatocellular carcinoma (HCC). The aim of this study was to investigate the expression of BMP4 in HCC and determine its correlation with tumor progression and prognosis. Immunohistochemistry assay was used to determine the expression of BMP4 in HCC and corresponding paracarcinomatous tissues from 156 patients. The potential prognostic value of BMP4 was investigated by comparing the survival rates between the BMP4-positive and BMP4-negative HCC patients. Immunohistochemically, BMP4 protein expression in the HCC tissues (120/156, 76.9%) was significantly higher than that in the paracarcinomatous tissues (19/156, 12.2%, P<0.01). The expression of BMP4 in HCC was associated with number of tumor nodules (P00.02), Edmondson grade (P00.03), TNM stage (P00.009), and vascular invasion (>P00.006). In univariate survival analysis, the significant associations of the BMP4 protein overexpression with shortened patients’ overall and disease-free survival were found (P00.001 and 0.006, respectively). Furthermore, its expression was found to be an independent factor for predicting both overall (P00.009) and disease-free survival (P00.022) of HCC in multivariate analysis. Our data suggest for the first time that BMP4 is overexpressed in HCC tissues and may also act as a novel marker for predicting the recurrence and prognosis of HCC patients after surgery. C1 [Guo, Xiaodong] Chinese PLA Postgraduate Medical School, 100853 Beijing, China. [Xiong, Lu] 302 Hospital of PLA, 100039 Beijing, China. [Zou, Lin] Chinese PLA General Hospital, 100853 Beijing, China. [Zhao, Jingmin] 302 Hospital of PLA, 100039 Beijing, China. RP Zhao, J (reprint author), 302 Hospital of PLA, 100039 Beijing, China. EM zhaojingmin302@163.com CR Yang H, Lin M, Xiong F, Yang Y, Nie X, McNutt MA, Zhou R, 2011, Combined lysosomal protein transmembrane 4 beta-35 and argininosuccinate synthetase expression predicts clinical outcome in hepatocellular carcinoma patients. Surg Today 41:810–817 Farazi PA, DePinho RA, 2006, Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer 6:674–687 Thorgeirsson SS, Grisham JW, 2002, Molecular pathogenesis of human hepatocellular carcinoma. Nat Genet 31:339–346 Chen D, Zhao M, Mundy GR, 2004, Bone morphogenetic proteins. Growth Factors 22:233–241 Simic P, Vukicevic S, 2005, Bone morphogenetic proteins in development and homeostasis of kidney. Cytokine Growth Factor Rev 16:299–308 Marki D, Celi T, Grkovi A, Spanjol J, Fukar Z, Grahovac B, Ethorevi G, Bobinac D, 2011, mRNA expression of bone morphogenetic proteins and their receptors in human renal cell carcinoma. Urol Int In press Yuen HF, Chan YP, Cheung WL, Wong YC, Wang X, Chan KW, 2008, The prognostic significance of BMP-6 signaling in prostate cancer. Mod Pathol 21:1436–1443 Motoyama K, Tanaka F, Kosaka Y, Mimori K, Uetake H, Inoue H, Sugihara K, Mori M, 2008, Clinical significance of BMP7 in human colorectal cancer. Ann Surg Oncol 15:1530–1537 Hogan BL, 1996, Bone morphogenetic proteins in development. Curr Opin Genet Dev 6:432–438 Virtanen S, Alarmo EL, Sandstrom S, Ampuja M, Kallioniemi A, 2011, Bone morphogenetic protein −4 and −5 in pancreatic cancer– novel bidirectional players. Exp Cell Res 317:2136–2146 McLean K, Gong Y, Choi Y, Deng N, Yang K, Bai S, Cabrera L, Keller E, McCauley L, Cho KR, Buckanovich RJ, 2011, Human ovarian carcinoma–associated mesenchymal stem cells regulate cancer stem cells and tumorigenesis via altered BMP production. J Clin Invest 121:3206–3219 Slattery ML, Lundgreen A, Herrick JS, Kadlubar S, Caan BJ, Potter JD, Wolff RK, 2011, Genetic variation in bone morphogenetic protein and colon and rectal cancer. Int J Cancer In press Qiao B, Johnson NW, Chen X, Li R, Tao Q, Gao J, 2011, Disclosure of a stem cell phenotype in an oral squamous cell carcinoma cell line induced by BMP-4 via an epithelial-mesenchymal transition. Oncol Rep 26:455–461 Ketolainen JM, Alarmo EL, Tuominen VJ, Kallioniemi A, 2010, Parallel inhibition of cell growth and induction of cell migration and invasion in breast cancer cells by bone morphogenetic protein 4. Breast Cancer Res Treat 124:377–386 Shon SK, Kim A, Kim JY, Kim KI, Yang Y, Lim JS, 2009, Bone morphogenetic protein-4 induced by NDRG2 expression inhibits MMP-9 activity in breast cancer cells. Biochem Biophys Res Commun 385:198–203 Hjertner O, Hjorth-Hansen H, Borset M, Seidel C, Waage A, Sundan A, 2001, Bone morphogenetic protein-4 inhibits proliferation and induces apoptosis of multiple myeloma cells. Blood 97:516–522 Maegdefrau U, Arndt S, Kivorski G, Hellerbrand C, Bosserhoff AK, 2011, Downregulation of hemojuvelin prevents inhibitory effects of bone morphogenetic proteins on iron metabolism in hepatocellular carcinoma. Lab Invest In press Ishak KG, Anthony PP, Sobin LH, 1994, Nonepithelial tumors. In: Ishak KG, ed, Histological typing of tumors of the liver. World Health Organization International Classification of Tumors. Springer, Berlin, pp 22–27 Wittekind C, 2006, Pitfalls in the classification of liver tumors. Pathologe 27:289–293 Gao Q, Qiu SJ, Fan J, Zhou J, Wang XY, Xiao YS, 2007, Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J Clin Oncol 25:2586–2593 Zhu XD, Zhang JB, Zhuang PY, Zhu HG, Zhang W, Xiong YQ, 2008, High expression of macrophage colony-stimulating factor in peritumoral liver tissue is associated with poor survival after curative resection of hepatocellular carcinoma. J Clin Oncol 26:2707–2716 Shen H, Huang GJ, Gong YW, 2003, Effect of transforming growth factor beta and bone morphogenetic proteins on rat hepatic stellate cell proliferation and trans-differentiation. World J Gastroenterol 9:784–787 Slattery ML, Lundgreen A, Herrick JS, Wolff RK, Caan BJ, 2011, Genetic variation in the transforming growth factor-β signaling pathway and survival after diagnosis with colon and rectal cancer. Cancer 117:4175–4183 Lombardo Y, Scopelliti A, Cammareri P, Todaro M, Iovino F, Ricci-Vitiani L, Gulotta G, Dieli F, de Maria R, Stassi G, 2011, Bone morphogenetic protein 4 induces differentiation of colorectal cancer stem cells and increases their response to chemotherapy in mice. Gastroenterology 140:297–309 LeeYC, ChengCJ, BilenMA, Lu JF, Satcher RL,Yu-Lee LY,Gallick GE,Maity SN, Lin SH(2011, BMP4 promotes prostate tumor growth in bone through osteogenesis. Cancer Res 71:5194–5203 Capasso M, Ayala F, Russo R, Avvisati RA, Asci R, Iolascon A, 2009, A predicted functional single-nucleotide polymorphism of bone morphogenetic protein-4 gene affects mRNA expression and shows a significant associationwith cutaneousmelanoma in Southern Italian population. J Cancer Res Clin Oncol 135:1799–1807 Deng H, Ravikumar TS, Yang WL, 2007, Bone morphogenetic protein-4 inhibits heat-induced apoptosis by modulating MAPK pathways in human colon cancer HCT116 cells. Cancer Lett 256:207–217 Hamada S, Satoh K, Hirota M, Fujibuchi W, Kanno A, Umino J, Ito H, Satoh A, Kikuta K, Kume K, Masamune A, Shimosegawa T, 2009, Expression of the calcium-binding protein S100P is regulated by bone morphogenetic protein in pancreatic duct epithelial cell lines. Cancer Sci 100:103–110 Piccirillo SG, Reynolds BA, Zanetti N, Lamorte G, Binda E, Broggi G, Brem H, Olivi A, Dimeco F, Vescovi AL, 2006, Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature 444:761–765 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 635 EP 640 DI 10.1007/s12253-011-9488-2 PG 6 ER PT J AU Katz, S Balogh, P Nagy, N Kiss, LA AF Katz, Sandor Balogh, Petra Nagy, Nandor Kiss, L Anna TI Epithelial-To-Mesenchymal Transition Induced by Freund’s Adjuvant Treatment in Rat Mesothelial Cells: A Morphological and Immunocytochemical Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Caveolin-1/caveolae; Epithelial-tomesenchymal transition; Mesothelial cell; Transdifferentiation ID Caveolin-1/caveolae; Epithelial-tomesenchymal transition; Mesothelial cell; Transdifferentiation AB Intraperitoneal injection of Freund’s adjuvant induces acute peritonitis. By the time of the Freund’s adjuvant treatment the flat, simple squamous epithelial cells became rounded, cuboidal shaped, many of them have lost their connection with the neighbouring cells and detached from the basement membrane. The macrophage markers’ (ED1, OX43 and CD68) expression also increased in the mesothelial cells and more mesothelin and anti-ED1 doublelabelled cells were found freely present close to the surface. The cytokeratin expression of the mesothelial cells has gradually decreased. At the 5th day of the inflammation practically there was no cytokeratin labelling present in the mesothelial cells and the mesothelin expression has significantly decreased. Parallel to this mesothelial cells started to express vimentin, a characteristic mesenchymal intermediate filament protein indicating that they gradually lost their epithelial character and gained mesenchymal phenotype. These results strongly suggest that under the effect of Freund’s adjuvant treatment (inflammation) mesothelial cells can undergo epithelial-to-mesenchymal transition and differentiate into phagocytotic (macrophage-like) cells. Studying the caveolae/caveolin-1 on the plasma membrane of mesothelial cells we found that the Freund’s adjuvant treatment has changed the cellular distribution of caveolin-1: as the inflammation progressed strong caveolin-1 labelling was found inside of the cytoplasm(in perinuclear localization) indicating that inflammation induced the caveolae internalization. These results indicate that caveolae/caveolin-1 might play important regulatory role in signal transduction leading to trasdifferentiation. C1 [Katz, Sandor] Semmelweis University, 1st Department of Anatomy, Tuzolto u. 58Budapest, Hungary. [Balogh, Petra] Semmelweis University, 1st Department of Anatomy, Tuzolto u. 58Budapest, Hungary. [Nagy, Nandor] Semmelweis University, 1st Department of Anatomy, Tuzolto u. 58Budapest, Hungary. [Kiss, L Anna] Semmelweis University, 1st Department of Anatomy, Tuzolto u. 58Budapest, Hungary. RP Katz, S (reprint author), Semmelweis University, 1st Department of Anatomy, Budapest, Hungary. EM katzsanya@gmail.com CR Savagner P, 2001, Leaving the neighbourhood: Molecular mechanisms involved during epithelial-mesenchymal transition. Bioesssay 23:912–923 Camenisch TD, Molin DG, Person A, Runyan RB, Gittenberger-de Groot AC, McDonald JA, Klewer SE, 2002, Temporal and distinct TGFß ligand requirements during mouse and avian endocardial cushon morphogenesis. Dev Biol 248:170–181 Piek E, Moustakas A, Kurisaki A, Heldin CH, ten Dijke P, 1999, TGF-(beta, type I receptor/Alk-5 and Smad proteins mediate epithelial to mesenchymal transdifferentiation in NMuMG breast epithelial cells. J Cell Sci 112(Suppl):4557–4568 Moustakas A, Pardali K, Gaal A, Heldin CH, 2002, Mechanisms of TGF-beta signalling in regulation of cell growth and differentiation. Immunol Lett 82:85–91 Kredict RT, 1999, The peritoneal membrane in chronic peritoneal dialysis patients. Kidney Int 55:341–356 Boyer B, Valles AM, Edme N, 2000, Induction and regulation of epithelial-mesenchymal transitions. Biochem Pharmacol 60:1091– 1099 Yang J, Liu Y, 2001, Dissection of key events in tubular epithelial to myofibroblast transition and its implication in renal interstitial fibrosis. Am J Pathol 159:1465–1475 Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA, 2002, Myofibroblasts and mechanoregulation of connective tissue remodelling. Nat Rev Mol Cell Biol 3:349–363 Katz S, Balogh P, Kiss AL, 2011, Mesothelial cells can detach from the mesentery and differentiate into macrophage-like cells. APMIS 119:782–793 Dijkstra CD, Dopp EA, Joling P, Kraal G, 1985, The heterogeneity of mononuclear phagocytes in lymphoid organs: distinct macrophages subpopulation in the rat recognized by monoclonal antibodies ED1, ED2 and ED3. Immunology 54:589–599 Wiese C, Rollentschek A, Kania G, Blyszczuk P, Tarasova KV, Wersto RP, Boheler KR, Wobus AM, 2004, Nestin expression—a property of multi-lineage progenitor cells? Cell Mol Life Sci 61:2510–2522 Yanez-Mo M, Lara-Pezzi E, Selgas R, Ramirez-Huesca M et al, 2003, Peritoneal dialysis and epithelial-to-mesenchymal transition of mesothelial cells. New England J Medicine 348:403–413 von Ruhland CJ, Campbell L, Gumbleton M, Jasani B, Newman G, 2004, Immunolocalization of caveolin-1 in rat and human mesothelium. JHistochem and Cytochem 52:1415–1425 Bishop AL, Hall A, 2000, Rho GTPases and their effector proteins. Biochem J 348:241–255 Braga VM, Machesky LM, Hall A, Hotchin NA, 1997, The small GTPases Rho and Rac are required for establishment of cadherin-dependent cell-cell contacts. J Cell Biol 137:1421– 1431 Hordjik PL, Klooster JP, van der Kammen RA, Michiels F, Oomen LC, Collard JG, 1997, Inhibition of invasion of epithelial cells by Tiam-1-Rac signalling. Science 278:1464–1466 Bakin AV, Rinehart C, Tomlinson AK, Arteaga CL, 2002, p38 mitogen-activated protein kinase is required for TGFß-mediated fibroblastic transdifferentiation and cell migration. J Cell Science 115:3193–3206 Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga CL, 2000, Phosphatidyl inositol 3-kinase function is required for TGFbeta-mediated epithelial to mesenchymal transition and cell migration. J Biol Chem 275:36803–36810 Patel HH, Murray F, Insel PA, 2008, Caveolae as organizers of pharmacologically relevant signa transduction molecules. Ann Rev Pharmacol Toxicol 48:359–391 Pelkmans L, Helenius A, 2002, Endocytosis via caveolae. Traffic 3:311–320 Krajewska WM, Maslowska I, 2004, Caveolins: structure and function in signal transduction. Cell Mol Biol Lett 9:195–220 Razani B, Zhang ZL, Bitzer M, von Gersdorff G, Bottinger EP, Lisanti MP, 2001, Caveolin-1 regulates transforming growth factor, TGF)-ß-SMAD signalling through an interaction with the TGF-ß type I receptor. J Biol Chem 276:6727–6738 Zwaagstra JC, El-Alfy M, O’Connor-McCourt MD, 2001, Transforming growth factor, TGF)-ß 1 internalization: modulation by ligand interaction with TGF-ß receptors types I and II and a mechanism that is distinct from clathrin-mediated endocytosis. J Biol Chem 276:27237–27245 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 641 EP 649 DI 10.1007/s12253-011-9489-1 PG 9 ER PT J AU Ebrahimkhani, S Asgharian, MA Nourinaier, B Ebrahimkhani, K Vali, N Abbasi, F Zali, RM AF Ebrahimkhani, Saeideh Asgharian, Mohammad Ali Nourinaier, Babak Ebrahimkhani, Khadijeh Vali, Nasrin Abbasi, Fatemeh Zali, Reza Mohammad TI Association of GSTM1, GSTT1, GSTP1 and CYP2E1 Single Nucleotide Polymorphisms with Colorectal Cancer in Iran SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glutathione S-transferases; Colorectal cancer; Cytochrome P4502E1; Iran ID Glutathione S-transferases; Colorectal cancer; Cytochrome P4502E1; Iran AB Colorectal cancer is a major cause of morbidity and mortality both globally and in Iran. The aim of this study was to determine the association between genetic polymorphisms of glutathione S-transferases P1, M1 and T1 (GSTP1, M1, T1) and susceptibility to colorectal cancer (CRC). Genotyping of GSTP1, GSTM1 and GSTT1 was performed by the use of pyrosequencing. One hundred cases and healthy controls were enrolled into this study. Mean GSTT1 polymorphism type was significantly (P<0.01) higher in cases as compared to controls (P<0.0001: OR, 2.43: 95% CI, 1.47-4). On the other hand there is no significant association between GSTM1, GSTP1 and colorectal cancer. GSTs measurement may be useful as a colorectal marker in colorectal cancer and biopsies obtained at colonoscopy can be used to measure tumor markers. C1 [Ebrahimkhani, Saeideh] Shahid Beheshti University of Medical Sciences, Research Center for Gastroenterology and Liver Disease, Taleghani st- EvinTehran, Iran. [Asgharian, Mohammad Ali] Islamic Azad University, Department of Cell and Molecular Biology, Tonekabon BranchTonekabon, Iran. [Nourinaier, Babak] Shahid Beheshti University of Medical Sciences, Research Center for Gastroenterology and Liver Disease, Taleghani st- EvinTehran, Iran. [Ebrahimkhani, Khadijeh] Zanjan University of Medical ScienceZanjan, Iran. [Vali, Nasrin] Shahid Beheshti University of Medical Sciences, Research Center for Gastroenterology and Liver Disease, Taleghani st- EvinTehran, Iran. [Abbasi, Fatemeh] Azarbaijan Tarbiat Molaem University, Department of Cell and Molecular BiologyTonekabon, Iran. [Zali, Reza Mohammad] Shahid Beheshti University of Medical Sciences, Research Center for Gastroenterology and Liver Disease, Taleghani st- EvinTehran, Iran. RP Zali, RM (reprint author), Shahid Beheshti University of Medical Sciences, Research Center for Gastroenterology and Liver Disease, Tehran, Iran. EM mrzali55@yahoo.com CR Guengerich FP, 1992, Metabolic activation of carcinogens. Pharmacol Ther 54:17–61 Guengerich FP, Shimada T, 1991, Oxidation of toxic and carcinogenic chemicals by human cytochrome P- 450 enzymes. Chem Res Toxicol 4:391–407 Doll R, Peto R, 1981, The causes of cancers: quantitative estimates of avoidable risks of cancers in the United States today. J Natl Cancer Inst 66(6):1191–1308 Chasseaud LF, 1979, The role of glutathione and glutathione S_transferases in the metabolism of chemical carcinogens and other electrophiles agents. Adv Cancer Res 29:175–274 Coles B, Ketterer B, 1990, The role of glutathione transferases in chemical carcinogenesis. Crit Rev Biochem MolBiol 25:47–70 Mucci LA, Wedren S, Tamimi RM, Trichopoulos D, Adami HO, 2001, The role of gene-environment interaction in the aetiology of human cancer: examples from cancers of the large bowel, lung and breast. J Intern Med 249:477–493 Kihara M, Kihara M, Noda K, 1995, Risk of smoking for squamous and small cell carcinomas of the lung modulated by combinations of CYP1A1 and GSTM1 gene polymorphisms in a Japanese population. Carcinogenesis 16:2331–2336 Lin DX, Tang YM, Peng Q, Lu SH, Ambrosone CB, Kadlubar FF, 1998, Susceptibility to esophageal cancer and genetic polymorphisms in glutathione s- transferases T1, P1, and M1 and cytochrome P450 2E1. Cancer Epidemiology, Biomarker & Prey 7:1013–1018 Nebert DW, McKinnon RA, Puga A, 1996, Human drug- metabolizing enzyme polymorphisms: effects on risk of toxicity and cancer. DNA Cell Biol 15:273–280 Cheng KK, Day NE, Duffy SW, Lam TH, Fok M, Wong J, 1992, Pickled vegetables in the aetiology of esophageal cancer in Hong Kong Chinese. Lancet 339:1314–1318 Hecht SS, 1998, Biochemistry, biology, and carcinogenicity of tobacco-specific N-nitrosamines. Chem Res Toxicol 11(6):559–603 Guengerich FP, Kim DH, Iwasaki M, 1991, Role of human cytochrome P-450IIE1 in the oxidation of many low molecular weight cancer suspects. Chem Res Toxicol 4:168–179 Gunter MJ,Watson MA, Loktionov AS et al, 2005, No association between cytochrome P450 and glutathione S-transferase gene polymorphisms and risk of colorectal adenoma: results from the UK flexible sigmoidoscopy screening trial. Canc Epidemiol Biomarkers Prev 14(4):1028–30 Nakajima T, Wang R-S, Nimura Y et al, 1996, Expression of cytochrome P450s and glutathione S- transferases in human esophagus with squamous- cell carcinomas. Carcinogenesis 17:1477–1481 Boyer TD, Kenney WC, 1985, Preparation, characterization and properties of glutathione S-transferases. In: Zakim D, Vessey D, eds, Biochemical pharmacology and toxicology.Wiley, New York Mannervik B, Awasthi YC, Board PG et al, 1992, Nomenclature for human glutathione S-transferases. Biochem J 282:305–306 Meyer DJ, Coles B, Pemble SE, Gilmore KS, Fraser GM, Ketterer B, 1991, θ, a new class of glutathione transferases purified from rat and man. Biochem J 274:409–414 Lin D, Meyer DJ, Ketterer B, Lang NP, Kadlubar FF, 1994, Effects of human and rat glutathione S-transferases on the covalent bonding of the N-acetoxy derivatives of heterocyclic amine carcinogens in vitro: a possible mechanism of organ specificity in their carcinogenesis. Cancer Res 54:4920–4926 Pemble S, Schroeder KR, Spenser SR et al, 1994, Human glutathione S-transferase, GSTT1): cDNA cloning and the characterization of a genetic polymorphism. Biochem J 300:271–276 EvansWE, RellingMV(1999, Pharmacogenomics: translating functional genomics into rational therapeutics. Science 286:487–91 Rebbeck TR, 1997, Molecular epidemiology of the human glutathione S-transferase genotypes GSTM1 and GSTT1 in cancer susceptibility. Cancer Epidemiol Biomark Prev 6:733–743 Strange RC, Lear JT, Fryer AA, 1998, Glutathione S-transferase polymorphisms: influence on susceptibility to cancer. Chem Biol Interact 24(111–112):351–364 Nomania H, Mohammadzadeh Ghobadloob S, Yaghmaeib B, Rezvaniea NA, Yaghmaeic K, 2005, Glutathione S-transferases activity in patients with colorectal cancer. Clin Biochem 38:621–624 Rajagopal R, Deakin M, Fawole AS et al, 2005, Glutathione Stransferase T1 polymorphisms are associated with outcome in colorectal cancer. Carcinogenesis 26(12):2157–2163 Srivastava SK, Singhal SS, Hu X, Awasthi YC, Zimniak P, Singh SV, 1999, Differential catalytic efficiency of allelic variants of human glutathione S-transferase Pi in catalyzing the glutathione conjugation of thiotepa. Arch Biochem Biophys 366:89–94 Houlston RS, 1999, Glutathione S-transferase M1 status and lung cancer risk: a meta-analysis. Cancer Epidemiol Biomark Prev 8:675–682 Baily R, Roodi N, Verrier CS, Yee CJ, Dupont WD, Parl F, 1998, Breast cancer and CYP1A1, GSTM1, and GSTT1 polymorphisms: evidence of a lack of association in Caucasians and African Americans. Cancer Res 58:65–70 Saadat I, Saadat M, 2001, Glutathione S-transferase M1 and T1 null genotypes and the risk of gastric and colorectal cancers. Cancer Letters 169:21–26 de Jong M, Nolte IM, te Meerman GJ et al, 2002, Low-penetrance genes and their involvement in colorectal cancer susceptibility. Cancer Epidemiol Biomarkers Prev 11:1332–52 Board PG, 1981, Gene deletion and partial deficiency of the glutathione S-transferase, ligandin, system in man. FEBS Lett 135:12–14 Pemble S, Schroeder KR, Spencer SS, Meyer DJ, Hallier E, Bolt HM, 1994, Human glutathione S-transferase, GSTT1): cDNA cloning and the characterisation of a genetic polymorphism. Biochem J 300:271–276 Ali-Osman F, Akande O, Antoun G, Mao JX, Buolamwini J, 1997, Molecular cloning, characterisation and expression in Escherichia coli of full-length cDNAs of three human glutathione S-transferase π variants. Evidence for differential catalytic activity of the encoded proteins. J Biol Chem 272:10004–11112 Casson AG, Zheng Z, Chiasson D et al, 2003, Associations between genetic polymorphisms of Phase I and II metabolizing enzymes, p53 and susceptibility to esophageal adenocarcinoma. Cancer Detect Prev 27:139–146 Tan W, Song N, Wang GQ et al, 2000, Impact of genetic polymorphisms in cytochrome P450 2E1 and glutathione S-transferases M1, T1, and P1 on susceptibility to esophageal cancer among high-risk individuals in China. Cancer Epidemiology Biomarkers Prev 9:551–556 Butler WJ, Ryan P, Roberts-Thomson IC, 2001, Metabolic genotypes and risk for colorectal cancer. J Gastroenterology Hepatol 16:631–635 Lu X M, Zang YM, Lin RY et al, 2005, Relationship between genetic polymorphisms of metabolizing enzymes CYP2E1, GSTM1 and Kazakh’s esophageal squamous cell cancer in Xinjiang, China. World J Gastroenterol 28: 11(24): 3651-3654. Xing D, Tan W, Song N, Lin D, 2000, Genetic polymorphism in hOGG1and susceptibility to esophageal cancer in Chinese. Zhonghuayixue Yichuanxue Zazhi 17:377–380 Tan W, Chen GF, Xing DY, Song CY, Kadlubar FF, Lin DX, 2001, Frequency of CYP2A6 gene deletion and its relation to risk of lung and esophageal cancer in the Chinese population. Int J Cancer 95:96–101 Tan W, Song N, Wang GQ et al, 2000, Impact of genetic polymorphisms in cytochrome P450 2E1 and glutathione S-transferases M1, T1, and P1 on susceptibility to esophageal cancer among high-risk individuals in China. Cancer Epidemiol Biomarkers Prev 9:551–556 Dong CH, Yu SZ, Chen GC, Zhao DM, Hu Y, 1998, Association of polymorphisms of glutathioneS transferase M1 and T1 genotypes with elevated aflatoxin and increased risk of primary liver cancer. Shijie Huaren Xiaohua Zazhi 6:463–466 Thong S, Wyllie AH, Barnes D, Wolf CR, Spurr NK, 1993, Relationship between the GSTMI genetic polymorphism and susceptibility to bladder, breast and colon cancer. Carcinogenesis 14:1821–1824 Chevenix-Trench G, Young J, Coggan M, Board P, 1995, Glutathione S-transferase Ml, and Tl polymorphisms: susceptibility to colon cancer and age of onset. Carcinogenesis 16:1655–1657 Katoh T, Nagata N, Kuroda Y et al, 1996, Glutathione Stransferase MI, GSTM1), and Tl(GSTT1, genetic polymorphism and susceptibility to gastric and colorectal adenocarcinoma. Carcinogenesis 17:1855–1859 Deakin M, Elder J, Hendrickse C et al, 1996, Glutathione Stransferase GSTI’l genotypes and susceptibility to cancer: studies of interactions with GSTM1 in lung, oral, gastric and colorectal cancers. Carcinogenesis 17:881–884 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 651 EP 656 DI 10.1007/s12253-011-9490-8 PG 6 ER PT J AU Li, Z Liao, Q Wu, Y Liao, M Hao, Y Zhang, Sh Song, Sh Li, B Zhang, Yd AF Li, Zhehai Liao, Qiande Wu, Yuchi Liao, Mingmei Hao, Yuqin Zhang, Shengbin Song, Shipeng Li, Bing Zhang, Yang-de TI Upregulation of a Disintegrin and Metalloprotease 8 Influences Tumor Metastasis and Prognosis in Patients with Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; A disintegrin and metalloprotease 8; Immunohistochemistry; Prognosis; Overall survival; Disease-free survival ID Osteosarcoma; A disintegrin and metalloprotease 8; Immunohistochemistry; Prognosis; Overall survival; Disease-free survival AB To investigate the clinicopathological and prognostic value of a disintegrin and metalloprotease 8 (ADAM8) in osteosarcoma. ADAM8 expression in osteosarcoma tissues was examined by immunohistochemistry in 69 patients. ADAM8 was positively expressed in 61 of 69 (88.4%) osteosarcoma specimens with cytoplasmic staining, and also increased in the specimens with recurrence (P00.008) and metastasis (P00.002). Patients with strong ADAM8 expression had significantly poorer overall survival (OS) and disease-free survival (DFS) (both P<0.001) when compared with the patients with the weak expression of ADAM8. On multivariate analysis, ADAM8 expression was found to be an independent prognostic factor for both OS (P<0.001) and DFS (P<0.001). Our results suggest for the first time that ADAM8 might be applied as a novel marker for the prediction of recurrence and metastasis potency and a significant indicator of poor prognosis for patients with osteosarcoma. C1 [Li, Zhehai] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, Xiangya Road 87, 410078 Changsha, Hunan, China. [Liao, Qiande] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, Xiangya Road 87, 410078 Changsha, Hunan, China. [Wu, Yuchi] Inner Mongolia Medical College, The Third Affiliated Hospital, Shaoxian Road 20, Kun District, 014010 Baotou, Inner Mongolia, China. [Liao, Mingmei] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, Xiangya Road 87, 410078 Changsha, Hunan, China. [Hao, Yuqin] Inner Mongolia Medical College, The Third Affiliated Hospital, Shaoxian Road 20, Kun District, 014010 Baotou, Inner Mongolia, China. [Zhang, Shengbin] Inner Mongolia Medical College, The Third Affiliated Hospital, Shaoxian Road 20, Kun District, 014010 Baotou, Inner Mongolia, China. [Song, Shipeng] Inner Mongolia Medical College, The Third Affiliated Hospital, Shaoxian Road 20, Kun District, 014010 Baotou, Inner Mongolia, China. [Li, Bing] Inner Mongolia Medical College, The Third Affiliated Hospital, Shaoxian Road 20, Kun District, 014010 Baotou, Inner Mongolia, China. [Zhang, Yang-de] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, Xiangya Road 87, 410078 Changsha, Hunan, China. RP Zhang, Yd (reprint author), Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410078 Changsha, China. EM zydjs@sohu.com CR Urakawa H, Nishida Y, Naruse T, Nakashima H, Ishiguro N, 2009, Cyclooxygenase-2 overexpression predicts poor survival in patients with high-grade extremity osteosarcoma: a pilot study. Clin Orthop Relat Res 467:2932–2938 Yao Y, Dong Y, Lin F et al, 2009, The expression of CRM1 is associated with prognosis in human osteosarcoma. Oncol Rep 21:229–235 Jaffe N, 2009, Adjuvant chemotherapy in osteosarcoma: an odyssey of rejection and vindication. In: Jaffe N, Bielack SS, Bruland OS, eds, Pediatric and adolescent osteosarcoma, cancer treatment and research. Springer, New York, p 152 Wang YC, Zheng LH, Ma BA et al, 2011, Clinical value of signal transducers and activators of transcription 3, STAT3, gene expression in human osteosarcoma. Acta Histochem 113:402–408 Dijkstra A, Postma DS, Noordhoek JA et al, 2009, Expression of ADAMs, “a disintegrin and metalloprotease”, in the human lung. Virchows Arch 454:441–449 Mochizuki S, Okada Y, 2007, ADAMs in cancer cell proliferation and progression. Cancer Sci 98:621–628 Hernandez I, Moreno JL, Zandueta C, Montuenga L, Lecanda F, 2010, Novel alternatively spliced ADAM8 isoforms contribute to the aggressive bone metastatic phenotype of lung cancer. Oncogene 29:3758–3769 Hall T, Pegg LE, Pauley AM, Fischer HD, Tomasselli AG, Zack MD, 2009, ADAM8 substrate specificity: influence of pH on preprocessing and proteoglycan degradation. Arch Biochem Biophys 491:106–111 Zack MD, Melton MA, Stock JL et al, 2009, Reduced incidence and severity of experimental autoimmune arthritis in mice expressing catalytically inactive A disintegrin and metalloproteinase 8, ADAM8). Clin Exp Immunol 158:246–256 Ishizuka H, Garcia-Palacios V, Lu G et al, 2011, ADAM8 enhances osteoclast precursor fusion and osteoclast formation in vitro and in vivo. J Bone Miner Res 26:169–181 Ishikawa N, Daigo Y, Yasui W, 2004, ADAM8 as a novel serological and histochemical marker for lung cancer. Clin Cancer Res 10:8363–8370 Roemer A, Schwettmann L, Jung M, 2004, The membrane proteases adams and hepsin are differentially expressed in renal cell carcinoma. Are they potential tumor markers? J Urol 172:2162– 2166 Fritzsche FR, Jung M, Xu C, 2006, ADAM8 expression in prostate cancer is associated with parameters of unfavorable prognosis. Virchows Arch 449:628–636 Ainola M, Li TF, Mandelin J et al, 2009, Involvement of a disintegrin and a metalloproteinase 8, ADAM8, in osteoclastogenesis and pathological bone destruction. Ann Rheum Dis 68:427–434 Choi SJ, Han JH, Roodman GD, 2001, ADAM8: a novel osteoclast stimulating factor. J Bone Miner Res 16:814–822 Mandelin J, Li TF, Hukkanen MV, 2003, Increased expression of a novel osteoclast-stimulating factor, ADAM8, in interface tissue around loosened hip prostheses. J Rheumatol 30:2033–2038 Egeblad M, Werb Z, 2002, New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2:161–174 Shiomi T, Okada Y, 2003, MT1-MMP and MMP-7 in invasion and metastasis of human cancers. Cancer Metastasis Rev 22:145–152 Wildeboer D, Naus S, Amy Sang QX, Bartsch JW, Pagenstecher A, 2006, Metalloproteinase disintegrins ADAM8 and ADAM19 are highly regulated in human primary brain tumors and their expression levels and activities are associated with invasiveness. J Neuropathol Exp Neurol 65:516–527 Valkovskaya N, Kayed H, Felix K et al, 2007, ADAM8 expression is associated with increased invasiveness and reduced patient survival in pancreatic cancer. J Cell Mol Med 11:1162–1174 Valkovskaya NV, 2008, Hypoxia-dependent expression of ADAM8 in human pancreatic cancer cell lines. Exp Oncol 30:129– 132 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 657 EP 661 DI 10.1007/s12253-011-9491-7 PG 5 ER PT J AU Carmignani, L Picozzi, S Casellato, S Bozzini, G Marenghi, C Macchi, A Lunelli, L Rubino, B Clemente, C AF Carmignani, Luca Picozzi, Stefano Casellato, Stefano Bozzini, Giorgio Marenghi, Carlo Macchi, Alberto Lunelli, Luca Rubino, Barbara Clemente, Claudio TI A Proposed New Technique in Prostate Cancer Tissue Bio-Banking: Our Experience with a New Protocol SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate; Cancer; Prostate cancer; Bio-bank; Tissue; Biopsy ID Prostate; Cancer; Prostate cancer; Bio-bank; Tissue; Biopsy AB The aim of our study, beyond validating a method of collecting and storing biological samples from patients with prostate cancer, was to validate an innovative biopsy method for the creation of a biobank of prostatic frozen tissues. Patients referred to our hospital between November 2008 and March 2010 to undergo radical prostatectomy were invited to participate in the study. Each patient’s data were stored in two databases (personal information and clinical database) while samples of urine, blood and its derivatives, fresh material and formalin-processed tissue were stored in a correlated biobank. The proposed method for collecting fresh material was to take samples of the neoplastic tissue by carrying out targeted biopsies in the area indicated by the biopsy mapping as the site of the malignancy, under manual palpation to identify the neoplastic nodule. The site of sampling was marked by an injection of India ink. 55 patients agreed to participate in the study. In 43 cases biopsies were correct, with a mean of 48% of core involved by tumour (range, 10–90%). Overall the tumour detection rate was 78.2%. The protocol for collecting biological material and the new method for collecting fresh tissue reduce internal steps and staff involved, thereby reducing all those variables that cause heterogeneity of material and changes in its quality. This process provides high quality, low cost material for research on prostate cancer. The features of the collection protocol mean that the protocol can also be used in non-academic centres with only limited research funds. C1 [Carmignani, Luca] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Picozzi, Stefano] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Casellato, Stefano] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Bozzini, Giorgio] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Marenghi, Carlo] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Macchi, Alberto] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Lunelli, Luca] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Rubino, Barbara] Istituto Clinico S. Ambrogio, Division of Pathology, Via Faravelli 16, 20149 Milan, Italy. [Clemente, Claudio] Istituto Clinico S. Ambrogio, Division of Pathology, Via Faravelli 16, 20149 Milan, Italy. RP Picozzi, S (reprint author), IRCCS Policlinico San Donato, University of Milan, Urology Department, 20097 San Donato Milanese, Italy. EM stepico@tin.it CR Dhir R, 2008, Prostate cancer biobanking. Curr Opin Urol 18:309–14 Melamed J, Datta MW, Becich MJ et al, 2004, The cooperative prostate cancer tissue resource: a specimen and data resource for cancer researchers. Clin Cancer Res 10:4614–21 Bova GS, Fox WM, Epstein JI, 1993, Methods of radical prostatectomy specimen processing: a novel technique for harvesting fresh prostate cancer tissue and review of processing techniques. Mod Pathol 6:201–7 Bonavina L, Laface L, Picozzi S et al, 2010, Proposal of a punch biopsy protocol as a pre-requisite for the establishment of a tissue bank from resected esophageal tumors. Pathol Oncol Res 16:457–60 Greene FL, Page DL, Fleming ID et al, 2002, AJCC Cancer Staging Manual, 6th edn. Springer, New York, NY Bosso N, Chinello C, Picozzi SC et al, 2008, Human urine biomarkers of renal cell carcinoma evaluated by ClinProt. Proteomics Clin Appl 2:1036–46 Hulmes JD, Bethea D, Ho K et al, 2004, An investigation of plasma collection, stabilization, and storage procedures for proteomic analysis of clinical samples. Clinical Proteomics 1:17–31 Thongboonkerd V, 2007, Proteomics of human body fluids: principles, methods, and applications. Humana Press Inc., New Jersey Gore JL, Shariat SF, Miles BJ et al, 2001, Optimal combinations of systematic sextant and laterally directed biopsies for the detection of prostate cancer. J Urol 165:1554–9 Etzioni R, Penson DF, Legler JM et al, 2002, Overdiagnosis due to prostate-specific antigen screening: Lessons from U.S. prostate cancer incidence trends. J Natl Cancer Inst 94:981–990 Johansson JE, Andren O, Andersson SO et al, 2004, Natural history of early, localized prostate cancer. JAMA 291:2713–9 Schroeder FH, Hugosson J, Roobol MJ et al, 2009, Screening and prostate-cancer mortality in a randomized European study. N Eng J Med 360:1320–8 http://grants.nih.gov/grants/guide/rfa-files/RFA-CA-99–012.html. Milanes-Yearsley M, Hammond ME, Pajak TF et al, 2002, Tissue micro-array: a cost and time-effective method for correlative studies by regional and national cancer study groups. Mod Pathol 15:1366–73 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 663 EP 668 DI 10.1007/s12253-011-9492-6 PG 6 ER PT J AU Nagy, Zs Horvath, O Kadas, J Valtinyi, D Laszlo, L Kopper, B Blasko, Gy AF Nagy, Zsuzsanna Horvath, Orsolya Kadas, Julia Valtinyi, Dorottya Laszlo, Larisza Kopper, Bence Blasko, Gyorgy TI D-Dimer as a Potential Prognostic Marker SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer; LMWH; D-dimer; prognostic marker; solid tumors ID cancer; LMWH; D-dimer; prognostic marker; solid tumors AB Malignant tumors are often accompanied by increased risk for procoagulant activity, thrombosis and embolism. As a marker indicating such disturbancies is D-dimer, a product of fibrinolysis. In this retrospective study almost 300 patients with malignant tumors were evaluated. During LMWH treatment (as thromboprophylaxis) the highest frequency of VTE with worst prognosis occurred in pancreatic cancer (partly due to the late discovery) followed by ovarian, colonic and breast cancers. Also, increased D-dimer level correlated with progression (stages) and high mortality rate. Furthermore, D-dimer showed very similar or better prognostic activity than the clinically widely used classic tumor markers and suggested to use it as an additional value. C1 [Nagy, Zsuzsanna] St. Imre Hospital, Department Clinical Oncology, Tetenyi ut 12-15, 1116 Budapest, Hungary. [Horvath, Orsolya] St. Imre Hospital, Department Clinical Oncology, Tetenyi ut 12-15, 1116 Budapest, Hungary. [Kadas, Julia] St. Imre Hospital, Department Clinical Oncology, Tetenyi ut 12-15, 1116 Budapest, Hungary. [Valtinyi, Dorottya] St. Imre Hospital, Department Clinical Oncology, Tetenyi ut 12-15, 1116 Budapest, Hungary. [Laszlo, Larisza] St. Imre Hospital, Department Clinical Oncology, Tetenyi ut 12-15, 1116 Budapest, Hungary. [Kopper, Bence] Semmelweis University, Department of Biophysics and Radiation BiologyBudapest, Hungary. [Blasko, Gyorgy] University of Debrecen, Department of Pharmacology and PharmacotherapyDebrecen, Hungary. RP Nagy, Zs (reprint author), St. Imre Hospital, Department Clinical Oncology, 1116 Budapest, Hungary. EM zsulacz@chello.hu CR Bouillard JB, Bouillaud S, 1823, De l’Obliteration des veines et de son influence sur la formation des hydropisies partielles: consideration sur la hydropisies passive et general. Arch GenMed 1:188–204 Trousseau A, 1865, Phlegmasia alba dolens. Clinique Medicale de l’Hotel Dieu de Paris 3:654–712 Blom JW, Doggen CJ, Osanto S, Rosendaal FR, 2005, malignancies, prothrombotic mutations, and the risk of venous thrombosis. JAMA 293:492–496 Murchinson JT, Wylie L, Stockton DL, 2004, Excess risk of cancer in patients with primary venous thromboembolism: a national, population-based cohort study. Br J Cancer 91:92–95 Noble S, Pasi J, 2010, Epidemiology and pathophysiology of cancer-associated thrombosis. Brit J Cancer 102:52–59 Kakkar AK, 2005, Low molecular weight heparin and survival in patients with malignant disease. Cancer Control 12:22–30 Kakkar AK, Levine MN, Kadziola Z, 2004, Low molecular weight heparin, therapy with deltaparin, and survival in advanced cancer: the fragmin advanced malignancy outcome study, FAMOUS). J Clin Oncol 22:1944–1948 Klerk CPW, Smorenburg SM, Otten H-M et al, 2005, The effect of low molecular weight heparin on survivcal in patients with advanced malignancy. J Clin Oncol 23:2130–2135 Tovari J, Berecky B, Gilly R et al, 2004, Effect of heparin treatment on the metastatization of melanoma in a preclinical model, in Hungarian). Magyar Onkologia 48:235–241 Lazo-Langer A, Gross GD, Spaans JN et al, 2007, The effect of low-molecular-weight heparin on cancer survival. A systematic review and meta-analysis of randomized trial. Thromb Hemostasis 5:729–737 Niers TMH, Bruggeman LW, Van Sluis GL et al, 2009, Long-term thrombin inhibition promotes cancer cell extravasation in a mouse model of experimental metastasis. Internat Soc Thrombosis Haemostasis 7:1595–1597 Mousa SA, Petersen LJ, 2009, Anticancer properties of lowmolecular- weight heparin: preclinical evidence. Thromb Haemost 102:258–267 Borsig I, 2010, Antimetastatic effect of heparins and modified heparins. Experimental evidence. Thrombosis Res 125(suppl 2): S66–S71 Kenessey I, Simon E, Futosi K et al, 2009, Antimigratory and antimetastatic effect of heparin-derived 4-18 unit oligosaccharides in a preclinical humanmelanoma metastasis model. Thromb Haemost 102:1265–1273 Nadir Y, Brenner B, 2010, Heparanase procoagulant effects and inhibition by heparins. Thromb Res 125(suppl 2):572–576 Kvolik S, Jukic M, Matijevic M et al, 2010, An overview of coagulation disorders in cancer patients. Surg Oncol 29:e33–e46 Khorana AA, Kunderer NM, Culakova E et al, 2008, Development and validation of a predictive model for chemotherapyassociated thrombosis. Blood 111:4902–4907 Doormaal FF et al, 2011, Randomized trial of the effect of the low molecular weight heparin nadroparin on survival in patients with cancer. J Clin Oncol 29:2701–2706 Zs N, Turcsik V, Gy B, 2009, The effect of LMWH, Nadroparin, on Tumor progression. Pathol Oncol Res Path Oncol Res 15:689– 692 Kulasingam V, Pavlou MR, Diamandis EP, 2010, Integrating high-throughput technologies in the quest for effective biomarkers for ovarian cancer. Nature Cancer Rev 10:371–378 Moore RG et al, 2009, A novel multiple marker bioassay utilizing HE4 and CA125 for the prediction of ovarian cancer in patients with a pelvic mass. Gynecol Oncol 112:40–46 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 669 EP 674 DI 10.1007/s12253-011-9493-5 PG 6 ER PT J AU Andjelic, MB Mihaljevic, SB Jakovic, RL AF Andjelic, M Bosko Mihaljevic, S Biljana Jakovic, R Ljubomir TI ABVD as the Treatment Option in Advanced Hodgkin’s Lymphoma Patients Older than 45 Years SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Advanced Hodgkin’s lymphoma; Elderly patients ID Advanced Hodgkin’s lymphoma; Elderly patients AB Advanced age is considered an unfavourable prognostic factor for Hodgkin’s lymphoma (HL). The optimal treatment for these patients is not yet defined, especially for the advanced stages. We analysed the outcome and prognostic relevance of patient and disease characteristics in 46 advanced stage HL patients who were older than 45 years, treated with ABVD. Elderly patients (>60 year) had a significantly higher rate of comorbidities (p<0.05). The complete remission rate was significantly lower in elderly patients and in patients with an IPS ≥3 (p<0.05, p<0.05, respectively). Elderly patients had significantly shorter event-free survival (p<0.01) and overall survival (p<0.01) compared to patients of 45–60 year. Extranodal disease, an IPS ≥3, bulky disease, an ESR>50 and the presence of a large mediastinal tumour mass didn’t have an influence on survival (p>0.05). The multivariate Cox regression analysis identified the age of >60 year as an independent prognostic factor. The prospective clinical trials seem to be needed for defining the optimal therapeutic approach in elderly patients. C1 [Andjelic, M Bosko] Clinical Center of Serbia, Clinic for Hematology, 2 Koste Todorovic St, 11000 Belgrade, Serbia. [Mihaljevic, S Biljana] Clinical Center of Serbia, Clinic for Hematology, 2 Koste Todorovic St, 11000 Belgrade, Serbia. [Jakovic, R Ljubomir] Clinical Center of Serbia, Clinic for Hematology, 2 Koste Todorovic St, 11000 Belgrade, Serbia. RP Andjelic, MB (reprint author), Clinical Center of Serbia, Clinic for Hematology, 11000 Belgrade, Serbia. EM boleoli@sezampro.rs CR Brusamolino E, Bacigalupo A, Barosi G et al, 2009, Classical Hodgkin’s lymphoma in adults: guidelines of the Italian Society of Hematology, the Italian Society of Experimental Hematology, and the Italian Group for Bone Marrow Transplantation on initial work-up, management, and follow-up. Haematologica 94:550–565 Klimm B, Engert A, 2009, Combined modality treatment of Hodgkin’s lymphoma. Cancer J 15:143–149 Klimm B, Diehl V, Engert A, 2007, Hodgkin’s lymphoma in the elderly: a different disease in patients over 60. Oncology, Williston Park, 21:982–990, discussion 990, 996, 998 passim Peterson BA, Pajak TF, Cooper MR et al, 1982, Effect of age on therapeutic response and survival in advanced Hodgkin’s disease. Cancer Treat Rep 66:889–898 Specht L, Nissen N, 1989, Hodgkin’s disease and age. Eur J Haematol 43:127–135 Bosi A, Ponticelli P, Casini C et al, 1989, Clinical data and therapeutic approach in elderly patients with Hodgkin’s disease. Haematologica 74:463–473 Diaz-Pavon JR, Cabanillas F, Majlis A, Hagemeister FB, 1995, Outcome of Hodgkin’s disease in elderly patients. Hematol Oncol 13:19–27 Klimm B, Eich HT, Haverkamp H et al, 2007, Poorer outcome of elderly patients treated with extended-field radiotherapy compared with involved-field radiotherapy after chemotherapy for Hodgkin’s lymphoma: an analysis from the German Hodgkin Study Group. Ann Oncol 18:357–363 Hasenclever D, Diehl V, 1998, A prognostic score for advanced Hodgkin’s disease. N Engl J Med 339:1506–1514 Levis A, Anselmo AP, Ambrosetti A et al, 2004, VEPEMB in elderly Hodgkin’s lymphoma patients. Results from an Intergruppo Italiano Linfomi, IIL, study. Ann Oncol 15:123–128 Wedelin C, Bjorkholm M, Biberfeld P, Holm G, Johansson B, Mellstedt H, 1984, Prognostic factors in Hodgkin’s disease with special reference to age. Cancer 53:1202–1208 Eghbali H, Hoerni-Simon G, de Mascarel I, Durand M, Chauvergne J, Hoerni B, 1984, Hodgkin’s disease in the elderly. A series of 30 patients aged older than 70 years. Cancer 53:2191–2193 Enblad G, Glimelius B, Sundstrom C, 1991, Treatment outcome in Hodgkin’s disease in patients above the age of 60: a population based study. Ann Oncol 2:297–302 Guinee VF, Giacco GG, Durand M et al, 1991, The prognosis of Hodgkin’s disease in older adults. J Clin Oncol 9:947–953 Erdkamp FL, Breed WP, Bosch LJ, Wijnen JT, Blijham GB, 1992, Hodgkin’s disease in the elderly. A registry-based analysis. Cancer 70:830–834 Levis A, Depaoli L, Urgesi A et al, 1994, Probability of cure in elderly Hodgkin’s disease patients. Haematologica 79:46–54 Forsyth PD, Bessell EM, Moloney AJ, Leach IH, Davies JM, Fletcher J, 1997, Hodgkin’s disease in patients older than 70 years of age: a registry-based analysis. Eur J Cancer 33:1638–1642 Landgren O, Algernon C, Axdorph U et al, 2003, Hodgkin’s lymphoma in the elderly with special reference to type and intensity of chemotherapy in relation to prognosis. Haematologica 88:438–444 Levis A, Depaoli L, Bertini M et al, 1996, Results of a low aggressivity chemotherapy regimen, CVP/CEB, in elderly Hodgkin’s disease patients. Haematologica 81:450–456 Zinzani PL, Magagnali M, Bendandi M et al, 2000, Efficacy of VBM regimen in the treatment of elderly patients with Hodgkin’s disease. Haematologica 85:729–732 Austin-Seymour MM, Hoppe RT, Cox RS, Rosenberg SA, Kaplan HS, 1984, Hodgkin’s disease in patients over 60 years old. Ann Intern Med 100:13–18 Canellos GP, Anderson JR, Propert K et al, 1992, Chemotherapy of advanced Hodgkin’s disease with MOPP, ABVD, or MOPP alternating with ABVD. N Engl J Med 327:1478–1484 Duggan DB, Petroni GR, Johnson JL et al, 2003, Randomized comparison of ABVD and MOPP/ABVD hybrid for the treatment of advanced Hodgkin’s disease: report of an intergroup trial. J Clin Oncol 21:607–614 Gobbi PG, Levis A, Chisesi T et al, 2005, ABVD versus modified Stanford V versus MOPPEBVCAD with optional and limited radiotherapy in intermediate-and advanced-stage Hodgkin’s lymphoma. Final results of a multicenter randomized trial by the Intergruppo Italiano Linfomi. J Clin Oncol 23:9198–9207 Johnson PW, Radford JA, Cullen MH et al, 2005, Comparison of ABVD and alternating or hybrid multidrug regimens for the treatment of advanced Hodgkin’s lymphoma: results of the United Kingdom lymphoma group. LY09 trial. J Clin Oncol 23:9208–9218 Canellos GP, Niedzwiecki D, 2002, Long-term follow-up of Hodgkin’s disease trial. N Engl J Med 346:1417–1418 Federico M, Luminari S, Iannitto E et al, 2009, ABVD compared with BEACOPP compared with CEC for the initial treatment of patients with advanced Hodgkin’s lymphoma: results from the HD2000 Gruppo Italiano per lo Studio dei Linfomi Trial. J Clin Oncol 27:805–811 Hoskin PJ, Lowry L, Horwich A et al, 2009, Randomized comparison of the Stanford V regimen and ABVD in the treatment of advanced Hodgkin’s lymphoma: United Kingdom National Cancer Research Institute Lymphoma Group Study ISRCTN 64141244. J Clin Oncol 27:5390–5396 Weekes CD, Vose JM, Lynch JC et al, 2002, Hodgkin’s disease in the elderly: improved treatment outcome with a doxorubicincontaining regimen. J Clin Oncol 20:1087–1093 Borchmann P, Engert A, 2010, The past: what we have learned in the last decade. Hematology Am Soc Hematol Educ Program 2010:101–107 Ballova V, Ruffer JU, Haverkamp H et al, 2005, A prospectively randomized trial carried out by the German Hodgkin Study Group, GHSG, for elderly patients with advanced Hodgkin’s disease comparing BEACOPP baseline and COPP-ABVD, study HD9elderly). Ann Oncol 16:124–131 Engert A, Schiller P, Josting A et al, 2003, Involved-field radiotherapy is equally effective and less toxic compared with extendedfield radiotherapy after four cycles of chemotherapy in patients with early-stage unfavorable Hodgkin's lymphoma: results of the HD8 trial of the German Hodgkin’s Lymphoma Study Group. J Clin Oncol 21:3601–3608 Raemaekers J, Kluin-Nelemans H, Teodorovic I et al, 2002, The achievements of the EORTC lymphoma group. European organisation for research and treatment of cancer. Eur J Cancer 38(Suppl 4):S107–S113 Boll B, Borchmann P, Topp MS et al, 2010, Lenalidomide in patients with refractory or multiple relapsed Hodgkin lymphoma. Br J Haematol 148:480–482 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 675 EP 680 DI 10.1007/s12253-011-9494-4 PG 6 ER PT J AU Savic, A Cemerikic-Martinovic, V Dovat, S Rajic, N Urosevic, I Sekulic, B Kvrgic, V Popovic, S AF Savic, Aleksandar Cemerikic-Martinovic, Vesna Dovat, Sinisa Rajic, Nebojsa Urosevic, Ivana Sekulic, Borivoj Kvrgic, Vanja Popovic, Stevan TI Angiogenesis and Survival in Patients with Myelodysplastic Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Myelodysplastic syndrome; Angiogenesis; Microvessel density; VEGF; Survival; Prognosis ID Myelodysplastic syndrome; Angiogenesis; Microvessel density; VEGF; Survival; Prognosis AB Angiogenesis has been implicated in the pathogenesis and prognosis of myelodysplastic syndrome (MDS). In this study, we investigated the relationship between microvessel density (MVD), vascular endothelial growth factor (VEGF) expression, common morphological and clinical factors, and survival in patients with MDS. We examined the MVD of paraffin-embedded bone marrow sections from 70 MDS patients and 31 controls. VEGF expression was determined in 50 patients and 20 controls. The median MVD in MDS patients was significantly higher than that in controls (p00.025), whereas there was no difference in VEGF expression between MDS patients and controls. In univariate analysis, increased MVD was associated with a shorter survival time (p00.023). However, in multivariate analysis, MVD was not an independent predictor of survival. The VEGF expression did not influence survival in univariate analysis. Survival was independently influenced by platelet count (p00.0073), cytogenetic risk category (p00.022), and transfusion dependence (p00.0073). Neither MVD nor VEGF expression were predictors for progression to acute myeloid leukemia in univariate analysis. Progression to acute myeloid leukemia was independently influenced only by the cytogenetic risk category (p00.022). This study confirmed increased MVD in MDS. It does not support an independent prognostic role of angiogenesis in MDS. C1 [Savic, Aleksandar] Clinical Center of Vojvodina, Faculty of Medicine, Clinic of Hematology, Hajduk Veljkova 1-3, 21000 Novi Sad, Serbia. [Cemerikic-Martinovic, Vesna] Pathohistology Laboratory Beolab, Resavska 60, 11000 Belgrade, Serbia. [Dovat, Sinisa] Pennsylvania State University, College of Medicine, Milton S. Hershey Medical Center, Children’s Hospital, Department of Pediatrics, H085, Division of Pediatric Hematology/Oncology, 500 University Drive, 17033 Hershey, PA, USA. [Rajic, Nebojsa] Clinical Center of Vojvodina, Faculty of Medicine, Clinic of Hematology, Hajduk Veljkova 1-3, 21000 Novi Sad, Serbia. [Urosevic, Ivana] Clinical Center of Vojvodina, Faculty of Medicine, Clinic of Hematology, Hajduk Veljkova 1-3, 21000 Novi Sad, Serbia. [Sekulic, Borivoj] Clinical Center of Vojvodina, Faculty of Medicine, Clinic of Hematology, Hajduk Veljkova 1-3, 21000 Novi Sad, Serbia. [Kvrgic, Vanja] Clinical Center of Vojvodina, Faculty of Medicine, Clinic of Hematology, Hajduk Veljkova 1-3, 21000 Novi Sad, Serbia. [Popovic, Stevan] Clinical Center of Vojvodina, Faculty of Medicine, Clinic of Hematology, Hajduk Veljkova 1-3, 21000 Novi Sad, Serbia. RP Savic, A (reprint author), Clinical Center of Vojvodina, Faculty of Medicine, Clinic of Hematology, 21000 Novi Sad, Serbia. EM asavic@uns.ac.rs CR Folkman J, 1995, Angiogenesis in cancer, vascular, rheumatoid, and other disease. Nat Med 1:27–31 Folkman J, 1971, Tumor angiogenesis: therapeutic implications. N Engl J Med 285:1182–1186 Padro T, Ruiz S, Bieker R et al, 2000, Increased angiogenesis in the bone marrow of patients with acute myeloid leukemia. Blood 95:2637–2644 Hussong JW, Rodgers JM, Shami PJ, 2000, Evidence of increased angiogenesis in patients with acute myeloid leukemia. Blood 95:309–313 Lundberg LG, Lerner R, Sundelin P et al, 2000, Bone marrow in polycythemia vera, chronic myelocytic leukemia, and myelofibrosis has an increased vascularity. Am J Pathol 157:15–19 Mesa RA, Hanson CA, Rajkumar SV et al, 2000, Evaluation and clinical correlations of bone marrow angiogenesis in myelofibrosis with myeloid metaplasia. Blood 96:3374–3380 Peterson L, Kini AR, 2001, Angiogenesis is increased in B-cell chronic lymphocytic leukemia. Blood 97:2529 Rajkumar SV, Leong T, Roche PC et al, 2000, Prognostic value of bone marrow angiogenesis in multiple myeloma. Clin Cancer Res 6:3111–3116 Aguayo A, Kantarjian H, Manshouri T et al, 2000, Angiogenesis in acute and chronic leukemias and myelodysplastic syndromes. Blood 96:2240–2245 Pruneri G, Bertolini F, Soligo D et al, 1999, Angiogenesis in myelodysplastic syndromes. Br J Cancer 81:1398–1401 Korkolopoulou P, Apostolidou E, Pavlopoulos PM et al, 2001, Prognostic evaluation of the microvascular network in myelodysplastic syndromes. Leukemia 15:1369–1376 Lundberg LG, Hellstrom-Lindberg E, Kanter-Lewensohn L et al, 2006, Angiogenesis in relation to clinical stage, apoptosis and prognostic score in myelodysplastic syndromes. Leuk Res 30:247– 253 Alexandrakis MG, Passam FH, Pappa CA et al, 2005, Relation between bone marrow angiogenesis and serum levels of angiogenin in patients with myelodysplastic syndromes. Leuk Res 29:41– 46 Gale NW, Yancopoulos GD, 1999, Growth factors acting via endothelial cell-specific receptor tyrosine kinases: VEGFs, angiopoietins, and ephrins in vascular development. Genes Dev 13:1055–1066 Bellamy WT, Richter L, Sirjani D et al, 2001, Vascular endothelial cell growth factor is an autocrine promoter of abnormal localized immature myeloid precursors and leukemia progenitor formation in myelodysplastic syndromes. Blood 97:1427–1434 Wimazal F, Krauth MT, Vales A et al, 2006, Immunohistochemical detection of vascular endothelial growth factor, VEGF, in the bone marrow in patients with myelodysplastic syndromes: correlation between VEGF expression and the FAB category. Leuk Lymphoma 47:451–460 Bennett JM, Catovsky D, Daniel MT et al, 1982, Proposals for the classification of the myelodysplastic syndromes. Br J Haematol 51:189–199 Brunning RD, Orazi A, Germing U et al, 2008, Myelodysplastic syndromes/Neoplasms, overview. In: Swerdlow SH, Campo E, Harris NL et al, eds, WHO classification of tumours of haematopoietic and lymphoid tissues, 4th edn. IARC, Lyon, pp 88–93 Greenberg P, Cox C, LeBeau MM et al, 1997, International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 89:2079–2088 Thiele J, Kvasnicka HM, Facchetti F et al, 2005, European consensus on grading bone marrow fibrosis and assessment of cellularity. Haematologica 90:1128–1132 Tricot G, De Wolf-Peeters C, Vlietinck R et al, 1984, Bone marrow histology in myelodysplastic syndromes. II. Prognostic value of abnormal localization of immature precursors in MDS. Br J Haematol 58:217–225 Perez-Atayde AR, Sallan SE, Tedrow U et al, 1997, Spectrum of tumor angiogenesis in the bone marrow of children with acute lymphoblastic leukemia. Am J Pathol 150:815–821 Keith T, Araki Y, Ohyagi M et al, 2007, Regulation of angiogenesis in the bone marrow of myelodysplastic syndromes transforming to overt leukaemia. Br J Haematol 137:206–215 Malcovati L, Germing U, Kuendgen A et al, 2007, Timedependent prognostic scoring system for predicting survival and leukemic evolution in myelodysplastic syndromes. J Clin Oncol 25:3503–3510 Verstovsek S, Estey E, Manshouri T et al, 2002, Clinical relevance of vascular endothelial growth factor receptors 1 and 2 in acute myeloid leukaemia and myelodysplastic syndrome. Br J Haematol 118:151–156 Gabrilove J, 2001, Angiogenic growth factors: autocrine and paracrine regulation of survival in hematologic malignancies. Oncologist 6(Suppl 5):4–7 Dias S, Hattori K, Zhu Z et al, 2000, Autocrine stimulation of VEGFR-2 activates human leukemic cell growth and migration. J Clin Invest 106:511–521 Padro T, Bieker R, Ruiz S et al, 2002, Overexpression of vascular endothelial growth factor, VEGF, and its cellular receptor KDR, VEGFR-2, in the bone marrow of patients with acute myeloid leukemia. Leukemia 16:1302–1310 Germing U, Hildebrandt B, Pfeilstocker M et al, 2005, Refinement of the international prognostic scoring system, IPSS, by including LDH as an additional prognostic variable to improve risk assessment in patients with primary myelodysplastic syndromes, MDS). Leukemia 19:2223–2231 Sperr WR, Wimazal F, Kundi M et al, 2001, Survival analysis and AML development in patients with de novo myelodysplastic syndromes: comparison of six different prognostic scoring systems. Ann Hematol 80:272–277 Horny HP, Sotlar K, Valent P, 2007, Diagnostic value of histology and immunohistochemistry in myelodysplastic syndromes. Leuk Res 31:1609–1616 Valent P, Horny HP, Bennett JM et al, 2007, Definitions and standards in the diagnosis and treatment of the myelodysplastic syndromes: consensus statements and report from a working conference. Leuk Res 31:727–736 Patsouris E, Katsarou O, Korkolopoulou P et al, 2004, Increased microvascular network in bone marrow of HIV-positive haemophilic patients. HIV Med 5:18–25 List A, Dewald G, Bennett J et al, 2006, Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. N Engl J Med 355:1456–1465 Raza A, Reeves JA, Feldman EJ et al, 2008, Phase 2 study of lenalidomide in transfusion-dependent, low-risk, and intermediate- 1 risk myelodysplastic syndromes with karyotypes other than deletion 5q. Blood 111:86–93 Musto P, 2004, Thalidomide therapy for myelodysplastic syndromes: current status and future perspectives. Leuk Res 28:325– 332 Giles FJ, Stopeck AT, Silverman LR et al, 2003, SU5416, a small molecule tyrosine kinase receptor inhibitor, has biologic activity in patients with refractory acute myeloid leukemia or myelodysplastic syndromes. Blood 102:795–801 Giles FJ, Bellamy WT, Estrov Z et al, 2006, The anti-angiogenesis agent, AG-013736, has minimal activity in elderly patients with poor prognosis acute myeloid leukemia, AML, or myelodysplastic syndrome, MDS). Leuk Res 30:801–811 Oh ST, Gotlib J, 2008, Antiangiogenic therapy in myelodysplastic syndromes: is there a role? Curr Hematol Malig Rep 3:10–18 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 681 EP 690 DI 10.1007/s12253-012-9495-y PG 10 ER PT J AU Ziadi, S Boughamoura, H Ben Maitig, M Ben Gacem, R Mestiri, S Chaabani, L Trimeche, M AF Ziadi, Sonia Boughamoura, Hatem Ben Maitig, Mahmoud Ben Gacem, Riadh Mestiri, Sarra Chaabani, Lotfi Trimeche, Mounir TI Immunodetection of SV40 T/t-antigens in Human Osteosrcoma in a Series of Tunisian Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; Simian Virus 40; Immunohistochemistry; Tunisia ID Osteosarcoma; Simian Virus 40; Immunohistochemistry; Tunisia AB Osteosarcoma is a primary bone malignancy that typically occurs during adolescence but also has a second incidence peak in the elderly. The etiology of osteosarcoma is not well understood. Recent investigations have identified SV40 DNA sequences in osteosarcomas, suggesting that SV40 may contribute to tumor development. However, these studies also demonstrated geographical differences in SV40-positive osteosarcomas. The purpose of this study was to determine the prevalence and clinicopathological characteristics of SV40 positive osteosarcoma in Tunisian patients. Fifty-six formalin-fixed paraffin-embedded specimens of osteosarcomas were retrospectively investigated. Samples investigated were clinical cases examined between 1990 and 2004 in the Laboratory of Pathology at the University Hospital Farhat-Hached of Sousse (Tunisia). The search for SV40 was performed by immunohistochemistry using the Pab108 antibody for the detection of the viral oncoproteins: large T antigen and small t antigen (T/t-ag). SV40 status was correlated with clinico-pathological data. T/t-ag immunostaning was detected in the tumor cells in 31/56 (55.4%) osteosarcoma cases. SV40 positivity was more frequent (83%) in patients older than 40 years (5/6 cases) than in patients under 40 years (52%, 26/50), but the difference does not reach statistical significance (p00.33). Moreover, the time between the onset of clinical symptoms and diagnosis was shorter for SV40 positive than SV40 negative cases (p00.08). However, the viral status did not differ significantly according to gender, tumor size, histological subtype, tumor location, or metastases. This study documents the presence SV40 T/t-antigens in a proportion of osteosarcomas in Tunisian patients. The expression of these viral oncoproteins supports the hypothesis that SV40 may have a role in the pathogenesis of this tumor. C1 [Ziadi, Sonia] Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. [Boughamoura, Hatem] Sahloul Hospital, Department of OrthopedicsSousse, Tunisia. [Ben Maitig, Mahmoud] Sahloul Hospital, Department of OrthopedicsSousse, Tunisia. [Ben Gacem, Riadh] Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. [Mestiri, Sarra] Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. [Chaabani, Lotfi] Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. [Trimeche, Mounir] Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. RP Trimeche, M (reprint author), Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. EM m_trimech@yahoo.fr CR Scholz RB, Kabisch H, Weber B et al, 1992, Studies of the RB1 gene and the p53 gene in human osteosarcomas. Pediatr Hematol Oncol 9:125–137 Szendroi M, 2006, Bone tumours. Int Orthop 30:435–436 Crawford LV, Cole CN, Smith AE et al, 1978, Organization and expression of early genes of simian virus 40. Proc Natl Acad Sci USA 75:117–121 De Luca A, Baldi A, Esposito V et al, 1997, The retinoblastoma gene family pRb/p105, p107, pRb2/p130 and simian virus-40 large T-antigen in human mesotheliomas. Nat Med 3:913–916 Mendoza SM, Konishi T, Miller CW, 1998, Integration of SV40 in human osteosarcoma DNA. Oncogene 17:2457–2462 Carbone M, Rizzo P, Procopio A et al, 1996, SV40-like sequences in human bone tumors. Oncogene 13:527–535 Cicala C, Pompetti F, Carbone M, 1993, SV40 induces mesotheliomas in hamsters. Am J Pathol 142:1524–1533 Shein HM, Enders JF, Levinthal JD, 1962, Transformation induced by simian virus 40 in human renal cell cultures. II. Cellvirus relationships. Proc Natl Acad Sci USA 48:1350–1357 Ray FA, Peabody DS, Cooper LJ et al, 1990, SV40 T antigen alone drives karyotype instability that precedes neoplastic transformation of human diploid fibroblasts. J Cell Biochem 42:13–31 Barbanti-Brodano G, Martini F, De Mattei M et al, 1998, BK and JC human polyomaviruses and simian virus 40: natural history of infection in humans, experimental oncogenicity, and association with human tumors. Adv Virus Res 50:69–99 Diamandopoulos GT, 1972, Leukemia, lymphoma, and osteosarcoma induced in the Syrian golden hamster by simian virus 40. Science 176:173–175 Coe JE, Green I, 1975, B-cell origin of hamster lymphoid tumors induced by simian virus 40. J Natl Cancer Inst 54:269–270 Bergsagel DJ, Finegold MJ, Butel JS et al, 1992, DNA sequences similar to those of simian virus SV40 in ependymomas and choroid plexus tumors of childhood. N Engl J Med 326:988–993 Carbone M, Pass HI, Rizzo P et al, 1994, Simian virus 40-like DNA sequences in human pleural mesothelioma. Oncogene 9:1781–1790 Yamamoto H, Nakayama T, Murakami H et al, 2000, High incidence of SV40-like sequences detection in tumour and peripheral blood cells of Japanese osteosarcoma patients. Br J Cancer 82:1677–1681 Vilchez RA, Madden CR, Kozinetz CA et al, 2002, Simian virus 40 and non-Hodgkin lymphoma. Lancet 359:817–823 Amara K, Trimeche T, Ziadi S et al, 2007, Presence of simian virus 40 DNA sequences in diffuse large B-cell lymphomas in Tunisia correlates with aberrant promoter hypermethylation of multiple tumor suppressor genes. Int J Cancer 121:2693–2702 Wong NA, Rae F, Herriot MM et al, 2003, SV40 Tag DNA sequences, present in small proportion of human hepatocellular carcinomas, are associated with reduction survival. J Clin Pathol 56:904–909 Pacini F, Vivaldi A, Santoro M et al, 1998, Simian virus 40-like DNA sequences in human papillary thyroid carcinomas. Oncogene 15:665–669 Hachana M, Trimeche M, Ziadi S et al, 2009, Evidence for a role of the Simian Virus 40 in human breast carcinomas. Breast Cancer Res Treat 113:43–58 Garcea RL, Imperiale MJ, 2003, Simian virus 40 infection of humans. J Virol 77:5039–5045 Barbanti-Brodano G, Sabbioni S, Martini F et al, 2004, Simian virus 40 infection in humans and association with human diseases: results and hypotheses. Virology 318:1–9 Sheppard HM, Corneillie SI, Espiritu C et al, 1999, New insights into the mechanism of inhibition of p53 by simian virus 40 large T antigen. Mol Cell Biol 19:2746–2753 Theile M, Strauss M, Luebbe L et al, 1980, SV40-induced somatic mutations: possible relevance to viral transformation. Cold Spring Harb Symp Quant Biol 44:377–382 Stewart N, Bacchetti S, 1991, Expression of SV40 large T antigen, but not small t antigen, is required for the induction of chromosomal aberrations in transformed human cells. Virology 180:49–57 Tognon M, Casalone R, Martini F et al, 1996, Large T antigen coding sequences of two DNA tumor viruses, BK and SV40, and nonrandom chromosome changes in two glioblastoma cell lines. Cancer Genet Cytogenet 90:17–23 Gaillard S, Fahrbach KM, Parkati R et al, 2001, Overexpression of simian virus 40 small-T antigen blocks centrosome function and mitotic progression in human fibroblasts. J Virol 75:9799–9807 Hurault de Ligny B, Godin M et al, 2003, Virological, epidemiological and pathogenic aspects of human polyomaviruses. Presse Med 32:656–658 Jasani B, Cristaudo A, Emri SA et al, 2001, Association of SV40 with human tumours. Semin Cancer Biol 11:49–61 Klein G, Powers A, Croce C, 2002, Association of SV40 with human tumors. Oncogene 21:1141–1149 Gazdar AF, Butel JS, Carbone M, 2002, SV40 and human tumours: myth, association or causality? Nat Rev Cancer 2:957–964 Heinsohn S, Scholz RB, Weber B, 2000, SV40 sequences in human osteosarcoma of German origin. Anticancer Res 20:4539–4545 Butel JS, Jafar S, Stewart AR, 1998, Detection of authentic SV40 DNA sequences in human brain and bone tumours. Dev Biol Stand 94:23–32 Galateau-Salle F, Bidet P, Iwatsubo Yet al, 1998, SV40-like DNA sequences in pleural mesothelioma, bronchopulmonary carcinoma, and non-malignant pulmonary diseases. J Pathol 184:252–257 Malkin D, Chilton-MacNeil S, Meister LA et al, 2001, Tissue specific expression of SV40 in tumors associated with Li- Fraumeni syndrome. Oncogene 20:4441–4449 Zhen HN, Zhang X, Bu XY et al, 1999, Expression of the simian virus 40 large tumour antigen, Tag, and formation of Tag–p53 and Tag–pRb complexes in human brain tumors. Cancer 86:2124–2132 Martinelli M, Martini F, Rinaldi E et al, 2002, Simian virus 40 sequences and expression of the viral large T antigen oncoprotein in human pleomorphic adenomas of parotid glands. Am J Pathol 161:1127–1133 Heinsohn S, Szendroi M, Bielack S et al, 2009, Evaluation of SV40 in osteosarcoma and healthy population: a Hungarian- German study. Oncol Rep 21:289–297 Lednicky JA, Stewart AR, Jenkins JJ 3rd et al, 1997, SV40 DNA in human osteosarcomas shows sequence variation among Tantigen genes. Int J Cancer 72:791–800 Lopez-Rios F, Illei PB, Rusch V et al, 2004, Evidence against a role for SV40 infection in human mesotheliomas and high risk of false-positive PCR results owing to presence of SV40 sequences in common laboratory plasmids. Lancet 364:1157–1166 Atkin SJ, Griffin BE, Dilworth SM, 2009, Polyoma virus and simian virus 40 as cancer models: history and perspectives. Semin Cancer Biol 19:211–27 Gjoerup O, Chang Y, 2010, Update on human polyomaviruses and cancer. Adv Cancer Res 106:1–51 Cutrone R, Lednicky J, Dunn G et al, 2005, Some oral poliovirus were contaminated with infectious SV40 after 1961. Cancer Res 65:10273–10279 Lednicky JA, Garcea RL, Bergsagel DJ, Butel JS, 1995, Natural simian virus 40 strains are present in human choroid plexus and ependymoma tumors. Virology 212:710–717 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 691 EP 696 DI 10.1007/s12253-012-9496-x PG 6 ER PT J AU Wang, L Yang, M Shan, L Qi, L Chai, C Zhou, Q Yao, K Wu, H Sun, W AF Wang, Lifeng Yang, Miling Shan, Lihui Qi, Lei Chai, Cuicui Zhou, Qiufeng Yao, Ke Wu, Hongmei Sun, Wenguang TI The Role of SPARC Protein Expression in the Progress of Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; SPARC; Angiogenesis; Cancer cells proliferation ID Gastric cancer; SPARC; Angiogenesis; Cancer cells proliferation AB We aimed to investigate the expression of SPARC (secreted protein, acidic and rich in cysteine) in gastric cancer and its relationship with tumor angiogenesis and cancer cells proliferation. Protein expression of SPARC, VEGF, CD34 and Ki-67 in 80 cases of gastric cancer and 30 cases of normal gastric tissue was evaluated by immunohistochemistry. CD34 staining was used as an indicator of microvessel density (MVD). Ki-67 labeling Index (LI) indicated cancer cells proliferation. Statistical analysis was used to investigate its relationship with clinical characteristics, tumor angiogenesis and cancer cells proliferation. SPARC expression was mainly in the stromal cells surrounding the gastric cancer cells, and was statistically significant differences between gastric cancer and normal gastric tissue (P<0.05). Both the expression of SPARC and VEGF were related to differentiation degree, clinical stage, Lauren classification and lymph node metastasis (P<0.05). Expression of SPARC was significantly negatively correlated with the expression of VEGF and MVD in gastric cancer tissues. Expression of SPARC was also negatively correlated with Ki-67-LI. Our findings suggest that both the expression of SPARC and VEGF are closed to tumor angiogenesis in gastric cancer, SPARC inhibited tumor angiogenesis but VEGF promoted tumor angiogenesis. SPARC also inhibited cells proliferation of gastric cancer. C1 [Wang, Lifeng] The First Clinical College of Harbin Medical University, Department of Pathology, 150001 Harbin, China. [Yang, Miling] The First Teaching Hospital, Zhengzhou University, Department of Pathology, 450003 Henan, China. [Shan, Lihui] The First Clinical College of Harbin Medical University, Department of Pathology, 150001 Harbin, China. [Qi, Lei] The First Clinical College of Harbin Medical University, Department of Pathology, 150001 Harbin, China. [Chai, Cuicui] The First Clinical College of Harbin Medical University, Department of Pathology, 150001 Harbin, China. [Zhou, Qiufeng] The First Clinical College of Harbin Medical University, Department of Pathology, 150001 Harbin, China. [Yao, Ke] The First Clinical College of Harbin Medical University, Department of Pathology, 150001 Harbin, China. [Wu, Hongmei] The First Clinical College of Harbin Medical University, Department of Pathology, 150001 Harbin, China. [Sun, Wenguang] The First Clinical College of Harbin Medical University, Department of Nutrition, 150001 Harbin, China. RP Wang, L (reprint author), The First Clinical College of Harbin Medical University, Department of Pathology, 150001 Harbin, China. EM hljwlf@yahoo.cn CR ZhanW, Han F, 2008, Surgical therapy of gastric cancer in china. J Pract Oncol 23:91–93 Klagsbrun MD, Amore PA, 1991, Regulators of angiogenesis. Annu Rev Physiol 53:217–239 Dvorak HF, Brown LF, Detmer MD et al, 1995, Vascualr permeability factor/vascular endothelial growth factor, microvascular hyperpermeability and angiogenesis. Am J Pathol 146:1029–1039 Raines EW, Lane TF, Iruela-Arispe ML et al, 1992, The extracellular glycoprotein SPARC interacts with platelet-derived growth factor(PDGF)-AB and-BB and inbibits the binding of PDGF to its receptors. Proc Natl Acad Sci USA 89:1281–1285 Ledda F, Bravo AI, Adris S et al, 1997, The expression of the secreted protein acidic and rich in cysteine, SPARC, is associated with the neoplastic progression of human melanoma. J Invest Dermatol 108:210–214 Hasselaar P, Sage EH, 1992, SPARC antagonizes the effect of bFGF on the migration of bovine aortic endothelial cells. J Cell Biochem 49:272–283 Zhao Z-S, Wang Yuan-Yu, 2010, SPARC Is Associated with Gastric Cancer Progression and Poor Survival of Patients. Clin Cancer Res 16(1):260–268 Volm M, Koomagi R, Mattern J et al, 1997, Prognostic value of vascular endothelial growth factor and its receptor Flt-1 in squamous cell lung cancer. Int J Cancer 74(1):64–68 Weidner N, Folkman J, Pozza F et al, 1992, Tumor angiogenesis:a new significant independent prognostic indicator in early-stage breast carcinoma. J Natl Cancer Inst 84(9):1875–1887 Ikeguchi M, Cai J, Yamane N et al, 1999, Clinical significance of spontaneous apoptosis in advanced gastric adenocarcinoma [J]. Cancer 85(11):2329–2335 Ellen JE, Sage H, 1996, Regulation of angiogenesis by SPARC and angiostatin: implications for tumor cell biology. Seminars in cancer biology 7:139–146 Yan Q, Sage EH, 1999, SPARC, a matricellular glycoprotein with important biological functions. J Histochem Cytochem 47, 12):1495–1505 Bradshaw AD, Sage EH, 2001, SPARC, a matricellular protein that functions in cellular differentiation and tissue response to injury. J Clin Invest 107(9):1049–1054 Bornstein P, Sage EH, 2002, Matricellular proteins: extracellular modulators of cell function. Curr Opin Cell Biol 14:608– 616 Watkins G, Douglas-Jones A, Bryce R et al, 2005, Increased levels of SPARC, osteonectin, in human breast cancer tissues and its association with clinical outcomes. Prostaglandins Leukot Essent Fatty Acids 72:267–272 Yamanaka M, Kanda K, Li NC et al, 2001, Analysis of the gene expression of SPARC and its prognostic value for bladder cancer. J Urol 166:2495–2499 Koukourakis MI, Giatromanolaki A, Brekken RA et al, 2003, Enhanced Expression of SPARC/Osteonectin in the Tumor associated Stroma of Non Small Cell Lung Cancer Is Correlated with Markers of Hypoxia/Acidity and with Poor Prognosis of Patients. Cancer Res 63:5376–5380 Said N, Frierson JR, Chernauskas D et al, 2009, The role of SPARC in the TRAMP model of prostate carcinogenesis and progression. Oncogene 28(39):3487–3498 Siina J, Arto K, Toru M et al, 2010, Gene Expression Analysis Identifies Over-expression of CXCL1, SPARC, SPP1, and SULF1 in Gastric Cancer. Gene Chromosomes &Cancer 49(1):28–39 Alexandre C, LisaJG RP et al, 2010, Anti-angiogenic SPARC peptides inhibit progression of neuroblastoma tumors. Molecular Cancer 9:138 Shanna AA, Lee BR, Andrew FM et al, 2010, Lack of host SPARC enhances vascular function and tumor spread in an orthotopic murine model of pancreatic carcinoma. Disease Models & Mechanisms 3(1):57–72 Schultz C, Lemke N, Ge S et al, 2002, Secreted protein acidic and rich in cysteine promotes glioma invasion and delays tumor growth in vivo [J]. Cancer Res 62(21):6270–6277 Berse B, Brown LF, Van D, 1992, Vascular permeability factor, vascular endothelial growth factor, gene is expressed differently in normal tissues, macrophages, and tumors. Mol Biol Cell 3:210– 211 Detmar M, Velasco P, Richard L et al, 2000, Expression of vascular endothelial growth factor induces an invasive phenotype in human squamous cell carcinomas. Am J Pathol 156:159– 167 Raspollini MR, Amunni G, Villanucci A et al, 2004, Prognostic significance of microvessel density and vascular endothelial growth factor expression in advanced ovarian serous carcinoma [J]. Int J Gynecol Cancer 14(5):815–823 Bremnes RM, Camps C, Sirera R et al, 2006, Angiogenesis in non-small cell lung cancer: the prognostic impact of neoangiogenesis and the cytokines VEGF and bFGF in tumours and blood. Lung Cancer 51(2):143–158 Yunker CK, Golembieski W, Lemke N et al, 2008, SPARCinduced increase in glioma matrix and decrease in vascularity are associated with reduced VEGF expression and secretion. Int J Cancer 122:2735–2743 Chlenski A, Liu S, Crawford SE et al, 2002, SPARC is a key Schwannian derived inhibitor controlling neuroblastoma tumor angiogenesis. Cancer Res 62:7357–7363 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 697 EP 702 DI 10.1007/s12253-012-9497-9 PG 6 ER PT J AU Livide, G Epistolato, CM Amenduni, M Disciglio, V Marozza, A Mencarelli, AM Toti, P Lazzi, S Hadjistilianou, Th De Francesco, S D’Ambrosio, A Renieri, A Ariani, F AF Livide, Gabriella Epistolato, Carmela Maria Amenduni, Mariangela Disciglio, Vittoria Marozza, Annabella Mencarelli, Antonietta Maria Toti, Paolo Lazzi, Stefano Hadjistilianou, Theodora De Francesco, Sonia D’Ambrosio, Alfonso Renieri, Alessandra Ariani, Francesca TI Epigenetic and Copy Number Variation Analysis in Retinoblastoma by MS-MLPA SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Retinoblastoma; MS-MLPA; Epigenetics; Copy number changes ID Retinoblastoma; MS-MLPA; Epigenetics; Copy number changes AB Retinoblastoma is the most common primary intraocular malignancy in children. Two step inactivation of RB1 (M1-M2) represents the key event in the pathogenesis of retinoblastoma but additional genetic and epigenetic events (M3-Mn) are required for tumor development. In the present study, we employed Methylation Specific Multiplex Ligation Probe Assay to investigate methylation status and copy number changes of 25 and 39 oncosuppressor genes, respectively. This technique was applied to analyse 12 retinoblastomas (5 bilateral and 7 unilateral) and results were compared to corresponding normal retina. We identified hypermethylation in seven new genes: MSH6 (50%), CD44 (42%), PAX5 (42%), GATA5 (25%), TP53 (8%), VHL (8%) and GSTP1 (8%) and we confirmed the previously reported hypermethylation of MGMT (58%), RB1 (17%) and CDKN2 (8%). These genes belong to key pathways including DNA repair, pRB and p53 signalling, transcriptional regulation, protein degradation, cell-cell interaction, cellular adhesion and migration. In the same group of retinoblastomas, a total of 29 copy number changes (19 duplications and 10 deletions) have been identified. Interestingly, we found deletions of the following oncosuppressor genes that might contribute to drive retinoblastoma tumorigenesis: TP53, CDH13, GATA5, CHFR, TP73 and IGSF4. The present data highlight the importance of epigenetic changes in retinoblastoma and indicate seven hypermethylated oncosuppressors never associated before to retinoblastoma pathogenesis. This study also confirms the presence of copy number variations in retinoblastoma, expecially in unilateral cases (mean 3 ±1.3) where these changes were found more frequently respect to bilateral cases (mean 1.4±1.1). C1 [Livide, Gabriella] University of Siena, Department of Biotechnology, Medical GeneticsSiena, Italy. [Epistolato, Carmela Maria] University of Siena, Department of Biotechnology, Medical GeneticsSiena, Italy. [Amenduni, Mariangela] University of Siena, Department of Biotechnology, Medical GeneticsSiena, Italy. [Disciglio, Vittoria] University of Siena, Department of Biotechnology, Medical GeneticsSiena, Italy. [Marozza, Annabella] University of Siena, Department of Biotechnology, Medical GeneticsSiena, Italy. [Mencarelli, Antonietta Maria] University of Siena, Department of Biotechnology, Medical GeneticsSiena, Italy. [Toti, Paolo] University of Siena, Department of Human Pathology and Oncology, Section of PathologySiena, Italy. [Lazzi, Stefano] University of Siena, Department of Human Pathology and Oncology, Section of PathologySiena, Italy. [Hadjistilianou, Theodora] Azienda Ospedaliera Universitaria Senese, Ophthalmological Science and NeuroscienceSiena, Italy. [De Francesco, Sonia] Azienda Ospedaliera Universitaria Senese, Ophthalmological Science and NeuroscienceSiena, Italy. [D’Ambrosio, Alfonso] University of Siena, Pediatrics DepartmentSiena, Italy. [Renieri, Alessandra] University of Siena, Department of Biotechnology, Medical GeneticsSiena, Italy. [Ariani, Francesca] University of Siena, Department of Biotechnology, Medical GeneticsSiena, Italy. RP Renieri, A (reprint author), University of Siena, Department of Biotechnology, Medical Genetics, Siena, Italy. EM renieri@unisi.it CR MacCarthy A, Draper GJ, Steliarova-Foucher E, Kingston JE, 2006, Retinoblastoma incidence and survival in European children, 1978–1997, Report from the Automated Childhood Cancer Information System project. Eur J Cancer 42:2092–2102 Xu XL, Fang Y, Lee TC, Forrest D, Gregory-Evans C, Almeida D, Liu A et al, 2009, Retinoblastoma has properties of a cone precursor tumor and depends upon cone-specific MDM2 signaling. Cell 137:1018–1031 Knudson AG Jr, 1971, Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci U S A 68:820–823 Friend SH, Bernards R, Rogelj S, Weinberg RA, Rapaport JM, Albert DM, Dryja TP, 1986, A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature 323:643–646 Vogel F, 1979, Genetics of retinoblastoma. Hum Genet 52:1–54 Onadim Z, Hogg A, Baird PN, Cowell JK, 1992, Oncogenic point mutations in exon 20 of the RB1 gene in families showing incomplete penetrance andmild expression of the retinoblastoma phenotype. Proc Natl Acad Sci U S A 89:6177–6181 Gallie BL, Campbell C, Devlin H, Duckett A, Squire JA, 1999, Developmental basis of retinal-specific induction of cancer by RB mutation. Cancer Res 59:1731s–1735s Corson TW, Gallie BL, 2007, One hit, two hits, three hits, more? Genomic changes in the development of retinoblastoma. Genes Chromosomes Cancer 46:617–634 Sampieri K, Amenduni M, Papa FT, Katzaki E, Mencarelli MA, Marozza A, Epistolato MC et al, 2009, Array comparative genomic hybridization in retinoma and retinoblastoma tissues. Cancer Sci 100:465–471 Pieretti M, Cavalieri C, Conway PS, Gallion HH, Powell DE, Turker MS, 1995, Genetic alterations distinguish different types of ovarian tumors. Int J Cancer 64:434–440 Herman JG, Latif F, Weng Y, Lerman MI, Zbar B, Liu S, Samid D et al, 1994, Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. Proc Natl Acad Sci U S A 91:9700–9704 Yates DR, Rehman I, Abbod MF, Meuth M, Cross SS, Linkens DA, Hamdy FC et al, 2007, Promoter hypermethylation identifies progression risk in bladder cancer. Clin Cancer Res 13:2046–2053 Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA, Kang GH et al, 2006, CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 38:787–793 Leong KJ, Wei W, Tannahill LA, Caldwell GM, Jones CE, Morton DG, Matthews GM et al, 2011, Methylation profiling of rectal cancer identifies novel markers of early-stage disease. Br J Surg 98:724–734 Harada K, Toyooka S, Maitra A, Maruyama R, Toyooka KO, Timmons CF, Tomlinson GE et al, 2002, Aberrant promoter methylation and silencing of the RASSF1A gene in pediatric tumors and cell lines. Oncogene 21:4345–4349 ChoyKW, Pang CP, To KF, Yu CB, Ng JS, Lam DS, 2002, Impaired expression and promotor hypermethylation of O6-methylguanine- DNA methyltransferase in retinoblastoma tissues. Invest Ophthalmol Vis Sci 43:1344–1349 Choy KW, Pang CP, Fan DS, Lee TC, Wang JH, Abramson DH, Lo KWet al, 2004, Microsatellite instability and MLH1 promoter methylation in human retinoblastoma. Invest Ophthalmol Vis Sci 45:3404–3409 Choy KW, Lee TC, Cheung KF, Fan DS, Lo KW, Beaverson KL, Abramson DH et al, 2005, Clinical implications of promoter hypermethylation in RASSF1A and MGMT in retinoblastoma. Neoplasia 7:200–206 Tosi GM, Trimarchi C, Macaluso M, La Sala D, Ciccodicola A, Lazzi S, Massaro-Giordano M et al, 2005, Genetic and epigenetic alterations of RB2/p130 tumor suppressor gene in human sporadic retinoblastoma: implications for pathogenesis and therapeutic approach. Oncogene 24:5827–5836 Harada K, Toyooka S, Shivapurkar N, Maitra A, Reddy JL, Matta H, Miyajima K et al, 2002, Deregulation of caspase 8 and 10 expression in pediatric tumors and cell lines. Cancer Res 62:5897– 5901 Cohen Y, Merhavi-Shoham E, Avraham RB, Frenkel S, Pe’er J, Goldenberg-Cohen N, 2008, Hypermethylation of CpG island loci of multiple tumor suppressor genes in retinoblastoma. Exp Eye Res 86:201–206 Jeuken JW, Cornelissen SJ, Vriezen M, Dekkers MM, Errami A, Sijben A, Boots-Sprenger SH et al, 2007, MS-MLPA: an attractive alternative laboratory assay for robust, reliable, and semiquantitative detection of MGMT promoter hypermethylation in gliomas. Lab Invest 87:1055–1065 Laird PW, 1997, Oncogenic mechanisms mediated by DNA methylation. Mol Med Today 3:223–229 Esteller M, 2007, Epigenetic gene silencing in cancer: the DNA hypermethylome. Hum Mol Genet 16 Spec No 1:R50-9 Gerson SL, 2004, MGMT: its role in cancer aetiology and cancer therapeutics. Nat Rev Cancer 4:296–307 Jones PA, Laird PW, 1999, Cancer epigenetics comes of age. Nat Genet 21:163–167 Richter S, Vandezande K, Chen N, Zhang K, Sutherland J, Anderson J, Han L et al, 2003, Sensitive and efficient detection of RB1 gene mutations enhances care for families with retinoblastoma. Am J Hum Genet 72:253–269 Nygren AO, Ameziane N, Duarte HM, Vijzelaar RN, Waisfisz Q, Hess CJ, Schouten JP et al, 2005, Methylation-specific MLPA, MS-MLPA): simultaneous detection of CpG methylation and copy number changes of up to 40 sequences. Nucleic Acids Res 33:e128 Pegg AE, Dolan ME, Moschel RC, 1995, Structure, function, and inhibition of O6-alkylguanine-DNA alkyltransferase. Prog Nucleic Acid Res Mol Biol 51:167–223 BignamiM, O'DriscollM, AquilinaG, Karran P, 2000, Unmasking a killer: DNA O(6)-methylguanine and the cytotoxicity of methylating agents. Mutat Res 462:71–82 Dumenco LL, Allay E, Norton K, Gerson SL, 1993, The prevention of thymic lymphomas in transgenic mice by human O6-alkylguanine- DNA alkyltransferase. Science 259:219–222 Becker K, Dosch J, Gregel CM, Martin BA, Kaina B, 1996, Targeted expression of human O(6)-methylguanine-DNA methyltransferase, MGMT, in transgenic mice protects against tumor initiation in two-stage skin carcinogenesis. Cancer Res 56:3244– 3249 Esteller M, Herman JG, 2004, Generating mutations but providing chemosensitivity: the role of O6-methylguanine DNA methyltransferase in human cancer. Oncogene 23:1–8 Toyooka S, Toyooka KO, Maruyama R, Virmani AK, Girard L, Miyajima K, Harada K et al, 2001, DNA methylation profiles of lung tumors. Mol Cancer Ther 1:61–67 Maruyama R, Toyooka S, Toyooka KO, Virmani AK, Zochbauer- Muller S, Farinas AJ, Minna JD et al, 2002, Aberrant promoter methylation profile of prostate cancers and its relationship to clinicopathological features. Clin Cancer Res 8:514–519 Skorpen F, Krokan HE, 1995, The methylation status of the gene for O6-methylguanine-DNA methyltransferase in human Mer+ and Mer- cells. Carcinogenesis 16:1857–1863 Wang L, Zhu D, Zhang C, Mao X, Wang G, Mitra S, Li BF et al, 1997, Mutations of O6-methylguanine-DNA methyltransferase gene in esophageal cancer tissues from Northern China. Int J Cancer 71:719–723 Horsfall MJ, Gordon AJ, Burns PA, Zielenska M, van der Vliet GM, Glickman BW, 1990, Mutational specificity of alkylating agents and the influence of DNA repair. Environ Mol Mutagen 15:107–122 Esteller M, Risques RA, Toyota M, Capella G, Moreno V, Peinado MA, Baylin SB et al, 2001, Promoter hypermethylation of the DNA repair gene O(6)-methylguanine-DNA methyltransferase is associated with the presence of G:C to A:T transition mutations in p53 in human colorectal tumorigenesis. Cancer Res 61:4689–4692 Nakamura M, Watanabe T, Yonekawa Y, Kleihues P, Ohgaki H, 2001, Promoter methylation of the DNA repair gene MGMT in astrocytomas is frequently associated with G:C –>A:T mutations of the TP53 tumor suppressor gene. Carcinogenesis 22:1715–1719 Wolf P, Hu YC, Doffek K, Sidransky D, Ahrendt SA, 2001, O(6)- Methylguanine-DNA methyltransferase promoter hypermethylation shifts the p53 mutational spectrum in non-small cell lung cancer. Cancer Res 61:8113–8117 Esteller M, Toyota M, Sanchez-Cespedes M, Capella G, Peinado MA, Watkins DN, Issa JP et al, 2000, Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is associated with G to A mutations in K-ras in colorectal tumorigenesis. Cancer Res 60:2368–2371 Esteller M, Herman JG, 2002, Cancer as an epigenetic disease: DNA methylation and chromatin alterations in human tumours. J Pathol 196:1–7 Iyer RR, Pluciennik A, Burdett V, Modrich PL, 2006, DNA mismatch repair: functions and mechanisms. Chem Rev 106:302– 323 Jiricny J, 2006, The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol 7:335–346 Jun SH, Kim TG, Ban C, 2006, DNA mismatch repair system. Classical and fresh roles. Febs J 273:1609–1619 Stojic L, Brun R, Jiricny J, 2004, Mismatch repair and DNA damage signalling. DNA Repair, Amst, 3:1091–1101 Hong Z, Jiang J, Hashiguchi K, Hoshi M, Lan L, Yasui A, 2008, Recruitment ofmismatch repair proteins to the site of DNA damage in human cells. J Cell Sci 121:3146–3154 Wu Y, Berends MJ, Mensink RG, Kempinga C, Sijmons RH, van Der Zee AG, Hollema H et al, 1999, Association of hereditary nonpolyposis colorectal cancer-related tumors displaying low microsatellite instability with MSH6 germline mutations. Am J Hum Genet 65:1291–1298 Wijnen J, de LeeuwW,Vasen H, van der KliftH,Moller P, Stormorken A, Meijers-Heijboer H et al, 1999, Familial endometrial cancer in female carriers of MSH6 germline mutations. Nat Genet 23:142–144 Huang J, Kuismanen SA, Liu T, Chadwick RB, Johnson CK, Stevens MW, Richards SK et al, 2001, MSH6 and MSH3 are rarely involved in genetic predisposition to nonpolypotic colon cancer. Cancer Res 61:1619–1623 Berends MJ, Wu Y, Sijmons RH, Mensink RG, van der Sluis T, Hordijk-Hos JM, de Vries EG et al, 2002, Molecular and clinical characteristics of MSH6 variants: an analysis of 25 index carriers of a germline variant. Am J Hum Genet 70:26–37 Goodfellow PJ, Buttin BM, Herzog TJ, Rader JS, Gibb RK, Swisher E, Look K et al, 2003, Prevalence of defective DNA mismatch repair and MSH6 mutation in an unselected series of endometrial cancers. Proc Natl Acad Sci U S A 100:5908–5913 van der Wal JE, Hermsen MA, Gille HJ, Schouten-Van Meeteren NY, Moll AC, Imhof SM, Meijer GA et al, 2003, Comparative genomic hybridisation divides retinoblastomas into a high and a low level chromosomal instability group. J Clin Pathol 56:26–30 Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan-Smith E, Dalton JD, Girtman K et al, 2007, Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature 446:758–764 Cobaleda C, Schebesta A, Delogu A, Busslinger M, 2007, Pax5: the guardian of B cell identity and function. Nat Immunol 8:463– 470 Liu W, Li X, Chu ES, Go MY, Xu L, Zhao G, Li L et al, 2011, Paired box gene 5 is a novel tumor suppressor in hepatocellular carcinoma through interaction with p53 signaling pathway.Hepatology 53:843–853 Sato H, Wang D, Kudo A, 2001, Dissociation of Pax-5 from KI and KII sites during kappa-chain gene rearrangement correlates with its association with the underphosphorylated form of retinoblastoma. J Immunol 166:6704–6710 Matsumura Y, Tarin D, 1992, Significance of CD44 gene products for cancer diagnosis and disease evaluation. Lancet 340:1053– 1058 Mayer B, Jauch KW, Gunthert U, Figdor CG, Schildberg FW, Funke I, Johnson JP, 1993, De-novo expression of CD44 and survival in gastric cancer. Lancet 342:1019–1022 Akiyama Y, Watkins N, Suzuki H, Jair KW, van Engeland M, Esteller M, Sakai H et al, 2003, GATA-4 and GATA-5 transcription factor genes and potential downstream antitumor target genes are epigenetically silenced in colorectal and gastric cancer.Mol Cell Biol 23:8429–8439 Guo M, Akiyama Y, House MG, Hooker CM, Heath E, Gabrielson E, Yang SC et al, 2004, Hypermethylation of the GATA genes in lung cancer. Clin Cancer Res 10:7917–7924 Guo M, House MG, Akiyama Y, Qi Y, Capagna D, Harmon J, Baylin SB et al, 2006, Hypermethylation of the GATA gene family in esophageal cancer. Int J Cancer 119:2078–2083 Hellebrekers DM, Lentjes MH, van den Bosch SM, Melotte V, Wouters KA, Daenen KL, Smits KM et al, 2009, GATA4 and GATA5 are potential tumor suppressors and biomarkers in colorectal cancer. Clin Cancer Res 15:3990–3997 Wen XZ, Akiyama Y, Pan KF, Liu ZJ, Lu ZM, Zhou J, Gu LK et al, 2010, Methylation of GATA-4 and GATA-5 and development of sporadic gastric carcinomas. World J Gastroenterol 16:1201–1208 KatoMV, Shimizu T, Ishizaki K, Kaneko A, Yandell DW, Toguchida J, Sasaki MS, 1996, Loss of heterozygosity on chromosome 17 and mutation of the p53 gene in retinoblastoma. Cancer Lett 106:75–82 Young AP, Schlisio S, Minamishima YA, Zhang Q, Li L, Grisanzio C, Signoretti S et al, 2008, VHL loss actuates a HIF-independent senescence programme mediated by Rb and p400. Nat Cell Biol 10:361–369 LeeWH, Morton RA, Epstein JI, Brooks JD, Campbell PA, Bova GS, Hsieh WS et al, 1994, Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proc Natl Acad Sci U S A 91:11733–11737 Indovina P, Acquaviva A, De Falco G, Rizzo V, Onnis A, Luzzi A, Giorgi F et al, 2009, Downregulation and aberrant promoter methylation of p16INK4A: a possible novel heritable susceptibility marker to retinoblastoma. J Cell Physiol 223:143–150 Sampieri K, Mencarelli MA, Epistolato MC, Toti P, Lazzi S, Bruttini M, De Francesco S et al, 2008, Genomic differences between retinoma and retinoblastoma. Acta Oncol 47:1483–1492 Matsumoto K, Takayama N, Ohnishi J, Ohnishi E, Shirayoshi Y, Nakatsuji N, Ariga H, 2001, Tumour invasion and metastasis are promoted in mice deficient in tenascin-X. Genes Cells 6:1101–1111 Levy P, Ripoche H, Laurendeau I, Lazar V, Ortonne N, Parfait B, LeroyK et al, 2007, Microarray-based identification of tenascin C and tenascin XB, genes possibly involved in tumorigenesis associated with neurofibromatosis type 1. Clin Cancer Res 13:398–407 Laurie NA, Donovan SL, Shih CS, Zhang J, Mills N, Fuller C, Teunisse A et al, 2006, Inactivation of the p53 pathway in retinoblastoma. Nature 444:61–66 Gustmann S, Klein-Hitpass L, Stephan H, Weber S, Bornfeld N, Kaulisch M, Lohmann DR et al, 2011, Loss at chromosome arm 16q in retinoblastoma: confirmation of the association with diffuse vitreous seeding and refinement of the recurrently deleted region. Genes Chromosomes Cancer 50:327–337 Gratias S, Rieder H, Ullmann R, Klein-Hitpass L, Schneider S, Boloni R, Kappler M et al, 2007, Allelic loss in a minimal region on chromosome 16q24 is associated with vitreous seeding of retinoblastoma. Cancer Res 67:408–416 Sato M, Mori Y, Sakurada A, Fujimura S, Horii A, 1998, The Hcadherin, CDH13, gene is inactivated in human lung cancer. Hum Genet 103:96–101 Scolnick DM, Halazonetis TD, 2000, Chfr defines a mitotic stress checkpoint that delays entry into metaphase. Nature 406:430–435 Yu X, Minter-Dykhouse K, Malureanu L, Zhao WM, Zhang D, Merkle CJ, Ward IM et al, 2005, Chfr is required for tumor suppression and Aurora A regulation. Nat Genet 37:401–406 Chakraborty S, Khare S, Dorairaj SK, Prabhakaran VC, Prakash DR, Kumar A, 2007, Identification of genes associated with tumorigenesis of retinoblastoma by microarray analysis. Genomics 90:344–353 Corn PG, Kuerbitz SJ, van Noesel MM, Esteller M, Compitello N, Baylin SB, Herman JG, 1999, Transcriptional silencing of the p73 gene in acute lymphoblastic leukemia and Burkitt's lymphoma is associated with 5′ CpG island methylation. Cancer Res 59:3352–3356 Kawano S, Miller CW, Gombart AF, Bartram CR, Matsuo Y, Asou H, Sakashita A et al, 1999, Loss of p73 gene expression in leukemias/lymphomas due to hypermethylation. Blood 94:1113–1120 Kuramochi M, Fukuhara H, Nobukuni T, Kanbe T, Maruyama T, Ghosh HP, Pletcher M et al, 2001, TSLC1 is a tumor-suppressor gene in human non-small-cell lung cancer. Nat Genet 27:427–430 Ganguly A, Shields CL, 2010, Differential gene expression profile of retinoblastoma compared to normal retina. Mol Vis 16:1292–1303 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 703 EP 712 DI 10.1007/s12253-012-9498-8 PG 10 ER PT J AU Jin, JSh Tsao, TY Sun, PCh Yu, ChP Tzao, Ch AF Jin, Jong-Shiaw Tsao, Tang-Yi Sun, Pei-Chang Yu, Cheng-Ping Tzao, Ching TI SAHA Inhibits the Growth of Colon Tumors by Decreasing Histone Deacetylase and the Expression of Cyclin D1 and Survivin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Histone deacetylase; Histone deacetylase inhibitor; Nude mice; Colon cancer ID Histone deacetylase; Histone deacetylase inhibitor; Nude mice; Colon cancer AB We studied the effects of suberoylanilide hydroxamic acid (SAHA), a histone deacetylase (HDAC) inhibitor, on colon cancer. The expression of HDACs in colorectal cancer specimens and the effects of SAHA on colon cancer cells and tumors of nude mice were assessed. Treatment with SAHA (3 μm) for 72 h induced downregulation of different subtypes of HDAC proteins and also induced acetylation of histone 3 and histone 4. SAHA significantly inhibited the expression of the oncogenic protein c-myc and also increased the expression of the p53 and Rb proteins. The immunohistochemical staining of HDACs, including HDAC1, HDAC2, HDAC3, and HDAC4, was significantly increased in colorectal adenocarcinoma specimens compared to healthy control tissues. In addition, murine studies showed that 100 mg/kg SAHA administered by intraperitoneal injection significantly induced tumor necrosis and inhibited the growth of colon tumors. Immunohistochemistry of the tumor tissues from nude mice revealed that SAHA inhibited the expression of different subtypes of histone deacetylase, the anti-apoptotic proteins cyclin D1, survivin, and also inhibited cell proliferative as determined by Ki67 expression. SAHA inhibited the growth of colon tumors by decreasing histone deacetylases and the expression of cyclin D1 and survivin in nude mice. C1 [Jin, Jong-Shiaw] Tungs’ Taichung Metro Harbor Hospital, Department of PathologyWuqi Township, Taichung County, Taiwan, Republic of China. [Tsao, Tang-Yi] Tungs’ Taichung Metro Harbor Hospital, Department of PathologyWuqi Township, Taichung County, Taiwan, Republic of China. [Sun, Pei-Chang] National Defense Medical Center, Tri-Service General Hospital, Department of PathologyNeihu, Taipei County, Taiwan, Republic of China. [Yu, Cheng-Ping] National Defense Medical Center, Tri-Service General Hospital, Department of PathologyNeihu, Taipei County, Taiwan, Republic of China. [Tzao, Ching] National Defense Medical Center, Tri-Service General Hospital, Division of Thoracic Surgery, Department of Surgery, No. 325, Sec. 2, Cheng-Kung Road, 114 Neihu, Taipei County, Taiwan, Republic of China. RP Tzao, Ch (reprint author), National Defense Medical Center, Tri-Service General Hospital, Division of Thoracic Surgery, Department of Surgery, 114 Neihu, Taiwan, Republic of China. EM doctor.jsjin@gmail.com CR Mayer RJ, 2009, Targeted therapy for advanced colorectal cancermore is not always better. N Eng J Med 360:623–625 WilkoW, Annika R, Silvia N, Aurelia N, Ann-Christin B, Manfred D, Volker G, Boehm M, Thomas B, Carsten D, 2008, Class I histone deacetylase expression has independent prognostic impact in human colorectal cancer: specific role of class I histone deacetylases in vitro and in vivo. Clin Cancer Res 14:1669–1677 Ito K, Adcock IM, 2002, Histone acetylation and histone deacetylation. Mol Biotechnol 20:99–106 Kuo MH, Allis CD, 1998, Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays 20:615–626 Kim DH, Kim M, Kwon HJ, 2003, Histone deacetylase in carcinogenesis and its inhibitors as anti-cancer agents. J Biochem Mol Biol 36:110–119 Marks PA, Richon VM, Breslow R, Rifkind RA, 2001, Histone deacetylase inhibitors as new cancer drugs. Curr Opin Oncol 13:477–483 Luong QT, O’Kelly J, Braunstein GD, Hershman JM, Koeffler HP, 2006, Antitumor activity of suberoylanilide hydroxamic acid against thyroid cancer cell lines in vitro and in vivo. Clin Cancer Res 12:5570–5577 Sun PC, Tzao C, Chen BH, Liu CW, Yu CP, Jin JS, 2010, Suberoylanilide hydroxamic acid induces apoptosis and sub-G1 arrest of 320 HSR colon cancer cells. J Biochem Sci 17:76–85 Jin JS, Hsieh DS, Loh SH, Chen A, Yao CW, Yen CY, 2006, Increasing expression of serine protease matriptase in ovary tumors: tissue microarray analysis of immunostaining score with clinicopathological parameters. Mod Pathol 19:447–452 Jin JS, Wu WY, Lin YF, 2006, Higher expression of epidermal growth factor receptor is associated with extracellular matrix metalloprotease inducer in colorectal adenocarcinoma: tissue microarray analysis of immunostaining score with clinicopathological parameters. Dis Markers 22:309–316 Jung M, 2001, Inhibitors of histone deacetylase as new anticancer agents. Curr Med Chem 8:1505–1511 Lindemann RK, Gabrielli B, Johnstone RW, 2004, Histonedeacetylase inhibitors for the treatment of cancer. Cell Cycle 3:779–788 Truchet I, Jozan S, Baron S, 2008, Estrogen and antiestrogendependent regulation of breast cancer cell proliferation in multicellular spheroids: Influence of cell microenvironment. Int J Oncol 32:1033–1039 Minucci S, Pelicci PG, 2006, Histone deacetylase inhibitors and the promise of epigenetic, and more, treatments for cancer. Nat Rev Cancer 6:38–51 Alessandro V, Cinzia V, Gessica F, Elena CC, Mirko B, Debora R, Prasun C, Chantal P, Raffaele DF, Paola G, Christian S, Stefania DM, 2004, Crystal structure of a eukaryotic zinc-dependent histone deacetylase, human HDAC8, complexed with a hydroxamic acid inhibitor. PNAS 101:15064–15069 Wang S, Yan-Neale Y, Cai R, Alimov I, Cohen D, 2006, Activation of mitochondrial pathway is crucial for tumor selective induction of apoptosis by LAQ824. Cell Cycle 5:1662–1668 Zbar AP, Kennedy PJ, Singh V, 2009, Functional outcome following restorative rectal cancer surgery. Acta Chir Iugosl 56:9–16 Kelly WK, O’Connor OA, Krug LM, 2005, Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J Clin Oncol 23:3923– 3931 Sakajiri S, Kumagai T, Kawamata N, Saitoh T, Said JW, Koeffler HP, 2005, Histone deacetylase inhibitors profoundly decrease proliferation of human lymphoid cancer cell lines. Exp Hematol 33:53–61 Bali P, Pranpat M, Swaby R, 2005, Activity of suberoylanilide hydroxamic acid against human breast cancer cells with amplification of Her-2. Clin Cancer Res 11:6382–6389 Rundall BK, Denlinger CE, Jones DR, 2005, Suberoylanilide hydroxamic acid combined with gemcitabine enhances apoptosis in non-small cell lung cancer. Surgery 138:360–367 Takai N, Desmond JC, Kumagai T, 2004, Histone deacetylase inhibitors have a profound antigrowth activity in endometrial cancer cells. Clin Cancer Res 10:1141–1149 Takai N, Kawamata N, Gui D, Said JW, Miyakawa I, Koeffler HP, 2004, Human ovarian carcinoma cells: histone deacetylase inhibitors exhibit antiproliferative activity and potently induce apoptosis. Cancer 101:2760–2770 Altieri DC, 2006, The case for survivin as a regulator of microtubule dynamics and cell-death decisions. Curr Opin Cell Biol 18:609–615 Noh EJ, Lim DS, Jeong G, Lee JS, 2009, An HDAC inhibitor, trichostatin A, induces a delay at G2/M transition, slippage of spindle checkpoint, and cell death in a transcription-dependent manner. Biochem Biophys Res Commun 378:326–331 Kim DH, Kundu JK, Surh YJ, 2011, Redox modulation of p53: mechanisms and functional significance. Mol Carcinog 50:222– 234 Shen F, Kirmani KZ, Xiao Z, Thirlby BH, Hickey RJ, Malkas LH, 2011, Nuclear protein isoforms: implications for cancer diagnosis and therapy. J Cell Biochem 112:756–760 Smith RA, Tang J, Tudur-Smith C, Neoptolemos JP, Ghaneh P, 2011, Meta-analysis of immunohistochemical prognostic markers in resected pancreatic cancer. Br J Cancer 104:1440–1451 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 713 EP 720 DI 10.1007/s12253-012-9499-7 PG 8 ER PT J AU Krzeslak, A Wojcik-Krowiranda, K Forma, E Jozwiak, P Romanowicz, H Bienkiewicz, A Brys, M AF Krzeslak, Anna Wojcik-Krowiranda, Katarzyna Forma, Ewa Jozwiak, Pawel Romanowicz, Hanna Bienkiewicz, Andrzej Brys, Magdalena TI Expression of GLUT1 and GLUT3 Glucose Transporters in Endometrial and Breast Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glucose transporters; mRNA expression; Protein expression; Breast cancer; Endometrial cancer ID Glucose transporters; mRNA expression; Protein expression; Breast cancer; Endometrial cancer AB Cancer cells have accelerated metabolism and high glucose requirements. The up-regulation of specific glucose transporters may represent a key mechanism by which malignant cells may achieve increased glucose uptake to support the high rate of glycolysis. In present study we analyzed the mRNA and protein expression of GLUT1 and GLUT3 glucose transporters by quantitative real-time polymerase chain reaction (Q-PCR) and Western blotting technique in 76 cases of endometrial carcinoma and 70 cases of breast carcinoma. SLC2A1 and SLCA2A3 mRNAs expression was found, respectively in 100% and 97.4% samples of endometrial cancers and only in 50% and 40% samples of breast cancers. In endometrial cancers GLUT1 and GLUT3 protein expression was identified in 67.1% and 30.3% of cases. Analogously, in breast cancers in 48.7% and 21% of samples, respectively. The results showed that both endometrial and breast poorly differentiated tumors (grade 2 and 3) had significantly higher GLUT1 and GLUT3 expression than well-differentiated tumors (grade 1). Statistically significant association was found between SLCA2A3 mRNA expression and estrogen and progesterone receptors status in breast cancers. GLUT1 has been reported to be involved in the uptake of glucose by endometrial and breast carcinoma cells earlier and the present study determined that GLUT3 expression is also involved. GLUT1 and GLUT3 seem to be important markers in endometrial and breast tumors differentiation. C1 [Krzeslak, Anna] Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, Pomorska 141/143, 90-236 Lodz, Poland. [Wojcik-Krowiranda, Katarzyna] Medical University of Lodz, Department of Gynecological Oncology, Pabianicka 62, 93-509 Lodz, Poland. [Forma, Ewa] Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, Pomorska 141/143, 90-236 Lodz, Poland. [Jozwiak, Pawel] Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, Pomorska 141/143, 90-236 Lodz, Poland. [Romanowicz, Hanna] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Rzgowska 281/289, 93-338 Lodz, Poland. [Bienkiewicz, Andrzej] Medical University of Lodz, Department of Gynecological Oncology, Pabianicka 62, 93-509 Lodz, Poland. [Brys, Magdalena] Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, Pomorska 141/143, 90-236 Lodz, Poland. RP Brys, M (reprint author), Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, 90-236 Lodz, Poland. EM zreg@biol.uni.lodz.pl CR Young CD, Anderson SM, 2008, Sugar and fat—that’s where it’s at: metabolic changes in tumors. Breast Cancer Res 10:202 Ganapathy V, Thangaraju M, Prasad PD, 2009, Nutrient transporters in cancer: relevance to Warburg hypothesis and beyond. Pharmacol Ther 121:29–40 Warburg O, 1930, The metabolizm of tumors. Constable, London Warburg O, 1956, On the origin of cancer cells. Science 123:309– 314 Thorens B, Mueckler M, 2010, Glucose transporters in the 21st Century. Am J Physiol Endocrinol Metab 298:E141–E145 Macheda ML, Rogers S, Best JD, 2005, Molecular and cellular regulation of glucose transporter, GLUT, proteins in cancer. J Cell Physiol 202:654–662 Younes M, Brown RW, Stephenson M, Gondo M, Cagle PT, 1997, Overexpression of Glut1 and Glut3 in stage I nonsmall cell lung carcinoma is associated with poor survival. Cancer 80:1046–1051 Kawamura T, Kusakabe T, Sugino T et al, 2001, Expression of glucose transporter-1 in human gastric carcinoma: association with tumor aggressiveness, metastasis, and patient survival. Cancer 92:634–641 Haber RS, Rathan A, Weiser KR et al, 1998, GLUT1 glucose transporter expression in colorectal carcinoma: a marker for poor prognosis. Cancer 83:34–40 Sakashita M, Aoyama N, Minami R et al, 2001, Glut1 expression in T1 and T2 stage colorectal carcinomas: its relationship to clinicopathological features. Eur J Cancer 37:204–209 Kalir T, Wang BY, Goldfischer M et al, 2002, Immunohistochemical staining of GLUT1 in benign, borderline and malignant ovarian epithelia. Cancer 94:1078–1082 Boado RJ, Black KL, Pardridge WM, 1994, Gene expression of GLUT3 and GLUT1 glucose transporters in human brain tumors. Mol Brain Res 27:51–57 Estilo CL, O-charoenrat P, Talbot S et al, 2009, Oral tongue cancer gene expression profiling: Identification of novel potential prognosticators by oligonucleotide microarray analysis. BMC Cancer 9:11 Ayala FR, Rocha RM, Carvalho KC et al, 2010, GLUT3 as potential prognostic markers for oral squamous cell carcinoma. Molecules 15:2374–2387 Baer S, Casaubon L, Schwartz MR, Marcogliese A, Younes M, 2002, Glut3 expression in biopsy specimens of laryngeal carcinoma is associated with poor survival. Laryngoscope 112:393–396 Matsuzu K, Segade F, Matsuzu U et al, 2004, Differential expression of glucose transporters in normal and pathological thyroid tissue. Thyroid 14:806–812 Tsukioka M, Matsumoto Y, Noriyuki M et al, 2007, Expression of glucose transporters in epithelial ovarian carcinoma: correlation with clinical characteristics and tumor angiogenesis. Oncol Rep 8:361–367 Rudlowski C, Moser M, Becker AJ et al, 2004, GLUT1 mRNA and protein expression in ovarian borderline tumors and cancer. Oncology 66:404–410 Tavassoli FA, Devilee P, 2003, World Health Organization classification of tumours. Pathology & genetics of tumours of the breast and female genital organs, vol 1. IARC, Lyon, pp 217–232 Goldman NA, Katz EB, Glenn AS et al, 2006, GLUT1 and GLUT8 in endometrium and endometrial adenocarcinoma. Mod Pathol 19:1429–1436 Wang BY, Kalir T, Sabo E et al, 2000, Immunohistochemical staining of GLUT1 in benign, hyperplastic, and malignant endometrial epithelia. Cancer 88:2774–2781 Younes M, Brown RW, Mody DR, Fernandez L, Laucirica R, 1995, GLUT1 expression in human breast carcinoma: correlation with known prognostic markers. Anticancer Res 15:2895–2898 Kang SS, Chun YK, Hur MH et al, 2002, Clinical significance of glucose transporter 1, GLUT1, expression in human breast carcinoma. Jpn J Cancer Res 93:1123–1128 Schmidt M, Voelker HU, Kapp M et al, 2010, Glycolytic phenotype in breast cancer: activation of Akt, up-regulation of GLUT1, TKTL1 and down-regulation of M2PK. J Cancer Res Clin Oncol 36:219–225 Ravazoula P, Batistatou A, Aletra C et al, 2003, Immunohistochemical expression of glucose transporter Glut1 and cyclin D1 in breast carcinomas with negative lymph nodes. Eur J Gynaecol Oncol 24:544–546 Younes M, Lechago LV, Somoano JR, Mosharaf M, Lechago J, 1997, Immunohistochemical detection of Glut3 in human tumors and normal tissues. Anticancer Res 17:2747–2750 Godoy A, Ulloa V, Rodriguez F et al, 2006, Differential subcellular distribution of glucose transporters GLUT1-6 and GLUT9 in human cancer: ultrastructural localization of GLUT1 and GLUT5 in breast tumor tissues. J Cell Physiol 207:614–627 Rivenzon-Segal D, Boldin-Adamsky S, Seger D, Seger R, Degani H, 2003, Glycolysis and glucose transporter 1 as markers of response to hormonal therapy in breast cancer. Int J Cancer 107:177–182 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 721 EP 728 DI 10.1007/s12253-012-9500-5 PG 8 ER PT J AU Gasparinho, GM Morgado, S Fonseca, R Chaves, P AF Gasparinho, Gabriela Maria Morgado, Sonia Fonseca, Ricardo Chaves, Paula TI Collision Metastases of Breast and Rectal Carcinoma – A Possible Role for Chemokines Receptors Expression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports C1 [Gasparinho, Gabriela Maria] Instituto Portugues de Oncologia de Lisboa Francisco Gentil, Servico de Anatomia Patologica, Rua Prof. Lima Basto, 1099-023 Lisbon, Portugal. [Morgado, Sonia] Instituto Portugues de Oncologia de Lisboa Francisco Gentil, Servico de Anatomia Patologica, Rua Prof. Lima Basto, 1099-023 Lisbon, Portugal. [Fonseca, Ricardo] Instituto Portugues de Oncologia de Lisboa Francisco Gentil, Servico de Anatomia Patologica, Rua Prof. Lima Basto, 1099-023 Lisbon, Portugal. [Chaves, Paula] Instituto Portugues de Oncologia de Lisboa Francisco Gentil, Servico de Anatomia Patologica, Rua Prof. Lima Basto, 1099-023 Lisbon, Portugal. RP Gasparinho, GM (reprint author), Instituto Portugues de Oncologia de Lisboa Francisco Gentil, Servico de Anatomia Patologica, 1099-023 Lisbon, Portugal. EM mggasparinho@gmail.com CR Sughayer MA, Zakarneh L, Abu-Shakra R, 2009, Collision metastasis of breast and ovarian adenocarcinoma in axillary lymph nodes: a case report and review of the literature. Pathol Oncol Res 15:423–427 Mattioli F, Masoni F, Ponti G, Rossi G, Molteni G, Alicandri- Ciufelli M, Presutti L, 2009, “Collision” metastasis from unknown primary squamous cell carcinoma and papillary microcarcinoma of thyroid presenting as lateral cervical cystic mass. Auris Nasus Larynx 36:372–375 Raman D, Baugher PJ, Thu YM, Richmond A, 2007, Minireview: role of chemokines in tumor growth. Cancer Lett 256:137–165 Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegul E, Zlotnik A, 2001, Involvement of chemokine receptors in breast cancer metastases. Nature 410:50– 56 Kodama J, Hasengaowa KT, Seki N, Matsuo T, Ojima Y, Nakamura K, Hongo A, Hiramatsu Y, 2007, Association of CXCR4 and CCR7 chemokine receptor expression and lymph node metastasis in human cervical cancer. Ann Oncol 18:70–76 Andre F, Cabioglu N, Assi H, Sabourin JC, Delaloge S, Sahin A, Broglio K, Spano JP, Combadiere C, Bucana C, Soria JC, Cristofanilli M, 2006, Expression of chemokine receptors predicts the site of metastatic relapse in patients with axillary node positive primary breast cancer. Ann Oncol 17:945–951 Mizell J, Smith M, Li BDL, Ampli F, Chu QD, 2009, Overexpression of CXCR4 in primary tumor of patients with Her2- negative breast cancer was predictive of a poor disease-free survival: a validation study. Ann Surg Oncol 16:2711–2716 Mburu YK, Wang J, Wood MA, Walker WH, Ferris RL, 2006, CCR7 mediates inflammation-associated tumor progression. Immunol Res 36:61–72 Ghadjar P, Coupland SE, Na IK, Noutsias M, Letsch A, Stroux A, Bauer S, Buhr HJ, Thiel E, Scheibenbogen C, Keilholz U, 2006, Chemokine receptor CCR6 expression level and liver metastases in colorectal cancer. J Clin Oncol 24:1910–1916 Ghadjar P, Rubie C, Aebersold DM, Keilholz U, 2009, Minireview: the chemokine CCL20 and its receptor CCR6 in human malignancy with focus on colorectal cancer. Int J Cancer 125:741–745 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 729 EP 732 DI 10.1007/s12253-011-9443-2 PG 4 ER PT J AU Kulcsar, I Szanto, A Varoczy, L Mehes, G Zeher, M AF Kulcsar, Istvan Szanto, Antonia Varoczy, Laszlo Mehes, Gabor Zeher, Margit TI Hodgkin’s Lymphoma Developed after Autologous Stem Cell Transplantation for Multiple Myeloma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports C1 [Kulcsar, Istvan] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, 4032 Debrecen, Hungary. [Szanto, Antonia] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, 4032 Debrecen, Hungary. [Varoczy, Laszlo] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, 4032 Debrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Zeher, Margit] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, 4032 Debrecen, Hungary. RP Kulcsar, I (reprint author), University of Debrecen, Medical and Health Center, 3rd Department of Medicine, 4032 Debrecen, Hungary. EM istvankulcsar@yahoo.com CR Curtis RE, Travis LB, Rowlings PA, et al., 1999, Risk of lymphoproliferative disorders after bone marrow transplantation: a multi-institutional study. Blood 94: 2208±2216. Lowe T, Bhatia S, Somlo G, 2007, Second malignancies after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 13(10):1121–1134 Gy V, Sebestyen A, Jaray J, Kopper L, 2008, Malignant tumors in the immunocompromised state following organ transplantation. Orvoskepzes 81:235–242 Fend F, Martinez A, Quintanilla-Martinez L et al, 2002, Clonally unrelated Hodgkin’s disease following autologous stem cell transplant for B-cell lymphoma. Br J Haematol 116, 2):329–333 Zambelli A, Lilleri D, Baldanti F, Scelsi M, Villani L, Da Prada GA, 2005, Hodgkin’s disease as unusual presentation of posttransplant lymphoproliferative disorder after autologous hematopoietic cell transplantation for malignant glioma. BMC Cancer 23, 5):109 Ludwig H, Strasser-Weippl K, Schreder M, Zojer N, 2007, Advances in the treatment of hematological malignancies: current treatment approaches in multiple myeloma. Ann Oncol 18(9):64–70 Stojanoski Z, Georgievski B, Cevreska L et al, 2008, Autologous stem-cell transplantation in patients with multiple myeloma. Prilozi 29(1):265–279 Jaffe ES, Zarate-Osorno A, Medeiros J, 1992, The interrelationship of Hodgkin’s disease and non-Hodgkin’s lymphomas— lessons learned from composite and sequential malignancies. Semin Diagn Pathol 9:297–303 Brauninger A, Hansmann ML, Strickler JG, Dummer R, Burg G, Rajewsky K, Kuppers R, 1999, Identification of common germinal-center B-cell precursors in two patients with both Hodgkin’s disease and non-Hodgkin’s lymphoma. N Engl J Med 340:1239–1247 Ohno T, Smir BN, Weisenburger DD, Gascoyne RD, Hinrichs SD, Chan WC, 1998, Origin of the Hodgkin/Reed-Sternberg cells in chronic lymphocytic leukemia with Hodgkin’s transformation. Blood 91:1757–1761 Rowlings PA, Curtis RE, Passweg JR et al, 1999, Increased incidence of Hodgkin’s disease after allogeneic bone marrow transplantation. J Clin Oncol 17(10):3122–3127 Ranganathan S, Webber S, Ahuja S, Jaffe R, 2004, Hodgkin-like posttransplant lymphoproliferative disorder in children: does it differ from posttransplant Hodgkin lymphoma? Pediatr Dev Pathol 7(4):348–360 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 733 EP 736 DI 10.1007/s12253-011-9477-5 PG 4 ER PT J AU Payer, E Miltenyi, Zs Simon, Zs Szabados, L Hegyi, K Mehes, G Illes, AF Payer, Edit Miltenyi, Zsofia Simon, Zsofia Szabados, Lajos Hegyi, Katalin Mehes, Gabor Illes, Arpad TI Uncommon Late Relapse of Angioimmunoblastic T-Cell Lymphoma after 16-Year Remission Period SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports C1 [Payer, Edit] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, 4032 Debrecen, Hungary. [Miltenyi, Zsofia] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, 4032 Debrecen, Hungary. [Simon, Zsofia] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, 4032 Debrecen, Hungary. [Szabados, Lajos] Scanomed Kft., Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Hegyi, Katalin] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Illes, Arpad] University of Debrecen, Medical and Health Center, 3rd Department of Medicine, Moricz Zs. krt. 22, 4032 Debrecen, Hungary. RP Payer, E (reprint author), University of Debrecen, Medical and Health Center, 3rd Department of Medicine, 4032 Debrecen, Hungary. EM payeredit@vipmail.hu CR (1997, A clinical evaluation of the International Lymphoma Study Group classification of non- Hodgkin’s lymphoma: the Non-Hodgkin’s Lymphoma Classification Project. Blood 89: 3909–3918 Mourad N, Mounier N, Briere J, Raffoux E, Delmer A, Feller A, Meijer CJ, Emile JF, Bouabdallah R, Bosly A, Diebold J, Haioun C, Coiffier B, Gisselbrecht C, Gaulard P, 2008, Groupe d'Etude des Lymphomes de l'Adulte: clinical, biologic, and pathologic features in 157 patients with angioimmunoblastic T-cell lymphoma treated within the Groupe d'Etude des Lymphomes de l'Adulte, GELA, trials. Blood 1;111(9): 4463–4470 Freter CE, Cossman J, 1993, Angioimmunoblastic lymphadenopathy with dysproteinemia. Semin Oncol 20:627–635 Brauninger A, Spieker T, Willenbrock K, Gaulard P, Wacker HH, Rajewsky K, Hansmann ML, Kuppers R, 2001, Survival and clonal expansion of mutating ‘forbidden’, immunoglobulin receptor-deficient, Epstein–Barr virus-infected B cells in angioimmunoblastic T cell lymphoma. J Exp Med 194:927–940 Anagnostopoulos I, Hummel M, Finn T, Tiemann M, Korbjuhn P, Dimmler C, Gatter K, Dallenbach F, Parwaresch MR, Stein H, 1992, Heterogeneous Epstein–Barr virus infection patterns in peripheral T-cell lymphoma of angioimmunoblastic lymphadenopathy type. Blood 80:1804–1812 Weiss LM, Jaffe ES, Liu XF, Chen YY, Shibata D, Medeiros LJ, 1992, Detection and localization of Epstein–Barr viral genomes in angioimmunoblastic lymphadenopathy and angioimmunoblastic lymphadenopathy-like lymphoma. Blood 79:1789–1795 Zettl A, Lee SS, Rudiger T, Starostik P, Marino M, Kirchner T, Ott M, Muller-Hermelink HK, Ott G, 2002, Epstein–Barr virus-associated B-cell lymphoproliferative disorders in angloimmunoblastic T-cell lymphoma and peripheral T-cell lymphoma, unspecified. Am J Clin Pathol 117:368–379 Brearley RL, Chapman J, Cullen MH, Horton MA, Stansfeld AG, Waters AH, 1979, Haematological features of angioimmunoblastic lymphadenopathy with dysproteinaemia. J Clin Pathol 32:356–360 Seehafer JR, Goldberg NC, Dicken CH, Su WP, 1980, Cutaneous manifestations of angioimmunoblastic lymphadenopathy. Arch Dermatol 116:41–45 Pieters R, Martens J, Dequeker J, 1982, Rheumatoid arthritis associated with bronchiolitis obliterans and immunoblastic sarcoma. Clin Rheumatol 1:35–40 Tobinai K, Minato K, Ohtsu T, Mukai K, Kagami Y, Miwa M, Watanabe S, Shimoyama M, 1988, Clinicopathologic, immunophenotypic, and immunogenotypic analyses of immunoblastic lymphadenopathy-like T-cell lymphoma. Blood 72:1000–1006 Siegert W, Nerl C, Agthe A, Engelhard M, Brittinger G, Tiemann M, Lennert K, Huhn D, 1995, Angioimmunoblastic lymphadenopathy, AILD)-type T-cell lymphoma: prognostic impact of clinical observations and laboratory findings at presentation. The Kiel Lymphoma Study Group. Ann Oncol 6:659–664 Pautier P, Devidas A, Delmer A, Dombret H, Sutton L, Zini JM, Nedelec G, Molina T, Marolleau JP, Brice P, 1999, Angioimmunoblastic-like T-cell non Hodgkin’s lymphoma: outcome after chemotherapy in 33 patients and review of the literature. Leuk Lymphoma 32:545–552 Ambepitiya GB, 1989, Angioimmunoblastic lymphadenopathy associated with thyroid disease. J Clin Pathol 42:668–669 Hamidou MA, El Kouri D, Audrain M, Grolleau JY, 2001, Systemic antineutrophil cytoplasmic antibody vasculitis associated with lymphoid neoplasia. Ann Rheum Dis 60:293–295 Suchi T, Lennert K, Tu LY, Kikuchi M, Sato E, Stansfeld AG, Feller AC, 1987, Histopathology and immunohistochemistry of peripheral T cell lymphomas: a proposal for their classification. J Clin Pathol 40:995–1015 Attygalle AD, Kyriakou C, Dupuis J, Grogg KL, Diss TC, Wotherspoon AC, Chuang SS, Cabecadas J, Isaacson PG, Du MQ, Gaulard P, Dogan A, 2007, Histologic evolution of angioimmunoblastic T-cell lymphoma in consecutive biopsies: clinical correlationand insights into natural history and disease progression. Am J Surg Pathol 31:1077–1088 Ree HJ, Kadin ME, Kikuchi M, Ko YH, Go JH, Suzumiya J, Kim DS, 1998, Angioimmunoblastic lymphoma, AILD-type T-cell lymphoma, with hyperplastic germinal centers. Am J Surg Pathol 22:643–655 Attygalle A, Al Jehani R, Diss TC, Munson P, Liu H, Du MQ, Isaacson PG, Dogan A, 2002, Neoplastic T cells in angioimmunoblastic T-cell lymphoma express CD10. Blood 99:627–633 Dunleavy K, Wilson WH, Jaffe ES, 2007, Angioimmunoblastic T cell lymphoma: pathobiological insights and clinical implications. Curr Opin Hematol 14:348–353 Siegert W, Nerl C, Meuthen I, Zahn T, Brack N, Lennert K, 1991, Huhn: recombinant human interferon-alpha in the treatment of angioimmunoblastic lymphadenopathy: results in 12 patients. Leukemia 5:892–895 Siegert W, Agthe A, Griesser H, Schwerdtfeger R, Brittinger G, Engelhard M, Kuse R, Tiemann M, Lennert K, Huhn D, 1992, Treatment of angioimmunoblastic lymphadenopathy, AILD)-type T-cell lymphoma using prednisone with or without the COPBLAM ⁄ IMVP-16 regimen. A multicenter study. Kiel Lymphoma Study Group. Ann Intern Med 117:364–370 Gisselbrecht C, Gaulard P, Lepage E, Coiffier B, Briere J, Haioun C, Cazals-Hatem D, Bosly A, Xerri L, Tilly H, Berger F, Bouhabdallah R, Diebold J, 1998, Prognostic significance of T-cell phenotype in aggressive non-Hodgkin’s lymphomas: Groupe d’Etude des Lymphomes de l’Adulte, GELA). Blood 92:76–82 Iannitto E, Ferreri AJ, Minardi V, Tripodo C, Kreipe HH, 2008, Angioimmunoblastic T-cell lymphoma. Crit Rev Oncol Hematol 68(3):264–271 Pizzolo G, Stein H, Josimovic-Alasevic O, Vinante F, Zanotti R, Chilosi M, Feller AC, Diamantstein T, 1990, Increased serum levels of soluble IL-2 receptor, CD30 and CD8 molecules, and gamma-interferon in angioimmunoblastic lymphadenopathy: possible pathogenetic role of immunoactivation mechanisms. Br J Haematol 75:485–488 Takeshita M, Sumiyoshi Y, Masuda Y, Ohshima K, Yoshida T, Kikuchi M, Muller H, 1993, Cytokine, interleukin-1 alpha, interleukin-1 beta, tumor necrosis factor alpha, and interleukin- 6)-possessing cells in lymph nodes of malignant lymphoma. Pathol Res Pract 189:18–25 Foss HD, Anagnostopoulos I, Herbst H, Grebe M, Ziemann K, Hummel M, Stein H, 1995, Patterns of cytokine gene expression in peripheral T-cell lymphoma of angioimmunoblastic lymphadenopathy type. Blood 85:2862–2869 Ohshima KS, Suzumiya J, Kawasaki C, Kanda M, Kikuchi M, 2000, Cytoplasmic cytokines in lymphoproliferative disorders: multiple cytokine production in angioimmunoblastic lymphadenopathy with dysproteinemia. Leuk Lymphoma 38:541–545 Jones D, Fletcher CD, Pulford K, Shahsafaei A, Dorfman DM, 1999, The T-cell activation markers CD30 and OX40 ⁄ CD134 are expressed in nonoverlapping subsets of peripheral T-cell lymphoma. Blood 93:3487–3493 Jones D, O’Hara C, Kraus MD, Perez-Atayde AR, Shahsafaei A, Wu L, Dorfman DM, 2000, Expression pattern of T-cellassociated chemokine receptors and their chemokines correlates with specific subtypes of T-cell non-Hodgkin lymphoma. Blood 96:685–690 Dorfman DM, Shahsafaei A, 2002, CD69 expression correlates with expression of other markers of Th1 T cell differentiation in peripheral T cell lymphomas. Hum Pathol 33:330–334 Dogan A, Attygalle AD, Kyriakou C, 2003, Angioimmunoblastic T-cell lymphoma. Br J Haematol 121(5):681–691 Paloczi K, Suranyi P, Nemes Z, Szegedi Gy, 1986, A study of lymphocyte subsets in patients with angioimmunoblastic lymphadenopathy. 62(4): 615–618 Pizzolo G, Vinante F, Agostini C, Zambello R, Trentin L, Masciarelli M, Chilosi M, Benedetti F, Dazzi F, Todeschini G, 1987, Immunologic abnormalities in angioimmunoblastic lymphadenopathy. Cancer 60:2412–2418 Willenbrock K, Brauninger A, Hansmann ML, 2007, Frequent occurrence of B-cell lymphomas in angioimmunoblastic T-cell lymphoma and proliferation of Epstein-Barr virus-infected cells in early cases. Br J Haematol 138(6):733–739 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2012 VL 18 IS 3 BP 737 EP 741 DI 10.1007/s12253-011-9475-7 PG 5 ER PT J AU Kopper, L AF Kopper, Laszlo TI Denosumab—A Powerful RANKL Inhibitor to Stop Lytic Metastases and Other Bone Loss Actions by Osteoclasts SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Denosumab; RANKL-inhibitor; Osteoclasts; Metastasis ID Denosumab; RANKL-inhibitor; Osteoclasts; Metastasis AB Denosumab is a perfect example on the targeted anticancer therapy. The inhibition of RANKL activity suppressed the osteoclasts’ resorptive function and so prevented skeletal related events. This effect is useful not only against bone metastases, but also in the treatment of other diseases caused by bone loss. In different solid tumors with bone metastasis the quality of life also improved, although the overall survival usually showed no change. On the market the main competitors for denosumab are still the bisphosphonates (questions of costs and reimbursement are not discussed) and some potential new agents e.g. Src kinases (as dasatinib, saracatinib, bosutinib), cathepsin K inhibitors, (e.g. odanacatib), and new selective estrogen receptor modulators (e.g. bazedoxifene, lasofoxifene). Nevertheless, today denosumab is one of the most powerful agents in bone-saving area. C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM kopper@korb1.sote.hu CR Baron R, Ferrari S, Russel RGG, 2011, Denosumab and bisphosphonates: Different mechanisms of action and effects. Bone 48:677–692 Brown JE, Coleman RE, 2011, Denosumab in patients with cancer —a surgical strike against the osteoclast. Nature Rev Clin Oncol 9:110–118 Martin T, Gooi JH, Sims NA, 2009, Molecular mechanisms in coupling of bone formation to resorption. Crit Rev Eukaryot Gene Expr 19:73–88 Bruzzaniti A, Baron R, 2006, Molecular regulation of osteoclast activity. Rev J Endocr Metab Disord 7:123–139 Theoleyre S, Wittrant Y, Tat SK et al, 2004, The molecular triad OPR/RANK/RANKL involvement in the orchestration of pathophysiological bone remodeling. Cytokine Growth Factor Rev 15:457–475 Weight HL, McCarthy HS, Middleton J, Marshall MJ, 2009, RANK, RANKL, and osteoprotegerin in bone biology and disease. Curr Rev Musculoskelet Med 2:56–64 Pederson L, Ruan M, Wetendorf JJ et al, 2008, Regulation of bone formationby osteoclasts involvesWnt/BMP signaling and the chemokine sphingosine-1-phosphate. Proc Natl Acad Sci USA 105:20764–20769 Sims N, 2010, EPHs and ephrins: many pathways to regulate osteoblasts and osteoclasts. IBMS BoneKEy 7:304–313 Dougall WC, Chaisson M, 2006, The RANK/RANKL/OPG triad in cancer induced bone diseases. Cancer Metast Rev 25:541–549 Akiyama T, Choong PF, Dass CR, 2010, RANK-Fc inhibits malignancy via inhibiting ERK activation and evoking caspase3- mediated anoikis in human osteosarcoma cells. Clin Exp Metast 27:207–215 Akiyama T et al, 2010, Systemic RANK-Fc protein therapy is efficacious against primary osteosarcoma growth in murine model via activity against osteoclasts. J Pharm Pharmacol 62:470–476 Russell RG, Watts NB, Ebetino FH, Rogers MJ, 2008, Mechanisms of action of bisphosphonates: similarities and differences and their potential influence on clinical efficacy. Osteoporos Int 19:733–759 Kostenuik P, Nguyen HG, McCabe J et al, 2009, Denosumab, a fully human monoclonal antibody to RNAKL inhibits bone resorption and increases BMD in knock-in mice that express chimeric, murine/human, RANKL. J Bone Miner Res 24:182–195 Weinstein RS, Roberson PK, Manolagas SC, 2009, Giant osteoclast formation and long-term oral bisphosphonate therapy. N Engl J Med 360:53–62 Bekker PJ, Holloway DI, Rasmussen AS et al, 2004, A singledose placebo-controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res 19:1059–1066 Lin JH, 1996, Bisphosphonates: a review of their pharmacokinetic properties. Bone 18:85–95 Cremers S, Sparidqans R et al, 2002, A pharmacokinetic and pharmacodynamic model for intravenous bisphosphonate, pamidronate, in osteoporosis. Eur J Clin Pharmacol 57:883–8890 Body JJ et al, 2003, A phase I study of ANGN-0007, a recombinant osteoprotegerin construct, in patients with multiple myeloma or breast carcinoma related bone metastases. Cancer 97(Suppl 3):887–892 Body JJ et al, 2006, Study of the biological receptor activator of nuclear factor kB ligand inhibitor, denosumab, in patients with multiple myeloma or bone metastases from breast cancer. Clin Cancer Res 12:1221–1228 Lipton A et al, 2007, Randomized, active-controlled phase II study of denosumab efficacy and safety in patients with breast cancerrelated bone metastases. J Clin Oncol 25:4431–4437 Ellis GK et al, 2008, Randomized trial of denosumab in patients receiving adjuvant aromatase inhibitors for non-metastatic breast cancer. J Clin Oncol 26:4875–4882 Shahinian VB, Kuo YF, Freeman JL, Goodwin JS, 2005, Risk of fracture after androgen deprivation for prostate cancer. N Engl J Med 352:154–164 Smith MR et al, 2006, Risk of clinical fractures after gonadotropin-releasing hormone agonist therapy for prostate cancer. J Urol 136–139 Smith MR et al, 2009, Denosumab in men receiving androgendeprivation therapy for prostate cancer. N Engl J Med 361:745– 755 Smith MR et al Denosumab and bone-metastasis-free survival in men with castration-resistant prostate cancer: results of a phase III, randomized, placebo-controlled trial. Lancet., DOI 10.1016/S0140- 6736(11)61226-9 Brodowitz T, O’Byrne K, Manegold C Bone matters in lung cancer. Ann Oncol., DOI 10.1093/annoncol/mds009 Fizazi K et al, 2009, Randomized phase II trial of denosunab in patients with bone metastases from prostate cancer, breast cancer, or other neoplasms after intravenous bisphosphonates. J Clin Oncol 27:1564–1571 Fizazi K, Carducci M, Smith M et al, 2011, Denosumab versus zoledronic acid for the treatment of bone metastases in men with castration-resistant prostate cancer. A randomised, double-blind study. The Lancet 377:813–822 Stopeck AT, Lipton A, Body J-J et al, 2010, Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J Clin Oncol 28:5132–5139 Henry DH et al, 2011, A randomized, double-blind study of denosumab versus zoledronic acid in the treatment of bone metastasis in patients with advanced cancer, excluding breast and prostate cancer, or multiple myeloma. J Clin Oncol 29:1125– 1132 Huang L, Xu J, Wood DJ, Zheng MH, 2000, Gene expression of osteoprotegerin ligand, osteoprotegerin, and receptor activator of NF-kB in giant cell tumour of bone: possible involvement in tumour-cell induced osteoclast-like cell formation. Am J Pathol 156:761–767 Thomas D, Henshaw R, Skubicz K et al, 2010, Denosumab in patients with giant-cell tumour of bone: an open-label, phase 2 study. Lancet Oncol 11:275–280 Saylor PJ, 2011, Denosumab –a new option for solid tumors metastatic to bone. Nature Rev Clin Oncol 8:322–324 Hofbauer LC, Rachner DT, Harnann C, 2011, From bone to breast and back—the bone cytokine RANKL and breast cancer. Breast Cancer Rev 13:107–108 Smith MR, Sand F, Colemann R et al, 2012, Denosumab and bone-metastasis-free survival in men with castration-rersistant prostate cancer: results of a phase 3, randomised, placebocontrolled trial. The Lancet 379:3946 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 743 EP 747 DI 10.1007/s12253-012-9538-4 PG 5 ER PT J AU Jokai, H Marschalko, M Csomor, J Szakonyi, J Kontar, O Barna, G Karpati, S Hollo, P AF Jokai, Hajnalka Marschalko, Marta Csomor, Judit Szakonyi, Jozsef Kontar, Orsolya Barna, Gabor Karpati, Sarolta Hollo, Peter TI Tissue-Specific Homing of Immune Cells in Malignant Skin Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Lymphocyte homing receptors; Cutaneous lymphocyte associated antigen; Malignant melanoma; Basal cell carcinoma; Squamous cell carcinoma; Cutaneous T-cell lymphoma ID Lymphocyte homing receptors; Cutaneous lymphocyte associated antigen; Malignant melanoma; Basal cell carcinoma; Squamous cell carcinoma; Cutaneous T-cell lymphoma AB Tissue-specific migration of immune cells involved both in physiological and pathological immune responses is a current research subject for medical science. Several homing molecules have been identified orchestrating extravasation of immune cells to certain peripheral nonlymphoid tissues such as gut, lung and skin. Regarding lymphocyte homing to skin, the first-line defense of human body cutaneous lymphocyte associated antigen (CLA) and a group of chemokine-chemokine receptor pairs are considered to be of crucial importance. The aim of the present review is to summarize existing knowledge about skin- and tumor-specific migration of immune cells playing a major pathogenetic role in host immune responses induced by nonlymphoid malignant skin tumors as well as in the development of primary cutaneous T-cell lymphomas (CTCL). Melanoma malignum, squamous and basal cell carcinoma evoke host immune responses and consequently a subset of reactive immune cells is recruited to site of the tumor. Regarding migratory process and exact functional role of these cells a growing number of data is available in literature. On the other hand tissue-specific immune cell homing is regarded as a key process in the pathogenesis of CTCL where malignant T-lymphocytes can be found in circulation and symptomatic skin. Hereby homing mechanism of malignant T-cells in mycosis fungoides and Sezary-syndrome as separate clinical entities of CTCL is discussed. A precise insight into the molecular background of skin- and tumor-specific immune cell migration can contribute to developing efficient vaccine therapies in non-lymphoid malignant skin tumors and beneficial treatment modalities in CTCL. C1 [Jokai, Hajnalka] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria u. 41, 1085 Budapest, Hungary. [Marschalko, Marta] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria u. 41, 1085 Budapest, Hungary. [Csomor, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Szakonyi, Jozsef] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria u. 41, 1085 Budapest, Hungary. [Kontar, Orsolya] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria u. 41, 1085 Budapest, Hungary. [Barna, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Karpati, Sarolta] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria u. 41, 1085 Budapest, Hungary. [Hollo, Peter] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria u. 41, 1085 Budapest, Hungary. RP Jokai, H (reprint author), Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, 1085 Budapest, Hungary. EM jokaihajnalka@gmail.com CR Kunkel EJ, Butcher EC, 2002, Chemokines and the tissuespecific migration of lymphocytes. Immunity 16:1–4 Hart AL, Ng SC, Mann E, Al-Hassi HO, Bernardo D, Knight SC, 2010, Homing of immune cells: role in homeostasis and intestinal inflammation. Inflamm Bowel Dis 16:1969–1977 Lalor PF, Curbishley SM, Adams DH, 2010, Identifying homing interactions in T-cell traffic in human disease. Methods Mol Biol 616:231–252 Villablanca EJ, Russo V, Mora JR, 2008, Dendritic cell migration and lymphocyte homing imprinting. Histol Histopathol 23:897– 910 Woodland DL, Kohlmeier JE, 2009, Migration, maintenance and recall of memory T cells in peripheral tissues. Nat Rev Immunol 9:153–161 Sigmundsdottir H, Butcher EC, 2008, Environmental cues, dendritic cells and the programming of tissue-selective lymphocyte trafficking. Nat Immunol 9:981–987 Nestle FO, Di Meglio P, Qin JZ, Nickoloff BJ, 2009, Skin immune sentinels in health and disease. Nat Rev Immunol 9:679–691 Clark RA, 2010, Skin-resident T cells: the ups and downs of on site immunity. J Invest Dermatol 130(2):362–370 Sigmundsdottir H, 2010, Improving topical treatments for skin diseases. Trends Pharmacol Sci 31:239–245 Fuhlbrigge RC, King SL, Sackstein R, Kupper TS, 2006, CD43 is a ligand for E-selectin on CLA+human Tcells. Blood 107:1421– 1426 Magro CM, Dyrsen ME, 2008, Cutaneous lymphocyte antigen expression in benign and neoplastic cutaneous B- and T-cell lymphoid infiltrates. J Cutan Pathol 35:1040–1049 Ni Z, Walcheck B, 2009, Cutaneous lymphocyte-associated antigen, CLA, T cells up-regulate P-selectin ligand expression upon their activation. Clin Immunol 133:257–264 Yamanaka K, Dimitroff CJ, Fuhlbrigge RC, Kakeda M, Kurokawa I, Mizutani H, Kupper TS, 2008, Vitamins A and D are potent inhibitors of cutaneous lymphocyte-associated antigen expression. J Allergy Clin Immunol 121:148–157.e3 Baeke F, Takiishi T, Korf H, Gysemans C, Mathieu C, 2010, Vitamin D: modulator of the immune system. Curr Opin Pharmacol 10:482–496 Duhen T, Geiger R, Jarrossay D, Lanzavecchia A, Sallusto F, 2009, Production of interleukin 22 but not interleukin 17 by a subset of human skin-homing memory T cells. Nat Immunol 10:857–863 Zlotnik A, Yoshie O, 2000, Chemokines: a new classification system and their role in immunity. Immunitiy 12:121–127 Sabat R, Philipp S, Hoflich C, Kreutzer S, Wallace E, Asadullah K, Volk HD, Sterry W, Wolk K, 2007, Immunopathogenesis of psoriasis. Exp Dermatol 16:779–798 Clark RA, Chong B, Mirchandani N, Brinster NK, Yamanaka K, Dowgiert RK, Kupper TS, 2006, The vast majority of CLA+ T cells are resident in normal skin. J Immunol 176:4431– 4439 Santamaria Babi LF, Moser R, Perez Soler MT, Picker LJ, Blaser K, Hauser C, 1995, Migration of skin-homing T cells across cytokine-activated human endothelial cell layers involves interaction of the cutaneous lymphocyte-associated antigen, CLA), the very late antigen-4, VLA-4), and the lymphocyte functionassociated antigen-1, LFA-1). J Immunol 154:1543–1550 Pals ST, de Gorter DJ, Spaargaren M, 2007, Lymphoma dissemination: the other face of lymphocyte homing. Blood 110:3102– 3111 Gelb AB, Smoller BR, Warnke RA, Picker LJ, 1993, Lymphocytes infiltrating primary cutaneous neoplasms selectively express the cutaneous lymphocyte-associated antigen, CLA). Am J Pathol 142:1556–1564 Clark WH Jr, From L, Bernardino EA, Mihm MC, 1969, The histogenesis and biologic behavior of primary human malignant melanomas of the skin. Cancer Res 29:705–727 Clemente CG, Mihm MC Jr, Bufalino R, Zurrida S, Collini P, Cascinelli N, 1996, Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer 77:1303–1310 Mullins IM, Slingluff CL, Lee JK, Garbee CF, Shu J, Anderson SG, Mayer ME, KnausWA, Mullins DW, 2004, CXC chemokine receptor 3 expression by activated CD8+ T cells is associated with survival in melanoma patients with stage III disease. Cancer Res 64:7697–7701 Day CL Jr, Sober AJ, Kopf AW, Lew RA, Mihm MC Jr, Hennessey P, Golomb FM, Harris MN, Gumport SL, Raker JW, Malt RA, Cosimi AB, Wood WC, Roses DF, Gorstein F, Postel A, Grier WR, Mintzis MN, Fitzpatrick TB, 1981, A prognostic model for clinical stage I melanoma of the upper extremity. The importance of anatomic subsites in predicting recurrent disease. Ann Surg 193:436–440 Tuthill RJ, Unger JM, Liu PY, Flaherty LE, Sondak VK, 2002, Risk assessment in localized primary cutaneous melanoma: a Southwest Oncology Group study evaluating nine factors and a test of the Clark logistic regression prediction model. Am J Clin Pathol 118:504–511 Oble DA, Loewe R, Yu P, Mihm MC Jr, 2009, Focus on TILs: prognostic significance of tumor infiltrating lymphocytes in human melanoma. Cancer Immun 9:3 Taylor RC, Patel A, Panageas KS, Busam KJ, Brady MS, 2007, Tumor-infiltrating lymphocytes predict sentinel lymph node positivity in patients with cutaneous melanoma. J Clin Oncol 25:869–875 Prieto PA, Durflinger KH, Wunderlich JR, Rosenberg SA, Dudley ME, 2010, Enrichment of CD8+ cells from melanoma tumorinfiltrating lymphocyte cultures reveals tumor reactivity for use in adoptive cell therapy. J Immunother 33:547–556 Viguier M, Lemaitre F, Verola O, Cho MS, Gorochov G, Dubertret L, Bachelez H, Kourilsky P, Ferradini L, 2004, Foxp3 expressing CD4+CD25(high, regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells. J Immunol 173:1444–1453 Weishaupt C, Munoz KN, Buzney E, Kupper TS, Fuhlbrigge RC, 2007, T-cell distribution and adhesion receptor expression in metastatic melanoma. Clin Cancer Res 13:2549–2556 Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME, 2008, Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer 8:299–308 Labarriere N, Dreno B, Jotereau F, 2008, Lymphocyte biomarkers of clinical responses to adoptive immunotherapy of malignant melanoma. Curr Cancer Ther Rev 4:125–129 Gattinoni L, Powell DJ Jr, Rosenberg SA, Restifo NP, 2006, Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol 6:383–393 Adams DH, Yannelli JR, Newman W, Lawley T, Ades E, Rosenberg SA, Shaw S, 1997, Adhesion of tumour-infiltrating lymphocytes to endothelium: a phenotypic and functional analysis. Br J Cancer 75:1421–1431 Rohde D, Schluter-Wigger W, Mielke V, von den Driesch P, von Gaudecker B, Sterry W, 1992, Infiltration of both T cells and neutrophils in the skin is accompanied by the expression of endothelial leukocyte adhesion molecule-1, ELAM-1): an immunohistochemical and ultrastructural study. J Invest Dermatol 98:794–799 Schadendorf D, Heidel J, Gawlik C, Suter L, Czarnetzki BM, 1995, Association with clinical outcome of expression of VLA-4 in primary cutaneous malignant melanoma as well as Pselectin and E-selectin on intratumoral vessels. J Natl Cancer Inst 87:366–371 Nooijen PT,Westphal JR, Eggermont AM, Schalkwijk C, Max R, de Waal RM, Ruiter DJ, 1998, Endothelial P-selectin expression is reduced in advanced primary melanoma and melanoma metastasis. Am J Pathol 152:679–682 Quezada SA, Peggs KS, Simpson TR, Shen Y, Littman DR, Allison JP, 2008, Limited tumor infiltration by activated T effector cells restricts the therapeutic activity of regulatory T cell depletion against established melanoma. J Exp Med 205:2125– 2138 Walshe TE, Dole VS, Maharaj AS, Patten IS, Wagner DD, D'Amore PA, 2009, Inhibition of VEGF or TGF-{beta} signaling activates endothelium and increases leukocyte rolling. Arterioscler Thromb Vasc Biol 29:1185–1192 Walshe TE, 2010, TGF-beta and microvessel homeostasis. Microvasc Res 80:166–173 Kiss J, Timar J, Somlai B, Gilde K, Fejos Z, Gaudi I, Ladanyi A, 2007, Association of microvessel density with infiltrating cells in human cutaneous malignant melanoma. Pathol Oncol Res 13:21–31 Barton GM, 2008, A calculated response: control of inflammation by the innate immune system. J Clin Invest 118:413–420 Krieg C, Boyman O, 2009, The role of chemokines in cancer immune surveillance by the adaptive immune system. Semin Cancer Biol 19:76–83 Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, McKee M, Gajewski TF, 2009, Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res 69:3077–3085 Navarini-Meury AA, Conrad C, 2009, Melanoma and innate immunity–aActive inflammation or just erroneous attraction? Melanoma as the source of leukocyte-attracting chemokines. Semin Cancer Biol 19:84–91 Richmond A, Yang J, Su Y, 2009, The good and the bad of chemokines/chemokine receptors in melanoma. Pigment Cell Melanoma Res 22:175–186 Somasundaram R, Herlyn D, 2009, Chemokines and the microenvironment in neuroectodermal tumor-host interaction. Semin Cancer Biol 19:92–96 Strieter RM, Belperio JA, Phillips RJ, Keane MP, 2004, CXC chemokines in angiogenesis of cancer. Semin Cancer Biol 14:195–200 Strieter RM, Burdick MD, Gomperts BN, Belperio JA, Keane MP, 2005, CXC chemokines in angiogenesis. Cytokine Growth Factor Rev 16:593–609 Cole KE, Strick CA, Paradis TJ, Ogborne KT, Loetscher M, Gladue RP, Lin W, Boyd JG, Moser B, Wood DE, Sahagan BG, Neote K, 1998, Interferon-inducible T cell alpha chemoattractant, I-TAC): a novel non-ELR CXC chemokine with potent activity on activated T cells through selective high affinity binding to CXCR3. J Exp Med 187:2009–2021 Kunz M, Toksoy A, Goebeler M, Engelhardt E, Brocker E, Gillitzer R, 1999, Strong expression of the lymphoattractant CX- C chemokine Mig is associated with heavy infiltration of T cells in human malignant melanoma. J Pathol 189:552–558 Graves DT, Barnhill R, Galanopoulos T, Antoniades HN, 1992, Expression of monocyte chemotactic protein-1 in human melanoma in vivo. Am J Pathol 140:9–14 Mellado M, de Ana AM, Moreno MC, Martinez C, Rodriguez- Frade JM, 2001, A potential immune escape mechanism by melanoma cells through the activation of chemokine-induced T cell death. Curr Biol 11:691–696 Fushimi T, Kojima A, Moore MA, Crystal RG, 2000, Macrophage inflammatory protein 3alpha transgene attracts dendritic cells to established murine tumors and suppresses tumor growth. J Clin Invest 105:1383–1393 Brose MS, Volpe P, Feldman M, Kumar M, Rishi I, Gerrero R, Einhorn E, Herlyn M, Minna J, Nicholson A, Roth JA, Albelda SM, Davies H, Cox C, Brignell G, Stephens P, Futreal PA, Wooster R, Stratton MR, Weber BL, 2002, BRAF and RAS mutations in human lung cancer and melanoma. Cancer Res 62:6997–7000 Robertson GP, 2005, Functional and therapeutic significance of Akt deregulation in malignant melanoma. Cancer Metastasis Rev 24:273–285 Hardy KM, Kirschmann DA, Seftor EA, Margaryan NV, Postovit LM, Strizzi L, Hendrix MJ, 2010, Regulation of the embryonic morphogen Nodal by Notch4 facilitates manifestation of the aggressive melanoma phenotype. Cancer Res 70:10340–10350 Yang CH, Fan M, Slominski AT, Yue J, Pfeffer LM, 2010, The role of constitutively activated STAT3 in B16 melanoma cells. Int J Interferon Cytokine Mediator Res 2010:1–7 Clark RA, Chong BF, Mirchandani N, Yamanaka K, Murphy GF, Dowgiert RK, Kupper TS, 2006, A novel method for the isolation of skin resident T cells from normal and diseased human skin. J Invest Dermatol 126:1059–1070 Clark RA, Huang SJ, Murphy GF,Mollet IG, Hijnen D,Muthukuru M, Schanbacher CF, Edwards V, Miller DM, Kim JE, Lambert J, Kupper TS, 2008, Human squamous cell carcinomas evade the immune response by down-regulation of vascular E-selectin and recruitment of regulatory T cells. J Exp Med 205:2221–2234 Hald J, Rasmussen N, Claesson MH, 1995, Tumour-infiltrating lymphocytes mediate lysis of autologous squamous cell carcinomas of the head and neck. Cancer Immunol Immunother 41:243– 250 Wei S, Kryczek I, Zou W, 2006, Regulatory T-cell compartmentalization and trafficking. Blood 108:426–431 Dummer R, Urosevic M, Kempf W, Hoek K, Hafner J, Burg G, 2003, Imiquimod in basal cell carcinoma: how does it work? Br J Dermatol 149(Suppl 66):57–58 Urosevic M, Maier T, Benninghoff B, Slade H, Burg G, Dummer R, 2003, Mechanisms underlying imiquimod-induced regression of basal cell carcinoma in vivo. Arch Dermatol 139:1325–1332 Verhaegh M, Beljaards R, Veraart J, Hoekzema R, Neumann M, 1998, Adhesion molecule expression in basal cell carcinoma. Eur J Dermatol 8:252–255 Kooy AJ, Prens EP, Van Heukelum A, Vuzevski VD, Van Joost T, Tank B, 1999, Interferon-gamma-induced ICAM-1 and CD40 expression, complete lack of HLA-DR and CD80, B7.1), and inconsistent HLA-ABC expression in basal cell carcinoma: a possible role for interleukin-10? J Pathol 187:351–357 Kooy AJ, Tank B, Vuzevski VD, van Joost T, Prens EP, 1998, Expression of interferongamma receptors and interferon-gammainduced up-regulation of intercellular adhesion molecule-1 in basal cell carcinoma; decreased expression of IFNgamma R and shedding of ICAM-1 as a means to escape immune surveillance. J Pathol 184:169–176 Kovach BT, Stasko T, 2005, Use of topical immunomodulators in organ transplant recipients. Dermatol Ther 18:19–27 Barnetson RS, Satchell A, Zhuang L, Slade HB, Halliday GM, 2004, Imiquimod induced regression of clinically diagnosed superficial basal cell carcinoma is associated with early infiltration by CD4 T cells and dendritic cells. Clin Exp Dermatol 29:639–643 De Giorgi V, Salvini C, Chiarugi A, Paglierani M, Maio V, Nicoletti P, Santucci M, Carli P, Massi D, 2009, In vivo characterization of the inflammatory infiltrate and apoptotic status in imiquimod-treated basal cell carcinoma. Int J Dermatol 48:312–321 Salasche S, 2002, Imiquimod 5 % cream: a new treatment option for basal cell carcinoma. Int J Dermatol 41(Suppl 1):16–20 Kaporis HG, Guttman-Yassky E, Lowes MA, Haider AS, Fuentes-Duculan J, Darabi K, Whynot-Ertelt J, Khatcherian A, Cardinale I, Novitskaya I, Krueger JG, Carucci JA, 2007, Human basal cell carcinoma is associated with Foxp3+ T cells in a Th2 dominant microenvironment. J Invest Dermatol 127:2391–2398 Kim EJ, Hess S, Richardson SK, Newton S, Showe LC, Benoit BM, Ubriani R, Vittorio CC, Junkins-Hopkins JM, Wysocka M, Rook AH, 2005, Immunopathogenesis and therapy of cutaneous T cell lymphoma. J Clin Invest 115:798–812 Berger CL, Tigelaar R, Cohen J, Mariwalla K, Trinh J, Wang N, Edelson RL, 2005, Cutaneous T-cell lymphoma: malignant proliferation of T-regulatory cells. Blood 105:1640–1647 Clark RA, 2009, Regulation gone wrong: a subset of Sezary patients have malignant regulatory T cells. J Invest Dermatol 129:2747–2750 Chong BF, Wilson AJ, Gibson HM, Hafner MS, Luo Y, Hedgcock CJ, Wong HK, 2008, Immune function abnormalities in peripheral blood mononuclear cell cytokine expression differentiates stages of cutaneous T-cell lymphoma/mycosis fungoides. Clin Cancer Res 14:646–653 Beyer M, Mobs M, Humme D, Sterry W, 2011, Pathogenesis of mycosis fungoides. J Dtsch Dermatol Ges 9:594–598 Heid JB, Schmidt A, Oberle N, Goerdt S, Krammer PH, Suri-Payer E, Klemke CD, 2009, FOXP3+CD25- tumor cells with regulatory function in Sezary syndrome. J Invest Dermatol 129:2875–2885 Capriotti E, Vonderheid EC, Thoburn CJ,Wasik MA, Bahler DW, Hess AD, 2008, Expression of T-plastin, FoxP3 and other tumorassociated markers by leukemic T-cells of cutaneous T-cell lymphoma. Leuk Lymphoma 49:1190–1201 Krejsgaard T, Gjerdrum LM, Ralfkiaer E, Lauenborg B, Eriksen KW, Mathiesen AM, Bovin LF, Gniadecki R, Geisler C, Ryder LP, Zhang Q,WasikMA, Odum N,Woetmann, 2008, AMalignant Tregs express low molecular splice forms of FOXP3 in Sezary syndrome. Leukemia 22:2230–2239 Bouaziz JD, Remtoula N, Bensussan A, Marie-Cardine A, Bagot M, 2010, Absolute CD3+ CD158k+lymphocyte count is reliable and more sensitive than cytomorphology to evaluate blood tumor burden in Sezary syndrome. Br J Dermatol 162:123–128 Campbell JJ, Clark RA, Watanabe R, Kupper TS, 2010, Sezary syndrome and mycosis fungoides arise from distinct T-cell subsets: a biologic rationale for their distinct clinical behaviors. Blood 116:767–771 Notohamiprodjo M, Segerer S, Huss R, Hildebrandt B, Soler D, Djafarzadeh R, Buck W, Nelson PJ, von Luettichau I, 2005, CCR10 is expressed in cutaneous T-cell lymphoma. Int J Cancer 115:641–647 Hwang ST, Janik JE, Jaffe ES, Wilson WH, 2008, Mycosis fungoides and Sezary syndrome. Lancet 371:945–957 Narducci MG, Scala E, Bresin A, Caprini E, Picchio MC, Remotti D, Ragone G, Nasorri F, Frontani M, Arcelli D, Volinia S, Lombardo GA, Baliva G, Napolitano M, Russo G, 2006, Skin homing of Sezary cells involves SDF-1-CXCR4 signaling and down-regulation of CD26/dipeptidylpeptidase IV. Blood 107:1108–1115 Wu XS, Lonsdorf AS, Hwang ST, 2009, Cutaneous T-cell lymphoma: roles for chemokines and chemokine receptors. J Invest Dermatol 129:1115–1119 Capriotti E, Vonderheid EC, Thoburn CJ, Bright EC, Hess AD, 2007, Chemokine receptor expression by leukemic T cells of cutaneous T-cell lymphoma: clinical and histopathological correlations. J Invest Dermatol 127:2882–2892 Sokolowska-Wojdylo M, Wenzel J, Gaffal E, Lenz J, Speuser P, Erdmann S, Abuzahra F, Bowman E, Roszkiewicz J, Bieber T, Tuting T, 2005, Circulating clonal CLA(+, and CD4(+, T cells in Sezary syndrome express the skin-homing chemokine receptors CCR4 and CCR10 as well as the lymph node-homing chemokine receptor CCR7. Br J Dermatol 152:258– 264 Drillenburg P, Pals ST, 2000, Cell adhesion receptors in lymphoma dissemination. Blood 95:1900–1910 Borowitz MJ, Weidner A, Olsen EA, Picker LJ, 1993, Abnormalities of circulating T-cell subpopulations in patients with cutaneous T-cell lymphoma: cutaneous lymphocyte-associated antigen expression on T cells correlates with extent of disease. Leukemia 7:859–863 Heald PW, Yan SL, Edelson RL, Tigelaar R, Picker LJ, 1993, Skin-selective lymphocyte homing mechanisms in the pathogenesis of leukemic cutaneous T-cell lymphoma. J Invest Dermatol 101:222–226 Chong BF, Murphy JE, Kupper TS, Fuhlbrigge RC, 2004, Eselectin, thymus- and activation-regulated chemokine/CCL17, and intercellular adhesion molecule-1 are constitutively coexpressed in dermal microvessels: a foundation for a cutaneous immunosurveillance system. J Immunol 172:1575–1581 Hoeller C, Richardson SK, Ng LG, Valero T, Wysocka M, Rook AH, Weninger W, 2009, In vivo imaging of cutaneous T-cell lymphoma migration to the skin. Cancer Res 69:2704–2708 Wu CS, Wang ST, Liao CY, Wu MT, 2008, Differential CCR4 expression and function in cutaneous T-cell lymphoma cell lines. Kaohsiung J Med Sci 24:577–590 Santamaria-Babi LF, 2004, CLA(+, T cells in cutaneous diseases. Eur J Dermatol 14:13–18 Saeki H, Tamaki K, 2006, Thymus and activation regulated chemokine, TARC)/CCL17 and skin diseases. J Dermatol Sci 43:75–84 Yagi H, Seo N, Ohshima A, Itoh T, Itoh N, Horibe T, Yoshinari Y, Takigawa M, Hashizume H, 2006, Chemokine receptor expression in cutaneous T cell and NK/T-cell lymphomas: immunohistochemical staining and in vitro chemotactic assay. Am J Surg Pathol 30:1111–1119 Kakinuma T, Sugaya M, Nakamura K, Kaneko F, Wakugawa M, Matsushima K, Tamaki K, 2003, Thymus and activationregulated chemokine, TARC/CCL17, in mycosis fungoides: serum TARC levels reflect the disease activity of mycosis fungoides. J Am Acad Dermatol 48:23–30 Ferenczi K, Fuhlbrigge RC, Pinkus J, Pinkus GS, Kupper TS, 2002, Increased CCR4 expression in cutaneous T cell lymphoma. J Invest Dermatol 119:1405–1410 Fierro MT, Comessatti A, Quaglino P, Ortoncelli M, Osella Abate S, Ponti R, Novelli M, Bernengo MG, 2006, Expression pattern of chemokine receptors and chemokine release in inflammatory erythroderma and Sezary syndrome. Dermatology 213:284–292 Sugaya M, 2010, Chemokines and cutaneous lymphoma. J Dermatol Sci 59:81–85 Kagami S, Sugaya M, Minatani Y, Ohmatsu H, Kakinuma T, Fujita H, Tamaki K, 2006, Elevated serum CTACK/CCL27 levels in CTCL. J Invest Dermatol 126:1189–1191 Fujita Y, Abe R, Sasaki M, Honda A, Furuichi M, Asano Y, Norisugi O, Shimizu T, Shimizu H, 2006, Presence of circulating CCR10+ T cells and elevated serum CTACK/CCL27 in the early stage of mycosis fungoides. Clin Cancer Res 12:2670–2675 Lu D, Duvic M, Medeiros LJ, Luthra R, Dorfman DM, Jones D, 2001, The T-cell chemokine receptor CXCR3 is expressed highly in low-grade mycosis fungoides. Am J Clin Pathol 115:413–421 Kallinich T, Muche JM, Qin S, Sterry W, Audring H, Kroczek RA, 2003, Chemokine receptor expression on neoplastic and reactive T cells in the skin at different stages of mycosis fungoides. J Invest Dermatol 121:1045–1052 Yamaguchi T, Ohshima K, Tsuchiya T, Suehuji H, Karube K, Nakayama J, Suzumiya J, Yoshino T, Kikuchi M, 2003, The comparison of expression of cutaneous lymphocyteassociated antigen, CLA), and Th1- and Th2- associated antigens in mycosis fungoides and cutaneous lesions of adult T-cell leukemia/lymphoma. Eur J Dermatol 13:553– 559 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 749 EP 759 DI 10.1007/s12253-012-9529-5 PG 11 ER PT J AU Hegyi, K Mehes, G AF Hegyi, Katalin Mehes, Gabor TI Mitotic Failures in Cancer: Aurora B Kinase and its Potential Role in the Development of Aneuploidy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Cell division; Mitotic failure; Aurora kinase; Aneuploidy; Cancer progression ID Cell division; Mitotic failure; Aurora kinase; Aneuploidy; Cancer progression AB One of the basic requirements during the process of cell division is to maintain genetic integrity and ensure normal ploidy. The family of Aurora kinases, composed of Aurora A, B and C, takes a major role in the control of centrosome cycle, mitotic entry, chromosome condensation and coordination of chromosomal movements. Deregulation of kinase expression was described in a series of different malignancies which was also associated with aneuploidy. Recently, Aurora kinases gained significant interest as potential therapeutic targets in oncology. While there is increasing evidence about the activities of Aurora A kinase during cancer progression, data are controversial regarding the role of Aurora B. In this review the biology of Aurora kinases and its potential relation to cancer progression is discussed with special focus on functional changes and determination of Aurora B kinase. C1 [Hegyi, Katalin] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. RP Mehes, G (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, 4032 Debrecen, Hungary. EM gabor.mehes@dote.hu CR Nigg EA, 2001, Mitotic kinases as regulators of cell division and its checkpoints. Nat Rev Mol Cell Biol 2(1):21–32., DOI 10.1038/ 35048096 Bolanos-Garcia VM, 2005, Aurora kinases. Int J Biochem Cell Biol 37(8):1572–1577., DOI 10.1016/j.biocel.2005.02.021 Katayama H, Brinkley WR, Sen S, 2003, The Aurora kinases: role in cell transformation and tumorigenesis. Cancer Metastasis Rev 22(4):451–464 Carmena M, Ruchaud S, Earnshaw WC, 2009, Making the Auroras glow: regulation of Aurora A and B kinase function by interacting proteins. Curr Opin Cell Biol 21(6):796–805., DOI 10.1016/j.ceb.2009.09.008 Carvajal RD, Tse A, Schwartz GK, 2006, Aurora kinases: new targets for cancer therapy. Clin Cancer Res 12(23):6869–6875., DOI 10.1158/1078-0432.CCR-06-1405 Carmena M, Earnshaw WC, 2003, The cellular geography of aurora kinases. Nat Rev Mol Cell Biol 4(11):842–854., DOI 10.1038/nrm1245 Salaun P, Rannou Y, Prigent C, 2008, Cdk1, Plks, Auroras, and Neks: the mitotic bodyguards. Adv Exp Med Biol 617:41–56 Marumoto T, Zhang D, Saya H, 2005, Aurora-A - a guardian of poles. Nat Rev Cancer 5(1):42–50., DOI 10.1038/nrc1526 Lukasiewicz KB, Lingle WL, 2009, Aurora A, centrosome structure, and the centrosome cycle. Environ Mol Mutagen 50(8):602–619., DOI 10.1002/em.20533 Mountzios G, Terpos E, Dimopoulos MA, 2008, Aurora kinases as targets for cancer therapy. Cancer Treat Rev 34(2):175–182., DOI 10.1016/j.ctrv.2007.09.005 Tsai MY, Wiese C, Cao K, Martin O, Donovan P, Ruderman J, Prigent C, Zheng Y, 2003, A Ran signalling pathway mediated by the mitotic kinase Aurora A in spindle assembly. Nat Cell Biol 5(3):242–248., DOI 10.1038/ncb936 Kufer TA, Sillje HH, Korner R, Gruss OJ, Meraldi P, Nigg EA, 2002, Human TPX2 is required for targeting Aurora-A kinase to the spindle. J Cell Biol 158(4):617–623., DOI 10.1083/jcb.200204155 Lapenna S, Giordano A, 2009, Cell cycle kinases as therapeutic targets for cancer. Nat Rev Drug Discov 8(7):547–566., DOI 10.1038/nrd2907 Dutertre S, Cazales M, Quaranta M, Froment C, Trabut V, Dozier C, Mirey G, Bouche JP, Theis-Febvre N, Schmitt E, Monsarrat B, Prigent C, Ducommun B, 2004, Phosphorylation of CDC25B by Aurora-A at the centrosome contributes to the G2-M transition. J Cell Sci 117(Pt 12):2523–2531., DOI 10.1242/jcs.01108 Berdnik D, Knoblich JA, 2002, Drosophila Aurora-A is required for centrosome maturation and actin-dependent asymmetric protein localization during mitosis. Curr Biol 12(8):640–647 Lanni JS, Jacks T, 1998, Characterization of the p53-dependent postmitotic checkpoint following spindle disruption. Mol Cell Biol 18(2):1055–1064 Meraldi P, Honda R, Nigg EA, 2002, Aurora-A overexpression reveals tetraploidization as a major route to centrosome amplification in p53−/− cells. EMBO J 21(4):483–492 Lengauer C, Kinzler KW, Vogelstein B, 1998, Genetic instabilities in human cancers. Nature 396(6712):643–649., DOI 10.1038/25292 Marumoto T, Hirota T, Morisaki T, Kunitoku N, Zhang D, Ichikawa Y, Sasayama T, Kuninaka S, Mimori T, Tamaki N, Kimura M, Okano Y, Saya H, 2002, Roles of aurora-A kinase in mitotic entry and G2 checkpoint in mammalian cells. Genes Cells 7(11):1173–1182 Dieterich K, Soto Rifo R, Faure AK, Hennebicq S, Ben Amar B, Zahi M, Perrin J, Martinez D, Sele B, Jouk PS, Ohlmann T, Rousseaux S, Lunardi J, Ray PF, 2007, Homozygous mutation of AURKC yields large-headed polyploid spermatozoa and causes male infertility. Nat Genet 39(5):661–665., DOI 10.1038/ ng2027 Sharif B, Na J, Lykke-Hartmann K, McLaughlin SH, Laue E, Glover DM, Zernicka-GoetzM, 2010, The chromosome passenger complex is required for fidelity of chromosome transmission and cytokinesis in meiosis of mouse oocytes. J Cell Sci 123(Pt 24):4292–4300., DOI 10.1242/jcs.067447 Slattery SD, Mancini MA, Brinkley BR, Hall RM, 2009, Aurora- C kinase supports mitotic progression in the absence of Aurora-B. Cell Cycle 8(18):2984–2994 Kimura M, Matsuda Y, Yoshioka T, Okano Y, 1999, Cell cycledependent expression and centrosome localization of a third human aurora/Ipl1-related protein kinase, AIK3. J Biol Chem 274(11):7334–7340 Hu HM, Chuang CK, Lee MJ, Tseng TC, Tang TK, 2000, Genomic organization, expression, and chromosome localization of a third aurora-related kinase gene, Aie1. DNA Cell Biol 19, 11):679–688., DOI 10.1089/10445490050199063 Li X, Sakashita G, Matsuzaki H, Sugimoto K, Kimura K, Hanaoka F, Taniguchi H, Furukawa K, Urano T, 2004, Direct association with inner centromere protein, INCENP, activates the novel chromosomal passenger protein, Aurora-C. J Biol Chem 279(45):47201–47211., DOI 10.1074/jbc.M403029200 Davies H, Hunter C, Smith R, Stephens P, Greenman C, Bignell G, Teague J, Butler A, Edkins S, Stevens C, Parker A, O’Meara S, Avis T, Barthorpe S, Brackenbury L, Buck G, Clements J, Cole J, Dicks E, Edwards K, Forbes S, Gorton M, Gray K, Halliday K, Harrison R, Hills K, Hinton J, Jones D, Kosmidou V, Laman R, Lugg R, Menzies A, Perry J, Petty R, Raine K, Shepherd R, Small A, Solomon H, Stephens Y, Tofts C, Varian J, Webb A, West S, Widaa S, Yates A, Brasseur F, Cooper CS, Flanagan AM, Green A, Knowles M, Leung SY, Looijenga LH, Malkowicz B, Pierotti MA, Teh BT, Yuen ST, Lakhani SR, Easton DF, Weber BL, Goldstraw P, Nicholson AG, Wooster R, Stratton MR, Futreal PA, 2005, Somatic mutations of the protein kinase gene family in human lung cancer. Cancer Res 65(17):7591–7595., DOI 10.1158/ 0008-5472.CAN-05-1855 Fernandez-Miranda G, Trakala M, Martin J, Escobar B, Gonzalez A, Ghyselinck NB, Ortega S, Canamero M, Perez de Castro I, Malumbres M, 2011, Genetic disruption of aurora B uncovers an essential role for aurora C during early mammalian development. Development 138(13):2661–2672., DOI 10.1242/dev.066381 Lin YS, Su LJ, Yu CT, Wong FH, Yeh HH, Chen SL, Wu JC, Lin WJ, Shiue YL, Liu HS, Hsu SL, Lai JM, Huang CY, 2006, Gene expression profiles of the aurora family kinases. Gene Expr 13, 1):15–26 Ulisse S, Delcros JG, Baldini E, Toller M, Curcio F, Giacomelli L, Prigent C, Ambesi-Impiombato FS, D’Armiento M, Arlot- Bonnemains Y, 2006, Expression of Aurora kinases in human thyroid carcinoma cell lines and tissues. Int J Cancer 119(2):275– 282., DOI 10.1002/ijc.21842 Adams RR, Wheatley SP, Gouldsworthy AM, Kandels-Lewis SE, Carmena M, Smythe C, Gerloff DL, Earnshaw WC, 2000, INCENP binds the Aurora-related kinase AIRK2 and is required to target it to chromosomes, the central spindle and cleavage furrow. Curr Biol 10(17):1075–1078 Lens SM, Voest EE, Medema RH, 2010, Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nat Rev Cancer 10(12):825–841., DOI 10.1038/nrc2964 Gassmann R, Carvalho A, Henzing AJ, Ruchaud S, Hudson DF, Honda R, Nigg EA, Gerloff DL, Earnshaw WC, 2004, Borealin: a novel chromosomal passenger required for stability of the bipolar mitotic spindle. J Cell Biol 166(2):179–191., DOI 10.1083/ jcb.200404001 Hendzel MJ, Wei Y, Mancini MA, Van Hooser A, Ranalli T, Brinkley BR, Bazett-Jones DP, Allis CD, 1997, Mitosis-specific phosphorylation of histone H3 initiates primarily within pericentromeric heterochromatin during G2 and spreads in an ordered fashion coincident with mitotic chromosome condensation. Chromosoma 106(6):348–360 Ruchaud S, Carmena M, Earnshaw WC, 2007, Chromosomal passengers: conducting cell division. Nat Rev Mol Cell Biol 8, 10):798–812., DOI 10.1038/nrm2257 Weaver BA, Cleveland DW, 2005, Decoding the links between mitosis, cancer, and chemotherapy: the mitotic checkpoint, adaptation, and cell death. Cancer Cell 8(1):7–12., DOI 10.1016/ j.ccr.2005.06.011 Dar AA, Goff LW, Majid S, Berlin J, El-Rifai W, 2010, Aurora kinase inhibitors–rising stars in cancer therapeutics? Mol Cancer Ther 9(2):268–278., DOI 10.1158/1535-7163.MCT-09-0765 Ota T, Suto S, Katayama H, Han ZB, Suzuki F, Maeda M, Tanino M, Terada Y, Tatsuka M, 2002, Increased mitotic phosphorylation of histone H3 attributable to AIM-1/Aurora-B overexpression contributes to chromosome number instability. Cancer Res 62, 18):5168–5177 Goto H, Tomono Y, Ajiro K, Kosako H, Fujita M, Sakurai M, Okawa K, Iwamatsu A, Okigaki T, Takahashi T, Inagaki M, 1999, Identification of a novel phosphorylation site on histone H3 coupled with mitotic chromosome condensation. J Biol Chem 274(36):25543–25549 Hsu JY, Sun ZW, Li X, Reuben M, Tatchell K, Bishop DK, Grushcow JM, Brame CJ, Caldwell JA, Hunt DF, Lin R, Smith MM, Allis CD, 2000, Mitotic phosphorylation of histone H3 is governed by Ipl1/aurora kinase and Glc7/PP1 phosphatase in budding yeast and nematodes. Cell 102(3):279–291 Crosio C, Fimia GM, Loury R, Kimura M, Okano Y, Zhou H, Sen S, Allis CD, Sassone-Corsi P, 2002, Mitotic phosphorylation of histone H3: spatio-temporal regulation by mammalian Aurora kinases. Mol Cell Biol 22(3):874–885 Perez-Cadahia B, Drobic B, Davie JR, 2009, H3 phosphorylation: dual role in mitosis and interphase. Biochem Cell Biol 87(5):695– 709., DOI 10.1139/O09-053 Kang TH, Park DY, Choi YH, Kim KJ, Yoon HS, Kim KT, 2007, Mitotic histone H3 phosphorylation by vaccinia-related kinase 1 in mammalian cells. Mol Cell Biol 27(24):8533–8546., DOI 10.1128/ MCB.00018-07 Loomis RJ, Naoe Y, Parker JB, Savic V, Bozovsky MR, Macfarlan T, Manley JL, Chakravarti D, 2009, Chromatin binding of SRp20 and ASF/SF2 and dissociation from mitotic chromosomes is modulated by histone H3 serine 10 phosphorylation. Mol Cell 33(4):450–461., DOI 10.1016/j.molcel.2009.02.003 Moy C, Oleykowski CA, Plant R, Greshock J, Jing J, Bachman K, Hardwicke MA, Wooster R, Degenhardt Y, 2011, High chromosome number in hematological cancer cell lines is a negative predictor of response to the inhibition of Aurora B and C by GSK1070916. J Transl Med 9:110., DOI 10.1186/ 1479-5876-9-110 Walsby E, Walsh V, Pepper C, Burnett A, Mills K, 2008, Effects of the aurora kinase inhibitors AZD1152-HQPA and ZM447439 on growth arrest and polyploidy in acute myeloid leukemia cell lines and primary blasts. Haematologica 93(5):662–669., DOI 10.3324/haematol.12148 Girdler F, Sessa F, Patercoli S, Villa F, Musacchio A, Taylor S, 2008, Molecular basis of drug resistance in aurora kinases. Chem Biol 15(6):552–562., DOI 10.1016/j.chembiol.2008.04.013 Qi G, Ogawa I, Kudo Y, MiyauchiM, Siriwardena BS, Shimamoto F, Tatsuka M, Takata T, 2007, Aurora-B expression and its correlation with cell proliferation and metastasis in oral cancer. Virchows Arch 450(3):297–302., DOI 10.1007/s00428-006-0360-9 KuraiM, Shiozawa T, ShihHC, Miyamoto T, Feng YZ,Kashima H, Suzuki A, Konishi I, 2005, Expression of Aurora kinases A and B in normal, hyperplastic, and malignant human endometrium: Aurora B as a predictor for poor prognosis in endometrial carcinoma. Hum Pathol 36(12):1281–1288., DOI 10.1016/ j.humpath.2005.09.014 Sorrentino R, Libertini S, Pallante PL, Troncone G, Palombini L, Bavetsias V, Spalletti-Cernia D, Laccetti P, Linardopoulos S, Chieffi P, Fusco A, Portella G, 2005, Aurora B overexpression associates with the thyroid carcinoma undifferentiated phenotype and is required for thyroid carcinoma cell proliferation. J Clin Endocrinol Metab 90(2):928–935., DOI 10.1210/jc.2004-1518 Garcia-Fernandez E, De Diego JI, Collantes-Bellido E, Mendiola M, Prim MP, Perez-Fernandez E, Miguel-Martin M, Nistal M, Hardisson D, 2011, Aurora B kinase expression in laryngeal squamous cell carcinoma and its prognostic implications. Histopathology 58(3):368–376., DOI 10.1111/j.1365-2559.2011. 03757.x Lin ZZ, Jeng YM, Hu FC, Pan HW, Tsao HW, Lai PL, Lee PH, Cheng AL, Hsu HC, 2010, Significance of Aurora B overexpression in hepatocellular carcinoma. Aurora B Overexpression in HCC. BMC Cancer 10:461., DOI 10.1186/1471-2407-10-461 Benten D, Keller G, Quaas A, Schrader J, Gontarewicz A, Balabanov S, Braig M, Wege H, Moll J, Lohse AW, Brummendorf TH, 2009, Aurora kinase inhibitor PHA-739358 suppresses growth of hepatocellular carcinoma in vitro and in a xenograft mouse model. Neoplasia 11(9):934–944 Jantscher F, Pirker C, Mayer CE, Berger W, Sutterluety H, 2011, Overexpression of Aurora-A in primary cells interferes with Sphase entry by diminishing Cyclin D1 dependent activities. Mol Cancer 10:28., DOI 10.1186/1476-4598-10-28 Cha TL, Chuang MJ, Wu ST, Sun GH, Chang SY, Yu DS, Huang SM, Huan SK, Cheng TC, Chen TT, Fan PL, Hsiao PW, 2009, Dual degradation of aurora A and B kinases by the histone deacetylase inhibitor LBH589 induces G2-M arrest and apoptosis of renal cancer cells. Clin Cancer Res 15(3):840–850., DOI 10.1158/1078-0432.CCR-08-1918 Katayama H, Sasai K, Czerniak BA, Carter JL, Sen S, 2007, Aurora-A kinase phosphorylation of Aurora-A kinase interacting protein, AIP, and stabilization of the enzyme-substrate complex. J Cell Biochem 102(5):1318–1331., DOI 10.1002/jcb.21421 Venoux M, Basbous J, Berthenet C, Prigent C, Fernandez A, Lamb NJ, Rouquier S, 2008, ASAP is a novel substrate of the oncogenic mitotic kinase Aurora-A: phosphorylation on Ser625 is essential to spindle formation and mitosis. Hum Mol Genet 17, 2):215–224., DOI 10.1093/hmg/ddm298 Ouchi M, Fujiuchi N, Sasai K, Katayama H, Minamishima YA, Ongusaha PP, Deng C, Sen S, Lee SW, Ouchi T, 2004, BRCA1 phosphorylation by Aurora-A in the regulation of G2 toMtransition. J Biol Chem 279(19):19643–19648., DOI 10.1074/jbc.M311780200 Pugacheva EN, Golemis EA, 2005, The focal adhesion scaffolding protein HEF1 regulates activation of the Aurora-A and Nek2 kinases at the centrosome. Nat Cell Biol 7(10):937–946., DOI 10.1038/ncb1309 Cazales M, Schmitt E, Montembault E, Dozier C, Prigent C, Ducommun B, 2005, CDC25B phosphorylation by Aurora-A occurs at the G2/M transition and is inhibited by DNA damage. Cell Cycle 4(9):1233–1238 Kunitoku N, Sasayama T, Marumoto T, Zhang D, Honda S, Kobayashi O, Hatakeyama K, Ushio Y, Saya H, Hirota T, 2003, CENP-A phosphorylation by Aurora-A in prophase is required for enrichment of Aurora-B at inner centromeres and for kinetochore function. Dev Cell 5(6):853–864 Kim Y, Holland AJ, Lan W, Cleveland DW, 2010, Aurora kinases and protein phosphatase 1 mediate chromosome congression through regulation of CENP-E. Cell 142(3):444–455., DOI 10.1016/ j.cell.2010.06.039 Huang YS, Jung MY, Sarkissian M, Richter JD, 2002, N-methyl- D-aspartate receptor signaling results in Aurora kinase-catalyzed CPEB phosphorylation and alpha CaMKII mRNA polyadenylation at synapses. EMBO J 21(9):2139–2148., DOI 10.1093/emboj/ 21.9.2139 Mendez R, Hake LE, Andresson T, Littlepage LE, Ruderman JV, Richter JD, 2000, Phosphorylation of CPE binding factor by Eg2 regulates translation of c-mos mRNA. Nature 404(6775):302– 307., DOI 10.1038/35005126 Ban R, Matsuzaki H, Akashi T, Sakashita G, Taniguchi H, Park SY, Tanaka H, Furukawa K, Urano T, 2009, Mitotic regulation of the stability of microtubule plus-end tracking protein EB3 by ubiquitin ligase SIAH-1 and Aurora mitotic kinases. J Biol Chem 284(41):28367–28381., DOI 10.1074/jbc.M109.000273 Jang MS, Sul JW, Choi BJ, Lee SJ, Suh JH, Kim NS, Kim WH, Lim DS, Lee CW, Kim E, 2008, Negative feedback regulation of Aurora-A via phosphorylation of Fas-associated factor-1. J Biol Chem 283(47):32344–32351., DOI 10.1074/jbc.M804199200 Birkenfeld J, Nalbant P, Bohl BP, Pertz O, Hahn KM, Bokoch GM, 2007, GEF-H1 modulates localized RhoA activation during cytokinesis under the control of mitotic kinases. Dev Cell 12, 5):699–712., DOI 10.1016/j.devcel.2007.03.014 Dar AA, Belkhiri A, El-Rifai W, 2009, The aurora kinase A regulates GSK-3beta in gastric cancer cells. Oncogene 28, 6):866–875., DOI 10.1038/onc.2008.434 Yang SC, Huang CH, Chen NJ, Chou CK, Lin CH, 2000, Functional implication of human serine/threonine kinase, hAIK, in cell cycle progression. J Biomed Sci 7(6):484–493 Yu CT, Hsu JM, Lee YC, Tsou AP, Chou CK, Huang CY, 2005, Phosphorylation and stabilization of HURP by Aurora-A: implication of HURP as a transforming target of Aurora-A. Mol Cell Biol 25, 14):5789–5800., DOI 10.1128/MCB.25.14.5789-5800.2005 Jang CY, Coppinger JA, Seki A, Yates JR 3rd, Fang G, 2009, Plk1 and Aurora A regulate the depolymerase activity and the cellular localization of Kif2a. J Cell Sci 122(Pt 9):1334–1341., DOI 10.1242/jcs.044321 Toji S, Yabuta N, Hosomi T, Nishihara S, Kobayashi T, Suzuki S, Tamai K, Nojima H, 2004, The centrosomal protein Lats2 is a phosphorylation target of Aurora-A kinase. Genes Cells 9, 5):383–397., DOI 10.1111/j.1356-9597.2004.00732.x Sakai H, Urano T, Ookata K, Kim MH, Hirai Y, Saito M, Nojima Y, Ishikawa F, 2002, MBD3 and HDAC1, two components of the NuRD complex, are localized at Aurora-A-positive centrosomes in M phase. J Biol Chem 277(50):48714–48723., DOI 10.1074/ jbc.M208461200 Mori D, Yano Y, Toyo-oka K, Yoshida N, Yamada M, Muramatsu M, Zhang D, Saya H, Toyoshima YY, Kinoshita K, Wynshaw- Boris A, Hirotsune S, 2007, NDEL1 phosphorylation by Aurora- A kinase is essential for centrosomal maturation, separation, and TACC3 recruitment. Mol Cell Biol 27(1):352–367., DOI 10.1128/ MCB.00878-06 Molli PR, Li DQ, Bagheri-Yarmand R, Pakala SB, Katayama H, Sen S, Iyer J, Chernoff J, Tsai MY, Nair SS, Kumar R, 2010, Arpc1b, a centrosomal protein, is both an activator and substrate of Aurora A. J Cell Biol 190(1):101–114., DOI 10.1083/jcb. 200908050 Katayama H, Sasai K, Kawai H, Yuan ZM, Bondaruk J, Suzuki F, Fujii S, Arlinghaus RB, Czerniak BA, Sen S, 2004, Phosphorylation by aurora kinase A induces Mdm2-mediated destabilization and inhibition of p53. Nat Genet 36(1):55–62., DOI 10.1038/ng1279 Khazaei MR, Puschel AW, 2009, Phosphorylation of the par polarity complex protein Par3 at serine 962 is mediated by aurora a and regulates its function in neuronal polarity. J Biol Chem 284, 48):33571–33579., DOI 10.1074/jbc.M109.055897 Seki A, Coppinger JA, Jang CY, Yates JR, Fang G, 2008, Bora and the kinase Aurora a cooperatively activate the kinase Plk1 and control mitotic entry. Science 320(5883):1655–1658., DOI 10.1126/science.1157425 Macurek L, Lindqvist A, Lim D, Lampson MA, Klompmaker R, Freire R, Clouin C, Taylor SS, Yaffe MB, Medema RH, 2008, Polo-like kinase-1 is activated by aurora A to promote checkpoint recovery. Nature 455(7209):119–123., DOI 10.1038/nature07185 Katayama H, Zhou H, Li Q, Tatsuka M, Sen S, 2001, Interaction and feedback regulation between STK15/BTAK/Aurora-A kinase and protein phosphatase 1 through mitotic cell division cycle. J Biol Chem 276(49):46219–46224., DOI 10.1074/jbc.M107540200 Jang CY, Coppinger JA, Yates JR 3rd, Fang G, 2011, Mitotic kinases regulate MT-polymerizing/MT-bundling activity of DDA3. Biochem Biophys Res Commun 408(1):174–179., DOI 10.1016/j.bbrc.2011.04.004 Wu JC, Chen TY, Yu CT, Tsai SJ, Hsu JM, Tang MJ, Chou CK, Lin WJ, Yuan CJ, Huang CY, 2005, Identification of V23RalASer194 as a critical mediator for Aurora-A-induced cellular motility and transformation by small pool expression screening. J Biol Chem 280(10):9013–9022., DOI 10.1074/jbc.M411068200 Lim KH, Brady DC, Kashatus DF, Ancrile BB, Der CJ, Cox AD, Counter CM, 2010, Aurora-A phosphorylates, activates, and relocalizes the small GTPase RalA. Mol Cell Biol 30(2):508– 523., DOI 10.1128/MCB.00916-08 Rong R, Jiang LY, Sheikh MS, Huang Y, 2007, Mitotic kinase Aurora-A phosphorylates RASSF1A and modulates RASSF1Amediatedmicrotubule interaction andM-phase cell cycle regulation. Oncogene 26(55):7700–7708., DOI 10.1038/sj.onc.1210575 LeRoy PJ, Hunter JJ, Hoar KM, Burke KE, Shinde V, Ruan J, Bowman D, Galvin K, Ecsedy JA, 2007, Localization of human TACC3 to mitotic spindles is mediated by phosphorylation on Ser558 by Aurora A: a novel pharmacodynamic method for measuring Aurora A activity. Cancer Res 67(11):5362–5370., DOI 10.1158/0008-5472.CAN-07-0122 Ohishi T, Hirota T, Tsuruo T, Seimiya H, 2010, TRF1 mediates mitotic abnormalities induced by Aurora-A overexpression. Cancer Res 70(5):2041–2052., DOI 10.1158/0008-5472.CAN-09-2008 Troiani S, Uggeri M, Moll J, Isacchi A, Kalisz HM, Rusconi L, Valsasina B, 2005, Searching for biomarkers of Aurora-A kinase activity: identification of in vitro substrates through a modified KESTREL approach. J Proteome Res 4(4):1296–1303., DOI 10.1021/pr050018e Harper M, Tillit J, Kress M, Ernoult-Lange M, 2009, Phosphorylation-dependent binding of human transcription factor MOK2 to lamin A/C. FEBS J 276(11):3137–3147., DOI 10.1111/ j.1742-4658.2009.07032.x Yasui Y, Urano T, Kawajiri A, Nagata K, Tatsuka M, Saya H, Furukawa K, Takahashi T, Izawa I, Inagaki M, 2004, Autophosphorylation of a newly identified site of Aurora-B is indispensable for cytokinesis. J Biol Chem 279(13):12997– 13003., DOI 10.1074/jbc.M311128200 Zeitlin SG, Shelby RD, Sullivan KF, 2001, CENP-A is phosphorylated by Aurora B kinase and plays an unexpected role in completion of cytokinesis. J Cell Biol 155(7):1147– 1157., DOI 10.1083/jcb.200108125 DeLuca JG, Gall WE, Ciferri C, Cimini D, Musacchio A, Salmon ED, 2006, Kinetochore microtubule dynamics and attachment stability are regulated by Hec1. Cell 127(5):969–982., DOI 10.1016/j.cell.2006.09.047 Goto H, Yasui Y, Nigg EA, Inagaki M, 2002, Aurora-B phosphorylates Histone H3 at serine28 with regard to the mitotic chromosome condensation. Genes Cells 7(1):11–17 Bishop JD, Schumacher JM, 2002, Phosphorylation of the carboxyl terminus of inner centromere protein, INCENP, by the Aurora B Kinase stimulates Aurora B kinase activity. J Biol Chem 277(31):27577–27580., DOI 10.1074/jbc.C200307200 Lan W, Zhang X, Kline-Smith SL, Rosasco SE, Barrett-Wilt GA, Shabanowitz J, Hunt DF, Walczak CE, Stukenberg PT, 2004, Aurora B phosphorylates centromeric MCAK and regulates its localization and microtubule depolymerization activity. Curr Biol 14(4):273–286., DOI 10.1016/j.cub.2004.01.055 Minoshima Y, Kawashima T, Hirose K, Tonozuka Y, Kawajiri A, Bao YC, Deng X, Tatsuka M, Narumiya S, May WS Jr, Nosaka T, Semba K, Inoue T, Satoh T, Inagaki M, Kitamura T, 2003, Phosphorylation by aurora B converts MgcRacGAP to a RhoGAP during cytokinesis. Dev Cell 4(4):549–560 Guse A,MishimaM, GlotzerM(2005, Phosphorylation of ZEN-4/ MKLP1 by aurora B regulates completion of cytokinesis. Curr Biol 15(8):778–786., DOI 10.1016/j.cub.2005.03.041 Sakita-Suto S, Kanda A, Suzuki F, Sato S, Takata T, Tatsuka M, 2007, Aurora-B regulates RNA methyltransferase NSUN2. Mol Biol Cell 18(3):1107–1117., DOI 10.1091/ mbc.E06-11-1021 Tanno Y, Kitajima TS, Honda T, Ando Y, Ishiguro K, Watanabe Y, 2010, Phosphorylation of mammalian Sgo2 by Aurora B recruits PP2A and MCAK to centromeres. Genes Dev 24, 19):2169–2179., DOI 10.1101/gad.1945310 Wheatley SP, Henzing AJ, Dodson H, Khaled W, Earnshaw WC, 2004, Aurora-B phosphorylation in vitro identifies a residue of survivin that is essential for its localization and binding to inner centromere protein, INCENP, in vivo. J Biol Chem 279(7):5655– 5660., DOI 10.1074/jbc.M311299200 Goto H, Yasui Y, Kawajiri A, Nigg EA, Terada Y, Tatsuka M, Nagata K, Inagaki M, 2003, Aurora-B regulates the cleavage furrow-specific vimentin phosphorylation in the cytokinetic process. J Biol Chem 278(10):8526–8530., DOI 10.1074/jbc.M210892200 Lee EC, Frolov A, Li R, Ayala G, Greenberg NM, 2006, Targeting Aurora kinases for the treatment of prostate cancer. Cancer Res 66, 10):4996–5002., DOI 10.1158/0008-5472.CAN-05-2796 Chieffi P, Cozzolino L, Kisslinger A, Libertini S, Staibano S, Mansueto G, De Rosa G, Villacci A, Vitale M, Linardopoulos S, Portella G, Tramontano D, 2006, Aurora B expression directly correlates with prostate cancer malignancy and influence prostate cell proliferation. Prostate 66(3):326–333., DOI 10.1002/pros.20345 Chieffi P, Troncone G, Caleo A, Libertini S, Linardopoulos S, Tramontano D, Portella G, 2004, Aurora B expression in normal testis and seminomas. J Endocrinol 181(2):263–270 Baldini E, Arlot-BonnemainsY,MottoleseM, Sentinelli S, Antoniani B, Sorrenti S, Salducci M, Comini E, Ulisse S, D’ArmientoM(2010, Deregulation of Aurora kinase gene expression in human testicular germ cell tumours. Andrologia 42(4):260–267., DOI 10.1111/j.1439- 0272.2009.00987.x Katayama H, Ota T, Jisaki F, Ueda Y, Tanaka T, Odashima S, Suzuki F, Terada Y, Tatsuka M, 1999, Mitotic kinase expression and colorectal cancer progression. J Natl Cancer Inst 91, 13):1160–1162 Bischoff JR, Anderson L, Zhu Y, Mossie K, Ng L, Souza B, Schryver B, Flanagan P, Clairvoyant F, Ginther C, Chan CS, Novotny M, Slamon DJ, Plowman GD, 1998, A homologue of Drosophila aurora kinase is oncogenic and amplified in human colorectal cancers. EMBO J 17(11):3052–3065., DOI 10.1093/ emboj/17.11.3052 Araki K, Nozaki K, Ueba T, Tatsuka M, Hashimoto N, 2004, High expression of Aurora-B/Aurora and Ipll-like midbodyassociated protein, AIM-1, in astrocytomas. J Neurooncol 67, 1–2):53–64 Li Y, Zhang ZF, Chen J, Huang D, Ding Y, Tan MH, Qian CN, Resau JH, Kim H, Teh BT, 2010, VX680/MK-0457, a potent and selective Aurora kinase inhibitor, targets both tumor and endothelial cells in clear cell renal cell carcinoma. Am J Transl Res 2(3):296–308 Ikezoe T, Yang J, Nishioka C, Tasaka T, Taniguchi A, Kuwayama Y, Komatsu N, Bandobashi K, Togitani K, Koeffler HP, Taguchi H, 2007, A novel treatment strategy targeting Aurora kinases in acute myelogenous leukemia. Mol Cancer Ther 6(6):1851–1857., DOI 10.1158/1535-7163.MCT-07-0067 Hamada M, Yakushijin Y, Ohtsuka M, Kakimoto M, Yasukawa M, Fujita S, 2003, Aurora2/BTAK/STK15 is involved in cell cycle checkpoint and cell survival of aggressive non-Hodgkin’s lymphoma. Br J Haematol 121(3):439–447 Smith SL, Bowers NL, Betticher DC, Gautschi O, Ratschiller D, Hoban PR, Booton R, Santibanez-Koref MF, Heighway J, 2005, Overexpression of aurora B kinase, AURKB, in primary nonsmall cell lung carcinoma is frequent, generally driven from one allele, and correlates with the level of genetic instability. Br J Cancer 93(6):719–729., DOI 10.1038/sj.bjc.6602779 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 761 EP 769 DI 10.1007/s12253-012-9534-8 PG 9 ER PT J AU Zeyaullah, M Patro, M Ahmad, I Ibraheem, K Sultan, P Nehal, M Ali, A AF Zeyaullah, M Patro, Mohan Ahmad, Irfan Ibraheem, Kawthar Sultan, P Nehal, Mohammad Ali, Arif TI Oncolytic Viruses in the Treatment of Cancer: A Review of Current Strategies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Oncolytic viruses; Tumor cells; Human cancer; Virotherapeutics; Combination therapy ID Oncolytic viruses; Tumor cells; Human cancer; Virotherapeutics; Combination therapy AB Oncolytic viruses are live, replication-competent viruses that replicate selectively in tumor cells leading to the destruction of the tumor cells. Tumor-selective replicating viruses offer appealing advantages over conventional cancer therapy and are promising a new approach for the treatment of human cancer. The development of virotherapeutics is based on several strategies. Virotherapy is not a new concept, but recent technical advances in the genetic modification of oncolytic viruses have improved their tumor specificity, leading to the development of new weapons for the war against cancer. Clinical trials with oncolytic viruses demonstrate the safety and feasibility of an effective virotherapeutic approach. Strategies to overcome potential obstacles and challenges to virotherapy are currently being explored. Systemic administrations of oncolytic viruses will successfully extend novel treatment against a range of tumors. Combination therapy has shown some encouraging antitumor responses by eliciting strong immunity against established cancer. C1 [Zeyaullah, M] Omar Al-Mukhtar University, Faculty of Medicine, Department of MicrobiologyAl-Baida, Libyan Arab Jamabiriya. [Patro, Mohan] Omar Al-Mukhtar University, Faculty of Medicine, Department of SurgeryAl-Baida, Libyan Arab Jamabiriya. [Ahmad, Irfan] Jamia Millia Islamia, Department of Biosciences, 110025 New Delhi, India. [Ibraheem, Kawthar] Omar Al-Mukhtar University, Faculty of Medicine, Department of MicrobiologyAl-Baida, Libyan Arab Jamabiriya. [Sultan, P] Omar Al-Mukhtar University, Faculty of Medicine, Department of BiochemistryAl-Baida, Libyan Arab Jamabiriya. [Nehal, Mohammad] Central University of Bihar (CUB)Patna, Bihar, India. [Ali, Arif] Jamia Millia Islamia (Central University), Department of Biotechnology, 110025 New Delhi, India. RP Zeyaullah, M (reprint author), Omar Al-Mukhtar University, Faculty of Medicine, Department of Microbiology, Al-Baida, Libyan Arab Jamabiriya. EM zeya786@gmail.com CR Biederer C, Ries S, Brandts CH et al, 2002, Replication-selective viruses for cancer therapy. J Mol Med 80:163–175 Chiocca AE, 2002, Oncolytic viruses. Nat Rev Cancer 2:938–950 Everts B, Van der Poel HG, 2005, Replication-selective oncolytic viruses in the treatment of cancer. Cancer Gene Ther 12(2):141–161 Guo ZS, Naik A, O’Malley ME et al, 2005, The enhanced tumor selectivity of an oncolytic vaccinia lacking the host range and antiapoptosis genes SPI-1 and SPI-2. Cancer Res 65:9991–9998 Guo ZS, Thorne SH, Bartlett DL, 2008, Oncolytic virotherapy: molecular targets in tumor-selective replication and carrier cellmediated delivery of oncolytic viruses. Biochim Biophys Acta 1785:217–231 Thorne SH, Hermiston T, Kirn D, 2005, Oncolytic virotherapy: approaches to tumor targeting and enhancing antitumor effects. Semin Oncol 32:537–548 Kirn D, Martuza RL, Zwiebel J, 2001, Replication-selective virotherapy for cancer: Biological principles, risk management and future directions. Nat Med 7:781–787 Harrington KJ, Vile RG, Melcher A et al, 2010, Clinical trials with oncolytic reovirus: moving beyond phase I into combinations with standard therapeutics. Cytokine Growth Factor Rev 21:91–98 Van DenWollenberg DJ, Van Den Hengel SK, Dautzenberg IJ et al, 2009, Modification of mammalian reoviruses for use as oncolytic agents. Expert Opin Biol Ther 9:1509–1520 Schirrmacher V, Fournier P, 2009, Newcastle disease virus: a promising vector for viral therapy, immune therapy, and gene therapy of cancer. Methods in molecular biology 542:565–605 Barber GN, 2005, VSV-tumor selective replication and protein translation. Oncogene 24:7710–7719 Kirn DH, Thorne SH, 2009, Targeted and armed oncolytic poxviruses: a novel multi-mechanistic therapeutic class for cancer. Nat Rev Cancer 9:64–71 Msaouel P, Iankov ID, Dispenzieri A et al, 2011, Attenuated Oncolytic Measles Virus Strains as Cancer Therapeutics. Curr Pharm Biotechnol. Toth K, Wold WS, 2010, Increasing the efficacy of oncolytic adenovirus vectors. Viruses 2:1844–1866 Kaur B, Chiocca EA, Cripe TP, 2011, Oncolytic HSV-1 Virotherapy: Clinical Experience and Opportunities for Progress. Curr Pharm Biotechnol. Goetz C, Gromeier M, 2010, Preparing an oncolytic poliovirus recombinant for clinical application against glioblastoma multiforme. Cytokine Growth Factor Rev 21:197–203 Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100:57–70 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Kirn DH, Wang Y, Le Boeuf F et al, 2007, Targeting of interferon-beta to produce a specific, multi-mechanistic oncolytic vaccinia virus. PLoS Med 4:e353 McCart JA, Ward JM, Lee J et al, 2001, Systemic cancer therapy with a tumor-selective vaccinia virus mutant lacking thymidine kinase and vaccinia growth factor genes. Cancer Res 61:8751–8757 DeWeese TL, van der Poel H, Li S et al, 2001, A phase I trial of CV706, a replication-competent, PSA selective oncolytic adenovirus, for the treatment of locally recurrent prostate cancer following radiation therapy. Cancer Res 61:7464–7472 Li Y, Yu DC, Chen Y et al, 2001, A hepatocellular carcinomaspecific adenovirus variant, CV890, eliminates distant human liver tumors in combination with doxorubicin. Cancer Res 61: 6428–6436 Yu DC, Chen Y, Dilley J et al, 2001, Antitumor synergy of CV787, a prostate cancer-specific adenovirus, and paclitaxel and docetaxel. Cancer Res 61:517–525 Rojas JJ, Guedan S, Searle PF et al, 2010, Minimal RB-responsive E1A promoter modification to attain potency, selectivity, and transgene-arming capacity in oncolytic adenoviruses. Mol Ther 18:1960–1971 Roelvink PW, Mi Lee G, Einfeld DA et al, 1999, Identification of a conserved receptor-binding site on the fiber proteins of CARrecognizing adenoviridae. Science 286:1568–1571 Douglas JT, Rogers BE, Rosenfeld ME et al, 1996, Targeted gene delivery by tropism-modified adenoviral vectors. Nat Biotechnol 14:1574–1578 Peng KW, Donovan KA, Schneider U et al, 2003, Oncolytic measles viruses displaying a single-chain antibody against CD38, a myeloma cell marker. Blood 101:2557–2562 Thorne SH, 2011, Immunotherapeutic potential of oncolytic vaccinia virus. Immunol Res 50:286–293 Breitbach CJ, Paterson JM, Lemay CG et al, 2007, Targeted inflammation during oncolytic virus therapy severely compromises tumor blood flow. Mol Ther 15:1686–1693 Breitbach CJ, Reid T, Burke J et al, 2010, Navigating the clinical development landscape for oncolytic viruses and other cancer therapeutics: No shortcuts on the road to approval. Cytokine Growth Factor Rev 21:85–89 Breitbach CJ, Thorne SH, Bell JC et al, 2011, Targeted and Armed Oncolytic Poxviruses for Cancer: The Lead Example of JX-594. Curr Pharm Biotechnol. Liu TC, Hwang T, Park BH et al, 2008, The targeted oncolytic poxvirus JX-594 demonstrates antitumoral, antivascular, and anti-HBV activities in patients with hepatocellular carcinoma. Mol Ther 16:1637–1642 Hermiston T, 2002, Fighting fire with fire: attacking the complexity of human tumors with armed therapeutic viruses. Curr Opin Mol Ther 4:334–342 Hermiston TW, Kuhn I, 2002, Armed therapeutic viruses: strategies and challenges to arming oncolytic viruses with therapeutic genes. Cancer Gene Ther 9:1022–1035 Bell J, 2010, Oncolytic viruses: an approved product on the horizon? Mol Ther 18:233–234 Cattaneo R, Miest T, Shashkova EV et al, 2008, Reprogrammed viruses as cancer therapeutics: targeted, armed and shielded. Nat Rev Microbiol 6:529–540 John LB, Howland LJ, Flynn JK et al, 2012, Oncolytic virus and anti–4-1BB combination therapy elicits strong antitumor immunity against established cancer. Cancer Res 72(7):1651–1660 Seton-Rogers S, 2012, Immunotherapy: combinations that work. Nat Rev Cancer 12:231 Vanneman M, Dranoff G, 2011, Combining immunotherapy and targeted therapies in cancer treatment. Nature Rev Cancer 12:237–251 Sinkovics JG, Horvath JC, 2008, Natural and genetically engineered viral agents for oncolysis and gene therapy of human cancers. Arch Immunol Ther Exp, Warsz, 56(Suppl 1):3s–59s Russell SJ, 2002, RNA viruses as virotherapy agents. Cancer Gene Ther 9:961–966 Norman KL, Lee PW, 2000, Reovirus as a novel oncolytic agent. J Clin Invest 105:1035–1038 Strong JE, Coffey MC, Tang D et al, 1998, The molecular basis of viral oncolysis: usurpation of the Ras signaling pathway by reovirus. EMBO J 17:3351–3362 Cornelis JJ, Salome N, Dinsart C et al, 2004, Vectors based on autonomous parvoviruses: novel tools to treat cancer? J Gene Med 6:S193–S202 Rommelaere J, Cornelis JJ, 1991, Antineoplastic activity of parvoviruses. J Virol Methods 33:233–251 Farassati F, Yang A, Lee PWK, 2001, Oncogenes in Ras signalling pathway dictate host-cell permissiveness to herpes simplex virus 1. Nat Cell Biol 3:745–750 He B, Gross M, Roizman B, 1997, The 134.5 protein of herpes simplex virus complexes with protein phosphatase 1 to dephosphorylate the subunit of the eukaryotic translation initiation factor 2 and preclude the shutoff of protein synthesis by doublestranded RNA-activated protein kinase. Proc Natl Acad Sci USA 94:843–848 Varghese S, Rabkin SD, 2002, Oncolytic herpes simplex virus vectors for cancer virotherapy. Cancer Gene Ther 9:967–978 Reid T, Galanis E, Abbruzzese J et al, 2001, Intra-arterial administration of a replicationselective adenovirus, dl1520, in patients with colorectal carcinoma metastatic to the liver: a Phase I trial. Gene Ther 8:1618–1626 Reid T, Warren R, Kirn D, 2002, Intravascular adenoviral agents in cancer patients: lessons from clinical trials. Cancer Gene Ther 9:979–986 Reid T, Galanis E, Abbruzzese J et al, 2002, Hepatic arterial infusion of a replication-selective oncolytic adenovirus, dl1520): Phase II viral, immunologic, and clinical endpoints. Cancer Res 62:6070–6079 Zeh HJ, Bartlett DL, 2002, Development of a replicationselective, oncolytic poxvirus for the treatment of human cancers. CancerGene Ther 9:1001–1012 Gromeier M, Alexander L, Wimmer E, 1996, Internal ribosomal entry site substitution eliminates neurovirulence in intergeneric poliovirus recombinants. Proc Natl Acad Sci USA 93:2370–2375 Gromeier M, Lachmann S, Rosenfeld MR et al, 2000, Intergeneric poliovirus recombinants for the treatment ofmalignant glioma. Proc Natl Acad Sci U S A 97:6803–6808 Mineta T, Rabkin SD, Yazaki T et al, 1995, Attenuated multimutated herpes simplex virus-1 for the treatment of malignant gliomas. Nat Med 1:938–943 Dobbelstein M, 2004, Replicating adenoviruses in cancer therapy. Curr Top Microbiol Immunol 273:291–334 Flint J, Shenk T, 1997, Viral transactivating proteins. Annu Rev Genet 31:177–212 de Stanchina E, McCurrach ME, Zindy F et al, 1998, E1A signaling to p53 involves the p19, ARF, tumor suppressor. Genes Dev 12:2434–2442 Kao CC, Yew PR, Berk AJ, 1990, Domains required for in vitro association between the cellular p53 and the adenovirus 2 E1B 55 K proteins. Virology 179:806–814 Yew PR, Berk AJ, 1992, Inhibition of p53 transactivation required for transformation by adenovirus early 1B protein. Nature 357:82–85 Yew PR, Liu X, Berk AJ, 1994, Adenovirus E1B oncoprotein tethers a transcriptional repression domain to p53. Genes Dev 8:190–202 Bischoff JR, Kirn DH, Williams A et al, 1996, An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science 274:373–376 McCormick F, 2000, ONYX-015 selectivity and the p14ARF pathway. Oncogene 19:6670–6672 Petit T, Davidson KK, Cerna C et al, 2002, Efficient induction of apoptosis by ONYX-015 adenovirus in human colon cancer cell lines regardless of p53 status. Anticancer Drugs 13:47–50 Ries SJ, Brandts CH, Chung AS et al, 2000, Loss of p14ARF in tumor cells facilitates replication of the adenovirus mutant dl1520, ONYX-015). Nat Med 6:1128–1133 Ries S, Korn WM, 2002, ONYX-015: mechanisms of action and clinical potential of a replication-selective adenovirus. Br J Cancer 86:5–11 Johnson L, Shen A, Boyle L et al, 2002, Selectively replicating adenoviruses targeting deregulated E2F activity are potent, systemic antitumor agents. Cancer Cell 1:325–337 Fueyo J, Gomez-Manzano C, Alemany R et al, 2000, A mutant oncolytic adenovirus targeting the Rb pathway produces antiglioma effect in vivo. Oncogene 19:2–12 Heise C, Hermiston T, Johnson L et al, 2000, An adenovirus E1A mutant that demonstrates potent and selective systemic antitumoral efficacy. Nat Med 6:1134–1139 Chen Y, De Weese T, Dilley J et al, 2001, CV706, a prostate cancer-specific adenovirus variant, in combination with radiotherapy produces synergistic antitumor efficacy without increasing toxicity. Cancer Res 61:5453–5460 Yu DC, Chen Yu, Seng M et al, 1999, The addition of adenovirus type 5 region E3 enables calydon virus 787 to eliminate distant prostate tumor xenografts. Cancer Res 59:4200–4203 Yu YA, Shabahang S, Timiryasova TMet al, 2004, Visualization of tumors and metastases in live animals with bacteria and vaccinia virus encoding light-emitting proteins. Nat Biotechnol 22:313–320 Kurihara T, Brough DE, Kovesdi I et al, 2000, Selectivity of a replication-competent adenovirus for human breast carcinoma cells expressing the MUC1 antigen. J Clin Invest 106:763–771 Huang TG, Savontaus MJ, Shinozaki K et al, 2003, Telomerasedependent oncolytic adenovirus for cancer treatment. Gene Ther 10:1241–1247 Hernandez-Alcoceba PM, Qian D et al, 2002, New oncolytic adenoviruses with hypoxia- and estrogen receptor-regulated replication. Hum Gene Ther 13:1737–1750 Post DE, Meir VEG, 2003, A novel hypoxia-inducible factor, HIF, activated oncolytic adenovirus for cancer therapy. Oncogene 22:2065–2072 Miyatake S, 2002, Gene therapy using tissue-specific replication competent HSV. Hum Cell 15:130–137 Dabrowska K, Opolski A, Wietrzyk J et al, 2004, Antitumor activity of bacteriophages in murine experimental cancer models caused possibly by inhibition of β3 integrin signaling pathway. Arch Virol 48:241–248 Dabrowska K, Opolski A, Wietrzyk J et al, 2004, Anticancer activity of bacteriophage T4 and its mutant HAP1 in mouse experimental tumor models. Anticancer Res 24:3991–3996 Eriksson F, Culp WD, Massey R et al, 2007, Tumor specific phage particles promote tumor regression in a mouse melanoma model. Cancer Immunol Immunother 56:677–687 Gorski A, Dabrowska K, Switala-Jelen K et al, 2003, New insights into the possible role of bacteriophages in host defense and disease. Med Immunol 2:1–10 Pajtasz-Piasecka E, Rossowska J, Duoe D et al, 2008, Bacteriophages support anti-tumor response initiated by DCbased vaccine against murine transplantable colon carcinoma. Immunol Lett 116:24–32 Ackerman WW, Kurtz H, 1952, A new host-virus system. Proc Soc Exp Biol 81:421–423 Aghi M, Visted T, Depinho RA et al, 2008, Oncolytic herpes virus with defective ICP6 specifically replicates in quiescent cells with homozygous genetic mutations in p16. Oncogene 27:4249– 4254 Aghi MS, Rabkin S, Martuza RL, 2007, Angiogenic response caused by oncolytic herpes simplex virus-induced reduced thrombospondin expression can be prevented by specific viral mutations or by administering thrombospondin-derived peptid. Cancer Res 67:440–444 Ajayi BB, Rabo JS, Baba SS, 2006, Rabies in apparently healthy dogs: histological and immunohistological studies. Niger Postgrad Med 13:128–134 Link N, Aubel C, Kelm JM et al, 2006, Therapeutic protein transduction of mammalian cells and mice by nucleic acid-free lentiviral nanoparticles. Nucleic Acids Res 34:e16 Voelkel C, Galla M, Maetzig T et al, 2010, Protein transduction from retroviral Gag precursors. Proc Natl Acad Sci USA 107:7805–7810 Zhang F, Cong L, Lodato S et al, 2011, Efficient construction of sequence specific TAL effectors for modulating mammalian transcription. Nature Biotech 29:149–153 Kruyt FA, Curiel DT, 2002, Toward a new generation of conditionally replicating adenoviruses: pairing tumor selectivity with maximal oncolysis. Hum Gene Ther 13:485–495 Wickham TJ, 2003, Ligand-directed targeting of genes to the site of disease. Nat Med 9:135–139 Zhou G, Ye GJ, Debinski W et al, 2002, Engineered herpes simplex virus 1 is dependent on IL13Ralpha 2 receptor for cell entry and independent of glycoprotein D receptor interaction. Proc Natl Acad Sci USA 99:15124–15129 Tseng JC, Levin B, Hurtado A et al, 2004, Systemic tumor targeting and killing by Sindbis viral vectors. Nat Biotechnol 22:70–77 Diaz RM, Galivo F, Kottke T et al, 2007, Oncolytic immunovirotherapy for melanoma using vesicular stomatitis virus. Cancer Res 67:2840–2848 Leveille S, Goulet M-L, Lichty BD et al, 2011, Vesicular stomatitis virus oncolytic treatment interferes with tumor-associated dendritic cell functions and abrogates tumor antigen presentation. J Virol 85:12160–12169 Melero I, Hervas-Stubbs S, Glennie M et al, 2007, Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer 7:95–106 Melero I, Shuford WW, Newby SA et al, 1997, Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat Med 3:682–685 Kim EM, Sivanandham M, Stavropoulos CI et al, 2001, Overview analysis of adjuvant therapies for melanomas - a special reference to results from vaccinia melanoma oncolysate adjuvant therapy trials. Surg Oncol 10:53–59 Rudin CM, Cohen EE, Papadimitrakopoulou VA et al, 2003, An attenuated adenovirus, ONYX-015, as mouthwash therapy for premalignant oral dysplasia. J Clin Oncol 21:4546–4552 Nemunaitis J, Ganly I, Khuri F et al, 2000, Selective replication and oncolysis in p53 mutant tumors with ONYX-015, an E1B- 55kD gene-deleted adenovirus, in patients with advanced head and neck cancer: a phase II trial. Cancer Res 60:6359–6366 Nemunaitis J, Khuri F, Ganly I et al, 2001, Phase II trial of intratumoral administration of ONYX-015, a replicationselective adenovirus, in patients with refractory head and neck cancer. J Clin Oncol 19:289–298 Nemunaitis J, Cunningham C, Buchanan A et al, 2001, Intravenous infusion of a replication-selective adenovirus, ONYX-015, in cancer patients: safety, feasibility and biological activity. Gene Ther 8:746–759 Garber K, 2006, China approves world’s first oncolytic virus therapy for cancer treatment. J Nat Can Inst 98:298–300 Frew SE, Sammut SM, Shore AF et al, 2008, Chinese health biotech and the billion-patient market. Nat Biotech 26:37–53 Barbellido AS, Trapero CJ, Sanchez CJ et al, 2008, Gene therapy in the management of oral cancer: review of literature. Medicina oral, patologia oral y cirugia bucal 13:E15–E21 Ganly I, Eckhardt SG, Rodriguez GI et al, 2000, A Phase I study of Onyx-015, an E1B attenuated adenovirus, administered intratumorally to patients with recurrent head and neck cancer. Clin Cancer Res 6:798–806 Lamont JP, Nemunaitis J, Kuhn JA et al, 2000, A prospective phase II trial of ONYX-015 adenovirus and chemotherapy in recurrent squamous cell carcinoma of the head and neck, the Baylor experience). Ann Surg Oncol 7:588–592 Khuri FR, Nemunaitis J, Ganly I et al, 2000, A controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat Med 6:879–885 Mulvihill S, Warren R, Venook A et al, 2001, Safety and feasibility of injection with an E1B-55 kDa gene-deleted, replication-selective adenovirus, ONYX-015, into primary carcinomas of the pancreas: a Phase I trial. Gene Ther 8:308–315 Hecht JR, Bedford RR, Abbruzzese JL et al, 2003, A phase I/II trial of intratumoral endoscopic ultrasound injection of ONYX-015 with intravenous gemcitabine in unresectable pancreatic carcinoma. Clin Cancer Res 9:555–561 Crompton AM, Kirn DH, 2007, From ONYX-105 to armed vaccinia virus: the education and evolution of oncholytic virus development. Curr Cancer Drug Targets 7:133–139 Vasey PA, Shulman LN, Campos S et al, 2002, Phase I trial of intraperitoneal injection of the E1B-55-kd-gene-deleted adenovirus ONYX-015, dl1520, given on days 1 through 5 every 3 weeks in patients with recurrent/refractory epithelial ovarian cancer. J Clin Oncol 20:1562–1569 Hamid O, Varterasian ML, Wadler S et al, 2003, Phase II trial of intravenous CI-1042 in patients with metastatic colorectal cancer. J Clin Oncol 21:1498–1504 Freytag SO, Khil M, Stricker H et al, 2002, Phase I study of replication-competent adenovirus-mediated double suicide gene therapy for the treatment of locally recurrent prostate cancer. Cancer Res 62:4968–4976 Freytag SO, Stricker H, Pegg J et al, 2003, Phase I study of replication-competent adenovirus-mediated double-suicide gene therapy in combination with conventional-dose three-dimensional conformal radiation therapy for the treatment of newly diagnosed, intermediate- to high-risk prostate cancer. Cancer Res 63:7497–7506 Markert JM, Medlock MD, Rabkin SD et al, 2000, Conditionally replicating herpes simplex virus mutant, G207 for the treatment of malignant glioma: results of a Phase I trial. Gene Ther 7:867–874 Rampling R, Cruickshank G, Papanastassiou V et al, 2000, Toxicity evaluation of replication-competent herpes simplex virus, ICP 34.5 null mutant 1716, in patients with recurrent malignant glioma. Gene Ther 7:859–866 Papanastassiou V, Rampling R, Fraser M et al, 2002, The potential for efficacy of the modified [ICP 34.5, -)] herpes simplex virus HSV1716 following intratumoural injection into human malignant glioma: a proof of principle study. Gene Ther 9:398– 406 Bennett JJ, Delman KA, Burt BM et al, 2002, Comparison of safety, delivery, and efficacy of two oncolytic herpes viruses, G207 and NV1020, for peritoneal cancer. Cancer Gene Ther 9:935–945 Yoon SS, Nakamura H, Carroll NM et al, 2000, An oncolytic herpes simplex virus type 1 selectively destroys diffuse liver metastases from colon carcinoma. FASEB J 14:301–311 Kuhn I, Harden P, Bauzon M et al, 2008, Directed evolution generates a novel oncolytic virus for the treatment of colon cancer. PLoS One 3:e2409 MacKie RM, Stewart B, Brown SM, 2001, Intralesional injection of herpes simplex virus 1716 in metastatic melanoma. Lancet 357:525–526 Csatary LK, Gosztonyi G, Szeberenyi J et al, 2004, MTH-68/H oncolytic viral treatment in human high-grade gliomas. J Neurooncol 67:83–93 Freeman AI, Zakay-Rones Z, Gomori JM et al, 2006, Phase I/II trial of intravenous NDV-HUJ oncolytic virus in recurrent glioblastoma multiforme. Mol Ther 13:221–228 Pecora AL, Rizvi N, Cohen GI et al, 2002, Phase I trial of intravenous administration of PV701, an oncolytic virus, in patients with advanced solid cancers. J Clin Oncol 20:2251–2266 Lal R, Harris D, Postel-Vinay S et al, 2009, Reovirus: rationale and clinical trial update. Curr Opin Mol Ther 11:532–539 Thirukkumaran C, Morris DG, 2009, Oncolytic viral therapy using reovirus. Methods Mol Biol 542:607–634 Vile R, Ando D, Kirn D, 2002, The oncolytic virotherapy treatment platform for cancer: unique biological and biosafety points to consider. Cancer Gene Ther 9:1062–1067 Wodarz D, 2003, Gene therapy for killing p53-negative cancer cells: use of replicating versus nonreplicating agents. Hum Gene Ther 14:153–159 Wein LM, Wu JT, Kirn DH, 2003, Validation and analysis of a mathematical model of a replication-competent oncolytic virus for cancer treatment: implications for virus design and delivery. Cancer Res 63:1317–1324 Wakimoto H, Ikeda K, Abe T et al, 2002, The complement response against an oncolytic virus is species-specific in its activation pathways. Mol Ther 5:275–282 Wong RJ, Patel SG, Kim SH et al, 2001, Cytokine gene transfer enhances herpes oncolytic therapy in murine squamous cell carcinoma. Hum Gene Ther 12:253–265 Ikeda K, Wakimoto H, Ichikawa T et al, 2000, Complement depletion facilitates the infection of multiple brain tumors by an intravascular, replication-conditional herpes simplex virus mutant. J Virol 74:4765–4775 Rauen KA, Sudilovsky D, Le JL et al, 2002, Expression of the coxsackie adenovirus receptor in normal prostate and in primary and metastatic prostate carcinoma: potential relevance to gene therapy. Cancer Res 62:3812–3818 Anders M, Christian C, McMahon M et al, 2003, Inhibition of the Raf/MEK/ERK pathway upregulates expression of the coxsackievirus and adenovirus receptor in cancer cells. Cancer Res 63:2088–2095 Araujo RP, Liotta LA, Petricoin EF, 2007, Proteins, drug targets and the mechanism they control: the simple truth about complex networks. Nat Rev Drug Discv 6:871–880 Hann CL, Brahmer JR, 2007, Who should receive epidermal growth factor receptor inhibitors for non-small cell lung cancer and when? Curr Treat Options Oncol 8:28–37 Campbell S, Gromeier, 2005, Oncolytic viruses for cancer therapy II. Cell-internal factors for conditional growth in neoplastic cells. Onkologie 28:209–215 Roberts MS, Lorence RM, Groene WS et al, 2006, Naturally occuring oncolytic viruses for the treatment of cancer. Curr Opin Mol Therap 8:314–321 Roberts MS, Lorence RM, Groene WS et al, 2006, Naturally occuring viruses for treatment of cancer. Disov Medici 6:217–222 Shah AC, Benos D, Gillespie GY et al, 2003, Oncolytic viruses: clinical applications as vectors for the treatment of malignant gliomas. J Neurooncol 65:203–226 Wong HH, Lemoine NR, Wang Y, 2010, Oncolytic viruses for cancer therapy: overcoming the obstacles. Viruses 2:78–106 Kohrt HE, Houot R, Weiskopf K et al, 2012, Stimulation of natural killer cells with a CD137-specific antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer. J Clin Invest 122:1066–1075 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 771 EP 781 DI 10.1007/s12253-012-9548-2 PG 11 ER PT J AU Omar, FMM Ito, K Nga, EM Soo, R Peh, KB Ismail, MT Thakkar, B Soong, R Ito, Y Salto-Tellez, M AF Omar, Feroz Mohd Mohd Ito, Kosei Nga, En Min Soo, Ross Peh, Keow Bee Ismail, Muliana Tuty Thakkar, Bhavin Soong, Richie Ito, Yoshiaki Salto-Tellez, Manuel TI RUNX3 Downregulation in Human Lung Adenocarcinoma is Independent of p53, EGFR or KRAS Status SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR; KRAS; Lung adenocarcinoma; p53; Promoter hypermethylation; RUNX3 ID EGFR; KRAS; Lung adenocarcinoma; p53; Promoter hypermethylation; RUNX3 AB RUNX3 aberrations play a pivotal role in the oncogenesis of breast, gastric, colon, skin and lung tissues. The aim of this study was to characterize further the expression of RUNX3 in lung cancers. To achieve this, a lung cancer tissue microarray (TMA), frozen lung cancer tissues and lung cell lines were examined for RUNX3 expression by immunohistochemistry, while the TMA was also examined for EGFR and p53 expression. RUNX3 promoter methylation status, and EGFR and KRAS mutation status were also investigated. Inactivation of RUNX3 was observed in 70% of the adenocarcinoma samples, and this was associated with promoter hypermethylation but not biased to EGFR/KRAS mutations. Our results suggest a central role of RUNX3 downregulation in pulmonary adenocarcinoma, which may not be dependent of other established cancer-causing pathways and may have important diagnostic and screening implications. C1 [Omar, Feroz Mohd Mohd] National University of Singapore, Cancer Science Institute of Singapore, #12-01, 14 Medical Drive, 117599 Singapore, Singapore. [Ito, Kosei] Nagasaki University, Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, 852-8523 Nagasaki, Japan. [Nga, En Min] National University Hospital, Department of Pathology, 5, Lower Kent Ridge Road, 119074 Singapore, Singapore. [Soo, Ross] National University of Singapore, Cancer Science Institute of Singapore, #12-01, 14 Medical Drive, 117599 Singapore, Singapore. [Peh, Keow Bee] National University of Singapore, Cancer Science Institute of Singapore, #12-01, 14 Medical Drive, 117599 Singapore, Singapore. [Ismail, Muliana Tuty] National University Hospital, Department of Pathology, 5, Lower Kent Ridge Road, 119074 Singapore, Singapore. [Thakkar, Bhavin] National University of Singapore, Cancer Science Institute of Singapore, #12-01, 14 Medical Drive, 117599 Singapore, Singapore. [Soong, Richie] National University of Singapore, Cancer Science Institute of Singapore, #12-01, 14 Medical Drive, 117599 Singapore, Singapore. [Ito, Yoshiaki] National University of Singapore, Cancer Science Institute of Singapore, #12-01, 14 Medical Drive, 117599 Singapore, Singapore. [Salto-Tellez, Manuel] National University of Singapore, Cancer Science Institute of Singapore, #12-01, 14 Medical Drive, 117599 Singapore, Singapore. RP Ito, Y (reprint author), National University of Singapore, Cancer Science Institute of Singapore, 117599 Singapore, Singapore. EM csiitoy@nus.edu.sg CR Brambilla E, Travis WD, Colby TV, Corrin B, Shimosato Y, 2001, The new World Health Organization classification of lung tumours. Eur Respir J 18(6):1059–1068 Mattson ME, Pollack ES, Cullen JW, 1987, What are the odds that smoking will kill you? Am J Public Health 77(4):425–431 Hessel PA, Gamble JF, McDonald JC, 2005, Asbestos, asbestosis, and lung cancer: a critical assessment of the epidemiological evidence. Thorax 60(5):433–436 Lubin JH, Boice JD Jr, 1997, Lung cancer risk from residential radon: meta-analysis of eight epidemiologic studies. J Natl Cancer Inst 89(1):49–57 Zhu CQ, ShihW, Ling CH, Tsao MS, 2006, Immunohistochemical markers of prognosis in non-small cell lung cancer: a review and proposal for a multiphase approach to marker evaluation. J Clin Pathol 59(8):790–800 Belinsky SA, Nikula KJ, Palmisano WA, Michels R, Saccomanno G, Gabrielson E et al, 1998, Aberrant methylation of p16INK4a is an early event in lung cancer and a potential biomarker for early diagnosis. Proc Natl Acad Sci 95(20):11891–11896 Bouchardy C, Benhamou S, Dayer P, 1996, The effect of tobacco on lung cancer risk depends on CYP2D6 activity. Cancer Res 56, 2):251–253 El-Zein RA, Schabath MB, Etzel CJ, Lopez MS, Franklin JD, Spitz MR, 2006, Cytokinesis-blocked micronucleus assay as a novel biomarker for lung cancer risk. Cancer Res 66(12):6449–6456 Marchand LL, Sivaraman L, Pierce L, Seifried A, Lum A, Wilkens LR et al, 1998, Associations of CYP1A1, GSTM1, and CYP2E1 polymorphisms with lung cancer suggest cell type specificities to tobacco carcinogens. Cancer Res 58(21):4858–4863 SchwartzAG, PrysakGM, Bock CH, CoteML, 2007, The molecular epidemiology of lung cancer. Carcinogenesis 28(3):507–518 Sellers TA, Potter JD, Bailey-Wilson JE, Rich SS, Rothschild H, Elston RC, 1992, Lung Cancer Detection and Prevention: Evidence for an Interaction between Smoking and Genetic Predisposition. Cancer Res. 52(9_Supplement):2694s-7 Shaw GL, Falk RT, Frame JN, Weiffenbach B, Nesbitt JC, Pass HI et al, 1998, Genetic polymorphism of CYP2D6 and lung cancer risk. Cancer Epidemiol Biomarkers Prev 7(3):215–219 Wistuba II, Mao L, Gazdar AF, 2002, Smoking molecular damage in bronchial epithelium. Oncogene 21(48):7298–7306 Yamabuki T, Takano A, Hayama S, Ishikawa N, Kato T, Miyamoto M et al, 2007, Dikkopf-1 as a novel serologic and prognostic biomarker for lung and esophageal carcinomas. Cancer Res 67, 6):2517–2525 Arenberg D, 2004, In search of the holy grail: lung cancer biomarkers. Chest 126(2):325–326 Altshuler D, Daly MJ, Lander ES, 2008, Genetic mapping in human disease. Science 322(5903):881–888 Duffy MJ, Crown J, 2008, A personalized approach to cancer treatment: how biomarkers can help. Clin Chem 54(11):1770– 1779 Brambilla C, Fievet F, Jeanmart M, de Fraipont F, Lantuejoul S, Frappat V et al, 2003, Early detection of lung cancer: role of biomarkers. Eur Respir J. 21(39_suppl):36S-44 Bangsow C, Rubins N, Glusman G, Bernstein Y, Negreanu V, Goldenberg D et al, 2001, The RUNX3 gene–sequence, structure and regulated expression. Gene 279(2):221–232 Ito Y, 2008, RUNX genes in development and cancer: regulation of viral gene expression and the discovery of RUNX family genes. Adv Cancer Res 99:33–76 Hanai J-i, Chen LF, Kanno T, Ohtani-Fujita N, Kim WY, GuoW-H et al, 1999, Interaction and Functional Cooperation of PEBP2/ CBF with Smads. Synergistic induction of the immunoglobulin germline Cα promoter. J Biol Chem 274(44):31577–31582 Ito K, Lim AC-B, Salto-Tellez M, Motoda L, Osato M, Chuang LSH et al, 2008, RUNX3 attenuates β-Catenin/T cell factors in intestinal tumorigenesis. Cancer Cell 14(3):226–237 Licchesi JDF, Westra WH, Hooker CM, Machida EO, Baylin SB, Herman JG, 2008, Epigenetic alteration ofWnt pathway antagonists in progressive glandular neoplasia of the lung. Carcinogenesis 29, 5):895–904 Homma N, Tamura G, Honda T, Matsumoto Y, Nishizuka S, Kawata S et al, 2006, Spreading of methylation within RUNX3 CpG island in gastric cancer. Cancer Sci 97(1):51–56 Lee JM, Kwon HJ, Bae SC, Jung HS, 2010, Lung tissue regeneration after induced injury in Runx3 KO mice. Cell Tissue Res 341(3):465–470 Lee KS, Lee YS, Lee JM, Ito K, Cinghu S, Kim JH et al, 2010, Runx3 is required for the differentiation of lung epithelial cells and suppression of lung cancer. Oncogene 29(23):3349–3361 Lau QC, Raja E, Salto-Tellez M, Liu Q, Ito K, Inoue Met al, 2006, RUNX3 is frequently inactivated by dual mechanisms of protein mislocalization and promoter hypermethylation in breast cancer. Cancer Res 66(13):6512–6520 Subramaniam MM, Chan JY, Soong R, Ito K, Ito Y, Yeoh KG et al, 2008, RUNX3 inactivation by frequent promoter hypermethylation and protein mislocalization constitute an early event in breast cancer progression. Breast Cancer Res Treat Ito K, Liu Q, Salto-Tellez M, Yano T, Tada K, Ida H et al, 2005, RUNX3, a novel tumor suppressor, is frequently inactivated in gastric cancer by protein mislocalization. Cancer Res 65(17):7743– 7750 Ku J-L, Kang S-B, Shin Y-K, Kang HC, Hong S-H, Kim I-J et al, 2004, Promoter hypermethylation downregulates RUNX3 gene expression in colorectal cancer cell lines. Oncogene 23(40):6736– 6742 Li QL, Ito K, Sakakura C, Fukamachi H, Inoue K, Chi XZ et al, 2002, Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell 109(1):113–124 Salto-Tellez M, Peh BK, Ito K, Tan SH, Chong PY, Han HC et al, 2006, RUNX3 protein is overexpressed in human basal cell carcinomas. Oncogene 25(58):7646–7649 Kim TY, Lee HJ, Hwang KS, Lee M, Kim JW, Bang YJ et al, 2004, Methylation of RUNX3 in various types of human cancers and premalignant stages of gastric carcinoma. Lab Invest 84, 4):479–484 Yanada M, Yaoi T, Shimada J, Sakakura C, Nishimura M, Ito K et al, 2005, Frequent hemizygous deletion at 1p36 and hypermethylation downregulate RUNX3 expression in human lung cancer cell lines. Oncol Rep 14(4):817–822 Lane D, 2004, p53 from pathway to therapy. Carcinogenesis 25, 7):1077–1081 Cheng Y-L, Lee S-C, Harn H-J, Chen C-J, Chang Y-C, Chen J-C et al, 2003, Prognostic prediction of the immunohistochemical expression of p53 and p16 in resected non-small cell lung cancer. Eur J Cardiothorac Surg 23(2):221–228 Maniwa Y, Yoshimura M, Obayashi C, Inaba M, Kiyooka K, Kanki M et al, 2001, Association of p53 gene mutation and telomerase activity in resectable non-small cell lung cancer*. Chest 120(2):589–594 Piyathilake CJ, Frost AR, Manne U, Weiss H, Heimburger DC, Grizzle WE, 2003, Nuclear accumulation of p53 is a potential marker for the development of squamous cell lung cancer in smokers*. Chest 123(1):181–186 Ettinger DS, 2006, Clinical implications of EGFR expression in the development and progression of solid tumors: focus on nonsmall cell lung cancer. Oncologist 11(4):358–373 Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW et al, 2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350(21):2129–2139 Jang TW, Oak CH, Chang HK, Suo SJ, Jung MH, 2009, EGFR and KRAS mutations in patients with adenocarcinoma of the lung. Korean J Intern Med 24(1):48–54 Shigematsu H, Lin L, Takahashi T, Nomura M, Suzuki M,Wistuba II et al, 2005, Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst 97(5):339–346 Schmid K, Oehl N, Wrba F, Pirker R, Pirker C, Filipits M, 2009, EGFR/KRAS/BRAF mutations in primary lung adenocarcinomas and corresponding locoregional lymph node metastases. Clin Cancer Res 15(14):4554–4560 Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst RS et al, 2005, Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol 23, 25):5900–5909 Tsunematsu T, Kudo Y, Iizuka S, Ogawa I, Fujita T, Kurihara H et al, 2009, RUNX3 has an oncogenic role in head and neck cancer. PLoS One 4(6):e5892 Yanagawa N, Tamura G, Oizumi H, Endoh M, Sadahiro M, Motoyama T, 2011, Inverse correlation between EGFR mutation and FHIT, RASSF1A and RUNX3 methylation in lung adenocarcinoma: relation with smoking status. Anticancer Res 31(4):1211– 1214 Das K, Mohd Omar MF, Ong CW, Bin Abdul Rashid S, Peh BK, Putti TC et al, 2008, TRARESA: a tissue microarray-based hospital systemfor biomarker validation and discovery. Pathology 40, 5):441–449 Yano T, Ito K, Fukamachi H, Chi X-Z, Wee H-J, Inoue K-i et al, 2006, The RUNX3 tumor suppressor upregulates bim in gastric epithelial cells undergoing transforming growth factor β-induced apoptosis. Mol Cell Biol 26(12):4474–4488 Soong R, Robbins PD, Dix BR, Grieu F, Lim B, Knowles S et al, 1996, Concordance between p53 protein overexpression and gene mutation in a large series of common human carcinomas. Hum Pathol 27(10):1050–1055 Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB, 1996, Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 93, 18):9821–9826 Krypuy M, Newnham G, Thomas D, Conron M, Dobrovic A, 2006, High resolution melting analysis for the rapid and sensitive detection of mutations in clinical samples: KRAS codon 12 and 13 mutations in non-small cell lung cancer. BMC Cancer 6(1):295 Chin TM, Anuar D, Soo R, Salto-Tellez M, Li WQ, Ahmad B et al, 2007, Detection of epidermal growth factor receptor variations by partially denaturing HPLC. Clin Chem 53(1):62–70 Araki K, Osaki M, Nagahama Y, Hiramatsu T, Nakamura H, Ohgi S et al, 2005, Expression of RUNX3 protein in human lung adenocarcinoma: implications for tumor progression and prognosis. Cancer Sci 96(4):227–231 Ito K, Inoue Ki, Bae SC, Ito Y, 2009, Runx3 expression in gastrointestinal tract epithelium: resolving the controversy. Oncogene 28(10):1379–1384 Li QL, Kim HR, Kim WJ, Choi JK, Lee YH, Kim HM et al, 2004, Transcriptional silencing of the RUNX3 gene by CpG hypermethylation is associated with lung cancer. Biochem Biophys Res Commun 314(1):223–228 Sato K, Tomizawa Y, Iijima H, Saito R, Ishizuka T, Nakajima T et al, 2006, Epigenetic inactivation of the RUNX3 gene in lung cancer. Oncol Rep 15(1):129–135 Yanagawa N, Tamura G, Oizumi H, Takahashi N, Shimazaki Y, Motoyama T, 2003, Promoter hypermethylation of tumor suppressor and tumor-related genes in non-small cell lung cancers. Cancer Sci 94, 7):589–592 Ibrahim AEK, Arends MJ, Silva A-L, Wyllie AH, Greger L, Ito Y et al, 2010, Sequential DNA methylation changes are associated with DNMT3B overexpression in colorectal neoplastic progression. Gut Soung YH, Lee JW, Kim SY, Seo SH, Park WS, Nam SW et al, 2005, Mutational analysis of EGFR and K-RAS genes in lung adenocarcinomas. Virchows Arch 446(5):483–488 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 783 EP 792 DI 10.1007/s12253-011-9485-5 PG 10 ER PT J AU Calabuig-Farinas, S Benso, GR Szuhai, K Machado, I Lopez-Guerrero, AJ de Jong, D Peydro, A San Miguel, T Navarro, L Pellin, A LLombart-Bosch, A AF Calabuig-Farinas, Silvia Benso, Gil Rosario Szuhai, Karoly Machado, Isidro Lopez-Guerrero, Antonio Jose de Jong, Danielle Peydro, Amando San Miguel, Teresa Navarro, Lara Pellin, Antonio LLombart-Bosch, Antonio TI Characterization of a New Human Cell Line (CH-3573) Derived from a Grade II Chondrosarcoma with Matrix Production SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bone tumor; Cell line; Chondrosarcoma; Grade II; Karyotype; Matrix ID Bone tumor; Cell line; Chondrosarcoma; Grade II; Karyotype; Matrix AB Chondrosarcomas are malignant cartilageforming tumors that represent the third most common malignant solid tumor of bone. In patients with grades II and III, local recurrence, increasing tumor size and dedifferentiation have been associated with lower survival rates. These biologically poorly-understood neoplasms vary considerably in clinical presentation and biological behavior. Cytogenetic studies have shown that heterogeneity is related to karyotypic complexity; moreover, alterations in the 9p21 locus and TP53 gene are related to disease progression. Despite the relatively high frequency of chondrosarcoma only a limited number of cell lines exist in the scientific community, limiting the possibility to study hypothesisderived research or primary drug interaction necessary for pre-clinical studies. We report a chondrosarcoma cell line, CH-3573, derived from a primary tumor that may serve as a useful tool for both in vitro and in vivo models to study the molecular pathogenesis. In addition, xenograft passages in nude mice were studied to characterize the genetic stability over the course of tumor progression. In contrary to other reported cell lines, an important feature of our established cell line was the retained matrix production, a characteristic feature of a conventional grade II chondrosarcoma. The cell line (CH-3573) was characterized by pathological, immunohistochemical and molecular genetic methods. C1 [Calabuig-Farinas, Silvia] University of Valencia, Department of Pathology, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain. [Benso, Gil Rosario] University of Valencia, Department of Pathology, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain. [Szuhai, Karoly] Leiden University, Medical Center, Department of Molecular Cell BiologyLeiden, The Netherlands. [Machado, Isidro] University of Valencia, Department of Pathology, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain. [Lopez-Guerrero, Antonio Jose] Fundacion Instituto Valenciano de Oncologia, Laboratory of Molecular BiologyValencia, Spain. [de Jong, Danielle] Leiden University, Medical Center, Department of Molecular Cell BiologyLeiden, The Netherlands. [Peydro, Amando] University of Valencia, Department of Pathology, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain. [San Miguel, Teresa] University of Valencia, Department of Pathology, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain. [Navarro, Lara] University of Valencia, Department of Pathology, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain. [Pellin, Antonio] University of Valencia, Department of Pathology, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain. [LLombart-Bosch, Antonio] University of Valencia, Department of Pathology, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain. RP LLombart-Bosch, A (reprint author), University of Valencia, Department of Pathology, 46010 Valencia, Spain. EM antonio.llombart@uv.es CR Evans HL, Ayala AG, Romsdahl MM, 1977, Prognostic factors in chondrosarcoma of bone: a clinicopathologic analysis with emphasis on histologic grading. Cancer 40(2):818–31 Rozeman LB, Cleton-Jansen AM, Hogendoorn PC, 2006, Pathology of primary malignant bone and cartilage tumour. Int Orthop 30, 6):437–44 Puri A, Shah M, Agarwal MG, Jambhekar NA, Basappa P, 2009, Chondrosarcoma of bone: does the size of the tumor, the presence of a pathologic fracture, or prior intervention have an impact on local control and survival? J Cancer Res Ther 5(1):14–9 Riedel RF, Larrier N, Dodd L, Kirsch D, Martinez S, Brigman BE, 2009, The clinical management of chondrosarcoma. Curr Treat Options Oncol 10(1–2):94–106 Takigawa M, Tajima K, Pan HO et al, 1989, Establishment of a clonal human chondrosarcoma cell line with cartilage phenotypes. Cancer Res 49(14):3996–4002 Takigawa M, Pan HO, Kinoshita A, Tajima K, Takano Y, 1991, Establishment from a human chondrosarcoma of a new immortal cell line with high tumorigenicity in vivo, which is able to form proteoglycan-rich cartilage-like nodules and to respond to insulin in vitro. Int J Cancer 48(5):717–25 Ikemoto S, Sugimura K, Yoshida N, Nakatani T, 2004, Chondrosarcoma of the urinary bladder and establishment of a human chondrosarcoma cell line, OCUU-6). Hum Cell 17(3):93–6 Kudo N, Ogose A, Hotta T et al, 2007, Establishment of novel human dedifferentiated chondrosarcoma cell line with osteoblastic differentiation. Virchows Arch 451(3):691–9 Yang L, Chen Q, Zhang S, Wang X, Li W, Wen J et al, 2009, A novel mutated cell line with characteristics of dedifferentiated chondrosarcoma. Int J Mol Med 24(4):427–35 Gil-Benso R, Lopez-Gines C, Lopez-Guerrero JA et al, 2003, Establishment and characterization of a continuous human chondrosarcoma cell line, ch-2879: comparative histologic and genetic studies with its tumor of origin. Lab Invest 83(6):877–87 Kalinski T, Krueger S, Pelz AF et al, 2005, Establishment and characterization of the permanent human cell line C3842 derived from a secondary chondrosarcoma in Ollier’s disease. Virchows Arch 446(3):287–99 Oshiro Y, Chaturvedi V, Hayden D et al, 1998, Altered p53 is associated with aggressive behavior of chondrosarcoma: a long term follow-up study. Cancer 83(11):2324–34 Terek RM, Healey JH, Garin-Chesa P, Mak S, Huvos A, Albino AP, 1998, p53 mutations in chondrosarcoma. Diagn Mol Pathol 7, 1):51–6 Scully SP, Layfield LJ, Harrelson JM, 1997, Prognostic markers in chondrosarcoma: evaluation of cell proliferation and of regulators of the cell cycle. Sarcoma; 1(2):79–87 Szuhai K, Tanke HJ, 2006, COBRA: combined binary ratio labeling of nucleic-acid probes for multi-color fluorescence in situ hybridization karyotyping. Nat Protoc 1(1):264–75 Lopez-Guerrero JA, Lopez-Gines C, Pellin A, Carda C, Llombart- Bosch A, 2004, Deregulation of the G1 to S-phase cell cycle checkpoint is involved in the pathogenesis of human osteosarcoma. Diagn Mol Pathol 13(2):81–91 Lopez-Guerrero JA, Pellin A, Noguera R, Carda C, Llombart- Bosch A, 2001, Molecular analysis of the 9p21 locus and p53 genes in Ewing family tumors. Lab Invest 81(6):803–14 Okamoto S, Hisaoka M, Ishida T et al, 2001, Extraskeletal myxoid chondrosarcoma: a clinicopathologic, immunohistochemical, and molecular analysis of 18 cases. Hum Pathol 32(10):1116– 24 Tallini G, Dorfman H, Brys P et al, 2002, Correlation between clinicopathological features and karyotype in 100 cartilaginous and chordoid tumours A report from the Chromosomes and Morphology, CHAMP, Collaborative Study Group. J Pathol 196(2):194– 203 Mandahl N, Gustafson P, Mertens F et al, 2002, Cytogenetic aberrations and their prognostic impact in chondrosarcoma Genes Chromosomes. Cancer 33(2):188–200 Bovee JV, Sciot R, Dal Cin P et al, 2001, Chromosome 9 alterations and trisomy 22 in central chondrosarcoma: a cytogenetic and DNA flow cytometric analysis of chondrosarcoma subtypes. Diagn Mol Pathol 10(4):228–35 Hallor KH, Staaf J, Bovee JV et al, 2009, Genomic profiling of chondrosarcoma: chromosomal patterns in central and peripheral tumors. Clin Cancer Res 15(8):2685–94 Rozeman LB, Szuhai K, Schrage YM et al, 2006, Arraycomparative genomic hybridization of central chondrosarcoma: identification of ribosomal protein S6 and cyclin-dependent kinase 4 as candidate target genes for genomic aberrations. Cancer 107, 2):380–8 Larramendy ML, Tarkkanen M, Valle J et al, 1997, Gains, losses, and amplifications of DNA sequences evaluated by comparative genomic hybridization in chondrosarcomas. Am J Pathol 150, 2):685–91 Yamaguchi T, Toguchida J, Wadayama B et al, 1996, Loss of heterozygosity and tumor suppressor gene mutations in chondrosarcomas. Anticancer Res 16(4A):2009–15 Eisenberg MB, Woloschak M, Sen C, Wolfe D, 1997, Loss of heterozygosity in the retinoblastoma tumor suppressor gene in skull base chordomas and chondrosarcomas. Surg Neurol 47, 2):156–60, discussion 60-1 Gunawan B, Weber M, Bergmann F, Wildberger J, Niethard FU, Fuzesi L, 2000, Clonal chromosome abnormalities in enchondromas and chondrosarcomas. Cancer Genet Cytogenet 120(2):127–30 Asp J, Inerot S, Block JA, Lindahl A, 2001, Alterations in the regulatory pathway involving p16, pRb and cdk4 in human chondrosarcoma. J Orthop Res 19(1):149–54 Jagasia AA, Block JA, Qureshi A et al, 1996, Chromosome 9 related aberrations and deletions of the CDKN2 and MTS2 putative tumor suppressor genes in human chondrosarcomas. Cancer Lett 105(1):91–103 Jagasia AA, Block JA, Diaz MO et al, 1996, Partial deletions of the CDKN2 and MTS2 putative tumor suppressor genes in a myxoid chondrosarcoma. Cancer Lett 105(1):77–90 Asp J, Brantsing C, Benassi MS et al, 2001, Changes in p14(ARF, do not play a primary role in human chondrosarcoma tissues. Int J Cancer 93(5):703–5 Schrage YM, Lam S, Jochemsen AG et al, 2009, Central chondrosarcoma progression is associated with pRb pathway alterations: CDK4 down-regulation and p16 overexpression inhibit cell growth in vitro. J Cell Mol Med 13(9A):2843–52 Moussavi-Harami F, Mollano A, Martin JA et al, 2006, Intrinsic radiation resistance in human chondrosarcoma cells. Biochem Biophys Res Commun 346(2):379–85 Rozeman LB, Hogendoorn PC, Bovee JV, 2002, Diagnosis and prognosis of chondrosarcoma of bone. Expert Rev Mol Diagn 2, 5):461–72 Bovee JV, van Royen M, Bardoel AF et al, 2000, Near-haploidy and subsequent polyploidization characterize the progression of peripheral chondrosarcoma. Am J Pathol 157(5):1587–95 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 793 EP 802 DI 10.1007/s12253-012-9505-0 PG 10 ER PT J AU Annunziata, C Buonaguro, L Buonaguro, MF Tornesello, LM AF Annunziata, Clorinda Buonaguro, Luigi Buonaguro, M Franco Tornesello, Lina Maria TI Characterization of the Human Papillomavirus (HPV) Integration Sites into Genital Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human Papillomavirus; HPV; Integration sites; Penile cancer; Vulvar Cancer ID Human Papillomavirus; HPV; Integration sites; Penile cancer; Vulvar Cancer AB Oncogenic HPVs have been found frequently integrated into human genome of invasive cancers and chromosomal localization has been extensively investigated in cervical carcinoma. Few studies have analyzed the HPV integration loci in other genital cancers. We have characterized the integration sites of HPV16 in invasive penile carcinoma by means of Alu-HPV-based PCR. Nucleotide sequence analysis of viral–human DNA junctions showed that HPV integration occurredin one case within the chromosome 8q21.3 region, in which the FAM92A1 gene is mapped, and in the second case inside the chromosome 16p13.3, within the intronic region of TRAP1 gene. These results confirm previous observations, summarized in a systematic review of the literature, on the HPV integration events in gene loci relevant to cancer pathogenesis. C1 [Annunziata, Clorinda] National Cancer Institute, Molecular Biology and Viral Oncology and AIDS Reference Centre, Cappella Cangiani, 80131 Naples, Italy. [Buonaguro, Luigi] National Cancer Institute, Molecular Biology and Viral Oncology and AIDS Reference Centre, Cappella Cangiani, 80131 Naples, Italy. [Buonaguro, M Franco] National Cancer Institute, Molecular Biology and Viral Oncology and AIDS Reference Centre, Cappella Cangiani, 80131 Naples, Italy. [Tornesello, Lina Maria] National Cancer Institute, Molecular Biology and Viral Oncology and AIDS Reference Centre, Cappella Cangiani, 80131 Naples, Italy. RP Tornesello, LM (reprint author), National Cancer Institute, Molecular Biology and Viral Oncology and AIDS Reference Centre, 80131 Naples, Italy. EM m.tornesello@istitutotumori.na.it;irccsvir@unina.it CR Zur Hausen H, 2002, Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer 2(5):342–350 Bouvard V, Baan R, Straif K et al, 2009, A review of human carcinogens–Part B: biological agents. Lancet Oncol 10(4):321– 322 Parkin DM, Bray F, 2006, Chapter 2: The burden of HPV-related cancers. Vaccine 24(Suppl 3):S11–S25 Dunne EF, Nielson CM, Stone KM et al, 2006, Prevalence of HPV infection among men: A systematic review of the literature. J Infect Dis 194(8):1044–1057 Bruni L, Diaz M, Castellsague X et al, 2010, Cervical Human Papillomavirus Prevalence in 5 Continents: Meta-Analysis of 1 Million Women with Normal Cytological Findings. J Infect Dis. Palefsky JM, 2007, HPV infection in men. Dis Markers 23, 4):261–272 Daling JR, Madeleine MM, Schwartz SM et al, 2002, A population-based study of squamous cell vaginal cancer: HPV and cofactors. Gynecol Oncol 84(2):263–270 Gillison ML, Shah KV, 2003, Chapter 9: Role of mucosal human papillomavirus in nongenital cancers. J Natl Cancer Inst Monogr, 31):57–65. Smith JS, Backes DM, Hoots BE et al, 2009, Human papillomavirus type-distribution in vulvar and vaginal cancers and their associated precursors. Obstet Gynecol 113(4):917–924 Steenbergen RD, De WJ, Wilting SM et al, 2005, HPV-mediated transformation of the anogenital tract. J Clin Virol 32(Suppl 1): S25–S33 Wentzensen N, Vinokurova S, von Knebel DM, 2004, Systematic review of genomic integration sites of human papillomavirus genomes in epithelial dysplasia and invasive cancer of the female lower genital tract. Cancer Res 64(11):3878–3884 Kraus I, Driesch C, Vinokurova S et al, 2008, The majority of viral-cellular fusion transcripts in cervical carcinomas cotranscribe cellular sequences of known or predicted genes. Cancer Res 68, 7):2514–2522 Matovina M, Sabol I, Grubisic G et al, 2009, Identification of human papillomavirus type 16 integration sites in high-grade precancerous cervical lesions. Gynecol Oncol 113(1):120–127 Couturier J, Sastre-Garau X, Schneider-Maunoury S et al, 1991, Integration of papillomavirus DNA near myc genes in genital carcinomas and its consequences for proto-oncogene expression. J Virol 65(8):4534–4538 Kalantari M, Villa LL, Calleja-Macias IE et al, 2008, Human papillomavirus-16 and −18 in penile carcinomas: DNA methylation, chromosomal recombination and genomic variation. Int J Cancer 123(8):1832–1840 Tornesello ML, Buonaguro FM, Meglio A et al, 1997, Sequence variations and viral genomic state of human papillomavirus type 16 in penile carcinomas from Ugandan patients. J Gen Virol 78(Pt 9):2199–2208 Pett M, Coleman N, 2007, Integration of high-risk human papillomavirus: a key event in cervical carcinogenesis? J Pathol 212, 4):356–367 Bodily J, Laimins LA, 2011, Persistence of human papillomavirus infection: keys to malignant progression. Trends Microbiol 19, 1):33–39 Kraus I, Driesch C, Vinokurova S et al, 2008, The majority of viral-cellular fusion transcripts in cervical carcinomas cotranscribe cellular sequences of known or predicted genes. Cancer Res 68, 7):2514–2522 Liu D, Hu J, Agorreta J et al, 2010, Tumor necrosis factor receptor-associated protein 1(TRAP1, regulates genes involved in cell cycle and metastases. Cancer Lett 296(2):194–205 Liang S, Gong F, Zhao X et al, 2009, Prokaryotic expression, purification of a new tumor-relative protein FAM92A1-289 and its characterization in renal cell carcinoma. Cancer Lett 276(1):81–87 Solimini NL, Luo J, Elledge SJ, 2007, Non-oncogene addiction and the stress phenotype of cancer cells. Cell 130(6):986–988 Liu D, Hu J, Agorreta J et al, 2010, Tumor necrosis factor receptor-associated protein 1(TRAP1, regulates genes involved in cell cycle and metastases. Cancer Lett 296(2):194–205 Nakanishi G, Fujii K, Asagoe K et al, 2009, Human papillomavirus genome integration in multifocal vulvar Bowen’s disease and squamous cell carcinoma. Clin Exp Dermatol 34, 8):e965–e967 Dall KL, Scarpini CG, Roberts I et al, 2008, Characterization of naturally occurring HPV16 integration sites isolated from cervical keratinocytes under noncompetitive conditions. Cancer Res 68, 20):8249–8259 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 803 EP 808 DI 10.1007/s12253-012-9507-y PG 6 ER PT J AU Chen, M Chen, Tsh Lu, Yy Liu, Chy Qu, Jl AF Chen, Min Chen, Tong-sheng Lu, Ying-ying Liu, Cheng-yi Qu, Jun-le TI Dihydroarteminsin-Induced Apoptosis is not Dependent on the Translocation of Bim to the Endoplasmic Reticulum in Human Lung Adenocarcinoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bim; Dihydroartemisinin (DHA); Endoplasmic reticulum (ER); Apoptosis; Human lung adenocarcinoma cells ID Bim; Dihydroartemisinin (DHA); Endoplasmic reticulum (ER); Apoptosis; Human lung adenocarcinoma cells AB Bim, a proapoptotic BH3-only member of Bcl-2 family, has been considered to play an important role in initiating mitochondrial apoptotic pathway. Our previous studies have shown the ability of dihydroarteminsin (DHA) to induce apoptosis in human lung adenocarcinoma (ASTC-a-1) cells. In this study, we investigated the function of Bim during DHA-induced apoptosis in ASTC-a-1 and another human lung adenocarcinoma (A549) cell lines. Confocal imaging of single living cell expressing GFP-BimL showed the translocation of Bim to endoplasmic reticulum (ER) rather than mitochondria during DHA-induced apoptosis.Moreover, we also found that DHA induced ER stress and an increase of Bim protein levels. However, silencing Bim by short hairpin RNA did not inhibit DHA-induced caspase-9 activation and cell apoptosis. Taken together, our results demonstrate for the first time that DHA induces Bim translocation to ER, but DHA-induced apoptosis is not dependent on Bim in ASTCa-1 and A549 cell lines. C1 [Chen, Min] South China Normal University, MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, 510631 Guangzhou, China. [Chen, Tong-sheng] South China Normal University, MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, 510631 Guangzhou, China. [Lu, Ying-ying] South China Normal University, MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, 510631 Guangzhou, China. [Liu, Cheng-yi] South China Normal University, Laboratory of Laser Sports Medicine, 510631 Guangzhou, China. [Qu, Jun-le] Shenzhen University, Institute of Optoelectronics, 518060 Shenzhen, China. RP Chen, Tsh (reprint author), South China Normal University, MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, 510631 Guangzhou, China. EM chentsh@scnu.edu.cn CR Wang XW, Xing D, Liu L et al, 2009, BimL directly neutralizes Bcl-xL to promote Bax activation during UV-induced apoptosis. FEBS Lett 583:1873–1879 Yin KJ, Hsu CY, Hu XY et al, 2006, Protein phosphatase 2A regulates bim expression via the Akt/FKHRL1 signaling pathway in amyloidpeptide-induced cerebrovascular endothelial cell death. J Neurosci 26:2290–2299 Schneiders UM, Schyschka L, Rudy A et al, 2009, BH3-only proteins Mcl-1 and Bim as well as endonuclease G are targeted in spongistatin 1–induced apoptosis in breast cancer cells. Mol Cancer Ther 8:2914–2925 Khawaja NR, Carre M, Kovacic H et al, 2008, Patupilone-induced apoptosis is mediated by mitochondrial reactive oxygen species through Bim relocalization to mitochondria. Mol Pharmacol 74:1072–1083 Szegezdi E, Logue SE, Gorman AM et al, 2006, Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep 7:880–885 Merino D, Giam M, Hughes PD et al, 2009, The role of BH3-only protein Bim extends beyond inhibiting Bcl-2–like prosurvival proteins. J Cell Biol 186:355–362 Morishima N, Nakanishi K, Tsuchiya K et al, 2004, Translocation of Bim to the endoplasmic reticulum, ER, mediates ER stress signaling for activation of caspase-12 during ER stress-induced apoptosis. J Biol Chem 279:50375–50381 Puthalakath H, O’Reilly LA, Gunn P et al, 2007, ER stress triggers apoptosis by activating BH3-only protein Bim. Cell 129:1337– 1349 Chin TY, Lin HC, Kuo JP et al, 2007, Dual effect of thapsigargin on cell death in porcine aortic smooth muscle cells. Am J Physiolcell Ph 292:383–395 Morishima N, Nakanishi K, Takenouchi H et al, 2002, An endoplasmic reticulum stress-specific caspase cascade in apoptosis. J Biol Chem 277:34287–34294 Dhingra V, Rao KV, Narasu ML, 2000, Current status of artemisinin and its derivatives as antimalarial drugs. Life Sci 66:279–300 Lu YY, Chen TS, Wang XP et al, 2010, Single-cell analysis of dihydroartemisinin-induced apoptosis through reactive oxygen species-mediated caspase-8 activation and mitochondrial pathway in ASTC-a-1 cells using fluorescence imaging techniques. J Biomed Opt 15:046028-1–16 Lu YY, Chen TS, Wang XP et al, 2010, The JNK inhibitor SP600125 enhances dihydroartemisinin-induced apoptosis by accelerating Bax translocation into mitochondria in human lung adenocarcinoma cells. FEBS Lett 584:4019–4026 Wang SJ, GaoY CH et al, 2010, Dihydroartemisinin inactivates NF-κB and potentiates the anti-tumor effect of gemcitabine on pancreatic cancer both in vitro and in vivo. Cancer Lett 293:99– 108 Handrick R, Ontikatze T, Bauer KD et al, 2010, Dihydroartemisinin induces apoptosis by a Bak-dependent intrinsic pathway. Mol Cancer Ther 9:2497–2510 Mercer AE, Maggs JL, Sun XM et al, 2007, Evidence for the involvement of carbon-centered radicals in the induction of apoptotic cell death by artemisinin compounds. J Biol Chem 282:9372–9382 Hou JM, Wang DS, Zhang RWet al, 2008, Experimental therapy of hepatoma with artemisinin and its derivatives: in vitro and in vivo activity, chemosensitization, and mechanisms of action. Clin Cancer Res 14:5519–5530 Zhang YJ, Xing D, Liu L, 2009, PUMA promotes Bax translocation by both directly interacting with Bax and by competitive binding to Bcl-XL during UV-induced apoptosis. Mol Biol Cell 20:3077–3087 Zhang WW, Wang ZP, Chen TS, 2010, Curcumol induces apoptosis via caspases-independent mitochondrial pathway in human lung adenocarcinoma ASTC-a-1 cells. Med Oncol 28:307–314 Stan SD, Hahm ER, Warin R et al, 2008, Withaferin A causes FOXO3a- and Bim-dependent apoptosis and inhibits growth of human breast cancer cells in vivo. Cancer Res 68:7661–7669 Nakayama Y, Endo M, Tsukano H et al, 2010, Molecular mechanisms of the LPS-induced non-apoptotic ER stress-CHOP pathway. J Biochem 147:471–483 Lu JJ, Chen SM, Zhang XWet al, 2010, The anti-cancer activity of dihydroartemisinin is associated with induction of iron-dependent endoplasmic reticulum stress in colorectal carcinoma HCT116 cells. Invest New Drug., DOI 10.1007/s10637-010-9481-8 Mohamed R, Mayo M, Dash R et al, 2010, Melanoma differentiation associated gene-7/interleukin-24 potently induces apoptosis in human myeloid leukemia cells through a process regulated by endoplasmic reticulum stress. Mol Pharmacol 78:1096–1104 Kim H, Tu HC, Ren D et al, 2009, Stepwise activation of BAX and BAK by tBID, BIM, and PUMA initiates mitochondrial apoptosis. Mol Cell 36:487–499 Gillissen B, Essmann F, Hemmati PG et al, 2007, Mcl-1 determines the Bax dependency of Nbk/Bik-induced apoptosis. J Cell Biol 179:701–715 Zhang LL, Zhang YJ, Xing D, 2010, LPLI inhibits apoptosis upstream of Bax translocation via a GSK-3β-inactivation mechanism. J Cell Physiol 224:218–228 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 809 EP 816 DI 10.1007/s12253-012-9508-x PG 8 ER PT J AU Attaallah, W Yilmaz, MA Erdogan, N Yalcin, S Aktan, A AF Attaallah, Wafi Yilmaz, Mine Ayse Erdogan, Nusret Yalcin, A. Suha Aktan, Ozdemir Ahmet TI Whey Protein Versus Whey Protein Hydrolyzate for the Protection of Azoxymethane and Dextran Sodium Sulfate Induced Colonic Tumors in Rats SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon cancer; Azoxymethane; Dextran sodium sulfate; Whey protein; Whey protein hydrolyzate ID Colon cancer; Azoxymethane; Dextran sodium sulfate; Whey protein; Whey protein hydrolyzate AB Recent studies have shown that whey protein has many useful effects including its anti-cancer effect. In this study we have compared the protective effect of dietary whey protein with whey protein hydrolyzate against azoxymethane and dextran sodium sulfate induced colon cancer in rats. We used a rat model of the colon cancer induced by administration of azoxymethane followed by repeated dextran sodium sulfate ingestion which causes multiple tumor development. Colon tissues were analyzed histologically in addition to biochemical analyses performed by measuring lipid peroxidation, protein oxidation and glutathione levels in both of colon and liver tissues of rats after sacrification. Macroscopic and microscopic tumors were identified in all groups that received azoxymethane followed by repeated dextran sodium sulfate. Group fed with whey protein hydrolyzate showed significantly less macroscopic and microscopic tumor development compared with group fed with whey protein. The protocol applied to generate an appropriate model of colon cancer was successful. Whey protein hydrolyzate was found to be more effective in preventing colon tumor development compared with whey protein. C1 [Attaallah, Wafi] Marmara University, School of Medicine, Department of General SurgeryIstanbul, Turkey. [Yilmaz, Mine Ayse] Marmara University, School of Medicine, Department of BiochemistryIstanbul, Turkey. [Erdogan, Nusret] Taksim Education and Research Hospital, Department of PathologyIstanbul, Turkey. [Yalcin, A. Suha] Marmara University, School of Medicine, Department of BiochemistryIstanbul, Turkey. [Aktan, Ozdemir Ahmet] Marmara University, School of Medicine, Department of General SurgeryIstanbul, Turkey. RP Attaallah, W (reprint author), Marmara University, School of Medicine, Department of General Surgery, Istanbul, Turkey. EM drwafi2003@yahoo.com CR Bissahoyo A, Pearsall RS, Hanlon K, Vicky A, Donna H, Virginia LG, David WT, 2005, Azoxymethane is a genetic backgrounddependent colorectal tumor initiator and promoter in mice: effects of dose, route, and diet. Toxicol Sci 88:340–345 Hakkak R, Korourian S, Ronis MJJ, Johnston JM, Badger TM, 2001, Dietary whey protein protect against azoxymethane-induced colon tumors in male rats. Cancer Epidem Biomar 10:555–558 Xiao R, Badger TM, Simmen FA, 2005, Dietary exposure to soy or whey proteins alters colonic golbal gene expression profiles during rat colon tumorigenesis. Mol Cancer 4:1 Belobrajdic DP, McIntosh GH, Owens JA, 2003, Whey proteins protect more than red meat against azoxymethane induced ACF in Wistar rats. Cancer Lett 198:43–51 Sandler RS, 1996, Epidemiology and risk factors for colorectal cancer. Gastroenterol Clin N 25:717–735 Potter JD, 1996, Nutrition and colorectal cancer. Cancer Cause Control 7:127–146 Bounous G, Papenburg R, Kongshavn PA, Gold P, Fleiszer D, 1990, Dietary whey protein inhibits the development of dimethylhydrazine induced malignancy. Tumor Biol 11:129–136 Papenburg R, Bounous G, Fleiszer D, Gold P, 1988, Dietary whey protein inhibits the development of dimethylhydrazine induced malignancy. Clin Invest Med 11:213–217 McIntosh GH, Regester GO, Le Leu RK, Royle PJ, Smithers GW, 1995, Dietary proteins protect against dimethylhydrazine-induced intestinal cancers in rats. J Nutr 125:809–816 Dryakova A, Pihlanto A, Marnila P, Ladislav C, Korhone HJT, 2010, Antioxidant properties of whey protein hydrolysates as measured by three methods. Eur Food Res Technol 230:865–874 Contreras MM, Hernandez-Ledesma B, Amigo L, Martin-Alvarez PJ, Recio I, 2011, Production of antioxidant hydrolyzates from a whey protein concentrate with thermolysin: optimization by response surface methodology. Food Sci Technol 44:9–15 Okayasu I, Hatakeyama S, Yamada M, Ohkusa T, Inagaki Y, Nakaya R, 1990, A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology 98:694–702 Okayasu I, Yamada M, Mikami T, Kano J, Ohkusa T, 2002, Dysplasia and carcinoma development in a repeated dextran sulfate sodium induced colitis model. J Gastroen Hepatol 17:1078–1083 Boivin GP, Washington K, Yang K, Ward JM, Pretlow TP, 2003, Pathology of mouse models of intestinal cancer: consensus report and recommendations. Gastroenterology 124:762–777 Okayasu I, Ohkusa T, Kajiura K, Kanno J, Sakamoto S, 1996, Promotion of colorectal neoplasia in experimental murine ulcerative colitis. Gut 39:87–92 Popivanova BK, Kitamura K, Wu Y, Kondo T, Kagaya T, Kaneko S, Oshima M, Fujii C, Mukaida N, 2008, Blocking TNF-α in mice reduces colorectal carcinogenesis associated with chronic colitis. J Clin Invest 118:560–570 Botsoglou NA, 1994, Rapid sensitive and specific TBA method for measuring lipid peroxidation in animal tissue, food and feedstuff samples. J Agr Food Chem 42:1931–1937 Levine RL, Garland D, Oliver CN et al, 1990, Determination of carbonyl content in oxidatively modified proteins. Method Enzymol 186:464–473 Browne RW, Armstrong D, 1998, Reduced glutathione and glutathione disulfide. Methods Mol Biol 108:347–352 Korhonen H, Pihlanto-Leppala A, Rantamaki P, Tupasela T, 1998, Impact of processing on bioactive proteins and peptides. Trends Food Sci Tech 9:307–319 Fearson ER, Vogalstein BA, 1990, Genetic model for colorectal tumorigenesis. Cell 61:759–767 Reddy BU, Maruyama H, 1986, Effects of dietary fish oil on azoxymethane induced colon carcinogenesis in male F344 rats. Cancer Res 46:3367–3370 Bounous G, 2000, Whey protein concentrate, WPC, and glutathione modulation in cancer treatment. Anticancer Res 20:4785–4792 Bounous G, Papenburg R, Kongshavn PA, Gold P, 1998, The immunoenhancing property of dietary whey protein concentrate. Clin Invest Med 11:271–278 Bounous G, Batist G, Gold P, 1989, Immunoenhancing property of dietary whey protein in mice: role of glutathione. Clin Invest Med 12:154–161 Lothian B, Grey V, Kimoff RJ, Lands LC, 2000, Treatment of obstructive airway disease with a cysteine donor protein supplement: a case report. Chest 117:914–916 Micke P, Beeh KM, Schlaak JF, Buhl R, 2001, Oral supplemention with whey proteins increases plasma glutathione levels of HIVinfected patients. Eur J Clin Invest 31:171–178 Micke P, Beeh KM, Buhl R, 2002, Effects of long term supplemention with whey proteins on plasma glutathione levels of HIVinfected patients. Eur J Nutr 41:12–18 Colbert LB, Decker EA, 1991, Antioxidant activity of an ultrafiltration permeate from acid whey. J Food Sci 56:1248–1250 Tong LM, Sasaki S, McClements DJ, Decker EA, 2000, Mechanisms of antioxidant activity of a high molecular weight fraction of whey. J Agric Food Chem 48:1473–1478 Rowlands JC, He L, Hakkak R, Ronis MJ, Badger TM, 2001, Soy and whey proteins downregulate DMBA-induced liver and mammary gland CYP1 expression in female rats. J Nutr 131:3281– 3287 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 817 EP 822 DI 10.1007/s12253-012-9509-9 PG 6 ER PT J AU Rubovszky, G Horvath, Zs Toth, E Lang, I Kasler, M AF Rubovszky, Gabor Horvath, Zsolt Toth, Erika Lang, Istvan Kasler, Miklos TI Significance of Histomorphology of Early Triple-Negative Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Triple-negative breast cancer; Vascular invasion; Necrosis; Irradiation ID Triple-negative breast cancer; Vascular invasion; Necrosis; Irradiation AB Triple-negative breast cancer (TNBC) is a heterogeneous disease. Possibly genetic characterisation provides the most appropriate information on tumour biology and prognosis, but it is only limitedly available in clinical practice. The aim of this investigation was to explore what additional prognostic information could be gained from detailed histomorphologic report. Patients and method: patients were selected retrospectively operated from 2005 to 2009 in one institution and charts were revised. Beyond age, tumour and nodal status, histologic grade and therapy, the additional pathologic characteristics were also involved in analysis: necrosis, lymphocytic infiltration, peritumoural vascular invasion (PVI), perineural invasion, DCIS extent and grade, perinodal spread, mitotic activity index (MAI). Results: 295 early TNBC were involved. In univariate survival analysis with a mean follow-up of 3.57 years the tumour size, the nodal status, type of operation (conservation or mastectomy), irradiation, PVI and perinodal spread proved to be significantly connected with both disease free survival (DFS) and breast cancer specific overall survival (BSOS), and necrosis and chemotherapy with BSOS. Necrosis analysed together with lymphocytic infiltrate showed greater predicting power. In multivariate analysis nodal metastasis, necrosis positive/lymphacytic infiltration negative status and lack of irradiation has significant negative impact on DFS (p0<0.0001 HR:1.98 [1.4–2.77], p0<0.017 HR:2.1 [1.1–3.8], p0<0.001 HR:0.25 [0.11–0.57], respectively) and BSOS (p0<0.0001 HR:2.47 [1.8–3.4], p0<0.017 HR:3.7 [1.6–8.2], p0<0.0017 HR:0.24 [0.1–0.58], respectively). For DFS perivascular invasion also showed significant effect (p0<0.042 HR:2.5 [1.0–6.0]). Nodal status was the strongest prognostic parameter but other histomorphologic parameters can be used for prognosis prediction. C1 [Rubovszky, Gabor] National Institute of OncologyBudapest, Hungary. [Horvath, Zsolt] National Institute of OncologyBudapest, Hungary. [Toth, Erika] National Institute of OncologyBudapest, Hungary. [Lang, Istvan] National Institute of OncologyBudapest, Hungary. [Kasler, Miklos] National Institute of OncologyBudapest, Hungary. RP Rubovszky, G (reprint author), National Institute of Oncology, Budapest, Hungary. EM garub@oncol.hu CR Rakha EA, El-Sayed ME, Green AR et al, 2007, Prognostic markers in triple-negative breast cancer. Cancer 109:25–32 Sorlie T, Perou CM, Tibshirani R et al, 2001, Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implication. Proc Natl Acad Sci U S A 98:10869–10874 Kreike B, van Kouwenhove M, van de Vijver MJ et al, 2007, Gene expression profiling and histopathological characterization of triple-negative /basal-like breast carcinomas. Breast Cancer Res 9:R65 Lehmann BD, Bauer JA, Chen X, Sanders ME, Bapsi Chakravarthy A, Shyr Yu, Pietenpol JA, 2011, Identification of human triplenegative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 121:2750–2767 Teschendorff AE, Caldas C, 2008, A robust classifier of high predictive value to identify good prognosis patients in ERnegative breast cancer. Breast Cancer Res 10:R73 Yerushalmi R, Hayes MM, Gelmon KA, 2009, Breast carcinoma– rare types: review of the literature. Ann Oncol 20:1763–1770 Mohammed RA, Ellis IO, Lee AH, Martin SG, 2009, Vascular invasion in breast cancer; an overview of recent prognostic developments and molecular pathophysiological mechanisms. Histopathology 55:1–9 VialeG,Giobbie-HurderA,Gusterson BA,Maiorano E,Mastropasqua MG, Sonzogni A, Mallon E, Colleoni M, Castiglione-Gertsch M, Regan MM, Price KN, Brown RW, Golouh R, Crivellari D, Karlsson P, Ohlschlegel C, Gelber RD, Goldhirsch A, Coates AS, 2010, Adverse prognostic value of peritumoral vascular invasion: is it abrogated by adequate endocrine adjuvant therapy? Results from two International Breast Cancer Study Group randomized trials of chemoendocrine adjuvant therapy for early breast cancer. Ann Oncol 21:245–254 Pinder SE, Ellis IO, Galea M, O’Rouke S, Blamey RW, Elston CW, 1994, Pathological prognostic factors in breast cancer. III. Vascular invasion: relationship with recurrence and survival in a large study with long-term follow-up. Histopathology 24:41–47 Sabatier R, Jacquemier J, Bertucci F, Esterni B, Finetti P, Azario F, Birnbaum D, Viens P, Goncalves A, Extra JM, 2011, Peritumoural vascular invasion: a major determinant of triple-negative breast cancer outcome. Eur J Cancer 47:1537–1545 Lende TH, Janssen EA, Gudlaugsson E, Voorhorst F, Smaaland R, van Diest P, Soiland H, Baak JP, 2011, In patients younger than age 55 years with lymph node-negative breast cancer, proliferation by mitotic activity index is prognostically superior to adjuvant! J Clin Oncol 29:852–858 GaleaMH, Blamey RW, Elston CE, Ellis IO, 1992, The Nottingham Prognostic Index in primary breast cancer. Breast Cancer Res Treat 22:207–219 Choi YL, Oh E, Park S, Kim Y, Park YH, Song K, Cho EY, Hong YC, Choi JS, Lee JE, Kim JH, Nam SJ, Im YH, Yang JH, Shin YK, 2010, Triple-negative, basal-like, and quintuple-negative breast cancers: better prediction model for survival. BMC Cancer 10:507 Schiffman SC, McMasters KM, Scoggins CR, Martin RC, Chagpar AB, 2011, Lymph node ratio: a proposed refinement of current axillary staging in breast cancer patients. J Am Coll Surg 213:45–52 Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, McShane LM, Paik S, Pegram MD, Perez EA, Press MF, Rhodes A, Sturgeon C, Taube SE, Tubbs R, Vance GH, van de Vijver M, Wheeler TM, Hayes DF, 2007, American Society of Clinical Oncology; College of American Pathologists. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol 25:118–145 Bidard FC, Conforti R, Boulet T, Michiels S, Delaloge S, Andre F, 2007, Does triple-negative phenotype accurately identify basallike tumour? An immunohistochemical analysis based on 143 ‘triple-negative’ breast cancers. Ann Oncol 18:1285–1286 Lin C, Chien SY, Chen LS, Kuo SJ, Chang TW, Chen DR, 2009, Triple negative breast carcinoma is a prognostic factor in Taiwanese women. BMC Cancer 9:192 Liu ZB, Liu GY, Yang WT et al, 2008, Triple-negative breast cancer types exhibit a distinct poor clinical characteristic in lymph node-negative Chinese patients. Oncol Rep 20:987–994 Bauer KR, Brown M, Cress RD et al, 2007, Descripitive analysis of estrogen receptor, ER)-negative, progesteron receptor, PR)- negative, and HER2-negative invasive breast cancer, the socalled triple-negative phenotype: a population based study from the California Cancer Registry. Cancer 109:1721–1728 Onitilio AA, Engel JM, Greenlee RT et al, 2009, Breast cancer subtypes based on ER/PR and Her2 expression: comparison of clinicopathologic features and survival. Clin Med Res 7:4–13 Carey LA, Perou CM, Livasy CA et al, 2008, Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA 295:2492–2502 Spitale A, Mazzola P, Soldini D et al, 2009, Breast cancer classification according to immunohistochemical markers: clinicipathologic features and short-term survival analysis in a population-based study from the South of Switzerland. Ann Oncol 20:628–635 Nofech-Mozes S, Trudeau M, Kahn HK, Dent R, Rawlinson E, Sun P, Narod SA, Hanna WM, 2009, Patterns of recurrence in the basal and non-basal subtypes of triple-negative breast cancers. Breast Cancer Res Treat 118:131–137 Mohammed RA, Ellis IO, Mahmmod AM, Hawkes EC, Green AR, Rakha EA, Martin SG, 2011, Lymphatic and blood vessels in basal and triple-negative breast cancers: characteristics and prognostic significance. Mod Pathol 24:774–785 Linderholm BK, Hellborg H, Johansson U, Elmberger G, Skoog L, Lehtio J, Lewensohn R, 2009, Significantly higher levels of vascular endothelial growth factor, VEGF, and shorter survival times for patients with primary operable triple-negative breast cancer. Ann Oncol 20:1639–1646 Liu HT, Ma R, Yang QF, Du G, Zhang CJ, 2009, Lymphangiogenic characteristics of triple negativity in node-negative breast cancer. Int J Surg Pathol 17:426–431 Colleoni M, Rotmensz N, Maisonneuve P, Sonzogni A, Pruneri G, Casadio C, Luini A, Veronesi P, Intra M, Galimberti V, Torrisi R, Andrighetto S, Ghisini R, Goldhirsch A, Viale G, 2007, Prognostic role of the extent of peritumoral vascular invasion in operable breast cancer. Ann Oncol 18:1632–1640 Oven Ustaalioglu BB, Bilici A, Kefeli U, Seker M, Yildirim E, Salepci T, Oncel M, Kement M, Gumus M, 2010, Does the metastatic lymph node ratio influence the disease-free survival of patients with breast cancer: single-center experiences. Oncology 79:105–111 Mersin H, Yildirim E, Berberoglu U, Gulben K, 2009, Triple negative phenotype and N-ratio are important for prognosis in patients with stage IIIB non-inflammatory breast carcinoma. J Surg Oncol 100:681–687 Viale G, Regan MM, Maiorano E, Mastropasqua MG, Dell’Orto P, Rasmussen BB, Raffoul J, Neven P, Orosz Z, Braye S, Ohlschlegel C, Thurlimann B, Gelber RD, Castiglione-Gertsch M, Price KN, Goldhirsch A, Gusterson BA, Coates AS, 2007, Prognostic and predictive value of centrally reviewed expression of estrogen and progesterone receptors in a randomized trial comparing letrozole and tamoxifen adjuvant therapy for postmenopausal early breast cancer: BIG 1–98. J Clin Oncol 25:3846–3852 Stuart-Harris R, Caldas C, Pinder SE, Pharoah P, 2008, Proliferation markers and survival in early breast cancer: a systematic review and meta-analysis of 85 studies in 32,825 patients. Breast 17:323–334 Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, Fitzgibbons PL, Francis G, Goldstein NS, Hayes M, Hicks DG, Lester S, Love R, Mangu PB, McShane L, Miller K, Osborne CK, Paik S, Perlmutter J, Rhodes A, Sasano H, Schwartz JN, Sweep FC, Taube S, Torlakovic EE, Valenstein P, Viale G, Visscher D, Wheeler T, Williams RB, Wittliff JL, Wolff AC, 2010, American Society of Clinical Oncology/College Of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol 28:2784–2795 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 823 EP 831 DI 10.1007/s12253-012-9510-3 PG 9 ER PT J AU Yuan, J Tu, Y Mao, X He, Sh Wang, L Fu, G Zong, J Zhang, Y AF Yuan, Jun Tu, Yanyang Mao, Xinggang He, Shiming Wang, Liang Fu, Guoqiang Zong, Jianhai Zhang, Yongsheng TI Increased Expression of FAT10 is Correlated with Progression and Prognosis of Human Glioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; FAT10; Immunochemistry assay; Quantitative real-time PCR; Western blot analysis; Prognosis ID Glioma; FAT10; Immunochemistry assay; Quantitative real-time PCR; Western blot analysis; Prognosis AB FAT10, as a small ubiquitin-like modifier, plays an important role in various cellular processes, including mitosis, immune response, and apoptosis, the dys-regulation of which may arise tumorigenesis. Therefore, the aim of this study was to examine the expression of FAT10 at gene and protein levels in glioma samples with different WHO grades and its association with survival. One hundred and twentyeight glioma specimens and 10 non-neoplastic brain tissues were collected. Immunohistochemistry assay, quantitative real-time PCR and Western blot analysis were carried out to investigate the expression of FAT10. Kaplan-Meier method and Cox’s proportional hazards model were used in survival analysis. Immunohistochemistry showed that FAT10 protein was over-expressed in glioma tissues. FAT10 mRNA and protein levels were both higher in glioma compared to control on real-time PCR and Western blot analysis (both P<0.001). Additionally, its expression levels increase from grade I to grade IV glioma according to the results of real-time PCR, immunohistochemistry analysis and Western blot. Moreover, the survival rate of FAT10-positive patients was significantly lower than that of FAT10-negative patients (P<0.001). We further confirmed that the increased expression of FAT10 was a significant and independent prognostic indicator in glioma by multivariate analysis (P<0.001). Our data provides convincing evidence for the first time that the increased expression of FAT10 at gene and protein levels is correlated with poor outcome in patients with glioma. FAT10 may promote the aggressiveness of glioma and may be a potential prognosis predictor of glioma. C1 [Yuan, Jun] Fourth Military Medical University, Tangdu Hospital, Administrative Department, 710038 Xi’an, China. [Tu, Yanyang] Fourth Military Medical University, Tangdu Hospital, Department of Emergency, 710038 Xi’an, China. [Mao, Xinggang] 254th Hospital of PLA, Department of Neurosurgery, 300142 Tianjin, China. [He, Shiming] Fourth Military Medical University, Tangdu Hospital, Department of Neurosurgery, 710038 Xi’an, China. [Wang, Liang] Fourth Military Medical University, Tangdu Hospital, Department of Neurosurgery, 710038 Xi’an, China. [Fu, Guoqiang] Fourth Military Medical University, Tangdu Hospital, Department of Emergency, 710038 Xi’an, China. [Zong, Jianhai] Fourth Military Medical University, Tangdu Hospital, Department of Neurosurgery, 710038 Xi’an, China. [Zhang, Yongsheng] Fourth Military Medical University, Tangdu Hospital, Administrative Department, 710038 Xi’an, China. RP Zhang, Y (reprint author), Fourth Military Medical University, Tangdu Hospital, Administrative Department, 710038 Xi’an, China. EM zhangys@fmmu.edu.cn CR Ohgaki H, Kleihues P, 2005, Epidemiology and etiology of gliomas. Acta Neuropathol 109:93 Louis DN, Ohgaki H, Wiestler OD, 2007, The 2007 WHO classifcation of tumors of the central nervous system. Acta Neuropathol 114:97–109 Mu YG, Chen MZ, Chen ZP, Zhou WN, Zhang XH, Sai K, 2004, Microsurgical technique of brain glioma—a report of 183 cases. Ai Zheng 23:1317–1321 Chang CY, Li MC, Liao SL, Huang YL, Shen CC, Pan HC, 2005, Prognostic and clinical implication of IL-6 expression in glioblastoma multiforme. J Clin Neurosci 12:930–933 Lin H, Wang Y, Zhang X, Liu B, Zhang W, Cheng J, 2011, Prognostic significance of kappaB-Ras1 expression in gliomas. Med Oncol In press Jentsch S, Pyrowolakis G, 2000, Ubiquitin and its kin: how close are the family ties? Trends Cell Biol 10:335–342 Li T, Santockyte R, Yu S, Shen RF, Tekle E, Lee CG, Yang DC, Chock PB, 2011, FAT10 modifies p53 and upregulates its transcriptional activity. Arch Biochem Biophys 509:164–169 Canaan A, Yu X, Booth CJ, Lian J, Lazar I, Gamfi SL, Castille K, Kohya N, Nakayama Y, Liu YC, Eynon E, Flavell R, Weissman SM, 2006, FAT10/diubiquitin-like protein-deficient mice exhibit minimal phenotypic differences. Mol Cell Biol 26:5180–5189 Hipp MS, Kalveram B, Raasi S, Groettrup M, Schmidtke G, 2005, FAT10, a ubiquitin-independent signal for proteasomal degradation. Mol Cell Biol 25:3483–3491 Raasi S, Schmidtke G, Groettrup M, 2001, The ubiquitin-like protein FAT10 forms covalent conjugates and induces apoptosis. J Biol Chem 276:35334–35343 Ji F, Jin X, Jiao CH, Xu QW, Wang ZW, Chen YL, 2009, FAT10 level in human gastric cancer and its relation with mutant p53 level, lymph node metastasis and TNM staging. World J Gastroenterol 15:2228–2233 Lukasiak S, Schiller C, Oehlschlaeger P, Schmidtke G, Krause P, Legler DF, Autschbach F, Schirmacher P, Breuhahn K, Groettrup M, 2008, Proinflammatory cytokines cause FAT10 upregulation in cancers of liver and colon. Oncogene 27:6068–6074 Qing X, French BA, Oliva J, French SW, 2011, Increased expression of FAT10 in colon benign, premalignant and malignant epithelial neoplasms. Exp Mol Pathol 90:51–54 Lee CG, Ren J, Cheong IS, Ban KH, Ooi LL, Yong Tan S, Kan A, Nuchprayoon I, Jin R, Lee KH, Choti M, Lee LA, 2003, Expression of the FAT10 gene is highly upregulated in hepatocellular carcinoma and other gastrointestinal and gynecological cancers. Oncogene 22:2592–2603 Brown RS, Wahl RL, 1993, Overexpression of Glut-1 glucose transporter in human breast cancer: an immunohistochemical study. Cancer 72:2979–2985 Ren J, Wang Y, Gao Y, Mehta SB, Lee CG, 2011, FAT10 mediates the effect of TNF-α in inducing chromosomal instability. J Cell Sci 124:3665–3675 Zhang DW, Jeang KT, Lee CG, 2006, p53 negatively regulates the expression of FAT10, a gene upregulated in various cancers. Oncogene 25:2318–2327 Ren J, Kan A, Leong SH, Ooi LL, Jeang KT, Chong SS, Kon OL, Lee CG, 2006, FAT10 plays a role in the regulation of chromosomal stability. J Biol Chem 281:11413–11421 Liu YC, Pan J, Zhang C, Fan W, Collinge M, Bender JR, Weissman SM, 1999, A MHC-encoded ubiquitin-like protein, FAT10, binds noncovalently to the spindle assembly checkpoint protein MAD2. Proc Natl Acad Sci U S A 96:4313–4318 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 833 EP 839 DI 10.1007/s12253-012-9511-2 PG 7 ER PT J AU Hewala, IT Abd El-Monaim, AN Anwar, M Ebied, AS AF Hewala, I Taha Abd El-Monaim, A Nadia Anwar, Medhat Ebied, A Samia TI The Clinical Significance of Serum Soluble Fas and p53 Protein in Breast Cancer Patients: Comparison with Serum CA 15-3 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Apoptosis; Soluble Fas; p53 protein; CA 15-3; Diagnosis; Surgery; FAC chemotherapy ID Breast cancer; Apoptosis; Soluble Fas; p53 protein; CA 15-3; Diagnosis; Surgery; FAC chemotherapy AB Serum sFas and p53 protein have been observed in breast cancer patients, but their clinical usefulness for diagnosis and therapy monitoring has not been clarified. The aim of this study was to compare the clinical utility of serumsFas and p53 protein with that of serumCA 15-3 as the most commonly used breast cancer tumor marker. Serum samples were taken from 35 normal healthy controls and 35 breast cancer patients before surgery, after 2 weeks of surgery and after six cycles of FAC chemotherapy. Serum sFas and p53 protein levels were measured using ELISA kits. Serum CA 15-3 levels were determined using IRMA kit. Mean Serum levels of sFas and CA 15-3 were significantly elevated while p53 protein was significantly declined in breast caner patients than controls. Serum p53 protein showed the greatest significant area under the ROC curve (84.3%) followed by sFas (80.5%), then CA 15-3 (78%). The sensitivity, specificity and cut-off value for diagnosing breast cancer patients were 84.2%, 82.6% and 2.88 U/ml for p53 protein, 83.3%, 68.2% and 497.3 pg/ml for sFas and 45.8%, 100% and 23 U/ml for CA15-3. Surgical removal of breast resulted in a significant decline in serum sFas level with no effect on serum p53 protein and CA 15-3 levels. Six cycles of chemotherapy resulted in a significant elevation in serum sFas level with no effect on serum p53 protein and CA 15-3 levels. sFas was significantly correlated with tumor grade. It could be concluded that although serum p53 protein is superior to sFas and CA15-3 for diagnosis of breast cancer patients, only sFas is useful for monitoring the response of breast cancer patients to surgery and chemotherapy if the effect of systemic inflammatory reactions is excluded. C1 [Hewala, I Taha] Alexandria University, Medical Research Institute, Department of Radiation Sciences, 165 El-Horria Avenue, El Hadara, 21561 Alexandria, Egypt. [Abd El-Monaim, A Nadia] Alexandria University, Medical Research Institute, Department of Cancer Management and Research, 165 El-Horria Avenue, El Hadara, 21561 Alexandria, Egypt. [Anwar, Medhat] Alexandria University, Medical Research Institute, Department of Experimental and Clinical Surgery, 165 El-Horria Avenue, El Hadara, 21561 Alexandria, Egypt. [Ebied, A Samia] Alexandria University, Medical Research Institute, Department of Applied Medical Chemistry, 165 El-Horria Avenue, El Hadara, 21561 Alexandria, Egypt. RP Hewala, IT (reprint author), Alexandria University, Medical Research Institute, Department of Radiation Sciences, 21561 Alexandria, Egypt. EM tahahewala@hotmail.com CR Tamakoshi 1A, Nakachi K, Ito Y, Lin Y, Yagyu K, Kikuchi 1S, et al, 2008, Soluble Fas level and cancer mortality: Findings from a nested case–control study within a large-scale prospective study. Int. J. Cancer 123: 1913–1916. Sheen-Chen SM, Chen HS, Eng HL, Chenw J, 2003, Circulating soluble Fas in patients with breast cancer. World J Surg 27:10–13 Jager J, Jansen R, Arends J, 2002, Clinical relevance of apoptotic markers in breast cancer not yet clear. Apoptosis 7:361–365 Evan G, Littlewood T, 1998, A matter of life and cell death. Science 94:695–698 Kolomecki K, Maciaszczyk P, Stepien T, Kuzdak K, Ulanska J, 2005, p53 concentration and soluble FasL(sFasL, serum level as indicators of apoptosis in patients with benign and malignant thyroid tumors. Bratisl Lek Listy 106:297–300 Bennett M, Macdonald K, Chen SW, Luzio JP, Simari R, Weissberg P, 1998, Cell surface trafficking of Fas: a rapid mechanism of p53- mediated apoptosis. Science 282:290–293 Balogh GA, Mailo DA, Corte MM, Roncoroni P, Nardi H, Vincen E et al, 2006)Mutant p53 protein in serumcould be used as amolecular marker in human breast cancer. Int J Oncol 28:995–1002 Duffy MJ, 1999, CA 15-3 and related mucins as circulating markers in breast cancer. Ann Clin Biochem 36:579–586 Keyhani M, Nasizadeh S, Dehghannejad A, 2005, Serum CA 15-3 measurment in breast cancer patients before and after mastectomy. Arch Iranian Med 8:263–266 Haskell CM, Lowitz BB, Casciato AD, 1985, Breast cancer. In: Casciato AD, Lowitz BB, eds, Manual of clinical oncology, 2nd edn. Little and Brown, Boston, pp 150–165 Rintoul RF, 1986, Operations on the breast. In: Farquhaerison,s text book of operative surgery, 7th ed. Churchill living stone, pub., 270-281 AbeloffMD, Lichter AS, Niederhuber JE, Pierce LJ, Aziz DC, 1995, Breast. In: Abeloff MD, Armitage JO, Licher AS, Niederhuber JE, eds, Clinical Oncology, Churchil livingstone Inc., Pub., 1617– 714 Munakata S, Watanabe O, Ohashi K, Morino H, 2005, Expression of Fas ligand and bcl-2 in cervical carcinoma and their prognostic significance. Am J Clin pathol 123:879–885 Zhang L, Levi E, Majumder P, Yu Y, Aboukameel A, Du J, 2007, Transactivator of transcription tagged cell cycle and apoptosis regulatory protein-1 peptides suppress the growth of human breast cancer cells in vitro and in vivo. Mol cancer Ther 6:1661–1672 Motyl T, Gajkowska B, Zarzy J, Gajewska M, Lamparska P, 2006, Apoptosis and autophagy in mammary gland remodeling and breast cancer chemotherapy. J Physiol Pharmacol 7:17–32 Habibagahi M, Jaberipour M, Fattahi MJ, Hashemi SB, Shariati M, 2010, High concentration of serum soluble Fas in patients with head and neck carcinoma: a comparative study before and after surgical removal of tumor. Middle East J Can 1:21–26 El-Sarha A, Magour G, Zaki S, El-Sammak M, 2009, Serum sFas and tumor tissue FasL negatively correlated with survival in Egyptian patients suffering from breast ductal carcinoma. Pathol Oncol Res 15:241–250 Ohbu M, Saegusa M, Okayasu I, 1995, Apoptosis and cellular proliferation in oesophageal squamous cell carcinoma: differences between keratinizing and nonkeratinizing types. Virchows arch 427:271–276 Pignataro L, Arisi E, Sambataro G, Corsi MM, 2003, Soluble Fas, sFas, and soluble Fas ligand, sFas-L, balance in laryngeal carcinoma before and after surgical treatment. J Sur Oncol 83:112– 115 Ugurel S, Rappl G, TilgenW, Reinhold U, 2001, Increased soluble CD95(sFas/CD95, serum level correlates with poor prognosis in melanoma patients. Clin Cancer Res 7:1282–1286 Shimizu M, Kondo M, Ito Y, Kume H, Suzuki R, Yamaki K, 2005, Soluble Fas and Fas ligand provide new information on metastasis and response to chemotherapy in SCLC patients. Canc Detect Prev 29:175–180 Iwase M, Kondo G, Watanabe H, Takaoka S, Uchida M, Ohashi M et al, 2006, Regulation of Fas-mediated apoptosis in neutrophils after surgery-induced acute inflammation. J Surg Res 134:114–123 Paunel-Gorgulu A, Flohe S, Scholz M, Windolf J, Logters T, 2011, Increased serum soluble Fas after major trauma is associated with delayed neutrophil apoptosis and development of sepsis. Critical care 15:R20 Donnelly SC, MacGregor I, Zamani A, Gordon MW, Robertson CE, Steedman DJ et al, 1995, Plasma elastase levels and the development of the adult respiratory distress syndrome. Am J Respir Crit Care Med 151:1428–1433 Bhatia R, Dent C, Topley N, Pallister I, 2006, Neutrophil priming for elastase release in adult blunt trauma patients. J Trauma 60:590–596 Nadal C, Maurel J, Gallego R, Castells A, Longaron R, Marmol M et al, 2005, Fas/Fas ligand ratio: a marker of Oxaliplatin-based intrinsic and acquired resistance in advanced colorectal cancer. Clin Cancer Res 11:4770–4774 Hewala TI, Abd EL-Moneim NA, Ebied SA, Sheta MI, Soliman K, Abu-ELenean A, 2010, Diagnostic and prognostic value of serum nitric oxide, tumor necrosis factor-a, basic fibroblast growth factor and copper as angiogenic markers in premenopausal breast cancer patients: a case-control study. BJBS 67:167–176 Gion M, Mione R, Leon AE, Dittadi R, 1999, Comparison of the diagnostic accuracy of CA27.29 and CA15-3 in primary breast cancer. Clin Chem 45:630–637 Duffy MJ, 2006, Serum tumor markers in breast cancer: are they of clinical value? Clin Chem 52:345–351 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 841 EP 848 DI 10.1007/s12253-012-9512-1 PG 8 ER PT J AU Zamirska, A Matusiak, Dziegiel, P Szybejko-Machaj, G Szepietowski, CJ AF Zamirska, Aleksandra Matusiak, Lukasz Dziegiel, Piotr Szybejko-Machaj, Grazyna Szepietowski, C Jacek TI Expression of Metallothioneins in Cutaneous Squamous Cell Carcinoma and Actinic Keratosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Metallothioneins; Squamous cell carcinoma; Actinic keratosis; Ki-67; Skin ID Metallothioneins; Squamous cell carcinoma; Actinic keratosis; Ki-67; Skin AB Metallothioneins (MT) are low-molecular weight proteins implicated in heavy metal detoxification, zinc and cooper homeostasis and cell protection against free radicals. In variety of cancers MT-overexpression was shown, but there are just a few studies on the role of MT in skin carcinogenesis. Current study was undertaken to evaluate MT and Ki-67 expression in pre-cancerous skin lesions as well as in fully developed skin cancers. 73 squamous cell carcinomas (SCC), 23 actinic keratoses (AK) and 20 normal skin samples were included in the study. In obtained paraffin sections immunohistochemical reactions were performed. MT-expression in SCC (mean 2.89±1.83) was significantly higher than in AK (mean 1.69±1.26)(p00.006) and higher than in normal skin (mean 2±0.79) (p00.0075). The MT-expression positively correlated with Ki-67 expression (R00.28; p00.017) in SCC and in AK (R00.49; p00.018). Various clinico-pathological variables, e.g. morphology, size of lesions and the depth of neoplastic infiltration were not associated to MT-expression in both SCC and AK. The grade of histological differentiation of SCC correlated positively with Ki-67 antigen (p<0.001) and did not correlate with MT-expression (p00.06). Ki-67 expression was higher in SCC and in AK than in healthy skin (p00,003). In SCC and in AK expression of Ki-67 antigen correlated positively with MT-expression (respectively p00.017 and p00.018). MT may serve as a good markers of proliferation in SCC and AK. MT-overexpression in SCC may suggest a potential role of MT in skin carcinogenesis. C1 [Zamirska, Aleksandra] Wroclaw Medical University, Department of Dermatology, Venereology and Allergology, Chalubinskiego 1, 50-368 Wroclaw, Poland. [Matusiak, Lukasz] Wroclaw Medical University, Department of Dermatology, Venereology and Allergology, Chalubinskiego 1, 50-368 Wroclaw, Poland. [Dziegiel, Piotr] Wroclaw Medical University, Department of Human Morphology and EmbryologyWroclaw, Poland. [Szybejko-Machaj, Grazyna] Wroclaw Medical University, Department of Dermatology, Venereology and Allergology, Chalubinskiego 1, 50-368 Wroclaw, Poland. [Szepietowski, C Jacek] Wroclaw Medical University, Department of Dermatology, Venereology and Allergology, Chalubinskiego 1, 50-368 Wroclaw, Poland. RP Matusiak, (reprint author), Wroclaw Medical University, Department of Dermatology, Venereology and Allergology, 50-368 Wroclaw, Poland. EM luke71@interia.pl CR Dziegiel P, 2004, Expression of metallothioneins in tumor cells. Pol J Pathol 55:3–12 Pedersen MO, Larsen A, Stoltenberg M et al, 2009, The role of metallothionein in oncogenesis and cancer prognosis. Prog Histochem Cytochem 44:29–64 Coyle P, Philcox JC, Carey LC et al, 2002, Metallothionein: the multipurpose protein. Cell Mol Life Sci 59:627–47 Palmiter RD, Findley SD, Whitmore TE et al, 1992, MT-III, a brain-specific member of the metallothionein gene family. Proc Natl Acad Sci USA 89:6333–7 Quaife CJ, Findley SD, Erickson JC et al, 1994, Induction of a new metallothionein isoform, MT-IV, occurs during differentiation of stratified squamous epithelia. Biochemistry 33:7250–9 Miles AT, Hawksworth GM, Beattie JH et al, 2000, Induction, regulation, degradation and biological significance of mammalian metallothioneins. Crit Rev Biochem Mol Biol 35:35–70 Dziegiel P, Jelen M, Muszczynska B et al, 2004, Role of metallothionein expression in non-small cell lung carcinomas. Rocz Akad Med Bialymst 49:43–45 Sato M, Kondoh M, 2002, Recent studies on metallothionein: protection against toxicity of heavy metals and oxygen free radicals. Tohoku J Exp Med 196:9–22 Dutsch-Wicherek M, Sikora J, Tomaszewska R, 2008, The possible biological role of metallothionein in apoptosis. Front Biosci 13:4029–38 Miyashita H, Sato Y, 2005, Metallothionein 1 is a downstream target of vascular endothelial zinc finger 1, VEZF1, in endothelial cells and participates in the regulation of angiogenesis. Endothelium 12:163–170 Nielsen AE, Bohr A, Penkowa M, 2007, The balance between life and death of cells: roles of metallothioneins. Biomarker Insights 2:99–111 Brown JJ, Xu H, William-Smith L et al, 2003, Evaluation of metallothionein and p53 expression as potential prognostic markers for laryngeal squamous cell carcinoma. Cell Mol Biol. 49 Online Pub:OL473-9 Cardoso SV, Silveira-Junior JB, De Carvalho Machado V et al, 2009, Expression of metallothionein and p53 antigens are correlated in oral squamous cell carcinoma. Anticancer Res 29:1189–93 Hishikawa Y, Koji T, Dhar DK et al, 1999, Metallothionein expression correlates with metastatic and proliferative potential in squamous cell carcinoma of the oesophagus. Br J Cancer 81, 4):712–20 Jin R, Bay BH, Chow VT et al, 2001, Metallothionein 1 F mRNA expression correlates with histological grade in breast carcinoma. Breast Cancer Res Treat 66(3):265–72 Mitropoulos D, Kyroudi-Voulgari A, Theocharis S et al, 2005, Prognostic significance of metallothionein expression in renal cell carcinoma. World J Surg Oncol 3:5 Ohshio G, Imamura T, Okada N et al, 1996, Immunohistpochemical study of metallothionein in pancreatic carcinomas. J Cancer Res Clin Oncol 122:351–5 Saga Y, Hashimoto H, Yachiku S et al, 2002, Immunohistochemical expression of metallothionein in human bladder cancer: correlation with histopathological parameters and patient survival. J Urol 168:2227–2231 Sens MA, Somji S, Garrett SH et al, 2001, Metallothionein isoform 3 overexpression is associated with breast cancers having a poor prognosis. Am JPathol 159:21–26 Szelachowska J, Dziegiel P, Jelen-Krzeszewska J et al, 2008, Prognostic significance of nuclear and cytoplasmic expression of metallothioneins as related to proliferative activity in squamous cell carcinoma. Histol Histopathol 23(7):843–851 Weinlich G, Eisendle K, Hassler E et al, 2006, Metallothionein overexpression as a highly significant prognostic factor in melanoma: a prospective study on 1270 patients. Br J Cancer 94:835–41 Kelley SL, Basu A, Teicher BA et al, 1988, Overexpression of metallothionein confers resistance to anticancer drugs. Science 241:1813–15 Cohn BA, 2000, From sunlight to actinic keratosis to squamous cell carcinoma. J Am Acad Dermatol 42:143–4 Ouhtit A, Ananthaswamy HN, 2001, A model for UV- induction of skin cancer. J Biomed Biotechnol 1:5–6 Nouri K, 2007, Skin cancer. Mc Graw Hill Professional Ackerman AB, 2003, Solar keratosis is squamous cell carcinoma. Arch Dermatol 139:1216–17 Anwar J, Wrone DA, Kimyai-Asadi A et al, 2004, The development of actinic keratosis into invasive squamous cell carcinoma: evidence and evolving classification schemes. Clin Dermatol 22:189–96 Cockerell CJ, Wharton JR, 2005, New histopathological classification of actinic keratosis, incipient intraepidermal squamous cell carcinoma). J Drugs Dermatol 4:462–7 Broders AC, 1926, Carcinoma: grading and practical application. Arch Pathol 2:376–80 Weedon D, Strutton G, 2002, Skin pathology, 2nd edn. Churchill Livingston, London RemmeleW, StegnerHE, 1987, Vorschlag zur einheitlichen definition eines immunoreaktiven score, IRS, fur den immunohistochemichen Ostrogerezeptor-Nachweis, ER-ICA, im Mammakarzinomgewebe. Pathologie 8:138–40 Karasawa M, Nishimura N, Nishimura H et al, 1991, Localization of metallothionein in hair follicles of normal skin and the basal cell layer of hyperplastic epidermis: possible association with cell proliferation. J Invest Dermatol 97:97–100 Van den Oord JJ, De LeyM(1994, Distribution ofmetallothionein in normal and pathological human skin. Arch Dermatol Res 286:62–8 Borges Junior PC, Ribeiro RI, Cardoso SV et al, 2007, Metallothionein immunolocalization in actinic skin nonmelanoma carcinomas. Appl Immunohistochem Mol Morphol 15:165–9 Sundelin K, Jadner M, Norberg-Spaak L et al, 1997, Metallothionein and Fas, CD95, are expressed in squamous cell carcinoma of the tongue. Eur J Cancer 33:1860–4 Dutsch-Wicherek M, Popiela TJ, Klimek M et al, 2005, Matallothionein stroma reaction in tumor adjacent healthy tissue in head and neck squamous cell carcinoma and brest adenocarcionoma. Neuro Endocrinol Lett 26:567–74 Muramatsu Y, Hasegawa Y, Fukano H et al, 2000, Metallothionein immunoreactivity in head and neck carcinomas; special reference to clinical behaviours and chemiotherapy responses. Anticancer Res 20:257–64 Goldmann T, Moorkamp A,Wiedorn KH et al, 2001, The prognostic value of the expression of collagenase IV, cathepsin D and metallothionein in squamous cell carcinomas of the skin determined by immunohistochemistry. Arch Dermatol Res 293:115–120 Veness MJ, 2007, High-risk cutaneous squamous cell carcinoma of the head and neck. J Biomed Biotechnol 2007(3):80572, Epub 2007 Apr 3 Son KD, Kim TJ, Lee YS et al, 2008, Comparative analysis of immunohistochemical markers with invasiveness and histologic differentiation in squamous cell carcinoma and basal cell carcinoma of the skin. J Surg Oncol 97:615–620 Dziegiel P, Forgacz J, Suder E et al, 2003, Prognostic significance of metallothionein expression in correlation with Ki-67 expression in adenocarcinomas of large intestine. Histol Histopathol 18:401–7 Jayasurya A, Bay BH, Yap WM et al, 2000, Proliferative potential in nasopharyngeal carcinoma: correlations with metallothionein expression and tissue zinc levels. Carcinogenesis 21:1809–12 Pastuszewski W, Dziegiel P, Krecicki T et al, 2007, Prognostic significance of metallothionein, p53 protein and Ki-67 antigen expression in laryngeal cancer. Anticancer Res 27:335–42 Rossen K, Haerslev T, Hou-Jensen K et al, 1997, Metallothionein expression in basaloid proliferations overlying dermatofibromas and basal cell carcinomas. Br J Dermatol 136:30–4 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 849 EP 855 DI 10.1007/s12253-012-9513-0 PG 7 ER PT J AU Kenessey, I Banki, B Mark, Varga, N Tovari, J Ladanyi, A Raso, E Timar, J AF Kenessey, Istvan Banki, Balazs Mark, Agnes Varga, Norbert Tovari, Jozsef Ladanyi, Andrea Raso, Erzsebet Timar, Jozsef TI Revisiting CB1 Receptor as Drug Target in Human Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CB1 receptor; Endocannabinoid; Human malignant melanoma; Metastasis ID CB1 receptor; Endocannabinoid; Human malignant melanoma; Metastasis AB Previous studies have indicated the antitumoral effect of human melanocytes, human melanoma cell lines expressing CB1 receptor (CB1), and of the peritumoral administration of endocannabinoids. In the present study, we systematically screened several human melanoma cell lines for the expression of CNR1 and demonstrated transcription of the authentic gene. The product of CNR1, the CB1 protein, was found localized to the cell membrane as well as to the cytoskeleton. Further, the studied human melanoma cell lines expressed functional CB1 since physiological and synthetic ligands, anandamide (AEA), Met- F-AEA, ACEA and AM251 showed a wide range of biological effects in vitro, for example anti-proliferative, proapoptotic and anti-migratory. More importantly, our studies revealed that systemic administration of a stable CB1 agonist, ACEA, into SCID mice specifically inhibited liver colonization of human melanoma cells. Since therapeutic options for melanoma patients are still very limited, the endocannabinoid-CB1 receptor system may offer a novel target. C1 [Kenessey, Istvan] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Banki, Balazs] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Mark, Agnes] National Institute of OncologyBudapest, Hungary. [Varga, Norbert] National Institute of OncologyBudapest, Hungary. [Tovari, Jozsef] National Institute of OncologyBudapest, Hungary. [Ladanyi, Andrea] National Institute of OncologyBudapest, Hungary. [Raso, Erzsebet] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. RP Kenessey, I (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM steveken12@yahoo.com CR Wang J, Ueda N, 2009, Biology of endocannabinoid synthesis system. Prostaglandins Other Lipid Mediat 89:112–119 Flygare J, Sander B, 2008, The endocannabinoid system in cancerpotential therapeutic target? Semin Cancer Biol 18:176–189 Mouslech Z, Valla V, 2009, Endocannabinoid system: an overview of its potential in current medical practice. Neuro Endocrinol Lett 30:153–179 Bifulco M, Di Marzo V, 2002, Targeting the endocannabinoid system in cancer therapy: a call for further research. Nat Med 8:547–550 Alexander A, Smith PF, Rosengren RJ, 2009, Cannabinoids in the treatment of cancer. Cancer Lett 285:6–12 Alpini G, Demorrow S, 2009, Changes in the endocannabinoid system may give insight into new and effective treatments for cancer. Vitam Horm 81:469–485 Fowler CJ, Gustafsson SB, Chung SC et al, 2010, Targeting the endocannabinoid system for the treatment of cancer–a practical view. Curr Top Med Chem 10:814–827 Bifulco M, Malfitano AM, Pisanti S et al, 2008, Endocannabinoids in endocrine and related tumours. Endocr Relat Cancer 15:391–408 Portella G, Laezza C, Laccetti P et al, 2003, Inhibitory effects of cannabinoid CB1 receptor stimulation on tumor growth and metastatic spreading: actions on signals involved in angiogenesis and metastasis. FASEB J 17:1771–1773 Pisanti S, Bifulco M, 2009, Endocannabinoid system modulation in cancer biology and therapy. Pharmacol Res 60:107–116 Bifulco M, Laezza C, Gazzerro P et al, 2007, Endocannabinoids as emerging suppressors of angiogenesis and tumor invasion, review). Oncol Rep 17:813–816 Sarfaraz S, Afaq F, Adhami VM et al, 2005, Cannabinoid receptor as a novel target for the treatment of prostate cancer. Cancer Res 65:1635–1641 Gazzerro P, Malfitano AM, Proto MC et al, 2010, Synergistic inhibition of human colon cancer cell growth by the cannabinoid CB1 receptor antagonist rimonabant and oxaliplatin. Oncol Rep 23:171–175 Biro T, Toth BI, Hasko G et al, 2009, The endocannabinoid system of the skin in health and disease: novel perspectives and therapeutic opportunities. Trends Pharmacol Sci 30:411–420 Casanova ML, Blazquez C, Martinez-Palacio J et al, 2003, Inhibition of skin tumor growth and angiogenesis in vivo by activation of cannabinoid receptors. J Clin Invest 111:43–50 Blazquez C, Carracedo A, Barrado L et al, 2006, Cannabinoid receptors as novel targets for the treatment of melanoma. FASEB J 20:2633–2635 Sosman JA, Puzanov I, 2006, Molecular targets in melanoma from angiogenesis to apoptosis. Clin Cancer Res 12:2376s–2383s Denkert C, Kobel M, Berger S et al, 2001, Expression of cyclooxygenase 2 in human malignant melanoma. Cancer Res 61:303–308 Tabolacci C, Lentini A, Provenzano B et al, 2010, Similar antineoplastic effects of nimesulide, a selective COX-2 inhibitor, and prostaglandin E1 on B16-F10 murine melanoma cells. Melanoma Res 20:273–279 Liu B, Khan WA, Hannun YA et al, 1995, 12(S)-hydroxyeicosatetraenoic acid and 13(S)-hydroxyoctadecadienoic acid regulation of protein kinase C-alpha in melanoma cells: role of receptor-mediated hydrolysis of inositol phospholipids. Proc Natl Acad Sci U S A 92:9323–9327 Raso E, Dome B, Somlai B et al, 2004, Molecular identification, localization and function of platelet-type 12-lipoxygenase in human melanoma progression, under experimental and clinical conditions. Melanoma Res 14:245–250 Ladanyi A, Timar J, Paku S et al, 1990, Selection and characterization of human melanoma lines with different liver-colonizing capacity. Int J Cancer 46:456–461 Dome B, Raso E, Dobos J et al, 2005, Parallel expression of alphaIIbbeta3 and alphavbeta3 integrins in human melanoma cells upregulates bFGF expression and promotes their angiogenic phenotype. Int J Cancer 116:27–35 Deli T, Varga N, Adam A et al, 2007, Functional genomics of calcium channels in human melanoma cells. Int J Cancer 121:55– 65 Grimaldi C, Pisanti S, Laezza C et al, 2006, Anandamide inhibits adhesion and migration of breast cancer cells. Exp Cell Res 312:363–373 Ramer R, Hinz B, 2008, Inhibition of cancer cell invasion by cannabinoids via increased expression of tissue inhibitor of matrix metalloproteinases-1. J Natl Cancer Inst 100:59–69 Hersey P, Zhuang L, Zhang XD, 2006, Current strategies in overcoming resistance of cancer cells to apoptosis melanoma as a model. Int Rev Cytol 251:131–158 Eberle J, Fecker LF, Hossini AM et al, 2008, Apoptosis pathways and oncolytic adenoviral vectors: promising targets and tools to overcome therapy resistance of malignant melanoma. Exp Dermatol 17:1–11 La Porta CA, 2009, Mechanism of drug sensitivity and resistance in melanoma. Curr Cancer Drug Targets 9:391–397 Sarnataro D, Grimaldi C, Pisanti S et al, 2005, Plasma membrane and lysosomal localization of CB1 cannabinoid receptor are dependent on lipid rafts and regulated by anandamide in human breast cancer cells. FEBS Lett 579:6343–6349 Osborne KD, Lee W, Malarkey EB, et al, 2009, Dynamic imaging of cannabinoid receptor 1 vesicular trafficking in cultured astrocytes. ASN Neuro 1. Bari M, Oddi S, De Simone C et al, 2008, Type-1 cannabinoid receptors colocalize with caveolin-1 in neuronal cells. Neuropharmacology 54:45–50 Engelman JA, 2009, Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer 9:550–562 Liu P, Cheng H, Roberts TM et al, 2009, Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov 8:627– 644 Madhunapantula SV, Robertson GP, 2009, The PTEN-AKT3 signaling cascade as a therapeutic target in melanoma. Pigment Cell Melanoma Res 22:400–419 Hudes GR, Berkenblit A, Feingold J et al, 2009, Clinical trial experience with temsirolimus in patients with advanced renal cell carcinoma. Semin Oncol 36(Suppl 3):S26–S36 Porta C, Figlin RA, 2009, Phosphatidylinositol-3-kinase/Akt signaling pathway and kidney cancer, and the therapeutic potential of phosphatidylinositol-3-kinase/Akt inhibitors. J Urol 182:2569– 2577 European public assessment report. Acomplia. http://www.ema.europa. eu/docs/en_GB/document_library/EPAR_-_Summary_for_ the_public/human/000666/WC500021282.pdf. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 857 EP 866 DI 10.1007/s12253-012-9515-y PG 10 ER PT J AU Jethon, A Pula, B Piotrowska, A Wojnar, A Rys, J Dziegiel, P Podhorska-Okolow, M AF Jethon, Aleksandra Pula, Bartosz Piotrowska, Aleksandra Wojnar, Andrzej Rys, Janusz Dziegiel, Piotr Podhorska-Okolow, Marzena TI Angiotensin II Type 1 Receptor (AT-1R) Expression Correlates with VEGF-A and VEGF-D Expression in Invasive Ductal Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Angiotensin receptors; VEGF; Angiogenesis; Lymphangiogenesis; Breast cancer ID Angiotensin receptors; VEGF; Angiogenesis; Lymphangiogenesis; Breast cancer AB Recent studies point to the involvement of angiotensin II (Ang II) receptor type 1 (AT-1R) on processes of metastasing, stimulation of invasiveness and angiogenesis in tumours. In this study, the correlation between intensity of AT-1R expression and expression of lymph- and angiogenesis markers in invasive ductal breast cancers (IDC) was examined. Immunohistochemical studies (IHC) were performed on archival material of 102 IDC cases. Only 28 (27.5%) cases manifested low AT-1R expression while 74 (72.5%) cases demonstrated a moderate or pronounced AT- 1R expression. Expression intensity of AT-1R was found to correlate with expressions of VEGF-A (r00.26; p00.008) and VEGF-D (r00.24; p00.015). Out of the examined markers of angiogenesis and lymphangiogenesis only the pronounced expression of VEGF-C was found to correlate with patient poor clinical outcome (p00.009). The positive correlation between AT-1R and VEGF-A and VEGF-D could point to stimulatory action of Ang II on their expression what might result in augmented lymph- and angiogenesis in IDC. C1 [Jethon, Aleksandra] Wroclaw Medical University, Department of Human Morphology and Embryology, Chalubinskiego 6a, 50-368 Wroclaw, Poland. [Pula, Bartosz] Wroclaw Medical University, Department of Human Morphology and Embryology, Chalubinskiego 6a, 50-368 Wroclaw, Poland. [Piotrowska, Aleksandra] Wroclaw Medical University, Department of Human Morphology and Embryology, Chalubinskiego 6a, 50-368 Wroclaw, Poland. [Wojnar, Andrzej] Lower Silesian Centre of Oncology, Department of PathomorphologyWroclaw, Poland. [Rys, Janusz] Maria Sklodowska - Curie Institute, Cracow Branch, Oncology Center, Department of Tumour PathologyKrakow, Poland. [Dziegiel, Piotr] Wroclaw Medical University, Department of Human Morphology and Embryology, Chalubinskiego 6a, 50-368 Wroclaw, Poland. [Podhorska-Okolow, Marzena] Wroclaw Medical University, Department of Human Morphology and Embryology, Chalubinskiego 6a, 50-368 Wroclaw, Poland. RP Podhorska-Okolow, M (reprint author), Wroclaw Medical University, Department of Human Morphology and Embryology, 50-368 Wroclaw, Poland. EM marzenna.podhorska-okolow@am.wroc.pl CR Ferlay J, Shin H-R, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127:2893–2917 Cuzick J, 2010, Breast cancer prevention in the developing world. Breast Cancer Res 12:S9 Chappell M, 2010, Angiotensin-converting enzyme 2 autoantibodies: further evidence for a role of the renin-angiotensin system in inflammation. Arthritis Res Ther 12:128 De Paepe B, Verstraeten VL, De Potter CR, Vakaet LA, Bullock GR, 2001, Growth stimulatory angiotensin II type-1 receptor is upregulated in breast hyperplasia and in situ carcinoma but not in invasive carcinoma. Histochem Cell Biol 116:247–254 De Gasparo M, Catt KJ, Inagami T, Wright JW, Unger T, 2000, International union of pharmacology. XXIII. The angiotensin II receptors. Pharmacol Rev 52:415–472 Gwathmey TM, Shaltout HA, Pendergrass KD, Pirro NT, Figueroa JP, Rose JC, Diz DI, Chappell MC, 2009, Nuclear angiotensin II type 2, AT2, receptors are functionally linked to nitric oxide production. Am J Physiol Renal Physiol 296:1484–1493 Muscella A, Greco S, Elia MG, Storelli C, Marsigliante S, 2002, Angiotensin II stimulation of Na+/K + ATPase activity and cell growth by calcium-independent pathway in MCF-7 breast cancer cells. J Endocrinol 173:315–323 Tahmasebi M, Barker S, Puddefoot JR, Vinson GP, 2006, Localisation of renin-angiotensin system, RAS, components in breast. Br J Cancer 95:67–74 Steckelings UM, Kaschina E, Unger T, 2005, The AT2 receptor— a matter of love and hate. Peptides 26:1401–1409 Bonnet F, Cao Z, Cooper ME, 2001, Apoptosis and angiotensin II: yet another renal regulatory system? Exp Nephrol 9:295–300 Deshayes F, Nahmias C, 2005, Angiotensin receptors: a new role in cancer? Trends Endocrinol Metabol 16:293–299 Fujimoto Y, Sasaki T, Tsuchida A, Chayama K, 2001, Angiotensin II type 1 receptor expression in human pancreatic cancer and growth inhibition by angiotensin II type 1 receptor antagonist. FEBS Letters 495:197–200 Goldfarb DA, Diz DI, Tubbs RR, Ferrario CM, Novick AC, 1994, Angiotensin II receptor subtypes in the human renal cortex and renal cell carcinoma. J Urol 151:208–213 Batra VK, Gopalakrishnan V, McNeill JR, Hickie RA, 1994, Angiotensin II elevates cytosolic free calcium in human lung adenocarcinoma cells via activation of AT1 receptors. Cancer Lett 76:19–24 Inwang ER, Puddefoot JR, Brown CL, Goode AW, Marsigliante S, Ho MM, Payne JG, Vinson GP, 1997, Angiotensin II type 1 receptor expression in human breast tissues. Br J Cancer 75:1279– 1283 Greco S, Elia MG, Muscella A, Storelli C, Marsigliante S, 2002, AT1 angiotensin II receptor mediates intracellular calcium mobilization in normal and cancerous breast cells in primary culture. Cell Calcium 32:1–10 Suzuki Y, Ruiz-Ortega M, Lorenzo O, Ruperez M, Esteban V, Egido J, 2003, Inflammation and angiotensin II. Int J Biochem Cell Biol 35:881–900 Puddefoot JR, Udeozo UK, Barker S, Vinson GP, 2006, The role of angiotensin II in the regulation of breast cancer cell adhesion and invasion. Endocr Relat Cancer 13:895–903 Herr D, Rodewald M, Fraser HM, Hack G, Konrad R, Kreienberg R, Wulff C, 2008, Potential role of Renin-Angiotensin-system for tumor angiogenesis in receptor negative breast cancer. Gynecol Oncol 109:418–425 Suganuma T, Ino K, Shibata K, Kajiyama H, Nagasaka T, Mizutani S, Kikkawa F, 2005, Functional expression of the angiotensin II type 1 receptor in human ovarian carcinoma cells and its blockade therapy resulting in suppression of tumor invasion, angiogenesis, and peritoneal dissemination. Clin Cancer Res 11:2686–2694 Khakoo AY, Sidman RL, Pasqualini R, Arap W, 2008, Does the renin-angiotensin system participate in regulation of human vasculogenesis and angiogenesis? Cancer Res 68:9112– 9115 Fox SB, Gasparini G, Harris AL, 2001, Angiogenesis: pathological, prognostic, and growth-factor pathways and their link to trial design and anticancer drugs. Lancet Oncol 2:278–289 Imanishi T, Hano T, Nishio I, 2004, Angiotensin II potentiates vascular endothelial growth factor-induced proliferation and network formation of endothelial progenitor cells. Hypertens Res 27:101–108 Wasa J, Sugiura H, Kozawa E, Kohyama K, Yamada K, Taguchi O, 2011, The tumor suppressive effect of angiotensin II type 1 receptor antagonist in a murine osteosarcoma model. Anticancer Res 31:123–127 Gomulkiewicz A, Podhorska-Okolow M, Szulc R, Smorag Z, Wojnar A, Zabel M, Dziegiel P, 2010, Correlation between metallothionein, MT, expression and selected prognostic factors in ductal breast cancers. Folia Histochem Cytobiol 48:242–248 Remmele W, Stegner HE, 1987, Recommendation for uniform definition of an immunoreactive score, IRS, for immunohistochemical estrogen receptor detection, ER-ICA, in breast cancer tissue. Pathologe 8:138–40 Willis LM, El-Remessy AB, Somanath PR, Deremer DL, Fagan SC, 2011, Angiotensin receptor blockers and angiogenesis: clinical and experimental evidence. Clin Sci 120:307–319 Greco S,Muscella A, EliaMG, Salvatore P, Storelli C,Marsigliante S, 2002, Activation of angiotensin II type I receptor promotes protein kinase C translocation and cell proliferation in human cultured breast epithelial cells. J Endocrinol 174:205–214 Rhodes DR, Ateeq B, Cao Q, Tomlins SA, Mehra R, Laxman B, Kalyana-Sundaram S, Lonigro RJ, Helgeson BE, Bhojani MS, Rehemtulla A, Kleer CG, Hayes DF, Lucas PC, Varambally S, Chinnaiyan AM, 2009, AGTR1 overexpression defines a subset of breast cancer and confers sensitivity to losartan, an AGTR1 antagonist. Proc Natl Acad Sci U S A 106:10284–10289 Kosugi M, Miyajima A, Kikuchi E, Kosaka T, Horiguchi Y, Murai M, 2007, Effect of angiotensin II type 1 receptor antagonist on tumor growth and angiogenesis in a xenograft model of human bladder cancer. Human Cell 20:1–9 Imai N, Hashimoto T, Kihara M, Yoshida S, Kawana I, Yazawa T, Kitamura H, Umemura S, 2006, Roles for host and tumor angiotensin II type 1 receptor in tumor growth and tumor-associated angiogenesis. Lab Invest 87:189–198 Fujita M, Hayashi I, Yamashina S, Fukamizu A, Itoman M, Majima M, 2005, Angiotensin type 1a receptor signalingdependent induction of vascular endothelial growth factor in stroma is relevant to tumor-associated angiogenesis and tumor growth. Carcinogenesis 26:271–279 Noguchi R, Yoshiji H, Ikenaka Y, Namisaki T, Kitade M, Kaji K, Yoshii J, Yanase K, Yamazaki M, Tsujimoto T, Kawaratani H, Fukui H, 2009, Synergistic inhibitory effect of gemcitabine and angiotensin type-1 receptor blocker, losartan, on murine pancreatic tumor growth via anti-angiogenic activities. Oncol Rep 22:355–360 Gu Y, Qi X, Guo S, 2008, Lymphangiogenesis induced by VEGFC and VEGF-D promotes metastasis and a poor outcome in breast carcinoma: a retrospective study of 61 cases. Clin Exp Metastasis 25:717–725 Mohammed RAA, Green A, El-Shikh S, Paish EC, Ellis IO, Martin SG, 2007, Prognostic significance of vascular endothelial cell growth factors -A, -C and -D in breast cancer and their relationship with angio- and lymphangiogenesis. Br J Cancer 96:1092–1100 Mylona E, NomikosA,Alexandrou P,Giannopoulou I,Keramopoulos A, Nakopoulou L, 2007, Lymphatic and blood vessel morphometry in invasive breast carcinomas: relation with proliferation and VEGF-C and -D proteins expression. Histol Histopathol 22:825–835 Gisterek I, Matkowski R, Kozlak J, Dus D, Lacko A, Szelachowska J, Kornafel J, 2007, Evaluation of prognostic value of VEGF-C and VEGF-D in breast cancer - 10 years follow-up analysis. Anticancer Res 27:2797–2802 Mylona E, Alexandrou P, Mpakali A, Giannopoulou I, Liapis G, Markaki S, Keramopoulos A, Nakopoulou L, 2007, Clinicopathological and prognostic significance of vascular endothelial growth factors, VEGF)-C and -D and VEGF receptor 3 in invasive breast carcinoma. Eur J Surg Oncol 33:294–300 van der Schaft DWJ, Pauwels P, Hulsmans S, Zimmermann M, van de Poll-Franse LV, Griffioen AW, 2007, Absence of lymphangiogenesis in ductal breast cancer at the primary tumor site. Cancer Lett 254:128–136 Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA, 2005, Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/ CXCL12 secretion. Cell 121:335–348 Eubank TD, Roberts RD, Khan M, Curry JM, Nuovo GJ, Kuppusamy P, Marsh CB, 2009, Granulocyte macrophage colony-stimulating factor inhibits breast cancer growth and metastasis by invoking an anti-angiogenic program in tumor-educated macrophages. Cancer Res 69:2133–2140 Joimel U, Gest C, Soria J, Pritchard LL, Alexandre J, Laurent M, Blot E, Cazin L, Vannier JP, Varin R, Li H, Soria C, 2010, Stimulation of angiogenesis resulting from cooperation between macrophages and MDA-MB-231 breast cancer cells: proposed molecular mechanism and effect of tetrathiomolybdate. BMC Cancer 10:375 Lambert DW, Clarke NE, Turner AJ, 2010, Not just angiotensinases: new roles for the angiotensin-converting enzymes. Cell Mol Life Sci 67:89–98 Gonzalez-Zuloeta Ladd AM, Arias Vasquez A, Sayed-Tabatabaei FA, Coebergh JW, Hofman A, Njajou O, Stricker B, van Duijn C, 2005, Angiotensin-converting enzyme gene insertion/deletion polymorphism and breast cancer risk. Cancer Epidemiol Biomarkers Prev 14:2143–2146 Gonzalez-Zuloeta Ladd AM, Arias Vasquez A, Siemes C, Yazdanpanah M, Coebergh JW, Hofman A, Stricker BH, van Duijn CM, 2007, Differential roles of angiotensinogen and angiotensin receptor type 1 polymorphisms in breast cancer risk. Breast Cancer Res Treat 10:299–304 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 867 EP 873 DI 10.1007/s12253-012-9516-x PG 7 ER PT J AU Giaginis, C Politi, E Alexandrou, P Sfiniadakis, J Kouraklis, G Theocharis, S AF Giaginis, Costantinos Politi, Ekaterini Alexandrou, Paraskevi Sfiniadakis, John Kouraklis, Gregory Theocharis, Stamatios TI Expression of Peroxisome Proliferator Activated Receptor-Gamma (PPAR-γ) in Human Non-small Cell Lung Carcinoma: Correlation with Clinicopathological Parameters, Proliferation and Apoptosis Related Molecules and Patients’ Survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PPAR-γ; Non-small cell lung carcinoma; Immunohistochemistry; Cell cycle; Cell proliferation; Apoptosis ID PPAR-γ; Non-small cell lung carcinoma; Immunohistochemistry; Cell cycle; Cell proliferation; Apoptosis AB Peroxisome proliferator-activated receptor-γ (PPAR-γ) has currently been considered as molecular target for the treatment of human metabolic disorders. PPAR-γ has also been implicated in the pathogenesis and progression of several types of cancer, being associated with cell differentiation, growth and apoptosis. The present study aimed to evaluate the clinical significance of PPAR-γ expression in non-small cell lung carcinoma (NSCLC). PPAR-γ protein expression was assessed immunohistochemically in tumoral samples of 67 NSCLC patients and was statistically analyzed in relation to clinicopathological parameters, proliferation and apoptosis related molecules and patients’ survival. Positive PPAR-γ expression was prominent in 30 (45 %) out of 67 NSCLC cases. PPAR-γ positivity was more frequently observed in squamous cell lung carcinoma cases compared to lung adenocarcinoma ones (p00.048). PPAR-γ positivity was significantly associated with bcl-2 positivity (p00.016) and borderline with c-myc positivity (p00.052), whereas non associations with grade of differentiation, TNM stage, Ki-67, p53, bax proteins’ expression and patients’ survival were noted. In the subgroup of squamous cell lung carcinoma cases, PPAR-γ positivity was significantly associated with tumor size (p00.038), while in lung adenocarcinoma ones with histopathological grade of differentiation (p00.026). The present study supported evidence for possible participation of PPAR-γ in the biological mechanisms underlying the carcinogenic evolution of the lung. Although the survival prediction using PPAR-γ expression as a marker seems uncertain, the observed correlation with apoptosis related proteins reinforces the potential utility of PPAR-γ ligands as cell cycle modulators in future therapeutic approaches in lung cancer. C1 [Giaginis, Costantinos] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias Street, 11527 Athens, Greece. [Politi, Ekaterini] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias Street, 11527 Athens, Greece. [Alexandrou, Paraskevi] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias Street, 11527 Athens, Greece. [Sfiniadakis, John] Naval Hospital Athens, Department of Pathology, 70, Dinokratous Street, 11521 Athens, Greece. [Kouraklis, Gregory] University of Athens, Medical School, Second Department of Propedeutic Surgery, 17, Ag. Thoma Street, 11527 Athens, Greece. [Theocharis, Stamatios] University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 75 Mikras Asias Street, 11527 Athens, Greece. RP Giaginis, C (reprint author), University of Athens, Medical School, Department of Forensic Medicine and Toxicology, 11527 Athens, Greece. EM cgiaginis@yahoo.gr CR Willson TM, Brown PJ, Strenbach DD, Henke BR, 2000, The PPARs: From orphan receptors to drug discovery. J Med Chem 43:527–550. Berger J, Moller DE, 2002, The mechanisms of action of PPARs. Ann Rev Med 53: 409–435. Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH, Kurokawa R, Rosenfeld MG, Willson TM, Glass CK, Milburn MV, 1998, Ligand binding and co-activator assembly of the peroxisome proliferators activated receptor-γ. Nature 395:137–143. Fajas L, Auboeuf D, Raspe E, Shoonjans K, Lefebvre AM, Saladin R, Najib J, Laville M, Fruchart JC, Deeb S, Vidal-Puig A, Flier J, Briggs MR, Staels B, Vidal H, Auwerx J, 1997, The organization, promoter analysis and expression of the human PPARγ gene. J Biol Chem 272:18779–1878. Feige JN, Gelman L, Michalik L, Desvergne B, Wahli W, 2006, From: molecular action to physiological outputs: Peroxisome proliferator-activated receptors are nuclear receptors at the crossroads of key cellular functions. Prog Lipid Res 45:120–159. Desvergne B, Wahli W, 1999, Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocr Rev 20:649–688. Hamza MS, Pott S, Vega VB, Thomsen JS, Kandhadayar GS, Ng PW, Chiu KP, Pettersson S,Wei CL, Ruan Y, Liu ET, 2009, De-novo identification of PPARgamma/RXR binding sites and direct targets during adipogenesis. PLoS One 4:e4907. Wysowski DK, Armstrong G, Governale L, 2003, Rapid increase in the use of oral Antidiabetic drugs in the United States, 1990– 2001. Diabetes Care 26:1852–1855. Sugden MC, Zariwala MG, Holness MJ, 2009, PPARs and the orchestration of metabolic fuel selection. Pharmacol Res 60:141– 150. Panigraphy D, Huang S, Kieran MW, Kaipainen A, 2005, PPARγ as a therapeutic target for tumor angiogenesis and metastasis. Cancer Biol Ther 4:687–693. Giaginis C, Tsantili-Kakoulidou A, Theocharis S, 2008, Peroxisome Proliferator-Activated Receptor-γ, PPAR-γ, ligands: potential pharmacological agents for targeting the angiogenesis signaling cascade in cancer. PPAR Res 2008:431763. Theocharis S, Margeli A, Vielh P, Kouraklis G, 2004, Peroxisome proliferator-activated receptor-gamma ligands as cell-cycle modulators. Cancer Treat Rev 30:545–554. Giaginis C, Tsantili-Kakoulidou A, Theocharis S, 2007, Peroxisome proliferator activated receptor-gamma ligands as bone turnover modulators. Expert Opin Investig Drugs 16:195–207. Rizzo G, Fiorucci S, 2006, PPARs and other nuclear receptors in inflammation. Curr Opin Pharm 6:421–427. Giaginis C, Tsourouflis G, Theocharis S, 2008, Peroxisome proliferator-activated receptor-gamma, PPAR-gamma, ligands: novel pharmacological agents in the treatment of ischemia reperfusion injury. Curr Mol Med 8:562–579. Giaginis C, Spanopoulou E, Theocharis S, 2008, PPAR-gamma signaling pathway in placental development and function: a potential therapeutic target in the treatment of gestational diseases. Expert Opin Ther Targets 12:1049–1063. Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60:277–300. D' Addario G, Felip E, 2009, Non-small-cell lung cancer: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol 20:68–70. Mendez D, Alshanqeety O, Warner KE, Lantz PM, Courant PN, 2011, The impact of declining smoking on radon-related lung cancer in the United States. Am J Public Health 101:310– 314 Alberg AJ, Brock MV, Samet JM, 2005, Epidemiology of lung cancer: looking to the future. J Clin Oncol 23:3175–3185. Nemenoff RA, Winn RA, 2005, Role of nuclear receptors in lung tumourigenesis. Eur J Cancer 41:2561–2568. Keshamouni VG, Han S, Roman J, 2007, Peroxisome proliferatoractivated receptors in lung cancer. PPAR Res 2007:90289 Tsubouchi Y, San H, Kawahito Y Mukai S, Yamada R, Kohno M, Inoue K, Hla T, Kondo M, 2000, Inhibition of human cancer cell growth by peroxisome proliferator-activated receptor-gamma agonists through induction of apoptosis. Biochem Biophys Res Commun 270:400–405. Keshamouni VG, Reddy RC, Arenberg DA, Joel B, Thannickal VJ, Kalemkerian GP, Standiford TJ, 2004, Peroxisome proliferator-activated receptor-γ activation inhibits tumor progression in non-small-cell lung cancer. Oncogene 23:100–108. Allred CD, Kilgore MW, 2005, Selective activation of PPARγ in breast, colon, and lung cancer cell lines. Mol Cell Endocrinol 235:21–29. Chang TH, Szabo E, 2000, Induction of differentiation and apoptosis by ligands of peroxisome proliferator-activated receptor gamma in non small cell lung cancer. Cancer Res 60:1129–1138. InoueK,KawahitoY, TsubouchiY, Yamada R,KohnoM, Hosokawa Y, Katoh D, Bishop-Bailey D, Hla T, Sano H, 2001, Expression of peroxisome proliferator-activated receptor, PPAR)-gamma in human lung cancer. Anticancer Res 21:2471–2476. Sasaki H, Tanahashi M, Yukiue H, Moiriyama S, Kobayashi Y, Nakashima Y, Kaji M, Kiriyama M, Fukai I, Yamakawa Y, Fujii Y, 2002, Decreased peroxisome proliferator-activated receptorgamma gene expression was correlated with poor prognosis in patients with lung cancer. Lung cancer 36:71–76. Theocharis S, Kanelli H, Politi E, Margeli A, Karkandaris C, Philippides T, Koutselinis A, 2002, Expression of peroxisome proliferator-activated receptor-gamma in non-small cell lung carcinoma: correlation with histological type and grade. Lung Cancer 36:249–255. Wick M, Hurteau G, Dessev C, Chan D, Geraci MW, Winn RA, Heasley LE, Nemenoff RA, 2002, Peroxisome proliferatoractivated receptor-gamma is a target of nonsteroidal antiinflammatory drugs mediating cyclooxygenase-independent inhibition of lung cancer cell growth. Mol Pharmacol 62:1207–1214. Han S, Sidell N, Fisher PB, Roman J, 2004, Up-regulation of p21 gene expression by peroxisome proliferator-activated receptor γ in human lung carcinoma cells. Clin Cancer Res 10:1911–1919. Li M, Lee TW, Mok TSK,Warner TD, Yim APC, Chen GG, 2005, Activation of peroxisome proliferator-activated receptor-γ by troglitazone, TGZ, inhibits human lung cell growth. J Cell Biochem 96:760–774. Li M, Lee TW, Yim AP, Mok TS, Chen GG, 2006, Apopotsis induced by troglitazone is both peroxisome proliferator-activated receptor-γ- and ERK-dependent in human non-small cell lung cancer cells. J Cell Physiol 209:428–438. Zou W, Liu X, Yue P, Khuri FR, Sun SY, 2007, PPARg ligands enhance TRAIL-induced apoptosis through DR5 upregulation and c-FLIP downregulation in human lung cancer cells. Cancer Biol Ther 6:99–106. Keshamouni VG, Arenberg DA, Reddy RC, Newstead MJ, Anthwal S, Standiford TJ, 2005, PPAR-gamma activation inhibits angiogenesis by blocking ELR+CXC chemokine production in non-small cell lung cancer. Neoplasia 7:294–301. Bren-Mattison Y, Van Putten V, Chan D, Winn R, Geraci MW, Nemenoff RA, 2005, Peroxisome proliferator-activated receptorgamma, PPARgamma, inhibits tumorigenesis by reversing the undifferentiated phenotype of metastatic non-small-cell lung cancer cells, NSCLC). Oncogene 24:1412–1422. Govindarajan R, Ratnasinghe L, Simmons DL, Siegel ER, Midathada MV, Kim L, Kim PJ, Owens RJ, Lang NP, 2007, Thiazolidinediones and the risk of lung, prostate, and colon cancer in patients with diabetes. J Clin Oncol 25:1476–1481. Greene FLPD, Fleming ID, Fritz AG, Balch CM, Haller DG, Morrow M, 2002, AJCC cancer staging manual, 6th edn. Springer, New York Nair A, Klusmann MJ, Jogeesvaran KH, Grubnic S, Green SJ, Vlahos I, 2011, Revisions to the TNM Staging of Non-Small Cell Lung Cancer: Rationale, Clinicoradiologic Implications, and Persistent Limitations. Radiographics 31:215–238. Lababede O, Meziane M, Rice T, 2011, Seventh edition of the cancer staging manual and stage grouping of lung cancer: quick reference chart and diagrams. Chest 139:183–189. Giaginis C, Katsamangou E, Tsourouflis G, Zizi-Serbetzoglou D, Kouraklis G, Theocharis S, 2009, Peroxisome proliferatoractivated receptor-gamma and retinoid X receptor-alpha expression in pancreatic ductal adenocarcinoma: association with clinicopathological parameters, tumor proliferative capacity, and patients' survival. Med Sci Monit 15: BR148-156. Theocharis S, Giaginis C, Parasi A, Margeli A, Kakisis J, Agapitos E, Kouraklis G, 2007, Expression of Peroxisome Proliferator- Activated Receptor-gamma in colon cancer: correlation with histopathological parameters, cell cycle related molecules and patients’ survival. Dig Dis Sci 52:2305–2311. Ondrey F, 2009, Peroxisome proliferator-activated receptor gamma pathway targeting in carcinogenesis: implications for chemoprevention. Clin Cancer Res 15:2–8. Sultana H, Kigawa J, Kanamori Y, Itamochi H, Oishi T, Sato S, Kamazawa S, Ohwada M, Suzuki M, Terakawa N, 2003, Chemosensitivity and p53-Bax pathway-mediated apoptosis in patients with uterine cervical cancer. Ann Oncol 14:214–219. Kirkin V, Joos S, Zornig M, 2004, The role of Bcl-2 family members in tumorigenesis. Biochim Biophys Acta 1644:229–249 Yang FG, Zhang ZW, Xin DQ, Shi CJ, Wu JP, Guo YL, Guan YF, 2005, Peroxisome proliferator-activated receptor γ ligands induce cell cycle arrest and apoptosis in human renal cell carcinoma cell lines. Acta Pharmacol. Sin 26:753–761. Liu JJ, Huang RW, Lin DJ, Peng J, Wu XY, Lin Q, Pan XL, Song YQ, Zhang MH, Hou M, Chen F, 2005, Expression of surviving and bax/bcl-2 in peroxisome proliferator activated receptor γ ligands induces apoptosis on human myeloid leukemia cells in vitro. Ann Oncol 16:455–459. Liu DC, Zang CB, Liu HY, Possinger K, Fan SG, Elstner E, 2004, A novel PPAR alpha/gamma dual agonist inhibits cell growth and induces apoptosis in human glioblastoma T98G cells. Acta Pharmacol Sin 25:1312–1319. Elstner E, Muller C, Koshizuka K, Williamson EA, Park D, Asou H, Shintaku P, Said JW, Heber D, Koeffler HP, 1998, Ligands for peroxisome proliferator activated receptor gamma and retinoic acid receptor inhibit growth and induce apoptosis of human breast cancer cells in vitro and in BNX mice. Proc Natl Acad Sci USA 95:8806–8811. Strakova N, Ehrmann J, Bartos J, Malikova J, Dolezel J, Kolar Z, 2005, Peroxisome proliferator-activated receptors, PPAR, agonists affect cell viability, apoptosis and expression of cell cycle related proteins in cell lines of gliab brain tumors. Neoplasma 52:126–136. Shimada T, Kojima K, Yoshiura K, Hiraishi H, Terano A, 2002, Characteristics of the peroxisome proliferator activated receptor γ, PPARγ, ligand induced apoptosis in colon cancer cells. Gut 50:658–664. Fulzele SV, Chatterjee A, Shaik MS, Jackson T, Ichite N, Singh M, 2007, 15-deoxy-detal 12,14-prostaglandin J2 enhances docetaxel anti-tumor activity against A549 and H460 non small cell lines and xenograft tumors. Anticancer Drugs 18:65–78. Yamakawa-KarakidaN, SugitaK, Inukai T, GoiK,NakamuraM,Uno K, Sato H, Kagami K, Barker N,Nakazawa S, 2002, Ligand activation of peroxisome proliferator activated receptor γ induces apoptosis of leukemia cells by down-regulating the c-myc gene expression via blockage of the Tcf-4 activity. Cell death Diff 9:513–526. Sikand K, Kaul D, Varma N, 2006, Receptor Ck-dependent signaling regulates hTERT gene transcription. BMC Cell Biology 7:2. Han SW, Greene ME, Pitts J, Wada RK, Sidell N, 2001, Novel expression and function of peroxisome proliferator-activated receptor gamma, PPARgamma, in human neuroblastoma cells. Clin Cancer Res 7:98–104. Zhang GY, Ahmed N, Riley C et al., 2005, Enhanced expression of peroxisome proliferator-activated receptor gamma in epithelial ovarian carcinoma. Br J Cancer 92:113–119. Lee TW, Chen GG, Xu H, Yip JH, Chak EC, Mok TS, Yim AP, 2003, Differential expression of inducible nitric oxide synthase and peroxisome proliferator-activated receptor γ in non-small cell lung carcinoma. Eur J Cancer 39:1296–301. Kim HJ, Hwang JY, Kim HJ, Choi WS, Lee JH, Kim HJ, Chang KC, Nishinaka T, Yabe-Nishimura C, Seo HG, 2007, Expression of a peroxisome proliferator-activated receptor gamma 1 splice variant that was identified in human lung cancers suppresses cell death induced by cisplatin and oxidative stress. Clin Cancer Res 13:2577–2583. Jiang WG, Redfern A, Bryce RP, Mansel RE, 2000, Peroxisome proliferator activated receptor-gamma, PPAR-gamma, mediates the action of gamma linolenic acid in breast cancer cells. Prostaglandins Leukot Essent Fatty Acids 62:119–127. Katagiri Y, Takeda K, Yu ZX, Ferrans VJ, Ozato K, Guroff G, 2000, Modulation of retinoid signaling through NGF-induced nuclear export of NGFI-B. Nat Cell Biol 2:435–440. Ikezoe T, Miller CW, Kawano S, Heaney A, Williamson EA, Hisatake J, Green E, Hofmann W, Taguchi H, Koeffler HP, 2001, Mutational analysis of the peroxisome proliferatoractivated receptor γ gene in human malignancies. Cancer Res 61:5307–5310. Sun Y, 2006, p53 and its downstream proteins as molecular targets of cancer. Mol Carcinog 45:409–415. Breuer RH, Postmus PE, Smit EF, 2005, Molecular pathology of non-small-cell lung cancer. Respiration 72:229–232. Viktorsson K, De Petris L, Lewensohn R, 2005, The role of p53 in treatment responses of lung cancer. Biochem Biophys Res Commun 331:868–880. Martin B, Paesman SM, Mascaux C, Berghmans T, Lothaire P, Meert A-P, Lafitte J-J, Sculier J-P, 2004, Ki-67 expression and patients survival in lung cancer: systematic review of the literature with meta-analysis. Br J Cancer 91:2018–2025 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 875 EP 883 DI 10.1007/s12253-012-9517-9 PG 9 ER PT J AU Zaczek, A Markiewicz, A Supernat, A Bednarz-Knoll, N Brandt, B Seroczynska, B Skokowski, J Szade, J Czapiewski, P Biernat, W Welnicka-Jaskiewicz, M Jassem, J AF Zaczek, Anna Markiewicz, Aleksandra Supernat, Anna Bednarz-Knoll, Natalia Brandt, Burkhardt Seroczynska, Barbara Skokowski, Jaroslaw Szade, Jolanta Czapiewski, Piotr Biernat, Wojciech Welnicka-Jaskiewicz, Marzena Jassem, Jacek TI Prognostic Value of TOP2A Gene Amplification and Chromosome 17 Polysomy in Early Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Chromosome 17 polysomy; TOP2A amplification; qPCR; FISH; Prognostic factor ID Breast cancer; Chromosome 17 polysomy; TOP2A amplification; qPCR; FISH; Prognostic factor AB The aim of this study was to analyze the occurrence of TOP2A gene amplification and chromosome 17 polysomy in patients with early breast cancer and to correlate the status of these alterations with the prognostic significance expressed as patients’ clinical features and survival. Such concurrent analyses of TOP2A gene status and chromosome 17 polysomy have not been performed before. Study group included 149 consecutive stage I-III patients administered standard multimodality treatment. TOP2A abnormalities were examined by standard fluorescence in situ hybridization (FISH) and developed by our group quantitative real-time PCR (qPCR). TOP2A amplification and deletion assessed by FISH were found in 23% and 7% of the tumours, respectively, and by qPCR in 31% and 11% of the tumours, respectively. Chromosome 17 polysomy was detected in 40% of the cases. TOP2A amplification (by qPCR) correlated with shorter diseasefree survival (p00.03) and overall survival (p00.047), and the prognostic value of TOP2A was confirmed in the multivariate analysis (HR03.22, 95% CI 1.09–9.56, p00.03). TOP2A gene amplification, but not chromosome 17 polysomy, carries negative prognostic information in early breast cancer. Given the aforementioned results, qPCR might serve as a prognostic tool in determining the patient’s prognosis. C1 [Zaczek, Anna] University of Gdansk and Medical University of Gdansk, Intercollegiate Faculty of Biotechnology, Laboratory of Cell Biology, Department of Medical Biotechnology, Debinki 1, 80-211 Gdansk, Poland. [Markiewicz, Aleksandra] University of Gdansk and Medical University of Gdansk, Intercollegiate Faculty of Biotechnology, Laboratory of Cell Biology, Department of Medical Biotechnology, Debinki 1, 80-211 Gdansk, Poland. [Supernat, Anna] University of Gdansk and Medical University of Gdansk, Intercollegiate Faculty of Biotechnology, Laboratory of Cell Biology, Department of Medical Biotechnology, Debinki 1, 80-211 Gdansk, Poland. [Bednarz-Knoll, Natalia] University Medical Centre, Institute of Tumour Biology, Hamburg-Eppendorf, Martinistrasse 52, 20-246 Hamburg, Germany. [Brandt, Burkhardt] University Medical Centre, Institute of Tumour Biology, Hamburg-Eppendorf, Martinistrasse 52, 20-246 Hamburg, Germany. [Seroczynska, Barbara] Medical University of Gdansk, Bank of Frozen Tissues and Genetic Specimen, Debinki 1, 80-211 Gdansk, Poland. [Skokowski, Jaroslaw] Medical University of Gdansk, Bank of Frozen Tissues and Genetic Specimen, Debinki 1, 80-211 Gdansk, Poland. [Szade, Jolanta] Medical University of Gdansk, Department of Pathomorphology, Debinki 7, 80-211 Gdansk, Poland. [Czapiewski, Piotr] Medical University of Gdansk, Department of Pathomorphology, Debinki 7, 80-211 Gdansk, Poland. [Biernat, Wojciech] Medical University of Gdansk, Department of Pathomorphology, Debinki 7, 80-211 Gdansk, Poland. [Welnicka-Jaskiewicz, Marzena] Medical University of Gdansk, Department of Oncology and Radiotherapy, Debinki 7, 80-211 Gdansk, Poland. [Jassem, Jacek] Medical University of Gdansk, Department of Oncology and Radiotherapy, Debinki 7, 80-211 Gdansk, Poland. RP Zaczek, A (reprint author), University of Gdansk and Medical University of Gdansk, Intercollegiate Faculty of Biotechnology, Laboratory of Cell Biology, Department of Medical Biotechnology, 80-211 Gdansk, Poland. EM azaczek@gumed.edu.pl CR Lehmann U, Glockner S, Kleeberger W, Feist H, Rv W, Kreipe H, 2000, Detection of gene amplification in archival breast cancer specimens by laser-assisted microdissection and quantitative realtime polymerase chain reaction. Am J Pathol 156(6):1855–1864 Mano MS, Rosa DD, Azambuja ED, Ismael GFV, Durbecq V, 2007, The 17q12-q21 amplicon: Her2 and topoisomerase-IIa and their importance to the biology of solid tumours. Cancer Treat Rev 33:64–77 Park K, Kim J, Lim S, Han S, 2003, Topoisomerase II-alpha, topoII, and HER2 amplification in breast cancers and response to preoperative doxorubicin chemotherapy. Eur J Cancer 39, 5):631–634., DOI S0959804902007451 Jarvinen TAH, Liu ET, 2003, HER-2/neu and topoisomerase IIα in breast cancer. Breast Cancer Res Treat 78:299–311 Durbecq V, Paesmans M, Cardoso F, Desmedt C, Di Leo A, Chan S, Friedrichs K, Pinter T, Van Belle S, Murray E, Bodrogi I, Walpole E, Lesperance B, Korec S, Crown J, Simmonds P, Perren TJ, Leroy JY, Rouas G, Sotiriou C, Piccart M, Larsimont D, 2004, Topoisomerase-II alpha expression as a predictive marker in a population of advanced breast cancer patients randomly treated either with single-agent doxorubicin or single-agent docetaxel. Mol Cancer Ther 3(10):1207–1214., DOI 3/10/1207 O’Malley FP, Chia S, Tu D, Shepherd LE, Levine MN, Bramwell VH, Andrulis IL, Pritchard KI, 2009, Topoisomerase II alpha and responsiveness of breast cancer to adjuvant chemotherapy. J Natl Cancer Inst 101(9):644–650., DOI djp067 Arriola E, Maria S, Rodriguez-Pinilla LMBK, Jones RL, James M, Savage K, Smith IE, Dowsett M, Reis-Filho JS, 2007, Topoisomerase II alpha amplification may predict benefit from adjuvant anthracyclines in HER2 positive early breast cancer. Breast Cancer Res Treat 106:181–189 Knoop AS, Knudsen H, Balslev E, Rasmussen BB, Overgaard J, Nielsen KV, Schonau A, Gunnarsdottir K, Olsen KE, Mouridsen H, Ejlertsen B, 2005, Retrospective analysis of Topoisomerase IIa amplifications and deletions as predictive markers in primary breast cancer patients randomly assigned to Cyclophosphamide, Methotrexate, and Fluorouracil or Cyclophosphamide, Epirubicin, and Fluorouracil: Danish Breast Cancer Cooperative Group. J Clin Oncol 23(20):7483–7490 Nielsen KV, Ejlertsen B, Moller S, Jorgensen JT, Knoop A, Knudsen H, Mouridsen HT, 2008, The value of TOP2A gene copy number variation as a biomarker in breast cancer: update of DBCG trial 89D. Acta Oncol 47(4):725–734., DOI 791851530 Park K, Han S, Gwak GH, Kim HJ, Kim J, Kim KM, 2006, Topoisomerase II-alpha gene deletion is not frequent as its amplification in breast cancer. Breast Cancer Res Treat 98(3):337–342., DOI 10.1007/s10549-006-9170-7 Shackney SE, Singh SG, Yakulis R, Smith CA, Pollice AA, Petruolo S, Waggoner A, Hartsock RJ, 1995, Aneuploidy in breast cancer: a fluorescence in situ hybridization study. Cytometry 22, 4):282–291., DOI 10.1002/cyto.990220404 Staaf J, Jonsson G, Ringner M, Vallon-Christersson J, Grabau D, Arason A, Gunnarsson H, Agnarsson BA, Malmstrom PO, Johannsson OT, Loman N, Barkardottir RB, Borg A, 2010, Highresolution genomic and expression analyses of copy number alterations in HER2-amplified breast cancer. Breast Cancer Res 12(3): R25., DOI bcr2568 Gunn S, Yeh IT, Lytvak I, Tirtorahardjo B, Dzidic N, Zadeh S, Kim J, McCaskill C, Lim L, Gorre M, Mohammed M, 2010, Clinical array-based karyotyping of breast cancer with equivocal HER2 status resolves gene copy number and reveals chromosome 17 complexity. BMC Cancer 10:396., DOI 1471-2407-10-396 Moelans CB, de Weger RA, van Diest PJ, 2009, Absence of chromosome 17 polysomy in breast cancer: analysis by CEP17 chromogenic in situ hybridization and multiplex ligationdependent probe amplification. Breast Cancer Res Treat 120, 1):1–7., DOI 10.1007/s10549-009-0539-2 Reinholz MM, Bruzek AK, Visscher DW, Lingle WL, Schroeder MJ, Perez EA, Jenkins RB, 2009, Breast cancer and aneusomy 17: implications for carcinogenesis and therapeutic response. Lancet Oncol 10(3):267–277., DOI S1470-2045(09)70063-4 Krishnamurti U, Hammers JL, Atem FD, Storto PD, Silverman JF, 2009, Poor prognostic significance of unamplified chromosome 17 polysomy in invasive breast carcinoma. Mod Pathol 22, 8):1044–1048., DOI modpathol200961 Watters AD, Going JJ, Cooke TG, Bartlett JM, 2003, Chromosome 17 aneusomy is associated with poor prognostic factors in invasive breast carcinoma. Breast Cancer Res Treat 77, 2):109–114 Vanden Bempt I, Van Loo P, Drijkoningen M, Neven P, Smeets A, Christiaens MR, Paridaens R, De Wolf-Peeters C, 2008, Polysomy 17 in breast cancer: clinicopathologic significance and impact on HER-2 testing. J Clin Oncol 26(30):4869–4874., DOI JCO.2007.13.4296 Torrisi R, Rotmensz N, Bagnardi V, Viale G, Curto BD, Dell’orto P, Veronesi P, Luini A, D’Alessandro C, Cardillo A, Goldhirsch A, Colleoni M(2007, HER2 status in early breast cancer: relevance of cell staining patterns, gene amplification and polysomy 17. Eur J Cancer 43(16):2339–2344., DOI S0959-8049(07)00560-6 Jarvinen T, Tanner M, Rantanen V, Barlund M, Borg A, Grenman S, 2000, Amplification and deletion of topoisomerase IIa associate with ErbB-2 amplification and affect sensitivity to topoisomerase II Inhibitor doxorubicin in breast cancer. Am J Pathol 156:839– 847 Zaczek A, Markiewicz A, Jaskiewicz J, Pienkowski T, Rhone P, Jassem J, Welnicka-Jaskiewicz M, 2010, Clinical evaluation of developed PCR-based method with hydrolysis probes for TOP2A copy number evaluation in breast cancer samples. Clin Biochem 43(10–11):891–898., DOI S0009-9120(10)00193-1 McShane LM, Altman DG, SauerbreiW, Taube SE, Gion M, Clark GM, 2005, REporting recommendations for tumour MARKer prognostic studies, REMARK). Br J Cancer 93(4):387–391., DOI 6602678 Romero A, Martin M, Cheang MC, Lopez Garcia-Asenjo JA, Oliva B, He X, de la Hoya M, Garcia Saenz J, Arroyo Fernandez M, Diaz Rubio E, Perou CM, Llopis TC, 2011, Assessment of Topoisomerase II α status in breast cancer by quantitative PCR, gene expression microarrays, immunohistochemistry, and fluorescence in situ hybridization. Am J Pathol 178(4):1453–1460., DOI S0002-9440(11)00042-3 Hicks DG, Yoder BJ, Pettay J, Swain E, Tarr S, Hartke M, Skacel M, Crowe JP, Budd T, Tubbs RR, 2004, The incidence of topoisomerase II-alpha genomic alterations in adenocarcinoma of the breast and their relationship to human epidermal growth factor receptor-2 gene amplification: A fluorescence in situ hybridization study. Hum Pathol 36(4):348–356 Di Leo A, Gancberg D, Larsimont D, Tanner M, Jarvinen T, Rouas G, Dolci S, Leroy JY, Paesmans M, Isola J, Piccart MJ, 2002, HER-2 amplification and topoisomerase IIalpha gene aberrations as predictive markers in node-positive breast cancer patients randomly treated either with an anthracycline-based therapy or with cyclophosphamide, methotrexate, and 5-fluorouracil. Clin Cancer Res 8(5):1107–1116 Konecny GE, Pauletti G, Untch M, Wang HJ, Mobus V, Kuhn W, Thomssen C, Harbeck N, Wang L, Apple S, Janicke F, Slamon DJ, 2010, Association between HER2, TOP2A, and response to anthracycline-based preoperative chemotherapy in high-risk primary breast cancer. Breast Cancer Res Treat 120(2):481–489., DOI 10.1007/s10549-010-0744-z Slamon DJ, Press MF, 2009, Alterations in the TOP2A and HER2 genes: association with adjuvant anthracycline sensitivity in human breast cancers. J Natl Cancer Inst 101(9):615–618., DOI djp092 Herrington CS, Leek RD, McGee JO, 1995, Correlation of numerical chromosome 11 and 17 imbalance with metastasis of primary breast cancer to lymph nodes. J Pathol 176(4):353–359., DOI 10.1002/path.1711760406 Ichikawa D, Hashimoto N, Hoshima M, Yamaguchi T, Sawai K, Nakamura Y, Takahashi T, Abe T, Inazawa J, 1996, Analysis of numerical aberrations of specific chromosomes by fluorescent in situ hybridization as a diagnostic tool in breast cancer. Cancer 77, 10):2064–2069., DOI 10.1002/(SICI)1097-0142(19960515, 77:10<2064::AID-CNCR15>3.0.CO;2-T McManus DT, Patterson AH, Maxwell P, Hamilton PW, Anderson NH, Caughley LM, Toner PG, 1999, Interphase cytogenetics of chromosomes 11 and 17 in fine needle aspirates of breast cancer. Hum Pathol 30(2):137–144 Tsukamoto F, Miyoshi Y, Egawa C, Kasugai T, Takami S, Inazawa J, Noguchi S, 2001, Clinicopathologic analysis of breast carcinoma with chromosomal aneusomy detected by fluorescence in situ hybridization. Cancer 93(2):165–170 Fehm T, Morrison L, Saboorian H, Hynan L, Tucker T, Uhr J, 2002, Patterns of aneusomy for three chromosomes in individual cells from breast cancer tumors. Breast Cancer Res Treat 75, 3):227–239 Nakopoulou L, Giannopoulou I, Trafalis D, Gakiopoulou H, Keramopoulos A, Davaris P, 2002, Evaluation of numeric alterations of chromosomes 1 and 17 by in situ hybridization in invasive breast carcinoma with clinicopathologic parameters. Appl Immunohistochem Mol Morphol 10(1):20–28 Wang S, Hossein Saboorian M, Frenkel EP, Haley BB, Siddiqui MT, Gokaslan S, Hynan L, Ashfaq R, 2002, Aneusomy 17 in breast cancer: its role in HER-2/neu protein expression and implication for clinical assessment of HER-2/neu status. Mod Pathol 15, 2):137–145., DOI 10.1038/modpathol.3880505 Ma Y, Lespagnard L, Durbecq V, Paesmans M, Desmedt C, Gomez-Galdon M, Veys I, Cardoso F, Sotiriou C, Di Leo A, Piccart MJ, Larsimont D, 2005, Polysomy 17 in HER-2/neu status elaboration in breast cancer: effect on daily practice. Clin Cancer Res 11(12):4393–4399., DOI 11/12/4393 Merola R, Mottolese M, Orlandi G, Vico E, Cognetti F, Sperduti I, Fabi A, Vitelli G, Cianciulli AM, 2006, Analysis of aneusomy level and HER-2 gene copy number and their effect on amplification rate in breast cancer specimens read as 2+ in immunohistochemical analysis. Eur J Cancer 42(10):1501–1506., DOI S0959- 8049(06)00319-4 Dowsett M, Hanna WM, Kockx M, Penault-Llorca F, Ruschoff J, Gutjahr T, Habben K, van de Vijver MJ, 2007, Standardization of HER2 testing: results of an international proficiency-testing ring study. Mod Pathol 20(5):584–591., DOI 3800774 Marchio C, Lambros MB, Gugliotta P, Di Cantogno LV, Botta C, Pasini B, Tan DS, Mackay A, Fenwick K, Tamber N, Bussolati G, Ashworth A, Reis-Filho JS, Sapino A, 2009, Does chromosome 17 centromere copy number predict polysomy in breast cancer? A fluorescence in situ hybridization and microarray-based CGH analysis. J Pathol 219(1):16–24., DOI 10.1002/path.2574 Beser AR, Tuzlali S, Guzey D, Dolek Guler S, Hacihanefioglu S, Dalay N, 2007, HER-2, TOP2A and chromosome 17 alterations in breast cancer. Pathol Oncol Res 13(3):180–185., DOI PAOR.2007.13.3.0180 Jarvinen TA, Tanner M, Barlund M, Borg A, Isola J, 1999, Characterization of topoisomerase II alpha gene amplification and deletion in breast cancer. Genes Chromosomes Cancer 26, 2):142–150., DOI 10.1002/(SICI)1098-2264(199910)26:2<142:: AID-GCC6>3.0.CO;2-B Brase JC, Schmidt M, Fischbach T, Sultmann H, Bojar H, Koelbl H, Hellwig B, Rahnenfuhrer J, Hengstler JG, Gehrmann MC, 2010, ERBB2 and TOP2A in breast cancer: a comprehensive analysis of gene amplification, RNA levels, and protein expression and their influence on prognosis and prediction. Clin Cancer Res 16(8):2391–2401., DOI 1078-0432.CCR-09-2471 Bartlett JM, Munro AF, Dunn JA, McConkey C, Jordan S, Twelves CJ, Cameron DA, Thomas J, Campbell FM, Rea DW, Provenzano E, Caldas C, Pharoah P, Hiller L, Earl H, Poole CJ, 2010, Predictive markers of anthracycline benefit: a prospectively planned analysis of the UK National Epirubicin Adjuvant Trial, NEAT/BR9601). Lancet Oncol 11(3):266–274., DOI S1470-2045, 10)70006-1 Downey L, Livingston RB, Koehler M, Arbushites M, Williams L, Santiago A, Guzman R, Villalobos I, Di Leo A, Press MF, 2010, Chromosome 17 polysomy without human epidermal growth factor receptor 2 amplification does not predict response to lapatinib plus paclitaxel compared with paclitaxel in metastatic breast cancer. Clin Cancer Res 16(4):1281–1288., DOI 1078- 0432.CCR-09-1643 Bartlett JMS, Munro A, Cameron DA, Thomas J, Prescott R, Twelves CJ, 2008, Type 1 receptor tyrosine kinase profiles identify patients with enhanced benefit from anthracyclines in the BR9601 adjuvant breast cancer chemotherapy trial. J Clin Oncol 28(31) Glynn RW, Miller N, Whelan MC, Kerin MJ, 2010, Topoisomerase 2 alpha and the case for individualized breast cancer therapy. Ann Surg Oncol 17(5):1392–1397., DOI 10.1245/ s10434-009-0855-0 Bednarz N, Eltze E, Semjonow A, Rink M, Andreas A, Mulder L, Hannemann J, Fisch M, Pantel K,Weier HU, Bielawski KP, Brandt B, 2010, BRCA1 loss preexisting in small subpopulations of prostate cancer is associated with advanced disease and metastatic spread to lymph nodes and peripheral blood. Clin Cancer Res 16, 13):3340–3348., DOI 1078-0432.CCR-10-0150 Salido M, Tusquets I, Corominas JM, Suarez M, Espinet B, Corzo C, Bellet M, Fabregat X, Serrano S, Sole F, 2005, Polysomy of chromosome 17 in breast cancer tumors showing an overexpression of ERBB2: a study of 175 cases using fluorescence in situ hybridization and immunohistochemistry. Breast Cancer Res 7(2): R267–273., DOI bcr996 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 885 EP 894 DI 10.1007/s12253-012-9518-8 PG 10 ER PT J AU Ding, Y Huang, X Liu, T Fu, Y Tan, Z Zheng, H Zhou, T Dai, J Xu, W AF Ding, Yan Huang, Xiaobing Liu, Taiping Fu, Yong Tan, Zhangping Zheng, Hong Zhou, Taoli Dai, Jigang Xu, Wenyue TI The Plasmodium Circumsporozoite Protein, a Novel NF-κB Inhibitor, Suppresses the Growth of SW480 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Circumsporozoite protein; Nuclear transcription factor κB; Human colon cancer cell line; Proliferation; Apoptosis ID Circumsporozoite protein; Nuclear transcription factor κB; Human colon cancer cell line; Proliferation; Apoptosis AB The blocking of NF-κB activation is a promising strategy for the treatment of colorectal cancer. The circumsporozoite protein (CSP), a key component of the sporozoite stage of the malaria parasite, was recently reported to block NF-κB activation in hepatocytes. Thus, we investigated the effect of the CSP on the growth of the human colon cancer cell line, SW480. We demonstrated that transfection with a recombinant plasmid expressing CSP inhibited the proliferation of SW480 in a dose-dependent manner and induced the apoptosis of SW480. A NF-κB gene reporter assay showed that both the CSP and its nuclear localization signal (NLS) motif could equally suppress the activation of NF-κB following the stimulation with human recombinant TNF-α in the SW480. Furthermore, western blot analysis indicated that NLS did not affect the phosphorylation and degradation of IκB, but could sharply inhibit the nuclear translocation of NF-κB in TNF-α stimulated SW480. Hence, our data suggest that the CSP might be explored as a new NF-κB inhibitor for the treatment of colorectal cancer. C1 [Ding, Yan] Third Military Medical University, Department of Pathogenic Biology, 30 Gaotanyan Zhengjie, Shapingba District, 400038 Chongqing, China. [Huang, Xiaobing] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, 400037 Chongqing, China. [Liu, Taiping] Third Military Medical University, Department of Pathogenic Biology, 30 Gaotanyan Zhengjie, Shapingba District, 400038 Chongqing, China. [Fu, Yong] Third Military Medical University, Department of Pathogenic Biology, 30 Gaotanyan Zhengjie, Shapingba District, 400038 Chongqing, China. [Tan, Zhangping] Third Military Medical University, Department of Pathogenic Biology, 30 Gaotanyan Zhengjie, Shapingba District, 400038 Chongqing, China. [Zheng, Hong] Third Military Medical University, Department of Pathogenic Biology, 30 Gaotanyan Zhengjie, Shapingba District, 400038 Chongqing, China. [Zhou, Taoli] Third Military Medical University, Department of Pathogenic Biology, 30 Gaotanyan Zhengjie, Shapingba District, 400038 Chongqing, China. [Dai, Jigang] The Third Military Medical University, Xinqiao Hospital, Department of Thoracic Surgery, 400037 Chongqing, China. [Xu, Wenyue] Third Military Medical University, Department of Pathogenic Biology, 30 Gaotanyan Zhengjie, Shapingba District, 400038 Chongqing, China. RP Dai, J (reprint author), The Third Military Medical University, Xinqiao Hospital, Department of Thoracic Surgery, 400037 Chongqing, China. EM 691057831@qq.com CR Weir HK, Thun MJ, Hankey BF, Ries LA, Howe HL, Wingo PA, Jemal A, Ward E, Anderson RN, Edwards BK, 2003, Annual report to the nation on the status of cancer, 1975–2000, featuring the uses of surveillance data for cancer prevention and control. J Natl Cancer Inst 95:1276–1299 Prat A, Casado E, Cortes J, 2007, New approaches in angiogenic targeting for colorectal cancer. World J Gastroenterol 13:5857– 5866 Rodriguez J, Zarate R, Bandres E, Viudez A, Chopitea A, Garcia- Foncillas J, Gil-Bazo I, 2007, Combining chemotherapy and targeted therapies in metastatic colorectal cancer. World J Gastroenterol 13:5867–5876 Hayden MS, Ghosh S, 2004, Signaling to NF-kappaB. Genes Dev 18:2195–2224 Maeda S, Omata M, 2008, Inflammation and cancer: role of nuclear factor-kappaB activation. Cancer Sci 99:836–842 Karin M, Greten FR, 2005, NF-kappaB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol 5:749–759 Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, Kasem S, Gutkovich-Pyest E, Urieli-Shoval S, Galun E, Ben Neriah Y, 2004, NF-kappaB functions as a tumour promoter in inflammationassociated cancer. Nature 431:461–466 Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, Kagnoff MF, Karin M, 2004, IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 118:285–296 Langowski JL, Zhang X, Wu L, Mattson JD, Chen T, Smith K, Basham B, McClanahan T, Kastelein RA, Oft M, 2006, IL-23 promotes tumour incidence and growth. Nature 442:461–465 Lewis CE, Pollard JW, 2006, Distinct role of macrophages in different tumor microenvironments. Cancer Res 66:605–612 Qiao L, Zhang H, Yu J, Francisco R, Dent P, Ebert MP, Rocken C, Farrell G, 2006, Constitutive activation of NF-kappaB in human hepatocellular carcinoma: evidence of a cytoprotective role. Hum Gene Ther 17:280–290 Nakshatri H, Bhat-Nakshatri P, Martin DA, Goulet RJ Jr, Sledge GW Jr, 1997, Constitutive activation of NF-kappaB during progression of breast cancer to hormone-independent growth. Mol Cell Biol 17:3629–3639 Lindholm PF, Bub J, Kaul S, Shidham VB, Kajdacsy-Balla A, 2000, The role of constitutive NF-kappaB activity in PC-3 human prostate cancer cell invasive behavior. Clin Exp Metastasis 18:471–479 WangW, Abbruzzese JL, Evans DB, Larry L, Cleary KR, Chiao PJ, 1999, The nuclear factor-kappa B RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells. Clin Cancer Res 5:119–127 Sakamoto K, Maeda S, Hikiba Y, Nakagawa H, Hayakawa Y, Shibata W, Yanai A, Ogura K, Omata M, 2009, Constitutive NFkappaB activation in colorectal carcinoma plays a key role in angiogenesis, promoting tumor growth. Clin Cancer Res 15:2248– 2258 Hagemann T, Wilson J, Kulbe H, Li NF, Leinster DA, Charles K, Klemm F, Pukrop T, Binder C, Balkwill FR, 2005, Macrophages induce invasiveness of epithelial cancer cells via NF-kappa B and JNK. J Immunol 175:1197–1205 DeBusk LM, Massion PP, Lin PC, 2008, IkappaB kinase-alpha regulates endothelial cell motility and tumor angiogenesis. Cancer Res 68:10223–10228 Stathopoulos GT, Sherrill TP, Han W, Sadikot RT, Yull FE, Blackwell TS, Fingleton B, 2008, Host nuclear factor-kappaB activation potentiates lung cancer metastasis. Mol Cancer Res 6:364–371 Luo JL, Maeda S, Hsu LC, Yagita H, Karin M, 2004, Inhibition of NF-kappaB in cancer cells converts inflammation- induced tumor growth mediated by TNFalpha to TRAIL-mediated tumor regression. Cancer Cell 6:297–305 Yang J, Pan WH, Clawson GA, Richmond A, 2007, Systemic targeting inhibitor of kappaB kinase inhibits melanoma tumor growth. Cancer Res 67:3127–3134 Scaife CL, Kuang J, Wills JC, Trowbridge DB, Gray P, Manning BM, Eichwald EJ, Daynes RA, Kuwada SK, 2002, Nuclear factor kappaB inhibitors induce adhesion-dependent colon cancer apoptosis: implications for metastasis. Cancer Res 62:6870–6878 Kunnumakkara AB, Guha S, Krishnan S, Diagaradjane P, Gelovani J, Aggarwal BB, 2007, Curcumin potentiates antitumor activity of gemcitabine in an orthotopic model of pancreatic cancer through suppression of proliferation, angiogenesis, and inhibition of nuclear factor-kappaB-regulated gene products. Cancer Res 67:3853–3861 Sandur SK, Deorukhkar A, Pandey MK, Pabon AM, Shentu S, Guha S, Aggarwal BB, Krishnan S, 2009, Curcumin modulates the radiosensitivity of colorectal cancer cells by suppressing constitutive and inducible NF-kappaB activity. Int J Radiat Oncol Biol Phys 75:534–542 Kunnumakkara AB, Diagaradjane P, Guha S, Deorukhkar A, Shentu S, Aggarwal BB, Krishnan S, 2008, Curcumin sensitizes human colorectal cancer xenografts in nudemice to gamma-radiation by targeting nuclear factor-kappaB-regulated gene products. Clin Cancer Res 14:2128–2136 Aggarwal BB, Shishodia S, Takada Y, Banerjee S, Newman RA, Bueso-Ramos CE, Price JE, 2005, Curcumin suppresses the paclitaxel-induced nuclear factor-kappaB pathway in breast cancer cells and inhibits lung metastasis of human breast cancer in nude mice. Clin Cancer Res 11:7490–7498 Baud V, Karin M, 2009, Is NF-kappaB a good target for cancer therapy? Hopes and pitfalls. Nat Rev Drug Discov 8:33–40 Umezawa K, 2006, Inhibition of tumor growth by NF-kappaB inhibitors. Cancer Sci 97:990–995 Bharti AC, Donato N, Singh S, Aggarwal BB, 2003, Curcumin, diferuloylmethane, down-regulates the constitutive activation of nuclear factor-kappa B and IkappaBalpha kinase in human multiple myeloma cells, leading to suppression of proliferation and induction of apoptosis. Blood 101:1053–1062 Schon M, Wienrich BG, Kneitz S, Sennefelder H, Amschler K, Vohringer V, Weber O, Stiewe T, Ziegelbauer K, Schon MP, 2008, KINK-1, a novel small-molecule inhibitor of IKKbeta, and the susceptibility of melanoma cells to antitumoral treatment. J Natl Cancer Inst 100:862–875 May MJ, D’ Acquisto F, Madge LA, Glockner J, Pober JS, Ghosh S, 2000, Selective inhibition of NF-kappaB activation by a peptide that blocks the interaction of NEMO with the IkappaB kinase complex. Science 289:1550–1554 Singh AP, Buscaglia CA, Wang Q, Levay A, Nussenzweig DR, Walker JR, Winzeler EA, Fujii H, Fontoura BM, Nussenzweig V, 2007, Plasmodium circumsporozoite protein promotes the development of the liver stages of the parasite. Cell 131:492–504 Uzunoglu S, Karaca B, Atmaca H, Kisim A, Sezgin C, Karabulut B, Uslu R, 2010, Comparison of XTT and Alamar blue assays in the assessment of the viability of various human cancer cell lines by AT-101, −/− gossypol). Toxicol Mech Methods 20:482–486 Kappe SH, Buscaglia CA, Nussenzweig V, 2004, Plasmodium sporozoitemolecular cell biology. Annu Rev Cell Dev Biol 20:29–59 Kalderon D, Roberts BL, Richardson WD, Smith AE, 1984, A short amino acid sequence able to specify nuclear location. Cell 39:499–509 Kumar KA, Sano G, Boscardin S, Nussenzweig RS, Nussenzweig MC, Zavala F, Nussenzweig V, 2006, The circumsporozoite protein is an immunodominant protective antigen in irradiated sporozoites. Nature 444:937–940 Karin M, 2006, Nuclear factor-kappaB in cancer development and progression. Nature 441:431–436 Frevert U, Galinski MR, Hugel FU, Allon N, Schreier H, Smulevitch S, Shakibaei M, Clavijo P, 1998, Malaria circumsporozoite protein inhibits protein synthesis in mammalian cells. EMBO J 17:3816–3826 LoTempio MM, Veena MS, Steele HL, Ramamurthy B, Ramalingam TS, Cohen AN, Chakrabarti R, Srivatsan ES, Wang MB, 2005, Curcumin suppresses growth of head and neck squamous cell carcinoma. Clin Cancer Res 11:6994–7002 Matsumoto G, Namekawa J, Muta M, Nakamura T, Bando H, Tohyama K, Toi M, Umezawa K, 2005, Targeting of nuclear factor kappaB Pathways by dehydroxymethylepoxyquinomicin, a novel inhibitor of breast carcinomas: antitumor and antiangiogenic potential in vivo. Clin Cancer Res 11:1287– 1293 Tatetsu H, Okuno Y, Nakamura M, Matsuno F, Sonoki T, Taniguchi I, Uneda S, Umezawa K, Mitsuya H, Hata H, 2005, Dehydroxymethylepoxyquinomicin, a novel nuclear factorkappaB inhibitor, induces apoptosis in multiple myeloma cells in an IkappaBalpha-independent manner. Mol Cancer Ther 4:1114– 1120 Starenki DV, Namba H, Saenko VA, Ohtsuru A, Maeda S, Umezawa K, Yamashita S, 2004, Induction of thyroid cancer cell apoptosis by a novel nuclear factor kappaB inhibitor, dehydroxymethylepoxyquinomicin. Clin Cancer Res 10:6821–6829 Moschos SJ, Chaudhary PM, Kirkwood JM, 2008, Resolving “kinks” of chemotherapy in melanoma. J Natl Cancer Inst 100:833–835 Bilsland AE, Anderson CJ, Fletcher-Monaghan AJ, McGregor F, Evans TR, Ganly I, Knox RJ, Plumb JA, Keith WN, 2003, Selective ablation of human cancer cells by telomerase-specific adenoviral suicide gene therapy vectors expressing bacterial nitroreductase. Oncogene 22:370–380 Wirth T, Zender L, Schulte B, Mundt B, Plentz R, Rudolph KL, Manns M, Kubicka S, Kuhnel F, 2003, A telomerase-dependent conditionally replicating adenovirus for selective treatment of cancer. Cancer Res 63:3181–3188 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 895 EP 902 DI 10.1007/s12253-012-9519-7 PG 8 ER PT J AU Wang, Y Wang, X Yang, Z Zhu, G Chen, D Meng, Z AF Wang, Yongzhi Wang, Xinghuan Yang, Zhonghua Zhu, Guangbin Chen, Dong Meng, Zhe TI Menthol Inhibits the Proliferation and Motility of Prostate Cancer DU145 Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Menthol; Migration; Proliferation; Prostate cancer; Transient receptor potential (TRP) channel ID Menthol; Migration; Proliferation; Prostate cancer; Transient receptor potential (TRP) channel AB In recent years, the transient receptor potential melastatin member 8 (TRPM8) channel has emerged as a promising prognostic marker and putative therapeutic target in prostate cancer. We have found that forced overexpression of TRPM8 in PC-3 cells can inhibit the cell proliferation and motility probably through the TRPM8 activation. In this study, we aimed to investigate whether activating the TRPM8 channel by its selective agonist menthol can inhibit the proliferation and motility of androgen-independent prostate cancer (AIPC) with remarkable expression of TRPM8. Menthol is a naturally occurring compound, which has been widely used in cosmetics and pharmaceutical products, and also as flavoring in food. DU145 cells are androgenindependent but have a remarkable expression of TRPM8. The demonstration of the existence of TRPM8 and the absence of TRPA1 in DU145 cells provided the foundation for the following experiments, because both TRPM8 and TRPA1 are molecular targets of menthol. The outcome of MTT assay indicated that menthol inhibited the cell growth (p<0.01). Cell cycle distribution and scratch assay analysis revealed that menthol induced cell cycle arrest at the G0/G1 phase (p<0.01). Furthermore, menthol inhibited the migration of DU145 cells by downregulating the focal-adhesion kinase. So it suggests that the activation of the existing TRPM8 channels may serve as a potential and pragmatic treatment for those AIPC with remarkable expression of TRPM8, and menthol is a useful compound for future development as an anticancer agent. C1 [Wang, Yongzhi] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. [Wang, Xinghuan] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. [Yang, Zhonghua] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. [Zhu, Guangbin] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. [Chen, Dong] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. [Meng, Zhe] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. RP Wang, X (reprint author), Wuhan University, Zhongnan Hospital, Department of Urology, 430071 Wuhan, China. EM urologistwxh@gmail.com CR Jemal A, Siegel R, Ward E et al, 2007, Cancer statistics, 2007. CA Cancer J Clin 57(1):43–66 Chen Y, Clegg NJ, Scher HI, 2009, Anti-androgens and androgendepleting therapies in prostate cancer: new agents for an established target. Lancet Oncol 10(10):981–991 Rosenthal SA, Sandler HM, 2010, Treatment strategies for highrisk locally advanced prostate cancer. Nat Rev Urol 7(1):31–38 Taplin ME, 2007, Drug insight: role of the androgen receptor in the development and progression of prostate cancer. Nat Clin Pract Oncol 4(4):236–244 Damber JE, Aus G, 2008, Prostate cancer. Lancet 371(9625):1710– 1721 Legrand G, Humez S, Slomianny C et al, 2001, Ca2+ pools and cell growth. Evidence for sarcoendoplasmic Ca2+-ATPases 2B involvement in human prostate cancer cell growth control. J Biol Chem 276(50):47608–47614 Thebault S, Flourakis M, Vanoverberghe K et al, 2006, Differential role of transient receptor potential channels in Ca2+ entry and proliferation of prostate cancer epithelial cells. Cancer Res 66(4):2038–2047 Vanoverberghe K, Vanden Abeele F, Mariot P et al, 2004, Ca2+ homeostasis and apoptotic resistance of neuroendocrinedifferentiated prostate cancer cells. Cell Death Differ 11(3):321–330 Skryma R, Mariot P, Bourhis XL et al, 2000, Store depletion and store-operated Ca2+ current in human prostate cancer LNCaP cells: involvement in apoptosis. J Physiol 527(Pt 1):71–83 Vanden Abeele F, Skryma R, Shuba Y et al, 2002, Bcl-2- dependent modulation of Ca(2+, homeostasis and store-operated channels in prostate cancer cells. Cancer Cell 1(2):169–179 Vanden Abeele F, Roudbaraki M, Shuba Y, Skryma R, Prevarskaya N, 2003, Store-operated Ca2+ current in prostate cancer epithelial cells. Role of endogenous Ca2+ transporter type 1. J Biol Chem 278(17):15381–15389 Monteith GR, McAndrew D, Faddy HM, Roberts-Thomson SJ, 2007, Calcium and cancer: targeting Ca2+ transport. Nat Rev Cancer 7(7):519–530 Tsavaler L, Shapero MH, Morkowski S, Laus R, 2001, Trp-p8, a novel prostate-specific gene, is up-regulated in prostate cancer and other malignancies and shares high homology with transient receptor potential calcium channel proteins. Cancer Res 61(9):3760– 3769 Henshall SM, Afar DE, Hiller J et al, 2003, Survival analysis of genome-wide gene expression profiles of prostate cancers identifies new prognostic targets of disease relapse. Cancer Res 63, 14):4196–4203 Yang ZH, Wang XH, Wang HP, Hu LQ, 2009, Effects of TRPM8 on the proliferation and motility of prostate cancer PC-3 cells. Asian J Androl 11(2):157–165 Patel T, Ishiuji Y, Yosipovitch G, 2007, Menthol: a refreshing look at this ancient compound. J Am Acad Dermatol 57(5):873–878 McKemy DD, Neuhausser WM, Julius D, 2002, Identification of a cold receptor reveals a general role for TRP channels in thermosensation. Nature 416(6876):52–58 Peier AM, Moqrich A, Hergarden AC et al, 2002, ATRP channel that senses cold stimuli and menthol. Cell 108(5):705–715 Zhang L, 2004, Evidence that TRPM8 is an androgen-dependent Ca2+ channel required for the survival of prostate cancer cells. Cancer Res 64(22):8365–8373 Valero M, Morenilla-Palao C, Belmonte C, Viana F, 2010, Pharmacological and functional properties of TRPM8 channels in prostate tumor cells. Pflugers Arch-Eur J Physiol 461(1):99– 114 Kim S, Nam J, Park E, Kim B, So I, Jeon J, 2009, Menthol regulates TRPM8-independent processes in PC-3 prostate cancer cells. BBA-Mol Basis Dis 1792(1):33–38 Saadoun S, Papadopoulos MC, Hara-Chikuma M, Verkman AS, 2005, Impairment of angiogenesis and cell migration by targeted aquaporin-1 gene disruption. Nature 434(7034):786–792 Story GM, Peier AM, Reeve AJ et al, 2003, ANKTM1, a TRP-like channel expressed in nociceptive neurons, is activated by cold temperatures. Cell 112(6):819–829 Bidaux G, Roudbaraki M, Merle C et al, 2005, Evidence for specific TRPM8 expression in human prostate secretory epithelial cells: functional androgen receptor requirement. Endocr Relat Cancer 12(2):367–382 RIFM, Research Institute for Fragrance Materials, Inc.,, 1975, Mutagenic evaluation of compound FDA 71–57, menthol. NTIS PB-245-444, FDA 71–268). Unpublishee report from Food and Drug Administration, 14 January. Report Number 5713, RIFM, Woodcliff Lake, NJ, USA Bernson VSM, Pettersson B, 1983, The toxicity of menthol in short-term bioassays. Chem-Biol Interactions 46:233–246 Yamamura H, Ugawa S, Ueda T, Morita A, Shimada S, 2008, TRPM8 activation suppresses cellular viability in human melanoma. Am J Physiol Cell Physiol 295:C296–C301 Lapenna S, Giordano A, 2009, Cell cycle kinases as therapeutic targets for cancer. Nat Rev Drug Discov 8(7):547–566 Pines J, 1995, Cyclins and cyclin-dependent kinases: a biochemical view. Biochem J 308(Pt 3):697–711 Brooke GN, Bevan CL, 2009, The role of androgen receptor mutations in prostate cancer progression. Curr Genomics 10, 1):18–25 Schlaepfer DD, Mitra SK, Ilic D, 2004, Control of motile and invasive cell phenotypes by focal adhesion kinase. Biochim Biophys Acta 1692(2–3):77–102 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 903 EP 910 DI 10.1007/s12253-012-9520-1 PG 8 ER PT J AU Zhang, Y Deng, Zsh Liao, Mm Wang, N Zhang, Xq Yu, Hy Zhang, Yd AF Zhang, Yang Deng, Zhan-sheng Liao, Ming-mei Wang, Ning Zhang, Xiao-qing Yu, Hai-yang Zhang, Yang-de TI Tumor Associated Glycoprotein-72 is a Novel Marker for Poor Survival in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Tumor associated glycoprotein-72; Clinicopathology; Prognosis; Disease-free survival; Overall survival ID Hepatocellular carcinoma; Tumor associated glycoprotein-72; Clinicopathology; Prognosis; Disease-free survival; Overall survival AB To investigate the relationship of tumor associated glycoprotein-72 (TAG-72) expression with clinicopathological features in hepatocellular carcinoma (HCC) patients. Sixty pairs of HCC and paracarcinomatous (PCLT) tissues, and 10 nomral liver (NL) tissues were collected for Western blot analysis, and 244 pairs of HCC and PCLT tissues were collected for immunohistochemistry analysis. TAG-72 protein expression was elevated significantly in HCC tissues compared with PCLT and NL tissues. Its increased expression was correlated with TNM stage, Edmondson-Steiner grade, vein invasion and multiple tumor nodes. It is noteworthy that the HCC patients with high TAG-72 expression had shorter overall survival and disease-free survival than the patients with low expression. Multivariate Cox regression analysis revealed that TAG-72 expression was an independent prognostic factor for HCC patients. The current study demonstrated for the first time that the increased expression of TAG-72 was correlated with poor survival in patients with HCC, indicating that TAG-72 is a novel prognostic marker for HCC. C1 [Zhang, Yang] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan Province, China. [Deng, Zhan-sheng] Central South University, The First Xiangya Hospital, Department of Orthopedics, 410008 Changsha, Hunan Province, China. [Liao, Ming-mei] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan Province, China. [Wang, Ning] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan Province, China. [Zhang, Xiao-qing] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan Province, China. [Yu, Hai-yang] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan Province, China. [Zhang, Yang-de] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, Hunan Province, China. RP Zhang, Yd (reprint author), Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, 410008 Changsha, China. EM zhangyd99@sina.cn CR Zhang MF, Zhang ZY, Fu J, Yang YF, Yun JP, 2009, Correlation between expression of p53, p21/WAF1, and MDM2 proteins and their prognostic significance in primary hepatocellular carcinoma. J Transl Med 7:110 Teo EK, Fock KM, 2001, Hepatocellular carcinoma: an Asian perspective. Dig Dis 19:263–268 Leoni S, Piscaglia F, Righini R, Bolondi L, 2006, Management of small hepatocellular carcinoma. Acta Gastroenterol Belg 69:230– 235 Jin B, Wang X, Jin Y et al, 2009, Detection of serum gastric cancer-associated MG7-Ag from gastric cancer patients using a sensitive and convenient ELISA method. Cancer Invest 27:227– 233 Molina R, Auge JM, Escudero JM et al, 2008, Mucins CA 125, CA 19.9, CA 15.3 and TAG-72.3 as tumor markers in patients with lung cancer: comparison with CYFRA 21–1, CEA, SCC and NSE. Tumour Biol 29:371–380 Chen L, Wang Y, Liu X et al, 2008, A new TAG-72 cancer marker peptide identified by phage display. Cancer Lett 272:122–132 Kim SJ, Hong HJ, 2007, Guided selection of human antibody light chains against TAG-72 using a phage display chain shuffling approach. J Microbiol 45:572–577 Chauhan SC, Vinayek N, Maher DM et al, 2007, Combined staining of TAG-72, MUC1, and CA125 improves labeling sensitivity in ovarian cancer: antigens for multi-targeted antibodyguided therapy. J Histochem Cytochem 55:867–875 Mohsin H, Jia F, Sivaguru G, Hudson MJ et al, 2006, Radiolanthanide-labeled monoclonal antibody CC49 for radioimmunotherapy of cancer: biological comparison of DOTA conjugates and 149Pm, 166Ho, and 177Lu. Bioconjug Chem 17:485– 492 Santos-Juanes J, Bernaldo de Quiros JF, Galache Osuna C et al, 2004, Apocrine carcinoma, adenopathies, and raised TAG-72 serum tumor marker. Dermatol Surg 30:566–569 Milenic DE, Brady ED, Garmestani K, Albert PS, Abdulla A, Brechbiel MW, 2010, Improved efficacy of alpha-particletargeted radiation therapy: dual targeting of human epidermal growth factor receptor-2 and tumor-associated glycoprotein 72. Cancer 116:1059–1066 Zou P, Xu S, Povoski SP et al, 2009, Near-infrared fluorescence labeled anti-TAG-72 monoclonal antibodies for tumor imaging in colorectal cancer xenograft mice. Mol Pharm 6:428–440 Oei AL, Sweep FC, Thomas CM, Boerman OC, Massuger LF, 2008, The use of monoclonal antibodies for the treatment of epithelial ovarian cancer. Int J Oncol 32:1145–1157 Ponnusamy MP, Venkatraman G, Singh AP et al, 2007, Expression of TAG-72 in ovarian cancer and its correlation with tumor stage and patient prognosis. Cancer Lett 251:247–257 Ouyang M, Wu W, Zou Y, Zhou J, Wang Z, Wan X, 2010, Immunoreactivity and prognostic value of tumor-associated glycoprotein 72 in primary gallbladder carcinoma. Surg Oncol 19:82– 87 Graves SS, Dearstyne E, Lin Y et al, 2003, Combination therapy with Pretarget CC49 radioimmunotherapy and gemcitabine prolongs tumor doubling time in a murine xenograft model of colon cancer more effectively than either monotherapy. Clin Cancer Res 9:3712–3721 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 911 EP 916 DI 10.1007/s12253-012-9521-0 PG 6 ER PT J AU Rekhi, B Sharique, A Basak, R Qureshi, SS Desai, SS Ramadwar, M Desai, BS Kurkure, P Jambhekar, AN AF Rekhi, Bharat Sharique, Ahmed Basak, Ranjan Qureshi, S Sajid Desai, S Saral Ramadwar, Mukta Desai, B Sangeeta Kurkure, Purna Jambhekar, A Nirmala TI Desmoplastic Small Round Cell Tumor-Clinicopathological Spectrum, Including Unusual Features and Immunohistochemical Analysis of 45 Tumors Diagnosed at a Tertiary Cancer Referral Centre, with Molecular Results t(11; 22) (p13; q12) (EWS-WT1) in Select Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Desmoplastic small round cell tumor; WT1; Roundcell tumors; Molecular analysis of synovialsarcomas; Immunohistochemistry of synovial sarcoma ID Desmoplastic small round cell tumor; WT1; Roundcell tumors; Molecular analysis of synovialsarcomas; Immunohistochemistry of synovial sarcoma C1 [Rekhi, Bharat] Tata Memorial Hospital, Department of Pathology, Dr E.B. Road, Parel, 400012 Mumbai, Maharashtra, India. [Sharique, Ahmed] Tata Memorial Hospital, Department of Pathology, Dr E.B. Road, Parel, 400012 Mumbai, Maharashtra, India. [Basak, Ranjan] Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Division of Molecular PathologyNavi Mumbai, Khargar, India. [Qureshi, S Sajid] Tata Memorial Hospital, Pediatric Surgical Oncology Services, Parel, 400012 Mumbai, Maharashtra, India. [Desai, S Saral] Tata Memorial Hospital, Department of Pathology, Dr E.B. Road, Parel, 400012 Mumbai, Maharashtra, India. [Ramadwar, Mukta] Tata Memorial Hospital, Department of Pathology, Dr E.B. Road, Parel, 400012 Mumbai, Maharashtra, India. [Desai, B Sangeeta] Tata Memorial Hospital, Department of Pathology, Dr E.B. Road, Parel, 400012 Mumbai, Maharashtra, India. [Kurkure, Purna] Tata Memorial Hospital, Department of Medical Oncology, Parel, 400012 Mumbai, Maharashtra, India. [Jambhekar, A Nirmala] Tata Memorial Hospital, Department of Pathology, Dr E.B. Road, Parel, 400012 Mumbai, Maharashtra, India. RP Rekhi, B (reprint author), Tata Memorial Hospital, Department of Pathology, 400012 Mumbai, India. EM rekhi.bharat@gmail.com CR Antonescu CR, Gerald W, 2002, Desmoplastic small round cell tumor. In: Fletcher CDM, Unni K, Mertens F, eds, Tumors of soft tissue and bone. Pathology and genetics. World Health Organization classification of tumors. IARC, Lyon, pp 216–219 Sestenhenn I, Davis CJ, Mostofi FK, 1987, Undifferentiated malignant epithelial tumors involving serosal surfaces of scrotum and abdomen in young males. J Urol 137(Suppl):214A Gerald WL, Rosai J, 1989, Case 2: desmoplastic small round cell tumor with divergent differentiation. Pediatr Pathol 9:177–183 Gonzalez-Crussi F, Crawford SE, Sun CC, 1990, Intra-abdominal desmoplastic small-cell tumors with divergent differentiation. Observations on three cases of childhood. Am J Surg Pathol 14:633–642 Gerald WL, Miller HK, Battifora H, Miettinen M, Silva EG, Rosai J, 1991, Intra-abdominal desmoplastic small round-cell tumor: report of 19 cases of a distinctive type of high-grade polyphenotypic malignancy affecting young individuals. Am J Surg Pathol 15:499–513 Sawyer JR, Tryka AF, Lewis JM, 1992, A novel reciprocal chromosome translocation t(11;22)(p13;q12, in an intra-abdominal desmoplastic small round-cell tumor. Am J Surg Pathol 16:411–416 Ordonez NG, el-Naggar AK, Ro JY, Silva EG, Mackay B, 1993, Intra-abdominal desmoplastic small cell tumor: a light microscopic, immunocytochemical, ultrastructural, and flow cytometric study. Hum Pathol 24:850–865 Dorsey BV, Benjamin LE, Rauscher F 3rd, Klencke B, Venook AP, Warren RS, Weidner N, 1996, Intra-abdominal desmoplastic small round-cell tumor: expansion of the pathologic profile. Mod Pathol 9:703–709 Amato RJ, Ellerhorst JA, Ayala AG, 1996, Intra-abdominal desmoplastic small cell tumor. Report and discussion of five cases. Cancer 78:845–851 Cummings OW, Ulbright TM, Young RH, Dei Tos AP, Fletcher CD, Hull MT, 1997, Desmoplastic small round cell tumors of the paratesticular region: a report of six cases. Am J Surg Pathol 21:219–225 Ordonez NG, 1998, Desmoplastic small round cell tumor: I: a histopathologic study of 39 cases with emphasis on unusual histological patterns. Am J Pathol 22(1303–13):1314–1327 Gerald WL, Ladanyi M, de Alava E, Cuatrecasas M, Kushner BH, LaQuaglia MP, Rosai J, 1998, Clinical, pathologic, and molecular spectrum of tumors associated with, 11;22)(p13;q12): desmoplastic small round-cell tumor and its variants. J Clin Oncol 16:3028– 3036 Lae ME, Roche PC, Jin L, Lloyd RV, Nascimento AG, 2002, Desmoplastic small round cell tumor: a clinicopathologic, immunohistochemical, and molecular study of 32 tumors. Am J Surg Pathol 26:823–835 Cao L, Ni J, Que N, Wu Z, Song Z, 2008, Desmoplastic small round cell tumor: a clinical, pathological, and immunohistochemical study of 18 Chinese cases. Int J Surg Pathol 16:257–262 Slomovitz BM, Giraotra M, Aledo A, Saqi A, Soslow RA, Spigland NA, Caputo TA, 2000, Desmoplastic small round cell tumor with primary ovarian involvement: case report and review. Gynecol Oncol 79:124–128 Rekhi B, Basak R, Desai S, Jambhekar NA, 2010, A t, 11; 22,, p13; q12, EWS-WT-1 positive desmoplastic small round cell tumor, DSRCT, of the maxilla- An unusual case indicating role of molecular diagnosis in round cell sarcomas. J Post Grad Med 56:206–210 Srivastava A, Rosenberg AE, Selig M, Rubin BP, Nielsen GP, 2005, Keratin-positive Ewing’s sarcoma: an ultrastructural study of 12 cases. Int J Surg Pathol 13:43–50 Kohashi K, Oda Y, Yamamoto H, Tamiya S, Oshiro Y, Izumi T, Taguchi T, Tsuneyoshi M, 2008, SMARCB1/INI1 protein expression in round cell soft tissue sarcomas associated with chromosomal translocations involving EWS: a special reference to SMARCB1/INI1 negative variant extraskeletal myxoid chondrosarcoma. Am J Surg Pathol 32:1168–1174 Nagel H, Hemmerlein B, Ruschenburg I, Huppe K, Droese M, 1998, The value of anti-calretinin antibody in the differential diagnosis of normal and reactive mesothelia versus metastatic tumors in effusion cytology. Pathol Res Pract 194:759–764 Engohan-Aloghe C, Aubain Sommerhausen Nde S, Noel JC, 2009, Ovarian involvement by desmoplastic small round cell tumor with leydig cell hyperplasia showing an unusual immunophenotype, cytokeratin negative, calretinin and inhibin positive, mimicking poorly differentiated sertoli leydig cell tumor. Int J Gynecol Pathol 28:579–583 Gerald WL, Rosai J, Ladanyi M, 1995, Characterization of the genomic breakpoint and chimeric transcripts in the EWS-WT1 gene fusion of desmoplastic small round cell tumor. Proc Natl Acad Sci U SA 92:1028–1032 Ladanyi M, Gerald W, 1994, Fusion of the EWS and WT1 genes in the Desmoplastic small round cell tumor. Cancer Res 54:2837– 2840 de Alava E, Ladanyi M, Rosai J, Gerald WL, 1995, Detection of chimeric transcripts in desmoplastic small round cell tumor and related developmental tumors by reverse transcriptase polymerase chain reaction. A specific diagnostic assay. Am J Pathol 147:1584– 1591 Schwarz RE, Gerald WL, Kushner BH, Coit DG, Brennan MF, La Quaglia MP, 1998, Desmoplastic small round cell tumors: prognostic indicators and results of surgical management. Ann Surg Oncol 5:416–422 Mazuryk M, Paterson AH, Temple W, Arthur K, Crabtree T, Stewart DA, 1998, Benefit of aggressive multimodality therapy with autologous stem cell support for intra-abdominal desmoplastic small round cell tumor. Bone Marrow Transplant 21:961–963 Hiralal GS, Thulkar S, Rao SK, 2007, Desmoplastic small round cell tumor of abdomen. Singapore Med J 48:e19 Chouli M, Viala J, Dromain C, Fizazi K, Duvillard P, Vanel D, 2005, Intra-abdominal desmoplastic small round cell tumors: CT findings and clinicopathological correlations in 13 cases. Eur J Radiol 54:438–442 Hayes-Jordan A, Anderson PM, 2011, The diagnosis and management of desmoplastic small round cell tumor: a review. Current Opinion Oncol 23:385–389 Hayes-Jordan A, Green H, Fitzgerald N, Xiao L, Anderson P, 2010, Novel treatment for desmoplastic small round cell tumor: hyperthermic intraperitoneal perfusion. J Paediatr Surg 45:1000–1006 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 917 EP 927 DI 10.1007/s12253-012-9522-z PG 11 ER PT J AU Hassumi-Fukasawa, KM Miranda-Camargo, AF Zanetti, RB Galano, FD Ribeiro-Silva, A Soares, GE AF Hassumi-Fukasawa, Kazue Marcela Miranda-Camargo, Alves Fabiana Zanetti, Riedo Bruna Galano, Faria Denise Ribeiro-Silva, Alfredo Soares, Garcia Edson TI Expression of BAG-1 and PARP-1 in Precursor Lesions and Invasive Cervical Cancer Associated with Human Papillomavirus (HPV) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BAG-1; PARP-1; HPV; Cervix; PCR; Immunohistochemistry ID BAG-1; PARP-1; HPV; Cervix; PCR; Immunohistochemistry AB Cervical cancer remains persistently the second most common malignancies among women worldwide, responsible for 500,000 new cases annually. Only in Brazil, the estimate is for 18,430 new cases in 2011. Several types of molecular markers have been studied in carcinogenesis including proteins associated with apoptosis such as BAG-1 and PARP-1. This study aims to demonstrate the expression of BAG-1 and PARP-1 in patients with low-grade squamous intraepithelial lesions (LSILs), high-grade squamous intraepithelial lesions (HSILs) and invasive squamous cell carcinomas (SCCs) of the uterine cervix and to verify a possible association with HPV infection. Fifty samples of LSILs, 50 samples of HSILs and 50 samples of invasive SCCs of the uterine cervix were analyzed by immunohistochemistry for BAG-1 and PARP-1 expression. PCR was performed to detect and type HPV DNA. BAG-1 expression levels were significantly different between LSILs and HSILs (p00,014) and between LSILs and SCCs (p00,014). In regards to PARP-1 expression, we found significant differences between the expression levels in HSILs and SCCs (p00,022). No association was found between BAG-1 expression and the presence of HPV. However, a significant association was found between PARP-1 expression and HPV positivity in the HSILs group (p00,021). In conclusion our research suggests that BAG-1 expression could contribute to the differentiation between LSIL and HSIL/SCC whereas PARP-1 could be useful to the differentiation between HSIL HPV-related and SCC. Further studies are needed to clarify the molecular aspects of the relationship between PARP-1 expression and HPV infection, with potential applications for cervical cancer prediction. C1 [Hassumi-Fukasawa, Kazue Marcela] University of Sao Paulo, Faculty of Medicine of Ribeirao Preto, Department of Pathology, Avenida dos Bandeirantes, 3900, 14049-900 Ribeirao Preto, Sao Paulo, Brazil. [Miranda-Camargo, Alves Fabiana] University of Sao Paulo, Faculty of Medicine of Ribeirao Preto, Department of Pathology, Avenida dos Bandeirantes, 3900, 14049-900 Ribeirao Preto, Sao Paulo, Brazil. [Zanetti, Riedo Bruna] University of Sao Paulo, Faculty of Medicine of Ribeirao Preto, Department of Pathology, Avenida dos Bandeirantes, 3900, 14049-900 Ribeirao Preto, Sao Paulo, Brazil. [Galano, Faria Denise] University of Sao Paulo, Faculty of Medicine of Ribeirao Preto, Department of Pathology, Avenida dos Bandeirantes, 3900, 14049-900 Ribeirao Preto, Sao Paulo, Brazil. [Ribeiro-Silva, Alfredo] University of Sao Paulo, Faculty of Medicine of Ribeirao Preto, Department of Pathology, Avenida dos Bandeirantes, 3900, 14049-900 Ribeirao Preto, Sao Paulo, Brazil. [Soares, Garcia Edson] University of Sao Paulo, Faculty of Medicine of Ribeirao Preto, Department of Pathology, Avenida dos Bandeirantes, 3900, 14049-900 Ribeirao Preto, Sao Paulo, Brazil. RP Hassumi-Fukasawa, KM (reprint author), University of Sao Paulo, Faculty of Medicine of Ribeirao Preto, Department of Pathology, 14049-900 Ribeirao Preto, Brazil. EM marcelahassumi@usp.br CR Nour NM, 2009, Cervical cancer: a preventable death. Rev Obstet Gynecol 4:240–244 Jayshree RS, Sreenivas A, Tessy M, Krishna S, 2009, Cell intrinsic & extrinsic factors in cervical carcinogenesis. Indian J Med Res 130:286–295 Townsend PA, Cutress RI, Carroll CJ, Lawrence KM, Scarabelli TM, Packham G, Stephanou A, Latchman DS, 2004, BAG-1 proteins protect cardiac myocytes from simulated ischemia/reperfusion- induced apoptosis via an alternate mechanism of cell survival independent of the proteasome. J Biol Chem 20:20723– 20728 Sharp A, Crabb SJ, Townsend PA, Cutress RI, Brimmell M, Wang X, Packham G, 2004, BAG-1 in carcinogenesis. Expert Rev Mol Med 6(7):1–15 Ribeiro-Silva A, 2007, Immunohistochemical features of a papillary squamous cell carcinoma of the endometrium with transitional cell differentiation. Diagn Pathol 2:26 Krajewska M, Turner BC, Shabaik A, Krajewski S, Reed JC, 2006, Expression of BAG-1 protein correlates with aggressive behavior of prostate cancers. Prostate 66(8):801–810 Porichi O, Nikolaidou ME, Apostolaki A, Arnogiannaki N, Papassideri I, Chatonidis I, Tserkezoglou A, Vorgias G, Kassanos D, Panotopoulou E, 2010, Isomorph expression of BAG-1 gene, ER and PR in endometrial cancer. Anticancer Res 30(10):4103–4108 Wood J, Lee SS, Hague A, 2009, Bag-1 proteins in oral squamous cell carcinoma. Oral Oncol 45(2):94–102 Rorke S, Murphy S, Khalifa M, Chernenko G, Tang SC, 2001, Prognostic significance of BAG-1 expression in nonsmall cell lung cancer. Int J Cancer 95(5):317–322 Masutani M, Nakagama H, Sugimura T, 2003, Poly(ADP-ribose, and carcinogenesis. Gene Chromosome Canc 38(4):339–348 Burkle A, 2001, Poly(APD-ribosyl)ation, a DNA damage-driven protein modification and regulator of genomic instability. Cancer Lett 163(1):1–5 Tong WM, Cortes U, Wang ZQ, 2001, Poly(ADP-ribose, polymerase: a guardian angel protecting the genome and suppressing tumorigenesis. Biochim Biophys Acta 1552(1):27–37 Bouchard VJ, Rouleau M, Poirier GG, 2003, PARP-1, a determinant of cell survival in response to DNA damage. Exp Hematol 31, 6):446–454 Koh DW, Dawson TM, Dawson VL, 2005, Mediation of cell death by poly(ADP-ribose, polymerase-1. Pharmacol Res 52(1):5–14 Bieche I, de Murcia G, Lidereau R, 1996, Poly(ADP-ribose, polymerase gene expression status and genomic instability in human breast cancer. Clin Cancer Res 2(7):1163–1167 Soldatenkov VA, Albor A, Patel BK, Dreszer R, Dritschilo A, Notario V, 1999, Regulation of the human poly(ADP-ribose, polymerase promoter by the ETS transcription factor. Oncogene 18(27):3954–3962 Shimizu S, Nomura F, Tomonaga T, Sunaga M, Noda M, Ebara M, Saisho H, 2004, Expression of poly(ADP-ribose, polymerase in human hepatocellular carcinoma and analysis of biopsy specimens obtained under sonographic guidance. Oncol Rep 12(4):821– 825 Ghabreau L, Roux JP, Frappart PO, Mathevet P, Patricot LM, Mokni M, Korbi S, Wang ZQ, Tong WM, Frappart L, 2004, Poly(ADP-ribose, polymerase-1, a novel partner of progesterone receptors in endometrial cancer and its precursors. Int J Cancer 109, 3):317–321 Nosho K, Yamamoto H, Mikami M, Taniguchi H, Takahashi T, Adachi Y, Imamura A, Imai K, Shinomura Y, 2006, Overexpression of poly(ADP-ribose, polymerase-1, PARP-1, in the early stage of colorectal carcinogenesis. Eur J Cancer 42(14):2374– 2381 Csete B, Lengyel Z, Kadar Z, Battyani Z, 2009, Poly(adenosine diphosphate-ribose, polymerase-1 expression in cutaneous malignant melanomas as a new molecular marker of aggressive tumor. Pathol Oncol Res 15(1):47–53 Xie X, Clausen OP, Boysen M, 2004, Bag-1 expression as a prognostic factor in tongue squamous cell carcinomas. Laryngoscope 114(10):1785–1790 Sanguinetti CJ, Dias Neto E, Simpson AJ, 1994, Rapid silver staining and recovery of PCR products separated on polyacrylamide gels. Biotechniques 17:914–921 Nikitakis NG, Sauk JJ, Papanicolaou SI, 2004, The role of apoptosis in oral disease: mechanisms; aberrations in neoplastic, autoimmune, infectious, hematologic, and developmental diseases; and therapeutic opportunities. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 97(4):476–490 Guimaraes MC, Goncalves MA, Soares CP, Bettini JS, Duarte RA, Soares EG, 2005, Immunohistochemical expression of p16INK4a and bcl-2 according to HPV type and to the progression of cervical squamous intraepithelial lesions. J Histochem Cytochem 53, 4):509–516 Cutress RI, Townsend PA, Sharp A, Maison A, Wood L, Lee R, Brimmell M, Mullee MA, Johnson PW, Royle GT, Bateman AC, Packham G, 2003, The nuclear BAG-1 isoform, BAG-1L, enhances oestrogen-dependent transcription. Oncogene, 32):4973–4982 Nadler Y, Camp RL, Giltnane JM, Moeder C, Rimm DL, Kluger HM, Kluger Y, 2008, Expression patterns and prognostic value of Bag-1 and Bcl-2 in breast cancer. Breast Cancer Res 10(2):R35 Sun N, Meng Q, Tian A, 2010, Expressions of the anti-apoptotic genes Bag-1 and Bcl-2 in colon cancer and their relationship. Am J Surg 200(3):341–345 Chang JL, Shyul JS, Lee WH, 2003, The expression of antiapoptotic protein BAG-1 in the squamous neoplastic changes of the uterine cervix. J Med Sci 23(2):105–112 Yang X, Hao Y, Ferenczy A, Tang SC, Pater A, 1999, Overexpression of Anti-apoptotic Gene BAG-1 in Human Cervical Cancer. Exp Cell Res 247:200–207 Takahashi N, Yanagihara M, Ogawa Y, Yamanoha B, Andoh T, 2003, Down-regulation of Bcl-2-interacting protein BAG-1 confers resistance to anti-cancer drugs. Biochem Biophys Res Commun 301, 3):798–803 Masutani M, Nakagama H, Sugimura T, 2005, Poly(ADP-ribosyl, ation in relation to cancer and autoimmune disease. Cell Mol Life Sci 62(7–8):769–783 Staibano S, Pepe S, LoMuzio L, Somma P, Mascolo M, Argenziano G, Scalvenzi M, Salvatore G, Fabbrocini G, Molea G, Bianco AR, Carlomagno C, De Rosa G, 2005, Poly(adenosine diphosphate-ribose, polymerase 1 expression in malignant melanomas from photoexposed areas of the head and neck region. Hum Pathol 36(7):724–731 Lo WY, Lai CC, Hua CH, Tsai MH, Huang SY, Tsai CH, Tsai FJ, 2007, S100A8 is identified as a biomarker of HPV18-infected oral squamous cell carcinomas by suppression subtraction hybridization, clinical proteomics analysis, and immunohistochemistry staining. J Proteome Res 6:2143–2151 Lee D, Jin Woo Kim JW, Kim K, Joel CO, Schreiber V, de Murcia JM, Choe J, 2002, Functional interaction between human papillomavirus type 18 E2 and poly(ADP-ribose, polymerase 1. Oncogene 21:5877–5885 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 929 EP 937 DI 10.1007/s12253-012-9523-y PG 9 ER PT J AU Wu, D Li, H Du, W Ji, X Liu, W Huang, Sh Xiao, Y AF Wu, Dan Li, Huiyu Du, Wen Ji, Xiaoxia Liu, Wei Huang, Shiang Xiao, Yi TI Mathematical Modeling of Therapeutic Strategies for Myeloid Malignancies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hematopoietic stem cells; Cancer stem cell; Mathematical modeling; Chemotherapy ID Hematopoietic stem cells; Cancer stem cell; Mathematical modeling; Chemotherapy AB The existence of malignant stem cells has been proven for hematopoietic disorder as well as some solid tumors. Although significant improvements in cancer therapy have been made, tumor recurrence is frequent and can partly be due to the absence of therapeutic target which tumor stem cells are regarded as. In this paper we shall explore different therapeutic scenarios for successful tumor treatment by using a predictive mathematical model based on the cell compartment method. In particular, we shall study the effects of the chemotherapeutic target rate and of the interval of G-CSF administration on therapy for myeloid malignancies through simulating chemotherapy with G-CSF (granulocyte colony-stimulating factor) support. The results indicate that if target rate is raised to an enough high value, the efficiency of chemotherapy increases so greatly that the tumor mature cells perish completely and normal mature cells are maintained at a normal level. Furthermore, the administration of G-CSF can increase the amount of the normal mature cells to a normal level. However, too long interval of G-CSF administration is demonstrated not propitious to patients’ healing. These results indicate that the simulations may be an effective approach to help designing therapeutic scenarios for successful tumor treatment by chemotherapy. C1 [Wu, Dan] Huazhong University of Science and Technology, Department of PhysicsWuhan, China. [Li, Huiyu] Huazhong University of Science and Technology, Tongji Medical College, Center of Stem cellWuhan, China. [Du, Wen] Huazhong University of Science and Technology, Tongji Medical College, Center of Stem cellWuhan, China. [Ji, Xiaoxia] Huazhong University of Science and Technology, Department of PhysicsWuhan, China. [Liu, Wei] Huazhong University of Science and Technology, Tongji Medical College, Center of Stem cellWuhan, China. [Huang, Shiang] Huazhong University of Science and Technology, Tongji Medical College, Center of Stem cellWuhan, China. [Xiao, Yi] Huazhong University of Science and Technology, Department of PhysicsWuhan, China. RP Xiao, Y (reprint author), Huazhong University of Science and Technology, Department of Physics, Wuhan, China. EM yxiao@mail.hust.edu.cn CR Reya T, Morrison SJ, Clarke MF, Weissman IL, 2001, Stem cells, cancer, and cancer stem cells. Nature 414:105–111 Abkowitz JL, Persik MT, Shelton GH, Ott RL, Kiklevich JV, Catlin SN, Guttorp P, 1995, Behavior of hematopoietic stem cells in a large animal. Proc Natl Acad Sci U S A 92:2031–2035 Nowak MA, Michor F, Iwasa Y, 2003, The linear process of somatic evolution. Proc Natl Acad Sci U S A 100:14966–14969 Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100:57–70 Hope KJ, Jin L, Dick JE, 2004, Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in selfrenewal capacity. Nat Immunol 5:738–743 Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF, Morrison SJ, 2003, Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature 425:962–967 Polakis P, 2000, Wnt signaling and cancer. Genes Dev 14:1837– 1851 Armitage P, Doll R, 1954, The age distribution of cancer and a multi-stage theory of carcinogenesis. Br J Cancer 8:1–12 Michor F, Hughes TP, Iwasa Y, Branford S, Shah NP, Sawyers CL, Nowak MA, 2005, Dynamics of chronic myeloid leukaemia. Nature 435:1267–1270 Komarova NL, Wodarz D, 2005, Drug resistance in cancer: principles of emergence and prevention. Proc Natl Acad Sci U S A 102:9714–9719 Bernard S, Belair J, Mackey MC, 2004, Bifurcations in a whiteblood- cell production model. C R Biol 327:201–210 Pujo-Menjouet L, Mackey MC, 2004, Contribution to the study of periodic chronic myelogenous leukemia. C R Biol 327:235–244 Colijn C, Mackey MC, 2005, A mathematical model of hematopoiesis— I. Periodic chronic myelogenous leukemia. J Theor Biol 237:117–132 Catlin SN, Guttorp P, Abkowitz JL, 2005, The kinetics of clonal dominance in myeloproliferative disorders. Blood 106:2688–2692 Engel C, Scholz M, Loeffler M, 2004, A computational model of human granulopoiesis to simulate the hematotoxic effects of multicycle polychemotherapy. Blood 104:2323–2331 Ostby I, Kvalheim G, Rusten LS, Grottum P, 2004, Mathematical modeling of granulocyte reconstitution after high-dose chemotherapy with stem cell support: effect of post-transplant G-CSF treatment. J Theor Biol 231:69–83 Wichmann HE, Loeffler M, 1985, Mathematical modeling of cell proliferation: stem cell regulation in hemopoiesis. CRC Press, Boca Raton, Fla Ganguly R, Puri IK, 2006, Mathematical model for the cancer stem cell hypothesis. Cell Prolif 39:3–14 Scholz M, Engel C, Loeffler M, 2005, Modelling human granulopoiesis under poly-chemotherapy with G-CSF support. J Math Biol 50:397–439 Dontu G, Al-Hajj M, Abdallah WA, Clarke MF, Wicha MS, 2003, Stem cells in normal breast development and breast cancer. Cell Proliferation 36:59–72 Brown MD, Gilmore PE, Hart CA, Samuel JD, Ramani VA, George NJ, Clarke NW, 2007, Characterization of benign and malignant prostate epithelial Hoechst 33342 side populations. Prostate 67:1384–1396 Alison MR, 2005, Liver stem cells: implications for hepatocarcinogenesis. Stem Cell Rev 1:253–260 Warner JK, Wang JC, Hope KJ, Jin L, Dick JE, 2004, Concepts of human leukemic development. Oncogene 23:7164–7177 Clarke MF, 2004, Neurobiology: At the root of brain cancer. Nature 432:281–282 Huang S, Terstappen LW, 1994, Formation of haematopoietic microenvironment and haematopoietic stem cells from single human bone marrow stem cells. Nature 368:664 Armitage P, Doll R, 1957, A two-stage theory of carcinogenesis in relation to the age distribution of human cancer. Br J Cancer 11:161–169 Michor F, Iwasa Y, Nowak MA, 2004, Dynamics of cancer progression. Nat Rev Cancer 4:197–205 Dick JE, 2003, Breast cancer stem cells revealed. Proc Natl Acad Sci U S A 100:3547–3549 Huang S, Terstappen LW, 1992, Formation of haematopoietic microenvironment and haematopoietic stem cells from single human bone marrow stem cells. Nature 360:745–749 Huang S, Law P, Francis K, Palsson BO, Ho AD, 1999, Symmetry of initial cell divisions among primitive hematopoietic progenitors is independent of ontogenic age and regulatory molecules. Blood 94:2595–2604 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 939 EP 947 DI 10.1007/s12253-012-9524-x PG 9 ER PT J AU Biesaga, B Niemiec, J Ziobro, M AF Biesaga, Beata Niemiec, Joanna Ziobro, Marek TI Microvessel Density and Status of p53 Protein as Potential Prognostic Factors for Adjuvant Anthracycline Chemotherapy in Retrospective Analysis of Early Breast Cancer Patients Group SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Early breast cancer patients; Anthracyclines; Angiogenesis; p53 status; Potential prognostic factors ID Early breast cancer patients; Anthracyclines; Angiogenesis; p53 status; Potential prognostic factors AB A considerable subgroup of patients with early breast cancer does not address benefits of anthracycline based chemotherapy. The aim of this retrospective study was to investigate the effect of microvessel density (MVD) and status of p53 protein on 5-year disease free survival (DFS) in the group of breast cancer patients treated with anthracyclines in adjuvant setting. Correlations between MVD, p53 status and other clinicopathological parameters were also assessed. MVD and p53 status were analyzed immunohistochemically in the group of 172 women with breast cancer in clinical stage T1-2, N1-N2, M0. There were 123 tumors (71.5 %) with lower MVD (≤214.8 microvesells/ mm2) and 49 (28.5 %) with higher MVD (>214.8 microvesells/mm2). The proportion of higher MVD tumors significantly increased in N2 (P00.000) and in estrogen (P00.046) or progesterone receptors (P00.029) negative tumors. p53 positivity was indicated in 50 cancers (29.1 %) and was significantly associated with higher grade (P00.000), steroid receptors negativity (P00.000), cytokeratin5/ 6 positivity (P00.026), topoisomerase IIα overexpression (P00.005) and higher proliferation rate (P00.001). In univariate analysis, higher MVD (P00.016) and p53 negativity (P00.023) were significantly related with longer DFS (median follow-up 36 months). In multivariate Cox regression analysis MVD was independently associated with DFS. These data suggest that higher MVD is favourable prognostic factors for early advanced breast cancer patients after adjuvant anthracycline based chemotherapy. C1 [Biesaga, Beata] Centre of Oncology, Department of Applied Radiobiology, Garncarska 11, 31-115 Krakow, Poland. [Niemiec, Joanna] Centre of Oncology, Department of Applied Radiobiology, Garncarska 11, 31-115 Krakow, Poland. [Ziobro, Marek] Centre of Oncology, Department of Medical Oncology, Garncarska 11, 31-115 Krakow, Poland. RP Biesaga, B (reprint author), Centre of Oncology, Department of Applied Radiobiology, 31-115 Krakow, Poland. EM z5biesag@cyfronet.pl CR Early Breast Cancer Trialists' Collaborative Group, EBCTCG,, 2005, Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet 365(9472):1687–717 Andre F, Pusztai L, 2006, Heterogeneity of breast cancer among patients and implications for patient selection for adjuvant chemotherapy. Pharm Res 23(9):1951–8 Biesaga B, Niemiec J, Ziobro M, Wysocka J, Kruczak A, 2011, Prognostic potential of topoisomerase IIα and HER2 in a retrospective analysis of early advanced breast cancer patients treated with adjuvant anthracycline chemotherapy. Breast 20(4):338–350 Weidner N, Semple JP, Welch WR, Folkman J, 1991, Tumor angiogenesis and metastasis correlation with invasive breast carcinoma. N Engl J Med 324:1–8 Gasparini G, Weidner N, Bevilacqua P, Maluta S, Dalla Palma P, Caffo O, Barbareschi M, Boracchi P, Marubini E, Pozza F, 1994, Tumor microvessel density, expression, tumor size, and peritumoral lymphatic vessel invasion are relevant prognostic markers in nodenegative breast carcinoma. J Clin Oncol 12(3):454–66 Heimann R, Ferguson D, Powers C, Recant WM, Weichselbaum RR, Hellman S, 1996, Angiogenesis as a predictor of long-term survival for patients with node-negative breast cancer. J Natl Cancer Inst 88(23):1764–69 Uzzan B, Nicolas P, Cucherat M, Perret GY, 2004, Microvessel density as a prognostic factor in women with breast cancer: a systematic review of the literature and meta-analysis. Cancer Res 64(9):2941–55 Nieto Y, Woods J, Nawaz F, Baron A, Jones RB, Shpall EJ, Nawaz S, 2007, Prognostic analysis of tumor angiogenesis, determined by microvessel density and expression of vascular endothelial growth factor, in high-risk primary breast cancer patients treated with high-dose chemotherapy. Br J Cancer 97(3):391–7 Tas F, Yavuz E, Aydiner A, Saip P, Disci R, Iplikci A, 2000, Angiogenesis and p53 protein expression in breast cancer: prognostic roles and interrelationships. Am J Clin Oncol 23(6):546–53 Viens P, Jacquemier J, Bardou VJ et al, 1999, Association of angiogenesis and poor prognosis in node-positive patients receiving anthracycline-based adjuvant chemotherapy. Breast Cancer Res Treat 54(3):205–12 Gluz O,Wild P, Liedtke C, Kates R et al, 2011, Tumor angiogenesis as prognostic and predictive marker for chemotherapy doseintensification efficacy in high-risk breast cancer patients within the WSG AM-01 trial. Breast Cancer Res Treat 126(3):643–51 Kanjanapanjapol S, Wongwaisayawan S, Phuwapraisirisan S, Wilasrusmee C, 2007, Prognostic significance of microvessel density in breast cancer of Thai women. JMed Assoc Thai 90(2):282–90 Hansen S, Sorensen FB, Vach W, Grabau DA, Bak M, Rose C, 2004, Microvessel density compared with the Chalkley count in a prognostic study of angiogenesis in breast cancer patients. Histopathology 44(5):428–36 Ludovini V, Sidoni A, Pistola L et al, 2003, Evaluation of the prognostic role of vascular endothelial growth factor and microvessel density in stages I and II breast cancer patients. Breast Cancer Res Treat 81(2):159–68 Guidi AJ, Berry DA, Broadwater G, Helmchen B, Bleiweiss IJ, Budman DR, Henderson IC,Norton L,Hayes DF, 2002, Association of angiogenesis and disease outcome in node-positive breast cancer patients treated with adjuvant cyclophosphamide, doxorubicin, and fluorouracil: a Cancer and Leukemia Group B correlative science study from protocols 8541/8869. J Clin Oncol 20(3):732–42 Ioachim E, Charchanti A, Charalabopoulos K, Tsanou H, Briasoulis E, Karavasilis V, Pavlidis N, Agnantis NJ, 2002, The prognostic evaluation of tumor angiogenesis in invasive breast carcinoma. Electronic Journal of Pathology and Histology 8.1 021–02 Protopapa E, Delides GS, Revesz L, 1993, Vascular density and the response of breast carcinomas to mastectomy and adjuvant chemotherapy. Eur J Cancer 29A(10):1391–3 Gunel N, Akcali Z, Coskun U, Akyol G, Yamac D, Yenidunya S, 2002, Prognostic importance of tumor angiogenesis in breast carcinoma with adjuvant chemotherapy. Pathol Res Pract 198(1):7–12 Fedier A, Moawad A, Haller U, Fink D, 2003, p53-deficient cells display increased sensitivity to anthracyclines after loss of the catalytic subunit of the DNA-dependent protein kinase. Int J Oncol 23(5):1431–7 Lowe SW, Bodis S, McClatchey A, Remington L, Ruley HE, Fisher DE, Housman DE, Jacks T, 1994, p53 status and the efficacy of cancer therapy in vivo. Science 266(5186):807–10 Malamou-Mitsi V, Gogas H, Dafni U et al, 2006, Evaluation of the prognostic and predictive value of p53 and Bcl-2 in breast cancer patients participating in a randomized study with dose-dense sequential adjuvant chemotherapy. Ann Oncol 17(10):1504–11 Faneyte IF, Peterse JL, Van Tinteren H et al, 2004, Predicting early failure after adjuvant chemotherapy in high-risk breast cancer patients with extensive lymph node involvement. Clin Cancer Res 10(13):4457–63 Mottolese M, Benevolo M, Del Monte G, Buglioni S, Papaldo P, Nistico C, Di Filippo F, Vasselli S, Vici P, Botti C, 2000, Role of P53 and BCL-2 in high-risk breast cancer patients treated with adjuvant anthracycline-based chemotherapy. J Cancer Res Clin Oncol 126(12):722–29 Thor AD, Berry DA, Budman DR et al, 1998, erbB-2, p53, and efficacy of adjuvant therapy in lymph node-positive breast cancer. J Natl Cancer Inst 90(18):1346–60 Chrisanthar R, Knappskog S, Lokkevik E et al, 2011, Predictive and prognostic impact of TP53 mutations and MDM2 promoter genotype in primary breast cancer patients treated with epirubicin or paclitaxel. PLoS One 6(4):e19249 Geisler S, Lonning PE, Aas T, Johnsen H, Fluge O, Haugen DF, Lillehaug JR, Akslen LA, Borresen-Dale AL, 2001, Influence of TP53 gene alterations and c-erbB-2 expression on the response to treatment with doxorubicin in locally advanced breast cancer. Cancer Res 61(6):2505–12 Niskanen E, Blomqvist C, Franssila K, Hietanen P, Wasenius VM, 1997, Predictive value of c-erbB-2, p53, cathepsin-D and histology of the primary tumor in metastatic breast cancer. Br J Cancer 76, 7):917–22 Kaplan EL, Meier P, 1958, Nonparametric estimation from incomplete observations. J Am Statl Association 53:457–81 Chien AJ, Moasser MM, 2008, Cellular mechanisms of resistance to anthracyclines and taxanes in cancer: intrinsic and acquired. Semin Oncol 35(2 Suppl 2):S1–S14 Khromova NV, Kopnin PB, Stepanova EV, Agapova LS, Kopnin BP, 2009, p53 hot-spot mutants increase tumor vascularization via ROS-mediated activation of the HIF1/VEGF-A pathway. Cancer Lett 276(2):143–51 Ushio-Fukai M, 2006, Redox signaling in angiogenesis: Role of NADPH oxidase. Cardiovascular Res 71:226–35 Rykala J, Przybylowska K, Majsterek I, Pasz-Walczak G, Sygut A, Dziki A, Kruk-Jeromin J, 2011, Angiogenesis markers quantification in breast cancer and their correlation with clinicopathological prognostic variables. Pathol Oncol Res 17(4):809–17 Martin L, Green B, Renshaw C, Lowe D, Rudland P, Leinster SJ, Winstanley J, 1997, Examining the technique of angiogenesis assessment in invasive breast cancer. Br J Cancer 76(8):1046–54 Dittmer D, Pati S, Zambetti G, Chu S, Teresky AK, Moore M, Finlay C, Levine AJ, 1993, Gain of function mutations in p53. Nat Genet 4(1):42–6 Chrisanthar R, Knappskog S, Lokkevik E et al, 2008, CHEK2 mutations affecting kinase activity together with mutations in TP53 indicate a functional pathway associated with resistance to epirubicin in primary breast cancer. PLoS ONE 3(8): e3062 Bug M, Dobbelstein M, 2011, Anthracyclines induce the accumulation of mutant p53 through E2F1-dependent and -independent mechanisms. Oncogene 30(33):3612–24 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 949 EP 960 DI 10.1007/s12253-012-9525-9 PG 12 ER PT J AU Kulkarni, A Thota, B Mallavarapu, RS Thennarasu, K Arivazhagan, A Santosh, V Chandramouli, AB AF Kulkarni, Aniruddh Thota, Balaram Mallavarapu, R Srividya Thennarasu, Kandavel Arivazhagan, Arimappamagan Santosh, Vani Chandramouli, Ashwathnarayanara Bangalore TI Expression Pattern and Prognostic Significance of IGFBP Isoforms in Anaplastic Astrocytoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Anaplastic astrocytoma; Insulin-like growth factor binding protein; Immunohistochemistry; Prognosis ID Anaplastic astrocytoma; Insulin-like growth factor binding protein; Immunohistochemistry; Prognosis AB The role of insulin- like growth factors and their regulatory proteins (IGFBP isoforms) in gliomas, particularly glioblastoma, has been a subject of active research in recent years. There is paucity of literature on their expression and impact on clinical outcome in anaplastic astrocytomas. To evaluate the expression patterns of IGFBP isoforms in anaplastic astrocytoma and correlate with clinical outcome, a retrospective study of 53 adult patients operated for supratentorial lobar anaplastic astrocytoma was performed. The protein expression of IGFBP isoforms (IGFBP-2, -3, -5 and -7), was studied by immunohistochemistry on all samples. The patients were followed up and outcome was documented. The median age at presentation in the present study was 35 years. The pattern of staining was intra cytoplasmic, homogenous and diffuse for IGFBP-2, -3 and -5 and granular for IGFBP-7. IGFBP-2 expression was significantly low in anaplastic astrocytoma as compared to other isoforms (P<0.001). IGFBP-3 expression was higher than the other isoforms. However, its’ expression correlated with favorable overall survival and demonstrated a trend towards significance on univariate analysis. The present study is the first of its kind to describe comprehensively the pattern of expression of IGFBP isoforms (IGFBP-2, -3, -5 and -7) in anaplastic astrocytomas. IGFBP-2 and IGFBP-3 expression patterns and correlation to prognosis were distinct in anaplastic astrocytoma patients, contradictory to what has been reported in glioblastoma, thus giving further evidence that anaplastic astrocytomas are molecularly distinct from glioblastoma. C1 [Kulkarni, Aniruddh] National Institute of Mental Health and Neurosciences, Department of NeurosurgeryBangalore, India. [Thota, Balaram] National Institute of Mental Health and Neurosciences, Department of NeuropathologyBangalore, India. [Mallavarapu, R Srividya] National Institute of Mental Health and Neurosciences, Department of NeuropathologyBangalore, India. [Thennarasu, Kandavel] National Institute of Mental Health and Neurosciences, Department of BiostatisticsBangalore, India. [Arivazhagan, Arimappamagan] National Institute of Mental Health and Neurosciences, Department of NeurosurgeryBangalore, India. [Santosh, Vani] National Institute of Mental Health and Neurosciences, Department of NeuropathologyBangalore, India. [Chandramouli, Ashwathnarayanara Bangalore] National Institute of Mental Health and Neurosciences, Department of NeurosurgeryBangalore, India. RP Santosh, V (reprint author), National Institute of Mental Health and Neurosciences, Department of Neuropathology, Bangalore, India. EM vani.santosh@gmail.com CR Bondy ML, Scheurer ME, Malmer B, Barnholtz-Sloan JS, Davis FG, Il'yasova D et al, 2008, Brain tumor epidemiology: consensus from the brain tumor epidemiology consortium. Cancer 113(7 Suppl):1953–1968 Central Brain Tumor Registry of the United States, CBTRUS,, 2008, Statistical Report: Primary Brain Tumors in the United States, 2000– 2004. Hinsdale, Ill: Central Brain Tumor Registry of the United States Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A et al, 2007, The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114(2):97–109 Nomiya T, Nemoto K, Kumabe T, Takai Y, Yamada S, 2007, Prognostic significance of surgery and radiation therapy in cases of anaplastic astrocytoma: retrospective analysis of 170 cases. J Neurosurg 106(4):575–581 Prados MD, Seiferheld W, Sandler HM, Buckner JC, Phillips T, Schultz C et al, 2004, Phase III randomized study of radiotherapy plus procarbazine, lomustine, and vincristine with or without BUdR for treatment of anaplastic astrocytoma: final report of RTOG 9404. Int J Radiat Oncol Biol Phys 58(4):1147–1152 Smith JS, Tachibana I, Passe SMet al, 2001, PTEN mutation, EGFR amplification, and outcome in patients with anaplastic astrocytoma and glioblastoma multiforme. J Natl Cancer Inst 93:1246–1256 Daumas-Duport C, Scheithauer BW, O’Fallon JR, Kelly P, 1988, Grading of astrocytomas: a simple and reproducible method. Cancer 62:2152–2165 Prados MD, Gutin PH, Phillips TL,Wara WM, Larson DA, Sneed PK et al, 1992, Highly anaplastic astrocytoma: a review of 357 patients treated between 1977 and 1989. Int J Radiat Oncol Biol Phys 23:3–8 Cunningham JM, Kimmel DW, Scheithauer BW, O’Fallon JR, Novotny PJ, Jenkins RB, 1997, Analysis of proliferation markers and p53 expression in gliomas of astrocytic origin: relationships and prognostic value. J Neurosurg 86:121–130 Hai Yan D, Parsons W, Jin G, Roger McLendon B et al, 2009, IDH1 and IDH2 mutations in gliomas. N Engl J Med 360(8):765– 773 Wang H, Fuller GN, Zhang W, 2004, Insulin-like growth factors and insulin-like growth factors binding proteins in CNS tumors. In: Zhang W, Fuller GN, eds, Genomic and molecular neurooncology. Jones and Bartlett Publishers, Sudbury, pp 119–130 Trojan J, Cloix JF, Ardourel MY, Chatel M, Anthony DD, 2007, Insulin-like growth factor type I biology and targeting in malignant gliomas. Neuroscience 145:795–811 Fuller GN, Rhee CH, Hess KR, Caskey LS, Wang R, Bruner JM et al, 1999, Reactivation of insulin-like growth factor binding protein 2 expression in glioblastoma multiforme: a revelation by parallel gene expression profiling. Cancer Res 59(17):4228–4232 Fukushima T, Kataoka H, 2007, Roles of insulin-like growth factor binding protein-2, IGFBP-2, in glioblastoma. Anticancer Res 27(6A):3685–3692 Santosh V, Arivazhagan A, Sreekanthareddy P, Srinivasan H, Thota B, Srividya MR et al, 2010, Grade-specific expression of insulin-like growth factor–binding proteins-2, -3, and −5 in astrocytomas: IGFBP-3 emerges as a strong predictor of survival in patients with newly diagnosed glioblastoma. Cancer Epidemiol Biomarkers Prev 19(6):1399–1408 Moore LM, Holmes KM, Smith SM, Wu Y, Tchougounova E, Uhrbom L et al, 2009, IGFBP2 is a candidate biomarker for Ink4a-Arf status and a therapeutic target for high-grade gliomas. Proc Natl Acad Sci U S A 106(39):16675–16679 Wang H, Shen W, Huang H, Hu L, Ramdas L, Zhou YH et al, 2003, Insulin-like growth factor binding protein 2 enhances glioblastoma invasion by activating invasion-enhancing genes. Cancer Res 63(15):4315–4321 McDonald KL, O'Sullivan MG, Parkinson JF, Shaw JM, Payne CA, Brewer JM et al, 2007, IQGAP1 and IGFBP2: valuable biomarkers for determining prognosis in glioma patients. J Neuropathol Exp Neurol 66(5):405–417 Reddy SP, Britto R, Vinnakota K, Aparna H, Sreepathi HK, Thota B et al, 2008, Novel glioblastoma markers with diagnostic and prognostic value identified through transcriptome analysis. Clin Cancer Res 14(10):2978–2987 Lin Y, Jiang T, Zhou K, Xu L, Chen B, Li G et al, 2009, Plasma IGFBP-2 levels predict clinical outcomes of patients with highgrade gliomas. Neuro Oncol 11(5):468–476 Wang H, Zhang W, Fuller GN, 2006, Overexpression of IGFBP5, but not IGFBP3, correlates with the histologic grade of human diffuse glioma: a tissue microarray and immunohistochemical study. Technol Cancer Res Treat 5(3):195–199 O'Han MK, Baxter RC, Schedlich LJ, 2009, Effects of endogenous insulin-like growth factor binding protein-3 on cell cycle regulation in breast cancer cells. Growth Factors 27(6):394–408 Vestey SB, Perks CM, Sen C, Calder CJ, Holly JM, Winters ZE, 2005, Immunohistochemical expression of insulin-like growth factor binding protein-3 in invasive breast cancers and ductal carcinoma in situ: implications for clinicopathology and patient outcome. Breast Cancer Res 7(1):R119–R129 Butt AJ, Martin JL, Dickson KA, McDougall F, Firth SM, Baxter RC, 2004, Insulin-like growth factor binding protein-3 expression is associated with growth stimulation of T47D human breast cancer cells: the role of altered epidermal growth factor signaling. J Clin Endocrinol Metab 89(4):1950–1956 Kim HS, Lee WJ, Lee SW, Chae HW, Kim DH, Oh Y, 2010, Insulin-like growth factor binding protein-3 induces G1 cell cycle arrest with inhibition of cyclin-dependent kinase 2 and 4 in MCF-7 human breast cancer cells. Horm Metab Res 42(3):165–172 Dupart JJ, Trent JC, Lee HY, Hess KR, Godwin AK, Taguchi T et al, 2009, Insulin-like growth factor binding protein-3 has dual effects on gastrointestinal stromal tumor cell viability and sensitivity to the anti-tumor effects of imatinib mesylate in vitro. Mol Cancer 8:99 Chang MH, Lee J, Han J, Park YH, Ahn JS, Park K et al, 2009, Prognostic role of insulin-like growth factor receptor-1 expression in small cell lung cancer. APMIS 117(12):861–869 Sztefko K, Hodorowicz-Zaniewska D, Popiela T, Richter P, 2009, IGF-I, IGF-II, IGFBP2, IGFBP3 and acid-labile subunit, ALS, in colorectal cancer patients before surgery and during 1 year follow up in relation to age. Adv Med Sci 54(1):51–58 Miyake H, Pollak M, Gleave ME, 2000, Castration-induced up-regulation of insulin-like growth factor binding protein-5 potentiates insulin-like growth factor-I activity and accelerates progression to androgen independence in prostate cancer models. Cancer Res 60(11):3058–3064 Hou XJ, Zhang YZ, Liu X, Meng LH, Qiao YB, 2009, Expressions of IGFBP-5, cFLIP in cervical intraepithelial neoplasia, cervical carcinoma and their clinical significances: a molecular pathology. J Exp Clin Cancer Res 28:70 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 961 EP 967 DI 10.1007/s12253-012-9526-8 PG 7 ER PT J AU Ye, F Jiao, J Zhou, C Cheng, Q Chen, H AF Ye, Feng Jiao, Jie Zhou, Caiyun Cheng, Qi Chen, Huaizeng TI Nucleotide Excision Repair Gene Subunit XPD is Highly Expressed in Cervical Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE XPD; Cervical squamous cell carcinoma; Nucleotide excision repair ID XPD; Cervical squamous cell carcinoma; Nucleotide excision repair AB One of the subunits in the mammalian transcription factor IIH complex,XPD (TFIIH p80), plays a significant role in the nucleotide excision repair pathway. Events such as abnormal DNA excision repair may be involved in the cervical carcinogenesis process. Expression of the human XPD protein was examined using immunohistochemistry in 84 normal cervical tissues and 148 cervical squamous cell carcinoma samples. Additionally, qRT-PCR was performed to analyse the XPD mRNA expression in 69 fresh normal cervical tissues and 110 cervical carcinoma samples. The relationships between XPD expression and various clinicopathological features (including age, FIGO stage, tumor size, stroma involvement, lymph node metastasis and histologic grade) were assessed. The XPD (TFIIH p80) protein was detected primarily in the cytoplasm.We found a statistically significant difference in XPD expression level in cervical carcinoma versus normal cervical tissue (Z0−7.302, P<0.000). Notably, XPD mRNA was significantly over-expressed in cervical carcinoma tissues but not in normal cervix tissues (t06.942, P<0.000). However, no statistically significant relationship was found regarding XPD expression and age, FIGO stage, tumor size, stroma involvement, lymph node metastasis or histologic grade (P< 0.089, 0.953, 0.809, 0.275, 0.421, 0.387 respectively). Our results showed that XPD was highly expressed in cervical squamous cell carcinoma tissues. A poorly understood change may occur during the XPD transcription process, resulting in the abnormal enrichment seen from mRNA to the protein level, thus leaving the protein unable to perform the normal function of excision repair. There is a need for further research in order to elucidate the specific mechanism involved. C1 [Ye, Feng] School of Medicine, Zhejiang University, Women’s Hospital, Women’s Reproductive Health Laboratory of Zhejiang Province, Xueshi Rd #2, 310006 Hangzhou, China. [Jiao, Jie] School of Medicine, Zhejiang University, Women’s Hospital, Department of Gynecological Oncology, Xueshi Rd #2, 310006 Hangzhou, China. [Zhou, Caiyun] School of Medicine, Zhejiang University, Women’s Hospital, Department of Pathology, Xueshi Rd #2, 310006 Hangzhou, China. [Cheng, Qi] School of Medicine, Zhejiang University, Women’s Hospital, Women’s Reproductive Health Laboratory of Zhejiang Province, Xueshi Rd #2, 310006 Hangzhou, China. [Chen, Huaizeng] School of Medicine, Zhejiang University, Women’s Hospital, Women’s Reproductive Health Laboratory of Zhejiang Province, Xueshi Rd #2, 310006 Hangzhou, China. RP Chen, H (reprint author), School of Medicine, Zhejiang University, Women’s Hospital, Women’s Reproductive Health Laboratory of Zhejiang Province, 310006 Hangzhou, China. EM chenhz@zju.edu.cn CR Wang J, Lv XW, Shi JP et al, 2007, Mechanisms involved in ceramide-induced cell cycle arrest in human hepatocarcinoma cells. World J Gastroenterol 13:1129–1134 Wang Y, Huang B, Edelmann L et al, 2010, Genes essential for cell viability that are linked to tumor suppressor genes play a role in cancer susceptibility. Cell Cycle 9:3144–3145 Falik-Zaccai TC, Keren Z, Slor H, 2009, The Versatile DNA Nucleotide Excision Repair, NER, and Its Medical Significance. Pediatr Endocrinol Rev 7:117–122 Li X, Urwyler O, Suter B, 2010, Drosophila Xpd regulates Cdk7 localization, mitotic kinase activity, spindle dynamics, and chromosome segregation. PLoS Genet 6:e1000876 Aloyz R, Xu ZY, Bello V et al, 2002, Regulation of cisplatin resistance and homologous recombinational repair by the TFIIH subunit XPD. Cancer Res 62:5457–5462 Chen J, Suter B, 2003, Xpd, a structural bridge and a functional link. Cell Cycle 2:503–506 Chen J, Larochelle S, Li X et al, 2003, Xpd/Ercc2 regulates CAK activity and mitotic progression. Nature 424:228–232 He X, Ye F, Zhang J et al, 2008, Susceptibility of XRCC3, XPD, and XPG genetic variants to cervical carcinoma. Pathobiology 75:356–363 Tavana O, Benjamin CL, Puebla-Osorio N et al, 2010, Absence of p53-dependent apoptosis leads to UV radiation hypersensitivity, enhanced immunosuppression and cellular senescence. Cell Cycle 9:3328–3336 Singh RP, Agarwal R, 2006, Natural flavonoids targeting deregulated cell cycle progression in cancer cells. Curr Drug Targets 7:345–354 Marchisio M, Bertagnolo V, Lanuti P et al, 2006, Nuclear protein kinase C-delta: a possible check-point of cell cycle progression. Int J Immunopathol Pharmacol 19:287–291 Mydlikova Z, Gursky J, Pirsel M, 2010, Transcription factor IIH —the protein complex with multiple functions. Neoplasma 57:287–290 Zhovmer A, Oksenych V, Coin F, 2010, Two sides of the same coin: TFIIH complexes in transcription and DNA repair. Scientific World Journal 10:633–643 Zurita M, Merino C, 2003, The transcriptional complexity of the TFIIH complex. Trends in Genetics 19:578–584 Oksenych V, de Jesus BB, Zhovmer A et al, 2009, Molecular insights into the recruitment of TFIIH to sites of DNA damage. Embo J 28:2971–2980 Jaitovich-Groisman I, Benlimame N, Slagle BL et al, 2001, Transcriptional regulation of the TFIIH transcription repair components XPB and XPD by the hepatitis B virus x protein in liver cells and transgenic liver tissue. J Bio Chem 276:14124–14132 Oksenych V, Coin F, 2010, The long unwinding road: XPB and XPD helicases in damaged DNA opening. Cell Cycle 9:90–96 Gao W, Romkes M, Zhong S et al, 2010, Genetic polymorphisms in the DNA repair genes XPD and XRCC1, p53 gene mutations and bladder cancer risk. Oncol Rep 24:257–262 Sturgis EM, Zheng R, Li L et al, 2000, XPD/ERCC2 polymorphisms and risk of head and neck cancer: a case–control analysis. Carcinogenesis 21:2219–2223 Butkiewicz D, Rusin M, Enewold L et al, 2001, Genetic polymorphisms in DNA repair genes and risk of lung cancer. Carcinogenesis 22:593–597 Vogel U, Hedayati M, Dybdahl M et al, 2001, Polymorphisms of the DNA repair gene XPD: correlations with risk of basal cell carcinoma revisited. Carcinogenesis 22:899–904 Long XD, Ma Y, Zhou YF et al, 2009, XPD codon 312 and 751 polymorphisms, and AFB1 exposure, and hepatocellular carcinoma risk. BMC Cancer 9:400 Aboussekhra A, Biggerstaff M, Shivji MK et al, 1995, Mammalian DNA nucleotide excision repair reconstituted with purified protein components. Cell 80:859–868 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 969 EP 975 DI 10.1007/s12253-012-9527-7 PG 7 ER PT J AU Kovacs, J Gurzu, S Jung, J Szederjesi, J Copotoiu, SM Copotoiu, R Azamfirei, L AF Kovacs, Judit Gurzu, Simona Jung, Janos Szederjesi, Janos Copotoiu, Sanda-Maria Copotoiu, Ruxandra Azamfirei, Leonard TI Clinico-Pathological Particularities of the Shock- Related Pancreatitis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute pancreatitis; Analgo-sedation; Hyperglycemia; Hypocalcemia; Severe hypotension ID Acute pancreatitis; Analgo-sedation; Hyperglycemia; Hypocalcemia; Severe hypotension AB Acute pancreatitis can develop in patients with shock due to the underlying diseases, surgical interventions or because of severe hypoperfusion. The aim of our work was to study the histological alterations of the pancreas in patients dying after cardiogenic, hypovolemic or septic shock, to demonstrate the presence and severity of pancreatic injury. We performed a retrospective study which included patients who died and who were autopsied after different types of shock, hospitalized between 2007–2009 in general and cardiac intensive care units. We excluded the patients with known pancreatic diseases. From 223 patients included in our study 39 presented necrotising hemorrhagic alteration of the pancreatic tissue. There were no differences in histological and immunohistochemical findings between the different etiopathogenetic types of shock. None of the patients had characteristic clinical signs for acute pancreatitis. The digestive symptoms, they presented, could be related to the underlying disease or to postoperative state. The common findings in these patients were prolonged and severe hypotension, associated renal dysfunction, leucocytosis, hyperglycemia and hypocalcemia. Pancreatitis can occur in patients with shock, due to prolonged hypoperfusion of the pancreas. It is difficult to diagnose it because clinical signs are altered due to severity of underlying disease or analgo-sedation commonly used in intensive care. We therefore recommend in patients with shock to consider the possible development of ischemic pancreatitis for prompt and efficient treatment. C1 [Kovacs, Judit] University of Medicine and Pharmacy Tg-Mures, Department of Anesthesia and Intensive CareTargu-Mures, Romania. [Gurzu, Simona] University of Medicine and Pharmacy Targu-Mures, Department of Pathology, Gheorghe Marinescu 38, 540193 Targu-Mures, Romania. [Jung, Janos] University of Medicine and Pharmacy Targu-Mures, Department of Pathology, Gheorghe Marinescu 38, 540193 Targu-Mures, Romania. [Szederjesi, Janos] University of Medicine and Pharmacy Tg-Mures, Department of Anesthesia and Intensive CareTargu-Mures, Romania. [Copotoiu, Sanda-Maria] University of Medicine and Pharmacy Tg-Mures, Department of Anesthesia and Intensive CareTargu-Mures, Romania. [Copotoiu, Ruxandra] University of Medicine and Pharmacy Tg-Mures, Department of Anesthesia and Intensive CareTargu-Mures, Romania. [Azamfirei, Leonard] University of Medicine and Pharmacy Tg-Mures, Department of Anesthesia and Intensive CareTargu-Mures, Romania. RP Gurzu, S (reprint author), University of Medicine and Pharmacy Targu-Mures, Department of Pathology, 540193 Targu-Mures, Romania. EM simonagurzu@yahoo.com CR Piton G, Barbot O, Manzon C et al, 2010, Acute ischemic pancreatitis following cardiac arrest: a case report. J Pancreas, Online, 11(5):456–459 Hackert T, Hartwig W, Fritz S et al, 2009, Ischemic acute pancreatitis: clinical features of 11 patients and review of the literature. Am J Surg 197(4):450–454 Sakorafas GH, Tsiotos GG, Sarr MG, 2000, Ischemia/reperfusion induced pancreatitis. Dig Surg 17:3–14 Schmitz-Moorman P, 1981, Comparative radiological and morphological study of the human pancreas. Acute necrotising pancreatitis in man. Path Res Pract 171:325–335 Dugernier T, Laterre PF, Reynaert MS, 2000, Ascites fluid in severe acute pancreatitis: from pathophysiology to therapy. Acta Gastroenterol Belg 63(3):264–268 Nordback I, Lauslahti K, 1986, Clinical pathology of acute necrotising pancreatitis. J Clin Pathol 39:68–74 Warshaw AL, O’ Hara PJ, 1978, Susceptibility of the pancreas to ischemic injury in shock. Ann Surg 188:197–201 Warzecha Z, Dembinski A, Ceranowicz P, 2004, Immunohistochemical expression of FGF-2, PDGF-A, VEGF and TGF beta RII in the pancreas in the course of ischemia/reperfusion-induced acute pancreatitis. J Physiol Pharmacol 55(4):791–810 Nandy D, Mukhopadhyay D, 2011, Growth factor mediated signaling in pancreatic pathogenesis. Review. Cancers 3:841–871 UedaT TY, Yasuda T, 2006, Vascular endothelial growth factor increases in serum and protects against the organ injuries in severe acute pancreatitis. J Surg Res 134(2):223–230 Beere HM, Wolf BB, Cain K et al, 2000, Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome. Nat Cell Biol 2(8):469–75 Bhagat L, Singh VP, Song AM et al, 2002, Thermal stress-induced HSP70 mediates protection against intrapancreatic trypsinogen activation and acute pancreatitis in rats. Gastroenterology 122, 1):156–65 Ethridge RT, Ehlers RA, Hellmich MR et al, 2000, Acute pancreatitis results in induction of heat shock proteins 70 and 27 and heat shock factor-1. Pancreas 21(3):248–56 Geissler HJ, Fischer UM, Grunert S et al, 2006, Incidence and outcome of gastrointestinal complications after cardiopulmonary bypass. Interact CardioVasc Thorac Surgery 5:239–242 Munoz A, Katerndahl DA, 2000, Problem-oriented diagnosis. Diagnosis and management of acute pancreatitis. Am Fam Physician 62:164–74 Wan S, Arifi AA, Chan CSYet al, 2002, Is hyperamylasemia after cardiac surgery due to cardiopulmonary bypass? Asian Cardiovasc Thorac Ann 10:115–118 Butler J, Mackway-Jones K, 2002, Serum amylase or lipase to diagnose pancreatitis in patients presenting with abdominal pain. Emerg Med J 19:430–431 Weir GC, Lesser PB, Drop LJ et al, 1975, The hypocalcemia of acute pancreatitis. Ann Intern Med 83(2):185–189 Khan JH, Lambert AM, Habib JH et al, 2006, Abdominal complications after heart surgery. Ann Thorac Surg 82:1796– 1801 Perez A, Ito H, Farivar RS et al, 2005, Risk factors and outcomes of pancreatitis after open heart surgery. Am J Surg 190(3):401– 405 Del-Castillo CF, Harringer W, Warshaw AL et al, 1991, Risk factors for pancreatic cellular injury after cardiopulmonary bypass. N Engl J Med 325:382–387 Kuo IM, Wang F, Liu KH et al, 2009, Post-gastrectomy acute pancreatitis in a patient with gastric carcinoma and pancreas divisum. World J Gastroenterol 15(36):4596–4600 Blom RLGM, van Heijl M, Busch ORC et al, 2009, Acute pancreatitis in the postoperative course after esophagectomy: a major complication described in 4 patients. Case Rep Gastroenterol 3:382–388 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 977 EP 981 DI 10.1007/s12253-012-9528-6 PG 5 ER PT J AU Wu, L Wang, Z Lu, R Jiang, W AF Wu, Lielin Wang, Zhiming Lu, Rongli Jiang, Wei TI Expression of High Mobility Group A2 is Associated with Poor Survival in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article C1 [Wu, Lielin] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan, China. [Wang, Zhiming] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan, China. [Lu, Rongli] Central South University, Xiangya Hospital, Department of Respiratory, 410008 Changsha, Hunan, China. [Jiang, Wei] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan, China. RP Wang, Z (reprint author), Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, China. EM wzm005@yahoo.com.cn CR Srivatanakul P, Sriplung H, Deerasamee S, 2004, Epidemiology of liver cancer: an overview. Asian Pac J Cancer Prev 5:118–125 Mann CD, Neal CP, Garcea G et al, 2007, Prognostic molecular markers in hepatocellular carcinoma: a systematic review. Eur J Cancer 43:979–992 Reeves R, Beckerbauer L, 2001, HMGI/Y proteins: flexible regulators of transcription and chromatin structure. Biochim Biophys Acta 1519:13–29 Fusco A, Fedele M, 2007, Roles of HMGA proteins in cancer. Nat Rev Cancer 7:899–910 Cleynen I, Van de Ven WJ, 2008, The HMGA proteins: a myriad of functions. Int J Oncol 32:289–305 Chiappetta G, Avantaggiato V, Visconti R et al, 1996, High level expression of the HMGI, Y, gene during embryonic development. Oncogene 13:2439–2446 Rogalla P, Drechsler K, Frey G et al, 1996, HMGI-C expression patterns in human tissues. Implications for the genesis of frequent mesenchymal tumors. Am J Pathol 149:775–779 Ashar HR, Fejzo MS, Tkachenko A et al, 1995, Disruption of the architectural factor HMGI-C: DNA-binding AT hook motifs fused in lipomas to distinct transcriptional regulatory domains. Cell 82:57–65 Mine N, Kurose K, Nagai H et al, 2001, Gene fusion involving HMGIC is a frequent aberration in uterine leiomyomas. J Hum Genet 46:408–412 Kazmierczak B, Meyer-Bolte K, Tran KH et al, 1999, A high frequency of tumors with rearrangements of genes of the HMGI, Y, family in a series of 191 pulmonary chondroid hamartomas. Gene Chromosome Canc 26:125–133 Rogalla P, Drechsler K, Kazmierczak B et al, 1997, Expression of HMGI-C, a member of the high mobility group protein family, in a subset of breast cancers: relationship to histologic grade. Mol Carcinog 19:153–156 Sarhadi VK, Wikman H, Salmenkivi K et al, 2006, Increased expression of high mobility group A proteins in lung cancer. J Pathol 209:206–212 Meyer B, Loeschke S, Schultze A et al, 2007, HMGA2 overexpression in non-small cell lung cancer. Mol Carcinog 46:503– 511 Abe N, Watanabe T, Suzuki Y et al, 2003, An increased highmobility group A2 expression level is associated with malignant phenotype in pancreatic exocrine tissue. Br J Cancer 89:2104– 2109 Belge G, Meyer A, Klemke M et al, 2008, Upregulation of HMGA2 in thyroid carcinomas: a novel molecular marker to distinguish between benign and malignant follicular neoplasias. Gene Chromosome Canc 47:56–63 Motoyama K, Inoue H, Nakamura Y et al, 2008, Clinical significance of high mobility group A2 in human gastric cancer and its relationship to let-7 microRNA family. Clin Cancer Res 14:2334– 2340 Livak KJ, Schmittquen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), Method. Method 25:402–408 Miyazawa J, Mitoro A, Kawashiri S et al, 2004, Expression of mesenchyme-specific gene HMGA2 in squamous cell carcinomas of the oral cavity. Cancer Res 64:2024–2029 Park S-M, Shell S, Radjabi AR et al, 2007, Let-7 prevents early cancer progression by suppressing expression of the embryonic gene HMGA2. Cell Cycle 6:2585–2590 Hristov AC, Cope L, Reyes MD et al, 2009, HMGA2 protein expression correlates with lymph node metastasis and increased tumor grade in pancreatic ductal adenocarcinoma. Mod Pathol 22:43–49 Vallone D, Battista S, Pierantoni GM et al, 1997, Neoplastic transformation of rat thyroid cells requires the junB and fra-1 gene induction which is dependent on the HMGI-C gene product. EMBO J 16:5310–5321 Fedele M et al, 2006, HMGA2 induces pituitary tumorigenesis by enhancing E2F1 activity. Cancer Cell 9:459–471 Thuault S, Valcourt U, Petersen M et al, 2006, Transforming growth factor employs HMGA2 to elicit epithelial-mesenchymal transition. J Cell Biol 174:175–183 Wang X, Liu X, Li AY et al, 2011, Overexpression of HMGA2 promotes metastasis and impacts survival of colorectal cancers. Clin Cancer Res 17:2570–2580 Langelotz C, Schmid P, Jakob C et al, 2003, Expression of highmobility- group-protein HMGI-C mRNA in the peripheral blood is an independent poor prognostic indicator for survival in metastatic breast cancer. Br J Cancer 88:1406–1410 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 983 EP 987 DI 10.1007/s12253-012-9514-z PG 5 ER PT J AU Gong, L Cai, Y Zhou, X Yang, H AF Gong, Liang Cai, Yun Zhou, Xiangdong Yang, Heping TI Activated Platelets Interact with Lung Cancer Cells Through P-Selectin Glycoprotein Ligand-1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Platelet activation; P-selectin; Pselectin glycoprotein ligand 1; RNA interference ID Lung cancer; Platelet activation; P-selectin; Pselectin glycoprotein ligand 1; RNA interference AB Hematogenous metastasis always leads to the poor prognosis of non-small cell lung cancers (NSCLC). Activated platelets are involved in hematogenous metastasis and may be a potential therapeutic target. P-selectin is an important adhesion molecule and expressed on the surface of activated platelets. P-selectin glycoprotein ligand-1 (PSGL-1) as a transmembrane protein is expressed on the surface of various cell types. P-selectin can bind to PSGL-1, and thereby initiate the platelet-mediated cell adhesion. The aim of the study was to investigate the degree of platelet activation in NSCLC and the roles of PSGL-1 in the activation of platelets. Purified platelets were obtained from NSCLC patients (40 lung adenocarcinomas and 26 lung squamous cell carcinomas), and P-selectin expression was detected by fluorescence-activated cell sorter. The population of peripheral blood platelets with P-selectin expression in lung adenocarcinoma was 63.16±25.44 %, and significantly higher than that in lung squamous cell carcinoma (35.97±17.19 %) and the healthy population (9.12± 7.66 %, n030). A specific small hairpin RNA (shRNA) for PSGL-1 was transfected into A549 human alveolar cell carcinoma cells. The expressions of PSGL-1 mRNA and protein were significantly reduced with the PSGL-1 shRNA (p<0.01). Furthermore, the knockdown of PSGL-1 also resulted in the significantly reduced aggregate formation of activated platelets and A549 cells. Thus, activated platelets may interact with lung cancer cells through PSGL-1. Inhibiting platelet activation and/or down-regulating PSGL-1 expression may be useful for suppression of tumor metastasis. C1 [Gong, Liang] the Third Military Medical University, Southwest Hospital, Department of Respiratory diseasesChongqing, China. [Cai, Yun] the Third Military Medical University, Southwest Hospital, Southwest Cancer InstituteChongqing, China. [Zhou, Xiangdong] the Third Military Medical University, Southwest Hospital, Department of Respiratory diseasesChongqing, China. [Yang, Heping] the Third Military Medical University, Southwest Hospital, Department of Respiratory diseasesChongqing, China. RP Yang, H (reprint author), the Third Military Medical University, Southwest Hospital, Department of Respiratory diseases, Chongqing, China. EM mkojima@163.com CR Jemal A, Siegel R, Ward E, Murray T, Xu J, Thun MJ, 2007, Cancer statistics. CA Cancer J Clin 57:43–66 Inamura K, Ishikawa Y, 2010, Lung cancer progression and metastasis from the prognostic point of view. Clin ExpMetastasis 27:389–397 Geiger TR, Peeper DS, 2009, Metastasis mechanisms. Biochim Biophys Acta 1796:293–308 Joyce JA, Pollard JW, 2009, Microenvironmental regulation of metastasis. Nat Rev Cancer 9:239–252 Borsig L, 2008, The role of platelet activation in tumor metastasis. Expert Rev Anticancer Ther 8:1247–1255 Sierko E, Wojtukiewicz MZ, 2007, Inhibition of platelet function, does it offer a chance of better cancer progression control? Semin Thromb Hemost 33:712–721 Kunita A, Kashima TG, Morishita Y, FukayamaM, Kato Y, Tsuruo T, Fujita N, 2007, The platelet aggregation-inducing factor aggrus/podoplanin promotes pulmonary metastasis. Am J Pathol 170:1337–1347 Gonzalez Barcala FJ, Garcia Prim JM, Moldes Rodriguez M, AlvarezFernandez J, Rey Rey MJ, Pose Reino A, Valdes Cuadrado L, 2010, Platelet count: association with prognosis in lung cancer. Med Oncol 27:357–362 Ludwig RJ, Schon MP, Boehncke WH, 2007, P-selectin: a common therapeutic target for cardiovascular disorders, inflammation and tumour metastasis. Expert Opin Ther Targets 11:1103–1117 Dimitroff CJ, Descheny L, Trujillo N, Kim R, Nguyen V, Huang W, Pienta KJ, Kutok JL, Rubin MA, 2005, Identification of leukocyte E-selectin ligands, P-selectin glycoprotein ligand-1 and E-selectin ligand-1, on human metastatic prostate tumor cells. Cancer Res 65:5750–5760 Baisse B, Galisson F, Giraud S, Schapira M, Spertini O, 2007, Evolutionary conservation of P-selectin glycoprotein ligand-1 primary structure and function. BMC Evol Biol 14:166 Raes G, Ghassabeh GH, Brys L, Mpofu N, Verschueren H, Vanhecke D, De Baetselier P, 2007, The metastatic T-cell hybridoma antigen/P-selectin glycoprotein ligand 1 is required for hematogenous metastasis of lymphomas. Int J Cancer 121:2646–2652 Baumann K, Kowalczyk D, Gutjahr T, Pieczyk M, Jones C, Wild MK, Vestweber D, Kunz H, 2009, Sulfated and non-sulfated glycopeptide recognition domains of P-selectin glycoprotein ligand 1 and their binding to P- and E-selectin. Angew Chem Int Ed Engl 48:3174–3178 Marathe DD, Buffone A Jr, Chandrasekaran EV, Xue J, Locke RD, Nasirikenari M, Lau JT, Matta KL, Neelamegham S, 2010, Fluorinated per-acetylated GalNAc metabolically alters glycan structures on leukocyte PSGL-1 and reduces cell binding to selectins. Blood 115:1303–1312 Thomas GM, Panicot-Dubois L, Lacroix R et al, 2009, Cancer cell-derived microparticles bearing P-selectin glycoprotein ligand 1 accelerate thrombus formation in vivo. JEM 206(9):1913–1927 Chambers AF,NaumovGN, Varghese HJ, Nadkarni KV,MacDonald IC, Groom AC, 2001, Critical steps in hematogenous metastasis, an overview. Surg Oncol Clin N Am 10:243–255 Sleeman J, Steeg PS, 2010, Cancer metastasis as a therapeutic target. Eur J Cancer 46:1177–1180 Tsuruo T, Fujita N, 2008, Platelet aggregation in the formation of tumor metastasis. Proc Jpn Acad Ser B Phys Biol Sci 84:189–198 Noble S, Pasi J, 2010, Epidemiology and pathophysiology of cancer-associated thrombosis. Br J Cancer 102:S2–S9 Barthel SR, Gavino JD, Descheny L, Dimitroff CJ, 2007, Targeting selectins and selectin ligands in inflammation and cancer. Expert Opin Ther Targets 11:1473–1491 Laubli H, Borsig L, 2010, Selectins promote tumor metastasis. Semin Cancer Biol 20:169–177 Iiizumi M, Mohinta S, Bandyopadhyay S, Watabe K, 2007, Tumor-endothelial cell interactions: Therapeutic potential. Microvasc Res 74:114–120 Ley K, 2009, Cell adhesion under flow. Microcirculation 16:1–2 Fuchs B, Budde U, Schulz A, Kessler CM, Fisseau C, Kannicht C, 2010, Flow-based measurements of von Willebrand factor, VWF, function: Binding to collagen and platelet adhesion under physiological shear rate. Thromb Res 125:239–245 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 989 EP 996 DI 10.1007/s12253-012-9531-y PG 8 ER PT J AU Roszak, A Mostowska, A Sowinska, A Lianeri, M Jagodzinski, PP AF Roszak, Andrzej Mostowska, Adrianna Sowinska, Anna Lianeri, Margarita Jagodzinski, P Pawel TI Contribution of IL12A and IL12B Polymorphisms to the Risk of Cervical Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical carcinoma; IL12A and IL12B; Polymorphisms ID Cervical carcinoma; IL12A and IL12B; Polymorphisms AB We studied the contribution of the IL12A 3′UTR G>A (rs568408) and IL12B 3′UTR A>C (rs3212227) polymorphisms to the risk of cervical cancer. These polymorphisms were genotyped in four hundred-five patients with cervical cancer and four hundred fifty unrelated healthy females from the Polish population. Logistic regression analysis adjusting for age, pregnancy, oral contraceptive use, tobacco smoking, and menopausal status revealed that the IL12B 3′UTR A>C polymorphism could be a genetic risk factor for cervical cancer. The adjusted odds ratio (OR) for patients with the A/C genotype vs A/A genotype was 1.557 (95 % CI01.173–2.066, p00.0178) and adjusted OR for the C/C or A/C genotype vs the A/A genotype was 1.635 (95 % CI01.241–2.153, p00.0125). However, logistic regression analysis did not show an association of the IL12A 3′UTR G>A polymorphism with cervical cancer development in the studied Polish population. Our studies confirmed that the IL12B 3′UTR A>C polymorphism may be a genetic risk factor for cervical cancer. C1 [Roszak, Andrzej] Greater Poland Cancer Center, Department of Radiotherapy and Gynecological OncologyPoznan, Poland. [Mostowska, Adrianna] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. [Sowinska, Anna] Poznan University of Medical Sciences Poznan, Department of Computer Science and StatisticsPoznan, Poland. [Lianeri, Margarita] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. [Jagodzinski, P Pawel] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. RP Jagodzinski, PP (reprint author), Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 60-781 Poznan, Poland. EM pjagodzi@am.poznan.pl CR Arbyn M, Raifu AO, Autier P et al, 2007, Burden of cervical cancer in Europe: estimates for 2004. Ann Oncol 18:1708–1715 Munoz N, Bosch FX, de Sanjose S et al, 2003, Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med 348:518–527 Walboomers JM, Jacobs MV, Manos MM et al, 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189:12–19 Giuliano AR, Harris R, Sedjo RL et al, 2002, Incidence, prevalence, and clearance of type-specific human papillomavirus infections: the Young Women’s Health Study. J Infect Dis 186:462–469 Magnusson PK, Lichtenstein P, Gyllensten UB, 2000, Heritability of cervical tumours. Int J Cancer 88:698–701 Hemminki K, Chen B, 2006, Familial risks for cervical tumors in full and half siblings: etiologic apportioning. Cancer Epidemiol Biomarkers Prev 15:1413–1414 Castellsague X, Munoz N, 2003, Chapter 3: cofactors in human papillomavirus carcinogenesis–role of parity, oral contraceptives, and tobacco smoking. J Natl Cancer Inst Monogr 20–28 Moreno V, Bosch FX, Munoz N et al, 2002, Effect of oral contraceptives on risk of cervical cancer in women with human papillomavirus infection: the IARC multicentric case–control study. Lancet 359:1085–1092 Wu TC, Kurman RJ, 1997, Analysis of cytokine profiles in patients with human papillomavirus-associated neoplasms. J Natl Cancer Inst 89:185–187 Scott M, Stites DP, Moscicki AB, 1999, Th1 cytokine patterns in cervical human papillomavirus infection. Clin Diagn Lab Immunol 6:751–755 Trinchieri G, 1998, Interleukin-12: a cytokine at the interface of inflammmation and immunity. Adv Immunol 70:83–243 Trinchieri G, 2003, Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol 3:133–146 Macatonia SE, Hosken NA, Litton M et al, 1995, Dendritic cells produce IL-12 and direct the development of Th1 cells from naive CD4+ T cells. J Immunol 154:5071–5079 D’Andrea A, Rengaraju M, Valiante NM et al, 1992, Production of natural killer cell stimulatory factor, interleukin 12, by peripheral blood mononuclear cells. J Exp Med 176:1387–1398 Cassatella MA, Meda L, Gasperini S et al, 1995, Interleukin-12 production by human polymorphonuclear leukocytes. Eur J Immunol 25:1–5 Colombo MP, Trinchieri G, 2002, Interleukin-12 in anti-tumor immunity and immunotherapy. Cytokine Growth Factor Rev 13:155–168 Brunda MJ, Luistro L, Warrier RR et al, 1993, Antitumor and antimetastatic activity of interleukin 12 against murine tumors. J Exp Med 178:1223–1230 Imagawa Y, Satake K, Kato Yet al, 2004, Antitumor and antiangiogenic effects of interleukin 12 gene therapy in murine head and neck carcinoma model. Auris Nasus Larynx 31:239–245 Sieburth D, Jabs EW, Warrington JA et al, 1992, Assignment of genes encoding a unique cytokine, IL12, composed of two unrelated subunits to chromosomes 3 and 5. Genomics 14:59–62 Lan Q, Zheng T, Rothman N et al, 2006, Cytokine polymorphisms in the Th1/Th2 pathway and susceptibility to non-Hodgkin lymphoma. Blood 107:4101–4108 Purdue MP, Lan Q, Kricker A et al, 2007, Polymorphisms in immune function genes and risk of non-Hodgkin lymphoma: findings from the New South Wales non-Hodgkin lymphoma study. Carcinogenesis 28:704–712 Nieters A, Yuan JM, Sun CL et al, 2005, Effect of cytokine genotypes on the hepatitis B virus-hepatocellular carcinoma association. Cancer 103:740–748 Lee KM, Shen M, Chapman RS et al, 2007, Polymorphisms in immunoregulatory genes, smoky coal exposure and lung cancer risk in Xuan Wei, China. Carcinogenesis 28:1437–1441 Hou L, El-Omar EM, Chen J et al, 2007, Polymorphisms in Th1- type cell-mediated response genes and risk of gastric cancer. Carcinogenesis 28:118–123 Navaglia F, Basso D, Zambon CF et al, 2005, Interleukin 12 gene polymorphisms enhance gastric cancer risk in H pylori infected individuals. J Med Genet 42:503–510 Brown EE, Fallin D, Ruczinski I et al, 2006, Associations of classic Kaposi sarcoma with common variants in genes that modulate host immunity. Cancer Epidemiol Biomark Prev 15:926–934 Howell WM, Turner SJ, Theaker JM et al, 2003, Cytokine gene single nucleotide polymorphisms and susceptibility to and prognosis in cutaneous malignant melanoma. Eur J Immunogenet 30:409–414 Landi S, Gemignani F, Bottari F et al, 2006, Polymorphisms within inflammatory genes and colorectal cancer. J Negat Results Biomed 5:15 Ben Chaaben A, Busson M, Douik H et al, 2011, Association of IL-12p40 +1188 A/C polymorphism with nasopharyngeal cancer risk and tumor extension. Tissue Antigens 78:148–151 Chen X, Han S,Wang S et al, 2009, Interactions of IL-12A and IL- 12B polymorphisms on the risk of cervical cancer in Chinese women. Clin Cancer Res 15:400–405 Kordi Tamandani DM, Shekari M, Suri Vet al, 2009, Interleukin- 12 gene polymorphism and cervical cancer risk. Am J Clin Oncol Han SS, Cho EY, Lee TS et al, 2008, Interleukin-12 p40 gene, IL12B, polymorphisms and the risk of cervical caner in Korean women. Eur J Obstet Gynecol Reprod Biol 140:71–75 Noguchi Y, Jungbluth A, Richards EC et al, 1996, Effect of interleukin 12 on tumor induction by 3-methylcholanthrene. Proc Natl Acad Sci USA 93:11798–11801 Nanni P, Nicoletti G, De Giovanni C et al, 2001, Combined allogeneic tumor cell vaccination and systemic interleukin 12 prevents mammary carcinogenesis in HER-2/neu transgenic mice. J Exp Med 194:1195–1205 Bais AG, Beckmann I, Ewing PC et al, 2007, Cytokine release in HR-HPV(+, women without and with cervical dysplasia, CIN II and III, or carcinoma, compared with HR-HPV(−, controls. Mediators Inflamm 24147 Kadish AS, Timmins P, Wang Yet al, 2002, Regression of cervical intraepithelial neoplasia and loss of human papillomavirus, HPV, infection is associated with cell-mediated immune responses to an HPV type 16 E7 peptide. Cancer Epidemiol Biomark Prev 11:483– 488 Jin HS, Park EK, Lee JM et al, 2005, Immunization with adenoviral vectors carrying recombinant IL-12 and E7 enhanced the antitumor immunity to human papillomavirus 16-associated tumor. Gynecol Oncol 97:559–567 Indrova M, Bieblova J, Rossowska J et al, 2009, HPV 16- associated tumours: IL-12 can repair the absence of cytotoxic and proliferative responses of tumour infiltrating cells after chemotherapy. Int J Oncol 34:173–179 Mikyskova R, Indrova M, Simova J et al, 2011, Genetically modified tumour vaccines producing IL-12 augment chemotherapy of HPV16-associated tumours with gemcitabine. Oncol Rep 25:1683–1689 Chen T, Liang W, Gao L et al, 2011, Association of single nucleotide polymorphisms in interleukin 12, IL-12A and -B, with asthma in a Chinese population. Hum Immunol 72:603–606 Morris GA, Edwards DR, Hill PC et al, 2011, Interleukin 12B, IL12B, genetic variation and pulmonary tuberculosis: a study of cohorts from The Gambia, Guinea-Bissau, United States and Argentina. PLoS One 6:e16656 Morahan G, McKinnon E, Berry J et al, 2009, Type I diabetes genetics consortium. Evaluation of IL12B as a candidate type I diabetes susceptibility gene using data from the type I diabetes genetics consortium. Genes Immun 10(Suppl 1):S64–S68 Nair RP, Stuart PE, Kullavanijaya P et al, 2010, Genetic evidence for involvement of the IL23 pathway in Thai psoriatics. Arch Dermatol Res 302:139–143 Filer C, Ho P, Smith RL et al, 2008, Investigation of association of the IL12B and IL23R genes with psoriatic arthritis. Arthritis Rheum 58:3705–3709 Phawong C, Ouma C, Tangteerawatana P et al, 2010, Haplotypes of IL12B promoter polymorphisms condition susceptibility to severe malaria and functional changes in cytokine levels in Thai adults. Immunogenetics 62:345–356 Cerhan JR, Wang S, Maurer MJ et al, 2007, Prognostic significance of host immune gene polymorphisms in follicular lymphoma survival. Blood 109:5439–5446 Morahan G, Huang D, Ymer SI et al, 2001, Linkage disequilibrium of a type 1 diabetes susceptibility locus with a regulatory IL12B allele. Nat Genet 27:218–221 Davoodi-Semiromi A, Yang JJ, She JX, 2002, IL-12p40 is associated with type 1 diabetes in Caucasian-American families. Diabetes 51:2334–2336 Seegers D, Zwiers A, Strober Wet al, 2002, ATaqI polymorphism in the 3′UTR of the IL-12 p40 gene correlates with increased IL-12 secretion. Genes Immun 3:419–423 Yilmaz V, Yentur SP, Saruhan-Direskeneli G, 2005, IL-12 and IL- 10 polymorphisms and their effects on cytokine production. Cytokine 30:188–194 Guo T, Yang S, Liu N et al, 2011, Association study of interleukin- 12A gene polymorphisms with Graves’ disease in two Chinese populations. Clin Endocrinol, Oxf, 74:125–129 Chen Y, Zheng T, Lan Q et al, 2011, Cytokine polymorphisms in Th1/Th2 pathway genes, body mass index, and risk of non- Hodgkin lymphoma. Blood 117:585–590 Liu L, Xu Y, Liu Z et al, 2011, IL12 polymorphisms, HBV infection and risk of hepatocellular carcinoma in a high-risk Chinese population. Int J Cancer 128:1692–1696 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 997 EP 1002 DI 10.1007/s12253-012-9532-x PG 6 ER PT J AU Hanajiri, R Ohashi, K Hirashima, Y Kakihana, K Kobayashi, T Yamashita, T Sakamaki, H Akiyama, H AF Hanajiri, Ryo Ohashi, Kazuteru Hirashima, Yuka Kakihana, Kazuhiko Kobayashi, Takeshi Yamashita, Takuya Sakamaki, Hisashi Akiyama, Hideki TI Second Allogeneic Transplantation for Relapsed Acute Leukemia after Initial Allogeneic Hematopoietic Stem Cell Transplantation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Second allogeneic hematopoietic stem cell transplantation; Acute leukemia; Relapse; Salvage treatment ID Second allogeneic hematopoietic stem cell transplantation; Acute leukemia; Relapse; Salvage treatment AB We retrospectively reviewed the medical records of 45 patients with relapsed acute leukemia after initial allogeneic hematopoietic stem ell transplantation (allo-HSCT). Among 45 patients, a total of 11 patients eventually underwent second allo-HSCT (HSCT-2). Median survival after relapse was 294 days (range, 135–942 days) for HSCT-2. Multivariate analysis showed significantly better survival for recipients of second allo-HSCT than for other patients (hazard ratio, 4.38; 95 % confidence interval, 1.45–13.2; P00.009). Although outcomes for patients with relapsed leukemia were generally poor, our results suggest that second HSCT could offer a survival advantage over other conventional salvage strategies. C1 [Hanajiri, Ryo] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Ohashi, Kazuteru] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Hirashima, Yuka] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Diseases Center, Pharmacy DivisionTokyo, Japan. [Kakihana, Kazuhiko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kobayashi, Takeshi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Yamashita, Takuya] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Sakamaki, Hisashi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Akiyama, Hideki] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. RP Ohashi, K (reprint author), Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 113-8677 Tokyo, Japan. EM k.ohashi@cick.jp CR Greinix HT, 2002, DLI or second transplant. Ann Hematol 81, Suppl 2):S34–S35 Mehta J, Powles R, Kulkarni S et al, 1997, Induction of graftversus- host disease as immunotherapy of leukemia relapsing after allogeneic transplantation: single-center experience of 32 adult patients. Bone Marrow Transplant 20:129–135 Locatelli F, 1998, The role of repeat transplantation of haemopoietic stem cells and adoptive immunotherapy in treatment of leukaemia relapsing following allogeneic transplantation. Br J Haematol 102:633–638 Wolff SN, 2002, Second hematopoietic stem cell transplantation for the treatment of graft failure, graft rejection or relapse after allogeneic transplantation. Bone Marrow Transplant 29:545–552 Kono N, Ohashi K, Sasaki E et al, 2001, Second allogeneic peripheral blood stem cell transplantation with fludarabine-based low-intensity conditioning regimen for relapsed myelodysplastic syndrome after allogeneic bone marrow transplantation. Int J Hematol 73:122–125 Kono N, Ohashi K, Okuyama Yet al, 2001, Treatment of relapsing Ph+ acute lymphoblastic leukemia with donor leukocyte infusion followed by quantitative monitoring of residual disease. Hematology 6:261–265 Schmid C, Labopin M, Nagler A et al, 2007, Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party. J Clin Oncol 25:4938–4945 Mehta J, Powles R, Treleaven J et al, 1997, Outcome of acute leukemia relapsing after bone marrow transplantation: utility of second transplants and adoptive immunotherapy. Bone Marrow Transplant 19:709–719 Mrsic M, Horowitz MM, Atkinson K et al, 1992, Second HLAidentical sibling transplants for leukemia recurrence. Bone Marrow Transplant 9:269–275 Bosi A, Laszlo D, Labopin M et al, 2001, Second allogeneic bone marrow transplantation in acute leukemia: results of a survey by the European Cooperative Group for Blood and Marrow Transplantation. J Clin Oncol 16:3675–3684 Barrett AJ, Locatelli F, Treleaven JG et al, 1991, Second transplants for leukaemic relapse after bone marrow transplantation: high early mortality but favorable effect of chronic GVHD on continued remission. A report by the EBMT Leukaemia Working Party. Br J Haematol 79:567–574 Michallet M, Tanguy ML, Socie G et al, 2000, Second allogeneic haematopoietic stem cell transplantation in relapsed acute and chronic leukaemias for patients who underwent a first allogeneic bone marrow transplantation: a survey of the Societe Francaise de Greffe de moelle, SFGM). Br J Haematol 108:400–407 Kishi K, Takahashi S, Gondo H et al, 1997, Second allogeneic bone marrow transplantation for post-transplant leukemia relapse: results of a survey of 66 cases in 24 Japanese institutes. Bone Marrow Transplant 19:461–466 Pawson R, Potter MN, Theocharous P et al, 2001, Treatment of relapse after allogeneic bone marrow transplantation with reduced intensity conditioning, FLAG +/− Ida, and second allogeneic stem cell transplant. Br J Haematol 115:622–629 Cheng H, Ohashi K, Kurosawa S et al, 2005, Busulfan and melphalan as a conditioning regimen for second peripheral blood stem cell transplantation in relapsed acute lymphoblastic leukemia after initial transplantation with total-body irradiation. Haema 8:83–90 Giralt SA, Champlin RE, 1994, Leukemia relapse after allogeneic bone marrow transplantation: a review. Blood 84:3603–3612 Kumar L, 1994, Leukemia: management of relapse after allogeneic bone marrow transplantation. J Clin Oncol 12:1710–1717 Savani BN, Mielke S, Reddy N et al, 2009, Management of relapse after allo-SCT for AML and the role of second transplantation. Bone Marrow Transplant 44:769–777 Kurosawa S, Fukuda T, Tajima K et al, 2009, Outcome of 93 patients with relapse or progression following allogeneic hematopoietic cell transplantation. Am J Hematol 84:815–820 Oran B, Giralt S, Couriel D et al, 2007, Treatment of AML and MDS relapsing after reduced-intensity conditioning and allogeneic hematopoietic stem cell transplantation. Leukemia 21:2540–2544 Arellano ML, Langston A, Winton E et al, 2007, Treatment of relapsed acute leukemia after allogeneic transplantation: a single center experience. Biol Blood Marrow Transplant 13:116–123 Mielcarek M, Storer BE, Flowers ME et al, 2007, Outcomes among patients with recurrent high-risk hematologic malignancies after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 13:1160–1168 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1003 EP 1008 DI 10.1007/s12253-012-9535-7 PG 6 ER PT J AU Lv, H Li, Q Qiu, W Xiang, J Wei, H Liang, H Sui, A Liang, J AF Lv, Hongying Li, Qicai Qiu, Wengsheng Xiang, Jinyu Wei, Hongjun Liang, Hua Sui, Aihua Liang, Jun TI Genetic Polymorphism of XRCC1 Correlated with Response to Oxaliplatin-Based Chemotherapy in Advanced Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal neoplasms/genetics; Polymorphism; Oxaliplatin/drug therapy; X-ray repair cross complementing 1/genetics ID Colorectal neoplasms/genetics; Polymorphism; Oxaliplatin/drug therapy; X-ray repair cross complementing 1/genetics AB To examine the association between genetic polymorphisms of XRCC1 Arg399Gln(G→A) and response to oxaliplatin-based chemotherapy in advanced colorectal cancer. XRCC1 genotypes of totally 99 patients(37 stage III, 62 stage IV)with advanced colorectal cancer treated with oxaliplatin-based chemotherapy were detected by TaqMan-MGB probe allelic discrimination method. And clinical response of 62 patients in stage IV after 2 to 3 cycles of chemotherapy were evaluated. Also time to progress (TTP) of all patients were evaluated. Of the genotype frequencies in all patients, up to 52.53 % were G/G genotype, 9.09 % were A/A genotype, and 38.38 % were G/A genotype. The response rate (CR+PR) of 62 patients in stage IV was 61.29% (19/31). Patients with G/G genotype showed enhanced respond to chemotherapy compared to those with G/A+A/A (x205.6, P00.029; OR03.845, 95 %CI01.231~12.01, P00.018). Individuals with the G/G genotype had a TTP of 10.0 (8.88–11.12) months, those with the G/A+A/A genotype had an TTP of 5.0(4.26–5.74) months. The log-rank test was marginally significant (x2029.20, P<0.01). The Cox proportional hazards model, adjusted for stage, performance status, and chemotherapy regimen, showed that only XRCC1 G/G genotypes increases the OR significantly (OR03.555; 95 % CI, 2.119~5.963; P<0.01). The results suggest that XRCC1 Arg399Gln polymorphisms is associated with the response to oxaliplantin-based chemotherapy and time to progression in advanced colorectal cancer in Chinese population. It is proposed that the XRCC1 Arg399Gln polymorphism should be routinely detected to screen patients who are more likely benefit from oxaliplantin-based treatment. C1 [Lv, Hongying] The Affiliated Hospital of Qingdao University, 266003 Qingdao, CN, China. [Li, Qicai] The Affiliated Hospital of Qingdao University, 266003 Qingdao, CN, China. [Qiu, Wengsheng] The Affiliated Hospital of Qingdao University, 266003 Qingdao, CN, China. [Xiang, Jinyu] The Affiliated Hospital of Qingdao University, 266003 Qingdao, CN, China. [Wei, Hongjun] Qingdao Municipal Hospital, Department of Pathology, 266027 Qingdao, CN, China. [Liang, Hua] Qingdao Oncology Hospital, Department of Oncology, 266074 Qingdao, CN, China. [Sui, Aihua] The Affiliated Hospital of Qingdao University, 266003 Qingdao, CN, China. [Liang, Jun] The Affiliated Hospital of Qingdao University, 266003 Qingdao, CN, China. RP Liang, J (reprint author), The Affiliated Hospital of Qingdao University, 266003 Qingdao, China. EM qdliangjun@163.com CR Pisani P, Parkin DM, Bray F, Ferlay J, 1999, Estimates of the worldwide mortality from 25 cancers in 1990. Int J Cancer 83, 1):18–29., DOI 10.1002/(SICI)1097-0215(19990924)83:1<18:: AID-IJC5>3.0.CO;2-M Faivre S, Chan D, Salinas R, Woynarowska B, Woynarowski JM, 2003, DNA strand breaks and apoptosis induced by oxaliplatin in cancer cells. Biochem Pharmacol 66(2):225–237 Lentz F, Tran A, Rey E, Pons G, Treluyer JM, 2005, Pharmacogenomics of fluorouracil, irinotecan, and oxaliplatin in hepatic metastases of colorectal cancer: clinical implications. Am J Pharmacogenomics 5(1):21–33 Kang D, 2003, Genetic polymorphisms and cancer susceptibility of breast cancer in Korean women. J Biochem Mol Biol 36(1):28–34 Friedberg EC, 2001, How nucleotide excision repair protects against cancer. Nat Rev Cancer 1(1):22–33., DOI 10.1038/35094000 Tian M, Shinkura R, Shinkura N, Alt FW, 2004, Growth retardation, early death, andDNArepair defects inmice deficient for the nucleotide excision repair enzyme XPF. Mol Cell Biol 24(3):1200–1205 Mort R,Mo L, McEwan C, MeltonDW(2003, Lack of involvement of nucleotide excision repair gene polymorphisms in colorectal cancer. Br J Cancer 89(2):333–337., DOI 10.1038/sj.bjc.6601061 Motycka TA, Bessho T, Post SM, Sung P, Tomkinson AE, 2004, Physical and functional interaction between the XPF/ERCC1 endonuclease and hRad52. J Biol Chem 279(14):13634–13639., DOI 10.1074/jbc.M313779200 Nagasubramanian R, Innocenti F, Ratain MJ, 2003, Pharmacogenetics in cancer treatment. Annu Rev Med 54:437–452., DOI 10.1146/annurev.med.54.101601.152352 Chacko P, Rajan B, Joseph T, Mathew BS, Pillai MR, 2005, Polymorphisms in DNA repair gene XRCC1 and increased genetic susceptibility to breast cancer. Breast Cancer Res Treat 89(1):15– 21., DOI 10.1007/s10549-004-1004-x Kiyohara C, Takayama K, Nakanishi Y, 2006, Association of genetic polymorphisms in the base excision repair pathway with lung cancer risk: a meta-analysis. Lung Cancer 54(3):267–283., DOI 10.1016/j.lungcan.2006.08.009 Stern MC, Siegmund KD, Conti DV, Corral R, Haile RW, 2006, XRCC1, XRCC3, and XPD polymorphisms as modifiers of the effect of smoking and alcohol on colorectal adenoma risk. Cancer Epidemiol Biomarkers Prev 15(12):2384–2390., DOI 10.1158/1055- 9965.EPI-06-0381 Zhang Z, Miao XP, Tan W, Guo YL, Zhang XM, Lin DX, 2006, Correlation of genetic polymorphisms in DNA repair genes ADPRT and XRCC1 to risk of gastric cancer. Ai Zheng 25(1):7–10 Kim DY, Kim YB, Choi HS, Kim JM, Suh K, 2006, Which gene is a dominant predictor of response during FOLFOX chemotherapy for the treatment of metastatic colorectal cancer, MTHFR or XRCC1 gene? Ann Surg Oncol 13(2):7–7 Stoehlmacher J, Ghaderi V, Iobal S, Groshen S, Tsao-Wei D, Park D, Lenz HJ, 2001, A polymorphism of the XRCC1 gene predicts for response to platinum based treatment in advanced colorectal cancer. Anticancer Res 21(4B):3075– 3079 Liu B,Wei J, Zou Z, Qian X, Nakamura T, ZhangW, Ding Y, Feng J, Yu L, 2007, Polymorphism of XRCC1 predicts overall survival of gastric cancer patients receiving oxaliplatin-based chemotherapy in Chinese population. Eur J Hum Genet 15(10):1049–1053., DOI 10.1038/sj.ejhg.5201884 Gurubhagavatula S, Liu G, Park S, Zhou W, Su L, Wain JC, Lynch TJ, Neuberg DS, Christiani DC, 2004, XPD and XRCC1 genetic polymorphisms are prognostic factors in advanced non-small-cell lung cancer patients treated with platinum chemotherapy. J Clin Oncol 22(13):2594–2601., DOI 10.1200/JCO.2004.08.067JCO. 2004.08.067 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1009 EP 1014 DI 10.1007/s12253-012-9536-6 PG 6 ER PT J AU Sobczuk, A Poplawski, T Blasiak, J AF Sobczuk, Anna Poplawski, Tomasz Blasiak, Janusz TI Polymorphisms of DNA Repair Genes in Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE XRCC1; ERCC2; hOGG1; Endometrial cancer; RFLP-PCR; BER; NER ID XRCC1; ERCC2; hOGG1; Endometrial cancer; RFLP-PCR; BER; NER AB Endometrial cancer belongs to the commonest malignancy in females. Its development may be associated with the high exposure of endometrium to exo- and endogenous estrogens. Estrogens produce DNA bulky adducts and oxidative base damages which are removed in nucleotide excision repair (NER) and base excision repair (BER) pathways. The reaction of endometrial cells to DNA damage may be crucial for their susceptibility to cancer transformation. This reaction is executed mainly by DNA repair, which can be modulated by the variability in the genes encoding DNA repair proteins. In this report we genotyped 4 polymorphisms of 3 DNA repair genes in 94 endometrial cancer patients and 114 age-matched cancer-free women using RFLP-PCR. The following polymorphisms were studied: p.Arg194Trp, p.Arg399Gln of the XRCC1 gene, p.Ser326Cys of the hOGG1 gene and p.Lys751Gln of the ERCC2 gene. We found an association between the ERCC2 751Gln variant and endometrial cancer occurrence (OR 3.95; 95 % CI 1.88–8.31). Gene-gene interaction between the ERCC2 751Gln and XRCC1 194Trp variants also increased the risk of endometrial cancer (OR 4.41; 95 % CI 2.01–9.67). The risk in the carriers of the ERCC2 751Gln variant was increased by a positive cancer history in first degree relatives (OR 4.97; 95 % CI 1.98–12.48). The risk of endometrial cancer was not alter by polymorphism p.Ser326Cys of the hOGG1 gene. The 751 Lys/Gln polymorphism of the ERCC2 gene may be linkedwith endometrial cancer occurrence and its effect can be potentiated by variants of the XRCC1 gene or first degree relatives positive cancer history. C1 [Sobczuk, Anna] Medical University of Lodz, Department of Gynaecology and ObstetricsLodz, Poland. [Poplawski, Tomasz] University of Lodz, Department of Molecular GeneticsLodz, Poland. [Blasiak, Janusz] University of Lodz, Department of Molecular GeneticsLodz, Poland. RP Blasiak, J (reprint author), University of Lodz, Department of Molecular Genetics, Lodz, Poland. EM jblasiak@biol.uni.lodz.pl CR Prat J, Gallardo A, Cuatrecasas M, Catasus L, 2007, Endometrial carcinoma: pathology and genetics. Pathology 39:72–87 Oda K, Stokoe D, Taketani Y, McCormick F, 2005, High frequency of coexistent mutations of PIK3CA and PTEN genes in endometrial carcinoma. Cancer Res 65:10669–10673 Velasco A, Bussaglia E, Pallares J et al, 2006, PIK3CA gene mutations in endometrial carcinoma: correlation with PTEN and K-RAS alterations. Hum Pathol 37:1465–1472 Liehr JG, Fang WF, Sirbasku DA, Ari-Ulubelen A, 1986, Carcinogenicity of catechol estrogens in Syrian hamsters. J Steroid Biochem 24:353–356 Kohno T, Kunitoh H, Toyama K et al, 2006, Association of the OGG1-Ser326Cys polymorphism with lung adenocarcinoma risk. Cancer Sci 97:724–728 Pachouri SS, Sobti RC, Kaur P, Singh J, 2007, Contrasting impact of DNA repair gene XRCC1 polymorphisms Arg399Gln and Arg194Trp on the risk of lung cancer in the north-Indian population. DNA Cell Biol 26:186–191 Poplawski T, Arabski M, Kozirowska D et al, 2006, DNA damage and repair in gastric cancer–a correlation with the hOGG1 and RAD51 genes polymorphisms. Mutat Res 601:83–91 Yin J, Vogel U, Ma Y, Qi R, Sun Z, Wang H, 2007, The DNA repair gene XRCC1 and genetic susceptibility of lung cancer in a northeastern Chinese population. Lung Cancer 56:153–160 Hatt L, Loft S, Risom L et al, 2008, OGG1 expression and OGG1 Ser326Cys polymorphism and risk of lung cancer in a prospective study. Mutat Res 639:45–54 Yin J, Vogel U, Ma Y, Guo L,Wang H, Qi R, 2006, Polymorphism of the DNA repair gene ERCC2 Lys751Gln and risk of lung cancer in a northeastern Chinese population. Cancer Genet Cytogenet 169:27–32 De Ruyck K, Szaumkessel M, De Rudder I et al, 2007, Polymorphisms in base-excision repair and nucleotide-excision repair genes in relation to lung cancer risk. Mutat Res 631:101–110 Brewster AM, Jorgensen TJ, Ruczinski I et al, 2006, Polymorphisms of the DNA repair genes ERCC2, Lys751Gln, and XRCC1, Arg399Gln and Arg194Trp): relationship to breast cancer risk and familial predisposition to breast cancer. Breast Cancer Res Treat 95:73–80 Patel AV, Calle EE, Pavluck AL, Feigelson HS, ThunMJ, Rodriguez C, 2005, A prospective study of XRCC1, X-ray crosscomplementing group 1, polymorphisms and breast cancer risk. Breast Cancer Res 7:1168–1173 Abdel-Rahman SZ, El-Zein RA, 2000, The 399Gln polymorphism in the DNA repair gene XRCC1 modulates the genotoxic response induced in human lymphocytes by the tobacco-specific nitrosamine NNK. Cancer Lett 159:63–71 Vodicka P, Stetina R, Polakova V et al, 2007, Association of DNA repair polymorphisms with DNA repair functional outcomes in healthy human subjects. Carcinogenesis 28:657– 664 Wang Y, Spitz MR, Zhu Y, Dong Q, Shete S, Wu X, 2003, From genotype to phenotype: correlating XRCC1 polymorphisms with mutagen sensitivity. DNA Repair 2:901–908 Silva SN, Moita R, Azevedo AP et al, 2007, Menopausal age and XRCC1 gene polymorphisms: role in breast cancer risk. Cancer Detect Prev 31:303–309 Fan L, Fuss JO, Cheng QJ, Arvai AS, Hammel M, Roberts VA, Cooper PK, Tainer JA, 2008, XPD helicase structures and activities: insights into the cancer and aging phenotypes from XPD mutations. Cell 133:789–800 Stern MC, Conway K, Li Y, Mistry K, Taylor JA, 2006, DNA repair gene polymorphisms and probability of TP53 mutation in bladder cancer. Mol Carcinog 45:715–719 Applebaum KM, Karagas MR, Hunter DJ et al, 2007, Polymorphisms in nucleotide excision repair genes, arsenic exposure, and non-melanoma skin cancer in New Hampshire. Environ Health Perspect 115:1231–1236 Unal M, Guven M, Batar B, Ozaydin A, Sarici A, Devranoglu K, 2007, Polymorphisms of DNA repair genes ERCC2 and XRCC1 and risk of cataract development. Exp Eye Res 85:328–334 Martucci CP, Fishman J, 1993, P450 enzymes of estrogen metabolism. Pharmacol Ther 57:237–257 Stack DE, Byun J, Gross ML, Rogan EG, Cavalieri EL, 1996, Molecular characteristics of catechol estrogen quinones in reactions with deoxyribonucleosides. Chem Res Toxicol 9:851–859 Dwivedy I, Devanesan P, Cremonesi P, Rogan E, Cavalieri E, 1992, Synthesis and characterization of estrogen 2,3- and 3,4- quinones. Comparison of DNA adducts formed by the quinones versus horseradish peroxidase-activated catechol estrogens. Chem Res Toxicol 5:828–833 Moser J, Kool H, Giakzidis I, Caldecott K, Mullenders LH, Fousteri MI, 2007, Sealing of chromosomal DNA nicks during nucleotide excision repair requires XRCC1 and DNA ligase III alpha in a cellcycle- specific manner. Mol Cell 27:311–323 Mortusewicz O, Leonhardt H, 2007, XRCC1 and PCNA are loading platforms with distinct kinetic properties and different capacities to respond to multiple DNA lesions. BMC Mol Biol 8:81 Fan J, Otterlei M, Wong HK, Tomkinson AE, Wilson DM 3rd, 2004, XRCC1 co-localizes and physically interacts with PCNA. Nucleic Acids Res 32:2193–2201 Tuimala J, Szekely G, Gundy S, Hirvonen A, Norppa H, 2002, Genetic polymorphisms of DNA repair and xenobioticmetabolizing enzymes: role in mutagen sensitivity. Carcinogenesis 23:1003–1008 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1015 EP 1020 DI 10.1007/s12253-012-9537-5 PG 6 ER PT J AU Chuangui, Ch Peng, T Zhentao, Y AF Chuangui, Chen Peng, Tang Zhentao, Yu TI The Expression of High Mobility Group Box 1 is Associated with Lymph Node Metastasis and Poor Prognosis in Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE High mobility group box 1 (HMGB1); Esophageal squamous cell carcinoma (ESCC); Vascular endothelial growth factor C (VEGF-C); Lymphangiogenesis ID High mobility group box 1 (HMGB1); Esophageal squamous cell carcinoma (ESCC); Vascular endothelial growth factor C (VEGF-C); Lymphangiogenesis AB The objective is to explore the expression of high mobility group box 1 (HMGB1) in esophageal squamous cell carcinoma (ESCC) and its relationship with lymph node metastasis and the prognosis of patients as well as possible mechanism. The expression of HMGB1, vascular endothelial growth factor C (VEGF-C) and lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1) in ESCC tissues, which were obtained from 72 patients who underwent radical esophagectomy, was detected through immunohistochemistry, firstly. The correlations between HMGB1 and VEGF-C, and micro-lymphatic vessel density (MLD), and lymph node metastasis, and the prognosis of patients, were analyzed by statistic analysis. The plasmid of small interference RNA (siRNA) targeting HMGB1, giving siHMGB1, was transfected into exponentially growing KYSE150 human esophageal squamous cancer cells and the expression of HMGB1 mRNA and protein was observed by Real-time PCR and Western Blot and the expression of VEGF-C was examined by ELISA. HMGB1 expressed highly in the nuclei and cytoplasm of carcinoma cells as well as the extracellular space in ESCC and was associated with lymph node metastasis, MLD, the expression of VEGF-C, TNM stage and the prognosis of patients (P<0.05 or P<0.01). In vitro, siHMGB1 inhibited the expression of HMGB1 mRNA and protein and the secretion of VEGF-C in KYSE150 cells. In ESCC, HMGB1 expresses highly and affects the prognosis of patients through regulating the expression of VEGF-C to promote lymphangiogenesis and lymph node metastasis, and HMGB1 might serve as the marker of progression and potential target for anti-lymphangiogenesis therapy. C1 [Chuangui, Chen] Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Prevention and Therapy, 300060 Tianjin, China. [Peng, Tang] Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Prevention and Therapy, 300060 Tianjin, China. [Zhentao, Yu] Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Prevention and Therapy, 300060 Tianjin, China. RP Zhentao, Y (reprint author), Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Prevention and Therapy, 300060 Tianjin, China. EM tracycheng2008@yahoo.com.cn CR Duff SE, Li C, Jeziorska M, Kumar S, Saunders MP, Sherlock D, O’Dwyer ST, Jayson GC, 2003, Vascular endothelial growth factors C and D and lymphangiogenesis in gastrointestinal tract malignancy. Br J Cancer 89:426–30 Aurello P, Rossi Del Monte S, D’Angelo F, Cicchini C, Ciardi A, Bellagamba R, Ravaioli M, Ramacciato G, 2009, Vascular endothelial growth factor C and microvessel density in gastric carcinoma: correlation with clinicopathological factors. Our experience and review of the literature. Oncol Res 17:405–11 Shimoda H, Takahashi Y, Kajiwara T, Kato S, 2003, Demonstration of the rat lymphatic vessels using immunohistochemistry and in situ hybridization for 5′- nucleotidase. Biomed Res 24:51–7 Liu P, Chen W, Zhu H, Liu B, Song S, Shen W, Wang F, Tucker S, Zhong B, Wang D, 2009, Expression of VEGF-C correlates with a poor prognosis based on analysis of prognostic factors in 73 patients with esophageal squamous cell carcinomas. Jpn J Clin Oncol 39:644–50 Czura CJ, Wang H, Tracey KJ, 2001, Dual roles for HMGB1: DNA binding and cytokine. J Endotoxin Res 7:315–21 Yao X, Zhao G, Yang H, Hong X, Bie L, Liu G, 2010, Overexpression of high-mobility group box 1 correlates with tumor progression and poor prognosis in human colorectal carcinoma. J Cancer Res Clin Oncol 136:677–84 Palumbo R, Sampaolesi M, De Marchis F, Tonlorenzi R, Colombetti S, Mondino A, Cossu G, BianchiME, 2004, Extracellular HMGB1, a signal of tissue damage, induces mesoangioblast migration and proliferation. J Cell Biol 164:441–49 Gnanasekar M, Thirugnanam S, Ramaswamy K, 2009, Short hairpin RNA, shRNA, constructs targeting high mobility group box-1, HMGB1, expression leads to inhibition of prostate cancer cell survival and apoptosis. Int J Oncol 34:425–431 Tang D, Kang R, Zeh HJ 3rd, Lotze MT, 2010, High-mobility group box 1 and cancer. Biochim Biophys Acta 1799:131–40 Kusume A, Sasahira T, Luo Y, Isobe M, Nakagawa N, Tatsumoto N, Fujii K, Ohmori H, Kuniyasu H, 2009, Suppression of dendritic cells by HMGB1 is associated with lymph node metastasis of human colon cancer. Pathobiology 76:155–62 Moriwaka Y, Luo Y, Ohmori H, Fujii K, Tatsumoto N, Sasahira T, Kuniyasu H, 2010, HMGB1 attenuates anti-metastatic defense of the lymph nodes in colorectal cancer. Pathobiology 77:17–23 Kang HJ, Lee H, Choi HJ, Youn JH, Shin JS, Ahn YH, Yoo JS, Paik YK, Kim H, 2009, Non-histone nuclear factor HMGB1 is phosphorylated and secreted in colon cancers. Lab Invest 89:948–59 Schlueter C, Weber H, Meyer B, Rogalla P, Roser K, Hauke S, Bullerdiek J, 2005, Angiogenetic signaling through hypoxia HMGB1: an angiogenetic switch molecule. Am J Path 166:1259–63 Weidner N, Semple JP, Welch WR, Folkman J, 1991, Tumor angiogenesis and metastasis –correlation in invasive breast carcinoma. N Engl J Med 324:1–8 Chen C, Sun J, Liu G, Chen J, 2009, Effect of small interference RNA targeting HIF-1α mediated by rAAV combined L-ascorbate on pancreatic tumors in athymic mice. Pathol Oncol Res 15:109–14 Tuomela J, Valta M, Seppanen J, Tarkkonen K, Vaananen HK, Harkonen P, 2009, Overexpression of vascular endothelial growth factor C increases growth and alters the metastatic pattern of orthotopic PC-3 prostate tumors. BMC Cancer 9:362 Zeh HJ 3rd, Lotze MT, 2005, Addicted to death: invasive cancer and the immune response to unscheduled cell death. J Immunother 28:1–9 Ito N, DeMarco RA,Mailliard RB, Han J, Rabinowich H, Kalinski P, Stolz DB, Zeh HJ 3rd, Lotze MT, 2007, Cytolytic cells induce HMGB1 release from melanoma cell lines. J Leukoc Biol 81:75–83 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1021 EP 1027 DI 10.1007/s12253-012-9539-3 PG 7 ER PT J AU Palanimuthu, D Baskaran, N Silvan, S Rajasekaran, D Manoharan, Sh AF Palanimuthu, Duraisamy Baskaran, Nagarethinam Silvan, Simon Rajasekaran, Duraisamy Manoharan, Shanmugam TI Lupeol, A Bioactive Triterpene, Prevents Tumor Formation During 7,12-Dimethylbenz(a)anthracene Induced Oral Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Antioxidants; DMBA; Lipid peroxidation; Lupeol; Oral cancer ID Antioxidants; DMBA; Lipid peroxidation; Lupeol; Oral cancer AB The oral cancer chemopreventive efficacy of lupeol, a bioactive triterpene, was assessed by monitoring the tumor incidence and using the status of phase I and II xenobiotic metabolizing enzymes, lipid peroxidation and antioxidants as biochemical end points during 7,12-dimethylbenz( a)anthracene (DMBA) induced hamster buccal pouch carcinogenesis. Oral tumors were developed in the buccal pouch of golden Syrian hamsters by painting with 0.5 % DMBA three times a week for 14 weeks. Well differentiated oral squamous cell carcinoma with marked abnormalities in the status of biochemical markers were noticed in hamsters treated with DMBA alone. Oral administration of lupeol at a dose of 50 mg/kg bw completely inhibited the formation of oral tumors and restored the status of biochemical markers during DMBA induced oral carcinogenesis. The present study thus demonstrates the chemopreventive potential of lupeol in DMBA induced oral carcinogenesis. The chemopreventive potential of lupeol is probably due to its antioxidant or free radical scavenging property and modulating effect on phase I and II xenobiotic metabolizing enzymes in favour of the excretion of carcinogenic metabolites during DMBA induced hamster buccal pouch carcinogenesis. C1 [Palanimuthu, Duraisamy] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai nagar, 608 002 Tamil Nadu, India. [Baskaran, Nagarethinam] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai nagar, 608 002 Tamil Nadu, India. [Silvan, Simon] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai nagar, 608 002 Tamil Nadu, India. [Rajasekaran, Duraisamy] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai nagar, 608 002 Tamil Nadu, India. [Manoharan, Shanmugam] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai nagar, 608 002 Tamil Nadu, India. RP Manoharan, Sh (reprint author), Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, 608 002 Tamil Nadu, India. EM sakshiman@rediffmail.com CR Rastogi T, Devesa S, Mangtani P, Mathew A, Cooper N, Kao R, Sinha R, 2008, Cancer incidence rates among South Asians in four geographic regions: India, Singapore, UK and US. Int J Epidemiol 37:147–160 Petti S, Mohd M, Scully C, 2012, Revisiting the association between alcohol drinking and oral cancer in nonsmoking and betel quid non-chewing individuals. Cancer Epidemiol 36:1–6 Saman DM, 2012, A review of the epidemiology of oral and pharyngeal carcinoma: update. Head Neck Oncol 4:1 Manoharan S, Vasantha Selvan M, Silvan S, Baskaran N, Singh AK, Vinoth Kumar V, 2010, Carnosic acid: a potent chemopreventive agent against oral carcinogenesis. Chem Biol Interact 188:616–622 Bhuvaneswari V, Abraham SK, Nagini S, 2005, Combinatorial antigenotoxic and anticarcinogenic effects of tomato and garlic through modulation of xenobiotic-metabolizing enzymes during hamster buccal pouch carcinogenesis. Nutrition 21:726–731 Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB, 2010, Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 49:1603–1616 Manoharan S, Kolanjiappan K, Suresh K, Panjamurthy K, 2005, Lipid peroxidation & antioxidants status in patients with oral squamous cell carcinoma. Indian J Med Res 122:529–534 Anusuya C, Manoharan S, 2011, Antitumor initiating potential of rosmarinic acid in 7,12-dimethylbenz(a)anthracene-induced hamster buccal pouch carcinogenesis. J Environ Pathol Toxicol Oncol 30:199–211 Bradford PG, Awad AB, 2010, Modulation of signal transduction in cancer cells by phytosterols. Biofactors 36:241–247 Wu SB, Su JJ, Sun LH, Wang WX, Zhao Y, Li H, Zhang SP, Dai GH, Wang CG, Hu JF, 2010, Triterpenoids and steroids from the fruits of Melia toosendan and their cytotoxic effects on two human cancer cell lines. J Nat Prod 73:1898–1906 Saleem M, 2009, Lupeol, a novel anti-inflammatory and anticancer dietary triterpene. Cancer Lett 285:109–115 Margareth BCG, Miranda JS, 2009, Biological activities of Lupeol. Int J Biomed Pharm Sci 3:46–66 Nigam N, Prasad S, Shukla Y, 2007, Prevents effects of lupeol on DMBA induced DNA alkylation alkylation damage in mouse skin. Food Chem Biol 45:2331–2335 Saleem M, Afaq F, Adhami VM, Mukthar H, 2004, Lupeol modulates NF-Kappa B and P13K/Akt pathways and inhibits skin cancer in CD-1 mice. Oncogene 23:5203–5214 Nagaraj M, Sunitha S, Varalakshmi P, 2000, Effect of lupeol, a pentacyclic triterpene, on the lipid peroxidation and antioxidant status in rat kidney after chronic cadmium exposure. J Appl Toxicol 20:413–417 Saleem M, Kaur S, Kweon M, Adhami VM, Afaq F, Mukhtar H, 2005, Lupeol, a fruit based triterpene, induces apoptotic death of human pancreatic adenocarcinoma cells via inhibition of Ras signaling pathway. Carcinogenesis 26:1956–1964 Yagi K, 1987, Lipid peroxides and human diseases. Chem Phys Lipids 45:337–351 Ohkawa H, Ohishi N, Yagi K, 1979, Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 95:351–358 Kakkar P, Das B, Viswanathan PN, 1984, A modified spectrophotometric assay of superoxide dismutase. Indian J Biochem Biophys 21:130–132 Sinha AK, 1972, Colorimetric assay of catalase. Anal Biochem 47:389–394 Rotruck JT, Pope AL, Ganther HE, Swanson AB, Hafeman DG, Hoekstra WG, 1973, Selenium: biochemical role as a component of glutathione peroxidase. Science 179:588–590 Beutler E, Kelly BM, 1963, The effect of sodium nitrite on red cell GSH. Experientia 19:96–97 Tietze F, 1969, Enzymic method for quantitative determination of nanogram amounts of total and oxidized glutathione: applications to mammalian blood and other tissues. Anal Biochem 27:502–522 Desai ID, 1984, Vitamin E analysis methods for animal tissues. Methods Enzymol 105:138–147 Palan PR, Mikhail MS, Basu J, Romney SL, 1991, Plasma levels of antioxidant beta-carotene and alpha-tocopherol in uterine cervix dysplasias and cancer. Nutr Cancer 15:13–20 Omura T, Sato R, 1964, The carbon monoxide binding pigment of liver microsomes. J Biol Chem 239:2370–2378 Habig WH, Pabst MJ, Jakoby WBC, 1974, Glutathione-Stransferases: the first enzymatic step in mercapturic acid formation. J Biol Chem249:7130–7139 Carlberg I, Mannervik B, 1985, Glutathione reductase. Methods Enzymol 113:484–490 Lowry OH, Rosebrough NJ, Farr AL, Randall RJ, 1951, Protein measurement with Folin phenol reagent. J Biol Chem 193:265–275 Vidjaya Letchoumy P, Chandra Mohan KV, Stegeman JJ, Gelboin HV, Hara Y, Nagini S, 2008, Pretreatment with black tea polyphenols modulates xenobiotic-metabolizing enzymes in an experimental oral carcinogenesis model. Oncol Res 17:75–85 Manoharan S, Sindhu G, Vinothkumar V, Kowsalya R, 2012, Berberine prevents 7,12-dimethylbenz[a]anthracene-induced hamster buccal pouch carcinogenesis: a biochemical approach. Eur J Cancer Prev 21:182–192 Silvan S, Manoharan S, Baskaran N, Anusuya C, Karthikeyan S, Prabhakar MM, 2011, Chemopreventive potential of apigenin in 7,12-dimethylbenz(a)anthracene induced experimental oral carcinogenesis. Eur J Pharmacol 670:571–577 Hata K, Hori K, Takahashi S, 2003, Role of p38 MAPK in lupeolinduced B16 2 F2 mouse melanoma cell differentiation. J Biochem 134:441–445 Prasad S, Nigam N, Kalra N, Shukla Y, 2008, Regulation of signaling pathways involved in lupeol induced inhibition of proliferation and induction of apoptosis in human prostate cancer cells. Mol Carcinog 47:916–924 Vinothkumar V, Manoharan S, 2011, Chemopreventive efficacy of geraniol against 7,12-dimethylbenz[a]anthracene-induced hamster buccal pouch carcinogenesis. Redox Rep 16:91–100 Subapriya R, Velmurugan B, Nagini S, 2005, Modulation of xenobiotic-metabolizing enzymes by ethanolic neem leaf extract during hamster buccal pouch carcinogenesis. J Exp Clin Cancer Res 24:223–230 Wnek SM, Kuhlman CL, Camarillo JM, Medeiros MK, Liu KJ, Lau SS, Gandolfi AJ, 2011, Interdependent genotoxic mechanisms of monomethylarsonous acid: role of ROS- induced DNA damage and poly(ADP-ribose, polymerase-1 inhibition in the malignant transformation of urothelial cells. Toxicol Appl Pharmacol 257:1–13 Balakrishnan S, Menon VP, Manoharan S, 2008, Ferulic acid inhibits 7,12-dimethylbenz[a]anthracene-induced hamster buccal pouch carcinogenesis. J Med Food 11:693–700 Kowsalya R, Vishwanathan P, Manoharan S, 2011, Chemopreventive potential of 18beta-glycyrrhetinic acid: an active constituent of liquorice, in 7,12-dimethylbenz(a)anthracene induced hamster buccal pouch carcinogenesis. Pak J Biol Sci 14:619–626 Manikandan P, Letchoumy PV, Gopalakrishnan M, Nagini S, 2008, Evaluation of Azadirachta indica leaf fractions for in vitro antioxidant potential and in vivo modulation of biomarkers of chemoprevention in the hamster buccal pouch carcinogenesis model. Food Chem Toxicol 46:2332–2343 Yamashita K, Lu H, Lu J, Chen G, Yokoyama T, Sagara Y, Manabe M, Kodama H, 2002, Effect of three triterpenoids, lupeol, betulin, and betulinic acid on the stimulus-induced superoxide generation and tyrosyl phosphorylation of proteins in human neutrophils. Clin Chim Acta 325:91–6 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1029 EP 1037 DI 10.1007/s12253-012-9541-9 PG 9 ER PT J AU Li, B Ge, Z Song, Sh Zhang, Sh Yan, H Huang, B Zhang, Y AF Li, Bing Ge, Zhiping Song, Shipeng Zhang, Shengbin Yan, Hong Huang, Boyun Zhang, Yangde TI Decreased Expression of SOX7 is Correlated with Poor Prognosis in Lung Adenocarcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung adenocarcinoma; SOX7; Immunohistochemistry; Quantitative real-time reverse transcription polymerase chain reaction; Prognosis ID Lung adenocarcinoma; SOX7; Immunohistochemistry; Quantitative real-time reverse transcription polymerase chain reaction; Prognosis AB Lung adenocarcinoma is the most frequently histologic subtype and the most histologically heterogeneous form of lung cancer. De-regulation of Wnt/β-catenin signaling pathway is implicated in lung carcinogenesis. SOX7, as a member of high mobility group (HMG) transcription factor family, plays a role in the modulation of the Wnt/β- catenin signaling pathway. However, the expression pattern and clinicopathological significance of SOX7 in patients with lung adenocarcinoma is still unclear. To address this problem, the SOX7 mRNA expression was detected by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). Immunohistochemical studies were performed on 288 pairs of adjacent normal lung and lung adenocarcinoma tissues with complete follow-up records. Association of SOX7 protein expression with clinical outcomes was evaluated using the Kaplan-Meier method and a multivariate Cox proportional hazards regression model. SOX7 mRNA expression was significantly down-regulated in lung adenocarcinoma compared with matched adjacent normal tissues (P<0.001). SOX7 protein was expressed in the cytoplasm of lung adenocarcinoma cells in 106/288 (36.8 %) of cases, whereas its immunoreactivities were predominantly located in the cytoplasm of the adjacent normal tissues. The reduced SOX7 expression was correlated with poor differentiation (P00.002), lymph node metastasis (P00.011) and advanced TNM stage (P00.006). Regarding patient survival, the overall survival and the disease-free survival rates were both significantly lower in patients with SOX7-negative tumors than in those with SOX7-positive tumors (P00.018 and 0.013, respectively). Multivariate analysis using a Cox proportional-hazards model demonstrated that SOX7 expression status was an independent prognostic factor predicting the overall survival and the disease-free survival of patients with lung adenocarcinoma (P00.021 and 0.016, respectively).Our data suggest that the decreased expression of SOX7 is an important feature of lung adenocarcinoma. The expression level of SOX protein may be a useful prognostic marker for patients with lung adenocarcinoma. C1 [Li, Bing] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, Xiangya Road 87Changsha, Hunan, China. [Ge, Zhiping] the Third Affiliated Hospital of Inner Mongolia Medical College (Baogang Hospital of Inner Mongolia), Department of Cardiology, Shaoxian Road 20, Kun DistrictBaotou, Inner Mongolia, China. [Song, Shipeng] the Third Affiliated Hospital of Inner Mongolia Medical College (Baogang Hospital of Inner Mongolia), Department of Genery Surgery, Shaoxian Road 20, Kun DistrictBaotou, Inner Mongolia, China. [Zhang, Shengbin] the Third Affiliated Hospital of Inner Mongolia Medical College (Baogang Hospital of Inner Mongolia), Department of Genery Surgery, Shaoxian Road 20, Kun DistrictBaotou, Inner Mongolia, China. [Yan, Hong] the second Affiliated Hospital of Baotou Medical College, Department of CardiologyBaotou, Inner Mongolia, China. [Huang, Boyun] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, Xiangya Road 87Changsha, Hunan, China. [Zhang, Yangde] Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, Xiangya Road 87Changsha, Hunan, China. RP Zhang, Y (reprint author), Central South University, Xiangya Hospital, National Hepatobiliary and Enteric Surgery Research Center, Changsha, China. EM zydjs@sohu.com CR Okamoto J, Kratz JR, Hirata T, Mikami I, Raz D, Segal M, Chen Z, Zhou HM, Pham P, Li H, Beltran A, Ray MR, Koizumi K, Shimizu K, Jablons D, He B, 2011, Downregulation of EMX2 is associated with clinical outcomes in lung adenocarcinoma patients. Clin Lung Cancer 12:237–44 Travis WD, Brambilla E, Muller-Hermelink HK, Harris CC, 2004, World Health Organization International Histological Classification of Tumours. Pathology and genetics of tumors of the lung, pleura, thymus and heart. IARC Press, Lyon TravisWD, Brambilla E, NoguchiM, Nicholson AG, Geisinger KR, Yatabe Y, Beer DG, Powell CA, Riely GJ, Van Schil PE, Garg K, Austin JH, Asamura H, Rusch VW, Hirsch FR, Scagliotti G, Mitsudomi T, Huber RM, Ishikawa Y, Jett J, Sanchez-Cespedes M, Sculier JP, Takahashi T, Tsuboi M, Vansteenkiste J, Wistuba I, Yang PC, Aberle D, Brambilla C, Flieder D, Franklin W, Gazdar A, Gould M, Hasleton P, Henderson D, Johnson B, Johnson D, Kerr K, Kuriyama K, Lee JS,Miller VA, Petersen I, Roggli V, Rosell R, Saijo N, Thunnissen E, Tsao M, Yankelewitz D, 2011, International association for the study of lung cancer/american thoracic society/european respiratory society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol 6:244–85 Katoh Y, Katoh M, 2005, Comparative genomics on SOX2 orthologs. Oncol Rep 14:797–800 Hosking BM, Wyeth JR, Pennisi DJ, Wang SC, Koopman P, Muscat GE, 2001, Cloning and functional analysis of the Sryrelated HMG box gene, Sox18. Gene 262:239–47 Herpers R, van de Kamp E, Duckers HJ, 2008, Schulte-Merker: redundant roles for sox7 and sox18 in arteriovenous specification in zebrafish. Circ Res 102:12–15 Cermenati S,Moleri S, Cimbro S, Corti P, Del Giacco L, Amodeo R, Dejana E, Koopman P, Cotelli F, BeltrameM(2008, Sox18 and Sox7 play redundant roles in vascular development. Blood 111:2657–66 Gandillet A, Serrano AG, Pearson S, Lie-A-Ling M, Lacaud G, Kouskoff V, 2009, Sox7-sustained expression alters the balance between proliferation and differentiation of hematopoietic progenitors at the onset of blood specification. Blood 114:4813–22 Katoh M, 2002, Expression of human SOX7 in normal tissues and tumors. Int J Mol Med 9:363–8 Zhang Y, Huang S, Dong W, Li L, Feng Y, Pan L, Han Z, Wang X, Ren G, Su D, Huang B, Lu J, 2009, SOX7, down-regulated in colorectal cancer, induces apoptosis and inhibits proliferation of colorectal cancer cells. Cancer Lett 277:29–37 Guo L, Zhong D, Lau S, Liu X, Dong XY, Sun X, Yang VW, Vertino PM, Moreno CS, Varma V, Dong JT, Zhou W, 2008, Sox7 is an independent checkpoint for beta-catenin function in prostate and colon epithelial cells. Mol Cancer Res 6:1421–30 Gong YD, Dong MS, Lee SB, Kim N, Bae MS, Kang NS, 2011, A novel 3-arylethynyl-substituted pyrido[2,3,-b]pyrazine derivatives and pharmacophore model as Wnt2/β-catenin pathway inhibitors in non-small-cell lung cancer cell lines. Bioorg Med Chem 19:5639–47 Pacheco-Pinedo EC, Durham AC, Stewart KM, Goss AM, Lu MM, Demayo FJ, Morrisey EE, 2011, Wnt/β-catenin signaling accelerates mouse lung tumorigenesis by imposing an embryonic distal progenitor phenotype on lung epithelium. J Clin Invest 121:1935–45 Xu X, Sun PL, Li JZ, Jheon S, Lee CT, Chung JH, 2011, Aberrant Wnt1/β-catenin expression is an independent poor prognostic marker of non-small cell lung cancer after surgery. J Thorac Oncol 6:716–24 Teng Y, Wang X, Wang Y, Ma D, 2010, Wnt/beta-catenin signaling regulates cancer stem cells in lung cancer A549 cells. Biochem Biophys Res Commun 392:373–9 Takash W, Canizares J, Bonneaud N, Poulat F, Mattei MG, Jay P, Berta P, 2001, SOX7 transcription factor: sequence, chromosomal localisation, expression, transactivation and interference with Wnt signalling. Nucleic Acids Res 29:4274–83 Kormish JD, Sinner D, Zorn AM, 2010, Interactions between SOX factors and Wnt/beta-catenin signaling in development and disease. Dev Dyn 239:56–68 Ding D, Xu H, Liang Q, Xu L, Zhao Y, Wang Y, 2011, Overexpression of Sox2 in C3H10T1/2 cells inhibits osteoblast differentiation through Wnt and MAPK signalling pathways. Int Orthop. In press Xiang R, Liao D, Cheng T, Zhou H, Shi Q, Chuang TS, Markowitz D, Reisfeld RA, Luo Y, 2011, Downregulation of transcription factor SOX2 in cancer stem cells suppresses growth and metastasis of lung cancer. Br J Cancer 104:1410–7 Titulaer MJ, Klooster R, Potman M, Sabater L, Graus F, Hegeman IM, Thijssen PE, Wirtz PW, Twijnstra A, Smitt PA, van der Maarel SM, Verschuuren JJ, 2009, SOX antibodies in small-cell lung cancer and Lambert-Eaton myasthenic syndrome: frequency and relation with survival. J Clin Oncol 27:4260–7 Vural B, Chen LC, Saip P, Chen YT, Ustuner Z, Gonen M, Simpson AJ, Old LJ, Ozbek U, Gure AO, 2005, Frequency of SOX Group B, SOX1, 2, 3, and ZIC2 antibodies in Turkish patients with small cell lung carcinoma and their correlation with clinical parameters. Cancer 103:2575–83 Friedman RS, Bangur CS, Zasloff EJ, Fan L, Wang T,Watanabe Y, Kalos M, 2004, Molecular and immunological evaluation of the transcription factor SOX-4 as a lung tumor vaccine antigen. J Immunol 172:3319–27 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1039 EP 1045 DI 10.1007/s12253-012-9542-8 PG 7 ER PT J AU Guo, J Niu, R Huang, W Zhou, M Shi, J Zhang, L Liao, H AF Guo, JingJing Niu, Rui Huang, Wenhui Zhou, Mengliang Shi, Jixing Zhang, Luyong Liao, Hong TI Growth Factors from Tumor Microenvironment Possibly Promote the Proliferation of Glioblastoma-Derived Stem-like Cells in Vitro SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma stem-like cells; Proliferation; Microenvironment; Growth factors ID Glioblastoma stem-like cells; Proliferation; Microenvironment; Growth factors AB Glioblastoma multiformis a lethal brain glial tumor characterized by low survival and high recurrence, partially attributed to the glioblastoma stem cells according to recent researches. Microenvironment or niche in tumor tissue is believed to provide essential support for the aberrant growth of tumor stem cells. In order to explore the effect of growth factors in tumor microenvironment on glioblastoma stem cells behavior, glioblastoma-derived stem-like cells (GDSCs) were isolated from adult human glioblastoma specimen with antibody against surface marker CD133 and were co-cultured with various tumor cells including U87MG cells, unsorted glioblastoma tumor cells, CD133- cells and normal rat primary astrocytes. Results suggested that tumor cells could promote GDSCs proliferation while non-tumor cells could not, and several growth factors were exclusively detected in the coculture system with tumor cells. It was concluded that growth factors derived from tumor microenvironment possibly contributed to the uncontrolled proliferation of GDSCs. C1 [Guo, JingJing] China Pharmaceutical University, Jiangsu Center for Drug Screening, Neurobiology Lab, 24# Tong Jiaxiang road, 210009 Nanjing, China. [Niu, Rui] China Pharmaceutical University, Jiangsu Center for Drug Screening, Neurobiology Lab, 24# Tong Jiaxiang road, 210009 Nanjing, China. [Huang, Wenhui] China Pharmaceutical University, Jiangsu Center for Drug Screening, Neurobiology Lab, 24# Tong Jiaxiang road, 210009 Nanjing, China. [Zhou, Mengliang] Nanjing University, School of Medicine, Jinling Hospital, Department of NeurosurgeryNanjing, China. [Shi, Jixing] Nanjing University, School of Medicine, Jinling Hospital, Department of NeurosurgeryNanjing, China. [Zhang, Luyong] China Pharmaceutical University, Jiangsu Center for Drug Screening, Neurobiology Lab, 24# Tong Jiaxiang road, 210009 Nanjing, China. [Liao, Hong] China Pharmaceutical University, Jiangsu Center for Drug Screening, Neurobiology Lab, 24# Tong Jiaxiang road, 210009 Nanjing, China. RP Liao, H (reprint author), China Pharmaceutical University, Jiangsu Center for Drug Screening, Neurobiology Lab, 210009 Nanjing, China. EM liaohong56@hotmail.com CR Vescovi AL, Galli R, Reynolds BA, 2006, Brain tumour stem cells. Nature reviews 6(6):425–436 Oliver TG, Wechsler-Reya RJ, 2004, Getting at the root and stem of brain tumors. Neuron 42(6):885–888 DeAngelis LM, 2001, Brain tumors. N Engl J Med 344(2):114–123 Kondo T, Setoguchi T, Taga T, 2004, Persistence of a small subpopulation of cancer stem-like cells in the C6 glioma cell line. Proc Natl Acad Sci U S A 101(3):781–786 Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB, 2004, Identification of human brain tumour initiating cells. Nature 432(7015):396–401 Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB, 2003, Identification of a cancer stem cell in human brain tumors. Cancer Res 63(18):5821–5828 Pfenninger CV, Roschupkina T, Hertwig F, Kottwitz D, Englund E, Bengzon J, Jacobsen SE, Nuber UA, 2007, CD133 is not present on neurogenic astrocytes in the adult subventricular zone, but on embryonic neural stem cells, ependymal cells, and glioblastoma cells. Cancer Res 67(12):5727–5736 Campos LS, 2005, Beta1 integrins and neural stem cells: making sense of the extracellular environment. Bioessays 27(7):698–707 Ohlstein B, Kai T, Decotto E, Spradling A, 2004, The stem cell niche: theme and variations. Curr Opin Cell Biol 16(6):693–699 Spradling A, Drummond-Barbosa D, Kai T, 2001, Stem cells find their niche. Nature 414(6859):98–104 Watt FM, Hogan BL, 2000, Out of Eden: stem cells and their niches. Science 287(5457):1427–1430, New York, NY Moore KA, Lemischka IR, 2006, Stem cells and their niches. Science 311(5769):1880–1885, New York, NY Fuchs E, Tumbar T, Guasch G, 2004, Socializing with the neighbors: stem cells and their niche. Cell 116(6):769–778 Reynolds BA, Weiss S, 1992, Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science 255(5052):1707–1710, New York, NY McCarthy KD, de Vellis J, 1980, Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. The Journal of cell biology 85(3):890–902 Schwartz JP, Wilson DJ, 1992, Preparation and characterization of type 1 astrocytes cultured from adult rat cortex, cerebellum, and striatum. Glia 5(1):75–80 Tropepe V, Sibilia M, Ciruna BG, Rossant J, Wagner EF, van der Kooy D, 1999, Distinct neural stem cells proliferate in response to EGF and FGF in the developing mouse telencephalon. Dev Biol 208(1):166–188 Bellows CG, Aubin JE, 1989, Determination of numbers of osteoprogenitors present in isolated fetal rat calvaria cells in vitro. Dev Biol 133(1):8–13 Liao H, HuangW, Schachner M, Guan Y, Guo J, Yan J, Qin J, Bai X, Zhang L, 2008, Beta 1 integrin-mediated effects of tenascin-R domains EGFL and FN6-8 on neural stem/progenitor cell proliferation and differentiation in vitro. J Biol Chem 283(41):27927–27936 Shen Q, Goderie SK, Jin L, Karanth N, Sun Y, Abramova N, Vincent P, Pumiglia K, Temple S, 2004, Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science 304(5675):1338–1340, New York, NY Lendahl U, Zimmerman LB, McKay RD, 1990, CNS stem cells express a new class of intermediate filament protein. Cell 60, 4):585–595 Yin AH, Miraglia S, Zanjani ED, Almeida-Porada G, Ogawa M, Leary AG, Olweus J, Kearney J, Buck DW, 1997, AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood 90(12):5002–5012 Uchida N, Buck DW, He D, Reitsma MJ, Masek M, Phan TV, Tsukamoto AS, Gage FH, Weissman IL, 2000, Direct isolation of human central nervous system stem cells. Proc Natl Acad Sci U S A 97(26):14720–14725 Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN, 2006, Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444(7120):756–760 Eramo A, Ricci-Vitiani L, Zeuner A, Pallini R, Lotti F, Sette G, Pilozzi E, Larocca LM, Peschle C, De Maria R, 2006, Chemotherapy resistance of glioblastoma stem cells. Cell death and differentiation 13(7):1238–1241 Liu G, Yuan X, Zeng Z, Tunici P, Ng H, Abdulkadir IR, Lu L, Irvin D, Black KL, Yu JS, 2006, Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Molecular cancer 5:67 Piccirillo SG, Binda E, Fiocco R, Vescovi AL, Shah K, 2009, Brain cancer stem cells. Journal of molecular medicine. Berlin, Germany Wang J, Sakariassen PO, Tsinkalovsky O, Immervoll H, Boe SO, Svendsen A, Prestegarden L, Rosland G, Thorsen F, Stuhr L, Molven A, Bjerkvig R, Enger PO, 2008, CD133 negative glioma cells form tumors in nude rats and give rise to CD133 positive cells. Int J Cancer 122(4):761–768 Joo KM, Kim SY, Jin X, Song SY, Kong DS, Lee JI, Jeon JW, Kim MH, Kang BG, Jung Y, Jin J, Hong SC, Park WY, Lee DS, Kim H, Nam DH, 2008, Clinical and biological implications of CD133- positive and CD133-negative cells in glioblastomas. Laboratory investigation; a journal of technical methods and pathology 88, 8):808–815 Beier D, Hau P, Proescholdt M, Lohmeier A, Wischhusen J, Oefner PJ, Aigner L, Brawanski A, Bogdahn U, Beier CP, 2007, CD133(+, and CD133(−, glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Res 67(9):4010–4015 Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, Vescovi A, 2004, Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res 64(19):7011–7021 Buick RN, Minden MD, McCulloch EA, 1979, Self-renewal in culture of proliferative blast progenitor cells in acute myeloblastic leukemia. Blood 54(1):95–104 Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, Hamner B, Oh EY, Gaber MW, Finklestein D, Allen M, Frank A, Bayazitov IT, Zakharenko SS, Gajjar A, Davidoff A, Gilbertson RJ, 2007, A perivascular niche for brain tumor stem cells. Cancer cell 11, 1):69–82 Li L, Neaves WB, 2006, Normal stem cells and cancer stem cells: the niche matters. Cancer Res 66(9):4553–4557 Hansen K, Wagner B, Hamel W, Schweizer M, Haag F, Westphal M, Lamszus K, 2007, Autophagic cell death induced by TrkA receptor activation in human glioblastoma cells. J Neurochem 103, 1):259–275 Schmidt AL, de Farias CB, Abujamra AL, Kapczinski F, Schwartsmann G, Brunetto AL, Roesler R, 2009, BDNF and PDE4, but not the GRPR. Regulate Viability of Human Medulloblastoma Cells, J Mol Neurosci Gruber-Olipitz M, Strobel T, Kang SU, John JP, Grotzer MA, Slavc I, Lubec G, 2009, Neurotrophin 3/TrkC-regulated proteins in the human medulloblastoma cell line DAOY. Electrophoresis 30, 3):540–549 Bassili M, Birman E, Schor NF, Saragovi HU, 2009, Differential roles of Trk and p75 neurotrophin receptors in tumorigenesis and chemoresistance ex vivo and in vivo. Cancer chemotherapy and pharmacology Golestaneh N, Mishra B, 2005, TGF-beta, neuronal stem cells and glioblastoma. Oncogene 24(37):5722–5730 Petschnik AE, Fell B, Tiede S, Habermann JK, Pries R, Kruse C, Danner S A novel xenogeneic co-culture system to examine neuronal differentiation capability of various adult human stem cells. PloS one 6, 9):e24944 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1047 EP 1057 DI 10.1007/s12253-012-9543-7 PG 11 ER PT J AU Petrov, VS Malkhasyan, K Ulyanin, YM Abdrakhmanov, FE Khasanov, ShR AF Petrov, V Semion Malkhasyan, A. Karen Ulyanin, Yurievich Mikhail Abdrakhmanov, F E Khasanov, Shamilyevich Rustem TI The Value of the Preoperative FISH Test in Unscreened Bladder Cancer Patients with TUR Indications SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; Urothelial carcinoma; FISH; Prediction; Bladder washings ID Bladder cancer; Urothelial carcinoma; FISH; Prediction; Bladder washings AB Patients with bladder cancer are still requiring close follow up with frequent cystoscopies. This study aims to assess the FISH analysis, as a procedure capable of highlighting the hidden features of a tumor and helping to individualize treatment tactics. The bladder washings of 50 primary bladder cancer patients were taken prior to TURB and analyzed with the commercial FISH assay UroVysion®. All patients were divided into groups according to the maximum stage and grade of the tumor. The sensitivity of the method was 81.5 %, 91.7 % and 100 % for the Ta, T1 and T2 stage groups, respectively. For the G1, G2 and G3 groups the sensitivity was 70 %, 100 % and 100 %, respectively. In addition, the rate of detecting genetically abnormal cells was significantly higher in the T2 stage compared to the Ta and combined Ta+T1 groups, as well as in the G3 group compared to the G1 and G2 groups. The mean signal number from each chromosome insignificantly increased with the stage and grade of the tumor. The detection of <40 % genetically abnormal cells predicted the absence of muscle invasion and a G3 tumor with more then 90 % reliability. The FISH method is highly sensitive in early bladder cancer detection and is able to predict the morphological character of a tumor even before surgery. C1 [Petrov, V Semion] Kazan Cancer CenterKazan, Russian Federation. [Malkhasyan, A. Karen] N. N. Blokhin Russian Cancer Research Center RAMSMoscow, Russian Federation. [Ulyanin, Yurievich Mikhail] Kazan Cancer CenterKazan, Russian Federation. [Abdrakhmanov, F E] Kazan Cancer CenterKazan, Russian Federation. [Khasanov, Shamilyevich Rustem] Kazan Cancer CenterKazan, Russian Federation. RP Malkhasyan, K (reprint author), N. N. Blokhin Russian Cancer Research Center RAMS, Moscow, Russian Federation. EM karen.malkhasyan@gmail.com CR Davydov M, Aksel E, 2009, Statistics of the malignant neoplasm in Russia and CIS countries in 2007. J Blokhin RCRC RAMS 20, N3(77) Matveev B, Kudashev B, Bukharkin B, Romanov V, Rubanov Yu, 2000, The role of fluorescent control in improvement of surgical radicalism in superficial bladder cancer. Urology 3:22–24 World Health Organization. World cancer report, 2003. IARC Press, Lyon, France, 2003, p 229 Jones JS, 2006, DNA-based molecular cytology for bladder cancer surveillance. Urology 67:35–45 Cairns P, Shaw ME, Knowles MA, 1993, Initiation of bladder cancer may involve deletion of a tumour-suppressor gene on chromosome 9. Oncogene 8:1083–1085 Orlow I, Lacombe L, Hannon GJ, Serrano M, Pellicer I, Dalbagni G, Reuter VE, Zhang ZF, Beach D, Cordon-Cardo C, 1995, Deletion of the p16 and p15 genes in human bladder tumors. J Natl Cancer Inst 87:1524–1529 Sokolova IA, Halling KC, Jenkins RB, Burkhardt HM, Meyer RG, Seelig SA, King W, 2000, The development of a multitarget, multicolor fluorescence in Situ hybridization assay for the detection of urothelial carcinoma in urine. J Mol Diagn 2: 116–123 Bashkatov S, Sevankaeva A, Karyakin O, Mikhailova G, Golub E, 2008, Fluorescent hybridization in situ: practical application in oncourology. Oncourology 3:9–15 Vorobcova I, Vasileva Z, Shkolnik M, Timofeeva D, Odincova M, Gappoev M, Urbanskiy A, 2011, The use of fluorescent in situ hybridization on the urine sediment cells for diagnostics of bladder cancer and it’s recurrence. Oncourology 1:73–78 Bubendorf L, Grilli B, Sauter G et al, 2001, Multiprobe FISH for enhanced detection of bladder cancer in voided urine specimens and bladder washings. Am J Clin Pathol 116(1):79–86 Degtyar P, Neulander E, Zirkin H et al, 2004, Fluorescence in situ hybridization performed on exfoliated urothelial cell in patients with transitional cell carcinomas of the bladder. Urology 63, 2):398–401 Lokeshwar VB, Habuchi T, Grossman B, 2005, Bladder tumor markers beyond cytology: international consensus on bladder tumor markers. Urology 66(suppl):35–63 Placer J, Espinet B, Salido M et al, 2002, Clinical utility of a multiprobe FISH assay in voided urine specimens for detection of bladder cancer and its recurrences, compared with urinary cytology. Eur Urol 42(6):547–552 Sevankaev A, Lushnikov E, Karyakin O, Mikhailova G, Gorban N, Bashkatov S, Golub E, Shkavrova T, Cepenko V, 2008, Clinical use of the FISH method in early detection of superficial bladder cancer. Oncourology 4:61–65 Richter J, Jiang F, Gorog JP et al, 1997, Marker ganetic differences between atage pTa and stage pT1 papillary bladder cancer detected by comparative genomic hybridization. Cancer Res 57:2860–2864 Babayan A, Bashkatov S, Karyakin O, Teplov A, Golovashenko M, Shkarupo V, Zaletaev D, Nemcova D, 2009, Molecular-genetic markers as disease course predictors in superficial bladder cancer. Oncourology 3:19–24 Olumi AF, Tsai YC, Nichols PW et al, 1990, Allelic loss of chromosome 17p distinguishes high grade from low grade transitional cell carcinomas of the bladder. Cancer Res 50:7081–7083 Babjuk M, Oosterlinck W, Sylvester R, Kaasinen E, Bohle A, Palou-Redorta J, Roupret M, 2011, EAU guidelines on nonmuscle- invasive urothelial carcinoma of the bladder, the 2011 update. Eur Urol 59(6):997–1008 Brauers A, Buettner R, Jakse G, 2001, Second resection and prognosis of primary high risk superficial bladder cancer: is cystectomy often too early? J Urol 165(3):808–810 Dutta SC, Smith JA Jr, Shappell SB et al, 2001, Clinical under staging of high risk nonmuscle invasive urothelial carcinoma treated with radical cystectomy. J Urol 166:490–493 Herr HW, Donat MS, 2008, Quality control in transurethral resection of bladder tumors. BJU Int 102:1242–1246 Jakse G, Algaba MPU, Osterlinck W, 2004, A second-look TUR in T1 transitional cell carcinoma: why? Eur Urol 45(5):539–546 Herr HW, 1999, The value of a second transurethral resection in evaluating patients with bladder tumors. J Urol 162:74–76 Paner GP, Zehnder P, Amin AM, Husain AN, Desai MM, 2011, Urothelial neoplasms of the urinary bladder occurring in young adult and pediatric patients: a comprehensive review of literature with implications for patient management. Adv Anat Pathol 18, 1):79–89 Magi-Galluzzi C, Epstein JI, 2004, Urothelial papilloma of the bladder: a review of 34 de novo cases. Am J Surg Pathol 28, 12):1615–1620 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1059 EP 1066 DI 10.1007/s12253-012-9544-6 PG 8 ER PT J AU Jeong, HE Koo, HD Lee, HS Bang, BK Park, HE Seol, SJ Lee, YJ Pyo, SJ Kim, HD Lee, JH Oh, S AF Jeong, Haeng Eun Koo, Hoe Dong Lee, Hyuk Sang Bang, Bae Ki Park, Hye Eun Seol, Soo Ji Lee, Yong Ji Pyo, Soo Jung Kim, Hoon Dong Lee, Jin Hee Oh, Sukjoong TI Aggressive Classical Kaposi’s Sarcoma Mimicking Malignant Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Classical Kaposi’s sarcoma; Human herpesvirus-8; Lymphoma ID Classical Kaposi’s sarcoma; Human herpesvirus-8; Lymphoma AB Classical Kaposi’s sarcoma is an unusual multifocal neoplasm of vascular endothelial cell origin, and considered a less malignant, slowly-progressing tumor. Although visceral involvement is occasionally seen in HIV/AIDS patients with KS, tumor dissemination to visceral lymph nodes in classical KS is very rare. A 72-yearold woman without any other relevant past medical history presented with anorexia, weight loss, night sweats, and skin eruptions. As the rapid progression of cytopenias and lymphadenopathy were observed, bone marrow biopsy and imaging were performed. Positron emission tomography showed disseminated lymphadenopathy in the cervical, axillary, mediastinal, inguinal, and abdomino-pelvic nodal areas. Inguinal lymph node biopsy was compatible with KS, positive for CD31, CD34, and human herpesvirus-8 by immunohistochemical stain. We report a case of aggressive classical KS mimicking aggressive malignant lymphoma. C1 [Jeong, Haeng Eun] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Internal Medicine, 108 Pyung-Dong, Chongno-Gu, 110-746 Seoul, South Korea. [Koo, Hoe Dong] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Internal Medicine, 108 Pyung-Dong, Chongno-Gu, 110-746 Seoul, South Korea. [Lee, Hyuk Sang] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Internal Medicine, 108 Pyung-Dong, Chongno-Gu, 110-746 Seoul, South Korea. [Bang, Bae Ki] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Internal Medicine, 108 Pyung-Dong, Chongno-Gu, 110-746 Seoul, South Korea. [Park, Hye Eun] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Internal Medicine, 108 Pyung-Dong, Chongno-Gu, 110-746 Seoul, South Korea. [Seol, Soo Ji] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Internal Medicine, 108 Pyung-Dong, Chongno-Gu, 110-746 Seoul, South Korea. [Lee, Yong Ji] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Internal Medicine, 108 Pyung-Dong, Chongno-Gu, 110-746 Seoul, South Korea. [Pyo, Soo Jung] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of PathologySeoul, South Korea. [Kim, Hoon Dong] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of PathologySeoul, South Korea. [Lee, Jin Hee] University of Ulsan College of Medicine, Asan Medical Center, Department of PathologySeoul, South Korea. [Oh, Sukjoong] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Internal Medicine, 108 Pyung-Dong, Chongno-Gu, 110-746 Seoul, South Korea. RP Koo, HD (reprint author), Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Internal Medicine, 110-746 Seoul, South Korea. EM dhkoo.smc@gmail.com CR Kaposi M, 1982, Idiopathic multiple pigmented sarcoma of the skin. CA Cancer J Clin 32:342–347 Antman K, Chang Y, 2000, Kaposi’s Sarcoma. N Engl J Med 342:1027–1038 Friedman-Kien AE, Saltzman BR, 1990, Clinical manifestations of classical, endemic African, and epidemic AIDS-associated Kaposi’s sarcoma. J Am Acad Dermatol 22:1237–1250 Brenner B, Weissmann-Brenner A, Rakowsky E et al, 2002, Classical Kaposi sarcoma. Cancer 95:1982–1987 Lospalluti M, Mastrolonardo M, Loconsole F et al, 1995, Classical Kaposi’s sarcoma: a survey of 163 cases observed in Bari, South Italy. Dermatology 191:104–108 Patel RM, Goldblum JR, Hsi ED, 2004, Immunohistochemical detection of human herpes virus-8 latent nuclear antigen-1 is useful in the diagnosis of Kaposi sarcoma. Mod Pathol 17:456–460 Chang Y, Cesarman E, Pessin MS et al, 1994, Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science 266:1865–1869 Osman M, Kubo T, Gill J et al, 1999, Identification of Human Herpesvirus 8-Specific Cytotoxic T-Cell Responses. J Virol 73:6136–6140 Cheuk W, Wong KOY, Wong CSC et al, 2004, Immunostaining for Human Herpesvirus 8 latent nuclear Antigen-1 helps distinguish Kaposi Sarcoma from its mimickers. Am J Clin Pathol 121:335–342 Brenner B, Rakowsky E, Katz A et al, 1999, Tailoring treatment for classical Kaposi’s sarcoma: comprehensive clinical guidelines. Int J Oncol 14:1097–1102 Von Roenn JH, Cianfrocca M, 2001, Treatment of Kaposi’s sarcoma. Canc Treat Res 104:127–148 Di Lorenzo G, Kreuter A, Di Trolio R et al, 2008, Activity and safety of pegylated liposomal doxorubicin as first-line therapy in the treatment of non-visceral classic Kaposi’s sarcoma: a multicenter study. J Invest Dermatol 128:1578–1580 Brambilla L, Romanelli A, Bellinvia M et al, 2008, Weekly paclitaxel for advanced aggressive classic Kaposi sarcoma: experience in 17 cases. Br J Dermatol 158:1339–1344 Zustovich F, Lombardi G, Pastorelli D, 2009, Important role of gemcitabine in the treatment of classic Kaposi’s sarcoma. Tumori 95:562–563 Krown SE, 2007, AIDS-associated Kaposi’s sarcoma: is there still a role for interferon alfa? Cytokine Growth Factor Rev 18:395–402 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1067 EP 1069 DI 10.1007/s12253-012-9545-5 PG 3 ER PT J AU Literati-Nagy, Zs Tory, K Literati-Nagy, B Kolonics, A Vigh, L Vigh, L Mandl, J Szilvassy, Z AF Literati-Nagy, Zsuzsanna Tory, Kalman Literati-Nagy, Botond Kolonics, Attila Vigh, Laszlo Vigh, Laszlo Mandl, Jozsef Szilvassy, Zoltan TI A Novel Insulin Sensitizer Drug Candidate—BGP-15—Can Prevent Metabolic Side Effects of Atypical Antipsychotics SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Atypical antipsychotic drugs; Side effects; BGP-15; Hydroxylamine derivative; Insulin resistance; Weight gain; Hyperinsulinaemic euglycaemic glucose clamp ID Atypical antipsychotic drugs; Side effects; BGP-15; Hydroxylamine derivative; Insulin resistance; Weight gain; Hyperinsulinaemic euglycaemic glucose clamp AB Atypical antipsychotic drugs (AAPD) are widely used to treat severe psychiatric disorders, have well documented metabolic side effects such as disturbances in glucose metabolism, insulin resistance and weight gain. It has been shown that BGP-15, a hydroxylamine derivative with insulin sensitizing activity can prevent AAPD provoked fat accumulation in adipocyte cultures, and insulin resistance in animal experiments and in healthy volunteers. The aim of this study was to compare the preventive effect of BGP-15 with conventional oral antidiabetics on metabolic side effects of AAPDs. We found that BGP-15 that does not belong to either conventional insulin sensitizers or oral antidiabetics, is able to counteract insulin resistance and weight gain provoked by antipsychotic agents in rats while rosiglitazone and metformin were not effective in the applied doses. Our results confirm that BGP-15 is a promising new drug candidate to control the metabolic side effects of atypical antipsychotics. Data indicate that this rat model is suitable to analyze the metabolic side effects of AAPDs and the protective mechanism of BGP-15. C1 [Literati-Nagy, Zsuzsanna] Semmelweis University, Department of Medical Chemistry, Molecular Biology and PathobiochemistryBudapest, Hungary. [Tory, Kalman] N-Gene Research and Development LtdBudapest, Hungary. [Literati-Nagy, Botond] Drug Research Center LtdBalatonfured, Hungary. [Kolonics, Attila] N-Gene Research and Development LtdBudapest, Hungary. [Vigh, Laszlo] Mecsek Pharma Research LtdPecs, Hungary. [Vigh, Laszlo] Hungarian Academy of Sciences, Biological Research CenterSzeged, Hungary. [Mandl, Jozsef] Semmelweis University, Department of Medical Chemistry, Molecular Biology and PathobiochemistryBudapest, Hungary. [Szilvassy, Zoltan] University of Debrecen, Department of Pharmacology and Pharmacotherapy, 4032 Debrecen, Hungary. RP Tory, K (reprint author), N-Gene Research and Development Ltd, Budapest, Hungary. EM trklmn@gmail.com CR Gardner DM, Baldessarini RJ, Waraich P, 2005, Modern antipsychotic drugs: a critical overview. CMAJ 172:1703–1711 Dwyer DS, Donohoe D, 2003, Induction of hyperglycemia in mice with atypical antipsychotic drugs that inhibit glucose uptake. Pharmacol Biochem Behav 75:255–260 Cohen T, Sundaresh S, Levine F, 2012, Antipsychotics activate the TGFβ pathway effector SMAD3. Molecular Psychiatry., DOI 10.1038/mp.2011.186 American Diabetes Association, American Psychiatric Association, American Association of Clinical Endocrinologists, North American Association for the Study of Obesity, 2004, Consensus development conference on antipsychotic drugs and obesity and diabetes. J Clin Psychiatry 65:267–272 De Hert M, van Eyck D, De Nayer A, 2006, Metabolic abnormalities associated with second generation antipsychotics: fact or fiction? Development of guidelines for screening and monitoring. Int Clin Psychopharmacol 21(suppl 2):S11–5 Kurthy M, Mogyorosi T, Nagy K, Kukorelli T, Jednakovits A, Talosi L et al, 2002, Effect of BRX-220 against peripheral neuropathy and insulin resistance in diabetic rat models. Ann N Y Acad Sci 967:482–9 Literati-Nagy B, Kulcsar E, Literati-Nagy Z, Buday B, Petrfai E, Horvath T, Tory K, Kolonics A, Fleming A, Mandl J, Koranyi L, 2009, Improvement of insulin sensitivity by a novel drug, BGP- 15, in insulin-resistant patients: a proof of concept randomized double-blind clinical trial. Horm Metab Res 41:374–380 Tim Crul, Noemi Toth, Stefano Piotto, Peter Literati-Nagy, Kalman Tory, Pierre Haldimann, Bernadett Kalmar, Linda Greensmith, Zsolt Torok, Gabor Balogh, Imre Gombos, Federica Campana, Simona Concilio, Ferenc Gallyas, Gabor Nagy, Zoltan Berente, Burcin Gungor, Maria Peter, Attila Glatz, Akos Hunya, Zsuzsanna Literati-Nagy, Laszlo Vigh Jr., Femke Hoogstra-Berends, Andre Heeres, Irma Kuipers, Lizette Loen, Jean-Paul Seerden, Deli Zhang, Roelien A M Meijering, Robert H Henning, Bianca JJM Brundel, Harm H Kampinga, Laszlo Koranyi, Zoltan Szilvassy, Jozsef Mandl, Balazs Sumegi, Mark A Febbraio, Ibolya Horvath, Philip L Hooper, Laszlo Vigh, 2012, Hydroximic acid derivatives: pleiotrophic HSP coinducers restoring homeostasis and robustness. Curr. Phar. Des, in press). Gombos I, Crul T, Piotto S, Gungor B, Torok Z, Balogh G, Peter M, Slotte PJ, Pilbat AN, Hunya A, Toth N, Literati-Nagy Z, Vigh L Jr, Glatz A, Brameshuber M, Schutz GJ, Hevener A, Febbraio MA, Horvath I, Vigh L, 2011, Membrane-lipid therapy in operation: the HSP co-inducer BGP-15 activates stress signal transduction pathways by remodeling plasma membrane rafts. PLoS One 6, 12):e28818 Hargitai J, Lewis H, Boros I, Racz T, Fiser A, Kurucz I, Benjamin I, Vigh L, Penzes Z, Csermely P, Latchman DS, 2003, Bimoclomol, a heat shock protein co-inducer, acts by the prolonged activation of heat shock factor-1. Biochem Biophys Res Commun 307(3):689–95 Chung J, Nguyen AK, Henstridge DC, Holmes AG, Chan MH, Mesa JL, Lancaster GI, Southgate RJ, Bruce CR, Duffy SJ, Horvath I, Mestril R, Watt MJ, Hooper PL, Kingwell BA, Vigh L, Hevener A, Febbraio MA, 2008, HSP72 protects against obesity-induced insulin resistance. Proc Natl Acad Sci USA 105:1739–1744 Literati-Nagy B, Peterfai E, Kulcsar E, Literati-Nagy Z, Buday B, Tory K, Mandl J, Sumegi B, Fleming A, Roth J, Koranyi L, 2010, Beneficial effect of the insulin sensitizer, HSP co-inducer, BGP- 15 on olanzapine-induced metabolic disorders. Brain Res Bull 83:340–344 Literati-Nagy Z, Tory K, Literati-Nagy B, Kolonics A, Torok Z, Gombos I, Balogh G, Vigh L Jr, Horvath I, Mandl J, Sumegi B, Hooper PL, Vigh L, 2012, The HSP co-inducer BGP-15 can prevent the metabolic side effects of the atypical antipsychotics. Cell Stress Chaperon., DOI 10.1007/s12192-012-0327-5 Ota M, Mori K, Nakashima A, Kaneko YS, Takahashi H, Ota A, 2005, Resistance to excessive bodyweight gain in risperidoneinjected rats. Clin Exp Pharmacol Physiol 32:279–87 Ota M, Mori K, Nakashima A, Kaneko YS, Fujiwara K, Itoh M, Nagasaka A, Ota A, 2003, Peripheral injection of risperidone, an atypical antipsychotic, alters the bodyweight gain of rats. Clin Exp Pharmacol Physiol 30:513–4 Porszasz R, Legvari G, Pataki T, Szilvassy J, Nemeth J, Kovacs P, Paragh G, Szolcsanyi J, Szilvassy Z, 2002, Sensory nitrergic nature of the hepatic insulin sensitizing substance mechanism in conscious rabbits. Eur J Pharmacol 17(1–3):211–2, 17 Wallenstein S, Zucker CL, Fleiss JL, 1980, Some statistical methods useful in circulation research. Circ Res 47(1):1–9 Cope MB, Nagy TR, Fernandez JR, Geary N, Casey DE, Allison DB, 2005, Antipsychotic drug-induced weight gain: development of an animal model. Int J Obes 29(6):607–614 Arulmozhi DK, Dwyer DS, Bodhankar SL, 2006, Antipsychotic induced metabolic abnormalities: an interaction study with various PPAR modulators in mice. Life Sci 79(19):1865–72 Panariello F, De Luca V, de Bartolomeis A, 2011, Weight Gain, Schizophrenia and Antipsychotics: New Findings from Animal Model and Pharmacogenomic Studies. Schizophrenia Research and Treatment., DOI 10.1155/2011/459284 Basu A, Meltzer HY, 2006, Differential trends in prevalence of diabetes and unrelated general medical illness for schizophrenia patients before and after the atypical antipsychotic era. Schizophr Res 8:99 Wu R-R, Zhao J-P, Guo X-F, He Y-Q, Fang M-S, Guo W-B, Chen J-D, Li L-H, 2008, Lifestyle intervention and metformin for treatment of antipsychotic-induced weight gain. A randomized controlled trial. JAMA 299:185 Baptista T, Rangel N, Fernandez V, Carrizo E, Fakih Yamily El, Uzcategui E, Galeazzi T, Gutierrez MA, Servigna M, Davila A, Uzcategui M, Serrano A, Connell L, Beaulieu S, de Baptista EA, 2006, Metformin for prevention of weight gain and insulin resistance with olanzapine: A double-blind placebo-controlled trial. Can J Psychiatry 51:192 Henderson DC, Fan X, Sharma B, Copeland PM, Borba CP, Boxill R, Freudenreich O, Cather C, Eden Evins A, Goff DC, 2009, A double-blind, placebo-controlled trial of rosiglitazone for clozapine-induced glucose metabolism impairment in patients with schizophrenia. Acta Psychiatr Scand 119(6):457–65 Baptista T, Rangel N, El Fakih Y, Uzcategui E, Galeazzi T, Beaulieu S, Araujo de Baptista E, 2009, Rosiglitazone in the assistance of metabolic control during olanzapine administration in schizophrenia: a pilot double-blind, placebo-controlled, 12- week trial. Pharmacopsychiatry 42(1):14–9 Wolski K, Steven E, 2007, Nissen, Kathy Wolski, 2007, Effect of Rosiglitazone on the Risk of Myocardial Infarction and Death from Cardiovascular Causes. N Engl J Med 356:2457–2471 Halmosi R, Berente Z, Osz E, Toth K, Literati-Nagy P, Sumegi B, 2001, Effect of poly(ADP-ribose, polymerase inhibitors on the ischemia-reperfusion-induced oxidative cell damage and mitochondrial metabolism in Langendorff heart perfusion system. Mol Pharmacol 59:1497–505 Szabados E, Literati-Nagy P, Farkas B, Sumegi B, 2000, BGP-15, a nicotinic amidoxime derivate protecting heart from ischemia reperfusion injury through modulation of poly(ADP-ribose, polymerase. Biochem Pharmacol 59:937–45 Zhang D, Ke L, Mackovicova K, Van Der Want JJ, Sibon OC et al, 2011, Effects of different small HSPB members on contractile dysfunction and structural changes in a Drosophila melanogaster model for atrial Fibrillation. J Mol Cell Cardiol 51:381–389 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1071 EP 1076 DI 10.1007/s12253-012-9546-4 PG 6 ER PT J AU Yadamsuren, EA Nagy, Sz Pajor, L Lacza, Bogner, B AF Yadamsuren, Enkh-Amar Nagy, Szilvia Pajor, Laszlo Lacza, Agnes Bogner, Barna TI Characteristics of Advanced- and Non Advanced Sporadic Polypoid Colorectal Adenomas: Correlation to KRAS Mutations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sporadic colorectal adenomas; Villous architecture; KRAS mutation; Adenoma progression ID Sporadic colorectal adenomas; Villous architecture; KRAS mutation; Adenoma progression AB The malignant potential of colorectal adenomas highly correlates with their pathological characteristics, such as size, histology and grade of dysplasia. Currently, based on these parameters, adenomas are characterized as "nonadvanced or advanced" and patient surveillance is adjusted accordingly. The aim of this study was to investigate the correlation between the KRAS mutations and characteristics of non-advanced and advanced colorectal adenomas for predicting the risk of increased malignant potential of adenomas that may influence the decision to offer follow-up endoscopic surveillance. We used a mutagenic polymerase chain reaction – restriction fragment length polymorphism method to determine KRAS mutations in 164 colorectal sporadic polypoid adenomas (51 non-advanced-, 113 advanced adenomas) and in 40 early colorectal carcinomas. The method of mutation detection was validated according to recommendation for KRAS mutation testing in colorectal carcinoma of the European Quality Assurance Program. The limit of detection of the assay was 3 % mutated DNA with a good reproducibility. Evaluation of pathological characteristics was performed according to European Guidelines for Quality Assurance in Colorectal Cancer Screening and Diagnosis. The morphological parameters of the adenoma such as size, histology, grade of dysplasia are highly correlated with one another: an increasing adenoma size raised the proportion of villous histology and degree of dysplasia (all p<0.0001). KRAS mutations were detected in 31 % of the non-advanced adenomas, in 57.5 % of the advanced adenomas and in 62.5 % of the early carcinomas. Most mutations occurred at codon 12 rather than at codon 13 (72 %, 82 %, 76 % versus 22 %, 17 %, 24 %, respectively). There was no significant difference in association of KRAS mutation with age, gender, location among non-advanced-, and advanced adenomas and early carcinomas. KRAS mutation was found more often in tubulovillous and villous adenomas, whereas wild-type KRAS was observed more frequently in tubular adenomas (P<0.0001) and there was an increased prevalence of KRAS mutations in larger adenomas (P<0.0001). In this study KRAS mutation occurred with the same frequency in adenomas with low-grade (48 %) and high-grade (50 %) dysplasia. KRAS mutation is very strongly associated with a villous architecture and through villous component expansion, KRAS mutations may increase risk of tumor progression in sporadic colorectal polypoid adenomas. C1 [Yadamsuren, Enkh-Amar] University of Pecs, Department of Pathology, Szigeti str. 12, 7624 Pecs, Hungary. [Nagy, Szilvia] University of Pecs, Department of Neurology, 7623 Pecs, Hungary. [Pajor, Laszlo] University of Pecs, Department of Pathology, Szigeti str. 12, 7624 Pecs, Hungary. [Lacza, Agnes] University of Pecs, Department of Pathology, Szigeti str. 12, 7624 Pecs, Hungary. [Bogner, Barna] University of Pecs, Department of Pathology, Szigeti str. 12, 7624 Pecs, Hungary. RP Bogner, B (reprint author), University of Pecs, Department of Pathology, 7624 Pecs, Hungary. EM bogner.barna@gmail.com CR Jemal A, Siegel R, Ward E et al, 2008, Cancer statistics, 2008. CA Cancer J Clin 58(2):71–96 Center MM, Jemal A, Ward E, 2009, International trends in colorectal cancer incidence rates. Cancer Epidemiol Biomarkers Prev 18:1688–1694 Strul H, Kariv R, Leshno M et al, 2006, The prevalence rate and anatomic location of colorectal adenoma and cancer detected by colonoscopy in average-risk individuals aged 40-80 years. Am J Gastroenterol 101:255–262 Carl CS, Ulrich L, Martin RB, 1999, Sensitive detection of K-ras mutations augments diagnosis of colorectal cancer metastases in liver. Cancer Res 59:5169–5175 Nagorni A, Katic V, Zivkovic V, Stanojevic G, 2004, Advanced colorectal adenoma. Arch Oncol 12(Suppl 1):59–60 Anke HS, Beatriz C, Meike W et al, 2010, Identification of key genes for carcinogenic pathways associated with colorectal adenoma-to-carcinoma progression. Tumor biol 31:89–96 Barry ELR, Baron JA, Grau MV et al, 2006, K-ras mutations in incident Sporadic colorectal adenomas. Cancer 106:1036–1040 Einspahr JG, Martinez E et al, 2006, Associations of Ki-ras protooncogene mutation and p53 gene overexpression in sporadic colorectal adenomas with demographic and clinicopathologic characteristic. Cancer Epidemiol Biomarkers Prev 15:1443–1450 Fearon ER, Vogelstein B, 1990, A genetic model for colorectal tumorigenesis. Cell 61:759–767 Pretlow TP, Brasitus TA, Fulton NC et al, 1993, K-ras mutations in putative preneoplastic lesions in human colon. J Nat Cancer Inst 85:200–204 Quirke Ph, Risio M, Lambert R et al, 2011, Quality assurance in pathology in colorectal cancer screening and diagnosis - European recommendations. Virchows Arch 458(1):1–19., DOI 10.1007/ s00428-010-0977-6 Winawer SJ, Zauber AG, Ho MN, O’Brien MJ et al, 1993, Prevention of colorectal cancer by colonoscopic polypectomy. N Engl J Med 329:1977–1981 Vieth M, Quirke P, Lambert R et al, 2011, Annex to Quirke et al. Quality assurance in pathology in colorectal cancer screening and diagnosis: annotations of colorectal lesions. Virchows Arch 458, 1):21-–30 Segnan N, Patnick J, von Karsa L, 2010, European guidelines for quality assurance in colorectal cancer screening and diagnosis, 1st edn. Publications Office of the European Union, Luxembourg WHO, 2000, Pathology and genetics of tumours in the digestive system. Carcinoma of the colon and rectum. In: Hamilton SR, Aaltonen LA, eds, World health organization international histological classification of tumours, vol 2. IARC, Lyon, pp 105–119 Abdulamir AS, Hafidh RR, Mahdi LK et al, 2008, The interplay between p53 and p21 tumor suppressor proteins in the transformation of colorectal adenoma to carcinoma. Am J Immunol 4(2):14–22 Van Krieken JHJM, Jung A, Kirchner T et al, 2008, KRAS mutation testing for predicting response to anti-EGFR therapy for colorectal carcinoma: proposal for an European quality assurance programme. Virchow Arch 453:417–431 Amicarelli G, Adlerstein D, Shehi E, Wang F, Makrigiorgos GM, 2006, Genotype-specific signal generation based on digestion of 3-Way DNA junctions: application to KRAS variation detection. Clin Chem 52(10):1855–1863 Krypuy M, Newnham GM, Thomas DM et al, 2006, High resolution melting analysis for the rapid and sensitive detection of mutations in clinical samples: KRAS codon 12 and 13 mutations in non-small cell lung cancer. BMC. Cancer 6:295 Brink M, de Goeij AFPM, Weijenberg MP et al, 2003, K-ras oncogene mutations in sporadic colorectal cancer in The Netherlands Cohort Study. Carcinogenesis 24(4):703–710 Schoen RE, Gerber LD, Margulies C, 1997, The pathologic measurement of polyp size is preferable to the endoscopic estimate. Gastrointest Endosc 46:492–496 Tsai FC, Strum WB, 2011, Prevalence of advanced adenomas in small and diminutive colon polyps using direct measurement of size. Dig Dis Sci 56(8):2384–2388., DOI 10.1007/s10620-011-1598-x Rashid A, Zahurak M, Goodman SN, Hamilton SR, 1999, Genetic epidemiology of mutated K-ras proto-oncogene, altered suppressor genes, and microsatellite instability in colorectal adenomas. Gut 44:826–833 Vogelstein B, Fearon ER, Hamilton SR et al, 1988, Genetic alterations during colorectal-tumor development. N Engl J Med 319:525–532 Morris RG, Curtis LJ, Romanowski P et al, 1996, Ki-ras mutations in adenomas: A characteristic of cancer-bearing colorectal mucosa. J Pathol 180:357–363 Eide TJ, 1986, Risk of colorectal cancer in adenoma-bearing individuals within a defined population. Int J Cancer 15:173–176 McLellan EA, Owen RA, Stepniewska KA et al, 1993, High frequency of K-ras mutations in sporadic colorectal adenomas. Gut 34:392–396 Jass JR, Baker K, Zlolec I et al, 2006, Advanced colorectal polyps with the molecular and morphological features of serrated polyps and adenomas: concept of a’fusion’ pathway to colorectal cancer. Histopathology 49:121–131 Risio M, 2010, The natural history of adenomas. Best Practice & Research Clinical Gastroenterology 24:271–280 Appelman HD, 2008, Con: High-grade dysplasia and villous features should not be part of the routine diagnosis of colorectal adenomas. Am J Gastroenterol 103:1329–1331 Rex DK, Goldblum JR, 2008, Pro: Villous elements and highgrade dysplasia help guide post-polypectomy colonoscopic surveillance. Am J Gastroenterol 103:1327–1329 Odze R, 2008, A Balancing view: Pathologist-Clinician interaction is essential. Am J Gastroenterol 103:1331–1333 Nusko G, Sachse R, Mansmann U et al, 1997, K-RAS-2 gene mutations as predictors of metachronous colorectal adenomas. Scand J Gastroenterol 32:1035–1041 Wang JY, Wang YH, Jao SW et al, 2006, Molecular mechanisms underlying the tumorigenesis of colorectal adenomas: correlation to activated K-ras oncogene. Oncol Rep 16:1245–1252 O’Brien MJ, Yang S, Mack C et al, 2006, Comparison of microsatellite instability, CpG island methylation phenotype, BRAF and KRAS status in serrated polyps and traditional adenomas indicates separate pathways to distinct colorectal carcinoma end points. Am J Surg Pathol 30:1491–1501 Risio M, Malacarne D, Giaretti W, 2005, KRAS transitions and villous growth in colorectal adenomas. Cell Oncology 27:363–366 Ishii T, Notohara K, Umapathy A et al, 2011, Tubular adenomas with minor villous changes show molecular features characteristic of tubulovillous adenomas. Am J Surg Pathol 35:212–220 Saini SD, Kim HM, Schoenfeld P, 2006, Incidence of advanced adenomas at surveillance colonoscopy in patients with a personal history of colon adenomas: a meta-analysis and systematic review. Gastrointest Endosc 64:614–626 Calistri D, Rengucci C, Seymour I et al, 2005, Mutation analysis of p53, K-ras, and BRAF genes in colorectal cancer progression. J Cell Physiol 204:484–488 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1077 EP 1084 DI 10.1007/s12253-012-9547-3 PG 8 ER PT J AU Gravante, G Ong, LS West, K McGregor, A Maddern, JG Metcalfe, SM Lloyd, MD Dennison, RA AF Gravante, Gianpiero Ong, Ling Seok West, Kevin McGregor, Angus Maddern, J Guy Metcalfe, S Matthew Lloyd, M David Dennison, R Ashley TI Patterns of Histological Changes following Hepatic Electrolytic Ablation in an Ex-Vivo Perfused Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ex-vivo; Liver; Histology; Pathology; Electrolytic ablation ID Ex-vivo; Liver; Histology; Pathology; Electrolytic ablation AB Electrolytic ablation (EA) destroys the liver by releasing toxic radicles and producing modifications in the local pH without increasing the tissue temperature. We assessed the histological changes produced by EA using an ex-vivo perfused model. Five porcine livers were harvested, preserved in ice and reperfused for six hours in an extracorporeal circuit using autologous normothermic blood. One hour after reperfusion EA was performed and liver biopsies collected at the end of the experiments. The main necrotic zone consisted of coagulative necrosis, sinusoidal dilatation and haemorrhage with an unusual morphological pattern. The coagulative necrosis and haemorrhage affected mainly the peripheral area of the lobule with relative sparing of the area surrounding the centrilobular vein. Contrasting with this sinusoidal dilatation appeared to be more prominent in the centrilobular area. EA produces patterns of tissue destruction that have not been observed with the more commonly used thermal techniques. Further studies should obtain more information about the influence of adjacent biliary and vascular structures so that appropriate clinical trials can be designed. C1 [Gravante, Gianpiero] Leicester General Hospital, Department of Hepatobiliary and Pancreatic Surgery, Gwendolen Road, LE5 4PW Leicester, UK. [Ong, Ling Seok] Leicester General Hospital, Department of Hepatobiliary and Pancreatic Surgery, Gwendolen Road, LE5 4PW Leicester, UK. [West, Kevin] University Hospitals of Leicester NHS Trust, Department of HistopathologyLeicester, UK. [McGregor, Angus] University Hospitals of Leicester NHS Trust, Department of HistopathologyLeicester, UK. [Maddern, J Guy] The Queen Elizabeth Hospital, Department of Surgery, University of AdelaideWoodville, Australia. [Metcalfe, S Matthew] Leicester General Hospital, Department of Hepatobiliary and Pancreatic Surgery, Gwendolen Road, LE5 4PW Leicester, UK. [Lloyd, M David] Leicester General Hospital, Department of Hepatobiliary and Pancreatic Surgery, Gwendolen Road, LE5 4PW Leicester, UK. [Dennison, R Ashley] Leicester General Hospital, Department of Hepatobiliary and Pancreatic Surgery, Gwendolen Road, LE5 4PW Leicester, UK. RP Gravante, G (reprint author), Leicester General Hospital, Department of Hepatobiliary and Pancreatic Surgery, LE5 4PW Leicester, UK. EM ggravante@hotmail.com CR Robertson GS, Wemyss-Holden SA, Dennison AR et al, 1998, Experimental study of electrolysis-induced hepatic necrosis. Br J Surg 85(9):1212–1216 Wemyss-Holden SA, de la M Hall P, Robertson GS et al, 2000, The safety of electrolytically induced hepatic necrosis in a pig model. Aust N Z J Surg 70(8):607–612 Baxter PS, Wemyss-Holden SA, Dennison AR et al, 1998, Electrochemically induced hepatic necrosis: the next step forward in patients with unresectable liver tumours? Aust N Z J Surg 68, 9):637–640 Turler A, Schaefer H, Schaefer N et al, 2000, Experimental lowlevel direct current therapy in liver metastases: influence of polarity and current dose. Bioelectromagnetics 21(5):395–401 Wemyss-Holden SA, Dennison AR, Finch GJ et al, 2002, Electrolytic ablation as an adjunct to liver resection: experimental studies of predictability and safety. Br J Surg 89(5):579–585 Metcalfe MS, Mullin EJ, Texler M et al, 2007, The safety and efficacy of radiofrequency and electrolytic ablation created adjacent to large hepatic veins in a porcine model. Eur J Surg Oncol 33, 5):662–667 Fosh BG, Finch JG, Lea M et al, 2002, Use of electrolysis as an adjunct to liver resection. Br J Surg 89(8):999–1002 Goldberg SN, Gazelle GS, Compton CC et al, 2000, Treatment of intrahepatic malignancy with radiofrequency ablation: radiologicpathologic correlation. Cancer 88(11):2452–2463 Tamaki K, Shimizu I, Oshio A et al, 2004, Influence of large intrahepatic blood vessels on the gross and histological characteristics of lesions produced by radiofrequency ablation in a pig liver model. Liver Int 24(6):696–701 Gravante G, Ong SL, Metcalfe MS, et al. The effects of radiofrequency ablation on the hepatic parenchyma: Histological bases for tumor recurrences. Surgical oncology Feb 26 Gravante G, Ong SL, Metcalfe MS et al, 2008, Hepatic microwave ablation: a review of the histological changes following thermal damage. Liver Int 28(7):911–921 Vanagas T, Gulbinas A, Sadauskiene I et al, 2009, Apoptosis is activated in an early period after radiofrequency ablation of liver tissue. Hepato-Gastroenterology 56(93):1095–1099 Nijkamp MW, van der Bilt JD, de Bruijn MT et al, 2009, Accelerated perinecrotic outgrowth of colorectal liver metastases following radiofrequency ablation is a hypoxia-driven phenomenon. Ann Surg 249(5):814–823 Gravante G, Ong SL, Metcalfe MS et al, 2009, Experimental application of electrolysis in the treatment of liver and pancreatic tumours: Principles, preclinical and clinical observations and future perspectives. Surgical oncology 2009 Dec 31 Gravante G, Ong SL, Metcalfe MS et al, 2008, Effects of hypoxia due to isovolemic hemodilution on an ex vivo normothermic perfused liver model. J Surg Res 2008 Nov 6: [Epub ahead of print] Gravante G, Ong SL, Metcalfe MS et al. Changes in acid–base balance during electrolytic ablation in an ex vivo perfused liver model. Am J Surg May 5 Gravante G, Ong SL, Metcalfe MS et al, 2009, Cytokine response to ischemia/reperfusion injury in an ex vivo perfused porcine liver model. Transplant Proc 41(4):1107–1112 von Euler H, Olsson JM, Hultenby K et al, 2003, Animal models for treatment of unresectable liver tumours: a histopathologic and ultra-structural study of cellular toxic changes after electrochemical treatment in rat and dog liver. Bioelectrochem, Amsterdam, Netherlands, 59(1–2):89–98 Hinz S, Egberts JH, Pauser U et al, 2008, Electrolytic ablation is as effective as radiofrequency ablation in the treatment of artificial liver metastases in a pig model. J Surg Oncol 98, 2):135–138 Berry DP, Garcea G, Vanderzon P et al, 2004, Augmenting the ablative effect of liver electrolysis: using two electrodes and the pringle maneuver. J Invest Surg 17(2):105–112 von Euler H, Nilsson E, Olsson JM et al, 2001, Electrochemical treatment, EChT, effects in rat mammary and liver tissue. In vivo optimizing of a dose-planning model for EChT of tumours. Bioelectrochem, Amsterdam, Netherlands, 54, 2):117–124 Berendson J, Simonsson D, 1994, Electrochemical aspects of treatment of tissue with direct current. Eur J Surg Suppl 574:111–115 Li K, Xin Y, Gu Y et al, 1997, Effects of direct current on dog liver: possible mechanisms for tumor electrochemical treatment. Bioelectromagnetics 18(1):2–7 Bhardwaj N, Dormer J, Ahmad F et al. Microwave ablation of the liver: a description of lesion evolution over time and an investigation of the heat sink effect. Pathology Dec;43(7):725–731 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1085 EP 1089 DI 10.1007/s12253-012-9549-1 PG 5 ER PT J AU Terada, T AF Terada, Tadashi TI Primary Esophageal Small Cell Carcinoma with Brain Metastasis and with CD56, KIT, and PDGFRA Expressions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Terada, Tadashi] Shizuoka City Shimizu Hospital, Department of Pathology, Miyakami 1231 Shimizu-Ku, 424–8636 Shizuoka, Japan. RP Terada, T (reprint author), Shizuoka City Shimizu Hospital, Department of Pathology, 424–8636 Shizuoka, Japan. EM piyo0111jp@yahoo.co.jp CR Miettinen M, Lasota J, 2005, KIT, CD117): a review on expression in normal and neoplastic tissue, and mutations and their clinicopathologic correlation. Appl Immunohistochem Mol Morphol 13:205–220 Miettinen M, Lasota J, 2006, Gastrointestinal stromal tumor: review on morphology, molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med 130:1466–1478 Dow N, Giblen G, Sobin LH, Miettinen M, 2006, Gastrointestinal stromal tumors: differential diagnosis. Semin Diagn Pathol 23:111–119 Sihto H, Sarlomo-Rikara M, Tynnienen O, Tanner M, Andersson LC, Franssila K, Nupponen NN, Joensuu H, 2005, KIT and platelet-derived growth factor receptor alpha tyrosine kinase gene mutations and KIT amplifications in human solid tumors. J Clin Oncol 23:49–57 Koide N, Saito H, Suzuki A, Sato T, Koiwai K, Nakamura N, Miyagawa S, 2007, Clinicopathologic features and histochemical analuses of proliferative activity and angiogenesis in small cell carcinoma of the esophagus. J Gastroenterol 42:932–938 Terada T, Kawaguchi M, Furukawa K, Sekido Y, Osamura Y, 2002, Minute mixed ductal-endocrine carcinoma of the pancreas with predominant intraductal growth. Pathol Int 52:740–746 Terada T, Kawaguchi M, 2005, Primary clear cell adenocarcinoma of the peritoneum. Tohoku J Exp Med 271:271–275 Terada T, 2009, Gastrointestinal stromal tumor of the uterus: a case report with genetic analyses of c-kit and PDGFRA genes. Int J Gynecol Oncol 28:29–34 Terada T, 2009, Primary extragastrointestinal stromal tumor of the transverse mesocolon without c-kit mutations but with PDGFRA mutations. Med Oncol 26:233–237 Terada T, 2008, Primary multiple extragastrointestinal stromal tumors of the omentum with different mutations of c-kit gene. World J Gastroenterol 14:7256–7259 Terada T, 2009, Mediastinal seminoma with multiple KIT gene mutations. Pathology 41:695–697 Terada T, 2009, Primary small cell carcinoma of the mediastinum: a case report with immunohistochemical and molecular genetic analyses of KIT and PDGFRA genes. Med Oncol 26:247–250 Terada T, 2010, Primary small cell carcinoma of the ureter: a case report involving immunohistochemical and molecular genetic analyses of KIT and PDGFRA genes. Pathology 42:101–102 Terada T, 2009, Autopsy case of primary small cell carcinoma of the urinary bladder: KIT and PDGFRA expression and mutations. Pathol Int 59:247–250 LaPoint RJ, Bourne PA, Wang HL, Xu H, 2007, Coexpression of c-kit and bcl-2 in small cell carcinoma and large cell neuroendocrine carcinoma of the lung. Appl Immunohistochem Mol Morphol 15:401–406 Lopes-Martin A, Ballenstin C, Garcia-Carbonero R, Castano A, Lopez-Rios F, Lopes-Encuentra A, Sanchez-CespedesM, Castellano D, Bartolomes A, Cortes Funes H, Paz-Ares L, 2007, Prognostic value of KIT expression in small cell lung carcinoma. Lung Cancer 56:405–413 Micke P, BasralM, Faldum A, Bittinger F, Ronnstrand L, Blaukat A, Beeh KM, Oesch F, Fischer B, Buhl R, Hengstler JG, 2003, Characterization of c-kit expression in small cell lung carcinoma: prognostic and therapeutic implications. Clin Cancer Res 9:188–194 Camps C, Sirera R, Bremnes RM, Garde J, Safont MJ, Blasco A, Berrocal A, Sanchez JJ, Calabuig C, Marrorell M, 2006, Analysis of c-kit expression in small cell lung carcinoma: prevalence and prognostic implications. Lung Cancer 52:343–347 Rossi G, Cavazza A, Marchioni A, Miggaldi M, Bavierli M, Fracciolongo N, Petruzzelli S, Longo L, Tamberi S, Crino L, 2003, KIT expression in small cell carcinoma of the lung: effects of chemotherapy. Mod Pathol 16:1041–1047 Naeem M, Dahiya M, Clark JI, Creech SD, Alkin S, 2002, Analysis of c-kit protein expression in small-cell lung carcinoma and its implications for prognosis. Hum Pathol 33:1182– 1187 Mojica WD, Saxena R, Starostik P, Cheney RT, 2005, CD117+ small cell lung cancer lacks the asp 816 val point mutation in exon 17. Histopathology 47:517–522 Boldrini L, Ursino S, Gisfredi S, Faviana P, Donati V, Camcci T, Lucchi M, Mussi A, Basolo F, Pingitore R, Fontanini G, 2004, Expression and mutational status of c-kit in small-cell lung cancer: prognostic relevance. Clin Cancer Res 15:4101–4108 Burger H, Den Bakker MA, Stoter G, Verweij J, Nooter K, 2003, Lack of c-kit exon 11 activating mutations in c-kit/CD117-positive SCLC tumor specimens. Eur J Cancer 39:793–799 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1091 EP 1093 DI 10.1007/s12253-011-9374-y PG 3 ER PT J AU Wang, Ch Fu, H Zhao, FG Wang, J Shi, Y AF Wang, Chunmeng Fu, H Zhao, Fang Gong Wang, Jingmei Shi, Yongquan TI CT Scan is not Everything in the Evaluation of a Patient with Gastrointestinal Tumors (GIST) Under Imatinib Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Wang, Chunmeng] Fudan University Shanghai Cancer Center, Abdominal Department, 270 Dong’an Road, 200032 Shanghai, China. [Fu, H] Fudan University Shanghai Cancer Center, Abdominal Department, 270 Dong’an Road, 200032 Shanghai, China. [Zhao, Fang Gong] Fudan University Shanghai Cancer Center, Abdominal Department, 270 Dong’an Road, 200032 Shanghai, China. [Wang, Jingmei] Fudan University Shanghai Cancer Center, Department of Pathology, 270 Dong’an Road, 200032 Shanghai, China. [Shi, Yongquan] Fudan University Shanghai Cancer Center, Abdominal Department, 270 Dong’an Road, 200032 Shanghai, China. RP Shi, Y (reprint author), Fudan University Shanghai Cancer Center, Abdominal Department, 200032 Shanghai, China. EM shiyingqiang@yeah.net CR Stroszczynski C, Jost D, Reichardt P, Chmelik P, Gaffke G, Kretzschmar A et al, 2005, Follow-up of gastrointestinal stromal tumours(GIST, during treatment with imatinib mesylate by abdominal MRI. Eur Radiol 15:2448–2456 Demetri GD, von Mehren M, Blanke CD, Van den Abbeele AD, Eisenberg B, Roberts PJ et al, 2002, Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 347:472–480 Phongkitkarun S, Phaisanphrukkun C, Jatchavala J, Sirachainan E, 2008, Assessment of gastrointestinal stromal tumors with computed tomography following treatment with imatinib mesylate. World J Gastroenterol 14:892–898 Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L et al, 2000, New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 92:205–216 Choi H, Charnsangavej C, de Castro Faria S, Tamm EP, Benjamin RS, Johnson MM et al, 2004, CT Evaluation of the response of gastrointestinal stromal tumors after Imatinib Mesylate treatment: a quantitative analysis correlated with FDG PET findings. AJR Am J Roentgenol 183:1619–1628 Benjamin RS, Choi H, Macapinlac HA, Burgess MA, Patel SR, Chen LL et al, 2007, We should desist using RECIST, at Least in GIST. J Clin Oncol 25:1760–1764 Choi H, Charnsangavej C, Faria SC, Macapinlac HA, Burgess MA, Patel SR et al, 2007, Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: proposal of new computed tomography response criteria. J Clin Oncol 25:1753–1759 Jager PL, Gietema JA, van der Graaf WT, 2004, Imatinib mesylate for the treatment of gastrointestinal stromal tumours: best monitored with FDG PET. Nucl Med Commun 25:433–438 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1095 EP 1097 DI 10.1007/s12253-011-9445-0 PG 3 ER PT J AU Ozturk, GO AF Ozturk, Goruroglu Ozlem TI HLA Alleles in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Ozturk, Goruroglu Ozlem] Cukurova University, Faculty of Medicine, Department of Clinical Biochemistry, Tibbi Biyokimya AD, 01330 Adana, Turkey. RP Ozturk, GO (reprint author), Cukurova University, Faculty of Medicine, Department of Clinical Biochemistry, 01330 Adana, Turkey. EM ozlem_goruroglu@yahoo.com CR Mahmoodi M, Nahvi H, Mahmoudi M, Kasaian A, Mohagheghi MA, Divsalar K, Nahavandian B, Jafari A, Ansarpour B, Moradi B, Aghamohammadi A, Amirzargar A, 2011, HLA-DRB1,-DQA1 and -DQB1 Allele and Haplotype Frequencies in Female Patients with Early Onset Breast Cancer. Pathol Oncol Res [In press] Gun FD, Ozturk OG, Polat A, Polat G, 2011, HLA class-II allele frequencies in Turkish breast cancer patients. Med Oncol [In press] NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1099 EP 1099 DI 10.1007/s12253-012-9501-4 PG 1 ER PT J AU Ostwal, V Rekhi, B Noronha, V Basak, R Desai, BS Maheshwari, A Prabhash, K AF Ostwal, Vikas Rekhi, Bharat Noronha, Vanita Basak, Ranjan Desai, B Sangeeta Maheshwari, Amita Prabhash, Kumar TI Primitive Neuroectodermal Tumor of Ovary in a Young Lady, Confirmed with Molecular and Cytogenetic Results—A Rare Case Report with a Diagnostic and Therapeutic Challenge SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Ostwal, Vikas] Tata Memorial Hospital, Department of Medical Oncology, ParelMumbai, India. [Rekhi, Bharat] Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Division of Molecular Pathology, KhargarNavi Mumbai, India. [Noronha, Vanita] Tata Memorial Hospital, Department of Medical Oncology, ParelMumbai, India. [Basak, Ranjan] Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Division of Molecular Pathology, KhargarNavi Mumbai, India. [Desai, B Sangeeta] Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Division of Molecular Pathology, KhargarNavi Mumbai, India. [Maheshwari, Amita] Tata Memorial Hospital, Department of Surgical Oncology (Gynaecology), ParelMumbai, India. [Prabhash, Kumar] Tata Memorial Hospital, Department of Medical Oncology, ParelMumbai, India. RP Rekhi, B (reprint author), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Division of Molecular Pathology, Navi Mumbai, India. EM rekhi.bharat@gmail.com CR Enzinger FM, Weiss SW, 1995, Primitive neuroectodermal tumors and related lesions. In: Enzinger FM, Weiss SW, eds, Soft tissue tumors, 3rd edn. Mosby-Year Book, St Louis, pp 929–964 Grier HE, 1997, The Ewing family of tumors: Ewing’s sarcoma and primitive neuroectodermal tumors. Pediatr Clin North Am 44:991–1004 Aurias A, Rimbaut C, Buffe D, Dubousett MA, 1983, Chromosomal translocations in Ewing’s sacoma. New Eng J Med 309:496–497 Marley EF, Liapis H, Humphrey PA, 1997, Primitive neuroectodermal tumor of the kidney-another enigma: a pathologic, immunohistochemical, and molecular diagnostic study. Am J Surg Pathol 21:354–359 Rekhi B, Qureshi S, Basak R, Menon S, Desai S, Medhi S, Kurkure P, Maheshwair AM, Jambhekar NA, 2010, A primary vaginal Ewing’s sarcoma/Primitive neuroectodermal tumor, PNET, in a young girl with t, 11; 22,, q24; q12,, EWSR1- FLI1, positive results: a case report. J Med Case Report 4:88 Lawlor ER, Murphy JI, Sorensen PH, Fryer CJ, 1997, Metastatic primitive neuroectodermal tumour of the ovary: successful treatment with mega-dose chemotherapy followed by peripheral blood progenitor cell rescue. Med Pediatr Oncol 29:308–312 Kawauchi S, Fukuda T, Miyamoto S, Yoshioka J, Shirahama S, Saito T, Tsukamoto N, 1998, Peripheral primitive neuroectodermal tumor of the ovary confirmed by CD99 immunostaining, karyotypic analysis, and RT-PCR for EWS/FLI-1 chimeric mRNA. Am J Surg Pathol 22:1417–1422 Shigeto K, Toshiro F, Shingo M, Jun-Ichi Y, Syuya S, Toshiaki S, 1998, Peripheral primitive neuroectodermal tumor of the ovary confirmed by CD99 immunostaining, karyotypic analysis and RTPCR for EWS/FLI-1 chimeric mRNA. Am J Surg Pathol 22:1417– 1422 Rangan A, Lobo FD, Rao AA, 2003, Primary primitive neuroectodermal tumor of the ovary: a case report. Indian J Pathol Microbiol 46:58–59 Kim KJ, Jang BW, Lee SK, Kim BK, Nam SL, 2004, A case of peripheral primitive neuroectodermal tumor of the ovary. Int J Gynecol Cancer 14:370–372 Ateser G, Yildiz O, Leblebici C, Mandel NM, Unal F, Turna H, Arikan I, Colcaki D, 2007, Metastatic primitive neuroectodermal tumor of the ovary in pregnancy. Int J Gynecol Cancer 17:266–269 Chow SN, Lin MC, Shen J, Wang S, Jong YJ, Chien CH, 2004, Analysis of chromosome abnormalities by comparative genomic hybridization in malignant peripheral primitive neuroectodermal tumor of the ovary. Gynecol Oncol 92:752–760 Bridge RS, Rajaram V, Dehner LP, Pfeifer JD, Perry A, 2006, Molecular diagnosis of Ewing sarcoma/primitive neuroectodermal tumor in routinely processed tissue: a comparison of two FISH strategies and RT-PCR in malignant round cell tumors. Mod Pathol 19:1–8 Kleinman GM, Young RH, Scully RE, 1993, Primary neuroectodermal tumors of the ovary. A report of 25 cases. Am J Surg Pathol 17:764–778 Anfinan NM, Sait KH, Al-Maghrabi JA, 2008, Primitive neuroectodermal tumor of the ovary. Saudi Med J 29:444–446 Ambros IM, Ambros PF, Strehl S, Kovar H, Gardner H, Salzer- Kuntschik M, 1991, MIC2 is a specific marker for Ewing’s sarcoma and peripheral primitive neuroectodermal tumors. Cancer 67:1886–1893 Gordon MD, Corless C, Renshaw AA, Beckstead J, 1998, CD99, keratin, and vimentin staining of sex cord-stromal tumors, normal ovary, and testis. Mod Pathol 11:769–773 Lin PP, Brody RI, Hamelin AC, Bradner JE, Healey JH, Ladanyi M, 1999, Differential transactivation by alternative EWS-FLI1 fusion proteins correlates with clinical heterogeneity in Ewing’s sarcoma. Cancer Res 59:1428–1432 Giovannini M, Biegel JA, Serra M, Wang JY, Wei YH, Nycum L, Emanuel BS, Evans GA, 1994, EWS-erg and EWS-Fli1 fusion transcripts in Ewing’s sarcoma and primitive neuroectodermal tumors with variant translocations. J Clin Invest 94:489–496 Kovar H, Aryee DN, Jug G, Henockl C, Schemper M, Delattre O, Thomas G, Gadner H, 1996, EWS/FLI-1 antagonists induce growth inhibition of Ewing tumor cells in vitro. Cell Growth Differ 7:429–437 Toretsky JA, Connell Y, Neckers L, Bhat NK, 1997, Inhibition of EWS-FLI-1 fusion protein with antisense oligodeoxynucleotides. J Neurooncol 31:9–16 Tanaka K, Iwakuma T, Harimaya K, Sato H, Iwamoto Y, 1997, EWS-Fli1 antisense oligodeoxynucleotide inhibits proliferation of human Ewing’s sarcoma and primitive neuroectodermal tumor cells. J Clin Invest 99:239–247 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2012 VL 18 IS 4 BP 1101 EP 1106 DI 10.1007/s12253-012-9503-2 PG 6 ER PT J AU Rusz, O Kahan, Zs AF Rusz, Orsolya Kahan, Zsuzsanna TI Bone Homeostasis and Breast Cancer: Implications for Complex Therapy and the Maintenance of Bone Integrity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Breast cancer; Bisphosphonates; Denosumab; Bone; Prevention ID Breast cancer; Bisphosphonates; Denosumab; Bone; Prevention AB The standard of care in bone metastases is antiresorptive therapy. If present in the bone, tumor cells induce a vicious cycle by stimulating the osteoclasts, which further accelerates tumor progression. The widely-used bisphosphonates or the new therapeutic option, denosumab an inhibitor of the receptor activator of NF-κB ligand (RANKL), interrupt this vicious cycle, inhibit tumor growth, and in clinical practice prevent skeleton-related events. Adjuvant oncological therapy, including chemotherapy and endocrine manipulations (ovarian ablation and tamoxifen in premenopausal, and aromatase inhibitors in postmenopausal women), increases the bone turnover and the risk of fracture. Awareness is essential for the diagnosis and treatment of cancer therapyinduced bone loss, or its prevention with appropriate calcium and vitamin D supplementation. A new possibility has been suggested for the prevention of relapse: the use of bisphosphonates in the adjuvant setting. Three large studies and their meta-analyses indicate that the inhibition of bone remodeling prevents the growth of dormant tumor cells and cancer relapse in the population of postmenopausal patients with a lowestrogen environment in the skeleton. The similar potential of a RANKL inhibitor is currently under evaluation. Since the maintenance of bone integrity is necessary for the prevention of both therapy-related side-effects and progression of the disease, the management of breast cancer at any stage requires a careful consideration of the bone homeostasis. C1 [Rusz, Orsolya] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. RP Kahan, Zs (reprint author), University of Szeged, Department of Oncotherapy, 6720 Szeged, Hungary. EM kahan.zsuzsanna@med.u-szeged.hu CR Van Poznak CH, Temin S, Yee GC et al, 2011, American society of clinical oncology executive summary of the clinical practice guideline update on the role of bone-modifying agents in metastatic breast cancer. J Clin Oncol 29:1221–1227 Guise T, 2010, Examining the metastatic niche: targeting the microenvironment. Semin Oncol 37:S2–S14 Yoneda T, Hiraga T, 2005, Crosstalk between cancer cells and bone microenvironment in bone metastasis. Biochem Biophys Res Commun 328:679–687 Karen MB, Carol VG, Andrea MM, 2008, The bone microenvironment in metastasis; what is special about bone? Cancer Metastasis Rev 27:41–55 Rogers MJ, Crockett JC, Coxon FP, Monkkonen J, 2011, Biochemical and molecular mechanisms of action of bisphosphonates. Bone 49:34–41 Clezardin P, Benzaid I, Croucher PI, 2011, Bisphosphonates in preclinical bone oncology. Bone 49:66–70 Kimachi K, Kajiya H, Nakayama S et al, 2011, Zoledronic acid inhibits RANK expression and migration of osteoclast precursors during osteoclastogenesis. N Schmied Arch Pharmacol 383:297– 308 Serge C, Socrates P, 2011, Pharmacology of bisphosphonates. Bone 49:42–49 Arboleya L, Alperi M, Alonso S, 2011, Adverse effects of bisphosphonates. Rheumatol Clin 7:189–197 Ross JR, Saunders Y, Edmonds PMet al, 2003, Systematic review of role of bisphosphonates on skeletal morbidity in metastatic cancer. BMJ http://www.bmj.com/content/327/7413/469. Accessed 30 August 2003 Wong MHF, Stockler MR, Pavlakis N, 2012, Bisphosphonates and other bone agents for breast cancer. Cochrane Database Syst Rev http:// onlinelibrary.wiley.com/doi/10.1002/14651858.CD003474.pub3/full. Accessed 30 April 2011 Costa L, Major PP, 2009, Effect of bisphosphonates on pain and quality of life in patients with bones metastases. Nat Clin Pract Oncol 6:163–174 Clemons M, Dranitsaris G, Ooi W, Cole DEC, 2008, A phase II trial evaluating the palliative benefit of second-line oral ibandronate in breast cancer patients with either a skeletal related event, SRE, or progressive bone metastases, BM, despite standard bisphosphonate, BP, therapy. Breast Canc Res Treat 108:79–85 Clemons MJ, Dranitsaris G, Ooi WS et al, 2006, Phase II trial evaluating the palliative benefit of second-line zoledronic acid in breast cancer patients with either a skeletal-related event or progressive bone metastases despite first-line bisphosphonate therapy. J Clin Oncol 24:4895–4900 Hillner BE, Ingle JN, Chlebowski RT et al, 2003, American society of clinical oncology 2003 update on the role of bisphosphonates and bone health issues in women with breast cancer. J Clin Oncol 21:4042–4057 Santini D, Fratto ME, Vincenzi B et al, 2009, Denosumab: the era of targeted therapies in bone metastatic disease. Curr Canc Drug Targets 9:834–842 Baron R, Ferrari S, Russell RGG, 2011, Denosumab and bisphosphonates: different mechanisms of action and effects. Bone 48:677–692 Kumagai Y, Hasunuma T, Padhi D, 2011, A randomized, doubleblind, placebo-controlled, single-dose study to evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of denosumab administered subcutaneously to postmenopausal Japanese women. Bone 49:1101–1107 Stopeck AT, Lipton A, Body JJ et al, 2010, Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J Clin Oncol 28:5132–5139 Bjarnason NH, Hitz M, Jorgensen NR, Vestergaard P, 2008, Adverse bone effects during pharmacological breast cancer therapy. Acta Oncol 47:747–754 Powles TJ, Hickish T, Kanis JA et al, 1996, Effect of tamoxifen on bone mineral density measured by dual-energy X-ray absorptiometry in healthy premenopausal and postmenopausal women. J Clin Oncol 14:78–84 Love RR, Mazess RB, Barden HS et al, 1992, Effects of tamoxifen on bone mineral density in postmenopausal women with breast cancer. N Engl J Med 326:852–856 Kristensen B, Ejlertsen B, Dalgaard P et al, 1994, Tamoxifen and bone metabolism in postmenopausal low-risk breast cancer patients: a randomized study. J Clin Oncol 12:992–997 Geisler J, Lonning PE, Krag LE et al, 2006, Changes in bone and lipid metabolism in postmenopausal women with early breast cancer after terminating 2-year treatment with exemestane: a randomised, placebo-controlled study. Eur J Cancer 42:2968–2975 Perez EA, Josse RG, Pritchard KI et al, 2006, Effect of letrozole versus placebo on bone mineral density in women with primary breast cancer completing 5 or more years of adjuvant tamoxifen: a companion study to NCIC CTGMA. 17. J Clin Oncol 24:3629–3635 Coombes RC, Kilburn LS, Snowdon CF et al, 2007, Survival and safety of exemestane versus tamoxifen after 2–3 years’ tamoxifen treatment, Intergroup Exemestane Study): a randomised controlled trial. Lancet 369:559–570 Coleman RE, Banks LM, Girgis SI et al, 2007, Skeletal effects of exemestane on bone-mineral density, bone biomarkers, and fracture incidence in postmenopausal women with early breast cancer participating in the Intergroup Exemestane Study, IES): a randomised controlled study. Lancet Oncol 8:119–127 Eastell R, Hannon RA, Cuzick J et al, 2006, Effect of an aromatase inhibitor on BMD and bone turnover markers: 2-year results of the anastrozole, tamoxifen, alone or in combination, ATAC, trial. J Bone Miner Res 21:1215–1223 Fogelman I, Blake GM, Blamey R et al, 2003, Bone mineral density in premenopausal women treated for node-positive early breast cancer with 2 years of goserelin or 6 months of cyclophosphamide, methotrexate and 5-fluorouracil, CMF). Osteoporos Int 14:1001–1006 Crew KD, Greenlee H, Capodice J et al, 2007, Prevalence of joint symptoms in postmenopausal women taking aromatase inhibitors for early-stage breast cancer. J Clin Oncol 25:3877–3883 Fontaine C, Meulemans A, Huizing M et al, 2008, Tolerance of adjuvant letrozole outside of clinical trials. Breast 17:376–381 Hadji P, Bundred N, 2007, Reducing the risk of cancer treatmentassociated bone loss in patients with breast cancer. Semin Oncol 34:S4–S10 Delmas PD, Balena R, Confravreux E et al, 1997, Bisphosphonate risedronate prevents bone loss in women with artificial menopause due to chemotherapy of breast cancer: a double-blind, placebocontrolled study. J Clin Oncol 15:955–962 Lester JE, Dodwell D, Purohit OP et al, 2008, Prevention of anastrozole-induced bone loss with monthly oral ibandronate during adjuvant aromatase inhibitor therapy for breast cancer. Clin Cancer Res 14:6336–6342 Gnant M, Mlineritsch B, Stoeger H et al, 2011, Adjuvant endocrine therapy plus zoledronic acid in premenopausal women with early-stage breast cancer: 62-month follow-up from the ABCSG- 12 randomised trial. Lancet Oncol 12:631–641 Brufsky AM, Harker WG, Beck JT, 2012, Final 5-year results of Z-FAST trial. Cancer 118:1192–1201 Eidtmann H, de Boer R, Bundred N et al, 2010, Efficacy of zoledronic acid in postmenopausal women with early breast cancer receiving adjuvant letrozole: 36-month results of the ZO-FAST study. Ann Oncol 21:2188–2194 Llombart A, Frassoldati A, Paija O et al, 2012, Immediate administration of zoledronic acid reduces aromatase inhibitor-associated bone loss in postmenopausal women with early breast cancer: 12- month analysis of the E-ZO-FAST trial. Clin Breast Canc 12:40–48 Hines SL, Mincey B, Dentchev T et al, 2009, Immediate versus delayed zoledronic acid for prevention of bone loss in postmenopausal women with breast cancer starting letrozole after tamoxifen- N03CC. Breast Canc Res Treat 117:603–609 Van Poznak C, Hannon RA, Mackey JR et al, 2010, Prevention of aromatase inhibitor-induced bone loss using risedronate: the SABRE trial. J Clin Oncol 28:967–975 Gnant M, Mlineritsch B, Luschin-Ebengreuth G et al, 2007, Zoledronic acid prevents cancer treatment-induced bone loss in premenopausal women receiving adjuvant endocrine therapy for hormone-responsive breast cancer: a report from the Austrian Breast and Colorectal Cancer StudyGroup. J Clin Oncol 25:820–828 GnantM,Mlineritsch B, Luschin-Ebengreuth G et al, 2008, Adjuvant endocrine therapy plus zoledronic acid in premenopausal women with early-stage breast cancer: 5-year follow-up of the ABCSG-12 bonemineral density substudy. Lancet Oncol 9:840–849 Gnant M, Mlineritsch B, Schippinger W et al, 2009, Endocrine therapy plus zoledronic acid in premenopausal breast cancer. N Engl J Med 360:679–691 Ellis GK, Bone HG, Chlebowski R et al, 2009, Effect of denosumab on bone mineral density in women receiving adjuvant aromatase inhibitors for non-metastatic breast cancer: subgroup analyses of a phase 3 study. Breast Canc Res Treat 118:81–87 Hadji P, Aapro MS, Body JJ et al, 2011, Management of aromatase inhibitor-associated bone loss in postmenopausal women with breast cancer: practical guidance for prevention and treatment. Ann Oncol 22:2546–2555 Powles T, Paterson A, McCloskey E et al, 2006, Reduction in bone relapse and improved survival with oral clodronate for adjuvant treatment of operable breast cancer [ISRCTN83688026]. Breast Canc Res 8:R13 Coleman RE, 2009, Adjuvant bisphosphonates in breast cancer: are we witnessing the emergence of a new therapeutic strategy? Eur J Cancer 45:1909–1915 Coleman RE, Marshall H, Cameron D et al, 2011, Breast-cancer adjuvant therapy with zoledronic acid. N Engl J Med 365:1396– 1405 Diel IJ, Solomayer EF, Costa SD et al, 1998, Reduction in new metastases in breast cancer with adjuvant clodronate treatment. N Engl J Med 339:357–363 Coleman RE, Winter MC, Cameron D et al, 2010, The effects of adding zoledronic acid to neoadjuvant chemotherapy on tumour response: exploratory evidence for direct anti-tumour activity in breast cancer. Br J Cancer 102:1099–1105 Yan T, Yin W, Zhou Q et al, 2012, The efficacy of zoledronic acid in breast cancer adjuvant therapy: a meta-analysis of randomised controlled trials. Eur J Cancer 48:187–195 Lee BL, Higgins MJ, Goss PE, 2012, Denosumab and the current status of bone-modifying drugs in breast cancer. Acta Oncol 51:157– 167 Cuzick J, DeCensi A, Arun B et al, 2011, Preventive therapy for breast cancer: a consensus statement. Lancet Oncol 12:496–503 Rennert G, Pinchev M, Rennert HS, 2010, Use of bisphosphonates and risk of postmenopausal breast cancer. J Clin Oncol 28:3577–3581 Chlebowski RT, Chen Z, Cauley JA et al, 2010, Oral bisphosphonate use and breast cancer incidence in postmenopausal women. J Clin Oncol 28:3582–3590 Marra M, Salzano G, Leonetti C et al, 2012, New self-assembly nanoparticles and stealth liposomes for the delivery of zoledronic acid: a comparative study. Biotechnol Adv 30:302–309 Salzano G, Marra M, Porru M et al, 2011, Self-assembly nanoparticles for the delivery of bisphosphonates into tumors. Int J Pharm 403:292–297 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 1 EP 10 DI 10.1007/s12253-012-9586-9 PG 10 ER PT J AU Pan, G Zhang, X Ren, J Lu, J Li, W Fu, H Zhang, Sh Li, J AF Pan, Guoqing Zhang, Xiangling Ren, Junyu Lu, Jianbo Li, Wenliang Fu, Hongmei Zhang, Shufang Li, Jun TI Semaphorin 5A, an Axon Guidance Molecule, Enhances the Invasion and Metastasis of Human Gastric Cancer through Activation of MMP9 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Semaphorin 5A; Gastric cancer; Invasion and metastasis; MMP9; ErK1/2 ID Semaphorin 5A; Gastric cancer; Invasion and metastasis; MMP9; ErK1/2 AB Semaphorin 5A, a member of semaphorin family, was originally identified as axonal guidance factor functioning during neuronal development. Previously, we showed that the expression of semaphorin 5A might contribute to the metastasis of gastric cancer. However, its functional roles and mechanism(s) in invasion and metastasis of gastric cancer remain unclear. By using human gastric caner cell lines Parental SGC7901, SGC7901-siScrambled and SGC7901-siSema 5A, we found that semaphorin 5A significantly promoted the invasive and metastatic abilities of gastric cancer cell in vitro. Semaphorin 5A increased the expression of MMP9 by activating phosphorylated ErK1/2 in gastric cancer cell. Furthermore, MEK inhibitor PD98059 and MMP9 antibody (Ab) significantly inhibited in vitro invasive and metastatic abilities induced by semaphorin 5A. Taken together, the present work revealed a novel function of semaphorin 5A that the existence of semaphorin 5A could promote invasion and metastasis of gastric cancer by regulating MMP9 expression, at least partially, via the MEK/ERKs signal transduction pathway. Semaphorin 5A and its regulated molecules could be the potential targets for cancer therapy. C1 [Pan, Guoqing] The First Affiliated Hospital of Kunming Medical College, Department of PathologyKunming, Yunnan, China. [Zhang, Xiangling] The First Affiliated Hospital of Kunming Medical College, Department of Blood TransfusionKunming, Yunnan, China. [Ren, Junyu] The First Affiliated Hospital of Kunming Medical College, Department of SurgeryKunming, Yunnan, China. [Lu, Jianbo] The First Affiliated Hospital of Kunming Medical College, Department of PathologyKunming, Yunnan, China. [Li, Wenliang] The First Affiliated Hospital of Kunming Medical College, Department of SurgeryKunming, Yunnan, China. [Fu, Hongmei] The First Affiliated Hospital of Kunming Medical College, Department of PathologyKunming, Yunnan, China. [Zhang, Shufang] Central South University, Affiliated Hai Kou Hospital, Xiangya Medical College, Central Laboratory, Renmin road #43, 570208 Haikou, Hainan, China. [Li, Jun] The First Affiliated Hospital of Kunming Medical College, Department of NephrologyKunming, Yunnan, China. RP Zhang, Sh (reprint author), Central South University, Affiliated Hai Kou Hospital, Xiangya Medical College, Central Laboratory, 570208 Haikou, China. EM haikuoyinyuan@163.com CR Parkin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics, 2002. CA Cancer J Clinic 55:74–108 Foukakis T, Lundell L, Gubanski M et al, 2007, Advances in the treatment of patients with gastric adenocarcinoma. Acta Oncol 46:277–285 Yazdani U, Terman JR, 2006, The semaphorins. Genome Biol 7:211–224 Kolodkin AL, Matthes DJ, Goodman CS, 1993, The semaphorin genes encode a family of transmembrane and secreted growth cone guidance molecules. Cell 75:1389–1399 Kruger RR et al, 2005, Semaphorins command cells to move. Nat Rev Mol Cell Biol 5556:789–800 Pan GQ, Ren HZ, Wen JF et al, 2009, Expression of semaphorin 5A and its receptor plexin B3 contributes to invasion and metastasis of gastric carcinoma. World J Gastroenterol 15:2800–2804 Pan G, Lv H, Ren H, Wang Y, Liu Y, Jiang H, Wen J, 2010, Elevated expression of semaphorin 5A in human gastric cancer and its implication in carcinogenesis. Life Sci 86:139–144 Laurens N, Engelse MA, Jungerius C, Lowik CW, van Hinsbergh VW et al, 2009, Single and combined effects of alphavbeta3- and alpha5betal-integrins on capillary tube formation in a human fibrinous matric. Angigenesis 12:275–285 Reddy KB, Nabha SM, Atanaskova N, 2003, Role of MAP kinase in tumor progression and invasion. Cancer Metastasis Rev 22:395– 403 Hilario JD, Rodino-Klapac LR, Wang C et al, 2009, Semaphorin 5A is a bifunctional axon guidance cue for axial motoneurons in vivo. Dev Biology 326:190–200 Sun Q, Nawabi-Ghasimi F, Basile JR, 2008, Semaphorins in vascular development and head and neck squamous cell carcinoma-induced angiogenesis. Oral oncology 44:523–531 Roth L, Koncina E, Satkauskas S et al, 2009, The many faces of semaphorins: from development to pathology. Cell Mol Life Science 66:649–666 Neufeld G, Shraga-Heled N, Lange T et al, 2005, Semaphorins in cancer. Front Biosci 10:751–760 Parsons SL, Watson SA, Collins HM, 1998, Gelatinase, MMP-2 and -9, expression in gastrointestinal malignancy. Br J Cancer 78:1495–1502 Artigiani S, Conrotto P, Fazzari P et al, 2004, Plexin-B3 is a functional receptor for semaphorin 5A. EMBO Reports 5:710–714 Serini G et al, 2009, Semaphorins and tumor angiogenesis. Angiogenesis 12:187–193 Schlomann U, Schwamborn JC, Muller M, Fassler R, Puschel AW, 2009, The stimulation of dendrite growth by Sema3A requires integrin engagement and focal adhesion kinase. J Cell Sci 15, 122):2034–2042 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 11 EP 18 DI 10.1007/s12253-012-9550-8 PG 8 ER PT J AU Zhu, Ch Wang, Q Xie, J Shi, J Zhou, X Li, D Xiong, F Zhang, L AF Zhu, Chunrong Wang, Qingcai Xie, Jing Shi, Jinfang Zhou, Xiumin Li, Dapeng Xiong, Feng Zhang, Lu TI Expression and Significance of RKIP and E-cadherin in Lung Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE RKIP; E-cadherin; Lung squamous cell carcinoma; RT-PCR; Western blot ID RKIP; E-cadherin; Lung squamous cell carcinoma; RT-PCR; Western blot AB The purpose of this study was to investigate the expression of Raf kinase inhibitor protein (RKIP) and epithelial cadherin (E-cadherin) in lung squamous cell carcinoma tissue and its correlation with the clinical pathology of lung squamous cell carcinoma. RKIP and E-cadherin mRNA (by RT-PCR) and protein (by western blotting) levels were monitored in carcinoma tissues and surrounding normal tissues from 86 lung squamous cell carcinoma cases, and their positive rates were calculated. The rates of positive RKIP and E-cadherin mRNA expression were significantly lower in lung squamous cell carcinoma than in the surrounding normal tissues (P<0.05). The positive expression rates were significantly lower in those with lymph node metastasis than in those without (P<0.05). The lower the degree of tumor differentiation, the lower the E-cadherin mRNA positive expression rate (P<0.05). The rates of positive RKIP and E-cadherin mRNA expression were significantly lower in patients at advanced (III, IV) stages than in patients at early (I, II) stages (p<0.05); this rate, however, was independent of gender, age, and tumor size (P>0.05). The protein levels of RKIP and E-cadherin were significantly lower in lung squamous cell carcinoma than in the surrounding normal tissues (P<0.05). The levels were significantly lower in patients with lymph node metastasis than in those without it (P<0.05). The lower the degree of tumor differentiation, the lower the protein level of E-cadherin (P< 0.05). Both RKIP and E-cadherin are tumor suppressors, their low expression levels may be associated with initiation, invasion and/or metastasis, as well as with the inhibition of lung squamous cell carcinoma differentiation. C1 [Zhu, Chunrong] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, No. 188, Shi Zi Rd, 215006 Suzhou, China. [Wang, Qingcai] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, No. 188, Shi Zi Rd, 215006 Suzhou, China. [Xie, Jing] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, No. 188, Shi Zi Rd, 215006 Suzhou, China. [Shi, Jinfang] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, No. 188, Shi Zi Rd, 215006 Suzhou, China. [Zhou, Xiumin] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, No. 188, Shi Zi Rd, 215006 Suzhou, China. [Li, Dapeng] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, No. 188, Shi Zi Rd, 215006 Suzhou, China. [Xiong, Feng] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, No. 188, Shi Zi Rd, 215006 Suzhou, China. [Zhang, Lu] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, No. 188, Shi Zi Rd, 215006 Suzhou, China. RP Zhu, Ch (reprint author), Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, 215006 Suzhou, China. EM zhucr2011@yeah.net CR Yeung K, Seitz T, Li S, Janosch P, McFerran B, Kaiser C, Fee F, Katsanakis KD, Rose DW, Mischak H, Sedivy JM, Kolch W, 1999, Suppression of Raf-1 kinase activity and MAP kinase signalling by RKIP. Nature 401:173–7 Odabaei G, Chatterjee D, Jazirehi AR, Goodglick L, Yeung K, Bonavida B, 2004, Raf-1 kinase inhibitor protein: structure, function, regulation of cell signaling, and pivotal role in apoptosis. Adv Cancer Res 91:169–200 Beshir AB, Ren G,Magpusao AN, Barone LM,Yeung KC, Fenteany G, 2010, Raf kinase inhibitor protein suppresses nuclear factorkappaB- dependent cancer cell invasion through negative regulation of matrix metalloproteinase expression. Cancer Lett 299:137–49 Zeng L, Imamoto A, Rosner MR, 2008, Raf kinase inhibitory protein, RKIP): a physiological regulator and future therapeutic target. Expert Opin Ther Targets 12:1275–87 Fu Z, Smith PC, Zhang L, Rubin MA, Dunn RL, Yao Z, Keller ET, 2003, Effects of raf kinase inhibitor protein expression on suppression of prostate cancer metastasis. J Natl Cancer Inst 95:878–89 Xu YF, Yi Y, Qiu SJ, Gao Q, Li YW, Dai CX, Cai MY, Ju MJ, Zhou J, Zhang BH, Fan J, 2010, PEBP1 downregulation is associated to poor prognosis in HCC related to hepatitis B infection. J Hepatol 53:872–879 Lin K, Baritaki S, Militello L, Malaponte G, Bevelacqua Y, Bonavida B, 2010, The role of B-RAF mutations in melanoma and the induction of EMT via dysregulation of the NF-kappaB/Snail/ RKIP/PTEN circuit. Genes Cancer 1:409–20 Al-Mulla F, Hagan S, Al-Ali W, Jacob SP, Behbehani AI, Bitar MS, Dallol A, Kolch W, 2008, Raf kinase inhibitor protein: mechanism of loss of expression and association with genomic instability. J Clin Pathol 61:524–9 Li HZ, Gao Y, Zhao XL, Liu YX, Sun BC, Yang J, Yao Z, 2009, Effects of raf kinase inhibitor protein expression on metastasis and progression of human breast cancer. Mol Cancer Res 7:832–40 van Roy F, Berx G, 2008, The cell-cell adhesion molecule Ecadherin. Cell Mol Life Sci 65:3756–88 Rachagani S, Senapati S, Chakraborty S, Ponnusamy MP, Kumar S, Smith LM, Jain M, Batra SK, 2011, Activated Kras(G12D, is associated with invasion and metastasis of pancreatic cancer cells through inhibition of E-cadherin. Br J Cancer 104:1038–48 Gu X, Ma C, Yuan D, Song Y, 2012, Circulating soluble intercellular adhesion molecule-1 in lung cancer: a systematic review. Transl Lung Cancer Res 1:36–44 Saad AA, Awed NM, Abd Elkerim NN, El-Shennawy D, Alfons MA, Elserafy ME, Darwish YW, Barakat EM, Ezz-Elarab SS, 2010, Prognostic significance of E-cadherin expression and peripheral blood micrometastasis in gastric carcinoma patients. Ann Surg Oncol 17:3059–67 Metro G, Crino L, 2012, Advances on EGFR mutation for lung cancer. Transl lung Cancer Res 1:5–13 Ellis PM, Vandermeer R, 2011, Delays in the diagnosis of lung cancer. J Thorac Dis 3:183–8 Shao WL, Wang DY, He JX, 2010, The role of gene expression profiling in early-stage non-small cell lung cancer. J Thorac Dis 2:89–99 Yang HH, Zhang Q, He JX, Lu WJ, 2010, Regulation of calcium signaling in lung cancer. J Thorac Dis 2:52–6 Xiao DK, He JX, 2010, Epithelial mesenchymal transition and lung cancer. J Thorac Dis 2:154–9 Korpanty G, Smyth E, Carney DN, 2011, Update on antiangiogenic therapy in non-small cell lung cancer: Are we making progress? J Thorac Dis 3:19–29 Shash E, Peccatori FA, Azim HA Jr, 2011, Optimizing the use of epidermal growth factor receptor inhibitors in advanced non-smalllung cancer, NSCLC). J Thorac Dis 3:57–64 McKeage MJ, Jameson MB, AS1404-201 Study Group Investigators, 2010, Comparative outcomes of squamous and nonsquamous non-small cell lung cancer, NSCLC, patients in phase II studies of ASA404, DMXAA)—retrospective analysis of pooled data. J Thorac Dis 2:199–204 Lee HC, Tian B, Sedivy JM,Wands JR, Kim M, 2006, Loss of Raf kinase inhibitor protein promotes cell proliferation and migration of human hepatoma cells. Gastroenterology 131:1208–17 Akaishi J, Onda M, Asaka S, Okamoto J, Miyamoto S, Nagahama M, Ito K, Kawanami O, Shimizu K, 2006, Growth-suppressive function of phosphatidylethanolaminebinding protein in anaplastic thyroid cancer. Anticancer Res 26:4437–42 Hagan S, Al-Mulla F, Mallon E, Oien K, Ferrier R, Gusterson B, Garcia JJ, Kolch W, 2005, Reduction of Raf-1 kinase inhibitor protein expression correlates with breast cancer metastasis. Clin Cancer Res 11:7392–7 Fu Z, Kitagawa Y, Shen R, Shah R, Mehra R, Rhodes D, Keller PJ, Mizokami A, Dunn R, Chinnaiyan AM, Yao Z, Keller ET, 2006, Metastasis suppressor gene Raf kinase inhibitor protein, RKIP, is a novel prognostic marker in prostate cancer. Prostate 66:248–56 Zeljko M, Pecina-Slaus N, Martic TN, Kusec V, Beros V, Tomas D, 2011, Molecular alterations of E-cadherin and beta-catenin in brain metastases. Front Biosci, Elite Ed, 3:616–24 Wang G, Dong W, Shen H, Mu X, Li Z, Lin X, Liu Y, Du J, 2011, A comparison of Twist and E-cadherin protein expression in primary non-small-cell lung carcinoma and corresponding metastases. Eur J Cardiothorac Surg. Mar 2. [Epub ahead of print] Sulzer MA, Leers MP, van Noord JA, Bollen EC, Theunissen PH, 1998, Reduced E-cadherin expression is associated with increased lymph node metastasis and unfavorable prognosis in non-small cell lung cancer. Am J Respir Crit Care Med 157:1319–23 Zigler M, Dobroff AS, Bar-Eli M, 2010, Cell adhesion: implication in tumor progression. Minerva Med 101:149–62 Yeung K, Janosch P, McFerran B, Rose DW, Mischak H, Sedivy JM, Kolch W, 2000, Mechanism of suppression of the Raf/MEK/extracellular signal-regulated kinase pathway by the raf kinase inhibitor protein. Mol Cell Biol 20:3079–85 Lan M, Kojima T, Osanai M, Chiba H, Sawada N, 2004, Oncogenic Raf-1 regulates epithelial to mesenchymal transition via distinct signal transduction pathways in an immortalized mouse hepatic cell line. Carcinogenesis 25:2385–95 Herfs M, Hubert P, Kholod N, Caberg JH, Gilles C, Berx G, Savagner P, Boniver J, Delvenne P, 2008, Transforming growth factor-beta1-mediated Slug and Snail transcription factor upregulation reduces the density of Langerhans cells in epithelial metaplasia by affecting E-cadherin expression. Am J Pathol 172:1391–402 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 19 EP 26 DI 10.1007/s12253-012-9552-6 PG 8 ER PT J AU Engelman, dFBM Grande, MR Naves, AM de Franco, FM de Paulo Castro Teixeira, V AF Engelman, de Fatima Brasil Miriam Grande, Mendes Rogerio Naves, Andery Marcelo de Franco, Fabiano Marcello de Paulo Castro Teixeira, Vicente TI Integrin-Linked Kinase (ILK) Expression Correlates with Tumor Severity in Clear Cell Renal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Immunohistochemistry; Tissue array analysis; Carcinoma; Renal cell; Integrin-linked kinase; Human ID Immunohistochemistry; Tissue array analysis; Carcinoma; Renal cell; Integrin-linked kinase; Human AB Integrin-linked kinase (ILK) is an unique intracellular serine/threonine kinase and adapter protein. When dysregulated, it has been associated with increased cell proliferation, anchorage-independent cell growth, evasion of apoptosis, angiogenesis, invasion of surrounding tissues, downregulation of E-cadherin expression, nuclear translocation of β-catenin and metastasis, all features of tumoral malignancy. The objective of the present work was to evaluate the expression of ILK in clear cell renal carcinomas (CCRC) as a possible prognostic indicator. ILK immunoexpression was evaluated in a tissue microarray (TMA) with 45 human CCRCs. In addition, the apoptotic and proliferative indices and the immuno-expression of β-catenin and Ecadherin were also evaluated. E-cadherin expression was significantly decreased in tumors with positive ILK expression in relation to those with negative immunoexpression (p00.011). ILK immunostaining was significantly increased in high-grade in comparison to low-grade CCRCs (p<0.0008). ILK expression was also associated with increased proliferative index (p<0.020), tumor size >7.0 cm (p<0.018) and with renal vein and capsule invasion (p<0.003 and p<0.00). Finally, tumors stage I and II (noninvasive) presented significantly reduced ILK immunoexpression when compared to stage III (locally invasive) (p<0.0028). ILK immunoexpression in CCRC increases with loss of intercellular adhesion, nuclear grading, increased proliferative index and Robson stage. Altogether, our data suggest a possible role for ILK in the progression of CRCC. C1 [Engelman, de Fatima Brasil Miriam] Universidade do Vale do Sapucai, Faculdade de Ciencias da Saude Dr Jose Antonio Garcia Coutinho, Avenida Alfredo Custodio de Paula 360, 37550000 Pouso Alegre, Minas Gerais, Brazil. [Grande, Mendes Rogerio] Universidade do Vale do Sapucai, Faculdade de Ciencias da Saude Dr Jose Antonio Garcia Coutinho, Avenida Alfredo Custodio de Paula 360, 37550000 Pouso Alegre, Minas Gerais, Brazil. [Naves, Andery Marcelo] Sao Paulo University School of Medicine, Department of Pathology, Rua Botucatu Edificio Lemos Torres 1° Andar, 04023-062 Sao Paulo, SP, Brazil. [de Franco, Fabiano Marcello] Sao Paulo University School of Medicine, Department of Pathology, Rua Botucatu Edificio Lemos Torres 1° Andar, 04023-062 Sao Paulo, SP, Brazil. [de Paulo Castro Teixeira, Vicente] Sao Paulo University School of Medicine, Department of Pathology, Rua Botucatu Edificio Lemos Torres 1° Andar, 04023-062 Sao Paulo, SP, Brazil. RP Engelman, dFBM (reprint author), Universidade do Vale do Sapucai, Faculdade de Ciencias da Saude Dr Jose Antonio Garcia Coutinho, 37550000 Pouso Alegre, Brazil. EM mi.engelman@uol.com.br CR Hannigan GE, Leung-Hagesteijn C, Fitz-Gibbon L et al, 1996, Regulation of cell adhesion and anchorage-dependent growth by a new beta 1-integrin-linked protein kinase. Nature 379:91–96 Dedhar S, Williams B, Hannigan G, 1999, Integrin-linked kinase, ILK): a regulator of integrin and growth-factor signalling. Trends Cell Biol 9:319–323 Wu C, Keightley SY, Leung-Hagesteijn C et al, 1998, Integrinlinked protein kinase regulates fibronectin matrix assembly, Ecadherin expression, and tumorigenicity. J Biol Chem 273:528– 536 Delcommenne M, Tan C, Gray V, Rue L, Woodgett J, Dedhar S, 1998, Phosphoinositide-3-OH kinase-dependent regulation of glycogen synthase kinase 3 and protein kinase B/AKT by the integrin-linked kinase. Proc Natl Acad Sci U S A 95:11211–11216 Hannigan G, Troussard AA, Dedhar S, 2005, Integrin-linked kinase: a cancer therapeutic target unique among its ILK. Nat Rev Cancer 5:51–63 Wu C, Dedhar S, 2001, Integrin-linked kinase, ILK, and its interactors: a new paradigm for the coupling of extracellular matrix to actin cytoskeleton and signaling complexes. J Cell Biol 155:505–510 Persad S, Attwell S, Gray V et al, 2000, Inhibition of integrinlinked kinase, ILK, suppresses activation of protein kinase B/Akt and induces cell cycle arrest and apoptosis of PTEN-mutant prostate cancer cells. Proc Natl Acad Sci U S A 97:3207–3212 Novak A, Hsu S-C, Leung-Hagesteijn C et al, 1998, Cell adhesion and the integrin-linked kinase regulate the LEF-1 and β-catenin signaling pathways. Proc Natl Acad Sci U S A 95:4374–4379 D’Amico M, Hulit J, Atullah DF et al, 2000, The integrin-linked kinase regulates the cyclin D1 gene through glycogen synthase kinase 3B and cAMP-responsive element-binding proteindependent pathways. J Biol Chem 75:32649–32657 Oloumi A, McPhee T, Dedhar S, 2004, Regulation of E-cadherin expression and beta-catenin/Tcf transcriptional activity by the integrin-linked kinase. Biochim Biophys Acta 169:1–15 Graff JR, Deddens JA, Konicek BW et al, 2001, Integrin-linked kinase expression increases with prostate tumor grade. Clin Cancer Res 7:1987–1991 Marotta A, Tan C, Gray V et al, 2001, Dysregulation of integrinlinked kinase, ILK, signaling in colonic polyposis. Oncogene 20:6250–6267 Marotta A, Parhar K, Owen D, Dedhar S, Salh B, 2003, Characterisation of integrin-linked kinase signalling in sporadic human colon cancer. Br J Cancer 88:1755–1762 Bravou V, Klironomos G, Papadaki E, Taraviras S, Varakis J, 2006, ILK over-expression in human colon cancer progression correlates with activation of beta-catenin, down-regulation of Ecadherin and activation of the Akt-FKHR pathway. J Pathol 208:91–99 Ito R, Oue N, Zhu X et al, 2003, Expression of integrin-linked kinase is closely correlated with invasion and metastasis of gastric carcinoma. Virchows Arch 442:118–123 Ahmed N, Riley C, Oliva K, Stutt E, Rice GE, Quinn MA, 2003, Integrin-linked kinase expression increases with ovarian tumour grade and is sustained by peritoneal tumour fluid. J Pathol 201:229–237 Dai DL, Makretsov N, Campos EI et al, 2003, Increased expression of integrin-linked kinase is correlated with melanoma progression and poor patient survival. Clin Cancer Res 9:4409–4414 Chung DH, Lee JI, Kook MC et al, 1998, ILK, beta1-integrinlinked protein kinase): a novel immunohistochemical marker for Ewing’s sarcoma and primitive neuroectodermal tumour. Virchows Arch 433:113–117 Takanami I, 2005, Increased expression of integrin-linked kinase is associated with shorter survival in non-small cell lung cancer. BMC Cancer 5:1 Okamura M, Yamaji S, Nagashima Yet al, 2007, Prognostic value of integrin beta1-ILK-pAkt signaling pathway in non-small cell lung cancer. Hum Pathol 38:1081–1091 Watzka SB, Rauscher-Potsch I, Stubenberger E et al, 2010, Immunoreactivity of integrin-linked kinase in primary non-small-cell lung cancer and survival after curative resection. Eur J Cardiothorac Surg 38:254–259 Papanikolaou S, Bravou V, Gyftopoulos K, Nakas D, Repanti M, Papadaki H, 2010, ILK expression in human basal cell carcinoma correlates with epithelial-mesenchymal transition markers and tumour invasion. Histopathology 56:799–809 Schaeffer DF, Assi K, Chan K et al, 2010, Tumor expression of integrin-linked kinase, ILK, correlates with the expression of the E-cadherin repressor snail: an immunohistochemical study in ductal pancreatic adenocarcinoma. Virchows Arch 456:261–268 Peroukides S, Bravou V, Varakis J, Alexopoulos A, Kalofonos H, Papadaki H, 2008, ILK overexpression in human hepatocellular carcinoma and liver cirrhosis correlates with activation of Akt. Oncol Rep 20:1337–1344 Goulioumis AK, Bravou V, Varakis J, Goumas P, Papadaki H, 2008, Integrin-linked kinase cytoplasmic and nuclear expression in laryngeal carcinomas. Virchows Arch 453:511–519 Matsui Y, Assi K, Ogawa O et al, 2012, The importance of integrin-linked kinase in the regulation of bladder cancer invasion. Int J Cancer 130:521–531 Yoshino S, Kato M, Okada K, 2000, Clinical significance of angiogenesis, proliferation and apoptosis in renal cell carcinoma. Anticancer Res 20:591–594 Kirkali Z, Yorukoglu K, Ozkara E, Kazimoglu H, Mungan U, 2001, Proliferative activity, angiogenesis and nuclear morphometry in renal cell carcinoma. Int J Urol 8:697–703 Kononen J, Bubendorf L, Kallioniemi A et al, 1998, Tissue microarrays for high-through put molecular profiling of tumor specimens. Nat Med 4:844–847 Kallakuri BVS, Sheehan CE, Winn-Deen E et al, 2001, Decreased expression of catenins, α and β), p120, CTNM and E-cadherin gene promoter methylation in prostatic adenocarcinomas. Cancer 92:2786–2795 Rao JY, Seligson D, Visapaa H et al, 2002, Tissue microarray analysis for cytoskeletal actin-associated biomarkes gelsolin and cadherin in urothelial carcinoma. Cancer 95:1247–1257 Skacel M, Skilton B, Pettay JD, Tubbs RR, 2002, Tissue microarrays: a powerful tool for high-throughput an analisis of clinical specimens—a review of the method whit validation data. Appl Immunohistochem Mol Morphol 10:1–6 Haase M, Gmach CC, Eke I, Hehlgans S, Baretton GB, Cordes N, 2008, Expression of integrin-linked kinase is increased in differentiated cells. J Histochem Cytochem 56:819–829 Thiery JP, 2004, Epithelial-mesenchymal transition: twist in development and metastasis. Cell 1014:1125–1136 Katagiri A, Watanabe R, Tomita Y, 2000, E-cadherin expression in renal cell cancer and its significance in metastasis and survival. Br J Cancer 85:801–804 Pecina-Slaus N, Gall-TroSelj K, Slaus M et al, 2004, Genetic changes of the E-cadherin and APC tumour suppressor genes in clear renal cell carcinoma. Pathology 36:145–151 Slaton JW, Inoue K, Perrotte P et al, 2001, Expression levels of genes that regulate metastasis and angiogenesis correlate with advanced pathological stage of renal cell carcinoma. Am J Pathol 158:735–743 Younes MN, Kim S, Yigitbasi OG et al, 2005, Integrin-linked kinase is a potential therapeutic target for anaplastic thyroid cancer. Mol Cancer Ther 4:1146–1156 Duxbury MS, Ito H, Benoit E, Waseem T, Ashley SW, Whang EE, 2005, RNA interference demonstrates a novel role for integrinlinked kinase as a determinant of pancreatic adenocarcinoma cell gemcitabine chemoresistance. Clin Cancer Res 11:3433–3438 Edwards LA, Thiessen B, Dragowska WH et al, 2005, Inhibition of ILK in PTEN-mutant human glioblastomas inhibits PKB/Akt activation, induces apoptosis, and delays tumor growth. Oncogene 24:3596–3605 Han L, Pamukcu R, Thompson J, 2002, β-catenin signaling: therapeutic strategies in oncology. Cancer Biol Ther 1:621–625 Guo L, Kuroda N, Miyazaki E et al, 2001, The complementary role of beta-catenin in diagnosing various subtypes of renal cell carcinomas and its up-regulation in conventional renal cell carcinomas with high nuclear grades. Oncol Rep 8:521–526 Kim YS, Kang YK, Kim JB, Han SA, Kim KI, Paik SR, 2000, β- catenin expression and mutational analysis in renal cell carcinomas. Pathol Int 50:725–730 Shiina H, Igawa M, Breault J et al, 2003, The human T-cell factor- 4 gene splicing isoforms, WNT pathway and apoptosis in renal cell carcinoma. Clin Cancer Res 9:2121–2132 Hindermann W, Berndt A, Wunderlich H, Katenkamp D, Kosmehl H, 1997, Quantitative evaluation of apoptosis and proliferation in renal cell carcinoma. Correlation to tumor subtype, cytological grade according to thoenes-classification and the occurrence of metastasis. Pathol Res Pract 193:1–7 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 27 EP 33 DI 10.1007/s12253-012-9554-4 PG 7 ER PT J AU Luo, Y Tian, L Ye, F Yi, M Chen, X Huang, Q AF Luo, Yanli Tian, Ling Ye, Feng Yi, Miaoying Chen, Xiafang Huang, Qian TI The Predictive Role of p16 Deletion, p53 Deletion, and Polysomy 9 and 17 in Pancreatic Ductal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic ductal adenocarcinoma; Prognosis; p16; p53; Deletion; Polysomy ID Pancreatic ductal adenocarcinoma; Prognosis; p16; p53; Deletion; Polysomy AB In this study, we investigated p53 and p16 deletions, and chromosome 9 and 17 amplifications in pancreatic ductal adenocarcinoma (PDAC), and further analyzed their associations with clinical characteristics and prognosis of PDAC. A total of 32 PDAC and 23 peritumoral tissues were collected. Molecular abnormalities of CEP9/p16 and CEP17/p53 were detected using Fluorescence in situ hybridization (FISH). Deletions of p16 and p53 were detected in 50 % and 65.7 % of PDAC, respectively. Polysomy 9 and 17 were identified in 75 % and 71.8 % of PDAC, respectively. No p16 and p53 deletion, polysomy 9 and 17 were identified in peritumoral tissues. We also observed significant correlations of p16 deletion, polysomy 9 and 17 with shorter survival of PDAC. P16 deletion, polysomy 9 and 17 are predictive markers for poor prognosis of PDAC patients, but p53 deletion is not associated with the clinical characteristics and prognosis of PDAC. C1 [Luo, Yanli] Shanghai Jiaotong University, First People’s Hospital, Experimental Research Center, 85 Wujin Road, 200080 Shanghai, China. [Tian, Ling] Shanghai Jiaotong University, First People’s Hospital, Experimental Research Center, 85 Wujin Road, 200080 Shanghai, China. [Ye, Feng] Shanghai Jiaotong University, First People’s Hospital, Experimental Research Center, 85 Wujin Road, 200080 Shanghai, China. [Yi, Miaoying] Shanghai Jiaotong University, First People’s Hospital, Experimental Research Center, 85 Wujin Road, 200080 Shanghai, China. [Chen, Xiafang] Shanghai Jiaotong University, First People’s Hospital, Experimental Research Center, 85 Wujin Road, 200080 Shanghai, China. [Huang, Qian] Shanghai Jiaotong University, First People’s Hospital, Experimental Research Center, 85 Wujin Road, 200080 Shanghai, China. RP Huang, Q (reprint author), Shanghai Jiaotong University, First People’s Hospital, Experimental Research Center, 200080 Shanghai, China. EM qhuang2012@yahoo.com CR Spratlin JL, Mulder KE, 2011, Looking to the future: biomarkers in the management of pancreatic adenocarcinoma. Int J Mol Sci 12:5895–5907 Singh P, Srinivasan R, Wig JD, 2011, Major molecular markers in pancreatic ductal adenocarcinoma and their roles in screening, diagnosis, prognosis, and treatment. Pancreas 40:644–652 Gansauge S, Gansauge F, Ramadani M, Stobbe H, Rau B, Harada N, Beger HG, 1997, Overexpression of cyclin D1 in human pancreatic carcinoma is associated with poor prognosis. Cancer Res 57:1634–1637 Stathopoulos GP, Androulakis N, Souglakos J, Stathopoulos J, Georgoulias V, 2008, Present treatment and future expectations in advanced pancreatic cancer. Anticancer Res 28:1303–1308 Cohenuram M, Saif MW, 2007, Epidermal growth factor receptor inhibition strategies in pancreatic cancer: past, present and the future. JOP 8:4–15 Saif MW, Karapanagiotou L, Syrigos K, 2007, Genetic alterations in pancreatic cancer. World J Gastroenterol 13:4423–4430 Martinez JD, 2010, Restoring p53 tumor suppressor activity as an anticancer therapeutic strategy. Future Oncol 6:1857–1862 Redston MS, Caldas C, Seymour AB, Hruban RH, da Costa L, Yeo CJ, Kern SE, 1994, p53 mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions. Cancer Res 54:3025–3033 Fry LC, Monkemuller K, Malfertheiner P, 2008, Molecular markers of pancreatic cancer: development and clinical relevance. Langenbecks Arch Surg 393:883–890 Garcea G, Neal CP, Pattenden CL, Steward WP, Berry DP, 2005, Molecular prognostic markers in pancreatic cancer: a systematic review. Eur J Cancer 41:2213–2236 Talar-Wojnarowska R, Malecka-Panas E, 2006, Molecular pathogenesis of pancreatic adenocarcinoma: potential clinical implications. Med Sci Monit 12:RA186–RA193 Caldas C, Hahn SA, da Costa LT, Redston MS, Schutte M, Seymour AB, Weinstein CL, Hruban RH, Yeo CJ, Kern SE, 1994, Frequent somatic mutations and homozygous deletions of the p16, MTS1, gene in independent poor prognostic predictor. Nat Genet 8:27–32 Naumann M, Savitskaia N, Eilert C, Schramm A, Kalthoff H, Schmiegel W, 1996, Frequent codeletion of p16/MTS1 and p15/ MTS2 and genetic alterations in p16/MTS1 in pancreatic tumors. Gastroenterology 110:1215–1224 Ohtsubo K, Watanabe H, Yamaguchi Y, Hu YX, Motoo Y, Okai T, Sawabu N, 2003, Abnormalities of tumor suppressor gene p16 in pancreatic carcinoma: immunohistochemical and genetic findings compared with clinicopathological parameters. J Gastroenterol 38:663–671 Jeong J, Park YN, Park JS, Yoon DS, Chi HS, Kim BR, 2005, Clinical significance of p16 protein expression loss and aberrant p53 protein expression in pancreatic cancer. Yonsei Med J 46:519– 525 Kim HG, Yamal JM, Xu XC, Hu W, Boiko I, Linares A et al, 2005, Cervical chromosome 9 polysomy: validation and use as a surrogate endpoint biomarker in a 4-HPR chemoprevention trial. Gynecol Oncol 99:S32–S37 Stoecklein NH, Luebke AM, Erbersdobler A, Knoefel WT, Schraut W, Verde PE et al, 2004, Copy number of chromosome 17 but not HER2 amplification predicts clinical outcome of patients with pancreatic ductal adenocarcinoma. J Clin Oncol 22:4737–4745 Chiosea S, Krasinskas A, Cagle PT, Mitchell KA, Zander DS, Dacic S, 2008, Diagnostic importance of 9p21 homozygous deletion in malignant mesotheliomas. Mod Pathol 21:742–747 Shan Z, Wu P, Zheng S, Tan W, Zhou H, Zuo Qi H et al, 2010, Wang, evaluation of upper urinary tract tumors by FISH in Chinese patients. Cancer Genet Cytogenet 203:238–246 Sobin LH, Gospodarowicz MK, 2009, Wittekind C UICC-TNM Classification of Malignant Tumours. Wiley-Liss, New York, pp 132–135 Korshunov A, Sycheva R, Golanov A, 2004, Molecular stratification of diagnostically challenging high-grade Gliomas composed of small cells: the utility of fluorescence in situ hybridization. Clin Cancer Res 10:7820–7826 Botti C, Pescatore B, Mottolese M, Sciarretta F, Greco C, Di Filippo F et al, 2000, Incidence of chromosomes 1 and 17 aneusomy in breast cancer and adjacent tissue: an interphase cytogenetic study. J Am Coll Surg 190:530–539 Tsiambas E, Kravvaritis C, Tsounis D, Salemis NS, Niotis A, Niotis TH et al, 2010, Correlation between different p53 expression patterns and chromosome 17 imbalances in pancreatic ductal adenocarcinoma based on tissue microarray analysis. J BUON 15:94–100 KubiliunN, Ribeiro A, FanYS,Rocha-Lima CM, Sleeman D, Merchan J et al, 2011, EUS-FNA with rescue fluorescence in situ hybridization for the diagnosis of pancreatic carcinoma in patients with inconclusive on-site cytopathology results. Gastrointest Endosc 74:541–754 Reinholz MM, Bruzek AK, Visscher DW, Lingle WL, Schroeder MJ, Perez EA et al, 2009, Breast cancer and aneusomy 17: implications for carcinogenesis and therapeutic response. Lancet Oncol 10:267–277 Koka V, Potti A, Koch M, Fraiman G, Mehdi S, Levitt R, 2002, Role of immunohistochemical identification of Her-2/neu and detection of variability in overexpression in pancreatic carcinoma. Anticancer Res 22:1593–1597 Saxby AJ, Nielsen A, Scarlett CJ, Clarkson A, Morey A, Gill A, Smith RC, 2005, Assessment of HER-2 status in pancreatic adenocarcinoma: correlation of immunohistochemistry, quantitative real-time RT-PCR, and FISH with aneuploidy and survival. Am J Surg Pathol 29:1125–1134 Panani AD, Maliaga K, Babanaraki A, Bellenis I, 2009, Numerical abnormalities of chromosome 9 and p16CDKN2A gene deletion detected by FISH in non-small cell lung cancer. Anticancer Res 29:4483–4487 Mariatos G, Gorgoulis VG, Zacharatos P, Kotsinas A, Vogiatzi T, Rassidakis G et al, 2000, Expression of p16(INK4A, and alterations of the 9p21-23 chromosome region in non-small-cell lung carcinomas: relationship with tumor growth parameters and ploidy status. Int J Cancer 89:133–141 Bruyere H, Sutherland H, Chipperfield K, Hudoba M, 2010, Concomitant and successive amplifications of MYC in APL-like leukemia. Cancer Genet Cytogenet 197:75–80 Zamecnikova A, 2011, Chromosomal translocation t(1;9)(q24; q34, in acute lymphoblastic leukemia patient involving the ABL1 gene. Leuk Res 35:e149–e150 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 35 EP 40 DI 10.1007/s12253-012-9555-3 PG 6 ER PT J AU Boissiere-Michot, F Denouel, A Boulle, N Guillaume, C Orsetti, B Lopez-Crapez, E Chateau, MCh Bibeau, F AF Boissiere-Michot, Florence Denouel, Amelie Boulle, Nathalie Guillaume, Carole Orsetti, Beatrice Lopez-Crapez, Evelyne Chateau, Marie-Christine Bibeau, Frederic TI The Non-Crosslinking Fixative RCL2®-CS100 is Compatible with Both Pathology Diagnosis and Molecular Analyses SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Fixative; Formalin substitute; Nucleic acids; Morphology ID Fixative; Formalin substitute; Nucleic acids; Morphology AB Formalin is the key agent for tissue fixation and pathological diagnosis. However, it poorly preserves nucleic acids and this can impair molecular studies. An alternative to formalin would be a fixative which can allow both morphologic and molecular analyses. To assess the suitability of such a fixative, breast (n011) and colon (n012) tumor samples were fixed in the non cross-linking RCL2®- CS100 fixative and compared to paired formalin-fixed and to frozen samples, the current standards for histology and molecular analyses, respectively. Sections from RCL2®- CS100-fixed samples showed good preservation of cellular and architectural morphology, suitable for routine diagnosis. Although some antibodies required change in the immunohistochemical procedures, quality of the immunohistochemical staining was comparable to that obtained after formalin fixation. HER2 chromogenic in situ hybridization was also successfully performed. High quality DNA could be isolated from RCL2®-CS100-fixed cancer tissues as evidenced by successful amplification of large DNA fragment, CGH array, KRAS and microsatellites genotyping. The quality of RNA from RCL2®-CS100-fixed samples was slightly decreased in comparison to that of RNA isolated from frozen samples, as evidenced by a decreased RNA integrity number but remained exploitable for molecular assays. Our results support the use of the RCL2®-CS100 fixative for histological diagnosis and recovery of high-quality nucleic acids for molecular applications. However, specific procedures for tissue handing and processing, essential to provide high-quality specimens, could limit its use to small target lesions which cannot be frozen without impairing their pathological evaluation. C1 [Boissiere-Michot, Florence] Val d’Aurelle Cancer Institute, Department of Pathology, 34 298 Montpellier, France. [Denouel, Amelie] Val d’Aurelle Cancer Institute, Department of Pathology, 34 298 Montpellier, France. [Boulle, Nathalie] Arnaud de Villeneuve Hospital, Department of Cellular Biology and Hormonology, 34 090 Montpellier, France. [Guillaume, Carole] Val d’Aurelle Cancer Institute, INSERM U896, 34 298 Montpellier, France. [Orsetti, Beatrice] Val d’Aurelle Cancer Institute, INSERM U896, 34 298 Montpellier, France. [Lopez-Crapez, Evelyne] Val d’Aurelle Cancer Institute, Department of Biology, 34 298 Montpellier, France. [Chateau, Marie-Christine] Val d’Aurelle Cancer Institute, Department of Pathology, 34 298 Montpellier, France. [Bibeau, Frederic] Val d’Aurelle Cancer Institute, Department of Pathology, 34 298 Montpellier, France. RP Boissiere-Michot, F (reprint author), Val d’Aurelle Cancer Institute, Department of Pathology, 34 298 Montpellier, France. EM Florence.Boissiere@montpellier.unicancer.fr CR Formaldehyde, 2-Butoxyethanol and 1-tert-Butoxypropan-2-ol. International Agency for Research on Cancer, 2004, IARC Monographs on the Evaluation of Carcinogenic Risks to Humans V. 88 Metz B, Kersten GF, Hoogerhout P, Brugghe HF, Timmermans HA, de Jong A, Meiring H, ten Hove J, Hennink WE, Crommelin DJ, Jiskoot W, 2004, Identification of formaldehyde-induced modifications in proteins: reactions with model peptides. J Biol Chem 279:6235–6243 Plenat F, Montagne K, Weinbreck N, Corby S, Champigneulle J, Antunes L, Bonnet C, Maire C, Monhoven N, 2006, Molecular consequences of fixation and tissue processing: the examples of nucleic acids and proteins. Ann Pathol 26:8–21 McGhee JD, von Hippel PH, 1975, Formaldehyde as a probe of DNA structure. I. Reaction with exocyclic amino groups of DNA bases. Biochemistry 14:1281–1296 Masuda N, Ohnishi T, Kawamoto S, Monden M, Okubo K, 1999, Analysis of chemical modification of RNA from formalin-fixed samples and optimization of molecular biology applications for such samples. Nucleic Acids Res 27:4436–4443 Hewitt SM, Lewis FA, Cao Y, Conrad RC, Cronin M, Danenberg KD, Goralski TJ, Langmore JP, Raja RG, Williams PM, Palma JF, Warrington JA, 2008, Tissue handling and specimen preparation in surgical pathology: issues concerning the recovery of nucleic acids from formalin-fixed, paraffin-embedded tissue. Arch Pathol Lab Med 132:1929–1935 Pikor LA, Enfield KS, Cameron H, Lam WL, 2011, DNA extraction from paraffin embedded material for genetic and epigenetic analyses. JoVE 49., DOI 10.3791/2763 Huijsmans CJ, Damen J, van der Linden JC, Savelkoul PH, Hermans MH, 2010, Comparative analysis of four methods to extract DNA from paraffin-embedded tissues: effect on downstream molecular applications. BMC Res Notes 3:239 Antonov J,Goldstein DR,OberliA, Baltzer A, Pirotta M, Fleischmann A, Altermatt HJ, Jaggi R, 2005, Reliable gene expression measurements from degraded RNA by quantitative real-time PCR depend on short amplicons and a proper normalization. Lab Invest 85:1040–1050 Coudry RA, Meireles SI, Stoyanova R, Cooper HS, Carpino A, Wang X, Engstrom PF, Clapper ML, 2007, Successful application of microarray technology to microdissected formalin-fixed, paraffin-embedded tissue. J Mol Diagn 9:70–79 Cronin M, Pho M, Dutta D, Stephans JC, Shak S, Kiefer MC, Esteban JM, Baker JB, 2004, Measurement of gene expression in archival paraffin-embedded tissues: development and performance of a 92-gene reverse transcriptase-polymerase chain reaction assay. Am J Pathol 164:35–42 Paris PL, 2009, Awhole-genome amplification protocol for a wide variety of DNAs, including those from formalin-fixed and paraffin-embedded tissue. Methods Mol Biol 556:89–98 Penland SK, Keku TO, Torrice C, He X, Krishnamurthy J, Hoadley KA,Woosley JT, Thomas NE, Perou CM, Sandler RS, Sharpless NE, 2007, RNA expression analysis of formalin-fixed paraffinembedded tumors. Lab Invest 87:383–391 Goldstein LJ, Gray R, Badve S, Childs BH, Yoshizawa C, Rowley S, Shak S, Baehner FL, Ravdin PM, Davidson NE, Sledge GW Jr, Perez EA, Shulman LN, Martino S, Sparano JA, 2008, Prognostic utility of the 21-gene assay in hormone receptor-positive operable breast cancer compared with classical clinicopathologic features. J Clin Oncol 26:4063–4071 Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, Baehner FL, Walker MG, Watson D, Park T, Hiller W, Fisher ER, Wickerham DL, Bryant J, Wolmark N, 2004, A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 351:2817–2826 Paik S, Tang G, Shak S, Kim C, Baker J, Kim W, Cronin M, Baehner FL, Watson D, Bryant J, Costantino JP, Geyer CE Jr, Wickerham DL, Wolmark N, 2006, Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptorpositive breast cancer. J Clin Oncol 24:3726–3734 Bueno-de-Mesquita JM, van Harten WH, Retel VP, van't Veer LJ, van Dam FS, Karsenberg K, Douma KF, van Tinteren H, Peterse JL, Wesseling J, Wu TS, Atsma D, Rutgers EJ, Brink G, Floore AN, Glas AM, Roumen RM, Bellot FE, van Krimpen C, Rodenhuis S, van de Vijver MJ, Linn SC, 2007, Use of 70-gene signature to predict prognosis of patients with node-negative breast cancer: a prospective community-based feasibility study, RASTER). Lancet Oncol 8:1079–1087 Buyse M, Loi S, van't Veer L, Viale G, Delorenzi M, Glas AM, d'Assignies MS, Bergh J, Lidereau R, Ellis P, Harris A, Bogaerts J, Therasse P, Floore A, Amakrane M, Piette F, Rutgers E, Sotiriou C, Cardoso F, Piccart MJ, 2006, Validation and clinical utility of a 70- gene prognostic signature for women with node-negative breast cancer. J Natl Cancer Inst 98:1183–1192 Glas AM, Floore A, Delahaye LJ, Witteveen AT, Pover RC, Bakx N, Lahti-Domenici JS, Bruinsma TJ, Warmoes MO, Bernards R, Wessels LF, van't Veer LJ, 2006, Converting a breast cancer microarray signature into a high-throughput diagnostic test. BMC Genomics 7:278., DOI 10.1186/1471-2164-7-278 van de Vijver MJ, He YD, van't Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, Parrish M, Atsma D, Witteveen A, Glas A, Delahaye L, van d V, Bartelink H, Rodenhuis S, Rutgers ET, Friend SH, Bernards R, 2002, A geneexpression signature as a predictor of survival in breast cancer. N Engl J Med 347:1999–2009 Denouel A, Boissiere-Michot F, Rochaix P, Bibeau F, Boulle N, 2011, An alternative fixative to formalin fixation for molecular applications: the RCL2®-CS100 approach. Methods Mol Biol 724:297–307 Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, McShane LM, Paik S, Pegram MD, Perez EA, Press MF, Rhodes A, Sturgeon C, Taube SE, Tubbs R, Vance GH, van de Vijver M, Wheeler TM, Hayes DF, American Society of Clinical Oncology/ College of American Pathologists, 2007, American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 131:18–43 Boissiere-Michot F, Lopez-Crapez E, Frugier H, Berthe ML, Ho-Pun-Cheung A, Assenat E, Maudelonde T, Lamy PJ, Bibeau F, 2012, KRAS genotyping in rectal adenocarcinoma specimens with low tumor cellularity after neoadjuvant treatment. Mod Pathol 25:731–739 Lopez-Crapez E, Chypre C, Saavedra J, Marchand J, Grenier J, 1997, Rapid and large-scale method to detect K-ras gene mutations in tumor samples. Clin Chem 43:936–942 Orsetti B, Nugoli M, Cervera N, Lasorsa L, Chuchana P, Rouge C, Ursule L, Nguyen C, Bibeau F, Rodriguez C, Theillet C, 2006, Genetic profiling of chromosome 1 in breast cancer: mapping of regions of gains and losses and identification of candidate genes on 1q. Br J Cancer 95:1439–1447 Bellet V, Boissiere F, Bibeau F, Desmetz C, Berthe ML, Rochaix P, Maudelonde T, Mange A, Solassol J, 2008, Proteomic analysis of RCL2 paraffin-embedded tissues. J Cell Mol Med 12:2027–2036 Delfour C, Roger P, Bret C, Berthe ML, Rochaix P, Kalfa N, Raynaud P, Bibeau F, Maudelonde T, Boulle N, 2006, RCL2, a new fixative, preserves morphology and nucleic acid integrity in paraffin-embedded breast carcinoma and microdissected breast tumor cells. J Mol Diagn 8:157–169 Lassalle S, Hofman V, Marius I, Gavric-Tanga V, Brest P, Havet K, Butori C, Selva E, Santini J, Mograbi B, Hofman P, 2009, Assessment of morphology, antigenicity, and nucleic acid integrity for diagnostic thyroid pathology using formalin substitute fixatives. Thyroid 19:1239–1248 Preusser M, Plumer S, Dirnberger E, Hainfellner JA, Mannhalter C, 2010, Fixation of brain tumor biopsy specimens with RCL2 results in well-preserved histomorphology, immunohistochemistry and nucleic acids. Brain Pathol 20:1010–1020 Tawfik O, Kimler BF, Davis M, Stasik C, Lai SM, Mayo MS, Fan F, Donahue JK, Damjanov I, Thomas P, Connor C, Jewell WR, Smith H, Fabian CJ, 2007, Grading invasive ductal carcinoma of the breast: advantages of using automated proliferation index instead of mitotic count. Virchows Arch 450:627–636 Williams C, Ponten F, Moberg C, Soderkvist P, Uhlen M, Ponten J, Sitbon G, Lundeberg J, 1999, A high frequency of sequence alterations is due to formalin fixation of archival specimens. Am J Pathol 155:1467–1471 Li J, Smyth P, Cahill S, Denning K, Flavin R, Aherne S, Pirotta M, Guenther SM, O'Leary JJ, Sheils O, 2008, Improved RNA quality and TaqMan Pre-amplification method, PreAmp, to enhance expression analysis from formalin fixed paraffin embedded, FFPE, materials. BMC Biotechnol 8:10., DOI 10.1186/1472-6750-8-10 McKinney MD, Moon SJ, Kulesh DA, Larsen T, Schoepp RJ, 2009, Detection of viral RNA from paraffin-embedded tissues after prolonged formalin fixation. J Clin Virol 44:39–42 Ribeiro-Silva A, Zhang H, Jeffrey SS, 2007, RNA extraction from ten year old formalin-fixed paraffin-embedded breast cancer samples: a comparison of column purification and magnetic beadbased technologies. BMC Mol Biol 8:118., DOI 10.1186/1471- 2199-8-118 Dotti I, Bonin S, Basili G, Nardon E, Balani A, Siracusano S, Zanconati F, Palmisano S, De Manzini N, Stanta G, 2010, Effects of formalin, methacarn, and fineFIX fixatives on RNA preservation. Diagn Mol Pathol 19:112–122 von Ahlfen S, Missel A, Bendrat K, Schlumpberger M, 2007, Determinants of RNA quality from FFPE samples. PLoS One 2: e1261., DOI 10.1371/journal.pone.0001261 Bustin SA, Nolan T, 2004, Pitfalls of quantitative real-time reverse-transcription polymerase chain reaction. J Biomol Tech 15:155–166 Ho-Pun-Cheung A, Bascoul-Mollevi C, Assenat E, Boissiere- Michot F, Bibeau F, Cellier D, Ychou M, Lopez-Crapez E, 2009, Reverse transcription-quantitative polymerase chain reaction: description of a RIN-based algorithm for accurate data normalization. BMC Mol Biol 10:31., DOI 10.1186/1471-2199-10-31 Strand C, Enell J, Hedenfalk I, Ferno M, 2007, RNA quality in frozen breast cancer samples and the influence on gene expression analysis–a comparison of three evaluation methods using microcapillary electrophoresis traces. BMC Mol Biol 8:38 Eltoum I, Fredenburgh HT, Myers R, Grizzle W, 2001, Introduction to the theory and practice of fixation of tissue. J Histotechnol 24:173–192 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 41 EP 53 DI 10.1007/s12253-012-9556-2 PG 13 ER PT J AU Juasook, A Boonmars, Th Wu, Z Loilome, W Veteewuthacharn, K Namwat, N Sudsarn, P Wonkchalee, O Sriraj, P Aukkanimart, R AF Juasook, Amornrat Boonmars, Thidarut Wu, Zhiliang Loilome, Watcharin Veteewuthacharn, Kulathida Namwat, Nissana Sudsarn, Pakkayanee Wonkchalee, Orasa Sriraj, Pranee Aukkanimart, Ratchadawan TI Immunosuppressive Prednisolone Enhances Early Cholangiocarcinoma in Syrian Hamsters with Liver Fluke Infection and Administration of N-nitrosodimethylamine SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Liver fluke; Cholangiocarcinogenesis; Immunosuppressed; Chronic inflammation; Histopathology ID Liver fluke; Cholangiocarcinogenesis; Immunosuppressed; Chronic inflammation; Histopathology AB Chronic infection with Opisthorchis viverrini for many years has been associated with the development of hepatobiliary diseases including cholangiocarcinoma. It is well known that inflammation is a key component of the tumormicroenvironment, and that chronic inflammation plays an important role in tumorigenesis. Therefore, in this study cholangiocarcinogenesis was induced in Syrian hamsters in order to observe the cancer-related inflammation. The Syrian hamsters were divided into 5 groups: uninfected controls; normal Syrian hamsters infected with O. viverrini (OV); immunosuppressed Syrian hamsters infected with O. viverrini (OVis); normal Syrian hamsters infected with O. viverrini and administered N-nitrosodimethylamine (CCA); and immunosuppressed Syrian hamsters infected with O. viverrini and administered N-nitrosodimethylamine (CCAis). Syrian hamster livers were later observed for gross pathology and histopathological changes; COX2 was analyzed by immunohistochemical staining. We found a decreased number of inflammatory cells surrounding the hepatic bile duct in the OVis group, but not in the OV and CCAis groups. However, in the CCAis group (with suppressed immunity) early appearance and greater severity of cholangiocarcinoma were observed; gross pathological examination revealed many cancer nodularities on the liver surface, and histopathological studies showed the presence of cancer cells, findings which correlated with the predominant expression of COX2. The present study suggests that host immune responses are intended to ameliorate pathology, and they are also crucially associated with pathogenesis in O. viverrini infection; the unbalancing of host immunity may enhance cancer-related inflammation. C1 [Juasook, Amornrat] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Boonmars, Thidarut] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Wu, Zhiliang] Gifu University, Graduate School of Medicine, Department of Parasitology, Yanagido1-1, 501-1194 Gifu, Japan. [Loilome, Watcharin] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Veteewuthacharn, Kulathida] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Namwat, Nissana] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Sudsarn, Pakkayanee] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Wonkchalee, Orasa] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Sriraj, Pranee] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Aukkanimart, Ratchadawan] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. RP Boonmars, Th (reprint author), Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. EM boonmars@yahoo.com;bthida@kku.ac.th CR Sripa B, Kaewkes S, Sithithaworn P, Mairiang E, Laha T, Smout M, Pairojkul C, Bhudhisawasdi V, Tesana S, Thinkamrop B, Bethony JM, Loukas A, Brindley PJ, 2007, Liver fluke induces cholangiocarcinoma. PLoS Med 4(7):e201 IARC, 2011, Opisthorchis viverrini and Clonorchis sinensis IARC. Monogr Eval Carcinog Risks Hum 100(7):347–376 Sripa B, 2003, Pathobiology of opisthorchiasis: an update. Acta Trop 88(3):209–220 Pinlaor S, Hiraku Y, Ma N, Yongvanit P, Semba R, Oikawa S, Murata M, Sripa B, Sithithaworn P, Kawanishi S, 2004, Mechanism of NO-mediated oxidative and nitrative DNA damage in hamsters infected with Opisthorchis viverrini: a model of inflammation-mediated carcinogenesis. Nitric Oxide 11(2):175– 183 Ames BN, Shigenaga MK, Hagen TM, 1993, Oxidants, antioxidants, and the degenerative diseases of aging. Proc Natl Acad Sci U S A 90(17):7915–7922 Coussens LM, Werb Z, 2002, Inflammation and cancer. Nature 420(6917):860–867 Boonmars T, Srisawangwong T, Srirach P, Kaewsamut B, Pinlaor S, Sithithaworn P, 2007, Apoptosis-related gene expressions in hamsters re-infected with Opisthorchis viverrini and re-treated with praziquantel. Parasitol Res 102(1):57–62 Ohshima H, Bandaletova TY, Brouet I, Bartsch H, Kirby G, Ogunbiyi F, Vatanasapt V, Pipitgool V, 1994, Increased nitrosamine and nitrate biosynthesis mediated by nitric oxide synthase induced in hamsters infected with liver fluke, Opisthorchis viverrini). Carcinogenesis 15(2):271–275 Mantovani A, Allavena P, Sica A, Balkwill F, 2008, Cancerrelated inflammation. Nature 454(7203):436–444 Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A, 2009, Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 30(7):1073–1081 Boonmars T, Srirach P, Kaewsamut B, Srisawangwong T, Pinlaor S, Pinlaor P, Yongvanit P, Sithithaworn P, 2008, Apoptosis-related gene expression in hamster opisthorchiasis post praziquantel treatment. Parasitol Res 102(3):447–455 Boonmars T, Boonjaraspinyo S, Kaewsamut B, 2009, Animal models for Opisthorchis viverrini infection. Parasitol Res 104, 3):701–703 Boonmars T, Wu Z, Boonjaruspinyo S, Puapairoj A, Kaewsamut B, Nagano I, Pinlaor S, Yongvanit P, Wonkchalee O, Juasook A, Sudsarn P, Srisawangwong T, 2010, Involvement of c-Ski oncoprotein in carcinogenesis of cholangiocacinoma induced by Opisthorchis viverrini and N-nitrosodimethylamine. Pathol Oncol Res 17(2):219–227 Trune DR, Kempton JB, Gross ND, 2006, Mineralocorticoid receptor mediates glucocorticoid treatment effects in the autoimmune mouse ear. Hear Res 212(1–2):22–32 Boonjaraspinyo S, Boonmars T, Aromdee C, Puapairoj A, Wu Z, 2011, Indirect effect of a turmeric diet: enhanced bile duct proliferation in Syrian hamsters with a combination of partial obstruction by Opisthorchis viverrini infection and inflammation by N-nitrosodimethylamine administration. Parasitol Res 108(1):7–14 Martin Sanz PHS, Bosca L, Casado M, 2006, Cyclooxygenase 2: understanding the pathophysiological role through genetically altered mouse models. Front Biosci 11:2876–2888 Takasu S, Tsukamoto T, Cao XY, Toyoda T, Hirata A, Ban H, Yamamoto M, Sakai H, Yanai T, Masegi T, Oshima M, Tatematsu M, 2008, Roles of cyclooxygenase-2 and microsomal prostaglandin E synthase-1 expression and beta-catenin activation in gastric carcinogenesis in N-methyl-N-nitrosourea-treated K19-C2mE transgenic mice. Cancer Sci 99(12):2356–2364 Sirica AE, Lai GH, Zhang Z, 2001, Biliary cancer growth factor pathways, cyclo-oxygenase-2 and potential therapeutic strategies. J Gastroenterol Hepatol 16(4):363–372 Endo K, Yoon BI, Pairojkul C, Demetris AJ, Sirica AE, 2002, ERBB-2 overexpression and cyclooxygenase-2 up-regulation in human cholangiocarcinoma and risk conditions. Hepatology 36, 2):439–450 Onizuka S, Tawara I, Shimizu J, Sakaguchi S, Fujita T, Nakayama E, 1999, Tumor rejection by in vivo administration of anti-CD25, interleukin-2 receptor alpha, monoclonal antibody. Cancer Res 59, 13):3128–3133 El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, Herrera J, Lissowska J, Yuan CC, Rothman N, Lanyon G, Martin M, Fraumeni JF Jr, Rabkin CS, 2000, Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature 404(6776):398–402 de Martel C, Llosa AE, Farr SM, Friedman GD, Vogelman JH, Orentreich N, Corley DA, Parsonnet J, 2005, Helicobacter pylori infection and the risk of development of esophageal adenocarcinoma. J Infect Dis 191(5):761–767 Zamarron BF, ChenW, 2011, Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci 7, 5):651–658 Grivennikov SI, Greten FR, Karin M, 2010, Immunity, inflammation, and cancer. Cell 140(6):883–899 Barth H, Robinet E, Liang TJ, Baumert TF, 2008, Mouse models for the study of HCV infection and virus-host interactions. J Hepatol 49(1):134–142 Park EJ, Lee JH, Yu GY, He G, Ali SR, Holzer RG, Osterreicher CH, Takahashi H, Karin M, 2010, Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 140(2):197–208 Lindor KD, Wiesner RH, Colwell LJ, Steiner B, Beaver S, LaRusso NF, 1991, The combination of prednisone and colchicine in patients with primary sclerosing cholangitis. Am J Gastroenterol 86(1):57–61 Angulo P, Batts KP, Jorgensen RA, LaRusso NA, Lindor KD, 2000, Oral budesonide in the treatment of primary sclerosing cholangitis. Am J Gastroenterol 95(9):2333–2337 Kleiman A, Tuckermann JP, 2007, Glucocorticoid receptor action in beneficial and side effects of steroid therapy: lessons from conditional knockout mice. Mol Cell Endocrinol 275(1–2):98–108 Giljaca V, Poropat G, Stimac D, Gluud C, 2010, Glucocorticosteroids for primary sclerosing cholangitis. Cochrane Database Syst Rev 1:CD004036 El-Awady RA, Saleh EM, Ezz M, Elsayed AM, 2011, Interaction of celecoxib with different anti-cancer drugs is antagonistic in breast but not in other cancer cells. Toxicol Appl Pharmacol 255, 3):271–286 Hayashi S, Sumi Y, Ueno N, Murase A, Takada J, 2011, Discovery of a novel COX-2 inhibitor as an orally potent anti-pyretic and anti-inflammatory drug: design, synthesis, and structure-activity relationship. Biochem Pharmacol 82(7):755–768 Roger T, Chanson AL, Knaup-Reymond M, Calandra T, 2005, Macrophage migration inhibitory factor promotes innate immune responses by suppressing glucocorticoid-induced expression of mitogen-activated protein kinase phosphatase-1. Eur J Immunol 35(12):3405–3413 Musil R, Schwarz MJ, Riedel M, Dehning S, Cerovecki A, Spellmann I, Arolt V, Muller N, 2011, Elevated macrophage migration inhibitory factor and decreased transforming growth factor-beta levels in major depression–no influence of celecoxib treatment. J Affect Disord 134(1–3):217–225 Coleman RE, 1992, Glucocorticoids in cancer therapy. Biotherapy 4(1):37–44 Small AJ, Loftus CG, Smyrk TC, Baron TH, 2008, A case of IgG4-associated cholangitis and autoimmune pancreatitis responsive to corticosteroids. Nat Clin Pract Gastroenterol Hepatol 5, 12):707–712 Fracchia M, Secreto P, Tabone M, Zaffino C, Pera A, Galatola G, 2000, Serum interferon gamma in primary biliary cirrhosis: effect of ursodeoxycholic acid and prednisone therapy alone and in combination. Eur J Gastroenterol Hepatol 12(4):463– 468 Yao LC, Baluk P, Feng J, McDonald DM, 2010, Steroid-resistant lymphatic remodeling in chronically inflamed mouse airways. Am J Pathol 176(3):1525–1541 Chapple KS, Cartwright EJ, Hawcroft G, Tisbury A, Bonifer C, Scott N, Windsor AC, Guillou PJ, Markham AF, Coletta PL, Hull MA, 2000, Localization of cyclooxygenase-2 in human sporadic colorectal adenomas. Am J Pathol 156(2):545–553 McLean MH, Murray GI, Fyfe N, Hold GL, Mowat NA, El-Omar EM, 2008, COX-2 expression in sporadic colorectal adenomatous polyps is linked to adenoma characteristics. Histopathology 52, 7):806–815 Jarnicki AG, Lysaght J, Todryk S, Mills KH, 2006, Suppression of antitumor immunity by IL-10 and TGF-beta-producing T cells infiltrating the growing tumor: influence of tumor environment on the induction of CD4+ and CD8+ regulatory T cells. J Immunol 177(2):896–904 Tiano HF, Loftin CD, Akunda J, Lee CA, Spalding J, Sessoms A, Dunson DB, Rogan EG, Morham SG, Smart RC, Langenbach R, 2002, Deficiency of either cyclooxygenase, COX)-1 or COX-2 alters epidermal differentiation and reduces mouse skin tumorigenesis. Cancer Res 62(12):3395–3401 An KP, AtharM, Tang X, Katiyar SK, Russo J, Beech J, Aszterbaum M, Kopelovich L, Epstein EH Jr, Mukhtar H, Bickers DR, 2002, Cyclooxygenase-2 expression in murine and human nonmelanoma skin cancers: implications for therapeutic approaches. Photochem Photobiol 76(1):73–80 Nakanuma YSB, Vatanasapt V, Leong A, Ponchon T, Ishak K, 2000, Intrahepatic cholangiocarcinoma. In: Hamilton SR, Aaltonen LA, eds, WHO classification of tumors, pathology and genetics. Tumours of the digestive system. IARC, Lyon, pp 173–180 Jinawath N, Chamgramol Y, Furukawa Y, Obama K, Tsunoda T, Sripa B, Pairojkul C, Nakamura Y, 2006, Comparison of gene expression profiles between Opisthorchis viverrini and non- Opisthorchis viverrini associated human intrahepatic cholangiocarcinoma. Hepatology 44(4):1025–1038 Hawrylowicz CM, 2005, Regulatory T cells and IL-10 in allergic inflammation. J Exp Med 202(11):1459–1463 Rubtsov YP, Rasmussen JP, Chi EY, Fontenot J, Castelli L, Ye X, Treuting P, Siewe L, Roers A, Henderson WR Jr, Muller W, Rudensky AY, 2008, Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 4):546–558 Barnes PJ, 2011, Glucocorticosteroids: current and future directions. Br J Pharmacol 163(1):29–43 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 55 EP 62 DI 10.1007/s12253-012-9557-1 PG 8 ER PT J AU Patonai, A Erdelyi-Belle, B Korompay, A Somoracz, Torzsok, P Kovalszky, I Barbai, T Raso, E Lotz, G Schaff, Zs Kiss, A AF Patonai, Attila Erdelyi-Belle, Boglarka Korompay, Anna Somoracz, Aron Torzsok, Peter Kovalszky, Ilona Barbai, Tamas Raso, Erzsebet Lotz, Gabor Schaff, Zsuzsa Kiss, Andras TI Molecular Characteristics of Fibrolamellar Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Fibrolamellar hepatocellular carcinoma; Hepatocellular carcinoma; Cholangiocellular carcinoma; Epidermal growth factor receptor; K-RAS mutation ID Fibrolamellar hepatocellular carcinoma; Hepatocellular carcinoma; Cholangiocellular carcinoma; Epidermal growth factor receptor; K-RAS mutation AB Fibrolamellar hepatocellular carcinoma (FLC) occurs in non-cirrhotic liver and the etiopathogenesis is still obscure. Both hepatocellular and cholangiocellular markers are expressed in the tumor, however, molecular alterations and altered pathways playing role in the tumor pathogenesis are not clearly identified. The purpose of the present study was to compare the expression level of EGFR, syndecan-1 and ßcatenin in FLC, conventional hepatocellular carcinoma (cHCC) and cholangiocellular carcinoma (CCC) and to investigate the possibility ofmutation both in EGFR and K-RAS. Eight FLCs were compared with 7 cHCCs, 7 CCCs and 5 normal liver samples. Cytokeratins 7, 8, 18, 19, HepPar1 (HSA), EGFR, syndecan-1 (CD138) and ß-catenin were detected by immunohistochemistry. In addition EGFR, ß-catenin and syndecan-1 were evaluated by digital morphometry and K-RAS, EGFR mutations in FLC cases using paraffin-embedded samples. All FLCs were positive for HepPar1 (HSA) and cytokeratins 7, 8, 18, but negative for cytokeratin 19 by immunohistochemistry. EGFR was significantly overexpressed in all three tumor types, being highest in FLCs (p<0,0001). EGFR, K-RAS mutation analyses revealed no mutations in exons studied in FLCs. Our findings proved that expression of EGFR is higher in FLC than in other types of primary malignant hepatic tumors and no KRAS mutation can be detected, so FLC is a good candidate for anti-EGFR treatment. C1 [Patonai, Attila] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Erdelyi-Belle, Boglarka] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Korompay, Anna] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Somoracz, Aron] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Torzsok, Peter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Barbai, Tamas] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Raso, Erzsebet] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM schaff.zsuzsa@med.semmelweis-univ.hu CR Abdul-Al HM, Wang G, Makhlouf HR, Goodman ZD, 2010, Fibrolamellar hepatocellular carcinoma: An immunohistochemical comparison with conventional hepatocellular carcinoma. Int J Surg Pathol 18:313–318 Kannangai R, Vivekanandan P, Martinez-Murillo F, Choti M, TorbensonM, 2007, Fibrolamellar carcinomas show overexpression of genes in the RAS, MAPK, PIK3, and xenobiotic degradation pathways. Hum Pathol 38:639–644 Liu S, Chan KW, Wang B, Qiao L, 2009, Fibrolamellar hepatocellular carcinoma. Am J Gastroenterol 104:2617–2624 Torbenson M, 2007, Review of the clinicopathologic features of fibrolamellar carcinoma. Adv Anat Pathol 14:217–223 Ward SC, Huang J, Tickoo SK, Thung SN, Ladanyi M, Klimstra DS, 2010, Fibrolamellar carcinoma of the liver exhibits immunohistochemical evidence of both hepatocyte and bile duct differentiation. Mod Pathol 23:1180–1190 Patonai A, Erdelyi-Belle B, Korompay A, Somoracz A, Straub BK, Schirmacher P, Kovalszky I, Lotz G, Kiss A, Schaff Z, 2011, Claudins and tricellulin in fibrolamellar hepatocellular carcinoma. Virchows Arch 458:679–688 Kakar S, Chen X, Ho C, Burgart LJ, Sahai V, Dachrut S, Yabes A, Jain D, Ferrell LD, 2009, Chromosomal changes in fibrolamellar hepatocellular carcinoma detected by array comparative genomic hybridization. Hum Pathol 22:134–141 International Consensus Group for Hepatocellular Neoplasia, 2009, Pathologic diagnosis of early hepatocellular carcinoma: A report of the International Consensus Group for Hepatocellular Neoplasia. Hepatology 49:658–664 Libbrecht L, Severi T, Cassiman D, Vander Borght S, Pirenne J, Nevens F, Verslype C, van Pelt J, Roskams T, 2006, Glypican-3 expression distinguishes small hepatocellular carcinomas from cirrhosis, dysplastic nodules, and focal nodular hyperplasia-like nodules. Am J Surg Pathol 30:1405–1411 Sirivatanauksorn Y, Sirivatanauksorn V, Lemoine NR, Williamson BR, Davidson BR, 2001, Genomic homogeneity in fibrolamellar carcinomas. Gut 49:82–86 Vivekanandan P, Torbenson M, 2008, Epigenetic insility is rare in fibrolamellar carcinomas but common in viral-associated hepatocellular carcinomas. Mod Pathol 21:670–675 Buckley AF, Burgart LJ, Kakar S, 2006, Epidermal growth factor receptor expression and gene copy number in fibrolamellar hepatocellular carcinoma. Hum Pathol 37:410–414 Llovet JM, Bruix J, 2008, Novel advancements in the management of hepatocellular carcinoma in 2008. J Hepatol 48: S20–S37 Ishak KG, Goodman ZD, Stocker JT, 2001, Tumors of the Liver and Intrahepatic Bile Ducts. Atlas of Tumor Pathology, Third Series, Fascicle 31. Armed Forces Institute of Pathology, Washington, DC Ikari A, Atomi K, Takiguchi A, Yamazaki Y, Miwa M, Sugatani J, 2009, Epidermal growth factor increases claudin-4 expression mediated by Sp1 elevation in MDCK cells. Biochem Biophys Res Commun 384:306–310 Kojima T, Murata M, Yamamoto T, Lan M, Imamura M, Son S, Takano K, Yamaguchi H, Ito T, Tanaka S, Chiba H, Hirata K, Sawada N, 2009, Tight junction proteins and signal transduction pathways in hepatocytes. Histol Histopathol 24:1463–1472 Cieply B, Zeng G, Proverbs-Singh T, Geller DA, Monga SPS, 2009, Unique phenotype of hepatocellular cancers with exon-3 mutations in beta-catenin gene. Hepatology 49:821–831 Vivekanandan P, Daniel H, Yeh MM, Torbenson M, 2010, Mitochondrial mutations in hepatocellular carcinomas and fibrolamellar carcinomas. Mod Pathol 23:790–798 Li W, Tan D, Zenali MJ, Brown RE, 2010, Constitutive activation of nuclear factor- kappa B, NF-kB, signaling pathway in fibrolamellar hepatocellular carcinoma. Int J Clin Exp Pathol 3:238–243 Sethi S, Tageja N, Singh J, Arabi H, Dave M, Badheka A, Revankar S, 2009, Hyperammonemic encephalopathy: A rare presentation of fibrolamellar hepatocellular carcinoma. Am J Med Sci 338:522–524 Roskams T, De Vos R, Van Damme DG, Desmet V, 1998, Heparan sulphate proteoglycan expression in primary liver tumors. J Pathol 185:190–297 Li H-G, Xie D-R, Shen X-M, Li H-H, Zeng H, Zeng Y-J, 2005, Clinicopathological significance of expression of paxilin, syndecan-1 and EMMPRIN in hepatocellular carcinoma. W J Gastroent 11:1445–1451 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 63 EP 70 DI 10.1007/s12253-012-9558-0 PG 8 ER PT J AU Bezic, J Samija-Projic, I Projic, P Ljubkovic, J Tomas-Zekic, S Marinovic-Guic, M Tomic, S AF Bezic, Josko Samija-Projic, Ivana Projic, Petar Ljubkovic, Jelena Tomas-Zekic, Sandra Marinovic-Guic, Maja Tomic, Snjezana TI Near-Diploid Hyperploidy in Early Breast Cancer (T1a,b) is Associated with Higher Risk of Lymph Node Involvement SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Early breast cancer; Nodal metastases; DNA flow cytometry; DNA index ID Early breast cancer; Nodal metastases; DNA flow cytometry; DNA index AB Due to the worldwide implementation of the mammographic screening program early breast cancer (T1a,b) has become more prevalent form of breast cancer. Although T1a,b breast cancers are generally associated with excellent prognosis, some of them, particularly those with lymph node involvement, has unfavourable outcome. Searching for additional prognostic factors, we investigated DNA content of 163 T1a,b cancers measured by DNA flow cytometry, and correlated it with regional lymph node status. T1a,b cancers were divided into four ploidy classes based on their DNA index (DI): hypodiploid (DI<0.95), diploid (DI 0.95–1.05), low-hyperploid (DI 1.06–1.3), and highhyperploid (DI>1.3). Diploid T1a,b cancers were associated with negative lymph node status (p<0.003). Among aneuploid cancers only low-hyperploid tumors were associated with positive lymph node status (p<0.03). The histopathological features of low-hyperploid group of T1a,b cancers did not differ from the other three ploidy groups of cancers, except for lower S-phase fraction of tumor cells in lowhyperploid group compared to high-hyperploid group (p<0.01). Our data showed that near-diploid hyperploid T1a,b cancers are associated with higher risk of lymph node involvement despite similar clinicopathological features shared with other ploidy classes of T1a,b tumors. C1 [Bezic, Josko] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Pathology, Forensic Medicine and Cytology, Spinciceva 1, 21 000 Split, Croatia. [Samija-Projic, Ivana] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Pathology, Forensic Medicine and Cytology, Spinciceva 1, 21 000 Split, Croatia. [Projic, Petar] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Pathology, Forensic Medicine and Cytology, Spinciceva 1, 21 000 Split, Croatia. [Ljubkovic, Jelena] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Pathology, Forensic Medicine and Cytology, Spinciceva 1, 21 000 Split, Croatia. [Tomas-Zekic, Sandra] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Pathology, Forensic Medicine and Cytology, Spinciceva 1, 21 000 Split, Croatia. [Marinovic-Guic, Maja] University of Split, School of MedicineSplit, Croatia. [Tomic, Snjezana] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Pathology, Forensic Medicine and Cytology, Spinciceva 1, 21 000 Split, Croatia. RP Bezic, J (reprint author), University of Split, School of Medicine, Clinical Hospital Center Split, Department of Pathology, Forensic Medicine and Cytology, 21 000 Split, Croatia. EM jbezic@mefst.hr CR Ernster VL, Barclay J, Kerlikowski K, Wilkie H, Ballard-Barbash R, 2000, Mortality among women with ductal carcinoma in situ of the breast in the population-based surveillance, epidemiology and end results program. Arch Int Med 160:953–958 Abner AL, Collins L, Peiro G, Recht A, Come S, Shulman LN, Silver B, Nixon A, Harris JR, Schnitt SJ, Connolly JL, 1998, Correlation of tumor size and axillary lymph node involvement with prognosis in patients with T1 breast carcinoma. Cancer 83:2502–2508 Kerlikowske K, Grady D, Rubin SM, Sandrock C, Ernster VL, 1995, Efficacy of screening mammography. A meta-analysis. JAMA 273:149–154 Chen YY, Schnitt SJ, 1998, Prognostic factors for patients with breast cancers 1 cm and smaller. Breast Cancer Res Treat 51:209– 225 Hanrahan EO, Valero V, Gonzales-Angulo AM, Hortobagyi GN, 2006, Prognosis and management of patients with node-negative invasive breast carcinoma that is 1 cm or smaller in size, stage 1; T1a, bN0M0): a review of the literature. J Clin Oncol 24:2113– 2122 Arnesson LG, Smeds S, Fagerberg G, 1994, Recurrence-free survival in patients with small breast cancers. Eur J Surg 160:271–276 Fentiman IS, Hyland MA, Chaudary MA, Gregory WM, 1996, Prognosis of patients with breast cancers up to 1 cm in diametar. Eur J Cancer 32:417–420 Bezic J, Samija-Projic I, Projic P, Ljubkovic J, Capkun V, Tomic S, 2011, Can we identify the group of small invasive, T1a, b, breast cancers with minimal risk of axillary lymph node involvement? A pathohistological and DNA flow cytometric study. Pathol Res Pract 207:438–442 Tavassoli FA, Devilee P, 2003, World Health Organization of tumors: pathology and genetics of tumors of the breast and female genital organs. IARC Press, Lyon Elston CW, Ellis IO, Goulding H, Pinder SE, 1998, Role of pathology in the prognosis and management of breast cancer. In: Elston CW, Ellis IO, eds, The breast, 3rd edn. Churchill Livingstone, Edinburgh, pp 385–433 Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19:403–410 Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, Fitzgibbons PL, Francis G, Goldstein NS, Hayes M, Hicks DG, Lester S, Love R, Mangu PB, McShane L, Miller K, Osborne CK, Paik S, Perlmutter J, Rhodes A, Sasano H, Schwartz JN, Sweep FC, Taube S, Torlakovic EE, Valenstein P, Viale G, Visscher D, Wheeler T, Williams RB, Wittliff JL, Wolff AC, 2010, American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol 28:2784–2795 Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, McShane LM, Paik S, Pegram MD, Perez EA, Press MF, Rhodes A, Sturgeon C, Taube SE, Tubbs R, Vance GH, van de Vijver M, Wheeler TM, Hayes DF, 2007, American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol 25:118–145 Hedley DW, Friedlander ML, Taylor IW, Rugg CA, Musgrove EA, 1983, Method for analysis of cellular DNA-content of paraffinembedded pathological material using flow cytometry. J Histochem Cytochem 31:1333–1335 Anderson TJ, Waller M, Ellis IO, Bobrow L, Moss S, 2004, Influence of annual mammography from age 40 on breast cancer pathology. Hum Pathol 35:1251–1259 Kirshtein B, Crystal P, Koretz M, Strano S, 2004, Dedicated screening mammography for diagnosis of small breast cancer. World J Surg 28:232–235 Ottesen GL, Christensen IJ, Larsen JK, Kerndrup GB, Hansen B, Andersen JA, 1995, DNA aneuploidy in early breast cancer. Br J Cancer 72:832–839 Ewers SB, Langstrom E, Baldetorp B, Killander D, 1984, Flowcytometric DNA analysis in primary breast carcinomas and clinicopathological correlations. Cytometry 5:408–419 Blegen H, Ghadimi BM, Jauho A, Zetterberg A, Eriksson E, Auer G, Ried T, 2001, Genetic instability promotes the acquisition of chromosomal imbalances in T1b and T1c breast adenocarcinomas. Anal Cell Pathol 22:123–131 Chadha M, Chabon AB, Friedmann P, Vikram B, 1994, Predictors of axillary lymph node metastases in patients with T1 breast cancer: a multivariate analysis. Cancer 73:350–353 Zanon C, Durando A, Geuna M, Clara R, Mobiglia A, Massobrio M, Palestro G, Pourshayesteh A, 1998, Flow cytometry in breast cancer: prognostic and surgical indications of the sparing of axillary lymph node dissection. Am J Clin Oncol 21:392–397 Pinto AE, Andre S, Nogueira M, Mendonca E, Soares J, 1997, Flow cytometric DNA hypertetraploidy is associated with unfavorable prognostic features in breast cancer. J Clin Pathol 50:591– 595 Beerman H, Kluin PM, Hermans J, van de Velde CJ, Cornelisse CJ, 1990, Prognostic significance of DNA-ploidy in a series of 690 primary breast cancer patients. Int J Cancer 45:34–39 Dutrillaux B, Gerbault-Seureau M, Remvikos Y, Zafrani B, Prieur M, 1991, Breast cancer genetic evolution: I. Data from cytogenetics and DNA content. Breast Cancer Res Treat 19:245–255 Molist R, Remvikos Y, Dutrillaux B, Muleris M, 2004, Characterization of a new cytogenetic subtype of ductal breast carcinoma. Oncogene 23:5986–5993 Remvikos Y, Vogt N, Muleris M, Salmon RJ, Malfoy B, Dutrillaux B, 1995, DNA-repeat instability is associated with colorectal cancers presenting minimal chromosome rearrangements. Genes Chromosomes Cancer 12:272–276 Ferno M, Baldetorp B, Borg A, Olsson H, Sigurdsson H, Killander D, 1992, Flow cytometric DNA index and S-phase fraction in breast cancer in relation to other prognostic variables and to clinical outcome. Acta Oncol 31:157–165 Toikkanen S, Joensuu H, Klemi P, 1989, The prognostic significance of nuclear DNA content in invasive breast cancer–a study with long-term follow-up. Br J Cancer 60:693–700 Elie C, Molist R, Asselain B, Dutrillaux B, Muleris M, 2005, Ductal breast carcinomas with whole chromosome gains as a particular subset of near-diploid tumors with different metastases free survival. Breast Cancer Res Treat 92:279–285 Wenger CR, Clark GM, 1998, S-phase fraction and breast cancer– a decade of experience. Breast Cancer Res Treat 51:255–265 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 71 EP 77 DI 10.1007/s12253-012-9559-z PG 7 ER PT J AU Zhang, Y Tan, YF Jiang, Ch Zhang, K Zha, TZ Zhang, M AF Zhang, Yun Tan, Yong-Fei Jiang, Chao Zhang, Kai Zha, Tian-Zhou Zhang, Miao TI High ADAM8 Expression is Associated with Poor Prognosis in Patients with Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ADAM8; Hepatocellular carcinoma; Biomarker; Prognosis; Immunohistochemical analysis ID ADAM8; Hepatocellular carcinoma; Biomarker; Prognosis; Immunohistochemical analysis AB In this study,we investigated the ADAM8 expression in hepatocellular carcinoma (HCC) and its correlation with clinicopathologic features,including the survival of patients with HCC. Furthermore,we examined the biological processes regulated by ADAM8 during the development of using HepG2 cell line as a model system. We used immunohistochemistry to compare ADAM8 protein expression in HCC and normal liver tissues and further analyze the ADAM8 protein expression in clinicopathologically characterized 105 HCC cases.We stably knocked down the endogenous expression level of ADAM8 in HepG2 cells with specific shRNA expressing lentiviral vector. Following the successful establishment of stable cells,we examined in vitro cell growth by MTT assay,anchorage-independent growth by soft-agar colony formation assay and cell migration/invasion by transwell and boyden chamber assay. And in addition,we also investigated the in vivo tumor growth by xenograft transplantation of HepG2 cells into nude mice. Protein expression level of ADAM8 was markedly higher in HCC tissues than that in the normal liver tissues (P<0.0058).In addition,high expression of ADAM8 protein was positively correlated with serum AFP elevation,tumor size,histological differentiation,tumor recurrence,tumor metastasis,and tumor stage. Patients with higher ADAM8 expression showed a significantly shorter overall survival time than patients with low ADAM8 expression. Multivariate analysis suggested that ADAM8 expression might be an independent prognostic indicator (p00.016) for the survival of patients with HCC. ADAM8-specific shRNA (shADAM8) successfully knocked down its endogenous expression in HepG2 cells. Compared to the parental and control shRNA-transfected (shCtrl) HepG2 cells,the shADAM8 cells exhibited significantly reduced in vitro cell growth,anchorageindependent growth,cell migration and invasion (p<0.05).In vivo,the xenograft transplants from shADAM8 cells gave rise to much smaller tumors as compared to those from shCtrl cells. High ADAM8 expression is associated with poor overall survival in patients with HCC. Down-regulation of ADAM8 inhibits the growth,anchorage-independent growth, migration and invasion of HepG2 cells. ADAM8 may be a potential target of antiangiogenic therapy for HCC. C1 [Zhang, Yun] Yixing People’s Hospital, Department of General Surgery, No. 75, Tongzhen Guan Rd, 214200 Yixing, China. [Tan, Yong-Fei] Yixing People’s Hospital, Department of General Surgery, No. 75, Tongzhen Guan Rd, 214200 Yixing, China. [Jiang, Chao] Yixing People’s Hospital, Department of General Surgery, No. 75, Tongzhen Guan Rd, 214200 Yixing, China. [Zhang, Kai] Yixing People’s Hospital, Department of General Surgery, No. 75, Tongzhen Guan Rd, 214200 Yixing, China. [Zha, Tian-Zhou] Yixing People’s Hospital, Department of General Surgery, No. 75, Tongzhen Guan Rd, 214200 Yixing, China. [Zhang, Miao] Yixing People’s Hospital, Department of General Surgery, No. 75, Tongzhen Guan Rd, 214200 Yixing, China. RP Tan, YF (reprint author), Yixing People’s Hospital, Department of General Surgery, 214200 Yixing, China. EM yongfeitan@yahoo.com.cn CR Shariff MI, Cox IJ, Gomaa AI, Khan SA, Gedroyc W, TaylorRobinson SD, 2009, Hepatocellular carcinoma:current trends in worldwide epidemiology, risk factors, diagnosis and therapeutics. Expert Rev Gastroenterol Hepatol 3:353–367 Lau WY, Lai EC, 2008, Hepatocellular carcinoma:current management and recent advances. Hepatobiliary Pancreat Dis Int 7:237–257 Koorey D, 2007, Hepatocellular carcinoma:prevention, detection and treatment in the real world. Intern Med J 37:513–515 Pleguezuelo M, Marelli L, Misseri M, Germani G, Calvaruso V, Xiruochakis E et al, 2008, TACE versus TAE as therapy for hepatocellular carcinoma. Expert Rev Anticancer Ther 8:1623–1641 Yoshiyama K, Higuchi Y, Kataoka M, Matsuura K, Yamamoto S, 1997, CD156(human ADAM8):expression, primary amino acid sequence, and gene location. Genomics 41(1):56–62 Yamamoto S, Higuchi Y, Yoshiyama K et al, 1999, ADAM family proteins in the immune system. Immunol Today 20(6):278–284 Schlomann U, Wildeboer D, Webster A et al, 2002, The metalloprotease disintegrin ADAM8. Processing by autocatalysis is required for proteolytic activity and cell adhesion. Biol Chem 277(50):48210–48219 Primakoff P, Myles DG, 2000, The ADAM gene family:surface proteins with adhesion and protease activity. Trends Genet 16, 2):83–87 Seals DF, Courtneidge SA, 2003, The ADAMs family of metalloproteases: multidomain proteins with multiple functions. Genes Dev 17(1):7–30 Fourie AM, Coles F, Moreno V, Karlsson L, 2003, Catalytic activity of ADAM8, ADAM15, and MDC-L, ADAM28, on synthetic peptide substrates and in ectodomain cleavage of CD23. J Biol Chem 278(33):30469–30477 Naus S, Richter M, Wildeboer D, Moss M, Schachner M, Bartsch JW, 2004, Ectodomain shedding of the neural recognition molecule CHL1 by the metalloprotease-disintegrin ADAM8 promotes neurite outgrowth and suppresses neuronal cell death. J Biol Chem 279(16):16083–16090 Karan D, Lin FC, Bryan M et al, 2003, Expression of ADAMs, a disintegrin and metalloproteases, and TIMP-3, tissue inhibitor of metalloproteinase-3, in human prostatic adenocarcinomas. Int J Oncol 23(5):1365–1371 O’Shea C, McKie N, Buggy Yet al, 2003, Expression of ADAM- 9 mRNA and protein in human breast cancer. Int J Cancer 105, 6):754–761 Zhu XD, Zhang JB, Zhuang PY, Zhu HG, Zhang W, Xiong YQ et al, 2008, High expression of macrophage colony-stimulating factor in peritumoral liver tissue is associated with poor survival after curative resection of hepatocellular carcinoma. J Clin Oncol 26:2707–2716 Valkovskaya N, Kayed H, Felix K, Hartmann D, Giese NA, Osinsky SP, Friess H, Kleeff J, 2007, ADAM8 expression is associated with increased invasiveness and reduced patient survival in pancreatic cancer. J Cell Mol Med 11(5):1162–1174 Valkovskaya NV, 2008, Hypoxia-dependent expression of ADAM8 in human pancreatic cancer cell lines. Exp Oncol 30(2):129–132 Kelly K, Crowley J, Bunn PA Jr et al, 2001, Randomized phase III trial of paclitaxel plus carboplatin versus vinorelbine plus cisplatin in the treatment of patients with advanced non-small-cell lung cancer:a Southwest Oncology Group trial. J Clin Oncol 19(13):3210–3218 Hennessy BT, Hanrahan EO, Daly PA, 2004, Non-Hodgkin lymphoma: an update. Lancet Oncol 5(6):341–353 Pujol JL, Grenier J, Daures JP, Daver A, Pujol H, Michel FB, 1993, Serum fragment of cytokeratin subunit 19 measured by CYFRA 21–1 immunoradiometric assay as a marker of lung cancer. Cancer Res 53(1):61–66 Miyake Y, Kodama T, Yamaguchi K, 1994, Pro-gastrin-releasing peptide(31–98, is a specific tumor marker in patients with small cell lung carcinoma. Cancer Res 54(8):2136–2140 Michael M, Babic B, Khokha R et al, 1999, Expression and prognostic significance of metalloproteinases and their tissue inhibitors in patients with small-cell lung cancer. J Clin Oncol 17, 6):1802–1808 Peng SY, Lai PL, Chu JS et al, 1993, Expression and hypomethylation of alpha-fetoprotein gene in unicentric and multicentric human hepatocellular carcinomas. Hepatology 17:35–41 Hsu HC, Jeng YM, Mao TL, Chu JS, Lai PL, Peng SY, 2000, b- Catenin mutations are associated with a subset of low stage hepatocellular carcinoma negative for hepatitis B virus and with favorable prognosis. Am J Pathol 157:763–770 Tuck AB, O'Malley FP, Singhal H et al, 1998, Osteopontin expression in a group of lymph node negative breast cancer patients. Int J Cancer 79:502–508 Ue T, Yokozaki H, Kitadai Y et al, 1998, Co-expression of osteopontin and CD44v9 in gastric cancer. Int J Cancer 79:127–132 Hwang YH, Choi JY, Kim S et al, 2004, Over-expression of c-raf- 1proto-oncogene in liver cirrhosis and hepatocellular carcinoma. Hepatol Res 29(2):113–121 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 79 EP 88 DI 10.1007/s12253-012-9560-6 PG 10 ER PT J AU Masood, N Kayani, AM AF Masood, Nosheen Kayani, Akhtar Mahmood TI Expression Patterns of Carcinogen Detoxifying Genes (CYP1A1, GSTP1 & GSTT1) in HNC Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Immunohistochemistry; GSTP1; CYP1A1; GSTT1 ID Immunohistochemistry; GSTP1; CYP1A1; GSTT1 AB Carcinogen detoxifying genes may be involved in pathogenesis of head and neck cancer (HNC). CYP1A1 is phase I enzyme that converts carcinogens into water soluble compounds which are easily excreted from body. GSTs constitute phase II detoxification enzymes that recognize these highly electrophilic compounds and detoxify them. Abnormal expression of these genes can potentially lead to cancer initiation. In present study, we analyzed protein expression of these genes in a total of 192 HNC patients and noncancerous healthy control serum samples screened for GSTs specific activity by ELISA. Furthermore, expression of these molecules was also determined in 49 HNC tissues/ adjacent control tissue by immunohistochemistry with specific antibodies. Mean serum GSTs specific activity was found to be 7.7 (+11.5)U/L in HNC patients and 11.4 (+7.5)U/L in controls. Significant decrease (P<0.05) in GSTs specific activity was observed in HNC patients compared with controls (P<0.001). Data for immunohistochemistry showed that CYP1A1 and GSTT1 was down expressed whereas GSTP1 was over expressed in HNC tissues compared with adjacent normal control tissues. Results of immunohistochemistry revealed 63 % HNC tissues had weak, 27 % moderate and 10 % strong staining for CYP1A1. For GSTT1, 27 % HNC tissues had no staining, 49 % weak staining, 16 % moderate and 8 % strong staining. Similarly for GSTP1, percentages for weak, moderate and strong staining were 6 %, 12 % and 82 % respectively. These reduced proteins observed in cancer patients highlight a potential breach on DNA repair mechanism when compared with control. Thus altered expression of these detoxifying molecules may collectively contribute to HNC development in Pakistani population. C1 [Masood, Nosheen] COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics LabIslamabad, Pakistan. [Kayani, Akhtar Mahmood] COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics LabIslamabad, Pakistan. RP Masood, N (reprint author), COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics Lab, Islamabad, Pakistan. EM nosheenmasood@hotmail.com CR Bhurgri Y, 2005, Cancer of the oral cavity- trends in Karachi south, 1995–2002). Asian Pac J Cancer Prev 6(1):22–26 Toru H, Masaharu Y, Shinji T, Kazuaki C, 2008, Genetic polymorphisms and head and neck cancer risk. Int J Oncol 32(5):945–973 Hanif M, Zaidi P, Kamal S, Hameed A, 2009, Institution based cancer incidence in a local population in Pakistan: nine year data analysis. Asian Pac J Cancer Prev 10(2):227–230 Guengerich FP, Simada T, 1991, Oxidation of toxic and carcinogenic chemicals by human cytochrome P 450 enzymes. Chem Res Toxicol 4:341–407 Masood N, Kayani MA, 2011, Mutational analysis of xenobiotic metabolizing genes, CYP1A1 and GSTP1, in sporadic head and neck cancer patients. Genet Mol Biol 34(4):533–538 Masood N, Kayani MA, Malik FA, Ishrat M, Baig RM, Faryal R, 2011, Genetic variations in carcinogen metabolizing genes associated with oral cancer in Pakistani population. Asian Pac J Cancer Prev 12:491–495 Masood N, Malik FA, Ishrat M, Baig RM, Kayani MA, 2011, A novel CYP1A1 gene polymorphism and the risk of head and neck cancer in Pakistani population. Afr J Biotechnol 10(27):5273–5280 Masood N, Ishrat M, Malik FA, Baig RM, Kayani MA, 2010, Association of GSTM1 and GSTT1 gene deletions with risk of head and neck cancer in Pakistan: a case control study. Asian Pac J Cancer Prev 11:881–885 Masood N, Malik FA, Kayani MA, 2011, Expression of xenobiotic metabolizing genes in head and neck cancer tissues. Asian Pac J Cancer Prev 12(2):377–382 Farin FM, Bigler LG, Oda D, McDougall JK, Omiecinski CJ, 1995, Expression of cytochrome P450 and microsomal exposide hydrolase in cervical and oral epithelial cells immortalized by human papillomavirus type 16 E6/E7 genes. Carcinogenesis 16:1670 Tu CP, Weiss MJ, Li NQ, Reddy CC, 1983, Tissue-specific expression of the rat glutathione S-transferases. J Biol Chem 8:4659–4662 Hayes JD, Pulford DJ, 1995, The glutathione S-transferase supergene family: regulation of GST and the contribution of the isoenzymes to cancer chemoprotection and drug resistance. Crit Rev Biochem Mol Biol 30(6):445–600 Tsuchida S, 1990, Purification and characterization of glutathione transferases with an activity toward nitroglycerin from human aorta and heart: multiplicity of the human class mu forms. J Biol Chem 13:7150–7157 Matthias C, Jahnke V, Hand P et al, 1999, Immunhistologische und molekular genetische untersuchungen des effektes der glutathion-S-transferase auf die entstehung von plattenepithelkarzinomen der Kopf-Hals-region, [immunohistologic and molecular genetic studies of the effect of glutathione-S-transferases on the development of squamous epithelial carcinomas in the area of the head-neck]). Laryngorhinootologie 78:182–188 Matthias C, Bockmuhl U, Jahnke Vet al, 1998, The glutathione Stransferase GSTP1 polymorphism: effects on susceptibility to oral/ pharyngeal and laryngeal carcinomas. Pharmacogenetics 8(1):1–6 Niitsu Y, Takahashi Y, Saito T, Hirata Y, Arisato N, Maruyama H, Kohgo Y, Listowsky I, 1989, Serum glutathione-S-transferase-pi as a tumor marker for gastrointestinal malignancies. Cancer 63, 2):317–323 Seidegard J, Pero RW, Miller DG et al, 1986, A glutathione transferase in human leukocytes as a marker for the susceptibility to lung cancer. Carcinogenesis 7(5):751–753 Lafuente A, Pujol F, Carretero P et al, 1993, Human glutathione S-transferase mu, GST mu, deficiency as a marker for the susceptibility to bladder and larynx cancer among smokers. Cancer Lett 68(1):49–54 Lafuente A, Molina R, Palou J et al, 1995, Phenotype of glutathione S-transferase mu, GSTM1, and susceptibility to malignant melanoma: MMM, multidisciplinary malignant melanoma, group. Br J Cancer 72(2):324–326 Patel BP, Raval GN, Rawal RM, Patel JB, Sainger RN, Patel MM, Shah MH, Patel DD, Patel PS, 2002, Serum glutathione-Stransferase and glutathione reductase activity in head and neck cancer patients. Neoplasma 49(4):260–266 Ferruzzi E, Franceschini R, Cazzolato G, Geroni C, Fowst C, Pastorino U, Tradati N, Tursi J, Dittadi R, Gion M, 2003, Blood glutathione as a surrogate marker of cancer tissue glutathione Stransferase activity in non-small cell lung cancer and squamous cell carcinoma of the head and neck. Eur J Cancer 39(7):1019– 1029 Van Der Zee AGJ, Van Ommen B, Meijer C, Hollema H, Van Bladeren PJ, 1992, Glutathione S-transferase activity and isoenzyme composition in benign ovarian tumors, untreated malignant ovarian tumors, and malignant ovarian tumors after platinum/ Cyclophosphamide chemotherapy. Br J Cancer 66:930–936 Sprem M, Babic D, Abramic M et al, 2001, Glutathione and glutathione S-transferase as early markers for ovarian carcinomas: case series. Croat Med J 42(6):624–629 Smart J, Daly AK, 2000, Variation in induced CYP1A1 levels: relationship to CYP1A1, Ah receptor and GSTM1 polymorphisms. Pharmacogenetics 10:11–24 Anttila S, Tuominen P, Hirvonen A et al, 2001, CYP1A1 levels in lung tissue of tobacco smokers and polymorphisms of CYP1A1 and aromatic hydrocarbon receptor. Pharmacogenetics 11:501–509 Wandel C, Witte JS, Hall JM, Stein CM, Wood AJ, Wilkinson GR, 2000, CYP3A activity in African American and European American men: population differences and functional effect of the CYP3A4*1B5′- promoter region polymorphism. Clin Pharmacol Ther 68:82–91 El-Rayes BF, Ali S, Heilbrun LK et al, 2003, Cytochrome p450 and glutathione transferase expression in human breast cancer. Clin Cancer Res 9:1705–1709 Murray GI, Shaw D, Weaver RJ, McKay JA, Ewen SW, Melvin WT, Burke MD, 1994, Cytochrome P450 expression in oesophageal cancer. Gut 35:599–603 Murray GI, Taylor VE, McKay JA, Weaver RJ, Ewen SW, Melvin WT, Burke MD, 1995, Expression of xenobiotic metabolizing enzymes in tumors of the urinary bladder. Int J Exp Pathol 76:271–276 Theo PJ, Johannes JM, Hennie MJ, Wilbert HM, Anne W, 1995, Glutathione S transferases and glutathione in human head and neck cancer. Carcinogenesis 16(3):619–624 David GB, Isabelle M, Jonathan H, 2007, Glutathione S transferase: differential expression of alpha, mu and pi isoenzymes in benign prostate, prostatic intraepithelial neoplasia, and prostatic adenocarcinoma. Hum Pathol 38:1394–1401 Ioanna AD, Demetrios AA, Demetrios AS, 2001, Genetic polymorphisms and transcriptional pattern analysis of CYP1A1, AhR, GSTM1, GSTP1 and GSTT1 genes in breast cancer. Int J Mol Med 8:79–87 Tanita J, Tsuchida S, Hozawa J, Sato K, 1993, Expression of glutathione S-transferase-pi in human squamous cell carcinomas of the pharynx and larynx. Loss after radiation therapy. Cancer 72:569–576 Chen YK, Lin LM, 1995, Immunohistochemical demonstration of epithelial glutathione S-transferase isoenzymes in normal, benign, premalignant and malignant human oral mucosa. J Oral Pathol Med 24:316–321 Bentz BG, Haines GK, Radosevich JA, 2000, Glutathione Stransferase pi in squamous cell carcinoma of the head and neck. Laryngoscope 110:1642–1647 Mulder TPJ, Manni JJ, Roelofs HMJ, Peters WHM, Wiersma A, 1995, Glutathione S-transferases and glutathione in human head and neck cancer. Carcinogenesis 16:619–624 Anna Y, Russell V, Malti K, Dan L, Morgan AM, Ie M, Robert JK, 2011, Immunohistochemical staining patterns of p53 can serve as a surrogate marker for TP53 mutations in ovarian carcinoma: an immunohistochemical and nucleotide sequencing analysis. Mod Pathol 24:1248–1253 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 89 EP 94 DI 10.1007/s12253-012-9563-3 PG 6 ER PT J AU Cserni, G Bori, R Maraz, R Leidenius, HKM Meretoja, JT Heikkila, SP Regitnig, P Luschin-Ebengreuth, G Zgajnar, J Perhavec, A Gazic, B Lazar, Gy Takacs, T Voros, A Audisio, AR AF Cserni, Gabor Bori, Rita Maraz, Robert Leidenius, H K Marjut Meretoja, J Tuomo Heikkila, S Paivi Regitnig, Peter Luschin-Ebengreuth, Gero Zgajnar, Janez Perhavec, Andraz Gazic, Barbara Lazar, Gyorgy Takacs, Tibor Voros, Andras Audisio, A Riccardo TI Multi-Institutional Comparison of Non-sentinel Lymph Node Predictive Tools in Breast Cancer Patients with High Predicted Risk of Further Axillary Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sentinel lymph node; Non-sentinel lymph node; Breast cancer; Nomogram; High risk; Axillary lymph node dissection ID Sentinel lymph node; Non-sentinel lymph node; Breast cancer; Nomogram; High risk; Axillary lymph node dissection AB Although axillary lymph node dissection (ALND) has been the standard intervention in breast cancer patients with sentinel lymph node (SLN) metastasis, only a small proportion of patients benefit from this operation, because most do not harbor additional metastases in the axilla. Several predictive tools have been constructed to identify patients with low risk of non-SLN metastasis who could be candidates for the omission of ALND. In the present work, predictive nomograms were used to predict a high (>50 %) risk of non-SLN metastasis in order to identify patients who would most probably benefit from further axillary treatment. Data of 1000 breast cancer patients with SLN metastasis and completion ALND from 5 institutions were tested in 4 nomograms. A subset of 313 patients with micrometastatic SLNs were also tested in 3 different nomograms devised for the micrometastatic population (the high risk cut-off being 20 %). Patients with a high predicted risk of non-SLN metastasis had higher rates of metastasis in the non-SLNs than patients with low predicted risk. The positive predictive values of the nomograms ranged from 44 % to 64 % with relevant inter-institutional variability. The nomograms for micrometastatic SLNs performed much better in identifying patients with low risk of non-SLN involvement than in high-risk-patients; for the latter, the positive predictive values ranged from 13 % to 20 %. The nomograms show inter-institutional differences in their predictive values and behave differently in different settings. They are worse in identifying high risk patients than low-risk ones, creating a need for new predictive models to identify high-risk patients. C1 [Cserni, Gabor] University of Szeged, Department of Pathology, Allomas u. 2, 6720 Szeged, Hungary. [Bori, Rita] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, 6000 Kecskemet, Hungary. [Maraz, Robert] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38, 6000 Kecskemet, Hungary. [Leidenius, H K Marjut] Helsinki University Central Hospital, Breast Surgery Unit, 00029 Helsinki, Finland. [Meretoja, J Tuomo] Helsinki University Central Hospital, Breast Surgery Unit, 00029 Helsinki, Finland. [Heikkila, S Paivi] Helsinki University Central Hospital, Department of Pathology, 00029 Helsinki, Finland. [Regitnig, Peter] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, 8036 Graz, Austria. [Luschin-Ebengreuth, Gero] Medical University of Graz, Department of Obstetrics and Gynecology, Auenbruggerplatz 25, 8036 Graz, Austria. [Zgajnar, Janez] Institute of Oncology, Department of Surgical Oncology, Zaloska c. 2, 1105 Ljubljana, Slovenia. [Perhavec, Andraz] Institute of Oncology, Department of Surgical Oncology, Zaloska c. 2, 1105 Ljubljana, Slovenia. [Gazic, Barbara] Institute of Oncology, Department of Pathology, Zaloska c. 2, 1105 Ljubljana, Slovenia. [Lazar, Gyorgy] University of Szeged, Department of Surgery, Pecsi u 6, 6720 Szeged, Hungary. [Takacs, Tibor] University of Szeged, Department of Surgery, Pecsi u 6, 6720 Szeged, Hungary. [Voros, Andras] University of Szeged, Department of Pathology, Allomas u. 2, 6720 Szeged, Hungary. [Audisio, A Riccardo] St Helens Teaching Hospital, Department of Surgery, Marshalls Cross Road, WA93DA St Helens, UK. RP Cserni, G (reprint author), University of Szeged, Department of Pathology, 6720 Szeged, Hungary. EM cserni@freemail.hu CR Lyman GH, Giuliano AE, Somerfield MR et al, 2005, American Society of Clinical Oncology guideline recommendations for sentinel lymph node biopsy in early-stage breast cancer. J Clin Oncol 23:7703–7720 Van Zee KJ, Manasseh DM, Bevilacqua JL et al, 2003, A nomogram for predicting the likelihood of additional nodal metastases in breast cancer patients with a positive sentinel node biopsy. Ann Surg Oncol 10:1140–1151 Hwang RF, Krishnamurthy S, Hunt KK et al, 2003, Clinicopathologic factors predicting involvement of nonsentinel axillary nodes in women with breast cancer. Ann Surg Oncol 10:248–254 Farshid G, Pradhan M, Kollias J, Gill PG, 2004, A decision aid for predicting non-sentinel node involvement in women with breast cancer and at least one positive sentinel node. Breast 13:494–501 Barranger E, Coutant C, Flahault A, Antoine M, Darai E, Uzan S, 2005, An axilla scoring system to predict non-sentinel lymph node status in breast cancer patients with sentinel lymph node involvement. Breast Cancer Res Treat 91:113–119 Degnim AC, Reynolds C, Pantvaidya G et al, 2005, Nonsentinel node metastasis in breast cancer patients: assessment of an existing and a new predictive nomogram. Am J Surg 190:543–550 Chagpar AB, Scoggins CR, Martin RC 2nd et al, 2006, Prediction of sentinel lymph node-only disease in women with invasive breast cancer. Am J Surg 192:882–887 Kohrt HE, Olshen RA, Bermas HR et al, 2008, New models and online calculator for predicting non-sentinel lymph node status in sentinel lymph node positive breast cancer patients. BMC Cancer 8:66 Cho J, Han W, Lee JW et al, 2008, A scoring system to predict nonsentinel lymph node status in breast cancer patients with metastatic sentinel lymph nodes: a comparison with other scoring systems. Ann Surg Oncol 15:2278–2286 Pal A, Provenzano E, Duffy SW, Pinder SE, Purushotham AD, 2008, A model for predicting non-sentinel lymph node metastatic disease when the sentinel lymph node is positive. Br J Surg 95:302–309 Houvenaeghel G, Nos C, Giard S et al, 2009, A nomogram predictive of non-sentinel lymph node involvement in breast cancer patients with a sentinel lymph node micrometastasis. Eur J Surg Oncol 35:690–695 Coufal O, Pavlik T, Fabian P et al, 2009, Predicting non-sentinel lymph node status after positive sentinel biopsy in breast cancer: What model performs the best in a Czech population? Pathol Oncol Res 15:733–740 Perhavec A, Perme MP, Hocevar M, Besic N, Zgajnar J, 2010, Ljubljana nomograms for predicting the likelihood of non-sentinel lymph node metastases in breast cancer patients with a positive sentinel lymph node. Breast Cancer Res Treat 119:357–366 Meretoja TJ, Strien L, Heikkila PS, Leidenius MH, 2012, A simple nomogram to evaluate the risk of nonsentinel node metastases in breast cancer patients with minimal sentinel node involvement. Ann Surg Oncol 19:567–576 Cserni G, 2007, Comparison of different validation studies on the use of the Memorial-Sloan Kettering Cancer Center nomogram predicting nonsentinel node involvement in sentinel node positive breast cancer patients. Am J Surg 194:699–700 Bilimoria KY, Bentrem DJ, Hansen NM et al, 2009, Comparison of sentinel lymph node biopsy alone and completion axillary lymph node dissection for node-positive breast cancer. J Clin Oncol 27:2946–2953 Yi M, Giordano SH, Meric-Bernstam F et al, 2010, Trends in and outcomes from sentinel lymph node biopsy, SLNB, alone vs. SLNB with axillary lymph node dissection for node-positive breast cancer patients: experience from the SEER database. Ann Surg Oncol 17(Suppl 3):343–351 Giuliano AE, McCall L, Beitsch P et al, 2010, Locoregional recurrence after sentinel lymph node dissection with or without axillary dissection in patients with sentinel lymph node metastasis. Ann Surg 252:426–433 Cserni G, Boross G, Maraz R et al, 2012, Multicentre validation of different predictive tools of non-sentinel lymph node involvement in breast cancer. Surg Oncol., DOI 10.1016/j.suronc.2011.12.001 Houvenaeghel G, Bannier M, Nos C et al, 2012, Non sentinel node involvement prediction for sentinel node micrometastases in breast cancer: nomogram validation and comparison with other models. Breast 21:204–249 Cserni G, Gregori D, Merletti F et al, 2004, Non-sentinel node metastases associated with micrometastatic sentinel nodes in breast cancer: metaanalysis of 25 studies. Br J Surg 91:1245–1252 Cserni G, Bori R, Sejben I et al, 2009, Analysis of predictive tools for further axillary involvement in patients with sentinel-lymphnode- positive, small, < or 015 mm, invasive breast cancer. Orv Hetil 150:2182–2188 Goldhirsch A, Wood WC, Coates AS et al, 2011, Strategies for subtypes—dealing with the diversity of breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol 22:1736– 1747 Tvedskov TF, Jensen MB, Lisse IM, Ejlertsen B, Balslev E, Kroman N, 2012, High risk of non-sentinel node metastases in a group of breast cancer patients with micrometastases in the sentinel node. Int J Cancer., DOI 10.1002/ijc.27499 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 95 EP 101 DI 10.1007/s12253-012-9553-5 PG 7 ER PT J AU Pagni, F Zannella, S Ronchi, S Garanzini, C Leone, EB AF Pagni, Fabio Zannella, Stefano Ronchi, Susanna Garanzini, Cristina Leone, Eugenio Biagio TI HER2 Status of Gastric Carcinoma and Corresponding Lymph Node Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Metastasis; HER2; IHC; HER2 status scoring system ID Gastric cancer; Metastasis; HER2; IHC; HER2 status scoring system AB Our goal is to verify HER2 status variability between primary tumor and metastatic site. Our second intention is to identify the most reliable criteria for pathological HER2 status assessment in gastric cancer node metastases since, at present, there is not a validated standard. 3 independent pathologists evaluated HER2 immunohistochemical and gene status (for IHC 2+ cases) in 34 gastric carcinoma metastatic lymph nodes and in their corresponding primary tumors. For primary gastric cancers, we followed the current HER2 assessment guidelines and for nodal metastases, we applied two immunohistochemical scoring systems with different cut-offs. The immunohistochemical inter-pathologists mean agreement was 71.4 % (κ00.45); a final score for each case was defined after collegial revision. By applying the two immunohistochemical criteria, we found 2 discordant cases, which can imply different pathological management.Moreover, a significantly different HER2 status between lymph node metastasis and primary tumor was obtained in 4 cases (concordance ratio 87.5 %). None of the patients would have undergone a different therapeutic pathway despite the scoring method applied. On the other hand we also detected a subset of patients who could have their therapeutic management changed, according to the differences between HER2 status in lymph nodes metastases and primary tumor. C1 [Pagni, Fabio] Desio Hospital, Department of PathologyDesio, Italy. [Zannella, Stefano] Desio Hospital, Department of PathologyDesio, Italy. [Ronchi, Susanna] Desio Hospital, Department of PathologyDesio, Italy. [Garanzini, Cristina] Desio Hospital, Department of PathologyDesio, Italy. [Leone, Eugenio Biagio] Desio Hospital, Department of PathologyDesio, Italy. RP Pagni, F (reprint author), Desio Hospital, Department of Pathology, Desio, Italy. EM petala.83@tiscali.it CR Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T, Aprile G, Kulikov E, Hill J, Lehle M, Ruschoff J, Kang YK, 2010, Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer, ToGA): a phase III, openlabel, randomised controlled trial. Lancet 376:687–97 Hoffmann M, Stoss O, Shi D, Buttner R, van de Vijver M, Kim W, Ochiai A, Ruschoff J, Henkel T, 2008, Assessment of a HER2 scoring system for gastric cancer: results from a validation study. Histopathology 52:797–805 Ruschoff J, Dietel M, Baretton G, Arbogast S, Walch A, Monges G, Chenard MP, Penault-Llorca F, Nagelmeier I, Schlake W, Hofler H, Kreipe HH, 2010, HER2 diagnostics in gastric cancer: guideline validation and development of standardized immunohistochemical testing. Virchows Arch 457:299–307 Wolff AC, Hammond EH M, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, McShane LM, Paik S, Pegram MD, Perez EA, Press MF, Rhodes A, Sturgeon C, Taube SE, Tubbs R, Vance GH, van de Vijver M, Wheeler TM, Hayes DF, 2007, American Society of Clinical Oncology/College of American Pathologists Guideline recommendations for Human Epidermal Growth Factor Receptor 2 testing in breast cancer. Arch Pathol Lab Med 131:18–43 Albarello L, Pecciarini L, Doglioni C, 2011, HER2 Testing in gastric cancer. Adv Anat Pathol 18:53–59 Bozzetti C, Negri FV, Lagrasta CA, Crafa P, Bassano C, Tamagnini I, Gardini G, Nizzoli R, Leonardi F, Gasparro D, Camisa R, Cavalli S, Silini EM, Ardizzoni A, 2011, Comparison of HER2 status in primary and paired metastatic sites of gastric carcinoma. Br J Cancer 104:1372–1376 Nassar A, Cohen C, Agersborg SS, Zhou W, Lynch KA, Albitar M, Barker EA, Vanderbilt BL, Thompson J, Heyman ER, Lange H, Olson A, Siddiqui MT, 2001, Trainable Immunohistochemical HER/neu Image Analysis. A multisite performance study using 260 breast tissue specimen. Arch Pathol Lab Med 135:896–902 Marx AH, Tharun L, Muth J, Dancau AM, Simon R, Yekebas E, Kaifi JT, Mirlacher M, Brummendorf TH, Bokemeyer C, Izbicki JR, Sauter G, 2009, HER-2 amplification is highly homogeneous in gastric cancer. Hum Pathol 40:769–777 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 103 EP 109 DI 10.1007/s12253-012-9564-2 PG 7 ER PT J AU Tavares, A Gandra, A Viveiros, F Cidade, C Maciel, J AF Tavares, Amelia Gandra, Antonio Viveiros, Fernando Cidade, Cassilda Maciel, Jorge TI Analysis of Clinicopathologic Characteristics and Prognosis of Gastric Cancer in Young and Older Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE Cancer gastric; Prognostic; Age ID Cancer gastric; Prognostic; Age AB Background: The worldwide incidence of gastric cancer is gradually declining, however it remains the fourth highest in cancer incidence and the second leading cause of cancer death. Gastric cancer in young people is a disturbing problem and the routine screening does not include people less than 35 years. The clinicopathological features of gastric carcinoma are said to differ between young and elderly patients and it is thought that the prognosis of this disease is worse for younger patients. It is also suggested that the diagnosis is usually made later or have a more aggressive behaviour. Although, others report that tumor staging and prognosis for young patients is similar to older patients and depends on whether the patients undergo a curative resection. All these data need more investigation and studies. Although Portugal has a high incidence of gastric cancer, no studies have yet been performed comparing the clinicopathologic features and prognosis of young and elderly patients with gastric cancer. Aims: This study intend to assess whether the clinicopathological features and prognosis of gastric cancer in young patients (YGC) is similar to older ones (OGC). Methods: Between 2000 and 2005, 406 patients with histological diagnosis of primary gastric cancer, treated in the Departments of Surgery and Oncology at the Centro Hospitalar of Vila Nova de Gaia / Espinho, were regularly followed at least for five years after surgery. These were reviewed retrospectively. Several variables were analyzed in young patients and compared with the elder ones. We used the chi-square and Fisher to evaluate the statistical association between categorical variables and t-test for numeric variables. Survival was estimated by the Kaplan- Meier method and used the log-rank test to assess differences in survival among different subgroups of patients. The criteria for statistical significance was p<0.05. Data analysis was performed using the SPSS 18. Results and Conclusions: With regard to resectability, 78 % of the tumors were resected in the group of younger patients, the surgery more frequently achieved was total gastrectomy with anastomosis in Y of Roux. In the elder group, about 62 % of the tumors were resected and BII gastrectomy was the most frequent surgery. The diffuse adenocarcinoma was the most frequent histological type in younger patients, whereas in older patients was intestinal adenocarcinoma. With regard to the stage in the first group there was a predominance of stages: IA and IV (26.1 %) in the second: IV (25.8 %). The survival for stage III e IV was significantly worst in YGC compared with OGC. C1 [Tavares, Amelia] Centro Hospitalar de Vila Nova de Gaia/EspinhoVila Nova de Gaia, Portugal. [Gandra, Antonio] Centro Hospitalar de Vila Nova de Gaia/EspinhoVila Nova de Gaia, Portugal. [Viveiros, Fernando] Centro Hospitalar de Vila Nova de Gaia/EspinhoVila Nova de Gaia, Portugal. [Cidade, Cassilda] Centro Hospitalar de Vila Nova de Gaia/EspinhoVila Nova de Gaia, Portugal. [Maciel, Jorge] Centro Hospitalar de Vila Nova de Gaia/EspinhoVila Nova de Gaia, Portugal. RP Tavares, A (reprint author), Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal. EM ameliassbtavares@gmail.com CR Parkin DM, 2004, International variation. Oncogene 23:6329– 6340 Parkin DM, Bray FI, Devesa SS, 2001, Cancer burden in the year 2000, the global picture. Eur J Cancer 37:S4–S66 Forman D, Newell DG, Fullerton F, Yarnell JW, Stacey AR, Wald N, Sitas F, 1991, Association between infection with Helicobacter Pylori and risk of gastric cancer: evidence from a prospective investigation. BMJ 302:1302–1305 Parsonnet J, Friedman GD, Vandersteen DP, Chang Y, Vogelman JH, Orentreich N, Sibley RK, 1991, Helicobacter pylori infection and the risk of gastric carcinoma. N Engl J Med 325:1127–1131 Milne AN, Sitarz R, Carvalho R, Carneiro F, Offerhaus GJ, 2007, Early onset gastric cancer: on the road to unraveling gastric carcinogenesis. Curr Mol Med 7:15–28 El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, Herrera J, Lissowska J, Yuan CC, Rothman N, Lanyon G, Martin M, Fraumeni JF Jr, Rabkin CS, 2000, Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature 404:398–402 Milne AN, Offerhaus GJA, 2010, Early-onset gastric cancer: Learning lessons from the young. World J Gastrointest Oncol 2, 2):59–64 Correa P, Shiao YH, 1994, Phenotypic and genotypic events in gastric carcinogenesis. Cancer Res 54:1941s–1943s Maehara Y, Emi Y, Tomisaki S, Oshiro T, Kakeji Y, Ichiyoshi Y, Sugimachi K, 1996, Age-related characteristics of gastric carcinoma in young and elderly patients. Cancer 77:1774–1780 Theuer CP, de Virgilio C, Keese G, French S, Arnell T, Tolmos J, Klein S, PowersW, Oh T, Stabile BE, 1996, Gastric adenocarcinoma in patients 40 years of age or younger. Am J Surg 172:473–476 Hamilton SR, Aaltonen LA. World Health Organization Classification of Tumors. Pathology and Genetics of Tumors of the Digestive System. Lyon, France: IARC Press, 2000: 204 Ramos De la Medina A, Salgado-Nesme N, Torres-Villalobos G, Medina-Franco H, 2004, Clinicopathologic characteristics of gastric cancer in a young patient population. J Gastrointest Surg 8:240–244 Medina-Franco H, Heslin MJ, Cortes-Gonzalez R, 2000, Clinicopathological characteristics of gastric carcinoma in young and elderly patients: a comparative study. Ann Surg Oncol 7:515–519 Matley PJ, Dent DM, Madden MV, Price SK, 1988, Gastric carcinoma in young adults. Ann Surg 208:593–596 Kokkola A, Sipponen P, 2001, Gastric carcinoma in young adults. Hepatogastroenterology 48:1552–1555 Lim S, Lee HS, Kim HS, Kim YI, Kim WH, 2003, Alteration of Ecadherin- mediated adhesion protein is common, but microsatellite instability is uncommon in young age gastric cancers. Histopathology 42:128–136 Maeta M, Yamashiro H, Oka A, Tsujitani S, Ikeguchi M, Kaibara N, 1995, Gastric cancer in the young, with special reference to 14 pregnancy-associated cases: analysis based on 2,325 consecutive cases of gastric cancer. J Surg Oncol 58:191–195 Derakhshan MH, Liptrot S, Paul J, Brown IL, Morrison D, McColl KE, 2009, Oesophageal and gastric intestinal-type adenocarcinomas show the same male predominance due to a 17 year delayed development in females. Gut 58:16–23 Furukawa H, Iwanaga T, Imaoka S, Hiratsuka M, Fukuda I, Kabuto T, Ishikawa O, Sasaki Y, 1989, Multifocal gastric cancer in patients younger than 50 years of age. Eur Surg Res 21:313–318 Suriano G, Oliveira C, Ferreira P, Machado JC, Bordin MC, De Wever O, Bruyneel EA, Moguilevsky N, Grehan N, Porter TR, Richards FM, Hruban RH, Roviello F, Huntsman D, Mareel M, Carneiro F, Caldas C, Seruca R, 2003, Identification of CDH1 germline missense mutations associated with functional inactivation of the E-cadherin protein in young gastric cancer probands. Hum Mol Genet 12:575–582 Suriano G, Yew S, Ferreira P, Senz J, Kaurah P, Ford JM, Longacre TA, Norton JA, Chun N, Young S, OliveiraMJ,Macgillivray B, Rao A, Sears D, Jackson CE, Boyd J, Yee C, Deters C, Pai GS, Hammond LS, McGivern BJ, Medgyesy D, Sartz D, Arun B, Oelschlager BK, UptonMP, Neufeld-KaiserW, Silva OE, Donenberg TR, Kooby DA, Sharma S, Jonsson BA, Gronberg H, Gallinger S, Seruca R, Lynch H, Huntsman DG, 2005, Characterization of a recurrent germ line mutation of the E-cadherin gene: implications for genetic testing and clinical management. Clin Cancer Res 11:5401–5409 Carneiro F, Oliveira C, Suriano G, Seruca R, 2008, Molecular pathology of familial gastric cancer, with an emphasis on hereditary diffuse gastric cancer. J Clin Pathol 61:25–30 Nakamura R, Saikawa Y, Takahashi T, Takeuchi H, Asanuma H, Yamada Y, Kitagawa Y, 2011, Retrospective analysis of prognostic outcome of gastric cancer in young patients. Int J Clin Oncol 16, 4):328–334 Ramos-De la Medina A, Salgado-Nesme N et al, 2004, Clinicopathologic characteristics of gastric cancer in a young patient population. J Gastrointest Surg 8(3):240–244 Koea JB, Karpeh MS, Brennan MF, 2000, Gastric cancer in young patients: demographic, clinicopathological, and prognostic factors in 92 patients. Ann Surg Oncol 7(5):346–351 Hiyama T, Tanaka S, Yoshihara M, Sasao S, Kose K, Shima H, Tuncel H, Ueno Y, Ito M, Kitadai Y, Yasui W, Haruma K, Chayama K, 2004, Chromosomal and microsatellite instability in sporadic gastric cancer. J Gastroenterol Hepatol 19:756–760 Wu M, Semba S, Oue N, Ikehara N, Yasui W, Yokozaki H, 2004, BRAF/K-ras mutation, microsatellite instability, and promoter hypermethylation of hMLH1/MGMT in human gastric carcinomas. Gastric Cancer 7:246–253 Hirahashi M, Yao T, Matsumoto T, Nishiyama K, Oya M, Iida M, Tsuneyoshi M, 2007, Intramucosal gastric adenocarcinoma of poorly differentiated type in the young is characterized by Helicobacter pylori infection and antral lymphoid hyperplasia. Mod Pathol 20(1):29–34 Chung HW, Noh SH, Lim JB, 2010, Analysis of demographic characteristics in 3242 young age gastric cancer patients in Korea. World J Gastroenterol 16(2):256–263 Smith BR, Stabile BE, 2009, Extreme aggressiveness and lethality of gastric adenocarcinoma in the very young. Arch Surg 144, 6):506–510 Santoro R, Carboni F, Lepiane P, Ettorre GM, Santoro E, 2007, Clinicopathological features and prognosis of gastric cancer in young European adults. Br J Surg 94(6):737–742 Lai JF, Kim S, Li C, Oh SJ, Hyung WJ, Choi WH, Choi SH, Wang LB, Noh SH, 2008, Clinicopathologic characteristics and prognosis for young gastric adenocarcinoma patients after curative resection. Ann Surg Oncol 15(5):1464–1469 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 111 EP 117 DI 10.1007/s12253-012-9530-z PG 7 ER PT J AU Rubovszky, G Nagy, T Godeny, M Szasz, A Lang, I AF Rubovszky, Gabor Nagy, Tunde Godeny, Maria Szasz, Andras Lang, Istvan TI Successful Treatment of Solitary Bone Metastasis of Non-Small Cell Lung Cancer with Bevacizumab and Hyperthermia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE Solitary bone metastases; Lung cancer; Hyperthermia; Bevacizumab ID Solitary bone metastases; Lung cancer; Hyperthermia; Bevacizumab AB Non-small cell lung cancer (NSCLC) represents 85 % of all malignant lung cancers. In metastatic disease the principle goal of palliative therapy is to prolong survival with least toxicity and best patients’ quality of life. Bevacizumab (BEV) has been approved as first line treatment in combination with platinum based chemotherapy and maintenance therapy in NSCLC. BEV can be added safely to several chemotherapeutic agents, however there is no data on coadministration with thermotherapy. Even in localized disease no robust evidence exists about the beneficial effect of loco-regional thermotherapy on overall survival, but it might be used successfully in symptom palliation. In this article a successful co-administration of BEV and hyperthermia is reported in a patient with monolocalized bone metastasis from previously operated NSCLC. This case suggests that electrohyperthermia can probably be incorporated in palliative therapy added not only to radiotherapy or chemotherapy but also to anti-angiogenic BEV treatment. C1 [Rubovszky, Gabor] National Institute of OncologyBudapest, Hungary. [Nagy, Tunde] National Institute of OncologyBudapest, Hungary. [Godeny, Maria] National Institute of OncologyBudapest, Hungary. [Szasz, Andras] Szt. Istvan University, Biotechnics DepartmentGodollo, Hungary. [Lang, Istvan] National Institute of OncologyBudapest, Hungary. RP Rubovszky, G (reprint author), National Institute of Oncology, Budapest, Hungary. EM garub@oncol.hu CR Wakelee HA, Bernardo P, Johnson DH et al, 2006, Changes in the natural history of nonsmall cell lung cancer, NSCLC)–comparison of outcomes and characteristics in patients with advanced NSCLC entered in Eastern Cooperative Oncology Group trials before and after 1990. Cancer 06:2208–2217 Yang P, Allen MS, Aubry MC et al, 2005, Clinical features of 5,628 primary lung cancer patients: experience at Mayo Clinic from 1997 to 2003. Chest 28:452–462 Govindan R, Page N, Morgensztern D et al, 2006, Changing epidemiology of small-cell lung cancer in the United States over the last 30 years: analysis of the surveillance, epidemiologic, and end results database. J Clin Oncol 24:4539–4544 Azzoli CG, Baker S, Temin S et al, 2009, American Society of Clinical Oncology Clinical Practice guideline update on chemotherapy for Stage IV non-Nmall-Cell Lung Cancer. J Clin Oncol 27:6251–6266 Sandler A, Gray R, Perry MC, 2006, Paclitaxel–carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 355:2542–2550 Reck M, von Pawel J, Zatloukal P, 2009, Phase iii trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer: avail. J Clin Oncol 27:1227–1234 Laskin J, Crino L, Tsai C, 2009, MO19390, SAIL): first-line bevacizumab-based therapy in advanced non-small cell lung cancer, nsclc)—outcome by chemotherapy regimen [abstract C2.5]. J Thorac Oncol 4(Suppl 1):S359 Hirsh V, 2008, Emerging safety data for bevacizumab in advanced non–small-cell lung cancer. Clin Lung Cancer 9(suppl 2):S62–S70 Crino L, Mezger J, Griesinger F et al, 2009, MO19390, SAIL): safety and efficacy of first-line bevacizumab, Bv)-based therapy in advanced non-small cell lung cancer, nsclc, [abstract 8043] Proc Am Soc. Clin Oncol 27:417s Fischbach N, Spigel D, Brahmer J, on behalf of the ARIES investigators, 2009, Preliminary safety and effectiveness of bevacizumab, bv, based treatment in subpopulations of patients, pts, with non-small cell lung cancer, nsclc, from the aries study: a bevacizumab, bv, treatment observational cohort study, ocs, [abstract 8040]. Proc Am Soc Clin Oncol 27:416s Mezger J, von Pawel J, Reck M, 2009, Bevacizumab, Bv, singleagent maintenance following Bv-based chemotherapy in patients with advanced non-small cell lung cancer, nsclc): results from an exploratory analysis of the avail study [abstract e19001] Proc Am Soc. Clin Oncol 27:e19001 Miller V, O’Connor P, Soh C, on behalf of the ATLAS Investigators, 2009, A randomized, double-blind, placebo-controlled, phase iiib trial, atlas, comparing bevacizumab, b, therapy with or without erlotinib, e, after completion of chemotherapy with b for firstline treatment of locally advanced, recurrent, or metastatic nonsmall cell lung cancer, nsclc, [abstract LBA8002] Proc Am Soc. Clin Oncol 27:799s Hildebrandt B, Wust P, Ahlers O et al, 2002, The cellular and molecular basis of hyperthermia. Clin Rev Oncol Haemat 43:33–56 Jiang Z, Yan W, Ming J et al, 2007, Docetaxel weekly regimen in conjunction with RF hyperthermia for pretreated locally advanced non-small cell lung cancer: a preliminary study. BMC Cancer 7:189 Shen H, Li XD, Wu CP et al, 2011, The regimen of gemcitabine and cisplatin combined with radio frequency hyperthermia for advanced non-small cell lung cancer: a phase II study. Int J Hyperthermia 27:27–32 Mitsumori M, Zeng ZF, Oliynychenko P et al, 2007, Regional hyperthermia combined with radiotherapy for locally advanced non-small cell lung cancers: a multi-institutional prospective randomized trial of the International Atomic Energy Agency. Int J Clin Oncol 12:192–198 Moon SD, Ohguri T, Imada H et al, 2011, Definitive radiotherapy plus regional hyperthermia with or without chemotherapy for superior sulcus tumors: a 20-year, single center experience. Lung Cancer 71:338–343 Sumiyoshi K, Strebel FR, Rowe RWet al, 2003, The effect of wholebody hyperthermia combined with ‘metronomic’ chemotherapy on rat mammary adenocarcinoma metastases. Int J Hyperthermia 19:103–118 Hermisson M, Weller M, 2000, Hyperthermia enhanced chemosensitivity of human malignant glioma cells. Anticancer Res 20, 3A):1819–1823 Szasz A, Szasz N, Szasz O, 2010, Oncothermia –principles and practices. Springer Verlag, Heidelberg Chun-Yi L, Yiing-Feng H, Hua-Tzu H et al, 2009, Lung cancer with solitary bone metastasis in the radius. Clin Nucl Med 34:684– 685 Shih WJ, Magoun S, Lahar B et al, 1998, An unusual case of a tibial metastasis as the clinical presentation of bronchogenic adenocarcinoma. J Nucl Med Technol 26:91–93 An SJ, Huang YS, Chen ZH, 2011, et al. Posttreatment plasma VEGF levels may be associated with the overall survival of patients with advanced non-small cell lung cancer treated with bevacizumab plus chemotherapy, Med Oncol [Epub ahead of print] Horn L, Dahlberg SE, Sandler AB et al, 2009, Phase II study of cisplatin plus etoposide and bevacizumab for previously untreated, extensive-stage small-cell lung cancer: Eastern Cooperative Oncology Group Study E3501. J Clin Oncol 27:6006– 6011 Herbst RS, Ansari R, Bustin F et al, 2011, Efficacy of bevacizumab plus erlotinib versus erlotinib alone in advanced non-smallcell lung cancer after failure of standard first-line chemotherapy, BeTa): a double-blind, placebo-controlled, phase 3 trial. Lancet 377:1846–1854 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 119 EP 122 DI 10.1007/s12253-012-9551-7 PG 4 ER PT J AU Haltrich, I Csoka, M Kovacs, G Torok, D Alpar, D Ottoffy, G Fekete, Gy AF Haltrich, Iren Csoka, Monika Kovacs, Gabor Torok, Dora Alpar, Donat Ottoffy, Gabor Fekete, Gyorgy TI Six Cases of Rare Gene Amplifications and Multiple Copy of Fusion Gene in Childhood Acute Lymphoblastic Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Childhood ALL; Gene amplification; FISH; Subtle rearrangement ID Childhood ALL; Gene amplification; FISH; Subtle rearrangement AB Cytogenetic aberrations are very important factors in risk assessment of childhood hematological malignancies. We report six childhood acute lymphoid leukemia (ALL) cases with rare cytogenetic aberrations: five with RUNX1, ABL1 or MLL proto-oncogene amplification and one case of multiple copies of ETV6/RUNX1 fusion genes. The simultaneous presence of two adverse genetic aberrations is of special interest: ETV6-RUNX1 fusion gene is associated with good prognosis and intrachromosomal amplification of the homologue RUNX1 gene is associated with poor prognosis. We also report a patient with MLL amplification, a unique finding in childhood T-ALL. Report of these subtle rearrangements contributes to our understanding of diagnostic and prognostic significance of these rare cytogenetic abnormalities. C1 [Haltrich, Iren] Semmelweis University, 2nd Department of Pediatrics, Tuzolto utca 7-9, 1094 Budapest, Hungary. [Csoka, Monika] Semmelweis University, 2nd Department of Pediatrics, Tuzolto utca 7-9, 1094 Budapest, Hungary. [Kovacs, Gabor] Semmelweis University, 2nd Department of Pediatrics, Tuzolto utca 7-9, 1094 Budapest, Hungary. [Torok, Dora] Semmelweis University, 2nd Department of Pediatrics, Tuzolto utca 7-9, 1094 Budapest, Hungary. [Alpar, Donat] University of Pecs, Department of PathologyPecs, Hungary. [Ottoffy, Gabor] University of Pecs, Department of PediatricsPecs, Hungary. [Fekete, Gyorgy] Semmelweis University, 2nd Department of Pediatrics, Tuzolto utca 7-9, 1094 Budapest, Hungary. RP Haltrich, I (reprint author), Semmelweis University, 2nd Department of Pediatrics, 1094 Budapest, Hungary. EM haltrich.iren@med.semmelweis-univ.hu CR Bruyere H, Sutherland H, Chipperfield K, Hudoba M, 2010, Concomitant and successive amplifications of MYC in APL-like leukemia. Cancer Genet Cytogenet 197:75–80 Gebhart E, 2005, Double minutes, cytogenetic equivalents of gene amplification, in human neoplasia - a review. Clin Transl Oncol 7:477–485 Attarbaschi A, Mann G, Panzer-Grumayer R et al, 2008, Minimal residual disease values discriminate between low and high relapse risk in children with B-cell precursor acute lymphoblastic leukemia and an intrachromosomal amplification of chromosome 21: the Austrian and German acute lymphoblastic leukemia Berlin- Frankfurt-Munster, ALL-BFM, trials. J Clin Oncol 26:3046–3050 Mohamed A, 2010, MLL amplification in leukemia. Atlas Genet Cytogenet Oncol Haematol. URL: http://AtlasGeneticsOncology.org/ Anomalies/MLLampliID1547.html Moorman AV, Ensor HM, Richards SM et al, 2010, Prognostic effect of chromosomal abnormalities in childhood B-cell precursor acute lymphoblastic leukaemia: results from the UK Medical Research Council ALL97/99 randomised trial. Lancet Oncol 11:429–438 Peter A, Heiden T, Taube T et al, 2009, Interphase FISH on TEL/ AML1 positive acute lymphoblastic leukemia relapses-analysis of clinical relevance of additional TEL and AML1 copy number changes. Eur J Haematol 83:420–32 Attarbaschi A, Mann G, Konig M et al, 2004, Incidence and relevance of secondary chromosome abnormalities in childhood TEL/AML1+ acute lymphoblastic leukemia: an interphase FISH analysis. Leukemia 18:1611–1616 Al-Sweedan SA, Neglia JP et al, 2007, Characteristics of patients with TEL-AML1-positive acute lymphoblastic leukemia with single or multiple fusions. Pediatr Blood Cancer 48:510–514 Moorman AV, Richards SM, Robinson HM et al, 2007, Prognosis of children with acute lymphoblastic leukemia, ALL, and intrachromosomal amplification of chromosome 21, iAMP21). Blood 109:2327–2330 Mikhail FM, Serry KA, Hatem N et al, 2002, AML1 gene overexpression in childhood acute lymphoblastic leukemia. Leukemia 16:658–668 PaisAP,Amare KadamPS, RajeGC et al, 2008, RUNX1 aberrations in ETV6/RUNX1-positive and ETV6/RUNX1-negative patients: its hemato-pathological and prognostic significance in a large cohort, 619 cases, of ALL. Pediatr Hematol Oncol 25:582–597 Kim HJ, Woo HY, Koo HH et al, 2004, ABL oncogene amplification with p16, INK4a, gene deletion in precursor T-cell acute lymphoblastic leukemia/lymphoma: report of the first case. Am J Hematol 76:360–363 Barber KE, Martineau M, Harewood L et al, 2004, Amplification of the ABL gene in T-cell acute lymphoblastic leukemia. Leukemia 18:1153–1156 Bernasconi P, Calatroni S, Giardini I et al, 2005, ABL1 amplification in T-cell acute lymphoblastic leukemia. Cancer Genet Cytogenet 162:146–150 Graux C, Cools J, Melotte C et al, 2004, Fusion ofNUP214 to ABL1 on amplified episomes in T-cell acute lymphoblastic leukemia. Nat Genet 36:1084–1089 Hagemeijer A, Graux C, 2010, ABL1 rearrangements in T-cell acute lymphoblastic leukemia. Genes Chromosomes Cancer 49:299–308 Quintas-Cardama A, Tong W, Manshouri T et al, 2008, Activity of tyrosine kinase inhibitors against human NUP214-ABL1-positive T cell malignancies. Leukemia 22:1117–1124 Sarova I, Brezinova J, Zemanova Z et al, 2010, Cytogenetic manifestation of chromosome 11 duplication/amplification in acute myeloid leukemia. Cancer Genet Cytogenet 199:121–127 Cuthbert G, Thompson K, McCullough S et al, 2000, MLL amplification in acute leukaemia: a United Kingdom Cancer Cytogenetics Group, UKCCG, study. Leukemia 14:1885–1891 Espinet B, Florensa L, Salido M, Sole F, 2003, MLL intrachromosomal amplification in a pre-B acute lymphoblastic leukemia. Haematologica 88:EIM03 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 123 EP 128 DI 10.1007/s12253-012-9533-9 PG 6 ER PT J AU Kontic, M Stojsic, J Stevic, R Bunjevacki, V Jekic, B Dobricic, V AF Kontic, Milica Stojsic, Jelena Stevic, Ruza Bunjevacki, Vera Jekic, Biljana Dobricic, Valerija TI Could Spindle Cell Lung Carcinoma be Considered and Treated as Sarcoma, According to its Clinical Course, Morphology, Immunophenotype and Genetic Finding? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Spindle cell lung carcinoma; Immunophenotype; Oncogenetic; p53; Sarcoma ID Spindle cell lung carcinoma; Immunophenotype; Oncogenetic; p53; Sarcoma AB The actual nature of spindle cell carcinoma has been debated extensively because of its rarity. It carries a poor prognosis, even when early-stage disease is diagnosed and resected. In view of the rarity and the significance of the histological diagnosis, we report a patient with rapidly progressing spindle cell lung carcinoma with soft tissue metastasis. Diagnosis was confirmed by immunohistochemistry finding. Analysis of the TP53 gene mutations by polymerase chain reaction and DNA sequencing revealed insertion of single thymine resulting in frameshift mutation in the exon 8. Prognosis of spindle cell lung carcinoma might be determined by the sarcoma component of the tumor and, based on that, we wonder if this type of lung carcinoma could be followed-up and treated by strategies for soft tissue sarcomas, because of its rapid, sarcomatous type of growth, beside the properly lung carcinoma oncological treatment. C1 [Kontic, Milica] Clinical Centre of Serbia, Institute for Lung Diseases and TuberculosisBelgrade, Serbia. [Stojsic, Jelena] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia. [Stevic, Ruza] Clinical Centre of Serbia, Institute of RadiologyBelgrade, Serbia. [Bunjevacki, Vera] University in Belgrade, Medical Faculty, Institute for Human GeneticsBelgrade, Serbia. [Jekic, Biljana] University in Belgrade, Medical Faculty, Institute for Human GeneticsBelgrade, Serbia. [Dobricic, Valerija] University in Belgrade, Medical Faculty, Clinical Centre of Serbia, Institute of NeurologyBelgrade, Serbia. RP Kontic, M (reprint author), Clinical Centre of Serbia, Institute for Lung Diseases and Tuberculosis, Belgrade, Serbia. EM milicakontic@yahoo.com CR Mainwaring MG, Poor C, Zander DS, Harman E, 2000, Complete remission of pulmonary spindle cell carcinoma after treatment with oral germanium sesquioxide. Chest 117:591–593 Yuki T, Sakuma T, Ohbayashi C, Yoshimura M et al, 2007, Pleomorphic carcinoma of the lung: a surgical outcome. J Thorac Cardiovasc Surg 134:399–404 Travis WD, Brambilla E, Muller-Hermelink HK, Harris CC et al, 2004, World health organisation classification of tumours. Pathology and genetics of the lung, pleura, thymus and heart. IARC Press, Lyon, pp 512–536 Travis WD, 2002, Pathology of lung cancer. Clin Chest Med 23:65–81 Attanoos RL, Papagiannis A, Suttinont P, Goddard H, Papotti M, Gibbs AR, 1998, Pulmonary giant cell carcinoma: pathological entity or morphological phenotype? Histopathology 32:225–231 Stojsic J, Stevic R, Kontic M, Stojsic Z, Drndarevic N, Bunjevacki V, Jekic B, 2011, Large cell lung carcinoma with unusual imaging feature. Immunophenotype and genetic finding. Pathol Oncol Res 17:175–179 Kim TS, Han J, Lee KS, Jeong YJ, Kwak SH, Byun HS, ChungMJ, Kim H, Kwon OJ, 2005, CT findings of surgically resected pleomorphic carcinoma of the lung in 30 patients. AJR 185:120–125 Kim TH, Kim SJ, Ryu YH, Lee HJ, Goo JM, Im J-G, 2004, Pleomorphic carcinoma of lung: comparison of CT features and pathologic findings. Radiology 232:554–559 Venissac N, Pop D, 2007, Sarcomatoid lung cancer, spindle/giant cells): an aggressive disease? J Thorac Cardiovasc Surg 134:619– 623 Kontic M, Stojsic J, Kacar-Kukric V, Jekic B, Bunjevacki V, 2010, Multidisciplinary approach in diagnosis of lung carcinoma. Exp Oncol 32:111–113 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 129 EP 133 DI 10.1007/s12253-012-9562-4 PG 5 ER PT J AU Villaran, J Loaiza-Bonilla, A Parra-Herran, C Pinto, A AF Villaran, Jorge Loaiza-Bonilla, Arturo Parra-Herran, Carlos Pinto, Andre TI Pelvic Angiosarcoma Occurring in a Postmenopausal Female: Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports C1 [Villaran, Jorge] University of Miami, Jackson Health System MiamiMiami, FL, USA. [Loaiza-Bonilla, Arturo] University of Miami, Jackson Health System MiamiMiami, FL, USA. [Parra-Herran, Carlos] Brigham and Women’s Hospital, Department of PathologyBoston, MA, USA. [Pinto, Andre] University of Miami, Jackson Health System MiamiMiami, FL, USA. RP Pinto, A (reprint author), University of Miami, Jackson Health System Miami, Miami, USA. EM Apinto1@med.miami.edu CR Mankin H, Hornicek F, 2005, Diagnosis, classification, and management of soft tissue sarcomas. Cancer Control 12(1):5– 21 Park M, Ravi V, Araujo D, 2010, Inhibiting the VEGF-VEGFR pathway in angiosarcoma, epithelioid hemangioendothelioma, and hemangiopericytoma/solitary fibrous tumor. Curr Opin Oncol 22, 4):351–5 Young R, Brown N, Reed M et al, 2010, Angiosarcoma. Lancet Oncol 11:983–91 Hart J, Mandavilli S, 2011, Epithelioid angiosarcoma: a brief diagnostic review and differential diagnosis. Arch Pathol Lab Med 135(2):268–72 Cambruzzi E, Pegas KL, Milani DM, Cruz RP, Guerra EH, Ferrari MB, 2010, Angiosarcoma arising in an ovarian fibroma: a case report. Pathol Res Int 842592 Bradford L, Swartz K, Rose S, 2010, Primary angiosarcoma of the ovary complicated by hemoperitoneum: a case report and review of the literature. Arch Gynecol Obstet 281:145–150 Bosmuller H, Gruber C, Haitchi-Petnehazy S, Dietmar Wagner D, Webersinke G, Hauptmann S, 2011, Primary angiosarcoma of the ovary with prominent fibrosis of the ovarian stroma. Case report of an 81-year old patient. Diagn Pathol 6:65–71 Ali J, Ko H, Owen D et al, 2006, Epithelioid angiosarcoma of the small bowel. Gastrointest Endosc 64(6):1018–21 Fayette J,Martin E, Piperno-Neumann S et al, 2007, Angiosarcomas, a heterogeneous group of sarcomas with specific behavior depending on primary site: a retrospective study of 161 cases. Ann Oncol 18:2030–2036 Aragon L, Terreros D, Ho H et al, 2011, Angiosarcoma of the ovary arising in a mucinous cystadenoma. J Clin Ultrasound 39:351–355 Davidson B, Abeler VM, 2005, Primary ovarian angiosarcoma presenting as malignant cells in ascites: case report and review of the literature. Diagn Cytopathol 32(5):307–9 Jhaa S, Chan K, Poole C et al, 2005, Pregnancy following recurrent angiosarcoma of the ovary- A case report and review of literature. Gynecol Oncol 97:935–937 Platt JS, Rogers SJ, Flynn EA, 1999, Primary angiosarcoma of the ovary: a case report and review of the literature. Gynecol Oncol 73:443–446 Ariyoshi K, Kawauchi S, Kaku T et al, 2000, Prognostic factors in ovarian carcinosarcoma: a clinicopathological and immunohistochemical analysis of 23 Cases. Histopathology 37:427–436 Treuner J, Koscielniak E, et al, 2003, Cooperative Weichteilsarkom Study CWS-2002p, Der Gesellschaft fur Padiatrische Onkologie und Hamatologie, GPOH); Fassung Contreras A,Malpica A, et al, 2009, Angiosarcoma arising inmature cystic teratoma of the ovary: a case report and review of the literature. International Journal of Gynecological Pathology 28(5) Cunningham M, Brooks J, Noumoff J, 1994, Treatment of primary ovarian angiosarcoma with ifosfamide and doxorubicin. Gynecol Oncol 53(2):265–8 Lahat G, Dhuka A, Hallev Hi et al, 2010, Angiosarcoma: clinical and molecular insights. Ann Surg 251:1098–1106 Furihata M, Takeuchi T, Iwata J et al, 1998, Primary ovarian angiosarcoma: a case report and literature review. Pathol Int 48:967–973 Nara M, Sasaki T, Shimura S et al, 1996, Diffuse alveolar hemorrhage caused by lung metastasis of ovarian angiosarcoma. Intern Med 35, 8):653–6 Virtanen A, Pukkala E, Auvinen A, 2007, Angiosarcoma after radiotherapy: a cohort study of 332 163 Finnish cancer patients. Br J Cancer 97:115–117 Iljazovic E, Tomic S, Mustedanagic-Mujanovic J et al, 2011, Angiosarcoma of the ovary in an 11 year old girl: case report and review of the literature. Bosn J Basic Med Sci 11(2):132–136 Penel N, Marreaud S, Robin YM et al, 2011, Angiosarcoma: state of the art and perspectives. Crit Rev Oncol Hematol 80(2):257–63 Bosmuller et al, 2011, Primary angiosarcoma of the ovary with prominent fibrosis of the ovarian stroma. Case report of an 81-year old patient. Diagnostic Pathology 6:65 Sood A, Sorosky J, Gelder M et al, 1998, Primary ovarian sarcoma: analysis of prognostic variables and the role of surgical cytoreduction. Cancer 82(9):1731–7 Yi D, Keng S, Jing-he L et al, 2011, Primary sarcoma of the ovary: clinicopathological characteristics, prognostic factors and evaluation of therapy. Chin Med J 124(9):1316–1321 Potter ME, Moradi M, Alexander CW et al, 1989, Value of serum 125Ca levels: does the result preclude second look? Gynecolog Oncology 33:201–3 Cherie D, Quesenberrya C, Chen A, 2005, Primary angiosarcoma of the ovary: a case report of stage I disease. Gynecol Oncol 99:218–221 Nielsen GP, Young RH, Prat J et al, 1997, Primary angiosarcoma of the ovary: a report of seven cases and review of the literature. Int J Gynecol Pathol 16(4):378–82 Bradford L, Swartz K, Rose S, 2010, Primary angiosarcoma of the ovary complicated by hemoperitoneum: a case report and review of the literature. Arch Gynecol Obstet 281:145–150 Serrano C, Garcia A, Brana I et al, 2010, Angiosarcoma of the ovary: is it always a lethal disease? J Clin Oncol 28(33):e675–e677 Cambruzzi E, Pegas KL, Milani DM et al, 2010, Angiosarcoma arising in an ovarian fibroma: a case report. Pathol Res Int 2010:842592 Kiesel H, Muller AM, Schmitt-Graeff A et al, 2009, Dramatic and durable efficacy of imatinib in an advanced angiosarcoma without detectable KIT and PDGFRA mutations. Cancer Biol Ther 8:319–321 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2013 VL 19 IS 1 BP 135 EP 139 DI 10.1007/s12253-012-9540-x PG 5 ER PT J AU Balogh, P Katz, S Kiss, LA AF Balogh, Peter Katz, Sandor Kiss, L Anna TI The Role of Endocytic Pathways in TGF-β Signaling SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE TGF-β; EMT; Endocytosis; Early endosome; Caveolae ID TGF-β; EMT; Endocytosis; Early endosome; Caveolae AB Transforming growth factor β (TGF-β) superfamily consists of numerous cytokins that regulate various cellular processes. TGF-β, the prototype of the family, signals through its cell surface serine/threonin kinase receptors and besides its role in cell differentiation, migration, adhesion etc. it is also able to induce epithelial-mesenchymal (EMT) transition via both Smadpathway and MAPK- pathway. Among the different types of epithelial-mesenchymal transition, type II that is described to be associated with wound healing, tissue regeneration, organ fibrosis and is induced upon inflammatory stimuli. It can be triggered by secretion of growth factors such as TGF-β, EGF. Different endocytic routes are used for the internalization of TGF-β ligand and its receptors and these pathways can control the activity of downstream events. Internalization via clathrin-coated vesicles promotes the signaling while the caveolamediated endocytosis plays important role in the termination of the events, although the steps of the latter event are less clear. The early endosome is considered a clue compartment in promoting the signaling. Recently published data suggest that the early endosome plays crucial role in the termination of the TGFβ signaling as well. It is not only maintain a special environment for the effective signaling but can direct the internalized cargos towards degradative pathways (multivesicular bodies, lysosomes). C1 [Balogh, Peter] Semmelweis University, 1st Department of Anatomy, Tuzolto u. 58, 1094 Budapest, Hungary. [Katz, Sandor] Semmelweis University, 1st Department of Anatomy, Tuzolto u. 58, 1094 Budapest, Hungary. [Kiss, L Anna] Semmelweis University, 1st Department of Anatomy, Tuzolto u. 58, 1094 Budapest, Hungary. RP Balogh, P (reprint author), Semmelweis University, 1st Department of Anatomy, 1094 Budapest, Hungary. EM balogh.petra@med.semmelweis-univ.hu CR Massague J, 1998, TGF-β signal transduction. Annu Rev Biochem 67:753–91 Shook D, Keller R, 2003, Mechanisms, mechanics and function of epithelial-mesenchymal transitions in early development. Mech Dev 120:1351–83 Strutz F, Zeisberg M, Ziyadeh FN, Yang CQ, Kalluri R, Muller GA, Neilson EG, 2002, Role of basic fibroblast growth factor-2 in epithelial-mesenchymal transformation. Kidney Int 61:1714–28 Kalluri R, Weinberg RA, 2009, The basics of epithelialmesenchymal transition. J Clin Invest 119:1420–28 Wrana JL, Attisano L, Wieser R, Ventura F, Massague J, 1994, Mechanism of activation of TGF-β receptor. Nature 370:341–47 Shi Y, Massague J, 2003, Mechanisms of TGF-β signaling from cell membrane to the nucleus. Cell 113:685–700 Moustakas A, Souchelnytskyi S, Heldin CH, 2001, Smad regulation in TGF-β signal transduction. J Cell Sci 11:4359–69 Zhang Y, Chang C, Gehling JD, Hemmati-Brivanlou A, Derynck R, 2001, Regulation of Smad degradation and activity by Smurf2, an E3 ubiquitin ligase. Proc Natl Acad Sci U S A 98:974–79 Tsukazaki T, Chiang TA, Davison AF, Attisano L, Wrana L, 1998, SARA, a FYVE domain protein that recruits Smad2 to the TGF-β receptor. Cell 95:779–91 Di Guglielmo GM, Le Roy C, Goodfellow AF, Wrana L, 2003, Distinct endocytic pathways regulate TGF-β receptor signalling and turnover. Nat Cell Biol 5:410–21 Chen YG, 2009, Endocytic regulation of TGF-β signaling. Cell Res 19:58–70 Chen YG, Wang Z, Ma Y, Zhang L, Lu Z, 2007, Endofin, a FYVE domain protein, interacts with Smad4 and facilitates transforming growth factor-beta signaling. J Biol Chem 282:9688–96 Gillooly DJ, Simonsen A, Stenmark H, 2001, Cellular functions of phosphatidylinositol 3-phosphate and FYVE domain proteins. Biochem J 355:249–58 Itoh F, Divecha N, Brocks L, Brocks L, Oomen L, Janssen H, Calafar J, Itoh S, Dijke PP, 2002, The FYVE domain in Smad anchor for receptor activation, SARA, is sufficient for localization of SARA in early endosomes and regulates TGF-beta/Smad signalling. Genes Cells 7:321–31 Le Roy C, Wrana L, 2005, Clathrin- and non-clathrin- mediated endocytic regulation of cell signalling. Nat Rev Cell Biol 6:112–26 Bonifacino JS, Lippincott-Schwartz J, 2003, Coat proteins: shaping membrane transport. Nat Rev Mol Cell Biol 4:409–14 Bonifacino JS, Traub LM, 2003, Signals for sorting of transmembrane proteins to endosomes and lysosomes. Annu Rev Biochem 72:395–447 Hayes S, Chawla A, Corvera S, 2002, TGFβ receptor internalization into EEA1-enriched early endosomes: role in signaling to Smad2. J Cell Biol 158:1239–49 Penheiter SG, Mitchell H, Garamszegi N, Edens M, Dore JJJ, Leof EB, 2002, Internalization-dependent and –independent requirements for trasfotming growth factor beta receptor signaling via the Smad pathway. Mol Cell Biol 22:4750–59 Xu L, Chen YG, Massague J, 2000, The nuclear import function of Smad2 is masked by SARA and unmasked by TGFb-dependent phosphorylation. Nat Cell Biol 2:559–62 Heldin CH, Moustakas A, 2012, Role of Smads in TGFβ signaling. Cell Tissue Res 347:21–36 Kurisaki A, Kose S, Yoneda Y, Heldin CH, Moustakas A, 2001, Transforming growth factor-beta induces nuclear import of Smad3 in an importin-beta1 and Ran-dependent manner. Mol Biol Cell 12:1079–91 Xiao Z, Latek R, Lodish HF, 2003, An extended bipartite nuclear localization signal in Smad4 is required for its nuclear import and transcriptional activity. Oncogene 22:1057–69 Mitchell H, Choudhury A, Pagano RE, Leof EB, 2004, Liganddependent and –independent transforming growth factor-beta receptor recycling regulated by clathrin-mediated endocytosis and Rab11. Mol Biol Cell 15:4166–78 Lisanti MP, Scherer P, Tang ZL, Sargiacomo M, 1994, Caveolae, caveolin and caveolin-rich membrane domains: a signalling hypothesis. Trends Cell Biol 4:231–35 Couet J, Li S, Okamoto T, Scherer P, Lisanti MP, 1997, Molecular and cellular biology of caveolae: paradoxes and plasticities. Trends Cardiovasc Med 7:103–10 Pelkmans L, Kartenbeck J, Helenius A, 2001, Caveolar endocytosis of simian virus 40 reveals a new two-step vesicular transport pathway to the ER. Nat Cell Biol 3:475–83 Nicols BJ, 2002, A distinct class of endosome mediates clathrinindependent endocytosis to the Golgi complex. Nat Cell Biol 4:374–78 Parton RG, Simons K, 2007, The multiple faces of caveolae. Nat Rev Mol Cell Biol 8:185–94 Kiss AL, Botos E, 2009, Endocytosis via caveolae: alternative pathway with distinct cellular compartments to avoid lysosomal degradation? J Cell Mol Med 13:1228–37 Hayer A, Stoaber M, Ritz D, Engel S, Meyer HH, Helenius A, 2010, Caveolin-1 is ubiquitinated and targeted to intraluminal vesicles in endolysosomes for degradation. J Cell Biol 191:615–29 Nakao A, Afrakhte M, Moren A, Nakayama T, Christian JL, Heuchel R, Itoh S, Kawabata M, Heldin NE, Heldin CH, PtP D, 1997, Identification of Smad7, a TGF-beta inducible antagonist of TGF-beta signalling. Nature 389:631–35 Hayashi H, Abdollah S, Qui Y, Cai J, Xu JJ, Grinnell BW, Richardson MA, Topper JN, Gimbrone MA Jr, Wrana JL, Falb D, 1997, The MAD-related protein Smad7 associates with the TGFbeta receptor and functions as an antagonist of TGFbeta signaling. Cell 89:1165–73 Zhang S, Fei T, Zhang L, Zhang R, Chen F, Ning Y, Han Y, Feng XH, Meng A, Chen YG, 2007, Smad7 antagonizes transforming growth factor beta signaling in the nucleus by interfering with functional Smad-DNA complex formation. Mol Cell Biol 27:4488– 99 Kavsak P, Rasmussen RK, Causing CG, Bonni S, Zhu H, Thomsen GH, Wrana JL, 2000, Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGF-beta receptor for degradation. Mol Cell 6:1365–75 Ebisawa T, Fukuchi M, Murakami G, Chiba T, Tanaka K, Imamura T, Miyazono K, 2001, Smurf1 interacts with transforming growth factor-beta type-I receptor through Smad7 and induces receptor degradation. J Biol Chem 276:12477–80 Itoh S, Landstrom M, Hermansson A, Itoh F, Heldin CH, Heldin NE, PtP D, 1998, Transforming growth factor beta1 induces nuclear export of inhibitory Smad7. J Biol Chem 273:29195–201 Kowanetz M, Lonn P, Vanlandewijck M, Kowanetz K, Heldin CH, Moustakas A, 2008, TGFbeta induces SIK to negatively regulate type I receptor kinase signaling. J Cell Biol 182:655–62 Bizet AA, Liu K, Tran-Khanh N, Saksena A, Vorstenbosch J, Finnson KW, Buschmann MD, Philip A, 2011, The TFG-β coreceptor, CD109, promotes internalization and degradation of TGF-β receptors. Biochim Biophys Acta 1813:742–53 Mukhopadhyay D, Riezman H, 2007, Proteasome-independent functions of ubiquitin in endocytosis and signaling. Science 315:201–205 Ciechanover A, 2005, Proteolysis: from the lysosome to ubiquitin and the proteasome. Mol Cel Biol 6:79–86 Felder S, Miller K, Moshren G, Ullrich A, Schlessinger J, Hopkins CR, 1990, Kinase activity controls the sorting of the epidermal growth factor receptor within the multivesicular body. Cell 61:623–34 Gruenberg J, Maxfield F, 1995, Membrane transport in the endocytic pathway. Curr Op Cell Biol 7:552–63 von Gersdoff G, Susztak K, Rezvani F, Bitzer M, Liang D, Bottinger EP, 2000, Smad3 and Smad4 mediate transcriptional activation of the human Smad7 promoter by transforming growth factor beta. J Biol Chem 275:11320–26 Katzmann DJ, Babst M, Emr DS, 2001, Ubiquitin-dependent sorting into the multivesicular body pathway requires the function of a conserved endosomal protein sorting complex, ESCRT-I. Cell 106:145–55 Soond SM, Chantry A, 2011, How ubiqitination regulates the TGF-β signalling pathway: new insights and new players. Bioessays 33:749–58 Ito I, Hanyu A, Wayama M, Goto N, Katsuno Y, Kawasaki S, Nakajima Y, Kajiro M, Komatsu Y, Fujimura A,Hirota R,Murayama A, Kimura K, Imamura T, Yanagisawa J, 2010, Estrogen inhibits transforming growth factor β signaling by promoting Smad2/3 degradation. J Biol Chem 285:14747–55 Derynck R, Zhang YE, 2003, Smad-dependent and Smadindependent pathways in TGF-β family signalling. Nature 424:577– 584 Moustakas A, Heldin CH, 2005, Non-Smad TGF-β signals. J Cell Sci 118:3573–3584 Chuderland D, Seger R, 2005, Protein-protein interactions in the regulation of the extracellular signal-regulated kinase. Mol Biothechnol 29:57–74 Adachi M, Fukuda M, Nishida E, 2000, Nuclear export of MAP kinase, Erk, involves a MAP kinase kinase, MEK)-dependent active transport mechanism. J Cell Biol 148:849–856 Chen RH, Sarnecki C, Blenis J, 1992, Nuclear localization and regulation of erk- and rsk-encoded protein kinase. Mol Cell Biol 12:915–927 Kamaraju AK, Roberts AB, 2005, Role of Rho/ROCK and p38 MAP kinase pathways in transforming growth factor-β-mediated Smad-dependent growth inhibition of human breast carcinom cells in vivo. J Biol Chem 280:1024–1036 Mori S, Matsuzaki K, Yoshida K, Furukawa F, Tahashi Y, Yamagata H, Sekimoto G, Seki T, Matsui H, NishizawaM, Fujisawa J, Okazaki K, 2004, TGF-β and HGF transmit the signals through JNKdependent Smad2/3 phosphorylation at the linker region. Oncogene 23:7416–7429 Kretzschmar M, Doody J, Timokhina I, Massague J, 1999, A mechanism of repression of TGF-β/Smad signaling by oncogenic Ras. Genes Dev 13:804–816 Engel M, McDonnell MA, Law BK, Moses HL, 1999, Interdependent Smad and JNK signaling in trasforming growth factor-β- mediated transcription. J Biol Chem 274:37413–37420 Kang JS, Liu C, Derynck R, 2009, New regulatory mechanisms of TGF-β receptor function. Trends Cell Biol 19:385–394 Galliher AJ, Schiemann WP, 2007, Src phosphorylates Tyr284 in TGF-β type II receptor and regulates TGF-β stimulation of p38 MAPK during brest cancer cell proliferation and invasion. Cancer Res 67:3752–3758 Taub N, Teis D, Ebner HL, Hess MW, Huber LA, 2007, Late endosomal traffic of epidermal growth factor receptor ensures spatial and temporal fidelity of mitogen-activated protein kinase signaling. Mol Biol Cell 18:4698–4710 Zehorai E, Yao Z, Plotnikov A, Seger R, 2010, The subcellular localization of MEK and ERK-A novel nuclear translocation signal, NTS, paves a way to nucleus. Mol Cell End 314:213–220 Zuo W, Chen YG, 2009, Specific activation of mitogen-activated protein kinase by transforming growth factor-β receptors in lipid rafts is required for epithelial cell plasticity. Mol Biol Cell 20:1020–1029 Galmiche A, Fueller J, Santel A, Krohne G, Witting I, Doye A, Rolando M, Flatau G, Lemichez E, Rapp UR, 2008, Isoformspecific interaction of C_RAF with mitochondria. J Biol Chem 283:14857–14866 Adachi M, Fukuda M, Nishida E, 1999, Two co-existing mechanisms for nuclear import of MAP kinase: passive diffusion of a monomer and active transport of a dimer. EMBO J 18:5347–5358 Fukuda M, Gotoh I, Gotoh Y, Nishida E, 1996, Cytoplasmic localization of MAP kinase kinase directed by its N-terminal, leucine-rich short amino acid sequence, which acts as a nuclear export signal. J Biol Chem 271:20024–20028 Fukuda M, Gotoh Y, Nishida E, 1997, Interaction of MAP kinase with MAP kinase kinase: its possible role in the control of nucleocytoplasmic transport of MAP kinase. EMBO J 16:1901–1908 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 141 EP 148 DI 10.1007/s12253-012-9595-8 PG 8 ER PT J AU Zamanian, M Veerakumarasivam, A Syahril, A Rosli, R AF Zamanian, Mohammadreza Veerakumarasivam, Abhi Syahril, Abdullah Rosli, Rozita TI Calreticulin and Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Calreticulin; Endoplasmic reticulum; Cancer; Malignant progression; Invasion; Metastasis ID Calreticulin; Endoplasmic reticulum; Cancer; Malignant progression; Invasion; Metastasis AB Calreticulin (CRT) as a multi-functional endoplasmic reticulum protein is involved in a spectrum of cellular processes which ranges from calcium homeostasis and chaperoning to cell adhesion and finally malignant formation and progression. Previous studies have shown a contributing role for CRT in a range of different cancers. This present review will focus on the possible roles of CRT in the progression of malignant proliferation and the mechanisms involved in its contribution to cancer invasion. C1 [Zamanian, Mohammadreza] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Genetic Medicine Research Center, 43400 Selangor, Serdang, Malaysia. [Veerakumarasivam, Abhi] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Genetic Medicine Research Center, 43400 Selangor, Serdang, Malaysia. [Syahril, Abdullah] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Genetic Medicine Research Center, 43400 Selangor, Serdang, Malaysia. [Rosli, Rozita] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Genetic Medicine Research Center, 43400 Selangor, Serdang, Malaysia. RP Rosli, R (reprint author), Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Genetic Medicine Research Center, 43400 Selangor, Malaysia. EM rozita@medic.upm.edu.my;r_rosli@yahoo.com CR Michalak M et al, 1999, Calreticulin: one protein, one gene, many functions. Biochem J 344(2):281–292 Goicoechea S et al, 2002, The anti-adhesive activity of thrombospondin is mediated by the N-terminal domain of cell surface calreticulin. J Biol Chem 277(40):37219–37228 Michalak M et al, 1992, Calreticulin. Biochem J 285(3):681–692 Michalak M, Robert Parker JM, Opas M, 2002, Ca2+ signaling and calcium binding chaperones of the endoplasmic reticulum. Cell Calcium 32(5–6):269–278 Rojiani MV et al, 1991, In vitro interaction of a polypeptide homologous to human Ro/SS-A antigen, calreticulin, with a highly conserved amino acid sequence in the cytoplasmic domain of integrin alpha subunits. Biochemistry 30:9859–9866 Burns K et al, 1994, Modulation of gene expression by calreticulin binding to the glucocorticoid receptor. Nature 367(6462):476–480 Gelebart P, Opas M, Michalak M, 2005, Calreticulin, a Ca2+−binding chaperone of the endoplasmic reticulum. Int J Biochem Cell Biol 37(2):260–266 Qiu Y, Michalak M, 2009, Transcriptional control of the calreticulin gene in health and disease. Int J Biochem Cell Biol 41, 3):531–538 Gold LI et al, 2010, Calreticulin: non-endoplasmic reticulum functions in physiology and disease. FASEB J 24(3):665–683 Nanney LB et al, 2008, Calreticulin enhances porcine wound repair by diverse biological effects. Am J Pathol 173(3):610–630 Gu VY et al, 2008, Calreticulin in human pregnancy and preeclampsia. Mol Hum Reprod 14(5):309–315 Pagh R et al, 2008, The chaperone and potential mannan-binding lectin, MBL, co-receptor calreticulin interacts with MBL through the binding site for MBL-associated serine proteases. FEBS J 275, 3):515–526 Yokoyama M, Hirata K-I, 2005, New function of calreticulin: calreticulin-dependent mRNA destabilization. Circ Res 97(10):961– 963 McCauliffe DP et al, 1992, The 5’-flanking region of the human calreticulin gene shares homology with the human GRP78, GRP94, and protein disulfide isomerase promoters. J Biol Chem 267(4):2557–2562 Waser M et al, 1997, Regulation of calreticulin gene expression by calcium. J Cell Biol 138(3):547–557 Nguyen TQ, Donald Capra J, Sontheimer RD, 1996, Calreticulin is transcriptionally upregulated by heat shock, calcium and heavy metals. Mol Immunol 33(4–5):379–386 Michalak M et al, 2009, Calreticulin, a multi-process calciumbuffering chaperone of the endoplasmic reticulum. Biochem J 417(3):651–666 Qiu Y et al, 2008, Regulation of the calreticulin gene by GATA6 and Evi-1 transcription factorsa€. Biochemistry 47(12):3697– 3704 Guo L et al, 2001, COUP-TF1 antagonizes Nkx2.5-mediated activation of the calreticulin gene during cardiac development. J Biol Chem 276:2797–2801 Lynch J et al, 2005, Calreticulin signals upstream of calcineurin and MEF2C in a critical Ca2+−dependent signaling cascade. J Cell Biol 170:37–47 Vig S et al, 2012, C/EBPα mediates the transcriptional suppression of human calreticulin gene expression by TNFα. Int J Cell Biol 44(1):113–122 Opas M et al, 1991, Regulation of expression and intracellular distribution of calreticulin, a major calcium binding protein of nonmuscle cells. J Cell Physiol 149(1):160–171 Yoon G-S et al, 2000, Nuclear matrix of calreticulin in hepatocellular carcinoma. Cancer Res 60(4):1117–1120 Gold LI, et al., 2006, Overview of the role for calreticulin in the enhancement of wound healing through multiple biological effects. The journal of investigative dermatology. Symposium proceedings/ the Society for Investigative Dermatology, Inc. [and] European Society for Dermatological Research 11(1):57–65 Huang J-B et al, 2004, Identification of channels promoting calcium spikes and waves in HT1080 tumor cells. Cancer Res 64, 7):2482–2489 Pallero MA et al, 2008, Thrombospondin 1 binding to calreticulin- LRP1 signals resistance to anoikis. FASEB J 22(11):3968–3979 Kageyama K et al, 2002, Overexpression of calreticulin modulates protein kinase B/Akt signaling to promote apoptosis during cardiac differentiation of cardiomyoblast H9c2 cells. J Biol Chem 277, 22):19255–19264 Okunaga T et al, 2006, Calreticulin, a molecular chaperone in the endoplasmic reticulum, modulates radiosensitivity of human glioblastoma U251MG cells. Cancer Res 66(17):8662–8671 LimS, et al., 2008, Enhanced calreticulin expression promotes calciumdependent apoptosis in postnatal cardiomyocytes. 25:390–396 Bini L et al, 1997, Protein expression profiles in human breast ductal carcinoma and histologically normal tissue. Electrophoresis 18(15):2832–2841 Chahed K et al, 2005, Expression of fibrinogen E-fragment and fibrin E-fragment is inhibited in the human infiltrating ductal carcinoma of the breast: the two-dimensional electrophoresis and MALDI-TOFmass spectrometry analyses. Int J Oncol 27(5):1425–1431 Kageyama S et al, 2004, Identification by proteomic analysis of calreticulin as a marker for bladder cancer and evaluation of the diagnostic accuracy of its detection in urine. Clin Chem 50, 5):857–866 Ayodele A et al, 2000, Polypeptide expression in prostate hyperplasia and prostate adenocarcinoma. Anal Cell Pathol 21(1):1–9 Kim Y et al, 2004, Glucoronic acid is a novel inducer of heat shock response. Mol Cell Biochem 259(1):23–33 Hong S-H et al, 2004, An autoantibody-mediated immune response to calreticulin isoforms in pancreatic cancer. Cancer Res 64(15):5504–5510 Du X-L et al, 2007, Proteomic profiling of proteins dysregulted in Chinese esophageal squamous cell carcinoma. J Mol Med 85, 8):863–875 Nishimori T et al, 2006, Proteomic analysis of primary esophageal squamous cell carcinoma reveals downregulation of a cell adhesion protein, periplakin. Proteomics 6(3):1011–1018 Chen CN et al, 2009, Association between color doppler vascularity index, angiogenesis-related molecules, and clinical outcomes in gastric cancer. J Surg Oncol 99(7):402–408 Vougas K et al, 2008, Two-dimensional electrophoresis and immunohistochemical study of calreticulin in colorectal adenocarcinoma and mirror biopsies. J Blacan Union Oncol 13(1):101–107 White TK, Zhu Q, Tanzer ML, 1995, Cell surface calreticulin is a putative mannoside lectin which triggers mouse melanoma cell spreading. J Biol Chem 270(27):15926–15929 Dissemond J et al, 2004, Differential downregulation of endoplasmic reticulum-residing chaperones calnexin and calreticulin in human metastatic melanoma. Cancer Lett 203(2):225–231 Helbling D et al, 2005, CBFB-SMMHC is correlated with increased calreticulin expression and suppresses the granulocytic differentiation factor CEBPA in AML with inv(16). Blood 106, 4):1369–1375 Chen CN et al, 2009, Identification of calreticulin as a prognosis marker and angiogenic regulator in human gastric cancer. Ann Surg Oncol 16(2):524–533 Lwin Z-M, et al., 2010, Clinicopathological significance of calreticulin in breast invasive ductal carcinoma. Mod Pathol Eric A et al, 2009, Effects of humoral immunity and calreticulin overexpression on postoperative course in breast cancer. Pathol Oncol Res 15(1):89–90 Pabst T et al, 2001, AML1-ETO downregulates the granulocytic differentiation factor C/EBP[alpha] in t(8;21, myeloid leukemia. Nat Med 7(4):444–451 Lu Y-C et al, 2011, Changes in tumor growth and metastatic capacities of j82 human bladder cancer cells suppressed by downregulation of calreticulin expression. Am J Pathol 179:1425–1433 Hayashi E et al, 2005, Proteomic profiling for cancer progression: differential display analysis for the expression of intracellular proteins between regressive and progressive cancer cell lines. Proteomics 5(4):1024–1032 Zhu J, 1996, Ultraviolet B irradiation and cytomegalovirus infection synergize to induce the cell surface expression of 52-kD/Ro antigen. Clin Exp Immunol 1(103):47–53 Alur M et al, 2009, Suppressive roles of calreticulin in prostate cancer growth and metastasis. Am J Pathol 175(2):882–890 Porcellini S et al, 2006, Regulation of peripheral T cell activation by calreticulin. J Exp Med 203(2):461–471 Dupuis M et al, 1993, The calcium-binding protein calreticulin is a major constituent of lytic granules in cytolytic T lymphocytes. J Exp Med 177(1):1–7 Andrin C et al, 1998, Interaction between a Ca2+−binding protein calreticulin and perforin, a component of the cytotoxic T-cell granules†. Biochemistry 37(29):10386–10394 Sipione S et al, 2005, Impaired cytolytic activity in calreticulindeficient CTLs. J Immunol 174(6):3212–3219 Obeid M et al, 2007, Ecto-calreticulin in immunogenic chemotherapy. Immunol Rev 220:22–34 Obeid M et al, 2007, Leveraging the immune system during chemotherapy: moving calreticulin to the cell surface converts apoptotic death from “silent” to immunogenic. Cancer Res 67, 17):7941–7944 Clarke C, Smyth MJ, 2007, Calreticulin exposure increases cancer immunogenicity. Nat Biotechnol 25(2):192–193 Zhou P et al, 2008, Calreticulin expression in the clonal plasma cells of patients with systemic light-chain, AL-, amyloidosis is associated with response to high-dose melphalan. Blood 111(2):549–557 Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100(1):57–70 Hayashida Y et al, 2006, Calreticulin represses E-cadherin gene expression in Madin-Darby Canine kidney cells via slug. J Biol Chem 281(43):32469–32484 Fresno Vara JA et al, 2004, PI3K/Akt signalling pathway and cancer. Cancer Treat Rev 30(2):193–204 Xu Q et al, 2005, Targeting Stat3 blocks both HIF-1 and VEGF expression induced by multiple oncogenic growth signaling pathways. Oncogene 24(36):5552–5560 Yu H, Pardoll D, Jove R, 2009, STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer 9(11):798–809 Luo M-L et al, 2006, Amplification and overexpression of CTTN, EMS1, contribute to the metastasis of esophageal squamous cell carcinoma by promoting cell migration and anoikis resistance. Cancer Res 66(24):11690–11699 Weaver AM, 2008, Cortactin in tumor invasiveness. Cancer Lett 265(2):157–166 Du XL et al, 2009, Calreticulin promotes cell motility and enhances resistance to anoikis through STAT3-CTTN-Akt pathway in esophageal squamous cell carcinoma. Oncogene 28(42):3714–3722 Dedhar S et al, 1994, Inhibition of nuclear hormone receptor activity by calreticulin. Nature 367(6462):480–483 Green S et al, 1988, The N-terminal DNA-binding ‘zinc finger’ of the oestrogen and glucocorticoid receptors determines target gene specificity. EMBO J 7(10):3037–3044 Platet N et al, 2000, Unliganded and liganded estrogen receptors protect against cancer invasion via different mechanisms. Mol Endocrinol 14(7):999–1009 Lusche DF, Wessels D, Soll DR, 2009, The effects of extracellular calcium on motility, pseudopod and uropod formation, chemotaxis, and the cortical localization of myosin II in Dictyostelium discoideum. Cell Motil Cytoskeleton 66(8):567–587 Fache S et al, 2005, Calcium mobilization stimulates Dictyostelium discoideum shear-flow-induced cell motility. J Cell Sci 118(15):3445–3458 Chantome A et al, 2009, KCa2.3 channel-dependent hyperpolarization increases melanoma cell motility. Exp Cell Res 315, 20):3620–3630 Titushkin I, Cho M, 2009, Regulation of cell cytoskeleton and membrane mechanics by electric field: role of linker proteins. Biophys J 96(2):717–728 Nakamura K et al, 2001, Functional specialization of calreticulin domains. J Cell Biol 154(5):961–972 Mesaeli N et al, 1999, Calreticulin is essential for cardiac development. J Cell Biol 144(5):857–868 Camacho P, Lechleiter JD, 1995, Calreticulin inhibits repetitive intracellular Ca2+ waves. Cell 82(5):765–771 Arnaudeau S et al, 2002, Calreticulin differentially modulates calcium uptake and release in the endoplasmic reticulum and mitochondria. J Biol Chem 277(48):46696–46705 Orr AWet al, 2003, Thrombospondin signaling through the calreticulin/ LDL receptor-related protein co-complex stimulates random and directed cell migration. J Cell Sci 116(14):2917–2927 Li SS, Forslow A, Sundqvist K-G, 2005, Autocrine regulation of T cell motility by calreticulin-thrombospondin-1 interaction. J Immunol 174(2):654–661 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 149 EP 154 DI 10.1007/s12253-012-9600-2 PG 6 ER PT J AU Singh, KA Pandey, A Tewari, M Kumar, R Sharma, A Pandey, PH Shukla, ShH AF Singh, Kumar Alok Pandey, Anshuman Tewari, Mallika Kumar, Rajiv Sharma, Anjana Pandey, P H Shukla, Shankar Hari TI Prospects of Nano–Material in Breast Cancer Management SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Nano materials; Breast cancer; Biomarkers; Diagnosis ID Nano materials; Breast cancer; Biomarkers; Diagnosis AB Breast cancer evaluation and early diagnosis are core complexity worldwide and an ambiguity for scientists till date. Nano-materials are innovative tools for rapid diagnosis and therapy, which may induce an immense result in the field of oncology. Their exceptional size-dependent properties make them special and superior materials and quite indispensable in several fields of the human activities. The major obstacle in finding cure for malignant breast cancer is to increase in development of resistances for tumors to the therapeutic treatments. The widespread mammo-graph particle is being developed by nations to diagnosis disease in primitive stage to decline the mortality rates caused by breast carcinoma. The advancement of nano-particle based diagnostic tools facilitates in evaluation and provides encouraging development in breast cancer therapeutics. In this compact review, efforts have been made to compose the current advancements in the area of functional nano-particles. Furthermore, in vivo and in vitro applications of nano-materials in breast cancer management are also discussed. C1 [Singh, Kumar Alok] Banaras Hindu University, Institute of Medical Sciences, Department of Surgical Oncology, 221005 Varanasi, India. [Pandey, Anshuman] Banaras Hindu University, Institute of Medical Sciences, Department of Surgical Oncology, 221005 Varanasi, India. [Tewari, Mallika] Banaras Hindu University, Institute of Medical Sciences, Department of Surgical Oncology, 221005 Varanasi, India. [Kumar, Rajiv] Amity University, Amity Institute of Biotechnology, Lucknow CampusLucknow, UP, India. [Sharma, Anjana] Amity University, Amity Institute of Biotechnology, Lucknow CampusLucknow, UP, India. [Pandey, P H] Banaras Hindu University, Faculty of Science, Department of BiochemistryVaranasi, India. [Shukla, Shankar Hari] Banaras Hindu University, Institute of Medical Sciences, Department of Surgical Oncology, 221005 Varanasi, India. RP Shukla, ShH (reprint author), Banaras Hindu University, Institute of Medical Sciences, Department of Surgical Oncology, 221005 Varanasi, India. EM wfsos1@gmail.com;harishukla@usa.net CR American Cancer Society, 2011, Breast cancer facts and figures 2011–2012. American Cancer Society, Inc., Atlanta Sasco AJ, 2001, Epidemiology of breast cancer: an environmental disease? APMIS 109(5):321–332 Singh AK, Pandey A, Tewari M, Pandey HP, Shukla HS, 2011, Epigenetic silencing of BRCA1 gene associated with demographic and pathologic factors in sporadic breast cancert: a study of Indian population. Eur J Cancer Prev., DOI 10.1097/CEJ.0b013e32834761a6 Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 2010 Howell A, 2010, The emerging breast cancer epidemic: early diagnosis and treatment. Breast Cancer Res 12(Suppl 4):S10 James PB, 2001, Current and future technologies for breast cancer imaging. Breast Cancer Res 3:14–16 Pandey A, Singh AK, Maurya SK, Rai R, Shukla HS, 2008, Nanoscience and their biological importance: human health and disease. Dig J Nanomater Biostruct 3(3):141–146 Yezhelyev M, Morris C, Gao X, Nie S, Lewis M, Cohen C et al, 2005, Multiple profiling of human breast cancer cell lines with quantum dots–Ab conjugates. Proc Am Assoc Cancer Res 46:510 Singh AK, Pandey A, Rai R, Tewari M, Pandey HP, Shukla HS, 2008, Nanomaterials as emerging tool in cancer diagnosis and treatment. Dig J Nanomater Biostruct 3(3):135–140 Gao XH, Cui YY, Levenson RM, Chung LWK, Nie SM, 2004, In vivo cancer targeting and imaging with semiconductor quantum dots. Nat Biotechnol 22:969–976 Hallahan D, Geng L, Qu S, Scarfone C, Giorgio T, Donnelly E et al, 2003, Integrin-mediated targeting of drug delivery to irradiated tumor blood vessels. Cancer Cell 3:63–74 Liberman A, Martinez HP, Ta CN, Barback CV, Mattrey RF, Kono Y, Blair SL, Trogler WC, Kummel AC, Wu Z, 2012, Hollow silica and silica-boron nano/microparticles for contrast-enhanced ultrasound to detect small tumors. Biomaterials 33, 20):5124–5129 Qiao G, Gao Y, Li N, Yu Z, Zhuo L, Tang B, 2011, Simultaneous detection of intracellular tumor mRNA with bi-color imaging based on a gold nanoparticle/molecular beacon. Chemistry 17, 40):11210–11215 Salata OV, 2004, Applications of nanoparticles in biology and medicine. J Nanobiotechnol 2:3 Tomalia DA, 2009, In quest of a systematic framework for unifying and defining nanoscience. J Nanopart Res 11(6):1251– 1310 Park JW, 2002, Liposome-based drug delivery in breast cancer treatment. Breast Cancer Res 4:95–99 Madani SY, Naderi N, Dissanayake O, Tan A, Seifalian AM, 2011, A new era of cancer treatment: carbon nanotubes as drug delivery tools. Int J Nanomedicine 6:2963–2979 Bianco A, Kostarelos K, Prato M, 2005, Applications of carbon nanotubes in drug delivery. Curr Opin Chem Biol 9:674–679 Duncan R, 2003, The dawning era of polymer therapeutics. Nat Rev Drug Discov 2:347–360 Lee CC, Mackay JA, Frechet JMJ, Szoka FC, 2005, Designing dendrimers for biological application. Nat Biotechnol 23:1517– 1526 Svenson S, Tomalia DA, 2005, Dendrimers in biomedical applications: reflections on the field. Adv Drug Deliv Rev 57:2106– 2129 Tasis D, Tagmatarchis N, Bianco A, Prato M, 2006, Chemistry of carbon nanotubes. Chem Rev 106:1105–1136 Artemov D, Mori N, Okollie B et al, 2003, MR molecular imaging of the Her-2/neu receptor in breast cancer cells using targeted iron oxide nanoparticles. Magn Reson Med 49:403–408 Chan P, Yuen T, Ruf F, Gonzalez-Maeso J, Sealfon SC, 2005, Method for multiplex cellular detection of mRNAs using quantum dot fl uorescent in situ hybridization. Nucleic Acids Res 33:161 Gao X,Yang L, Petros JAet al, 2005, In vivomolecular and cellular imaging with quantum dots. Curr Opin Biotechnol 16:63–72 Medintz IL, Uyeda HT, Goldman ER, Mattoussi H, 2005, Quantum dot bioconjugates for imaging, labelling and sensing. Nat Mater 4:435–446 Burtea C, Ballet S, Laurent S, Rousseaux O, Dencausse A, Gonzalez W et al, 2012, Development of a magnetic resonance imaging protocol for the characterization of atherosclerotic plaque by using vascular cell adhesion molecule-1 and apoptosis-targeted ultrasmall superparamagnetic iron oxide derivatives. Arterioscler Thromb Vasc Biol 32(6):e36–e48 Perez OD, Nolan GP, 2002, Simultaneous measurement of multiple active kinase states using polychromatic flow cytometry. Nat Biotechnol 20:155–162 Xing J, Zeng J, Yang J, Kong T, Xu T, Roa Wet al, 2007, Goldbased nanoparticles for breast cancer diagnosis and treatment. In ISCAS: 2882–2885 Wang J, O’Toole M, Massey A, Biswas S, Nantz M, Achilefu S et al, 2011, Highly specific, NIR fluorescent contrast agent with emission controlled by gold nanoparticle. Adv Exp Med Biol 915:149–154 El-Sayed I, Huang X, Macheret F, Humstoe JO, Kramer R, El- Sayed M, 2007, Effect of plasmonic gold nanoparticles on benign and malignant cellular autofluorescence: a novel probe for fluorescence based detection of cancer. Technol Cancer Res Treat 6(5):403–412 Kumar A, Boruah BM, Liang X, 2011, Gold nanoparticles: promising nanomaterials for the diagnosis of cancer and HIV/ AIDS. J Nanomater., DOI 10.1155/2011/202187 Chen J, Wang D, Xi J, Au L, Siekkinen A, Warsen A et al, 2007, Immuno gold nanocages with tailored optical properties for targeted photothermal destruction of cancer cells. Nano Lett 7:1318–1322 Au L, Zheng D, Zhou F, Li ZY, Li X, Xia Y, 2008, A quantitative study on the photothermal effect of immuno gold nanocages targeted to breast cancer cells. ACS Nano 2(8):1645–1652 Zhou HS, Honma I, Komiyama H, Haus JW, 1994, Controlled synthesis and quantum-size effect in gold-coated nanoparticles. Phys Rev B 50(16):12052–12056 Gobin AM, Watkins EM, Quevedo E, Colvin VL, West JL, 2010, Nearinfrared- resonant gold/gold sulfide nanoparticles as a photothermal cancer therapeutic agent. Small 6(6):745–752 Day ES, Bickford LR, Slater JH, Riggall NS, Drezek RA, West JL, 2010, Antibody-conjugated gold-gold sulfide nanoparticles as multifunctional agents for imaging and therapy of breast cancer. Int J Nanomedicine 5:445–454 Lu W, Arumugam SR, Senapati D, Singh AK, Arbneshi T, Khan SA et al, 2010, Multifunctional oval-shaped gold-nanoparticlebased selective detection of breast cancer cells using simple colorimetric and highly sensitive two-photon scattering assay. ACS Nano 4(3):1739–1749 Dreaden EC, Mwakwari SC, Sodji QH, Oyelere AK, El-Sayed MA, 2009, Tamoxifen-poly(ethylene glycol)-thiol gold nanoparticle conjugates: enhanced potency and selective delivery for breast cancer treatment. Bioconjug Chem 20(12):2247–2253 Sitharaman B,Wilson L, 2007)Gadofullerenes and gadonanotubes: a new paradigm for high-performance magnetic resonance imaging contrast agent probes. J Biomed Nanotechnol 3(4):342–352 Liu Z, Cai W, He L, Nakayama N, Chen K, Sun X, 2007, In vivo biodistribution and highly efficient tumour targeting of carbon nanotubes in mice. Nat Nanotechnol 2(1):47–52 De La Zerda A, Zavaleta C, Keren S, Vaithilingam S, Bodapati S, Liu Z et al, 2008, Carbon nanotubes as photoacoustic molecular imaging agents in living mice. Nat Nanotechnol 3:557–562 Kievit FM, Zhang M, 2011, Cancer nanotheranostics: improving imaging and therapy by targeted delivery across biological barriers. Adv Mater 23(36):H217–H247 Beg S, Rizwan M, Sheikh AM, Hasnain MS, Anwer K, Kohli K, 2011, Advancement in carbon nanotubes: basics, biomedical applications and toxicity. J Pharm Pharmacol 63(2):141–163 Bhirde AA, Patel V, Gavard J et al, 2009, Targeted killing of cancer cells in vivo and in vitro with EGF-directed carbon nanotube-based drug delivery. ACS Nano 3(2):307–316 Shea JD, Kosmas P, Hagness SC, Van Veen BD, 2009, Contrastenhanced microwave breast imaging. 13th International Symposium on Antenna Technology and Applied Electromagnetics, ANTEM, and the Canadian Radio Science Meeting, ANTEM/URSI); Banff, Canada, ISBN: 978-1-4244-2980-6 Shea JD, Kosmas P, Hagness SC, Van Veen BD, 2010, Contrastenhanced microwave imaging of breast tumors. Inverse Probl 26., DOI 10.1088/0266-5611/26/7/074009 Mashal A, Sitharaman B, Li X, Avti PK, Sahakian AV, Booske JH, 2010, Toward carbon-nanotube-based theranostic agents for microwave detection and treatment of breast cancer: enhanced dielectric and heating response of tissue-mimicking materials. IEEE Trans Biomed Eng 57(8):1831–1834 Rodriguez-Luccioni HL, Latorre-Esteves M, Mendez-Vega J, Soto O, Rodriguez AR, Rinaldi C et al, 2011, Enhanced reduction in cell viability by hyperthermia induced by magnetic nanoparticles. Int J Nanomedicine 6:373–380 Ito A, Shinkai M, Honda H, Kobayashi T, 2005, Medical application of functionalized magnetic nanoparticles. J Biosci Bioeng 100(1):1–11 Will O, Purkayastha S, Chan C, Athanasiou T, Darzi AW, Gedroyc W et al, 2005, Diagnostic precision of nanoparticleenhanced MRI for lymph-node metastases: a meta-analysis. Lancet Oncol 7:52–60 Tran LA, Wilson LJ, 2011, Nanomedicine: making controllable magnetic drug delivery possible for the treatment of breast cancer. Breast Cancer Res 13(2):303 Gillies ER, Frechet JM, 2005, Dendrimers and dendritic polymers in drug delivery. Drug Discov Today 10:35–43 Svenson S, 2009, Dendrimers as versatile platform in drug delivery applications. Eur J Pharm Biopharm 71:445–462 Kono K, Liu M, Frechet JM, 1999, Design of dendritic macromolecules containing folate or methotrexate residues. Bioconjug Chem 10:1115–1121 Franc G, Kakkar AK, 2009, Diels–Alder “click” chemistry in designing dendritic macromolecules. Chem Eur J 15:5630–5639., DOI 10.1002/chem.200900252 Quintana A, Raczka E, Piehler L, Lee I, Myc A, Majoros I et al, 2002, Design and function of a dendrimerbased therapeutic nanodevice targeted to tumor cells through the folate receptor. Pharm Res 19:1310–1316 Patri AK, Myc A, Beals J, Thomas TP, Bander NH, Baker JR, 2004, Synthesis and in vitro testing of J591 antibody-dendrimer conjugates for targeted prostate cancer therapy. Bioconjug Chem 15:1174–1181 Shukla R, Thomas TP, Peters J, Kotlyar A, Myc A, Baker JR, 2005, Tumor angiogenic vasculature targeting with PAMAM dendrimer–RGD conjugates. Chem Commun 46:5739–5741 Shukla R, Thomas TP, Peters JL, Desai AM, Kukowska-Latallo J, Patri AK et al, 2006, HER2 specific tumor targeting with dendrimer conjugated anti-HER2 mAb. Bioconjug Chem 17:1109–1115 Hirsjarvi S, Passirani C, Benoit JP, 2011, Passive and active tumour targeting with nanocarriers. Curr Drug Discov Technol 8(3):188–196 Huynh NT, Roger E, Lautram N, Benoit JP, Passirani C, 2010, The rise and rise of stealth nanocarriers for cancer therapy: passive versus active targeting. Nanomedicine, Lond, 5(9):1415–1433 Krukemeyer MG, Krenn V, Jakobs M, Wagner W, 2012, Magnetic drug targeting in a rhabdomyosarcoma rat model using magnetite-dextran composite nanoparticle-bound mitoxantrone and 0.6 tesla extracorporeal magnets - sarcoma treatment in progress. J Drug Target 20(2):185–193 Torchilin VP, 2010, Passive and active drug targeting: drug delivery to tumors as an example. Handb Exp Pharmacol, 197): 3–53., DOI 10.1007/978-3-642-00477-3_1 Myc A, Kukowska-Latallo J, Cao P, Swanson B, Battista J, Dunham T, Baker JR, 2010, Targeting the efficacy of a dendrimer-based nanotherapeutic in heterogeneous xenograft tumors in vivo. Anticancer Drugs 21(2):186 Leatherdale CA, Woo WK, Mikulec FV, Bawendi MG, 2002, On the absorption cross section of CdSe nanocrystal quantum dots. J Phys Chem B 106:7619–7622 Wu XY, Liu HJ, Liu JQ, Haley KN, Treadway JA, Larson JP et al, 2003, Immunofluorescent labeling of cancer marker Her2 and other cellular targets with semiconductor quantum dots. Nat Biotechnol 21:41–46 Nisman R, Dellaire G, Ren Y, Li R, Bazett-Jones DP, 2004, Application of quantum dots as probes for correlative fluorescence, conventional and energy-filtered transmission electron microscopy. J Histochem Cytochem 52:13–18 Dubertret B, Skourides P, Norris DJ, Noireaux V, Brivanlou AH, Libchaber A, 2002, In vivo imaging of quantum dots encapsulated in phospholipid micelles. Science 298:1759–1762 Voura EB, Jaiswal JK, Mattoussi H, Simon SM, 2004, Tracking metastatic tumor cell extravasation with quantum dot nanocrystals and fluorescence emission-scanning microscopy. Nat Med 10:993–998 Lewin M, Carlesso N, Tung CH, Tang XW, Cory D, Scadden DT et al, 2000, Tat peptide-derivatized magnetic nanoparticles allow in vivo tracking and recovery of progenitor cells. Nat Biotechnol 18:410–414 Akerman ME, Chan WCW, Laakkonen P, Bhatia SN, Ruoslahti E, 2002, Nanocrystal targeting in vivo. Proc Natl Acad Sci USA 99:12617–12621 Chen C, Sun SR, Gong YP, Qi CB, Peng CW, Yang XQ, Liu SP, Peng J, Zhu S, Hu MB, Pang DW, Li Y, 2011, Quantum dotsbased molecular classification of breast cancer by quantitative spectroanalysis of hormone receptors and HER2. Biomaterials 32, 30):7592–7599 Mattoussi H, Mauro JM, Goldman ER, Anderson GP, Sundar VC, Mikulec FV et al, 2000, Self-assembly of CdSe-ZnS quantum dot bioconjugates using an engineered recombinant protein. J Am Chem Soc 122:12142–12150 Gao XH, Chan WCW, Nie SM, 2002, Quantum-dot nanocrystals for ultrasensitive biological labeling and multicolor optical encoding. J Biomed Opt 7:532–537 Liu G, Swierczewska M, Lee S, Chena X, 2010, Functional nanoparticles for molecular imaging guided gene delivery. Nano Today 5:524–539 Chang SS, Reuter VE, HestonWDW, Gaudin PB, 2001)Metastatic renal cell carcinoma neovasculature expresses prostate-specific membrane antigen. Urology 57:801–805 Jaiswal JK, Simon SM, 2004, Potentials and pitfalls of fluorescent quantum dots for biological imaging. Trends Cell Biol 14:9 Agrawal A, Deo R, Wang GD, Wang MD, Nie S, 2008, Nanometer-scale mapping and single-molecule detection with color-coded nanoparticle probes. PNAS 105(9):3298–3303 Mankoff DA, 2008, Molecular imaging as a tool for translating breast cancer science. Breast Cancer Res 10(Suppl 1):S3., DOI 10.1186/bcr2126 Doubrovin M, Serganova I, Mayer-Kuckuk P, Ponomarev V, Blasberg RG, 2004, Multimodality in vivo molecular genetic imaging. Bioconjug Chem 15:1376–1388 Hay BP, Werner EJ, Raymond KN, 2004, Estimating the number of bound waters in Gd(III, complexes revisited. Improved methods for the prediction of q-values. Bioconjug Chem 15:1496–1502 Caravan P, Ellison JJ, McMurry TJ, Lauffer RB, 1999, Gadolinium, III, chelates as MRI contrast agents: structure, dynamics, and applications. Chem Rev 99:2293–2352 Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R, 2007, Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol 2:751–760 Alexis F, Rhee JW, Richie JP, Radovic-Moreno AF, Langer R, Farokhzad OC, 2008, New frontiers in nanotechnology for cancer treatment. Urol Oncol 26:74–85 Sofou S, 2007, Surface-active liposomes for targeted cancer therapy. Nanomedicine, Lond, 2(5):711–724 Noble CO, Kirpotin DB, Hayes ME, Mamot C, Hong K, Park JW et al, 2004, Development of ligand-targeted liposomes for cancer therapy. Expert Opin Ther Targets 8:335–353 Wang T, D’Souza GGM, Bedi D, Fagbohun OA, Potturi LP, Papahadjopoulos-Sternberg B et al, 2010, Enhanced binding and killing of target tumor cells by drugloadedliposomes modified with tumor-specific phage fusioncoat protein. Nanomedicine, Lond, 5(4):563–574 Morgan TT, Muddana HS, Altinoglu EI, Rouse SM, Tabakovic A, Tabouillot T et al, 2008, Encapsulation of organic molecules in calcium phosphate nanocomposite particles for intracellular imaging and drug delivery. Nano Lett 8(12):4108–4115 Muddana HS, Morgan TT, Adair JH, Butler PJ, 2009, Photophysics of Cy3-encapsulated calcium phosphate nanoparticles. Nano Lett 4:1559–1566 Barth BM, Sharma R, Altinoglu EI, Morgan TT, Shanmugavelandy SS, Kaiser JM et al, 2010, Bioconjugation of calcium phosphosilicate composite nanoparticles for selective targeting of human breast and pancreatic cancers in vivo. ACS Nano 4(3):1279–1287 Hung O, 2006, Drug transformation: advances in drug delivery systems. Can J Anaesth 53(11):1074–1077 Jiang W, Kim BY, Rutka JT, Chan WC, 2007, Advances and challenges of nanotechnology-based drug delivery systems. Expert Opin Drug Deliv 4(6):621–633 Sebak S, Mirzaei M, Malhotra M, Kulamarva A, Prakash S, 2010, Human serum albumin nanoparticles as an efficient noscapine drug delivery system for potential use in breast cancer: preparation and in vitro analysis. Int J Nanomedicine 20(5):525– 532 DiMasi JA, Hansen RW, Grabowski HG, 2003, The price of innovation: new estimates of drug development costs. J Health Econ 22:151–185 Marchetti S, Schellens JH, 2007, The impact of FDA and EMEA guidelines on drug development in relation to Phase 0 trials. Br J Cancer 97:577–581 Ferrari M, 2005, Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer 5:161–171 Ferrari M, 2005, Nanovector therapeutics. Curr Opin Chem Biol 9:343–346 Ferrari M, Downing G, 2005)Medical nanotechnology: shortening clinical trials and regulatory pathways? BioDrugs 19:203–210 Lu C, Stewart DJ, Lee JJ, Ji L, Ramesh R et al, 2012, Phase I clinical trial of systemically administered TUSC2(FUS1)- nanoparticles mediating functional gene transfer in humans. PLoS One 7(4):e34833., DOI 10.1371/journal.pone.0034833 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 155 EP 165 DI 10.1007/s12253-013-9609-1 PG 11 ER PT J AU Aiad, AH Bashandy, AM Abdou, GA Zahran, AA AF Aiad, A Hayam Bashandy, A Manar Abdou, Gaber Asmaa Zahran, A Ahmad TI Significance of AgNORs and Ki-67 Proliferative Markers in Differential Diagnosis of Thyroid Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ag NORs; Ki 67 labeling index; Follicular carcinoma; Follicular adenoma; Papillary carcinoma ID Ag NORs; Ki 67 labeling index; Follicular carcinoma; Follicular adenoma; Papillary carcinoma AB We aimed to assess the utility of quantitative analysis of AgNORs and Ki67 labeling index (LI) in the differential diagnosis of different thyroid lesions. This study included: 25 papillary carcinomas, 7 follicular carcinomas, 21 follicular adenomas and 27 nodular goiters. Using a semiautomatic image analysis system, Ag NORs parameters were measured and calculated including: total area of AgNORs, mean Ag NOR number in nuclei, nuclear area, mean area of AgNOR dots per each nucleus, number of central and marginal AgNOR dots, and the relative ratio of total area of AgNOR dots/total area of nucleus. Ki67 immunostaining was performed and the LI was determined. There was a significant difference between groups of thyroid lesions regarding total area of AgNORs, Ag NOR number and number of marginal Ag NOR dots. According to receiver operating characteristic curve, Ag NORs number 02.91 and marginal Ag NORs02.67 were useful cut off values above which follicular carcinoma can be diagnosed with 100 % sensitivity, 79 % specificity, 76 % PPV, 100 % NPV and 85 % diagnostic accuracy for both parameters. Mean Ki67 LI in our study was 14.12±2.29, 61.42±3.77, 34.90±3.49 and 18.60±1.96 for papillary carcinoma, follicular carcinoma, follicular adenoma and nodular goiter respectively. Ki67 LI showed statistically significant difference between follicular carcinoma and follicular adenoma (p<0.026) and between papillary carcinoma and follicular adenoma (p<0.007). Quantification of Ag NORs and Ki67 LI could be used as helpful ancillary methods in the differentiation between different thyroid lesions. C1 [Aiad, A Hayam] Menoufiya University, Faculty of Medicine, Department of PathologyShebein Elkom, Egypt. [Bashandy, A Manar] Menoufiya University, Faculty of Medicine, Department of AnatomyShebein Elkom, Egypt. [Abdou, Gaber Asmaa] Menoufiya University, Faculty of Medicine, Department of PathologyShebein Elkom, Egypt. [Zahran, A Ahmad] Shebin El-Kom Educational Hospital, ENT DepartmentShebein Elkom, Egypt. RP Aiad, AH (reprint author), Menoufiya University, Faculty of Medicine, Department of Pathology, Shebein Elkom, Egypt. EM hayamaiad@yahoo.com CR Baloch ZW, LiVolsi VA, 2002, Follicular-patterned lesions of the thyroid: the bane of the pathologist. Am J Clin Pathol 117(1):143– 150 Rosai J, 2004, Thyroid gland. In: Rosai J, ed, Rosai and Ackerman’s surgical pathology, volume, 2), Ch, 9), 9th edn. Mosby, Edinburgh, pp 515–594 Mase T, Funahashi H, Koshikawa T, Imai T, Nara Y, Tanaka Y, Nakao A, 2003, HBME-1 immunostaining in thyroid tumors especially in follicular neoplasm. Endocr J 50(2):173–177 Rezk S, Brynes RK, Nelson V, Thein M, Patwardhan N, Fischer A, Khan A, 2004, beta-Catenin expression in thyroid follicular lesions: potential role in nuclear envelope changes in papillary carcinomas. Endocr Pathol 15(4):329–337 Aiad HA, Kandil MA, Asaad NY, El-Kased AM, El-Goday SF, 2008, Galectin-3 immunostaining in cytological and histopathological diagnosis of thyroid lesions. J Egypt Natl Canc Inst 20, 1):36–46 Rivera M, Ricarte-Filho J, Patel S, Tuttle M, Shaha A, Shah JP, Fagin JA, Ghossein RA, 2010, Encapsulated thyroid tumors of follicular cell origin with high grade features, high mitotic rate/ tumor necrosis): a clinicopathologic and molecular study. Hum Pathol 41(2):172–180 Volante M, Collini P, Nikiforov YE, Sakamoto A, Kakudo K, Katoh R, Lloyd RV, LiVolsi VA, Papotti M, Sobrinho-Simoes M, Bussolati G, Rosai J, 2007, Poorly differentiated thyroid carcinoma: the Turin proposal for the use of uniform diagnostic criteria and an algorithmic diagnostic approach. Am J Surg Pathol 31, 8):1256–1264 Sofiadis A, Tani E, Foukakis T, Kjellman P, Skoog L, Hoog A, Wallin G, Zedenius J, Larsson C, 2009, Diagnostic and prognostic potential of MIB-1 proliferation index in thyroid fine needle aspiration biopsy. Int J Oncol 35(2):369–374 Slowinska-Klencka D, Klencki M, Popowicz B, Sporny S, Lewinski A, 2004, Multiparameter analysis of AgNOR in thyroid lesions: comparison with PCNA expression. Histol Histopathol 19(3):785– 792 Mehrotra A, Goel MM, Singh K, 2002, Ki-67 and AgNOR proliferative markers as diagnostic adjuncts to fine needle aspiration cytology of thyroid follicular lesions. Anal Quant Cytol Histol 24, 4):205–211 Howell WM, 1982, Selective staining of nucleolus organiser regions, NORs). In: Busch H, Rothblum L, eds, The cell nucleus. Vol IX. Academic, New York, pp 89–143 Ploton D, Menager M, Jeannesson P, Himber G, Pigeon F, Adnet JJ, 1986, Improvement in the staining and in the visualization of the argyrophilic proteins of the nucleolar organizer region at the optical level. Histochem J 18(1):5–14 Bukhari MH, Niazi S, Khan SA, Hashmi I, Perveen S, Qureshi SS, Chaudhry NA, Qureshi GR, Hasan M, 2007, Modified method of AgNOR staining for tissue and interpretation in histopathology. Int J Exp Pathol 88(1):47–53 Canet V, Montmasson MP, Usson Y, Giroud F, Brugal G, 2001, Correlation between silver-stained nucleolar organizer region area and cell cycle time. Cytometry 43(2):110–116 Kanematsu E, Matsui H, Deguchi T, Yamamoto O, Korematsu M, Kobayashi A, Nezasa S, Yamamoto N, Takeuchi T, Tanaka T, Kawada Y, 1997, Signifcance of AgNOR counts for distinguishing carcinoma from adenoma and hyperplasia in parathyroid gland. Hum Pathol 28(4):421–427 Augustynowicz A, Dzieciol J, Barwijuk-Machala M, Dadan J, Puchalski Z, Sulkowski S, 2004, Assessment of proliferative activity of thyroid Hurthle cell tumors using PCNA, Ki-67 and AgNOR methods. Folia Histochem Cytobiol 42(3):165–168 Kummoona R, Jabbar A, Kareem Al-Rahal D, 2008, Proliferative activity in oral carcinomas studied with Ag-NOR and electron microscopy. Ultrastruct Pathol 32(4):139–146 Gerdes J, Lemke H, Baisch H, Wacker HH, Schwab U, Stein H, 1984, Cell cycle analysis of a cell proliferation associated human nuclear antigen defined by the monoclonal antibody Ki-67. J Immunol 133:1710–1715 Kjellman P, Zedenius J, Lundell G, Backdahl M, Farnebo LO, Hamberger B, Larsson C, Wallin G, 2006, Predictors of outcome in patients with papillary thyroid carcinoma. Eur J Surg Oncol 32, 3):345–352 Ito Y, Miyauchi A, Kakudo K, Hirokawa M, Kobayashi K, Miya A, 2010, Prognostic significance of ki-67 labeling index in papillary thyroid carcinoma. World J Surg 34(12):3015–3021 Mar KC, Eimoto T, Nagaya S, Tateyama H, 2006, Cell proliferation marker MCM2, but not Ki67, is helpful for distinguishing between minimally invasive follicular carcinoma and follicular adenoma of the thyroid. Histopathology 48:801–807 Mehrotra P, Gonzalez MA, Johnson SJ, Coleman N, Wilson JA, Davies BR, Lennard TW, 2006, Mcm-2 and Ki-67 have limited potential in preoperative diagnosis of thyroid malignancy. Laryngoscope 116:1434–1438 Krolicka A, Kobierzycki C, Pula B, Podhorska-Okolow M, Piotrowska A, Rzeszutko M, Rzeszutko W, Rabczynski J, Domoslawski P, Wojtczak B, Dawiskiba J, Dziegiel P, 2010, Comparison of metallothionein, MT, and Ki-67 antigen expression in benign and malignant thyroid tumours. Anticancer Res 30(12):4945–4949 Heikkila A, Siironen P, Hagstrom J, Heiskanen I, Sankila R, Louhimo J, Haglund C, Arola J, 2010, Follicular thyroid neoplasm: clinicopathologic features suggesting malignancy. APMIS 118(11):846–854 Pujani M, Arora B, Pujani M, Singh SK, Tejwani N, 2010, Role of Ki-67 as a proliferative marker in lesions of thyroid. Indian J Cancer 47(3):304–307 Delellis RA, Lloyd RV, Heitz PU, Eng C, eds,, 2004, World Health Organization classification of tumors. Pathology and genetics of tumors of endocrine organs. IARC Press, Lyon Aubele M, Biesterfeld S, Derenzini M, Hufnagl P, Martin H, Ofner D, Ploton D, Ruschoff J, 1994, Guidelines of AgNOR quantitation. Zentralbl Pathol 140:107–108 Hufnagl P, Guski H, Schulz HJ, 1994, Measuring of AgNORs using image analysis. Zentralbl Pathol 140(1):31–35 Slowinska-Klencka D, Klencki M, Popowicz B, Lewinski A, 2003, AgNOR quantification in the diagnosis of follicular pattern thyroid lesions. Anal Quant Cytol Histol 25(6):347–352 Solymosi T, Toth V, Sapi Z, Bodo M, Gal I, Csanate L, 1996, Diagnostic value of AgNORs method in thyroid cytopathology: correlation with morphometric measurements. Diagn Cytopathol 14:140–144 Asorta S, Sharma J, 2008, Role of AgNors in thyroid lesions on fine needle aspiration cytology smears. J Cytol 25:18–22 Cornianu M, Milos L, Golu L, Taban S, Milos A, 2006, Proliferative activity of thyroid hurthle cell tumors. Acta Endocrinologica, Buc, 3:269–282 Lewy-Trenda I, Janezukowics J, Wierzchniewska L, 2006, Practical application of proliferation markers, MIB-I, PCNA, AgNOR, expression analysis of differential diagnostics of nodular thyroid lesions. Wiad Lek 59(1-2):32–37, abstract Ciobanu D, Caruntu ID, Vulpoi C, Florea N, Giusca SE, 2006, Morphometric parameters and silver stain used in diagnosis of thyroid follicular diseases. Rom J Morphol Embryol 47(4):323–330 Ruschoff J, Prasser C, Cortez T, Hohne HM, Hohenberger W, Hofstadter F, 1993, Diagnostic value of AgNOR staining in follicular cell neoplasms of the thyroid: comparison of evaluation methods and nucleolar features. Am J Surg Pathol 17(12):1281– 1288 Hepburn PJ, Glynne-Jones E, Goddard L, Gee JM, Harper ME, 1995, Cell proliferation in prostatic carcinoma: comparative analysis of Ki-67, MIB-1 and PCNA. Histochem J 27(3):196–203 Ronckers C, Ron E, 2006, Thyroid cancer. In, Freedman LS, Edwards BK, Ries LAG, Young JL, eds). Cancer Incidence in Four Member Countries, Cyprus, Egypt, Israel, and Jordan, of the Middle East Cancer Consortium, MECC, Compared with US SEER. National Cancer Institute. NIH Pub. No. 06-5873. Bethesda, MD, pp 121–130 Erickson LA, Jin L, Wollan PC, Thompson GB, van Heerden J, Lloyd RV, 1998, Expression of p27/kip1 and Ki-67 in benign and malignant thyroid tumors. Mod Pathol 11(2):169–174 Erickson LA, Jin L, Goellner JR, Lohse C, Pankratz VS, Zukerberg LR, Thompson GB, van Heerden JA, Grant CS, Lloyd R, 2000, Pathologic features, proliferative activity, and cyclin D1 expression in Hurthle cell neoplasms of the thyroid. Mod Pathol 13(2):186–192 Horii A, Yoshida J, Sakai M, Okamoto S, Honjo Y, Mitani K, Hattori K, Kubo T, 1999, Ki-67 positive fractions in benign and malignant thyroid tumours: application of flow cytometry. Acta Otolaryngol 119(5):617–620 Rickert D, Mittermayer C, Lindenfelser R, Biesterfeld S, 2000, MIB-1 immunohistometry of follicular adenoma and follicular carcinoma of the thyroid gland. Anal Quant Cytol Histol 22, 3):229–234 Erickson LA, Yousef OM, Jin L, Lohse CM, Pankratz VS, Lloyd RV, 2000, p27kip1 expression distinguishes papillary hyperplasia in Graves’ disease from papillary thyroid carcinoma. Mod Pathol 13(9):1014–1019 Zidan J, Karen D, Stein M, Rosenblatt E, Basher W, Kuten A, 2003, Pure versus follicular variant of papillary thyroid carcinoma: clinical features, prognostic factors, treatment and survival. Cancer 97(5):1181–1185 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 167 EP 175 DI 10.1007/s12253-012-9565-1 PG 9 ER PT J AU Youlin, K Jianwei, Z Xin, G Li, Z Xiaodong, W Xiuheng, L Hengchen, Z Zhiyuan, Ch AF Youlin, Kuang Jianwei, Zhang Xin, Gou Li, Zhang Xiaodong, Weng Xiuheng, Liu Hengchen, Zhu Zhiyuan, Chen TI 4-1BB Protects Dendritic Cells from Prostate Cancer-Induced Apoptosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Dendritic cells; Co-stimulatory molecules; 4-1BB; Prostate cancer ID Dendritic cells; Co-stimulatory molecules; 4-1BB; Prostate cancer AB It has been shown that human prostate cancer (PCa) cells induced apoptotic death of the most potent antigen-presenting cells, dendritic cells (DCs), which are responsible for the induction of specific antitumor immune responses. Here, we investigated the function of 4-1BB on protecting DCs from prostate cancer-induced apoptosis with an agonistic mAb to 4-1BB. RM-1 cells and DCs were coincubated for 48 h and DC apoptosis was assessed by Annexin Vassay. TNF-α and IL-12 production were assessed by enzyme-linked immunosorbent assay (ELISA) and Bcl-2 and Bcl-xL on DCs were analyzed by Western blot. We have shown that co-incubation of RM-1 cells with DCs is accompanied by an increased level of DCs apoptosis. Triggering 4-1BB on DCs resulted in increased resistance of DCs to RM-1 cells-induced apoptosis, which was owing to the up-regulated expression of Bcl-2 and Bcl-xL, and increased secretion of TNF-αand IL-12. These results demonstrate that triggering 4-1BB on DCs could increased resistance of DCs to PCa-induced apoptosis. C1 [Youlin, Kuang] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 400016 Chongqing, China. [Jianwei, Zhang] First Affiliated Hospital of Zhengzhou University, Department of UrologyZhengzhou, China. [Xin, Gou] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 400016 Chongqing, China. [Li, Zhang] Wuhan University, Renmin Hospital, Department of UrologyWuhan, China. [Xiaodong, Weng] Wuhan University, Renmin Hospital, Department of UrologyWuhan, China. [Xiuheng, Liu] Wuhan University, Renmin Hospital, Department of UrologyWuhan, China. [Hengchen, Zhu] Wuhan University, Renmin Hospital, Department of UrologyWuhan, China. [Zhiyuan, Chen] Wuhan University, Renmin Hospital, Department of UrologyWuhan, China. RP Xin, G (reprint author), Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 400016 Chongqing, China. EM kyl361@163.com CR Jemal A, Siegel R, Ward E et al, 2008, Cancer statistics, 2008. CA Cancer J Clin 58:71–96 Kuang Y, Weng X, Liu X et al, 2010, Anti-tumor immune response induced by dendritic cells transduced with truncated PSMA IRES 4-1BBL recombinant adenoviruses. Cancer Lett 293:254–262 Lodge PA, Jones LA, Bader RA et al, 2000, Dendritic cell-based immunotherapy of prostate cancer: immune monitoring of a phase II clinical trial. Cancer Res 60:829–833 Tjoa BA, Simmons SJ, Bowes VA et al, 1998, Evaluation of phase I/II clinical trials in prostate cancer with dendritic cells and PSMA peptides. Prostate 36:39–44 Nencioni A, Grunebach F, Schmidt SM et al, 2008, The use of dendritic cells in cancer immunotherapy. Crit Rev Oncol Hematol 65:191–199 Aalamian M, Pirtskhalaishvili G, Nunez A et al, 2001, Human prostate cancer regulates generation and maturation of monocytederived dendritic cells. Prostate 46:68–75 Pirtskhalaishvili G, Shurin GV, Esche C et al, 2000, Cytokinemediated protection of human dendritic cells from prostate cancerinduced apoptosis is regulated by the Bcl-2 family of proteins. Br J Cancer 83:506–513 Pirtskhalaishvili G, Shurin GV, Esche C et al, 2001, TNF-alpha protects dendritic cells from prostate cancer-induced apoptosis. Prostate Cancer Prostatic Dis 4:221–227 Laderach D, Movassagh M, Johnson A et al, 2002, 4-1BB costimulation enhances human CD8(+, T cell priming by augment-ing the proliferation and survival of effector CD8(+, Tcells. Int Immunol 14:1155–1167 Vinay DS, Kwon BS, 1999, Differential expression and costimulatory effect of 4-1BB, CD137, and CD28 molecules on cytokine-induced murine CD8(+, Tc1 and Tc2 cells. Cell Immunol 192:63–71 Gullo C, Koh LK, Pang WL et al, 2010, Inhibition of proliferation and induction of apoptosis in multiple myeloma cell lines by CD137 ligand signaling. PLoS One 5:e10845 Lin GH, Liu Y, Ambagala T et al, 2010, Evaluating the cellular targets of anti-4-1BB agonist antibody during immunotherapy of a pre-established tumor in mice. PLoS One 5:e11003 Melero I, Shuford WW, Newby SA et al, 1997, Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors. Nat Med 3:682–685 Wilcox RA, Chapoval AI, Gorski KS et al, 2002, Cutting edge: expression of functional CD137 receptor by dendritic cells. J Immunol 168:4262–4267 Nagaraj S, Pisarev V, Kinarsky L et al, 2007, Dendritic cell-based full-length survivin vaccine in treatment of experimental tumors. J Immunother 30:169–179 Jahnisch H, Fussel S, Kiessling A et al, 2010, Dendritic cell-based immunotherapy for prostate cancer. Clin Dev Immunol 2010:517493 Troy A, Davidson P, Atkinson C et al, 1998, Phenotypic characterisation of the dendritic cell infiltrate in prostate cancer. J Urol 160:214–219 Amdur RJ, Parsons JT, Fitzgerald LT et al, 1990, The effect of overall treatment time on local control in patients with adenocarcinoma of the prostate treated with radiation therapy. Int J Radiat Oncol Biol Phys 19:1377–1382 Futagawa T, Akiba H, Kodama T et al, 2002, Expression and function of 4-1BB and 4-1BB ligand on murine dendritic cells. Int Immunol 14:275–286 Lee SW, Park Y, So T et al, 2008, Identification of regulatory functions for 4-1BB and 4-1BBL in myelopoiesis and the development of dendritic cells. Nat Immunol 9:917–926 Choi BK, Kim YH, Kwon PM et al, 2009, 4-1BB functions as a survival factor in dendritic cells. J Immunol 182:4107–4115 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 177 EP 181 DI 10.1007/s12253-012-9566-0 PG 5 ER PT J AU Mahjabeen, I Baig, MR Masood, N Sabir, M Inayat, U Malik, AF Kayani, AM AF Mahjabeen, Ishrat Baig, Mehmood Ruqia Masood, Nosheen Sabir, Maimoona Inayat, Uzma Malik, Arshad Faraz Kayani, Akhtar Mahmood TI Genetic Variations in XRCC1 Gene in Sporadic Head and Neck Cancer (HNC) Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HNC; SSCP; DNA; Carcinogenesis; Mutational analysis; XRCC1 ID HNC; SSCP; DNA; Carcinogenesis; Mutational analysis; XRCC1 AB DNA repair gene polymorphisms have been implicated as susceptibility factors in cancer development. It is possible that DNA repair polymorphisms may also influence the risk of gene mutation. Polymorphisms in the DNA repair gene XRCC1 have been indicated to have a contributive role in DNA adduct formation and an increased risk of cancer development. 300 head and neck cancer patients and 150 controls were included in this study. PCR-single-strand conformation polymorphism and DNA sequencing were used to analyze the whole exonic region of XRCC1 in head and neck cancer patients. Sequence analysis revealed two missense and two silent mutations in our study. Frequency of silent mutations; Pro206Pro (rs915927) and Gln632Gln (rs3547) was calculated as 0.16 (16 %) and 0.30 (30 %) respectively. Whereas, the frequency of missense mutations; Arg399Gln (rs25487) and Tyr576Asn (rs2307177) was calculated as 0.27 (27 %) and 0.28 (28 %) respectively. In our study, incidence of these mutations was found higher in larynx cancer (p<0.005) as compared to oral cavity and pharynx cancer. Our finding suggests that the polymorphic XRCC1 gene may contribute to risk of developing head and neck cancer. To our knowledge, this is the first report that XRCC1 is associated with increased risk of head and neck cancer in a Pakistani population. C1 [Mahjabeen, Ishrat] COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics LabIslamabad, Pakistan. [Baig, Mehmood Ruqia] COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics LabIslamabad, Pakistan. [Masood, Nosheen] COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics LabIslamabad, Pakistan. [Sabir, Maimoona] COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics LabIslamabad, Pakistan. [Inayat, Uzma] COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics LabIslamabad, Pakistan. [Malik, Arshad Faraz] COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics LabIslamabad, Pakistan. [Kayani, Akhtar Mahmood] COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics LabIslamabad, Pakistan. RP Kayani, AM (reprint author), COMSATS Institute of Information Technology, Department of Biosciences, Cancer Genetics Lab, Islamabad, Pakistan. EM mkayani@comsats.edu.pk CR Parkin DM, Pisani P, Ferley J, 1999, Global cancer statistics. CA Cancer J Clin 49:33–64 Scully C, Field JK, Tanzawa H, 2000, Genetic aberrations in oral or head and neck squamous cell carcinoma, SCCHN, Carcinogen metabolism, DNA repair and cell cycle control. Oral Oncol 36:256–263 Marsin S, Vidal AE, Sossou M, Menissier-de Murcia J, Le Page F, Boiteux S, de Murcia G, Radicella JP, 2003, Role of XRCC1 in the coordination and stimulation of oxidative DNA damage repair initiated by the DNA glycosylase hOGG1. J Biol Chem 27844068-44074 Marintchev A, Mullen MA, Maciejewski MW, Pan B, Gryk MR, Mullen GP, 1999, Solution structure of the single-strand break repair protein XRCC1-N-terminal domain. Nat Struct Biol 6:884–893 Mao L, Hong WK, 2004, How does human papillomavirus contribute to head and neck cancer development? J Nat Cancer Inst 13:978–979 Taningher M, Malacarne D, Izzotti A, Ugolini D, Parodi S, 1999, Drug metabolism polymorphisms as modulators of cancer susceptibility. Mutat Res 436:227–261 Nash RA, Caldecott KW, Barnes DE, Lindahl T, 1997, XRCC1 protein interacts with one of two distinct forms of DNA ligase III. Biochemistry 36:5207–5211 Chacko P, Rajan B, Joseph T, Mathew BS, Pillai MR, 2005, Polymorphisms in DNA repair gene XRCC1 and increased genetic susceptibility to breast cancer. Breast Cancer Res Treat 89:15–21 Fan J, Otterlei M, Wong HK, Tomkinson AE, Wilson DM, 2004, XRCC1 co-localizes and physically interacts with PCNA. Nucleic Acids Res 322193–2201 Vierhapper H, Bieglmayer C, Heinze G, Baumgartner-Parzer SM, 2004, Frequency of RET protooncogene mutations in patients with normal and with moderately elevated, 50–100 pg/ml, pentagastrin-stimulated serum concentrations of calcitonin. Thyroid 14:580–583 Yin J, Qi R, Ma Y, Sun Z, Wang H, 2007, Genetic polymorphisms of DNA Repair Gene: XRCC1 Pro206Pro and Gln632Gln in a Chinese Han population. Biochem Genet 45:815–821 Hung RJ, Hall J, Brennan P, Boffetta P, 2005, Genetic polymorphisms in the base excision repair pathway and cancer risk: a HuGE review. Am J Epidemiol 162:925–942 Schreiber V, Ame JC, Dolle P, Schultz I, Rinaldi B, Fraulob V, Menissier-de Murcia J, De-murcia G, 2007, Poly(ADP-ribose, Polymerase-2, PARP-2, Is Required for Efficient Base Excision DNA Repair in Association with PARP-1 and XRCC1. J Biol Chem 277:23028–23036 Campalans A, Marsin S, Nakabeppu Y, Connor TR, Boiteux S, Radicella JP, 2005, XRCC1 interactions with multiple DNA glycosylases: a model for its recruitment to base excision repair. DNA Repair 4:826–835 Berquist BR, Singh DK, Jinshui F, Daemyung K, Elizabeth G, Avanti K, Vilhelm AB, Eric JA, Alan ET, David MW, 2010, Functional capacity of XRCC1 protein variants identified in DNA repair-deficient Chinese hamster ovary cell lines and the human population. Nucleic Acids Res 38:5023–5035 Holmila R, Luce D, Bornholdt J, Heikkila P, Suitiala T, Fevotte J, Cyr D, Hansen J et al, 2009, Mutations in TP53 tumor suppressor gene in wood dust related sinonasal cancer. Int J Cancer., DOI 10.1002/ijc.25064 Vidal AE, Boiteux, S, Hickson ID, Radicella JP, 2001, XRCC1 coordinates the initial and late stages of DNA abasic site repair through protein–protein interactions. EMBO J 206530–6539 Holmila R, Bornholdt J, Suitiala T, Cyr D, Dictor M, Steiniche T, Wolff H, Wallim H et al, 2010, Profile of T53 mutations in sinonasal cancer. Mutation Res 686:9–14 Talamini R, Bosetti C, Vecchia CL, Maso LD, Levi F, Bidoli E, Negri E, Pasche C et al, 2002, Combined effect of tobacco and alcohol on laryngeal cancer risk: a case–control study. Cancer Causes and Control 13:957–964 Yin J, Vogel U, Ma Y, Qi R, Sun Z, Wang H, 2007, The DNA repair gene XRCC1 and genetic susceptibility of lung cancer in a northeastern Chinese population. Lung Cancer 56:153–160 Majumder M, Indra D, Roy PD, Datta S, Ray JG, Panda CK, Roy B, 2009, Variant haplotypes at XRCC1 and risk of oral leukoplakia in HPV non-infected samples. J Oral Pathol Med 38:174–180 Settheetham-Ishida W, Yuenyao P, Natphopsuk S, Settheetham D, Ishida T, 2011, Genetic Risk of DNA Repair Gene Polymorphisms, XRCC1 and XRCC3, for High Risk Human Papillomavirus Negative Cervical Cancer in Northeast Thailand. APJCP 12:963–966 Lamerdin JE, Montgomery MA, Stilwagen SA, Scheidecker LK, Tebbs RS, Brookman KW, Thompson LH, Carrano AV, 1995, Genomic sequence comparison of the human and mouse XRCC1 DNA repair gene regions. Genomics 25:547–554 Ramachandran S, Ramadas K, Hariharan R, Kumar RR, Pillai MR, 2005, Single nucleotide polymorphisms of DNA repair genes XRCC1 and XPD and its molecular mapping in Indian oral cancer. Oral oncology 42:350–362 Yin J, Vogel U, Gerdes LU, Dybdahl M, Bolund L, Nexo BA, 2003, Twelve single nucleotide polymorphisms on chromosome 19q13.2–3: Linkage disequilibria and associations with basal cell carcinoma in Danish psoriatic patients. Biochem Genet 41:27–37 Au WW, Salama SA, Sierra-Torres CH, 2003, Functional characterization of polymorphisms in DNA repair genes using cytogenetic challenge assays. Environ Health Perspect 111:1843–1850 Jbic-allele, DBSNP of NCBI, [dhttp://www.ncbi.nlm.nlh.gov/SNP] David-Beabes GL, London SJ, 2001, Genetic polymorphism of XRCC1 and lung cancer risk among African–Americans and Caucasians. Lung Cancer 34:333–339 Cao Y, Miao X, Huang M, Deng L, Hu L, Ernberg I, Zeng Y, Lin D, Shao J, 2006, Polymorphisms of XRCC1 genes and risk of nasopharyngeal carcinoma in the Cantonese population. BMC Cancer., DOI 10.1186/1471-2407-6-167 Abdel-rahman SZ, Soliman AS, Bondy ML, Omar S, El-Badawy SA, Khaled HM, Seifeldin IA, Levin B, 2000, Inheritance of the 194Trp and the 399Gln variant alleles of the DNA repair gene XRCC1 are associated with increased risk of early-onset colorectal carcinoma in Egypt. Cancer Lett 159:79–86 Xu Z, Hua LX, Qian LX, Yang J, Wang XR, Zhang W, Wu HF, 2007, Relationship between XRCC1 polymorphisms and susceptibility to prostatte cancer in men from Han, Southern China. Asian J Androl 9:331–338 Park JY, Lee SY, Jeon HS, Bae NC, Chae SC, Joo S, Kim CH, Park JH, 2002, Polymorphism of the DNA repair gene XRCC1 and risk of primary lung cancer. Cancer Epidemiol Biomark Prev 11:23–27 Shen H, Xu Y, Yu R, Qin Y, Zhou L, Wang X, Spitz MR, 2000, Polymorphism of the DNA repair gene XRCC1 and risk of gastric cancer in a Chinese population. Int J Cancer 88:601–606 Yeh CC, Sung FC, Tang R, Chang-Chieh CR, Hsieh LL, 2005, Polymorphisms of the XRCC1, XRCC3, & XPD genes and colorectal cancer risk: A case–control study in Taiwan. BMC Cancer 5:8 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 183 EP 188 DI 10.1007/s12253-012-9567-z PG 6 ER PT J AU Chaisuparat, R Rojanawatsirivej, S Yodsanga, S AF Chaisuparat, Risa Rojanawatsirivej, Somsri Yodsanga, Somchai TI Ribosomal Protein S6 Phosphorylation is Associated with Epithelial Dysplasia and Squamous Cell Carcinoma of the Oral Cavity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ribosomal protein S6; Epithelial dysplasia; Oral squamous cell carcinoma; Tumor progression ID Ribosomal protein S6; Epithelial dysplasia; Oral squamous cell carcinoma; Tumor progression AB Ribosomal protein S6 (RPS6), a downstream effector of the mammalian target of rapamycin pathway (mTOR), is activated in many cancers including oral squamous cell carcinoma (OSCC). However, the role of RPS6 in the progression of potentially malignant disorders (or premalignant lesions) to OSCC is unknown. The purpose of this study was to examine the expression of RPS6 in epithelial dysplasia and OSCC to determine the association of RPS6 in tumor progression. In our study, an immunohistochemical analysis of RPS6 was performed on tissue microarrays containing 30 control samples, 15 epithelial dysplasia cases, and 53 OSCC cases. Correlations between the clinicopathologic features of OSCC and RPS6 expression were analyzed using the Chi-square test. We found RPS6 phosphorylation (p-RPS6) in 15/30 (50 %) control normal oral mucosa samples, 15/15 (100 %) epithelial dysplasia cases, and 47/53 (88.68 %) OSCC cases. The frequency of p-RPS6 in epithelial dysplasia or OSCC showed a statistically significant difference compared to control (P<0.001). However, there were no significant correlations between p-RPS6 and the clinicopathologic features of OSCC. Our findings suggest that RPS6 activation is associated with the early events of tumor progression, suggesting p-RPS6 as a potential marker for early detection of oral cancer. C1 [Chaisuparat, Risa] Chulalongkorn University, Faculty of Dentistry, Department of Oral Pathology, Henry-Dunant Road, Pathumwan, 10330 Bangkok, Thailand. [Rojanawatsirivej, Somsri] Chulalongkorn University, Faculty of Dentistry, Department of Oral Pathology, Henry-Dunant Road, Pathumwan, 10330 Bangkok, Thailand. [Yodsanga, Somchai] Chulalongkorn University, Faculty of Dentistry, Department of Oral Pathology, Henry-Dunant Road, Pathumwan, 10330 Bangkok, Thailand. RP Chaisuparat, R (reprint author), Chulalongkorn University, Faculty of Dentistry, Department of Oral Pathology, 10330 Bangkok, Thailand. EM risa.c@chula.ac.th CR Johnson NW, Jayasekara P, Amarasinghe AA, 2011, Squamous cell carcinoma and precursor lesions of the oral cavity: epidemiology and aetiology. Periodontol 2000 57(1):19–37 Warnakulasuriya S, 2009, Global epidemiology of oral and oropharyngeal cancer. Oral Oncol 45:309–316 Napier SS, Speight PM, 2008, Natural history of potentially malignant oral lesions and conditions: an overview of the literature. J Oral Pathol Med 37:1–10 Mithani SK, Mydlarz WK, Grumbine FL et al, 2007, Molecular genetics of premalignant oral lesions. Oral Dis 13:126–133 Ruvinsky I, Meyuhas O, 2006, Ribosomal protein S6 phosphorylation: from protein synthesis to cell size. Trends Biochem Sci 31:342–348 Vivanco I, Sawyers CL, 2002, The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer 2:489–501 Ruvinsky I, Katz M, Dreazen A et al, 2009, Mice deficient in ribosomal protein S6 phosphorylation suffer from muscle weakness that reflects a growth defect and energy deficit. PLoS One 4: e5618 Ruvinsky I, Sharon N, Lerer T, Cohen H, Stolovich-Rain M, Nir T, Dor Y, Zisman P, Meyuhas O, 2005, Ribosomal protein S6 phosphorylation is a determinant of cell size and glucose homeostasis. Genes Dev 19(18):2199–2211 Clark C, Shah S, Herman-Ferdinandez L et al, 2010, Teasing out the best molecular marker in the AKT/mTOR pathway in head and neck squamous cell cancer patients. Laryngoscope 120:1159–1165 Molinolo AA, Hewitt SM, Amornphimoltham P et al, 2007, Dissecting the Akt/mammalian target of rapamycin signaling network: emerging results from the head and neck cancer tissue array initiative. Clin Cancer Res 13:4964–4973 Kononen J, Bubendorf L, Kallioniemi A et al, 1998, Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4:844–847 Bose S, Chandran S, Mirocha JM et al, 2006, The Akt pathway in human breast cancer: a tissue-array-based analysis. Mod Pathol 19:238–245 Pantuck AJ, Seligson DB, Klatte T et al, 2007, Prognostic relevance of the mTOR pathway in renal cell carcinoma: implications for molecular patient selection for targeted therapy. Cancer 109:2257–2267 Bjornsti MA, Houghton PJ, 2004, The TOR pathway: a target for cancer therapy. Nat Rev Cancer 4:335–348 Scheper MA, Chaisuparat R, Nikitakis NG et al, 2008, Expression and alterations of the PTEN/AKT/mTOR pathway in ameloblastomas. Oral Dis 14:561–568 Amornphimoltham P, Patel V, Sodhi A et al, 2005, Mammalian target of rapamycin, a molecular target in squamous cell carcinomas of the head and neck. Cancer Res 65:9953–9961 Liu Y, Hidayat S, Su WH et al, 2007, Expression and activity of mTOR and its substrates in different cell cycle phases and in oral squamous cell carcinomas of different malignant grade. Cell Biochem Funct 25:45–53 Garnis C, Buys TP, Lam WL, 2004, Genetic alteration and gene expression modulation during cancer progression. Mol Cancer 3:9 Vitale-Cross L, Czerninski R, Amornphimoltham P et al, 2009, Chemical carcinogenesis models for evaluating molecular-targeted prevention and treatment of oral cancer. Cancer Prev Res, Phila, 2:419–422 Feng CJ, Li HJ, Li JN et al, 2008, Expression of Mcm7 and Cdc6 in oral squamous cell carcinoma and precancerous lesions. Anticancer Res 28:3763–3769 Inoue H, Miyazaki Y, Kikuchi K et al, 2012, Podoplanin expression during dysplasia-carcinoma sequence in the oral cavity. Tumour Biol 33:183–194 Jham BC, Costa NL, Silva JM et al, 2012, Midkine expression in oral squamous cell carcinoma and leukoplakia. J Oral Pathol Med 41:21–26 Kato Y, Uzawa K, Yamamoto N et al, 2007, Overexpression of Septin1: possible contribution to the development of oral cancer. Int J Oncol 31:1021–1028 Montebugnoli L, Cervellati F, Cocchi R et al, 2010, Immunohistochemical expression of p16(INK4A, protein as a helpful marker of a subset of potentially malignant oral epithelial lesions: study on a series with long-term follow-up. Histopathology 57:528–534 Nomura H, Uzawa K, Yamano Y et al, 2009, Down-regulation of plasma membranous Annexin A1 protein expression in premalignant and malignant lesions of the oral cavity: correlation with epithelial differentiation. J Cancer Res Clin Oncol 135:943–949 Torres-Rendon A, Roy S, Craig GT et al, 2009, Expression of Mcm2, geminin and Ki67 in normal oral mucosa, oral epithelial dysplasias and their corresponding squamous-cell carcinomas. Br J Cancer 100:1128–1134 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 189 EP 193 DI 10.1007/s12253-012-9568-y PG 5 ER PT J AU Li, L Zhang, A Cao, X Chen, J Xia, Y Zhao, H Shen, A AF Li, Liren Zhang, Aixian Cao, Xiaolei Chen, Jing Xia, Yunfei Zhao, Hui Shen, Aiguo TI General Transcription Factor IIB Overexpression and a Potential Link to Proliferation in Human Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human hepatocellular carcinoma (HCC); General transcription factor IIB (TFIIB); Cell proliferation; Pathogenesis ID Human hepatocellular carcinoma (HCC); General transcription factor IIB (TFIIB); Cell proliferation; Pathogenesis AB The general transcription factor IIB (TFIIB) plays a central role in preinitiation complex (PIC) assembly, providing a bridge between promoter-bound TFIID and RNA Polymerase II (RNA POLII). TFIIB functionally counteracts the transcriptional activation of hepatitis B virus X protein (HBx), which has been shown to play a role in the development of human hepatocellular carcinoma (HCC). However, the function of TFIIB in HCC remains unclear. In this article, we demonstrate that TFIIB plays an important role in HCC pathogenesis. TFIIB expression was immunohistochemically examined in a series of 100 HCC tissue specimens. The expression level of TFIIB showed significant correlation with the histological grade (P<0.030), the level of AFP (P<0.011) and the proliferation marker Ki-67 (P<0.0002). High TFIIB expression level correlated with poor survival. Western blot analysis also confirmed that the TFIIB protein was overexpressed in HCC tissue compared to benign normal tissue. Additionally, Western blot and qRT-PCR analyses showed a high expression level of TFIIB protein in the HCC cell lines SMMC7721, HepG2, BEL7404, and Huh7 and the immortalized normal line BEL7702 but a lower expression in the normal Chang hepatocyte cell line. Following the release of Huh7 cells from serum starvation, the expression of TFIIB was upregulated. A cell growth assay suggested that TFIIB was involved in the proliferation and growth of HCC cells. In conclusion, our results demonstrate that TFIIB overexpression may play essential roles in the pathogenesis of hepatocellular carcinoma by affecting the proliferation of HCC cells. C1 [Li, Liren] Affiliated Hospital of Nantong University, Department of Internal Medicine, 226001 Nantong, China. [Zhang, Aixian] Affiliated Hospital of Nantong University, Department of Internal Medicine, 226001 Nantong, China. [Cao, Xiaolei] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, China. [Chen, Jing] Affiliated Hospital of Nantong University, Department of Surgery, 226001 Nantong, China. [Xia, Yunfei] Affiliated Hospital of Nantong University, Department of Internal Medicine, 226001 Nantong, China. [Zhao, Hui] Affiliated Hospital of Nantong University, Department of Internal Medicine, 226001 Nantong, China. [Shen, Aiguo] Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, 226001 Nantong, China. RP Shen, A (reprint author), Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, 226001 Nantong, China. EM jsntzax@163.com CR Feng H, Cheng AS, Tsang DP et al, 2011, Cell cycle-related kinase is a direct androgen receptor-regulated gene that drives β-catenin/ T cell factor-dependent hepatocarcinogenesis. J Clin Invest 121:3159–3175 Hsieh A, Kim HS, Lim SO et al, 2011, Hepatitis B viral X protein interacts with tumor suppressor adenomatous polyposis coli to activate Wnt/β-catenin signaling. Cancer Lett 300:162–172 Lau WY, 2000, Primary liver tumors. Semin Surg Oncol 19:135– 144 Llovet JM, Bruix J, 2008, Molecular targeted therapies in hepatocellular carcinoma. Hepatology 48:1312–1327 El-Serag HB, Rudolph KL, 2007, Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 132:2557–2576 Ueda S, Basaki Y, Yoshie M et al, 2006, PTEN/Akt signaling through epidermal growth factor receptor is prerequisite for angiogenesis by hepatocellular carcinoma cells that is susceptible to inhibition by gefitinib. Cancer Res 66:5346–5353 Baek HJ, Lim SC, Kitisin K et al, 2008, Hepatocellular cancer arises from loss of transforming growth factor beta signaling adaptor protein embryonic liver fodrin through abnormal angiogenesis. Hepatology 48:1128–1137 Deng WS, Roberts SGE, 2007, TFIIB and the regulation of transcription by RNA polymerase II. Chromosoma 116:417–429 Albert TK, Grote K, Boeing S et al, 2010, Basal core promoters control the equilibrium between negative cofactor 2 and preinitiation complexes in human cells. Genome Biol 11:R33 Sevilimedu A, Shi H, Lis JT, 2008, TFIIB aptamers inhibit transcription by perturbing PIC formation at distinct stages. Nucleic Acids Res 36:3118–3127 Wang YM, Fairley JA, Roberts SGE, 2010, Phosphorylation of TFIIB links transcription initiation and termination. Curr Biol 20:548–553 Liu X, Bushnell DA, Wang D et al, 2010, Structure of an RNA polymerase II–TFIIB complex and the transcription initiation mechanism. Science 327:206–209 Simone C, Robert OJ, 2010, The linker domain of basal transcription factor TFIIB controls distinct recruitment and transcription stimulation functions. Nucleic Acids Res 39:464–474 Haviv I, Shamay M, Doitsh G et al, 1998, Hepatitis B virus pX targets TFIIB in transcription coactivation. Mol Cell Biol 18:1562– 1569 Yang Y, Zheng L, Chen Y, 2000, Study of HBV X protein and RMP, an RPB5 mediate protein competitively interacting with general transcription factor TF2B. Zhonghua Gan Zang Bing Za Zhi 8:15–17 Yang H, Gu J, Zheng Q et al, 2011, RPB5-mediating protein is required for the proliferation of hepatocellular carcinoma cells. J Biol Chem 286:11865–11874 Liu Z, Wang D, Shao B et al, 2011, Increased expression of transcription initiation factor IIB after rat traumatic brain injury. J Mol Histol 42:265–271 Lau WY, Lai EC, 2008, Hepatocellular carcinoma: current management and recent advances. Hepatobiliary Pancreat 7:237–257 Li X, Pan Y, Fan R et al, 2008, Adenovirus-delivered CIAPIN1 small interfering RNA inhibits HCC growth in vitro and in vivo. Carcinogenesis 29:1587–1593 Hahn S, 2004, Structure and mechanism of the RNA polymerase II transcription machinery. Nat Struct Mol Biol 11:394–403 Wada C, Kasai K, Kameya T et al, 1992, A general transcription initiation factor, human transcription factor IID, overexpressed in human lung and breast carcinoma and rapidly induced with serum stimulation. Cancer Res 52:307–313 Wei W, Gu JX, Zhu CQ et al, 2003, Interaction with general transcription factor IIF, TFIIF, is required for the suppression of activated transcription by RPB5-mediating protein, RMP). Cell Res 13:111–120 Qadri I, Fatima K, AbdeL-Hafiz H, 2011, Hepatitis B virus X protein impedes the DNA repair via its association with transcription factor, TFIIH. BMC Microbiol 11:48 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 195 EP 203 DI 10.1007/s12253-012-9569-x PG 9 ER PT J AU Ji, HX Zhao, Q Pan, JH Shen, WH Chen, ZW Zhou, ZS AF Ji, Hui-Xiang Zhao, Qian Pan, Jin-Hong Shen, Wen-Hao Chen, Zhi-Wen Zhou, Zhan-Song TI Association of BLCA-4 Hypomethylation in Blood Leukocyte DNA and the Risk of Bladder Cancer in a Chinese Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BLCA-4; Hypomethylation; Bladder cancer; Blood leukocyte ID BLCA-4; Hypomethylation; Bladder cancer; Blood leukocyte AB Global DNA hypomethylation has been associated with increased risk for cancers of the colorectum, bladder, breast, head and neck, and testicular germ cells. The aim of this study was to examine whether global hypomethylation measured at BLCA-4 repeat regions through bisulfite pyrosequencing in blood leukocyte DNA is associated with the risk of bladder cancer(BC). A total of 312 bladder cancer patients and 361 healthy control subjects were included in Chongqing, China. Global methylation in blood leukocyte DNA was estimated by analyzing BLCA-4 repeats using bisulfite-polymerase chain reaction (PCR) and pyrosequencing. The median methylation level in BC cases (percentage of 5-methylcytosine (5 mC)075.7 %) was significantly lower than that in controls (79.7 % 5 mC) (P<0.002, Wilcoxon rank-sum test). The odds ratios (ORs) of BC for individuals in the third, second, and first (lowest) quartiles of BLCA-4 methylation were 1.2 (95 % confidence interval (CI) 0.8– 1.9), 1.6 (95 % CI 1.1–2.3), and 2.7 (95 % CI 1.5–3.8) (P for trend <0.001), respectively, compared to individuals in the fourth (highest) quartile. A 2.1-fold (95 % CI 1.5–2.8) increased risk of BC was observed among individuals with BLCA-4 methylation below the median compared to individuals with higher (>median) BLCA-4 methylation. Our results demonstrate for the first time that individuals with global hypomethylation measured in BLCA-4 repeats in blood leukocyte DNA have an increased risk for BC. Our data provide the evidence that BLCA-4 hypomethylation may be a useful biomarker for poor prognosis of patients with BC. C1 [Ji, Hui-Xiang] Southwest Hospital Affilated to Third Military Medical University, Urology Department, No. 33, Gaotanyanzheng RD, 400038 Chongqing, China. [Zhao, Qian] Kunming General Hospital of Chengdu Military Region, Urology Department, No. 212, Daguan RD, 650032 Kunming, China. [Pan, Jin-Hong] Southwest Hospital Affilated to Third Military Medical University, Urology Department, No. 33, Gaotanyanzheng RD, 400038 Chongqing, China. [Shen, Wen-Hao] Southwest Hospital Affilated to Third Military Medical University, Urology Department, No. 33, Gaotanyanzheng RD, 400038 Chongqing, China. [Chen, Zhi-Wen] Southwest Hospital Affilated to Third Military Medical University, Urology Department, No. 33, Gaotanyanzheng RD, 400038 Chongqing, China. [Zhou, Zhan-Song] Southwest Hospital Affilated to Third Military Medical University, Urology Department, No. 33, Gaotanyanzheng RD, 400038 Chongqing, China. RP Zhou, ZS (reprint author), Southwest Hospital Affilated to Third Military Medical University, Urology Department, 400038 Chongqing, China. EM zhouzs@yahoo.com.cn CR Parkin MP, 2008, The global burden of urinary bladder cancer. Scand J Urol Nephrol 42:12–20 Morrison AS, 1984, Advances in the etiology of urothelial cancer. Urol Clin North Am 11:557–566 Burch JD, Rohan TE, Howe GR, Risch HA, Hill GB, Steele R, Miller AB, 1989, Risk of bladder cancer by source and type of tobacco exposure: a case-control study. Int J Cancer 44:622–628 Miller BA, Kolonel LN, Bernstein L. Racial/Ethnic Patterns of Cancer in the United States 1988–1992 Das PM, Singal R, 2004, DNA methylation and cancer. J Clin Oncol 22:4632–4642 Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J, Gray JW, Leonhardt H, Jaenisch R, 2003, Induction of tumors in mice by genomic hypomethylation. Science 300:489–492 Zhu Z-Z, Sparrow D, Hou L, Tarantini L, Bollati V, Litonjua AA, Zanobetti A, Vokonas P, Wright RO, Baccarelli A, 2011, Repetitive element hypomethylation in blood leukocyte DNA and cancer incidence, prevalence, and mortality in elderly individuals: the normative aging study. Canc Causes Contr 22:437–447 Wilson MJ, Shivapurkar N, Poirier LA, 1984, Hypomethylation of hepatic nuclear DNA in rats fed with a carcinogenic methyldeficient diet. Biochem J 218:987–990 Thomas GA, Williams ED, 1992, Production of thyroid tumours in mice by demethylating agents. Carcinogenesis 13:1039–1042 Pufulete M, Al-Ghnaniem R, Leather AJ, Appleby P, Gout S, Terry C, Emery PW, Sanders TA, 2003, Folate status, genomic DNA hypomethylation, and risk of colorectal adenoma and cancer: a case control study. Gastroenterology 124:1240–1248 Lim U, Flood A, Choi SW, Albanes D, Cross AJ, Schatzkin A, Sinha R, Katki HA, Cash B, Schoenfeld P, Stolzenberg-Solomon R, 2008, Genomic methylation of leukocyte DNA in relation to colorectal adenoma among asymptomatic women. Gastroenterology 134:47–55 Bollati V, Baccarelli A, Hou L, Bonzini M, Fustinoni S, Cavallo D, Byun HM, Jiang J, Marinelli B, Pesatori AC, Bertazzi PA, Yang AS, 2007, Changes in DNA methylation patterns in subjects exposed to low-dose benzene. Cancer Res 67:876–880 Rusiecki JA, Baccarelli A, Bollati V, Tarantini L, Moore LE, Bonefeld-Jorgensen EC, 2008, Global DNA hypomethylation is associated with high serum-persistent organic pollutants in Greenlandic Inuit. Environ Health Perspect 116:1547–1552 Breton CV, Byun HM, Wenten M, Pan F, Yang A, Gilliland FD, 2009, Prenatal tobacco smoke exposure affects global and genespecific DNA methylation. Am J Respir Crit Care Med 180:462– 467 Liu F, Killian JK, Yang M, Walker RL, Hong JA, Zhang M, Davis S, Zhang Y, Hussain M, Xi S, Rao M, Meltzer PA, Schrump DS, 2010, Epigenomic alterations and gene expression profiles in respiratory epithelia exposed to cigarette smoke condensate. Oncogene 29:3650–3664 Wan YJ, Li YY, Xia W, Chen J, Lv ZQ, Zeng HC, Zhang L, Yang WJ, Chen T, Lin Y, Wei J, Xu SQ, 2010, Alterations in tumor biomarker GSTP gene methylation patterns induced by prenatal exposure to PFOS. Toxicology 274:57–64 Hsiung DT, Marsit CJ, Houseman EA, Eddy K, Furniss CS, McClean MD, Kelsey KT, 2007, Global DNA methylation level in whole blood as a biomarker in head and neck squamous cell carcinoma. Canc Epidemiol Biomarkers Prev 16:108–114 Moore LE, Pfeiffer RM, Poscablo C, Real FX, Kogevinas M, Silverman D, Garcia-Closas R, Chanock S, Tardon A, Serra C, Carrato A, Dosemeci M, Garcia-Closas M, Esteller M, Fraga M, Rothman N, Malats N, 2008, Genomic DNA hypomethylation as a biomarker for bladder cancer susceptibility in the Spanish bladder cancer study: a case-control study. Lancet Oncol 9:359–366 Choi JY, James SR, Link PA, McCann SE, Hong CC, Davis W, Nesline MK, Ambrosone CB, Karpf AR, 2009, Association between global DNA hypomethylationin leukocytes and risk of breast cancer. Carcinogenesis 30:1889–1897 Cho YH, Yazici H, Wu HC, Terry MB, Gonzalez K, Qu M, Dalay N, Santella RM, 2010, Aberrant promoter hypermethylation and genomic hypomethylation in tumor, adjacent normal tissues and blood from breast cancer patients. Anticancer Res 30:2489–2496 Mirabello L, Savage SA, Korde L, Gadalla SM, Greene MH, 2010, BLCA-4 methylation is inherited in familial testicular cancer kindreds. BMC Med Genet 11:77 Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W et al, 2001, Initial sequencing and analysis of the human genome. Nature 409, 6822):860–921 Getzenberg RH, Konety BR, Oeler TA, Quigley MM, Hakam A, Becich MJ, Bahnson RR, 1996, Bladder cancer-associated nuclear matrix proteins. Cancer Res 56:1690–1694 Shirodkar SP, Lokeshwar VB, 2009, Potential new urinary markers in the early detection of bladder cancer. Curr Opin Urol 19:488–493 Mostofi FK, Sorbin LH, Torloni H, 1973, Histologic Typing of Urinary Bladder Tumours. International classification of tumours, No 10. WHO, Geneva Hsieh CL, 2000, Dynamics of DNA methylation pattern. Curr Opin Genet Dev 10:224–228 Gluckman PD, Hanson MA, Cooper C, Thornburg KL, 2008, Effect of in utero and early-life conditions on adult health and disease. N Engl J Med 359:61–73 Bjornsson HT, Sigurdsson MI, Fallin MD, Irizarry RA, Aspelund T, Cui H, YuW, Rongione MA, Ekstrom TJ, Harris TB, Launer LJ, Eiriksdottir G, Leppert MF, Sapienza C, Gudnason V, Feinberg AP, 2008, Intra-individual change over time in DNA methylation with familial clustering. JAMA 299:2877–2883 Hillemacher T, Frieling H, Moskau S, Muschler MA, Semmler A, Kornhuber J, Klockgether T, Bleich S, Linnebank M, 2008, Global DNA methylation is influenced by smoking behaviour. Eur Neuropsychopharmacol 18:295–298 Feinberg AP, Ohlsson R, Henikoff S, 2006, The epigenetic progenitor origin of human cancer. Nat Rev Genet 7:21–33 Lamprecht B,Walter K, Kreher S, Kumar R, Hummel M, Lenze D, Kochert K, Bouhlel MA, Richter J, Soler E, Stadhouders R, Johrens K, Wurster KD, Callen DF, Harte MF, Giefing M, Barlow R, Stein H, Anagnostopoulos I, Janz M, Cockerill PN, Siebert R, Dorken B, Bonifer C, Mathas S, 2010, Derepression of an endogenous long terminal repeat activates the CSF1R proto-oncogene in human lymphoma. Nat Med 16:571–579 Esteller M, 2008, Epigenetics in cancer. N Engl J Med 358:1148– 1159 Esteller M, 2007, Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet 8:286–298 Konety BR, Nguyen TS, Brenes G, Sholder A, Lewis N, Bastacky S, Potter DM, Getzenberg RH, 2000, Clinical usefulness of the novel marker BLCA-4 for the detection of bladder cancer. J Urol 164:634–639 Van Le TS, Miller R, Barder T, Babjuk M, Potter DM, Getzenberg RH, 2005, Highly specific urine-based marker of bladder cancer. Urology 66:1256–1260 Wolff EM, Byun HM, Han HF, Sharma S, Nichols PW, Siegmund KD, Yang AS, Jones PA, Liang G, 2010, Hypomethylation of a LINE-1 promoter activates an alternate transcript of the MET oncogene in bladders with cancer. PLoS Genet 6:e1000917 Suzuki H, Toyota M, Sato H, Sonoda T, Sakauchi F, Mori M, 2006, Roles and causes of abnormal DNA methylation in gastrointestinal cancers. Asian Pac J Cancer Prev 7:177–185 Schernhammer ES, Giovannucci E, Kawasaki T, Rosner B, Fuchs CS, Ogino S, 2010, Dietary folate, alcohol and B vitamins in relation to LINE-1 hypomethylation in colon cancer. Gut 59:794– 799 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 205 EP 210 DI 10.1007/s12253-012-9570-4 PG 6 ER PT J AU Cserni, G Bezsenyi, I Marko, L AF Cserni, Gabor Bezsenyi, Istvanne Marko, Laszlo TI Patients’ Choice on Axillary Lymph Node Dissection Following Sentinel Lymph Node Micrometastasis — First Report on Prospective Use of a Nomogram in Very Low Risk Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sentinel lymph node; Non sentinel lymph node; Micrometastasis; Nomogram; Patient’s choice; Axillary lymph node dissection ID Sentinel lymph node; Non sentinel lymph node; Micrometastasis; Nomogram; Patient’s choice; Axillary lymph node dissection AB The optimal locoregional treatment of patients diagnosed with sentinel node (SN) micrometastasis is controversial. A previously reported and validated nomogram was used to calculate the risk of non-SN metastasis in patients with SN micrometastasis over a period of 2 years. Patients were given detailed information about the risk, consequences and treatment options of non-SN involvement, the risk and potential complications of unnecessary completion axillary lymph node dissection (ALND), the imperfectness of the nomogram, and other factors that may influence their selection of further treatment. They also received a questionnaire to monitor factors influencing their decisions. Of the 25 patients participating in the study, 10 have opted for ALND. The only factor that seemed to influence their choice was fear from disease recurrence. Giving detailed information to SN micrometastatic patients is a patient-centered alternative to current recommendations on performing ALND in all such patients or omitting ALND in all of them. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, 6000 Kecskemet, Hungary. [Bezsenyi, Istvanne] University of Szeged, Department of Pathology, Allomas u. 2, 6720 Szeged, Hungary. [Marko, Laszlo] University of Szeged, Department of Pathology, Allomas u. 2, 6720 Szeged, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, 6000 Kecskemet, Hungary. EM cserni@freemail.hu CR Chu KU, Turner RR, Hansen NM, Brennan MB, Bilchik A, Giuliano AE, 1999, Do all patients with sentinel node metastasis from breast carcinoma need complete axillary node dissection? Ann Surg 229:536–541 Degnim AC, Griffith KA, Sabel MS et al, 2003, Clinicopathologic features of metastasis in nonsentinel lymph nodes of breast carcinoma patients. Cancer 98:2307–2315 Van la Parra RF, Peer PG, Ernst MF, Bosscha K, 2011, Metaanalysis of predictive factors for non-sentinel lymph node metastases in breast cancer patients with a positive SLN. Eur J Surg Oncol 37:290–299 Kumar S, Bramlage M, Jacks LM et al, 2010, Minimal disease in the sentinel lymph node: how to best measure sentinel node micrometastases to predict risk of additional non-sentinel lymph node disease. Ann Surg Oncol 17:2909–2919 Cserni G, Gregori D, Merletti F et al, 2004, Non-sentinel node metastases associated with micrometastatic sentinel nodes in breast cancer: metaanalysis of 25 studies. Br J Surg 91:1245–1252 Van Deurzen CH, de Boer M, Monninkhof EM et al, 2008, Nonsentinel lymph node metastases associated with isolated breast cancer cells in the sentinel node. J Natl Cancer Inst 100:1574–1580 Giuliano AE, Dale PS, Turner RR, Morton DL, Evans SW, Krasne DL, 1995, Improved axillary staging of breast cancer with sentinel lymphadenectomy. Ann Surg 222:394–401 Cserni G, Amendoeira I, Apostolikas N et al, 2003, Pathological work-up of sentinel lymph nodes in breast cancer. Review of current data to be considered for the formulation of guidelines. Eur J Cancer 39:1654–1667 Hermanek P, Hutter RV, Sobin LH, Wittekind C, 1999, International Union Against Cancer. Classification of isolated tumor cells and micrometastasis. Cancer 86:2668–2673 Sobin LH, Wittekind C, eds,, 2002, UICC TNM classification of malignant tumours, 6th edn. Wiley, New York Greene FL, Page DL, Fleming ID et al, eds,, 2002, AJCC cancer staging handbook – TNM classification of malignant tumors, 6th edn. Springer Verlag, New York Lyman GH, Giuliano AE, Somerfield MR et al, 2005, American Society of Clinical Oncology guideline recommendations for sentinel lymph node biopsy in early-stage breast cancer. J Clin Oncol 23:7703–7720 Goldhirsch A, Ingle JN, Gelber RD et al, 2009, Thresholds for therapies: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2009. Ann Oncol 20:1319–1329 Goldhirsch A, Wood WC, Coates AS et al, 2011, Strategies for subtypes — dealing with the diversity of breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol 22:1736–1747 Bilimoria KY, Bentrem DJ, Hansen NM et al, 2009, Comparison of sentinel lymph node biopsy alone and completion axillary lymph node dissection for node-positive breast cancer. J Clin Oncol 27:2946–2953 Lazar G, Besznyak I, Boross G et al, 2010, Modern surgical treatment of breast cancer — 2nd Consensus Conference. Magy Seb 63:132–140 Cserni G, Bianchi S, Boecker W et al, 2005, Improving the reproducibility of diagnosing micrometastases and isolated tumor cells. Cancer 103:358–367 Roberts CA, Beitsch PD, Litz CE et al, 2003, Interpretative disparity among pathologists in breast sentinel lymph node evaluation. Am J Surg 186:324–329 De Mascarel I, MacGrogan G, Debled M, Brouste V, Mauriac L, 2008, Distinction between isolated tumor cells and micrometastases in breast cancer: is it reliable and useful? Cancer 112:1672–1678 Turner RR, Weaver DL, Cserni G et al, 2008, Nodal stage classification for breast carcinoma: improving interobserver reproducibility through standardized histologic criteria and image-based training. J Clin Oncol 26:258–263 Sobin L, Gospodarowicz M, Wittekind C, eds,, 2009, UICC TNM classification of malignant tumours, 7th edn. Wiley, New York Edge SB, Byrd DR, Compton CC et al, eds,, 2009, AJCC Cancer Staging Handbook: From the AJCC Cancer Staging Manual. Springer, New York Houvenaeghel G, Nos C, Giard S et al, 2009, A nomogram predictive of non-sentinel lymph node involvement in breast cancer patients with a sentinel lymph node micrometastasis. Eur J Surg Oncol 35:690–695 Cserni G, Bori R, Sejben I et al, 2009, Analysis of predictive tools for further axillary involvement in patients with sentinel-lymphnode- positive, small, < or 015 mm, invasive breast cancer. Orv Hetil 150:2182–2188 Salhab M, Patani N, Mokbel K, 2011, Sentinel lymph node micrometastasis in human breast cancer: an update. Surg Oncol 20: e195–e206 Meretoja TJ, Strien L, Heikkila PS, Leidenius MH, 2012, A simple nomogram to evaluate the risk of nonsentinel node metastases in breast cancer patients with minimal sentinel node involvement. Ann Surg Oncol 19:567–576 Houvenaeghel G, Bannier M, Nos C et al, 2012, Non sentinel node involvement prediction for sentinel node micrometastases in breast cancer: nomogram validation and comparison with other models. Breast 21:204–209 Cserni G, Boross G, Maraz R et al, 2012, Multicentre validation of different predictive tools of non-sentinel lymph node involvement in breast cancer. Surg Oncol 21:59–65 Cserni G, Bori R, Maraz R et al, 2012, Multi-institutional comparison of non-sentinel lymph node predictive tools in breast cancer patients with high predicted risk of further axillary metastasis. Pathol Oncol Res., DOI 10.1007/s12253-012-9553-5 Yi M, Giordano SH, Meric-Bernstam F et al, 2010, Trends in and outcomes from sentinel lymph node biopsy, SLNB, alone vs SLNB with axillary lymph node dissection for node-positive breast cancer patients: experience from the SEER database. Ann Surg Oncol 17(Suppl 3):343–351 Giuliano AE, McCall L, Beitsch P et al, 2010, Locoregional recurrence after sentinel lymph node dissection with or without axillary dissection in patients with sentinel lymph node metastasis. Ann Surg 252:426–433 Kahan Z, Varga K, Dudas R, Nyari T, Thurzo L, 2006, Collaborative/ active participation per se does not decrease anxiety in breast cancer. Pathol Oncol Res 12:93–101 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 211 EP 216 DI 10.1007/s12253-012-9571-3 PG 6 ER PT J AU Candelier, JJ Frappart, L Yadaden, T Poaty, H Picard, JY Prevot, S Coullin, P AF Candelier, Jean-Jacques Frappart, Lucien Yadaden, Tarik Poaty, Henriette Picard, Jean-Yves Prevot, Sophie Coullin, Philippe TI Altered p16 and Bcl-2 Expression Reflects Pathologic Development in Hydatidiform Moles and Choriocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Trophoblast; Uterine cancer; Differentiation; p16; Bcl-2 ID Trophoblast; Uterine cancer; Differentiation; p16; Bcl-2 AB Abnormal trophoblast differentiation is the main cause of gestational trophoblast diseases in the case of hydatidiform moles and choriocarcinomas. Here we investigated the expression patterns of two gene products, p16 and Bcl-2, implicated in cell cycle regulation and apoptosis, respectively, using immunohistochemistry during normal placenta differentiation, hydatidiform moles (partial, complete and invasive) and post-molar choriocarcinomas. The p16 protein shows a gradual expression in cytotrophoblast of normal villous, from a p16 weak proliferative phenotype to a p16 strong invasive phenotype reaching a maximum around 17 weeks of gestation. The expression pattern in cytotrophoblast was similar in moles in contrast to the villous mesenchyme of invasive moles where p16 was strongly expressed. Bcl-2 expression was syncytiotrophoblast specific in normal placenta and moles and increased gradually during normal differentiation. The results explain the persistence of normal and molar villous fragments during their development and their dramatic invasion in the uterine arteries in case of invasive moles. In choriocarcinomas the weak Bcl-2 expression is associated with weak p16 expression indicating a great apoptotic and proliferative potentials. The results suggest that strong p16 expression in the villous mesenchyme may be responsible in part of the morbidity of the moles, and the key of cancer progression in the choriocarcinomas would be a fast cell-cycle turnover. C1 [Candelier, Jean-Jacques] INSERM U782 Endocrinologie et genetique de la reproduction et du developpement, 32 rue des Carnets, 92140 Clamart, France. [Frappart, Lucien] Hopital E. Herriot, Service central d’anatomie et de cytologie pathologiques, place d’Arsonval, 69437 Lyon, France. [Yadaden, Tarik] Centre Hospitalier, Service d’anatomie et de cytologie pathologiques, 194, avenue Rubillard, 72037 Le Mans, France. [Poaty, Henriette] INSERM U782 Endocrinologie et genetique de la reproduction et du developpement, 32 rue des Carnets, 92140 Clamart, France. [Picard, Jean-Yves] INSERM U782 Endocrinologie et genetique de la reproduction et du developpement, 32 rue des Carnets, 92140 Clamart, France. [Prevot, Sophie] Hopital A. Beclere, Service d’anatomie et de cytologie pathologiques, 157 rue de la Porte de Trivaux, 92141 Clamart, France. [Coullin, Philippe] INSERM U782 Endocrinologie et genetique de la reproduction et du developpement, 32 rue des Carnets, 92140 Clamart, France. RP Candelier, JJ (reprint author), INSERM U782 Endocrinologie et genetique de la reproduction et du developpement, 92140 Clamart, France. EM jean-jacques.candelier@u-psud.fr CR Goffin F, Munaut C, Malassine A, Evain-Brion D, Frankenne F, Fridman V, Dubois M, Uzan S, Merviel P, Foidart JM, 2003, Evidence of a limited contribution of feto-maternal interactions to trophoblast differentiation along the invasive pathway. Tissue Antigens 62:104–116 Gruslin A, Qiu Q, Tsang BK, 2001, X-linked inhibitor of apoptosis protein expression and the regulation of apoptosis during human placental development. Biol Reprod 64:1264–1272 Harris LK, Keogh RJ, Wareing M, Baker PN, Cartwright JE, Aplin JD, Whitley GS, 2006, Invasive trophoblasts stimulate vascular smooth muscle cell apoptosis by a fas ligand-dependent mechanism. Am J Pathol 169:1863–1874 Genbacev O, McMaster MT, Fisher SJ, 2000, A repertoire of cell cycle regulators whose expression is coordinated with human cytotrophoblast differentiation. Am J Pathol 157:1337–1351 Kamb A, Gruis NA, Weaver-Feldhaus J, Liu Q, Harshman K, Tavtigian SV, Stockert E, Day RS III, Johnson BE, Skolnick MH, 1994, A cell cycle regulator potentially involved in genesis of many tumor types. Science 264:436–440 Yang H, Xie Y, Yang R, Wei SL, Xi Q, 2008, Expression of p16, INK4a, in mouse endometrium and its effect during blastocyst implantation. Sheng Li Xue Bao 60:547–552 Duan J, Zhang Z, Tong T, 2001, Senescence delay of human diploid fibroblast induced by anti-sense p16INK4a expression. J Biol Chem 276:48325–48331 Ignatov A, Bischoff J, Schwarzenau C, Krebs T, Kuester D, Herrmann K, Costa SD, Roessner A, SemczukA, Schneider-Stock R, 2008, P16 alterations increase the metastatic potential of endometrial carcinoma. Gynecol Oncol 111:365–371 AhmedMN, Kim K, Haddad B, Berchuck A, QumsiyehMB, 2000, Comparative genomic hybridization studies in hydatidiform moles and choriocarcinoma: amplification of 7q21-q31 and loss of 8p12-p21 in choriocarcinoma. Cancer Genet Cytogenet 116:10–15 Xue WC, Chan KY, Feng HC, Chiu PM, Ngan HY, Tsao SW, Cheung AN, 2004, Promoter hypermethylation of multiple genes in hydatidiform mole and choriocarcinoma. J Mol Diagn 6:326– 334 Smith SC, Baker PN, Symonds EM, 1997, Placental apoptosis in normal human pregnancy. Am J Obstet Gynecol 177:57–65 Kim CJ, Choe YJ, Yoon BH, Kim CW, Chi JG, 1995, Patterns of bcl-2 expression in placenta. Pathol Res Pract 191:1239–1244 Huppertz B, Kadyrov M, Kingdom JC, 2006, Apoptosis and its role in the trophoblast. Am J Obstet Gynecol 195:29–39 Danihel L, Gomolcak P, Korbel M, Pruzinec J, Vojtassak J, Janik P, Babal P, 2002, Expression of proliferation and apoptotic markers in human placenta during pregnancy. Acta Histochem 104:335– 338 Danihel L, Porubsky J, Vojtassak J, Breitenecker G, 1994, Trophoblastic disease. II. Immunohistochemical and cytogenetic parameters of partial hydatidiform moles. Cesk Patol 30:80–84 Danihel L, Porubsky J, Zaviacic T, Vojtassak J, Breitenecker G, 1994, Trophoblastic disease. I. Use of immunohistochemistry in the diagnosis of complete hydatidiform moles. Cesk Patol 30:76– 79 Maruo T, Ishihara N, Samoto T, Murakoshi H, Laoag-Fernandez JB, Matsuo H, 2001, Regulation of human trophoblast proliferation and apoptosis during pregnancy. Early Pregnancy 5:28–29 Murakoshi H, Matsuo H, Laoag-Fernandez JB, Samoto T, Maruo T, 2003, Expression of Fas/Fas-ligand, Bcl-2 protein and apoptosis in extravillous trophoblast along invasion to the decidua in human term placenta. Endocr J 50:199–207 Pegoraro L, Palumbo A, Erikson J, Falda M, Giovanazzo B, Emanuel BS, Rovera G, Nowell PC, Croce CM, 1984, A 14;18 and an 8;14 chromosome translocation in a cell line derived from an acute B-cell leukemia. Proc Natl Acad Sci U S A 81:7166–7170 Tsujimoto Y, Finger LR, Yunis J, Nowell PC, Croce CM, 1984, Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18, chromosome translocation. Science 226:1097–1099 Reed JC, 1998, Bcl-2 family proteins. Oncogene 17:3225–3236 Seckl MJ, Fisher RA, Salerno G, Rees H, Paradinas FJ, Foskett M, Newlands ES, 2000, Choriocarcinoma and partial hydatidiform moles. Lancet 356:36–39 Philippe E, Boue J, Boue A, 1980, Trophoblastic gestational diseases. Triploid syndrome, perivillous trophoblastic hyperplasia, trophoblastic pseudotumor, trophoblastic microcarcinoma and carcinoma. Ann Anat Pathol, Paris, 25:13–38 Jacobs PA, Szulman AE, Funkhouser J, Matsuura JS, Wilson CC, 1982, Human triploidy: relationship between parental origin of the additional haploid complement and development of partial hydatidiform mole. Ann Hum Genet 46:223–231 Bifulco C, Johnson C, Hao L, Kermalli H, Bell S, Hui P, 2008, Genotypic analysis of hydatidiform mole: an accurate and practical method of diagnosis. Am J Surg Pathol 32:445–451 Couillin P, Ravise N, Afoutou JM, Chaibi R, Azoulay M, Hors J, Oury JF, Boue J, Boue A, 1987, HLA and molar pregnancies, triploidies, hydatidiform moles and choriocarcinoma). Etiological and epidemiological study. Ann Genet 30:197–208 Jacobs PA, Hunt PA, Matsuura JS, Wilson CC, Szulman AE, 1982, Complete and partial hydatidiform mole in Hawaii: cytogenetics, morphology and epidemiology. Br J Obstet Gynaecol 89:258–266 Couillin P, Afoutou JM, Faye O, Ravise N, Correa P, Boue A, 1985, Androgenetic origin of African complete hydatidiform moles demonstrated by HLA markers. Hum Genet 71:113–116 Steigrad SJ, 2003, Epidemiology of gestational trophoblastic diseases. Best Pract Res Clin Obstet Gynaecol 17:837–847 Berkowitz RS, Goldstein DP, 2009, Current management of gestational trophoblastic diseases. Gynecol Oncol 112:654–662 Matsui H, Iitsuka Y, Yamazawa K, Tanaka N, Seki K, Sekiya S, 2003, Changes in the incidence of molar pregnancies. A population-based study in Chiba Prefecture and Japan between 1974 and 2000. Hum Reprod 18:172–175 El-Maarri O, Seoud M, Coullin P, Herbiniaux U, Oldenburg J, Rouleau G, Slim R, 2003, Maternal alleles acquiring paternal methylation patterns in biparental complete hydatidiform moles. Hum Mol Genet 12:1405–1413 Deveault C, Qian JH, ChebaroW, Ao A, Gilbert L, Mehio A, Khan R, Tan SL, Wischmeijer A, Coullin P, Xie X, Slim R, 2009, NLRP7 mutations in women with diploid androgenetic and triploid moles: a proposed mechanism for mole formation. Hum Mol Genet 18:888–897 Ngan HY, Bender H, Benedet JL, Jones H,Montruccoli GC, Pecorelli S, 2003, Gestational trophoblastic neoplasia, FIGO 2000 staging and classification. Int J Gynaecol Obstet 83(Suppl 1):175–177 Latour P, Boutrand L, Levy N, Bernard R, Boyer A, Claustrat F, Chazot G, Boucherat M, Vandenberghe A, 2001, Polymorphic short tandem repeats for diagnosis of the Charcot-Marie-Tooth 1A duplication. Clin Chem 47:829–837 Noguer-Dance M, Abu-Amero S, Al-Khtib M, Lefevre A, Coullin P, Moore GE, Cavaille J, 2010, The primate-specific microRNA gene cluster, C19MC, is imprinted in the placenta. Hum Mol Genet 19:3566–3582 Kar M, Ghosh D, Sengupta J, 2007, Histochemical and morphological examination of proliferation and apoptosis in human first trimester villous trophoblast. Hum Reprod 22:2814–2823 Serrano M, Hannon GJ, Beach D, 1993, A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature 366:704–707 Tsatsaris V, Goffin F, Munaut C, Brichant JF, Pignon MR, Noel A, Schaaps JP, Cabrol D, Frankenne F, Foidart JM, 2003, Overexpression of the soluble vascular endothelial growth factor receptor in preeclamptic patients: pathophysiological consequences. J Clin Endocrinol Metab 88:5555–5563 Herath CB, Ishiwata H, Shiojima S, Kadowaki T, Katsuma S, Ushizawa K, Imai K, Takahashi T, Hirasawa A, Takahashi S, Izaike Y, Tsujimoto G, Hashizume K, 2006, Developmental aberrations of liver gene expression in bovine fetuses derived from somatic cell nuclear transplantation. Cloning Stem Cells 8:79–95 Bozec A, Bakiri L, Hoebertz A, Eferl R, Schilling AF, Komnenovic V, ScheuchH, PriemelM, Stewart CL,Amling M,Wagner EF, 2008, Osteoclast size is controlled by Fra-2 through LIF/LIF-receptor signalling and hypoxia. Nature 454:221–225 Kim KR, Park BH, Hong YO, Kwon HC, Robboy SJ, 2009, The villous stromal constituents of complete hydatidiform mole differ histologically in very early pregnancy from the normally developing placenta. Am J Surg Pathol 33:176–185 Ishioka S, Ezaka Y, Umemura K, Hayashi T, Endo T, Saito T, 2007, Proteomic analysis of mechanisms of hypoxia-induced apoptosis in trophoblastic cells. Int J Med Sci 4:36–44 Ishihara N, Matsuo H, Murakoshi H, Laoag-Fernandez JB, Samoto T, Maruo T, 2002, Increased apoptosis in the syncytiotrophoblast in human term placentas complicated by either preeclampsia or intrauterine growth retardation. Am J Obstet Gynecol 186:158– 166 Poaty H, Coullin P, Peko JF, Dessen P, Diatta AL, Valent A, Leguern E, Prevot S, Gombe-Mbalawa C, Candelier JJ, Picard JY, Bernheim A, 2012, Genome-wide high-resolution aCGH analysis of gestational choriocarcinomas. PLoS One 7:e29426 Huppertz B, Kadyrov M, Kingdom JC, 2006, Apoptosis and its role in the trophoblast. Am J Obstet Gynecol 195:29–39 Ishihara N, Matsuo H, Murakoshi H, Laoag-Fernandez J, Samoto T, Maruo T, 2000, Changes in proliferative potential, apoptosis and Bcl-2 protein expression in cytotrophoblasts and syncytiotrophoblast in human placenta over the course of pregnancy. Endocr J 47:317–327 Heazell AE, Crocker IP, 2008, Live and let die - regulation of villous trophoblast apoptosis in normal and abnormal pregnancies. Placenta 29:772–783 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 217 EP 227 DI 10.1007/s12253-012-9572-2 PG 11 ER PT J AU Obeidat, RB Matalka, II Mohtaseb, AA Al-Kaisi, SN AF Obeidat, R Basil Matalka, I Ismail Mohtaseb, A Alia Al-Kaisi, S Nabih TI Selected Immuno-Histochemical Markers in Curettage Specimens and their Correlation with Final Pathologic Findings in Endometrial Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Estrogen receptor; Progesterone receptor; p53; Ki-67; Her2/neu; Endometrial cancer ID Estrogen receptor; Progesterone receptor; p53; Ki-67; Her2/neu; Endometrial cancer AB To assess the immuno-histochemical expression of various markers in, endometrial biopsies of patients with endometrial cancer, and to correlate their expression with the final pathologic findings. Sixty-two patients with primary endometrial cancer who underwent surgical treatment were included in this study. Immuno-histochemical expression of estrogen receptor (ER), progesterone receptor (PR), p53, bcl-2, Her-2/neu and Ki-67 were assessed in curettage specimens, and review of the final pathology report from hysterectomy specimens was carried out. The expression of these markers in curettage was correlated with the final tumor characteristics obtained on hysterectomy specimens. Both ER and PR were significantly more expressed in endometrioid type (EC) than nonendometrioid type (NEC) (P value of 0.004 and 0.012). On the contrary, P53, Her-2 and Ki-67 showed higher positivity in NEC than EC (P value of 0.005, 0.025 and 0.002). Positive expression of ER and PR was significantly associated with low grade tumors and superficial myometrial invasion, whereas positive expression of Her-2 and Ki-67 was significantly associated with higher grade lesions, and deep myometrial invasion. Moreover, a statistically significant inverse relationship was observed between the positivity of P53, Her-2 and Ki-67 and the positivity of ER, PR. We found that determination of immuno-histochemical markers in curettage specimens might be helpful in predicting the final pathologic findings in patients with endometrial cancer. This might be helpful in planning the extensivity of the surgery. C1 [Obeidat, R Basil] Jordan University of Science and Technology, Department of Obstetrics and Gynecology, 21110 Irbid, Jordan. [Matalka, I Ismail] Jordan University of Science and Technology, Department of Pathology and Laboratory MedicineIrbid, Jordan. [Mohtaseb, A Alia] Jordan University of Science and Technology, Department of Pathology and Laboratory MedicineIrbid, Jordan. [Al-Kaisi, S Nabih] King Hussein Cancer Center, Department of PathologyAmman, Jordan. RP Obeidat, RB (reprint author), Jordan University of Science and Technology, Department of Obstetrics and Gynecology, 21110 Irbid, Jordan. EM b_obeidat@hotmail.com CR Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics. CA Cancer J Clin 59(4):225–249 Bokhman JV, 1983, Two pathogenetic types of endometrial carcinoma. Gynecol Oncol 15(1):10–17 Announcements, 1989, FIGO stages-1988 revision. Gynecol Oncol 35:125–127 Yokoyama Y, Maruyama H, Sato S et al, 1997, Indispensability of pelvic and paraaortic lymphadenectomy in endometrial cancers. Gynecol Oncol 64:411–417 Mariani A, Keeney GL, Aletti G et al, 2004, Endometrial carcinoma: paraaortic dissemination. Gynecol Oncol 92:833–838 Kinkel K, Kaji Y, Yu KK et al, 1999, Radiologic staging in patients with endometrial cancer: a meta-analysis. Radiology 212:711–718 Larson DM, Connor GP, Broste SK, Krawisz BR, Johnson KK, 1996, Prognostic significance of gross myometrial invasion with endometrial cancer. Obstet Gynecol 88:396–398 Zorlu CG, Kuscu E, Ergun Y, Aydogdu T, Cobanoglu O, Erdas O, 1993, Intraoperative evaluation of prognostic factors in stage I endometrial cancer by frozen section: how reliable? Acta Obstet Gynecol Scand 72:382–385 Sanjuan A, Cobo T, Pahisa J, Escaramis G, Ordi J, Ayuso JR, Garcia S, Hernandez S, Torne A, Martinez Roman S, Lejarcegui JA, Vanrell JA, 2006, Preoperative and intraoperative assessment of myometrial invasion and histologic grade in endometrial cancer: role of magnetic resonance imaging and frozen section. Int J Gynecol Cancer 16(1):385–390 Vorgias G, Hintipas E, Katsoulis M, Kalinoglou N, Dertimas B, Akrivos T, 2002, Intraoperative gross examination of myometrial invasion and cervical infiltration in patients with endometrial cancer: decision-making accuracy. Gynecol Oncol 85:483–486 Geisinger KR, Marschall RB, Kute TE, Homesley HD, 1986, Correlation of female sex steroid hormone receptors with histologic and ultrastructural differentiation in adenocarcinoma of endometrium. Cancer 58:1506–1517 Kleine W, Maier T, Geyer H, Phleiderer A, 1990, Estrogen and progesterone receptors in endometrial cancer and their prognostic relevance. Gynecol Oncol 38:59–65 Creasman WT, Soper JT, McCarty KS Jr, McCarty KS Sr, Hinshaw W, Clarke-Pearson DL, 1985, Influence of cytoplasmic steroid receptor content on prognosis of early stage endometrial carcinoma. Am J Obstet Gynecol 151:922–932 Ferrandina G, Ranelletti FO, Gallotta Vet al, 2005, Expression of cyclooxygenase-2, COX-2), receptors for estrogen, ER, and progesterone, PR), p53, Ki-67, and neu protein in endometrial cancer. Gynecol Oncol 98:383–389 Halperin R, Zehavi S, Habler L et al, 2001, Comparative immunohistochemical study of endometrioid and serous papillary carcinoma of endometrium. Eur J Gynaecol Oncol 22:122–126 Kadar N, Malfetano JH, Homesley HD, 1993, Steroid receptor concentrations in endometrial carcinoma: effect on survival in surgically staged patients. Gynecol Oncol 50:281–286 Morris PC, Anderson JR, Anderson B et al, 1995, Steroid hormone receptor content and lymph node status in endometrial cancer. Gynecol Oncol 56:406–411 Geisler JP, Wiemann MC, Zhou Z et al, 1996, p53 as a prognostic indicator in endometrial cancer. Gynecol Oncol 61:245–248 Cherchi PL, Marras V, Capobianco G et al, 2001, Prognostic value of p53, c-erb-B2 and MIB-1 in endometrial carcinoma. Eur J Gynaecol Oncol 22:451–453 Lax SF, Pizer ES, Ronnett BM et al, 1998, Clear cell carcinoma of the endometrium is characterized by adistinctive profile of p53, Ki- 67, estrogen, and progesterone receptor expression. Hum Pathol 29:551–558 Ohkouchi T, Sakuragi N, Watari H et al, 2002, Prognostic significance of Bcl-2, p53 overexpression, and lymph node metastasis in surgically staged endometrial carcinoma. Am J Obstet Gynecol 187:353–359 Lukes AS, Kohler MF, Pieper CF et al, 1994, Multivariable analysis of DNA ploidy, p53, and HER-2/neu as prognostic factors in endometrial cancer. Cancer 73:2380–2385 Mariani A, Sebo TJ, Webb MJ et al, 2003, Molecular and histopathologic predictors of distant failure in endometrial cancer. Cancer Detect Prev 27:434–441 Salvesen HB, Iversen OE, Akslen LA, 1998, Identification of high-risk patients by assessment of nuclear Ki-67 expression in a prospective study of endometrial carcinomas. Clin Cancer Res 4:2779–2785 Salvesen HB, Iversen OE, Akslen LA, 1999, Prognostic significance of angiogenesis and Ki-67, p53, and p21 expression: a population-based endometrial carcinoma study. J Clin Oncol 17:1382–1390 Erdem O, Erdem M, Dursum A et al, 2003, Angiogenesis, p53 and bcl-2 expression as prognostic indicators in endometrial cancer: comparison with traditional clinicopathological variables. Int J Gynecol Pathol 22:254–260 Sakuragi N, Ohkouchi T, Hareyama H et al, 1998, Bcl-2 expression and prognosis of patients with endometrial carcinoma. Int J Cancer 79:153–158 Kounelis S, Kapranos N, Kouri E, Coppola D, Papadaki H, Jones MW, 2000, Immunohistochemical profile of endometrial adenocarcinoma: a study of 61 cases and review of the literature. Mod Pathol 13(4):379–388 Appel MLM, Edelweis MI, Fleck J et al, 2008, p53 and bcl-2 as prognostic markers in endometrial carcinoma. Pathol Oncol Res 14:23–30 Markova I, Duskova M, Lubusky M, Kudela M, Zapletalova J, Prochazka M, Pilka R, 2010, Selected immunohistochemical prognostic factors in endometrial cancer. Int J Gynecol Cancer 20, 4):576–582 Konecny GE, Agarwal R, Keeney GA, Winterhoff B, Jones MB, Mariani A, Riehle D, Neuper C, Dowdy SC, Wang HJ, Morin PJ, Podratz KC, 2008, Claudin-3 and claudin-4 expression in serous papillary, clear-cell, and endometrioid endometrial cancer. Gynecol Oncol 109(2):263–269 Sobel G, Nemeth J, Kiss A, Lotz G, Szabo I, Udvarhelyi N, Schaff Z, Paska C, 2006, Claudin 1 differentiates endometrioid and serous papillary endometrial adenocarcinoma. Gynecol Oncol 103(2):591–598 Dupont J, Wang X, Marshall DS, Leitao M, Hedvat CV, Hummer A et al, 2004, Wilms Tumor Gene, WT1, and p53 expression in endometrial carcinomas: a study of 130 cases using a tissue microarray. Gynecol Oncol 94(2):449–455 Ohno S, Dohi S, Ohno Y, Kyo S, Sugiyama H, Suzuki N, Inoue M, 2009, Immunohistochemical detection of WT1 protein in endometrial cancer. Anticancer Res 29(5):1691–1695 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 229 EP 235 DI 10.1007/s12253-012-9573-1 PG 7 ER PT J AU Ji, YI Lee, BY Kang, YJ Jo, JO Lee, HS Kim, YH Kim, YO Lee, Ch Koh, BS Kim, A Lee, YJ Jung, HM Ock, SM Cha, HJ AF Ji, Yong-Il Lee, Bo-Young Kang, Yun-Jeong Jo, Jin-Ok Lee, Ho Sang Kim, Yeol Heung Kim, Young-Ok Lee, Chulmin Koh, Bong Suk Kim, Ari Lee, Young Ji Jung, Hyung Min Ock, Sun Mee Cha, Hee-Jae TI Expression Patterns of Thymosin β4 and Cancer Stem Cell Marker CD133 in Ovarian Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thymosin β4; CD133; Ovarian cancer; Metastasis; Cancer stem cells; Stomach cancer ID Thymosin β4; CD133; Ovarian cancer; Metastasis; Cancer stem cells; Stomach cancer AB Thymosin β4 (Tβ4), a small acidic actin binding peptide, is overexpressed in a side population of cancer stem cells and CD133-positive colorectal cancer stem cells. In order to understand the relationship between Tβ4 and CD133, we studied the expression patterns of Tβ4 and CD133 in ovarian cancers. The expression patterns of Tβ4 and CD133 were studied in normal ovaries, primary ovarian cancers, metastatic ovarian cancers, primary stomach cancers, and normal stomachs by Western blot and immunohistochemistry. Expression patterns and co-localization of Tβ4 and CD133 were examined by immunofluorescence and confocal laser-scanning microscopy. Tβ4 is overexpressed in primary ovarian cancers, but not in primary stomach cancers, when compared with normal controls. However, Tβ4 levels in metastatic stomach cancers to the ovary are significantly upregulated compared with levels in normal stomachs and primary stomach cancers. These results suggest that Tβ4 levels are related to tumorigenesis in ovarian cancers and metastasis in stomach cancers. The expression of Tβ4 in normal ovaries and normal stomachs was weak, but was colocalized with CD133 expression. Tβ4 expression was also co-localized with CD133 expression in primary ovarian carcinomas, metastatic ovarian cancers from stomach cancers and primary stomach cancers. These data suggest that Tβ4 expression is strongly related to CD133 expression and is a characteristic of stem cells or cancer stem cells. C1 [Ji, Yong-Il] Kosin University College of Medicine, Department of Obstetrics and GynecologyBusan, South Korea. [Lee, Bo-Young] Kosin University College of Medicine, Department of Parasitology and Genetics, 34, Annam-dong, Seo-gu, 602-703 Busan, South Korea. [Kang, Yun-Jeong] Kosin University College of Medicine, Department of Parasitology and Genetics, 34, Annam-dong, Seo-gu, 602-703 Busan, South Korea. [Jo, Jin-Ok] Kosin University College of Medicine, Department of Parasitology and Genetics, 34, Annam-dong, Seo-gu, 602-703 Busan, South Korea. [Lee, Ho Sang] Kosin University, Department of SurgeryBusan, South Korea. [Kim, Yeol Heung] Kosin University College of Medicine, Department of Obstetrics and GynecologyBusan, South Korea. [Kim, Young-Ok] Kosin University College of Medicine, Department of PathologyBusan, South Korea. [Lee, Chulmin] Inje university, Haeunade Paik Hospital, Department of Obstetrics and GynecologyBusan, South Korea. [Koh, Bong Suk] Inje university, Sanggye Paik Hospital, Department of Obstetrics and GynecologySeoul, South Korea. [Kim, Ari] Catholic University of Daegu, School of Medicine, Departments of Obstetrics and GynecologyDaegu, South Korea. [Lee, Young Ji] Wonkwang University, College of Medicine, Department of Obstetrics and GynecologyIksan, South Korea. [Jung, Hyung Min] Konkuk University, School of Medicine, Department of Obstetrics and GynecologySeoul, South Korea. [Ock, Sun Mee] Kosin University College of Medicine, Department of Parasitology and Genetics, 34, Annam-dong, Seo-gu, 602-703 Busan, South Korea. [Cha, Hee-Jae] Kosin University College of Medicine, Department of Parasitology and Genetics, 34, Annam-dong, Seo-gu, 602-703 Busan, South Korea. RP Cha, HJ (reprint author), Kosin University College of Medicine, Department of Parasitology and Genetics, 602-703 Busan, South Korea. EM hcha@kosin.ac.kr CR Low TL, Goldstein AL, 1982, Chemical characterization of thymosin beta 4. J Biol Chem 257:1000–1006 Safer D, Golla R et al, 1990, Isolation of a 5-kilodalton actinsequestering peptide from human blood platelets. Proc Natl Acad Sci U S A 87:2536–2540 Safer D, Elzinga M et al, 1991, Thymosin beta 4 and Fx, an actinsequestering peptide, are indistinguishable. J Biol Chem 266:4029–4032 Cassimeris L, Safer D et al, 1992, Thymosin beta 4 sequesters the majority of G-actin in resting human polymorphonuclear leukocytes. J Cell Biol 119:1261–1270 Van Troys M, Dewitte D et al, 1996, The actin binding site of thymosin beta 4 mapped by mutational analysis. EMBO J 15:201–210 Grant DS, RoseWet al, 1999, Thymosin beta4 enhances endothelial cell differentiation and angiogenesis. Angiogenesis 3:125–135 Malinda KM, Goldstein AL et al, 1997, Thymosin beta 4 stimulates directional migration of human umbilical vein endothelial cells. FASEB J 11:474–481 Malinda KM, Sidhu GS et al, 1999, Thymosin beta4 accelerates wound healing. J Invest Dermatol 113:364–368 Philp D, Nguyen M et al, 2004, Thymosin beta4 increases hair growth by activation of hair follicle stem cells. FASEB J 18:385–387 Philp D, Goldstein AL et al, 2004, Thymosin beta4 promotes angiogenesis, wound healing, and hair follicle development. Mech Ageing Dev 125:113–115 Philp D, St-Surin S et al, 2007, Thymosin beta 4 induces hair growth via stem cell migration and differentiation. Ann N Y Acad Sci 1112:95–103 Hall AK, 1994, Molecular interactions between G-actin, DNase I and the beta-thymosins in apoptosis: a hypothesis. Med Hypotheses 43:125–131 Iguchi K, Usami Y et al, 1999, Decreased thymosin beta4 in apoptosis induced by a variety of antitumor drugs. Biochem Pharmacol 57:1105–1111 Niu M, Nachmias VT, 2000, Increased resistance to apoptosis in cells overexpressing thymosin beta four: a role for focal adhesion kinase pp125FAK. Cell Adhes Commun 7:311–320 Moon EY, Song JH et al, 2007, Actin-sequestering protein, thymosin-beta-4, TB4), inhibits caspase-3 activation in paclitaxel-induced tumor cell death. Oncol Res 16:507–516 Young JD, Lawrence AJ et al, 1999, Thymosin beta 4 sulfoxide is an anti-inflammatory agent generated by monocytes in the presence of glucocorticoids. Nat Med 5:1424–1427 Sosne G, Qiu P et al, 2007, Thymosin beta 4 suppression of corneal NFkappaB: a potential anti-inflammatory pathway. Exp Eye Res 84:663–669 Larsson LI, Holck S, 2007, Occurrence of thymosin beta4 in human breast cancer cells and in other cell types of the tumor microenvironment. Hum Pathol 38:114–119 Santelli G, Califano D et al, 1999, Thymosin beta-10 gene overexpression is a general event in human carcinogenesis. Am J Pathol 155:799–804 Hall AK, 1991, Differential expression of thymosin genes in human tumors and in the developing human kidney. Int J Cancer 48:672–677 Ji P, Diederichs S et al, 2003, MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene 22:8031–8041 Wang WS, Chen PM et al, 2004, Overexpression of the thymosin beta-4 gene is associated with increased invasion of SW480 colon carcinoma cells and the distant metastasis of human colorectal carcinoma. Oncogene 23:6666–6671 Kobayashi T, Okada F et al, 2002, Thymosin-beta4 regulates motility and metastasis of malignant mouse fibrosarcoma cells. Am J Pathol 160:869–882 Cha HJ, Jeong MJ et al, 2003, Role of thymosin beta4 in tumor metastasis and angiogenesis. J Natl Cancer Inst 95:1674–1680 Huang HC, Hu CH et al, 2007, Thymosin beta4 triggers an epithelial-mesenchymal transition in colorectal carcinoma by upregulating integrin-linked kinase. Oncogene 26:2781–2790 Ricci-Vitiani L, Pagliuca A et al, 2008, Colon cancer stem cells. Gut 57:538–548 Kondo T, Setoguchi T et al, 2004, Persistence of a small subpopulation of cancer stem-like cells in the C6 glioma cell line. Proc Natl Acad Sci U S A 101:781–786 Ricci-Vitiani L, Lombardi DG et al, 2007, Identification and expansion of human colon-cancer-initiating cells. Nature 445:111–115 Huang EH, Hynes MJ et al, 2009, Aldehyde dehydrogenase 1 is a marker for normal and malignant human colonic stem cells, SC, and tracks SC overpopulation during colon tumorigenesis. Cancer Res 69:3382–3389 Liu G, Yuan X et al, 2006, Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer 5:67 Shmelkov SV, Butler JM et al, 2008, CD133 expression is not restricted to stem cells, and both CD133+ and CD133- metastatic colon cancer cells initiate tumors. J Clin Invest 118:2111–2120 Miraglia S, Godfrey W et al, 1997, A novel five-transmembrane hematopoietic stem cell antigen: isolation, characterization, and molecular cloning. Blood 90:5013–5021 Yin AH, Miraglia S et al, 1997, AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood 90:5002–5012 Singh SK, Clarke ID et al, 2003, Identification of a cancer stem cell in human brain tumors. Cancer Res 63:5821–5828 Singh SK, Hawkins C et al, 2004, Identification of human brain tumour initiating cells. Nature 432:396–401 Collins AT, Berry PA et al, 2005, Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 65:10946– 10951 Suetsugu A, Nagaki M et al, 2006, Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells. Biochem Biophys Res Commun 351:820–824 Yin S, Li J et al, 2007, CD133 positive hepatocellular carcinoma cells possess high capacity for tumorigenicity. Int J Cancer 120:1444–1450 Hermann PC, Huber SL et al, 2007, Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1:313–323 Eramo A, Lotti F et al, 2008, Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ 15:504–514 O’Brien CA, Pollett A et al, 2007, A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445:106–110 Todaro M, Alea MP et al, 2007, Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4. Cell Stem Cell 1:389–402 Steiniger SC, Coppinger JA et al, 2008, Quantitative mass spectrometry identifies drug targets in cancer stem cell-containing side population. Stem Cells 26:3037–3046 Ricci-Vitiani L, Mollinari C et al, 2010, Thymosin beta4 targeting impairs tumorigenic activity of colon cancer stem cells. FASEB J 24:4291–4301 Piek JM, van Diest PJ et al, 2008, Ovarian carcinogenesis: an alternative hypothesis. Adv Exp Med Biol 622:79–87 Bridda A, Padoan I et al, 2007, Peritoneal mesothelioma: a review. MedGenMed 9:32 Jo JO, Kang YJ et al, 2011, Thymosin beta4 expression in human tissues and in tumors using tissue microarrays. Appl Immunohistochem Mol Morphol 19:160–167 Vermeulen L, Sprick MR et al, 2008, Cancer stem cells–old concepts, new insights. Cell Death Differ 15:947–958 Hollier BG, Evans K et al, 2009, The epithelial-to-mesenchymal transition and cancer stem cells: a coalition against cancer therapies. J Mammary Gland Biol Neoplasia 14:29–43 Wang ZY, Zeng FQ et al, 2010, Evaluation of thymosin beta4 in the regulation of epithelial-mesenchymal transformation in urothelial carcinoma. Urol Oncol 30:167–176 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 237 EP 245 DI 10.1007/s12253-012-9574-0 PG 9 ER PT J AU Aneiros-Fernandez, J Arias-Santiago, S Arias-Santiago, B Herrero-Fernandez, M Carriel, V Aneiros-Cachaza, J Lopez-Valverde, A Cutando-Soriano, A AF Aneiros-Fernandez, Jose Arias-Santiago, Salvador Arias-Santiago, Borja Herrero-Fernandez, Maria Carriel, Victor Aneiros-Cachaza, Jose Lopez-Valverde, Antonio Cutando-Soriano, Antonio TI MT1 Melatonin Receptor Expression in Warthin’s Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Immunohistochemistry; Melatonin; MT1 receptor; Normal parotid gland ID Immunohistochemistry; Melatonin; MT1 receptor; Normal parotid gland AB We contribute the first immunohistochemical study of MT1 melatonin receptor in Warthin’s tumor and normal parotid gland. All 14Warthin’s tumors studied showed intense cytoplasmic positivity for MT1 receptor in all cylindrical epithelial cells lining spaces and a less intense positivity in basal cells. The lymphoid component accompanying the tumor was always negative for MT1 receptor. The parotid structure surrounding the tumor showed intense cytoplasmic positivity in all cells lining excretory ducts (lobar and lobulillar), with a lesser and focal positivity in cells of the acinar component. The biological activity of MT1 receptor in epithelial cells lining parotid excretory ducts may resemble its activity in Warthin’s tumor cells.Hence, we propose Warthin’s tumor as a useful positive control in immunohistochemical studies of MT1 melatonin receptor. C1 [Aneiros-Fernandez, Jose] San Cecilio University Hospital, Department of PathologyGranada, Spain. [Arias-Santiago, Salvador] San Cecilio University Hospital, Department of DermatologyGranada, Spain. [Arias-Santiago, Borja] University of Granada, School of DentistryGranada, Spain. [Herrero-Fernandez, Maria] University of Granada, School of DentistryGranada, Spain. [Carriel, Victor] University of Granada, Tissue Engineering GroupGranada, Spain. [Aneiros-Cachaza, Jose] San Cecilio University Hospital, Department of PathologyGranada, Spain. [Lopez-Valverde, Antonio] University of Salamanca, School of DentistrySalamanca, Spain. [Cutando-Soriano, Antonio] University of Granada, School of DentistryGranada, Spain. RP Cutando-Soriano, A (reprint author), University of Granada, School of Dentistry, Granada, Spain. EM acutando@ugr.es CR Dubocovich ML, Rivera-Bermudez MA, Gerdin MJ et al, 2003, Molecular pharmacology, regulation and function of mammalian melatonin receptors. Front Biosci 8:d1093–d1108 Slaugenhaupt SA, Roca AL, Liebert CB et al, 1995, Mapping of the gene for the Mel1a-melatonin receptor to human chromosome 4, MTNR1A, and mouse chromosome 8, Mtnr1a). Genomics 27:355–357 Reppert SM, Godson C, Mahle CD et al, 1995, Molecular characterization of a second melatonin receptor expressed in human retina and brain: the Mel1b melatonin receptor. Proc Natl Acad Sci U S A 92:8734–8738 Reppert SM, Weaver DR, Godson C, 1996, Melatonin receptors step into the light: cloning and classification of subtypes. Trends Pharmacol Sci 17:100–102 Vanĕcek J, 1988, Melatonin binding sites. J Neurochem 51:1436– 1440 Lissoni P, Tancini G, Barni S, Paolorossi F, Ardizzoia A, Conti A, Maestroni G, 1997, Treatment of cancer chemotherapy-induced toxicity with the pineal hormone melatonin. Support Care Canc 5:126–129 Hunt AE, Al-GhoulWM, Gillette MU et al, 2001, Activation of MT, 2, melatonin receptors in rat suprachiasmatic nucleus phase advances the circadian clock. Am J Physiol Cell Physiol 280:C110–C118 Ekmekcioglu C, 2006, Melatonin receptors in humans: biological role and clinical relevance. Biomed Pharmacother 60:97–108 Rogelsperger O, Ekmekcioglu C, Jager W et al, 2009, Coexpression of the melatonin receptor 1 and nestin in human breast cancer specimens. J Pineal Res 46:422–432 Dillon DC, Easley SE, Asch BB et al, 2002, Differential expression of high-affinity melatonin receptors, MT1, in normal and malignant human breast tissue. Am J Clin Pathol 118:451–458 Toma CD, Svoboda M, Arrich F et al, 2007, Expression of the melatonin receptor, MT, 1 in benign and malignant human bone tumors. J Pineal Res 43:206–213 Girgert R, Hanf V, Emons G et al, 2009, Membrane-bound melatonin MT1 receptor down-regulates estrogen responsive genes in breast cancer cells. J Pineal Res 47:23–31 Yuan L, Collins AR, Dai J et al, 2002, MT(1, melatonin receptor overexpression enhances the growth suppressive effect of melatonin in human breast cancer cells. Mol Cell Endocrinol 192:147–156 Ying SW, Niles LP, Crocker C, 1993, Human malignant melanoma cells express high-affinity receptors for melatonin: antiproliferative effects of melatonin and 6-chloromelatonin. Eur J Pharmacol 246:89–96 Shiu SY, Law IC, Lau KWet al, 2003, Melatonin slowed the early biochemical progression of hormone-refractory prostate cancer in a patient whose prostate tumor tissue expressed MT1 receptor subtype. J Pineal Res 35:177–182 Aust S, Thalhammer T, Humpeler S et al, 2004, The melatonin receptor subtype MT1 is expressed in human gallbladder epithelia. J Pineal Res 36:43–48 Claustrat B, Brun J, Chazot G, 2005, The basic physiology and pathophysiology of melatonin. Sleep Med Rev 9:11–24 Reiter RJ, 1991, Pineal melatonin: cell biology of its synthesis and of its physiological interactions. Endocr Rev 12:151–180 Reiter RJ, 2003, Melatonin: clinical relevance: best practice & research clinical. Endocrinol Metab 17:273–285 Cutando A, Lopez-Valverde A, Arias-Santiago S, DE Vicente J, DE Diego RG, 2012, Role of melatonin in cancer treatment. Anticancer Res 32:2747–2753 Cutando A, Aneiros-Fernandez J, Lopez-Valverde A, Arias- Santiago S, Aneiros-Cachaza J, Reiter RJ, 2011, A new perspective in Oral health: potentialimportance and actions of melatonin receptors MT1, MT2, MT3, and RZR/ROR in the oral cavity. Arch Oral Biol 56:944–950 Cutando A, Aneiros-Fernandez J, Aneiros-Cachaza J, Arias- Santiago S, 2011, Melatonin and cancer: current knowledge and its application to oral cavity tumours. J Oral Pathol Med 40(8):593–597 Bubenik GA, 2002, Gastrointestinal melatonin, localization, function and clinical relevante. Dig Dis Sci 47:2336–2348 Raikhlin NT N, Kvetnoy IM, Tolkachev VN, 1975, Melatonin may be synthesised in enterochromaffin cells. Nature 255:344– 345 Aras HC, Ekstrom J, 2008, Melatonin-evoked in vivo secretion of protein and amylase from the parotid gland of the anaesthetised rat. J Pineal Res 45:413–421 Dubocovich ML, Markowska M, 2005, Functional MT1 and MT2 melatonin receptors in mammals. Endocrine 27:101–110 Sobrinho-Simoes M, Maximo V, 2006, Warthin’s tumour. Virchows Arch 448:877–878 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 247 EP 250 DI 10.1007/s12253-012-9575-z PG 4 ER PT J AU Huang, XB Li, J Zheng, L Zuo, GH Han, KQ Li, HY Liang, P AF Huang, Xiao-Bing Li, Jing Zheng, Lu Zuo, Guo-Hua Han, Ke-Qiang Li, Hong-Yan Liang, Ping TI Bioinformatics Analysis Reveals Potential Candidate Drugs for HCC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Cirrhosis; Sub-pathways; Small molecule drugs ID Hepatocellular carcinoma; Cirrhosis; Sub-pathways; Small molecule drugs AB In our study, we used the GSE17967 series to identify differentially expressed genes between cirrhosis and hepatocellular carcinoma, aiming to analyse the mechanism of the progression of cirrhosis to hepatocellular carcinoma and identify the sub-pathways closely related to this progression, and find the small molecule drugs to interfere this progression. From the result of our study, we find that many small molecule drugs closely related with carcinoma have been linked by our method. We also find some new small molecule drugs related to this progression. It is demonstrated that bioinformatics analysis is useful in identification of the candidate drugs in hepatocellular carcinoma. C1 [Huang, Xiao-Bing] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, NO.2 Xin Qiao Street, Sha Ping Ba District, 400037 Chongqing, China. [Li, Jing] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, NO.2 Xin Qiao Street, Sha Ping Ba District, 400037 Chongqing, China. [Zheng, Lu] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, NO.2 Xin Qiao Street, Sha Ping Ba District, 400037 Chongqing, China. [Zuo, Guo-Hua] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, NO.2 Xin Qiao Street, Sha Ping Ba District, 400037 Chongqing, China. [Han, Ke-Qiang] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, NO.2 Xin Qiao Street, Sha Ping Ba District, 400037 Chongqing, China. [Li, Hong-Yan] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, NO.2 Xin Qiao Street, Sha Ping Ba District, 400037 Chongqing, China. [Liang, Ping] Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, NO.2 Xin Qiao Street, Sha Ping Ba District, 400037 Chongqing, China. RP Liang, P (reprint author), Third Military Medical University, Xinqiao Hospital, Department of Hepatobiliary Surgery, 400037 Chongqing, China. EM PiangLiang2012@hotmail.com CR El-Serag HB, Rudolph KL, 2007, Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 132, 7):2557–2576., DOI 10.1053/j.gastro.2007.04.061 Avila MA, Berasain C, Sangro B, Prieto J, 2006, New therapies for hepatocellular carcinoma. Oncogene 25(27):3866–3884., DOI 10.1038/sj.onc.1209550 Yuan R, Jiang C, Hong K, Yu X, Wu L, Liu T, Liu X, Tang X, Cai H, Shao J, 2011, Genetic variation in the Fat10 gene is associated with risk of hepatocellular carcinoma in a Chinese population. Asian Pac J Cancer Prev 12(8):2117–2122 Johnson PJ, Williams R, 1987, Cirrhosis and the aetiology of hepatocellular carcinoma. J Hepatol 4(1):140–147 Colombo M, de Franchis R, Del Ninno E, Sangiovanni A, De Fazio C, Tommasini M, Donato MF, Piva A, Di Carlo V, Dioguardi N, 1991, Hepatocellular carcinoma in Italian patients with cirrhosis. N Engl J Med 325(10):675–680., DOI 10.1056/ NEJM199109053251002 Aravalli RN, Steer CJ, Cressman EN, 2008, Molecular mechanisms of hepatocellular carcinoma. Hepatology 48(6):2047–2063., DOI 10.1002/hep. 22580 Spies M, Dasu MR, Svrakic N, Nesic O, Barrow RE, Perez-Polo JR, Herndon DN, 2002, Gene expression analysis in burn wounds of rats. Am J Physiol Regul Integr Comp Physiol 283(4):R918– R930., DOI 10.1152/ajpregu.00170.2002 Llovet JM, Bruix J, 2008, Molecular targeted therapies in hepatocellular carcinoma. Hepatology 48(4):1312–1327., DOI 10.1002/ hep. 22506 Su H, Hu N, Yang HH, Wang C, Takikita M, Wang QH, Giffen C, Clifford R, Hewitt SM, Shou JZ, Goldstein AM, Lee MP, Taylor PR, 2011, Global gene expression profiling and validation in esophageal squamous cell carcinoma and its association with clinical phenotypes. Clin Cancer Res 17(9):2955–2966., DOI 10.1158/ 1078-0432.CCR-10-2724 Kanehisa M, 2002, The KEGG database. Novartis Found Symp 247:91–101; discussion 101–103, 119–128, 244–152 Smyth GK, 2004, Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 3:Article3., DOI 10.2202/1544-6115.1027 Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I, 2001, Controlling the false discovery rate in behavior genetics research. Behav Brain Res 125(1–2):279–284 Zhou L, Liu C, Meng FD, Qu K, Tian F, Tai MH, Wei JC, Wang RT, 2012, Long-term prognosis in hepatocellular carcinoma patients after hepatectomy. Asian Pac J Cancer Prev 13(2):483– 486 Dreesen O, Brivanlou AH, 2007, Signaling pathways in cancer and embryonic stem cells. Stem Cell Rev 3(1):7–17 Drummond DC, Noble CO, Kirpotin DB, Guo Z, Scott GK, Benz CC, 2005, Clinical development of histone deacetylase inhibitors as anticancer agents. Annu Rev Pharmacol Toxicol 45:495–528., DOI 10.1146/annurev.pharmtox.45.120403.095825 Shankar S, Srivastava RK, 2008, Histone deacetylase inhibitors: mechanisms and clinical significance in cancer: HDAC inhibitorinduced apoptosis. Adv Exp Med Biol 615:261–298., DOI 10.1007/ 978-1-4020-6554-5_13 Minucci S, Pelicci PG, 2006, Histone deacetylase inhibitors and the promise of epigenetic, and more, treatments for cancer. Nat Rev Cancer 6(1):38–51., DOI 10.1038/nrc1779 Marks PA, Breslow R, 2007, Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol 25(1):84–90., DOI 10.1038/nbt1272 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 251 EP 258 DI 10.1007/s12253-012-9576-y PG 8 ER PT J AU Lu, Y Sun, G Liu, G Shi, Y Han, Y Yu, F Xiang, X Li, W Xiao, H Liu, X Li, Sh AF Lu, Yifang Sun, Guogui Liu, Geling Shi, Yanping Han, Ying Yu, Fang Xiang, Xiuxiu Li, Weijuan Xiao, Hongzhen Liu, Xiuling Li, Sha TI Clinical Significance of Mannose-Binding Lectin Expression in Thyroid Carcinoma Tissues SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thyroid carcinoma; Apoptosis; Mannosebinding lectin ID Thyroid carcinoma; Apoptosis; Mannosebinding lectin AB Mannose-binding lectin (MBL) plays an important role in the host defence against pathogens and carcinogenesis. This study aimed to analyze differential expression of MBL protein in thyroid cancer tissues and then to investigate the effects of rhMBL in thyroid cancer cells. Tissue specimens from 45 thyroid carcinoma patients and 45 adenoma patients were recruited for immunohistochemical analysis of MBL expression. Cell viability, apoptosis, RT-PCR and Western blot assays were used to detect changes in tumor cell viability, apoptosis, and gene expression, respectively, after treatment of thyroid cancer cells with rhMBL. MBL was differentially expressed in papillary thyroid carcinoma, adenoma, and the distant normal tissues (0.322±0.008, 0.227±0.003, and 0.113 ±0.003, respectively, P<0.05). MBL expression was associated with the advanced disease stage, histological grade, or lymph nodemetastasis in cancer patients (P<0.05).Moreover, rhMBL treatment of thyroid cancer cells reduced tumor cell viability but induced apoptosis in a dose- and time-dependent manner. rhMBL treatment also downregulated Bcl2 protein expression in thyroid cancer cells (P<0.05). In addition, expression p53 protein was increased in thyroid cancer cells after rhMBL treatment (P<0.05). The data from the current study demonstrate that MBL overexpression is associated with advanced thyroid carcinomas, and rhMBL treatment significantly reduced viability but induced apoptosis of thyroid cancer cell lines. Further studies will clarify whether overexpressed MBL in thyroid cancer tissues is functional. C1 [Lu, Yifang] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Sun, Guogui] Tangshan People’s Hospital, Department of ChemoradiotherapyTangshan, China. [Liu, Geling] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Shi, Yanping] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Han, Ying] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Yu, Fang] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Xiang, Xiuxiu] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Li, Weijuan] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Xiao, Hongzhen] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Liu, Xiuling] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Li, Sha] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. RP Liu, G (reprint author), Tangshan Workers Hospital, Department of Endocrinology (Section I), Tangshan, China. EM lyf800218@163.com CR DeLellis RA, Lloyd RV, Heitz PU, Eng C, eds,, 2004, World Health Organization classification of tumours. Pathology and genetics of tumours of endocrine organs. IARC, Lyon Nikiforov YE, 2009, Thyroid tumors: classification and general considerations. In: Nikiforov YE, Biddinger PW, Thompson LDR, eds, Diagnostic pathology and molecular genetcis of the thyroid. Lippincott Williams & Wilkins, Baltimore, pp 94–102 Kimura ET, Nikiforova MN, Zhu Z, Knauf JA, Nikiforov YE, Fagin JA, 2003, High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/ PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res 63(7):1454–1457 Frattini M, Ferrario C, Bressan P, BalestraD,De Cecco L,Mondellini P, Bongarzone I, Collini P, Gariboldi M, Pilotti S, PierottiMA, Greco A, 2004, Alternative mutations of BRAF, RET and NTRK1 are associated with similar but distinct gene expression patterns in papillary thyroid cancer. Oncogene 23(44):7436–7440 Loris R, 2002, Principles of styuctures of animal and plant lectins. Biochim Biophys Acta 1572(2-3):198–208 Fujita T, 2002, Evolution of the lectin-complement pathway and its role in innate immunity. Nat Rev Immunol 2(5):346–353 Bernig T, Boersma BJ, Howe TM, Welch R, Yadavalli S, Staats B, Mechanic LE, Chanock SJ, Ambs S, 2007, The mannose-binding lectin, MBL2, haplotype and breast cancer: an association study in African American and Caucasian women. Carcinogenesis 28, 4):828–836 Wang FY, Tahara T, Arisawa T, Shibata T, Yamashita H, Nakamura M, Yoshioka D, Okubo M, Maruyama N, Kamano T, Kamiya Y, Nakamura M, Fujita H, Nagasaka M, Iwata M, Takahama K, Watanabe M, Nakano H, Hirata I, 2008, Mannan-binding lectin, MBL, polymorphism and Gastric Cancer Risk in Japanese Population. Dig Dis Sci 53(11):2904–2908 Amon PE, Robyn MM, 2003, Impact of mannose-binding lectin on susceptibility to infectious diseases. Clin Infect Dis 37(11):1496–1505 Ma Y, Uemura K, Oka S, Kozutsumi Y, Kawasaki N, Kawasaki T, 1999, Antitumor activity of mannan-binding protein in vivo as revealed by a virus expression system:mannan-binding proteindependent cell-mediated cytotoxicity. Biochem 96(2):371–375 Vang Petersen S, Thiel S, Jensenius JC, 2001, The mannanbinding lectin pathway of complement activation: biology and disease association. Mol lmmunol 38(2–3):133–149 Gu Y, Xia Z, 2008, Studies of mannose-binding lectin. Grad Med J 21:537–541 Garcia-Laorden MI, Rua-Figueroa I, Perez-Aciego P, Rodriguez- Perez JC, Citores MJ, Alamo F, Erausquin C, Rodriguez-Gallego C, 2003, Mannose binding lectin polymorphisms as a diseasemodulating facter in women with systemic lupus erythematosus from Canary Islands, Spain. J Rheumatol 30(4):740–746 Tumer MW, 2002, The role of mannose-binding lectin in health and desease. HK J Paediatr 40(7):134–142 Boniotto M, Braida L, Baldas V, Not T, Ventura A, Vatta S, Radillo O, Tedesco F, Percopo S, Montico M, Amoroso A, Crovella S, 2005, Evidence of a correlation between mannose binding lectin and celiac disease: a model for other autoimmune diseases. J Mol Med 83(4):308–315 Stuart LM, Takahashi K, Shi L, Savill J, Ezekowitz RA, 2005, Mannose-binding lectin-deficient mice display defective apoptotic cells. J Immunol 174(6):3220–3226 Koch A, Melbye M, Sorensen P, Homoe P, Madsen HO, Molbak K, Hansen CH, Andersen LH, Hahn GW, Garred P. Acute respiratory tract infections and mannose-binding lectin insufficiency during early childhood. Uqeskr Laeqer 164(10):5635-5640 van der Zwet WC, Catsburg A, van Elburg RM, Savelkoul PH, Vandenbroucke-Grauls CM, 2008, Mannose binding Lectin, MBL, genotype in relation to risk of nosocomial infection in preterm neonates in the neonatal intensive care unit. Clin Microbiol Infect 14(2):130–135 Minchinton RM, Dean MM, Clark TR, Heatley S, Mullighan CG, 2002, Analysis of the relationship between mannose-binding lectin, MBL, genotype, MBL levels and function in an Australian blood donor population. Seand J Immunol 56(6):630–641 Liu G, Zhang H, Li J, 2006, mRNA differential display cloning of thyroid cancer-related genes. Chin J Clin Oncol 33:621–624 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 259 EP 266 DI 10.1007/s12253-012-9577-x PG 8 ER PT J AU Takacs, T Paszt, A Simonka, Zs Abraham, Sz Borda, B Ottlakan, A Ormandi, K Lazar, M Voros, A Kahan, Zs Lazar, Gy AF Takacs, Tibor Paszt, Attila Simonka, Zsolt Abraham, Szabolcs Borda, Bernadett Ottlakan, Aurel Ormandi, Katalin Lazar, Mate Voros, Andras Kahan, Zsuzsanna Lazar, Gyorgy TI Radioguided Occult Lesion Localisation Versus Wire-Guided Lumpectomy in the Treatment of Non-Palpable Breast Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-palpable; Breast cancer; Wire-guided localisation; Radioguided occult lesion localisation; Surgical margins ID Non-palpable; Breast cancer; Wire-guided localisation; Radioguided occult lesion localisation; Surgical margins AB The purpose of this study was to compare the two methods—guidewire localisation and the radioguided occult lesion localisation—used in the localisation and surgical removal of non-palpable breast tumours. This retrospective study enrolled patients diagnosed with nonpalpable malignant breast tumours. In this study either guidewire localisation (GWL, n069) or radioguided occult lesion localisation (ROLL, n0321) was used for the detection and removal of the tumours. The two methods were compared with regards to preoperative localisation time, operating time, removed specimen volume, the pathological tumour size, the presence of positive surgical margins and postoperative complications. Furthermore, we have also investigated other factors that could have an impact on the frequency of positive resection margins. The localisation time was significantly shorter in the ROLL group, both with ultrasound guidance (5.7±1.44 min vs. 21.6±2.37 min, p<0.05) and with radiographic guidance (21.8±3.1 min vs. 41.6±3.75 min, p<0.021) as well. No significant difference was observed between the two methods in terms of operating time, removed specimen volume and pathological tumour size, or the presence of positive resection margins, or the occurrence of postoperative wound infections. The size of the tumour (ROLL, GWL grps), the presence of a multifocal tumour (ROLL grp), the presence of an extensive in situ breast carcinoma around the invasive cancer (ROLL, GWL grps) and the volume of the removed breast specimen (GWL grp) significantly increased the frequency of positive resection margins. We recommend the use of the ROLL method for the removal of nonpalpable breast tumours as it has a much shorter localisation time, and it is a simpler surgical technique as well. C1 [Takacs, Tibor] University of Szeged, Department of SurgerySzeged, Hungary. [Paszt, Attila] University of Szeged, Department of SurgerySzeged, Hungary. [Simonka, Zsolt] University of Szeged, Department of SurgerySzeged, Hungary. [Abraham, Szabolcs] University of Szeged, Department of SurgerySzeged, Hungary. [Borda, Bernadett] University of Szeged, Department of SurgerySzeged, Hungary. [Ottlakan, Aurel] University of Szeged, Department of SurgerySzeged, Hungary. [Ormandi, Katalin] Euromedic Diagnostics Hungary LtdSzeged, Hungary. [Lazar, Mate] Euromedic Diagnostics Hungary LtdSzeged, Hungary. [Voros, Andras] University of Szeged, Department of PathologySzeged, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of OncotherapySzeged, Hungary. [Lazar, Gyorgy] University of Szeged, Department of SurgerySzeged, Hungary. RP Lazar, Gy (reprint author), University of Szeged, Department of Surgery, Szeged, Hungary. EM lg@surg.szote.u-szeged.hu CR Verkooijen HM, Peeters PHM, Pijnappel RM, Koot VCM, Schipper MEI, Rinkes IHMB, 2000, Diagnostic accuracy of needle-localized open breast biopsy for impalpable breast disease. Br J Surg 87(3):344–347 Kopans DB, DeLuca S, 1980, A modified needle-hook wire technique to simplify preoperative localisation of occult breast lesions. Radiology 134(3):781 Zurrida S, Galimberti V, Monti S, Luini, 1998, A radioguided localisation of occult breast lesions. Breast 7(1):11–13 Gennari R, Galimberti V, De Cicco C, Zurrida S, Zerwes F, Pigatto F et al, 2000, Use of technetium-99m-labeled colloid albumin for preoperative and intraoperative localisation of nonpalpable breast lesions. J Am Coll Surg 190(6):692–698, discussion 698–699 Ronka R, Krogerus L, Leppanen E, von Smitten K, Leidenius M, 2004, Radio-guided occult lesion localisation in patients undergoing breast-conserving surgery and sentinel node biopsy. Am J Surg 187(4):491–496 Gallegos Hernandez JF, Tanis PJ, Deurloo EE, Nieweg OE, Th Rutgers EJ, Kroon BB et al, 2004, Radio-guided surgery improves outcome of therapeutic excision in non-palpable invasive breast cancer. Nucl Med Commun 25(3):227–232 Zgajnar J, Hocevar M, Frkovic-Grazio S, Hertl K, Schweiger E, Besic N, 2004, Radioguided occult lesion localisation, ROLL, of the nonpalpable breast lesions. Neoplasma 51:385–389 Nadeem R, Chagla LS, Harris O, Desmond S, Thind R, Titterrell C et al, 2005, Occult breast lesions: a comparison between radioguided occult lesion localisation, ROLL, vs. wire-guided lumpectomy, WGL). Breast 14(4):283–289 Thind CR, Desmond S, Harris O, Nadeem R, Chagla LS, Audisio RA, 2005, Radio-guided localisation of clinically occult breast lesions, ROLL): a DGH experience. Clin Radiol 60:681–686 Medina-Franco H, Abarca-Perez L, Garcia-Alvarez MN, Ulloa- Gomez JL, Romero-Trejo C, Sepulveda-Mendez J, 2008, Radioguided occult lesion localisation, ROLL, versus wireguided lumpectomy for non-palpable breast lesions: a randomized prospective evaluation. J Surg Oncol 97(2):108–111 De Cicco C, Trifiro G, Intra M, Marotta G, Ciprian A, Frasson A et al, 2004, Optimised nuclear medicine method for tumor marking and sentinel node detection in occult primary breast lesions. Eur J Nucl Med Mol Imaging 31(3):349–354 Pathology Reporting of Breast Disease, 2005, A joint document incorporating the third edition of the NHS Breast Screening Programme Guidelines for Pathology Reporting in Breast. Cancer Screening and the second edition of the Royal College of Pathologists’ Minimum Dataset for Breast Cancer Histopathology. NHSBSP Publication No 58. January Luini A, Zurrida S, Paganelli G, Galimberti V, Sacchini V, Monti S et al, 1999, Comparison of radioguided excision with wire localisation of occult breast lesions. Br J Surg 86(4):522–525 Rampaul RS, Bagnall M, Burrell H, Pinder SE, Evans AJ, Macmillan RD, 2004, Randomized clinical trial comparing radioisotope occult lesion localisation and wire-guided excision for biopsy of occult breast lesions. Br J Surg 91(12):1575–1577 Strnad P, Rob L, Halaska MG, Chod J, Zuntova A, Moravcova Z, 2006, Radioguided occult lesion localisation in combination with detection of the sentinel lymph node in non-palpable breast cancer tumours. Eur J Gynaecol Oncol 27(3):236–238 Moreno M, Wiltgen JE, Bodanese B, Schmitt RL, Gutfilen B, da Fonseca LM, 2008, Radioguided breast surgery for occult lesion localisation—correlation between two methods. J Exp Clin Cancer Res 15(27):29 Mariscal Martinez A, Sola M, de Tudela AP, Julian JF, Fraile M, Vizcaya S et al, 2009, Radioguided localisation of nonpalpable breast cancer lesions: randomized comparison with wire localisation in patients undergoing conservative surgery and sentinel node biopsy. AJR Am J Roentgenol 193(4):1001–1009 Lovrics PJ, Cornacchi SD, Vora R, Goldsmith CH, Kahnamoui K, 2011, Systematic review of radioguided surgery for non-palpable breast cancer. Eur J Surg Oncol 37(5):388–397 Lovrics PJ, Goldsmith CH, Hodgson N, McCready D, Gohla G, Boylan C et al, 2011, A multicentered, randomized, controlled trial comparing radioguided seed localisation to standard wire localisation for nonpalpable, invasive and in situ breast carcinomas. Ann Surg Oncol 18(12):3407–3414, Epub 2011 Apr 30 Besic N, Zgajnar J, Hocevar M, Rener M, Frkovic-Grazio S, Snoj N et al, 2002, Breast biopsy with wire localisation: factors influencing complete excision of nonpalpable carcinoma. Eur Radiol 12, 11):2684–2689 Schaefer FK, Eden I, Schaefer PJ, Peter D, Jonat W, Heller M et al, 2007, Factors associated with one step surgery in case of nonpalpable breast cancer. Eur J Radiol 64(3):426–431 van Esser S, Peters NH, van den Bosch MA, Mali WP, Peeters PH, Borel Rinkes IH et al, 2009, Surgical outcome of patients with core-biopsy-proven nonpalpable breast carcinoma: a large cohort follow-up study. Ann Surg Oncol 16(8):2252–2258 Jackman RJ, Marzoni FA Jr, 1997, Needle-localized breast biopsy: why do we fail? Radiology 204(3):677–684 Smitt MC, Nowels KW, Zdeblick MJ, Jeffrey S, Carlson RW, Stockdale FE et al, 1995, The importance of the lumpectomy surgical margin status in long-term results of breast conservation. Cancer 76(2):259–267 Gage I, Schnitt SJ, Nixon AJ, Silver B, Recht A, Troyan SL et al, 1996, Pathologic margin involvement and the risk of recurrence in patients treated with breast-conserving therapy. Cancer 78, 9):1921–1928 Luini A, Rososchansky J, Gatti G, Zurrida S, Caldarella P, Viale G et al, 2009, The surgical margin status after breast-conserving surgery: discussion of an open issue. Breast Cancer Res Treat 113(2):397–402 Freedman G, Fowble B, Hanlon A, Nicolaou N, Fein D, Hoffman J et al, 1999, Patients with early stage invasive cancer with close or positive margins treated with conservative surgery and radiation have an increased risk of breast recurrence that is delayed by adjuvant systemic therapy. Int J Radiat Oncol Biol Phys 44, 5):1005–1015 Park CC, Mitsumori M, Nixon A, Recht A, Connolly J, Gelman R et al, 2000, Outcome at 8 years after breast-conserving surgery and radiation therapy for invasive breast cancer: influence of margin status and systemic therapy on local recurrence. J Clin Oncol 18, 8):1668–1675 Tartter PI, Kaplan J, Bleiweiss I, Gajdos C, Kong A, Ahmed S et al, 2000, Lumpectomy margins, reexcision, and local recurrence of breast cancer. Am J Surg 179(2):81–85 DiBiase SJ, Komarnicky LT, Heron DE, Schwartz GF, Mansfield CM, 2002, Influence of radiation dose on positive surgical margins in women undergoing breast conservation therapy. Int J Radiat Oncol Biol Phys 53(3):680–686 Mirza NQ, Vlastos G, Meric F, Buchholz TA, Esnaola N, Singletary SE et al, 2002, Predictors of locoregional recurrence among patients with early-stage breast cancer treated with breastconserving therapy. Ann Surg Oncol 9(3):256–265 Intra M, Veronesi P, Mazzarol G et al, 2003, Axillary sentinel lymph node biopsy in patients with pure ductal carcinoma in situ of the breast. Arch Surg 138(3):309–313 Zavagno G, Carcoforo P, Marconato R, Franchini Z, Scalco G, Burelli P et al, 2005, Role of axillary sentinel lymph node biopsy in patients with pure ductal carcinoma in situ of the breast. BMC Cancer 5:28 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 267 EP 273 DI 10.1007/s12253-012-9578-9 PG 7 ER PT J AU Shen, Y Liu, Y Liu, Sh Zhang, A AF Shen, Yizhen Liu, Yi Liu, Shaoge Zhang, Aimin TI Toll-like Receptor 4 Gene Polymorphisms and Susceptibility to Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Toll-like receptor 4; Polymorphism; Bladder cancer ID Toll-like receptor 4; Polymorphism; Bladder cancer AB Bladder cancer is one of the most common malignancies in the world. Toll-like receptor 4 (TLR4) plays important roles in regulating innate immunity and may affect the development of cancers. Polymorphisms in TLR4 gene have been shown to be associated with impaired immune responses. Here, we investigated the association of TLR4 polymorphisms with bladder cancer. Four TLR4 polymorphisms (-2242T/C, Asp299Gly, Thr399Il3, and +3725G/C) were genotyped in a total number of 436 bladder cancer patients and 522 healthy controls. Data were analyzed using the Chi-square test. Results showed that the prevalence of TLR4 +3725GC and CC genotypes were significantly increased in bladder cancer cases than in controls (odds ratio [OR]01.58, 95 % confidence interval [CI]01.19–2.10, p0 0.0015, and OR02.33, 95%CI01.52–3.58, p<0.0001, respectively). Also, the frequency of TLR4 +3725C allele was significantly higher in bladder cancer patients (p<0.0001). The -2242T/C, Asp299Gly and Thr399Il3 polymorphisms did not reveal any significant differences between cases and controls. Stratification analysis of the clinical features in the patients demonstrated that cases with invasive cancer were correlated with higher numbers of +3725GC and CC genotype (p<0.0004 and p<0.0231). In conclusion, these results indicate that TLR4 +3725G/C polymorphism may be a novel risk factor for bladder cancer in the Chinese population. C1 [Shen, Yizhen] General Hospital of Jinan Military Command, Department of Urology, 25 Shifan Road, 250031 Jinan, Shandong, China. [Liu, Yi] General Hospital of Jinan Military Command, Department of Urology, 25 Shifan Road, 250031 Jinan, Shandong, China. [Liu, Shaoge] General Hospital of Jinan Military Command, Department of Urology, 25 Shifan Road, 250031 Jinan, Shandong, China. [Zhang, Aimin] General Hospital of Jinan Military Command, Department of Urology, 25 Shifan Road, 250031 Jinan, Shandong, China. RP Zhang, A (reprint author), General Hospital of Jinan Military Command, Department of Urology, 250031 Jinan, China. EM zhangiaimin@126.com CR Parkin DM, 2004, International variation. Oncogene 23:6329– 6340 Yee DS, Ishill NM, Lowrance WT et al, 2011, Ethnic differences in bladder cancer survival. Urology 78:544–549 Cohen SM, Shirai T, Steineck G, 2000, Epidemiology and etiology of premalignant and malignant urothelial changes. Scand J Urol Nephrol Suppl 34:105–115 Chow JC, Young DW, Golenbock DT et al, 1999, Toll-like receptor 4 mediates lipopolysaccharide-induced signal transduction. J Biol Chem 274:10689–10692 Shi H, Kokoeva MV, Inouye K et al, 2006, TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 116:3015–3025 Schreibelt G, Tel J, Sliepen KH et al, 2010, Tolllike receptor expression and function in human dendritic cell subsets: implications for dendritic cell-based anti-cancer immunotherapy. Cancer Immunol Immunother 59:1573–1582 Liu H, Komai-Koma M, Xu D et al, 2006, Toll-like receptor 2 signaling modulates the functions of CD4+CD25+ regulatory T cells. Proc Natl Acad Sci U S A 103:7048–7053 Sutmuller RP, den Brok MH, Kramer M et al, 2006, Toll-like receptor 2 controls expansion and function of regulatory T cells. J Clin Invest 116:485–494 Kabelitz D, 2007, Expression and function of Toll-like receptors in T lymphocytes. Curr Opin Immunol 19:39–45 Huang B, Zhao J, Li H et al, 2005, Toll-like receptors on tumor cells facilitate evasion of immune surveillance. Cancer Res 65:5009–5014 Kelly MG, Alvero AB, Chen R et al, 2006, TLR-4 signaling promotes tumor growth and paclitaxel chemoresistance in ovarian cancer. Cancer Res 66:3859–3868 Arbour NC, Lorenz E, Schutte BC et al, 2000, TLR4 mutations are associated with endotoxin hyporesponsiveness in humans. Nat Genet 25:187–191 de Oliveira JG, Silva AE, 2012, Polymorphisms of the TLR2 and TLR4 genes are associated with risk of gastric cancer in a Brazilian population. World J Gastroenterol 18:1235–1242 Akil I, Ozkinay F, Onay H, 2012, Assessment of Toll-like receptor-4 gene polymorphism on pyelonephritis and renal scar. Int J Immunogenet 39:303–307 Agundez JA, Garcia-Martin E, Devesa MJ, 2012, Polymorphism of the TLR4 gene reduces the risk of hepatitis C virus-induced hepatocellular carcinoma. Oncology 82:35–40 Creely SJ, McTernan PG, Kusminski CM, Fisher M, 2007, Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am J Physiol Endocrinol Metab 292:E740–E747 Manolakis AC, Kapsoritakis AN, Tiaka EK et al, 2011, TLR4 gene polymorphisms: evidence for protection against type 2 diabetes but not for diabetes-associated ischaemic heart disease. Eur J Endocrinol 165:261–267 Chen K, Wang YT, Gu W et al, 2010, Functional significance of the Toll-like receptor 4 promoter gene polymorphisms in the Chinese Han population. Crit Care Med 38:1292–1299 Sato K, Yoshimura A, Kaneko T, 2012, A single nucleotide polymorphism in 3′-untranslated region contributes to the regulation of toll-like receptor 4 translation. J Biol Chem 287:25163– 25172 Hishida A, Matsuo K, Goto Y et al, 2009, Toll-like receptor 4 +3725G/C polymorphism, Helicobacter pylori seropositivity, and the risk of gastric atrophy and gastric cancer in Japanese. Helicobacter 14:47–53 Zhou L, Wei B, Xing C et al, 2011, Polymorphism in 3′-untranslated region of toll-like receptor 4 gene is associated with protection from hepatitis B virus recurrence after liver transplantation. Transpl Infect Dis 13:250–258 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 275 EP 280 DI 10.1007/s12253-012-9579-8 PG 6 ER PT J AU Miao, Y Li, AL Wang, L Fan, ChF Zhang, XP Xu, HT Yang, LH Liu, Y Wang, EH AF Miao, Yuan Li, Ai-Lin Wang, Liang Fan, Chui-Feng Zhang, Xiu-Peng Xu, Hong-Tao Yang, Lian-He Liu, Yang Wang, En-Hua TI Overexpression of NEDD9 is Associated with Altered Expression of E-Cadherin, β-Catenin and N-Cadherin and Predictive of Poor Prognosis in non-Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; NEDD9; Epithelial-mesenchymal transition; Lymphatic metastasis; Prognosis ID Non-small cell lung cancer; NEDD9; Epithelial-mesenchymal transition; Lymphatic metastasis; Prognosis AB Neural precursor cell expressed, developmentally down-regulated 9 (NEDD9) is overexpressed in multiple tumor types, where it is thought to regulate tumor cell metastasis and act as a trigger of the epithelial-mesenchymal transition (EMT). Loss of E-cadherin/β-catenin and upregulation of Ncadherin are hallmarks of the EMT. The expression and correlation of NEDD9 with E-cadherin, β-catenin and Ncadherin in lung cancer are poorly characterized. We examined NEDD9, E-cadherin, β-catenin and N-cadherin protein expression in 105 cases of non-small cell lung carcinoma (NSCLC), including 43 cases of squamous cell carcinoma and 62 cases of lung adenocarcinoma, and the corresponding normal lung tissues using immunohistochemistry. NEDD9 was overexpressed in 56.2% (59/105) of the NSCLC samples compared to normal lung tissue. Overexpression of NEDD9 correlated with abnormal expression of E-cadherin, β-catenin and N-cadherin (P<0.001, P<0.008 and P<0.027, respectively). Additionally, overexpression of NEDD9 correlated positively with lymph node metastasis in NSCLC (Chisquare test; P<0.015). The mean overall survival of NSCLC patients overexpressing NEDD9 (39.10±6.49 months) was markedly shorter than patients with normal NEDD9 expression (56.67±7.44 months; Log-Rank, P<0.001). Moreover, for patients with adenocarcinoma or squarmous cell carcinoma, the survival is also dramatically poorer upon overexpression of NEDD9. In multivariate analysis, overexpression of NEDD9 (P<0.013) and TNM stage (P<0.001) were significant independent prognostic factors for overall survival in NSCLC. In conclusion, overexpression of NEDD9 correlates with altered expression of EMT markers, increased lymph node metastasis and poorer survival in lung cancer. C1 [Miao, Yuan] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of PathologyShenyang, China. [Li, Ai-Lin] the First Affiliated Hospital of China Medical University, Department of RadiotherapyShenyang, China. [Wang, Liang] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of PathologyShenyang, China. [Fan, Chui-Feng] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of PathologyShenyang, China. [Zhang, Xiu-Peng] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of PathologyShenyang, China. [Xu, Hong-Tao] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of PathologyShenyang, China. [Yang, Lian-He] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of PathologyShenyang, China. [Liu, Yang] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of PathologyShenyang, China. [Wang, En-Hua] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of PathologyShenyang, China. RP Wang, EH (reprint author), China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, Shenyang, China. EM wangeh@hotmail.com CR Travis WD, 2011, Pathology of lung cancer. Clin Chest Med 32, 4):669–692 Jemal A, Siegel R, Ward E et al, 2006, Cancer statistics, 2006. CA Cancer J Clin 56(2):106–130 Singh M, Cowell L, Seo S, O’Neill G, Golemis E, 2007, Molecular basis for HEF1/NEDD9/Cas-L action as a multifunctional co-ordinator of invasion, apoptosis and cell cycle. Cell Biochem Biophys 48(1):54–72 O’Neill GM, Seo S, Serebriiskii IG, Lessin SR, Golemis EA, 2007, A new central scaffold for metastasis: parsing HEF1/ Cas-L/NEDD9. Cancer Res 67(19):8975–8979 Tikhmyanova N, Little JL, Golemis EA, 2010, CAS proteins in normal and pathological cell growth control. Cell Mol Life Sci 67, 7):1025–1048 Law SF, Estojak J, Wang B, Mysliwiec T, Kruh G, Golemis EA, 1996, Human enhancer of filamentation 1, a novel p130cas-like docking protein, associates with focal adhesion kinase and induces pseudohyphal growth in Saccharomyces cerevisiae. Mol Cell Biol 16(7):3327–3337 Natarajan M, Stewart JE, Golemis EA et al, 2006, HEF1 is a necessary and specific downstream effector of FAK that promotes the migration of glioblastoma cells. Oncogene 25(12):1721–1732 Rozenberg GI, Monahan KB, Torrice C, Bear JE, Sharpless NE, 2010, Metastasis in an orthotopic murine model of melanoma is independent of RAS/RAF mutation. Melanoma Res 20(5):361– 371 Kim M, Gans JD, Nogueira C et al, 2006, Comparative oncogenomics identifies NEDD9 as a melanoma metastasis gene. Cell 125(7):1269–1281 Li Y, Bavarva JH,Wang Z et al, 2011, HEF1, a novel target of Wnt signaling, promotes colonic cell migration and cancer progression. Oncogene 30(23):2633–2643 Kim SH, Xia D, Kim SW, Holla V, Menter DG, Dubois RN, 2010, Human enhancer of filamentation 1 Is a mediator of hypoxiainducible factor-1alpha-mediated migration in colorectal carcinoma cells. Cancer Res 70(10):4054–4063 Kong C, Wang C, Wang L et al, 2011, NEDD9 is a positive regulator of epithelial-mesenchymal transition and promotes invasion in aggressive breast cancer. PLoS One 6(7):e22666 Minn AJ, Gupta GP, Siegel PM et al, 2005, Genes that mediate breast cancer metastasis to lung. Nature 436(7050):518–524 Ji H, Ramsey MR, Hayes DN et al, 2007, LKB1 modulates lung cancer differentiation and metastasis. Nature 448(7155):807–810 Thiery JP, Acloque H, Huang RY, Nieto MA, 2009, Epithelialmesenchymal transitions in development and disease. Cell 139, 5):871–890 Tikhmyanova N, Golemis EA, 2011, NEDD9 and BCAR1 negatively regulate E-cadherin membrane localization, and promote E-cadherin degradation. PLoS One 6(7):e22102 Bargon SD, Gunning PW, O’Neill GM, 2005, The Cas family docking protein, HEF1, promotes the formation of neurite-like membrane extensions. Biochim Biophys Acta 1746(2):143–154 Travis WD, Brambilla E, Muller-Hermelink HK, Haris CC, 2004, Pathology and genetics: Tumours of the lung, pleura, thymus and heart. IARC Press, Lyon Goldstraw P, 2011, Updated staging system for lung cancer. Surg Oncol Clin N Am 20(4):655–666 Miao Y, Liu N, Zhang Yet al, 2010, p120ctn isoform 1 expression significantly correlates with abnormal expression of E-cadherin and poor survival of lung cancer patients. Med Oncol 27(3):880– 886 Xu HT, Wang L, Lin D et al, 2006, Abnormal beta-catenin and reduced axin expression are associated with poor differentiation and progression in non-small cell lung cancer. Am J Clin Pathol 125(4):534–541 Prudkin L, Liu DD, Ozburn NC et al, 2009, Epithelial-tomesenchymal transition in the development and progression of adenocarcinoma and squamous cell carcinoma of the lung. Mod Pathol 22(5):668–678 Fashena SJ, Einarson MB, O’Neill GM, Patriotis C, Golemis EA, 2002, Dissection of HEF1-dependent functions in motility and transcriptional regulation. J Cell Sci 115(Pt 1):99–111 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 281 EP 286 DI 10.1007/s12253-012-9580-2 PG 6 ER PT J AU Sulfikkarali, N Krishnakumar, N Manoharan, Sh Nirmal, MR AF Sulfikkarali, Nechikkad Krishnakumar, Narendran Manoharan, Shanmugam Nirmal, Madhavan Ramadas TI Chemopreventive Efficacy of Naringenin-Loaded Nanoparticles in 7,12-dimethylbenz(a)anthracene Induced Experimental Oral Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Nanochemoprevention; DMBA; Hamster buccal pouch carcinogenesis; Naringenin ID Nanochemoprevention; DMBA; Hamster buccal pouch carcinogenesis; Naringenin AB Nanochemoprevention has been introduced recently as a novel approach for improving phytochemicals bioavailability and anti-tumor effect. The present study is designed to evaluate the chemopreventive efficacy of prepared naringenin-loaded nanoparticles (NARNPs) relative to efficacy of free naringenin (NAR) against 7,12-dimethyl benz(a)anthracene (DMBA)-induced oral carcinogenesis by evaluating the status of lipid peroxidation, antioxidants and immunoexpression patterns of proliferating cell nuclear antigen (PCNA) and p53 proteins. Transmission electron microscope (TEM) and dynamic light scattering (DLS) investigations have confirmed a narrow size distribution of the prepared nanoparticles (40–90 nm) with ~88 % encapsulation efficiency. Oral squamous cell carcinoma (OSCC) was developed in the buccal pouch of golden Syrian hamsters by painting with 0.5 % DMBA in liquid paraffin three times a week for 14 weeks. DMBA painted animals revealed the morphological changes, hyperplasia, dysplasia and welldifferentiated squamous cell carcinoma. Moreover, the status of lipid peroxidation, antioxidants and immunoexpression of PCNA and p53 were significantly altered during DMBA-induced oral carcinogenesis. Oral administration of NARNPs (50 mg NAR/kg body weight/day) to DMBA-treated animals completely prevented the tumor formation as compared to the free NAR and significantly reduced the degree of histological lesions, in addition to restoration of the status of biochemical and molecular markers during oral carcinogenesis. In addition, NARNPs have more potent anti-lipid peroxidative, antiproliferative effect and antioxidant potentials compared to free NAR in DMBA-induced oral carcinogenesis. In conclusion, the present study suggests that NARNPs could be a potentially useful drug carrier system for targeted delivery of naringenin for cancer chemoprevention. C1 [Sulfikkarali, Nechikkad] Annamalai University, Department of Physics, 608 002 Annamalainagar, Tamilnadu, India. [Krishnakumar, Narendran] Annamalai University, Department of Physics, 608 002 Annamalainagar, Tamilnadu, India. [Manoharan, Shanmugam] Annamalai University, Department of Biochemistry and Biotechnology, 608 002 Annamalainagar, Tamilnadu, India. [Nirmal, Madhavan Ramadas] Annamalai University, Rajah Muthiah Dental College & Hospital, Department of Oral and Maxillofacial Pathology, 608 002 Annamalainagar, Tamilnadu, India. RP Krishnakumar, N (reprint author), Annamalai University, Department of Physics, 608 002 Annamalainagar, India. EM nskumarphyamu@gmail.com CR Warnakulasurya S, 2010, Living with oral cancer: epidemiology with particular reference to prevalence and life-style changes the influence survival. Oral Oncol 46:407–410 Nagini S, 2009, Of human and hamsters: the hamster buccal pouch carcinogenesis model as a paradigm for oral carcinogenesis and chemoprevention. Anti Cancer Agents Med Chem 9:843–852 Dipple A, Piggot M, Moschel RC, Constantino N, 1983, Evidence that binding of 7,12-dimethylbenz(a)anthracene to DNA in mouse embryo cell cultures results in extensive substitution of both adenine and guanine residues. Can Res 43:4132–4135 Shklar G, 1999, Development of experimental oral carcinogenesis and its impact on current oral cancer research. J Dent Res 78:1768–1772 Nagini S, Vidjaya Letchoumy P, Thangavelu A, Ramachandran CR, 2009, Of humans and hamsters: a comparative evaluation of carcinogen activation, DNA damage, cell proliferation apoptosis, invasive and angiogenesis in oral cancer patients and hamster buccal pouch carcinomas. Oral Oncol 45:31–37 Harish kumar G, Vidya priyadarsini R, Vinothini G et al, 2010, The neem limonoids azadirachtin and nimbolide inhibit cell proliferation and induce apoptosis in an animal model of oral oncogenesis. Invest New Drugs 28:392–401 Amaro MI, Helder Vila-Real JR, Eduardo-Figueira M et al, 2009, Anti-inflammatory activity of naringin and biosynthesised naringenin by naringinase immomobilized in microstructured materials in a model of DDS-induced colitis in mice. Food Res Int 42:1010– 1017 Choi JS, Park KY, Moon SH, Rhee SH et al, 1994, Antimutagenic effect of plant flavonoids in the salmonella assay system. Arch Pharm Res 17:71–75 Lee CH, Jeong TS, Choi YK et al, 2001, Antiatherogenic effect of citrus flavonoids, naringin and naringenin, associated with hepatic ACAT and aorticVCAM-1 and MCP-1 in high cholesterol-fed rabbits. Biophys Res Commun 284:681–688 Ekambaram G, Rajendran P, Magesh V, Sakthisekaran D, 2008, Naringenin reduces tumor size and weight loss in N-methyl-Nnitro- N-nitrosoguanidine-induced gastric carcinogenesis in rats. Nutr Res 28:106–112 Yen FL, Wu TH, Lin LT et al, 2008, Naringenin-loaded nanoparticles improve the physicochemical properties and the hepatoprotective effect of naringenin in orally-administrated rats with CCl4-induced acute liver failure. Pharm Res 26:893–902 Wen J, Liu B, Yuan E, Ma Y et al, 2010, Preperation and physicochemical properties of the complex of naringenin with hydroxypropyl-β-cyclodextrin. Molecules 15:4401–4447 Semalty A, Semalty M, Singh D, Rawat MSM, 2010, Preparation and characterization of phospholipid complexes of naringenin for effective drug delivery. J Incl Phenom Macrocycl Chem 67:253–260 Krishnakumar N, Sulfikkarali N, Rajendra Prasad N, Karthikeyan S, 2011, Enhanced anticancer activity of naringenin-loaded nanoparticles in human cervical, HeLa, cancer cells. Biomed Prev Nutr 1:223–231 Alonso JM, 2004, Nanomedicines for overcoming biological barriers. Biomed Pharmacother 58:168–172 Siddiqui IA, Adhami VM, Bharali DJ et al, 2009, Introducing nanochemoprevention as a novel approach for cancer control: proof of principle with green tea polyphenol epigallocatechin-3- gallate. Cancer Res 69:712–716 Okhawa H, Ohishi N, Yagi K, 1979, Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 95:351–358 Beutler E, Kelly BM, 1963, The effect of sodium nitrite on red cell GSH. Experientia 19:96–97 Rotruck JT, PopeAL,Ganther HE et al, 1973, Selenium: biochemical role as component of gluthaione peroxidase. Science 179:588–599 Kakkar P, Das B, Viswanathan PN, 1984, A modified spectrophotometric assay of superoxide dismutase. Indian J Biochem Biophys 21:130–132 Sinha AK, 1972, Calorimetric assay of catalase. Anal Biochem 47:389–394 Palan PR, Mikhail MS, Basu J, Romney SL, 1991, Plasma levels of antioxidant beta-carotene and alpha-tocopherol in uterine cervix dysplasias and cancer. Nutr Cancer 15:13–20 Panjamurthy K, Manoharan S, Nirmal MR, Vellaichamy L, 2009, Protective role of Withaferin-A on immuno expression of p53 and bcl-2 in 7, 12-dimethylbenz(a)anthracene-induced experimental oral carcinogenesis. Invest New Drugs 27:447–452 Lyzogubov V, Khozhaenko Y, Usenko V et al, 2008, Immunohistochemical analysis of Ki-67, PCNA and S6K1/2 expression in human breast cancer. Exp Oncol 27:141–144 Acharya SDF, Sahoo SK, 2009, Targeted epidermal growth factor receptor nanoparticle bioconjugates for breast cancer therapy. Biomaterials 30:5737–5750 Manoharan S, Vasanthaselvan M, Silvan S et al, 2010, Carnosic acid: A potent chemopreventive agent against oral carcinogenesis. Chem Bio Inter 188:616–622 Krishnakumar N, Manoharan S, Palaniappan PLRM, 2009, Chemopreventive efficacy of piperine in 7,12-dimethylbenz(a)anthrazene( DMBA)-induced hamster buccal pouch carcinogenesis: an FT-IR study. Food Chem Toxicol 47:2813–2820 Wu G, Fang YZ, Yang S et al, 2004, Glutathione metabolism and its implication for health. J Nutr 134:489–492 Silvan S, Manoharan S, Baskaran N et al, 2011, Chemopreventive potential of apigenin in 7, 12-dimethylbenz, a)anthracene induced experimental oral carcinogenices. Eur J Pharmcol 670:571–577 McCormik D, Hall PA, 1992, The complexities of proliferating cell nuclear antigen. Histopathology 21:591–594 Shin DM, Voravud N, Ro JY et al, 1993, Sequential increases in proliferating cell nuclear antigen expression in head and neck tumorigenesis: a potential biomarker. J Natl Cancer Int 85:971– 978 Chans KW, Sarraj S, Lin SS et al, 2000, P53 expression, p53 and Ha-ras mutation and telomerase activation during nitrosaminemediated hamster pouch carcinogenesis. Carcinogenesis 21:1441–1451 Das N, Sahoo SK, 2011, Epithelial cell adhesion molecule targeted nutlin-3a loaded immunonanoparticles for cancer therapy. Acta Biomaterialia 7:355–369 Mohanty C, Sahoo SK, 2010, The in vitro stability and in vivo pharmacokinetics of curcumin prepared as an aqueous nanoparticulate formulation. Biomaterials 3:6597–6611 Acharya S, Sahoo SK, 2011, PLGA nanoparticles containing various anticancer agents and tumor delivery by EPR effect. Adv Drug Del 63:170–183 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 287 EP 296 DI 10.1007/s12253-012-9581-1 PG 10 ER PT J AU Nyari, AT Ottoffy, G Bartyik, K Thurzo, L Solymosi, N Cserni, G Parker, L McNally, JQR AF Nyari, Andras Tibor Ottoffy, Gabor Bartyik, Katalin Thurzo, Laszlo Solymosi, Norbert Cserni, Gabor Parker, Louise McNally, J Q Richard TI Spatial Clustering of Childhood Acute Lymphoblastic Leukaemia in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute lymphoblastic leukaemia; Aetiology; Childhood; Cluster analysis; Environmental exposure; Spatial distribution; Gender specific effect; Male predominance ID Acute lymphoblastic leukaemia; Aetiology; Childhood; Cluster analysis; Environmental exposure; Spatial distribution; Gender specific effect; Male predominance AB The aetiology of childhood acute lymphoblastic leukaemia has been linked with spatially heterogeneous environmental exposures. The presence of spatial clustering would be consistent with geographically localized environmental exposures over long periods of time. The present study is the first to examine spatial clustering amongst children aged 0– 4 years using population-based data from Hungary. The data set consisted of 134 children diagnosed with acute lymphoblastic leukaemia who were resident in part of Hungary during the period 1981–2000. Two levels of spatial aggregation were examined: counties and settlements. The Potthoff-Whittinghill and Moran I autocorrelation methods were used to test for spatial clustering. Additionally, an evaluation of the environmental changes during the study period was considered. Specifically analyses were carried out on sub-periods to investigate a possible effect of the Chernobyl catastrophe. There was statistically significant spatial clustering both at the county (estimate of extra-Poisson variation (β)=0.56, P=0.04) and settlement levels (estimate of extra-Poisson variation (β)=0.68, P=0.0003). At county level, the finding was attributable to clustering amongst female cases, but at settlement level, the finding was limited to male cases. There was significant spatial autocorrelation in the sub-periods immediately following the accident (1986–1990 & 1991–1995), but not before 1986, nor after 1995. A significant autocorrelation was observed during the 5 year period immediately following the accident (1986–1990, global Moran I=0.1334, p=0.005). The centre of significant excesses of ALL cases was located in the county of Baranya. Our study is consistent with an environmental aetiology for acute lymphoblastic leukaemia in children associated with constant exposure to an, as yet unknown, environmental factor in small geographical areas. Although a possible effect of the Chernobyl accident was found in the autocorrelation analysis, the role of chance cannot be excluded. C1 [Nyari, Andras Tibor] University of Szeged, Department of Medical Informatics, 6701 Szeged, Hungary. [Ottoffy, Gabor] University of Pecs, Department of PediatricsPecs, Hungary. [Bartyik, Katalin] University of Szeged, Department of PediatricsSzeged, Hungary. [Thurzo, Laszlo] University of Szeged, Department of OncotherapySzeged, Hungary. [Solymosi, Norbert] Saint Stephen University Budapest, Department of Animal Hygiene, Herd-health and Veterinary EtiologyBudapest, Hungary. [Cserni, Gabor] University of Szeged, Department of PathologySzeged, Hungary. [Parker, Louise] Dalhousie University, IWK Health Centre, Department of PediatricsHalifax, Canada. [McNally, J Q Richard] Newcastle University, Institute of Health and SocietyNewcastle upon Tyne, UK. RP Nyari, AT (reprint author), University of Szeged, Department of Medical Informatics, 6701 Szeged, Hungary. EM Nyari.Tibor@med.u-szeged.hu CR McNally RJQ, Eden TOB, 2004, An infectious aetiology for childhood acute leukaemia: a review of the evidence. Br J Haematol 127:243–263 McNally RJ, Alexander FE, Vincent TJ et al, 2009, Spatial clustering of childhood cancer in Great Britain during the period 1969– 1993. Int J Cancer 124:932–936 Knox EG, Gilman E, 1992, Leukaemia clusters in Great Britain. 2. Geographical concentrations. J Epidemiol Community Health 46:573–576 Petridou E, Alexander FE, Trichopoulos D, Revinthi K, Dessypris N, Wray N et al, 1997, Aggregation of childhood leukemia in geographic areas of Greece. Cancer Causes Control 8:239–245 Alexander FE, Chan LC, Lam TH et al, 1997, Clustering of childhood leukaemia in Hong Kong: association with the childhood peak and common acute lymphoblastic leukaemia and with population mixing. Br J Cancer 75:457–463 Alexander FE, Boyle P, Carli PM et al, 1998, Spatial clustering of childhood leukaemia: summary results from the EUROCLUS project. Br J Cancer 77:818–824 McNally RJQ, Alston RD, Cairns DP, Eden OB, Birch JM, 2003, Geographical and ecological analyses of childhood acute leukaemias and lymphomas in north-west England. Br J Haematol 123:60–65 Hjalmars U, Kulldorff M, Gustafsson G, Nagarwalla N, 1996, Childhood leukaemia in Sweden: using GIS and a spatial scan statistic for cluster detection. Stat Med 15:707–715 Schmiedel S, Blettner M, Kaatsch P, Schuz J, 2010, Spatial clustering and space-time clusters of leukemia among children in Germany, 1987–2007. Eur J Epidemiol 25:627–633 Bellec S, Hemon D, Rudant J, Goubin A, Clavel J, 2006, Spatial and space-time clustering of childhood acute leukaemia in France from 1990 to 2000: a nationwide study. Br J Cancer 94:763–770 Potthoff RF, Whittinghill M, 1966, Testing for homogeneity: II. The Poisson distribution. Biometrika 53:183–190 Moran PAP, 1950, Notes on continuous stochastic phenomena. Biometrika 37(1):17–23., DOI 10.2307/2332142 Nyari TA,Kajtar P, BartyikK, Thurzo L,McNally R, Parker L, 2008, Seasonal variation of childhood acute lymphoblastic leukaemia is different between girls and boys. Pathol Oncol Res 14(4):423–428 Nyari TA, Kajtar P, Bartyik K, Thurzo L, Parker L, 2006, Childhood acute lymphoblastic leukaemia in relation to population mixing around the time of birth in South Hungary. Pediatr Blood Cancer 47:944–948 Cotterill SJ, Parker L, Malcolm AJ, Reid M, More L, Craft AW, 2000, Incidence and survival for cancer in children and young adults in the North of England, 1968–1995: a report from the Northern Region Young Persons’ Malignant Disease Registry. Br J Cancer 83:397–403 Smith M, 1997, Considerations on a possible viral etiology for B– precursor acute lymphoblastic leukemia of childhood. J Immunother 20:89–100 Alexander FE, Boyle P, eds,, 1996, Methods for investigating localized clustering of disease. IARC Scientific Publications No. 135, Lyon Black RJ, Sharp L, Urquhart JD, 1991, An analysis of the geographical distribution of childhood leukaemia and non-Hodgkin lymphomas in Great Britain using areas of approximately equal population size. In: Draper GJ, ed, The geographical epidemiology of childhood leukaemia and non-Hodgkin lymphoma in Great Britain 1966–1983. HMSO, London, pp 61–68 Cuzick J, Edwards R, 1990, Spatial clustering for inhomogeneous populations, with discussion). J Royal Stat Soc Ser B 52:73–104 Openshaw S, Charlton M, Craft AW, Birch JM, 1988, Investigation of leukaemia clusters by the use of a geographical analysis machine. Lancet I:272–273 Besag J, Newell J, 1991, The detection of clusters in rare diseases. J R Stat Soc Ser A 154:143–155 Diggle PJ, Chetwynd AG, 1991, Second-order analysis of spatial clustering for inhomogeneous populations. Biometrics 47:1155– 1163 Anderson NH, Titterington DM, 1996, Some methods for investigating spatial clustering, with epidemiological applications. J R Stat Soc Ser A 160:87–105 Oliver MA, Muir KR, Webster R et al, 1992, A geostatistical approach to the analysis of patterns of rare disease. J Pub Health Med 14:280–289 Kulldorff M, Nagarwall N, 1995, Spatial disease clusters: disease and inference. Stat Med 14:799–810 Hakulinen T, Hovi L, Karkinen-Jaaskelainen PK, Saxen L, 1973, Association between influenza during pregnancy and childhood leukaemia. Br Med J 4(5887):265–267 Nyari TA, Dickinson HO, Parker L, 2003, Childhood cancer in relation to infections in the community during pregnancy and around the time of birth. Int J Cancer 104(6):772–777 Torok S, Borgulya G, Lobmayer P, Jakab Z, Schuler D, Fekete G, 2005, Childhood leukaemia incidence in Hungary, 1973–2002. Interpolation model for analysing the possible effects of the Chernobyl accident. Eur J Epidemiol 20:899–906 Dickinson HO, Parker L, 2002, Leukaemia and non-Hodgkin’s lymphoma in children of male Sellafield radiation workers. Int J Cancer 99:437–444 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 297 EP 302 DI 10.1007/s12253-012-9582-0 PG 6 ER PT J AU Zhou, P Wu, LL Wu, KM Jiang, W Li, Jd Zhou, Ld Li, XY Chang, Sh Huang, Y Tan, H Zhang, GW He, F Wang, ZM AF Zhou, Peng Wu, Lie-Lin Wu, Ke-Min Jiang, Wei Li, Jin-dong Zhou, Le-du Li, Xin-Ying Chang, Shi Huang, Yun Tan, Hui Zhang, Ge-Wen He, Feng Wang, Zhi-Ming TI Overexpression of MMSET is Correlation with Poor Prognosis in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; MMSET; Clinicopathology; Prognosis; Disease-free survival; Overall survival ID Hepatocellular carcinoma; MMSET; Clinicopathology; Prognosis; Disease-free survival; Overall survival AB The multiple myeloma SET domain (MMSET) involved in the t(4;14)(p16;q32) chromosomal translocation encodes a histone lysine methyltransferase. High expression of MMSET is common translocation in multiple myeloma (MM) and is associated with the worst prognosis. Recent studies have shown that overexpression of MMSET is significant in other tumor types compared to their normal tissues. However, little is known about its role in hepatocellular carcinoma (HCC). In these study we investigate the expression of MMSET in HCC and to make correlations with clinicopathologic features. Twenty-eight pairs of HCC and adjacent non-tumor tissues, and eight normal liver tissues were collected for MMSET detection by western blotting and real time-PCR analysis. Immunohistochemistry was used to determine the expression of MMSET in HCC and adjacent non-tumor tissues from 103 patients. Overexpression of MMSET was significantly associated with Edmondson stage, vascular invasion. Moreover, Kaplan- Meier curves showed that MMSET upregulated was associated with shorter overall survival and disease-free survival in HCC patient. In conclusion, our study demonstrates for the first time that overexpression of MMSET is an independent prognostic factor and is correlated with poor survival in HCC patients. C1 [Zhou, Peng] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Wu, Lie-Lin] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Wu, Ke-Min] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Jiang, Wei] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Li, Jin-dong] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Zhou, Le-du] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Li, Xin-Ying] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Chang, Shi] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Huang, Yun] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Tan, Hui] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Zhang, Ge-Wen] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [He, Feng] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. [Wang, Zhi-Ming] Central South University, Xiangya Hospital, Department of General Surgery, No.87 Xiangya Road, 410008 Changsha, China. RP Wang, ZM (reprint author), Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, China. EM wzm005@yahoo.com.cn CR Di Bisceglie AM, 2004, Issues in screening and surveillance for hepatocellular carcinoma. Gastroenterology 127:S104–S107 Szabo E, Paska C, Kaposi Novak P, Schaff Z, Kiss A, 2004, Similarities and differences in hepatitis B and C virus induced hepatocarcinogenesis. Pathol Oncol Res 10(1):5–11 El-Serag HB, Rudolph KL, 2007, Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 132, 7):2557–2576 Xie B, Wang DH, Spechler SJ, 2012, Sorafenib for treatment of hepatocellular carcinoma: a systematic review. Dig Dis Sci 57, 5):1122–1129 Martin C, Zhang Y, 2005, The diverse functions of histone lysine methylation. Nat Rev Mol Cell Biol 6(11):838–849 Li Y, Trojer P, Xu CF et al, 2009, The target of the NSD family of histone lysine methyltransferases depends on the nature of the substrate. J Biol Chem 284(49):34283–34295 Stec I, Wright TJ, van Ommen GJ et al, 1998, WHSC1, a 90 kb SET domain-containing gene, expressed in early development and homologous to a Drosophila dysmorphy gene maps in the Wolf- Hirschhorn syndrome critical region and is fused to IgH in t(4;14, multiple myeloma. Hum Mol Genet 7(7):1071–1082 Chesi M, Nardini E, Lim RS, Smith KD, Kuehl WM, Bergsagel PL, 1998, The t(4;14, translocation in myeloma dysregulates both FGFR3 and a novel gene, MMSET, resulting in IgH/MMSET hybrid transcripts. Blood 92(9):3025–3034 Keats JJ, Reiman T, Belch AR, Pilarski LM, 2006, Ten years and counting: so what do we know about t(4;14)(p16;q32, multiple myeloma. Leuk Lymphoma 47(11):2289–2300 Hudlebusch HR, Skotte J, Santoni-Rugiu E et al, 2011, MMSET is highly expressed and associated with aggressiveness in neuroblastoma. Cancer Res 71(12):4226–4235 Li J, Yin C, Okamoto H, Mushlin H et al, 2008, Identification of a novel proliferation-related protein, WHSC1 4a, in human gliomas. Neuro Oncol 10(1):45–51 Kang HB, Choi Y, Lee JM et al, 2009, The histone methyltransferase, NSD2, enhances androgen receptor-mediated transcription. FEBS Lett 583(12):1880–1886 Rhodes DR, Yu J, Shanker K et al, 2004, ONCOMINE: a cancer microarray database and integrated data-mining platform. Neoplasia 6(1):1–6 Kassambara A, Klein B, Moreaux J, 2009, MMSET is overexpressed in cancers: link with tumor aggressiveness. Biochem Biophys Res Commun 379(4):840–845 Hudlebusch HR, Santoni-Rugiu E, Simon R et al, 2011, The histone methyltransferase and putative oncoprotein MMSET is overexpressed in a large variety of human tumors. Clin Cancer Res 17(9):2919–2933 Okabe H, Satoh S, Kato T et al, 2001, Genome-wide analysis of gene expression in human hepatocellular carcinomas using cDNA microarray: identification of genes involved in viral carcinogenesis and tumor progression. Cancer Res 61(5):2129–2137 Lauring J, Abukhdeir AM, Konishi H et al, 2008, The multiple myeloma associated MMSET gene contributes to cellular adhesion, clonogenic growth, and tumorigenicity. Blood 111(2):856–864 Garlisi CG, Uss AS, Xiao H et al, 2001, A unique mRNA initiated within a middle intron of WHSC1/MMSET encodes a DNA binding protein that suppresses human IL-5 transcription. Am J Respir Cell Mol Biol 24(1):90–98 Kim JY, Kee HJ, Choe NW et al, 2008, Multiple-myelomarelated WHSC1/MMSET isoform RE-IIBP is a histone methyltransferase with transcriptional repression activity. Mol Cell Biol 28(6):2023–2034 Keats JJ, Reiman T, Maxwell CA et al, 2003, In multiple myeloma, t(4;14)(p16;q32, is an adverse prognostic factor irrespective of FGFR3 expression. Blood 101(4):1520–1529 Marango J, Shimoyama M, Nishio H et al, 2008, The MMSET protein is a histone methyltransferase with characteristics of a transcriptional corepressor. Blood 111(6):3145–3154 Saez B, Martin-Subero JI, Lahortiga I et al, 2007, Simultaneous translocations of FGFR3/MMSET and CCND1 into two different IGH alleles in multiple myeloma: lack of concurrent activation of both proto-oncogenes. Cancer Genet Cytogenet 175(1):65–68 Martinez-Garcia E, Popovic R et al, 2011, The MMSET histone methyl transferase switches global histone methylation and alters gene expression in t(4;14, multiple myeloma cells. Blood 117, 1):211–220 Hudlebusch HR, Theilgaard-Monch K, Lodahl M, Johnsen HE, Rasmussen T, 2005, Identification of ID-1 as a potential target gene of MMSET in multiple myeloma. Br J Haematol 130, 5):700–708 Toyokawa G, Cho HS, Masuda K et al, 2011, Histone lysine methyltransferase Wolf-Hirschhorn syndrome candidate one is involved in human carcinogenesis through regulation of the Wnt pathway. Neoplasia 13(10):887–898 Pei H, Zhang L, Luo K et al, 2011, MMSET regulates histone H4K20 methylation and 53BP1 accumulation at DNA damage sites. Nature 470(7332):124–128 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 303 EP 309 DI 10.1007/s12253-012-9583-z PG 7 ER PT J AU Cui, J Xu, G Liu, J Pang, Z Florholmen, J Cui, G AF Cui, Jing Xu, Gang Liu, Jinzhong Pang, Zhigang Florholmen, Jon Cui, Guanglin TI The Expression of Non-Mast Histamine in Tumor Associated Microvessels in Human Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Histamine; Angiogenesis; Colorectal cancer ID Histamine; Angiogenesis; Colorectal cancer AB Angiogenesis is essential for the growth, expansion and metastasis of human colorectal cancers (CRCs). Histamine produced by mast cells is a potent proangiogenic factor. However, the significance of non-mast cell expressing histamine in the tumor microenvironment remains unknown. In this study, we evaluated the histamine positive microvessels with the specific marker for biosynthesis of histamine L-histidine decarboxylase (HDC) in the CRC tumor microenvironment. The relationship between HDC positive microvessel density (HDC-MVD) and clinical pathological parameters was assessed. The results revealed that HDC-MVD in the tumor microenvironment of CRCs was significantly increased as compared with the controls. CRC patients with lymph node invasion had a particularly higher density of HDC-MVD than those without. The density of HDC-MVD accounted for ~79 % of CD34 positive MVD in CRCs and double IHC analysis demonstrated that these HDC positive microvessels were mostly CD34 positive microvessels and with a high proliferative activity. Our results suggest that histamine expressed in microvessels could be an additional cellular source and involved in the cancer invasion through promoting angiogenesis in human CRCs. C1 [Cui, Jing] the Second Affiliated Hospital of Zhengzhou University, Department of MedicineZhengzhou, Henan, China. [Xu, Gang] the Second Affiliated Hospital of Zhengzhou University, Department of MedicineZhengzhou, Henan, China. [Liu, Jinzhong] the Fourth Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, Henan, China. [Pang, Zhigang] the Second Affiliated Hospital of Zhengzhou University, Department of Gastrointestinal SurgeryZhengzhou, Henan, China. [Florholmen, Jon] University of Tromso, Faculty of Medicine, Institute of Clinical Medicine, Laboratory of Gastroenterology & NutritionTromso, Norway. [Cui, Guanglin] the Second Affiliated Hospital of Zhengzhou University, Department of MedicineZhengzhou, Henan, China. RP Cui, G (reprint author), the Second Affiliated Hospital of Zhengzhou University, Department of Medicine, Zhengzhou, China. EM guanglin.cui@uit.no CR Timar J, Csuka O, Orosz Z, Jeney A, Kopper L, 2001, Molecular pathology of tumor metastasis. I. Predictive pathology. Pathol Oncol Res POR 7:217–230 Hubbard J, Grothey A, 2010, Antiangiogenesis agents in colorectal cancer. Curr Opin Oncol 22:374–380 Saclarides TJ, 1997, Angiogenesis in colorectal cancer. Surg Clin N Am 77:253–260 Habibollahi P, Jamshidiha M, Daryani NE, Jahanzad I, Ghahremani MH et al, 2010, Correlation between inducible nitric oxide synthase and cyclooxygenase-2 expression in human colorectal adenocarcinoma: a cross-sectional study. Pathol Oncol Res POR 16:327–335 Jutel M, Blaser K, Akdis CA, 2005, Histamine in allergic inflammation and immune modulation. Int Arch Allergy Immunol 137:82–92 Watanabe T, Ohtsu H, 2002, L-histidine decarboxylase as a probe in studies on histamine. Chem Rec 2:369–376 Natori T, Sata M, Nagai R, Makuuchi M, 2005, Cimetidine inhibits angiogenesis and suppresses tumor growth. Biomed Pharmacother 59:56–60 Tomita K, Izumi K, Okabe S, 2003, Roxatidine- and cimetidineinduced angiogenesis inhibition suppresses growth of colon cancer implants in syngeneic mice. J Pharmacol Sci 93:321–330 Nielsen HJ, Hammer JH, Moesgaard F, Kehlet H, 1991, Possible role of histamine-2 receptor antagonists for adjuvant treatment in colorectal cancer. Clinical review. Eur J Surg 157:437–441 Nielsen HJ, Christensen IJ, Moesgaard F, Kehlet H, 2002, Ranitidine as adjuvant treatment in colorectal cancer. Br J Surg 89:1416–1422 Malaviya R, Uckun FM, 2000, Histamine as an autocrine regulator of leukemic cell proliferation. Leuk Lymphoma 36:367–373 Bowrey PF, King J, Magarey C, Schwartz P, Marr P et al, 2000, Histamine, mast cells and tumour cell proliferation in breast cancer: does preoperative cimetidine administration have an effect? Br J Cancer 82:167–170 Watson SA, Wilkinson LJ, Robertson JF, Hardcastle JD, 1993, Effect of histamine on the growth of human gastrointestinal tumours: reversal by cimetidine. Gut 34:1091–1096 Boer K, Darvas Z, Baki M, Kaszas I, Pal Z et al, 2003, Expression of histidine decarboxylase in human colonic cancer cells and adenomatous polyps. Inflamm Res 52(Suppl 1):S76–S77 Norrby K, 1997, Mast cells and de novo angiogenesis: angiogenic capability of individual mast-cell mediators such as histamine, TNF, IL-8 and bFGF. Inflamm Res 46(Suppl 1):S7–S8 Cui G, Waldum HL, 2007, Physiological and clinical significance of enterochromaffin-like cell activation in the regulation of gastric acid secretion. World J Gastroenterol 13:493–496 Tippens AS, Gruetter CA, 2004, Detection of histidine decarboxylase mRNA in human vascular smooth muscle and endothelial cells. Inflamm Res 53:215–216 El-Ackad TM, Brody MJ, 1975, Evidence for non-mast cell histamine in the vascular wall. Blood Vessels 12:181–191 Gruetter CA, Bailey B, Easterling L, Szarek JL, 2002, Release of non-mast cell histamine from rat aorta. Life Sci 70:1709–1717 Rhee J, Hoff PM, 2005, Angiogenesis inhibitors in the treatment of cancer. Expert Opin Pharmacother 6:1701–1711 Masini E, Fabbroni V, Giannini L, Vannacci A, Messerini L et al, 2005, Histamine and histidine decarboxylase up-regulation in colorectal cancer: correlation with tumor stage. Inflamm Res 54, Suppl 1):S80–S81 Ai W, Takaishi S, Wang TC, Fleming JV, 2006, Regulation of L-histidine decarboxylase and its role in carcinogenesis. Prog Nucleic Acid Res Mol Biol 81:231–270 Kuefner MA, Schwelberger HG, Hahn EG, Raithel M, 2008, Decreased histamine catabolism in the colonic mucosa of patients with colonic adenoma. Dig Dis Sci 53:436–442 Li Z, Liu J, Tang F, Liu Y, Waldum HL et al, 2008, Expression of non-mast cell histidine decarboxylase in tumor-associated microvessels in human esophageal squamous cell carcinomas. APMIS 116:1034–1042 Weidner N, Semple JP, Welch WR, Folkman J, 1991, Tumor angiogenesis and metastasis–correlation in invasive breast carcinoma. N Engl J Med 324:1–8 Yang JJ, Ma YL, Zhang P, Chen HQ, Liu ZH et al, 2011, Histidine decarboxylase is identified as a potential biomarker of intestinal mucosal injury in patients with acute intestinal obstruction. Mol Med 17:1323–1337 Norrby K, 2002, Mast cells and angiogenesis. APMIS 110:355– 371 Ghosh AK, Hirasawa N, Ohtsu H, Watanabe T, Ohuchi K, 2002, Defective angiogenesis in the inflammatory granulation tissue in histidine decarboxylase-deficient mice but not in mast celldeficient mice. J Exp Med 195:973–982 Ghosh AK, Hirasawa N, Ohuchi K, 2001, Enhancement by histamine of vascular endothelial growth factor production in granulation tissue via H(2, receptors. Br J Pharmacol 134:1419–1428 Cianchi F, Cortesini C, Schiavone N, Perna F, Magnelli L et al, 2005, The role of cyclooxygenase-2 in mediating the effects of histamine on cell proliferation and vascular endothelial growth factor production in colorectal cancer. Clin Cancer Res 11:6807– 6815 Hellstrand K, Brune M, Naredi P, Mellqvist UH, Hansson M et al, 2000, Histamine: a novel approach to cancer immunotherapy. Cancer Investig 18:347–355 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 311 EP 316 DI 10.1007/s12253-012-9584-y PG 6 ER PT J AU Zizhen, Z Hui, C Yanying, Sh Danping, Sh Jiahua, L Chao, H Xingzhi, N AF Zizhen, Zhang Hui, Cao Yanying, Shen Danping, Shen Jiahua, Liu Chao, He Xingzhi, Ni TI Correlation Between Immunophenotype Classification and Clinicopathological Features in Chinese Patients with Primary Gastric Diffuse Large B-Cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Diffuse large B-cell lymphoma; Immunophenotype; Primary gastric lymphoma; Prognosis ID Diffuse large B-cell lymphoma; Immunophenotype; Primary gastric lymphoma; Prognosis AB Recent studies have shown that diffuse large Bcell lymphoma (DLBCL) can be classified into germinal center B-cell–like (GCB) and non-GCB phenotypes by immunohistochemical staining. The aim of this study was to investigate the correlation of immunophenotypic classification with clinicopathological features in Chinese patients with primary gastric DLBCL to further our knowledge of this disease. Seventy-three patients with a histopathological diagnosis of primary gastric DLBCL were studied. Immunohistochemistry was carried out using the EnVision method to detect the expression of CD10, Bcl-6, and MUM1. The clinicopathologic features and follow-up data were analyzed using the Kaplan–Meier method, log-rank test, and χ2 test. Expression of CD10 was observed in 21.9 % (16/73) of patients, Bcl-6 in 72.6 % (53/73), and MUM1 in 74.0 % (54/73). According to these data, 32.9 % (24/73) of the cases belonged to GCB subtype and 67.1 % (49/73) belonged to non-GCB subtype. There was a significant difference in tumor size and local lymph node metastasis between the GCB and non-GCB groups (P<0.05). Complications in the GCB group (4.2 %) occurred less frequently than those in the non-GCB group (18.4 %); however, this difference was not significant (P>0.05). Survival analysis revealed that patients in the GCB group had an increased 5- year survival rate compared to those in the non-GCB group (58.5 % vs 35.7 %, χ2=3.939, P<0.05). The 5-year survival rate of patients undergoing R-CHOP chemotherapy was significantly longer than that of patients in the CHOP group (74.7 % vs 37.5 %, χ2=4.185, P<0.05). The immunophenotype classification of primary gastric DLBCL, which is closely related to the tumor size and local lymph nodes metastasis, was found to have prognostic significance. C1 [Zizhen, Zhang] Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital, Department of General Surgery, Dongfang Road 1630, 200127 Shanghai, China. [Hui, Cao] Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital, Department of General Surgery, Dongfang Road 1630, 200127 Shanghai, China. [Yanying, Shen] Medical College of Shanghai Jiao Tong University, Ren Ji Hospital, Department of Pathology, Dongfang Road 1630, 200127 Shanghai, China. [Danping, Shen] Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital, Department of General Surgery, Dongfang Road 1630, 200127 Shanghai, China. [Jiahua, Liu] Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital, Department of General Surgery, Dongfang Road 1630, 200127 Shanghai, China. [Chao, He] Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital, Department of General Surgery, Dongfang Road 1630, 200127 Shanghai, China. [Xingzhi, Ni] Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital, Department of General Surgery, Dongfang Road 1630, 200127 Shanghai, China. RP Xingzhi, N (reprint author), Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital, Department of General Surgery, 200127 Shanghai, China. EM zzzhang16@hotmail.com CR Connor J, Ashton-key M, 2007, Gastric and intestinal diffuse large B-cell lymphomas are clinically and immunophenotypically different. An immunohistochemical and clinical study. Histopathology 51:697–703 Mitchell KA, Finn WG, Owens SR, 2008, Differences in germinal center and non-germinal center phenotype in gastric and intestinal diffuse large B-cell lymphomas. Leuk Lymphoma 49:1717–1723 Chen LP, Lin SJ, Yu MS, 2011, Nonmucosa-associated lymphoid tissue lymphomas of gastric and intestinal origin differ in their clinical features: a single institute experience. J Chin Med Assoc 74:400–407 Ohashi S, Segawa K, Okamura S, Urano H, Kanamori S, Ishikawa H, Hara K, Hukutomi A, Shiral K, Maeda M, 2000, A clinicopathologic study of gastric mucosa-associated lymphoid tissue lymphoma. Cancer 88:2210–2219 Timm S, Sailer M, Fuchs KH, Greiner A, 2001, First successful treatment of a primary high-grade gastric MALT lymphoma by eradication therapy for Helicobacter pylori. Gastroenterology 121:1025–1026 Liu H, Ruskon-Foumestraux A, Lavergne-Slove A, Ye H, Molina T, Bouhnik Y, Hamoudi RA, Diss TC, Doqan A, Meqraud F, Rambaud JC, Du MQ, Isaacson PG, 2001, Resistance of t(11;18, positive gastric mucosa-associated lymphoid tissue lymphoma to Helicobacter pylori eradication therapy. Lancet 357:39–40 Hans CP,Weisenburger DD, Greiner TC, Gascoyne RD, Delabie J, Ott G, Muller-Hermelink HK, Campo E, Braziel RM, Jaffe ES, Pan Z, Farinha P, Smith LM, Falini B, Banham AH, Rosenwald A, Staudt LM, Connors JM, Armitaqe JO, Chan WC, 2004, Confirmation of the molecular classification of diffuse large Bcell lymphoma by immunohistochemistry using a tissue microarray. Blood 103:275–282 Dawson IMP, Cornes JS, Morson BC, 1961, Primary malignant lymphoid tumors of the intestinal tract: report of 37 cases with a study of factors influencing prognosis. Br J Surg 49:80–89 Alizadeh AA, Eisen MB, Davis ER, Ma C, Lossos IS, Rosenwald A, Boldrick JC, Sabet H, Tran T, Yu X, Powell JI, Yang L, Marti GE, Moore T, Hudson J Jr, Lu L, Lewis DB, Tibshirani R, Sherlock G, Chan WC, Greiner TC, Weisenburger DD, Armitaqe JO, Warnke R, Levy R, Wilson W, Grever MR, Byrd JC, Botstein D, Brown PO, Staudt LM, 2000, Distinct types of diffuse large Bcell lymphoma identified by gene expression profiling. Nature 403:503–511 Rosenwald A,Wright G, Chan WC, Connors JM, Campo E, Fisher RI, Gascoyne RD, Muller-Hermelink HK, Smeland EB, Giltnane JM, Hurt EM, Zhao H, Averett L, Yang L, Wilson WH, Jaffe ES, Simon R, Klausner RD, Powell J, Duffey PL, Longo DL, Greiner TC, Weisenburger DD, Sanger WG, Dave BJ, Lynch JC, Vose J, Armitage JO, Montserrat E, Lopez-Guillermo A, Grogan TM, Miller TP, LeBlanc M, Ott G, Kvaloy S, Delabie J, Holte H, Krajci P, Stokke T, Staudt LM, Lymphoma/Leukemia Molecular Profiling Project, 2002, The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med 346:1937–1947 Wright G, Tan B, Rosenwald A, Hurt EH, Wiestner A, Staudt LM, 2003, A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma. Proc Natl Acad Sci USA 100:9991–9996 Monti S, Savage KJ, Kutok JL, Feuerhake F, Kurtin P,MihmM,Wu B, Pasqualucci L, Neuberg D, Aguiar RC, Dal Cin P, Ladd C, Pinkus GS, Salles G, Harris NL, Dalla-Favera R, Habermann TM, Aster JC, Golub TR, Shipp MA, 2005, Molecular profiling of diffuse large Bcell lymphoma identifies robust subtypes including one characterized by host inflammatory response. Blood 105:1851–1861 Barrans SL, Carter I, Owen RG, Davies FE, Patmore RD, Haynes AP, Morgan GJ, Jack AS, 2002, Germinal center phenotype and bcl-2 expression combined with the International Prognostic Index improves patient risk stratification in diffuse large B-cell lymphoma. Blood 99:1136–1143 Colomo L, Lopez-Guillermo A, Perales M, Rives S, Martinez A, Bosch F, Colomer D, Falini B, Montserrat E, Campo E, 2003, Clinical impact of the differentiation profile assessed by immunophenotyping in patients with diffuse large B-cell lymphoma. Blood 101:78–84 Berglund M, Thunberg U, Amini RM, Book M, Roos G, Erlanson M, Linderoth J, Dictor M, Jerkeman M, Cavallin-Stahl E, Sundstrom C, Rehn-Eriksson S, Backlin C, Hagberg H, Rosenquist R, Enblad G, 2005, Evaluation of immunophenotype in diffuse large B-cell lymphoma and its impact on prognosis. Mod Pathol 18:1113–1120 van Imhoff GW, Boerma EJ, van der Holt B, Schuuring E, Verdonck LF, Kluin-Nelemans HC, Kluin PM, 2006, Prognostic impact of germinal center-associated proteins and chromosomal breakpoints in poor-risk diffuse large B-cell lymphoma. J Clin Oncol 24:4135–4142 Choi WW, Weisenburger DD, Greiner TC, Piris MA, Banham AH, Delabie J, Braziel RM, Geng H, Iqbal J, Lenz G, Vose JM, Hans CP, Fu K, Smith LM, Li M, Liu Z, Gascoyne RD, Rosenwald A, Ott G, Rimsza LM, Campo E, Jaffe ES, Jaye DL, Staudt LM, Chan WC, 2009, A new immunostain algorithm classifies diffuse large B-cell lymphoma into molecular subtypes with high accuracy. Clin Cancer Res 15:5494–5502 Meyer PN, Fu K, Greiner TC, Smith LM, Delabie J, Gascoyne RD,Ott G, Rosenwald A, Braziel RM, Campo E, Vose JM, Lenz G, Staudt LM, Chan WC, Weisenburger DD, 2010, Immunohistochemical methods for predicting cell of origin and survival in patients with diffuse large B-cell lymphoma treated with Rituximab. J Clin Oncol 29:200–207 Coiffier B, Lepage E, Briere J, Herbrecht R, Tilly H, Bouabdallah R, Morel P, Van Den Neste E, Salles G, Gaulard P, Reyes F, Lederlin P, Gisselbrecht C, 2002, CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med 346:235–242 Habermann TM, Weller EA, Morrison VA, Gascoyne RD, Cassileth PA, Cohn JB, Dakhil SR, Woda B, Fisher RI, Peterson BA, Horning SJ, 2006, Rituximab-CHOP versus CHOP alone or with maintenance rituximab in older patients with diffuse large Bcell lymphoma. J Clin Oncol 24:3121–3127 Pfreundschuh M, Trumper L, Osterborg A, Pettengell R, Trneny M, Imrie K, Ma D, Gill D, Walewski J, Zinzani PL, Stahel R, Kvaloy S, Shpilberg O, Jaeger U, Hansen M, Lehtinen T, Lopez- Guillermo A, Corrado C, Scheliga A, Milpied N, Mendila M, Rashford M, Kuhnt E, Loeffler M, MabThera International Trial Group, 2006, CHOP-like chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with goodprognosis diffuse large-B-cell lymphoma: a randomised controlled trial by the MabThera International Trial, MInT, Group. Lancet Oncol 7:379–391 Fu K, Weisenburger DD, Choi WW, Perry KD, Smith LM, Shi X, Hans CP, Greiner TC, Bierman PJ, Bociek RG, Armitage JO, Chan WC, Vose JM, 2008, Addition of rituximab to standard chemotherapy improves the survival of both the germinal center B-celllike and non-germinal center B-cell-like subtypes of diffuse large B-cell lymphoma. J Clin Oncol 26:4587–4594 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 317 EP 322 DI 10.1007/s12253-012-9585-x PG 6 ER PT J AU Toth, V Somlai, B Hatvani, Zs Szakonyi, J Gaudi, I Karpati, S AF Toth, Veronika Somlai, Beata Hatvani, Zsofia Szakonyi, Jozsef Gaudi, Istvan Karpati, Sarolta TI Melanoma Screening in a Hungarian Nuclear Power Plant SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sun exposure; Hazard of ionizing radiation; Nuclear power plant; Melanoma screening; Melanoma incidence ID Sun exposure; Hazard of ionizing radiation; Nuclear power plant; Melanoma screening; Melanoma incidence AB The industrial use of the ionizing radiation (IR) particularly stresses the safe work, regular health control is inevitable. Since previous occupational cohorts reported contradictory data on the incidence of melanoma among nuclear industry workers, and in few publications significant increase of it has been described, our clinic was requested by the industry to screen malignant skin tumours among the workers of a power plant. Within a year we have investigated 556 workers, 275 females and 281 males. Out of them 283, majorly males had been officially confirmed as to be employed at hazardous, but strictly controlled environment for an average of 18 years (1–32 years). To distinguish between IR and environmental UV (UVA+UVB) induced cutaneous malignancies we determined the sun and tanning bed exposure of the workers. One in situ melanoma developed in a woman with type I skin, bullous sunburns in the history, who had worked in safe environment for 26 years. Basal cell carcinoma was identified in two men, each of them worked for more than 20 years with IR (in hazardous environment). One had type I skin, the other had type II skin. These results didn’t differ significantly (chi-squared test; p00, 2437 and 1, 0) from the national population data and the results of Euromelanoma screening campaign in Hungary. Our data clearly show, that 1./UV exposure and skin type should be evaluated in occupation cohort studies. 2./The melanoma incidence was not significantly higher among the employees of the power plant than in the general Hungarian population, according to the results of our study, the only Hungarian power plant is safe as far as the skin carcinogenesis is concerned. C1 [Toth, Veronika] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Somlai, Beata] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Hatvani, Zsofia] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Szakonyi, Jozsef] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Gaudi, Istvan] National Institute of Oncology, Rath Gyorgy street 7-9, 1122 Budapest, Hungary. [Karpati, Sarolta] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. RP Toth, V (reprint author), Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, 1085 Budapest, Hungary. EM toveroka@yahoo.com CR Austin DF, Reynolds PJ, Snyder MA, Biggs MW, Stubbs HA, 1981, Malignant melanoma among employees of Lawrence Livermore National Laboratory. Lancet 2:712–716 Acquavella JF,Wilkinson GS, Tietjen GL, Key CR, Voelz GL, 1982, Malignant melanoma incidence at the Los Alamos National Laboratory. Lancet 319:883–884 Acquavella JF, Wilkinson GS, Tietjen GL, Key CR, Stebbings JH, Voelz GL, 1983, A melanoma case–control study at the Los Alamos National Laboratory. Health Phys 45:587–592 Johnson CJ, 1984, Cancer incidence in an area of radioactive fallout downwind from the Nevada Test Site. JAMA 251:230–236 Smith PG, Douglas AJ, 1986, Mortality of workers at the Sellafield plant of British Nuclear Fuels. Br Med J 293:845–854 Kendall GM, Muirhead CR, MacGibbon BH, O’Hagan JA, Conquest AJ, Goodill AA et al, 1992, Mortality and occupational exposure to radiation: first analysis of the National Registry for Radiation Workers. Br Med J 304:220–225 Carpenter L, Higgins C, Douglas A, Fraser C, Beral V, Smith P, 1994, Combined analysis of mortality in three United Kingdom nuclear industry workforces, 1946–1988. Radiat Res 138:224–238 Austin DF, Reynolds P, 1997, Investigation of an excess of melanoma among employees of the Lawrence Livermore National Laboratory. Am J Epidemiol 145:524–531 Moore DH 2nd, Patterson HW, Hatch F, Discher D, Schneider JS, Bennett D et al, 1997, Case–controll study of malignant melanoma among employees of the Lawrence Livermore National Laboratory. Am J Ind Med 32:377–391 Sont WN, Zielinski JM, Ashmore JE, Jiang H, Krewski D, Fair ME et al, 2001, First analysis of cancer incidence and occupational radiation exposure based on the National Dose Registry of Canada. Am J Epidemiol 153:309–318 Whorton MD, Moore ND, Seward JP, Noonan KA, Mendelsohn ML, 2004, Cancer incidence rates among Lawrence Livermore National Laboratory, LLNL, employees: 1974–1997. Am J Ind Med 45:24–33 Zielinski JM, Gamer MJ, Krewski D, Ashmore JP, Band PR, Fair ME et al, 2005, Decreases in occupational exposure to ionizing radiation among Canadian dental workers. J Can Dent Assoc 71:29–33 Telle-Lamberton M, Samson E, Caer S, Bergot D, Bard D, Bermann F et al, 2007, External radiation exposure andmortality in a cohort of French nuclear workers. Occup Environ Med 64:694–700 Cardis E, Vrijheid M, Blettner M, Gilbert E, Hakama M, Hill C et al, 2007, The 15-Country Collaborative Study of Cancer Risk among Radiation Workers in the Nuclear Industry: estimates of radiation-related cancer risks. Radiat Res 167:396–416 Gun RT, Parson J, Crouch P, Ryan P, Hiller JE, 2008, Mortality and cancer incidence of Australian participants in the British nuclear tests in Australia. Occup Environ Med 65:843–848 Jacob P, Ruhm W, Walsh L, Blettner M, Hammer G, Zeeb H, 2009, Cancer risk of radiation workers larger than expected? Occup Environ Med 66:789–796 Lens MB, Dawes M, 2004, Global perspectives of contemporary epidemiological trends of cutaneous malignant melanoma. Br J Dermatol 150:179–185 Garbe C, McLeod GR, Buettner PG, 2000, Time trends of cutaneous melanoma in Queensland, Australia and Central Europe. Cancer 89:1269–1278 Turai I, Souskevich G, 2001, The radiological accident in Lilo, Gerogia. Official joint report of the IAEA and the WHO. International Atomic Energy Agency, Vienna Link EM, Flanagan K, Michalowski AS, Blower PJ, 1999, Lowdoses of ionising radiation induce melanoma metastases and trigger the immune system—adrenal axis feedback loop. Eur J Cancer 35:1526–1533 Charles MW, 2007, Radon exposure of the skin: I. Biological effects J Radiol Prot 27:231–252 Lei U, Masmas TN, Frentz G, 2001, Occupational non-melanoma skin cancer. Acta Derm Venerol 81:415–417 Gawkrodger DJ, 2004, Occupational skin cancers. Occup Med 54:458–463 Gaudi I, Kasler M, 2003, New cases of melanoma as documented in the National Cancer Registry. Hungarian Oncology 47:13–17 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 323 EP 328 DI 10.1007/s12253-012-9587-8 PG 6 ER PT J AU Televantou, D Karkavelas, G Hytiroglou, P Lampaki, S Iliadis, G Selviaridis, P Polyzoidis, SK Fountzilas, G Kotoula, V AF Televantou, Despina Karkavelas, George Hytiroglou, Prodromos Lampaki, Sofia Iliadis, George Selviaridis, Panagiotis Polyzoidis, S Konstantinos Fountzilas, George Kotoula, Vassiliki TI DARPP32, STAT5 and STAT3 mRNA Expression Ratios in Glioblastomas are Associated with Patient Outcome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma subtyping; IDH-negative disease; DARPP32; STAT3; STAT5A; STAT5B; Geneexpression; Survivin ID Glioblastoma subtyping; IDH-negative disease; DARPP32; STAT3; STAT5A; STAT5B; Geneexpression; Survivin AB Based on recent developments in glioblastoma subtyping, we examined DARPP32 (PPP1R1B), a neuronal marker against STAT5 and STAT3 that are pro-oncogenic in glioblastoma. mRNA ratios of DARPP32, STAT1, STAT3, STAT5A and STAT5B were assessed in routinely diagnosed gliomas s including a series of glioblastomas from patients (n067) treated with chemoradiotherapy (temozolomide), out of which 88 % had sequencing validated IDH-negative disease. DARPP32/STAT1 (p00.0007), DARPP32/STAT3 (p0 0.0004) and DARPP32/STAT5B (p00.0039) ratios were significantly higher in grade II and III as compared to grade IV tumours. The same high ratios were also associated with absence of immunohistochemically assessed AKT/PKB phosphorylation and survivin protein expression. High DARPP32/ STAT3,DARPP32/STAT5B, and STAT5B/STAT3 ratios were associated with longer patient progression free (PFS) and overall survival (OS). Upon multivariate analysis, total/subtotal removal of the tumour (HR:0.431; 95%CI:0.241–0.771, Wald p00.005), high DARPP32/STAT5B (HR:0.341; 95%CI:0.169–0.690; Wald p00.003) and STAT5B/STAT3 mRNA ratios (HR:0.480; 95%CI:0.280–0.824; Wald p0 0.008) were independent favorable parameters for prolonged PFS. Extent of surgery (HR:0.198; 95%CI:0.101–0.390; p< 0.001) and high DARPP32/STAT5A ratios (HR:0.320; 95%CI:0.160–0.638, p00.001) were independently predictive for longer OS. The presented approach is applicable for prospective validation and appears promising towards an effective glioblastoma patient stratification in addition to IDH mutations. These data may contribute to understanding the biology of gliomas with respect to their potential neuronal characteristics and justify STAT-inhibiting therapeutic interventions in the same tumour system. C1 [Televantou, Despina] Aristotle University of Thessaloniki, AHEPA University Hospital, Pathology Department, 54006 Thessaloniki, Greece. [Karkavelas, George] Aristotle University of Thessaloniki, AHEPA University Hospital, Pathology Department, 54006 Thessaloniki, Greece. [Hytiroglou, Prodromos] Aristotle University of Thessaloniki, AHEPA University Hospital, Pathology Department, 54006 Thessaloniki, Greece. [Lampaki, Sofia] Aristotle University of Thessaloniki School of Medicine, Department of Medical OncologyThessaloniki, Greece. [Iliadis, George] Department of Radiation OncologyThessaloniki, Greece. [Selviaridis, Panagiotis] Aristotle University of Thessaloniki, AHEPA Hospital, 1st Neurosurgical DepartmentThessaloniki, Greece. [Polyzoidis, S Konstantinos] Aristotle University of Thessaloniki, AHEPA Hospital, 1st Neurosurgical DepartmentThessaloniki, Greece. [Fountzilas, George] Aristotle University of Thessaloniki School of Medicine, Department of Medical OncologyThessaloniki, Greece. [Kotoula, Vassiliki] Aristotle University of Thessaloniki, AHEPA University Hospital, Pathology Department, 54006 Thessaloniki, Greece. RP Kotoula, V (reprint author), Aristotle University of Thessaloniki, AHEPA University Hospital, Pathology Department, 54006 Thessaloniki, Greece. EM vkotoula@auth.gr CR WHO, 2007)WHO Classification of Tumors of the Central Nervous System. International Agency for Research on Cancer, City Wen PY, Kesari S, 2008, Malignant gliomas in adults. N Engl J Med 359:492–507 Riemenschneider MJ, Jeuken JW, Wesseling P, Reifenberger G, 2010, Molecular diagnostics of gliomas: state of the art. Acta Neuropathol 120:567–584 Yan H, Bigner DD, Velculescu V, Parsons DW, 2009, Mutant metabolic enzymes are at the origin of gliomas. Cancer Res 69:9157–9159 Ohgaki H, Kleihues P, 2011, Genetic profile of astrocytic and oligodendroglial gliomas. Brain Tumor Pathol 28:177–183 Lai A, Kharbanda S, Pope WB et al, 2011, Evidence for sequenced molecular evolution of IDH1 mutant glioblastoma from a distinct cell of origin. J Clin Oncol 29:4482–4490 Hartmann C, Hentschel B, Wick Wet al, 2011, Patients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: implications for classification of gliomas. Acta Neuropathol 120:707–718 Phillips HS, Kharbanda S, Chen R et al, 2006, Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 9:157–173 Verhaak RG, Hoadley KA, Purdom E et al, 2010, Integrated Genomic Analysis Identifies Clinically Relevant Subtypes of Glioblastoma Characterized by Abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17:98–110 Brennan C,Momota H, Hambardzumyan D et al, 2009, Glioblastoma subclasses can be defined by activity among signal transduction pathways and associated genomic alterations. PLoS One 4:e7752 Noushmehr H, Weisenberger DJ, Diefes K et al, 2010, Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17:510–522 Stupp R, Tonn JC, Brada M, Pentheroudakis G, 2010, High-grade malignant glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 21(Suppl 5):v190–v193 Stupp R, Hegi ME, Mason WP et al, 2009, Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10:459– 466 CGARN, 2008, Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455:1061–1068 Carro MS, Lim WK, Alvarez MJ et al, 2010, The transcriptional network for mesenchymal transformation of brain tumours. Nature 463:318–325 de la Iglesia N, Puram SV, Bonni A, 2009, STAT3 regulation of glioblastoma pathogenesis. Curr Mol Med 9:580–590 Anderson JC, McFarland BC, Gladson CL, 2008, New molecular targets in angiogenic vessels of glioblastoma tumours. Expert Rev Mol Med 10:e23 Liang QC, Xiong H, Zhao ZW et al, 2009, Inhibition of transcription factor STAT5b suppresses proliferation, induces G1 cell cycle arrest and reduces tumor cell invasion in human glioblastoma multiforme cells. Cancer Lett 273:164–171 Haybaeck J, Obrist P, Schindler CU, Spizzo G, Doppler W, 2007, STAT-1 expression in human glioblastoma and peritumoral tissue. Anticancer Res 27:3829–3835 Hemmings HC Jr, Greengard P, Tung HY, Cohen P, 1984, DARPP-32, a dopamine-regulated neuronal phosphoprotein, is a potent inhibitor of protein phosphatase-1. Nature 310:503–505 Brene S, Lindefors N, Ehrlich M et al, 1994, Expression of mRNAs encoding ARPP-16/19, ARPP-21, and DARPP-32 in human brain tissue. J Neurosci 14:985–998 Svenningsson P, Nishi A, Fisone G, Girault JA, Nairn AC, Greengard P, 2004, DARPP-32: an integrator of neurotransmission. Annu Rev Pharmacol Toxicol 44:269–296 Yger M, Girault JA, 2011, DARPP-32, Jack of All Trades… Master of Which? Frontiers in behavioral neuroscience 5:56 Gu L, Waliany S, Kane SE, 2009, Darpp-32 and its truncated variant t-Darpp have antagonistic effects on breast cancer cell growth and herceptin resistance. PLoS One 4:e6220 Hansen C, Howlin J, Tengholm A et al, 2009, Wnt-5a-induced phosphorylation of DARPP-32 inhibits breast cancer cell migration in a CREB-dependent manner. J Biol Chem 284:27533–27543 Bogush A, Pedrini S, Pelta-Heller J et al, 2007, AKT and CDK5/ p35 mediate brain-derived neurotrophic factor induction of DARPP-32 in medium size spiny neurons in vitro. J Biol Chem 282:7352–7359 Belkhiri A, Dar AA, Zaika A, Kelley M, El-Rifai W, 2008, t- Darpp promotes cancer cell survival by up-regulation of Bcl2 through Akt-dependent mechanism. Cancer Res 68:395–403 Chen F, Xu Y, Luo Y et al, 2010, Down-regulation of Stat3 decreases invasion activity and induces apoptosis of human glioma cells. J Mol Neurosci 40:353–359 Konnikova L, Kotecki M, Kruger MM, Cochran BH, 2003, Knockdown of STAT3 expression by RNAi induces apoptosis in astrocytoma cells. BMC Cancer 3:23 Stupp R, Mason WP, van den Bent MJ et al, 2005, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352:987–996 Zhang F,Wang ZM, Liu HYet al, 2011, Application of RT-PCR in formalin-fixed and paraffin-embedded lung cancer tissues. Acta Pharmacol Sinica 31:111–117 Sanchez-Navarro I, Gamez-Pozo A, Gonzalez-Baron M et al, 2010, Comparison of gene expression profiling by reverse transcription quantitative PCR between fresh frozen and formalinfixed, paraffin-embedded breast cancer tissues. Biotechniques 48:389–397 Jeuken JW, Cornelissen SJ, Vriezen M et al, 2007, MS-MLPA: an attractive alternative laboratory assay for robust, reliable, and semiquantitative detection of MGMT promoter hypermethylation in gliomas. Lab Invest 87:1055–1065 Mizoguchi M, Betensky RA, Batchelor TT, Bernay DC, Louis DN, Nutt CL, 2006, Activation of STAT3, MAPK, and AKT in malignant astrocytic gliomas: correlation with EGFR status, tumor grade, and survival. J Neuropathol Exp Neurol 65:1181–1188 Spaulding B, Pan D, Ghadersohi A et al, 2006, Characterization of the 12C4 survivin monoclonal antibody and insight into the expression of survivin in human adult tissues. Histopathology 49:622–633 UematsuM, Ohsawa I,Aokage Tet al, 2005, Prognostic significance of the immunohistochemical index of survivin in glioma: a comparative study with the MIB-1 index. J Neurooncol 72:231–238 Preusser M, Gelpi E, Matej R et al, 2005, No prognostic impact of survivin expression in glioblastoma. Acta Neuropathol 109:534–538 Johannessen AL, Torp SH, 2006, The clinical value of Ki-67/ MIB-1 labeling index in human astrocytomas. Pathol Oncol Res 12:143–147 Stupp R, Roila F, 2009, Malignant glioma: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol 20, Suppl 4):126–128 Feltus FA, Groner B, Melner MH, 1999, Stat5-mediated regulation of the human type II 3beta-hydroxysteroid dehydrogenase/delta5- delta4 isomerase gene: activation by prolactin. Mol Endocrinol 13:1084–1093 Waxman DJ, 2000, Growth hormone pulse-activated STAT5 signalling: a unique regulatory mechanism governing sexual dimorphism of liver gene expression. Novartis Found Symp 227:61–74, discussion 75–81 Viglietto G, Motti ML, Bruni P et al, 2002, Cytoplasmic relocalization and inhibition of the cyclin-dependent kinase inhibitor p27, Kip1, by PKB/Akt-mediated phosphorylation in breast cancer. Nat Med 8:1136–1144 Shirai K, Suzuki Y, Oka K et al, 2009, Nuclear survivin expression predicts poorer prognosis in glioblastoma. J Neurooncol 91:353–358 Mellai M, Caldera V, Patrucco A, Annovazzi L, Schiffer D, 2008, Survivin expression in glioblastomas correlates with proliferation, but not with apoptosis. Anticancer Res 28:109–118 Brantley EC, Benveniste EN, 2008, Signal transducer and activator of transcription-3: a molecular hub for signaling pathways in gliomas. Mol Cancer Res 6:675–684 Bhat KP, Salazar KL, Balasubramaniyan V et al, 2011, The transcriptional coactivator TAZ regulates mesenchymal differentiation in malignant glioma. Genes Dev 25:2594–2609 Cooper LA, Gutman DA, Chisolm C et al, 2012, The tumor microenvironment strongly impacts master transcriptional regulators and gene expression class of glioblastoma. Am J Pathol 180:2108–2119 Kohsaka S, Wang L, Yachi K et al., 2012, STAT3 inhibition overcomes temozolomide resistance in glioblastoma by downregulating MGMT expression. Mol Cancer Ther Lo HW, Cao X, Zhu H, Ali-Osman F, 2008, Constitutively activated STAT3 frequently coexpresses with epidermal growth factor receptor in high-grade gliomas and targeting STAT3 sensitizes them to Iressa and alkylators. Clin Cancer Res 14:6042–6054 Chumbalkar V, Latha K, Hwang Y et al, 2011, Analysis of phosphotyrosine signaling in glioblastoma identifies STAT5 as a novel downstream target of DeltaEGFR. J Proteome Res 10:1343–1352 Kostianovsky AM, Maier LM, Anderson RC, Bruce JN, Anderson DE, 2008, Astrocytic regulation of human monocytic/microglial activation. J Immunol 181:5425–5432 Tan SH, Nevalainen MT, 2008, Signal transducer and activator of transcription 5A/B in prostate and breast cancers. Endocr Relat Cancer 15:367–390 Walker SR, Nelson EA, Zou L et al, 2009, Reciprocal effects of STAT5 and STAT3 in breast cancer. Mol Cancer Res 7:966–976 Aubry L, Bugi A, Lefort N, Rousseau F, Peschanski M, Perrier AL, 2008, Striatal progenitors derived from human ES cells mature into DARPP32 neurons in vitro and in quinolinic acid-lesioned rats. Proc Natl Acad Sci U S A 105:16707–16712 Beckler A,Moskaluk CA, Zaika A et al, 2003, Overexpression of the 32-kilodalton dopamine and cyclic adenosine 3′,5′-monophosphateregulated phosphoprotein in common adenocarcinomas. Cancer 98:1547–1551 He F, Sun YE, 2007, Glial cells more than support cells? Int J Biochem Cell Biol 39:661–665 Visnyei K, Onodera H, Damoiseaux R et al, 2011, A molecular screening approach to identify and characterize inhibitors of glioblastoma stem cells. Mol Cancer Ther 10:1818–1828 Sarbassov DD, Ali SM, Sabatini DM, 2005, Growing roles for the mTOR pathway. Curr Opin Cell Biol 17:596–603 Fan QW, Weiss WA, 2012, Inhibition of PI3K-Akt-mTOR signaling in glioblastoma by mTORC1/2 inhibitors. Methods Mol Biol 821:349–359 Koul D, Shen R, Bergh S et al, 2006, Inhibition of Akt survival pathway by a small-molecule inhibitor in human glioblastoma. Mol Cancer Ther 5:637–644 Zhao J, Tenev T, Martins LM, Downward J, Lemoine NR, 2000, The ubiquitin-proteasome pathway regulates survivin degradation in a cell cycle-dependent manner. J Cell Sci 113(Pt 23):4363– 4371 Espina V, Edmiston KH, Heiby M et al, 2008, A portrait of tissue phosphoprotein stability in the clinical tissue procurement process. Mol Cell Proteomics 7:1998–2018 Weller M, Stupp R, HegiM,WickW, 2012, Individualized targeted therapy for glioblastoma: fact or fiction? Cancer J 18:40–44 Marko NF, Quackenbush J, Weil RJ, 2011, Why is there a lack of consensus on molecular subgroups of glioblastoma? Understanding the nature of biological and statistical variability in glioblastoma expression data. PLoS One 6:e20826 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2013 VL 19 IS 2 BP 329 EP 343 DI 10.1007/s12253-012-9588-7 PG 15 ER PT J AU Muzes, Gy Sipos, F Csomor, J Sreter, L AF Muzes, Gyorgyi Sipos, Ferenc Csomor, Judit Sreter, Lidia TI Multicentric Castleman’s Disease: A Challenging Diagnosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Multicentric Castleman’s disease; HHV-8; IL-6; Differential diagnosis; Therapy; Monoclonal antibodies ID Multicentric Castleman’s disease; HHV-8; IL-6; Differential diagnosis; Therapy; Monoclonal antibodies AB Multicentric Castleman’s disease (MCD) is a sytemic disorder with flares of non-specific symptoms suggestive of a chronic inflammatory syndrome. It is typically accompanied by generalized lymphadenopathy and multiorgan involvement. Histologically, two main variants of Castleman’s disease exist, the hyalin vascular type and the plasma cell variant. Upon localization unicentric (localized), and multicentric (diffuse, systemic) subtypes can be distinguished with more different disease outcomes. Patients often exhibit acute phase reactions and several autoimmune phenomena, and are at high risk for developing malignancies. Both the idiopathic and the HHV-8-driven infectious forms of MCD represent distinct disease entities with a less favorable prognosis. The induction of human IL-6 excess via yet unknown upstream mechanisms, and overexpression of viral IL-6 by HHV-8 can pivotally influence MCD biology. Based on the role of IL-6 in pathogenesis, MCD is also designated as IL-6 lymphadenopathy. To date there are no direct therapeutic evidences, but having been translated to daily practice the main regulatory factors may serve as promising therapeutic targets. C1 [Muzes, Gyorgyi] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. [Csomor, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1088 Budapest, Hungary. [Sreter, Lidia] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi u. 46, 1088 Budapest, Hungary. RP Muzes, Gy (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM muzes.gyorgyi@med.semmelweis-univ.hu CR Castleman B, Towne VW, 1954, Case records of the Massachusetts General Hospital; weekly clinicopathological exercises; founded by Richard C. Cabot. N Engl J Med 251:396–400 Herrada J, Cabanillas F, Rice L, Manning J, Pugh W, 1998, The clinical behavior of localized and multicentric Castleman disease. Ann Intern Med 128:657–662 Roca B, 2009, Castleman’s disease. A review. AIDS Rev 11:3–7. Keller AR, Hochholzer L, Castleman B, 1972, Hyaline-vascular and plasma-cell types of giant lymph node hyperplasia of the mediastinum and other locations. Cancer 29:670–683 Gaba AR, Stein RS, Sweet DL, Variakojis D, 1978, Multicentric giant lymph node hyperplasia. Am J Clin Pathol 69:86–90 Peterson BA, Frizzera G, 1993, Multicentric Castleman’s disease. Semin Oncol 20:636–647 Maslovsky I, Uriev L, Lugassy G, 2000, The heterogeneity of Castleman disease: report of five cases and review of the literature. Am J Med Sci 320:292–295 Waterston A, Bower M, 2004, Fifty years of multicentric Castleman’s disease. Acta Oncol 43:698–704 Frizzera G, Peterson BA, Bayrd ED, Goldman A, 1985, A systemic lymphoproliferative disorder with morphologic features of Castleman’s disease: clinical findings and clinicopathologic correlations in 15 patients. J Clin Oncol 3:1202–1216 Frizzera G, 2001, Atypical lymphoproliferative disorders. In: Knowles DM, ed, Neoplastic hematopathology. Lippincott,Williams and Wilkins, Philadelphia, pp 595–621 Collins LS, Fowler A, Tong CY, de Ruiter A, 2006, Multicentric Castleman’s disease in HIV infection. Int J STD AIDS 17:19–24 Soulier J, Grollet L, Oksenhendler E et al, 1995, Kaposi’s sarcomaassociated herpesvirus-like DNA sequences in multicentric Castleman’s disease. Blood 86:1276–1280 Cronin DM, Warnke RA, 2009, Castleman disease: an update on classification and the spectrum of associated lesions. Adv Anat Pathol 16:236–246 Shahidi H, Myers JL, Kvale PA, 1995, Castleman’s disease. Mayo Clin Proc 70:969–977 Seida A, Wada J, Morita Y et al, 2004, Multicentric Castleman’s disease associated with glomerular microangiopathy and MPGNlike lesion: does vascular endothelial cell-derived growth factor play causative or protective roles in renal injury? Am J Kidney Dis 43:E3–E9 Suneja S, Chidambaram M, Herzenberg AM, Bargman JM, 2009, Kidney involvement in multicentric castleman disease. Am J Kidney Dis 53:550–554 Guihot A, Couderc LJ, Agbalika F et al, 2005, Pulmonary manifestations of multicentric Castleman’s disease in HIV infection: a clinical, biological and radiological study. Eur Respir J 26:118– 125 Weisenburger DD, Nathwani BN, Winberg CD, Rappaport H, 1985, Multicentric angiofollicular lymph node hyperplasia: a clinicopathologic study of 16 cases. Hum Pathol 16:162– 172 Gessain A, Sudaka A, Briere J et al, 1996, Kaposi sarcomaassociated herpes-like virus, human herpesvirus type 8, DNA sequences in multicentric Castleman’s disease: is there any relevant association in non-human immunodeficiency virus-infected patients? Blood 87:414–416 Izuchukwu IS, Tourbaf K, Mahoney MC, 2003, An unusual presentation of Castleman’s disease: a case report. BMC Infect Dis 3:20 Oksenhendler E, Duarte M, Soulier J et al, 1996, Multicentric Castleman’s disease in HIV infection: a clinical and pathological study of 20 patients. AIDS 10:61–67 Oksenhendler E, Boulanger E, Galicier L et al, 2002, High incidence of Kaposi sarcoma-associated herpesvirus-related non-Hodgkin lymphoma in patients with HIV infection and multicentric Castleman disease. Blood 99:2331–2336 Larroche C, Cacoub P, Soulier J et al, 2002, Castleman’s disease and lymphoma: report of eight cases in HIV-negative patients and literature review. Am J Hematol 69:119–126 Du MQ, Liu H, Diss TC et al, 2001, Kaposi sarcoma-associated herpesvirus infects monotypic, IgM lambda, but polyclonal naive B cells in Castleman disease and associated lymphoproliferative disorders. Blood 97:2130–2136 Dupin N, Diss TL, Kellam P et al, 2000, HHV-8 is associated with a plasmablastic variant of Castleman disease that is linked to HHV- 8-positive plasmablastic lymphoma. Blood 95:1406–1412 Kojima M, Nakamura S, Nishikawa M, Itoh H, Miyawaki S, Masawa N, 2005, Idiopathic multicentric Castleman’s disease. A clinicopathologic and immunohistochemical study of five cases. Pathol Res Pract 201:325–332 Kojima M, Nakamura S, Itoh H et al, 1997, Systemic lupus erythematosus, SLE, lymphadenopathy presenting with histopathologic features of Castleman’ disease: a clinicopathologic study of five cases. Pathol Res Pract 193:565–571 Ioachim HL, Cronin W, Roy M, Maya M, 1990, Persistent lymphadenopathies in people at high risk for HIV infection. Clinicopathologic correlations and long-term follow-up in 79 cases. Am J Clin Pathol 93:208–218 Kojima M, Nakamura S, Shimizu K et al, 2004, Clinical implication of idiopathic plasmacytic lymphadenopathy with polyclonal hypergammaglobulinemia: a report of 16 cases. Int J Surg Pathol 12:25–30 Frizzera G, Banks PM, Massarelli G, Rosai J, 1983, A systemic lymphoproliferative disorder with morphologic features of Castleman’s disease. Pathological findings in 15 patients. Am J Surg Pathol 7:211–231 Koo CH, Nathwani BN, Winberg CD, Hill LR, Rappaport H, 1984, Atypical lymphoplasmacytic and immunoblastic proliferation in lymph nodes of patients with autoimmune disease, autoimmune- disease-associated lymphadenopathy). Med, Baltimore, 63:274–290 Bianchi MM, Narvaez J, Santo P et al, 2009, Multicentric Castleman’s disease mimicking adult-onset Still’s disease. Joint Bone Spine 76:304–307 Bonekamp D, Horton KM, Hruban RH, Fishman EK, 2011, Castleman disease: the great mimic. Radiographics 31:1793– 1807 Polizzotto MN, Uldrick TS, Hu D, Yarchoan R, 2012, Clinical manifestations of Kaposi sarcoma herpesvirus lytic activation: multicentric Castleman disease, KSHV-MCD, and the KSHV inflammatory cytokine syndrome. Front Microbiol 3:73 Nishimoto N, Kishimoto T, 2006, Interleukin 6: from bench to bedside. Nat Clin Pract Rheumatol 2:619–626 Kishimoto T, 2006, Interleukin-6: discovery of a pleiotropic cytokine. Arthritis Res Ther 8:S2 Kishimoto T, 1989, The biology of interleukin-6. Blood 74:1–10 Vinzio S, Ciarloni L, Schlienger JL, Rohr S, Mechine A, Goichot B, 2008, Isolated microcytic anemia disclosing a unicentric Castleman disease: the interleukin-6/hepcidin pathway? Eur J Intern Med 19:367–369 Nemeth E, Valore EV, Territo M, Schiller G, Lichtenstein A, Ganz T, 2003, Hepcidin, a putative mediator of anemia of inflammation, is a type II acute-phase protein. Blood 101:2461–2463 Sullivan RJ, Pantanowitz L, Casper C, Stebbing J, Dezube BJ, 2008, HIV/AIDS: epidemiology, pathophysiology, and treatment of Kaposi sarcoma-associated herpesvirus disease: Kaposi sarcoma, primary effusion lymphoma, and multicentric Castleman disease. Clin Infect Dis 47:1209–1215 Moore PS, Chang Y, 1995, Detection of herpesvirus-like DNA sequences in Kaposi’s sarcoma in patients with and without HIV infection. N Engl J Med 332:1181–1185 Davis DA, Humphrey RW, Newcomb FM et al, 1997, Detection of serum antibodies to a Kaposi’s sarcoma-associated herpesvirusspecific peptide. J Infect Dis 175:1071–1079 Kedes DH, Ganem D, Ameli N, Bacchetti P, Greenblatt R, 1997, The prevalence of serum antibody to human herpesvirus 8, Kaposi sarcoma-associated herpesvirus, among HIV-seropositive and high-risk HIV-seronegative women. JAMA 277:478–481 Pauk J, Huang ML, Brodie SJ et al, 2000, Mucosal shedding of human herpesvirus 8 in men. N Engl J Med 343:1369–1377 Regamey N, Tamm M, Wernli M et al, 1998, Transmission of human herpesvirus 8 infection from renal-transplant donors to recipients. N Engl J Med 339:1358–1363 Levi JE, Nascimento MC, Sumita LM et al, 2011, Non-detection of human herpesvirus 8, HHV-8, DNA in HHV-8-seropositive blood donors from three Brazilian regions. PLoS One 6:e23546 Damania B, 2007, DNA tumor viruses and human cancer. Trends Microbiol 15:38–44 Fukumoto H, Kanno T, Hasegawa H, Katano H, 2011, Pathology of Kaposi’s Sarcoma-Associated Herpesvirus infection. Front Microbiol 2:175 Belec L, Mohamed AS, Authier FJ et al, 1999, Human herpesvirus 8 infection in patients with POEMS syndrome-associated multicentric Castleman’s disease. Blood 93:3643–3653 Yamasaki S, Iino T, Nakamura M et al, 2003, Detection of human herpesvirus-8 in peripheral blood mononuclear cells from adult Japanese patients with multicentric Castleman’s disease. Br J Haematol 120:471–477 Darazam IA, Mansouri SD, Karimi S et al, 2010, A 25-year-old woman with cough, constitutional symptoms and lymphadenopathy. Tanaffos 9:80–83 Schulte KM, Talat N, 2010, Castleman’s disease–a two compartment model of HHV8 infection. Nat Rev Clin Oncol 7:533–543 Martro E, Cannon MJ, Dollard SC et al, 2004, Evidence for both lytic replication and tightly regulated human herpesvirus 8 latency in circulating mononuclear cells, with virus loads frequently below common thresholds of detection. J Virol 78:11707–11714 Neipel F, Albrecht JC, Ensser A et al, 1997, Human herpesvirus 8 encodes a homolog of interleukin-6. J Virol 71:839–842 Cesarman E, 2011, Gammaherpesvirus and lymphoproliferative disorders in immunocompromised patients. Cancer Lett 305:163– 174 El-Daly H, Bower M, Naresh KN, 2010, Follicular dendritic cells in multicentric Castleman disease present human herpes virus type 8, HHV8)-latent nuclear antigen 1, LANA1, in a proportion of cases and is associated with an enhanced T-cell response. Eur J Haematol 84:133–136 El-Osta HE, Kurzrock R, 2011, Castleman’s disease: from basic mechanisms to molecular therapeutics. Oncologist 16:497–511 Casper C, 2005, The aetiology and management of Castleman disease at 50 years: translating pathophysiology to patient care. Br J Haematol 129:3–17 Dispenzieri A, Gertz MA, 2005, Treatment of Castleman’s disease. Curr Treat Options Oncol 6:255–266 Gill PS, Loureiro C, Bernstein-Singer M, Rarick MU, Sattler F, Levine AM, 1989, Clinical effect of glucocorticoids on Kaposi sarcoma related to the acquired immunodeficiency syndrome, AIDS). Ann Intern Med 110:937–940 Lajoie G, Kumar S, Min KW, Silva FG, 1995, Renal thrombotic microangiopathy associated with multicentric Castleman’s disease. Report of two cases. Am J Surg Pathol 19:1021–1028 Mylona EE, Baraboutis IG, Lekakis LJ, Georgiou O, Papastamopoulos V, Skoutelis A, 2008, Multicentric Castleman’s disease in HIV infection: a systematic review of the literature. AIDS Rev 10:25–35 Seo HY, Kim EB, Kim JW, Shin BK, Kim SJ, Kim BS, 2009, Complete remission in a patient with human herpes virus-8 negative multicentric Castleman disease using CHOP chemotherapy. Cancer Res Treat 41:104–107 Bower M, Fox P, Fife K, Gill J, Nelson M, Gazzard B, 1999, Highly active anti-retroviral therapy, HAART, prolongs time to treatment failure in Kaposi’s sarcoma. AIDS 13:2105–2111 Zietz C, Bogner JR, Goebel FD, Lohrs U, 1999, An unusual cluster of cases of Castleman’s disease during highly active antiretroviral therapy for AIDS. N Engl J Med 340:1923–1924 Berezne A, Agbalika F, Oksenhendler E, 2004, Failure of cidofovir in HIV-associated multicentric Castleman disease. Blood 103:4368–4369 Casper C, Nichols WG, Huang ML, Corey L, Wald A, 2004, Remission of HHV-8 and HIV-associated multicentric Castleman disease with ganciclovir treatment. Blood 103:1632–1634 Bower M, Powles T, Williams S et al, 2007, Brief communication: rituximab in HIV-associated multicentric Castleman disease. Ann Intern Med 147:836–839 Gerard L, Berezne A, Galicier L et al, 2007, Prospective study of rituximab in chemotherapy-dependent human immunodeficiency virus associated multicentric Castleman’s disease: ANRS 117 CastlemaB Trial. J Clin Oncol 25:3350–3356 Law AB, Ryan G, Lade S, Prince HM, 2010, Development of Kaposi’s sarcoma after complete remission of multicentric Castlemans disease with rituximab therapy in a HHV8-positive, HIV-negative patient. Int J Hematol 91:347–348 Nishimoto N, Sasai M, Shima Y et al, 2000, Improvement in Castleman’s disease by humanized anti-interleukin-6 receptor antibody therapy. Blood 95:56–61 Nishimoto N, Kanakura Y, Aozasa K et al, 2005, Humanized antiinterleukin- 6 receptor antibody treatment of multicentric Castleman disease. Blood 106:2627–2632 Matsuyama M, Suzuki T, Tsuboi H et al, 2007, Anti-interleukin-6 receptor antibody, tocilizumab, treatment of multicentric Castleman’s disease. Intern Med 46:771–774 Higuchi T, Nakanishi T, Takada K et al, 2010, A case of multicentric Castleman’s disease having lung lesion successfully treated with humanized anti-interleukin-6 receptor antibody, tocilizumab. J Korean Med Sci 25:1364–1367 Muzes G, Sipos F, Csomor J, Sreter L, 2012, Successful tocilizumab treatment in a patient with HHV8-positive and HIVnegative multicentric Castleman’s disease of plasma cell type nonresponsive to R-CVP therapy., DOI 10.1111/apm.12029 van Rhee F, Fayad L, Voorhees P et al, 2010, Siltuximab, a novel anti-interleukin-6 monoclonal antibody, for Castleman’s disease. J Clin Oncol 28:3701–3708 Starkey CR, Joste NE, Lee FC, 2006, Near-total resolution of multicentric Castleman disease by prolonged treatment with thalidomide. Am J Hematol 81:303–304 Hess G,Wagner V, Kreft A, Heussel CP, Huber C, 2006, Effects of bortezomib on pro-inflammatory cytokine levels and transfusion dependency in a patient with multicentric Castleman disease. Br J Haematol 134:544–545 Sobas MA, Alonso Vence N, Diaz Arias J, Bendana Lopez A, Fraga Rodriguez M, Bello Lopez JL, 2010, Efficacy of bortezomib in refractory form of multicentric Castleman disease associated to poems syndrome, MCD-POEMS variant). Ann Hematol 89:217– 219 Galeotti C, Tran TA, Franchi-Abella S, Fabre M, Pariente D, Kone-Paut I, 2008, IL-1RA agonist, anakinra, in the treatment of multifocal castleman disease: case report. J Pediatr Hematol Oncol 30:920–924 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 345 EP 351 DI 10.1007/s12253-013-9619-z PG 7 ER PT J AU Turi, K Barabas, P Csurgay, K Lehner, Gy Lorincz, Nemeth, Zs AF Turi, Katalin Barabas, Peter Csurgay, Katalin Lehner, Gyorgy Lorincz, Adam Nemeth, Zsolt TI An Analysis of the Epidemiological and Etiological Factors of Oral Tumors of Young Adults in a Central-Eastern European Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Young adult; Oral cancer; Epidemiology; Etiology; Human papilloma virus ID Young adult; Oral cancer; Epidemiology; Etiology; Human papilloma virus AB The etiology of tumors in young age is not precisely known yet, but studies on the topic generally agree that in this group of patients the traditionally known behavioural risk factors (tobacco and alcohol abuse) play no or a significantly less important role. Oral squamous cell carcinoma occurring at a young age is a topic of utmost importance that is extensively and intensively researched as, while the overall incidence of oral cancer is decreasing worldwide, that of squamous cell carcinoma diagnosed in young adults is steadily increasing. The present article aims at presenting the main questions and characteristics of tumors in young adults in Central-Eastern Europe and in developed West European countries as contrasted to tumors found in middle aged and elderly patients. Factors influencing the development of oral cancer include regulatory factors of the cell cycle, the inherited vulnerability of the genetic code of certain proteins and the presence of HPV infection with an oncogenic genotype. The connections of HPV infection and genetic damages are studied intensively. It is known that the prevalence of oral HPV infections is growing with a background of potentially changing sexual habits. It is debated, however, whether smoking and alcohol consumption could have a connection to HPV-associated oral cancer and whether the spread of HPV in itself could be an explanation for the growing occurrence of young-age tumors. There is no consensus in the literature as to the prognostic significance of age. Some research groups have found a better life expectancy for young patients, while other authors found a worse prognosis for these patients. It is known that the prognosis of head and neck tumors, the prevalence of HPV infections as well as genetic mutations show regional and ethnic variations. This might be explained by differences in the degree of development of a preventive system, in the quality of care and in the attitudes of young patients towards visiting a doctor. The study is made difficult by incomparable patient selection criteria as well as by the question of the intraoral localisation of tumors as an independent risk factor. C1 [Turi, Katalin] Semmelweis University, Department of Oral and Maxillofacial Surgery, Maria str. 52, 1085 Budapest, Hungary. [Barabas, Peter] Semmelweis University, Department of Oral and Maxillofacial Surgery, Maria str. 52, 1085 Budapest, Hungary. [Csurgay, Katalin] Semmelweis University, Department of Oral and Maxillofacial Surgery, Maria str. 52, 1085 Budapest, Hungary. [Lehner, Gyorgy] Semmelweis University, Department of Oral and Maxillofacial Surgery, Maria str. 52, 1085 Budapest, Hungary. [Lorincz, Adam] Semmelweis University, Department of Oral and Maxillofacial Surgery, Maria str. 52, 1085 Budapest, Hungary. [Nemeth, Zsolt] Semmelweis University, Department of Oral and Maxillofacial Surgery, Maria str. 52, 1085 Budapest, Hungary. RP Nemeth, Zs (reprint author), Semmelweis University, Department of Oral and Maxillofacial Surgery, 1085 Budapest, Hungary. EM nemeth.zsolt@dent.semmelweis-univ.hu CR Chen JK, Eisenberg E et al, 1991, Changing trends in oral cancers in the United States, 1935 to 1985: a Connecticut study. J Oral Maxillofac Surg 49:1152–1158 Annertz K, Anderson H et al, 2002, Incidence and survival of squamous cell carcinoma of the tongue in Scandinavia, with special reference to young adults. Int J Cancer 101:95–99 Llewellyn CD, Johnson NW, Warnakulasuriya KA, 2004, Risk factors for oral cancer in newly diagnosed patients aged 45 years and younger: a case–control study in Southern England. J Oral Pathol Med 33:525–532 BDA, 2000, Opportunistic oral cancer screening. British Dental Association Occasional Paper No 6, London Schantz SP, Yu GP, 2002, Head and neck cancer incidence trends in young Americans, 1973–1997, with a special analysis for tongue cancer. Arch Otolaryngol Head Neck Surg 128:268–274 National Cancer Registry, http://www.honcology.hu/portal/page/ portal/OOI/MEDICAL_ATTENDANCE/medical_departments/ MD_901 Muller S, Pan Yet al, 2008, Changing trends in oral squamous cell carcinoma with particular reference to young patients:1971–2006. The Emory university experience. Head Neck Pathol 2:60–66 Siriwardena BS, Tilakaratne A et al, 2006, Demographic, aetiological and survival differences of oral squamous cell carcinoma in the young and the old in Sri Lanka. Oral Oncol 42:831–836 Brown LM, Check DP, Devesa SS, 2012, Oral cavity and pharynx cancer incidence trends by subsite in the United States: changing gender patterns. J Oncol 2012:649498., DOI 10.1155/2012/649498 Horn C, Thaker HM, Tampakopoulou DA et al, 2001, Tongue lesions in the pediatric population. Otolaryngol Head Neck Surg 124:164–169 Siegelmann-Danieli N, Hanlon A, Ridge JA et al, 1998, Oral tongue cancer in patients less than 45 years old: institutional experience and comparison with older patients. J Clin Oncol 16:745–753 Llewellyn CD, Johnson NW, Warnakulasurya KA, 2001, Risk factors for squamosus cell carcinoma of the oral cavity in young people-a comprehensive literature review. Oral Oncol 37:401–418 http://ec.europa.eu/public_opinion/archives/ebs/ebs_332_en.pdf http://ec.europa.eu/health/tobacco/products/index_en.htm Hashibe M, Brennan P, Chuang SC et al, 2009, Interaction between alcohol and tobacco use and the risk of head and neck cancer: pooled analysis in the international head and neck cancer epidemiology consortium. Cancer Epidemiol Biomarkers Prev 18:541–550 Llewellyn CD, Linklater K, Bell J et al, 2004, An analysis of risk factors for oral cancer in young people: a case–control study. Oral Oncol 40:304–313 Bachar G, Hod R, Goldstein DP, Irish JC et al, 2011, Outcome of oral tongue squamosus cell carcinoma in patients with or without the known risk factors. Oral Oncol 47:45–50 Rothenberg SM, Ellisen LW, 2012, The molecular pathogenesis of head and neck squamous cell carcinoma. J Clin Invest 122:1951– 1957 Popvic B, Jekic B, Novakovic I et al, 2010, Cancer gene alterations and HPV infection in oral squamosus cell carcinoma. Int J Oral Maxillofac Surg 39:909–915 Paterson IC, Eveson JW, Prime SS, 1996, Molecular changes in oral cancer may reflect aetiology and ethnic origin. Eur J Cancer B Oral Oncol 32:150–153 Sheikh Ali MA, Gunduz M, Nagatsuka H et al, 2008, Expression and mutation analysis of epidermal growth factor receptor in head and neck squamous cell carcinoma. Cancer Sci 99:1589–1594 Szabo B, Nelhubel AG, Karpati A et al, 2011, Clinical significance of genetic alterations and expression of epidermal growth factor receptor, EGFR, in head and neck squamous cell carcinomas. Oral Oncol 47:487–496 Hama T, Yuza Y, Saito Y et al, 2009, Prognostic significance of epidermal growth factor receptor phosphorylation and mutation in head and neck squamous cell carcinoma. Oncologist 14:900–908 Licitra L, Mesia R, Rivera F et al, 2011, Evaluation of EGFR gene copy number as a predictive biomarker for the efficacy of cetuximab in combination with chemotherapy in the first-line treatment of recurrent and/or metastatic squamous cell carcinoma of the head and neck: EXTREME study. Ann Oncol 22:1078–1087 Smilek P, Neuwirthova J, Jarkovsky J et al, 2012, Epidermal growth factor receptor, EGFR, expression and mutations in the EGFR signaling pathway in correlation with anti-EGFR therapy in head and neck squamous cell carcinomas. Neoplasma 59:508–515 Timar J, Csuka O, Remenar E et al, 2005, Progression of head and neck squamous cell cancer. Cancer Metastasis Rev 24:107–127 Csuka O, Remenar E, Koronczay K et al, 1997, Predictive value of p53, Bcl2 and bax in the radiotherapy of head and neck cancer. Pathol Oncol Res 3:204–210 Aswini YB, 2009, The genomics of oral cancer and wound healing. J Indian Soc Pedod Prevent Dent 27:2–5 Alvasiri NS, Mehdi MJ, Alam MS et al, 2012, PTEN-mediated Akt activation contributes to the reduced apoptosis among Indian oral squamosus carcinoma patients. J Cancer Res Clin Oncol 138:103–109 Lu Y, Lin YZ, Lapushin R et al, 1999, The PTEN/MMAC1/TEP tumor suppressor gene decreases cell growth and induces apoptosis and inoikis in breast cancer cells. Oncogene 18:7034–7045 Weng LP, Brown JL, Eng C, 2001, PTEN induces apoptosis and cell cycle arrest through phosphoinositol-3-kinase/Akt-dependent and -independent pathways. Hum Mol Genet 10:237–242 Wang Z, Sturgis EM, Zhang Y et al, 2011, Combined p53-related genetic variant together with HPV infection increase oral cancer risk. Int J Cancer 131:251–258 O’Regan EM, Toner ME, Finn SP et al, 2008, p16(INK4A, Genetic and epigenetic profiles differ in relation to age and site in head and neck squamosus cell carcinomas. Hum Pathol 39:452–458 Harris SL, Thorne LB, Seaman WT et al, 2011, Association of p16(INK4a, overexpression with improved outcomes in young patients with squamous cell cancers of the oral tongue. Head Neck 33:1622–1627 Zhang ZJ, Hao K, Shi R et al, 2011, Glutathione S-tranferase M1, GSTM1, and glutathione S-transferase T1, GSTT1, null polimorphisms, smoking, and their interaction in oral cancer: a huge review and meta-analysis. Am J Epidemiol 173:847–857 Gawecki W, Kostrzewska-Poczekai M, Gajecka M et al, 2007, The role of genetic factor in etiopathogenesis of squamosus cell carcinoma of the head and neck in young adults. Eur Arch Otorhinolaryngol 12:1459–1465 Toner M, O’Regan EM, 2009, Head and neck squamosus cell carcinoma in the young: a spectrum or a distinct group? part 2. Head Neck Pathol 3:249–251 Lukits J, Remenar E, Raso E et al, 2007, Molecular identification, expression and prognostic role of estrogen- and progesterone receptors in head and neck cancer. Int J Oncol 30:155–160 Remenar E, Szamel I, Budai B et al, 2007, Increase of hypophyseal hormone levels in male head and neck cancer patients. Pathol Oncol Res 13:341 Timar J, Forster-Horvath C, Lukits J et al, 2003, The effect of leukocyte interleukin injection, multikine®, treatment on the peritumoral and intratumoral subpopulation of mononuclear cells and on tumor epithelia:a possible New approach to augmenting sensitivity to radiation therapy and chemotherapy in oral cancer-a multicenter phase I/II clinical trial. Laryngoscope 113:2206–2217 Stransky N, Egloff AM, Tward AD et al, 2011, The mutational landscape of head and neck squamous cell carcinoma. Science 333:1157–1160 Kumaraswamy KL, Vidhya M, 2011, Human papilloma virus and oral infections: an update. J Cancer Res Ther 7:120–127 Smith EM, Hoffman HT, Summersgill KS et al, 1998, Human papilloma virus and risk of oral cancer. Laryngoscope 108:1098– 1103 Gillison ML, Broutian T et al, 2012, Prevalence of oral HPV infection in the United States, 2009–2010. JAMA 307:693–703., DOI 10.1001/jama.2012.101 D’Souza G, Kreimer AR, Viscidi R et al, 2007, Case-controll study of human papillomavirus and oropharyngeal cancer. N Engl J Med 356:1944–1956 Cleveland JL, Junger ML et al, 2011, The connection between human papillomavirus and oropharyngeal squamosus cell carcinomas in the United States: implications for dentistry. J Am Dent Assoc 142:915–924 Syrjanen S, 2010, The role of human papilloma virus infection in head and neck cancers. Ann Oncol 21:243–245 Rombaldi RL, Serafini EP et al, 2009, Perinatal transmission of human papillomavirus DNA. Virol J 6:83–95 Rombaldi RL, Serafini EP et al, 2008, Transplacental transmission of human papillomavirus. Virol J 5:106–120 Kreimer AR, Glifford GM, Boyle P et al, 2005, Human papillomavirus types in head and neck squamosus carcinomas worldwide: a systematic review. Cancer Epidemiol Biomark Prev 14:467–475 Cerovac C, Sarcevic B, Kralj Z et al, 1996, Detection of human papillomavirus, HPV, type 6, 16 and 18 in head and neck squamosus cell carcinomas by in situ hybridization. Neoplasma 43:185–194 Klozar J, Kratochvil V, Salakova M, 2008, HPV status and regional metastasis in the prognosis of oral and oropharyngeal cancer. Eur Arch Otolaryngol 265:75–82 Kansky AA, Poljak M, Seme K et al, 2003, Human papillomavirus DNA in oral squamosus cell carcinomas and normal oral mucosa. Acta Virol 47:11–16 Kansky AA, Seme K, Maver PJ et al, 2006, Human papillomaviruses, HPV, in tissue specimens of oral squamosus cell papillomas and normal oral mucosa. Anticancer Res 26:3197–3202 Szentirmay Z, Polus Z, Tamas L et al, 2005, Human papillomavirus in head and neck cancer: molecular biology and clinicopathological correlations. Cancer Metastasis Rev 24:19–34 Joo JH, Jung CK et al, 2010, High-risk human papillomavirus and cervical lymph node metastasis in patient with oropharyngeal cancer. Head Neck 34:10–14 Lill C, Kornek G et al, 2011, Survival of patients with HPVpositive oropharyngeal cancer after radiochemotherapy is significantly enhanced. Wien Klin Wochenschr 123:215–221 Kozomara RJ, Brankovic-Magic MV, Jovic NR et al, 2007, Prognostic significance of TP53 mutations in oral squamous cell carcinoma with human papilloma virus infection. Int J Biol Markers 22:252–257 Hong AM, Dobbins TA et al, 2010, Human papillomavirus predicts outcome in oropharyngeal cancer in patients treated primarily with surgery or radiation therapy. Br J Cancer 103:1510–1517 Na II, Kang HJ, Cho SY et al, 2007, EGFR mutations and human papillomavirus in squamous cell carcinoma of tongue and tonsil. Eur J Cancer 43:520–526 Byers RM, 1975, Squamous cell carcinoma of the oral tongue in patients less than thirty years of age. Am J Surg 130:475–478 Garavello W, Spreafico R et al, 2007, Oral tongue cancer in young patients: a matched analysis. Oral Oncol 43:894–897 Hagemann M, Zbaeren P, 2002/2003, Squamous Cell Carcinoma of the Tongue in Young. Oto-Rhino-Laryngol Nova 12:295–299 Sarkaria JN, Harari PM, 1994, Oral tongue cancer in young adults less than 40 years of age: rational for agressive therapy. Head Neck 16:107–111 Mallet Y, Avalaos N et al, 2009, Head and neck cancer in young people: a series of 52 SCCs of the oral tongue in patients aged 35 years or less. Acta Otolaryngol 129:1503– 1508 Gawecki W, Szyfter K, Szyfter W et al, 2007, Clinical and histopatological analysis of squamosus cell carcinoma of the head and neck in young adults. Otolaryngol Pol 61:11–16 Pytinia KB, Grant JR, Etzel CJ et al, 2004, Matched analysis of survival in patients with squamosus cell carcinoma of the head and neck diagnosed before and after 40 years of age. Arch Otolanryngol Head Neck Surg 130:869–873 Falaki F, Dalirsani Z, Pakfetrat A et al, 2011, Clinical and hystopatological analysis of oral squamosus cell carcinoma of young patients in Mashad, Iran: A retrospective study and review of literatures. Med Oral Patol Oral Cir Bucal 16:473–477 Zigon G, Berrino F, Gatta G et al, 2011, Prognosis for head and neck cancers in Europe diagnosed in 1995–1999: a populationbased study. Ann Oncol 22:165–174 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 353 EP 363 DI 10.1007/s12253-013-9628-y PG 11 ER PT J AU Pecina-Slaus, N AF Pecina-Slaus, Nives TI Merlin, the NF2 Gene Product SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE NF2 gene; Merlin; Schwannoma; Neurinoma ID NF2 gene; Merlin; Schwannoma; Neurinoma AB Merlin, the protein product of NF2 gene, is one of the most versatile tumor suppressors capable of integrating different mechanisms that regulate cell proliferation, motility, survival and signaling pathways underlying and governing those mechanisms. Merlin is considered a member of the band 4.1 families of cytoskeleton-associated proteins also called ERM family and acts as tumor suppressor. The main cause for transformation of Schwann cells into schwannomas is credited to the inactivation of the neurofibromin 2 (NF2) gene and the consecutive loss of its protein merlin. Recent scientific advances improved our understanding of pathogenic mechanisms involving NF2 gene. The present review brings genetic properties of NF2 gene, molecular characteristics of merlin, summarizes mutational spectra and explains merlin’s multifunctional roles regarding its involvement in neurofibromatosis associated tumorigenesis. C1 [Pecina-Slaus, Nives] University of Zagreb, School of Medicine, Croatian Institute for Brain Research, Laboratory of Neurooncology, Salata 12, 10000 Zagreb, Croatia. RP Pecina-Slaus, N (reprint author), University of Zagreb, School of Medicine, Croatian Institute for Brain Research, Laboratory of Neurooncology, 10000 Zagreb, Croatia. EM nina@mef.hr CR Rouleau GA, Merel P, Lutchman M, Sanson M, Zucman J, Marineau C, Hoang-Xuan K, Demczuk S, Desmaze C, Plougastel B et al, 1993, Alteration in a new gene encoding a putative membrane-organizing protein causes neurofibromatosis type 2. Nature 363:515–521 Trofatter JA, MacCollin MM, Rutter JL, Murrell JR, Duyao MP, Parry DM, Eldridge R, Kley N, Menon AG, Pulaski K et al, 1993, A novel moesin-, ezrin-, radixin-like gene is a candidate for the neurofibromatosis 2 tumor suppressor. Cell 72:791–800 Gusella JF, Ramesh V, MacCollin M, Jacoby LB, 1999, Merlin: the neurofibromatosis 2 tumor suppressor. Biochim Biophys Acta 1423:M29–M36 Stamenkovic I, Yu Q, 2010, Merlin, a “magic” linker between the extracellular cues and intracellular signaling pathways that regulate cell motility, proliferation, and survival. Curr Protein Pept Sci 11:471–484 Sughrue MI, Yeung AH, Rutkowski MJ, Cheung SW, Parsa AT, 2011, Molecular biology of familial and sporadic vestibular schwannomas: implications for novel therapeutics. J Neurosurg 114:359–366 Ammoun S, Hanemann CO, 2011, Emerging therapeutic targets in schwannomas and other merlin-deficient tumors. Nat Rev Neurol 7:3992–3999 Zhou L, Hanemann CO, 2012, Merlin, a multi-suppressor from cell membrane to the nucleus. FEBS Lett 586:1403–1408 Entrez, the life science search engine, http://www.ncbi.nlm.nih.gov/ sites/gquery) McClatchey AI, 2007, Neurofibromatosis. Annu Rev Pathol Mech Dis 2:191–216 Shimizu T, Seto A, Maita N, Hamada K, Tsukita S, Hakoshima T, 2002, Structural basis for neurofibromatosis type 2. Crystal structure of the merlin FERM domain. J Biol Chem 277:10332–10336 Hanemann CO, 2008, Magic but treatable? Tumours due to loss of Merlin. Brain 131:606–615 UniProtKB/Swiss-Prot, http://www.uniprot.org/uniprot/P35240# section_comments) Laulajainen M, Melikova M, Muranen T, Carpen O, Gronholm M, 2012, Dictinct overlapping sequences at the carboxy-terminus of merlin regulate its tumor suppressor and morphogenic activity. J Cell Mol Med 16:2161–2175 Carroll SL, 2012, Molecular mechanisms promoting the pathogenesis of Schwann cell neoplasms. Acta Neuropathol 123:321–348 Alfthan K, Heiska L, Gronholm M, Renkema GH, Carpen O, 2004, Cyclic AMP-demendent protein kinase phosphorylates merlin at serine 518 independently of p21 activated kinase and promotes merlin-ezrin heterodimerization. J Biol Chem 279:18559–18566 McClatchey AI, Saotome I, Ramesh V, Gusella JF, Jacks T, 1997, The NF2 tumor suppressor gene product is essential for extraembryonic development immediately prior to gastrulation. Genes Dev 11:1253–1265 Hanemann CO, Evans DG, 2006, News on the genetics, epidemiology, medical care and translational research of Schwannomas. J Neurol 253:1533–1541 Giovannini M, Robanus-Maandag E, van der Valk M, Niwa- Kawakita M, Abramowski V, Goutebroze L, Woodruff JM, Berns A, Thomas G, 2000, Conditional biallelic NF2 mutation in the mouse promotes manifestations of human neurofibromatosis type 2. Genes Dev 14:1617–1630 Lutchman M, Rouleau GA, 1995, The neurofibromatosis type 2 gene product, schwannomin, suppresses growth of NIH 3T3 cells. Cancer Res 55:2270–2274 Tikoo A, Varga M, Ramesh V, Gusella J, Maruta H, 1994, An anti- Ras function of neurofibromatosis type 2 gene product, NF2/ Merlin). J Biol Chem 269:23387–23390 Scoles DR, 2008, The merlin interacting proteins reveal multiple targets for NF2 therapy. Biochim Biophys Acta 1785:32–54 Morrison H, Sherman LS, Legg J, Banine F, Isacke C, Haipek CA, Gutmann DH, Ponta H, Herrlich P, 2001, The NF2 tumor suppressor gene product, merlin, mediates contact inhibition of growth through interactions with CD44. Genes Dev 15:968–980 Okada T, Lopez-Lago M, Giancotti FG, 2005, Merlin/NF-2 mediates contact inhibition of growth by suppressing recruitment of Rac to the plasma membrane. J Cell Biol 171:361–371 Lallemand D, Curto M, Saotome I, Giovannini M, McClatchey AI, 2003, NF2 deficiency promotes tumorigenesis and metastasis by destabilizing adherens junctions. Genes Dev 17:1090–1100 Li W, Cooper J, Karajannis MA, Giancotti FG, 2012, Merlin: a tumour suppressor with functions at the cell cortex and in the nucleus. EMBO Rep 13:204–215 Kim H, Kwak NJ, Lee JY, Choi BH, Lim Y, Ko YJ, Kim YH, Huh PW, Lee KH, Rha HK, Wang YP, 2004, Merlin neutralizes the inhibitory effect of Mdm2 on p53. J Biol Chem 279:7812–7818 Fernandez-Valle C, Tang Y, Ricard J, Rodenas-Ruano A, Taylor A, Hackler E, Biggerstaff J, Iacovelli J, 2002, Paxillin binds schwannomin and regulates its density-dependent localization and effect on cell morphology. Nat Genet 31:354–362 Morrison H, Sperka T, Manent J, Giovannini M, Ponta H, Herrlich P, 2007, Merlin/neurofibromatosis type 2 suppresses growth by inhibiting the activation of Ras and Rac. Cancer Res 67:520–527 Fraenzer JT, Pan H, Minimo L Jr, Smith GM, Knauer D, Hung G, 2003, Overexpression of the NF2 gene inhibits schwannoma cell proliferation through promoting PDGFR degradation. Int J Oncol 23:1493–1500 Okada M, Wang Y, Jang SW, Tang X, Neri LM, Ye K, 2009, Akt phosphorylation of merlin enhances its binding to phosphatidylinositols and inhibits the tumor-suppressive activities of merlin. Cancer Res 69:4043–4051 Bosco EE, Nakai Y, Hennigan RF, Ratner N, Zheng Y, 2010, NF2- deficient cells depend on the Rac1-canonical Wnt signaling pathway to promote the loss of contact inhibition of proliferation. Oncogene 29:2540–2549 Zhou L, Ercolano E, Ammoun S, Schmid MC, Barczyk MA, Hanemann CO, 2011, Merlin-deficient human tumors show loss of contact inhibition and activation of Wnt/β-Catenin signaling linked to the PDGFR/Src and Rac/PAK pathways. Neoplasia 13:1101–1112 Lau YKI, Murray LB, Houshmandi SS, Nu Y, Gutmann DH, Yu Q, 2008, Merlin is a potent inhibitor of glioma growth. Cancer Res 68:5733–5742 Evans DG, Huson SM, Donnai D, Neary W, Blair V, Teare D, Newton V, Strachan T, Ramsden R, Harris R, 1992, A genetic study of type 2 neurofibromatosis in the United Kingdom. I. Prevalence, mutation rate, fitness, and confirmation of maternal transmission effect on severity. J Med Genet 29:841–846 Jacoby LB, MacCollin M, Barone R, Ramesh V, Gusella JF, 1996, Frequency and distribution of NF2 mutations in schwannomas. Genes Chromosom Cancer 17:45–55 Irving RM, Harada T, Moffat DA, Hardy DG, Whittaker JL, Xuereb JH, Maher ER, 1997, Somatic neurofibromatosis type 2 gene mutations and growth characteristics in vestibular schwannoma. Am J Otol 18:754–760 The Human Gene Mutation Database, http://www.hgmd.cf.ac.uk/ ac/gene.php?gene=NF2) Antinheimo J, Sallinen SL, Sallinen P, Haapasalo H, Helin H, Horelli-Kuitunen N, Wessman M, Sainio M, Jaaskelainen J, Carpen O, 2000, Genetic aberrations in sporadic and neurofibromatosis 2, NF2)-associated Schwannomas studied by comparative genomic hybridization, CGH). Acta Neurochir 142:1099–1105 Ahronowitz I, Xin W, Kiely R, Sims K, MacCollin M, Nunes FP, 2007, Mutational spectrum of NF2 gene: a meta-analysis of 12 years of research and diagnostic laboratory findings. Hum Mutat 28:1–12 Welling DB, Akhmametyeva EM, Daniels RL, Lasak JM, Zhu L, Miles-Markley BA, Chang LS, 2000, Analysis of the human neurofibromatosis type 2 gene promoter and its expression. Otolaryngology 123:413–418 Gutmann DH, Geist RT, Xu H, Kim JS, Saporito-Irwin S, 1998, Defects in neurofibromatosis 2 protein function can arise at multiple levels. Hum Mol Genet 7:335–345 Fong B, Barkhoudarian G, Pezeshkian P, Parsa AT, Gopen Q, Yang I, 2011, The molecular biology and novel treatments of vestibular schwannomas. J Neurosurg 115:906–914 Scheithauer BW, Louis DN, Hunter S, Woodruff JM, Antonescu CR, 2007, Tumours of the cranial and paraspinal nerves. Schwannoma. In: Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, eds, WHO classification of tumours of the central nervous system, 4th edn. International Agency for Research on Cancer, Lyon, pp 152–155 Ghosh A, Talwar OP, Pradhan SV, 2010, Tumour and tumour-like conditions of peripheral nerve origin: ten years’ experience. Kathmandu Univ Med J 29:97–101 Celis-Aguilar E, Lassaletta L, Torres-MartinM, Rodrigues FY, Nistal M, Castresana JS, Gavilan J, Ray JA, 2012, The molecular biology of vestibular schwannomas and its association with hearing loss: a review. Genet Res Int. Article ID 856157:10., DOI 10.1155/2012/ 856157 Irving RM, Moffat DA, Hardy DG, Barton DE, Xuereb JH, Maher ER, 1993, Molecular genetic analysis of the mechanism of tumorigenesis in acoustic neuroma. Arch Otolaryngol Head Neck Surg 119:1222–1228 Hitotsumatsu T, Iwaki T, Kitamoto T, Mizoguchi M, Suzuki SO, Hamada Y, Fukui M, Tateishi J, 1997, Expression of neurofibromatosis 2 protein in human brain tumors: an immunohistochemical study. Acta Neuropathol 93:225–232 Lee DJ, Maseyesva B, Westra W, Long D, Niparko JK, Califano J, 2006, Microsatellite analysis of recurrent vestibular Schwannoma, acoustic neuroma, following stereotactic radiosurgery. Otol Neurotol 27:213–219 Pecina-Slaus N, Zeljko M, Pecina HI, Nikuseva Martic T, Bacic N, Tomas D, Hrascan R, 2012, Frequency of loss of heterozygosity of the NF2 gene in schwannomas from Croatian patients. Croat Med J 53:321–327 Bian LG, Sun QF, Tirakotai W, Zhao WG, Shen JK, Luo QZ, Bertalanffy H, 2005, Loss of heterozygosity on chromosome 22 in sporadic schwannoma and its relation to the proliferation of tumor cells. Chin Med J, Engl, 118:1517–1524 Hadfield KD, Smith MJ, Urquhart JE, Wallace AJ, Bowers NL, King AT, Rutherford SA, Trump D, Newman WG, Evans DG, 2010, Rates of loss of heterozygosity and mitotic recombination in NF2 schwannomas, sporadic vestibular schwannomas and schwannomatosis schwannomas. Oncogene 29:6216–6221 Mantripragada KK, Buckley PG, Benetkiewicz M, De Bustos C, Hirvela C, Jarbo C, Bruder CE, Wensman H, Mathiesen T, Nyberg G, Papi L, Collins VP, Ichimura K, Evans G, Dumanski JP, 2003, High-resolution profiling of an 11 Mb segment of human chromosome 22 in sporadic schwannoma using array-CGH. Int J Oncol 22:615–622 Warren C, James LA, Ramsden RT, Wallace A, Baser ME, Varley JM, Evans DG, 2003, Identification of recurrent regions of chromosome loss and gain in vestibular schwannomas using comparative genomic hybridization. J Med Genet 40:802–806 Koutsimpelas D, Felmeden U, Mann WJ, Brieger J, 2011, Analysis of cytogenetic aberrations in sporadic vestibular schwannoma by comparative genomic hybridization. J Neurooncol 103:437–443 Aarhus M, Bruland O, Saetran HA, Mork SJ, Lund-Johansen M, Knappskog PM, 2010, Global gene expression profiling and tissue microarray reveal novel candidate genes and down-regulation of the tumor suppressor gene CAV1 in sporadic vestibular schwannomas. Neurosurgery 67:998–1019 Ueki K, Wen-Bin C, Narita Y, Asai A, Kirino T, 1999, Tight association of loss of merlin expression with loss of heterozygosity at chromosome 22q in sporadic meningiomas. Cancer Res 59:5995–5998 Kros J, de Greve K, van Tilborg A, Hop W, Pieterman H, Avezaat C, Lekane dit Deprez R, Zwarthoff E, 2001, NF2 status of meningiomas is associated with tumour localization and histology. J Pathol 194:367–372 Fuller CE, Perry A, 2005, Molecular diagnostics in central nervous system tumors. Adv Anat Pathol 12:180–194 Claus EB, Bondy ML, Wiemels JL, Wrensch M, Black PM, 2005, Epidemiology of intracranial meningioma. Neurosurgery 57:1088–1094 Riemenschneider MJ, Perry A, Reifenberger G, 2006, Histological classification and molecular genetics of meningiomas. Lancet Neurol 5:1045–1054 Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P, 2007, The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114:97–109 Lee JY, Finkelstein S, Hamilton RL, Rekha R, King JT Jr, Omalu B, 2004, Loss of heterozygosity analysis of benign, atypical, and anaplastic meningiomas. Neurosurgery 55:1163–1173 Ragel BT, Jensen RL, 2005, Molecular genetics of meningiomas. Neurosurg Focus 119:E9 Barnholtz-Sloan JS, Kruchko C, 2007, Meningiomas: causes and risk factors. Neurosurg Focus 23:E2 Simon M, Bostrom JP, Hartmann C, 2007, Molecular genetics of meningiomas: from basic research to potential clinical applications. Neurosurgery 60:787–798 Pecina-Slaus N, Nikuseva Martic T, Deak AJ, Zeljko M, Hrascan R, 2010, Genetic and protein changes of E-cadherin in meningiomas. J Cancer Res Clin Oncol 136:695–702 Hulsebos TJ, Plomp AS, Wolterman RA, Robanus-Maandag EC, Baas F, Wesseling P, 2007, Germline mutation of INI1/ SMARCB1 in familial Schwannomatosis. Am J Hum Genet 80:805–810 Plotkin SR, Blakeley JO, Evans DG, Hanemann CO, Hulsebos TJ, Hunter-Schaedle K, Kalpana GV, Korf B, Messiaen L, Papi L, Ratner N, Sherman LS, Smith MJ, Stemmer-Rachamimov AO, Vitte J, Giovannini M, 2013, Update from the 2011 International Schwannomatosis Workshop: from genetics to diagnostic criteria. Am J Med Genet 161:405–416 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 365 EP 373 DI 10.1007/s12253-013-9644-y PG 9 ER PT J AU Jenei, Z Bardi, E Magyar, TM Horvath, Paragh, Gy Kiss, Cs AF Jenei, Zoltan Bardi, Edit Magyar, Tunde Maria Horvath, Agnes Paragh, Gyorgy Kiss, Csongor TI Anthracycline Causes Impaired Vascular Endothelial Function and Aortic Stiffness in Long Term Survivors of Childhood Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Childhood cancer; Survivors; Endothelial dysfunction; Stiffness ID Childhood cancer; Survivors; Endothelial dysfunction; Stiffness AB Vascular and endothelial functions were investigated in long term survivors of childhood cancer exposed to anthracycline treatment. We enrolled 96 long-term survivors (57 males and 39 females, mean age 14.9±5.3 year) of different childhood cancers and 72 age-, sex-, bodyweightand blood pressure matched controls (39 males and 33 females, mean age 13.7±4.9 year). Aortic stiffness was characterized by echocardiography. Brachial artery endothelial function was assessed by flow-mediated dilatation (FMD%) and nitrate-mediated dilatation (NTG%). Results were compared between three subgroups: anthracycline treated, only chemotherapy treated and control subgroups. The cumulative anthracycline dose was less than 350 mg/ m2. The healthy control subgroup had a significantly greater FMD response (13.13±2.40 %), and lower stiffness index (2.08±0.6) than both the anthracycline (7.12±6.28 % and 6.45±3.25, respectively) and only chemotherapy treated (10.17±4.23 % and 4.12±2.32, respectively) subgroups. In the anthracycline treated subgroup a significantly (p<0.01) lower FMD% response, and higher stiffness index were detected than in the only chemotherapy treated subgroup. Higher triglyceride level, higher cumulative anthracycline dose and lower age at the start of treatment were found to be associated independently with impairment of FMD% response and aortic stiffness. We found a significant negative correlation between FMD and aortic stiffness (p<0.001) and a positive correlation between FMD and distensibility (p<0.0001) Childhood cancer long term survivors exposed to anthracycline treatment exhibit a marked preclinical vasculopathy, characterized by endothelial dysfunction and increased arterial stiffness, contributing to a deteriorated cardiovascular function. C1 [Jenei, Zoltan] University of Debrecen, Faculty of Medicine, Department of Internal Medicine, Nagyerdei krt. 98, 4025 Debrecen, Hungary. [Bardi, Edit] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-OncologyDebrecen, Hungary. [Magyar, Tunde Maria] University of Debrecen, Medical and Health Science Center, Department of NeurologyDebrecen, Hungary. [Horvath, Agnes] University of Debrecen, Faculty of Medicine, Department of Internal Medicine, Nagyerdei krt. 98, 4025 Debrecen, Hungary. [Paragh, Gyorgy] University of Debrecen, Faculty of Medicine, Department of Internal Medicine, Nagyerdei krt. 98, 4025 Debrecen, Hungary. [Kiss, Csongor] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-OncologyDebrecen, Hungary. RP Jenei, Z (reprint author), University of Debrecen, Faculty of Medicine, Department of Internal Medicine, 4025 Debrecen, Hungary. EM jenei@internal.med.unideb.hu CR Yeh ET, Tong AT, Lenihan DJ et al, 2004, Cardiovascular complications of cancer therapy. Diagnosis, pathogenesis, and management. Circulation 109:3122–3131 Shankar SM, Marina N, Hudson MM et al, 2008, Cardiovascular Disease Task Force of the Children’s Oncology Group. Monitoring for cardiovascular disease in survivors of childhood cancer: report from the Cardiovascular Disease Task Force of the Children’s Oncology Group. Pediatrics 121:387–396 Hochster H, Wasserheit C, Speyer J, 1995, Cardiotoxicity and cardioprotection during chemotherapy. Curr Opin Oncol 7:304–309 Bristow MR, Thompson PD, Martin RP, et al. Early anthracycline cardiotoxicity. Am J Dengel DR, Ness KK, Glasser SP et al, 2008, Endothelial function in young adult survivors of childhood acute lymphoblastic leukemia. J Pediatr Hematol Oncol 30:20–25 Oeffinger KC, Buchanan GR, Eshelman DA et al, 2001, Cardiovascular risk factors in young adult survivors of childhood acute lymphoblastic leukemia. J Pediatr Hematol Oncol 23:424–430 Widlansky ME, Gokce N, Keaney JF Jr et al, 2003, The clinical implications of endothelial dysfunction. J Am Coll Cardiol 42:1149–1160 Arnett DK, Evans GW, Riley WA, 1994, Arterial stiffness: a new cardiovascular risk factor? Am J Epidemiol 140:669–682 Murata T, Yamawaki H, Yoshimoto R et al, 2001, Chronic effect of doxorubicin on vascular endothelium assess by organ culture study. Life Sci 69:2685–2695 Wu S, Ko Y, Teng M et al, 2002, Adriamycin-induced cardiomyocyte and endothelial cell apoptosis: in vitro and in vivo studies. J Mol Cell Cardiol 34:1595–1607 Amy YC, Clifford C, Gary D et al, 2006, Anthracyclines cause endothelial injury in pediatric cancer patients: a pilot study. J Clin Oncol 24:925–928 Bardi E, Olah AV, Bartyik K et al, 2004, Late effects on renal glomerular and tubular function in childhood cancer survivors. Pediatr Blood Cancer 43(6):668–673 Schrappe M, Reiter A, Zimmermann M et al, 2000, Long-term results of four consecutive trials in childhood ALL performed by the ALL-BFM study group from 1981 to 1995, Berlin-Frankfurt- Munster. Leukemia 14:2205–2222 Creutzig U, Ritter J, Zimmermann M et al, 2001, Improved treatment results in high-risk pediatric acute myeloid leukemia patients after intensification with high-dose cytarabine and mitoxanthrone: results of Study Acute Myeloid Leukemia-Berlin-Frankfurt-Munster 93. J Clin Oncol 19:2705–2713 Reiter A, Schrappe M, Parwaresch R et al, 1995, Non-Hodgkin’s lymphomas of childhood and adolescence: results of a treatment stratified for biologic subtypes and stage – a report of the Berlin- Frankfurt-Munster Group. J Clin Oncol 13:359–372 Schellong G, Potter R, Bramswig J et al, 1999, High cure rates and reduced long-term toxicity in pediatric Hodgkin’s disease: the German-Austrian multicenter trial DAL-HD-90. The German- Austrian Pediatric Hodgkin’s Disease Study Group. J Clin Oncol 17:3736–3744 Marx M, Langer T, Graf N et al, 2002, Multicentre analysis of anthracycline-induced cardiotoxicity in children following treatment according to the nephroblastoma studies SIOP No. 9/GPOH and SIOP 93-01/GPOH. Med Pediatr Oncol 39:18–24 Ozaki T, Flege S, Kevric M et al, 2003, Osteosarcoma of the pelvis: experience of the Cooperative Osteosarcoma Study Group. J Clin Oncol 21:334–341 Koscielniak E, Jurgens H, Winkler K et al, 1992, Treatment of soft tissue sarcoma in childhood and adolescence. A report of the German Cooperative Soft Tissue Sarcoma Study. Cancer 70:2557–2567 Tweddle DA, Pinkerton CR, Lewis IJ et al, 2001, OPEC/OJEC for stage 4 neuroblastoma in children over 1 year of age. Med Pediatr Oncol 36:239–242 Celermajer DS, Sorensen KE, Gooch VM et al, 1992, Noninvasive detection of endothelial dysfunction in children and adults at risk for atherosclerosis. Lancet 340:1111–1115 Kelly AS, Kaiser DR, Dengel DR et al, 2004, Comparison of Bmode and echo-tracking methods of assessing flow-mediated dilation. Ultrasound Med Biol 30:1447–14 Celik T, Iyisoy A, Acikel C et al, 2008, The comparative effects of metoprolol and perindopril on aortic elasticity in young patients with prehypertension. Blood Press Monit 13:169–176 Lacombe F, Dart A, Dewar E et al, 1992, Arterial elastic properties in man: a comparison of echo-Doppler indices of aortic stiffness. Eur Heart J 13:1040–1045 Fahey M, Ko HH, Srivastava S et al, 2009, A comparison of echocardiographic techniques in determination of arterial elasticity in the pediatric population. Echocardiography 26:567–573 Chow AY, Chin C, Dahl G, Rosenthal DN, 2006, Anthracyclines cause endothelial injury in pediatric cancer patients: a pilot study. J Clin Oncol 24:925–928 Chaosuwannakit N, D’Agostino R Jr, Hamilton CA et al, 2010, Aortic stiffness increases upon receipt of anthracycline chemotherapy. J Clin Oncol 28(1):166–172 Hashimoto M, Akishita M, Eto M et al, 1995, Modulation of endothelial-dependent flow-mediated dilatation of the brachial artery by sex and menstrual cycle. Circulation 92:3431– 3435 Lewis TV, Dart AM, Chin-Dusting JP, 1999, Endotheliumdependent relaxation by acetylcholine is impaired in hypertriglyceridemic humans with normal levels of plasma LDL cholesterol. J Am Coll Cardiol 33:805–812 Lupattelli G, Lombardini R, Schillaci G et al, 2000, Flowmediated vasoactivity and circulating adhesion molecules in hypertriglyceridemia: association with small, dense LDL cholesterol particles. Am Heart J 140:521–526 Lundman P, Eriksson MJ, Stuhlinger M et al, 2001, Mild to moderate hypertriglyceridemia in young men is associated with endothelial dysfunction and increased plasma concentrations of asymmetric dimethylarginine. J Am Coll Cardiol 38:111–116 Ceriello A, Taboga C, Tonutti L et al, 2002, Evidence for an independent and cumulative effect of postprandial hypertriglyceridemia and hyperglycemia on endothelial dysfunction and oxidative stress generation. Circulation 106:1211–1218 Bae JH, Bassenge E, Lee HJ et al, 2001, Postprandial hypertriglyceridemia impairs endothelial function by enhanced oxidant stress. Atherosclerosis 115:517–523 Marjolen W, Sebastian JN, Aart-Jan L et al, 2010, The metabolic syndrome in adult survivors of childhood cancer, a Review. J Pediatr Hematol Oncol 32:171–179 de Bor H, Blok GJ, Voerman HJ et al, 1994, Serum lipid levels in growth hormone-deficient men. Metab: Clin Exptl 43:199–203 Angelina B, Olivecrona H, Ericsson S et al, 1993, Growth hormone and low-density lipoproteins. Acta Endocrinol 128, suppl2):26–28 Darzy KH, 2009, Radiation-induced hypopituitarism after cancer therapy: who, how and when to test. Nat Clin Pract Endocrinol Metab 5(2):88–89 Kitajima M, Hirai T, Maruyama N et al, 2007, Asymptomatic cystic changes in the brain of children after cranial irradiation: frequency, latency, and relationship to age. Neuroradiology 49, 5):411–417 Duquaine D, Hirsch GA, Chakrabarti A et al, 2003, Rapid-onset endothelial dysfunction with adriamycin: evidence for a dysfunctional nitric oxide synthase. Vasc Med 8:101–107 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 375 EP 383 DI 10.1007/s12253-012-9589-6 PG 9 ER PT J AU Yahyapour, Y Shamsi-Shahrabadi, M Mahmoudi, M Motevallian, A Siadati, S Shefaii, S Shirvani, ShJ Mollaie, H Monavari, HRS Keyvani, H AF Yahyapour, Yousef Shamsi-Shahrabadi, Mahmoud Mahmoudi, Mahdi Motevallian, Abbas Siadati, Sepideh Shefaii, S Shirvani, Shokri Javad Mollaie, R. Hamidreza Monavari, Hamid Reza Seyed Keyvani, Hossein TI High-Risk and Low-Risk Human Papillomavirus in Esophageal Squamous Cell Carcinoma at Mazandaran, Northern Iran SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE High-risk and Low-risk HPV; Genotyping; ESCC; Mazandaran; North of Iran ID High-risk and Low-risk HPV; Genotyping; ESCC; Mazandaran; North of Iran AB Cancers are the second most common cause of nonaccidental deaths in Iran, following cardiovascular deaths. Mazandaran, near the Caspian Littoral at north of Iran have identified as a several-high incidence area for Esophageal Squamous Cell Carcinoma (ESCC) in the world. Several associated risk factors, such as dietary and cultural habits, infectious agents, nutritional deficiencies, too much use of tobacco and alcohol and infection to certain DNA tumor viruses (HPVs), including environmental and genetic factors are attributed to this disease. To explore this issue, we analyzed HPV DNA prevalence and HPV types together in relation to tumor sites a high-incidence population. Archived tissue blocks from 46, 69 and 62 upper, middle and lower third of esophagus, respectively from ESCC patients were evaluated for the presence of HPV DNA by PCR using the degenerate HPV L1 consensus primer pairs MY09/MY11. The positive specimens were evaluated by Real-time PCR to determine HPV genotypes. From the 49 HPV positive cases, of ESCC patients, 5 (23.1%), 11 (55 %) and 9 (56.3 %) of upper, middle and lower third of ESCC specimens, respectively were positive by at least one high and one low-risk HPV genotypes. In general, HPV45 and HPV11 were the most common high- risk and low-risk HPV genotypes in HPV L1 positive cases, respectively, followed by HPV6, HPV52 and HPV39. Therefore, the high prevalence of HPV DNA in different anatomical sites of ESCC patients from the Mazandaran region in North of Iran provides more evidence for a role of HPV in this cancer. C1 [Yahyapour, Yousef] Babol University of Medical Sciences, Infectious Diseases & Tropical Medicine Research CenterBabol, Iran. [Shamsi-Shahrabadi, Mahmoud] Tehran University of Medical Sciences, Faculty of Medicine, Department of Virology and Antimicrobial Resistance Research CenterTehran, Iran. [Mahmoudi, Mahdi] School of Public Health, Tehran University of Medical Sciences, Department of Epidemiology and BiostatisticsTehran, Iran. [Motevallian, Abbas] School of Public Health, Tehran University of Medical Sciences, Department of Epidemiology and BiostatisticsTehran, Iran. [Siadati, Sepideh] Babol University of Medical Sciences, Infectious Diseases & Tropical Medicine Research CenterBabol, Iran. [Shefaii, S] Babol University of Medical Sciences, Infectious Diseases & Tropical Medicine Research CenterBabol, Iran. [Shirvani, Shokri Javad] Babol University of Medical Sciences, Infectious Diseases & Tropical Medicine Research CenterBabol, Iran. [Mollaie, R. Hamidreza] Tehran University of Medical Sciences, Faculty of Medicine, Department of Virology and Antimicrobial Resistance Research CenterTehran, Iran. [Monavari, Hamid Reza Seyed] Tehran University of Medical Sciences, Faculty of Medicine, Department of Virology and Antimicrobial Resistance Research CenterTehran, Iran. [Keyvani, Hossein] Tehran University of Medical Sciences, Faculty of Medicine, Department of Virology and Antimicrobial Resistance Research CenterTehran, Iran. RP Monavari, HRS (reprint author), Tehran University of Medical Sciences, Faculty of Medicine, Department of Virology and Antimicrobial Resistance Research Center, Tehran, Iran. EM hrmonavari@yahoo.com CR Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55(2):74–108 Naghavi M, 2000, Death report from 10 provinces in Iran. Ministry of Health, Tehran Kamangar F, Dores GM, Anderson WF, 2006, Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol 24(14):2137–2150 Li MH, Li P, Li PJ, 1980, Recent progress in research on esophageal cancer in China. Adv Cancer Res 33:173–249 Li JY, 1982, Epidemiology of esophageal cancer in China. Natl Cancer Inst Monogr 62:113–120 Siddiqi M, Preussmann R, 1989, Esophageal cancer in Kashmir— an assessment. J Cancer Res Clin Oncol 115:111–117 Kodsi BE, Wickremesinghe C, Kozinn PJ, Iswara K, Goldberg PK, 1976, Candida esophagitis: a prospective study of 27 cases. Gastroenterology 71:715–719 Jarrett WF, 1987, The leeuwenhoek lecture, 1986. Environmental carcinogens and papillomaviruses in the pathogenesis of cancer. Proc R Soc Lond B Biol Sci 231:11 Goff JS, 1988, Infectious causes of esophagitis. Annu Rev Med 39:163–169 Syrjanen KJ, 2002, HPV infections oesophageal cancer. J Clin Pathol 55:721–728 Engel LS, Chow WH, Vaughan TL, Gammon M, Risch H, Stanford J et al, 2003, Population attributable risks of esophageal and gastric cancers. J Natl Cancer Inst 95(18):1404–1413 Brown LM, Hoover R, Silverman D, Baris D, Hayes R, Swansonet GM et al, 2001, Excess incidence of squamous cell esophageal cancer among US black men: role of social class and other risk factors. Am J Epidemiol 153(2):114–122 Kamangar F, Qiao YL, Schiller JT et al, 2006, Human papillomavirus serology and the risk of esophageal and gastric cancers: results from a cohort in a high-risk region in China. Int J Cancer 119:579–584 Lu XM, Monnier-Benoit S, Mo LZ et al, 2008, Human papillomavirus in esophageal squamous cell carcinoma of the high-risk Kazakh ethnic group in Xinjiang, China. Eur J Surg Oncol 34:765– 770 Nasrollahzadeh D, Kamangar F, Aghcheli K, Sotoudeh M, Islami F, Abnet C et al, 2008, Opium, tobacco, and alcohol use in relation to oesophageal squamous cell carcinoma in a high-risk area of Iran. Br J Cancer 98(11):1857–1863 Akbari MR, Malekzadeh R, Nasrollahzadeh D, Amanian D, Sun P, Islami F et al, 2006, Familial risks of esophageal cancer among the Turkmen population of the Caspian Littoral of Iran. Int J Cancer 119(5):1047–1051 Kamangar F, Strickland PT, Pourshams A, Malekzadeh R, Boffetta P, Roth M et al, 2005, High exposure to polycyclic aromatic hydrocarbons may contribute to high risk of esophageal cancer in northeastern Iran. Anticancer Res 25(1B):425–428 Taghavi N, Nasrollahzadeh D, Merat S, Yazdanbod A, Hormazdi M, Sotoudeh M et al, 2007, Epidemiology of upper gastrointestinal cancers in Iran: a sub site analysis of 761 cases. World J Gastroenterol 13(40):5367–5370 Akbari MR, Malekzadeh R, Nasrollahzadeh D, Amanian D, Islami F, Li S et al, 2008, Germline BRCA2 mutations and the risk of esophageal squamous cell carcinoma. Oncogene 27(9):1290–1296 Islami F, Nasrollahzadeh D, Kamangar F, 2008, Socioeconomic status in relation to esophageal cancer in a high-risk area of Iran. Gastroenterology 134(4):301–301 Munoz N, Bosch FX, de Sanjose S et al, 2003, Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med 348(6):518–527 de Villiers EM, Fauquet C, Broker TR, Bernard HU, zur Hausen H, 2004, Classification of papillomaviruses. Virology 324(1):17–27 Bahnassy AA, Zekri AR, Abdallah S, El-Shehaby AM, Sherif GM, 2005, Human papillomavirus infection in Egyptian esophageal carcinoma: correlation with p53, p21, mdm2, C-erbB2 and impact on survival. Pathol Int 55:53–62 Castillo A, Aguayo F, Koriyama C et al, 2006, Human papillomavirus in esophageal squamous cell carcinoma in Colombia and Chile. World J Gastroenterol 12:6188–6192 Souto Damin AP, Guedes Frazzon AP, de Carvalho Damin D et al, 2006, Detection of human papillomavirus DNA in squamous cell carcinoma of the esophagus by auto-nested PCR. Dis Esophagus 19:64–68 Pantelis A, Pantelis D, Ruemmele P et al, 2007, p53 Codon 72 polymorphism, loss of heterozygosity and high-risk human papillomavirus infection in a low-incidence German esophageal squamous cell carcinoma patient cohort. Oncol Rep 17:1243–1248 Koh JS, Lee SS, Baek HJ, Kim YI, 2008, No association of highrisk human papilloma virus with esophageal squamous cell carcinomas among Koreans, as determined by polymerase chain reaction. Dis Esophagus 21:114–117 Eslami Far A, Aghakhani A, Hamkar R, Ramezani A, Froutan Pishbigar H et al, 2007, Frequency of human papillomavirus infection in esophageal squamous cell carcinoma in Iranian patients. Scand J Infect Dis 39:58–62 Shuyama K, Castillo A, Aguayo F et al, 2007, Human papillomavirus in high- and low-risk areas of oesophageal squamous cell carcinoma in China. Br J Cancer 96:1554–1559 Lambert R, Hainaut P, 2007, Esophageal cancer: cases and causes, part I). Endoscopy 39:550–555 Kamangar F, Malekzadeh R, Dawsey SM, Saidi F, 2007, Esophageal cancer in Northeastern Iran: a review. Arch Iran Med 10:70– 82 Islami F, Kamangar F, Aghcheli K, Fahimi S, Semnani S, Taghavi N et al, 2004, Epidemiologic features of upper gastrointestinal tract cancers in Northeastern Iran. Br J Cancer 90:1402–1406 Moradi AV, de Villers EM, Mokhtari-Azad T, Mahmoudi M, Hazrati B et al, 2002, Detection of human papillomavirus DNA by PR in esophageal squamous cell carcinoma from Turkmen Sahra, North-East of Iran. Iranian Biomed J 6(1):19–23 Wang X, Tian X, Liu F, Zhao Y, Sun M, Chen D et al, 2010, Detection of HPV DNA in esophageal cancer specimens from different regions and ethnic groups: a descriptive study. BMC Cancer 10:19 Zhang D, Zhang Q, Zhou L, Huo L, Zhang Y, Shen Z et al, 2010, Comparison of prevalence, viral load, physical status and expression of human papillomavirus- 16, −18 and −58 in esophageal and cervical cancer: a case–control study. BMC Cancer 10:650–658 Zhang QY, Zhang DH, Shen ZY, Xu LY, Li EM, Au WW, 2011, Infection and integration of human papillomavirus in esophageal carcinoma. Int J Hyg Environ Health 214:156–161 Matsha T, Erasmus R, Kafuko AB, Mugwanya D, Stepien A, Parker MI, 2002, Human papillomavirus associated with esophageal cancer. J Clin Pathol 55:587–590 Matsha T, Donninger H, Erasmus RT et al, 2007, Expression of p53 and its homolog, p73, in HPV DNA positive oesophageal squamous cell carcinomas. Virology 369:182–190 Farhadi M, Tahmasebi Z, Merat S, Kamangar F, Nasrollahzadeh D, Malekzadeh R, 2005, Human papillomavirus in squamous cell carcinoma of esophagus in a high-risk population. World J Gastroenterol 11(8):1200–1203 Dreilich M, Bergqvist M, Moberg M, Daniel Brattstrom D, Gustavsson I, Bergstrom S, 2006, High-risk human papilloma virus, HPV, and survival in patients with esophageal carcinoma: a pilot study. BMC Cancer 6:94 Herrera-Goepfert R, Lizano M, Akiba S, Carrillo-Garcia A, Becker-D’Acosta M, 2009, Human papilloma virus and esophageal carcinoma in a Latin-American region. World J Gastroenterol 15(25):3142–3147 de Villiers EM, Gunst K, Stein H, Scherubl H, 2004, Esophageal squamous cell cancer in patients with head and neck cancer: prevalence of human papillomavirus DNA sequences. Int J Cancer 109:253–258 D’Souza G, Kreimer AR, Viscidi R, Pawlita M, Fakhry C, Koch WM, Westra WH, Gillison ML, 2007, Case–control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 356:1944–1956 Rintala MA, Grenman SE, Puranen MH, Isolauri E, Ekblad U, Kero PO, Syrjanen SM, 2005, Transmission of high-risk human papillomavirus, HPV, between parents and infant: a prospective study of HPV in families in Finland. J Clin Microbiol 43:376–381 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 385 EP 391 DI 10.1007/s12253-012-9590-0 PG 7 ER PT J AU Je, ME Yoo, JN Lee, HS AF Je, Mi Eun Yoo, Jin Nam Lee, Hyung Sug TI Somatic Mutation of PARK2 Tumor Suppressor Gene is not Common in Common Solid Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PARK2; Tumor suppressor; Mutation; Cancers ID PARK2; Tumor suppressor; Mutation; Cancers AB Recent studies identified that PARK2 gene was a candidate tumor suppressor gene in colorectal cancers and glioblastomas. The aim of this study was identify whether PARK2 somatic mutation is present in other solid tumor as well. In this study, we analyzed the entire coding sequences of human PARK2 gene in gastric, colorectal, breast, lung and prostate carcinoma by single-strand conformation polymorphism (SSCP) and subsequent direct DNA sequencing. We found two missense mutations (p.Ser9Thr and p.Gly450Val) in colon carcinomas (4.3 %), which were not overlapped with the known PARK2 mutations. Our data suggest that somatic mutational events in PARK2 gene may be rare in colorectal, gastric, prostate, breast and lung carcinomas and may not play an important role in the development of these cancers. C1 [Je, Mi Eun] The Catholic University of Korea, College of Medicine, Cancer Research Institute, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Cancer Research Institute, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Cancer Research Institute, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Cancer Research Institute, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Yoshii SR, Kishi C, Ishihara N, Mizushima N, 2011, Parkin mediates proteasome-dependent protein degradation and rupture of the outer mitochondrial membrane. J Biol Chem 286:19630– 19640 Kahle PJ, Haass C, 2004, How does parkin ligate ubiquitin to Parkinson’s disease? EMBO Rep 5:681–685 Farrer MJ, 2006, Genetics of Parkinson disease: paradigm shifts and future prospects. Nat Rev Genet 7:306–318 Abbas N, Lucking CB, Ricard S, Durr A, Bonifati V, De Michele G, Bouley S, Vaughan JR, Gasser T, Marconi R, Broussolle E, Brefel-Courbon C, Harhangi BS, Oostra BA, Fabrizio E, Bohme GA, Pradier L, Wood NW, Filla A, Meco G, Denefle P, Agid Y, Brice A, 1999, A wide variety of mutations in the parkin gene are responsible for autosomal recessive parkinsonism in Europe. French Parkinson’s Disease Genetics Study Group and the European Consortium on Genetic Susceptibility in Parkinson’s Disease. Hum Mol Genet 8:567–574 Cesari R, Martin ES, Calin GA, Pentimalli F, Bichi R, McAdams H, Trapasso F, Drusco A, Shimizu M, Masciullo V, D’Andrilli G, Scambia G, Picchio MC, Alder H, Godwin AK, Croce CM, 2003, Parkin, a gene implicated in autosomal recessive juvenile parkinsonism, is a candidate tumor suppressor gene on chromosome 6q25-q27. Proc Natl Acad Sci U S A 100:5956–5961 Picchio MC, Martin ES, Cesari R, Calin GA, Yendamuri S, Kuroki T, Pentimalli F, Sarti M, Yoder K, Kaiser LR, Fishel R, Croce CM, 2004, Alterations of the tumor suppressor gene Parkin in nonsmall cell lung cancer. Clin Cancer Res 10:2720–2724 Fujiwara M, Marusawa H, Wang HQ, Iwai A, Ikeuchi K, Imai Y, Kataoka A, Nukina N, Takahashi R, Chiba T, 2008, Parkin as a tumor suppressor gene for hepatocellular carcinoma. Oncogene 27:6002–6011 Poulogiannis G, McIntyre RE, Dimitriadi M, Apps JR,Wilson CH, Ichimura K, Luo F, Cantley LC, Wyllie AH, Adams DJ, Arends MJ, 2010, PARK2 deletions occur frequently in sporadic colorectal cancer and accelerate adenoma development in Apc mutant mice. Proc Natl Acad Sci U S A 107:15145–15150 Veeriah S, Taylor BS, Meng S, Fang F, Yilmaz E, Vivanco I, Janakiraman M, Schultz N, Hanrahan AJ, Pao W, Ladanyi M, Sander C, Heguy A, Holland EC, Paty PB, Mischel PS, Liau L, Cloughesy TF, Mellinghoff IK, Solit DB, Chan TA, 2010, Somatic mutations of the Parkinson’s disease-associated gene PARK2 in glioblastoma and other human malignancies. Nat Genet 42:77–82 Lee JW, Soung YH, Kim SY, Lee HW, Park WS, Nam SW, Kim SH, Lee JY, Yoo NJ, Lee SH, 2005, PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas. Oncogene 24:1477–1480 Kim MS, Hur SY, Yoo NJ, Lee SH, 2010, Mutational analysis of FOXL2 codon 134 in granulosa cell tumour of ovary and other human cancers. J Pathol 221:147–152 Kim MS, Oh JE, Kim YR, Park SW, Kang MR, Kim SS, Ahn CH, Yoo NJ, Lee SH, 2010, Somatic mutations and losses of expression of microRNA regulation-related genes AGO2 and TNRC6A in gastric and colorectal cancers. J Pathol 221:139–146 Kim YR, Oh JE, Kim MS, Kang MR, Park SW, Han JY, Eom HS, Yoo NJ, Lee SH, 2010, Oncogenic NRF2 mutations in squamous cell carcinomas of oesophagus and skin. J Pathol 220:446–451 Hayashi K, 1992, PCR-SSCP: a simple and sensitive method for detection of mutations in the genomic DNA. PCR Methods Appl 1:34–38 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 393 EP 395 DI 10.1007/s12253-012-9591-z PG 3 ER PT J AU Du, X Zhao, H Zang, L Song, N Yang, T Dong, R Yin, J Wang, Ch Lu, J AF Du, Xilin Zhao, Huadong Zang, Li Song, Nuan Yang, Tao Dong, Rui Yin, Jikai Wang, Chengguo Lu, Jianguo TI Overexpression of Histone Deacetylase 2 Predicts Unfavorable Prognosis in Human Gallbladder Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Primary gallbladder carcinoma; Histone deacetylase 2; Clinicopathology; Overall survival; Disease-free survival ID Primary gallbladder carcinoma; Histone deacetylase 2; Clinicopathology; Overall survival; Disease-free survival AB As important regulators of chromatin, histone deacetylases (HDACs) are involved in silencing tumor suppressor genes. HDAC2, a member of HDACs, has been demonstrated to be implicated in the development and progression of various human malignancies; however, its expression and role in human primary gallbladder carcinoma (PGC) are not fully understood. Therefore, we conducted this study to address this problem. The subjects were 136 patients underwent resection for PGC. Immunostainings for HDAC2 were performed on these archival tissues. The correlation of HDAC2 expression with clinicopathological characteristics including survival was analyzed. HDAC2 was positively expressed in the nucleus of tumor cells in 86.0 % (117/136) of PGC but not in the normal epithelium of the gallbladder. Additionally, there was a significant difference in the incidence of positive nodal metastasis between high and low HDAC2 expression groups (P00.001). The incidences of advanced clinical stage (P00.005) and pathologic T stage (P<0.001), and higher histologic grade (P<0.001) were respectively higher in the high HDAC2 expression group than in the low group. Moreover, univariate and multivariate analyses revealed the high HDAC2 expression to be an independent prognostic factor for both overall and disease-free survival of patients with PGC. High HDAC2 expression was correlated with a high incidence of lymph node metastasis and aggressive tumor progression of PGC. It also was an independent prognostic factor for poorer overall and disease-free survival in patients. Therefore, detection of HDAC2 expression may help us screen patients at increased risk of malignant behavior for PGC. C1 [Du, Xilin] Fourth Military Medical University, Tangdu Hospital, Department of general surgery, 710038 Xi’an, China. [Zhao, Huadong] Fourth Military Medical University, Tangdu Hospital, Department of general surgery, 710038 Xi’an, China. [Zang, Li] Fourth Military Medical University, Tangdu Hospital, Department of general surgery, 710038 Xi’an, China. [Song, Nuan] Fourth Military Medical University, Tangdu Hospital, Department of general surgery, 710038 Xi’an, China. [Yang, Tao] Fourth Military Medical University, Tangdu Hospital, Department of general surgery, 710038 Xi’an, China. [Dong, Rui] Fourth Military Medical University, Tangdu Hospital, Department of general surgery, 710038 Xi’an, China. [Yin, Jikai] Fourth Military Medical University, Tangdu Hospital, Department of general surgery, 710038 Xi’an, China. [Wang, Chengguo] Fourth Military Medical University, Tangdu Hospital, Department of general surgery, 710038 Xi’an, China. [Lu, Jianguo] Fourth Military Medical University, Tangdu Hospital, Department of general surgery, 710038 Xi’an, China. RP Wang, Ch (reprint author), Fourth Military Medical University, Tangdu Hospital, Department of general surgery, 710038 Xi’an, China. EM chengguo@fmmu.edu.cn CR Misra S, Chaturvedi A, Misra NC, Sharma ID, 2003, Carcinoma of the gallbladder. Lancet Oncol 4:167–176 Chen Y, Chen Y, Yu G, Ding H, 2011, Lymphangiogenic and angiogentic microvessel density in gallbladder carcinoma. Hepatogastroenterology 58:20–25 Araida T, Higuchi R, Hamano M, Kodera Y, Takeshita N, Ota T, Yoshikawa T, Yamamoto M, Takasaki K, 2009, Should the extrahepatic bile duct be resected or preserved in R0 radical surgery for advanced gallbladder carcinoma? results of a Japanese Society of Biliary Surgery Survey: a multicenter study. Surg Today 39:770– 779 Kokudo N, Makuuchi M, Natori T, Sakamoto Y, Yamamoto J, Seki M, Noie T, Sugawara Y, Imamura H, Asahara S, Ikari T, 2003, Strategies for surgical treatment of gallbladder carcinoma based on information available before resection. Arch Surg 138:741–750 Davies GF, Ross AR, Arnason TG, Juurlink BH, Harkness TA, 2010, Troglitazone inhibits histone deacetylase activity in breast cancer cells. Cancer Lett 288:236–250 Brandl A, Heinzel T, Kramer OH, 2009, Histone deacetylases: salesmen and customers in the post-translational modification market. Biol Cell Auspices Eur Cell Biol Organ 101:193–205 Ishdorj G, Graham BA, Hu X, Chen J, Johnston JB, Fang X, Gibson SB, 2008, Lysophosphatidic acid protects cancer cells from histone deacetylase, HDAC, inhibitor-induced apoptosis through activation of HDAC. J Biol Chem 283:16818–16829 Mehdi O, Francoise S, Sofia CL, Urs G, Kevin Z, Bernard S, Igor S, Anabela CD, Dominique L, Eric M, Ali O, 2012, HDAC gene expression in pancreatic tumor cell lines following treatment with the HDAC inhibitors panobinostat, LBH589, and trichostatine, TSA). Pancreatology 12:146–155 Spiegel S, Milstien S, Grant S, 2012, Endogenous modulators and pharmacological inhibitors of histone deacetylases in cancer therapy. Oncogene 31:537–551 Lee JH, Jeong EG, Choi MC, Kim SH, Park JH, Song SH, Park J, Bang YJ, Kim TY, 2010, Inhibition of histone deacetylase 10 induces thioredoxin-interacting protein and causes accumulation of reactive oxygen species in SNU-620 human gastric cancer cells. Mol Cells 30:107–112 Xu LN, Wang X, Zou SQ, 2008, Effect of histone deacetylase inhibitor on proliferation of biliary tract cancer cell lines. World J Gastroenterol 14:2578–2581 Yamaguchi J, Sasaki M, Sato Y, Itatsu K, Harada K, Zen Y, Ikeda H, Nimura Y, Nagino M, Nakanuma Y, 2010, Histone deacetylase inhibitor, SAHA, and repression of EZH2 synergistically inhibit proliferation of gallbladder carcinoma. Cancer Sci 101:355–362 Kitamura T, Connolly K, Ruffino L, Ajiki T, Lueckgen A, Digiovanni J, Kiguchi K., 2012, The therapeutic effect of histone deacetylase inhibitor PCI-24781 on gallbladder carcinoma in BK5.erbB2 mice. J Hepatol. In press Willis-Martinez D, Richards HW, Timchenko NA, Medrano EE, 2010, Role of HDAC1 in senescence, aging, and cancer. Exp Gerontol 45:279–285 Schuler S, Fritsche P, Diersch S, Arlt A, Schmid RM, Saur D, Schneider G, 2010, HDAC2 attenuates TRAIL-induced apoptosis of pancreatic cancer cells. Mol Cancer 9:80 Jung KH, Noh JH, Kim JK, Eun JW, Bae HJ, Xie HJ, Chang YG, Kim MG, Park H, Lee JY, Nam SW, 2012, HDAC2 overexpression confers oncogenic potential to human lung cancer cells by deregulating expression of apoptosis and cell cycle proteins. J Cell Biochem 113:2167–2177 Patani N, Jiang WG, Newbold RF, Mokbel K, 2011, Histonemodifier gene expression profiles are associated with pathological and clinical outcomes in human breast cancer. Anticancer Res 31:4115–4125 Quint K, Agaimy A, Di Fazio P, Montalbano R, Steindorf C, Jung R, Hellerbrand C, Hartmann A, Sitter H, Neureiter D, Ocker M, 2011, Clinical significance of histone deacetylases 1, 2, 3, and 7: HDAC2 is an independent predictor of survival in HCC. Virchows Arch 459:129–139 Aghdassi A, Sendler M, Guenther A, Mayerle J, Behn CO, Heidecke CD, Friess H, Buchler M, Evert M, Lerch MM, Weiss FU, 2012, Recruitment of histone deacetylases HDAC1 and HDAC2 by the transcriptional repressor ZEB1 downregulates E-cadherin expression in pancreatic cancer. Gut 61:439– 448 Ramsey MR, He L, Forster N, Ory B, Ellisen LW, 2011, Physical association of HDAC1 and HDAC2 with p63 mediates transcriptional repression and tumor maintenance in squamous cell carcinoma. Cancer Res 71:4373–4379 de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB, 2003, Histone deacetylases, HDACs): characterization of the classical HDAC family. Biochem J 370:737–749 Khier H, Bartl S, Schuettengruber B, Seiser C, 1999, Molecular cloning and characterization of themouse histone deacetylase 1 gene: integration of a retrovirus in 129SV mice. Biochim Biophys Acta 1489:365–373 Brunmeir R, Lagger S, Seiser C, 2009, Histone deacetylase HDAC1/HDAC2-controlled embryonic development and cell differentiation. Int J Dev Biol 53:275–289 Yamaguchi T, Cubizolles F, Zhang Y, Reichert N, Kohler H, Seiser C, Matthias P, 2010, Histone deacetylases 1 and 2 act in concert to promote the G1-to-S progression. Genes Dev 24:455–469 Jurkin J, Zupkovitz G, Lagger S, Grausenburger R, Hagelkruys A, Kenner L, Seiser C, 2011, Distinct and redundant functions of histone deacetylases HDAC1 and HDAC2 in proliferation and tumorigenesis. Cell Cycle 10:406–412 Huang BH, Laban M, Leung CH, Lee L, Lee CK, Salto-Tellez M, Raju GC, Hooi SC, 2005, Inhibition of histone deacetylase 2 increases apoptosis and p21Cip1/WAF1 expression, independent of histone deacetylase 1. Cell Death Differ 12:395–404 Harms KL, Chen X, 2007, Histone deacetylase 2 modulates p53 transcriptional activities through regulation of p53-DNA binding activity. Cancer Res 67:3145–3152 Zhu P, Martin E, Mengwasser J, Schlag P, Janssen KP, Gottlicher M, 2004, Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis. Cancer Cell 5:455–463 Chang HH, Chiang CP, Hung HC, Lin CY, Deng YT, Kuo MY, 2009, Histone deacetylase 2 expression predicts poorer prognosis in oral cancer patients. Oral Oncol 45:610–614 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 397 EP 403 DI 10.1007/s12253-012-9592-y PG 7 ER PT J AU Wang, K Jia, Z Zou, J Zhang, A Wang, G Hao, J Wang, Y Yang, Sh Pu, P AF Wang, Kun Jia, Zhifan Zou, Jian Zhang, Anling Wang, Guangxiu Hao, Jianwei Wang, Yirong Yang, Shuxu Pu, Peiyu TI Analysis of hsa-miR-30a-5p Expression in Human Gliomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; miR-30a-5p; Ex ID Glioma; miR-30a-5p; Ex AB Our previous study demonstrated that miR-30a-5p was upregulated in six malignant glioma cell lines by micro- RNA(miRNA) array. For further verification of this finding, the expression of miR-30a-5p in 7 more malignant glioma cell lines, 43 freshly resected glioma samples and 75 archival paraffin embedded glioma specimens with different grade of malignancy were examined by qRT-PCR and in situ hybridization (ISH). Here, we present the first evidence that miR-30a-5p is overexpressed in glioma cell lines and glioma samples as compared to the normal brain tissues (NBTs), and its expression level is positively correlated with tumor grade of malignancy. It is concluded that miR-30a-5p may have the potential as a diagnostic or prognostic marker of gliomas and as the target of miRNA-based glioma therapy in further studies. C1 [Wang, Kun] Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. [Jia, Zhifan] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, Heping District, 300052 Tianjin, China. [Zou, Jian] School of Medicine, Zhejiang University, Women’s Hospital, Women’s Reproductive Health Laboratory of Zhejiang Province, 310006 Hangzhou, China. [Zhang, Anling] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, Heping District, 300052 Tianjin, China. [Wang, Guangxiu] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, Heping District, 300052 Tianjin, China. [Hao, Jianwei] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, Heping District, 300052 Tianjin, China. [Wang, Yirong] Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. [Yang, Shuxu] Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. [Pu, Peiyu] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, Heping District, 300052 Tianjin, China. RP Pu, P (reprint author), Tianjin Medical University General Hospital, Department of Neurosurgery, 300052 Tianjin, China. EM pupeiyu33@hotmail.com CR Eyler CE, Foo WC, LaFiura KM et al, 2008, Brain cancer stem cells display preferential sensitivity to Akt inhibition. Stem Cells 26:3027–3036 Geiger GA, Fu W, Kao GD, 2008, Temozolomide-mediated radiosensitization of human glioma cells in a zebrafish embryonic system. Cancer Res 68:3396–3404 Zheng Y, Lin L, Zheng Z, 2008, TGF-alpha induces upregulation and nuclear translocation of Hes1 in glioma cell. Cell Biochem Funct 26:692–700 Lee RC, Ambros V, 2001, An extensive class of small RNAs in Caenorhabditis elegans. Science 294:862–864 Lau NC, Lim LP, Weinstein EG et al, 2001, An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science 294:858–862 Lagos-Quintana M, Rauhut R, Lendeckel W et al, 2001, Identification of novel genes coding for small expressed RNAs. Science 294:853–858 Bartel DP, 2009, MicroRNAs: target recognition and regulatory functions. Cell 136:215–233 Caldas C, Brenton JD, 2005, Sizing up miRNAs as cancer genes. Nat Med 11:712–714 Calin GA, Sevignani C, Dumitru CD et al, 2004, Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A 101:2999–3004 Calin GA, Croce CM, 2006, Genomics of chronic lymphocytic leukemia microRNAs as new players with clinical significance. Semin Oncol 33:167–173 Hayashita Y, Osada H, Tatematsu Y et al, 2005, A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res 65:9628– 9632 Ciafre SA, Galardi S, Mangiola A et al, 2005, Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun 334:1351–1358 He H, Jazdzewski K, Li W et al, 2005, The role of microRNA genes in papillary thyroid carcinoma. Proc Natl Acad Sci U S A 102:19075–19080 Iorio MV, Ferracin M, Liu CG et al, 2005, MicroRNA gene expression deregulation in human breast cancer. Cancer Res 65:7065–7070 Metzler M, Wilda M, Busch K et al, 2004, High expression of precursor microRNA-155/BIC RNA in children with Burkitt lymphoma. Genes Chromosom Cancer 39:167–169 Michael MZ, O' Connor SM, van Holst Pellekaan NG et al, 2003, Reduced accumulation of specific microRNAs in colorectal neoplasia. Mol Cancer Res 1:882–891 Murakami Y, Yasuda T, Saigo K et al, 2006, Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene 25:2537–2545 Takamizawa J, Konishi H, Yanagisawa K et al, 2004, Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res 64:3753–3756 You Y, Pu P, Huang Q et al, 2006, Antisense telomerase RNA inhibits the growth of human glioma cells in vitro and in vivo. Int J Oncol 28:1225–1232 Livak KJ, Schmittgen TD, 2001, Schmittgen: analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T), method. Methods 25:402– 408 Chan JA, Krichevsky AM, Kosik KS, 2005, MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res 65:6029–6033 Dong H, Siu H, Luo L et al, 2010, Investigation gene and micro- RNA expressionin in glioblastoma. BMC Genomics 11(3):S16 Hummel R, Maurer J, Haier J, 2011, MicroRNAs in brain tumors: a new diagnostic and therapeutic perspective? Mol Neurobiol 44:223–234 Li J, Donath S, Li Y et al, 2010, miR-30 regulates mitochondrial fission through targeting p53 and the dynamin-related protein-1 pathway. PLoS Genet 6:e1000795 Martinez I, Cazalla D, Almstead LL et al, 2011, miR-29 and miR- 30 regulate B-Myb expression during cellular senescence. Proc Natl Acad Sci U S A 108:522–527 Yu F, Deng H, Yao H et al, 2010, Mir-30 reduction maintains selfrenewal and inhibits apoptosis in breast tumor-initiating cells. Oncogene 29:4194–4204 Gaziel-Sovran A, Segura MF, Di Micco R et al, 2011, miR-30b/ 30d regulation of GalNAc transferases enhances invasion and immunosuppression during metastasis. Cancer Cell 20:104–118 Calin GA, Liu CG, Sevignani C et al, 2004, MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proc Natl Acad Sci U S A 101:11755–11760 Volinia S, Calin GA, Liu CG et al, 2006, A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 103:2257–2261 Schetter AJ, Leung SY, Sohn JJ et al, 2008, MicroRNA expression profiles associated with prognosis and therapeutic outcome in colon adenocarcinoma. Jama 299:425–436 Garzon R, Volinia S, Liu CG et al, 2008, MicroRNA signatures associated with cytogenetics and prognosis in acute myeloid leukemia. Blood 111:3183–3189 Zou ZY, Wu LP, Ding HY et al, 2012, MicroRNA-30a sensitizes tumor cells to cis-platinum via suppressing beclin 1-mediated autophagy*. J Biol Chem 287:4148–4156 Baraniskin A, Birkenkamp-Demtroder K, Maghnouj A et al, 2012, MiR-30a-5p suppresses tumor growth in colon carcinoma by targeting DTL. Carcinog 33(4):732–739 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 405 EP 411 DI 10.1007/s12253-012-9593-x PG 7 ER PT J AU Abdel-Aziz, A Mohamed, AM Akl, MFF Taha, NMA AF Abdel-Aziz, Azza Mohamed, Ali Mie Akl, Mohamed Farouk Fatma Taha, Nageeb M Ahmed TI Survivin Expression in Medulloblastoma: A Possible Marker for Survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Medulloblastoma; survivin; survival; antiapoptosis ID Medulloblastoma; survivin; survival; antiapoptosis AB Medulloblastomas are highly invasive tumors which are generally disseminated at the time of diagnosis. High and continued morbidity and mortality have prompted the search for new biologic markers that might be used for targeted therapy to minimise treatment related side effects. In this work, we studied the positive expression of survivin in medulloblastoma and investigated its relation to clinical, pathologic data and survival. Tumor tissue specimens from 47 patients with medulloblastoma who underwent primary surgical treatment from June 2002 to June 2006 at the Mansoura university hospital, Egypt were collected. Paraffin sections of all samples were submitted for immunohistochemistry using anti-survivin antibody. The relation between the percentage of positive survivin cells with clinical, pathological and survival data was evaluated Results: In 47 cancer tissue specimens, one case large-cell-anaplastic (1.12 %), tweleve cases desmoplastic (25.53 %) and 34 cases classic medulloblastomas (72.34 %). The immunohistochemical expression of survivin was nulear with moderate intensity. It does not correlate with either age or sex. There was a significant negative correlation of survivin expression with survival (p<0.001), where negative survivin immunostaining was associated with prolonged overall and disease free survival, while survivin expression was associated with shortened survival. Conclusion: Survivin expression correlate with the clinical outcome with poor prognosis and could be a potential predictive factor for recurrence or metastasis. C1 [Abdel-Aziz, Azza] Mansoura University, Faculty of Medicine, Department of PathologyMansoura, Egypt. [Mohamed, Ali Mie] Mansoura University, Faculty of Medicine, Department of PathologyMansoura, Egypt. [Akl, Mohamed Farouk Fatma] Mansoura University, Faculty of Medicine, Clinical Oncology and Nuclear Medicine DepartmentMansoura, Egypt. [Taha, Nageeb M Ahmed] Mansoura University, Faculty of Medicine, Neurosurgery departmentMansoura, Egypt. RP Akl, MFF (reprint author), Mansoura University, Faculty of Medicine, Clinical Oncology and Nuclear Medicine Department, Mansoura, Egypt. EM fatmaakl@yahoo.com CR Bowers DC, Gargan L, Weprin BE, Muline AF, Elterman RD, Munoz L, Giller CA, Winick NJ, 2007, Impact of site of tumor recurrence upon survival for children with recurrent or progressive medulloblastoma. J Neurosurg 107(1 Suppl Pediatrics): 5–10 Rossi A, Caracciolo V, Russo G, Reiss K, Giordano A, 2008, Medulloblastoma: from molecular pathology to therapy review. Clin Cancer Res 14(4):971–976 Sasaki T, Lopes MBS, Hankins GR, Helm GA, 2002, Expression of survivin, an inhibitor of apoptosis protein, in tumors of the nervous system. Acta Neuropathol 104:105–109 Li XN, Shu Q, Su JM, Adesina AM, Wong KK, Perlaky L, Antalffy BA, Blaney SM, Lau CC, 2007, Differential expression of survivin splice isoforms in medulloblastomas. Neuropathol Appl Neurobiol 33:67–76 Liu R, Mitchell DA, 2010, Survivin as an immunotherapeutic target for adult and pediatric malignant brain tumors. Cancer Immunol Immunother 59:183–193 Fangusaro JR, Jiang Y, Holloway MP, Caldas H, Singh V, Boue DR, Hayes J, Altura RA, 2005, Survivin, Survivin-2B, and Survivin-deltaEx3 expression in medulloblastoma: biologic markers of tumour morphology and clinical outcome. British J Cancer 92:359–365 Chang CH, Housepian EM, Herbert C Jr, 1969, An operative staging system and a megavoltage radiotherapeutic technic for cerebellar medulloblastomas. Radiology 93:1351–1359 Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Eds.,, 2007, WHO Classification of tumours of the central nervous system. IARC: Lyon 8:132–140 Packer RJ, Vezina G, 2008, Management of and Prognosis with Medulloblastoma. Ther Crossroads Arch Neurol 65(11):1419– 1424 Packer RJ, Cogen P, Vezina G, Rorke LB, 1999, Medulloblastoma: clinical and biologic aspects. Neuro-Oncology 1:232–250 Gilbertson RJ, 2004, Medulloblastoma: signalling a change in treatment. Lancet Oncol 5:209–18 Roberts RO, Lynch CF, Jones MP, Hart MN, 1991, Medulloblastoma: a population-based study of 532 cases. J Neuropathol Exp Neurol 50:134–144 Giordana MT, Schiffer P, Lanotte M, Girardi P, Chio A, 1999, Epidemiology of adult medulloblastoma. Int J Cancer 80:689– 692 Koeller KK, Rushing EJ, 2003, From the archives of the AFIP Medulloblastoma: a comprehensive review with radiologicpathologic correlation. AFIP ARCHIVES 23:1613–1637 Li F, Ambrosini G, Chu EY, Plescia J, Tognin S, Marchisio PC, Altieri DC, 1998, Control of apoptosis and mitotic spindle checkpoint by survivin. Nature 396:580–584 Ambrosini G, Adida C, Altieri DC, 1997, A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 3, 8):917–921 Deveraux QL, Reed JC, 1999, IAP family proteins—suppressors of apoptosis. Genes Dev 13:239–252 Pizem J, Cort A, Zadravec-Zaletel L, Popovic M, 2005, Survivin is a negative prognostic marker in medulloblastoma. Neuropathol Appl Neurobiol 31(4):422–8 Faccion RS, Ferreira RM, Grabois MF, Fonseca TC, de Oliveira JA, Maia RC, 2011, Lack of prognostic significance of survivin in pediatric medulloblastoma. Pathol Oncol Res 17(4):899– 908 Haberler C, Slavc I, Czech T, Gelpi E, Heinzl H, Budka H, Urban C, Scarpatetti M, Ebetsberger-Dachs G, Schindler C, Jones N, Klein-Franke A, Maier H, Jauk B, Kiefer A, Hainfellner JA, 2006, Histopathological prognostic factors in medulloblastoma: high expression of survivin is related to unfavourable outcome. Eur J Cancer 42(17):2996–3003 Duo-binW, Yong-ming J, Ming-xue NU, et al., 2009–01, Survivin Expression in Medulloblastoma and its Clinical Significance West China Medical Journal Bodey B, Bodey V, Siegel SE, Kaiser HE, 2004, Survivin expression in childhood medulloblastomas: a possible diagnostic and prognostic marker. In vivo 18:713–718 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 413 EP 419 DI 10.1007/s12253-012-9594-9 PG 7 ER PT J AU Smardova, J Liskova, K Ravcukova, B Kubiczkova, L Sevcikova, S Michalek, J Svitakova, M Vybihal, V Kren, L Smarda, J AF Smardova, Jana Liskova, Kvetoslava Ravcukova, Barbora Kubiczkova, Lenka Sevcikova, Sabina Michalek, Jaroslav Svitakova, Miluse Vybihal, Vaclav Kren, Leos Smarda, Jan TI High Frequency of Temperature-Sensitive Mutants of p53 in Glioblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma; p53 tumor suppressor; FASAY; Temperature-sensitive mutations; p53 gene deletion ID Glioblastoma; p53 tumor suppressor; FASAY; Temperature-sensitive mutations; p53 gene deletion AB Glioblastoma is the most common and the most aggressive type of brain cancer. Aberrations of the RTK/RAS/PI3K-, p53-, and RB cell signaling pathways were recognized as a core requirement for pathogenesis of glioblastoma. The p53 tumor suppressor functions as a transcription factor transactivating expression of its target genes in response to various stress stimuli. We determined the p53 status in 36 samples of glioblastoma by functional analyses FASAY and split assay. Seventeen p53 mutations were detected and further analyzed by cDNA and gDNA sequencing in 17 patients (47.2 %). Fifteen (88.2 %) of the mutations were missense mutations causing amino acid substitutions, seven of them exhibited temperature-sensitivity. Two mutations were determined as short deletions, one of them causing formation of premature termination codon in position 247. Fluorescent in situ hybridization revealed the loss of the p53- specific 17p13.3 locus in four of 33 analyzed samples (12 %). In 12 out of 30 samples (40 %), the p53 protein accumulation was shown by immunoblotting. There was high (80 %) concordance between the presence of the clonal p53 mutation and the p53 protein accumulation. C1 [Smardova, Jana] University Hospital Brno, Department of Pathology, Jihlavska 20, 625 00 Brno, Czech Republic. [Liskova, Kvetoslava] University Hospital Brno, Department of Pathology, Jihlavska 20, 625 00 Brno, Czech Republic. [Ravcukova, Barbora] Centre for Cardiovascular Surgery and Transplantation, Molecular Genetics LaboratoryBrno, Czech Republic. [Kubiczkova, Lenka] Masaryk University, Faculty of Medicine, Department of Pathological PhysiologyBrno, Czech Republic. [Sevcikova, Sabina] Masaryk University, Faculty of Medicine, Department of Pathological PhysiologyBrno, Czech Republic. [Michalek, Jaroslav] Masaryk University, Faculty of Medicine, Department of PharmacologyBrno, Czech Republic. [Svitakova, Miluse] University Hospital Brno, Department of Pathology, Jihlavska 20, 625 00 Brno, Czech Republic. [Vybihal, Vaclav] Faculty Hospital Brno, Department of NeurosurgeryBrno, Czech Republic. [Kren, Leos] University Hospital Brno, Department of Pathology, Jihlavska 20, 625 00 Brno, Czech Republic. [Smarda, Jan] Masaryk University, Faculty of Science, Department of Experimental BiologyBrno, Czech Republic. RP Smardova, J (reprint author), University Hospital Brno, Department of Pathology, 625 00 Brno, Czech Republic. EM janasmarda@seznam.cz CR Kleihues P, Louis DN, Scheithauer BW, Rorke LB, Reifenberger G, Burger PC, Cavenee WK, 2002, The WHO classification of tumors of the nervous system. J Neuropathol Exp Neurol 61:215– 225 Parsons DW, Jones S, Zhang X et al, 2008, An integrated genomic analysis of human glioblastoma multiforme. Science 321:1807–1812 Cancer Genome Atlas Research Network, 2008, Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455:1061–1068 Harris SL, Levine AJ, 2005, The p53 pathway: positive and negative feedback loops. Oncogene 24:2899–2908 Sherr CJ, 2004, Principles of tumor suppression. Cell 116:235– 246 Haupt Y, Maya R, Kazaz A, Oren M, 1997, Mdm2 promotes the rapid degradation of p53. Nature 387:296–299 Honda R, Tanaka H, Yasuda H, 1997, Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett 420:25– 27 Wu XW, Bayle JH, Olson D, Levine AJ, 1993, The p53 mdm-2 autoregulatory feedback loop. Genes Dev 7:1126–1132 Watanabe K, Sato K, Biernat W, Tachibana O, von Ammon K, Ogata N, Yonekawa Y, Kleihues P, Ohgaki H, 1997, Incidence and timing of p53 mutations during astrocytoma pregression in patients with multiple biopsies. Clin Cancer Res 3:523–530 Kraus JA, Glesmann N, Beck M, Krex D, Klockgether T, Schackert G, Schlegel U, 2000, Molecular analysis of the PTEN, TP53 and CDKN2A tumor suppressor genes in long-term survivors of glioblastoma multiforme. J Neuorooncol 48:89–94 Schiebe M, Ohneseit P, Hoffmann W, Meyermann R, Rodemann HP, Bamberg M, 2000, Analysis of mdm2 and p53 gene alterations in glioblastoma and its correlation with clinical factors. J Neurooncol 49:197–203 Schmidt MC, Antweiler S, Urban N, Mueller W, Kuklik A, Meyer- Puttlitz B, Wiestler OD, Louis DN, Fimmers R, von Deimling A, 2002, Impact of genotype and morphology on the prognosis of glioblastoma. J Neuropathol Exp Neurol 61:321–328 Peraud A, Kreth FW, Wiestler OD, Kleihues P, Reulen HJ, 2002, Prognostic impact of TP53 mutations and p53 protein overexpression in supratentorial WHO grade II astrocytomas and oligoastrocytomas. Clin Cancer Res 8:1117–1124 Batchelor TT, Betensky RA, Esposito JM, Pham LDD, Dorfman MV, Piscatelli N, Jhung S, Rhee D, Louis DN, 2004, Agedependent prognostic effects of genetic alterations in glioblastoma. Clin Cancer Res 10:228–233 Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL, Burkhard C, Schuler D, Probst-Hensch M, Maiorka PC, Baeza N, Pisani P, Yonekawa Y, Yasargil MG, Lutolf UM, Kleihues P, 2004, Genetic pathways to glioblastoma: a population-based study. Cancer Res 64:6892–6899 Rich JN, Hans C, Jones B, Iversen ES, McLendon RE, Rasheed BKA, Dobra A, Dressman HK, Bigner DD, Nevins JR, West M, 2005, Gene expression profiling and genetic markers in glioblastoma survival. Cancer Res 65:4051–4058 Momota H, Narita Y, Matsushita Y, Miyakita Y, Shibui S, 2010, p53 abnormality and tumor invasion in patients with malignant astrocytoma. Brain Tumor Pathol 27:95–101 Birner P, Piribauer M, Fischer I, Gatterbauer B, Marosi C, Ungersbock K, Rossler K, Budka H, Hainfellner JA, 2002, Prognostic relevance of p53 protein expression in glioblastoma. Oncol Rep 9:703–707 Srividya MR, Thota B, Arivazhagan A, Thennarasu K, Balasubramaniam A, Chandramouli BA, Hegde AS, Santosh V, 2010, Age-dependent prognostic effects of EGFR/p53 alterations in glioblastoma: study on a prospective cohort of 140 uniformly treated adult patients. J Clin Pathol 63:687–691 Flaman JM, Frebourg T, Moreau V, Charbonnier F, Martin C, Chappuis P, Sappino AP, Limacher JM, Bron L, Benhattar J, Tada M, Van Meir EG, Estreicher A, Iggo RD, 1995, A simple p53 functional assay for screening cell lines, blood, and tumors. Proc Natl Acad Sci USA 92:3963–3967 Smardova J, Nemajerova A, Trbusek M, Vagunda V, Kovarik J, 2001, Rare somatic p53 mutation identified in breast cancer: a case report. Tumor Biol 22:59–66 Ishioka C, Frebourg T, Yan YX, Vidal M, Friend SH, Schmidt S, Iggo R, 1993, Screening patients for heterozygous p53 mutations using a functional assay in yeast. Nat Genet 5:124–129 Waridel F, Estreicher A, Bron L, Flaman JM, Fontolliet C, Monnier P, Frebourg T, Iggo R, 1997, Field cancerisation and polyclonal p53 mutation in the upper aerodigestive tract. Oncogene 14:163–169 Petitjean A, Mathe E, Kato S, Ishioka C, Tavtigian SV, Hainaut P, Olivier M, 2007, Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mut 28:6292– 6299 Grochova D, Vankova J, Damborsky J, Ravcukova B, Smarda J, Vojtesek B, Smardova J, 2008, Analysis of transactivation capability and conformation of p53 temperature-dependent mutants and their reactivation by amifostine in yeast. Oncogene 27:1243–1252 Van Meir EG, Kikuchi T, Tada M, Li H, Diserens AC, Wojcik BE, Huang HJS, Friedmann T, de Tribolet N, Cavenee WK, 1994, Analysis of the p53 gene and its expression in human glioblastoma cells. Cancer Res 54:649–652 Tada M, Iggo RD, Waridel F, Nozaki M, Matsumoto R, Sawamura Y, Shinohe Y, Ikeda J, Abe H, 1997, Reappraisal of p53 mutations in human malignant astrocytic neoplasms by p53 functional assay: comparison with conventional structural analyses. Mol Carcinogen 18:171–176 Tada M, Matsumoto R, Iggo RD, Onimaru R, Shirato H, Sawamura Y, Shinohe Y, 1998, Selective sensitivity to radiation of cerebral glioblastomas harbouring p53 muttations. Cancer Res 58:1793–1797 Olivier M, Taniere P, 2010, Somatic mutations in cancer prognosis and prediction: lessons from TP53 and EGFR genes. Curr Opin Oncol 23:88–92 Kraus JA, Wenghoefer M, Glesmann N, Mohr S, Beck M, Schmidt MC, Schroder R, Berweiler U, RoggendorfW,Diete S, DietzmannK, Hauser K, Muller B, Fimmers R, von Deimling A, Schlegel U, 2001, TP53 gene mutations, nuclear p53 accumulation, expression of Waf/ p21, Bcl-2, and CD95, APO-1/Fas, proteins are not prognostic factors in de novo glioblastoma multiforme. J Neurooncol 52:263–272 Shiraishi K, Kato S, Han SY, Liu W, Otsuka K, Sakayori M, Ishida T, Takeda M, Kanamaru R, Ochuchi N, Ishioka C, 2004, Isolation of temperature-sensitive p53 mutations from a comprehensive missense mutation library. J Biol Chem 279:348–355 Stefancikova L, Moulis M, Fabian P, Vasova I, Zedek F, Ravcukova B, Muzik J, Kuglik P, Vranova V, Falkova I, Hrabalova R, Smardova J, 2011, Prognostic impact of p53 aberrations for R-CHOP-treated patiens with difuse large B-cell Lymphoma. Int J Oncol 39:1413–1420 Pavlova S, Mayer J, Koukalova H, Smardova J, 2003, High frequency of temperature-sensitive mutations of p53 tumor suppressor in acute myeloid leukemia revealed by functional assay in yeast. Int J Oncol 23:121–131 Smardova J, Ksicova K, Binkova H, Krpensky A, Pavlova S, Rottenberg J, Koukalova H, 2004, Analysis of tumor suppressor p53 status in head and neck squamous cell carcinoma. Oncol Rep 11:923–929 Smardova J, Vagunda V, Jandakova E, Vagundova M, Koukalova H, Kovarik J, Zaloudik J, 1999, p53 status in breast carcinomas revealed by FASAY correlates well with p53 protein accumulation determined by immunohistochemistry. Neoplasma 46:384–389 Nenutil R, Smardova J, Pavlova S, Hanzelkova Z,Muller P, Fabian P, Hrstka R, Janotova P, Radina M, Lane DP, Coates PJ, Vojtesek B, 2005, Discriminating functional and non-functional p53 in human tumours by p53 and MDM2 immunohistochemistry. J Pathol 207:251–259 Fulci G, Ishii N, Maurici D, Gernert KM, Hainaut P, Kaur B, Van Meir EG, 2002, Initiation of human astrocytoma by clonal evolution of cells with progressive loss of p53 functions in a patient with a 283H TP53 germ-line mutation: evidence for a precursor lesion. Cancer Res 62:2897–2905 Sidranski D, Mikkelsen T, Schwechheimer K, Rosenblum ML, Cavanee W, Vogelstein B, 1992, Clonal expansion of p53 mutant cells is associated with brain tumor progression. Nature 355:846– 847 James CD, Carlbom E, Nordenskjold M, Collins VP, Cavenee WK, 1988, Mitotic recombination of chromosome 17 in astrocytomas. Proc Natl Acad Sci USA 86:2858–2862 Bigner SH, Mark J, Burger PC, Mahaley MS, Bullard DE, Muhlbaier LH, Bigner DD, 1988, Specific chromosomal abnormalities in malignant human gliomas. Cancer Res 88:405–411 James CD, Carlbom E, Dumanski JP, Hansen M, Nordenskold M, Collins VP, Cavenee WK, 1989, Clonal genomic alterations in glioma malignancy stages. Cancer Res 48:5546–5551 von Deimling A, Eibl RH, Ohgaki H, Louis DN, von Ammon K, Petersen I, Kliehues P, Chung RY, Wiestler OD, Seizinger BR, 1992, p53 mutations are associated with 17p allelic loss in grade II and III atsrocytomas. Cancer Res 52:2987–2990 Campomenosi P, Monti P, Aprile A, Abbondandolo A, Frebourg T, Gold B, Crook T, Inga A, Resnick MA, Iggo R, Fronza G, 2001, p53 mutants can often transactivate promoters containing p21 but not Bax or PIG3 responsive elements. Oncogene 20:3573–3579 Di Como C, Prives C, 1998, Human tumor-derived p53 proteins exhibit binding site selectivity and temperature sensitivity for transactivation in yeast-based assay. Oncogene 19:2527–2539 Dearth LR, Qian H, Wang T, Baroni TE, Zeng J, Chen SW, Yi SY, Brachmann RK, 2007, Inactive full-length p53 mutants lacking dominant wild-type p53 inhibition highlight loss of heterozygosity as an important aspect of p53 status in human cancers. Carcinogenesis 28:289–298 Yamamoto S, Tada M, Lee CC, Masuda C, Wanibuchi H, Yoshimura R, Wada S, Yamamoto K, Kishimoto T, Fukushima S, 2000, p53 status in multiple human urothelial cancers: assessment for clonality by yeast p53 functional assay in combination with p53 immunohistochemsitry. Jpn J Canc Res 91:181–189 Scharer E, Iggo R, 1992, Mammalian p53 can function as a transcription factor in yeast. Nucl Acid Res 20:1539–1545 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 421 EP 428 DI 10.1007/s12253-012-9596-7 PG 8 ER PT J AU Wang, Q Wang, N Shao, G Qian, J Shen, D Fei, Y Mao, W Wu, D AF Wang, Qiong Wang, Nanyao Shao, Guoyi Qian, Jianzhong Shen, Dong Fei, Yanhua Mao, Weidong Wu, Dan TI Relationship Between Gastric Cancer Tau Protein Expression and Paclitaxel Sensitivity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Tau protein; Paclitaxel; Sensitivity ID Gastric cancer; Tau protein; Paclitaxel; Sensitivity AB The abnormal expression of Tau protein in breast cancer tissue affects paclitaxel sensitivity. The abnormal expression also exists in gastric carcinoma. Therefore, we speculate that the expression levels of Tau protein is closely related to paclitaxel sensitivity in gastric cancer, thus affecting the efficacy of paclitaxel. In this study, we used immunohistochemical methods to detect Tau protein expression levels in 47 cases of gastric cancer specimens. We also used Western blot to detect the level of Tau protein expression in gastric cancer cell lines and to check the efficacy of paclitaxel in vitro application. Findings indicate that Tau protein expression rate can reach as high as (+ +–+ + +) 63.83 % in gastric cancer. Paclitaxel induces inhibition and apoptosis with low expression of Tau protein in gastric cancer cell lines (P<0.05). The level of Tau protein expression is significantly correlated with paclitaxel efficacy. If confirmed by further studies, the Tau protein can be another useful marker of gastric cancer, thereby leading to the application of paclitaxel in cancer treatment. C1 [Wang, Qiong] The Affiliated Jiangyin Hospital of Southeast University Medical College, Department of Oncology, 214400 Wuxi, China. [Wang, Nanyao] The Affiliated Jiangyin Hospital of Southeast University Medical College, Department of Oncology, 214400 Wuxi, China. [Shao, Guoyi] The Affiliated Jiangyin Hospital of Southeast University Medical College, Department of Surgery, 214400 Wuxi, China. [Qian, Jianzhong] The Affiliated Jiangyin Hospital of Southeast University Medical College, Department of Pathology, 214400 Wuxi, China. [Shen, Dong] The Affiliated Jiangyin Hospital of Southeast University Medical College, Department of Oncology, 214400 Wuxi, China. [Fei, Yanhua] The Affiliated Jiangyin Hospital of Southeast University Medical College, Department of Oncology, 214400 Wuxi, China. [Mao, Weidong] The Affiliated Jiangyin Hospital of Southeast University Medical College, Department of Oncology, 214400 Wuxi, China. [Wu, Dan] The Affiliated Jiangyin Hospital of Southeast University Medical College, Department of Oncology, 214400 Wuxi, China. RP Wang, Q (reprint author), The Affiliated Jiangyin Hospital of Southeast University Medical College, Department of Oncology, 214400 Wuxi, China. EM qiongwangcn@163.com CR Zheng LZ, Chen Q, 2005, Gastric cancer chemotherapy status. Gastroenterology 10:178–181 Koizumi W, 2005, Available options in chemotherapy for advanced gastric cancer: the current developments in Japan. Expert Opin Pharmacother 6:225–231 Scartozzi M, Galizia E, Verdecchia L et al, 2007, Chemotherapy for advanced gastric cancer: across the years for a standard of care. Expert Opin Pharmaeother 8:797–808 Gu Y, Oyama F, Ihara Yet al, 1996, Tau is widely expressed in rat tissues. J Neurochem 67:1235–1244 Shen ZL, Qu MH, He HJ et al, 2008, HeLa, HEK293, SH-SY5Y, Tau protein in cells. Res Prog Biochem Biophys 35:1364–1370 Muszynska-Roslan K, Krawczuk-Rybak M, Protas PT et al, 2006, Level of Tau protein in children treated for acute lymphoblastic leukemia. Pediatr Neurol 34:367–371 Van CE, Moiseyenko VM, Tjulandin S et al, 2006, Phase Ul study of docetaxel and eisplatin plus fluorouracil compared with eisphtin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 study group. J Clin Oncol 24:4991–4997 Yan S, 2006, Antitumor handbook. Beijing: Peking University Medical Press 11:178 Wang F, Han R, 2002, Development of research for drugresistance mechanism of taxol. Ai Zheng 21:439–442 Li Z, Wang LM, Geng MY, 2006, Tau protein and nerve cell death. Chin Clinical Rehabilitation 10:124–126 Rossi G, Dalpra L, Crosti F et al, 2008, A new function of microtubule-associated protein tau: involvement in chromosome stability. Cell Cycle 7:1788–1794 Pentheroudakis G, Kalogeras KT, Wirtz RM et al, 2009, Gene expression of estrogen receptor, progesterone receptor and microtubule-associated protein Tau in high-risk early breast cancer: a quest for molecular predictors of treatment benefit in the context of a Hellenic Cooperative Oncology Group trial. Breast Cancer Res Treat 116:131–143 Pusztai L, Jeong JH, Gong Y et al, 2009, Evaluation of microtubule-associated protein-Tau expression as a prognostic and predictive marker in the NSABP-B 28 randomized clinical trial. J Clin Oncol 10:4287–4292 Andre F, Hatzis C, Anderson K et al, 2007, Microtubuleassociated protein-tau is a bifunctional predictor of endocrine sensitivity and chemotherapy resistance in estrogen receptorpositive breast cancer. Clin Cancer Res 13:2061–2067 Wagner P, Wang B, Clark E et al, 2005, Microtubule Associated Protein, MAP)-Tau: a novel mediator of paclitaxel sensitivity in vitro and in vivo. Cell Cycle 4:1149–1152 Rouzier R, Rajan R, Wagner P et al, 2005, Microtubule-associated protein tau: a marker of paclitaxel sensitivity in breast cancer. Proc Natl Acad Sci USA 102:8315–8320 Chambonniere ML, Mosnier-Damet M, Mosnier JF et al, 2001, Expression of microtubule-associated protein tau by gastrointestinal stromal tumors. Hum Pathol 32:1166–1173 Mimori K, Sadanaga N, Yoshikawa Y et al, 2006, Reduced tau expression in gastric cancer can identify candidates for successful Paclitaxel treatment. Br J Cancer 94:1894–1897 Feng R, Bao YZ, Peng CW et al, 2010, Tau gene expression and sensitivity of gastric cancer in paclitaxel treatment. Theory Pract Surg 15:432–437 Qiu LX, Qian XP, Liu B, 2009, Research progress on Taxane drug efficacy forecast molecular. Modern Oncol 17:1583–1584 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 429 EP 435 DI 10.1007/s12253-012-9598-5 PG 7 ER PT J AU Snyder, DA Dulin-Smith, NA Houston, HR Durban, NA Brisbin, JB Oostra, DTy Marshall, TJ Kahwash, MB Pierson, RCh AF Snyder, D Andrew Dulin-Smith, N Ashley Houston, H Ronald Durban, N Ashley Brisbin, J Bethany Oostra, D Tyler Marshall, T Jordan Kahwash, M Basil Pierson, R Christopher TI Expression Pattern of Id Proteins in Medulloblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Medulloblastoma; Id proteins; Id2; Id3; Cerebellum ID Medulloblastoma; Id proteins; Id2; Id3; Cerebellum AB Inhibitor of DNA binding or inhibitor of differentiation (Id) proteins are up regulated in a variety of neoplasms, particularly in association with high-grade, poorly differentiated tumors, while differentiated tissues show little or no Id expression. The four Id genes are members of the helix-loop-helix (HLH) family of transcription factors and act as negative regulators of transcription by binding to and sequestering HLH complexes. We tested the hypothesis that Id proteins are overexpressed in medulloblastoma by performing immunohistochemistry using a medulloblastoma tissue microarray with 45 unique medulloblastoma and 11 normal control cerebella, and antibodies specific for Id1, Id2, Id3, and Id4. A semi-quantitative staining score that took staining intensity and the proportion of immunoreactive cells into account was used. Id1 was not detected in normal cerebella or in medulloblastoma cells, but 78 % of tumors showed strong Id1 expression in endothelial nuclei of tumor vessels. Id2 expression was scant in normal cerebella and increased in medulloblastoma (median staining score: 4). Id3 expression was noted in some neurons of the developing cerebellar cortex, but it was markedly up regulated in medulloblastoma (median staining score: 12) and in tumor endothelial cells. Id4 was not expressed in normal cerebella or in tumor cells. Id2 or Id3 overexpression drove proliferation in medulloblastoma cell lines by altering the expression of critical cell cycle regulatory proteins in favor of cell proliferation. This study shows that Id1 expression in endothelial cells may contribute to angiogenic processes and that increased expression of Id2 and Id3 in medulloblastoma is potentially involved in tumor cell proliferation and survival. C1 [Snyder, D Andrew] Nationwide Children’s Hospital, The Research InstituteColumbus, OH, USA. [Dulin-Smith, N Ashley] Nationwide Children’s Hospital, The Research InstituteColumbus, OH, USA. [Houston, H Ronald] Nationwide Children’s Hospital, Department of Pathology and Laboratory MedicineColumbus, OH, USA. [Durban, N Ashley] Nationwide Children’s Hospital, The Research InstituteColumbus, OH, USA. [Brisbin, J Bethany] Nationwide Children’s Hospital, The Research InstituteColumbus, OH, USA. [Oostra, D Tyler] Nationwide Children’s Hospital, The Research InstituteColumbus, OH, USA. [Marshall, T Jordan] Nationwide Children’s Hospital, The Research InstituteColumbus, OH, USA. [Kahwash, M Basil] Nationwide Children’s Hospital, The Research InstituteColumbus, OH, USA. [Pierson, R Christopher] Nationwide Children’s Hospital, The Research InstituteColumbus, OH, USA. RP Pierson, RCh (reprint author), Nationwide Children’s Hospital, The Research Institute, Columbus, USA. EM christopher.pierson@nationwidechildrens.org CR Louis DN OH, Wiestler OD, Cavenee WK, eds,, 2007, WHO classification of tumors of the central nervous system. Lyon Ris MD, Packer R, Goldwein J, Jones-Wallace D, Boyett JM, 2001, Intellectual outcome after reduced-dose radiation therapy plus adjuvant chemotherapy for medulloblastoma: a Children’s Cancer Group study. J Clin Oncol 19(15):3470–3476 Perk J, Iavarone A, Benezra R, 2005, Id family of helix-loop-helix proteins in cancer. Nat Rev Cancer 5(8):603–614 Jen Y, Manova K, Benezra R, 1997, Each member of the Id gene family exhibits a unique expression pattern in mouse gastrulation and neurogenesis. Dev Dyn 208(1):92–106 Lyden D, Young AZ, Zagzag D, YanW, GeraldW, O’Reilly R et al, 1999, Id1 and Id3 are required for neurogenesis, angiogenesis and vascularization of tumour xenografts. Nature 401(6754):670–677 Neuman T, Keen A, Zuber MX, Kristjansson GI, Gruss P, Nornes HO, 1993, Neuronal expression of regulatory helix-loop-helix factor Id2 gene in mouse. Dev Biol 160(1):186–195 Fong S, Debs RJ, Desprez PY, 2004, Id genes and proteins as promising targets in cancer therapy. Trends Mol Med 10(8):387– 392 Ruzinova MB, Benezra R, 2003, Id proteins in development, cell cycle and cancer. Trends Cell Biol 13(8):410–418 Bain G, Cravatt CB, Loomans C, Alberola-Ila J, Hedrick SM,Murre C, 2001, Regulation of the helix-loop-helix proteins, E2A and Id3, by the Ras-ERK MAPK cascade. Nat Immunol 2(2):165–171 Deed RW, Hara E, Atherton GT, Peters G, Norton JD, 1997, Regulation of Id3 cell cycle function by Cdk-2-dependent phosphorylation. Mol Cell Biol 17(12):6815–6821 Yokota Y, Mori S, 2002, Role of Id family proteins in growth control. J Cell Physiol 190(1):21–28 Zebedee Z, Hara E, 2001, Id proteins in cell cycle control and cellular senescence. Oncogene 20(58):8317–8325 Fong S, Itahana Y, Sumida T, Singh J, Coppe JP, Liu Y et al, 2003, Id-1 as a molecular target in therapy for breast cancer cell invasion and metastasis. Proc Natl Acad Sci U S A 100, 23):13543–13548 Loveys DA, Streiff MB, Kato GJ, 1996, E2A basic-helix-loophelix transcription factors are negatively regulated by serum growth factors and by the Id3 protein. Nucleic Acids Res 24, 14):2813–2820 Tzeng SF, 2003, Inhibitors of DNA binding in neural cell proliferation and differentiation. Neurochem Res 28(1):45–52 Tzeng SF, de Vellis J, 1998, Id1, Id2, and Id3 gene expression in neural cells during development. Glia 24(4):372–381 Ellmeier W, Aguzzi A, Kleiner E, Kurzbauer R, Weith A, 1992, Mutually exclusive expression of a helix-loop-helix gene and Nmyc in human neuroblastomas and in normal development. EMBO J 11(7):2563–2571 Duncan M, DiCicco-Bloom EM, Xiang X, Benezra R, Chada K, 1992, The gene for the helix-loop-helix protein, Id, is specifically expressed in neural precursors. Dev Biol 154(1):1–10 Ellmeier W, Weith A, 1995, Expression of the helix-loop-helix gene Id3 during murine embryonic development. Dev Dyn 203, 2):163–173 Vandeputte DA, Troost D, Leenstra S, Ijlst-Keizers H, Ramkema M, Bosch DA et al, 2002, Expression and distribution of id helix-loop-helix proteins in human astrocytic tumors. Glia 38, 4):329–338 Kee Y, Bronner-Fraser M, 2005, To proliferate or to die: role of Id3 in cell cycle progression and survival of neural crest progenitors. Genes Dev 19(6):744–755 Prabhu S, Ignatova A, Park ST, Sun XH, 1997, Regulation of the expression of cyclin-dependent kinase inhibitor p21 by E2A and Id proteins. Mol Cell Biol 17(10):5888–5896 Ayrault O, Zindy F, Rehg J, Sherr CJ, Roussel MF, 2009, Two tumor suppressors, p27Kip1 and patched-1, collaborate to prevent medulloblastoma. Mol Cancer Res 7(1):33–40 Miyazawa K, Himi T, Garcia V, Yamagishi H, Sato S, Ishizaki Y, 2000, A role for p27/Kip1 in the control of cerebellar granule cell precursor proliferation. J Neurosci 20(15):5756–5763 Phi JH, Kim JH, Eun KM,Wang KC, Park KH, Choi SA et al, 2010, Upregulation of SOX2, NOTCH1, and ID1 in supratentorial primitive neuroectodermal tumors: a distinct differentiation pattern from that of medulloblastomas. J Neurosurg Pediatr 5(6):608–614 Taylor MD, Northcott PA, Korshunov A, Remke M, Cho YJ, Clifford SC et al, 2012, Molecular subgroups of medulloblastoma: the current consensus. Acta Neuropathol 123(4):465–472 Oliver TG, Grasfeder LL, Carroll AL, Kaiser C, Gillingham CL, Lin SM et al, 2003, Transcriptional profiling of the Sonic hedgehog response: a critical role for N-myc in proliferation of neuronal precursors. Proc Natl Acad Sci U S A 100(12):7331–7336 Lasorella A, Stegmuller J, Guardavaccaro D, Liu G, Carro MS, Rothschild G et al, 2006, Degradation of Id2 by the anaphasepromoting complex couples cell cycle exit and axonal growth. Nature 442(7101):471–474 Iavarone A, Garg P, Lasorella A, Hsu J, Israel MA, 1994, The helix-loop-helix protein Id-2 enhances cell proliferation and binds to the retinoblastoma protein. Genes Dev 8(11):1270–1284 Lasorella A, Noseda M, Beyna M, Yokota Y, Iavarone A, 2000, Id2 is a retinoblastoma protein target and mediates signalling by Myc oncoproteins. Nature 407(6804):592–598 Ohtani N, Zebedee Z, Huot TJ, Stinson JA, Sugimoto M, Ohashi Yet al, 2001, Opposing effects of Ets and Id proteins on p16INK4a expression during cellular senescence. Nature 409(6823):1067–1070 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 437 EP 446 DI 10.1007/s12253-012-9599-4 PG 10 ER PT J AU Mu, F Liu, ShP Zhou, XL Chen, JB Li, HB Zuo, JSh Xu, KCh AF Mu, Feng Liu, Shu-Peng Zhou, Xu-Long Chen, Ji-Bing Li, Hai-Bo Zuo, Jian-Sheng Xu, Ke-Cheng TI Prevention of Needle-Tract Seeding by Two-Step Freezing after Lung Cancer Biopsy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Biopsy; Percutaneous cryoablation; Lung cancer; Pathology ID Biopsy; Percutaneous cryoablation; Lung cancer; Pathology AB Fine-needle aspiration biopsy is a method to detect malignancy for undetermined pulmonary nodules, but has the potential to spread malignant cells from the tumor to the pleural cavity or chest wall. We developed a two-step freezing method to avoid needle-tract seeding, by use of percutaneous cryoablation after biopsy but before the biopsy needle was removed. A man aged 72 years was admitted because of a large mass in right upper lobe. After biopsy, the patient underwent surgery. Pathological assessment of the resected tumor showed that tissue around the biopsy probe and cryoprobe had been killed before needle withdrawal. C1 [Mu, Feng] Chinese Academy of Science, Institutes of Biomedicine and Health, No. 91 Judezhong Road, Haizhu District, 510305 Guangzhou, Guangdong, China. [Liu, Shu-Peng] Chinese Academy of Science, Institutes of Biomedicine and Health, No. 91 Judezhong Road, Haizhu District, 510305 Guangzhou, Guangdong, China. [Zhou, Xu-Long] Chinese Academy of Science, Institutes of Biomedicine and Health, No. 91 Judezhong Road, Haizhu District, 510305 Guangzhou, Guangdong, China. [Chen, Ji-Bing] Chinese Academy of Science, Institutes of Biomedicine and Health, No. 91 Judezhong Road, Haizhu District, 510305 Guangzhou, Guangdong, China. [Li, Hai-Bo] Chinese Academy of Science, Institutes of Biomedicine and Health, No. 91 Judezhong Road, Haizhu District, 510305 Guangzhou, Guangdong, China. [Zuo, Jian-Sheng] Chinese Academy of Science, Institutes of Biomedicine and Health, No. 91 Judezhong Road, Haizhu District, 510305 Guangzhou, Guangdong, China. [Xu, Ke-Cheng] Chinese Academy of Science, Institutes of Biomedicine and Health, No. 91 Judezhong Road, Haizhu District, 510305 Guangzhou, Guangdong, China. RP Chen, JB (reprint author), Chinese Academy of Science, Institutes of Biomedicine and Health, 510305 Guangzhou, China. EM jibingchen@yahoo.com.cn CR Moloo Z, Finley RJ, Lefcoe MS, Turner-Smith L, Craig ID, 1985, Possible spread of bronchogenic carcinoma to the chest wall after a transthoracic fine needle aspiration biopsy. A case report. Acta Cytol 29:167–169 Sacchini V, Galimberti V, Marchini S, Luini A, 1989, Percutaneous transthoracic needle aspiration biopsy: a case report of implantation metastasis. Eur J Surg Oncol 15:179–183 Seyfer AE, Walsh DS, Graeber GM, Nuno IN, Eliasson AH, 1989, Chest wall implantation of lung cancer after thin-needle aspiration biopsy. Ann Thorac Surg 48:284–286 Voravud N, Shin DM, Dekmezian RH, Dimery I, Lee JS, Hong WK, 1992, Implantation metastasis of carcinoma after percutaneous fine-needle aspiration biopsy. Chest 102:313–315 Sing RF, Kefalides PT, Mette SA, Fallahnejad M, 1996, Chest wall metastasis after percutaneous fine-needle aspiration biopsy. J Am Osteopath Assoc 96:546–547 Komiya T, Kusunoki Y, Kobayashi M, Hirashima T, Yana T, Masuda N et al, 1997, Transcutaneous needle biopsy of the lung. Acta Radiol 38:821–825 Sawabata N, Ohta M, Maeda H, 2000, Fine-needle aspiration cytologic technique for lung cancer has a high potential of malignant cell spread through the tract. Chest 118:936–939 Vignoli M, Rossi F, Chierici C, Terragni R, De Lorenzi D, Stanga M et al, 2007, Needle tract implantation after fine needle aspiration biopsy, FNAB, of transitional cell carcinoma of the urinary bladder and adenocarcinoma of the lung. Schweiz Arch Tierheilkd 149:314–318 Inoue M, Nakatsuka S, Yashiro H, Ito N, Izumi Y, Yamauchi Y et al, 2012, Percutaneous cryoablation of lung tumors: feasibility and safety. J Vasc Interv Radiol 23:295–302, quiz 305 Yamauchi Y, Izumi Y, Kawamura M, Nakatsuka S, Yashiro H, Tsukada N et al, 2011, Percutaneous cryoablation of pulmonary metastases from colorectal cancer. PLoS One 6:e27086 Yamauchi Y, Izumi Y, Hashimoto K, Inoue M, Nakatsuka S, Kawamura M et al, 2011, Needle-tract seeding after percutaneous cryoablation for lung metastasis of colorectal cancer. Ann Thorac Surg 92:e69–e71 Kawamura M, Izumi Y, Tsukada N, Asakura K, Sugiura H, Yashiro H et al, 2006, Percutaneous cryoablation of small pulmonary malignant tumors under computed tomographic guidance with local anesthesia for nonsurgical candidates. J Thorac Cardiovasc Surg 131:1007–1013 Niu L, Li J, Chen J, Zhou L, Wu B, Zeng J et al, 2012, Comparison of dual- and triple-freeze protocols for pulmonary cryoablation in a Tibet pig model. Cryobiology 64:245–249 Niu L, Zhou L, Korpan NN, Wu B, Tang J, Mu F et al, 2012, Experimental study on pulmonary cryoablation in a porcine model of normal lungs. Technol Cancer Res Treat 11:389–394 Hashimoto K, Izumi Y, Yamauchi Y, Yashiro H, Inoue M, Nakatsuka S et al, 2012, Prediction of the critical thermal zone during pulmonary cryoablation on computed tomography from correlated experimental and clinical findings. J Thorac Cardiovasc Surg., DOI 10.1016/j.jtcvs.2012.03.029 Hinshaw JL, Littrup PJ, Durick N, Leung W, Lee FT Jr, Sampson L et al, 2010, Optimizing the protocol for pulmonary cryoablation: a comparison of a dual- and triple-freeze protocol. Cardiovasc Intervent Radiol 33:1180–1185 Permpongkosol S, Nicol TL, Link RE, Varkarakis I, Khurana H, Zhai QJ et al, 2007, Differences in ablation size in porcine kidney, liver, and lung after cryoablation using the same ablation protocol. AJR Am J Roentgenol 188:1028–1032 Nakatsuka S, Yashiro H, Inoue M, Kuribayashi S, Kawamura M, Izumi Y et al, 2010, On freeze-thaw sequence of vital organ of assuming the cryoablation for malignant lung tumors by using cryoprobe as heat source. Cryobiology 61:317–326 Berner A, Lund-Iversen M, Nesland JM, 2011, Fine needle aspirations in oncology. Arkh Patol 73:21–26 Ivorra A, 2011, Electrochemical prevention of needle-tract seeding. Ann Biomed Eng 39:2080–2089 Azlan CA, Mohd Nasir NF, Saifizul AA, Faizul MS, Ng KH, Abdullah BJ, 2007, A low cost solution for post-biopsy complications using available RFA generator and coaxial core biopsy needle. Australasian physical & engineering sciences in medicine/ supported by the Australasian College of Physical Scientists in Medicine and the Australasian Association of Physical Sciences in Medicine 30: 288–291 Littrup PJ, Jallad B, Vorugu V, Littrup G, Currier B, GeorgeMet al, 2009, Lethal isotherms of cryoablation in a phantom study: effects of heat load, probe size, and number. J Vasc Interv Radiol 20:1343–1351 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 447 EP 450 DI 10.1007/s12253-012-9601-1 PG 4 ER PT J AU Sassi, A Popielarski, M Synowiec, E Morawiec, Z Wozniak, K AF Sassi, Agnieszka Popielarski, Marcin Synowiec, Ewelina Morawiec, Zbigniew Wozniak, Katarzyna TI BLM and RAD51 Genes Polymorphism and Susceptibility to Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Genepolymorphism; BLM gene; RAD51 gene; DNA double-strand breaks ID Breast cancer; Genepolymorphism; BLM gene; RAD51 gene; DNA double-strand breaks AB DNA repair by homologous recombination is one of the main processes of DNA double strand breaks repair. In the present work we performed a case-control study (304 cases and 319 controls) to check an association between the genotypes of the c.-61 G>T and the g.38922 C>G polymorphisms of the RAD51 gene and the g.96267 A>C and the g.85394 A>G polymorphisms of the BLM gene and breast cancer occurrence. Genotypes were determined in DNA from peripheral blood by PCR-RLFP and by PCRCTPP. We observed an association between breast cancer occurrence and the T/G genotype (OR 4.41) of the c.-61 G>T-RAD51 polymorphism, the A/A genotype (OR 1.69) of the g.85394 A>G-BLM polymorphism and the A/A genotype (OR 2.49) of the g.96267 A>C-BLM polymorphism. Moreover, we demonstrated a correlation between intra- and intergenes genotypes combinations and breast cancer occurrence. We found a correlation between progesterone receptor expression and the T/G genotype (OR 0.57) of the c.-61 G>T- RAD51 polymorphism. We also found a correlation between the T/G genotype (OR 1.86) and the T/T genotype (OR 0.56) of the c.-61 G>T- RAD51 polymorphism and the lymph node metastasis. We showed an association between the A/A genotype (OR 2.45) and the A/C genotype (OR 0.41) of the g.96267 A>C-BLM polymorphism and G3 grade of tumor. Our results suggest that the variability of the RAD51 and BLM genes may play a role in breast cancer occurrence. This role may be underlined by a common interaction between these genes. C1 [Sassi, Agnieszka] University of Lodz, Department of Molecular GeneticsLodz, Poland. [Popielarski, Marcin] University of Lodz, Department of Molecular GeneticsLodz, Poland. [Synowiec, Ewelina] University of Lodz, Department of Molecular GeneticsLodz, Poland. [Morawiec, Zbigniew] University of Lodz, Department of Molecular GeneticsLodz, Poland. [Wozniak, Katarzyna] University of Lodz, Department of Molecular GeneticsLodz, Poland. RP Wozniak, K (reprint author), University of Lodz, Department of Molecular Genetics, Lodz, Poland. EM wozniak@biol.uni.lodz.pl CR Patel KJ, Yu VP, Lee H, Corcoran TFC, Evans MJ, Colledge WH, Friedman LS, Ponder BA, Venkitaraman AR, 1998, Involvement of BRCA2 in DNA repair. Mol Cell 1:347–357 Moynahan ME, Chiu W, Koller BH, Jasin M, 1999, BRCA1 controls homology-directed DNA repair. Mol Cell 4:511–518 Thompson LH, Schild D, 2002, Recombinational DNA repair and human disease. Mutat Res 509:49–78 Helleday T, Lo J, van Gent DC, Engelward BP, 2007, DNA double-strand break repair: from mechanistic understanding to cancer treatment. DNA Repair 6:923–935 Hsu HM, Wang HC, Chen ST, Hsu GC, Shen CY, Yu JC, 2007, Breast cancer risk is associated with the genes encoding the DNA double-strand break repair Mre11/Rad50/Nbs1 complex. Cancer Epidemiol Biomarkers Prev 16:2024–2032 Ralhan R, Kaur J, Kreienberg R, Wiesmuller L, 2007, Links between DNA double strand break repair and breast cancer: accumulating evidence from both familial and nonfamilial cases. Cancer Lett 248:1–17 Lieber MR, 2010, The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu Rev Biochem 79:181–211 Thompson LH, Schild D, 2001, Homologous recombinational repair of DNA ensures mammalian chromosome stability. Mutat Res 477:131–153 Mansour WY, Schumacher S, Rosskopf R, Rhein T, Schmidt- Petersen F, Gatzemeier F, Haag F, Borgmann K, Willers H, Dahm- Daphi J, 2008, Hierarchy of nonhomologous end-joining, singlestrand annealing and gene conversion at site-directed DNA doublestrand breaks. Nucleic Acids Res 36:4088–4098 Tracker J, 2005, The RAD51 gene family, genetic instability and cancer. Cancer Lett 219:125–135 Sung P, Krejci L, Van Komen S, Sehorn MG, 2003, Rad51 recombinase and recombination mediators. J Biol Chem 278:42729–42732 Balakrishnan K, Krishnan NM, Kulkarni A, Rao BJ, 2009, Human Rad51 mediated DNA unwinding is facilitated by conditions that favour Rad51-dsDNA aggregation. BMC Biochem 10:1–15 Van Mameren J, Modesti M, Kanaar R, Wyman C, Peterman EJ, Wuite GJ, 2009, Counting RAD51 proteins disassembling from nucleoprotein filaments under tension. Nature 457:745–748 Karow JK, Constantinou A, Li JL, West SC, Hickson ID, 2000, The Bloom’s syndrome gene product promotes branch migration of Holliday junctions. Proc Natl Acad Sci USA 97:6504–6508 Wu L, Davies SL, Levitt NC, Hickson ID, 2001, Potential role for the BLM helicase in recombinational repair via a conserved interaction with RAD51. J Biol Chem 276:19375–19381 Sung P, Robberson DL, 1995, DNA strand exchange mediated by a RAD51-ssDNA nucleoprotein filament with polarity opposite to that of RecA. Cell 82:453–461 Bugreev DV, Mazina OM, Mazin AV, 2009, Bloom syndrome helicase stimulates RAD51 DNA strand exchange activity through a novel mechanism. J Biol Chem 39:26349–26359 Tambini CE, Spink KG, Ross CJ, Hill MA, Thacker J, 2010, The importance of XRCC2 in RAD51-related DNA damage repair. DNA Repair 9:517–525 Yoshikawa K, Ogawa T, Baer R, Hemmi H, Honda K, Yamauchi A, Inamoto T, Ko K, Yazumi S, Motoda H, Kodama H, Noguchi S, Gazdar AF, Yamaoka Y, Takahashi R, 2000, Abnormal expression of BRCA1 and BRCA2-interactive DNA-repair proteins in breast carcinomas. Int J Cancer 88:28–36 Gonzales R, Silva JM, Dominguez G, Garcia JM, Martinez G, Vargas J, Provencio M, Espana P, Bonilla F, 1999, Detection of loss of heterozygosity at RAD51, RAD52, RAD54 and BRCA1 and BRCA2 loci in breast cancer: pathological correlations. Br J Cancer 81:503–509 Chester N, Babbe H, Pinkas J, Manning C, Leder P, 2006, Mutation of the murine Bloom’s syndrome gene produces global genome destabilization. Mol Cell Biol 26:6713–6726 Sliwinski T, Krupa R, Majsterek I, Rykala J, Kolacinska A, Morawiec Z, Drzewoski J, Zadrozny M, Blasiak J, 2005, Polymorphisms of the BRCA2 and RAD51 genes in breast cancer. Breast Cancer Res Treat 94:105–109 Synowiec E, Stefanska J, Morawiec Z, Blasiak J, Wozniak K, 2008, Association between DNA damage, DNA repair genes variability and clinical characteristics in breast cancer patients. Mutat Res 648:65–72 Synowiec E, Krupa R, Morawiec Z, Wasylecka M, Dziki L, Morawiec J, Blasiak J, Wozniak K, 2010, Efficacy of DNA double-strand breaks repair in breast cancer is decreased in carriers of the variant allele of the UBC9 gene c.73G>A polymorphism. Mutat Res 694:31–38 Krupa R, Synowiec E, Pawlowska E, Morawiec Z, Sobczuk A, Zadrozny M, Wozniak K, Blasiak J, 2009, Polymorphism of the homologous recombination repair genes RAD51 and XRCC3 in breast cancer. Exp Mol Path 87:32–35 Ding S, JCh Y, Chen ST, Hsu GC, Kuo SJ, Lin YH, Wu PE, Shen CY, 2009, Genetic variants of BLM interact with RAD51 to increase breast cancer susceptibility. Carcinogenesis 30:43–49 Hadjisavvas A, Loizidou MA, Middleton N, Michael T, Papachristoforou R, Kakouri E, Daniel M, Papadopoulos P, Malas S, Marcou Y, Kyriacou K, 2010, An investigation of breast cancer risk factors in Cyprus: a case control study. BMC Cancer 10:1–10 Wang Y, Cortez D, Yazdi P, Neff N, Elledge SJ, Qin J, 2000, BASC, a super complex of BRCA1-associated proteins involved in the recognition and repair of aberrant DNA structure. Genes Dev 14:927–939 Bergeron KL, Murphy EL, Brown LW, Almeida KH, 2011, Critical interaction domains between Bloom Syndrome protein and RAD51. Protein J 30:1–8 Bugreev DV, Yu X, Egelman EH, Mazin AV, 2007, Novel proand anti-recombination activities of the Bloom’s syndrome helicase. Genes Dev 21:3085–3094 Ouyang KJ, Woo LL, Zhu J, Huo D, Matunis MJ, Ellis NA, 2009, SUMO modification regulates BLM and RAD51 interaction at damaged replication forks. PLoS Biol 7:1–12 Le Scodan R, Cizeron-Clairac G, Fourme E, Meseure D, Vacher S, Spyratos F, de la Lande B, Cvitkovic F, Lidereau R, Bieche I, 2010, DNArepair gene expression and risk of locoregional relapse in breast cancer patients. Int J Radiat Oncol Biol Phys 78:328–336 Barbano R, Copetti M, Perrone G, Pazienza V, Muscarella LA, Balsamo T, Storlazzi CT, Ripoli M, Rinaldi M, Valori VM, Latiano TP, Maiello E, Stanziale P, Carella M, Mangia A, Pellegrini F, Bisceglia M, Muda AO, Altomare V, Murgo R, Fazio VM, Parrella P, 2011, High RAD51 mRNA expression characterize estrogen receptor-positive/progesteron receptor-negative breast cancer and is associated with patient’s outcome. Int J Cancer 129:536–545 Bachrati CZ, Hickson JD, 2003, RecQ helicases: suppressors of tumorigenesis and premature aging. Biochem J 374:577–606 Ma CX, Sanchez CG, Ellis MJ, 2009, Predicting endocrine therapy responsiveness in breast cancer patients. Oncology 23:133–142 Revillon F, Bonneterre J, Peyrat JP, 1998, ERBB2 oncogene in human breast cancer and its clinical significance. Eur J Cancer 34:791–808 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 451 EP 459 DI 10.1007/s12253-013-9602-8 PG 9 ER PT J AU Wang, LP Chen, ShW Zhuang, ShM Li, H Song, M AF Wang, Li-Ping Chen, Shu-Wei Zhuang, Shi-Min Li, Huan Song, Ming TI Galectin-3 Accelerates the Progression of Oral Tongue Squamous Cell Carcinoma via a Wnt/β-catenin-Dependent Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Galectin-3; Oral tongue squamous cell carcinoma; Wnt/β-catenin; Clinicopathological significance; Mechanism ID Galectin-3; Oral tongue squamous cell carcinoma; Wnt/β-catenin; Clinicopathological significance; Mechanism AB The purpose of this study was to elucidate the clinicopathological significance and mechanism of action of galectin-3 in oral tongue squamous cell carcinoma (OTSCC). Here, the expression of galectin-3 was quantified in OTSCC (n=68) and paired OTSCC and normal surrounding tissues (n=10) using immunohistochemical staining. Tca8113 OTSCC cells were transfected with a plasmid expressing galectin-3 cDNA or siRNA against galectin-3. Cell proliferation, migration and invasion were measured using the MTT assay, Matrigel-coated Transwell migration assay and wound healing assay. The effect of galectin-3 on the Wnt/β-catenin signaling pathway and epithelial mesenchymal transition (EMT) were investigated using a plasmid expressing the Wnt antagonist dickkopf 1 (DKK1) and Western blotting. Galectin-3 was expressed at significantly higher levels in OTSCC than the normal adjacent tissues; galectin-3 expression correlated strongly with pathological stage, pathological grade and lymph node invasion in OTSCC. Overexpression of galectin-3 promoted Tca8113 cell proliferation, migration and invasion, upregulated Wnt protein expression, activated β-catenin and induced the EMT; knockdown of galectin-3 had the opposite effects. Co-transfection of Tca8113 cells overexpressing galectin-3 with the Wnt antagonist DKK1 reduced the ability of galectin-3 to increase cell proliferation, migration and invasion, reduced upregulation of Wnt, inhibited β-catenin activation and abrogated the EMT, demonstrating that the Wnt/β-catenin signaling pathway mediated the effects of galectin-3. Galectin-3 plays an important role in the progression of OTSCC via activation of the Wnt/β-catenin signaling pathway. C1 [Wang, Li-Ping] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. [Chen, Shu-Wei] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. [Zhuang, Shi-Min] The Third Affiliated Hospital of Sun Yat-sen University, Department of Otolaryngology-Head & Neck SurgeryGuangzhou, China. [Li, Huan] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. [Song, Ming] Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 651 Dongfeng Dong Road, 510060 Guangzhou, China. RP Song, M (reprint author), Sun Yat-sen University Cancer Center, Department of Head and Neck Surgery, 510060 Guangzhou, China. EM songming@sysucc.org.cn CR Cooper JS, Porter K, Mallin K et al, 2009, National Cancer Database report on cancer of the head and neck: 10-year update. Head Neck 31:748–758 Chitapanarux I, Lorvidhaya V, Sittitrai P et al, 2006, Oral cavity cancers at a young age: analysis of patient, tumor and treatment characteristics in Chiang Mai University Hospital. Oral Oncol 42:83–88 Hiratsuka H, Miyakawa A, Nakamori K, Kido Y, Sunakawa H, Kohama G, 1997, Multivariate analysis of occult lymph node metastasis as a prognostic indicator for patients with squamous cell carcinoma of the oral cavity. Cancer 80:351–356 Kowalski LP, Bagietto R, Lara JR, Santos RL, Silva JJ, Magrin J, 2000, Prognostic significance of the distribution of neck node metastasis from oral carcinoma. Head Neck 22:207–214 Lo WL, Kao SY, Chi LY, Wong YK, Chang RC, 2003, Outcomes of oral squamous cell carcinoma in Taiwan after surgical therapy: factors affecting survival. J Oral Maxillofac Surg 61:751–758 Cooper DN, 2002, Galectinomics: finding themes in complexity. Biochim Biophys Acta 1572:209–231 Cooper DN, Barondes SH, 1999, God must love galectins; he made so many of them. Glycobiology 9:979–984 Yang RY, Rabinovich GA, Liu FT, 2008, Galectins: structure, function and therapeutic potential. Expert Rev Mol Med 10:e17 Krzeslak A, Lipinska A, 2004, Galectin-3 as a multifunctional protein. Cell Mol Biol Lett 9:305–328 Chiu CG, Strugnell SS, Griffith OL et al, 2010, Diagnostic utility of galectin-3 in thyroid cancer. Am J Pathol 176:2067–2081 Wang Y, Nangia-Makker P, Tait L, Balan V, Hogan V, Pienta KJ, Raz A, 2009, Regulation of prostate cancer progression by galectin-3. Am J Pathol 174:1515–1523 Zhao Q, Guo X, Nash GB, Stone PC, Hilkens J, Rhodes JM, Yu LG, 2009, Circulating galectin-3 promotes metastasis by modifying MUC1 localization on cancer cell surface. Cancer Res 69:6799–6806 Hill M, Mazal D, Biron VA et al, 2010, A novel clinically relevant animal model for studying galectin-3 and its ligands during colon carcinogenesis. J Histochem Cytochem 58:553–565 Miranda FA, Hassumi MK, Guimaraes MC et al, 2009, Galectin-3 overexpression in invasive laryngeal carcinoma, assessed by computer-assisted analysis. J Histochem Cytochem 57:665–673 Honjo Y, Inohara H, Akahani S et al, 2000, Expression of cytoplasmic galectin-3 as a prognostic marker in tongue carcinoma. Clin Cancer Res 6:4635–4640 Alves PM, Godoy GP, Gomes DQ et al, 2011, Significance of galectins-1, -3, -4 and -7 in the progression of squamous cell carcinoma of the tongue. Pathol Res Pract 207:236–240 Matarrese P, Fusco O, Tinari N et al, 2000, Galectin-3 overexpression protects from apoptosis by improving cell adhesion properties. Int J Cancer 85:545–554 Giannopoulou M, Dai C, Tan X, Wen X, Michalopoulos GK, Liu Y, 2008, Hepatocyte growth factor exerts its anti-inflammatory action by disrupting nuclear factor-kappaB signaling. Am J Pathol 173:30–41 Kliment CR, Englert JM, Gochuico BR, Yu G, Kaminski N, Rosas I, Oury TD, 2009, Oxidative stress alters syndecan-1 distribution in lungs with pulmonary fibrosis. J Biol Chem 284:3537–3545 Duan L, Yao J, Wu X, Fan M, 2006, Growth suppression induced by Notch1 activation involves Wnt-beta-catenin down-regulation in human tongue carcinoma cells. Biol Cell 98:479–490 Fracalossi AC, Silva MS, Oshima CT, Ribeiro DA, 2010, Wnt/ beta-catenin signalling pathway following rat tongue carcinogenesis induced by 4-nitroquinoline 1-oxide. Exp Mol Pathol 88:176– 183 Lo ML, 2001, A possible role for the WNT-1 pathway in oral carcinogenesis. Crit Rev Oral Biol Med 12:152–165 Moon RT, Bowerman B, BoutrosM, PerrimonN, 2002, The promise and perils ofWnt signaling through beta-catenin. Science 296:1644– 1646 Yan D, Avtanski D, Saxena NK, Sharma D, 2012, Leptin-induced epithelial-mesenchymal transition in breast cancer cells requires beta-catenin activation via Akt/GSK3- and MTA1/Wnt1 proteindependent pathways. J Biol Chem 287:8598–8612 Howard S, Deroo T, Fujita Y, Itasaki N, 2011, A positive role of cadherin in Wnt/beta-catenin signalling during epithelialmesenchymal transition. PLoS One 6:e23899 Kawano Y, Kypta R, 2003, Secreted antagonists of the Wnt signalling pathway. J Cell Sci 116:2627–2634 Kocemba KA, Groen RW, van Andel H, Kersten MJ, Mahtouk K, Spaargaren M, Pals ST, 2012, Transcriptional silencing of the Wnt-antagonist DKK1 by promoter methylation is associated with enhanced Wnt signaling in advanced multiple myeloma. PLoS One 7:e30359 Liu FT, Rabinovich GA, 2005, Galectins as modulators of tumour progression. Nat Rev Cancer 5:29–41 Saussez S, Lorfevre F, Lequeux T et al, 2008, The determination of the levels of circulating galectin-1 and -3 in HNSCC patients could be used to monitor tumor progression and/or responses to therapy. Oral Oncol 44:86–93 Shimura T, Takenaka Y, Tsutsumi S, Hogan V, Kikuchi A, Raz A, 2004, Galectin-3, a novel binding partner of beta-catenin. Cancer Res 64:6363–6367 Weinberger PM, Adam BL, Gourin CG et al, 2007, Association of nuclear, cytoplasmic expression of galectin-3 with beta-catenin/ Wnt-pathway activation in thyroid carcinoma. Arch Otolaryngol Head Neck Surg 133:503–510 Song S, Mazurek N, Liu C et al, 2009, Galectin-3 mediates nuclear beta-catenin accumulation and Wnt signaling in human colon cancer cells by regulation of glycogen synthase kinase-3beta activity. Cancer Res 69:1343–1349 Staal FJ, Sen JM, 2008, The canonical Wnt signaling pathway plays an important role in lymphopoiesis and hematopoiesis. Eur J Immunol 38:1788–1794 Logan CY, Nusse R, 2004, The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol 20:781–810 Gan XQ, Wang JY, Xi Y, Wu ZL, Li YP, Li L, 2008, Nuclear Dvl, c-Jun, beta-catenin, and TCF form a complex leading to stabilization of beta-catenin-TCF interaction. J Cell Biol 180:1087–1100 Hu MC, Rosenblum ND, 2005, Smad1, beta-catenin and Tcf4 associate in a molecular complex with the Myc promoter in dysplastic renal tissue and cooperate to control Myc transcription. Development 132:215–225 Udhayakumar G, Jayanthi V, Devaraj N, Devaraj H, 2007, Interaction of MUC1 with beta-catenin modulates the Wnt target gene cyclinD1 in H. pylori-induced gastric cancer. Mol Carcinog 46:807–817 Rahmani M, Carthy JM, McManus BM, 2012, Mapping of the Wnt/beta-catenin/TCF response elements in the human versican promoter. Methods Mol Biol 836:35–52 Zeilstra J, Joosten SP, Wensveen FM et al, 2011, WNT signaling controls expression of pro-apoptotic BOK and BAX in intestinal cancer. Biochem Biophys Res Commun 406:1–6 Sancho R, Nateri AS, de Vinuesa AG, Aguilera C, Nye E, Spencer- Dene B, Behrens A, 2009, JNK signalling modulates intestinal homeostasis and tumourigenesis in mice. EMBO J 28:1843–1854 Saadeddin A, Babaei-Jadidi R, Spencer-Dene B, Nateri AS, 2009, The links between transcription, beta-catenin/JNK signaling, and carcinogenesis. Mol Cancer Res 7:1189–1196 Giles RH, van Es JH, Clevers H, 2003, Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta 1653:1– 24 Levy R, Biran A, Poirier F, Raz A, Kloog Y, 2011, Galectin-3 mediates cross-talk between K-Ras and Let-7c tumor suppressor microRNA. PLoS One 6:e27490 Elad-Sfadia G, Haklai R, Balan E, Kloog Y, 2004, Galectin-3 augments K-Ras activation and triggers a Ras signal that attenuates ERK but not phosphoinositide 3-kinase activity. J Biol Chem 279:34922–34930 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 461 EP 474 DI 10.1007/s12253-013-9603-7 PG 14 ER PT J AU Zhang, K Bai, P Shi, Sh Zhou, B Wang, Y Song, Y Rao, L Zhang, L AF Zhang, Kui Bai, Peng Shi, Shaoqing Zhou, Bin Wang, Yanyun Song, Yaping Rao, Li Zhang, Lin TI Association of Genetic Variations in RTN4 3’-UTR with Risk of Uterine Leiomyomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE RTN4; Uterine Leiomyoma (UL); Polymorphism; Genetic susceptibility ID RTN4; Uterine Leiomyoma (UL); Polymorphism; Genetic susceptibility AB This pilot case-control study was conducted to test the hypothesis that the TATC (rs71682890) and CAA (rs34917480) insertion/deletion polymorphisms of RTN4 3’- UTR are associated with the susceptibility to uterine leiomyoma (UL). The study recruited 286 premenopausal women with UL and 450 unrelated postmenopausal women not presenting the disease as control subjects. The polymorphisms of rs71682890 and rs34917480 were genotyped with the method of polymerase chain reaction polyacrylamide gel electrophoresis (PCR - PAGE). No statistically significant association was observed between the TATC insertion/deletion polymorphism and UL risk. However, increased UL risk was identified to be significantly associated with CAA insertion/deletion polymorphism in the recessive and codominant model. The present study provided evidence for the first time that CAA polymorphism in RTN4 3’-UTR, but not TATC polymorphism may be involved in susceptibility to UL. C1 [Zhang, Kui] Sichuan University, West China School of Preclinical and Forensic Medicine, Department of Forensic Biology, 610041 Chengdu, Sichuan, China. [Bai, Peng] Sichuan University, West China School of Preclinical and Forensic Medicine, Department of Forensic Biology, 610041 Chengdu, Sichuan, China. [Shi, Shaoqing] Sichuan University, West China School of Preclinical and Forensic Medicine, Department of Immunology, 610041 Chengdu, Sichuan, China. [Zhou, Bin] West China Second University Hospital, Sichuan University, West China Institute of Women and Children’s Health, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Laboratory of Molecular Translational Medicine, 610041 Chengdu, Sichuan, China. [Wang, Yanyun] West China Second University Hospital, Sichuan University, West China Institute of Women and Children’s Health, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Laboratory of Molecular Translational Medicine, 610041 Chengdu, Sichuan, China. [Song, Yaping] West China Second University Hospital, Sichuan University, West China Institute of Women and Children’s Health, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Laboratory of Molecular Translational Medicine, 610041 Chengdu, Sichuan, China. [Rao, Li] West China Hospital of Sichuan University, Department of Cardiology, 610041 Chengdu, Sichuan, China. [Zhang, Lin] Sichuan University, West China School of Preclinical and Forensic Medicine, Department of Forensic Biology, 610041 Chengdu, Sichuan, China. RP Zhang, L (reprint author), Sichuan University, West China School of Preclinical and Forensic Medicine, Department of Forensic Biology, 610041 Chengdu, China. EM zhanglin@scu.edu.cn CR Duhan N, 2011, Current and emerging treatments for uterine myoma - an update. Int J Womens Health 3:231–241 Hart RKY, Yeong CT, Seed P, Taylor A, Braude P, 2001, A prospective controlled study of the effect of intramural uterine fibroids on the outcome of assisted conception. Hum Reprod 16:2411–2417 Ptacek TSC, Walker CL, Sell SM, 2007, Physical mapping of distinct 7q22 deletions in uterine leiomyoma and analysis of a recently annotated 7q22 candidate gene. Cancer Genet Cytogenet 174:116–120 Morgan Ortiz FPRB, Elorriaga Garcia E, Baez Barraza J, Quevedo Castro E, Peraza Garay Fde J, 2011, Uterine leiomyomas during pregnancy and its impact on obstetric outcome. Ginecol Obstet Mex 79(8):467–473 Lee EJ, Kong G, Lee SH, Rho SB, Park CS, Kim BG, Bae DS, Kavanagh JJ, Lee JH, 2005, Profiling of differentially expressed genes in human uterine leiomyomas. Int J Gynecol Cancer 15, 1):146–154., DOI 10.1111/j.1048-891x.2005.15016.x Flynn MJM, Datta S, Myers E, 2006, Health care resource use for uterine fibroid tumors in the United States. Am J Obstet Gynecol 195(4):955–964 Luoto RKJ, Rutanen EM, Taipale P, Perola M, Koskenvuo M, 2000, Heritability and risk factors of uterine fibroids–the Finnish Twin Cohort study. Maturitas 37(1):15–26 Chen Y, Tang X, Cao X, Chen H, Zhang X, 2006, Human Nogo-C overexpression induces HEK293 cell apoptosis via a mechanism that involves JNK-c-Jun pathway. Biochem Biophys Res Commun 348(3):923–928., DOI 10.1016/j.bbrc.2006.07.166 Shimakage MIN, Ohshima K, Kawahara K, Oka T, Yasui K, Matsumoto K, Inoue H, Watari A, Higashiyama S, Yutsudo M, 2006, Down-regulation of ASY/Nogo transcription associated with progression of adult T-cell leukemia/lymphoma. Int J Cancer 119(7):1648–1653 Jung TYJS, Lee KH, Cao VT, Jin SG, Moon KS, Kim IY, Kang SS, Kim HS, Lee MC, 2011, Nogo-A expression in oligodendroglial tumors. Neuropathology 31(1):11–19 Yicun Chen XT, Zhang X, Zhuang L, 2009, New mutations of Nogo-C in hepatocellular carcinoma. Mol Biol Rep 36:377–380 Novak G, Tallerico T, 2006, Nogo A, B and C expression in schizophrenia, depression and bipolar frontal cortex, and correlation of Nogo expression with CAA/TATC polymorphism in 3’-UTR. Brain Res 1120(1):161–171., DOI 10.1016/j.brainres.2006.08.071 Novak G, Kim D, Seeman P, Tallerico T, 2002, Schizophrenia and Nogo: elevated mRNA in cortex, and high prevalence of a homozygous CAA insert. Brain Res Mol Brain Res 107(2):183–189 Zhou B, Rao L, Li Y, Gao L, Li C, Chen Y, Xue H, Liang W, Lv M, Song Y, Peng Y, Zhang L, 2009, The association between dilated cardiomyopathy and RTN4 3’UTR insertion/deletion polymorphisms. Clin Chim Acta 400(1–2):21–24., DOI 10.1016/j.cca.2008.09.028 Chen Y, Zhou B, Li H, Peng Y, Wang Y, Rao L, 2011, Analysis of RTN4 3’UTR insertion/deletion polymorphisms in ventricular septal defect in a Chinese Han population. DNA Cell Biol 30(5):323– 327., DOI 10.1089/dna.2010.1116 Shi S, Zhou B, Wang Y, Chen Y, Zhang K, Wang K, Quan Y, Song Y, Rao L, Zhang L, 2012, Genetic variation in RTN4 3’-UTR and susceptibility to cervical squamous cell carcinoma. DNA Cell Biol 31(6):1088–1094., DOI 10.1089/dna.2011.1548 Sole X, Guino E, Valls J, Iniesta R, Moreno V, 2006, SNPStats: a web tool for the analysis of association studies. Bioinformatics 22:1928–1929 Wei TGA, Qian HR, Su C, Helvering LM, Kulkarini NH, Shou J, N'Cho M, Bryant HU, Onyia JE, 2007, DNA microarray data integration by ortholog gene analysis reveals potential molecular mechanisms of estrogen-dependent growth of human uterine fibroids. BMC Womens Health 7:5 Hiroshi Ishikawa SR, Demura M, Rademaker AW, Kasai T, Inoue M, Usui H, Shozu M, Bulun SE, 2009, High aromatase expression in uterine leiomyoma tissues of African-American women. J Clin Endocrinol Metab 94:1752–1756 Ishwad CSFR, Hanley K, Davare J, Meloni AM, Sandberg AA, Surti U, 1997, Two discrete regions of deletion at 7q in uterine leiomyomas. Genes Chromosomes Cancer 19:156–160 Xing YPPW, Morton CC, 1997, The del(7q, subgroup in uterine leiomyomata: genetic and biologic characteristics. Further evidence for the secondary nature of cytogenetic abnormalities in the pathobiology of uterine leiomyomata. Cancer Genet Cytogenet 98(69–74):69 Meloni AMSU, Sandberg AA, 1991, Deletion of chromosome 13 in leiomyomas of the uterus. Cancer Genet Cytogenet 53:199– 203 Jennelle C, Hodge KTC, Huyck KL, Somasundaram P, Panhuysen CIM, Stewart EA, Morton CC, 2009, Uterine leiomyomata and decreased height: a common HMGA2 predisposition allele. Hum Genet 125(3):257–263 Bowden WSJ, Kovanci E, Rajkovic A, 2009, Detection of novel copy number variants in uterine leiomyomas using high-resolution SNP arrays. Mol Hum Reprod 15(9):563–568 Yaqin Chen SZ, Xiang R, 2010, RTN3 and RTN4: candidate modulators in vascular cell apoptosis and atherosclerosis. J Cell Biochem 111:797–800 Oertle T, Huber C, van der Putten H, Schwab ME, 2003, Genomic structure and functional characterisation of the promoters of human and mouse nogo/rtn4. J Mol Biol 325(2):299–323 Yang J, Yu L, Bi AD, Zhao SY, 2000, Assignment of the human reticulon 4 gene, RTN4, to chromosome 2p14–2p13 by radiation hybrid mapping. Cytogenet Cell Genet 88(1–2):101–102 GrandPre T, Nakamura F, Vartanian T, Strittmatter SM, 2000, Identification of the Nogo inhibitor of axon regeneration as a Reticulon protein. Nature 403(6768):439–444., DOI 10.1038/ 35000226 Chen YC, Lu DD, Cao XR, Zhang XR, 2005, RTN4-C gene expression in hepatocellular carcinoma and its influence on SMMC7721 cell growth and apoptosis. Yi Chuan Xue Bao 32, 9):891–897 Zheng H, Xue S, Lian F, Wang YY, 2011, A novel promising therapy for vein graft restenosis: overexpressed Nogo-B induces vascular smooth muscle cell apoptosis by activation of the JNK/ p38 MAPK signaling pathway. Med Hypotheses 77(2):278–281., DOI 10.1016/j.mehy.2011.04.035 Watari A, Yutsudo M, 2003, Multi-functional gene ASY/Nogo/ RTN-X/RTN4: apoptosis, tumor suppression, and inhibition of neuronal regeneration. Apoptosis 8(1):5–9 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 475 EP 479 DI 10.1007/s12253-013-9604-6 PG 5 ER PT J AU Le Donne, M Giuffre, G Caruso, C Nicotina, AP Alibrandi, A Scalisi, R Simone, A Chiofalo, B Triolo, O AF Le Donne, Maria Giuffre, Giuseppe Caruso, Carmela Nicotina, Antonio Piero Alibrandi, Angela Scalisi, Rosalba Simone, Angela Chiofalo, Benito Triolo, Onofrio TI Human Papillomavirus Types Distribution in Eastern Sicilian Females with cervical lesions. A Correlation with Colposcopic and Histological Findings SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human papillomavirus; Genotypes; Distribution; Colposcopy; Cervical lesion ID Human papillomavirus; Genotypes; Distribution; Colposcopy; Cervical lesion AB To determine human papillomavirus (HPV) types distribution in cervical lesions in a Southern Italian female population in Messina and their relationship between HPV type and grade of colposcopic and histopathological abnormality, a total of 253 women aged 17–68 years, with previous cytological abnormalities, were included in this study. HPVDNA testing, colposcopy and biopsy were performed. For each sample, cervical cells were collected by centrifugation and DNA was extracted, followed by a PCR-based HPV-DNA assay and reverse dot blot genotyping. HPV-16 was found the most common type (46.6%) followed by HPV-31 (26.9%), −6 (18.6 %), −58 (8.8 %), −18 (6.7 %), −66 (5.7 %), −52 and −53 (4.7 %). Out of 62 women with abnormal transformation zone (ATZ) area compatible with squamous intraepithelial lesion (SIL) or cervical cancer (CC), 64.5 % was found high risk (HR) HPV-positive. Moreover the severity of the colposcopic diagnosis was positively correlated with the higher HPV oncogenicity risk (HPV-16 P=0.023; and HPV-53 P=0.047). The HPV-16 was found the most prevalent type within each histological category: 66.7%, 31.2%, 44%and 37.2%of CC, high grade (H)SIL, low grade (L)SIL and chronic cervicitis respectively; followed by HPV-31 present in 25 %, 8 %, and 13.3 % of HSIL, LSIL and chronic cervicitis respectively. A higher HPV incidence than the rest of Italy was found, in agreement with that detected by other authors for the South of the country. These data provide further information about the types prevalence in women with cervical lesions living in Eastern Sicily, suggesting the introduction of new targeted vaccines against a wider spectrum of HPV. C1 [Le Donne, Maria] Azienda Ospedaliera Universitaria, ‘Policlinico G. Martino’, Dipartimento di Scienze Pediatriche, Ginecologiche, Microbiologiche e Biomediche, Via Consolare Valeria, 98125 Messina, Italy. [Giuffre, Giuseppe] Azienda Ospedaliera Universitaria, Department of Human Pathology, Via Consolare Valeria, 98125 Messina, Italy. [Caruso, Carmela] Azienda Ospedaliera Universitaria, ‘Policlinico G. Martino’, Dipartimento di Scienze Pediatriche, Ginecologiche, Microbiologiche e Biomediche, Via Consolare Valeria, 98125 Messina, Italy. [Nicotina, Antonio Piero] Azienda Ospedaliera Universitaria, Department of Human Pathology, Via Consolare Valeria, 98125 Messina, Italy. [Alibrandi, Angela] University of Messina, Department of Economical, Business and Environmental Sciences and Quantitative Methods, Via dei Verdi n.75, 98124 Messina, Italy. [Scalisi, Rosalba] Azienda Ospedaliera Universitaria, ‘Policlinico G. Martino’, Dipartimento di Scienze Pediatriche, Ginecologiche, Microbiologiche e Biomediche, Via Consolare Valeria, 98125 Messina, Italy. [Simone, Angela] Azienda Ospedaliera Universitaria, Department of Human Pathology, Via Consolare Valeria, 98125 Messina, Italy. [Chiofalo, Benito] Azienda Ospedaliera Universitaria, ‘Policlinico G. Martino’, Dipartimento di Scienze Pediatriche, Ginecologiche, Microbiologiche e Biomediche, Via Consolare Valeria, 98125 Messina, Italy. [Triolo, Onofrio] Azienda Ospedaliera Universitaria, ‘Policlinico G. Martino’, Dipartimento di Scienze Pediatriche, Ginecologiche, Microbiologiche e Biomediche, Via Consolare Valeria, 98125 Messina, Italy. RP Le Donne, M (reprint author), Azienda Ospedaliera Universitaria, ‘Policlinico G. Martino’, Dipartimento di Scienze Pediatriche, Ginecologiche, Microbiologiche e Biomediche, 98125 Messina, Italy. EM marialedonne@tin.it CR Giorgi Rossi P, Bisanzi S, Paganini I et al, 2010, Prevalence of HPV high and low risk types in cervical samples from the Italian general population: a population based study. BMC Infect Dis 20(10):214 Agarossi A, Ferrazzi E, Parazzini F, Perno CF, Ghisoni L, 2009, Prevalence and type distribution of high-risk human papillomavirus infection in women undergoing voluntary cervical cancer screening in Italy. J Med Virol 81:529–535 Ammatuna P, Giovannelli L, Matranga D, Ciriminna S, Perino A, 2008, Prevalence of genital human papilloma virus infection and genotypes among young women in Sicily, south Italy. Cancer Epidemiol Biomarkers Prev 17:2002–2006 Del Prete R, Di Taranto AM, Lipsi MR, Nirchio V, Antonetti R, Miragliotta G, 2008, Prevalence and genotypes identification of human papillomavirus infection in a population of south Italy. J Clin Virol 42:211–214 Capra G, Giovannelli L, Bellavia C et al, 2008, HPV genotype prevalence in cytologically abnormal cervical samples from women living in south Italy. Virus Res 133:195–200 Gargiulo F, De Francesco MA, Schreiber C et al, 2007, Prevalence and distribution of single and multiple HPV infections in cytologically abnormal cervical samples from Italian women.Virus Res 125:176–182 Sandri MT, Riggio D, Salvatici M et al, 2009, Typing of human papillomavirus in women with cervical lesions: prevalence and distribution of different genotypes. J Med Virol 81:271–277 Sideri M, Cristoforoni P, Casadio C et al, 2009, Distribution of human papillomavirus genotypes in invasive cervical cancer in Italy: a representative, single institution case series. Vaccine 27(Suppl 1):A30–A33 Dal Bello B, Spinillo A, Alberizzi P, Cesari S, Gardella B, Silini EM, 2009, Time trends of human papillomavirus type distribution in Italian women with cervical intraepithelial neoplasia, CIN). Gynecol Oncol 115:262–266 Broccolo F, Chiari S, Piana A et al, 2009, Prevalence and viral load of oncogenic human papillomavirus types associated with cervical carcinoma in a population of North Italy. J Med Virol 81:278–287 Carozzi FM, Tornesello ML, Burroni E et al, 2010, Prevalence of human papilloma types in high-grade cervical intraepithelian neoplasia and cancer in Italy. Cancer Epidemiol biomarkers Prev 19:2389–2400 Giorgi Rossi P, Chini F, Bisanzi S et al, 2011, Distribution of high and low risk HPV types by cytological status: a population based study from Italy. Infect Agent Cancer 20(6):2 Tornesello ML, Duraturo ML, Botti G et al, 2006, Italian HPV Working Group: Prevalence of alpha-papillomavirus genotypes in cervical squamous intraepithelial lesions and invasive cervical carcinoma in the italian population. J Med Virol 78:1663– 1672 Kay P, Meehan K, Williamson AL, 2002, The use of nested polymerase chain reaction and restriction fragment length polymorphism for the detection and typing of mucosal human papillomaviruses in samples containing low copy numbers of viral DNA. J Virol Methods 105:159–170 Qu W, Jiang G, Cruz Y et al, 1997, PCR detection of human papillomavirus: comparison between MY09/MY11 and GP5+/ GP6+ primer systems. J Clin Microbiol 35:1304–1310 Giovannelli L, Lama A, Capra G, Giordano V, Arico P, Ammatuna P, 2004, Detection of human papillomavirus DNA in cervical samples: analysis of the new PGMY-PCR compared to the Hybrid Capture II and MY-PCR assays and a two-step nested PCR assay. J Clin Microbiol 42:3861–3864 Giuffre G, Simone A, Todaro P et al, 2010, Detection of genotyping of human papillomavirus in gynaecologic outpatients of Messina, eastern Sicily, Italy. Oncol Rep 23:745–750 Agodi A, Barchitta M, La Rosa N et al, 2009, Human papillomavirus infection:low-risk and high-risk genotypes in women in Catania, Sicily. Int J Gynecol Cancer 19:1094–1098 Arbyn M, Benoj I, Simoens C, Bogers J, Beutels P, Depuydt C, 2009, Prevaccination distribution of human papilloma types in women attending at cervical cancer screening in Belgium. Cancer Epidemiol Biomarkers Prev 18:321–330 Clifford GM, Rashida K, Rana RK et al, 2005, Human papillomavirus genotype distribution in low-grade cervical lesions: comparison by geographic region and with cervical cancer. Cancer Epidemiol Biomarkers Prev 14:1157–1164 De Francesco MA, Gargiulo F, Schreiber C, Ciravolo G, Salinaio F, Manca N, 2005, Detection and genotyping of human papillomavirus in cervical samples from Italian patients. J Med Virol 75:588–592 Massad LS, Jeronimo J, Katki HA, Schiffman M, 2009, The accuracy of colposcopic grading for detection of high grade cervical intraepithelial neoplasia. Low Genit Tract Dis 13:137–144 Sideri M, Spolti N, Spinaci L et al, 2004, Interobserver variability of colposcopic interpretations and consistency with final histologic results. J Lower Genital Tract Dis 8:212–216 Atkins KA, Jeronimo J, Stoler MH, for the ALTS Group, 2006, Description of patients with squamous cell carcinoma in the atypical squamous cells of undetermined significance/low-grade squamous intraepithelial lesion triage study. Cancer, Cancer Cytopathol, 108:212–221 Milne DS, Wadehra V, Mennim D, Wagstaff TI, 1999, A prospective follow-up study of women with colposcopically unconfirmed positive cervical smears. Br J Obstet Gynaecol 106:38–41 Zuna RE, Allen RA, Moore WE, Lu Y, Mattu R, Dunn ST, 2007, Distribution of HPV genotypes in 282 women with cervical lesions: evidence for three categories of intraepithelial lesions based on morphology and HPV type. Mod Pathol 20:167–174 Lonky NM, Felix JC, Yathi MN, Wolde-Tsadik G, 2003, Triage of atypical squamous cells of undetermined significance with hybrid capture. II. Colposcopy and histologic human papillomavirus correlation. Obstet Gynecol 101:481–489 Guyot A, Fox J, Karim S, Kyi M, 2004, Possible causes of lowsensitivity of HPV-DNA testing for CIN2/3. Acta Obstet Gynecol Scand 83:217–218 Jastania R, Geddie WR, Chapman W, Boerner S, 2006, Characteristics of apparently false-negative digene hybrid capture 2 high risk HPV DNA testing. Am J Clin Pathol 125:223–228 Castle PE, Jeronimo J, Schiffman M et al, 2006, Age-related changes of the cervix influence human papillomavirus type distribution. Cancer Res 66:1218–1224 Zuna RE, Wang SS, Rosenthal DL, Jeronimo J, Schiffman M, Solomon D, 2005, Determinants of human papillomavirusnegative, low-grade squamous intraepithelial lesions in the atypical squamous cells of undetermined significance/low-grade squamous intraepithelial lesions Triage Study, ALTS). Cancer 105:253–262 Evans MF, Adamson CS-C, Papillo JL, St. John TL, Leiman G, Cooper K, 2006, Distribution of human papillomavirus types in ThinPrep Papanicolaou tests classified according to the Bethesda terminology and correlations with patient age and biopsy outcomes. Cancer 106:1054–1064 Caruso C, Le Donne M, Antico F et al, 2005, Colposcopy vs hybrid capture II assay in detection of cervical human papilloma virus infection. Eur J Gynaecol Oncol 26:303–305 Cobo F, Concha A, Ortiz M, 2009, Human papillomavirus, HPV, type distribution in females with abnormal cervical cytology. A correlation with histological study. Open Virol J 3:60–66 Ibanez R, Moreno-Crespi J, Sarda M et al, 2012, Prediction of cervical intraepithelial neoplasia grade 2+, CIN2+, using HPV DNA testing after a diagnosis of atypical squamous cells of undetermined significance, ASC-US, in Catalonia, Spain. BMC Infect Dis 12:25 Dane C, Batmaz G, Dane B, Cetin A, 2009, Screening properties of human papillomavirus testing for predicting cervical intraepithelial neoplasia in atypical squamous cells of undetermined significance and low-grade squamous intraepithelial lesion smears: a prospective study. Ann Diagn Pathol 13:73–77 Dexeus S, Cararach M, Dexeus D, 2002, The role of colposcopy in modern gynecology. Eur J Gynaecol Oncol 23:269–277 Ronco G, Anttila A, 2011, The best management of HPV positive women. EUROGIN 2011—HPV associated diseases and cancer— from reality now to the future Lisbon, Portugal—May 8– 11, CS 2–3 Ronco G, Biggeri A, Confortini M et al, 2012, Health technology assessment report: HPV Dna based primary screening for cervical cancer precursors. Epidemiol Prev 36(3–4 Suppl 1):1–72 Schiffman M, Wentzensen N, Wacholder S, Kinney W, Gage JC, Castle PE, 2011, Human papillomavirus testing in the prevention of cervical cancer. J Natl Cancer Inst 103:368–383 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 481 EP 487 DI 10.1007/s12253-013-9605-5 PG 7 ER PT J AU Liu, Ch Cui, Q Shu, C Guo, J Li, D AF Liu, Cheng Cui, Qiu Shu, Cuili Guo, Jun Li, Dingfeng TI Comprehensive Treatment Based on Intra-arterial Chemotherapy for Distal Femur Neoplasms SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intra-arterial chemotherapy; SIDS; Distal femur; Bone neoplasms ID Intra-arterial chemotherapy; SIDS; Distal femur; Bone neoplasms AB To investigate the clinical efficacy of intra-arterial chemotherapy by subcutaneous implantable delivery system (SIDS) in the treatment of distal femur neoplasm. From March 2002 to December 2009, 51 patients were treated with SIDS intra-femoral artery chemotherapy, followed by customized prosthetic reconstruction, including 45 patients of osteosarcoma and 6 malignant fibrous histiocytoma in distal femur. The average follow-up period was 64 months (ranging between 24 and 116 months) to track on the efficacy of chemotherapy, which shows that 96.1 % of patients got pain relief, 70.6 % of patients had significant radiological change, and 82.4 % of paitents with medium to severe pathological variation responded to the chemotherapy. Local recurrence happened in 4 cases, 3 cases are alive with disease, 4 cases died because of pulmonary metastases and other 40 patients are free of local recurrence or distant metastasis. SIDS intra-femur artery chemotherapy can improve clinical outcome of DFN, and provide effective method for treatment of DFN when combined with customized prosthetic reconstruction. C1 [Liu, Cheng] 307 Hospital of PLA, Department of Orthopedics, NO 8 East Street, Fengtai District, 100071 Beijing, China. [Cui, Qiu] 307 Hospital of PLA, Department of Orthopedics, NO 8 East Street, Fengtai District, 100071 Beijing, China. [Shu, Cuili] 307 Hospital of PLA, Department of Orthopedics, NO 8 East Street, Fengtai District, 100071 Beijing, China. [Guo, Jun] 307 Hospital of PLA, Department of Orthopedics, NO 8 East Street, Fengtai District, 100071 Beijing, China. [Li, Dingfeng] 307 Hospital of PLA, Department of Orthopedics, NO 8 East Street, Fengtai District, 100071 Beijing, China. RP Li, D (reprint author), 307 Hospital of PLA, Department of Orthopedics, 100071 Beijing, China. EM 307yygk@sina.com CR Wilkins RM, Cullen JW, Camozzi AB et al, 2005, Improved survival in primary nonmetastatic pediatric osteosarcoma of the extremity. Clin Orthop Relat Res 438:128–136 Zhang HJ, Yang JJ, Lu JP et al, 2009, Use of intra-arterial chemotherapy and embolization before limb salvage surgery for osteosarcoma of the lower extremity. Cardiovasc Intervent Radiol 32, 4):672–678 Abe S, Tateishi A, Ogawa K et al, 2001, Long-term local intensive preoperative chemotherapy and joint-preserving conservative surgery for osteosarcoma around the knee. Orthopedics 24(7):671–676 Wilkins RM, Cullen JW, Odom L et al, 2003, Superior survival in treatment of primary nonmetastatic pediatric osteosarcoma of the extremity. Ann Surg Oncol 10(5):498–507 Cullen JW, Jamroz BA, Stevens SL et al, 2005, The value of serial arteriography in osteosarcoma: delivery of chemotherapy, determination of therapy duration, and prediction of necrosis. J Vasc Interv Radiol 16(8):1107–1119 Dingfeng L, Qiu C, Haitao F, 2005, Development of adjustable prosthesis and its preliminary application in limb salvage treatment of osteosarcoma in the lower segment of femur. Chin J Reparative Reconstr Surg 19(7):534–536 Li DF, Cui Q, Wang Q, Su H et al, 2006, Application of I125 brachytherapy combined artificial joint prosthesis in malignant osteoand soft-tissue sarcoma. Cancer Biother Radiopharm 21(2):167–172 Cui Q, Li DF, Liu C et al, 2011, Two case-reports of the limb salvage treatment of osteosarcoma consolidated with obvious pathological fractures. Pathol Oncol Res 17:973–979 Wu CC, Pritsch T, Shehadeh A et al, 2008, The anterior popliteal approach for popliteal exploration, distal femoral resection, and endoprosthetic reconstruction. J Arthroplasty 23(2):254–262 Hugate RR, Wilkins RM, Kelly CM et al, 2008, Intraarterial chemotherapy for extremity osteosarcoma and MFH in adults. Clin Orthop Relat Res 466(6):1292–1301 Gelderblom H, Jinks RC, Sydes M et al, 2011, Survival after recurrent osteosarcoma: data from 3 European Osteosarcoma Intergroup, EOI, randomized controlled trials. Eur J Cancer 47(6):895–902 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 489 EP 493 DI 10.1007/s12253-013-9606-4 PG 5 ER PT J AU Giordano, G Pizzi, S Azzoni, C Bottarelli, L D’Adda, T AF Giordano, Giovanna Pizzi, Silvia Azzoni, Cinzia Bottarelli, Lorena D’Adda, Tiziana TI Primary Squamous Cell Carcinoma of the Endometrium Unrelated to Human Papilloma Virus: A Molecular Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial squamous carcinoma; Allelic imbalance; Aberrant methylation of promoters ID Endometrial squamous carcinoma; Allelic imbalance; Aberrant methylation of promoters AB In this paper we report a molecular study of a case of Primary Endometrial Squamous Carcinoma (PESC), in which a Human Papilloma Virus (HPV) infection had been previously excluded by Polymerase Chain Reaction (PCR). The studies performed in an effort to explain the carcinogenesis included immunohistochemical over-expression of p53 and p16 proteins as previously observed in our own papers, plus microsatellite analysis of D10S1765 at 10q23.3 (PTEN) and TP53 at 17p13.1 (P53) as well as the methylation status of the of BRCA1 and p16 promoters using specific PCRs. In this rare malignancy, we found allelic imbalance (AI) at 17p13.1 (P53). Instead, AI at D10S1765 (PTEN) gene was absent. The genetic alteration of p53, with hyper-expression of p53 protein and an absence of abnormalities in the PTEN gene are consistent with the similarities between Uterine Serous Carcinoma (USC) and our case of PESC. The aberrant methylation of both p16 and BCAR1 promoters was not detected in our case. This finding too could imply that ESC is more similar to Uterine Serous Carcinoma than Uterine Endometrioid Carcinoma (UEC). Moreover, the lack of aberrant methylation of p16, which is in accordance with over-expression of p16 immunoreactivity, in the absence of HPV infection may be related to other unknown genetic alterations. In our opinion, it is hard to reach any definite conclusion concerning the carcinogenesis of PESC, because of its rarity and the very few molecular studies reported in the literature. Further studies with more numerous cases and larger molecular analyses are mandatory for this malignancy, to confirm whether it is more closely related to papillary endometrial cancer than to endometrioid carcinoma. C1 [Giordano, Giovanna] University of Parma, Institute of Pathology, via Antonio Gramsci, 14 43126 Parma, Italy. [Pizzi, Silvia] University of Parma, Institute of Pathology, via Antonio Gramsci, 14 43126 Parma, Italy. [Azzoni, Cinzia] University of Parma, Institute of Pathology, via Antonio Gramsci, 14 43126 Parma, Italy. [Bottarelli, Lorena] University of Parma, Institute of Pathology, via Antonio Gramsci, 14 43126 Parma, Italy. [D’Adda, Tiziana] University of Parma, Institute of Pathology, via Antonio Gramsci, 14 43126 Parma, Italy. RP Giordano, G (reprint author), University of Parma, Institute of Pathology, 14 43126 Parma, Italy. EM giovanna.giordano@unipr.it CR Klaes R, Friedrich T, Spitkovsky D et al, 2001, Overexpression of p16ink4a as a specific marker for dysplastic and neoplastic epithelial cells of the cervix uteri. Int J Cancer 92:276–284 Kataoka A, Nishida T, Sugiyama T, Hori K, Honda S, Yakushiji M, 1997, Squamous cell carcinoma of the endometrium with human papillomavirus type 31 and without tumor suppressor gene p53 mutation. Gynecol Oncol 65:180–184 Pins MR, Young RH, Crum CP, Leach IH, Scully RE, 1997, Cervical squamous cell carcinoma in situ with intraepithelial extension to the upper genital tract and invasion of tubes and ovaries: report of a case with human papilloma virus analysis. Int J Gynecol Pathol 16:272–278 Wang Z, Dong J, Eyzaguirre EJ, Tang WW, Eltorky MA, Qiu S, 2009, Detection of human papilloma virus subtypes 16 and P16(ink4a, in invasive squamous cell carcinoma of the fallopian tube and concomitant squamous cell carcinoma in situ of the cervix. J Obstet Gynaecol Res 35:385–389 Bifulco G, Giampaolino P, Mandato VD, Morra I, Nappi C, Insabato L, 2011, Primary squamous cell carcinoma of the endometrium: a case report. Eur J Gynaecol Oncol 32:350–352 Goodrich S, Kebria-Moslemi M, Broshears J, Sutton GP, Rose P. Primary squamous cell carcinoma of the endometrium: two cases and a review of the literature. Diagn Cytopathol. 2012 Jan 12., DOI 10.1002/dc.22814. [Epub ahead of print] Giordano G, D’Adda T, Merisio C, Gnetti L, 2005, Primary squamous carcinoma of the endometrium: a case report with immunohistochemical and molecular study. Gynecol Oncol 96:876– 879 Giordano G, Azzoni C, D’Adda T, Merisio C, 2007, P16INK4a overexpression independent of Human Papilloma Virus, HPV, infection in rare subtypes of endometrial carcinomas. Pathol Res Pract 7:533–538 D’Adda T, Bottarelli L, Azzoni C et al, 2005, Malignancyassociated X chromosome allelic losses in foregut endocrine neoplasms: further evidence from lung tumors. Mod Pathol 18:795– 805 Naidoo R, Chetty R, 1998, The application of microsatellites in molecular pathology. Pathol Oncol Res 4:310–315 Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB, 1996, Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 93:9821–9826 Esteller M, 2008, Epigenetics in cancer. N Engl J Med 358:1148– 1159 Weissenbach J, Gyapay G, Dib C et al, 1992, A second-generation linkage map of the human genome. Nature 359:794–801 Peltomaki P, Lothe RA, Aaltonen LA et al, 1993, Microsatellite instability is associated with tumors that characterize the hereditary non-polyposis colorectal carcinoma syndrome. Cancer Res 53:5853– 5855 Ionov Y, Peinado MA, Malkhosyan S, Shibata D, Perucho M, 1993, Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature 363:558–561 Wirtz HC, Muller W, Noguchi T et al, 1998, Prognostic value and clinicopathological profile of microsatellite instability in gastric cancer. Clin Cancer Res 4:1749–1754 Duggan BD, Felix JC, Muderspach LI, Tourgeman D, Zheng J, Shibata D, 1994, Microsatellite instability in sporadic endometrial carcinoma. J Natl Cancer Inst 86:1216–1221 Steele RJC, Thompson AM, Hall PA, Lane DP, 1998, The p53 tumour suppressor gene. Br J Surg 85:1460–1467 Ziyaie D, Hupp TR, Thompson AM, 2000, p53 and breast cancer. Breast 9:239–246 Rogel A, Popliker M, Webb CG, Oren M, 1985, p53 cellular tumor antigen: analysis of mRNA levels in normal adult tissues, embryos, and tumors. Mol Cell Biol 5:2851–2855 Finlay CA, Hinds PW, Tan TH, Eliyahu D, Oren M, Levine AJ, 1988, Activating mutations for transformation by p53 produce a gene product that forms an hsc70-p53 complex with an altered half-life. Mol Cell Biol 8:531–539 Lakshmi S, Nair MB, Jayaprakash PG, Rajalekshmy TN, Nair MK, Pillai MR, 1997, p53 protein and tumorigenesis in the uterine cervix. Gen Diagn Pathol 142:281–287 Reihsaus E, Kohler M, Kraiss S et al, 1990, Regulation of the level of the oncoprotein p53 in non transformed and transformed cells. Oncogene 5:137–145 Munn KE, Walker RA, Menasce L, Varley JM, 1996, Mutation of the TP53 gene and allelic imbalance at chromosome 17p13 in ductal carcinoma in situ. Br J Cancer 74:1578–1585 Steck PA, Perhouse MA, Jasser SA et al, 1997, Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet 15:356–362 Li J, Yen C, Liaw D et al, 1997, PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 275:1943–1946 Bussaglia E, del Rio E, Matias-Guiu X, Prat J, 2000, PTEN mutations in endometrial carcinomas: a molecular and clinicopathologic analysis of 38 cases. Hum Pathol 31:312–317 Levine RL, Cargile CB, Blazes MS, van Rees B, Kurman RJ, Ellenson LH, 1998, PTEN mutations and microsatellite instability in complex atypical hyperplasia, a precursor lesion to uterine endometrioid carcinoma. Cancer Res 58:3254–3258 Maxwell GL, Risinger JI, Gumbs C et al, 1998, Mutation of the PTEN tumor suppressor gene in endometrial hyperplasias. Cancer Res 58:2500–2503 Mutter GL, 2001, PTEN, a protean tumor suppressor. Am J Pathol 158:1895–1898 Omura-Minamisawa M, Diccianni MB, Chang RC et al, 2001, p16/p14(ARF, cell cycle regulatory pathways in primary neuroblastoma: p16 expression is associated with advanced stage disease. Clin Cancer Res 7:3481–3490 Esteller M, Silva JM, Dominguez G et al, 2000, Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst 92:564–569 Wong YF, Chung TK, Cheung TH et al, 1999, Methylation of p16INK4A in primary gynecologic malignancy. Cancer Lett 136:231–235 Miki Y, Swensen J, Shattuck-Eidens D et al, 1994, A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 266:66–71 Venkitaraman AR, 2002, Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell 108:171–182 Kerr P, Ashworth A, 2001, New complexities for BRCA1 and BRCA2. Curr Biol 11:R668–R676 Shen S-X, Weaver Z, Xu X et al, 1998, A targeted disruption of the murine BRCA1 gene causes gamma-irradiation hypersensitivity and genetic instability. Oncogene 17:3115–3124 Foray N, Randrianarison V, Mrot D, Perricaudet M, Lenoir G, Feunteun J, 1999, Gamma-rays-induced death of human cells carrying mutations of BRCA1 or BRCA2. Oncogene 18:7334– 7342 Xu X, Wagner KU, Larson D et al, 1999, Conditional mutation of BRCA1 in mammary epithelial cells results in blunted ductal morphogenesis and tumour formation. Nat Genet 22:37–43 Bianco T, Chenevix-Trench G, Walsh DC, Cooper JE, Dobrovic A, 2000, Tumour-specific distribution of BRCA1 promoter region methylation supports a pathogenetic role in breast and ovarian cancer. Carcinogenesis 21:147–151 Baldwin RL, Nemeth E, Tran H et al, 2000, BRCA1 promoter region hypermethylation in ovarian carcinoma: a population-based study. Cancer Res 60:5329–5333 Risinger JI, Hayes AK, Berchuck A, Barrett JC, 1997, PTEN/ MMAC1 mutations in endometrial cancers. Cancer Res 57:4736– 4738 Seeber LM, Zweemer RP, Marchionni L et al, 2010, Methylation profiles of endometrioid and serous endometrial cancers. Endocr Relat Cancer 17:663–673 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 495 EP 499 DI 10.1007/s12253-013-9607-3 PG 5 ER PT J AU Dede, K Mersich, T Besznyak, I Zarand, A Salamon, F Baranyai, Zs Landherr, L Jakab, F Bursics, A AF Dede, Kristof Mersich, Tamas Besznyak, Istvan Zarand, Attila Salamon, Ferenc Baranyai, Zsolt Landherr, Laszlo Jakab, Ferenc Bursics, Attila TI Bevacizumab Treatment Before Resection of Colorectal Liver Metastases: Safety, Recovery of Liver Function, Pathologic Assesment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal liver metastases; Preoperative chemotherapy; Bevacizumab; Complications; Functional liver recovery ID Colorectal liver metastases; Preoperative chemotherapy; Bevacizumab; Complications; Functional liver recovery AB Patients with metastatic colorectal cancer receive chemotherapy prior liver resection more and more frequently. This preoperative treatment has many effects which have to be analysed, like the safety of liver resection, toxicity, tissue regeneration, radiological and pathological response and survival data. The aim of the study was to evaluate the safety of bevacizumab containing preoperative chemotherapy and functional recovery of the liver after resection for colorectal liver metastases (CLM) and to analyse radiological and pathological data. Data of three groups of 120 consecutive patients —(1) CTX+BV: cytotoxic chemotherapy + bevacizumab, (2) CTX: cytotoxic chemotherapy, (3) NC: no treatment before liver resection—were analysed. Postoperative liver function and complications were compared, clinical, radiological and pathological data were evaluated. Between 01.12.2006 and 31.12.2010 41 resections was performed after chemotherapy + bevacizumab (CTX+BV) and 27 resections was performed after preoperative chemotherapy without bevacizumab (CTX). There were 60 hepatic resections in this period without neoadjuvant treatment (NC). 8 patients had repeated resections. The postoperative complication rate was 40 % but there was no statistical difference between the groups (P=0.72). Only the type of resection was associated with a significantly higher complication rate (p=0.03). The subgroup of patients, who received irinotecan had a higher complication rate in the CTX group than in the BV+CTX group (55 % vs 41 %). Preoperative administration of bevacizumab was associated with higher peak postoperative AST, ALT levels but did not affect functional recovery of the liver. The RECIST system was not able to predict the outcome after chemotherapy in every patient and in many cases this system overestimated the effect of chemotherapy. On histopathological examination the presence of necrosis was not associated with chemotherapy or pathological response. Use of chemotherapy before hepatic resection of CLM was not associated with a significant increase in complication rates. The functional recovery of the liver was not affected by the preoperative administration of chemotherapy. The use of combined neoadjuvant chemotherapy is safe before hepatic resection. C1 [Dede, Kristof] Fovarosi Onkormanyzat Uzsoki utcai Korhaz, Sebeszeti-Onkosebeszeti Osztaly, Uzsoki utca 29, 1145 Budapest, Hungary. [Mersich, Tamas] Fovarosi Onkormanyzat Uzsoki utcai Korhaz, Sebeszeti-Onkosebeszeti Osztaly, Uzsoki utca 29, 1145 Budapest, Hungary. [Besznyak, Istvan] Fovarosi Onkormanyzat Uzsoki utcai Korhaz, Sebeszeti-Onkosebeszeti Osztaly, Uzsoki utca 29, 1145 Budapest, Hungary. [Zarand, Attila] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Salamon, Ferenc] Fovarosi Uzsoki utcai Korhaz, PathologiaBudapest, Hungary. [Baranyai, Zsolt] Szent Imre Hospital, Department of SurgeryBudapest, Hungary. [Landherr, Laszlo] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai KozpontBudapest, Hungary. [Jakab, Ferenc] Fovarosi Onkormanyzat Uzsoki utcai Korhaz, Sebeszeti-Onkosebeszeti Osztaly, Uzsoki utca 29, 1145 Budapest, Hungary. [Bursics, Attila] Fovarosi Onkormanyzat Uzsoki utcai Korhaz, Sebeszeti-Onkosebeszeti Osztaly, Uzsoki utca 29, 1145 Budapest, Hungary. RP Dede, K (reprint author), Fovarosi Onkormanyzat Uzsoki utcai Korhaz, Sebeszeti-Onkosebeszeti Osztaly, 1145 Budapest, Hungary. EM dede.kristof@gmail.com CR Adam R, Delvart V, Pascal G et al, 2004, Rescue surgery for unresectable colorectal liver metastases downstaged by chemotherapy: a model to predict long-term survival. Ann Surg 240:644–657 Kesmodel B, Ellis L et al, 2008, Preoperative bevacizumab does not significantly increase postoperative complication rates in patients undergoing hepatic surgery for colorectal cancer liver metastases. J Clin Oncol 26(32):5224–5260 Wicherts DA, de Haas RJ, Adam R et al, 2011, Impact of bevacizumab on functional recovery and histology of the liver after resection of colorectal metastases. Br J Surg 98(3):399–407 Folprecht G, Gruenberger T, Bechstein WO et al, 2010, Tumour response and secondary resectability of colorectal liver metastases following neoadjuvant chemotherapy with cetuximab: the CELIM randomised phase 2 trial. Lancet Oncol 11(1):38–47 Van Cutsem E, Rivera F, Berry S et al, 2009, Safety and efficacy of firstline bevacizumab with FOLFOX, XELOX, FOLFIRI and fluoropyrimidines in metastatic colorectal cancer: the BEAT study. Ann Oncol 20(11):1842–1847 Scoggins CR, Campbell ML, Landry CS et al, 2009, Preoperative chemotherapy does not increase morbidity or mortality of hepatic resection for colorectal cancer metastases. Ann Surg Oncol 16, 1):35–41 Vauthey JN, Pawlik TM, Ribero D et al, 2006, Chemotherapy regimen predicts steatohepatitis and an increase in 90-day mortality after surgery for hepatic colorectal metastases. J Clin Oncol 24, 13):2065–2072 Ribero D, Wang H, Donadon M et al, 2007, Bevacizumab improves pathologic response and protects against hepatic injury in patients treated with oxaliplatin-based chemotherapy for colorectal liver metastases. Cancer 110(12):2761–2767 Klinger M, Eipeldauer S, Hacker S et al, 2009, Bevacizumab protects against sinusoidal obstruction syndrome and does not increase response rate in neoadjuvant XELOX/FOLFOX therapy of colorectal cancer liver metastases. Eur J Surg Oncol 35(5):515– 520 D’Angelica M, Kornprat P, Gonen M et al, 2007, Lack of evidence for increased operative morbidity after hepatectomy with perioperative use of bevacizumab: a matched casecontrol study. An Surg Oncol 14:759–765 Ellis LM, Curley SA et al, 2005, Surgical resection after downsizing of colorectal liver metastasis in the era of Bevacizumab. J Clin Oncol 23(22):4853–4855 Reddy SK, Morse MA, Hurwitz HI et al, 2008, Addition of bevacizumab to irinotecan and oxaliplatin based preoperative chemotherapy regimens does not increase morbidity after resection of colorectal liver metastases. J Am Coll Surg 206:96–106 Mahfud M, Breitenstein S, El-Badry AM et al, 2010, Impact of preoperative bevacizumab on complications after resection of colorectal liver metastases: case-matched control study. World J Surg 34(1):92–100 Therasse P, Arbuck SG, Eisenhauer EA et al, 2000, New guidelines to evaluate the response to treatment is solid tumors. J Natl Cancer Inst 92:205–216 Dindo D, Demartines N, Clavien PA, 2004, Classification of surgical complications a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg 240:205–213 Dan G, Blazer III, Kishi Y et al, 2008, Pathologic response to preoperative chemotherapy: a new outcome end point after resection of hepatic colorectal metastases. J Clin Oncol 25(33):5344–5351 Nordlinger B, Vauthey J-N, Poston G et al, 2010, The timing of chemotherapy and surgery for the treatment of colorectal liver metastases. Clin Colorectal Cancer 9(4):212–218 Spelt L, Andersson B, Nilsson J, Andersson R, 2012, Prognostic models for outcome following liver resection for colorectal cancer metastases: a systematic review. EJSO 38:16–24 Gruenberger B, Tamandl D, Schueller J et al, 2008, Bevacizumab, capecitabine, and oxaliplatin as neoadjuvant therapy for patients with potentially curable metastatic colorectal cancer. J Clin Oncol 26(11):1830–1835 Nordlinger B, Sorbye H, Glimelius B et al, 2008, Perioperative chemotherapywith FOLFOX4 and surgery versus surgery alone for resectable liver metastases from colorectal cancer, EORTC Intergroup trial 40983): a randomisedcontrolled trial. Lancet 371, 9617):1007–1016 Chua TC, Saxena A, Liauw A et al, 2010, Systematic review of randomized and nonrandomized trials of the clinical response and outcomes of neoadjuvant systemic chemotherapy for resectable colorectal liver metastases. Ann Surg Oncol 17(2):492–501 Nordlinger B, Van Cutsem E, Gruenberger T, European Colorectal Metastases Treatment Group; Sixth International Colorectal Liver Metastases Workshop et al, 2009, Combination of surgery and chemotherapy and the role of targeted agents in the treatment of patients with colorectal liver metastases: recommendations from an expert panel. Ann Oncol 20(6):985–992 Lehmann K, Rickenbacher A, Weber A, Pestalozzi BC, Clavien PA, 2012, Chemotherapy before liver resection of colorectal metastases: friend or foe? Ann Surg 255:237–247 Scappaticci FA, Fehrenbacher L, Cartwright T et al, 2005, Surgical woundhealing complications in metastatic colorectal cancer patients treated with bevacizumab. J Surg Oncol 91:173–180 Tamandl D, Gruenberger B, Klinger M et al, 2010, Liver resection remains a safe procedure after neoadjuvant chemotherapy including bevacizumab: a case-controlled study. Ann Surg 252(1):124– 130 Kishi Y, Zorzi D, Contreras CM et al, 2010, Extended preoperative chemotherapy does not improve pathologic response and increases postoperative liver insufficiency after hepatic resection for colorectal liver metastases. Ann Surg Oncol 17(11):2870–2876 Gordon MS, Margolin K, Talpaz M et al, 2001, Phase I safety and pharmacokineticstudy of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J Clin Oncol 19(3):843–850 Chun YS, Vauthey JN, Boonsirikamchai P et al, 2009, Association of computed tomography morphologic criteria with pathologic response and survival in patients treated with bevacizumab for colorectal livermetastases. JAMA 302(21):2338–2344 Rubbia-Brandt L, Giostra E, Brezault C et al, 2007, Importance of histological tumor response assessment in predicting the outcome in patients with colorectal liver metastases treated with neoadjuvant chemotherapy followed by liver surgery. Ann Oncol 18:299–304 Dipen MM, Kopetz S, Boonsirikamchai P et al, 2010, Tumor thickness at the tumor-normal interface: a novel pathologic indicator of chemotherapy response in hepatic colorectal metastases. Am J Surg Pathol 34:1287–1294 Li Chang HH, Leeper WR, Chan G, Quan D, Driman DK, 2012, Infarct-like necrosis. A distinct form of necrosis seen in colorectal carcinoma liver metastases treated with perioperative chemotherapy. Am J Surg Pathol 36:570–576 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 501 EP 508 DI 10.1007/s12253-013-9608-2 PG 8 ER PT J AU de Abreu Pereira, D Areias, RV Franco, FM Benitez, CMM do Nascimento, MC de Azevedo, MC Alves, G AF de Abreu Pereira, Denise Areias, Rabello Vivian Franco, Felipe Marco Benitez, Carlos Moreira Manuel do Nascimento, Moreira Cristina de Azevedo, Maria Carolina Alves, Gilda TI Measurement of HER2 in Saliva of Women in Risk of Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Circulating HER2; Saliva; NAF; Breast cancer; Impalpable lesion ID Circulating HER2; Saliva; NAF; Breast cancer; Impalpable lesion AB HER2 amplification can be present in ductal carcinoma in situ (DCIS). The aim of the present study was to test the feasibility of measuring soluble HER2 in the saliva of patients at risk of breast cancer towards early diagnosis and prognosis.Women with lesions classified as 4 according to BIRADS and women with spontaneous nipple discharge (NAF) were recruited for this study. Quantification of soluble HER2 in saliva was performed using the enzyme immunoassay ELISA. Median values of HER2 were quantified in saliva of the control groups and in the patient groups. The statistical test nonparametric Mann–Whitney was applied for the evaluation of median differences. Although the medians increased with the severity of the clinical status, no significant difference was found in all possibilities (p> 0.05) when comparing the medians among the patients groups. Interestingly, inter-individual HER2 quantity variations in the saliva were detected in this study in some subjects from each group. Considering possible interindividual variations, research on saliva-based circulating HER2 has to be reinforced to ensure its correct application in diagnosis, treatment and in follow-up of breast cancer patients. Older and current issues surrounding the controversy about the appropriate methods for HER2 evaluation are discussed. C1 [de Abreu Pereira, Denise] Instituto Nacional de Cancer, Servico de Hematologia, Pc da Cruz Vermelha, 23, 6º andarRio de Janeiro, Brazil. [Areias, Rabello Vivian] Instituto Nacional de Cancer, Servico de Hematologia, Pc da Cruz Vermelha, 23, 6º andarRio de Janeiro, Brazil. [Franco, Felipe Marco] Instituto Nacional de Cancer, Servico de Hematologia, Pc da Cruz Vermelha, 23, 6º andarRio de Janeiro, Brazil. [Benitez, Carlos Moreira Manuel] Instituto Nacional de Cancer, Servico de Hematologia, Pc da Cruz Vermelha, 23, 6º andarRio de Janeiro, Brazil. [do Nascimento, Moreira Cristina] Instituto Nacional de Cancer, Divisao de Patologia, Av. Cordeiro da Graca, 156, Santo CristoRio de Janeiro, Brazil. [de Azevedo, Maria Carolina] Instituto Nacional de Cancer, Servico de Hematologia, Pc da Cruz Vermelha, 23, 6º andarRio de Janeiro, Brazil. [Alves, Gilda] Instituto Nacional de Cancer, Servico de Hematologia, Pc da Cruz Vermelha, 23, 6º andarRio de Janeiro, Brazil. RP Alves, G (reprint author), Instituto Nacional de Cancer, Servico de Hematologia, Rio de Janeiro, Brazil. EM gbrown@inca.gov.br CR Lang JE, Kuerer HM, 2007, Breast ductal secretions: clinical features, potential uses, and possible applications. Cancer Control 14(4):350–359 Wong DT, 2006, Salivary diagnostics powered by nanotechnologies, proteomics and genomics. J Am Dent Assoc 137(3):313–321 Streckfus C, Bigler L, Dellinger T, Pfeifer M, Rose A, Thigpen JT, 1999, CA 15–3 and c-erbB-2 presence in the saliva of women. Clin Oral Investig 3(3):138–143 Streckfus C, Bigler L, Dellinger T, Dai X, Kingman A, Thigpen JT, 2000, The presence of soluble c-erbB-2 in saliva and serum among women with breast carcinoma: a preliminary study. Clin Cancer Res 6(6):2363–2370 Streckfus C, Bigler L, Dellinger T, Dai X, Cox WJ, McArthur A, Kingman A, Thigpen JT, 2001, Reliability assessment of soluble c-erbB-2 concentrations in the saliva of healthy women and men. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 91(2):174–179 Streckfus C, Bigler L, 2005, The use of soluble, salivary c-erbB-2 for the detection and post-operative follow-up of breast cancer in women: the results of a five-year translational research study. Adv Dent Res 18(1):17–24 Esteva FJ, Cheli CD, Fritsche H, Fornier M, Slamon D, Thiel RP, Luftner D, Ghani F, 2005, Clinical utility of serum HER2/neu in monitoring and prediction of progression-free survival in metastatic breast cancer patients treated with trastuzumab-based therapies. Breast Cancer Res 7(4):R436–R443 McArthur H, 2009, An overview of HER-targeted therapy with lapatinib in breast cancer. Adv Ther 26(3):263–271 Zhou X, Cella D, Cameron D, Amonkar MM, Segreti A, Stein S, Walker M, Geyer CE, 2009, Lapatinib plus capecitabine versus capecitabine alone for HER2+, ErbB2+, metastatic breast cancer: quality-of-life assessment. Breast Cancer Res Treat 117(3):577– 589 Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, Thorsen T, Quist H, Matese JC, Brown PO, Botstein D, Lonning PE, Borresen-Dale AL, 2001, Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA 98(19):10869–10874 Barnes DM, Bartkova J, Camplejohn RS, Gullick WJ, Smith PJ, Millis RR, 1992, Overexpression of the c-erbB-2 oncoprotein: why does this occur more frequently in ductal carcinoma in situ than in invasive mammary carcinoma and is this of prognostic significance? Eur J Cancer 28(2–3):644–648 Allred DC, Clark GM, Molina R, Tandon AK, Schnitt SJ, Gilchrist KW, Osborne CK, Tormey DC, McGuireWL, 1992)Overexpression of HER-2/neu and its relationship with other prognostic factors change during the progression of in situ to invasive breast cancer. Hum Pathol 23(9):974–979 Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL, 1987, Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235(4785):177–182 Clark GM, McGuire WL, 1991, Follow-up study of HER-2/neu amplification in primary breast cancer. Cancer Res 51(3):944–948 Schroeter CA, De Potter CR, Rathsmann K, Willighagen RG, Greep JC, 1992, c-erbB-2 positive breast tumours behave more aggressively in the first years after diagnosis. Br J Cancer 66(4):728–734 Bianchi S, Paglierani M, Zampi G, Cardona G, Cataliotti L, Bonardi R, Ciatto S, 1993, Prognostic significance of c-erbB-2 expression in node negative breast cancer. Br J Cancer 67(3):625– 629 Press MF, Pike MC, Chazin VR, Hung G, Udove JA, Markowicz M, Danyluk J, Godolphin W, Sliwkowski M, Akita R, Paterson MC, Slnmoir DJ, 1993, Her-2/neu expression in node-negative breast cancer: direct tissue quantitation by computerized image analysis and association of overexpression with increased risk of recurrent disease. Cancer Res 53(20):4960–4970 Meijer SL, Wesseling J, Smit VT, Nederlof PM, Hooijer GK, Ruijter H, Arends JW, Kliffen M, van Gorp JM, Sterk L, van de Vijver MJ, 2011, HER2 gene amplification in patients with breast cancer with equivocal IHC results. J Clin Pathol 64(12):1069– 1072 Dowsett M, Hanna WM, Kockx M, Penault-Llorca F, Ruschoff J, Gutjahr T, Habben K, van de Vijver MJ, 2007, Standardization of HER2 testing: results of an international proficiency-testing ring study. Mod Pathol 20(5):584–591 Wludarski SC, Lopes LF, Berto E, Silva TR, Carvalho FM, Weiss LM, Bacchi CE, 2011, HER2 testing in breast carcinoma: very low concordance rate between reference and local laboratories in Brazil. Appl Immunohistochem MolMorphol 19(2):112– 118 Wilking U, Karlsson E, Skoog L, Hatschek T, Lidbrink E, Elmberger G, Johansson H, Lindstrom L, Bergh J, 2011, HER2 status in a population-derived breast cancer cohort: discordances during tumor progression. Breast Cancer Res Treat 125(2):553– 561 Aitken SJ, Thomas JS, Langdon SP, Harrison DJ, Faratian D, 2010, Quantitative analysis of changes in ER, PR and HER2 expression in primary breast cancer and paired nodal metastases. Ann Oncol 21(6):1254–1261 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 509 EP 513 DI 10.1007/s12253-013-9610-8 PG 5 ER PT J AU Kovacs, AK Kenessey, I Timar, J AF Kovacs, Attila Kristof Kenessey, Istvan Timar, Jozsef TI Skin Metastasis of Internal Cancers: A Single Institution Experience SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Skin metastasis; Visceral organs; Regional distribution ID Skin metastasis; Visceral organs; Regional distribution AB Skin metastatization of internal cancers are rare and a few studies are available analyzing its clinicopathological features. The reported incidence of skin metastasis is influenced by two factors: the relative proportion of cancers covered by skin in the various cohorts and the large differences in the prevalences of various cancer types. Futhermore, the anatomical distribution of skin metastases of various cancer types is aslo not well known. Therefore we have collected a skin metastasis cohort of biopsy and authopsy cases (n=80) from the archive of our department and analysed its clinicopathologic features. The adjusted skin metastasis prevalence data of various inner cancers indicated that kidney-, lung- and colorectal cancers have a strong positive preference for skin metastatisation while pancreatic cancer has a negative one. We have provided evidences that lower gastrointestinal- and genitourinary cancers preferred infradiaphragmatic skin regions unlike upper gastrointestinal cancers while lung- and kidney cancers preferred supradiaphragmatic regions. We have also detected that ventral skin regional metastasis is slightly more prevalent irrespective of the cancer type. Our study provide the first statistical data for the variations in skin preference of metastatisation among various cancer types as well as for the significant variations in their regional distributions. C1 [Kovacs, Attila Kristof] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93, 1091 Budapest, Hungary. [Kenessey, Istvan] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93, 1091 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, Ulloi u. 93, 1091 Budapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM jtimar@gmail.com CR Brownstein MH, Helwig EB, 1972, Patterns of cutaneous metastasis. Arch Dermatol 105(6):862–868 Lookingbill DP, Spangler N, Helm KF, 1993, Cutaneous metastases in patients with metastatic carcinoma: a retrospective study of 4020 patients. J Am Acad Dermatol 29:228–236 Nashan D, Muller ML, Falco MB, Reichenberger S, Szeimies RM, Bruckner-Tuderman L, 2009, Cutaneous metastases of visceral tumours: a review. J Cancer Res Clin Oncol 135:1–14 Hussein MRA, 2010, Skin metastasis: a pathologist’s perspective. J Cutan Pathol 37(9):1–20 Alcaraz I, Cerroni L, Rutten A, Kutzner H, Requena L, 2012, Cutaneous metastases from internal malignancies: a clinicopathologic and immunohistochemical review. Am J Dermatopathol 34(4):347–393 Fernandez-Flores A, 2010, Cutaneous metastases: a study of 78 biopsies from 69 patients. Am J Dermatopathol 32(3):222–239 Lookingbill DP, Spangler N, Sexton FM, 1990, Skin involvement as the presenting sign of internal carcinoma: a retrospective study of 7316 cancer patients. J Am Acad Dermatol 22:19–26 Bobba RK, Odem JL, Doll DC, Perry MC, 2012, Skin metastases in non-small cell lung cancer. Am J Med Sci 344:59–62 Pereira WA, Humaire CR, Silva CS, Fernandes LH, 2011, Sister Mary Joseph’s nodule: a sign of internal malignancy. Dermatol 86: S118–S120 Saeed S, Keehn CA, Morgan MB, 2004, Cutaneous metastasis: a clinical, pathological and immunohistochemical appraisal. J Cutan Pathol 31:419–430 Sariya D, Ruth K, McDonnell RA, Cusack C, Xu XW, Elenitsas R, Seykora J, Pasha T, Zhang P, Baldassano M, Lessin SR, Wu H, 2007, Clinicopathologic correlation of cutaneous metastases. Arch Dermatol 143:613–620 Hu SC, Chen GS, Wu CS, Chai CY, Chen WT, Lan CC, 2009, Rates of cutaneous metastases from different internal malignancies: experience from a Taiwanese medical center. J Am Acad Dermatol 60(3):379–387 Garrido MJM, Ponce CP, Martinez JLS, Scylla DM, Mena AC, 2006, Cutaneous metastases of lung cancer. Clin Transl Oncol 8:330–333 Dorairajan LN, Hemal AK, Aron M, Rajeev TP, Nair M, Seth A, Dogra PN, Gupta NP, 1999, Cutaneous metastases in renal cell carcinoma. Urol Int 63:164–167 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 515 EP 520 DI 10.1007/s12253-013-9611-7 PG 6 ER PT J AU Wang, M Zhang, D Wang, R Rui, Y Zhou, J Wang, R Zhou, B Huang, X Yang, L Li, Y Hu, J Zhou, Z Sun, X AF Wang, Mojin Zhang, Dan Wang, Rui Rui, Yuanyi Zhou, Jin Wang, Rong Zhou, Bin Huang, Xiaoran Yang, Lie Li, Yuan Hu, Jiankun Zhou, Zongguang Sun, Xiaofeng TI A-Kinase Anchoring Proteins 10 Expression in Relation to 2073A/G Polymorphism and Tumor Progression in Patients with Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE A-kinase anchoring proteins 10; Immunohistochemical staining; Western blot; Polymorphism; Colorectal cancer ID A-kinase anchoring proteins 10; Immunohistochemical staining; Western blot; Polymorphism; Colorectal cancer AB The cAMP/PKA signalling events regulated by A-kinase anchoring proteins 10 (AKAP10) is involved in tumorigenesis. Previous study showed that AKAP10 polymorphism (2073 A/G, I646V) was associated with colorectal cancer risk. However, there was no literature reporting the role of AKAP10 in the pathogenesis of colorectal cancer. The aim of the study was to investigate the clinicopathologic significance of A-kinase anchoring proteins 10 (AKAP 10) expression and the relationship with its polymorphism in colorectal cancer. The expression of AKAP10 was determined by immunohistochemical staining (IHC) and western blot assay on colorectal cancer (n=176), adenoma (n=87) and distant normal mucosa (n=72). 176 patients with colorectal cancer were genotyped for AKAP10 2073A/G polymorphism by TaqMan RT-PCR. We found that the positive expression rate of AKAP10 in colorectal cancer (59 %) was significantly higher than those in adenoma (39 %) and distant normal mucosa (42 %) (P=0.004). There was no significant difference between adenoma and distant normal mucosa (P=0.741). Positive AKAP10 staining was correlated with deeper tumor invasion (P<0.001), lymph nodes metastasis (P=0.022), advanced tumor stage (P<0.001) and poorly differentiated degree (P=0.003). Compared with AA genotype (52 %), positive expression of AKAP10 was significantly increased in colorectal cancer patients with the variant (AG+GG) genotypes (68 %, P=0.033). It was concluded that AKAP10 may play an important role in the development and progression of colorectal cancer. C1 [Wang, Mojin] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 37 Guo Xue Xiang, 610041 Chengdu, China. [Zhang, Dan] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 37 Guo Xue Xiang, 610041 Chengdu, China. [Wang, Rui] Sichuan University, West China Hospital, Department of GastroenterologyChengdu, China. [Rui, Yuanyi] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 37 Guo Xue Xiang, 610041 Chengdu, China. [Zhou, Jin] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 37 Guo Xue Xiang, 610041 Chengdu, China. [Wang, Rong] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 37 Guo Xue Xiang, 610041 Chengdu, China. [Zhou, Bin] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 37 Guo Xue Xiang, 610041 Chengdu, China. [Huang, Xiaoran] Sichuan University, West China Hospital, Department of PathologyChengdu, China. [Yang, Lie] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 37 Guo Xue Xiang, 610041 Chengdu, China. [Li, Yuan] West China Hospital, Sichuan University, Institute of Digestive Surgery and State Key Laboratory of Biotherapy, Department of Pediatric SurgeryChengdu, China. [Hu, Jiankun] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 37 Guo Xue Xiang, 610041 Chengdu, China. [Zhou, Zongguang] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 37 Guo Xue Xiang, 610041 Chengdu, China. [Sun, Xiaofeng] University of Linkoping, Faculty of Health Sciences, Department of Clinical and Experimental Medicine, Division of OncologyLinkoping, Sweden. RP Zhou, Z (reprint author), Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 610041 Chengdu, China. EM zhou_767@163.com CR Vogelstein B, Fearon ER, Hamilton SR et al, 1988, Genetic alterations during colorectal-tumor development. N Engl J Med 319:525–532 Cho YS, Park YG, Lee YN et al, 2000, Extracellular protein kinase A as a cancer biomarker: its expression by tumor cells and reversal by a myristatelacking Ca and RIIβ subunit overexpression. Proc Natl Acad Sci USA 97:835–840 Wong W, Scott JD, 2004, AKAP signalling complexes: focal points in space and time. Nat Rev Mol Cell Biol 5:959–970 Huang L, Durick K, Weiner JA, Chun J, Taylor SS, 1997, DAKAP2, a novel protein kinase A anchoring protein with a putative RGS domain. Proc Natl Acad Sci USA 94:11184–11189 Hamuro Y, Burns L, Canaves J, Hoffman R, Taylor SS, Woods V, 2002, Domain organization of D-AKAP2 revealed by enhanced deuterium exchange-mass spectrometry, DXMS). J Mol Biol 321:703–714 Sheng M, Sala C, 2001, PDZ domains and the organization of supramolecular complexes. Annu Rev Neurosci 24:1–29 Wang L, Sunahara RK, Krumins A et al, 2001, Cloning and mitochondrial localization of full-length D-AKAP2, a protein kinase A anchoring protein. Proc Natl Acad Sci USA 98:3220–3225 Burns-Hamuro LL, Barraclough DM, Taylor SS, 2004, Identification and functional analysis of dual-specific A kinaseanchoring protein-2. Methods Enzymol 390:354–374 Neary CL, Nesterova M, Cho YS, Cheadle C, Becker KG, Cho-Chung YS, 2004, Protein kinase A isozyme switching: eliciting differential cAMP signaling and tumor reversion. Oncogene 23:8847–8856 Cross TG, Scheel-Toellner D, Henriquez NV, Deacon E, Salmon M, Lord JM, 2000, Serine/threonine protein kinases and apoptosis. Exp Cell Res 256:34–41 Tasken K, Skalhegg BS, Tasken KA et al, 1997, Structure, function, and regulation of human cAMP-dependent protein kinases. Adv Second Messenger Phosphoprotein Res 31:191–204 Wirtenberger M, Schmutzhard J, Hemminkil K et al, 2007, The functional genetic variant Ile646Val located in the kinase binding domain of the A-kinase anchoring protein 10 is associated with familial breast cancer. Carcinogenesis 28:423–426 Wang MJ, Zhou ZG, Wang L et al, 2009, The Ile646Val, 2073A>G, Polymorphism in the kinase-binding domain of A-kinase anchoring protein 10 and the risk of colorectal cancer. Oncology 76:199–204 Compton CC, Greene FL, 2004, The staging of colorectal cancer: 2004 and beyond. CA Cancer J Clin 54:295–308 Yang L, Zhang H, Zhou ZG, Yan H, Adell G, Sun XF, 2011, Biological function and prognostic significance of peroxisome proliferator-activated receptor δ in rectal cancer. Clin Cancer Res 17:3760–3770 Dodge-Kafka KL, Soughayer J, Pare GC et al, 2005, The protein kinase A anchoring protein mAKAP coordinates two integrated cAMP effector pathways. Nature 437:574–578 Bradbury AW, Carter DC, Miller WR, Cho-Chung YS, Clair T, 1994, Protein kinase A, PKA, regulatory subunit expression in colorectal cancer and related mucosa. Br J Cancer 69:738–742 Bold RJ, Alpard S, Ishizuka J, Townsend CM Jr, Thompson JC, 1994, Growth-regulatory effect of gastrin on human colon cancer cell lines is determined by protein kinase a isoform content. Regul Pept 53:61–70 Nesterova MV, Johnson N, Cheadle C et al, 2006, Autoantibody cancer biomarker: extracellular protein kinase A. Cancer Res 66:8971–8974 Hensley HH, Hannoun-Levi JM, Hachem P et al, 2011, PKA knockdown enhances cell killing in response to radiation and androgen deprivation. Int J Cancer 128:962–973 Chen TC, Hinton DR, Zidovetzki R, Hofman FM, 1998, Upregulation of the cAMP/PKA pathway inhibits proliferation, induces differentiation, and leads to apoptosis in malignant gliomas. Lab Invest 78:165–174 Siddappa R, Mulder W, Steeghs I et al, 2009, cAMP/PKA signaling inhibits osteogenic differentiation and bone formation in rodent models. Tissue Eng Part A 15:2135–2143 Loniewska B, Clark JS, Kaczmarczyk M et al, 2012, Possible counter effect in newborns of 1936A>G, I646V, polymorphism in the AKAP10 gene encoding A-kinase-anchoring protein 10. J Perinatol 32:230–234 Zukowski M, Bohatyrewicz R, Biernawska J et al, 2009, Association of the A1936G, rs203462, of A-kinase anchoring protein 10 polymorphisms with QT interval prolongation during kidney transplantation. Transplant Proc 41:3036–3038 Kammerer S, Burns-Hamuro LL, Ma Y et al, 2003, Amino acid variant in the kinase binding domain of dual-specific A kinaseanchoring protein 2: a disease susceptibility polymorphism. Proc Natl Acad Sci USA 100:4066–4071 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 521 EP 527 DI 10.1007/s12253-013-9612-6 PG 7 ER PT J AU Xiong, Sw Lin, Tx Xu, Kw Dong, W Ling, Xh Jiang, Fn Chen, G Zhong, Wd Huang, J AF Xiong, Si-wei Lin, Tian-xin Xu, Ke-wei Dong, Wen Ling, Xiao-hui Jiang, Fu-neng Chen, Guo Zhong, Wei-de Huang, Jian TI MicroRNA-335 Acts as a Candidate Tumor Suppressor in Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; microRNA-335; Tumor suppressor; Clinicopathological feature; Prognosis ID Prostate cancer; microRNA-335; Tumor suppressor; Clinicopathological feature; Prognosis AB MicroRNA-335 (miR-335) acts as a tumor suppressor or a tumor promoter in different human malignancies. However, the involvement of miR-335 in prostate cancer (PCa) is still unclear. The purpose of this study was to investigate the functional and clinical significance of miR-335 in PCa. miR-335 expression in 3 PCa cell lines (LNCaP/DU145/PC3) and in 20 clinical PCa tissues were detected by real-time quantitative reverse transcriptase-PCR compared with corresponding controls. The function of miR-335 was investigated for cell proliferation, invasion and migration in PCa cells transfected with agents containing EGFP-miR-335 expression vector. Additionally, miR-335 expression in 104 clinical PCa tissues was detected by in situ hybridization. Its assocaitions with clinicopathological features and prognosis in patients with PCa were also determined. miR-335 was significantly down-regulated in PCa cell lines than in the normal prostate cell line (P<0.01). With the similar results in vitro, the reduced expression of miR-335 was also found in human PCa tissues comparing with paired adjacent benign prostate tissues (P<0.05). Moreover, the increased expression of miR-335 suppressed cell proliferation, invasion and migration of PCa cell lines in vitro. Turning to its clinical significance, the low expression of miR-335 was significantly associated with high Gleason Score (P=0.04), advanced clinical stage (P=0.04), and positive metastasis (P=0.02), but not with prognosis in PCa patients. Our data demonstrated for the first time the inhibitory effect of miR-335 on cell proliferation and invasion for PCa cells. The loss of this microRNA might be associated with clinical progression of PCa patients. C1 [Xiong, Si-wei] Sun Yat-sen University, Sun Yat-sen Memorial Hospital, Department of Urology, 510120 Guangzhou, China. [Lin, Tian-xin] Sun Yat-sen University, Sun Yat-sen Memorial Hospital, Department of Urology, 510120 Guangzhou, China. [Xu, Ke-wei] Sun Yat-sen University, Sun Yat-sen Memorial Hospital, Department of Urology, 510120 Guangzhou, China. [Dong, Wen] Sun Yat-sen University, Sun Yat-sen Memorial Hospital, Department of Urology, 510120 Guangzhou, China. [Ling, Xiao-hui] Guangzhou Medical College, First Affiliated Hospital of Guangzhou Medical College, Urology Key Laboratory of Guangdong Province, 510180 Guangzhou, China. [Jiang, Fu-neng] Guangzhou Medical College, First Affiliated Hospital of Guangzhou Medical College, Urology Key Laboratory of Guangdong Province, 510180 Guangzhou, China. [Chen, Guo] Guangzhou Medical College, First Affiliated Hospital of Guangzhou Medical College, Urology Key Laboratory of Guangdong Province, 510180 Guangzhou, China. [Zhong, Wei-de] Guangzhou Medical College, First Affiliated Hospital of Guangzhou Medical College, Urology Key Laboratory of Guangdong Province, 510180 Guangzhou, China. [Huang, Jian] Sun Yat-sen University, Sun Yat-sen Memorial Hospital, Department of Urology, 510120 Guangzhou, China. RP Huang, J (reprint author), Sun Yat-sen University, Sun Yat-sen Memorial Hospital, Department of Urology, 510120 Guangzhou, China. EM Yehjn@yahoo.com.cn CR Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics, 2012. CA Cancer J Clin 62:10–29 Utomo NB, Mochtar CA, Umbas R, 2012, Primary hormonal treatment in localized and locally advanced prostate cancer: effectiveness and survival predictive factors. Acta Med Indones 44:10–15 Dasgupta S, Srinidhi S, Vishwanatha JK, 2012, Oncogenic activation in prostate cancer progression and metastasis: molecular insights and future challenges. J Carcinog 11:4 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281–297 Calin GA, Liu CG, Sevignani C, Ferracin M, Felli N, Dumitru CD, Shimizu M, Cimmino A, Zupo S, Dono M, Dell’Aquila ML, Alder H, Rassenti L, Kipps TJ, Bullrich F, Negrini M, Croce CM, 2004, MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proc Natl Acad Sci USA 101:11755–11760 Porkka KP, Pfeiffer MJ, Waltering KK, Vessella RL, Tammela TL, Visakorpi T, 2007, MicroRNA expression profiling in prostate cancer. Cancer Res 67:6130–6135 Shu M, Zhou Y, Zhu W, Zhang H, Wu S, Chen J, Yan G, 2012, MicroRNA 335 is required for differentiation of malignant glioma cells induced by activation of cAMP/protein kinase A pathway. Mol Pharmacol 81:292–298 Nakanishi N, Nakagawa Y, Tokushige N, Aoki N, Matsuzaka T, Ishii K, Yahagi N, Kobayashi K, Yatoh S, Takahashi A, Suzuki H, Urayama O, Yamada N, Shimano H, 2009, The up-regulation of microRNA-335 is associated with lipid metabolism in liver and white adipose tissue of genetically obese mice. Biochem Biophys Res Commun 385:492–496 Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, Gerald WL, Massague J, 2008, Endogenous human microRNAs that suppress breast cancer metastasis. Nature 451:147–152 Wang F, Zheng Z, Guo J, Ding X, 2010, Correlation and quantitation of microRNA aberrant expression in tissues and sera from patients with breast tumor. Gynecol Oncol 119:586–593 Sorrentino A, Liu CG, Addario A, Peschle C, Scambia G, Ferlini C, 2008, Role of microRNAs in drug-resistant ovarian cancer cells. Gynecol Oncol 111:478–486 Shu M, Zheng X, Wu S, Lu H, Leng T, Zhu W, Zhou Y, Ou Y, Lin X, Lin Y, Xu D, Zhou Y, Yan G, 2011, Targeting oncogenic miR- 335 inhibits growth and invasion of malignant astrocytoma cells. Mol Cancer 10:59 Chen JH, He HC, Jiang FN et al, 2012, Analysis of the specific pathways and networks of prostate cancer for gene expression profiles in the Chinese population. Med Oncol 29:1972–1984 Chen C, Ridzon DA, Broomer AJ et al, 2005, Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res 33:e179 Nuovo GJ, 2008, In situ detection of precursor and mature microRNAs in paraffin embedded, formalin fixed tissues and cell preparations. Methods 44:39–46 Wang L, Alcon A, Yuan H, Ho J, Li QJ, Martins-Green M, 2011, Cellular and molecular mechanisms of pomegranate juice-induced anti-metastatic effect on prostate cancer cells. Integr Biol 3:742–754 Vickers MM, Bar J, Gorn-Hondermann I, Yarom N, Daneshmand M, Hanson JE, Addison CL, Asmis TR, Jonker DJ, Maroun J, Lorimer IA, Goss GD, Dimitroulakos J, 2012, Stage-dependent differential expression of microRNAs in colorectal cancer: potential role as markers of metastatic disease. Clin Exp Metastasis 29:123–132 Marcucci G, Maharry K, Radmacher MD, Mrόzek K, Vukosavljevic T, Paschka P, 2008, Prognostic significance of, and gene and microRNA expression signatures associated with, CEBPA mutations in cytogenetically normal acute myeloid leukemia with highrisk molecular features: a Cancer and Leukemia Group B Study. J Clin Oncol 26:5078–5087 Soon PS, Tacon LJ, Gill AJ, Bambach CP, Sywak MS, Campbell PR, 2009, miR-195 and miR-483-5p identified as predictors of poor prognosis in adrenocortical cancer. Clin Cancer Res 15:7684– 7692 Xu Y, Zhao F, Wang Z, Song Y, Luo Y, Zhang X, Jiang L, Sun Z, Miao Z, Xu H, 2012, MicroRNA-335 acts as a metastasis suppressor in gastric cancer by targeting Bcl-w and specificity protein 1. Oncogene 31:1398–1407 Wang YX, Zhang XY, Zhang BF, Yang CQ, Chen XM, Gao HJ, 2010, Initial study of microRNA expression profiles of colonic cancer without lymph node metastasis. J Dig Dis 11:50–54 Zhang H, Luo XQ, Zhang P, Huang LB, Zheng YS, Wu J, Zhou H, Qu LH, Xu L, Chen YQ, 2009, MicroRNA patterns associated with clinical prognostic parameters and CNS relapse prediction in pediatric acute leukemia. PLoS One 4:e7826 Ronchetti D, Lionetti M, Mosca L, Agnelli L, Andronache A, Fabris S, Deliliers GL, Neri A, 2008, An integrative genomic approach reveals coordinated expression of intronic miR-335, miR-342, and miR-561 with deregulated host genes in multiple myeloma. BMC Med Genomics 1:37 Wang J, Ruan K, 2010, miR-335 is involved in the rat epididymal development by targeting the mRNA of RASA1. Biochem Biophys Res Commun 402(2):222–227 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 529 EP 537 DI 10.1007/s12253-013-9613-5 PG 9 ER PT J AU Pappa, AC Tsirakis, G Boula, A Sfiridaki, A Psarakis, EF Alexandrakis, GM Stathopoulos, NE AF Pappa, A Constantina Tsirakis, George Boula, Anna Sfiridaki, Aikaterini Psarakis, E Fotios Alexandrakis, G Michael Stathopoulos, N Efstathios TI The Significance of non Correlation Between Interleukin-8 Serum Levels with Bone Marrow Microvascular Density in Patients with Myeloma Multiple SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Myeloma; Cytokines; Bone marrow; Inflammation ID Myeloma; Cytokines; Bone marrow; Inflammation AB In multiple myeloma (MM), angiogenesis plays a substantial role in disease progression. Interleukin-8 (IL-8), a pro-inflammatory chemokine with potent pro-angiogenic properties, has been implicated in the pathophysiology of MM. The aim of the study is to measure serum levels of IL- 8 in MM patients and to correlate them with markers of angiogenesis, such as circulating levels of platelet derived growth factor-AB (PDGF-AB) and angiogenin (Ang), and bone marrow microvascular density (MVD). Fifty-three newly diagnosed MM patients, 23 of them, who reached plateau phase after effective treatment and 20 healthy controls, were studied. Serum levels of PDGF-AB, Ang and IL- 8 were measured by ELISA, whereas bone marrow MVD was estimated by immunohistochemical staining of vessels with anti-CD31. All measured parameters were higher in MM patients compared to controls and in increased disease stages. They all also significantly decreased in plateau phase. IL-8 correlated positively with Ang and PDGF-AB, but not with MVD. The circulating levels of IL-8, PDGFAB and Ang are elevated in patients with MM. The lack of correlation between IL-8 with MVD suggests that its levels represent the inflammatory element of MM disease and the participation in angiogenesis process is rather complex with multifactorial mechanisms. C1 [Pappa, A Constantina] University Hospital of Heraklion, Hematology Department, Knossou Avenue, 71409 Heraklion, Greece. [Tsirakis, George] University Hospital of Heraklion, Hematology Department, Stavrakia, 71110 Heraklion, Greece. [Boula, Anna] University Hospital of Heraklion, Hematology Department, Knossou Avenue, 71409 Heraklion, Greece. [Sfiridaki, Aikaterini] University Hospital of Heraklion, Hematology Department, Knossou Avenue, 71409 Heraklion, Greece. [Psarakis, E Fotios] University Hospital of Heraklion, Hematology Department, Knossou Avenue, 71409 Heraklion, Greece. [Alexandrakis, G Michael] University Hospital of Heraklion, Hematology Department, Stavrakia, 71110 Heraklion, Greece. [Stathopoulos, N Efstathios] Medical School of the University of Crete, Department of Pathology, Stavrakia, 71110 Heraklion, Greece. RP Alexandrakis, GM (reprint author), University Hospital of Heraklion, Hematology Department, 71110 Heraklion, Greece. EM alexandm@med.uoc.gr CR Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC, 2007, Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer 7:585–598 Sezer O, Niemoller K, Eucker J, Jacob C, Kaufmann O, Zavrski I, Dietel M, Possinger K, 2000, Bone marrow microvessel density is a prognostic factor for survival in patients with multiple myeloma. Ann Hemal 79:574–577 Tsirakis G, Pappa C, Kanellou P, Stratinaki M, Xekalou A, Psarakis F, Sakellaris G, Alegakis A, Stathopoulos E, Alexandrakis M, 2012, Role of platelet-derived growth factor- AB in tumour growth and angiogenesis in relation with other angiogenic cytokines in multiple myeloma. Hematol Oncol 30:131–136 Kastritis E, Roussou M, Michalis M, Gavriatopoulou M, Michalis E, Migkou M, Delimpasi S, Kyrtsonis MC, Gogos D, Liapis K, Charitaki E, Repousis P, Terpos E, Dimopoulos A, 2010, On behalf of the Greek Myeloma Study Group. High levels of serum angiogenic growth factors in patients with AL amyloidosis: comparisons with normal individuals and multiple myeloma patients. Br J Haematol 150:587–591 Tsirakis G, Pappa C, Kaparou M, Katsomitrou V, Xatzivasili A, Alegakis T, Xekalou A, Stathopoulos E, Alexandrakis M, 2011, Assessment of proliferating cell nuclear antigen and its relationship with proinflammatory cytokines and parameters of disease activity in multiple myeloma patients. Eur J Histochem 55(e21):113–116 Singh R, Varney M, 2000, Il-8 expression in malignant melanoma: implications in growth and metastasis. Histol Histopathol 15:843– 849 Kline M, Donovan K, Wellik L, Jin W, Moon-Tasson L, Xiong Y, Witsig TE, Kumar S, Rajkumar S, Lust J, 2007, Cytokine and chemokine profiles in multiple myeloma; significance of stromal interaction and correlation of IL-8 production with disease progression. Leuk Res 31:591–598 Pappa C, Tsirakis G, Kanellou P, Kaparou M, Stratinaki M, Xekalou A, Alegakis A, Boula A, Stathopoulos E, Alexandrakis M, 2011, Monitoring serum levels ELR + CXC chemokines and the relationship between microvessel density and angiogenic growth factors in multiple myeloma. Cytokine 56:616–620 Alexandrakis MG, Passam FH, Pappa CA, Dambaki C, Sfakiotaki G, Alegakis AK, Kyriakou DS, Stathopoulos E, 2004, Expression of proliferating cell nuclear antigen, PCNA, in multiple myeloma: its relationship to bone marrow microvessel density and other factors of disease activity. Int J Immunopathol Pharmacol 17:49– 56 Wright D, Bowman E, Wagers A, Butcher E, Weissman I, 2002, Hematopoietic stem cells are uniquely selective in the migratory response to chemokines. J Exp Med 195:1145–1154 Qazi BS, Tang K, Qazi A, 2011, Recent advances in underlying pathologies provide insight into interleukin-8 expression-mediated inflammation and angiogenesis. Int J Inflamm 2011:908468 Dobreva I, Waeber G, James RW, Widmann C, 2006, Interleukin- 8 secretion by fibroblasts induced by low density lipoproteins is p38 MAPK-dependent and leads to cell spreading and wound closure. J Biol Chem 281:199–205 Aggarwal R, Ghobrial IM, Roodman GD, 2006, Chemokines in multiple myeloma. Exp Hematol 34:1289–1295 Benelli R, Lorusso G, Albini A, Noonan DM, 2006, Cytokines and chemokines as regulators of angiogenesis in health and disease. Curr Pharm Des 12:3101–3115 Markovina S, Callander NS, O'Connor SL, Xu G, Shi Y, Leith CP, Kim K, Trivedi P, Kim J, Hematti P, Miyamoto S, 2010, Bone marrow stromal cells from multiple myeloma patients uniquely induce bortezomib resistant NF-kappaB activity in myeloma cells. Mol Canc 9:176 Benelli R, Morini M, Carrozzino F, Ferrari N, Minghelli S, Santi L, Cassatella M, Noonan DM, Albini A, 2002, Neutrophils as a key cellular target for angiostatin: implications for regulation of angiogenesis and inflammation. FASEB J 16:267–269 Benelli R, Albini A, Noonan D, 2003, Neutrophils and angiogenesis: potential initiators of the angiogenic cascade. Chem Immunol Allergy 83:167–181 Scapini P, Morini M, Tecchio C, Minghelli S, Di Carlo E, Tanghetti E, Albini A, Lowell C, Berton G, Noonan DM, Cassatella MA, 2004, CXCL1/macrophage inflammatory protein-2-induced angiogenesis in vivo is mediated by neutrophil-derived vascular endothelial growth factor-A. J Immunol 172:5034–5040 Li A, Varney ML, Valasek J, Godfrey M, Dave BJ, Singh RK, 2005, Autocrine role of interleukin-8 in induction of endothelial cell proliferation, survival, migration and MMP-2 production and angiogenesis. Angiogenesis 8:63–71 Huang S, Mills L, Mian B, Tellez C, McCarty M, Yang XD, Gudas JM, Bar-Eli M, 2002, Fully humanized neutralizing antibodies to interleukin-8, ABX-IL8, inhibit angiogenesis, tumor growth, and metastasis of human melanoma. Am J Pathol 161:125–134 Dong X, Han ZC, Yang R, 2007, Angiogenesis and antiangiogenic therapy in hematologic malignancies. Crit Rev Oncol Hematol 62:105–118 Negaard HF, Iversen N, Bowitz-Lothe IM, Sandset PM, Steinsvik B, Ostenstad B, Iversen PO, 2009, Increased bone marrow microvascular density in haematological malignancies is associated with differential regulation of angiogenic factors. Leukemia 23:162– 169 Rakjumar V, Mesa R, Fonseca R, Schroeder G, Plevak M, Dispenzieri A, Lacy M, Lust J, Witzig T, Gertz M, Kule R, Russell S, Greipp P, 2002, Bone marrow angiogenesis in 400 patients with monoclonal gammopathy of undetermined significance, multiple myeloma and primary amyloidosis. Clin Canc Res 8:2210–2216 Zhang J, Cao R, Zhang Y, Jia T, Cao Y, Wahlberg E, 2009, Differential roles of PDGFR-alpha and PDGFR-beta in angiogenesis and vessel stability. FASEB J 23:153–163 Miyagaki T, Sugaya M, Suga H, Akamata K, Ohmatsu H, Fujita H, Asano Y, Tada Y, Kadono T, Sato S, 2012, Angiogenin levels are increased in lesional skin and sera in patients with erythrodermic cutaneous T cell lymphoma. Arch Dermatol Res 304:401–406 Hu GF, Riordan JF, 1993, Angiogenin enhances actin acceleration of plasminogen activation. Biochem Biophys Res Commun 197:682–687 Hu GF, Chang SI, Riordan JF, Vallee BL, 1991, An angiogeninbinding protein from endothelial cells. Proc Natl Acad Sci USA 88:2227–2231 Dungwa J, Uparkar U, May M, Ramani P, 2012, Angiogenin upregulation correlates with adverse clinicopathological and biological factors, increased microvascular density and poor patient outcome in neuroblastomas. Histopathology 60:911–923 Li S, Ibaragi S, Hu GF, 2011, Angiogenin as a molecular target for the treatment of prostate cancer. Curr Canc Ther Rev 7:83–90 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 539 EP 543 DI 10.1007/s12253-013-9614-4 PG 5 ER PT J AU Nikuseva-Martic, T Serman, L Zeljko, M Vidas, Gasparov, S Zeljko, MH Kosovic, M Pecina-Slaus, N AF Nikuseva-Martic, Tamara Serman, Ljiljana Zeljko, Martina Vidas, Zeljko Gasparov, Slavko Zeljko, Marija Hrvojka Kosovic, Marin Pecina-Slaus, Nives TI Expression of Secreted Frizzled-Related Protein 1 and 3, T-cell Factor 1 and Lymphoid Enhancer Factor 1 in Clear Cell Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SFRP1; SFRP3; TCF1; LEF1; cRCC ID SFRP1; SFRP3; TCF1; LEF1; cRCC AB Frequency and mortality of renal cell carcinoma (RCC) are increasing for decades. However, the molecular background of RCC tumorigenesis is still poorly understood. In current study we investigated the expression of TCF/LEF and SFRP family members (SFRP1 and SFRP3) to gain a better understanding of biological signaling pathways responsible for epidemiology and clinical parameters of clear cell RCC (cRCC). Thirty-six pairs of paraffinembedded clear cRCC and adjacent nontumoral tissues samples using immunohistochemistry (IHC) were analyzed and compared with corresponding clinicopathological parameters. Immunohistochemistry indicated statistically significant decreased SFRP3 expression in tumor tissues but no consistency in SFRP1 expression in analyzed normal and tumor tissue. The TCF1 expression level was significantly weaker in normal tissue compared to tumor samples while LEF1 protein levels were significantly weaker in tumor tissue. To our knowledge, this is the first report on analysis of the expression of transcription factors TCF1 and LEF1 in clear cell renal cell carcinoma and their comparison with Wnt signal pathway antagonists belonging to SFRP family. C1 [Nikuseva-Martic, Tamara] University of Zagreb, School of Medicine, Department of Biology, Salata 3, 10000 Zagreb, Croatia. [Serman, Ljiljana] University of Zagreb, School of Medicine, Department of Biology, Salata 3, 10000 Zagreb, Croatia. [Zeljko, Martina] “Merkur” University Hospital, Department of Internal MedicineZagreb, Croatia. [Vidas, Zeljko] “Merkur” University Hospital, Department of UrologyZagreb, Croatia. [Gasparov, Slavko] “Merkur” University Hospital, Department of Clinical Pathology and CytologyZagreb, Croatia. [Zeljko, Marija Hrvojka] “Merkur” University Hospital, Department of Internal MedicineZagreb, Croatia. [Kosovic, Marin] University of Zagreb, School of Medicine, Department of Physics and BiophysicsZagreb, Croatia. [Pecina-Slaus, Nives] University of Zagreb, School of Medicine, Department of Biology, Salata 3, 10000 Zagreb, Croatia. RP Nikuseva-Martic, T (reprint author), University of Zagreb, School of Medicine, Department of Biology, 10000 Zagreb, Croatia. EM tmartic@mef.hr CR Moon RT, Kohn AD, De Ferrari GV, Kaykas A, 2004, WNT and hcatenin signaling: diseases and therapies. Nat Rev Genet 5:691–701 Nelson WJ, Nusse R, 2004, Convergence of Wnt, h-catenin, and cadherin pathways. Science 303:1483–1487 Waterman ML, 2004, Lymphoid enhancer factor/T cell factor expression in colorectal cancer. Cancer Metastasis Rev 23:41– 52 Schilham MW, Clevers H, 1998, HMG box containing transcription factors in lymphocyte differentiation. Semin Immunol 10:127–132 Brantjes H, Barker N, van Es J, Clevers H, 2002, TCF: Lady Justice casting the final verdict on the outcome of Wnt signalling. Biol Chem 383:255–261 Reya T, Clevers H, 2005, Wnt signalling in stem cells and cancer. Nature 434:843–850 Logan CY, Nusse R, 2004, The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol 20:781–810 Dennis S, Aikawa M, Szeto W, d’Amore PA, Papkoff J, 1999, A secreted frizzled related protein, FrzA, selectively associates with Wnt-1 protein and regulates wnt-1 signaling. J Cell Sci 112:3815–3820 Uren A, Reichsman F, Anest V, Taylor WG, Muraiso K, Bottaro DP, Cumberledge S, Rubin JS, 2000, Secreted frizzled related protein-1 binds directly to Wingless and is a biphasic modulator of Wnt signaling. J Biol Chem 275:4374–4382 Caldwell GM, Jones C, Gensberg K, Jan S, Hardy RG, Byrd P, Chughtai S, Wallis Y, Matthews GM, Morton DG, 2004, The Wnt antagonist sFRP1 in colorectal tumorigenesis. Cancer Res 64:883–888 Weibel ER, 1979, Stereological methods, vol. 1, practical methods for biological morphometry. Academic, London, pp 1–415 Wicksell SD, 1925, The corpuscle problem. A mathematical study of a biometric problem. Biometrika 17:84–99 Wicksell SD, 1926, The corpuscle problem. Second memoir. Case of ellipsoidal corpuscles. Biometrika 18:151–172 Shi Y, He B, You L, Jablons DM, 2007, Roles of secreted frizzledrelated proteins in cancer. Acta Pharmacol Sin 28(9):1499–1504 Gumz ML, Zou H, Kreinest PA, Childs AC, Belmonte LS, LeGrand SN, Wu KJ, Luxon BA, Sinha M, Parker AS, Sun LZ, Ahlquist DA, Wood CG, Copland JA, 2007, Secreted frizzledrelated protein 1 loss contributes to tumor phenotype of clear cell renal cell carcinoma. Clin Cancer Res 13:4740–4749 Dahl E, Wiesmann F, Woenckhaus M, Stoehr R, Wild PJ, Veeck J, Knuchel R, Klopocki E, Sauter G, Simon R, Wieland WF, Walter B, Denzinger S, Hartmann A, Hammerschmied CG, 2007, Frequent loss of SFRP1 expression in multiple human solid tumours: association with aberrant promoter methylation in renal cell carcinoma. Oncogene 26:5680–5691 Awakura Y, Nakamura E, Ito N, Kamoto T, Ogawa O, 2008, Methylation-associated silencing of SFRP1 in renal cell carcinoma. Oncol Rep 20:1257–1263 Rubin JS, Barshishat-Kupper M, Feroze-Merzoug F, Xi ZF, 2006, Secreted WNT antagonists as tumor suppressors: pro and con. Front Biosci 11:2093–2105 Saini S, Liu J, Yamamura S, Majid S, Kawakami K, Hirata H, Dahiya R, 2009, Functional significance of secreted frizzledrelated protein 1 in metastatic renal cell carcinomas. Cancer Res 69(17):6815–6822 Joesting MS, Perrin S, Elenbaas B, Fawell SE, Rubin JS, Franco OE, Hayward SW, Cunha GR, Marker PC, 2005, Identification of SFRP1 as a candidate mediator of stromal-to-epithelial signaling in prostate cancer. Cancer Res 65(22):10423–10430 Peichel CL, Kozak CA, Luyten FP, Vogt TF, 1998, Evaluation of mouse Sfrp3/Frzb1 as a candidate for the lst, Ul, and Far mutants on chromosome 2. Mamm Genome 9(5):385–387 Zi X, Guo Y, Simoneau AR, Hope C, Xie J, Holcombe RF, Hoang BH, 2005, Expression of Frzb/secreted Frizzled-related protein 3, a secreted Wnt antagonist, in human androgen-independent prostate cancer PC-3 cells suppresses tumor growth and cellular invasiveness. Cancer Res 65(21):9762–9770 Pecina-Slaus N, Nikuseva Martic T, Deak AJ, Zeljko M, Hrascan R, Tomas D, Musani V, 2010, Genetic and protein changes of Ecadherin in meningiomas. J Cancer Res Clin Oncol 136(5):695–702 Hirata H, Hinoda Y, Ueno K, Majid S, Saini S, Dahiya R, 2010, Role of secreted frizzled-related protein 3 in human renal cell carcinoma. Cancer Res 70(5):1896–1905 Lustig B, Behrens J, 2003, The Wnt signaling pathway and its role in tumor development. J Cancer Res Clin Oncol 129(4):199–221 van de Wetering M, Cavallo R, Dooijes D, van Beest M, van Es J, Loureiro J, Ypma A, Hursh D, Jones T, Bejsovec A, Peifer M, Mortin M, Clevers H, 1997, Armadillo coactivates transcription driven by the product of the drosophila segment polarity gene dTCF. Cell 88(6):789–799 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 545 EP 551 DI 10.1007/s12253-013-9615-3 PG 7 ER PT J AU Perez, OL Crivaro, A Barbisan, G Poleri, L Golijow, DC AF Perez, Orlando Luis Crivaro, Andrea Barbisan, Gisela Poleri, Lucia Golijow, Daniel Carlos TI XRCC2 R188H (rs3218536), XRCC3 T241M (rs861539) and R243H (rs77381814) Single Nucleotide Polymorphisms in Cervical Cancer Risk SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; HPV; XRCC2; XRCC3; Single Nucleotide Polymorphism ID Cervical cancer; HPV; XRCC2; XRCC3; Single Nucleotide Polymorphism AB Human Papillomavirus (HPV) is the main cause of cervical cancer and its precursor lesions. Transformation may be induced by several mechanisms, including oncogene activation and genome instability. Individual differences in DNA damage recognition and repair have been hypothesized to influence cervical cancer risk. The aim of this study was to evaluate whether the double strand break gene polymorphisms XRCC2 R188H G>A (rs3218536), XRCC3 T241M C>T (rs861539) and R243H G>A (rs77381814) are associated to cervical cancer in Argentine women. A case control study consisting of 322 samples (205 cases and 117 controls) was carried out. HPV DNA detection was performed by PCR and genotyping of positive samples by EIA (enzyme immunoassay). XRCC2 and 3 polymorphisms were determined by pyrosequencing. The HPV-adjusted odds ratio (OR) of XRCC2 188 GG/AG genotypes was OR=2.4 (CI=1.1–4.9, p=0.02) for cervical cancer. In contrast, there was no increased risk for cervical cancer with XRCC3 241 TT/CC genotypes (OR=0.48; CI=0.2–1; p=0.1) or XRCC3 241 CT/CC (OR=0.87; CI=0.52–1.4; p=0.6). Regarding XRCC3 R243H, the G allele was almost fixed in the population studied. In conclusion, although the sample size was modest, the present data indicate a statistical association between cervical cancer and XRCC2 R188H polymorphism. Future studies are needed to confirm these findings. C1 [Perez, Orlando Luis] University of La Plata, Faculty of Veterinary Genetics, IGEVET-CONICET (Institute of Veterinary Genetics-National Council of Scientific and Technical Investigations), 60th and 118th Street, PC B1900 La Plata, Buenos Aires, Argentina. [Crivaro, Andrea] University of La Plata, Faculty of Veterinary Genetics, IGEVET-CONICET (Institute of Veterinary Genetics-National Council of Scientific and Technical Investigations), 60th and 118th Street, PC B1900 La Plata, Buenos Aires, Argentina. [Barbisan, Gisela] University of La Plata, Faculty of Veterinary Genetics, IGEVET-CONICET (Institute of Veterinary Genetics-National Council of Scientific and Technical Investigations), 60th and 118th Street, PC B1900 La Plata, Buenos Aires, Argentina. [Poleri, Lucia] University of La Plata, Faculty of Veterinary Genetics, IGEVET-CONICET (Institute of Veterinary Genetics-National Council of Scientific and Technical Investigations), 60th and 118th Street, PC B1900 La Plata, Buenos Aires, Argentina. [Golijow, Daniel Carlos] University of La Plata, Faculty of Veterinary Genetics, IGEVET-CONICET (Institute of Veterinary Genetics-National Council of Scientific and Technical Investigations), 60th and 118th Street, PC B1900 La Plata, Buenos Aires, Argentina. RP Perez, OL (reprint author), University of La Plata, Faculty of Veterinary Genetics, IGEVET-CONICET (Institute of Veterinary Genetics-National Council of Scientific and Technical Investigations), PC B1900 La Plata, Argentina. EM perezlo@gmail.com CR Wright TC, 2009, Natural history of HPV infections. J Fam Pract 58:3–7 Magnusson P, Lichtenstein P, Gyllensten U, 2000, Heritability of cervical tumours. Int J Cancer 88:698–701 Barbisan G, Perez LO, Difranza L, Fernandez CJ, Ciancio NE, Golijow CD, 2011, XRCC1 Arg399Gln polymorphism and risk for cervical cancer development in Argentine women. Eur J Gynaecol Oncol 32:274–279 Huang J, Ye F, Chen H, Lu W, Xie X, 2007, The nonsynonymous single nucleotide polymorphisms of DNA repair gene XRCC1 and susceptibility to the development of cervical carcinoma and high-risk human papillomavirus infection. Int J Gynecol Cancer 17:668–675 Roszak A, Lianeri M, Jagodzinski PP, 2011, Involvement of the XRCC1 Arg399Gln gene polymorphism in the development of cervical carcinoma. Int J Biol Markers 26(4):216–220 Settheetham-IshidaW, Yuenyao P, Natphopsuk S, Settheetham D, Ishida T, 2011, Genetic risk of DNA repair gene polymorphisms, XRCC1 and XRCC3, for high risk human papillomavirus negative cervical cancer in Northeast Thailand. Asian Pac J Cancer Prev 12:963–966 Thacker J, 2005, The RAD51 gene family, genetic instability and cancer. Cancer Lett 219:125–135 Udumudi A, Jaiswal M, Rajeswari N et al, 1998, Risk assessment in cervical dysplasia patients by single cell gel electrophoresis assay: a study of DNA damage and repair. Mutat Res 30:195–205 Cortes-Gutierrez EI, Davila-Rodriguez MI, Fernandez JL et al, 2011, DNA damage in women with cervical neoplasia evaluated by DNA breakage detection-fluorescence in situ hybridization. Anal Quant Cytol Histol 33:175–181 Duensing S, Munger K, 2002, The human papillomavirus type 16 E6 and E7 oncoproteins independently induce numerical and structural chromosome instability. Cancer Res 62:7075–7082 Kadaja M, Sumerina A, Verst T, Ojarand M, Ustav E et al, 2007, Genomic instability of the host cell induced by the human papillomavirus replication machinery. EMBO J 26:2180–2191 Kadaja M, Sumerina A, Verst T, Ojarand M, Ustav E, Ustav M, 2007, Genomic instability of the host cell induced by the human papillomavirus replication machinery. EMBO J 26:2180–2191 Kadaja M, Silla T, Ustav E, Ustav M, 2009, Papillomavirus DNA replication - frominitiation to genomic instability. Virology 384:360–368 Kadaja M, Isok-Paas H, Laos T, Ustav E, Ustav M, 2009, Mechanism of genomicinstability in cells infected with the highrisk human papillomaviruses. PLoSPathog 5:e1000397 Vineis P, Manuguerra M, Kavvoura FK, Guarrera S, Allione A, Rosa F et al, 2009, A field synopsis on low-penetrance variants in DNA repair genes and cancer susceptibility. J Natl Cancer Inst 101:24–36 Rafii S, O’Regan P, Xinarianos G, Azmy I, Stephenson T, Reed M et al, 2002, A potential role for the XRCC2 R188H polymorphic site in DNA-damage repair and breast cancer. Hum Mol Genet 11:1433–1438 Loizidou MA, Michael T, Neuhausen SL, Newbold RF, Marcou Y, Kakouri E et al, 2008, Genetic polymorphisms in the DNA repair genes XRCC1, XRCC2 and XRCC3 and risk of breast cancer in Cyprus. Breast Cancer Res Treat 112:575–579 Romanowicz-Makowska H, Smolarz B, Zadrozny M, Westfal B, Baszczynski J, Polac I, Sporny S, 2011, Single nucleotide polymorphisms in the homologous recombination repair genes and breast cancer risk in polish women. Tohoku J Exp Med 224:201–208 Rafii S, Lindblom A, Reed M, Meuth M, Cox A, 2003, A naturally occurring mutation in an ATP-binding domain of the recombination repair gene XRCC3 ablates its function without causing cancer susceptibility. Hum Mol Genet 12:915–923 De Ruyck K, Van Eijkeren M, Claes K, Morthier R, De Paepe A, Vral A et al, 2005, Radiation-induced damage to normal tissues after radiotherapy in patients treated for gynecologic tumors: association with single nucleotide polymorphisms in XRCC1, XRCC3, and OGG1 genes and in vitro chromosomal radiosensitivity in lymphocytes. Int J Radiat Oncol Biol Phys 15:1140–1149 He X, Ye F, Zhang J, Cheng Q, Shen J, Chen H, 2008, Susceptibility of XRCC3, XPD, and XPG genetic variants to cervical carcinoma. Pathobiology 75:356–363 Wang SS, Bratti MC, Rodriguez AC, Herrero R, Burk RD, Porras C et al, 2009, Common variants in immune and DNA repair genes and risk for human papillomavirus persistence and progression to cervical cancer. J Infect Dis 199:20–30 Miller SA, Dykes DD, Polesky HF, 1988, A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16:1215 Ting Y, Manos MM, 1990, Detection and typing of genital human Papillomavirus. In: Innis MA, Gelfand DH, Sninsky JJ, White TJ, eds, PCR protocols. Academic, San Diego, p 356 Evander M, Edlund K, Bodun E et al, 1992, Comparison of a onestep and two-step polimerase chain reaction with degenerate general primers in a population-based study of human Papillomavirus infection in young Swedish women. J Clin Microbiol 30:987–992 Soderlund-Strand A, Rymark P, Andersson P et al, 2005, Comparison between the Hybrid Capture II Test and a PCRBased Human Papillomavirus detection method for diagnosis and posttreatment follow-up of cervical intraepithelial neoplasia. J ClinMicrobiol 43:3260–3266 Kuschel B, Auranen A, McBride S, Novik KL, Antoniou A, Lipscombe JM et al, 2002, Variants in DNA double-strand break repair genes and breast cancer susceptibility. Hum Mol Genet 11:1399–1407 Jiao L, Hassan MM, Bondy ML, Wolff RA, Evans DB, Abbruzzese JL et al, 2008, XRCC2 and XRCC3 gene polymorphism and risk of pancreatic cancer. Am J Gastroenterol 103:360–367 Han J, Hankinson SE, Zhang SM, De Vivo I, Hunter DJ, 2004, Interaction between genetic variations in DNA repair genes and plasma folate on breast cancer risk. Cancer Epidemiol Biomarkers Prev 13:520–524 Pearce CL, Near AM, Van Den Berg DJ, Ramus SJ, Gentry- Maharaj A, Menon U et al, 2009, Validating genetic risk associations for ovarian cancer through the International Ovarian Cancer Association Consortium. Br J Cancer 100:412–20, Erratum in: Cunningham JC, 2009, Br J Cancer 101:1805 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 553 EP 558 DI 10.1007/s12253-013-9616-2 PG 6 ER PT J AU Brankovic, SA Brajuskovic, NG Mircetic, DJ Nikolic, ZZ Kalaba, BP Vukotic, DV Tomovic, MS Cerovic, JS Radojicic, AZ Savic-Pavicevic, LD Romac, PS AF Brankovic, S Ana Brajuskovic, N Goran Mircetic, D Jovan Nikolic, Z Zorana Kalaba, B Predrag Vukotic, D Vinka Tomovic, M Sasa Cerovic, J Snezana Radojicic, A Zoran Savic-Pavicevic, L Dusanka Romac, P Stanka TI Common Variants at 8q24 are Associated with Prostate Cancer Risk in Serbian Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Association study; Polymorphism; Single nucleotide polymorphism (SNP); 8q24 ID Prostate cancer; Association study; Polymorphism; Single nucleotide polymorphism (SNP); 8q24 AB Previous studies have shown correlation between single nucleotide polymorphisms (SNPs) at 8q24 and prostate cancer (PCa) risk. This study aimed to evaluate possible association between genotypes and alleles of 8q24 polymorphisms (rs1447295, rs4242382, rs6983267, rs7017300, and rs7837688) and PCa risk and progression. 150 patients with PCa, 150 patients with benign prostatic hyperplasia (BPH), and 100 healthy controls selected from the general population were recruited for this study. SNPs were genotyped by using PCR-RFLP analysis. There was a significant positive association between the A allele of the SNP rs4242382 and PCa risk [PCa vs. BPH comparison, P=0.014 for the best-fitting dominant model; odds ratio (OR) =1.98; 95 % confidence interval (95%CI) 1.14–3.43].We found evidence (P=0.0064) of association between PCa risk and rs7017300 (heterozygote OR= 1.60; 95%CI 0.95–2.69) when comparing genotype distributions in PCa and BPH patients. The association between T allele rs7837688 and PCa riskwas determined in PCa vs. BPH comparison with the best-fitting model of inheritance being log-additive (P=0.0033; OR=2.14, 95%CI 1.27–3.61). Odds ratio for carriers of rs6983267 TT genotype under recessive model of association with PCa was found to be 0.36 (PCa vs. control comparison, P=0.0029; 95%CI 0.19–0.71). For rs1447295, deviation from Hardy-Weinberg equilibrium was observed in BPH patients and controls. We found no association between parameters of PCa progression and five 8q24 SNPs. Locus 8q24 harbors genetic variants associated with PCa risk in Serbian population. C1 [Brankovic, S Ana] University of Belgrade, Faculty of BiologyBelgrade, Serbia. [Brajuskovic, N Goran] University of Belgrade, Faculty of BiologyBelgrade, Serbia. [Mircetic, D Jovan] University of Belgrade, Faculty of BiologyBelgrade, Serbia. [Nikolic, Z Zorana] University of Belgrade, Faculty of BiologyBelgrade, Serbia. [Kalaba, B Predrag] University of Belgrade, Faculty of BiologyBelgrade, Serbia. [Vukotic, D Vinka] Clinical Centre, Department of UrologyBelgrade, Serbia. [Tomovic, M Sasa] Clinical Centre, Clinical Department of SurgeryBelgrade, Serbia. [Cerovic, J Snezana] Military Medical Academy, Institute of PathologyBelgrade, Serbia. [Radojicic, A Zoran] University of Belgrade, Faculty of Organizational ScienceBelgrade, Serbia. [Savic-Pavicevic, L Dusanka] University of Belgrade, Faculty of BiologyBelgrade, Serbia. [Romac, P Stanka] University of Belgrade, Faculty of BiologyBelgrade, Serbia. RP Brajuskovic, NG (reprint author), University of Belgrade, Faculty of Biology, Belgrade, Serbia. EM brajuskovic@bio.bg.ac.rs CR Hamano T, Matsui H, Sekine Y, Ohtake N, Nakata S, Suzuki K, 2010, Association of SNP rs1447295 and microsatellite marker DG8S737 with familial prostate cancer and high grade disease. J Urol 184:738–742 Chang YM, Kung HJ, Evans C, 2007, Nonreceptor tyrosine kinases in prostate cancer. Neoplasia 9:90–100 Terada N, Tsuchiya N, Ma Z, Shimizu Y, Kobayashi T, Nakamura E et al, 2008, Association of genetic polymorphisms at 8q24 with the risk of prostate cancer in a Japanese population. Prostate 68:1689–1695 Schulz W, Hatina J, 2006, Epigenetics of prostate cancer: beyond DNA methylation. J Cell Mol Med 10:100–125 Vrdoljak E, Wojtukiewicz MZ, Pienkowski T, Bodoky G, Berzinec P, Finek J et al, 2011, South Eastern European Research Oncology Group. Cancer epidemiology in Central, South and Eastern European countries. Croat Med J 52:478–487 Cancer Registry of Central Serbia, Institute of Public Health of the Republic of Serbia 1999–2009. Available at: http://www.batut.org.rs/ Accessed: March 2, 2012 Wasserman NF, Aneas I, Nobrega MA, 2010, An 8q24 gene desert variant associated with prostate cancer risk confers differential in vivo activity to a MYC enhancer. Genome Res 20:1191–1197 Zheng SL, Sun J, Wiklund F, Smith S, Stattin P, Li G et al, 2008, Cumulative association of five genetic variants with prostate cancer. N Engl J Med 358:910–919 Liu H, Wang B, Han C, 2011, Meta-analyses of genome-wide and replication association studies on prostate cancer. Prostate 71:209–224 Amundadottir LT, Sulem P, Gudmundsson J, Helgason A, Baker A, Agnarsson BA et al, 2006, A common variant associated with prostate cancer in European and African populations. Nat Genet 38:652–658 Wang L, McDonnell SK, Slusser JP, Hebbring SJ, Cunningham JM, Jacobsen SJ et al, 2007, Two common chromosome 8q24 variants are associated with increased risk for prostate cancer. Cancer Res 67:2944–2950 Gleason DF, Mellinger GT, 1974, Prediction of prognosis for prostatic adenocarcinoma by combined histological grading and clinical staging. J Urol 111:58–64 Catalona WJ, Hudson MA, Scardino PT, Richie JP, Ahmann FR, Flanigan RC et al, 1994, Selection of optimal prostate specific antigen cutoffs for early detection of prostate cancer: receiver operating characteristic curves. J Urol 152:2037–2042 D’Amico AV, Whittington R, Malkowicz SB, Schultz D, Blank K, Broderick GA et al, 1998, Biochemical outcome after radical prostatectomy, external beam radiation therapy, or interstitial radiation therapy for clinically localized prostate cancer. JAMA 280:969–974 Medeiros RM, Morais A, Vasconcelos A, Costa S, Pinto D, Oliveira J et al, 2002, Outcome in prostate cancer: association with endothelial nitric oxide synthase Glu-Asp298 polymorphism at exon 7. Clin Cancer Res 8:3433–3437 Wallis Y, Morrell N, 2011, Automated DNA sequencing. Methods Mol Biol 688:173–185 Dianat SS, Margreiter M, Eckersberger E, Finkelstein J, Kuehas F, Herwig R et al, 2009, Gene polymorphisms and prostate cancer: the evidence. BJU Int 104:1560–1572 Zheng SL, Sun J, Cheng Y, Li G, Hsu FC, Zhu Y et al, 2007, Association between two unlinked loci at 8q24 and prostate cancer risk among European Americans. J Natl Cancer Inst 99:1525–1533 Freedman ML, Haiman CA, Patterson N, McDonald GJ, Tandon A, Waliszewska A et al, 2006, Admixture mapping identifies 8q24 as a prostate cancer risk locus in African-American men. Proc Natl Acad Sci USA 103:14068–14073 Chen M, Huang YC, Ko IL, Yang S, Chang YH, Huang W et al, 2009, The rs1447295 at 8q24 is a risk variant for prostate cancer in Taiwanese men. J Urol 74:698–701 Schumacher FR, Feigelson HC, Cox DG, Haiman CA, Albanes D, Buring J et al, 2007, A common 8q24 variant in prostate and breast cancer from a large nested case–control study. Cancer Res 67:2951–2956 Salinas CA, Kwon E, Carlson CS, Koopmeiners JS, Feng Z, Karyadi DM et al, 2008, Multiple independent genetic variants in the 8q24 region are associated with prostate cancer risk. Cancer Epidemiol Biomark Prev 17:1203–1213 Penney KL, Salinas CA, Pomerantz M, Schumacher FR, Beckwith CA, Lee GS et al, 2009, Evaluation of 8q24 and 17q risk loci and prostate cancer mortality. Clin Cancer Res 15:3223–3230 Okobia MN, Zmuda JM, Ferrell RE, Patrick AL, Bunker CH, 2011, Chromosome 8q24 variants are associated with prostate cancer risk in a high risk population of African ancestry. Prostate 71:1054–1063 Cussenot O, Azzouzi AR, Bantsimba-Malanda G, Gaffroy C, Mangin P, Cormier L et al, 2008, Effect of genetic variability within 8q24 on aggressiveness patterns at diagnosis and familial status of prostate cancer. Clin Cancer Res 14:5635–5639 Troutman SM, Sissung TM, Cropp CD, Venzon DJ, Spencer SD, Adesunloye BA et al, 2012, Racial disparities in the association between variants on 8q24 and prostate cancer: a systematic review and meta-analysis. Oncologist 17:312–320 Suuriniemi M, Agalliu I, Schaid DJ, Johanneson B, McDonnell SK, Iwasaki L et al, 2007, Confirmation of a positive association between prostate cancer risk and a locus at chromosome 8q24. Cancer Epidemiol Biomark Prev 16:809–814 Os’kina NA, Boiarskikh UA, Lazarev AF, Petrova VD, Ganov DI, Tonacheva OG et al, 2012, Association of chromosome 8q24 variants with prostate cancer risk in the Siberian region of Russia and meta-analysis. Mol Biol, Mosk, 46:234–241 Joung JY, Park S, Yoon H, Lee SJ, Park WS, Seo HK et al, 2012, Association of common variations of 8q24 with the risk of prostate cancer in Koreans and a review of the Asian population. BJU Int 110:E318–E325 Wokolorczyk D, Gliniewicz B, Stojewski M, Sikorski A, Zlowocka E, Debniak T et al, 2010, The rs1447295 and DG8S737 markers on chromosome 8q24 and cancer risk in the Polish population. Eur J Cancer Prev 19:167–171 Zeegers MP, Khan HS, Schouten LJ, van Dijk B, Goldbohn AR, Shalken J et al, 2010, Genetic marker polymorphisms on chromosome 8q24 and prostate cancer in the Dutch population: DG8S737 may not be the causative variant. Eur J Hum Genet 19:118–120 Robbins C, Torres JB, Hooker S, Bonilla C, Hernandez W, Candreva A et al, 2007, Confirmation study of prostate cancer risk variants at 8q24 in African Americans identifies a novel risk locus. Genome Res 17:1717–1722 Murphy AB, Ukoli F, Freeman V, Bennett F, AikenW, Tulloch T et al, 2012, 8q24 risk alleles in West African and Caribbean men. Prostate 72:1366–1373 Pal P, Xi H, Guha S, Sun G, Helfand BT, Meeks JJ et al, 2009, Common variants in 8q24 are associated with risk for prostate cancer and tumor aggressiveness in men of European ancestry. Prostate 69:1548–1556 Beuten J, Gelfond JA, Martinez-Fierro ML, Weldon KS, Crandall AC, Rojas-Martinez A et al, 2009, Association of chromosome 8q variants with prostate cancer risk in Caucasian and Hispanic men. Carcinogenesis 30:1372–1379 Bao BY, Pao JB, Lin VC, Huang CN, Chang TY, Lan YH et al, 2010, Individual and cumulative association of prostate cancer susceptibility variants with clinicopathologic characteristics of the disease. Clin Chim Acta 411:1232–1237 Fitzgerald LM, Kwon EM, Koopmeiners JS, Salinas CA, Standford JL, Ostrander EA, 2009, Analysis of recently identified prostate cancer susceptibility loci in a population-based study: associations with family history and clinical features. Clin Cancer Res 15:3231–3237 Yeager M, Xiao N, Hayes RB, Bouffard P, Desany B, Burdet L et al, 2008, Comprehensive resequence analysis of a 136 kb region of human chromosome 8q24 associated with prostate and colon cancers. Hum Genet 124:161–170 Sun J, Lange EM, Isaacs SD, Liu W, Wiley KE, Lange L et al, 2008, Chromosome 8q24 risk variants in hereditary and nonhereditary prostate cancer patients. Prostate 68:489–497 Xu Z, Bensen JT, Smith GJ, Mohler JL, Taylor JA, 2011, GWAS SNP replication among African American and European American men in the North Carolina—Louisiana prostate cancer project. Prostate 71:881–891 Chan JY, Li H, Singh O, Mahajan A, Ramasamy S, Subramaniyan K, et al., 2012, 8q24 and 17q prostate cancer susceptibility loci in a multiethnic Asian cohort. Urol Oncol 1–8 Papanikolopoulou A, Landt O, Ntoumas K, Bolomitis S, Tyritzis SI, Constantinides C et al, 2012, The multi-cancer marker, rs6983267, located at region 3 of chromosome 8q24, is associated with prostate cancer in Greek patients but does not contribute to the aggressiveness of the disease. Clin Chem Lab Med 50:379–385 Cochran WG, 1954, Some methods for strengthening the common χ2 tests. Biometrics 10:417–451 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 559 EP 569 DI 10.1007/s12253-013-9617-1 PG 11 ER PT J AU Literati-Nagy, Zs Tory, K Literati-Nagy, B Bajza, Vigh, L Vigh, L Mandl, J Szilvassy, Z AF Literati-Nagy, Zsuzsanna Tory, Kalman Literati-Nagy, Botond Bajza, Agnes Vigh, Laszlo Vigh, Laszlo Mandl, Jozsef Szilvassy, Zoltan TI Synergetic Insulin Sensitizing Effect of Rimonabant and BGP-15 in Zucker-Obes Rats SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Rimonabant; BGP-15; Insulin resistance; Obesity; Capsaicin; Glucose clamp ID Rimonabant; BGP-15; Insulin resistance; Obesity; Capsaicin; Glucose clamp AB Abdominal obesity is referred for as a common pathogenic root of multiple risk factors, which include insulin resistance, dyslipidemia, hypertension, and a pro-atherogenic and pro-inflammatory state. Irrespective of its psychiatric side effects, rimonabant through blocking cannabinoid-1 receptor (CB1R) induces an increase in whole body insulin sensitivity. The aim of this work was to study the effect of selected doses of another insulin sensitizer compound BGP-15, and rimonabant on insulin resistance in Zucker obese rats with a promise of inducing insulin sensitization together at lower doses than would have been expected by rimonabant alone. We found that BGP-15 potentiates the insulin sensitizing effect of rimonabant. The combination at doses, which do not induce insulin sensitization by themselves, improved insulin signaling. Furthermore our results suggest that capsaicin-induced signal may play a role in insulin sensitizing effect of both molecules. Our data might indicate that a lower dose of rimonabant in the treatment of insulin resistance and type 2 diabetes is sufficient to administer, thus a lower incidence of the unfavorable psychiatric side effects of rimonabant are to be expected. C1 [Literati-Nagy, Zsuzsanna] Semmelweis University, Department of Medical Chemistry, Molecular Biology and PathobiochemistryBudapest, Hungary. [Tory, Kalman] N-Gene Research and Development LtdBudapest, Hungary. [Literati-Nagy, Botond] Drug Research Center LtdBalatonfured, Hungary. [Bajza, Agnes] N-Gene Research and Development LtdBudapest, Hungary. [Vigh, Laszlo] Mecsek Pharma Research LtdPecs, Hungary. [Vigh, Laszlo] Hungarian Academy of Sciences, Biological Research CenterSzeged, Hungary. [Mandl, Jozsef] Semmelweis University, Department of Medical Chemistry, Molecular Biology and PathobiochemistryBudapest, Hungary. [Szilvassy, Zoltan] University of Debrecen, Department of Pharmacology and Pharmacotherapy, 4032 Debrecen, Hungary. RP Tory, K (reprint author), N-Gene Research and Development Ltd, Budapest, Hungary. EM trklmn@gmail.com CR Obesity and overweight WHO fact sheet N°311, 2012) Howlett AC, Breivogel CS, Childers SR, Deadwyler SA, Hampson RE, Porrino LJ, 2004, Cannabinoid physiology and pharmacology: 30 years of progress. Neuropharmacology 47(Suppl 1):345–358 Scheen AJ, Finer N, Hollander P, Jensen MD, Van Gaal LF, RIODiabetes Study Group, 2006, Efficacy and tolerability of rimonabant in overweight or obese patients with type 2 diabetes: a randomised controlled study. Lancet 368:1660–1672 Literati-Nagy B, Kulcsar E, Literati-Nagy Z, Buday B, Petrfai E, Horvath T, Tory K, Kolonics A, Fleming A, Mandl J, Koranyi L, 2009, Improvement of insulin sensitivity by a novel drug, BGP- 15, in insulin-resistant patients: a proof of concept randomized double-blind clinical trial. Horm Metab Res 41:374–380 Tim C, Noemi T, Stefano P, Peter L-N, Kalman T, Pierre H, Bernadett K, Linda G, Zsolt T, Gabor B, Imre G, Federica C, Simona C, Ferenc G, Gabor N, Zoltan B, Burcin G, Maria P, Attila G, Akos H, Zsuzsanna L-N, Vigh L Jr, Femke H-B, Andre H, Irma K, Lizette L, Jean-Paul S, Deli Z, Meijering RAM, Henning RH, Brundel BJJM, Kampinga HH, Laszlo K, Zoltan S, Jozsef M, Balazs S, Febbraio MA, Ibolya H, Hooper PL, Laszlo V, 2013, Hydroximic acid derivatives: pleiotrophic HSP coinducers restoring homeostasis and robustness. Curr Pharm Des 19(00) Literati-Nagy Z, Tory K, Literati-Nagy B, Kolonics A, Vigh L Jr, Vigh L, Mandl J, Szilvassy Z, 2012, A novel insulin sensitizer drug candidate-BGP-15-can prevent metabolic side effects of atypical antipsychotics. Pathol Oncol Res 18(4):1071–1076 Gombos I, Crul T, Piotto S, Gungor B, Torok Z, Balogh G, Peter M, Slotte PJ, Pilbat AN, Hunya A, Toth N, Literati-Nagy Z, Vigh L Jr, Glatz A, Brameshuber M, Schutz GJ, Hevener A, Febbraio MA, Horvath I, Vigh L, 2011)Membrane-lipid therapy in operation: the HSP co-inducer BGP-15 activates stress signal transduction pathways by remodeling plasma membrane rafts. PLoS One 6(12):e28818 Chung J, Nguyen AK, Henstridge DC, Holmes AG, Chan MH, Mesa JL, Lancaster GI, Southgate RJ, Bruce CR, Duffy SJ, Horvath I, Mestril R, Watt MJ, Hooper PL, Kingwell BA, Vigh L, Hevener A, Febbraio MA, 2008, HSP72 protects against obesity-induced insulin resistance. Proc Natl Acad Sci USA 105:1739–1744 Literati-Nagy Z, Tory K, Literati-Nagy B, Kolonics A, Torok Z, Gombos I, Balogh G, Vigh L Jr, Horvath I, Mandl J, Sumegi B, Hooper PL, Vigh L, 2012, The HSP co-inducer BGP-15 can prevent the metabolic side effects of the atypical antipsychotics. Cell Stress Chaperones 17(4):517–521 Guillot E, Coste A, Angel I, 1996, Involvement of capsaicinsensitive nerves in the regulation of glucose tolerance in diabetic rats. Life Sci 59(12):969–977 Porszasz R, Legvari G, Pataki T, Szilvassy J, Nemeth J, Kovacs P, Paragh G, Szolcsanyi J, Szilvassy Z, 2002, Sensory nitrergic nature of the hepatic insulin sensitizing substance mechanism in conscious rabbits. Eur J Pharmacol 443(1–3):211–212 Wallenstein S, Zucker CL, Fleiss JL, 1980, Some statistical methods useful in circulation research. Circ Res 47(1):1–9 Schafer A, Pfrang J, Neumuller J, Fiedler S, Ertl G, Bauersachs J, 2008, The cannabinoid receptor-1 antagonist rimonabant inhibits platelet activation and reduces pro-inflammatory chemokines and leukocytes in Zucker rats. Br J Pharmacol 154(5):1047–1054 Szilvassy J, Sziklai I, Sari R, Nemeth J, Peitl B, Porszasz R, Lonovics J, Szilvassy Z, 2006, Neurogenic insulin resistance in guinea-pigs with cisplatin-induced neuropathy. Eur J Pharmacol 531(1–3):217–225 Hooper PL, Hooper JJ, 2005, Loss of defense against stress: diabetes and heat shock proteins. Diabetes Technol Ther 7:204–208 Hooper PL, Hooper PL, 2009, Inflammation, heat shock proteins, and type 2 diabetes. Cell Stress Chaperones 14:113–115 Weiss Y, Bromberg, P. Goloubinoff, 2009, Activation of the heat shock response by membrane dependent calcium channel receptors effect in inflammation and cancer Electronic poster Vienna Kang JH, Kim CS, Han IS, Kawada T, Yu R, 2007, Capsaicin, a spicy component of hot peppers, modulates adipokine gene expression and protein release from obese-mouse adipose tissues and isolated adipocytes, and suppresses the inflammatory responses of adipose tissue macrophages. FEBS Lett 581(23):4389–4396 Kunos G, Tam J, 2011, The case for peripheral CB1 receptor blockade in the treatment of visceral obesity and its cardiometabolic complications. Br J Pharmacol 163(7):1423–1431 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 571 EP 575 DI 10.1007/s12253-013-9620-6 PG 5 ER PT J AU Kanjer, K Tatic, S Neskovic-Konstantinovic, Z Abu Rabi, Z Nikolic-Vukosavljevic, D AF Kanjer, Ksenija Tatic, Svetislav Neskovic-Konstantinovic, Zora Abu Rabi, Zaki Nikolic-Vukosavljevic, Dragica TI Treatment Response to Preoperative Anthracycline-Based Chemotherapy in Locally Advanced Breast Cancer: The Relevance of Proliferation and Apoptosis Rates SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breastcancer; Primary chemotherapy; Ki-67; AI; Growth index ID Breastcancer; Primary chemotherapy; Ki-67; AI; Growth index AB Objectives were to evaluate the relevance of proliferating fraction (Ki-67) along with apoptotic index (AI) which denoted growth index (Ki-67/AI ratio, GI) to predict pathological response to preoperative chemotherapy, and the pattern of their modifications following chemotherapy in women with locally advanced breast cancer. Archivalmaterial of diagnostic biopsies and surgical specimens from 106 patients were examined. Response rate to chemotherapy in this group was 95 %, eight (8 %) patients achieved a pathological complete remission (pCR) and five (5 %) had a progressive/stable disease (PD/SD). The expression of Ki-67 and AI were assessed using immunohistochemistry and in situ DNA nick labeling assay respectively. Higher baseline level of Ki-67 and GI were associated with an improved pathological response (p=0.0001 and p=0.008), but the degree of correlation with GI was no greater than that with Ki-67 alone. Ki-67 below 1 % highly indicated a lack of tumor response. High AI which characterized the opposite chemo-sensitive tumors, pCR vs. PD/SD (p=0.72) implied that treatment response was not influenced by the "presence" or "absence" of apoptosis. A significant decrease in Ki-67 (p<0.001), AI (p =0.035) and GI (p=0.008) was found following chemotherapy, but percentage change in biomarker values revealed an increase in a number of cases. Higher initial Ki-67 and AI was associated with profound reduction of GI and raising value of GI after treatment, respectively. Such a variance of a given parameter elicited by chemotherapy may have various impact on disease outcome. C1 [Kanjer, Ksenija] Institute of Oncology and Radiology of Serbia, Department of Experimental Oncology, Pasterova 14Belgrade, Serbia. [Tatic, Svetislav] University of Belgrade, Faculty of Medicine, Institute for Pathology, Dr Subotica 1Belgrade, Serbia. [Neskovic-Konstantinovic, Zora] Institute of Oncology and Radiology of Serbia, Department of Medical Oncology, Pasterova 14Belgrade, Serbia. [Abu Rabi, Zaki] Institute of Oncology and Radiology of Serbia, Department of Experimental Oncology, Pasterova 14Belgrade, Serbia. [Nikolic-Vukosavljevic, Dragica] Institute of Oncology and Radiology of Serbia, Department of Experimental Oncology, Pasterova 14Belgrade, Serbia. RP Kanjer, K (reprint author), Institute of Oncology and Radiology of Serbia, Department of Experimental Oncology, Belgrade, Serbia. EM ksenijakanjer@gmail.com CR Hayes DF, 2005, Primary therapy of early breast cancer: 9th International St. Gallen consensus conference. Prognostic and predictive factors revisited. Breast 14:493–499 Goldhirsch A, Glick JH, Gelber RD, Coates AS, Thurlimann B, Senn HJ, Panel Members, 2005, Meeting highlights: international expert consensus on the primary therapy of early breast cancer 2005. Ann Oncol 16:1569–1583 Tewari M, Krishnamurthy A, Shukla H, 2008, Predictive markers of response to neoadjuvant chemotherapy in breast cancer. Surg Oncol 17:301–311 Hugh J, Hanson J, CheangMCU, Nielsen TO, Perou CM, Dumontet C et al, 2009, Breast cancer subtypes and response to docetaxel in node-positive breast cancer: use of an immunohistochemical definition in the BCIRG 001 Trial. J Clin Oncol 27:1168–1176 Albain KS, Barlow WE, Shak S, Hortobagyi GN, Livingston RB, Yeh IT et al, 2010, Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with nodepositive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis of a randomized trial. Lancet Oncol 11:55–65 Fisher B, Mamounas E, 1995, Preoperative chemotherapy: a model for studying the biology and therapy of primary breast cancer. J Clin Oncol 13:537–540 Loonning PE, 2003, Study of suboptimum treatment response: lessons from breast cancer. Lancet Oncol 4:177–185 Pomier Y, Sordet O, Antony S, Hayward RL, Kohn KW, 2004, Apoptosis defects and chemotherapy resistance: molecular interaction maps and networks. Oncogene 23:2934–2949 Johnstone RW, Ruefli AA, Lowe SW, 2002, Apoptosis: a link between cancer genetics and chemotherapy. Cell 108:153–164 Fulda S, Debatin KM, 2006, Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene 25:4798–4811 Tamm I, Schriever F, Dorken B, 2001, Apoptosis: implications of basis research for clinical oncology. Lancet Oncol 2:33–42 Harper-Wynne CL, Sacks NP, Shenton K, MacNeill FA, Sauven P, Laidlaw I et al, 2002, Comparision of the systemic and intratumoral effects of tamoxifen and the aromatase inhibitor vorozole in postmenopausal patients with primary breast cancer. J Clin Oncol 20:1026–1035 Cleator S, Parton M, Dowsett M, 2002, The biology of neoadjuvant therapy for breast cancer. Endoc Rel Cancer 9:183–195 Archer CD, Parton M, Smith IE, Ellis PA, Salter J, Ashley S et al, 2003, Early changes in apoptosis and proliferation following primary chemotherapy for breast cancer. Br J Cancer 89:1035–1041 Scarff RF, Torloni H, 1981, Histological Typing of Breast Tumors. International Histological Classification of Tumors, No 2. Geneva, WHO Bloom HJG, Richardson WW, 1957, Histological grading and prognosis in breast cancer; a study of 1409 cases of which 359 have been followed for 15 years. Br J Cancer 11:337–359 Kaufmann M, Hortobagyi GN, Goldhirsch A, Scholl S, Makris A, Valaqussa P et al, 2006, Recommendations from an international expert panel on the use of neoajuvant, primary, systemic treatment of operable breast cancer: an update. J Clin Oncol 24:1940–1949 Sneige N, Kemp B, Pusztai L, Asmar L, Hortobagyi GN, 2001, Chemotherapy-induced histologic changes in mastectomy specimens and their potential significance. Breast 10:492–500 Gavrieli Y, Sherman Y, Ben-Sasson SA, 1992, Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. J Cell Biol 119:493–501 Bonadonna G, Valagussa P, Brambilla C, Ferrari L, Moliterni A, Terenziani M et al, 1998, Primary chemotherapy in operable breast cancer: eight year experiance at the Milan Cancer Institutte. J Clin Oncol 16:93–100 Guarneri V, Broglio K, Kau SW, Cristofanilli M, Buzdar AU, Valero V et al, 2006, Prognostic value of pathologic complete response after primary chemotherapy in relation to hormone receptor status and other factors. J Clin Oncol 24:1037–1044 Bertheau P, Lerebours F, Mounier N, de Roquancourt A, Espie M, Clot P et al, 2005, Prognostic significance of a combined clinicopathologic score for response to primary systemic therapy in locally advanced breast cancer. Oncol Rep 14:513–520 Smith IE, Lipton L, 2001, Preoperative/neoadjuvant medical therapy for early breast cancer. Lancet Oncol 2:561–570 Lipponen P, 1999, Apoptosis in breast cancer: relationship with other pathological parameters. Endoc Relat Cancer 6:13–16 Urruticoechea A, Smith IS, Dowset M, 2005, Proliferation marker Ki-67 in early breast cancer. J Cli Oncol 23:7212–7220 Ellis PA, Smith IE, Detre S, 1998, Reduced apoptosis and proliferation and increased bcl-2 levels in residual breast cancer following preoperative chemotherapy. Breast Cancer Res Treat 48:107–116 Remvikos Y, Beuzeboc P, Zajdela A, Voillemot N, Magdalenat H, Pouillart P, 1989, Correlation of pretreatment proliferative activity of breast cancer with the response to cytotoxic chemotherapy. J Nat Cancer Inst 20:1383–1387 Campiglio M, Somenzi G, Olgiati C, Beretta G, Balsari A et al, 2003, Role of proliferation in HER-2 status predicted response to doxorubicin. Int J Cancer 105:568–573 Collecchi P, Baldini E, Giannessi P, Naccarato AG, Passoni A, Gardin G et al, 1998, Primary chemotherapy in locally advanced breast cancer, LABC): effects on tumor proliferative activity, bcl-2 expression and the relationship between tumor regression and biological markers. Eur J Cancer 34:1701–1703 Wang J, Buchholz TA, Middleton LP, Allred DC, Tucker SL, Kuerer HM et al, 2002, Assessment of histologic features and expression of biomarkers in predicting pathologic response to anthracycline-based neoadjuvant chemotherapy in patients with breast carcinoma. Cancer 94:3107–3114 Pohl G, Rudas M, Taucher S, Stranzl T, Steger GG, Jakesz R et al, 2003, Expression of cell cycle regulatory proteins in breast carcinomas before and after preoperative chemotherapy. Breast Cnacer Res Treat 78:97–103 Faneyte IF, Schrama JG, Peterse JL, Remijnse PL, Rodenhuis S, van de Vijver MJ, 2003, Breast cancer response to neoadjuvant therapy: predictive markers and relation with outcome. Br J Cancer 88:406–412 Ogston KN, Miller ID, Schofield AC, Spyrantis A, Pavlidou E, Sarkar TK et al, 2004, Can patients to benefit from primary chemotherapy for breast cancer be predicted before commencement of treatment? Breast Cancer Res Treat 86:181–189 Vincent–Salomon A, Rousseau A, Jouve M, Beuzeboc P, Sigal- Zafrani B, Freneaux P et al, 2004, Proliferation markers predictive of the pathological response and disease outcome of patients with breast carcinomas treated by antracycline-based preoperative chemotherapy. Eur J Cancer 40:1502–1508 Prisack HB, Karreman C, Modlich O, Audretsch W, Danae M, Rezai M et al, 2005, Predictive biological markers for response of invasive breast cancer to anthracycline /cyclophosphamide-based primary, radio-)chemotherapy. Anticancer Res 25:4615–4621 Fasching PA, Heusinger L, Niklos M, Hein A, Bayer CM, Rauh C et al, 2011, Ki-67, chemotherapy response, and prognosis in breast cancer patients receiving neoadjuvant treatment. BMC Cancer 11:486–498 Penault-Llorca F, Abrial C, Raoelfils I, Chollet P, Cayre A, Mouret-Reynier M-A et al, 2008, Changes and predictive and prognostic value of the mitotic index, Ki-67, Cyclin D1, and Cyclo-oxigenase-2 in 710 operable breast cancer patients treated with neoadjuvant chemotherapy. Oncologist 13:1235–1245 Bozzetti C, Musolino A, Camisa R, Bisagni G, Flora M, Bassano C et al, 2006, Evaluation of HER-2/Neu Amplification and other biological markers as predictors of response to neoadjuvant anthracycline-based chemotherapy in primary breast cancer: the role of anthracycline dose intensity. Am J Clin Oncol 2:171–177 Bottini A, Berruti A, Bersiga A, Brizzi MP, Bruzzi P, Aguggini S et al, 2001, Relationship between tumour shrinkage and reduction in Ki-67 expression after primary chemotherapy in human breast cancer. Br J Cancer 85:1106–1112 Petit T, Wilt M, Millon R, Rodier JF, Borel C, Mors R et al, 2004, Comparative value of tumor grade, hormonal receptors, Ki-67, HER-2 and topoisomerase II alpha status as predictive markers in breast cancer patients treated with neoadjuvant anthracyclinebased chemotherapy. Eur J Cancer 40:205–211 Tordai A, Wang J, Andre F, Liedtke C, Yan K, Sotiriou C et al, 2008, Evaluation of biological pathways involved in chemotherapy response in breast cancer. Breast Cancer Res 10:R37 vonMinckwitz G, Sinn HP, Raab G, Loibl S, Blohmer JU, Eidtmann H et al, 2008, Clinical response after two cycles compared to HER2, Ki-67, p53, and bcl-2 in independently predicting a pathological complete response after preoperative chemotherapy in patients with operable carcinoma of the breast. Breast Cancer Res 10:R30 Lee J, Im YH, Lee SH, Cho EY, Choi YL, Ko YH et al, 2008, Evaluation of ER and Ki-67 proliferation index as prognostic factors for survival following neoadjuvant chemotherapy with doxorubicin/docetaxel for locally advanced breast cancer. Cancer Chemother Pharmacol 61:569–577 Burcombe RJ, Makris A, Richman PI, Daley FM, Noble S, Pittam M et al, 2005, Evaluation of ER, PR, HER-2 and Ki-67 as predictors of response to neoadjuvant antracycline chemotherapy for operable breast cancer. Br J Cancer 92:147–155 Burcombe RJ,Wilson GD, Dowsett M, Khan I, Richman PI, Daley F et al, 2006, Evaluation of Ki-67 proliferation and apoptotic index before, during and after neoadjuvant chemotherapy for primary breast cancer. Breast Cancer Res 8:R31 Arpino G, Ciocca DR, Weiss H, Allred DC, Daguerre P, Vargas- Roig L et al, 2005, Predictive value of apoptosis, proliferation, HER-2, and topoisomerase IIα for anthracycline chemotherapy in locally advanced breast cancer. Breast Cancer Res Treat 92:69–75 Kariya S, Ogawa Y, Nishioka A, Moriki T, Ohnishi T, Ito S et al, 2005, Relationship between hormonal receptors, HER-2, p53 protein, bcl-2, and MIB-1 status and the antitumor effects of neoadjuvant anthracycline-based chemotherapy in invasive breast cancer patients. Radiation Med 23:189–194 Castedo M, Perfettini JL, Roumier T, Andreau K, Medema R, Kroemer G, 2004, Cell death by mitotic catastrophe: a molecular definition. Oncogene 23:2825–2837 Shay JW, Roninson IB, 2004, Hallmarks of senensce in carcinogenesis and cancer therapy. Oncogene 23:2919–2933 Tiezzi DG, De Andrade JM, Candido Dos Reis FJ, Cosiski Marrana HR, Ribeiro-Silva A, Tiezzi MG et al, 2006, Apoptosis induced by neoadjuvant chemotherapy in breast cancer. Pathology 38:21–27 Rein DT, Schondorf T, Breidenbach M, Janat MM, Weikelt A, Gohring UJ et al, 2000, Lack of correlation between p53 expression, bcl-2 expression, apoptosis and ex vivo chemosensitivity in advanced human breast cancer. Anticancer Res 20:5069–5072 Buchholz TA, Davis DW, McConkey DJ, Symmans WF, Valero V, Jhingran A et al, 2003, Chemotherapy-induced apoptosis and bcl- 2 levels correlate with breast cancer response to chemotherapy. Cancer J 9:33–41 Vargas-Roig LM, Cuello-Carrion FD, Fernandez-Eskobar N, Daguerre P, Leuzzi M, Ibarra J et al, 2008, Prognostic value of bcl-2 in breast cancer patients treated with neoadjuvant anthracycline based chemotherapy. Molecular Oncol 2:102–111 Navarrete MAH, Maier CM, Falzoni R, de Azavedo Quadros LG, Lima GR, Baracat EC, Nazario ACP, 2005, Assessment of the proliferative, apoptotic and cellular renovation indices of the human mammary epithelium during the follicular and lutheal phases of the menstrual cycle. Breast Cancer Res 7:306–313 Aas T, Geisler S, Helle H, Borrensen-Dale AL, Lonning PE, Akslen LA, 2005, Prognostic and predictive value of changes in tumor cell proliferation in locally advanced breast cancer primarily treated with doxorubicin. Oncol Reports 13:525–530 Zunino F, Perego P, Pilotti S, Pratesi G, Supino R, Arcamone F, 1997, Role of apoptotic response in cellular resistance to cytotoxic agents. Pharmacol Ther 76:177–185 Frassoldati A, Adami F, Banzi C, Criscuolo M, Piccinini L, Silingardi V, 1997, Changes of biological features in breast cancer cells determined by primary chemotherapy. Breast Cancer Res Treat 44:185–192 Takada M, Kataoka A, Masazaku T, Bando H, Toyama K, Horiguchi S et al, 2004, A close association between alteration in growth kinetics by neoadjuvant therapy and survival outcome in primary breast cancer. Int J Oncol 25:397–406 Shao ZM, Li J, Wu J, Qi-Xia H, Zhen-Zoi S, Fontana JA, Barsky SH, 1999, Neo-adjuvant chemotherapy for operable breast cancer induces apoptosis. Breast Cancer Res Treat 53:263–269 Dowsett M, A’Hern RA, Salter J, Zabaglo L, Smith IE, 2009, Who would thought a single Ki-67 measurement would be predict longterm outcome? Breast Cancer Res 11:S15 Assersohn LA, Salter J, Powles TJ, A’hern R, Makris A, Gregory RK et al, 2003, Studies of potential utility of Ki-67 as a predictive molecular marker of clinical response in primary breast cancer. Breast Cancer Res Treat 82:113–123 Dowsett M, Smith IE, Ebbs SR, Dixon JM, Skene A, Griffith C et al, 2006, Proliferation and apoptosis as marker of benefit in neoadjuvant endocrine therapy of breast cancer. Clin Cancer Res 12:1024–1029 Jones RL, Salter J, A’Hern R, Nerurkar A, Parton M, Reis-Filho JS et al, 2009, The prognostic significance of Ki-67 before and after neoadjuvant chemotherapy in breast cancer. Breast Cancer Res Treat 116:53–68 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 577 EP 588 DI 10.1007/s12253-013-9621-5 PG 12 ER PT J AU Maraz, R Boross, G Ambrozay, Svebis, M Cserni, G AF Maraz, Robert Boross, Gabor Ambrozay, Eva Svebis, Mihaly Cserni, Gabor TI Selective Ductectomy for the Diagnosis and Treatment of Intraductal Papillary Lesions Presenting with Single Duct Discharge SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intraductal papilloma; Breast cancer; Ductography; Selective ductectomy ID Intraductal papilloma; Breast cancer; Ductography; Selective ductectomy AB Solitary ductal papilloma of the breast, although considered a benign disorder has a potential association with carcinomas. We studied and analyzed the role of selective ductectomy (SD) for the diagnosis and treatment of intraductal lesions presenting with single duct discharge and ductography suggestive of intraductal (papillary) lesions. During a ten-year-period, files of patients presenting with single (or rarely dual) duct discharge were retrospectively reviewed. The examinations included mammography, ductography and ultrasonography and cytology of the fluid discharged from the duct in all patients. Patients treated with SD were considered further and their histological diagnosis and treatment were analyzed. The series included 100 patients. In 6 cases malignancy was found in the specimen consisting of four in situ and two invasive ductal carcinomas. These 6 patients had a second operation and this was followed by adjuvant treatment. Nine further patients had atypical ductal hyperplasia in or around papillomas and one patient had lobular neoplasia around her papilloma. In the present series, the incidence of carcinoma associated with the clinical suspicion of papillary lesions was 6%, and further 10% had low grade neoplastic proliferations resulting in the diagnosis of atypical papillomas or atypical ductal hyperplasia or lobular neoplasia around the papilloma, indicating that single duct discharge may be a symptom a malignancy, and that ductal papillomas have malignant potential. For such a low risk and grade of malignancy simple follow-up could be one option, but in some cases SD could be applied to relieve the patients from symptoms and establish a diagnosis. C1 [Maraz, Robert] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38, 6000 Kecskemet, Hungary. [Boross, Gabor] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38, 6000 Kecskemet, Hungary. [Ambrozay, Eva] Bacs-Kiskun County Teaching Hospital, Breast Diagnostic UnitKecskemet, Hungary. [Svebis, Mihaly] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38, 6000 Kecskemet, Hungary. [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of PathologyKecskemet, Hungary. RP Maraz, R (reprint author), Bacs-Kiskun County Hospital, Department of Surgery, 6000 Kecskemet, Hungary. EM marazrobert2010@gmail.com CR Greif F, Sharon E, Shechtman I, Morgenstern S, Gutman H, 2010, Carcinoma within solitary ductal papilloma of the breast. EJSO 36:384–386 Maxwell AJ, 2009, Ultrasound guided vacuum-assisted excision of breast papillomas: review of 6-years experience. Clin Radiol 64:801–806 Haagensen CD Diseases of the Breast 3rd ed, 1986, Solitary intraductal papilloma. Philadelphia: WB Saunders, pp 137–175 Hou M-F, Huang T-J, Liu G-C, 2005, The diagnostic value of galactography in patients with nipple discharge. Clin Imaging 25:74–81 Bruce L, Daniel R, Gardner W, Robyn L, Birdwell K, Nowels W, Denise J, 2003, Magnetic resonance imaging of intraductal papilloma of the breast. Mag Reson Im 21:887–892 Hunerbein M, Dubowy A, Raubach M et al, 2007, Gradient index ductoscopy and intraductal biopsy of intraductal breast lesions. Am J Surg 194:511–514 Leah S, Feldman SM, Balassanian R et al, 2004, Association of breast cancer with papillary lesions identified at percutaneous image-guided breast biopsy. Am J Surg 188:365–370 Amendoeira I, Apostolikas N, Bellocq JP et al, 2006, Quality assurance guidelines for pathology. In: Perry N, Broeders M, de Wolf C, Tornberg S, Holland R, von Karsa L, eds, European guidelines for quality assurance in breast cancer screening and diagnosis, 4th edn. European Comission, Luxemburg, pp 219–311 Cserni G, 2012, Benign apocrine papillary lesions of the breast lacking or virtually lacking myoepihelial cells- potential pitfalls in diagnosing malignancy. APMIS 120:249–252 Gennari R, Galimberti V, De Cicco C et al, 1999, Use of technetium-99m-labeled colloid albumin for preoperative and intraoperative localization of nonpalpable breast lesions. J Am Coll Surg 1(90):692–699 Leis HP Jr, 1989, Management of nipple discharge. World J Surg 13:736–742 Buhl-Jorgensen SE, Fischermann K, Johansen H et al, 1968, Cancer risk in intraductal papilloma and papillomatosis. Surg Gynecol Obstet 127:1307–1312 WAl Sarakbi, Worku D, Escobar PF, Mokbel K, 2006, Breast papillomas: current managment with a focus on a new diagnostic and therapeutic modality. International Seminars in Surgical Oncology SneigeN(2000, Fine-needle aspiration cytology of in situ epithelial cell proliferation in the breast. Am J Clin Pathol 113(5 Suppl 1):S38–S48 Kefah M, Pedro FE, Tadaharu M, 2005, Mammary ductoscopy: current status and future prospects. EJSO 31:3–8 Shen K-W, Wu J, Lu J-S et al, 2001, Fiberoptic ductoscopy for breast cancer patients with nipple discharge. Surg Endosc 15:1340–1345 Atkins H,Wolff B, 1964, Discharge from the nipple. BJS 51:602–606 Urban JA, 1963, Excision of the major duct system of the breast. Cancer 16:516–520 Dietz JR, Crowe JP, Grundfest S, Arrigan S, Kim JA, 2002, Directed duct excision by using mammary ductoscopy in patients with pathologic nipple discharge. Surgery 132:582–587 Cardenosa G, Eklund GW, 1991, Benign papillary neoplasms of the breast: mammographic findings. Radiology 181:751–755 Ali-Fehmi R, Carolin K, Wallis T, Visscher DW, 2003, Clinicopathologic analysis of breast lesions associated with multiple papillomas. Hum Pathol 34:234–239 Page DL, Salhany KE, Jensen RA, Dupont WD, 1996, Subsequent breast carcinoma risk after biopsy with atypia in a breast papilloma. Cancer 78:258–266 O’Malley F, Visscher D, MacGrogan G et al, 2012, Intraductal papillary lesions. In: Lakhani SR, Ellis IO, Schnitt SJ et al, eds, WHO classification of tumours of the breast, 4th edn. International Agency for Research on Cancer, Lyon, pp 99–109 Cserni G, 2011, The current TNM classification of breast carcinomas: controversial issues in eraly breast cancer. memo 4:144–148 Sakr R, Rouzier R, Salem C, Antoine M, Chopier J, Darai E et al, 2008, Risk of breast cancer associated with papilloma. EJSO 34:1304–1308 El-Sayed ME, Rakha EA, Reed J, Lee AHS, Evans AJ, Ellis IO, 2008, Predictive value of needle core biopsy diagnoses of lesions of uncertain malignant potential, B+, in abnormalities detected by mammographic screening. Hystopathology 53:650–657 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 589 EP 595 DI 10.1007/s12253-013-9622-4 PG 7 ER PT J AU Zhu, W Chen, L Ai, Z AF Zhu, Wei Chen, Li Ai, Zhilong TI Candidate Agents for Papillary Thyroid Cancer Identified by Gene Expression Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary thyroid carcinoma; Differentially expressed genes; Small molecules ID Papillary thyroid carcinoma; Differentially expressed genes; Small molecules AB A better understanding of the molecular mechanisms involved in papillary thyroid cancer (PTC) is needed to manage these patients effectively. Our objectives were to expand our understanding of this disease, and to identify biologically active small molecules capable to reverse PTC. We downloaded gene expression data of PTC from Gene Expression Omnibus database and employed computational bioinformatics analysis to compare gene expression patterns with normal tissues. Small molecules that induced inverse gene changes to the PTC were identified. A total of 2,154 differentially expressed genes (DEGs) with a false discovery rate of 0.01 were identified. These 2,154 DEGs were significantly enriched in 17 pathways, including pathways associated with signal transduction, tumorigenesis and lipid or amino acid metabolism. In addition, we identified large amount of small molecules that capable to reverse PTC. We found a group of small molecules that can provide new ideas for the therapeutic studies in PTC. These drugs are clearly a direction that warrants additional consideration. C1 [Zhu, Wei] Fudan University, Hua Shan Hospital, Department of General Surgery, No.180 Fenglin Road, Xuhui District, 200032 Shanghai, China. [Chen, Li] Fudan University, Hua Shan Hospital, Department of General Surgery, No.180 Fenglin Road, Xuhui District, 200032 Shanghai, China. [Ai, Zhilong] Fudan University, Hua Shan Hospital, Department of General Surgery, No.180 Fenglin Road, Xuhui District, 200032 Shanghai, China. RP Ai, Z (reprint author), Fudan University, Hua Shan Hospital, Department of General Surgery, 200032 Shanghai, China. EM aizhilongazl@hotmail.com CR Hundahl SA, Fleming ID, Fremgen AM, Menck HR, 1998, A National Cancer Data Base report on 53,856 cases of thyroid carcinoma treated in the U.S., 1985–1995. Cancer 83(12):2638– 2648., DOI 10.1002/(SICI)1097-0142(19981215)83:12<2638:: AID-CNCR31>3.0.CO;2-1 Leenhardt L, Grosclaude P, Cherie-Challine L, 2004, Increased incidence of thyroid carcinoma in france: a true epidemic or thyroid nodule management effects? Report from the French Thyroid Cancer Committee. Thyroid 14(12):1056–1060., DOI 10.1089/ thy.2004.14.1056 Davies L, Welch HG, 2006, Increasing incidence of thyroid cancer in the United States, 1973–2002. JAMA 295(18):2164–2167., DOI 10.1001/jama.295.18.2164 Wang Y, Wang W, 2012, Increasing incidence of thyroid cancer in Shanghai, China, 1983–2007. Asia Pac J Public Health., DOI 10.1177/ 1010539512436874 Olaleye O, Ekrikpo U, Moorthy R, Lyne O, Wiseberg J, Black M, Mitchell D, 2011, Increasing incidence of differentiated thyroid cancer in South East England: 1987–2006. Eur Arch Otorhinolaryngol 268(6):899–906., DOI 10.1007/s00405-010-1416-7 Ramirez-Vick M, Nieves-Rodriguez M, Lugaro-Gomez A, Perez- Irizarry J, 2011, Increasing incidence of thyroid cancer in Puerto Rico, 1985–2004. P R Health Sci J 30(3):109–115 Capezzone M, Morabito E, Bellitti P, Giannasio P, De Sanctis D, Bruno R, 2007, Increasing incidence of thyroid cancer in Basilicata: an Italian study. J Endocrinol Invest 30(6):507–512 Hall SF, Walker H, Siemens R, Schneeberg A, 2009, Increasing detection and increasing incidence in thyroid cancer. World J Surg 33(12):2567–2571., DOI 10.1007/s00268-009-0226-9 Sanabria A, Dominguez LC, Vega V, Osorio C, 2011, Prognosis of patients with thyroid cancer who do not undergo surgical treatment: a SEER database analysis. Clin Transl Oncol 13(9):692– 696., DOI 10.1007/s12094-011-0716-8 Cady B, Rossi R, 1988, An expanded view of risk-group definition in differentiated thyroid carcinoma. Surgery 104(6):947–953 Santacroce L, Gagliardi S, SA K, 2009, Thyroid, papillary carcinoma: treatment & medication. eMedicine Oncol 12:17 Legakis I, Syrigos K, 2011, Recent advances in molecular diagnosis of thyroid cancer. J Thyroid Res 2011:384213., DOI 10.4061/ 2011/384213 Xing M, 2007, BRAF mutation in papillary thyroid cancer: pathogenic role, molecular bases, and clinical implications. Endocr Rev 28(7):742–762., DOI 10.1210/er.2007-0007 Fusco A, Grieco M, Santoro M, Berlingieri MT, Pilotti S, Pierotti MA, Della Porta G, Vecchio G, 1987, A new oncogene in human thyroid papillary carcinomas and their lymph-nodal metastases. Nature 328(6126):170–172., DOI 10.1038/328170a0 Santoro M, Melillo RM, Grieco M, Berlingieri MT, Vecchio G, Fusco A, 1993, The TRK and RET tyrosine kinase oncogenes cooperate with ras in the neoplastic transformation of a rat thyroid epithelial cell line. Cell Growth Differ 4(2):77–84 Nikiforova MN, Lynch RA, Biddinger PW, Alexander EK, Dorn GW 2nd, Tallini G, Kroll TG, Nikiforov YE, 2003, RAS point mutations and PAX8-PPAR gamma rearrangement in thyroid tumors: evidence for distinct molecular pathways in thyroid follicular carcinoma. J Clin Endocrinol Metab 88(5):2318–2326 Placzkowski KA, Reddi HV, Grebe SK, Eberhardt NL, McIver B, 2008, The role of the PAX8/PPARgamma fusion oncogene in thyroid cancer. PPAR Res 2008:672829., DOI 10.1155/2008/672829 Malaguarnera R, Vella V, Vigneri R, Frasca F, 2007, p53 family proteins in thyroid cancer. Endocr Relat Cancer 14(1):43–60., DOI 10.1677/erc.1.01223 Knauf JA, Kuroda H, Basu S, Fagin JA, 2003, RET/PTC-induced dedifferentiation of thyroid cells is mediated through Y1062 signaling through SHC-RAS-MAP kinase. Oncogene 22(28):4406– 4412., DOI 10.1038/sj.onc.1206602 Mitsutake N, Miyagishi M, Mitsutake S, Akeno N, Mesa C Jr, Knauf JA, Zhang L, Taira K, Fagin JA, 2006, BRAF mediates RET/PTC-induced mitogen-activated protein kinase activation in thyroid cells: functional support for requirement of the RET/PTCRAS- BRAF pathway in papillary thyroid carcinogenesis. Endocrinology 147(2):1014–1019., DOI 10.1210/en.2005-0280 He H, Jazdzewski K, Li W, Liyanarachchi S, Nagy R, Volinia S, Calin GA, Liu CG, Franssila K, Suster S, Kloos RT, Croce CM, de la Chapelle A, 2005, The role of microRNA genes in papillary thyroid carcinoma. Proc Natl Acad Sci U S A 102(52):19075– 19080., DOI 10.1073/pnas.0509603102 Team RDC, 2008, R: a language and environment for statistical computing. R Foundation for Statistical Computing. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP, 2003, Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4(2):249–264., DOI 10.1093/biostatistics/4.2.249 Smyth GK, 2004, Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Statistical applications in genetics and molecular biology 3:Article3., DOI 10.2202/1544-6115.1027 Benjamini Y, Hochberg Y, 1995, Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B, Methodological, 57:289–300 Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G, 2000, Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 25(1):25–29., DOI 10.1038/75556 Yu G, Wang LG, Han Y, He QY, 2012, clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16(5):284–287., DOI 10.1089/omi.2011.0118 Kanehisa M, 2002, The KEGG database. Novartis Found Symp 247:91–101, discussion 101-103, 119-128, 244-152 da Huang W, Sherman BT, Lempicki RA, 2009, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4(1):44–57., DOI 10.1038/nprot.2008.211 Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, Reich M, Hieronymus H, Wei G, Armstrong SA, Haggarty SJ, Clemons PA, Wei R, Carr SA, Lander ES, Golub TR, 2006, The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313(5795):1929–1935., DOI 10.1126/science.1132939 Donato R, 2001, S100: a multigenic family of calcium-modulated proteins of the EF-hand type with intracellular and extracellular functional roles. Int J Biochem Cell Biol 33(7):637–668 Heizmann CW, Fritz G, Schafer BW, 2002, S100 proteins: structure, functions and pathology. Front Biosci 7:d1356–1368 Roth J, Vogl T, Sorg C, Sunderkotter C, 2003, Phagocyte-specific S100 proteins: a novel group of proinflammatory molecules. Trends Immunol 24(4):155–158 Kanamori T, Takakura K, Mandai M, Kariya M, Fukuhara K, Sakaguchi M, Huh NH, Saito K, Sakurai T, Fujita J, Fujii S, 2004, Increased expression of calcium-binding protein S100 in human uterine smooth muscle tumours. Mol Hum Reprod 10(10):735–742., DOI 10.1093/molehr/gah100 Rust R, Visser L, van der Leij J, Harms G, Blokzijl T, Deloulme JC, van der Vlies P, Kamps W, Kok K, Lim M, Poppema S, van den Berg A, 2005, High expression of calcium-binding proteins, S100A10, S100A11 and CALM2 in anaplastic large cell lymphoma. Br J Haematol 131(5):596–608., DOI 10.1111/j.1365- 2141.2005.05816.x Ohuchida K, Mizumoto K, Ohhashi S, Yamaguchi H, Konomi H, Nagai E, Yamaguchi K, Tsuneyoshi M, Tanaka M, 2006, S100A11, a putative tumor suppressor gene, is overexpressed in pancreatic carcinogenesis. Clin Cancer Res 12(18):5417–5422., DOI 10.1158/1078-0432.CCR-06-0222 Memon AA, Sorensen BS, Meldgaard P, Fokdal L, Thykjaer T, Nexo E, 2005, Down-regulation of S100C is associated with bladder cancer progression and poor survival. Clin Cancer Res 11(2 Pt 1):606–611 Lesniak W, Slomnicki LP, Filipek A, 2009, S100A6—new facts and features. Biochem Biophys Res Commun 390(4):1087–1092., DOI 10.1016/j.bbrc.2009.10.150 Salama I, Malone PS, Mihaimeed F, Jones JL, 2008, A review of the S100 proteins in cancer. Eur J Surg Oncol 34(4):357–364., DOI 10.1016/j.ejso.2007.04.009 DeRycke MS, Andersen JD, Harrington KM, Pambuccian SE, Kalloger SE, Boylan KL, Argenta PA, Skubitz AP, 2009, S100A1 expression in ovarian and endometrial endometrioid carcinomas is a prognostic indicator of relapse-free survival. Am J Clin Pathol 132(6):846–856., DOI 10.1309/ AJCPTK87EMMIKPFS Cossu-Rocca P, Contini M, Brunelli M, Festa A, Pili F, Gobbo S, Eccher A, Mura A, Massarelli G, Martignoni G, 2009, S-100A1 is a reliable marker in distinguishing nephrogenic adenoma from prostatic adenocarcinoma. Am J Surg Pathol 33(7):1031–1036., DOI 10.1097/PAS.0b013e31819c6ff9 Camby I, Nagy N, Lopes MB, Schafer BW, Maurage CA, Ruchoux MM, Murmann P, Pochet R, Heizmann CW, Brotchi J, Salmon I, Kiss R, Decaestecker C, 1999, Supratentorial pilocytic astrocytomas, astrocytomas, anaplastic astrocytomas and glioblastomas are characterized by a differential expression of S100 proteins. Brain Pathol 9(1):1–19 Gogas H, Eggermont AM, Hauschild A, Hersey P, Mohr P, Schadendorf D, Spatz A, Dummer R, 2009, Biomarkers in melanoma. Ann Oncol 20(Suppl 6):vi8–13., DOI 10.1093/annonc/mdp251 Thanos D, Du W, Maniatis T, 1993, The high mobility group protein HMG I(Y, is an essential structural component of a virus-inducible enhancer complex. Cold Spring Harb Symp Quant Biol 58:73–81 Fusco A, Fedele M, 2007, Roles of HMGA proteins in cancer. Nat Rev Cancer 7(12):899–910., DOI 10.1038/nrc2271 Peluso S, Chiappetta G, 2010, High-mobility group A, HMGA, proteins and breast cancer. Breast Care, Basel, 5(2):81–85., DOI 10.1159/000297717 Abe N, Watanabe T, Sugiyama M, Uchimura H, Chiappetta G, Fusco A, Atomi Y, 1999, Determination of high mobility group I(Y, expression level in colorectal neoplasias: a potential diagnostic marker. Cancer Res 59(6):1169–1174 Tamimi Y, van der Poel HG, Karthaus HF, Debruyne FM, Schalken JA, 1996, A retrospective study of high mobility group protein I(Y, as progression marker for prostate cancer determined by in situ hybridization. Br J Cancer 74(4):573– 578 Masciullo V, Baldassarre G, Pentimalli F, Berlingieri MT, Boccia A, Chiappetta G, Palazzo J, Manfioletti G, Giancotti V, Viglietto G, Scambia G, Fusco A, 2003, HMGA1 protein over-expression is a frequent feature of epithelial ovarian carcinomas. Carcinogenesis 24(7):1191–1198., DOI 10.1093/carcin/bgg075 Frasca F, Rustighi A, Malaguarnera R, Altamura S, Vigneri P, Del Sal G, Giancotti V, Pezzino V, Vigneri R, Manfioletti G, 2006, HMGA1 inhibits the function of p53 family members in thyroid cancer cells. Cancer Res 66(6):2980–2989., DOI 10.1158/0008- 5472.CAN-05-2637 Pucci E, Chiovato L, Pinchera A, 2000, Thyroid and lipid metabolism. Int J Obes Relat Metab Disord 24(Suppl 2): S109–112 Liu LF, Desai SD, Li TK, Mao Y, Sun M, Sim SP, 2000, Mechanism of action of camptothecin. Ann N Y Acad Sci 922:1–10 Bomgaars L, Berg SL, Blaney SM, 2001, The development of camptothecin analogs in childhood cancers. Oncologist 6(6):506– 516 Souhami RL, Prankerd TA, 1970, Daunorubicin in acute leukaemia. Postgrad Med J 46(535):272–275 Gottlieb AJ, Weinberg V, Ellison RR, Henderson ES, Terebelo H, Rafla S, Cuttner J, Silver RT, Carey RW, Levy RN et al, 1984, Efficacy of daunorubicin in the therapy of adult acute lymphocytic leukemia: a prospective randomized trial by cancer and leukemia group B. Blood 64(1):267–274 Fox EJ, 2006, Management of worsening multiple sclerosis with mitoxantrone: a review. Clin Ther 28(4):461–474., DOI 10.1016/ j.clinthera.2006.04.013 Periti P, della Cuna GR, Pannuti F, Mazzei T, Preti P, Martoni A, Mini E, 1985, First-line combination chemotherapy with mitoxantrone and cyclophosphamide in advanced breast cancer. Invest New Drugs 3(2):167–171 Ho AD, Lipp T, Ehninger G, Illiger HJ, Meyer P, Freund M, Hunstein W, 1988, Combination of mitoxantrone and etoposide in refractory acute myelogenous leukemia—an active and welltolerated regimen. J Clin Oncol 6(2):213–217 Johnson K, Liu L, Majdzadeh N, Chavez C, Chin PC, Morrison B, Wang L, Park J, Chugh P, Chen HM, D’Mello SR, 2005, Inhibition of neuronal apoptosis by the cyclindependent kinase inhibitor GW8510: identification of 3′ substituted indolones as a scaffold for the development of neuroprotective drugs. J Neurochem 93(3):538–548., DOI 10.1111/ j.1471-4159.2004.03004.x Fomina-Yadlin D, Kubicek S, Vetere A, He KH, Schreiber SL, Wagner BK, 2012, GW8510 increases insulin expression in pancreatic alpha cells through activation of p53 transcriptional activity. PLoS One 7(1):e28808., DOI 10.1371/journal. pone.0028808 Lahusen T, De Siervi A, Kunick C, Senderowicz AM, 2003, Alsterpaullone, a novel cyclin-dependent kinase inhibitor, induces apoptosis by activation of caspase-9 due to perturbation in mitochondrial membrane potential. Mol Carcinog 36(4):183–194., DOI 10.1002/mc.10114 Shimaoka K, Schoenfeld DA, DeWys WD, Creech RH, DeConti R, 1985, A randomized trial of doxorubicin versus doxorubicin plus cisplatin in patients with advanced thyroid carcinoma. Cancer 56(9):2155–2160 Williams SD, Birch R, Einhorn LH, 1986, Phase II evaluation of doxorubicin plus cisplatin in advanced thyroid cancer: a Southeastern Cancer Study Group Trial. Cancer Treat Rep 70(3):405– 407 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2013 VL 19 IS 3 BP 597 EP 604 DI 10.1007/s12253-013-9625-1 PG 8 ER PT J AU Vuity, D Bogdan, S Csurgay, K Sapi, Z Nemeth, Zs AF Vuity, Drazsen Bogdan, Sandor Csurgay, Katalin Sapi, Zoltan Nemeth, Zsolt TI Malignant Fibrous Histiocytoma/Undifferentiated High-Grade Pleomorphic Sarcoma of the Maxillary Sinus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Malignant fibrous histiocytoma; Undifferentiated high-grade pleomorphic sarcoma; Orbitoethmoidal spread; Young age; Lymphatic metastasis ID Malignant fibrous histiocytoma; Undifferentiated high-grade pleomorphic sarcoma; Orbitoethmoidal spread; Young age; Lymphatic metastasis AB Malignant fibrous histiocytoma (MFH) also known as undifferentiated high-grade pleomorphic sarcoma (UHPS) is a soft tissue sarcoma, composed of undifferentiated mesenchymal tumors possessed fibrohistiocytic morphology without definite true histiocytic differentiation. Head and neck localization is very rare, showing an incidence ranging from 4% to 10% in different series of investigations. The most frequent involved sites in UHPS are the neck and parotid, followed by the scalp, face, anterior skull base and orbit. Upper aerodigestive tract, lateral skull base and ear are rare locations. The incidence of the lymphatic metastases is also rare. The aim of this article is to report a case of UHPS in the maxillary sinus with palatal, orbital and ethmoidal involvement, with lymphatic metastasis and its surgical treatment. In addition, we review the literature of similar cases of the past 12 years. C1 [Vuity, Drazsen] Semmelweis University, Department of Oral and Maxillofacial SurgeryBudapest, Hungary. [Bogdan, Sandor] Semmelweis University, Department of Oral and Maxillofacial SurgeryBudapest, Hungary. [Csurgay, Katalin] Semmelweis University, Department of Oral and Maxillofacial SurgeryBudapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Nemeth, Zsolt] Semmelweis University, Department of Oral and Maxillofacial SurgeryBudapest, Hungary. RP Vuity, D (reprint author), Semmelweis University, Department of Oral and Maxillofacial Surgery, Budapest, Hungary. EM drazsen@gmail.com CR Ozzello L, Stout AP, Murray MR, 1963, Cultural characteristics of malignant histiocytomas and fibrous xanthomas. Cancer 16:331– 344 Matushansky I, Charytonowicz E, Mills J, Siddiqi S, Hricik T, Cordon-Cardo C, 2009, MFH classification: differentiating undifferentiated pleomorphic sarcoma in the 21st Century. Expert Rev Anticancer Ther 9:1135–1144 Enzinger FM, 1986, Malignant fibrous histiocytoma 20 years after Stout. Am J Surg Pathol 10(Suppl 1):43–53 Dehner LP, 1988, Malignant fibrous histiocytoma. Nonspecific morphologic pattern, specific pathologic entity, or both? Arch Pathol Lab Med 112:236–237 Fletcher CD, 1992, Pleomorphic malignant fibrous histiocytoma: fact or fiction? A critical reappraisal based on 159 tumors diagnosed as pleomorphic sarcoma. Am J Surg Pathol 16:213–228 Fletcher CD, Unni KK, Mertens F, 2002, Pathology and genetics of tumours of soft tissue and bone, World Health Organization Classification of Tumours). IARC Press, Lyon Fletcher CD, 2006, The evolving classification of soft tissue tumours: an update based on the new WHO classification. Histopathology 48:3–12 Daugaard S, 2004, Current soft-tissue sarcoma classifications. Eur J Cancer 40:543–548 Murphey MD, 2007, World Health Organization classification of bone and soft tissue tumors: modifications and implications for radiologists. Semin Musculoskelet Radiol 11:201–214 Randall RL, Albritton KH, Ferney BJ, Layfield L, 2004, Malignant fibrous histiocytoma of soft tissue: an abandoned diagnosis. Am J Orthop, Belle Mead NJ, 33:602–608 Bentz BG, Singh B, Woodruff J, Brennan M, Shah JP, Kraus D, 2004, Head and neck soft tissue sarcomas: a multivariate analysis of outcomes. Ann Surg Oncol 11:619–628 Park SW, Kim HJ, Lee JH, Ko YH, 2009, Malignant fibrous histiocytoma of the head and neck: CT and MR imaging findings. Am J Neuroradiol 30:71–76 Sarrami AH, Setareh M, Afshar-Moghaddam N, Izadinejad M, Saadatnia M, 2011, A case of intracranial malignant fibrous histiocytoma. J Res Med Sci 16:968–973 Al-Agha OM, Igbokwe AA, 2008, Malignant fibrous histiocytoma: between the past and the present. Arch Pathol Lab Med 132:1030–1035 Todoroki T, Kondo T, Sugahara S, Morishita Y, Mori K, Ohno T, 2011, Long-term survivor of relapsed MFH on the thigh treated with autologous formalin-fixed tumor vaccine, AFTV, combined with limb-sparing surgery and radiotherapy. World J Surg Oncol 9:96 Matsumoto S, Ahmed AR, Kawaguchi N, Manabe J, Matsushita Y, 2003, Results of surgery for malignant fibrous histiocytomas of soft tissue. Int J Clin Oncol 8:104–109 Huvos AG, 1976, Primary malignant fibrous histiocytoma of bone; clinicopathologic study of 18 patients. N Y State J Med 76:552–559 Weiss SW, Enzinger FM, 1978, Malignant fibrous histiocytoma: an analysis of 200 cases. Cancer 41:2250–2266 Jin GP, Zhao M, Qi JX, Jiang HG, Yu SG, 2003, Malignant fibrous histiocytoma of head and neck: clinical analysis of 21 cases. Ai Zheng 22:523–525 Wu TH, Shih CW, Huang JS, Wang CH, Yeh KY, 2012, Unusual hematogenous brain metastasis in malignant fibrous histiocytoma of the maxillary sinus. Int J Clin Oncol 17:69–74 Satomi T, Watanabe M, Kaneko T, Matsubayashi J, Nagao T, Chiba H, 2011, Radiation-induced malignant fibrous histiocytoma of the maxilla. Odontology 99:203–208 Yanagi Y, Watanabe M, Kaneko T, Matsubayashi J, Nagao T, Chiba H, 2010, A case of malignant fibrous histiocytoma of the maxillary sinus. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 109:99–104 Agnihotri R, Bhat KM, Bhat GS, 2008, A rare case of malignant fibrous histiocytoma of the gingiva. J Periodontol 79:955–960 Seper L, Schwab R, Kiattavorncharoen S, Buchter A, Bankfalvi A, Joos U, Piffko J, Kruse-Loesler B, 2007, Malignant fibrous histiocytoma of the face: report of a case. Head Face Med 3:36 Sabesan T, Xuexi W, Yongfa Q, Pingzhang T, Ilankovan V, 2006, Malignant fibrous histiocytoma: outcome of tumours in the head and neck compared with those in the trunk and extremities. Br J Oral Maxillofac Surg 44:209–212 Chan YW, Guo YC, Tsai TL, Tsay SH, Lin CZ, 2004, Malignant fibrous histiocytoma of the maxillary sinus presenting as toothache. J Chin Med Assoc 67:104–107 Yamaguchi S, Nagasawa H, Suzuki T, Fujii E, Iwaki H, Takagi M, Amagasa T, 2004, Sarcomas of the oral and maxillofacial region: a review of 32 cases in 25 years. Clin Oral Investig 8:52–55 Sato T, Kawabata Y, Morita Y, Noikura T, Mukai H, Kawashima K, Sugihara K, 2001, Radiographic evaluation of malignant fibrous histiocytoma affecting maxillary alveolar bone: a report of 2 cases. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 92:116– 123 Mardinger O, Givol N, Talmi YP, Taicher S, 2001, Osteosarcoma of the jaw. The Chaim Sheba Medical Center experience. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 91:445–451 Pandey M, Chandramohan K, Thomas G, Mathew A, Sebastian P, Somanathan T, Abraham EK, Rajan B, Krishnan Nair M, 2003, Soft tissue sarcoma of the head and neck region in adults. Int J Oral Maxillofac Surg 32:43–48 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 605 EP 609 DI 10.1007/s12253-013-9640-2 PG 5 ER PT J AU Xie, B Chen, J Liu, B Zhan, J AF Xie, Biao Chen, Jinhui Liu, Bin Zhan, Junkun TI Klotho Acts as a Tumor Suppressor in Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Klotho; Insulin/IGF1 axis; Autophagy; Wnt signaling; ROS ID Klotho; Insulin/IGF1 axis; Autophagy; Wnt signaling; ROS AB The klotho gene is a classical "aging suppressor" gene. Its roles in the pathology of chronic kidney diseases have been well documented. However, the role of Klotho in tumorigenesis, cancer progression, and prognosis is attracting more and more attention. Recent studies have shown that Klotho participates in the progression of several types of human cancers. Klotho functions as a tumor suppressor by inhibiting insulin/IGF1, p53/p21, and Wnt signaling. Silencing klotho gene expression is mainly mediated through promoter hypermethylation and histone deacetylation in cancer. Klotho has been proposed to take part in cell proliferation, survival, autophagy, and resistance to anti-cancer therapies. C1 [Xie, Biao] 8th Changsha Hospital, Department of General Surgery, 410015 Changsha, Hunan, China. [Chen, Jinhui] 8th Changsha Hospital, Department of General Surgery, 410015 Changsha, Hunan, China. [Liu, Bin] 8th Changsha Hospital, Department of General Surgery, 410015 Changsha, Hunan, China. [Zhan, Junkun] Central South University, Second Xiangya Hospital, Department of Geriatric Medicine, 410011 Changsha, Hunan, China. RP Zhan, J (reprint author), Central South University, Second Xiangya Hospital, Department of Geriatric Medicine, 410011 Changsha, China. EM zhanjunkun@yahoo.com CR Kuro-o M, Matsumura Y, Aizawa H et al, 1997, Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 390:45–51 Kurosu H, Yamamoto M, Clark JD et al, 2005, Suppression of aging in mice by the hormone Klotho. Science 309:1829–1833 Torres PU, Prie D, Beck L, De Brauwere D, Leroy C, Friedlander G, 2009, Klotho gene, phosphocalcic metabolism, and survival in dialysis. J Ren Nutr 19:50–56 Kuro-o M, 2012, Klotho in health and disease. Curr Opin Nephrol Hypertens 21:362–368 Lu L, Katsaros D, Wiley A, de la Longrais IA, Puopolo M, Yu H, 2008, Klotho expression in epithelial ovarian cancer and its association with insulin-like growth factors and disease progression. Cancer Invest 26:185–192 Wolf I, Levanon-Cohen S, Bose S et al, 2008, Klotho: a tumor suppressor and a modulator of the IGF-1 and FGF pathways in human breast cancer. Oncogene 27:7094–7105 Wolf I, Laitman Y, Rubinek T et al, 2010, Functional variant of KLOTHO: a breast cancer risk modifier among BRCA1 mutation carriers of Ashkenazi origin. Oncogene 29:26–33 Laitman Y, Kuchenbaecker KB, Rantala J et al, 2012, The KL-VS sequence variant of Klotho and cancer risk in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 132:1119–1126 Chen B,Wang X, ZhaoW,Wu J, 2010, Klotho inhibits growth and promotes apoptosis in human lung cancer cell line A549. J Exp Clin Cancer Res 29:99–106 Lee J, Jeong DJ, Kim J et al, 2010, The antiaging gene KLOTHO is a novel target for epigenetic silencing inhuman cervical carcinoma. Mol Cancer 9:109–118 Wang LJ,Wang X,Wang XJ et al, 2011, Klotho is silenced through promoter hypermethylation in gastric cancer. Am J Cancer Res 1:111–119 Abramovitz L, Rubinek T, Ligumsky H et al, 2011, KL1 internal repeat mediates klotho tumor suppressor activities and inhibits bFGF and IGF-I signaling in pancreatic cancer. Clin Cancer Res 17:4254–4266 Bartucci M, Morelli C, Mauro L, Ando S, Surmacz E, 2001, Differential insulin-like growth factor I receptor signaling and function in estrogen receptor, ER)-positive MCF-7 and ER-negative MDA-MB-231 breast cancer cells. Cancer Res 61:6747–6754 Pan J, Zhong J, Gan LH et al, 2011, Klotho, an anti-senescence related gene, is frequently inactivated through promoter hypermethylation in colorectal cancer. Tumour Biol 32:729–735 Rubinek T, Shulman M, Israeli S, et al., 2011, Epigenetic silencing of the tumor suppressor klotho in human breast cancer. Breast Cancer Res Treat [Epub ahead of print] Wang X, Chen B, Xu W, Liu S, Zhao W, Wu J, 2011, Combined effects of klotho and soluble CD40 ligand on A549 lung cancer cells. Oncol Rep 25:1465–1472 Doi S, Zou Y, Togao O et al, 2011, Klotho inhibits transforming growth factor-beta1, TGF-beta1, signaling and suppresses renal fibrosis and cancer metastasis in mice. Biol Chem 286:8655–8665 Shu G, Xie B, Ren F, et al., 2012, Restoration of klotho expression induces apoptosis and autophagy in hepatocellular carcinoma cells. Cell Oncol, Dordr, [Epub ahead of print] Xie B, Zhou J, Yuan L, et al., 2012, Epigenetic silencing of Klotho expression correlates with poor prognosis of human hepatocellular carcinoma. Hum Pathol Pollak MN, Schernhammer ES, Hankinson SE, 2004, Insulin-like growth factors and neoplasia. Nat Rev Cancer 4:505–518 Evans DS, Kapahi P, Hsueh WC, Kockel L, 2011, TOR signaling never gets old: aging, longevity and TORC1 activity. Ageing Res Rev 10:225–237 Carboni JM, Lee AV, Hadsell DL et al, 2005, Tumor development by transgenic expression of a constitutively active insulin-like growth factor I receptor. Cancer Res 5:3781–3787 Lopez T, Hanahan D, 2002, Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 1:339–353 Yakar S, Leroith D, Brodt P, 2005, The role of the growth hormone/ insulin-like growth factor axis in tumor growth and progression: Lessons from animal models. Cytokine Growth Factor Rev 16:407–420 Yakar S, Leroith D, Brodt P, 2005, The role of the growth hormone/ insulin-like growth factor axis in tumor growth and progression: Lessons fromanimalmodels. Cytokine Growth Factor Rev 6:407–420 Usuda J, Ichinose S, Ishizumi T et al, 2011, Klotho is a novel biomarker for good survival in resected large cell neuroendocrine carcinoma of the lung. Lung Cancer 72:355–359 Usuda J, Ichinose S, Ishizumi T et al, 2011, Klotho predicts good clinical outcome in patients with limited-disease small cell lung cancer who received surgery. Lung Cancer 74:332–337 Xie B, Zhou J, Shu G et al, 2013, Restoration of klotho gene expression induces apoptosis and autophagy in gastric cancer cells: tumor suppressive role of klotho in gastric cancer. Cancer Cell Int 13:18 Chihara Y, Rakugi H, Ishikawa K et al, 2006, Klotho protein promotes adipocyte differentiation. Endocrinology 147:3835–3842 Lorenzi O, Veyrat-Durebex C, Wollheim CB et al, 2010, Evidence against a direct role of klotho in insulin resistance. Pflugers Arch 459:465–473 Duce JA, Podvin S, Hollander Wet al, 2008, Gene profile analysis implicates Klotho as an important contributor to aging changes in brain white matter of the rhesus monkey. Glia 56:106–117 Choi BH, Kim CG, Lim Y, Lee YH, Shin SY, 2010, Transcriptional activation of the human Klotho gene by epidermal growth factor in HEK293 cells; role of Egr-1. Gene 450:121–127 Shen J, Stass SA, Jiang F, 2013, MicroRNAs as potential biomarkers in human solid tumors. Cancer Lett 329:125–136 Liang R, Bates DJ, Wang E, 2009, Epigenetic Control of MicroRNA Expression and Aging. Curr Genomics 10:184–193 Maes OC, An J, Sarojini H, Wang E, 2008, Murine microRNAs implicated in liver functions and aging process. Mech Ageing Dev 129:534–541 Logan CY, Nusse R, 2004, The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol 20:781–810 Klaus A, Birchmeier W, 2008, Wnt signalling and its impact on development and cancer. Nat Rev Cancer 8:387–398 Suzuki H, Watkins DN, Jair KW et al, 2004, Epigenetic inactivation of SFRP genes allows constitutive WNTsignaling in colorectal cancer. Nat Genet 36:417–422 Urakami S, Shiina H, Enokida H et al, 2006, Combination analysis of hypermethylated Wnt-antagonist family genes as a novel epigenetic biomarker panel for bladder cancer detection. Clin Cancer Res 12:2109–2116 Nojima M, Suzuki H, Toyota M et al, 2007, Frequent epigenetic inactivation of SFRP genes and constitutive activation of Wnt signaling in gastric cancer. Oncogene 26:4699–4713 Suzuki H, Toyota M, Carraway H et al, 2008, Frequent epigenetic inactivation of Wnt antagonist genes in breast cancer. Br J Cancer 98:1147–1156 Camilli TC, Xu M, O’Connell MP et al, 2011, Loss of Klotho during melanoma progression leads to increased filamin cleavage, increased Wnt5A expression, and enhanced melanoma cell motility. Pigment Cell Melanoma Res 24:175–186 Liu H, Fergusson MM, Castilho RM et al, 2007, Augmented Wnt signaling in a mammalian model of accelerated aging. Science 317:803–806 Kirstetter P, Anderson K, Porse BT, Jacobsen SE, Nerlov C, 2006, Activation of the canonical Wnt pathway leads to loss of hematopoietic stem cell repopulation and multilineage differentiation block. Nat Immunol 7:1048–1056 Nusse R, 2008, Wnt signaling and stem cell control. Cell Res 18:523–527 Castilho RM, Squarize CH, Chodosh LA,Williams BO, Gutkind JS, 2009, mTOR mediates Wnt-induced epidermal stem cell exhaustion and aging. Cell Stem Cell 4:279–289 Scheller M, Huelsken J, Rosenbauer F et al, 2006, Hematopoietic stem cell and multilineage defects generated by constitutive betacatenin activation. Nat Immunol 7:1037–1047 Wang Y, Sun Z, 2009, Current understanding of klotho. Ageing Res Rev 8:43–51 Stone JR, Yang S, 2006, Hydrogen peroxide: a signaling messenger. Antioxid Redox Signal 8:243–270 Mahadev K, Wu X, Zilbering A et al, 2001, Hydrogen peroxide generated during cellular insulin stimulation is integral to activation of the distal insulin signaling cascade in 3T3-L1 adipocytes. J Biol Chem 276:48662–48669 Colavitti R, Pani G, Bedogni B et al, 2002, Reactive oxygen species as downstream mediators of angiogenic signaling by vascular endothelial growth factor receptor-2/KDR. J Biol Chem 277:3101–3108 Wang Y, Sun Z, 2009, Klotho gene delivery prevents the progression of spontaneous hypertension and renal damage. Hypertension 54:810–817 Mitobe M, Yoshida T, Sugiura H et al, 2005, Oxidative stress decreases klotho expression in a mouse kidney cell line. Nephron Exp Nephrol 101:67–74 Kuro-o M, 2008, Klotho as a regulator of oxidative stress and senescence. Biol Chem 389:233–241 Panayiotidis M, 2008, Reactive oxygen species, ROS, in multistage carcinogenesis. Cancer Lett 266:3–5 Lee SB, Cho ES, Yang HS, Kim H, Um HD, 2005, Serum withdrawal kills U937 cells by inducing a positive mutual interaction between reactive oxygen species and phosphoinositide 3-kinase. Cell Signal 17:197–204 Cao Q, Mak KM, Lieber CS, 2006, DLPC and SAMe prevent alpha1(I, collagen mRNA up-regulation in human hepatic stellate cells, whether caused by leptin or menadione. Biochem Biophys Res Commun 350:50–55 Nicco C, Laurent A, Chereau C, Weill B, Batteux F, 2005, Differential modulation of normal and tumor cell proliferation by reactive oxygen species. Biomed Pharmacother 59:169–174 Lopez-Lazaro M, 2007, Dual role of hydrogen peroxide in cancer: possible relevance to cancer chemoprevention and therapy. Cancer Lett 252:1–8 Zielonka J, Kalyanaraman B, 2008, “ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis”–a critical commentary. Free Radic Biol Med 45:1217–1219 Scherz-Shouval R, Shvets E, Fass E et al, 2007, Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J 26:1749–1760 Juhasz G, Erdi B, Sass M, Neufeld TP, 2007, Atg7-dependent autophagy promotes neuronal health, stress tolerance, and longevity but is dispensable for metamorphosis in Drosophila. Genes Dev 21:3061–3066 Kuma A, Hatano M, Matsui M et al, 2004, The role of autophagy during the early neonatal starvation period. Nature 432:1032–1036 Melendez A, Talloczy Z, Seaman M et al, 2003, Autophagy genes are essential for dauer development and life-span extension in C. elegans. Science 301:1387–1391 Yamamoto M, Clark JD, Pastor JV et al, 2005, Regulation of oxidative stress by the anti-aging hormone klotho. J Biol Chem 280:38029–38034 Saito K, Ishizaka N, Mitani H, Ohno M, Nagai R, 2003, Iron chelation and a free radical scavenger suppress angiotensin IIinduced downregulation of klotho, an anti-aging gene, in rat. FEBS Lett 551:58–62 Afanas’ev I, 2010, Reactive oxygen species and age-related genes p66shc, Sirtuin, FOX03 and Klotho in senescence. Oxid Med Cell Longev 3:77–85 Iwasaki Y, Nishiyama M, Taguchi T et al, 2009, Insulin exhibits short-term anti-inflammatory but long-term proinflammatory effects in vitro. Mol Cell Endocrinol 298:25–32 Salminen A, Huuskonen J, Ojala J et al, 2008, Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging. Ageing Res Rev 7:83–105 Salminen A, Kaarniranta K, 2009, Regulation of the aging process by autophagy. Trends Mol Med 15:217–224 Eskelinen EL, Saftig P, 2004, Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim Biophys Acta 1793:664–673 Gozuacik D, Kimchi A, 2004, Autophagy as a cell death and tumor suppressor mechanism. Oncogene 23:2891–2906 Tanida I, Minematsu-Ikeguchi N, Ueno T, Kominami E, 2005, Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy 1:84–91 Bunney TD, KatanM(2010, Phosphoinositide signalling in cancer: beyond PI3K and PTEN. Nat Rev Cancer 10:342–352 Huang J, Manning BD, 2009, A complex interplay between Akt, TSC2 and the two mTOR complexes. Biochem Soc Trans 37:217– 222 Degtyarev M, De Maziere A, Orr C et al, 2008, Akt inhibition promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic agents. J Cell Biol 183:101–116 Arico S, Petiot A, Bauvy C et al, 2001, The tumor suppressor PTEN positively regulates macroautophagy by inhibiting the phosphatidylinositol 3-kinase/protein kinase B pathway. J Biol Chem 276:35243–35246 Mammucari C, Milan G, Romanello Vet al, 2007, FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 6:458–471 Iida RH, Kanko S, Suga T,MoritoM, Yamane A, 2011, Autophagiclysosomal pathway functions in the masseter and tongue muscles in the klotho mouse, a mouse model for aging. Mol Cell Biochem 348:89–98 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 611 EP 617 DI 10.1007/s12253-013-9663-8 PG 7 ER PT J AU Orosz, E Ember, I Gombos, K Toth, L Tarpay, Pap, Otto, Sz AF Orosz, Eniko Ember, Istvan Gombos, Katalin Toth, Laszlo Tarpay, Adam Pap, Akos Otto, Szabolcs TI Alternatives for the Intensive Follow-Up After Curative Resection of Colorectal Cancer. Potential Novel Biomarkers for the Recommendations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Colorectal cancer; Follow-up; New strategy; New biomarkers ID Colorectal cancer; Follow-up; New strategy; New biomarkers AB Early diagnosis of recurrence and metastasis of colorectal cancer following surgery of curative intent is of vital importance in terms of survival and quality of life. The consistent implementation of appropriate patient follow-up strategy is therefore essential. Debates over the methodology, evaluation and strategy of follow-up have been known for many years, and continue today. By introducing several follow-up models, the present paper offers different options featuring certain individual, national and international, conceptual and financial aspects. Colorectal cancer is an important public health concern due to its destructive nature and frequency, it is therefore essential to develop new monitoring strategies, involving new biomarkers and extensive clinical validation. Since the recurrence rate is very high in high-risk patients, the improvement of individual patient risk estimates and the utilization of a corresponding follow-up model require broad international co-operation and common practice, along with the determination of optimal levels of evidence. C1 [Orosz, Eniko] National Institute of Oncology, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Ember, Istvan] University of Pecs, Faculty of Medicine, Department of Public HealthPecs, Hungary. [Gombos, Katalin] University of Pecs, Faculty of Medicine, Department of Public HealthPecs, Hungary. [Toth, Laszlo] National Institute of Oncology, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Tarpay, Adam] National Institute of Oncology, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Pap, Akos] National Institute of Oncology, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Otto, Szabolcs] National Institute of Oncology, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. RP Otto, Sz (reprint author), National Institute of Oncology, 1122 Budapest, Hungary. EM sz.otto@oncol.hu CR Bray F, Sankila F, Ferlay J et al, 2002, Estimates ef cancer incidence and mortality in Europe in 1995. Eur J Cancer 38:99–166 Ferlay J, Autier P, Boniol M et al, 2007, Estimates of the cancer incidence and mortality in Europe in 2006. Ann Oncol 18:581–592 Otto S, 2003, Cancer epidemiology in Hungary and the Bela Johan National Program for the decade of health. Pathol Oncol Res 2:126–130 Yang L, Fujimoto J, Qiu D et al, 2010, Trends in cancer mortality in the elderly in Japan, 1970–2007. Ann Oncol 21:389–396 Bosetti C, Levi F, Rosato Vet al, 2011, Recent trends in colorectal cancer mortality in Europe. Int J Cancer 129:180–191 Malvezzi M, Bertuccio P, Levi F et al, 2012, European cancer mortality predictions for the year 2012. Ann Oncology 23:1044– 1052 Verdecchia A, Francisci S, Brenner H et al, 2007, Recent cancer survival in Europe: a 2000–02 period analysis of EUROCARE-4 data. Lancet Oncol 8:784–796 Majek O, Gondos A, Jansen L et al, 2012, Survival from colorectal cancer in Germany in the early 2 Ist century. Br J Cancer 106:1875–1880 Rachet B, Woods LM, Mitry E et al, 2008, Cancer survival in England and Wales at the end of the 20th century. Br J Cancer 99:S2–S10 Aslam MI, Taylor K, Pringle JH et al, 2009, MicroRNAs are novel biomarkers of colorectal cancer. Br J Surg 96:702–710 La Vecchia C, Bosetti C, Lucchini F et al, 2010, Cancer mortality in Europe, 2000–2004, and overview of trends since 1975. Ann Oncol 21:1323–1360 Tusnady G, Gaudi I, Rejto L et al, 2008, Survival chances of Hungarian cancer patients in the National Cancer Registry. Hungarian Oncol 52:339–349 Olesen F, Hansen RP, Vedsted P, 2009, Delay in diagnosis: the experience in Denmark. Br J Cancer 101:55–58 Schmoll HJ, Van Cutsem E, Stein A et al, 2012, ESMO Consensus Guidelines for management of patients with colon and rectal cancer. A personalized approach to clinical decision making. Ann Oncol 23:2479–2516 Baca B, Beart RW, Etzioni DA, 2011, Surveillance after colorectal cancer resection: a systematic review. Dis Colon Rectum 54:1036– 1048 Levin B, Liberman DA, McFarland B et al, 2008, American Cancer Society Colorectal Cancer Advisory Group. Screening and surveillance for the early detection of colorectal cancer and adenomatous polyps, 2008: a joint guideline from the American Cancer Society, the US Multy-Society Task Force on colorectal cancer, and the American College of Radiology. Gastroenterology 134:1570–1595 Dunn BK,Wagner PD, Anderson D et al, 2010, Molecular markers for early detection. Semin Oncol 37:224–242 Diamandis EP, 2010, Cancer Biomarkers: can we turn recent failures into success? J Natl Cancer Inst 102:1462–1467 Bertagnolli MM, 2009, The forest and the trees:pathways and proteins as colorectal cancer biomarkers. J Clin Oncol 27:5866– 5867 Jung Y, Lee S, Choi H-S et al, 2011, Clinical validation of colorectal cancer biomarkers identified from bioinformatics analysis of public expression data. Clin Cancer Res 17:700–709 Gutman S, Kessler LG, 2006, The US Food and Drug Administration perspective on cancer biomarker development. Nature Rev/ Cancer 6:565–571 McShane LM, Hayes DF, 2012, Publication of tumos marker research result: the necesity for complete and transparent reporting. J Clin Oncol 30:4223–4232 Labianca R, Nordlinger B, Beretta GD et al, 2010, Primary colon cancer: ESMO Clinical Practice Guidelines for diagnosis, adjuvant treatment and follow-up. Ann Oncol 21(Suppl 5):v70–v77 Griesenberg D, Nurnberg R, Bahlo M et al, 1999, CEA, TPS, CA 19–9 and CA 72–4 and the fecal occult blood test in the preoperative diagnosis and follow-up after resective surgery of colorectal cancer. Anticancer Res 19:2443–2450 Hohnbec JRL, Topolcan O, Pikner R et al, 2000, The significance of CEA, CA 19–9 and CA 72–4 in the detection of colorectal carcinoma recurrence. Anticancer Res 20:5237–5244 Lane JM, Chow E, Young GP et al, 2010, Interval fecal immunochemical testing in a colonoscopic surveillance program speeds detection of colorectal neoplasia. Gastroenterology 139:1918– 1926 Morris AM, Gryfe R, Thorson AG et al, 2011, Canadian Association of General Surgeons, the American College of Surgeons, the Canadian Society of Colorectal Surgeons, and the American Society of Colorectal Surgeons: evidence based review in surgery – colorectal surgery. Dis Colon Rectum 54:1598–1600 Engstrom PF, Arnoletti JP, Benson AP et al, 2009, NCCN Clinical Practice Guidelines in Oncology: colon cancer. J Natl Compr Cancer Netw 7:778–831 Criblez D, 2007, Revidierte Konsensus-Empfehlungen zur Nachsorge nach koloskopischer Polypektomie und nach kurativ operiertem kolorektalem Karzinom. Schweiz Med Forum 7:994–999 Viehl CT, Ochsner A, von Holzen U et al, 2010, Inadequate quality of surveillance after curative surgery for colon cancer. Ann Surg Oncol 17:2663–2669 Tsitikis VL, Maliraldy K, Green EA et al, 2009, Postoperative surveillance recommendations for early stage colon cancer based on result from the clincal outcomes of surgical therapy trial. J Clin Oncol 27:3671–3676 Desch CE, Benson AB III, Somerfield MR et al, 2005, Colorectal Cancer Surveillance: 2005 Update of an American Society of Clinical Oncology Practice Guideline. J Clin Oncol 23:8512– 8519 Fora A, Patta A, Attwood K et al, 2012, Intensive radiographic and biomarker surveillance in stage II and III colorectal cancer. Oncology 82:41–47 Andreoni B, Chiappa A, Bertani E et al, 2007, Surgical outcomes for colon and rectal cancer over a decade: results from a consecutive monocentric experience in 902 unselected patients. World J Surg Oncol 5:73 Jeffrey M, Hickey BE, Hider PN, 2007, Follow-up strategies for patients treated for non-metastatic colorectal cancer. Cochrane Database Syst Rev CD002200 Yakabe T, Nakafusa Y, Sumi K et al, 2010, Clinical significance of CEA and CA19-9 in postoperative follow-up of colorectal cancer. Ann Surg Oncol 17:2349–2356 Chang GJ, Kaiser AM, Mills S et al, 2012, Practice parameters for the management of colon cancer. Dis Colon Rectum 55:831–843 Borie F, Daures JP, Millat B et al, 2004, Cost and effectiveness of follow-up examinations in patients with colorectal cancer resected for cure in a French population-based study. J Gastrointest Surg 8:552–558 Hara M, Sato M, Takahashi H et al, 2010, Does serum Carcinoembryonic Antigen elevation in patients with postoperatve stage II colorectal cancer indicate recurrence ? Comparison with stage III. J Surg Oncol 102:154–157 Renehan AG, Egger M, Saunders MP et al, 2002, Impact on survival of intensive follow-up after curative resection for colorectal cancer: systematic review and meta-analysis of randomised trials. Br Med J 324:813 Holt A, Nelson RA, Lai L, 2010, Surveillance with serial serum carcinoembryonic levels detect colorectal cancer recurrences in patients who are initial nonsecretors. Am Surg 76:1100–1103 Mulder SA, Van Leerdam ME, Ouwendijk RJ et al, 2007, Attendance at surveillance endoscopy of patients with adenoma or colorectal cancer. Scand J Gastroenterol 42:66–71 Herzlinger RE, Parsa-Parsi R, 2004, Consumer-driven health care: lessons from Switzerland. JAMA 292:1213–1220 Grossmann I, de Bock GH, van de Velde et al, 2007, Resalts of a national survey among Dutch surgeons treating patients with colorectal carcinoma. Current opinion about follow-up, treatment of metastasis, and reasons to revise follow-up practice. Colorect Dis 9:782–792 Locker G, Hamilton S, Harris J et al, 2006, ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol 24:5313–5327 Van der Geest LGM, Krijnen P, Wouters MWJM et al, 2012, Improved guideline compliance after a 3-year audit of multidisciplinary colorectal cancer care in the western part of the Netherlands. J Surg Oncol 106:1–9 Ho JM, Juurlink DN, Cavalcanti RB, 2010, Hypokalemia following polyethylene glycol-based bowel preparation for colonoscopy in older hospitalized patients with significant comorbidities. Ann Pharmacother 44:466–470 Miller A, Steele S, 2012, Novell molecular screening approaches in corectal cancer. J Surg Oncol 105:459–467 Stoop EM, de Haan MC, de Wijkerslooth TR et al, 2012, Participation and yield of colonoscopy versus non-cathartic CT colonography in population-based screening for colorectal cancer: a randomised controlled trial. Lancet Oncol 13:55–64 Barbas AS, Turley RS, Mantyh CR et al, 2012, Effect of surgeon specialization on long-term survival following. Colon cancer resection at an NCI - designated cancer center. J Surg Oncol 106:219– 223 Jones RP, Jackson R, Dunne DFJ et al, 2012, Systematic review and meta-analysis of follow-up after hepatectomy for colorectal liver metastases. Br J Surg 99:477–486 Lyass S, Zamir G, Matot I et al, 2001, Combined colon and hepatic resection for synchronous colorectal liver metastases. J Surg Oncol 78:17–21 Gomez D, Sangha VK, Morris-Stiff G, 2010, Outcomes of intensive surveillance after resection of hepatic colorectal metastases. Br J Surg 97:1552–1560 Tarantino I, Warschkow R, Worni M et al, 2012, Elevated preoperative CEA is associated with worse survival in stage I-III cancer patiens. Br J Cancer 107:266–274 Lin JK, Lin CC, Yang SH et al, 2011, Early postoperative CEA level is a better prognostic indicator than is preoperative CEA level in predicting prognosis of patiens with curable colorectal cancer. Int J Colorectal Dis 26(9):1135–1141 Biomarkers Definitions Working Group, 2001, Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 69:89–95 Saijo N, 2012, Critical comments for roles of biomarkers. Cancer Treat Rev 38:63–67 Trape J, Fiella X, Alsina-Donadeu et al, 2011, Increased plasma concentrations of tumour markers in the absence of neoplasia. Clin Chem Lab Med 49(10):1605–1620 Canil CM, Tannok IF, 2002, Doctor’s dilemma: incorporating tumor markers into clinical decision-making. Semin Oncol 29:286–293 Kern SE, 2012, Why your new cancer biomarker may never work: recurrent patterns and remarkable diversity in biomarker failures. Cancer Ress 72(23):6097–6101 Lecomte T, Ceze N, Dorval E et al, 2010, Circulating free tumor DNA and colorectal cancer. Gastroen Clin Biol 34:662–681 Ryan BM, Lefort F, McManus R et al, 2003, A prospective study of circulating mutant KRAS2 in the serum of patients with colorectal neoplasia: strong prognostic indicator in postoperatve folow-up. Gut 52:101–108 Rose JS, Serna DS, Martin KL, 2012, Influence of KRAS mutation status in metachronous and synchronous metastatic colorectal adenocarcinoma. Cancer 118:6243–6252 De Vos T, Tetzner R, Model F et al, 2009, Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem 55:1337–1346 Matsubara N, 2012, Epigenetic regulation and colorectal cancer. Dis Colon Rectum 55:96–104 Cassinotti E, Melson J, Ligetti T et al, 2012, DNA methylation patters in blood of patients with colorectal cancer and adenomatous colorectal polyps. Int J Cancer 131:1153–1157 Ahlquist DA, Zou H, Domanico M et al, 2012, Next-generation stool DNA test accurately detects colorectal cancer and large adenomas. Gastroenterol 142:248–256 Miranda E, Bianchi P, Destro A et al, 2013, Genetic and epigenetic alterations in primary colorectal cancers and related lymph node and liver metastases. Cancer 119:266–276 Baldus SE, Schaefer KL, Engels R et al, 2010, Prevalence and heterogeneity of KRAS, BRAF, and PIK3CA mutations in primary colorectal adenocarcinomas and their corresponding metastases. Clin Cancer Res 16:790–799 Cohen SJ, Punt CJA, Iannotti N et al, 2008, Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol 26:3213–3221 Lowes LE, Hegley BD, KeeneyMet al, 2012, User-defined protein marker assay development for characterization of circulating tumor cells using the Cell Search® System. Cytometry Part A 81A:983– 995 Sato N, Hayashi N, Imamura Y et al, 2012, Usefulness of Transcription-Reserve Transcription Concerted reaction method for detecting ciculating tumor cells in patiens with colorectal cancer. Ann Surg Oncol 19:2060–2065 Flamini E, Mercatali L, Nanni O et al, 2006, Free DNA and carcinoembryonic antigen serum levels: an important combination for diagnosis of colorectal cancer. Clin Cancer Res 12:6985–6988 Phillipp AB, Stieber P, Nagel D et al, 2012, Prognostic role of methylated free circulating DNA in colorectal cancer. Int J Cancer 131:2308–2319 Chan C-C, Fan C-W, Kuo Y-B et al, 2010, Multiple serological biomarkers for colorectal cancer detection. Int J Cancer 126:1683– 1690 Lawrie CH, Gal S, Dunlop HM et al, 2008, Detection of elevated levels of tumour-associatedmicroRNAs in serum of patients with diffuse large B-cell lymphoma. Br J Haematol 141:672– 675 Huang Z, Huang D, Ni S et al, 2010, Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. Int J Cancer 127:118–126 Brase J,Wuttig D, Kuner R et al, 2010, Serum microRNAs as noninvasive biomarkers for cancer. Mol Cancer 9:306 Shibuya H, Linuma H, Shimada R et al, 2010, Clinicopathological and prognostic value of microRNA-21 and microRNA-155 in colorectal cancer. Oncology 79:313–320 Nishida N, Yamashita S, Minori K et al, 2012, MicroRNA-10b is a prognostic indicator in colorectal cancer and confers resistance to the chemotherapeutic agent 5-Fluorouracil in colorectal cancer cells. Ann Surg Oncol 19:3065–3071 Schwarzenbach H, Hoon DSB, Pantel K, 2011, Cell-free nucleic acids as biomarkers in cancer patients. Nature Rev/Cancer 11:426–436 Kosaka N, Iguchi H, Ochiya T, 2010, Circulating microRNA in body fluid: a new potential biomarker for cancer diagnosis and prognosis. Cancer Sci 101:2087–2092 Mitchell PS, Parkin RK, Kroh EM et al, 2008, Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci 105:10513–10518 Nair VS, Maeda LS, Ioannidis JPA, 2012, Clinical outcome prediction by microRNAs in human cancer. A systematic review. Reviews/ JNCI 104:528–540 Chen C, Ba Y, Ma L et al, 2008, Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 18:997–1006 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 619 EP 629 DI 10.1007/s12253-013-9672-7 PG 11 ER PT J AU Patki, J Pawar, SS AF Patki, M. Jyoti Pawar, S Sagar TI HSP90: Chaperone-me-not SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Hsp90; Cancer hallmarks; Hsp90 inhibitors; Tumor selectivity; Client proteins; Inhibitor classes ID Hsp90; Cancer hallmarks; Hsp90 inhibitors; Tumor selectivity; Client proteins; Inhibitor classes AB With increasing understanding of the molecular basis of carcinogenesis, its progression and metastasis, the cancer therapy has shifted from empirical approaches to targeting specific molecules that regulate the complex network of signalling pathways for cell survival and proliferation. These include key players in malignant transformation like protein kinases, transcription factors, steroid hormone receptors, cell cycle regulators, signal transduction proteins and regulators of apoptosis. Almost all these proteins depend upon the molecular chaperone Hsp90 for their proper folding, stability and function and thus are a part of the Hsp90 clientele. Dependence of these proteins on Hsp90 makes this chaperone an appealing target for cancer therapeutics. Inhibition of Hsp90 can affect multiple oncogenic pathways simultaneously. Moreover Hsp90 inhibitors selectively kill cancer cells compared to normal cells and cancer cells have greater dependence on Hsp90 for the maintenance of intracellular protein homeostasis. All this has led to a rapid pace discovery of Hsp90 clients as well as chemical inhibitors of Hsp90. The role of hsp90 in cancer, tumor selectivity of Hsp90 inhibitors and the current status of Hsp90 inhibitors are discussed in the present review. C1 [Patki, M. Jyoti] Padmashree Dr D.Y. Patil University, Department of Biotechnology and Bioinformatics, Sector-15, Plot-50, CBD BelapurNavi Mumbai, India. [Pawar, S Sagar] University of Mumbai, Chikitsak Samuha’s Patkar-Varde College, Department of Zoology, S.V. Road, Goregaon (W)Mumbai, India. RP Patki, J (reprint author), Padmashree Dr D.Y. Patil University, Department of Biotechnology and Bioinformatics, Navi Mumbai, India. EM jyotitope8@gmail.com CR Neckers L, Ivy SP, 2003, Heat shock protein 90. Curr Opin Oncol 15:419–424 Chiosis G, Vilenchik M, KimJ, Solit D, 2004, Hsp90: the vulnerable chaperone. Drug Discov Today 9:881–888 Goetz MP, Toft DO, Ames MM, Erichman C, 2003, The Hsp90 chaperone complex as a novel target for cancer therapy. Ann Oncol 14:1169–1176 Li Y, Zhang T, Sun D, 2009, New developments in Hsp90 inhibitors as anti-cancer therapeutics: mechanisms, clinical perspective and more potential. Drug Resist Updat 12(1–2):17–27 Wandinger SK, Richter K, Buchner J, 2008, The Hsp90 chaperone machinery. J Biol Chem 283:18473–18477., DOI 10.1074/jbc. R800007200 Richter K, Soroka J, Skalniak L, Leskovar A, Hessling M, Reinstein J, Buchner J, 2008, Conserved conformational changes in the ATPase cycle of human Hsp90. J Biol Chem 283:17757–17765 Pearl LH, Prodromou C, Workman P, 2008, The Hsp90 molecular chaperone: an open and shut case for treatment. Biochem J 410:439– 453 Dollins ED, Warren JJ, Immormino RM, Gewirth DT, 2007, Structures of GRP94-nucleotide complexes reveal mechanistic differences between the hsp90 chaperones. Mol Cell 28:41–56 Travers J, Sharp S, Workman P, 2012, HSP90 inhibition: twopronged exploitation of cancer dependencies. Drug Discov Today 00, 00,, in press) Kasibhatla S, Tseng B, 2003, Why target apoptosis in cancer treatment? Mol Cancer Ther 2:573–580 Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100:57–70 Hanahan D, Weinberg RA, 2011, The hallmarks of cancer: the next generation. Cell 144:646–674., DOI 10.1016/j.cell.2011.02.013 Bagatell R, Whitesell L, 2004, Altered Hsp90 function in cancer: a unique therapeutic opportunity. Mol Cancer Ther 3(8):1021–1030 McCarthy MM, Pick E, Kluger Y, Gould-Rothberg B, Lazova R, Camp RL, 2008, HSP90 as a marker of progression in melanoma. Ann Oncol 19:590–594 Workman P, Burrows F, Neckers L, Rosen N, 2007, Drugging the cancer chaperone HSP90: combinatorial therapeutic exploitation of oncogene addiction and tumor stress. AnnNYAcad Sci 1113:202–216 Sidera K, Patsavoudi E, 2009, Extracellular Hsp90: an emerging target for cancer therapy. Curr Signal Transduct Ther 4:51–58 Sreedhar AS, Kalmar E, Csermely P, Shen YF, 2004, Hsp90 isoforms: functions, expression and clinical importance. FEBS Lett 562:11–15 Neckers L, 2002, Hsp90 inhibitors as novel cancer chemotherapeutic agents. Trends Mol Med 8:S55–S61 Workman P, 2004, Combinatorial attack on multistep oncogenesis by inhibiting the Hsp90 molecular chaperone. Cancer Lett 206:149–157 PrattWB, 1998, The Hsp90 based chaperone system: involvement in signal transduction from a variety of hormone and growth factor receptors. Proc Soc Exp Biol Med 217:420–434 Neckers L, 2007, Heat shock protein 90: the cancer chaperone. J Biosci 32:517–530 Jaattela M, 1999, Escaping cell death: survival proteins in cancer. Exp Cell Res 248:30–43 Pasquale EB, 2008, Eph-ephrin bi-directional signaling in physiology and disease. Cell 133(1):38–52 Bohonowych JE, Gopal U, Issacs JS, 2010, Hsp90 as a gatekeeper of tumor angiogenesis: clinical promise and potential pitfalls. J Oncol., DOI 10.1155/2010/412985 Powers MV, Workman P, 2006, Targeting the multiple signaling pathways by heat shock protein 90 molecular chaperone inhibitors. Endocr Relat Cancer 13(1):125–135 Neckers L, 2006, Using natural product inhibitors to validate HSP90 as a molecular target in cancer. Curr Med Chem 6:1163–1171 Zhang H, Burrows F, 2004, Targeting multiple signal transduction pathways through inhibition of Hsp90. J Mol Med 82:488–499 Gava LM, Ramos CHI, 2009, Human 90Kda Heat shock protein Hsp90 as a target for cancer therapeutics. Curr Chem Biol 3:330–341 Issacs JS, 2005, Heat shock protein 90 inhibitors in antineoplastic therapy: is it all wrapped up? Expert Opin Investig Drugs 14(6):569– 589 Pecorino L, 2008, Molecular biology of cancer.Mechanisms, targets and therpeutics, 2nd edn. Oxford University Press Vilenchik M, Solit D, Basso M, Huezo H, Lucas B, Huazhong H, Rosen N, Spampinato C, Modrich P, Chiosis G, 2004, Targeting widerange oncogenic transformation via PU24FCl, a specific inhibitor of tumor Hsp90. Chem Biol 11:787–797 Kamal A, Thao L, Sensintaffar J, Zhang L, Boehm MF, Fritz LC, Burrows FJ, 2003, A high-affinity conformation of Hsp90 confers tumor selectivity on Hsp90 inhibitors. Nature 425:407–410 Workman P, 2003, Auditing the pharmacological accounts for hsp90 molecular chaperone inhibitors: unfolding the relationship between pharmacokinetics and pharmacodynamics.Mol Cancer Ther 2:131–138 Workman P, 2003, Overview: translating Hsp90 biology into Hsp90 drugs. Curr Cancer Drug Targets 3:297–300 Chiosis G, Neckers L, 2006, Tumor selectivity of Hsp90 inhibitors— the explanat-ion remains elusive. ACS Chem Biol 1:279–284 Maroney AC et al, 2006, Dihydroquinone ansamycins: toward resolving the conflict between low in vitro affinity and high cellular potency of geldanamycin derivatives. Biochemistry 45:5678–5685 Solit DB, Rosen N, 2006, Hsp90: a novel target for cancer therapy. Curr Top Med Chem 6:1205–1214 Xu W, Yuan X, Beebe K et al, 2007, Loss of Hsp90 association upregulates Src dependent ErbB2 activity. Mol Cell Biol 27:220–228 Sharma S et al, 1998, Targeting of the protein chaperone, HSP90, by the transformation suppressing agent, radicicol. Oncogene 16:2639– 2645 Burlingson JA et al, 2007, Development of Novobiocin analogues that manifest anti-proliferative activity against several cancer cell lines. J Org Chem 73:2130–2137 Donnelly A, Blagg BSJ, 2008, Novobiocin and Hsp90 inhibitors of C terminal nucleotide binding pocket. CurrMed Chem 15(26):2702– 2717 Wangl Yet al, 2010, STA-9090AsmallmoleculeHsp90 inhibitor for the potential treatment of cancer. Curr Opin Investig Drugs 11(12):1466–1476 Hastings JM, Hadden MK, Blagg BSJ, 2008, Synthesis and evaluation of Derrubone and select analogues. J Org Chem 73:369–373 Nicoll M, 2008, XL 888, a novel synthetic, orally bio-available inhibitor of Hsp90., Presented at the AACR-NCI-EORTC International Conference, Molecular Targets and Cancer Therapeutics, Discovery, Biology, and Clinical Applications, October 21–24, 2008, Geneva, Switzerland. Exelixis Research and Development, Exelixis inc; South San rancisco, CA, USA) Wettstein D et al, 2008, MPC-3100: a non-natural product Hsp90 inhibitor with anti-tumor activity in pre-clinical models. 20th EORTC-NCI-AACR, October 21–24 in Geneva, Switzerland AmolinsMW, Blagg BSJ, 2009, Natural product inhibitors of Hsp90: potential leads for drug discovery.Mini Rev Med Chem 9(2):140–152 Vasko RC et al, 2010, Mechanistic studies of Sansalvamide A-Amide: an allosteric modulator of Hsp90. ACS Med Chem Lett 1:4–8 Brandt GEL et al, 2008, Gedunin, a novel Hsp90 inhibitor: semi synthesis of derivatives and preliminary structure activity relationships. J Med Chem 51(20):6495–6502., DOI 10.1021/jm8007486 Winssinger N, Barluenga S, 2007, Chemistry and biology of resorcyclic acid lactones. Chem Commun :22–36., DOI 10.1039/ b610344h Rodriguez RA, 2008, Structure-activity of Sansalvamide A derivatives and their apoptotic activity in pancreatic cancer cell line PL-45. Mex Chem Soc 52(3):201–211 Yi F, Regan L, 2008, A novel class of small molecule inhibitors of Hsp90. ACS Chem Biol 3(10):645–654 Sun X, Kenney SC, 2010, Hsp90 inhibitors: a potential treatement for latent EBV infection? Cell Cycle 9(9):1665–1666 Gorska M et al, 2012, Geldanamycin and its derivatives as Hsp90 inhibitors. Front Biosci 17:2269–2277 Shapiro G, 2011, STA-9090, Ganetespib, and AT13387. 9th International Symposium on Targeted Cancer Therapies Paris, France Trepel J et al, 2010, Targeting the dynamicHsp90 complex in cancer. Nat Rev Cancer 10:537–549., DOI 10.1038/nrc2887 Eskew JD, 2011, Development and characterization of novel Cterminal inhibitor of Hsp90 in androgen dependent and independent prostate cancer cells. BMC Cancer 11:468 Mereles D, Hunstein W, 2011, Epigallocatechin-3-gallate, EGCG, for clinical trials:more pitfalls than promises? Int JMol Sci 12:5592– 5603., DOI 10.3390/ijms12095592 Shelton SN et al, 2009, KU135 A novel novobiocin derived C-terminal inhibitor of 90 Kda heat shock protein exerts potent anti-proliferative effects on human luekemic cells. Mol Pharmacol 76(6):1314–1322 Chiosis G et al, 2002, Development of a purine-scaffold novel class of Hsp90 binders that inhibit the proliferation of cancer cells and induce the degradation of Her2 tyrosine kinase. Bioorg Med Chem 10:3555–3564 Ho N, Li A, Li S, Zhang H, 2012, Heat shock protein 90 and role of its chemical inhibitors in treatment of hematologic malignancies. Pharmaceuticals 5:779–801., DOI 10.3390/ph5080779 Piper PW, Millson SH, 2011, Mechanisms of resistance to Hsp90 inhibitor drugs: a complex mosaic emerges. Pharmaceuticals 4:1400– 1422 Dymock BW, Barrill X, Brough PA et al, 2005, Novel, potent smallmolecule inhibitors of the molecular chaperone Hsp90 discovered through structure-based design. J Med Chem 48:4212–4215 Stuhmer T et al, 2008, Signalling profile and anti-tumor activity of the novel Hsp90 inhibitor NVP-AUY922 in multiple myeloma. Leukemia 22:1604–1612 Samuel T et al, 2010, AUY922 a novel Hsp90 inhibitor: final results of a first-in-human study in patients with advanced solid malignancies. Am Soc Clin Oncol Ann Meet 46:Abs 2528 Okawa Yet al, 2009, SNX-2112, a selective hsp90 inhibitor, potently inhibits tumor cell growth, angiogenesis and osteoclastogenesis in multiple myeloma and other hematologic tumors by abrogating signalling via Akt and Erk. Blood 113:846–855 Richardson et al, 2010, Br J Haematol 150:438–445 Neckers L, Workman P, 2012, Hsp90 molecular chaperone inhibitors: are we there yet? Clin Cancer Res 18(1):64–76., DOI 10.1158/ 1078-0432.CCR-11-1000 Zhang H, Chung D, Yang YC, 2006, Identification of new biomarkers for clinical trials of Hsp90 inhibitors. Mol Cancer Ther 5:1256–1264., DOI 10.1158/1535-7163.MCT-05-0537 Smith-Jones PM et al, 2006, Early tumor response to Hsp90 therapy using HER2 PET: comparison with 18F-FDG PET. J Nucl Med 47:793–796 Solit DB, Chiosis G, 2008, Development and applications of Hsp90 inhibitors. Drug Discov Today 13(1/2):38–43 Oikonomopoulou K et al, 2009, Evaluation of prostate-specific antigen as a novel biomarker of Hsp90 inhibition. Clin Biochem 42(8):16–17 Adeela K, Burrows FJ, 2009, Hsp90 inhibitors as selective anticancer drugs. Discov Med. http://www.discoverymedicine.com/Adeela- Kamal/2009/07/12/hsp90-inhibitors-as-selectable-anticancer-drugs/ Sliutz G, Karlseder J, Tempfer C, Orel L, Holzer G, Simon M, 1996, Drug resistance against gemcitabine and topotecan mediated by constitutive Hsp70 overexpression in vitro: implication of quercetin as sensitizer in chemotherapy. Br J Cancer 74:172– 177 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 631 EP 640 DI 10.1007/s12253-013-9675-4 PG 10 ER PT J AU Qiang, F Guangguo, R Yongtao, H Dandan, D Hong, Y AF Qiang, Fang Guangguo, Ren Yongtao, Han Dandan, Dong Hong, Yang TI Multidrug Resistance in Primary Tumors and Metastases in Patients with Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal cancer; Lymph node metastasis; Multidrug resistance; Protein expression ID Esophageal cancer; Lymph node metastasis; Multidrug resistance; Protein expression AB Studies have demonstrated that radical esophagectomy can significantly prolong disease-free survival and improve the survival rate of patients with T3 or T4 esophageal cancer and lymph node metastasis. Multidrug resistant cancer cells have active efflux mechanisms that prevent the accumulation of chemotherapeutic drugs in the cells. The purpose of this study was to compare the expression of five MDR related proteins between primary tumors in patients with thoracic esophageal squamous cell carcinoma (ESCC) and metastatic cancer in lymph nodes to explore the clinical significance of heterogeneity in MDR metastatic cancer cells. Fifty-four patients with ESCC and lymph node metastasis were included. All patients underwent subtotal esophagectomy and D2/D3 lymph node resection. The expression of lung resistance-related protein (LRP), Pglycoprotein, topoisomerase-II, thymidylate synthase, and glutathione S-transferase P1–1 (GST-π) were determined in the primary tumors and lymph nodes via immunohistochemistry. The expression of LRP was significantly different between the primary tumors and lymph nodes (P=0. 026). No significant differences were found for the other four proteins, and protein expression was not associated with either degree of differentiation or disease stage. It was also found that GST-π was expressed in all patients in both the primary tumors and lymph nodes, suggesting that the design and application of chemotherapeutic protocols capable of reducing GST-π expression may be beneficial for patients with ESCC. Additional research regarding the clinical utility of MDR protein expression in ESCC is warranted to design effective chemotherapeutic protocols. C1 [Qiang, Fang] Cancer Hospital of Sichuan Province, Department of Thoracic Surgery, No. 55, 4th section of Remin South Rd, 610000 Chengdu, Sichuan province, China. [Guangguo, Ren] Cancer Hospital of Sichuan Province, Department of Thoracic Surgery, No. 55, 4th section of Remin South Rd, 610000 Chengdu, Sichuan province, China. [Yongtao, Han] Cancer Hospital of Sichuan Province, Department of Thoracic Surgery, No. 55, 4th section of Remin South Rd, 610000 Chengdu, Sichuan province, China. [Dandan, Dong] The People’s Hospital of Sichuan Province, Department of PathologyChengdu, China. [Hong, Yang] The People’s Hospital of Sichuan Province, Department of PathologyChengdu, China. RP Qiang, F (reprint author), Cancer Hospital of Sichuan Province, Department of Thoracic Surgery, 610000 Chengdu, China. EM fangqiang111@126.com CR Wolfard A, Paszt A, Szentpali K et al, 2011, Efficacy and drawbacks of neoadjuvant chemoradiotherapy in squamous cell carcinoma of the thoracic esophagus. Hepatogastroenterology 58:1214– 1219 Lee PC, Port JL, Paul S et al, 2009, Predictors of long-term survival after resection of esophageal carcinoma with nonregional nodal metastases. Ann Thorac Surg 88:186–192 Liu HC, Hung SK, Huang CJ et al, 2005, Esophagectomy for locally advanced esophageal cancer, followed by chemoradiotherapy and adjuvant chemotherapy. World J Gastroenterol 11:5367–5372 Shiozaki A, Yamagishi H, Itoi H et al, 2005, Long-term administration of low-dose cisplatin plus 5-fluorouracil prolongs the postoperative survival of patients with esophageal cancer. Oncol Rep 13:667–672 Ando N, Iizuka T, Ide H et al, 2003, Surgery plus chemotherapy compared with surgery alone for localized squamous cell carcinoma of the thoracic esophagus: a Japan Clinical Oncology Group Study–JCOG9204. J Clin Oncol 21:4592–4596 Lee J, Lee KE, Im YH et al, 2005, Adjuvant chemotherapy with 5- fluorouracil and cisplatin in lymph node-positive thoracic esophageal squamous cell carcinoma. Ann Thorac Surg 80:1170–1175 Ando N, Kato H, Igaki H et al, 2012, A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5- fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus, JCOG9907). Ann Surg Oncol 19:68–74 Pottgen C, Stuschke M, 2012, Radiotherapy versus surgery within multimodality protocols for esophageal cancer–a meta-analysis of the randomized trials. Cancer Treat Rev 38:599–604 Yamamoto Y, Yamai H, Seike J et al, 2012, Prognosis of esophageal squamous cell carcinoma in patients positive for human epidermal growth factor receptor family can be improved by initial chemotherapy with docetaxel, fluorouracil, and cisplatin. Ann Surg Oncol 19:757–765 Donnenberg VS, Donnenberg AD, 2005, Multiple drug resistance in cancer revisited: the cancer stem cell hypothesis. J Clin Pharmacol 45:872–877 Takebayashi Y, Akiyama S, Natsugoe S et al, 1998, The expression of multidrug resistance protein in human gastrointestinal tract carcinomas. Cancer 82:661–666 Tiwari AK, Sodani K, Dai CL et al, 2011, Revisiting the ABCs of multidrug resistance in cancer chemotherapy. Curr Pharm Biotechnol 12:570–594 Stewart DJ, 2010, Tumor and host factors that may limit efficacy of chemotherapy in non-small cell and small cell lung cancer. Crit Rev Oncol Hematol 75:173–234 Ikeda K, Oka M, Narasaki F et al, 1998, Lung resistance-related protein gene expression and drug sensitivity in human gastric and lung cancer cells. Anticancer Res 18:3077–3080 Watson RG, Muhale F, Thorne LB et al, 2010, Amplification of thymidylate synthetase in metastatic colorectal cancer patients pretreated with 5-fluorouracil-based chemotherapy. Eur J Cancer 46:3358–3364 Burgess DJ, Doles J, Zender L et al, 2008, Topoisomerase levels determine chemotherapy response in vitro and in vivo. Proc Natl Acad Sci U S A 105:9053–9058 Gan SY, Zhong XY, Xie SM, 2010, Expression and significance of tumor drug resistance related proteins and betacatenin in esophageal squamous cell carcinoma. Chin J Cancer 29:300–305 Becker TE, Ellsworth RE, Deyarmin B et al, 2008, The genomic heritage of lymph node metastases: implications for clinical management of patients with breast cancer. Ann Surg Oncol 15:1056–1063 Fleming ID, Cooper JS, Henson DE, 1997, Digestive system: Esophagus. In: Flemming ID, ed, AJCC Cancer Staging Manual, 5th edn. Lippincott Williams & Wilkins, Philadelphia, pp 65–69 Sabattini E, Bisgaard K, Ascani S et al, 1998, The EnVision++ system: a new immunohistochemical method for diagnostics and research. Critical comparison with the APAAP, ChemMate, CSA, LABC, and SABC techniques. J Clin Pathol 51:506–511 Vosse BA, Seelentag W, Bachmann A et al, 2007, Background staining of visualization systems in immunohistochemistry: comparison of the Avidin-Biotin Complex system and the EnVision + system. Appl Immunohistochem Mol Morphol 15:103–107 Kelty CJ, Kennedy CW, Falk GL, 2010, Ratio of metastatic lymph nodes to total number of nodes resected is prognostic for survival in esophageal carcinoma. J Thorac Oncol 5:1467–1471 Zhang HL, Chen LQ, Liu RL et al, 2010, The number of lymph node metastases influences survival and International Union Against Cancer tumor-node-metastasis classification for esophageal squamous cell carcinoma. Dis Esophagus 23:53–58 Zhang J, Chen HQ, Zhang YW et al, 2008, Adjuvant chemotherapy in oesophageal cancer: a meta-analysis and experience from the Shanghai Cancer Hospital. J Int Med Res 36:875–882 Wang LS, Chow KC, Wu YC et al, 2004, Inverse expression of dihydrodiol dehydrogenase and glutathione-S-transferase in patients with esophageal squamous cell carcinoma. Int J Cancer 111:246–251 Gillet JP, Gottesman MM, 2011, Advances in the molecular detection of ABC transporters involved in multidrug resistance in cancer. Curr Pharm Biotechnol 12:686–692 Orina JN, Calcagno AM, Wu CP et al, 2009, Evaluation of current methods used to analyze the expression profiles of ATP-binding cassette transporters yields an improved drug-discovery database. Mol Cancer Ther 8:2057–2066 Shi H, Lu D, Shu Y et al, 2008, Expression of multidrugresistance- related proteins P-glycoprotein, glutathione-Stransferases, topoisomerase-II and lung resistance protein in primary gastric cardiac adenocarcinoma. Cancer Invest 26:344–351 Funke S, Timofeeva M, Risch A et al, 2010, Genetic polymorphisms in GST genes and survival of colorectal cancer patients treated with chemotherapy. Pharmacogenomics 11:33–41 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 641 EP 648 DI 10.1007/s12253-013-9623-3 PG 8 ER PT J AU Kaira, K Toyoda, M Shino, M Sakakura, K Takahashi, K Tominaga, H Oriuchi, N Kanai, Y Oyama, T Chikamatsu, K AF Kaira, Kyoichi Toyoda, Minoru Shino, Masato Sakakura, Koichi Takahashi, Katsumasa Tominaga, Hideyuki Oriuchi, Noboru Kanai, Yoshikatsu Oyama, Tetsunari Chikamatsu, Kazuaki TI Clinicopathological Significance of L-type Amino Acid Transporter 1 (LAT1) Expression in Patients with Adenoid Cystic Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adenoid cystic carcinoma; LAT1; CD98; Salivary gland; Prognostic factor ID Adenoid cystic carcinoma; LAT1; CD98; Salivary gland; Prognostic factor AB The expression of L-type amino acid transporter 1 (LAT1) is correlated with tumor cell growth and survival. However, the clinicopathological significance of LAT1 expression in adenoid cystic carcinoma (ACC) remains to be elucidated. The aim of this study is to investigate the clinicopathological significance of LAT1 expression in ACC. A total of 30 patients with ACC were retrospectively reviewed. Tumor sections were stained by immunohistochemistry for LAT1, p53, and CD98, and cell proliferation and microvessel density (MVD) were determined by Ki-67 and CD34, respectively. High LAT1 and CD98 expression were observed in 27 % (8/30) and 23 % (7/30) of samples, respectively (p>0.999). The high expression of LAT1 was significantly correlated with cell proliferation (Ki-67) and the cell cycle regulator p53. By univariate analysis, solid histological pattern, maxillary tumor site, LAT1, CD98, Ki-67 and p53 were significantly associated with poor prognosis. Multivariate analysis demonstrated that the high expression of LAT1 was an independent prognostic factor for predicting poor prognosis after surgical resection. LAT1 is a promising clinical marker to predict the outcome after surgery in patients with ACC. C1 [Kaira, Kyoichi] Gunma University, Graduate School of Medicine, Department of Medicine and Molecular Science, Showa-machi, 371-8511 Maebashi, Gunma, Japan. [Toyoda, Minoru] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Gunma, Japan. [Shino, Masato] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Gunma, Japan. [Sakakura, Koichi] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Gunma, Japan. [Takahashi, Katsumasa] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Gunma, Japan. [Tominaga, Hideyuki] Gunma University, Gunma Graduate School of Medicine, Department of Molecular ImagingMaebashi, Gunma, Japan. [Oriuchi, Noboru] Gunma University, Graduate School of Medicine, Department of Diagnostic Radiology and Nuclear medicineMaebashi, Gunma, Japan. [Kanai, Yoshikatsu] Osaka University, Graduate School of Medicine, Division of Bio-system PharmacologyOsaka, Japan. [Oyama, Tetsunari] Gunma University, Graduate School of Medicine, Department of Diagnostic PathologyMaebashi, Gunma, Japan. [Chikamatsu, Kazuaki] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Gunma, Japan. RP Kaira, K (reprint author), Gunma University, Graduate School of Medicine, Department of Medicine and Molecular Science, 371-8511 Maebashi, Japan. EM kkaira1970@yahoo.co.jp CR Kokemueller H, Eckardt A, Brachvogel P et al, 2004, Adenoid cystic carcinoma of the head and neck-a 20 years experience. Int J Oral Maxillofac Surg 33:25–31 Perzin KH, Gullane P, Clairmont AC, 1978, Adenoid cystic carcinomas arising in salivary glands: a correlation of histologic features and clinical course. Cancer 42:265–282 Christensen HN, 1990, Role of amino acid transport and countertransport in nutrition and metabolism. Physiol Rev 70:43–77 McGivan JD, Pastor-Anglada M, 1994, Regulatory and molecular aspects ofmammalian amino acid transport. Biochem J 299:321–334 Kanai Y, Segawa H, Miyamoto K et al, 1998, Expression cloning and characterization of a transporter for large neutral amino acids activated by the heavy chain of 4F2 antigen, CD98). J Biol Chem 273:23629–23632 Yanagida O, Kanai Y, Chairoungdua A et al, 2001, Human L-type amino acid transporter 1, LAT 1): characterization of function and expression in tumor cell lines. Biochim Biophys Acta 1514:291– 302 Kobayashi H, Ishii Y, Takayama T, 2005, Expression of L-type amino acid transporter 1, LAT1, in esophageal carcinoma. J Surg Oncol 90:233–238 Kaira K, Oriuchi N, Imai H et al, 2008, Prognostic significance of L-type amino acid transporter 1 expression in resectable stage I-III nonsmall cell lung cancer. Br J Cancer 98:742–748 Nakanishi K, Ogata S, Matsuo H et al, 2007, Expression of LAT1 predicts risk of progression of transitional cell carcinoma of the upper urinary tract. Virchows Arch 451:681–690 Nawashiro H, Otani N, Shinomiya N et al, 2006, L-type amino acid transporter 1 as a potential molecular target in human astrocytic tumors. Int J Cancer 119:484–492 Sakata T, Ferdous G, Tsuruta T et al, 2009, L-type amino acid transporter 1 as a novel biomarker for high-grade malignancy in prostate cancer. Pathol Int 59:7–18 Ichinoe M, Mikami T, Yoshida T et al, 2011, High expression of L-type amino-acid transporter 1, LAT1, in gastric carcinomas: Comparison with non-cancerous lesions. Pathol Int 61:281–289 Furuya M, Horiguchi J, Nakajima H et al, 2012, Correlation of L-type amino acid transporter 1 and CD98 expression with triple negative breast cancer prognosis. Cancer Sci 103:382– 389 Kaira K, Oriuchi N, Takahashi T et al, 2011, LAT1 expression is closely associated with hypoxic markers and mTOR in resected non-small cell lung cancer. Am J Transl Res 3:468– 478 Kaira K, Sunose Y, Arakawa K et al, 2012, Prognostic significance of L-type amino acid transporter 1 expression in surgically resected pancreatic cancer. Br J Cancer 107:632–638 Chikamatsu K, Shino M, Sakakura K et al, 2007, Expression of thymidylate synthase and dihydropyrimidine dehydrogenase in adnoid cystic carcinoma of the head and neck: correlation with clinical outcome. Oral Oncol 43:662–669 Arriaga M, Curtin H, Takahashi H et al, 1990, Staging proposal auditory meatus carcinoma based on preoperative clinical examination and computed tomography findings. Ann Otol Rhinol Laryngol 99:714–721 Norberg-Spaak L, Dardick I, Ledin T, 2000, Adenoid cystic carcinoma: use of cell proliferation, Bcl-2 expression, histologic grade, and clinical stage as predictors of clinical outcome. Head Neck 22:489–497 Zhang J, Peng B, Chen X, 2005, Expression of nuclear factor B, inducible nitric oxide sunthase, and vascular endothelial growth factor in adenoid cystic carcinoma of salivary glands: correlations with the angiogenesis and clinical outcome. Clin Cancer Res 11:7334–7343 Da Cruz Perez DE, de Abreu AF, Nishimoto IN et al, 2006, Prognositc factors in head and neck adenoid cystic carcinoma. Oral Oncol 42:139–146 Garden AS,Weber RS, Morrison WH et al, 1995, The influence of positive margins and nerve invasion in adenoid cystic carcinoma of the head and neck treated with surgery and radiation. Int J Radiat Oncol Phys 32:619–626 Chummun S, Mclean NR, Kelly CG et al, 2001, Adenoid cystic carcinoma of the head and neck. Br J Plast Surg 54:476–480 Carlinfante G, Lazzaretti M, Ferrari S et al, 2005, p53, bcl-2 and Ki-67 expression in adenoid cystic carcinoma of the palate. A clinoco-pathologic study of 21 cases with long-term follow-up. Pathol Res Pract 200:791–799 Kaira K, Oriuchi N, Imai H et al, 2009, Prognostic significance of L-type amino acid transporter 1, LAT1, and 4F2hc heavy chain, CD98, expression in stage I pulmonary adenocarcinoma. Lung Cancer 66:120–126 Jaso J, Mlhotra R, 2011, Adenoid cystic carcinoma. Arch Pathol Lab Med 135:511–515 Laurie SA, Ho AL, Fury MG et al, 2011, Systemic therapy in the management of metastatic or locally recurrent adenoid cystic carcinoma of the salivary glands: a systemic review. Lancet Oncol 12:815–824 Park NS, Kim SG, Kim HK et al, 2008, Characterization of amino acid transport system L in HTB-41 human salivary gland epidermoid carcinoma cell. Anticancer Res 28:2649–2656 Kim CH, ParK KJ, Park JR et al, 2006, The RNA interference of amino acid transporter LAT1 inhibits the growth of KB human oral cancer cells. Anticancer Res 26:2943–2948 Kim CS, Cho SH, Chun HS et al, 2008, BCH, an inhibitor of system L amino acid transporters, induces apoptosis in cancer cells. Biol Pharm Bull 31:1096–1100 Imai H, Kaira K, Oriuchi N et al, 2010, Inhibition of L-type amino acid transporter 1 has antitumor activity in non-small cell lung cancer. Anticancer Res 30:4819–4828 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 649 EP 656 DI 10.1007/s12253-013-9624-2 PG 8 ER PT J AU Sikalidis, KA Fitch, DM Fleming, ESh AF Sikalidis, K Angelos Fitch, D Mark Fleming, E Sharon TI Diet Induced Obesity Increases the Risk of Colonic Tumorigenesis in Mice SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Obesity; Colon cancer; C57BL/6J; Diet induced obesity; AOM ID Obesity; Colon cancer; C57BL/6J; Diet induced obesity; AOM AB A large body of epidemiological data indicates that obesity increases the risk of colon cancer in humans. There are limited studies using rodent models where the relationship between obesity and colon cancer has been studied. In this study, wild-type diet-induced obese (DIO) mice and lean wild-type controls were used to investigate the influence of obesity on the risk of colon cancer. We hypothesized that the obese phenotype would exhibit increased colonic tumorigenesis. Colon cancer was chemically induced by injecting the mice with azoxymethane (AOM) at levels that we experimentally determined to result in equivalent AOM concentrations in circulating blood. Risk of colon cancer was assessed via microscopic examination of entire colons for aberrant crypts, aberrant crypt foci and proliferation levels. The DIO mice were found to have significantly more aberrant crypts and aberrant crypt foci as well as increased proliferation of colonocytes per mouse compared to wild-type control mice, supporting the epidemiological data that obesity increases the risk of colonic tumorigenesis. C1 [Sikalidis, K Angelos] University of California, Department of Nutritional Sciences and Toxicology, 94720 Berkeley, CA, USA. [Fitch, D Mark] University of California, Department of Nutritional Sciences and Toxicology, 94720 Berkeley, CA, USA. [Fleming, E Sharon] University of California, Department of Nutritional Sciences and Toxicology, 94720 Berkeley, CA, USA. RP Sikalidis, KA (reprint author), University of California, Department of Nutritional Sciences and Toxicology, 94720 Berkeley, USA. EM angelos_sikalidis@dfci.harvard.edu CR Murphy TK, Calle EE, Rodriguez C, Khan HS, Thun MJ, 2000, Body mass index and colon cancer mortality in a large prospective study. Am J Epidemiol 152(9):847–854 Larson SC, Wolk A, 2007, Obesity and colon and rectal cancer risk: a meta-analysis of prospective studies. Am J Clin Nutr 86(3):556–565 Wolin KY, Yan Y, Colditz GA, 2011, Physical activity and risk of colon adenoma: a meta-analysis. Br J Cancer 104(5):882–885 Oxantenko AS, Bardia A, Vierkant RA, Wang AH, Anderson KE et al, 2010, Body size and incident colorectal cancer: a prospective study of older women. Cancer Prev Res, Phila, 3(12):1608–1620 Martinez ME, Giovannucci E, Spiegelman D, Hunter DJ, Willett WC, Colditz GA, 1997, Leisure-time physical activity, body size, and colon cancer in women. Nurses’ health study research group. J Natl Cancer Inst 89(13):948–955 Bird CL, Frankl HD, Lee ER, Haile RW, 1998, Obesity, weight gain, large weight changes, and adenomatous polyps of the left colon and rectum. Am J Epidemiol 147(7):670–680 Nock NL, Plummer SJ, Thompson CL, Casey G, Li L, 2011, FTO polymorphisms are associated with adult body mass index, BMI, and colorectal adenomas in African-Americans. Carcinogenesis 32(5):748–756 Stein B, Anderson JC, Rajapakse R, Alpern ZA, Messina CR, Walker G, 2010, Body mass index as a predictor of colorectal neoplasia in ethnically diverse screening population. Dig Dic Sci 55(10):2945–2952 Ford ES, 1999, Body mass index and colon cancer in a national sample of adult US men and women. Am J Epidemiol 150(4):390– 398 Caan BJ, Coates AO, Slattery ML, Potter JD, Quesenberry CP Jr, Edwards SM, 1998, Body size and the risk of colon cancer in a large case-control study. Int J Obes Relat Metab Disord 22(2):178– 184 Russo A, Franceschi S, La Vecchia C, Dal Maso L, Montella M et al, 1998, Body size and colorectal-cancer risk. Int J Cancer 78(2):161–165 Wolin KY, Carson K, Colditz GA, 2010, Obesity and cancer. Oncologist 15(6):556–565 Giacosa A, Franceschi S, La Vecchia C, Favero A, Andreatta R, 1999, Energy intake, overweight, physical exercise and colorectal cancer risk. Eur J Canc Prev 8(Suppl 1):S53–S60 Franco A, Sikalidis AK, Solis Herruzo JA, 2005, Colorectal cancer: influence of diet and lifestyle factors. Rev Esp Enferm Dig 97(6):432–448 Sikalidis AK, Varamini B, 2011, Roles of hormones and signaling molecules in describing the relationship between obesity and colon cancer. Pathol Oncol Res 17(4):785–790 Lee WM, Lu S, Medline A, Archer MC, 2001, Susceptibility of lean and obese Zucker rats to tumorigenesis induced by N-methyl- N-nitrosourea. Cancer Lett 162(2):155–160 Guerre-Millo M, Gervois P, Raspe E, Madsen L, Poulain P et al, 2000, Peroxisome proliferator-activated receptor alpha activators improve insulin sensitivity and reduce adiposity. J Biol Chem 275(22):16638–16642 Tsubura A, Lai YC, Miki H, Sasaki T, Uehara N et al, 2011, Review: animal models of N-Methyl-N-nitrosourea-induced mammary cancer and retinal degeneration with special emphasis on therapeutic trials. Vivo 25(1):11–22 Hirose Y, Hata K, Kuno T, Yoshida K, Sakata K et al, 2004, Enhancement of development of azoxymethane-induced colonic premalignant lesions in C57BL/KsJ-db/db mice. Carcinogenesis 25(5):821–825 Pilvi TK, Storvik M, Louhelainen M, Merasto S, Korpela R, Mervaala EM, 2009, Effect of dietary calcium and dairy proteins on the adipose tissue gene expression profile in diet-induced obesity. J Nutrigenet Nutrigenomics 1(5):240–251 Bartolomucci A, Bresciani E, Bulgarelli I, Rigamonti AE, Pascucci T et al, 2009, Chronic intracerebroventricular injection of TLQP- 21 prevents high fat diet induced weight gain in fast weightgaining mice. Genes Nutr 4(1):49–57 Huang X-F, Han M, South T, Storlien L, 2003, Altered levels of POMC, AgRP and MC4-R mRNA expression in the hypothalamus and other parts of the limbic system of mice prone or resistant to chronic high-energy diet-induced obesity. Brain Res 992(1):9–19 Huang X-F, Xin X, McLennan P, Storlien L, 2004, Role of fat amount and type in ameliorating diet-induced obesity: insights at the level of hypothalamic arcurate nucleus leptin receptor, neuropeptide Y and pro-opiomelanocortin mRNA expression. Diabetes Obes Metab 6(1):35–44 Petro AE, Cotter J, Cooper DA, Peters JC, Surwit SJ, 2004, Fat, carbohydrate and calories in the development of diabetes and obesity in the C57BL/6J mouse. Metabolism 53(4):454–457 Moraes RC, Blondet A, Birkenkamp-Demtroeder K, Tirard J, Orntoft TF et al, 2003, Study of the alteration of gene expression in adipose tissue of diet-induced obesity, DIO, mice by microarray and RT-PCR analyses. Endocrinology 144(11):4773–4782 Kwon O, Chung HY, Yu W, Bae HI, Chae YS et al, 2012, Clinical significance of insulin-like growth factor gene polymorphisms with survival in patients with gastrointestinal stromal tumors. J Korean Surg Soc 82(5):288–295 Weber RV, Stein DE, Scholes J, Kral JG, 2000, Obesity potentiates AOM-induced colon cancer. Dig Dis Sci 45(5):890–895 Corpet DE, Tache S, 2002, Most effective colon cancer chemopreventive agents in rats: a systematic review of aberrant crypt foci and tumor data, ranked by potency. Nutr Canc 43(1):1–21 Roncucci L, Pedroni M, Vaccina F, Benatti P, Marzona L, De Pol A, 2000, Aberrant crypt foci in colorectal carcinogenesis. Cell and crypt dynamics. Cell Prolif 33(1):1–18 Bird PR, Good CK, 2000, The significance of aberrant crypt foci in understanding the pathogenesis of colon cancer. Toxicol Lett 112–113:295–402 Paulsen JE, Steffensen I-L, Namork E, Alexander J, 1994, Scanning electron microscopy of aberrant crypt foci in rat colon. Carcinogenesis 15(10):2371–2373 Lacy ER, Kuwayama H, Cowart KS, King JS, Deutz AH, Sistrunk S, 1991, A rapid, accurate, immunohistochemical method to label proliferating cells in the digestive tract. A comparison with tritiated thymidine. Gastroenterology 100(1):259–262 Garcia SB, da Costa Barros LT, Turatti A, Martinello F, Modiano P et al, 2006, The anti-obesity agent Orlistat is associated to increase in colonic preneoplastic markers in rats treated with a chemical carcinogen. Cancer Lett 240(2):221–224 Frankenberry KA, Somasundar P, McFadden DW, Vona-Davis LC, 2004, Leptin induces cell migration and the expression of growth factors in human prostate cancer cells. Am J Surg 188(5):560–565 Cleary MP, Grande JP, Maihle NJ, 2004, Effect of high fat diet on body weight and mammary tumor latency in MMTV-TGF-alpha mice. Int J Obes Relat Metab Dis 28(8):956–962 Newmark HL, Yang K, Lipkin M, Kopelovich L, Liu Y et al, 2001, Western-style diet induces benign and malignant neoplasms in the colon of normal C57Bl/6 mice. Carcinogenesis 22(11):1871–1875 Thornton WH Jr, MacDonald RS, 1997, Dietary fat quantity and composition induce changes in proliferation and membrane lipids in rat colon cells. Ann Nutr Metab 41(4):260–268 Slattery ML, Potter J, Caan B, Edwards S, Coates A et al, 1997, Energy balance and colon cancer—Beyond physical activity. Cancer Res 57(1):75–80 Slattery ML, Lundgreen A, Bondurant KL, Wolff RK, 2011, Tumor necrosis factor-related genes and colon and rectal cancer. Int J Mol Epidemiol Genet 2(4):328–338 Newmark HL, Yang K, Kurihara N, Fan K, Augenlicht LH, Lipkin M, 2009, Western-style diet-induced colonic tumors and their modulation by calcium and vitamin D in C57Bl/6 mice: a preclinical model for human sporadic colon cancer. Carcinogenesis 30(1):88–92 Kiely JM, Noh JH, Pitt HA, Swartz-Basile DA, 2005, Impaired intestinal cell proliferation and cell death in leptin-deficient obese mice. J Parent Ent Nutr 29(1):30–35 Sohn OS, Fiala ES, Requeijo SP, Weisburger JH, Gonzales FJ, 2001, Differential effects of CYP2E1 status on the metabolic activation of the colon carcinogens azoxymethane and methylazoxymethanol. Cancer Res 61(23):8435–8440 Steinbach G, Heymsfield S, Olansen NE, Tighe A, Holt PR, 1994, Effect of caloric restriction on colonic proliferation in obese persons: implications for colon cancer prevention. Cancer Res 54(5):1194–1197 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 657 EP 666 DI 10.1007/s12253-013-9626-0 PG 10 ER PT J AU Huang, GL Qiu, JH Li, BB Wu, JJ Lu, Y Liu, XY He, Z AF Huang, Guo-Liang Qiu, Jin-Hua Li, Bin-Bin Wu, Jing-Jing Lu, Yan Liu, Xing-Yan He, Zhiwei TI Prolyl Isomerase Pin1 Regulated Signaling Pathway Revealed by Pin1 +/+ and Pin1 −/− Mouse Embryonic Fibroblast Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pin1; Pathway; Cancer; Immune ID Pin1; Pathway; Cancer; Immune AB Pin1 (peptidylprolyl cis/trans isomerase, NIMA-interacting 1) plays a key role in a number of diseases including cancer and Alzheimer disease. Previous studies have identified a wide range of phosphoproteins as Pin1 substrates. Related pathways were analyzed separately. The aim of this study was to provide a comprehensive picture involving Pin1 regulation. A genome-wide mRNA expression microarray was carried out using the RNA isolation from Pin1 +/+ and Pin1 −/− mouse embryonic fibroblast (MEF) cells. Signaling pathways regulated by Pin1 were analyzed with the utility of KEGG pathway and GO annotation. An expression pattern regulated by Pin1 was revealed. A total of 606 genes, 375 being up-regulated and 231 down-regulated, were differentially expressed when comparing Pin1 +/+ to Pin1 −/− MEF cells. Totally 48 pathways were shown to be regulated by Pin1 expression in KEGG pathway analysis. In the GO annotation system, 19 processes on biological processes, 15 processes on cellular components, and 18 processes on molecular functions were found to be in the regulation of Pin1 expression. Pathways related to immune system and cancer showed most significant association with Pin1 regulation. Pin1 is an important regulator in a wide range of signaling pathways that were related to immune system and cancer. C1 [Huang, Guo-Liang] Guangdong Medical University, Cancer Research Institute, No. 1 Xincheng Road, 523808 Dongguan, China. [Qiu, Jin-Hua] Huizhou First Hospital, Department of Neurology, 516000 Huizhou, China. [Li, Bin-Bin] Guangdong Medical University, Cancer Research Institute, No. 1 Xincheng Road, 523808 Dongguan, China. [Wu, Jing-Jing] Guangdong Medical University, Cancer Research Institute, No. 1 Xincheng Road, 523808 Dongguan, China. [Lu, Yan] Guangdong Medical University, Cancer Research Institute, No. 1 Xincheng Road, 523808 Dongguan, China. [Liu, Xing-Yan] Guangdong Medical University, Cancer Research Institute, No. 1 Xincheng Road, 523808 Dongguan, China. [He, Zhiwei] Guangdong Medical University, Cancer Research Institute, No. 1 Xincheng Road, 523808 Dongguan, China. RP He, Z (reprint author), Guangdong Medical University, Cancer Research Institute, 523808 Dongguan, China. EM zhiweihe688@yahoo.com CR Lu KP, Hanes SD, Hunter T, 1996, A human peptidyl-prolyl isomerase essential for regulation of mitosis. Nature 380(6574):544–547 Lu PJ, Zhou XZ, Shen M, Lu KP, 1999, Function ofWWdomains as phosphoserine- or phosphothreonine-binding modules. Science 283(5406):1325–1328 Ranganathan R, Lu KP, Hunter T, Noel JP, 1997, Structural and functional analysis of the mitotic rotamase Pin1 suggests substrate recognition is phosphorylation dependent. Cell 89(6):875–886 Yaffe MB, Schutkowski M, Shen M, Zhou XZ, Stukenberg PT, Rahfeld JU, Xu J, Kuang J, KirschnerMW, Fischer G, Cantley LC, Lu KP, 1997, Sequence-specific and phosphorylation-dependent proline isomerization: a potential mitotic regulatory mechanism. Science 278(5345):1957–1960 Liou YC, Ryo A, Huang HK, Lu PJ, Bronson R, Fujimori F, Uchida T, Hunter T, Lu KP, 2002, Loss of Pin1 function in the mouse causes phenotypes resembling cyclin D1-null phenotypes. Proc Natl Acad Sci U S A 99(3):1335–1340 Ryo A, Nakamura M, Wulf G, Liou YC, Lu KP, 2001, Pin1 regulates turnover and subcellular localization of beta-catenin by inhibiting its interaction with APC. Nat Cell Biol 3(9):793–801 Zacchi P, Gostissa M, Uchida T, Salvagno C, Avolio F, Volinia S, Ronai Z, Blandino G, Schneider C, Del Sal G, 2002, The prolyl isomerase Pin1 reveals a mechanism to control p53 functions after genotoxic insults. Nature 419(6909):853–857 Zheng H, You H, Zhou XZ, Murray SA, Uchida T, Wulf G, Gu L, Tang X, Lu KP, Xiao ZX, 2002, The prolyl isomerase Pin1 is a regulator of p53 in genotoxic response. Nature 419(6909):849–853 Wulf GM, Ryo A, Wulf GG, Lee SW, Niu T, Petkova V, Lu KP, 2001, Pin1 is overexpressed in breast cancer and cooperates with Ras signaling in increasing the transcriptional activity of c-Jun towards cyclin D1. EMBO J 20(13):3459–3472 Moretto-Zita M, Jin H, Shen Z, Zhao T, Briggs SP, Xu Y, 2010, Phosphorylation stabilizes Nanog by promoting its interaction with Pin1. Proc Natl Acad Sci U S A 107(30):13312–13317 Monje P, Hernandez-Losa J, Lyons RJ, Castellone MD, Gutkind JS, 2005, Regulation of the transcriptional activity of c-Fos by ERK. A novel role for the prolyl isomerase PIN1. J Biol Chem 280(42):35081–35084 Pathan N, Aime-Sempe C, Kitada S, Basu A, Haldar S, Reed JC, 2001, Microtubule-targeting drugs induce bcl-2 phosphorylation and association with Pin1. Neoplasia 3(6):550–559 Liou YC, Zhou XZ, Lu KP, 2011, Prolyl isomerase Pin1 as a molecular switch to determine the fate of phosphoproteins. Trends Biochem Sci 36(10):501–514 Lee TH, Pastorino L, Lu KP, 2011, Peptidyl-prolyl cis-trans isomerase Pin1 in ageing, cancer and Alzheimer disease. Expert Rev Mol Med 13:e21 Finn G, Lu KP, 2008, Phosphorylation-specific prolyl isomerase Pin1 as a new diagnostic and therapeutic target for cancer. Curr Cancer Drug Targets 8(3):223–229 Fujimori F, Takahashi K, Uchida C, Uchida T, 1999, Mice lacking Pin1 develop normally, but are defective in entering cell cycle from G(0, arrest. Biochem Biophys Res Commun 265(3):658–663 Patterson TA, Lobenhofer EK, Fulmer-Smentek SB, Collins PJ, Chu TM, Bao W, Fang H, Kawasaki ES, Hager J, Tikhonova IR, Walker SJ, Zhang L, Hurban P, de Longueville F, Fuscoe JC, Tong W, Shi L, Wolfinger RD, 2006, Performance comparison of onecolor and two-color platforms within the MicroArray Quality Control, MAQC, project. Nat Biotechnol 24(9):1140–1150 Guo Y, Guo H, Zhang L, Xie H, Zhao X, Wang F, Li Z, Wang Y, Ma S, Tao J, Wang W, Zhou Y, Yang W, Cheng J, 2005, Genomic analysis of anti-hepatitis B virus, HBV, activity by small interfering RNA and lamivudine in stable HBV-producing cells. J Virol 79(22):14392–14403 Ang LS, Boivin WA, Williams SJ, Zhao H, Abraham T, Carmine- Simmen K, McManus BM, Bleackley RC, Granville DJ, 2011, Serpina3n attenuates granzyme B-mediated decorin cleavage and rupture in a murine model of aortic aneurysm. Cell Death Dis 2: e209 Rodvold JJ, Mahadevan NR, Zanetti M, 2012, Lipocalin 2 in cancer: when good immunity goes bad. Cancer Lett 316(2):132– 138 Mulero JJ, Pace AM, Nelken ST, Loeb DB, Correa TR, Drmanac R, Ford JE, 1999, IL1HY1: a novel interleukin-1 receptor antagonist gene. Biochem Biophys Res Commun 263(3):702–706 Tanaka K, Imoto I, Inoue J, Kozaki K, Tsuda H, Shimada Y, Aiko S, Yoshizumi Y, Iwai T, Kawano T, Inazawa J, 2007, Frequent methylation-associated silencing of a candidate tumor-suppressor, CRABP1, in esophageal squamous-cell carcinoma. Oncogene 26(44):6456–6468 Takasu S, Mutoh M, Takahashi M, Nakagama H, 2012, Lipoprotein lipase as a candidate target for cancer prevention/ therapy. Biochem Res Int 2012:398697 Bolcun-Filas E, Costa Y, Speed R, Taggart M, Benavente R, De Rooij DG, Cooke HJ, 2007, SYCE2 is required for synaptonemal complex assembly, double strand break repair, and homologous recombination. J Cell Biol 176(6):741–747 Esnault S, Shen ZJ, Malter JS, 2008, Pinning down signaling in the immune system: the role of the peptidyl-prolyl isomerase Pin1 in immune cell function. Crit Rev Immunol 28(1):45–60 Esnault S, Braun RK, Shen ZJ, Xiang Z, Heninger E, Love RB, Sandor M, Malter JS, 2007, Pin1 modulates the type 1 immune response. PLoS One 2(2):e226 Tun-Kyi A, Finn G, Greenwood A, Nowak M, Lee TH, Asara JM, Tsokos GC, Fitzgerald K, Israel E, Li X, Exley M, Nicholson LK, Lu KP, 2011, Essential role for the prolyl isomerase Pin1 in Tolllike receptor signaling and type I interferon-mediated immunity. Nat Immunol 12(8):733–741 Bane FT, Bannon JH, Pennington SR, Campiani G, Williams DC, Zisterer DM, Mc Gee MM, 2009, The microtubule-targeting agents, PBOX-6 [pyrrolobenzoxazepine 7-[(dimethylcarbamoyl)oxy]-6-(2- naphthyl)pyrrolo-[2,1-d], 1,5)-benzoxazepine] and paclitaxel, induce nucleocytoplasmic redistribution of the peptidyl-prolyl isomerases, cyclophilin A and pin1, in malignant hematopoietic cells. J Pharmacol Exp Ther 329(1):38–47 AyalaG,WangD,Wulf G, Frolov A, Li R, Sowadski J,Wheeler TM, Lu KP, Bao L, 2003, The prolyl isomerase Pin1 is a novel prognostic marker in human prostate cancer. Cancer Res 63(19):6244–6251 Yuan WC, Lee YR, Huang SF, Lin YM, Chen TY, Chung HC, Tsai CH, Chen HY, Chiang CT, Lai CK, Lu LT, Chen CH, Gu DL, Pu YS, Jou YS, Lu KP, Hsiao PW, Shih HM, Chen RH, 2011, A Cullin3-KLHL20 Ubiquitin ligase-dependent pathway targets PML to potentiate HIF-1 signaling and prostate cancer progression. Cancer Cell 20(2):214–228 Bao L, Kimzey A, Sauter G, Sowadski JM, Lu KP, Wang DG, 2004, Prevalent overexpression of prolyl isomerase Pin1 in human cancers. Am J Pathol 164(5):1727–1737 Kuramochi J, Arai T, Ikeda S, Kumagai J, Uetake H, Sugihara K, 2006, High Pin1 expression is associated with tumor progression in colorectal cancer. J Surg Oncol 94(2):155–160 Kim CJ, Cho YG, Park YG, Nam SW, Kim SY, Lee SH, Yoo NJ, Lee JY, Park WS, 2005, Pin1 overexpression in colorectal cancer and its correlation with aberrant beta-catenin expression. World J Gastroenterol 11(32):5006–5009 Atkinson GP, Nozell SE, Harrison DK, Stonecypher MS, Chen D, Benveniste EN, 2009, The prolyl isomerase Pin1 regulates the NFkappaB signaling pathway and interleukin-8 expression in glioblastoma. Oncogene 28(42):3735–3745 He J, Zhou F, Shao K, Hang J, Wang H, Rayburn E, Xiao ZX, Lee SW, Xue Q, Feng XL, Shi SS, Zhang CY, Zhang S, 2007, Overexpression of Pin1 in non-small cell lung cancer, NSCLC, and its correlation with lymph node metastases. Lung Cancer 56(1):51–58 Tan X, Zhou F, Wan J, Hang J, Chen Z, Li B, Zhang C, Shao K, Jiang P, Shi S, Feng X, Lv N, Wang Z, Ling Y, Zhao X, Ding D, Sun J, Xiong M, He J, 2010, Pin1 expression contributes to lung cancer: Prognosis and carcinogenesis. Cancer Biol Ther 9(2):111– 119 Saegusa M, Hashimura M, Kuwata T, 2010, Pin1 acts as a modulator of cell proliferation through alteration in NF-kappaB but not beta-catenin/TCF4 signalling in a subset of endometrial carcinoma cells. J Pathol 222(4):410–420 Plotnikov A, Zehorai E, Procaccia S, Seger R, 2011, The MAPK cascades: signaling components, nuclear roles and mechanisms of nuclear translocation. Biochim Biophys Acta 1813(9):1619–1633 Pan CQ, Liou YC, Low BC, 2010, Active Mek2 as a regulatory scaffold that promotes Pin1 binding to BPGAP1 to suppress BPGAP1-induced acute Erk activation and cell migration. J Cell Sci 123(Pt 6):903–916 Nakano A, Koinuma D, Miyazawa K, Uchida T, Saitoh M, Kawabata M, Hanai J, Akiyama H, Abe M, Miyazono K, Matsumoto T, Imamura T, 2009, Pin1 down-regulates transforming growth factor-beta, TGF-beta, signaling by inducing degradation of Smad proteins. J Biol Chem 284(10):6109–6115 Kim MR, Choi HS, Heo TH, Hwang SW, Kang KW, 2008, Induction of vascular endothelial growth factor by peptidylprolyl isomerase Pin1 in breast cancer cells. Biochem Biophys Res Commun 369(2):547–553 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 667 EP 675 DI 10.1007/s12253-013-9629-x PG 9 ER PT J AU Zhou, L Qiu, T Xu, J Wang, T Wang, J Zhou, X Huang, Z Zhu, W Shu, Y Liu, P AF Zhou, Li Qiu, Tianzhu Xu, Jing Wang, Tongshan Wang, Jian Zhou, Xin Huang, Zebo Zhu, Wei Shu, Yongqian Liu, Ping TI miR-135a/b Modulate Cisplatin Resistance of Human Lung Cancer Cell Line by Targeting MCL1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE miR-135a/b; Cisplatin resistance; Apoptosis; MCL1; Lung cancer ID miR-135a/b; Cisplatin resistance; Apoptosis; MCL1; Lung cancer AB microRNAs (miRNAs) are short non-coding RNA molecules, which post-transcriptionally regulate genes expression and play crucial roles in diverse biological processes, such as development, differentiation, apoptosis, and proliferation. Here, we investigated the possible role of miRNAs in the development of drug resistance in human lung cancer cell line. We found that miR-135a/b were downregulated while MCL1 was upregulated in A549/CDDP (cisplatin) cells, compared with the parental A549 cells. In vitro drug sensitivity assay demonstrated that overexpression of miR-135a/b sensitized A549/CDDP cells to cisplatin. The luciferase activity of MCL1 3′-untranslated region-based reporter constructed in A549/CDDP cells suggested that MCL1 was the direct target gene of miR-135a/b. Enforced miR-135a/b expression reduced MCL1 protein level and sensitized A549/CDDP cells to CDDP-induced apoptosis. Taken together, our findings first suggested that hsa-miR-135a/b could play a role in the development of CDDP resistance in lung cancer cell line at least in part by modulation of apoptosis via targeting MCL1. C1 [Zhou, Li] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 300 Guangzhou Road, 210029 Nanjing, China. [Qiu, Tianzhu] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 300 Guangzhou Road, 210029 Nanjing, China. [Xu, Jing] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 300 Guangzhou Road, 210029 Nanjing, China. [Wang, Tongshan] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 300 Guangzhou Road, 210029 Nanjing, China. [Wang, Jian] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 300 Guangzhou Road, 210029 Nanjing, China. [Zhou, Xin] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 300 Guangzhou Road, 210029 Nanjing, China. [Huang, Zebo] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 300 Guangzhou Road, 210029 Nanjing, China. [Zhu, Wei] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 300 Guangzhou Road, 210029 Nanjing, China. [Shu, Yongqian] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 300 Guangzhou Road, 210029 Nanjing, China. [Liu, Ping] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 300 Guangzhou Road, 210029 Nanjing, China. RP Liu, P (reprint author), Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 210029 Nanjing, China. EM liu-ping@csco.org.cn CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127:2893–917 Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM, 2006, Targeting multidrug resistance in cancer. Nat Rev Drug Disc 5:219–34 Rabik CA, Dolan ME, 2007, Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat Rev 33:9–23 Johnstone RW, Ruefli AA, Lowe SW, 2002, Apoptosis: A link between cancer genetics and chemotherapy. Cell 108:153–64 Zhang K, Mack P, Wong KP, 1998, Glutathione-related mechanisms in cellular resistance to anticancer drugs. Int J Oncol 12:871–82 Konopleva M, Tabe Y, Zeng Z, Andreeff M, 2009, Therapeutic targeting of microenvironmental interactions in leukemia: Mechanisms and approaches. Drug Resist Updat 12:103–113 Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeulen L, Iovino F, Tripodo C, Russo A, Gulotta G, Medema JP, Stassi G, 2007, Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4. Cell Stem Cell 1:389–402 Voulgari A, Pintzas A, 2009, Epithelial-mesenchymal transition in cancer metastasis: Mechanisms, markers and strategies to overcome drug resistance in the clinic. Biochim Biophys Acta 1796(2):75–90 Wang B, Yang H, Huang YZ, Yan RH, Liu FJ, Zhang JN, 2010, Biologic characteristics of the side population of human small cell lung cancer cell line H446. Chin J Cancer 29:254–60 Sarkar FH, Li Y, Wang Z, Kong D, Ali S, 2010, Implication of microRNAs in drug resistance for designing novel cancer therapy. Drug Resist Updat 13:57–66 Bartel DP, 2004, MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 116:281–97 Aigner A, 2011, MicroRNAs, miRNAs, in cancer invasion and metastasis: Therapeutic approaches based on metastasis-related miRNAs. J Mol Med, Berl, 89:445–57 Rottiers V, Najafi-Shoushtari SH, Kristo F, Gurumurthy S, Zhong L, Li Y, Cohen DE, Gerszten RE, Bardeesy N, Mostoslavsky R, Naar AM, 2011, MicroRNAs in metabolism and metabolic diseases. Cold Spring Harb Symp Quant Biol 76:225–33 Cho WC, 2010, MicroRNAs: Potential biomarkers for cancer diagnosis, prognosis and targets for therapy. Int J Biochem Cell Biol 42:1273–81 Chen J, Tian W, Cai H, He H, Deng Y, 2011, Down-regulation of microRNA-200c is associated with drug resistance in human breast cancer. Med Oncol., DOI 10.1007/s12032-011-0117-4 Xu K, Liang X, Cui D, Wu Y, Shi W, Liu J, 2011, miR-1915 inhibits Bcl-2 to modulate multidrug resistance by increasing drugsensitivity in human colorectal carcinoma cells. Mol Carcinog., DOI 10.1002/mc.21832 Li H, Hui L, XuW, 2012, miR-181a sensitizes a multidrug-resistant leukemia cell line K562/A02 to daunorubicin by targeting BCL-2. Acta Biochim Biophys Sin 44:269–77 Zhang S,Wan Y, Pan T, Gu X, Qian C, Sun G, Sun L, XiangY,Wang Z, Shi L, 2012, MicroRNA-21 inhibitor sensitizes human glioblastoma U251 stem cells to chemotherapeutic drugTemozolomide. J Mol Neurosci 47:346–56 Ru P, Steele R, Hsueh EC, Ray RB, 2011, Anti-miR-203 upregulates SOCS3 expression in breast cancer cells and enhances cisplatin chemosensitivity. Genes Cancer 2:720–7 Fu X, Tian J, Zhang L, Chen Y, Hao Q, 2012, Involvement of microRNA-93, a new regulator of PTEN/Akt signaling pathway, in regulation of chemotherapeutic drug cisplatin chemosensitivity in ovarian cancer cells. FEBS Lett 586:1279–86 Zhu W, Zhu D, Lu S, Wang T, Wang J, Jiang B, Shu Y, Liu P, 2012, miR-497 modulates multidrug resistance of human cancer cell lines by targeting BCL2. Med Oncol 29:384–91 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), method. Methods 25:402–8 Yamaue H, Tanimura H, Noguchi K, Hotta T, Tani M, Tsunoda T, Iwahashi M, Tamai M, Iwakura S, 1992, Chemosensitivity testing of fresh human gastric cancer with highly purified tumor cells using the MTT assay. Br J Cancer 66:794–9 Yamaue H, Tani M, Onishi H, Kinoshita H, Nakamori M, Yokoyama S, Iwahashi M, Uchiyama K, 2002, Locoregional chemotherapy for patients with pancreatic cancer intraarterial adjuvant chemotherapy after pancreatectomy with portal vein resection. Pancreas 25:366–72 Xia L, Zhang D, Du R, Pan Y, Zhao L, Sun S, Hong L, Liu J, Fan D, 2008, miR-15b and miR-16 modulate multidrug resistance by targeting BCL2 in human gastric cancer cells. Int J Cancer 123:372–9 Quinn BA, Dash R, Azab B, Sarkar S, Das SK, Kumar S, Oyesanya RA, Dasgupta S, Dent P, Grant S, Rahmani M, Curiel DT, Dmitriev I, Hedvat M, Wei J, Wu B, Stebbins JL, Reed JC, Pellecchia M, Sarkar D, Fisher PB, 2011, Targeting Mcl-1 for the therapy of cancer. Expert Opin Investig Drugs 20:1397–411 Wu S, Lin Y, Xu D, Chen J, Shu M, Zhou Y, ZhuW, Su X, Zhou Y, Qiu P, Yan G, 2011, MiR-135a functions as a selective killer of malignant glioma. Oncogene., DOI 10.1038/onc.2011.551 Navarro A, Diaz T, Martinez A, Gaya A, Pons A, Gel B, Codony C, Ferrer G, Martinez C, Montserrat E, Monzo M, 2009, Regulation of JAK2 by miR-135a: Prognostic impact in classic Hodgkin lymphoma. Blood 114:2945–51 Chen Y, Zhang J, Wang H, Zhao J, Xu C, Du Y, Luo X, Zheng F, Liu R, Zhang H, Ma D, 2012, miRNA-135a promotes breast cancer cell migration and invasion by targeting HOXA10. BMC Cancer 12:111 Matsuyama H, Suzuki HI, Nishimori H, Noguchi M, Yao T, Komatsu N, Mano H, Sugimoto K, Miyazono K, 2011, miR- 135b mediates NPM-ALK-driven oncogenicity and renders IL- 17-producing immunophenotype to anaplastic large cell lymphoma. Blood 118:6881–92 Hummel R, Hussey DJ, Haier J, 2010, MicroRNAs: Predictors and modifiers of chemo- and radiotherapy in different tumour types. Eur J Cancer 46:298–311 Perez-Tomas R, 2006, Multidrug resistance: Retrospect and prospects in anti-cancer drug treatment. Curr Med Chem 13:1859–76 Borowski E, Bontemps-Gracz MM, Piwkowska A, 2005, Strategies for overcoming ABC-transporters-mediated multidrug resistance, MDR, of tumor cells. Acta Biochim Pol 52:609–27 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 677 EP 683 DI 10.1007/s12253-013-9630-4 PG 7 ER PT J AU Sun, G Hu, W Lu, Y Wang, Y AF Sun, Guogui Hu, Wanning Lu, Yifang Wang, Yadi TI A Meta-Analysis of HIF-1α and Esophageal Squamous Cell Carcinoma (ESCC) Risk SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ESCC; HIF-1α; Meta-analysis; Survival ID ESCC; HIF-1α; Meta-analysis; Survival AB To investigate the correlation of hypoxiainducible factor-1α (HIF-1α) expression with clinical prognosis and efficacy of radiochemotherapy in esophageal squamous cell carcinoma (ESCC). Studies assessing the clinical or prognostic significance of HIF-1α expression in ESCC published prior to December 2011 were selected by searching PubMed, EMBASE, Cochrane Library, and (China National Knowledge Infrastructure) CNKI. A metaanalysis was performed to clarify the impact of HIF-1α expression on clinicopathological parameters or survival in ESCC. A total of 16 studies met the inclusion criteria, which included 1261 patients with ESCC. Accordingly, the level of HIF-1α expression in esophageal tissues of patients with ESCC was significantly higher than that in normal patients (odds ratio, OR=33.111, 95 % confidence interval, CI= 11.912–92.040). The expression of HIF-1α correlated with the depth of invasion (OR=1.701, 95 % CI=1.076–4.705), clinical TNM stage (OR=2.160, 95%CI=1.516–3.077), as well as lymph node metastasis (OR=2.393, 95 % CI= 1.319–4.344), regardless of differentiation grading (OR= 1.185, 95 % CI=0.859–1.635). Furthermore, there was a significant association of increased HIF-1α expression with poorer radiochemotherapy outcomes, 2-year overall survival (OR=0.219, 95 % CI=0.104–0.461) and survival (OR= 0.320, 95 % CI=0.115–0.887, P<0.05) in patients with ESCC. In addition, HIF-1α expression correlated with VEGF expression in the ESCCs (OR=4.635, 95%CI= 2.591–8.292). Increased expression of HIF-1α plays an important role in the malignant biology of ESCC resulting in significantly poorer radiochemotherapy outcomes and 2- year overall survival. HIF-1α expression may be a prognostic factor, as well as a potential target for therapy in patients with ESCC. C1 [Sun, Guogui] Tangshan People’s Hospital, Department of Chemoradiotherapy, 063000 Tangshan, Hebei Province, China. [Hu, Wanning] Tangshan People’s Hospital, Department of Chemoradiotherapy, 063000 Tangshan, Hebei Province, China. [Lu, Yifang] Tangshan Workers Hospital, Department of Endocrinology (Section I), 063000 Tangshan, Hebei Province, China. [Wang, Yadi] The Military General Hospital of Beijing PLA, Department of Radiotherapy, 100700 Beijing, China. RP Hu, W (reprint author), Tangshan People’s Hospital, Department of Chemoradiotherapy, 063000 Tangshan, China. EM guogui_sun@hotmail.com CR Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics, 2012. CA Cancer J Clin 62:10–29 Zhang GH, Mai RQ, Huang B, 2010, Meta-analysis of ADH1B and ALDH2 polymorphisms and esophageal cancer risk in China. WJG 16:6020–6025 Enzinger PC, Mayer RJ, 2003, Esophageal cancer. N Engl J Med 349:2241–2252 Yang SJ, Yokoyama A, Yokoyama T, Huang YC, Wu SY, Shao Y, Niu J, Wang J, Liu Y, Zhou XQ, Yang CX, 2010, Relationship between genetic polymorphisms of ALDH2 and ADH1B and esophageal cancer risk: a meta-analysis. World J Gastroenterol 16:4210–4220 Liang Y, Liu JL, Wu Y, Zhang ZY, Wu R, 2011, Cyclooxygenase- 2 polymorphisms and susceptibility to esophageal cancer: a metaanalysis. Tohoku J Exp Med 223:137–144 Knechtel G, Szkandera J, Stotz M, Hofmann G, Langsenlehner U, Krippl P, Samonigg H, RennerW, Langner C,DehchamaniD,Gerger A, 2010, Single nucleotide polymorphisms in the hypoxia-inducible factor-1 gene and colorectal cancer risk. Mol Carcinog 49:805–809 Unruh A, Ressel A, Mohamed HG, Johnson RS, Nadrowitz R, Richter E, Katschinski DM, Wenger RH, 2003, The hypoxiainducible factor-1 alpha is a negative factor for tumor therapy. Oncogene 22:3213–3220 Adams JM, Difazio LT, Rolandelli RH, Lujan JJ, Hasko G, Csoka B, Selmeczy Z, Nemeth ZH, 2009, HIF-1 a key mediator in hypoxia. Acta Physiol Hung 96:19–28 Smith TG, Robbins PA, Ratcliffe PJ, 2008, The human side of hypoxia-inducible factor. Br J Haematol 141:325–334 Kuphal S, Winklmeier A, Warnecke C, Bosserhoff AK, 2010, Constitutive HIF-1 activity in malignant melanoma. Eur J Cancer 46:1159–1169 Liu JL, Zhou BJ, Zhang Y, 2007, Expression and biological significance of HPA and HIF-1alpha in human esophageal squamous cell carcinoma. Zhonghua Zhong Liu Za Zhi[Chin J Oncol] 29:689–692 Yu ZT, Zhao HF, Shang XB, 2008, Expression of hypoxiainducible factor-1alpha and vessel endothelial growth factor in esophageal squamous cell carcinoma and clinico-pathological significance thereof. Zhonghua Yi Xue Za Zhi 88:2465–2469 Matsuyama T, Nakanishi K, Hayashi T, Yoshizumi Y, Aiko S, Sugiura Y, Tanimoto T, Uenoyama M, Ozeki Y, Maehara T, 2005, Expression of hypoxia-inducible factor-1alpha in esophageal squamous cell carcinoma. Cancer Sci 96:176–182 Kurokawa T, Miyamoto M, Kato K, Cho Y, Kawarada Y, Hida Y, Shinohara T, Itoh T, Okushiba S, Kondo S, Katoh H, 2003, Overexpression of hypoxia-inducible-factor 1alpha(HIF-1alpha, in oesophageal squamous cell carcinoma correlates with lymph node metastasis and pathologic stage. Br J Cancer 89:1042–1047 Sohda M, Ishikawa H, Masuda N, Kato H, Miyazaki T, Nakajima M, Fukuchi M, Manda R, Fukai Y, Sakurai H, Kuwano H, 2004, Pretreatment evaluation of combined HIF-1alpha, p53 and p21 expression is a useful and sensitive indicator of response to radiation and chemotherapy in esophageal cancer. Int J Cancer 110:838–844 Tzao C, Lee SC, Tung HJ, Hsu HS, Hsu WH, Sun GH, Yu CP, Jin JS, Cheng YL, 2008, Expression of hypoxia-inducible factor, HIF)-1alpha and vascular endothelial growth factor, VEGF)-D as outcome predictors in resected esophageal squamous cell carcinoma. Dis Markers 25:141–148 Ogawa K, Chiba I, Morioka T, Shimoji H, Tamaki W, Takamatsu R, Nishimaki T, Yoshimi N, Murayama S, 2011, Clinical significance of HIF-1alpha expression in patients with esophageal cancer treated with concurrent chemoradiotherapy. Anticancer Res 31:2351–2359 Kimura S, Kitadai Y, Tanaka S, Kuwai T, Hihara J, Yoshida K, Toge T, Chayama K, 2004, Expression of hypoxia-inducible factor, HIF)-1alpha is associated with vascular endothelial growth factor expression and tumour angiogenesis in human oesophageal squamous cell carcinoma. Eur J Cancer 40:1904–1912 Talks KL, Turley H, Gatter KC, Maxwell PH, Pugh CW, Ratcliffe PJ, Harris AL, 2000, The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alp:a in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol 157:411–421 Wu XA, Sun Y, Fan QX, Wang LX, Wang RL, 2007, Relationship between the expression of hypoxia-inducible factor-1α and chemotherapy response in esophageal squamous cell carcinoma. Chinese Med J Peking 87:1783–1786 Zhang HZ, Wang YZ, Xu N, Zhu SC, Liu B, 2007, Expression and clinical significance of HIF-1α, VEGF and Survivin in esophageal squamous cell carcinoma. Chin-Ger J Clin Oncol 6:339–344 Guo LJ, Chen ZJ, Chen CZ, Hong CQ, Wu X, 2009, Expressions of HIF-1α and COX-2 in esophageal squamous cell carcinoma and their correlation with intratumoral MVD. Chin J Radiol Med Prot 29:502–505 Wu XA, Fan QX, Wang LX, Sun Y, Zhao PR, 2006, ReIationship between HIF-lα expression and angiogenesis in esophageal sguamous cell carcinoma tissue. J Zhengzhou Univ 41:1111–1114 Hu JL, Chen P, Liu W, Li ZY, Chen YS, Wu G, 2010, Expression of VEGF16sb and HIF-1α in esophageal squamous carcinoma tissues and their significance. Cancer Res Prev Treat 1:9–11 Carmeliet P, Jain RK, 2011, Molecular mechanisms and clinical applications of angiogenesis. Nature 473:298–307 Valastyan S, Weinberg RA, 2011, Tumor metastasis: molecular insights and evolving paradigms. Cell 147:275–292 Gebski V, Burmeister B, Smithers BM, Foo K, Zalcberg J, Simes J, 2007, Survival benefits from neoadjuvant chemoradiotherapy or chemotherapy in oesophageal carcinoma: a meta-analysis. Lancet Oncol 8:226–234 Zhao J, Li L, Wei S, Gao Y, Chen Y, Wang G, Wu Z, 2011, Clinicopathological and prognostic role of cyclin D1 in esophageal squamous cell carcinoma: a meta-analysis. Dis Esophagus 25:520– 526 Yu WW, Guo YM, Zhu M, Cai XW, Zhu ZF, Zhao WX, Fu XL, 2011, Clinicopathological and prognostic significance of EGFR over-expression in esophageal squamous cell carcinoma: a metaanalysis. Hepatogastroenterology 58:426–431 Fang P, Jiao S, Zhang X, Liu Z, Wang H, Gao Y, Luo H, Chen T, Shi L, 2011, Meta-analysis of ALDH2 variants and esophageal cancer in Asians. Asian Pac J Cancer Prev 12:2623–2627 Miyazawa M, Yasuda M, Fujita M, Kajiwara H, Hirabayashi K, Takekoshi S, Hirasawa T, Murakami M, Ogane N, Kiguchi K, Ishiwata I, Mikami M, Osamura RY, 2009, Therapeutic strategy targeting the mTOR-HIF-1alpha-VEGF pathway in ovarian clear cell adenocarcinoma. Pathol Int 59:19–27 Grebhardt S, Veltkamp C, Strobel P, Mayer D, 2012, Hypoxia and HIF-1 increase S100A8 and S100A9 expression in prostate cancer. Int J Cancer Natsuizaka M, Naganuma S, Kagawa S, Ohashi S, Ahmadi A, Subramanian H, Chang S, Nakagawa KJ, Ji X, Liebhaber SA, Klein-Szanto AJ, Nakagawa H, 2012, Hypoxia induces IGFBP3 in esophageal squamous cancer cells through HIF-1alpha-mediated mRNA transcription and continuous protein synthesis. FASEB J 26:2620–2630 Tang N, Wang L, Esko J, Giordano FJ, Huang Y, Gerber HP, Ferrara N, Johnson RS, 2004, Loss of HIF-1alpha in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis. Cancer Cell 6:485–495 Arany Z, Foo SY, Ma Y, Ruas JL, Bommi-Reddy A, Girnun G, Cooper M, Laznik D, Chinsomboon J, Rangwala SM, Baek KH, Rosenzweig A, Spiegelman BM, 2008, HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1alpha. Nature 451:1008–1012 Buchler P, Reber HA, Buchler MW, Friess H, Lavey RS, Hines OJ, 2004, Antiangiogenic activity of genistein in pancreatic carcinoma cells is mediated by the inhibition of hypoxia-inducible factor- 1 and the down-regulation of VEGF gene expression. Cancer-Am Cancer Soc 100:201–210 Pore N, Jiang Z, Gupta A, Cerniglia G, Kao GD, Maity A, 2006, EGFR tyrosine kinase inhibitors decrease VEGF expression by both hypoxia-inducible factor, HIF)-1-independent and HIF-1- dependent mechanisms. Cancer Res 66:3197–3204 Pore N, Gupta AK, Cerniglia GJ, Maity A, 2006, HIV protease inhibitors decrease VEGF/HIF-1alpha expression and angiogenesis in glioblastoma cells. Neoplasia 8:889–895 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 685 EP 693 DI 10.1007/s12253-013-9631-3 PG 9 ER PT J AU Pather, S Mohamed, Z McLeod, H Pillay, K AF Pather, Sugeshnee Mohamed, Zainab McLeod, Heather Pillay, Komala TI Large Cell Lymphoma: Correlation of HIV Status and Prognosis with Differentiation Profiles Assessed by Immunophenotyping SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Diffuse large B cell lymphoma; Plasmablastic lymphoma; Prognosis; HAART; HIV; CD4; Bcl-2; CD38 ID Diffuse large B cell lymphoma; Plasmablastic lymphoma; Prognosis; HAART; HIV; CD4; Bcl-2; CD38 AB Diffuse large B cell lymphoma (DLBCL) and plasmablastic lymphoma (PBL) represent aggressive non- Hodgkin lymphomas, particularly in the setting of HIV infection. Since the introduction of highly active antiretroviral therapy (HAART), recent studies have documented improved survival outcome in patients with AIDS-related lymphomas. This study contributes a South African perspective by correlating the HIV status and prognosis of DLBCL and PBL with differentiation profiles assessed by immunophenotyping. Analysis of the morphologic, immunophenotypic and clinicopathologic features of 52 cases of DLBCL and 9 cases of de novo PBL was performed. The overall survival of patients with PBL was poorer than that of DLBCL (logrank p value 0.002). Despite HAART, the overall survival with DLBCL and HIV infection was significantly poorer than HIV negative patients with DLBCL (p value <0.001). Profound immunosuppression was evident in the HIV positive group as the mean CD4 count was 151 cells/mm3 in DLBCL and 61 cells/mm3 in PBL. HIV positive patients were significantly younger at presentation with greater likelihood of extranodal lymphoma. When Hans’ and Muris’ algorithmic stratification of DLBCL were applied, no statistical significance was demonstrated (p values 0.188 and 0.399 respectively). However, when Bcl-2 expression occurred in germinal center-type DLBCL (Hans’ defined), improved survival was conferred by the germinal center immunophenotype (p value 0.007). The study demonstrates that DLBCL and PBL have significant potential for aggressive behaviour and poor outcome in the setting of profound immunosuppression due to HIV infection. Further studies are required to assess the effect of targeted-immunotherapy (Rituximab) in combination with recent amendment of the South African national antiretroviral treatment guidelines which has created tremendous potential for improved survival in patients with AIDS-related non-Hodgkin B-cell lymphomas. C1 [Pather, Sugeshnee] University of Cape Town, Groote Schuur Hospital, Division of Anatomical Pathology, National Health Laboratory ServiceCape Town, South Africa. [Mohamed, Zainab] University of Cape Town, Groote Schuur Hospital, Department of Radiation OncologyCape Town, South Africa. [McLeod, Heather] University of Cape Town, Groote Schuur Hospital, Division of Anatomical Pathology, National Health Laboratory ServiceCape Town, South Africa. [Pillay, Komala] University of Cape Town, Red Cross Children’s Hospital, Division of Anatomical Pathology, National Health Laboratory ServiceCape Town, South Africa. RP Pather, S (reprint author), University of Cape Town, Groote Schuur Hospital, Division of Anatomical Pathology, National Health Laboratory Service, Cape Town, South Africa. EM Sugeshnee.pather@nhls.ac.za CR Stein H, Warnke RA, Chan WC, Jaffe ES, Chan JKC, Gatter KC, Campo E, 2008, Diffuse large B cell lymphoma, not otherwise specified. In: Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Thiele J, Vardiman JW, eds, World Health Organisation classification of tumours of haematopoietic and lymphoid tissues, 4th edn. IARC Press, Lyon, pp 233–237 Hoffmann C, Tieman M, Schrader C, Janssen D, Wolf E, Vierbuchen M, Parwaresch R, Ernestus K, Plettenberg A, Stoehr A, Fatkenheuer G, Wyen C, Oette M, Horst HA, 2005, AIDS-related B-cell lymphoma, ARL): correlation of prognosis with differentiation profiles assessed by immunophenotyping. Blood 106:1762–1769 Colomo L, Loong F, Rives S, Pittaluga S, Martinez A, Lopez- Guillermo A, Ojanguren J, Romagosa V, Jaffe ES, Campo E, 2004, Diffuse large B-cell Lymphomas with plasmablastic differentiation represent a heterogeneous group of disease entities. Am J Surg Pathol 28:736–747 Hans CP,Weisenburger DD, Greiner TC, Gascoyne RD, Delabie J, Ott G, Muller-Hermelink HK, Campo E, Braziel RM, Jaffe ES, Pan Z, Farinha P, Smith LM, Falini B, Banham AH, Rosenwald A, Staudt LM, Connors JM, Armitage JO, Chan WC, 2004, Confirmation of the molecular classification of diffuse large Bcell lymphoma by immunohistochemistry using a tissue microarray. Blood 103(1):275–282 Chang CC, McClintock S, Cleveland RP, Trzpuc T,VesoleDH, Logan B, Kajdacsy-Balla A, Perkins SL, 2004, Immunohistochemical expression patterns of germinal center and activation B-cell markers correlate with prognosis in diffuse large B-cell lymphoma. Am J Surg Pathol 28:464–470 Tumwine LK, Agostinelli C, Campidelli C, Othieno E, Wabinga H, Righi S, Falini B, Piccaluga PP, Byarugaba W, Pileri SA, 2009, Immunohistochemical and other prognostic factors in B cell non Hodgkin lymphoma patients, Kampala, Uganda. BMC Clin Pathol 9:1–7 Amen F, Horncastle D, Elderfield K, Banham AH, Bower M, Macdonald D, Kanfer E, Naresh KN, 2007, Absence of cyclin- D2 and Bcl-2 expression within the germinal centre type of diffuse large B-cell lymphoma identifies a very good prognostic subgroup of patients. Histopathology 51:70–79 Muris JJF, Meijer CJLM, Vos W, van Krieken JH, Jiwa NM, Ossenkoppele GJ, Oudejans JJ, 2006, Immunohistochemical profiling based on Bcl-2, CD10 and MUM1 expression improves risk stratification in patients with primary nodal diffuse large B cell lymphoma. J Pathol 208:714–723 Choi WW, Weisenburger DD, Greiner TC, Piris MA, Banham AH, Delabie J, Braziel RM, Geng H, Iqbal J, Lenz G, Vose JM, Hans CP, Fu K, Smith LM, Li M, Liu Z, Gascoyne RD, Rosenwald A, Ott G, Rimsza LM, Campo E, Jaffe ES, Jaye DL, Staudt LM, Chan WC, 2009, A new immunostain algorithm classifies diffuse large B-cell lymphoma into molecular subtypes with high accuracy. Clin Cancer Res 15:5494–5502 Alizadeh AA, EisenMB, Davis RE,Ma C, Lossos IS, Rosenwald A, Boldrick JC, Sabet H, Tran T, Yu X, Powell JI, Yang L, Marti GE, Moore T, Hudson J Jr, Lu L, Lewis DB, Tibshirani R, Sherlock G, Chan WC, Greiner TC, Weisenburger DD, Armitage JO, Warnke R, Levy R, Wilson W, Grever MR, Byrd JC, Botstein D, Brown PO, Staudt LM, 2000, Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 403:503–511 Madan R, Gormley R, Dulau A, Xu D, Walsh D, Ramesh KH, Cannizaro L, Tamas EF, Kumar P, Sparano J, LeValley A, Xue X, Bhattacharyya PK, Ioachim HL, Ratech H, 2006, AIDS and non- AIDS diffuse large B-cell lymphomas express different antigen profiles. Mod Pathol 19:438–446 Banks PM, Keller RH, Li C-Y, White WL, 1978, Case report malignant lymphoma of plasmablastic identity: a neoplasm with both “immunoblastic” and plasma cellular features. Am J Med 64(5):906–909 Delecluse HJ, Anagnostopoulos I, Dallenbach F, Hummel M, Marafioti T, Schneider U, Huhn D, Schmidt-Westhausen A, Reichart PA, Gross U, Stein H, 1997, Plasmablastic lymphomas of the oral cavity: a new entity associated with the human immunodeficiency virus infection. Blood 89:1413–1420 Stein H, Harris NL, Campo E, 2008, Plasmablastic lymphoma. In: Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Thiele J, Vardiman JW, eds, World Health Organisation classification of tumours of haematopoietic and lymphoid tissues, 4th edn. IARC Press, Lyon, pp 256–257 Desai J, Mitnick RJ, Henry DH, Llena J, Sparano JA, 1999, Patterns of central nervous system recurrence in patients with systemic human immunodeficiency virus-associated non- Hodgkin lymphoma. Cancer 86(9):1840–1847 Sarker D, Thirlwell C, Nelson M, Gazzard B, Bower M, 2003, Leptomeningeal disease in AIDS-related non-Hodgkin’s lymphoma. AIDS 17:861–865 Republic of South Africa. Department of Health. National Antiretroviral Treatment Guidelines. Jacana, 2004. Available from http://southafrica.usembassy.gov/media/2004‐doh‐art‐guidelines.pdf Warnke RA, Strauchen JA, Burke JS, Hoppe RT, Campbell BA, Dorfman RF, 1982, Morphologic types of diffuse large B-cell lymphoma. Cancer 50:690–695 Ambinder RF, 2001, Epstein-Barr virus associated lymphoproliferations in the AIDS setting. Eur J Cancer 10:1209– 1216 Nyman H, 2010, Prognostic molecular factors and algorithms in diffuse large B-cell lymphoma. [Academic thesis] University of Helsinki Chan JKC, 2007, Tumors of the lymphoreticular system—part A: the lymph node. In: Fletcher CDM, ed, Diagnostic histopathology of tumors vol 2, 3rd edn. Churchill-Livingstone Elsevier, London, pp 1139–1288 Antinori A, Cingolani A, Alba L, Ammassari A, Serraino D, Ciancio BC, Palmieri F, De Luca A, Larocca LM, Ruco L, Ippolito G, Cauda R, 2001, Better response to chemotherapy and prolonged survival in AIDS-related lymphomas responding to highly active antiretroviral therapy. AIDS 15:1483–1491 Navarro JT, Ribera JM, Oriol A, 2001, Influence of highly active antiretroviral therapy on response to treatment and survival in patients with acquired immunodeficiency syndrome-related non- Hodgkin’s lymphoma treated with CHOP. Br J Haematol 112:909– 915 Besson C, Goubar A, Gabarre J, RozenbaumW, Pialoux G, Chatelet FP, Katlama C, Charlotte F, Dupont B, Brousse N, Huerre M, Mikol J, Camparo P, Mokhtari K, Tulliez M, Salmon‐Ceron D, Boue F, Costagliola D, Raphael M, 2001, Changes in AIDS-related lymphoma since the era of highly active antiretroviral therapy. Blood 98:2339–2344 Seaberg EC, Wiley D, Martinez‐Maza O, Chmiel JS, Kingsley L, Tang Y, Margolick JB, Jacobson LP, Multicenter AIDS Cohort Study, MACS,, 2010, Cancer incidence in the multicenter AIDS cohort study before and during the HAART era 1984–2007. Cancer 116(23):5507–5516 Franceschi S, Lise M, Clifford GM, Rickenbach M, Levi F, Maspoli M, Bouchardy C, 2010, Changing patterns of cancer incidence in the early- and late-HAART periods: the Swiss HIV Cohort Study. Br J Cancer 103(3):416–422 Republic of South Africa. Department of Health. The South African Antiretroviral Treatment Guidelines 2010. Available from http://www.uj.ac.za/EN/CorporateServices/ioha/Documentation/ Documents/ART%20Guideline.pdf van Eeden A. New HIV treatment guidelines: 15 August 2011 [Internet] Available from http://www.samedical.org/newsroom/ media-releases/archived-media-releases/new-hiv-treatmentguidelines- 15-august-2011.html Evison J, Jost J, Ledergerber B, Jost L, Strasser F, Weber R, 1999, HIV-associated non-Hodgkin’s lymphoma: highly active antiretroviral therapy improves remission rate of chemotherapy. AIDS 13:732–734 Thiessard F, Morlat P, Marimoutou C, Labouyrie E, Ragnaud JM, Pellegrin JL, Dupon M, Dabis F, the Groupe d’Epidemiologie Clinique du SIDA en Aquitaine, GECSA,, 2000, Prognostic factors after non-Hodgkin’s lymphoma in patients infected with the human immunodefciency virus: aquitaine cohort, France. Cancer 88:1696–1702 Coiffier B, 2001, Diffuse large cell lymphoma. Curr Opin Oncol 13:325–334 Habermann TM, Weller EA, Morrison VA et al, 2006, Rituximab- CHOP versus CHOP alone or with maintenance rituximab in older patients with diffuse large B-cell lymphoma. J Clin Oncol 24:3121–3127 Pfreundschuh M, Trumper L, Osterborg A et al, 2006, CHOPlike chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with good-prognosis diffuse large-B-cell lymphoma: a randomised controlled trial by the MabThera International Trial, MInT, Group. Lancet Oncol 7:379–391 Sehn LH, Donaldson J, Chhanabhai M et al, 2005, Introduction of combined CHOP plus rituximab therapy dramatically improved outcome of diffuse large B-cell lymphoma in British Columbia. J Clin Oncol 23:5027–5033 Hockenbery D, Nunez G, Milliman C et al, 1990, Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature 348:334–336 Cory S, 1995, Regulation of lymphocyte survival by the bcl-2 gene family. Annu Rev Immunol 13:513–543 Reed JC, 1997, Bcl-2 family proteins: regulators of apoptosis and chemoresistance in hematologic malignancies. Semin Hematol 34:9–19 Obermann EC, Csato M, Dirnhofer S, Tzankov A, 2009, BCL2 gene aberration as an IPI-independent marker for poor outcome in non-germinal-centre diffuse large B cell lymphoma. J Clin Pathol 62:903–907 Hermine O, Haioun C, Lepage E, d’Agay MF, Briere J, Lavignac C, Fillet G, Salles G, Marolleau JP, Diebold J, Reyas F, Gaulard P, 1996, Prognostic significance of bcl-2 protein expression in aggressive non-Hodgkin’s lymphoma. Blood 87:265–272 Gascoyne R, Adomat S, Krajewski S, Krajewska M, Horsman DE, Tolcher AW, O’Reilly SE, Hoskins P, Coldman AJ, Reed JC, Connors JM, 1997, Prognostic significance of bcl-2 protein expression and Bcl-2 gene rearrangement in diffuse aggressive non- Hodgkin’s lymphoma. Blood 90:244–251 Barrans SL, Carter I, Owen RG, Davies FE, Patmore RD, Haynes AP, Morgan GJ, Jack AS, 2002, Germinal center phenotype and bcl-2 expression combined with the International Prognostic Index improves patient risk stratification in diffuse large B-cell lymphoma. Blood 99:1136–1143 Ohshima K, Kawasaki C, Muta H, Muta K, Deyev V, Haraoka S, Suzumiya J, Podack ER, Kikuchi M, 2001, CD10 and Bcl10 expression in diffuse large B-cell lymphoma: CD10 is a marker of improved prognosis. Histopathology 39(2):156–162 Liu YJ, Johnson GD, Gordon J, Maclennan IC, 1992, Germinal centres in T-cell-dependent antibody responses. Immunol Today 13:17–21 Cutrona G, Dono M, Pastorino S, Ulivi M, Burgio VL, Zupo S, Roncella S, Ferrarini M, 1997, The propensity to apoptosis of centrocytes and centroblasts correlates with elevated levels of intracellular myc protein. Eur J Immunol 27:234–238 Cutrona G, Leanza N, Ulivi M, Melioli G, Burgio VL, Mazzarello G, Gabutti G, 1999, Expression of CD10 by human Tcells that undergo apoptosis both in vitro and in vivo. Blood 94(9):3067–3076 Cutrona G, Tasso P, Dono M, Roncella S, Ulivi M, Carpaneto EM, Fontana V, Comis M, Morabito F, Spinelli M, Frascella E, Boffa LC, Basso G, Pistoia, Ferrarini M, 2002, CD10 is a marker for cycling cells with propensity to apoptosis in childhood ALL. Br J Cancer 86(11):1776–1785 Lossos IS, Jones CD, Warnke R, Natkunam Y, Kaizer H, Zehnder JL, Tibshirani R, Levy R, 2001, Expression of a single gene, BCL- 6, strongly predicts survival in patients with diffuse large B-cell lymphoma. Blood 98:945–951 Winter JN, Weller EA, Horning SJ, Krajewska M, Variakojis D, Habermann TM, Fisher RI, Kurtin PJ, Macon WR, Chhanabhai M, Felgar RE, Hsi ED, Medeiros LJ, Weick JK, Reed JC, Gascoyne RD, 2006, Prognostic significance of Bcl-6 protein expression in DLBCL treated with CHOP or R-CHOP: a prospective correlative study. Blood 107:4207–4213 Levine AM, 2006, AIDS-related lymphoma. Semin Oncol Nurs 22:80–89 Castillo J, Pantanowithz L, Dezube BJ, 2008, HIV-associated plasmablastic lymphoma: a literature review of 112 cases. Am J Hematol 83:804–809 Flaitz CM, Nichols, Walling DM, Hicks MJ, 2002, Case report. Plasmablastic lymphoma: an HIV-associated entity with primary oral manifestations. Oral Oncol 38:96–102 Chabay P, De Matteo E, Lorenzetti M, Gutierrez M, Narbaitz M, Aversa L, Preciado MV, 2009, Case report: vulvar plasmablastic lymphoma in a HIV-positive child: a novel extraoral localisation. J Clin Pathol 62:644–646 Radhakrishnan R, Suhas S, Kumar RV, Krishnanand G, Srinivasan R, Rao NN, 2005, Plasmablastic lymphoma of the oral cavity in an HIV-positive child. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 100:725–731 Gogia A, Bakhshi S, 2010, Letter to the editor. Plasmablastic lymphoma of oral cavity in an HIV negative child. Pediatr Blood Cancer 55:390–391 Pather S, MacKinnon D, Padayachee RS, 2012, Plasmablastic lymphoma in paediatric patients: Clinicopathologic study of three cases. Ann Diagn Pathol. Accepted for publication Aug 2012 Scheper MA, Nikitakis NG, Fernandes R, Gocke CD, Ord RA, Sauk JJ, 2005, Oral plasmablastic lymphoma in an HIV-negative patient: a case report and review of the literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 100:198–206 Lin F, Zhang K, Quiery AT, Prichard J, Schuerch C, 2004, Plasmablastic Lymphoma of the cervical lymph nodes in a human immunodeficiency virus-negative patient. A case report and review of the literature. Arch Pathol Lab Med 128:581– 584 Guan B, Zhang X, Hu W, Rao Q, Wang Y, Zhu Y, Wang H, Ma H, Zhou H, Zhou X, 2011, Plasmablastic lymphoma of the oral cavity in an HIV-negative patient. Ann Diagn Pathol 15(6):436–440 Chetty R, Hlatswayo N, Muc R, Sabaratnam R, Gatter K, 2003, Plasmablastic lymphoma in HIV+patients: an expanding spectrum. Histopathology 42:605–609 Dong HY, Scadden DT, de Leval L, Tang Z, Isaacson PG, Harris NL, 2005, Plasmablastic lymphoma in HIV-positive patients: an aggressive Epstein-Barr virus–associated extramedullary plasmacytic neoplasm. Am J Surg Pathol 29:1633–1641 Rochford R, Hobbs MV, Garnier JL, Cooper NR, Cannon MJ, 1993, Plasmacytoid differentiation of Epstein-Barr virustransformed B cells in vivo is associated with reduced expression of viral latent genes. Proc Natl Acad Sci U S A 90:352–356 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 695 EP 705 DI 10.1007/s12253-013-9632-2 PG 11 ER PT J AU Skapa, P Robova, H Rob, L Zamecnik, J AF Skapa, Petr Robova, Helena Rob, Lukas Zamecnik, Josef TI p16INK4a Immunoprofiles of Squamous Lesions of the Uterine Cervix–Implications for the Reclassification of Atypical Immature Squamous Metaplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Uterine cervix; p16; Cytokeratin 17; Cervical intraepithelial neoplasia; Atypical immature squamous metaplasia; Flat condyloma ID Uterine cervix; p16; Cytokeratin 17; Cervical intraepithelial neoplasia; Atypical immature squamous metaplasia; Flat condyloma AB p16INK4a immunoprofiles of non-precancerous and dysplastic squamous cervical lesions were defined and applied to the reclassification of atypical immature squamous metaplasia (AIM). The immunoexpression of cytokeratin 17 (CK 17) in AIM was also evaluated. Totally, 295 cervical cone biopsies representing squamous metaplasia, reactive changes, koilocytosis, flat condyloma, CIN I, CIN II, CIN III and AIM were subjected to p16INK4a immunohistochemistry. AIM cases were analyzed using CK 17 antibody. Typical p16INK4a immunoprofiles for the metaplastic, LSIL/HPV and HSIL phenotypes were recorded and used for the categorization of AIM into particular phenotype groups. Results were correlated with CK 17 immunoexpression. All CIN II and CIN III lesions, all but one case of CIN I and all flat condylomas overexpressed p16INK4a. Other non-precancerous lesions, including koilocytosis, were predominantly negative. Contrary to the sporadic and focal immunostaining, diffuse positivity was associated with the dysplastic features of the lesion. CIN II and CIN III were characterized by a diffuse, strong/weak, full-thickness staining, whereas CIN I showed a heterogeneous diffuse/focal, weak/strong, lower half positivity. One third of AIM lesions may be reclassified as HSIL, one third as LSIL/HPV and one third shows metaplastic phenotype. All AIM cases with metaplastic and LSIL/HPV phenotypes expressed CK 17 diffusely, whereas focal positivity slightly prevailed in AIM with HSIL phenotype. We conclude that p16INK4a immunohistochemistry is a supporting method for the differential diagnosis of cervical lesions, which may be especially useful for the reclassification of AIM. The efficacy of CK 17 immunohistochemistry seems to be controversial for these purposes. C1 [Skapa, Petr] Charles University and University Hospital Motol, 2nd Medical School, Department of Pathology and molecular medicine, V Uvalu 84, 150 06 Prague, Czech Republic. [Robova, Helena] University Hospital Motol, Charles University, 2nd Medical School, Department of Obstetrics and Gynecology, V Uvalu 84Prague, Czech Republic. [Rob, Lukas] University Hospital Motol, Charles University, 2nd Medical School, Department of Obstetrics and Gynecology, V Uvalu 84Prague, Czech Republic. [Zamecnik, Josef] Charles University and University Hospital Motol, 2nd Medical School, Department of Pathology and molecular medicine, V Uvalu 84, 150 06 Prague, Czech Republic. RP Skapa, P (reprint author), Charles University and University Hospital Motol, 2nd Medical School, Department of Pathology and molecular medicine, 150 06 Prague, Czech Republic. EM petr.skapa@lfmotol.cuni.cz CR McCluggage WG, Walsh MY, Thornton CM, Hamilton PW, Date A, Caughley LM, Bharucha H, 1998, Inter- and intra-observer variation in the histopathological reporting of cervical squamous intraepithelial lesions using a modified Bethesda grading system. Br J Obstet Gynaecol 105(2):206–210 Stoler MH, Schiffman M, 2001, Interobserver reproducibility of cervical cytologic and histologic interpretations: realistic estimates from the ASCUS-LSIL Triage Study. JAMA 285(11):1500–1505 Carreon JD, ShermanME, GuillenD, Solomon D, Herrero R, Jeronimo J, Wacholder S, Rodriguez AC, Morales J, Hutchinson M, Burk RD, Schiffman M, 2007, CIN2 is a much less reproducible and less valid diagnosis than CIN3: results from a histological review of populationbased cervical samples. Int J Gynecol Pathol 26(4):441–446 Crum CP, Egawa K, Fu YS, Lancaster WD, Barron B, Levine RU, Fenoglio CM, Richart RM, 1983, Atypical immature metaplasia, AIM). A subset of human papilloma virus infection of the cervix. Cancer 51(12):2214–2219 Geng L, Connolly DC, Isacson C, Ronnett BM, Cho KR, 1999, Atypical immature metaplasia, AIM, of the cervix: is it related to high-grade squamous intraepithelial lesion, HSIL)? Hum Pathol 30(3):345–351 Iaconis L, Hyjek E, Ellenson LH, Pirog EC, 2007, p16 and Ki-67 immunostaining in atypical immature squamous metaplasia of the uterine cervix: correlation with human papillomavirus detection. Arch Pathol Lab Med 131(9):1343–1349 Duggan MA, Akbari M, Magliocco AM, 2006, Atypical immature cervical metaplasia: immunoprofiling and longitudinal outcome. Hum Pathol 37(11):1473–1481 O’Neill CJ, McCluggage WG, 2006, p16 expression in the female genital tract and its value in diagnosis. Adv Anat Pathol 13(1):8–15 Branca M, Ciotti M, Santini D, Di Bonito L, Giorgi C, Benedetto A, Paba P, Favalli C, Costa S, Agarossi A, Alderisio M, Syrjanen K, 2004, p16(INK4A, expression is related to grade of CIN and highrisk human papillomavirus but does not predict virus clearance after conization or disease outcome. Int J Gynecol Pathol 23(4):354–365 Martens JE, Arends J, Van der Linden PJ, De Boer BA, Helmerhorst TJ, 2004, Cytokeratin 17 and p63 are markers of the HPV target cell, the cervical stem cell. Anticancer Res 24(2B):771–775 Smedts F, Ramaekers F, Troyanovsky S, Pruszczynski M, Robben H, Lane B, Leigh I, Plantema F, Vooijs P, 1992, Basal-cell keratins in cervical reserve cells and a comparison to their expression in cervical intraepithelial neoplasia. Am J Pathol 140(3):601–612 Ikeda K, Tate G, Suzuki T, Mitsuya T, 2008, Coordinate expression of cytokeratin 8 and cytokeratin 17 immunohistochemical staining in cervical intraepithelial neoplasia and cervical squamous cell carcinoma: an immunohistochemical analysis and review of the literature. Gynecol Oncol 108(3):598–602 Regauer S, Reich O, 2007, CK17 and p16 expression patterns distinguish, atypical, immature squamous metaplasia from highgrade cervical intraepithelial neoplasia, CIN III). Histopathology 50(5):629–635 Klaes R, Friedrich T, Spitkovsky D, Ridder R, Rudy W, Petry U, Dallenbach-Hellweg G, Schmidt D, von Knebel Doeberitz M, 2001, Overexpression of p16(INK4A, as a specific marker for dysplastic and neoplastic epithelial cells of the cervix uteri. Int J Cancer 92(2):276–284 Keating JT, Cviko A, Riethdorf S, Riethdorf L, Quade BJ, Sun D, Duensing S, Sheets EE, Munger K, Crum CP, 2001, Ki-67, cyclin E, and p16INK4 are complimentary surrogate biomarkers for human papilloma virus-related cervical neoplasia. Am J Surg Pathol 25(7):884–891 Sano T, Oyama T, Kashiwabara K, Fukuda T, Nakajima T, 1998, Expression status of p16 protein is associated with human papillomavirus oncogenic potential in cervical and genital lesions. Am J Pathol 153(6):1741–1748 OmoriM, Hashi A, Nakazawa K, Yuminamochi T, Yamane T, Hirata S, Katoh R, Hoshi K, 2007, Estimation of prognoses for cervical intraepithelial neoplasia 2 by p16INK4a immunoexpression and high-risk HPV in situ hybridization signal types. Am J Clin Pathol 128(2):208–217 Focchi GR, Silva ID, Nogueira-de-Souza NC, Dobo C, Oshima CT, Stavale JN, 2007, Immunohistochemical expression of p16(INK4A, in normal uterine cervix, nonneoplastic epithelial lesions, and low-grade squamous intraepithelial lesions. J Low Genit Tract Dis 11(2):98–104 Wright TC, Massad LS, Dunton CJ, Spitzer M, Wilkinson EJ, Solomon D, 2007, 2006 consensus guidelines for the management of women with cervical intraepithelial neoplasia or adenocarcinoma in situ. Am J Obstet Gynecol 197(4):340–345 Negri G, Vittadello F, Romano F, Kasal A, Rivasi F, Girlando S, Mian C, Egarter-Vigl E, 2004, p16INK4a expression and progression risk of low-grade intraepithelial neoplasia of the cervix uteri. Virchows Arch 445(6):616–620 Wang JL, Zheng BY, Li XD, Angstrom T, LindstromMS,Wallin KL, 2004, Predictive significance of the alterations of p16INK4A, p14ARF, p53, and proliferating cell nuclear antigen expression in the progression of cervical cancer. Clin Cancer Res 10(7):2407–2414 Hariri J, Oster A, 2007, The negative predictive value of p16INK4a to assess the outcome of cervical intraepithelial neoplasia 1 in the uterine cervix. Int J Gynecol Pathol 26(3):223–228 Castle PE, SchiffmanM,Wheeler CM, Solomon D, 2009, Evidence for frequent regression of cervical intraepithelial neoplasia-grade 2. Obstet Gynecol 113(1):18–25 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 707 EP 714 DI 10.1007/s12253-013-9633-1 PG 8 ER PT J AU Otrock, KZ Saab, J Aftimos, G Nasr, F Farhat, SF Khairallah, S Abadjian, G Ghosn, M Sidani, H Ibrahim, A Tawil, A Ghorra, C Meguerian, Z Mokaddem, W Dayeh, W Salem, Z Chahine, G Bitar, N Mugharbel, A Makdessi, J Khater, Ch El Hajj, M Abi Gerges, D Sfeir, Ch Kattan, J Ibrahim, K Saade, M Sadek, H Mahfouz, AR Kharfan-Dabaja, AM Zaatari, G Bazarbachi, A AF Otrock, K Zaher Saab, Jad Aftimos, Georges Nasr, Fady Farhat, S Fadi Khairallah, Saad Abadjian, Gerard Ghosn, Marwan Sidani, Hassan Ibrahim, Ahmad Tawil, Ayman Ghorra, Claude Meguerian, Zarouhie Mokaddem, Walid Dayeh, Walid Salem, Ziad Chahine, Georges Bitar, Nizar Mugharbel, Anas Makdessi, Joseph Khater, Christina El Hajj, Mirna Abi Gerges, Dany Sfeir, Charles Kattan, Joseph Ibrahim, Khaled Saade, Michel Sadek, Hussein Mahfouz, A Rami Kharfan-Dabaja, A Mohamed Zaatari, Ghazi Bazarbachi, Ali TI A Collaborative Nationwide Lymphoma Study in Lebanon: Incidence of Various Subtypes and Analysis of Associations with Viruses SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lymphoma; Incidence; Subtypes; Viruses; EBV; Lebanon ID Lymphoma; Incidence; Subtypes; Viruses; EBV; Lebanon AB Incidence of various Hodgkin (HL) and non- Hodgkin lymphoma (NHL) subtypes and association with viruses in Lebanon are not known. We undertook a nationwide study of 272 patients diagnosed with lymphoma in 2007. HL comprised 32.7%(n=89) of cases while NHL represented 67.3 % (n=183). Consistent with the literature, nodular sclerosis was the most predominant HL subtype (n=57/89). Among NHL, B-cell NHL represented 88 % (n=161/183), T-cell NHL 9 % (n=17/183), whereas in 2.7 % it was not classifiable. The B-cell NHL comprised predominantly diffuse large B-cell lymphoma (46 %) and follicular lymphoma (23 %). 81 cases were reviewed by a panel of pathologists with 87.6 % concordance rate. Serology was negative for hepatitis C in 122 tested cases. HIV was positive in 2 cases. Two adult T-cell leukemia/lymphoma were HTLV-I positive. EBV IgG were positive in 88.5 % of cases. 38 EBV seropositive cases [27 NHL (24 B-cell, 3 T-cell) and 11 HL] were studied for EBV genome expression using EBV-encoded RNA (EBER)-in situ hybridization. EBER expression was positive in 8 (21 %) cases (6 HL, 2 T-cell NHL). The distribution of lymphoma subtypes in Lebanon appears similar to that of Western countries. The high rate of EBV positivity in HL and T-cell lymphoma by EBER deserves further investigation. C1 [Otrock, K Zaher] American University of Beirut Medical Center, Department of Pathology and Laboratory MedicineBeirut, Lebanon. [Saab, Jad] American University of Beirut Medical Center, Department of Pathology and Laboratory MedicineBeirut, Lebanon. [Aftimos, Georges] Institut National de PathologieBeirut, Lebanon. [Nasr, Fady] Hotel-Dieu de France University HospitalBeirut, Lebanon. [Farhat, S Fadi] Hammoud Hospital University Medical CenterSaida, Lebanon. [Khairallah, Saad] Institut National de PathologieBeirut, Lebanon. [Abadjian, Gerard] Hotel-Dieu de France University HospitalBeirut, Lebanon. [Ghosn, Marwan] Hotel-Dieu de France University HospitalBeirut, Lebanon. [Sidani, Hassan] Makassed General HospitalBeirut, Lebanon. [Ibrahim, Ahmad] Makassed General HospitalBeirut, Lebanon. [Tawil, Ayman] American University of Beirut Medical Center, Department of Pathology and Laboratory MedicineBeirut, Lebanon. [Ghorra, Claude] Hotel-Dieu de France University HospitalBeirut, Lebanon. [Meguerian, Zarouhie] Saint Georges HospitalBeirut, Lebanon. [Mokaddem, Walid] Islamic HospitalTripoli, Lebanon. [Dayeh, Walid] Hammoud Hospital University Medical CenterSaida, Lebanon. [Salem, Ziad] American University of Beirut Medical Center, Department of Internal Medicine, 6044 Beirut, Lebanon. [Chahine, Georges] Hotel-Dieu de France University HospitalBeirut, Lebanon. [Bitar, Nizar] Sahel General HospitalBeirut, Lebanon. [Mugharbel, Anas] Makassed General HospitalBeirut, Lebanon. [Makdessi, Joseph] Saint Georges HospitalBeirut, Lebanon. [Khater, Christina] Trad HospitalBeirut, Lebanon. [El Hajj, Mirna] Saint Georges HospitalBeirut, Lebanon. [Abi Gerges, Dany] Middle East Institute of HealthBeirut, Lebanon. [Sfeir, Charles] Saint Georges HospitalAjaltoun, Lebanon. [Kattan, Joseph] Hotel-Dieu de France University HospitalBeirut, Lebanon. [Ibrahim, Khaled] Hammoud Hospital University Medical CenterSaida, Lebanon. [Saade, Michel] Rizk HospitalBeirut, Lebanon. [Sadek, Hussein] Bekaa General HospitalBekaa, Lebanon. [Mahfouz, A Rami] American University of Beirut Medical Center, Department of Pathology and Laboratory MedicineBeirut, Lebanon. [Kharfan-Dabaja, A Mohamed] American University of Beirut Medical Center, Department of Internal Medicine, 6044 Beirut, Lebanon. [Zaatari, Ghazi] American University of Beirut Medical Center, Department of Pathology and Laboratory MedicineBeirut, Lebanon. [Bazarbachi, Ali] American University of Beirut Medical Center, Department of Internal Medicine, 6044 Beirut, Lebanon. RP Bazarbachi, A (reprint author), American University of Beirut Medical Center, Department of Internal Medicine, 6044 Beirut, Lebanon. CR Morton LM, Wang SS, Devesa SS, Hartge P, Weisenburger DD, Linet MS, 2006, Lymphoma incidence patterns by WHO subtype in the United States, 1992–2001. Blood 107(1):265–276., DOI 10.1182/blood-2005-06-2508 Roman E, Smith AG, 2011, Epidemiology of lymphomas. Histopathology 58(1):4–14., DOI 10.1111/j.1365-2559.2010.03696.x Smith A, Roman E, Howell D, Jones R, Patmore R, Jack A, 2010, The Haematological Malignancy Research Network, HMRN): a new information strategy for population based epidemiology and health service research. Br J Haematol 148(5):739–753., DOI 10.1111/j.1365-2141.2009.08010.x Cancer Facts and Figures 2010, Accessed February 16, 2011) Ekstrom-Smedby K, 2006, Epidemiology and etiology of non- Hodgkin lymphoma—a review. Acta Oncol 45(3):258–271., DOI 10.1080/02841860500531682 Zuckerman E, Zuckerman T, Levine AM, Douer D, Gutekunst K, Mizokami M, Qian DG, Velankar M, Nathwani BN, Fong TL, 1997, Hepatitis C virus infection in patients with B-cell non- Hodgkin lymphoma. Ann Intern Med 127(6):423–428 Muller AM, Ihorst G, Mertelsmann R, Engelhardt M, 2005, Epidemiology of non-Hodgkin’s lymphoma, NHL): trends, geographic distribution, and etiology. Ann Hematol 84(1):1–12., DOI 10.1007/s00277-004-0939-7 Suarez F, Lortholary O, Hermine O, Lecuit M, 2006, Infectionassociated lymphomas derived from marginal zone B cells: a model of antigen-driven lymphoproliferation. Blood 107(8):3034–3044., DOI 10.1182/blood-2005-09-3679 Yamaguchi K, Takatsuki K, 1993, Adult T cell leukaemia–lymphoma. Baillieres Clin Haematol 6(4):899–915 Baris D, Zahm SH, 2000, Epidemiology of lymphomas. Curr Opin Oncol 12(5):383–394 Cartwright R, Brincker H, Carli PM, Clayden D, Coebergh JW, Jack A, McNally R, Morgan G, de Sanjose S, Tumino R, Vornanen M, 1999, The rise in incidence of lymphomas in Europe 1985– 1992. Eur J Cancer 35(4):627–633 Bosetti C, Levi F, Ferlay J, Lucchini F, Negri E, La Vecchia C, 2008, Incidence and mortality from non-Hodgkin lymphoma in Europe: the end of an epidemic? Int J Cancer 123(8):1917–1923., DOI 10.1002/ijc.23722 Greiner TC, Medeiros LJ, Jaffe ES, 1995, Non-Hodgkin’s lymphoma. Cancer 75(1 Suppl):370–380 Eltom MA, Jemal A, Mbulaiteye SM, Devesa SS, Biggar RJ, 2002, Trends in Kaposi’s sarcoma and non-Hodgkin’s lymphoma incidence in the United States from 1973 through 1998. J Natl Cancer Inst 94(16):1204–1210 Clarke CA, Glaser SL, 2002, Changing incidence of non-Hodgkin lymphomas in the United States. Cancer 94(7):2015–2023., DOI 10.1002/cncr.10403 Carreon JD, Morton LM, Devesa SS, Clarke CA, Gomez SL, Glaser SL, Sakoda LC, Linet MS, Wang SS, 2008, Incidence of lymphoid neoplasms by subtype among six Asian ethnic groups in the United States, 1996–2004. Cancer Causes Control 19(10):1171–1181., DOI 10.1007/s10552-008-9184-z Nakatsuka S, Aozasa K, 2006, Epidemiology and pathologic features of Hodgkin lymphoma. Int J Hematol 83(5):391–397., DOI 10.1532/IJH97.05184 Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55(2):74–108 Mueller N, 1994, Another view of the epidemiology of non- Hodgkin’s lymphoma. Oncology, Williston Park, 8(9):83 Glaser SL, Swartz WG, 1990, Time trends in Hodgkin’s disease incidence. The role of diagnostic accuracy. Cancer 66(10):2196– 2204 Jaffe ES, Harris N, Stein H, Vardiman JW, 2001, Pathology and genetics of tumours of haematopoietic and lymphoid tissues. IARC Press, Lyon Shamseddine A, Sibai AM, Gehchan N, Rahal B, El-Saghir N, Ghosn M, Aftimos G, Chamsuddine N, Seoud M, 2004, Cancer incidence in postwar Lebanon: findings from the first national population-based registry, 1998. Ann Epidemiol 14(9):663–668., DOI 10.1016/j.annepidem.2003.12.002 Castella A, Joshi S, Raaschou T, Mason N, 2001, Pattern of malignant lymphoma in the United Arab Emirates—a histopathologic and immunologic study in 208 native patients. Acta Oncol 40(5):660–664 Bamanikar S, Thunold S, Devi KR, Bamanikar A, 1995, The pattern of malignant lymphoma in Oman. J Trop Med Hyg 98(5):351–354 Akhtar SS, Haque IU, Wafa SM, El-Saka H, Saroor AM, Nadrah HM, 2009, Malignant lymphoma in Al-Qassim, Saudi Arabia, reclassified according to the WHO classification. Saudi Med J 30(5):677–681 Haddadin WJ, 2005, Malignant lymphoma in Jordan: a retrospective analysis of 347 cases according to the World Health Organization classification. Ann Saudi Med 25(5):398–403 Shome DK, George SM, Al-Hilli F, Satir AA, 2004, Spectrum of malignant lymphomas in Bahrain. Leitmotif of a regional pattern. Saudi Med J 25(2):164–167 Temmim L, Baker H, Al-Jarallah M, Manguno H, Madda JP, Sinowatz F, 2004, Clinical characteristics and pathological classification of non-Hodgkin’s lymphoma in Kuwait. Results of a collaborative study with the International Lymphoma Study Group, ILSG). Leuk Lymphoma 45(9):1865–1871., DOI 10.1080/ 10428190410001697386 Kadin ME, Berard CW, Nanba K, Wakasa H, 1983, Lymphoproliferative diseases in Japan and Western countries: proceedings of the United States–Japan Seminar, September 6 and 7, 1982, in Seattle, Washington. Hum Pathol 14(9):745–772 Ho FC, Todd D, Loke SL, Ng RP, Khoo RK, 1984, Clinicopathological features of malignant lymphomas in 294 Hong Kong Chinese patients, retrospective study covering an eightyear period. Int J Cancer 34(2):143–148 Ko YH, Kim CW, Park CS, Jang HK, Lee SS, Kim SH, Ree HJ, Lee JD, Kim SW, Huh JR, 1998, REAL classification of malignant lymphomas in the Republic of Korea: incidence of recently recognized entities and changes in clinicopathologic features. Hematolymphoreticular Study Group of the Korean Society of Pathologists. Revised European–American lymphoma. Cancer 83(4):806–812., DOI 10.1002/(SICI)1097-0142(19980815, 83:4<806::AID-CNCR26>3.0.CO;2-V Shih LY, Liang DC, 1991, Non-Hodgkin’s lymphomas in Asia. Hematol Oncol Clin N Am 5(5):983–1001 Ameen R, Sajnani KP, Albassami A, Refaat S, 2010, Frequencies of non-Hodgkin’s lymphoma subtypes in Kuwait: comparisons between different ethnic groups. Ann Hematol 89(2):179–184., DOI 10.1007/s00277-009-0801-z Al-Kuraya K, Narayanappa R, Al-Dayel F, El-Solh H, Ezzat A, Ismail H, Belgaumi A, Bavi P, Atizado V, Sauter G, Simon R, 2006, Epstein-Barr virus infection is not the sole cause of high prevalence for Hodgkin’s lymphoma in Saudi Arabia. Leuk Lymphoma 47(4):707–713., DOI 10.1080/10428190500286879 Allemani C, Sant M, De Angelis R, Marcos-Gragera R, Coebergh JW, 2006, Hodgkin disease survival in Europe and the U.S.: prognostic significance of morphologic groups. Cancer 107(2):352–360., DOI 10.1002/cncr.21995 Makar RR, Saji T, Junaid TA, 2003, Epstein–Barr virus expression in Hodgkin’s lymphoma in Kuwait. Pathol Oncol Res 9(3):159–165 Al-Shemmari SH, Al-Humood S, Ameen R, Kamlesh S, Nemec J, Varghese A, 2004, Hodgkin’s disease: Kuwait experience. Med Princ Pract 13(4):201–205., DOI 10.1159/000078316 Proctor IE, McNamara C, Rodriguez-Justo M, Isaacson PG, Ramsay A, 2011, Importance of expert central review in the diagnosis of lymphoid malignancies in a regional cancer network. J Clin Oncol 29(11):1431–1435., DOI 10.1200/JCO.2010.31.2223 Lester JF, Dojcinov SD, Attanoos RL, O’Brien CJ, Maughan TS, Toy ET, Poynton CH, 2003, The clinical impact of expert pathological review on lymphoma management: a regional experience. Br J Haematol 123(3):463–468 Ferri C, Caracciolo F, Zignego AL, La Civita L, Monti M, Longombardo G, Lombardini F, Greco F, Capochiani E, Mazzoni A et al, 1994, Hepatitis C virus infection in patients with non-Hodgkin’s lymphoma. Br J Haematol 88(2):392–394 Pozzato G, Mazzaro C, Crovatto M, Modolo ML, Ceselli S, Mazzi G, Sulfaro S, Franzin F, Tulissi P, Moretti M et al, 1994, Lowgrade malignant lymphoma, hepatitis C virus infection, and mixed cryoglobulinemia. Blood 84(9):3047–3053 Dal Maso L, Franceschi S, 2006, Hepatitis C virus and risk of lymphoma and other lymphoid neoplasms: a meta-analysis of epidemiologic studies. Cancer Epidemiol Biomarkers Prev 15(11):2078–2085., DOI 10.1158/1055-9965.EPI-06-0308 Hausfater P, Cacoub P, Sterkers Y, Thibault V, Amoura Z, NguyenL, Ghillani P, Leblond V, Piette JC, 2001, Hepatitis C virus infection and lymphoproliferative diseases: prospective study on 1,576 patients in France. AmJ Hematol 67(3):168–171., DOI 10.1002/ajh.1101 Udomsakdi-Auewarakul C, Auewarakul P, Sukpanichnant S, Muangsup W, 2000, Hepatitis C virus infection in patients with non-Hodgkin lymphoma in Thailand. Blood 95(11):3640–3641 Salem Z, Nuwaiyri-Salti N, Ramlawi F, Ramia S, 2003, Hepatitis C virus infection in Lebanese patients with B-cell non-Hodgkin’s lymphoma. Eur J Epidemiol 18(3):251–253 Irani-Hakime N, Musharrafieh U, Samaha H, Almawi WY, 2006, Prevalence of antibodies against hepatitis B virus and hepatitis C virus among blood donors in Lebanon, 1997–2003. Am J Infect Control 34(4):241–243., DOI 10.1016/j.ajic.2005.06.009 Uchiyama T, Yodoi J, Sagawa K, Takatsuki K, Uchino H, 1977, Adult T-cell leukemia: clinical and hematologic features of 16 cases. Blood 50(3):481–492 Bazarbachi A, Ghez D, Lepelletier Y, Nasr R, de The H, El-Sabban ME, Hermine O, 2004, New therapeutic approaches for adult Tcell leukaemia. Lancet Oncol 5(11):664–672., DOI 10.1016/S1470- 2045(04)01608-0 Bitar N, Hajj HE, Houmani Z, Sabbah A, Otrock ZK, Mahfouz R, Zaatari G, Bazarbachi A, 2009, Adult T-cell leukemia/lymphoma in theMiddle East: first report of two cases from Lebanon. Transfusion 49(9):1859–1864., DOI 10.1111/j.1537-2995.2009.02223.x Spano JP, Atlan D, Breau JL, Farge D, 2002, AIDS and non- AIDS-related malignancies: a new vexing challenge in HIVpositive patients. Part I: Kaposi’s sarcoma, non-Hodgkin’s lymphoma, and Hodgkin’s lymphoma. Eur J Intern Med 13(3):170–179 Dal Maso L, Serraino D, Franceschi S, 2001, Epidemiology of AIDS-related tumours in developed and developing countries. Eur J Cancer 37(10):1188–1201 Rabkin CS, 1994, Epidemiology of AIDS-related malignancies. Curr Opin Oncol 6(5):492–496 Carbone A, Gloghini A, Dotti G, 2008, EBV-associated lymphoproliferative disorders: classification and treatment. Oncologist 13(5):577–585., DOI 10.1634/theoncologist.2008-0036 Jones JF, Shurin S, Abramowsky C, Tubbs RR, Sciotto CG, Wahl R, Sands J, Gottman D, Katz BZ, Sklar J, 1988, T-cell lymphomas containing Epstein–Barr viral DNA in patients with chronic Epstein–Barr virus infections. N Engl J Med 318(12):733–741., DOI 10.1056/NEJM198803243181203 Rezk SA, Weiss LM, 2007, Epstein–Barr virus-associated lymphoproliferative disorders. Hum Pathol 38(9):1293–1304., DOI 10.1016/j.humpath.2007.05.020 Shibata D, Hansmann ML, Weiss LM, Nathwani BN, 1991, Epstein–Barr virus infections and Hodgkin’s disease: a study of fixed tissues using the polymerase chain reaction. Hum Pathol 22(12):1262–1267 Evans AS, Gutensohn NM, 1984, A population-based case–control study of EBV and other viral antibodies among persons with Hodgkin’s disease and their siblings. Int J Cancer 34(2):149–157 Pallesen G, Hamilton-Dutoit SJ, Rowe M, Young LS, 1991, Expression of Epstein–Barr virus latent gene products in tumour cells of Hodgkin’s disease. Lancet 337(8737):320–322 Anagnostopoulos I, Herbst H, Niedobitek G, Stein H, 1989, Demonstration of monoclonal EBV genomes in Hodgkin’s disease and Ki-1-positive anaplastic large cell lymphoma by combined Southern blot and in situ hybridization. Blood 74(2):810–816 Staratschek-Jox A, Kotkowski S, Belge G, Rudiger T, Bullerdiek J, Diehl V, Wolf J, 2000, Detection of Epstein–Barr virus in Hodgkin-Reed-Sternberg cells: no evidence for the persistence of integrated viral fragments inLatent membrane protein-1, LMP-1)- negative classical Hodgkin’s disease. Am J Pathol 156(1):209– 216., DOI 10.1016/S0002-9440(10)64721-9 Khan G, Coates PJ, Kangro HO, Slavin G, 1992, Epstein Barr virus, EBV, encoded small RNAs: targets for detection by in situ hybridisation with oligonucleotide probes. J Clin Pathol 45(7):616–620 Khan G, 2009, Screening for Epstein–Barr virus in Hodgkin’s lymphoma. Methods Mol Biol 511:311–322., DOI 10.1007/978-1- 59745-447-6_13 Enblad G, Sandvej K, Sundstrom C, Pallesen G, Glimelius B, 1999, Epstein–Barr virus distribution in Hodgkin’s disease in an unselected Swedish population. Acta Oncol 38(4):425–429 Chang KL, Albujar PF, Chen YY, Johnson RM, Weiss LM, 1993, High prevalence of Epstein–Barr virus in the Reed–Sternberg cells of Hodgkin’s disease occurring in Peru. Blood 81(2):496–501 Katebi M, Sharifi N, Tarhini M, Otrock ZK, Bazarbachi A, Kchour G, 2008, Frequency of Epstein–Barr virus expression in various histological subtypes of Hodgkin’s lymphoma. Histopathology 52(6):775–777., DOI 10.1111/j.1365-2559.2008.03019.x Mourad W, Alsohaibani MO, Saddik M, Bazerbashi S, 1998, Epstein–Barr virus expression in Hodgkin’s disease: correlation with histologic subtypes and T and B lymphocyte distribution. Ann Saudi Med 18(4):296–300 Almasri NM, Khalidi HS, 2004, Epstein–Barr virus expression in Hodgkin’s disease in Jordan. Saudi Med J 25(6):770–775 Levine PH, Pallesen G, Ebbesen P, Harris N, Evans AS, Mueller N, 1994, Evaluation of Epstein–Barr virus antibody patterns and detection of viral markers in the biopsies of patients with Hodgkin’s disease. Int J Cancer 59(1):48–50 Enblad G, Sandvej K, Lennette E, Sundstrom C, Klein G, Glimelius B, Pallesen G, 1997, Lack of correlation between EBV serology and presence of EBV in the Hodgkin and Reed– Sternberg cells of patients with Hodgkin’s disease. Int J Cancer 72(3):394–397., DOI 10.1002/(SICI)1097-0215(19970729, 72:3<394::AID-IJC3>3.0.CO;2-K Axdorph U, Porwit-MacDonald A, Sjoberg J, Grimfors G, Ekman M, Wang W, Biberfeld P, Bjorkholm M, 1999, Epstein–Barr virus expression in Hodgkin’s disease in relation to patient characteristics, serum factors and blood lymphocyte function. Br J Cancer 81(7):1182–1187., DOI 10.1038/sj.bjc.6690827 Abdel-Fattah MM, Yassine OG, 2007, Non-Hodgkin’s lymphomas in Alexandria, Egypt; incidence rates and trend study, 1995– 2004). Eur J Cancer Prev 16(5):479–485., DOI 10.1097/ 01.cej.0000243858.91642.c9 Wu XC, Andrews P, Chen VW, Groves FD, 2009, Incidence of extranodal non-Hodgkin lymphomas among whites, blacks, and Asians/Pacific Islanders in the United States: anatomic site and histology differences. Cancer Epidemiol 33(5):337–346., DOI 10.1016/j.canep.2009.09.006 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 715 EP 722 DI 10.1007/s12253-013-9634-0 PG 8 ER PT J AU Madaras, L Baranyak, Zs Kulka, J Szasz, MA Kovacs, A Lan, HP Szekely, B Dank, M Nagy, T Kiss, O Harsanyi, L Barbai, T Kenessey, I Tokes, AM AF Madaras, Lilla Baranyak, Zsuzsanna Kulka, Janina Szasz, Marcell Attila Kovacs, Attila Lan, Huong Phan Szekely, Borbala Dank, Magdolna Nagy, Tibor Kiss, Orsolya Harsanyi, Laszlo Barbai, Tamas Kenessey, Istvan Tokes, Anna Maria TI Retrospective Analysis of Clinicopathological Characteristics and Family History Data of Early-Onset Breast Cancer: A Single-Institutional Study of Hungarian Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Early-onset; Family history ID Breast cancer; Early-onset; Family history AB Patients at young age (≤35 years) diagnosed with breast cancer (BC) are considered to have poor prognosis. The aim of the present study was to retrospectively analyse clinicopathological characteristics and prognosis in a group of young BC patients. We included women diagnosed with invasive breast carcinoma younger than/or at the age of 35 years. Between 1999 and 2009, 107 women with earlyonset BC were selected from the database of the 2nd Department of Pathology at Semmelweis University. For clinicopathological comparison, 55 women (36–45 years), 214 women (46–65 years), 110 women (66–75 years) and 58 women (76≤years) were also included in the analysis. Family history, clinicopathological and follow-up data were analysed. The tissue specimens were reviewed for histological type, nuclear grade, and estrogen receptor (ER), progesterone receptor (PgR), Ki67 and HER2 status (IHC4). The mean age in the study group was 31.6 years at the time of diagnosis. Histology showed a high incidence of grade III tumours in this group of patients (67.9 %), while only four cases (3.8 %) were considered grade I. According to the immunohistochemical results, 35.3 % of the study cases were considered as Luminal B (LumB: either being higly proliferative or co-expressing HER2) and 33.3 % as triple negative breast carcinomas (TNBC). The detailed questionnaire related to family history was completed and received in 49/107 cases (45.8 %). Analysis of these data revealed an affected family history of breast or ovarian carcinoma in first and second degree relatives in 51.0 %. A high proportion (52.0 %) of TNBC was observed among young women with a family history of the disease. Survival analysis of the 107 patients showed that 25 (23.3 %) women died until 31 December 2012. No significant difference in survival was detectable considering the regimen of systemic treatment (p=0.188). Regarding clinicopathological parameters, the immunophenotypes, grade, pT and pN values differred substantially between the age groups (p=0.001, for all), and the shortest relapse-free survival was seen among the youngest BC patients. This analysis illustrates that breast cancer arising in young women is characterized by the presence of less favorable subtypes such as LumB and TNBC. The increased proportion of TNBC was especially remarquable in the group of patients presenting with family history of the disease. The fact that a high rate of death occured and no significant difference in OS were notable regarding the scheme of systemic therapies (neoadjuvant vs. adjuvant) highlight the necessity of the development of new treatment strategies. C1 [Madaras, Lilla] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Baranyak, Zsuzsanna] MH Honvedkorhaz, Sebeszeti OsztalyBudapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Szasz, Marcell Attila] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kovacs, Attila] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Lan, Huong Phan] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Szekely, Borbala] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Dank, Magdolna] Semmelweis University, Department of Radiology and OncotherapyBudapest, Hungary. [Nagy, Tibor] Szt. Istvan University, Biotechnics Department, Szent-Gyorgyi Albert u. 4., 2100 Godollo, Hungary. [Kiss, Orsolya] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Harsanyi, Laszlo] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Barbai, Tamas] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kenessey, Istvan] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Tokes, Anna Maria] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. RP Tokes, AM (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM ta@med.semmelweis-univ.hu CR Tusnady G, Gaudi I, Rejto L, Kasler M, Szentirmay Z, 2008, Survival chances of hungarian cancer patients in the national cancer registry. Magy Onkol 52:339–349 American Cancer SocietyWorkshop on Breast Cancer Detection in Younger Women, 1993, A current assessment. Proceedings. New York, February 1–2, 1993. Cancer 72(4 suppl):1428–1497 Paluch-Shimon S, Wolf I, Sadetzki S, Gluck I, Oberman B et al, 2011, Association between very young age and adverse characteristics of breast cancer at presentation amongst Israeli women. Am J Clin Oncol 34:219–222 Nixon AJ, Neuberg D, Hayes DF, Gelman R, Connolly JL et al, 1994, Relationship of patient age to pathologic features of the tumor and prognosis for patients with stage I or II breast cancer. J Clin Oncol 12:888–894 Fredholm H, Eaker S, Frisell J, Holmberg L, Fredriksson I et al, 2009, Breast cancer in young women: poor survival despite intensive treatment. PLoS One 4:e7695 Aebi S, Gelber S, Castiglione-Gertsch M, Gelber RD, Collins J et al, 2000, Is chemotherapy alone adequate for young women with oestrogen-receptor-positive breast cancer? Lancet 355:1869–1874 Kim K, Chie EK, Han W, Noh DY, Oh DY et al, 2011, Age <40Years is an independent prognostic factor predicting inferior overall survival in patients treated with breast conservative therapy. Breast J 17:75–78 Bonnier P, Romain S, Charpin C, Lejeune C, Tubiana N et al, 1995, Age as a prognostic factor in breast cancer: relationship to pathologic and biologic features. Int J Cancer 62:138–144 Colleoni M, Rotmensz N, Robertson C, Orlando L, Viale G et al, 2002, Very young women, <35 years, with operable breast cancer: features of disease at presentation. Ann Oncol 13:273–279 Lin CH, Lu YS, Huang CS, Kuo KT, Wang CC et al, 2011, Prognostic molecular markers in women aged 35 years or younger with breast cancer: is there a difference from the older patients? J Clin Pathol 64:781–787 Abenhaim HA, Azoulay L, Holcroft CA, Bure LA, Assayag J et al, 2012, Incidence, risk factors, and obstetrical outcomes of women with breast cancer in pregnancy. Breast J 18:564–568 Chun J, Pocock B, Joseph KA, El-Tamer M, Klein L et al, 2009, Breast cancer risk factors in younger and older women. Ann Surg Oncol 16:96–99 Ford D, Easton DF, Stratton M, Narod S, Goldgar D et al, 1998, Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. The Breast Cancer Linkage Consortium. Am J Hum Genet 62:676–689 Van Der Looij M, Szabo C, Besznyak I, Liszka G, Csokay B et al, 2000, Prevalence of founder BRCA1 and BRCA2 mutations among breast and ovarian cancer patients in Hungary. Int J Cancer 86:737–740 Antoniou AC, Hardy R, Walker L, Evans DG, Shenton A et al, 2008, Predicting the likelihood of carrying a BRCA1 or BRCA2 mutation: validation of BOADICEA, BRCAPRO, IBIS, Myriad and the Manchester scoring system using data from UK genetics clinics. J Med Genet 45:425–431 Berry DA, Iversen ES Jr, Gudbjartsson DF, Hiller EH, Garber JE et al, 2002, BRCAPRO validation, sensitivity of genetic testing of BRCA1/BRCA2, and prevalence of other breast cancer susceptibility genes. J Clin Oncol 20:2701–2712 van Harssel JJ, van Roozendaal CE, Detisch Y, Brandao RD, Paulussen AD et al, 2010, Efficiency of BRCAPRO and Myriad II mutation probability thresholds versus cancer history criteria alone for BRCA1/2 mutation detection. Fam Cancer 9:193–201 Hickey M, Peate M, Saunders CM, Friedlander M, 2009, Breast cancer in young women and its impact on reproductive function. Hum Reprod Update 15:323–339 Gaudet MM, Press MF, Haile RW, Lynch CF, Glaser SL, et al., 2011, Risk factors by molecular subtypes of breast cancer across a population-based study of women 56 years or younger. Breast Cancer Res Treat 130:587–597 Han W, Kim SW, Park IA, Kang D, Youn YK et al, 2004, Young age: an independent risk factor for disease-free survival in women with operable breast cancer. BMC Cancer 4:82 Fodor J, Mozsa E, Zaka Z, Polgar C, Major T, 2005, Local relapse in young, < or = 40 years, women with breast cancer after mastectomy or breast conserving surgery: 15-year results. Magy Onkol 49:205–208, 203 Claus EB, RischNJ, ThompsonWD(1990, Age at onset as an indicator of familial risk of breast cancer. Am J Epidemiol 131:961–972 Collaborative Group of Hormonal Factors in Breast Cancer, 2001, Familial breast cancer: collaborative reanalysis of individual data from 52 epidemiological studies including 58,209 women with breast cancer and 101,986 women without the disease. Lancet 358:1389–1399 Loman N, Johannsson O, Kristoffersson U, Olsson H, Borg A, 2001, Family history of breast and ovarian cancers and BRCA1 and BRCA2 mutations in a population-based series of early-onset breast cancer. J Natl Cancer Inst 93:1215–1223 Evans DG, Howell A, Ward D, Lalloo F, Jones JL et al, 2011, Prevalence of BRCA1 and BRCA2 mutations in triple negative breast cancer. J Med Genet 48:520–522 Jiang X, Castelao JE, Chavez-Uribe E, Fernandez Rodriguez B, Celeiro Munoz C et al, 2012, Family history and breast cancer hormone receptor status in a Spanish cohort. PLoS One 7:e29459 van der Hage JA,Mieog JS, van de Velde CJ, Putter H, BartelinkH et al, 2011, Impact of established prognostic factors and molecular subtype in very young breast cancer patients: pooled analysis of four EORTC randomized controlled trials. Breast Cancer Res 13:R68 Kheirelseid EA, Boggs JM, Curran C, Glynn RW, Dooley C et al, 2011, Younger age as a prognostic indicator in breast cancer: a cohort study. BMC Cancer 11:383 Anders CK, Fan C, Parker JS, Carey LA, Blackwell KL et al, 2011, Breast carcinomas arising at a young age: unique biology or a surrogate for aggressive intrinsic subtypes? J Clin Oncol 29:e18–e20 Anders CK, Hsu DS, Broadwater G, Acharya CR, Foekens JA et al, 2008, Young age at diagnosis correlates with worse prognosis and defines a subset of breast cancers with shared patterns of gene expression. J Clin Oncol 26:3324–3330 Ahn SH, Son BH, Kim SW, Kim SI, Jeong J et al, 2007, Poor outcome of hormone receptor-positive breast cancer at very young age is due to tamoxifen resistance: nationwide survival data in Korea–a report from the Korean Breast Cancer Society. J Clin Oncol 25:2360–2368 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 723 EP 729 DI 10.1007/s12253-013-9635-z PG 7 ER PT J AU Kim, HJ Baek, TH Yim, SH Kim, HK Jeong, SH Kang, BH Oh, Ss Lee, GH Kim, WJ Kim, DK AF Kim, Heon Joo Baek, Tae-Hwa Yim, Sun Hyun Kim, Hyun Kyo Jeong, Seong-Hoo Kang, Bum Ho Oh, Sang-seok Lee, Gu Hee Kim, Wha Jae Kim, Dong Kwang TI Collagen Triple Helix Repeat Containing-1 (CTHRC1) Expression in Invasive Ductal Carcinoma of the Breast: The Impact on Prognosis and Correlation to Clinicopathologic Features SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CTHRC1; Immunohistochemistry; Metastasis; Breast cancer ID CTHRC1; Immunohistochemistry; Metastasis; Breast cancer AB CTHRC1 has been known as a regulator of collagen expression and cell migration. The aim of this research was to clarify the clinicopathologic significance of CTHRC1 expression in human breast cancer. 22 cases of breast cancer tissues, randomly selected from clinically diagnosed patients, showed a significant increase of CTHRC1 mRNA expression compared to the normal tissue from the same patients using RT-PCR and real-time PCR. Additionally we investigated breast cancers from 189 patients by immunohistochemistry (IHC). A high level of CTHRC1 expression was observed in 111 (58.7 %) out of 189 breast cancer patients and the expression was significantly correlated with histologic grade (P=0.026), nodal status (P<0.001), and TNM pathologic stage (P=0.002). High CTHRC1 expression was associated with a shorter recurrence free survival (P=0.008). Taken together, the results showed that CTHRC1 over-expression was significantly associated with clinicopathological factors of poor prognosis in invasive ductal carcinoma. CTHRC1 could be used as a supplementary prognostic biomarker and a potential therapeutic target in breast cancer. C1 [Kim, Heon Joo] Eulji University, School of Medicine, Department of Pathology, 301-070 Daejeon, South Korea. [Baek, Tae-Hwa] Eulji University, School of Medicine, Department of Pathology, 301-070 Daejeon, South Korea. [Yim, Sun Hyun] Eulji University, School of Medicine, Department of Pathology, 301-070 Daejeon, South Korea. [Kim, Hyun Kyo] Eulji University, School of Medicine, Preventive Medicine, 301-070 Daejeon, South Korea. [Jeong, Seong-Hoo] Chonbuk National University, Medical School, Department of Surgery, 560-182 Chonju, South Korea. [Kang, Bum Ho] Korea Research Institute of Bioscience and Biotechnology, Medical Genomics Research Center, 305-333 Daejeon, South Korea. [Oh, Sang-seok] Gyeongsang National University, PMBBRC, Division of Applied Life Science (BK21), 660-701 Jinju, South Korea. [Lee, Gu Hee] Korea Research Institute of Bioscience and Biotechnology, Medical Genomics Research Center, 305-333 Daejeon, South Korea. [Kim, Wha Jae] Korea Research Institute of Bioscience and Biotechnology, Medical Genomics Research Center, 305-333 Daejeon, South Korea. [Kim, Dong Kwang] Gyeongsang National University, PMBBRC, Division of Applied Life Science (BK21), 660-701 Jinju, South Korea. RP Kim, DK (reprint author), Gyeongsang National University, PMBBRC, Division of Applied Life Science (BK21), 660-701 Jinju, South Korea. EM kdkim88@gnu.ac.kr CR American Cancer Society, 2011, Global Cancer Facts & Figures 2nd edition http://www.cancer.org/research/cancerfactsfigures/ globalcancerfactsfigures/global-facts-figures-2nd-ed Gonzalez-Angulo AM, Morales-Vasquez F, Hortobagyi GN, 2007, Overview of resistance to systemic therapy in patients with breast cancer. Adv Exp Med Biol 608:1–22 Reis-Filho JS, Pusztai L, 2011, Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet 378:1812–1823 Russnes HG, Navin N, Hicks J, Borresen-Dale AL, 2011, Insight into the heterogeneity of breast cancer through next-generation sequencing. J Clin Invest 121:3810–3818 Prat A, Ellis MJ, Perou CM, 2011, Practical implications of geneexpression- based assays for breast oncologists. Nat Rev Clin Oncol 9:48–57 Pyagay P, Heroult M, Wang Q, Lehnert W, Belden J, Liaw L, Friesel RE, Lindner V, 2005, Collagen triple helix repeat containing 1, a novel secreted protein in injured and diseased arteries, inhibits collagen expression and promotes cell migration. Circ Res 96:261–268 Diatchenko L, Lau YF, Campbell AP, Chenchik A, Moqadam F, Huang B, Lukyanov S, Lukyanov K, Gurskaya N, Sverdlov ED, Siebert PD, 1996, Suppression subtractive hybridization: a method for generating differentially regulated or tissue-specific cDNA probes and libraries. Proc Natl Acad Sci U S A 93:6025–6030 Durmus T, LeClair RJ, Park KS, Terzic A, Yoon JK, Lindner V, 2006, Expression analysis of the novel gene collagen triple helix repeat containing-1, Cthrc1). Gene Expr Patterns 6:935–940 LeClair RJ, Durmus T, Wang Q, Pyagay P, Terzic A, Lindner V, 2007, Cthrc1 is a novel inhibitor of transforming growth factorbeta signaling and neointimal lesion formation. Circ Res 100:826– 833 LeClair R, Lindner V, 2007, The role of collagen triple helix repeat containing 1 in injured arteries, collagen expression, and transforming growth factor beta signaling. Trends Cardiovasc Med 17:202–205 Tang L, Dai DL, Su M, Martinka M, Li G, Zhou Y, 2006, Aberrant expression of collagen triple helix repeat containing 1 in human solid cancers. Clin Cancer Res 12:3716–3722 Turashvili G, Bouchal J, Baumforth K, Wei W, Dziechciarkova M, Ehrmann J, Klein J, Fridman E, Skarda J, Srovnal J, Hajduch M, Murray P, Kolar Z, 2007, Novel markers for differentiation of lobular and ductal invasive breast carcinomas by laser microdissection and microarray analysis. BMC Cancer 7:55 Ip W, Wellman-Labadie O, Tang L, Su M, Yu R, Dutz J, Wang Y, Huang S, Zhang X, Huang C, Zhou Y, 2011, Collagen triple helix repeat containing 1 promotes melanoma cell adhesion and survival. J Cutan Med Surg 15:103–110 Orloff M, Peterson C, He X, Ganapathi S, Heald B, Yang YR, Bebek G, Romigh T, Song JH, Wu W, David S, Cheng Y, Meltzer SJ, Eng C, 2011, Germline mutations in MSR1, ASCC1, and CTHRC1 in patients with Barrett esophagus and esophageal adenocarcinoma. JAMA 306:410–419 Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H, Porter D, Hu M, Chin L, Richardson A, Schnitt S, Sellers WR, Polyak K, 2004, Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell 6:17–32 West RB, Nuyten DS, Subramanian S, Nielsen TO, Corless CL, Rubin BP, Montgomery K, Zhu S, Patel R, Hernandez-Boussard T, Goldblum JR, Brown PO, van de Vijver M, van de Rijn M, 2005, Determination of stromal signatures in breast carcinoma. PLoS Biol 3:e187 Kharaishvili G, Cizkova M, Bouchalova K, Mgebrishvili G, Kolar Z, Bouchal J, 2011, Collagen triple helix repeat containing 1 protein, periostin and versican in primary and metastatic breast cancer: an immunohistochemical study. J Clin Pathol 64:977–982 Robbins P, Pinder S, de Klerk N, Dawkins H, Harvey J, Sterrett G, Ellis I, Elston C, 1995, Histological grading of breast carcinomas: a study of interobserver agreement. Hum Pathol 26:873–879 Elston CW, Ellis IO, 2002, Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 41:154–161 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, Panel members, 2011, Strategies for subtypes–dealing with the diversity of breast cancer: highlights of the St. Gallen international expert consensus on the primary therapy of early breast cancer. Ann Oncol 22:1736–1747 Chomczynski P, Sacchi N, 1987, Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem 162:156–159 Kimura H, Kwan KM, Zhang Z, Deng JM, Darnay BG, Behringer RR, Nakamura T, de Crombrugghe B, Akiyama H, 2008, Cthrc1 is a positive regulator of osteoblastic bone formation. PLoS One 3:e3174 Li J, Cao J, Li M, Yu Y, Yang Y, Xiao X, Wu Z, Wang L, Tu Y, Chen H, 2011, Collagen triple helix repeat containing-1 inhibits transforming growth factor-b1-induced collagen type I expression in keloid. Br J Dermatol 164:1030–1036 Tano K, Mizuno R, Okada T, Rakwal R, Shibato J, Masuo Y, Ijiri K, Akimitsu N, 2010, MALAT-1 enhances cell motility of lung adenocarcinoma cells by influencing the expression of motilityrelated genes. FEBS Lett 584:4575–4580 Yamamoto S, Nishimura O, Misaki K, Nishita M, Minami Y, Yonemura S, Tarui H, Sasaki H, 2008, Cthrc1 selectively activates the planar cell polarity pathway of Wnt signaling by stabilizing the Wnt-receptor complex. Dev Cell 15:23–36 Wang L, Xiang YN, Zhang YH, Tu YT, Chen HX, 2011, Collagen triple helix repeat containing-1 in the differential diagnosis of dermatofibrosarcoma protuberans and dermatofibroma. Br J Dermatol 164:135–140 Li Y, Hively WP, Varmus HE, 2000, Use of MMTV-Wnt-1 transgenic mice for studying the genetic basis of breast cancer. Oncogene 19:1002–1009 Wu Y, Zhou BP, 2008, New insights of epithelial-mesenchymal transition in cancer metastasis. Acta Biochim Biophys Sin, Shanghai, 40:643–650 Ji X, Woodard AS, Rimm DL, Fearon ER, 1997, Transcriptional defects underlie loss of E-cadherin expression in breast cancer. Cell Growth Differ 8:773–778 Cowin P, Rowlands TM, Hatsell SJ, 2005, Cadherins and catenins in breast cancer. Curr Opin Cell Biol 17:499–508 Junghans D, Haas IG, Kemler R, 2005, Mammalian cadherins and protocadherins: about cell death, synapses and processing. Curr Opin Cell Biol 17:446–452 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 731 EP 737 DI 10.1007/s12253-013-9636-y PG 7 ER PT J AU Wu, K Hu, G He, X Zhou, P Li, J He, B Sun, W AF Wu, Kemin Hu, Guohuang He, Xin Zhou, Peng Li, Jian He, Bin Sun, Weijia TI MicroRNA-424-5p Suppresses the Expression of SOCS6 in Pancreatic Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic cancer; miR-424-5p; SOCS6; ERK1/2 signaling pathway ID Pancreatic cancer; miR-424-5p; SOCS6; ERK1/2 signaling pathway AB MicroRNAs (miRNAs) are a group of small noncoding RNA molecules predicted to control the activity of about 30 % of all protein-coding genes in mammals. The expression of microRNA-424-5p (miR-424-5p) has been shown to vary in multiple hematological and solid organ malignancies, such as pancreatic cancer. This study aimed to characterize the function of upregulated miR-424-5p in pancreatic cancer and show how downstream suppressor of cytokine-induced signaling 6 (SOCS6) is negatively regulated by miR-424-5p. MiR-424-5p and SOCS6 expression was detected using quantitative real-time PCR (qRT-PCR) in pancreatic cancer tissues and adjacent non-tumorous ductal epithelium tissues. Luciferase reporter assays were used to assess SOCS6 as a target of miR-424-5p. The downstream effect of SOCS6 was measured by qRT-PCR after miR-424- 5p inhibition and SOCS6 upregulation. The functions of miR- 424-5p in vitro in pancreatic cancer cells were measured by migration and invasion assays and flow cytometry. Results suggested miR-424-5p was significantly upregulated in pancreatic cancer and suppress the expression of SOCS6, and miR-424-5p increased proliferation,migration and invasion of pancreatic cancer cells, while inhibited cell apoptosis. It was concluded that miR-424-5p is frequently upregulated in pancreatic cancer and modulates ERK1/2 signaling pathway by negatively regulating SOCS6. C1 [Wu, Kemin] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan Province, China. [Hu, Guohuang] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan Province, China. [He, Xin] Central South University, Xiangya Hospital, Department of AnesthesiologyChangsha, China. [Zhou, Peng] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan Province, China. [Li, Jian] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan Province, China. [He, Bin] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan Province, China. [Sun, Weijia] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan Province, China. RP Sun, W (reprint author), Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, China. EM sunweijia2010@126.com CR Li D, Xie K, Wolff R, Abbruzzese JL, 2004, Pancreatic cancer. Lancet 363:1049 Guo X, Cui Z, 2005, Current diagnosis and treatment of pancreatic cancer in China. Pancreas 31:13 Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics, 2009. CA Cancer J Clin 59:225 Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60:277 Cameron JL, Riall TS, Coleman J, Belcher KA, 2006, One thousand consecutive pancreaticoduodenectomies. Ann Surg 244:10 Wagner M, Redaelli C, Lietz M, Seiler CA, Friess H, Buchler MW, 2004, Curative resection is the single most important factor determining outcome in patients with pancreatic adenocarcinoma. Br J Surg 91:586 Bartel DP, 2004, MicroRNA’s: genomics biogenesis, mechanism, and function. Cell 116:281 Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR, 2005, MicroRNA expression profiles classify human cancers. Nature 435:834 Garzon R, Calin GA, Croce CM, 2009, MicroRNAs in cancer. Annu Rev Med 60:167 Navon R, Wang H, Steinfeld I, Tsalenko A, Ben-Dor A, Yakhini Z, 2009, Novel rank-based statistical methods reveal microRNAs with differential expression in multiple cancer types. PLoS One 4:e8003 Kozomara A, Griffiths-Jones S, 2011, miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res 39:D152 Wang X, Wang J, Ma H, Zhang J, Zhou X, 2012, Downregulation of miR-195 correlates with lymph node metastasis and poor prognosis in colorectal cancer. Med Oncol 29:919 Donnem T, Fenton CG, Lonvik K, Berg T, Eklo K, Andersen S, Stenvold H, Al-Shibli K, Al-Saad S, Bremnes RM, Busund LT, 2012, MicroRNA signatures in tumor tissue related to angiogenesis in non-small cell lung cancer. PLoS One 7:e29671 Chen X, Hu H, Guan X, Xiong G, Wang Y, Wang K, Li J, Xu X, Yang K, Bai Y, 2012, CpG island methylation status of miRNAs in esophageal squamous cell carcinoma. Int J Cancer 130:1607 Imig J, Motsch N, Zhu JY, Barth S, Okoniewski M, Reineke T, Tinguely M, Faggioni A, Trivedi P, Meister G, Renner C, Grasser FA, 2011, microRNA profiling in Epstein-Barr virus-associated Bcell lymphoma. Nucleic Acids Res 39:1880 Shen Y, Li Y, Ye F, Wang F, Wan X, Lu W, Xie X, 2011, Identification of miR-23a as a novel microRNA normalizer for relative quantification in human uterine cervical tissues. Exp Mol Med 43:358 Lee EJ, Gusev Y, Jiang J, Nuovo GJ, Lerner MR, Frankel WL, Morgan DL, Postier RG, Brackett DJ, Schmittgen TD, 2007, Expression profiling identifies microRNA signature in pancreatic cancer. Int J Cancer 120:1046 Lai RH, Hsiao YW, Wang MJ, Lin HY, Wu CW, Chi CW, Li AF, Jou YS, Chen JY, 2010, SOCS6, down-regulated in gastric cancer, inhibits cell proliferation and colony formation. Cancer Lett 288:75 Hwang MN, Min CH, Kim HS, Lee H, Yoon KA, Park SY, Lee ES, Yoon S, 2007, The nuclear localization of SOCS6 requires the n-terminal region and negatively regulates Stat3 protein levels. Biochem Biophys Res Commun 360:333 Storojeva I, Boulay JL, Ballabeni P, Buess M, Terracciano L, Laffer U, Mild G, Herrmann R, Rochlitz C, 2005, Prognostic and predictive relevance of DNAM-1, SOCS6 and CADH-7 genes on chromosome 18q in colorectal cancer. Oncology 68:246 SasiW, Jiang WG, Sharma A, Mokbel K, 2010, Higher expression levels of SOCS 1,3,4,7 are associated with earlier tumour stage and better clinical outcome in human breast cancer. BMC Cancer 10:178 Bayle J, Letard S, Frank R, Dubreuil P, De Sepulveda P, 2004, Suppressor of cytokine signaling 6 associates with KIT and regulates KIT receptor signaling. J Biol Chem 279:12249 Krebs DL, Uren RT, Metcalf D, Rakar S, Zhang JG, Starr R, De Souza DP, Hanzinikolas K, Eyles J, Connolly LM, Simpson RJ, Nicola NA, Nicholson SE, Baca M, Hilton DJ, Alexander WS, 2002, SOCS-6 binds to insulin receptor substrate 4, and mice lacking the SOCS-6 gene exhibit mild growth retardation. Mol Cell Biol 22:4567 Boucher MJ, Morisset J, Vachon PH, Reed JC, Laine J, Rivard N, 2000, MEK/ERK signaling pathway regulates the expression of Bcl-2, Bcl-X(L), and Mcl-1 and promotessurvival of human pancreatic cancer cells. J Cell Biochem 79:355 Mardin WA, Mees ST, 2009, MicroRNAs: novel diagnostic and therapeutic tools for pancreatic ductal adenocarcinoma? Ann Surg Oncol 16:3183 Rachagani S, Kumar S, Batra SK, 2010, MicroRNA in pancreatic cancer, pathological, diagnostic and therapeutic implications. Cancer Lett 292:8 Wang YX, Zhang XY, Zhang BF, Yang CQ, Chen XM, Gao HJ, 2010, Initial study of microRNA expression profiles of colonic cancer without lymph node metastasis. J Dig Dis 11:50 Pallasch CP, Patz M, Park YJ, Hagist S, Eggle D, Claus R, Debey- Pascher S, Schulz A, Frenzel LP, Claasen J, Kutsch N, Krause G, Mayr C, Rosenwald A, Plass C, Schultze JL, Hallek M, Wendtner CM, 2009, miRNA deregulation by epigenetic silencing disrupts suppression of the oncogene PLAG1 in chronic lymphocytic leukemia. Blood 114:3255 Xu J, Li Y,Wang F,Wang X, Cheng B, Ye F, Xie X, Zhou C, LuW, 2012, Suppressed miR-424 expression via upregulation of target gene Chk1 contributes to the progression of cervical cancer. Oncogene., DOI 10.1038/onc.2012.121 Fulci V, Colombo T, Chiaretti S, Messina M, Citarella F, Tavolaro S, Guarini A, Foa R, Macino G, 2009, Characterization of B- and T-lineage acute lymphoblastic leukemia by integrated analysis of MicroRNA and mRNA expression profiles. Genes Chromosomes Cancer 48:1069 Forrest AR, Kanamori-Katayama M, Tomaru Y, Lassmann T, Ninomiya N, Takahashi Y, de Hoon MJ, Kubosaki A, Kaiho A, Suzuki M, Yasuda J, Kawai J, Hayashizaki Y, Hume DA, Suzuki H, 2010, Induction of microRNAs, mir-155, mir-222, miR-424 and mir-503, promotes monocytic differentiation through combinatorial regulation. Leukemia 24:460 Schmeier S, MacPherson CR, Essack M, Kaur M, Schaefer U, Suzuki H, Hayashizaki Y, Bajic VB, 2009, Deciphering the transcriptional circuitry of microRNA genes expressed during human monocytic differentiation. BMC Genom 10:595 Rosa A, Ballarino M, Sorrentino A, Sthandier O, De Angelis FG, Marchioni M, Masella B, Guarini A, Fatica A, Peschle C, Bozzoni I, 2007, The interplay between the master transcription factor PU.1 and miR-424 regulates human monocyte/macrophage differentiation. Proc Natl Acad Sci U S A 104:19849 Krebs DL, Hilton DJ, 2000, SOCS: physiological suppressors of cytokine signaling. J Cell Sci 113:2813 Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A, Hong SM, Fu B, Lin MT, Calhoun ES, Kamiyama M, Walter K, Nikolskaya T, Nikolsky Y, Hartigan J, Smith DR, Hidalgo M, Leach SD, Klein AP, Jaffee EM, Goggins M, Maitra A, Iacobuzio-Donahue C, Eshleman JR, Kern SE, Hruban RH, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW, 2008, Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321:1801–1806 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 739 EP 748 DI 10.1007/s12253-013-9637-x PG 10 ER PT J AU Zhu, M Xu, Y Mao, X Gao, Y Shao, L Yan, F AF Zhu, Mingchen Xu, Yijun Mao, Xuelian Gao, Yanfang Shao, Lijia Yan, Feng TI Overexpression of Metastasis-Associated in Colon Cancer-1 Associated with Poor Prognosis in Patients with Esophageal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MACC1; Immunohistochemistry; Prognosis; Esophageal caner ID MACC1; Immunohistochemistry; Prognosis; Esophageal caner AB Recent studies have shown that expression of metastasis-associated in colon cancer-1(MACC1) is observed in different types of cancer and plays an important role in tumor metastasis. However, the expression of MACC1 and its possible role in esophageal cancer remains unknown. In this study, we determined the expression of MACC1 in esophageal cancer by utilizing immunohistochemistry and analyzed the relationship between the expression and esophageal cancer prognosis. Immunohistochemistry results showed that 47 of 85 cancer lesions (55.2 %) were stained positive, and high expression of MACC1 was correlated with the node metastasis and TNM stage (P<0.05). The Kaplan-Meier survival curve showed that patients with high MACC1 expression had significantly reduced overall 5-year survival rates (P=0.004). Cox regression analysis revealed that high expression of MACC1 was associated with increased risk of death (hazard ratio [HR] =2.25) in patients with esophageal cancer. These findings suggested that high expression of MACC1 was correlated with progression and metastasis of esophageal cancer and might serve as a novel prognostic marker for patients with esophageal cancer. C1 [Zhu, Mingchen] Jiangsu Cancer Hospital, Department of Clinical Laboratory, 42 Baiziting Road, 210009 Nanjing, Jiangsu, China. [Xu, Yijun] Nanjing First Hospital & Nanjing Medical University First Hospital, Department of Gastroenterology, 68 Changle Road, 210006 Nanjing, Jiangsu, China. [Mao, Xuelian] Jiangsu Cancer Hospital, Department of Clinical Laboratory, 42 Baiziting Road, 210009 Nanjing, Jiangsu, China. [Gao, Yanfang] Jiangsu Cancer Hospital, Department of Clinical Laboratory, 42 Baiziting Road, 210009 Nanjing, Jiangsu, China. [Shao, Lijia] Jiangsu Cancer Hospital, Department of Clinical Laboratory, 42 Baiziting Road, 210009 Nanjing, Jiangsu, China. [Yan, Feng] Jiangsu Cancer Hospital, Department of Clinical Laboratory, 42 Baiziting Road, 210009 Nanjing, Jiangsu, China. RP Yan, F (reprint author), Jiangsu Cancer Hospital, Department of Clinical Laboratory, 210009 Nanjing, China. EM yanfeng2007@sohu.com CR Pisani P, Parkin DM, Bray F et al, 1999, Erratum: estimates of the worldwide mortality from 25 cancers in 1990. Int. J. Cancer, 83, 18–29, 1999). Int J Cancer 83:870–873 Enzinger PC, Mayer RJ, 2003, Esophageal cancer. N Engl J Med 349:2241–2252 Blanchard P, Quero L, Hennequin C, 2009, Prognostic and predictive factors of oesophageal carcinoma. Bull Cancer 96:379–389 Lin YC, Wu MY, Li DR et al, 2004, Prognostic and clinicopathological features of E-cadherin, alpha-catenin, beta-catenin, gamma-catenin and cyclin D1 expression in human esophageal squamous cell carcinoma. World J Gastroenterol 10:3235–3239 Michaylira CZ, Wong GS, Miller CG et al, 2010, Periostin, a cell adhesion molecule, facilitates invasion in the tumor microenvironment and annotates a novel tumor-invasive signature in esophageal cancer. Cancer Res 70:5281–5292 Kadowaki T, Shiozaki H, Inoue M et al, 1994, E-cadherin and alphacatenin expression in human esophageal cancer. Cancer Res 54:291–296 Chen M, Cai E, Huang J et al, 2012, Prognostic value of vascular endothelial growth factor expression in patients with esophageal cancer: a systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev 21:1126–1134 Stoker M, Gherardi E, Perryman M et al, 1987, Scatter factor is a fibroblast-derived modulator of epithelial cell mobility. Nature 327:239–242 Lesko E, Majka M, 2008, The biological role of HGF-MET axis in tumor growth and development of metastasis. Front Biosci 13:1271–1280 Horiguchi N, Takayama H, Toyoda M et al, 2002, Hepatocyte growth factor promotes hepatocarcinogenesis through c-Met autocrine activation and enhanced angiogenesis in transgenic mice treated with diethylnitrosamine. Oncogene 21:1791–1799 Zhang YW, Vande Woude GF, 2003, HGF/SF-met signaling in the control of branching morphogenesis and invasion. J Cell Biochem 88:408–417 Vande Woude GF, Jeffers M, Cortner J et al, 1997, Met-HGF/SF: tumorigenesis, invasion and metastasis. CIBA Found Symp 212:119–130, discussion 130–2, 148–154 Gumustekin M, Kargi A, Bulut G et al, 2011, HGF/c-Met overexpressions, but not met mutation, correlates with progression of non-small cell lung cancer. Pathol Oncol Res 18:209–218 Toschi L, Janne PA, 2008, Single-agent and combination therapeutic strategies to inhibit hepatocyte growth factor/ MET signaling in cancer. Clin Cancer Res 14:5941–5946 Birchmeier C, Birchmeier W, Gherardi E et al, 2003, Met, metastasis, motility and more. Nat Rev Mol Cell Biol 4:915–925 Okano J, Shiota G, Matsumoto K et al, 2003, Hepatocyte growth factor exerts a proliferative effect on oval cells through the PI3K/AKT signaling pathway. Biochem Biophys Res Commun 309:298–304 Ye M, Hu D, Tu L et al, 2008, Involvement of PI3K/Akt signaling pathway in hepatocyte growth factor-induced migration of uveal melanoma cells. Investig Ophthalmol Vis Sci 49:497–504 Stein U,Walther W, Arlt F et al, 2009, MACC1, a newly identified key regulator of HGF-MET signaling, predicts colon cancer metastasis. Nat Med 15:59–67 Shirahata A, Sakata M, Kitamura Y et al, 2010, MACC 1 as a marker for peritoneal-disseminated gastric carcinoma. Anticancer Res 30:3441–3444 Chundong G, Uramoto H, Onitsuka T et al, 2011, Molecular diagnosis of MACC1 status in lung adenocarcinoma by immunohistochemical analysis. Anticancer Res 31:1141–1145 Shirahata A, Fan W, Sakuraba K et al, 2011, MACC 1 as a marker for vascular invasive hepatocellular carcinoma. Anticancer Res 31:777–780 Lang AH, Geller-Rhomberg S, Winder T et al, 2012, A common variant of the MACC1 gene is significantly associated with overall survival in colorectal cancer patients. BMC Cancer 12:20 Qiu J, Huang P, Liu Q et al, 2011, Identification of MACC1 as a novel prognostic marker in hepatocellular carcinoma. J Transl Med 9:166 Steeg PS, 2006, Tumor metastasis: mechanistic insights and clinical challenges. Nat Med 12:895–904 Gupta GP, Massague J, 2006, Cancer metastasis: building a framework. Cell 127:679–695 Fidler IJ, 2002, Critical determinants of metastasis. Semin Cancer Biol 12:89–96 Liang P, Pardee AB, 1992, Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction. Science 257:967–971 Peschard P, Park M, 2007, From Tpr-Met to Met, tumorigenesis and tubes. Oncogene 26:1276–1285 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 749 EP 753 DI 10.1007/s12253-013-9638-9 PG 5 ER PT J AU Cireap, N Narita, D AF Cireap, Natalia Narita, Diana TI Molecular Profiling of ADAM12 and ADAM17 Genes in Human Malignant Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ADAM12; ADAM17; Melanoma; Gene expression; Cytokines ID ADAM12; ADAM17; Melanoma; Gene expression; Cytokines AB ADAM12 and ADAM17 proteins belong to a family of transmembrane disintegrin-containing metalloproteinases (ADAMs) involved in the proteins ectodomain shedding and cell-cell and cell-matrix interactions. However, the specific biological functions of ADAMs are still unclear and, until now, these proteins were not investigated yet in melanoma. The aim of this study was to analyze the splicing variants of ADAM12 (L and S) and ADAM17 gene expression in melanoma at transcriptional and translational level in comparison with control (non-tumor) tissues. Taking in account that ADAM17 sheddase is involved in the modulation of TNF-α (tumor necrosis factor alpha), we analyzed also this cytokine in the plasma of the same patients before any treatment, and we compared the results with healthy controls. Quantitative-RT-PCR and immunohistochemistry were used to analyze ADAM12 and ADAM17 genes expression and the analysis of TNF-α expression was carried out in the plasma using ELISA. We demonstrated that ADAM12L splicing variant together with ADAM17 gene are strongly overexpressed in melanomas, whereas ADAM12S, although up-regulated when compared with the non-tumor controls, the difference was not statistically significant. When we compared the levels of expression for the ADAMs genes according to the tumor stage, we observed that all three investigated genes were significantly overexpressed in advanced stage in comparison with early stage melanomas. In the plasma of the same patients, the expression of TNF-α was up-regulated and significantly correlated with the expression of ADAM17 and respectively, with the advanced tumor stage. C1 [Cireap, Natalia] University of Medicine and Pharmacy, Department of Surgical OncologyTimisoara, Romania. [Narita, Diana] University of Medicine and Pharmacy, Biochemistry Department, E. Murgu 2, 300041 Timisoara, Romania. RP Narita, D (reprint author), University of Medicine and Pharmacy, Biochemistry Department, 300041 Timisoara, Romania. EM diana_narita@yahoo.com;diananarita@umft.ro CR Altekruse SF, Kosary CL, Krapcho M et al, 2010, SEER Cancer Statistics Review 1975–2007, National Cancer Institute, Bethesda, MD http://seer.cancer.gov/csr/1975_2007/, based on November 2009 SEER data submission, posted to the SEER web site Peschon JJ, SlackJ L, Reddy P et al, 1998, An essential role for ectodomain shedding in mammalian development. Science 282:1281–1284 Ohtsu H, Dempsey PJ, Eguchi S, 2006, ADAMs as mediators of EGF receptor transactivation by G-protein-coupled receptors. Am J Physiol Cell Physiol 291:C1–C10 Mochizuki A, Okada Y, 2007, ADAMs in cancer cell proliferation and progression. Cancer Sci 98:621–628 Murphy G, 2008, The ADAMs: signaling scissors in the tumor microenvironment. Nat Rev Cancer 8:929–941 Edwards DR, Handsley MM, Pennington CJ, 2008, The ADAM metalloproteinases. Mol Asp Med 29:258–289 Blobel CP, 2005, ADAMS: key components in EGFR signalling and development. Nat Rev Mol Cell Biol 6:32–43 Arribas J, Bech-Serra JJ, Santiago-Josefat B, 2006, ADAMs, cell migration and cancer. Cancer Metastasis Rev 25:57–68 Wewer UM, Morgelin M, Holck P et al, 2006, ADAM12 is a fourleafed clover. The excised prodomain remains bound to the mature enzyme. J Biol Chem 281:9418–9422 Duffy MJ, McKiernan E, O’Donovan N, McGowan P, 2009, Role of ADAMs in cancer formation and progression. Clin Cancer Res 15:1140–1144 Iba K, Albrechtsen R, Gilpin B et al, 2000, The cysteine-rich domain of human ADAM 12 supports cell adhesion through syndecans and triggers signaling events that lead to β1 integrindependent cell spreading. J Cell Biol 149:1143–1156 Atfi A, Dumont E, Colland F et al, 2007, The disintegrin and metalloproteinase ADAM12 contributes to TGF-β signaling through interaction with the type II receptor. J Cell Biol 178:201–208 Dyczynska E, Syta E, Sun D, Zolkiewska A, 2008, Breast cancerassociated mutations in metalloprotease disintegrin ADAM12 interfere with the intracellular trafficking and processing of the protein. Int J Cancer 122:2634–2640 Le Pabic H, Bonnier D, Wewer UM et al, 2003, ADAM12 in human liver cancers: TGF-β-regulated expression in stellate cells is associated with matrix remodeling. Hepatology 37:1056–1066 Borrell-Pages M, Rojo F, Albanell J, Baselga J, Arribas J, 2003, TACE is required for the activation of the EGFR by TGF-alpha in tumors. EMBO J 22:1114–1124 Lu X, Lu D, Scully M, KakkarV(2008)ADAMproteins—therapeutic potential in cancer. Curr Cancer Drug Targets 8:720–732 Shi Z, XuW, Loechel F,Wewer UM,Murphy LJ, 2000)ADAM12, a disintegrin and metalloprotease, interacts with insulin-like growth factor-binding protein-3. J Biol Chem 275:18574–18580 Laigaard J, Sorensen T, Frohlich C et al, 2003, ADAM12: a novel first-trimester maternal serum marker for Down syndrome. Prenat Diagn 23:1086–1091 Roy R, Wewer UM, Zurakowski D, Pories SE, Moses MA, 2004, ADAM12 cleaves extracellular matrix proteins and correlates with cancer status and stage. J Biol Chem 279:51323–51330 Pories SE, Zurakowski D, Roy R et al, 2008, Urinary metalloproteinases: noninvasive biomarkers for breast cancer risk assessment. Cancer Epidemiol Biomarkers Prev 17:1034–1042 McGowan PM, McKiernan E, Bolster F et al, 2008, ADAM-17 predicts adverse outcome in patients with breast cancer. Ann Oncol 19:1075–1081 O’Shea C, McKie N, Buggy Y et al, 2003, Expression of ADAM-9 mRNA and protein in human breast cancer. Int J Cancer 105:754–761 Lendeckel U, Kohl J, Arndt M et al, 2005, Increased expression of ADAM family members in human breast cancer and breast cancer cell lines. J Cancer Res Clin Oncol 31:41–48 Kuefer R, Day KC, Kleer CG et al, 2006, ADAM15 disintegrin is associated with aggressive prostate and breast cancer diseases. Neoplasia 8:319–329 Kveiborg M, Albrechtsen R, Couchman JR, Wewer UM, 2008, Cellular roles of ADAM12 in health and disease. Int J Biochem Cell Biol 40:1685–1702 Jacobsen J, Wewer UM, 2009, Targeting ADAM12 in human disease: head, body or tail? Curr Pharm Des 15:2300–2310 Frohlich C, Albrechtsen R, Dyrskjot L et al, 2006)Molecular profiling of ADAM12 in human bladder cancer. Clin Cancer Res 12:7359–7368 Kveiborg M, Frohlich C, Albrechtsen R et al, 2005, A role for ADAM 12 in breast tumor progression and stromal cell apoptosis. Cancer Res 65:4754–4760 Peduto L, Reuter VE, Sehara-Fujisawa A et al, 2006, ADAM12 is highly expressed in carcinoma-associated stroma and is required for mouse prostate tumor progression. Oncogene 25:5462–5466 Sorensen HP, Vives RR, Manetopoulos C et al, 2008, Heparan sulfate regulates ADAM12 through a molecular switch mechanism. J Biol Chem 46:31920–31932 Black RA, Rauch CT, Kozlosky CJ et al, 1997, A metalloproteinase disintegrin that releases tumour necrosis factor-alpha from cells. Nature 385:729–733 Zheng Y, Saftig P, Hartmann D, Blobel C, 2004, Evaluation of the contribution of different ADAMs to tumor necrosis factor α, TNF α, shedding and of the function of the TNF α ectodomain in ensuring selective stimulated shedding by the TNF α convertase, TACE/ADAM17). J Biol Chem 41:42898–42906 Kawaguchi N, Horiuchi K, Becherer JD et al, 2007, Different ADAMs have distinct influences on Kit ligand processing: phorbol-ester-stimulated ectodomain shedding of Kitl1 by ADAM17 is reduced by ADAM19. J Cell Sci 120:943–952 Horiuchi K, Le Gall S, Schulte M et al, 2007, Substrate selectivity of epidermal growth factor-receptor ligand sheddases and their regulation by phorbol esters and calcium influx. Mol Biol Cell 18:176–188 Franovic A, Robert I, Smith K et al, 2006, Multiple acquired renal carcinoma tumor capabilities abolished upon silencing of ADAM17. Cancer Res 66:8083–8090 Tanaka Y, Miyamoto S, Suzuki SO et al, 2005, Clinical significance of heparin-binding epidermal growth factor-like growth factor and a disintegrin and metalloprotease 17 expression in human ovarian cancer. Clin Cancer Res 11:4783–4792 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method. Methods 25:402–408 Narita D, Seclaman E, Ursoniu S, Anghel A, 2011, Increased expression of ADAM12 and ADAM17 genes in laser-capture microdissected breast cancers and correlations with clinical and pathological characteristics. Acta Histochem., DOI 10.1016/j.acthis.2011.03.009 Rocks N, Paulissen G, Quesada Calvo F et al, 2006, Expression of a disintegrin and metalloprotease, ADAM and ADAMTS, enzymes in human non-small-cell lung carcinomas, NSCLC). Br J Cancer 94:724–730 Kodama T, Ikeda E, Okada A et al, 2004, ADAM12 is selectively overexpressed in human glioblastomas and is associated with glioblastoma cell proliferation and shedding of heparin-binding epidermal growth factor. Am J Pathol 165:1743–1753 Moss ML, Stoeck A, Yan W, Dempsey PJ, 2008, ADAM10 as a target for anti-cancer therapy. Curr Pharm Biotechnol 9:2–8 Horiuchi K, Zhou HM, Kelly K, Manova K, Blobel CP, 2005, Evaluation of the contributions of ADAMs 9, 12, 15, 17, and 19 to heart development and ectodomain shedding of neuregulins beta1 and beta 2. Dev Biol 283:459–471 Zhou BB, 2005, Targeting ligand cleavage to inhibit the ErbB pathway in cancer. Ann NY Acad Sci 1059:56–60 Ziober BL, Chen YQ, Ramos DM, Waleh N, Kramer RH, 1999, Expression of the alpha7 beta1 laminin receptor suppresses melanoma growth and metastatic potential. Cell Growth Differ 10:479–490 Kramer RH, Vu MP, Cheng YF et al, 1991, Laminin-binding integrin alpha 7 beta 1: functional characterization and expression in normal and malignant melanocytes. Cell Regul 2:805–817 Oh ST, Cho BK, Schramme A et al, 2009, Hair-cycle dependent differential expression of ADAM 10 and ADAM 12, an immunohistochemical analysis in human hair follicles in situ. Dermatoendocrinol 1:46–53 Harsha A, Stojadinovic O, Brem H et al, 2008, ADAM12: a potential target for the treatment of chronic wounds. J Mol Med 86:961–969 Zigrino P, Mauch C, Fox JW, Nischt R, 2005, ADAM-9 expression in human skin melanoma and melanoma cell lines. Int J Cancer 116:853–859 Sunnarborg SW, Hinkle CL, Stevenson M et al, 2002, Tumor necrosis factor-alpha converting enzyme, TACE, regulates epidermal growth factor receptor ligand availability. J Biol Chem 277:12838–12845 Fowlkes JL, Winkler MK, 2002, Exploring the interface between metallo-proteinase activity and growth factor and cytokine bioavailability. Cytokine Growth Factor Rev 13:277–287 Lazar-Molnar E, Hegyesi H, Toth S, Falus A, 2000, Autocrine and paracrine regulation by cytokines and growth factors in melanoma. Cytokines 12:547–554 Elias EG, Hasskamp JH, Sharma BK, 2010, Cytokines and growth factors expressed by human cutaneous melanoma. Cancers 2:794–808 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 755 EP 762 DI 10.1007/s12253-013-9639-8 PG 8 ER PT J AU Arul, D Subramanian, P AF Arul, Duraikannu Subramanian, Perumal TI Naringenin (Citrus Flavonone) Induces Growth Inhibition, Cell Cycle Arrest and Apoptosis in Human Hepatocellular Carcinoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Naringenin; Hepatocellular carcinoma cells; Cell proliferation; Apoptosis ID Naringenin; Hepatocellular carcinoma cells; Cell proliferation; Apoptosis AB Search for new substances with antiproliferative activity and apoptosis inducing potential towards HepG2 cells is important since HCC is notoriously resistant to conventional chemotherapy. Dietary phytochemicals with significant anti-proliferative and apoptosis inducing potential are considered as agents promising for cancer therapy. Naringenin, a common dietary flavonoid abundantly present in fruits and vegetables, is believed to possess strong cytotoxic activity in numerous types of cancer cells. However, the detailed molecular mechanisms of its antiproliferative effects and apoptosis induction are still unclear. In this study, we investigated antiproliferative and apoptosisinducing effect of naringenin in human hepatocellular carcinoma HepG2 cells. Naringenin was shown to inhibit the proliferation of HepG2 cells resulted partly from an accumulation of cells in the G0/G1 and G2/M phase of the cell cycle. Naringenin induced a rapid accumulation of p53, which might account for the naringenin-induced G0/G1 and G2/M phase arrests in Hep G2 cells. In addition, naringenin have been shown to induce apoptosis as evidenced by nuclei damage and increased proportion of apoptotic cells detected by flow cytometry analysis. Naringenin triggered the mitochondrial-mediated apoptosis pathway as shown by an increased ratio of Bax/Bcl-2, subsequent release of cytochrome C, and sequential activation of caspase-3. Our results showed that naringenin had inhibitory effect on the growth of HepG2 cell line through inhibition of cell proliferation and apoptosis induction. The elucidation of the drug targets of naringenin on inhibition of tumor cells growth should enable further development of naringenin for liver cancer therapy. C1 [Arul, Duraikannu] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalainagar, 608 002 Tamil Nadu, India. [Subramanian, Perumal] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalainagar, 608 002 Tamil Nadu, India. RP Arul, D (reprint author), Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, 608 002 Tamil Nadu, India. EM sent.biochem@gmail.com CR Semela D, Heim M, 2011, Hepatocellular carcinoma. Ther Umsch 68:213–217 Wysocki PJ, 2010, Targeted therapy of hepatocellular cancer. Expert Opin Investig Drugs 19:265–274 Zender L, Kubicka S, 2008, Molecular pathogenesis and targeted therapy of hepatocellular carcinoma. Onkologie 31:550–5 Ye SL, Takayama T, Geschwind J, Marrero JA, Bronowicki JP, 2010, Current approaches to the treatment of early hepatocellular carcinoma. Oncologist 15:34–41 Zhang ZM, Guo JX, Zhang ZC, Jiang N, Zhang ZY, Pan LJ, 2011, Therapeutic options for intermediate-advanced hepatocellular carcinoma. World J Gastroenterol 17:1685–1689 Tan AC, Konczak I, Sze DM, Ramzan I, 2011, Molecular pathways for cancer chemoprevention by dietary phytochemicals. Nutr Cancer 63:495–505 Diaz-Laviada I, 2010, Effect of capsaicin on prostate cancer cells. Future Oncol 6:1545–1550 Lee KW, Bode AM, Dong Z, 2011, Molecular targets of phytochemicals for cancer prevention. Nat Rev Cancer 11:211–218 Harapu CD, Miron A, Cuciureanu M, Cuciureanu R, 2010, Flavonoids-bioactive compounds in fruits juice. Rev Med Chir Soc Med Nat Iasi 114:1209–1214 Wahle KW, Brown I, Rotondo D, Heys SD, 2011, Plant phenolics in the prevention and treatment of cancer. Adv Exp Med Biol 698:36–51 Clere N, Faure S, Martinez MC, Andriantsitohaina R, 2011, Anticancer properties of flavonoids: Roles in various stages of carcinogenesis. Cardiovasc Hematol Agents Med Chem 9:62–77 Renugadevi J, Prabu SM, 2010, Cadmium-induced hepatotoxicity in rats and the protective effect of naringenin. Exp Toxicol Pathol 62:171–181 Sabarinathan D, Mahalakshmi P, Vanisree AJ, 2011, Naringenin, a flavanone inhibits the proliferation of cerebrally implanted C6 glioma cells in rats. Chem Biol Interact 189:26–36 Ekambaram G, Rajendran P, Magesh V, Sakthisekaran D, 2008, Naringenin reduces tumor size and weight lost in N-methyl-N’- nitro-N-nitrosoguanidine-induced gastric carcinogenesis in rats. Nutr Res 28:106–112 Qin L, Jin L, Lu L, Lu X, Zhang C, Zhang F, Liang W, 2011, Naringenin reduces lung metastasis in a breast cancer resection model. Protein Cell 2:507–516 Koopman G, Reutelingsperger CP, Kuijten GA, Keehnen RM, Pals ST, Van Oers MH, 1994, Annexin V for flow cytometric detection of phosphatidylserine expression on B cells undergoing apoptosis. Blood 84:1415–1420 Bradford MM, 1976, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72:248–254 Laemmli UK, 1970, Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227:680–685 Knowles LM, Zigrossi DA, Tauber RA, Hightower C, Milner JA, 2000, Flavonoids suppress androgen-independent human prostate tumor proliferation. Nutr Cancer 38:116–122 Wang H, Li YL, She WZ, Guo GQ, Jiang ZY, Cen YZ, Fan ZY, 2007, Studies on antiproliferative effect of flavones compounds isolated from Yao herb medicines. Zhong Yao Cai 30:980–984 Lin E, Lin WH, Wang SY, Chen CS, Liao JW, Chang HW, Chen SC, Lin KY, Wang L, Yang HL, Hseu YC, 2012, Flavokawain B inhibits growth of human squamous carcinoma cells: Involvement of apoptosis and cell cycle dysregulation in vitro and in vivo. J Nutr Biochem 23:368–378 Zhong Z, Chen X, Tan W, Xu Z, Zhou K, Wu T, Cui L, Wang Y, 2011, Germacrone inhibits the proliferation of breast cancer cell lines by inducing cell cycle arrest and promoting apoptosis. Eur J Pharmacol 667:50–55 Suzuki K, Matsubara H, 2011, Recent advances in p53 research and cancer treatment. J Biomed Biotechnol 2011:978312., DOI 10.1155/2011/978312 Giono LE, Manfredi JJ, 2006, The p53 tumor suppressor participates in multiple cell cycle checkpoints. J Cell Physiol 209:13–20 Taylor WR, Stark GR, 2001, Regulation of the G2/M transition by p53. Oncogene 20:1803–1815 Alshatwi AA, Shafi G, Hasan TN, Al-Hazzani AA, Alsaif MA, Alfawaz MA, Lei KY, Munshi A, 2011, Apoptosis-mediated inhibition of human breast cancer cell proliferation by lemon citrus extract. Asian Pac J Cancer Prev 12:1555–1559 Park HS, Kim GY, Nam TJ, Deuk Kim N, Hyun Choi Y, 2011, Antiproliferative activity of fucoidan was associated with the induction of apoptosis and autophagy in AGS human gastric cancer cells. J Food Sci 26:77–83 Tan AC, Konczak I, Ramzan I, Sze DM, 2011, Native Australian fruit polyphenols inhibit cell viability and induce apoptosis in human cancer cell lines. Nutr Cancer 63:444–55 Chidambara Murthy KN, Jayaprakasha GK, Kumar V, Rathore KS, Patil BS, 2011, Citrus limonin and its glucoside inhibit colon adenocarcinoma cell proliferation through apoptosis. J Agric Food Chem 59:2314–2323 Plati J, Bucur O, Khosravi-Far R, 2011, Apoptotic cell signaling in cancer progression and therapy. Integr Biol, Camb, 4:279–296 Kelly PN, Strasser A, 2011, The role of Bcl-2 and its pro-survival relatives in tumourigenesis and cancer therapy. Cell Death Differ 18:1414–1424 Martinou JC, Youle RJ, 2011, Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Dev Cell 21:92–101 Wang G, Zhan Y, Wang H, Li W, 2012, ABT-263 sensitizes TRAIL-resistant hepatocarcinoma cells by downregulating the Bcl-2 family of anti-apoptotic protein. Can Chemother Pharmacol 69:799–805 Jin CY, Park C, Lee JH, Chung KT, Kwon TK, Kim GY, Choi BT, Choi YH, 2009, Naringenin-induced apoptosis is attenuated by Bcl-2 but restored by the small molecule Bcl-2 inhibitor, HA 14–1, in human leukemia U937 cells. Toxicol In Vitro 23:259–65 Park JH, Jin CY, Lee BK, Kim GY, Choi YH, Jeong YK, 2008, Naringenin induces apoptosis through downregulation of Akt and caspase-3 activation in human leukemia THP-1 cells. Food Chem Toxicol 46:3684–3690 Lee ER, Kang YJ, Choi HJ, Kang GH, Kim JH, Kim BW, Han YS, Nah SY, Paik HD, Park YS, 2007, Cho SG, 2007, induction of apoptotic cell death by synthetic naringenin derivatives in human lung epithelial carcinoma A549 cells. Biol Pharm Bull 30:2394–8 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 763 EP 770 DI 10.1007/s12253-013-9641-1 PG 8 ER PT J AU Huang, L Wang, Sh Li, ShSh Yang, XM AF Huang, Li Wang, Shuang Li, Shi-Sheng Yang, Xin-Ming TI Prognostic Significance of Beclin-1 Expression in Laryngeal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Beclin-1; Laryngeal squamous cell carcinoma; Metastasis; Prognosis ID Beclin-1; Laryngeal squamous cell carcinoma; Metastasis; Prognosis AB Beclin-1 plays a critical role in the regulation of autophagy, apoptosis, differentiation and the development and progression of cancer. The aim of the present investigation was to analyze the Beclin-1 protein expression and to assess its prognostic significance in tissue of laryngeal squamous cell carcinoma (LSCC). Beclin-1 protein expression in 82 primary laryngeal squamous cell carcinoma and 40 paracarcinoma non-tumor tissue samples was analyzed by immunohistochemistry and correlated with clinicopathological parameters and patients’ outcome. The expression of Beclin-1 in tumor tissues was significantly lower than that in non-tumor tissues (P=0.035). Reduced Beclin-1 expression was significantly correlated with lymph node metastases (P=0.021). Kaplan–Meier survival estimates showed a significant correlation between Beclin-1 expression and patient’s survival rate (log-rank P<0.05).Multivariate Cox proportional hazards model analysis confirmed that lymph node metastases (P=0.048) and Beclin-1 expression (P=0.029) were statistically significant, independent prognostic factors for LSCC. Our findings suggested that decreased Beclin-1 expression and lymph node metastases, as examined by immunohistochemistry, are both independent biomarker for poor prognosis of patients with LSCC. C1 [Huang, Li] Sichuan University, West China Hospital, Department of Otolaryngology-Head and Neck Surgery, 37, Guoxue Lane, 610041 Chengdu, China. [Wang, Shuang] Central South University, The Second XiangYa Hospital, Department of Otolaryngology-Head and Neck Surgery, 410011 Changsha, Hunan, China. [Li, Shi-Sheng] Central South University, The Second XiangYa Hospital, Department of Otolaryngology-Head and Neck Surgery, 410011 Changsha, Hunan, China. [Yang, Xin-Ming] Central South University, The Second XiangYa Hospital, Department of Otolaryngology-Head and Neck Surgery, 410011 Changsha, Hunan, China. RP Yang, XM (reprint author), Central South University, The Second XiangYa Hospital, Department of Otolaryngology-Head and Neck Surgery, 410011 Changsha, China. EM x16y2003@yahoo.com.cn CR Carvalho AL, Nishimoto IN, Califano JA, Kowalski LP, 2005, Trends in incidence and prognosis for head and neck cancer in the United States: a site-specific analysis of the SEER database. Int J Cancer 114:806–816 Assuncao Guimaraes C, Linden R, 2004, Programmed cell death. Apoptosis and alternative deathstyles. Eur J Biochem 271:1638–1650 Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T et al, 2008, Cancer statistics, 2008. CA Cancer J Clin 58:71–96 Cosetti M, Yu GP, Schantz SP, 2008, Five-year survival rates and time trends of laryngeal cancer in the US population. Arch Otolaryngol Head Neck Surg 134:370–379 Chu CT, Zhu J, Dagda R, 2007, Beclin1-independent pathway of damage-induced mitophagy and autophagic stress: implications for neurodegeneration and cell death. Autophagy 3:663–666 Kihara A, Kabeya Y, Ohsumi Y, Yoshimori T, 2001, Beclinphosphatidylinositol 3-kinase complex functions at the trans- Golgi network. EMBO Rep 2:330–335 Yue Z, Jin S, Yang C, Levine AJ, Heintz N, 2003, Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci U S A 100:15077–15082 Ahn CH, Jeong EG, Lee JW, Kim MS, Kim SH, Kim SS, Yoo NJ, Lee SH, 2007, Expression of beclin-1, an autophagy-related protein, in gastric and colorectal cancers. APMIS 115:1344–1349 Smeele LE, Leemans CR, Langendijk JA, Tiwari R, Slotman BJ, van DerWaal I, Snow GB, 2000, Positive surgical margins in neck dissection specimens in patients with head and neck squamous cell carcinoma and the effect of radiotherapy. Head Neck 22:559–563 Thabet MH, Talaat M, Rizk AM, 2000, Pitfalls in the surgical management of cancer of the larynx and hypopharynx. Otolaryngol Head Neck Surg 123:482–487 Sobin LH, Wittekind CH, 2002, TNM classification of malignant tumours, UICC), 6th edn. Wiley-Liss, New York Li BX, Li CY, Peng RQ,Wu XJ,Wang HY,Wan DS, Zhu XF, Zhang XS, 2009, The expression of beclin 1 is associated with favorable prognosis in stage IIIB colon cancers. Autophagy 5:303–306 Kim HS, Lee SH, Do SI, Lim SJ, Park YK, Kim YW, 2011, Clinicopathologic correlation of beclin-1 expression in pancreatic ductal adenocarcinoma. Pathol Res Pract 207:247–252 Mathew R, Kongara S, Beaudoin B, Karp CM, Bray K, Degenhardt K, Chen G, Jin S, White E, 2007, Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev 21:1367–1381 Levine B, Klionsky DJ, 2004, Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell 6:463–477 Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, Thomas-Tikhonenko A, Thompson CB, 2007, Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest 117:326–336 Aita VM, Liang XH, Murty VV, Pincus DL, Yu W, Cayanis E, Kalachikov S, Gilliam TC, Levine B, 1999, Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics 59:59–65 Mathew R, White E, 2011, Autophagy in tumorigenesis and energy metabolism: friend by day, foe by night. Curr Opin Genet Dev 21:113–119 Kenific CM, Thorburn A, Debnath J, 2010, Autophagy and metastasis: another double-edged sword. Curr Opin Cell Biol 22:241– 245 Karantza-Wadsworth V, Patel S, Kravchuk O, Chen G,Mathew R, Jin S, White E, 2007, Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev 21:1621–1635 Leemans CR, Tiwari R, Nauta JJ, van derWaal I, Snow GB, 1994, Recurrence at the primary site in head and neck cancer and the significance of neck lymph node metastases as a prognostic factor. Cancer 73:187–190 Shen Y, Li DD, Wang LL, Deng R, Zhu XF, 2008, Decreased expression of autophagy-related proteins in malignant epithelial ovarian cancer. Autophagy 4:1067–1068 Karantza-Wadsworth V, White E, 2007, Role of autophagy in breast cancer. Autophagy 3:610–613 Shi YH, Ding ZB, Zhou J, Qiu SJ, Fan J, 2009, Prognostic significance of Beclin 1-dependent apoptotic activity in hepatocellular carcinoma. Autophagy 5:380–382 Cabanillas R, Rodrigo JP, Astudillo A, Dominguez F, Suarez C, Chiara MD, 2007, P53 expression in squamous cell carcinomas of the supraglottic larynx and its lymph node metastases: new results for an old question. Cancer 109:1791–1798 Leibel SA, Scott CB, Mohiuddin M, Marcial VA, Coia LR, Davis LW, Fuks Z, 1991, The effect of local-regional control on distant metastatic dissemination in carcinoma of the head and neck: results of an analysis from the RTOG head and neck database. Int J Radiat Oncol Biol Phys 21:549–556 Wan XB, Fan XJ, ChenMY, Xiang J, Huang PY, Guo L et al, 2010, Elevated Beclin 1 expression is correlated with HIF-1alpha in predicting poor prognosis of nasopharyngeal carcinoma. Autophagy 6:395–404 Koukourakis MI, Giatromanolaki A, Sivridis E, Pitiakoudis M, Gatter KC, Harris AL, 2010, Beclin 1 over-and underexpression in colorectal cancer: distinct patterns relate to prognosis and tumour hypoxia. Br J Cancer 103:1209–1214 Huang X, Bai HM, Chen L, Li B, Lu YC, 2010, Reduced expression of LC3B-II and Beclin 1 in glioblastoma multiforme indicates a down-regulated autophagic capacity that relates to the progression of astrocytic tumors. J Clin Neurosci 17:1515–1519 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 771 EP 777 DI 10.1007/s12253-013-9642-0 PG 7 ER PT J AU Tastemir-Korkmaz, D Demirhan, O Kuleci, S Hasturk, S AF Tastemir-Korkmaz, Deniz Demirhan, Osman Kuleci, Sedat Hasturk, Serap TI There is no Significant Association Between Death Receptor 4 (DR4) Gene Polymorphisms and Lung Cancer in Turkish Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Death receptor 4; Polymorphisms ID Lung cancer; Death receptor 4; Polymorphisms AB Death receptor 4 (DR4) gene is a candidate tumor supressor gene that has a role in apoptotic pathway. It was reported in literature that polymorphisms in DR4 gene lead to susceptibility to many cancers. In accordance with this information, we aimed to investigate the association between G422A, C626G, A683C and A1322G polymorphisms in DR4 gene and lung cancer. We selected 60 patients with lung cancer (LC) and 30 healthy, sex and age matched volunteers randomly. Four polymorhisms, G422A, C626G, A683C and A1322G, in DR4 gene were analyzed with Polymerase Change Reaction (PCR) – Restriction Fragment Lenght Polymorphism (RFLP) and Amplification Refractory Mutation System (ARMS) techniques in both groups. Our results showed that there are no statistically significances between the patients and controls in terms of the G422A, C626G, A683C and A1322G polymorphisms in DR4 gene (p>0,05). Our findings showed no role of DR4 gene polymorhisms in susceptibility to LC and provide a plausible explanation for DR4 genetic heterogeneity in LC susceptibility. C1 [Tastemir-Korkmaz, Deniz] Adiyaman University, Vocational School of Health Services, TR-02040 Adiyaman, Turkey. [Demirhan, Osman] Cukurova University, Faculty of Medicine, Department of Medical Biology and GeneticsAdana, Turkey. [Kuleci, Sedat] Cukurova University, Faculty of Medicine, Department of Chest DiseasesAdana, Turkey. [Hasturk, Serap] Acibadem Hospital, Department of Chest DiseasesAdana, Turkey. RP Tastemir-Korkmaz, D (reprint author), Adiyaman University, Vocational School of Health Services, TR-02040 Adiyaman, Turkey. EM deniz-tbio@hotmail.com CR Dechant MJ, Fellenberg J, Scheuerpflug CG, Ewerbeck V, Debatin K-M, 2004, Mutation Analysis of the apoptotic “death-receptors” and the adaptors TRADD and FADD/MORT-1 in osteosarcoma tumor samples and osteosarcoma cell lines. Int J Cancer 109:661– 667 Hazra A, Chamberlain RM, Grossman HB, Zhu Y, Spitz MR, Wu X, 2003, Death receptor 4 and bladder cancer risk. Cancer Res 63:1157–1159 Fisher MJ, Virmani AK, Wu L, Aplenc R, Harper JC, Powell SM, Rebbeck TR, Sidransky D, Gazdar AF, El-Deiry WS, 2001, Nucleotide substitution in the ectodomain of TRAIL receptor DR4 is associated with lung cancer and head and neck cancer. Clin Cancer Res 7:1688–1697 Martinez-Ferrandis JI, Rodriguez-Lopez R, Milne RL, Gonzalez E, Cebolla E, Chirivella I, Zamora P, Arias JI, Palacios S, Cervantes A, Diez O, Benitez J, Armengod ME, 2007, Polymorphisms in TRAIL receptor genes and risk of breast cancer in Spanish women. Cancer Biomark 3(2):89–93 Horak P, Pils D, Roessler M, Tomek S, Elandt K, Zeillinger R, Zielinski C, Krainer M, 2005, Common death receptor 4, DR4, polymorphisms do not predispose to ovarian cancer. Gynecol Oncol 97:514–518 Seitz S, Wassmuth P, Fischer J, Nothnagel A, Jandrig B, Schlag PM, Scherneck S, 2002, Mutation analysis and mRNA expression of trail-receptors in human breast cancer. Int J Cancer 102:117–128 Lee SH, Shin MS, Kim HS, Lee HK, Park WS, Kim SY, Lee JH, Han SY, Park JY, Oh RR, Kang CS, Kim KM, Jang JJ, Nam SW, Lee JY, Yoo NJ, 2001, Somatic mutations of TRAIL-receptor 1 and TRAIL-receptor 2 genes in non-Hodgkin’s lymphoma. Oncogene 20(3):399–403 Ulybina YM, Kuligina ES, Mitiushkina NV, Rozanov ME, Ivantsov AO, Ponomariova DN, Togo AV, Levchenko EV, Shutkin VA, Brenister SI, Devilee P, Zhivotovsky B, Hirvonen A, Imyanitov EN, 2009, Coding polymorphisms in Casp5, Casp8 and DR4 genes may play a role in predisposition to lung cancer. Cancer Lett 278(2):183–191 Wang M, Wang M, Cheng G, Zhang Z, Fu G, Zhang Z, 2009, Genetic variants in the death receptor 4 gene contribute to susceptibility to bladder cancer. Mutat Res 661:85–92 Frank B, Shanmugam KS, Beckmann L, Hemminki K, Brenner H, Hoffmeister M, Chang-Claude J, Burwinkel B, 2006, Death receptor 4 variants and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev 15(10):2002–2005 Lu G, Bing X, Qi S, Chun L, 2007, Association of DR4 gene polymorphism in Chinese patients with gastroduodenal diseases. Med J Wuhan Univ 28:93–95 Wolf S, Mertens D, Pscherer A, Schroeter P, Winkler D, Grone HJ, Hofele C, Hemminki K, Kumar R, SteineckG,Dohner H, Stilgenbauer S, Lichter P, 2006, Ala228 variant of trail receptor 1 affecting the ligand binding site is associated with chronic lymphocytic leukemia, mantle cell lumphoma, prostate cancer, head and neck squamous cell carcinoma and bladder cancer. Int J Cancer 118:1831–1835 Hymowitz SG, Christinger HW, Fuh G, Ultsch M, O’Connell M, Kelley RF, Ashkenazi A, de Vos AM, 1999, Triggering cell death: the crystal structure of Apo2L/TRAIL in a complex with death receptor 5. Mol Cell 4:563–571 Chen B, Liu S, Wang X-L, Xu W, Li Y, Zhao W-H, Wu, 2009, TRAIL-R1 polymorphisms and cancer susceptibility: an evidencebased meta-analysis. Eur J Cancer 45:2598–2605 Fernandez V, Jares P, Bea S, Salaverria I, Guino E, de Sanjose S, Colomer D, Ott G, Montserrat E, Campo E, 2004, Frequent polymorphic changes but not mutations of TRAIL receptors DR4 and DR5 in mantle cell lymphoma and other B-cell lymphoid neoplasms. Haematologica 89:1322–1331 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 779 EP 784 DI 10.1007/s12253-013-9643-z PG 6 ER PT J AU Juasook, A Aukkanimart, R Boonmars, Th Sudsarn, P Wonkchalee, N Laummaunwai, P Sriraj, P AF Juasook, Amornrat Aukkanimart, Ratchadawan Boonmars, Thidarut Sudsarn, Pakkayanee Wonkchalee, Nadchanan Laummaunwai, Porntip Sriraj, Pranee TI Tumor-Related Gene Changes in Immunosuppressive Syrian Hamster Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Immunosuppressive; Cholangiocarcinogenesis; Tumor-related gene; Immunohistochemistry ID Immunosuppressive; Cholangiocarcinogenesis; Tumor-related gene; Immunohistochemistry AB The results of a previous study demonstrated that prednisolone enhanced cholangiocarcinogenesis. Therefore, to clarify molecular changes during immunosuppressive cholangiocarcinogenesis, Syrian hamsters were divided into 8 groups: uninfected controls; immunosuppressed Syrian hamsters using prednisolone (P); normal Syrian hamsters administered N-nitrosodimethylamine (ND); immunosuppressed Syrian hamsters administered N-nitrosodimethylamine (NDis); normal Syrian hamsters infected with Opisthorchis viverrini (OV); immunosuppressed Syrian hamsters infected with O. viverrini (OVis); normal Syrian hamsters infected with O. viverrini and administered N-nitrosodimethylamine (CCA); and immunosuppressed Syrian hamsters infected with O. viverrini and administered N-nitrosodimethylamine (CCAis). Syrian hamster livers were used for analysis of tumor-related gene expression and immunohistochemistry through cytokeratin 19 (CK19) and proliferating cell nuclear antigen (PCNA) staining. The tumor-related gene expression results show that CCAis groups at all time points exhibited upregulation of COX-2, IL-6, SOD1, CAT and iNOS and downregulation of p53, which correlated with the predominant expression of CK19 and PCNA in liver tissue. These results suggest that prednisolone enhances cholangiocarcinoma development, which was confirmed by molecular changes. C1 [Juasook, Amornrat] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Aukkanimart, Ratchadawan] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Boonmars, Thidarut] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Sudsarn, Pakkayanee] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Wonkchalee, Nadchanan] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Laummaunwai, Porntip] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Sriraj, Pranee] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. RP Boonmars, Th (reprint author), Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. EM boonmars@yahoo.com;bthida@kku.ac.th CR Juasook A, Boonmars T,Wu Z, LoilomeW,VeteewuthacharnK,Namwat N, Sudsarn P, Wonkchalee O, Sriraj P, Aukkanimart R, 2013, Immunosuppressive prednisolone enhances early cholangiocarcinoma in Syrian hamsters with liver fluke infection and administration of Nnitrosodimethylamine. Pathol Oncol Res 19(1):55–62 Boonmars T, Srirach P, Kaewsamut B, Srisawangwong T, Pinlaor S, Pinlaor P, Yongvanit P, Sithithaworn P, 2008, Apoptosis-related gene expression in hamster opisthorchiasis post praziquantel treatment. Parasitol Res 102(3):447–455 Boonmars T, Wu Z, Boonjaruspinyo S, Pinlaor S, Nagano I, Takahashi Y, Kaewsamut B, Yongvanit P, 2009, Alterations of gene expression of RB pathway in Opisthorchis viverrini infection-induced cholangiocarcinoma. Parasitol Res 105(5):1273–1281 Boonmars T, Wu Z, Boonjaruspinyo S, Puapairoj A, Kaewsamut B, Nagano I, Pinlaor S, Yongvanit P,Wonkchalee O, Juasook A, Sudsarn P, Srisawangwong T, 2010, Involvement of c-Ski oncoprotein in carcinogenesis of cholangiocacinoma induced by Opisthorchis viverrini and N-nitrosodimethylamine. Pathol Oncol Res 17(2):219–227 Trune DR, Kempton JB, Gross ND, 2006, Mineralocorticoid receptor mediates glucocorticoid treatment effects in the autoimmune mouse ear. Hear Res 212(1–2):22–32 Boonjaraspinyo S, Boonmars T, Aromdee C, Puapairoj A, Wu Z, 2011, Indirect effect of a turmeric diet: enhanced bile duct proliferation in Syrian hamsters with a combination of partial obstruction by Opisthorchis viverrini infection and inflammation by Nnitrosodimethylamine administration. Parasitol Res 108(1):7–14 Laothong U, Pinlaor P, Hiraku Y, Boonsiri P, Prakobwong S, Khoontawad J, Pinlaor S, 2010, Protective effect of melatonin against Opisthorchis viverrini-induced oxidative and nitrosative DNA damage and liver injury in hamsters. J Pineal Res 49(3):271–282 Grivennikov SI, Greten FR, Karin M, 2010, Immunity, inflammation, and cancer. Cell 140(6):883–899 Barth H, Robinet E, Liang TJ, Baumert TF, 2008, Mouse models for the study of HCV infection and virus-host interactions. J Hepatol 49(1):134–142 Park EJ, Lee JH, Yu GY, He G, Ali SR, Holzer RG, Osterreicher CH, Takahashi H, Karin M, 2010, Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 140(2):197–208 IARC, 2011, A review of human carcinogens part B: biological agents, Opisthorchis viverrini and Clonorchis sinensis). IARC Monogr Eval Carcinog Risks Hum 100:347–376 Onizuka S, Tawara I, Shimizu J, Sakaguchi S, Fujita T, Nakayama E, 1999, Tumor rejection by in vivo administration of anti-CD25, interleukin-2 receptor alpha, monoclonal antibody. Cancer Res 59(13):3128–3133 El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, Herrera J, Lissowska J, Yuan CC, Rothman N, Lanyon G, Martin M, Fraumeni JF Jr, Rabkin CS, 2000, Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature 404(6776):398–402 de Martel C, Llosa AE, Farr SM, Friedman GD, Vogelman JH, Orentreich N, Corley DA, Parsonnet J, 2005, Helicobacter pylori infection and the risk of development of esophageal adenocarcinoma. J Infect Dis 191(5):761–767 Zamarron BF, ChenW, 2011, Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci 7(5):651–658 Pinlaor S, Hiraku Y,Ma N, Yongvanit P, Semba R, Oikawa S, Murata M, Sripa B, Sithithaworn P, Kawanishi S, 2004, Mechanism of NOmediated oxidative and nitrative DNA damage in hamsters infected with Opisthorchis viverrini: a model of inflammation-mediated carcinogenesis. Nitric Oxide 11(2):175–183 Pinlaor S,MaN,HirakuY,Yongvanit P, Semba R, Oikawa S, Murata M, Sripa B, Sithithaworn P, Kawanishi S, 2004, Repeated infection with Opisthorchis viverrini induces accumulation of 8-nitroguanine and 8- oxo-7,8-dihydro-2′-deoxyguanine in the bile duct of hamsters via inducible nitric oxide synthase. Carcinogenesis 25(8):1535–1542 Wu Z, Boonmars T, Boonjaraspinyo S, Nagano I, Pinlaor S, Puapairoj A, Yongvanit P, Takahashi Y, 2011, Candidate genes involving in tumorigenesis of cholangiocarcinoma induced by Opisthorchis viverrini infection. Parasitol Res 109(3):657–673 Loilome W, Yongvanit P, Wongkham C, Tepsiri N, Sripa B, Sithithaworn P, Hanai S, Miwa M, 2006, Altered gene expression in Opisthorchis viverrini-associated cholangiocarcinoma in hamster model. Mol Carcinog 45(5):279–287 Howe LR, Subbaramaiah K, Brown AM, Dannenberg AJ, 2001, Cyclooxygenase-2: a target for the prevention and treatment of breast cancer. Endocr Relat Cancer 8(2):97–114 Lakatos PL, Lakatos L, 2008, Risk for colorectal cancer in ulcerative colitis: changes, causes and management strategies. World J Gastroenterol 14(25):3937–3947 Dolcet X, Llobet D, Pallares J, Matias-Guiu X, 2005, NF-kB in development and progression of human cancer. Virchows Arch 446(5):475–482 Sakamoto K, Maeda S, Hikiba Y, Nakagawa H, Hayakawa Y, Shibata W, Yanai A, Ogura K, Omata M, 2009, Constitutive NF-kappaB activation in colorectal carcinoma plays a key role in angiogenesis, promoting tumor growth. Clin Cancer Res 15(7):2248–2258 Fernandez-Majada V, Aguilera C, Villanueva A, Vilardell F, Robert-Moreno A, Aytes A, Real FX, Capella G, Mayo MW, Espinosa L, Bigas A, 2007, Nuclear IKK activity leads to dysregulated notch-dependent gene expression in colorectal cancer. Proc Natl Acad Sci U S A 104(1):276–281 Leong KG, Karsan A, 2006, Recent insights into the role of Notch signaling in tumorigenesis. Blood 107(6):2223–2233 Koga H, Sakisaka S, Ohishi M, Kawaguchi T, Taniguchi E, Sasatomi K, Harada M, Kusaba T, Tanaka M, Kimura R, Nakashima Y, Nakashima O, Kojiro M, Kurohiji T, Sata M, 1999, Expression of cyclooxygenase-2 in human hepatocellular carcinoma: relevance to tumor dedifferentiation. Hepatology 29(3):688–696 Martin-Sanz P,Mayoral R, Casado M, Bosca L, 2010, COX-2 in liver, from regeneration to hepatocarcinogenesis: what we have learned from animal models? World J Gastroenterol 16(12):1430–1435 Kundu JK, Surh YJ, 2008, Inflammation: gearing the journey to cancer. Mutat Res 659(1–2):15–30 Burstein E, Fearon ER, 2008, Colitis and cancer: a tale of inflammatory cells and their cytokines. J Clin Invest 118(2):464–467 McCord JM, Fridovich I, 1969, Superoxide dismutase. An enzymic function for erythrocuprein, hemocuprein). J Biol Chem 244(22):6049–6055 Cobbs CS, Levi DS, Aldape K, Israel MA, 1996, Manganese superoxide dismutase expression in human central nervous system tumors. Cancer Res 56(14):3192–3195 Izutani R, Asano S, Imano M, Kuroda D, Kato M, Ohyanagi H, 1998, Expression of manganese superoxide dismutase in esophageal and gastric cancers. J Gastroenterol 33(6):816–822 Toh Y, Kuninaka S, Oshiro T, Ikeda Y, Nakashima H, Baba H, Kohnoe S, Okamura T, Mori M, Sugimachi K, 2000, Overexpression of manganese superoxide dismutase mRNA may correlate with aggressiveness in gastric and colorectal adenocarcinomas. Int J Oncol 17(1):107–112 Thomsen LL, Miles DW, Happerfield L, Bobrow LG, Knowles RG, Moncada S, 1995, Nitric oxide synthase activity in human breast cancer. Br J Cancer 72(1):41–44 Cobbs CS, Brenman JE, Aldape KD, Bredt DS, Israel MA, 1995, Expression of nitric oxide synthase in human central nervous system tumors. Cancer Res 55(4):727–730 Masri FA, Comhair SA, Koeck T, Xu W, Janocha A, Ghosh S, Dweik RA, Golish J, Kinter M, Stuehr DJ, Erzurum SC, Aulak KS, 2005, Abnormalities in nitric oxide and its derivatives in lung cancer. Am J Respir Crit Care Med 172(5):597–605 Klotz T, Bloch W, Volberg C, Engelmann U, Addicks K, 1998, Selective expression of inducible nitric oxide synthase in human prostate carcinoma. Cancer 82(10):1897–1903 Lagares-Garcia JA, Moore RA, Collier B, Heggere M, Diaz F, Qian F, 2001, Nitric oxide synthase as a marker in colorectal carcinoma. Am Surg 67(7):709–713 Hajri A, Metzger E, Vallat F, Coffy S, Flatter E, Evrard S, Marescaux J, Aprahamian M, 1998, Role of nitric oxide in pancreatic tumour growth: in vivo and in vitro studies. Br J Cancer 78(7):841–849 Ekmekcioglu S, Ellerhorst J, Smid CM, Prieto VG, Munsell M, Buzaid AC, Grimm EA, 2000, Inducible nitric oxide synthase and nitrotyrosine in human metastatic melanoma tumors correlate with poor survival. Clin Cancer Res 6(12):4768–4775 Calvisi DF, Pinna F, Ladu S, Pellegrino R, MuroniMR, Simile MM, Frau M, Tomasi ML, De Miglio MR, Seddaiu MA, Daino L, Sanna V, Feo F, Pascale RM, 2008, Aberrant iNOS signaling is under genetic control in rodent liver cancer and potentially prognostic for the human disease. Carcinogenesis 29(8):1639–1647 Cahlin C, Gelin J, Delbro D, Lonnroth C, Doi C, Lundholm K, 2000, Effect of cyclooxygenase and nitric oxide synthase inhibitors on tumor growth in mouse tumor models with and without cancer cachexia related to prostanoids. Cancer Res 60(6):1742–1749 van der Woude CJ, Kleibeuker JH, Jansen PL, Moshage H, 2004, Chronic inflammation, apoptosis and, pre-)malignant lesions in the gastro-intestinal tract. Apoptosis 9(2):123–130 Mates JM(2000, Effects of antioxidant enzymes in the molecular control of reactive oxygen species toxicology. Toxicology 153(1–3):83–104 Marra M, Sordelli IM, Lombardi A, Lamberti M, Tarantino L, Giudice A, Stiuso P, Abbruzzese A, Sperlongano R, Accardo M, Agresti M, Caraglia M, Sperlongano P, 2011, Molecular targets and oxidative stress biomarkers in hepatocellular carcinoma: an overview. J Transl Med., DOI 10.1186/1479-5876-9-171 Min JY, Lim SO, Jung G, 2010, Downregulation of catalase by reactive oxygen species via hypermethylation of CpG island II on the catalase promoter. FEBS Lett 584(11):2427–2432 Sen S, Kawahara B, Chaudhuri G, 2012, Maintenance of higher H2O2 levels, and its mechanism of action to induce growth in breast cancer cells: important roles of bioactive catalase and PP2A. Free Radic Biol Med 53(8):1541–1551 Alawadi S, Ghabreau L, Alsaleh M, Abdulaziz Z, Rafeek M, Akil N, Alkhalaf M, 2011, P53 gene polymorphisms and breast cancer risk in Arab women. Med Oncol 28(3):709–715 Hollstein M, Sidransky D, Vogelstein B, Harris CC, 1991, p53 mutations in human cancers. Science 253(5015):49–53 O’Dell MR, Huang JL, Whitney-Miller CL, Deshpande V, Rothberg P, Grose V, Rossi RM, Zhu AX, Land H, Bardeesy N, Hezel AF, 2012, Kras(G12D, and p53 mutation cause primary intrahepatic cholangiocarcinoma. Cancer Res 72(6):1557–1567 Kishimoto T, Akira S, Narazaki M, Taga T, 1995, Interleukin-6 family of cytokines and gp130. Blood 86(4):1243–1254 Chung YC, Chang YF, 2003, Serum interleukin-6 levels reflect the disease status of colorectal cancer. J Surg Oncol 83(4):222–226 Schneider MR, Hoeflich A, Fischer JR, Wolf E, Sordat B, Lahm H, 2000, Interleukin-6 stimulates clonogenic growth of primary and metastatic human colon carcinoma cells. Cancer Lett 151(1):31–38 Park J, Tadlock L, Gores GJ, Patel T, 1999, Inhibition of interleukin 6-mediated mitogen-activated protein kinase activation attenuates growth of a cholangiocarcinoma cell line. Hepatology 30(5):1128–1133 Meng F, Yamagiwa Y, Ueno Y, Patel T, 2006, Over-expression of interleukin-6 enhances cell survival and transformed cell growth in human malignant cholangiocytes. J Hepatol 44(6):1055–1065 Frampton G, Invernizzi P, Bernuzzi F, Pae HY, Quinn M, Horvat D, Galindo C, Huang L, McMillin M, Cooper B, Rimassa L, DeMorrow S, 2012, Interleukin-6-driven progranulin expression increases cholangiocarcinoma growth by an Akt-dependent mechanism. Gut 61(2):268–277 Alvaro D, 2009, Serum and bile biomarkers for cholangiocarcinoma. Curr Opin Gastroenterol 25(3):279–284 Okada K, Shimizu Y, Nambu S, Higuchi K, Watanabe A, 1994, Interleukin-6 functions as an autocrine growth factor in a cholangiocarcinoma cell line. J Gastroenterol Hepatol 9(5):462–467 Sugawara H, Yasoshima M, Katayanagi K, Kono N, Watanabe Y, Harada K, Nakanuma Y, 1998, Relationship between interleukin-6 and proliferation and differentiation in cholangiocarcinoma. Histopathology 33(2):145–153 Sripa B, Thinkhamrop B, Mairiang E, Laha T, Kaewkes S, Sithithaworn P, Periago MV, Bhudhisawasdi V, Yonglitthipagon P, Mulvenna J, Brindley PJ, Loukas A, Bethony JM, 2012, Elevated plasma IL-6 associates with increased risk of advanced fibrosis and cholangiocarcinoma in individuals infected by Opisthorchis viverrini. PLoS Negl Trop Dis 6(5):e1654 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 785 EP 794 DI 10.1007/s12253-013-9645-x PG 10 ER PT J AU Yanamoto, S Yamada, Shi Takahashi, H Kawasaki, G Ikeda, H Shiraishi, T Fujita, Sh Ikeda, T Asahina, I Umeda, M AF Yanamoto, Souichi Yamada, Shin-ichi Takahashi, Hidenori Kawasaki, Goro Ikeda, Hisazumi Shiraishi, Takeshi Fujita, Shuichi Ikeda, Tohru Asahina, Izumi Umeda, Masahiro TI Predictors of Locoregional Recurrence in T1-2N0 Tongue Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tongue cancer; Recurrence; Neoadjuvant chemotherapy; Surgical margin ID Tongue cancer; Recurrence; Neoadjuvant chemotherapy; Surgical margin AB Locoregional recurrence of oral tongue squamous cell carcinoma (OTSCC) has been considered a poor prognostic entity in terms of survival rate. The purpose of this study was to evaluate the incidence of locoregional recurrence and to identify significant risk factors for locoregional recurrence in early-stage OTSCC. We retrospectively reviewed the records of 58 patients who underwent radical surgery for T1-2N0 OTSCC. The local recurrence and regional recurrence rates were 10.3 % (6/58 patients) and 15.5 % (9/58 patients) in this study, respectively. The survival rate of patients with local recurrence was 66.7 %, which was significantly lower than that (96.2 %) of patients without local recurrence, whereas the survival rates of patients with or without regional recurrence were not significantly difference. Pattern of invasion (POI), neoadjuvant chemotherapy (NAC) and the status of the surgical margin were identified as factors influencing local recurrence. In particular, the status of the deep surgical margin was a high potential independent risk factor. The deep surgical margin was resected closely in many NAC-treated cases, suggesting that NAC may lead to local recurrence and a poor outcome. No efficacy of NAC was observed, suggesting that the standard treatment for early OTSCC is surgery alone. C1 [Yanamoto, Souichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yamada, Shin-ichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Takahashi, Hidenori] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Kawasaki, Goro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Ikeda, Hisazumi] Nagasaki University Graduate School of Biomedical Sciences, Department of Regenerative Oral Surgery, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Shiraishi, Takeshi] Nagasaki University Graduate School of Biomedical Sciences, Department of Regenerative Oral Surgery, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Fujita, Shuichi] Nagasaki University Graduate School of Biomedical Sciences, Department of Oral Pathology and Bone Metabolism, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Ikeda, Tohru] Nagasaki University Graduate School of Biomedical Sciences, Department of Oral Pathology and Bone Metabolism, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Asahina, Izumi] Nagasaki University Graduate School of Biomedical Sciences, Department of Regenerative Oral Surgery, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Umeda, Masahiro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. RP Yanamoto, S (reprint author), Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 852-8588 Nagasaki, Japan. EM syana@nagasaki-u.ac.jp CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global Cancer Statistics 2011. CA Cancer J Clin 61:69–90 Warnakulasuriya S, 2009, Global epidemiology of oral and oropharyngeal cancer. Oral Oncol 45:309–316 National Cancer Institute, 2012). Surveillance, Epidemiology, and End Results, SEER, web site. http://seer.cancer.gov/statistics/. Accessed 15 March 2012 Rusthoven KE, Raben D, Song JI, Kane M, Altoos TA, Chen C, 2010, Survival and patterns of relapse in patients with oral tongue cancer. J Oral Maxillofac Surg 68:584–589 Ganly I, Patel S, Shah J, 2012, Early stage squamous cell cancer of the oral tongue-clinicopathologic features affecting outcome. Cancer 118:101–111 Spiro RH, Guillamondegui O, Paulino AF, Huvos AG, 1999, Pattern of invasion and margin assessment in patients with oral tongue cancer. Head Neck 21:408–413 Goto M, Hasegawa Y, Terada A, Hyodo I, Hanai N, Ijichi K, Yamada H, Fujimoto Y, Ogawa T, 2005, Prognostic significance of late cervical metastasis and distant failure in patients with stage I and II oral tongue cancers. Oral Oncol 41:62–69 Brandwein-Gensler M, Teixeira MS, Lewis CM, Lee B, Rolnitzky L, Hille JJ, Genden E, Urken ML, Wang BY, 2005, Oral squamous cell carcinoma. Histologic risk assessment, but not margin status, is strongly predictive of local disease-free and overall survival. Am J Surg Pathol 29:167–178 Pentenero M, Gandolfo S, Carrozzo M, 2005, Importance of tumor thickness and depth of invasion in nodal involvement and prognosis of oral squamous cell carcinoma a review of the literature. Head Neck 27:1080–1091 Vered M, Dayan D, Dobriyan A, Yahalom R, Shalmon B, Barshack I, Bedrin L, Talmi YP, Taicher S, 2010, Oral tongue squamous cell carcinoma: recurrent disease is associated with histopathologic risk score and young age. J Cancer Res Clin Oncol 136:1039–1048 El-Naaj IA, Leiser Y, Shveis M, Sabo E, Peled M, 2011, Incidence of oral cancer occult metastasis and survival of T1-T2N0 oral cancer patients. J Oral Maxillofac Surg 69:2674–2679 Shah JP, Gil Z, 2009, Current concepts in management of oral cancer. Oral Oncol 45:394–401 Brennan S, Corry J, Kleid S, Porceddu S, Yuen K, Rischin D, Peters LJ, 2010, Prospective trial to evaluate staged neck dissection or elective neck radiotherapy in patients with ct-staged T1-2 N0 squamous cell carcinoma of the oral tongue. Head Neck 32:191–198 Nag S, Cano ER, Demanes J, Puthawala AA, Vikram B, 2001, The American brachytherapy society recommendations for high-doserate brachytherapy for head-and-neck carcinoma. Int J Radia Oncol Biol Phys 50:1190–1198 Mazeron JJ, Ardiet JM, Haie-Meder C, Kovacs G, Levendag P, Peiffert D, Polo A, Rovirosa A, Strnad V, 2009, GEC-ESTRO recommendations for brachytherapy for head and neck squamous cell carcinomas. Radiother Oncol 91:150–156 Umeda M, Komatsubara H, Ojima Y, Minamikawa T, Shibuya Y, Yokoo S, Ishii J, Komori T, 2005, A comparison of brachytherapy and surgery for the treatment of stage I-II squamous cell carcinoma of the tongue. Int J Oral Maxillofac Surg 34:739–744 Sobin LH, Wittekind C, 2002, UICC TNM classification of malignant tumours, 6th edn. Wiley, New York Pindborg JJ, Reichart PA, Smith CJ, van der Waal I, eds,, 1997, World Health Organization histological typing of cancer and precancer of the oral mucosa, 2nd edn. Springer, New York Bryne M, Boysen M, Alfsen CG, Abeler VM, Sudbo J, Nesland JM, Kristensen GB, Piffko J, Bankfalvi A, 1998, The invasive front of carcinomas. The most important area for tumor prognosis? Anticancer Res 18:4757–4764 Asakage T, Yokose T, Mukai K, Tsugane S, Tsubono Y, Asai M, Ebihara S, 1998, Tumor thickness predicts cervical metastasis in patients with stage I/II carcinoma of the tongue. Cancer 82:1443–1448 Kurokawa H, Yamashita Y, Takeda S, Zhang M, Fukuyama H, Takahashi T, 2002, Risk factors for late cervical lymph node metastases in patients with stage I or II carcinoma of the tongue. Head Neck 24:731–736 Lim SC, Zhang S, Ishii G, Endoh Y, Kodama K, Miyamoto S, Hayashi R, Ebihara S, Cho JS, Ochiai A, 2004, Predictive markers for late cervical metastasis in stage I and II invasive squamous cell carcinoma of the oral tongue. Clin Cancer Res 10:166–172 Sparano A, Weinstein G, Chalian A, Yodul M, Weber R, 2004, Multivariate predictors of occult neck metastasis in early oral tongue cancer. Otolaryngol Head Neck Surg 131:472–476 Huang SH, Hwang D, Lockwood G, Goldstein DP, O’Sullivan B, 2009, Predictive value of tumor thickness for cervical lymphnode involvement in squamous cell carcinoma of the oral cavity. A meta-analysis of reported studies. Cancer 115:1489–1497 Fan KH, Wang HM, Kang CJ, Lee LY, Huang SF, Lin CY, Chen EYC, Chen IH, Liao CT, Chang JTC, 2010, Treatment results of postoperative radiotherapy on squamous cell carcinoma of the oral cavity: coexistence of multiple minor risk factors results in higher recurrence rates. Int J Radiation Oncology Biol Phys 77:1024–1029 Kernohan MD, Clark JR, Gao K, Ebrahimi A, Milross CG, 2010, Predicting the prognosis of oral squamous cell carcinoma after first recurrence. Arch Otolaryngol Head Neck Surg 136:1235–1239 Huang TY, Hsu LP, Wen YH, Huang TT, Chou YF, Lee CF, Yang MC, Chang YK, Chen PR, 2010, Predictors of locoregional recurrence in early stage oral cavity cancer with free surgical margins. Oral Oncol 45:49–55 O-charoenrat PO, Pillai G, Patel S, Fisher C, Archer D, Eccles S, Rhys-Evans P, 2003, Tumour thickness predicts cervical nodal metastases and survival in early oral tongue cancer. Oral Oncol 39:386–390 Al-Rajhi N, Khafaga Y, El-Husseiny J, Saleem M, Mourad W, Al- Otieschan A, Al-Amro A, 2000, Early stage carcinoma of oral tongue: prognostic factors for local control and survival. Oral Oncol 36:508–514 Yuen APW, Lam KY, Lam LK, Ho CM, Wong A, Chow TL, Yuen WF, Wei WI, 2002, Prognostic factors of clinically stage I and II oral tongue carcinoma-a comparative study of stage, thickness, shape, growth pattern, invasive front malignancy grading, Martines-Gimen score, and pathologic features. Head Neck 24:513–520 Keski-Santti H, Atula T, Tikka J, Hollmen J, Makitie AA, Leivo I, 2007, Predictive value of histopathologic parameters in early squamous cell carcinoma of oral tongue. Oral Oncol 43:1007–1013 Pignon JP, Bourhis J, Domenge C, Designe L, 2000, Chemotherapy added to locoregional treatment for head and neck squamouscell carcinoma: three meta-analyses of updated individual data. Lancet 355:949–955 Kirita T, Ohgi K, Kawakami M, Miyawaki S, Okamoto M, Yamanaka Y, Sugimura M, 2002, Primary tumour resection of tongue carcinoma based on response to preoperative therapy. Int J Oral Maxillofac Surg 31:267–272 Licitra L, Grandi C, Guzzo M, Mariani L, Lo Vullo S, Valvo F, Quattrone P, Valagussa P, Bonadonna G,Molinari R, Cantu G, 2003, Primary chemotherapy in resectable oral cavity squamous cell cancer: a randomized controlled trial. J Clin Oncol 21:327–333 Olasz L, Szalma J, Orsi E, Tornoczky T, Marko T, Nyarady Z, 2010, Neoadjuvant chemotherapy: does it have benefits for surgeon in the treatment of advanced squamous cell cancer of the oral cavity? Pathol Oncol Res 16:207–212 Lore JM, Diaz-Ordaz E, Spaulding M, Chary K, Kaufman S, Lawrence W, Hong F, Gerold T, Sundquist N, Barrali RA, 1995, Improved survival with preoperative chemotherapy followed by tumor response for advanced squamous cell carcinoma of the head and neck. Am J Surg 170:506–511 Okura M, Hiranuma T, Adachi T, Ogura T, Aikawa T, Yoshioka H, Hayashido Y, Kogo M, Matsuya T, 1998, Induction chemotherapy is associated with an increase in the incidence of locoregional recurrence in patients with carcinoma of the oral cavity: results from a single institution. Cancer 82:804–815 Bernier J, Domenge C, Ozsahin M, Matuszewska K, Lefebvre JL, Greiner RH, Giralt J, Maingon P, Rolland F, Michel B, Cognetti F, Bourhis J, Kirkpatrick A, van Glabbeke M, 2004, Postoperative irradiation with or without concomitant chemotherapy for locally advanced head and neck cancer. N Eng J Med 350:1945–1952 Kurita H, Nakanishi Y, Nishizawa R, Xiao T, Kamata T, Koike T, Kobayashi H, 2010, Impact of different surgical margin conditions on local recurrence of oral squamous cell carcinoma. Oral Oncol 46:814–817 Sutton DN, Brown JS, Rogers SN, Vaughan ED, Woolgar JA, 2003, The prognostic implications of the surgical margin in oral squamous cell carcinoma. Int J Oral Maxillofac Surg 32:30–34 Upile T, Fisher C, Jerjes W, El Maaytah M, Searle A, Archer D, Michaels L, Rhys-Evans P, Hopper C, Howard D,Wright A, 2007, The uncertainty of the surgical margin in the treatment of head and neck cancer. Oral Oncol 43:321–326 Umeda M, Shigeta T, Takahashi H, Minamikawa T, Komatsubara H, Oguni A, Shibuya Y, Komori T, 2011, Cinical evaluation of Lugol’s iodine staining in the treatment of stage I-II squamous cell carcinoma of the tongue. Int J Oral Maxillofac Surg 40:593–596 Kim JJ, Tannock IF, 2005, Repopulation of cancer cells during therapy: an important cause of treatment failure. Nat Rev Cancer 5:516–525 Bourhis J, Wilson G, Wibault P, Janot F, Bosq J, Armand JP, Luboinski B, Malaise EP, Eschwege F, 1994, Rapid tumor cell proliferation after induction chemotherapy in oropharyngeal cancer. Laryngoscope 104:468–472 Penel N, Fournier C, Lefebvre D, Roussel-Delvallez M, Sarini J, Kar A, Mallet Y, Lefebvre JL, 2004, Previous chemotherapy as a predictor of wound infections in nonmajor head and neck surgery: results of a prospective study. Head Neck 26:513–517 Penel N, Fournier C, Lefebvre D, Lefebvre JL, 2005, Multivariate analysis of risk factors for wound infection in head and neck squamous cell carcinoma surgery with opening of mucosa. Study of 260 surgical procedures. Oral Oncol 41:294–303 Rahima B, Shingaki S, Nagata M, Saito C, 2004, Prognostic significance of perineural invasion in oral and oropharyngeal carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 97:423–431 Gonzalez-Garcia R, Naval-Gias L, Roman-Romero L, Sastre-Perez J, Rodriguez-Campo FJ, 2009, Local recurrences and second primary tumors from squamous cell carcinoma of the oral cavity: a retrospective analytic study of 500 patients. Head Neck 31:1168– 1180 Ord RA, Kolokythas A, Reynolds MA, 2006, Surgical salvage for local and regional recurrence in oral cancer. J Oral Maxillofac Surg 64:1409–1414 Schwartz GJ, Mehta RH, Wenig BL, Shaligram C, Portugal LG, 2000, Salvage treatment for recurrent squamous cell carcinoma of the oral cavity. Head Neck 22:34–41 Sklenicka S, Gardiner S, Dierks EJ, Potter BE, Bell RB, 2010, Survival analysis and risk factors for recurrence in oral squamous cell carcinoma: Does surgical salvage affect outcome? J Oral Maxillofac Surg 68:1270–1275 Kaya S, Yilmaz T, Gursel B, Sarac S, Sennaroglu L, 2001, The value of elective neck dissection in treatment of cancer of the tongue. Am J Otolaryngol 22:59–64 Lim YC, Lee JS, Koo BS, Kim SH, Kim YH, Choi EC, 2006, Treatment of contralateral N0 neck in early squamous cell carcinoma of the oral tongue: elective neck dissection versus observation. Laryngoscope 116:461–465 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 795 EP 803 DI 10.1007/s12253-013-9646-9 PG 8 ER PT J AU Toth, V Hatvani, Zs Somlai, B Harsing, J Laszlo, FJ Karpati, S AF Toth, Veronika Hatvani, Zsofia Somlai, Beata Harsing, Judit Laszlo, F Janos Karpati, Sarolta TI Risk of Subsequent Primary Tumor Development in Melanoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Melanoma; Survivor; Cancer risk; Subsequent primary tumor ID Melanoma; Survivor; Cancer risk; Subsequent primary tumor AB Incidence of subsequent malignant tumor development in 740 patients with primary cutaneous melanoma verified between 2006 and 2010 at the Semmelweis University was studied retrospectively and was compared to data of sex and age matched Hungarian population. The follow-up period was 1499 personyears for the whole group from the diagnosis of index melanoma with an average of 2 years. Standardized incidence rate (SIR) was established as the ratio of observed and expected values. The risk of all subsequent malignancies was 15- and 10-fold higher in males (SIR: 15.42) and in females (SIR: 10.55) with melanoma, than in the general population. The increased cancer risk resulted mainly from the significantly higher skin tumor development: SIR values were 160.39 and 92.64 for additional invasive melanoma and 342.28 and 77.04 for subsequent in situ melanoma in males and females, respectively. Non-melanoma skin cancers also notably contributed to the higher risk, the SIR was elevated in both genders to the same extent (males: 17.12, females: 17.55). The risk was also significantly higher for extracutaneous tumor development like chronic lymphocytic leukemia, colon and kidney cancer (both genders), non-Hodgkin’s lymphoma, cervical cancer (females), and bladder carcinoma (males). These data underline the importance of patient education and the necessity of frequent medical follow up, including a close-up dermatological screening of melanoma survivors for further malignancies. C1 [Toth, Veronika] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Hatvani, Zsofia] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Somlai, Beata] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Harsing, Judit] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Laszlo, F Janos] University of Debrecen, Department of Computer Science, Kassai street 26, 4028 Debrecen, Hungary. [Karpati, Sarolta] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. RP Toth, V (reprint author), Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, 1085 Budapest, Hungary. EM toveroka@yahoo.com CR Wassberg C, Thorn M, Yuen J, Ringborg U, Hakulinen T, 1996, Second primary cancers in patients with cutaneous malignant melanoma: a population-based study in Sweden. Br J Cancer 73:255–259 Levi F, LaVecchia C, Randimbison L, Te VC, Erler G, 1997, Incidence of invasive cancers following cutaneous malignant melanoma. Int J Cancer Suppl 72:776–779 Wassberg C, Thorn M, Yuen J, Hakulinen T, Ringborg U, 1999, Cancer risk in patients with earlier diagnosis of cutaneous melanoma in situ. Int J Cancer 83:314–317 Kroumpouzos G, Konstadoulakis MM, Cabral H, Karakousis CP, 2000, Risk of basal cell and squamous cell carcinoma in persons with prior cutaneous melanoma. Dermatol Surg 26:547–550 Goggins WB, Finkelstein DM, Tsao H, 2001, Evidence for an association between cutaneous melanoma and non-Hodgkin lymphoma. Cancer 91:874–880 Schmid-Wendtner MH, Baumert J, Wendtner CM, Plewig G, Volkenandt M, 2001, Risk of second primary malignancies in patients with cutaneous melanoma. Br J Dermatol 145:981–985 Crocetti E, Carli P, 2004, Risk of second primary cancers, other than melanoma, in an Italian population-based cohort of cutaneous malignant melanoma patients. Eur J Cancer Prev 13:33–37 Lens MB, Newton-Bishop JA, 2005, An association between cutaneous melanoma and non-Hodgkin’s lymphoma: pooled analysis of published data with a review. Ann Oncol 16:460–465 Wu YH, Kim GH, Wagner JD, Hood AF, Chuang TY, 2006, The association between malignant melanoma and noncutaneous malignancies. Int J Dermatol 45:529–534 Freedman DM, Miller BA, Tucker MA, 2006, New malignancies following melanoma of skin, eye melanoma, and nonmelanoma eye cancer. In: Curtis RE, Freedman DM, Ron E et al, eds, New malignancies among cancer survivors: SEER cancer registries, 1973-2000. National Cancer Institute, Bethesda Crocetti E, Guzzinati S, Paci E, Falcini F, Zanetti R, Vercelli M et al, 2008, The risk of developing a second, different, cancer among 14 560 survivors of malignant cutaneous melanoma: a study by AIRTUM, the Italian Network of Cancer Registries). Melanoma Res 18:230–234 Cantwell MM, Murray LJ, Catney D, Donnelly D, Autier P, Boniol M et al, 2009, Second primary cancers in patients with skin cancer: a population-based study in Northern Ireland. Br J Cancer 100:174–177 Spanogle JP, Clarke CA, Aroner S, Swetter SM, 2010, Risk of second primary malignancies following cutaneous melanoma diagnosis: a population-based study. J Am Acad Dermatol 62:757–767 Bradford PT, Freedman DM, Goldstein AM, Tucker MA, 2010, Increased risk of second primary cancers after a diagnosis of melanoma. Arch Dermatol 146:265–272 Balamurugan A, Rees JR, Kosary C, Rim SH, Li J, Stewart SL, 2011, Subsequent primary cancers among men and women with in situ and invasive melanoma of the skin. J Am Acad Dermatol 65(5 Suppl 1):69–77 Bhatia S, Estrada-Batres L, Maryon T, Bogue M, Chu D, 1999, Second primary tumors in patients with cutaneous malignant melanoma. Cancer 86:2014–2020 Wolff J, Wollina U, 2000, Second malignancies in melanoma patients in Thuringia. J Eur Acad Dermatol Venereol 14:479–483 Manganoni AM, Pavoni L, Farisoglio C, Sereni E, Chiudinelli M, Calzavara-Pinton P, 2012, Association between multiple cutaneous melanoma and other primary neoplasms. Clin Exp Dermatol 37:857– 861 National Cancer Registry, 2001-2010, National Institute of Oncology http://www.honcology.hu/portal/page/portal/OOI/MEDICAL_ ATTENDANCE/medical_departments/MD_901/Statisztika Rothman KJ, Boice JD, 1979, Epidemiologic analysis with a programmable calculator, NIH publication 79-1649). US Government Printing Office, Washington DC Pearson K, 1900, On the criterion that a given system of deviations from the probable in the case of a correlated system of variables is such that it can be reasonably supposed to have arisen from random sampling. Philos Mag 50:157–175 Yates F, 1934, Contingency table involving small numbers and the χ2 test. Suppl J R Stat Soc 1:217–235 Ward KA, Lazovich D, Hordinsky MK, 2012, Germline melanoma susceptibility and prognostic genes: a review of the literature. J Am Acad Dermatol 67:1055–1067 Read J, 2013, Recent advances in cutaneous melanoma: towards a molecular model and targeted treatment. Australas J Dermatol., DOI 10.1111/ajd.12013 Boussemart L, Routier E, Mateus C, Opletalova K, Sebille G, Kamsu-Kom N et al, 2013, Prospective study of cutaneous sideeffects associated with the BRAF inhibitor vemurafenib: a study of 42 patients. Ann Oncol 2013 Feb 13. [Epub ahead of print] Su F, Viros A, Milagre C, Trunzer K, Bollag G, Spleiss O et al, 2012, RAS mutations in cutaneous squamous-cell carcionomas in patients treated with BRAF inhibitors. N Engl J Med 366:207–215 Chapman PB et al, 2012, Development of colonic adenomas and gastric polyps in BRAF mutant melanoma patients treated with vemurafenib. Pigment Cell Melanoma Res 25:847 Callahan MK, Rampal R, Harding JJ, Klimek VM, Chung YR, Merghoub T et al, 2012, Progression of RAS-mutant leukemia during RAF inhibitor treatment. N Engl J Med 367:2316–2321 Smalley KS, Sondak VK, 2013, Targeted therapy for melanoma: is double hitting a home run? Nat Rev Clin Oncol 10:5–6 Grant WB, 2012, Ultraviolet exposure and non-Hodgkin’s lymphoma: beneficial and adverse effects? Cancer Causes Control 23:653–655 Bertrand KA, Chang ET, Abel GA, Zhang SM, Spiegelman D, Qureshi AA et al, 2011, Sunlight exposure, vitamin D, and risk of non-Hodgkin lymphoma in the Nurses’ Health Study. Cancer Causes Control 22:1731–1741 Grant WB, 2012, Update on evidence that support a role of solar ultraviolet-B irradiance in reducing cancer risk. Anticancer Agents Med Chem 13:140–146 Grant WB, 2002, An ecologic study of dietary and solar ultraviolet-B links to breast carcinoma mortality rates. Cancer 94:272–281 Grant WB, Garland CF, 2002, The association of solar ultraviolet- B, UVB, with reducing risk of cancer: multifactorial ecologic analysis of geographic variation in age-adjusted cancer mortality rates. Anticancer Res 26:2687–2699 Boscoe FP, Schymura MJ, 2006, Solar ultraviolet-B exposure and cancer incidence and mortality in the United States, 1993–2002. BMC Cancer 6:264 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 805 EP 810 DI 10.1007/s12253-013-9647-8 PG 6 ER PT J AU Yang, Q Du, J Zu, L AF Yang, Qing Du, Jun Zu, Lingling TI Overexpression of CD73 in Prostate Cancer is Associated with Lymph Node Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; CD73; Immunohistochemistry; Lymph node; Metastasis ID Prostate cancer; CD73; Immunohistochemistry; Lymph node; Metastasis AB Prostate cancer is the most common malignancy in men in Europe and North America. At present, it is becoming an increasingly common cancer in China. CD73 (ecto-5′-nucleotidase) is a glycosylphosphatidylinositol (GPI)-linked 70-kDa cell surface enzyme. It is also broadly expressed in many types of tissues. Recent studies have showed that CD73 is widely expressed on malignancies and is up-regulated in cancerous tissues. Consequently, we analyzed the expression of CD73 in prostate cancer tissue. The expression of the CD73 protein was evaluated by Immunohistochemistry staining in 116 tissue specimens. The expression was further examined by quantitative real-time PCR (qRT-PCR) and Western blot in the same set of patients. The intense cell membrane staining for the CD73 protein was observed. The expression of CD73 in lymph node non-metastasizing prostate cancer tissues can be seen at low levels, and is generally undetectable. RT-PCR and Western blot showed that the expression of CD73 in lymph node metastasizing prostate cancer was higher compared with non-metastasizing ones. These results suggest that CD73 could be considered as a relevant-specific target for molecular therapy of prostate cancer metastasis. C1 [Yang, Qing] Key Laboratory of Cancer Prevention and Therapy Tianjin, Tianjin Medical University Cancer Institute and Hospital, Department of Genitourinary Oncology, 300060 Tianjin, China. [Du, Jun] Key Laboratory of Cancer Prevention and Therapy Tianjin, Tianjin Medical University Cancer Institute and Hospital, Department of Genitourinary Oncology, 300060 Tianjin, China. [Zu, Lingling] Tianjin Medical University General Hospital, Tianjin Lung Cancer InstituteTianjin, China. RP Yang, Q (reprint author), Key Laboratory of Cancer Prevention and Therapy Tianjin, Tianjin Medical University Cancer Institute and Hospital, Department of Genitourinary Oncology, 300060 Tianjin, China. EM yangqingtj@163.com CR Ruan Y, YuW, Cheng F et al, 2012, Detection of prostate stem cell antigen expression in human prostate cancer using quantum-dotbased technology. Sensors, Basel, 12(5):5461–5470 Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics. CA Cancer J Clin 60(5):277–300 Liu Z, Ren G, Shangguan C et al, 2012, ATRA inhibits the proliferation of DU145 prostate cancer cells through reducing the methylation level of HOXB13 gene. PLoS One 7(7):e40943 Resta R, Yamashita Y, Thompson LF, 1998, Ecto-enzyme and signaling functions of lymphocyte CD73. Immunol Rev 161:95–109 Colgan SP, Eltzschig HK, Eckle T, Thompson LF, 2006, Physiological roles for ecto-5′-nucleotidase, CD73). Purinergic Signal 2(2):351–360 Zhang B, 2010, CD73: a novel target for cancer immunotherapy. Cancer Res 70(16):6407–6411 Niemela J, Henttinen T, Yegutkin GG et al, 2004, IFN-alpha induced adenosine production on the endothelium: a mechanism mediated by CD73, ecto-50-nucleotidase, upregulation. J Immunol 172(3):1646–1653 Jin D, Fan J, Wang L et al, 2010, CD73 on tumor cells impairs antitumor T-cell responses: a novel mechanism of tumor-induced immune suppression. Cancer Res 70(6):2245–2255 Leth-Larsen R, Lund R, Hansen HVet al, 2009, Metastasis-related plasma membrane proteins of human breast cancer cells identified by comparative quantitative mass spectrometry. Mol Cell Proteomics 8(6):1436–1439 Strohmeier GR, Lencer WI, Patapoff TW et al, 1997, Surface expression, polarization, and functional significance of CD73 in human intestinal epithelia. J Clin Invest 99(11):2588–2601 Lennon PF, Taylor CT, Stahl GL, Colgan SP, 1998, Neutrophilderived 50-adenosine monophosphate promotes endothelial barrier function via CD73-mediated conversion to adenosine and endothelial A2B receptor activation. J Exp Med 188(8):1433– 1443 Spychala J, 2000, Tumor-promoting functions of adenosine. Pharmacol Ther 87(2–3):161–173 Canbolat O, Durak I, Cetin R et al, 1996, Activities of adenosine deaminase, 59-nucleotidase, guanase, and cytidine deaminase enzymes in cancerous and non-cancerous human breast tissues. Breast Cancer Res Treat 37(2):189–193 Wang L, Zhou X, Zhou T et al, 2008, Ecto-5′-nucleotidase promotes invasion, migration and adhesion of human breast cancer cells. J Cancer Res Clin Oncol 134(3):365–372 Yegutkin GG, Marttila-Ichihara F, Karikoski M et al, 2011, Altered purinergic signaling in CD73-deficient mice inhibits tumor progression. Eur J Immunol 41(5):1231–1241 Wu XR, He XS, Chen YF et al, 2012, High expression of CD73 as a poor prognostic biomarker in human colorectal cancer. J Surg Oncol 106(2):130–137 Liu N, Fang XD, Vadis Q, 2012, CD73 as a novel prognostic biomarker for human colorectal cancer. J Surg Oncol 106(7):918– 919 Cappellari AR, Vasques GJ, Bavaresco L et al, 2012, Involvement of ecto-5′-nucleotidase/CD73 in U138MG glioma cell adhesion. Mol Cell Biochem 359(1–2):315–322 Kondo T, Nakazawa T, Murata SI, Katoh R, 2006, Expression of CD73 and its ecto-5′-nucleotidase activity are elevated in papillary thyroid carcinomas. Histopathology 48(5):612–614 Wang L, Fan J, Thompson LF et al, 2011, CD73 has distinct roles in non hematopoietic and hematopoietic cells to promote tumor growth in mice. J Clin Invest 121(6):2371–2382 Stagg J, Divisekera U, Duret H et al, 2011, CD73-deficient mice have increased antitumor immunity and are resistant to experimental metastasis. Cancer Res 71(8):2892–2900 Stagg J, Divisekera U, McLaughlin N et al, 2010, Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis. Proc Natl Acad Sci USA 107(4):1547–1552 Lee H, Lin EC, Liu L, Smith JW, 2003, Gene expression profiling of tumor xenografts: in vivo analysis of organ-specific metastasis. Int J Cancer 107(4):528–534 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 811 EP 814 DI 10.1007/s12253-013-9648-7 PG 4 ER PT J AU Wang, F Liu, Y Zhang, H AF Wang, Fei Liu, Yulong Zhang, Hao TI Loss of MTSS1 Expression is an Independent Prognostic Factor for Hilar Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cholangiocarcinoma; MTSS1; Immunohistochemistry; Prognosis ID Cholangiocarcinoma; MTSS1; Immunohistochemistry; Prognosis AB Metastasis suppressor 1 (MTSS1) is a novel metastasis suppressor gene in a variety of cancers. This study aimed to detect MTSS1 expression in normal and cancerous tissue specimens from Chinese patients with hilarcholangiocarcinoma to determine the association with clinicopathological parameters and survival. Tissue microarrays containing normal and tumor specimens were constructed using paraffin blocks from 61 patients for immunohistochemical analysis of MTSS1 expression. A subgroup of these tissues was verified by Western blot analysis. MTSS1 protein was expressed in 24 of 61 cases (39.3 %) of tumor tissues, compared to that in 22 of 26 (84.6 %) of non-neoplastic bile duct epithelium and in 26 of 26 (100 %) of adjacent normal liver cells. Loss of MTSS1 expression was associated with lymph node metastases of cholangiocelluarcarcinoma and tumor cell de-differentiation. MTSS1 expression inversely associated with tumor recurrence and overall survival of the patients by univariate and multivariate analyses. MTSS1 expression was significantly decreased in human hilarcholangiocarcinoma and lost MTSS1 expression was associated with pooroverall survival and tumor recurrences in cholangiocarcinoma patients. Thus, MTSS1 expression represents an independent predictor for tumor recurrence and overall survival in patients with cholangiocarcinoma. C1 [Wang, Fei] Eastern Hepatobiliary Hospital, Department of SurgeryShanghai, China. [Liu, Yulong] the Second Affiliated Hospital of Soochow University, Emergency CenterSuzhou, China. [Zhang, Hao] Fudan University, Hua Shan Hospital, Department of General Surgery, Wulumuqi Middle Road 12, 200040 Shanghai, China. RP Zhang, H (reprint author), Fudan University, Hua Shan Hospital, Department of General Surgery, 200040 Shanghai, China. EM drhao@hotmail.com CR Ito F, Cho CS, Rikkers LF, Weber SM, 2009, Hilar cholangiocarcinoma: Current management. Ann Surg 250(2):210–218 Otani K, Chijiiwa K, Kai M, Ohuchida J, Nagano M, Tsuchiya K, Kondo K, 2008, Outcome of surgical treatment of hilar cholangiocarcinoma. J Gastrointest Surg 12(6):1033–1040 Liu CL, Fan ST, Lo CM, Tso WK, Lam CM, Wong J, 2006, Improved operative and survival outcomes of surgical treatment for hilar cholangiocarcinoma. Br J Surg 93(12):1488–1494 Sawada T, Kita J, Rokkaku K, Kato M, Shimoda M, Kubota K, 2008, Outcome of surgical resection for hilar cholangiocarcinoma in elderly patients. Hepatogastroenterology 55(88):1971–1974 Seyama Y, Makuuchi M, 2007, Current surgical treatment for bile duct cancer. World J Gastroenterol 13(10):1505–1515 Maeno H, Ono T, Yamanoi A, Nagasue N, 2007, Our experiences in surgical treatment for hilar cholangiocarcinoma. Hepatogastroenterology 54(75):669–673 Nixdorf S, Grimm MO, Loberg R, Marreiros A, Russell PJ, Pienta KJ, Jackson P, 2004, Expression and regulation of MIM, Missing In Metastasis), a novel putative metastasis suppressor gene, and MIM-B, in bladder cancer cell lines. Cancer Lett 215(2):209–220 Utikal J, Gratchev A, Muller-Molinet I, Oerther S, Kzhyshkowska J, Arens N, Grobholz R, Kannookadan S, Goerdt S, 2006, The expression of metastasis suppressor MIM/MTSS1 is regulated by DNA methylation. Int J Cancer 119(10):2287–2293 Wang Y, Liu J, Smith E, Zhou K, Liao J, Yang GY, Tan M, Zhan X, 2007, Downregulation of missing in metastasis gene, MIM, is associated with the progression of bladder transitional carcinomas. Cancer Invest 25(2):79–86 Parr C, Jiang WG, 2009, Metastasis suppressor 1, MTSS1, demonstrates prognostic value and anti-metastatic properties in breast cancer. Eur J Cancer 45(9):1673–1683 Liu K,Wang G, Ding H, Chen Y, Yu G,Wang J Downregulation of metastasis suppressor 1(MTSS1, is associated with nodal metastasis and poor outcome in Chinese patients with gastric cancer. BMC Cancer 10:428 Hicks DG, Yoder BJ, Short S, Tarr S, Prescott N, Crowe JP, Dawson AE, Budd GT, Sizemore S, Cicek M et al, 2006, Loss of breast cancer metastasis suppressor 1 protein expression predicts reduced disease-free survival in subsets of breast cancer patients. Clin Cancer Res 12(22):6702–6708 Ma S, Guan XY, Lee TK, Chan KW, 2007, Clinicopathological significance of missing in metastasis B expression in hepatocellular carcinoma. Hum Pathol 38(8):1201–1206 Callahan CA, Ofstad T, Horng L, Wang JK, Zhen HH, Coulombe PA, Oro AE, 2004, MIM/BEG4, a Sonic hedgehog-responsive gene that potentiates Gli-dependent transcription. Genes Dev 18(22):2724–2729 Wang D, Zhang H, Fang Z, Yu G: Annexin-1 downregulation is associated with clinical outcome in chinese patients with hilar cholangiocarcinoma. Eur Surg Res 45(3–4):151–157 Liu JH, Song LB, Zhang X, Guo BH, Feng Y, Li XX, Liao WT, Zeng MS, Huang KH, 2008, Bmi-1 expression predicts prognosis for patients with gastric carcinoma. J Surg Oncol 97(3):267–272 Urano N, Fujiwara Y, Doki Y, Tsujie M, Yamamoto H, Miyata H, Takiguchi S, Yasuda T, Yano M, Monden M, 2006, Overexpression of hypoxia-inducible factor-1 alpha in gastric adenocarcinoma. Gastric Cancer 9(1):44–49 Tsai HM, Chuang CH, Lin XZ, Chen CY, 2009, Factors relating to the short term effectiveness of percutaneous biliary drainage for hilar cholangiocarcinoma. World J Gastroenterol 15(41):5206–5210 Tsalis K, Vasiliadis K, Kalpakidis V, Christoforidis E, Avgerinos A, Botsios D, Megalopoulos A, Haidich AB, Betsis D, 2007, A single-center experience in the management of Altemeier-Klatskin tumors. J Gastrointestin Liver Dis 16(4):383–389 Ramacciato G, Nigri G, Bellagamba R, Petrucciani N, Ravaioli M, Cescon M, Del Gaudio M, Ercolani G, Di Benedetto F, Cautero N et al, 2010, Univariate and multivariate analysis of prognostic factors in the surgical treatment of hilar cholangiocarcinoma. Am Surg 76(11):1260–1268 Murakami Y, Uemura K, Sudo T, Hashimoto Y, Nakashima A, Kondo N, Sakabe R, Ohge H, Sueda T, 2011, Prognostic factors after surgical resection for intrahepatic, hilar, and distal cholangiocarcinoma. Ann Surg Oncol 18(3):651–658 Li Q, Li HK, Hao XS, 2009, Analysis of the surgical outcome and prognostic factors for hilar cholangiocarcinoma. ZhonghuaWai Ke Za Zhi 47(2):94–97 Kirimlioglu H, Turkmen I, Bassullu N, Dirican A, Karadag N, Kirimlioglu V, 2009, The expression of matrix metalloproteinases in intrahepatic cholangiocarcinoma, hilar, Klatskin tumor), middle and distal extrahepatic cholangiocarcinoma, gallbladder cancer, and ampullary carcinoma: role of matrix metalloproteinases in tumor progression and prognosis. Turk J Gastroenterol 20(1):41–47 Fiorentino M, Altimari A, D’Errico A, Gabusi E, Chieco P, Masetti M, Grigioni WF, 2001, Low p27 expression is an independent predictor of survival for patients with either hilar or peripheral intrahepatic cholangiocarcinoma. Clin Cancer Res 7(12):3994–3999 Thelen A, Scholz A, Benckert C, Schroder M, Weichert W, Wiedenmann B, Neuhaus P, Jonas S, 2008, Microvessel density correlates with lymph node metastases and prognosis in hilar cholangiocarcinoma. J Gastroenterol 43(12):959–966 Nishihara Y, Aishima S, Hayashi A, Iguchi T, Fujita N, Taketomi A, Honda H, Tsuneyoshi M, 2009, CD10+ fibroblasts are more involved in the progression of hilar/extrahepatic cholangiocarcinoma than of peripheral intrahepatic cholangiocarcinoma. Histopathology 55(4):423–431 Bershteyn M, Atwood SX, Woo WM, Li M, Oro AE: MIM and cortactin antagonism regulates ciliogenesis and hedgehog signaling. Dev Cell 19(2):270–283. Bompard G, Sharp SJ, Freiss G, Machesky LM, 2005, Involvement of Rac in actin cytoskeleton rearrangements induced by MIM-B. J Cell Sci 118(Pt 22):5393–5403 Lin J, Liu J, Wang Y, Zhu J, Zhou K, Smith N, Zhan X, 2005, Differential regulation of cortactin and N-WASP-mediated actin polymerization by missing in metastasis, MIM, protein. Oncogene 24(12):2059–2066 Mattila PK, Pykalainen A, Saarikangas J, Paavilainen VO, Vihinen H, Jokitalo E, Lappalainen P, 2007, Missing-in-metastasis and IRSp53 deform PI(4,5)P2-rich membranes by an inverse BAR domain-like mechanism. J Cell Biol 176(7):953–964 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 815 EP 820 DI 10.1007/s12253-013-9649-6 PG 6 ER PT J AU Ma, J Ma, J Meng, Q Zhao, ZSh Xu, Wj AF Ma, Jie Ma, Jun Meng, Qun Zhao, Zhong-Sheng Xu, Wen-juan TI Prognostic Value and Clinical Pathology of MACC-1 and c-MET Expression in Gastric Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; MACC-1; c-MET; Progression; Prognosis ID Gastric cancer; MACC-1; c-MET; Progression; Prognosis AB This study was to assess the expression of MACC-1 and c-MET in gastric cancer, and to correlate this expression with clinicohistological parameters and patient prognosis. Total RNA was extracted from cancer tissue and adjacent normal mucosa from frozen biopsy specimens of 30 patients with gastric cancer, and MACC-1 expression was assessed by RTPCR. MACC-1 and c-MET protein expression were also assessed in paraffin-embedded tissues obtained from 436 tumor mucosa and 92 normal mucosa specimens by immunohistochemistry. The correlation between MACC-1 and c-MET expression and clinicopathological factors (age, sex, histology, tumor depth, lymph node status and vessel invasion) were also evaluated. RT-PCR analysis revealed that MACC-1 expression was significantly higher in cancerous mucosa compared with normal tissue. Immunohistochemical analysis indicated that MACC-1 and c-MET were moderately or strongly expressed in gastric cancer tissue, whereas expression was weak or absent in non-cancer tissue. Expression of MACC-1 or c-MET was significantly associated with larger tumor size, deeper tumor invasion, presence of lymph node metastasis, lymphatic involvement, venous invasion, distant metastasis and advanced clinical stage. However, only MACC-1 exhibited significantly greater expression in carcinomas from the higher age group. The intensity of MACC-1 and c-MET expression was also positively correlated. Survival analysis of the 436 gastric cancer patients revealed that patients in clinical stages I, II and III exhibiting lower MACC-1 and c-MET expression had a higher 5-year survival rate compared with patients expressing high levels of these proteins. Multivariate analysis revealed that MACC-1 and c-MET may be independent prognostic indexes of gastric carcinoma (P<0.01). Our findings confirm that MACC-1 and c-MET expression is strongly related to gastric cancer stage and degree of malignancy, and is inversely correlated to patient prognosis. Thus, MACC-1 and c-MET may interact to promote tumorigenesis and their expression may be used as independent prognostic markers in gastric cancer. C1 [Ma, Jie] Zhejiang Provincial People’s Hospital, Department of PathologyHangzhou, Zhejiang, China. [Ma, Jun] GongShu District People’s Hospital, Department of MedcineHangzhou, Zhejiang, China. [Meng, Qun] Zhejiang Provincial People’s Hospital, Department of PathologyHangzhou, Zhejiang, China. [Zhao, Zhong-Sheng] Zhejiang Provincial People’s Hospital, Department of PathologyHangzhou, Zhejiang, China. [Xu, Wen-juan] Zhejiang Provincial People’s Hospital, Department of PathologyHangzhou, Zhejiang, China. RP Zhao, ZSh (reprint author), Zhejiang Provincial People’s Hospital, Department of Pathology, Hangzhou, China. EM zhaozhongsheng1950@163.com CR Steeg PS, 2006, Tumor metastasis: mechanistic insights and clinical challenges. Nat Med 12:895–904 Smith BR, Stabile BE, 2009, Extreme aggressiveness and lethality of gastric adenocarcinoma in the very young. Arch Surg 144(6):506–10 Birchmeier C, Birchmeier W, Gherardi E et al, 2003, Met, metastasis, motility and more. Nat Rev Mol Cell Biol 4(12):915–925 Okines AF, Reynolds AR, Cunningham D, 2011, Targeting angiogenesis in esophagogastric adenocarcinoma. Oncologist 16(6):844–58 Lai L, Goldschneider I, 2001, Cutting edge: identification of a hybrid cytokine consisting of IL-7 and the beta-chain of the hepatocyte growth consisting of IL-7 and the beta-chain of the hepatocyte growth factor/scatter factor. Immun 167:3550–3554 Stein U, Ahher W, Arlt F et al, 2009, MACC1, a newly identifiedkey regulator of HGF–MET signaling, predicts colon cancermetastasis. Nat Med 15(1):59–67 Lee HS, Lee HK, Kim HS, Yang HK, Kim WH, 2003, Tumour suppressor gene expression correlates with gastric cancer prognosis. J Pathol 200:39–46 Lee HS, Lee HK, Kim HS, Yang HK, Kim YI, Kim WH, 2001, MUC1, MUC2, MUC5AC, and MUC6 expressions in gastric carcinomas: their roles as prognostic indicators. Cancer 92:1427–34 Zhang D, Salto-TellezM, Putti TC,Do E,Koay ES, 2003, Reliability of tissue microarrays in detecting protein expression and gene amplification in breast cancer. Mod Pathol 16:79–84 Lee HS, Cho SB, Lee HE et al, 2007, Protein expression profiling and molecular classification of gastric cancer by the tissue array method. Clin Cancer Res 13(14):4154–63 Mizokami K, Kakeji Y, Oda S et al, 2006, Clinicopathologic significance of hypoxia-inducible factor 1 alpha overexpression in gastric carcinomas. J Surg Oncol 94:149–154 Song LB, Liao WT, Mai HQ et al, 2006, The clinical significance of twist expression in nasopharyngeal carcinoma. Cancer Lett 242:258–65 KolevY, Uetake H, Iida S, Ishikawa T, Kawano T, SugiharaK, 2007, Prognostic significance of VEGF expression in correlation with COX-2, microvessel density, and clinicopathological characteristics in human gastric carcinoma. Ann Surg Oncol 14(10):2738–47 Xu ZY, Chen JS, Shu YQ, 2010, Gene expression profile towards the prediction of patient survival of gastric cancer. Biomed Pharmacother 64(2):133–9 Weidner KM, Arakaki N, Hartmann G et al, 1991, Evidence for the identity of human scatter factor and human hepatocyte growth factor. Proc Nat Acad Sci USA 88:7001–7005 Sierra JR, Tsao MS, 2011, c-MET as a potential therapeutic target and biomarker in cancer. Ther Adv Med Oncol 3(1 Suppl):S21–35 Galimi F, Torti D, Sassi F et al, 2011, Genetic and expression analysis of MET, MACC1, and HGF in metastatic colorectal cancer: response to met inhibition in patient xenografts and pathologic correlations. Clin Cancer Res 17(10):3146–56 Arlt F, Stein U, 2009, Colon cancer metastasis: MACC1 and Met as metastatic pacemakers. Int J Biochem Cell Biol 41(12):2356–9 Kokoszynska K, Krynski J, Rychlewski L et al, 2009, Unexpected domain composition of MACC1 links MET signaling and apoptosis. Acta Biochim Pol 56(2):317–23 Lang AH, Geller-Rhomberg S, Winder T et al, 2012, A common variant of the MACC1 gene is significantly associated with overall survival in colorectal cancer patients. BMC Cancer 12(1):20 Stein U, Dahlmann M, Walther W, 2010, MACC1—more than metastasis? Facts and predictions about a novel gene. J Mol Med, Berl, 88(1):11–8 Qiu J, Huang P, Liu Q, Hong J, Li B, Lu C, Wang L, Wang J, Yuan Y, 2011, Identification of MACC1 as a novel prognostic marker in hepatocellular carcinoma. J Transl Med 9:166 Chundong G, Uramoto H, Onitsuka T et al, 2011, Molecular diagnosis of MACC1 status in lung adenocarcinoma by immunohistochemical analysis. Anticancer Res 31(4):1141–5 Aoba-ku Y, 2010, MACC 1 as a marker for peritoneal-disseminated gastric carcinoma. Anticancer Res 30:3441–3444 Zhang R, Shi H, Chen Z et al, 2011, Effects of metastasis-associated in colon cancer 1 inhibition by small hairpin RNA on ovarian carcinoma OVCAR-3 cells. J Exp Clin Cancer Res 30:83 Shirahata A, Shinmura K, Kitamura K et al, 2010, MACC 1 as a marker for advanced colorectal carcinoma. Anticancer Res 30:3441–3444 Boardman LA, 2009, Overexpression of MACC1 leads to downstream activation of HGF/MET and potentiates metastasisand recurrence of colorectal cancer. Genome Med 1(4):361–364 Brand-Saberi B, Muller TS, Wilting J et al, 1996, Scatter factor/ hepatocyte growth factor, SF/HGF, induces emigration of myogenic cells at interlimb level in vivo. Dev Biol 179(1):303–8 Borowiak M, Garratt AN, Wustefeld T, Strehle M, Trautwein C, Birchmeier C, 2004, Met provides essential signals for liver regeneration. Proc Natl Acad Sci U S A 101:10608–10613 Bahary N, Zon L, 2001, Development. Endothelium–chicken soup forthe endoderm. Science 294(5542):530–531 Wu FS, Zheng SS, Wu LJ et al, 2006, Study on the pregnomic value of hepatocyte growth factor and c-met for patients with hepatocellular carcinoma. Zhonghua Wai lke za zlli 44(9):603–608 Chen BK, Ohtsuki Y, Furihata M et al, 1999, Overexpression of c-Metprotein in human thyroid tumors correlated with lymph node metastasis and clinicopathologic stage. Pathol Res Pract 195(6):427–433 Ghoussoub RA, Dillon DA, D’Aquila T et al, 1998, Expression of c-metis a strong independent prognostic factor in breast carcinoma. Cancer 82(8):1513–1520 Camp RL, Rimm EB, Goedecke S et al, 1999, Met expression is associated with poor outcome in patients with axillary lymph node negative breast carcinoma. Cancer 86:2259–2265 Zhang RT, Shi HR, Huang HL et al, 2011, Expressions of MACC1, HGF, and C-met protein in epithelial ovarian cancer and their significance. Nan Fang Yi Ke Da Xue Xue Bao 31(9):1551–5 Di Renzo MF, Olivero M, Giacomini A et al, 1995, Overexpression and amplification of the met/HGF receptor gene during the progression of colorectal cancer. Clin Cancer Res 1(2):147– 54 Nakajima M, Sawada H, Yamada Y et al, 1999, The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer 85(9):1894–902 Ma PC, Jagadeeswaran R, Jagadeesh S et al, 2005, Functional expression and mutations of c-Met and its therapeutic inhibition with SU11274 and small interfering RNA in non-small cell lung cancer. Cancer Res 65(4):1479–88 Migliore C, Martin V, Leoni VP et al, 2012, MiR-1 downregulation cooperates withMACC1 in promoting METoverexpression in human colon cancer. Clin Cancer Res 18(3):737–47 Herynk MH, Stoeltzing O, Reinmuth N et al, 2003, Down regulation of c-Met inhibits growth in the liver of human colorectal carcinoma cells. Cancer Res 63(11):2990–299 Date K, Matsumoto K, Kuba K et al, 1998, Inhibition of tumor growth and invasion by afour-kringleantagonis t(HGF/NK4, for heap-tocyte growth factor. Oncogene 17(23):3045–3051 Stein U, Schlag PM, 2007, Clinical, biological, and molecular aspects of metastasis in colorectalcancer. Recent Res Cancer Res 176:61–8 Kokoszynska K, Krynski J, Rychlewski L et al, 2009, Unexpected domain composition of MACC1 links MET signaling and apoptosis. Acta Biochim Pol 56:317–323 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 821 EP 832 DI 10.1007/s12253-013-9650-0 PG 12 ER PT J AU Chen, HJ Yan, HH Yang, JJ Chen, ZH Su, J Zhang, XCh Wu, YL AF Chen, Hua-Jun Yan, Hong-Hong Yang, Jin-Ji Chen, Zhi-Hong Su, Jian Zhang, Xu-Chao Wu, Yi-Long TI Disease Flare After EGFR Tyrosine Kinase Inhibitor Cessation Predicts Poor Survival in Patients with Non-small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; Epidermal growth factor receptor; Tyrosine kinase inhibitor; Cessation; Disease flare ID Non-small cell lung cancer; Epidermal growth factor receptor; Tyrosine kinase inhibitor; Cessation; Disease flare AB Available study revealed non-small cell lung cancer (NSCLC) patients faced a risk of disease flare after cessation of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) treatment. There was no data concerning the prognostic value of disease flare. This study aimed to investigate the prevalence of disease flare in a Chinese cohort, and analyzed its prediction to survival. A cohort of 227 NSCLC patients with acquired resistance to EGFR TKI was retrospectively analyzed. Prevalence and clinical features of disease flare after TKI cessation were reviewed. Survival data were analyzed between patients with flare and those without flare. EGFR gene mutations in tumors were detected. Twenty of 227 (8.8 %) patients were determined with disease flare after TKI cessation. The median interval from TKI cessation to disease flare was 7 days (range 3–18). Forty percent of patients complained of deteriorated dyspnea attributable to malignant effusion. Thirty percent of patients had progressive lesions in the brain. After TKI cessation 35 % of flare patients died before challenge of subsequent treatment. No response was observed in 30 % of flare patients undergoing subsequent chemotherapy. When compared with the non-flare group, patients with disease flare demonstrated comparable progression-free survival (10.1 vs. 9.9 months; P=0.973), shorter post-TKI survival (4.1 vs. 6.1 months; P<0.001), and a significantly poor overall survival (16.6 vs. 21.6 months; P=0.002). Disease flare after cessation of EGFR TKI occurred in Chinese NSCLC population and predicted a poor survival. C1 [Chen, Hua-Jun] Guangdong General Hospital, Guangdong Lung Cancer InstituteGuangzhou, China. [Yan, Hong-Hong] Guangdong General Hospital, Guangdong Lung Cancer InstituteGuangzhou, China. [Yang, Jin-Ji] Guangdong General Hospital, Guangdong Lung Cancer InstituteGuangzhou, China. [Chen, Zhi-Hong] Guangdong General Hospital, Guangdong Lung Cancer InstituteGuangzhou, China. [Su, Jian] Guangdong General Hospital, Guangdong Lung Cancer InstituteGuangzhou, China. [Zhang, Xu-Chao] Guangdong General Hospital, Guangdong Lung Cancer InstituteGuangzhou, China. [Wu, Yi-Long] Guangdong General Hospital, Guangdong Lung Cancer InstituteGuangzhou, China. RP Wu, YL (reprint author), Guangdong General Hospital, Guangdong Lung Cancer Institute, Guangzhou, China. EM syylwu@live.cn CR Mok TS, Wu YL, Thongprasert S et al, 2009, Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 361:947–957 Mitsudomi T, Morita S, Yatabe Y et al, 2010, Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor, WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 11:121–128 Zhou CC, Wu YL, Chen G et al, 2011, Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer, OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 12:735–742 Rosell R, Carcereny E, Gervais R et al, 2012, Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small cell lung cancer, EURTAC): a multicentre, open-label, randomized phase 3 trial. Lancet Oncol 13:239–246 Bean J, Brennan C, Shih JY et al, 2007, MET amplification occurs with or without T790M mutation in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci USA 104:20932–20937 Engelman JA, Zejnullahu K, Mitsudomi T et al, 2007, MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316:1039–1043 Pao W, Miller VA, Politi KA et al, 2005, Acquired resistance of lung adenocarcinoma to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PloS Med 2:e73 Oxnard GR, Arcila ME, Sima CS et al, 2011, Acquired resistance to EGFR tyrosine kinase inhibitors in EGFR-mutant lung cancer: distinct natural history of patients with tumors harboring the T790M mutation. Clin Cancer Res 17:1616–1622 Ettinger DS, Akerley W, Bepler G et al, 2010, Non-small cell lung cancer. J Natl Compr Canc Netw 8:740–801 Chaft JE, Oxnard GR, Sima CS et al, 2011, Disease flare after tyrosine kinase inhibitor discontinuation in patients with EGFRmutant lung cancer and acquired resistance to erlotinib or gefitinib: implications for clinical trial design. Clin Cancer Res 17:6298–6303 Kim YH, Fukuhara A, Mishima M, 2011, Should epidermal growth factor receptor-tyrosine kinase inhibitor be continued beyond progressive disease? Case Rep Oncol 4:470–474 Beasley MB, Brambilla E, Travis WD, 2005, The 2004 world health organization classification of lung tumors. Semin Roentgenol 40:90–97 Detterbeck FC, Boffa DJ, Tanoue LT, 2009, The new lung cancer staging system. Chest 136:260–271 Lee YJ, Choi HJ, Kim SK et al, 2010, Frequent central nervous system failure after clinical benefit with epidermal growth factor receptor tyrosine kinase inhibitors in Korean patients with nonsmall- cell lung cancer. Cancer 116:1336–1343 Therasse P, Arbuck SG, Eisenhauer EA et al, 2000, New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst 92:205–216 Lynch TJ, Bell DW, Sordella R et al, 2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350:2129–2139 Zhou Q, Zhang XC, Chen ZH et al, 2011, Relative abundance of EGFR mutations predicts benefit from gefitinib treatment for advanced non-small-cell lung cancer. J Clin Oncol 29:3316–3321 Riely GJ, Kris MG, Zhao B et al, 2007, Prospective assessment of discontinuation and reinitiation of erlotinib or gefitinib in patients with acquired resistance to erlotinib or gefitinib followed by the addition of everolimus. Clin Cancer Res 13:5150–5155 Omuro AM, Kris MG, Miller VA et al, 2005, High incidence of disease recurrence in the brain and leptomeninges in patients with non-small cell lung carcinoma after response to gefitinib. Cancer 103:2344–2348 Penel N, Brichet A, Prevost B et al, 2001, Prognostic factors of synchronous brain metastases from lung cancer. Lung Cancer 33:143–154 Ampil F, Caldito G, Milligan S et al, 2007, The elderly with synchronous non-small cell lung cancer and solitary brain metastasis: does palliative thoracic radiotherapy have a useful role? Lung Cancer 57:60–65 Sharma SV, Lee DY, Li B et al, 2010, A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141:69–80 Becker A, Crombag L, Heideman DA et al, 2011, Retreatment with erlotinib: regain of TKI sensitivity following a drug holiday for patients with NSCLC who initially responded to EGFR-TKI treatment. Eur J Cancer 47:2603–2606 Heon S, Nishino M, Goldberg SB et al., 2012, Response to EGFR tyrosine kinase inhibitor, TKI, retreatment after a drug-free interval in EGFR-mutant advanced non-small cell lung cancer, NSCLC, with acquired resistance. In: ASCO Annual Meeting [Abstract 7525] Mitsudomi T, Morita S, Yatabe Y et al., 2012, Updated overall survival results of WJTOG 3405, a randomized phase III trial comparing gefitinib, G, with cisplatin plus docetaxel, CD, as the first-line treatment for patients with non-small cell lung cancer harboring mutations of the epidermal growth factor receptor, EGFR). In: ASCO Annual Meeting [Abstract 7521] Zhou CC, Wu YL, Liu XQ et al., 2012, Overall survival, OS, results from OPTIMAL, CTONG0802), a phase III trial of erlotinib, E, versus carboplatin plus gemcitabine, GC, as first-line treatment for Chinese patients with EGFR mutation-positive advanced non-small cell lung cancer, NSCLC). In: ASCO Annual Meeting [Abstract 7520] NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 833 EP 838 DI 10.1007/s12253-013-9651-z PG 6 ER PT J AU Menschikowski, M Hagelgans, A Schuler, U Froeschke, S Rosner, A Siegert, G AF Menschikowski, Mario Hagelgans, Albert Schuler, Ulrich Froeschke, Susanne Rosner, Andrea Siegert, Gabriele TI Plasma Levels of Phospholipase A2-IIA in Patients with Different Types of Malignancies: Prognosis and Association with Inflammatory and Coagulation Biomarkers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer; Inflammation; Secreted phospholipase A2; C-reactive protein; Fibrinogen ID Cancer; Inflammation; Secreted phospholipase A2; C-reactive protein; Fibrinogen AB It is well-known that the plasma level of group IIA phospholipase A2 (sPLA2-IIA) is increased in patients with malignant diseases, but whether the up-regulated enzyme expression is directly related to tumorigenesis or a consequence of tumor-associated inflammation remains unresolved. In this study we analyzed circulating levels of sPLA2-IIA, C-reactive protein (CRP), fibrinogen, factor VIII (FVIII), von Willebrand factor (vWF), and antithrombin as biomarkers of inflammation and coagulation in patients with various types of malignancies. Underlying tumor entities were lung, esophageal, gastric, pancreatic, colorectal, head and neck, and hepatocellular carcinomas as well as multiple myeloma and non-Hodgkin’s lymphoma. Plasma levels of sPLA2-IIA are shown to be markedly increased in all types of analysed malignancies in comparison to the normal range (22.8±4.5 μg/L versus <1.9 μg/L). Levels of sPLA2-IIA correlate positively with CRP (p<0.001), fibrinogen (p<0.01), FVIII (p<0.05), and vWF (p<0.05) and negatively with antithrombin levels (p<0.05). Kaplan- Meier analyses revealed a statistically prolonged survival time of patients with lower sPLA2-IIA concentrations (<4 μg/L) in comparison to those with elevated concentrations (>4 μg/L) of this enzyme. In conclusion, the study shows that the measurement of plasma sPLA2-IIA levels has prognostic values in patients with different types of malignancies. The association of sPLA2-IIA levels with CRP, fibrinogen, FVIII, and vWF levels supports the importance of inflammatory processes for the up-regulation of sPLA2-IIA during cancer progression. C1 [Menschikowski, Mario] Technische Universitat Dresden, Medizinische Fakultat, Institut fur Klinische Chemie und Laboratoriumsmedizin, Fetscherstrasse 74, 01307 Dresden, Germany. [Hagelgans, Albert] Technische Universitat Dresden, Medizinische Fakultat, Institut fur Klinische Chemie und Laboratoriumsmedizin, Fetscherstrasse 74, 01307 Dresden, Germany. [Schuler, Ulrich] Technische Universitat Dresden, Medizinische Fakultat, Medizinische Klinik und Poliklinik IDresden, Germany. [Froeschke, Susanne] Technische Universitat Dresden, Medizinische Fakultat, Medizinische Klinik und Poliklinik IDresden, Germany. [Rosner, Andrea] Technische Universitat Dresden, Medizinische Fakultat, Medizinische Klinik und Poliklinik IDresden, Germany. [Siegert, Gabriele] Technische Universitat Dresden, Medizinische Fakultat, Institut fur Klinische Chemie und Laboratoriumsmedizin, Fetscherstrasse 74, 01307 Dresden, Germany. RP Menschikowski, M (reprint author), Technische Universitat Dresden, Medizinische Fakultat, Institut fur Klinische Chemie und Laboratoriumsmedizin, 01307 Dresden, Germany. EM Mario.Menschikowski@uniklinikum-dresden.de CR Menschikowski M, Hagelgans A, Siegert G, 2006, Secretory phospholipase A2 of group IIA: is it an offensive or a defensive player during atherosclerosis and other inflammatory diseases? Prostaglandins Other Lipid Mediat 79:1–33 Triggiani M, Granata F, Giannattasio G, Marone G, 2005, Secretory phospholipases A2 in inflammatory and allergic diseases: not just enzymes. J Allergy Clin Immunol 116:1000–1006 Scott KF, Sajinovic M, Hein J, Nixdorf S, Galettis P, Liauw W, de Souza P, Dong Q, Graham GG, Russell PJ, 2010, Emerging roles for phospholipase A2 enzymes in cancer. Biochimie 92:601–610 Mirtti T, Laine VJO, Hiekkanen H, Hurme S, Rowe O, Nevalainen TJ, Kallajoki M, Alanen K, 2009, Group IIA phospholipase A2 as a prognostic marker in prostate cancer: relevance to clinicopathological variables and disease-specific mortality. Acta Pathol Microbiol Immunol Scand 117:151–161 Laye JP, Gill JH, 2003, Phospholipase A2 expression in tumours: a target for therapeutic intervention? Drug Discov Today 8:710–716 Yamashita S, Yamashita J, Sakamoto K, Inada K, Nakashima Y, Murata K, Saishoji T, Nomura K, Ogawa M, 1993, Increased expression of membrane-associated phospholipase A2 shows malignant potential of human breast cancer cells. Cancer 71:3058–3064 Asano T, Shoda J, Ueda T, Kawamoto T, Todoroki T, Shimonishi M, Tanabe T, Sugimoto Y, Ichikawa A, Mutoh M, Tanaka N, Miwa M, 2002, Expressions of cyclooxygenase-2 and prostaglandin Ereceptors in carcinoma of the gallbladder: crucial role of arachidonate metabolism in tumor growth and progression. Clin Cancer Res 8:1157–1167 Ogawa M, Yamashita S, Sakamoto K, Ikei S, 1991, Elevation of serumgroup II phospholipaseA2 in patientswith cancers of digestive organs. Res Commun Chem Pathol Pharmacol 74:241–244 Yamashita S, Ogawa M, Sakamoto K, Abe T, Arakawa H, Yamashita J, 1994, Elevation of serum group II phospholipase A2 levels in patients with advanced cancer. Clin Chim Acta 228:91–99 Oleksowicz L, Liu Y, Bracken RB, Gaitonde K, Burke B, Succop P, Levin L, Dong Z, Lu S, 2012, Secretory phospholipase A2-IIa is a target gene of the HER/HER2-elicited pathway and a potential plasma biomarker for poor prognosis of prostate cancer. Prostate 72:1140–1149 Kupert E, Anderson M, Liu Y, Succop P, Levin L, Wang J, Wikenheiser-Brokamp K, Chen P, Pinney SM, Macdonald T, Dong Z, Starnes S, Lu S, 2011, Plasma secretory phospholipase A2-IIA as a potential biomarker for lung cancer in patients with solitary pulmonary nodules. BMC Cancer 11:513 Nevalainen TJ, Haapamaki MM, Gronroos JM, 2000, Roles of secretory phospholipases A2 in inflammatory diseases and trauma. Biochim Biophys Acta 1488:83–90 Kallajoki M, Alanen KA, Nevalainen M, Nevalainen TJ, 1998, Group II phospholipase A2 in human male reproductive organs and genital tumors. Prostate 35:263–272 Abe T, Sakamoto K, Kamohara H, Hirano Y, Kuwahara N, Ogawa M, 1997, Group II phospholipase A2 is increased in peritoneal and pleural effusions in patients with various types of cancer. Int J Cancer 74:245–250 Nevalainen TJ, 1993, Serum phospholipase A2 in inflammatory disease. Clin Chem 39:2453–2459 Balkwill F, Mantovani A, 2001, Inflammation and cancer: back to Virchow? Lancet 357:539–545 Coussens LM, Werb Z, 2002, Inflammation and cancer. Nature 420:860–867 Moore MM, Chua W, Charles KA, Clarke SJ, 2010, Inflammation and cancer: causes and consequences. Clin Pharmacol Ther 87:504–508 Roxburgh CS, McMillan DC, 2010, Role of systemic inflammatory response in predicting survival in patients with primary operable cancer. Future Oncol 6:149–163 Mahmoud FA, Rivera NI, 2002, The role of C-reactive protein as a prognostic indicator in advanced cancer. Curr Oncol Rep 4:250–255 McCall P, Catlow J, McArdle PA, McMillan DC, Edwards J, 2011, Tumoral C-reactive protein and nuclear factor kappa-B expression are associated with clinical outcome in patients with prostate cancer. Cancer Biomark 10:91–99 Auwerda JJ, Sonneveld P, de Maat MP, Leebeek FW, 2007, Prothrombotic coagulation abnormalities in patients with newly diagnosed multiple myeloma. Haematologica 92:279–280 Battistelli S, Stefanoni M, Lorenzi B, Dell’avanzato R, Varrone F, Pascucci A et al, 2008, Coagulation factor levels in non-metastatic colorectal cancer patients. Int J Biol Markers 23:36–41 Minnema MC, Fijnheer R, De Groot PG, Lokhorst HM, 2003, Extremely high levels of von Willebrand factor antigen and of procoagulant factor VIII found in multiple myeloma patients are associated with activity status but not with thalidomide treatment. J Thromb Haemost 1:445–449 Palumbo JS, Kombrinck KW, Drew AF, Grimes TS, Kiser JH, Degen JL, Bugge TH, 2000, Fibrinogen is an important determinant of the metastatic potential of circulating tumor cells. Blood 96:3302–3309 Hanebutt FL, Rolf N, Loesel A, Kuhlisch E, Siegert G, Knoefler R, 2008, Evaluation of desmopressin effects on haemostasis in children with congenital bleeding disorders. Haemophilia 14:524–530 Palumbo JS, Degen JL, 2010, Mechanisms coupling the hemostatic system to colitis-associated cancer. Thromb Res 125(Suppl 2):S39–43 Yigit E, Gonullu G, Yucel I, Turgut M, Erdem D, Cakar B, 2008, Relation between hemostatic parameters and prognostic/ predictive factors in breast cancer. Eur J Intern Med 19:602–607 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Grivennikov SI, Greten FR, Karin M, 2010, Immunity, inflammation, and cancer. Cell 140:883–899 Menschikowski M, Hagelgans A, Fuessel S, Mareninova OA, Neumeister V, Wirth MP, Siegert G, 2012, Serum levels of secreted group IIA phospholipase A2 in benign prostatic hyperplasia and prostate cancer: A biomarker for inflammation or neoplasia? Inflammation 35:1113–1138 Rintala EM, Nevalainen TJ, 1993, Synovial-type, group II, phospholipase A2 in serum of febrile patients with haematological malignancy. Eur J Haematol 50:11–16 Orlowski RZ, Baldwin AS Jr, 2002, NF-κB as a therapeutic target in cancer. Trends Mol Med 8:385–389 Grivennikov SI, Karin M, 2010, Dangerous liaisons: STAT3 and NF-κB collaboration and crosstalk in cancer. Cytokine Growth Factor Rev 21:11–19 Fan Y, Dutta J, Gupta N, Fan G, Gelinas C, 2008, Regulation of programmed cell death by NF-κB and its role in tumorigenesis and therapy. Adv Exp Med Biol 615:223–250 Voleti B, Agrawal A, 2005, Regulation of basal and induced expression of C-reactive protein through an overlapping element for OCT-1 and NF-κB on the proximal promoter. J Immunol 175:3386–3390 Begbie M, Notley C, Tinlin S, Sawyer L, Lillicrap D, 2000, The Factor VIII acute phase response requires the participation of NF- κB and C/EBP. Thromb Haemost 84:216–222 Minnema MC, Chang AC, Jansen PM, Lubbers YT, Pratt BM, Whittaker BG et al, 2000, Recombinant human antithrombin III improves survival and attenuates inflammatory responses in baboons lethally challenged with Escherichia coli. Blood 95:1117–1123 Oelschlager C, Romisch J, Staubitz A, Stauss H, Leithauser B, Tillmanns H, Holschermann H, 2002, Antithrombin III inhibits nuclear factor κB activation in human monocytes and vascular endothelial cells. Blood 99:4015–4020 Kim TH, Bae JS, 2010, Inhibitory effects of antithrombin on the expression of secretory group IIA phospholipase A2 in endothelial cells. BMB Rep 43:604–608 Adams RA, Schachtrup C, Davalos D, Tsigelny I, Akassoglou K, 2007, Fibrinogen signal transduction as a mediator and therapeutic target in inflammation: lessons from multiple sclerosis. Curr Med Chem 14:2925–2936 Rubel C, Gomez S, Fernandez GC, Isturiz MA, Caamano J, Palermo MS, 2003, Fibrinogen-CD11b/CD18 interaction activates the NF-κB pathway and delays apoptosis in human neutrophils. Eur J Immunol 33:1429–1438 Fuller GM, Zhang Z, 2001, Transcriptional control mechanism of fibrinogen gene expression. Ann N Y Acad Sci 936:469–479 Graff JR, Konicek BW, Deddens JA, Chedid M, Hurst BM, Colligan B et al, 2001, Expression of group IIa secretory phospholipase A2 increases with prostate tumor grade. Clin Cancer Res 7:3857–3861 Jiang J, Neubauer BL, Graff JR, Chedid M, Thomas JE, Roehm NW et al, 2002, Expression of group IIA secretory phospholipase A2 is elevated in prostatic intraepithelial neoplasia and adenocarcinoma. Am J Pathol 160:667–671 Morgenbesser SD, McLaren RP, Richards B, Zhang M, Akmaev VR, Winter SF et al, 2007, Identification of genes potentially involved in the acquisition of androgen-independent and metastatic tumor growth in an autochthonous genetically engineered mouse prostate cancer model. Prostate 67:83–106 Sved P, Scott KF, McLeod D, King NJ, Singh J, Tsatralis T et al, 2004, Oncogenic action of secreted phospholipase A2 in prostate cancer. Cancer Res 64:6934–6940 Baden LR, Bensinger W, Angarone M, Casper C, Dubberke ER, Freifeld AG, Garzon R, Greene JN, Greer JP, Ito JI, Karp JE, Kaul DR, King E, Mackler E, Marr KA, Montoya JG, Morris- Engemann A, Pappas PG, Rolston K, Segal B, Seo SK, Swaminathan S, Naganuma M, Shead DA, 2012, Prevention and treatment of cancer-related infections. J Natl Compr Cancer Netw 10:1412–1445 Buhmeida A, Bendardaf R, Hilska M, Laine J, Collan Y, Laato M, Syrjanen K, Pyrhonen S, 2009, PLA2, group IIA phospholipase A2, as a prognostic determinant in stage II colorectal carcinoma. Ann Oncol 20:1230–1235 Ganesan K, Ivanova T, Wu Y, Rajasegaran V, Wu J, Lee MH et al, 2008, Inhibition of gastric cancer invasion and metastasis by PLA2G2A, a novel beta-catenin/TCF target gene. Cancer Res 68:4277–4286 Birts CN, Barton CH, Wilton DC, 2010, Catalytic and non-catalytic functions of human IIA phospholipase A2. Trends Biochem Sci 35:28–35 Menschikowski M, Hagelgans A, Gussakovsky E, Kostka H, Paley EL, Siegert G, 2008, Differential expression of secretory phospholipases A2 in normal and malignant prostate cell lines: regulation by cytokines, cell signaling pathways, and epigenetic mechanisms. Neoplasia 10:279–286 Menschikowski M, Hagelgans A, Kostka H, Eisenhofer G, Siegert G, 2008, Involvement of epigenetic mechanisms in the regulation of secreted phospholipase A2 expressions in Jurkat leukemia cells. Neoplasia 10:1195–1203 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 839 EP 846 DI 10.1007/s12253-013-9652-y PG 8 ER PT J AU Jia, Z Wang, K Zhang, A Wang, G Kang, Ch Han, L Pu, P AF Jia, Zhifan Wang, Kun Zhang, Anling Wang, Guangxiu Kang, Chunsheng Han, Lei Pu, Peiyu TI miR-19a and miR-19b Overexpression in Gliomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; miR-19 expression; Real-time PCR; In situ hybridization; PTEN ID Glioma; miR-19 expression; Real-time PCR; In situ hybridization; PTEN AB Astrocytic gliomas are the most common type of human primary brain tumors with poor prognosis. MicroRNAs(miRs) are frequently deregulated in gliomas and play an oncogenic or tumor suppressor role. In our previous study we found that miR-19a and miR-19b were upregulated in malignant glioma cell lines by microRNA array. For further validation of this finding, the expression of miR- 19a and miR-19b was detected by qRT-PCR and in situ hybridization(ISH) in 8 malignant glioma cell lines, 43 freshly resected glioma samples and 75 archival paraffin embedded glioma specimens with different grades of malignancy in the present study. The results demonstrate that miR-19a and miR- 19b are overexpressed in glioma cell lines and astrocytic glioma tissues, and their expression level is positively correlated with tumor grades. Additionally, the tumor suppressor gene PTEN is identified as the target ofmiR-19a and miR-19b by Luciferase assay. It is speculated that miR-19a and miR- 19b may have an oncogenic role in gliomagenesis at least partially via the negative regulation of PTEN and the molecular mechanism of gliomagenesis in which miR 19a and miR- 19b involved should be investigated further. C1 [Jia, Zhifan] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, 300052 Tianjin, China. [Wang, Kun] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, 300052 Tianjin, China. [Zhang, Anling] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, 300052 Tianjin, China. [Wang, Guangxiu] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, 300052 Tianjin, China. [Kang, Chunsheng] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, 300052 Tianjin, China. [Han, Lei] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, 300052 Tianjin, China. [Pu, Peiyu] Tianjin Medical University General Hospital, Department of Neurosurgery, 152 An-Shan Road, 300052 Tianjin, China. RP Pu, P (reprint author), Tianjin Medical University General Hospital, Department of Neurosurgery, 300052 Tianjin, China. EM zhfjia@hotmail.com CR Louis DN, 2006, Molecular pathology of malignant gliomas. Annu Rev Pathol 1:97–117 Stupp R, Mason WP, van den Bent MJ et al, 2005, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352:987–996 Chen L, Li C, Zhang R et al, 2011, miR-17-92 cluster microRNAs confers tumorigenicity in multiple myeloma. Cancer Lett 309(1):62–70 Takakura S, Mitsutake N, Nakashima M et al, 2008, Oncogenic role of miR-17-92 cluster in anaplastic thyroid cancer cells. Cancer Sci 99(6):1147–1154 Ruan JS, Chen HZ, Kurgan L et al, 2008, HuMiTar: a sequencebased method for prediction of human microRNA targets. Algorithm Mol Biol 3:16 You Y, Pu P, Huang Q et al, 2006, Antisense telomerase RNA inhibits the growth of human glioma cells in vitro and in vivo. Int J Oncol 28:1225–1232 Chan JA, Krichevsky AM, Kosik KS, 2005, MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res 65:6029–6033 Zhang Y, Dutta A, Abounader R, 2012, The role of microRNAs in glioma initiation and progression. Front Biosci 17:700–712 Ota A, Tagawa H, Karnan S et al, 2004, Identification and characterization of a novel gene, C13orf25, as a target for 13q31-q32 amplification in malignant lymphoma. Cancer Res 64:3087–3095 Hayashita Y, Osada H, Tatematsu Y et al, 2005, A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res 65:9628–9632 Tagawa H, Karube K, Tsuzuki S et al, 2007, Synergistic action of the microRNA-17 poly-cistron and Myc in aggressive cancer development. Cancer Sci 98:1482–1490 Liang Z, Li Y, Huang K et al, 2011, Regulation of miR-19 to breast cancer chemoresistance through targeting PTEN. Pharm Res 28(12): 3091–3100 Ye H, Liu X, Lv M et al, 2012, MicroRNA and transcription factor co-regulatory network analysis reveals miR-19 inhibits CYLD in Tcell acute lymphoblastic leukemia. Nucleic Acids Res 40(12): 5201–5214 Baraniskin A, Kuhnhenn J, Schlegel U et al, 2011, Identification of microRNAs in the cerebrospinal fluid as marker for primary diffuse large B-cell lymphoma of the central nervous system. Blood 117(11): 3140–3146 Conkrite K, Sundby M, Mukai S et al, 2011, miR-17-92 cooperates with RB pathway mutations to promote retinoblastoma. Genes Dev 25(16):1734–1745 Zhang X, Yu H, Lou JR et al, 2011, MicroRNA-19, miR-19, regulates tissue factor expression in breast cancer cells. J Biol Chem 286(2):1429–1435 Mavrakis KJ, Wolfe AL, Oricchio E, Palomero T et al, 2010, Genome-wide RNA-mediated interference screen identifies miR- 19 targets in Notch-induced T-cell acute lymphoblastic leukaemia. Nat Cell Biol 12(4):372–379 Ping M, Han Y-C, Bete D et al, 2009, Genetic dissection of the miR-17-92 cluster of microRNAs in Myc-induced B-cell lymphomas. Genes Dev 23(24):2806–2811 Grillari J, Hackl M, Grillari-Voglauer R, 2010, miR-17-92 cluster: ups and downs in cancer and aging. Biogerontology 11(4):501–506 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 847 EP 853 DI 10.1007/s12253-013-9653-x PG 7 ER PT J AU Gasljevic, G Lamovec, J Contreras, AJ Zadnik, V Blas, M Gasparov, S AF Gasljevic, Gorana Lamovec, Janez Contreras, Antonio Juan Zadnik, Vesna Blas, Mateja Gasparov, Slavko TI HER2 in Gastric Cancer: An Immunohistochemical Study on Tissue Microarrays and the Coressponding Whole-Tissue Sections with a Supplemental Fish Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE FISH; Gastric carcinoma; HER2; Immunohistochemistry; TMA; Whole sections ID FISH; Gastric carcinoma; HER2; Immunohistochemistry; TMA; Whole sections AB Since focal HER2 expression is an issue in GC, TMA construction from the paraffin-embedded surgicallyobtained tissue may not reflect its real status. The aim of this study was to assess the HER2 status in tissue microarrays (TMAs) and the corresponding whole sections using HercepTest immunohistochemistry (IHC), and to correlate it and to assess the concordance of HER2 IHC and fluorescence in situ hybridization (FISH) in TMAs. Concordance of the HER2 expression status for 302 cases of gastric cancer using 9 paired TMAs was evaluated using a 2-mm core size and 305 corresponding whole sections. Concordance of the IHC and FISH HER2 status was compared. In addition,, the HER2 status was compared to clinicopathological characteristics and patients’ survival. Using the whole-section approach, HER2 over-expression was found in 25.2 % (HER2 3+ 6.6 %, HER2 2+ 18.7 %) of tumours. The overall concordance of IHC between the cores and the whole section was 84.9 %; 15.1 % of the tumours showed HER2 amplification. The overall concordance of IHC and FISH on cores was 75.7 %. The level of amplification correlated with the IHC score. Relationship between the intestinal and papillary types and tumour grade was observed for tumours with over-expression and amplification, whereas tumour location was related only to over-expression. There was a statistically significant difference in the overall survival of the patients, which was related to HER2 amplification. In conclusion, good concordance of the IHC HER2 results between tissue cores in TMA and whole sections, and excellent concordance of the IHC and FISH results on tissue cores was found. At least a part of the observed IHC HER2 heterogeneity could very likely be explained by fixation artifacts.With adequate fixation, a higher concordance of IHC HER2 between the cores and the whole sections can be expected. The TMA approach could enable an easier analysis of more than one representative tumour block. C1 [Gasljevic, Gorana] Institute of Oncology, Department of Pathology, Zaloska 2, 1000 Ljubljana, Slovenia. [Lamovec, Janez] Institute of Oncology, Department of Pathology, Zaloska 2, 1000 Ljubljana, Slovenia. [Contreras, Antonio Juan] Institute of Oncology, Department of Pathology, Zaloska 2, 1000 Ljubljana, Slovenia. [Zadnik, Vesna] Institute of Oncology, Cancer Registry, Zaloska 2, 1000 Ljubljana, Slovenia. [Blas, Mateja] Institute of Oncology, Department of Pathology, Zaloska 2, 1000 Ljubljana, Slovenia. [Gasparov, Slavko] “Merkur” University Hospital, Department of Clinical Pathology and Cytology, Zajceva 19, 10 000 Zagreb, Croatia. RP Gasljevic, G (reprint author), Institute of Oncology, Department of Pathology, 1000 Ljubljana, Slovenia. EM ggasljevic@onko-i.si CR Kamangar F, Dores GM, Anderson WF, 2006, Patterns of cancer incidence, mortality and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol 24:2137–2150 Cunningham SC, Kamangar F, Kim MP et al, 2005, Survival after gastric adenocarcinoma resection: eighteen-year experience at single institution. J Gastrointest Surg 9:725–728 Faycal J, Bessaguet C, Nousbaum JB et al, 2005, Epidemiology and long term survival of gastric carcinoma in the French district of Finestere between 1984 and 1995. Gastroenterol Clin Biol 29:23–32 Tsubono Y, Hisamichi S, 2000, Screening for gastric cancer in Japan. Gastric Cancer 3:9–18 Ohtsu A, 2008, Chemotherapy for metastatic gastric cancer: past, present and future. Gastroenterology 43:256–264 Menard S, Casalini P, Campiglio M, Pupa S, Agresti R, Tagliabue E, 2001, HER2 overexpresion in various tumor types, focusing on its relationship to the development of invasive breast cancer. Ann Oncol 12:S15–S19 Scholl S, Beuzeboc P, Pouillart P, 2001, Targeting HER2 in other tumor types. Ann Oncol 12:S81–S87 Ross JS, Slodkowska A, Symmans F, Pusztai L, Ravdin PM, Hortobagyi GN, 2009, The HER-2 receptor and breast cancer: ten years of targeted anti-HER-2 therapy and personalized medicine. Oncologist 14:320–368 Gravalos C, Jimeno A, 2008, HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol 19:1523–1529 Jǿrgensen JT, 2010, Targeted HER2 treatment in advanced gastric cancer. Oncology 78:26–33 Hofmann M, Stoss O, Shi D et al, 2008, Assessment of a HER2 scoring system for gastric cancer: results from a validation study. Histopathology 52:797–805 Allgayer H, Babic R, Gruetzner KU, Tarabichi A, Schildberg FW, Heiss MM, 2000, c-erbB-2 is of independent prognostic relevance in gastric cancer and is associated with the expression of tumorassociated protease systems. J Clin Oncol 18(11):2201–2209 Yano T, Toshihiko D, Ohtsu A et al, 2006, Comparison of HER2 gene amplification assessed by fluorescence in situ hybridization and HER2 protein expression assessed by immunohistochemistry in gastric cancer. Oncol Rep 15:65–71 Tanner M, Hollmen M, Junttila TT et al, 2005, Amplification of HER-2 in gastric carcinoma: association with Topoisomerase IIα gene amplification, intestinal type, poor prognosis and sensitivity to trastuzumab. Ann Oncol 16:273–278 Lordick F, Bang YJ, Kang YK et al., 2007, HER2-positive advanced gastric cancer: similar HER2 positivity levels to breast cancer. P3541, ECCO 14, Barcelona, Spain, September Ruschoff J, Dietel M, Baretton G et al, 2010, HER2 diagnostics in gastric cancer—guideline validation and development of standardized immunohistochemical testing. Virchows Arch 457(3):299–307 Park DI, Yun JW, Park JH et al, 2006, HER2/neu amplification is an independent prognostic factor in gastric carcinoma. Dig Dis Sci 51:1371–1379 Drev P, Frkovic-Grazio S, Bracko M, 2008, Tissue microarrays for routine diagnostic assessment of HER2 status in breast carcinoma. Appl Immunohistochem Mol Morphol 16(2):179–184 Grabsch H, Sivakumar S, Gray S, Gabbert HE, Muller W, 2010, HER2 expression in gastric cancer: rare, heterogeneous and of no prognostic value—conclusions from 924 cases of two independent series. Cell Oncol 32:57–65 Lee S, de Boer WB, Fermoyle S, Platten M, Kumarasinghe MP, 2011, Human epidermal growth factor receptor 2 testing in gastric carcinoma: issues related to heterogeneity in biopsies and resections. Histopathology 59:832–840 Kunz PL, Mojtahed A, Fisher G et al, 2012, HER2 expression in gastric and gastroesophageal junction adenocarcinoma in a US population: clinicopathologic analysis with proposed approach to HER2 assessment. Appl Immunohistochem Mol Morphol 20(1):13–24 Park SY, Hwang HS, Park HJ et al, 2012, Comprehensive analysis of HER2 expression and gene amplification in gastric cancers using immunohistochemistry and in situ hybridization: which scoring system should we use? Hum Pathol 43(3):413–422 Marx AH, Simon R, Sauter G, 2010, HER-2 amplification is highly homogeneous in gastric cancer-reply. Hum Pathol 41:305–306 Moelans CB, Milne AN, Morsink FH, Offerhaus GA, van Diest PJ, 2011, Low frequency of HER2 amplification and overexpression in early onset gastric cancer. Cell Oncol 34:89–95 Penault-Llorca F, Bilous M, Dowset M et al, 2009, Emerging technologies for assesing HER2 amplification. Am J Clin Pathol 132:539–548 Simon R, Mirlacher M, Sauter G, 2003, Tissue microarrays in cancer diagnosis. Expert Rev Mol Diagn 3:421–430 Sauter G, Mirlacher M, 2002, Tissue microarrys for predictive molecular pathology. J Clin Pathol 55:575–576 Bang YJ, Van Cutsem E, Feyereislova A et al, 2010, Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastroesophageal junction cancer, ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376:687–697 Bang YJ, Chung HC, Xu JM et al., 2009, Pathological features of advanced gastric cancer: relationship to human epidermal growth factor receptor 2 positivity in the global screeening programme of the ToGA trial. J Clin Oncol 27(Suppl.15s); Abstract 4556 Barros-Silva JD, Leitao D, Afonso L et al, 2009, Association of ERBB2 gene status with histopathological parameters and disease specific survival in gastric carcinoma patients. BJC 100:487–493 Kim MA, Jung EJ, Lee SH et al, 2007, Evaluation of HER2 gene status in gastric carcinoma using immunohistochemistry, fluorescence in situ hybridization and real-time quantitative polymerase chain reaction. Hum Pathol 38:1386–1393 Zheng HC, Li X, Hara T, Masuda S et al, 2008, Mixed-type gastric carcinomas exhibit more aggressive features and indicate the histogenesis of carcinomas. Virchows Arch 452:525– 534 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 855 EP 865 DI 10.1007/s12253-013-9654-9 PG 11 ER PT J AU Sikalidis, KA Fitch, DM Fleming, ESh AF Sikalidis, K Angelos Fitch, D Mark Fleming, E Sharon TI Risk of Colonic Cancer is Not Higher in the Obese Lepob Mouse Model Compared to Lean Littermates SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Leptin; Aberrant crypts; Tumors; Proliferation; Azoxymethane ID Leptin; Aberrant crypts; Tumors; Proliferation; Azoxymethane AB Epidemiological data suggest that obesity increases the risk of colorectal cancer in humans. Given that diet-induced obesity mouse models verified the epidemiological data, the present study aimed to determine whether obese C57BL/6J-Lepob male mice (a different obesity in vivo model) were at greater risk of colonic cancer than their lean male littermates. Risk of colonic tumorigenesis was assessed by numbers of aberrant crypts, aberrant crypt foci and colonic tumors. Proliferation of the colonic epithelia was assessed histochemically following administration of BrdU. Availability of the procarcinogen, azoxymethane (AOM) to target tissues was assessed by quantifying via HPLC plasma AOM concentrations during the 60 min period following AOM injection. When obese and lean mice were injected with azoxymethane (AOM) at doses calculated to provide equivalent AOM levels per kg lean body mass, obese animals had significantly fewer aberrant crypts/colon and fewer aberrant crypt foci/colon than the lean animals. Tumors were identified in the colonic mucosa of lean (4 tumors in 14 mice) but not obese (0 tumors in 15 mice) mice. Colonic cell proliferation was not significantly different for obese and lean mice. Because these results were unexpected, plasma AOM concentrations were measured and were found to be lower in the obese than lean mice. When plasma AOM levels were comparable for the lean and obese mice, the Lepob mice continued to have significantly fewer aberrant crypt foci/colon than the lean mice, but differences were not statistically different for aberrant crypts/colon. Interestingly, obese Lepob mice did not exhibit increased risk of colonic cancer as expected. Instead, Lepob mice exhibited equivalent or lower risk of colon cancer when compared to the lean group. These results taken together with in vivo results from diet-induced obesity studies, imply that leptin may be responsible for the increased risk of colon cancer associated with obesity. C1 [Sikalidis, K Angelos] University of California, Department of Nutritional Sciences and Toxicology, 94720 Berkeley, CA, USA. [Fitch, D Mark] University of California, Department of Nutritional Sciences and Toxicology, 94720 Berkeley, CA, USA. [Fleming, E Sharon] University of California, Department of Nutritional Sciences and Toxicology, 94720 Berkeley, CA, USA. RP Sikalidis, KA (reprint author), University of California, Department of Nutritional Sciences and Toxicology, 94720 Berkeley, USA. EM angelos_sikalidis@dfci.harvard.edu CR Caan BJ, Coates AO, Slattery ML, Potter JD, Quesenberry CP Jr, Edwards SM(1998, Body size and the risk of colon cancer in a large case–control study. Int J Obes Relat Metab Disord 22(2):178–184 Boyle T, Keegel T, Bull F, Heyworth J, Fritschi L, 2012, Physical activity and risks of proximal and distal colon cancers: a systematic review and meta-analysis. J Natl Cancer Inst 104(20):1548–1561 Kono S, Handa K, Hayabuchi H, Kiyohara C, Inoue H et al, 1999, Obesity, weight gain and risk of colon adenomas in Japanese men. Jpn J Cancer Res 90(8):805–811 Suh S, Kang M, Kim MY, Chung HS, Kim SK et al, 2011, Korean type 2 diabetes patients have multiple adenomatous polyps compared to non-diabetic controls. J Korean Med Sci 26(9):1196–1200 Wolin KY, Yan Y, Colditz GA, 2011, Physical activity and risk of colon adenoma: a meta-analysis. Br J Cancer 104(5):882–885 Ma Y, Yang Y,Wang F, Zhang P, Shi C et al, 2013, Obesity and risk of colorectal cancer: a systematic review of prospective studies. PLoS One 8(1):e53916 Maruthur NM, Bolen S, Gudzune K, Brancati FL, Clark JM, 2012, Body mass index and colon cancer screening: a systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev 21(5):737–746 Ford ES, 1999, Body mass index and colon cancer in a national sample of adult US men and women. Am J Epidemiol 150(4):390–398 Murphy TK, Calle EE, Rodriguez C, Khan HS, Thun MJ, 2000, Body mass index and colon cancer mortality in a large prospective study. Am J Epidemiol 152(9):847–854 Gerhardsson de Verdier M, Hagman U, Steineck G, Rieger A, Norell SE, 1990, Diet, body mass and colorectal cancer: a casereferent study in Stockholm. Int J Cancer 46(5):832–838 Bostick RM, Potter JD, Kushi LH, Sellers TA, Steinmetz KA et al, 1994, Sugar, meat, and fat intake, and non-dietary risk factors for colon cancer incidence in Iowa women, United States). Cancer Causes Control 5(1):38–52 Yu XF, Wang YQ, Zou J, Dong J, 2012, A meta-analysis of the effects of energy intake on risk of digestive cancers. World J Gastreneterol 18(48):7362–7370 Bergstrom A, Pisani P, Tenet V, Wolk A, Adami HO, 2001, Overweight as an avoidable cause of cancer in Europe. Int J Cancer 91(3):421–430 Tashiro M, Akiyama T, Yoshikawa I, Kume K, Otsuki M, 2004, Obesity as a risk factor for colorectal polyps in Japanese patients. Gut 53(1):156 Lee WM, Lu S, Medline A, Archer MC, 2001, Susceptibility of lean and obese Zucker rats to tumorigenesis induced by N-methyl- N-nitrosourea. Cancer Lett 162(2):155–160 Hakkak R, Holley AW, Macleod SL, Simpson PM, Fuchs GJ et al, 2005, Obesity promotes 7,12-dimethylbenz(a)anthracene-induced mammary tumor development in female zucker rats. Breast Cancer Res 7(5):R627–R633 Weber RV, Stein DE, Scholes J, Kral JG, 2000, Obesity potentiates AOM-induced colon cancer. Dig Dis Sci 45(45):890–895 Hata K, Kubota M, Shimizu M, Moriwaki H, Kuno T et al, 2011, C57BL/KsJ-db/db-Apc Mice Exhibit an Increased Incidence of Intestinal Neoplasms. Int J Mol Sci 12(11):8133–8145 Sikalidis AK, Fitch MD, Fleming SE, 2013, diet induced obesity increases the risk of colonic tumorigenesis in mice. Pathol Oncol Res., DOI 10.1007/s12253-013-9626-0 Wolter S, Frank N, 1982, Metabolism of 1,2-dimethylhydrazine in isolated perfused rat liver. Chem Biol Interact 42(3):335–344 Bird RP, 1987, Observation and quantification of aberrant crypts in the murine colon treated with a colon carcinogen: preliminary findings. Cancer Lett 37(2):147–151 Lacy ER, Kuwayama H, Cowart KS, King JS, Deutz AH, Sistrunk S, 1991, A rapid, accurate, immunohistochemical method to label proliferating cells in the digestive tract. A comparison with tritiated thymidine. Gastroenterology 100(1):259–262 Park CM, Reid PE, Walker DC, MacPherson BR, 1987, A simple, practical ‘swiss roll’ method of preparing tissues for paraffin or methacrylate embedding. J Microsc 145(1):115–120 Chaudhary M, Mandir N, Fitzgerald AJ, Howard JK, Lord GM et al, 2000, Starvation, leptin and epithelial cell proliferation in the gastrointestinal tract of the mouse. Digestion 61(4):223–229 Meier U, Gressner AM, 2004, Endocrine regulation of energy metabolism: review of pathobiochemical and clinical chemical aspects of leptin, ghrelin, adiponectin, and resistin. Clin Chem 50(9):1511– 1525 Kuate D, Etoundi BC, Azantsa BK, Kengne AP, Ngondi JL, Oben JE, 2010, The use of LeptiCore in reducing fat gain and managing weight loss in patients with metabolic syndrome. Lipids Health Dis 9:20 Procaccini C, Jirillo E, Matarese G, 2012, Leptin as an immunomodulator. Mol Aspects Med 33(1):35–45 Harle P, Straub RH, 2006, Leptin is a link between adipose tissue and inflammation. Ann NYAcad Sci 1069:454–462 La Cava A, Alviggi C, Matarese G, 2003, Unraveling the multiple roles of leptin in inflammation and autoimmunity. J Mol Med, Berl, 82(1):4–11 Zhou Z, Neupane M, Zhou HR, Wu D, Chang CC, et al., 2012, Leptin differentially regulate STAT3 activation in ob/ob mouse adipose mesenchymal stem cells. Nutr Metab, Lond, 9(1):109 Mykoniatis A, Anton PM, Wlk M, Wang CC, Ungsunan L et al, 2003, Leptin mediates Clostridium difficile toxin A-induced enteritis in mice. Gastroenterology 124(3):683–691 Busso N, So A, Chobaz-Peclat V, Morard C, Martinez-Soria E et al, 2002, Leptin signaling deficiency impairs humoral and cellular immune responses and attenuates experimental arthritis. J Immunol 168(2):875–882 Siegmund B, Lehr HA, Fantuzzi G, 2002, Leptin: a pivotal mediator of intestinal inflammation in mice. Gastroenterology 122(7):2011– 2025 Mackey-Lawrence NM, Petri WA Jr, 2012, Leptin and mucosal immunity. Mucosal Immunol 5(5):472–479 Mantovani A, 2010, Molecular pathways linking inflammation and cancer. Curr Mol Med 10(4):369–373 Germano G, Allavena P, Mantovani A, 2008, Cytokines as a key component of cancer-related inflammation. Cytokine 43(3):374–379 Greten FR, Eckmann L, Greten TF, Park JM, Li ZW et al, 2004, IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 118(30):285–296 Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S et al, 2004, NF-kappaB functions as a tumour promoter in inflammationassociated cancer. Nature 431(7007):461–466 Somasundar P, Riggs D, Jackson B, Vona-Davis L, McFadden DW, 2003, Leptin stimulates esophageal adenocarcinoma growth by nonapoptotic mechanisms. Am J Surg 186(5):575–578 Watanabe N, Takai K, Imai K, Shimizu M, Naiki T et al, 2011, Increased levels of serum leptin are a risk factor for the recurrence of stage I/II hepatocellular carcinoma after curative treatment. J Clin Biochem Nutr 49(3):153–158 Drew JE, 2012, Molecular mechanisms linking adipokines to obesityrelated colon cancer: focus on leptin. Proc Nutr Soc 71(1):175–180 Hardwick JC, Van Den Brink GR, Offerhaus GJ, Van Deventer SJ, Peppelenbosch MP, 2001, Leptin is a growth factor for colonic epithelial cells. Gastroenterology 121(1):79–90 Padidar S, Farguharson AJ, Williams LM, Kearney R, Arthur JR, Drew JE, 2012, High-fat diet alters gene expression in the liver and colon: links to increased development of aberrant crypt foci. Dig Dis Sci 57(7):1866–1874 Hoda MR, Theil G, Mohammed N, Fischer K, Fornara P, 2012, The adipocyte-derived hormone leptin has proliferative actions on androgen-resistant prostate cancer cells linking obesity to advanced stages of prostate cancer. J Oncol 2012:280386 Nachat-Kappes R, Pinel A, Combe K, Lamas B, Farges MC et al, 2012, Effects of enriched environment on COX-2, leptin and eicosanoids in a mouse model of breast cancer. PLoS One 7(12):e51525 Hu X, Juneja SC, Maihle NJ, Cleary MP, 2002, Leptin-a growth factor in normal and malignant breast cells and for normal mammary gland development. J Natl Cancer Inst 94(22):1704– 1711 Jaffe T, Schwartz B, 2008, Leptin promotes motility and invasiveness in human colon cancer cells by activating multiple signaltransduction pathways. Int J Cancer 123(11):2543–2556 Iversen PO, Drevon CA, Reselnd JE, 2002, Prevention of leptin binding to its receptor suppresses rat leukemic cell growth by inhibiting angiogenesis. Blood 100(12):4123–4128 Hirose Y, Hata K, Kuno T, Yoshida K, Sakata K et al, 2004, Enhancement of development of azoxymethane-induced colonic premalignant lesions in C57BL/KsJ-db/db mice. Carcinogenesis 25(5):821–825 Aparicio T, Guilmeau S, Goiot H, Tsocas A, Laigneau JP et al, 2004, Leptin reduces the development of the initial precancerous lesions induced by azoxymethane in the rat colonic mucosa. Gastroenterology 126(2):499–510 Sikalidis AK, Varamini B, 2011, Roles of hormones and signaling molecules in describing the relationship between obesity and colon cancer. Pathol Oncol Res 17(4):785–790 Franco A, Sikalidis AK, Solis Herruzo JA, 2005, Colorectal cancer: influence of diet and lifestyle factors. Rev Esp Enferm Dig 97(6):432– 448 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 867 EP 874 DI 10.1007/s12253-013-9656-7 PG 8 ER PT J AU Bubis, G Hilly, O Bubis, R Halpern, M Schwartz, A Koren, R Rath-Wolfson, L AF Bubis, Golan Hilly, Ohad Bubis, Roy Halpern, Marisa Schwartz, Ariel Koren, Rumelia Rath-Wolfson, Lea TI A New Ki-67 / E-Cadherin Cocktail Reduces Inter-observer Variation of the Calculated Proliferative Index SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE Ki-67; E-Cadherin; Cocktail immunostain; Proliferative index; Infiltrating duct carcinoma of breast ID Ki-67; E-Cadherin; Cocktail immunostain; Proliferative index; Infiltrating duct carcinoma of breast AB The proliferative index in breast carcinoma is usually calculated by the percentage of the Ki-67 positive cells out of the total number of malignant cells. In order to reduce the inter-observer variability of the calculated proliferative index a cocktail of antibodies against E-Cadherin and Ki-67 (Ki/Cad Cocktail) is presented. The cocktailwas applied on 59 cases of infiltrating duct carcinoma of breast and compared to the consecutive slides stained for Ki-67 alone. The Ki/Cad cocktail has the advantage that by adding the anti E-Cadherin antibody, all the malignant epithelial cells are highlighted and can be differentiated from other proliferating cells. Statistical analysis proved that the cocktail increases the inter-observer agreement from 89 % to 97 % as compared to the Ki-67 alone and also reduces the overlap between the cancer grades. C1 [Bubis, Golan] Hadassah University, Department of BiologyJerusalem, Israel. [Hilly, Ohad] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Otolaryngology-Head and Neck SurgeryTel Aviv, Israel. [Bubis, Roy] Tel-Aviv University, Raymond and Beverly School of Physics and AstronomyTel Aviv, Israel. [Halpern, Marisa] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of PathologyTel Aviv, Israel. [Schwartz, Ariel] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of PathologyTel Aviv, Israel. [Koren, Rumelia] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of PathologyTel Aviv, Israel. [Rath-Wolfson, Lea] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of PathologyTel Aviv, Israel. RP Hilly, O (reprint author), Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of Otolaryngology-Head and Neck Surgery, Tel Aviv, Israel. EM hillyo@clalit.org.il CR DeSantis C, Siegel R, Bandi P, Jemal A, 2011, Breast cancer statistics, 2011. CA Cancer J Clin 61:409–418., DOI 10.3322/caac.20134 Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA, 2010, Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol 11:174–183 Assersohn L, Salter J, Powles TJ, A’hern R, Makris A, Gregory RK, Chang J,DowsettM(2003, Studies of the potential utility ofKi67 as a predictive molecular marker of clinical response in primary breast cancer. Breast Cancer Res Treat 82:113–123 Urruticoechea A, Smith IE, Dowsett M, 2005, Proliferation marker Ki-67 in early breast cancer. J Clin Oncol 23:7212–7220 Lindboe CF, Torp SH, 2002, Comparison of Ki-67 equivalent antibodies. J Clin Pathol 55:467–471 Cooper LS,Gillett CE, Smith P, Fentiman IS, Barnes DM(1998, Cell proliferation measured by MIB1 and timing of surgery for breast cancer. Br J Cancer 77:1502–1507 ThorAD, Liu S,MooreDH2nd, Edgerton SM(1999)Comparison of mitotic index, in vitro bromodeoxyuridine labeling, and MIB-1 assays to quantitate proliferation in breast cancer. J Clin Oncol 17:470–477 Rudolph P, Alm P, Heidebrecht HJ, Bolte H, Ratjen V, Baldetorp B, FernoM, Olsson H, Parwaresch R, 1999, Immunologic proliferation marker Ki-S2 as prognostic indicator for lymph node-negative breast cancer. J Natl Cancer Inst 91:271–278 Mengel M, von Wasielewski R, Wiese B, Rudiger T, Muller- Hermelink HK, Kreipe H, 2002, Inter-laboratory and inter-observer reproducibility of immunohistochemical assessment of the Ki-67 labelling index in a large multi-centre trial. J Pathol 198:292–299 DowsettM,Nielsen TO,A’Hern R, Bartlett J, CoombesRC, Cuzick J, EllisM, Henry NL, Hugh JC, Lively T, McShane L, Paik S, Penault- Llorca F, Prudkin L, ReganM, Salter J, Sotiriou C, Smith IE, Viale G, Zujewski JA, Hayes DF, 2011, International Ki-67 in Breast Cancer Working Group. Assessment of Ki67 in breast cancer: recommendations fromthe InternationalKi67 inBreastCancerworking group. J Natl Cancer Inst 103:1656–1664 Gal R, Halpern M, Gertzmann H, Schwartz A, Rath-Wolfson L, Koren R, 2011, Ki-67 & E-Cadherin cocktail: an immunochemical stain for accurate estimation of the proliferative index in infiltrating duct carcinoma of breast. Med Con 4:9–11 Bland JM, Altman DG, 1986, Statistical methods for assessing agreement between two methods of clinical measurement. Lancet 1:307–310 Landis JR,KochGG(1977, The measurement of observer agreement for categorical data. Biometrics 33:159–174 Yaziji H, Eisen R, Wick M, Badve S, Cartun R, Haas T, Marolt M, Hicks D,Martin A, Barry T, Alsabeh R, Taylor J, Fulton R, Goldsmith J, Shen S, Taylor C, Swanson P, 2012, Immunohistochemistry cocktails are here to stay: Center for Medicare and Medicaid Services should revise its new reimbursement policy. Am J Clin Pathol 138:10– 11 Orchard G, 2002, Evaluation of melanocytic neoplasms: application of a pan-melanoma antibody cocktail. Br J Biomed Sci 59:196–202 Sanderson SO, Sebo TJ,Murphy LM, NeumannR, Slezak J,Cheville JC, 2004, An analysis of the p63/alpha-methylacyl coenzyme A racemase immunohistochemical cocktail stain in prostate needle biopsy specimens and tissuemicroarrays. Am J Clin Pathol 121:220– 225 Tacha DE,Miller RT, 2004, Use of p63/P504S monoclonal antibody cocktail in immunohistochemical staining of prostate tissue. Appl Immunohistochem Mol Morphol 12:75–78 Hameed O, Sublett J, Humphrey PA, 2005, Immunohistochemical stains for p63 and alpha-methylacyl-CoA racemase, versus a cocktail comprising both, in the diagnosis of prostatic carcinoma: a comparison of the immunohistochemical staining of 430 foci in radical prostatectomy and needle biopsy tissues. Am J Surg Pathol 29:579–587 Jiang Z, Li C, Fischer A, Dresser K, Woda BA, 2005, Using an AMACR, P504S)/34betaE12/p63 cocktail for the detection of small focal prostate carcinoma in needle biopsy specimens. Am J Clin Pathol 123:231–236 Qureshi HS, Linden MD, Divine G, Raju UB, 2006, E-cadherin status in breast cancer correlates with histologic type but does not correlate with established prognostic parameters. Am J Clin Pathol 125:377–385 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2013 VL 19 IS 4 BP 875 EP 879 DI 10.1007/s12253-013-9655-8 PG 5 ER PT J AU Robert, M Robert, L AF Robert, A. Matthieu Robert, Ladislas TI Xanthine Oxido-Reductase, Free Radicals and Cardiovascular Disease. A Critical Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Xanthine oxidase; Free radicals; ROS; Cardiovascular disease; Allopurinol ID Xanthine oxidase; Free radicals; ROS; Cardiovascular disease; Allopurinol AB Free radical mediated pathologies occupy a special place in medical semiology and in mechanistic interpretation of diseases. Free radicals, or better reactive oxygen species (ROS) or reactive nitrogen species (RNS) play also an important role in cell signaling. This is the basis of the ambivalent (Jekyll – Hyde) situation of ROS in biology and pathology. Aging itself is attributed by a popular theory to free radicals. A number of ROS – scavenging substances and procedures were described without however reaching credibility for their therapeutic value. An interesting exception is the xanthine oxido - reductase produced ROS and their role in cardiovascular disease. Allopurinol inhibition of xanthine oxido – reductase was shown to be efficient in some cases of cardiovascular diseases. Another important aspect of xanthine oxido – reductase produced ROS is their antibacterial capacity considered to be of importance with newborns fed on milk rich in this enzyme as well as at the gastrointestinal barrier. This ambivalent role of xanthine oxido – reductase justifies this review on the basic enzymatic mechanisms involved, derived ROS production, their role in the abovementioned biological processes and especially the interest of the inhibition of this enzyme as a preventive or curative measure in some cardiovascular pathologies. C1 [Robert, A. Matthieu] Hopital Hotel Dieu, Laboratoire de Recherche Ophtalmologique, 7 rue Jean-Baptiste Lully, 94440 Paris, France. [Robert, Ladislas] Hopital Hotel Dieu, Laboratoire de Recherche Ophtalmologique, 7 rue Jean-Baptiste Lully, 94440 Paris, France. RP Robert, L (reprint author), Hopital Hotel Dieu, Laboratoire de Recherche Ophtalmologique, 94440 Paris, France. EM lrobert5@orange.fr CR Robert L, Labat-Robert J, Robert AM, 2010, Vieillissement cellulaire, telomeres et maladies liees a l’age. Medecine & Longevite 2:151–161., DOI 10.1016/j.mlong.2010.07.005 Harman D, 1955, Aging — a theory based on free radical and information theory U.C.R.L. Publ., 3078, Univ. of Calif. Comfort A, 1979, The biology of senescence, 3rd edn. Churchill Livingstone, Edinburgh & London, 1979 Emerit I, Eds BC, 1992, Free radicals and aging. Birkhauser Verlag, Basel, 1992 Edeas M, 2009, Anti-oxydants, controverses et perspectives: comment expliquer l’echec des etudes cliniques utilisant des antioxydants. J Soc Biol 203:271–280 Blokhina O, Virolainen E, Fagerstedt KV, 2003, Antioxidants, oxidative damage and oxygen deprivation stress: a review. Ann Bot 91: 179–194 Martin HM, Hancock JT, Salisbury V, Harrison R, 2004, Minireview. Role of xanthine oxydoreductase as an antimicrobial agent. Infect Immun 72:4933–4939 Miller SL, Urey HC, 1959, Organic compound synthesis on the primitive earth. Science 130:245–251 Garattini E, Mendel R, Romao MJ, Wright R, Terao M, 2003, Review article. Mammalian molybdo-flavoenzymes, an expanding family of proteins: structure, genetics, regulation, function and pathophysiology. Biochem J 372:15–32 Drury AN, Szent-Gyorgyi A, 1929, The physiological activity of adenine compounds with especial reference to their action upon the mammalian heart. J Physiol 68:213–237 Eltzschig HK, Sitkovsky MV, Robson SC, 2012, Purinergic signaling during inflammation. New Engl J Med 367:2322–2333 Patton S, Keenan TW, 1975, The milk fat globule membrane. Biochim Biophys Acta 415:273–309 Dowben RM, Brunner JR, Philpott DE, 1967, Studies on milk fat globule membranes. Biochim Biophys Acta 135:1–10 Polonovski M, Robert M, Robert L, 1950, Cinetique de la revelation de la xanthinedehydrase du lait de vache. Bull Soc Chim Biol 32:862–867 Polonovski M, Baudu L, Robert M, Robert L, 1950, Donnees nouvelles sur la revelation de la xanthine-dehydrase du lait par les agents chimiques. Bull Soc Chim Biol 32:855–861 Polonovski M, Robert L, Robert M, 1950, Une nouvelle methode d’extraction et de purification de la xanthine dehydrase du lait. Bull Soc Chim Biol 32:868–871 Robert L, Basset J, 1953, Action des hautes pressions sur la xanthinedehydrase du lait. Bull Soc Chim Biol 35:1375–1380 Robert L, Nolla N, 1953, Action des ultrasons sur la xanthinedehydrase du lait. Bull Soc Chim Biol 35:1363–1373 Robert L, Polonovski M, 1955, Activation and inactivation of milk xanthineoxydase by physicochemical means. In: Discussions of the Faraday Society, N° 20, The Physical Chemistry of Enzymes The Aberdeen University Press Ltd; 6 Upper Kirkgate, Aberdeen, pp 54–65 Avis PG, Berger F, Bray RC, Shooter KV, 1954, A crystalline material with xanthine oxidase activity. Nature 173: 1230–1233 Avis PG, Bergel F, Bray RC, 1955, Cellular constituents. The chemistry of xanthine oxidase. Pat I. The preparation of a crystalline xanthine oxidase from cow’s milk. J Chem Soc April 1100–1105 Avis PG, Bergel F, Bray RC, James DWF, Shooter KV, 1956, Cellular constituents. The chemistry of xanthine oxidase. Part II. The homogeneity of crystalline metalloflavoprotein fractions. J Chem Soc 252:1212–1219 Avis PG, Bergel F, Bray RC, 1956, Cellular constituents. The chemistry of xanthine oxidase. Part III. Estimations of the co-factors and the catalytic activities of enzyme fractions from cow’s milk. J Chem Soc 253:1219–1226 Bergel F, Bray RC, 1959, The chemistry of xanthine oxidase. 4. The problems of enzyme inactivation and stabilisation. Biochem J 73: 182–192 Bray RC, Chisholm AJ, Hart LI, Meriwether LS, Watts DC, 1966, Studies on the composition andmechanismof action ofmilk xanthine oxidase. In: Slater EC, ed, Flavins and flavoproteins. Elsevier Publishing Company, Amsterdam, pp 117–132 Haddow A, de Lamirande G, Bergel F, Bray RC, Gilbert DA, 1958, Anti-tumour and biochemical effects of purified bovine xanthine oxidase in C3H and C mice. Nature 182:1144–1146 Mackler B, Mahler HR, Green DE, 1954, Studies on metalloflavoproteins. I. Xanthine oxydase. A molybdoflavoprotein. J Biol Chem 210:149–164 Berry CE, Hare JM, 2004, Xanthine oxidoreductase and cardiovascular disease: molecular mechanisms and pathophysiological implications. J Physiol 555:589–606 Granger DN, Rutili G,McCord J, 1981, Superoxide radicals in feline intestinal ischemia. Gastroenterology 81:22–29 Carden DI, Granger DN, 2000, Pathology of ischemia-reperfusion injury. J Pathol 190:255–266 Harrison R, 2002, Structure and function of xanthine oxidoreductase: where are we now? Free Radic Biol Med 33:774–797 Granger DN, Rutili G, McCord JM, 1981, Superoxide radicals in feline intestinal ischemia. Gastroenterology 81:22–29 Abadeh S, Killacky J, Benboubetra M, Harrison R, 1992, Purification and partial characterization of xanthine oxidase from human milk. Biochim Biophys Acta 1117:25–32 Vorbach C, Scriven A, Capecchi MR, 2002, The housekeeper gene xanthine oxidoreductase is necessary for milk fat droplet enveloping and secretion: gene sharing in the lactating mammary gland. Genes Dev 16:3223–3235 Simmonds HA, 1994, Purine and pyrimidine disorders. In: Holton JS, ed, The Inherited metabolic diseases, 2nd edn. Churchill Livingstone, New York, pp 297–307 Delbarre F, Cartier P, Auscher C, De Gery A, HametM(1970, Gouttes enzymopathiques. Dyspurinies par deficit en Hypoxanthine-Guanine- Phosphoribosyl-Transferase. Frequence et caracteres cliniques de l’anenzymose. Presse Med 78:729–734 Ea H-K, Bardin T, Jinnah HA, Aral B, Liote F, Ceballos - Picot I, 2009, Severe Gouty Arthritis and mild neurologic symptoms due to F199C, a newly identified variant of the hypoxanthine guanine phosphoribosyltransferase. Arthritis Rheum 60:2201–2220 Lewers JC, Ceballos-Picot I, Shirley TL,Mockel L, Egami K, Jinnah HA, 2008, Consequences of impaired purine recycling in dopaminergic neurons. Neuroscience 152:761–772 Bollee G, Dollinger C, Boutaud L, Guillemot D, Bensman A, Harambat J, Deteix P, Daudon M, Knebelmann B, Ceballos-Picot I, 2010, Phenotype and genotype characterization of adenine phosphoribosyltransfetrase deficiency. J Am Soc Nephrol 4:679–688 Jinnah HA, Ceballos-Picot I, Torres RJ, Visser JE, Schretlen DJ, Verdu A, Larovere LE, Chen S-J, Cossu A et al, 2010, Attenuated variants of Lesch-Nyhan disease. Brain 133:671– 689 Li H, Samouilov A, Liu X, Zweier JL, 2003, Characterization of the magnitude and kinetics of xanthine oxidase – catalysed nitrate reduction: evaluation of its role in nitrite and nitric oxide generation in anoxic tissues. Biochemistry 42:1150–1159 Kayyali US, Donaldson C, Huang H, Abdelnour R, Hassoun PM, 2001, Phosphorylation of xanthine dehydrogenase/oxidase in hypoxia. J Biol Chem 276:14359–14365 Adachi T, Fukushima T, Usami Y, Hirano K, 1993, Binding of human xanthine oxidase to sulphated glycosaminoglycans on the endothelial cell surface. Biochem J 289:523–527 Brown JM, Terrada LS, GrossoMA,Whitmann GJ, Velasco SE, Patt A, Harken AH, Repine JE, 1988, Xanthine oxidase produces hydrogen peroxide which contributes to reperfusion injury of ischemic, isolated, perfused rat hearts. J Clin Invest 81:1297–1301 Wang W, Sawicki G, Schulz R, 2002, Peroxinitrite – induced myocardial injury is mediated through matrix metalloproteinase-2. Cardiovasc Res 53:165–174 Wang W, Schulze CJ, Suarez-Pinzon WL, Dyck JRB, Sawicki G, Schulz R, 2002, Intracellular action of matrix metalloproteinase - 2 accounts for acute myocardial ischemia and reperfusion injury. Circulation 106:1543–1549 Campbell DL, Stamler JS, Strauss HC, 1996, Redox modulation of L-type calcium channels in ferret ventricular myocytes dual mechanism regulation by nitric oxide and S-nitrosothiols. J General Physiol 108:277–293 Ohara Y, Peterson TE, Harrison DG, 1993, Hypercholesterolemia increases endothelial superoxide anion production. J Clin Invest 91: 2546–2551 Burns CM,Wortmann RL, 2012, Latest evidence on gout management: what the clinician needs to know. Ther Adv Chronic Dis 3(6):271–286 Truglio JJ, Theis K, Leimkuhler S, Rappa R, Rajagopalan KV, Kisker C, 2002, Crystal structures of the active and alloxanthine-inhibited forms of xanthine dehydrogenase from Rhodobacter capsulatus. Structure 10:115–125 Sanders S, Eisenthal RS, Harrison R, 1997, NADH oxidase activity of human xanthine oxidoreductase –generation of superoxide anion. Eur J Biochem 245:541–548 Brown JM, Terada LS, Grosso MA,Whitmann GJ, Velasco SE, Patt A, Harken AH, Repine JE, 1988, Xanthine oxidase produces hydrogen peroxide which contributes to reperfusion injury of ischemic, isolated perfused rat hearts. J Clin Invest 81:1297–1301 Grisham MB, Hernandez LA, Granger DN, 1986, Xanthine oxidase and neutrophil infiltration in intestinal ischemia. Am J Physiol 251: G567–G574 Gimpel JA, Lahpor JR, van der Molen A, Damen J, Hitchcock JF, 1995, Reduction of reperfusion injury of human myocardium by allopurinol: a clinical study. Free Radic Biol Med 19:251–255 Guan W, Osanai T, Kamada T, Hanada H, Ishizaka H, Onodera H, IwasaA, Fujita N, Kudo S,OhkuboTOK, 2003, Effect of allopurinol pretreatment on free radical generation after primary coronary angioplasty for acute myocardial infarction. J Cardiovasc Pharmacol 41: 699–705 Butler R, Morris AD, Belch JJF, Hill A, Struthers AD, 2000, Allopurinol normalizes endothelial dysfunction in type 2 diabetics with mild hypertension. Hypertension 35:746–751 Nakazono K, Watanabe N, Matsuno K, Sasaki J, Sato T, Iwasaki T, 2003, Does superoxide underlie the pathogenesis of hypertension? Proc Natl Acad Sci USA 88:10045–10048 Kogler H, Fraser H, McCune S, Altschuld R, Marban E, 2003, Disproportionate enhancement of myocardial contractility by the xanthine oxidase inhibitor oxypurinol in failing rat myocardium. Cardiovasc Res 59:582–592 Cappola TP, Kass DA, Nelson GS, Berger RD, Rosas GO, Kobeissi ZA, Marban E, Hare JM, 2001, Allopurinol improves myocardial efficiency in patients with idiopathic dilated cardiomyopathy. Circulation 104:2407–2411 Feig DI, Kang DH, Johnson RJ, 2008, Uric acid and cardiovascular risk. N Engl J Med 359:1811–18 21 Galiardi AC, Miname MH, Santos RD, 2009, Uric acid: a marker of increased cardiovascular risk. Atherosclerosis 202:11–17 Krishnan E, Baker JF, Furst DE, Schumacher HR, 2006, Gout and the risk of acute myocardial infarction. Arthritis Rheum 54:2688– 2696 Krishnan E, Pandya BJ, Lingala B, Hariri A, Dabbous O, 2012, Hyperuricemia and untreated gout are poor prognostic markers among those with a recent acute myocardial infarction. Arthritis Res Ther 14:R10 Baker JF, Schumacher HR, Krishnan E, 2007, Serum uric acid level and risk for peripheral arterial disease: analysis of data from the multiple risk factor intervention trial. Angiology 58:450–457 Krishnan E, Svendsen K, Neaton JD, Grandits G, Kuller RH, 2008, Long-term cardiovascular mortality among middle – aged men with gout. Arch Intern Med 168:1104–1110 Choi HK, Curhan G, 2007, Independent impact of gout on mortality and risk for coronary heart disease. Circulation 116:894–900 Hanassoulis G, Brophy JM, Richard H, Pilote L, 2010, Gout, allopurinol use and heart failure outcomes. Arch Intern Med 170:1358–1364 Noman A, Ang DS, Ogston S, Lang CC, Struthers AD, 2010, Effect of high - dose allopurinol on exercise in patients with chronic stable angina: a randomized, placebo controlled crossover trial. Lancet 375:2161–2167 Feig DI, Soletsky B, Johnson RJ, 2008, Effect of allopurinol on blood pressure of adolescents with newly diagnosed essential hypertension: a randomized trial. JAMA 300:924–932 Kelkar A, Kuo A, Frishman WH, 2011, Allopurinol as a cardiovascular drug. Cardiol Rev 19:265–271 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 1 EP 10 DI 10.1007/s12253-013-9698-x PG 10 ER PT J AU Bittner, N Ostoros, Gy Geczi, L AF Bittner, Nora Ostoros, Gyula Geczi, Lajos TI New Treatment Options for Lung Adenocarcinoma - in View of Molecular Background SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Non-small cell lung cancer (NSCLC); Adenocarcinoma; Targeted therapies; Signal transduction pathway; Tyrosine kinase inhibitors; Monoclonal antibodies; EGFR mutation; KRAS; EML-4ALK; VEGFR ID Non-small cell lung cancer (NSCLC); Adenocarcinoma; Targeted therapies; Signal transduction pathway; Tyrosine kinase inhibitors; Monoclonal antibodies; EGFR mutation; KRAS; EML-4ALK; VEGFR AB Lung cancer is the leading cause of cancer related mortality all over the world, and a number of developments have indicated future clinical benefit recently. The development of molecular pathology methods has become increasingly important in the prediction of chemotherapy sensitivity and mutation analysis to identify driver mutations as important targets of new therapeutic agents. The most significant changes in the treatment of NSCLC revealed in new pathologic classification and in the introduction of molecularly targeted therapies, which include monoclonal antibodies and small molecule tyrosine kinase inhibitors. The side effects of these agents are generally better tolerated than those of conventional chemotherapy and show higher efficacy. The most important factor follows: histology subtypes, gene mutation status, patients’ selection, drug toxicities and occurence of drug resistance. In the advanced disease, the hope of cure is less than 3 %, but improvements in survival have been clearly achieved. Some years ago the median lung cancer survival rate was 10–12 months, now in case of available specific molecular targets, a significant increase in median survival rates to 24–36 months has been achieved. These agents give an opportunity to provide a new standard of care. Therefore testing EGFR mutations and ALK rearrangements in patients with advanced lung adenocarcinoma should be incorporated into routine clinical practice. This review focuses on the rationale for targeted agents and new treatment possibilities in case of advanced lung adenocarcinoma. C1 [Bittner, Nora] National Institute of OncologyBudapest, Hungary. [Ostoros, Gyula] National Koranyi Institute of PulmonologyBudapest, Hungary. [Geczi, Lajos] National Institute of OncologyBudapest, Hungary. RP Bittner, N (reprint author), National Institute of Oncology, Budapest, Hungary. EM nora_bittner@yahoo.ca CR Travis WD, Brambilla E et al, 2011, International association for the study of lung cancer/American thoracic society/European respiratory society international multidisciplinary classification of lung adenocarcinoma. J Thoracic Oncol 6:2244–2285 Mino-Kenudson M et al, 2010, A novel, high sensitive antibody allows for the routine detection of ALK – rearranged lung adenocarcinomas by standard immunohistochemistry. Clin Cancer Res 16: 1561–1571 Shiller JH, 2002, Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 2:92–98 NSCLC Meta –Analysis Collaborative Group, 2008, Chemotherapy in addition to supportive care improves survival in advanced nonsmall- cell lung cancer: a systematic review and meta-analysis of individual patient data from 16 randomised controlled trials. J Clin Oncol 28:4617–4625 Azolli CG, 2009, American society of clinical oncology clinical practice guideline update on chemotherapy for stage IV non-smallcell lung cancer. J Clin Oncol 36:6251–6266 Ettinger DS, 2010, Non-small cell lung cancer. J Natl Compr Canc Netw 7:740–801 Paz-Ares L, 2012, Maintenance therapy with pemetrexed plus best supportive care versus placebo plus best supportive care after induction therapy with pemetrexed plus cisplatin for advanced nonsquamosus non-small cell lung cancer, PARAMOUNT): a double – blind, phase 3, randomised controlled trial. Lancet Oncol 3:247–255 Paz-Ares L, PPARAMOUNT, 2012, Final overall survival, OS, result of the phase III study of maintenance pemetrexed, pem, plus best supportive care, BSC, versus placebo, plb, plus BSC immediatly following induction treatment with pem plus cisplatin, cis, for advanced nonsquamosus, NS, non-small cell lung cancer, NSCLC). J Clin Oncol 30:(Suppl; abstr LBA7507) Pennel NA, 2009, Combined inhibition of the VEGFR and EGFR signalling pathways in the treatment of NSCLC. Oncologist 14:399– 411 Oxanard GR, Binder A, 2013, New targetable oncogenes in Nonsmall- cell lung cancer. J Clin Oncol 31:1097–1104 Gaughan EM, 2011, Genotype-driven therapies for non-small cell lung cancer: focus on EGFR, KRAS and ALK gene abnormalities. Ther Adv Med Oncol 3:113–125 Hynes NE, Lane H, 2005, ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer 5:341–354 Gazdar AF, 2009, Acitvating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical responses to EGFR tyrosine kinase inhibitors. Oncogene 28:24–31 Lee YJ et al, 2011, Lung cancer in never smokers: change of a mindset in the molecular era. Lung Cancer 72:9–15 Reinersman JM et al, 2011, Frequency of EGFR and KRAS mutations in lung adenocarcinoma in African Americans. J Thorac Oncol 6:28–31 Patrick JR, Thomas ES, 2013, KRAS mutation: shouldWe test for it, and does it matter? J Clin Oncol 31:1112–1121 Guan JL et al, 2013, KRAS mutation in patients with lung cancer: a predictor for poor prognosis but Not for EGFR-TKIs or chemotherapy. Ann Surg Oncol 4:1381–11388 FukuokaM(2009, Biomarker analyses from a phase III, randomised, open-label, first-line study of gefitinib(G, versus carboplatin/ paclitaxel(C/P, in clinically selected patients with advanced nonsmall cell lung cancer, NSCLC, in Asia, IPASS, J Clin Oncol 27:, suppl 15) Shepherd FA, 2005, Erlotinib in previously treated non-small cell lung cancer. N Eng Med 353:123–132 Capuzzo F, 2010, Erlotinib as maintenance treatment in advanced non-small cell lung cancer: a multicentre, randomised, placebo – controlled phase III study. Lancet Oncol 6:521–529 Zhou C, Wu YL, 2010, Efficacy results from the randomised phase III OPTIMAL, CTONG 0802, study comparing first-line Erlotinib versus carboplatin, CBDCA, plus gemcitabine, GEM, in Chinese advanced non small cell lung cancer, NSCLC, patients, PTS, with EGFR activating mutations. Ann Oncol 21(supp.8):LBA 13 Garassino MC, 2012, TAILOR: A phase III trial comparing erlotinib with docetaxel as the second-line treatment of NSCLC patients with wild –type, wt, EGFR. J Clin Oncol 30:(abstr LBA7501) Rosell R, Carcereny E et al, 2013, Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer, EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 3: 239–246 Kim ES, Hirsh Vet al, 2008, Gefitinib versus docetaxel in previously treated non-small-cell lung cancer, INTEREST, a randomised phase III trial. The Lancet 372:1809–1818 Fukuoka M, Wu Y, 2011, Biomarker analyses and final overall survival results from a phase III, randomised, open –label, first-line study of Gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced Non-small-cell lung cancer in Asia, IPASS). J Clin Oncol 29:2866–2874 Mok TS, 2009, Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Eng J Med 361:947–957 Inoue A, MinegishiM,MaemondoM(2010, Final results of a phase II study of first- line gefitinib for elderly patients, PTS, with advanced non-small cell lung cancer, NSCLC, harbour epidermal growth factor receptor, EGFR, mutations. NEJ 003 study. Ann Oncol 21:(Suppl 8, Abstract:3559 The Pirker R, Pereira JR, 2012, EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: analysis of data from the phase 3 FLEX study. Lancet Oncol 1:33–42 Teblutt N, Pedersen MW, 2013, Targeting the ERBB family in cancer: couples therapy. Nat Rev Cancer 13:663–673 Miller VA, 2012, Phase IIB/III double-blind randomised trial of afatinib, BIBW 2992, an irreversibile inhibitor of EGFR/HER1 and HER2)+best supportive care, BSC, versus placebo+BSC in patients with NSCLC failing 1-2 lines of chemotehrapy and erlotinib or gefitinib, LUX-LUNG 1, Ann Oncol 21:(Suppl 8,LBA1) Chi YJ, 2012, LUX-Lung 3: A randomized, open-label, phase III study of afatinib versus pemetrexed and cisplatin as first-line treatment for patients with advanced adenocarcinoma of the lung harboring EGFR-activating mutations. J Clinic Oncol 30:18(Suppl. LBA 7500) Pao W, 2004, Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with second mutation in the EGFR kinase ddomain. PloS Med 2:2129–2139 Janjigian YY, Smit EF, Horn L, et al., 2012, Activity of afatinib/ cetuximab in patients, pts, with EGFR mutant non-small cell lung cancer, NSCLC, and acquired resistance, AR, to EGFR inhibitors. Ann Oncol 23:(Suppl 9, abstr. 1227 Sequist LV, Besse B, 2010, Neratinib, an irreversible Pan-ErB receptor tyrosine kinase inhibitor: result of phase II trial in patients with advanced Non-small-cell lung cancer. J Clin Oncol 18:3076–3083 Maemondo M, Inoue A, 2010, Gefinitib or chemotherapy for nonsmall- cell lung cancer with mutated EGFR. N Eng J Med 362:2380– 2388 Pao W, Hutchinson KE, 2012, Chipping away the lung cancer genome. Nat Med 18:349.351 Yano S,Yamada T, 2011, Hepatocyte growth factor expression in EGFR mutant lung cancer with intrinsic and acquired resistance to tyrosine kinase inhibitors in japanase cohort. J Thorac Oncol 6:2011–2017 Oxnard GR, Arcila ME, 2011, New strategies on overcoming acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in lung cancer. Clin Cancer Res 17:5530–5537 Ohashi K, Maruvka YE, 2013, Epidermal growth factor receptor tyrosine kinase inhibitor –resistant disease. J Clin Oncol 31:1070– 1080 Shikada Y, 2005, Platelet-derived growth factor-A is an essential and autocrine regulators of vascular endothelial growth factor expression in non-small cell lung carcinomas. Cancer Res 6:7241–7248 Sandler A, 2006, Paclitaxel-carboplatin alone or with bevacizumab for non-small cell lung cancer. N Eng J Med 24:2542–2550 Crino L, Dansin E, 2010, Safety and efficacy of first line bevacizumab based in advanced non-squamosus non-small cell lung cancer, SAIL MO19390, Phase 4 study. Lancet Oncol 8:733–740 Cho BC, Kim BH, Soo RA, 2010, The role of monoclonal antibody in combination with first-line chemotherapy in Asian patients with advancedNon-small cell lung cancer the role ofmonoclonal antibody in combination with first-line chemotherapy in Asian patients with advanced Non-small cell lung cancer. Yonsei Med J 1:1–8 Schiller JH, Larson T, 2009, Efficacy and safety of axitinib, AG- 013736 AG, in patients with advanced Non-small-cell lung cancer: results from phase II study. J Clin Oncol 23:3836–3841 Chue QS, 2009, Aflibercept(AVE0005, an alternative strategy for inhibiting tumour angiogenesis by vascular endothelial growth factors. Expert Opin Biol Ther 2:263–271 Goss GD, 2010, Randomised, double-blind trial of carboplatin and paclitaxel with either daily oral cediranib or placebo in advanced nonsmall cell lung cancer: NCIC clinical trial group BR24 study. J Clin Oncol 1:49–55 Wedge SR, 2002, ZD6474 inhibits vascular endothelial growth factor signalling, angiogenesis, and tumor growth following oral administration. Cancer Res 62:4645–4655 Novello S, 2009, Phase II study of continuous daily sunitinib dosing in patients with previously treated advanced non-small cell lung cancer. Br J Cancer 9:1543–1548 Blumenschein GR Jr, 2009, Phase II, multicenter, uncontrolled trial of single –agent sorafenib in patients with relapsed or refractory, advanced non –small cell lung cancer. J Clin Oncol 26:4274– 4280 Polverino A, 2006, AMG 706, an oral, multikinase inhibitor that selectively targets vascular endothelial growth factor, platelet –derived gowth facror and kit receptors, potently inhibits angiogenesis and induces regression in tumor xenografts. Cancer Res 17:8715– 8721 Blumenschein GR, Kabbinavar F, 2011, Randomised open label phase II study of motesanib or bevacizumab in combination with paclitaxel and carboplatin, P/C, for advanced nonsquamosus nonsmall cell lung cancer, NSCLC). Ann Oncol 9:2057–2067 Aggarwall C, 2012, Antiangiogenic agents in the the treatment of non-small cell lung cancer:where do we stand and where are we headed? Cancer Biol Ther 5:247–263 SodaM(2007, Identification of the transforming EML4-ALKfusion gene in non –small cell lung cancer. Nature 448:561–567 Ardini E, Magnaghi P, 2010, Anaplastic lymphoma kinase: role in specifis tumours, and development of small molecule inhibitors for cancer therapy. Cencer letters 299:81–94 Capuzzo F, Marchetti A, 2009, Increased MET gene copy number negatively affects survival of surgical resected non-small-cell lung cancer patients. J Clin Oncol 27:1667–1674 Bergethon K, Shaw AT, 2012, ROS1 rearrangements define a unique molecular class of lung cancer. J Clin Oncol 30:863–870 McDermott U, 2008, Genomic alterations of anaplastic lymphoma kinase may sensitize tumors to anaplastic lymphoma kinase inhibitors. Cancer Res 68:3389–3395 PaoW, Girard N, 2011, New driver mutations in non-small-cell lung cancer. Lancet Oncol 12:175–180 Settleman J, 2009, Cell culture modelling of genotype-directed sensitivity to selective kinase inhibitors: targeting the anaplastic lymphoma kinase, ALK). Semin Oncol S36–41 Kwak EL, 2010, Anaplastic lymphoma kinase inhibitions in nonsmall cell lung cancer. N Eng J Med 363:1693–1703 Gadgeel M, Bepler G, 2011, Crizotinib: an anaplastic lymphoma kinase inhibitor. Future Oncol 8:947–953 Alice Tsang S, Ranee Mehra, 2013, Clinical activity of the ALK inhibitor LDK378 in advanced, ALK-positive NSCLC. J Clin Oncol 31:(suppl, Abstr 8010 Sequist LV, Akerley Wl, 2010, Final results from AR197-209: a global randomised placebo –controlled phase II clinical trial of erlotinib PLUS ARQ197 versus erlotinib plus placebo in previously treated EGFR- inhibitor naive patients with advanced non-small cell lung cancer, NSCLC). Ann Oncol 21:(Suppl.8, Abstr 3630 RobinsonKW, Sandler AB, 2013, The role ofMET receptor tyrosine kinase in Non-small cell lung cancer and clinical development of targeted anti-MET agents. Oncologist 18:115–122 Herbst RS, SandlerA, 2008, Bevacizumab and erlotinib: a promising new approach to the treatment of advanced NSCLC. Oncologist 13: 1166–1176 Haisworth J, 2008, A phase III multicenter, placebo-controlled, double-blind, randomised clinical trial to evaluate the efficacy of bevacizumab(Avastin, in combination with erlotinib, Tarceva, compared with chemotherapy alone for treatment of advanced non-small cell lung cancer after failure of standard first line chemotherapy. J Thorac Oncol 3:(Suppl.4) Kabbinavar F, Miller VA, 2010, Overall survival, OS, in ATLAS, a phase IIIb trial comparing bevacizumab(B, therapy with or without erlotinib(E, after completion of chemoterapy with B for first line treatment of locally advanced, recurrent, or metastatic non-small cell lung cancer, NSCLC, J Clin Oncol 28:(Suppl 15)abstr 7526 Gandara D, 2009, S0536: carboplatin, paclitaxel, cetuximab and bevacizumab followed by cetuximab and bevacizumab maintenance in advanced NSCLC: a SWOG phase II study. J Clin Oncol 27:, Suppl.15, abstr 8015 Lind JS, Dingemans AMC, 2009, A phase II study of erlotinib and sorafenib in chemotherapy-naive patients with locally advanced/ metastatic non-small cell lung cancer. Clin Cancer Res 16:3078–3087 Herbst RS, Gordon MS et al., 2013, A study of MPD3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic tumors. J Clin Oncol 31:(suppl, Abstr 3000 Dasanu CA, Sethi N, 2012, Immune alterations and emerging immunotherapeutic approaches in lung cancer. Expert Opin Biol 12: 923–937 Samuels Y, 2004, High frequency of mutations of the PIK3CA gene in human cancers. Science 304:554 Malaga D, 2008, Activating E17K mutation in the gene encoding the protein kinase AKT1 in a subset of squamous cell carcinoma of the lung. Cell cycle 7:665–669 Naoki K, 2002, Missense mutations of the BRAF gene in human lung adenocarcinoma. Cancer Res 62:7001–7003 Marks JL, 2008, Novel MEK 1 mutation identified by mutational analysis of epidermal growth factor receptor signalling pathway genes in lung adenocarcinoma. Cancer Res 68:5524–5528 Gridelli C, Maione P, 2008, The potential Role of mTOR inhibitor sin Non-Small-Cell Lung Cancer. Oncologist 2:139–147 Dong S, Zhang XC, 2012, Everolimus synergizes with gefitinib in non-small-cell lung cancer cell lines resistant to epidermal growth factor receptor tyrosine kinase inhibitors. Cancer Chemother Pharmacol 5:707–716 Belani CP, Goss G, 2011, Recent clinical developments and rationale for combining targeted agents in non-small cell lung cancer, NSCLC). Cancer Treat Rev 38:173–178 FrierichMJ, 2011, NSCLC drug targets acquire New visibility. J Nat Cancer Inst 5:366 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 11 EP 25 DI 10.1007/s12253-013-9719-9 PG 15 ER PT J AU Gao, D Li, Sh AF Gao, Dongwei Li, Sha TI Stimuli-induced Organ-specific Injury Enhancement of Organotropic Metastasis in a Spatiotemporal Regulation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Stimuli; Organ-specific injury; Organotropic metastasis; Spatiotemporal regulation ID Stimuli; Organ-specific injury; Organotropic metastasis; Spatiotemporal regulation AB The relationship between inflammation and tumorigenesis has been established. Recently, inflammation is also reported to be a drive force for cancer metastasis. Further evidences show that various stimuli directly induced-injury in a specific organ can also promote metastasis in this organ, which include epidemiological reports, clinical series and experimental studies. Each type of cancer has preferential sites for metastasis, which is also due to inflammatory factors that are released by primary cancer to act on these sites and indirectly induce injuries on them. Host factors such as stress,fever can also influence distant metastasis in a specific site through stimulation of immune and inflammatory effects. The five aspects support an idea that specific-organ injury directly induced by various stimuli or indirectly induced by primary tumor or host factors activation of proinflammatory modulators can promote metastasis in this organ through a spatiotemporal regulation, which has important implications for personalized prediction, prevention and management of cancer metastasis. C1 [Gao, Dongwei] 536 Hospital of PLA, 29# Xiadu street, 810007 Xining, Qinghai Province, China. [Li, Sha] Lanzhou General Hospital of PLA, Department of Radiation Oncology, 333# Southern Binhe Road, 730050 Lanzhou, Gansu Province, China. RP Gao, D (reprint author), 536 Hospital of PLA, 810007 Xining, China. EM gdw3152007@hotmail.com CR Balkwill F, Mantovani A, 2001, Inflammation and cancer: Back to Virchow? Lancet 357:539–545 Hans C, 2004, At the crossroads of inflammation and cancer. Cell 118:671–674 Lu H, Ouyang W, Huang C, 2006, Inflammation, a key event in cancer development. Mol Cancer Res 4:221–233 Schottenfeld D, Beebe-Dimmer J, 2006, Chronic inflammation: A common and important factor in the pathogenesis of neoplasia. CA Cancer J Clin 56:69–83 Grivennikov SI, Karin M, 2010, Inflammation and oncogenesis: A vicious connection. Curr Opin Genet Dev 20:65–71 Nowacki MP, Janik P, Nowacki PM, 1996, Inflammation and metastases. Med Hypotheses 47:193–196 Mantovani A, 2009, Cancer: Inflaming metastasis. Nature 457:36– 37 Solinas G, Marchesi F, Garlanda C, Mantovani A, Allavena P, 2010, Inflammation-mediated promotion of invasion and metastasis. Cancer Metastasis Rev 29:243–248 Jones FS, Rous P, 1914, On the cause of the localization of secondary tumors at points of injury. J Exp Med 20:404– 412 Brody JS, Spira A, 2006, State of the art. Chronic obstructive pulmonary disease, inflammation, and lung cancer. Proc Am Thorac Soc 3:535–537 Walser T, Cui X, Yanagawa J, Lee JM, Heinrich E, Lee G, Sharma S, Dubinett SM, 2008, Smoking and lung cancer: The role of inflammation. Proc Am Thorac Soc 5:811–815 Calle EE, Miracle-McMahill HL, Thun MJ, Heath CJ, 1994, Cigarette smoking and risk of fatal breast cancer. Am J Epidemiol 139:1001–1007 Yu GP, Ostroff JS, Zhang ZF, Tang J, Schantz SP, 1997, Smoking history and cancer patient survival: A hospital cancer registry study. Cancer Detect Prev 21:497–509 Shaw HM, Milton GW, 1981, Smoking and the development of metastases from malignant melanoma. Int J Cancer 28:153–156 Scanlon EF, Suh O, Murthy SM, Mettlin C, Reid SE, Cummings KM, 1995, Influence of smoking on the development of lung metastases from breast cancer. Cancer 75:2693–2699 Murin S, Inciardi J, 2001, Cigarette smoking and the risk of pulmonary metastasis from breast cancer. Chest 119:1635–1640 Abrams JA, Lee PC, Port JL, Altorki NK, Neugut AI, 2008, Cigarette smoking and risk of lung metastasis from esophageal cancer. Cancer Epidemiol Biomarkers Prev 17:2707–2713 Meliska CJ, Stunkard ME, Gilbert DG, Jensen RA, Martinko JM, 1995, Immune function in cigarette smokers who quit smoking for 31 days. J Allergy Clin Immunol 95:901–910 Mayhan WG, Sharpe GM, 1998, Nicotine impairs histamineinduced increases in macromolecular efflux: Role of oxygen radicals. J Appl Physiol 84:1589–1595 Conklin BS, Zhao W, Zhong DS, Chen C, 2002, Nicotine and cotinine up-regulate vascular endothelial growth factor expression in endothelial cells. Am J Pathol 160:413–418 Lu Q, Newton J, Gabino-Miranda G, Ortiz M, Harrington EO, Rounds SIS, 2010, Cigarette smoke extract increases lung endothelial permeability through oxidative stress-mediated alterations of small GTPases. Am J Resp Crit Care 181:A3427 Padua D, Zhang XH, Wang Q, Nadal C, Gerald WL, Gomis RR, Massague J, 2008, TGFbeta primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell 133:66–77 O’Byrne PM, Postma DS, 1999, The many faces of airway inflammation. Asthma and chronic obstructive pulmonary disease. Asthma Research Group. Am J Respir Crit CareMed 159:S41–S63 Bosse Y, Rola-PleszczynskiM, 2007, Controversy surrounding the increased expression of TGF beta 1 in asthma. Respir Res 8:66 Fidler IJ, 1990, Critical factors in the biology of human cancer metastasis: Twenty-eighth G.H.A. clowes memorial award lecture. Cancer Res 50:6130–6138 Mannel DN, Orosz P, Hafner M, Falk W, 1994, Mechanisms involved in metastasis enhanced by inflammatory mediators. Circ Shock 44:9–13 Fidler IJ, 1995, Critical factors in the biology of human cancer metastasis. Am Surg 61:1065–1066 Bogenrieder T, Herlyn M, 2003, Axis of evil: Molecular mechanisms of cancer metastasis. Oncogene 22:6524–6536 LinWW, KarinM(2007, A cytokine-mediated link between innate immunity, inflammation, and cancer. J Clin Invest 117:1175–1183 Liao D, Luo Y, Markowitz D, Xiang R, Reisfeld RA, 2009, Cancer associated fibroblasts promote tumor growth and metastasis by modulating the tumor immune microenvironment in a 4 T1 murine breast cancer model. PLoS One 4:e7965 Finger EC, Giaccia AJ, 2010, Hypoxia, inflammation, and the tumor microenvironment in metastatic disease. Cancer Metastasis Rev 29:285–293 Erez N, Coussens LM, 2011, Leukocytes as paracrine regulators of metastasis and determinants of organ-specific colonization. Int J Cancer 128:2536–2544 Yang L, 2010, TGFbeta and cancer metastasis: An inflammation link. Cancer Metastasis Rev 29:263–271 Taranova AG, Maldonado DR, Vachon CM, Jacobsen EA, Abdala- Valencia H, McGarry MP, Ochkur SI, Protheroe CA, Doyle A, Grant CS, Cook-Mills J, Birnbaumer L, Lee NA, Lee JJ, 2008, Allergic pulmonary inflammation promotes the recruitment of circulating tumor cells to the lung. Cancer Res 68:8582–8589 Formenti SC, Demaria S, 2009, Systemic effects of local radiotherapy. Lancet Oncol 10:718–726 Dao TL, Kovaric J, 1962, Incidence of pulmonary and skin metastases in women with breast cancer who received postoperative irradiation. Surgery 52:203–212 Oosterling SJ, van der Bij GJ, van Egmond M, van der Sijp JR, 2005, Surgical trauma and peritoneal recurrence of colorectal carcinoma. Eur J Surg Oncol 31:29–37 Nagai S, Fujii T, Kodera Y, Kanda M, Sahin TT, Kanzaki A, Hayashi M, Sugimoto H, Nomoto S, Takeda S, Morita S, Nakao A, 2011, Recurrence pattern and prognosis of pancreatic cancer after pancreatic fistula. Ann Surg Oncol 18:2329–2337 Allon I, Pessing A, Kaplan I, Allon DM, Hirshberg A, 2013, Metastatic tumors to the gingiva, the presence of teeth as a contributing factor- a literature analysis. J Periodontol Black JW, 1964, The localisation of metastatic brown-pearce carcinoma in granulation tissue. Br J Cancer 18:143–145 Benisch B, Toker C, 1971, Metastasis to the site of recent trauma. JAMA 216:2142–2143 Cohen HJ, Laszlo J, 1972, Influence of trauma on the unusual distribution of metastases from carcinoma of the larynx. Cancer 29:466–471 Cunha-Gomes D, Prasad R, Bhathena HM, Kavarana NM, 1999, Tumor implantation at the flap donor site: A case report. Acta Chir Plast 41:75–76 Shine T, Wallack MK, 1981, Inflammatory oncotaxis after testing the skin of the cancer patient. Cancer 47:1325–1328 Marley NF, Marley WM, 1982, Skin metastases in an area of radiation dermatitis. Arch Dermatol 118:129–131 Ito H, Kubo A, Shigematsu N, Hashimoto S, 1984, Skin metastases within the previous radiation field after prophylactic postoperative radiotherapy for breast cancer. Clin Exp Metastasis 2:235–239 Carr RJ, Gilbert PM, 1986, Tumour implantation to a temporalis muscle flap donor site. Br J Oral Maxillofac Surg 24:102–106 Climo MS, EnosWF, King PG, 1989, Squamous cell carcinoma of skin developing in a skin graft donor site. Br J Plast Surg 42:118 Betke M, Suss R, Hohenleutner U, Lubke S, Eckert F, 1993)Gastric carcinoma metastatic to the site of a congenitalmelanocytic nevus. J Am Acad Dermatol 28:866–869 Trefzer U, Schwurzer-Voit M, Audring H, Jahn S, Thies E, SterryW, 1998, Multiple melanoma metastases in split-thickness skin graft donor sites. J Am Acad Dermatol 38:997–998 Serrano-Ortega S, Buendia-Eisman A, Ortega DOR, Linares SJ, 2000, Melanoma metastasis in donor site of full-thickness skin graft. Dermatology 201:377–378 Pradhan S, Asthana AK, Sharan GK, Kumar M, Sharma OP, 2006, Recurrence of carcinoma cervix in the scar of previous cesarean section: A case report. Int J Gynecol Cancer 16:900–904 Erol B, Ufuk U, Husamettin T, Yasin U, Aslihan C, Koray K, 2008, True hematogenous metastases of melanoma on contralateral skin graft donor site: A case report. Melanoma Res 18:443–446 Marenco F, Fava P,Macripo G, Quaglino P, Savoia P, BernengoMG, 2009, Cutaneous melanoma metastases arising on a split-skin graft donor site. Dermatol Surg 35:1282–1285 Hussain A, Ekwobi C, Watson S, 2011, Metastatic implantation squamous cell carcinoma in a split-thickness skin graft donor site. J Plast Reconstr Aesthet Surg 64:690–692 Dunn PT, Bigler CF, 1997, Metastasis in an electrodesiccation and curettage scar. J Am Acad Dermatol 36:117–118 Ferguson PC, Sommerville S, Grimer RJ, 2004, Possible metastasis of osteosarcoma to a remote biopsy site: a case report. Clin Orthop Relat Res 424:216–220 Mincheff TV, 2005, Metastatic spread to a percutaneous gastrostomy site from head and neck cancer: Case report and literature review. JSLS 9:466–471 Hameed H, Khan YI, 2009, Metastasis of carcinosarcoma of oesophagus to gastrostomy site. Br J Oral Maxillofac Surg 47:643– 644 El SaghirN, Elhajj I,Geara F, HouraniM(2005, Trauma-associated growth of suspected dormant micrometastasis. Bmc Cancer 5:94 FukushimaM, Katagiri A,Mori T,Watanabe T, Katayama Y, 2010, Case of skull metastasis from hepatocellular carcinoma at the site of skull fracture. No Shinkei Geka 38:371–377 Walter ND, Rice PL, Redente EF, Kauvar EF, Lemond L, Aly T, Wanebo K, Chan ED, 2011, Wound healing after trauma may predispose to lung cancer metastasis: Review of potential mechanisms. Am J Respir Cell Mol Biol 44:591–596 Bergqvist D, Mattsson J, 1978, Solitary calcaneal metastasis as the first sign of gastric cancer. A case report. Ups JMed Sci 83:115–118 De Simone P, Carrai P, Morelli L, Coletti L, Petruccelli S, Filipponi F, Doria R, Menichetti F, Vannozzi R, 2005, Posttransplant hepatocellular carcinoma metastasis at a skull trauma site. Transplantation 80:1358–1359 Yip KM, Lin J, Kumta SM, 1996, A pelvic osteosarcoma with metastasis to the donor site of the bone graft. A case report. Int Orthop 20:389–391 Wondergem JH, Holscher HC, Arndt JW, Bakker W, Pauwels EK, 1987, A long term scintigraphic bone abnormality after trauma: A pitfall in the survey for metastatic disease. Eur J NuclMed 13:321– 323 Ivarsson L, 1976, Pulmonary metastasis formation after trauma. Acta Chir Scand Suppl 463:1–46 Kukita K, Shirakawa T, Kojima A, Yoshida H, Yoshida K, Tokuomi H, Kurano R, 1992, An autopsy case of pulmonary metastasis of cholangiocellular carcinoma associated withmarked fibrotic change of the lungs. Nihon Kyobu Shikkan Gakkai Zasshi 30:1738–1742 Nielsen SL, Posner JB, 1983, Brain metastasis localized to an area of infarction. J Neurooncol 1:191–195 Rodriguez RE, Valero V, Watanakunakorn C, 1986, Salmonella focal intracranial infections: Review of the world literature, 1884- 1984, and report of an unusual case. Rev Infect Dis 8:31–41 Palazzo FF, New NE, Cullen PT, 2000, An unusual cause of thigh pain in colonic cancer. Inflammatory oncotaxis? Acta Chir Belg 100:28–30 Magee T, Rosenthal H, 2002, Skeletal muscle metastases at sites of documented trauma. AJR Am J Roentgenol 178:985–988 Currall VA, Dixon JH, 2008, Synovial metastasis: An unusual cause of pain after total knee arthroplasty. J Arthroplasty 23:631– 636 Dionigi G, Uccella S, Gandolfo M, Lai A, Bertocchi V, Rovera F, Tanda ML, 2008, Solitary intrathyroidal metastasis of renal clear cell carcinoma in a toxic substernal multinodular goiter. Thyroid Res 1:6 DerHagopian RP, Sugarbaker EV, Ketcham A, 1978, Inflammatory oncotaxis. JAMA 240:374–375 Murin S, Pinkerton KE, Hubbard NE, Erickson K, 2004, The effect of cigarette smoke exposure on pulmonary metastatic disease in a murine model of metastatic breast cancer. Chest 125:1467–1471 Kim H, Zamel R, Bai XH, Liu M, 2013, PKC activation induces inflammatory response and cell death in human bronchial epithelial cells. PLoS One 8:e64182 Gopalakrishna R, Chen ZH, Gundimeda U, 1994, Tobacco smoke tumor promoters, catechol and hydroquinone, induce oxidative regulation of protein kinase C and influence invasion andmetastasis of lung carcinoma cells. Proc Natl Acad Sci U S A 91:12233–12237 Blache D, 1995, Involvement of hydrogen and lipid peroxides in acute tobacco smoking-induced platelet hyperactivity. AmJ Physiol 268:H679–H685 Kunita A, Kashima TG,Morishita Y, Fukayama M, Kato Y, Tsuruo T, Fujita N, 2007, The platelet aggregation-inducing factor aggrus/ podoplanin promotes pulmonary metastasis. Am J Pathol 170: 1337–1347 Yan L, Cai Q, Xu Y, 2013, The ubiquitin-CXCR4 axis plays an important role in acute lung infection-enhanced lung tumor metastasis. Clin Cancer Res 19:4706–4716 Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegui E, Zlotnik A, 2001, Involvement of chemokine receptors in breast cancer metastasis. Nature 410:50–56 Smith HA, Kang Y, 2013, Acute infection induces a metastatic niche: A double menace for cancer patients. Clin Cancer Res 19: 4547–4549 Carmel RJ, Brown JM, 1977, The effect of cyclophosphamide and other drugs on the incidence of pulmonary metastases in mice. Cancer Res 37:145–151 Milas L, Malenica B, Allegretti N, 1979, Enhancement of artificial lung metastases in mice caused by cyclophosphamide. Cancer Immunol Immunother 6:191–196 Yamauchi K, Yang M, Hayashi K, Jiang P, Yamamoto N, Tsuchiya H, Tomita K, Moossa AR, Bouvet M, Hoffman RM, 2008, Induction of cancer metastasis by cyclophosphamide pretreatment of host mice: An opposite effect of chemotherapy. Cancer Res 68: 516–520 Gasic GJ, Iwakawa A, Gasic TB, Viner ED, Milas L, 1984, Leech salivary gland extract from Haementeria officinalis, a potent inhibitor of cyclophosphamide- and radiation-induced artificial metastasis enhancement. Cancer Res 44:5670–5676 Man S, Zhang Y, Gao W, Yan L, Ma C, 2008, Cyclophosphamide promotes pulmonary metastasis on mouse lung adenocarcinoma. Clin Exp Metastasis 25:855–864 Wu YJ, Muldoon LL, Dickey DT, Lewin SJ, Varallyay CG, Neuwelt EA, 2009, Cyclophosphamide enhances human tumor growth in nude rat xenografted tumor models. Neoplasia 11:187– 195 Sleijfer S, 2001, Bleomycin-induced pneumonitis. Chest 120:617– 624 Chung MP, Monick MM, Hamzeh NY, Butler NS, Powers LS, Hunninghake GW, 2003, Role of repeated lung injury and genetic background in bleomycin-induced fibrosis. Am J Respir Cell Mol Biol 29:375–380 Schmidt R, Ruppert C, Markart P, Lubke N, Ermert L, Weissmann N, Breithecker A, Ermert M, SeegerW, Gunther A, 2004, Changes in pulmonary surfactant function and composition in bleomycininduced pneumonitis and fibrosis. Toxicol Appl Pharmacol 195: 218–231 Opdenakker G, Van Damme J, 1992, Chemotactic factors, passive invasion and metastasis of cancer cells. Immunol Today 13:463– 464 Hiratsuka S, Nakamura K, Iwai S, Murakami M, Itoh T, Kijima H, Shipley JM, Senior RM, Shibuya M, 2002, MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lungspecific metastasis. Cancer Cell 2:289–300 MuellerMM, Fusenig NE, 2004, Friends or foes - bipolar effects of the tumour stroma in cancer. Nat Rev Cancer 4:839–849 Shimo T, Kubota S, Yoshioka N, Ibaragi S, Isowa S, Eguchi T, Sasaki A, Takigawa M, 2006, Pathogenic role of connective tissue growth factor, CTGF/CCN2, in osteolytic metastasis of breast cancer. J Bone Miner Res 21:1045–1059 Affara NI, Coussens LM, 2007, IKKalpha at the crossroads of inflammation and metastasis. Cell 129:25–26 Solinas G, Germano G, Mantovani A, Allavena P, 2009, Tumorassociated macrophages, TAM, as major players of the cancerrelated inflammation. J Leukoc Biol 86:1065–1073 Valastyan S, Weinberg RA, 2011, Tumor metastasis: Molecular insights and evolving paradigms. Cell 147:275–292 Orr FW, Young L, King GM, Adamson IY, 1988, Preferential growth of metastatic tumors at the pleural surface of mouse lung. Clin Exp Metastasis 6:221–232 Adamson IY, Orr FW, Young L, 1986, Effects of injury and repair of the pulmonary endothelium on lung metastasis after bleomycin. J Pathol 150:279–287 Orr FW, Adamson IY, Young L, 1986, Promotion of pulmonary metastasis in mice by bleomycin-induced endothelial injury. Cancer Res 46:891–897 Orr FW, Adamson IY, Young L, 1986, Quantification of metastatic tumor growth in bleomycin-injured lungs. Clin Exp Metastasis 4: 105–116 Saiki I, Milas L, Hunter N, Fidler IJ, 1986, Treatment of experimental lung metastasis with local thoracic irradiation followed by systemic macrophage activation with liposomes containing muramyl tripeptide. Cancer Res 46:4966–4970 van den Brenk HA, Kelly H, 1974, Potentiating effect of prior local irradiation of the lungs on pulmonary metastases. Br J Radiol 47: 332–336 Brown JM, 1973, The effect of lung irradiation on the incidence of pulmonary metastases in mice. Br J Radiol 46:613–618 Withers HR, Milas L, 1973, Influence of preirradiation of lung on development of artificial pulmonary metastases of fibrosarcoma in mice. Cancer Res 33:1931–1936 Biswas S, Guix M, Rinehart C, Dugger TC, Chytil A, Moses HL, Freeman ML, Arteaga CL, 2007, Inhibition of TGF-beta with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J Clin Invest 117:1305–1313 Vincic L, Orr FW, Warner DJ, Suyama KL, Kay JM, 1989, Enhanced cancer metastasis after monocrotaline-induced lung injury. Toxicol Appl Pharmacol 100:259–270 Orr FW, Adamson IY, Young L, 1985, Pulmonary inflammation generates chemotactic activity for tumor cells and promotes lung metastasis. Am Rev Respir Dis 131:607–611 Adamson IYYLOF, 1987, Tumor metastasis after hyperoxic injury and repair of the pulmonary endothelium. Lab Invest 57:71–77 Kobayashi T, Todoroki T, Sato H, 1987, Enhancement of pulmonary metastasis of murine fibrosarcoma NR-FS by ozone exposure. J Toxicol Environ Health 20:135–145 Orr FW, Warner DJ, 1987, Effects of neutrophil-mediated pulmonary endothelial injury on the localization and metastasis of circulating Walker carcinosarcoma cells. Invasion Metastasis 7:183–196 Orr FW, Warner DJ, 1990, Effects of systemic complement activation and neutrophil-mediated pulmonary injury on the retention and metastasis of circulating cancer cells in mouse lungs. Lab Invest 62: 331–338 Kennel SJ, Lankford TK, Ullrich RL, Jamasbi RJ, 1988, Enhancement of lung tumor colony formation by treatment of mice with monoclonal antibodies to pulmonary capillary endothelial cells. Cancer Res 48:4964–4968 Orr FW, Adamson IY, Warner D, Leroyer V, Werner L, Shaughnessy S, Young L, 1988, The effects of oxygen radical– mediated pulmonary endothelial damage on cancer metastasis.Mol Cell Biochem 84:189–198 Hafner M, Orosz P, Kruger A, Mannel DN, 1996, TNF promotes metastasis by impairing natural killer cell activity. Int J Cancer 66: 388–392 Wu WJ, Pruett SB, 1999, Ethanol decreases host resistance to pulmonary metastases in a mouse model: Role of natural killer cells and the ethanol-induced stress response. Int J Cancer 82:886–892 Harmey JH, Bucana CD, LuW, Byrne AM, McDonnell S, Lynch C, Bouchier-Hayes D, Dong Z, 2002, Lipopolysaccharide-induced metastatic growth is associated with increased angiogenesis, vascular permeability and tumor cell invasion. Int J Cancer 101:415–422 Gupta GP, Nguyen DX, Chiang AC, Bos PD, Kim JY, Nadal C, Gomis RR, Manova-Todorova K, Massague J, 2007, Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature 446:765–770 Stathopoulos GT, Sherrill TP, Han W, Sadikot RT, Yull FE, Blackwell TS, Fingleton B, 2008, Host nuclear factor-kappaB activation potentiates lung cancer metastasis. Mol Cancer Res 6: 364–371 Wang W, Xu GL, Jia WD, Ma JL, Li JS, Ge YS, Ren WH, Yu JH, Liu WB, 2009, Ligation of TLR2 by versican: A link between inflammation and metastasis. Arch Med Res 40:321–323 Kim S, Takahashi H, Lin W, Descargues P, Grivennikov S, Kim Y, Luo J, KarinM(2009, Carcinoma-produced factors activatemyeloid cells through TLR2 to stimulate metastasis. Nature 457:102–106 Maru Y, 2010, Premetastatic milieu explained by TLR4 agonistmediated homeostatic inflammation. Cell Mol Immunol 7:94–99 Orosz P, Echtenacher B, FalkW, Ruschoff J,Weber D,Mannel DN, 1993, Enhancement of experimental metastasis by tumor necrosis factor. J Exp Med 177:1391–1398 Ai-Aql ZS, Alagl AS, Graves DT, Gerstenfeld LC, Einhorn TA, 2008, Molecular mechanisms controlling bone formation during fracture healing and distraction osteogenesis. J Dent Res 87:107– 118 Robling AG, Castillo AB, Turner CH, 2006, Biomechanical and molecular regulation of bone remodeling. Annu Rev Biomed Eng 8: 455–498 Lee JY, Murphy SM, Scanlon EF, 1994, Effect of trauma on implantation of metastatic tumor in bone in mice. J Surg Oncol 56:178–184 Vicent S, Luis-Ravelo D, Anton I, Garcia-Tunon I, Borras-Cuesta F, Dotor J,De Las RJ, Lecanda F, 2008, A novel lung cancer signature mediates metastatic bone colonization by a dualmechanism. Cancer Res 68:2275–2285 Das RL, Pathangey LB, Tinder TL, Schettini JL, Gruber HE, Mukherjee P, 2009, Breast-cancer-associated metastasis is significantly increased in a model of autoimmune arthritis. Breast Cancer Res 11:R56 Roy LD, Ghosh S, Pathangey LB, Tinder TL, Gruber HE, Mukherjee P, 2011, Collagen induced arthritis increases secondary metastasis in MMTV-PyV MT mouse model of mammary cancer. Bmc Cancer 11:365 Steeg PS, 2006, Tumor metastasis: Mechanistic insights and clinical challenges. Nat Med 12:895–904 Roodman GD, 2004, Mechanisms of bone metastasis. N Engl J Med 350:1655–1664 Taichman RS, Cooper C, Keller ET, Pienta KJ, Taichman NS, McCauley LK, 2002, Use of the stromal cell-derived factor-1/ CXCR4 pathway in prostate cancer metastasis to bone. Cancer Res 62:1832–1837 Kitaori T, Ito H, Schwarz EM, Tsutsumi R, Yoshitomi H, Oishi S, Nakano M, Fujii N, Nagasawa T, Nakamura T, 2009, Stromal cellderived factor 1/CXCR4 signaling is critical for the recruitment of mesenchymal stemcells to the fracture site during skeletal repair in a mouse model. Arthritis Rheum 60:813–823 Barash H, GE R, Edrei Y, Ella E, Israel A, Cohen I, Corchia N, Ben-Moshe T, Pappo O, Pikarsky E, Goldenberg D, Shiloh Y, Galun E, Abramovitch R, 2010, Accelerated carcinogenesis following liver regeneration is associated with chronic inflammation-induced double-strand DNA breaks. Proc Natl Acad Sci U S A 107:2207–2212 Fisher B, Fisher ER, 1959, Experimental studies of factors influencing hepatic metastases. II. Effect of partial hepatectomy. Cancer 12: 929–932 Murthy SM, Goldschmidt RA, Rao LN, AmmiratiM, Buchmann T, Scanlon EF, 1989, The influence of surgical trauma on experimental metastasis. Cancer 64:2035–2044 Uotani H, Yamashita I, Nagata T, Kishimoto H, Kashii Y, Tsukada K, 2001, Induction of E-selectin after partial hepatectomy promotes metastases to liver in mice. J Surg Res 96:197–203 Harun N, Nikfarjam M,Muralidharan V, Christophi C, 2007, Liver regeneration stimulates tumor metastases. J Surg Res 138:284–290 Wu Y, Brodt P, Sun H,MejiaW, Novosyadlyy R,NunezN, Chen X, Mendoza A, Hong SH, Khanna C, Yakar S, 2010, Insulin-like growth factor-I regulates the liver microenvironment in obese mice and promotes liver metastasis. Cancer Res 70:57–67 Nijkamp MW, Hoogwater FJ, Govaert KM, Steller EJ, Verheem A, Kranenburg O, Borel RI, 2011, A role for CD95 signaling in ischemia/reperfusion-induced invasion and outgrowth of colorectal micrometastases in mouse liver. J Surg Oncol 104:198–204 Doi K, Horiuchi T, Uchinami M, Tabo T, Kimura N, Yokomachi J, Yoshida M, Tanaka K, 2002, Hepatic ischemia-reperfusion promotes liver metastasis of colon cancer. J Surg Res 105:243–247 Nicoud IB, Jones CM, Pierce JM, Earl TM, Matrisian LM, Chari RS, Gorden DL, 2007, Warm hepatic ischemia-reperfusion promotes growth of colorectal carcinoma micrometastases in mouse liver via matrix metalloproteinase-9 induction. Cancer Res 67: 2720–2728 Nguyen DX, Bos PD,Massague J, 2009, Metastasis: From dissemination to organ-specific colonization. Nat Rev Cancer 9:274–284 Uetsuji S, Yamamura M, Yamamichi K, Okuda Y, Takada H, Hioki K, 1992, Absence of colorectal cancer metastasis to the cirrhotic liver. Am J Surg 164:176–177 Melato M, Laurino L, Mucli E, Valente M, Okuda K, 1989, Relationship between cirrhosis, liver cancer, and hepaticmetastases. An autopsy study. Cancer 64:455–459 Seymour K, Charnley RM, 1999, Evidence that metastasis is less common in cirrhotic than normal liver: A systematic review of postmortem case–control studies. Br J Surg 86:1237–1242 Qi K, Qiu H, Sun D, Minuk GY, Lizardo M, Rutherford J, Orr FW, 2004, Impact of cirrhosis on the development of experimental hepatic metastases by B16F1 melanoma cells in C57BL/6 mice. Hepatology 40:1144–1150 Kuriyama S, Yamazaki M, Mitoro A, Tsujimoto T, Kikukawa M, Tsujinoue H, Nakatani T, Toyokawa Y, Yoshiji H, Fukui H, 1999, Hepatocellular carcinoma in an orthotopic mouse model metastasizes intrahepatically in cirrhotic but not in normal liver. Int J Cancer 80:471–476 Berghoff AS, Lassmann H, Preusser M, Hoftberger R, 2013, Characterization of the inflammatory response to solid cancer metastases in the human brain. Clin Exp Metastasis 30:69–81 NodaM, Seike T, Fujita K, Yamakawa Y, KidoM, Iguchi H, 2009, The role of immune cells in brainmetastasis of lung cancer cells and neuron-tumor cell interaction. Ross Fiziol Zh Im IMSechenova 95: 1386–1396 Bos PD, Zhang XH, Nadal C, Shu W, Gomis RR, Nguyen DX, Minn AJ, van de Vijver MJ, Gerald WL, Foekens JA, Massague J, 2009, Genes that mediate breast cancer metastasis to the brain. Nature 459:1005–1009 WilhelmI,Molnar J, Fazakas C, Hasko J, Krizbai IA, 2013, Role of the blood–brain barrier in the formation of brain metastases. Int J Mol Sci 14:1383–1411 Seelbach M, Chen L, Powell A, Choi YJ, Zhang B, Hennig B, Toborek M, 2010, Polychlorinated biphenyls disrupt blood–brain barrier integrity and promote brain metastasis formation. Environ Health Perspect 118:479–484 Coisne C, Engelhardt B, 2011, Tight junctions in brain barriers during central nervous system inflammation. Antioxid Redox Signal 15:1285–1303 Esposito P, Gheorghe D, Kandere K, Pang X, Connolly R, Jacobson S, Theoharides TC, 2001, Acute stress increases permeability of the blood–brain-barrier through activation of brainmast cells. Brain Res 888:117–127 Esposito P, Chandler N, Kandere K, Basu S, Jacobson S, Connolly R, Tutor D, Theoharides TC, 2002, Corticotropin-releasing hormone and brain mast cells regulate blood–brain-barrier permeability induced by acute stress. J Pharmacol Exp Ther 303:1061–1066 Theoharides TC, Asadi S, Patel AB, 2013, Focal brain inflammation and autism. J Neuroinflammation 10:46 Theoharides TC, Cochrane DE, 2004, Critical role of mast cells in inflammatory diseases and the effect of acute stress. J Neuroimmunol 146:1–12 Rozniecki JJ, Sahagian GG, Kempuraj D, Tao K, Jocobson S, Zhang B, Theoharides TC, 2010, Brain metastases of mouse mammary adenocarcinoma is increased by acute stress. Brain Res 1366: 204–210 Li A, Rockne KJ, Sturchio N, SongW, Ford JC,Wei H(2009, PCBs in sediments of the great lakes–distribution and trends, homolog and chlorine patterns, and in situ degradation. Environ Pollut 157:141– 147 Sipos E, Chen L, Andras IE, Wrobel J, Zhang B, Pu H, Park M, Eum SY, Toborek M, 2012, Proinflammatory adhesion molecules facilitate polychlorinated biphenyl-mediated enhancement of brain metastasis formation. Toxicol Sci 126:362–371 Van Den Brenk HA, Kelly H, 1973, Stimulation of growth of metastases by local x-irradiation in kidney and liver. Br J Cancer 28:349–353 Ammirati MRLMM, 1989, Partial nephrectomy in mice with milliwatt carbon dioxide laser and its influence on experimental metastasis. J Surg Oncol 3:153–159 AokiY, ShimuraH, Li H,Mizumoto K,DateK, TanakaM(1999, A model of port-sitemetastases of gallbladder cancer: The influence of peritoneal injury and its repair on abdominal wall metastases. Surgery 125:553–559 Hopkins MP, Dulai RM, Occhino A, Holda S, 1999, The effects of carbon dioxide pneumoperitoneum on seeding of tumor in port sites in a rat model. Am J Obstet Gynecol 181:1329–1334 Ost MC, Patel KP, Rastinehad AR, Chu PY, Anderson AE, Smith AD, Lee BR, 2008, Pneumoperitoneum with carbon dioxide inhibits macrophage tumor necrosis factor-alpha secretion: Source of transitional-cell carcinoma port-site metastasis, with prophylactic irrigation strategies to decrease laparoscopic oncologic risks. J Endourol 22:105–112 Jones DB, Guo LW, Reinhard MK, Soper NJ, Philpott GW, Connett J, Fleshman JW, 1995, Impact of pneumoperitoneum on trocar site implantation of colon cancer in hamster model. Dis Colon Rectum 38:1182–1188 Bouvy ND, Marquet RL, Jeekel H, Bonjer HJ, 1996, Impact of gas(less, laparoscopy and laparotomy on peritoneal tumor growth and abdominal wall metastases. Ann Surg 224(694– 700):700–701 van den Tol PM, van Rossen EE, van Eijck CH, Bonthuis F, Marquet RL, Jeekel H, 1998, Reduction of peritoneal trauma by using nonsurgical gauze leads to less implantation metastasis of spilled tumor cells. Ann Surg 227:242–248 Zeamari S, Roos E, Stewart FA, 2004, Tumour seeding in peritoneal wound sites in relation to growth-factor expression in early granulation tissue. Eur J Cancer 40:1431–1440 Li F, Tiede B, Massague J, Kang Y, 2007, Beyond tumorigenesis: Cancer stem cells in metastasis. Cell Res 17:3–14 Schier AF, 2003, Chemokine signaling: Rules of attraction. Curr Biol 13:R192–R194 Kucia M, Reca R, Miekus K, Wanzeck J, Wojakowski W, Janowska-Wieczorek A, Ratajczak J, Ratajczak MZ, 2005, Trafficking of normal stem cells and metastasis of cancer stem cells involve similar mechanisms: Pivotal role of the SDF-1-CXCR4 axis. Stem Cells 23:879–894 Arwert EN, Hoste E, Watt FM, 2012, Epithelial stem cells, wound healing and cancer. Nat Rev Cancer 12:170–180 Korkaya H, Liu S, Wicha MS, 2011, Regulation of cancer stem cells by cytokine networks: Attacking cancer’s inflammatory roots. Clin Cancer Res 17:6125–6129 Shipitsin M, Campbell LL, Argani P, Weremowicz S, Bloushtain- Qimron N, Yao J, Nikolskaya T, Serebryiskaya T, BeroukhimR, Hu M, Halushka MK, Sukumar S, Parker LM, Anderson KS, Harris LN, Garber JE, RichardsonAL, Schnitt SJ,Nikolsky Y,Gelman RS, Polyak K, 2007)Molecular definition of breast tumor heterogeneity. Cancer Cell 11:259–273 Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordon- Cardo C, Guise TA, Massague J, 2003, A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3:537–549 Minn AJ, Gupta GP, Siegel PM, Bos PD, ShuW, Giri DD, Viale A, Olshen AB, Gerald WL, Massague J, 2005, Genes that mediate breast cancer metastasis to lung. Nature 436:518–524 Mueller L, Goumas FA, Affeldt M, Sandtner S, Gehling UM, Brilloff S, Walter J, Karnatz N, Lamszus K, Rogiers X, Broering DC, 2007, Stromal fibroblasts in colorectal liver metastases originate from resident fibroblasts and generate an inflammatory microenvironment. Am J Pathol 171:1608–1618 Steeg PS, 2005, Cancer biology: Emissaries set up newsites.Nature 438:750–751 Duda DG, Duyverman AM, Kohno M, Snuderl M, Steller EJ, Fukumura D, Jain RK, 2010, Malignant cells facilitate lung metastasis by bringing their own soil. Proc Natl Acad Sci U S A 107: 21677–21682 Tan W, Zhang W, Strasner A, Grivennikov S, Cheng JQ, Hoffman RM, Karin M, 2011, Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL-RANK signalling. Nature 470:548–553 Al-Shibli KIDTAS, 2008, Prognostic effect of epithelial and stromal lymphocyte infiltration in non-small cell lung cancer. Clin Cancer Res 14:5220–5227 Hiratsuka S, Watanabe A, Aburatani H, Maru Y, 2006, Tumourmediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nat Cell Biol 8:1369– 1375 Ring BZ, Ross DT, 2005, Predicting the sites of metastases. Genome Biol 6:241 Chang HY, Nuyten DS, Sneddon JB, Hastie T, Tibshirani R, Sorlie T, Dai H, He YD, Van’T VL, Bartelink H, van de Rijn M, Brown PO, van de Vijver MJ, 2005, Robustness, scalability, and integration of a wound-response gene expression signature in predicting breast cancer survival. Proc Natl Acad Sci U S A 102:3738–3743 Novak K, 2005, A healing process. Nat Rev Cancer 5(4):244–244 Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, Gerald WL, Massague J, 2008, Endogenous human microRNAs that suppress breast cancer metastasis. Nature 451:147–152 Midwood KS, Orend G, 2009, The role of tenascin-C in tissue injury and tumorigenesis. J Cell Commun Signal 3:287–310 Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, Lovat F, Fadda P, Mao C, Nuovo GJ, Zanesi N, Crawford M, Ozer GH, Wernicke D, Alder H, Caligiuri MA, Nana-Sinkam P, Perrotti D, Croce CM, 2012, MicroRNAs bind to toll-like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci U S A 109:E2110–E2116 Sung SY,Hsieh CL,Wu D, Chung LW, Johnstone PA, 2007, Tumor microenvironment promotes cancer progression, metastasis, and therapeutic resistance. Curr Probl Cancer 31:36–100 Lorusso G, Ruegg C, 2008, The tumor microenvironment and its contribution to tumor evolution toward metastasis. Histochem Cell Biol 130:1091–1103 Rofstad EK, 2000, Microenvironment-induced cancer metastasis. Int J Radiat Biol 76:589–605 MargarettenNC,WitschiHR, 1988, Effects of hyperoxia on growth of experimental lung metastasis. Carcinogenesis 9:433–439 Vaupel P, Kallinowski F, Okunieff P, 1989, Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: A review. Cancer Res 49:6449–6465 Rofstad EK, Halsor EF, 2002, Hypoxia-associated spontaneous pulmonary metastasis in human melanoma xenografts: Involvement of microvascular hot spots induced in hypoxic foci by interleukin 8. Br J Cancer 86:301–308 Rofstad EK, Mathiesen B, Kindem K, Galappathi K, 2006, Acidic extracellular pH promotes experimental metastasis of human melanoma cells in athymic nude mice. Cancer Res 66:6699–6707 Brizel DM, Scully SP, Harrelson JM, Layfield LJ, Bean JM, Prosnitz LR, Dewhirst MW, 1996, Tumor oxygenation predicts for the likelihood of distant metastases in human soft tissue sarcoma. Cancer Res 56:941–943 Budhu A, Forgues M, Ye QH, Jia HL, He P, Zanetti KA, Kammula US, Chen Y, Qin LX, Tang ZY, Wang XW, 2006, Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment. Cancer Cell 10:99–111 Ben-Eliyahu MBAS, 2010, Surgery as a double-edged sword: A clinically feasible approach to overcome the metastasis-promoting effects of surgery by blunting stress and prostaglandin responses. Cancers 2:1929–1951 Zhao T, Xia WH, Zheng MQ, Lu CQ, Han X, Sun YJ, 2008, Surgical excision promotes tumor growth and metastasis by promoting expression of MMP-9 and VEGF in a breast cancer model. Exp Oncol 30:60–64 Camphausen K, Moses MA, Beecken WD, Khan MK, Folkman J, O’Reilly MS, 2001, Radiation therapy to a primary tumor accelerates metastatic growth in mice. Cancer Res 61:2207–2211 von Essen CF, 1991, Radiation enhancement of metastasis: A review. Clin Exp Metastasis 9:77–104 Nathanson SD, Nelson L, Anaya P, Havstad S, Hetzel FW, 1991, Development of lymph node and pulmonary metastases after local irradiation and hyperthermia of footpad melanomas. Clin Exp Metastasis 9:377–392 Vikram B, Strong EW, Shah JP, Spiro R, 1984, Failure at distant sites followingmultimodality treatment for advanced head and neck cancer. Head Neck Surg 6:730–733 Demicheli R, Retsky MW, Hrushesky WJ, Baum M, Gukas ID, 2008, The effects of surgery on tumor growth: A century of investigations. Ann Oncol 19:1821–1828 Pidgeon GP, Harmey JH, Kay E, Da CM, Redmond HP, Bouchier- Hayes DJ, 1999, The role of endotoxin/lipopolysaccharide in surgically induced tumour growth in a murine model of metastatic disease. Br J Cancer 81:1311–1317 Al-Sahaf O,Wang JH, Browne TJ, Cotter TG, Redmond HP, 2010, Surgical injury enhances the expression of genes that mediate breast cancer metastasis to the lung. Ann Surg 252:1037–1043 Yan T, Yin W, Zhou L, Jiang Y, Shen Z, Shao Z, Lu J, 2010, Postoperative fever: The potential relationship with prognosis in node negative breast cancer patients. PLoS One 5:e15903 Nowacki MP, Szymendera JJ, 1983, The strongest prognostic factors in colorectal carcinoma. Surgicopathologic stage of disease and postoperative fever. Dis Colon Rectum 26:263–268 Lutgendorf SK, Sood AK, Antoni MH, 2010, Host factors and cancer progression: Biobehavioral signaling pathways and interventions. J Clin Oncol 28:4094–4099 Li S, Sun Y, Gao D, 2013, Role of the nervous system in cancer metastasis. Oncol Lett 5:1101–1111 Moreno-Smith M, Lutgendorf SK, SoodAK, 2010, Impact of stress on cancer metastasis. Future Oncol 6:1863–1881 Wu W, Yamaura T, Murakami K, Murata J, Matsumoto K, Watanabe H, Saiki I, 2000, Social isolation stress enhanced liver metastasis of murine colon 26-L5 carcinoma cells by suppressing immune responses in mice. Life Sci 66:1827–1838 WuW, Yamaura T,Murakami K, OgasawaraM, Hayashi K,Murata J, Saiki I, 1999, Involvement of TNF-alpha in enhancement of invasion and metastasis of colon 26-L5 carcinoma cells in mice by social isolation stress. Oncol Res 11:461–469 Sloan EK, Priceman SJ, Cox BF, Yu S, Pimentel MA, Tangkanangnukul V, Arevalo JM, Morizono K, Karanikolas BD, Wu L, Sood AK, Cole SW(2010, The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res 70:7042–7052 Chiang AC, Massague J, 2008, Molecular basis of metastasis. N Engl J Med 359:2814–2823 Benish M, Bartal I, Goldfarb Y, Levi B, Avraham R, Raz A, Ben- Eliyahu S, 2008, Perioperative use of beta-blockers and COX-2 inhibitors may improve immune competence and reduce the risk of tumor metastasis. Ann Surg Oncol 15:2042–2052 Ewertz M, Jensen MB, Gunnarsdottir KA, Hojris I, Jakobsen EH, Nielsen D, Stenbygaard LE, Tange UB, Cold S, 2011, Effect of obesity on prognosis after early-stage breast cancer. J ClinOncol 29: 25–31 Sestak I, Distler W, Forbes JF, Dowsett M, Howell A, Cuzick J, 2010, Effect of body mass index on recurrences in tamoxifen and anastrozole treated women: An exploratory analysis from the ATAC trial. J Clin Oncol 28:3411–3415 Sinicrope FA, Dannenberg AJ, 2011, Obesity and breast cancer prognosis: Weight of the evidence. J Clin Oncol 29:4–7 Nicolson GL, 1988, Organ specificity of tumor metastasis: Role of preferential adhesion, invasion and growth of malignant cells at specific secondary sites. Cancer Metastasis Rev 7:143–188 Okada F, Shionoya H, Kobayashi M, Kobayashi T, Tazawa H, Onuma K, Iuchi Y, Matsubara N, Ijichi T, Dugas B, Hosokawa M, 2006, Prevention of inflammation-mediated acquisition of metastatic properties of benign mouse fibrosarcoma cells by administration of an orally available superoxide dismutase. Br J Cancer 94: 854–862 Fidler IJ, Balasubramanian K, Lin Q, Kim SW, Kim SJ, 2010, The brain microenvironment and cancer metastasis.Mol Cells 30:93–98 Seike T, Fujita K, Yamakawa Y, Kido MA, Takiguchi S, Teramoto N, Iguchi H, Noda M, 2011, Interaction between lung cancer cells and astrocytes via specific inflammatory cytokines in the microenvironment of brain metastasis. Clin Exp Metastasis 28:13–25 Wang N, Thuraisingam T, Fallavollita L, Ding A, Radzioch D, Brodt P, 2006, The secretory leukocyte protease inhibitor is a type 1 insulin-like growth factor receptor-regulated protein that protects against liver metastasis by attenuating the host proinflammatory response. Cancer Res 66:3062–3070 Pantel K, Brakenhoff RH, 2004, Dissecting the metastatic cascade. Nat Rev Cancer 4:448–456 Alix-Panabieres C, Riethdorf S, Pantel K, 2008, Circulating tumor cells and bone marrow micrometastasis. Clin Cancer Res 14:5013– 5021 Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, Zhu Z, Hicklin D, Wu Y, Port JL, Altorki N, Port ER, Ruggero D, Shmelkov SV, Jensen KK, Rafii S, Lyden D, 2005, VEGFR1- Positive haematopoietic bone marrow progenitors initiate the premetastatic niche. Nature 438:820–827 Kaplan RN, Psaila B, Lyden D, 2006, Bone marrow cells in the ‘pre-metastatic niche’: Within bone and beyond. Cancer Metastasis Rev 25:521–529 Herzog EL, Chai L, Krause DS, 2003, Plasticity of marrow-derived stem cells. Blood 102:3483–3493 Uccelli A, Moretta L, Pistoia V, 2008, Mesenchymal stem cells in health and disease. Nat Rev Immunol 8:726–736 RojasM, Xu J,Woods CR,Mora AL, SpearsW, Roman J, Brigham KL, 2005, Bone marrow-derived mesenchymal stem cells in repair of the injured lung. Am J Respir Cell Mol Biol 33:145–152 YamadaM, Kubo H, Kobayashi S, Ishizawa K, NumasakiM, Ueda S, Suzuki T, Sasaki H, 2004, Bone marrow-derived progenitor cells are important for lung repair after lipopolysaccharide-induced lung injury. J Immunol 172:1266–1272 Kallis YN, Alison MR, Forbes SJ, 2007, Bone marrow stem cells and liver disease. Gut 56:716–724 Mareel M, Oliveira MJ, Madani I, 2009, Cancer invasion and metastasis: Interacting ecosystems. Virchows Arch 454:599–622 Allawi Z, Cuzick J, BaumM, 2011, Does trauma or an intercurrent surgical intervention lead to a short-term increase in breast cancer recurrence rates? Ann Oncol Holmes MD, Chen WY, Li L, Hertzmark E, Spiegelman D, Hankinson SE, 2010, Aspirin intake and survival after breast cancer. J Clin Oncol 28:1467–1472 Motoyama S, Miura M, Hinai Y, Maruyama K, Usami S, Saito H, Minamiya Y, Satoh S, Murata K, Suzuki T, Ogawa J, 2009, CRP genetic polymorphism is associated with lymph node metastasis in thoracic esophageal squamous cell cancer. Ann Surg Oncol 16: 2479–2485 Clinchy B, Fransson A, Druvefors B, Hellsten A, Hakansson A, Gustafsson B, Sjodahl R, Hakansson L, 2007, Preoperative interleukin-6 production by mononuclear blood cells predicts survival after radical surgery for colorectal carcinoma. Cancer 109: 1742–1749 Alcover J, Filella X, Luque P, Molina R, Izquierdo L, Auge JM, Alcaraz A, 2010, Prognostic value of IL-6 in localized prostatic cancer. Anticancer Res 30:4369–4372 Ganz PA, Cole SW(2011, Expanding our therapeutic options: Beta blockers for breast cancer? J Clin Oncol 29:2612–2616 Barron TI, Connolly RM, Sharp L, Bennett K, Visvanathan K, 2011, Beta blockers and breast cancer mortality: A populationbased study. J Clin Oncol 29:2635–2644 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 27 EP 42 DI 10.1007/s12253-013-9734-x PG 16 ER PT J AU Zhang, XM Ma, ZW Wang, Q Wang, JN Yang, JW Li, XD Li, H Men, TY AF Zhang, Xiao-Ming Ma, Zhong-Wei Wang, Qiang Wang, Jian-Ning Yang, Ji-Wei Li, Xian-Duo Li, Hao Men, Tong-Yi TI A New RNA-Seq Method to Detect the Transcription and Non-coding RNA in Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; RNA-seq; Transcription; Non-coding RNA; lncRNA ID Prostate cancer; RNA-seq; Transcription; Non-coding RNA; lncRNA AB Prostate cancer is a big killer in many regions especially American men, and this year, the diagnosed rate rises rapidly.We aimed to find the biomarker or any changing in prostate cancer patients. With the development of next generation sequencing, much genomic alteration has been found. Here, basing on the RNA-seq result of human prostate cancer tissue, we tried to find the transcription or non-coding RNA expressed differentially between normal tissue and prostate cancer tissue. 10 T sample data is the RNA-seq data for prostate cancer tissue in this study, we found the differential gene is TFF3-Trefoil factor 3, which was more than seven fold change from prostate cancer tissue to normal tissue, and the most outstanding transcript is C15orf21. Additionally, 9 lncRNAs were found according our method. Finally, we found the many important non-coding RNA related to prostate cancer, some of them were long non-coding RNA (lncRNA). C1 [Zhang, Xiao-Ming] Qianfoshan Hospital Affiliated to Shandong University, Department of Urology, No.16766 Jingshi Road, 250014 Jinan, Shandong Province, China. [Ma, Zhong-Wei] Hospital of Shandong Aluminum Corporation, Department of Urology, 255069 Zibo, China. [Wang, Qiang] 309 Hospital of the Chinese People’s Liberation Army, Department of Urology, 100091 Beijing, China. [Wang, Jian-Ning] Qianfoshan Hospital Affiliated to Shandong University, Department of Urology, No.16766 Jingshi Road, 250014 Jinan, Shandong Province, China. [Yang, Ji-Wei] Qianfoshan Hospital Affiliated to Shandong University, Department of Urology, No.16766 Jingshi Road, 250014 Jinan, Shandong Province, China. [Li, Xian-Duo] Qianfoshan Hospital Affiliated to Shandong University, Department of Urology, No.16766 Jingshi Road, 250014 Jinan, Shandong Province, China. [Li, Hao] Hospital of Shandong Aluminum Corporation, Department of Urology, 255069 Zibo, China. [Men, Tong-Yi] Qianfoshan Hospital Affiliated to Shandong University, Department of Urology, No.16766 Jingshi Road, 250014 Jinan, Shandong Province, China. RP Men, TY (reprint author), Qianfoshan Hospital Affiliated to Shandong University, Department of Urology, 250014 Jinan, China. EM mentongyish3316@sina.com CR Berger MF, Lawrence MS, Demichelis F, Drier Y, Cibulskis K, Sivachenko AY, Sboner A, Esgueva R, Pflueger D, Sougnez C, 2011, The genomic complexity of primary human prostate cancer. Nature 470(7333):214–220 Barbieri CE, Baca SC, Lawrence MS, Demichelis F, Blattner M, Theurillat JP, White TA, Stojanov P, Van Allen E, Stransky N, 2012, Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet 44(6):685–689 Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, Quist MJ, Jing X, Lonigro RJ, Brenner JC, 2012, The mutational landscape of lethal castration-resistant prostate cancer. Nature 487(7406):239–243 Morin RD, Bainbridge M, Fejes A, Hirst M, Krzywinski M, Pugh TJ, McDonald H, Varhol R, Jones SJM, Marra MA, 2008, Profiling the HeLa S3 transcriptome using randomly primed cDNA and massively parallel short-read sequencing. Biotechniques 45(1):81–94 Maher CA, Kumar-Sinha C, Cao X, Kalyana-Sundaram S, Han B, Jing X, Sam L, Barrette T, Palanisamy N, Chinnaiyan AM, 2009, Transcriptome sequencing to detect gene fusions in cancer. Nature 458(7234):97–101 Pflueger D, Terry S, Sboner A, Habegger L, Esgueva R, Lin PC, Svensson MA, Kitabayashi N, Moss BJ, MacDonald TY, 2011, Discovery of non-ETS gene fusions in human prostate cancer using next-generation RNA sequencing. Genome Res 21(1):56–67 Sahu A, Iyer MK, Chinnaiyan AM, 2012, Insights into Chinese prostate cancer with RNA-seq. Cell Res 22(5):786–788 Mattick JS, 2001, Non-coding RNAs: the architects of eukaryotic complexity. EMBO reports 2(11):986–991 Stefani G, Slack FJ, 2008, Small non-coding RNAs in animal development. Nat Rev Mol Cell Biol 9(3):219–230 Mercer TR, Dinger ME, Mattick JS, 2009, Long non-coding RNAs: insights into functions. Nat Rev Genet 10(3):155–159 Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, 2010, Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464(7291):1071–1076 Ren S, Peng Z, Mao JH, Yu Y, Yin C, Gao X, Cui Z, Zhang J, Yi K, Xu W, 2012, RNA-seq analysis of prostate cancer in the Chinese population identifies recurrent gene fusions, cancer-associated long noncoding RNAs and aberrant alternative splicings. Cell Res 22(5):806–821 Leinonen R, Akhtar R, Birney E, Bower L, Cerdeno-Tarraga A, Cheng Y, Cleland I, Faruque N, Goodgame N, Gibson R, 2011, The European nucleotide archive. Nucleic Acids Res 39(suppl 1):D28–D31 Trapnell C, Pachter L, Salzberg SL, 2009, TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25(9):1105–1111 Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, Pimentel H, Salzberg SL, Rinn JL, Pachter L, 2012, Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc 7(3):562–578 Jiang H, Wong WH, 2009, Statistical inferences for isoform expression in RNA-Seq. Bioinformatics 25(8):1026–1032 Toung JM, Morley M, Li M, Cheung VG, 2011, RNA-sequence analysis of human B-cells. Genome Res 21(6):991–998 Garraway IP, Seligson D, Said J, Horvath S, Reiter RE, 2004, Trefoil factor 3 is overexpressed in human prostate cancer. Prostate 61(3):209–214 Laxman B, Morris DS, Yu J, Siddiqui J, Cao J, Mehra R, Lonigro RJ, Tsodikov A, Wei JT, Tomlins SA, 2008, A first-generation multiplex biomarker analysis of urine for the early detection of prostate cancer. Cancer Res 68(3):645 Vestergaard EM, Borre M, Poulsen SS, Nexo E, Torring N, 2006, Plasma levels of trefoil factors are increased in patients with advanced prostate cancer. Clin Cancer Res 12(3):807–812 AtherMH, Abbas F, Faruqui N, IsrarM, Pervez S, 2004, Expression of pS2 in prostate cancer correlates with grade and Chromogranin A expression but not with stage. BMC Urol 4(1):14 Tomlins SA, Laxman B, Dhanasekaran SM, Helgeson BE, Cao X, Morris DS, Menon A, Jing X, Cao Q, Han B, 2007, Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature 448(7153):595–599 Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A, Rinn JL, 2011, Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 25(18):1915–1927 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 43 EP 50 DI 10.1007/s12253-013-9618-0 PG 8 ER PT J AU Lu, C Liu, G Cui, X Zhang, J Wei, L Wang, Y Yang, X Liu, Y Cong, X Lv, L Ni, R Huang, X AF Lu, Cuihua Liu, Guoliang Cui, Xiaopeng Zhang, Jing Wei, Lixian Wang, Yingying Yang, Xiaojing Liu, Yanhua Cong, Xia Lv, Liting Ni, Runzhou Huang, Xiaodong TI Expression of SGTA Correlates with Prognosis and Tumor Cell Proliferation in Human Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Small glutamine-rich TPR-containing protein A (SGTA); Prognosis; Proliferation; Cell cycle ID Hepatocellular carcinoma; Small glutamine-rich TPR-containing protein A (SGTA); Prognosis; Proliferation; Cell cycle AB To investigate the potential role of small glutaminerich TPR-containing protein A (SGTA) in hepatocarcinogenesis, immunohistochemistry and Western blot were performed to detect the expression of SGTA in clinical Hepatocellular carcinoma (HCC) samples, adjacent nontumorous liver tissues and HCC cell lines. In addition, expression of SGTA was correlated with clinicopathological variables and univariate andmultivariate survival analyses were performed to determine the prognostic significance. Moreover, the biological significance of the aberrant expression of SGTA was investigated in vitro. Both immunohistochemistry evaluation and Western blot analyses demonstrated that SGTA was overexpressed in HCC tissues compared with adjacent nontumorous liver tissues. Expression of SGTA directly correlated with the histological grades of HCC and high expression of SGTA was associated with a poor prognosis. SGTA depletion by siRNA inhibited cell proliferation, blocked S-phase and mitotic entry in Huh7 cells. Western blot analyses showed that SGTA depletion decreased cyclin A and cyclin B levels. Taken together, owing to overexpression of SGTA in HCC and its important role in predicting poor prognosis and the development of HCC, SGTA could be a potential prognostic marker and therapeutic target of HCC. C1 [Lu, Cuihua] Medical College of Nantong University, Affiliated Hospital of Nantong University, Department of Digestion, 226001 Nantong, Jiangsu, China. [Liu, Guoliang] Medical College of Nantong University, Affiliated Hospital of Nantong University, Department of Digestion, 226001 Nantong, Jiangsu, China. [Cui, Xiaopeng] Medical College of Nantong University, Affiliated Hospital of Nantong University, Department of Digestion, 226001 Nantong, Jiangsu, China. [Zhang, Jing] Medical College of Nantong University, Affiliated Hospital of Nantong University, Department of Digestion, 226001 Nantong, Jiangsu, China. [Wei, Lixian] Medical College of Nantong University, Affiliated Hospital of Nantong University, Department of Digestion, 226001 Nantong, Jiangsu, China. [Wang, Yingying] Medical College of Nantong University, Department of Immunology and Microbiology, 226001 Nantong, Jiangsu, China. [Yang, Xiaojing] Medical College of Nantong University, Department of Immunology and Microbiology, 226001 Nantong, Jiangsu, China. [Liu, Yanhua] Medical College of Nantong University, Affiliated Hospital of Nantong University, Department of Digestion, 226001 Nantong, Jiangsu, China. [Cong, Xia] Medical College of Nantong University, Affiliated Hospital of Nantong University, Department of Digestion, 226001 Nantong, Jiangsu, China. [Lv, Liting] Medical College of Nantong University, Department of Immunology and Microbiology, 226001 Nantong, Jiangsu, China. [Ni, Runzhou] Medical College of Nantong University, Affiliated Hospital of Nantong University, Department of Digestion, 226001 Nantong, Jiangsu, China. [Huang, Xiaodong] Medical College of Nantong University, Affiliated Hospital of Nantong University, Department of Digestion, 226001 Nantong, Jiangsu, China. RP Ni, R (reprint author), Medical College of Nantong University, Affiliated Hospital of Nantong University, Department of Digestion, 226001 Nantong, China. EM nirunzhou@sina.cn CR Zhou P,Wu LL,Wu KM et al, 2012, Overexpression of MMSET is correlation with poor prognosis in hepatocellular carcinoma. Pathol Oncol Res 19(2):303–9 Xia LM, Huang WJ, Tian DA et al, 2012, Overexpression of forkhead box C1 promotes tumor metastasis and indicates poor prognosis in hepatocellular carcinoma. Hepatology 57(2):610–24 Guo XD, Xiong L, Zou L, Zhao JM, 2012, Upregulation of bone morphogenetic protein 4 is associated with poor prognosis in patients with hepatocellular carcinoma. Pathol Oncol Res 18(3):635–640 Yang H, Xiong FX, Lin M, Yang Y, Nie X, Zhou RL, 2010, LAPTM4B-35 overexpression is a risk factor for tumor recurrence and poor prognosis in hepatocellular carcinoma. J Cancer Res Clin Oncol 136(2):275–281 Zhao ZK, Dong P, Gu J, et al, 2012, Overexpression of LSD1 in hepatocellular carcinoma: a latent target for the diagnosis and therapy of hepatoma. Tumour Biol 34(1):173–80 Callahan MA, Handley MA, Lee YH, Talbot KJ, Harper JW, Panganiban AT, 1998, Functional interaction of human immunodeficiency virus type 1 Vpu and Gag with a novel member of the tetratricopeptide repeat protein family. J Virol 72(10):8461 Cziepluch C, Kordes E, Poirey R, Grewenig A, Rommelaere J, Jauniaux JC, 1998, Identification of a novel cellular TPRcontaining protein, SGT, that interacts with the nonstructural protein NS1 of parvovirus H-1. J Virol 72(5):4149–4156 Kordes E, Savelyeva L, Schwab M, Rommelarere J, Jauniaux JC, Cziepluch C, 1998, Isolation and characterization of human SGT and identification of homologues in Saccharomyces cerevisiae and Caenorhabditis elegans. Genomics 52(1):90–94 Wang HZ, Shen HL,Wang YL et al, 2005, Overexpression of small glutamine-rich TPR-containing protein promotes apoptosis in 7721 cells. FEBS Lett 579(5):1279–1284 Buchanan GB, Ricciardelli C, Jonathan M et al, 2007, Control of androgen receptor signaling in prostate cancer by the cochaperone small glutamine rich tetratricopeptide repeat containing protein alpha. Cancer Res 67(20):10087–10096 Moritz A, Li Y, Guo AL et al, 2010, Akt-RSK-S6 kinase signaling networks activated by oncogenic receptor tyrosine kinases. Sci Signal 3(136):ra64 Mochida S, Hunt T, 2012, Protein phosphatases and their regulation in the control of mitosis. EMBO Rep 13(3):197–203 Winnefeld M, Rommelaere J, Cziepluch C, 2004, The human small glutamine-rich TPR-containing protein is required for progress through cell division. Exp Cell Res 293(1):43–57 Winnefeld M, Grewenig A, Schnolzer M et al, 2006, Human SGT interacts with Bag-6/Bat-3/Scythe and cells with reduced levels of either protein display persistence of few misaligned chromosomes and mitotic arrest. Exp Cell Res 312(13):2500–2514 Chan KS, Koh CG, Li HY, 2012, Mitosis-targeted anti-cancer therapies: where they stand. Cell Death Dis 3:e411 He J, Wu J, Xu N et al, 2012, MiR-210 disturbs mitotic progression through regulating a group ofmitosis-related genes. Nucleic Acids Res 41(1):498–508 Lu MD, Ma JB, XueWQet al, 2009, The expression and prognosis of FOXO3a and Skp2 in human hepatocellular carcinoma. Pathol Oncol Res 15(4):679–687 Ke Q, Ji JL, Cheng C et al, 2009, Expression and prognostic role of Spy1 as a novel cell cycle protein in hepatocellular carcinoma. Exp Mol Pathol 87(3):167–172 Wang YC, Liu F, Mao F et al, 2013, Interaction with CCNH/CDK7 stabilizes CtBP2 and promotes cancer cell migration. J Biol Chem 288(13):9028–9034 Deng Q,Wang Q, ZongWYet al, 2010, E2F8 contributes to human hepatocellular carcinoma via regulating cell proliferation. Cancer Res 70(2):782–791 Smith A, Simanski S, Fallahi M, Ayad NG, 2007, Redundant ubiquitin ligase activities regulate wee1 degradation and mitotic entry. Cell Cycle 6(22):2795–2799 McFarlane C, Kelvin AA, de la Vega M et al, 2010, The deubiquitinating enzyme USP17 is highly expressed in tumor biopsies, is cell cycle regulated, and is required for G1-S progression. Cancer Res 70(8):3329–3339 Uchida F, Uzawa K, Kasamatsu A et al, 2012, Overexpression of cell cycle regulator CDCA3 promotes oral cancer progression by enhancing cell proliferation with prevention of G1 phase arrest. BMC Cancer 12:321 Hur W, Rhim H, Jung CK et al, 2010, SOX4 overexpression regulates the p53-mediated apoptosis in hepatocellular carcinoma: clinical implication and functional analysis in vitro. Carcinogenesis 31(7):1298–1307 Miyai K, Yamamoto S, Iwaya K et al, 2012, Altered expression of p27(Kip1, -interacting cell-cycle regulators in the adult testicular germ cell tumors: potential role in tumor development and histological progression. APMIS 120(11):890–900 Demonacos C, Krstic-Demonacos M, La Thangue NB et al, 2001, A TPR motif cofactor contributes to p300 activity in the p53 response. Mol Cell 8(1):71–84 Xiang SL, Kumano T, Iwasaki SI, Sun X, Yoshioka K, Yamamoto KC, 2001, The J domain of Tpr2 regulates its interaction with the proapoptotic and cell-cycle checkpoint protein, Rad9. Biochem Biophys Res Commun 287(4):932–940 Imai Y, Soda M, Hatakeyama S et al, 2002, CHIP is associated with Parkin, a gene responsible for familial Parkinson’s disease, and enhances its ubiquitin ligase activity. Mol Cell 10(1):55–67 Dai Q, Zhang C, Wu Y et al, 2003, CHIP activates HSF1 and confers protection against apoptosis and cellular stress. EMBO J 22(20):5446–5458 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 51 EP 60 DI 10.1007/s12253-013-9657-6 PG 10 ER PT J AU Doki, N Ohashi, K Oshikawa, G Kobayashi, T Kakihana, K Sakamaki, H AF Doki, Noriko Ohashi, Kazuteru Oshikawa, Gaku Kobayashi, Takeshi Kakihana, Kazuhiko Sakamaki, Hisashi TI Clinical Outcome of Hematopoietic Stem Cell Transplantation for Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia (Ph + ALL): Experience From a Single Institution SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Philadelphia Chromosome-positive acute lymphoblastic leukemia (Ph + ALL); Allogeneic hematopoietic stem cell transplantation (allo-HSCT); Tyrosine kinase inhibitor (TKI); Complete remission (CR) ID Philadelphia Chromosome-positive acute lymphoblastic leukemia (Ph + ALL); Allogeneic hematopoietic stem cell transplantation (allo-HSCT); Tyrosine kinase inhibitor (TKI); Complete remission (CR) AB To identify factors affecting transplant outcome, data from 65 Philadelphia Chromosome-positive acute lymphoblastic leukemia (Ph + ALL) patients who had undergone allogeneic hematopoietic transplantation (allo-HSCT) in our institution were analyzed. The probability of OS (overall survival) and DFS (disease free-survival) at 3 years after allo- HSCT was 40.1 % and 38 %, respectively. Multivariate analysis showed that gender and disease status (p=0.0059, p=0.0039, respectively) were significant factors for OS. Among 51 patients with CR (complete remission), multivariate analysis showed that the factors associated with OS included gender (p=0.014), number of white blood cell at diagnosis (p=0.025), and the source of stem cells (bone marrow versus. cord blood; BM stem cell source was associated with favorable OS, p=0.042). Twenty-one patients relapsed after allo-HSCT with a median of 207 days (range, 19–1,324 days). The estimated cumulative incidence of relapse at 3 years was 39.4%. Patients with CR showed a lower relapse rate at 3 years (34.2 %) when compared with patients with non-CR (62.7 %). Among 21 patients, eight patients received imatinib-based chemotherapy and 13 received chemotherapy without imatinib before HSCT. In terms of treatment after relapse, seven patients received chemotherapy with imatinib and 13 received chemotherapy without imatinib. Five patients underwent a second HSCT. One patient survived, and 20 patients died. In this study, disease status at time of allo-HSCT had a significant impact on OS, DFS, and relapse. Imatinib administration given before allo-HSCT was not associated with favorable outcome. Second-generation tyrosine kinase inhibitors may be more suitable candidates for Ph + ALL before and after allo-HSCT. C1 [Doki, Noriko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Ohashi, Kazuteru] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Oshikawa, Gaku] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kobayashi, Takeshi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kakihana, Kazuhiko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Sakamaki, Hisashi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. RP Doki, N (reprint author), Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 113-8677 Tokyo, Japan. EM n-doki@cick.jp CR Gleissner B, Gokbuget N, Bartram CR et al, 2002, Leading prognostic relevance of the BCR-ABLtranslocation in adult acute Blineage lymphoblastic leukemia: a prospective study of the German Multicenter Traial Group and confirmed polymerase chain reaction analysis. Blood 99:1536–1543 Vinetti M, Fazi P, Cimino G et al, 2007, Imatinib plus steroids induces complete remissons and prolonged survival in elderly Philadelphia chromosome-positive patients with acute lymphoblastic leukemia without additional chemotherapy:results of the Gruppo Italiano Malattie Ematologiche dell’Adulto, GI-MEMA, LAL0201-B protocol. Blood 109:3676–3678 Lee S, Kim YJ, Min CK et al, 2005, The effect of first-line imatinib interim therapy on the outcome of allogeneic stem cell transplantation in adults with newly diagnosed Philadelphia chromosomepositive acute lymphoblastic leukemia. Blood 105:3449–3457 Lee HJ, Thompson JE, Wang ES et al, 2011, Philadelphia chromosome-positive acute lymphoblastic leukemia: current treatment and future perspectives. Cancer 117:1583–1594 Yanada M, Naoe T, Iida H et al, 2005, Myeloablative allogeneic hematopoietic stem cell transplantation for Philadelphia chromosomepositive acute lymphoblastic leukemia in adults: significant roles of total body irradiation and chronic graft-versus-host disease. BMT 26:867–872 Kebriaei P, Saliba R, Rondon G et al, 2012, Long-term follow-up of allogeneic hematopoietic stem cell transplantation for patients with Philadelphia chromosome-positive acute lymphoblastic leukemia: impact of tyrosine kinase inhibitors on treatment outcomes. Biol Blood Marrow Transplant 18:584–592 Fielding AK, Rowe JM, Richards SM et al, 2009, Prospective outcome data on 267 unselected adult patients with Philadelphia chromosome-positive acute lymphoblastic leukemia confirms superiority of allogeneic transplantation over chemotherapy in the pre-imatinib era: results from the International ALL Trial MRC UKALLXII/ECOG 2993. Blood 113:4489–4496 Mizuta S, Matsuo K, Yagasaki F et al, 2011, Pre-transplant imatinib-based therapy improves the outcome of allogeneic hematopoietic stem cell transplantation for BCR-ABL-positive acute lymphoblastic leukemia. Leukemia 25:41–47 Mizuta S, Matsuo K, Maeda T et al, 2012, Prognostic factors influencing clinical outcome of allogeneic hematopoietic stem cell transplantation following imatinib-based therapy in BCR-ABLpositive ALL. Blood Cancer J 2:e72 Spinelli O, Peruta B, Tosi M et al, 2007, Clearance of minimal residual disease after allogeneic stem cell transplantation and the prediction of the clinical outcome of adult patints with high-risk acute lymphoblastic leukemia. Haematologica 92:612–618 Burke MJ, Troz B, Baker KS et al, 2009, Allo-hematopoietic cell transplantation for Ph chromosome-positive ALL: impact of imatinib on relapse and survival. BMT 43:107–113 Carpenter PA, Snyder DS, Flowers ME et al, 2007, Prophilactic administration of imatinib after hematopoietic cell transplantation for high-risk Philadelphia chromosome-positive leukemia. Blood 109:2791–2793 O’Hare T, Walters DK, Stoffregen EP et al, 2005, In vitro activity of Bcr-Abl inhibitors AMN107 and BMS-354825 against clinically relevant imatinib-resistant Abl kinase domain mutants. Cancer Res 65:4500–4505 Ravandi F, O’Brein S, Thomas D et al, 2010, First report of phase 2 study of dasatinib with hyper-CVAD for the frontline treatment of patients with Philadelphia chromosome-positive, Ph+, acute lymphoblastic leukemia. Blood 116:2070–2077 Foa R, Vitale A, Vifnwrri M et al, 2011, Dasatinib as first-line treatment for adult patients with Philadelphia chromosome-positive acute lymphoblastic leukemia. Blood 118:6521–6528 Caocci G, Vacca A, Ledda A et al, 2012, Prophylactic and preemptive therapy with dasatinib after hematopoietic stem cell transplantation for Philadelphia chromosome-positive acute lymphoblastic leukemia. Biol Blood Marrow Transplant 18:652–654 Teng CL, Yu JT, Chen HC, Hwang WL, 2013, Maintenance therapy with dasatinib after allogeneic hematopoietic stem cell transplantation in Philadelphia chromosome-positive acute lymphoblastic leukemia. Ann Hematol., DOI 10.1007/s00277−012-1670-4 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 61 EP 66 DI 10.1007/s12253-013-9658-5 PG 6 ER PT J AU Wozniak, K Krupa, R Synowiec, E Morawiec, Z AF Wozniak, Katarzyna Krupa, Renata Synowiec, Ewelina Morawiec, Zbigniew TI Polymorphism of UBC9 Gene Encoding the SUMO-E2- Conjugating Enzyme and Breast Cancer Risk SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE UBC9 gene and protein; Breast cancer; Sumoylation ID UBC9 gene and protein; Breast cancer; Sumoylation AB UBC9 protein (E2-conjugating enzyme) plays a key role in post-translation modification named sumoylation. Proteins, which are sumoylated take part in many cellular processes including cell growth, maintaining the genome integrity and stability and cancer development. The aim of this study was to investigate an association between three polymorphisms of the UBC9 gene: c.73G>A (rs11553473), c.430T>G (rs75020906) and g.1289209T>C (rs7187167) and a risk of ductal breast cancer occurrence. We performed a case-control study in 181 breast cancer cases and 277 controls using PCR-RLFP and ASO-PCR. In the case of the 430T>G polymorphism of the UBC9 gene lack of variability suggests that there is not a polymorphic site in polish population. We observed that a risk of breast cancer occurrence is elevated in patients with the G/A genotype (OR 5.03; 95 % Cl 3.05–8.28), the A/A genotype (OR 11.3; 95 % Cl 4.24–30.3) and the A allele (OR 6.86; 95 % Cl 4.43–10.6) of the c.73G>A polymorphism. In the case of the g.1289209T>C polymorphism we found a correlation between estrogen receptor (ER) expression and the T/T genotype (OR 0.22; 95 % Cl 0.07–0.64) and the T allele (OR 0.53; 95 % Cl 0.32–0.88). We also found a correlation between the T/T genotype (OR 4.13; 95 % Cl 1.21–14.1) and the T allele (OR 2.09; 95 % Cl 1.07–4.08) of the g.1289209T>C polymorphism with triple negative breast cancer. Our results suggest that the variability of the UBC9 gene can play a role in breast cancer occurrence. C1 [Wozniak, Katarzyna] University of Lodz, Department of Molecular GeneticsLodz, Poland. [Krupa, Renata] University of Lodz, Department of Molecular GeneticsLodz, Poland. [Synowiec, Ewelina] University of Lodz, Department of Molecular GeneticsLodz, Poland. [Morawiec, Zbigniew] N. Copernicus Hospital, Department of Surgical OncologyLodz, Poland. RP Wozniak, K (reprint author), University of Lodz, Department of Molecular Genetics, Lodz, Poland. EM wozniak@biol.uni.lodz.pl CR Seeler J-S, Bischof O, Nacerddine K, Dejean A, 2007, SUMO, the three rs and cancer. CTMI 313:49–71 Gill G, 2004, SUMO and ubiquitin in the nucleus: different functions, similar mechanisms? Genes Dev 18:2046–2259 Liu LB, Omata W, Kojima I, Shibata H, 2007, The SUMO conjugating enzyme Ubc9 is a regulator of GLUT4 turnover and targeting to the insulin-responsive storage compartment in 3T3-1 adipocytes. Diabetes 56:1977–1985 Zhu S, Sachdeva M, Wu F, Lu Z, Mo YY, 2010, Ubc9 promotes breast cell invasion and metastasis in a sumoylation-independent manner. Oncogene 29:1763–1772 Ronen O, Malone JP, Kay P, Bivens C, Hall K, Paruchuri LP,Mo YY, Robbins KT, Ran S, 2009, Expression of a novel marker, Ubc9, in squamous cell carcinoma of the head and neck. Head Neck 31:845–855 Driscoll JJ, Pelluru D, Lefkimmiatis K, Fulciniti M, Prabhala RH, Greipp PR, Barlogie B, Tai YT, Anderson KC, Shaughnessy JD Jr, Annunziata CM, Munshi NC, 2010, The sumoylation pathway is dysregulated in multiple myeloma and is associated with adverse patient outcome. Blood 115:2827–2834 Moschos SJ, Jukic DM, Athanassiou C, Bhargava R, Dacic S, Wang X, Kuan SF, Fayewicz SL, Galambos C, Acquafondata M, Dhir R, Becker D, 2010, Expression analysis of Ubc9, the single small ubiquitin-like modifier, SUMO, E2 conjugating enzyme, in normal and malignant tissues. Hum Pathol 9:1286–1298 Wu F, Zhu S, Ding Y, Beck WT, Mo YY, 2009, MicroRNAmediated regulation for Ubc9 expression in cancer cells. Clin Cancer Res 15:1550–1557 Li Y, Lu J, Prochownik EV, 2007, Dual role for SUMO E2 conjugase Ubc9 in modulating the transforming and growthpromoting properties of the HMGA1b architectural transcription factor. J Biol Chem 282:13363–13371 Poukka H, Karvonen U, Janne OA, Palvimo JJ, 2000, Covalent modification of the androgen receptor by small ubiquitin-like modifier 1, SUMO-1). Proc Natl Acad Sci U S A 97:14145–14150 Sentis S, Le Romancer M, Bianchin C, Rostan MC, Corbo L, 2005, Sumoylation of the estrogen receptor alpha hinge region regulates its transcriptional activity. Mol Endocrinol 19:2671–2684 Abdel-Hafiz HA, Horwitz KB, 2012, Control of progesterone receptor transcriptional synergy by SUMOylation and deSUMOylation. BMC Mol Biol 13:, DOI 10.1186/1471-2199-13-10 Galanty Y, Belotserkovskaya R, Coates J, Polo S, Miller KM, Jackson SP, 2009, Mammalian SUMO E3-ligases PIAS1 and PIAS4 promote responses to DNA double-strand breaks. Nature 462:935–940 Morris JR, Boutell C, Keppler M, Densham R, Weekes D, Alamshah A, Butler L, Galanty Y, Pangon L, Kiuchi T, Ng T, Solomon E, 2009, The SUMO modification pathway is involved in the BRCA1 response to genotoxic stress. Nature 462:886–891 Thompson LH, Schild D, 2002, Recombinational DNA repair and human disease. Mutat Res 509:49–78 Hsu HM, Wang HC, Chen ST, Hsu GC, Shen CY, Yu JC, 2007, Breast cancer risk is associated with the genes encoding the DNA double-strand break repair Mre11/Rad50/Nbs1 complex. Cancer Epidemiol Biomarkers Prev 16:2024–2032 Ralhan R, Kaur J, Kreienberg R, Wiesmuller L, 2007, Links between DNA double strand break repair and breast cancer: accumulating evidence from both familial and nonfamilial cases. Cancer Lett 248:1–17 Mao Z, Jiang Y, Liu X, Seluanov A, Gorbunova V, 2009, DNA repair by homologous recombination, but not by non-homologous end joining, is elevated in breast cancer cells. Neoplasia 11:683– 691 Qin Y, Xu J, Aysola K, Begum N, Reddy V, Chai Y, Grizzle WE, Partridge EE, Reddy ESP, Rao VN, 2011, Ubc9 mediates nuclear localization and growth suppression of BRCA1 and BRCA1a proteins. J Cell Physiol 226:3355–3367 Anders CK, Carey LA, 2009, Biology, metastatic patterns, and treatment of patients with triple-negative breast cancer. Clin Breast Cancer 9:S73–S81 de Ruijter TC, Veeck J, de Hoon JPJ, van Engeland M, Tjan- Heijnen VC, 2011, Characteristics of triple-negative breast cancer. J Cancer Res Clin Oncol 137:183–192 Ratanaphan A, 2012, A DNA repair BRCA1 estrogen receptor and targeted therapy in breast cancer. Int J Mol Sci 13:14898–14916 Xu J, Watkins T, Reddy A, Reddy ESP, Rao VN, 2009, A novel mechanism whereby BRCA1/1a/1b fine tunes the dynamic complex interplay between SUMO-dependent/independent activities of Ubc9 on E2-induced ERα activation/repression and degradation in breast cancer cells. Int J Oncol 34:939–949 Dunnebier T, Bermejo JL, Haas S, Fischer HP, Pierl CB, Justenhoven C, Brauch H, Baisch C, Gilbert M, Harth V, Spickenheuer A, Rabstein S, Pesch B, Bruning T, Ko YD, Hamann U, 2009, Common variants in the UBC9 gene encoding the SUMO-conjugating enzyme are associated with breast tumor grade. Int J Cancer 125:596–602 Dunnebier T, Bermejo JL, Haas S, Fischer HP, Pierl CB, Justenhoven C, Brauch H, Baisch C, Gilbert M, Harth V, Spickenheuer A, Rabstein S, Pesch B, Bruning T, Ko YD, Hamann U, 2010, Polymorphisms in the UBC9 and PIAS3 genes of the SUMO-conjugating system and breast cancer risk. Breast Cancer Res Treat 121:185–194 Synowiec E, Krupa R, Morawiec Z,WasyleckaM, Dziki L, Morawiec J, Blasiak J,Wozniak K, 2010, Efficacy of DNA double-strand breaks repair in breast cancer is decreased in carriers of the variant allele of the UBC9 gene c.73G>A polymorphism. Mutat Res 694:31–38 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 67 EP 72 DI 10.1007/s12253-013-9659-4 PG 6 ER PT J AU Zhou, F Xu, J Ding, G Cao, L AF Zhou, Fan Xu, Jiewei Ding, Guoping Cao, Liping TI Overexpressions of CK2β and XIAP are Associated with Poor Prognosis of Patients with Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Protein kinase CK2; Casein kinase 2; X-Linked inhibitor of apoptosis protein; Cholangiocarcinoma; Prognosis ID Protein kinase CK2; Casein kinase 2; X-Linked inhibitor of apoptosis protein; Cholangiocarcinoma; Prognosis AB To investigate the expressions of casein kinase II β(CK2β) and X-Linked inhibitor of apoptosis protein (XIAP) in cholangiocarcinoma (CCA) and evaluated their correlations with major clinicopathologic features and patients’ survival. Fifty CCA specimens and 20 normal liver tissues were included in the study. Immunohistochemical staining was used to determine the expression levels of CK2β, XIAP in normal and CCA tissues. The relationships of CK2β and XIAP expressions with clinicopathologic parameters and clinical outcome were evaluated. High immunostaining of CK2β and XIAP were observed in 66 % (33/50) and 68 % (34/50) of CCA tissues, which were significantly higher than that of normal liver tissues 0 % (0/20) and 25 % (5/20). The high expression of CK2β was significantly associated with TNM stage (P=0.036), histological grade (P=0.020) and high serum CEA level(P=0.010), while high expression of XIAP was only associated with TNM stage(P=0.014) and high serum CEA level(P=0.001). By univariant analysis, patients with high expression of CK2β and XIAP demonstrate significantly poorer overall survival (P=0.003 vs P=0.018). Cox regression model showed that positive expression of CK2βis an independent factor of prognosis (P=0.004). The expressions of CK2β and XIAP in CCA tissues showed strong correlations with the tumor progression, CK2β may be applied as a potential prognostic marker for CCA. C1 [Zhou, Fan] School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. [Xu, Jiewei] School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. [Ding, Guoping] School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. [Cao, Liping] School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. RP Cao, L (reprint author), School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. EM cao@zju.edu.cn CR Blechacz B, Gores GJ, 2008, Cholangiocarcinoma: advances in pathogenesis, diagnosis, and treatment. Hepatology 48:308–321., DOI 10.1002/hep.22310 Hsing AW, Gao YT, Han TQ et al, 2007, Gallstones and the risk of biliary tract cancer: a population-based study in China. Br J Cancer 97:1577–1582., DOI 10.1038/sj.bjc.6604047 Shaib YH, Davila JA, McGlynn K et al, 2004, Rising incidence of intrahepatic cholangiocarcinoma in the United States: a true increase? J Hepatol 40:472–477., DOI 10.1016/j.jhep.2003.11.030 Patel T, 2001, Increasing incidence and mortality of primary intrahepatic cholangiocarcinoma in the United States. Hepatology 33:1353–1357., DOI 10.1053/jhep. 2001.25087 Zhang P, Davis AT, Ahmed K, 1998, Mechanism of protein kinase CK2 association with nuclear matrix: role of disulfide bond formation. J Cell Biochem 69:211–220 Graham KC, Litchfield DW, 2000, The regulatory β subunit of protein kinase CK2 mediates formation of tetrameric CK2 complexes. J Biol Chem 275:5003–5010 Ahmed K, 1999, Nuclear matrix and protein kinase CK2 signaling. Crit Rev Eukaryot Gene Expr 9:329–336 Tawfic S, Faust RA, Gapany M et al, 1996, Nuclear matrix as an anchor for protein kinase CK2 nuclear signalling. J Cell Biochem 62:165–171 Guo C, Yu S, Davis AT et al, 2001, A potential role of nuclear matrix-associated protein kinase CK2 in protection against druginduced apoptosis in cancer cells. J Biol Chem 276:5992–5999., DOI 10.1074/jbc.M004862200 Ahmad KA,Wang G, Unger G et al, 2008, Protein Kinase CK2 - A key suppressor of apoptosis. Adv Enzyme Regul 48:179–187., DOI 10.1016/j.advenzreg.2008.04.002 Wang G, Ahmad KA, Ahmed K, 2006, Role of Protein kinase CK2 in the regulation of tumor necrosis factor–related apoptosis inducing ligand–induced apoptosis in prostate cancer cells. Cancer Res 66:2242–2249., DOI 10.1158/0008-5472.CAN-05-2772 Hamacher R, Saur D, Fritsch R et al, 2007, Casein kinase II inhibition induces apoptosis in pancreatic cancer cells. Oncol Rep 18:695–701 Wang G, Ahmad KA, Ahmed K, 2005, Modulation of receptor mediated apoptosis by CK2. Mol Cell Biochem 274:201–205 Wang G, Unger G, Ahmad KA et al, 2005, Downregulation of CK2 induces apoptosis in cancer cells-a potential approach to cancer therapy. Mol Cell Biochem 274:77–84 Wang G, Ahmad KA, Harris NH et al, 2008, Impact of protein kinase CK2 on inhibitor of apoptosis proteins in prostate cancer cells. Mol Cell Biochem 316:91–97., DOI 10.1007/s11010-008- 9810-9 Ishak KG, Anthony PP, Sobin LH, 1994, Nonepithelial tumors. In: Ishak KG, ed, World Health Organization International Classification of Tumors. Springer, Berlin, pp 22–27 Wittekind C, 2006, Pitfalls in the classification of liver tumors. Pathology 27:289–293., DOI 10.1007/s00292-006-0834-1 Liu YF, Zhao R, Guo S et al, 2011, Expression and clinical significance of hepatoma-derived growth factor as a prognostic factor in human hilar cholangiocarcinoma. Ann Surg Oncol 18:872–879., DOI 10.1245/s10434-010-1303-x Lin KY, Fang CL, Chen Y et al, 2010, Overexpression of nuclear protein kinase CK2β subunit and prognosis in human gastric carcinoma. Ann Surg Oncol 17:1695–1702., DOI 10.1245/s10434-010- 0911-9 Laramas M, Pasquier D, Filhol O et al, 2007, Nuclear localization of protein kinase CK2 catalytic subunit, CK2α, is associated with poor prognostic factors in human prostate cancer. Eur J Cancer 43:928–934., DOI 10.1016/j.ejca.2006.11.021 Zhao M, Ma J, Zhu HYet al, 2011, Apigenin inhibits proliferation and induces apoptosis in human multiple myeloma cells through targeting the trinity of CK2, Cdc37 and Hsp90. Mol Cancer 10:104., DOI 10.1186/1476-4598-10-104 Huang XY, Ke AW, Shi GM et al, 2010, Overexpression of CD151 as an adverse marker for intrahepatic cholangiocarcinoma patients. Cancer 16:5440–5451., DOI 10.1002/cncr. 25485 Boonjaraspinyo S, Boonmars T, Kaewkes S et al, 2012, Downregulated expression of HSP70 in correlation with clinicopathology of cholangiocarcinoma. Pathol Oncol Res 18:227–237., DOI 10. 1007/s12253-011-9432-5 Yuan RH, Jeng YM, Hu RH et al, 2011, Role of p53 and β-catenin mutations in conjunction with CK19 expression on early tumor recurrence and prognosis of hepatocellular carcinoma. J Gastrointest Surg 15:321–329., DOI 10.1007/s11605-010-1373-x NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 73 EP 79 DI 10.1007/s12253-013-9660-y PG 7 ER PT J AU Yongxiang, W Liang, G Qinshu, Sh AF Yongxiang, Wang Liang, Gao Qinshu, Shao TI Apoptosis of Human Pancreatic Carcinoma PC-2 Cells by an Antisense Oligonucleotide Specific to Point Mutated K-ras SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic carcinoma; K-ras gene; Antisense oligonucleotide; Target gene ID Pancreatic carcinoma; K-ras gene; Antisense oligonucleotide; Target gene AB The prognosis of pancreatic carcinoma is poor due to the difficulty in early diagnosis, insensitivity to routine therapies and limited understanding of its pathological mechanisms. Gene therapy is now becoming an important strategy for the treatment of pancreatic carcinoma, which includes antisense gene therapy. In this study, we investigated the effect of an antisense oligonucleotide specific to point mutated K-ras on the apoptosis of human pancreatic carcinoma cells in vitro. Human pancreatic carcinoma PC-2 cells were transfected with an antisense oligonucleotide specific to a K-ras point mutation by liposomes. The effect of the antisense oligonucleotide on the apoptosis of PC-2 cells was studied using flow cytometry, TUNEL, and phase contrast microscopy. An apoptotic peak was observed in the experimental group, and most cells were arrested at the G1 phase with few cells at the S phase. The numbers of apoptotic cells in the experimental group increased as indicated by TUNEL and phase contrast microscopy. An antisense oligonucleotide specific to a K-ras point mutation promotes apoptosis in PC-2 cells in vitro perhaps by inhibition of ras gene expression. C1 [Yongxiang, Wang] Zhejiang Provincial People’s Hospital, Department of General Surgery, 310014 Hangzhou, China. [Liang, Gao] Zhejiang Provincial People’s Hospital, Department of Oncology, 310014 Hangzhou, China. [Qinshu, Shao] Zhejiang Provincial People’s Hospital, Department of General Surgery, 310014 Hangzhou, China. RP Liang, G (reprint author), Zhejiang Provincial People’s Hospital, Department of Oncology, 310014 Hangzhou, China. EM yxwang910@yahoo.com.cn CR Zongzheng J, Yongxiang W, Xi C, Tao W, 2003, Peripancreatic artery ligation and artery infusion chemotherapy for advanced pancreatic carcinoma. Chin Med J 116:89–92 Luttges J, Hahn S, Kloppel G, 2004, Where and when does pancreatic carcinoma start? Med Klin, Munich, 99:191–195 Shuanzeng W, Tonghua L, Hongrui L, Jie G, 2003, Unique GGT→GTT mutation at K-ras codon 12 in six human pancreatic cancer cell lines from Chinese patients. Chin Med J 116:1585–1587 Luttges J, Stigge C, Pacena M, Kloppel G, 2004, Rare ductal adenocarcinoma of the pancreas in patients younger than age 40 years. Cancer 100:173–182 Wei S, Liang Z, Gao J,Wu S, Liu H, Liu T, 2005, Patterns of K-ras codon 12 and 13 mutations found in pancreatic adenocarcinoma of 30 Chinese patients by microdissection, PCR and direct sequencing. J Gastroenterol Hepatol 20:67–72 Matsubayashi H, Watanabe H, Ajioka Y, Nishikura K, Yamano M, Seki T et al, 2000, Different amounts of K-ras mutant epithelial cells in pancreatic carcinoma and mass-forming pancreatitis. Pancreas 21:77–85 Kita K, Saito S, Morioka CY,Watanabe A, 1999, Growth inhibition on human pancreatic cancer cell lines by anti-sense oligonucleotides specific to mutated K-ras genes. Int J Cancer 80:553–558 Higgins KA, Peres JR, Coleman TA, Dorshkind K, McComasWA, Sarmiento UM et al, 1993, Anti-sense inhibition of the p65 submit of NF-k B block tumorigenecity and causes tumor regression. Proc Nat Acad Sci 90:9901–9905 Tonghua L, Zhiyong W, Lei Z, 1997, The inhibitary effect of modified Antisense oligonucleotide on the growth of pancreatic carcinoma cell and the expression of target gene. Chin J Patho 26:147– 151 Hoffmann B, Lieberman DA, 1994, Molecular regulation of apoptosis: differentiation/growth arrest primary response genes, protooncogenes, and tumor suppressor genes as positive and negative modulation. Oncogene 9:1807–1812 Jaattela M, 1999, Escaping cell death: survival proteins in cancer. Exp Cell Res 248:30–43 Arya J, Finlayson CA, Shames BD et al, 2000, Stimulated apoptosis as an antineoplastic strategy. Surgery 127:366–369 Humenil D, Neel H, Lacout C et al, 1994, Infection with a Kirstenretrovirus can induce a multiplicity of tumorigenic phenotypes in the interleukin-3-dependent FDC-P1 cells. Exp Hematol 22:178– 185 Kinoshita T, Yokota T, Aria KI et al, 1995, Regulation of bcl-2 expression by oncogene ras protein in hematopoietic cells. Oncogene 10:2207–2212 Arends MJ, McGregor AH, Wyllie AH, 1994, Apoptosis is inversely related to necrosis and determines net growth in tumors bearing constitutively expressed myc, ras, and HPVoncogenes. Am J Pathol 144:1045–1052 Yong-xiang W, Liang G, Zong-zheng J, 2007, Inhibitary effects of antisense oligonucleotide specific to K-ras point mutation on the target gene expression in human pancreatic carcinoma cells. Chin Med J 120:1448–1450 Ward RL, Todd AV, Santiago F et al, 1997, Activation of the K-ras in colorectal neoplasms is associated with decreased apoptosis. Cancer 70:1106–1113 Peli J, Schroter M, Rudaz C et al, 1999, Oncogenic Ras inhibits Fas ligand-mediated apoptosis by downregulating the expression of Fas. EMBO J 18:1824–1831 Potten CS, Li YQ, O’Connor PL et al, 1992, A possible explanation for the differential cancer incidence in the intestine, based on distribution of the cytotoxic effects of carcinogens in the murine large bowel. Carcinogenesis 3:2305–2312 Mizukami Y, Ura H, Obara T et al, 2001, Requirement of cjun N-terminal kinase for apoptotic cell death induced by farnesyltransferase inhibitor, farnesylamine, in human pancreatic cancer cells. Biochem Biophys Res Commun 288(1):198– 204 Fenton RG, Hixon JA, Wright PW et al, 1998, Inhibition of Fas, CD95, expression and fas-mediated apoptosis by oncogenic ras. Cancer Res 58:3391–3400 Fushida S, Nishimura GI, Shimizu K et al, 2001, Human pancreatic cancer cells disable function of fas receptors at several levels in fas signal transduction pathway. Int J Oncol 18:311– 316 Gulbins E, Bissonnette R, Mahboubi A et al, 1995, Fas induced apoptosis is mediated via a cermide-initiated RAS signaling pathway. Immunity 2:341–351 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 81 EP 85 DI 10.1007/s12253-013-9661-x PG 5 ER PT J AU Igci, ZY Erkilic, S Igci, M Arslan, A AF Igci, Ziya Yusuf Erkilic, Suna Igci, Mehri Arslan, Ahmet TI MCM3 Protein Expression in Follicular and Classical Variants of Papillary Thyroid Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary thyroid carcinoma; Follicular variant of papillary thyroid carcinoma; Classic variant of papillary thyroid carcinoma; MCM; MCM3 ID Papillary thyroid carcinoma; Follicular variant of papillary thyroid carcinoma; Classic variant of papillary thyroid carcinoma; MCM; MCM3 AB Minichromosome maintenance (MCM) proteins are needed as licensors in the DNA replication of eukaryotic cells and transcriptional control of MCM genes has critical role in the regulation of MCM functions. Different MCM protein family members are proposed as diagnostic or prognostic markers in various cancers due to their increased proliferative potential. Among MCM family members, minichromosome maintenance protein 3 (MCM3) expressions in both mRNA and protein levels were shown to be associated with papillary thyroid carcinoma (PTC). But, the usability of MCM3 in some histological variants of PTC might be controversial due to tissue specific molecular heterogeneities. In follicular variant of papillary thyroid carcinoma (FVPTC), a number of genes including MCM3 were shown to be differentially expressed which were specific to this kind of variant. Using immunohistochemistry method, MCM3 protein expression levels were compared in FVPTC, classic variant of papillary thyroid carcinoma (CVPTC), and multi-nodular goiter (MNG) tissues in a group of 32 cases. There was meaningful differences between MNG vs. FVPTC (p=0.016) and MNG vs. CVPTC (p=0.019) while there was no significant difference in the comparison FVPTC vs. CVPTC (p=0.15). Four of the 5 CVPTC cases having surrounding tissue invasion had high expression values. For FVPTC and CVPTC, MCM3 protein expression results were parallel to our previous mRNA expression study while there was downregulation in protein expression despite the increased expression of MCM3 mRNA in MNG suggesting tissue-specific post-transcriptional events in benign thyroid neoplasms of which should be focused on. Moreover, the relatively lower MCM3 protein expression in FVPTC comparing to CVPTC could be due to a different tumorigenic pathway favored in this type of tissue. C1 [Igci, Ziya Yusuf] University of Gaziantep, Faculty of Medicine, Department of Medical Biology, 27310 Gaziantep, Turkey. [Erkilic, Suna] Gaziantep University, Faculty of Medicine, Department of PathologyGaziantep, Turkey. [Igci, Mehri] University of Gaziantep, Faculty of Medicine, Department of Medical Biology, 27310 Gaziantep, Turkey. [Arslan, Ahmet] University of Gaziantep, Faculty of Medicine, Department of Medical Biology, 27310 Gaziantep, Turkey. RP Igci, ZY (reprint author), University of Gaziantep, Faculty of Medicine, Department of Medical Biology, 27310 Gaziantep, Turkey. EM igci@gantep.edu.tr CR Jemal A, Siegel R, Xu J,Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60:277–300 Fagin JA, Mitsiades N, 2008, Molecular pathology of thyroid cancer: diagnostic and clinical implications. Best Pract Res Clin Endocrinol Metab 22:955–969 Erickson L, Lloyd R, 2010, Well-differentiated papillary thyroid carcinoma. In: Hunt J, ed, Molecular pathology of endocrine diseases. Springer, New York, pp 57–71 Ertek S, Yilmaz NC, Cicero AF, Vurupalmaz O, Demiroz AS, Erdogan G, 2012, Increasing diagnosis of thyroid papillary carcinoma follicular variant in south-east Anatolian region: comparison of characteristics of classical papillary and follicular variant thyroid cancers. Endocr Pathol 23:157–160 Liu J, Singh B, Tallini G, Carlson DL, Katabi N, Shaha A, Tuttle RM, Ghossein RA, 2006, Follicular variant of papillary thyroid carcinoma: a clinicopathologic study of a problematic entity. Cancer 107:1255–1264 LiVolsi VA, 2011, Papillary thyroid carcinoma: an update. Mod Pathol 24(Suppl 2):S1–S9 Salajegheh A, Petcu EB, Smith RA, Lam AK, 2008, Follicular variant of papillary thyroid carcinoma: a diagnostic challenge for clinicians and pathologists. Postgrad Med J 84:78–82 Igci YZ, Arslan A, Akarsu E, Erkilic S, Igci M, Oztuzcu S, Cengiz B, Gogebakan B, Cakmak EA, Demiryurek AT, 2011, Differential expression of a set of genes in follicular and classic variants of papillary thyroid carcinoma. Endocr Pathol 22:86–96 Tye BK, 1999, MCM proteins in DNA replication. Annu Rev Biochem 68:649–686 Maine GT, Sinha P, Tye BK, 1984, Mutants of S. cerevisiae defective in the maintenance of minichromosomes. Genetics 106:365–385 Ha SA, Shin SM, Namkoong H, Lee H, Cho GW, Hur SY, Kim TE, Kim JW, 2004, Cancer-associated expression of minichromosome maintenance 3 gene in several human cancers and its involvement in tumorigenesis. Clin Cancer Res 10:8386–8395 Giaginis C, Vgenopoulou S, Vielh P, Theocharis S, 2010, MCM proteins as diagnostic and prognostic tumor markers in the clinical setting. Histol Histopathol 25:351–370 Lee YS, Ha SA, Kim HJ, Shin SM, Kim HK, Kim S, Kang CS, Lee KY, Hong OK, Lee SH, Kwon HS, Cha BY, Kim JW, 2010, Minichromosome maintenance protein 3 is a candidate proliferation marker in papillary thyroid carcinoma. Exp Mol Pathol 88:138–142 Lei M, 2005, The MCM complex: its role in DNA replication and implications for cancer therapy. Curr Cancer Drug Targets 5:365–380 Melck A, Masoudi H, Griffith OL, Rajput A, Wilkins G, Bugis S, Jones SJ,Wiseman SM, 2007, Cell cycle regulators show diagnostic and prognostic utility for differentiated thyroid cancer. Ann Surg Oncol 14:3403–3411 Musahl C, Holthoff HP, Lesch R, Knippers R, 1998, Stability of the replicative Mcm3 protein in proliferating and differentiating human cells. Exp Cell Res 241:260–264 Endl E, Kausch I, Baack M, Knippers R, Gerdes J, Scholzen T, 2001, The expression of Ki-67, MCM3, and p27 defines distinct subsets of proliferating, resting, and differentiated cells. J Pathol 195:457–462 Zhang P, Zuo H, Ozaki T, Nakagomi N, Kakudo K, 2006, Cancer stem cell hypothesis in thyroid cancer. Pathol Int 56:485–489 Shibru D, Chung KW, Kebebew E, 2008, Recent developments in the clinical application of thyroid cancer biomarkers. Curr Opin Oncol 20:13–18 Salabe GB, 2001, Pathogenesis of thyroid nodules: histological classification? Biomed Pharmacother 55:39–53 Madine MA, Swietlik M, Pelizon C, Romanowski P, Mills AD, Laskey RA, 2000, The roles of the MCM, ORC, and Cdc6 proteins in determining the replication. J Struct Biol 129:198–210 Zhu M,Wang F, Yan F, Yao PY, Du J, Gao X,Wang X,Wu Q,Ward T, Li J, Kioko S, Hu R, Xie W, Ding X, Yao X, 2008, Septin 7 interacts with centromere-associated protein E and is required for its kinetochore localization. J Biol Chem 283:18916–18925 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 87 EP 91 DI 10.1007/s12253-013-9662-9 PG 5 ER PT J AU Chang, L Guo, F Wang, Y Lv, Y Huo, B Wang, L Liu, W AF Chang, Liang Guo, Fengjie Wang, Yudong Lv, Yalei Huo, Bingjie Wang, Long Liu, Wei TI MicroRNA-200c Regulates the Sensitivity of Chemotherapy of Gastric Cancer SGC7901/DDP Cells by Directly Targeting RhoE SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MicroRNA-200c; Gastric cancer; RhoE; Drug resistance ID MicroRNA-200c; Gastric cancer; RhoE; Drug resistance AB Gastric cancer remains a worldwide burden as the second leading cause of cancer-related death. Drug resistance of chemotherapy looms as a major clinical obstacle to successful treatment. Recent evidence indicated that miRNA-200c can restore the sensitivity of NSCLC cells to cisplatin and cetuximab. The expression of miRNA-200c and RhoE were investigated in gastric cancer tissues and cells (SGC7901 and SGC7901/DDP) by qRT-PCR. A luciferase reporter assay was done to understand the potential correlation between miRNA-200c and RhoE. Pre-miR-200c was transfected in SGC7901/DDP cells to confirm whether miRNA-200c could regulate RhoE expression. RhoE was knocked down to explore the role of RhoE on sensitivity of chemotherapy in gastric cancer by MTT. Western blot analysis was performed to further explore the mechanism of RhoE in regulating drug resistance. The results showed that miRNA-200c was significantly lower in cancerous tissues than those in the paired normal tissues, whereas the expression of RhoE was just the opposite. The significant difference of miRNA-200c and RhoE were observed between SGC7901 cells and SGC7901/DDP cells. miRNA-200c has target sites in the 3’-UTR of RhoE mRNA by luciferase reporter assay. Transfection of pre-miR-200c reduces RhoE expression. Meanwhile, the knockdown of RhoE enhanced the sensitivity of SGC7901/DDP cells and changed expression of some genes. These suggested that miRNA-200c regulated the sensitivity of chemotherapy to cisplatin (DDP) in gastric cancer by possibly targeting RhoE. C1 [Chang, Liang] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 12 Jiankang RoadShijiazhuang, Hebei Province, China. [Guo, Fengjie] Tianjin Medical University General Hospital, Tianjin Lung Cancer InstituteTianjin, China. [Wang, Yudong] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 12 Jiankang RoadShijiazhuang, Hebei Province, China. [Lv, Yalei] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 12 Jiankang RoadShijiazhuang, Hebei Province, China. [Huo, Bingjie] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 12 Jiankang RoadShijiazhuang, Hebei Province, China. [Wang, Long] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 12 Jiankang RoadShijiazhuang, Hebei Province, China. [Liu, Wei] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 12 Jiankang RoadShijiazhuang, Hebei Province, China. RP Liu, W (reprint author), Fourth Hospital of Hebei Medical University, Department of Medical Oncology, Shijiazhuang, China. EM changliang1081@163.com CR Lee JH, Kim KM, Cheong JH, Noh SH, 2012, Current management and future strategies of gastric cancer. Yonsei Med J 53(2):248–257 Hudler P, 2012, Genetic aspects of gastric cancer instability. Sci World J 2012:761909 Zhang H, Sun LL, Meng YL, Song GY, Hu JJ, Lu P, Ji B, 2011, Survival trends in gastric cancer patients of Northeast China.World J Gastroenterol 17(27):3257–3262 Maconi G, Manes G, Porro GB, 2008, Role of symptoms in diagnosis and outcome of gastric cancer. World J Gastroenterol 14(8):1149–1155 Sun F, Lu X, Li H, Peng Z,Wu K,Wang G, Tong Q, 2012, Special AT-rich sequence binding protein 1 regulates themultidrug resistance and invasion of human gastric cancer cells. Oncol Lett 4(1):156–162 Schoof CR, Botelho EL, Izzotti A, Vasques Ldos R, 2012, MicroRNAs in cancer treatment and prognosis. Am J Cancer Res 2(4):414–433 Dontu G, de Rinaldis E, 2010, MicroRNAs: shortcuts in dealing with molecular complexity? Breast Cancer Res 12(1):301 Davalos V, Moutinho C, Villanueva A, Boque R, Silva P, Carneiro F, Esteller M, 2012, Dynamic epigenetic regulation of the microRNA-200 family mediates epithelial and mesenchymal transitions in human tumorigenesis. Oncogene 31(16):2062–2074 Roybal JD, Zang Y, Ahn YH, Yang Y, Gibbons DL, Baird BN, Alvarez C, Thilaganathan N, Liu DD, Saintigny P, Heymach JV, Creighton CJ, Kurie JM, 2011, miR-200 inhibits lung adenocarcinoma cell invasion and metastasis by targeting Flt1/VEGFR1. Mol Cancer Res 9(1):25–35 Yu J, Ohuchida K, Mizumoto K, Sato N, Kayashima T, Fujita H, Nakata K, Tanaka M, 2010, MicroRNA, hsa-miR-200c, is an independent prognostic factor in pancreatic cancer and its upregulation inhibits pancreatic cancer invasion but increases cell proliferation. Mol Cancer 9:169 Ceppi P, Mudduluru G, Kumarswamy R, Rapa I, Scagliotti GV, Papotti M, Allgayer H, 2010, Loss of miR-200c expression induces an aggressive, invasive, and chemoresistant phenotype in non-small cell lung cancer. Mol Cancer Res 8(9):1207–1216 Chen J, Tian W, Cai H, He H, Deng Y, 2012, Down-regulation of microRNA-200c is associated with drug resistance in human breast cancer. Med Oncol 29(4):2527–2534 Madigan JP, Bodemann BO, Brady DC, Dewar BJ, Keller PJ, Leitges M, Philips MR, Ridley AJ, Der CJ, Cox AD, 2009, Regulation of Rnd3 localization and function by protein kinase C alpha-mediated phosphorylation. Biochem J 424(1):153–161 Riento K, Villalonga P, Garg R, Ridley A, 2005, Function and regulation of RhoE. Biochem Soc Trans 33(Pt 4):649–651 Villalonga P, Guasch RM, Riento K, Ridley AJ, 2004, RhoE inhibits cell cycle progression and Ras-induced transformation. Mol Cell Biol 24(18):7829–7840 Li K, Lu Y, Liang J, Luo G, Ren G, Wang X, Fan D, 2009, RhoE enhances multidrug resistance of gastric cancer cells by suppressing Bax. Biochem Biophys Res Commun 379(2):212–216 Issabekova A, Berillo O, Regnier M, Anatoly I, 2012, Interactions of intergenic microRNAs with mRNAs of genes involved in carcinogenesis. Bioinformation 8(11):513–518 Hong L, Han Y, Lu Q, Zhang H, Zhao Q,Wu K, Fan D, 2012, Drug resistance-related microRNAs in esophageal cancer. Expert Opin Biol Ther 12(11):1487–1494 Burk U, Schubert J,Wellner U, Schmalhofer O, Vincan E, Spaderna S, Brabletz T, 2008, A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep 9(6):582–589 Bracken CP, Gregory PA, Kolesnikoff N, Bert AG, Wang J, Shannon MF, Goodall GJ, 2008, A double-negative feedback loop between ZEB1-SIP1 and the microRNA-200 family regulates epithelial-mesenchymal transition. Cancer Res 68(19):7846–7854 Jurmeister S, Baumann M, Balwierz A, Keklikoglou I, Ward A, Uhlmann S, Zhang JD, Wiemann S, Sahin O, 2012, MicroRNA- 200c represses migration and invasion of breast cancer cells by targeting actin-regulatory proteins FHOD1 and PPM1F. Mol Cell Biol 32(3):633–651 Lo WL, Yu CC, Chiou GY, Chen YW, Huang PI, Chien CS, Tseng LM, Chu PY, Lu KH, Chang KW, Kao SY, Chiou SH, 2011, MicroRNA-200c attenuates tumour growth and metastasis of presumptive head and neck squamous cell carcinoma stem cells. J Pathol 223(4):482–495 Cochrane DR, Spoelstra NS, Howe EN, Nordeen SK, Richer JK, 2009, MicroRNA-200c mitigates invasiveness and restores sensitivity to microtubule-targeting chemotherapeutic agents. Mol Cancer Ther 8(5):1055–1066 Leskela S, Leandro-Garcia LJ, Mendiola M, Barriuso J, Inglada- Perez L, Munoz I, Martinez-Delgado B, Redondo A, de Santiago J, Robledo M, Hardisson D, Rodriguez-Antona C, 2010, The miR- 200 family controls beta-tubulin III expression and is associated with paclitaxel-based treatment response and progression-free survival in ovarian cancer patients. Endocr Relat Cancer 18(1):85–95 Collett GP, Goh XF, Linton EA, Redman CW, Sargent IL, 2012, RhoE is regulated by cyclic AMP and promotes fusion of human BeWo choriocarcinoma cells. PLoS One 7(1):e30453 Poch E, Minambres R, Mocholi E, Ivorra C, Perez-Arago A, Guerri C, Perez-Roger I, Guasch RM, 2007, RhoE interferes with Rb inactivation and regulates the proliferation and survival of the U87 human glioblastoma cell line. Exp Cell Res 313(4):719–731 Cuiyan Z, Jie H, Fang Z, Kezhi Z, Junting W, Susheng S, Xiaoli F, Ning L, Xinhua M, Zhaoli C, Kang S, Bin Q, Baozhong L, Sheng C, Meihua X, Jie H, 2007, Overexpression of RhoE in Non-small Cell Lung Cancer, NSCLC, is associated with smoking and correlates with DNA copy number changes. Cancer Biol Ther 6(3):335–342 Klein RM, Higgins PJ, 2011, A switch in RND3-RHOA signaling is critical for melanoma cell invasion following mutant-BRAF inhibition. Mol Cancer 10:114 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 93 EP 98 DI 10.1007/s12253-013-9664-7 PG 6 ER PT J AU Nelhubel, AGy Karoly, B Szabo, B Lotz, G Kiss, A Tovari, J Kenessey, I AF Nelhubel, A Gyorgyi Karoly, Boroka Szabo, Balazs Lotz, Gabor Kiss, Andras Tovari, Jozsef Kenessey, Istvan TI The Prognostic Role of Claudins in Head and Neck Squamous Cell Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Claudin; Head and neck squamous cell carcinoma; Prognosis; Immunohistochemistry; Tight junction ID Claudin; Head and neck squamous cell carcinoma; Prognosis; Immunohistochemistry; Tight junction AB The expression of tight junction proteins is frequently altered in epithelial cancers. The loss of cell-cell adhesion associates with enhanced metastatic potential, which underlies the role of altered expression pattern of tight junction component claudins (CLDNs). Our study assessed the expression of CLDN 1, 2, 3, 4, 7, 8 and 10 in squamous cell carcinoma of the head and neck region (HNSCC) including oropahrynx, larynx, and hypopharynx in comparison to normal epithelial tissue of the same patient. The surgical samples were examined by tissue microarray and immunohistochemistry, the expression was calculated by H-score, which took account of intensity and percentage of positivity as well. Both normal and cancerous tissue proved negative for CLDN 3, 8 and 10. Normal epithelia showed mild expression of CLDN 4, but the minimal positivity disappeared in squamous cancer. In case of CLDN 1 and CLDN 7 we demonstrated significantly increased intensity in cancer, while CLDN 2 showed decreased expression compared with normal epithelium. The normal polarity and distribution of claudins were lost in HNSCC. Moreover, preserved expression of CLDN 2 (but not that of 1 and 7) was associated with better survival, which suggested a potential prognostic role of CLDN 2. C1 [Nelhubel, A Gyorgyi] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Karoly, Boroka] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Szabo, Balazs] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Tovari, Jozsef] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Kenessey, Istvan] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. RP Kenessey, I (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM steveken12@yahoo.com CR Parkin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Sant M, Aareleid T, Berrino F et al, 2003, EUROCARE-3: Survival of cancer patients diagnosed 1990–94–results and commentary. Ann Oncol 14(Suppl 5):v61–118 Dobrossy L, 2005, Epidemiology of head and neck cancer: Magnitude of the problem. Cancer Metastasis Rev 24:9–17 Guntinas-Lichius O, Wendt T, Buentzel J et al, 2010, Head and neck cancer in Germany: A site-specific analysis of survival of the thuringian cancer registration database. J Cancer Res Clin Oncol 136:55–63 Zimmermann M, Zouhair A, Azria D et al, 2006, The epidermal growth factor receptor, EGFR, in head and neck cancer: Its role and treatment implications. Radiat Oncol 1:11 Goerner M, Seiwert TY, Sudhoff H, 2010, Molecular targeted therapies in head and neck cancer–an update of recent developments. Head Neck Oncol 2:8 Pereira CS, Oliveira MV, Fraga CA et al, 2012, Impact of the epithelial dysplasia grading and Ki67 proliferation index in the adjacent non-malignant mucosa on recurrence and survival in head and neck squamous cell carcinoma. Pathol Res Pract 208:651–656 Cardesa A, Nadal A, 2011, Carcinoma of the head and neck in the HPV era. Acta Dermatovenerol Alp Panonica Adriat 20:161–173 Smilek P, Dusek L, Vesely K et al, 2005, Prognostic significance of mitotic and apoptotic index and the DNA cytometry in head and neck cancer. Neoplasma 52:199–207 Sadick M, Schoenberg SO, Hoermann K et al, 2012, Current oncologic concepts and emerging techniques for imaging of head and neck squamous cell cancer. GMS Curr Top Otorhinolaryngol Head Neck Surg 11:Doc08 Morin PJ, 2005, Claudin proteins in human cancer: Promising new targets for diagnosis and therapy. Cancer Res 65:9603–9606 Tsukita S, Furuse M, Itoh M, 2001, Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol 2:285–293 Prat A, Parker JS, Karginova O et al, 2010, Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res 12:R68 Szasz AM, Nemeth Z, Gyorffy B et al, 2011, Identification of a claudin-4 and E-cadherin score to predict prognosis in breast cancer. Cancer Sci 102:2248–2254 Rangel LB, Agarwal R, D'Souza Tet al, 2003, Tight junction proteins claudin-3 and claudin-4 are frequently overexpressed in ovarian cancer but not in ovarian cystadenomas. Clin Cancer Res 9:2567–2575 Michl P, Barth C, Buchholz M et al, 2003, Claudin-4 expression decreases invasiveness and metastatic potential of pancreatic cancer. Cancer Res 63:6265–6271 Nichols LS, Ashfaq R, Iacobuzio-Donahue CA, 2004, Claudin 4 protein expression in primary and metastatic pancreatic cancer: Support for use as a therapeutic target. Am J Clin Pathol 121:226–230 Long H, Crean CD, Lee WH et al, 2001, Expression of clostridium perfringens enterotoxin receptors claudin-3 and claudin-4 in prostate cancer epithelium. Cancer Res 61:7878–7881 Borka K, Kaliszky P, Szabo E et al, 2007, Claudin expression in pancreatic endocrine tumors as compared with ductal adenocarcinomas. Virchows Arch 450:549–557 Landers KA, Samaratunga H, Teng L et al, 2008, Identification of claudin-4 as a marker highly overexpressed in both primary and metastatic prostate cancer. Br J Cancer 99:491–501 Szasz AM, Nyirady P, Majoros A et al, 2010, beta-catenin expression and claudin expression pattern as prognostic factors of prostatic cancer progression. BJU Int 105:716–722 Torzsok P, Riesz P, Kenessey I et al, 2011, Claudins and ki-67: Potential markers to differentiate low- and high-grade transitional cell carcinomas of the urinary bladder. J Histochem Cytochem 59:1022–1030 Sobel G, Paska C, Szabo I et al, 2005, Increased expression of claudins in cervical squamous intraepithelial neoplasia and invasive carcinoma. Hum Pathol 36:162–169 Suzuki M, Kato-Nakano M, Kawamoto S et al, 2009, Therapeutic antitumor efficacy of monoclonal antibody against Claudin-4 for pancreatic and ovarian cancers. Cancer Sci 100:1623–1630 Neesse A, Griesmann H, Gress TM et al, 2012, Claudin-4 as therapeutic target in cancer. Arch Biochem Biophys 524:64–70 Gyorffy H, Holczbauer A, Nagy P et al, 2005, Claudin expression in Barrett's esophagus and adenocarcinoma. Virchows Arch 447:961–968 Al Moustafa AE, Alaoui-Jamali MA, Batist G et al, 2002, Identification of genes associated with head and neck carcinogenesis by cDNA microarray comparison between matched primary normal epithelial and squamous carcinoma cells. Oncogene 21:2634–2640 Ouban A, Hamdan H, Hakam A et al, 2012, Claudin-1 expression in squamous cell carcinomas of different organs: Comparative study of cancerous tissues and normal controls. Int J Surg Pathol 20:132–138 Szabo B, Nelhubel GA, Karpati A et al, 2011, Clinical significance of genetic alterations and expression of epidermal growth factor receptor, EGFR, in head and neck squamous cell carcinomas. Oral Oncol 47:487–496 Szekely E, Torzsok P, Riesz P et al, 2011, Expression of claudins and their prognostic significance in noninvasive urothelial neoplasms of the human urinary bladder. J Histochem Cytochem 59:932–941 McCarty KS Jr, Szabo E, Flowers JL et al, 1986, Use of a monoclonal anti-estrogen receptor antibody in the immunohistochemical evaluation of human tumors. Cancer Res 46:4244s–4248s Miyamoto K, Kusumi T, Sato F et al, 2008, Decreased expression of claudin-1 is correlated with recurrence status in esophageal squamous cell carcinoma. Biomed Res 29:71–76 Morita K, Tsukita S, Miyachi Y, 2004, Tight junctionassociated proteins, occludin, ZO-1, claudin-1, claudin-4, in squamous cell carcinoma and Bowen's disease. Br J Dermatol 151:328–334 Tokes AM, Kulka J, Paku S et al, 2005, Claudin-1, -3 and −4 proteins and mRNA expression in benign and malignant breast lesions: a research study. Breast Cancer Res 7:R296–305 Kramer F, White K, Kubbies M et al, 2000, Genomic organization of claudin-1 and its assessment in hereditary and sporadic breast cancer. Hum Genet 107:249–256 Resnick MB, Konkin T, Routhier J et al, 2005, Claudin-1 is a strong prognostic indicator in stage II colonic cancer: a tissue microarray study. Mod Pathol 18:511–518 Yoshizawa K, Nozaki S, Kato A et al, 2013, Loss of claudin-7 is a negative prognostic factor for invasion and metastasis in oral squamous cell carcinoma. Oncol Rep 29:445–450 Hewitt KJ, Agarwal R, Morin PJ, 2006, The claudin gene family: Expression in normal and neoplastic tissues. BMC Cancer 6:186 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 99 EP 106 DI 10.1007/s12253-013-9665-6 PG 8 ER PT J AU Krawczyk, P Nicos, M Ramlau, R Powrozek, T Wojas-Krawczyk, K Sura, S Jarosz, B Szumilo, J Warda, E Mazurkiewicz, T Sawicki, M Milanowski, J AF Krawczyk, Pawel Nicos, Marcin Ramlau, Rodryg Powrozek, Tomasz Wojas-Krawczyk, Kamila Sura, Sylwia Jarosz, Bozena Szumilo, Justyna Warda, Edward Mazurkiewicz, Tomasz Sawicki, Marek Milanowski, Janusz TI The Incidence of EGFR-Activating Mutations in Bone Metastases of Lung Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung adenocarcinoma; EGFR gene mutations; Bone metastases; EGFR tyrosine kinase inhibitors ID Lung adenocarcinoma; EGFR gene mutations; Bone metastases; EGFR tyrosine kinase inhibitors AB Poor prognosis of lung adenocarcinoma is associated with early occurrence of distant metastases. This type of non-small-cell lung carcinoma more frequently involves EGFR gene abnormalities, which determine the efficacy of EGFR tyrosine kinase inhibitor therapies (EGFR TKIs). It is probable that genetic abnormalities present in primary tumor will also be present in metastases. Unfortunately little is known about the incidence of these mutations in the metastases and about the effectiveness ofmolecularly targeted therapy in such patients. Formalin-fixed, paraffin-embedded tumor tissue was prepared from 431 samples of primary adenocarcinoma, 61 of adenocarcinoma central nervous system (CNS) metastases and 8 of adenocarcinoma bone metastases. The presence of exon 19 deletions was examined using the PCR technique and amplified PCR product fragment length analysis. The ASP-PCR technique was used to evaluate the L858R substitutions in exon 21, and the results were analyzed using ALF Express II sequencer. In the adenocarcinoma metastases to bone obtained from 8 patients, deletions in exon 19 of the EGFR gene were revealed in 3 smoking men and one nonsmoking woman, while L858R substitution in exon 21 was found in one smoking woman and one man of unknown smoking status. The incidence of EGFR gene mutations in the bonemetastases was 75 %, in the primary adenocarcinoma - 12.8 %, and in the adenocarcinoma metastases to CNS - 14.75 %. Five patients with EGFR gene mutation revealed in bone metastases were treated with EGFR TKIs; the majority of them had a satisfactory response to therapy. C1 [Krawczyk, Pawel] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Nicos, Marcin] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Ramlau, Rodryg] Greater Poland Center of Pulmonology and Thoracic Surgery of Eugenia and Janusz ZeylandPoznan, Poland. [Powrozek, Tomasz] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Wojas-Krawczyk, Kamila] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Sura, Sylwia] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Jarosz, Bozena] Medical University of Lublin, Neurosurgery and Pediatric Neurosurgery DepartmentLublin, Poland. [Szumilo, Justyna] Medical University of Lublin, Department of Clinical PathomorphologyLublin, Poland. [Warda, Edward] Medical University of Lublin, Orthopedics and Traumatology DepartmentLublin, Poland. [Mazurkiewicz, Tomasz] Medical University of Lublin, Orthopedics and Traumatology DepartmentLublin, Poland. [Sawicki, Marek] Medical University of Lublin, Thoracic Surgery DepartmentLublin, Poland. [Milanowski, Janusz] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. RP Krawczyk, P (reprint author), Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, 20-954 Lublin, Poland. EM krapa@poczta.onet.pl CR Jemal A, Bray F, Center MM et al, 2011, Global cancer statistic. CA Cancer J Clin 61:69–90 Kadara H, Kabbout M, Wistuba II, 2011, Pulmonary adenocarcinoma: a renewed entity in 2012. Respirology 17:50–65 Kalikaki A, Koutsopoulos A, Trypaki M et al, 2008, Comparison of EGFR and K-RAS gene status between primary tumours and corresponding metastases in NSCLC. Br J Cancer 99:923–929 Petrelli F, Borgonovo K, Cabiddu Met al, 2012, Efficacy of EGFR tyrosine kinase inhibitors in patients with EGFR-mutated nonsmall- cell lung cancer: a meta-analysis of 13randomized trials. Clin Lung Cancer 13:107–114 Italiano A, Vandenbos FB, Otto J et al, 2006, Comparison of the epidermal growth factor receptor gene and protein in primary non-small-cell-lung cancer and metastatic sites: implications for treatment with EGFR-inhibitors. Ann Oncol 17:981–985 Reungwetwattana T, Weroha SJ, Molina JR, 2012, Oncogenic pathways, molecularly targeted therapies, and highlighted clinical trials in non-small-cell lung cancer, NSCLC). Clin Lung Cancer 13:252–266 Sun L, Zhang Q, Luan H et al, 2011, Comparison of KRAS and EGFR gene status between primary non-small cell lung cancer and local lymph node metastases: implications for clinical practice. J Exp Clin Cancer Res 30:30–38 Sugiura H, Yamada K, Sugiura T et al, 2008, Predictors of survival in patients with bone metastasis of lung cancer. Clin Orthop Relat Res 466:729–736 Suva LJ, Washam C, Nicholas RW et al, 2011, Bone metastasis: mechanisms and therapeutic opportunities. Nat Rev Endocrinol 7:208–218 Theriault RL, Theriault RL, 2012, Biology of bone metastases. Cancer Control 19:92–101 Badalian G, Barbai T, Raso E et al, 2007, Phenotype of bone metastases of non-small cell lung cancer: epidermal growth factor receptor expression and K-RAS mutational status. Pathol Oncol Res 13:99–104 Patel LR, Camacho DF, Shiozawa Y et al, 2011, Mechanisms of cancer cell metastasis to the bone: a multistepprocess. Future Oncol 7:1285–1297 Krawczyk P, Remiszewski W, Czekajska-Chehab E et al, 2009, Evident clinical response to erlotinib as third-line treatment in EGFR FISH, +, male smoker patient with adenocarcinoma of lung. Am J Case Rep 10:166–171 Kunimasa K, Masuda G, Watanabe N et al, 2012, Diffuse metastases of lung adenocarcinoma with EGFR mutation. Intern Med 51:685–686 Togashi Y, Masago K, Kubo T et al, 2011, Association of diffuse, random pulmonary metastases, including miliary metastases, with epidermal growth factor receptor mutations in lung adenocarcinoma. Cancer 117:819–825 Matsumoto S, Takahashi K, Iwakawa R et al, 2006, Frequent EGFR mutations in brain metastases of lung adenocarcinoma. Int J Cancer 119:1491–1494 Furugaki K, Moriya Y, Iwai T, 2011, Erlotinib inhibits osteolytic bone invasion of human non-small-cell lung cancer cell line NCIH292. Clin Exp Metastasis 28:649–659 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 107 EP 112 DI 10.1007/s12253-013-9667-4 PG 6 ER PT J AU Naserpour Farivar, T Johari, P Najafipour, R Farzam, A Nasirian, N HajManouchehri, F Jahani Hashemi, H Azimi, A Bahrami, M AF Naserpour Farivar, Taghi Johari, Pouran Najafipour, Reza Farzam, Amir Nasirian, Neda HajManouchehri, Fatemeh Jahani Hashemi, Hassan Azimi, Akram Bahrami, Mohammad TI The Relationship Between Gastric Cancer and Helicobacter Pylori in Formaldehyde Fixed Paraffin Embedded Gastric Tissues of Gastric Cancer Patients-Scorpion Real-Time PCR Assay Findings SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Scorpion; Real-time; Helicobacter pylori; Gastric; Cancer ID Scorpion; Real-time; Helicobacter pylori; Gastric; Cancer AB Gastric cancer is the second leading cause of cancerrelated deaths worldwide and it seems that environmental and lifestyle factors and infection with Helicobacter pylori (H.pylori) have had a major role in the etiology of gastric cancer. The aim of this study was to investigate the presence of H. pylori DNA in archival gastric tissues of patients with gastric cancer disease by rapid, sensitive and specific technique of Scorpion Realtime PCR. This retrospective cross-sectional study was performed on 285 paraffin embedded gastric specimens of patients who were pathologically proved for gastric cancer admitted in Bou- Ali, Shahid Rajaie and Dehkhoda hospitals and Bahar and Farzam private laboratory in Qazvin city in Iran during 2009 and 150 paraffin embedded pathological specimens of patients with other proved diagnosis other than gastric cancer. Results of our Scorpion Realtime PCR analysis showed that DNA of H. pylori DNA was present in 78.42 % of our total specimens. Modified McMullen’s Staining of paraffin embedded sections was positive in 210 patients. Also we were not able to finding significant relationship between demographic characteristics of our studied patients and presence of H. pylori DNA in their formaldehyde fixed paraffin embedded gastric tissues samples. Existence of H. pylori in gastric tissue samples of patients with gastric cancer is controversial and our results indicated that in our studied specimens prevalence of H.pylori was significantlymore than recent published reports. C1 [Naserpour Farivar, Taghi] Qazvin University of Medical Sciences, Cellular and Molecular Reasearch CenterQazvin, Iran. [Johari, Pouran] Qazvin University of Medical Sciences, Cellular and Molecular Reasearch CenterQazvin, Iran. [Najafipour, Reza] Qazvin University of Medical Sciences, Cellular and Molecular Reasearch CenterQazvin, Iran. [Farzam, Amir] Qazvin University of Medical Sciences, Cellular and Molecular Reasearch CenterQazvin, Iran. [Nasirian, Neda] Qazvin University of Medical Sciences, Cellular and Molecular Reasearch CenterQazvin, Iran. [HajManouchehri, Fatemeh] Qazvin University of Medical Sciences, Cellular and Molecular Reasearch CenterQazvin, Iran. [Jahani Hashemi, Hassan] Qazvin University of Medical Sciences, Cellular and Molecular Reasearch CenterQazvin, Iran. [Azimi, Akram] Qazvin University of Medical Sciences, Cellular and Molecular Reasearch CenterQazvin, Iran. [Bahrami, Mohammad] Qazvin University of Medical Sciences, Cellular and Molecular Reasearch CenterQazvin, Iran. RP Najafipour, R (reprint author), Qazvin University of Medical Sciences, Cellular and Molecular Reasearch Center, Qazvin, Iran. EM rnajafipour@gmail.com CR Ferlay J, Bray F, Pisani P, 2001, GLOBOCAN 2000: Cancer incidence, mortality and prevalence worldwide, version 1.0. IARC Cancer Base No 5. IARC Pres, Lyon, 1 Crew KD, Neuget AI, 2006, Epidemiology of gastric cancer.World J Gastroenterol 12:354–363 Tiwari SK, Manoj G, Vasanth Kumar G, Sivaram G, Hassan SI, 2008, Prognostic significance of genotyping Helicobacter pylori infection in patients in younger age groups with gastric cancer. Postgrad Med J 84:193–197 Ries LAG, Kosary CL, Hankey BF, 1994, International Agency for Research on CancerWorking Group on the Evaluation of Carcinogenic Risks to Humans. Schistosomes, liver flukes, and Helicobacter pylori. International Agency for Research on Cancer, Lyon, pp 177–240 Fox JG, Wang TC, 2007, Inflammation, atrophy, and gastric cancer. J Clin Invest 117:60–69 Naserpour Farivar T, Pahlevan AK, Johari P, 2012, Assessment of Helicobacter pylori prevalence by scorpion Real-time PCR in chronic tonsillitis patients. J Global Infec Dis 4:38–42 Correa P, Piazuelo MB, 2008, Natural history of Helicobacter pylori infection. Dig Liver Dis 40:490–496 McNamara D, El-Omar E, 2008, Helicobacter pylori infection and the pathogenesis of gastric cancer: a paradigm for host-bacterial interactions. Dig Liver Dis 40:504–509 Fock KM, Dhamodaran S, Teo EK, 2006, Is H. pylori genotypes significant in Asian patients with gastric cancer and GERD. Gut 55(Suppl. V):A258 Erzin Y, Koksal V, Altun S, 2006, Prevalence of Helicobacter pylori vacA, cagA, cagE, iceA, babA2 genotypes and correlation with clinical outcome in Turkish patients with dyspepsia. Helicobacter 11:574–580 Chomvarin C, Namwat W, Chaicumpar K, 2008, Prevalence of Helicobacter pylori vacA, cagA, cagE, iceA and babA2 genotypes in Thai dyspeptic patients. Int J Infect Dis 12:30–36 Ghasemi A, Shirazi MH, Ranjbar R, 2008, The prevalence of cagA and cagE genes in Helicobacter pylori strains isolated from different patient groups by polymerase chain reaction. Pak J Biol Sci 11:2579–2583 Babus V, Strnad M, Presecki V, 1998, Helicobacter pylori and gastric cancer in Croatia. Cancer Lett 125:9–15 Uemura N, Okamoto S, Yamamoto S, 2001, Helicobacter pylori infection and the development of gastric cancer. N Engl J Med 345:784–789 Martinez T, Hernandez-Suarez G, Mercedes Bravo M, Trujillo E, Quiroga A, Albis R, 2011, Association of interleukin-1 genetic polymorphism and CagA positive Helicobacter pylori with gastric cancer in Colombia]. Rev Med Chil 139:1313– 1321 Khanna AK, Seth P, Nath G, 2002, Correlation of Helicobacter pylori and gastric carcinoma. J Postgrad Med 48:27–35 Rudi J, Muller M, von Herbay A, 1995, Lack of association of Helicobacter pylori seroprevalence and gastric cancer in a population with low gastric cancer incidence. Scand J Gastroenterol 30:958–963 Burucoa C, Garnier M, Silvain C, Fauchere JL, 2008, Quadruplex real-time pcr assay using allele-specific scorpion primers for detection of mutations conferring clarithromycin resistance to helicobacter pylori. J Clin Microbiol 46:2320–2326 Hashemi MR, Rahnavardi M, Bikdeli B, Dehghani Zahedani M, 2006, H pylori infection among 1000 southern Iranian dyspeptic patients. World J Gastroenterol 12(34):5479–5482 Bauer B, Meyer TF, 2011, The human gastric pathogen helicobacter pylori and its association with gastric cancer and ulcer disease. Ulcers 2011, Article ID: 40157, 23 pages McColl K, 2010, Helicobacter pylori infection. N Engl J Med 362:1597–1604 Fuccio L, Eusebi LH, Bazzoli F, 2010, Gastric cancer, Helicobacter pylori infection and other risk factors. World J Gastrointest Oncol 2:342–347 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 113 EP 117 DI 10.1007/s12253-013-9669-2 PG 5 ER PT J AU Lokamani, I Looi, ML Md Ali, AS Mohd Dali, ZHA Ahmad Annuar, AM Jamal, R AF Lokamani, Ilambarthi Looi, Mee-Lee Md Ali, Aishah Siti Mohd Dali, Zailani Hatta Ahmad Ahmad Annuar, Azrif Muhammad Jamal, Rahman TI Gelsolin and Ceruloplasmin as Potential Predictive Biomarkers for Cervical Cancer by 2D-DIGE Proteomics Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE 2D-DIGE; Cervical neoplasia; Gelsolin; ceruloplasmin; proteomics ID 2D-DIGE; Cervical neoplasia; Gelsolin; ceruloplasmin; proteomics AB This study aimed to identify candidate proteins which may serve as potential biological markers for cervical cancer using 2D-DIGE. Serum samples of controls, patients with cervical intraepithelial neoplasia grade 3 (CIN 3), squamous cell carcinoma of early (SCC I and II) and late (SCC III and IV) stage were subjected to 2D-DIGE. Differentially expressed spots were identified by tandem mass spectrometry. Validation of candidate proteins in serum and tissue samples were then performed by ELISA and immunohistochemistry (IHC) analysis respectively. A total of 20 differentially expressed proteins were identified. These proteins were found to play key roles in the apoptosis pathway, complement system, various types of transportation such as hormones, fatty acids, lipid, vitamin E and drug transportation, coagulation cascade, regulation of iron and immunologic response. Based on their functional relevancy to the progression of various cancers, 4 proteins namely the complement factor H, CD5- like antigen, gelsolin and ceruloplasmin were chosen for further validation using ELISA. Biological network analysis showed that ceruloplasmin and gelsolin are closely interacted with the oncogene NF-κb. These two proteins were further validated using the IHC. Gelsolin and ceruloplasmin may serve as potential predictive biomarkers for the progression of high grade lesions. C1 [Lokamani, Ilambarthi] Universiti Kebangsaan Malaysia, UKM Medical Molecular Biology Institute (UMBI), Level 7, Clinical Block, UKM Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, 56000 Kuala Lumpur, Malaysia. [Looi, Mee-Lee] Universiti Kebangsaan Malaysia, UKM Medical Molecular Biology Institute (UMBI), Level 7, Clinical Block, UKM Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, 56000 Kuala Lumpur, Malaysia. [Md Ali, Aishah Siti] Universiti Kebangsaan Malaysia, Faculty of Medicine, Department of Pathology, 56000 Kuala Lumpur, Malaysia. [Mohd Dali, Zailani Hatta Ahmad] Universiti Kebangsaan Malaysia, Obstetrics and Gynecology Department, 56000 Kuala Lumpur, Malaysia. [Ahmad Annuar, Azrif Muhammad] Universiti Kebangsaan Malaysia, Faculty of Medicine, Radiology Department, 56000 Kuala Lumpur, Malaysia. [Jamal, Rahman] Universiti Kebangsaan Malaysia, UKM Medical Molecular Biology Institute (UMBI), Level 7, Clinical Block, UKM Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, 56000 Kuala Lumpur, Malaysia. RP Jamal, R (reprint author), Universiti Kebangsaan Malaysia, UKM Medical Molecular Biology Institute (UMBI), 56000 Kuala Lumpur, Malaysia. EM rahmanj@ppukm.ukm.edu.my CR Fung ET, 2010, A recipe for proteomics diagnostic test development: the OVA1 test, from biomarker discovery to FDA clearance. Clin Chem 56:327–329 Teramoto R, Minagawa H, Honda M, Miyazaki K, Tabuse Y et al, 2008, Protein expression profile characteristic to hepatocellular carcinoma revealed by 2D-DIGE with supervised learning. Biochimica et Biophysica Acta - Proteins and Proteomics 1784: 764–772 Zhou G, Li H, DeCamp D, Chen S, Shu H et al, 2002, 2D differential in-gel electrophoresis for the identification of esophageal scans cell cancer-specific protein markers. Mol Cell Proteomics : MCP 1:117–124 Katayama M, Nakano H, Ishiuchi A,WuW, Oshima R et al, 2006, Protein pattern difference in the colon cancer cell lines examined by two-dimensional differential in-gel electrophoresis and mass spectrometry. Surg Today 36:1085–1093 Murphy L, Henry M, Meleady P, Clynes M, Keenan J, 2008, Proteomic investigation of taxol and taxotere resistance and invasiveness in a squamous lung carcinoma cell line. Biochimica et Biophysica Acta, BBA, - Proteins & Proteomics 1784:1184–1191 Orenes-Pinero E, CortonM, Gonzalez-Peramato P, Algaba F, Casal I et al, 2007, Searching urinary tumor markers for bladder cancer using a two-dimensional differential gel electrophoresis, 2D-DIGE, approach. J Proteome Res 6:4440–4448 Kakisaka T, Kondo T, Okano T, Fujii K, Honda Ket al, 2007, Plasma proteomics of pancreatic cancer patients by multi-dimensional liquid chromatography and two-dimensional difference gel electrophoresis, 2D-DIGE): up-regulation of leucine-rich alpha-2-glycoprotein in pancreatic cancer. J Chromatogr B 852:257–267 Arnouk H, Merkley MA, Podolsky RH, Stoppler H, Santos C et al, 2009, Characterization of molecular markers indicative of cervical cancer progression. Proteomics Clin Appl 3:516–527 Jacobs JM, Adkins JN, Qian WJ, Liu T, Shen Y et al, 2005, Utilizing human blood plasma for proteomic biomarker discovery. J Proteome Res 4:1073–1085 Richart RM(1995, Screening. The next century. Cancer 76:1919–1927 Ostor AG, 1993, Natural history of cervical intraepithelial neoplasia: a critical review. Int J Gynecol Pathol 12:186–192 Sadler L, Saftlas A, Wang W, Exeter M, Whittaker J et al, 2004, Treatment for cervical intraepithelial neoplasia and risk of preterm delivery. JAMA 291:2100–2106 Huang HL, Stasyk T, Morandell S, Dieplinger H, Falkensammer G et al, 2006, Biomarker discovery in breast cancer serum using 2-D differential gel electrophoresis/MALDI-TOF/TOF and data validation by routine clinical assays. Electrophoresis 27:1641–1650 Watari H, Ohta Y, Hassan MK, Xiong Y, Tanaka S et al, 2008, Clusterin expression predicts survival of invasive cervical cancer patients treated with radical hysterectomy and systematic lymphadenectomy. Gynecol Oncol 108:527–532 Abdul-Rahman PS, Lim BK, Hashim OH, 2007, Expression of high-abundance proteins in sera of patients with endometrial and cervical cancers: analysis using 2-DE with silver staining and lectin detection methods. Electrophoresis 28:1989–1996 Bae SM, Lee CH, Cho YL, Nam KH, Kim YW et al, 2005, Twodimensional gel analysis of protein expression profile in squamous cervical cancer patients. Gynecol Oncol 99:26–35 Lin YW, Lai HC, Lin CY, Chiou JY, Shui HA et al, 2006, Plasma proteomic profiling for detecting and differentiating in situ and invasive carcinomas of the uterine cervix. Int J Gynecol Cancer 16:1216–1224 Lee KH, Yim EK, Kim CJ, Namkoong SE, Um SJ et al, 2005, Proteomic analysis of anti-cancer effects by paclitaxel treatment in cervical cancer cells. Gynecol Oncol 98:45–53 Yim EK, Lee KH, Namkoong SE, Um SJ, Park JS, 2006, Proteomic analysis of ursolic acid-induced apoptosis in cervical carcinoma cells. Cancer Lett 235:209–220 Koya RC, Fujita H, Shimizu S, Ohtsu M, Takimoto M et al, 2000, Gelsolin inhibits apoptosis by blocking mitochondrial membrane potential loss and cytochrome c release. J Biol Chem 275:15343– 15349 Li Y, Qu P, Wu L, Li B, Du H et al., 2011, Api6/AIM/Spα/CD5L Overexpression in alveolar type II epithelial cells induces spontaneous lung adenocarcinoma. Cancer Res 71(16):5488–5499 Sun HQ, Yamamoto M, Mejillano M, Yin HL, 1999, Gelsolin, a multifunctional actin regulatory protein. J Biol Chem 274:33179– 33182 Fujita H, Okada F, Hamada J, Hosokawa M, Moriuchi Tet al, 2001, Gelsolin functions as a metastasis suppressor in B16-BL6 mouse melanoma cells and requirement of the carboxyl-terminus for its effect. Int J Cancer 93:773–780 Bucki R, Levental I, Kulakowska A, Janmey PA, 2008, Plasma gelsolin: function, prognostic value, and potential therapeutic use. Curr Protein Pept Sci 9:541–551 Yin HL, Kwiatkowski DJ, Mole JE, Cole FS, 1984, Structure and biosynthesis of cytoplasmic and secreted variants of gelsolin. J Biol Chem 259:5271–5276 Asch HL, Winston JS, Edge SB, Stomper PC, Asch BB, 1999, Down-regulation of gelsolin expression in human breast ductal carcinoma in situ with and without invasion. Breast Cancer Res Treat 55:179–188 Van den Abbeele A, De Corte V, Van Impe K, Bruyneel E, Boucherie C et al, 2007, Downregulation of gelsolin family proteins counteracts cancer cell invasion in vitro. Cancer Lett 255:57–70 De Corte V, Bruyneel E, Boucherie C, Mareel M, Vandekerckhove J et al, 2002, Gelsolin-induced epithelial cell invasion is dependent on Ras-Rac signaling. EMBO J 21:6781–6790 Sun H, Lin K, Yin HL, 1997, Gelsolin modulates phospholipase C activity in vivo through phospholipid binding. J Cell Biol 138: 811–820 Singh SS, Chauhan A, Murakami N, Chauhan VP, 1996, Profilin and gelsolin stimulate phosphatidylinositol 3-kinase activity. Biochemistry 35:16544–16549 Gangadharan B, Antrobus R, Dwek RA, Zitzmann N, 2007, Novel serum biomarker candidates for liver fibrosis in hepatitis C patients. Clin Chem 53:1792–1799 Sarrias MR, Rosello S, Sanchez-Barbero F, Sierra JM, Vila J et al, 2005, A role for human Sp alpha as a pattern recognition receptor. J Biol Chem 280:35391–35398 Gray J, Chattopadhyay D, Beale GS, Patman GL, Miele L et al, 2009, A proteomic strategy to identify novel serum biomarkers for liver cirrhosis and hepatocellular cancer in individuals with fatty liver disease. BMC Cancer 9:271 Rutkowski MJ, Sughrue ME, Kane AJ, Ahn BJ, Fang S et al, 2010, The complement cascade as a mediator of tissue growth and regeneration. Inflamm Res 59:897–905 Rutkowski MJ, Sughrue ME, Kane AJ, Mills SA, Parsa AT, 2010, Cancer and the complement cascade. Mol Cancer Res 8:1453–1465 Ajona D, Castano Z, Garayoa M, Zudaire E, Pajares MJ et al, 2004, Expression of complement factor H by lung cancer cells: effects on the activation of the alternative pathway of complement. Cancer Res 64:6310–6318 Cheng Z, Corey MJ, Parepalo M, Majno S, Hellwage J et al, 2005, Complement Factor H as a marker for detection of bladder cancer. Clin Chem 51(5):856–863 Junnikkala S, Hakulinen J, Jarva H, Manuelian T, Bjorge L et al, 2002, Secretion of soluble complement inhibitors factor H and factor H-like protein, FHL-1, by ovarian tumour cells. Br J Cancer 87:1119–1127 Kinders R, Jones T, Root R, Bruce C, Murchison H et al, 1998, Complement factor H or a related protein is a marker for transitional cell cancer of the bladder. Clin Cancer Res 4:2511–2520 Bradley DT, Zipfel PF, Hughes AE, 2011, Complement in age-related macular degeneration: a focus on function. Eye, Lond, 25:683–693 Ambrus JL Jr, Peters MG, Fauci AS, Brown EJ, 1990, The Ba fragment of complement factor B inhibits human B lymphocyte proliferation. J Immunol 144:1549–1553 Doustjalali SR, Yusof R, Yip CH, Looi LM, Pillay B et al, 2004, Aberrant expression of acute-phase reactant proteins in sera and breast lesions of patients with malignant and benign breast tumors. Electrophoresis 25:2392–2401 Bloomston M, Zhou JX, Rosemurgy AS, Frankel W, Muro-Cacho CA et al, 2006, Fibrinogen gamma overexpression in pancreatic cancer identified by large-scale proteomic analysis of serum samples. Cancer Res 66:2592–2599 Seriramalu R, Pang WW, Jayapalan JJ, Mohamed E, Abdul-Rahman PS et al, 2010, Application of champedak mannose-binding lectin in the glycoproteomic profiling of serum samples unmasks reduced expression of alpha-2 macroglobulin and complement factor B in patients with nasopharyngeal carcinoma. Electrophoresis 31:2388–2395 Huang X, 2008, Does iron have a role in breast cancer? Lancet Oncol 9:803–807 Liehr JG, Jones JS, 2001, Role of iron in estrogen-induced cancer. Curr Med Chem 8:839–849 Raju KS, Alessandri G, Ziche M, Gullino PM(1982, Ceruloplasmin, copper ions, and angiogenesis. J Natl Cancer Inst 69:1183–1188 Coates RJ, Weiss NS, Daling JR, Rettmer RL, Warnick GR, 1989, Cancer risk in relation to serum copper levels. Cancer Res 49:4353– 4356 Pejovic M, Djordjevic V, Ignjatovic I, Stamenic T, Stefanovic V, 1997, Serum levels of some acute phase proteins in kidney and urinary tract urothelial cancers. Int Urol Nephrol 29:427–432 Doustjalali SR, Yusof R, Govindasamy GK, Bustam AZ, Pillay B et al, 2006, Patients with nasopharyngeal carcinoma demonstrate enhanced serum and tissue ceruloplasmin expression. J Med Invest 53:20–28 Onizuka K, Migita S, Yamada H, Matsumoto I, 1999, Serum protein fractions in patients with laryngeal cancer undergoing radiation therapy. Possibility as a prognostic factor. Fukuoka Igaku Zasshi 90:46–58 Sampson MT, Kakkar AK, 2002, Coagulation proteases and human cancer. Biochem Soc Trans 30:201–207 Dolcet X, Llobet D, Pallares J, Matias X, 2005, NF-kB in development and progression of human cancer. Virchows archiv 446:8 Escarcega RO, Fuentes-Alexandro S, Garcia-Carrasco M, Gatica A, Zamora A, 2007, The transcription factor nuclear factor-kappa B and cancer. Clin Oncol, R Coll Radiol, 19:154–161 Persichini T, Maio N, di Patti MC, Rizzo G, Colasanti M et al, 2010, Interleukin-1beta induces ceruloplasmin and ferroportin-1 gene expression via MAP kinases and C/EBPbeta, AP-1, and NFkappaB activation. Neurosci Lett 484:133–138 EunDW, Ahn SH, You JS, Park JW, Lee EK et al, 2007, PKCepsilon is essential for gelsolin expression by histone deacetylase inhibitor apicidin in human cervix cancer cells. Biochem Biophys Res Commun 354:769–775 Liao CJ, Wu TI, Huang YH, Chang TC, Wang CS et al, 2011, Overexpression of gelsolin in human cervical carcinoma and its clinicopathological significance. Gynecol Oncol 120:10 Ni XG, Zhou L,Wang GQ, Liu SM, Bai XF et al, 2008, The ubiquitinproteasome pathway mediates gelsolin protein downregulation in pancreatic cancer. Mol Med 14:582–589 Li R, Chen WC, Pang XQ, Hua C, Li L et al, 2009, Expression of CD40 and CD40L in gastric cancer tissue and its clinical significance. Int J Mol Sci 10:3900–3917 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 119 EP 129 DI 10.1007/s12253-013-9670-9 PG 11 ER PT J AU Vosmik, M Laco, J Sirak, I Beranek, M Hovorkova, E Vosmikova, H Drastikova, M Hodek, M Zoul, Z Odrazka, K Petera, J AF Vosmik, Milan Laco, Jan Sirak, Igor Beranek, Martin Hovorkova, Eva Vosmikova, Hana Drastikova, Monika Hodek, Miroslav Zoul, Zdenek Odrazka, Karel Petera, Jiri TI Prognostic Significance of Human Papillomavirus (HPV) Status and Expression of Selected Markers (HER2/neu, EGFR, VEGF, CD34, p63, p53 and Ki67/MIB-1) on Outcome After (Chemo-) Radiotherapy in Patients with Squamous Cell Carcinoma of Uterine Cervix SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Uterine cervix; Immunohistochemistry; Squamous cell carcinoma; Prognostic factors; Human papillomavirus ID Uterine cervix; Immunohistochemistry; Squamous cell carcinoma; Prognostic factors; Human papillomavirus AB The aim of the retrospective study was to evaluate prognostic significance of human papillomavirus (HPV) status and expression of epidermal growth factor receptor (EGFR), human epidermal growth factor receptor type 2 (HER2/neu), vascular endothelial growth factor (VEGF), CD34 antigen, tumor suppressors p63 and p53, and Ki67/ MIB-1 in squamous cell carcinoma of the uterine cervix (SCCC) treated with radiotherapy or chemoradiotherapy. Seventy-two consecutive patients with SCCC, diagnosed and treated with (chemo-) radiotherapy with a curative intent at the University Hospital Hradec Kralove between August 1998 and August 2008, were enrolled in the study. The median follow-up period was 57 months (range 5–152). The tested biological factors were evaluated by polymerase chain reaction (HPV status) and by immunohistochemistry (remaining above mentioned markers) from archival paraffin embedded original diagnostic tumor samples. A statistical significant correlation was observed between low expression of p63 and poor overall survival (p =0.001), although the complete response probability was influenced with borderline statistical significance (p=0.05). However, the results could be affected by the statistical error due to the small number of p63 negative patients. HPV positivity and EGFR staining intensity was associated with higher complete response probability (p =0.038 and p =0.044, resp.). All other results were not significant. Neither HPV positivity nor EGFR staining intensity were reflected in the overall survival evaluation. In conclusion, the presented study did not confirm any apparently significant association of the suggested markers with prognosis of SCCC in patients treated with (chemo-) radiotherapy. C1 [Vosmik, Milan] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, Sokolska 581, 500 05 Hradec Kralove, Czech Republic. [Laco, Jan] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec KraloveHradec Kralove, Czech Republic. [Sirak, Igor] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec KraloveHradec Kralove, Czech Republic. [Beranek, Martin] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec KraloveHradec Kralove, Czech Republic. [Hovorkova, Eva] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec KraloveHradec Kralove, Czech Republic. [Vosmikova, Hana] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec KraloveHradec Kralove, Czech Republic. [Drastikova, Monika] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec KraloveHradec Kralove, Czech Republic. [Hodek, Miroslav] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec KraloveHradec Kralove, Czech Republic. [Zoul, Zdenek] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec KraloveHradec Kralove, Czech Republic. [Odrazka, Karel] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec KraloveHradec Kralove, Czech Republic. [Petera, Jiri] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec KraloveHradec Kralove, Czech Republic. RP Vosmik, M (reprint author), Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, 500 05 Hradec Kralove, Czech Republic. EM milan.vosmik@fnhk.cz CR Green JA, Kirwan JM, Tierney JF, Symonds P, Fresco L, Collingwood M, Williams CJ, 2001, Survival and recurrence after concomitant chemotherapy and radiotherapy for cancer of the uterine cervix: a systematic review and meta-analysis. Lancet 358:781– 786 Gondi V, Bentzen SM, Sklenar KL, Dunn EF, Petereit DG, Tannehill SP, Straub M, Bradley KA, 2012, Severe late toxicities following concomitant chemoradiotherapy compared to radiotherapy alone incervical cancer: an inter-era analysis. Int J Radiat Oncol Biol Phys 84:973–982., DOI 10.1016/j.ijrobp.2012.01.064 Petera J, Sirak I, Beranek M, Vosmik M, Drastikova M, Paulikova S, Soumarova R, 2011, Molecular predictive factors of outcome of radiotherapy in cervical cancer. Neoplasma 58:469–475 Sirak I, Petera J, Odrazka K, Dolezel M, Zoul Z, Vaculikova M, 2006, Chemotherapy intensity importance in concurrent chemoradiotherapy of locally advanced cervical cancer. Ceska Gynekol 71:308–311 Petera J, Odrazka K, Dolezel M, Zoul Z, Vaculikova M, Sefrova J, Sirak I, 2007, Extended field radiotherapy and high-dose brachytherapy combined with chemotherapy in patients with locally advanced cervical carcinoma. Ceska Gynekol 72:126–130 Sirak I, Petera J, Zoul Z, 2008, The influence of cisplatin dose upon survival in concurrent chemoradiotherapy of locally advanced cervical carcinoma with weekly cisplatin. Acta Medica, Hradec Kralove, 51:95–99 Siriaunkgul S, Suwiwat S, Settakorn J, Khunamornpong S, Tungsinmunkong K, Boonthum A, Chaisuksunt V, Lekawanvijit S, Srisomboon J, Thorner PS, 2008, HPV genotyping in cervical cancer in Northern Thailand: adapting the linear array HPV assay for use on paraffin-embedded tissue. Gynecol Oncol 108:555–560., DOI 10.1016/j.ygyno.2007.11.016 Harima Y, Sawada S, Nagata K, Sougawa M, Ohnishi T, 2002, Human papilloma virus, HPV, DNA associated with prognosis of cervical cancer after radiotherapy. Int J Radiat Oncol Biol Phys 52:1345–1351 Lindel K, Burri P, Studer HU, Altermatt HJ, Greiner RH, Gruber G, 2005, Human papillomavirus status in advanced cervical cancer: predictive and prognostic significance for curative radiation treatment. Int J Gynecol Cancer 15:278–284 Herrington CS, 1999, Do HPV-negative cervical carcinomas exist?– revisited. J Pathol 189:1–3 Lee CM, Shrieve DC, Zempolich KA, Lee RJ, Hammond E, Handrahan DL, Gaffney DK, 2005, Correlation between human epidermal growth factor receptor family, EGFR, HER2, HER3, HER4), phosphorylated Akt, P-Akt), and clinical outcomes after radiation therapy in carcinoma of the cervix. Gynecol Oncol 99:415–421 Yamashita H, Murakami N, Asari T, Okuma K, Ohtomo K, Nakagawa K, 2008, Correlation among six biologic factors, p53, p21(WAF1), MIB-1, EGFR, HER2, and Bcl-2, and clinical outcomes after curative chemoradiation therapy in squamous cell cervical cancer. Int J Radiat Oncol Biol Phys 74:1165–1172., DOI 10.1016/j.ijrobp.2008.09.005 Niibe Y, Nakano T, Ohno T, Suzuki Y, Oka K, Tsujii H, 2003, Prognostic significance of c-erbB-2/HER2 expression in advanced uterine cervical carcinoma with para-aortic lymph node metastasis treated with radiation therapy. Int J Gynecol Cancer 13:849–855 Shen L, Shui Y, Wang X, Sheng L, Yang Z, Xue D, Wei Q, 2008, EGFR and HER2 expression in primary cervical cancers and corresponding lymph node metastases: implications for targeted radiotherapy. BMC Cancer 8:232., DOI 10.1186/1471-2407-8-232 Noordhuis MG, Eijsink JJ, Ten Hoor KA, Roossink F, Hollema H, Arts HJ, Pras E, Maduro JH, Reyners AK, de Bock GH, Wisman GB, Schuuring E, van der Zee AG, 2009, Expression of epidermal growth factor receptor, EGFR, and activated EGFR predict poor response to, chemo)radiation and survival in cervical cancer. Clin Cancer Res 15:7389–7397., DOI 10.1158/1078-0432.CCR-09-1149 Cho NH, Kim YB, Park TK, Kim GE, Park K, Song KJ, 2003, P63 and EGFR as prognostic predictors in stage IIB radiation-treated cervical squamous cell carcinoma. Gynecol Oncol 91:346–353 Loncaster JA, Cooper RA, Logue JP, Davidson SE, Hunter RD, West CM, 2000, Vascular endothelial growth factor, VEGF, expression is a prognostic factor for radiotherapy outcome in advanced carcinoma of the cervix. Br J Cancer 83:620–625 Kang JO, Hong SE, 2004, The prognostic effect of VEGF expression in squamous cell carcinoma of the cervix treated with radiation therapy alone. J Korean Med Sci 19:693–697 Gaffney DK, Haslam D, Tsodikov A, Hammond E, Seaman J, Holden J, Lee RJ, Zempolich K, Dodson M, 2003, Epidermal growth factor receptor, EGFR, and vascular endothelial growth factor, VEGF, negatively affect overall survival in carcinoma of the cervix treated with radiotherapy. Int J Radiat Oncol Biol Phys 56:922–928 Randall LM, Monk BJ, Darcy KM, Tian C, Burger RA, Liao SY, Peters WA, Stock RJ, Fruehauf JP, 2009, Markers of angiogenesis in high-risk, early-stage cervical cancer: a Gynecologic Oncology Group study. Gynecol Oncol 112:583–589., DOI 10.1016/j.ygyno. 2008.11.013 Oka K, Suzuki Y, Nakano T, 2000, Expression of p27 and p53 in cervical squamous cell carcinoma patients treated with radiotherapy alone: radiotherapeutic effect and prognosis. Cancer 88:2766–2773 Jain D, Srinivasan R, Patel FD, Kumari Gupta S, 2003, Evaluation of p53 and Bcl-2 expression as prognostic markers in invasive cervical carcinoma stage IIb/III patients treated by radiotherapy. Gynecol Oncol 88:22–28 Lindstrom AK, Stendahl U, Tot T, Lidstrom BM, Hellberg D, 2007, Predicting the outcome of squamous cell carcinoma of the uterine cervix using combinations of individual tumor marker expressions. Anticancer Res 27:1609–1615 Ebara T, Mitsuhashi N, Saito Y, Sakurai H, Hasegawa M, Takahashi M, Takahashi T, Hayakawa K, Niibe H, 1996, Prognostic significance of immunohistochemically detected p53 protein expression in stage IIIB squamous cell carcinoma of the uterine cervix treated with radiation therapy alone. Gynecol Oncol 63:216–218 Ishikawa H, Mitsuhashi N, Sakurai H, Maebayashi K, Niibe H, 2001, The effects of p53 status and human papillomavirus infection on the clinical outcome of patients with stage IIIB cervical carcinoma treated with radiation therapy alone. Cancer 91:80–89 Wootipoom V, Lekhyananda N, Phungrassami T, Boonyaphiphat P, Thongsuksai P, 2004, Prognostic significance of Bax, Bcl-2, and p53 expressions in cervical squamous cell carcinoma treated by radiotherapy. Gynecol Oncol 94:636–642 Nakano T, Oka K, Ishikawa A, Morita S, 1998, Immunohistochemical prediction of radiation response and local control in radiation therapy for cervical cancer. Cancer Detect Prev 22:120–128 Pillai MR, Jayaprakash PG, Nair MK, 1998, Tumour-proliferative fraction and growth factor expression as markers of tumour response to radiotherapy in cancer of the uterine cervix. J Cancer Res Clin Oncol 124:456–461 Suzuki M, Tsukagoshi S, Saga Y, Ohwada M, Sato I, 2000, Assessment of proliferation index with MIB-1 as a prognostic factor in radiation therapy for cervical cancer. Gynecol Oncol 79:300–304 Oka K, Arai T, 1996, MIB1 growth fraction is not related to prognosis in cervical squamous cell carcinoma treated with radiotherapy. Int J Gynecol Pathol 15:23–27 Levine EL, Renehan A, Gossiel R, Davidson SE, Roberts SA, Chadwick C, Wilks DP, Potten CS, Hendry JH, Hunter RD et al, 1995, Apoptosis, intrinsic radiosensitivity and prediction of radiotherapy response in cervical carcinoma. Radiother Oncol 37:1–9 Sobin LH, Wittekind C, 2002, UICC international union against cancer TNM classification of malignant tumours, 6th edn. Wiley- Liss, New York NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 131 EP 137 DI 10.1007/s12253-013-9674-5 PG 7 ER PT J AU Bardi, E Csoka, M Garai, I Szegedi, I Muller, J Gyorke, T Kajary, K Nemes, K Kiss, Cs Kovacs, G AF Bardi, Edit Csoka, Monika Garai, Ildiko Szegedi, Istvan Muller, Judit Gyorke, Tamas Kajary, Kornelia Nemes, Karolina Kiss, Csongor Kovacs, Gabor TI Value of FDG-PET/CT Examinations in Different Cancers of Children, Focusing on Lymphomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Childhood cancers; FDG-PET/CT; Hodgkin; Non-Hodgkin lymphoma; High grade solid tumors ID Childhood cancers; FDG-PET/CT; Hodgkin; Non-Hodgkin lymphoma; High grade solid tumors AB The aim of the study was to assess sensitivity and specificity of FDG-PET/CT in different forms of childhood cancer. We retrospectively evaluated the results dedicated of 162 FDG-PET/CT examinations of 86 children treated with: Hodgkin lymphoma (HL; n=31), non-Hodgkin lymphoma (NHL; n=30) and other high grade solid tumors (n=25). Patients were admitted and treated in two departments of pediatric hematology and oncology in Hungary. FDG-PET/CT was performed for staging (n=25) and for posttreatment evaluation (n=137). Imaging was performed in three FDG-PET/CT Laboratories, using dedicated PET/CT scanners. False positive results were defined as resolution or absence of disease progression over at least 1 year on FDG-PET/CT-scans without any intervention. In some cases histopathological evaluation of suspicious lesions was performed. Fals negative results were defined as negative FDG-PET/CT results in case of active malignancy. Positive predictive values (PPV) and negative predictive values (NPV) were calculated. NPV was 100 %. The highest PPV was observed in high grade solid tumors (81 %), followed by HL (65 %) and NHL (61 %). There was a major difference of PPV in different histological types of HL (50 % in HL of mixed-cellularity subtype, 90 % in nodular sclerosing, and 100 % in lymphocyte-rich and lymphocyte depleted HL). We treated one patient with nodular lymphocyte predominant HL, who had 5 false positive FDG-PET/CT results. PPV of Tand B-lineage NHL were similar (60%and 62 %, respectively). We observed an interesting difference of PPV in different stages of HL and NHL. In HL PPV was higher in early than in advanced disease forms: 66 % in stage II HL and 60 % in stage III HL, whereas there was an inverse relationship between PPV and disease stages in NHL 0%in stage I and II patients, 67%in stage III and 100 % in stage IV patients. PPV was lower in males (54 %) than in females (65 %). PPV were 64 % vs. 58 % in patients under vs. over 10 years of age. Negative FDG-PET/ CT results during follow-up reliably predict the absence of malignancy. Positive FDG-PET/CT scan results in general have a low PPV. The relatively high PPV in patients with histologically proven high grade solid tumors, advanced stages of NHL and with nodular sclerosing, lymphocyte-rich and lymphocyte depleted subtypes of HL warrant a confirmation by biopsy, whereas the watch-and-wait approach can be used in other forms of childhood cancer patients with a positive FDG-PET/ CT result in course of follow-up examinations. C1 [Bardi, Edit] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-OncologyDebrecen, Hungary. [Csoka, Monika] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Garai, Ildiko] Scanomed Kft.Debrecen, Hungary. [Szegedi, Istvan] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-OncologyDebrecen, Hungary. [Muller, Judit] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Gyorke, Tamas] Semmelweis University, Department of Radiology and OncotherapyBudapest, Hungary. [Kajary, Kornelia] Pozitron Diagnosztika KftBudapest, Hungary. [Nemes, Karolina] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Kiss, Csongor] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-OncologyDebrecen, Hungary. [Kovacs, Gabor] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. RP Bardi, E (reprint author), University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-Oncology, Debrecen, Hungary. EM editbardi@hotmail.com CR London K, Cross S, Onikul E, Dalla-Pozza L, Howman-Giles R, 2011,, 18)F-FDG PET/CT in paediatric lymphoma: comparison with conventional imaging. Eur J Nucl Med Mol Imaging 38:274–284 Rhodes MM, Delbeke D, Whitlock JA, Martin W, Kuttesch JF, Frangoul HA, Shankar S, 2006, Utility of FDG-PET/CT in follow up of children treated for Hodgkin and non-Hodgkin lymphoma. J Pediatr Hematol Oncol 28:300–306 KleisM, Daldrup-Link H,MatthayK et al, 2009, Diagnostic value of PET CT for the staging and restaging of pediatric tumors. Eur J Nucl Med Mol Imaging 36:23–36 Portwine C, Marriott C, Barr RD, 2010, PET imaging for pediatric oncology: an assessment of the evidence. Pediatr Blood Cancer 55:1048–1061 Nanni C, RubelloD, Castellucci P, FarsadM, Franchi R, Rampin L, Al- Nahhas A, Fanti S, 2006, 18F-FDG PET/CT fusion imaging in paediatric solid extracranial tumours. Biomed Pharmacother 60:593–606 Franzius C, Juergens KU, Schober O, 2006, Is PET CT necessary in pediatric oncology? For Eur J Nucl Med Mol Imaging 33:960–965 Jadvar H, Connoly LP, Fahey FH et al, 2007, PET and PET CT in pediatric oncology. Semin Nucl Med 37:316–331 Shulkin BL, Mitchell DS, Ungar DR et al, 1995, Neoplasms in a pediatric population: 2-[F-18]-fluoro-2-deoxy-D-glucose PET studies. Radiology 94:3277–3284 Hawkins DS, Rajendran JG, Conrad EU III et al, 2002, Evaluation of chemotherapy response in pediatric bone sarcomas by -[F-18]-fluoro- 2-deoxy-D-glucose positron emission tomography. Cancer 94:3277– 3284 Wegner EA, Barrington SF, Kingston JE et al, 2005, The impact of PET scanning on management of paediatric oncology patients. Eur J Nucl Med Mol Imaging 32:23–30 Bar-Sever Z, Keidar Z, Ben-Barak A et al, 2007, The incremental value of, 18, F-FDG PET/CT in pediatric malignancies. Eur J Nucl Med Mol Imaging 34:628–629 Frazius C, Juergens KU,Vormoor J, 2006, PET/CTwith diagnostic CT in the evaluation of childhood sarcoma.AmJ Roentgenol 186:581–582 McCarville MB, Christie R, Daw NC, Spunt SL, Kaste SC, 2005, PET/CT in evaluation of childhood sarcomas. Am J Roentgenol 184:1293–1304 Miller E, Metser U, Avrahami G et al, 2006, Role of 18FFDG PET CT in staging and follow up of lymphoma in pediatric and young adult patients. J Comput Assist Tomogr 30:689–694 Schoder H, Noy A, Gonen M et al, 2005, Intensity of 18 fluorodeoxyglucose uptake in positron emission tomography distinguishes between indolent and aggressive non-Hodgkin’s lymphoma. J Clin Oncol 23:4643–4651 Mody RJ, Bui C, Hutchinson RJ et al, 2010, FDG PET imaging in childhood sarcomas. Pediatr Blood Cancer 54:222–227 Kim DW, Jung SA, Kim CG, Park SA, 2010, The efficacy of dual time point F-18 FDG PET imaging for grading of brain tumors. Clin Nucl Med 35:400–403 Heyman S, Evans EA, D’Angio GJ, 1998, I-131 metiaiodobenzylguanidine: diagnostic use in neuroblastoma patients in relapse. Med Pediatr Oncol 16:337–340 Colavolpe C, Ued JE, Cammilleri S et al, 2008, Utility of FDG PET CT in the follow up of neuroblastoma which became MIBIGnegative. Pediatr Blood Cancer 51:828–831 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 139 EP 143 DI 10.1007/s12253-013-9676-3 PG 5 ER PT J AU Arzt, L Quehenberger, F Halbwedl, I Mairinger, Th Popper, HH AF Arzt, Lisa Quehenberger, Franz Halbwedl, Iris Mairinger, Thomas Popper, H Helmut TI BAP1 Protein is a Progression Factor in Malignant Pleural Mesothelioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BRCA1-associated protein1 (BAP1); Malignant pleuralmesothelioma; Asbestos exposure; Overall survival ID BRCA1-associated protein1 (BAP1); Malignant pleuralmesothelioma; Asbestos exposure; Overall survival AB Human malignant pleural mesothelioma (MPM) is an aggressive cancer due to former asbestos exposure with little knowledge about prognostic factors of outcome and resistance to conventional therapy. BRCA1-associated protein 1 (BAP1) is a tumor suppressor gene that is frequently lost in MPM. Germline mutations of BAP1 predispose to several different tumors including malignant mesothelioma. Our study aimed to clarify if asbestos exposure has an influence on BAP1 expression and if BAP1 expression could be used as a prognostic factor of outcome. An immunohistochemical staining for BAP1 was performed on 123 MPM tissue samples and the expression levels have been correlated with asbestos exposure and overall survival time. BAP1 expression was not associated with asbestos exposure but we detected a significant effect of BAP1 expression on overall survival time - the higher the BAP1 expression (non-mutated BAP1), the shorter the overall survival. BAP1 mutation has been linked to nonasbestos induced familial mesotheliomas, which usually belong to the long survivor group and BAP1 is most probably functioning differently than in sporadic cases. Further investigations need to be performed to characterize the BAP1 mutations and to identify the BAP1 downstream targets in MPM. C1 [Arzt, Lisa] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, 8036 Graz, Austria. [Quehenberger, Franz] Medical University of Graz, Institute for Medical Informatics, Statistics and Documentation, Auenbruggerplatz 2/V, 8036 Graz, Austria. [Halbwedl, Iris] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, 8036 Graz, Austria. [Mairinger, Thomas] Helios Klinikum Berlin, Institute of Pathology, Walterhoferstraße 11, 14156 Berlin, Germany. [Popper, H Helmut] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, 8036 Graz, Austria. RP Arzt, L (reprint author), Medical University of Graz, Department of Pathology, 8036 Graz, Austria. EM lisa.arzt@medunigraz.at CR Tomasetti M, Amati M, Santarelli L, Alleva R, Neuzil J, 2009, Malignant mesothelioma: biology, diagnosis and therapeutic approaches. Curr Mol Pharmacol 2(2):190–206 Paleari L, Rotolo N, Imperatori A, Puzone R, Sessa F, Franzi F, Meacci E, Camplese P, Cesario A, Paganuzzi M, 2009, Osteopontin is not a specific marker in malignant pleural mesothelioma. Int J Biol Markers 24(2):112–117 Belli C, Fennell D, Giovannini M, Gaudino G, Mutti L, 2009, Malignant pleural mesothelioma: current treatments and emerging drugs. Expert Opin Emerg Drugs 14(3):423–437., DOI 10.1517/ 14728210903074563 Neragi-Miandoab S, 2006, Multimodality approach in management of malignant pleural mesothelioma. Eur J Cardiothorac Surg 29(1):14–19., DOI 10.1016/j.ejcts.2005.10.008 Treasure T, Sedrakyan A, 2004, Pleural mesothelioma: little evidence, still time to do trials. Lancet 364(9440):1183–1185., DOI 10. 1016/s0140-6736(04)17108-0 Moore AJ, Parker RJ, Wiggins J, 2008, Malignant mesothelioma. Orphanet J Rare Dis 3:34., DOI 10.1186/1750-1172-3-34 Montjoy C, Parker J, Petsonk L, Luis T, Fallon K, 2009, Mesothelioma review. W V Med J 105(3):13–16 Gordon GJ, Appasani K, Parcells JP, Mukhopadhyay NK, Jaklitsch MT, Richards WG, Sugarbaker DJ, Bueno R, 2002, Inhibitor of apoptosis protein-1 promotes tumor cell survival in mesothelioma. Carcinogenesis 23(6):1017–1024 He B, You L, Uematsu K, Zang K, Xu Z, Lee AY, Costello JF, McCormick F, Jablons DM, 2003, SOCS-3 is frequently silenced by hypermethylation and suppresses cell growth in human lung cancer. Proc Natl Acad Sci U S A 100(24):14133–14138., DOI 10. 1073/pnas.2232790100 Xia C, Xu Z, Yuan X, Uematsu K, You L, Li K, Li L, McCormick F, Jablons DM, 2002, Induction of apoptosis in mesothelioma cells by antisurvivin oligonucleotides. Mol Cancer Ther 1(9):687–694 Ceresoli GL, Locati LD, Ferreri AJ, Cozzarini C, Passoni P, Melloni G, Zannini P, Bolognesi A, Villa E, 2001, Therapeutic outcome according to histologic subtype in 121 patientswithmalignant pleural mesothelioma. Lung Cancer 34(2):279–287 Flores RM, Zakowski M, Venkatraman E, Krug L, Rosenzweig K, Dycoco J, Lee C, Yeoh C, Bains M, Rusch V, 2007, Prognostic factors in the treatment of malignant pleural mesothelioma at a large tertiary referral center. J Thorac Oncol 2(10):957–965., DOI 10.1097/ JTO.0b013e31815608d9 Proietti L, Spicuzza L, Di Maria A, Polosa R, Sebastian Torres E, Asero V, Di Maria GU, 2006, Non-occupational malignant pleural mesothelioma due to asbestos and non-asbestos fibres.Monaldi Arch Chest Dis 65(4):210–216 Testa JR, Cheung M, Pei J, Below JE, Tan Y, Sementino E, Cox NJ, Dogan AU, Pass HI, Trusa S, Hesdorffer M, Nasu M, Powers A, Rivera Z, Comertpay S, Tanji M, Gaudino G, Yang H, Carbone M, 2011, Germline BAP1 mutations predispose to malignant mesothelioma. Nat Genet 43(10):1022–1025., DOI 10.1038/ng.912 Jensen DE, ProctorM,Marquis ST, GardnerHP, Ha SI, Chodosh LA, Ishov AM, Tommerup N, Vissing H, Sekido Y,Minna J, Borodovsky A, Schultz DC, Wilkinson KD, Maul GG, Barlev N, Berger SL, Prendergast GC, Rauscher FJ 3rd, 1998, BAP1: a novel ubiquitin hydrolase which binds to the BRCA1 RING finger and enhances BRCA1-mediated cell growth suppression. Oncogene 16(9):1097– 1112 Jensen DE, Rauscher FJ 3rd, 1999, BAP1, a candidate tumor suppressor protein that interacts with BRCA1. Ann N Y Acad Sci 886:191–194 Ventii KH, Devi NS, Friedrich KL, Chernova TA, Tighiouart M, Van Meir EG, Wilkinson KD, 2008, BRCA1-associated protein-1 is a tumor suppressor that requires deubiquitinating activity and nuclear localization. Cancer Res 68(17):6953–6962., DOI 10.1158/0008-5472. can-08-0365 Harbour JW, Onken MD, Roberson ED, Duan S, Cao L,Worley LA, Council ML, Matatall KA, Helms C, Bowcock AM, 2010, Frequent mutation of BAP1 in metastasizing uveal melanomas. Science 330(6009):1410–1413., DOI 10.1126/science.1194472 Wiesner T, Obenauf AC, Murali R, Fried I, Griewank KG, Ulz P, Windpassinger C,WackernagelW, Loy S,Wolf I, Viale A, Lash AE, PirunM, Socci ND, Rutten A, Palmedo G, Abramson D, Offit K, Ott A, Becker JC, Cerroni L, Kutzner H, Bastian BC, Speicher MR, 2011, Germline mutations in BAP1 predispose to melanocytic tumors. Nat Genet 43(10):1018–1021., DOI 10.1038/ng.910 Abdel-Rahman MH, Pilarski R, Cebulla CM, Massengill JB, Christopher BN, Boru G, Hovland P, Davidorf FH, 2011, Germline BAP1 mutation predisposes to uveal melanoma, lung adenocarcinoma, meningioma, and other cancers. J Med Genet 48(12):856–859., DOI 10.1136/jmedgenet-2011-100156 Carbone M, Korb Ferris L, Baumann F, Napolitano A, Lum CA, Flores EG, Gaudino G, Powers A, Bryant-Greenwood P, Krausz T, Hyjek E, Tate R, Friedberg J, Weigel T, Pass HI, Yang H, 2012, BAP1 cancer syndrome: malignant mesothelioma, uveal and cutaneous melanoma, and MBAITs. J Transl Med 10(1):179., DOI 10.1186/ 1479-5876-10-179 Wiesner T, Fried I, Ulz P, Stacher E, Popper H,Murali R, Kutzner H, Lax S, Smolle-Juttner F, Geigl JB, Speicher MR, 2012, Toward an improved definition of the tumor spectrum associated with BAP1 germline mutations. J Clin Oncol., DOI 10.1200/jco.2011.41.2965 Flores RM, Pass HI, Seshan VE, Dycoco J, Zakowski M, CarboneM, Bains MS, Rusch VW, 2008, Extrapleural pneumonectomy versus pleurectomy/decortication in the surgical management of malignant pleural mesothelioma: results in 663 patients. J Thorac Cardiovasc Surg 135(3):620–626., DOI 10.1016/j.jtcvs.2007.10.054, 626 e621-623 Flores RM, 2009, Surgical options in malignant pleural mesothelioma: extrapleural pneumonectomy or pleurectomy/decortication. Semin Thorac Cardiovasc Surg 21(2):149–153., DOI 10.1053/j. semtcvs.2009.06.008 Bott M, Brevet M, Taylor BS, Shimizu S, Ito T, Wang L, Creaney J, Lake RA, Zakowski MF, Reva B, Sander C, Delsite R, Powell S, Zhou Q, Shen R, Olshen A, Rusch V, LadanyiM, 2011, The nuclear deubiquitinase BAP1 is commonly inactivated by somatic mutations and 3p21.1 losses in malignant pleural mesothelioma. Nat Genet 43(7):668–672., DOI 10.1038/ng.855 Ladanyi M, Zauderer MG, Krug LM, Ito T, McMillan R, Bott M, Giancotti FG, 2012, New strategies in pleural mesothelioma: BAP1 and NF2 as novel targets for therapeutic development and risk assessment. Clin Cancer Res., DOI 10.1158/1078- 0432.ccr-11-2375 Yoshikawa Y, Sato A, Tsujimura T, Emi M, Morinaga T, Fukuoka K, Yamada S, Murakami A, Kondo N, Matsumoto S, Okumura Y, Tanaka F, Hasegawa S, Nakano T, Hashimoto-Tamaoki T, 2012, Frequent inactivation of the BAP1 gene in epithelioid-typemalignant mesothelioma. Cancer Sci 103(5):868–874., DOI 10.1111/j.1349- 7006.2012.02223.x Kittler R, Pelletier L, Heninger AK, Slabicki M, Theis M, Miroslaw L, Poser I, Lawo S, Grabner H, Kozak K,Wagner J, Surendranath V, Richter C, Bowen W, Jackson AL, Habermann B, Hyman AA, Buchholz F, 2007, Genome-scale RNAi profiling of cell division in human tissue culture cells. Nat Cell Biol 9(12):1401–1412., DOI 10. 1038/ncb1659 Mallery DL, Vandenberg CJ, Hiom K, 2002, Activation of the E3 ligase function of the BRCA1/BARD1 complex by polyubiquitin chains. EMBO J 21(24):6755–6762 Schlabach MR, Luo J, Solimini NL, Hu G, Xu Q, Li MZ, Zhao Z, Smogorzewska A, Sowa ME, Ang XL, Westbrook TF, Liang AC, Chang K, Hackett JA, Harper JW, Hannon GJ, Elledge SJ, 2008, Cancer proliferation gene discovery through functional genomics. Science 319(5863):620–624., DOI 10.1126/science.1149200 Illei PB, Rusch VW, ZakowskiMF, LadanyiM(2003, Homozygous deletion of CDKN2A and codeletion of the methylthioadenosine phosphorylase gene in the majority of pleural mesotheliomas. Clin Cancer Res 9(6):2108–2113 Dacic S, Kothmaier H, Land S, Shuai Y, Halbwedl I, Morbini P, Murer B, Comin C, Galateau-Salle F, Demirag F, Zeren H, Attanoos R, GibbsA, Cagle P, PopperH(2008, Prognostic significance of p16/ cdkn2a loss in pleural malignant mesotheliomas. Virchows Arch 453(6):627–635., DOI 10.1007/s00428-008-0689-3 Thurneysen C, Opitz I, Kurtz S, Weder W, Stahel RA, Felley-Bosco E, 2009, Functional inactivation of NF2/merlin in human mesothelioma. Lung Cancer 64(2):140–147., DOI 10.1016/j.lungcan.2008.08. 014 Machida YJ, Machida Y, Vashisht AA, Wohlschlegel JA, Dutta A, 2009, The deubiquitinating enzyme BAP1 regulates cell growth via interaction with HCF-1. J Biol Chem 284(49):34179–34188., DOI 10. 1074/jbc.M109.046755 Misaghi S, Ottosen S, Izrael-Tomasevic A, Arnott D, Lamkanfi M, Lee J, Liu J, O’Rourke K, Dixit VM,Wilson AC, 2009, Association of C-terminal ubiquitin hydrolase BRCA1-associated protein 1 with cell cycle regulator host cell factor 1. Mol Cell Biol 29(8):2181– 2192., DOI 10.1128/mcb.01517-08 Sowa ME, Bennett EJ, Gygi SP, Harper JW, 2009, Defining the human deubiquitinating enzyme interaction landscape. Cell 138(2):389–403., DOI 10.1016/j.cell.2009.04.042 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 145 EP 151 DI 10.1007/s12253-013-9677-2 PG 7 ER PT J AU Catrina (Ene), MA Borze, I Guled, M Costache, M Leen, G Sajin, M Ionica, E Chitu, A Knuutila, S AF Catrina (Ene), Maria Ana Borze, Ioana Guled, Mohamed Costache, Mariana Leen, Gayle Sajin, Maria Ionica, Elena Chitu, Aura Knuutila, Sakari TI MicroRNA Expression Profiles in Kaposi’s Sarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Array profiling; FFPE; Kaposi sarcoma; miRNA; Quantitative reverse transcriptase polymerase chain reaction ID Array profiling; FFPE; Kaposi sarcoma; miRNA; Quantitative reverse transcriptase polymerase chain reaction AB Kaposi’s sarcoma (KS) is a mesenchymal tumor, caused by Human herpesvirus 8 (HHV8) with molecular and cytogenetic changes poorly understood. To gain further insight on the underlying molecular changes in KS, we performed microRNA (miRNA) microarray analysis of 17 Kaposi’s sarcoma specimens. Three normal skin specimens were used as controls. The most significant differentially expressed miRNA were confirmed by quantitative reverse transcriptase polymerase chain reaction (RT-PCR). We detected in KS versus normal skin 185 differentially expressed miRNAs, 76 were upregulated and 109 were downregulated. The most significantly downregulated miRNAs were miR-99a, miR- 200 family, miR-199b-5p, miR-100 and miR-335, whereas kshv-miR-K12-4-3p, kshv-miR-K12-1, kshv-miR-K12-2, kshv-miR-K12-4-5p and kshv-miR-K12-8 were significantly upregulated. High expression levels of kshv-miR-K12-1 (p=0.004) and kshv-miR-K12-4-3p (p=0.001) was confirmed by RT-PCR. The predicted target genes for differentially expressed miRNAs included genes which are involved in a variety of cellular processes such as angiogenesis (i.e. THBS1) and apoptosis (i.e. CASP3, MCL1), suggesting a role for these miRNAs in Kaposi’s sarcoma pathogenesis. C1 [Catrina (Ene), Maria Ana] University of Bucharest, Faculty of Biology, Department of Biochemistry and Molecular Biology, 050095 Bucharest, Romania. [Borze, Ioana] University of Helsinki, Haartman Institute and HUSLAB, Department of Pathology, Haartmaninkatu 3, 00014 Helsinki, Finland. [Guled, Mohamed] University of Helsinki, Haartman Institute and HUSLAB, Department of Pathology, Haartmaninkatu 3, 00014 Helsinki, Finland. [Costache, Mariana] Department of Pathology, 050471 Bucharest, Romania. [Leen, Gayle] University of Helsinki, Haartman Institute and HUSLAB, Department of Pathology, Haartmaninkatu 3, 00014 Helsinki, Finland. [Sajin, Maria] Department of Pathology, 050471 Bucharest, Romania. [Ionica, Elena] University of Bucharest, Faculty of Biology, Department of Biochemistry and Molecular Biology, 050095 Bucharest, Romania. [Chitu, Aura] University of Bucharest, Faculty of Biology, Department of Biochemistry and Molecular Biology, 050095 Bucharest, Romania. [Knuutila, Sakari] University of Helsinki, Haartman Institute and HUSLAB, Department of Pathology, Haartmaninkatu 3, 00014 Helsinki, Finland. RP Knuutila, S (reprint author), University of Helsinki, Haartman Institute and HUSLAB, Department of Pathology, 00014 Helsinki, Finland. EM sakari.knuutila@helsinki.fi CR Foreman KE, 2001, Kaposis sarcoma: the role of HHV-8 and HIV-1 in pathogenesis. Expert Rev Mol Med 2001:1–17 Feller L, Lemmer J, Wood NH et al, 2007, HIV-associated oral kaposi sarcoma and HHV-8: a review. J Int Acad Periodontol 9:129–136 Ambros V, 2003, MicroRNA pathways in flies and worms: growth, death, fat, stress, and timing. Cell 113:673–676 Antman K, Chang Y, 2000, Kaposi’s sarcoma. N Engl J Med 342:1027–1038 Kiuru-Kuhlefelt S, Sarlomo-Rikala M, Larramendy ML et al, 2000, FGF4 and INT2 oncogenes are amplified and expressed in kaposi’s sarcoma. Mod Pathol 13:433–437 Sun R, Lin SF, Staskus K et al, 1999, Kinetics of kaposi’s sarcomaassociated herpesvirus gene expression. J Virol 73:2232–2242 Raggo C, Ruhl R, McAllister S et al, 2005, Novel cellular genes essential for transformation of endothelial cells by kaposi’s sarcomaassociated herpesvirus. Cancer Res 65:5084–5095 Samols MA, Skalsky RL, Maldonado AM et al, 2007, Identification of cellular genes targeted by KSHV-encoded microRNAs. PLoS Pathog 3:e65 Croce CM, 2009, Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet 10:704–714 Sassen S, Miska EA, Caldas C, 2008, MicroRNA: implications for cancer. Virchows Arch 452:1–10 Volinia S, Calin GA, Liu CG et al, 2006, A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 103:2257–2261 Lu J, Getz G, Miska EA et al, 2005, MicroRNA expression profiles classify human cancers. Nature 435:834–838 Borze I, Guled M, Musse S et al, 2010, MicroRNA microarrays on archive bone marrow core biopsies of leukemias-method validation. Leuk Res 35:188–195 Zhang X, Chen J, Radcliffe T et al, 2008, An array-based analysis of microRNA expression comparingmatched frozen and formalin-fixed paraffin-embedded human tissue samples. J Mol Diagn 10:513–519 Mosakhani N, Sarhadi VK, Borze I et al, 2012)MicroRNA profiling differentiates colorectal cancer according to KRAS status. Genes Chromosom Cancer 51:1–9 Schmittgen TD, Livak KJ, 2008, Analyzing real-time PCR data by the comparative C(T, method. Nat Protoc 3:1101–1108 Griffiths-Jones S, Saini HK, van Dongen S et al, 2008, miRBase: tools for microRNA genomics. Nucleic Acids Res 36:D154–D158 Lewis BP, Burge CB, Bartel DP, 2005, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120:15–20 Betel D, Wilson M, Gabow A et al, 2008, The microRNA.org resource: targets and expression. Nucleic Acids Res 36:D149–D153 Wang X, El Naqa IM, 2008, Prediction of both conserved and nonconserved microRNA targets in animals. Bioinformatics 24:325–332 Wang X, 2008, miRDB: a microRNA target prediction and functional annotation database with a wiki interface. RNA 14:1012–1017 Nuovo GJ, Wu X, Volinia S et al, 2010, Strong inverse correlation between microRNA-125b and human papillomavirus DNA in productive infection. Diagn Mol Pathol 19:135–143 Schickel R, Boyerinas B, Park SM et al, 2008, MicroRNAs: key players in the immune system, differentiation, tumorigenesis and cell death. Oncogene 27:5959–5974 Merimsky O, Jiveliouk I, Sagi-Eisenberg R, 2008, Targeting mTOR in HIV-negative classic kaposi’s sarcoma. Sarcoma 2008:825093 Gregory PA, Bert AG, Paterson EL et al, 2008, The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol 10:593–601 Karasek M, 2007, Differentiation of postembryonic skin endothelial cells. J Investig Dermatol 127:2710–2712 Park SM, Gaur AB, Lengyel E et al, 2009, The miR-200 family determines the epithelial phenotype of cancer cells by targeting the Ecadherin repressors ZEB1 and ZEB2. Genes Dev 22:894–907 O’Hara AJ, Wang L, Dezube BJ et al, 2009, Tumor suppressor microRNAs are underrepresented in primary effusion lymphoma and kaposi sarcoma. Blood 113:5938–5941 Park SM, Shell S, Radjabi AR et al, 2007, Let-7 prevents early cancer progression by suppressing expression of the embryonic gene HMGA2. Cell Cycle 6:2585–2590 Johnson SM, Grosshans H, Shingara J et al, 2005, RAS is regulated by the let-7 microRNA family. Cell 120:635–647 Zhang B, Pan X, CobbGP et al, 2007)microRNAs as oncogenes and tumor suppressors. Dev Biol 302:1–12 Moustakas A, Pardali K, Gaal A et al, 2002, Mechanisms of TGFbeta signaling in regulation of cell growth and differentiation. Immunol Lett 82:85–91 Di Bartolo DL, Cannon M, Liu YF et al, 2008, KSHV LANA inhibits TGF-beta signaling through epigenetic silencing of the TGF-beta type II receptor. Blood 111:4731–4740 Seo T, Park J, Choe J, 2005, Kaposi’s sarcoma-associated herpesvirus viral IFN regulatory factor 1 inhibits transforming growth factorbeta signaling. Cancer Res 65:1738–1747 Tomita M, Choe J, Tsukazaki T et al, 2004, The kaposi’s sarcomaassociated herpesvirus K-bZIP protein represses transforming growth factor beta signaling through interaction with CREB-binding protein. Oncogene 23:8272–8281 Meng F, Henson R, Wehbe-Janek H et al, 2007, MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 133:647–658 Zhu S, Si ML, Wu H et al, 2007, MicroRNA-21 targets the tumor suppressor gene tropomyosin 1, TPM1). J Biol Chem 282:14328–14336 Moore PS, Chang Y, 2001)Molecular virology of kaposi’s sarcomaassociated herpesvirus. Philos TransR Soc Lond B Biol Sci 356:499– 516 Mott JL, Kobayashi S, Bronk SF et al, 2007, Mir-29 regulates mcl-1 protein expression and apoptosis. Oncogene 26:6133–6140 Marshall V, Martro E, Labo N et al, 2010, Kaposi sarcoma, KS)- associated herpesvirus microRNA sequence analysis and KS risk in a european AIDS-KS case control study. J Infect Dis 202:1126–1135 Hansen A, Henderson S, Lagos D et al, 2010, KSHV-encoded miRNAs target MAF to induce endothelial cell reprogramming. Genes Dev 24:195–205 Cai X, Lu S, Zhang Z et al, 2005, Kaposi’s sarcoma-associated herpesvirus expresses an array of viral microRNAs in latently infected cells. Proc Natl Acad Sci U S A 102:5570–5575 Qin Z, Kearney P, Plaisance K et al, 2010, Pivotal advance: Kaposi’s sarcoma-associated herpesvirus, KSHV)-encoded microRNA specifically induce IL-6 and IL-10 secretion by macrophages and monocytes. J Leukoc Biol 87:25–34 Bellare P, Ganem D, 2009, Regulation of KSHV lytic switch protein expression by a virus-encoded microRNA: an evolutionary adaptation that fine-tunes lytic reactivation. Cell Host Microbe 6:570–575 Suffert G, Malterer G, Hausser J et al, 2011, Kaposi’s sarcoma herpesvirus microRNAs target caspase 3 and regulate apoptosis. PLoS Pathog 7:e1002405 Abend JR, Uldrick T, Ziegelbauer JM, 2010, Regulation of tumor necrosis factor-like weak inducer of apoptosis receptor protein, TWEAKR, expression by kaposi’s sarcoma-associated herpesvirus microRNA prevents TWEAK-induced apoptosis and inflammatory cytokine expression. J Virol 84:12139–121351 Ziegelbauer JM, Sullivan CS, Ganem D, 2009, Tandem array-based expression screens identify host mRNA targets of virus-encoded microRNAs. Nat Genet 41:130–134 Calin GA, Sevignani C, Dumitru CD et al, 2004, Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A 101:2999–3004 Pyakurel P, Montag U, Castanos-Velez E et al, 2006, CGH of microdissected kaposi’s sarcoma lesions reveals recurrent loss of chromosome Y in early and additional chromosomal changes in late tumour stages. AIDS 20:1805–1812 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 153 EP 159 DI 10.1007/s12253-013-9678-1 PG 7 ER PT J AU Zhang, J Xu, H Ren, F Yang, Y Chen, B Zhang, F AF Zhang, Jinnan Xu, Haiyan Ren, Fang Yang, Yijin Chen, Bin Zhang, Fengchun TI Analysis of Clinicopathological Features and Prognostic Factors of Desmoplastic Small Round Cell Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Desmoplastic small round cell tumor; Prognosis ID Desmoplastic small round cell tumor; Prognosis AB Desmoplastic small round cell tumor (DSRCT) is a relatively uncommon and highly aggressive malignancy in young males. It is associated with a poor outcome, due in part to missed diagnosis. To characterize the clinical pathological features of DSRCT in Chinese patients and to find out the characteristics of treatment and prognostic factors, the authors collected and analyzed the clinical information of 48 cases. A total of 48 cases of DSRCT between March 1995 and March 2012were retrospectively reviewed and analyzed. The clinical information, histological results and survival data of the patients were collected. Median age was 26.96±14.09 years with a range of 6–66 years. Thirty-three patients (68.75 %) were seen before 30 years old, and 15 patients (31.25 %) were diagnosed after 30 years old. The male-to-female ratio is 3.36 :1. Among them, 37 cases presented with tumors in the abdominal or pelvic cavity; the other 11 cases had extraabdominal tumors. The most common symptoms were abdominal pain (19/48, 39.58 %) and palpable mass (12/48, 25.00 %). The percentage of patients received surgery, complete surgery, and chemotherapy was 79.17 %, 37.50 %, and 52.08 %, respectively. Median follow-up duration was 2.67 years. Median overall survival for all patients was 24.33 months (95 % CI: 9.74–38.92 months) and median event-free survival for all patients was 8.00 months (95 % CI: 5.13–10.89 months). Univariate analysis revealed that surgery, effective debulking surgery, chemotherapy and any two or more combined therapeutics were significant prognostic factors for longer overall survival (p<0.05). Cox regression analysis showed complete surgery was an independent prognostic factor. Standard therapy for DSRCT consists of combination of surgical resection and postoperative chemotherapy. Complete surgery is an independent prognostic factor and should be further investigated. C1 [Zhang, Jinnan] Jilin University, China-Japan Union Hospital, Department of Neurosurgery, 130031 Changchun, China. [Xu, Haiyan] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, 215021 Suzhou, China. [Ren, Fang] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, 215021 Suzhou, China. [Yang, Yijin] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, 215021 Suzhou, China. [Chen, Bin] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, 215021 Suzhou, China. [Zhang, Fengchun] Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, 215021 Suzhou, China. RP Zhang, F (reprint author), Shanghai Jiaotong University School of Medicine, Suzhou Kowloon Hospital, Department of Oncology, 215021 Suzhou, China. EM fczhang2004@163.com CR Gerald WL, Rosai J, 1989, Case 2: desmoplastic small round cell tumor with divergent differentiation. Pediatr Pathol 9:177–183 Dufresne A, Cassier P, Couraud L, Marec-Berard P,Meeus P, Alberti L, Blay JY, 2012, Desmoplastic small round cell tumor: current management and recent findings. Sarcoma 2012:714986 Rekhi B, Ahmed S, Basak R, Qureshi SS, Desai SS, Ramadwar M, Desai SB, Kurkure P, Jambhekar NA, 2012, Desmoplastic small round cell tumor-clinicopathological spectrum, including unusual features and immunohistochemical analysis of 45 tumors diagnosed at a tertiary cancer referral centre, with molecular results t(11; 22,, p13; q12,, EWS-WT1, in select cases. Pathol Oncol Res 18:917– 927 Gerald WL, Ladanyi M, de Alava E , Cuatrecasas M, Kushner BH, LaQuaglia MP, Rosai J, 1998, Clinical, pathologic, and molecular spectrum of tumors associated with t(11;22)(p13;q12): desmoplastic small round-cell tumor and its variants. J Clin Oncol 16:3028–3036 Lal DR, SuWT, Wolden SL, Loh KC, Cuatrecasas M, Kushner BH, LaQuaglia MP, Rosai J, 2005, Results of multimodal treatment for desmoplastic small round cell tumors. J Pediatr Surg 40:251–255 Gil A, Gomez Portilla A, Brun EA, Sugarbaker PH, 2004, Clinical perspective on desmoplastic small round-cell tumor. Oncology 67:231–242 PernaMJ, Streck CJ, 2012, Alarge solitary desmoplastic small round cell tumor. Am Surg 78:316–317 Jordan AH, Pappo A, 2012, Management of desmoplastic small round-cell tumors in children and young adults. J Pediatr Hematol Oncol 34:S73–S75 Chang F, 2006, Desmoplastic small round cell tumors: cytologic, histologic, and immunohistochemical features. Arch Pathol LabMed 130:728–732 Harmon RL, Sugarbaker PH, 2005, Prognostic indicators in peritoneal carcinomatosis from gastrointestinal cancer. Int Semin Surg Oncol 2:3 Kallianpur AA, Shukla NK, Deo SV, Yadav P, Mudaly D, Yadav R, Palaniappan RM, 2012, Updates on the multimodality management of desmoplastic small round cell tumor. J Surg Oncol 105:617–621 Biswas G, Laskar S, Banavali SD, Gujral S, Kurkure PA, Muckaden M, Parikh PM, Nair CN, 2005, Desmoplastic small round cell tumour: extra abdominal and abdominal presentations and the results of treatment. Indian J Cancer 42:78–84 Hayes-Jordan A, Anderson PM, 2011, The diagnosis and management of desmoplastic small round cell tumor: a review. Curr Opin Oncol 23:385–389 Kushner BH, LaQuaglia MP, Wollner N, Meyers PA, Lindsley KL, Ghavimi F, Merchant TE, Boulad F, Cheung NK, Bonilla MA, Crouch G, Kelleher JF Jr, Steinherz PG, Gerald WL, 1996, Desmoplastic small round cell tumor: prolonged progression-free survival with aggressive multimodality therapy. J Clin Oncol 14:1515–1531 Bertuzzi A, Castagna L, Nozza A, Quagliuolo V, Siracusano L, Balzarotti M, Compasso S, Alloisio M, Soto Parra H, Santoro A, 2002, High-dose chemotherapy in poor-prognosis adult small round cell tumours: clinical and molecular results from a prospective study. J Clin Oncol 20:2181–2188 Schwarz RE, Gerald WL, Kushner BH, Coit DG, Brennan MF, La Quaglia MP, 1998, Desmoplastic small round cell tumors: prognostic indicators and results of surgical management. Ann Surg Oncol 5:416–422 Ordonez NG, 1998, Desmoplastic small round cell tumor: I: a histopathologic study of 39 cases with emphasis on unusual histological patterns. Am J Surg Pathol 22:1303–1313 Watanabe T, Miyamoto S, Kitagori K, Horimatsu T, Morita S, Mashimo Y, Ezoe Y, Muto M, Chiba T, 2012, A case of long-term survival of metastatic desmoplastic small round cell tumor treated with multimodal therapy. Oncol Lett 3:30–34 Quaglia MP, Brennan MF, 2000, The clinical approach to desmoplastic small round cell tumor. Surg Oncol 9:77–81 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 161 EP 168 DI 10.1007/s12253-013-9679-0 PG 8 ER PT J AU Maraz, R Boross, G Pap-Szekeres, J Rajtar, M Ambrozay, Cserni, G AF Maraz, Robert Boross, Gabor Pap-Szekeres, Jozsef Rajtar, Maria Ambrozay, Eva Cserni, Gabor TI Internal Mammary Sentinel Node Biopsy in Breast Cancer. Is it Indicated? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Internalmammary nodes; Sentinel node biopsy ID Breast cancer; Internalmammary nodes; Sentinel node biopsy AB Axillary sentinel node (A-SN) biopsy is a standard procedure in breast cancer surgery. Sampling of intenal mammary sentinel nodes (IM-SN) is not performed routinly, although it is also considered an important prognostic factor of breast cancer. The role of this latter procedure was investigated in cases of IM-SN visualized on lymphoscintigraphy. Between January 2001 and June 2012 1542 patients with clinically node negative operable primary breast cancer had sentinel node biopsy (SNB). Both axillary and IM-SN were sampled (whenever detected), based on lymphoscintigraphy, intraoperative gamma probe detection and blu dye mapping. Lymphoscintigraphy showed IM-SN in 83 cases. IM-SN biopsy (IM-SNB) was succesfull in 77 patients (93 %). A total of 86 IM-SNs were removed. IM-SN involvement was identified in 14 cases, representing 18 % of patients who underwent IM-SNB. This included macrometastases (MAC) in 5 cases, micrometastases (MIC) in 2 cases, isolated tumor cells (ITC) in 7 cases. No significant differences were found between patients with and without IM-SN involvement in terms of age, tumor location, tumor size, axillary involvement, tumor grade or estrogen receptor status. The IM-SN involvement has lead to new therapeutic indications in 2 cases (2.6 %), both of them due to MAC in the IM-SN: in 1 case change in chemotherapy and in 1 case change in radiotherapy, with the addition of iradiation of the internal mammary chain. Based on this series and information from the literature, we conclude that the indication for an IM-SNB procedure is very limited and its routine use should not be recommended. C1 [Maraz, Robert] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38, 6000 Kecskemet, Hungary. [Boross, Gabor] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38, 6000 Kecskemet, Hungary. [Pap-Szekeres, Jozsef] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38, 6000 Kecskemet, Hungary. [Rajtar, Maria] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. [Ambrozay, Eva] Bacs-Kiskun County Teaching Hospital, Breast Diagnostic UnitKecskemet, Hungary. [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of PathologyKecskemet, Hungary. RP Maraz, R (reprint author), Bacs-Kiskun County Hospital, Department of Surgery, 6000 Kecskemet, Hungary. EM marazrobert2010@gmail.com CR Krag D,Weaver D, Ashikaga Tet al, 1998, The sentinel node in breast cancer—a multicenter validation study. N Engl J Med 339:941–946 Giuliano AE, Jones RC, Brennan M, Statman R, 1997, Sentinel lymphadenectomy in breast cancer. J Clin Oncol 15:2345–2350 Veronesi U, Paganelli G, Galimberti V et al, 1997, Sentinel-node biopsy to avoid axillary dissection in breast cancer with clinically negative lymph-nodes. Lancet 349:1864–1867 Uren RF, Howman-Giles RB, Thompson JF et al, 1995, Mammary lymphoscintigraphy in breast cancer. J Nucl Med 36:1775–1780 Lamonica D, Edge SB, Hurd T et al, 2003, Mammographic and clinical predictors of drainage patterns in breast lymphoscintigrams obtained during sentinel node procedures. Clin NuclMed 28:558–564 Van der Ent FW, Kengen RA, van der Pol HA et al, 2001, Halsted revisited: internal mammary sentinel lymph node biopsy in breast cancer. Ann Surg 234:79–84 Estourgie SH, Nieweg OE, Valdes Olmos RA et al, 2004, Lymphatic drainage patterns from the breast. Ann Surg 239:232–237 Estourgie SH, Tanis PJ, Nieweg OE et al, 2003, Should the hunt for internal mammary chain sentinel nodes begin? An evaluation of 150 breast cancer patients. Ann Surg Oncol 10:935–941 Cserni G, Pap Szekeres J, 2001, Internal mammary lymph nodes and sentinel node biopsy in breast cancer. Surg Oncol 10:25–33 Veronesi U, Cascinelli N, Bufalino R et al, 1983, Risk of internal mammary lymph node metastases and its relevance on prognosis of breast cancer patients. Ann Surg 198:681 Kuenen-Boumeester V, Menke-Pluymers M, de Kanter AY et al, 2003, Ultrasound-guided fine needle aspiration cytology of axillary lymph nodes in breast cancer patients. A preoperative staging procedure. Eur J Cancer 39:170–174 Sapino A, Cassoni P, Zanon E et al, 2003, Ultrasonographicallyguided fine-needle aspiration of axillary lymph nodes: role in breast cancer management. Br J Cancer 88:702–706 Ambrozay E, Cserni G, Serenyi P et al, 2004, The role of axillary nodal staging during preoperative breast diagnostics. Eur J Cancer Suppl 2:92–93 Gennari R, Galimberti V, De Cicco C et al, 2000, Use of technetium- 99m-labeled colloid albumin for preoperative and intraoperative localization of nonpalpable breast lesions. J Am Coll Surg 1 90:692–699 Cserni G, 2002, Complete sectioning of axillary sentinel nodes in patients with breast cancer. Analysis of two different step sectioning and immunohistochemistry protocols in 246 patients. J Clin Pathol 55:926–931 Greene FL, Page DL, Fleming ID et al, eds,, 2002, AJCC Cancer Staging Hanbook New York. Springer, Berlin, p 469 Hultborn KA, Larsson LG, Ragnhult I, 1955, The lymph drainage from the breast to the axillary and parasternal lymph nodes, studied with the aid of colloidal AU198. Acta Radiol 43:52–64 Turner-Warwick RT, 1959, The lymphatics of the breast. Br J Surg 46:574–582 Heuts EM, van der Ent FW, Hulsewe KWE, von Meyenfeldt MF, Voogd EM, 2009, Internal mammary lymph drainage and sentinel node biopsy in breast cancer- A study on 1008 patients. Eur J Cancer Surgery 35:252–257 Nathan J, Boyages J, French JR, Ung OA, 2008, Internal mammary sentinel nodes: ignore, irradiate or operate? Eur J Cancer 45:789–794 Veronesi U, Cascinelli N, Greco M et al, 1985, Prognosis of breast cancer patients after mastectomy and dissection of internal mammary nodes. Ann Surg 202(6):702–707 Madsen E, Gobardhan PD, Bongers V et al, 2007, The impact on post-surgical treatment of sentinel lymph node biopsy of internal mammary lymph nodes in patients with breast cancer. Ann Surg Oncol 14:1486–1492 Roumen RMH, Geuskens LM, Valkenburg JGH et al, 1999, In search of the true sentinel node by different injection techniques in breast cancer patients. Eur J Surg Oncol 25:347–351 Borgstein PJ, Meijer S, Pijpers RJ, van Diest PJ, 2000, Functional lymphatic anatomy for sentinel node biopsy in breast cancer: echoes from the past and the periareolar blue method. Ann Surg 1(81–89) Spillane AJ, Noushi F, Cooper RA, Gebski V, Uren RF, 2009, Highresolution lymphoscintigraphy is essential for recognition of the significance of internal mammary nodes in breast cancer. Ann Oncol 20:977–984 Sugg SL, Ferguson DJ, Posner MC, Heimann R, 2000, Should internal mammary nodes be sampled in the sentinel lymph node era? Ann Surg Oncical Oncol 7:188–192 Van der Ent FW, Kengen RA, van der Pol HA, Povel JA, Stroeken HJ, Hoofwijk AG, 2001, Halsted revisited: internal mammary sentinel lymph node biopsy in breast cancer. Ann Surg 234(1):79–84 Overgaard M, Jensen MB, Overgaard J et al, 1999, Postoperative radiotherapy in high-risk postmenopausal breast-cancer patients given adjuvant tamoxifen: Danisch Breast Cancer Cooperative Group DBCG 82c randomised trial. Lancet 353:1641–1648 Ragaz J, Jackson SM, Le N et al, 1997, Adjuvant radiotherapy and chemotherapy in node-positive premenopausal women with breast cancer. N Engl J Med 337:956–962 Whelan TJ, Julian J,Wright J et al, 2000)Does locoregional radiation therapy improve survival in breast cancer? A meta-analysis. J Clin Oncol 18:1220–1229 Obedian E, Hafty BG, 1999, Internal mammary nodal irradiation in conservatively managed breast cancer patients: is there a benefit? Int J Radiat Oncol Biol Phys 44(5):997–1003 Freedman GM, Fowble BL, Nicolaou N et al, 2000, Should internal mammary lymph nodes in breast cancer be the target for the radiation oncologist? Int J Radiat Oncol Biol Phys 46:805–814 Jatoi I, 2003, Internal mammary sentinel nodes in primary breast cancer. Curr Med Res Opin 19:567–569 Fowble B, Hanlon A, Freedman G et al, 2000, Internal mammary node irradiation neither decreases distantmetastases nor improves survival in stage I and II breast cancer. Int J Radiat Oncol Biol Phys 47:883–894 Matzinger O, Heimsoth I, Poortmans P et al, 2010, Toxicity at three years with and without irradiation of the internal mammary and medial supraclavicular lymph node chain in stage I to III breast cancer, EORTC trial 22922/10925, Act. Oncol 49:24–34 ClarkeM, Collins R, Darby S et al, 2005, Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: an overview of the randomised trials. Lancet 366(9503):2087–2106 Early Breast Cancer Trialists’ Collaborative Group, EBCTGC,, 2005, Effects of chemotherapy and hormonal therapy for early breast cancer: effects on recurrence and 15-year survival: an overview of the randomised trials. Lancet 365:1687–1717 Dutch national guideline for the treatment of breast cancer, version 2.0, Feb 2012). http://www.oncoline.nl/mammacarcinoom. Accessed 22 Feb 2012 Coombs NJ, Boyages J, French JR et al, 2009, Internal mammary sentinel nodes: ignore, irradiate or operate? Eur J Cancer 45:789–794 Heuts EM, van der Ent FW, HulseweKWE, vonMeyenfeldtMF, Voogd EM, 2009, Results of tailored treatment for breast cancer patients with internal mammary lymph node metastases. The Breast 18:254–258 Postma EL, van Wieringen S, Hobbelink MG et al, 2012, Sentinel lymph node biopsy of the internal mammary chain in breast cancer. Breast Cancer Res Treat 134:735–774 Leidenius MHK, Krogerud LA, Toivonen TS et al, 2006, The clinical value of parasternal sentinel node biopsy in breast cancer. Ann Surg Oncol 13(3):321–326 Cserni G, 2007, What is a positive sentinel lymph node in a breast cancer patient? A practical approach. Breast 16:152–160 Lazar G, Besznyak I, Boross G et al, 2010, Az emlorak korszeru sebeszi kezelese. M Onkol 54:227–236 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 169 EP 177 DI 10.1007/s12253-013-9680-7 PG 9 ER PT J AU Feng, Qj Zhang, F Huang, Xy Wu, Zx AF Feng, Qing-jing Zhang, Feng Huang, Xiao-yun Wu, Zhi-xiang TI Effectiveness and Complications of Anthracycline and Taxane in the Therapy of Breast Cancer: A Meta-analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Meta-analysis; Breast cancer; Anthracycline; Taxane ID Meta-analysis; Breast cancer; Anthracycline; Taxane AB Objective: To compare the efficacy and safety between anthracycline & taxane and anthracycline in the treatment of breast cancer. Methods: Computer-assisted literature search was performed with PubMed, MEDLINE, EMBASE and Cochrane Controlled Trials Register (CCTR) to identify pertinent literatures. Software RevMan 5.0 was used for statistical analysis. The measurement of interest outcomes included severe neurotoxicity, death without breast cancer recurrences, leukemia, venous thrombus and severe cardiotoxicity. Results: A total of 10 randomized controlled trial studies (RCTs) containing 18,198 cases were selected in this metaanalysis. Of which, 9,902 cases were treated with anthracycline & taxane and 8,296 cases treated with anthracycline alone as control. Anthracycline& taxane showed lower risks of incident leukemia (RR=0.40; 95 % CI: 0.18, 0.90), venous thrombus (RR=0.49; 95 % CI: 0.29, 0.84) and severe cardiotoxicity (RR=0.41, 95%CI: 0.26, 0.66), but higher risks of incident severe neurotoxicity (RR=5.97; 95 % CI: 1.72, 20.65) and non-recurrent death (RR=1.79; 95%CI: 1.06, 3.04), compared to anthracycline alone. Conclusion: Clinically important differences exist for general safety in favour of the anthracycline & taxane rather than anthracycline alone. However, as a result of tumor recurrent rate, anthracycline might be superior to anthracycline & taxane. A longer duration of follow-up and a larger number of cases are required to better assess the efficacy and safety profile of the treatment of breast cancer. C1 [Feng, Qing-jing] Yiwu Maternity and Child Care Hospital, Department of Breast Surgery, 322000 Yiwu, China. [Zhang, Feng] School of Medicine, Zhejiang University, Women’s Hospital, Department of Surgery, No. 1 Xueshi Road, 310006 Hangzhou, China. [Huang, Xiao-yun] Yiwu Maternity and Child Care Hospital, Department of Breast Surgery, 322000 Yiwu, China. [Wu, Zhi-xiang] Shanghai Jiao Tong University, The International Peace Maternity and Child Health Hospital of China welfare Institute, Department of Breast Surgery, 200030 Shanghai, China. RP Zhang, F (reprint author), School of Medicine, Zhejiang University, Women’s Hospital, Department of Surgery, 310006 Hangzhou, China. EM zhang3772@gmail.com CR Jemal A, Siegel R,Ward E,Murray T, Xu J, Thun MJ, 2007, Cancer statistics, 2007. CA Cancer J Clin 57(1):43–66 Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF, 2003, Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci 100(7):3983–3988 De Laurentiis M, Cancello G, D’Agostino D, Giuliano M, Giordano A, Montagna E, Lauria R, Forestieri V, Esposito A, Silvestro L, 2008, Taxane-based combinations as adjuvant chemotherapy of early breast cancer: a meta-analysis of randomized trials. J Clin Oncol 26(1):44–53 Kaufmann M, Hortobagyi GN, Goldhirsch A, Scholl S, Makris A, Valagussa P, Blohmer JU, Eiermann W, Jackesz R, Jonat W, Lebeau A, Loibl S, MillerW, Seeber S, Semiglazov V, Smith R, Souchon R, Stearns V, Untch M, von Minckwitz G, 2006, Recommendations from an international expert panel on the use of neoadjuvant, primary, systemic treatment of operable breast cancer: an update. J Clin Oncol 24(12):1940–1949., DOI 10.1200/JCO.2005.02.6187 Kaufmann M, von Minckwitz G, Rody A, 2005, Preoperative, neoadjuvant, systemic treatment of breast cancer. Breast 14(6):576–581., DOI 10.1016/j.breast.2005.08.010 Jarvinen TA, Tanner M, Rantanen V, Barlund M, Borg A, Grenman S, Isola J, 2000, Amplification and deletion of topoisomerase IIalpha associate with ErbB-2 amplification and affect sensitivity to topoisomerase II inhibitor doxorubicin in breast cancer. Am J Pathol 156(3):839–847 Burgess DJ, Doles J, Zender L, Xue W, Ma B, McCombie WR, Hannon GJ, Lowe SW, Hemann MT, 2008, Topoisomerase levels determine chemotherapy response in vitro and in vivo. Proc Natl Acad Sci U S A 105(26):9053–9058., DOI 10.1073/pnas.0803513105 Trudeau M, Charbonneau F, Gelmon K, Laing K, Latreille J, Mackey J, McLeod D, Pritchard K, Provencher L, Verma S, 2005, Selection of adjuvant chemotherapy for treatment of node-positive breast cancer. Lancet Oncol 6(11):886–898., DOI 10.1016/S1470-2045(05)70424-1 Miglietta A, Bocca C, Gabriel L, 2002, Comparative studies on biological activity of certain microtubule-interacting taxanes. Chem Biol Interact 139(3):283–299 Fromes Y, Gounon P, Veitia R, Bissery MC, Fellous A, 1996, Influence of microtubule-associated proteins on the differential effects of paclitaxel and docetaxel. J Protein Chem 15(4):377–388 Jones SE, Savin MA, Holmes FA, O’Shaughnessy JA, Blum JL, Vukelja S, McIntyre KJ, Pippen JE, Bordelon JH, Kirby R, Sandbach J, Hyman WJ, Khandelwal P, Negron AG, Richards DA, Anthony SP, Mennel RG, Boehm KA, Meyer WG, Asmar L, 2006, Phase III trial comparing doxorubicin plus cyclophosphamide with docetaxel plus cyclophosphamide as adjuvant therapy for operable breast cancer. J Clin Oncol 24(34):5381–5387., DOI 10.1200/JCO.2006.06.5391 Bria E, Giannarelli D, Felici A, Peters WP, Nistico C, Vanni B, Cuppone F, Cognetti F, Terzoli E, 2005, Taxanes with anthracyclines as first-line chemotherapy for metastatic breast carcinoma. Cancer 103(4):672–679., DOI 10.1002/cncr.20757 Bria E, Nistico C, Cuppone F, Carlini P, CiccareseM, MilellaM, Natoli G, Terzoli E, Cognetti F, Giannarelli D, 2006, Benefit of taxanes as adjuvant chemotherapy for early breast cancer: pooled analysis of 15,500 patients. Cancer 106(11):2337–2344., DOI 10.1002/cncr.21886 De Laurentiis M, Cancello G, D’Agostino D, Giuliano M, Giordano A, Montagna E, Lauria R, Forestieri V, Esposito A, Silvestro L, Pennacchio R, Criscitiello C, Montanino A, Limite G, Bianco AR, De Placido S, 2008, Taxane-based combinations as adjuvant chemotherapy of early breast cancer: a meta-analysis of randomized trials. J Clin Oncol 26(1):44–53., DOI 10.1200/JCO.2007.11.3787 Boccardo F, Amadori D, Guglielmini P, Sismondi P, Farris A, Agostara B, Gambi A, Catalano G, Faedi M, Rubagotti A, 2010, Epirubicin followed by cyclophosphamide, methotrexate and 5- fluorouracil versus paclitaxel followed by epirubicin and vinorelbine in patients with high-risk operable breast cancer. Oncology 78(3– 4):274–281., DOI 10.1159/000315735 Burnell M, Levine MN, Chapman JA, Bramwell V, Gelmon K, Walley B, Vandenberg T, Chalchal H, Albain KS, Perez EA, Rugo H, Pritchard K, O’Brien P, Shepherd LE, 2010, Cyclophosphamide, epirubicin, and Fluorouracil versus dose-dense epirubicin and cyclophosphamide followed by Paclitaxel versus Doxorubicin and cyclophosphamide followed by Paclitaxel in node-positive or high-risk node-negative breast cancer. J Clin Oncol 28(1):77–82., DOI 10.1200/ JCO.2009.22.1077 Ellis P, Barrett-Lee P, Johnson L, Cameron D,Wardley A,O’Reilly S, VerrillM, Smith I, Yarnold J, Coleman R, Earl H, Canney P, Twelves C, Poole C, Bloomfield D, Hopwood P, Johnston S, Dowsett M, Bartlett JM, Ellis I, Peckitt C, Hall E, Bliss JM, 2009, Sequential docetaxel as adjuvant chemotherapy for early breast cancer, TACT): an open-label, phase III, randomised controlled trial. Lancet 373(9676):1681–1692., DOI 10.1016/S0140-6736(09)60740-6 Francis P, Crown J, Di Leo A, Buyse M, Balil A, Andersson M, Nordenskjold B, Lang I, Jakesz R,Vorobiof D,Gutierrez J, van Hazel G, Dolci S, Jamin S, Bendahmane B, Gelber RD, Goldhirsch A, Castiglione-Gertsch M, Piccart-Gebhart M, 2008, Adjuvant chemotherapy with sequential or concurrent anthracycline and docetaxel: Breast InternationalGroup 02–98 randomized trial. JNatl Cancer Inst 100(2):121–133., DOI 10.1093/jnci/djm287 Gianni L, Baselga J, EiermannW, Porta VG, Semiglazov V, Lluch A, Zambetti M, Sabadell D, Raab G, Cussac AL, Bozhok A, Martinez- Agullo A, GrecoM, Byakhov M, Lopez JJ,MansuttiM, Valagussa P, Bonadonna G, 2009, Phase III trial evaluating the addition of paclitaxel to doxorubicin followed by cyclophosphamide, methotrexate, and fluorouracil, as adjuvant or primary systemic therapy: European Cooperative Trial in Operable Breast Cancer. J Clin Oncol 27(15):2474–2481., DOI 10.1200/JCO.2008.19.2567 Goldstein LJ,O’Neill A, Sparano JA, PerezEA, ShulmanLN,Martino S, Davidson NE, 2008, Concurrent doxorubicin plus docetaxel is not more effective than concurrent doxorubicin plus cyclophosphamide in operable breast cancer with 0 to 3 positive axillary nodes: North American Breast Cancer Intergroup Trial E 2197. J Clin Oncol 26(25):4092–4099., DOI 10.1200/JCO.2008.16.7841 Martin M, Rodriguez-Lescure A, Ruiz A, Alba E, Calvo L, Ruiz- Borrego M, Munarriz B, Rodriguez CA, Crespo C, de Alava E, Lopez Garcia-Asenjo JA, Guitian MD, Almenar S, Gonzalez-Palacios JF, Vera F, Palacios J, Ramos M, Gracia Marco JM, Lluch A, Alvarez I, SeguiMA, Mayordomo JI, Anton A, Baena JM, Plazaola A,Modolell A, Pelegri A,Mel JR, Aranda E, Adrover E, Alvarez JV, Garcia Puche JL, Sanchez-Rovira P, Gonzalez S, Lopez-Vega JM, 2008, Randomized phase 3 trial of fluorouracil, epirubicin, and cyclophosphamide alone or followed by Paclitaxel for early breast cancer. J Natl Cancer Inst 100(11):805–814., DOI 10.1093/jnci/djn151 Martin M, Segui MA, Anton A, Ruiz A, Ramos M, Adrover E, Aranda I, Rodriguez-Lescure A, Grosse R, Calvo L, Barnadas A, Isla D, Martinez del Prado P, Ruiz Borrego M, Zaluski J, Arcusa A, Munoz M, Lopez Vega JM, Mel JR, Munarriz B, Llorca C, Jara C, Alba E, Florian J, Li J, Lopez Garcia-Asenjo JA, Saez A, Rios MJ, Almenar S, Peiro G, Lluch A, 2010, Adjuvant docetaxel for highrisk, node-negative breast cancer. N Engl JMed 363(23):2200–2210., DOI 10.1056/NEJMoa0910320 Polyzos A, Malamos N, Boukovinas I, Adamou A, Ziras N, Kalbakis K, Kakolyris S, Syrigos K, Papakotoulas P, Kouroussis C, Karvounis N, Vamvakas L, Christophyllakis C, Athanasiadis A, Varthalitis I, Georgoulias V, Mavroudis D, 2010, FEC versus sequential docetaxel followed by epirubicin/cyclophosphamide as adjuvant chemotherapy in women with axillary node-positive early breast cancer: a randomized study of the Hellenic Oncology Research Group, HORG). Breast Cancer Res Treat 119(1):95–104., DOI 10.1007/s10549-009-0468-0 Roche H, Fumoleau P, SpielmannM, Canon JL, Delozier T, Serin D, Symann M, Kerbrat P, Soulie P, Eichler F, Viens P, Monnier A, Vindevoghel A, Campone M, Goudier MJ, Bonneterre J, Ferrero JM, Martin AL, Geneve J, Asselain B, 2006, Sequential adjuvant epirubicin-based and docetaxel chemotherapy for node-positive breast cancer patients: the FNCLCC PACS 01 Trial. J Clin Oncol 24(36):5664–5671., DOI 10.1200/JCO.2006.07.3916 Michaud LB, 2008, Treatment-experienced breast cancer. Am J Health Syst Pharm 65(10 Supplement 3):S4–S9 Paik S, Tang G, Shak S, Kim C, Baker J, KimW, CroninM, Baehner FL, Watson D, Bryant J, 2006, Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor–positive breast cancer. J Clin Oncol 24(23):3726–3734 Lee CW, Chi KN, 2000, The standard of reporting of health-related quality of life in clinical cancer trials. J Clin Epidemiol 53(5):451–458 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 179 EP 184 DI 10.1007/s12253-013-9681-6 PG 6 ER PT J AU Rehders, A Anlauf, M Adamowsky, I Ghadimi, HM Klein, S Antke, Ch Cupisti, K Stoecklein, HN Knoefel, TW AF Rehders, Alexander Anlauf, Martin Adamowsky, Ilona Ghadimi, H Markus Klein, Sarah Antke, Christina Cupisti, Kenko Stoecklein, H Nikolas Knoefel, T Wolfram TI Is Minimal Residual Lymph Node Disease in Papillary Thyroid Cancer of Prognostic Impact? An Analysis of The Epithelial Cell Adhesion Molecule EpCAM in Lymph Nodes of 40 pN0 Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary thyroid cancer; Lymphadenectomy; Radio iodine treatment; Tumor cell dissemination; Immunohistochemistry ID Papillary thyroid cancer; Lymphadenectomy; Radio iodine treatment; Tumor cell dissemination; Immunohistochemistry AB This study was aimed to assess the extend of nodal microdissemination in patients with pN0 papillary thyroid carcinoma (PTC) using immunohistochemical analysis. In early stage PTC both, systematic lymphadenectomy as well as radio iodine treatment, aimed to eliminate occult nodal tumor involvement, are under controversial debate, since little is known about the extend of lymphatic microdissemination in these patients. Formalin embedded samples of the resected lymph nodes were systematically screened for the presence of disseminated tumor cells using immunohistochemistry (monoclonal antibody Ber-EP4). Clinical and histopathological parameters as well as the post-operative course were recorded. Survival data were analysed by the Kaplan-Meier method and the log rank test. Overall 321 lymph nodes of 40 patients were screened immunohistochemically. In 12.5 % of the patients disseminated occult tumor cells were diagnosed. In addition to tumor resection 90 % of the patients underwent adjuvant radio-iodine treatment. The mean observation period in our collective was 72 months. The detection of disseminated tumor cells did not correlate with clinicopathologic risk parameters and did not have significant influence on the prognosis of these patients. Immunohistochemical analysis enables the detection of disseminated tumor cells in patients with pN0 PTC. This finding seems to support the application of adjuvant radio iodine, even in early tumor stages. C1 [Rehders, Alexander] Heinrich Heine University, Department of SurgeryDusseldorf, Germany. [Anlauf, Martin] Heinrich Heine University, Institute of PathologyDusseldorf, Germany. [Adamowsky, Ilona] Heinrich Heine University, Department of SurgeryDusseldorf, Germany. [Ghadimi, H Markus] Heinrich Heine University, Department of SurgeryDusseldorf, Germany. [Klein, Sarah] Heinrich Heine University, Department of SurgeryDusseldorf, Germany. [Antke, Christina] Heinrich Heine University, Department of Nuclear MedicineDusseldorf, Germany. [Cupisti, Kenko] Heinrich Heine University, Department of SurgeryDusseldorf, Germany. [Stoecklein, H Nikolas] Heinrich Heine University, Department of SurgeryDusseldorf, Germany. [Knoefel, T Wolfram] Heinrich Heine University, Department of SurgeryDusseldorf, Germany. CR DeGroot L, Kaplan E, McCormick M et al, 1990, Natural history, treatment, and course of papillary thyroid carcinoma. J Clin Endocrinol Metab 71:414–424 Reynolds RM, Weir J, Stockton DL et al, 2005, Changing trends in incidence and mortality of thyroid cancer in Scotland. Clin Endocrinol 62:152–162 ItoY,MiyauchiA, Jikuzono Tet al, 2007, Risk factors contributing to a poor prognosis of papillary thyroid carcinoma; validity of UICC/AJCC TNM classification and stage grouping. World J Surg 31:838–848 Noguchi S, Murakami N, Yamashita H et al, 1998, Papillary thyroid carcinoma: modified radical neck dissection improves prognosis. Arch Surg 133:276–280 Cranshaw IM, Carnaille B, 2008)Micrometastases in thyroid cancer. An important finding? Surg Oncol 17:253–258 Borson-Chazot F, Causeret S, Lifante JC et al, 2004, Predictive factors for recurrence of from a series of 74 children and adolescents with differentiated thyroid cancer. World J Surg 28:1088–1092 Cooper DS, Doherty GM, Haigen BR et al, 2006, Management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid 16:109–142 Wada N, Duh QY, Sugino K et al, 2003, Lymph node metastasis from 259 papillary thyroid microcarcinomas. Ann Surg 237:399–407 Ito Y, Tomoda C, Uruno T et al, 2006, Clinical significance of metastasis to the central compartment frompapillary microcarcinoma of the thyroid. World J Surg 30:91–99 Lundgren CI, Hall P, Dickman PWet al, 2006, Clinically significant prognostic factors for differentiated thyroid carcinoma: a populationbased, nested case control study. Cancer 106:524–531 Sawka AM, Brierley JD, Tsang RWet al, 2008, An updated review and commentary examining the effectiveness of radioactive iodine remnant ablation in well differentiated thyroid cancer. Endocrinol Metab Clin North Am 37:457–480 Izbicki JR, Hosch SB, Pichlmeier U et al, 1997, Prognostic value of immunohistochemically identifiable tumor cells in lymph nodes of patients with completely resected esophageal cancer. N Engl J Med 337:1188–1194 Rosenberg R, Friedrichs J, Gertler R et al, 2004, Prognostic evaluation and review of immunohistochemically detected disseminated tumor cells in peritumoral lymph nodes of patients with pN0 colorectal cancer. Int J Colorectal Dis 19:430–437 Kurahara H, Takao S, Maemura K et al, 2007, Impact of lymph node micrometastasis in patients with pancreatic head cancer.World J Surg 31:483–490 Hermanek P, Hutter RV, Sobin LH et al, 1999, International Union Against Cancer. Classification of isolated tumor cells and micrometastasis. Cancer 86:2668–2673 Sobin LH, Wittekind C, eds,, 1997, International Union Against Cancer, UICC). TNM classification of malignant tumors, 5th edn. Wiley, New York De Klerk JMH, De Keizer B, Zelissen PMJ et al, 2000, Fixed dosage of 131I for remnant ablation with differentiated thyroid carcinoma without pre-ablative diagnostic 131 scintigraphy. Nucl Med Commun 21:529–532 Scheunemann P, Stoecklein NH, Rehders A et al, 2007, Frequency and prognostic significance of occult tumor cells in lymph nodes in patients with adenocarcinoma of the papilla of Vater. HPB, Oxford, 9:135–139 Latza U, Niedobitek G, Schwarting R et al, 1990, Ber EP4: new monoclonal antibody,which distinguishes epithelia frommesothelial. J Clin Pathol 43:213–219 Stoecklein NH, Siegmund A, Scheunemann P et al, 2006, Ep-CAM expression in squamous cell carcinoma of the esophagus: a potential therapeutic target and prognostic marker. BMC Cancer 6:135–143 Hsu SM, Raine L, 1981, Protein A, avidin and biotin in immunohistochemistry. J Histochem Cytochem 29:1349–1353 Kaplan EL, Meier P, 1958, Nonparametric estimation from incomplete observations. J Am Stat Assoc 53:457–481 Toniato A, Boschin I, Casara D et al, 2008, Papillary thytoid carcinoma: factors influencing recurrence and survival. Ann Surg Oncol 15:1518–1522 Cappelli C, Castellano M, Braga M et al, 2007, Aggressiveness and outcome of papillary thyroid carcinoma, PTC, versus microcarcinoma, PMC): a mono-institutional experience. J Surg Oncol 95:555–560 Ito Y, Higashiyama T, Takamura Yet al, 2007, Risk factors for recurrence to the lymph node in papillary thyroid carcinoma patients without preoperatively detectable lateral node metastasis: validity of prophylactic modified radical neck dissection. World J Surg 31:2085–2091 Lo CY, Chan WF, Lam KYet al, 2004, Optimizing the treatment of AMES high-risk papillary thyroid carcinoma.World J Surg 28:1103– 1109 Monchik JM, Simon CJ, Caragacianu DL et al, 2009, Does failure to performprophylactic levelVI node dissection leave persistent disease detectable by ultrasonography in patients with low-risk papillary carcinoma of the thyroid? Surgery 146:1182–1187 Rosenbaum MA, McHenry CR, 2009, Central neck dissection for papillary thyroid cancer. Arch Otolaryngol Head Neck Surg 135:1092– 1097 Mazzaferri EL, Doherty GM, Steward DL, 2009, The pros and cons of prophylactic central compartment lymph node dissection for papillary thyroid carcinoma. Thyroid 19:683–689 Hughes CJ, Shaha AR, Shah JP et al, 1996, Impact of lymph node metastasis in differentiated carcinoma of the thyroid; a matched pair analysis. Head Neck 18:127–132 Cady B, Sedgwick CE, MeissnerWAet al, 1979, Risk factor analysis in differentiated thyroid cancer. Cancer 43:810–820 Lugo TG, Braun S, Cote RJ et al, 2003, Detection and measurement of occult disease for the prognosis of solid tumors. J Clin Oncol 21:2609– 2615 Qubain SW, Nakano S, Baba M et al, 2002, Distribution of lymph node micrometastasis in pN0 well-differentiated thyroid carcinoma. Surgery 131:249–256 Bardet S, Malville E, Rame JP et al, 2008, The pros and cons of prophylactic central compartment lymph node dissection for papillary thyroid carcinoma. Eur J Endocrinol 158:551–560 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 185 EP 190 DI 10.1007/s12253-013-9682-5 PG 6 ER PT J AU Samulak, D Grosman-Dziewiszek, P Michalska, MM Mojs, E Samulak, K Romanowicz, H Smolarz, B AF Samulak, Dariusz Grosman-Dziewiszek, Patrycja Michalska, M Magdalena Mojs, Ewa Samulak, Katarzyna Romanowicz, Hanna Smolarz, Beata TI Evaluation of Expression of the PTEN Gene, Oestrogen and Progesterone Receptors as Diagnostic and Predictive Factors in Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial cancer; Oestrogen and progesterone receptors; PTEN gene; Predictive factor ID Endometrial cancer; Oestrogen and progesterone receptors; PTEN gene; Predictive factor AB Endometrial cancer belongs to the commonest malignancy in females after breast cancer,malignant neoplasm of female genitals in Europe and North America but there is still not significant improvement as far as the curability of this neoplasm is concerned, especially its advanced forms. That is why there is need to define new factors that could be not only diagnostic but also predictve factors. In present study we analyzed the mRNA PTEN expression by quantitative realtime polymerase chain reaction (Q-PCR) in 123 women of endometrial carcinoma and 14 women of control group. Moreover we assessed oestrogen (ER) and progesterone receptors (PgR) in all cases.We defined the correlation between expression of PTEN gene and receptors and between PTEN expression and maturity grade of cancer. Neoplasm advancement grade G1was diagnosed in 82.11%of patients (n =101), G2 in 9.76 % of patients (n =12) and G3 in 8.13 % of patients (n =10). Presence of ER and PgR and decreased expression of PTEN gene was found in majority of patients with endometrial cancer (79.12 % and 59.34 % respectively) and the most numerous group was with weak expression of ER and strong expression of PgR. There was no statistically significant difference in gene expression depending on receptors expression nor maturity grade of cancer (p >0.05). Evaluation of expression of PTEN gene may turn out to be a very useful tool aimed at qualifying patients for different therapies of endometrial cancer and at searching of new diagnostic and therapeutic methods of this cancer independently on its receptor status nor maturity grade of cancer. C1 [Samulak, Dariusz] Poznan University of Medical Sciences, Cathedral of Mother’s and Child’s Health, ul. Polna 33, 60-535 Poznan, Poland. [Grosman-Dziewiszek, Patrycja] Wroclaw Medical University, Department of Human Morphology and Embryology, ul. Chalubinskiego 6a, 50-368 Wroclaw, Poland. [Michalska, M Magdalena] Regional Hospital in Kalisz, Department of Obstetrics and Gynaecology, ul. Torunska 7, 62-800 Kalisz, Poland. [Mojs, Ewa] Poznan University of Medical Sciences, Department of Clinical PsychologyPoznan, Poland. [Samulak, Katarzyna] University of Medical Sciences in Poznan, Student Scientific Society, ul. Polna 33, 60-535 Poznan, Poland. [Romanowicz, Hanna] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. [Smolarz, Beata] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. RP Smolarz, B (reprint author), Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, 93-338 Lodz, Poland. EM smolbea@wp.pl CR Ferlay J, Bray F, Pisani P, Parkin DM, 2004, GLOBOCAN, 2002. Cancer incidence, mortality and prevalence worldwide. IARC CancerBase. IARC Press, Lyon Jemal A,Murray T,Ward E, Samuels A, Tiwari RC, Ghafoor A et al, 2005, Cancer statistics 2005. CA Cancer J Clin 55:10–30 Bray F, Loos AH, Oostindier M, Weiderpass E, 2005, Geographic and temporal variations in cancer of the corpus uteri: incidence and mortality in pre- and postmenopausal women in Europe. Int J Cancer 117:123–131 Salvesen HB, Akslen LA, 2002, Molecular pathogenesis and prognostic factors in endometrial carcinoma. APMIS 110:673–689 Hayes MP, Wang H, Espinal-Witter R, Douglas W, Solomon GJ, Baker SJ, Ellenson LH, 2006, PIK3CA and PTEN mutations in uterine endometrioid carcinoma and complex atypical hyperplasia. Clin Cancer Res 12:5932–5935 Oda K, Stokoe D, Taketani Y, McCormick F, 2005, High frequency of coexistent mutations of PIK3CA and PTEN genes in endometrial carcinoma. Cancer Res 65:10669–10673 Blanco-Aparico C, Renner O, Leal J, Carnero A, 2007, PTEN, more than AKT pathway. Carcicnogenesis 28:1379–1386 Stemke-Hale K, Gonzalez-Angulo AM, Lluch A, Neve RM, Kuo WL, Davies M, Carey M, Hu Z, Guan Y, Sahin A, Symmans WF, Pusztai L, Nolden LK, Horlings H, BernsK, HungMC, van deVijver MJ, Valero V, Gray JW, Bernards R, Mills GB, Hennessy BT, 2008, An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res 68:6084–6091 Levine DA, Bogomolniy F, Yee CJ, Lash A, Barakat RR, Borgen PI, Boyd J, 2005, Frequentmutation of the PIK3CA gene in ovarian and breast cancers. Clin Cancer Res 11:2875–2878 Bansal N, Yendluri V,Wenham RM(2009, The molecular biology of endometrial cancers and the implications for pathogenesis, classification, and targeted therapies. Cancer Control 16:8–13 Mutter GL, Lin MC, Fitzerald JT, Kum JB, Baak JPA, Lees JA, Wenig LP, Eng C, 2000, Altered PTEN expression as a diagnostic marker for the earliest endometrial precancers. J Natl Cancer Inst 92: 924–931 Doll A, AbalM, RigauM, MongeM, GonzalezM, Demajo S, Colas E, Llaurado M, Alazzouzi H, Planaguma J, Lohmann MA, Garcia J, Castellvi S, Ramon y Cajal J, Gil-Moreno A, Xercavins J,Alameda F, Reventos J, 2008, Novel molecular profiles of endometria cancernew light through old windows. J Steroid Biochem Mol Biol 108: 221–229 Engelsen IB, Akslen L, Salvesen HB, 2009, Biologic markers in endometrial cancer treatment. APMIS 117:693–707 Piekarski J, 2005, Should we start routine assessment of expression of PTEN protein in patients with breast cancer? J Oncol 55:480–484 Maxwell GL, Risinger JI, Gumbs C, Shaw H, Bentley RC, Barrett JC, Berchuck A, Futreal PA et al, 1998, Mutation of the PTEN tumor suppressor gene in endometrial hyperplasias. Cancer Res 58:2500– 2503 Sarbassov D, Guertin DA, Ali SM, SabatinM(2005, Phosphorylation and regulation of AKT/PKB by the Rictor-mTOR Complex. Science 304:1098–1101 Samarnthai N, Hall K, Yeh IT, 2010, Molecular profiling of endometrial malignancies. Obstet Gynecol Int 2010:162363 Lax SF, 2007, Molecular genetic changes in epithelial, stromal and mixed neoplasms of the endometrium. Pathology 39:46–54 Lax SF, 2004, Molecular genetic pathways in various types of endometrial carcinoma: from a phenotypical to a molecularbased classification. Virchows Arch 444:213–223 Hecht JL, Mutter GL, 2006, Molecular and pathologic aspects of endometrial carcinogenesis. J Clin Oncol 24:4783–4791 Pilarski R, 2009, Cowden syndrome: a critical review of the clinical literature. J Genet Couns 18:13–27 Llobet D, Pallares J, Yeramian A, Santacana M, Eritja N, Velasco A, Dolcet X,Matias-Guiu X, 2009, Molecular pathology of endometrial carcinoma: practical aspects from the diagnostic and therapeutic viewpoints. J Clin Pathol 62:777–785 Matias-guiu X, Catasus L, Bussaglia E, LagardaH,GarciaA, Pons C, Munoz J, Arguelles R, Machin P, Prat J, 2001, Molecular pathology of endometrial hyperplasia and carcinoma. Hum Pathol 32:569–577 Prat J, Gallardo A, Cuatrecasas M, Catasus L, 2007, Endometrial carcinoma: pathology and genetics. Pathology 39:72–87 Tashiro H, Blazes MS, Wu R, Cho KR, Bose S, Wang SI, Li J, Parsons R, Ellenson LH, 1997, Mutations in PTEN are frequent in endometrial carcinoma but rare in other common gynecological malignancies. Cancer Res 57:3935–3940 Liu FS, 2007, Molecular carcinogenesis of endometrial cancer. Taiwan J Obstet Gynecol 46:26–32 Salvesen HB, MacDonald N, Ryan A, Jacobs IJ, Lynch ED, Akslen LA, Das S, 2001, PTEN methylation is associated with advanced stage and microsatellite instability in endometrial carcinoma. Int J Cancer 91:22–26 Latta E, Chapman WB, 2002, PTEN mutations and evolving concepts in endometrial neoplasia. Curr Opin Obstet Gynecol 14:59–65 Salvesen HB, Stefansson I, Kretzschmar EI, Gruber P, MacDonald ND, Ryan A, Jacobs IJ, Akslen LA, Das S, 2004, Significance of PTEN alterations in endometrial carcinoma: a population-based study of mutations, promoter methylation and PTEN protein expression. Int J Oncol 25:1615–1623 Mackay HJ, Gallinger S, Tsao MS, McLachlin CM, Tu D, Keiser K, Eisenhauer EA, Oza AM, 2010, Prognostic value of microsatellite instability, MSI, and PTEN expression in women with endometrial cancer: results from studies of the NCIC Clinical Trials Group, NCIC CTG). Eur J Cancer 46:1365–1373 Erkanli S, Kayaselcuk F, Kuscu E, Bagis T, Bolat F, Haberal A, Demirhan B, 2006, Expression of survivin, PTEN and p27 in normal, hyperplastic, and carcinomatous endometrium. Int J Gynecol Cancer 16:1412–1418 Inaba F, Kawamata H, Teramoto T, Fukasawa I, Inaba N, Fujimori T, 2005, PTEN and p53 abnormalities are indicative and predictive factors for endometrial carcinoma. Oncol Rep 13:17–24 Kagan J, Srivastava S, 2005, Mitochondria as a target for early detection and diagnosis of cancer. Crit Rev Clin Lab Sci 42:453–472 Sobczuk A, Smolarz B, Romanowicz-Makowska H, Pertynski T, 2006, MMAC/PTEN gene expression in endometrial cancer: RTPCR studies. Pol J Pathol 57:137–140 Bogusiewicz M, Semczuk A, Gogacz M, Skomra D, Jakowicki JA, Rechberger T, 2006, Lack of correlation between leptin receptor expression and PI3-K/Akt signaling pathway proteins immunostaining in endometrioid-type endometrial carcinomas. Cancer Lett 238: 61–68 Temkin SM, Fleming G, 2009, Current treatment of metastatic endometrial cancer. Cancer Control 16:38–45 Gadducci A, Tana R, Cosio S, Fanucchi A, Genazzani AR, 2008, Molecular target therapies in endometrial cancer: from the basic research to the clinic. Gynecol Endocrinol 24:239–249 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 191 EP 196 DI 10.1007/s12253-013-9684-3 PG 6 ER PT J AU Qian, L Luo, Q Zhao, X Huang, J AF Qian, Liqiang Luo, Qingquan Zhao, Xiaojing Huang, Jia TI Pathways Enrichment Analysis for Differentially Expressed Genes in Squamous Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Squamous lung cancer; Oncogenesis mechanism; Potential pathways; Target genes ID Squamous lung cancer; Oncogenesis mechanism; Potential pathways; Target genes AB Squamous lung cancer (SQLC) is a common type of lung cancer, but its oncogenesis mechanism is not so clear. The aim of this study was to screen the potential pathways changed in SQLC and elucidate the mechanism of it. Published microarray data of GSE3268 series was downloaded from Gene Expression Omnibus (GEO). Significance analysis of microarrays was performed using software R, and differentially expressed genes (DEGs) were harvested. The functions and pathways of DEGs were mapped in Gene Otology and KEGG pathway database, respectively. A total of 2961 genes were filtered as DEGs between normal and SQLC cells. Cell cycle and metabolism were the mainly changed functions of SQLC cells. Meanwhile genes such as MCM, RFC, FEN1, and POLD may induce SQLC through DNA replication pathway, and genes such as PTTG1, CCNB1, CDC6, and PCNA may be involved in SQLC through cell cycle pathway. It is demonstrated that pathway analysis is useful in the identification of target genes in SQLC. C1 [Qian, Liqiang] Shanghai JiaoTong University, Shanghai Chest Hospital, Shanghai Lung Cancer Center, NO.241 Huaihai Road, 200030 Shanghai, China. [Luo, Qingquan] Shanghai JiaoTong University, Shanghai Chest Hospital, Shanghai Lung Cancer Center, NO.241 Huaihai Road, 200030 Shanghai, China. [Zhao, Xiaojing] Shanghai JiaoTong University, Shanghai Chest Hospital, Shanghai Lung Cancer Center, NO.241 Huaihai Road, 200030 Shanghai, China. [Huang, Jia] Shanghai JiaoTong University, Shanghai Chest Hospital, Shanghai Lung Cancer Center, NO.241 Huaihai Road, 200030 Shanghai, China. RP Luo, Q (reprint author), Shanghai JiaoTong University, Shanghai Chest Hospital, Shanghai Lung Cancer Center, 200030 Shanghai, China. EM luoqingquanlqq@hotmail.com CR Tseng C-Y, Huang Y-C, Su S-Y, Huang J-Y, Lai C-H, Lung C-C, Ho C-C, Liaw Y-P, 2012, Cell type specificity of female lung cancer associated with sulfur dioxide from air pollutants in Taiwan: an ecological study. BMC Publ Health 12(1):4 Giangreco A, Lu L, Vickers C, Teixeira VH, Groot KR, Butler CR, Ilieva EV, George PJ, Nicholson AG, Sage EK, 2012, β–Catenin determines upper airway progenitor cell fate and preinvasive squamous lung cancer progression by modulating epithelial–mesenchymal transition. J Pathol 226(4):575–587 Sequoia Ecosystem and Recreation Preserve Act of 1999, 1999,, trans: Rep. George E. Brown J). 106th Congress edn Hammerman P, 2012, How far away is targeted treatment for squamous cell lung cancer? Oncology Times UK 9(11):21 Wu M, Tu T, Huang Y, Cao Y, 2013, Suppression subtractive hybridization identified differentially expressed genes in lung adenocarcinoma: ERGIC3 as a novel lung cancer-related gene. BMC Cancer 13(1):44 Fang X, Netzer M, Baumgartner C, Bai C, Wang X, 2012, Genetic network and gene set enrichment analysis to identify biomarkers related to cigarette smoking and lung cancer. Cancer Treat Rev Wachi S, Yoneda K, Wu R, 2005, Interactome-transcriptome analysis reveals the high centrality of genes differentially expressed in lung cancer tissues. Bioinformatics 21(23):4205–4208 Team RC, 2008, R: a language and environment for statistical computing. R Found Stat Comput Davis S, Meltzer PS, 2007, GEOquery: a bridge between the Gene Expression Omnibus, GEO, and BioConductor. Bioinformatics 23(14):1846–1847 Diboun I,Wernisch L, Orengo C, Koltzenburg M, 2006, Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma. BMC Genomics 7(1):252 Smyth GK, 2004, Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 3(1):3 BenjaminiY, HochbergY, 1995, Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B, Methodological):289–300 Hulsegge I, Kommadath A, Smits MA, 2009, Globaltest and GOEAST: two different approaches for Gene Ontology analysis. In: BMC proceedings. BioMed Central Ltd, p S10 Zheng Q, Wang X-J, 2008, GOEAST: a web-based software toolkit for Gene Ontology enrichment analysis. Nucleic Acids Res 36(suppl 2):W358–W363 Zhang JD, Wiemann S, 2009, KEGGgraph: a graph approach to KEGG PATHWAY in R and bioconductor. Bioinformatics 25(11): 1470–1471 Weiss J, Sos ML, Seidel D, Peifer M, Zander T, Heuckmann JM, Ullrich RT, Menon R, Maier S, Soltermann A, 2010, Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med 2(62):62ra93–62ra93 Tye BK, 1999, MCM proteins in DNA replication. Annu Rev Biochem 68(1):649–686 Kikuchi J, Kinoshita I, Shimizu Y, Kikuchi E, Takeda K, Aburatani H, Oizumi S, Konishi J, Kaga K, Matsuno Y, 2011, Minichromosome maintenance, MCM, protein 4 as a marker for proliferation and its clinical and clinicopathological significance in non-small cell lung cancer. Lung Cancer 72(2):229–237 Kim YR, Song SY, Kim SS, An CH, Lee SH, Yoo NJ, 2010, Mutational and expressional analysis of RFC3, a clamp loader in DNA replication, in gastric and colorectal cancers. Hum Pathol 41(10):1431–1437 Balakrishnan L, Bambara RA, 2013, Flap Endonuclease 1. Annu Rev Biochem, 0) Yang M, Guo H, Wu C, He Y, Yu D, Zhou L, Wang F, Xu J, Tan W, Wang G, 2009, Functional FEN1 polymorphisms are associated with DNA damage levels and lung cancer risk. Hum Mutat 30(9):1320–1328 Allera-Moreau C, Rouquette I, Lepage B, Oumouhou N,Walschaerts M, Leconte E, Schilling V, Gordien K, Brouchet L, DelisleM(2012, DNA replication stress response involving PLK1, CDC6, POLQ, RAD51 and CLASPIN upregulation prognoses the outcome of early/mid-stage non-small cell lung cancer patients. Oncogenesis 1(10):e30 Li H, Yin C, Zhang B, Sun Y, Shi L, Liu N, Liang S, Lu S, Liu Y, Zhang J, 2013, PTTG1 promotes migration and invasion of human non-small cell lung cancer cells and is modulated by miR-186. Carcinogenesis Andriani F, Roz E, Caserini R, Conte D, Pastorino U, Sozzi G, Roz L, 2012, Inactivation of both FHIT and p53 cooperate in deregulating proliferation-related pathways in lung cancer. J Thorac Oncol 7(4): 631 Daraselia N, Wang Y, Budoff A, Lituev A, Potapova O, Monforte J, Ossovskaya V, 2011, Pathway analysis of primary human non-small cell lung cancer, NSCLC). J Clin Oncol, Meeting Abstracts), p 10573 Yang I-P, Tsai H-L, Hou M-F, Chen K-C, Tsai P-C, Huang S-W, Chou W-W, Wang J-Y, Juo S-HH, 2012, MicroRNA-93 inhibits tumor growth and early relapse of human colorectal cancer by affecting genes involved in the cell cycle. Carcinogenesis 33(8):1522– 1530 Zhao Y, Li X, Sui X, Tang X, Qin H, Ren H, 2010, Expression and significance of PCNA and Caspase-3 in the tissue of lung cancer. Chin J Cell Mol Immunol 26(2):154 Groeger AM, CaputiM, Esposito V, Baldi A, Rossiello R, Santini D, Mancini A, Kaiser HE, Baldi F, 2000, Expression of p21 in non small cell lung cancer relationship with PCNA. Anticancer Res 20(5A):3301 Li C, Johnson DE, 2013, Liberation of functional p53 by proteasome inhibition in human papilloma virus-positive head and neck squamous cell carcinoma cells promotes apoptosis and cell cycle arrest. Cell Cycle 12(6) NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 197 EP 202 DI 10.1007/s12253-013-9685-2 PG 6 ER PT J AU Chen, XJ Shen, J Zhang, GB Chen, WCh AF Chen, Xiao-Juan Shen, Jin Zhang, Guang-Bo Chen, Wei-Chang TI B7-H6 Protein Expression has no Prognostic Significance in Human Gastric Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE B7-H6; Gastric carcinoma; Adjacent non-tumor tissues; Immunohistochemistry ID B7-H6; Gastric carcinoma; Adjacent non-tumor tissues; Immunohistochemistry AB B7-H6, a novel member of the B7 family which binds to NKp30 to trigger antitumor NK cell cytotoxicity and cytokine secretion. Recently, B7-H family has been reported to be a negative regulator of the immune response in patients with gastric carcinoma.However, no reports have investigated the clinical significance of B7-H6 expression in human gastric cancer. We present the first study to the clinicopathological and prognostic value of B7-H6 in primary gastric tumors and adjacent non-tumor tissues at the protein level. Here we show that B7-H6 immunoreactivity was expressed in 6/60 (10 %) gastric tumors and 8/43 (18.60 %) adjacent non-tumor tissues. No statistical difference was found between B7-H6 expression and various prognostic factors; however, B7-H6-positive carcinomas were significantly associated with a higher differentiation (p =0.047). The survival analysis did not confirm the prognostic significance of B7-H6 expression in gastric cancer patients. Our data suggest that B7-H6, as detected by immunohistochemistry, is of limited value as a prognostic marker for gastric cancer. C1 [Chen, Xiao-Juan] The First Affiliated Hospital of Soochow University, Department of Gastroenterology, 215006 Suzhou, China. [Shen, Jin] The First Affiliated Hospital of Soochow University, Department of Gastroenterology, 215006 Suzhou, China. [Zhang, Guang-Bo] The First Affiliated Hospital of Soochow University, Department of Gastroenterology, 215006 Suzhou, China. [Chen, Wei-Chang] The First Affiliated Hospital of Soochow University, Department of Gastroenterology, 215006 Suzhou, China. RP Chen, WCh (reprint author), The First Affiliated Hospital of Soochow University, Department of Gastroenterology, 215006 Suzhou, China. EM chenwcdoc@163.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer Clin 61:69–90., DOI 10.3322/caac Zang X, Allison JP, 2007, The B7 family and cancer therapy. Costimulation and coinhibition. Clin Cancer Res 13:5271–5279 Greaves P, Gribben JG, 2013, The role of B7 family molecules in hematologic malignancy. Blood 121:734–744., DOI 10.1182/blood- 2012-10-385591 Lu B, Chen L, Liu L, Zhu Y, Wu C, Jiang J, Zhang X, 2011, T-cellmediated tumor immune surveillance and expression of B7 coinhibitory molecules in cancers of the upper gastrointestinal tract. Immunol Res 50:269–275., DOI 10.1007/s12026-011-8227-9 Capece D, Verzella D, Fischietti M, Zazzeroni F, Alesse E, 2012, Targeting costimulatory molecules toimprove antitumor immunity. J Biomed Biotechnol 2012:926321., DOI 10.1155/2012/926321 Seliger B, Quandt D, 2012, The expression, function, and clinical relevance of B7 family members in cancer. Cancer Immunol Immunother 61:1327–1341., DOI 10.1007/s00262-012-1293-6 Brandt CS, Baratin M, Yi EC, Kennedy J, Gao Z, Fox B, Haldeman B, Ostrander CD, Kaifu T, Chabannon C, Moretta A,West R, XuW, Vivier E, Levin SD, 2009, The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans. J Exp Med 206:1495–1503., DOI 10.1084/ jem.20090681 Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, 2009, AJCC cancer staging manual, 7th edn. Springer-verlag, New York Wu C, Zhu Y, Jiang J, Zhao J, Zhang XG, Xu N, 2006, Immunohistochemical localization of programmed death-1 ligand-1, PD-L1, in gastric carcinoma and its clinical significance. Acta Histochem 108: 19–24 Liu SM, Meng Q, Zhang QX, 2008, Expression and significance of B7-H1 and its receptor PD-1 in human gastric carcinoma. Zhonghua Zhong Liu Za Zhi 30:192–195 Sun J, Xu K,Wu C,WangY, HuY, ZhuY, ChenY, Shi Q,Yu G, Zhang X, 2007, PD-L1 expression analysis in gastric carcinoma tissue and blocking of tumor-associated PD-L1 signaling by two functional monoclonal antibodies. Tissue Antigens 69:19–27 Wu CP, Jiang JT, TanM, Zhu YB, JiM, Xu KF, Zhao JM, ZhangGB, ZhangXG(2006, Relationship between co-stimulatorymolecule B7- H3 expression and gastric carcinoma histology and prognosis.World J Gastroenterol 12:457–459 Arigami T, Uenosono Y, Hirata M, Yanagita S, Ishigami S, Natsugoe S, 2011, B7-H3 expression in gastric cancer: a novel molecular blood marker for detecting circulating tumor cells. Cancer Sci 102:1019–1024., DOI 10.1111/j.1349-7006.2011. 01877.x Arigami T, Uenosono Y, Ishigami S, Hagihara T, Haraguchi N, Natsugoe S, 2011, Clinical significance of the B7-H4 coregulatory moleculeas a novel prognostic marker in gastric cancer. World J Surg 35:2051–2057., DOI 10.1007/s00268-011- 1186-4 Jiang J, Zhu Y,Wu C, Shen Y,WeiW, Chen L, ZhengX, Sun J, Lu B, Zhang X, 2010, Tumor expression of B7-H4 predicts poor survival of patients suffering from gastric cancer. Cancer Immunol Immunother 59:1707–1714 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 203 EP 207 DI 10.1007/s12253-013-9686-1 PG 5 ER PT J AU Yerrabothala, S Shaaban, H Capo, G Maroules, M Debari, AV AF Yerrabothala, Swaroopa Shaaban, Hamid Capo, Gerardo Maroules, Michael Debari, A Vincent TI The Impact of Diabetes Mellitus on Breast Cancer Outcomes: A Single Center Retrospective Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Practice Guideline DE Diabetes mellitus; Breast neoplasms; Survival ID Diabetes mellitus; Breast neoplasms; Survival AB Diabetes mellitus has been implicated to affect the prognostic outcomes of patients with various types of cancer. This study explores the impact of diabetes mellitus on the survival outcomes of patients with all stages of breast cancer. We performed a retrospective analysis of 255 patients with all stages of breast cancer. Survival outcomes were compared for diabetic and non-diabetic patients. A greater percent of patients in the non-diabetic group (54.1 %) presented with early-stage (stage 0 and 1) cancer than diabetics for which 41.2 % presented with stage 0 or 1 breast cancer; however this difference did not achieve statistical significance (p=0.068). Overall, we observed a significant difference in survival between the diabetics and non-diabetic subjects (p=0.001). Even after adjustment for all covariates and after stratification for Body Mass Index (BMI), diabetics were found to have a poorer prognosis in terms of survival time. In patients with breast cancer, diabetes mellitus is an independent predictor of lower overall survival rates, even after adjusting for other comorbidities. Primary caregivers and oncologists alike should aggressively screen breast cancer patients for diabetes mellitus and vice versa. C1 [Yerrabothala, Swaroopa] Seton Hall University School of Health and Medical Sciences, Trinitas Regional Medical CenterElizabeth, USA. [Shaaban, Hamid] St Joseph’s Regional Medical Center, Seton Hall University School of Health and Medical Sciences, St Michael’s Medical Center, 111 central avenue, 07102 Newark, NJ, USA. [Capo, Gerardo] Seton Hall University School of Health and Medical Sciences, Trinitas Regional Medical CenterElizabeth, USA. [Maroules, Michael] Seton Hall University School of Health and Medical Sciences, St Joseph’s Regional Medical CenterPaterson, USA. [Debari, A Vincent] Seton Hall University School of Health and Medical SciencesSouth Orange, NJ, USA. RP Shaaban, H (reprint author), St Joseph’s Regional Medical Center, Seton Hall University School of Health and Medical Sciences, St Michael’s Medical Center, 07102 Newark, USA. EM hamidshaaban@gmail.com CR American Cancer Society, 2008, Breast cancer facts and figures 2007–2008. American Cancer Society, Atlanta King H, Aubert R, Herman W, 1998, Global burden of diabetes, 1995–2025. Diabetes Care 21:1414–1431 Wolf I, Rubinek T, 2008, Diabetes mellitus and breast cancer. In: Masur K, Thevenod F, Zanker KS, eds, Diabetes and cancer. epidemiological evidence and molecular links. Front diabetes. Vol 19. Karger, Basel, pp 97–113 Chapman JA, Meng D, Shepherd L, Parulekar W, Ingle JN, Muss HB et al, 2008, Competing causes of death from a randomized trial of extended adjuvant endocrine therapy for breast cancer. J Natl Cancer Inst 100:252–260 Van der Burg B, Rutteman GR, Blankenstein MA, de Laat SW, van Zoelen EJ, 1988, Mitogenic stimulation of human breast cancer cells in a growth factor-defined medium: synergistic action of insulin and estrogen. J Cell Physiol 134:101–108 Papa V, Costantino A, Belfiore A, 1997, Insulin receptor what role in breast cancer? Trends Endocrinol Metab 8:306–312 BaroneBB, YehHC, Snyder CF,PeairsKS, SteinKB, Derr RL, 2008, Long-term all-cause mortality in cancer patients with preexisting diabetes mellitus: a systematic review and meta-analysis. JAMA 300:2754–2764 Larsson SC, Mantzoros CS, Wolk A, 2007, Diabetes mellitus and risk of breast cancer: a meta-analysis. Int J Cancer 121:856–862 Peairs KS, Barone BB, Snyder CF, Yeh HC, Stein KB, Derr R et al, 2011, Diabetes mellitus and breast cancer outcomes: a systematic review and meta-analysis. J Clin Oncol 29:40–46 Kaplan MA, Pekkolay Z, Kucukoner M, Inal A, Urakci Z, Ertugrul H et al, 2011, Type 2 diabetes mellitus and prognosis in early stage breast cancer women. Med Oncol 29(3):1576–1580 Unterburger P, Sinop A, NoderW, Berger MR, Fink M, Edler L et al, 1990, Diabetes mellitus and breast cancer. A retrospective followup study. Onkologie 13:17–20 Maruthur NM, Bolen S, Brancati FL, Clarke JM, 2009, Obesity and mammography: a systematic review and meta-analysis. J Gen Intern Med 24:665–677 Calle EE, Kaaks R, 2004, Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer 4:579–591 Pollak MN, Schernhammer ES, Hankinson SE, 2004, Insulin-like growth factors and neoplasia. Nat Rev Cancer 4:505–518 Chlebowski RT, McTiernan A, Wactawski-Wende J, Manson JE, Aragaki AK, Rohan T et al, 2012, Diabetes, metformin, and breast cancer in postmenopausal women. J Clin Oncol 30(23):2844–2852 Goodwin PJ, Pritchard KI, Ennis M, Clemons M, Graham M, Fantus IG, 2008, Insulin-lowering effects of metformin in women with early breast cancer. Clin Breast Cancer 8:501–505 Monami M, Lamanna C, Balzi D, Marchionni N, Mannucci E, 2009, Sulphonylureas and cancer: a case–control study. Acta Diabetol 46:279–284 Bowker SL, Yasui Y, Veugelers P, Johnson JA, 2010, Glucose-lowering agents and cancer mortality rates in type 2 diabetes: assessing effects of time-varying exposure. Diabetologia 53:1631–1637 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 209 EP 214 DI 10.1007/s12253-013-9666-5 PG 6 ER PT J AU Fernandez-Vega, I Quirk, J Norwood, LF Sibtain, AN Laxton, R Bodi, I AF Fernandez-Vega, Ivan Quirk, Jennifer Norwood, L Fiona Sibtain, A Naomi Laxton, Ross Bodi, Istvan TI Gliomatosis Cerebri Type 1 with Extensive Involvement of the Spinal Cord and BRAF V600E Mutation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; Gliomatosis cerebri; BRAF V600E; Brain; Spinal cord ID Glioma; Gliomatosis cerebri; BRAF V600E; Brain; Spinal cord AB Gliomatosis cerebri (GC) is a rare neoplasm in which there is a diffuse cerebral infiltration by malignant glial cells with relative conservation of the underlying structures. A 67-year-old lady was admitted complaining of balance problems, troubled breathing, stuttered speech, decreased mobility, progressive ataxia and also some mild cognitive problems. MRI demonstrated ill defined T2 hyperintensity with mild mass effect mainly involving the brain stem and cerebellar hemispheres, with minor signal abnormalities extending supratentorially along the corticospinal tracts. The imaging appearances were static over a year. No biopsy was performed. The patient received palliative care and died 2 years after initial presentation. Macroscopic examination of the brain showed an extensive firm white-grey lesion predominantly in the cerebellar white matter, the brainstem, spreading to the full length of the spinal cord and invading the sensory ganglia. Histology revealed an extensively infiltrating diffuse glioma with small elongated fusiform nuclei. Diagnosis of GC type 1 was made. Molecular genetic tests revealed BRAF V600E mutation, while no IDH1 & IDH2 mutations were found. GC should be taken into account in the differential diagnoses mainly when there is rapid clinical deterioration without clear evidence of radiological progression. Extensive spinal cord infiltration has been reported only in 9 % and BRAF V600E mutation was detected only in one case in GC previously. Future clinical trials should address whether BRAF V600E mutant brain tumour patients will benefit from BRAF V600E-directed targeted therapies. C1 [Fernandez-Vega, Ivan] Hospital Universitario Central de Asturias, Department of PathologyOviedo, Spain. [Quirk, Jennifer] Princess Royal University Hospital, Department of NeurologyOrpington, UK. [Norwood, L Fiona] Princess Royal University Hospital, Department of NeurologyOrpington, UK. [Sibtain, A Naomi] King’s College Hospital, Department of NeuroradiologyLondon, UK. [Laxton, Ross] King’s College Hospital, Clinical NeuropathologyLondon, UK. [Bodi, Istvan] King’s College Hospital, Clinical NeuropathologyLondon, UK. RP Bodi, I (reprint author), King’s College Hospital, Clinical Neuropathology, London, UK. EM istvan.bodi@nhs.net CR Nevin S, 1938, Gliomatosis cerebri. Brain 61:170–191 Taillibert S, Chodkiewicz C, Laigle-Donadey F, Napolitano M, Cartalat-Carel S, Sanson M, 2006, Gliomatosis cerebri: a review of 296 cases from the ANOCEF database and the literature. J Neurooncol 76(2):201–205., DOI 10.1007/s11060-005-5263-0 Herrlinger U, Felsberg J, Kuker W, Bornemann A, Plasswilm L, Knobbe CB, Strik H, Wick W, Meyermann R, Dichgans J, Bamberg M, Reifenberger G, Weller M, 2002, Gliomatosis cerebri: molecular pathology and clinical course. Ann Neurol 52(4):390–399., DOI 10.1002/ana.10297 Thomas RP, Xu LW, Lober RM, Li G, Nagpal S, 2013, The incidence and significance of multiple lesions in glioblastoma. J Neurooncol 112(1):91–97., DOI 10.1007/s11060-012-1030-1 Lantos PLBJ, 2000, Gliomatosis cerebri. In: Kleihues P, Cavence WK, eds, Pathology and genetics of tumours of the nervous system. IARC Press, Lyon, pp 92–93 Setty P, Hammes J, Rothamel T, Vladimirova V, Kramm CM, Pietsch T, Waha A, 2010, A pyrosequencing-based assay for the rapid detection of IDH1 mutations in clinical samples. J Mol Diagn 12(6):750–756., DOI 10.2353/jmoldx.2010.090237 Song X, Andrew Allen R, Terence Dunn S, Fung KM, Farmer P, Gandhi S, Ranjan T, Demopoulos A, Symons M, Schulder M, Li JY, 2011, Glioblastoma with PNET-like components has a higher frequency of isocitrate dehydrogenase 1, IDH1, mutation and likely a better prognosis than primary glioblastoma. Int J Clin Exp Pathol 4(7):651–660 Park S, Suh YL, Nam DH, Kim ST, 2009, Gliomatosis cerebri: clinicopathologic study of 33 cases and comparison of mass forming and diffuse types. Clin Neuropathol 28(2):73–82 Troost D, Kuiper H, Valk J, Fleury P, 1987, Gliomatosis cerebri, report of a clinically diagnosed and histologically confirmed case. Clin Neurol Neurosurg 89(1):43–47 LA PiccirilliM, SalvatiM(2006, Gliomatosis cerebri treatment in 11 elderly patients. J Exp Clin Cancer Res 25(2):183–187 Kim DG, Yang HJ, Park IA, Chi JG, Jung HW, Han DH, Choi KS, Cho BK, 1998, Gliomatosis cerebri: clinical features, treatment, and prognosis. Acta Neurochirurgica 140(8):755–762., DOI 10.1007/ s007010050176 Kong DS, Kim ST, Lee JI, Suh YL, Lim do H, Kim WS, Kwon KH, Park K, Kim JH, Nam DH, 2010, Impact of adjuvant chemotherapy for gliomatosis cerebri. BMC Cancer 10:424., DOI 10.1186/1471- 2407-10-424 Narasimhaiah D, Miquel C, Verhamme E, Desclee P, Cosnard G, Godfraind C, 2012, IDH1 mutation, a genetic alteration associated with adult gliomatosis cerebri. Neuropathology 32(1):30–37., DOI 10. 1111/j.1440-1789.2011.01216.x Artigas J, Cervos-Navarro J, Iglesias JR, Ebhardt G, 1985, Gliomatosis cerebri: clinical and histological findings. Clin Neuropathol 4(4):135–148 Corbett JJ, Newman NM, 1981, Symptomatic leptomeningeal metastases preceding other manifestations of occult primary brain tumors. Surg Neurol 15(5):362–367 Karaca MAM, Hicsonmez A, Guney Y, Kurtman C, 2006, Cases of glioblastoma multiforme metastasizing to spinal cord. Neurol India 54:428–430 Lawton CD, Nagasawa DT, Yang I, Fessler RG, Smith ZA, 2012, Leptomeningeal spinal metastases from glioblastoma multiforme: treatment and management of an uncommon manifestation of disease. J Neurosurg Spine 17(5):438–448., DOI 10.3171/2012.7. SPINE12212 Balko MG, Blisard KS, Samaha FJ, 1992, Oligodendroglial gliomatosis cerebri. Hum Pathol 23(6):706–707 Capper D, Weissert S, Balss J, Habel A, Meyer J, Jager D, Ackermann U, Tessmer C, Korshunov A, Zentgraf H, Hartmann C, vonDeimlingA(2010, Characterization of R132Hmutation-specific IDH1 antibody binding in brain tumors. Brain Pathol 20(1):245–254., DOI 10.1111/j.1750-3639.2009.00352.x Seiz M, Tuettenberg J, Meyer J, Essig M, Schmieder K, Mawrin C, von Deimling A, Hartmann C, 2010, Detection of IDH1mutations in gliomatosis cerebri, but only in tumors with additional solid component: evidence for molecular subtypes. Acta Neuropathol 120(2): 261–267., DOI 10.1007/s00401-010-0701-2 Desestret V, Ciccarino P, Ducray F, Criniere E, Boisselier B, Labussiere M, Polivka M, Idbaih A, Kaloshi G, von Deimling A, Hoang-Xuan K, Delattre JY,Mokhtari K, SansonM(2011, Prognostic stratification of gliomatosis cerebri by IDH1 R132H and INA expression. J Neurooncol 105(2):219–224., DOI 10.1007/s11060-011-0587-4 Kwon MJ, Kim ST, Kong DS, Lee D, Park S, Kang SY, Song JY, Nam DH, Kato Y, Choi YL, Suh YL, 2012, Mutated IDH1 is a favorable prognostic factor for type 2 gliomatosis cerebri. Brain Pathol 22(3):307–317., DOI 10.1111/j.1750-3639.2011.00532.x Mawrin C, Kirches E, Schneider-Stock R, Boltze C, Vorwerk CK, vonMawrin A, vonDeimling A, Stoltenburg-Didinge G, Bornemann A, Romeike B, Sellhaus B, Dietzmann K, 2005, Alterations of cell cycle regulators in gliomatosis cerebri. J Neurooncol 72(2):115–122., DOI 10.1007/s11060-004-2061-z LiuW, Lv G, Li Y, Li L,Wang B, 2011, Downregulation of CDKN2A and suppression of cyclin D1 gene expressions in malignant gliomas. J Exp Clin Cancer Res 30:76., DOI 10.1186/1756-9966-30-76 Di Giovanni S,Movsesyan V, Ahmed F, Cernak I, Schinelli S, Stoica B, Faden AI, 2005, Cell cycle inhibition provides neuroprotection and reduces glial proliferation and scar formation after traumatic brain injury. Proc Natl Acad Sci U S A 102(23):8333–8338., DOI 10. 1073/pnas.0500989102 Kouri FM, Jensen SA, Stegh AH, 2012, The role of Bcl-2 family proteins in therapy responses ofmalignant astrocytic gliomas: Bcl2L12 and beyond. Sci World J 2012:838916., DOI 10.1100/2012/838916 Mawrin C, Lins H, Kirches E, Schildhaus HU, Scherlach C, Kanakis D, Dietzmann K, 2003, Distribution of p53 alterations in a case of gliomatosis cerebri. Hum Pathol 34(1):102–106., DOI 10.1053/hupa. 2003.1 Wan PTGM, Roe SM, Lee S, NiculescuDuvaz D, Good VM, Jones CM, Marshall CJ, Springer CJBD, Marais R, 2004, Cancer genome project.Mechanismof activation of the RAF-ERKsignaling pathway by oncogenic mutations of B-RAF. Cell 116:855–867., DOI 10.1016/ S0092-8674(04)00215-6 Capper D, Berghoff AS, von DeimlingA, PreusserM(2012, Clinical neuropathology practice news 2-2012: BRAF V600E testing. Clin Neuropathol 31(2):64–66 Schindler G, Capper D, Meyer J, Janzarik W, Omran H, Herold- Mende C, Schmieder K, Wesseling P, Mawrin C, Hasselblatt M, Louis DN, Korshunov A, Pfister S, Hartmann C, Paulus W, Reifenberger G, von Deimling A, 2011, Analysis of BRAF V600E mutation in 1,320 nervous system tumors reveals high mutation frequencies in pleomorphic xanthoastrocytoma, ganglioglioma and extra-cerebellar pilocytic astrocytoma. Acta Neuropathol 121(3): 397–405., DOI 10.1007/s00401-011-0802-6 Kleinschmidt-DeMasters BK, Aisner DL, Birks DK, Foreman NK, 2013, Epithelioid GBMs show a high percentage of BRAF V600E mutation. Am J Surg Pathol 37(5):685–698., DOI 10.1097/PAS. 0b013e31827f9c5e Huillard E, Hashizume R, Phillips JJ, Griveau A, Ihrie RA, Aoki Y, Nicolaides T, Perry A, Waldman T, McMahon M, Weiss WA, Petritsch C, James CD, Rowitch DH, 2012, Cooperative interactions of BRAFV600E kinase and CDKN2A locus deficiency in pediatric malignant astrocytoma as a basis for rational therapy. Proc Natl Acad Sci U S A 109(22):8710–8715., DOI 10.1073/pnas.1117255109 Nicolaides TP, Li H, Solomon DA, Hariono S, Hashizume R, Barkovich K, Baker SJ, Paugh BS, Jones C, Forshew T, Hindley GF, Hodgson JG, Kim JS, Rowitch DH, Weiss WA, Waldman TA, James CD, 2011, Targeted therapy for BRAFV600E malignant astrocytoma. Clin Cancer Res 17(24):7595–7604., DOI 10.1158/ 1078-0432.CCR-11-1456 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 215 EP 220 DI 10.1007/s12253-013-9732-z PG 6 ER PT J AU Kim, SM Yoo, JN Lee, HS AF Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Expressional and Mutational Analysis of CREBBP Gene in Gastric and Colorectal Cancers with Microsatellite Instability SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Cancer Research Institute, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Cancer Research Institute, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Cancer Research Institute, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Cancer Research Institute, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Gonzalez GA, Montminy MR, 1989, Cyclic AMP stimulates somatostatin gene transcription by phosphorylation of CREB at serine 133. Cell 59:675–680 Iyer NG, Ozdag H, Caldas C, 2004, p300/CBP and cancer. Oncogene 23:4225–4231 Bartsch O, KressW, Kempf O, Lechno S, Haaf T, Zechner U, 2010, Inheritance and variable expression in Rubinstein-Taybi syndrome. Am J Med Genet A 152A:2254–2261 Gui Y, Guo G, Huang Y, Hu X, Tang A, Gao S, Wu R, Chen C, Li X, Zhou L, He M, Li Z, Sun X, JiaW, Chen J, Yang S, Zhou F, Zhao X,Wan S, Ye R, Liang C, Liu Z, Huang P, Liu C, Jiang H,Wang Y, Zheng H, Sun L, Liu X, Jiang Z, Feng D, Chen J, Wu S, Zou J, Zhang Z, Yang R, Zhao J, Xu C, YinW, Guan Z, Ye J, Zhang H, Li J, Kristiansen K, Nickerson ML, Theodorescu D, Li Y, Zhang X, Li S, Wang J, Yang H, Wang J, Cai Z, 2011, Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat Genet 43:875–878 Borrow J, Stanton VP Jr, Andresen JM, Becher R, Behm FG, Chaganti RS, Civin CI, Disteche C, Dube I, Frischauf AM, Horsman D, Mitelman F, Volinia S, Watmore AE, Housman DE, 1996, The translocation t(8;16)(p11;p13, of acute myeloid leukaemia fuses a putative acetyltransferase to the CREB-binding protein. Nat Genet 14:33–41 Mullighan CG, Zhang J, Kasper LH, Lerach S, Payne-Turner D, Phillips LA, Heatley SL, Holmfeldt L, Collins-Underwood JR, Ma J, Buetow KH, Pui CH, Baker SD, Brindle PK, Downing JR, 2011, CREBBP mutations in relapsed acute lymphoblastic leukaemia. Nature 471:235–239 Pasqualucci L, Dominguez-Sola D, Chiarenza A, Fabbri G, Grunn A, Trifonov V, Kasper LH, Lerach S, Tang H, Ma J, Rossi D, Chadburn A, Murty VV, Mullighan CG, Gaidano G, Rabadan R, Brindle PK, Dalla-Favera R, 2011, Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature 471:189–195 Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, Meltzer SJ, Rodriguez-Bigas MA, Fodde R, Ranzani GN, Srivastava S, 1998, A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58: 5248–5257 Kim MS, Hur SY, Yoo NJ, Lee SH, 2010, Mutational analysis of FOXL2 codon 134 in granulosa cell tumour of ovary and other human cancers. J Pathol 221:147–152 Kim MS, Oh JE, Kim YR, Park SW, Kang MR, Kim SS, Ahn CH, Yoo NJ, Lee SH, 2010, Somatic mutations and losses of expression of microRNA regulation-related genes AGO2 and TNRC6A in gastric and colorectal cancers. J Pathol 221:139–146 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2014 VL 20 IS 1 BP 221 EP 222 DI 10.1007/s12253-013-9668-3 PG 2 ER PT J AU Reptova, S Trtkova, SK Kolar, Z AF Reptova, Silvie Trtkova, Smesny Katerina Kolar, Zdenek TI Direct Detection of the AR-E211 G>A Gene Polymorphism from Blood and Tissue Samples Without DNA Isolation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gene polymorphism; Prostate cancer; Androgen receptor; Direct method ID Gene polymorphism; Prostate cancer; Androgen receptor; Direct method AB The pathogenesis of prostate cancer (CaP) involves alterations in a gene structure of the androgen receptor (AR). The single nucleotide polymorphism AR-E211 G>A localized in exon 1 of the AR gene (G1733A) was detected using direct polymerase chain reaction and restriction digestion (PCRRFLP) method on blood and tissue samples without prior DNA isolation. We used blood samples of patients with a diagnosis of benign prostatic hyperplasia (BPH) or CaP. From monitored group of CaP patients were selected specimen in formalin-fixed paraffin-embedded tissue blocks with morphology of BPH and CaP. The main objective of our study was to develop a method based the direct PCR-RFLP analysis from blood and tissue without prior DNA isolation for faster genotyping analysis of a large number of samples. We found no statistically significant differences in allelic % of the ARE211 G>A polymorphism between BPH and CaP patients (p≤0.8462). Genotyping of the AR-E211 G>A variant in blood was not identical with tumor tissue genotyping analysis. Significant agreement between blood and tissue AR-E211 G>A polymorphism only in non-tumor tissue focus was confirmed. Although we analyzed a limited number of the tissue samples, we suppose that a presence of the minor allele A may be associated with cancer transformation-induced changes of the modified AR gene. C1 [Reptova, Silvie] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular Pathology, Hnevotinska 3, 775 15 Olomouc, Czech Republic. [Trtkova, Smesny Katerina] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular Pathology, Hnevotinska 3, 775 15 Olomouc, Czech Republic. [Kolar, Zdenek] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular Pathology, Hnevotinska 3, 775 15 Olomouc, Czech Republic. RP Trtkova, SK (reprint author), Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular Pathology, 775 15 Olomouc, Czech Republic. EM trtkova@post.cz CR Casella R, Maduro MR, Lipshultz LI, Lamb DJ, 2001, Significance of the polyglutamine tract polymorphism in the androgen receptor. Urology 5:651–656 Gottlieb B, Beitel LK, Wu JH, Trifiro M, 2004, The androgen receptor gene mutations database, ARDB): 2004 update.Hum Mutat 23:527–533 Beilin J, Ball EMA, Favaloro JM, Zajac JD, 2000, Effect of the androgen receptor CAG repeat polymorphism on transcriptional activity: specifity in prostate and non-prostate cell lines. J Mol Endocrinol 25:85–96 Zitzmann M, Nieschlag E, 2003, The CAGrepeat polymorphismwithin the androgen receptor gene and maleness. Int J Androl 26:76–83 Chang BL, Zheng SL, Hawkins GA, Isaacs SD, Wiley KE, Turner A et al, 2002, Polymorphic GGC repeats in the androgen receptor gene are associated with hereditary and sporadic prostate cancer risk. Hum Genet 110:122–129 Chen C, Lamharzi N, Weiss NS, Etzioni R, Dightman DA, Barnett et al, 2002, Androgen receptor polymorphisms and the incidence of prostate cancer. Cancer Epidemiol Biomarkers Prev 11:985–986 Ferro P, Catalano MG, Dell’Eva R, Fortunati N, Pfeffer U, 2002, The androgen receptor CAG repeat: a modifier of carcinogenesis? Mol Cell Endocrinol 193:109–120 Mononen N, Ikonen T, Autio V, Rokman A, Matikainen MP, Teuvo L et al, 2002, Androgen receptor CAG polymorphism and prostate cancer risk. Hum Genet 111:166–171 Silva NB, Koff WJ, Biolchi V, Brenner C, Biolo KD, Spritzer PM et al, 2008, Polymorphic CAG and GGC repeat lengths in the androgen receptor gene and prostate cancer risk: analysis of a Brazilian population. Cancer Invest 26:74–80 Edwards A, Hammond HA, Jin L, Caskey CT, Chakraborty R, 1992, Genetic variation at five trimeric and tetrameric tandem repeat loci in four human population groups. Genomic 12:241–253 Irvine RA, Yu MC, Ross RK, Coetzee GA, 1995, The CAG and GGC microsatellites of the androgen receptor gene are in linkage disequilibrium in men with prostate cancer. Cancer Res 55:1937– 1940 Hsing AW, Tsao L, Devesa SS, 2000, International trends and patterns of prostate cancer incidence and mortality. Int J Cancer 85:60–67 Lu J, Danielsen M, 1996, A StuI polymorphism in the human androgen receptor gene, AR). Clin Genet 49:323–324 Ross RK, Pike MC, Coetzee GA, Reichardt JKV, Yu MC, Feigelson H, 1998, Androgen metabolism and prostate cancer: Establishing a model of genetic susceptibility. Cancer Res 58:4497– 4504 Gray IC, Campbell DA, Spurr NK, 2000, Single nucleotide polymorphisms as tools in human genetics. Hum Mol Genet 9:2403– 2408 Shen IC, Rideout WM, Jones PA, 1994, The rate of hydrolytic deamination of 5-methylcytosine in double-stranded DNA. Nucleic Acids Res 22:972–974 Ellis JA, Stebbing M, Harrap SB, 2001, Polymorphism of the androgen receptor gene is associated with male pattern baldness. J Invest Dermatol 116:452–455 Sasaki M, Karube A, Karube Y,Watari M, Sakuragi N, Fujimoto S, Dahiya R, 2005, GGC and StuI polymorphism on the androgen receptor gene in endometrial cancer patients. Biochem Biophys Res Commun 329:100–104 Hayes VM, Severi G, Eggleton SA, Padilla EJD, Southey MC, Sutherland RL, Hopper JL, Giles GG, 2005, The E211 G>A androgen receptor polymorphism is associated with a decreased risk of metastatic prostate cancer and androgenetic alopecia. Cancer Epidemiol Biomarkers Prev 14:993–996 Medeiros R, Vasconcelos A, Costa S, Pinto D, Morais A, Oliveira J, Lopees C, 2003, Steroid hormone genotypes ARStuI and ER325 are linked to the progression of human prostate cancer. Cancer Genet Cytogenet 141:91–96 Ribeiro ML, Santos A, Carvalho-Salles AB, Hackel C, 2002, Allelic frequencies of six polymorphic markers for risk of prostate cancer. Braz J Med Biol Res 35:205–213 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 223 EP 227 DI 10.1007/s12253-013-9671-8 PG 5 ER PT J AU Liu, Ch Cui, Q Guo, J Li, D Zeng, Y AF Liu, Cheng Cui, Qiu Guo, Jun Li, Dingfeng Zeng, Yanjun TI Intra-Arterial Intervention Chemotherapy for Sarcoma and Cancerous Ulcer Via an Implanted Pump SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sarcoma ulcer; Cancerous ulcer; Chemotherapy pump; Intra-arterial chemotherapy ID Sarcoma ulcer; Cancerous ulcer; Chemotherapy pump; Intra-arterial chemotherapy AB To observe the efficacy of intra-arterial chemotherapy with subcutaneously implanted pump for soft tissue sarcoma in extremities and cancerous ulcer. 31 patients with ulcerative skin squamous cell carcinoma or sarcoma in extremities who received treatment during the period from July 2003 to November 2011 at our hospital were recruited, including 15 male and 16 female patients, aging between 14 and 83 with average age of 49 years old. 10 patients had tumor in upper extremities and 21 patients in lower extremities. The pathological types of studied cases include 9 cases with skin squamous cell carcinoma, 6 cases with synovial sarcoma, 5 cases with malignant fibrous histiocytoma, 3 cases with liposarcoma, 3 cases with osteosarcoma, 2 cases with malignant melanoma, 2 cases with epidermoid sarcoma, and 1 case with protuberans. The main symptoms of cancerous ulcer were pain, infection and hemorrhage; All the studied patients were administrated with cisplatin and doxorubicin by intra-arterial chemotherapy pump, and the patients with squamous cell carcinoma were additionally applied with bleomycin and patients with malignant melanoma were additionally applied with dacarbazine. The chemotherapy efficiency was observed after at 3 cycles of intra-arterial chemotherapy. The total remission rate of pain (RR) was 87 %, and total remission rate of ulcer cicatrization (RR) was 71 %, with ulcer cicatrizing spontaneously in 9 cases and obvious homeostasis in 5 cases with bleeding ulcers. 19 patients underwent surgery after chemotherapy, in which 16 cases had limbsalvage surgery and 3 cases underwent lower leg amputation after chemotherapy, and 3 patients out of 16 cases had local recurrence (19 %). The subcutaneous intra-arterial targeting chemotherapy could be applied to treat refractory sarcoma and cancerous ulcer in extremities to significantly increase the chemotherapeutic concentration at tumor area so as to effectively constrain the tumor rupture induced main symptoms including pain, infection and bleeding, which would help to make a decreased blood supplied and well defined tumor boundary to finally decrease the recurrence rate. C1 [Liu, Cheng] Affiliated Hospital of Academy of Military Medical Sciences, Department of Orthopedics (307 Hospital of PLA), 100071 Beijing, China. [Cui, Qiu] Affiliated Hospital of Academy of Military Medical Sciences, Department of Orthopedics (307 Hospital of PLA), 100071 Beijing, China. [Guo, Jun] Affiliated Hospital of Academy of Military Medical Sciences, Department of Orthopedics (307 Hospital of PLA), 100071 Beijing, China. [Li, Dingfeng] Affiliated Hospital of Academy of Military Medical Sciences, Department of Orthopedics (307 Hospital of PLA), 100071 Beijing, China. [Zeng, Yanjun] Beijing University of Technology, Biomechanics and Medical Information Institute, 100022 Beijing, China. RP Zeng, Y (reprint author), Beijing University of Technology, Biomechanics and Medical Information Institute, 100022 Beijing, China. EM yjzeng@bjut.edu.cn CR Yan S, Jichang Z, eds,, 2003)Manual for clinical cancer medicine[M]. People’s Medical Publishing House, Beijing, p 106 Mei W, Xizhi G, 1982, WHO standards for evaluating the cancer treatment effects. Chin J Oncol 4(4):300 Xu HW, Ren F, Chen Wet al, 2012, Osseous metastasis of cutaneous squamous cell carcinoma treated successfully with oxaliplatin, tegafur and leucovorin combination chemotherapy: a case report. Int J Clin Exp Med 5(1):87–91 Hong G, Yaling G, 2005, Clinical observation of using Alpha interferon to treat cancerous ulcer. J Clin Oncol 10(4):416–417 Wang YF, Que HF, Xu JN et al, 2012, Assessment of external methods of traditional Chinese medicine in patients with chronic ulcer of the lower extremities: study protocol of a multicenter, randomized, parallel-group, prospective trial. Zhong Xi Yi Jie He Xue Bao 10(2):166–175 Wang SQ, Goldberg LH, 2007, Pulsed-dye laser treatment of nonhealing chronic ulcer with hypergranulation tissue. Arch Dermatol 143(6):700–702 Siqi F, Jianxing S, 2010, Free flap transplantation in the treatment of 1 case of huge chronic ulcer induced by breast cancer. Chin J Aesthet Med 19(7):978–979 Guo J, Cui Q, Liu C et al, 2013, Clinical report on transarterial neoadjuvant chemotherapy of malignant fibrous histiocytoma in soft tissue. Clin Transl Oncol 15:370–375 Cheng L, Dingfeng L, Ju Z et al, 2009, Comprehensive treatment of malignant tumor of distal femur based on interventional chemotherapy. Chin J Clin Oncol Rehabil 3(4):250–253 Cui Q, Li DF, Liu C et al, 2011, Two case-reports of the limb salvage treatment of osteosarcoma consolidated with obvious pathological fractures. Pathol Oncol Res 17:973–979 Li D, Cui Q, Liu Yet al, 2011, Chemotherapy response analysis for osteosarcoma with intra-arterial chemotherapy by subcutaneous implantable delivery system. Pathol Oncol Res 17(4):947–953 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 229 EP 234 DI 10.1007/s12253-013-9673-6 PG 6 ER PT J AU Kosmaczewska, A Ciszak, L Swierkot, J Szteblich, A Wiland, P Frydecka, I AF Kosmaczewska, Agata Ciszak, Lidia Swierkot, Jerzy Szteblich, Aleksandra Wiland, Piotr Frydecka, Irena TI Alterations in Both the Activatory and Inhibitory Potential of Peripheral Blood CD4+ T Cells in Rheumatoid Arthritis Patients Correlate with Disease Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD28; CD40L; CTLA-4; Rheumatoid arthritis; Disease progression; Therapy ID CD28; CD40L; CTLA-4; Rheumatoid arthritis; Disease progression; Therapy AB The chronic nature of rheumatoid arthritis (RA) suggests immune dysfunction, including persistent systemic activation. Therefore, we evaluated the activatory and inhibitory potential as well as proliferative activity of peripheral blood (PB) CD4+ T cells from RA patients in different stages of the disease and after different therapeutic interventions.We found that CD4+ T cells from RA patients were activated in vivo concerning decreased CD28 expression and increase of CD40L, CD69, and CTLA-4 expression; however, the extent of stimulation was suboptimal when compared to healthy controls. Consequently, impaired proliferative activities of these cells were found in all patients irrespective of the active disease duration. Treatment with methotrexate (MTX) and/or inhibitors of TNF-alpha (iTNF) did not significantly influence systemic activation in RA patients, which corresponded with the maintenance of inflammation markers; however, partial restoration of CD28 and CTLA-4 expression as well as clinical improvement were observed. In patients with early disease (the MTX group), we noted higher capacity of CD4+ T cells for restoration of T cell function, whereas cells from the iTNF group with progressive disease remained with a proliferative defect after the treatment. In conclusion, our study demonstrates that the dysregulated expression of molecules interfering with CD4+ Tcell signaling may result in functional impairment of the effector T cells and correlates with disease progression. C1 [Kosmaczewska, Agata] Polish Academy of Sciences, Institute of Immunology and Experimental TherapyWroclaw, Poland. [Ciszak, Lidia] Polish Academy of Sciences, Institute of Immunology and Experimental TherapyWroclaw, Poland. [Swierkot, Jerzy] Wroclaw Medical University, Department of Rheumatology and Internal MedicineWroclaw, Poland. [Szteblich, Aleksandra] Polish Academy of Sciences, Institute of Immunology and Experimental TherapyWroclaw, Poland. [Wiland, Piotr] Wroclaw Medical University, Department of Rheumatology and Internal MedicineWroclaw, Poland. [Frydecka, Irena] Polish Academy of Sciences, Institute of Immunology and Experimental TherapyWroclaw, Poland. RP Kosmaczewska, A (reprint author), Polish Academy of Sciences, Institute of Immunology and Experimental Therapy, Wroclaw, Poland. EM kosmacz@iitd.pan.wroc.pl CR Weyand CM, 2000, New insights into the pathogenesis of rheumatoid arthritis. Rheumatology 39:13–18 Lenschow DJ, Walunas TL, Bluestone JA, 1996, CD28/B7 system of T cell costimulation. Annu Rev Immunol 14:233–258 Elgueta R, Benson MJ, Vries VC, Wasiuk A, Guo YX, Noelle RJ, 2009)Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol Rev 229:152–171 Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Greene JM et al, 1994, CTLA-4 can function as a negative regulator of T cell activation. Immunity 1:405–413 Krummel MF, Allison JP, 1995, CD28 and CTLA-4 have opposing effects on the response of Tcells to stimulation. J Exp Med 182:459–465 Lindsten T, Lee KP, Harris ES, Petryniak B, Craihead N, Reynolds PJ et al, 1993, Characterization of CTLA-4 structure and expression on human T cells. J Immunol 151:3489–3499 Noelle RJ, Roy M, Shepherd DM, Stamenkovic I, Ledbetter JA, Aruffo A, 1992, A 39-kDa protein signal for an activated helper T cells binds CD40 and transduces the signal for cognate activation of B cell. Proc Natl Acad Sci 89:6550–6554 Kim YO, Kim HJ, Kim SK, Chung JH, Hong SJ, 2010, Association of the CD28/CTLA4/ICOS polymorphisms with susceptibility to rheumatoid arthritis. Clin Chem Lab Med 48:345–353 Kremer JM,Westhovens R, LeonM,DiGiorgio E, Alten R, Steinfeld S et al, 2003, Treatment of rheumatoid arthritis by selective inhibition of T-cell activation with fusion protein CTLA-4Ig. N Engl J Med 349:1907–1915 Cao J, Zou L, Luo P, Chen P, Zhang L, 2012, Increased production of circulating soluble co-stimulatory molecules CTLA-4, CD28 and CD80 in patients with rheumatoid arthritis. Int Immunopharmacol 14:585–592 Goules A, Tzioufas AG, Manousakis MN, Kirou KA, Crow MK, Routsias JG, 2006, Elevated levels of soluble CD40 ligand, sCD40L, in serum of patients with systemic autoimmune diseases. J Autoimmun 26:165–171 Berner B, Wolf G, Hummel KM, Muller GA, Reuss-Borst MA, 2000, Increased expression of CD40 ligand, CD154, on CD4+ T cells as a marker of disease activity in rheumatoid arthritis. Ann Rheum Dis 59:190–195 Early GS, Zhao W, Burns CM, 1996, Anti-CD40 ligand antibody treatment prevents the development of lupus-like nephritis in a subset of a New Zealand black x New Zealand white mice. Response correlates with the absence of an anti-antibody response. J Immunol 157:3159–3164 Kawashima M, Miossec P, 2005, mRNA quantification of T-bet, GATA-3, IFN-gamma, and IL-4 shows a defective Th1 immune response in the peripheral blood from rheumatoid arthritis patients: link with disease activity. J Clin Immunol 25:209–214 van Roon JAG,Verhoef CM, van Roy JLAMet al, 1997, Decrease in peripheral type 1 over type 2 T cell cytokine production in patients with rheumatoid arthritis correlates with an increase in severity of disease. Ann Rheum Dis 56:656–660 Garcia de Tena J, Manzano L, Leal JC, San Antonio E, Sualdea V, Alvarez-Mon M, 2004, Active Crohn’s disease patients show a distinctive expansion of circulating memory CD4+CD45RO+ CD28null T cells. J Clin Immunol 24:185–196 Martens PB, Goronzy JJ, Schaid D, Weyand CM, 1997, Expansion or unusual CD4+ T cells in severe rheumatoid arthritis. Arthritis Rheum 40:1106–1114 Duftner C, Goldberger C, Falkenbach A,Wurzner R, Falkensammer B, Pfeiffer FP et al, 2003, Prevalence, clinical relevance and characterization of circulating cytotoxic CD4+CD28- T cells in ankylosing spondylitis. Arthritis Res Ther 5:R292–R300 Markovic-Plese S, Cortese I, Wandinger KP, McFarland HF, Martin R, 2001, CD4+CD28- costimulation-independent T cells in multiple sclerosis. J Clin Invest 108:1185–1194 Bryl E, Vallejo AN, Matteson EL, Witkowski JM, Weyand CM, Goronzy JJ, 2005, Modulation of CD28 expression with anti-tumor necrosis factor alpha therapy in rheumatoid arthritis. Arthritis Rheum 52:2996–3003 Schmidt D, Goronzy JJ, Weyand CM, 1996, CD4+CD7-CD28- T cells are expanded in rheumatoid arthritis and are characterized by autoreactivity. J Clin Invest 97:2027–2037 Pawlowska J, Smolenska Z, Zdrojewski Z, Witkowski JM, Bryl E, 2012, Changes in proliferation kinetics of T cells: a new predictive cellular biomarkers for early rheumatoid arthritis? J Clin Immunol., DOI 10.1007/s10875-012-9692-1 Bryl E, Vellejo AN, Weyand CM, Goronzy JJ, 2001, Down-regulation of CD28 expression by TNF-alpha. J Immunol 167:3231–3238 Vallejo AN, Weyand CM, Goronzy JJ, 2004, T-cell senescence: a culprit of immune abnormalities in chronic inflammation and persistent infection. Trends Mol Med 10:119–124 Feldmann M, Maini RN, 2001, Anti-TNF-alpha therapy of rheumatoid arthritis: what have we learned? Annu Rev Immunol 19:163–196 Valenzuela HF, Effros RB, 2002, Divergent telomerase and CD28 expression patterns in human CD4 and CD8 Tcells following repeated encounters with the same antigenic stimulus. Clin Immunol 105: 117–125 Yamashita K, Fukushima K, 2004, The carbohydrate recognition by cytokines modulates their physiological activities. Glycoconj J 21:31–34 Pawlik A,Ostanek L, Brzosko I, BrzoskoM,MasiukM, Machalinski B et al, 2003, The expansion of CD4+CD28- T cells in patients with rheumatoid arthritis. Arthritis Res Ther 5:R210–R213 Kansas GS, Wood GS, Tedder TF, 1991, Expression, distribution, and chemistry of human CD39. Role in activation-associated homotypic adhesion of lymphocytes. J Immunol 146:2235–2244 Liao J, Liang G, Xie S, Zhao H, Zuo X, Li F et al, 2012, CD40L demethylation in CD4+ T cells from women with rheumatoid arthritis. Clin Immunol 145:13–18 Warrington KJ, Vallejo AN, Weyand CM, Goronzy JJ, 2003, CD28 loss in senescent CD4+ T cells; reversal by interleukin-12 stimulation. Blood 101:3543–3549 Kosmaczewska A, Bilinska M, Ciszak L, Noga L, Pawlak E, Szteblich A et al, 2007, Different patterns of activation markers expression and CD4+ T-cell responses to ex vivo stimulation in patients with clinically quiescent multiple sclerosis. J Neuroimmunol 186:137–146 Cope AP, Londei M, Chu NR, Cohen SB, Elliott MJ, Brennan FM, Maini RN, Feldmann M, 1994, Chronic exposure to tumor necrosis factor, TNF, in vitro impairs the activation of T cells through the T cell receptor/CD3 complex; reversal in vivo by anti-TNF antibodies in patients with rheumatoid arthritis. J Clin Invest 94:749–760 Koetz K, Bryl E, Spickschen K, O’FallonWM, Goronzy JJ,Weyand CM, 2000, T cell homeostasis in patients with rheumatoid arthritis. Proc Natl Acad Sci U S A 97:9203–9208 Wagner U, PiererM,Wahle M, Moritz F, Kaltenhauser S, Hantzschel H, 2004, Ex vivo homeostatic proliferation of CD4+ T cells in rheumatoid arthritis is dysregulated and driven by membraneanchored TNF-alpha. J Immunol 173:2825–2833 Hodes RJ, Hathcock KS, Weng NP, 2002, Telomeres in B and T cells. Nat Rev Immunol 2:699–706 Parish ST, Wu JE, Effros RB, 2009, Modulation of T lymphocyte replicative senescence via TNF-alpha inhibition: role of caspase-3. J Immunol 182:4237–4243 Fujii H, Shao L, Colmegna I, Goronzy JJ, Weyand CM, 2009, Telomerase insufficiency in rheumatoid arthritis. Proc Natl Acad Sci U S A 106:4360–4365 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 235 EP 243 DI 10.1007/s12253-013-9687-0 PG 9 ER PT J AU Wang, BH Jiang, BZ Li, M AF Wang, Bin Hong Jiang, Biao Zhi Li, Min TI Research on the Typical miRNA and Target Genes in Squamous Cell Carcinoma and Adenocarcinoma of Esophagus Cancer with DNA Microarray SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal cancer; Differentially expressed miRNA; Target genes interaction network; Function and pathway enrichment analysis ID Esophageal cancer; Differentially expressed miRNA; Target genes interaction network; Function and pathway enrichment analysis AB To identify the typically expressed miRNAs in squamous cell carcinoma (SCC) and adenocarcinoma (ADC) of esophagus cancer and their target genes, and explore the related functions and pathways, providing potential biomarkers for esophageal carcinoma diagnosis and treatment. Gene expression profile GSE13937 was downloaded from Gene Expression Omnibus database which includes 152 samples, paired non-cancerous and cancerous, 44 SCC cases and 32 ADC cases; the differentially expressed miRNAs were identified with limma packages in R language after the data were normalized. Selected differentially expressed miRNAs were further analyzed using bioinformatics methods. Firstly, verified targets of miRNAs in two miRNA databases: miRecods and miRTarBase were integrated to select the targets genes of differentially expressed miRNAs. Next, String software was used to construct the target genes interaction network. Finally, function and pathway enrichment analysis of genes in the interaction network was carried out with Gestalt software. Up-regulated hsa-miR-21 and down-regulated hsamiR- 203 were identified by comparing normal and cancer tissue samples, and the targets genes regulated by these two miRNAs were most significantly related to cell cycle function and pathway, especially in the phase of G1/S. The two differentially expressed miRNA: hsa-miR-21 and hsa-miR-203 provide evidence for early diagnosis and treatment of esophageal carcinoma. The functions and pathways of target genes shows that deep understanding of cell cycle G1/S will help to illustrate the relationship between cell cycle regulation and pathogenesis of esophageal cancer. C1 [Wang, Bin Hong] Fengxian District Central Hospital, Sixth People’s Hospital in Southern Branch, Affiliated to Shanghai Jiao Tong University, Department of Cardiac-Thoracic Surgery, 201499 Shanghai, China. [Jiang, Biao Zhi] Fengxian District Central Hospital, Sixth People’s Hospital in Southern Branch, Affiliated to Shanghai Jiao Tong University, Department of Cardiac-Thoracic Surgery, 201499 Shanghai, China. [Li, Min] Fengxian District Central Hospital, Sixth People’s Hospital in Southern Branch, Affiliated to Shanghai Jiao Tong University, Department of Cardiac-Thoracic Surgery, 201499 Shanghai, China. RP Li, M (reprint author), Fengxian District Central Hospital, Sixth People’s Hospital in Southern Branch, Affiliated to Shanghai Jiao Tong University, Department of Cardiac-Thoracic Surgery, 201499 Shanghai, China. CR Enzinger PC, Mayer RJ, 2003, Esophageal cancer. N Engl J Med 349(23):2241–2252 Mathe EA, Nguyen GH, Bowman ED, Zhao Y, Budhu A, Schetter AJ, Braun R, Reimers M, Kumamoto K, Hughes D, 2009, MicroRNA expression in squamous cell carcinoma and adenocarcinoma of the esophagus: associations with survival. Clin Cancer Res 15(19):6192–6200 Enzinger PC, Mayer RJ, 2008, Oesophageal cancer. In: Baert A, ed, Encyclopedia of diagnostic imaging. Springer, Berlin, pp 1401– 1401., DOI 10.1007/978-3-540-35280-8_1772 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116(2):281–297., DOI 10.1016/S0092-8674(04)00045-5 Zhao Y, Srivastava D, 2007, A developmental view of microRNA function. Trends Biochem Sci 32(4):189 Zhang B, Pan X, Cobb GP, Anderson TA, 2007, microRNAs as oncogenes and tumor suppressors. Dev Biol 302(1):1–12 Feber A, Xi L, Luketich JD, Pennathur A, Landreneau RJ, Wu M, Swanson SJ, Godfrey TE, Litle VR, 2008, MicroRNA expression profiles of esophageal cancer. J Thorac Cardiovasc Surg 135(2):255–260 YeY,Wang KK,Gu J,Yang H, Lin J,Ajani JA,Wu X(2008, Genetic variations inmicroRNA-related genes are novel susceptibility loci for esophageal cancer risk. Cancer Prev Res 1(6):460–469 YangM, Liu R, Sheng J, Liao J,Wang Y, Pan E, GuoW, Pu Y, Yin L, 2013, Differential expression profiles of microRNAs as potential biomarkers for the early diagnosis of esophageal squamous cell carcinoma. Oncol Rep 29(1):169–176 Hiyoshi Y, Kamohara H, Karashima R, Sato N, Imamura Y, Nagai Y, Yoshida N, Toyama E, Hayashi N, Watanabe M, 2009, MicroRNA- 21 regulates the proliferation and invasion in esophageal squamous cell carcinoma. Clin Cancer Res 15(6):1915–1922 Kano M, Seki N, Kikkawa N, Fujimura L, Hoshino I, Akutsu Y, Chiyomaru T, Enokida H, Nakagawa M, Matsubara H, 2010, miR– 145, miR–133a and miR–133b: tumor–suppressive miRNAs target FSCN1 in esophageal squamous cell carcinoma. Int J Cancer 127(12):2804–2814 Sonkoly E,Wei T, Lorie EP, Suzuki H, KatoM, Torma H, Stahle M, Pivarcsi A, 2009, Protein kinase C-dependent upregulation of miR- 203 induces the differentiation of human keratinocytes. J Investig Dermatol 130(1):124–134 Troyanskaya O, CantorM, Sherlock G, Brown P, Hastie T, Tibshirani R, Botstein D, Altman RB, 2001, Missing value estimation methods for DNA microarrays. Bioinformatics 17(6):520–525 Fujita A, Sato J, Rodrigues L, Ferreira C, Sogayar M, 2006, Evaluating different methods of microarray data normalization. BMC Bioinforma 7(1):469 Gentleman R, Carey V, Huber W, Irizarry R, Dudoit S, 2005, Bioinformatics and computational biology solutions using R and Bioconductor, vol 746718470. Springer, New York BenjaminiY, HochbergY, 1995, Controlling the false discovery rate: a practical and powerful approach tomultiple testing. J R Stat Soc Ser B, Methodological, 289–300 Xiao F, Zuo Z, Cai G, Kang S, Gao X, Li T, 2009, miRecords: an integrated resource for microRNA–target interactions. Nucleic Acids Res 37(suppl 1):D105–D110 Hsu S-D, Lin F-M, Wu W-Y, Liang C, HuangW-C, Chan W-L, Tsai W-T, Chen G-Z, Lee C-J, Chiu C-M, 2011, miRTarBase: a database curates experimentally validated microRNA–target interactions. Nucleic Acids Res 39(suppl 1):D163–D169 Li S,ArmstrongCM, Bertin N,GeH,Milstein S, BoxemM,Vidalain P-O, Han J-DJ, Chesneau A, Hao T, 2004, A map of the interactome network of the metazoan C. elegans. Sci Signal 303(5657):540 Szklarczyk D, Franceschini A, Kuhn M, Simonovic M, Roth A, Minguez P, Doerks T, Stark M, Muller J, Bork P, 2011, The STRING database in 2011: functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res 39(suppl 1):D561–D568 Nam D, Kim S-Y, 2008, Gene-set approach for expression pattern analysis. Brief Bioinform 9(3):189–197 Zhang B, Kirov S, Snoddy J, 2005)WebGestalt: an integrated system for exploring gene sets in various biological contexts. Nucleic Acids Res 33(suppl 2):W741–W748 Duncan D, Prodduturi N, Zhang B, 2010, WebGestalt2: an updated and expanded version of the Web-based Gene Set Analysis Toolkit. BMC Bioinforma 11(Suppl 4):P10 Ec L, 2002, MicroRNAs are complementary to 3′UTR sequence motifs that mediate negative post-transcriptional regulation. Nat Genet 30(4):363–364 Fassan M, Realdon S, Pizzi M, Balistreri M, Battaglia G, Zaninotto G, Ancona E, RuggeM(2012, Programmed cell death 4 nuclear loss and miR–21 or activated Akt overexpression in esophageal squamous cell carcinogenesis. Dis Esophagus 25(3):263–268 Zhang J-g, Wang J-j, Zhao F, Liu Q, Jiang K, Yang G-h, 2010, MicroRNA-21, miR-21, represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer, NSCLC). Clin Chim Acta 11–12:846–852., DOI 10.1016/j.cca.2010.02.074 Xiong B, Cheng Y, Ma L, Zhang C, 2013, MiR-21 regulates biological behavior through the PTEN/PI-3 K/Akt signaling pathway in human colorectal cancer cells. Int J Oncol 42(1):219–228 Y-a Z, Zhang T, Zhao J-B, X-p W, Jiang T, Gu Z-P, X-n W, X-f L, 2010, The adenovirus-mediated transfer of PTEN inhibits the growth of esophageal cancer cells in vitro and in vivo. Biosci Biotechnol Biochem 74(4):736–740 Yuan T, Cantley L, 2008, PI3K pathway alterations in cancer: variations on a theme. Oncogene 27(41):5497–5510 Ofir M, Ben-Shachar B, Hacohen D, Ginsberg D, 2012, Abstract A17: MiR-15,miR-16 and miR-21 are direct transcriptional targets of E2F1 that limit E2F-induced proliferation and apoptosis. Cancer Res 72(2 Supplement):A17–A17 Schuldt A, 2011, Cell cycle: E2F1 ensures the endocycle. Nat Rev Mol Cell Biol 12(12):768–769 Furuta M, K-i K, Tanaka S, Arii S, Imoto I, Inazawa J, 2010, miR-124 and miR-203 are epigenetically silenced tumor-suppressive microRNAs in hepatocellular carcinoma. Carcinogenesis 31(5):766–776 Bian K, Fan J, Zhang X, Yang X-W, Zhu H-Y, Wang L, Sun J-Y, Meng Y-L, Cui P-C, Cheng S-Y, 2012, MicroRNA-203 leads to G1 phase cell cycle arrest in laryngeal carcinoma cells by directly targeting survivin. FEBS Lett 586(6):804–809 Bueno MJ, Perez de Castro I, Gomez de Cedron M, Santos J, Calin GA, Cigudosa Juan C, Croce CM, Fernandez-Piqueras J,Malumbres M, 2008, Genetic and epigenetic silencing of MicroRNA-203 enhances ABL1 and BCR-ABL1 oncogene expression. Cancer Cell 13(6):496–506., DOI 10.1016/j.ccr.2008.04.018 Cunningham J, Vierkant R, Sellers T, Phelan C, Rider D, Liebow M, Schildkraut J, Berchuck A, Couch F,Wang X, 2009, Cell cycle genes and ovarian cancer susceptibility: a tagSNP analysis. Br J Cancer 101(8):1461–1468 Lena A, Shalom-Feuerstein R, di Val Cervo PR, Aberdam D, Knight R, Melino G, Candi E, 2008, miR-203 represses ‘stemness’ by repressing ΔNp63. Cell Death Differ 15(7):1187–1195 Lau CPY, Ng PKS, Li MS, Tsui SKW, Huang L, Kumta SM(2013, p63 regulates cell proliferation and cell cycle progression-associated genes in stromal cells of giant cell tumor of the bone. Int J Oncol 42(2):437 Mishina T, Dosaka-Akita H, Hommura F, Nishi M, Kojima T, Ogura S, Shimizu M, Katoh H, Kawakami Y, 2000, Cyclin E expression, a potential prognostic marker for non-small cell lung cancers. Clin Cancer Res 6(1):11–16 Blagosklonny MV, Pardee AB, 2001, Exploiting cancer cell cycling for selective protection of normal cells. Cancer Res 61(11):4301– 4305 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 245 EP 252 DI 10.1007/s12253-013-9688-z PG 8 ER PT J AU Kawakita, A Yanamoto, S Yamada, Shi Naruse, T Takahashi, H Kawasaki, G Umeda, M AF Kawakita, Akiko Yanamoto, Souichi Yamada, Shin-ichi Naruse, Tomofumi Takahashi, Hidenori Kawasaki, Goro Umeda, Masahiro TI MicroRNA-21 Promotes Oral Cancer Invasion via the Wnt/β-Catenin Pathway by Targeting DKK2 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MiR-21; Oral tongue squamous cell carcinoma; DKK2; β-catenin; Tumor invasion ID MiR-21; Oral tongue squamous cell carcinoma; DKK2; β-catenin; Tumor invasion AB MicroRNA-21 (miR-21) is overexpressed in a wide variety of cancers and has been related to cellular proliferation, apoptosis, and invasion; however, the function of miR-21 is unknown in oral tongue squamous cell carcinoma (OTSCC). The purpose of this study was to examine miR-21 expression in OTSCC, correlate it with clinicopathological factors, and investigate its contribution to OTSCC cell invasion. MiR-21 expression in 79 primary OTSCCs was evaluated using locked nucleic acid in situ hybridization, and correlation was examined with the clinicopathological factors. To determine the miR-21 target, we searched for molecular genes involved in tumor invasion using the commonly cited prediction program miRanda. In an OTSCC cell line, SCC25 cells, we further evaluated whether miR-21 contributes to cell invasiveness by blocking its expression with a specific knockdown LNA probe and confirmed the direct target by Matrigel invasion assay and Western blotting. MiR-21 overexpression was detected in 60 of 79 cases (75.9 %) and correlated with the pattern of invasion (P =0.016). We selected DKK2 as a Wnt/antagonist involved in tumor invasion. MiR-21 overexpression was significantly correlated with the DKK2-/β-catenin- immunohistochemical phenotype. Knockdown ofmiR-21 significantly decreased the invasion potential of SCC25 cells with up-regulated DKK2. It was found that miR-21 is overexpressed and associated with tumor invasion in OTSCC, and that miR-21 promotes OTSCC cell invasion via the Wnt/β-catenin pathway by targeting DKK2 in vitro. These results suggest that miR-21 may be a potential therapeutic target for OTSCC treatment. C1 [Kawakita, Akiko] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yanamoto, Souichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yamada, Shin-ichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Naruse, Tomofumi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Takahashi, Hidenori] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Kawasaki, Goro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Umeda, Masahiro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. RP Yanamoto, S (reprint author), Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 852-8588 Nagasaki, Japan. EM syana@nagasaki-u.ac.jp CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics 2011. CA Cancer J Clin 61:69–90 Warnakulasuriya S, 2009, Global epidemiology of oral and oropharyngeal cancer. Oral Oncol 45:309–316 Rogers NS, Brown SJ,Woolgar AJ, Lowe D, Magennis P, Shaw JR, Sutton D, Errington D, Vaughan D, 2009, Survival following primary surgery for oral cancer. Oral Oncol 45:201–211 Yanamoto S, Kawasaki G, Yoshitomi I, Mizuno A(2002, P53, mdm2, and p21 expression in oral squamous cell carcinomas: relationship with clinicopathologic factors. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 94:593–600 Sessions GD, Lenox J, Spector JG, Chao C, Chaudry AO, 2003, Analysis of treatment results for base of tongue cancer. Laryngoscope 113:1252–1261 Kurokawa H, Zhang M, Matsumoto S, Yamashita Y, Tomoyose T, Tanaka T, Fukuyama H, Takahashi T, 2005, The high prognostic value of the histologic grade at the deep invasive front of tongue squamous cell carcinoma. J Oral Pathol Med 34:329–333 Bryne M, Koppang SH, Lilleng R, Kjaerheim A, 1992, Malignacy grading of the deep invasive margins of oral squamous cell carcinoma has high prognostic value. J Pathol 166:375–381 Bryne M, Jenssen N, Boysen M, 1995, Histologic grading in the deep invasive front of T1 and T2 glottic squamous cell carcinomas has high prognostic value. Virchows Arch 427:277–281 Bartel PD, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281–297 Hannon JG, Rossi JJ, 2004, Unlocking the potential of the human genome with RNA interference. Nature 431:371–378 Esquela-Kerscher A, Slack JF, 2006, Oncomirs-microRNAs with a role in cancer. Nat Rev Cancer 6:259–269 Calin AG, Croce MC, 2006, MicroRNA signatures in human cancers. Nat Rev Cancer 6:857–866 Wu HB, Xiong PX, Jia J, Zhang FW, 2011, MicroRNAs: new actors in the oral cancer scene. Oral Oncol 47:314–319 Volinia S, Calin AG, Liu GC, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, Prueitt LR, Yanaihara N, Lanza G, Scarpa A, Vecchione A, Negrini M, Harris CC, Croce MC, 2006, A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA 103:2257–2261 Krichevsky MA, Gabriely G, 2009, MiR-21: a small multi-faceted RNA. J Cell Mol Med 13:39–53 Fu X, Han Y, Wu Y, Zhu X, Lu X, Mao F, Wang X, He X, Zhao Y, Zhao Y, 2011, Prognostic role of microRNA-21 in various carcinomas: a systematic review and meta-analysis. Eur J Clin Invest 41: 1245–1253 Yan XL, Huang FX, Shao Q, Huang YM, Deng L,Wu LQ, Zeng XY, Shao YJ, 2008, MicroRNA miR-21 overexpression in human breast cancer is associated with advanced clinical stage, lymph node metastasis and patient poor prognosis. RNA 14:2348–2360 HwangHJ,Voortman J,Giovannetti E, SteinbergMS, Leon GL, Kim YT, Funel N, Park KJ, Kim AM, Kang HG, KimWS, Del ChiaroM, Peters JG, Giaccone G, 2010, Identification of microRNA-21 as a biomarker for chemoresistance and clinical outcome following adjuvant therapy in resectable pancreatic cancer. PLoS ONE 5:e10630 Asangani AI, Rasheed AS, Nikolova AD, Leupold HJ, Colburn HN, Post S, Allgayer H, 2008, MicroRNA-21, miR-21, posttranscriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 27:2128–2136 Reis PP, Tomenson M, Cervigne KN, Machado J, Jurisica I, Pintilie M, Sukhai AM, Perez-Ordonez B, Grenman R, GilbertWR, Gullane JP, Irish CJ, Kamel-Reid S, 2010, Programmed cell death 4 loss increases tumor cell invasion and is regulated by miR-21 in oral squamous cell carcinoma. Mol Cancer 9:238 Zhang GB, Li FJ, Yu QB, Zhu GZ, Liu YB, Yan M, 2012, MicroRNA-21 promotes tumor proliferation and invasion in gastric cancer by targeting PTEN. Oncol Rep 27:1019–1026 Han L,YueX, Zhou X, LanMF, YouG,ZhangW, Zhang LK,Cheng QJ,Yu ZS, Pu YP, Jiang T, Kang SC, 2012, MicroRNA-21 expression is regulated by β-catenin/STAT3 pathway and promotes glioma cell invasion by direct targeting RECK. CNS Neurosci Ther 18:573–583 Li T, Li SR, Li HY, Zhong S, Chen YY, ZhangMC, HuMM, Shen JZ, 2012, MiR-21 as an independent biochemical recurrence predictor and potential therapeutic target for prostate cancer. J Urol 187:1466– 1472 Yanamoto S, Kawasaki G, Yoshitomi I, Iwamoto T, Hirata K, Mizuno A, 2007, Clinicopathologic significance of EpCAM expression in squamous cell carcinoma of the tongue and its possibility as a potential target for tongue cancer gene therapy. Oral Oncol 43:869–877 Li J, Huang H, Sun L, Yang M, Pan C, ChenW, Wu D, Lin Z, Zeng C, Yao Y, Zhang P, Song E, 2009, MiR-21 indicates poor prognosis in tongue squamous cell carcinomas as an apoptosis inhibitor. Clin Cancer Res 15:3998–4008 Moriyama T, Ohuchida K, Mizumoto K, Yu J, Sato N, Nabae T, Takahata S, Toma H, Nagai E, Tanaka M, 2009, MicroRNA-21 modulates biological functions of pancreatic cancer cells including their proliferation, invasion, and chemoresistance. Mol Cancer Ther 8:1067–1074 Zheng J,XueH,Wang T, JiangY,LiuB, Li J,LiY,WangW, ZhangB, Sun M, 2011, MiR-21 downregulates the tumor suppressor p12CDK2AP1 and stimulates cell proliferation and invasion. J Cell Biochem 112:872–880 Ren J, Zhu D, LiuM, Sun Y, Tian L, 2010, Downregulation of miR- 21 modulates Ras expression to promote apoptosis and suppress invasion of laryngeal squamous cell carcinoma. Eur J Cancer 46: 3409–3416 Liu LZ, Wang H, Liu J, Wang XZ, 2013, MicroRNA-21, miR-21, expression promotes growth,metastasis, and chemo- or radioresistance in non-small cell lung cancer cells by targeting PTEN. Mol Cell Biochem 374:35–45 HanM, LiuM, Wang Y, Chen X, Xu J, SunY, Zhao L,QuH, Fan Y, Wu C, 2012, Antagonism of miR-21 reverses epithelial-mesenchymal transition and cancer stem cell phenotype through AKT/ERK1/2 inactivation by targeting PTEN. PLoS ONE 7:e39520 Krupnik EV, Sharp DJ, Jiang C, Robison K, Chickering WT, Amaravadi L, Brown ED, Guyot D, Mays G, Leiby K, Chang B, Duong T, Goodeari DA, Gearing PD, Sokol YS, MaCarthy AS, 1999, Functional and structural diversity of the human Dickkopf gene family. Gene 238:301–313 Mao B, Niehrs C, 2003, Kremen2 modulates Dickkopf2 activity during Wnt/LRP6 signaling. Gene 302:179–183 Zhu J, Zhang S, Gu L, Di W, 2012, Epigenetic silencing of DKK2 and Wnt signal pathway components in human ovarian carcinoma. Carcinogenesis 33:2334–2343 Hirata H, Hinoda Y, Nakajima K, Kawamoto K, Kikuno N, Kawakami K, Yamamura S, Ueno K, Majid S, Saini S, Ishii N, Dahiya R, 2009, Wnt antagonist gene DKK2 is epigenetically silenced and inhibits renal cancer progression through apoptotic and cell cycle pathways. Clin Cancer Res 15:5678–5687 Clvers H, Nusse R, 2012)Wnt/β-catenin signaling and disease. Cell 149:1192–1205 Betel D, KoppalA,Agius P, Sander C, Leslie C, 2010, Comprehensive modeling of microRNA targets predicts functional non-conserved and non-canonical sites. Genome Biol 11:R90 Yang HC, Yue J, Fan M, Pfeffer ML, 2010, IFN induces miR-21 through a signal transducer and activator of transcription 3-dependent pathway as a suppressive negative feedback on IFN-induced apoptosis. Cancer Res 70:8108–8116 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 253 EP 261 DI 10.1007/s12253-013-9689-y PG 9 ER PT J AU Lan, YJ Chen, H Chen, JQ Lei, QH Zheng, M Shao, ZR AF Lan, Yan-Ju Chen, Huan Chen, Jia-Qi Lei, Qiu-Hua Zheng, Min Shao, Zhe-Ren TI Immunolocalization of Vimentin, Keratin 17, Ki-67, Involucrin, β-Catenin and E-Cadherin in Cutaneous Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Squamous cell carcinoma; Epithelial-mesenchymal transition; Immunolocalization ID Squamous cell carcinoma; Epithelial-mesenchymal transition; Immunolocalization AB Skin squamous cell carcinoma (SCC) is a subtype of very aggressive skin cancers. To investigate if epithelialmesenchymal transition (EMT), a process for epitheloid cells losing their polarity and cohesiveness and transform into spindle-shaped cells, occurs in skin SCC. By using immunofluorescence, we defined the immunolocalization of vimentin, Keratin 17, β-catenin, E-cadherin, Ki-67 and involucrin, in SCC samples. Our results show reduced activity of involucrin and E-cadherin, and increased expression of Ki-67, β-catenin, Keratin 17 and vimentin in SCC. These data propose that EMT really occurs in poorly differentiated SCC and keratin 17 and involucrin may be another two biomarkers for EMT. C1 [Lan, Yan-Ju] Lishui Central Hospital, Department of DermatologyLishui, China. [Chen, Huan] Lishui Central Hospital, Department of DermatologyLishui, China. [Chen, Jia-Qi] Zhejiang University School of Medicine, Second Affiliated Hospital, Department of DermatologyHangzhou, China. [Lei, Qiu-Hua] Zhejiang University School of Medicine, Second Affiliated Hospital, Department of DermatologyHangzhou, China. [Zheng, Min] Zhejiang University School of Medicine, Second Affiliated Hospital, Department of DermatologyHangzhou, China. [Shao, Zhe-Ren] Zhejiang University School of Medicine, Second Affiliated Hospital, Department of Plastic Surgery, 88 Jiefang Rd, 310009 Hangzhou, China. RP Shao, ZR (reprint author), Zhejiang University School of Medicine, Second Affiliated Hospital, Department of Plastic Surgery, 310009 Hangzhou, China. EM shaozju@gmail.com CR Thiery JP, Sleeman JP, 2006, Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol 7(2): 131–142., DOI 10.1038/nrm1835 Said NA, Williams ED, 2011, Growth factors in induction of epithelial-mesenchymal transition and metastasis. Cells Tissues Organs 193(1–2):85–97., DOI 10.1159/000320360 Kalluri R, Neilson EG, 2003, Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 112(12):1776–1784., DOI 10. 1172/JCI20530 Hugo H, Ackland ML, Blick T, Lawrence MG, Clements JA, Williams ED, Thompson EW, 2007, Epithelial–mesenchymal and mesenchymal–epithelial transitions in carcinoma progression. J Cel Physiol 213(2):374–383., DOI 10.1002/jcp.21223 Kalluri R, Weinberg RA, 2009, The basics of epithelial-mesenchymal transition. J Clin Invest 119(6):1420–1428., DOI 10.1172/JCI39104 Acloque H, Adams MS, Fishwick K, Bronner-Fraser M, Nieto MA, 2009, Epithelial-mesenchymal transitions: the importance of changing cell state in development and disease. J Clin Invest 119(6):1438– 1449., DOI 10.1172/JCI38019 ZeisbergM, Neilson EG, 2009, Biomarkers for epithelial-mesenchymal transitions. J Clin Invest 119(6):1429–1437., DOI 10.1172/JCI36183 Mani SA, GuoW, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J,Weinberg RA(2008, The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133(4): 704–715., DOI 10.1016/j.cell.2008.03.027 Man XY, Finnson KW, BaronM, Philip A, 2012, CD109, a TGF-beta co-receptor, attenuates extracellular matrix production in scleroderma skin fibroblasts. Arthritis Res Ther 14(3):R144., DOI 10.1186/ar3877 Marks R, 1996, Squamous cell carcinoma. Lancet 347(9003):735–738 Gerdes J, Lemke H, Baisch H, Wacker HH, Schwab U, Stein H, 1984, Cell cycle analysis of a cell proliferation-associated human nuclear antigen defined by the monoclonal antibody Ki-67. J Immunol 133(4):1710–1715 Scholzen T, Gerdes J, 2000, The Ki-67 protein: from the known and the unknown. J Cel Physiol 182(3):311–322., DOI 10.1002/(SICI, 1097-4652(200003)182:3<311::AID-JCP1>3.0.CO;2-9 Pich A, Chiusa L, Navone R, 2004, Prognostic relevance of cell proliferation in head and neck tumors. Ann Oncol: official journal of the European Society for Medical Oncology/ESMO 15(9):1319– 1329., DOI 10.1093/annonc/mdh299 Iyer PV, Leong AS, 1992, Poorly differentiated squamous cell carcinomas of the skin can express vimentin. J Cutan Pathol 19(1):34–39 ChangHW, Lee YS, Nam HY, HanMW, Kim HJ,Moon SY, Jeon H, Park JJ, Carey TE, Chang SE, Kim SW, Kim SY, 2012, Knockdown of beta-catenin controls both apoptotic and autophagic cell death through LKB1/AMPK signaling in head and neck squamous cell carcinoma cell lines. Cell Signal., DOI 10.1016/j.cellsig.2012.12.020 Depianto D, Kerns ML, Dlugosz AA, Coulombe PA, 2010, Keratin 17 promotes epithelial proliferation and tumor growth by polarizing the immune response in skin. Nat Genet 42(10):910–914., DOI 10. 1038/ng.665 Proby CM, Churchill L, Purkis PE, GloverMT, Sexton CJ, Leigh IM, 1993, Keratin 17 expression as a marker for epithelial transformation in viral warts. Am J Pathol 143(6):1667–1678 Wei KJ, Zhang L, Yang X, Zhong LP, Zhou XJ, Pan HY, Li J, Chen WT, Zhang ZY, 2009, Overexpression of cytokeratin 17 protein in oral squamous cell carcinoma in vitro and in vivo. Oral Dis 15(1): 111–117., DOI 10.1111/j.1601-0825.2008.01501.x Toyoshima T, Koch F, Kaemmerer P, Vairaktaris E, Al-Nawas B, Wagner W, 2009, Expression of cytokeratin 17 mRNA in oral squamous cell carcinoma cells obtained by brush biopsy: preliminary results. J Oral Pathol Med: official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology 38(6):530–534., DOI 10.1111/j.1600-0714.2009.00748.x Kitamura R, Toyoshima T, Tanaka H, Kawano S, Kiyosue T, Matsubara R, Goto Y, Hirano M, Oobu K, Nakamura S, 2012, Association of cytokeratin 17 expression with differentiation in oral squamous cell carcinoma. J Cancer Res Clin Oncol 138(8):1299– 1310., DOI 10.1007/s00432-012-1202-6 Rice RH, Green H, 1977, The cornified envelope of terminally differentiated human epidermal keratinocytes consists of crosslinked protein. Cell 11(2):417–422 Bhargava G, Rifas L, Makman MH, 1979, Presence of epidermal growth factor receptors and influence of epidermal growth factor on proliferation and aging in cultured smooth muscle cells. J Cell Physiol 100(2):365–374., DOI 10.1002/jcp.1041000217 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 263 EP 266 DI 10.1007/s12253-013-9690-5 PG 4 ER PT J AU Orsini, G Legitimo, A Failli, A Ferrari, P Nicolini, A Spisni, R Miccoli, P Consolini, R AF Orsini, Giulia Legitimo, Annalisa Failli, Alessandra Ferrari, Paola Nicolini, Andrea Spisni, Roberto Miccoli, Paolo Consolini, Rita TI Quantification of Blood Dendritic Cells in Colorectal Cancer Patients During the Course of Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Circulating dendritic cells; Colorectal cancer; Immunosuppression; Flow cytometry ID Circulating dendritic cells; Colorectal cancer; Immunosuppression; Flow cytometry AB Colorectal cancer is a malignancy with poor prognosis that might be associated with defective immune function. The aim of the present study was to investigate circulating dendritic cells in colorectal cancer patients, in order to contribute to elucidate tumor-escape mechanisms and to point out a possible correlation with the clinical condition of the disease. Therefore, we enumerated ex vivo myeloid and plasmacytoid dendritic cells, through multicolor flow cytometry, in 26 colorectal patients and 33 healthy controls. Furthermore we performed several analyses at determined time points in order to define the immunological trend of cancer patients after surgery and other conventional treatments. At the preoperative time point the absolute number of plasmacytoid dendritic cells in cancer patients was significantly reduced in comparison to controls, this result being mainly referred to stage III-IV patients. The number of myeloid dendritic cells did not show any significant difference compared to healthy controls; interestingly the expression of the tolerogenic antigen CD85k was significantly higher on cancer patients’ myeloid dendritic cells than controls’. At the following samplings, circulating dendritic cell absolute number did not show any difference compared to controls. Conclusively the impairment of the number of circulating dendritic cells may represent one of the tumor escape mechanisms occurring in colorectal cancer. These alterations seem to be correlated to cancer progression. Our work sheds light on one of dendritic cell-based tumor immune escape mechanisms. This knowledge may be useful to the development of more effective immunotherapeutic strategies. C1 [Orsini, Giulia] University of Pisa, Department of Experimental and Clinical Medicine, via Roma 67, 56126 Pisa, Italy. [Legitimo, Annalisa] University of Pisa, Department of Experimental and Clinical Medicine, via Roma 67, 56126 Pisa, Italy. [Failli, Alessandra] Azienda Ospedaliero-Universitaria PisanaPisa, Italy. [Ferrari, Paola] University of Pisa, Department of Experimental and Clinical Medicine, via Roma 67, 56126 Pisa, Italy. [Nicolini, Andrea] University of Pisa, Department of Experimental and Clinical Medicine, via Roma 67, 56126 Pisa, Italy. [Spisni, Roberto] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical AreaPisa, Italy. [Miccoli, Paolo] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical AreaPisa, Italy. [Consolini, Rita] University of Pisa, Department of Experimental and Clinical Medicine, via Roma 67, 56126 Pisa, Italy. RP Orsini, G (reprint author), University of Pisa, Department of Experimental and Clinical Medicine, 56126 Pisa, Italy. EM giulia.orsini@for.unipi.it;gl84@hotmail.it CR Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Galon J, Fridman WH, Pages F, 2007, The adaptive immunologic microenvironment in colorectal cancer: a novel perspective. Cancer Res 67:1883–1886 Dalerba P, Maccalli C, Casati C, Castelli C, Parmiani G, 2003, Immunology and immunotherapy of colorectal cancer. Crit Rev Oncol Hematol 46:33–57 Roxburgh CS, McMillan DC, 2012, The role of the in situ local inflammatory response in predicting recurrence and survival in patients with primary operable colorectal cancer. Cancer Treat Rev 38: 451–466 Dadabayev AR, Sandel MH, Menon AG, Morreau H, Melief CJ, Offringa R, van der Burg SH, Janssen-van Rhijn C, Ensink NG, Tollenaar RA, van de Velde CJ, Kuppen PJ, 2004, Dendritic cells in colorectal cancer correlatewith other tumor-infiltrating immune cells. Cancer Immunol Immunother 53:978–986 Sandel MH, Dadabayev AR, Menon AG, Morreau H, Melief CJ, Offringa R, van der Burg SH, Janssen-van Rhijn CM, Ensink NG, Tollenaar RA, van de Velde CJ, Kuppen PJ, 2005, Prognostic value of tumor-infiltrating dendritic cells in colorectal cancer: role of maturation status and intratumoral localization. Clin Cancer Res 11: 2576–2582 Nagorsen D, Voigt S, Berg E, Stein H, Thiel E, Loddenkemper C, 2007, Tumor-infiltrating macrophages and dendritic cells in human colorectal cancer: relation to local regulatory T cells, systemic T-cell response against tumor-associated antigens and survival. J Transl Med 5:62 Schakel K, 2009, Dendritic cells–why can they help and hurt us. Exp Dermatol 18:264–273 Mortellaro A, Wong SC, Fric J, Ricciardi-Castagnoli P, 2010, The need to identify myeloid dendritic cell progenitors in human blood. Trends Immunol 31:18–23 Reizis B, 2010, Regulation of plasmacytoid dendritic cell development. Curr Opin Immunol 22:206–211 Palucka K, Ueno H, Zurawski G, Fay J, Banchereau J, 2010, Building on dendritic cell subsets to improve cancer vaccines. Curr Opin Immunol 22:258–263 Fricke I, Gabrilovich DI, 2006, Dendritic cells and tumor microenvironment: a dangerous liaison. Immunol Invest 35:459–483 Lenahan C, Avigan D, 2006, Dendritic cell defects in patients with cancer: mechanisms and significance. Breast Cancer Res 8:101 Mazzolini G, Murillo O, Atorrasagasti C, Dubrot J, Tirapu I, Rizzo M, Arina A, Alfaro C, Azpilicueta A, Berasain C, Perez-Gracia JL, Gonzalez A, Melero I, 2007, Immunotherapy and immunoescape in colorectal cancer. World J Gastroenterol 13:5822–5831 Della Bella S, Gennaro M, Vaccari M, Ferraris C, Nicola S, Riva A, Clerici M, Greco M, Villa ML, 2003, Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br J Cancer 89:1463–1472 Pinzon-Charry A, Maxwell T, Lopez JA, 2005, Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol 83:451–461 Ormandy LA, Farber A, Cantz T, Petrykowska S, Wedemeyer H, Horning M, Lehner F, Manns MP, Korangy F, Greten TF, 2006, Direct ex vivo analysis of dendritic cells in patients with hepatocellular carcinoma. World J Gastroenterol 12:3275–3282 Sakakura K, Chikamatsu K, Takahashi K, Whiteside TL, Furuya N, 2006, Maturation of circulating dendritic cells and imbalance of Tcell subsets in patients with squamous cell carcinoma of the head and neck. Cancer Immunol Immunother 55:151–159 McCarter MD, Baumgartner J, Escobar GA, Richter D, Lewis K, RobinsonW,Wilson C, Palmer BE, Gonzalez R, 2007, Immunosuppressive dendritic and regulatory Tcells are upregulated inmelanoma patients. Ann Surg Oncol 14:2854–2860 Sciarra A, Lichtner M, Autran GA,Mastroianni C, Rossi R,Mengoni F, Cristini C, Gentilucci A, Vullo V, Di Silverio F, 2007, Characterization of circulating blood dendritic cell subsets DC123+, lymphoid, and DC11C+, myeloid, in prostate adenocarcinoma patients. Prostate 67:1–7 Yamamoto T, Yanagimoto H, Satoi S, Toyokawa H, Yamao J, Kim S, Terakawa N, Takahashi K, Kwon AH, 2012, Circulating myeloid dendritic cells as prognostic factors in patients with pancreatic cancer who have undergone surgical resection. J Surg Res 173:299–308 Ambe K, Mori M, Enjoji M, 1989, S-100 protein-positive dendritic cells in colorectal adenocarcinomas. Distribution and relation to the clinical prognosis. Cancer 63:496–503 Nakayama Y, Inoue Y, Minagawa N, Katsuki T, Nagashima N, Onitsuka K, Tsurudome Y, Sako T, Hirata K, Nagata N, Itoh H, 2003, Relationships between S-100 protein-positive cells and clinicopathological factors in patients with colorectal cancer. Anticancer Res 23:4423–4426 Inoue Y, Nakayama Y, Minagawa N, Katsuki T, Nagashima N, Matsumoto K, Shibao K, Tsurudome Y, Hirata K, Nagata N, Itoh H, 2005, Relationship between interleukin-12-expressing cells and antigen-presenting cells in patients with colorectal cancer. Anticancer Res 25:3541–3546 Yuan A, Steigen SE, Goll R, Vonen B, Husbekk A, Cui G, Florholmen J, 2008, Dendritic cell infiltration pattern along the colorectal adenoma-carcinoma sequence. APMIS 116:445–456 Gulubova MV, Ananiev JR, Vlaykova TI, Yovchev Y, Tsoneva V, Manolova IM, 2012, Role of dendritic cells in progression and clinical outcome of colon cancer. Int J Colorectal Dis 27:159–169 Huang A, Gilmour JW, Imami N, Amjadi P, Henderson DC, Allen- Mersh TG, 2003, Increased serum transforming growth factor-beta1 in human colorectal cancer correlates with reduced circulating dendritic cells and increased colonic Langerhans cell infiltration. Clin Exp Immunol 134:270–278 Bellik L, Gerlini G, Parenti A, Ledda F, Pimpinelli N, Neri B, Pantalone D, 2006, Role of conventional treatments on circulating and monocyte-derived dendritic cells in colorectal cancer. Clin Immunol 121:74–80 Schmidt MA, Fortsch C, Schmidt M, Rau TT, Fietkau R, Distel LV, 2012, Circulating regulatory T cells of cancer patients receiving radiochemotherapy may be useful to individualize cancer treatment. Radiother Oncol 104:131–138 Orsini G, Legitimo A, Failli A,Massei F, Biver P, Consolini R, 2012, Enumeration of human peripheral blood dendritic cells throughout the life. Int Immunol 24:347–356 Cella M, Dohring C, Samaridis J, Dessing M, Brockhaus M, Lanzavecchia A, Colonna M, 1997, A novel inhibitory receptor, ILT3, expressed on monocytes, macrophages, and dendritic cells involved in antigen processing. J Exp Med 185:1743–1751 Vlad G, Chang CC, Colovai AI, Berloco P, Cortesini R, Suciu-Foca N, 2009, Immunoglobulin-like transcript 3: A crucial regulator of dendritic cell function. Hum Immunol 70:340–344 Ju XS, Hacker C, Scherer B, Redecke V, Berger T, Schuler G, Wagner H, Lipford GB, Zenke M, 2004, Immunoglobulin-like transcripts ILT2, ILT3 and ILT7 are expressed by human dendritic cells and down-regulated following activation. Gene 331:159–164 Penna G, Roncari A, Amuchastegui S, Daniel KC, Berti E, Colonna M, Adorini L, 2005, Expression of the inhibitory receptor ILT3 on dendritic cells is dispensable for induction of CD4+Foxp3+ regulatory T cells by 1,25-dihydroxyvitamin D3. Blood 106:3490–3497 Koos D, Josephs SF, Alexandrescu DT, Chan RC, Ramos F, Bogin V, Gammill V, Dasanu CA, De Necochea-Campion R, Riordan NH, Carrier E, 2010, Tumor vaccines in 2010: need for integration. Cell Immunol 263:138–147 Palucka K, Banchereau J, 2012, Cancer immunotherapy via dendritic cells. Nat Rev Cancer 12:265–277 Nencioni A, Grunebach F, Schmidt SM, Muller MR, Boy D, Patrone F, Ballestrero A, Brossart P, 2008, The use of dendritic cells in cancer immunotherapy. Crit Rev Oncol Hematol 65:191–199 Michielsen AJ, Hogan AE, Marry J, Tosetto M, Cox F, Hyland JM, Sheahan KD, O’Donoghue DP, Mulcahy HE, Ryan EJ, O’Sullivan JN, 2011, Tumour tissue microenvironment can inhibit dendritic cell maturation in colorectal cancer. PLoS One 6:e27944 Takahashi K, Toyokawa H, Takai S, Satoi S, Yanagimoto H, Terakawa N, Araki H, Kwon AH, Kamiyama Y, 2006, Surgical influence of pancreatectomy on the function and count of circulating dendritic cells in patients with pancreatic cancer. Cancer Immunol Immunother 55:775–784 Lou Y, Liu C, Kim GJ, Liu YJ, Hwu P,Wang G, 2007, Plasmacytoid dendritic cells synergize with myeloid dendritic cells in the induction of antigen-specific antitumor immune responses. J Immunol 178: 1534–1541 Hartmann E,Wollenberg B, Rothenfusser S,WagnerM,Wellisch D, Mack B, Giese T, Gires O, Endres S, Hartmann G, 2003, Identification and functional analysis of tumor-infiltrating plasmacytoid dendritic cells in head and neck cancer. Cancer Res 63:6478–6487 Perez-Cabezas B, Naranjo-Gomez M, Fernandez MA, Grifols JR, Pujol-Borrell R, Borras FE, 2007, Reduced numbers of plasmacytoid dendritic cells in aged blood donors. Exp Gerontol 42:1033–1038 Ratta M, Fagnoni F, Curti A, Vescovini R, Sansoni P, Oliviero B, Fogli M, Ferri E, Della Cuna GR, Tura S, Baccarani M, Lemoli RM, 2002, Dendritic cells are functionally defective in multiple myeloma: the role of interleukin-6. Blood 100:230–237 Brimnes MK, Vangsted AJ, Knudsen LM, Gimsing P, Gang AO, Johnsen HE, Svane IM(2010, Increased level of both CD4+FOXP3+ regulatory T cells and CD14+HLA-DR-/low myeloid-derived suppressor cells and decreased level of dendritic cells in patients with multiple myeloma. Scand J Immunol 72:540–547 Hoffmann TK, Muller-Berghaus J, Ferris RL, Johnson JT, Storkus WJ, Whiteside TL, 2002, Alterations in the frequency of dendritic cell subsets in the peripheral circulation of patients with squamous cell carcinomas of the head and neck. Clin Cancer Res 8:1787–1793 Yanagimoto H, Takai S, Satoi S, Toyokawa H, Takahashi K, Terakawa N, Kwon AH, Kamiyama Y, 2005, Impaired function of circulating dendritic cells in patients with pancreatic cancer. Clin Immunol 114:52–60 Wilkinson R, Kassianos AJ, Swindle P, Hart DN, Radford KJ, 2006, Numerical and functional assessment of blood dendritic cells in prostate cancer patients. Prostate 66:180–192 Suciu-Foca N, Cortesini R, 2007, Central role of ILT3 in the T suppressor cell cascade. Cell Immunol 248:59–67 Lissoni P, Malugani F, Bonfanti A, Bucovec R, Secondino S, Brivio F, Ferrari-Bravo A, Ferrante R, Vigore L, Rovelli F, Mandala M, Viviani S, Fumagalli L, Gardani GS, 2001, Abnormally enhanced blood concentrations of vascular endothelial growth factor, VEGF, in metastatic cancer patients and their relation to circulating dendritic cells, IL-12 and endothelin-1. J Biol Regul Homeost Agents 15:140– 144 Beckebaum S, Zhang X, Chen X, Yu Z, Frilling A, Dworacki G, Grosse-Wilde H, Broelsch CE, Gerken G, Cicinnati VR, 2004, Increased levels of interleukin-10 in serum from patients with hepatocellular carcinoma correlate with profound numerical deficiencies and immature phenotype of circulating dendritic cell subsets. Clin Cancer Res 10:7260–7269 Bennaceur K, Chapman JA, Touraine JL, Portoukalian J, 2009, Immunosuppressive networks in the tumour environment and their effect in dendritic cells. Biochim Biophys Acta 1795:16–24 Bindea G, Mlecnik B, Fridman WH, Galon J, 2011, The prognostic impact of anti-cancer immune response: a novel classification of cancer patients. Semin Immunopathol 33:335–340 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 267 EP 276 DI 10.1007/s12253-013-9691-4 PG 10 ER PT J AU Theocharis, S Klijanienko, J Giaginis, C Alexandrou, P Patsouris, E Sastre-Garau, X AF Theocharis, Stamatios Klijanienko, Jerzy Giaginis, Constantinos Alexandrou, Paraskevi Patsouris, Efstratios Sastre-Garau, Xavier TI Ephrin Receptor (Eph) -A1, -A2, -A4 and -A7 Expression in Mobile Tongue Squamous Cell Carcinoma: Associations with Clinicopathological Parameters and Patients Survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mobile tongue squamous cell carcinoma; Ephrin receptors; Clinicopathological parameters; Prognosis; Immunohistochemistry ID Mobile tongue squamous cell carcinoma; Ephrin receptors; Clinicopathological parameters; Prognosis; Immunohistochemistry AB Ephrin receptors (Ephs) are frequently overexpressed in a wide variety of human malignant tumors, being associated with tumor growth, invasion, metastasis and angiogenesis. The present study aimed to evaluate the clinical significance of Eph- A1, -A2, -A4 and -A7 protein expression in mobile tongue squamous cell carcinoma (SCC). Eph-A1, -A2, -A4 and -A7 protein expression was assessed immunohistochemically on 37 mobile tongue SCC tissue samples and was analyzed in relation with clinicopathological characteristics, overall and disease-free patients’ survival. All the examined mobile tongue SCC cases were found positive for Eph-A1, -A2, -A4 and -A7. Significant associations were noted between high Eph-A1, -A4 and -A7 expression and absence of lymph node metastases (p=0.0263, p=0.0461 and p=0.0461, respectively). High Eph-A1, -A2 and -A7 expression was significantly more frequently observed in patients presenting absence of vascular invasion (p =0.0444), dense stromal inflammatory reaction (p=0.0063) and female gender (p=0.0327), respectively. Mobile tongue SCC patients with high Eph-A7 expression presented longer overall and disease-free survival compared to those with low Eph-A7 expression (log-rank test, p=0.0093 and p=0.0164, respectively). In multivariate analysis, Eph-A7 expression was identified as independent prognostic factor of overall survival (Coxregression analysis, p=0.0426). The present study supported evidence that Ephs may participate in the malignant transformation of mobile tongue SCC, reinforcing their utility as clinical markers for patients’ management and prognosis, as also as targets for potential therapeutic intervention in tongue chemoprevention. C1 [Theocharis, Stamatios] Institut Curie, Department of PathologyParis, France. [Klijanienko, Jerzy] Institut Curie, Department of PathologyParis, France. [Giaginis, Constantinos] University of Athens, Medical School, Department of Pathology, 75 M. Asias str., Goudi, GR11527 Athens, Greece. [Alexandrou, Paraskevi] University of Athens, Medical School, Department of Pathology, 75 M. Asias str., Goudi, GR11527 Athens, Greece. [Patsouris, Efstratios] University of Athens, Medical School, Department of Pathology, 75 M. Asias str., Goudi, GR11527 Athens, Greece. [Sastre-Garau, Xavier] Institut Curie, Department of PathologyParis, France. RP Theocharis, S (reprint author), Institut Curie, Department of Pathology, Paris, France. EM theocharis@ath.forthnet.gr CR Zhang J, Hughes SE, 2006, Role of the ephrin and Ephrin receptor tyrosine kinase families in angiogenesis and development of the cardiovascular system. J Pathol 208:453–461 Pasquale EB, 2008, Eph-ephrin bidirectional signaling in physiology and disease. Cell 133:38–52 Nakamoto M, Bergemann AD, 2002, Diverse role for the Eph family of receptor tyrosine kinases in carcinogenesis. Microsc Res Tech 59: 58–67 Surawska H, Ma PC, Salgia R, 2004, The role of ephrins and Eph receptors in cancer. Cytokine Growth Factor Rev 15:419–433 Cheng N, Brantley DM, Chen J, 2002, The ephrins and Eph receptors in angiogenesis. Cytokine Growth Factor Rev 13:75–85 Brandley-SiedersDM, Chen J, 2007, Eph receptor tyrosine kinase in angiogenesis: from development to disease. Angiogenesis 7:17–28 Castano J, Davalos V, Schwartz S Jr, Arango D, 2008, EPH receptors in cancer. Histol Histopathol 23:1011–1023 IretonRC, Chen J, 2005, EphA2 receptor tyrosine kinase as a promising target for cancer therapeutics. Curr Cancer Drug Targets 5:149–157 Brantley-Sieders D, Schmidt S, Parker M, Chen J, 2004, Eph receptor tyrosine kinases in tumor and tumor microenvironment. Curr Pharm Des 10:3431–3442 HeroultM, Schaffner F,AugustinHG(2004, Eph receptor and ephrin ligand-mediated interactions during angiogenesis and tumor progression. Exp Cell Res 312:642–650 Canto MT, Devesa SS, 2002, Oral cavity and pharynx cancer incidence in the United States, 1975–1988. Oral Oncol 38:610–617 Jemal A, Siegel R,Ward E, Murray T, Xu J, Thun MJ, 2007, Cancer statistics. CA Cancer J Clin 57:43–66 Brenner H, 2002, Long-term survival rates of cancer patients achieved by the end of the 20th century: a period analysis. Lancet 360:1131–1135 Shiboski CH, Schmidt BL, Jordan RC, 2005, Tongue and tonsil carcinoma: increasing trends in the U.S. population ages 20–44 years. Cancer 103:1843–1849 Sano D, Myers JN, 2007, Metastasis of squamous cell carcinoma of the oral tongue. Cancer Metastasis Rev 26:645–662 Kantola S, Parikka M, Jokinen K et al, 2000, Prognostic factors in tongue cancer-relative importance of demographic, clinical and histopathological factors. Br J Cancer 83:614–619 Wangsa D, Ryott M, Avall-Lundquist E et al, 2008, Ki-67 expression predicts locoregional recurrence in stage I oral tongue carcinoma. Br J Cancer 99:1121–1128 Giaginis C, Tsourouflis G, Zizi-Serbetzoglou A, Kouraklis G, Chatzopoulou E, Theocharis S, 2010, Clinical significance of Ephrin, Eph)-A1, -A2, -A4, -A5 and -A7 receptors in pancreatic ductal adenocarcinoma. Pathol Oncol Res 16:267–276 Karidis NP, Giaginis C, Tsourouflis G, Alexandrou P, Delladetsima I, Theocharis S, 2011, Eph-A2 and Eph-A4 expression in human benign and malignant thyroid lesions: an immunohistochemical study. Med Sci Monit 17:BR257–BR265 Brandwein-Gensler M, Teixeira MS, Lewis CM et al, 2005, Oral squamous cell carcinoma: histological risk assessment, but not margin status, is strongly predictive of local disease-free and overall survival. Am J Surg Pathol 29:167–178 Shintani S, Matsura H, Hasegawa Y, Nakayama B, Fujimoto Y, 1997, The relationship of shape of tumor invasion to depth of invasion and cervical lymph node metastasis in squamous cell carcinoma of the tongue. Oncology 54:463–467 Barnes L, Eveson JW, Reichert P, Sidransky D, 2005, World Health Organization classifications tumours. Pathology and genetics of head and neck tumours. IARC Press, Lyon Po Wing Yen A, Lam KY, Lam LK et al, 2002, Prognostic factors clinically stage I and II oral tongue carcinoma: a comparative study of stage, thickness, shape, growth pattern, invasive front malignancy grading,Martinez-Gimeno score and pathologic features. Head Neck 24:513–520 Klijanienko J, el-Naggar AK, de Braud F, 1995, Tumor vascularization, mitotic index, histopathologic grade, and DNA ploidy in the assessment of 114 head and neck squamous cell carcinomas. Cancer 75:1649–1656 Theocharis S, Klijanienko J, Giaginis C et al, 2011, Histone deacetylase, HDAC)-1 and −2 expression in mobile tongue squamous cell carcinoma: associations with clinicopathological parameters and patients survival. J Oral Med Pathol 40:706– 714 Theocharis S, Klijanienko J, Giaginis C et al, 2011, Metallothionein expression in mobile tongue squamous cell carcinoma: associations with clinicopathological parameters and patients survival. Histopathology 59:514–525 Dong Y, Wang J, Sheng Z et al, 2009, Downregulation of EphA1 in colorectal carcinomas correlates with invasion and metastasis. Mod Pathol 22:151–160 Saito T, Masuda N,Miyazaki Tet al, 2004, Expression of EphA2 and E-cadherin in colorectal cancer: correlation with cancer metastasis. Oncol Rep 11:605–611 Abraham S, Knapp DW, Cheng L et al, 2006, Expression of EphA2 and Ephrin A-1 in carcinoma of the urinary bladder. Clin Cancer Res 12:353–360 HolmR,Knopp S, Suo Z, Trope C,Nesland JM(2007, Expression of EphA2 and EphrinA-1 in vulvar carcinomas and its relation to prognosis. J Clin Pathol 60:1086–1091 Hafner C, Becker B, LandthalerM, Vogt T, 2006, Expression profile of Eph receptors and ephrin ligands in human skin and downregulation of EphA1 in nonmelanoma skin cancer. Mod Pathol 19:1369–1377 Lin YG, Han LY, Kamat AA et al, 2007, EphA2 overexpression is associated with angiogenesis in ovarian cancer. Cancer 109:332–340 Yuan W, Chen Z, Wu S et al, 2009, Expression of EphA2 and Ecadherin in Gastric Cancer: correlated with Tumor Progression and Lymphogenous Metastasis. Pathol Oncol Res 15:473–478 Zeng G, Hu Z, Kinch MS et al, 2003, High-level expression of EphA2 receptor tyrosine kinase in prostatic intraepithelial neoplasia. Am J Pathol 163:2271–2276 Kinch MS, Moore MB, Harpole DH, 2003, Predictive value of the EphA2 receptor tyrosine kinase in lung cancer recurrence and survival. Clin Cancer Res 9:613–618 Shao Z, Zhang WF, Chen XM, Shang ZJ, 2008, Expression of EphA2 and VEGF in squamous cell carcinoma of the tongue: correlation with the angiogenesis and clinical outcome. Oral Oncol 44:1110–1117 Kamat AA, Coffey D, MerrittWMet al, 2009, EphA2 overexpression is associated with lack of hormone receptor expression and poor outcome in endometrial cancer. Cancer 115:2684–2692 Miyazaki T, Kato H, Fukuchi M, Nakajima M, Kuwano H, 2003, EphA2 overexpression correlates with poor prognosis in esophageal squamous cell carcinoma. Int J Cancer 103:657–663 Wang LF, Fokas E, Bieker M et al, 2008, Increased expression of EphA2 correlates with adverse outcome in primary and recurrent glioblastoma multiforme patients. Oncol Rep 19:151–156 Herrem CJ, Tatsumi T, Olson KS et al, 2005, Expression of EphA2 is prognostic of disease-free interval and overall survival in surgically treated patients with renal cell carcinoma. Clin Cancer Res 11:226–231 Oki M, Yamamoto H, Taniguchi H, Adachi Y, Imai K, Shinomura Y, 2008, Overexpression of the receptor tyrosine kinase EphA4 in human gastric cancers. World J Gastroenterol 14:5650–5656 Wang LF, Fokas E, Juricko J et al, 2008, Increased expression of EphA7 correlates with adverse outcome in primary and recurrent glioblastoma multiforme patients. BMC Cancer 8:79 Pasquale EB, 2010, Eph receptors and ephrins in cancer: bidirectional signalling and beyond. Nat Rev Cancer 10:165–180 Tandon M, Vemula SV, Mittal SK, 2011, Emerging strategies for EphA2 receptor targeting for cancer therapeutics. Expert Opin Ther Targets 15:31–51 Landen CN Jr, Chavez-Reyes A, Bucana C et al, 2005, Therapeutic EphA2 gene targeting in vivo using neutral liposomal small interfering RNA delivery. Cancer Res 65:6910–6918 Shahzad MM, Lu C, Lee JW et al, 2009, Dual targeting of EphA2 and FAK in ovarian carcinoma. Cancer Biol Ther 8:1027–1034 Zhuang G, Brantley-Sieders DM, Vaught D et al, 2010, Elevation of receptor tyrosine kinase EphA2 mediates resistance to trastuzumab therapy. Cancer Res 70:299–308 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 277 EP 284 DI 10.1007/s12253-013-9692-3 PG 8 ER PT J AU Li, J Yang, ZL Zou, Q Yuan, Y Li, J Liang, L Zhen, G Chen, S AF Li, Jiequn Yang, Zhu-Lin Zou, Qiong Yuan, Yuan Li, Jinghe Liang, Lufeng Zhen, Guixiang Chen, Senlin TI Squamous Cell/Adenosquamous Carcinomas and Adenocarcinomas of the Gallbladder: An Immunohistochemistry Study of Prognostic Markers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gallbladdercancer; Adenocarcinoma; Squamous cell carcinoma; Adenosquamous carcinoma; KRT19; hASH1; Prognosis; Metastasis ID Gallbladdercancer; Adenocarcinoma; Squamous cell carcinoma; Adenosquamous carcinoma; KRT19; hASH1; Prognosis; Metastasis AB Gallbladder cancers (GBCs) are highly aggressive and lethal diseases. However, the key molecular mechanisms responsible for the progression and prognosis of GBCs have not been identified. No biological markers for effectively identifying GBC subtypes have been reported. In this study the expression of keratin 19 (KRT19) and human achaetescute homolog 1 (hASH1) proteins in 46 squamous cell/ adenosquamous carcinomas (SC/ASC) and 80 adenocarcinomas (AC) were examined using immunohistochemistry. Negative KRT19 or positive hASH1 expression were significantly associated with lymph node metastasis, invasion and TNM stage of SC/ASC patients. In contrast, positive KRT19 and hASH1 expression were significantly associated with large tumor size, lymph metastasis, invasion, and TNM stage in AC patients. Univariate Kaplan–Meier analysis showed that loss of KRT19 or elevated hASH1 expression significantly correlated with decreased survival in SC/ASC patients. In contrast, positive KRT19 and hASH1 expression correlated with a shorter survival time in AC patients. Multivariate Cox regression analysis showed that negative KRT19 expression or positive hASH1 expression was an independent poor-prognostic predictor in SC/ASC, but positive KRT19 and hASH1 expression were poor-prognostic factors in AC patients. Our study suggested that hASH1 can be used to determine the malignancy of SC/ASC and AC tumors and is associated with poor prognosis. In contrast, KRT19 is a protective factor in AC patients but a sign of malignancy in SC/ ASC patients. C1 [Li, Jiequn] Central South University, The Second Xiangya Hospital, Department of General Surgery, 410011 Changsha, Hunan, China. [Yang, Zhu-Lin] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, Hunan, China. [Zou, Qiong] Central South University, Third Xiangya Hospital, Department of Pathology, 410013 Changsha, Hunan, China. [Yuan, Yuan] Central South University, Third Xiangya Hospital, Department of Pathology, 410013 Changsha, Hunan, China. [Li, Jinghe] Central South University, Xiangya Basic Medical College, Department of Pathology, 410078 Changsha, Hunan, China. [Liang, Lufeng] Hunan Provincial People’s Hospital, Department of Hepatobiliary and Pancreatic Surgery, 410007 Changsha, Hunan, China. [Zhen, Guixiang] Loudi Central Hospital, Department of Pathology, 417011 Loudi, Hunan, China. [Chen, Senlin] Hunan Provincial Tumor Hospital, Department of Pathology, 410013 Changsha, Hunan, China. RP Yang, ZL (reprint author), Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, China. EM zhulinyang@yahoo.com CR Jemal A, Siegel R, Ward E et al, 2008, Cancer statistics, 2008. CA Cancer J Clin 58:71–96 Jayaraman S, Jarnagin WR, 2010, Management of gallbladder cancer. Gastroenterol Clin N Am 39:331–342 Bamburg JR, Wiggan OP, 2002, ADF/cofilin and actin dynamics in disease. Trends Cell Biol 12:598–605 Roa JC, Tapia O, Cakir A et al, 2011, Squamous cell and adenosquamous carcinomas of the gallbladder: clinicopathological analysis of 34 cases identified in 606 carcinomas. Mod Pathol 24: 1069–1078 Wang W, Eddy R, Condeelis J, 2007, The cofilin pathway in breast cancer invasion and metastasis. Nat Rev Cancer 7:429–440 Nishihara K, Nagai E, Izumi Y, Yamaguchi K, Tsuneyoshi M, 1994, Adenosquamous carcinoma of the gallbladder: a clinicopathological, immunohistochemical and flow-cytometric study of twenty cases. Jpn J Cancer Res 85:389–399 Kondo M, Dono K, Sakon M et al, 2002, Adenosquamous carcinoma of the gallbladder. Hepato-Gastroenterology 49:1230–1234 Park SB, Kim YH, Rho HL, Chae GB, Hong SK, 2012, Primary carcinosarcoma of the gallbladder. J Korean Surg Soc 82:54–58 Barak V, Goike H, Panaretakis KW, Einarsson R, 2004, Clinical utility of cytokeratins as tumor markers. Clin Biochem 37:529–540 Lacroix M, 2006, Significance, detection and markers of disseminated breast cancer cells. Endocr Relat Cancer 13: 1033–1067 Prasad ML, Pellegata NS, Huang Y, Nagaraja HN, de la Chapelle A, Kloos RT, 2005, Galectin-3, fibronectin-1, CITED-1, HBME1 and cytokeratin-19 immunohistochemistry is useful for the differential diagnosis of thyroid tumors. Mod Pathol 18:48–57 Chu PG, Weiss LM, 2002, Keratin expression in human tissues and neoplasms. Histopathology 40:403–439 Nozato M, Kaneko S, Nakagawara A, Komuro H, 2013, Epithelialmesenchymal transition-related gene expression as a new prognostic marker for neuroblastoma. Int J Oncol 42:134–140 Jain R, Fischer S, Serra S, Chetty R, 2010, The use of Cytokeratin 19, CK19, immunohistochemistry in lesions of the pancreas, gastrointestinal tract, and liver. Appl Immunohistochem Mol Morphol 18:9–15 Axelson H, 2004, The Notch signaling cascade in neuroblastoma: role of the basic helix-loop-helix proteins HASH-1 and HES-1. Cancer Lett 204:171–178 Ball DW(2004, Achaete-scute homolog-1 and Notch in lung neuroendocrine development and cancer. Cancer Lett 204:159–169 Sippel RS, Carpenter JE, Kunnimalaiyaan M, Chen H, 2003, The role of human achaete-scute homolog-1 in medullary thyroid cancer cells. Surgery 134:866–871 Kunnimalaiyaan M, Traeger K, Chen H, 2005, Conservation of the Notch1 signaling pathway in gastrointestinal carcinoid cells. Am J Physiol Gastrointest Liver Physiol 289:G636–G642 Shida T, Furuya M, Nikaido T et al, 2005, Aberrant expression of human achaete-scute homologue gene 1 in the gastrointestinal neuroendocrine carcinomas. Clin Cancer Res 11(2 Pt 1):450–458 MikiM, Ball DW, Linnoila RI, 2012, Insights into the achaete-scute homolog-1 gene, hASH1, in normal and neoplastic human lung. Lung Cancer 75:58–65 Jensen-Taubman S, Wang XY, Linnoila RI, 2010, Achaete-scute homologue-1 tapers neuroendocrine cell differentiation in lungs after exposure to naphthalene. Toxicol Sci 117:238–248 Liu DC, Yang ZL, 2011, Overexpression of EZH2 and loss of expression of PTEN is associated with invasion, metastasis, and poor progression of gallbladder adenocarcinoma. Pathol Res Pract 207: 472–478 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 285 EP 292 DI 10.1007/s12253-013-9693-2 PG 8 ER PT J AU Shi, B Wang, X Han, X Liu, P Wei, W Li, Y AF Shi, Baomin Wang, Xiuyan Han, Xujie Liu, Pengfei Wei, Weiwei Li, Yan TI Functional Modules Analysis Based on Coexpression Network in Pancreatic Ductal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Co-expression network; Functional modules; Pancreatic ductal adenocarcinoma ID Co-expression network; Functional modules; Pancreatic ductal adenocarcinoma AB Pancreatic ductal adenocarcinoma (PDAC) is the most common epithelial, exocrine pancreatic malignancy, accounting formore than 80%of themalignant neoplasms of the pancreas. Although the molecular basis of pancreatic cancer is now better understood than ever before, there remains a long distance from being completely understood. In this study, we identified the differentially expressed genes (DEGs) in PDAC tissue compared with normal tissue and constructed a coexpression network by computing the pairwise correlation coefficient between the DEGs. We applied a statistical approach of MCODE to cluster genes in the coexpression network. Ten functional modules were identified in this network. Our results strongly suggest that dysregulations of immune response, homeostasis and cell adhesion may significantly contribute to the development and progression of PDAC. Results from this study will provide the groundwork for the understanding of PDAC. Future studies are needed to confirm some of the possible interactions suggested by this study. C1 [Shi, Baomin] Tongji Hospital of Tongji University, Department of General Surgery, 389 xincun road, Putuo district, 200065 Shanghai, China. [Wang, Xiuyan] Tongji Hospital of Tongji University, Department of UltrasonographyShanghai, China. [Han, Xujie] Tongji Hospital of Tongji University, Department of General Surgery, 389 xincun road, Putuo district, 200065 Shanghai, China. [Liu, Pengfei] Tongji Hospital of Tongji University, Department of General Surgery, 389 xincun road, Putuo district, 200065 Shanghai, China. [Wei, Weiwei] Tongji Hospital of Tongji University, Department of General Surgery, 389 xincun road, Putuo district, 200065 Shanghai, China. [Li, Yan] Tongji Hospital of Tongji University, Department of General Surgery, 389 xincun road, Putuo district, 200065 Shanghai, China. RP Shi, B (reprint author), Tongji Hospital of Tongji University, Department of General Surgery, 200065 Shanghai, China. EM shibaominsbm@hotmail.com CR Alexakis N, Halloran C, RaratyM, Ghaneh P, Sutton R, Neoptolemos JP, 2004, Current standards of surgery for pancreatic cancer. Br J Surg 91(11):1410–1427., DOI 10.1002/bjs.4794 Siegel R,Ward E, Brawley O, Jemal A, 2011, Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 61(4):212–236 Ghaneh P, Costello E, Neoptolemos JP, 2007, Biology and management of pancreatic cancer. Gut 56(8):1134–1152 Li D, Xie K, Wolff R, Abbruzzese JL, 2004, Pancreatic cancer. Lancet 363(9414):1049–1057., DOI 10.1016/S0140-6736(04)15841-8 Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A, Hong SM, Fu B, Lin MT, Calhoun ES, Kamiyama M, Walter K, Nikolskaya T, Nikolsky Y, Hartigan J, Smith DR, Hidalgo M, Leach SD, Klein AP, Jaffee EM, Goggins M, Maitra A, Iacobuzio-Donahue C, Eshleman JR, Kern SE, Hruban RH, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW, 2008, Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321(5897):1801–1806 Carter H, Samayoa J, Hruban RH, Karchin R, 2010, Prioritization of driver mutations in pancreatic cancer using cancer-specific highthroughput annotation of somatic mutations, CHASM). Cancer Biol Ther 10(6):582–587 Zhang G, Schetter A, He P, Funamizu N, Gaedcke J, Ghadimi BM, Ried T, Hassan R, Yfantis HG, Lee DH, Lacy C, Maitra A, Hanna N, Alexander HR, Hussain SP, 2012, DPEP1 inhibits tumor cell invasiveness, enhances chemosensitivity and predicts clinical outcome in pancreatic ductal adenocarcinoma. PLoS One 7(2):e31507., DOI 10. 1371/journal.pone.0031507 Perez-Mancera PA, Rust AG, van der Weyden L, Kristiansen G, Li A, Sarver AL, Silverstein KA, Grutzmann R, Aust D, Rummele P, Knosel T, Herd C, Stemple DL, Kettleborough R, Brosnan JA, Morgan R, Knight S, Yu J, Stegeman S, Collier LS, ten Hoeve JJ, de Ridder J, Klein AP, GogginsM, Hruban RH, Chang DK, Biankin AV, Grimmond SM,Wessels LF,Wood SA, Iacobuzio-Donahue CA, Pilarsky C, Largaespada DA, Adams DJ, Tuveson DA, 2012, The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma. Nature 486(7402):266–270., DOI 10.1038/nature11114 VincentA,Omura N, Hong SM, Jaffe A, Eshleman J, GogginsM(2011, Genome-wide analysis of promoter methylation associated with gene expression profile in pancreatic adenocarcinoma. Clin Cancer Res 17(13):4341–4354., DOI 10.1158/1078-0432.CCR-10-3431 Badea L, HerleaV, Dima SO,Dumitrascu T, Popescu I, 2008, Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia. Hepatogastroenterology 55(88):2016–2027 Badea L, Herlea V, Dima SO, Dumitrascu T, Popescu I, 2008, Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia. Hepatogastroenterology 55(88): 2015–2026 Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP, 2003, Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4(2):249–264., DOI 10.1093/biostatistics/4.2.249 Team RDC, 2008, R: A language and environment for statistical computing. R Foundation for Statistical Computing da Huang W, Sherman BT, Lempicki RA, 2009, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4(1):44–57 Tusher VG, Tibshirani R, Chu G, 2001, Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A 98(9):5116–5121., DOI 10.1073/pnas.091062498 Bader GD, Hogue CW, 2003, An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics 4:2 Allocco DJ, Kohane IS, Butte AJ, 2004, Quantifying the relationship between co-expression, co-regulation and gene function. BMC Bioinformatics 5:18., DOI 10.1186/1471-2105-5-18 Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T, 2003, Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13(11):2498–2504., DOI 10.1101/gr.1239303 Hiraoka N, Onozato K, Kosuge T, Hirohashi S, 2006, Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clin Cancer Res 12(18):5423–5434 Gross S, Walden P, 2008, Immunosuppressive mechanisms in human tumors: why we still cannot cure cancer. Immunol Lett 116(1):7–14 Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA, 2006, Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 20(10):1218–1249 Gloushankova NA, 2008, Changes in regulation of cell-cell adhesion during tumor transformation. Biochemistry, Mosc, 73(7):742–750 Tlsty TD, 1998, Cell-adhesion-dependent influences on genomic instability and carcinogenesis. Curr Opin Cell Biol 10(5):647–653 Deer EL, Gonzalez-Hernandez J, Coursen JD, Shea JE, Ngatia J, Scaife CL, Firpo MA, Mulvihill SJ, 2010, Phenotype and genotype of pancreatic cancer cell lines. Pancreas 39(4):425–435., DOI 10.1097/ MPA.0b013e3181c15963 Zhou W, Capello M, Fredolini C, Racanicchi L, Piemonti L, Liotta LA, Novelli F, Petricoin EF, 2011, Phosphoproteomic analysis of pancreatic ductal adenocarcinoma cells reveals differential phosphorylation of cell adhesion, cell junction and structural proteins. J Proteomics Bioinforma 4:170–178 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 293 EP 299 DI 10.1007/s12253-013-9694-1 PG 7 ER PT J AU Manoochehri, M Karbasi, A Bandehpour, M Kazemi, B AF Manoochehri, Mehdi Karbasi, Ashraf Bandehpour, Mojgan Kazemi, Bahram TI Down-Regulation of BAX Gene During Carcinogenesis and Acquisition of Resistance to 5-FU in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Carcinogenesis; 5-FU; Chemoresistance; Apoptosis ID Colorectal cancer; Carcinogenesis; 5-FU; Chemoresistance; Apoptosis AB Carcinogenesis and resistance to chemotherapy could be as results of expression variations in apoptosis regulating genes. Changes in the expression of apoptosis interfering genes may contribute to colorectal carcinogenesis and resistance to 5-Flourouracil (5-FU) during treatment schedule period. The present study aimed to evaluate the expression of pro-apoptotic and anti-apoptotic genes in colorectal cancer tumor tissues, normal adjacent tissues, and tumor colorectal cancer cell line during acquiring resistance to 5-FU in HT-29 based on Bolus treatment protocol. The normal and tumor tissues were obtained from hospital after surgery and total RNA was extracted for expression analysis. The HT-29 colorectal cancer cell line was cultured and exposed with 5-FU in three stages based on Bolus protocol. The MTT assay and Real Time PCR were carried out to determine the sensitivity to the drug and expression of desired genes, respectively. The obtained data showed that Proapoptotic genes, BAX and BID, were down-regulated in resistant derivate cells compared to wild type HT-29 cells. On the other hand Antiapoptotic genes, CIAP1 and XIAP, showed upregulation in resistant cells compared to wild type ones. Furthermore, BAX and FAS genes showed down-regulation in tumor samples in comparison to normal adjacent tissues. In conclusion, the results of our study suggest that BAX down-regulation could contribute as an important factor during both colorectal carcinogenesis and cell resistance to 5-FU. C1 [Manoochehri, Mehdi] Shahid Beheshti University of Medical Sciences, Cellular and Molecular Biology Research CenterTehran, Iran. [Karbasi, Ashraf] Baqyiatallah Research Center for Gastroenterology and Liver Diseases, Gastroenterology and Hepatology DepartmentTehran, Iran. [Bandehpour, Mojgan] Shahid Beheshti University of Medical Sciences, Cellular and Molecular Biology Research CenterTehran, Iran. [Kazemi, Bahram] Shahid Beheshti University of Medical Sciences, Cellular and Molecular Biology Research CenterTehran, Iran. RP Kazemi, B (reprint author), Shahid Beheshti University of Medical Sciences, Cellular and Molecular Biology Research Center, Tehran, Iran. EM kazemi@sbmu.ac.ir CR Herszenyi L, Tulassay Z, 2010, Epidemiology of gastrointestinal and liver tumors. Eur Rev Med Pharmacol Sci 14(4):249–258 Naishadham D, Lansdorp-Vogelaar I, Siegel R, Cokkinides V, Jemal A(2011, State disparities in colorectal cancermortality patterns in the United States. Cancer Epidemiol Biomarkers Prev 20(7):1296–1302 PourhoseingholiMA, 2012, Increased burden of colorectal cancer in Asia. World J Gastrointest Oncol 4(4):68–70 Park S, Bae J, Nam BH, Yoo KY, 2008, Aetiology of cancer in Asia. Asian Pac J Cancer Prev 9(3):371–380 Koehler A, Bataille F, Schmid C, Ruemmele P, Waldeck A, Blaszyk H, Hartmann A, Hofstaedter F, DietmaierW(2004, Gene expression profiling of colorectal cancer and metastases divides tumours according to their clinicopathological stage. J Pathol 204(1):65–74 Fukushima T, Takenoshita S, 2001, Colorectal carcinogenesis. Fukushima J Med Sci 47(1):1–11 Watson AJ, 2004, Apoptosis and colorectal cancer. Gut 53(11): 1701–1709 ShaibW, Lee V, Saif MW, 2009, Bolus 5-fluorouracil as an alternative modality to infusion 5-fluorouracil in a patient with rectal cancer and capecitabine-induced cardiotoxicity. In Vivo 23(5):821–826 Berger FG, Berger SH, 2006, Thymidylate synthase as a chemotherapeutic drug target: where are we after fifty years? Cancer Biol Ther 5(9):1238–1241 Buroker TR, O’Connell MJ, Wieand HS, Krook JE, Gerstner JB, Mailliard JA, Schaefer PL, Levitt R, Kardinal CG, Gesme DH Jr, 1994, Randomized comparison of two schedules of fluorouracil and leucovorin in the treatment of advanced colorectal cancer. J Clin Oncol 12(1):14–20 Piedbois P, Rougier P, Buyse M, Pignon JP, Ryan L, Hansen R, Zee B, Weinerman B, Pater J, Leichman C, Macdonald J, Benedetti J, Lokich J, Fryer J, Brufman G, Isacson R, Laplanche A, Levy E, 1998, Efficacy of intravenous continuous infusion of fluorouracil compared with bolus administration in advanced colorectal cancer. Meta-analysis Group In Cancer. J Clin Oncol 16(1):301–308 SchmidtWM, KalipciyanM, Dornstauder E, Rizovski B, Steger GG, Sedivy R, Mueller MW, Mader RM, 2004, Dissecting progressive stages of 5-fluorouracil resistance in vitro using RNA expression profiling. Int J Cancer 112(2):200–212 Zhang N, Yin Y, Xu SJ, Chen WS, 2008, 5-Fluorouracil: mechanisms of resistance and reversal strategies. Molecules 13(8):1551–1569 Peters GJ, Backus HH, Freemantle S, van Triest B, Codacci-Pisanelli G, van derWilt CL, Smid K, Lunec J, Calvert AH, Marsh S,McLeod HL, Bloemena E, Meijer S, Jansen G, van Groeningen CJ, Pinedo HM, 2002, Induction of thymidylate synthase as a 5-fluorouracil resistance mechanism. Biochim Biophys Acta 1587(2–3):194–205 Grem JL, 2005, Screening for dihydropyrimidine dehydrogenase deficiency. Clin Cancer Res 11(14):5067–5068 Arnold CN, Goel A, Boland CR, 2003, Role of hMLH1 promoter hypermethylation in drug resistance to 5-fluorouracil in colorectal cancer cell lines. Int J Cancer 106(1):66–73 Akao Y, Noguchi S, Iio A, Kojima K, Takagi T, Naoe T, 2011, Dysregulation of microRNA-34a expression causes drug-resistance to 5-FU in human colon cancer DLD-1 cells. Cancer Lett 300(2):197–204 Choi S, Ku JL, 2011, Resistance of colorectal cancer cells to radiation and 5-FU is associated with MELK expression. Biochem Biophys Res Commun 412(2):207–213 Humeniuk R,Menon LG,Mishra PJ, Gorlick R, Sowers R, RodeW, Pizzorno G, Cheng YC, Kemeny N, Bertino JR, Banerjee D, 2009, Decreased levels of UMP kinase as a mechanism of fluoropyrimidine resistance. Mol Cancer Ther 8(5):1037–1044 Violette S, Poulain L, Dussaulx E, Pepin D, Faussat AM, Chambaz J, Lacorte JM, Staedel C, Lesuffleur T, 2002, Resistance of colon cancer cells to long-term 5-fluorouracil exposure is correlated to the relative level of Bcl-2 and Bcl-X(L, in addition to Bax and p53 status. Int J Cancer 98(4):498–504 Shi X, Liu S, Kleeff J, Friess H, Buchler MW, 2002, Acquired resistance of pancreatic cancer cells towards 5-fluorouracil and gemcitabine is associated with altered expression of apoptosisregulating genes. Oncology 62(4):354–362 Meng J, Zhang HH, Zhou CX, Li C, Zhang F, Mei QB, 2012, The histone deacetylase inhibitor trichostatin A induces cell cycle arrest and apoptosis in colorectal cancer cells via p53-dependent and - independent pathways. Oncol Rep 28(1):384–388 Pizzorno G, Handschumacher RE, 1995, Effect of clinically modeled regimens on the growth response and development of resistance in human colon carcinoma cell lines. Biochem Pharmacol 49(4):559–565 Igney FH, Krammer PH, 2002, Death and anti-death: tumour resistance to apoptosis. Nat Rev Cancer 2(4):277–288 Luo X, Budihardjo I, Zou H, Slaughter C, Wang X, 1998, Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors.Cell 94(4):481–490 Elmore S, 2007, Apoptosis: a review of programmed cell death. Toxicol Pathol 35(4):495–516 Kaufmann T, Strasser A, Jost PJ, 2012, Fas death receptor signalling: roles of Bid and XIAP. Cell Death Differ 19(1):42–50 Stevenson L, AllenWL, Proutski I, Stewart G, Johnston L,McCloskey K, Wilson PM, Longley DB, Johnston PG, 2011, Calbindin 2, CALB2, regulates 5-fluorouracil sensitivity in colorectal cancer by modulating the intrinsic apoptotic pathway. PLoS One 6(5):e20276 Samuel S, Fan F, Dang LH, Xia L, Gaur P, Ellis LM(2011, Intracrine vascular endothelial growth factor signaling in survival and chemoresistance of human colorectal cancer cells. Oncogene 30(10):1205–1212 Sax JK, Fei P,MurphyME, Bernhard E, Korsmeyer SJ, El-DeiryWS, 2002, BID regulation by p53 contributes to chemosensitivity. Nat Cell Biol 4(11):842–849 Miao J, Chen GG, Chun SY, Chak EC, Lai PB, 2004, Bid sensitizes apoptosis induced by chemotherapeutic drugs in hepatocellular carcinoma. Int J Oncol 25(3):651–659 Li Y, Jian Z, Xia K, Li X, Lv X, Pei H, Chen Z, Li J, 2006, XIAP is related to the chemoresistance and inhibited its expression by RNA interference sensitize pancreatic carcinoma cells to chemotherapeutics. Pancreas 32(3):288–296 Nomura T, YamasakiM, Nomura Y,Mimata H, 2005, Expression of the inhibitors of apoptosis proteins in cisplatin-resistant prostate cancer cells. Oncol Rep 14(4):993–997 Karasawa H, Miura K, Fujibuchi W, Ishida K, Kaneko N, Kinouchi M, OkabeM, Ando T,Murata Y, Sasaki H, Takami K, Yamamura A, Shibata C, Sasaki I, 2009, Down-regulation of cIAP2 enhances 5-FU sensitivity through the apoptotic pathway in human colon cancer cells. Cancer Sci 100(5):903–913 Katkoori VR, Suarez-Cuervo C, Shanmugam C, Jhala NC, Callens T, Messiaen L, Posey J 3rd, Bumpers HL,Meleth S, GrizzleWE, Manne U, 2010, Bax expression is a candidate prognostic and predictive marker of colorectal cancer. J Gastrointest Oncol 1(2):76–89 Guillen-Ahlers H, Suckow MA, Castellino FJ, Ploplis VA, 2010, Fas/CD95 deficiency in ApcMin/+ mice increases intestinal tumor burden. PLoS One 5(2):e9070 Butler LM, Dobrovic A, Bianco T, Cowled PA, 2000, Promoter region methylation does not account for the frequent loss of expression of the Fas gene in colorectal carcinoma. Br J Cancer 82(1):131–135 Petak I, Danam RP, Tillman DM, Vernes R, Howell SR, Berczi L, Kopper L, Brent TP, Houghton JA, 2003, Hypermethylation of the gene promoter and enhancer region can regulate Fas expression and sensitivity in colon carcinoma. Cell Death Differ 10(2):211–217 Zhang L, Yu J, Park BH, Kinzler KW, Vogelstein B, 2000, Role of BAX in the apoptotic response to anticancer agents. Science 290(5493):989–992 Jansson A, Sun XF, 2002, Bax expression decreases significantly from primary tumor to metastasis in colorectal cancer. J Clin Oncol 20(3):811–816 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 301 EP 307 DI 10.1007/s12253-013-9695-0 PG 7 ER PT J AU Dong-Feng, Z Ting, L Yong, Z Cheng, Ch Xi, Z Pei-Yan, K AF Dong-Feng, Zeng Ting, Liu Yong, Zhang Cheng, Chang Xi, Zhang Pei-Yan, Kong TI The TPO/c-MPL Pathway in the Bone Marrow may Protect Leukemia Cells from Chemotherapy in AML Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thrombopoietin; c-MPL; Acutemyeloid leukemia; Chemotherapy resistance; Relapse ID Thrombopoietin; c-MPL; Acutemyeloid leukemia; Chemotherapy resistance; Relapse AB Accumulating evidence indicates that the interaction of human LSCs (leukemic stem cells) with the hematopoietic microenvironment, mediated by the thrombopoietin (TPO)/c-MPL pathway, may be an underlying mechanism for resistance to cell cycle–dependent cytotoxic chemotherapy. However, the role of TPO/c-MPL signaling in AML (acute myelogenous leukemia) chemotherapy resistance hasn’t been fully understood. The c-MPL and TPO levels in different AML samples were measured by flow cytometry and ELISA. We also assessed the TPO levels in the osteoblasts derived from bone mesenchymal stem cells (BMSCs). The survival rate of an AML cell line that had been co-cultured with different BMSC-derived osteoblasts was measured to determine the IC50 of an AML chemotherapy drug daunorubicin (DNR). The levels of TPO/c-MPL in the initial and relapse AML patients were significantly higher than that in the control (P <0.05). The osteoblasts derived from AML patients’ BMSCs secreted more TPO than the osteoblasts derived from normal control BMSCs (P <0.05). A strong positive correlation between the TPO level and c-MPL expression was found in the bone marrow mononuclear cells of the relapse AML patients. More importantly, the IC50 of DNR in the HEL + AML-derived osteoblastswas the highest among all co-culture systems. High level of TPO/c-MPL signaling may protect LSCs from chemotherapy in AML. The effects of inhibition of the TPO/c-MPL pathway on enhancing the chemotherapy sensitivity of AML cells, and on their downstream effector molecules that direct the interactions between patientderived blasts and leukemia repopulating cells need to be further studied. C1 [Dong-Feng, Zeng] Third Military Medical University, Xinqiao Hospital, Department of Hematology, 400037 Chongqing, China. [Ting, Liu] Third Military Medical University, Daping Hospital, Department of Ophtalmology, 400042 Chongqing, China. [Yong, Zhang] Chengdu General Hospital of Chengdu Military Region, Department of Hematology, 610000 Chengdu, China. [Cheng, Chang] Third Military Medical University, Xinqiao Hospital, Department of Hematology, 400037 Chongqing, China. [Xi, Zhang] Third Military Medical University, Xinqiao Hospital, Department of Hematology, 400037 Chongqing, China. [Pei-Yan, Kong] Third Military Medical University, Xinqiao Hospital, Department of Hematology, 400037 Chongqing, China. RP Pei-Yan, K (reprint author), Third Military Medical University, Xinqiao Hospital, Department of Hematology, 400037 Chongqing, China. EM kongpeiyanpp1312@hotmail.com CR She M, Niu X, Chen X, Li J, Zhou M, He Y, Le Y, Guo K, 2012, Resistance of leukemic stem-like cells in AML cell line KG1a to natural killer cell-mediated cytotoxicity. Cancer Lett 318(2):173– 179., DOI 10.1016/j.canlet.2011.12.017 Nervi B, Ramirez P, Rettig MP, Uy GL, Holt MS, Ritchey JK, Prior JL, Piwnica-Worms D, Bridger G, Ley TJ, DiPersio JF, 2009, Chemosensitization of acute myeloid leukemia, AML, following mobilization by the CXCR4 antagonist AMD3100. Blood 113(24): 6206–6214., DOI 10.1182/blood-2008-06-162123 Saito Y, Kitamura H, Hijikata A, Tomizawa-Murasawa M, Tanaka S, Takagi S, Uchida N, Suzuki N, Sone A, Najima Y, Ozawa H, Wake A, Taniguchi S, Shultz LD, Ohara O, Ishikawa F, 2010, Identification of therapeutic targets for quiescent, chemotherapy-resistant human leukemia stem cells. Sci TranslMed 2(17):17ra19., DOI 10.1126/ scitranslmed.3000349 Lane SW, Scadden DT, Gilliland DG, 2009, The leukemic stem cell niche: current concepts and therapeutic opportunities. Blood 114(6): 1150–1157., DOI 10.1182/blood-2009-01-202606 Blanco TM, Mantalaris A, Bismarck A, Panoskaltsis N, 2010, The development of a three-dimensional scaffold for ex vivo biomimicry of human acute myeloid leukaemia. Biomaterials 31(8):2243–2251., DOI 10.1016/j.biomaterials.2009.11.094 Ishikawa FYS, Saito Y, HijikataA, KitamuraH, Tanaka S,Nakamura R, Tanaka T, Tomiyama H, Saito N, Fukata M, Miyamoto T, Lyons B, Ohshima K, Uchida N, Taniguchi S, Ohara O, Akashi K, Harada M, Shultz LD, 2007, Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol 25:1315–1321 Zeng Z, Shi YX, Samudio IJ,Wang RY, Ling X, Frolova O, LevisM, Rubin JB, Negrin RR, Estey EH, Konoplev S, Andreeff M, Konopleva M, 2009, Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML. Blood 113(24):6215–6224., DOI 10.1182/ blood-2008-05-158311 Jawad MSC, Mony U, Grundy M, Russell NH, Pallis M, 2010, Analysis of factors that affect in vitro chemosensitivity of leukaemic stem and progenitor cells to gemtuzumab ozogamicin, Mylotarg, in acute myeloid leukaemia. Leukemia 24:74–80 Parmar A, Marz S, Rushton S, Holzwarth C, Lind K, Kayser S, Dohner K, Peschel C, Oostendorp RA, Gotze KS, 2011, Stromal niche cells protect early leukemic FLT3-ITD+ progenitor cells against first-generation FLT3 tyrosine kinase inhibitors. Cancer Res 71(13):4696–4706., DOI 10.1158/0008-5472.CAN-10-4136 Lane SW, Wang YJ, Lo Celso C, Ragu C, Bullinger L, Sykes SM, Ferraro F, Shterental S, Lin CP, Gilliland DG, Scadden DT, Armstrong SA, Williams DA, 2011, Differential niche and Wnt requirements during acute myeloid leukemia progression. Blood 118(10): 2849–2856., DOI 10.1182/blood-2011-03-345165 Guo YBS, Luo M, 1999, Study of thrombopoietin and its recptor in the acute myeloid leukemia. Zhonghua Xue Ye Xue Za Zhi 20:120– 123 Schroder JK, Kolkenbrock S, Tins J, Kasimir-Bauer S, Seeber S, Schutte J, 2000, Analysis of thrombopoietin receptor, c-mpl, mRNA expression in de novo acute myeloid leukemia. Leuk Res 24(5):401– 409 Li L, Xie T, 2005, Stem cell niche: structure and function. Annu Rev Cell Dev Biol 21:605–631., DOI 10.1146/annurev.cellbio.21.012704. 131525 Adams GB, Scadden DT, 2006, The hematopoietic stem cell in its place. Nat Immunol 7(4):333–337 LE Wilson A, Trumpp A, 2009, Balancing dormant and selfrenewing hematopoietic stem cells. Curr Opin Genet Dev 19:461– 468 Yoshihara HAF, Hosokawa K, Hagiwara T, Takubo K, Nakamura Y, Gomei Y, Iwasaki H, Matsuoka S, Miyamoto K, Miyazaki H, Takahashi T, Suda T, 2007, Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche. Cell Stem Cell 1:685–697 Matsumura IKY, Kato T, Ikeda H, Ishikawa J, Horikawa Y, Hashimoto K, Moriyama Y, Tsujimura T, Nishiura T, 2005, Growth response of acute myeloblastic leukemia cells to recombinant human thrombopoietin. Blood 86:703–709 Corazza FHC, D’Hondt S, Ferster A, Kentos A, Benoit Y, Sariban E, 2006, Circulating thrombopoietin as an in vivo growth factor for blast cells in acute myeloid leukemia. Blood 107:2525–2530 Kaushansky K, 2005, The molecular mechanisms that control thrombopoiesis. J Clin Invest 115:3339–3347 Sitnicka E, Lin N, Priestley GV, Fox N, Broudy VC, Wolf NS, Kaushansky K, 1996, The effect of thrombopoietin on the proliferation and differentiation of murine hematopoietic stem cells. Blood 87(12):4998–5005 Ku H, Yonemura Y, Kaushansky K, Ogawa M, 1996, Thrombopoietin, the ligand for theMpl receptor, synergizes with steel factor and other early acting cytokines in supporting proliferation of primitive hematopoietic progenitors of mice. Blood 87(11):4544– 4551 Fox N, Priestley G, Papayannopoulou T, Kaushansky K, 2002, Thrombopoietin expands hematopoietic stem cells after transplantation. J Clin Invest 110(3):389–394., DOI 10.1172/JCI15430 Wu YX, Wang HY, Wang W, 2010, Measurement and clinical significance of serum TPO and LDH levels in patients with myelodysplastic syndrome and acute leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 18(3):671–674 Wetzler MBM, Bernstein SH, Blumenson L, Stewart C, Barcos M, Mrozek K, Block AW, Herzig GP, Bloomfield CD, 1997, Expression of c-mpl mRNA, the receptor for thrombopoietin, in acute myeloid leukemia blasts identifies a group of patients with poor response to intensie chemotherapy. Clin Oncol 15:2262–2268 Arai F, Yoshihara H, Hosokawa K, Nakamura Y, Gomei Y, Iwasaki H, Suda T, 2009, Niche regulation of hematopoietic stem cells in the endosteum. Ann N Y Acad Sci 1176:36–46., DOI 10.1111/j.1749- 6632.2009.04561.x NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 309 EP 317 DI 10.1007/s12253-013-9696-z PG 9 ER PT J AU Mozes, P Szanto, E Tiszlavicz, L Barzo, P Cserhati, A Fodor, E Hideghety, K AF Mozes, Petra Szanto, Erika Tiszlavicz, Laszlo Barzo, Pal Cserhati, Adrienne Fodor, Emese Hideghety, Katalin TI Clinical Course of Central Neurocytoma with Malignant Transformation—An Indication for Craniospinal Irradiation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Central neurocytoma; Craniospinal dissemination; Radiotherapy; MIB-1 labeling index ID Central neurocytoma; Craniospinal dissemination; Radiotherapy; MIB-1 labeling index AB Central neurocytoma is generally considered to be a benign tumor and the literature suggests that a cure may be attained by surgery ± adjuvant focal irradiation. However, there is a need for change in the therapeutic strategy for the subgroup of patients with aggressive central neurocytoma. An example case is presented and the literature on central neurocytoma cases with malignant features and dissemination via the cerebrospinal fluid is reviewed and the radiotherapeutic strategies available for central neurocytoma treatment is discussed. Nineteen cases including the present report with a malignant course and cerebrospinal fluid dissemination have been described to date, most of them involving an elevated MIB-1 labeling index. Our case exhibited atypical central neurocytoma with an initially elevated MIB-1 labeling index (25–30 %). The primary treatment included surgery and focal radiotherapy. Three years later the disease had disseminated throughout the craniospinal axis. A good tumor response and symptom relief were achieved with repeated radiation and temozolomide chemotherapy. Central neurocytoma with an initially high proliferation activity has a high tendency to spread via the cerebrospinal fluid. The chemo- and radiosensitivity of the tumor suggest a more aggressive adjuvant therapy approach. Cases with a potential for malignant transformation should be identified and treated appropriately, including irradiation of the entire neuroaxis and adjuvant chemotherapy may be considered. C1 [Mozes, Petra] University of Szeged, Department of Oncotherapy, Koranyi fasor 12., 6720 Szeged, Hungary. [Szanto, Erika] University of Szeged, Department of Oncotherapy, Koranyi fasor 12., 6720 Szeged, Hungary. [Tiszlavicz, Laszlo] University of Szeged, Department of Pathology, Allomas u. 2., 6720 Szeged, Hungary. [Barzo, Pal] University of Szeged, Department of Neurosurgery, Semmelweis u. 6., 6720 Szeged, Hungary. [Cserhati, Adrienne] University of Szeged, Department of Oncotherapy, Koranyi fasor 12., 6720 Szeged, Hungary. [Fodor, Emese] University of Szeged, Department of Oncotherapy, Koranyi fasor 12., 6720 Szeged, Hungary. [Hideghety, Katalin] University of Szeged, Department of Oncotherapy, Koranyi fasor 12., 6720 Szeged, Hungary. RP Mozes, P (reprint author), University of Szeged, Department of Oncotherapy, 6720 Szeged, Hungary. EM mozespetra@gmail.com CR Hassoun J, Gambareli D, Grisoli F, PelletW, Salamon G, Pellisier JF, Toga M, 1982, Central neurocytoma—an electron-microscopic study of 2 cases. Acta Neuropathol 56:151–156 Leenstra JL, Rodriguez FJ, Frechette CM, Giannini C, Stafford SL, Pollock BE, Schild SE, Scheithauer BW, Jenkins RB, Buckner JC, Brown PD, 2007, Central neurocytoma: management recommendations based on a 35-year experience. Int J Radiat Oncol Biol Phys 67: 1145–1154 Rades D, Fehlauer F, Lamszus K, Schild SE, Hagel C, Westphal M, Alberti W, 2005, Well-differentiated neurocytoma: what is the best available treatment? Neuro-Oncol 7:77–83 Schmidt MH, Gottfried ON, von Koch CS, Chang SM, McDermott MW, 2004, Central neurocytoma: a review. J Neurooncol 66:377– 384 Vasiljevic A, Francois P, Loundou A, Fevre-Montange M, Jouvet A, Roche PH, Figarella-Branger D, 2012, Prognostic factors in central neurocytomas: a multicenter study of 71 cases. Am J Surg Pathol 36: 220–227 Kane AJ, SughrueME, RutkowskiMJ, Tihan T, ParsaAT, 2011, The molecular pathology of central neurocytomas. J Clin Neurosci 18:1– 6 Kubota T, Hayashi M, Kawano H, Kabuto M, Sato K, Ishise J, Kawamoto K, Shirataki K, Iizuka H, Tsunoda S, Katsuyama J, 1991, Central neurocytoma - immunohistochemical and ultrastructural-study. Acta Neuropathol 81:418–427 Von Deimling A, Janzer R, Kleihues P, Wiestler OD, 1990, Patterns of differentiation in central neurocytoma—an immunohistochemical study of 11 biopsies. Acta Neuropathol 79:473–479 Coca S, Moreno M, Martos JA, Rodriguez J, Barcena A, Vaquero J, 1994, Neurocytoma of spinal-cord. Acta Neuropathol 87:537–540 Hassoun J, Soylemezoglu F, Gambarelli D, Figarella-Branger D, Von Ammon K, Kleihues P, 1993, Central neurocytoma—a synopsis of clinical and histological features. Brain Pathol 3:297–306 Kim DG, Chi JG, Parks SH, Chang KH, Lee SH, Jung HW, Kim HJ, Cho BK, Choi KS, Han DH, 1992, Intraventricular neurocytoma - clinicopathological analysis of 7 cases. J Neurosurg 76:759–765 Figarella-Branger D, Soylemezoglu F, Kleihues P, Hassoun J, Cavenee WK, 2000, Central neurocytoma. In: Kleihues P, ed, World health classification of tumours. Tumours of the nervous system. Pathology and genetics. IARC, Lyon, pp 107–109 Sharma MC, Deb P, Sharma S, Sarkar C, 2006, Neurocytoma: a comprehensive review. Neurosurg Rev 29:270–285 Giangaspero F, Cenacchi G, Losi L, Cerasoli S, Bisceglia M, Burger PC, 1997, Extraventricular neoplasms with neurocytoma features—a clinicopathological study of 11 cases. Am J Surg Pathol 21:206–212 Ferreol E, Sawaya R, de Courten-Myers GM(1989, Primary cerebral neuro-blastoma, neurocytoma, in adults. J Neurooncol 7:121–128 Kulkarni V, Rajshekhar V, Haran RP, Chandi SM, 2002, Long-term outcome in patients with central neurocytoma following stereotactic biopsy and radiation therapy. Br J Neurosurg 16:126–132 Sharma MC, Rathore A, Karak AK, Sarkar C, 1998, A study of proliferative markers in central neurocytoma. Pathology 30:355–359 Sharma MC, Sarkar C, Karak AK, Gaikwad S, Mahapatra AK, MehtaVS, 1999, Intraventricular neurocytoma: a clinicopathological study of 20 cases with review of the literature. J ClinNeurosci 6:319– 323 Sharma S, Sarkar C, Gaikwad S, Suri A, SharmaMC, 2005, Primary neurocytoma of the spinal cord: a case report and review of literature. J Neurooncol 74:47–52 Jaiswal S, VijM, Rajput D,Mehrotra A, Jaiswal AK, Srivastava AK, Behari S, Krishnani N, 2011, A clinicopathological, immunohistochemical and neuroradiological study of eight patients with central neurocytoma. J Clin Neurosci 18:334–339 Chen CM, Chen KH, Jung SM, Hsu HC, Wang CM, 2008, Central neurocytoma: 9 case series and review. Surg Neurol 70:204–209 Chen H, Zhou R, Liu J, Tang J, 2012, Central neurocytoma. J Clin Neurosci 19:849–853 Chen CL, Shen CC, Wang J, Lu CH, Lee HT, 2008, Central neurocytoma: a clinical, radiological and pathological study of nine cases. Clin Neurol Neurosurg 110:129–136 Ishiuchi S, Nakazato Y, IinoM, Ozawa S, Tamura M, Ohye C, 1998, In vitro neuronal and glial production and differentiation of human central neurocytoma cells. J Neurosci Res 51:526–535 Ishiuchi S, Tamura M, 1997, Central neurocytoma: an immunohistochemical, ultrastructural and cell culture study. Acta Neuropathol 94:425–435 Kawashima M, Suzuki SO, Doh-ura K, Iwaki T, 2000, alpha- Synuclein is expressed in a variety of brain tumors showing neuronal differentiation. Acta Neuropathol 99:154–160 Nakagawa K, Aoki Y, Sakata K, Sasaki Y,Matsutani M, Akanuma A, 1993, Radiation-therapy of well-differentiated neuroblastoma and central neurocytoma. Cancer 72:1350–1355 Namiki J, Nakatsukasa M, Murase I, Yamazaki K, 1998, Central neurocytoma presenting with intratumoral hemorrhage 15 years after initial treatment by partial removal and irradiation—case report. Neurol Med 38:278–282 Nishio S, Takeshita I, Kaneko Y, Fukui M, 1992, Cerebral neurocytoma—a new subset of benign neuronal tumors of the cerebrum. Cancer 70:529–537 Kim DG, Kim JS, Chi JG, Park SH, Jung HW, Choi KS, Han DH, 1996, Central neurocytoma: proliferative potential and biological behavior. J Neurosurg 84:742–747 Kim DG, Paek SH, Kim IH, Chi JG, Jung HW, Han DH, Choi KS, Cho BK, 1997, Central neurocytoma—the role of radiation therapy and long term outcome. Cancer 79:1995–2002 Kim DH, Suh YL, 1997, Pseudopapillary neurocytoma of temporal lobe with glial differentiation. Acta Neuropathol 94:187–191 Kim DG, Choe WJ, Chang KH, Song IC, Han MH, Jung HW, Cho BK, 2000, In vivo proton magnetic resonance spectroscopy of central neurocytomas. Neurosurgery 46:329–333 Kim CY, Paek SH, Jeong SS, Chung HT, Han JH, Park CK, Jung HW, Kim DG, 2007, Gamma knife radiosurgery for central neurocytoma—primary and secondary treatment. Cancer 110:2276– 2284 Rades D, Schild SE, 2006, Treatment recommendations for the various subgroups of neurocytomas. J Neurooncol 77:305–309 Soylemezoglu F, Scheithauer BW, Esteve J, Kleihues P, 1997, Atypical central neurocytoma. J Neuropathol Exp Neurol 56:551–556 Mackenzie IRA, 1999, Central neurocytoma—histologic atypia, proliferation potential, and clinical outcome. Cancer 85:1606–1610 Lenzi J, SalvatiM, Raco A, FratiA, Piccirilli M, Delfini R, 2006, Central neurocytoma: a novel appraisal of a polymorphic pathology—our experience and a review of the literature. Neurosurg Rev 29:286–292 Rades D, Schild SE, Fehlauer F, 2004, Prognostic value of theMIB- 1 labeling index for central neurocytomas. Neurology 62:987–989 Lee J, Chang SM,McDermottMW, Parsa AT, 2003, Intraventricular neurocytomas. Neurosurg Clin N Am 14:483–508 Paek SH, Han JH, Kim JW, Park CK, Jung HW, Park SH, Kim IH, Kim DG, 2008, Long-term outcome of conventional radiation therapy for central neurocytoma. J Neurooncol 90:25–30 Schild SE, Scheithauer BW,Haddock MG, SchiffD, Burger PC,Wong WW, Lyons MK, 1997, Central neurocytomas. Cancer 79:790–795 Rades D, Fehlauer F, Schild SE, 2004, Treatment of atypical neurocytomas. Cancer 100:814–817 Rades D, Schild SE, Ikezaki K, Fehlauer F, 2003, Defining the optimal dose of radiation after incomplete resection of central neurocytomas. Int J Radiat Oncol Biol Phys 55:373–377 Rades D, Schild SE, 2006, Value of postoperative stereotactic radiosurgery and conventional radiotherapy for incompletely resected typical neurocytomas. Cancer 106:1140–1143 Neto MC, Ramina R, de Meneses MS, Arruda WO, Milano JB, 2003, Peritioneal dissemination from central neurocytoma—case report. Arq Neuropsiquiatr 61:1030–1034 Stapleton CJ, Walcott BP, Kahle KT, Codd PJ, Nahed BV, Chen L, Robison NJ, Delalle I, Goumnerova LC, Jackson EM, 2012, Diffuse central neurocytoma with craniospinal dissemination. J Clin Neurosci 19:163–166 Amini E, Roffidal T, Lee A, Fuller GN, Mahajan A, Ketonen L, Kobrinsky N, Cairo MS, Wells RJ, Wolff JEA, 2008, Central neurocytoma responsive to topotecan, ifosfamide, carboplatin. Pediatr Blood Cancer 51:137–140 Elek G, Slowik F, Eross L, Toth S, Szabo Z, Balint K, 1999, Central neurocytoma with malignant course: neuronal and glial differentiation and craniospinal dissemination. Pathol Oncol Res 5: 155–159 Eng DY, DeMonte F, Ginsberg L, Fuller GN, Jaeckle K, 1997, Craniospinal dissemination of central neurocytoma—report of two cases. J Neurosurg 86:547–552 Brandes AA, Amista P, GardimanM, Volpin L, Danieli D, Guglielmi B, Carollo C, Pinna G, Turazzi S, Monfardini S, 2000, Chemotherapy in patients with recurrent and progressive central neurocytoma. Cancer 88:169–174 Ando K, Ishikura R, Morikawa T, Nakao N, Ikeda J, Matsumoto T, Arita N, 2002, Central neurocytomawith craniospinal dissemination. Magn Reson Med Sci 1:179–182 Tran H, Medina-Flores R, Cerilli LA, Phelps J, Lee FC, Wong G, Turner P, 2010, Primary disseminated central neurocytoma: cytological andMRI evidence of tumor spread prior to surgery. J Neurooncol 100:291–298 Ogawa Y, Sugawara T, Seki H, Sakuma T, 2006, Central neurocytomas with MIB-1 labeling index over 10% showing rapid tumor growth and dissemination. J Neurooncol 79:211–216 Takao H, Nakagawa K, Ohtomo K, 2003, Central neurocytoma with craniospinal dissemination. J Neurooncol 61:255–259 Amagasa M, Yuda F, Sat S, Kojima H, 2008, Central neurocytoma with remarkably large rosette formation and rapid malignant progression: a clinicopathological follow-up study with autopsy report. Clin Neuropathol 27:252–257 Tomura N, Hirano H, Watanabe O, Watarai J, Itoh Y, Mineura K, Kowada M, 1997, Central neurocytoma with clinically malignant behavior. Am J Neuroradiol 18:1175–1178 Cook DJ, Christie SD, Macaulay RJB, Rheaume DE, Holness RO, 2004, Fourth ventricular neurocytoma: case report and review of the literature. Can J Neurol Sci 31:558–564 Bertalanffy A, Roessler K, Koperek O, Gelpi E, Prayer D, Knosp E, 2005, Recurrent central neurocytomas. Cancer 104:135–142 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 319 EP 325 DI 10.1007/s12253-013-9697-y PG 7 ER PT J AU Horvath, K Pete, I Vereczkey, I Dudnyikova, A Godeny, M AF Horvath, Katalin Pete, Imre Vereczkey, Ildiko Dudnyikova, Anna Godeny, Maria TI Evaluation of the Accuracy of Preoperative MRI in Measuring Myometrial Infiltration in Endometrial Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MRI; Endometriumcarcinoma; Myometrial infiltration; Benign uterine lesions ID MRI; Endometriumcarcinoma; Myometrial infiltration; Benign uterine lesions AB The aim of our study was to evaluate the diagnostic performance of magnetic resonance imaging (MRI) in the pretreatment evaluation of myometrium invasion in endometrial cancer. Our retrospective study concerns 89 patients with endometrial cancer, who had preoperative MR evaluation of myometrium invasion and we compared it with histological results. Considering histological type and grade, we had excluded patients with poor prognosis, and separately evaluated those cases where the depth of myometrium invasion is the main prognostic factor determining the choice treatment. Of the 89 cases MRI had accurately evaluated the depth of myometrial invasion in 75 patients. Based on data from all cases, we found the sensitivity of detection of deep myometrial infiltration by MRI (Sv) 71 %, specificity (Sp) 92 %, accuracy (Acc) 84 %, positive predictive value (PPV) 86 % and negative predictive value (NPV) 83 %. Excluding patients with poor prognosis according to histology and grade, these data were Sv 71 %, Sp 95 %, Acc 87 %, PPV 90 %, NPV 84 %. In conclusion, MRI is an efficient diagnostic tool in assessing myometrial infiltration, which is necessary for proper preoperative staging and therapy planning, including evaluation of the necessity of lymphadenectomy. Certain factors may interfere with evaluation of MRI results, thus hindering the precise determination of the level of myometrial infiltration. C1 [Horvath, Katalin] National Institute of Oncology, Center of RadiologyBudapest, Hungary. [Pete, Imre] National Institute of Oncology, Center of GynaecologyBudapest, Hungary. [Vereczkey, Ildiko] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Dudnyikova, Anna] National Institute of Oncology, Center of GynaecologyBudapest, Hungary. [Godeny, Maria] National Institute of Oncology, Center of RadiologyBudapest, Hungary. RP Horvath, K (reprint author), National Institute of Oncology, Center of Radiology, Budapest, Hungary. EM vakhor2@gmail.com CR Bray F, Dos Santos Silva I, Moller H, Weiderpas E, 2005, Endometrial cancer incidence trends in Europe: Underlying determinants and prospects for prevention. Cancer Epidemiol Biomarkers Prev 14:1132–1142 American Cancer Society. Cancer Facts and Figures 2011. http://www.cancer.org/Research/CancerFactsFigures/Cancer FactsFigures/cancer-facts-figures-2011 Hungarian National Cancer Registry. http://www.oncol.hu/rakreg/ stat2/nrr_2010.html Fisher B, Costantino JP,Wickerham DL, Redmond CK, Kavanah M, Cronin WM et al, 1998, Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P- 1 Study. J Natl Cancer Inst 90:1371–1388 Pecorelli S, 2009, Revised FIGO staging for carcinoma of the vulva, cervix, and endometrium. Int J Gynaecol Obstet 105:103–104 Sala E, Wakely S, Senior E, Lomas D, 2007, MRI of Malignant Neoplasms of the Uterine Corpus and Cervix. Am J Roentgenol 188: 1577–1587 Lester SC, 2010, Manual of surgical pathology, 3rd Edition, Elsevier FIGO Committee on Gynecologic Oncology, 2009, Revised FIGO staging for carcinoma of the vulva, cervix and endometrium. Int J Gynecol Obstet 105:103–104 Harris M, Taylor G. Medical Statistics Made Easy, 2nd Edition 2008, Scion. Levine DA, De Los Santos JF, Fleming GF, Barakat RR, Markman M, Randall ME, 2010, Handbook for principles and practice of gynecologic oncology. Lippincott, Williams and Wilkins Chen JK, Cheung MK, Huh WK, Osann K, Husain A, Teng NN, Kapp DS, 2006, Therapeutic role of lymph node resection in endometrioid corpus cancer: a study of 12,333 patients. Cancer 107:1823–1830 Kitchener H, Swart AM, Quian Q, Amos C, Parmar MK, 2009, Efficacy of systematic pelvic lymphadenectomy in endometrial cancer, MRC ASTEC trial): a randomized study. Lancet 373:125– 136 Balleyguier C, Sala E, Da Cuncha T, Bergman A, Brkljacic B, Danza F et al, 2011, Staging of uterine cervical cancer withMRI: guidelines of the European Society of Urogenital Radiology. Eur Radiol 21: 1102–1110 Kinkel K, Forstner R, Danza FM, Oleaga L, Cunha TM, Bergman A et al, 2009, Staging of endometrial cancer with MRI: guidelines of the European Society of Urogenital Imaging; European Society of Urogenital Imaging. Eur Radiol 19:1565–1574 Rockall AG,Merony R, Sohaib SA, Reynolds K, Alexander-Sefre F, Shepherd JH, Jacobs I, Reznek EH, 2007, Evaluation of endometrial carcinoma on magnetic resonance imaging. Int J Gynecol Cancer 17: 188–196 Chung HH, Kang SB, Cho JY, KimJW, ParkNH, Song YS, KimSH, Lee HP, 2007, Accuracy of MR imaging for the prediction of myometrial invasion of endometrial carcinoma. Gynecol Oncol 104:654–659 Rechichi G, Galimberti S, Signorelli M, Perego P, Valsecchi MG, Sironi S, 2010, Myometrial invasion in endometrial cancer: diagnostic performance of diffusion-weighted MR imaging at 1,5-T. Eur Radiol 20:754–762 Nakao Y, Yokoyama M, Hara K, Koyamatsu Y, Yasunaga M, Araki Y, Watanabe Y, Iwasaka T, 2006, MR imaging in endometrial carcinoma as a diagnostic tool for the absence of myometrial invasion. Gynecol Oncol 102:343–347 Ortashi O, Jain S, Emannuel O, Henry R, Wood A, Evans J, 2008, Evaluation of the sensitivity, specificity, positive and negative predictive values of preoperative magnetic resonance imaging for staging endometrial cancer. A prospective study of 100 cases at the Dorset cancer centre. Eur J Obstet Gynecol Reprod Biol 137:232– 235 Hwang JH, Lee KW, Lee JK, 2009, Magnetic resonance imaging for assessment of deep endometrial invasion for patients with endometrial carcinoma. Aust N Z J Obstet Gynaecol 49:537–541 Emlik D, Ozdemir S, Celik C, Karakose S, 2010, Preoperative assessment of myometrial and cervical invasion in endometrial carcinoma: comparison of multi-section dynamic MR imaging using a 3D FLASH technique and T2 weighted MR imaging. J Med Imag Radiat Oncol 54:202–210 McComiskey MH, McCluggage WG, Grey A, Harley I, Dobbs S, Nagar HA, 2012, Diagnostic accuracy of magnetic resonance imaging in endometrial cancer. Int J Gynecol Cancer 22:1020–1025 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 327 EP 333 DI 10.1007/s12253-013-9699-9 PG 7 ER PT J AU Yu, Y Huang, T Wu, Y Ma, X Yu, G Qi, J AF Yu, Yongjiang Huang, Tao Wu, Yu Ma, Xiaorong Yu, Guopeng Qi, Jun TI In-Vitro and In-Vivo Imaging of Prostate Tumor Using NaYF4: Yb, Er Up-Converting Nanoparticles SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NaYF4:Yb; Er; Nanoparticles; Prostate tumor cells; Biological imaging ID NaYF4:Yb; Er; Nanoparticles; Prostate tumor cells; Biological imaging AB The aim of this study was to investigate the feasibility of prostate tumor bioimaging both in vitro and in vivo using an upconversion fluorophore, NaYF4: Yb, Er nanoparticles. Luminescent signals of human prostate cancer cells (CWR22R and LNCaP) labeled with NaYF4: Yb, Er nanoparticles were detected by laser scanning confocal microscope, while Cy3 or FITC was used as control probe. Mouse-human prostate cancer model was developed by subcutaneously injecting the CWR22R cells into BALB/c nude mice to investigate the in-vivo imaging properties of NaYF4:Yb, Er nanoparticles. Both CWR22R and LNCaP cells could phagocytose NaYF4:Yb, Er nanoparticles in vitro, and the cellular uptake of CWR22R cells was much higher than that of LNCaP cells (95.42±3.47% vs. 51.63±6.43 %), which made us choose the former for the further study. CWR22R cells pre-labeled with NaYF4:Yb, Er nanoparticles showed no obvious decrease of fluorescence intensity (P >0.05) after light exposure, while the fluorescence intensity of Cy3 or FITC labeled cells decreased rapidly with prolonged bleaching (P <0.05). Furthermore, the in-vivo results showed that the prostate cancer cells pre-labeled with or without NaYF4:Yb, Er nanoparticles formed tumors 4 weeks after injection, and the tumor length-diameter of the nanoparticle group and the control group was (10.3±2.0) mm and (9.8±2.5) mm, respectively. Significant upconversion fluorescence signals were observed in the tumors of the nanoparticle group when being excited at 980 nmby a NIR laser. In summary, the results suggest that as an intensive fluorescence imaging label agent, NaYF4:Yb, Er nanoparticles possess unique features and can be used for imaging prostate tumor cells both in vitro and in vivo by phagocytosis. C1 [Yu, Yongjiang] Shanghai Jiaotong University School of Medicine, Xinhua Hospital, Department of Urology, No.1665 Kongjiang Road, Yangpu District, 200092 Shanghai, China. [Huang, Tao] Shanghai Jiaotong University School of Medicine, Xinhua Hospital, Department of Urology, No.1665 Kongjiang Road, Yangpu District, 200092 Shanghai, China. [Wu, Yu] Shanghai Jiaotong University School of Medicine, Xinhua Hospital, Department of Urology, No.1665 Kongjiang Road, Yangpu District, 200092 Shanghai, China. [Ma, Xiaorong] Shanghai Jiaotong University School of Medicine, Xinhua Hospital, Department of Plastic Surgery, 200092 Shanghai, China. [Yu, Guopeng] Shanghai Jiaotong University School of Medicine, Xinhua Hospital, Department of Urology, No.1665 Kongjiang Road, Yangpu District, 200092 Shanghai, China. [Qi, Jun] Shanghai Jiaotong University School of Medicine, Xinhua Hospital, Department of Urology, No.1665 Kongjiang Road, Yangpu District, 200092 Shanghai, China. RP Qi, J (reprint author), Shanghai Jiaotong University School of Medicine, Xinhua Hospital, Department of Urology, 200092 Shanghai, China. EM jasonqi@sh163.net CR Ren J, Wang F, Wei G, Yang Y, Liu Y, Wei M, Huan Y, Larson AC, Zhang Z, 2012, MRl of prostate cancer antigen expression for diagnosis and immunotherapy. PLoS One 7(6):e38350., DOI 10. 1371/journal.pone.0038350 Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60(5):277–300., DOI 10.3322/caac.20073caac.20073 Haas GP, Delongchamps N, Brawley OW, Wang CY, de la Roza G, 2008, The worldwide epidemiology of prostate cancer: perspectives from autopsy studies. Can J Urol 15(1):3866–3871 Abdolahi M, Shahbazi-Gahrouei D, Laurent S, Sermeus C, Firozian F, Allen BJ, Boutry S, Muller RN, 2013, Synthesis and in vitro evaluation of MR molecular imaging probes using J591 mAbconjugated SPIONs for specific detection of prostate cancer. Contrast MediaMol Imaging 8(2):175–184., DOI 10.1002/cmmi.1514 Ferrari M, 2005, Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer 5(3):161–171., DOI 10.1038/nrc1566 Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R, 2007, Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol 2(12):751–760., DOI 10.1038/nnano.2007.387 He F, Yang P, Wang D, Li C, Niu N, Gai S, Zhang M, 2011, Preparation and up-conversion luminescence of hollow La2O3:Ln, Ln = Yb/Er, Yb/Ho)microspheres. Langmuir 27(9):5616–5623., DOI 10.1021/la200506q Chatterjee DK, Rufaihah AJ, Zhang Y, 2008, Upconversion fluorescence imaging of cells and small animals using lanthanide doped nanocrystals. Biomaterials 29(7):937–943., DOI 10.1016/j. biomaterials.2007.10.051 Idris NM, Li Z, Ye L, Sim EK, Mahendran R, Ho PC, Zhang Y, 2009, Tracking transplanted cells in live animal using upconversion fluorescent nanoparticles. Biomaterials 30(28):5104–5113., DOI 10. 1016/j.biomaterials.2009.05.062 Yu M, Li F, Chen Z, Hu H, Zhan C, Yang H, Huang C, 2009, Laser scanning up-conversion luminescence microscopy for imaging cells labeled with rare-earth nanophosphors. Anal Chem 81(3):930–935., DOI 10.1021/ac802072d10.1021/ac802072d Xiong L, Chen Z, Tian Q, Cao T, Xu C, Li F, 2009, High contrast upconversion luminescence targeted imaging in vivo using peptidelabeled nanophosphors. Anal Chem 81(21):8687–8694., DOI 10.1021/ ac901960d Wang C, Tao H, Cheng L, Liu Z, 2011, Near-infrared light induced in vivo photodynamic therapy of cancer based on upconversion nanoparticles. Biomaterials 32(26):6145–6154., DOI 10.1016/j. biomaterials.2011.05.007 Mader HS, Kele P, Saleh SM, Wolfbeis OS, 2010, Upconverting luminescent nanoparticles for use in bioconjugation and bioimaging. Curr Opin Chem Biol 14(5):582–596., DOI 10.1016/j.cbpa.2010.08.014 LinM, Zhao Y,Wang S, Liu M, Duan Z, Chen Y, Li F, Xu F, Lu T, 2012, Recent advances in synthesis and surface modification of lanthanide-doped upconversion nanoparticles for biomedical applications. Biotechnol Adv 30(6):1551–1561., DOI 10.1016/j. biotechadv.2012.04.009 Soukka T, Rantanen T, Kuningas K, 2008, Photon upconversion in homogeneous fluorescence-based bioanalytical assays. Ann N Y Acad Sci 1130:188–200., DOI 10.1196/annals.1430.0271130/1/188 Wang F, Liu X, 2009, Recent advances in the chemistry of lanthanide-doped upconversion nanocrystals. Chem Soc Rev 38(4): 976–989., DOI 10.1039/b809132n Lim SF, Ryu WS, Austin RH, 2010, Particle size dependence of the dynamic photophysical properties of NaYF4:Yb, Er nanocrystals. Opt Express 18(3):2309–2316., DOI 10.1364/OE.18.002309195128 Cao T, Yang T, Gao Y, Yang Y, Hu H, Li F, 2010, Water-soluble NaYF 4 : Yb/Er upconversion nanophosphors: synthesis, characteristics and application in bioimaging. Inorg Chem Commun 13(3):392–394 WangM,Mi CC,WangWX, Liu CH,Wu YF, Xu ZR,Mao CB, Xu SK, 2009, Immunolabeling and NIR-excited fluorescent imaging of HeLa cells by using NaYF(4):Yb, Er upconversion nanoparticles. ACS Nano 3(6):1580–1586., DOI 10.1021/nn900491j Egerer K, Roggenbuck D, Hiemann R,WeyerMG, Buttner T, Radau B, Krause R, Lehmann B, Feist E, Burmester GR, 2010, Automated evaluation of autoantibodies on human epithelial-2 cells as an approach to standardize cell-based immunofluorescence tests. Arthritis Res Ther 12(2):R40., DOI 10.1186/ar2949ar2949 Chan KH, Kwan JS, Ho PW, Ho JW, Chu AC, Ramsden DB, 2010, Aquaporin-4 autoantibodies in neuromyelitis optica spectrum disorders: comparison between tissue-based and cell-based indirect immunofluorescence assays. J Neuroinflammation 7:50., DOI 10. 1186/1742-2094-7-501742-2094-7-50 Xiong L, Yu M, ChengM, Zhang M, Zhang X, Xu C, Li F, 2009, A photostable fluorescent probe for targeted imaging of tumour cells possessing integrin alpha(v)beta(3). Mol Biosyst 5(3):241–243., DOI 10.1039/b820576k Yu M, Zhao Q, Shi L, Li F, Zhou Z, Yang H, Yi T, Huang C, 2008, Cationic iridium(III, complexes for phosphorescence staining in the cytoplasm of living cells. Chem Commun, Camb, 18:2115–2117., DOI 10.1039/b800939b Smith AM, Duan H, Mohs AM, Nie S, 2008, Bioconjugated quantum dots for in vivo molecular and cellular imaging. Adv Drug Deliv Rev 60(11):1226–1240., DOI 10.1016/j.addr.2008.03.015 Xiong LQ, Chen ZG, Yu MX, Li FY, Liu C, Huang CH, 2009, Synthesis, characterization, and in vivo targeted imaging of aminefunctionalized rare-earth up-converting nanophosphors. Biomaterials 30(29):5592–5600., DOI 10.1016/j.biomaterials.2009.06.015 Lim SF, Riehn R, RyuWS, Khanarian N, Tung CK, Tank D, Austin RH, 2006, In vivo and scanning electron microscopy imaging of upconverting nanophosphors in Caenorhabditis elegans. Nano Lett 6(2):169–174., DOI 10.1021/nl0519175 Zhang P, Rogelj S, Nguyen K,Wheeler D, 2006, Design of a highly sensitive and specific nucleotide sensor based on photon upconverting particles. J Am Chem Soc 128(38):12410–12411., DOI 10.1021/ja0644024 Chen Z, Chen H, Hu H, YuM, Li F, Zhang Q, Zhou Z, Yi T, Huang C, 2008, Versatile synthesis strategy for carboxylic acid-functionalized upconverting nanophosphors as biological labels. J Am Chem Soc 130(10):3023–3029., DOI 10.1021/ja076151k Bruchez M Jr, Moronne M, Gin P, Weiss S, Alivisatos AP, 1998, Semiconductor nanocrystals as fluorescent biological labels. Science 281(5385):2013–2016 Wu X, Gong S, Roy-Burman P, Lee P, Culig Z, 2013, Current mouse and cellmodels in prostate cancer research. Endocr Relat Cancer., DOI 10.1530/ERC-12-0285 Bocci G, Di Paolo A, Danesi R, 2013, The pharmacological bases of the antiangiogenic activity of paclitaxel. Angiogenesis., DOI 10.1007/ s10456-013-9334-0 WangHZ,Wang HY, Liang RQ, Ruan KC, 2004)Detection of tumor marker CA125 in ovarian carcinoma using quantum dots. Acta Biochim Biophys Sin, Shanghai, 36(10):681–686 Wu X, Liu H, Liu J, Haley KN, Treadway JA, Larson JP, Ge N, Peale F, Bruchez MP, 2003, Immunofluorescent labeling of cancer marker Her2 and other cellular targetswith semiconductor quantumdots. Nat Biotechnol 21(1):41–46., DOI 10.1038/nbt764nbt764 Ness JM, Akhtar RS, Latham CB, Roth KA, 2003, Combined tyramide signal amplification and quantum dots for sensitive and photostable immunofluorescence detection. J Histochem Cytochem 51(8):981–987 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 335 EP 341 DI 10.1007/s12253-013-9700-7 PG 7 ER PT J AU Wang, Sh Lu, Sh Geng, Sh Ma, Sh Liang, Z Jiao, B AF Wang, Shuai Lu, Shengkui Geng, Shaomei Ma, Shucheng Liang, Zhaohui Jiao, Baohua TI Decreased expression of microRNA-206 correlates with poor clinical outcome in patients with malignant astrocytomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microRNA-206; Astrocytoma; Expression; Prognosis ID microRNA-206; Astrocytoma; Expression; Prognosis AB MicroRNA-206 (miR-206) has been proved to function as a tumor suppressor in several types of human malignant cancers. More recently, it has been demonstrated that the ectopic expression of miR-206 significantly inhibited the proliferation and promoted apoptosis at the early stages in glioma cell U343. In order to investigate the clinical significance of miR-206 expression in human astrocytoma, quantitative real-time polymerase chain reaction (qRT-PCR) analysis was used to characterize the expression patterns of miR-206 in 108 astrocytoma and 20 normal brain tissues. As the results, the expression levels of miR-206 in astrocytoma tissues were significantly lower than those in normal brain tissues (P <0.001). Additionally, the decreased expression of miR-206 in astrocytoma was significantly associated with advanced pathological grade (P =0.008), low Karnofsky performance score (KPS, P =0.02), and large tumor size (P=0.01). Moreover, Kaplan-Meier survival and Cox regression analyses showed that low miR-206 expression (P <0.001) and advanced pathological grade (P =0.02) were independent factors predicting poor prognosis for astrocytomas. In conclusion, this is the first report of the differential expression of miR-206 in human astrocytoma tissues. MiR-206 could be a valuable marker of astrocytoma progression and low miR-206 expression is associated with poor overall survival in patients with malignant astrocytomas. C1 [Wang, Shuai] Second Hospital of Hebei Medical University, Department of Neurosurgery, No. 215, Hepingxi Road, 050000 Shijiazhuang, Hebei Province, China. [Lu, Shengkui] Second Hospital of Hebei Medical University, Department of Neurosurgery, No. 215, Hepingxi Road, 050000 Shijiazhuang, Hebei Province, China. [Geng, Shaomei] Second Hospital of Hebei Medical University, Department of Neurosurgery, No. 215, Hepingxi Road, 050000 Shijiazhuang, Hebei Province, China. [Ma, Shucheng] Second Hospital of Hebei Medical University, Department of Neurosurgery, No. 215, Hepingxi Road, 050000 Shijiazhuang, Hebei Province, China. [Liang, Zhaohui] Second Hospital of Hebei Medical University, Department of Neurosurgery, No. 215, Hepingxi Road, 050000 Shijiazhuang, Hebei Province, China. [Jiao, Baohua] Second Hospital of Hebei Medical University, Department of Neurosurgery, No. 215, Hepingxi Road, 050000 Shijiazhuang, Hebei Province, China. RP Jiao, B (reprint author), Second Hospital of Hebei Medical University, Department of Neurosurgery, 050000 Shijiazhuang, China. EM jbhhbey@163.com CR Ohgaki H, Kleihues P, 2005, Epidemiology and etiology of gliomas. Acta Neuropathol 109:93–108 Rousseau A, Mokhtari K, Duyckaerts C, 2008, The 2007 WHO classification of tumors of the central nervous system-what has changed? Curr Opin Neurol 21:720–727 Zhao W, Wang C, Wang J, Ge A, Li Y, Li W, Lu Y, 2011, Relationship between CacyBP/SIP expression and prognosis in astrocytoma. J Clin Neurosci 18:1240–1244 Finkelstein SD, Mohan D, Hamilton RL, Sasatomi E, Swalsky PA, Lieberman FS, 2004, Microdissection-based genotyping assists discrimination of reactive gliosis from glioma. Am J Clin Pathol 121:671–678 Zhang X, Yang H, Gong B, Jiang C, Yang L, 2012, Combined gene expression and protein interaction analysis of dynamic modularity in glioma prognosis. J Neurooncol 107:281–288 Legler JM, Ries LA, Smith MA, Warren JL, Heineman EF, Kaplan RS, Linet MS, 1999, Cancer surveillance series [corrected]: brain and other central nervous system cancers: recent trends in incidence and mortality. J Natl Cancer Inst 91:1382–1390 Ross JS, Carlson JA, Brock G, 2007, miRNA: the new gene silencer. AmJ Clin Pathol 128:830–836 Brown BD, Gentner B, Cantore A, Colleoni S, AmendolaM, Zingale A, Baccarini A, Lazzari G, Galli C, Naldini L, 2007, Endogenous microRNA can be broadly exploited to regulate transgene expression according to tissue, lineage and differentiation state. Nat Biotechnol 25:1457–1467 Jiang L,Mao P, Song L,Wu J, Huang J, Lin C, Yuan J, Qu L, Cheng SY, Li J, 2010, miR-182 as a prognostic marker for glioma progression and patient survival. Am J Pathol 177:29–38 Lages E, Guttin A, El AtifiM, Ramus C, Ipas H, Dupre I, Rolland D, Salon C, Godfraind C, deFraipont F, Dhobb M, Pelletier L, Wion D, Gay E, Berger F, Issartel JP, 2011, MicroRNA and target protein patterns reveal physiopathological features of glioma subtypes. PLoS One 6:e20600 Rao SA, Santosh V, Somasundaram K, 2010, Genome-wide expression profiling identifies deregulated miRNAs in malignant astrocytoma. Mod Pathol 23:1404–1417 Nohata N, Hanazawa T, Enokida H, Seki N, 2012, microRNA-1/ 133a andmicroRNA-206/133b clusters: dysregulation and functional roles in human cancers. Oncotarget 3:9–21 Townley-TilsonWH, Callis TE,Wang D, 2010, MicroRNAs 1, 133, and 206: critical factors of skeletal and cardiac muscle development, function, and disease. Int J Biochem Cell Biol 42:1252–1255 Yan D, Dong Xda E, ChenX,Wang L, Lu C,Wang J,Qu J, Tu L, 2009, MicroRNA-1/206 targets c-Met and inhibits rhabdomyosarcoma development. J Biol Chem 284:29596–29604 Wang X, Ling C, Bai Y, Zhao J, 2011, MicroRNA-206 is associated with invasion and metastasis of lung cancer. Anat Rec, Hoboken, 294:88–92 Missiaglia E, Shepherd CJ, Patel S, Thway K, Pierron G, Pritchard- Jones K, Renard M, Sciot R, Rao P, Oberlin O, Delattre O, Shipley J, 2010, MicroRNA-206 expression levels correlate with clinical behaviour of rhabdomyosarcomas. Br J Cancer 102:1769–1777 Chen X, Yan Q, Li S, Zhou L, Yang H, Yang Y, Liu X, Wan X, 2012, Expression of the tumor suppressor miR-206 is associated with cellular proliferative inhibition and impairs invasion in ERalpha-positive endometrioid adenocarcinoma. Cancer Lett 314:41–53 Kondo N, Toyama T, Sugiura H, Fujii Y, Yamashita H, 2008, miR- 206 Expression is down-regulated in estrogen receptor alpha-positive human breast cancer. Cancer Res 68:5004–5008 Wang R, Hu Y, Song G, Hao CJ, Cui Y, Xia HF, Ma X, 2012, MiR- 206 regulates neural cells proliferation and apoptosis via Otx2. Cell Physiol Biochem 29:381–390 Ciafre SA, Galardi S, Mangiola A, Ferracin M, Liu CG, Sabatino G, Negrini M, Maira G, Croce CM, Farace MG, 2005, Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun 334:1351–1358 Gaur AB, Holbeck SL, Colburn NH, Israel MA, 2011, Downregulation of Pdcd4 by mir-21 facilitates glioblastoma proliferation in vivo. Neuro Oncol 13:580–590 Song G, Zhang Y, Wang L, 2009, MicroRNA-206 targets notch3, activates apoptosis, and inhibits tumor cell migration and focus formation. J Biol Chem 284:31921–31927 Zhou W, Li Y, Wang X, Wu L, Wang Y, 2011, MiR-206-mediated dynamic mechanism of the mammalian circadian clock. BMC Syst Biol 5:141 Dey BK, Gagan J, Dutta A, 2011, miR-206 and -486 induce myoblast differentiation by downregulating Pax7. Mol Cell Biol 31:203–214 Miyachi M, Tsuchiya K, Yoshida H, Yagyu S, Kikuchi K, Misawa A, Iehara T, Hosoi H, 2010, Circulating musclespecific microRNA, miR-206, as a potential diagnostic marker for rhabdomyosarcoma. Biochem Biophys Res Commun 400: 89–93 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 343 EP 348 DI 10.1007/s12253-013-9701-6 PG 6 ER PT J AU Liu, Y Song, J Li, Y Zhao, Y Ju, Q Zhou, G Li, G AF Liu, Yanhong Song, Jie Li, Yuehui Zhao, Yanjie Ju, Qiang Zhou, Guohua Li, Guancheng TI Monoclonal Antibody Preparation and Expression Profile Analysis of a Novel Hepatoma Associated Gene SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Hepatoma associated gene; Monoclonal antibody; Immunohistochemistry; Expression profile ID Hepatocellular carcinoma; Hepatoma associated gene; Monoclonal antibody; Immunohistochemistry; Expression profile AB Hepatoma associated gene (HTA), a gene screened and cloned by our previous research, was specifically expressed in certain kinds of tumors and had a cancer-promoting role in hepatocellular carcinoma (HCC). To further elucidate the mechanism of HTA in hepatoma carcinogenesis and its potential role as a cancer biomarker, refolded HTA protein (HTA) was obtained by prokaryotic recombinant expression system and immobilized metal affinity chromatography. Then anti-HTA monoclonal antibody (mAb) was produced by hybridoma technique. Using the high titer anti-HTA mAb with high specificity obtained, the expression profile of HTA was analysed by immunohistochemistry staining. It showed that HTA expressed specifically in some kinds of tumors, and didn’t express in almost any of the normal tissues. The positive expression rate and expression quantity of HTA was significantly higher in HCC tissues than in hepatic cirrhosis tissues, hepatic fibrosis tissues and normal hepatic tissues. The expression of HTA was positively correlated with hepatoma carcinogenic process. C1 [Liu, Yanhong] Key Laboratory of Carcinogenesis Ministry of Health, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, 410078 Changsha, Hunan, China. [Song, Jie] Key Laboratory of Carcinogenesis Ministry of Health, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, 410078 Changsha, Hunan, China. [Li, Yuehui] Key Laboratory of Carcinogenesis Ministry of Health, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, 410078 Changsha, Hunan, China. [Zhao, Yanjie] Key Laboratory of Carcinogenesis Ministry of Health, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, 410078 Changsha, Hunan, China. [Ju, Qiang] Key Laboratory of Carcinogenesis Ministry of Health, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, 410078 Changsha, Hunan, China. [Zhou, Guohua] Key Laboratory of Carcinogenesis Ministry of Health, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, 410078 Changsha, Hunan, China. [Li, Guancheng] Key Laboratory of Carcinogenesis Ministry of Health, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, 410078 Changsha, Hunan, China. RP Li, G (reprint author), Key Laboratory of Carcinogenesis Ministry of Health, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion Ministry of Education, 410078 Changsha, China. EM libsun@163.com CR El-Serag HB, 2011, Hepatocellular carcinoma. N Engl J Med 365(12):1118–1127 Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics, 2012. CA Cancer J Clin 62(1):10–29 DuBray BJ, Chapman WC, Anderson CD, 2011, Hepatocellular carcinoma: a review of the surgical approaches to management. Mo Med 108(3):195–198 Michor F, Iwasa Y, Nowak MA, 2004, Dynamics of cancer progression. Nat Rev Cancer 4(3):197–205 Fartoux L, Decaens T, 2011, Contribution of biomarkers and imaging in the management of hepatocellular carcinoma. Clin Res Hepatol Gastroenterol 35(Suppl 1):S21–S30 Jia HL, Ye QH, Qin LX et al, 2007, Gene expression profiling reveals potential biomarkers of human hepatocellular carcinoma. Clin Cancer Res 13(4):1133–1139 Lee IN, Chen CH, Sheu JC et al, 2005, Identification of human hepatocellular carcinoma-related biomarkers by two-dimensional difference gel electrophoresis and mass spectrometry. J Proteome Res 4(6):2062–2069 Steel LF, Shumpert D, Trotter M et al, 2003, A strategy for the comparative analysis of serum proteomes for the discovery of biomarkers for hepatocellular carcinoma. Proteomics 3(5):601–609 Marrero JA, Lok AS, 2004, Newer markers for hepatocellular carcinoma. Gastroenterology 127(5 Suppl 1):S113–S119 Qin LX, Tang ZY, 2004, Recent progress in predictive biomarkers for metastatic recurrence of human hepatocellular carcinoma: a review of the literature. J Cancer Res Clin Oncol 130(9):497–513 Liu Y, Li Y, Guo F et al, 2010, Identification of HTA as a novelspecific marker for human hepatocellular carcinoma. J Cancer Res Clin Oncol 136(8):1187–1192 Liu Y, Zhao Y, Ju Q et al, 2013, Molecular clone and functional study of a novel hepatoma associated gene. Int J Oncol 42(3): 1105–1112 Clement JM, Jehanno M, Popescu O et al, 1996, Expression and biological activity of genetic fusions between MalE, the maltose binding protein from Escherichia coli and portions of CD4, the Tcell receptor of the AIDS virus. Protein Expr Purif 8(3):319–331 SpangfortMD, Ipsen H, Sparholt SH et al, 1996, Characterization of purified recombinant Bet v 1 with authentic N-terminus, cloned in fusion with maltose-binding protein. Protein Expr Purif 8(3):365– 373 Li Y, 2011, Recombinant production of antimicrobial peptides in Escherichia coli: a review. Protein Expr Purif 80(2):260–267 Eiberle MK, Jungbauer A, 2010, Technical refolding of proteins: Do we have freedom to operate? Biotechnol J 5(6):547–559 Burgess RR, 2009, Refolding solubilized inclusion body proteins. Methods Enzymol 463:259–282 Kohler G, Milstein C, 1975, Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256(5517):495– 497 Gerdes J, Lemke H, Baisch H et al, 1984, Cell cycle analysis of a cell proliferation-associated human nuclear antigen defined by the monoclonal antibody Ki-67. J Immunol 133(4):1710–1715 Kolaskar AS, Tongaonkar PC, 1990, A semi-empirical method for prediction of antigenic determinants on protein antigens. Febs Lett 276(1–2):172–174 Tong JC, Tan TW, Ranganathan S, 2007, Methods and protocols for prediction of immunogenic epitopes. Brief Bioinform 8(2):96–108 Zhang L, Udaka K,Mamitsuka H et al, 2012, Toward more accurate pan-specific MHC-peptide binding prediction: a review of current methods and tools. Brief Bioinform 13(3):350–364 Feitelson MA, Reis HM, Tufan NL et al, 2009, Putative roles of hepatitis B x antigen in the pathogenesis of chronic liver disease. Cancer Lett 286(1):69–79 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 349 EP 356 DI 10.1007/s12253-013-9703-4 PG 8 ER PT J AU Hunyadi, J Andras, Cs Szabo, I Szanto, J Szluha, K Sipka, S Kovacs, P Kiss, A Szegedi, Gy Altorjay, I Sapy, P Antal-Szalmas, P Toth, L Fazekas, Gy Rajnavolgyi, AF Hunyadi, Janos Andras, Csilla Szabo, Imre Szanto, Janos Szluha, Kornelia Sipka, Sandor Kovacs, Peter Kiss, Attila Szegedi, Gyula Altorjay, Istvan Sapy, Peter Antal-Szalmas, Peter Toth, Laszlo Fazekas, Gyorgy Rajnavolgyi, Eva TI Autologous Dendritic Cell Based Adoptive Immunotherapy of Patients with Colorectal Cancer—A Phase I-II Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Dendritic cell; Vaccination; Colorectal cancer; Autologous ID Dendritic cell; Vaccination; Colorectal cancer; Autologous AB Dendritic cell-based active immunotherapies of cancer patients are aimed to provoke the proliferation and differentiation of tumor-specific CD4+ and CD8+ Tlymphocytes towards protective effector cells. Isolation and in vitro differentiation of circulating blood monocytes has been established a reasonable platform for adoptively transferred DC-based immunotherapies. In the present study the safety and tolerability of vaccination by autologous tumor cell lysates (oncolysate)- or carcinoembriogenic antigen (CEA)-loaded DCs in patients with colorectal cancer was investigated in a phase I-II trial. The study included 12 patients with histologically confirmed colorectal cancer (Dukes B2-C stages). Six of the patients received oncolysate-pulsed, whereas the other six received recombinant CEA-loaded autologous DCs. The potential of the tumor antigen-loaded DCs to provoke the patient’s immune system was studied both in vivo and in vitro. The clinical outcome of the therapy evaluated after 7 years revealed that none of the six patients treated with oncolysate-loaded DCs showed relapse of colorectal cancer, whereas three out of the six patients treated with CEA-loaded DCs died because of tumor relapse. Immunization with both the oncolysate- and the CEA-loaded autologous DCs induced measurable immune responses, which could be detected in vivo by cutaneous reactions and in vitro by lymphocyte proliferation assay. Our results show that vaccination by autologous DCs loaded with autologous oncolysates containing various tumor antigens represents a well tolerated therapeuticmodality in patients with colorectal cancer without any detectable adverse effects. Demonstration of the efficacy of such therapy needs further studies with increased number of patients. C1 [Hunyadi, Janos] University of Debrecen, Medical and Health Science Center, Clinical Centre for Cell Therapy, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Andras, Csilla] University of Debrecen, Department of Oncology, Nagyerdei krt. 98, 4012 Debrecen, Hungary. [Szabo, Imre] University of Debrecen, Department of Oncology, Nagyerdei krt. 98, 4012 Debrecen, Hungary. [Szanto, Janos] University of Debrecen, Department of Oncology, Nagyerdei krt. 98, 4012 Debrecen, Hungary. [Szluha, Kornelia] University of Debrecen, Department of Oncology, Nagyerdei krt. 98, 4012 Debrecen, Hungary. [Sipka, Sandor] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Kovacs, Peter] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Kiss, Attila] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Szegedi, Gyula] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Altorjay, Istvan] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Sapy, Peter] University of Debrecen, Department of Surgery and Operative TechniquesDebrecen, Hungary. [Antal-Szalmas, Peter] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Toth, Laszlo] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Fazekas, Gyorgy] University of Debrecen, Medical and Health Science Center, Clinical Centre for Cell Therapy, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Rajnavolgyi, Eva] University of Debrecen, Faculty of Medicine, Division of Clinical ImmunologyDebrecen, Hungary. RP Szabo, I (reprint author), University of Debrecen, Department of Oncology, 4012 Debrecen, Hungary. EM szabo.imre@med.unideb.hu CR Van-den-Eynde BJ, Van-der-Bruggen P, 1997, T cell defined tumor antigens. Curr Opin Immunol 9:684–693 Salgaller ML, Thurner M, Bartsch G, Boynton AL, Murphy GP, 1999, Report from the International Union Agaist Cancer, UICC, Tumor Biology Committee: UICC Workshop on the use of dendritic cells in cancer clinical trials. Cancer 86:2674–2683 Sokolof MH, Vogelzang N, 1999, The ongoing evolution of dendritic cell therapy. Cancer 86:2593–2596 Knuth A, Wolfel T, Klehmann E, Boon T, Buschenfelde KM, 1989, Cytolytic T-cell clones against an autologous human melanoma: specificity study and definition of three antigens by immunoselection. Proc Natl Acad Sci U S A 86:2804–2808 Davis ID, Jefford M, Parente P, Cebon J, 2003, Rational approaches to human cancer immunotherapy. J Leuk Biol 73:3–29 Liviu V, Titu E, John R, Monson E, Greenman J, 2002, The role of CD8+ T cells in immune responses to colorectal cancer. Cancer Immunol Immunother 51:235–247 Rock KL, Gamble S, Rothstein L, 1990, Presentation of exogenous antigen with class I major histocompatibility complex molecules. Science 249:918–921 Tatsumi T, Takehara T, Kanto T, Miyagi T, Kuzushita N, Sugimoto Y, Jinushi M, Kasahara A, Sasaki Y, Hori M, Hayashi N, 2001, Adminstration of interleukin 12 enhances the therapeutic efficacy of dendritic cell-based tumor vaccines in mouse hepatocellular carcinoma. Cancer Res 61:7563–7567 Tuting T, Wilson CC, Martin DM, Kasamon YL, Rowles J, Ma DI, Slingluff CLJ,Wagner SN, Bruggen P, Baar J, LotzeMT, StorkusWJ, 1998, Autologous human monocyte-derived dendritic cells genetically modified to express melanoma antigens elicit primary cytotoxic T cell responses in vitro: enhancement by co-transfection of genes encoding the Th1-biasing cytokines IL-12 and IFN alfa. J Immunol Methods 160:1139–1147 Shimizu K, Fields RC, Giedlin M, Mule JJ, 1999, Systemic adminstration of interleukin 2 enhances the therapeutic effficacy of dendritic cell-based tumor vaccine. Proc Natl Acad Sci USA 96:2268– 2273 Chen S, Akbar SM, Tanimoto K, Ninomiya T, Iuchi H, Michitaka K, Horiike N, Onji M, 2000, Absence of CD83(+)-positive mature and activated dendritic cells at cancer nodules from patients with hepatocellular carcinoma: relevance to hepatocarcinogenesis. Cancer Lett 148:49–57 Van-de-Velde AL, Berneman ZN, Van-Tendeloo VF, 2008, Immunotherapy of hematological malignancies using dendritic cells. Bull Cancer 95:320–326 Draube A, Klein-Gonzalez N, Mattheus S, Brillant C, Hellmich M, EngertA, von Bergwelt-BaildonM(2011, Dendritic cell based tumor vaccination in prostate and renal cell cancer: a systematic review and meta-analysis. LoS One 6:e18801 Alexandrescu DT, Ichim TE, Riordan NH, Marincola FM, Di-Nardo A, Kabigting FD, Dasanu CA, 2010, Immunotherapy for melanoma: current status and perspectives. J Immunother 33:570–590 Mazzolini G, Murillo O, Atorrasagasti C, Dubrot J, Tirapu I, Rizzo M, Arina A, Alfaro C, Azpilicueta A, Berasain C, Perez-Gracia JL, Gonzales A, Melero I, 2007, Immunotherapy and immunoescape in colorectal cancer. World J Gastroenterol 13:5822–5831 Dauer M, Schnurr M, Eigler A, 2008, Dendritic cell-based cancer vaccination: quo vadis? Expert Rev Vaccines 7:1041–1053 Nencioni A, Grunebach F, Schmidt SM, Muller MR, Boy D, Patrone F, Ballestrero A, Brossart P, 2008, The use of dendritic cells in cancer immunotherapy. Crit Rev Oncol Hematol 65:191–199 Vulink A, Radford KJ, Melief C, Hart DN, 2008, Dendritic cells in cancer immunotherapy. Adv Cancer Res 99:363–407 Erreni M, Mantovani A, Allavena P, 2011, Tumor-associated Macrophages, TAM, and Inflammation in Colorectal Cancer. Cancer Microenviron 4:141–154 Roxburgh C, McMillan D, 2010, Role of systemic inflammatory response in predicting survival in patients with primary operable cancer. Future Oncol 6:149–163 Roxburgh C, McMillan D, 2012, The role of the in situ local inflammatory response in predicting recurrence and survival in patients with primary operable colorectal cancer. Cancer Treat Rev 38(5):451–466., DOI 10.1016/j.ctrv.2011.09.001, Epub 2011 Sep 25 database-online http://www.cancer.org/Cancer/Colonand RectumCancer/DetailedGuide/colorectal-cancer-survival-rates. Last Medical Review: 05/24/2012; Last Revised: 01/17/2013 Palucka K, Banchereau J, Mellman I, 2010, Designing vaccines based on biology of human dendritic cell subsets. Immunity 33:464–478 Nestle F, Banchereau J, Hart D, 2001, Dendritic cells: on the move from bench to bedside. Nat Med 7:761–765 Figdor C, Jd V, Lesterhuis W, Melief C, 2004, Dendritic cell immunotherapy: mapping the way. Nat Med 10:475–480 Dhodapkar MV, Krasovsky J, Steinman RM, Bhardwaj N, 2000, Mature dendritic cells boost functionally superior CD8(+, Tcell in humans without foreign helper epitopes. J Clin Invest 105:9– 14 Jonuleit H, Giesecke-Tuettenberg A, Tuting T, Thurner-Schuler B, Stuge TB, Paragnik L, Kandemir A, Lee PP, Schuler G, Knop J, Enk AH, 2001, A comparison of two types of dendritic cells as adjuvants for the induction of melanoma-specific T-cell responses in humans following intranodal injection. Int J Cancer 93:243–251 Banchereau J, Palucka AK, DhodapkarM, Burkeholder S, Taquet N, Rolland A, Taquet S, Coquery S, Wittkowski KM, Bhardwaj N, 2001, Immune and clinical responses in patients with metastatic melanoma to CD34(+, progenitro-derived dendritic cell vaccine. Cancer Res 61:6451–6458 Schuler-Thurnmer B, Schultz ES, Berger TG, Weinlich G, Ebner S, Woerl P, Bender A, Feuerstein B, Fritsch PO, Romani N, Schuler G, 2002, Rapid induction of tumor-specific type 1 T helper cells in metastatic melanoma patients by vaccination with mature cryopreserved, peptide-loaded monocyte-derived dendritic cells. J Exp Med 195:1279–1288 Martin-Fontecha A, Sebastiani S, Hopken U, Uguccioni M, Lipp M, Lanzavecchia A, Sallusto F, 2003, Regulation of dendritic cell migration to the draining lymph node: impact on T lymphocyte traffic and priming. J Exp Med 198:615–621 Jefford M, Maraskovsky E, Cebon J, Davies ID, 2001, The use of dendritic cells in cancer therapy. Lancet Oncol 2(6):343–353 Steinman RM, Dhodapkar M, 2001, Active immunization against cancer with dendritic cells: the near future. Int J Cancer 94:459–473 Cerundolo V, Hermans I, Salio M, 2004, Dendritic cells: a journey from laboratory to clinic. Nat Immunol 5:7–10 Cranmer L, Trevor K, Hersh E, 2004, Clinical applications of dendritic cell vaccination in the treatment of cancer. Cancer Immunol Immunother 53:275–306 Kavanagh B, Ko A, Venook A, Margolin K, Zeh H, Lotze M, Schillinger B, Liu W, Lu Y, Mitsky P, Schilling M, Bercovici N, Loudovaris M, Guillermo R, Lee SM, Bender J, Mills B, Fong L, 2007, Vaccination of metastatic colorectal cancer patients with matured dendritic cells loaded withmultiplemajor histocompatibility complex class I peptides. J Immunother 30:762–772 Mocellin S, Rossi C, Lise M, Nitti D, 2004, Colorectal cancer vaccines: principles, results, and perspectives. Gastroenterology 127:1821–1837 Mosolits S, Ullenhag G, Mellstedt H, 2005, Therapeutic vaccination in patients with gastrointestinal malignancies. A review of immunological and clinical results. Ann Oncol 16:847–862 Nagorsen D, Thiel E, 2006, Clinical and immunologic responses to active specific cancer vaccines in human colorectal cancer. Clin Cancer Res 12:3064–3069 Ridgway D, 2003, The first 1000 dendritic cell vaccinees. Cancer Invest 21:873–886 Rosenberg S, 2004, Development of effective immunotherapy for the treatment of patients with cancer. J Am Coll Surg 198:685–696 Rosenberg S, Yang J, Restifo N, 2004, Cancer immunotherapy: moving beyond current vaccines. Nat Med 10:909–915 Lesterhuis WJ, De Vries IJ, Schreibelt G, Schuurhuis DH, Aarntzen EH, De Boer A, Scharenborg NM, Van De Rakt M, Hesselink EJ, Figdor CG, Adema GJ, Punt CJ, 2010, Immunogenicity of dendritic cells pulsed with CEA peptide or transfected with CEA mRNA for vaccination of colorectal cancer patients. Anticancer Res 30:5091– 5097 Okuno K, Sugiura F, Itoh K, Yoshida K, Tsunoda T, Nakamura Y, 2012, Recent advances in active specific cancer vaccine treatment for colorectal cancer. Curr Pharm Biotechnol(Feb 14), PMID: 22339221 Sakakibara M, Kanto T, Hayakawa M, Kuroda S, Miyatake H, Itose I, Miyazaki M, Kakita N, Higashitani K, Matsubara T, Hiramatsu N, Kasahara A, Takehara T, Hayashi N, 2011, Comprehensive immunological analyses of colorectal cancer patients in the phase I/ II study of quickly matured dendritic cell vaccine pulsed with carcinoembryonic antigen peptide. Cancer Immunol Immunother 60:1565–1575 Zhou Q, Peng R, Wu X, Xia Q, Hou J, Ding Y, Zhou Q, Zhang X, Pang Z, Wan D, Zeng Y, Zhang X, 2010, The density of macrophages in the invasive front is inversely correlated to liver metastasis in colon cancer. J Transl Med 8:13 Nagaraj S, Gabrilovich DI, 2012, Regulation of suppressive function ofmyeloid-derived suppressor cells byCD4(+, Tcells. Semin Cancer Biol 22(4):282–288., DOI 10.1016/j.semcancer.2012.01.010, Epub 2012 Jan 31 Erdman S, Sohn J, Rao V, Nambiar P, Ge Z, Fox J, Schauer D, 2005, CD4+CD25+ regulatory lymphocytes induce regression of intestinal tumors in ApcMin/+ mice. Cancer Res 65:3998–4004 Chaput N, Louafi S, Bardier A, Charlotte F, Vaillant J, Menegaux F, Rosenzwajg M, Lemoine F, Klatzmann D, Taieb J, 2009, Identification of CD8+CD25+Foxp3+ suppressive T cells in colorectal cancer tissue. Gut 58:520–529 NagorsenD,Voigt S, Berg E, SteinH, Thiel E, Loddenkemper C, 2007, Tumor-infiltrating macrophages and dendritic cells in human colorectal cancer: relation to local regulatory T cells, systemic T-cell response against tumor-associated antigens and survival. J Transl Med 29:62 Gilboa E, 2001, The risk of autoimmunity asssociated with tumor immunotherapy. Nat Immunol 2:789–792 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 357 EP 365 DI 10.1007/s12253-013-9704-3 PG 9 ER PT J AU Valibeigi, B Amirghofran, Z Golmoghaddam, H Hajihosseini, R Kamazani, MF AF Valibeigi, Behnaz Amirghofran, Zahra Golmoghaddam, Hossein Hajihosseini, Reza Kamazani, M Fatemeh TI Fas Gene Variants in Childhood Acute Lymphoblastic Leukemia and Association with Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute lymphoblastic leukemia; Fas; Polymorphism ID Acute lymphoblastic leukemia; Fas; Polymorphism AB Fas molecule is one of the main important molecules involved in apoptotic cell death. Single nucleotide polymorphisms in the promoter of Fas gene at positions −1377G/A and −670 A/G may affect its expression and play an important role in the pathology of leukemia. In the present study the association between these polymorphisms and risk of the development of acute lymphoblastic leukemia (ALL) in children with ALL compared to cancer-free control subjects was examined by polymerase chain reaction- based restriction fragment length polymorphism. The relationship between the polymorphisms and clinical and laboratory features of the patients and response to therapy were determined. No significant differences in genotype and allele frequencies between the patients and the control subjects at positions −670 and −1377 were detected. Evaluation of the prognostic factors revealed an association between the GG genotype at position −670 and liver involvement in ALL patients (p < 0.04). Although patients with −1377 AA genotype showed shorter mean complete remission duration, the result of survival analysis did not reach to be significant. In conclusion, results of this study showed no contribution of Fas genotypes at positions −670 and −1377 to risk of ALL in children. The association of Fas GG genotype at position −670 with liver involvement in the patients may show its important role in prognosis of ALL. C1 [Valibeigi, Behnaz] Payame Noor University, Tehran CenterTehran, Iran. [Amirghofran, Zahra] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Golmoghaddam, Hossein] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. [Hajihosseini, Reza] Payame Noor University, Tehran CenterTehran, Iran. [Kamazani, M Fatemeh] Medical School, Shiraz University of Medical Sciences, Department of ImmunologyShiraz, Iran. RP Amirghofran, Z (reprint author), Medical School, Shiraz University of Medical Sciences, Department of Immunology, Shiraz, Iran. EM amirghz@sums.ac.ir CR Daneshbod Y, Amirghofran Z, Tabei SZ, 2005, Bcl-2 expression in acute myelogenous leukemia: the relation to myeloid antigen expression and response to therapy in Iranian patients. Neoplasma 52:109–114 Amirghofran Z, Monabati A, Gholijani N, 2004, Androgen receptor expression in relation to apoptosis and the expression of cell cycle related proteins in prostate cancer. Pathol Oncol Res 10:37–41 Kaufmann T, Strasser A, Jost PJ, 2012, Fas death receptor signalling: roles of Bid and XIAP. Cell Death Differ 19:42–50 Lavrik IN, Krammer PH, 2012, Regulation of CD95/Fas signaling at the DISC. Cell Death Differ 19:36–41 Itoh N,Yonehara S, 2000, The polypeptide encoded by the cDNAfor human cell surface antigen FAS can mediate apoptosis. Cell 66:233– 243 Inazawa J, Itoh N, Abe T, Nagata S, 1992, Assignment of the human Fas antigen gene, Fas, to 10q24.1. Genomics 14:821–822 Behrmann I,Walczak H,Krammer PH, 1994, Structure of the human APO-1 gene. Eur J Immunol 24:3057–3062 MuschenM,Warskulat U, BeckmannMW(2000, Defining CD95 as a tumor suppressor gene. J Mol Med 78:312–325 Sibley K, Rollinson S, Allan JM et al, 2003, Functional Fas promoter polymorphism are associated with increased risk of acute myeloid leukemia. Cancer Res 63:4327–4330 Wu J, Siddiqui J, Nihal M, Vonderheid EC et al, 2011, Structural alterations of the FAS gene in cutaneous T-cell lymphoma, CTCL). Arch Biochem Biophys 15:185–191 Zhang Z, Xue H, GongWet al, 2009, FAS promoter polymorphisms and cancer risk: a meta-analysis based on 34 case-control studies. Carcinogenesis 30:487–493 Lei D, Sturgis EM, Wang LE et al, 2010, FAS and FASLG genetic variants and risk for second primary malignancy in patients with squamous cell carcinoma of the head and neck. Cancer Epidemiol Biomarkers Prev 19:1484–1491 Cao Y, Miao XP, Huang MY et al, 2010, Polymorphisms of death pathway genes FAS and FASL and risk of nasopharyngeal carcinoma. Mol Carcinog 49:944–950 Shao P, Ding Q, Qin C et al, 2011, Functional polymorphisms in cell death pathway genes FAS and FAS ligand and risk of prostate cancer in a Chinese population. Prostate 71:1122–1130 Zhang W, Li C, Wang J, He C, 2012, Functional polymorphisms in FAS/FASL system contribute to the risk of occurrence but not progression of gastric cardiac adenocarcinoma. Hepatogastroenterology 59:141–146 Zhang X, Miao X, Sun T et al, 2005, Functional polymorphisms in cell death pathway genes Fas and FasL contribute to risk of lung cancer. J Med Genet 42:479–484 Scholl V, Stefanoff CG, Hassan R et al, 2007, Spector N, Renault IZ. Mutations within the 5′ region of FAS/CD95 gene in nodal diffuse large B-cell lymphoma. Leuk Lymphoma 48: 957–963 Peter AM, Kohfink B, Martin H et al, 1999, Defective apoptosis due to a point mutation in the death domain of CD95 associated with autoimmune lymphoproliferative syndrome, T-cell lymphoma, and Hodgkin’s disease. Exp Hematol 27:868–874 Landowski TH, Qu N, Buyuksal I, Painter JS, Dalton WS, 1997, Mutations in the Fas antigen in patients with multiple myeloma. Blood 90:4266–4270 Farre L, Bittencourt AL, Silva-Santos G et al, 2008, Fas 670 promoter polymorphism is associated to susceptibility, clinical presentation, and survival in adult T cell leukemia. J Leukoc Biol 83:220–222 Nair RR, Khanna A, Singh K, 2012, Association of FAS −1377 G> AandFAS−670A>Gfunctional polymorphisms of FAS gene of cell death pathway with recurrent early pregnancy loss risk. J Reprod Immunol 93:114–118 Zhao XF, Reitz M, Chen QC, Stass S, 2011, Pathogenesis of early leukemia and lymphoma. Cancer Biomark 9:341–374 Traver D, Akashi K,Weissman IL, Lagasse E, 1998, Mice defective in two apoptosis pathways in the myeloid lineage develop acute myeloblastic leukemia. Immunity 9:47–57 Chavez-Galan L, Arenas-Del AngelMC, Zenteno E et al, 2009, Cell death mechanisms induced by cytotoxic lymphocytes. Cell Mol Immunol 6:15–25 Arasteh JM, Sarvestani EK, Aflaki E, Amirghofran Z, 2010, Fas gene polymorphisms in systemic lupus erythematosus and serum levels of some apoptosis-related molecules. Immunol Investig 39: 27–38 Villa-Morales M, Fernandez-Piqueras J, 2012, Targeting the Fas/ FasL signaling pathway in cancer therapy. Expert Opin Ther Targets 16:85–101 Rozenfeld-GranotG, Toren A,AmariglioN(2001)Mutation analysis of the FAS and TNFR apoptotic cascade genes in hematological malignancies. Exp Hematol 29:228–233 Beltinger C, Kurz E, Bohler T, 1998, CD95, APO-1/Fas, mutations in childhood T-lineage acute lymphoblastic leukemia. Blood 15: 3943–3951 Beltinger C, Bohler T, Karawajew L et al, 1998)Mutation analysis of CD95, APO-1/Fas, in childhood B-lineage acute lymphoblastic leukaemia. Br J Haematol 102:722–728 Kim HJ, Jin XM, Kim HN et al, 2010, Fas and FasL polymorphisms are not associated with acutemyeloid leukemia risk in Koreans.DNA Cell Biol 29:619 Mehta PA, Gerbing RB, Alonzo TA et al, 2008, FAS promoter polymorphism: outcome of childhood acute myeloid leukemia. A children’s oncology group report. Clin Cancer Res 1:7896–7899 Amirghofran Z, Daneshbod Y, Gholijani N, Esmaeilbeig M, 2011, The influence of Bcl-2 and myeloid antigen expression on response to therapy in childhood acute lymphoblastic leukemia. Arch IranMed 14:170–174 Amirghofran Z, Daneshbod Y, Gholijani N, 2009, Bcl-2 in combination tomyeloid antigen expression in adult acute lymphoblastic leukemia and prognostic outcome. Oncol Res 17:447–454 Fathi M, Amirghofran Z, Shahriari M, 2012, Soluble Fas and Fas ligand and prognosis in children with acute lymphoblastic leukemia. Med Oncol 29:2046–2052 Sunter NJ, Scott K, Hills R et al, 2012, A functional variant in the core promoter of the CD95 cell death receptor gene predicts prognosis in acute promyelocytic leukemia. Blood 5:196–205 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 367 EP 374 DI 10.1007/s12253-013-9705-2 PG 8 ER PT J AU Cai, T Xiao, J Wang, Zf Liu, Q Wu, H Qiu, Yz AF Cai, Tao Xiao, Jie Wang, Zhi-fei Liu, Qiang Wu, Hao Qiu, Yuan-zheng TI Identification of Differentially Coexpressed Genes in Gonadotrope Tumors and Normal Pituitary Using Bioinformatics Methods SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gene; Gonadotrope tumors; Pituitary; Bioinformatics ID Gene; Gonadotrope tumors; Pituitary; Bioinformatics AB To investigate the underlying molecular mechanisms of pituitary tumor by using the microarray expression profiles of pituitary tumor and normal tissue samples. The gene expression profile of GSE26966 was downloaded from Gene Expression Omnibus, including nine normal samples and 14 pituitary tumor samples. The differentially coexpressed genes (DEGs) were identified by Affy package in R Software. The functional and pathway enrichment analysis of the screened DEGs were performed by DAVID. Then, differential coexpression networks were contructed and further analyzed. Functional and pathway enrichment analysis of the 1220 identified DEGs revealed that phosphatidylinositol signaling system, p53 signaling pathway and inositol phosphate metabolism were disturbed in pituitary tumors. The degree of DLK1, CDKN2A and ITGA4 in the constructed differential coexpression network was 46, 45 and 44, respectively. In addition, MPP2 and ASAP2 were the obvious hub genes in the constructed differential coexpression network. Through exploring genes in the differential coexpression networks, the results suggested that DLK1, CDKN2A, ITGA4, MPP2 and ASAP2 may potentially be used as biomarkers for pituitary tumor. C1 [Cai, Tao] The First Xiangya Hospital of Central South University, Department of OtolaryngologyChangsha, Hunan Province, China. [Xiao, Jie] The Third Xiangya Hospital of Central South University, Department of EmergencyChangsha, Hunan Province, China. [Wang, Zhi-fei] The Third Xiangya Hospital of Central South University, Department of NeurosurgeryChangsha, Hunan Province, China. [Liu, Qiang] The Third Xiangya Hospital of Central South University, Department of NeurosurgeryChangsha, Hunan Province, China. [Wu, Hao] The Third Xiangya Hospital of Central South University, Department of NeurosurgeryChangsha, Hunan Province, China. [Qiu, Yuan-zheng] The First Xiangya Hospital of Central South University, Department of OtolaryngologyChangsha, Hunan Province, China. RP Qiu, Yz (reprint author), The First Xiangya Hospital of Central South University, Department of Otolaryngology, Changsha, China. EM songyechun@hotmail.com CR Michaelis KA, Knox AJ, Xu M, Kiseljak-Vassiliades K, Edwards MG, GeraciM, Kleinschmidt-DeMasters BK, Lillehei KO,Wierman ME, 2011, Identification of growth arrest and DNA-damageinducible gene beta, GADD45beta, as a novel tumor suppressor in pituitary gonadotrope tumors. Endocrinology 152(10):3603–3613., DOI 10.1210/en.2011-0109 Alexander JM, BillerBM, BikkalH, Zervas NT, Arnold A, Klibanski A, 1990, Clinically nonfunctioning pituitary tumors are monoclonal in origin. J Clin Invest 86(1):336–340., DOI 10.1172/JCI114705 Pivonello R, Matrone C, Filippella M, Cavallo LM, Di Somma C, Cappabianca P, Colao A, Annunziato L, Lombardi G, 2004, Dopamine receptor expression and function in clinically nonfunctioning pituitary tumors: comparison with the effectiveness of cabergoline treatment. J Clin Endocrinol Metab 89(4):1674–1683 Yoshida D, Kim K, Yamazaki M, Teramoto A, 2005, Expression of hypoxia-inducible factor 1α and cathepsin D in pituitary adenomas. Endocr Pathol 16(2):123–131., DOI 10.1385/ep:16:2:123 Thapar K, Scheithauer BW, Kovacs K, Pernicone PJ, Laws ERJ, 1996, p53 expression in pituitary adenomas and carcinomas: correlation with invasiveness and tumor growth fractions. Neurosurgery 38(4):765–771 Lee HK, Hsu AK, Sajdak J, Qin J, Pavlidis P, 2004, Coexpression analysis of human genes across many microarray data sets. Genome Res 14(6):1085–1094., DOI 10.1101/gr.1910904 McInturff JE, Modlin RL, KimJ, 2005, The role of toll-like receptors in the pathogenesis and treatment of dermatological disease. J Investig Dermatol 125(1):1–8 KA M, AJ K, M X, K-V K, MG E, M G, BK K-D KOL, ME W, 2011, Identification of growth arrest and DNA-damage-inducible gene beta, GADD45beta). Endocrinology 152(10):3603–3613., DOI 10.1210/en2011-0109 Liu BH, Yu H, Tu K, Li C, Li YX, Li YY, 2010, DCGL: an R package for identifying differentially coexpressed genes and links from gene expression microarray data. Bioinformatics 26(20):2637– 2638., DOI 10.1093/bioinformatics/btq471 Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I, 2001, Controlling the false discovery rate in behavior genetics research. Behav Brain Res 125(1–2):279–284 Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA, 2003, DAVID: database for annotation, visualization, and integrated discovery. Genome Biol 4(5):P3 de la Fuente A, 2010, From ‘differential expression’ to ‘differential networking’—identification of dysfunctional regulatory networks in diseases. Trends Genet 26(7):326–333., DOI 10.1016/j.tig.2010.05.001 Altenberger T, Bilban M, Auer M, Knosp E, Wolfsberger S, Gartner W, Mineva I, Zielinski C,Wagner L, Luger A, 2006, Identification of DLK1 variants in pituitary- and neuroendocrine tumors. Biochem Biophys Res Commun 340(3):995–1005., DOI 10.1016/j.bbrc.2005. 12.094 Cheunsuchon P, Zhou Y, Zhang X, Lee H, Chen W, Nakayama Y, Rice KA, Tessa Hedley-Whyte E, Swearingen B, Klibanski A, 2011, Silencing of the imprinted DLK1-MEG3 locus in human clinically nonfunctioning pituitary adenomas. Am J Pathol 179(4):2120–2130., DOI 10.1016/j.ajpath.2011.07.002 Seemann N, Kuhn D, Wrocklage C, Keyvani K, Hackl W, Buchfelder M, Fahlbusch R, Paulus W, 2001, CDKN2A/p16 inactivation is related to pituitary adenoma type and size. J Pathol 193(4):491–497., DOI 10.1002/path.833 Frost SJ, Simpson DJ, Clayton RN, Farrell WE, 1999, Transfection of an inducible p16/CDKN2A construct mediates reversible growth inhibition and G1 arrest in the AtT20 pituitary tumor cell line. Mol Endocrinol 13(11):1801–1810 Tani Y, Inoshita N, Sugiyama T, Kato M, Yamada S, Shichiri M, Hirata Y, 2010, Upregulation of CDKN2A and suppression of cyclin D1 gene expressions in ACTH-secreting pituitary adenomas. Eur J Endocrinol 163(4):523–529., DOI 10.1530/EJE- 10-0245EJE-10-0245 Chang E, Park DI, Kim YJ, Kim BK, Park JH, Kim HJ, Cho YK, Sohn CI, Jeon WK, Kim BI, Kim HD, Kim DH, Kim YH, 2010, Detection of colorectal neoplasm using promoter methylation of ITGA4, SFRP2, and p16 in stool samples: a preliminary report in Korean patients. Hepato-Gastroenterology 57(101):720–727 Saramaki OR, Porkka KP, Vessella RL, Visakorpi T, 2006, Genetic aberrations in prostate cancer by microarray analysis. Int J Cancer 119(6):1322–1329., DOI 10.1002/ijc.21976 Katoh M, Katoh M, 2004, Identification and characterization of human MPP7 gene and mouse Mpp7 gene in silico. Int J Mol Med 13(2):333–338 Kon S, Tanabe K, Watanabe T, Sabe H, Satake M, 2008, Clathrin dependent endocytosis of E-cadherin is regulated by the Arf6GAP isoform SMAP1. Exp Cell Res 314(7):1415–1428., DOI 10.1016/j. yexcr.2007.11.006 Thalappilly S, Suliman M, Gayet O, Soubeyran P, Hermant A, Lecine P, Iovanna JL, Dusetti NJ, 2008, Identification of multi-SH3 domaincontaining protein interactome in pancreatic cancer: a yeast two-hybrid approach. Proteomics 8(15):3071–3081., DOI 10.1002/pmic.200701157 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 375 EP 380 DI 10.1007/s12253-013-9706-1 PG 6 ER PT J AU Zhang, X Nie, Y Li, X Wu, G Huang, Q Cao, J Du, Y Li, J Deng, R Huang, D Chen, B Li, Sh Wei, B AF Zhang, Xiangyang Nie, Yuqiang Li, Xiaorong Wu, Guifu Huang, Qun Cao, Jie Du, Yanlei Li, Junda Deng, Ruoyu Huang, Dongshen Chen, Baozhi Li, Shang Wei, Baojun TI MicroRNA-181a Functions as an Oncomir in Gastric Cancer by Targeting the Tumour Suppressor Gene ATM SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE miR-181a; micro-RNA; Gastric cancer; Target gene; ATM ID miR-181a; micro-RNA; Gastric cancer; Target gene; ATM AB Based on our previous experiments, this study is to further investigate the functional significance of miR-181a and its target gene in gastric cancer. Expression of miR-181a was detected by qRT-PCR in three normal gastric tissues and three human gastric cancer cell lines (SGC-7901, MGC-803, and BGC-823 cells). After transfection with miR-181a inhibitor, proliferation, apoptosis, migration, and invasion of the SGC-7901 cells were evaluated. Ataxia-telangiectasia mutation (ATM) was predicted as a target gene of miR-181a with bioinformatics analysis, and was verified by lucifersae reporter assay. Expression of ATM protein in HEK293T cells and tissues was measured by Western Blot. Expression of ATM mRNA in HEK293T cells was measured by RT-PCR. Compared with three non-tumour tissues, the expression of miR-181a in three gastric cancer cells was significantly increased by 26.68, 14.83 and 14.96 folds; Compared with Negative Control(NC) and blank groups, transfection of miR- 181a inhibitor led to inhibition of SGC7901 cell proliferation, invasion, and migration as well as promotion of apoptosis. A luciferase reporter assay demonstrated that ATM was a direct target of miR-181a, miR-181a mimics transfection down regulated ATM mRNA and protein expression. There was inverse correlation between miR-181a and ATM protein expression in gastric cancer and normal gastric tissues. Our study demonstrates that over-expression of miR-181a might be involved in development of gastric cancer by promoting proliferation and inhibiting apoptosis probably through directly targeting ATM. miR-181a modulation may be a potential strategy for the development of miRNA-based therapy of gastric cancer. C1 [Zhang, Xiangyang] Shenzhen Futian Hospital of TCM, Department of Gastroenterology, No. 6001 North Central Avenue of Futian, 518034 Shenzhen, China. [Nie, Yuqiang] Guangzhou Medical University, Guangzhou First Municipal People’s Hospital, Department of Gastroenterology, Guangzhou Key Laboratory of Digestive Disease, No.1 Panfu Road, 510180 Guangzhou, China. [Li, Xiaorong] Shenzhen Futian Hospital of TCM, Department of Gastroenterology, No. 3025 Middle Shennan Road, 518033 Shenzhen, China. [Wu, Guifu] Shenzhen Futian Hospital of TCM, Department of Gastroenterology, No. 3025 Middle Shennan Road, 518033 Shenzhen, China. [Huang, Qun] Shenzhen Futian Hospital of TCM, Department of Gastroenterology, No. 3025 Middle Shennan Road, 518033 Shenzhen, China. [Cao, Jie] Guangzhou Medical University, Guangzhou First Municipal People’s Hospital, Department of Gastroenterology, Guangzhou Key Laboratory of Digestive Disease, No.1 Panfu Road, 510180 Guangzhou, China. [Du, Yanlei] Guangzhou Medical University, Guangzhou First Municipal People’s Hospital, Department of Gastroenterology, Guangzhou Key Laboratory of Digestive Disease, No.1 Panfu Road, 510180 Guangzhou, China. [Li, Junda] People’s Hospital Of New District Longhua Shenzhen, Department of Gastroenterology, Jianshe East Road of Longhua New City, 518019 Shenzhen, China. [Deng, Ruoyu] People’s Hospital Of New District Longhua Shenzhen, Department of Gastroenterology, Jianshe East Road of Longhua New City, 518019 Shenzhen, China. [Huang, Dongshen] Guangzhou Chest Hospital, 510180 Guangzhou, China. [Chen, Baozhi] Lanzhou University Second Hospital, 730046 Lanzhou, China. [Li, Shang] Jiuquan City Health School, 735000 Lanzhou, China. [Wei, Baojun] People’s Hospital of Gansu Province, 730060 Lanzhou, China. RP Nie, Y (reprint author), Guangzhou Medical University, Guangzhou First Municipal People’s Hospital, Department of Gastroenterology, Guangzhou Key Laboratory of Digestive Disease, 510180 Guangzhou, China. EM yuqiangnie@126.com CR Kamangar F, Dores GM, Anderson WF, 2006, Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol 24(14):2137–2150 Tamura G, 2006, Alterations of tumour suppressor and tumourrelated genes in the development and progression of gastric cancer. World J Gastroenterol 12(2):192–198 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116(2):281–297 Esquela-Kerscher A, Slack FJ, 2006, Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer 6(4):259–269 Calin GA, Dumitru CD, Shimizu M et al, 2002, Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A 99(24):15524–15529 Bonci D, Coppola V, Musumeci M et al, 2008, The miR-15a-miR- 16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat Med 14(11):1271–1277 Petrocca F, Visone R, Onelli MR et al, 2008, E2F1-regulated microRNAs impair TGFbeta-dependent cell-cycle arrest and apoptosisin gastric cancer. Cancer Cell 13(3):272–286 Kim YK, Yu J, Han TS et al, 2009, Functional links between clustered microRNAs: suppression of cell-cycleinhibitors by microRNAclusters in gastric cancer. Nucleic Acids Res 37(5):1672–1681 Ueda T, Volinia S, Okumura H et al, 2010, Relation between microRNAexpression and progression and prognosis of gastriccancer: a microRNA expression analysis. Lancet Oncol 11(2):136–146 Chen CZ, Li L, Lodish HF, Bartel DP, 2004, MicroRNAs modulate hematopoietic lineage differentiation. Science, 80-, 303(5654):83–86 Li QJ, Chau J, Ebert PJ et al, 2007, miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell 129(1):147–161 Neilson JR, Zheng GX, Burge CB, Sharp PA, 2007, Dynamic regulation of miRNA expression in ordered stages of cellular development. Genes Dev 21(5):578–589 Kazenwadel J, Michael MZ, Harvey NL, 2010, Prox1 expression is negatively regulated by miR-181 in endothelial cells. Blood 116(13): 2395–2401 Naguibneva I, Ameyar-Zazoua M, Polesskaya A et al, 2006, The microRNA miR-181 targets the homeobox protein Hox-A11 during mammalianmyoblast differentiation. Nat Cell Biol 8(3): 278–284 Marcucci G, Radmacher MD, Maharry K et al, 2008, MicroRNA expression in cytogenetically normal acute myeloid leukemia.NEngl J Med 358(18):1919–1928 Pallasch CP, Patz M, Park YJ et al, 2009, miRNA deregulation by epigenetic silencing disrupts suppression of the oncogenePLAG1 in chronic lymphocytic leukemia. Blood 114(15):3255–3264 Pichiorri F, Suh SS, Ladetto M et al, 2008, MicroRNAs regulate critical genes associated with multiple myeloma pathogenesis. Proc Natl Acad Sci U S A 105(35):12885–12890 Wang Y, Yu Y, Tsuyada A et al, 2011, Transforming growth factorbeta regulates the sphere-initiating stem cell-likefeature in breast cancer throughmiRNA-181 and ATM. Oncogene 30(12):1470–1480 Ji J, Yamashita T, Budhu A et al, 2009, Identification of microRNA- 181 by genome-wide screening as a critical player inEpCAMpositive hepatic cancer stem cells. Hepatology 50(2):472–480 Bhattacharya SD, Garrison J, Guo H et al, 2010, Micro-RNA-181a regulates osteopontin-dependent metastatic function inhepatocellular cancer cell lines. Surgery 148(2):291–297 Yao Y, Suo AL, Li ZF et al, 2009, MicroRNA profiling of human gastric cancer. Mol Med Report 2(6):963–970 Herzog KH, Chong MJ, Kapsetaki M, Morgan JI, McKinnon PJ, 1998, Requirement for ATMin ionizing radiation-induced cell death in the developingcentral nervous system. Science, 80-, 280(5366): 1089–1091 Derheimer FA, Kastan MB, 2010, Multiple roles of ATM in monitoring and maintaining DNA integrity. FEBS Lett 584(17): 3675–3681 Thompson D, Duedal S, Kirner J et al, 2005, Cancer risks and mortality in heterozygous ATM mutation carriers. J Natl Cancer Inst 97(11):813–822 Mandriota SJ, Buser R, Lesne L et al, 2010, Ataxia telangiectasia mutated, ATM, inhibition transforms human mammary glandepithelial cells. J Biol Chem 285(17):13092–13106 Kang B, Guo RF, Tan XH, Zhao M, Tang ZB, Lu YY, 2008, Expression status of ataxia-telangiectasia-mutated gene correlated with prognosisin advanced gastric cancer.Mutat Res 638(1–2):17–25 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), Method. Methods 25(4):402–408 Lewis BP, Shih IH, Jones-RhoadesMW, Bartel DP, Burge CB, 2003, Prediction of mammalian microRNA targets. Cell 115(7):787–798 Krek A, Grun D, Poy MN et al, 2005, Combinatorial microRNA target predictions. Nat Genet 37(5):495–500 Betel D, Wilson M, Gabow A, Marks DS, Sander C, 2008, The microRNA.org resource: targets and expression. Nucleic Acids Res 36(Database issue):D149–D153 Smith PK, Krohn RI, Hermanson GT et al, 1985, Measurement of protein using bicinchoninic acid. Anal Biochem 150(1):76–85 Du Y, Xu Y, Ding L et al, 2009, Down-regulation of miR-141 in gastric cancer and its involvement in cell growth. J Gastroenterol 44(6):556–561 O’Donnell KA,Wentzel EA, Zeller KI, Dang CV,Mendell JT, 2005, c-Myc-regulated microRNAs modulate E2F1 expression. Nature 435(7043):839–843 Tchernitsa O, Kasajima A, Schafer R et al, 2010, Systematic evaluation of the miRNA-ome and its downstream effects on mRNA expression identifies gastric cancer progression. J Pathol 222(3):310–319 Ciafre SA, Galardi S,Mangiola A et al, 2005, Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun 334(4):1351–1358 GaoW, Yu Y, Cao H et al, 2010, Deregulated expression of miR-21, miR-143 and miR-181a in non small cell lungcancer is related to clinicopathologic characteristics or patient prognosis. Biomed Pharmacother 64(6):399–408 Shin KH, Bae SD, Hong HS, Kim RH, Kang MK, Park NH, 2011, miR-181a shows tumour suppressive effect against oral squamous cell carcinomacells by downregulating K-ras. Biochem Biophys Res Commun 404(4):896–902 Jazdzewski K, Boguslawska J, Jendrzejewski J et al, 2011, Thyroid hormone receptor beta, THRB, is a major target gene for microRNAsderegulated in papillary thyroid carcinoma, PTC). J Clin Endocrinol Metab 96(3):E546–E553 Noh H, Hong S, Dong Z, Pan ZK, Jing Q, Huang S, 2011, Impaired microRNA processing facilitates breast cancer cell invasion by up regulating urokinase-type plasminogen activator expression. Genes Cancer 2(2):140–150 Toller IM, Neelsen KJ, Steger M et al, 2011, Carcinogenic bacterial pathogen Helicobacter pylori triggers DNA double-strandbreaks and a DNA damage response in its host cells. Proc Natl Acad Sci U S A 108(36):14944–14949 Zhu KQ, Zhang SJ, 2003, Involvement of ATM/ATR-p38 MAPK cascade in MNNG induced G1-S arrest. World J Gastroenterol 9(9): 2073–2077 Lavin MF, Kozlov S, 2007, ATM activation and DNA damage response. Cell Cycle 6(8):931–942 Bartkova J, Bakkenist CJ, Rajpert-De ME et al, 2005, ATM activation in normal human tissues and testicular cancer. Cell Cycle 4(6):838–845 Song SY, Kang MR, Yoo NJ, Lee SH, 2010, Mutational analysis of mononucleotide repeats in dual specificity tyrosinephosphatase genes in gastric and colon carcinomas with microsatellite instability. APMIS 118(5):389–393 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 381 EP 389 DI 10.1007/s12253-013-9707-0 PG 9 ER PT J AU Voros, A Csorgo, E Kovari, B Lazar, P Kelemen, Gy Cserni, G AF Voros, Andras Csorgo, Erika Kovari, Bence Lazar, Peter Kelemen, Gyongyi Cserni, Gabor TI The Use of Digital Images Improves Reproducibility of the Ki-67 Labeling Index as a Proliferation Marker in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Core-biopsy; Ki-67; Proliferation marker; Reproducibility; Digital assessment ID Breast cancer; Core-biopsy; Ki-67; Proliferation marker; Reproducibility; Digital assessment AB The proportion of Ki-67 immunostained cells (Ki- 67 labeling index, LI) is one of the most commonly used histology methods for estimating proliferation of breast carcinomas. Although the Ki-67 LI is used in treatment decision making, its reproducibility shows variation in different studies, and is generally less then optimal. The aim of the present study was to investigate how the use of a standardized, partially digitalized counting method could affect reproducibility of determining the Ki-67 LI. Thirty breast cancer core-biopsy samples were stained with B-56, SP-6 and MIB-1 monoclonal Ki-67 antibodies. Each sample was represented by a single digital photograph taken with a x20 objective. Four investigators determined the Ki-67 LI on these digital images by estimation, then by counting with the help of a grid overlaid on the same images. Altogether 720 evaluations were made by 4 independent pathologists. Good to excellent correlation was found between estimations and calculations of each observer. Kappa values >0.6 suggest substantial inter-observer agreement when classifying the cases into a 15 % and 30 % cut-off determined three-tiered or a 4-quarter-based four-tiered categorization, which is better than the fair reproducibility gained on the real slides in a previous study. The results also suggest that the type of the antibody may also impact on the consistency of both estimating and calculating the Ki-67 LIs. The results indicate that counting on digital images may significantly improve reproducibility of determining the KI-67 LI. Interestingly, estimation on the same images is not worse, but is obviously faster and more convenient. C1 [Voros, Andras] University of Szeged, Department of Pathology, Allomas u 2, 6725 Szeged, Hungary. [Csorgo, Erika] University of Szeged, Department of Pathology, Allomas u 2, 6725 Szeged, Hungary. [Kovari, Bence] University of Szeged, Department of Pathology, Allomas u 2, 6725 Szeged, Hungary. [Lazar, Peter] University of Szeged, Department of Pathology, Allomas u 2, 6725 Szeged, Hungary. [Kelemen, Gyongyi] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Cserni, Gabor] University of Szeged, Department of Pathology, Allomas u 2, 6725 Szeged, Hungary. RP Voros, A (reprint author), University of Szeged, Department of Pathology, 6725 Szeged, Hungary. EM andrasvoros@libero.it CR Urruticoechea A, Smith IE, Dowsett M, 2005, Proliferation marker Ki-67 in early breast cancer. J Clin Oncol 23:7212–20 ColozzaM, Azambuja E, Cardoso F, Sotiriou C, LarsimontD, Piccart MJ, 2005, Proliferative markers as prognostic and predictive tools in early breast cancer: where are we now? Ann Oncol 16:1723–39 Jalava P, Kuopio T, Juntti-Patinen L, Kotkansalo T, Kronqvist P, Collan Y, 2006, Ki67 immunohistochemistry: a valuable marker in prognostication but with a risk of misclassification: proliferation subgroups formed based on Ki67 immunoreactivity and standardized mitotic index. Histopathology 48:674–82 de Azambuja E, Cardoso F, de Castro G, Jr CM, Mano DV et al, 2007, Ki-67 as prognostic marker in early breast cancer: a metaanalysis of published studies involving 12,155 patients. Br J Cancer 96:1504–13 DowsettM, Nielsen TO,A’Hern R, Bartlett J, Coombes RC, Cuzick J et al, 2011, Assessment of Ki67 in breast cancer: recommendations from the International Ki67 in Breast Cancer Working Group. J Natl Cancer Inst 103:1–9 Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA, 2010, Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol 11:174–83 Billgren AM, Tani E, Liedberg A, Skoog L, Rutqvist LE, 2002, Prognostic significance of tumor cell proliferation analyzed in fine needle aspirates from primary breast cancer. Breast Cancer Res Treat 71:161–70 Liu S, Edgerton SM, Moore DH 2nd, Thor AD, 2001, Measures of cell turnover, proliferation and apoptosis, and their association with survival in breast cancer. Clin Cancer Res 7:1716–23 Seshadri R, Leong AS, McCaul K, Firgaira FA, Setlur V, Horsfall DJ, 1996, Relationship between p53 gene abnormalities and other tumour characteristics in breast-cancer prognosis. Int J Cancer 69:135–41 Luporsi E, Andre F, Spyratos F, Martin PM, Jacquemier J, Penault- Llorca F, Tubiana-Mathieu N, Sigal-Zafrani B, Arnould L, Gompel A, Egele C, Poulet B, Clough KB, Crouet H, Fourquet A, Lefranc JP, Mathelin C, Rouyer N, Serin D, Spielmann M, Haugh M, Chenard MP, Brain E, de Cremoux P, Bellocq JP, 2012, Ki-67: level of evidence andmethodological considerations for its role in the clinical management of breast cancer: analytical and critical review. Breast Cancer Res Treat 132:895–915 Viale G, Giobbie-Hurder A, Regan MM, Coates AS, Mastropasqua MG, Dell’Orto P et al, 2008, Prognostic and predictive value of centrally reviewed Ki-67 labeling index in postmenopausal women with endocrine-responsive breast cancer: results from Breast International Group Trial 1–98 comparing adjuvant tamoxifen with letrozole. J Clin Oncol 26:5569–75 Viale G, Regan MM, Mastropasqua MG, Maffini F, Maiorano E, Colleoni M, Price KN, Golouh R, Perin T, Brown RW, Kovacs A, Pillay K, Ohlschlegel C, Gusterson BA, Castiglione-Gertsch M, Gelber RD, Goldhirsch A, Coates AS, International Breast Cancer Study Group, 2008, Predictive value of tumor Ki-67 expression in two randomized trials of adjuvant chemoendocrine therapy for nodenegative breast cancer. J Natl Cancer Inst 100:207–12 Goldhirsch A, Ingle JN, Gelber RD, Coates AS, Thurlimann B, Senn HJ, 2009, Thresholds for therapies: highlights of the St Gallen International Expert Consensus on the primary therapy of early breast cancer 2009. Ann Oncol 20:1319–29 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, 2011, Strategies for subtypes–dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol 22:1736–47 Beresford MJ,Wilson GD,Makris A, 2006)Measuring proliferation in breast cancer: practicalities and applications. Breast Cancer Res 8: 216 Gerdes J, Li L, Schlueter C, Duchrow M, Wohlenberg C, Gerlach C et al, 1991, Immunobiochemical and molecular biologic characterization of the cell proliferation-associated nuclear antigen that is defined by monoclonal antibody Ki-67. Am J Pathol 138:867–73 Gerdes J, Schwab U, Lemke H, Stein H, 1983, Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer 31:13–20 Goodson WH 3rd, Moore DH 2nd, Ljung BM, Chew K, Mayall B, Smith HS,Waldman FM(2000, The prognostic value of proliferation indices: a study with in vivo bromodeoxyuridine and Ki-67. Breast Cancer Res Treat 59:113–23 Fasching PA, Heusinger K, Haeberle L, Niklos M, Hein A, Bayer CM, Rauh C, Schulz-Wendtland R, Bani MR, Schrauder M, Kahmann L, Lux MP, Strehl JD, Hartmann A, Dimmler A, Beckmann MW, Wachter DL, 2011, Ki67, chemotherapy response, and prognosis in breast cancer patients receiving neoadjuvant treatment. BMC Cancer 11:486 Jung SY, HanW, Lee JW, Ko E, Kim E, Yu JH, Moon HG, Park IA, OhDY, ImSA, KimTY, Hwang KT, KimSW, Noh DY, 2009, Ki-67 expression gives additional prognostic information on St. Gallen 2007 and adjuvant! Online risk categories in early breast cancer. Ann Surg Oncol 16:1112–21 Mohammed ZM, McMillan DC, Elsberger B, Going JJ, Orange C, Mallon E, Doughty JC, Edwards J, 2012, Comparison of visual and automated assessment of Ki-67 proliferative activity and their impact on outcome in primary operable invasive ductal breast cancer. Br J Cancer 106:383–8 Kashiwagi S, YashiroM, Takashima T, Aomatsu N, Ikeda K, Ogawa Y, Ishikawa T, Hirakawa K, 2011, Advantages of adjuvant chemotherapy for patients with triple-negative breast cancer at Stage II: usefulness of prognosticmarkers E-cadherin andKi67. Breast Cancer Res 13:R122 Murri AM, Hilmy M, Bell J, Wilson C, McNicol AM, Lannigan A, Doughty JC, McMillan DC, 2008, The relationship between the systemic inflammatory response, tumour proliferative activity, Tlymphocytic and macrophage infiltration, microvessel density and survival in patients with primary operable breast cancer. Br J Cancer 99:1013–9 Fasanella S, Leonardi E, Cantaloni C, Eccher C, Bazzanella I, Aldovini D, Bragantini E, Morelli L, Cuorvo LV, Ferro A, Gasperetti F, Berlanda G, Dalla Palma P, Barbareschi M, 2011, Proliferative activity in human breast cancer: Ki-67 automated evaluation and the influence of different Ki-67 equivalent antibodies. Diagn Pathol., DOI 10.1186/1746-1596-6-S1-S7 Gnant M, Harbeck N, Thomssen C, 2011, St. Gallen 2011: summary of the consensus discussion. Breast Care, Basel, 6: 136–141 Spyratos F, Ferrero-Pous M, Trassard M, Hacene K, Phillips E, Tubiana-Hulin M, Le Doussal V, 2002, Correlation between MIB- 1 and other proliferation markers: clinical implications of the MIB-1 cutoff value. Cancer 94:2151–9 Jones RL, Salter J, A’Hern R, Nerurkar A, Parton M, Reis-Filho JS, Smith IE, Dowsett M, 2010, Relationship between oestrogen receptor status and proliferation in predicting response and long-term outcome to neoadjuvant chemotherapy for breast cancer. Breast Cancer Res Treat 119:315–23 Dowsett M, A’Hern R, Salter J, Zabaglo L, Smith IE, 2009, Who would have thought a single Ki67 measurement would predict longterm outcome? Breast Cancer Res., DOI 10.1186/bcr2434 Voros A, Csorgo E, Nyari T, Cserni G, 2012, An intra- and interobserver reproducibility analysis of the Ki-67 proliferation marker assessment on core biopsies of breast cancer patients and its potential clinical implications. Pathobiology 80:111–118 Fleiss JL, 1980, The measurement of interrater agreement. In: Fleiss JL, ed, Statistical methods for rates and proportions, 2nd edn. John Wiley and Sons Inc, New York, pp 212–236 Landis JR,KochGG(1977, Themeasurement of observer agreement for categorical data. Biometrics 33:159–174 Zs V, Diebold J, Dommann-Scherrer C, Frick H, Kaup D, Noske A, Obermann E, Ohlschlegel C, Padberg B, Rakozy C, Sancho Oliver S, Schobinger-Clement S, Schreiber-Facklam H, Singer G, Tapia C, Wagner U, Mastropasqua M, Viale G, Lehr H-A, 2012, How reliable is Ki-67 immunohistochemistry in grade 2 breast carcinomas? A QA study of the SwissWorking Group of Breast- and Gynecopathologists. Plos One 7:e37379 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 391 EP 397 DI 10.1007/s12253-013-9708-z PG 7 ER PT J AU Li, N Abe, Sh Kurata, M Abe-Suzuki, Sh Onishi, I Kirimura, S Murayama, T Hidaka, M Kawano, F Kitagawa, M AF Li, Na Abe, Shinya Kurata, Morito Abe-Suzuki, Shiho Onishi, Iichiroh Kirimura, Susumu Murayama, Toshihiko Hidaka, Michihiro Kawano, Fumio Kitagawa, Masanobu TI Over-Expression of Cancerous Inhibitor of PP2A (CIP2A) in Bone Marrow Cells from Patients with a Group of High-Risk Myelodysplastic Syndromes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CIP2A; c-MYC; Myelodysplastic syndromes; Bone marrow; Immunohistochemistry ID CIP2A; c-MYC; Myelodysplastic syndromes; Bone marrow; Immunohistochemistry AB Cancerous inhibitor of PP2A (protein phosphatase 2A) (CIP2A) is an inhibitor of PP2A, a phosphatase and tumor suppressor that regulates cell proliferation, differentiation, and survival. The aim of this study was to investigate whether CIP2A plays a role in the progression of myelodysplastic syndromes (MDS). Immunohistochemical analysis revealed that a fraction patients having refractory anemia with excess blasts (RAEB)-1 (4 out of 12) and RAEB-2 (10 out of 14) exhibited significant expression of CIP2A in bone marrow hematopoietic cells, while all patients with refractory cytopenia with unilineage or multilineage dysplasia (RCUD/ RCMD) (0 out of 18) and the control group (0 out of 17) were negative. CIP2A was mainly expressed by the MPO-positive myeloid series of cells and partly by the CD34-positive cells in association with the expression of phosphorylated c-MYC (pc- MYC) protein and the cell cycle-related proteins Ki-67, MCM2, and geminin. The percentage of p-c-MYC-positive cells in the bone marrow of CIP2A-positive MDS cases was significantly higher than that in CIP2A-negative MDS cases (P <0.01). The expression levels of mRNA for CIP2A and PP2A exhibited positive correlation in MDS/control bone marrow. These results suggest that up-regulated expression of CIP2A might play a role in the proliferation of blasts in the MDS bone marrow and in disease progression in at least some cases. C1 [Li, Na] Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Department of Comprehensive Pathology, 1-5-45 Yushima, Bunkyo-ku, 113-8519 Tokyo, Japan. [Abe, Shinya] Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Department of Comprehensive Pathology, 1-5-45 Yushima, Bunkyo-ku, 113-8519 Tokyo, Japan. [Kurata, Morito] Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Department of Comprehensive Pathology, 1-5-45 Yushima, Bunkyo-ku, 113-8519 Tokyo, Japan. [Abe-Suzuki, Shiho] Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Department of Comprehensive Pathology, 1-5-45 Yushima, Bunkyo-ku, 113-8519 Tokyo, Japan. [Onishi, Iichiroh] Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Department of Comprehensive Pathology, 1-5-45 Yushima, Bunkyo-ku, 113-8519 Tokyo, Japan. [Kirimura, Susumu] Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Department of Comprehensive Pathology, 1-5-45 Yushima, Bunkyo-ku, 113-8519 Tokyo, Japan. [Murayama, Toshihiko] National Hospital Organization, Kumamoto Medical Center, Department of PathologyKumamoto, Japan. [Hidaka, Michihiro] National Hospital Organization, Kumamoto Medical Center, Department of Internal MedicineKumamoto, Japan. [Kawano, Fumio] National Hospital Organization, Kumamoto Medical Center, Department of Internal MedicineKumamoto, Japan. [Kitagawa, Masanobu] Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Department of Comprehensive Pathology, 1-5-45 Yushima, Bunkyo-ku, 113-8519 Tokyo, Japan. RP Abe, Sh (reprint author), Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Department of Comprehensive Pathology, 113-8519 Tokyo, Japan. EM abenchi.pth2@tmd.ac.jp CR Bennett JM(2005, A comparative review of classification systems in myelodysplastic syndromes, MDS). Semin Oncol 32:S3–S10 Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Vardiman JW, 2008, WHO classification of tumours of haematopoietic and lymphoid tissues. IARC PRESS, Lyon Benesch M, Platzbecker U, Ward J, Deeg HJ, Leisenring W, 2003, Expression of FLIP(Long, and FLIP(Short, in bone marrow mononuclear and CD34+ cells in patients with myelodysplastic syndrome: correlation with apoptosis. Leukemia 17:2460–2466 Campioni D, Secchiero P, Corallini F, Melloni E, Capitani S, Lanza F, Zauli G, 2005, Evidence for a role of TNF-related apoptosisinducing ligand, TRAIL, in the anemia of myelodysplastic syndromes. Am J Pathol 166:557–563 Claessens YE, Bouscary D, Dupont JM, Picard F, Melle J, Gisselbrecht S, Lacombe C, Dreyfus F, Mayeux P, Fontenay- Roupie M, 2002, In vitro proliferation and differentiation of erythroid progenitors from patients withmyelodysplastic syndromes: evidence for Fas-dependent apoptosis. Blood 99:1594–1601 Kitagawa M, Yamaguchi S, Takahashi M, Tanizawa T, Hirokawa K, Kamiyama R, 1998, Localization of Fas and Fas ligand in bone marrow cells demonstrating myelodysplasia. Leukemia 12:486–492 Zamai L, Secchiero P, Pierpaoli S, Bassini A, Papa S, Alnemri ES, Guidotti L, VitaleM, Zauli G, 2000, TNF-related apoptosis-inducing ligand, TRAIL, as a negative regulator of normal human erythropoiesis. Blood 95:3716–3724 Zang DY, Goodwin RG, Loken MR, Bryant E, Deeg HJ, 2001, Expression of tumor necrosis factor-related apoptosis-inducing ligand, Apo2L, and its receptors in myelodysplastic syndrome: effects on in vitro hemopoiesis. Blood 98:3058–3065 Kitagawa M, Kamiyama R, Kasuga T, 1993, Expression of the proliferating cell nuclear antigen in bone marrow cells from patients with myelodysplastic syndromes and aplastic anemia. Hum Pathol 24:359–363 Kitagawa M, Saito I, Kuwata T, Yoshida S, Yamaguchi S, Takahashi M, Tanizawa T, Kamiyama R, Hirokawa K, 1997, Overexpression of tumor necrosis factor, TNF)-alpha and interferon, IFN)-gamma by bone marrow cells from patients with myelodysplastic syndromes. Leukemia 11:2049–2054 Raza A, Gezer S, Mundle S, Gao XZ, Alvi S, Borok R, Rifkin S, Iftikhar A, ShettyV, ParcharidouA(1995, Apoptosis in bonemarrow biopsy samples involving stromal and hematopoietic cells in 50 patients with myelodysplastic syndromes. Blood 86:268–276 Sawanobori M, Yamaguchi S, Hasegawa M, Inoue M, Suzuki K, Kamiyama R, Hirokawa K, Kitagawa M, 2003, Expression of TNF receptors and related signaling molecules in the bone marrow from patients with myelodysplastic syndromes. Leuk Res 27:583–591 Katsoulidis E, Li Y, Yoon P, Sassano A, Altman J, Kannan- Thulasiraman P, Balasubramanian L, Parmar S, Varga J, Tallman MS, Verma A, Platanias LC, 2005, Role of the p38 mitogenactivated protein kinase pathway in cytokine-mediated hematopoietic suppression in myelodysplastic syndromes. Cancer Res 65:9029–9037 Junttila MR, Puustinen P, Niemela M, Ahola R, Arnold H, Bottzauw T, Ala-aho R, Nielsen C, Ivaska J, Taya Y, Lu SL, Lin S, Chan EK, Wang XJ, Grenman R, Kast J, Kallunki T, Sears R, Kahari VM, Westermarck J, 2007, CIP2A inhibits PP2A in human malignancies. Cell 130:51–62 Junttila MR, Westermarck J, 2008, Mechanisms of MYC stabilization in human malignancies. Cell Cycle 7:592–596 MumbyM(2007, PP2A: unveiling a reluctant tumor suppressor. Cell 130:21–24 Khanna A, Bockelman C, Hemmes A, Junttila MR, Wiksten JP, Lundin M, Junnila S, Murphy DJ, Evan GI, Haglund C, Westermarck J, Ristimaki A, 2009, MYC-dependent regulation and prognostic role of CIP2A in gastric cancer. J Natl Cancer Inst 101: 793–805 Bockelman C, Lassus H, Hemmes A, Leminen A, Westermarck J, Haglund C, Butzow R, Ristimaki A, 2011, Prognostic role of CIP2A expression in serous ovarian cancer. Br J Cancer 105:989–995 Come C, Laine A, Chanrion M, Edgren H, Mattila E, Liu X, Jonkers J, Ivaska J, Isola J, Darbon JM, Kallioniemi O, Thezenas S, Westermarck J, 2009, CIP2A is associated with human breast cancer aggressivity. Clin Cancer Res 15:5092–5100 Lucas CM, Harris RJ, Giannoudis A, Copland M, Slupsky JR, Clark RE, 2011, Cancerous inhibitor of PP2A, CIP2A, at diagnosis of chronic myeloid leukemia is a critical determinant of disease progression. Blood 117:6660–6668 Cristobal I, Garcia-Orti L, Cirauqui C, Alonso MM, Calasanz MJ, Odero MD, 2011, PP2A impaired activity is a common event in acute myeloid leukemia and its activation by forskolin has a potent anti-leukemic effect. Leukemia 25:606–614 Yamamoto K, Abe S, Nakagawa Y, Suzuki K, Hasegawa M, Inoue M, Kurata M, Hirokawa K, Kitagawa M, 2004, Expression of IAP family proteins in myelodysplastic syndromes transforming to overt leukemia. Leuk Res 28:1203–1211 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T), Method. Methods 25:402–408 Arroyo JD, Hahn WC, 2005, Involvement of PP2A in viral and cellular transformation. Oncogene 24:7746–7755 Janssens V, Goris J, Van Hoof C, 2005, PP2A: the expected tumor suppressor. Curr Opin Genet Dev 15:34–41 WangX, CunninghamM, Zhang X, Tokarz S, Laraway B, Troxell M, Sears RC, 2011, Phosphorylation regulates c-Myc’s oncogenic activity in the mammary gland. Cancer Res 71:925–936 Niemela M, Kauko O, Sihto H, Mpindi JP, Nicorici D, Pernila P, Kallioniemi OP, Joensuu H, Hautaniemi S, Westermarck J, 2012, CIP2A signature reveals the MYC dependency of CIP2A-regulated phenotypes and its clinical association with breast cancer subtypes. Oncogene 31:4266–4278 Mukhopadhyay A, Saddoughi SA, Song P, Sultan I, Ponnusamy S, Senkal CE, Snook CF, Arnold HK, Sears RC, Hannun YA, Ogretmen B, 2009, Direct interaction between the inhibitor 2 and ceramide via sphingolipid-protein binding is involved in the regulation of protein phosphatase 2A activity and signaling. FASEB J 23:751–763 Zhang X, Farrell AS, Daniel CJ, Arnold H, Scanlan C, Laraway BJ, Janghorban M, Lum L, Chen D, Troxell M, Sears R, 2012, Mechanistic insight into Myc stabilization in breast cancer involving aberrant Axin1 expression. Proc Natl Acad Sci U S A 109:2790– 2795 ChenKF, Liu CY, LinYC,Yu HC, Liu TH,Hou DR, Chen PJ, Cheng AL, 2010, CIP2A mediates effects of bortezomib on phospho-Akt and apoptosis in hepatocellular carcinoma cells. Oncogene 29:6257– 6266 Wei S, Chen X, Rocha K, Epling-Burnette PK, Djeu JY, Liu Q, Byrd J, Sokol L, Lawrence N, Pireddu R, Dewald G, Williams A, Maciejewski J, List A, 2009, A critical role for phosphatase haplodeficiency in the selective suppression of deletion 5q MDS by lenalidomide. Proc Natl Acad Sci U S A 106:12974–12979 Abe S, Kurata M, Suzuki S, Yamamoto K, Aisaki K, Kanno J, Kitagawa M, 2012, Minichromosome maintenance 2 bound with retroviral Gp70 is localized to cytoplasm and enhances DNAdamage- induced apoptosis. PLoS One 7:e40129 de Souza Fernandez T, Silva ML, De Souza J, De Paula MT, Abdelhay E, 1996, c-MYC amplification in a case of progression from MDS to AML, M2). Cancer Genet Cytogenet 86:183–184 Taira N, Mimoto R, Kurata M, Yamaguchi T, Kitagawa M, Miki Y, Yoshida K, 2012, DYRK2 priming phosphorylation of c-Jun and c- Myc modulates cell cycle progression in human cancer cells. J Clin Invest 122:859–872 Di Bonito M, Cantile M, Collina F, Scognamiglio G, Cerrone M, La Mantia E, Barbato A, Liguori G, Botti G, 2012, Overexpression of cell cycle progression inhibitor geminin is associated with tumor stem-like phenotype of triple-negative breast cancer. J Breast Cancer 15:162–171 Yu G, Liu G, Dong J, Jin Y, 2013, Clinical implications of CIP2A protein expression in breast cancer. Med Oncol 30:524 Shetty A, Loddo M, Fanshawe T, Prevost AT, Sainsbury R,Williams GH, Stoeber K, 2005, DNA replication licensing and cell cycle kinetics of normal and neoplastic breast. Br J Cancer 93:1295–1300 Shinnick KM, Eklund EA, McGarry TJ, 2010, Geminin deletion from hematopoietic cells causes anemia and thrombocytosis in mice. J Clin Invest 120:4303–4315 Ohno Y, Yasunaga S, Janmohamed S, Ohtsubo M, Saeki K, Kurogi T, Mihara K, Iscove NN, Takihara Y, 2013, Hoxa9 transduction induces hematopoietic stem and progenitor cell activity through direct down-regulation of geminin protein. PLoS One 8:e53161 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 399 EP 407 DI 10.1007/s12253-013-9709-y PG 9 ER PT J AU Kim, K Kim, DH Chae, WS Shin, JH Kim, JH Do, SI Lee, JH Koo, HJ Pyo, JS Sohn, HJ AF Kim, Kyungeun Kim, Dong-Hoon Chae, Wan Seoung Shin, Jun-Ho Kim, Joo Hong Do, Sung-Im Lee, Joo Hyun Koo, Hae Ji Pyo, Jung-Soo Sohn, Hee Jin TI Expression of Cell Cycle-Related Proteins, p16, p53 and p63 as Important Prognostic Markers in Gallbladder Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gallbladder; Adenocarcinoma; p16 protein; p53 protein; p63 protein ID Gallbladder; Adenocarcinoma; p16 protein; p53 protein; p63 protein AB Gallbladder cancer, the most common biliary tract malignancy, is a highlymalignant neoplasm. In the present work, we have analyzed the significance of cell cycle-related proteins to predict prognosis and to provide guidance for optimal therapeutic decision-making in patients with gallbladder adenocarcinoma. The expressions of p16, p21, p27, p53, p63, cyclin D1, bcl-2 and bcl-6 were examined in a tissue microarray constructed from 96 cases of gallbladder adenocarcinoma by immunohistochemistry and correlated with clinicopathologic prognostic factors. Expression of p16 was correlated with a low pT stage, adenoma background and good prognosis. Cases with p63 expression showed a higher T stage, more frequent perineural invasion and poor prognosis when compared to cases without p63 expression. Over-expression of p53 or loss of p53 was associated with poor tumor differentiation, frequent distant metastasis and low disease-specific survival rate. The expressions of p21, p27, bcl- 2, bcl-6 and cyclin D1 were not significant prognostic factors for gallbladder adenocarcinoma. These results indicate that p16, p63 and p53 can be used as prognostic markers in gallbladder adenocarcinoma; especially p53 and p63 as poor prognostic markers and p16 as a favorable prognostic marker. C1 [Kim, Kyungeun] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 108, Pyeong-dong, Jongro-gu, 100-634 Seoul, South Korea. [Kim, Dong-Hoon] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 108, Pyeong-dong, Jongro-gu, 100-634 Seoul, South Korea. [Chae, Wan Seoung] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 108, Pyeong-dong, Jongro-gu, 100-634 Seoul, South Korea. [Shin, Jun-Ho] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of SurgerySeoul, South Korea. [Kim, Joo Hong] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Internal MedicineSeoul, South Korea. [Do, Sung-Im] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 108, Pyeong-dong, Jongro-gu, 100-634 Seoul, South Korea. [Lee, Joo Hyun] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 108, Pyeong-dong, Jongro-gu, 100-634 Seoul, South Korea. [Koo, Hae Ji] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 108, Pyeong-dong, Jongro-gu, 100-634 Seoul, South Korea. [Pyo, Jung-Soo] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 108, Pyeong-dong, Jongro-gu, 100-634 Seoul, South Korea. [Sohn, Hee Jin] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 108, Pyeong-dong, Jongro-gu, 100-634 Seoul, South Korea. RP Sohn, HJ (reprint author), Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 100-634 Seoul, South Korea. EM jhpath.sohn@samsung.com CR Miller G, Jarnagin WR, 2008, Gallbladder carcinoma. Eur J Surg Oncol 34(3):306–312 Jayaraman S, Jarnagin WR, 2010, Management of gallbladder cancer. Gastroenterol Clin North Am 39(2):331–342 Goldin RD, Roa JC, 2009, Gallbladder cancer: a morphological and molecular update. Histopathology 55(2):218–229 Pietenpol JA, Kastan M, 2004, Control of the cell cycle. In: Abeloff M, Armitage J, Niederhuber J, eds, Clinical oncology. 3rd edn. Chruchill & Livingstone, pp 81–95 Bosman FT, Carneiro F, Hruban RH, Theise ND, 2010, WHO Classification of Tumours of the Digestive System. 4th edn., Lyon Edge SB, Byrd DR, Compton CC, April GF, Greene FL, Trotti A, 2010, AJCC cancer staging manual, 7th edn. Springer, New York Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100(1):57–70 Sherr CJ, 1996, Cancer cell cycles. Science 274(5293):1672–1677 Gerdes B, Ramaswamy A, Ziegler A, Lang SA, KerstingM, Baumann R, Wild A, Moll R, Rothmund M, Bartsch DK, 2002, p16INK4a is a prognostic marker in resected ductal pancreatic cancer: an analysis of p16INK4a, p53, MDM2, an Rb. Annals of surgery 235(1):51–59 Tanaka R, Wang D, Morishita Y, Inadome Y, Minami Y, Iijima T, Fukai S,Goya T,NoguchiM(2005, Loss of function of p16 gene and prognosis of pulmonary adenocarcinoma. Cancer 103(3):608–615 Choi HJ, Yun SS, Kim HJ, Choi JH, 2010, Expression of p16 protein in gallbladder carcinoma and its precancerous conditions. Hepatogastroenterology 57(97):18–21 Lynch BC, Lathrop SL, Ye D, Ma TY, Cerilli LA, 2008, Expression of the p16(INK4a, gene product in premalignant and malignant epithelial lesions of the gallbladder. Ann Diagn Pathol 12(3):161–164 Feng Z, Chen J,Wei H, Gao P, Shi J, Zhang J, Zhao F, 2011, The risk factor of gallbladder cancer: hyperplasia of mucous epithelium caused by gallstones associates with p16/CyclinD1/CDK4 pathway. Exp Mol Pathol 91(2):569–577 Quan ZW, Wu K, Wang J, Shi W, Zhang Z, Merrell RC, 2001, Association of p53, p16, and vascular endothelial growth factor protein expressions with the prognosis and metastasis of gallbladder cancer. J Am Coll Surg 193(4):380–383 Shi YZ, Hui AM, Li X, Takayama T, Makuuchi M, 2000, Overexpression of retinoblastoma protein predicts decreased survival and correlates with loss of p16INK4 protein in gallbladder carcinomas. Clin Cancer Res 6(10):4096–4100 Tadokoro H, Shigihara T, Ikeda T, TakaseM, SuyamaM(2007, Two distinct pathways of p16 gene inactivation in gallbladder cancer. World J Gastroenterol 13(47):6396–6403 Rai R, Tewari M, Kumar M, Singh AK, Shukla HS, 2011, p53: its alteration and gallbladder cancer. Eur J Cancer Prev 20(2):77–85 Kanthan R, Radhi JM, Kanthan SC, 2000, Gallbladder carcinomas: an immunoprognostic evaluation of P53, Bcl-2, CEA and alphafetoprotein. Can J Gastroenterol 14(3):181–184 Misra S, Chaturvedi A, Goel MM, Mehrotra R, Sharma ID, Srivastava AN, Misra NC, 2000, Overexpression of p53 protein in gallbladder carcinoma in North India. Eur J Surg Oncol 26(2):164–167 Chang HJ, Yoo BC, Kim SW, Lee BL, KimWH, 2007, Significance of PML and p53 protein as molecular prognostic markers of gallbladder carcinomas. Pathol Oncol Res 13(4):326–335 Ajiki T, OnoyamaH,YamamotoM,Asaka K, Fujimori T, Maeda S, Saitoh Y, 1996, p53 protein expression and prognosis in gallbladder carcinoma and premalignant lesions. Hepato-gastroenterology 43(9):521–526 Washington K, Gottfried MR, 1996, Expression of p53 in adenocarcinoma of the gallbladder and bile ducts. Liver 16(2):99–104 Kim YW, Huh SH, Park YK, Yoon TY, Lee SM, Hong SH, 2001, Expression of the c-erb-B2 and p53 protein in gallbladder carcinomas. Oncol Rep 8(5):1127–1132 da Rocha AO, Coutinho LM, Scholl JG, Leboutte LD, 2004, The value of p53 protein expression in gallbladder carcinoma: analysis of 60 cases. Hepato-gastroenterology 51(59):1310–1314 Hidalgo Grau LA, Badia JM, Salvador CA, Monso TS, Canaleta JF, Nogues JM, Sala JS, 2004, Gallbladder carcinoma: the role of p53 protein overexpression and Ki-67 antigen expression as prognostic markers. HPB, Oxford, 6(3):174–180 TakagawaM,Muguruma N, Oguri K, Imoto Y, Okamoto K, Ii K, Ito S, 2005, Prediction of prognosis in gallbladder carcinoma by mucin and p53 immunohistochemistry. Dig Dis Sci 50(8):1410–1413 Wang SN, Chung SC, Tsai KB, Chai CY, ChangWT, Kuo KK, Chen JS, Lee KT, 2006, Aberrant p53 expression and the development of gallbladder carcinoma and adenoma. Kaohsiung JMed Sci 22(2):53–59 Hong SM, Cho H,Moskaluk CA,Yu E, Zaika AI, 2007, p63 and p73 expression in extrahepatic bile duct carcinoma and their clinical significance. J Mol Histol 38(3):167–175 Ramalho FS, Ramalho LN, Della Porta L, Zucoloto S, 2006, Comparative immunohistochemical expression of p63 in human cholangiocarcinoma and hepatocellular carcinoma. J Gastroenterol Hepatol 21(8):1276–1280 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 409 EP 415 DI 10.1007/s12253-013-9710-5 PG 7 ER PT J AU Virag, J Kenessey, I Haberler, Ch Piurko, V Balint, K Dome, B Timar, J Garami, M Hegedus, B AF Virag, Jozsef Kenessey, Istvan Haberler, Christine Piurko, Violetta Balint, Katalin Dome, Balazs Timar, Jozsef Garami, Miklos Hegedus, Balazs TI Angiogenesis and Angiogenic Tyrosine Kinase Receptor Expression in Pediatric Brain Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Astrocytoma; Ependymoma; Medulloblastoma; Tyrosine kinase receptor; Targeted therapy ID Astrocytoma; Ependymoma; Medulloblastoma; Tyrosine kinase receptor; Targeted therapy AB Tumor angiogenesis and receptor tyrosine kinases (RTK) are major novel targets in anticancer molecular therapy. Accordingly, we characterized the vascular network and the expression pattern of angiogenic RTK in the most frequent pediatric brain tumors. In a retrospective collection of 44 cases (14 astrocytoma, 16 ependymoma and 14 medulloblastoma), immunohistochemistry for VEGFR1, VEGFR2, PDGFRα, PDGFRβ, and c-Kit as well as microvessel labeling with CD34 and SMA were conducted on surgical specimens. We found a significantly higher vascular density in ependymoma. Glomeruloid formations were abundant in medulloblastoma but rare or almost absent in astrocytoma and ependymoma, respectively. C-Kit and VEGFR2 labeled blood vessels were more abundant in ependymoma than in the other two types of tumors. In contrast, medulloblastoma contained higher number of PDGFRα expressing vessels. In tumor cells, we found no VEGFR2 but VEGFR1 expression in all three tumor types. PDGFRαwas strongly expressed on the tumor cells in all three malignancies, while PDGFRβ tumor cell expression was present in the majority of medulloblastoma cases. Interestingly, small populations of c-Kit expressing cancer cells were found in a number of medulloblastoma and ependymoma cases. Our study suggests that different angiogenic mechanisms are present in ependymoma and medulloblastoma. Furthermore ependymoma patients may benefit from anti-angiogenic therapies based on the high vascularization as well as the endothelial expression of c-kit and VEGFR2. The expression pattern of the receptors on tumor cells also suggests the targeting of specific angiogenic tyrosine kinase receptors may have direct antitumor activity. Further preclinical and biomarker driven clinical investigations are needed to establish the application of tyrosine kinase inhibitors in the treatment of pediatric brain tumors. C1 [Virag, Jozsef] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Kenessey, Istvan] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Haberler, Christine] Medical University of Vienna, Department of NeuropathologyVienna, Austria. [Piurko, Violetta] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Balint, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Dome, Balazs] Medical University of Vienna, Department of Thoracic SurgeryVienna, Austria. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Garami, Miklos] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Hegedus, Balazs] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. RP Hegedus, B (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM balazs.hegedus@meduniwien.ac.at CR Goudar RK et al, 2005, Combination therapy of inhibitors of epidermal growth factor receptor/vascular endothelial growth factor receptor 2, AEE788, and the mammalian target of rapamycin, RAD001, offers improved glioblastoma tumor growth inhibition. Mol Cancer Ther 4(1):101–112 Goldman CK et al, 1993, Epidermal growth factor stimulates vascular endothelial growth factor production by human malignant glioma cells: a model of glioblastoma multiforme pathophysiology. Mol Biol Cell 4(1):121–133 Jain RK et al, 2007, Angiogenesis in brain tumours. Nat Rev Neurosci 8(8):610–622 Lopes MB, 2003, Angiogenesis in brain tumors. Microsc Res Tech 60(2):225–230 Zagzag D et al, 2000, Molecular events implicated in brain tumor angiogenesis and invasion. Pediatr Neurosurg 33(1):49–55 Zagzag D et al, 2000, Vascular apoptosis and involution in gliomas precede neovascularization: a novel concept for glioma growth and angiogenesis. Lab Invest 80(6):837–849 Zagzag D et al, 2000, Expression of hypoxia-inducible factor 1alpha in brain tumors: association with angiogenesis, invasion, and progression. Cancer 88(11):2606–2618 Haberler C et al, 2006, Immunohistochemical analysis of plateletderived growth factor receptor-alpha, -beta, c-kit, c-abl, and arg proteins in glioblastoma: possible implications for patient selection for imatinib mesylate therapy. J Neurooncol 76(2):105–109 Chan AS et al, 1998, Expression of vascular endothelial growth factor and its receptors in the anaplastic progression of astrocytoma, oligodendroglioma, and ependymoma.AmJ Surg Pathol 22(7):816–826 Gilhuis HJ et al, 2006, Three-dimensional, 3D, reconstruction and quantitative analysis of the microvasculature in medulloblastoma and ependymoma subtypes. Angiogenesis 9(4):201–208 Sie M et al, 2010, Tumour vasculature and angiogenic profile of paediatric pilocytic astrocytoma; is it much different from glioblastoma? Neuropathol Appl Neurobiol 36(7):636–647 Bartels U et al, 2006, Vascularity and angiogenesis as predictors of growth in optic pathway/hypothalamic gliomas. J Neurosurg 104(5 Suppl):314–320 Korshunov A, Golanov A, Timirgaz V, 2002, Immunohistochemical markers for prognosis of ependymal neoplasms. J Neurooncol 58(3): 255–270 Wagemakers M et al, 2010, Tumor vessel biology in pediatric intracranial ependymoma. J Neurosurg Pediatr 5(4):335–341 Puputti M et al, 2010, Expression of KIT receptor tyrosine kinase in endothelial cells of juvenile brain tumors. Brain Pathol 20(4):763– 770 Dome B et al, 2007, Alternative vascularization mechanisms in cancer: Pathology and therapeutic implications. Am J Pathol 170(1):1–15 Selby DM et al, 1997, Are endothelial cell patterns of astrocytomas indicative of grade? In Vivo 11(5):371–375 Holash J et al, 1999, Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science 284(5422): 1994–1998 Vredenburgh JJ et al, 2007, Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol 25(30):4722–4729 Green RM et al, 2009, Bevacizumab for recurrent ependymoma. Neurology 73(20):1677–1680 Aguilera DG, Goldman S, Fangusaro J, 2011, Bevacizumab and irinotecan in the treatment of children with recurrent/refractory medulloblastoma. Pediatr Blood Cancer 56(3):491–494 BeneschMet al, 2008, Compassionate use of bevacizumab, Avastin, in children and young adults with refractory or recurrent solid tumors. Ann Oncol 19(4):807–813 Couec ML et al, 2012, Bevacizumab and irinotecan in children with recurrent or refractory brain tumors: toxicity and efficacy trends. Pediatr Blood Cancer 59(1):34–38 Peyrl A et al, 2012, Antiangiogenic metronomic therapy for children with recurrent embryonal brain tumors. Pediatr Blood Cancer 59(3): 511–517 Maris JM et al, 2008, Initial testing of the VEGFR inhibitor AZD2171 by the pediatric preclinical testing program. Pediatr Blood Cancer 50(3):581–587 Meco D et al, 2010, Dual inhibitor AEE788 reduces tumor growth in preclinical models of medulloblastoma. Transl Oncol 3(5):326–335 Joensuu H et al, 2005, Amplification of genes encoding KIT, PDGFRalpha and VEGFR2 receptor tyrosine kinases is frequent in glioblastoma multiforme. J Pathol 207(2):224–231 de Groot JF, Gilbert MR, 2007, New molecular targets in malignant gliomas. Curr Opin Neurol 20(6):712–718 Slongo ML et al, 2007, Functional VEGF and VEGF receptors are expressed in human medulloblastomas. Neuro Oncol 9(4):384–392 Blom T et al, 2010, Amplification and overexpression of KIT, PDGFRA, and VEGFR2 in medulloblastomas and primitive neuroectodermal tumors. J Neurooncol 97(2):217–224 Shiba K, Matsumoto T, Hirota S, 2009, Small-molecule inhibitors against KIT and PDGFRs especially in GISTs. Gan To Kagaku Ryoho 36(7):1080–1085 Cetin N, Dienel G, Gokden M, 2005, CD117 expression in glial tumors. J Neurooncol 75(2):195–202 Gomes AL et al, 2007, Molecular alterations of KIT oncogene in gliomas. Cell Oncol 29(5):399–408 Chilton-Macneill S et al, 2004, C-kit expression and mutational analysis in medulloblastoma. Pediatr Dev Pathol 7(5):493–498 Enguita-German M et al, 2011, KIT expression and methylation in medulloblastoma and PNET cell lines and tumors. J Neurooncol 103(2):247–253 Razis E et al, 2009, Phase II study of neoadjuvant imatinib in glioblastoma: evaluation of clinical and molecular effects of the treatment. Clin Cancer Res 15(19):6258–6266 Puputti M et al, 2006, Amplification of KIT, PDGFRA, VEGFR2, and EGFR in gliomas. Mol Cancer Res 4(12):927–934 Thorarinsdottir HK et al, 2008, Protein expression of platelet-derived growth factor receptor correlates with malignant histology and PTEN with survival in childhood gliomas. Clin Cancer Res 14(11):3386–3394 Bielen A et al, 2011, Enhanced efficacy of IGF1R inhibition in pediatric glioblastoma by combinatorial targeting of PDGFRalpha/ beta. Mol Cancer Ther 10(8):1407–1418 Gilbertson RJ et al, 2006, Mutational analysis of PDGFR-RAS/ MAPK pathway activation in childhood medulloblastoma. Eur J Cancer 42(5):646–649 Gilbertson RJ, Clifford SC, 2003, PDGFRB is overexpressed in metastatic medulloblastoma. Nat Genet 35(3):197–198 Abouantoun TJ, MacDonald TJ, 2009, Imatinib blocks migration and invasion of medulloblastoma cells by concurrently inhibiting activation of platelet-derived growth factor receptor and transactivation of epidermal growth factor receptor. Mol Cancer Ther 8(5):1137–1147 Abouantoun TJ, Castellino RC, MacDonald TJ, 2011, Sunitinib induces PTEN expression and inhibits PDGFR signaling and migration of medulloblastoma cells. J Neurooncol 101(2):215–226 Herrington B,KieranMW(2009, Small molecule inhibitors in children with malignant gliomas. Pediatr Blood Cancer 53(3):312–317 Goumnerova LC, 1996, Growth factor receptors and medulloblastoma. J Neurooncol 29(1):85–89 Goldbrunner RH et al, 1999, Different vascular patterns of medulloblastoma and supratentorial primitive neuroectodermal tumors. Int J Dev Neurosci 17(5–6):593–599 Sundberg C et al, 2001, Glomeruloid microvascular proliferation follows adenoviral vascular permeability factor/vascular endothelial growth factor-164 gene delivery. Am J Pathol 158(3):1145–1160 Jain S et al, 2012, Incremental increase in VEGFR1(+, hematopoietic progenitor cells and VEGFR2(+, endothelial progenitor cells predicts relapse and lack of tumor response in breast cancer patients. Breast Cancer Res Treat 132(1):235–242 Scully S et al, 2012, Transdifferentiation of glioblastoma stem-like cells into mural cells drives vasculogenic mimicry in glioblastomas. J Neurosci 32(37):12950–12960 Francescone R et al, 2012, Glioblastoma-derived tumor cells induce vasculogenic mimicry through Flk-1 protein activation. J Biol Chem 287(29):24821–24831 El Hallani S et al, 2010, A new alternative mechanism in glioblastoma vascularization: tubular vasculogenic mimicry. Brain 133(Pt 4):973–982 Nico B et al, 2010, Intussusceptive microvascular growth in human glioma. Clin Exp Med 10(2):93–98 Moreno L, et al., 2012, Role of platelet derived growth factor receptor, PDGFR, over-expression and angiogenesis in ependymoma. J Neurooncol Zavalhia LS et al, 2012, Evaluation of the expression of C-kit, CD117, in ependymomas and oligodendrogliomas. Dis Markers 33(2):61–68 Yao J et al, 2011, Expression of a functional VEGFR-1 in tumor cells is a major determinant of anti-PlGF antibodies efficacy. Proc Natl Acad Sci U S A 108(28):11590–11595 Lichtenberger BMet al, 2010, Autocrine VEGF signaling synergizes with EGFR in tumor cells to promote epithelial cancer development. Cell 140(2):268–279 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 417 EP 426 DI 10.1007/s12253-013-9711-4 PG 10 ER PT J AU Moghbeli, M Forghanifard, MM Aarabi, A Mansourian, A Abbaszadegan, RM AF Moghbeli, Meysam Forghanifard, Mahdi Mohammad Aarabi, Azadeh Mansourian, Akram Abbaszadegan, Reza Mohammad TI Clinicopathological Sex- Related Relevance of Musashi1 mRNA Expression in Esophageal Squamous Cell Carcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal squamous cell carcinoma; Musashi1 (Msi1); Expressional analysis; Real-time PCR ID Esophageal squamous cell carcinoma; Musashi1 (Msi1); Expressional analysis; Real-time PCR AB The cancer stem cell theory is considered as the spotlight of cancer biology, in which a subpopulation of tumor cells show unlimited proliferative and self renewal capacities. Post-transcriptional regulation is involved in different cellular functions such as cell differentiation and proliferation which results in cellular diversity. Musashi1 (Msi1) is one of the most important RNA-binding proteins (RBPs) which are involved in translational inhibition. Although, Msi1 targets are largely unknown, p21WAF-1, a cell cycle regulator, and Numb, inhibitor of notch signaling pathway, are well-known factors which are suppressed by the Msi1 in normal and cancer stem cells. Msi1 expression in tumor tissues from 53 ESCC patients was compared to normal tissues using realtime polymerase chain reaction (PCR). Msi1 was significantely overexpressed in 41.5 % of tumor samples and we observed a significant correlation between Msi1 expression and sex, in which the males had shown a higher level of Msi1 expression in comparison with the females (2.00 Vs 0.78 fold changes, p =0.05). In this study, we assessed whether Msi1 is expressed in ESCC samples suggesting this protein as a novel cancer stem cell marker which requires further studies. C1 [Moghbeli, Meysam] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, 9196773117 Mashhad, Iran. [Forghanifard, Mahdi Mohammad] Islamic Azad University, Damghan Branch, Department of BiologyDamghan, Iran. [Aarabi, Azadeh] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, 9196773117 Mashhad, Iran. [Mansourian, Akram] Mashhad University of Medical Sciences, Ghaem Hospital, Department of PathologyMashhad, Iran. [Abbaszadegan, Reza Mohammad] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, 9196773117 Mashhad, Iran. RP Abbaszadegan, RM (reprint author), Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, 9196773117 Mashhad, Iran. EM abbaszadeganmr@mums.ac.ir CR Gholamin M, Moaven O, Memar B, Farshchian M, Naseh H, Malekzadeh R, Sotoudeh M, Rajabi-Mashhadi MT, Forghani MN, Farrokhi F, Abbaszadegan MR, 2009, Overexpression and interactions of interleukin-10, transforming growth factor beta, and vascular endothelial growth factor in esophageal squamous cell carcinoma. World J Surg 33(7):1439–1445 Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A, Weinberg RA, 2008, An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet 40(5):499–507 Pardal R, Clarke MF, Morrison SJ, 2003, Applying the principles of stem-cell biology to cancer. Nat Rev Cancer 3(12):895–902 Schwanhausser B, Busse D, Li N, Dittmar G, Schuchhardt J,Wolf J, ChenW, SelbachM(2011)Global quantification of mammalian gene expression control. Nature 473(7347):337–342 Jackson RJ, Hellen CU, Pestova TV, 2010, The mechanism of eukaryotic translation initiation and principles of its regulation. Nat Rev Mol Cell Biol 11(2):113–127 Sonenberg N, Hinnebusch AG, 2009, Regulation of translation initiation in eukaryotes: mechanisms and biological targets. Cell 136(4):731–745 Grzmil M, Hemmings BA, 2012, Translation regulation as a therapeutic target in cancer. Cancer Res 72(16):3891–3900 Battelli C, Nikopoulos GN,Mitchell JG, Verdi JM(2006, The RNAbinding protein Musashi-1 regulates neural development through the translational repression of p21WAF-1. Mol Cell Neurosci 31(1):85– 96 Imai T, Tokunaga A, Yoshida T, Hashimoto M, Mikoshiba K, Weinmaster G, Nakafuku M, Okano H, 2001, The neural RNAbinding proteinMusashi1 translationally regulates mammalian numb gene expression by interactingwith itsmRNA.Mol Cell Biol 21(12): 3888–3900 Kawahara H, Imai T, Imataka H, TsujimotoM,Matsumoto K, Okano H, 2008, Neural RNA-binding protein Musashi1 inhibits translation initiation by competing with eIF4G for PABP. J Cell Biol 181(4): 639–653 Okano H, Kawahara H, ToriyaM, Nakao K, Shibata S, Imai T, 2005, Function of RNA-binding protein Musashi-1 in stem cells. Exp Cell Res 306(2):349–356 Guo M, Jan LY, Jan YN, 1996, Control of daughter cell fates during asymmetric division: interaction of Numb and Notch. Neuron 17(1): 27–41 DiMarcotullio L, Ferretti E, Greco A, De Smaele E, Po A, SicoMA, Alimandi M, Giannini G, Maroder M, Screpanti I, Gulino A, 2006, Numb is a suppressor of Hedgehog signalling and targets Gli1 for Itch-dependent ubiquitination. Nat Cell Biol 8(12):1415–1423 McGill MA, McGlade CJ, 2003, Mammalian numb proteins promote Notch1 receptor ubiquitination and degradation of the Notch1 intracellular domain. J Biol Chem 278(25):23196–23203 Wakamatsu Y, Maynard TM, Jones SU, Weston JA, 1999, NUMB localizes in the basal cortex of mitotic avian neuroepithelial cells and modulates neuronal differentiation by binding to NOTCH-1. Neuron 23(1):71–81 Glazer RI, Wang XY, Yuan H, Yin Y, 2008, Musashi1: a stem cell marker no longer in search of a function. Cell Cycle 7(17):2635–2639 Wang XY, Yin Y, Yuan H, Sakamaki T, Okano H, Glazer RI, 2008, Musashi1 modulates mammary progenitor cell expansion through proliferin-mediated activation of the Wnt and Notch pathways. Mol Cell Biol 28(11):3589–3599 Devgan V, Mammucari C, Millar SE, Brisken C, Dotto GP, 2005, p21WAF1/Cip1 is a negative transcriptional regulator of Wnt4 expression downstream of Notch1 activation. Genes Dev 19(12): 1485–1495 Li L, Yuan H,Weaver CD, Mao J, Farr GH 3rd, Sussman DJ, Jonkers J, Kimelman D, Wu D, 1999, Axin and Frat1 interact with dvl and GSK, bridging Dvl to GSK in Wnt-mediated regulation of LEF-1. Embo J 18(15):4233–4240 Toda M, Iizuka Y, Yu W, Imai T, Ikeda E, Yoshida K, Kawase T, Kawakami Y, Okano H, Uyemura K, 2001, Expression of the neural RNA-binding protein Musashi1 in human gliomas. Glia 34(1):1–7 GotteM,Wolf M, Staebler A, Buchweitz O, Kelsch R, Schuring AN, Kiesel L, 2008, Increased expression of the adult stem cell marker Musashi-1 in endometriosis and endometrial carcinoma. J Pathol 215(3):317–329 Shu HJ, Saito T, Watanabe H, Ito JI, Takeda H, Okano H, Kawata S, 2002, Expression of the Musashi1 gene encoding the RNA-binding protein in human hepatoma cell lines. Biochem Biophys Res Commun 293(1):150–154 Schulenburg A, Cech P, Herbacek I, Marian B, Wrba F, Valent P, Ulrich-Pur H, 2007, CD44-positive colorectal adenoma cells express the potential stem cell markers musashi antigen, msi1, and ephrin B2 receptor, EphB2). J Pathol 213(2):152–160 Forghanifard MM, Moaven O, Farshchian M, Montazer M, Raeisossadati R, Abdollahi A, Moghbeli M, Nejadsattari T, Parivar K, Abbaszadegan MR, 2012, Expression analysis elucidates the roles ofMAML1 and Twist1 in esophageal squamous cell carcinoma aggressiveness and metastasis. Ann Surg Oncol 19(3):743–749 Kaneko Y, Sakakibara S, Imai T, Suzuki A, Nakamura Y, Sawamoto K, Ogawa Y, Toyama Y, Miyata T, Okano H, 2000, Musashi1: an evolutionally conserved marker for CNS progenitor cells including neural stem cells. Dev Neurosci 22(1–2):139–153 Siddall NA, McLaughlin EA, Marriner NL, Hime GR, 2006, The RNA-binding protein Musashi is required intrinsically to maintain stem cell identity. Proc Natl Acad Sci U S A 103(22):8402–8407 Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, Bronner-Fraser M, Kornblum HI, 2003, Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci U S A 100(25):15178–15183 Yokota N, Mainprize TG, Taylor MD, Kohata T, Loreto M, Ueda S, Dura W, Grajkowska W, Kuo JS, Rutka JT, 2004, Identification of differentially expressed and developmentally regulated genes in medulloblastoma using suppression subtraction hybridization. Oncogene 23(19):3444–3453 Clement V, Sanchez P, de Tribolet N, Radovanovic I, Ruiz i Altaba A, 2007, HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr Biol 17(2):165–172 Gonzalez F, BarraganMonasterioM, Tiscornia G, Montserrat Pulido N, Vassena R, Batlle Morera L, Rodriguez Piza I, Izpisua Belmonte JC, 2009, Generation of mouse-induced pluripotent stem cells by transient expression of a single nonviral polycistronic vector. Proc Natl Acad Sci U S A 106(22):8918–8922 Stadtfeld M, Maherali N, Borkent M, Hochedlinger K, 2010, A reprogrammable mouse strain from gene-targeted embryonic stem cells. Nat Methods 7(1):53–55 Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, Slukvin II, Thomson JA, 2009, Human induced pluripotent stem cells free of vector and transgene sequences. Science 324(5928):797–801 Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin II, Thomson JA, 2007, Induced pluripotent stem cell lines derived from human somatic cells. Science 318(5858): 1917–1920 Potten CS, Booth C, Tudor GL, Booth D, Brady G, Hurley P, Ashton G, Clarke R, Sakakibara S, Okano H, 2003, Identification of a putative intestinal stem cell and early lineage marker; musashi-1. Differentiation 71(1):28–41 Bobryshev YV, Freeman AK, Botelho NK, Tran D, Levert-Mignon AJ, Lord RV, 2010, Expression of the putative stem cell marker Musashi-1 in Barrett’s esophagus and esophageal adenocarcinoma. Dis Esophagus 23(7):580–589 Nikpour P, Baygi ME, Steinhoff C, Hader C, Luca AC, Mowla SJ, Schulz WA, 2011, The RNA binding protein Musashi1 regulates apoptosis, gene expression and stress granule formation in urothelial carcinoma cells. J Cell Mol Med 15(5):1210–1224 Nikpour P, Emadi-Baygi M, Mohhamad-Hashem F, Maracy MR, Haghjooy-Javanmard S, 2013, MSI1 overexpression in diffuse type of gastric cancer. Pathol Res Pract 209(1):10–13 TodaroM, FrancipaneMG, Medema JP, Stassi G, 2010, Colon cancer stem cells: promise of targeted therapy. Gastroenterology 138(6): 2151–2162 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 427 EP 433 DI 10.1007/s12253-013-9712-3 PG 7 ER PT J AU Zhang, F Yang, J Li, Z AF Zhang, Fan Yang, Junlan Li, Zijian TI Trastuzumab-Induced Systemic Capillary Leak Syndrome in a Breast Cancer Patient SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SCLS; Trastuzumab; Chemotherapy; Breast cancer ID SCLS; Trastuzumab; Chemotherapy; Breast cancer AB Systemic capillary leak syndrome (SCLS) is a rare health condition. It is characterized by recurrent episodes of generalized edema and severe hypotension along with hypoproteinemia. The condition is under recognized because of its nonspecific signs and symptoms, and high mortality rate. SCLS triggered by trastuzumab, a target drug for Her2- positive breast cancer patients, has not been previously reported. A 59-year-old Chinese woman, diagnosed with breast cancer with accompanying liver and bone metastasis, was treated with 3 cycles of docetaxel with capecitabine and a regimen of 12 cycles of capecitabine with trastuzumab. The patient developed systemic capillary leak syndrome during the 16th cycle of chemotherapy. Post-diagnosis treatment regimen is also presented in the current case report. SCLS has been previously observed in breast cancer patients. However, SCLS incidence post-chemotherapeutic treatment with trastuzumab has not been reported elsewhere. Hence, our report highlights the need for rigorous investigation of the side effects of trastuzumab usage and the increasing need of insightful diagnosis to manage any incidence of SCLS. The case provides valuable experience for treating the uncommon adverse effects of trastuzumab in Her2-positive breast cancer patients. C1 [Zhang, Fan] PLA General Hospital, Tumor Center, Key LaboratoryBeijing, China. [Yang, Junlan] PLA General Hospital, Tumor Center, Oncology Department, No. 28 Fuxing Road, Haidian District, 100853 Beijing, China. [Li, Zijian] PLA No.161 Hospital, Oncology DepartmentWuhan, China. RP Yang, J (reprint author), PLA General Hospital, Tumor Center, Oncology Department, 100853 Beijing, China. EM yangjunlan1123@126.com CR Clarkson B, Thompson D, Horwith M et al, 1960, Cyclical edema and shock due to increased capillary permeability. Am J Med 29:193–216 Dhir V, Arya V, Malav IC et al, 2007, Idiopathic systemic capillary leak syndrome, SCLS): case report and systematic review of cases reported in the last 16 years. Intern Med 46:899–904 de Martino M, Sasso L, Pirozzi F et al, 2009, Systemic capillary leak syndrome or Clarkson’s disease: a case report. Intern Emerg Med 4: 357–358 Druey KM, Greipp PR, 2010, Narrative review: the systemic capillary leak syndrome. Ann Intern Med 153:90–98 Hollenberg J, Frykman J, Lundberg LG et al, 2010, A case report of systemic capillary leak syndrome, Clarkson’s disease). Acta Anaesthesiol Scand 54:649–652 Guillaume M, Tolsma V, Colombe B et al, 2011, Idiopathic systemic capillary leak syndrome: a case report with cardiac involvement. Rev Med Interne 32:e69–e71 Hudis CA, 2007, Trastuzumab–mechanism of action and use in clinical practice. N Engl J Med 357:39–51 Bencsath KP, Reu F, Dietz J et al, 2011, Idiopathic systemic capillary leak syndrome preceding diagnosis of infiltrating lobular carcinoma of the breast with quiescence during neoadjuvant chemotherapy. Mayo Clin Proc 86:260–261 Kai-Feng W, Hong-Ming P, Hai-Zhou L et al, 2011, Interleukin-11- induced capillary leak syndrome in primary hepatic carcinoma patients with thrombocytopenia. BMC Cancer 11:204 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 435 EP 437 DI 10.1007/s12253-013-9713-2 PG 3 ER PT J AU Xu, D Yu, Y Zhu, Y Huang, T Chen, Y Qi, J AF Xu, Ding Yu, Yongjiang Zhu, Yunkai Huang, Tao Chen, Yaqing Qi, Jun TI A new Model Consists of Intravesical Prostatic Protrusion, Prostate Volume and Serum Prostatic-Specific Antigen in the Evaluation of Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intravesical prostatic protrusion; Prostate specific antigen; Prostate cancer; Diagnostic accuracy ID Intravesical prostatic protrusion; Prostate specific antigen; Prostate cancer; Diagnostic accuracy AB The Prostate-specific antigen (PSA) level is largely used to diagnose prostate cancer (PCa) in last decades. However, its specificity is low in patients with a PSA level ranging from 4.0 to 10.0 ng/ml. This study aims to define the correlation between intravesical prostatic protrusion (IPP) and PSA and to establish a new model to predict PCa. A total of 339 patients order than 45 years examined between October 2010 and June 2012 were enrolled. Eligible patients were recommended for transrectal ultrasonography (TRUS)-guided prostate biopsies after measuring total prostate volume (TPV), tranzisional zone volume (TZV) and IPP. The levels of total PSA (tPSA), free PSA (fPSA) were analyzed by using Hybritech calibrated Access tPSA and fPSA assays. A new mathematical model, named IPP removed PCa predicting score (IRPPS), consists of tPSA, TZV and IPP was established. The predictive accuracy of IRPPS, PSA density (PSAD), %PSA and tPSA were compared using receiveroperator characteristic (ROC) analysis. Eighty-six patients had PSA levels of 4.0–10.0 ng/ml. Twenty of them were diagnosed as PCa. Using ROC curves, the areas under the curve for IRPPS, PSAD and %PSA and tPSA were 0.786, 0.768 and 0.664 and 0.585, respectively. We suggested IPP grade had a significant relationship with serum tPSA levels. The predictive accuracy of IRPPS was higher than the other 3 indictors. C1 [Xu, Ding] XinHua Hospital Affiliate to Shanghai Jiao Tong University School of Medicine, Department of UrologyShanghai, China. [Yu, Yongjiang] XinHua Hospital Affiliate to Shanghai Jiao Tong University School of Medicine, Department of UrologyShanghai, China. [Zhu, Yunkai] XinHua Hospital Affiliate to Shanghai Jiao Tong University School of Medicine, Department of UltrasoundShanghai, China. [Huang, Tao] XinHua Hospital Affiliate to Shanghai Jiao Tong University School of Medicine, Department of UrologyShanghai, China. [Chen, Yaqing] XinHua Hospital Affiliate to Shanghai Jiao Tong University School of Medicine, Department of UltrasoundShanghai, China. [Qi, Jun] XinHua Hospital Affiliate to Shanghai Jiao Tong University School of Medicine, Department of UrologyShanghai, China. RP Qi, J (reprint author), XinHua Hospital Affiliate to Shanghai Jiao Tong University School of Medicine, Department of Urology, Shanghai, China. EM qijunqijunqqjj@hotmail.com CR Stephan C, Jung K, Lein M, Diamandis EP, 2007, PSA and other tissue kallikreins for prostate cancer detection. Eur J Cancer 43(13): 1918–1926., DOI 10.1016/j.ejca.2007.06.006 Tchetgen MB, Oesterling JE, 1997, The effect of prostatitis, urinary retention, ejaculation, and ambulation on the serum prostate-specific antigen concentration. Urol Clin North Am 24(2):283–291 Schroder FH, Roobol MJ, 2009, Defining the optimal prostatespecific antigen threshold for the diagnosis of prostate cancer. Curr Opin Urol 19(3):227–231., DOI 10.1097/MOU.0b013e328329a2d0 Lee JW, Ryu JH, Yoo TK, Byun SS, Jeong YJ, Jung TY, 2012, Relationship between Intravesical prostatic protrusion and postoperative outcomes in patients with benign prostatic hyperplasia. Korean J Urol 53(7):478–482., DOI 10.4111/kju.2012.53.7.478 Lim KB, Ho H, Foo KT, Wong MY, Fook-Chong S, 2006, Comparison of intravesical prostatic protrusion, prostate volume and serum prostatic-specific antigen in the evaluation of bladder outlet obstruction. Int J Urol 13(12):1509–1513., DOI 10.1111/j. 1442-2042.2006.01611.x Lee LS, Sim HG, Lim KB, Wang D, Foo KT, 2010, Intravesical prostatic protrusion predicts clinical progression of benign prostatic enlargement in patients receiving medical treatment. Int J Urol 17(1): 69–74., DOI 10.1111/j.1442-2042.2009.02409.x Yuen JS, Ngiap JT, Cheng CW, Foo KT, 2002, Effects of bladder volume on transabdominal ultrasound measurements of intravesical prostatic protrusion and volume. Int J Urol 9(4):225–229 Milonas D, Trumbeckas D, 2003, Prostate-specific antigen and transition zone index - powerful predictors for acute urinary retention in men with benign prostatic hyperplasia. Medicina, Kaunas, 39(11): 1071–1077 Catalona WJ, Partin AW, Slawin KM, Brawer MK, Flanigan RC, Patel A, Richie JP, deKernion JB,Walsh PC, Scardino PT, Lange PH, Subong EN, Parson RE, Gasior GH, Loveland KG, Southwick PC, 1998, Use of the percentage of free prostate-specific antigen to enhance differentiation of prostate cancer from benign prostatic disease: a prospective multicenter clinical trial. JAMA 279(19): 1542–1547 Catalona WJ, Partin AW, Sanda MG, Wei JT, Klee GG, Bangma CH, SlawinKM, Marks LS, Loeb S, BroylesDL, Shin SS,CruzAB, Chan DW, Sokoll LJ, Roberts WL, van Schaik RH, Mizrahi IA, 2011, A multicenter study of [−2]pro-prostate specific antigen combined with prostate specific antigen and free prostate specific antigen for prostate cancer detection in the 2.0 to 10.0 ng/ml prostate specific antigen range. J Urol 185(5):1650–1655., DOI 10.1016/j.juro.2010.12.032 Reis LO, Barreiro GC, Baracat J, Prudente A, D’Ancona CA, 2008, Intravesical protrusion of the prostate as a predictive method of bladder outlet obstruction. Int Braz J Urol 34(5):627–633, discussion 634–627 Chia SJ, Heng CT, Chan SP, Foo KT, 2003, Correlation of intravesical prostatic protrusion with bladder outlet obstruction. BJU Int 91(4):371–374 Nose H, Foo KT, Lim KB, Yokoyama T, Ozawa H, Kumon H, 2005, Accuracy of two noninvasive methods of diagnosing bladder outlet obstruction using ultrasonography: intravesical prostatic protrusion and velocity-flow video urodynamics. Urology 65(3):493–497., DOI 10.1016/j.urology.2004.10.014 Keqin Z, Zhishun X, Jing Z, Haixin W, Dongqing Z, Benkang S, 2007, Clinical significance of intravesical prostatic protrusion in patients with benign prostatic enlargement. Urology 70(6):1096– 1099., DOI 10.1016/j.urology.2007.08.008 Laniado ME, Ockrim JL, Marronaro A, Tubaro A, Carter SS, 2004, Serum prostate-specific antigen to predict the presence of bladder outlet obstruction in men with urinary symptoms. BJU Int 94(9): 1283–1286., DOI 10.1111/j.1464-410X.2004.05158.x van Renterghem K, van Koeveringe G, Achten R, van Kerrebroeck P, 2009, Long-term clinical outcome of diagnostic transurethral resection of the prostate in patients with elevated prostate-specific antigen level andminor lower urinary tract symptoms. Urol Int 83(1): 60–65., DOI 10.1159/000224870 Kuo HC, 1999, Clinical prostate score for diagnosis of bladder outlet obstruction by prostate measurements and uroflowmetry. Urology 54(1):90–96 Hammerer PG, McNeal JE, Stamey TA, 1995, Correlation between serum prostate specific antigen levels and the volume of the individual glandular zones of the human prostate. J Urol 153(1): 111–114., DOI 10.1097/00005392-199501000-00038 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 439 EP 443 DI 10.1007/s12253-013-9714-1 PG 5 ER PT J AU Lu, Rl Hu, ChP Yang, Hp Li, Yy Gu, Qh Wu, L AF Lu, Rong-li Hu, Cheng-Ping Yang, Hua-ping Li, Yuan-yuan Gu, Qi-hua Wu, Lielin TI Biological Characteristics and Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors Efficacy of EGFR Mutation and its Subtypes in Lung Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung adenocarcinoma (LAC); Epidermal growth factor receptor (EGFR); Erlotinib; Gefitinib; Exon 19 deletions (del 19); Exon 21 point mutation (L858R) ID Lung adenocarcinoma (LAC); Epidermal growth factor receptor (EGFR); Erlotinib; Gefitinib; Exon 19 deletions (del 19); Exon 21 point mutation (L858R) AB Mutation of epidermal growth factor receptor (EGFR) gene has been reported to be present in lung adenocarcinoma (LAC). In this study, we extensively investigated the impact of patients’ biological characteristics on EGFR mutation and the impact of EGFR mutation subtypes on targeted therapy of advanced LAC. We examined EGFR exons18to21status in169 LAC patients by direct sequencing to study the impact of patients’ biological characteristics on the EGFR mutational spectrum. And then, 59 patients with advanced LAC harboring EGFR exon 19 deletions(del 19) or exon 21 point mutation(L858R) mutations received first-line treatment of gefitinib or erlotinib, the efficacy of treatment, and the progression-free survival (PFS) of these patients were recorded. The frequency of the EGFR mutation and its subtypes and the variables associated with the EGFR mutation after removing the confound factors were investigated by the logistic analysis using all samples (n =169). The EGFR mutation was significantly associated with well-differentiated tumor and excessive household cooking fumes(P <0.05). The deletions in exon 19 were more frequently associated with well-differentiated tumor (P <0.05). The overall frequency of the EGFR mutation was 49%. Then the impact of EGFR mutation subtypes on targeted therapy were investigated by the retrospective analysis on 59 advanced LAC patients with del 19 or L858R mutations and treated first-line with erlotinib or gefitinib. The deletions in exon 19 got longer PFS (P < 0.05). But there were no differences in PFS between erlotinib therapy and gefitinib therapy. EGFR mutations were more frequently in high tumor differentiation and excessive household cooking fumes LAC. The del 19 mutation rate is relatively high with a high differentiation degree in advanced lung adenocarcinoma. The deletions in exon 19 may benefit more from first-line targeted therapy of advanced LAC compared with exon 21 point mutation L858R. There was no significant difference between the efficacy of gefitinib and erlotinib treatments associated with EGFR mutation and its subtypes. C1 [Lu, Rong-li] Central South University, Xiangya Hospital, Department of Respiratory, 410008 Changsha, Hunan, China. [Hu, Cheng-Ping] Central South University, Xiangya Hospital, Department of Respiratory, 410008 Changsha, Hunan, China. [Yang, Hua-ping] Central South University, Xiangya Hospital, Department of Respiratory, 410008 Changsha, Hunan, China. [Li, Yuan-yuan] Central South University, Xiangya Hospital, Department of Respiratory, 410008 Changsha, Hunan, China. [Gu, Qi-hua] Central South University, Xiangya Hospital, Department of Respiratory, 410008 Changsha, Hunan, China. [Wu, Lielin] Central South University, Xiangya Hospital, Department of General Surgery, 410008 Changsha, Hunan, China. RP Hu, ChP (reprint author), Central South University, Xiangya Hospital, Department of Respiratory, 410008 Changsha, China. EM huchengp28@126.com CR Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics, 2012. CA Cancer J Clin 62(1):10–29 Herbst RS, Heymach JV, Lippman SM(2008, Lung cancer. N Engl J Med 359(13):1367–1380 Mok TS, Wu YL, Thongprasert S et al, 2009, Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 361(10):947–957 Mitsudomi T, Morita S, Yatabe Y et al, 2010, Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor, WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 11(2):121–128 Lynch TJ, Bell DW, Sordella R et al, 2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350(21):2129– 2139 Paez JG, Janne PA, Lee JC et al, 2004, EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304(5676):1497–1500 Mok TS, Wu YL, Thongprasert S et al, 2009, Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 361(10):947–957 Yang CH, Yu CJ, Shih JY et al, 2008, Specific EGFR mutations predict treatment outcome of stage IIIB/IV patients with chemotherapy-naive non-small-cell lung cancer receiving first-line gefitinib monotherapy. J Clin Oncol 26(16):2745–2753 Rosell R, Moran T, Queralt C et al, 2009, Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med 361(10):958–967 Thatcher N, Chang A, Parikh P et al, 2005, Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study, Iressa Survival Evaluation in Lung Cancer). Lancet 366(9496):1527–1537 Shepherd FA, Rodrigues PJ, Ciuleanu T et al, 2005, Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 353(2):123–132 Pao W, Miller V, Zakowski M et al, 2004, EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A 101(36):13306–13311 Taron M, Ichinose Y, Rosell R et al, 2005, Activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor are associated with improved survival in gefitinib-treated chemorefractory lung adenocarcinomas. Clin Cancer Res 11(16): 5878–5885 Uramoto H, Sugio K, Oyama T et al, 2006, Epidermal growth factor receptor mutations are associated with gefitinib sensitivity in nonsmall cell lung cancer in Japanese. Lung Cancer 51(1):71–77 Li M, Yin Z, Guan P et al, 2008, XRCC1 polymorphisms, cooking oil fume and lung cancer in Chinese women nonsmokers. Lung Cancer 62(2):145–151 Ko YC, Cheng LS, Lee CH et al, 2000, Chinese food cooking and lung cancer in women nonsmokers.AmJ Epidemiol 151(2):140–147 Metayer C,Wang Z, Kleinerman RA et al, 2002, Cooking oil fumes and risk of lung cancer in women in rural Gansu. China Lung Cancer 35(2):111–117 Lin Y, Cai L, 2012, Environmental and dietary factors and lung cancer risk among Chinese women: a case–control study in Southeast China. Nutr Cancer 64(4):508–514 Yang HH, Chien SM, Lee HL et al, 2007, Emission of trans,trans-2, 4-decadienal from restaurant exhausts to the atmosphere. Atmos Environ, 41: 5327–5333 Chang LW, Lo WS, Lin P, 2005, Trans, trans-2,4-decadienal, a product found in cooking oil fumes, induces cell proliferation and cytokine production due to reactive oxygen species in human bronchial epithelial cells. Toxicol Sci 87(2):337–343 Meng-hong S, Fei Y, Lei S, 2011, Detection of epidermal growth factor receptor mutations in non-small-cell lung carcinoma by direct sequencing and correlations with clinicopathological characteristics and sample types. Chinese J Pathol 40(10):655–659 Miao-Jin F, Hai-Gang L, Zhi-Qiang L et al, 2011, Relationship of epidermal growth factor receptor gene mutations, clinicopathologic features and prognosis in patients with non-small cell lung cancer. Chinese J Pathol 40(10):679–682 Tomizawa Y, Iijima H, Sunaga N et al, 2005, Clinicopathologic significance of the mutations of the epidermal growth factor receptor gene in patients with non-small cell lung cancer. Clin Cancer Res 11(19 Pt 1):6816–6822 Tam IY, Chung LP, Suen WS et al, 2006, Distinct epidermal growth factor receptor and KRAS mutation patterns in non-small cell lung cancer patients with different tobacco exposure and clinicopathologic features. Clin Cancer Res 12(5):1647–1653 Rosell R, Moran T, Queralt C et al, 2009, Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med 361(10):958–967 Shigematsu H, Gazdar AF, 2006, Somatic mutations of epidermal growth factor receptor signaling pathway in lung cancers. Int J Cancer 118(2):257–262 Kim ST, Lee J, Kim JH et al, 2010, Comparison of gefitinib versus erlotinib in patients with nonsmall cell lung cancer who failed previous chemotherapy. Cancer 116(12):3025–3033 Kim ST, Uhm JE, Lee J et al, 2012, Randomized phase II study of gefitinib versus erlotinib in patients with advanced non-small cell lung cancer who failed previous chemotherapy. Lung Cancer 75(1):82–88 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 445 EP 451 DI 10.1007/s12253-013-9715-0 PG 7 ER PT J AU Paschke, L Rucinski, M Ziolkowska, A Zemleduch, T Malendowicz, W Kwias, Z Malendowicz, KL AF Paschke, Lukasz Rucinski, Marcin Ziolkowska, Agnieszka Zemleduch, Tomasz Malendowicz, Witold Kwias, Zbigniew Malendowicz, K Ludwik TI ZFP91—A Newly Described Gene Potentially Involved in Prostate Pathology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Benign prostate hyperplasia; Prostate cancer; Prostate cancer cell lines; ZFP91 ID Benign prostate hyperplasia; Prostate cancer; Prostate cancer cell lines; ZFP91 AB In search for novel molecular targets in benign prostate hyperplasia (BPH), a PCR Array based screening of 84 genes was performed. Of those, expression of ZFP91 (ZFP91 zinc finger protein) was notably upregulated. Limited data concerning the function of ZFP91 product show that it is a potential transcription factor upregulated in human acute myelogenous leukemia and most recently found to be the noncanonical NF-κB pathway regulator. In order to test this finding on a larger number of samples, prostate specimens were obtained from patients undergoing adenomectomy for BPH (n =21), and as a control, from patients undergoing radical cystectomy for bladder cancer (prostates unchanged pathologically, n =18). Similar studies were performed on cultured human prostate cancer cell lines: LNCaP, DU145, 22Rv1, PC-3; as well as normal prostate epithelial cells-PrEC. Methods employed included: Human Obesity PCR Array (Qiagen), QPCR and Western blotting. QPCR studies confirmed significant overexpression of ZFP91 in BPH samples. On a protein level, however, comparison between normal and BPH prostates revealed insignificant differences. As for prostate cell lines examined, all expressed ZFP91 mRNA. Western blotting analysis showed markedly higher protein levels of ZFP91 in all cancer cell lines in comparison with normal (PrEC) cells. In conclusion, the upregulated ZFP91 mRNA in BPH, not accompanied by parallel changes in ZFP91 protein levels, together with ZFP91 protein abundance in prostate cancer cell lines suggest ZFP91 involvement in these prostate diseases. C1 [Paschke, Lukasz] Poznan University of Medical Sciences, Department of Histology and Embryology, 6 Swiecicki St, 60-781 Poznan, Poland. [Rucinski, Marcin] Poznan University of Medical Sciences, Department of Histology and Embryology, 6 Swiecicki St, 60-781 Poznan, Poland. [Ziolkowska, Agnieszka] Poznan University of Medical Sciences, Department of Histology and Embryology, 6 Swiecicki St, 60-781 Poznan, Poland. [Zemleduch, Tomasz] Poznan University of Medical Sciences, Department of Histology and Embryology, 6 Swiecicki St, 60-781 Poznan, Poland. [Malendowicz, Witold] Poznan University of Medical Sciences, Department of Histology and Embryology, 6 Swiecicki St, 60-781 Poznan, Poland. [Kwias, Zbigniew] Poznan University of Medical Sciences, Department of Urology and UrooncologyPoznan, Poland. [Malendowicz, K Ludwik] Poznan University of Medical Sciences, Department of Urology and UrooncologyPoznan, Poland. RP Paschke, L (reprint author), Poznan University of Medical Sciences, Department of Histology and Embryology, 60-781 Poznan, Poland. EM paschkelukasz@gmail.com CR Tewari R, Rajender S, Natu SM, Dalela D, Goel A, Goel MM, Tandon P, 2012, Diet, obesity, and prostate health: are we missing the link? Int J Androl 33:763–776 Allott EH, Masko EM, Freedland SJ, 2013, Obesity and prostate cancer: weighing the evidence. Eur Urol 63:800–809 Kopp RP, Han M, Partin AW, Humphreys E, Freedland SJ, Parsons JK, 2011, Obesity and prostate enlargement in men with localized prostate cancer. BJU Int 108:1750–1755 Parsons JK, Carter HB, Partin AW, Windham BG, Metter EJ, Ferrucci L, Landis P, Platz EA, 2006, Metabolic factors associated with benign prostatic hyperplasia. J Clin EndocrinolMetab 91:2562– 2568 Unoki M, Okutsu J, Nakamura Y, 2003, Identification of a novel human gene, ZFP91, involved in acute myelogenous leukemia. Int J Oncol 22:1217–1223 Saotome Y, Winter CG, Hirsh D, 1995, A widely expressed novel C2H2 zinc-finger protein with multiple consensus phosphorylation sites is conserved in mouse and man. Gene 152:233–238 Tompkins V, Hagen J, Zediak VP, Quelle DE, 2006, Identification of novel ARF binding proteins by two-hybrid screening. Cell Cycle 5: 641–646 Jin X, Jin HR, JungHS, Lee SJ, Lee JH, Lee JJ, 2010)An atypical E3 ligase zinc finger protein 91 stabilizes and activates NF-kappaBinducing kinase via Lys63-linked ubiquitination. J Biol Chem 285: 30539–30547 Lee JJ, Lee J-H, Lee K, HongY-S, Jin X, 2008, Therapeutic agent for cancer, inflammation, and auto-immune disease containing inhibitor of Zinc Finger Protein 91. US Patent 20,080,248,024 Jin HR, Jin X, Lee JJ, 2010, Zinc-finger protein 91 plays a key role in LIGHT-induced activation of non-canonical NF-kappaB pathway. Biochem Biophys Res Commun 400:581–586 Malendowicz W, Rucinski M, Macchi C, Spinazzi R, Ziolkowska A, Nussdorfer GG, Kwias Z, 2006, Leptin and leptin receptors in the prostate and seminal vesicles of the adult rat. Int JMol Med 18:615–618 Paschke L, Zemleduch T, Rucinski M, Ziolkowska A, Szyszka M, Malendowicz LK, 2010, Adiponectin and adiponectin receptor system in the rat adrenal gland: ontogenetic and physiologic regulation, and its involvement in regulating adrenocortical growth and steroidogenesis. Peptides 31:1715–1724 Malendowicz W, Szyszka M, Ziolkowska A, Rucinski M, Kwias Z, 2011, Elevated expression of orexin receptor 2, HCRTR2, in benign prostatic hyperplasia is accompanied by lowered serum orexin A concentrations. Int J Mol Med 27:377–383 RucinskiM, Ziolkowska A, Szyszka M,Hochol A,Malendowicz LK, 2012, Evidence suggesting that ghrelin O-acyl transferase inhibitor acts at the hypothalamus to inhibit hypothalamo-pituitaryadrenocortical axis function in the rat. Peptides 35:149–159 Ohl F, Jung M, Xu C, Stephan C, Rabien A, Burkhardt M, Nitsche A, Kristiansen G, Loening SA, Radonic A, Jung K, 2005, Gene expression studies in prostate cancer tissue: which reference gene should be selected for normalization? J Mol Med, Berl, 83:1014– 1024 Rucinski M, Ziolkowska A, Neri G, Trejter M, Zemleduch T, Tyczewska M, Nussdorfer GG, Malendowicz LK, 2007, Expression of neuromedins S and U and their receptors in the hypothalamus and endocrine glands of the rat. Int J Mol Med 20:255–259 Maier T, Guell M, Serrano L, 2009, Correlation of mRNA and protein in complex biological samples. FEBS Lett 583:3966– 3973 Taniguchi Y, Choi PJ, Li GW, Chen H, Babu M, Hearn J, Emili A, Xie XS, 2010, Quantifying E. coli proteome and transcriptome with single-molecule sensitivity in single cells. Science 329:533–538 Maier T, Schmidt A, Guell M, Kuhner S, Gavin AC, Aebersold R, Serrano L, 2011, Quantification of mRNA and protein and integration with protein turnover in a bacterium. Mol Syst Biol 7:511 Vogel C, Marcotte EM, 2012, Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet 13:227–232 Chen G, Gharib TG, Huang CC, Taylor JM, Misek DE, Kardia SL, Giordano TJ, Iannettoni MD, Orringer MB, Hanash SM, Beer DG, 2002, Discordant protein and mRNA expression in lung adenocarcinomas. Mol Cell Proteomics MCP 1:304–313 Koumenis C, Alarcon R, Hammond E, Sutphin P, Hoffman W, Murphy M, Derr J, Taya Y, Lowe SW, Kastan M, Giaccia A, 2001, Regulation of p53 by hypoxia: dissociation of transcriptional repression and apoptosis from p53-dependent transactivation. Mol Cell Biol 21:1297–1310 Vallabhapurapu S, Karin M, 2009, Regulation and function of NFkappaB transcription factors in the immune system. Annu Rev Immunol 27:693–733 Harhaj EW, Dixit VM, 2011, Deubiquitinases in the regulation of NF-kappaB signaling. Cell Res 21:22–39 Sun SC, 2011, Non-canonical NF-kappaB signaling pathway. Cell Res 21:71–85 Xiao G, Fu J, 2011, NF-kappaB and cancer: a paradigm of Yin-Yang. Am J Cancer Res 1:192–221 Jain G, Cronauer MV, Schrader M, Moller P, Marienfeld RB, 2012, NF-kappaB signaling in prostate cancer: a promising therapeutic target? World J Urol 30:303–310 Gasparian AV, Yao YJ, Kowalczyk D, Lyakh LA, Karseladze A, Slaga TJ, Budunova IV, 2002, The role of IKK in constitutive activation of NF-kappaB transcription factor in prostate carcinoma cells. J Cell Sci 115:141–151 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 453 EP 459 DI 10.1007/s12253-013-9716-z PG 7 ER PT J AU Liu, K Gu, MM Chen, HL You, QSh AF Liu, Kun Gu, Ming-Ming Chen, Hong-Lin You, Qing-Sheng TI NOB1 in Non-small-cell Lung Cancer: Expression Profile and Clinical Significance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small-cell lung cancer; NOB1; Tumor–specific gene ID Non-small-cell lung cancer; NOB1; Tumor–specific gene AB Nin one binding (NOB1) gene has been reported up-regulated in several types of cancer. The aim of this study was to investigate the expression profile of NOB1 in nonsmall- cell lung cancer (NSCLC) and assess the clinical significance. qRT-PCR was used in the detection of NOB1 mRNA expression both in NSCLC tissue and in adjacent normal lung tissue. Western blot analysis and immunohistochemistry were used in the detection of NOB1 protein expression. The clinicopathological implications of NOB1 were analyzed statistically. It was confirmed by RT-qPCR that expression of NOB1 mRNA in NSCLC cells was higher than in human lung cells (P <0.05), and NOB1 mRNA was also over-expressed in NSCLC tissue when compared with adjacent tissue and normal lung tissue (P <0.05). Western blot analysis showed that NOB1 protein was significant increased in NSCLC cell lines compared with human lung cell line. Western blot analysis and immunohistochemistry showed that NOB1 protein was significant increased in NSCLC tissue compared with adjacent tissue and normal lung tissue (P <0.05). There were significant associations between NOB1 expression and TNM stage, lymph node metastasis, and histopathological grade (P <0.05), but not gender, age, smoke, or tumor diameter (P >0.05). Our results suggest that enhanced expression of NOB1 gene plays an important role in the occurrence and development of NSCLC. NOB1 may be a potential therapeutic target in NSCLC. C1 [Liu, Kun] Affiliated Hospital of Nantong University, Department of Cardiothoracic Surgery, Xi Si Road 20#, 226001 Nantong, Jiangsu Province, China. [Gu, Ming-Ming] Affiliated Hospital of Nantong University, Department of Cardiothoracic Surgery, Xi Si Road 20#, 226001 Nantong, Jiangsu Province, China. [Chen, Hong-Lin] Nantong University, Qi Xiu Road 19#, 226001 Nantong, Jiangsu Province, China. [You, Qing-Sheng] Affiliated Hospital of Nantong University, Department of Cardiothoracic Surgery, Xi Si Road 20#, 226001 Nantong, Jiangsu Province, China. RP You, QSh (reprint author), Affiliated Hospital of Nantong University, Department of Cardiothoracic Surgery, 226001 Nantong, China. EM pphss@126.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63(1):11–30 Lamanna AC, Karbstein K, 2009, NOB1 binds the single-stranded cleavage site D at the 30-end of 18S rRNAwith its PIN domain. Proc Natl Acad Sci U S A 106:14259–14264 Lin S,MengW, ZhangWet al, 2013, Expression of the NOB1 gene and its clinical significance in papillary thyroid carcinoma. J Int Med Res 41(3):568–572 Lin Y, Peng S, Yu H, Teng H, Cui M, 2012, RNAi-mediated downregulation of NOB1 suppresses the growth and colonyformation ability of human ovarian cancer cells. Med Oncol 29(1): 311–317 Lu Z, Guo Q, Shi A, Xie F, Lu Q, 2012, Downregulation of NIN/ RPN12 binding protein inhibit the growth of human hepatocellular carcinoma cells. Mol Biol Rep 39(1):501–507 Huang WY, Chen DH, Ning L, Wang LW, 2012, siRNA mediated silencing of NIN1/RPN12 binding protein 1 homolog inhibits proliferation and growth of breast cancer cells. Asian Pac J Cancer Prev 13(5):1823–1827 Zhang Y, Ni J, Zhou G et al, 2005, Cloning, expression and characterization of the human NOB1 gene. Mol Biol Rep 32: 185–189 Ettinger DS, AkerleyW, Borghaei H et al, 2012, Non-small cell lung cancer. J Natl Compr Canc Netw 10(10):1236–1271 Ettinger DS, Akerley W, Borghaei H et al, 2013, Non-Small Cell Lung Cancer, Version 2.2013. J Natl Compr Canc Netw 11(6):645– 653 Coate LE, John T, Tsao MS, Shepherd FA, 2009, Molecular predictive and prognostic markers in non-small-cell lung cancer. Lancet Oncol 10(10):1001–1010 Zhu CQ, Shih W, Ling CH, Tsao MS, 2006, Immunohistochemical markers of prognosis in non-small cell lung cancer: a review and proposal for a multiphase approach to marker evaluation. J Clin Pathol 59(8):790–800 Oyama T, Osaki T, Baba T, Nagata Y, Mizukami M, So T, Nakata S, Ichiki Y, Uramoto H, Sugaya M, Yoshimatsu T, Morita M, Hanagiri T, Sugio K, Kawamoto T, Yasumoto K, 2005, Molecular genetic tumor markers in non-small cell lung cancer. Anticancer Res 25(2B): 1193–1196 Tone Y, Tanahashi N, Tanaka K, Fujimuro M, Yokosawa H, Toh-e A, 2000, NOB1p, a new essential protein, associates with the 26S proteasome of growing saccharomyces cerevisiae cells. Gene 243(1–2):37–45 Tone Y, Toh-E A. NOB1p is required for biogenesis of the 26S proteasome and degraded upon its maturation in Saccharomyces cerevisiae. Genes Dev. 2002,15;16(24):3142– 57. Tu Y, Chen C, Pan J, Xu J, Zhou ZG,Wang CY(2012, The Ubiquitin Proteasome Pathway, UPP, in the regulation of cell cycle control and DNA damage repair and its implication in tumorigenesis. Int J Clin Exp Pathol 5(8):726–738 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 461 EP 466 DI 10.1007/s12253-013-9717-y PG 6 ER PT J AU Grigoryeva, SE Cherdyntseva, VN Karbyshev, SM Volkomorov, VV Stepanov, I Zavyalova, VM Perelmuter, MV Buldakov, AM Afanasjev, GS Tuzikov, AS Bukurova, AY Lisitsyn, AN Beresten, FS AF Grigoryeva, S Evgeniya Cherdyntseva, V Nadezhda Karbyshev, S Mikhail Volkomorov, V Viktor Stepanov, IvanV Zavyalova, V Marina Perelmuter, M Vladimir Buldakov, A Mikhail Afanasjev, G Sergey Tuzikov, A Sergey Bukurova, A Yulia Lisitsyn, A Nikolai Beresten, F Sergey TI Expression of Cyclophilin A in Gastric Adenocarcinoma Patients and Its Inverse Association with Local Relapses and Distant Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Stomach cancer; Two-dimensional gel electrophoresis; Proteomics; Mass spectrometry; Cancer markers ID Stomach cancer; Two-dimensional gel electrophoresis; Proteomics; Mass spectrometry; Cancer markers AB The aim of this study was to identify new protein markers of the intestinal and diffuse type gastric adenocarcinoma and to determine their relation to local relapses and distantmetastasis. Using two-dimensional gel electrophoresis, we searched for proteins that are overexpressed in the intestinal and/or diffuse type gastric adenocarcinoma, as compared to matched normal mucosa samples with further change confirmation by Western blot. Expression of the selected proteins was further assessed by immunohistocemistry in a large panel of gastric adenocarcinoma with various clinicopathological features. Expression level of cyclophilin A measured with western blot appeared to be increased on average ten times in 63 % of gastric adenocarcinoma vs. paired samples of normal mucosa. The frequency of immunihistochemistry detected cyclophilin A protein expression was found to be equal in tumor of both histotypes, but staining intensity was higher in intestinal versus diffuse types of gastric adenocarcinoma. cyclophilin A protein expression appeared to be lower in deeply invading glandular and cribriform structures of intestinal tumors, as well as in discretely placed groups of the intestinal tumor cells. Local relapses as well as distant metastases registered within 3 year follow up were observed to occur much less frequently in patients with positive cyclophilin A immunostaining in gastric tumors. Analysis of cyclophilin A expression has a potential value for prognosis of gastric adenocarcinoma recurrence and distant metastasis. C1 [Grigoryeva, S Evgeniya] Siberian Branch of RAMS, Cancer Research Institute, 5 Kooperativny Street, 634050 Tomsk, Russian Federation. [Cherdyntseva, V Nadezhda] Siberian Branch of RAMS, Cancer Research Institute, 5 Kooperativny Street, 634050 Tomsk, Russian Federation. [Karbyshev, S Mikhail] Siberian Branch of RAMS, Cancer Research Institute, 5 Kooperativny Street, 634050 Tomsk, Russian Federation. [Volkomorov, V Viktor] Siberian Branch of RAMS, Cancer Research Institute, 5 Kooperativny Street, 634050 Tomsk, Russian Federation. [Stepanov, IvanV] Siberian State Medical UniversityTomsk, Russian Federation. [Zavyalova, V Marina] Siberian State Medical UniversityTomsk, Russian Federation. [Perelmuter, M Vladimir] Siberian Branch of RAMS, Cancer Research Institute, 5 Kooperativny Street, 634050 Tomsk, Russian Federation. [Buldakov, A Mikhail] Siberian Branch of RAMS, Cancer Research Institute, 5 Kooperativny Street, 634050 Tomsk, Russian Federation. [Afanasjev, G Sergey] Siberian Branch of RAMS, Cancer Research Institute, 5 Kooperativny Street, 634050 Tomsk, Russian Federation. [Tuzikov, A Sergey] Siberian Branch of RAMS, Cancer Research Institute, 5 Kooperativny Street, 634050 Tomsk, Russian Federation. [Bukurova, A Yulia] Russian Academy of Sciences, Engelhardt Institute of Molecular BiologyMoscow, Russian Federation. [Lisitsyn, A Nikolai] Russian Academy of Sciences, Engelhardt Institute of Molecular BiologyMoscow, Russian Federation. [Beresten, F Sergey] Russian Academy of Sciences, Engelhardt Institute of Molecular BiologyMoscow, Russian Federation. RP Grigoryeva, SE (reprint author), Siberian Branch of RAMS, Cancer Research Institute, 634050 Tomsk, Russian Federation. EM grigorieva@oncology.tomsk.ru CR Jemal A, Center M, DeSantis C, Ward E, 2010, Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol Biomarkers Prev 19(8):1893–1907 Mizoue T, Yoshimura T, Tokui N, Hoshiyama Y, Yatsuya H, Sakata K, Kondo T, Kikuchi S, Toyoshima H, Hayakawa N, Tamakoshi A, Ohno Y, Fujino Y, Kaneko S, 2003, Prospective study of screening for stomach cancer in Japan. Japan Collaborative Cohort Study Group Int J Cancer 106(1):103–107 Washington K, 2010, 7th Edition of the AJCC Cancer Staging Manual: Stomach. Ann Surg Oncol 17:3077–3079 VolkomorovV, Grigoryeva E, KrasnovG, Litviakov N, Tsyganov M, KarbyshevM, Zavyalova M, Afanasуev S, Cherdyntseva N, Lisitsyn N, Beresten S, 2013, Search for potential gastric cancer markers using miRNA databases and gene expression analysis. Exp Oncol 35(1):2–7 Obchoei S, Weakley SM, Wongkham S, Wongkham C, Sawanyawisuth K, Yao Q, Chen C, 2011, Cyclophilin A enhances cell proliferation and tumor growth of liver fluke-associated cholangiocarcinoma. Mol Cancer 10:102 Nigro P, SatohK, O’DellM, SoeN, Cui Z,Mohan A,Abe J, Alexis J, Sparks J, Berk B, 2011, Cyclophilin A is an inflammatory mediator that promotes atherosclerosis in apolipoprotein E-deficient mice. J Exp Med 208(1):53–66 Lee J, 2010, Role of Cyclophilin A during Oncogenesis. Arch Pharm Res 33:181–187 Campa MJ, Wang MZ, Howard B, Fitzgerald MC, Patz EF, 2003, Protein expression profiling identifies macrophage migration inhibitory factor and cyclophilin A as potential molecular targets in nonsmall-cell lung cancer. Cancer Res 63:1652–1656 Cecconi D, Astner H, Donadelli M, Palmieri M, Missiaglia E, Hamdan M, Scarpa A, Righetti PG, 2003, Proteomic analysis of pancreatic ductal carcinoma cells treated with 5-aza-2′- deoxycytidine. Electrophoresis 24:4291–4303 Lim SO, Park SJ, KimW, Park SG, Kim HJ, Kim YI, Sohn TS, Noh JH, Jung G, 2002, Proteome analysis of hepatocellular carcinoma. Biochem Biophys Res Commun 291:1031–1037 Yang J, Li A, Yang Y, Li X, 2011, Identification of cyclophilin A as a potential prognostic factor for clear-cell renal cell carcinoma by comparative proteomic analysis. Cancer Biol Ther 11(5):535–546 Zheng J, Koblinski JE, Dutson LV, Feeney YB, Clevenger CV(2008, Prolyl isomerase cyclophilin A regulation of Janus-activated kinase 2 and the progression of human breast cancer. Cancer Res 68:7769– 7778 Hathout Y, Riordan K, Gehrmann M, Fenselau C, 2002, Differential protein expression in the cytosol fraction of an MCF-7 breast cancer cell line selected for resistance toward melphalan. J Proteome Res 1: 435–442 Krasnov GS, Oparina NY, Hankin SL, Mashkova TD, Ershov AN, Zatsepina OG, Karpov VL, Beresten SF, 2009, Identification of proteins with altered expression in colorectal cancer by means of 2D-proteomics. Mol Biol, Mosk, 43:321–328 Mortz E, Krogh T, Vorum H, Gоrg A, 2001, Improved silver staining protocols for high sensitivity protein identification using matrixassisted laser desorption/ionization-time of flight analysis. Proteomics 1:1359–1363 Stepanov IV, Zavizlova MV, Grigor’eva ES, Bukurova YA, Afanas’ev SG, Cherdyntseva NV, 2010, Clinico-morphological and genetics features of diffuse and intestinal gastric adenocarcinomas. Siberian journal of oncology 40:55–66, in Russian) El-Rifai W, Moskaluk CA, Abdrabbo MK, Harper J, Yoshida C, Riggins GJ, Frierson HF Jr, Powell SM, 2002, Gastric Cancers Over express S100A Calcium-binding Proteins. Cancer Research 62:6823–6826 Oien KA, Vass KJ, Downie I, Fullarton G, Keith NW, 2003, Profiling, comparison and validation of gene expression in gastric carcinoma and normal stomach. Oncogene 22:4287–4300 Ikeguchia M, Fukudaa K, Okaa S, Yamaguchia K, Hisamitsua K, Tsujitania S, Sakatanib T, Uedac T, Kaibaraa N, 2001, Clinicopathological Significance of Cathepsin D Expression in Gastric Adenocarcinoma. Oncology 61:71–78 Sribenja S, Li M, Wongkham S, Wongkham C, Yao Q, Chen C, 2009, Advances in thymosin beta10 research: differential expression, molecular mechanisms, and clinical implications in cancer and other conditions. Cancer Invest 27(10):1016–1022 Liaudet-Coopman E, Beaujouin M, Derocq D, Garcia M, Glondu- Lassis M, Laurent-Matha V, Prebois C, Rochefort H, Vignon F, 2006, Cathepsin D: newly discovered functions of a long-standing aspartic protease in cancer and apoptosis. Cancer Lett 237(2):167– 179 Salama I,Malone PS, Mihaimeed F, Jones JL, 2008, A review of the S100 proteins in cancer. Eur J Surg Oncol 34(4):357–364 Adachi Y, Yasuda K, Inomata M, Sato K, Shiraishi N, Kitano S, 2000, Pathology and prognosis of gastric carcinoma: well versus poorly differentiated type. Cancer 89:1418–1424 Kim JP, Lee JH, Kim SJ, Yu HJ, Yang HK, 1998, Clinicopathologic characteristics and prognostic factors in 10783 patients with gastric cancer. Gastric Cancer 1:125–133 Yoshihara T, Kadota Y, Yoshimura Y, Tatano Y, Takeuchi N, Okitsu H, Umemoto A, Yamauchi T, Itoh K, 2006, Proteomic alteration in gastic adenocarcinomas from Japanese patients.Molecular Cancer 5: 75 Li Z, Zhao X, Bai S, Wang Z, Chen L, Wei Y, Huang C, 2008, Proteomics Identification of Cyclophilin A as a Potential Prognostic Factor and Therapeutic Target in Endometrial Carcinoma. Mol Cell Proteomics 7:1810–1823 Grigoryeva E, Bukurova Y, Cherdyntseva N, Afanasуev S, Komarova T, Rodicheva N, Beresten S, 2009, 2Dproteomics of gastric cancer: identification of proteins with increased synthesis in tumor tissue. Siberian journal of oncology 5:37–42, in Russian) Ryu JW, Kim HJ, Lee YS, Myong NH, Hwang CH, Lee GS, Yom HC, 2003, The proteomics approach to find biomarkers in gastric cancer. J Korean Med Sci 18(4):505–509 Bai Z, Ye Y, Liang B, Xu F, Zhang H, Zhang Y, Peng J, Shen D, Cui Z, Zhang Z, Wang S, 2011, Proteomics-based identification of a group of apoptosis-related proteins and biomarkers in gastric cancer. Int J Oncol 38(2):375–383 Choi KJ, Piao YJ, Lim MJ, Kim JH, Ha J, Choe W, Kim SS, 2007, Overexpressed cyclophilin A in cancer cells renders resistance to hypoxia- and cisplatin-induced cell death. Cancer Res 67:3654–3662 Qi YJ, He QY, Ma YF, Du YW, Liu GC, Li YJ, Tsao GS, Ngai SM, Chiu JF, 2008, Proteomic identification ofmalignant transformationrelated proteins in esophageal squamous cell carcinoma. J Cell Biochem 104:1625–1635 Howard BA, Zheng Z, Campa MJ, Wang MZ, Shama A, Haura E, Herndon JE II, FitzgeraldMC, Bepler G, Patz EF, 2004, Translating biomarkers into clinical practice: prognostic implications of cyclophilin A and macrophage migratory inhibitory factor identified from protein expression profiles in non-small cell lung cancer. Lung Cancer 46:313–323 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 467 EP 473 DI 10.1007/s12253-013-9718-x PG 7 ER PT J AU Gou, X Chen, H Jin, F Wu, W Li, Y Long, J Gong, X Luo, M Bi, T Li, Z He, Q AF Gou, Xiaoxia Chen, Haixia Jin, Feng Wu, Weili Li, Yuanyuan Long, Jinhua Gong, Xiuyun Luo, Mengyalan Bi, Ting Li, Zhuolin He, Qianyong TI Expressions of CD147, MMP-2 and MMP-9 in Laryngeal Carcinoma and its Correlation with Poor Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Laryngeal carcinoma; Extracellularmatrix metalloproteinase induced factor; Matrix metalloproteinase-2; Matrix metalloproteinase-9; Immunohistochemistry ID Laryngeal carcinoma; Extracellularmatrix metalloproteinase induced factor; Matrix metalloproteinase-2; Matrix metalloproteinase-9; Immunohistochemistry AB The objectives of this study are to investigate the expressions of matrix metalloproteinase inducing factor (CD147), matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9) in laryngeal tumor tissues and its significant correlation with tumor infiltration, metastasis and prognosis. Laryngeal tumor tissue from 48 laryngeal cancer patients with complete clinical information was collected. The laryngitis tissue from 15 patients were collected as control group. Immunohistochemical analysis for CD147, MMP-2 and MMP-9 was performed for all the tissue. The results showed the expression rates of CD147, MMP-2 and MMP-9 in laryngeal carcinoma were 87.5 %, 75.0 % and 79.2 % respectively, significantly higher than those (26.7 %, 6.7 %, and 33.3 % respectively) in the control group are (P <0.01). High expression of CD147, MMP-2 and MMP-9 related to the clinical stages and lymphatic metastasis of laryngeal carcinoma. Univariate survival analysis showed that the 5-year survival of laryngeal carcinoma patients with low expression of CD147, MMP-2 and MMP-9was 83.3 %, 83.3 % and 90 % respectively, while the patients with high expression had 5-year survival at 25 %, 7.7 % and 18.2 % respectively (P <0.05). Multiple regression analysis showed that high expression of MMP-9 was independently associated with poor prognosis (P <0.05). High expression of CD147, MMP-2 and MMP-9 were related with laryngeal carcinoma invasion and metastasis. High expressions of CD147,MMP-2 and MMP-9 were all predictive factors of poor prognosis of laryngeal carcinoma. C1 [Gou, Xiaoxia] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. [Chen, Haixia] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. [Jin, Feng] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. [Wu, Weili] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. [Li, Yuanyuan] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. [Long, Jinhua] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. [Gong, Xiuyun] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. [Luo, Mengyalan] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. [Bi, Ting] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. [Li, Zhuolin] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. [He, Qianyong] The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. RP Jin, F (reprint author), The Affiliated Hospital of Guiyang Medical College, Guizhou Cancer Hospital, Department of Oncology, 550003 Guiyang, China. EM jinf8865@yeah.net CR Nabeshima K, Iwasaki H, Koga K, Hojo H, Suzumiya J, Kikuchi M, 2006, Emmprin, basigin/CD147):matrix metalloproteinase modulator and multifunctional cell recognition molecule that plays a critical role in cancer progression. Pathol Int 56:359–367 Tang Y, Kesavan P, Nakada MT, Yan L, 2004, Tumor-stroma interaction: positive feedback regulation of extracellular matrix metalloproteinase inducer, EMMPRIN, expression and matrix metalloproteinase-dependent generation of soluble EMMPRIN. Mol Cancer Res 2:73–80 Kato Y, Yamashita T, Ishikawa M, 2002, Relationship between expression of matrix metalloproteinase-2 and matrix metalloproteinase-9 and invasion ability of cervical cancer cells. Oncol Rep 9:565–569 Egeblad M, Werb Z, 2002, New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2:161–174 Jodele S, Blavier L, Yoon JM, DeClerk YA, 2006)Modifying the soil to affect the seed: Role of stromal-derived matrix metalloproteinases in cancer progression. Cancer Metastasis Rev 25:35–43 Li Y, Xu J, Chen L, Zhong WD, Zhang Z, Mi L, Zhang Y, Liao CG, Bian HJ, Jiang JL, Yang XM, Li XY, Fan CM, Zhu P, Fu L, Chen ZN, 2009, HAb18G, CD147), a cancer-associated biomarker and its role in cancer detection. Histopathology 54:677–687 Guo H, Li R, Zucker S, Toole BP, 2000, EMMPRIN, CD147), an inducer of matrix metalloproteinase synthesis, also binds interstitial collagenase to the tumor cell surface. Cancer Res 60:888–891 Riethdorf S, Reimers N, Assmann V, Kornfeld JW, Terracciano L, Sauter G, Pantel K, 2006, High incidence of EMMPRIN expression in human tumors. Int J Cancer 119:1800–1810 Noguchi Y, Sato T, Hirata M, Hara T, Ohama K, Ito A, 2003, Identification and characterization of extracellular matrix metalloproteinase inducer in humane ndometrium during the menstrual cycle in vivo and in vitro. J Clin Endocrinol Metab 88:6063–6072 Kanekura T, Chen X,Kanzaki T, 2002, Basigin, CD147)is expressed on melanoma cells and induces tumor cell invasion by stimulating production of matrix metalloproteinases by fibroblasts. Int J Cancer 99:520–528 Xu J, Shen ZY, Chen XG, Zhang Q, Bian HJ, Zhu P, Xu HY, Song F, Yang XM, Mi L, Zhao QC, Tian R, Feng Q, Zhang SH, Li Y, Jiang JL, Li L, Yu XL, Zhang Z, Chen ZN, 2007, A randomized controlled trial of Licartin for preventing hepatoma recurrence after liver transplantation. Hepatology 45:269–276 Su J, ChenX, Kanekura T, 2009)A CD147-targeting siRNA inhibits the proliferation, invasiveness, and VEGF production of human malignant melanoma cells by down regulating glycolysis.Cancer Lett 273:140–147 Kuang YH, Chen X, Su J, Wu LS, Li J, Chang J, Qiu Y, Chen ZS, Kanekura T, 2008, Proteome analysis of multidrug resistance of human oral squamous carcinoma cells using CD147 silencing. J Proteome Res 7:4784–4791 Jia L,WeiW, Cao J, Xu H, Miao J, Zhang J, 2009, Silencing CD147 inhibits tumor progression and increases chemosensitivity inmurine lymphoid neoplasm P388D1 cells. Ann Hematol 88(8):753–760 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 475 EP 481 DI 10.1007/s12253-013-9720-3 PG 7 ER PT J AU Kapoor, Sh AF Kapoor, Shailendra TI Tumor Growth Attenuating Effects of Naringenin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Kapoor, Shailendra] University of ChicagoChicago, IL, USA. RP Kapoor, Sh (reprint author), University of Chicago, Chicago, USA. EM shailendrakapoor@yahoo.com CR Sulfikkarali N, Krishnakumar N, Manoharan S, Nirmal RM, 2012, Chemopreventive efficacy of naringenin-loaded nanoparticles in 7,12- dimethylbenz(a)anthracene induced experimental oral carcinogenesis. Pathol Oncol Res 19:287–296 Sabarinathan D, Vanisree AJ, 2012, Plausible role of naringenin against cerebrally implanted C6 glioma cells in rats. Mol Cell Biochem 375:171–178 Sabarinathan D, Mahalakshmi P, Vanisree AJ, 2011, Naringenin, a flavanone inhibits the proliferation of cerebrally implanted C6 glioma cells in rats. Chem Biol Interact 189:26–36 Sabarinathan D, Mahalakshmi P, Vanisree AJ, 2010, Naringenin promote apoptosis in cerebrally implanted C6 glioma cells. Mol Cell Biochem 345:215–222 Ekambaram G, Rajendran P, Magesh V, Sakthisekaran D, 2008, Naringenin reduces tumor size and weight lost in N-methyl-N′-nitro- N-nitrosoguanidine-induced gastric carcinogenesis in rats. Nutr Res 28:106–112 Ekambaram G, Rajendran P, Devaraja R, Muthuvel R, Sakthisekaran D, 2008, Impact of naringenin on glycoprotein levels in N-methyl-N′- nitro-N-nitrosoguanidine-induced gastric carcinogenesis in rats. Anticancer Drugs 19:885–890 Subramanian P, Arul D, 2012, Attenuation of NDEA-induced hepatocarcinogenesis by naringenin in rats. Cell Biochem Funct 31: 511–517 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2014 VL 20 IS 2 BP 483 EP 483 DI 10.1007/s12253-013-9702-5 PG 1 ER PT J AU Al-Dasooqi, N Wardill, RH Gibson, JR AF Al-Dasooqi, Noor Wardill, R Hannah Gibson, J Rachel TI Gastrointestinal Mucositis: The Role of MMP-Tight Junction Interactions in Tissue Injury SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Matrix metalloproteinases; Tight junctions; Gut toxicity; Mucositis; Chemotherapy ID Matrix metalloproteinases; Tight junctions; Gut toxicity; Mucositis; Chemotherapy AB Chemotherapy for cancer causes significant gut toxicity known as mucositis. The pathogenesis of mucositis is ill defined. Recent clinical research guidelines have highlighted epithelial junctional complexes as emerging targets within mucositis research. Given the robust biological evidence linking tight junctions and matrix metalloproteinases, key mediators of mucositis, tight junction proteins have received significant attention. Despite this, the link between tight junctions, matrix metalloproteinases and mucositis development is yet to be established. This critical review therefore aims to describe the role of matrix metalloproteinases in mucositis, and how matrix metalloproteinase-dependent tight junction disruption may contribute to the pathobiology of mucositis. C1 [Al-Dasooqi, Noor] University of Adelaide, School of Medical Sciences, North Terrace, 5005 Adelaide, Australia. [Wardill, R Hannah] University of Adelaide, School of Medical SciencesAdelaide, Australia. [Gibson, J Rachel] University of Adelaide, School of Medical SciencesAdelaide, Australia. RP Al-Dasooqi, N (reprint author), University of Adelaide, School of Medical Sciences, 5005 Adelaide, Australia. EM noor.al-dasooqi@adelaide.edu.au CR Keefe D, SchubertMet al, 2007, Updated clinical practice guidelines for the prevention and treatment of mucositis. Cancer 109:820–831 Sonis S, 2004, The pathobiology of mucositis. Nat Rev Cancer 4: 277–284 Sonis S, Shklar Tet al, 1990, An animal model for mucositis induced by cancer chemotherapy. Oral Surg Oral Med Oral Pathol 69:437– 443 Paris F, Fuks Z et al, 2001, Endothelial apoptosis as the primary lesion initiating intestinal radiation damage in mice. Science 293: 293–297 Logan R, Stringer A et al, 2009, Is the pathobiology of chemotherapy-induced alimentary tract mucositis influenced by the type of mucotoxic drug administered? Cancer Chemother Pharmacol 63:239–251 Stringer A, Gibson R et al, 2009, Irinotecan-induced mucositis manifesting as diarrhoea corresponds with an amended intestinal flora and mucin profile. Int J Exp Pathol 90:489–499 Stringer A, Gibson R et al, 2009, Chemotherapy-induced changes to microflora: evidence and implications of change. Curr Drug Metab 10:79–83 Al-Dasooqi N, Bowen J et al, 2011, Irinotecan-induced alterations in intestinal cell kinetics and extracellular matrix component expression in the dark agouti rat. Int J Exp Pathol 92:357–365 Al-Dasooqi N, Gibson R et al, 2010, Matrix metalloproteinases are possible mediators for the development of alimentary tract mucositis in the DA rat. Exp Biol Med 235:1244–1256 Ma T, Iwamoto G et al, 2004, TNF-alpha-induced increase in intestinal epithelial tight junction permeability requires NF-kappa B activation. Am J Physiol Gastrointest Liver Physiol 286:G367–G376 Blijlevens NM, Donnelly JP, de Pauw BE, 2005, Prospective evaluation of gut mucosal barrier injury following various myeloablative regimens for haematopoietic stem cell transplant. Bone Marrow Transplant 35(7):707–711 Walsh-Reitz M, Huang E et al, 2005, AMP-18 protects barrier function of colonic epithelial cells: role of tight junction proteins. Am J Physiol Gastrointest Liver Physiol 289:G163–G171 Wardill H, Bowen J, Gibson R, 2013, Chemotherapy-induced gut toxicity: are alterations to intestinal tight junctions pivotal? Cancer Chemother Pharmacol 70:627–635 Hollander D, 1999, Intestinal permeability, leaky gut, and intestinal disorders. Curr Gastroenterol Rep 1:410–416 Gonzalez-Mariscal L, Tapia R, Chamorro D, 2008, Crosstalk of tight junction components with signaling pathways. Biochim Biophys Acta 1778(3):729–756 Keefe D, Cummins AG et al, 1997, Effect of high-dose chemotherapy on intestinal permeability in humans. Clin Sci 92:385–389 Keefe D, 2000, Chemotherapy for cancer causes apoptosis that precedes hypoplasia in crypts of the small intestine in humans. Gut 47:632–637 Hamada K, Shitara Yet al, 2010, Zonula Occluden-1 alterations and enhances intestinal permeability in methotrexate-treated rats. Cancer Chemother Pharmacol 66:1031–1038 Nakao T, Kurita N et al, 2012, Irinotecan injures tight junction and causes bacterial translocation in rat. J Surg Res 173(2):341–347 Youmba SB, Belmonte L et al, 2011, Methotrexate Modulates Tight Junctions Through NF-kappaB, MEK and JNK Pathways. J Pediatr Gastroenterol Nutr Stringer A, Al-Dasooqi N et al, 2013, Biomarkers of chemotherapyinduced diarrhoea: a clinical study of intestinal microbiome alterations, inflammation and circulating matrix metalloproteinases. Support Care Cancer. [Epub ahead of print] Sengupta N,MacDonald T, 2007, The role of matrix metalloproteinases in stromal/epithelial interactions in the gut. Physiology 22:401– 409 Clark I, Swingler T et al, 2008, The regulation of matrix metalloproteinases and their inhibitors. Int J Biochem Cell Biol 40:1362– 1378 Manicone A, McGuire J, 2008, Matrix metalloproteinases as modulators of inflammation. Semin Cell Dev Biol 19:34–41 Wolf M, Albrecht S, Marki C, 2008, Proteolytic processing of Chemokines: implications in physiological and pathological conditions. Int J Biochem Cell Biol 40:1185–1198 Klein T, Bischoff R, 2011, Physiology and pathophysiology of matrix metalloproteinases. Amino Acids 41:271–290 Chakraborti S, Mandal M et al, 2003, Regulation of matrix metalloproteinases: an overview. Mol Cell Biochem 253:269–285 Pender S, MacDonald T, 2004, Matrix metalloproteinases and the gut- new roles for old enzymes. Curr Opin Pharmacol 4:546–550 Van Wart H, Birkedal-Hansen H, 1990, The cysteine switch: a principle of regulation of metalloproteinase activity with potential applicability to the entire matrix metalloproteinase gene family. Proc Natl Acad Sci U S A 87:5578–5582 Dhaouadi T, Sfar I et al, 2007, Role of immune system, apoptosis and angiogenesis in pathogenesis of rheumatoid arthritis and joint destruction, a systematic review. Tunis Med 85:991–998 Sorsa T, Tjaderhane L et al, 2006, Matrix metalloproteinases: contribution to pathogenesis, diagnosis and treatment of periodontal inflammation. Ann Med 38:306–321 Vandenbroucke R, Dejonckheere E, and Libert C, 2011, A therapeutic role for MMP inhibitors in lung diseases? Eur Respir J [Epub ahead of print] Parks W, Wilson C, Lopez-Boado Y, 2004, Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol 4:617–629 Shipley J, Wesselschmidt R et al, 1996, Metalloelastase is required for macrophage-mediated proteolysis and matrix invasion in mice. Proc Natl Acad Sci 93:3942–3946 Warner R, Lewis C et al, 2001, The role of metalloelastase in immune complex-induced acute lung injury. Am J Pathol 158: 2139–2144 Lanone S, Zheng T et al, 2002, Overlapping and enzyme-specific contributions of matrix metalloproteinases-9 and -12 in IL-13- induced inflammation and remodeling. J Clin Invest 110:463–474 Itoh T,Matsuda H et al, 2002, The role ofmatrixmetalloproteinase-2 and matrix metalloproteinase-9 in antibody-induced arthritis. J Immunol 169:2643–2647 Mudgett J, Hutchinson N et al, 1998, Susceptability of stromelysin-1 deficient mice to collagen-induced arthritis and cartilage destruction. Arthritis Rheum 41:110–121 Corry D, Rishi K et al, 2002, Decreased allergic lung inflammatory cell egression and increased susceptability to asphyxiation inMMP2- deficiency. Nat Immunol 3:347–353 HartzellW, Shapiro S, 1999, Marcophage elastase prevents Gemella morbillorum infection and improves outcome following murine bone marrow transplantation. Chest 116:31S–32S Burke B, 2004, The role of matrix metalloproteinase 7 in innate immunity. Immunobiology 209:51–56 Page-McCaw A, Ewald A,Werb Z, 2007)Matrix metalloproteinases and the regulation of tissue remodelling. Nat Rev Mol Cell Biol 8: 221–233 Will C, Fromm M, Muller D, 2008, Claudin tight junction proteins: novel aspects in paracellular transport. Perit Dial Int J Int Soc Perit Dial 28(6):577–584 Anderson JM, Balda MS, Fanning AS, 1993, The structure and regulation of tight junctions. Curr Opin Cell Biol 5(5):772–778 Fanning AS, Anderson JM, 2009, Zonula occludens-1 and -2 are cytosolic scaffolds that regulate the assembly of cellular junctions. Ann N YAcad Sci 1165:113–120 Katsuno T, Umeda K et al, 2008, Deficiency of zonula occludens-1 causes embryonic lethal phenotype associated with defected yolk sac angiogenesis and apoptosis of embryonic cells. Mol Biol Cell 19(6): 2465–2475 Shin K, Margolis B, 2006, ZOning out tight junctions. Cell 126(4): 647–649 Umeda K, Ikenouchi J et al, 2006, ZO-1 and ZO-2 independently determine where claudins are polymerized in tight-junction strand formation. Cell 126(4):741–754 Bertiaux-Vandaele N, Youmba SB et al, 2011, The expression and the cellular distribution of the tight junction proteins are altered in irritable bowel syndrome patients with differences according to the disease subtype. Am J Gastroenterol 106(12):2165–2173 Schulzke JD, Ploeger S et al, 2009, Epithelial tight junctions in intestinal inflammation. Ann N YAcad Sci 1165:294–300 Turner J, 2009, Intestinal mucosal barrier function in health and disease. Nat Rev Immunol 9:799–809 Cummins PM, 2012, Occludin: one protein, many forms. Mol Cell Biol 32(2):242–250 Schulzke JD, Gitter AH et al, 2005, Epithelial transport and barrier function in occludin-deficient mice. Biochim Biophys Acta 1669(1): 34–42 Ulluwishewa D, Anderson RC et al, 2011, Regulation of tight junction permeability by intestinal bacteria and dietary components. J Nutr 141(5):769–776 Fazeny-Dorner B, Veitl M et al, 2002, Alterations in intestinal permeability following the intensified polydrug-chemotherapy IFADIC, ifosfamide, Adriamycin, dacarbazine). Cancer Chemother Pharmacol 49(4):294–298 Wardill H, Bowen J and Gibson R, 2013, Irinotecan disrupts tight junction protein occludin in the rat small intestine, in Multinational Association for Supprotive Care in Cancer. J Support Care Cancer. Berlin, Germany Bauer A, Burgers H et al, 2010)Matrix metalloproteinase-9 mediates hypoxia-induced vascular leakage in the brain via tight junction rearrangement. J Cereb Blood Flow Metab 30:837–848 Lischper M, Beuck S et al, 2010, Metalloproteinase mediated occludin cleavage in the cerebral microcapillary endothelium under pathological conditions. Brain Res 1326:114–127 Vermeer P, Denker J et al, 2009, MMP9 modulates tight junction integrity and cell viability in human airway epithelia. Am J Physiol Lung Cell Mol Physiol 296:L751–L762 Blecharz K, Haghikia A et al, 2010, Glucocorticoid effects on endothelial barrier function in the murine brain endothelial cell line cEND incubated with sera frompatients with multiple sclerosis.Mult Scler 16:293–302 Liu W, Hendren J et al, 2009, Normobaric hyperoxia attenuates early blood–brain barrier disruption by inhibiting MMP-9-mediated occludin degradation in focal cerebral ischemia. J Neurochem 108:811–820 Chen W, Hartman R, Ayer R, Marcantonio S, Kamper J, Tang J, Zhang JH, 2009, Matrix metalloproteinases inhibition provides neuroprotection against hypoxia-ischemia in the developing brain. J Neurochem 111:726–736 Higashida T, Kreipke CW, Rafols JA, Peng C, Schafer S, Schafer P, Ding JY, Dornbos D 3rd, Li X, Guthikonda M, Rossi NF, Ding Y, 2011, The role of hypoxia-inducible factor-1alpha, aquaporin-4 and matrix metalloproteinase-9 in blood–brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg 114:92– 101 Siu M, Lee W, Cheng C, 2003, The interplay of collagen IV, tumor necrosis factor-alpha, gelatinase B, matrixmetalloprotease-9), and tissue inhibitor of metalloproteases-1 in the basal lamina regulates Sertoli celltight junction dynamics in the rat testis. Endocrinology 144:371–387 Gorodeski G, 2007, Estrogen decrease in tight junction resistance involves matrix metalloproteinase-7-mediated remodeling of occludin. Endocrinology 148:218–231 Jeong S, Ledee D et al, 2012, Interaction of clusterin and matrix metalloproteinase-9 and its implication for epithelial homeostasis and inflammation. Am J Pathol 180:2028–2039 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 485 EP 491 DI 10.1007/s12253-013-9733-y PG 7 ER PT J AU Holczbauer, Gyongyosi, B Lotz, G Torzsok, P Kaposi-Novak, P Szijarto, A Tatrai, P Kupcsulik, P Schaff, Zs Kiss, A AF Holczbauer, Agnes Gyongyosi, Benedek Lotz, Gabor Torzsok, Peter Kaposi-Novak, Pal Szijarto, Attila Tatrai, Peter Kupcsulik, Peter Schaff, Zsuzsa Kiss, Andras TI Increased Expression of Claudin-1 and Claudin-7 in Liver Cirrhosis and Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocarcinogenesis; Hepatitis C virus; Cirrhosis; Tight junction; Claudin ID Hepatocarcinogenesis; Hepatitis C virus; Cirrhosis; Tight junction; Claudin AB Claudins have been reported to be differentially regulated in malignancies and implicated in the process of carcinogenesis and tumor progression. Claudin-1 has been described as key factor in the entry of hepatitis C virus (HCV) into hepatocytes and as promoter of epithelialmesenchymal transition in liver cells. The objective of the current study was to characterize claudin expression in hepatocellular carcinoma (HCC) as well as HCC-surrounding and normal liver samples with respect to cirrhosis and HCV-infection. Expression of claudin-1, -2, -3, -4, and −7 was measured by morphometric analysis of immunohistochemistry, and Western blotting in 30 HCCs with 30 corresponding nontumorous tissues and 6 normal livers. Claudin-1 and −7 protein expression was found significantly elevated in cirrhosis when compared with non-cirrhotic liver. HCCs developed in cirrhotic livers showed even higher expression of claudin-1 contrary to decreased claudin-7 expression when compared with cirrhosis. With reference to HCV status, HCCs or surrounding livers of HCV-infected samples did not show significant alterations in claudin expression when compared with HCV-negative specimens. Cirrhotic transformation associates with elevated claudin-1 and -7 expressions in both nontumorous liver and HCC. The fact that no significant differences in claudin expression were found regarding HCV-positivity in our sample set suggests that HCV-infection alone does not induce a major increase in the total amount of its entry co-factor claudin-1. Increased expression of claudin-1 seems to be a consequence of cirrhotic transformation and might contribute to a more effective HCV entry and malignant transformation. C1 [Holczbauer, Agnes] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Gyongyosi, Benedek] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Torzsok, Peter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Kaposi-Novak, Pal] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Szijarto, Attila] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Tatrai, Peter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Kupcsulik, Peter] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. RP Kiss, A (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM kiss.andras@med.semmelweis-univ.hu CR Ferenci P, Fried M, Labrecque D, Bruix J, Sherman M, Omata M, Heathcote J, Piratsivuth T, Kew M, Otegbayo JA, Zheng SS, Sarin S, Hamid SS, Modawi SB, Fleig W, Fedail S, Thomson A, Khan A, Malfertheiner P, Lau G, Carillo FJ, Krabshuis J, Le Mair A, 2010, Hepatocellular carcinoma, HCC): a global perspective. J Clin Gastroenterol 44(4):239–245., DOI 10.1097/MCG.0b013e3181d46ef2 Bosch FX, Ribes J, Diaz M, Cleries R, 2004, Primary liver cancer: worldwide incidence and trends. Gastroenterology 127(5 Suppl 1): S5–S16 Furuse M, Sasaki H, Fujimoto K, Tsukita S, 1998, A single gene product, claudin-1 or -2, reconstitutes tight junction strands and recruits occludin in fibroblasts. J Cell Biol 143(2):391–401 Furuse M, Sasaki H, Tsukita S, 1999, Manner of interaction of heterogeneous claudin species within and between tight junction strands. J Cell Biol 147(4):891–903 Morita K, Furuse M, Fujimoto K, Tsukita S, 1999, Claudin multigene family encoding four-transmembrane domain protein components of tight junction strands. Proc Natl Acad Sci U S A 96(2):511–516 Pileri P, Uematsu Y, Campagnoli S, Galli G, Falugi F, Petracca R, Weiner AJ, Houghton M, Rosa D, Grandi G, Abrignani S, 1998, Binding of hepatitis C virus to CD81. Science 282(5390):938–941 Scarselli E, Ansuini H, Cerino R, RoccaseccaRM, Acali S, Filocamo G, Traboni C, Nicosia A, Cortese R, Vitelli A, 2002, The human scavenger receptor class B type I is a novel candidate receptor for the hepatitis C virus. EMBO J 21(19):5017–5025 Evans MJ, von Hahn T, Tscherne DM, Syder AJ, Panis M, Wolk B, Hatziioannou T, McKeating JA, Bieniasz PD, Rice CM, 2007, Claudin-1 is a hepatitis C virus co-receptor required for a late step in entry. Nature 446(7137):801–805., DOI 10.1038/nature05654 Ploss A, Evans MJ, Gaysinskaya VA, Panis M, You H, de Jong YP, Rice CM, 2009, Human occludin is a hepatitis C virus entry factor required for infection of mouse cells. Nature 457(7231):882–886., DOI 10.1038/nature07684 Harris HJ, Davis C, Mullins JG, Hu K, Goodall M, Farquhar MJ, Mee CJ, McCaffrey K,Young S, Drummer H, Balfe P, McKeating JA, 2010, Claudin association with CD81 defines hepatitis C virus entry. J Biol Chem 285(27):21092–21102., DOI 10.1074/jbc.M110.104836 Krieger SE, Zeisel MB, Davis C, Thumann C, Harris HJ, Schnober EK, Mee C, Soulier E, Royer C, Lambotin M, Grunert F, Dao Thi VL, Dreux M, Cosset FL, McKeating JA, Schuster C, Baumert TF, 2010, Inhibition of hepatitis C virus infection by anti-claudin-1 antibodies is mediated by neutralization of E2-CD81-claudin-1 associations. Hepatology 51(4):1144–1157., DOI 10.1002/hep.23445 Fofana I, Krieger SE, Grunert F, Glauben S, Xiao F, Fafi-Kremer S, Soulier E, Royer C, Thumann C, Mee CJ, McKeating JA, Dragic T, Pessaux P, Stoll-Keller F, Schuster C, Thompson J, Baumert TF, 2010)Monoclonal anti-claudin 1 antibodies prevent hepatitis C virus infection of primary human hepatocytes. Gastroenterology 139, 3): 953–964, 964 e951-954., DOI 10.1053/j.gastro.2010.05.073 Mensa L, Crespo G, Gastinger MJ, Kabat J, Perez-del-Pulgar S, Miquel R, Emerson SU, Purcell RH, Forns X, 2011, Hepatitis C virus receptors claudin-1 and occludin after liver transplantation and influence on early viral kinetics. Hepatology 53(5):1436–1445., DOI 10.1002/hep.24110 Reynolds GM, Harris HJ, Jennings A, Hu K, Grove J, Lalor PF, Adams DH, Balfe P, Hubscher SG,McKeating JA, 2008, Hepatitis C virus receptor expression in normal and diseased liver tissue. Hepatology 47(2):418–427., DOI 10.1002/hep.22028 Holczbauer A, Gyongyosi B, Lotz G, Szijarto A, Kupcsulik P, Schaff Z, Kiss A, 2013, Distinct claudin expression profiles of hepatocellular carcinoma and metastatic colorectal and pancreatic carcinomas. J Histochem Cytochem: Off J Histochem Soc 61(4):294–305., DOI 10. 1369/0022155413479123 Yoon CH, Kim MJ, Park MJ, Park IC, Hwang SG, An S, Choi YH, Yoon G, Lee SJ, 2010, Claudin-1 acts through c-Abl-protein kinase Cdelta, PKCdelta, signaling and has a causal role in the acquisition of invasive capacity in human liver cells. J Biol Chem 285(1):226–233., DOI 10.1074/jbc.M109.054189 SuhY,Yoon CH,Kim RK, Lim EJ,OhYS,Hwang SG, An S,Yoon G, GyeMC, Yi JM, Kim MJ, Lee SJ, 2012, Claudin-1 induces epithelialmesenchymal transition through activation of the c-Abl-ERK signaling pathway in human liver cells. Oncogene 32:4873–4882., DOI 10.1038/ onc.2012.505 Ishak K, Baptista A, Bianchi L, Callea F, De Groote J, Gudat F, Denk H, Desmet V, Korb G, MacSween RN et al, 1995, Histological grading and staging of chronic hepatitis. J Hepatol 22(6):696–699 Lodi C, Szabo E, Holczbauer A, Batmunkh E, Szijarto A, Kupcsulik P, Kovalszky I, Paku S, Illyes G, Kiss A, Schaff Z, 2006, Claudin-4 differentiates biliary tract cancers from hepatocellular carcinomas. Mod Pathol: Off J U S Can Acad Pathol, Inc 19(3):460–469., DOI 10. 1038/modpathol.3800549 Grotegut S, von Schweinitz D, Christofori G, Lehembre F, 2006, Hepatocyte growth factor induces cell scattering through MAPK/Egr-1-mediated upregulation of Snail. EMBO J 25(15): 3534–3545., DOI 10.1038/sj.emboj.7601213 Kojima T, Takano K, Yamamoto T, Murata M, Son S, Imamura M, Yamaguchi H, Osanai M, Chiba H, Himi T, Sawada N, 2008, Transforming growth factor-beta induces epithelial to mesenchymal transition by down-regulation of claudin-1 expression and the fence function in adult rat hepatocytes. Liver Int: Off J Int Assoc Stud Liver 28(4):534–545., DOI 10.1111/j.1478-3231.2007.01631.x Takaki Y, Hirai S, Manabe N, Izumi Y, Hirose T, Nakaya M, Suzuki A, Mizuno K, Akimoto K, Tsukita S, Shuin T, Ohno S, 2001, Dynamic changes in protein components of the tight junction during liver regeneration. Cell Tissue Res 305(3):399–409 Kominsky SL, Argani P, Korz D, Evron E, Raman V, Garrett E, Rein A, Sauter G, Kallioniemi OP, Sukumar S, 2003, Loss of the tight junction protein claudin-7 correlates with histological grade in both ductal carcinoma in situ and invasive ductal carcinoma of the breast. Oncogene 22(13):2021–2033., DOI 10.1038/sj.onc.1206199 Tokes AM, Kulka J, Paku S, Mathe M, Paska C, Lodi C, Kiss A, Schaff Z, 2005, The expression of five different claudins in invasive breast carcinomas: comparison of pT1pN1 and pT1pN0 tumors. Pathol Res Pract 201(8–9):537–544 Michl P, Barth C, Buchholz M, Lerch MM, Rolke M, Holzmann KH, Menke A, Fensterer H, Giehl K, LohrM, Leder G, Iwamura T, Adler G, Gress TM, 2003, Claudin-4 expression decreases invasiveness and metastatic potential of pancreatic cancer. Cancer Res 63(19):6265–6271 Korompay A, Borka K, Lotz G, Somoracz A, Torzsok P, Erdelyi- Belle B, Kenessey I, Baranyai Z, Zsoldos F, Kupcsulik P, Bodoky G, Schaff Z, Kiss A, 2012, Tricellulin expression in normal and neoplastic human pancreas. Histopathology 60(6B):E76–E86., DOI 10. 1111/j.1365-2559.2012.04189.x Sobel G, Paska C, Szabo I, Kiss A, Kadar A, Schaff Z, 2005, Increased expression of claudins in cervical squamous intraepithelial neoplasia and invasive carcinoma. Hum Pathol 36(2):162–169., DOI 10.1016/j.humpath.2004.12.001 Schmelzer E, Wauthier E, Reid LM, 2006, The phenotypes of pluripotent human hepatic progenitors. Stem Cells 24(8):1852– 1858., DOI 10.1634/stemcells.2006-0036 Schmelzer E, Zhang L, Bruce A, Wauthier E, Ludlow J, Yao HL, Moss N, MelhemA, McClelland R, TurnerW, Kulik M, Sherwood S, Tallheden T, Cheng N, Furth ME, Reid LM, 2007, Human hepatic stem cells from fetal and postnatal donors. J Exp Med 204(8):1973– 1987., DOI 10.1084/jem.20061603 YovchevMI, Grozdanov PN, Joseph B, Gupta S, DabevaMD, 2007, Novel hepatic progenitor cell surface markers in the adult rat liver. Hepatology 45(1):139–149., DOI 10.1002/hep.21448 Gonzalez-Mariscal L, Lechuga S, Garay E, 2007, Role of tight junctions in cell proliferation and cancer. Prog Histochem Cytochem 42(1):1–57., DOI 10.1016/j.proghi.2007.01.001 Hirohashi S, Kanai Y, 2003, Cell adhesion system and human cancer morphogenesis. Cancer Sci 94(7):575–581 Lal-Nag M, Morin PJ, 2009, The claudins. Genome Biol 10(8):235., DOI 10.1186/gb-2009-10-8-235 Harris HJ, FarquharMJ, Mee CJ, Davis C, Reynolds GM, Jennings A, Hu K, Yuan F, Deng H, Hubscher SG, Han JH, Balfe P, McKeating JA, 2008, CD81 and claudin 1 coreceptor association: role in hepatitis C virus entry. J Virol 82(10):5007–5020., DOI 10.1128/JVI.02286-07 Orban E, Szabo E, Lotz G, Kupcsulik P, Paska C, Schaff Z, Kiss A, 2008, Different expression of occludin and ZO-1 in primary and metastatic liver tumors. Pathol Oncol Res POR 14(3):299–306., DOI 10.1007/s12253-008-9031-2 Schluter H,Wepf R,Moll I, FrankeWW(2004, Sealing the live part of the skin: the integrated meshwork of desmosomes, tight junctions and curvilinear ridge structures in the cells of the uppermost granular layer of the human epidermis. Eur J Cell Biol 83(11–12):655–665., DOI 10.1078/0171-9335-00434 Higashi Y, Suzuki S, Sakaguchi T, Nakamura T, Baba S, Reinecker HC, Nakamura S, Konno H, 2007, Loss of claudin-1 expression correlates with malignancy of hepatocellular carcinoma. J Surg Res 139(1):68–76., DOI 10.1016/j.jss.2006.08.038 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 493 EP 502 DI 10.1007/s12253-013-9683-4 PG 10 ER PT J AU Rekhi, B Vogel, U Basak, R Desai, BS Jambhekar, AN AF Rekhi, Bharat Vogel, Ulrich Basak, Ranjan Desai, B Sangeeta Jambhekar, A Nirmala TI Clinicopathological and Molecular Spectrum of Ewing Sarcomas/PNETs, Including Validation of EWSR1 Rearrangement by Conventional and Array FISH Technique in Certain Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ewing sarcoma; PNET; EWS-FLI1; EWSR1 rearrangement; FISH in soft tissue tumors; Molecular pathology of soft tissue sarcomas; Array FISH ID Ewing sarcoma; PNET; EWS-FLI1; EWSR1 rearrangement; FISH in soft tissue tumors; Molecular pathology of soft tissue sarcomas; Array FISH AB Over the years, a wide clinicopathological spectrum has been identified within Ewing family of tumors (EFTs). As these tumors are chemosensitive, their correct and timely identification is necessary. The aims of this study were (1) to present the diverse clinicopathological and molecular profile of EFTs in our settings, (2) to identify a pragmatic approach for diagnosing EFTs, especially for application of ancillary techniques, namely RT-PCR for specific transcripts (EWS-FLI1, EWS-ERG) and FISH for EWSR1 gene rearrangement, in certain cases and (3) to show the utility of tissue microarray in establishing a new FISH test. Fifty-eight EFTs were identified in 38 males and 20 females within an age-range of 1–65 years (median, 16),mostly in lower extremities (14) (24.1 %). Therapeutically, most patients underwent neoadjuvant chemotherapy with subsequent surgery. Histopathologically, diagnosis of EFTs was initially offered in 41/58 (70.6 %) tumors. On review, 59 % tumors showed diffuse pattern, while 41 % displayed rosettes. Immunohistochemically, tumor cells were mostly diffusely positive for CD99 (48/52) (92.3 %); FLI-1 (17/18) (94.4 %); variably for BCL2 (16/18) (88.8 %), synaptophysin (6/20) (35 %), S100-P (2/7) (28.5 %), CD56 (2/5) (40 %), NSE (2/5) (40 %), calponin (3/4) (75 %), EMA (5/24) (20.8 %) and CK (3/24) (12.5%), the latter two mostly focally. Fifty five tumors were EWS-FLI1 positive, while a single tumor was EWS-ERG positive. Sensitivity for PCR was 61 %. EWSR1 rearrangement was detected by FISH in 12/13 Ewing sarcomas/ PNETs. Sensitivity for EWSR1 test was 92.3 % and specificity was 100 %. Thirty-eight tumors, including 14 molecular confirmed EFTs and 21 other tumors were tested for EWSR1 rearrangement. Among 21 unrelated tumors, EWSR1 rearrangement was detected in fewmyoepithelial tumors, occasional desmoplastic small round cell tumor and an extraskeletal myxoid chondrosarcoma. Further, a tissue microarray with a separate set of 8 EFTs, confirmed at another laboratory was analysed for validation of EWSR1 rearrangement test. 23/28 (82.1 %) tissue cores of the tissue microarray, stained by FISH were interpretable, including EWSR1 rearrangement, detected in 20/28 tissue cores; not detected in 3 liver cores and uninterpretable in 5 (17.8 %) cores. Classical EFTs can be diagnosed with diffuse, membranous CD99 positivity, intranuclear FLI1 positivity and LCA negativity in malignant round cells. In unconventional cases, it is indispensable to reveal the concomitant fusion m-RNA by RT-PCR. In case of negative molecular results, it is necessary to prove EWSR1 rearrangement by FISH. These tests should be interpreted with clinicopathological correlation. Tissue microarrays for FISH are useful during validation of a new test, especially when sarcomas like EFTs show less genetic heterogeneity within tumor cells. C1 [Rekhi, Bharat] Tata Memorial Hospital, Department of Pathology, ParelMumbai, Maharashtra, India. [Vogel, Ulrich] Eberhard-Karls-University, University Hospital Tuebingen, Institute of Pathology, Leibermeisterstrasse 8, 72076 Tuebingen, Germany. [Basak, Ranjan] Tata Memorial Hospital, Department of Pathology, ParelMumbai, Maharashtra, India. [Desai, B Sangeeta] Tata Memorial Hospital, Department of Pathology, ParelMumbai, Maharashtra, India. [Jambhekar, A Nirmala] Tata Memorial Hospital, Department of Pathology, ParelMumbai, Maharashtra, India. RP Rekhi, B (reprint author), Tata Memorial Hospital, Department of Pathology, Mumbai, India. EM rekhi.bharat@gmail.com CR Delattre O, Zucman J, Melot T, Garau XS, Zucker JM, Lenoir GM, Ambros PF, Sheer D, Turc-Carel C, Triche TJ, Aurias A, Thomas G, 1994, The Ewing family of tumors–a subgroup of small-round-cell tumors defined by specific chimeric transcripts. N Engl J Med 331: 294–299 Dome JS, Rodriguez-Galindo C, Spunt SL, Santana VM, 2008, Pediatric solid tumors: Ewing’s sarcoma family tumors. In: Abeloff MD, Armitage JO, Niederhuber JE, KastanMB,McKennaWG(eds, Abeloff’s clinical oncology, 4th edn. Elsevier, Philadelphia, pp 2085– 2091 Khoury JD, 2005, Ewing sarcoma family of tumors. Adv Anat Pathol 12:212–220 Ushigome S, Machinami R, Sorensen PH, 2002, Ewing sarcoma/ Primitive neuroectodermal tumor. In: Fletcher CDM, Unni K, Mertens F, eds, Tumors of soft tissue and bone. Pathology and genetics. World Health Organization classification of tumors. IARC Press, Lyon, pp 298–300 Aurias A, Rimbaut C, Buffe D, Dubousset J, Mazabraud A, 1983, Translocation of chromosome 22 in Ewing’s sarcoma. C R Seances Acad Sci III 296:1105–1107 Aurias A, Rimbaut C, Buffe D, Zucker JM, Mazabraud A, 1984, Translocation involving chromosome 22 in Ewing’s sarcoma. A cytogenetic study of four fresh tumors. Cancer Genet Cytogenet 12: 21–25 Zucman J, Melot T, Desmaze C, Ghysdael J, Plougastel B, Peter M, Zucker JM, Triche TJ, Sheer D, Turc-Carel C, Ambros P, Combaret V, Lenoir G, Aurias A, Thomas G, Delattre O, 1993, Combinatorial generation of variable fusion proteins in the Ewing family of tumors. EMBO J 12:4481–4487 Urano F, Umezawa A, Yabe H, Hong W, Yoshida K, Fujinaga K, Hata J, 1998)Molecular analysis of Ewing’s sarcoma: another fusion gene, EWS-E1AF, available for diagnosis. Jpn J Cancer Res 89:703– 711 Shing DC, McMullan DJ, Roberts P, Smith K, Chin SF, Nicholson J, Tillman RM, Ramani P, Cullinane C, Coleman N, 2003, FUS/ERG gene fusions in Ewing’s tumors. Cancer Res 63:4568–4576 Barr FG, Womer RB, 2007, Molecular diagnosis of Ewing family tumors: too many fusions… ? J Mol Diagn 9:437–440 de Alava E, Kawai A, Healey JH, Fligman I, Meyers P, Huvos AG, GeraldWL, Jhanwar SC, Argani P,Antonescu CR, Pardo-Mindan FJ, Ginsberg J, Womer R, Lawlor ER, Wunder J, Andrulis I, Sorensen PHB, Barr FG, LadanyiM, 1998, EWS-FLI1 fusion transcript structure is an independent determinant of prognosis in Ewing’s sarcoma. J Clin Oncol 16:1248–1255 Hauben E, van den Broek LC, VanMarck E, Hogendoorn PC, 2001, Adamantinoma-like Ewing’s sarcoma and Ewing’s-like adamantinoma. The t, 11; 22), t, 21; 22, status. J Pathol 195:218–221 Folpe AL, Goldblum JR, Rubin BP, Shehata BM, LiuW, Dei TosAP, Weiss SW, 2005, Morphologic and immunophenotypic diversity in Ewing family tumors: a study of 66 genetically confirmed cases. Am J Surg Pathol 29:1025–1033 Schuetz AN, Rubin BP, Goldblum JR, Shehata B,Weiss SW, LiuW, Wick MR, Folpe AL, 2005, Intercellular junctions in Ewing sarcoma/primitive neuroectodermal tumor: additional evidence of epithelial differentiation. Mod Pathol 18:1403–1410 Llombart-Bosch A, Machado I, Navarro S, Bertoni F, Bacchini P, Alberghini M, Karzeladze A, SavelovN, Petrov S,Alvarado-Cabrero I, Mihaila D, Terrier P, Lopez-Guerrero JA, Picci P, 2009, Histological heterogeneity of Ewing’s sarcoma/PNET: an immunohistochemical analysis of 415 genetically confirmed cases with clinical support. Virchows Arch 455:397–411 Ishii N, Hiraga H, Sawamura Y, Shinohe Y, Nagashima K, 2001, Alternative EWS-FLI1 fusion gene and MIC2 expression in peripheral and central primitive neuroectodermal tumors. Neuropathology 21:40–44 Jimenez RE, Folpe AL, Lapham RL, Ro JY, O’Shea PA, Weiss SW, Amin MB, 2002, Primary Ewing’s sarcoma/primitive neuroectodermal tumor of the Kidney. A clinicopathologic and immunohistochemical analysis of 11 cases. Am J Surg Pathol 26:320– 327 Karpate A, Menon S, Basak R, Yuvaraja TB, Tongaonkar HB, Desai SB, 2012, Ewing sarcoma/primitive neuroectodermal tumor of the kidney: clinicopathologic analysis of 34 cases. Ann Diagn Pathol 16: 267–274 Gardner LJ, Ayala AG, Monforte HL, Dunphy CH, 2004, Ewing sarcoma/peripheral primitive neuroectodermal tumor: adult abdominal tumors with an Ewing sarcoma gene rearrangement demonstrated by fluorescence in situ hybridization in paraffin sections. Appl Immunohistochem Mol Morphol 12:160–165 Rekhi B, Qureshi S, Basak R, Desai SB, Medhi S, Kurkure P, Menon S, Maheshwari A, Jambhekar NA, 2010, Primary vaginal Ewing’s sarcoma or primitive neuroectodermal tumor in a 17-year-old woman: a case report. J Med Case Rep 4:88 Ostwal V, Rekhi B, Noronha V, Basak R, Desai SB, Maheshwari A, Prabhash K, 2012, Primitive neuroectodermal tumor of ovary in a young lady, confirmed with molecular and cytogenetic resultsa rare case report with a diagnostic and therapeutic challenge. Pathol Oncol Res 18:1104–1106. Meier VS, Kuhne T, Jundt G, Gudat F, 1998)Molecular diagnosis of Ewing tumors: improved detection of EWS-FLI-1 and EWS-ERG chimeric transcripts and rapid determination of exon combinations. Diagn Mol Pathol 7:29–35 Yamaguchi U, Hasegawa T, Morimoto Y, Tateishi U, Endo M, Nakatani F, Kawai A, Chuman H, Beppu Y, Endo M, Kurotaki H, Furuta K, 2005, A practical approach to the clinical diagnosis of Ewing’s sarcoma/primitive neuroectodermal tumor and other small round cell tumors sharing EWS rearrangement using new fluorescence in situ hybridisation probes for EWSR1 on formalin fixed, paraffin wax embedded tissue. J Clin Pathol 58:1051–1056 Bridge RS, Rajaram V, Dehner LP, Pfeifer JD, Perry A, 2006, Molecular diagnosis of Ewing sarcoma/primitive neuroectodermal tumor in routinely processed tissue: a comparison of two FISH strategies and RT-PCR in malignant round cell tumors. Mod Pathol 19:1–8 Jambhekar NA, Bagwan IN, Ghule P, Shet TM, Chinoy RF, Agarwal S, Joshi R, Amare Kadam PS, 2006, Comparative analysis of routine histology, immunohistochemistry, reverse transcriptase polymerase chain reaction, and fluorescence in situ hybridization in diagnosis of Ewing family of tumors. Arch Pathol Lab Med 130:1813–1818 Liu BY, Yang Y, Du J, Zhang Y,Wang H, Zheng J, 2008, Application of the in situ hybridization with EWS dual-color break-apart fluorescence probe and anti-CD99 and anti-FLI-1 antibodies in the diagnosis of Ewing’s sarcoma/primitive neuroectodermal tumor. Beijing Da Xue Xue Bao 40:358–362 Gamberi G, Cocchi S, Benini S,MagagnoliG,Morandi L,Kreshak J, Gambarotti M, Picci P, Zanella L, Alberghini M, 2011, Molecular diagnosis in Ewing family tumors: the Rizzoli experience–222 consecutive cases in four years. J Mol Diagn 13:313–324 Vural C, Uluoglu O, Akyurek N, Oguz A, Karadeniz C, 2011, The evaluation of CD99 immunoreactivity and EWS/FLI1 translocation by fluorescence in situ hybridization in central PNETs and Ewing’s sarcoma family of tumors. Pathol Oncol Res 17:619–625 Qian X, Jin L, Shearer BM, Ketterling RP, Jalal SM, Lloyd RV, 2005, Molecular diagnosis of Ewing’s sarcoma/primitive neuroectodermal tumor in formalin-fixed paraffin-embedded tissues by RT-PCR and fluorescence in situ hybridization. DiagnMol Pathol 14:23–28 Bridge JA, Cushman-Vokoun AM, 2011, Molecular diagnostics of soft tissue tumors. Arch Pathol Lab Med 135:588–601 Romeo S, Dei Tos AP, 2010, Soft tissue tumors associated with EWSR1 translocation. Virchows Arch 456:219–234 Sugimura H, Mori H, Nagura K, Kiyose S, Tao H, Isozaki M, Igarashi H, Shinmura K, Hasegawa A, Kitayama Y, Tanioka F, 2010, Fluorescence in situ hybridization analysis with a tissue microarray: ‘FISH and chips’ analysis of pathology archives. Pathol Int 60:543–550 Chen H, McClain D, Jhawar SC, Agaram NP, Hameed M, 2012, Complex interphase fluorescent in situ hybridization patterns of EWSR1 gene in Ewing sarcoma using break apart probes. Abstract 28. USCAP Kojima T, Asami S, Chin M, Yoshida Y, Mugishima H, Suzuki T, 2002, Detection of chimeric genes in Ewing’s sarcoma and its clinical applications. Biol Pharm Bull 25:991–994 Parija T, Shirley S, Uma S, Rajalekshmy KR, Ayyappan S, Rajkumar T, 2005, Type 1, 11; 22,, q24:q12, translocation is common in Ewing’s sarcoma/peripheral neuroectodermal tumor in south Indian patients. J Biosci 303:371–376 Chen N, Zhou Q, 2005, Constructing tissue microarrays without prefabricating recipient blocks. A novel approach. Am J Clin Pathol 124:103–107 Machado I, Noguera R, Pellin A, Lopez-Guerrero JA, Piqueras M, Navarro S, Llombart-Bosch A, 2009, Molecular diagnosis of ewing sarcoma family of tumors: a comparative analysis of 560 cases with FISH and RT-PCR. Diagn Mol Pathol 18:189–19938 Perlman EJ, Dickman PS, Askin FB, Grier HE, Miser JS, Link MP, 1994, Ewing’s sarcoma–routine diagnostic utilization ofMIC2 analysis: a Pediatric Oncology Group/Children’s Cancer Group Intergroup Study. Hum Pathol 25:304–307 Folpe AL, Hill CE, Parham DM, O’Shea PA, Weiss SW, 2000, Immunohistochemical detection of FLI-1 protein expression: a study of 132 round cell tumors with emphasis on CD99-positive mimics of Ewing’s sarcoma/primitive neuroectodermal tumor. Am J Surg Pathol 24:1657–1662 Kavalar R, Pohar Marinsek Z, Jereb B, Cagran B, Golouh R, 2009, Prognostic value of immunohistochemistry in the Ewing’s sarcoma family of tumors. Med Sci Monit 15:CR442–CR452 Mhawech-Fauceglia P, Herrmann F, Penetrante R, Beck A, Sait S, Block AM, Odunsi K, Fisher J, Balos L, Cheney RT, 2006, Diagnostic utility of FLI-1 monoclonal antibody and dual-colour, break-apart probe fluorescence in situ, FISH, analysis in Ewing’s sarcoma/primitive neuroectodermal tumor, EWS/PNET). A comparative study with CD99 and FLI-1 polyclonal antibodies. Histopathology 49:569–575 Llombart-Bosch A, Navarro S, 2001, Immunohistochemical detection of EWS and FLI-1 proteinss in Ewing sarcoma and primitive neuroectodermal tumors: comparative analysis with CD99, MIC-2, expression. Appl Immunohistochem Mol Morphol 9: 255–260 Downing JR, Head DR, Parham DM, Douglass EC, Hulshof MG, Link MP, Motroni TA, Grier HE, Curcio-Brint AM, Shapiro DN, 1993, Detection of the, 11;22)(q24;q12, translocation of Ewing’s sarcoma and peripheral neuroectodermal tumor by reverse transcription polymerase chain reaction. Am J Pathol 143:1294–1300 Sorensen PH, Liu XF, Delattre O, Rowland JM, Biggs CA, Thomas G, Triche TJ, 1993, Reverse transcriptase PCR amplification of EWS/FLI-1 fusion transcripts as a diagnostic test for peripheral primitive neuroectodermal tumors of childhood. Diagn Mol Pathol 2:147–157 Dockhorn-Dworniczak B, Schafer KL, Dantcheva R, Blasius S, Winkelmann W, Strehl S, Burdach S, van Valen F, Jurgens H, BockerW(1994, Diagnostic value of themolecular genetic detection of the t(11;22, translocation in Ewing’s tumors. Virchows Arch 425: 107–112 Guillou L, 2008, Contribution of molecular biology and markers to the prognosis and management of patients with soft tissue sarcoma. Pathol Case Rev 13:69–77 Kumar R, Rekhi B, Shirazi N, PaisA, Amare P, Gawde D, Jambhekar N, 2008, Spectrum of cytomorphological features, including literature review, of an extraskeletal myxoid chondrosarcoma with t(9;22)(q22;q12,, TEC/EWS, results in one case. Diagn Cytopathol 36:868–875 Bridge JA, 2008, Contribution of cytogenetics to the management of poorly differentiated sarcomas. Ultrastruct Pathol 32:63–71 Diaz LK, Gupta R, Kidwai N, Sneige N,Wiley EL, 2004, The use of TMA for interlaboratory validation of FISH testing for detection of HER2 gene amplification in breast cancer. J Histochem Cytochem 52:501–507 de Alava E, Antonescu CR, Panizo A, Leung D, Meyers PA, Huvos AG, Pardo-Mindan FJ, Healey JH, Ladanyi M, 2000, Prognostic impact of P53 status in Ewing sarcoma. Cancer 89:783–792 Huang HY, Illei PB, Zhao Z, Mazumdar M, Huvos AG, Healey JH, Wexler LH, Gorlick R,Meyers P, LadanyiM(2005, Ewing sarcomas with p53 mutation or p16/p14ARF homozygous deletion: a highly lethal subset associated with poor chemoresponse. J Clin Oncol 23: 548–558 Balamuth NJ,Womer RB, 2010, Ewing’s sarcoma. Lancet Oncol 11: 184–1192 Pierron G, Tirode F, Lucchesi C, Reynaud S, Ballet S, Cohen-Gogo S, PerrinV, Coindre JM,Delattre O, 2012, A new subtype of bone sarcoma defined by BCOR-CCNB3 gene fusion. Nat Genet 44:461–466 Italiano A, Sung YS, Zhang L, Singer S, Maki RG, Coindre JM, Antonescu CR, 2012, High prevalence of CIC fusion with doublehomeobox, DUX4, transcription factors in EWSR1-negative undifferentiated small blue round cell sarcomas. Genes Chromosome Cancer 51:207–218 Ladanyi M, Bridge JA, 2000, Contribution of molecular genetic data to the classification of sarcomas. Hum Pathol 31: 532–538 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 503 EP 516 DI 10.1007/s12253-013-9721-2 PG 14 ER PT J AU Choi, JY Yoo, JN Lee, HS AF Choi, Jin Youn Yoo, Jin Nam Lee, Hyung Sug TI Down-regulation of ROBO2 Expression in Prostate Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ROBO2; Prostate cancer; Expression ID ROBO2; Prostate cancer; Expression AB Several lines of evidence exist that axon guidance genes are involved in cancer pathogenesis. Axon guidance genes ROBO1 and ROBO2 are candidate tumor suppressor genes (TSG). The aim of our study was to address whether ROBO1 and ROBO2 expressions are altered in prostate cancers (PCA). In this study, we analyzed ROBO1 and ROBO2 expressions in 107 PCAs. In the immunohistochemistry, loss of ROBO2 expression was identified in 66 % of PCAs and was significantly higher than that in normal cells (p< 0.001). By contrast, there was no significant difference of ROBO1 expression between normal and PCAs. Our results indicate that axon guidance protein ROBO2 is frequently lost in PCA and that ROBO2 might be involved in PCA pathogenesis as a candidate TSG. C1 [Choi, Jin Youn] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Topczewska JM, Postovit LM, Margaryan NV, Sam A, Hess AR, Wheaton WW, Nickoloff BJ, Topczewski J, Hendrix MJ, 2006, Embryonic and tumorigenic pathways converge via Nodal signaling: role in melanoma aggressiveness. Nat Med 12:925–932 Mehlen P, Delloye-Bourgeois C, Chedotal A, 2011, Novel roles for Slits and netrins: axon guidance cues as anticancer targets. Nat Rev Cancer 11:188–197 Tseng RC, Lee SH, Hsu HS, Chen BH, TsaiWC, Tzao C,Wang YC, 2010, SLIT2 attenuation during lung cancer progression deregulates beta-catenin and E-cadherin and associates with poor prognosis. Cancer Res 70:543–551 Prasad A, Paruchuri V, Preet A, Latif F, Ganju RK, 2008, Slit-2 induces a tumor-suppressive effect by regulating beta-catenin in breast cancer cells. J Biol Chem 283:26624–26633 Xian J, Clark KJ, Fordham R, Pannell R, Rabbitts TH, Rabbitts PH, 2001, Inadequate lung development and bronchial hyperplasia in mice with a targeted deletion in the Dutt1/Robo1 gene. Proc Natl Acad Sci U S A 98:15062–15066 Mitra S, Mazumder-Indra D, Mondal RK, Basu PS, Roy A, Roychoudhury S, Panda CK, 2012, Inactivation of SLIT2- ROBO1/2 pathway in premalignant lesions of uterine cervix: clinical and prognostic significances. PLoS One 7:e38342 Sundaresan V, Chung G, Heppell-Parton A, Xiong J, Grundy C, Roberts I, James L, Cahn A, Bench A, Douglas J, Minna J, Sekido Y, Lerman M, Latif F, Bergh J, Li H, Lowe N, Ogilvie D, Rabbitts P, 1998, Homozygous deletions at 3p12 in breast and lung cancer. Oncogene 17:1723–1729 Dallol A, Forgacs E, Martinez A, Sekido Y, Walker R, Kishida T, Rabbitts P, Maher ER, Minna JD, Latif F, 2002, Tumour specific promoter region methylation of the human homologue of the Drosophila Roundabout gene DUTT1, ROBO1, in human cancers. Oncogene 21:3020–3028 Ghosh S, Ghosh A, Maiti GP, Alam N, Roy A, Roychoudhury S, Panda CK(2009, Alterations of ROBO1/DUTT1 andROBO2 loci in early dysplastic lesions of head and neck: clinical and prognostic implications. Hum Genet 125:189–198 Latil A, Chene L, Cochant-Priollet B, Mangin P, Fournier G, Berthon P, Cussenot O, 2003, Quantification of expression of netrins, slits and their receptors in human prostate tumors. Int J Cancer 103:306–315 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Kim MS, Hur SY, Yoo NJ, Lee SH, 2010, Mutational analysis of FOXL2 codon 134 in granulosa cell tumour of ovary and other human cancers. J Pathol 221:147–152 Kim MS, Oh JE, Kim YR, Park SW, Kang MR, Kim SS, Ahn CH, Yoo NJ, Lee SH, 2010, Somatic mutations and losses of expression of microRNA regulation-related genes AGO2 and TNRC6A in gastric and colorectal cancers. J Pathol 221:139–146 Biankin AV, Waddell N, Kassahn KS et al, 2012, Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491:399–405 Ong CK, Subimerb C, Pairojkul C et al, 2012, Exome sequencing of liver fluke-associated cholangiocarcinoma. Nat Genet 44:690–693 Berger MF, Lawrence MS, Demichelis F et al, 2011, The genomic complexity of primary human prostate cancer. Nature 470:214–220 Barbieri CE, Baca SC, LawrenceMS et al, 2012, Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet 44:685–689 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 517 EP 519 DI 10.1007/s12253-013-9722-1 PG 3 ER PT J AU Seki, S Fujiwara, M Matsuura, M Fujita, Sh Ikeda, H Umeda, M Asahina, I Ikeda, T AF Seki, Sachiko Fujiwara, Mutsunori Matsuura, Masaaki Fujita, Shuichi Ikeda, Hisazumi Umeda, Masahiro Asahina, Izumi Ikeda, Tohru TI Prognostic Value of Podoplanin Expression in Oral Squamous Cell Carcinoma―A Regression Model Auxiliary to UICC Classification SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Podoplanin; Oral squamous cell carcinoma; Prognosis; Clinical stage; Statistical analysis ID Podoplanin; Oral squamous cell carcinoma; Prognosis; Clinical stage; Statistical analysis AB Podoplanin, a type I transmembrane glycoprotein with an effect of platelet aggregation, has been reported to be one of the possible prognostic factors of oral squamous cell carcinoma (OSCC). However, the biological significance of podoplanin is largely unclear. The aim of this study was to develop a practicalmodel for the prediction of prognosis using the grade of podoplanin expression, and also to evaluate the biological function of podoplanin. Eighty-two specimens of patients with previously untreated OSCC, who underwent either biopsy or surgery, were histopathologically and immunohistochemically analyzed. These 82 cases were composed of 66 well-differentiated, 10 moderately differentiated and 6 poorly differentiated OSCC. Podoplanin was successfully immunostained in 78 specimens, and was detected in most cases, but the frequency of positive cells varied. The prognosis of patients with more than 50 % podoplaninpositive tumor cells was significantly poorer than that of the other patients. Multivariate hazards regression analysis suggested that a linear combination of covariates, OSCC patients with more or less than 50 % podoplanin expression, age of more or less than 70 years old,mode of invasion and T3, T4 or T2 versus T1 of the UICC T-stage classification was the most effective model for evaluating the prognosis of OSCC patients. Additionally, podoplanin expression had a significant relationship to UICC clinical stage and the expression of Ki- 67. An effective regression model using podoplanin expression was developed for evaluating the prognosis of OSCC and the biological significance of podoplanin was suggested to be associated with the growth and/or progression of OSCC. C1 [Seki, Sachiko] Nagasaki University Graduate School of Biomedical Sciences, Department of Oral Pathology and Bone MetabolismNagasaki, Japan. [Fujiwara, Mutsunori] Japanese Red Cross Medical Center, Department of Clinical PathologyTokyo, Japan. [Matsuura, Masaaki] Japanese Foundation for Cancer Research, Genome Center of JFCR, and Division of Cancer Genomics, Cancer Institute of JFCR, Bioinformatics GroupTokyo, Japan. [Fujita, Shuichi] Nagasaki University Graduate School of Biomedical Sciences, Department of Oral Pathology and Bone MetabolismNagasaki, Japan. [Ikeda, Hisazumi] Nagasaki University Graduate School of Biomedical Sciences, Department of Regenerative Oral SurgeryNagasaki, Japan. [Umeda, Masahiro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral OncologyNagasaki, Japan. [Asahina, Izumi] Nagasaki University Graduate School of Biomedical Sciences, Department of Regenerative Oral SurgeryNagasaki, Japan. [Ikeda, Tohru] Nagasaki University Graduate School of Biomedical Sciences, Department of Oral Pathology and Bone MetabolismNagasaki, Japan. RP Ikeda, T (reprint author), Nagasaki University Graduate School of Biomedical Sciences, Department of Oral Pathology and Bone Metabolism, Nagasaki, Japan. EM tohrupth@nagasaki-u.ac.jp CR Toyoshima M, Nakajima M, Yamori T, Tsuruo T, 1995, Purification and characterization of the platelet-aggregating sialoglycoprotein gp44 expressed by highly metastatic variant cells of mouse colon adenocarcinoma 26. Cancer Res 55(4):767–773 Kato Y, Fujita N, Kunita A, Sato S, Kaneko M, Osawa M, Tsuruo T, 2003, Molecular identification of Aggrus/T1alpha as a platelet aggregation-inducing factor expressed in colorectal tumors. J Biol Chem 278(51):51599–51605., DOI 10.1074/jbc.M309935200 Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E, Diem K, Weninger W, Tschachler E, Alitalo K, Kerjaschki D, 1999, Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: podoplanin as a specific marker for lymphatic endothelium. Am J Pathol 154(2):385–394., DOI 10.1016/s0002-9440(10)65285-6 Schacht V, Ramirez MI, Hong YK, Hirakawa S, Feng D, Harvey N, Williams M, Dvorak AM, Dvorak HF, Oliver G, Detmar M, 2003, T1alpha/podoplanin deficiency disrupts normal lymphatic vasculature formation and causes lymphedema. EMBO J 22(14):3546–3556., DOI 10.1093/emboj/cdg342 Martin-Villar E, Scholl FG, Gamallo C, Yurrita MM, Munoz-Guerra M, Cruces J, Quintanilla M, 2005, Characterization of human PA2.26 antigen, T1alpha-2, podoplanin), a small membrane mucin induced in oral squamous cell carcinomas. Int J Cancer 113(6):899– 910., DOI 10.1002/ijc.20656 Yuan P, Temam S, El-Naggar A, Zhou X, Liu DD, Lee JJ, Mao L, 2006, Overexpression of podoplanin in oral cancer and its associationwith poor clinical outcome. Cancer 107(3):563–569., DOI 10.1002/cncr.22061 Kreppel M, Drebber U, Wedemeyer I, Eich HT, Backhaus T, Zoller JE, Scheer M, 2011, Podoplanin expression predicts prognosis in patients with oral squamous cell carcinoma treated with neoadjuvant radiochemotherapy. Oral Oncol 47(9):873–878., DOI 10.1016/j. oraloncology.2011.06.508 Kreppel M, Scheer M, Drebber U, Ritter L, Zoller JE, 2010, Impact of podoplanin expression in oral squamous cell carcinoma: clinical and histopathologic correlations. Virchows Arch 456(5):473–482., DOI 10.1007/s00428-010-0915-7 Bartuli FN, Luciani F, Caddeo F, Compagni S, Piva P, Ottria L, Arcuri C, 2012, Podoplanin in the development and progression of oral cavity cancer: a preliminary study. Oral Implantol, Rome, 5(2– 3):33–41 Funayama A, Cheng J, Maruyama S, Yamazaki M, Kobayashi T, SyafriadiM, Kundu S, Shingaki S, Saito C, Saku T, 2011, Enhanced expression of podoplanin in oral carcinomas in situ and squamous cell carcinomas. Pathobiology 78(3):171–180., DOI 10.1159/ 000324926 Kato Y, Kaneko M, Sata M, Fujita N, Tsuruo T, Osawa M, 2005, Enhanced expression of Aggrus, T1alpha/podoplanin), a plateletaggregation- inducing factor in lung squamous cell carcinoma. Tumour Biol 26(4):195–200., DOI 10.1159/000086952 Kato Y, Sasagawa I, KanekoM, Osawa M, Fujita N, Tsuruo T, 2004, Aggrus: a diagnostic marker that distinguishes seminoma from embryonal carcinoma in testicular germ cell tumors. Oncogene 23(52): 8552–8556., DOI 10.1038/sj.onc.1207869 Shibahara J, Kashima T, Kikuchi Y, Kunita A, Fukayama M, 2006, Podoplanin is expressed in subsets of tumors of the central nervous system. Virchows Arch 448(4):493–499., DOI 10.1007/s00428-005- 0133-x Wu HM, Ren GX, Wang LZ, Zhang CY, Chen WT, Guo W, 2012, Expression of podoplanin in salivary gland adenoid cystic carcinoma and its association with distant metastasis and clinical outcomes. Mol Med Report 6(2):271–274., DOI 10.3892/mmr.2012.911 Xu Y, Ogose A, Kawashima H, Hotta T, Ariizumi T, Li G, Umezu H, Endo N, 2011, High-level expression of podoplanin in benign and malignant soft tissue tumors: immunohistochemical and quantitative real-time RT-PCR analysis. Oncol Rep 25(3):599–607., DOI 10.3892/ or.2011.1141 Ariizumi T, Ogose A, Kawashima H, Hotta T, Li G, Xu Y, Umezu H, Sugai M, Endo N, 2010, Expression of podoplanin in human bone and bone tumors: New marker of osteogenic and chondrogenic bone tumors. Pathol Int 60(3):193–202., DOI 10.1111/j.1440-1827.2009. 02510.x Zustin J, Scheuer HA, Friedrich RE, 2010, Podoplanin expression in human tooth germ tissues and cystic odontogenic lesions: an immunohistochemical study. J Oral Pathol Med 39(1):115–120., DOI 10. 1111/j.1600-0714.2009.00853.x Gonzalez-Alva P, Tanaka A, Oku Y, Miyazaki Y, Okamoto E, Fujinami M, Yoshida N, Kikuchi K, Ide F, Sakashita H, Kusama K, 2010, Enhanced expression of podoplanin in ameloblastomas. J Oral Pathol Med 39(1):103–109., DOI 10.1111/j.1600-0714.2009. 00818.x Padgett DM, Cathro HP, Wick MR, Mills SE, 2008, Podoplanin is a better immunohistochemical marker for sarcomatoid mesothelioma than calretinin. Am J Surg Pathol 32(1):123–127., DOI 10.1097/PAS. 0b013e31814faacf Tateyama H, Sugiura H, Yamatani C, Yano M, 2011, Expression of podoplanin in thymoma: its correlation with tumor invasion, nodal metastasis, and poor clinical outcome. Hum Pathol 42(4):533–540., DOI 10.1016/j.humpath.2010.08.012 Tong L, Yuan S, Feng F, Zhang H, 2012, Role of podoplanin expression in esophageal squamous cell carcinoma: a retrospective study. Dis Esophagus 25(1):72–80., DOI 10.1111/j.1442-2050.2011. 01211.x Rodrigo JP, Garcia-Carracedo D, Gonzalez MV, Mancebo G, Fresno MF, Garcia-Pedrero J, 2010, Podoplanin expression in the development and progression of laryngeal squamous cell carcinomas. Mol Cancer 9:48., DOI 10.1186/1476-4598-9-48 Suzuki H, Onimaru M, Koga T, Takeshita M, Yano T, Maehara Y, Nakamura S, Sueishi K, 2011, High podoplanin expression in cancer cells predicts lower incidence of nodal metastasis in patients with lung squamous cell carcinoma. Pathol Res Pract 207(2):111–115., DOI 10.1016/j.prp.2010.11.006 Seki S, FujiwaraM,MatsuuraM, Fujita S, Ikeda H, Asahina I, Ikeda T, 2011, Prediction of outcome of patients with oral squamous cell carcinoma using vascular invasion and the strongly positive expression of vascular endothelial growth factors. Oral Oncol 47(7):588– 593., DOI 10.1016/j.oraloncology.2011.04.013 Jakobsson PA, Eneroth CM, Killander D, Moberger G, Martensson B, 1973, Histologic classification and grading of malignancy in carcinoma of the larynx. Acta Radiol Ther Phys Biol 12(1):1–8 Schacht V, Dadras SS, Johnson LA, Jackson DG, Hong YK, Detmar M, 2005, Up-regulation of the lymphatic marker podoplanin, a mucin-type transmembrane glycoprotein, in human squamous cell carcinomas and germ cell tumors. Am J Pathol 166(3):913–921., DOI 10.1016/s0002-9440(10)62311-5 Akaike H, 1973, Information theory and an extention of the maximum likelihood principle. 2nd International Symposium on Information Theory. Budapest. First published Ihaka R, Gentleman R, 1996, R: a language for data analysis and graphics. J Comp Graph Stat 5:299–314 Astarita JL, Acton SE, Turley SJ, 2012, Podoplanin: emerging functions in development, the immune system, and cancer. Front Immunol 3:283., DOI 10.3389/fimmu.2012.00283 Cueni LN, Hegyi I, Shin JW, Albinger-Hegyi A, Gruber S, Kunstfeld R, Moch H, Detmar M, 2010, Tumor lymphangiogenesis and metastasis to lymph nodes induced by cancer cell expression of podoplanin. Am J Pathol 177(2):1004–1016., DOI 10.2353/ajpath. 2010.090703 Klimowicz AC, Bose P, Nakoneshny SC, Dean M, Huang L, Chandarana S, Magliocco AM, Wayne Matthews T, Brockton NT, Dort JC, 2012, Basal Ki67 expression measured by digital image analysis is optimal for prognostication in oral squamous cell carcinoma. Eur J Cancer 48(14):2166–2174., DOI 10.1016/j.ejca.2012.04. 010 Mashhadiabbas F, Mahjour F, Mahjour SB, Fereidooni F, Hosseini FS, 2012, The immunohistochemical characterization of MMP-2, MMP-10, TIMP-1, TIMP-2, and podoplanin in oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol 114(2):240– 250., DOI 10.1016/j.oooo.2012.04.009 Kawaguchi H, El-Naggar AK, Papadimitrakopoulou V, Ren H, Fan YH, Feng L, Lee JJ, Kim E, HongWK, Lippman SM,Mao L, 2008, Podoplanin: a novel marker for oral cancer risk in patients with oral premalignancy. J Clin Oncol 26(3):354–360., DOI 10.1200/jco.2007. 13.4072 Atsumi N, Ishii G, Kojima M, Sanada M, Fujii S, Ochiai A, 2008, Podoplanin, a novel marker of tumor-initiating cells in human squamous cell carcinoma A431. Biochem Biophys Res Commun 373(1): 36–41., DOI 10.1016/j.bbrc.2008.05.163 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 521 EP 528 DI 10.1007/s12253-013-9723-0 PG 8 ER PT J AU Varszegi, D Duga, B Melegh, IB Sumegi, K Kisfali, P Maasz, A Melegh, B AF Varszegi, Dalma Duga, Balazs Melegh, I Bela Sumegi, Katalin Kisfali, Peter Maasz, Anita Melegh, Bela TI Hodgkin Disease Therapy Induced Second Malignancy Susceptibility 6q21 Functional Variants in Roma and Hungarian Population Samples SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hodgkin’s lymphoma; PRDM1; Genetics; Susceptibility ID Hodgkin’s lymphoma; PRDM1; Genetics; Susceptibility AB Patients treated successfully for pediatric Hodgkin’s lymphoma are known to develop secondary malignancies; care is already taken in treatment to prevent this adverse effect. Recent GWAS study identified rs4946728 and rs1040411 noncoding SNPs located between PRDM1 and ATG1 genes on chromosome 6q21 as risk factors for secondary malignancies in patients formerly treated with radiotherapy for pediatric Hodgkin disease. We investigated the allele frequencies of these two SNPs in biobanked, randomly selected DNA of average, apparently healthy Hungarians (n =277) and in samples of Roma (n =279) population living Hungary. The risk allele frequency for rs4946728 was 79.4 % in Hungarian and 83.5 % in Roma samples, while for rs1040411 it was 56.4%in Hungarian and 55.8%in Roma samples. These values are quite similar in the two populations, and are rather high. The values are higher than those frequencies observed in the controls (rs4946728: 59.1 % and rs1040411: 39.6 %, p < 0.05), and are in the range of the cases (86 % and 68.2 %, respectively) of the above original GWAS study. Our findings suggest, that beside the already taken precautions, genetic characterization of Hungarian pediatric Hodgkin patients seems to be advantageous prior to the treatment of their disease. C1 [Varszegi, Dalma] University of Pecs, Department of Dermatology, Venereology and OncodermatologyPecs, Hungary. [Duga, Balazs] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, 7624 Pecs, Hungary. [Melegh, I Bela] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, 7624 Pecs, Hungary. [Sumegi, Katalin] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, 7624 Pecs, Hungary. [Kisfali, Peter] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, 7624 Pecs, Hungary. [Maasz, Anita] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, 7624 Pecs, Hungary. [Melegh, Bela] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, 7624 Pecs, Hungary. RP Melegh, B (reprint author), University of Pecs, Department of Medical Genetics and Child Development, 7624 Pecs, Hungary. EM bela.melegh@aok.pte.hu CR Friedman DL, Whitton J, Leisenring W, Mertens AC, Hammond S, Stovall M, Donaldson SS, Meadows AT, Robison LL, Neglia JP, 2010, Subsequent neoplasms in 5-year survivors of childhood cancer: the Childhood Cancer Survivor Study. J Natl Cancer Inst 102: 1083–1095 Meadows AT, Friedman DL, Neglia JP, Mertens AC, Donaldson SS, Stovall M, Hammond S, Yasui Y, Inskip PD, 2009, Second neoplasms in survivors of childhood cancer: findings fromthe Childhood Cancer Survivor Study cohort. J Clin Oncol 27:2356–2362 O’Brien MM, Donaldson SS, Balise RR, Whittemore AS, Link MP, 2010, Second malignant neoplasms in survivors of pediatric Hodgkin’s lymphoma treated with low-dose radiation and chemotherapy. J Clin Oncol 28:1232–1239 Best T, Li D, Skol AD, Kirchhoff T, Jackson SA, Yasui Y, Bhatia S, Strong LC, Domchek SM, Nathanson KL, Olopade OI, Huang RS, Mack TM, Conti DV, Offit K, CozenW, Robison LL, Onel K, 2011, Variants at 6q21 implicate PRDM1 in the etiology of therapy-induced second malignancies after Hodgkin’s lymphoma. Nat Med 17:941– 943 Keller AD,Maniatis T, 1991, Identification and characterization of a novel repressor of beta-interferon gene expression. Genes Dev 5: 868–879 Shaffer AL, LinKI, Kuo TC,Yu X, Hurt EM, RosenwaldA, Giltnane JM, Yang L, Zhao H, Calame K, Staudt LM, 2002, Blimp-1 orchestrates plasma cell differentiation by extinguishing the mature B cell gene expression program. Immunity 17:51–62 Shapiro-Shelef M, Calame K, 2005, Regulation of plasma-cell development. Nature reviews. Immunology 5:230–242 Chang DH, Angelin-Duclos C, Calame K, 2000, BLIMP-1: trigger for differentiation of myeloid lineage. Nat Immunol 1:169–176 Yan J, Jiang J, Lim CA, Wu Q, Ng HH, Chin KC, 2007, BLIMP1 regulates cell growth through repression of p53 transcription. Proc Natl Acad Sci U S A 104:1841–1846 Lin FR, Kuo HK, Ying HY, Yang FH, Lin KI, 2007, Induction of apoptosis in plasma cells by B lymphocyte-induced maturation protein-1 knockdown. Cancer Res 67:11914–11923 Nadasi E, Gyurus P, Czako M, Bene J, Kosztolanyi S, Fazekas S, Domosi P, Melegh B, 2007, Comparison of mtDNA haplogroups in Hungarians with four other European populations: a small incidence of descents with Asian origin. Acta Biol Hung 58:245–256 Semino O, Passarino G, Quintana-Murci L, Liu A, Beres J, Czeizel A, Santachiara-Benerecetti AS, 2000, MtDNA and Y chromosome polymorphisms in Hungary: inferences from the palaeolithic, neolithic and Uralic influences on the modern Hungarian gene pool. Eur J HumGenet 8:339–346 Tomory G, Csanyi B, Bogacsi-Szabo E, Kalmar T, Czibula A, Csosz A, Priskin K, Mende B, Lango P, Downes CS, Rasko I, 2007, Comparison of maternal lineage and biogeographic analyses of ancient and modern Hungarian populations. Am J Phys Anthropol 134: 354–368 Sipeky C, Lakner L, Szabo M, Takacs I, TamasiV, Polgar N, FalusA, Melegh B, 2009, Interethnic differences of CYP2C9 alleles in healthy Hungarian and Roma population samples: relationship to worldwide allelic frequencies. Blood Cells Mol Dis 43:239–242 Sipeky C, Csongei V, Jaromi L, Safrany E, Polgar N, Lakner L, Szabo M, Takacs I, Melegh B, 2009, Vitamin K epoxide reductase complex 1, VKORC1, haplotypes in healthy Hungarian and Roma population samples. Pharmacogenomics 10:1025–1032 Kalaydjieva L, Gresham D, Calafell F, 2001, Genetic studies of the Roma, Gypsies): a review. BMC Medical Genetics 2:5 Bombard Y, Abelson J, Simeonov D, Gauvin FP, 2013, Citizens’ perspectives on personalized medicine: a qualitative public deliberation study. Eur J Hum Genet Emmert-Streib F, 2012, Personalized medicine: Has it started yet? A reconstruction of the early history. Front Genet 3:313 Grosse SD, Khoury MJ, 2006, What is the clinical utility of genetic testing? Genetics in Med: Off J Am College ofMed Genetics 8:448– 450 Pasic MD, Samaan S, Yousef GM, 2013, Genomic medicine: new frontiers and new challenges. Clin Chem 59:158–167 Marshall E, 2011, Human genome 10th anniversary. Waiting for the revolution. Science 331:526–529 Behrens C, Travis LB, Wistuba II, Davis S, Maitra A, Clarke EA, Lynch CF, Glimelius B, Wiklund T, Tarone R, Gazdar AF, 2000, Molecular changes in second primary lung and breast cancers after therapy for Hodgkin’s disease. Cancer Epidemiology. Biomarkers and Prevention 9:1027–1035 Constine LS, Tarbell N, Hudson MM, Schwartz C, Fisher SG, Muhs AG, Basu SK, Kun LE, Ng A, Mauch P, Sandhu A, Culakova E, Lyman G, Mendenhall N, 2008, Subsequent malignancies in children treated for Hodgkin’s disease: associations with gender and radiation dose. Int J Radiat Oncol Biol Phys 72:24–33 Neglia JP, Friedman DL, Yasui Y, Mertens AC, Hammond S, Stovall M, Donaldson SS, Meadows AT, Robison LL, 2001, Second malignant neoplasms in 5-year survivors of childhood cancer: childhood cancer survivor study. J Natl Cancer Inst 93:618–629 Robison LL, Green DM, Hudson M, Meadows AT, Mertens AC, Packer RJ, Sklar CA, Strong LC, Yasui Y, Zeltzer LK, 2005, Longterm outcomes of adult survivors of childhood cancer. Cancer 104: 2557–2564 Sipeky C, Csongei V, Jaromi L, Safrany E,Maasz A, Takacs I, Beres J, Fodor L, Szabo M, Melegh B, 2011, Genetic variability and haplotype profile of MDR1, ABCB1, in Roma and Hungarian population samples with a review of the literature. Drug Metababolism and Pharmacokinet 26:206–215 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 529 EP 533 DI 10.1007/s12253-013-9724-z PG 5 ER PT J AU Chen, K Wu, D Bai, Y Zhu, X Chen, Z Wang, Ch Zhao, Y Li, M AF Chen, Kai Wu, Dajiang Bai, Yushu Zhu, Xiaodong Chen, Ziqiang Wang, Chuanfeng Zhao, Yingchuan Li, Ming TI Fuzzy Clustering Analysis of Osteosarcoma Related Genes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; Gene sequences; Fuzzy clustering; Runx2 ID Osteosarcoma; Gene sequences; Fuzzy clustering; Runx2 AB Osteosarcoma is the most common malignant bone-tumor with a peak manifestation during the second and third decade of life. In order to explore the influence of genetic factors on the mechanism of osteosarcoma by analyzing the inter relationship between osteosarcoma and its related genes, and then provide potential genetic references for the prevention, diagnosis and treatment of osteosarcoma, we collected osteosarcoma related gene sequences in Genebank of National Center for Biotechnology Information (NCBI) and local alignment analysis for a pair of sequences was carried out to identify the measurement association among related sequences. Then fuzzy clustering method was used for clustering analysis so as to contact the unknown genes through the consistent osteosarcoma related genes in one class. From the result of fuzzy clustering analysis, we could classify the osteosarcoma related genes into two groups and deduced that the genes clustered into one group had similar function. Based on this knowledge, we found more genes related to the pathogenesis of osteosarcoma and these genes could exert similar function as Runx2, a risk factor confirmed in osteosarcoma, this study may help better understand the genetic mechanism and provide new molecular markers and therapies for osteosarcoma. C1 [Chen, Kai] Changhai Hospital, Department of Orthopedics, 200433 Shanghai, China. [Wu, Dajiang] Changhai Hospital, Department of Orthopedics, 200433 Shanghai, China. [Bai, Yushu] Changhai Hospital, Department of Orthopedics, 200433 Shanghai, China. [Zhu, Xiaodong] Changhai Hospital, Department of Orthopedics, 200433 Shanghai, China. [Chen, Ziqiang] Changhai Hospital, Department of Orthopedics, 200433 Shanghai, China. [Wang, Chuanfeng] Changhai Hospital, Department of Orthopedics, 200433 Shanghai, China. [Zhao, Yingchuan] Changhai Hospital, Department of Orthopedics, 200433 Shanghai, China. [Li, Ming] Changhai Hospital, Department of Orthopedics, 200433 Shanghai, China. RP Li, M (reprint author), Changhai Hospital, Department of Orthopedics, 200433 Shanghai, China. EM minglil@hotmail.com CR Bielack SS, Kempf-Bielack B, Delling G, Exner GU, Flege S, Helmke K, Kotz R, Salzer-Kuntschik M, Werner M, Winkelmann W, 2002, Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol 20:776–790 Fletcher C.D, Unni K.K, Mertens F, 2002, Pathology & genetics: tumours of soft tissue and bone, World Health Organization Levine AJ, 1997, p53, the cellular gatekeeper review for growth and division. Cell 88:323–331 Kansara M, Thomas DM, 2007, Molecular pathogenesis of osteosarcoma. DNA Cell Biol 26:1–18 Sjin RTT, Naspinski J, Birsner A, Li C, Chan R, Lo K, Gillies S, Zurakowski D, Folkman J, Samulski J, 2006, Endostatin therapy reveals a U-shaped curve for antitumor activity. Cancer Gene Ther 13:619–627 Mohammed RAA, Green A, El-Shikh S, Paish E, Ellis I, Martin S, 2007, Prognostic significance of vascular endothelial cell growth factors-A,-C and-D in breast cancer and their relationship with angioand lymphangiogenesis. Br J Cancer 96:1092–1100 IIDA K, NOBORI T,MATSUMINE A, ISAKA A, Seto M, Shiraishi T, Uchida A, 2003, Effect of retinoblastoma tumor suppressor gene expression on chemosensitivity of human osteosarcoma cell lines. Oncol Rep 10:1961–1965 Ek ET, Dass CR, Contreras KG, Choong PF, 2007, Inhibition of orthotopic osteosarcoma growth and metastasis by multitargeted antitumor activities of pigment epithelium-derived factor. Clinical Exp Metastasis 24:93–106 Stricker S, Fundele R, Vortkamp A, Mundlos S, 2002, Role of Runx genes in chondrocyte differentiation. Dev Biol 245:95–108 Pratap J, Galindo M, Zaidi SK, Vradii D, Bhat BM, Robinson JA, Choi J-Y, Komori T, Stein JL, Lian JB, 2003, Cell growth regulatory role of Runx2 during proliferative expansion of preosteoblasts. Cancer Res 63:5357–5362 Choi J-Y, Pratap J, Javed A, Zaidi SK, Xing L, Balint E, Dalamangas S, Boyce B, Van Wijnen AJ, Lian JB, 2001, Subnuclear targeting of Runx/Cbfa/AML factors is essential for tissue-specific differentiation during embryonic development. Proc Natl Acad Sci 98:8650–8655 Ping G, Liangtian S, Shilun L, Lei S, 1996, A theoretical study of the effect of porous media on the dew point pressure of a gas condensate, in, SPE Gas Technology Symposium Saeidi, L. Handy, 1974, Flow and phase behavior of gas condensate and volatile oils in porous media, in, SPE California Regional Meeting Thompson JD, Higgins DG, Gibson TJ, 1994, CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Res 22:4673–4680 Altschul SF, GishW, MillerW, Myers EW, Lipman DJ, 1990, Basic local alignment search tool. J Mol Biol 215:403–410 Casey R, 2005, Blast sequences aid in genomics and proteomics. Business Intelligence Network Gustafson DE, Kessel WC, 1978, Fuzzy clustering with a fuzzy covariance matrix, in, Decision and Control including the 17th Symposium on Adaptive Processes, 1978 I.E. Conference on, IEEE, pp. 761–766 Dembele D, Kastner P, 2003, Fuzzy C-means method for clustering microarray data. Bioinformatics 19:973–980 Pickert L, Reuter I, Klawonn F, Wingender E, 1998, Transcription regulatory region analysis using signal detection and fuzzy clustering. Bioinformatics 14:244–251 Bezdek JC, 1981, Pattern recognition with fuzzy objective function algorithms, Kluwer Academic Publishers Eisen MB, Spellman PT, Brown PO, Botstein D, 1998, Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci 95:14863–14868 Banerjee C, Javed A, Choi J-Y, Green J, Rosen V, Van Wijnen AJ, Stein JL, Lian JB, Stein GS, 2001, Differential regulation of the two principal Runx2/Cbfa1 n-terminal isoforms in response to bone morphogenetic protein-2 during development of the osteoblast phenotype. Endocrinology 142:4026–4039 Stein GS, Lian JB, 1993, Molecular mechanisms mediating proliferation/differentiation interrelationships during progressive development of the osteoblast phenotype. Endocr Rev 14:424–442 Stewart M, TerryA,HuM,O’haraM, Blyth K, Baxter E, Cameron E, Onions D, Neil J, 1997, Proviral insertions induce the expression of bone-specific isoforms of PEBP2αA, CBFA1): evidence for a new myc collaborating oncogene. Proc Natl Acad Sci 94:8646–8651 Langdon C, Kerr C, Hassen M, Hara T, Arsenault AL, Richards CD, 2000, Murine oncostatin m stimulates mouse synovial fibroblasts in vitro and induces inflammation and destruction in mouse joints in vivo. Am J Pathol 157:1187–1196 Horn D, Fitzpatrick WC, Gompper PT, Ochs V, Bolton-Hansen M, Zarling J, Malik N, Todaro GJ, Linsley PS, 1990, Regulation of cell growth by recombinant oncostatin M. Growth Factors 2:157–165 Grant SL, Begley CG, 1999, The oncostatin M signalling pathway: reversing the neoplastic phenotype? Molecular Med Today 5:406– 412 Klausen P, Pedersen L, Jurlander J, Baumann H, 2000, OncostatinM and interleukin 6 inhibit cell cycle progression by prevention of p27kip1 degradation in HepG2 cells. Oncogene 19:3675 Halfter H, Lotfi R, Westermann R, Young P, Ringelstein EB, Stogbauer FT, 1998, Inhibition of growth and induction of differentiation of glioma cell lines by oncostatin M, OSM). Growth Factors 15:135–147 Bellido T, Borba VZ, Roberson P, Manolagas SC, 1997, Activation of the Janus kinase/STAT, signal transducer and activator of transcription, signal transduction pathway by interleukin-6-type cytokines promotes osteoblast differentiation. Endocrinology 138: 3666–3676 de Hooge AS, van de Loo FA, Bennink MB, de Jong DS, Arntz OJ, Lubberts E, Richards CD, van den Berg WB, 2002, Adenoviral transfer of murine oncostatin M elicits periosteal bone apposition in knee joints of mice, despite synovial inflammation and up-regulated expression of interleukin-6 and receptor activator of nuclear factor- κB ligand. Am J Pathol 160:1733–1743 Liu F, Aubin J, Malaval L, 2002, Expression of leukemia inhibitory factor, LIF)/interleukin-6 family cytokines and receptors during in vitro osteogenesis: differential regulation by dexamethasone and LIF. Bone 31:212 Wang Z-Q, Ovitt C, Grigoriadis AE, Mohle-Steinlein U, Ruther U, Wagner EF, 1992, Bone and haematopoietic defects in mice lacking c-fos Grigoriadis AE, Wang Z-Q, Cecchini MG, Hofstetter W, Felix R, Fleisch HA, Wagner EF, 1994, c-Fos: a key regulator of osteoclastmacrophage lineage determination and bone remodeling. Science 266:443–448 Grigoriadis AE, Wang Z-Q, Wagner EF, 1995, Fos and bone cell development: lessons from a nuclear oncogene. Trends Genet 11: 436–441 Chambers T, Fuller K, 1985, Bone cells predispose bone surfaces to resorption by exposure of mineral to osteoclastic contact. J Cell Sci 76:155–165 Fuller K, Gallagher A, Chambers T, 1991, Osteoclast resorption— stimulating activity is associated with the osteoblast cell surface and/ or the extracellular matrix. Biochem Biophys Res Commun 181:67–73 Rozen N, Ish-Shalom S, Rachmiel A, Stein H, Lewinson D, 2000, Interleukin-6 modulates trabecular and endochondral bone turnover in the nude mouse by stimulating osteoclast differentiation. Bone 26: 469–474 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 535 EP 539 DI 10.1007/s12253-013-9725-y PG 5 ER PT J AU Sheng, L Xiong, M Li, C Meng, X AF Sheng, Long Xiong, Maoming Li, Cong Meng, Xiangling TI Reversing Multidrug-Resistant by RNA Interference Through Silencing MDR1 Gene in Human Hepatocellular Carcinoma Cells Subline Bel-7402/ADM SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human hepatocellular carcinoma; siRNA; MDRl gene; Reversal; Bel-7402/ADM ID Human hepatocellular carcinoma; siRNA; MDRl gene; Reversal; Bel-7402/ADM AB Multidrug resistance (MDR) in hepatocellular carcinoma (HC) significantly impedes the effect of chemotherapy and is considered as a primary reason leading to its recurrences andmetastasis. The aimof present study was to explore new molecular targets for the reversal of MDR in HC by screening the adriamycin (ADM)-induced, human MDR-resistant HC cell subline Bel-7402/ADM. Small interfering RNAs (siRNAs) of four (MDR1si326, MDR1si1513, MDR1si2631 and MDR1si3071) targeting MDR1 were designed and transfected into Bel-7402/ADM cell strains. The experiments involved the following: mRNA expression of MDR1 gene by RT-PCR, P-glycoprotein (P-gp) expression by Western blot, intracellular ADM accumulation flow cytometry, and IC50 of ADM by a cytotoxic MTT assay. Four siRNAs reversed MDR in HC mediated by MDR1 to varying degrees. The expression level of MDR1 mRNA in cells of MDR1si326 or MDR1si2631 group (0.190±0.038 or 0.171± 0.011) was more decreased. The expression level of P-gp in cells of MDR1si326 group was the lowest. The accumulation of ADM in cells of MDR1si326 or MDR1si2631 group (77.0±3.5 or 75.4±2.9) was more increased. The IC50 of cells to ADM was lowest in MDR1si326 group (11.32±0.69 mg/L). Compared with other three siRNAs, MDR1si326 performed the optimal reversal effect of drug resistance in human HC Bel-7402/ADM. C1 [Sheng, Long] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, China. [Xiong, Maoming] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, China. [Li, Cong] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, China. [Meng, Xiangling] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, China. RP Xiong, M (reprint author), The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, China. EM xiongmd@aliyun.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90., DOI 10.3322/ caac.20107 Perz JF, Armstrong GL, Farrington LA, Hutin YJ, Bell BP, 2006, The contributions of hepatitis B virus and hepatitis C virus infections to cirrhosis and primary liver cancer worldwide. J Hepatol 45:529– 538., DOI 10.1016/j.jhep.2006.05.013 Zhuo L, Liu J,Wang B, GaoM, HuangA, 2013, DifferentialmiRNA expression profiles in hepatocellular carcinoma cells and drugresistant sublines. Oncol Rep 29:555–562., DOI 10.3892/or.2012.2155 Davidson A, Dick G, Pritchard-Jones K, Pinkerton R, 2002, EVE/ cyclosporin, etoposide, vincristine, epirubicin with high-dose cyclosporin)- chemotherapy selected for multidrug resistance modulation. Eur J Cancer 38:2422–2427., DOI 10.1016/S0959-8049(02)00493-8 Tsang TY, Tsang SW, Lai KP, Tsang WP, Co NN, Kwok TT, 2009, Facilitation of drug resistance development by gamma-irradiation in human cancer cells. Oncol Rep 22:921–926., DOI 10.3892/or_ 00000518 Fantappie O, Solazzo M, Lasagna N, Platini F, Tessitore L, Mazzanti R, 2007, P-glycoprotein mediates celecoxib-induced apoptosis in multiple drug-resistant cell lines. Cancer Res 67:4915–4923., DOI 10.1158/0008-5472.CAN-06-3952 Roy KR, Reddy GV, Maitreyi L, Agarwal S, Achari C, Vali S, Reddanna P, 2010, Celecoxib inhibits MDR1 expression through COX-2-dependent mechanism in human hepatocellular carcinoma, HepG2, cell line. Cancer Chemother Pharmacol 65:903–911., DOI 10.1007/s00280-009-1097-3 Keaney J, Campbell M, Humphries P, 2011, From RNA interference technology to effective therapy: howfar havewe come and howfar to go? Ther Deliv 2:1395–1406., DOI 10.4155/tde.11.109 Tschaharganeh D, Ehemann V, Nussbaum T, Schirmacher P, Breuhahn K, 2007, Non-specific effects of siRNAs on tumor cells with implications on therapeutic applicability using RNA interference. Pathol Oncol Res 13:84–90., DOI 10.1007/BF02893482 Bora RS, Gupta D, Mukkur TK, Saini KS, 2012, RNA interference therapeutics for cancer: challenges and opportunities, review). Mol Med Rep 6:9–15., DOI 10.3892/mmr.2012.871 McManus MT, Sharp PA, 2002, Gene silencing in mammals by small interfering RNAs. Nat Rev Genet 3:737–747., DOI 10.1038/ nrg908 Bulk E, Hascher A, Liersch R, Mesters RM, Diederichs S, Sargin B, Gerke V, Hotfilder M, Vormoor J, Berdel WE, Serve H, Muller-Tidow C, 2008, Adjuvant therapy with small hairpin RNA interference prevents non-small cell lung cancer metastasis development in mice. Cancer Res 68:1896–1904., DOI 10.1158/0008-5472.CAN-07- 2390 Yoshida Y, Wang IC, Yoder HM, Davidson NO, Costa RH, 2007, The forkhead box M1 transcription factor contributes to the development and growth of mouse colorectal cancer. Gastroenterology 132:1420–1431., DOI 10.1053/j.gastro.2007. 01.036 Iorns E, Lord CJ, Turner N, Ashworth A, 2007, Utilizing RNA interference to enhance cancer drug discovery. Nat Rev Drug Discov 6:556–568., DOI 10.1038/nrd2355 Elbashir SM, Martinez J, Patkaniowska A, Lendeckel W, Tuschl T, 2001, Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate. EMBO J 20:6877–6888., DOI 10.1093/emboj/20.23.6877 Wilda M, Fuchs U, Wossmann W, Borkhardt A, 2002, Killing of leukemic cells with a BCR/ABL fusion gene by RNA interference, RNAi). Oncogene 21:5716–5724., DOI 10.1038/sj.onc.1205653 Mirza A, Basso A, Black S, Malkowski M, Kwee L, Pachter JA, Lachowicz JE, Wang Y, Liu S, 2005, RNA interference targeting of A1 receptor-overexpressing breast carcinoma cells leads to diminished rates of cell proliferation and induction of apoptosis. Cancer Biol Ther 4:1355–1360., DOI 10.4161/cbt.4.12.2196 Zhang PH, Zou L, Tu ZG, 2006, RNAi-hTERT inhibition hepatocellular carcinoma cell proliferation via decreasing telomerase activity. J Surg Res 131:143–149., DOI 10.1016/j.jss.2005.09.017 Farra R, Dapas B, Pozzato G, Scaggiante B, Agostini F, Zennaro C, GrassiM, Rosso N, Giansante C, Fiotti N, Grassi G, 2011, Effects of E2F1-cyclin E1-E2 circuit down regulation in hepatocellular carcinoma cells. Dig Liver Dis 43:1006–1014., DOI 10.1016/j.dld.2011.07. 007 Nieth C, Priebsch A, Stege A, Lage H, 2003, Modulation of the classical multidrug resistance, MDR, phenotype by RNA interference, RNAi). FEBS Lett 545:144–150., DOI 10.1016/S0014-5793(03, 00523-4 Sun Y, LiuM, Yang B, Li B, Lu J, 2008, Role of siRNA silencing of MMP-2 gene on invasion and growth of laryngeal squamous cell carcinoma. Eur Arch Otorhinolaryngol 265:1385–1391., DOI 10. 1007/s00405-008-0684-y Zhao P, Zhang Y, Sun M, He Y, 2008, Reversion of multidrug resistance in human glioma by RNA interference. Neurol Res 30: 562–566., DOI 10.1179/174313208X297869 Hua J, Mutch DG, Herzog TJ, 2005, Stable suppression of MDR-1 gene using siRNA expression vector to reverse drug resistance in a human uterine sarcoma cell line. Gynecol Oncol 98:31–38., DOI 10. 1016/j.ygyno.2005.03.042 Halder J, Landen CN, Jr., Lutgendorf SK, Li Y, Jennings NB, Fan D, Nelkin GM, Schmandt R, Schaller MD, Sood AK, 2005, Focal adhesion kinase silencing augments docetaxel-mediated apoptosis in ovarian cancer cells. Clin Cancer Res 11:8829–8836., DOI 10. 1158/1078-0432.CCR-05-1728 Zhao Y, Liu J, Hong Q, Yang C, Chen L, Chen Y, Wang Q, Zhao K, Jin W, 2010, Involvement of MyoD and PEA3 in regulation of transcription activity of MDR1 gene. Acta Biochim Biophys Sin, Shanghai, 42:900–907., DOI 10.1093/abbs/gmq094 Klosek SK, Nakashiro K, Hara S, Goda H, Hamakawa H, 2008, Stat3 as a molecular target in RNA interference-based treatment of oral squamous cell carcinoma. Oncol Rep 20:873–878., DOI 10.3892/ or_00000085 Olejniczak M, Galka P, Krzyzosiak WJ, 2010, Sequence-nonspecific effects of RNA interference triggers and microRNA regulators. Nucleic Acids Res 38:1–16., DOI 10.1093/nar/gkp829 Chan GN, Bendayan R, 2011, Molecular and functional characterization of P-glycoprotein in vitro. Methods Mol Biol 686:313–336., DOI 10.1007/978-1-60761-938-3_15 Zhong X, Xiong M, Meng X, Gong R, 2010, Comparison of the multi-drug resistant human hepatocellular carcinoma cell line Bel- 7402/ADM model established by three methods. J Exp Clin Cancer Res 29:115., DOI 10.1186/1756-9966-29-115 Speth PA, Linssen PC, Boezeman JB,Wessels HM, Haanen C, 1987, Cellular and plasma adriamycin concentrations in long-term infusion therapy of leukemia patients. Cancer Chemother Pharmacol 20:305– 310., DOI 10.1007/BF00262581 Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T, 2001, Duplexes of 21-nucleotide RNAs mediate RNA interference in culturedmammalian cells. Nature 411:494–498., DOI 10.1038/35078107 Vorobjeva MA, Davydova AS, Venyaminova AG, 2011, Artificial hammerhead ribozymes: engineering and applications. Russian Chem Rev 80:127., DOI 10.1070/RC2011v080n02ABEH004142 Gaspar P, Moura G, Santos MA, Oliveira JL, 2013, mRNA secondary structure optimization using a correlated stem-loop prediction. Nucleic Acids Res 41:e73., DOI 10.1093/nar/gks1473 Fujiwara T, Yada T, 2013, miRNA-target prediction based on transcriptional regulation. BMC Genomics 14(Suppl 2):S3., DOI 10.1186/1471- 2164-14-S2-S3 Miyagishi M, Hayashi M, Taira K, 2003, Comparison of the suppressive effects of antisense oligonucleotides and siRNAs directed against the same targets in mammalian cells. Antisense Nucleic Acid Drug Dev 13:1–7., DOI 10.1089/108729003764097296 Hinrichsen L, Harborth J, Andrees L, Weber K, Ungewickell EJ, 2003, Effect of clathrin heavy chain- and alpha-adaptin-specific small inhibitory RNAs on endocytic accessory proteins and receptor trafficking in HeLa cells. J Biol Chem 278:45160–45170., DOI 10. 1074/jbc.M307290200 Filipowicz W, Jaskiewicz L, Kolb FA, Pillai RS, 2005, Posttranscriptional gene silencing by siRNAs and miRNAs. Curr Opin Struct Biol 15:331–341., DOI 10.1016/j.sbi.2005.05.006 Ambudkar SV, Sauna ZE, Gottesman MM, Szakacs G, 2005, A novel way to spread drug resistance in tumor cells: functional intercellular transfer of P-glycoprotein, ABCB1). Trends Pharmacol ScI 26:385–387., DOI 10.1016/j.tips.2005.06.001 Labroille G, Belloc F, Bilhou-Nabera C, Bonnefille S, Bascans E, Boisseau MR, Bernard P, Lacombe F, 1998, Cytometric study of intracellular P-gp expression and reversal of drug resistance. Cytometry 32:86-94., DOI 10.1002/(SICI)1097-0320(19980601, 32:2<86::AID-CYTO3>3.0.CO;2-C Cordon-Cardo C, O’Brien JP, Boccia J, Casals D, Bertino JR, Melamed MR, 1990, Expression of the multidrug resistance gene product, P-glycoprotein, in human normal and tumor tissues. J Histochem Cytochem 38:1277–1287., DOI 10.1177/38.9.1974900 Miller DS, Nobmann SN, Gutmann H, Toeroek M, Drewe J, Fricker G, 2000, Xenobiotic transport across isolated brain microvessels studied by confocal microscopy. Mol Pharmacol 58:1357–1367., DOI 10.1124/mol.58.6.1357 Miller DS, 2010, Regulation of P-glycoprotein and other ABC drug transporters at the blood–brain barrier. Trends Pharmacol Sci 31:246– 254., DOI 10.1016/j.tips.2010.03.003 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 541 EP 548 DI 10.1007/s12253-013-9726-x PG 8 ER PT J AU Zhou, Y Wan, Ch Liu, Y Lv, L Chen, B Ni, R Huang, Y Li, Y Zheng, X Yang, D Mao, G Xue, Q AF Zhou, Yiqun Wan, Chunhua Liu, Yifei Lv, Liting Chen, Buyou Ni, Runzhou Huang, Yuexia Li, Yangcheng Zheng, Xiaodong Yang, Dunpeng Mao, Guoxin Xue, Qun TI Polycomb Group Oncogene RING1 is Over-expressed in Non-Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; RING1; Prognosis ID Non-small cell lung cancer; RING1; Prognosis AB Ring finger protein 1 (RING1) have recently been reported to be related to aggressive tumor features in Prostate Cancer and urothelial carcinoma of the bladder. However, the role of RING1 in non-small-cell lung cancer (NSCLC) tumorigenesis has never been elucidated. So we aimed at investigating the potential role of RING1 in NSCLC. RING1 expression was evaluated by Immunoblot in 8 paired fresh lung cancer tissues and immunohistochemistry on 69 paraffinembedded sections from 2006 to 2009. Furthermore, flowcytometry and RNA interference were performed to analyse the role of RING1 in A549 cells. We showed that the expression level of RING1 was significant increased in lung cancer as compared with the adjacent normal tissue. High expression level of RING1 was associated with TNM stage (P =0.013), and RING1 was positively related with proliferation marker Ki67 (P <0.05). Moreover, RING1 knockdown induces growth suppression of human lung cancer cells through G1/ S cell cycle phase arrest in vitro. Kaplan–Meier survival curves showed that high expression level of RING1 was associated with poor prognosis (P =0.03). On the basis of these results, we suggested that RING1 protein expression may be a favorable independent prognostic parameter for non-small cell lung cancer. C1 [Zhou, Yiqun] Affiliated Hospital of Nantong University, Department of Thoracic Surgery, 226001 Nantong, Jiangsu, China. [Wan, Chunhua] Nantong University, Department of Public Health, 226001 Nantong, Jiangsu, China. [Liu, Yifei] Affiliated Hospital of Nantong University, Department of Oncology, 226001 Nantong, Jiangsu, China. [Lv, Liting] Affiliated Hospital of Nantong University, Department of Oncology, 226001 Nantong, Jiangsu, China. [Chen, Buyou] Affiliated Hospital of Nantong University, Department of Oncology, 226001 Nantong, Jiangsu, China. [Ni, Runzhou] Affiliated Hospital of Nantong University, Department of Gastroenterology, 226001 Nantong, Jiangsu, China. [Huang, Yuexia] Affiliated Hospital of Nantong University, Department of Gastroenterology, 226001 Nantong, Jiangsu, China. [Li, Yangcheng] Affiliated Hospital of Nantong University, Department of Thoracic Surgery, 226001 Nantong, Jiangsu, China. [Zheng, Xiaodong] Affiliated Hospital of Nantong University, Department of Oncology, 226001 Nantong, Jiangsu, China. [Yang, Dunpeng] Affiliated Hospital of Nantong University, Department of Thoracic Surgery, 226001 Nantong, Jiangsu, China. [Mao, Guoxin] Affiliated Hospital of Nantong University, Department of Oncology, 226001 Nantong, Jiangsu, China. [Xue, Qun] Affiliated Hospital of Nantong University, Department of Thoracic Surgery, 226001 Nantong, Jiangsu, China. RP Mao, G (reprint author), Affiliated Hospital of Nantong University, Department of Oncology, 226001 Nantong, China. EM ntmgx123@163.com CR Buchenau P, Hodgson J, Strutt H, Arndt-Jovin DJ, 1998, The distribution of poly-comb-group proteins during cell division and development in Drosophila embryos: impact on models for silencing. J Cell Biol 141(2):469–481 Caldas C, Aparicio S, 1999, Cellmemory and cancer–the story of the trithorax and Poly-comb group genes. Cancer Metastasis Rev 18(2): 313–329 Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P et al, 2002, Role of histone H3 lysine 27 methylation in Poly-combgroup silencing. Science 298(5595):1039–1043 CaoW, Ribeiro Rde O, Liu D, Saintigny P, Xia R, Xue Yet al, 2012, EZH2 promotes malignant behaviors via cell cycle dysregulation and its mRNA level associates with prognosis of patient with non-small cell lung cancer. PLoS One 7(12):e52984 Cooper CS, Foster CS, 2009, Concepts of epigenetics in prostate cancer development. Br J Cancer 100(2):240–245 Czermin B,Melfi R, McCabe D, Seitz V, Imhof A, Pirrotta V, 2002, Drosophila enhancer of Zeste/ESC complexes have a histone H3 methyltransferase activity that marks chromosomal Poly-comb sites. Cell 111(2):185–196 Dimri GP, Martinez JL, Jacobs JJ, Keblusek P, Itahana K, Van Lohuizen M et al, 2002, The Bmi-1 oncogene induces telomerase activity and immortalizes human mammary epithelial cells. Cancer Res 62(16):4736–4745 Ferraiuolo MA, Rousseau M, Miyamoto C, Shenker S, Wang XQ, Nadler M et al, 2010, The three-dimensional architecture of Hox cluster silencing. Nucleic Acids Res 38(21):7472–7484 Fussbroich B, Wagener N, Macher-Goeppinger S, Benner A, Falth M, Sultmann H et al, 2011, EZH2 depletion blocks the proliferation of colon cancer cells. PLoS One 6(7):e21651 Goebl MG(1991, The bmi-1 and mel-18 gene products define a new family of DNA-binding proteins involved in cell proliferation and tumorigenesis. Cell 66(4):623 Hinz S, Kempkensteffen C, Christoph F, Krause H, Schrader M, Schostak M et al, 2008, Expression parameters of the poly-comb group proteins BMI1, SUZ12, RING1 and CBX7 in urothelial carcinoma of the bladder and their prognostic relevance. Tumour Biol 29(5):323–329 Jacobs JJ, Scheijen B, Voncken JW, Kieboom K, Berns A, van LohuizenM, 1999, Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF. Genes Dev 13(20):2678–2690 Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60(5):277–300 Ke Q, Ji J, Cheng C, Zhang Y, LuM,Wang Yet al, 2009, Expression and prognostic role of Spy1 as a novel cell cycle protein in hepatocellular carcinoma. Exp Mol Pathol 87(3):167–172 Kerr KM, 2013, Clinical relevance of the new IASLC/ERS/ATS adenocarcinoma classification. J Clin Pathol 66(10):832–8 Kovalchuk O, Tryndyak VP, Montgomery B, Boyko A, Kutanzi K, Zemp F et al, 2007, Estrogen-induced rat breast carcinogenesis is characterized by alterations in DNA methylation, histone modifications and aberrant micro-RNA expression. Cell Cycle 6(16):2010– 2018 Li H, Bitler BG, Vathipadiekal V, Maradeo ME, Slifker M, Creasy CL et al, 2012, ALDH1A1 is a novel EZH2 target gene in epithelial ovarian cancer identified by genome-wide approaches. Cancer Prev Res, Phila, 5(3):484–491 Li Z, Qu L, Dong Q, Huang B, Li H, Tang Z et al, 2012, Overexpression of CARMA3 in non-small-cell lung cancer is linked for tumor progression. PLoS One 7(5):e36903 Lu, C., Liu, G., Cui, X., Zhang, J., Wei, L., Wang, Y., et al., 2013). Expression of SGTA Correlates with Prognosis and Tumor Cell Proliferation in Human Hepatocellular Carcinoma. Pathol Oncol Res Mathers CD, Loncar D, 2006, Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med 3(11):e442 Qin H, Wang J, Liang Y, Taniguchi Y, Tanigaki K, Han H, 2004, RING1 inhibits transactivation of RBP-J by Notch through interaction with LIM protein KyoT2. Nucleic Acids Res 32(4):1492–1501 Raaphorst FM,Meijer CJ, Fieret E, Blokzijl T, Mommers E, Buerger H et al, 2003, Poorly differentiated breast carcinoma is associated with increased expression of the human poly-comb group EZH2 gene. Neoplasia 5(6):481–488 Raaphorst FM, Vermeer M, Fieret E, Blokzijl T, Dukers D, Sewalt RG et al, 2004, Site-specific expression of poly-comb-group genes encoding the HPC-HPH/PRC1 complex in clinically defined primary nodal and cutaneous large B-cell lymphomas. Am J Pathol 164(2): 533–542 Satijn DP, Otte AP, 1999, RING1 interacts with multiple Poly-combgroup proteins and displays tumorigenic activity. Mol Cell Biol 19(1):57–68 Satijn DP, GunsterMJ, van der Vlag J, Hamer KM, SchulW, Alkema MJ et al, 1997, RING1 is associated with the poly-comb group protein complex and acts as a transcriptional repressor. Mol Cell Biol 17(7):4105–4113 Satijn DP, Olson DJ, van der Vlag J, Hamer KM, Lambrechts C, Masselink H et al, 1997, Interference with the expression of a novel human poly-comb protein, hPc2, results in cellular transformation and apoptosis. Mol Cell Biol 17(10):6076–6086 Shin YJ, Kim JH, 2012, The role of EZH2 in the regulation of the activity of matrix metalloproteinases in prostate cancer cells. PLoS One 7(1):e30393 Travis WD, Brambilla E, Muller-Hermelink HK et al, 2004, World Health Organisation Classification of Tumors. Pathology and Genetics of tumors of the lung. Thymus and Heart, p 1 van Leenders GJ, Dukers D, Hessels D, van den Kieboom SW, Hulsbergen CA, Witjes JA et al, 2007, Poly-comb-group oncogenes EZH2, BMI1, and RING1 are over-expressed in prostate cancer with adverse pathologic and clinical features. Eur Urol 52(2):455–463 Wang Y, Fei M, Cheng C, Zhang D, Lu J, He S et al, 2008, Jun activation domain-binding protein 1 negatively regulate p27 kip1 in non-Hodgkin’s lymphomas. Cancer Biol Ther 7(3):460–467 Wang Y, Yang S, Ni Q, He S, Zhao Y, Yuan Q et al, 2012, Overexpression of forkhead box J2 can decrease the migration of breast cancer cells. J Cell Biochem 113(8):2729–2737 Watanabe H, Soejima K, Yasuda H, Kawada I, Nakachi I, Yoda S et al, 2008, Deregulation of histone lysine methyltransferases contributes to oncogenic transformation of human bronchoepithelial cells. Cancer Cell Int 8:15 Xu X, Yamamoto H, Sakon M, YasuiM, Ngan CY, Fukunaga H et al, 2003, Over-expression of CDC25A phosphatase is associated with hyper-growth activity and poor prognosis of human hepatocellular carcinomas. Clin Cancer Res 9(5):1764–1772 Zhang Y, Peng C, Wu G, Wang Y, Liu R, Yang S et al, 2011, Expression of NLK and its potential effect in ovarian cancer chemotherapy. Int J Gynecol Cancer 21(8):1380–1387 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 549 EP 556 DI 10.1007/s12253-013-9727-9 PG 8 ER PT J AU Fernandes, FB Coates, J Odashiro, NA Quezada, C Huynh, A Odashiro, RP Odashiro, M Burnier, NM AF Fernandes, F Bruno Coates, James Odashiro, N Alexandre Quezada, Carlos Huynh, Aimee Odashiro, R Patricia Odashiro, Macanori Burnier, N Miguel TI Hypoxia-Inducible Factor-1α and its Role in the Proliferation of Retinoblastoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hypoxic; Hypoxic-inducible factor; Retinoblastoma; Knockdown; Proliferation ID Hypoxic; Hypoxic-inducible factor; Retinoblastoma; Knockdown; Proliferation AB In order to better understand the role of HIF-1α in the proliferation of the retinoblastoma cells, a siRNA knockdown of HIF-1α followed by a proliferation assay was performed. Further sequencing was then carried out in order to assess knockdown efficiency and expression of HIF-1α. Upregulation of HIF-1α gene expression in CoCl2-treated retinoblastoma cells was demonstrated via melting curve analysis from PCR tests and was further analyzed using western blot and densitometry analysis. Reduction of HIF-1α expression in retinoblastoma, post HIF-1α knockdown, was observed after siRNA transfection into Y-79 cells. Knockdown of HIF-1α resulted in a significant decrease in proliferation thereby demonstrating that HIF-1α is involved in promoting survival and proliferation in retinoblastoma cells. Stabilization of HIF-1α in retinoblastoma cells using CoCl2 was unsuccessful. C1 [Fernandes, F Bruno] The McGill University Health Center & Henry C. Witelson Ocular Pathology Laboratory, Department of Ophthalmology and Pathology, 3775 University St., Room 216, H3A 2B4 Montreal, QC, Canada. [Coates, James] The McGill University Health Center & Henry C. Witelson Ocular Pathology Laboratory, Department of Ophthalmology and Pathology, 3775 University St., Room 216, H3A 2B4 Montreal, QC, Canada. [Odashiro, N Alexandre] McMaster University, Department of Pathology and Molecular MedicineHamilton, ON, Canada. [Quezada, Carlos] The McGill University Health Center & Henry C. Witelson Ocular Pathology Laboratory, Department of Ophthalmology and Pathology, 3775 University St., Room 216, H3A 2B4 Montreal, QC, Canada. [Huynh, Aimee] The McGill University Health Center & Henry C. Witelson Ocular Pathology Laboratory, Department of Ophthalmology and Pathology, 3775 University St., Room 216, H3A 2B4 Montreal, QC, Canada. [Odashiro, R Patricia] The McGill University Health Center & Henry C. Witelson Ocular Pathology Laboratory, Department of Ophthalmology and Pathology, 3775 University St., Room 216, H3A 2B4 Montreal, QC, Canada. [Odashiro, Macanori] Federal University of Mato Grosso do SulCampo Grande, MS, Brazil. [Burnier, N Miguel] The McGill University Health Center & Henry C. Witelson Ocular Pathology Laboratory, Department of Ophthalmology and Pathology, 3775 University St., Room 216, H3A 2B4 Montreal, QC, Canada. RP Quezada, C (reprint author), The McGill University Health Center & Henry C. Witelson Ocular Pathology Laboratory, Department of Ophthalmology and Pathology, H3A 2B4 Montreal, Canada. EM quezadarc@hotmail.com CR Arean C, OrellanaME, Abourbih D, Abreu C, Pifano I, Burnier MN Jr, 2010, Expression of vascular endothelial growth factor in retinoblastoma. Arch Ophthalmol 128(2):223–229 Boutrid H, Jockovich ME, Murray TG, Pina Y, Feuer WJ, Lampidis TJ, Cebulla CM, 2008, Targeting hypoxia, a novel treatment for advanced retinoblastoma. Investig Ophthalmol Vis Sci 49(7):2799– 2805 Kunz M, Ibrahim SM, 2003, Molecular responses to hypoxia in tumor cells. Mol Cancer 2:23 Mayes PA, Dolloff NG, Daniel CJ, Liu JJ, Hart LS, Kuribayashi K, Allen JE, Jee DI, Dorsey JF, Liu YY et al, 2011, Overcoming hypoxia-induced apoptotic resistance through combinatorial inhibition of GSK-3beta and CDK1. Cancer Res 71(15):5265–5275 Hockel M, Vaupel P, 2001, Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 93(4): 266–276 Piret JP, Mottet D, Raes M, Michiels C, 2002, CoCl2, a chemical inducer of hypoxia-inducible factor-1, and hypoxia reduce apoptotic cell death in hepatoma cell line HepG2. Ann N YAcad Sci 973:443– 447 Semenza GL, 2003, Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3(10):721–732 Kaur B, Khwaja FW, Severson EA, Matheny SL, Brat DJ, Van Meir EG, 2005, Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro-Oncology 7(2):134–153 Loboda A, Jozkowicz A, Dulak J, 2010, HIF-1 and HIF-2 transcription factors—similar but not identical. Mol Cells 29(5):435–442 Mendez O, Zavadil J, Esencay M, Lukyanov Y, Santovasi D, Wang SC, Newcomb EW, Zagzag D, 2010, Knock down of HIF-1alpha in glioma cells reduces migration in vitro and invasion in vivo and impairs their ability to form tumor spheres. Mol Cancer 9:133 Vaupel P, Kelleher DK, HockelM(2001, Oxygen status ofmalignant tumors: pathogenesis of hypoxia and significance for tumor therapy. Semin Oncol 28(2 Suppl 8):29–35 Pina Y, Decatur C, Murray T, Houston S, Gologorsky D, Cavalcante M, Cavalcante L, Hernandez E, Celdran M, Feuer W et al, 2011, Advanced retinoblastoma treatment: targeting hypoxia by inhibition of the mammalian target of rapamycin, mTOR, in LH(BETA)T(AG, retinal tumors. Clin Ophthalmol 5:337–343 Yuan Y, Hilliard G, Ferguson T, Millhorn DE, 2003, Cobalt inhibits the interaction between hypoxia-inducible factor-alpha and von Hippel-Lindau protein by direct binding to hypoxia-inducible factor-alpha. J Biol Chem 278(18):15911–15916 Grasselli F, Basini G, Bussolati S, Bianco F, 2005, Cobalt chloride, a hypoxia-mimicking agent, modulates redox status and functional parameters of cultured swine granulosa cells. Reprod Fertil Dev 17(7):715–720 Cikos S, Bukovska A, Koppel J, 2007, Relative quantification of mRNA: comparison of methods currently used for real-time PCR data analysis. BMC Mol Biol 8 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(T)(-Delta Delta C, method. Methods 25(4):402–408 Chang CI, Kim HA, Dua P, Kim S, Li CJ, Lee DK, 2011, Structural diversity repertoire of gene silencing small interferingRNAs.Nucleic Acid Ther 21(3):125–131 Burr DB, Molina SA, Banerjee D, Low DM, Takemoto DJ, 2011, Treatment with connexin 46 siRNA suppresses the growth of human Y79 retinoblastoma cell xenografts in vivo. Exp Eye Res 92(4):251– 259 Manohar SM, Padgaonkar AA, Jalota-Badhwar A, Sonawane V, Rathos MJ, Kumar S, Joshi KS, 2011, A novel inhibitor of hypoxia-inducible factor-1 alpha P3155 also modulates PI3K pathway and inhibits growth of prostate cancer cells. BMC Cancer 11 Kaelin WG, 1999, Functions of the retinoblastoma protein. Bioessays 21(11):950–958 Wang GL, Semenza GL, 1993, Desferrioxamine induces erythropoietin gene-expression and hypoxia-inducible factor-I DNA-binding activity—implications for models of hypoxia signal-transduction. Blood 82(12):3610–3615 Jantsch J, Wiese M, Schodel J, Castiglione K, Glasner J, Kolbe S, Mole D, Schleicher U, Eckardt KU, Hensel M et al, 2011, Toll-like receptor activation and hypoxia use distinct signaling pathways to stabilize hypoxia-inducible factor 1 alpha, HIF1A, and result in differential HIF1A-dependent gene expression. J Leukoc Biol 90(3):551–562 Fletcher CDM, 1995, Diagnostic histopathology of tumors. Churchill Livingstone, Edinburgh Huang X, Ding L, Bennewith KL, Tong RT, Welford SM, Ang KK, Story M, Le QT, Giaccia AJ, 2009, Hypoxia-inducible mir-210 regulates normoxic gene expression involved in tumor initiation. Mol Cell 35(6):856–867 Berra E,Milanini J, RichardDE, Le GallM,Vinals F, Gothie E, Roux D, Pages G, Pouyssegur J, 2000, Signaling angiogenesis via p42/p44 MAP kinase and hypoxia. Biochem Pharmacol 60(8):1171–1178 Mahon PC, Hirota K, Semenza GL, 2001, FIH-1: a novel protein that interacts with HIF-1 alpha and VHL to mediate repression of HIF-1 transcriptional activity. Gene Dev 15(20):2675–2686 Wang GL, Semenza GL, 1993, General involvement of hypoxiainducible factor 1 in transcriptional response to hypoxia. Proc Natl Acad Sci U S A 90(9):4304–4308 Berra E, Pages G, Pouyssegur J, 2000, MAP kinases and hypoxia in the control of VEGF expression. Cancer Metastasis Rev 19(1–2): 139–145 de Souza N, 2007, Too much of a good thing. Nat Methods 4(5):386 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 557 EP 563 DI 10.1007/s12253-013-9728-8 PG 7 ER PT J AU Ujj, Zs Buglyo, G Udvardy, M Vargha, Gy Biro, S Rejto, L AF Ujj, Zsofia Buglyo, Gergely Udvardy, Miklos Vargha, Gyorgy Biro, Sandor Rejto, Laszlo TI WT1 Overexpression Affecting Clinical Outcome in Non-Hodgkin Lymphomas and Adult Acute Lymphoblastic Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE WT1 gene; Overexpression; Non-Hodgkin lymphoma; Diffuse large B-cell lymphoma; Mantle cell lymphoma; Acute lymphoblastic leukemia ID WT1 gene; Overexpression; Non-Hodgkin lymphoma; Diffuse large B-cell lymphoma; Mantle cell lymphoma; Acute lymphoblastic leukemia AB The Wilms tumor 1 (WT1) gene has a complex role as a transcriptional regulator, acting as tumor suppressor or oncogene in different malignancies. The prognostic role of its overexpression has been well-studied in leukemias, especially acute myeloid leukemia (AML), but not in lymphomas. For the first time to our knowledge, we present a study demonstrating the correlation of WT1 expression and survival in various non-Hodgkin lymphomas. We also studied the prognostic implications of WT1 overexpression in adult acute lymphoblastic leukemia (ALL). In our sample of 53 patients— 25 with diffuse large B-cell lymphoma (DLBCL), 8 with mantle cell lymphoma (MCL), 9 with peripheral T-cell lymphoma (PTCL), 2 with Burkitt’s lymphoma, 2 with mucosa-associated lymphoid tissue (MALT) lymphoma, and 7 with B-cell ALL—, we measured WT1 mRNA from blood samples by quantitative RT-PCR, and divided the patients into subgroups based on the level of expression. Kaplan–Meier survival curves were drawn and compared using the logrank test. In the sample of DLBCL patients, the difference in overall and disease-free survival between WT1-positive and negative subgroups was significant (p=0.0475 and p=0.0004, respectively), and in a few observed cases, a sudden increase in WT1 expression signified a relapse soon followed by death. Disease-free survival curves in MCL and ALL were similarly suggestive of a potential role played by WT1. In PTCL, though WT1-positivity was detected in 4 out of 9 cases, it did not seem to affect survival. The few cases of MALT and Burkitt’s lymphoma all proved to be WT1-negative. C1 [Ujj, Zsofia] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Buglyo, Gergely] University of Debrecen, Department of Human Genetics, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Udvardy, Miklos] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Vargha, Gyorgy] University of Debrecen, Department of Human Genetics, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Biro, Sandor] University of Debrecen, Department of Human Genetics, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Rejto, Laszlo] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. RP Buglyo, G (reprint author), University of Debrecen, Department of Human Genetics, 4032 Debrecen, Hungary. EM gbuglyo@hotmail.com CR Call KM, Glaser T, Ito CY, Buckler AJ, Pelletier J, Haber DA, Rose EA, Kral A, Yeger H, Lewis WH, 1990, Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms’ tumor locus. Cell 60(3):509–520 Hohenstein P, Hastie ND, 2006, The many facets of the Wilms’ tumour gene, WT1. Hum Mol Genet 15:R196–R201 Haber DA, Sohn RL, Buckler AJ, Pelletier J, Call KM, Housman DE, 1991, Alternative splicing and genomic structure of the Wilms tumor gene WT1. Proc Natl Acad Sci U S A 88(21): 9618–9622 Dallosso AR, Hancock AL, Brown KW, Williams AC, Jackson S, Malik K, 2004, Genomic imprinting at the WT1 gene involves a novel coding transcript, AWT1, that shows deregulation in Wilms’ tumours. Hum Mol Genet 13:405–415 Hastie ND, 2001, Life, sex, and WT1 isoforms – three amino acids can make all the difference. Cell 106:391–394 Morrison AA, Viney RL, Ladomery MR, 2008, The posttranscriptional roles of WT1, a multifunctional zinc-finger protein. Biochim Biophys Acta 1785:55–62 Miwa H, Beran M, Aunders GF, 1992, Expression of the Wilms tumor gene, WT1, in human leukemias. Leukemia 6:405–409 Bergmann L, Miething C, Maurer U, Brieger J, Karakas T, Weidmann E, Hoelzer D, 1997, High levels of Wilms’ tumor gene, wt1)mRNA in acute myeloid leukemias are associated with a worse long-term outcome. Blood 90:1217–1225 GlienkeW, Maute L, Koehl U, Esser R, Milz E, Bergmann L, 2007, Effective treatment of leukemic cell lines with wt1 siRNA. Leukemia 21:2164–2170 Candoni A, Toffoletti E, Gallina R, Simeone E, Chiozzotto M, Volpetti S, Fanin R, 2011, Monitoring of minimal residual disease by quantitative WT1 gene expression following reduced intensity conditioning allogeneic stem cell transplantation in acute myeloid leukemia. Clin Transplant 25(2):308–316 Nowakowska-Kopera A, Sacha T, Florek I, ZawadaM, Czekalska S, Skotnicki AB, 2009, Wilms’ tumor gene 1 expression analysis by real-time quantitative polymerase chain reaction for monitoring of minimal residual disease in acute leukemia. Leuk Lymphoma 50(8): 1326–1332 Boublikova L, Kalinova M, Ryan J, Quinn F, O’Marcaigh A, Smith O, Browne P, Stary J, McCann SR, Trka J, Lawler M, 2006, Wilms’ tumor gene 1, WT1, expression in childhood acute lymphoblastic leukemia: a wide range of WT1 expression levels, its impact on prognosis and minimal residual disease monitoring. Leukemia 20(2):254–263 Busse A, Gokbuget N, Siehl JM, Hoelzer D, Schwartz S, Rietz A, Thiel E, Keilholz U, 2009, Wilms’ tumor gene 1, WT1, expression in subtypes of acute lymphoblastic leukemia, ALL, of adults and impact on clinical outcome. Ann Hematol 88(12):1199–1205 Sadek HA, El-Metnawey WH, Shaheen IA, Korshied MM, Mohamed AS, 2011, Quantitative assessment of Wilms tumor 1, WT1, gene transcripts in Egyptian acute lymphoblastic leukemia patients. J Investig Med 59(8):1258–1262 Drakos E, Rassidakis GZ, Tsioli P, Lai R, Jones D, Medeiros LJ, 2005, Differential expression of WT1 gene product in non- Hodgkin lymphomas. Appl Immunohistochem Mol Morphol 13: 132–137 Mazrouei S, Ziaei A, Tanhaee AP, Keyhanian K, Esmaeili M, Baradaran A, SalehiM(2012, Apoptosis inhibition or inflammation: the role of NAIP protein expression in Hodgkin and non-Hodgkin lymphomas compared to non-neoplastic lymph node. J Inflamm Lond 9:4 Hoelzer D, Thiel E, Loffler H, Bodenstein H, Plaumann L, Buchner T, Urbanitz D, Koch P, Heimpel H, Engelhardt R, 1983, Recruiting patients and results of a preliminary study on the therapy of acute lymphatic leukemia and acute undifferentiated leukemia in adults. Onkologie 6(4):170–174 Wang YF, Yang YL, Gao ZF, Zhou CJ, Gregg X, Shi YF, Wang J, Yang XF, Ke XY, 2012, Clinical and laboratory characteristics of systemic anaplastic large cell lymphoma in Chinese patients. J Hematol Oncol 5:38 Vadasz Z, Shasha-Lavsky H, Nov Y, Bejar J, Lurie M, Tadmor T, Attias D, 2012)Wilms’ tumor gene 1: a possible new proangiogenic factor in hodgkin lymphoma. Appl ImmunohistochemMol Morphol., DOI 10.1097/PAI.0b013e318259852a Gurbuxani S, 2007, WT1 expression in myelodysplastic syndrome. Leuk Lymphoma 48(3):456–457 Perotti D, Mondini P, Giardini R, Ferrari A, Massimino M, Gambirasio F, Pierotti MA, Fossati-Bellani F, Radice P, 1998, No evidence of WT1 involvement in a Burkitt’s lymphoma in a patient with Denys-Drash syndrome. Ann Oncol 9(6):627–631 Tosello V, MansourMR, Barnes K, PaganinM, SulisML, Jenkinson S, Allen CG, Gale RG, Linch DC, Palomero T, Real P,Murty V, Yao X, Richards SM, Goldstone A, Rowe J, Basso G,Wiernik PH, Paietta E, Pieters R,Horstmann M,Meijerink JPP, FerrandoAA(2009)WT1 mutations in T-ALL. Blood 114(5):1038–1045 Heesch S, Goekbuget N, Stroux A, Tanchez JO, Schlee C, Burmeister T, Schwartz S, Blau O, Keilholz U, Busse A, Hoelzer D, Thiel E, Hofmann WK, Baldus CD, 2010, Prognostic implications of mutations and expression of the Wilms tumor 1, WT1, gene in adult acute T-lymphoblastic leukemia. Haematologica 95(6):942–949 Oka Y, Tsuboi A, Oji Y, Kawase I, Sugiyama H, 2008, WT1 peptide vaccine for the treatment of cancer. CurrOpin Immunol 20(2):211–220 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 565 EP 570 DI 10.1007/s12253-013-9729-7 PG 6 ER PT J AU Franchi, A Innocenti, RDD Palomba, A Miligi, L Paiar, F Franzese, C Santucci, M AF Franchi, Alessandro Innocenti, Rossi Degli Duccio Palomba, Annarita Miligi, Lucia Paiar, Fabiola Franzese, Ciro Santucci, Marco TI Low Prevalence of K-RAS, EGF-R and BRAF Mutations in Sinonasal Adenocarcinomas. Implications for Anti-EGFR Treatments SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sinonasal adenocarcinoma; Intestinal type adenocarcinoma; EGFR; KRAS; BRAF ID Sinonasal adenocarcinoma; Intestinal type adenocarcinoma; EGFR; KRAS; BRAF AB We have previously shown that a subset of sinonasal intestinal-type adenocarcinomas (ITAC) shows activation of the epidermal growth factor-receptor (EGFR) pathway. In this study we examine the status of the EGFR, KRAS and BRAF genes in a series of sinonasal intestinal (ITAC) and nonintestinal type adenocarcinomas (non-ITAC). Eighteen ITACs and 12 non-ITACs were studied immunohistochemically for EGFR expression. Point mutations were analyzed for EGFR exons 19 and 21, KRAS exon 2 and BRAF exon 15 by direct sequencing. Non-ITACs showed significantly higher expression of EGFR (p =0.015). Mutation analysis revealed one ITAC with EGFR and one ITAC with KRAS mutation, while two non-ITACs presented mutation of BRAF.We conclude that a subset of sinonasal adenocarcinomas shows overexpression of EGFR, while activating mutations of the signaling cascade downstream of EGFR are rare, suggesting that these tumors could be good candidates for anti-EGFR therapies. C1 [Franchi, Alessandro] AOU Careggi, Department of Surgery and Translational Medicine, Largo Brambilla 3, 50134 Florence, Italy. [Innocenti, Rossi Degli Duccio] University of Florence, Department of Clinical and Experimental MedicineFlorence, Italy. [Palomba, Annarita] Azienda Ospedaliera Universitaria Careggi, Unit of HistopathologyFlorence, Italy. [Miligi, Lucia] ISPO – Cancer Prevention and Research Institute, Unit of Environmental and Occupational EpidemiologyFlorence, Italy. [Paiar, Fabiola] Azienda Ospedaliera Universitaria Careggi, Unit of Radiation OncologyFlorence, Italy. [Franzese, Ciro] Azienda Ospedaliera Universitaria Careggi, Unit of Radiation OncologyFlorence, Italy. [Santucci, Marco] AOU Careggi, Department of Surgery and Translational Medicine, Largo Brambilla 3, 50134 Florence, Italy. RP Franchi, A (reprint author), AOU Careggi, Department of Surgery and Translational Medicine, 50134 Florence, Italy. EM franchi@unifi.it CR Dulguerov P, Jacobsen MS, Allal AS, Lehmann W, Calcaterra T, 2001, Nasal and paranasal sinus carcinoma: are we making progress? A series of 220 patients and a systematic review. Cancer 92: 3012–3029 Turner JH, Reh DD, 2012, Incidence and survival in patients with sinonasal cancer: a historical analysis of population-based data. Head Neck 34:877–885 Franchi A, Miligi L, Palomba A, Giovannetti L, Santucci M, 2011, Sinonasal carcinomas: recent advances in molecular and phenotypic characterization and their clinical implications. Crit Rev Oncol Hematol 79:265–277 Custodio A, Feliu J, 2013, Prognostic and predictive biomarkers for epidermal growth factor receptor-targeted therapy in colorectal cancer: Beyond KRAS mutations. Crit Rev Oncol Hematol 85:45–81 Franchi A, Fondi C, Paglierani M, Pepi M, Gallo O, Santucci M, 2009, Epidermal growth factor receptor expression and gene copy number in sinonasal intestinal type adenocarcinoma. Oral Oncol 45: 835–838 Franchi A, Palomba A, Massi D, Biancalani M, Sardi I, Gallo O, Santucci M, 2006, Low-grade salivary type tubulo-papillary adenocarcinoma of the sinonasal tract. Histopathology 48:881–884 Garcia-Inclan C, Lopez F, Perez-Escuredo J, Cuesta-Albalad MP, Vivanco B, Centeno I, Balbin M, Suarez C, Llorente JL, Hermsen MA, 2012, EGFR status and KRAS/BRAF mutations in intestinaltype sinonasal adenocarcinomas. Cell Oncol 35:443–450 Szablewski V, Solassol J, Poizat F, LarrieuxM, Crampette L, Mange A, Bascoul-Mollevi C, Costes V, 2013, EGFR expression and KRAS and BRAF mutational status in intestinal-type sinonasal adenocarcinoma. Int J Mol Sci 14:5170–5181 Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA, 2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350:2129–2139 Taron M, Ichinose Y, Rosell R, Mok T, Massuti B, Zamora L, Mate JL, Manegold C, Ono M, Queralt C, Jahan T, Sanchez JJ, Sanchez- Ronco M, Hsue V, Jablons D, Sanchez JM, Moran T, 2005, Activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor are associated with improved survival in gefitinib-treated chemorefractory lung adenocarcinoma. Clin Cancer Res 11:5878–5885 Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, LindemanN, Boggon TJ, NaokiK, SasakiH, FujiiY, Eck MJ, Sellers WR, Johnson BE, Meyerson M, 2004, EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304:1497–1500 Wu TT, Barnes L, Bakker A, Swalsky PA, Finkelstein SD, 1996, Kras- 2 and p53 genotyping of intestinal-type adenocarcinoma of the nasal cavity and paranasal sinuses. Mod Pathol 9:199–204 Saber AT, Nielsen LR, Dictor M, Hagmar L, Mikoczy Z, Wallin H, 1998, K-ras mutations in sinonasal adenocarcinomas in patients occupationally exposed to wood or leather dust. Cancer Lett 126: 59–65 Yom SS, Rashid A, Rosenthal DI, Elliott DD, Hanna EY,Weber RS, El-Naggar AK, 2005, Genetic analysis of sinonasal adenocarcinoma phenotypes: distinct alterations of histogenetic significance. Mod Pathol 18:315–319 FrattiniM, Perrone F, Suardi S, Balestra D, Caramuta S, Colombo F, Licitra L, Cantu G, PierottiMA, Pilotti S, 2006, Phenotype-genotype correlation: challenge of intestinal-type adenocarcinoma of the nasal cavity and paranasal sinuses. Head Neck 28:909–915 Bornholdt J, Hansen J, Steiniche T, Dictor M, Antonsen A,Wolff H, Schlunssen V, Holmila R, Luce D, Vogel U, Husgafvel-Pursiainen K, Wallin H, 2008, K-ras mutations in sinonasal cancers in relation to wood dust exposure. BMC Cancer 8:53 Lopez F, Garcia Inclan C, Perez-Escuredo J, AlvarezMarcos C, Scola B, Suarez C, Llorente JL, Hermsen MA, 2012, KRAS and BRAF mutations in sinonasal cancer. Oral Oncol 48:692–697 Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, De Dosso S,Mazzucchelli L, Frattini M, Siena S, Bardelli A, 2008, Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol 26: 5705–5712 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 571 EP 579 DI 10.1007/s12253-013-9730-1 PG 9 ER PT J AU Madaras, L Kovacs, AK Szasz, MA Kenessey, I Tokes, AM Szekely, B Baranyak, Zs Kiss, O Dank, M Kulka, J AF Madaras, Lilla Kovacs, Attila Kristof Szasz, Marcell Attila Kenessey, Istvan Tokes, Anna-Maria Szekely, Borbala Baranyak, Zsuzsanna Kiss, Orsolya Dank, Magdolna Kulka, Janina TI Clinicopathological Features and Prognosis of Pregnancy Associated Breast Cancer – A Matched Case Control Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Breast cancer; Pregnancy associated breast cancer; Pregnancy; Postpartum; Young women ID Breast cancer; Pregnancy associated breast cancer; Pregnancy; Postpartum; Young women AB Pregnancy Associated Breast Cancer (PABC) manifests during pregnancy or within a year following delivery. We sought to investigate differences in management, outcome, clinical, histopathology and immunohistochemistry (IHC) characteristics of PABC and matched controls in a retrospective case control study. PABC and control patients were selected from breast cancer cases of women ≤45 years, diagnosed in the 2nd Department of Pathology, Semmelweis University, Budapest, Hungary between 1998 and 2012. Histopathology information on tumor type, grade, size, T, N, lympho-vascular invasion (LVI), Nottingham Prognostic Index (NPI), associated in situ lesions and IHC charcteristics: ER, PgR, HER2, Ki67, p53 were recorded, IHC-based subtype was assessed, clinical, management and outcome data were analysed. Thirty-one breast cancer cases were pregnancy related. Clinical management data did not differ in cases and controls. Histopathology of disease at presentation was not significantly different, but NPI assessed the PABC group as having poor, whereas controls as having intermediate prognosis. Associated in situ lesion was more often high grade Extensive Intraductal Carcinoma Component (EIC) in PABC. Triple negative and LuminalB prol tumors predominated in PABC. Disease-free and overall survival was inferior compared to controls. PABC patients with LuminalB prol and Triple negative tumors had inferior outcomes. On multivariate analysis inferior prognosis of PABC was associated with pregnancy. Our study has demonstrated inferior outcome of PABC. Difference in tumor biology is reflected by the predominance of triple negative and LuminalB tumors in PABC. The strength of the study is the analysis of complete pathology and IHC data. C1 [Madaras, Lilla] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. [Kovacs, Attila Kristof] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. [Szasz, Marcell Attila] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. [Kenessey, Istvan] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. [Tokes, Anna-Maria] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. [Szekely, Borbala] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. [Baranyak, Zsuzsanna] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. [Kiss, Orsolya] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. [Dank, Magdolna] Semmelweis University, 1st Department of Internal Medicine, Tomo u 25-29, 1083 Budapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93., 1091 Budapest, Hungary. RP Madaras, L (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM lilla.madaras@gmail.com CR Johansson ALV, Andersson TM-L, Hsieh C-C, Cnattingius S, Lambe M, 2011, Increased mortality in women with breast cancer detected during pregnancy and different periods postpartum. Cancer Epidemiol Biomark Prev 20(9):1865–1872., DOI 10.1158/1055- 9965.epi-11-0515 Woo Jc YTHTC, 2003, Breast cancer in pregnancy: a literature review. Arch Surg 138(1):91–98., DOI 10.1001/archsurg.138.1.91 Lyons T, Schedin P, Borges V, 2009, Pregnancy and breast cancer: when they collide. J Mammary Gland Biol Neoplasia 14(2):87–98., DOI 10.1007/s10911-009-9119-7 Liu Q, Wuu J, Lambe M, Hsieh S-F, Ekbom A, Hsieh C-C, 2002, Transient increase in breast cancer risk after giving birth: postpartum period with the highest risk, Sweden). Cancer Causes Control 13(4): 299–305., DOI 10.1023/a:1015287208222 Lambe M, Hsieh C, Trichopoulos D, Ekbom A, Pavia M, Adami HO, 1994, Transient increase in the risk of breast cancer after giving birth. New England J Med 331(1):5–9., DOI 10.1056/NEJM19940 7073310102 Nugent P, Connell TXO, 1985, Breast cancer and pregnancy. Arch Surg 120(11):1221–1224 Ali SA, Gupta S, Sehgal R, Vogel V, 2012, Survival outcomes in pregnancy associated breast cancer: a retrospective case control study. Breast J 18(2):139–144., DOI 10.1111/j.1524-4741.2011. 01201.x MoreiraWB, Brandao EC, Soares AN, Lucena CEM, Antunes CMF, 2010, Prognosis for patients diagnosed with pregnancy-associated breast cancer: a paired case–control study. Sao PauloMed J 128:119– 124 Bonnier P, Romain S, Dilhuydy JM, Bonichon F, Julien JP, Charpin C, Lejeune C,Martin PM, Piana L, 1997, Influence of pregnancy on the outcome of breast cancer: a case–control study. Int J Cancer 72(5):720–727., DOI 10.1002/(sici)1097-0215(19970904)72:5<720:: aid-ijc3>3.0.co;2-u Azim HA, Santoro L, Russell-Edu W, Pentheroudakis G, Pavlidis N, Peccatori FA, 2012, Prognosis of pregnancyassociated breast cancer: a meta-analysis of 30 studies. Cancer Treat Rev 38(7):834–842 Anders CK, Hsu DS, Broadwater G, Acharya CR, Foekens JA, Zhang Y, Wang Y, Marcom PK, Marks JR, Febbo PG, Nevins JR, Potti A, Blackwell KL, 2008, Young age at diagnosis correlates with worse prognosis and defines a subset of breast cancers with shared patterns of gene expression. J Clin Oncol 26(20):3324–3330., DOI 10. 1200/jco.2007.14.2471 Middleton LP, AminM, GwynK, Theriault R, Sahin A, 2003, Breast carcinoma in pregnant women. Cancer 98(5):1055–1060., DOI 10. 1002/cncr.11614 Hahn KME, Johnson PH, Gordon N, Kuerer H, Middleton L, Ramirez M, Yang W, Perkins G, Hortobagyi GN, Theriault RL, 2006, Treatment of pregnant breast cancer patients and outcomes of children exposed to chemotherapy in utero. Cancer 107(6):1219– 1226., DOI 10.1002/cncr.22081 Halaska MJ, Pentheroudakis G, Strnad P, Stankusova H, Chod J, Robova H, Petruzelka L, Rob L, Pavlidis N, 2009, Presentation, management and outcome of 32 patients with pregnancy-associated breast cancer: a matched controlled study. Breast J 15(5):461–467., DOI 10.1111/j.1524-4741.2009.00760.x Azim HA Jr, Botteri E, Renne G, Dell’Orto P, Rotmensz N, Gentilini O, Sangalli C, Pruneri G, Di Nubila B, Locatelli M, Sotiriou C, Piccart M, Goldhirsch A, Viale G, Peccatori FA, 2012, The biological features and prognosis of breast cancer diagnosed during pregnancy: a case–control study. Acta Oncol 51(5):653–661., DOI 10. 3109/0284186X.2011.636069 Murphy CG,Mallam D, Stein S, Patil S, Howard J, Sklarin N, Hudis CA, GemignaniML, Seidman AD, 2012, Current or recent pregnancy is associated with adverse pathologic features but not impaired survival in early breast cancer. Cancer 118(13):3254–3259., DOI 10. 1002/cncr.26654 Genin A-S, Lesieur B, Gligorov J, Antoine M, Selleret L, Rouzier R, 2012, Pregnancy-associated breast cancers: do they differ from other breast cancers in young women? Breast Edinb Scotl 21(4):550–555 Pilewskie M, Gorodinsky P, Fought A, Hansen N, Bethke K, Jeruss J, Scholtens D, Khan S, 2012, Association between recency of last pregnancy and biologic subtype of breast cancer. Ann Surg Oncol 19(4):1167–1173., DOI 10.1245/s10434- 011-2104-6 Reed W, Hannisdal E, Skovlund E, Thoresen S, Lilleng P, Nesland JM, 2003, Pregnancy and breast cancer: a population-based study. Virchows Archiv 443(1):44–50., DOI 10.1007/s00428-003-0817-z Hammond MEH, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, Fitzgibbons PL, Francis G, Goldstein NS, Hayes M, Hicks DG, Lester S, Love R, Mangu PB, McShane L, Miller K, Osborne CK, Paik S, Perlmutter J, Rhodes A, Sasano H, Schwartz JN, Sweep FCG, Taube S, Torlakovic EE, Valenstein P, Viale G, Visscher D, Wheeler T, Williams RB, Wittliff JL, Wolff AC, 2010, American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for Immunohistochemical Testing of Estrogen and Progesterone Receptors in breast cancer. J Clin Oncol 28(16):2784–2795., DOI 10.1200/jco. 2009.25.6529 Wolff AC, Hammond MEH, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, McShane LM, Paik S, Pegram MD, Perez EA, Press MF, Rhodes A, Sturgeon C, Taube SE, Tubbs R, Vance GH, van de Vijver M, Wheeler TM, Hayes DF, 2007, American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for Human Epidermal Growth Factor Receptor 2 testing in breast cancer. Arch Pathol Lab Med 131(1):18–43., DOI 10. 1043/1543-2165(2007)131[18:asocco]2.0.co;2 CheangMCU, Chia SK, Voduc D, Gao D, Leung S, Snider J,Watson M, Davies S, Bernard PS, Parker JS, PerouCM, EllisMJ,Nielsen TO, 2009, Ki67 Index, HER2 status, and prognosis of patients with luminal B breast cancer. J Nat Cancer Inst 101(10):736–750., DOI 10.1093/jnci/djp082 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn H-J, Members P, 2011, Strategies for subtypes—dealing with the diversity of breast cancer: highlights of the St Gallen International Expert Consensus on the primary therapy of early breast cancer 2011. Ann Oncol 22(8):1736–1747., DOI 10.1093/ annonc/mdr304 Beadle BM, Woodward WA, Middleton LP, Tereffe W, Strom EA, Litton JK, Meric-Bernstam F, Theriault RL, Buchholz TA, Perkins GH, 2009, The impact of pregnancy on breast cancer outcomes in women ≤35 years. Cancer 115(6):1174–1184., DOI 10.1002/cncr.24165 Azim HA Jr, Santoro L, Russell-Edu W, Pentheroudakis G, Pavlidis N, Peccatori FA, 2012, Prognosis of pregnancy-associated breast cancer: a meta-analysis of 30 studies. Cancer Treat Rev 38(7):834– 842., DOI 10.1016/j.ctrv.2012.06.004 Guinee F, Hess KR, Taylor SH, Olsson H, Moller T, Fahey T, Gladikov JV, van den Blink JW, Bonichon F, Dische S, Yates JW, Cleton F, 1994, Effect of pregnancy on prognosis for young women with breast cancer. Lancet 343(8913):1587–1589 Kollias J,Murphy C, Elston C, Ellis I, Robertson J, Blamey R, 1999, The prognosis of small primary breast cancers. Eur J Cancer 35(6): 908–912 Albergaria A, Ricardo S, Milanezi F, Carneiro V, Amendoeira I, Vieira D, Cameselle-Teijeiro J, Schmitt F, 2011, Nottingham prognostic index in triple-negative breast cancer: a reliable prognostic tool? BMC Cancer 11(1):299 Collins LC, Achacoso N, Nekhlyudov L, Fletcher SW, Haque R, Quesenberry CPJ, Puligandla B, Alshak NS, Goldstein LC, Gown AM, Schnitt SJ, Habel LA, 2009, Relationship Between Clinical and Pathologic Features of Ductal Carcinoma In Situ and Patient Age: An Analysis of 657 Patients. The American Journal of Surgical Pathology 33, 12):1802–1808 Schnitt SJ, 2010, Local outcomes in ductal carcinoma in situ based on patient and tumor characteristics. JNCI Monographs 2010(41): 158–161., DOI 10.1093/jncimonographs/lgq031 Goldstein NS, Vicini FA, Kestin LL, Thomas M, 2000, Differences in the pathologic features of ductal carcinoma in situ of the breast based on patient age. Cancer 88(11):2553– 2560., DOI 10.1002/1097-0142(20000601)88:11<2553::AIDCNCR18> 3.0.CO;2-V Vincent-Salomon A, Lucchesi C, Gruel N, Raynal V, Pierron G, Goudefroye R, Reyal F, Radvanyi F, Salmon R, Thiery J-P, Sastre- Garau X, Sigal-Zafrani B, Fourquet A, Delattre O, 2008, Integrated genomic and transcriptomic analysis of ductal carcinoma in situ of the breast. Clin Cancer Res 14(7):1956–1965., DOI 10.1158/1078- 0432.ccr-07-1465 Lopez-Garcia MA, Geyer FC, Lacroix-Triki M, Marchio C, Reis- Filho JS, 2010, Breast cancer precursors revisited:molecular features and progression pathways. Histopathology 57(2):171–192., DOI 10. 1111/j.1365-2559.2010.03568.x Fornetti J, Martinson H, Borges V, Schedin P, 2012, Emerging targets for the prevention of pregnancy-associated breast cancer. Cell Cycle 11(4):639–640 O’Brien J, Schedin P, 2009, Macrophages in breast cancer: do involution macrophages account for the poor prognosis of pregnancy-associated breast cancer? J Mammary Gland Biol Neoplasia 14(2):145–157., DOI 10.1007/s10911-009-9118-8 O’Brien J, Lyons T,Monks J, Lucia MS,Wilson RS, Hines L, Y-g M, Borges V, Schedin P, 2010, Alternatively activated macrophages and collagen remodeling characterize the postpartum involuting mammary gland across species. Am J Pathol 176(3):1241–1255 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 581 EP 590 DI 10.1007/s12253-013-9735-9 PG 10 ER PT J AU Jancsik, AV Gelencser, G Maasz, G Schmidt, J Molnar, AG Wittmann, I Olasz, L Mark, L AF Jancsik, A Veronika Gelencser, Gabor Maasz, Gabor Schmidt, Janos Molnar, A Gergo Wittmann, Istvan Olasz, Lajos Mark, Laszlo TI Salivary Proteomic Analysis of Diabetic Patients for Possible Oral Squamous Cell Carcinoma Biomarkers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Diabetes mellitus; MALDI TOF/TOF mass spectrometry; Oral squamous cell carcinoma; Salivary biomarkers; Type-2 diabetes ID Diabetes mellitus; MALDI TOF/TOF mass spectrometry; Oral squamous cell carcinoma; Salivary biomarkers; Type-2 diabetes AB Since oral squamous cell carcinoma (OSCC) is one of the most important causes of death worldwide, the prevention and early detection plays a crucial role. Recent epidemiological studies have incriminated diabetes as a risk factor for the development of OSCC, as well as oral premalignant lesions. As for the last 20 years diabetes and oral squamous cell carcinoma rates have been increasing rapidly, therefore a reliable detection method of major saliva proteins as possible biomarkers for OSCC is of key priority. In this study we collected whole saliva samples from patients with diabetes and from healthy subjects. To reduce the risk of failure and to keep the investigation good reproducible, we proposed an examination and saliva collecting technique. The proteins were analyzed using SDS-PAGE and MALDI TOF/TOF mass spectrometry. Our findings show that the expression of Annexin A8, Peroxiredoxin-2 and Tyrosine kinase is elevated by patients having diabetes. All these proteins have been previously described in cancer saliva samples also in OSCC. Our current findings showed that testing saliva may be an effective and reliable method for detecting oral cancer in early stages. C1 [Jancsik, A Veronika] University of Pecs, Department of Oral and Maxillofacial Surgery, 5 Dischka G. str, 7620 Pecs, Hungary. [Gelencser, Gabor] University of Pecs, Department of Oral and Maxillofacial Surgery, 5 Dischka G. str, 7620 Pecs, Hungary. [Maasz, Gabor] University of Pecs, Institute of Biochemistry and Medical Chemistry, 12 Szigeti str, 7624 Pecs, Hungary. [Schmidt, Janos] University of Pecs, Institute of Biochemistry and Medical Chemistry, 12 Szigeti str, 7624 Pecs, Hungary. [Molnar, A Gergo] University of Pecs, 2nd Department of Medicine and Nephrological Center, 1 Pacsirta str, 7624 Pecs, Hungary. [Wittmann, Istvan] University of Pecs, 2nd Department of Medicine and Nephrological Center, 1 Pacsirta str, 7624 Pecs, Hungary. [Olasz, Lajos] University of Pecs, Department of Oral and Maxillofacial Surgery, 5 Dischka G. str, 7620 Pecs, Hungary. [Mark, Laszlo] University of Pecs, Institute of Biochemistry and Medical Chemistry, 12 Szigeti str, 7624 Pecs, Hungary. RP Mark, L (reprint author), University of Pecs, Institute of Biochemistry and Medical Chemistry, 7624 Pecs, Hungary. EM laszlo.mark@aok.pte.hu CR Wong DT, 2006, Salivary diagnostics powered by nanotechnologies, proteomics and genomics. J Am Dent Ass 137:313–321 Segal A, Wong DT, 2008, Salivary diagnostics: enhancing disease detection and making medicine better. Eur J Dent Educ 12(suppl): 22S–29S Szanto I, Mark L, Bona A, Maasz G, Sandor B, Gelencser G, Turi Z, Gallyas F Jr, 2012, High-throughput screening of saliva for early detection of oral cancer: a pilot study. Technol Cancer Res Treat 11(2):181–188 Jarai T, Maasz G, Burian A, Bona A, Jambor E, Gerlinger I, Mark L, 2012, Mass spectrometry-based salivary proteomics for the discovery of head and neck squamous cell carcinoma. Pathol Oncol Res 18(3):623–628 Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Argiris A, Karamouzis MV, Raben D et al, 2008, Head and neck cancer. Lancet 371:1695–1709 Decker J, Glodstein JC, 1982, Risk factors in head and neck cancer. N Engl J Med 306:1151–1155 Hintao J, Teanpaisan R, Chongsuvivatwong V et al, 2007, The microbiological profiles of saliva, supragingival and subgingival plaque and dental caries in adults with and without type 2 diabetes mellitus. Oral Microbiol Immunol 22:175–181 Lamster IB, Lalla E, Borgnakke WS, Taylor GW, 2008, The relationship between oral health and diabetes mellitus. J Am Dent Ass 139:19S–24S American Academy of Periodontology, 1996, Position paper: diabetes and periodontal diseases. J Periodontol 67:166–176 Negrato CA, Tarzia O, 2010, Buccal alterations in diabetes mellitus. Diabetol Metab Syndr 15:2–3 Lu H, Ouyang W, Huang C, 2006, Inflammation, a key event in cancer development. Mol Cancer Res 4(4):221–233 Ujpal M, Matos O, Bidok G, Somogyi A, Szabo G, Suba Z, 2004, Diabetes and oral tumors in Hungary. Diabetes Care 27:770–774 Albrecht M, Banoczy J, Dinya E, Tamas G Jr, 1992, Occurence of oral leukoplakia and lichen planus in diabetes mellitus. J Oral Path Med 21:364–365 NavazeshM(1993)Methods for collecting saliva. Ann NYAcad Sci 694:72–77 Laemmli UK, 1970, Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227: 680–685 Vairaktaris E, 2004, Diabetes and oral oncogenesis. Anticancer Res 27:4185–4194 Farnaes L, Ditzel HJ, 2003, Dissecting the cellular functions of annexin XI using recombinant human annexin XI-specific autoantibodies cloned by phage display. J Biol Chem 278(35): 33120–33126 Tomas A, Futter C, Moss SE, 2004, Annexin 11 is required for midbody formation and completion of the terminal phase of cytokinesis. J Cell Biol 165(6):813–822 Rand JH, 2000, The annexinopathies: a new category of diseases. Biochim Biophys Acta 1498:169–173 Paweletz CP, Ornstein DK, Roth MJ, Bichsel VE, Gillespie JW, Calvert VS, Vocke CD, Hewitt SM, Duray PH, Herring J, Wang QH, Hu N, Linehan WM, Taylor PR, Liotta LA, Emmert-Buck MR, Petricoin EF, 2000, Loss of annexin 1 correlates with early onset of tumorigenesis in esophageal and prostate carcinoma. Cancer Res 60:6293–6297 Wood ZA, Poole LB, Karplus PA, 2003, Peroxiredoxin evolution and the regulation of hydrogen peroxide signaling. Science 300:650– 653 Lu Y, Liu J, Chengzhao L, Wang H, Jiang Y, Wang Y, Yang P, He F, 2010, Peroxiredoxin 2: a potential biomarker for early diagnosis of Hepatitis B Virus related liver fibrosis identified by proteomic analysis of the plasma. BMC Gastroenterol 10:115 Hanks SK, Quinn AM, Hunter T, 1998, The protein kinase family: conserved features and deduced phylogeny of the catalytic domains. Science 241(4861):42–52 Weinberg RA, 2007, The biology of cancer. New York: Garland Science, Taylor & Francis Group, LLC. pp 757–759 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 591 EP 595 DI 10.1007/s12253-013-9736-8 PG 5 ER PT J AU Forma, E Wojcik-Krowiranda, K Jozwiak, P Szymczyk, A Bienkiewicz, A Brys, M Krzeslak, A AF Forma, Ewa Wojcik-Krowiranda, Katarzyna Jozwiak, Pawel Szymczyk, Agnieszka Bienkiewicz, Andrzej Brys, Magdalena Krzeslak, Anna TI Topoisomerase IIβ Binding Protein 1 c.*229C>T (rs115160714) Gene Polymorphism and Endometrial Cancer Risk SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Topoisomerase IIβ binding protein 1; Polymorphism; Genetic variation; Endometrial cancer ID Topoisomerase IIβ binding protein 1; Polymorphism; Genetic variation; Endometrial cancer AB TopBP1 (topoisomerase IIβ binding protein 1) protein is involved in DNA replication, DNA damage checkpoint response and transcriptional regulation. In this study we investigated whether alterations in the TopBP1 gene can influence the risk of endometrial cancer. We examined the association between five single nucleotide polymorphisms (rs185903567, rs116645643, rs115160714, rs116195487, and rs112843513) located in the 3′UTR region of the TopBP1 gene and endometrial cancer risk as well as allelespecific gene expression. One hundred twenty-one endometrial cancer patients were genotyped for these SNPs. Allelespecific TopBP1 mRNA and protein expressions were determined by real time PCR and western blotting methods, respectively. Only one SNP (rs115160714) showed an association with endometrial cancer. Compared to homozygous common allele carriers, heterozygous for the T variant had significantly increased risk of endometrial cancer [adjusted odds ratio (OR)=5.59, 95 % confidence interval (CI): 1.96–15.91, p=0.0003]. Mean TopBP1 mRNA and protein expression were higher in the individuals with the CT genotype. There was a significant association between the rs115160714 and tumor grade and FIGO classification. Most carriers of minor allele had a high grade tumors (G3) classified as FIGO III/IV. The results of our study raise a possibility that a genetic variation of TopBP1 may be implicated in the etiology of endometrial cancer. C1 [Forma, Ewa] University of Lodz, Department of Cytobiochemistry, Pomorska 141/143, 90-236 Lodz, Poland. [Wojcik-Krowiranda, Katarzyna] Medical University of Lodz, Department of Gynecological Oncology, Pabianicka 62, 93-509 Lodz, Poland. [Jozwiak, Pawel] University of Lodz, Department of Cytobiochemistry, Pomorska 141/143, 90-236 Lodz, Poland. [Szymczyk, Agnieszka] University of Lodz, Department of Cytobiochemistry, Pomorska 141/143, 90-236 Lodz, Poland. [Bienkiewicz, Andrzej] Medical University of Lodz, Department of Gynecological Oncology, Pabianicka 62, 93-509 Lodz, Poland. [Brys, Magdalena] University of Lodz, Department of Cytobiochemistry, Pomorska 141/143, 90-236 Lodz, Poland. [Krzeslak, Anna] University of Lodz, Department of Cytobiochemistry, Pomorska 141/143, 90-236 Lodz, Poland. RP Krzeslak, A (reprint author), University of Lodz, Department of Cytobiochemistry, 90-236 Lodz, Poland. EM zreg@biol.uni.lodz.pl CR Amant F, Moerman P, Neven P, Timmerman D, Van Limbergen E, Vergote I, 2005, Endometrial cancer. Lancet 366:491–505 Segev Y, Iqbal J, Lubinski J et al, 2013, Hereditary Breast Cancer Study Group. The incidence of endometrial cancer in women with BRCA1 and BRCA2 mutations: an international prospective cohort study. Gynecol Oncol., DOI 10.1016/j.ygyno.2013.03.027 Forma E, Brys M, Krajewska W, 2011, TopBP1 in DNA damage response. In: Kruman I, ed, DNA repair/book 4. INTECH Open Access, Rijeka, pp 281–304 Glover JNM, 2006, Insights into the molecular basis of human hereditary breast cancer from studies of the BRCA1 BRCT domain. Fam Cancer 5:89–93 Sokka M, Parkkinen S, Pospiech H, Syvaoja JE, 2010, Function of TopBP1 in genome stability. Subcell Biochem 50:119–141 RodriguezMC, Songyang Z, 2008, BRCT domains: phosphopeptide binding and signaling modules. Front Biosci 13:5905–5915 Smits VA, Warmerdam DO, Martin Y, Freire R, 2010, Mechanisms of ATR-mediated checkpoint signalling. Front Biosci 15:840–853 Forma E, Brzezianska E, Krzeslak A et al, 2012, Association between the c.*229C>T polymorphism of the topoisomerase IIβ binding protein 1, TopBP1, gene and breast cancer. Mol Biol Rep 40:3493–3502 Bilbao C, Ramirez R, Rodriguez G et al, 2010, Double strand break repair components are frequent targets of microsatellite instability in endometrial cancer. Eur J Cancer 46:2821–2827 Zighelboim I, Schmidt AP, Gao F et al, 2009, ATR mutation in endometrioid endometrial cancer is associated with poor clinical outcomes. J Clin Oncol 27:3091–3096 Forma E, Krzeslak A, Bernaciak M, Romanowicz-Makowska H, Brys M, 2012, Expression of TopBP1 in hereditary breast cancer. Mol Biol Rep 39:7795–7804 Liu K, Bellam N, Lin HYet al, 2009, Regulation of p53 by TopBP1: a potential mechanism for p53 inactivation in cancer. Mol Cell Biol 29:2673–2693 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 597 EP 602 DI 10.1007/s12253-013-9737-7 PG 6 ER PT J AU Feng, L Tao, L Dawei, H Xuliang, L Xiaodong, L AF Feng, Liu Tao, Lin Dawei, He Xuliang, Li Xiaodong, Luo TI HIF-1α Expression Correlates with Cellular Apoptosis, Angiogenesis and Clinical Prognosis in Rectal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hypoxia-inducible factor 1 alpha (HIF-1α); Rectal neoplasm; Apoptosis; Neovascularization ID Hypoxia-inducible factor 1 alpha (HIF-1α); Rectal neoplasm; Apoptosis; Neovascularization AB Regional hypoxia caused by accelerated cell proliferation and overgrowth is an important characteristic of neoplasm. Hypoxia can cause a series of changes in gene transcription and protein expression, thereby not only inducing tumor cell resistance to radiotherapy and chemotherapy but also promoting tumor invasion and metastasis. This study aimed to investigate the relationship between HIF-1α expression and cellular apoptosis, angiogenesis and clinical prognosis in rectal carcinoma. In 113 rectal carcinoma cases, cellular apoptosis was analyzed by the in situ terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay, whereas the levels of HIF-1α expression, VEGF expression, microvessel density (MVD) and lymphatic vessel density(LVD) were examined by immunohistochemical staining. HIF-1 expression was detected in 67 of 113 rectal carcinoma cases (59.3 %). A positive correlation was found among HIF-1α expression, cellular apoptosis and angiogenesis. The 5-year survival rate in the HIF-1α-negative group was significantly higher than that in the HIF-1α- positive group (81.34 % versus 50 %, P<0.05). According to the Cox regression analysis, HIF-1α expression, VEGF expression and cellular apoptosis index were independent risk factors for clinical prognosis in rectal carcinoma. Aberrant HIF-1α expression correlates with apoptosis inhibition, angiogenesis and poor prognosis in rectal carcinoma. C1 [Feng, Liu] Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), the Children’s Hospital, Chongqing Medical University, Department of Urinary Surgery, 400014 Chongqing, China. [Tao, Lin] Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), the Children’s Hospital, Chongqing Medical University, Department of Urinary Surgery, 400014 Chongqing, China. [Dawei, He] Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), the Children’s Hospital, Chongqing Medical University, Department of Urinary Surgery, 400014 Chongqing, China. [Xuliang, Li] Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), the Children’s Hospital, Chongqing Medical University, Department of Urinary Surgery, 400014 Chongqing, China. [Xiaodong, Luo] Chongqing Medical University, the Second Affiliated Hospital, Department of Obstetrics and Gynecology, 400010 Chongqing, China. RP Xiaodong, L (reprint author), Chongqing Medical University, the Second Affiliated Hospital, Department of Obstetrics and Gynecology, 400010 Chongqing, China. EM cchliuf@163.com CR Gupta SC, Kim JH, Prasad S et al, 2010, Regulation of survival, proliferation, invasion, angiogenesis, and metastasis of tumor cells through modulation of inflammatory pathways by nutraceuticals. Cancer Metastasis Rev 29(3):405–434 de Krijger I, Mekenkamp LJ, Punt CJ et al, 2011, MicroRNAs in colorectal cancer metastasis. J Pathol 224(4):438–447 Arvelo F, Cotte C, 2009, Hypoxia in cancer malignity. Invest Clin 50(4):529–546 Greijer AE, van der Groep P, Kemming D et al, 2005, Up-regulation of gene expression by hypoxia is medi-ated predominantly by hypoxia-inducible factor 1, HIF-1). J Pathol 206:291–304 Belozerov VE, Van Meir EG, 2005, Hypoxia inducible factor-1: a novel target for cancer therapy. Anticancer Drugs 16(9):901–909 Mucaj V, Shay JE, SimonMC, 2012, Effects of hypoxia and HIFs on cancer metabolism. Int J Hematol 95(5):464–670 Semenza GL, 2000, HIF-1: mediator of physiological and pathophysiological responses to hypoxia. J Appl Physiol 88(4):1474–1480 Yeung SJ, Pan J, Lee MH, 2008, Roles of p53, MYC and HIF-1 in regulating glycolysis - the seventh hallmark of cancer. Cell Mol Life Sci 65(24):3981–3999 Rapisarda A, Melillo G, 2012, Role of the VEGF/VEGFR axis in cancer biology and therapy. Adv Cancer Res 114:237–267 Kerbel RS, 2008, Tumor angiogenesis. N Engl JMed 358(19):2039– 2049 Merritt WM, Sood AK, 2007, Markers of angiogenesis in ovarian cancer. Dis Markers 23(5–6):419–431 Menrad H, Werno C, Schmid T et al, 2010, Roles of hypoxiainducible factor-1alpha, HIF-1alpha, versus HIF-2alpha in the survival of hepatocellular tumor spheroids. Hepatology 51(6):2183– 2192 Ardyanto TD, Osaki M, Nagahama Y et al, 2008, Down-regulation of cobalt-induced HIF-1alpha expression correlates with cell proliferation and apoptosis in human gastric carcinoma cells. Oncol Rep 19(2):339–343 Hao X, Du M, Bishop AE et al, 1998, Imbalance between proliferation and apoptosis in development of colorectal carcinoma. Virchows Arch 433:523–527 Benchabane H, Ahmed Y, 2009, The adenomatous polyposis coli tumor suppressor and Wnt signaling in the regulation of apoptosis. Adv Exp Med Biol 656:75–84 Lampropoulos P, Zizi-Sermpetzoglou A, Rizos S et al, 2012, TGFbeta signalling in colon carcinogenesis. Cancer Lett 314(1):1–7 Liu W, Shen SM, Zhao XY et al, 2012, Targeted genes and interacting proteins of hypoxia inducible factor-1. Int J Biochem Mol Biol 3(2):165–178 Zhong H, DeMarzo AM, Laughner E et al, 1999, Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res 59:5830–5835 Koh MY, Spivak-Kroizman TR, Powis G, 2010, HIF-1alpha and cancer therapy. Recent Results Cancer Res 180:15–34 Simiantonaki N, Taxeidis M, Jayasinghe C et al, 2008, Hypoxiainducible factor 1 alpha expression increases during colorectal carcinogenesis and tumor progression. BMC Cancer 8:320 Kotch LE, Iyer NV, Laughner E et al, 1999, Defective vascularization of HIF-1alpha-null embryos is not associated with VEGF deficiency but with mesenchymal cell death. Dev Biol 209:254–267 Tsuzuki Y, Fukumura D, Oosthuyse B et al, 2000, Vascular endothelial growth factor, VEGF, modulation by targeting hypoxia-inducible factor-1alpha–> hypoxia re-sponse element–> VEGF cascade differentially regulates vascular response and growth rate in tumors. Cancer Res 60:6248–6252 Rey S, Semenza GL, 2010, Hypoxia-inducible factor-1-dependent mechanisms of vascularization and vascular remodelling. Cardiovasc Res 86(2):236–242 Duffy JP, Eibl G, Reber HA et al, 2003, Influence of hypoxia and neoangiogenesis on the growth of pancreatic cancer.Mol Cancer 2:12 Suzuki H, Tomida A, Tsuruo T, 2001, Dephosphorylated hypoxiainducible factor 1alpha as a mediator of p53-dependent apoptosis during hypoxia. Oncogene 20(41):5779–5788 Piret JP, Mottet D, Raes M et al, 2002, Is HIF-1alpha a pro- or an anti-apoptotic protein? Biochem Pharmacol 64(5–6):889–892 Price DJ,Miralem T, Jiang S et al, 2001, Role of vascular endothelial growth factor in the stimulation of cellular invasion and signaling of breast cancer cells. Cell Growth Differ 12(3):129–135 Bos R, van Diest PJ, van der Groep P et al, 2004, Expression of hypoxia-inducible factor-1alpha and cell cycle proteins in invasive breast cancer are estrogen receptor related. Breast Cancer Res 6(4): R450–R459 Nagy JA, Vasile E, Feng D et al, 2002, Vascular permeability factor/ vascular endothelial growth factor induces lymphangiogenesis as well as angiogenesis. J Exp Med 196(11):1497–1506 Mohammed RA, Green A, El-Shikh S et al, 2007, Prognostic significance of vascular endothelial cell growth factors -A, -C and -D in breast cancer and their relationship with angio- and lymphangiogenesis. Br J Cancer 96(7):1092–1100 Gao Y, ZhongWX,Mu DB et al, 2008, Distributions of angiogenesis and lymphangiogenesis in gastrointestinal intramucosal tumors. Ann Surg Oncol 15(4):1117–1123 Lee S, Choi I, Hong YK, 2010, Heterogeneity and plasticity of lymphatic endothelial cells. Semin Thromb Hemost 36(3):352–361 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 603 EP 610 DI 10.1007/s12253-013-9738-6 PG 8 ER PT J AU Wang, Y Zheng, T AF Wang, Yonggang Zheng, Tianying TI Screening of Hub Genes and Pathways in Colorectal Cancer with Microarray Technology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Pathway interaction network; Protein-protein interaction networksz analysis; p53 signaling pathway ID Colorectal cancer; Pathway interaction network; Protein-protein interaction networksz analysis; p53 signaling pathway AB Here we intend to identify key genes and pathways in the pathogenesis of colorectal cancer (CRC) through analyzing microarray data with bioinformatic tools. The gene expression profile dataset GSE23878 was downloaded from Gene Expression Omnibus and differentially expressed genes (DEGs) were screened out using Student’s t-test. GO function and KEGG pathway enrichment analyses were performed for these DEGs with the DAVID online tool. Interaction network was constructed among the over-represented pathways based on the protein-protein interactions within the pathways. Besides, the protein interaction information obtained from HPRD database were applied to constructed protein-protein interaction networks among the DEGs and hub genes and function module were screened out. A total of 2,296 DEGs were obtained and they were enriched in 34 pathways. An interaction network was constructed among 32 pathways, in which p53 signaling pathway acted as the hub pathway as it showed the highest node degree. The protein-protein interaction network comprised 1,481 interaction relationships among 332 genes which included 40 DEGs. Further analysis revealed that theses DEGs formed 7 functionmodules and many genes, such as PDGFRB, MET, FZD2, CCND1, PRKCB, ARHGEF6, JUP, WNT2, WNT5A and WNT11 were key genes in the networks. The DEGs and disturbed biological functions uncovered in present study may play important roles in the development of CRC and can contribute to the understanding on molecular mechanisms of CRC. Further these DEGs we obtained can be acted as potential biomarkers for diagnosis and therapy of CRC. C1 [Wang, Yonggang] Shandong University, Cancer Center, Qilu Hospital, Department of Chemotherapy, NO. 107 West Wenhua Road, 250012 Jinan, Shandong Province, China. [Zheng, Tianying] Shandong University, Cancer Center, Qilu Hospital, Department of Chemotherapy, NO. 107 West Wenhua Road, 250012 Jinan, Shandong Province, China. RP Zheng, T (reprint author), Shandong University, Cancer Center, Qilu Hospital, Department of Chemotherapy, 250012 Jinan, China. EM zhentianyingzh@hotmail.com CR WHO. Cancer, fact sheet N°297, World Health Organization, reviewed January 2013 http://wwwwhoint/mediacentre/factsheets/ fs297/en/indexhtml O’Connell JB, Maggard MA, Ko CY, 2004, Colon cancer survival rates with the new American Joint Committee on Cancer sixth edition staging. J Natl Cancer Inst 96(19):1420–1425 Ramaswamy S, Tamayo P, Rifkin R, Mukherjee S, Yeang C-H, Angelo M, Ladd C, Reich M, Latulippe E, Mesirov JP, 2001, Multiclass cancer diagnosis using tumor gene expression signatures. Proc Natl Acad Sci 98(26):15149–15154 Lanza G, FerracinM, Gafa R, Veronese A, Spizzo R, Pichiorri F, C-g L, Calin GA, Croce CM, Negrini M, 2007, mRNA/microRNA gene expression profile in microsatellite unstable colorectal cancer. Mol Cancer 6(1):54 Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, Smits AM, Bos JL, 1988, Genetic alterations during colorectal-tumor development. N Engl J Med 319(9):525–532 Law DJ, Olschwang S, Monpezat J-P, Lefrancois D, Jagelman D, Petrelli NJ, Thomas G, Feinberg AP, 1988, Concerted nonsyntenic allelic loss in human colorectal carcinoma. Science 241(4868):961– 965 Vogelstein B, Fearon ER, Kern SE, Preisinger A, Nakamura Y,White R, 1989, Allelotype of colorectal carcinomas. Science 244(4901): 207–211 Thiagalingam S, Lengauer C, Leach FS, Schutte M, Hahn SA, Overhauser J, Willson JK, Markowitz S, Hamilton SR, Kern SE, 1996, Evaluation of candidate tumour suppressor genes on chromosome 18 in colorectal cancers. Nat Genet 13(3):343–346 Peltomaki P, 2003, Role of DNA mismatch repair defects in the pathogenesis of human cancer. J Clin Oncol 21(6):1174–1179 Lynch HT, Lynch J, 2000, Lynch syndrome: genetics, natural history, genetic counseling, and prevention. J Clin Oncol 18(suppl 1):19s– 31s DeRisi J, Penland L, Brown PO, Bittner ML, Meltzer PS, Ray M, Chen Y, Su YA, Trent JM, 1996, Use of a cDNA microarray to analyse gene expression patterns in human cancer. Nat Genet 14(4): 457–460., DOI 10.1038/ng1296-457 Uddin S, Ahmed M, Hussain A, Abubaker J, Al-Sanea N, AbdulJabbar A, Ashari LH, Alhomoud S, Al-Dayel F, Jehan Z, Bavi P, Siraj AK, Al-Kuraya KS, 2011, Genome-wide expression analysis of Middle Eastern colorectal cancer reveals FOXM1 as a novel target for cancer therapy. Am J Pathol 178(2):537–547 Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP, 2003, Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4(2):249–264 Bolstad BM, Irizarry RA, Astrand M, Speed TP, 2003, A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19(2):185–193 Hochberg Y, Benjamini Y, 1990, More powerful procedures for multiple significance testing. Stat Med 9(7):811–818 da Huang W, Sherman BT, Lempicki RA, 2009, Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 37(1):1–13 Peri S, Navarro JD, Amanchy R, Kristiansen TZ, Jonnalagadda CK, Surendranath V, Niranjan V, Muthusamy B, Gandhi TK, Gronborg M, Ibarrola N, Deshpande N, Shanker K, Shivashankar HN, Rashmi BP, Ramya MA, Zhao Z, Chandrika KN, Padma N, Harsha HC, Yatish AJ, Kavitha MP, Menezes M, Choudhury DR, Suresh S, Ghosh N, Saravana R, Chandran S, Krishna S, Joy M, Anand SK, Madavan V, Joseph A, Wong GW, Schiemann WP, Constantinescu SN, Huang L, Khosravi-Far R, Steen H, TewariM, Ghaffari S, Blobe GC, Dang CV, Garcia JG, Pevsner J, Jensen ON, Roepstorff P, Deshpande KS, Chinnaiyan AM, Hamosh A, Chakravarti A, Pandey A, 2003, Development of human protein reference database as an initial platform for approaching systems biology in humans. Genome Res 13(10):2363–2371 Palla G, Derenyi I, Farkas I, Vicsek T, 2005, Uncovering the overlapping community structure of complex networks in nature and society. Nature 435(7043):814–818 Adamcsek B, Palla G, Farkas IJ, Derenyi I, Vicsek T, 2006, CFinder: locating cliques and overlapping modules in biological networks. Bioinformatics 22(8):1021–1023 Malumbres M, Barbacid M, 2009, Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer 9(3):153–166 Kastan MB, Bartek J, 2004, Cell-cycle checkpoints and cancer. Nature 432(7015):316–323 MolaeiM, YadollahzadehM, Almasi S, Shivarani S, Fatemi SR, Zali MR, 2011, Sporadic colorectal polyps and mismatch repair proteins. Indian J Pathol Microbiol 54(4):725–729 Tajima A, Iwaizumi M, Tseng-Rogenski S, Cabrera BL, Carethers JM, 2011, Both hMutSalpha and hMutSss DNA mismatch repair complexes participate in 5-fluorouracil cytotoxicity. PLoSOne 6(12): e28117 Ramirez-Ramirez MA, Sobrino-Cossio S, de la Mora-Levy JG, Hernandez-Guerrero A, Macedo-Reyes Vde J, Maldonado- Martinez HA, Alonso-Larraga JO, Ramirez-Solis ME, 2012, Loss of expression of DNA mismatch repair proteins in aberrant crypt foci identified in vivo by magnifying colonoscopy in subjects with hereditary nonpolyposic and sporadic colon rectal cancer. J Gastrointest Cancer 43(2):209–214., DOI 10.1007/s12029-011-9303-z Nishitani H, Lygerou Z, 2002, Control of DNA replication licensing in a cell cycle. Genes Cells 7(6):523–534 Bandura JL, Calvi BR, 2002, Duplication of the genome in normal and cancer cell cycles. Cancer Biol Ther 1(1):8–13 Macarthur M, Hold GL, El-Omar EM, 2004, Inflammation and Cancer II. Role of chronic inflammation and cytokine gene polymorphisms in the pathogenesis of gastrointestinal malignancy. Am J Physiol Gastrointest Liver Physiol 286(4):G515–G520 Theodoropoulos G, Papaconstantinou I, Felekouras E, Nikiteas N, Karakitsos P, Panoussopoulos D, Ch Lazaris A, Patsouris E, Bramis J, Gazouli M, 2006, Relation between common polymorphisms in genes related to inflammatory response and colorectal cancer. World J Gastroenterol 12(31):5037 Allen JI Molecular biology of colon polyps and colon cancer. In: Seminars in surgical oncology, 1995.Wiley Online Library, pp 399– 405 Stegh AH, 2012, Targeting the p53 signaling pathway in cancer therapy—the promises, challenges and perils. Expert Opin Ther Targets 16(1):67–83., DOI 10.1517/14728222.2011.643299 Bunz F, Dutriaux A, Lengauer C, Waldman T, Zhou S, Brown JP, Sedivy JM, Kinzler KW, Vogelstein B, 1998, Requirement for p53 and p21 to sustain G2 arrest after DNA damage. Science 282(5393): 1497–1501 TongWM, HandeMP, Lansdorp PM,Wang ZQ, 2001, DNA strand break-sensing molecule poly(ADP-Ribose, polymerase cooperates with p53 in telomere function, chromosome stability, and tumor suppression. Mol Cell Biol 21(12):4046–4054., DOI 10.1128/MCB. 21.12.4046-4054.2001 Achanta G, Sasaki R, Feng L, Carew JS, Lu W, Pelicano H, Keating MJ, Huang P, 2005, Novel role of p53 in maintaining mitochondrial genetic stability through interaction with DNA Pol gamma. EMBO J 24(19):3482–3492., DOI 10.1038/sj.emboj.7600819 Liu G, Parant JM, Lang G, Chau P, Chavez-Reyes A, El- Naggar AK, Multani A, Chang S, Lozano G, 2004, Chromosome stability, in the absence of apoptosis, is critical for suppression of tumorigenesis in Trp53 mutant mice. Nat Genet 36(1):63–68., DOI 10.1038/ng1282 Iglesias P, Salas A, Costoya JA, 2012, The maintenance of mitochondrial genetic stability is crucial during the oncogenic process. Commun Integr Biol 5(1):34–38 Lengauer C, Kinzler KW, Vogelstein B, 1997, Genetic instability in colorectal cancers. Nature 386(6625):623–627., DOI 10.1038/ 386623a0 Wehler TC, Frerichs K, Graf C, Drescher D, Schimanski K, Biesterfeld S, Berger MR, Kanzler S, Junginger T, Galle PR, Moehler M, Gockel I, Schimanski CC, 2008, PDGFRalpha/beta expression correlates with the metastatic behavior of human colorectal cancer: a possible rationale for a molecular targeting strategy. Oncol Rep 19(3):697–704 Kuwai T, Nakamura T, Sasaki T, Kitadai Y, Kim J-S, Langley R, Fan D, Wang X, Do K-A, Kim S-J, Fidler I, 2008, Targeting the EGFR, VEGFR, and PDGFR on colon cancer cells and stromal cells is required for therapy. Clin Exp Metastasis 25(4):477–489., DOI 10. 1007/s10585-008-9153-7 Di Renzo MF, Olivero M, Giacomini A, Porte H, Chastre E, Mirossay L, Nordlinger B, Bretti S, Bottardi S, Giordano S, 1995, Overexpression and amplification of the met/HGF receptor gene during the progression of colorectal cancer. Clin Cancer Res 1(2): 147–154 Fukuura T, Miki C, Inoue T, Matsumoto K, Suzuki H, 1998, Serum hepatocyte growth factor as an index of disease status of patients with colorectal carcinoma. Br J Cancer 78(4):454 Gherardi E, Birchmeier W, Birchmeier C, Woude GV, 2012, Targeting MET in cancer: rationale and progress. Nat Rev Cancer 12(2):89–103 Herynk MH, Stoeltzing O, Reinmuth N, Parikh NU, Abounader R, Laterra J, Radinsky R, EllisLM, Gallick GE, 2003, Down-regulation of c-Met inhibits growth in the liver of human colorectal carcinoma cells. Cancer Res 63(11):2990–2996 FrontiniMJ, Nong Z, Gros R, Drangova M, O’Neil C, RahmanMN, Akawi O, Yin H, Ellis CG, Pickering JG, 2011, Fibroblast growth factor 9 delivery during angiogenesis produces durable, vasoresponsive microvessels wrapped by smooth muscle cells. Nat Biotechnol 29(5):421–427 Suzuki H,Watkins DN, Jair KW, Schuebel KE,Markowitz SD, Chen WD, Pretlow TP, Yang B, Akiyama Y, Van Engeland M, Toyota M, Tokino T, Hinoda Y, Imai K, Herman JG, Baylin SB, 2004, Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat Genet 36(4):417–422., DOI 10. 1038/ng1330 Segditsas S, Tomlinson I, 2006, Colorectal cancer and genetic alterations in the Wnt pathway. Oncogene 25(57):7531–7537 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 611 EP 618 DI 10.1007/s12253-013-9739-5 PG 8 ER PT J AU Xuan, Qj Wang, Jx Nanding, A Wang, Zp Liu, H Lian, X Zhang, Qy AF Xuan, Qi-jia Wang, Jing-xuan Nanding, Abiyasi Wang, Zhi-peng Liu, Hang Lian, Xin Zhang, Qing-yuan TI Tumor-Associated Macrophages are Correlated with Tamoxifen Resistance in the Postmenopausal Breast Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Tumor-associated macrophages; Epidermal growth factor receptor; Tamoxifen resistance; Obesity ID Breast cancer; Tumor-associated macrophages; Epidermal growth factor receptor; Tamoxifen resistance; Obesity AB Tumor-associated macrophages (TAMs) have been correlated with increased angiogenesis and poor prognosis in breast cancer. However, the precise role of TAMs in tamoxifen resistance remains unclear. We used immunohistochemical method to examine the expression of epidermal growth factor receptor (EGFR) and CD163+ macrophages in 100 breast cancer tissues. The clinical and biological features of 100 patients were estrogen receptor (ER)-positive and human epidermal growth factor receptor 2(Her-2)-negative tumors. The tamoxifen resistant tissues (n=48) were the surgical excision samples frompatientswho developed recurrence ormetastasis at the time of adjuvant tamoxifen treatment. The tamoxifen resistant tissues were contrast to tamoxifen sensitive tissues (n=52). Positive staining for EGFR and CD163+ macrophages were observed in 21 samples (43.8 %) and in 26 samples (54.2 %) respectively in tamoxifen resistance group, which were higher than that of tamoxifen sensitive group (P=0.001 and P=0.000279 respectively). Significant positive correlations were found between the expression of EGFR and CD163+ macrophages (r=0.567, P<0.01). CD163+ macrophages were positively correlated with tumor size, lymph node metastasis and obesity. Obesity was also related to tamoxifen resistance (P<0.05). The patients with higher density of CD163+ macrophages infiltration suffered fromshorter time to develop recurrence or metastasis (P<0.05). TAMs may be associated with tamoxifen resistance. Further studies are needed to investigate the potential mechanism between TAMs and tamoxifen resistance. C1 [Xuan, Qi-jia] Harbin Medical University, Cancer Hospital, Department of Medical Oncology, Haping Road 150 of Nangang District, 150081 Harbin, Heilongjiang, China. [Wang, Jing-xuan] Harbin Medical University, Cancer Hospital, Department of Medical Oncology, Haping Road 150 of Nangang District, 150081 Harbin, Heilongjiang, China. [Nanding, Abiyasi] Tumor Hospital of Harbin Medical University, Department of Pathology, 150081 Harbin, Heilongjiang, China. [Wang, Zhi-peng] Harbin Medical University, Cancer Hospital, Department of Medical Oncology, Haping Road 150 of Nangang District, 150081 Harbin, Heilongjiang, China. [Liu, Hang] Harbin Medical University, Cancer Hospital, Department of Medical Oncology, Haping Road 150 of Nangang District, 150081 Harbin, Heilongjiang, China. [Lian, Xin] Harbin Medical University, Cancer Hospital, Department of Medical Oncology, Haping Road 150 of Nangang District, 150081 Harbin, Heilongjiang, China. [Zhang, Qing-yuan] Harbin Medical University, Cancer Hospital, Department of Medical Oncology, Haping Road 150 of Nangang District, 150081 Harbin, Heilongjiang, China. RP Zhang, Qy (reprint author), Harbin Medical University, Cancer Hospital, Department of Medical Oncology, 150081 Harbin, China. EM zqyxsci@126.com CR Key TJ, Verkasalo PK, Banks E, 2001, Epidemiology of breast cancer. Lancet Oncol 2:133–140 Musgrove EA, Sutherland RL, 2009, Biological determinants of endocrine resistance in breast cancer. Nat Rev Cancer 9:631–643 Gradishar WJ, 2004, Tamoxifen-What next? Oncologist 9:378–384 Ring A, Dowsett M, 2004, Mechanisms of tamoxifen resistance. Endocr Relat Cancer 11:643–658 Beelen K, Zwart W, Linn SC, 2012, Can predictive biomarkers in breast cancer guide adjuvant endocrine therapy? Nat Rev Clin Oncol 9:529–541 Pollard JW, 2008, Macrophages define the invasive microenvironment in breast cancer. J Leukoc Biol 84:623–630 Mantovani A, 2005, Cancer: inflammation by remote control. Nature 435:752–753 Balkwill F, Mantovani A, 2001, Inflammation and cancer: back to Virchow? Lancet 357:539–545 Sica A, Schioppa T, Mantovani A et al, 2006, Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur J Cancer 42:717–727 Mantovani A, Sica A, Sozzani S et al, 2004, The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 25:677–686 Lau SK, Chu PG, Weiss LM, 2004, CD163 A specific marker of macrophages in paraffin-embedded tissue samples. Am J Clin Pathol 122:794–801 Leek RD, Harris AL, 2002, Tumor-associated macrophages in breast cancer. J Mammary Gland Biol Neoplasia 7:177–189 Soeda S, Nakamura N, Ozeki T et al, 2008, Tumor-associated macrophages correlate with vascular space invasion and myometrial invasion in endometrial carcinoma. Gynecol Oncol 109:122–128 Konecny GE, Santos L, Winterhoff B et al, 2008, HER2 gene amplification and EGFR expression in a large cohort of surgically staged patients with nonendometrioid, type II, endometrial cancer. Br J Cancer 100:89–95 Garcia-Becerra R, Santos N, Diaz L et al, 2012, Mechanisms of resistance to endocrine therapy in breast cancer: focus on signaling pathways, miRNAs and genetically based resistance. Int J Mol Sci 14:108–145 McKenna NJ, Lanz RB, O’Malley BW, 1999, Nuclear receptor coregulators: cellular and molecular biology. Endocr Rev 20:321–344 Frasor J, Danes JM, Komm B et al, 2003, Profiling of estrogen up-and down-regulated gene expression in human breast cancer cells: insights into gene networks and pathways underlying estrogenic control of proliferation and cell phenotype. Endocrinol 144:4562–4574 Deroo BJ, Korach KS, 2006, Estrogen receptors and human disease. J Clin Invest 116:561–570 Norris JD, Fan D, Kerner SA et al, 1997, Identification of a third autonomous activation domain within the human estrogen receptor. Mol Endocrinol 11:747–754 Nicholson RI, Hutcheson IR, Knowlden JM et al, 2004, Nonendocrine pathways and endocrine resistance observations with antiestrogens and signal transduction inhibitors in combination. Clin Cancer Res 10:346s–354s Arpino G, Green SJ, Allred DC et al, 2004, HER-2 amplification, HER-1 expression, and tamoxifen response in estrogen receptorpositive metastatic breast cancer a southwest oncology group study. Clin Cancer Res 10:5670–5676 Tang X, 2013, Tumor-associated macrophages as potential diagnostic and prognostic biomarkers in breast cancer. Cancer Lett Volodko N, Reiner A, Rudas M et al, 1998, Tumour-associated macrophages in breast cancer and their prognostic correlations. Breast 7:99–105 Mahmoud SMA, LeeAHS, Paish EC et al, 2012, Tumour-infiltrating macrophages and clinical outcome in breast cancer. J Clin Pathol 65: 159–163 Morris PG, Hudis CA, Giri D et al, 2011, Inflammation and increased aromatase expression occur in the breast tissue of obese women with breast cancer. Cancer Prev Res, Phila, 4:1021–1029 Monteiro R, Azevedo I, 2010, Chronic inflammation in obesity and the metabolic syndrome.Mediat Inflamm., DOI 10.1155/2010/289645 Cinti S,Mitchell G, Barbatelli G et al, 2005, Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res 46:2347–2355 Zhao Y, Agarwal VR, Mendelson CR et al, 1996, Estrogen biosynthesis proximal to a breast tumor is stimulated by PGE2 via cyclic AMP, leading to activation of promoter II of the CYP19, aromatase, gene. Endocrinology 137:5739–5742 Irahara N, Miyoshi Y, Taguchi T et al, 2006, Quantitative analysis of aromatase mRNA expression derived from various promoters, I.4, I.3, PII and I.7, and its association with expression of TNF-alpha, IL- 6 and COX-2 mRNAs in human breast cancer. Int J Cancer 118: 1915–1921 Bachelot T, Ray-Coquard I, Menetrier-Caux C et al, 2003, Prognostic value of serum levels of interleukin 6 and of serum and plasma levels of vascular endothelial growth factor in hormone-refractory metastatic breast cancer patients. Br J Cancer 88:1721–1726 Dandona P, Weinstock R, Thusu K et al, 1998, Tumor necrosis factor-alpha in sera of obese patients: fall with weight loss. J Clin Endocrinol Metab 83:2907–2910 Leek RD, Hunt NC, Landers RJ et al, 2000, Macrophage infiltration is associated with VEGF and EGFR expression in breast cancer. J Pathol 190:430–436 Yang J, Liao D, Chen C et al, 2013, Tumor associated macrophages regulate murine breast cancer stem cells through a novel paracrine EGFR/Stat3/Sox-2 signaling pathway. Stem Cells 31:248–258 Vlaicu P,Mertins P, ThomasMet al, 2013)Monocytes/macrophages support mammary tumor invasivity by co-secreting lineage-specific EGFR ligands and a STAT3 activator. BMC Cancer 13:197 Condeelis J, Segall JE, 2003, Intravital imaging of cell movement in tumours. Nat Rev Cancer 3:921–930 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 619 EP 624 DI 10.1007/s12253-013-9740-z PG 6 ER PT J AU Ahmed, AR Shawky, EAA Hamed, HR AF Ahmed, Allah Rehab Shawky, El-Aty Abd Hamed, Hamdy Rasha TI Prognostic Significance of Cyclin D1 and E-cadherin Expression in Laryngeal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE E-cadherin; CyclinD1; Laryngeal squamous cell carcinoma; Prognosis ID E-cadherin; CyclinD1; Laryngeal squamous cell carcinoma; Prognosis AB Cyclin D1 and E-cadherin are important factors in the progression and metastasis of cancers. Their role in laryngeal carcinoma has been studied with conflicting results. To define the frequency of cyclin D1 and E-cadherin expression and its correlation with both the clinicopathological characteristics and prognosis of patients with laryngeal squamous cell carcinoma (LSCC). Tumor tissue samples from 75 patients with laryngeal squamous cell carcinoma were examined for cyclin D1 and E-cadherin expression by immunohistochemistry. The relationship between the expression of both molecules and the age and sex of the patient, tumor site, tumor differentiation, lymph node metastasis, tumor invasiveness, TNM stages, tumor recurrence and overall survival was analyzed. Cyclin D1 was found to be a significant independent prognostic factor of lymph node metastasis (p=0.000). The multivariate analysis revealed that cyclin D1 and E-cadherin expression wasn’t an independent prognostic factor of local recurrence free survival (LRFS) in patients with LSCC (P=0.56 and 0.28) respectively. However, the univariate analysis revealed a significant association between them and LRFS (p=0.003 and 0.000) respectively. Also, the group of high cyclin D1 /low Ecadherin expression had the poorest prognosis, so they might serve as potential predictors of the prognosis of the patients with LSCC. E-cadherin was found to affect the overall survival (OAS) significantly by the univariate analysis (p=0.01). However, by the multivariate analysis the TNM stage was the only independent prognostic factor of OAS (p<0.05). Cyclin D1 can be used as an independent prognostic marker of lymph node metastasis in patients with LSCC and can help to identify those patients with clinically negative lymph nodes but with considerable risk for occult metastasis. Detection of cyclin D1 and E-cadherin status in LSCC may contribute to the identification of patients with high risk factors of local recurrence. However, they don’t appear to be better prognostic predictors than other established markers in LSCC. C1 [Ahmed, Allah Rehab] Mansoura University, Faculty of Medicine, Department of PathologyMansoura, Egypt. [Shawky, El-Aty Abd] Mansoura University, Faculty of Medicine, Department of PathologyMansoura, Egypt. [Hamed, Hamdy Rasha] Mansoura University, Faculty of Medicine, Clinical Oncology and Nuclear Medicine DepartmentMansoura, Egypt. RP Hamed, HR (reprint author), Mansoura University, Faculty of Medicine, Clinical Oncology and Nuclear Medicine Department, Mansoura, Egypt. EM rashahamdy532@yahoo.com CR Luke CG, Yeoh E, Roder DM, 2008, Exploring trends in laryngeal cancer incidence, mortality and survival: implications for research and cancer control. Asian Pac J Cancer Prev 9(3):397–402 Freedman LS, Edwards BK, Ries LAG et al, 2006, Cancer incidence in four member countries, Cyprus, Egypt, Israel, and Jordan, of the Middle East Cancer Consortium, MECC, compared with US SEER. National Cancer Institute. NIH Pub. No. 06–5873. Bethesda, MD. Chapter title; Overview and Summary Data;, 1): pp 1–27. Edited by Freedman LS, Edwards BK, Ries LAG, et al Torrente MC, Rodrigo JP, Haigentz M et al, 2011, Human papillomavirus infections in laryngeal cancer. Head Neck 33(4):581–586 Krecicki T, Smigiel R, Fraczek M et al, 2004, Studies of the cell cycle regulatory proteins, P16, cyclinD1and retinoblastoma protein in laryngeal carcinoma tissue. J Laryngol Otol 118(9):676–680 Cristobal E, Arribas B, Tardio J et al, 2000, Analysis of the cyclin D1/p16/pRb pathway in parathyroid adenomas. Endocr Pathol 11(3): 259–266 vanDiest PJ, Michalides RJAM, Jannink I et al, 1997, Cyclin D1 expression in invasive breast cancer: correlations and prognostic value. Am J Pathol 150(2):705–711 Ishikawa T, Furihata M, Ohtsuki Y et al, 1998, Cyclin D1 overexpression related to retinoblastoma protein expression as a prognostic marker in human oesophageal squamous cell carcinoma. Br J Cancer 77(1):92–97 Wangefjord S, Manjer J, Gaber A et al, 2011, Cyclin D1 expression in colorectal cancer is a favourable prognostic factor inmen but not in women in a prospective, population-based cohort study. Biol Sex Differ 2:10 Ito Y, Matsuura N, Sakon M et al, 1999, Expression and prognostic roles of the G1-S modulators in hepatocellular carcinoma: p27 independently predicts the recurrence. Hepatology 30(1):90–99 Fracchiolla NS, Pruneri G, Pignataro L et al, 1997, Molecular and immunohistochemical analysis of the bcl-1/cyclin D1 gene in laryngeal squamous cell carcinomas: correlation of protein expression with lymph node metastases and advanced clinical stage. Cancer 79(6):1114–1121 Do NY, Park SY, Lim SC, 2004, The role of E-cadherin/B2-catenin complex and cyclin D1 in head and neck squamous cell carcinoma. Cancer Res Treat 36(1):72–78 Berx G, Staes K, van Hengel J et al, 1995, Cloning and characterization of the human invasion suppressor gene E-cadherin, CDH1). Genomics 26(2):281–289 Kashibuchi K, Tomita K, Schalken JA et al, 2006, The prognostic value of E-cadherin, alpha-, beta-, and gamma-catenin in urothelial cancer of the upper urinary tract. Eur Urol 49(5):839–845 Loddo M, Kingsbury SR, Rashid M et al, 2009, Cell-cycle-phase progression analysis identifies unique phenotypes of major prognostic and predictive significance in breast cancer. Br J Cancer 100(6): 959–970 Barnes L, Eveson JW, Reichart PA et al, 2005, World Health Organization classification of tumors: pathology and genetics of tumors of the head and neck. Lyon: IARC. Squamous cell carcinoma in; Hypopharynx, larynx and trachea;, 3): p p121. Edited by Cardesa A, Gale N, Nadal A et al Edge SB, Byrd DR, Compton CC et al, 2010, Larynx. In: AJCC cancer staging manual, 7th edn. Springer, New York, pp 57–62 Larizadeh MH, Damghani MA, Tabrizchi H et al, 2009, Expression of E-cadherin in squamous cell carcinoma of the larynx and its correlation with clinicopathological features. J Med Sci 9(1):41–45 Leon X, Lopez M, Garcia J et al, 2008, Recurrent laryngeal squamous cell carcinoma: rTNM versus composite laryngeal recurrence staging system. Proposal for a modification of the CLRSS to improve patient classification. Head Neck 30(7):939–945 Awada A, Lalami Y, 2005, Molecular markers, molecular-targeted therapies and taxanes: how to integrate the progress into clinical research and practice for the management of head and neck cancers. Curr Opin Oncol 17(3):209–211 Zhang Y, Xu Z, Jun-Xi Wang J et al, 2012, G1/S-specific cyclin-D1 might be a prognostic biomarker for patients with laryngeal squamous cell carcinoma. Asian Pac J Cancer Prev 13(5):2133–2137 Pignataro L, Sambataro G, Pagani D et al, 2005, Clinico-prognostic value of D-type cyclins and p27 in laryngeal cancer patients: a review. Acta Otorhinolaryngol Ital 25(2):75–85 Vielba R, Eng JBC, Ispizua A et al, 2003, Cyclin D1 as prognostic factors in squamous cell carcinoma of the larynx. Laryngoscope 113(1):167–172 Galera-Ruiz H, Rios-Moreno MJ, Gonzalez-Campora R, 2012, The cadherin-catenin complex in laryngeal squamous cell carcinoma. Eur Arch Otorhinolaryngol 269(4):1183–1188 Li JJ, Zhang GH, Yang XM et al, 2012, Reduced E-cadherin expression is associated with lymph node metastases in laryngeal squamous cell carcinoma. Auris Nasus Larynx 39(2):186–192 PaksoyM, Hardal U, Caglar C, 2011, Expression of cathepsin D and E-cadherin in primary laryngeal cancers correlation with neck lymph node involvement. J Cancer Res Clin Oncol 137(9):1371–1377 Rodrigo JP, Dominguez F, Alvarez C et al, 2002, Expression of Ecadherin in squamous cell carcinomas of the supraglottic larynx with correlations to clinicopathological features. Eur J Cancer 38(8): 1059–1064 Rodrigo JP, Dominguez F, Suarez V et al, 2007, Focal adhesion kinase and E-cadherin as markers for nodal metastasis in laryngeal cancer. Arch Otolaryngol 133(2):145–150 Michalides RJAM, van Veelen NMJ, Kristel PMP et al, 1997, Overexpression of cyclin D1 indicates a poor prognosis in squamous cell carcinoma of the head and neck. Arch Otolaryngol Head Neck Surg 123(5):497–502 Pignataro L, Capaccio P, Pruneri G et al, 1996, P53 and cyclin D1 protein expression in glottic laryngeal squamous cell carcinomas involving the anterior commissure, pT1bN0M0). Int J Oncol 3(9): 553–557 Snow GB, Patel P, Leemans CR et al, 1992)Management of cervical lymph nodes in patients with head and neck cancer. Eur Arch Otorhinolaryngol 249:187–194 Zou J, Yang H, Chen F et al, 2010, Prognostic significance of fascin- 1 and E-cadherin expression in laryngeal squamous cell carcinoma. Eur J Cancer Prev 19(1):11–17 Pruneri G, Pignataro L, Valentini S et al, 2005, Cyclin D3 immunoreactivity is an independent predictor of survival in laryngeal squamous cell carcinoma. Clin Cancer Res 11(1):242–248 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 625 EP 633 DI 10.1007/s12253-014-9741-6 PG 9 ER PT J AU Oskina, N Ermolenko, N Boyarskih, AU Lazarev, A Petrova, V Ganov, D Tonacheva, O Lifschitz, G Filipenko, M AF Oskina, А. Natalja Ermolenko, А. Natalya Boyarskih, Aleksandrovna Ulyana Lazarev, F. Aleksandr Petrova, D. Valentina Ganov, I. Dmitriy Tonacheva, G. Olga Lifschitz, I. Galina Filipenko, L. Maxim TI Associations Between SNPs Within Antioxidant Genes and the Risk of Prostate Cancer in the Siberian Region of Russia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE GPX1; GSTP1; MnSOD; Prostate cancer; Single nucleotide polymorphisms (SNPs); SOD2 ID GPX1; GSTP1; MnSOD; Prostate cancer; Single nucleotide polymorphisms (SNPs); SOD2 AB In the present study we investigated the association of a number of polymorphic changes in antioxidant system genes (SNPs rs1050450 in the GPX1 gene, rs1695 and rs1138272 in the GSTP1 gene and rs4880 in the MnSOD gene) with the risk of prostate cancer. The association of disease stage and PSA levels with specific genotypes was also analyzed. A study was conducted with the participation of 736 Russian men. We compared the frequency of occurrence of the studied alleles in patients with prostate cancer (392) to a control group (344) of men without a history of cancer. Genotyping was performed by real-time PCR. Comparison of frequencies of alleles and genotypes were performed using logistic regression analysis. No statistically significant association with the risk of prostate cancer was found for any of the SNPs studied (p>0.05). For SNP rs1695 in the GSTP1 gene, a correlation with cancer disease stage was observed: a GG genotype is significantly more common in patients with PCa in the 3rd and 4th stage than 1st and 2nd (OR[95%CI]= 2.66[1.15–6.18], p=0.02). Both studied SNPs of GSTP1 gene are associated with the level of PSA: the GG rs1695 and the TT rs1138272 genotypes are associated with higher PSA levels (p=1.5*10−3). C1 [Oskina, А. Natalja] Institute of Chemical Biology and Fundamental MedicineNovosibirsk, Russian Federation. [Ermolenko, А. Natalya] Institute of Chemical Biology and Fundamental MedicineNovosibirsk, Russian Federation. [Boyarskih, Aleksandrovna Ulyana] Institute of Chemical Biology and Fundamental MedicineNovosibirsk, Russian Federation. [Lazarev, F. Aleksandr] N. N. Blokhin Russian Cancer Research Center RAMSBarnaul, Russian Federation. [Petrova, D. Valentina] N. N. Blokhin Russian Cancer Research Center RAMSBarnaul, Russian Federation. [Ganov, I. Dmitriy] N. N. Blokhin Russian Cancer Research Center RAMSBarnaul, Russian Federation. [Tonacheva, G. Olga] Krasnoyarsk Regional Oncology CenterKrasnoyarsk, Russian Federation. [Lifschitz, I. Galina] Institute of Chemical Biology and Fundamental MedicineNovosibirsk, Russian Federation. [Filipenko, L. Maxim] Institute of Chemical Biology and Fundamental MedicineNovosibirsk, Russian Federation. RP Oskina, N (reprint author), Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russian Federation. EM nattasha.o@gmail.com CR Ames BN, Gold LS,WillettWC(1995, The causes and prevention of cancer. Proc Natl Acad Sci U S A 92(12):5258–5265 Feig DI, Reid TM, Loeb LA, 1994, Reactive oxygen species in tumorigenesis. Cancer Res 54(7 Suppl):1890s–1894s Dreher D, Junod AF, 1996, Role of oxygen free radicals in cancer development. Eur J Cancer 32A(1):30–38 Matsui A, Ikeda T, Enomoto K, Hosoda K, Nakashima H, Omae K, Watanabe M, Hibi T, Kitajima M, 2000, Increased formation of oxidative DNA damage, 8-hydroxy-2′-deoxyguanosine, in human breast cancer tissue and its relationship to GSTP1 and COMT genotypes. Cancer Lett 151(1):87–95 Forsberg L, de Faire U, Morgenstern R, 2001, Oxidative stress, human genetic variation, and disease. Arch Biochem Biophys 389(1):84–93 Li Y,Huang TT, Carlson EJ, Melov S, Ursell PC, Olson JL, Noble LJ, Yoshimura MP, Berger C, Chan PH,Wallace DC, Epstein CJ, 1995, Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nat Genet 11(4):376–381 Rosenblum JS, Gilula NB, Lerner RA, 1996, On signal sequence polymorphisms and diseases of distribution. Proc Natl Acad Sci U S A 93(9):4471–4473 Sutton A, Khoury H, Prip-Buus C, Cepanec C, Pessayre D, Degoul F, 2003, The Ala16Val genetic dimorphism modulates the import of human manganese superoxide dismutase into rat liver mitochondria. Pharmacogenetics 13(3):145–157 Baker AM, Oberley LW, Cohen MB, 1997, Expression of antioxidant enzymes in human prostatic adenocarcinoma. Prostate 32(4): 229–233 Wang S,Wang F, Shi X, Dai J, Peng Y, Guo X,Wang X, Shen H, Hu Z, 2009, Association between manganese superoxide dismutase, MnSOD, Val-9Ala polymorphism and cancer risk - a metaanalysis. Eur J Cancer 45(16):2874–2881., DOI 10.1016/j.ejca Ishida K, Morino T, Takagi K, Sukenaga Y, 1987, Nucleotide sequence of a human gene for glutathione peroxidase. Nucleic Acids Res 15(23):10051 Kucukgergin C, Gokpinar M, Sanli O, Tefik T, Oktar T, Seckin S, 2011, Association between genetic variants in glutathione peroxidase 1, GPx1, gene, GPx activity and the risk of prostate cancer. Minerva Urol Nefrol 63(3):183–190 Arsova-Sarafinovska Z, Matevska N, Eken A, Petrovski D, Banev S, Dzikova S, Georgiev V, Sikole A, Erdem O, Sayal A, Aydin A, Dimovski AJ, 2009, Glutathione peroxidase 1, GPX1, genetic polymorphism, erythrocyte GPX activity, and prostate cancer risk. Int Urol Nephrol 41(1):63–70., DOI 10.1007/s11255-008-9407-y Choi JY, Neuhouser ML, Barnett M, Hudson M, Kristal AR, Thornquist M, King IB, Goodman GE, Ambrosone CB, 2007, Polymorphisms in oxidative stress-related genes are not associated with prostate cancer risk in heavy smokers. Cancer Epidemiol Biomarkers Prev 16(6):1115–1120 Board PG, Webb GC, Coggan M, 1989, Isolation of a cDNA clone and localization of the human glutathione S-transferase 3 genes to chromosome bands 11q13 and 12q13-14. Ann Hum Genet 53(Pt 3): 205–213 Nelson WG, De Marzo AM, DeWeese TL, Isaacs WB, 2004, The role of inflammation in the pathogenesis of prostate cancer. J Urol 172(5 Pt 2):S6–S11, discussion S11–12 Lee WH, Morton RA, Epstein JI, Brooks JD, Campbell PA, Bova GS, HsiehWS, IsaacsWB, NelsonWG(1994, Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proc Natl Acad Sci U S A 91(24):11733–11737 Lin X, Tascilar M, Lee WH, Vles WJ, Lee BH, Veeraswamy R, Asgari K, Freije D, van Rees B, Gage WR, Bova GS, Isaacs WB, Brooks JD, DeWeese TL, De MarzoAM, NelsonWG, 2001, GSTP1 CpG island hypermethylation is responsible for the absence of GSTP1 expression in human prostate cancer cells. Am J Pathol 159(5):1815–1826 Wigginton JE, Cutler DJ, Abecasis GR, 2005, A note on exact tests of Hardy-Weinberg equilibrium. Am J Hum Genet 76(5):887–893 Gaunt TR, Rodriguez S, Day IN, 2007, Cubic exact solutions for the estimation of pairwise haplotype frequencies: implications for linkage disequilibrium analyses and a web tool ‘CubeX’. BMC Bioinforma 8:428 Schaid DJ, Rowland CM, Tines DE, Jacobson RM, Poland GA, 2002, Score tests for association between traits and haplotypes when linkage phase is ambiguous. Am J Hum Genet 70(2):425–434 Emerit I, 1994, Reactive oxygen species, chromosome mutation, and cancer: possible role of clastogenic factors in carcinogenesis. Free Radic Biol Med 16(1):99–109 Woodson K, Tangrea JA, Lehman TA, Modali R, TaylorKM, Snyder K, Taylor PR, Virtamo J, Albanes D, 2003, Manganese superoxide dismutase, MnSOD, polymorphism, alpha-tocopherol supplementation and prostate cancer risk in the alpha-tocopherol, beta-carotene cancer prevention study, Finland). Cancer Causes Control 14(6): 513–518 Rose DP, Boyar AP, Wynder EL, 1986, International comparisons of mortality rates for cancer of the breast, ovary, prostate, and colon, and per capita food consumption. Cancer 58(11):2363–2371 Fair WR, Fleshner NE, Heston W, 1997, Cancer of the prostate: a nutritional disease? Urology 50(6):840–848 Fleshner NE, Kucuk O, 2001, Antioxidant dietary supplements: rationale and current status as chemopreventive agents for prostate cancer. Urology 57(4 Suppl 1):90–94 Wei H, 1992, Activation of oncogenes and/or inactivation of antioncogenes by reactive oxygen species. Med Hypotheses 39(3):267– 270 Hainaut P, Milner J, 1993, Redox modulation of p53 conformation and sequence-specific DNA binding in vitro. Cancer Res 53(19): 4469–4473 Navone NM, Troncoso P, Pisters LL, Goodrow TL, Palmer JL, Nichols WW, von Eschenbach AC, Conti CJ, 1993, p53 protein accumulation and genemutation in the progression of human prostate carcinoma. J Natl Cancer Inst 85(20):1657–1669 Yossepowitch O, Pinchuk I, Gur U, Neumann A, Lichtenberg D, Baniel J, 2007, Advanced but not localized prostate cancer is associated with increased oxidative stress. J Urol 178(4 Pt 1):1238–1243, discussion 1243-1234 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 635 EP 640 DI 10.1007/s12253-014-9742-5 PG 6 ER PT J AU Yang, X Liu, G Xiao, H Yu, F Xiang, X Lu, Y Li, W Liu, X Li, Sh Shi, Y AF Yang, Xiaolin Liu, Geling Xiao, Hongzhen Yu, Fang Xiang, Xiuxiu Lu, Yifang Li, Weijuan Liu, Xiuling Li, Sha Shi, Yanping TI TPX2 Overexpression in Medullary Thyroid Carcinoma Mediates TT Cell Proliferation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Medullary thyroid cancer; TPX2; siRNA; Proliferation; Apoptosis ID Medullary thyroid cancer; TPX2; siRNA; Proliferation; Apoptosis AB TPX2 (targeting protein for xenopus kinesinlike protein 2), a microtubule-associated protein, plays an important role in the formation of the mitotic spindle. Abnormal expression of TPX2 in various types of malignant tumors has been reported, but less is known for medullary thyroid cancer (MTC). We investigated the expression of TPX2 in human MTC tissues and its potential use as a therapeutic target. Immunohistochemical analysis of TPX2 expression was performed for 32 cases of MTC and 8 cases of normal thyroid. TPX2 expression was found to be significantly higher in MTC compared to normal thyroid tissues (P<0.05), and to be associated with tumor size, lymph node metastasis, and advanced disease stage. The cellular effects of TPX2 knockdown, including cell proliferation, apoptosis, cell cycle diffusions, and mitotic gene expression were investigated using small interfering RNA (siRNA). TPX2- siRNA caused G1 and G2-phase cell cycle arrest, inhibited cell proliferation, and induced apoptosis. TPX2-siRNA also downregulated Aurora-A and cyclinB1 protein expression in MTC cells and enhanced the expression of p53 protein (P<0.05). These results suggest that TPX2 may be of potential use as a new marker for MTC prognosis and therapy. C1 [Yang, Xiaolin] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Liu, Geling] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Xiao, Hongzhen] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Yu, Fang] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Xiang, Xiuxiu] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Lu, Yifang] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Li, Weijuan] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Liu, Xiuling] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Li, Sha] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Shi, Yanping] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. RP Liu, G (reprint author), Tangshan Workers Hospital, Department of Endocrinology (Section I), Tangshan, China. EM liugeling1963@163.com CR Simard EP, Ward EM, Siegel R, Jemal A, 2012, Cancers with increasing incidence trends in the United States: 1999 through 2008. CA Cancer J Clin 62(2):118–128 Trimboli P, Ulisse S, Graziano F, Marzullo A, Ruggieri M, Calvanese A, Piccirilli F, Cavaliere R, Fumarola A,D’ArmientoM(2006, Trend in thyroid carcinoma size, age at diagnosis, and histology in a retrospective study of 500 cases diagnosed over 20 years. Thyroid 16(11):1151–1155 Rahmani N, Abbas Hashemi S, Fazli M, Raisian M, 2013, Clinical management and outcomes of papillary, follicular and medullary thyroid cancer surgery. Med Glas, Zenica, 10(1):164–167 Janssen A, Medema RH, 2012, Genetic instability: tipping the balance. Oncogene., DOI 10.1038/onc.2012.576 LinD, Ma Y, Xiao T, Guo S,HanN, SuK, Yi S, Fang J,Cheng S,Gao Y, 2006, TPX2 expression and its significance in squamous cell carcinoma of lung. Zhonghua Bing Li Xue Za Zhi Chin J Pathol 35(9):540 Shigeishi H, Ohta K, Hiraoka M, Fujimoto S, Minami M, Higashikawa K, Kamata N, 2009, Expression of TPX2 in salivary gland carcinomas. Oncol Rep 21(2):341 Mohsenifar J, Almassi-Aghdam M, Mohammad-Taheri Z, Zare K, Jafari B, Atri M, Mortazavi S-H, Azadeh P, Bagherzadeh M, Rahimi F, 2007, Prognostic values of proliferative markers ki-67 and repp86 in breast cancer. Arch Iran Med 10(1):27–31 Baldini E, Arlot-Bonnemains Y, Sorrenti S,Mian C, Pelizzo MR, De Antoni E, Palermo S, Morrone S, Barollo S, Nesca A, Moretti CG, D’Armiento M, Ulisse S, 2011, Aurora kinases are expressed in medullary thyroid carcinoma, MTC, and their inhibition suppresses in vitro growth and tumorigenicity of the MTC derived cell line TT. BMC Cancer 11:411 Schmittgen TD, Livak KJ, 2008, Analyzing real-time PCR data by the comparative CT method. Nat Protoc 3(6):1101–1108 Wittmann T, Hyman A, Desai A, 2001, The spindle: a dynamic assembly of microtubules and motors. Nat Cell Biol 3:E28–E34 Aguirre-Portoles C, Bird AW, Hyman A et al, 2012, TPX2 controls spindle integrity, genome stability, and tumor development. Cancer Res 72(6):1518–1528 Warner SL, Stephens BJ, Nwokenkwo S, Hostetter G, Sugeng A, Hidalgo M, Trent JM, Han H, Von Hoff DD, 2009, Validation of TPX2 as a potential therapeutic target in pancreatic cancer cells. Clin Cancer Res 15(21):6519–6528 Tonon G, Wong K-K, Maulik G, Brennan C, Feng B, Zhang Y, Khatry DB, Protopopov A, You MJ, Aguirre AJ, 2005, Highresolution genomic profiles of human lung cancer. Proc Natl Acad Sci U S A 102(27):9625–9630 Sillars-Hardebol AH, Carvalho B, Tijssen M, Belien JA, de Wit M, Delis-van Diemen PM, Ponten F, van de Wiel MA, Fijneman RJ, Meijer GA, 2012, TPX2 and AURKA promote 20q amplicon-driven colorectal adenoma to carcinoma progression. Gut 61(11):1568–1575 Chang H, Wang J, Tian Y, Xu J, Gou X, Cheng J, 2012, The TPX2 gene is a promising diagnostic and therapeutic target for cervical cancer. Oncol Rep 27(5):1353–1359 Li B, Qi XQ, Chen X, Huang X, Liu GY, Chen HR, Huang CG, Luo C, Lu YC, 2010, Expression of targeting protein for Xenopus kinesin-like protein 2 is associated with progression of human malignant astrocytoma. Brain Res 1352:200–207 Morgan-Lappe SE, Tucker LA, Huang X, Zhang Q, Sarthy AV, Zakula D, Vernetti L, Schurdak M, Wang J, Fesik SW, 2007, Identification of Ras-related nuclear protein, targeting protein for xenopus kinesin-like protein 2, and stearoyl-CoA desaturase 1 as promising cancer targets from an RNAi-based screen. Cancer research 67(9):4390–4398 Carson DA, Lois A, 1995, Cancer progression and p53. Lancet 346(8981):1009–1011 Pascreau G, Eckerdt F, Lewellyn AL, Prigent C, Maller JL, 2009, Phosphorylation of p53 is regulated by TPX2-Aurora A in xenopus oocytes. J Biol Chem 284(9):5497–5505 Marumoto T, Zhang D, Saya H, 2005, Aurora-A—a guardian of poles. Nat Rev Cancer 5(1):42–50 Ferchichi I, Sassi Hannachi S, Baccar A et al, 2013, Assessment of Aurora A kinase expression in breast cancer: A tool for early diagnosis? Dis Markers 34(2):63–69 Lee ECY, Frolov A, Li R, Ayala G, Greenberg NM, 2006, Targeting Aurora kinases for the treatment of prostate cancer. Cancer research 66(10):4996–5002 Ulisse S, Delcros JG, Baldini E, Toller M, Curcio F, Giacomelli L, Prigent C, Ambesi-Impiombato FS, D’Armiento M, Arlot- Bonnemains Y, 2006, Expression of Aurora kinases in human thyroid carcinoma cell lines and tissues. Int J Cancer 119(2):275– 282 Kufer TA, SilljeHH, Korner R, Gruss OJ, Meraldi P, Nigg EA, 2002, Human TPX2 is required for targeting Aurora-A kinase to the spindle. J Cell Biol 158(4):617–623 Bayliss R, Sardon T, Ebert J, Lindner D, Vernos I, Conti E, 2004, Determinants for Aurora-A activation and Aurora-B discrimination by TPX2. Cell Cycle 3(4):402–405 Eyers PA, Erikson E, Chen LG,Maller JL, 2003, Anovelmechanism for activation of the protein kinase Aurora A. Curr Biol CB 13(8):691 Katayama H, Sasai K, Kawai H, Yuan Z-M, Bondaruk J, Suzuki F, Fujii S, Arlinghaus RB, Czerniak BA, Sen S, 2003, Phosphorylation by aurora kinase A induces Mdm2-mediated destabilization and inhibition of p53. Nat Genet 36(1):55–62 Liu Q, Kaneko S, Yang L, Feldman RI, Nicosia SV, Chen J, Cheng JQ, 2004, Aurora-A abrogation of p53 DNA binding and transactivation activity by phosphorylation of serine 215. J Biol Chem 279(50):52175–52182 Rieder CL, 2011, Mitosis in vertebrates: the G2/M and M/A transitions and their associated checkpoints. Chromosom Res 19(3):291– 306 Sasayama T, Marumoto T, Kunitoku N, Zhang D, Tamaki N, Kohmura E, Saya H, Hirota T, 2005, Over-expression of Aurora-A targets cytoplasmic polyadenylation element binding protein and promotes mRNA polyadenylation of Cdk1 and cyclin B1. Genes Cells 10(7):627–638 Marumoto T, Hirota T, Morisaki T, Kunitoku N, Zhang D, Ichikawa Y, Sasayama T, Kuninaka S, Mimori T, Tamaki N, 2002, Roles of aurora‐A kinase in mitotic entry and G2 checkpoint in mammalian cells. Genes Cells 7(11):1173–1182 Moore JD, 2013, In the wrong place at the wrong time: does cyclin mislocalization drive oncogenic transformation? Nat Rev Cancer 13(3):201–208 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 641 EP 648 DI 10.1007/s12253-014-9743-4 PG 8 ER PT J AU Posfai, Irsai, G Illes, Mehes, G Marton, I Molnar, Cs Csipo, I Barath, S Gergely, L AF Posfai, Eva Irsai, Gabor Illes, Arpad Mehes, Gabor Marton, Imelda Molnar, Csaba Csipo, Istvan Barath, Sandor Gergely, Lajos TI Evaluation of Significance of Lymphocyte Subpopulations and Non-specific Serologic Markers in B-cell Non-Hodgkin’s Lymphoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lymphocyte subpopulations; Non-specific serologicmarkers; Non-Hodgkin’s lymphoma; Immuno-chemotherapy; Host immune system ID Lymphocyte subpopulations; Non-specific serologicmarkers; Non-Hodgkin’s lymphoma; Immuno-chemotherapy; Host immune system AB The use of rituximab brought attention to the hosts’ immune system and to the microenvironment in non- Hodgkin’s lymphoma cases. Our aim was to identify prognostic factors that can be measured easily to indicate the current state of the patient’s immune status and possible reaction against malignant cells. In the retrospective analysis (2000–2008), 66 patients diagnosed with B-cell non- Hodgkin’s lymphomas were enrolled (40 women, 26 men; mean age: 51 years).White blood cells, lymphocytes, CD3 +; CD4 +; CD8+T-cells, immunoglobulin types A; G; M, anticardiolipin antibody isotypes A; G; M; and levels of beta-2- microglobulin were measured before the initiation of the first cycle of chemotherapy, during and after 4-weeks treatment. As for CD 3+ T-lymphocytes, the absolute CD 3+ T – lymphocyte numbers were higher before (0.78×109/L) versus during (0.27×109/L) treatment, and increased percentages were detected in pre- (66.57 %) and post-treatment (75.32 %). Absolute numbers of CD 8+ T-lymphocyte levels showed reduction before (0.26×109/L) versus during (0.10× 109/L) therapy, but were elevated after (0.28×109/L) treatment, while increased percentage before (21.99 %) versus after (29.85 %), and during (24.56 %) versus after (29.85 %) therapy were seen. Average white blood cell numbers were increased before (9.71×109/L) versus during (12.07×109/L) treatment, while decreased numbers could be observed, after (5.47×109/L) treatment. IgA levels were decreased before (2.51 g/L) versus after (1.63 g/L) therapy. IgG levels were higher before (12.25 g/L) vs. after (8.64 g/L) treatment. IgM levels were decreased before (1.76 g/L) and after (0.83 g/L) as well as before (1.76 g/L) versus during (0.73 g/L) treatment. Anti-cardiolipin antibody type A level were decreased before (2.76 U/ml) versus after (2.49 U/ml) treatment. Decreased level of beta-2-microglobulin could be observed before (2.91 mg/L) versus post (2.28 mg/L) chemotherapy. Findings may provide better insight into the effects of immuno-chemotherapy on the hosts’ immune system. C1 [Posfai, Eva] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Irsai, Gabor] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Marton, Imelda] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Molnar, Csaba] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Csipo, Istvan] University of Debrecen, Medical and Health Science Center, Regional Laboratory for ImmunologyDebrecen, Hungary. [Barath, Sandor] University of Debrecen, Medical and Health Science Center, Regional Laboratory for ImmunologyDebrecen, Hungary. [Gergely, Lajos] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. RP Gergely, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM lgergely@med.unideb.hu CR Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60(5):277–300., DOI 10.3322/caac.20073 Laszlo J, Grizzle J, Jonsson U, Rundles RW, 1962, Comparative study of mannitol mustard, cyclophosphamide, and nitrogen mustard in malignant lymphomas. Cancer Chemother Rep 16:247–250 Liu WM, Meyer B, Dalgleish AG, 2009, How immunotherapy can enhance the response to other modalities and improve outcome and quality of life. J BUON 14(Suppl 1):S103–S109 Ferrara F, Ravasio R, 2008, Cost-effectiveness analysis of the addition of rituximab to CHOP in young patients with good-prognosis diffuse large-B-cell lymphoma. Clin Drug Investig 28(1):55–65 Plosker GL, Figgitt DP, 2003, Rituximab: a review of its use in non- Hodgkin’s lymphoma and chronic lymphocytic leukaemia. Drugs 63(8):803–843 Hernberg M, 1999, Lymphocyte subsets as prognostic markers for cancer patients receiving immunomodulative therapy. Med Oncol 16(3):145–153 Wahlin BE, Sander B, Christensson B, Ostenstad B, Holte H, Brown PD, Sundstrom C, Kimby E, 2012, Entourage: the immune microenvironment following follicular lymphoma. Blood Cancer J 2(1):e52., DOI 10.1038/bcj.2011.53 Herreros B, Sanchez-Aguilera A, Piris MA, 2008, Lymphoma microenvironment: culprit or innocent? Leukemia 22(1):49–58., DOI 10. 1038/sj.leu.2404970 Alvaro T, LejeuneM, Escriva P, Pons LE, Bosch R, Jaen J, Lopez C, Salvado MT, de Sanjose S, 2009, Appraisal of immune response in lymphoproliferative syndromes: a systematic review. Crit Rev Oncol Hematol 70(2):103–113., DOI 10.1016/j.critrevonc.2008.09.013 Alvaro T, de la Cruz-Merino L, Henao-Carrasco F, Villar Rodriguez JL, Vicente Baz D, CodesManuel de VillenaM, ProvencioM(2010, Tumor microenvironment and immune effects of antineoplastic therapy in lymphoproliferative syndromes. J Biomed Biotechnol., DOI 10. 1155/2010/846872 Cheson BD, Pfistner B, Juweid ME, Gascoyne RD, Specht L, Horning SJ, Coiffier B, Fisher RI, Hagenbeek A, Zucca E, Rosen ST, Stroobants S, Lister TA, Hoppe RT, Dreyling M, Tobinai K, Vose JM, Connors JM, Federico M, Diehl V, 2007, Revised response criteria for malignant lymphoma. J Clin Oncol 25(5):579–586., DOI 10.1200/JCO.2006.09.2403 Bairey O, Blickstein D, Monselise Y, Lahav J, Stark P, Prokocimer M, Nativ HM, Kirgner I, Pazgal I, Shaklai M, 2006, Antiphospholipid antibodies may be a new prognostic parameter in aggressive non-Hodgkin’s lymphoma. Eur J Haematol 76(5):384– 391., DOI 10.1111/j.1600-0609.2005.00620.x Genvresse I, Luftner D, Spath-Schwalbe E, Buttgereit F, 2002, Prevalence and clinical significance of anticardiolipin and antibeta2- glycoprotein-I antibodies in patients with non-Hodgkin’s lymphoma. Eur J Haematol 68(2):84–90 Talaulikar D, Choudhury A, Shadbolt B, Brown M, 2008, Lymphocytopenia as a prognostic marker for diffuse large B cell lymphomas. Leuk Lymphoma 49(5):959–964., DOI 10.1080/ 10428190801959026 Oki Y, Yamamoto K, Kato H, Kuwatsuka Y, Taji H, Kagami Y, Morishima Y, 2008, Low absolute lymphocyte count is a poor prognostic marker in patients with diffuse large B-cell lymphoma and suggests patients’ survival benefit from rituximab. Eur J Haematol 81(6):448–453., DOI 10.1111/j.1600-0609.2008.01129.x Porrata LF, Ristow K, Habermann TM, Witzig TE, Inwards DJ, Markovic SN, 2009, Absolute lymphocyte count at the time of first relapse predicts survival in patients with diffuse large B-cell lymphoma. Am J Hematol 84(2):93–97., DOI 10.1002/ajh.21337 Cox MC, Nofroni I, Ruco L, Amodeo R, Ferrari A, La Verde G, Cardelli P, Montefusco E, Conte E, Monarca B, Aloe-Spiriti MA, 2008, Low absolute lymphocyte count is a poor prognostic factor in diffuse-large-B-cell-lymphoma. Leuk Lymphoma 49(9):1745–1751., DOI 10.1080/10428190802226425 Gergely L, Vancsa A, Miltenyi Z, Simon Z, Barath S, Illes A, 2011, Pretreatment T lymphocyte numbers are contributing to the prognostic significance of absolute lymphocyte numbers in B-cell non- Hodgkins lymphomas. Pathol Oncol Res 17(2):249–255., DOI 10. 1007/s12253-010-9306-2 Simon Z, Illes A,Miltenyi Z,Magyari F, Varoczy L, Peter N, Gergely L, 2012, Immunologic changes in diffuse large B-cell lymphomas after rituximab-CHOP treatment: own data and review of the literature. Orv Hetil 153(42):1658–1666., DOI 10.1556/OH.2012.29471 Varoczy L, Gergely L, Miltenyi Z, Aleksza M, Illes A, 2005, Can CD3+/HLA-DR+ activated T cells predict the prognosis of non- Hodgkin’s lymphoma patients? Immunol Lett 97(1):155–157., DOI 10.1016/j.imlet.2004.10.005 Czuczman MS, Grillo-Lopez AJ, Alkuzweny B, Weaver R, Larocca A, McLaughlin P, 2006, Prognostic factors for non-Hodgkin’s lymphoma patients treated with chemotherapy may not predict outcome in patients treated with rituximab. Leuk Lymphoma 47(9):1830– 1840., DOI 10.1080/10428190600709523 Wahlin BE, Sander B, Christensson B, Kimby E, 2007, CD8+ T-cell content in diagnostic lymph nodes measured by flow cytometry is a predictor of survival in follicular lymphoma. Clin Cancer Res 13(2 Pt 1):388–397., DOI 10.1158/1078-0432.CCR-06-1734 Yano Y, Ueda Y, Itoh T, Fuji N, Okugawa K, Naito K, Imura K, Kohara J, Hayashi T, Nakane K,Matsuura Y, Kawai K, Yamagishi H, 2006, A new strategy using autologous dendritic cells and lymphokine-activated killer cells for cancer immunotherapy: efficient maturation of DCs by co-culture with LAK cells in vitro. Oncol Rep 16(1):147–152 Imura K, Ueda Y, Hayashi T, Itoh T, Shimizu K, Tamai H, Yano Y, Naito K, Kohara J, Nakane K, Matsuura Y, Takeda A, Takeda T, Kawai K, Yamagishi H, 2006, Induction of cytotoxic T lymphocytes against human cancer cell lines using dendritic cell-tumor cell hybrids generated by a newly developed electrofusion technique. Int J Oncol 29(3):531–539 Melbye M, Smedby KE, Lehtinen T, Rostgaard K, Glimelius B, Munksgaard L, Schollkopf C, Sundstrom C, Chang ET, Koskela P, Adami HO, Hjalgrim H, 2007, Atopy and risk of non-Hodgkin lymphoma. J Natl Cancer Inst 99(2):158–166., DOI 10.1093/jnci/ djk019 Gergely L, Danko A, Csipo I, Varoczy L, Sipka S, Zeher M, Illes A, 2005, Antibodies against extractable nuclear antigen in non- Hodgkin lymphoma patients. Scand J Immunol 61(4):343–346., DOI 10.1111/j.1365-3083.2005.01567.x NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 649 EP 654 DI 10.1007/s12253-014-9744-3 PG 6 ER PT J AU Gu, JM Jang, IB AF Gu, Jin Mi Jang, Ik Byung TI Clinicopathologic Significance of Sox2, CD44 and CD44v6 Expression in Intrahepatic Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intrahepatic cholangiocarcinoma; Sox2; CD44; CD44v6; Survival ID Intrahepatic cholangiocarcinoma; Sox2; CD44; CD44v6; Survival AB Embryonic stem cells (ESC) and cancer stem cells (CSC) have a capacity for self-renewal and differentiation into multiple cell lineages. Sox2 plays a critical role in ESC and has been shown to participate in carcinogenesis and tumor progression in many human cancers. CD44 and CD44v6 are putative CSC markers and their association with tumor progression, metastasis, and tumor relapse after treatment has been demonstrated. We evaluated the immunoexpression of Sox2, CD44, and CD44v6 in 85 cases of Intrahepatic cholangiocarcinomas (IHCC) and assessed their prognostic significance. Sox2 expression showed a significant association with lymph node metastasis (p=0.025), T4 stage (p= 0.046), and worse overall survival (p=0.047). Greater expression of Sox2 was observed in IHCC with poor differentiation, vascular invasion, and stage IV, without statistical significance (p>0.05). CD44 expression showed an association with periductal infiltrative type (p=0.034), poor differentiation (p=0.012), and vascular invasion (p=0.009). CD44v6 expression was evident in patients with stage IV (p=0.019). These results demonstrated that Sox2 expression is associated with aggressive behavior and poor overall survival in IHCC. C1 [Gu, Jin Mi] Yeungnam University, College of Medicine, Department of Pathology, 170, Hyeonchung-ro, Nam-guDaegu, South Korea. [Jang, Ik Byung] Yeungnam University, College of Medicine, Department of Internal MedicineDaegu, South Korea. RP Gu, JM (reprint author), Yeungnam University, College of Medicine, Department of Pathology, Daegu, South Korea. EM mjgu@yu.ac.kr CR Zhang C, Bai DS, Huang XYet al, 2013, Prognostic significance of Capn4 overexpression in intrahepatic cholangiocarcinoma. PLoS One 8:e54619 Ruan J,Wei B, Xu Z et al, 2013, Predictive value of Sox2 expression in transurethral resection specimens in patients with T1 bladder cancer. Med Oncol 30:445., DOI 10.1007/s12032-012-0445-z Guo Y, Liu S, Wang P et al, 2011, Expression profile of embryonic stem cell-associated genes Oct4, Sox2 and Nanog in human gliomas. Histopathology 59:763–775 Ling GQ, Chen DB, Wang BQ et al, 2012, Expression of the pluripotency markers Oct3/4, Nanog and Sox2 in human breast cancer cell lines. Oncol Lett 4:1264–1268 Ben-Porath I, Thomson MW, Carey VJ et al, 2008, An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet 40:499–507 Kitamura H, Torigoe T, Hirohashi Yet al, 2013, Prognostic impact of the expression of ALDH1 and SOX2 in urothelial cancer of the upper urinary tract. Mod Pathol 26:117–124 Brcic L, Sherer CK, Shuai Y et al, 2012, Morphologic and clinicopathologic features of lung squamous cell carcinomas expressing Sox2. Am J Clin Pathol 138:712–718 Neumann J, Bahr F, Horst D et al, 2011, SOX2 expression correlates with lymph-node metastases and distant spread in right-sided colon cancer. BMC Cancer 11:518., DOI 10.1186/1471-2407-11-518 Lengerke C, Fehm T, Kurth R et al, 2011, Expression of the embryonic stem cell marker SOX2 in early-stage breast carcinoma. BMC Cancer 11:42., DOI 10.1186/1471-2407-11-42 Rodriguez-Pinilla SM, Sarrio D,Moreno-Bueno G et al, 2007, Sox2: a possible driver of the basal-like phenotype in sporadic breast cancer. Mod Pathol 20:474–481 Han X, Fang X, Lou X et al, 2012, Silencing SOX2 induced mesenchymal-epithelial transition and its expression predicts liver and lymph node metastasis of CRC patients. PLoS One 7:e41335 Huang P, Qiu J, Li B et al, 2011, Role of Sox2 and Oct4 in predicting survival of hepatocellular carcinoma patients after hepatectomy. Clin Biochem 44:582–589 Wilbertz T, Wagner P, Petersen K et al, 2011, SOX2 gene amplification and protein overexpression are associated with better outcome in squamous cell lung cancer. Mod Pathol 24:944–953 Velcheti V, Schalper K, Yao X et al, 2013, High SOX2 levels predict better outcome in non-small cell lung carcinomas. PLoS One 8: e61427 Otsubo T, Akiyama Y, Yanagihara K et al, 2008, SOX2 is frequently downregulated in gastric cancers and inhibits cell growth through cell-cycle arrest and apoptosis. Br J Cancer 98:824–831 Matsuoka J, Yashiro M, Sakurai K et al, 2012, Role of the stemness factors sox2, oct3/4, and nanog in gastric carcinoma. J Surg Res 174: 130–135 Nanashima A, Hatachi G, Tsuchiya T et al, 2013, Clinical significances of cancer stem cells markers in patients with intrahepatic cholangiocarcinoma who underwent hepatectomy. Anticancer Res 33:2107–2114 Wakamatsu Y, Sakamoto N, Oo HZ et al, 2012, Expression of cancer stem cell markers ALDH1, CD44 and CD133 in primary tumor and lymph node metastasis of gastric cancer. Pathol Int 62:112–119 Palagani V, El Khatib M, Krech T et al, 2012, Decrease of CD44- positive cells correlates with tumor response to chemotherapy in patients with gastrointestinal cancer. Anticancer Res 32:1747–1755 Okayama H, Kumamoto K, Saitou K et al, 2009, CD44v6, MMP-7 and nuclear Cdx2 are significant biomarkers for prediction of lymph node metastasis in primary gastric cancer. Oncol Rep 22:745–755 Klatte T, Seligson DB, Rao JYet al, 2010, Absent CD44v6 expression is an independent predictor of poor urothelial bladder cancer outcome. J Urol 183:2403–2408 Sun C, Sun L, Li Y et al, 2013, Sox2 expression predicts poor survival of hepatocellular carcinoma patients and it promotes liver cancer cell invasion by activating Slug. Med Oncol 30:503., DOI 10. 1007/s12032-013-0503-1 Pongcharoen P, Jinawath A, Tohtong R, 2011, Silencing of CD44 by siRNA suppressed invasion, migration and adhesion to matrix, but not secretion of MMPs, of cholangiocarcinoma cells. Clin Exp Metastasis 28:827–839 Zlobec I, Gunthert U, Tornillo L, Iezzi G, Baumhoer D, Terracciano L et al, 2009, Systematic assessment of the prognostic impact of membranous CD44v6 protein expression in colorectal cancer. Histopathology 55:564–575 Avoranta ST, Korkeila EA, Syrjanen KJ et al, 2012, Lack of CD44 variant 6 expression in rectal cancer invasive front associates with early recurrence. World J Gastroenterol 18:4549–4556 da Cunha CB, Oliveira C,Wen X et al, 2010, De novo expression of CD44 variants in sporadic and hereditary gastric cancer. Lab Investig 90:1604–1614 Afify AM, Tate S, Durbin-Johnson B et al, 2011, Expression of CD44s and CD44v6 in lung cancer and their correlation with prognostic factors. Int J Biol Markers 26:50–57 Ashida K, Terada T, Kitamura Y et al, 1998, Expression of Ecadherin, alpha-catenin, beta-catenin, and CD44, standard and variant isoforms, in human cholangiocarcinoma: an immunohistochemical study. Hepatology 27:974–982 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 655 EP 660 DI 10.1007/s12253-014-9745-2 PG 6 ER PT J AU Zhang, ShL Yuan, F Guan, X Li, J Liu, LX Sun, J Liu, B Ma, W Deng, MF AF Zhang, Shun Lu Yuan, Fang Guan, Xuan Li, Juan Liu, Lian Xin Sun, Jing Liu, Bo Ma, Wei Deng, Mei Feng TI Association of Genetic Polymorphisms in HSD17B1, HSD17B2 and SHBG Genes with Hepatocellular Carcinoma Risk SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SHBG; HSD17B1; HSD17B2; HCC ID SHBG; HSD17B1; HSD17B2; HCC AB Genetic polymorphisms of enzymes involved in estrogen synthesizing/transporting can influence the risk of hormone-dependent diseases. The incidence rate and relative risk for hepatocellular carcinoma (HCC) are higher in men than in women. This study was conducted to explore the relationship of single nucleotide polymorphisms (SNPs) in 17 β-Hydroxysteroid dehydrogenases (HSD17B1 and HSD17B2) and sex hormone-binding globulin (SHBG) genes with the risk of HCC within Chinese Han population. Polymorphisms of HSD17B1 rs676387, HSD17B2 rs8191246 and SHBG rs6259 were genotyped in 253 HCC patients and 438 healthy control subjects using the polymerase chain reaction–restriction fragment length polymorphism (PCR–RFLP). Significantly increased HCC risk was found to be associated with T allele of rs676387 and G allele of rs8191246. Increased HCC risks were found in different genetic model (TT genotype in a recessive model, T allele carriers in a dominant model, TT genotype and TG genotype in a codominant model for HSD17B1 rs676387, G allele carriers in a dominant model and AG genotype in a codominant model for HSD17B2 rs8191246, respectively). No association between SHBG rs6259 and HCC risk was observed. The present study provided evidence that HSD17B1 rs676387 and HSD17B2 rs8191246 were association with HCC development. Further studies in diverse ethnic population with larger sample size were recommended to confirm the findings. C1 [Zhang, Shun Lu] Cheng Du Medical College, Department of Pathology and Pathophysiology, 610500 Chengdu, China. [Yuan, Fang] Sichuan University, West China School of Preclinical and Forensic Medicine, Department of Immunology, 610041 Chengdu, China. [Guan, Xuan] Chengdu Medical College, Department of Experimental Technology, 610500 Chengdu, China. [Li, Juan] Cheng Du Medical College, Department of Pathology and Pathophysiology, 610500 Chengdu, China. [Liu, Lian Xin] Cheng Du Medical College, Department of Pathology and Pathophysiology, 610500 Chengdu, China. [Sun, Jing] Cheng Du Medical College, Department of Pathology and Pathophysiology, 610500 Chengdu, China. [Liu, Bo] Cheng Du Medical College, Department of Lab Medicine, 610500 Chengdu, China. [Ma, Wei] Cheng Du Medical College, Department of Lab Medicine, 610500 Chengdu, China. [Deng, Mei Feng] Cheng Du Medical College, Department of Pathology and Pathophysiology, 610500 Chengdu, China. RP Zhang, ShL (reprint author), Cheng Du Medical College, Department of Pathology and Pathophysiology, 610500 Chengdu, China. EM zhangls2012@163.com CR Parkin DM, Bray F, Ferlay J, Pisani P, 2001, Estimating the world cancer burden: Globocan 2000. Int J Cancer 94(2):153–156 Bosch FX, Ribes J, Cleries R, Diaz M, 2005, Epidemiology of hepatocellular carcinoma. Clin Liver Dis 9(2):191–211., DOI 10. 1016/j.cld.2004.12.009 Tan A, Yeh SH, Liu CJ, Cheung C, Chen PJ, 2008, Viral hepatocarcinogenesis: from infection to cancer. Liver Int Off J Int Assoc Study Liver 28(2):175–188., DOI 10.1111/j.1478-3231.2007. 01652.x Gomaa AI, Khan SA, Toledano MB, Waked I, Taylor-Robinson SD, 2008, Hepatocellular carcinoma: epidemiology, risk factors and pathogenesis. World J Gastroenterol 14(27):4300–4308 De Maria N, Manno M, Villa E, 2002, Sex hormones and liver cancer. Mol Cell Endocrinol 193(1–2):59–63 Giannitrapani L, Soresi M, La Spada E, Cervello M,D’Alessandro N, Montalto G, 2006, Sex hormones and risk of liver tumor. Ann N Y Acad Sci 1089:228–236., DOI 10.1196/annals.1386.044 Pasqualini JR, 2004, The selective estrogen enzyme modulators in breast cancer: a review. Biochim Biophys Acta 1654(2):123–143., DOI 10.1016/j.bbcan.2004.03.001 Vihko P, IsomaaV, Ghosh D, 2001, Structure and function of 17betahydroxysteroid dehydrogenase type 1 and type 2. Mol Cell Endocrinol 171(1–2):71–76 Tsuchiya Y, Nakajima M, Yokoi T, 2005, Cytochrome P450- mediated metabolism of estrogens and its regulation in human. Cancer Lett 227(2):115–124., DOI 10.1016/j.canlet.2004.10.007 Berube D, Seralini GE, Gagne R, Hammond GL, 1990, Localization of the human sex hormone-binding globulin gene, SHBG, to the short arm of chromosome 17, 17p12–p13). Cytogenet Cell Genet 54(1–2):65–67 Mahato D, Goulding EH, Korach KS, Eddy EM, 2001, Estrogen receptor-alpha is required by the supporting somatic cells for spermatogenesis. Mol Cell Endocrinol 178(1–2):57–63 Kato S, Masuhiro Y, Watanabe M, Kobayashi Y, Takeyama KI, Endoh H, Yanagisawa J, 2000, Molecular mechanism of a crosstalk between oestrogen and growth factor signalling pathways. Genes Cells 5(8):593–601 Kahn SM, Hryb DJ, Nakhla AM, Romas NA, Rosner W, 2002, Sex hormone-binding globulin is synthesized in target cells. J Endocrinol 175(1):113–120 Fortunati N, Becchis M, Catalano MG, Comba A, Ferrera P, Raineri M, Berta L, Frairia R, 1999, Sex hormone-binding globulin, its membrane receptor, and breast cancer: a new approach to the modulation of estradiol action in neoplastic cells. J Steroid Biochem Mol Biol 69(1–6):473–479 Cui Y, Shu XO, Cai Q, Jin F, Cheng JR, Cai H, Gao YT, Zheng W, 2005, Association of breast cancer risk with a common functional polymorphism, Asp327Asn, in the sex hormone-binding globulin gene. Cancer Epidemiol Biomarkers Prev 14(5):1096–1101., DOI 10. 1158/1055-9965.EPI-04-0721 Kataoka N, Cai Q, Xu WH, Xiang YB, Cai H, Zheng W, Shu XO, 2007, Association of endometrial cancer risk with a functional polymorphism, Asp(327)Asn, in the sex hormone-binding globulin gene. Cancer 109(7):1296–1302., DOI 10.1002/cncr.22531 Garcia-Closas M, Brinton LA, Lissowska J, Richesson D, Sherman ME, Szeszenia-Dabrowska N, Peplonska B, Welch R, Yeager M, Zatonski W, Chanock SJ, 2007, Ovarian cancer risk and common variation in the sex hormone-binding globulin gene: a populationbased case–control study. BMC Cancer 7:60., DOI 10.1186/1471- 2407-7-60 Berndt SI, Chatterjee N, Huang WY, Chanock SJ, Welch R, Crawford ED, Hayes RB, 2007, Variant in sex hormone-binding globulin gene and the risk of prostate cancer. Cancer Epidemiol Biomarkers Prev 16(1):165–168., DOI 10.1158/1055-9965.EPI-06- 0689 Cong RJ, Huang ZY, Cong L, Ye Y,Wang Z, Zha L, Cao LP, SuXW, Yan J, Li YB, 2012, Polymorphisms in genes HSD17B1 and HSD17B2 and uterine leiomyoma risk in Chinese women. Arch Gynecol Obstet 286(3):701–705., DOI 10.1007/s00404-012-2328-0 Napoli N, Varadharajan A, Rini GB, Del Fiacco R, Yarramaneni J, Mumm S, Villareal DT, Armamento-Villareal R, 2009, Effects of polymorphisms of the sex hormone-binding globulin, SHBG, gene on free estradiol and bone mineral density. Bone 45(6):1169–1174., DOI 10.1016/j.bone.2009.08.001 Johnson FL, Lerner KG, Siegel M, Feagler JR, Majerus PW, Hartmann JR, Thomas ED, 1972, Association of androgenicanabolic steroid therapy with development of hepatocellular carcinoma. Lancet 2(7790):1273–1276 Baum JK, Bookstein JJ, Holtz F, Klein EW, 1973, Possible association between benign hepatomas and oral contraceptives. Lancet 2(7835):926–929 Peltoketo H, Luu-The V, Simard J, Adamski J, 1999, 17betahydroxysteroid dehydrogenase, HSD)/17-ketosteroid reductase, KSR, family; nomenclature and main characteristics of the 17HSD/KSR enzymes. J Mol Endocrinol 23(1):1–11 Normand T, Narod S, Labrie F, Simard J, 1993, Detection of polymorphisms in the estradiol 17 beta-hydroxysteroid dehydrogenase II gene at the EDH17B2 locus on 17q11-q21. Hum Mol Genet 2(4): 479–483 Mannermaa A, Peltoketo H, Winqvist R, Ponder BA, Kiviniemi H, Easton DF, Poutanen M, Isomaa V, Vihko R, 1994, Human familial and sporadic breast cancer: analysis of the coding regions of the 17 beta-hydroxysteroid dehydrogenase 2 gene, EDH17B2, using a single-strand conformation polymorphism assay. Hum Genet 93(3): 319–324 Feigelson HS, Cox DG, Cann HM, Wacholder S, Kaaks R, Henderson BE, Albanes D, Altshuler D, Berglund G, Berrino F, Bingham S, Buring JE, Burtt NP, Calle EE, Chanock SJ, Clavel- Chapelon F, Colditz G, Diver WR, Freedman ML, Haiman CA, Hankinson SE, Hayes RB, Hirschhorn JN, Hunter D, Kolonel LN, Kraft P, LeMarchand L, Linseisen J,ModiW, Navarro C, Peeters PH, Pike MC, Riboli E, Setiawan VW, Stram DO, Thomas G, Thun MJ, Tjonneland A, Trichopoulos D, 2006, Haplotype analysis of the HSD17B1 gene and risk of breast cancer: a comprehensive approach to multicenter analyses of prospective cohort studies. Cancer Res 66(4):2468–2475., DOI 10.1158/0008-5472.can-05-3574 Kraft P, Pharoah P, Chanock SJ, Albanes D, Kolonel LN, Hayes RB, Altshuler D, Andriole G, Berg C, Boeing H, Burtt NP, Bueno-de- Mesquita B, Calle EE, Cann H, Canzian F, Chen YC, Crawford DE, Dunning AM, Feigelson HS, Freedman ML, Gaziano JM, Giovannucci E, Gonzalez CA, Haiman CA, Hallmans G, Henderson BE, Hirschhorn JN, Hunter DJ, Kaaks R, Key T, Le Marchand L, Ma J, Overvad K, Palli D, Pike MC, Riboli E, Rodriguez C, Setiawan WV, Stampfer MJ, Stram DO, Thomas G, ThunMJ, Travis R, Trichopoulou A, Virtamo J,Wacholder S, 2005, Genetic variation in the HSD17B1 gene and risk of prostate cancer. PLoS Genet 1(5):e68., DOI 10.1371/journal.pgen.0010068 Gaudet MM, Chanock S, Dunning A, Driver K, Brinton LA, Lissowska J, Peplonska B, Pharoah P, Garcia-Closas M, 2008, HSD17B1 genetic variants and hormone receptor-defined breast cancer. Cancer Epidemiol Biomark Prev 17(10):2766–2772., DOI 10. 1158/1055-9965.epi-07-2891 Husen B, Psonka N, Jacob-Meisel M, Keil C, Rune GM, 2000, Differential expression of 17beta-hydroxysteroid dehydrogenases types 2 and 4 in human endometrial epithelial cell lines. J Mol Endocrinol 24(1):135–144 Moghrabi N, Head JR, Andersson S, 1997, Cell type-specific expression of 17 beta-hydroxysteroid dehydrogenase type 2 in human placenta and fetal liver. J Clin Endocrinol Metab 82(11): 3872–3878 Andersson S, Moghrabi N, 1997, Physiology and molecular genetics of 17 beta-hydroxysteroid dehydrogenases. Steroids 62(1):143–147 Elo JP, Harkonen P, Kyllonen AP, Lukkarinen O, Poutanen M, Vihko R, Vihko P, 1997, Loss of heterozygosity at 16q24.1-q24.2 is significantly associated with metastatic and aggressive behavior of prostate cancer. Cancer Res 57(16):3356–3359 SuMT, Chen CH, Kuo PH, Hsu CC, Lee IW, Pan HA, Chen YT, Kuo PL, 2010, Polymorphisms of estrogen-related genes jointly confer susceptibility to human spermatogenic defect. Fertil Steril 93(1):141– 149., DOI 10.1016/j.fertnstert.2008.09.030 Dunning AM, Dowsett M, Healey CS, Tee L, Luben RN, Folkerd E, Novik KL, Kelemen L, Ogata S, Pharoah PD, Easton DF, Day NE, Ponder BA, 2004, Polymorphisms associated with circulating sex hormone levels in postmenopausal women. J Natl Cancer Inst 96(12):936–945 Forsti A, Jin Q, Grzybowska E, Soderberg M, Zientek H, Sieminska M, Rogozinska-Szczepka J, Chmielik E, Utracka-Hutka B, Hemminki K, 2002, Sex hormone-binding globulin polymorphisms in familial and sporadic breast cancer. Carcinogenesis 23(8):1315– 1320 Cunningham JM, Hebbring SJ, McDonnell SK, Cicek MS, Christensen GB, Wang L, Jacobsen SJ, Cerhan JR, Blute ML, Schaid DJ, Thibodeau SN, 2007, Evaluation of genetic variations in the androgen and estrogen metabolic pathways as risk factors for sporadic and familial prostate cancer. Cancer Epidemiol Biomarkers Prev 16(5):969–978., DOI 10.1158/1055-9965.epi-06-0767 Shimizu I, Ito S, 2007, Protection of estrogens against the progression of chronic liver disease. Hepatol Res Off J Japan Soc Hepatol 37(4):239–247., DOI 10.1111/j.1872-034X.2007.00032.x NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 661 EP 666 DI 10.1007/s12253-014-9746-1 PG 6 ER PT J AU Shin, E Yu, YD Kim, DS Won, HN AF Shin, Eun Yu, Young-Dong Kim, Dong-Sik Won, Hee Nam TI Adiponectin Receptor Expression Predicts Favorable Prognosis in Cases of Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Adiponectin; Immunohistochemistry; Prognosis ID Hepatocellular carcinoma; Adiponectin; Immunohistochemistry; Prognosis AB Obesity influences risk, progression and prognosis of various cancers including hepatocellular carcinoma (HCC). Adipose-tissue-derived adipokines has been considered to be involved in tumorigenesis and adiponecin, one such adipokine, has antiproliferative effect on obesity-related malignancies, though variable signal pathway mediated by adiponectin receptors-AdipoR1 and AdipoR2. In this study, we investigated expression of adiponectin and adiponectin receptors in tumor and non-tumorous hepatic tissues of HCC patients and its clinicopathological significance. We collected 75 HCC tissues and 70 non-tumorous hepatic tissues from HCC patients who underwent surgical resection. The tissue microarrays were constructed and immunohistochemical study for adiponectin, AdipoR1 and AipoR2 was performed. Adiponectin and AdipoR1 expression rates were significantly lower in HCC than non-neoplastic hepatic tissues (82.7 % vs. 97.1 % and 24.0 % vs. 90 %, P=0.005 and <0.001, respectively). Immunopositivity for adiponectin was associated with small tumor size, low Edmonson-Steiner grade and absence of other organ invasion (P=0.015, 0.021 and 0.028, respectively). AdipoR1 expression had association with absence of vascular invasion (P=0.028) and AdipoR2 expression was correlated with lower histologic grade and low pathologic Tstage (P=0.003 and 0.008, respectively). Cox regression analysis revealed that low expression of AdipoR1 and AdipoR2 were associated with increased risk of recurrence and death, respectively (hazard ration = 3.222 and 14.797, respectively). These findings suggest that loss of adiponectin, and adiponectin receptors expression is associated with aggressive clinicopathological features of HCC and AdipoR1 and AdipoR2 might serve as the independent prognostic factors for HCC patients. C1 [Shin, Eun] Seoul National University Bundang Hospital, Department of Pathology, 173-82 Gumiro, Bundang-gu, Seongnam, 463-707 Gyeonggi, South Korea. [Yu, Young-Dong] Korea University, College of Medicine, Department of Surgery, 126-1 Anam-dong 5 ga, Seongbuk-gu, 163-705 Seoul, South Korea. [Kim, Dong-Sik] Korea University, College of Medicine, Department of Surgery, 126-1 Anam-dong 5 ga, Seongbuk-gu, 163-705 Seoul, South Korea. [Won, Hee Nam] Korea University, College of Medicine, Department of Pathology, 126-1 Anam-dong 5 ga, Seongbuk-gu, 163-705 Seoul, South Korea. RP Won, HN (reprint author), Korea University, College of Medicine, Department of Pathology, 163-705 Seoul, South Korea. EM nhw@korea.ac.kr CR Wolk A, Gridley G, Svensson M, Nyren O, McLaughlin JK, Fraumeni JF, Adam HO, 2001, A prospective study of obesity and cancer risk, Sweden). Cancer Causes Control 12:13–21 Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M, 2008, Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet 371:569– 578 Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ, 2003, Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med 348:1625–1638 Moller H, Mellemgaard A, Lindvig K, Olsen JH, 1994, Obesity and cancer risk: a Danish record-linkage study. Eur J Cancer 30A:344– 350 Kadowaki T, Yamauchi T, 2005, Adiponectin and adiponectin receptors. Endocr Rev 26:439–451 Mantzoros C, Petridou E, Dessypris N, Chavelas C, Dalamaga M, Alexe DM, Papadiamantis Y, Markopoulos C, Spanos E, Chrousos G, Trichopoulos D, 2004, Adiponectin and breast cancer risk. J Clin Endocrinol Metab 89:1102–1107 Wei EK, Giovannucci E, Fuchs CS, Willett WC, Mantzoros CS, 2005, Low plasma adiponectin levels and risk of colorectal cancer in men: a prospective study. J Natl Cancer Inst 97:1688–1694 Goktas S, Yilmaz MI, Caglar K, Sonmez A, Kilic S, Bedir S, 2005, Prostate cancer and adiponectin. Urology 65:1168–1172 Sugiyama M, Takahashi H, Hosono K, Endo H, Kato S, Yoneda K, Nozaki Y, Fujita K, Yoneda M, Wada K, Nakagama H, Nakajima A, 2009, Adiponectin inhibits colorectal cancer cell growth through the AMPK/mTOR pathway. Int J Oncol 34:339–344 Dos Santos E, Benaitreau D, Dieudonne MN, Leneveu MC, Serazin V, Giudicelli Y, Pecquery R, 2008, Adiponectin mediates an antiproliferative response in human MDA-MB 231 breast cancer cells. Oncol Rep 20:971–977 Arditi JD, Venihaki M, Karalis KP, Chrousos GP, 2007, Antiproliferative effect of adiponectin on MCF7 breast cancer cells: a potential hormonal link between obesity and cancer. Horm Metab Res 39:9–13 Bub JD, Miyazaki T, Iwamoto Y, 2006, Adiponectin as a growth inhibitor in prostate cancer cells. Biochem Biophys Res Commun 340:1158–1166 Kang JH, Lee YY, Yu BY, Yang BS, Cho KH, Yoon DK, Roh YK, 2005, Adiponectin induces growth arrest and apoptosis of MDAMB- 231 breast cancer cell. Arch Pharm Res 28:1263–1269 Dieudonne MN, Bussiere M, Dos Santos E, Leneveu MC, Giudicelli Y, Pecquery R, 2006, Adiponectin mediates antiproliferative and apoptotic responses in human MCF7 breast cancer cells. Biochem Biophys Res Commun 345:271–279 Goldstein BJ, Scalia R, 2004, Adiponectin: a novel adipokine linking adipocytes and vascular function. J Clin Endocrinol Metab 89:2563– 2568 Luo Z, Saha AK, Xiang X, Ruderman NB, 2005, AMPK, the metabolic syndrome and cancer. Trends Pharmacol Sci 26:69–76 Chou SH, Tseleni-Balafouta S, Moon HS, Chamberland JP, Liu X, Kavantzas N,Mantzoros CS, 2010, Adiponectin receptor expression in human malignant tissues. Horm Cancer 1:136–145 Petridou ET, Mitsiades N, Gialamas S, Angelopoulos M, Skalkidou A, Dessypris N, Hsi A, Lazaris N, Polyzos A, Syrigos C, Brennan AM, Tseleni-Balafouta S, Mantzoros CS, 2007, Circulating adiponectin levels and expression of adiponectin receptors in relation to lung cancer: two case– control studies. Oncology 73:261–269 Pfeiler G, Hudelist G,Wulfing P, Mattsson B, Konigsberg R, Kubista E, Singer CF, 2010, Impact of AdipoR1 expression on breast cancer development. Gynecol Oncol 117:134–138 Dalamaga M, Migdalis I, Fargnoli JL, Papadavid E, Bloom E, Mitsiades N, Karmaniolas K, Pelecanos N, Tseleni-Balafouta S, Dionyssiou-Asteriou A, Mantzoros CS, 2009, Pancreatic cancer expresses adiponectin receptors and is associated with hypoleptinemia and hyperadiponectinemia: a case–control study. Cancer Causes Control 20:625–633 Saxena NK, Fu PP, Nagalingam A, Wang J, Handy J, Cohen C, Tighiouart M, Sharma D, Anania FA, 2010, Adiponectin modulates C-jun N-terminal kinase and mammalian target of rapamycin and inhibits hepatocellular carcinoma. Gastroenterology 139:1762–1773, 1773 e1761-1765 Sharma D,Wang J, Fu PP, Sharma S, Nagalingam A,Mells J, Handy J, Page AJ, Cohen C, Anania FA, Saxena NK, 2010, Adiponectin antagonizes the oncogenic actions of leptin in hepatocellular carcinogenesis. Hepatology 52:1713–1722 Shen C, Zhao CY, Zhang R, Qiao L, 2012, Obesity-related hepatocellular carcinoma: roles of risk factors altered in obesity. Front Biosci 17:2356–2370 Caldwell SH, Crespo DM, Kang HS, Al-Osaimi AM, 2004, Obesity and hepatocellular carcinoma. Gastroenterology 127:S97–S103 YuMW, ShihWL, Lin CL, Liu CJ, Jian JW, Tsai KS, Chen CJ, 2008, Body-mass index and progression of hepatitis B: a population-based cohort study in men. J Clin Oncol 26:5576–5582 Lai MS, Hsieh MS, Chiu YH, Chen TH, 2006, Type 2 diabetes and hepatocellular carcinoma: a cohort study in high prevalence area of hepatitis virus infection. Hepatology 43:1295–1302 Otani K, Kitayama J, Kamei T, Soma D, Miyato H, Yamauchi T, Kadowaki T, Nagawa H, 2010, Adiponectin receptors are downregulated in human gastric cancer. J Gastroenterol 45:918–927 Michalakis K, Williams CJ, Mitsiades N, Blakeman J, Balafouta- Tselenis S, Giannopoulos A, Mantzoros CS, 2007, Serum adiponectin concentrations and tissue expression of adiponectin receptors are reduced in patients with prostate cancer: a case control study. Cancer Epidemiol Biomarkers Prev 16:308–313 Chen DC, Chung YF, Yeh YT, Chaung HC, Kuo FC, Fu OY, Chen HY, HouMF, Yuan SS, 2006, Serum adiponectin and leptin levels in Taiwanese breast cancer patients. Cancer Lett 237:109–114 Petridou E, Mantzoros C, Dessypris N, Koukoulomatis P, Addy C, Voulgaris Z, Chrousos G, Trichopoulos D, 2003, Plasma adiponectin concentrations in relation to endometrial cancer: a case–control study in Greece. J Clin Endocrinol Metab 88:993–997 Kim AY, Lee YS, Kim KH, Lee JH, Lee HK, Jang SH, Kim SE, Lee GY, Lee JW, Jung SA, Chung HY, Jeong S, Kim JB, 2010, Adiponectin represses colon cancer cell proliferation via AdipoR1- and -R2-mediated AMPK activation. Mol Endocrinol 24:1441–1452 Kelesidis I, Kelesidis T, Mantzoros CS, 2006, Adiponectin and cancer: a systematic review. Br J Cancer 94:1221–1225 Tsukada T, Fushida S, Harada S, Terai S, Yagi Y, Kinoshita J, Oyama K, Tajima H, Fujita H, Ninomiya I, Fujimura T, Ohta T, 2011, Adiponectin receptor-1 expression is associated with good prognosis in gastric cancer. J Exp Clin Cancer Res 30:107 Abdul-Ghafar J, Oh SS, Park SM,Wairagu P, Lee SN, Jeong Y, Eom M, Yong SJ, Jung SH, 2013, Expression of adiponectin receptor 1 is indicative of favorable prognosis in non-small cell lung carcinoma. Tohoku J Exp Med 229:153–162 Yamamoto N, Oshima T, Yoshihara K, Sato T, Yamada R, Fujii S, Nagano Y, Shiozawa M, Akaike M, Wada N, Rino Y, Kunisaki C, Masuda M, Tanaka K, Imada T, 2009, Reduced expression of the AdipoR1 gene is correlated with venous invasion in colorectal cancer. Mol Med Rep 2:555–559 Byeon JS, Jeong JY, Kim MJ, Lee SM, Nam WH, Myung SJ, Kim JG, Yang SK, Kim JH, Suh DJ, 2010, Adiponectin and adiponectin receptor in relation to colorectal cancer progression. Int J Cancer 127: 2758–2767 Ishikawa M, Kitayama J, Yamauchi T, Kadowaki T, Maki T, Miyato H, Yamashita H, Nagawa H, 2007, Adiponectin inhibits the growth and peritoneal metastasis of gastric cancer through its specific membrane receptors AdipoR1 and AdipoR2. Cancer Sci 98:1120–1127 Man K, Ng KT, Xu A, Cheng Q, Lo CM, Xiao JW, Sun BS, Lim ZX, Cheung JS, Wu EX, Sun CK, Poon RT, Fan ST, 2010, Suppression of liver tumor growth and metastasis by adiponectin in nude mice through inhibition of tumor angiogenesis and downregulation of Rho kinase/IFN-inducible protein 10/matrix metalloproteinase 9 signaling. Clin Cancer Res 16:967–977 Barresi V, Grosso M, Giuffre G, Tuccari G, Barresi G, 2009, The expression of adiponectin receptors Adipo-R1 and Adipo-R2 is associated with an intestinal histotype and longer survival in gastric carcinoma. J Clin Pathol 62:705–709 ChiuYC, Shieh DC, TongKM, Chen CP, HuangKC, Chen PC, Fong YC, Hsu HC, Tang CH, 2009, Involvement of AdipoR receptor in adiponectin-induced motility and alpha2beta1 integrin upregulation in human chondrosarcoma cells. Carcinogenesis 30:1651–1659 Tang CH, Lu ME, 2009, Adiponectin increases motility of human prostate cancer cells via adipoR, p38, AMPK, and NF-kappaB pathways. Prostate 69:1781–1789 Pfeiler G, Treeck O, Wenzel G, Goerse R, Hartmann A, Schmitz G, Ortmann O, 2009, Influence of insulin resistance on adiponectin receptor expression in breast cancer. Maturitas 63:253–256 Howard JM, Beddy P, Ennis D, Keogan M, Pidgeon GP, Reynolds JV, 2010, Associations between leptin and adiponectin receptor upregulation, visceral obesity and tumour stage in oesophageal and junctional adenocarcinoma. Br J Surg 97:1020–1027 Wang Y, Lam JB, Lam KS, Liu J, Lam MC, Hoo RL,Wu D, Cooper GJ, Xu A, 2006, Adiponectin modulates the glycogen synthase kinase-3beta/beta-catenin signaling pathway and attenuates mammary tumorigenesis of MDA-MB-231 cells in nude mice. Cancer Res 66:11462–11470 Grossmann ME, Nkhata KJ, Mizuno NK, Ray A, Cleary MP, 2008, Effects of adiponectin on breast cancer cell growth and signaling. Br J Cancer 98:370–379 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 667 EP 675 DI 10.1007/s12253-014-9747-0 PG 9 ER PT J AU Sukosd, F Ivanyi, B Pajor, L AF Sukosd, Farkas Ivanyi, Bela Pajor, Laszlo TI Accurate Determination of the Pathological Stage with Gross Dissection Protocol for Radical Cystectomy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer stage; Gross dissection; Pathology report; Radical cystectomy; Total embedding; Urinary bladder carcinoma; Whole mount section ID Cancer stage; Gross dissection; Pathology report; Radical cystectomy; Total embedding; Urinary bladder carcinoma; Whole mount section AB The current protocol for reporting urinary bladder cancer in radical cystectomies may exhibit limitations in the diagnostic accuracy, such as a risk of understaging, especially in cases with prostatic involvement. Difficulty can arise in the verification of stage pT0, and the assessment of surgical margins is suboptimal. We have developed a daily gross dissection protocol practice where radical cystectomies are totally embedded and evaluated histologically in wholemount sections. We report here on the first 138 consecutive specimens from 2008 to the first quarter of 2012 inclusive. The incidence of the cancer stages was compared with data on 15,586 radical cystectomies from the literature. The differences were analyzed with the one-sample z-test (p<0.05). The following emerged from and our series and the literature data: pT0 8.7 % and 6.1 %; pTa 0.7 % and 2.9 %; pTis 2.9 % and 6 %; pT1 15.2 % and 15.5 %; pT2 21 % and 23.3 %; pT3 34.8 % and 34.3 %; and pT4 16.7 % and 11 %, respectively. Our findings closely reflected the means of the published statistical data based on a large number of cases. The differences were due to the more detailed processing: the case numbers in groups from pTis to pT2 were comparatively low, while those in groups pT3 and pT4 were higher. The difference in group pT4 was significant (p=0.0494).With this method, only those samples were regarded as pT0 in which the granulomatous area and the hemosiderin deposition indicative of the earlier intervention were observable and the entire preparation was tumor-free. C1 [Sukosd, Farkas] University of Szeged, Department of Pathology, 2 Allomas u, 6720 Szeged, Hungary. [Ivanyi, Bela] University of Szeged, Department of Pathology, 2 Allomas u, 6720 Szeged, Hungary. [Pajor, Laszlo] University of Szeged, Department of Urology, 57 Kalvaria u, 6725 Szeged, Hungary. RP Sukosd, F (reprint author), University of Szeged, Department of Pathology, 6720 Szeged, Hungary. EM sukosd.farkas@med.u-szeged.hu CR Parkin DM, 2008, The global burden of urinary bladder cancer. Scand J Urol Nephrol Suppl, 218)42:12–20 Cheng L, Montironi R, Davidson DD, Lopez-Beltran A, 2009, Staging and reporting of urothelial carcinoma of the urinary bladder. Mod Pathol 22(Suppl 2):S70–S95 Herr HW, 1992, Staging invasive bladder tumors. J Surg Oncol 51: 217–220 Shariat SF, Palapattu GS, Karakiewicz PI, Rogers CG, Vazina A, Bastian PJ et al, 2007, Discrepancy between clinical and pathologic stage: impact on prognosis after radical cystectomy. Eur Urol 51: 137–149, discussion 149-151 Stein JP, Lieskovsky G, Cote R, Groshen S, Feng AC, Boyd S et al, 2001, Radical cystectomy in the treatment of invasive bladder cancer: long-term results in 1,054 patients. J Clin Oncol 19:666–675 Teloh HA, 1957, Methods in surgical pathology. Thomas Chapter 26, Springfield, IL, pp 80–82 Lopez-Beltran A, Bassi PF, Pavone-Macaluso M, Montironi R, 2004, Handling and pathology reporting of specimens with carcinoma of the urinary bladder, ureter, and renal pelvis. A joint proposal of the European Society of Uropathology and the Uropathology Working Group. Virchows Arch 445:103–110 (2011, Rosai and Ackerman’s surgical pathology, 10th edn. In: Rosai J. Mosby, St. Louis, MO, pp 2913–2914 van Dijk PR, Ploeg M, Aben KK, Weijerman PC, Karthaus HF, van Berkel JT et al, 2011, Downstaging of TURBT-based muscle-invasive bladder cancer by radical cystectomy predicts better survival. ISRN Urol 2011:458930 Hautmann RE, de Petriconi RC, Pfeiffer C, Volkmer BG, 2012, Radical cystectomy for urothelial carcinoma of the bladder without neoadjuvant or adjuvant therapy: long-term results in 1100 patients. Eur Urol 61:1039–1047 Herrmann E, Stoter E, van Ophoven A, Bierer S, Bolenz C, Hertle L et al, 2008, The prognostic impact of pelvic lymph node metastasis and lymphovascular invasion on bladder cancer. Int J Urol 15:607– 611 Madersbacher S, HochreiterW, Burkhard F, Thalmann GN, Danuser H, Markwalder R et al, 2003, Radical cystectomy for bladder cancer today–a homogeneous series without neoadjuvant therapy. J Clin Oncol 21:690–696 Mallen Mateo E, Gil Martinez P, Gil Sanz MJ, Sancho Serrano C, Pascual Regueriro D, Rioja Sanz LA, 2006, Stage pT0 bladder tumors after radical cystectomy: a review of our series. Actas Urol Esp 30:763–771 MayM, Bastian PJ, Burger M, Bolenz C, Trojan L, Herrmann E et al, 2011, Multicenter evaluation of the prognostic value of pT0 stage after radical cystectomy due to urothelial carcinoma of the bladder. BJU Int 108:E278–E283 Roupret M, Drouin SJ, Larre S, Neuzillet Y, Botto H, Hitier M et al, 2011, Oncologic outcomes and survival in pT0 tumors after radical cystectomy in patients without neoadjuvant chemotherapy: results from a large multicentre collaborative study. Ann Surg Oncol 18: 3833–3838 Rink M, Ehdaie B, Cha EK, Green DA, Karakiewicz PI, Babjuk M et al, 2012, Stage-specific impact of tumor location on oncologic outcomes in patients with upper and lower tract urothelial carcinoma following radical surgery. Eur Urol 62(4):677–684 Rodriguez Faba O, Palou J, Rosales A, Breda A, Algaba F, Urdaneta G et al, 2011, Clinical predictive factors of poor outcome in patients with stage pT0 disease at radical cystectomy. J Urol 186:442–447 Takahashi A, Tsukamoto T, Tobisu K, Shinohara N, Sato K, Tomita Yet al, 2004, Radical cystectomy for invasive bladder cancer: results of multi-institutional pooled analysis. Jpn J Clin Oncol 34:14–19 Tilki D, Reich O, Svatek RS, Karakiewicz PI, KassoufW, Novara G et al, 2010, Characteristics and outcomes of patients with clinical carcinoma in situ only treated with radical cystectomy: an international study of 243 patients. J Urol 183:1757–1763 Tollefson MK, Boorjian SA, Farmer SA, Frank I, 2012, Downstaging to non-invasive urothelial carcinoma is associated with improved outcome following radical cystectomy for patients with cT2 disease. World J Urol 30(6):795–799 Vickers AJ, Cronin AM, Kattan MW, Gonen M, Scardino PT, Milowsky MI et al, 2009, Clinical benefits of a multivariate prediction model for bladder cancer: a decision analytic approach. Cancer 115:5460–5469 Yu RJ, Stein JP, Cai J, Miranda G, Groshen S, Skinner DG, 2006, Superficial, pT2a, and deep, pT2b, muscle invasion in pathological staging of bladder cancer following radical cystectomy. J Urol 176: 493–498, discussion 498-499 Greene FL, Page DL, Flemming ID et al, 2002, American joint committee on cancer staging manual. Springer, New York Soto EA, Friedell GH, Tiltman AJ, 1977, Bladder cancer as seen in giant Histologic sections. Cancer 39:447–455 JewettHJ, 1977, The historical development of the staging of bladder tumors: personal reminiscences. Urol Surv 27:37–40 Boudreaux KJ Jr, Clark PE, LowranceWT, Rumohr JA, BarocasDA, Cookson MS et al, 2009, Comparison of american joint committee on cancer pathological stage T2a versus T2b urothelial carcinoma: analysis of patient outcomes in organ confined bladder cancer. J Urol 181:540–545, discussion 546 Tilki D, Reich O, Karakiewicz PI, Novara G, Kassouf W, Ergun S et al, 2010, Validation of the AJCC TNM substaging of pT2 bladder cancer: deep muscle invasion is associated with significantly worse outcome. Eur Urol 58:112–117 Scosyrev E, Yao J, Messing E, 2010, Microscopic invasion of perivesical fat by urothelial carcinoma: implications for prognosis and pathology practice. Urology 76:908–913, discussion 914 Donat SM, Genega EM, HerrHW, Reuter VE, 2001, Mechanisms of prostatic stromal invasion in patients with bladder cancer: clinical significance. J Urol 165:1117–1120 Shen SS, Lerner SP, Muezzinoglu B, Truong LD, Amiel G, Wheeler TM, 2006, Prostatic involvement by transitional cell carcinoma in patients with bladder cancer and its prognostic significance. Hum Pathol 37:726–734 Montironi R, Cheng L, Mazzucchelli R, Scarpelli M, Kirkali Z, Montorsi F et al, 2009, Critical evaluation of the prostate from cystoprostatectomies for bladder cancer: insights from a complete sampling with the whole mount technique. Eur Urol 55:1305–1309 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 677 EP 685 DI 10.1007/s12253-014-9748-z PG 9 ER PT J AU Novak Kujundzic, R Grbesa, I Ivkic, M Kruslin, B Konjevoda, P Gall Troselj, K AF Novak Kujundzic, Renata Grbesa, Ivana Ivkic, Mirko Kruslin, Bozo Konjevoda, Pasko Gall Troselj, Koraljka TI Possible Prognostic Value of BORIS Transcript Variants Ratio in Laryngeal Squamous Cell Carcinomas – a Pilot Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Laryngeal squamous cell carcinoma; Prognostic value; BORIS; Transcript variant; Immunohistochemistry; RT-PCR ID Laryngeal squamous cell carcinoma; Prognostic value; BORIS; Transcript variant; Immunohistochemistry; RT-PCR AB BORIS is a paralog of a highly conserved, multifunctional chromatin factor CTCF. Unlike CTCF, which has been shown to possess tumor-suppressive properties, BORIS belongs to the "cancer/testis antigen" family normally expressed only in germ cells and aberrantly activated in a variety of tumors. The consequences of BORIS expression, relative abundance of its isoforms, and its role in carcinogenesis have not been completely elucidated. It activates transcription of hTERT and MYC, genes relevant for laryngeal carcinoma progression. In this study, BORIS expression has been analyzed at the transcriptional level by RT-PCR and protein level by semi-quantitative immunohistochemistry in 32 laryngeal squamous cell carcinomas and adjacent nontumorous tissue. BORIS was detected in 44 % (14/32) laryngeal squamous cell carcinoma samples, while it was detected only in one normal, tumor-adjacent tissue sample. Tree based survival analysis, using the recursive partitioning algorithm mvpart, extracted the ratio of relative abundance of BORIS transcript variants containing exon 7 (BORIS 7+) and those lacking exon 7 (BORIS 7−) as an independent prognostic factor associated with disease relapse during a 5-year followup period. Patients having BORIS 7+/BORIS 7− ratio ≥1 had a higher rate of disease relapse than patients with BORIS 7+/ BORIS 7− ratio <1. Hazard ratio for that group, based on Cox Proportional Hazard Regression, was 3.53. This is the first study analyzing expression of BORIS protein and transcript variants in laryngeal squamous cell carcinoma relative to its possible prognostic value for recurrence and overall survival. C1 [Novak Kujundzic, Renata] Ruder Boskovic Institute, Laboratory for Epigenomics, Bijenicka cesta 54, 10002 Zagreb, Croatia. [Grbesa, Ivana] Ruder Boskovic Institute, Laboratory for Epigenomics, Bijenicka cesta 54, 10002 Zagreb, Croatia. [Ivkic, Mirko] Sestre Milosrdnice University Hospital, Department of Otorhinolaryngology and Head and Neck Surgery, 10000 Zagreb, Croatia. [Kruslin, Bozo] University Hospital Sisters of Charity, Ljudevit Jurak Department of Pathology, 10000 Zagreb, Croatia. [Konjevoda, Pasko] Ruder Boskovic Institute, Center for NMR, Bijenicka cesta 54, 10002 Zagreb, Croatia. [Gall Troselj, Koraljka] Ruder Boskovic Institute, Laboratory for Epigenomics, Bijenicka cesta 54, 10002 Zagreb, Croatia. RP Novak Kujundzic, R (reprint author), Ruder Boskovic Institute, Laboratory for Epigenomics, 10002 Zagreb, Croatia. EM rnovak@irb.hr CR Loukinov DI, Pugacheva E, Vatolin S, Pack SD, Moon H, Chernukin I,Mannan P, Larsson E, Kanduri C, Vostrov AA, Cui H, Niemitz EL, Rasko JE, Docquier FM, Kistler M, Breen JJ, Zhuang Z, Quitschke WW, Renkawitz R, Klenova EM, Feinberg AP, Ohlsson R, Morse HC 3rd, Lobanenkov VV, 2002, BORIS, a novel male germ-linespecific protein associated with epigenetic reprogramming events, shares the same 11-zinc-finger domain with CTCF, the insulator protein involved in reading imprinting marks in the soma. Proc Natl Acad Sci U S A 99:6806–6811 Vatolin S, Abdullaev Z, Pack SD, Flanagan PT, Custer M, Loukinov DI, Pugacheva E, Hong JA, Morse H 3rd, Schrump DS, Risinger JI, Barrett JC, Lobanenkov VV, 2005, Conditional expression of the CTCF-paralogous transcriptional factor BORIS in normal cells results in demethylation and derepression of MAGE-A1 and reactivation of other cancer-testis genes. Cancer Res 65:7751–7762 Hong JA, Kang Y, Abdullaev Z, Flanagan PT, Pack SD, Fischete MR, Adnani MT, Loukinov DI, Vatolin S, Risinger JI, Custer M, Chen GA, Zhao M, Nguyen DM, Barrett JC, Lobanenkov VV, Schrump DS, 2005, Reciprocal binding of CTCF and BORIS to the NY-ESO-1 promoter coincides with derepression of this cancertestis gene in lung cancer cells. Cancer Res 65:7763–7774 Hoffmann MJ, Muller M, Engers R, Schulz WA, 2006, Epigenetic control of CTCFL/BORIS and OCT4 expression in urogenital malignancies. Biochem Pharmacol 72:1577–1588 Klenova EM, Morse HC, Ohlsson R, Lobanenkov VV, 2002, The novel BORIS+CTCF gene family is uniquely involved in epigenetics of normal biology and cancer. Semin Cancer Biol 12:399–414 Ohlsson R, Renkawitz R, Lobanenkov V, 2001, CTCF is a uniquely versatile transcription regulator linked to epigenetics and disease. Trends Genet 17:520–527 Klenova E, Ohlsson R, 2005, Poly(ADP-ribosyl)ation and epigenetics: is CTCF PARt of the plot? Cell Cycle 4:96–101 Jelinic P, Stehle J-C, Shaw P, 2006, The testis-specific factor CTCFL cooperates with the protein methyltransferase PRMT7 in H19 imprinting control region methylation. PLoS Biol 4:1910–1922 Fiorentino FP, Macaluso M, Miranda F, Montanari M, Russo A, Bagella L, Giordano A, 2011, CTCF and BORIS regulate Rb2/ p130 gene transcription: a novel mechanism and a new paradigm for understanding the biology of lung cancer. Mol Cancer Res 9:225– 233 Filipova GN, Fagerlie S, Klenova EM, Myers C, Dehner Y, Goodwin G, Neiman PE, Collins SJ, Lobanenkov VV, 1996, An exceptionally conserved transcriptional repressor, CTCF, employs different combinations of zinc fingers to bind diverged promoter sequences of avian and mammalian c-myc oncogenes. Mol Cell Biol 16:2802–2813 Pugacheva EM, Suzuki T, Pack SD, Kosaka-Suzuki N, Yoon J, Vostrov AA, Barsov E, Strunnikov AV, Morse HC III, Loukinov D, Lobanenkov V, 2010, The structural complexity of the human BORIS gene in gametogenesis and cancer. PLoS One 5:e13872 Renaud S, Loukinov D, Alberti L, Vostrov A, Kwon Y-W, Bosman FT, Lobanenkov V, Benhattar J, 2011, BORIS/CTCFL-mediated transcriptional regulation of the hTERT telomerase gene in testicular and ovarian tumor cells. Nucleic Acids Res 39:862–873 Ulaner GA,VuTHLT, Hu J-F, YaoX-M,YangY,GorlickR,Meyers P, Healey J, LadanyiM, Hoffman AR, 2003, Loss of imprinting of IGF2 and H19 in osteosarcoma is accompanied by reciprocal methylation changes of a CTCF-binding site. Hum Mol Genet 12:535–549 D’Arcy V, Abdullaev ZK, Pore N, Docquier F, Torrano V, Chernukin I, SmartM, Farrar D, MetodievM, Fernandez N, Richard C, Delgado DM, Lobanenkov V, Klenova E, 2006, The potential of BORIS detected in the leukocytes of breast cancer patients as an early marker of tumorigenesis. Clin Cancer Res 12:5978–5986 Looijenga LH, Hersmus R, Gillis AJ, Pfundt R, Stoop HJ, van Gurp RJ, Veltman J, Beverloo HB, van Drunen E, van Kessel AG, Pera RR, Schneider DT, Summersgill B, Shipley J, McIntyre A, van der Spek P, Schoenmakers E, Oosterhuis JW, 2006, Genomic and expression profiling of human spermatocytic seminomas: primary spermatocyte as tumorigenic precursor and DMRT1 as candidate chromosome 9 gene. Cancer Res 66:290–302 Risinger JI, Chandramouli GVR, Maxwell GL, Custer M, Pack S, Loukinov D, Aprelikova O, Litzi T, Schrump DS, Murphy SK, Berchuck A, Lobanenkov V, Barrett JC, 2007, Global expression analysis of cancer/testis genes in uterine cancers reveals a high incidence of BORIS expression. Clin Cancer Res 13:1713–1719 Woloszynska-Read A, James SR, Link PA, Yu J, Odunsi K, Karpf AR, 2007, DNA methylation-dependent regulation of BORIS/ CTCFL expression in ovarian cancer. Cancer Immun 7:21 D’Arcy V, Pore N, Docquier F, Abdullaev ZK, Chernukhin I, Kita GX, Rai S, Smart M, Farrar D, Pack S, Lobanenkov V, Klenova E, 2008, BORIS, a paralogue of the transcription factor, CTCF, is aberrantly expressed in breast tumors. Br J Cancer 98:571–579 Kholmanskikh O, Loriot A, Brasseur F, De Plaen E, De Smet C, 2008, Expression of BORIS in melanoma: lack of association with MAGE-A1 activation. Int J Cancer 122:777–784 Smith IM, Glazer CA, Mithani SK, Ochs MF, Sun W, Bhan S, Vostrov A, Abdullaev Z, Lobanenkov V, Gray A, Liu C, Chang SS, Ostrow KL, Westra WH, Begum S, Dhara M, Califano J, 2009, Coordinated activation of candidate proto-oncogenes and cancer testes antigens via promoter demethylation in head and neck cancer and lung cancer. PLoS One 4:e4961 Smiraglia DJ, Smith LT, Lang JC, Rush LJ, Dai Z, Schuller DE, Plass C, 2003, Differential targets of CpG island hypermethylation in primary and metastatic head and neck squamous cell carcinoma, HNSCC). J Med Genet 40:25–33 Schmezer P, Plass C, 2008, Epigenetic aspects in carcinomas of the head and neck. HNO 56:594–602 Larynx D et al, 2010, In: Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, eds, AJCC cancer staging manual, 7th edn. Springer, New York, pp 57–62 Grbesa I, Ivkic M, Pegan B, Gall-Troselj K, 2006, Loss of imprinting and promoter usage of the IGF2 in laryngeal squamous cell carcinoma. Cancer Lett 238:224–229 rpart by Terry M Therneau, Beth Atkinson. R port of rpart by Brian Ripley Rripley@stats.ox.ac.uk>. Some routines from vegan – Jari Oksanen < jari.oksanen@oulu.fi > Extensions and adaptations of rpart to mvpart by Glenn De’ath., 2013, mvpart:Multivariate partitioning. R package version 1.6-1. http://CRAN.R-project.org/package=mvpart. Accessed 15 Jan 2014 R Core Team, 2013, R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. URL http://www.R-project.org/ Accessed 15 Jan 2014 Schumacher M, Hollander N, Schwarzer G, Willi Sauerbrei W, 2006, Prognostic factor studies. In: Crowley J, Ankerst DP, eds, Handbook of statistics in clinical oncology, 2nd edn. Chapman & Hall/CRC, Boca Raton, pp 289–333 Liu X, 2013, Survival analysis. Wiley, Chichester Ragin CC,Modugno F, Gollin SM(2007, The epidemiology and risk factors of head and neck cancer: a focus on human papillomavirus. J Dent Res 86:104–114 Poschl G, Stickel F, Wang XD, Seitz HK, 2004, Alcohol and cancer: genetic and nutritional aspects. Proc Nutr Soc 63:65– 71 Flatley JE, McNeir K, Balasubramani L, Tidy J, Stuart EL, Young TA, Powers HJ, 2009, Folate status and aberrant DNA methylation are associated with HPV infection and cervical pathogenesis. Cancer Epidemiol Biomarkers Prev 18:2782– 2789 McCarty KS Jr, Szabo E, Flowers JL, Cox EB, Leight GS, Miller L, Konrath J, Soper JT, Budwit DA, Creasman WT, Seigler HF, McCarty KS Sr, 1986, Use of a monoclonal anti-estrogen receptor antibody in the immunohistochemical evaluation of human tumors. Cancer Res 46:4244s–4248s Rosa-Garrido M, Ceballos L, Alonso-Lecue P, Abraira C, Delgado MD, Gandarillas A, 2012, A cell cycle role for the epigenetic factor CTCF-L/BORIS. PLoS One 7:e39371 Grbesa I, Marinkovic M, Ivkic M, Kruslin B, Novak-Kujundzic R, Pegan B, Bogdanovic O, Bedekovic V, Gall-Troselj K, 2008, Loss of imprinting of IGF2 and H19, loss of heterozygosity of IGF2R and CTCF, and helicobacter pylori infection in laryngeal squamous cell carcinoma. J Mol Med, Berl, 86:1057–1066 Hogarth C, Itman C, Jans DA, Loveland KL, 2005, Regulated nucleocytoplasmic transport in spermatogenesis: a driver of cellular differentiation? BioEssays 27:1011–1025 Nguyen P, Bar-Sela G, Sun L, Bisht KS, Cui H, Kohn E, Feinberg AP, Gius D, 2008, BAT3 and SET1A form a complex with CTCFL/ BORIS to modulate H3K4 histone dimethylation and gene expression. Mol Cell Biol 28:6720–6729 Ozaki T, Hanaoka E, Naka M, Nakagawara A, Sakiyama S, 1999, Cloning and characterization of rat BAT3 cDNA. DNA Cell Biol 18: 503–512 Manchen ST, Hubberstey AV, 2001, Human Scythe contains a functional nuclear localization sequence and remains in the nucleus during staurosporine-induced apoptosis. Biochem Biophys Res Commun 287:1075–1082 Tsukahara T, Kimura S, Ichimiya S, Torigoe T, Kawaguchi S, Wada T, Yamashita T, Sato N, 2009, Scythe/BAT3 regulates apoptotic cell death induced by papillomavirus binding factor in human osteosarcoma. Cancer Sci 100:47–53 EhrlichM(2002, DNA methylation in cancer: too much, but also too little. Oncogene 21:5400–5413 Renaud S, Pugacheva EM, DelgadoMD, Braunschweig R, Abdullaev Z, Loukinov D, Benhattar J, Lobanenkov V, 2007, Expression of the CTCF-paralogous cancer-testis gene, brother of the regulator of imprinted sites, BORIS), is regulated by three alternative promoters modulated by CpG methylation and by CTCF and p53 transcription factors. Nucleic Acids Res 35:7372–7388 HinesWC, Bazarov AV,Mukhopadhyay R,Yaswen P, 2010, BORIS, CTCFL, is not expressed in most human breast cell lines and high grade breast carcinomas. PLoS One 5:e9738 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 687 EP 695 DI 10.1007/s12253-014-9749-y PG 9 ER PT J AU Kwon, YS Lee, JH Kim, B Park, JW Kwon, KT Kang, HS Kim, Sh AF Kwon, Young Sun Lee, Jae-Ho Kim, Bora Park, Jong-Wook Kwon, Kyu Taeg Kang, Hee Sun Kim, Shin TI Complexity in Regulation of microRNA Machinery Components in Invasive Breast Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microRNA biogenesis; DGCR8; AGO2; Ki-67; Invasive breast carcinoma ID microRNA biogenesis; DGCR8; AGO2; Ki-67; Invasive breast carcinoma AB Altered expression of microRNA (miRNA) machinery components may play an important role in breast cancer progression. The objective of the current study was to evaluate Drosha, the DiGeorge syndrome critical region gene 8 (DGCR8), Dicer, and Argonaute 2 (AGO2) mRNA expression in invasive breast carcinoma (IBC) and to assess the value of clinical parameters on their expression. By using quantitative real-time PCR, we examined the expression of the four miRNA machinery components in 52 breast tumor tissues which are diagnosed as invasive ductal carcinoma and adjacent non-neoplastic tissues. In the present study, decreased mRNA expression levels of major miRNA machinery components were observed in IBC. The altered mRNA expression levels of DGCR8 and AGO2 are positively correlated with to each other. This study revealed for the first time that expression alterations of DGCR8 are significantly associated with estrogen receptor and Ki-67 status in IBC. Moreover, AGO2 mRNA expression level was significantly correlated with N stage. These results provided evidences that downregulated the four miRNA machinery components may play an important role in breast pathobiology and that DGCR8 and AGO2 might be associated with important clinical factors. C1 [Kwon, Young Sun] Keimyung University, School of Medicine, Department of Pathology, 1095 Dalgubeoldaero, Dalseo-Gu, 704-701 Daegu, South Korea. [Lee, Jae-Ho] Keimyung University, School of Medicine, Department of Anatomy, 1095 Dalgubeoldaero, Dalseo-Gu, 704-701 Daegu, South Korea. [Kim, Bora] Keimyung University, School of Medicine, Department of Immunology, 1095 Dalgubeoldaero, Dalseo-Gu, 704-701 Daegu, South Korea. [Park, Jong-Wook] Keimyung University, School of Medicine, Department of Immunology, 1095 Dalgubeoldaero, Dalseo-Gu, 704-701 Daegu, South Korea. [Kwon, Kyu Taeg] Keimyung University, School of Medicine, Department of Immunology, 1095 Dalgubeoldaero, Dalseo-Gu, 704-701 Daegu, South Korea. [Kang, Hee Sun] Keimyung University, School of Medicine, Department of General Surgery, 1095 Dalgubeoldaero, Dalseo-Gu, 704-701 Daegu, South Korea. [Kim, Shin] Keimyung University, School of Medicine, Department of Immunology, 1095 Dalgubeoldaero, Dalseo-Gu, 704-701 Daegu, South Korea. RP Kim, Sh (reprint author), Keimyung University, School of Medicine, Department of Immunology, 704-701 Daegu, South Korea. EM god98005@dsmc.or.kr CR Abd El-Rehim DM, Ball G, Pinder SE, Rakha E, Paish C, Robertson JF, Macmillan D, Blamey RW, Ellis IO, 2005, High-throughput protein expression analysis using tissue microarray technology of a large well-characterised series identifies biologically distinct classes of breast cancer confirming recent cDNA expression analyses. Int J Cancer 116(3):340–350., DOI 10.1002/ijc.21004 Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C, Cowan D, Dressler L, Akslen LA, Ragaz J, Gown AM, Gilks CB, van de Rijn M, Perou CM, 2004, Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res 10(16):5367–5374., DOI 10.1158/1078-0432.CCR-04-0220 Perou CM, Sorlie T, EisenMB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen- Dale AL, Brown PO, BotsteinD(2000)Molecular portraits of human breast tumours. Nature 406(6797):747–752., DOI 10.1038/35021093 Rakha EA, Reis-Filho JS, Ellis IO, 2008, Basal-like breast cancer: a critical review. J Clin Oncol 26(15):2568–2581., DOI 10.1200/JCO. 2007.13.1748 Diaz LK, Cryns VL, SymmansWF, Sneige N, 2007, Triple negative breast carcinoma and the basal phenotype: from expression profiling to clinical practice. Adv Anat Pathol 14(6):419–430., DOI 10.1097/ PAP.0b013e3181594733 Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, Thorsen T, Quist H, Matese JC, Brown PO, Botstein D, Lonning PE, Borresen-Dale AL, 2001, Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98(19):10869–10874., DOI 10. 1073/pnas.191367098 Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA, 2010, Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol 11(2):174–183., DOI 10.1016/S1470- 2045(09)70262-1 Dowsett M, Nielsen TO, A'Hern R, Bartlett J, Coombes RC, Cuzick J, Ellis M, Henry NL, Hugh JC, Lively T, McShane L, Paik S, Penault-Llorca F, Prudkin L, Regan M, Salter J, Sotiriou C, Smith IE, Viale G, Zujewski JA, Hayes DF, International Ki-67 in Breast Cancer Working G, 2011, Assessment of Ki67 in breast cancer: recommendations from the International Ki67 in Breast Cancer working group. J Natl Cancer Inst 103(22):1656–1664., DOI 10. 1093/jnci/djr393 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116(2):281–297 Papachristou DJ, Korpetinou A, Giannopoulou E, Antonacopoulou AG, Papadaki H, Grivas P, Scopa CD, Kalofonos HP, 2011, Expression of the ribonucleases Drosha, Dicer, and Ago2 in colorectal carcinomas. Virchows Arch Int J Pathol 459(4):431–440., DOI 10. 1007/s00428-011-1119-5 Mockenhaupt S, Schurmann N, Grimm D, 2011, When cellular networks run out of control: global dysregulation of the RNAi machinery in human pathology and therapy. Prog Mol Biol Transl Sci 102:165–242., DOI 10.1016/B978-0-12-415795-8.00006-4 Lee Y, Han J, Yeom KH, Jin H, Kim VN, 2006, Drosha in primary microRNA processing. Cold Spring Harb Symp Quant Biol 71:51– 57., DOI 10.1101/sqb.2006.71.041 Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, Cooch N, Shiekhattar R, 2004, The Microprocessor complex mediates the genesis of microRNAs. Nature 432(7014):235–240., DOI 10. 1038/nature03120 TijstermanM, Plasterk RH, 2004, Dicers at RISC; the mechanism of RNAi. Cell 117(1):1–3 Sand M, Skrygan M, Georgas D, Arenz C, Gambichler T, Sand D, Altmeyer P, Bechara FG, 2012, Expression levels of the microRNA maturing microprocessor complex component DGCR8 and the RNA-induced silencing complex, RISC, components argonaute-1, argonaute-2, PACT, TARBP1, and TARBP2 in epithelial skin cancer. Mol Carcinog 51(11):916–922., DOI 10.1002/mc.20861 Merritt WM, Lin YG, Han LY, Kamat AA, Spannuth WA, Schmandt R, Urbauer D, Pennacchio LA, Cheng JF, Nick AM, Deavers MT, Mourad-Zeidan A, Wang H, Mueller P, Lenburg ME, Gray JW, Mok S, Birrer MJ, Lopez-Berestein G, Coleman RL, Bar-Eli M, Sood AK, 2008, Dicer, Drosha, and outcomes in patients with ovarian cancer. N Engl J Med 359(25):2641–2650., DOI 10.1056/NEJMoa0803785 Kim B, Lee JH, Park JW, Kwon TK, Baek SK, Hwang I, Kim S, 2013, An essential microRNA maturing microprocessor complex component DGCR8 is up-regulated in colorectal carcinomas. Clin Exp Med., DOI 10.1007/s10238-013-0243-8 Grelier G, Voirin N, Ay AS, Cox DG, Chabaud S, Treilleux I, Leon-Goddard S, Rimokh R, Mikaelian I, Venoux C, Puisieux A, Lasset C, Moyret-Lalle C, 2009, Prognostic value of Dicer expression in human breast cancers and association with the mesenchymal phenotype. Br J Cancer 101(4):673–683., DOI 10. 1038/sj.bjc.6605193 Dedes KJ, Natrajan R, Lambros MB, Geyer FC, Lopez-Garcia MA, Savage K, Jones RL, Reis-Filho JS, 2011, Down-regulation of the miRNA master regulators Drosha and Dicer is associated with specific subgroups of breast cancer. Eur J Cancer 47(1):138–150., DOI 10. 1016/j.ejca.2010.08.007 Passon N, Gerometta A, Puppin C, Lavarone E, Puglisi F, Tell G, Di Loreto C, Damante G, 2012, Expression of Dicer and Drosha in triple-negative breast cancer. J Clin Pathol 65(4):320–326., DOI 10. 1136/jclinpath-2011-200496 YanM, Huang HY,Wang T,Wan Y, Cui SD, Liu ZZ, Fan QX, 2012, Dysregulated expression of dicer and drosha in breast cancer. Pathol Oncol Res 18(2):343–348., DOI 10.1007/s12253-011-9450-3 Khoshnaw SM, Rakha EA, Abdel-Fatah TM, Nolan CC, Hodi Z, Macmillan DR, Ellis IO, Green AR, 2012, Loss of Dicer expression is associated with breast cancer progression and recurrence. Breast Cancer Res Treat 135(2):403–413., DOI 10. 1007/s10549-012-2169-3 Chiosea S, Jelezcova E, Chandran U, Acquafondata M, McHale T, Sobol RW, Dhir R, 2006, Up-regulation of dicer, a component of the MicroRNA machinery, in prostate adenocarcinoma. Am J Pathol 169(5):1812–1820., DOI 10.2353/ ajpath.2006.060480 Sugito N, Ishiguro H, Kuwabara Y, Kimura M, Mitsui A, Kurehara H, Ando T, Mori R, Takashima N, Ogawa R, Fujii Y, 2006, RNASEN regulates cell proliferation and affects survival in esophageal cancer patients. Clin Cancer Res 12(24):7322–7328., DOI 10. 1158/1078-0432.CCR-06-0515 Martin MG, Payton JE, Link DC, 2009, Dicer and outcomes in patients with acute myeloid leukemia, AML). Leuk Res 33(8): e127., DOI 10.1016/j.leukres.2009.02.003 Harvey JM, Clark GM, Osborne CK, Allred DC, 1999, Estrogen receptor status by immunohistochemistry is superior to the ligandbinding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol 17(5):1474–1481 Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JM, Bilous M, Fitzgibbons P, Hanna W, Jenkins RB, Mangu PB, Paik S, Perez EA, Press MF, Spears PA, Vance GH, Viale G, Hayes DF, 2013, Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: american society of clinical oncology/college of american pathologists clinical practice guideline update. J Clin Oncol 31(31):3997–4013., DOI 10.1200/JCO.2013.50.9984 Schmittgen TD, Livak KJ, 2008, Analyzing real-time PCR data by the comparative C(T, method. Nat Protoc 3(6):1101–1108 Shen J, Stass SA, Jiang F, 2012, MicroRNAs as potential biomarkers in human solid tumors. Cancer Lett., DOI 10.1016/j.canlet.2012.11.001 Li PY, He FC, Zhou GQ, 2011, Association of human microRNA related genetic variations with cancer. Yi chuan = Hereditas/ Zhongguo yi chuan xue hui bian ji 33(8):870–878 Schmittgen TD, 2008, Regulation of microRNA processing in development, differentiation and cancer. J CellMol Med 12(5B):1811– 1819., DOI 10.1111/j.1582-4934.2008.00483.x Han L, Zhang A, Zhou X, Xu P, Wang GX, Pu PY, Kang CS, 2010, Downregulation of Dicer enhances tumor cell proliferation and invasion. Int J Oncol 37(2):299–305 Faggad A, Budczies J, Tchernitsa O, Darb-Esfahani S, Sehouli J, Muller BM, Wirtz R, Chekerov R, Weichert W, Sinn B, Mucha C, Elwali NE, Schafer R, Dietel M, Denkert C, 2010, Prognostic significance of Dicer expression in ovarian cancer-link to global microRNA changes and oestrogen receptor expression. J Pathol 220(3):382–391., DOI 10.1002/path.2658 Cheng C, Fu X, Alves P, Gerstein M, 2009, mRNA expression profiles show differential regulatory effects of microRNAs between estrogen receptor-positive and estrogen receptor-negative breast cancer. Genome Biol 10(9):R90., DOI 10.1186/gb-2009-10-9-r90 Erson AE, Petty EM, 2009, miRNAs and cancer: new research developments and potential clinical applications. Cancer Biol Ther 8(24):2317–2322 Yu Z, Baserga R, Chen L, Wang C, Lisanti MP, Pestell RG, 2010, microRNA, cell cycle, and human breast cancer. Am J Pathol 176(3): 1058–1064., DOI 10.2353/ajpath.2010.090664 Caudle AS, Yi M, Hoffman KE, Mittendorf EA, Babiera GV, Hwang RF, Meric-Bernstam F, Sahin AA, Hunt KK, 2013, Impact of identification of internal mammary sentinel lymph node metastasis in breast cancer patients. Ann Surg Oncol., DOI 10.1245/s10434-013-3276-z Liu Y, Yin W, Yan T, Du Y, Shao Z, Lu J, 2013, The clinical significance of Ki-67 as a marker of prognostic value and chemosensitivity prediction in hormone-receptor-positive breast cancer: a meta-analysis of the published literature. Curr Med Res Opin., DOI 10.1185/03007995.2013.833088 Pathmanathan N, Balleine RL, 2013, Ki67 and proliferation in breast cancer. J Clin Pathol 66(6):512–516., DOI 10.1136/jclinpath-2012-201085 Foulkes WD, Smith IE, Reis-Filho JS, 2010, Triple-negative breast cancer. N Engl J Med 363(20):1938–1948., DOI 10.1056/ NEJMra1001389 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 697 EP 705 DI 10.1007/s12253-014-9750-5 PG 9 ER PT J AU Kropotova, SE Zinovieva, LO Zyryanova, FA Dybovaya, IV Prasolov, SV Beresten, FS Oparina, YN Mashkova, DT AF Kropotova, S Ekaterina Zinovieva, L Olga Zyryanova, F Alisa Dybovaya, I Vera Prasolov, S Vladimir Beresten, F Sergey Oparina, Yu Nina Mashkova, D Tamara TI Altered Expression of Multiple Genes Involved in Retinoic Acid Biosynthesis in Human Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Retinoic acid biosynthesis; Gene expression; Tumor progression ID Colorectal cancer; Retinoic acid biosynthesis; Gene expression; Tumor progression AB All-trans-retinoic acid (atRA), the oxidized form of vitamin A (retinol), regulates a wide variety of biological processes, such as cell proliferation and differentiation. Multiple alcohol, retinol and retinaldehyde dehydrogenases (ADHs, RDHs, RALDHs) as well as aldo-keto reductases (AKRs) catalyze atRA production. The reduced atRA biosynthesis has been observed in several human tumors, including colorectal cancer. However, subsets of atRA-synthesizing enzymes have not been determined in colorectal tumors. We investigated the expression patterns of genes involved in atRA biosynthesis in normal human colorectal tissues, primary carcinomas and cancer cell lines by RT-PCR. These genes were identified using transcriptomic data analysis (expressed sequence tags, RNA-sequencing, microarrays). Our results indicate that each step of the atRA biosynthesis pathway is dysregulated in colorectal cancer. Frequent and significant decreases in the mRNA levels of the ADH1B, ADH1C, RDHL, RDH5 and AKR1B10 genes were observed in a majority of colorectal carcinomas. The expression levels of the RALDH1 gene were reduced, and the expression levels of the cytochrome CYP26A1 gene increased. The human colon cancer cell lines showed a similar pattern of changes in the mRNA levels of these genes. A dramatic reduction in the expression of genes encoding the predominant retinoloxidizing enzymes could impair atRA production. The most abundant of these genes, ADH1B and ADH1C, display decreased expression during progression from adenoma to early and more advanced stage of colorectal carcinomas. The diminished atRA biosynthesis may lead to alteration of cell growth and differentiation in the colon and rectum, thus contributing to the progression of colorectal cancer. C1 [Kropotova, S Ekaterina] Russian Academy of Sciences, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russian Federation. [Zinovieva, L Olga] Russian Academy of Sciences, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russian Federation. [Zyryanova, F Alisa] Russian Academy of Sciences, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russian Federation. [Dybovaya, I Vera] Russian Academy of Sciences, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russian Federation. [Prasolov, S Vladimir] Russian Academy of Sciences, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russian Federation. [Beresten, F Sergey] Russian Academy of Sciences, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russian Federation. [Oparina, Yu Nina] Russian Academy of Sciences, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russian Federation. [Mashkova, D Tamara] Russian Academy of Sciences, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russian Federation. RP Mashkova, DT (reprint author), Russian Academy of Sciences, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russian Federation. EM tamashkova@yandex.ru CR Di Caro V, Phillips B, Engman C, Harnaha J, Trucco M, Giannoukakis N, 2013, Retinoic acid-producing, ex-vivo-generated human tolerogenic dendritic cells induce the proliferation of immunosuppressive B lymphocytes. Clin Exp Immunol 174(2):302–317 Morikawa K, Hanada H, Hirota K, Nonaka M, Ikeda C, 2013, Alltrans retinoic acid displaysmultiple effects on the growth, lipogenesis and adipokine gene expression of AML-I preadipocyte cell line. Cell Biol Int 37(1):36–46 Blomhoff R, Blomhoff HK, 2006, Overview of retinoid metabolism and function. J Neurobiol 66(7):606–630 Harrison EH, 2012, Mechanisms involved in the intestinal absorption of dietary vitamin A and provitamin A carotenoids. Biochim Biophys Acta 1821(1):70–77 Napoli JL, 2012, Physiological insights into all-trans-retinoic acid biosynthesis. Biochim Biophys Acta 1821(1):152–167 Kumar S, Sandell LL, Trainor PA, Koentgen F, Duester G, 2012, Alcohol and aldehyde dehydrogenases: retinoid metabolic effects in mouse knockout models. Biochim Biophys Acta 1821(1):198–205 Pares X, Farres J, Kedishvili N, Duester G, 2008, Medium- and short-chain dehydrogenase/reductase gene and protein families: medium-chain and short-chain dehydrogenases/reductases in retinoid metabolism. Cell Mol Life Sci 65(24):3936–3949 Duester G, Farres J, Felder MR et al, 1999, Recommended nomenclature for the vertebrate alcohol dehydrogenase gene family. Biochem Pharmacol 58(3):389–395 Estonius M, Svensson S, Hoog J-O, 1996, Alcohol dehydrogenase in human tissues: localization of transcripts coding for five classes of the enzyme. FEBS Lett 397(2–3):338–342 Gallego O, Belyaeva OV, Porte S et al, 2006, Comparative functional analysis of human medium-chain dehydrogenases, short-chain dehydrogenases/reductases and aldo-keto reductases with retinoids. Biochem J 399(1):101–109 Molotkov A, Deltour L, Foglio MH, Cuenca AE, Duester G, 2002, Distinct retinoid metabolic functions for alcohol dehydrogenase genes Adh1 and Adh4 in protection against vitamin A toxicity or deficiency revealed in double null mutant mice. J Biol Chem 277(16): 13804–13811 Molotkov A, Fan X, Deltour L et al, 2002, Stimulation of retinoic acid production and growth by ubiquitously expressed alcohol dehydrogenase Adh3. Proc Natl Acad Sci U S A 99(8):5337–5342 Belyaeva OV, Johnson MP, Kedishvili NY, 2008, Kinetic analysis of human enzyme RDH10 defines the characteristics of a physiologically relevant retinol dehydrogenase. J Biol Chem 283(29):20299– 20308 Crosas B, Hyndman DJ, Gallego O, Martras S, Pares X, Flynn TG, Farres J, 2003, Human aldose reductase and human small intestine aldose reductase are efficient retinal reductases: Consequences for retinoid metabolism. Biochem J 373(Pt 3):973–979 Ross AC, Zolfaghari R, 2011, Cytochrome P450s in the regulation of cellular retinoic acid metabolism. Annu Rev Nutr 31:65–87 Bushue N,Wan YJ, 2010, Retinoid pathway and cancer therapeutics. Adv Drug Deliv Rev 62(13):1285–1298 Nadauld LD, Shelton DN, Chidester S, Yost HJ, Jones DA, 2005, The zebrafish retinol dehydrogenase, rdh1l, is essential for intestinal development and is regulated by the tumor suppressor adenomatous polyposis coli. J Biol Chem 280(34): 30490–30495 NilesRM,Wilhelm SA, Thomas P, Zamcheck N, 1988, The effect of sodium butyrate and retinoic acid on growth and CEA production in a series of human colorectal tumor cell lines representing different states of differentiation. Cancer Invest 6(1):39–45 Reynolds S, Rajagopal S, Chakrabarty S, 1998, Differentiationinducing effect of retinoic acid, difluoromethylornithine, sodium butyrate and sodium suramin in human colon cancer cells. Cancer Lett 134(1):53–60 Ang HL, Deltour L, Zgombic-Knight M, Wagner MA, Duester G, 1996, Expression patterns of class I and class IV alcohol dehydrogenase genes in developing epithelia suggest a role for alcohol dehydrogenase in local retinoic acid synthesis. Alcohol Clin Exp Res 20(6):1050–1064 Parlesak A, Menzl I, Feuchter A, Bode JC, Bode C, 2000, Inhibition of retinol oxidation by ethanol in the rat liver and colon. Gut 47(6): 825–831 Jette C, Peterson PW, Sandoval IT, Manos EJ, Hadley E, IrelandCM, Jones DA, 2004, The tumor suppressor adenomatous polyposis coli and caudal related homeodomain protein regulate expression of retinol dehydrogenase L. J Biol Chem 279(33):34397–34405 Pasquali D, Thaller C, Eichele G, 1996, Abnormal level of retinoic acid in prostate cancer tissues. J Clin Endocrinol Metab 81(6):2186– 2191 Mira-y-Lopez R, ZhengWL, Kuppumbatti YS, Rexer B, Jing Y, Ong DE, 2000, Retinol conversion to retinoic acid is impaired in breast cancer cell lines relative to normal cells. J Cell Physiol 185(2):302– 309 Williams SJ, Cvetkovic D, Hamilton TC, 2009, Vitamin A metabolism is impaired in human ovarian cancer. Gynecol Oncol 112(3): 637–645 MatsumotoM, Yokoyama H, Suzuki H, Shiraishi-Yokoyama H, Hibi T, 2005, Retinoic acid formation from retinol in the human gastric mucosa: role of class IV alcohol dehydrogenase and its relevance to morphological changes. Am J Physiol Gastrointest Liver Physiol 289(3):G429–G433 Fukumoto S, Yamauchi N, Moriguchi H et al, 2005, Overexpression of the aldo-keto reductase family protein AKR1B10 is highly correlated with smokers’ non-small cell lung carcinomas. Clin Cancer Res 11(5):1776–1785 Chiang CP, Jao SW, Lee SP et al, 2012, Expression pattern, ethanolmetabolizing activities, and cellular localization of alcohol and aldehyde dehydrogenases in human large bowel: association of the functional polymorphisms of ADH and ALDH genes with hemorrhoids and colorectal cancer. Alcohol 46(1):37–49 Zaitseva M, Vollenhoven BJ, Rogers PA, 2007, Retinoic acid pathway genes show significantly altered expression in uterine fibroids when compared with normal myometrium. Mol Hum Reprod 13(8): 577–585 Ashla AA, Hoshikawa Y, Tsuchiya H et al, 2010, Genetic analysis of expression profile involved in retinoid metabolism in non-alcoholic fatty liver disease. Hepatol Res 40(6):594–604 O’Shaughnessy PJ, AbelM, Charlton HM, Hu B, Johnston H, Baker PJ, 2007, Altered expression of genes involved in regulation of vitamin A metabolism, solute transportation, and cytoskeletal function in the androgen-insensitive tfm mouse testis. Endocrinology 148:2914–2924 Sandell LL, Lynn ML, Inman KE, McDowell W, Trainor PA, 2012, RDH10 oxidation of Vitamin A is a critical control step in synthesis of retinoic acid during mouse embryogenesis. PLoSOne 7(2):e30698 Collins MD, Eckhoff C, Chahoud I, Bochert G, Nau H, 1992, 4- Methylpyrazole partially ameliorated the teratogenicity of retinol and reduced the metabolic formation of all-trans-retinoic acid in the mouse. Arch Toxicol 66(9):652–659 Boleda MD, Julia P, Moreno A, Pares X, 1989, Role of extrahepatic alcohol dehydrogenase in rat ethanol metabolism. Arch Biochem Biophys 274(1):74–81 Chiang CP, Jao SW, Lee SP, Chen PC, Chung CC, Lee SL, Nieh S, Yin SJ, 2012, Expression pattern, ethanol-metabolizing activities, and cellular localization of alcohol and aldehyde dehydrogenases in human large bowel: association of the functional polymorphisms of ADH and ALDH genes with hemorrhoids and colorectal cancer. Alcohol 46(1):37–49 Chase JR, Poolman MG, Fell DA, 2009, Contribution of NADH increases to ethanol’s inhibition of retinol oxidation by human ADH isoforms. Alcohol Clin Exp Res 33(4):571–580 Mashkova TD, Oparina NIu, Zinov’eva OL et al, 2006, Transcription of TIMP3, DAPK1, and AKR1B10 in squamous cell lung cancer. Mol Biol, Mosk, 40(6):1047–1054 Scuric Z, Stain SC, Anderson WF, Hwang JJ, 1998, New member of aldose reductase family proteins overexpressed in human hepatocellular carcinoma. Hepatology 27(4):943–950 Kropotova ES, Zinov’eva OL, Zyrianova AF et al, 2013, Expression of genes involved in retinoic acid biosynthesis in human gastric cancer. Mol Biol, Mosk, 47(2):317–330 Kropotova ES, Tychko RA, Zinov’eva OL et al, 2010, Downregulation of AKR1B10 gene expression in colorectal cancer. Mol Biol, Mosk, 44(2):243–250 Roberts ES, Vaz AD, Coon MJ, 1992, Role of isozymes of rabbit microsomal cytochrome P-450 in the metabolism of retinoic acid, retinol, and retinal. Mol Pharmacol 41(2):427–433 Martin HJ, Maser E, 2009, Role of human aldo-ketoreductase AKR1B10 in the protection against toxic aldehydes. Chem Biol Interact 178(1–3):145–150 Westerlund M, Belin AC, FelderMR, Olson L, Galter D, 2007, High and complementary expression patterns of alcohol and aldehyde dehydrogenases in the gastrointestinal tract: implications for Parkinson’s disease. FEBS J 274(5):1212–1223 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 707 EP 717 DI 10.1007/s12253-014-9751-4 PG 11 ER PT J AU Oberlander, M Alkemade, H Bunger, S Ernst, F Thorns, Ch Braunschweig, T Habermann, J AF Oberlander, Martina Alkemade, Hendrik Bunger, Stefanie Ernst, Floris Thorns, Christoph Braunschweig, Till Habermann, K. Jens TI A ‘Waterfall’ Transfer-based Workflow for Improved Quality of Tissue Microarray Construction and Processing in Breast Cancer Research SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tissue microarray (TMA); Biomarkers; High-throughput technique; Immunohistochemistry; Immunofluorescence staining ID Tissue microarray (TMA); Biomarkers; High-throughput technique; Immunohistochemistry; Immunofluorescence staining AB A major focus in cancer research is the identification of biomarkers for early diagnosis, therapy prediction and prognosis. Hereby, validation of target proteins on clinical samples is of high importance. Tissue microarrays (TMAs) represent an essential advancement for high-throughput analysis by assembling large numbers of tissue cores with high efficacy and comparability. However, limitations along TMA construction and processing exist. In our presented study, we had to overcome several obstacles in the construction and processing of high-density breast cancer TMAs to ensure good quality sections for further research. Exemplarily, 406 breast tissue cores from formalin-fixed and paraffin embedded samples of 245 patients were placed onto three recipient paraffin blocks. Sectioning was performed using a rotary microtome with a "waterfall" automated transfer system. Sections were stained by immunohistochemistry and immunofluorescence for nine proteins. The number and quality of cores after sectioning and staining was counted manually for each marker. In total, 97.1 % of all cores were available after sectioning, while further 96 % of the remaining cores were evaluable after staining. Thereby, normal tissue cores were more often lost compared to tumor tissue cores. Our workflow provides a robust method for manufacturing high-density breast cancer TMAs for subsequent IHC or IF staining without significant sample loss. C1 [Oberlander, Martina] University of Lubeck and University Medical Center Schleswig-Holstein, Department of Surgery, Section for Translational Surgical Oncology and Biobanking, Campus Lubeck, Ratzeburger Allee 160, 23538 Lubeck, Germany. [Alkemade, Hendrik] University of Lubeck and University Medical Center Schleswig-Holstein, Department of Surgery, Section for Translational Surgical Oncology and Biobanking, Campus Lubeck, Ratzeburger Allee 160, 23538 Lubeck, Germany. [Bunger, Stefanie] University of Lubeck and University Medical Center Schleswig-Holstein, Department of Surgery, Section for Translational Surgical Oncology and Biobanking, Campus Lubeck, Ratzeburger Allee 160, 23538 Lubeck, Germany. [Ernst, Floris] University of Lubeck and University Medical Center Schleswig-Holstein, Department of Surgery, Section for Translational Surgical Oncology and Biobanking, Campus Lubeck, Ratzeburger Allee 160, 23538 Lubeck, Germany. [Thorns, Christoph] University of Schleswig-Holstein, Institute of Pathology, Campus LubeckLubeck, Germany. [Braunschweig, Till] Medical Faculty of the Technical University of Aachen, Department of Pathology, Pauwelsstraße 30, 52074 Aachen, Germany. [Habermann, K. Jens] University of Lubeck and University Medical Center Schleswig-Holstein, Department of Surgery, Section for Translational Surgical Oncology and Biobanking, Campus Lubeck, Ratzeburger Allee 160, 23538 Lubeck, Germany. RP Habermann, J (reprint author), University of Lubeck and University Medical Center Schleswig-Holstein, Department of Surgery, Section for Translational Surgical Oncology and Biobanking, 23538 Lubeck, Germany. EM habermann@biobank.uni-luebeck.de CR Martini M, Vecchione L, Siena S, Tejpar S, Bardelli A, 2012, Targeted therapies: how personal should we go? Nat Rev Clin Oncol 9(2):87–97., DOI 10.1038/nrclinonc.2011.164 Braunschweig T, Chung JY, Hewitt SM, 2005, Tissue microarrays: bridging the gap between research and the clinic. Expert Rev Proteomics 2(3):325–336., DOI 10.1586/14789450.2.3.325 Hewitt SM, Takikita M, Abedi-Ardekani B, Kris Y, Bexfield K, Braunschweig T, Chung JY, 2008, Validation of proteomic-based discovery with tissue microarrays. Proteomics Clin Appl 2(10–11): 1460–1466., DOI 10.1002/prca.200800003 Battifora H, 1986, The multitumor, sausage, tissue block: novel method for immunohistochemical antibody testing. Lab Invest 55(2):244–248 Wan WH, Fortuna MB, Furmanski P, 1987, A rapid and efficient method for testing immunohistochemical reactivity of monoclonal antibodies against multiple tissue samples simultaneously. J Immunol Methods 103(1):121–129 Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi O-P, 1998, Tissue microarrays for high-throughputmolecular profiling of tumor specimens. Nat Med 4(7):844–847 Rimm DL, Nielsen TO, Jewell SD, Rohrer DC, Broadwater G, Waldman F, Mitchell KA, Singh B, Tsongalis GJ, Frankel WL, Magliocco AM, Lara JF, Hsi ED, Bleiweiss IJ, Badve SS, Chen B, Ravdin PM, Schilsky RL, Thor A, Berry DA, 2011, Cancer and leukemia group B Pathology Committee guidelines for tissue microarray construction representing multicenter prospective clinical trial tissues. J Clin Oncol 29(16):2282–2290., DOI 10.1200/JCO.2010.33. 2023 Gately K, Kerr K, O’Byrne K, 2011, Design, construction, and analysis of cell line arrays and tissue microarrays for gene expression analysis. Methods Mol Biol 784:139–153., DOI 10.1007/978-1- 61779-289-2_10 Gillett CE, Springall RJ, Barnes DM, Hanby AM, 2000, Multiple tissue core arrays in histopathology research: a validation study. J Pathol 192(4):549–553., DOI 10.1002/1096-9896(2000) Hewitt SM, 2004, Design, construction, and use of tissue microarrays. MethodsMol Biol 264:61–72., DOI 10.1385/1-59259-759-9:061 OnozatoML, Hammond S,MerrenM, Yagi Y, 2013, Evaluation of a completely automated tissue-sectioning machine for paraffin blocks. J Clin Pathol 66(2):151–154., DOI 10.1136/jclinpath-2011-200205 Torhorst J, Bucher C, Kononen J, Haas P, Zuber M, Kochli OR, Mross F, Dieterich H, Moch H, Mihatsch M, Kallioniemi OP, Sauter G, 2001, Tissue microarrays for rapid linking of molecular changes to clinical endpoints. Am J Pathol 159(6):2249–2256., DOI 10.1016/ S0002-9440(10)63075-1 ZhangD, Salto-TellezM, Putti TC, Do E, Koay ES, 2003, Reliability of tissue microarrays in detecting protein expression and gene amplification in breast cancer. Mod Pathol 16(1):79–84., DOI 10.1097/01. MP.0000047307.96344.93 Buesa RJ, 2010, Productivity standards for histology laboratories. Ann Diagn Pathol 14(2):107–124., DOI 10.1016/j.anndiagpath.2009. 12.005 Yang XR, Charette LA, Garcia-Closas M, Lissowska J, Paal E, Sidawy M, Hewitt SM, Rimm DL, Sherman ME, 2006, Construction and validation of tissuemicroarrays of ductal carcinoma in situ and terminal duct lobular units associated with invasive breast carcinoma. Diagn Mol Pathol 15(3):157–161., DOI 10.1097/01.pdm. 0000213453.45398.e0 Dancau AM, Simon R, Mirlacher M, Sauter G, 2010, Tissue microarrays. Methods Mol Biol 576:49–60., DOI 10.1007/978-1-59745- 545-9_4 Fedor HL, De Marzo AM, 2005, Practical methods for tissue microarray construction. Methods Mol Med 103:89–101 Shi SR, Key ME, Kalra KL, 1991, Antigen retrieval in formalinfixed, paraffin-embedded tissues: an enhancement method for immunohistochemical staining based on microwave oven heating of tissue Sections. J Histochem Cytochem 39(6):741–748 Tsao SC, Wu CC, Wen CH, Chai CY, Chen YT, 2013, Improved technique for manually constructing tissue microarrays for large-core arrays. Appl Immunohistochem Mol Morphol/Off Publ Soc ApplI mmunohistochem 21(1):85–89., DOI 10.1097/PAI.0b013e3182553527 Hoos A, Cordon-Cardo C, 2001, Tissue microarray profiling of cancer specimens and cell lines: opportunities and limitations. Lab Invest 81(10):1331–1338 Shebl AM, Zalata KR, Amin MM, El-Hawary AK, 2011, An inexpensive method of small paraffin tissue microarrays using mechanical pencil tips. Diagn Pathol 6:117., DOI 10.1186/1746-1596-6-117 Kajdacsy-Balla A, Geynisman JM, Macias V, Setty S, Nanaji NM, Berman JJ, Dobbin K, Melamed J, Kong X, BoslandM, Orenstein J, Bayerl J, Becich MJ, Dhir R, Datta MW, 2007, Practical aspects of planning, building, and interpreting tissue microarrays: the Cooperative Prostate Cancer Tissue Resource experience. J Mol Histol 38(2):113–121., DOI 10.1007/s10735-006-9054-5 Camp RL, Neumeister V, Rimm DL, 2008, A decade of tissue microarrays: progress in the discovery and validation of cancer biomarkers. J Clin Oncol 26(34):5630–5637., DOI 10.1200/JCO. 2008.17.3567 Catchpoole D, Mackie N, McIver S, Chetcuti A, Henwood A, Graf N, Arbuckle S, 2011, Tape transfer sectioning of tissue microarrays introduces nonspecific immunohistochemical staining artifacts. Biotech Histochem : Off Publ Biol Stain Comm 86(6):421–428., DOI 10.3109/10520295.2010.527859 Kampf C, Olsson I, Ryberg U, Sjostedt E, Ponten F, 2012, Production of tissue microarrays, immunohistochemistry staining and digitalization within the human protein atlas. J Vis Exp:, 63)., DOI 10.3791/3620 Fergenbaum JH, Garcia-Closas M, Hewitt SM, Lissowska J, Sakoda LC, Sherman ME, 2004, Loss of antigenicity in stored sections of breast cancer tissue microarrays. Cancer epidemiology, biomarkers& prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology 13, 4):667–672 Hoos A, Urist MJ, Stojadinovic A, Mastorides S, Dudas ME, Leung DH, Kuo D, Brennan MF, Lewis JJ, Cordon-Cardo C, 2001, Validation of tissue microarrays for immunohistochemical profiling of cancer specimens using the example of human fibroblastic tumors. Am J Pathol 158(4):1245–1251., DOI 10.1016/S0002-9440(10, 64075-8 Mucci NR, Akdas G, Manely S, Rubin MA, 2000, Neuroendocrine expression in metastatic prostate cancer: evaluation of high throughput tissue microarrays to detect heterogeneous protein expression. Human Pathol 31(4):406–414., DOI 10.1053/hp.2000.7295 Watanabe A, Cornelison R, Hostetter G, 2005, Tissue microarrays: applications in genomic research. Expert Rev Mol Diagn 5(2):171– 181., DOI 10.1586/14737159.5.2.171 Packeisen J, Korsching E, Herbst H, Boecker W, Buerger H, 2003, Demystified…tissue microarray technology. Mol Pathol 56(4):198– 204 Henriksen KL, Rasmussen BB, Lykkesfeldt AE, Moller S, Ejlertsen B, Mouridsen HT, 2007, Semi-quantitative scoring of potentially predictive markers for endocrine treatment of breast cancer: a comparison between whole sections and tissue microarrays. J Clin Pathol 60(4):397–404., DOI 10.1136/jcp.2005.034447 Divito KA, Berger AJ, Camp RL, Dolled-Filhart M, Rimm DL, Kluger HM, 2004, Automated quantitative analysis of tissue microarrays reveals an association between high Bcl-2 expression and improved outcome in melanoma. Cancer Res 64(23):8773–8777., DOI 10.1158/0008-5472.CAN-04-1387 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 719 EP 726 DI 10.1007/s12253-014-9752-3 PG 8 ER PT J AU Lang, D Marwitz, S Heilenkotter, U Schumm, W Behrens, O Simon, R Reck, M Vollmer, E Goldmann, T AF Lang, S. Dagmar Marwitz, Sebastian Heilenkotter, Uwe Schumm, W Behrens, Oliver Simon, Ronald Reck, Martin Vollmer, Ekkehard Goldmann, Torsten TI Transforming Growth Factor-Beta Signaling Leads to uPA/PAI-1 Activation and Metastasis: A Study on Human Breast Cancer Tissues SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Metastasis; Breast cancer; Urokinase-type plasminogen activator; Plasminogen activator inhibitor-1; Transforming growth factor beta; Mothers against decapentaplegic homolog 3; BMP Activin membrane-bound inhibitor homolog ID Metastasis; Breast cancer; Urokinase-type plasminogen activator; Plasminogen activator inhibitor-1; Transforming growth factor beta; Mothers against decapentaplegic homolog 3; BMP Activin membrane-bound inhibitor homolog AB Metastasis represents a major problem in the treatment of patients with advanced primary breast cancer. Both Transforming Growth Factor-Beta (TGF-β) signaling and Plasminogen Activator (PA) components, urokinasetype Plasminogen Activator (uPA) and Plasminogen Activator Inhibitor-1 (PAI-1) represent a complex network crucial for such enhanced invasiveness of tumors and imply high prognostic/predictive and promising therapeutic potential. Therefore, protein expression of specific effector molecules comprising the main parts of the TGF-β signaling pathway were determined in HOPE-fixed human tumor tissues through IHC (Scoring) using tissue microarray (TMA) technique and correlated with respective uPA and PAI-1 levels determined earlier in the same TMAs through optimized IHC and semi-quantitative image analysis. TGF-β signaling was active in vast majority (96 %) of the tumor samples and 88 % of all cases were significantly correlated with established metastasis markers uPA and PAI-1. In addition, TGF-β was also closely associated with tumor size, nodal status and two steroid hormone receptors. Consistent interrelationships between TGF-β, PA components and additional tumor characteristics underline the superiority of such more comprising data with regards to confirming TGF-β signaling as a promising target system to inhibit metastasis in advanced breast cancer. C1 [Lang, S. Dagmar] Research Center Borstel, Clinical and Experimental Pathology, Parkallee 3a, 23845 Borstel, Germany. [Marwitz, Sebastian] Research Center Borstel, Clinical and Experimental Pathology, Parkallee 3a, 23845 Borstel, Germany. [Heilenkotter, Uwe] Holsteinisches Breast Center, Gynecological Hospital, Clinical Center ItzehoeItzehoe, Germany. [Schumm, W] Holsteinisches Breast Center, imland Clinic Rendsburg, PathologyRendsburg, Germany. [Behrens, Oliver] Holsteinisches Breast Center, imland Clinic RendsburgRendsburg, Germany. [Simon, Ronald] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Reck, Martin] Airway Research Center North (ARCN)- Member of the German Center for Lung Research (DZL)Borstel, Germany. [Vollmer, Ekkehard] Research Center Borstel, Clinical and Experimental Pathology, Parkallee 3a, 23845 Borstel, Germany. [Goldmann, Torsten] Research Center Borstel, Clinical and Experimental Pathology, Parkallee 3a, 23845 Borstel, Germany. RP Lang, D (reprint author), Research Center Borstel, Clinical and Experimental Pathology, 23845 Borstel, Germany. EM dlang@fz-borstel.de CR Welch DR, Steeg PS, Rinker-Schaeffer CW, 2000, Molecular biology of breast cancer metastasis. Genetic regulation of human breast carcinoma metastasis. Breast Cancer Res 2:408–412 Lyer S, Wang Z-G, Akhtari M, Zhao W, Seth P, 2005, Targeting TGFβ signaling for cancer therapy. Cancer Biol Ther 4(3):261–266 Malinowsky K, Bollner C, Hipp S, Berg D, Schmitt M, Becker KF, 2010, UPA and PAI-1 analysis from fixed tissues—new perspectives for a known set of predictive markers. Curr Med Chem 17: 4370–4377, Review) Jo M, Eastman BM, Webb DL, Stoletov K, Klemke R, Gonias SL, 2010, Cell signaling by urokinase-type plasminogen activator receptor induces stem cell-like properties in breast cancer cells. Cancer Res 70(21):8948–8958 Harbeck N, SchmittM, VetterM, Krol J, Paepke D, Uhlig M, Paepke S, Janicke F, Geurts-Moespot A, von Minckwitz G, Sweep F, Thomssen C, 2008, Prospective biomarker trials Chemo NO and NNBC-3 Europe validate the clinical utility of invasion markers uPA and PAI-1 in node-negative breast cancer. Breast Care 3(suppl 2):11– 15 Kreienberg R, Albert U-S, Follmann M, Kopp I, Kuhn T,Wockel A, Zemmler T, 2012, Interdisziplinare S-3 Leitlinie fur Diagnostik, Therapie und Nachsorge des Mammakarzinoms. Published by German Cancer Society e.V Shiou S-R, Datta PK, Dhawan P, Law BK, Yingling JM, Dixon DA, Beauchamp RD, 2006, Smad4-dependent regulation of urokinase plasminogen activator secretion and RNA stability associated with invasiveness by autocrine and paracrine transforming growth factor- β. J Biol Chem 281:33971–33981 Song X, Thalacker FW, Nilsen-Hamilton M, 2012, Synergistic and multidimensional regulation of plasminogen activator inhibitor type 1 expression by transforming growth factor type β and epidermal growth factor. J Biol Chem 287:12520–12528 Kohn EA, Yang YA, Du Z, Nagano Y, Van Schyndle CM, Herrmann MA, Heldman M, Chen J-Q, Stuelten CH, Flanders KC, 2012, Wakefield LM, 2012, Biological responses of TGF-β in the mammary epitheliumshow a complex dependency on Smad3 gene dosage with important implications for tumor progression. Mol Cancer Res 10(10):1389–1399 Kocic J, Bugarski D, Santibanez JF, 2012, Smad3 is essential for transforming growth factor-β1-induced urokinase type plasminogen activator expression and migration in transformed keratinocytes. Eur J Cancer 48:1550–1557 Onichtchouk D, Chen YG, Dosch R, Gawantka V, Delius H, Massaque J, Niehrs C, 1999, Silencing of TGF-beta signaling by the pseudoreceptor BAMBI. Nature 401:480–485 Dromann D, Rupp J, Rohmann K, Osbahr S, Ulmer A, Marwitz S, Roschmann K, Abdullah M, Schultz H, Vollmer E, Zabel P, Dalhoff K, Goldmann T, 2010, The TGF-beta- pseudoreceptor BAMBI is strongly expressed in COPD lungs and regulated by nontypeable Haemophilus influenzae. Respir Res 11:67 Ito I, Hanyu A, Wayama M, Goto N, Katsuno N, Kawasaki S, Nakajima Y, Kajiro M, Komatsu Y, Fujimura A, Hirota R, Murayama A, Kimura K, Imamura T, Yanagisawa J, 2010, Estrogen inhibits transforming growth factor β signaling by promoting Smad2/3 degradation. J Biol Chem 285:14747–14755 Goto N, Hiyoshi H, Ito I, Tsuchiya M, Nakajima Y, Yanagisawa J, 2011, Estrogen and antiestrogens alter breast cancer invasiveness by modulating the transforming growth factor-β signaling pathway. Cancer Sci 102:1501–1508 Lang DS, Heilenkotter U, SchummW, Behrens O, Simon R, Vollmer E, Goldmann T, 2013, Optimized immunohistochemistry in combination with image analysis: a reliable alternative to quantitative ELISA determination of uPA and PAI-1 for routine risk group discrimination in breast cancer. Breast., DOI 10.1016/j.breast.2012.12. 011 Schulz H, Kahler D, Branscheid D, Vollmer E, Zabel P, Goldmann T, 2008, TKTL1 is overexpressed in a large portion of non-small cell lung cancer specimens. Diagn Pathol 3:35 Vollmer E, Galle J, Lang DS, Loeschke S, Schultz H, Goldmann T, 2006, The HOPE technique opens up a multitude of new possibilities in pathology. Rom J Morphol Embryol 47(1):15–19, Review) Marwitz S, Abdullah M, Vock C, Fine JS, Visvanathan S, Gaede KI, Hauber HP, Zabel P, Goldmann T, 2011, HOPE-BAL: improved molecular diagnostics by application of a novel technique for fixation and paraffin embedding. J Histochem Cytochem 59(6):601–614 Tobar N, Villar V, Santibanez JF, 2010, ROS-NFkB mediates TGF- β1-induced expression of urokinase-type plasminogen activator, matrix metalloproteinase-9 and cell invasion. Mol Cell Biochem 340: 195–202 Giehl K, Imamichi Y, Menke A, 2007, Smad4-independent TGF–β signaling in tumor cell migration. Cells Tissues Organs 185:123–130 Wakefield LM, Roberts AB, 2002, TGF-b signaling: positive and negative effects on tumorigenesis. Curr Opin Genet Dev 12:22–29, Review) Wolff C, Malinoswky K, Berg D, Schragner K, Schuster T,Walch A, Bronger H, Hofler H, Becker K-F, 2011, Signalling networks associated with urokinase-type plasminogen activator, uPA, and ist inhibitor PAI-1 in breast cancer tissues: new insights from protein microarray analysis. J Pathol 223:54–63 Malinowsky K, Wolff C, Berg D, Schuster T, Walch A, Bronger H, Mannsperger H, Schmidt C, Korf U, Hofler H, Becker K-F, 2012, uPA and PAI-1-related signaling pathways differ between primary breast cancers and lymph node metastases. Trans Oncol 5:98–104 Czekay R-P, Wilkins-Port CE, Higgins SP, Freytag J, Overstreet JM, Klein RM, Higgins CE, Samarakoon R, Higgins PJ, 2011, PAI-1: an integrator of cell signaling and migration. Int J Cell Biol 2011:1–9, Review) Huang SS, Ling T-Y, Tseng W-F, Huang Y-W, Tang F-M, Leal SM, Huang JS, 2003, FASEB J 17:2068–2080 Khin SS, Kitazawa R,Win N, Aye TT,Mori K, Kondo T, Kitazawa S, 2009, BAMBI gene is epigenetically silenced in subset of highgrade bladder. Int J Cancer 125:328–338 Ganapathy V, Rongrong G, Grazioli A, Xie W, Banach-Petrosky W, Kang Y, Lonning S, McPherson J, Yingling JM, Biswas S, Mundy GR, Reiss M, 2010, Targeting the transforming growth factor–β pathway inhibits human basal-like breast cancer metastasis. Mol Cancer 9:122 Liu J, Liao S, Diop-Frimpong B, Chen W, Goel S, Naxerova K, Ancukiewicz M, Boucher Y, Jain RK, Xu L, 2012, TGF-β blockage improves the distribution and efficiacy of therapeutics in breast carcinoma by normalizing the tumor stroma. PNAS 109:16618–16623 Matise LA, Palmer TD, Ashby WJ, Nashabi A, Chytil A, Aakre M, Pickup MW, Gorska AE, Zijlstra A, Moses HL, 2012, Lack of transforming growth factor-β signaling promotes collective cancer cell invasion through tumor-stromal crosstalk. Breast Cancer Res 14: R98 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 727 EP 732 DI 10.1007/s12253-014-9753-2 PG 6 ER PT J AU Liu, G Xie, B Gong, L Zhou, J Shu, G AF Liu, Ganglei Xie, Biao Gong, Lin Zhou, Jianping Shu, Guoshun TI The Expression of p66Shc Protein in Benign, Premalignant, and Malignant Gastrointestinal Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Colorectal cancer; p66Shc; Neoplasia; Hyperplastic polyps; Immunohistochemistry ID Gastric cancer; Colorectal cancer; p66Shc; Neoplasia; Hyperplastic polyps; Immunohistochemistry AB ROS produced from Oxidative stress have long been recognized to be involved in carcinogenesis. p66Shc generates H2O2 by oxidizing cytochrome c, and its expression has been reported to be elevated in several tumors. However, the expression of p66Shc in gastric cancer has not been reported, and its role in colorectal cancer has not been well elucidated. This study investigated p66Shc expression in benign, premalignant, and malignant gastric and colorectal lesions. p66Shc expression in 146 gastric tumors, 136 colorectal tumors, 45 gastric hyperplastic polyps, 33 gastric low-grade intraepithelial neoplasias, 41 gastric high-grade intraepithelial neoplasias, 42 colorectal hyperplastic polyps, 21 colorectal low-grade intraepithelial neoplasias, 38 colorectal high-grade intraepithelial neoplasias, and 30 normal gastric and colorectal tissues was measured by immunohistochemistry. Most normal gastric and colorectal tissues exhibited low or no p66Shc expression (93.4 %), while most gastric and colorectal tumors exhibited moderate to high p66Shc expression (78.1 %– 80.9 %). The p66Shc expression in normal gastric and colorectal tissues were significantly lower than that in the lowgrade intraepithelial neoplasias (p<0.05), high-grade intraepithelial neoplasias (p<0.01), and gastric adenocarcinomas (p<0.01 or <0.001). No differences in p66Shc expression were observed in gastric and colorectal hyperplastic polyps compared to the normal tissues. No statistically significant differences in p66Shc expression were observed between patients with different disease stages, different tumor grades, and with or without lymph node metastasis in gastric and colorectal cancers. In conclusion, p66Shc may be involved in the carcinogenesis of gastric and colorectal cancers and could be a marker for the diagnosis of gastric and colorectal cancers. C1 [Liu, Ganglei] Central South University, Second Xiangya Hospital, Department of Geriatric Surgery, 410011 Changsha, Hunan, China. [Xie, Biao] Central South University, Second Xiangya Hospital, Department of Geriatric Surgery, 410011 Changsha, Hunan, China. [Gong, Lin] Central South University, Second Xiangya Hospital, Department of Geriatric Surgery, 410011 Changsha, Hunan, China. [Zhou, Jianping] Central South University, Second Xiangya Hospital, Department of Geriatric Surgery, 410011 Changsha, Hunan, China. [Shu, Guoshun] Central South University, Second Xiangya Hospital, Department of Geriatric Surgery, 410011 Changsha, Hunan, China. RP Zhou, J (reprint author), Central South University, Second Xiangya Hospital, Department of Geriatric Surgery, 410011 Changsha, China. EM zhoujiangping88@yahoo.com CR Ray PD, Huang BW, Tsuji Y, 2012, Reactive oxygen species, ROS, homeostasis and redox regulation in cellular signaling. Cell Signal 24:981–990 Azad MB, Chen Y, Gibson SB, 2009, Regulation of autophagy by reactive oxygen species, ROS): implications for cancer progression and treatment. Antioxid Redox Signal 11:777–790 Pelicano H, Carney D,Huang P, 2004, ROS stress in cancer cells and therapeutic implications. Drug Resist Updat 7:97–110 Storz P, 2005, Reactive oxygen species in tumor progression. Front Biosci 10:1881–1896 Oberley LW, 2005, Mechanism of the tumor suppressive effect of MnSOD overexpression. Biomed Pharmacother 59:143–148 Wiseman H, Halliwell B, 1996, Damage to DNA by reactive oxygen and nitrogen species: role in inflammatory disease and progression to cancer. Biochem J 313:17–29 Leto TL, Geiszt M, 2006, Role of Nox family NADPH oxidases in host defense. Antioxid Redox Signal 8:1549–1561 Yoshida S, Masaki T, Feng H, Yuji J, Miyauchi Y, Funaki T et al, 2004, Enhanced expression of adaptor molecule p46 Shc in nuclei of hepatocellular carcinoma cells: study of LEC rats. Int J Oncol 25: 1089–1096 Ravichandran KS, 2001, Signaling via Shc family adapter proteins. Oncogene 20:6322–6330 Rajendran M, Thomes P, Zhang L, Veeramani S, Lin MF, 2010, p66Shc–a longevity redox protein in human prostate cancer progression and metastasis: p66Shc in cancer progression and metastasis. Cancer Metastasis Rev 29:207–222 Migliaccio E, Giorgio M, Mele S, Pelicci G, Reboldi P, Pandolfi PP et al, 1999, The p66shc adaptor protein controls oxidative stress response and life span in mammals. Nature 402:309–313 Giorgio M, Migliaccio E, Orsini F, Paolucci D, Moroni M, Contursi C et al, 2005, Electron transfer between cytochrome c and p66 Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell 122:221–233 Jackson JG, Yoneda T, Clark GM, Yee D, 2000, Elevated levels of p66 Shc are found in breast cancer cell lines and primary tumors with high metastatic potential. Clin Cancer Res 6:1135–1139 Abdollahi A, Gruver BN, Patriotis C, Hamilton TC, 2003, Identification of epidermal growth factor-responsive genes in normal rat ovarian surface epithelial cells. Biochem Biophys Res Commun 18:188–197 Park YJ, Kim TY, Lee SH, Kim H, Kim SW, Shong M et al, 2005, p66shc expression in proliferating thyroid cells is regulated by thyrotropin receptor signaling. Endocrinology 146:2473–2480 Galimov ER, Sidorenko AS, Tereshkova AV, Pletiushkina OI, Cherniak BV, Chumakov PM, 2012, P66shc action on resistance of colon carcinoma RKO cells to oxidative stress. Mol Biol, Mosk, 46: 139–146 Grossman SR, Lyle S, Resnick MB, Sabo E, Lis RT, Rosinha E et al, 2007, p66 Shc tumor levels show a strong prognostic correlation with disease outcome in stage IIA colon cancer. Clin Cancer Res 13: 5798–5804 Lee MS, Igawa T, Chen SJ, Van Bemmel D, Lin JS, Lin FF et al, 2004, p66Shc protein is upregulated by steroid hormones in hormone-sensitive cancer cells and in primary prostate carcinomas. Int J Cancer 108:672–678 Hamilton SR, Aaltonen LA, 2000, WHO classification of tumours: pathology and genetics of tumours of the digestive system. IARC Press, Lyon Dango S, SienelW, Schreiber M, Stremmel C, Kirschbaum A, Pantel K, Passlick B, 2008, Elevated expression of carcino-embryonic antigen-related cell adhesionmolecule 1, CEACAM-1, is associated with increased angiogenic potential in non-small-cell lung cancer. Lung Cancer 60:426–433 Yukimasa S, Masaki T, Yoshida S, Uchida N, Watanabe S, Usuki H, Yoshiji H, Maeta T, Ebara K, Nakatsu T, Kurokohchi K, Kuriyama S, 2005, Enhanced expression of p46 Shc in the nucleus and p52 Shc in the cytoplasm of human gastric cancer. Int J Oncol 26:905–911 Ventura A, Luzi L, Pacini S, Baldari CT, Pelicci PG, 2002, The p66Shc longevity gene is silenced through epigenetic modifications of an alternative promoter. J Biol Chem 277:22370–22376 TrineiM, GiorgioM, Cicalese A, Barozzi S, Ventura A, Migliaccio E et al, 2002, Ap53-p66Shc signalling pathway controls intracellular redox status, levels of oxidationdamagedDNAand oxidative stressinduced apoptosis. Oncogene 21:3872–3878 Pellegrini M, Pacini S, Baldari CT, 2005, p66SHC: the apoptotic side of Shc proteins. Apoptosis 10:13–18 Pani G, Koch OR, Galeotti T, 2009, The p53-p66shc-Manganese Superoxide Dismutase, MnSOD, network: a mitochondrial intrigue to generate reactive oxygen species. Int J Biochem Cell Biol 41:1002–1005 Nicco C, Laurent A, Chereau C, Weill B, Batteux F, 2005, Differential modulation of normal and tumor cell proliferation by reactive oxygen species. Biomed Pharmacother 59:169–174 Xiao D, Singh SV, 2010, p66Shc is indispensable for phenethyl isothiocyanate-induced apoptosis in human prostate cancer cells. Cancer Res 70:3150–3158 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 733 EP 739 DI 10.1007/s12253-014-9754-1 PG 7 ER PT J AU Marsovszky, L Resch, DM Visontai, Zs Nemeth, J AF Marsovszky, Laszlo Resch, D Miklos Visontai, Zsuzsanna Nemeth, Janos TI Confocal Microscopy of Epithelial and Langerhans Cells of the Cornea in Patients Using Travoprost Drops Containing Two Different Preservatives SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Benzalkonium chloride; Confocal microscopy; Langerhans cell; Polyquaternium ID Benzalkonium chloride; Confocal microscopy; Langerhans cell; Polyquaternium AB The recently developed confocal cornea microscopy offers the opportunity to examine pathologies of the cornea and to gain insight into the activity of innate immunity. We aimed to investigate the corneal epithelial and Langerhans cell (LC) densities along with dry eye parameters in primary openangle glaucoma (POAG) subjects, treated with either of two commercially available travoprost 0.004 % topical medications containing different preservatives. (1: benzalkonium chloride 0.015 % (TravBAK) and 2: polyquaternium-1 (PQ) 0.001%(TravPQ). Consecutive case series of nineteen POAG patients on TravBAK (mean age: 64.8±13.6 years), nineteen POAG patients on TravPQ (mean age: 66.8±11.3 years) and nineteen age-matched healthy control subjects (63.8±8.2 years). Ocular surface disease index (OSDI), lid parallel conjunctival folds (LIPCOF), Schirmer test (ST) and tear break up time (TBUT) were assessed, and then corneal epithelial and LC densities were investigated with confocal microscopy. Tear production was significantly reduced in both glaucoma patient groups compared to healthy individuals (p<0.05). TBUT was significantly reduced and epithelial cell densities were significantly greater in patients treated with TravBAK compared to healthy individuals (p<0.05 for all). LC densities were greater in both glaucoma groups compared to control subjects (p<0.05 for all). Travoprost therapy may compromise ocular surface. The limited alertness of the corneal immune system found in patients with TravPQ can be considered as indicators of a less disturbed ocular surface and better controlled corneal homeostasis. C1 [Marsovszky, Laszlo] Semmelweis University, Department of Ophthalmology, Maria u. 39, 1085 Budapest, Hungary. [Resch, D Miklos] Semmelweis University, Department of Ophthalmology, Maria u. 39, 1085 Budapest, Hungary. [Visontai, Zsuzsanna] Semmelweis University, Department of Ophthalmology, Maria u. 39, 1085 Budapest, Hungary. [Nemeth, Janos] Semmelweis University, Department of Ophthalmology, Maria u. 39, 1085 Budapest, Hungary. RP Marsovszky, L (reprint author), Semmelweis University, Department of Ophthalmology, 1085 Budapest, Hungary. EM marsovsz@hotmail.com CR Martone G, Frezzotti P, Tosi GM, Traversi C, Mittica V, Malandrini A, Pichierri P, Balestrazzi A, Motolese PA, Motolese I, Motolese E, 2009, An in vivo confocal microscopy analysis of effects of topical antiglaucoma therapy with preservative on corneal innervation and morphology. Am J Ophthalmol 147:725–735 Epstein SP, Chen D, Asbell PA, 2009, Evaluation of biomarkers of inflammation in response to benzalkonium chloride on corneal and conjunctival epithelial cells. J Ocul Pharmacol and Ther 25:415–424 Noecker RJ, Herrygers LA, Anwaruddin R, 2004, Corneal and conjunctival changes caused by commonly used glaucoma medications. Cornea 23:490–496 Whitson JT, Petroll WM, 2012, Corneal epithelial cell viability following exposure to ophthalmic solutions containing preservatives and/or antihypertensive agents. Adv Ther 29:874–888 Horsley MB, Kahook MY, 2009, Effects of prostaglandin analog therapy on the ocular surface of glaucoma patients. Clin Ophthalmol 3:291–295 Labbe A, Pauly A, Liang H, Brignole-Baudouin F,Martin C,Warnet JM, Baudouin C, 2006, Comparison of toxicological profiles of benzalkonium chloride and polyquaternium-1: an experimental study. J Ocul Pharmacol 22:267–278 Aptel F, Cucherat M, Denis P, 2008, Efficacy and tolerability of prostaglandin analogs: a meta-analysis of randomized controlled clinical trials. J Glaucoma 17:667–673 Hamrah P, Huq SO, Liu Y, Zhang Q, Dana MR, 2003, Corneal immunity is mediated by heterogeneous population of antigenpresenting cells. J Leuk Biol 74:172–178 Hazlett LD, McClellan SM, Hume EB, Dajcs JJ, O’Callaghan RJ, 1999, Willcox, M.D. Extended wear contact lens wear usage induces Langerhans cell migration into cornea. Exp Eye Res 69:575– 577 Zhivov A, Stave J, Vollmar B, Guthoff R, 2007, In vivo confocal microscopic evaluation of Langerhans cell density and distribution in the corneal epithelium of healthy volunteers and contact lens wearers. Cornea 26:47–54 Marsovszky L, Resch MD, Nemeth J, Toldi G, Medgyesi E, Kovacs L, BalogA, 2013, In vivo confocal microscopic evaluation of corneal Langerhans cell density, and distribution and evaluation of dry eye in rheumatoid arthritis. Innate Immun 19:348–354 Pult H, Purslow C, Murphy PJ, 2011, The relationship between clinical signs and dry eye symptoms. Eye 25:502–510 Zhivov A, Stave J, Vollmar B, Guthoff R, 2005, In vivo confocal microscopic evaluation of Langerhans cells density and distribution in the normal corneal epithelium. Graefe’s Arch Clin Exp Ophthalmol 243:1056–1061 Marsovszky L, Nemeth J, ReschMD, Toldi G, Legany N, Kovacs L, Balog A, 2013, Corneal Langerhans cell and dry eye examinations in ankylosing spondylitis. Innate Immun Aug 19 Baratz KH, Nau CB, Winter EJ, McLaren JW, Hodge DO, Herman DC, Bourne WM, 2006, Effects of glaucoma medications on corneal endothelium, keratocytes, and subbasal nerves among participants in the Ocular Hypertension Treatment Study. Cornea 25:1046–1052 Ammar DA, Kahook MY, 2011, Effects of benzalkonium chlorideor polyquad-preserved fixed combination glaucoma medications on human trabecular meshwork cells. Mol Vision 17:1806–1813 Guenoun JM, Baudouin C, Rat P, Pauly A, Warnet JM, Brignole- Baudouin F, 2005, In vitro comparison of cytoprotective and antioxidative effects of latanoprost, travoprost, and bimatoprost on conjuctiva-derived epithelial cells. Inv Ophth Vis Sci 46: 4594–4599 Villani E, Beretta S, De Capitani M, Galimberti D, Viola F, Ratiglia R, 2011, In vivo confocal microscopy of meibomian glands in Sjogren’s syndrome. Invest Ophthalmol Vis Sci 52: 933–939 Pisella PJ, Pouliquen P, Baudouin C, 2002, Prevalence of ocular symptoms and signs with preserved and preservative-free glaucoma medications. Br J Ophthalmol 86:418–423 Zhivov A, Kraak R, Bergter H, Kundf G, Beck R, Guthoff RF, 2010, Influence of benzalkonium chloride on Langerhans cells in corneal epithelium and development of dry eye in healthy volunteers. Curr Eye Res 35:762–769 Gery I, Davies P, Derr J, Krett N, Barranger JA, 1981, Relationship between production and release of lymphocyte-activating factor, interleukin-1, by murine macrophages: 1. Effects of various agents. Cell Immunol 64:293–303 Niederkorn JY, Peeler JS, Mellon J, 1989, Phagocytosis of particulate antigens by corneal epithelial cells stimulate interleukin-1 secretion and migration of Langerhans cells into the central cornea. Reg Immunol 2:83–90 Dekaris I, Zhu SN, Dana MR, 1999, TNF-alpha regulates corneal Langerhans cell migration. J Immunol 162:4235–4239 Hattori T, Chauhan SK, Lee H, Ueno H, Dana MR, 2011, Characterization of Langerin-Expressing Dendritic Cell Subsets in the Normal Cornea. Invest Ophthalmol Vis Sci 52:4598–4604 Zhivov A, Stachs O, Kraak R, Stave J, Guthoff RF, 2006, In vivo confocal microscopy of the ocular surface. Ocul Surf 4: 81–93 Liang H, Brignole-Baudouin F, Riancho L, Baudouin C, 2012, Reduced in vivo ocular surface toxicity with polyquad-preserved travoprost versus benzalkonium-preserved travoprost or latanoprost ophthalmic solutions. Ophthalmic Res 48:89–101 Paimela T, Ryhanen T, Kauppinen A, Marttila L, Salminen A, Kaarniranta K, 2012, The preservative polyquaternium-1 increases cytoxicity and NFkappaB linked inflammation in human corneal epithelial cells. Mol Vis 18:1189–1196 Ranno S, Fogagnolo P, Rossetti L, Orzalesi N, Nucci P, 2011, Changes in corneal parameters at confocal microscopy in treated glaucoma patients. Clin Ophthalmol 5:1037–1042 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 741 EP 746 DI 10.1007/s12253-014-9755-0 PG 6 ER PT J AU Wu, Ch Yang, Sh Sun, Z Han, X Ye, Y Liu, Sh AF Wu, Chunyu Yang, Shunfang Sun, Zhenping Han, Xianghui Ye, Yiyi Liu, Sheng TI Characterization of the Attenuation of Breast Cancer Bone Metastasis in Mice by Zoledronic Acid Using 99mTc bone Scintigraphy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bone metastasis; Breast cancer; 99mTc-MDP bone scintigraphy; Zoleronic acid ID Bone metastasis; Breast cancer; 99mTc-MDP bone scintigraphy; Zoleronic acid AB Metastatic breast cancer often metastasizes to bone. The purposes of the study were (1) to evaluate the use of 99mTc-MDP bone scintigraphy for detection of metastatic bone lesions, and (2) to determine the efficacy of zoledronic acid in mice with breast cancer bone metastasis. All tumorbearing mice were analyzed with radionuclide bone scintigraphy, X-ray, and histological analysis. The metastatic bone tissue was also harvested and analyzed by western blotting and real-time qPCR. Interestingly, zoledronic acid significantly decreased both the tumor burden and the incidence of bone metastasis in mice. In addition, histomorphometric, stereological, and molecular biology analyses demonstrated that zoledronic acid may function to inhibit breast cancer cell growth in the bone microenvironment and regulate the function of osteoblasts and osteoclasts in tumor-bearing mice. Finally, the attenuation of breast cancer bone metastasis using zoledronic acid can be accurately characterized by 99mTc bone scintigraphy in mice. C1 [Wu, Chunyu] Shanghai University of Traditional Chinese Medicine, Longhua Hospital, Department of Breast Surgery, 725 South Wanping Road, 200032 Shanghai, China. [Yang, Shunfang] Shanghai JiaoTong University, Shanghai Chest Hospital, Department of Nuclear Medicine, 200030 Shanghai, China. [Sun, Zhenping] Shanghai University of Traditional Chinese Medicine, Longhua Hospital, Department of Breast Surgery, 725 South Wanping Road, 200032 Shanghai, China. [Han, Xianghui] Shanghai University of Traditional Chinese Medicine, Longhua Hospital, Pharmacology Laboratory of Traditional Chinese Medicine, 200032 Shanghai, China. [Ye, Yiyi] Shanghai University of Traditional Chinese Medicine, Longhua Hospital, Pharmacology Laboratory of Traditional Chinese Medicine, 200032 Shanghai, China. [Liu, Sheng] Shanghai University of Traditional Chinese Medicine, Longhua Hospital, Department of Breast Surgery, 725 South Wanping Road, 200032 Shanghai, China. RP Liu, Sh (reprint author), Shanghai University of Traditional Chinese Medicine, Longhua Hospital, Department of Breast Surgery, 200032 Shanghai, China. EM sliu_tcm@163.com CR Chen WQ, Zeng HM, Zheng RS, Zhang SW, He J, 2012, Cancer incidence and mortality in China, 2007. Chin J Cancer Res 24:1–8 Linos E, Spanos D, Rosner BA, Linos K, Hesketh T, Qu JD et al, 2008, Effects of reproductive and demographic changes on breast cancer incidence in China: a modeling analysis. J Natl Cancer Inst 100:1352–1360 Lipton A, Uzzo R, Amato RJ, Ellis GK, Hakimian B, Roodman GD et al, 2009, The science and practice of bone health in oncology: managing bone loss and metastasis in patients with solid tumors. J Natl Compr Cancer Netw Suppl 7:S1–S29 Roodman GD, 2004, Mechanisms of bone metastasis. N Engl J Med 350:1655–1664 Anderson KC, Alsina M, Bensinger W, Biermann JS, Chanan-Khan A, Cohen AD, et al, 2010, NCCN clinical practice guidelines in oncology: multiple myeloma, 2010, v.2. Available at: http://www. 24hmb.com/UpLoad/Editor/2010/1/4/2010010471312017.pdf Luo KW, Ko CH, Yue GG, Lee MY, Siu WS, Lee JK et al, 2013, Anti-tumor and anti-osteolysis effects of the metronomic use of zoledronic acid in primary and metastatic breast cancer mouse models. Cancer Lett 339:42–48 Insalaco L, Di Gaudio F, TerrasiM, Amodeo V, Caruso S, Corsini LR et al, 2012, Analysis of molecular mechanisms and anti-tumoural effects of zoledronic acid in breast cancer cells. J Cell Mol Med 16: 2186–2195 Hiraga T, Williams PJ, Ueda A, Tamura D, Yoneda T, 2004, Zoledronic acid inhibits visceral metastases in the 4T1/luc mouse breast cancer model. Clin Cancer Res 10:4559–4567 Jeong J, Lee KS, Choi YK, Oh YJ, Lee HD, 2011, Preventive effects of zoledronic acid on bone metastasis in mice injected with human breast cancer cells. J Korean Med Sci 26:1569–1575 Koto K, Horie N, Kimura S, Murata H, Sakabe T, Matsui T et al, 2008, Clinically relevant dose of zoledronic acid inhibits spontaneous lung metastasis in a murine osteosarcoma model. Cancer Lett 274:271–278 Rachner TD, Singh SK, Schoppet M, Benad P, Bornhauser M, Ellenrieder V et al, 2010, Zoledronic acid induces apoptosis and changes the TRAIL/OPG ratio in breast cancer cells. Cancer Lett 287:109–116 Hiraga T, Williams PJ, Mundy GR, Yoneda T, 2001, The bisphosphonate ibandronate promotes apoptosis in MDA-MB-231 human breast cancer cells in bone metastases. Cancer Res 61:4418–4424 Yoneda T,Willams PJ, Hiraga T, NiewolnaM, Nishimura R, 2001)A bone-seeking clone exhibits different biological properties from the MDA-MB-231 parental human breast cancer cells and a brainseeking clone in vivo and in vitro. J Bone Miner Res 16:1486–1495 El-Abdaimi K, Ste-Marie LG, Papavasiliou V, Dion N, Cardinal PE, Huang D et al, 2003, Pamidronate prevents the development of skeletal metastasis in nude mice transplanted with human breast cancer cells by reducing tumor burden within bone. Int J Oncol 22: 883–890 Even-Sapir E, 2005, Imaging of malignant bone involvement by morphologic, scintigraphic, and hybrid modalities. J Nucl Med 46: 1356–1367 Hamaoka T, Madewell JE, Podoloff DA, Hortobagyi GN, Ueno NT, 2004, Bone imaging in metastatic breast cancer. J Clin Oncol 22: 2942–2953 Padhani A, Husband J, 1998, Bone metastases. In: Husband JES, Reznek RH, eds, Imaging in oncology oxford. Isis Medical Media Ltd, U.K., pp 765–787 Yang S, Dong Q, Yao M, Shi M, Ye J, Zhao L et al, 2009, Establishment of an experimental human lung adenocarcinoma cell line SPC-A-1BM with high bone metastases potency by 99mTc– MDP bone scintigraphy. Nucl Med Biol 36:313–321 Wu C, Sun Z, Ye Y, Han X, Song X, Liu S, 2013, Psoralen inhibits bone metastasis of breast cancer in mice. Fitoterapia 91:205–210 Chen M, Ye Y, Ji G, Liu J, 2010, Hesperidin upregulates heme oxygenase–1 to attenuate hydrogen peroxide-induced cell damage in hepatic L02 cells. J Agric Food Chem 58:3330–3335 Mandal MN, Vasireddy V, Jablonski MM, Wang X, Heckenlively JR, Hughes BA et al, 2006, Spatial and temporal expression of MFRP and its interaction with CTRP5. Invest Ophthalmol Vis Sci 47:5514–5521 Coleman RE, Marshall H, Cameron D, Dodwell D, Burkinshaw R, KeaneMet al, 2011, Breast-cancer adjuvant therapy with zoledronic acid. N Engl J Med 365:1396–1405 Costa L, 2007, Bisphosphonates: reducing the risk of skeletal complications from bone metastasis. Breast 16:S16–S20 Gallo M, Luca AD, Lamura L, Normanno N, 2012, Zoledronic acid blocks the interaction between mesenchymal stem cells and breast cancer cells: implications for adjuvant therapy of breast cancer. Ann Oncol 23:597–604 Niikura N, Liu J, Hayashi N, Palla S, Tokuda Y, Hortobagyi G et al, 2012, Retrospective analysis of antitumor effects of zoledronic acid in breast cancer patients with bone-only metastases. Cancer 118: 2039–2047 Fournier P, Boissier S, Filleur S, Guglielmi J, Cabon F, Colombel M et al, 2002, Bisphosphonates inhibit angiogenesis in vitro and testosterone-stimulated vascular regrowth in the ventral prostate in castrated rats. Cancer Res 62:6538– 6544 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2014 VL 20 IS 3 BP 747 EP 754 DI 10.1007/s12253-014-9756-z PG 8 ER PT J AU Somoracz, Korompay, A Torzsok, P Patonai, A Erdelyi-Belle, B Lotz, G Schaff, Zs Kiss, A AF Somoracz, Aron Korompay, Anna Torzsok, Peter Patonai, Attila Erdelyi-Belle, Boglarka Lotz, Gabor Schaff, Zsuzsa Kiss, Andras TI Tricellulin Expression and its Prognostic Significance in Primary Liver Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Tricellulin; Hepatocellular carcinoma; Cholangiocellular carcinoma; Prognosis ID Tricellulin; Hepatocellular carcinoma; Cholangiocellular carcinoma; Prognosis AB Numerous data suggest that altered expression of tight junction proteins such as occludin and claudins plays important role in carcinogenesis. However, little is known about tricellulin, a transmembrane tight junction protein concentrated where three epithelial cells meet. We aimed to characterize tricellulin expression in normal and cirrhotic liver in comparison to primary hepatic neoplasms. Tricellulin expression of 20 control livers, 12 cirrhotic livers, 32 hepatocell u l a r carcinomas (HCC), and 20 i n t rahepatic cholangiocarcinomas (iCCC) was investigated by immunohistochemistry and Western blotting. Co-localization of tricellulin with claudin-1, -4, and MRP2 was studied using double immunofluorescence. Scattered tricellulin immunopositivity was restricted to biliary pole of hepatocytes confirmed by co-localization with MRP2. Moreover, spottedlike reaction was observed between bile duct epithelial cells. In 40 % of HCCs marked tricellulin overexpression was measured regardless of tumor grades. In iCCCs, however, tricellulin expression decreased parallel with dedifferentiation. In HCCs high tricellulin expression, in iCCCs low tricellulin expression correlated with poor prognosis. Co-localization with MRP2 might substantiate that tricellulin plays role in blood-biliary barrier. Overexpressed tricellulin in a subset of HCCs correlated with unfavorable prognosis. Similar to ductal pancreatic adenocarcinoma, higher grades of iCCCs were associated with decreased tricellulin expression correlating with poor prognosis. C1 [Somoracz, Aron] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Korompay, Anna] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Torzsok, Peter] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Patonai, Attila] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Erdelyi-Belle, Boglarka] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Kiss, A (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM kiss.andras@med.semmelweis-univ.hu CR Zigler M, Dobroff AS, Bar-Eli M, 2010, Cell adhesion: implication in tumor progression. Minerva Med 101:149–162 Turksen K, Troy TC, 2011, Junctions gone bad: claudins and loss of the barrier in cancer. Biochim Biophys Acta 1816:73–79 Martin TA, Mason MD, Jiang WG, 2011, Tight junctions in cancer metastasis. Front Biosci 16:898–936 Ikenouchi J, FuruseM, Furuse K et al, 2005, Tricellulin constitutes a novel barrier at tricellular contacts of epithelial cells. J Cell Biol 171: 939–945 Chiba H, Osanai M, Murata M et al, 2008, Transmembrane proteins of tight junctions. Biochim Biophys Acta 1778:588–600 Riazuddin S, Ahmed ZM, Fanning AS et al, 2006, Tricellulin is a tight-junction protein necessary for hearing. Am J Hum Genet 79: 1040–1051 Chishti MS, Bhatti A, Tamim S et al, 2008, Splice-site mutations in the TRIC gene underlie autosomal recessive nonsyndromic hearing impairment in Pakistani families. J Hum Genet 53:101–105 Kondoh A, Takano K, Kojima T et al, 2011, Altered expression of claudin-1, claudin-7, and tricellulin regardless of human papilloma virus infection in human tonsillar squamous cell carcinoma. Acta Otolaryngol 131:861–868 Korompay A, Borka K, Lotz G et al, 2012, Tricellulin expression in normal and neoplastic human pancreas. Histopathology 60(6B):E76– E78 Patonai A, Erdelyi-Belle B, Korompay A et al, 2011, Claudins and tricellulin in fibrolamellar hepatocellular carcinoma. Virchows Arch 458:679–688 Parkin DM, Bray F, Ferlay J et al, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Hammill CW, Wong LL, 2008, Intrahepatic cholangiocarcinoma: a malignancy of increasing importance. J Am Coll Surg 207:594–603 Nemeth Z, Szasz AM, Tatrai P et al, 2009, Claudin-1, -2, -3, -4, -7, - 8, and -10 protein expression in biliary tract cancers. J Histochem Cytochem 57:113–121 Lodi C, Szabo E, Holczbauer A et al, 2006, Claudin-4 differentiates biliary tract cancers from hepatocellular carcinomas. Mod Pathol 19: 460–469 Liu S, Yang W, Shen L et al, 2009, Tight junction proteins claudin- 1 and occludin control hepatitis C virus entry and are downregulated during infection to prevent superinfection. J Virol 83:2011– 2014 Evans MJ, von Hahn T, Tscherne DM et al, 2007, Claudin-1 is a hepatitis C virus co-receptor required for a late step in entry. Nature 446:801–805 Sakaguchi T, Suzuki S, Higashi H et al, 2008, Expression of tight junction protein claudin-5 in tumor vessels and sinusoidal endothelium in patients with hepatocellular carcinoma. J Surg Res 147:123– 131 Higashi Y, Suzuki S, Sakaguchi T et al, 2007, Loss of claudin-1 expression correlates with malignancy of hepatocellular carcinoma. J Surg Res 139:68–76 Huang GW, Ding X, Chen SL et al, 2011, Expression of claudin 10 protein in hepatocellular carcinoma: impact on survival. J Cancer Res Clin Oncol 137:1213–1218 Cheung ST, Leung KL, Ip YC et al, 2005, Claudin-10 expression level is associated with recurrence of primary hepatocellular carcinoma. Clin Cancer Res 11:551–556 Shinozaki A, Shibahara J, Noda N et al, 2011, Claudin-18 in biliary neoplasms. Its significance in the classification of intrahepatic cholangiocarcinoma. Virchows Arch 459:73–80 Kojima T, Yamamoto T, Murata M et al, 2003, Regulation of the blood-biliary barrier: interaction between gap and tight junctions in hepatocytes. Med Electron Microsc 36:157–164 Meertens L, Bertaux C, Cukierman L et al, 2008, The tight junction proteins claudin-1, -6, and -9 are entry cofactors for hepatitis C virus. J Virol 82:3555–3560 Zheng A, Yuan F, Li Yet al, 2007, Claudin-6 and claudin-9 function as additional coreceptors for hepatitis C virus. J Virol 81:12465–12471 French AD, Fiori JL, Camilli TC et al, 2009, PKC and PKA phosphorylation affect the subcellular localization of claudin-1 in melanoma cells. Int J Med Sci 6:93–101 Terry S, NieM,MatterKet al, 2010, Rho signaling and tight junction functions. Physiology, Bethesda, 25:16–26 Gonzalez-Mariscal L, Lechuga S, Garay E, 2007, Role of tight junctions in cell proliferation and cancer. Prog Histochem Cytochem 42:1–57 Lal-Nag M, Morin PJ, 2009, The claudins. Genome Biol 10:235 de Oliveira SS, de Oliveira IM, De Souza W et al, 2005, Claudins upregulation in human colorectal cancer. FEBS Lett 579:6179–6185 Yoshida T, Kinugasa T, Akagi Yet al, 2011, Decreased expression of claudin-1 in rectal cancer: a factor for recurrence and poor prognosis. Anticancer Res 31:2517–2525 ShinHI, KimBH, ChangHS et al, 2011, Expression of claudin-1 and -7 in clear cell renal cell carcinoma and its clinical significance. Korean J Urol 52:317–322 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 755 EP 764 DI 10.1007/s12253-014-9758-x PG 10 ER PT J AU Michalopoulos, N Papavramidis, STh Karayannopoulou, G Pliakos, I Papavramidis, TS Kanellos, I AF Michalopoulos, Nickos Papavramidis, S Theodossis Karayannopoulou, Georgia Pliakos, Ioannis Papavramidis, T Spiros Kanellos, Ioannis TI Neuroendocrine Tumors of Extrahepatic Biliary Tract SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Neuroendocrine tumor; Carcinoid; Biliary neoplasm; Klatskin tumor; Extrahepatic biliary duct obstruction ID Neuroendocrine tumor; Carcinoid; Biliary neoplasm; Klatskin tumor; Extrahepatic biliary duct obstruction AB Neuroendocrine tumors of the extrahepatic bile ducts (EBNETs) are very rare. The aim of the present review is to elucidate the characteristics of EBNETs, their treatment and prognosis. An exhaustive systematic review of the literature was performed from 1959 up-to-date. One hundred articles, describing 150 cases were collected. Each article was carefully analyzed and a database was created. The most common symptoms were jaundice (60.3 %) and pruritus (19.2 %). Cholelithiasis co-existed in 15 cases (19.2 %). Hormone- and vasoactive peptide- related symptoms were present in only 7 cases (9 %). The most frequent sites were found to be the common hepatic duct and the proximal common bile duct (19.2 %). Surgical management was considered the main treatment for EBNETs, while excision of extrahepatic biliary tree (62.82 %) with portal vein lymphadenectomy (43.6 %) was the most popular procedure. EBNETs are extremely rare. Their rarity makes their characterization particularly difficult. Up to date the final diagnosis is made after surgery by pathology and immunohistochemistry findings. The present analysis of the existing published cases elucidates many aspects of these tumours, giving complete clinicopathological documentation. C1 [Michalopoulos, Nickos] Aristotle University of Thessaloniki, AHEPA University Hospital, 3rd Department of Surgery, 85 Karakasi, 54453 Thessaloniki, Greece. [Papavramidis, S Theodossis] Aristotle University of Thessaloniki, AHEPA University Hospital, 3rd Department of Surgery, 85 Karakasi, 54453 Thessaloniki, Greece. [Karayannopoulou, Georgia] Aristotle University of Thessaloniki, AHEPA University Hospital, Pathology DepartmentThessaloniki, Greece. [Pliakos, Ioannis] Aristotle University of Thessaloniki, AHEPA University Hospital, 3rd Department of Surgery, 85 Karakasi, 54453 Thessaloniki, Greece. [Papavramidis, T Spiros] Aristotle University of Thessaloniki, AHEPA University Hospital, 3rd Department of Surgery, 85 Karakasi, 54453 Thessaloniki, Greece. [Kanellos, Ioannis] Aristotle University of Thessaloniki, AHEPA University Hospital, 3rd Department of Surgery, 85 Karakasi, 54453 Thessaloniki, Greece. RP Michalopoulos, N (reprint author), Aristotle University of Thessaloniki, AHEPA University Hospital, 3rd Department of Surgery, 54453 Thessaloniki, Greece. EM nickos.michalopoulos@gmail.com CR Are C, Gonen M, D’Angelina M, RP D m, Fong Y, Blumgart LH, 2006, Differential diagnosis of proximal biliary obstruction. Surgery 140:756–763 Modlin IM, Shapiro MD, Kidd M, 2005, An analysis of rare carcinoid tumors: Clarifying these clinical conundrums. World J Surg 29:92–101 Modlin IM, Sandor A, 1997, An analysis of 8305 cases of carcinoids tumors. Cancer 79:813–819 Capella C, Solcia E, Sobin LH, Arnold R, 2000, Endocrine tumours of the gallbladder and extrahepatic bile ducts. In: Hamilton R, Aaltonen LA, eds, Pathology and Genetics of Tumours of the Digestive System. WHO Classification of Tumours. IARC Press, Lyon, pp 214–266 Davies AJ, 1959, Carcinoidtumors, argentaffinomata). Ann R CollSurgEngl 25:277–280 Ferrone CR, Tang LH, D’AngelicaM, DeMatteo RP, Blumgart LH, Klimstra DS et al, 2007, Extrahepatic bile duct carcinoid tumors: malignant biliary obstruction with a good prognosis. J AmCollSurg 205(2):357–61 Sethi H, Madanur M, Srinivasan P, Portmann B, Heaton N, Rela M, 2007, Non-functioning well-differentiated neuroendocrine tumor of the extrahepat i c bi l e duct: an unusual suspect? HepatobiliaryPancreat Dis Int 6(5):549–52 Pilz E, 1961, Ubereinkarzinoid des ductuscholedochus, in German). ZentralblChir 86:1588–1590 Little JM, Gibson AA, Kay AW, 1968, Primary common bile-duct carcinoid. Br J Surg 55:147–149 Godwin JD, 1975, Carcinoid tumours. An analysis of 2,837 cases. Cancer 36:560–569 Bergdahl L, 1976, Carcinoid tumours of the biliary tract. Aust N Z J Surg 46:136–138 Judge DM, Dickman PS, Trapukdi BS, 1976, Nonfunctioning argyrophilictumour, APUDoma, of the hepatic duct: simplified methods of detecting biogenic amines in tissue. Am J PatholClin 66:40–45 Schwesinger G, 1978, Carcinoid tumor of the bile ducts, in German). Z ArztlFortbild, Jena, 72(16):796–8 Gerlock AJ Jr, Muhletaler CA, 1979, Primary common bile duct carcinoid. GastrointestRadiol 4:263–264 Sbaffi E, Panucci A, Reale R, Persia A, Gucciardo G, 1980, A case of hepato-biliary carcinoid, in Italian). Chir Ital 32(4):896–902 Machado MC, Penteado S, Bacchella T, Betallero A, da Cunha JE, Faintuch J, 1981, Carcinoid tumor, apudoma, of the junction of the main hepatic ducts. Rev HospClinFac Med Sao Paulo 36:139–142 Vitaux J, Salmon RJ, Languille O, Buffet C, Martin E, Chaput JC, 1981, Carcinoid tumor of the common bile duct. Am J Gastroenterol 76:360–362 Abe V, Itoh H, Yoshida M, 1983, Carcinoid tumor of the common bile duct: report of a case. Stomach Intestine 18:543–548 Steenbergen VW, Fevery J, Vanstabel J, Geboes K, Ponette E, Kerremans R, 1983, Case report: Fourteen-year follow-up an apudoma of the bile ducts at the hilum of the liver. Gastroenterol 1585–1591 Goodman ZD, Albores-Saavedra J, Lundblad DM, 1984, Somatostatinoma of the cystic duct. Cancer 53:498–502 Jutte DL, Bell RH Jr, Penn I, Powers J, Kolinjivadi J, 1986, Carcinoid tumor of the biliary system. Case report and literature review. Dig Dis Sci 32:763–769 Alexander IA, Thomson KR, Kune GA, 1986, Primary common bile duct carcinoid: demonstration by computerised tomography, ultrasonography and angiography. AustralasRadiol 30:34–37 Badowski A, Sosnicki W, Dumanski Z, Kaszynski H, 1986, Carcinoid tumor of the common bile duct, Article in Polish). WiadLek 39(10):694–9 Nicolescu PG, Popescu A, 1986, Carcinoid tumor of the cystic duct, in Rumanian). MorpholEmbryol, Bucur, 32:275–277 Gastinger I, Schutze U, Beetz G, Lippert H, 1987, Obstructive jaundice caused by a carcinoid tumor of the hepatocholedochal duct, in German). ZentralblChir 112(18):1170–5 Bickerstaff DR, RossWB, 1987, Carcinoid of the biliary tree: a case report and review of the literature. J R CollSurgEdinb 32:48–51 AnezMS, Zabala Nava R, Zabaleta P, Villegas SR, 1987, Carcinoid tumor in aberrant pancreas of the choledochus, in Spanish). GEN 41(3):108–112 ReinhardtD, ThieleH,Weber JC,HofmannWJ, 1988, Carcinoid of the ductuscholedochus. A rare cause of obstructive jaundice, in German). Chirurg 59:683–687 Fujita N, Mochizuki F, Lee S, Satoh K, Kobayashi G, Yano A, 1989, Carcinoid tumour of the bile duct: case report. GastrointestRadiol 14:151–154 Chittal SM, Ra PM, 1989, Carcinoid of the cystic duct. Histopathology 15:643–646 Yoshida N, Ueno Y, Sumino Y,Wakabayashi M, Atobe T, 1989, A case of carcinoid tumor of the common hepatic duct, in Japanese). Nippon ShokGakkaiZasshi 86(11):2617–21 Van der Wal AC, Van Leeuwen DJ, Walford N, 1989, Small cell neuroendocrine, oat cell, tumour of the common bile duct. Histopathology 16:398–400 Wrazildo W, Gamroth A, Hofmann WJ, Koch K, 1989, PrimaresKarzinoid des ductuccholedochus. Radiologe 29:191–194 Bumin C, Ormeci N, DolapciM, Gungor S, 1990, Carcinoid tumor of the biliary duct. IntSurg 75:262–264 Fellows IW, Leach IH, Smith PG, Toghill PJ, Doran J, 1990, Carcinoid tumour of the common bile duct–a novel complication of von Hippel-Lindau syndrome. Gut 31(6):728–9 Carle JP, Tarasco A, Daude M, Nayraud P, Gislon J, 1990, Carcinoid tumor of the common bile duct in a 13-year-old girl. Presse Med 19(42):1946–7 BrownWM, Henderson JM, Kennedy JC, 1990, Carcinoid tumour of the bile duct. A case report and literature review. Am Surg 56: 343–346 Hsu W, Deziel DJ, Gould VE, Warren WH, Gooch GT, Staren ED, 1991, Neuroendocrine differentiation and prognosis of extrahepatic biliary tract carcinoma. Surgery 110:604–610 Barron-Rodriguez LP, Manivel JC, Mendez-Sanchez N, Jessurun J, 1991, Carcinoid tumor of the common bile duct: evidence for its origin in metaplastic endocrine cells. Am J Gastroenterol 86:1073– 1076 Besznyak I, Tasnadi G, Hirsch T, Koves I, Schaff Z, Lapis K, 1991, Hepato-lobectomy for carcinoid tumor of the hepatic duct, in Hungarian with English abstract). OrvHetil 132:591–594 Angeles-Angeles A, Quintanilla-Martinez L, Larriva-Sahd J, 1991, Primary carcinoid tumor of the common bile duct: Immunohistochemical characterization of a case a review of the literature. Am J Clin Pathol 96:341–344 Newman K, Vates T, Duffy L, Anderson K, 1992, Pancreatoduodenectomy with preservation of the stomach and pylorus: a safe and effective alternative in children. J PediatrSurg 27:1334–1335 Dixon J, Lisehora G, Lee MY, 1992, Carcinoid tumour of the common bile duct. ContempSurg 41:37–40 Ueyama T, Ding J, Hashimoto H, Tsuneyoshi M, Enjoji M, 1992, Carcinoid tumor arising in the wall of a congenital bile duct cyst. Arch Pathol Lab Med 166:291–293 Rugge M, Sonego F,Militello C, Guido M, Ninfo V, 1992, Primary carcinoid tumor of the cystic and common bile ducts. Am J SurgPathol 16:802–807 Gembala RS, Friedman AC, Friedman AC, Radecki PD, Ball DS, Hartman GG et al, 1993, Carcinoid of the intrahepatic ducts. Abdom Imaging 18:242–244 Sankary HN, Foster P, Frye E, Williams JW, 1995, Carcinoid tumours of the extrahepatic bile duct: an unusual cause of bile duct obstruction. Liver TransplSurg 1:122–123 Mandujano-Vera G, Angeles-Angeles A, 1995, Cruz-HernadezJ, Sansores-Perez M, Larriva-Sahd J. Gastrinoma of the common bile duct: Immunohistochemical and ultrastructural study of a case. J ClinGastroenterol 20:321–324 Belli G, Rotondano G, D’Agostino A, Marano I, 1996, Primary extrahepatic bile duct carcinoids. HPB Surg 9:101–105 Kopelman D, ScheinM, Kerner H, Bahuss H, HashmonaiM(1996, Carcinoid tumor of the common bile duct. HPB Surg 10:41–43 Meyer CN, Gersdsen J, Christensen N, 1997, Limited surgical excision of a localized carcinoid tumor of the cystic duct. DigSurg 14:423–425 Hao L, Friedman AL, Navarro VJ, West B, Robert ME, 1996, Carcinoid tumor of the common bile duct producing gastrin and serotonin. J ClinGastroenterol 23:63–65 Nahas SC, Lourencao JL, 1998, GazoniE, MottCde B, Borba MR, Pinto Filho JOet al. Tumour carcinoide de ducto biliar comum: relato de um caso, in Portuguese with English abstract). Rev Hosp Clin Fa Med Sao Paulo 53:26–28 Shah IA, Schlageter MO, Wong SX, Gani OS, 1998, Carcinoid tumor of the cystic duct. Dig Surg 15:372–373 Bembenek A, Lotterer E, Machens A, Cario H, Krause U, Holzhausen H, 1998, Neuroendocrine tumor of the common hepatic duct: a rare case of extrahepatic jaundice in adolescence. Surgery 123:712 Oikawa I, Hirata K, 1998, KatsuramakiT, Mukaiya M, Sasaki K, Satoh M. Neuroendocrine carcinoma of extrahepatic biliary tract with positive immunostaining for gastrin-releasing peptide: report of a case. Surg Today 28:1192–1195 Aronsky D, Z’graggen K, Stauffer E, Lange J, Klaiber C, 1999, Primary Neuroendocrine Tumors of the Cystic Duct. Digestion 60: 493–496 MartignoniME, Friess H, 1999, LubkeD, UhlW,Maurer C,Muller M et al. Study of a Primary Gastrinoma in the Common Hepatic Duct – A Case Report. Digestion 60:187–190 Ross AC, Hurley JB, Hay WB, Rusnak CH, Petrunia DM, 1999, Carcinoids of the common bile duct: a case report and literature review. Can J Surg 42:59–63 Perakath B, Chandran BS, Srivastava VM, Nair A, Sitaram V, 1999, Carcinoid tumor of common hepatic duct. Indian J Gastroenterol 18:127 Hermina M, Starling J, Warner TF, 1999, Carcinoid tumor of the cystic duct. Pathol Res Pract 195:707–710 Chamberlain RS, Blumgart LH, 1999, Carcinoid tumors of the extrahepatic bile duct. A rare cause malignant biliary obstruction. Cancer 86:1956–1965 Chan C, Medina-Franco H, Bell W, Lazenby A, Vickers S, 2000, Carcinoid tumor of the hepatic duct presenting as a Klatskin tumor i n an adolescen t and review of worl d l i te ra t u re . Hepatogastroenterology 47:519–521 Lombardi A, Batignani G, Nesi G, 2000, Primary carcinoid tumour of the common bile duct. HBP 2:95–100 Juturi JV, Maghfoor I, Doll DC, Evans ML, 2000, A case of biliary carcinoid presenting with pancreatitis and obstructive jaundice. Am J Gastroenterol 95:2973–2974 Maitra A, Krueger JE, Tascilar M, Offerhaus GJ, Angeles-Angeles A, Klimstra DS et al, 2000, Carcinoid tumors of the extrahepatic bile ducts: a study of seven cases. Am J SurgPathol 24:1501–1510 Jensen SI, Pless T, Mortensen MB, 2001, Carcinoid tumor of the choledochus mimicking a bileduct stone during endoscopic ultrasonography and endoscopic retrograde cholangiography. Endoscopy 33:100 Turrion VS, Salas C, Alvira LG, Jimenez M, Lucena JL, Ardaiz J, 2002, Carcinoid tumour of the common bile duct: an exceptional indication for liver transplantation. Transplant Proc 34:264 Pawlik TM, Shah S, Eckhauser FE, 2003, Carcinoid tumor of the biliary tract: treating a rare cause of duct obstruction. Am Surg 69: 98–101 Podnos YD, Jimenez JC, Zainabadi K, Ji P, Cooke J, Bussutil RW et al, 2003, Carcinoid tumors of the common bile duct: report of two cases. Surg Today 33:553–555 Volpe CM, Pryor JP, Caty M, Doerr RJ, 2003, An adolescent with bile duct carcinoid tumor. Med Pediatric Oncol 40(2):137–138 El Rass ZS,Mohsine RM, Berger F, Thierry P, Partensky CC, 2004, Endocrine tumors of the extrahepatic bile ducts. Pathological and clinical aspects, surgical management and outcome. Hepatogastrenterology 51:1295–1300 Menezes AA, Diver AJ, McCance D, Diamond T, 2004, Carcinoid tumour of the extrahepatic bile duct–report of a case and literature review. Ulster Med J 73(1):59–62 Ligato S, Furmaga W, Cartun RW, Hull D, Tsongalis GJ, 2005, Primary carcinoid tumor of the common hepatic duct: a rare case with immunohistochemical and molecular findings. OncolRep 13: 543–546 Nesi G, Lombarti A, Batignani G, Ficari F, Rubio CA, Tonelli F, 2006, Well-differentiated endocrine tumor of the distal common bile duct: a case study and literature review. Virchows Arch 449: 104–111 Pithawala M, Mittal G, 2005, PrabhuRY, Kantharia CV, Joshi A. Supe A Carcinoid tumor of bile duct Indian J Gastroenterol 24(6): 262–3 Hubert C, Sempoux C, Berquin A, Deprez F, Jamar F, Giqot JF, 2005, Bile duct carcinoids tumors: an uncommon disease but with a good prognosis? Hepato-Gastroenterology 52:1042–1047 Tzimas GN, Vali K, Deschenes M, Marcus VA, Barkun JS, Tchervenkov JI et al, 2006, Liver transplantation for metastases from a bile duct carcinoid. HPB 8:67–68 Kim DH, SongMH, Kim DH, 2006)Malignant carcinoid tumor of the common bile duct: report of a case. Surg Today 36(5):485–9 Caglikulekci M, Dirlik M, Aydin O, Ozer C, Colak T, Daq A et al, 2006, Carcinoid tmour of the common bile duct: report of a case and a review of the literature. Acta ChirBelg 106(1):112–5 Honda H, Hayashi S, Sekiguchi Y, Tsukadaira T, Nakamura K, 2006, A case of the extrahepatic bile duct carcinoid tumor, in Japanese with English abstract). Nippon ShokakibyoGakkaiZasshi 103(10):1169–75 John BM, SongMH, Park YS, Jo YJ, Kim SH, Lee HH et al, 2006, A case of carcinoid tumor of the common bile duct, in Korean with English abstract). Korean J Gastroenterol 47(4):320–3 Todoroki T, Sano T, Yamada S, Hirahara N, Toda N, Tsukada K et al, 2007, Clear cell carcinoid tumor of the distal common bile duct.W J SurgOnc 17:5–6 Colombo F, Carrara B, Rosato S, Ruck F, Simonetti A, Soccio M, 2007, Carcinoid tumours of the bile ducts, in Italian with English abstract). ChirItal 59(3):405–10 Stavridi F, Chong H, Chan S, Goldsmith C, Reddy M, Glees J, Benepal T, 2007, Neuroendocrine tumour of the cystic duct: a case report and literature review. J Gastrointest Cancer 38(1):32–3 Nafidi O, Nguyen BN, Roy A, 2008, Carcinoid tumor of the common bile duct: A rare complication of vonHippel-Lindau syndrome. W J Gastroenterol 14(8):1299–1301 Schmitt TM, Bonatti H, Hagspiel KD, Iezzoni J, Northup P, Pruett TL, 2008, Carcinoid of bile duct difurcation. J Am Coll Surgeons 206(2):399 Gusani NJ, Marsh JW, Nalesnik MA, Tublin ME, Gamblin TC, 2008, Carcinoid of the extra-hepatic bile duct: a case report with long-term follow up and review of literature. The Am Surgeon 74: 87–91 Ferekouras E, Petrou A, Bramis K, Prassas E, Papaconstantinou I, Dimitriou N et al, 2009, Malignant carcinoid tumor of the cystic duct: a rare cause of bile duct obstruction. HepatobiliaryPancreat Dis Int 8(6):640–645 Albores-Saavedra J, Batich K, Hossain S, Henson DE, Schwartz AM, 2009, Carcinoid tumors and small cell carcinimas of the gallbladder and extrahepatic bile ducts: a comparative study based on 221 cases from the surveillance, epidimiology and end results program. Annals of diagnostic pathology 13:378–383 Tonnhofer U, Balassy C, Reck CA, Koller A, Horcher E, 2009, Neuroendocrine tumor of the common hepatic duct, mimicking a choledochal cyst in a 6-year-old child. J PediartSurg 44:22–25 Price TN, Thompson GB, Lewis JT, Lioyd RV, Young WF, 2009, Zollinger-Ellison syndrome due to primary gastrinoma of the extrahepatic biliary tree: three case reports and review of the literature. EndocrPract 15(7):737–739 Malecki EA, Acosta R, Twaddell W, Heller T, Manning MA, Darwin P, 2009, Endoscopic diagnosis of a biliary neuroendocrine tumor. Gastrointest Endoscopy 70(6):1275–1276 Noronha YS, Raza AS, 2010, Well-differentiated neuroendocrine, carcinoid, tumors of the extrahepatic biliary ducts. Arch Pathol Lab Med 134(7):1075–9 Tsalis K, Vrakas G, Geroukis T, Cheva A, Roidos GN, Lazarides C, 2010, Primary neuroendocrine tumor of the extrahepatic biliary tree mimicking Klatskin tumor. J Gastrointestin Liver Dis 19(3): 341–2 Lee JH, Lee KG, Oh YH, Paik SS, Park HK, Lee KS, 2011, Carcinoid tumors of the extrahepatic biliary tract: report of four cases. Surg Today 41(3):430–5 Kim J, LeeWJ, Lee SH, Lee KB, Ryu JK, Kim YT, Kim SW, Yoon YB, Hwang JH, Han HS,Woo SM, Park SJ, 2011, Clinical features of 20 patients with curatively resected biliary neuroendocrine tumours. Dig Liver Dis 43(12):965–70 Squillaci S, Marchione R, Piccolomini M, Colombo F, Bucci F, Bruno M, Bisceglia M, 2010, Well-differentiated neuroendocrine carcinoma, malignant carcinoid, of the extrahepatic biliary tract: report of two cases and literature review. APMIS 118(8):543–56 Zhan J, Bao G, Hu X, GaoW, Ruo X, Gong J, Zhu Q, Liu Y, 2010, Carcinoid tumor of the common bile duct in children: a case report. J Pediatr Surg 45(10):2061–3 Song LY, Hu XL, Zhao LS, Ning PR, 2011, Carcinoid tumor of common bile duct: report of a case in pediatric patient. HonghuaBingLiXueZaZhi 40(1):54–5 Athanasopoulos PG, Arkadopoulos N, Stafyla V, Tympa A, Kairi E, Ryzman-Louloudis C, Smyrniotis V, 2011, A rare combination of an endocrine tumour of the common bile duct and a follicular lymphoma of the ampulla of Vater: a case report and review of the literature. World J SurgOncol 14(9):4 Cappell MS, Killeen TC, Jury R, 2011, Common bile duct carcinoid mimicking the clinical, EUS, and ERCP findings of cholangiocarcinoma: a rare but potentially curable cause of obstructive jaundice. ClinGastroenterolHepatol 9(11):112–3 Ioannidis O, Cheva A, Paraskevas G, Chatzopoulos S, Kotronis A, Papadimitriou N, Konstantara A, Makrantonakis A, Kakoutis E, 2012, Neuroendocrine tumor of the cystic duct. Acta Gastroenterol Belg 75(3):357–60 Bhalla P, Powle V, Shah RC, Jagannath P, 2012, Neuroendocrine tumor of common hepatic duct. Indian J Gastroenterol 31(3):144–146 Rosai J, 2011, The origin of neuroendocrine tumors and the neural crest saga. Mod Pathol 24:S53–7 Fontaine J, Le Douarin NM(1977, Analysis of endoderm formation in the avian blastoderm by the use of quailchick chimaeras. The problem of the neurectodermal origin of the cells of the APUD series. J Embryol Exp Morphol 41:209–222 Modlin IM, Lye KD, KiddM(2003, A 5-decade analysis of 13,715 carcinoid tumors. Cancer 97:934–959 Kloeppel G, Heintz PU, 1996, CapellaC, Solera E. Pathology and nomenaclature of human gastrointestinal neuroendocrine, carcinoid, tumors and related lesions. W J Surg 20:132–141 Rindi G, Arnold R, Bosman FT, Capella C, Klimstra DS, 2010, Klo¨ppel G et al. Nomenclature and classification of neuroendocrineneoplasms of the digestive system. In: Bosman FT, Carneiro F, Hruban RH, Theise ND, eds, WHO Classification ofTumors of the Digestive System, 4th edn. International Agency for Research on Cancer, Lyon, pp 13–14 Walter T, Brixi-Benmansour H, Lombard-Bohas C, CadiotG, 2012, New treatment strategies in advanced neuroendocrine tumours. Dig Liver Dis 44(2):95–105 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 765 EP 775 DI 10.1007/s12253-014-9808-4 PG 11 ER PT J AU Eder, K Kalman, B AF Eder, Katalin Kalman, Bernadette TI Molecular Heterogeneity of Glioblastoma and its Clinical Relevance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Glioblastoma; Inter-tumor heterogeneity; Intra-tumor heterogeneity; Molecular classification; Molecular heterogeneity; Primary brain tumor ID Glioblastoma; Inter-tumor heterogeneity; Intra-tumor heterogeneity; Molecular classification; Molecular heterogeneity; Primary brain tumor AB Glioblastoma is the most common intracranial malignancy and constitutes about 50%of all gliomas. Both intertumor and intra-tumor histological heterogeneity had been recognized by the early 1980-ies. Recent works using novel molecular platforms provided molecular definitions of these tumors. Based on comprehensive genomic sequence analyses, The Cancer Genome Atlas Research Network (TCGA) cataloged somatic mutations and recurrent copy number alterations in glioblastoma. Robust transcriptome and epigenome studies also revealed inter-tumor heterogeneity. Integration and cluster analyses of multi-dimensional genomic data lead to a new classification of glioblastoma tumors into subtypes with distinct biological features and clinical correlates. However, multiple observations also revealed tumor area-specific patterns of genomic imbalance. In addition, genetic alterations have been identified that were common to all areas analyzed and other alterations that were area specific. Analyses of intratumor transcriptome variations revealed that in more than half of the examined cases, fragments from the same tumor mass could be classified into at least two different glioblastoma molecular subgroups. Intra-tumor heterogeneity of molecular genetic profiles in glioblastoma may explain the difficulties encountered in the validation of oncologic biomarkers, and contribute to a biased selection of patients for single target therapies, treatment failure or drug resistance. In this paper, we summarize the currently available literature concerning interand intra-tumor molecular heterogeneity of glioblastomas, and call attention to the importance of this topic in relation to the growing efforts in routine molecular diagnostics and personalized therapy. C1 [Eder, Katalin] Vas County Markusovszky Hospital, Markusovszky Street 5, 9700 Szombathely, Hungary. [Kalman, Bernadette] Vas County Markusovszky Hospital, Markusovszky Street 5, 9700 Szombathely, Hungary. RP Eder, K (reprint author), Vas County Markusovszky Hospital, 9700 Szombathely, Hungary. EM ederkati@gmail.com CR Dolecek TA, Propp JM, Stroup NE, Kruchko C, 2012, CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005–2009. Neuro-Oncol 14(suppl 5):v1–v49 Olar A, Aldape KD, 2014, Using the molecular classification of glioblastoma to inform personalized treatment. J Pathol 232(2): 165–177 Burger PC, Green SB, 1987, Patient age, histologic features, and length of survival in patients with glioblastoma multiforme. Cancer 59(9):1617–1625 Shapiro JR, YungWK, ShapiroWR, 1981, Isolation, karyotype, and clonal growth of heterogeneous subpopulations of human malignant gliomas. Cancer Res 41(6):2349–2359 Loeper S, Romeike B, Heckmann N, Jung V, Henn W, Feiden W, Zang K, Urbschat S, 2001, Frequent mitotic errors in tumor cells of genetically micro-heterogeneous glioblastomas. Cytogenet Genome Res 94(1–2):1–8 Coons SW, Johnson PC, Shapiro JR, 1995, Cytogenetic and flow cytometry DNA analysis of regional heterogeneity in a low grade human glioma. Cancer Res 55(7):1569–1577 Misra A, Chattopadhyay P, Dinda AK, Sarkar C, Mahapatra AK, Hasnain SE, Sinha S, 2000, Extensive intra-tumor heterogeneity in primary human glial tumors as a result of locus non-specific genomic alterations. J Neuro-Oncol 48(1):1–12 Harada K, Nishizaki T, Ozaki S, Kubota H, Ito H, Sasaki K, 1998, Intratumoral cytogenetic heterogeneity detected by comparative genomic hybridization and laser scanning cytometry in human gliomas. Cancer Res 58(20):4694–4700 Walker C, du Plessis DG, Joyce KA, Machell Y, Thomson-Hehir J, Al Haddad SA, Broome JC, Warnke PC, 2003, Phenotype versus genotype in gliomas displaying inter-or intratumoral histological heterogeneity. Clin Cancer Res 9(13):4841–4851 Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, Hahn WC, Ligon KL, Louis DN, Brennan C, 2007, Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev 21(21):2683–2710 McLendon R, Friedman A, Bigner D, Van Meir EG, Brat DJ, Mastrogianakis GM, Olson JJ, Mikkelsen T, Lehman N, Aldape K, 2008, Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nat 455(7216):1061–1068 Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL, Olivi A, McLendon R, Rasheed BA, Keir S, Nikolskaya T, Nikolsky Y, Busam DA, Tekleab H, Diaz LA Jr, Hartigan J, Smith DR, Strausberg RL, Marie SK, Shinjo SM, Yan H, Riggins GJ, Bigner DD, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW, 2008, An integrated genomic analysis of human glioblastoma multiforme. Sci 321(5897):1807–1812., DOI 10.1126/ science.1164382 Verhaak RG, Hoadley KA, Purdom E,Wang V, Qi Y,WilkersonMD, Miller CR, Ding L, Golub T, Mesirov JP, Alexe G, Lawrence M, O’Kelly M, Tamayo P, Weir BA, Gabriel S, Winckler W, Gupta S, Jakkula L, Feiler HS, Hodgson JG, James CD, Sarkaria JN, Brennan C, Kahn A, Spellman PT,Wilson RK, Speed TP, Gray JW,Meyerson M, Getz G, Perou CM, Hayes DN, 2010, Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17(1):98–110., DOI 10.1016/j.ccr.2009.12.020 Masica DL, Karchin R, 2011, Correlation of somatic mutation and expression identifies genes important in human glioblastoma progression and survival. Cancer Res 71(13):4550–4561 Cerami E, Demir E, Schultz N, Taylor BS, Sander C, 2010, Automated network analysis identifies core pathways in glioblastoma. PLoS One 5(2):e8918 Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, Davies H, Teague J, Butler A, Stevens C, 2007, Patterns of somatic mutation in human cancer genomes. Nat 446(7132):153–158 Stratton MR, Campbell PJ, Futreal PA, 2009, The cancer genome. Nat 458(7239):719–724 Ahn J-Y, Hu Y, Kroll TG, Allard P, Ye K, 2004, PIKE-A is amplified in human cancers and prevents apoptosis by up-regulating Akt. Proc Natl Acad Sci U S A 101(18):6993–6998 Qi Q, He K, Liu X, Pham C, Meyerkord C, Fu H, Ye K, 2013, Disrupting the PIKE-A/Akt interaction inhibits glioblastoma cell survival, migration, invasion and colony formation. Oncog 32(8): 1030–1040 Frattini V, Trifonov V, Chan JM, Castano A, Lia M, Abate F, Keir ST, Ji AX, Zoppoli P, Niola F, 2013, The integrated landscape of driver genomic alterations in glioblastoma. Nat Genet 45(10):1141–1149 Pugh TJ, Weeraratne SD, Archer TC, Krummel DAP, Auclair D, Bochicchio J, Carneiro MO, Carter SL, Cibulskis K, Erlich RL, 2012, Medulloblastoma exome sequencing uncovers subtypespecific somatic mutations. Nat 488(7409):106–110 Zhang J, Benavente CA, McEvoy J, Flores-Otero J, Ding L, Chen X, Ulyanov A,Wu G,WilsonM,Wang J, 2012, A novel retinoblastoma therapy from genomic and epigenetic analyses. Nat 481(7381):329– 334 Network CGAR, 2013, Integrated genomic characterization of endometrial carcinoma. Nat 497(7447):67–73 FreijeWA, Castro-Vargas FE, Fang Z, Horvath S, Cloughesy T, Liau LM, Mischel PS, Nelson SF, 2004, Gene expression profiling of gliomas strongly predicts survival. Cancer Res 64(18):6503–6510 LiangY, DiehnM,WatsonN, BollenAW, AldapeKD, NicholasMK, Lamborn KR, Berger MS, Botstein D, Brown PO, 2005, Gene expression profiling reveals molecularly and clinically distinct subtypes of glioblastomamultiforme. Proc Natl Acad Sci U S A 102(16): 5814–5819 Phillips HS, Kharbanda S, Chen R, ForrestWF, Soriano RH,Wu TD, Misra A, Nigro JM, Colman H, Soroceanu L, 2006, Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 9(3):157–173 Kong J, Cooper LA, Wang F, Gutman DA, Gao J, Chisolm C, Sharma A, Pan T, Van Meir EG, Kurc TM, 2011, Integrative, multimodal analysis of glioblastoma using TCGA molecular data, pathology images, and clinical outcomes. Biomed Eng, IEEE Trans on 58(12):3469–3474 Kim Y-W, Koul D, Kim SH, Lucio-Eterovic AK, Freire PR, Yao J, Wang J, Almeida JS, Aldape K, Yung WA, 2013, Identification of prognostic gene signatures of glioblastoma: a study based on TCGA data analysis. Neuro-Oncol 15(7):829–839 Doucette T, Rao G, Rao A, Shen L, Aldape K,Wei J, Dziurzynski K, Gilbert M, Heimberger AB, 2013, Immune heterogeneity of glioblastoma subtypes: extrapolation from the cancer genome atlas. Cancer Immunol Res 1(2):112–122 Jones PA, Baylin SB, 2007, The epigenomics of cancer. Cell 128(4): 683–692 Noushmehr H, Weisenberger DJ, Diefes K, Phillips HS, Pujara K, Berman BP, Pan F, Pelloski CE, Sulman EP, Bhat KP, Verhaak RG, Hoadley KA, Hayes DN, Perou CM, Schmidt HK, Ding L, Wilson RK, Van Den Berg D, Shen H, Bengtsson H, Neuvial P, Cope LM, Buckley J, Herman JG, Baylin SB, Laird PW, Aldape K, 2010, Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17(5):510–522., DOI 10. 1016/j.ccr.2010.03.017 Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, ChakravartyD, Sanborn JZ, Berman SH(2013, The somatic genomic landscape of glioblastoma. Cell 155(2):462– 477 Li B, Senbabaoglu Y, Peng W, Yang M-L, Xu J, Li JZ, 2012, Genomic estimates of aneuploid content in glioblastoma multiforme and improved classification. Clin Cancer Res 18(20):5595–5605 Iorio MV, Croce CM, 2012, MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med 4(3):143–159 Kim T-M, Huang W, Park R, Park PJ, Johnson MD, 2011, A developmental taxonomy of glioblastoma defined and maintained by MicroRNAs. Cancer Res 71(9):3387–3399 Li R, Gao K, Luo H,Wang X, Shi Y, Dong Q, LuanW, You Y, 2014, Identification of intrinsic subtype-specific prognostic microRNAs in primary glioblastoma. J Exp Clin Cancer Res 33(1):9 Godlewski J, Newton H, Chiocca E, Lawler S, 2010, MicroRNAs and glioblastoma; the stem cell connection. Cell Death Differ 17(2): 221–228 Hermansen SK, Kristensen BW, 2013, MicroRNA biomarkers in glioblastoma. J Neuro-Oncol 114(1):13–23 Dirks PB, 2010, Brain tumor stem cells: the cancer stem cell hypothesis writ large. Mol Oncol 4(5):420–430 Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB, 2004, Identification of human brain tumour initiating cells. Nat 432(7015):396–401 Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, Vescovi A, 2004, Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res 64(19):7011–7021 Lottaz C, Beier D, Meyer K, Kumar P, Hermann A, Schwarz J, Junker M, Oefner PJ, Bogdahn U, Wischhusen J, 2010, Transcriptional profiles of CD133+ and CD133− glioblastomaderived cancer stem cell lines suggest different cells of origin. Cancer Res 70(5):2030–2040 De Bacco F, Casanova E, Medico E, Pellegatta S, Orzan F, Albano R, Luraghi P, Reato G, D’Ambrosio A, Porrati P, 2012, The MET oncogene is a functional marker of a glioblastoma stem cell subtype. Cancer Res 72(17):4537–4550 Weller M, Stupp R, Hegi ME, van den BentM, Tonn JC, Sanson M, WickW, Reifenberger G, 2012, Personalized care in neuro-oncology coming of age: why we need MGMT and 1p/19q testing for malignant glioma patients in clinical practice. Neuro-Oncol 14(suppl 4): iv100–iv108 Marko NF, Quackenbush J, Weil RJ, 2011, Why is there a lack of consensus on molecular subgroups of glioblastoma? Understanding the nature of biological and statistical variability in glioblastoma expression data. PLoS One 6(7):e20826 Nobusawa S, Lachuer J,Wierinckx A, Kim YH, Huang J, Legras C, Kleihues P, Ohgaki H, 2010, Intratumoral patterns of genomic imbalance in glioblastomas. Brain Pathol 20(5):936–944., DOI 10.1111/j. 1750-3639.2010.00395.x Snuderl M, Fazlollahi L, Le LP, Nitta M, Zhelyazkova BH, Davidson CJ, Akhavanfard S, Cahill DP, Aldape KD, Betensky RA, 2011, Mosaic amplification of multiple receptor tyrosine kinase genes in glioblastoma. Cancer Cell 20(6):810–817 Little SE, Popov S, Jury A, Bax DA, Doey L, Al-Sarraj S, Jurgensmeier JM, Jones C, 2012, Receptor tyrosine kinase genes amplified in glioblastoma exhibit a mutual exclusivity in variable proportions reflective of individual tumor heterogeneity. Cancer Res 72(7):1614–1620 Szerlip NJ, Pedraza A, Chakravarty D, Azim M, McGuire J, Fang Y, Ozawa T, Holland EC, Huse JT, Jhanwar S, 2012, Intratumoral heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma defines subpopulations with distinct growth factor response. Proc Natl Acad Sci 109(8):3041–3046 Sottoriva A, Spiteri I, Piccirillo SG, Touloumis A, Collins VP, Marioni JC, Curtis C, Watts C, Tavare S, 2013, Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc Natl Acad Sci U S A 110(10):4009–4014., DOI 10.1073/ pnas.1219747110 Purow B, Schiff D, 2009, Advances in the genetics of glioblastoma: are we reaching critical mass? Nat Rev Neurol 5(8):419–426 Ikediobi O, 2008, Somatic pharmacogenomics in cancer. Pharmacogenomics J 8(5):305–314 Inda M-d-M, Bonavia R, Seoane J, 2014, Glioblastoma multiforme: a look inside its heterogeneous nature. Cancers 6(1):226–239 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 777 EP 787 DI 10.1007/s12253-014-9833-3 PG 11 ER PT J AU Najbauer, J Kraljik, N Nemeth, P AF Najbauer, Joseph Kraljik, Nikola Nemeth, Peter TI Glioma Stem Cells: Markers, Hallmarks and Therapeutic Targeting by Metformin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Cancer stem cells; Glioma; Glioma stem cells; Metformin; Therapeutic targeting ID Cancer stem cells; Glioma; Glioma stem cells; Metformin; Therapeutic targeting AB Malignant gliomas are among the deadliest primary brain tumors. Despite multimodal therapy and advances in chemotherapy, imaging, surgical and radiation techniques, these tumors remain virtually incurable. Glioma stem cells may be responsible for resistance to traditional therapies and tumor recurrence. Therefore, elimination of glioma stem cells may be crucial for achieving therapeutic efficacy. Metformin, a small molecule drug widely used in the therapy of type 2 diabetes, has shown significant anti-tumor effects in patients with breast cancer and prostate cancer. Recent preclinical data suggest that metformin also has therapeutic effects against glioma. Here we review the markers and hallmarks of glioma stem cells, and the molecular mechanisms involved in therapeutic targeting of glioma stem cells by metformin. C1 [Najbauer, Joseph] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7624 Pecs, Hungary. [Kraljik, Nikola] Institute of Public Health for the Osijek-Baranja County, F. Krezme 1, 31000 Osijek, Croatia. [Nemeth, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7624 Pecs, Hungary. RP Najbauer, J (reprint author), University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, H-7624 Pecs, Hungary. EM jnajbauer@yahoo.com CR Wen PY, Kesari S, 2008)Malignant gliomas in adults. N Engl J Med 359:492–507 Stupp R, HegiME, MasonWP, van den Bent MJ, TaphoornMJ et al, 2009, Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10:459–466 Westphal M, Lamszus K, 2011, The neurobiology of gliomas: from cell biology to the development of therapeutic approaches. Nat Rev Neurosci 12:495–508 Gilbertson RJ, Rich JN, 2007, Making a tumour’s bed: glioblastoma stem cells and the vascular niche. Nat Rev Cancer 7:733–736 Frank RT, Aboody KS, Najbauer J, 2011, Strategies for enhancing antibody delivery to the brain. Biochim Biophys Acta 1816:191–198 Minchinton AI, Tannock IF, 2006, Drug penetration in solid tumours. Nat Rev Cancer 6:583–592 Neuwelt EA, Bauer B, Fahlke C, Fricker G, Iadecola C et al, 2011, Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci 12:169–182 Norden AD, Young GS, Setayesh K, Muzikansky A, Klufas R et al, 2008, Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurol 70:779–787 Visvader JE, 2011, Cells of origin in cancer. Nature 469:314–322 Visvader JE, Lindeman GJ, 2012, Cancer stem cells: current status and evolving complexities. Cell Stem Cell 10:717–728 Nguyen LV,Vanner R, Dirks P, Eaves CJ, 2012, Cancer stemcells: an evolving concept. Nat Rev Cancer 12:133–143 Bjerkvig R, Tysnes BB, Aboody KS, Najbauer J, Terzis AJ, 2005, Opinion: the origin of the cancer stem cell: current controversies and new insights. Nat Rev Cancer 5:899–904 Filatova A, Acker T, Garvalov BK, 2013, The cancer stem cell niche, s): the crosstalk between glioma stem cells and their microenvironment. Biochim Biophys Acta 1830:2496–2508 Binello E, Germano IM, 2011, Targeting glioma stem cells: a novel framework for brain tumors. Cancer Sci 102:1958–1966 Facchino S, Abdouh M, Bernier G, 2011, Brain cancer stem cells: current status on glioblastoma multiforme. Cancers, Basel, 3:1777– 1797 Vescovi AL, Galli R, Reynolds BA, 2006, Brain tumour stem cells. Nat Rev Cancer 6:425–436 Eramo A, Ricci-Vitiani L, Zeuner A, Pallini R, Lotti F et al, 2006, Chemotherapy resistance of glioblastoma stem cells. Cell Death Differ 13:1238–1241 Bao S,Wu Q, McLendon RE, Hao Y, Shi Q et al, 2006, Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444:756–760 Dean M, Fojo T, Bates S, 2005, Tumour stem cells and drug resistance. Nat Rev Cancer 5:275–284 Singh SK, Clarke ID, Hide T, Dirks PB, 2004, Cancer stem cells in nervous system tumors. Oncogene 23:7267–7273 Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J et al, 2004, Identification of human brain tumour initiating cells. Nature 432: 396–401 Persano L, Rampazzo E, Basso G, Viola G, 2013, Glioblastoma cancer stem cells: role of the microenvironment and therapeutic targeting. Biochem Pharmacol 85:612–622 Huse JT, Holland EC, 2010, Targeting brain cancer: advances in the molecular pathology of malignant glioma and medulloblastoma. Nat Rev Cancer 10:319–331 Quinn BJ, Kitagawa H, Memmott RM, Gills JJ, Dennis PA, 2013, Repositioningmetformin for cancer prevention and treatment. Trends Endocrinol Metab 24:469–480 Dunaif A, 2008, Drug insight: insulin-sensitizing drugs in the treatment of polycystic ovary syndrome–a reappraisal. Nat Clin Pract Endocrinol Metab 4:272–283 Pollak M, 2012, The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer 12:159–169 Emami Riedmaier A, Fisel P, Nies AT, Schaeffeler E, Schwab M, 2013, Metformin and cancer: from the old medicine cabinet to pharmacological pitfalls and prospects. Trends Pharmacol Sci 34: 126–135 Bost F, Sahra IB, LeMarchand-Brustel Y, Tanti JF, 2012, Metformin and cancer therapy. Curr Opin Oncol 24:103–108 Pernicova I, Korbonits M, 2014, Metformin–mode of action and clinical implications for diabetes and cancer. Nat Rev Endocrinol 10:143–156 Inzucchi SE, 2002, Oral antihyperglycemic therapy for type 2 diabetes: scientific review. JAMA 287:360–372 Witters LA, 2001, The blooming of the French lilac. J Clin Invest 108:1105–1107 Isakovic A, Harhaji L, Stevanovic D, Markovic Z, Sumarac- DumanovicMet al, 2007, Dual antiglioma action of metformin: cell cycle arrest andmitochondria-dependent apoptosis. CellMol Life Sci 64:1290–1302 Shackelford DB, Shaw RJ, 2009, The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat Rev Cancer 9:563–575 Liu X, Chhipa RR, Pooya S, Wortman M, Yachyshin S et al, 2014, Discrete mechanisms of mTOR and cell cycle regulation by AMPK agonists independent of AMPK. Proc Natl Acad Sci U S A 111: E435–444 Wurth R, Pattarozzi A, Gatti M, Bajetto A, Corsaro A et al, 2013, Metformin selectively affects human glioblastoma tumor-initiating cell viability: a role for metformin-induced inhibition of Akt. Cell Cycle 12:145–156 Sunayama J, Sato A, Matsuda K, Tachibana K, Watanabe E et al, 2011, FoxO3a functions as a key integrator of cellular signals that control glioblastoma stem-like cell differentiation and tumorigenicity. Stem Cells 29:1327–1337 Sato A, Sunayama J, Okada M, Watanabe E, Seino S et al, 2012, Glioma-initiating cell elimination bymetformin activation of FOXO3 via AMPK. Stem Cells Transl Med 1:811–824 Labuzek K, Suchy D, Gabryel B, Bielecka A, Liber S et al, 2010, Quantification of metformin by the HPLC method in brain regions, cerebrospinal fluid and plasma of rats treated with lipopolysaccharide. Pharmacol Rep 62:956–965 Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD, 2005, Metformin and reduced risk of cancer in diabetic patients. BMJ 330:1304–1305 Bowker SL, Majumdar SR, Veugelers P, Johnson JA, 2006, Increased cancer-related mortality for patients with type 2 diabetes who use sulfonylureas or insulin. Diabetes Care 29: 254–258 Jiralerspong S, Palla SL, Giordano SH,Meric-Bernstam F, Liedtke C et al, 2009, Metformin and pathologic complete responses to neoadjuvant chemotherapy in diabetic patients with breast cancer. J Clin Oncol 27:3297–3302 Chlebowski RT, McTiernan A, Wactawski-Wende J, Manson JE, Aragaki AK et al, 2012, Diabetes, metformin, and breast cancer in postmenopausal women. J Clin Oncol 30:2844– 2852 Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K, 2009, Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Cancer Res 69:7507–7511 Hirsch HA, Iliopoulos D, Struhl K, 2013, Metformin inhibits the inflammatory response associated with cellular transformation and cancer stem cell growth. Proc Natl Acad Sci U S A 110:972–977 Margel D, Urbach DR, Lipscombe LL, Bell CM, Kulkarni G et al, 2013, Metformin use and all-cause and prostate cancer-specific mortality among men with diabetes. J Clin Oncol 31:3069–3075 Aboody KS, Najbauer J, Metz MZ, D’Apuzzo M, Gutova M et al, 2013, Neural stem cell-mediated enzyme/prodrug therapy for glioma: preclinical studies. Sci Transl Med 5:184ra159 Lee C, Raffaghello L, Brandhorst S, Safdie FM, Bianchi G et al, 2012, Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med 4:124ra127 Clarke J, Penas C, Pastori C, Komotar RJ, Bregy A et al, 2013, Epigenetic pathways and glioblastoma treatment. Epigenetics 8:785– 795 Najbauer J, Huszthy PC, BarishME, Garcia E,Metz MZ et al, 2012, Cellular host responses to gliomas. PLoS One 7:e35150 Dimov I, Tasic-Dimov D, Conic I, Stefanovic V, 2011, Glioblastoma multiforme stem cells. Sci World J 11:930–958 Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H, 2012, The brain tumor microenvironment. Glia 60:502–514 Cheng L, Huang Z, Zhou W, Wu Q, Donnola S et al, 2013, Glioblastoma stem cells generate vascular pericytes to support vessel function and tumor growth. Cell 153:139–152 Kourelis TV, Siegel RD, 2012, Metformin and cancer: new applications for an old drug. Med Oncol 29:1314–1327 Del Barco S, Vazquez-Martin A, Cufi S, Oliveras-Ferraros C, Bosch- Barrera J et al, 2011, Metformin: multi-faceted protection against cancer. Oncotarget 2:896–917 Rattan R, Ali Fehmi R,Munkarah A, 2012, Metformin: an emerging new therapeutic option for targeting cancer stem cells and metastasis. J Oncol 2012:928127 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 789 EP 797 DI 10.1007/s12253-014-9837-z PG 9 ER PT J AU Chen, W Jiang, X Luo, Z AF Chen, Wei Jiang, Xiaofei Luo, Zhuang TI WWP2: A Multifunctional Ubiquitin Ligase Gene SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE WWP2 (AIP2); Ubiquitylation; Cancer ID WWP2 (AIP2); Ubiquitylation; Cancer AB The ubiquitin-proteasome system plays an important role in various celluar processes. WWP2, a recently identified ubiquitin E3 ligase, has been proved a multifunctional gene by degradation a series of targets via ubiquitindependent proteasome system, including PETN, Smads, Oct4, EGR2, TIRF and so. Hereafter, we reviewed the recent research process about the function of WWP2. C1 [Chen, Wei] Sichuan North Medical College, Department of Pathophysiology, 637100 Nanchong, China. [Jiang, Xiaofei] The Seventh People’s Hospital of Chengdu, Department of General Surgery, 610041 Chengdu, China. [Luo, Zhuang] Kunming Medical University, First Affiliated Hospital, Department of Respiratory Diseases, 650032 Kunming, China. RP Luo, Z (reprint author), Kunming Medical University, First Affiliated Hospital, Department of Respiratory Diseases, 650032 Kunming, China. EM skyny4511@126.com CR Bernassola F, Karin M, Ciechanover A, Melino G, 2008, The HECT family of E3 ubiquitin ligases: multiple players in cancer development. Cancer Cell 14(1):10–21 Chen C, Matesic LE, 2007, The Nedd4-like family of E3 ubiquitin ligases and cancer. Cancer Metastasis Rev 26(3–4):587–604 Harvey KF, Kumar S, 1999, Nedd4-like proteins: an emerging family of ubiquitin-protein ligases implicated in diverse cellular functions. Trends Cell Biol 9(5):166–169 Ingham RJ, Gish G, Pawson T, 2004, The Nedd4 family of E3 ubiquitin ligases: functional diversity within a common modular architecture. Oncogene 23(11):1972–1984 Shearwin-Whyatt L, Dalton HE, Foot N, Kumar S, 2006, Regulation of functional diversity within the Nedd4 family by accessory and adaptor proteins. BioEssays : news and rev in mol, cell and dev biol 28(6):617–628 Kerscher O, Felberbaum R, Hochstrasser M, 2006, Modification of proteins by ubiquitin and ubiquitin-like proteins. Annu Rev Cell Dev Biol 22:159–180 Navon A, Ciechanover A, 2009, The 26 S proteasome: from basic mechanisms to drug targeting. J of biol chem 284(49):33713–33718 Adhikary S, Marinoni F, Hock A, Hulleman E, Popov N, Beier R, Bernard S, Quarto M, Capra M, Goettig S et al, 2005, The ubiquitin ligase HectH9 regulates transcriptional activation by Myc and is essential for tumor cell proliferation. Cell 123(3):409–421 Alexandru G, Graumann J, Smith GT, Kolawa NJ, Fang R, Deshaies RJ, 2008, UBXD7 binds multiple ubiquitin ligases and implicates p97 in HIF1alpha turnover. Cell 134(5):804–816 Zhong Q, GaoW,Du F,WangX(2005)Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell 121(7):1085–1095 Melino G, Gallagher E, Aqeilan RI, Knight R, Peschiaroli A, Rossi M, Scialpi F, Malatesta M, Zocchi L, Browne G et al, 2008, Itch: a HECT-type E3 ligase regulating immunity, skin and cancer. Cell Death Differ 15(7):1103–1112 Kralj M, Tusek-Bozic L, Frkanec L, 2008, Biomedical potentials of crown ethers: prospective antitumor agents. ChemMedChem 3(10): 1478–1492 Pirozzi G, McConnell SJ, Uveges AJ, Carter JM, Sparks AB, Kay BK, Fowlkes DM, 1997, Identification of novel human WW domain-containing proteins by cloning of ligand targets. J of biol chem 272(23):14611–14616 Wood JD, Yuan J, Margolis RL, Colomer V, Duan K, Kushi J, Kaminsky Z, Kleiderlein JJ, Sharp AH, Ross CA, 1998, Atrophin- 1, the DRPLA gene product, interacts with two families of WW domain-containing proteins. Mol Cell Neurosci 11(3):149–160 Xu H, Wang W, Li C, Yu H, Yang A,Wang B, Jin Y, 2009, WWP2 promotes degradation of transcription factor OCT4 in human embryonic stem cells. Cell Res 19(5):561–573 Soond SM, Chantry A, 2011, Selective targeting of activating and inhibitory Smads by distinct WWP2 ubiquitin ligase isoforms differentially modulates TGFbeta signalling and EMT. Oncogene 30(21): 2451–2462 Lu PJ, ZhouXZ, ShenM, LuKP, 1999, Function ofWWdomains as phosphoserine- or phosphothreonine-binding modules. Science 283(5406):1325–1328 Liao B, Jin Y(2010)Wwp2mediatesOct4 ubiquitination and its own auto-ubiquitination in a dosage-dependent manner. Cell Res 20(3): 332–344 Wiesner S, Ogunjimi AA, Wang HR, Rotin D, Sicheri F, Wrana JL, Forman-Kay JD, 2007, Autoinhibition of the HECT-type ubiquitin ligase Smurf2 through its C2 domain. Cell 130(4):651–662 Yamoah K, Oashi T, Sarikas A, Gazdoiu S, Osman R, Pan ZQ, 2008, Autoinhibitory regulation of SCF-mediated ubiquitination by human cullin 1’s C-terminal tail. Proc Natl Acad Sci U S A 105(34):12230– 12235 Kobashigawa Y, Tomitaka A, Kumeta H, Noda NN, Yamaguchi M, Inagaki F, 2011, Autoinhibition and phosphorylation-induced activationmechanisms of human cancer and autoimmune disease-related E3 protein Cbl-b. Proc Natl Acad Sci U S A 108(51):20579–20584 Chou YC, Keszei AF, Rohde JR, Tyers M, Sicheri F, 2012, Conserved structural mechanisms for autoinhibition in IpaH ubiquitin ligases. J of biol chem 287(1):268–275 Chaugule VK, Burchell L, Barber KR, Sidhu A, Leslie SJ, Shaw GS, Walden H, 2011, Autoregulation of Parkin activity through its ubiquitin-like domain. The EMBO j 30(14):2853–2867 Maddika S, Kavela S, Rani N, Palicharla VR, Pokorny JL, Sarkaria JN, Chen J, 2011, WWP2 is an E3 ubiquitin ligase for PTEN. Nat Cell Biol 13(6):728–733 Pardal R, Clarke MF, Morrison SJ, 2003, Applying the principles of stem-cell biology to cancer. Nat Rev Cancer 3(12):895–902 Al-Hajj M, Clarke MF, 2004, Self-renewal and solid tumor stem cells. Oncogene 23(43):7274–7282 Beachy PA, Karhadkar SS, Berman DM, 2004, Tissue repair and stem cell renewal in carcinogenesis. Nature 432(7015):324–331 Gidekel S, Pizov G, Bergman Y, Pikarsky E, 2003, Oct-3/4 is a dosedependent oncogenic fate determinant. Cancer Cell 4(5):361–370 Qian YW, Chen Y, YangW, Fu J, Cao J, Ren YB, Zhu JJ, Su B, Luo T, Zhao XF et al, 2012, p28(GANK, prevents degradation of Oct4 and promotes expansion of tumor-initiating cells in hepatocarcinogenesis. Gastroenterology 142(7):1547–1558 Zhi X, Chen C, 2012, WWP1: a versatile ubiquitin E3 ligase in signaling and diseases. Cell and mol life sci : CMLS 69(9):1425– 1434 Xu HM, Liao B, Zhang QJ,Wang BB, Li H, Zhong XM, Sheng HZ, Zhao YX, Zhao YM, Jin Y, 2004, Wwp2, an E3 ubiquitin ligase that targets transcription factor Oct-4 for ubiquitination. J of biol chem 279(22):23495–23503 Lu D, Davis MP, Abreu-Goodger C, Wang W, Campos LS, Siede J, Vigorito E, SkarnesWC, Dunham I, Enright AJ et al, 2012, MiR-25 regulatesWwp2 and Fbxw7 and promotes reprogramming of mouse fibroblast cells to iPSCs. PLoS ONE 7(8):e40938 Chen A, Gao B, Zhang J, McEwen T, Ye SQ, Zhang D, Fang D, 2009, The HECT-type E3 ubiquitin ligase AIP2 inhibits activationinduced T-cell death by catalyzing EGR2 ubiquitination. Mol Cell Biol 29(19):5348–5356 Yang Y, Liao B,Wang S, Yan B, Jin Y, Shu HB,Wang YY, 2013, E3 ligase WWP2 negatively regulates TLR3-mediated innate immune response by targeting TRIF for ubiquitination and degradation. Proceedings of the National Academy of Sciences of the United States of America McDonald FJ, Western AH, McNeil JD, Thomas BC, Olson DR, Snyder PM, 2002, Ubiquitin-protein ligase WWP2 binds to and downregulates the epithelial Na(+, channel. Am j of physiol Renal physiol 283(3):F431–F436 Foot NJ, Dalton HE, Shearwin-Whyatt LM,Dorstyn L, Tan SS, Yang B, Kumar S, 2008, Regulation of the divalent metal ion transporter DMT1 and iron homeostasis by a ubiquitin-dependent mechanism involving Ndfips and WWP2. Blood 112(10):4268–4275 Li H, Zhang Z, Wang B, Zhang J, Zhao Y, Jin Y, 2007, Wwp2- mediated ubiquitination of the RNA polymerase II large subunit in mouse embryonic pluripotent stemcells.Mol Cell Biol 27(15):5296– 5305 Marcucci R, Brindle J, Paro S, Casadio A, Hempel S, Morrice N, Bisso A, Keegan LP, Del Sal G, O’Connell MA, 2011, Pin1 and WWP2 regulate GluR2 Q/R site RNA editing by ADAR2 with opposing effects. The EMBO j 30(20):4211–4222 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 799 EP 803 DI 10.1007/s12253-014-9838-y PG 5 ER PT J AU Hu, X Xin, Y Xiao, Y Zhao, J AF Hu, Xiaobin Xin, Yan Xiao, Yuping Zhao, Jing TI Overexpression of YAP1 is Correlated with Progression, Metastasis and Poor Prognosis in Patients with Gastric Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE YAP1; Gastriccarcinoma; Metastasis; Prognosis ID YAP1; Gastriccarcinoma; Metastasis; Prognosis AB YAP1 is overexpressed in numerous cancers, but its molecular mechanism in the carcinogenesis and clinic significance in tumor diagnosis and prognosis remains to be determined. We attempted to analyze the clinicopathologic significance of YAP1 expression and the correlation of the YAP1 levels with the progression, metastasis and prognosis of patients with gastric carcinoma. By immunohistochemistry, we determined YAP1 expression in 214 of primary gastric carcinoma (GC), 167 of matched normal gastric mucosa, 40 of chronic atrophic gastritis, 11 of dysplasia and 73 of intestinal metaplasia. The positive rate of YAP1 in gastric carcinoma was significantly higher than that in normal gastric mucosa, chronic atrophic gastritis and intestinal metaplasia. In the gastric cancers with lymph node metastasis, the positive rate of YAP1 was much higher than that in the group without lymph nodemetastasis. Moreover, gastric cancer patients with YAP1 overexpression demonstrated poorer prognosis than those with YAP1 negative staining. Finally, multivariate analysis of 191 patients with gastric carcinoma indicated that YAP1 overexpression, the invasion depth and lymph node metastasis were high hazard factors for gastric carcinoma. Our results demonstrated that YAP1 overexpression is correlated to the progression, lymph node metastasis and poor prognosis of gastric carcinoma, suggesting that overexpression of YAP1 might be an adjuvant factor for predicting lymph node metastasis, and a useful biomarker for the diagnosis and prediction of prognosis in patients with gastric cancers. C1 [Hu, Xiaobin] No. 1 Hospital of China Medical University, Cancer Institute and Department of Pediatrics, 155 North Nanjing Street, Heping District, 110001 Shenyang, Liaoning Province, China. [Xin, Yan] China Medical University, The First Affiliated Hospital, Department of General Surgery, 155 North Nanjing Street, Heping District, 110001 Shenyang, Liaoning Province, China. [Xiao, Yuping] China Medical University, The First Affiliated Hospital, Department of General Surgery, 155 North Nanjing Street, Heping District, 110001 Shenyang, Liaoning Province, China. [Zhao, Jing] China Medical University, The First Affiliated Hospital, Department of General Surgery, 155 North Nanjing Street, Heping District, 110001 Shenyang, Liaoning Province, China. RP Xin, Y (reprint author), China Medical University, The First Affiliated Hospital, Department of General Surgery, 110001 Shenyang, China. EM yxin@mail.cmu.edu.cn CR Jackson C, Cunningham D, Oliveira J, 2009, Gastric cancer: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol 20:34–36 Thun MJ, DeLancey JO, CenterMM, Jemal A,Ward EM(2010, The global burden of cancer: priorities for prevention. Carcinogenesis 31: 100–110 Zhao B,Wei X, LiW, Udan RS, Yang Q, KimJ, Xie J, Ikenoue T, Yu J, Li L, Zheng P, Ye K, Chinnaiyan A, Halder G, Lai ZC, Guan KL, 2007, Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev 21:2747–2761 Dong J, Feldmann G, Huang J, Wu S, Zhang N, Comerford SA, Gayyed MF, Anders RA, Maitra A, Pan D, 2007, Elucidation of a universal size-control mechanism in drosophila and mammals. Cell 130:1120–1133 Zender L, Spector MS, XueW, Flemming P, Cordon-Cardo C, Silke J, Fan ST, Luk JM, Wigler M, Hannon GJ, Mu D, Lucito R, Powers S, Lowe SW, 2006, Identification and validation of oncogenesin liver cancer using an integrative oncogenomic approach. Cell 125: 1253–1267 ZhangX, George J, Deb S, Degoutin JL, Takano EA, Fox SB, AOCS Study group, Bowtell DD, Harvey KF, 2011, The Hippo pathway transcriptional co-activator, YAP, is an ovarian cancer oncogene. Oncogene 30:2810–2822 Goulev Y, Fauny JD, Gonzalez-Marti B, Flagiello D, Silber J, Zider A, 2008, SCALLOPED interacts with YORKIE, the nuclear effector of the Hippo tumor-suppressor pathway in Drosophila. Curr Biol 18: 435–441 Zhang L, Ren F, Zhang Q, Chen Y, Wang B, Jiang J, 2008, The TEAD/TEF family of transcription factor Scal-loped mediates Hippo signaling in organ size control. Dev Cell 14:377–387 Wu S, Liu Y, Zheng Y, Dong J, Pan D, 2008, TheTEAD/TEF family protein Scalloped mediates transcrip-tional output of the Hippo growth-regulatory pathway. Dev Cell 14:388–398 Huang J, Wu S, Barrera J, Matthews K, Pan D, 2005, The Hipposignaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Dro sophila homolog of YAP. Cell 122:421–434 Camargo FD, Gokhale S, Johnnidis JB, Fu D, Bell GW, Jaenisch R, Brummelkamp TR, 2007, YAP1 increases organ size and expands undifferentiated progenitor cells. Curr Biol 17:2054–2060 OverholtzerM, Zhang J, Smolen GA,Muir B, LiW, Sgroi DC, Deng CX, Brugge JS, Haber DA, 2006, Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon. Proc Natl Acad Sci U S A 103:12405–12410 Tufail R, Jorda M, Zhao W, Reis I, Nawaz Z, 2011, Loss of Yesassociated protein, YAP, expression is associated with estrogen and progesterone receptors negativity in invasive breast carcinomas. Breast Cancer Res Treat 131:743–750 Steinhardt AA, Gayyed MF, Klein AP, Dong J, Maitra A, Pan D, Montgomery EA, Anders RA, 2008, Expression of Yes-associated protein in common solid tumors. Hum Pathol 39:1582–1589 Zhang J, Ji JY, Yu M, Overholtzer M, Smolen GA, Wang R, Brugge JS, Dyson NJ, Haber DA, 2009, YAP-dependent induction of amphiregulin identifies a non-cell-autonomous component of the Hippo pathway. Nat Cell Biol 11:1444–1450 Xu MZ, Yao TJ, Lee NP, Ng IO, Chan YT, Zender L, Lowe SW, Poon RT, Luk JM, 2009, Yes-associated protein is an independent prognostic marker in hepatocellular carcinoma. Cancer 115:4576–4585 Lam-Himlin DM, Daniels JA, Gayyed MF, Dong J, Maitra A, Pan D, Montgomery EA, Anders RA, 2006, The hippo pathway in human upper gastrointestinal dysplasia and carcinoma: a novel oncogenic pathway. Int J Gastrointest Cancer 37:103– 109 KangW, Tong JH, Chan AW, Lee TL, Lung RW, Leung PP, So KK, Wu K, Fan D, Yu J, Sung JJ, To KF, 2011, Yes-associated protein 1 exhibits oncogenic property in gastric cancer and its nuclear accumulation associates with poor prognosis. Clin Cancer Res 17:2130– 2139 Da CL, Xin Y, Zhao J, Luo XD, 2009, Significance and relationship between Yes-associated protein and survivin expression in gastric carcinoma and precancerous lesions. World J Gastroenterol 15:4055–4061 Song M, Cheong JH, Kim H, Noh SH, Kim H, 2012, Nuclear expression of Yes-associated protein 1 correlates with poor prognosis in intestinal type gastric cancer. Anticancer Res 32:3827–3834 Zhang J, Xu ZP, Yang YC, Zhu JS, Zhou Z, Chen WX, 2012, Expression of Yes-associated protein in gastric adenocarcinoma and inhibitory effects of its knockdown on gastric cancer cell proliferation and metastasis. Int J Immunopathol Pharmacol 25:583–590 Zhang J, Yang YC, Zhu JS, Zhou Z, Chen WX, 2012, Clinicopathologic characteristics of YES-associated protein 1 overexpression and its relationship to tumor biomarkers in gastric cancer. Int J Immunopathol Pharmacol 25:977–987 Zhou GX, Li XY, Zhang Q, Zhao K, Zhang CP, Xue CH, Yang K, Tian ZB, 2013, Effects of the hippo signaling pathway in human gastric cancer. Asian Pac J Cancer Prev 14:5199–5205 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 805 EP 811 DI 10.1007/s12253-014-9757-y PG 7 ER PT J AU Becsagh, P Szakacs, O AF Becsagh, Peter Szakacs, Orsolya TI Setting Up a Probe Based, Closed Tube Real-Time PCR Assay for Focused Detection of Variable Sequence Alterations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE qPCR; Taqman probe; Simple probe; c-Kit; Mutations; DNA ID qPCR; Taqman probe; Simple probe; c-Kit; Mutations; DNA AB During diagnostic workflow when detecting sequence alterations, sometimes it is important to design an algorithm that includes screening and direct tests in combination. Normally the use of direct test, which is mainly sequencing, is limited. There is an increased need for effective screening tests, with "closed tube" during the whole process and therefore decreasing the risk of PCR product contamination. The aim of this study was to design such a closed tube, detection probe based screening assay to detect different kind of sequence alterations in the exon 11 of the human c-kit gene region. Inside this region there are variable possible deletions and single nucleotide changes. During assay setup, more probe chemistry formats were screened and tested. After some optimization steps the taqman probe format was selected. C1 [Becsagh, Peter] Roche Hungary LtdBudaors, Hungary. [Szakacs, Orsolya] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. RP Becsagh, P (reprint author), Roche Hungary Ltd, Budaors, Hungary. EM peter.becsagh@roche.com CR Antonescu CR, Sommer G, Sarran L, Tschernyavsky SJ, Riedel E, Woodruff JM, Robson M, Maki R, Brennan MF, Ladanyi M, DeMatteo RP, Besmer P, 2003, Association of KIT exon 9 mutations with nongastric primary site and aggressive behavior: KIT mutation analysis and clinical correlates of 120 gastrointestinal stromal tumors. Clin Cancer Res 9(9):3329–3337 Kondiah K, Swanepoel R, Paweska JT, Burt FJ, 2010, A simpleprobe real-time PCR assay for genotyping reassorted and nonreassorted isolates of Crimean-Congo hemorrhagic fever virus in southern Africa. J Virol Methods 169(1):34–38 Grannemann S, Landt O, Breuer S, Blomeke B, 2005, LightTyper assay with locked-nucleic-acid-modified oligomers for genotyping of the toll-like receptor 4 polymorphisms A896G and C1196T. Clin Chem 51(8):1523–1525 Frances F, Corella D, Sorli JV, Guillen M, Gonzalez JI, Portoles O, 2005, Validating a rapid method for detecting common polymorphisms in the APOA5 gene by melting curve analysis using LightTyper. Clin Chem 51:1279–1282 Frances F, Portoles O, Sorli JV, Guillen M, Gonzalez JI, Corella D, 2008, Single tube optimisation of APOE genotyping based on melting curve analysis. Clin Biochem 41(10–11):923–926 McGuigan FE, Ralston SH, 2002, Single nucleotide polymorphism detection: allelic discrimination using TaqMan. Psychiatr Genet 12(3):133–136 Hui L, DelMonte T, Ranade K, 2008, Genotyping using the TaqMan assay. Curr Protoc Hum Genet., DOI 10.1002/0471142905.hg0210s56 Zhou L, Myers AN, Vandersteen JG, Wang L, Wittwer CT, 2004, Closed-tube genotyping with unlabeled oligonucleotide probes and a saturating DNA dye. Clin Chem 50(8):1328–1335 Roche Applied Science, 2004, LightCycler Manual, Technical Note No. LC 18/2004: Assay formats for use in real-time PCR. https:// www.roche-applied-science.com/sis/rtpcr/lightcycler/ lightcycler—docs/technical—notes/lc—18.pdf Mackay J, Landt O, 2007, Real-time PCR fluorescent chemistries. Methods Mol Biol 353:237–261 Korbie DJ, Mattick JS, 2008, Touchdown PCR for increased specificity and sensitivity in PCR amplification. Nat Protoc 3(9):1452–1456 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 813 EP 818 DI 10.1007/s12253-014-9759-9 PG 6 ER PT J AU Alshenawy, AH AF Alshenawy, AlSaeid Hanan TI Utility of Immunohistochemical Markers in Diagnosis of Follicular Cell Derived Thyroid Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thyroid carcinoma; Immunohistochemistry; CD56; HBME-1; Gatectin-3; CK19 ID Thyroid carcinoma; Immunohistochemistry; CD56; HBME-1; Gatectin-3; CK19 AB Differentiating the follicular derived lesions can be challenging. Although immunohistochemistry is generally accepted as a useful ancillary technique in the diagnosis, controversy exists regarding the best marker or combination of markers to distinguish each lesion from its mimics. In this study, we aimed at evaluating multiple markers to compare their sensitivity and usefulness, and to find out if a combination of the evaluated markers can be of additional value in discriminating thyroid lesions. The study included two groups of follicular derived thyroid lesions; benign group (Grave’s disease, nodular goiter, Hashimoto’s and adenoma) and malignant group (papillary, follicular carcinoma, well differentiated tumors of unknown malignant potential and follicular tumour of unknown malignant potential). Immunohistochemical evaluation of CD56, HBME-1, Gaectin-3 and CK19 were done. The sensitivity, specificity for each marker and their combination were calculated. Each marker was sensitive and specific for certain lesion but the sensitivity and specificity was increased when use combination of markers. Although no single marker is completely sensitive and specific for follicular thyroid lesions, the combination of CD56, HBME-1, Gaectin-3 and CK19 attains high sensitivity and specificity in diagnosis. C1 [Alshenawy, AlSaeid Hanan] Tanta University, Faculty of Medicine, Department of Pathology, 25 Hamdy Gado streetTanta, Egypt. RP Alshenawy, AH (reprint author), Tanta University, Faculty of Medicine, Department of Pathology, Tanta, Egypt. EM hanan_alshenawy@yahoo.com CR Ying YL, Hans M, Job K et al, 2008, Combined immunostaining with galectin-3, fibronectin-1, CITED-1, Hector Battifora mesothelial-1, cytokeratin-19, peroxisome proliferator-activated receptor-g, and sodium/iodide symporter antibodies for the differential diagnosis of non-medullary thyroid carcinoma. Eur J Endocrinol 158:375–384 Guido F, Esther DR, Marco R et al, 2011, Follicular thyroid neoplasms can be classified as low- and high-risk according to HBME-1 andGalectin-3 expression on liquid-based fine-needle cytology. Eur J Endocrinol 165:447–453 Yoon LC, Mi KK, Jin-Won S et al, 2005, Immunoexpression of HBME-1, high molecular weight cytokeratin, cytokeratin 19, thyroid transcription factor-1, and E-cadherin in thyroid carcinomas. J Korean Med Sci 20(5):853–859 Yasuhiro I, Hiroshi Y, Chisato T et al, 2005, HBME-1 expression in follicular tumor of the thyroid: an investigation of whether it can be used as amarker to diagnose follicular carcinoma. AnticancerRes 25:179–182 Arturs O, Zenons N, Ilze S et al, 2012, Immunohistochemical expression of HBME-1, E-cadherin, and CD56 in the differential diagnosis of thyroid nodules. Medicina, Kaunas, 48(10):507–514 Mauro P, Jaime R, Roberta DP et al, 2005, Galectin-3 and HBME-1 expression in well-differentiated thyroid tumors with follicular architecture of uncertain malignant potential. Mod Pathol 18:541–546 Dina ED, Ahmed N, Salem A, 2008, Application of CD56, P63 and CK19 immunohistochemistry in the diagnosis of papillary carcinoma of the thyroid. Diagn Pathol 3–5 Jeffrey L, Bhuvanesh S, Giovanni T et al, 2006, Follicular variant of papillary thyroid carcinoma. A clinicopathologic study of a problematic entity. Cancer 107(6):1255–1264 Debdas B, Ram ND, Uttara C et al, 2012, Cytokeratin 19 immunoreactivity in the diagnosis of papillary thyroid carcinoma. Indian J Med Paediatr Oncol 33(2):107–111 RashaMA, Lobna SS, 2012, Potential diagnostic utility of CD56 and claudin-1 in papillary thyroid carcinoma and solitary follicular thyroid nodules. J Egypt Natl Cancer Inst 24:175–184 Husain AS, Jining F, Farah T et al, 2009, Differential expression of galectin-3, CK19, HBME1, and Ret oncoprotein in the diagnosis of thyroid neoplasms by fine needle aspiration biopsy. Cytojournal 6:18 Young JP, Soo HK, Dong CK et al, 2007, Diagnostic value of galectin-3, HBME-1, cytokeratin 19, high molecular weight cytokeratin, cyclin D1 and p27kip1 in the differential diagnosis of thyroid nodules. J Korean Med Sci 22(4):621–628 Sandrine R, Brahim E, Sergio F et al, 2002, Changes in galectin-7 and cytokeratin-19 expression during the progression of malignancy in thyroid tumors: diagnostic and biological implications.Mod Pathol 15(12):1294–1301 Prasad ML, Pellegata NS, Huang Y et al, 2005, Galectin-3, fibronectin-1, CITED-1, HBME1 and cytokeratin-19 immunohistochemistry is useful for the differential diagnosis of thyroid tumors. Mod Pathol 18:48–57 de Matos PS, Ferreira AP, de Oliveira FF et al, 2005, Usefulness of HBME-1, cytokeratin 19 and galectin-3 immunostaining in the diagnosis of thyroid malignancy. Histopathology 47:391–401 Ozolins A, Narbuts Z, Strumfa I et al, 2010, Diagnostic utility of immunohistochemical panel in various thyroid pathologies. Langenbecks Arch Surg 395(7):885–891 DeLellis RA, 2006, Pathology and genetics of thyroid carcinoma. J Surg Oncol 94(8):662–669 Chan JK, 2002, Strict criteria should be applied in the diagnosis of encapsulated follicular variant of papillary thyroid carcinoma. Am J Clin Pathol 117:16–18 Williams ED, 2000, Two proposals regarding the terminology of thyroid tumors. Int J Surg Pathol 8:181–183 Park WY, Jeong SM, Lee JH et al, 2009, Diagnostic value of decreased expression of CD56 protein in papillary carcinoma of the thyroid gland. Basic Appl Pathol 2:63–68 Nasr MR, Mukhopadhyay S, Zhang S et al, 2006, Immunohistochemical markers in diagnosis of papillary thyroid carcinoma: utility of HBME 1 combined with CK-19 immunostaining. Mod Pathol 19:1631–1637 Husain AS, Bo J, John B et al, 2010, Utility of immunohistochemical markers in differentiating benign from malignant follicular-derived thyroid nodules. Diagn Pathol 5:9 Qingbin S, Deguang W, Yi L et al, 2011, Diagnostic significance of CK19, TG, Ki67 and galectin-3 expression for papillary thyroid carcinoma in the northeastern region of China. Diagn Pathol 6:126– 131 Shin MK, Kim JW, Ju Y, 2011, CD56 and high molecular weight keratin as dignostic markers of papillary thyroid carcinoma. Korean J Pathol 45:477–484 Etem H, Ozekinci S, Mizrak B et al, 2010, The role of CD56, HBME-1, and p63 in follicular neoplasms of the thyroid. J Pathol 26:238–242 MiettinenM, Karkkainen P, 1996, Differential reactivity ofHBME-1 and CD15 antibodies in benign and malignant thyroid tumours. Virchows Arch 429:213–219 Cheung CC, Ezzat S, Freeman JL e t a l, 2001, Immunohistochemical diagnosis of papillary thyroid carcinoma. Mod Pathol 14:338–342 Gasbarri A, Martegani MP, Del Prete F et al, 1999, Galectin-3 and CD44v6 isoforms in the preoperative evaluation of thyroid nodules. J Clin Oncol 17:3494–3499 Saggiorato E, De Pompa R, Volante M et al, 2005, Characterization of thyroid ‘follicular neoplasms’ in fine-needle aspiration cytological specimens using a panel of immunohistochemical markers: a proposal for clinical application. Endocrinol Relat Cancer 12:305–317 Orlandi F, Saggiorato E, Pivano G et al, 1998, Galectin-3 is a presurgical marker of human thyroid carcinoma. Cancer Res 58: 3015–3020 Kovacs RB, Foldes J, Winkler G et al, 2003, The investigation of galectin-3 in diseases of the thyroid gland. Eur J Endocrinol 149(5): 449–453 Bartolazzi A, Gasbarri A, Papotti M et al, 2001, Application of an immunodiagnostic method for improving preoperative diagnosis of nodular thyroid lesions. Lancet 357:1644–1650 Collet JF, Fajac A, 2006, Galectin-3 immunodetection in thyroid fine-needle aspirates: technical procedure and results. Ann Pathol 26:347–351 KimMJ, Kim HJ, Hong SJ et al, 2006, Diagnostic utility of galectin- 3 in aspirates of thyroid follicular lesions. Acta Cytol 50:28–34 Sahoo S, Hoda SA, Rosai J et al, 2001, Cytokeratin 19 immunoreactivity in the diagnosis of papillary thyroid carcinoma. Am J Clin Pathol 116:696–702 Guyetant S,Michalak S,Valo I et al, 2003, Diagnosis of the follicular variant of papillary thyroid carcinoma significance of immunohistochemistry. Ann Pathol 23:11–20 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 819 EP 828 DI 10.1007/s12253-014-9760-3 PG 10 ER PT J AU Tang, Y Jiang, L Tang, W AF Tang, Yongyong Jiang, Li Tang, Wei TI Decreased Expression of NPRL2 in Renal Cancer Cells is Associated with Unfavourable Pathological, Proliferation and Apoptotic Features SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NPRL2; Renal cancer carcinoma; Transfection; Proliferation; Apoptosis ID NPRL2; Renal cancer carcinoma; Transfection; Proliferation; Apoptosis AB The tumor suppressor gene nitrogen permease regulator-like 2(NPRL2) NPRL2 expressed obviously in many normal human tissues, but reduced in expression in many human tumors significantly. In this study, we detected the expression of NPRL2 in 78 clear cell renal cell carcinoma (ccRCC) by immunohistochemistry and correlated it with clinicopathological parameters. Meanwhile, the function of NPRL2 in human ccRCC was further explored after transfected recombinant expressing plasmids pEGFP-N1- NPRL2 into human renal cancer 786-0 cells. NPRL2 protein showed high expression in 67 of 78 cases of adjacent normal tissues (85.9 %), which was significantly higher than that in ccRCC tissues (23/78, 29.5 %). Clinic pathological analysis showed that NPRL2 expression was significantly correlated with histological grade (P=0.044), TNM stage (P=0.025) and lymph node metastasis (P=0.028). MTT assay demonstrated that NPRL2 could obviously inhibit renal cancer cell proliferation. Flow cytometric analysis revealed that NPRL2 could induce renal cancer cells apoptosis and arrest the cell cycle in G0/G1 phase. In conclusion, NPRL2 is closely correlated to unfavourable pathological, proliferation and apoptotic features in ccRCC. C1 [Tang, Yongyong] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, No.1 Medical College Road, Yuzhong District, 400016 Chongqing, China. [Jiang, Li] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, No.1 Medical College Road, Yuzhong District, 400016 Chongqing, China. [Tang, Wei] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, No.1 Medical College Road, Yuzhong District, 400016 Chongqing, China. RP Tang, W (reprint author), Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 400016 Chongqing, China. EM tangwei2060@163.com CR Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63:11–30 Campbell SC, Novick AC, 2002, Renal tumors. In:Walsh PC, Retik AB, Vaughan ED, Wein AJ, eds, Kavoussi LR, Novick AC, Partin AW, Peters CA, ed.), Campbell’s urology. Elsevier, Philadelphia, pp 2672–2686 Jemal A, Siegel R, Ward E et al, 2008, Cancer statistics, 2008. CA Cancer J Clin 58:71–96 Arsanious A, Bjarnason GA, Yousef GM, 2009, From bench to bedside: current and future applications of molecular profiling in renal cell carcinoma. Mol Cancer 8(1):20 Jiang Z, Chu PG, Woda BA, Liu Q, Balaji KC, Rock KL, Wu CL, 2008, Combination of quantitative IMP3 and tumor stage: a new system to predict metastasis for patients with localized renal cell carcinomas. Clin Cancer Res 14:5579–5584 Lee JW, Bae SH, Jeong JW et al, 2004, Hypoxia-inducible factor, HIF-1)alpha: its protein stability and biological functions. Exp Mol Med 36(1):1–12 Hirota, Yan L, Tsunoda Tet al, 2006, Genome-wide gene expression profiles of clear cell renal cell carcinoma: identification of molecular targets for treatment of renal cell carcinoma. Int J Oncol 29(4):799– 827 Lerman MI, Minna JD, 2000, The 630-kb lung cancer homozygous deletion region on human chromosome 3p21.3: identification and evaluation of the resident candidate tumor suppressor genes. The International Lung Cancer Chromosome 3p21.3 Tumor Suppressor Gene Consortium. Cancer Res 60(21):6116–6133 Li J, Wang F, Haraldson et al, 2004, Functional characterization of the candidate tumor suppressor gene NPRL2/G21 located in 3p21.3C. Cancer Res 64(18):6438–6443 Sasatomi E, Finkelstein SD, Woods JD et al, 2002, Comparison of accumulated allele loss between primary tumor and lymph node metastasis in stage II non-small cell lung carcinoma: implications for the timing of lymph nodemetastasis and prognostic value. Cancer Res 62(9):2681–2689 Senchenko VN, Anedchenko EA, Kondratieva TT et al, 2010, Simultaneous down-regulation of tumor suppressor genes RBSP3/ CTDSPL, NPRL2/G21 and RASSF1A in primary non-small cell lung cancer. BMC Cancer 10:75 Otani S, Takeda S, Yamada S, Sakakima Y et al, 2009, The tumor suppressor NPRL2 in hepatocellular carcinoma plays an important role in progression and can be served as an independent prognostic factor. J Surg Oncol 100(5):358–363 Zabarovsky ER, Lerman MI, Minna JD, 2002, Tumor suppressor genes on chromosome 3p involved in the pathogenesis of lung and other cancers. Oncogene 21(45):6915–6935 Yi Lo PH, Chung Leung AC, Xiong W et al, 2006, Expression of candidate chromosome 3p21.3 tumor suppressor genes and downregulation of BLU in some esophageal squamous cell carcinomas. Cancer Lett 234(2):184–192 Chow LS, Lo KW, Kwong J et al, 2004, RASSF1A is a target tumor suppressor from 3p21.3 in nasopharyngeal carcinoma. Int J Cancer 109(6):839–847 Li J, Wang F, Protopopov A et al, 2004, Inactivation of RASSF1C during in vivo tumor growth identifies it as a tumor suppressor gene. Oncogene 23(35):5941–5949 Wang F, Grigorieva EV, Li J et al, 2008, HYAL1 and HYAL2 inhibit tumour growth in vivo but not in vitro. PLoS One 3(8):e3031 Senchenko et al, 2010, Simultaneous down-regulation of tumor suppressor genes RBSP3/CTDSPL, NPRL2/G21 and RASSF1A in primary non-small cell lung cancer. BMC Cancer 10:75 Jayachandran G, Ueda K, Wang B, Roth JA, Ji L, 2010, NPRL2 sensitizes human non-small cell lung, NSCLC, cells to cisplatin treatment by regulating components in the DNA repair pathway. PLoS One 5(8):e11994 Otani S, Takeda S, Yamada S et al, 2009, The tumor suppressor NPRL2 in hepatocellular carcinoma plays an important role in progression and can be served as an independent prognostic factor. J Surg Oncol 100:358–363 Edge SB, Byrd DR, Compton CC et al, 2009, AJCC cancer staging manual, 7th edn. Springer Verlag, New York Eble JN, Sauter G, Epstein JI, Sesterhenn IA, eds,, 2004, World Health Organization classification of tumors. Pathology and genetics of tumors of the urinary system and male genital organs. IARC Press, Lyon, pp 16–18 Xue et al, 2012, Overexpression of FoxM1 is associated with tumor progression in patients with clear cell renal cell carcinoma. J Transl Med 10:200 Otani S, Takeda S, Yamada S et al, 2009, The tumor supperessor NPRL2 in hepatocellular carcinoma plays an important role in progression and can be served as an independent prognostic factor. J Surg Oncol 100(5):358–363 Gao Y, Wang J, Fan G, 2012–2013, NPRL2 is an independent prognostic factor of osteosarcoma. Cancer Biomark 12(1):31–36 Yogurtcu B,Hatemi I,Aydin I et al, 2012)NPRL2 gene expression in the progression of colon tumors. Genet Mol Res 11(4):4810–4816 Ji L, Nishizaki M, Gao B et al, 2002, Expression of several genes in the human chromosome 3p21.3 homozygous deletion region by an adenovirus vector results in tumor suppressor activities in vitro and in vivo. Cancer Res 62(9):2715–2720 Parasons SL,Watson SA, Brown PD et al, 1997, Matrix metalloproteinases. Br J Surg 84(2):160–166 Economidou F, Tzortzaki EG, Schiza S et al, 2007, Microsatellite DNA analysis does not distinguish malignant from benign pleural effusions. Oncol Rep 18(6):1507–1512 Castagnaro A, Marangio E, Verduri A et al, 2007, Microsatellite analysis of induced sputum DNA in patients with lung cancer in heavy smokers and in healthy subjects. Exp Lung Res 33(6): 289–301 Kurata A, Katayama R, Watanabe T et al, 2008, TUSC4/NPRL2, a novel PDK1-interacting protein, inhibits PDK1 tyrosine phosphorylation and its downstream signaling. Cancer Sci 99(9):1827–1834 Kim JH, Kim H, Lee KY et al, 2006, Genetic polymorphisms of ataxia telangiectasia mutated affect lung cancer risk. HumMol Genet 15(7):1181–1186 Wang YX, Zhu SC, Feng W, Li J, Su JW, Li R, 2006, Effect on retardation of G2/M phase in esophageal carcinoma cells transfected with CHK1 and CHK2 shRNA after irradiation. Zhonghua Zhong Liu Za Zhi 28(8):572–577 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 829 EP 837 DI 10.1007/s12253-014-9761-2 PG 9 ER PT J AU Raza, Y Khan, A Farooqui, A Mubarak, M Facista, A Akhtar, ShS Khan, S Kazi, IJ Bernstein, C Kazmi, USh AF Raza, Yasir Khan, Adnan Farooqui, Amber Mubarak, Muhammad Facista, Alex Akhtar, Shakeel Syed Khan, Saeed Kazi, Iqbal Javed Bernstein, Carol Kazmi, Urooj Shahana TI Oxidative DNA Damage as a Potential Early Biomarker of Helicobacter pylori Associated Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Helicobacter pylori; Gastritis; 8-hydroxy-2′-deoxyguanosine; Gastric cancer; Immunohistochemistry ID Helicobacter pylori; Gastritis; 8-hydroxy-2′-deoxyguanosine; Gastric cancer; Immunohistochemistry AB Helicobacter pylori infection is an established risk factor for gastritis, gastric ulcer, peptic ulcer and gastric cancer. CagA +ve H. pylori has been associated with oxidative DNA damage of gastric mucosa but their combined role in the development of gastric cancer is still unknown. Here we compare the combined expression of cagA and 8-hydroxy-2′-deoxyguanosine (8-OHdG) in normal, gastritis and gastric cancer tissues. Two hundred gastric biopsies from patients with dyspeptic symptoms, 70 gastric cancer tissue samples and 30 gastric biopsies from non-dyspeptic individuals (controls) were included in this study and 8-OHdG was detected by immunohistochemistry (IHC). Histological features and the presence of H. pylori infection were demonstrated by Hematoxylin and Eosin (HE), Giemsa and alcian blue-periodic acid- Schiff ± diastase (AB-PAS ± D) staining. DNA was extracted from tissues and polymerase chain reaction (PCR) performed to determine the presence of ureaseA and cagA genes of H. pylori. The results showed the presence of H. pylori in 106 (53 %) gastric biopsies out of 200 dyspeptic patients, including 70 (66 %) cases of cagA + ve H. pylori. The presence of cagA gene and high expression of 8-OHdG was highly correlated with severe gastric inflammation and gastric cancer particularly, in cases with infiltration of chronic inflammatory cells (36.8 % cagA + ve, 18 %), neutrophilic activity (47.2 %, 25.5 %), intestinal metaplasia (77.7 %, 35.7 %) and intestinal type gastric cancer (95 %, 95.4 %) (p≤ 0.01). In conclusion, H. Pylori cagA gene expression and the detection of 8-OHdG adducts in gastric epithelium can serve as potential early biomarkers of H. Pylori-associated gastric carcinogenesis. C1 [Raza, Yasir] University of Karachi, Department of Microbiology, Immunology and Infectious Diseases Research LaboratoryKarachi, Pakistan. [Khan, Adnan] University of Karachi, Department of Microbiology, Immunology and Infectious Diseases Research LaboratoryKarachi, Pakistan. [Farooqui, Amber] University of Karachi, Department of Microbiology, Immunology and Infectious Diseases Research LaboratoryKarachi, Pakistan. [Mubarak, Muhammad] Sindh Institute of Urology and Transplantation, Department of HistopathologyKarachi, Pakistan. [Facista, Alex] University of Arizona, College of Medicine, Department of Cellular and Molecular MedicinePhoenix, AZ, USA. [Akhtar, Shakeel Syed] Civil Hospital Karachi, Department of Surgery and MedicineKarachi, Pakistan. [Khan, Saeed] University of Karachi, Department of Microbiology, Immunology and Infectious Diseases Research LaboratoryKarachi, Pakistan. [Kazi, Iqbal Javed] Sindh Institute of Urology and Transplantation, Department of HistopathologyKarachi, Pakistan. [Bernstein, Carol] University of Arizona, College of Medicine, Department of Cellular and Molecular MedicinePhoenix, AZ, USA. [Kazmi, Urooj Shahana] University of Karachi, Department of Microbiology, Immunology and Infectious Diseases Research LaboratoryKarachi, Pakistan. RP Mubarak, M (reprint author), Sindh Institute of Urology and Transplantation, Department of Histopathology, Karachi, Pakistan. EM drmubaraksiut@yahoo.com CR Auten RL, Davis JM, 2009, Oxygen toxicity and reactive oxygen species: the devil is in the details. Pediatr Res 66:121–127 Klaunig JE, Kamendulis LM, Hocevar BA, 2010, Oxidative stress and oxidative damage in carcinogenesis. Toxicol Pathol 38:96–109 Inokuma T, Haraguchi M, Fujita F, Tajima Y, Kanematsu T, 2009, Oxidative stress and tumor progression in colorectal cancer. Hepatogastroenterology 56:343–347 Smoot DT, Elliott TB, Verspaget HW, Jones D, Allen CR, Vernon KG et al, 2000, Influence of Helicobacter pylori on reactive oxygeninduced gastric epithelial cell injury. Carcinogenesis 21:2091–2095 Farinati F, Cardin R, Russo VM, Busatto G, Franco M, Rugge M, 2003, Helicobacter pylori CagA status, mucosal oxidative damage and gastritis phenotype: a potential pathway to cancer? Helicobacter 8:227–234 Varon C, Mosnier JF, Lehours P, Matysiak-Budnik T, Megraud F, 2009, Gastric carcinogenesis and Helicobacter pylori infection. Methods Mol Biol 511:237–265 Valavanidis A, Vlachogianni T, Fiotakis C, 2009, 8-hydroxy-2′- deoxyguanosine, 8-OHdG): a critical biomarker of oxidative stress and carcinogenesis. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 27:120–139 Kuchino Y, Mori F, Kasai H, Inoue H, Iwai S, Miura K et al, 1987, Misreading of DNA templates containing 8-hydroxydeoxyguanosine at the modified base and at adjacent residues. Nature 327:77–79 Lee SF, Pervaiz S, 2011, Assessment of oxidative stress-induced DNA damage by immunoflourescent analysis of 8-OxodG. Methods Cell Biol 103:99–113 Moller H, Heseltine E, Vainio H, 1995, Working group report on schistosomes, liver flukes and Helicobacter pylori. Int J Cancer 60: 587–589 Marshall BJ, Warren JR, 1984, Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet 1: 1311–1315 Yamaoka Y, 2008, Increasing evidence of the role of Helicobacter pylori SabA in the pathogenesis of gastroduodenal disease. J Infect Dev Ctries 2:174–181 Asahi M, Azuma T, Ito S, Ito Y, Suto H, Nagai Y et al, 2000, Helicobacter pylori CagA protein can be tyrosine phosphorylated in gastric epithelial cells. J Exp Med 191:593–602 Odenbreit S, Puls J, Sedlmaier B, Gerland E, Fischer W, Haas R, 2000, Translocation of Helicobacter pylori CagA into gastric epithelial cells by type IV secretion. Science 287:1497–1500 Danese S, Cremonini F, Armuzzi A, Candelli M, Papa A, Ojetti V et al, 2001, Helicobacter pylori CagA-positive strains affect oxygen free radicals generation by gastric mucosa. Scand J Gastroenterol 36: 247–250 Suzuki H, Suzuki M, Imaeda H, Hibi T, 2009, Helicobacter pylori and microcirculation. Microcirculation 16:547–558 Gonzalez CA, Figueiredo C, Lic CB, Ferreira RM, Pardo ML, Ruiz Liso JM et al, 2011, Helicobacter pylori cagA and vacAgenotypes as predictors of progression of gastric preneoplastic lesions: a long-term follow-up in a high-risk area in Spain. Am J Gastroenterol 106:867– 874 Blaser MJ, Perez-Perez GI, Kleanthous H, Cover TL, Peek RM, Chyou PH et al, 1995, Infection with Helicobacter pylori strains possessing cagA is associated with an increased risk of developing adenocarcinoma of the stomach. Cancer Res 55:2111–2115 Singh K, Ghoshal UC, 2006, Causal role of Helicobacter pylori infection in gastric cancer: an Asian enigma. World J Gastroenterol 12:1346–1351 Qureshi TZ, Bilal R, Saleem K, Zafar S, 2008, General prevalence of Helicobacter pylori infection in dyspeptic population of Islamabad, Pakistan. Nucleus 45:157–162 Taj Y, Essa F, Kazmi SU, Abdullah E, 2003, Sensitivity and specificity of various diagnostic tests in the detection of Helicobacter pylori. J Coll Physicians Surg Pak 13:90–93 Khan SS, Zulfiqar A, Danish KF, Sauwal M, Bashir S, Zaman S, 2008, Prevalence of H. Pylori infection in patients with gastroduodenal disease in Pakistan. Rawal Med J 33:1–6 Yakoob J, Jafri W, Jafri N, Islam M, Abid S, Hamid S et al, 2005, Prevalence of non-Helicobacter pylori duodenal ulcer in Karachi, Pakistan. World J Gastroenterol 11:3562–3565 Peek RM Jr, Blaser MJ, 2002, Helicobacter pylori and gastrointestinal tract adenocarcinomas. Nat Rev Cancer 2:28–37 Dixon MF, Genta RM, Yardley JH, Correa P, 1996, Classification and grading of gastritis. The updated Sydney System. International Workshop on the Histopathology of Gastritis, Houston 1994. Am J Surg Pathol 20:1161–1181 Khan S, Jaffer NN, Khan MN, RaiMA, Shafiq M, Ali A et al, 2007, Human papillomavirus subtype 16 is common in Pakistani women with cervical carcinoma. Int J Infect Dis 11:313–317 Ashton-Key M, Diss TC, Isaacson PG, 1996, Detection of Helicobacter pylori in gastric biopsy and resection specimens. J Clin Pathol 49:107–111 Qabandi AA,Mustafa AS, Siddique I, Khajah AK, Madda JP, Junaid TA, 2005, Distribution of vacA and cagA genotypes of Helicobacter pylori in Kuwait. Acta Trop 93:283–288 Papa A, Danese S, Sgambato A, Ardito R, Zannoni G, Rinelli A et al, 2002, Role of Helicobacter pylori CagA +infection in determining oxidative DNA damage in gastric mucosa. Scand J Gastroenterol 37: 409–413 Rasheed F, Khan A, Kazmi SU, 2009, Bacteriological analysis, antimicrobial susceptibility and detection of 16S rRNA gene of Helicobacter pylori by PCR in drinking water samples of earthquake affected areas and other parts of Pakistan. Malay J Microbiol 5:123– 127 Gao L, Michel A,Weck MN, Arndt V, Pawlita M, Brenner H, 2009, Helicobacter pylori infection and gastric cancer risk: evaluation of 15H. pylori proteins determined by novel multiplex serology. Cancer Res 69:6164–6170 Kido M, Watanabe N, Aoki N, Iwamoto S, Nishiura H, Maruoka R et al, 2011, Dual roles of CagA protein in Helicobacter pyloriinduced chronic gastritis in mice. Biochem Biophys Res Commun 412:266–272 Hatakeyama M, 2009, Helicobacter pylori and gastric carcinogenesis. J Gastroenterol 44:239–248 Konturek PC, Konturek SJ, Brzozowski T, 2009, Helicobacter pylori infection in gastric cancerogenesis. J Physiol Pharmacol 60:3–21 Davies GR, Simmonds NJ, Stevens TR, Sheaff MT, Banatvala N, Laurenson IF et al, 1994, Helicobacter pylori stimulates antral mucosal reactive oxygenmetabolite production in vivo. Gut 35:179–185 Farinati F, Cardin R, Cassaro M, BortolamiM, Nitti D, Tieppo C et al, 2008, Helicobacter pylori, inflammation, oxidative damage and gastric cancer: a morphological, biological and molecular pathway. Eur J Cancer Prev 17:195–200 Katsurahara M, Kobayashi Y, Iwasa M, Ma N, Inoue H, Fujita N et al, 2009, Reactive nitrogen species mediate DNA damage in Helicobacter pylori-infected gastric mucosa. Helicobacter 14:552– 558 Everett SM,White KL,Drake IM, Schorah CJ, Calvert RJ, Skinner C et al, 2002, The effect of Helicobacter pylori infection on levels of DNA damage in gastric epithelial cells. Helicobacter 7:271–280 Wroblewski LE, Peek RM Jr, Wilson KT, 2010, Helicobacter pylori and gastric cancer: factors that modulate disease risk. Clin Microbiol Rev 23:713–739 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 839 EP 846 DI 10.1007/s12253-014-9762-1 PG 8 ER PT J AU Xing, J Cao, G Fu, Ch AF Xing, Junjie Cao, Guangwen Fu, Chuangang TI HMGA1 Interacts with β-Catenin to Positively Regulate Wnt/β-Catenin Signaling in Colorectal Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; HMGA1; β-catenin; Wnt signaling pathway ID Colorectal cancer; HMGA1; β-catenin; Wnt signaling pathway AB The high mobility group A1 (HMGA1) protein plays an important role in numerous biological processes, such as embryogenesis, cell proliferation, differentiation, apoptosis and carcinogenesis.Wnt/β-catenin signaling pathway plays a key role in development and cancer. Although previous reports have shown HMGA1 protein level can be induced by Wnt/β-catenin signaling pathway, however, the specific mechanism of HMGA1 on regulating Wnt/β-catenin signaling remains unclear. Here, we reported that HMGA1 interacted with β-catenin by using coimmunoprecipitation approach with exogenous and endogenous protein samples. HMGA1 positively regulated Wnt/β-catenin signaling, as determined by that HMGA1 increased the TOP-FLASH activity in a dose-dependent manner and β-catenin downstream target gene expression. Moreover, HMGA1 induced proliferation of colorectal cancer cells. Mechanistically, HMGA1 increased the β-catenin–TCF4 complex formation. Importantly, there was a correlation between HMGA1 and β-catenin expression in human colorectal cancer tissues. In summary, HMGA1 positively regulates Wnt/β-catenin signaling through interacting with β-catenin, which leads to increase the β-catenin–TCF4 complex formation. This suggests that targeting HMGA1 may be a useful therapeutic option in clinical application. C1 [Xing, Junjie] Second Military Medical University, Changhai Hospital, Department of Colorectal SurgeryShanghai, China. [Cao, Guangwen] Second Military Medical University, Department of EpidemiologyShanghai, China. [Fu, Chuangang] Second Military Medical University, Changhai Hospital, Department of Colorectal SurgeryShanghai, China. RP Fu, Ch (reprint author), Second Military Medical University, Changhai Hospital, Department of Colorectal Surgery, Shanghai, China. EM fugang416@126.com CR Fusco A, Fedele M, 2007, Roles of HMGA proteins in cancer. Nat Rev Cancer 7:899–910 Shah SN, Kerr C, Cope L et al, 2012, HMGA1 reprograms somatic cells into pluripotent stem cells by inducing stem cell transcriptional networks. PLoS One 7:e48533 Di Cello F, Shin J, Harbom K, Brayton C, 2013, Knockdown of HMGA1 inhibits human breast cancer cell growth and metastasis in immunodeficient mice. Biochem Biophys Res Commun 434:70–74 Chiappetta G, Ottaiano A, Vuttariello E et al, 2010, HMGA1 protein expression in familial breast carcinoma patients. Eur J Cancer 46: 332–339 Takeuchi I, Takaha N, Nakamura T et al, 2012, High mobility group protein AT-hook 1, HMGA1, is associated with the development of androgen independence in prostate cancer cells. Prostate 72:1124– 1132 Pang B, Fan H, Zhang IYet al, 2012, HMGA1 expression in human gliomas and its correlation with tumor proliferation, invasion and angiogenesis. J Neurooncol 106:543–549 Hillion J, Smail SS, Di Cello F et al, 2012, The HMGA1-COX-2 axis: a key molecular pathway and potential target in pancreatic adenocarcinoma. Pancreatology 12:372–379 Nezhad MH, Drieschner N, Helms S et al, 2010, 6p21 rearrangements in uterine leiomyomas targeting HMGA1. Cancer Genet Cytogenet 203:247–252 BushBM, Brock AT, Deng JA, Nelson RA Jr, Sumter TF, 2013, The Wnt/β-catenin/T-cell factor 4 pathway up-regulates high-mobility group A1 expression in colon cancer. Cell BiochemFunct 31:228– 236 Zhang Y, Ma T, Yang S et al, 2011, High-mobility group A1 proteins enhance the expression of the oncogenic miR-222 in lung cancer cells. Mol Cell Biochem 357:363–371 Clevers H, Nusse R, 2012)Wnt/β-catenin signaling and disease. Cell 149:1192–1205 MacDonald BT, Tamai K, He X, 2009, Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell 17:9–26 Rosenbluh J, Nijhawan D, 2012, Cox AG: β-Catenin-driven cancers require a YAP1 transcriptional complex for survival and tumorigenesis. Cell 151:1457–1473 Akaboshi S,Watanabe S, Hino Yet al, 2009, HMGA1 is induced by Wnt/beta-catenin pathway and maintains cell proliferation in gastric cancer. Am J Pathol 175:1675–1685 KonsavageWMJr, Kyler SL, Rennoll SA, Jin G, Yochum GS, 2012, Wnt/β-catenin signaling regulates Yes-associated protein, YAP, gene expression in colorectal carcinoma cells. J Biol Chem 287:11730– 11739 Guo RJ, Funakoshi S, Lee HH, Kong J, Lynch JP, 2010, The intestine-specific transcription factor Cdx2 inhibits beta-catenin/ TCF transcriptional activity by disrupting the beta-catenin-TCF protein complex. Carcinogenesis 31:159–166 Emami KH, Nguyen C,Ma H et al, 2004, A small molecule inhibitor of beta-catenin/CREB-binding protein transcription. Proc Natl Acad Sci U S A 101:12682–12687 Shah SN, Resar LM, 2012, High mobility group A1 and cancer: potential biomarker and therapeutic target. Histol Histopathol 27: 567–579 Giannini G, Cerignoli F, Mellone M, Massimi I, Ambrosi C, Rinaldi C, Gulino A, 2005, Molecular mechanism of HMGA1 deregulation in human neuroblastoma. Cancer Lett 228:97–104 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 847 EP 851 DI 10.1007/s12253-014-9763-0 PG 5 ER PT J AU Huang, P Cao, K Zhao, H AF Huang, Ping Cao, Kejian Zhao, Heng TI Screening of Critical Genes in Lung Adenocarcinoma via Network Analysis of Gene Expression Profile SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung adenocarcinoma; Gene expression profile; Differentially expressed gene; Gene expression core signature; Functional enrichment analysis; Network clustering ID Lung adenocarcinoma; Gene expression profile; Differentially expressed gene; Gene expression core signature; Functional enrichment analysis; Network clustering AB Biomarker discovery is of great importance in diagnosis and treatment of diseases. In present study, a number of differentially expressed genes (DEGs) were identified for lung adenocarcinoma via comparative analysis of gene expression data. A gene expression core signature was generated for four types of lung adenocarcinoma (EGFR-mutated, KRAS-mutated, ALK-mutated and triple-negative adenocarcinoma). Functional enrichment analysis with DAVID tools revealed that up-regulated genes were mainly associated with cell cycle while down-regulated genes were mainly involved in vasculature development and cell adhesion. Then it was used to retrieve relevant small molecule drugs with Connectivity map and trichostatin A was predicted to be the top candidate drug for treatment of lung cancer. Network clustering was performed with MCL in cytoscape to identify sub-networks and several hub genes were obtained: CDC25C, ICT1, TK1 and EZH2. These genes play important roles in the progression of lung cancer and some have been suggested as potential biomarkers. Therefore, our findings are beneficial in deepening the understandings about the pathogenesis and providing directions for future researches. C1 [Huang, Ping] Shanghai Jiao Tong University, Shanghai Chest Hospital, Department of Thoracic Surgery, NO.241 Huaihaixi Road, 200030 Shanghai, China. [Cao, Kejian] Shanghai Jiao Tong University, Shanghai Chest Hospital, Department of Thoracic Surgery, NO.241 Huaihaixi Road, 200030 Shanghai, China. [Zhao, Heng] Shanghai Jiao Tong University, Shanghai Chest Hospital, Department of Thoracic Surgery, NO.241 Huaihaixi Road, 200030 Shanghai, China. RP Zhao, H (reprint author), Shanghai Jiao Tong University, Shanghai Chest Hospital, Department of Thoracic Surgery, 200030 Shanghai, China. EM H_zhang28@163.com CR Bronte G, Rizzo S, La Paglia L, Adamo V, Siragusa S, Ficorella C, Santini D, Bazan V, Colucci G, Gebbia N, Russo A, 2010, Driver mutations and differential sensitivity to targeted therapies: a new approach to the treatment of lung adenocarcinoma. Cancer Treat Rev 36(Suppl 3):S21–29., DOI 10.1016/S0305-7372(10)70016-5 Gerber DE,Minna JD, 2010, ALK inhibition for non-small cell lung cancer: from discovery to therapy in record time. Cancer Cell 18(6): 548–551 Herbst RS, Heymach JV, Lippman SM(2008, Lung cancer. N Engl J Med 359(13):1367–1380., DOI 10.1056/NEJMra0802714 Janku F, Stewart DJ, Kurzrock R, 2010, Targeted therapy in nonsmall- cell lung cancer–is it becoming a reality? Nat Rev Clin Oncol 7(7):401–414., DOI 10.1038/nrclinonc.2010.64 PaoW, Girard N, 2011, New driver mutations in non-small-cell lung cancer. Lancet Oncol 12(2):175–180., DOI 10.1016/S1470-2045(10, 70087-5 Dai M, Wang P, Boyd AD, Kostov G, Athey B, Jones EG, Bunney WE, Myers RM, Speed TP, Akil H, Watson SJ, Meng F, 2005, Evolving gene/transcript definitions significantly alter the interpretation of GeneChip data. Nucleic Acids Res 33(20):e175 Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP, 2003, Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4(2):249–264., DOI 10.1093/biostatistics/4.2.249 da Huang W, Sherman BT, Lempicki RA, 2009, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4(1):44–57 Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, Reich M, Hieronymus H, Wei G, Armstrong SA, Haggarty SJ, Clemons PA, Wei R, Carr SA, Lander ES, Golub TR, 2006, The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313(5795):1929–1935 Licata L, Briganti L, Peluso D, Perfetto L, Iannuccelli M, Galeota E, Sacco F, Palma A, Nardozza AP, Santonico E, Castagnoli L, Cesareni G, 2012, MINT, the molecular interaction database: 2012 update. Nucleic Acids Res 40(Database issue):D857–861., DOI 10.1093/nar/ gkr930 Chatr-Aryamontri A, Breitkreutz BJ, Heinicke S, Boucher L, Winter A, Stark C, Nixon J, Ramage L, Kolas N, O’Donnell L, Reguly T, Breitkreutz A, Sellam A, Chen D, Chang C, Rust J, Livstone M, Oughtred R, Dolinski K, Tyers M, 2013, The BioGRID interaction database: 2013 update.NucleicAcids Res 41(D1):D816–823., DOI 10. 1093/nar/gks1158 Keshava Prasad TS, Goel R, Kandasamy K, Keerthikumar S, Kumar S, Mathivanan S, Telikicherla D, Raju R, Shafreen B, Venugopal A, Balakrishnan L, Marimuthu A, Banerjee S, Somanathan DS, Sebastian A, Rani S, Ray S, Harrys Kishore CJ, Kanth S, Ahmed M, Kashyap MK, Mohmood R, Ramachandra YL, Krishna V, Rahiman BA, Mohan S, Ranganathan P, Ramabadran S, Chaerkady R, Pandey A, 2009, Human Protein Reference Database–2009 update. Nucleic Acids Res 37(Database issue):D767–772., DOI 10.1093/ nar/gkn892 Enright AJ, Van Dongen S, Ouzounis CA, 2002, An efficient algorithm for large-scale detection of protein families. Nucleic Acids Res 30(7):1575–1584 Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T, 2003, Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13(11):2498–2504 Zhang QC, Jiang SJ, Zhang S, Ma XB, 2012, Histone deacetylase inhibitor trichostatin A enhances anti-tumor effects of docetaxel or erlotinib in A549 cell line. Asian Pac J Cancer Prev 13(7):3471–3476 Henry CJ, 2010, Biomarkers in veterinary cancer screening: applications, limitations and expectations. Vet J 185(1):10–14 Chen ZH, Huang SQ, Wang Y, Yang AZ, Wen J, Xu XH, Chen Y, Chen QB,Wang YH, He E, 2011, Serological thymidine kinase 1 is a biomarker for early detection of tumours—a health screening study on 35,365 people, using a sensitive chemiluminescent dot blot assay. Sensors 11(12):11064–11080 Korkmaz T, Seber S, Okutur K, Basaran G, Yumuk F, Dane F, Ones T, Polat O, Madenci OC, Demir G, Turhal NS, 2013, Serum thymidine kinase 1 levels correlates with FDG uptake and prognosis in patients with non small cell lung cancer. Biomarkers 18(1):88–94., DOI 10.3109/1354750X.2012.738250 Xu Y, Shi QL,Ma H, Zhou H, Lu Z,Yu B, Zhou X, Eriksson S,He E, Skog S, 2012, High thymidine kinase 1, TK1, expression is a predictor of poor survival in patients with pT1 of lung adenocarcinoma. Tumour Biol 33(2):475–483., DOI 10.1007/s13277-011-0276-0 Peng C-Y, Graves PR, Thoma RS,Wu Z, Shaw AS, Piwnica-Worms H, 1997, Mitotic and G2 checkpoint control: regulation of 14-3-3 protein binding by phosphorylation of Cdc25C on serine-216. Science 277(5331):1501–1505 Krause K, Haugwitz U, Wasner M, Wiedmann M, Mossner J, Engeland K, 2001, Expression of the cell cycle phosphatase cdc25C is down-regulated by the tumor suppressor protein p53 but not by p73. Biochem Biophys Res Commun 284(3):743–750 Carmazzi Y, Iorio M, Armani C, Cianchetti S, Raggi F, Neri T, Cordazzo C, Petrini S, Vanacore R, Bogazzi F, Paggiaro P, Celi A, 2012, The mechanisms of nadroparin-mediated inhibition of proliferation of two human lung cancer cell lines. Cell Prolif 45(6):545– 556., DOI 10.1111/j.1365-2184.2012.00847.x Li QQ, Wang G, Huang F, Li JM, Cuff CF, Reed E, 2013, Sensitization of lung cancer cells to cisplatin by beta-elemene is mediated through blockade of cell cycle progression: antitumor efficacies of beta-elemene and its synthetic analogs. Med Oncol 30(1):488., DOI 10.1007/s12032-013-0488-9 van Belzen N, Diesveld MP, van der Made AC, Nozawa Y, Dinjens WN, Vlietstra R, Trapman J, Bosman FT, 1995, Identification of mRNAs that showmodulated expression during colon carcinoma cell differentiation. Eur J Biochem 234(3):843–848 van Belzen N, Dinjens WN, Eussen BH, Bosman FT, 1998, Expression of differentiation-related genes in colorectal cancer: possible implications for prognosis. HistolHistopathol 13(4):1233–1242 Handa Y, Hikawa Y, Tochio N, Kogure H, Inoue M, Koshiba S, Guntert P, Inoue Y, Kigawa T, Yokoyama S, Nameki N, 2010, Solution structure of the catalytic domain of the mitochondrial protein ICT1 that is essential for cell vitality. JMol Biol 404(2):260–273., DOI 10.1016/j.jmb.2010.09.033 Richter R, Rorbach J, Pajak A, Smith PM,Wessels HJ, Huynen MA, Smeitink JA, Lightowlers RN, Chrzanowska-Lightowlers ZM(2010, A functional peptidyl-tRNA hydrolase, ICT1, has been recruited into the human mitochondrial ribosome. EMBO J 29(6):1116–1125., DOI 10.1038/emboj.2010.14 Bachmann IM, Halvorsen OJ, Collett K, Stefansson IM, Straume O, Haukaas SA, Salvesen HB, Otte AP, Akslen LA, 2006, EZH2 expression is associated with high proliferation rate and aggressive tumor subgroups in cutaneous melanoma and cancers of the endometrium, prostate, and breast. J Clin Oncol 24(2):268–273 Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Tomlins SA, Ghosh D, Sewalt RG, Otte AP, Hayes DF, 2003, EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci U S A 100(20):11606– 11611 Weikert S, Christoph F, Kollermann J,MullerM, SchraderM, Miller K, Krause H, 2005, Expression levels of the EZH2 polycomb transcriptional repressor correlate with aggressiveness and invasive potential of bladder carcinomas. Int J Mol Med 16(2):349 CaoW, Ribeiro Rde O, Liu D, Saintigny P, Xia R, Xue Y, Lin R,Mao L, Ren H, 2012, EZH2 promotes malignant behaviors via cell cycle dysregulation and its mRNA level associates with prognosis of patient with non-small cell lung cancer. PLoS One 7(12):e52984 Lin YW, Ren LL, Xiong H, DuW, Yu YN, Sun TT,Weng YR,Wang ZH,Wang JL,Wang YC, Cui Y, Sun DF, Han ZG, Shen N, ZouWP, Xu J, Chen HY, Cao WB, Hong J, Fang JY, 2013, Role of STAT3 and Vitamin D Receptor in EZH2-mediated invasion of human colorectal cancer. J Pathol., DOI 10.1002/path.4179 Zhang J-G, Guo J-F, Liu D-L, Liu Q, Wang J-J, 2011, MicroRNA- 101 exerts tumor-suppressive functions in non-small cell lung cancer through directly targeting enhancer of zeste homolog 2. J Thorac Oncol 6(4):671–678 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 853 EP 858 DI 10.1007/s12253-014-9764-z PG 6 ER PT J AU Shabayek, IM Sayed, MO Attaia, AH Awida, AH Abozeed, H AF Shabayek, I Marwa Sayed, M Ola Attaia, A Hanan Awida, A Heba Abozeed, Hamdy TI Diagnostic Evaluation of Urinary Angiogenin (ANG) and Clusterin (CLU) as Biomarker for Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder; Cancer; Cytology; Angiogenin; Clusterin; Sensitivity ID Bladder; Cancer; Cytology; Angiogenin; Clusterin; Sensitivity AB Bladder carcinoma is an important worldwide health problem. Both cystoscopy and urine cytology used in detecting bladder cancer suffer from drawbacks where cystoscopy is an invasive method and urine cytology shows low sensitivity in low grade tumors. This study validates easier and less time-consuming techniques to evaluate the value of combined use of angiogenin and clusterin in comparison and combination with voided urine cytology in the detection of bladder cancer patients. This study includes malignant (bladder cancer patients, n=50), benign (n=20) and healthy (n=20) groups. The studied groups were subjected to cystoscopic examination, detection of bilharzial antibodies, urine cytology, and estimation of urinary angiogenin and clusterin by ELISA. The overall sensitivity and specificity were 66 and 75 % for angiogenin, 70 and 82.5 % for clusterin and 46 and 80 % for voided urine cytology. Combined sensitivity of voided urine cytology with the two studied biomarkers was 88 % which is higher than the combined sensitivity of both markers alone (82 %) and that of the cytology with each marker (76 and 80 %) for angiogenin and clusterin respectively. In conclusion, combined use of the cytology with the studied biomarkers can improve the sensitivity for detecting bladder cancer, and may be very useful in monitoring the effectiveness of antiangiogenic and apoptotic therapies in bladder cancer. C1 [Shabayek, I Marwa] Future University, Faculty of Pharmaceutical Sciences and Pharmaceutical Technologies, Department of BiochemistryCairo, Egypt. [Sayed, M Ola] Azhar University, Faculty of Pharmacy, Department of BiochemistryCairo, Egypt. [Attaia, A Hanan] Azhar University, Faculty of Pharmacy, Department of BiochemistryCairo, Egypt. [Awida, A Heba] Future University, Faculty of Pharmaceutical Sciences and Pharmaceutical Technologies, Department of BiochemistryCairo, Egypt. [Abozeed, Hamdy] Azhar University, Faculty of Medicine, Urology DepartmentCairo, Egypt. RP Shabayek, IM (reprint author), Future University, Faculty of Pharmaceutical Sciences and Pharmaceutical Technologies, Department of Biochemistry, Cairo, Egypt. EM biochemistry.fue@gmail.com CR American Cancer Society, 2013, Cancer facts and figures 2013. American Cancer Society, Atlanta Zaghloul S, 2012, Bladder cancer and schistosomiasis. J Egypt Natl Cancer Inst 24:151–159 Eissa S, Shabayek M, Ismail M, Abd-Elateef A, 2010, Diagnostic evaluation of apoptosis inhibitory gene and tissue inhibitor matrix metalloproteinase-2 in patients with bladder cancer. IUBMB Life 62(5):394–399 Urquidi V, Goodison S, Kim J, Chang M, Dai Y, Rosser CJ, 2012, Vascular enothelail growth factor, carbonic anhydrase 9, and angiogenin as urinary biomarkers for bladder cancer detection. Urology 79(5):1185.e1–1185.e6 Mitra A, 2010, Urine cytologic analysis: special techniques for bladder cancer detection. In: Connection. 169–177 Yafi A, Brimo F, Auger M, Aprikian A, Tanguay S, Kassouf W, 2013, Is the performance of urinary cytology as high as reported historically? A contemporary analysis in the detection and surveillance of bladder cancer. Urol Oncol Semin Orig Investig 32(1):27.e1–6. Budman L, Kassouf W, Steinberg JR, 2008, Biomarkers for detection and surveillance of bladder cancer. Can Urol Assoc J 2:212–221 Li S, Hu G, 2010, Angiogenin-mediated rRNA transcription in cancer and neurodegeneration. Int J Biochem Mol Biol 1(1):26–35 Eissa S, Swellam M, Labib RA, El-Zayat T, El Ahmady O, 2009, A panel of angiogenic factors for early bladder cancer detection: enzyme immunoassay and Western blot. J Urol 181(3):1353–1360 Hazzaa M, Elashry M, Afifi K, 2010, Clusterin as a diagnostic and prognostic marker for transitional cell carcinoma of the bladder. Pathol Oncol Res 16(1):101–109 Miyake H, Hara I, Gleave M, Eto H, 2004, Protection of androgen dependent human prostate cancer cells from oxidative stressinduced DNA damage by overexpression of clusterin and its modulation by androgen. Prostate 61:318–323 American Joint Committee on Cancer, 2010, AJCC cancer staging manual. Urinary bladder, 7th edn. Springer, New York, pp 497–502 Felix AS, Soliman AS, Khaled H, Zaghloul MS, Banerjee M, El-Baradie M, 2008, The changing patterns of bladder cancer in Egypt over the past 26 years. Cancer Causes Control 19(4): 421–429 ZhengYL,Amr S, Saleh DA, Dash C, Ezzat S,MikhailNN,Gouda I, Loay I, Hifnawy T, Abdel-Hamid M, Khaled H, Wolpert B, Abdel- Aziz MA, Loffredo CA, 2012, Urinary bladder cancer risk factors in Egypt: a multicenter case–control study. Cancer Epidemiol Biomark Prev 21(3):537–546 SalemHK,Mahfouz S, 2012, Changing patterns, age, incidence, and pathologic types, of schistosoma-associated bladder cancer in Egypt in the past decade. Urology 79(2):379–383 Ahmedin J, Ram CT, TaylorM, Asma G, Alicia S, ElizabethW, Eric JF, Michael JT, 2004, American Cancer Society. Bladder Cancer J Clin 96:54–56 Burger M, Catto JW, Dalbagni G, Grossman HB, Herr H, Karakiewicz P, Kassouf W, Kiemeney LA, La Vecchia C, Shariat S, Lotan Y, 2013, Epidemiology and risk factors of urothelial bladder cancer. Eur Urol 63(2):234–241 Gho YS et al, 2002, Antiplasmin activity of a peptide that binds to the receptor-binding site of angiogenin. J Biol Chem 277(12):9690–9694 Yang Y, Sun M, Wang L, Jiao B, 2013, HIFs, angiogenesis, and cancer. J Cell Biochem 114:967–974 Wong P,Ulyanova T, Organisciak D, Bennett S, Lakins J,Arnold JM, Kutty RK, Tenniswood M, vanVeen T,DarrowRM, Chader G, 2001, Expression of multiple forms of clusterin during light-induced retinal degeneration. Curr Eye Res 23:157–165 Trougakso I, So A, Jansen B, Gleave ME, Gonos ES, 2004, Silencing expression of the clusterin/apolipoprotein J gene in human cancer cells using small interfering RNA induces spontaneous apoptosis, reduced growth ability, and cell sensitization to genotoxic and oxidative stress. Cancer Res 64:1834–1842 Leskov K, Klokov D, Li J, Boothman DA, 2003, Synthesis and functional analyses of nuclear clusterin, a cell death protein. J Biol Chem 278:11590–11600 Stejskal D, Fiala R, 2006, Evaluation of serum and urine clusterin as a potential tumor marker for urinary bladder cancer. Neoplasma 53(4):343–346 Kim N, Yoo J, Han J, 2012, Human nuclear clusterin mediates apoptosis by interacting with bcl-xl through c-terminal coiled coil domain. J Cell Physiol 227:1157–1167 Kruger S,Mahnken A, Kausch I, Feller AC, 2006, Value of clusterin immunoreactivity as a predictive factor in muscle-invasive urothelial bladder carcinoma. Urology 67(1):105–109 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 859 EP 866 DI 10.1007/s12253-014-9765-y PG 8 ER PT J AU Sipos, F Muzes, Gy Furi, I Spisak, S Wichmann, B Germann, MT Constantinovits, M Krenacs, T Tulassay, Zs Molnar, B AF Sipos, Ferenc Muzes, Gyorgyi Furi, Istvan Spisak, Sandor Wichmann, Barnabas Germann, M Tiana Constantinovits, Miklos Krenacs, Tibor Tulassay, Zsolt Molnar, Bela TI Intravenous Administration of a Single-Dose Free-Circulating DNA of Colitic Origin Improves Severe Murine DSS-Colitis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Free-circulating DNA; DSS-colitis; Toll-like receptor 9; Proinflammatory cytokines; Tissue regeneration ID Free-circulating DNA; DSS-colitis; Toll-like receptor 9; Proinflammatory cytokines; Tissue regeneration AB In inflammatory bowel diseases the presence of free-circulating DNA (fcDNA) sequences in the sera is an established phenomenon, albeit its real biological function still remains unclear. In our study the immunobiologic effects of a single-dose, intravenously administered fcDNA of normal and colitic origin were assayed in DSS-colitic and control mice. In parallel with disease and histological activity evaluations changes of the TLR9 and inflammatory cytokine signaling gene expression profiles were assayed in isolated cells of the lamina propria. Intravenously administered colitisderived fcDNA displayed a more prominent beneficial action regarding the clinical and histological severity of DSS-colitis than that of fcDNA of normal origin. Systemic administration of colitis-derived fcDNA significantly altered the expression of certain TLR9-related and proinflammatory cytokine genes in a clinically favorable manner. Presumably due to induction of severe colitis, the subsequent marked inflammatory environment may result changes in fcDNA with a potential to promote the downregulation of inflammation and improvement of tissue regeneration. Elucidating mechanisms of innate immune alterations by nucleic acids may provide further insight into the etiology of inflammatory bowel diseases, and develop the basis of novel nucleic acid-based immunotherapies. C1 [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Muzes, Gyorgyi] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Furi, Istvan] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Spisak, Sandor] Harvard Medical School, Dana-Farber Cancer Institute, Department of Medical OncologyBoston, MA, USA. [Wichmann, Barnabas] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Germann, M Tiana] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Constantinovits, Miklos] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Street 46, 1088 Budapest, Hungary. RP Sipos, F (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM dr.siposf@gmail.com CR van Dieren JM, Kuipers EJ, Samsom JN et al, 2006, Revisiting the immunomodulators tacrolimus, methotrexate, and mycophenolate mofetil: their mechanisms of action and role in the treatment of IBD. Inflamm Bowel Dis 12:311–327 Tan EM, Schur PH, Carr RI et al, 1966, Deoxybonucleic acid, DNA, and antibodies to DNA in the serum of patients with systemic lupus erythematosus. J Clin Invest 11:1732–1740 Leon SA, Shapiro B, Sklaroff DM et al, 1977, Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 3:646– 650 Kohler C, Barekati Z, Radpour R et al, 2011, Cell-free DNA in the circulation as a potential cancer biomarker. Anticancer Res 8:2623– 2628 Steinman CR, 1975, Free DNA in serum and plasma from normal adults. J Clin Invest 56:512–515 Rauh P, Rickes S, Fleischhacker M, 2003, Microsatellite alterations in free-circulating serum DNA in patients with ulcerative colitis. Dig Dis 21:363–366 Swarup V, Rajeswari MR, 2007, Circulating, cell-free, nucleic acids—a promising, non-invasive tool for early detection of several human diseases. FEBS Lett 581:795–799 Gosu V, Basith S, Kwon OP et al, 2012, Therapeutic applications of nucleic acids and their analogues in Toll-like receptor signaling. Molecules 17:13503–13529 Nierkens S, den Brok MH, Garcia Z et al, 2011, Immune adjuvant efficacy of CpG oligonucleotide in cancer treatment is founded specifically upon TLR9 function in plasmacytoid dendritic cells. Cancer Res 71:6428–6437 Liang X, Moseman EA, Farrar MA et al, 2010, Toll-like receptor 9 signaling by CpG-B oligodeoxynucleotides induces an apoptotic pathway in human chronic lymphocytic leukemia B cells. Blood 115:5041–5052 Sipos F, Muzes G, 2011, Isolated lymphoid follicles in colon: switch points between inflammation and colorectal cancer? World J Gastroenterol 17:1666–1673 Dieleman LA, Ridwan BU, TennysonGS et al, 1994, Dextran sulfate sodium-induced colitis occurs in severe combined immunodeficient mice. Gastroenterology 107:1643–1652 Egger B, Bajaj-Elliott M, MacDonald TT et al, 2000, Characterisation of acute murine dextran sodium sulphate colitis: cytokine profile and dose dependency. Digestion 62:240–248 Rachmilewitz D, Karmeli F, Takabayashi K et al, 2002, Immunostimulatory DNA ameliorates experimental and spontaneous murine colitis. Gastroenterology 122:1428–1441 CiorbaMA, Bettonville EE, McDonald KG et al, 2010, Induction of IDO-1 by immunostimulatory DNA limits severity of experimental colitis. J Immunol 184:3907–3916 Dou S, Smith M, Wang Yet al, 2013, Intraperitoneal injection is not always a suitable alternative to intravenous injection for radiotherapy. Cancer Biother Radiopharm Musch E, Lutfi T, von Stein P et al, 2013, Topical treatment with the toll-like receptor agonist DIMS0150 has potential for lasting relief of symptoms in patients with chronic active ulcerative colitis by restoring glucocorticoid sensitivity. Inflamm Bowel Dis 19:283–292 Murthy SN, Cooper HS, Shim H et al, 1993, Treatment of dextran sulfate sodium-induced murine colitis by intracolonic cyclosporin. Dig Dis Sci 38:1722–1734 te Velde AA, VerstegeMI, Hommes DW(2006, Critical appraisal of the current practice in murine TNBS-induced colitis. Inflamm Bowel Dis 12:995–999 Duijvestein M,WildenbergME,WellingMMet al, 2011, Pretreatment with interferon-γ enhances the therapeutic activity of mesenchymal stromal cells in animal models of colitis. Stem Cells 29:1549–1558 Lee JS, Oka K, ObaraMet al, 2009, Improved isolation methods for mucosal leukocytes from small and large intestines in rats. Biosci Biotechnol Biochem 73:1732–1740 Hart AL, Al-Hassi HO, Rigby RJ et al, 2005, Characteristics of intestinal dendritic cells in inflammatory bowel diseases. Gastroenterology 129:50–65 Howe R, Dillon S, Rogers L et al, 2009, Evidence for dendritic celldependent CD4(+, T helper-1 type responses to commensal bacteria in normal human intestinal lamina propria. Clin Immunol 131:317–332 StensonWF, 2008, Toll-like receptors and intestinal epithelial repair. Curr Opin Gastroenterol 24:103–107 Ishii KJ, Akira S, 2005, Innate immune recognition of nucleic acids: beyond toll-like receptors. Int J Cancer 117:517–523 Ishii KJ, Gursel I, GurselMet al, 2004, Immunotherapeutic utility of stimulatory and suppressive oligodeoxynucleotides. Curr Opin Mol Ther 6:166–174 Krieg AM, Wu T, Weeratna R et al, 1998, Sequence motifs in adenoviral DNA block immune activation by stimulatory CpG motifs. Proc Natl Acad Sci U S A 95:12631–12636 Xavier RJ, Podolsky DK, 2007, Unravelling the pathogenesis of inflammatory bowel disease. Nature 448:427–434 Rachmilewitz D, Katakura K, Karmeli F et al, 2004, Toll-like receptor 9 signalingmediates the anti-inflammatory effects of probiotics in murine experimental colitis. Gastroenterology 126:520–528 Gormally E, Caboux E, Vineis P et al, 2007, Circulating free DNA in plasma or serum as biomarker of carcinogenesis: practical aspects and biological significance. Mutat Res 635:105–117 Stroun M, Maurice P, Vasioukhin V et al, 2000, The origin and mechanism of circulating DNA. Ann N YAcad Sci 906:161–168 Model F, Osborn N, Ahlquist D et al, 2007, Identification and validation of colorectal neoplasia-specific methylation markers for accurate classification of disease. Mol Cancer Res 5:153–163 Li L, Choi JY, Lee KM et al, 2012, DNA methylation in peripheral blood: a potential biomarker for cancer molecular epidemiology. J Epidemiol 22:384–394 Toth K, Sipos F, Kalmar A et al, 2012, Detection of methylated SEPT9 in plasma is a reliable screening method for both left- and right-sided colon cancers. PLoS One 7:e46000 Philipp AB, Stieber P, Nagel D et al, 2012, Prognostic role of methylated free circulating DNA in colorectal cancer. Int J Cancer 131:2308–2319 Gorges TM, Schiller J, Schmitz A et al, 2012, Cancer therapy monitoring in xenografts by quantitative analysis of circulating tumor DNA. Biomarkers 17:498–506 Muzes G,Molnar B, Tulassay Z et al, 2012, Changes of the cytokine profile in inflammatory bowel diseases. World J Gastroenterol 18: 5848–5861 Dieleman LA, Palmen MJ, Akol H et al, 1998, Chronic experimental colitis induced by dextran sulphate sodium, DSS, is characterized by Th1 and Th2 cytokines. Clin Exp Immunol 114:385–391 Rykova EY, Morozkin ES, Ponomaryova AA et al, 2012, Cell-free and cell-bound circulating nucleic acid complexes: mechanisms of generation, concentration and content. Expert Opin Biol Ther 12(Suppl 1):S141–S153 Iriyama C, Tomita A, Hoshino H et al, 2012, Using peripheral blood circulating DNAs to detect CpG global methylation status and genetic mutations in patients with myelodysplastic syndrome. Biochem Biophys Res Commun 419:662–669 Constantinovits M, Sipos F, Molnar B et al, 2012, Organizer and regulatory role of colonic isolated lymphoid follicles in inflammation. Acta Physiol Hung 99:344–352 Saleh M, Trinchieri G, 2011, Innate immune mechanisms of colitis and colitis-associated colorectal cancer. Nat Rev Immunol 11:9–20 Muroi M, Tanamoto K, 2008, TRAF6 distinctively mediates MyD88- and IRAK-1-induced activation of NF-kappaB. J Leukoc Biol 83:702–707 Kawagoe T, Sato S, Matsushita K et al, 2008, Sequential control of Toll-like receptor-dependent responses by IRAK1 and IRAK2. Nat Immunol 9:684–691 Kawai T, Akira S, 2007, Antiviral signaling through pattern recognition receptors. J Biochem 141:137–145 Zhu J, Mohan C, 2010, Toll-like receptor signaling pathways— therapeutic opportunities. Mediat Inflamm 2010:781235 Takeda K, Kaisho T, Akira S, 2003, Toll-like receptors. Annu Rev Immunol 21:335–376 Berglund M, Thomas JA, Fredin MF et al, 2009, Gender dependent importance of IRAK-1 in dextran sulfate sodium induced colitis. Cell Immunol 259:27–32 Honda K, Yanai H, Mizutani T et al, 2004, Role of a transductionaltranscriptional processor complex involving MyD88 and IRF-7 in Toll-like receptor signaling. Proc Natl Acad Sci U S A 101:15416–15421 Honda K, Yanai H, Negishi H et al, 2005, IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature 434:772–777 Uematsu S, Sato S, YamamotoMet al, 2005, Interleukin-1 receptorassociated kinase-1 plays an essential role for Toll-like receptor, TLR)7- and TLR9-mediated interferon-{alpha} induction. J Exp Med 201:915–923 Nenci A, Becker C, Wullaert A et al, 2007, Epithelial NEMO links innate immunity to chronic intestinal inflammation. Nature 446:557–561 Moss BL, Elhammali A, Fowlkes T et al, 2012, Interrogation of inhibitor of nuclear factor κB α/nuclear factor κB, IκBα/NF-κB, negative feedback loop dynamics: from single cells to live animals in vivo. J Biol Chem 287:31359–31370 Lee J, Mo JH, Katakura K et al, 2006, Maintenance of colonic homeostasis by distinctive apical TLR9 signalling in intestinal epithelial cells. Nat Cell Biol 8:1327–1336 Katakura K, Lee J, Rachmilewitz D et al, 2005, Toll-like receptor 9- induced type I IFN protects mice from experimental colitis. J Clin Invest 115:695–702 Sivakumar PV,Westrich GM, Kanaly S et al, 2002, Interleukin 18 is a primary mediator of the inflammation associated with dextran sulphate sodium induced colitis: blocking interleukin 18 attenuates intestinal damage. Gut 50:812–820 Takagi H, Kanai T, Okazawa A et al, 2003, Contrasting action of IL- 12 and IL-18 in the development of dextran sodiumsulphate colitis in mice. Scand J Gastroenterol 38:837–844 Toth K, Galamb O, Spisak S et al, 2009, Free circulating DNA based colorectal cancer screening from peripheral blood: the possibility of the methylated septin 9 gene marker. Orv Hetil 150:969–977 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 867 EP 877 DI 10.1007/s12253-014-9766-x PG 11 ER PT J AU Aoki, J Tsubokura, M Kakihana, K Oshikawa, G Kobayashi, T Doki, N Sakamaki, H Ohashi, K AF Aoki, Jun Tsubokura, Masaharu Kakihana, Kazuhiko Oshikawa, Gaku Kobayashi, Takeshi Doki, Noriko Sakamaki, Hisashi Ohashi, Kazuteru TI The Predictive Value for Pulmonary Infection by Area Over the Neutrophil Curve (D-index) in Patients Who Underwent Reduced Intensity Hematopoietic Stem Cell Transplantation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE D-index; cD-index; D21-index; Pulmonary infection; Reduced intensity conditioning regimen ID D-index; cD-index; D21-index; Pulmonary infection; Reduced intensity conditioning regimen AB We evaluated the predictive value of the D-index for pulmonary infection in the early phase of reduced intensity stem cell transplantation (RIST). Out of 68 patients, ten patients developed a pulmonary infection within 100 days after RIST. Both the D-index and the cD-index were higher in the patients with pulmonary infection than in the control group (P=0.009, P=0.042, respectively). The best sensitivity and specificity, calculated with receiver operating characteristic curves, showed that the D-index was superior to the duration of neutropenia in predicting pulmonary infection. We also evaluated the utility of a cumulative D-index until 21 days after RIST (D21-index). The D21-index was higher in the patients with pulmonary infection (P=0.047). The cutoff value of the D21-index was lower than that of the D-index (8650 vs. 11000) with comparable sensitivity and specificity. Our results demonstrate that the D21-index, as well as the D-index, are useful tools for the prediction of pulmonary infection in RIST. C1 [Aoki, Jun] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Tsubokura, Masaharu] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kakihana, Kazuhiko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Oshikawa, Gaku] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kobayashi, Takeshi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Doki, Noriko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Sakamaki, Hisashi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Ohashi, Kazuteru] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. RP Kakihana, K (reprint author), Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 113-8677 Tokyo, Japan. EM kakihana@cick.jp CR Engels EA, Ellis CA, Supran SE, Schmid CH, Barza M, Schenkein DP, Koc Y, Miller KB, Wong JB, 1999, Early infection in bone marrow transplantation: quantitative study of clinical factors that affect risk. Clin Infect Dis 28(2):256–266., DOI 10.1086/515103 Kruger W, Russmann B, Kroger N, Salomon C, Ekopf N, Elsner HA, Kaulfers PM, Mack D, Fuchs N, Durken M, Kabisch H, Erttmann R, Zander AR, 1999, Early infections in patients undergoing bone marrow or blood stem cell transplantation–a 7 year single centre investigation of 409 cases. Bone Marrow Transplant 23(6):589–597., DOI 10. 1038/sj.bmt.1701614 Mossad SB, Longworth DL, Goormastic M, Serkey JM, Keys TF, Bolwell BJ, 1996, Early infectious complications in autologous bone marrow transplantation: a review of 219 patients. Bone Marrow Transplant 18(2):265–271 Sparrelid E, Hagglund H, Remberger M, Ringden O, Lonnqvist B, Ljungman P, Andersson J, 1998, Bacteraemia during the aplastic phase after allogeneic bone marrow transplantation is associated with early death from invasive fungal infection. Bone Marrow Transplant 22(8):795–800., DOI 10.1038/sj.bmt.1701404 Portugal RD, Garnica M, Nucci M, 2009, Index to predict invasive mold infection in high-risk neutropenic patients based on the area over the neutrophil curve. J Clin Oncol 27(23):3849–3854., DOI 10.1200/ JCO.2008.21.0856 Kimura S, Oshima K, Sato K, Sato M, Terasako K, Nakasone H, Kikuchi M, Okuda S, Kako S, Yamazaki R, Tanaka Y, Tanihara A, Nishida J, Kanda Y, 2010, Retrospective evaluation of the area over the neutrophil curve index to predict early infection in hematopoietic stem cell transplantation recipients. Biol Blood Marrow Transplant 16(10):1355–1361., DOI 10.1016/j.bbmt.2010.04.012 Kolbe K, Domkin D, Derigs HG, Bhakdi S, Huber C, Aulitzky WE, 1997, Infectious complications during neutropenia subsequent to peripheral blood stem cell transplantation. Bone Marrow Transplant 19(2):143–147., DOI 10.1038/sj.bmt.1700621 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 879 EP 883 DI 10.1007/s12253-014-9768-8 PG 5 ER PT J AU Li, F Wang, W Li, L Chang, Y Su, D Guo, G He, X Li, M AF Li, Fenqiang Wang, Wenhui Li, Li Chang, Yaowen Su, Dongjun Guo, Gang He, Xuewen Li, Mingxiang TI An Effective Therapy to Painful Bone Metastases: Cryoablation Combined with Zoledronic Acid SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pain; Bone metastases; Cryoablation; Zoledronic acid; Combined therapy; Efficacy ID Pain; Bone metastases; Cryoablation; Zoledronic acid; Combined therapy; Efficacy AB Approximately half or more of patients diagnosed with late malignant tumors may suffer from metastatic bone pain, effective palliation of pain becomes an important part of comprehensive therapy formalignant tumors. In this study, we examined the efficacy and safety of the combined regimen of cryoablation and zoledronic acid in patients of bonemetastatic pain. A total of 84 subjects were randomly divided into three groups, and underwent treatments of cryoablation plus zoledronic acid, cryoablation alone, zoledronic acid alone between June 2009 and March 2012. Patients responses had been assessed for a total of 6 months by using the Brief Pain Inventory (BPI)-Short Form. The results showed that the mean response of worst and average pain significantly dropped at week 2 (all P<0.05) in group with cryoablation treatment but at week 4 (all P<0.05) in group with zoledronic acid treatment. While between week 16 and week 24, zoledronic acid treatments showed more durable response to worst and average pain compared to cryoablation (all P<0.05). Cryoablation plus zoledronic acid regimen showed significant drop in worst and average pain between week 1 and week 4 compared to zoledronic acid alone (all P<0.05) and more durable effect on bone metastatic pain between week 12 and week 24 than cryoablation alone (all P<0.05). Additionally, no serious adverse effects and complication were observed by this combination use. In conclusion, cryoablation combined with zoledronic acid was safe and effective regimen and showed its superiority of fast response and durable effect on painful bone metastases. C1 [Li, Fenqiang] Lanzhou University First Hospital, Department of Interventional Radiology, No.1 Donggang West Road, 730000 Lanzhou, China. [Wang, Wenhui] Lanzhou University First Hospital, Department of Interventional Radiology, No.1 Donggang West Road, 730000 Lanzhou, China. [Li, Li] Lanzhou University First Hospital, Department of Interventional Radiology, No.1 Donggang West Road, 730000 Lanzhou, China. [Chang, Yaowen] Lanzhou University First Hospital, Department of Interventional Radiology, No.1 Donggang West Road, 730000 Lanzhou, China. [Su, Dongjun] Lanzhou University First Hospital, Department of Interventional Radiology, No.1 Donggang West Road, 730000 Lanzhou, China. [Guo, Gang] Lanzhou University First Hospital, Department of Interventional Radiology, No.1 Donggang West Road, 730000 Lanzhou, China. [He, Xuewen] Lanzhou University First Hospital, Department of Interventional Radiology, No.1 Donggang West Road, 730000 Lanzhou, China. [Li, Mingxiang] Lanzhou University First Hospital, Department of Interventional Radiology, No.1 Donggang West Road, 730000 Lanzhou, China. RP Wang, W (reprint author), Lanzhou University First Hospital, Department of Interventional Radiology, 730000 Lanzhou, China. EM wangwenhui1968@163.com CR Yoh K, Kubota K, Ohmatsu H, Goto K, Niho S, Ohe Y, 2012, Feasibility study of zoledronic acid plus cisplatin-docetaxel as firstline treatment for advanced non-small cell lung cancer with bone metastases. Anticancer Res 32:4131–4135 Martin M, Bell R, Bourgeois H, Brufsky A, Diel I, Eniu A, Fallowfield L, Fujiwara Y, Jassem J, Paterson AH, Ritchie D, Steger GG, Stopeck A, Vogel C, Fan M, Jiang Q, Chung K, Dansey R, Braun A, 2012, Bone-related complications and quality of life in advanced breast cancer: results from a randomized phase III trial of denosumab versus zoledronic acid. Clin Cancer Res 18:4841– 4849 Fujino M, Suzuki K, Nishio M, Nishiyama N, Osaka Y, 2011, Strategy of radiation therapy for bonemetastases andMSCC in breast cancer patients. Breast Cancer 18:238–243 Saylor PJ, Lee RJ, Smith MR, 2011, Emerging therapies to prevent skeletal morbidity in men with prostate cancer. J Clin Oncol 29: 3705–3714 Woodward E, Jagdev S, McParland L, Clark K, Gregory W, Newsham A, Rogerson S, Hayward K, Selby P, Brown J, 2011, Skeletal complications and survival in renal cancer patients with bone metastases. Bone 48:160–166 Lien K, Zeng L, Zhang L, Nguyen J, Di Giovanni J, Popovic M, Jamani R, Cramarossa G, Culleton S, Chow E, 2012, Predictive factors for well-being in advanced cancer patients referred for palliative radiotherapy. Clin Oncol, R Coll Radiol, 24:443–451 Smith HS, Barkin RL, 2014, Painful boney metastases. Am J Ther 21:106–130 FarhanghiM, Holmes RA, Volkert WA, Logan KW, Singh A, 1992, Samarium- 153-EDTMP: pharmacokinetic, toxicity and pain response using an escalating dose schedule in treatment of metastatic bone cancer. J Nucl Med 33:1451–1458 Smith HS, 2011, Painful osseous metastases. Pain Physician 14: E373–E403 Callstrom MR, Atwell TD, Charboneau JW, Farrell MA, Goetz MP, Rubin J, Sloan JA, Novotny PJ, Welch TJ, Maus TP, Wong GY, Brown KJ, 2006, Painful metastases involving bone: percutaneous image-guided cryoablation –prospective trial interim analysis. Radiology 241:572–580 CallstromMR, DupuyDE, Solomon SB, Beres RA, Littrup PJ, Davis KW, Paz-Fumagalli R, Hoffman C, Atwell TD, Charboneau JW, Schmit GD, Goetz MP, Rubin J, Brown KJ, Novotny PJ, Sloan JA, 2013, Percutaneous image-guided cryoablation of painful metastases involving bone: multicenter trial. Cancer 119:1033–1041 Choi J, Raghavan M, 2012, Diagnostic imaging and image-guided therapy of skeletal metastases. Cancer Control 19:102–112 Castaneda Rodriguez WR, Callstrom MR, 2011, Effective pain palliation and prevention of fracture for axial-loading skeletal metastases using combined cryoablation and cementoplasty. Tech Vasc Interv Radiol 14:160–169 de Freitas RM, de Menezes MR, Cerri GG, Gangi A, 2011, Sclerotic vertebral metastases: pain palliation using percutaneous image-guided cryoablation. Cardiovasc Intervent Radiol 34(Suppl 2):S294–S299 Callstrom MR, Charboneau JW, 2007, Image-guided palliation of painful metastases using percutaneous ablation. Tech Vasc Interv Radiol 10:120–131 Paparella S, Finkelberg E, Varisco D, Tondelli E, Rocco F, 2011)Use of zoledronic acid in patients with prostate cancer bone metastases: control of pain and musculoskeletal complications. Urologia 78:300– 304 Yamada K, Yoshimura M, Kaise H, Ogata A, Ueda N, Tokuuye K, Kohno N, 2012, Concurrent use of Sr-89 chloride with zoledronic acid is safe and effective for breast cancer patients with painful bone metastases. Exp Ther Med 3:226–230 Saad F, Eastham J, 2010, Zoledronic Acid improves clinical outcomes when administered before onset of bone pain in patients with prostate cancer. Urology 76:1175–1181 Ishiwata T, Hirose T, Hirama M, Miura K, Iwakami S, Tominaga S, Adachi M, Takahashi K, 2011, A feasibility study of zoledronic acid combined with carboplatin/nedaplatin plus paclitaxel in patients with non-small cell lung cancerwith bonemetastases. Tumori 97:568–572 Lopez-OlivoMA, Shah NA, PrattG, Risser JM, Symanski E, Suarez- Almazor ME, 2012, Bisphosphonates in the treatment of patients with lung cancer and metastatic bone disease: a systematic review and meta-analysis. Support Care Cancer 20:2985–2998 Cleeland CS, Gonin R, Hatfield AK, Edmonson JH, Blum RH, Stewart JA, Pandya KJ, 1994, Pain and its treatment in outpatients with metastatic cancer. N Engl J Med 330:592–596 Foro Arnalot P, Fontanals AV, Galceran JC, Lynd F, Latiesas XS, de Dios NR, Castillejo AR, Bassols ML, Galan JL, Conejo IM, Lopez MA(2008, Randomized clinical trialwith two palliative radiotherapy regimens in painful bone metastases: 30 Gy in 10 fractions compared with 8 Gy in single fraction. Radiother Oncol 89:150–155 Farrar JT, Young JP Jr, LaMoreaux L, Werth JL, Poole RM, 2001, Clinical importance of changes in chronic pain intensity measured on an 11- point numerical pain rating scale. Pain 94:149–158 Rosen LS, Gordon D, Tchekmedyian NS, Yanagihara R, Hirsh V, Krzakowski M, Pawlicki M, De Souza P, Zheng M, Urbanowitz G, Reitsma D, Seaman J, 2004, Long-term efficacy and safety of zoledronic acid in the treatment of skeletal metastases in patients with nonsmall cell lung carcinoma and other solid tumors: a randomized, Phase III, double-blind, placebo-controlled trial. Cancer 100: 2613–2621 Pozzi S, Raje N, 2011, The role of bisphosphonates in multiple myeloma: mechanisms, side effects, and the future. Oncologist 16:651–662 Fu D, He X, Yang S, Xu W, Lin T, Feng X, 2011, Zoledronic acid inhibits vasculogenic mimicry in murine osteosarcoma cell line in vitro. BMC Musculoskelet Disord 12:146 Insalaco L, Di Gaudio F, Terrasi M, Amodeo V, Caruso S, Corsini LR, Fanale D, Margarese N, Santini D, Bazan V, Russo A, 2012, Analysis of molecular mechanisms and anti-tumoural effects of zoledronic acid in breast cancer cells. J Cell Mol Med 16:2186–2195 Ottewell PD, Woodward JK, Lefley DV, Evans CA, Coleman RE, Holen I, 2009, Anticancer mechanisms of doxorubicin and zoledronic acid in breast cancer tumor growth in bone. Mol Cancer Ther 8: 2821–2832 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 885 EP 891 DI 10.1007/s12253-014-9769-7 PG 7 ER PT J AU Pocza, T Sebestyen, A Turanyi, E Krenacs, T Mark, Sticz, BT Jakab, Zs Hauser, P AF Pocza, Timea Sebestyen, Anna Turanyi, Eszter Krenacs, Tibor Mark, Agnes Sticz, Bela Tamas Jakab, Zsuzsanna Hauser, Peter TI mTOR Pathway As a Potential Target In a Subset of Human Medulloblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Medulloblastoma; mTORC1; mTOR inhibitors; Survival; Daoy; Pediatric; Brain; Tumor; Rapamycin ID Medulloblastoma; mTORC1; mTOR inhibitors; Survival; Daoy; Pediatric; Brain; Tumor; Rapamycin AB As mammalian Target of Rapamycin (mTOR) plays role in protein synthesis and metabolism, mTOR pathway activation is involved in the pathogenesis of several types of tumors. Our aim was to elucidate its role in medulloblastoma in terms of prognosis and as a therapeutic target. Members of activated mTOR complex 1 (mTORC1) pathway, phosphomTOR (p-mTOR) and phospho-S6 (p-S6) were examined by immunohistochemistry in formalin fixed paraffin embedded samples of 40 patients with medulloblastoma, and results were compared to clinical features and survival of patients. In proliferation assays, Daoy and UW228–2 medulloblastoma cell lines were tested by rapamycin, an mTORC1 inhibitor, and NVP-BEZ235, a dual mTOR and phosphatidylinositol 3- kinase (PI3K) inhibitor, each in monotherapy and in combination with cytostatic drugs (cisplatin, etoposide). Components of mTORC1 and mTORC2 complexes were also examined in these cell lines. Neither presence of p-mTOR (32.5 %) nor p-S6 (32.5 %) correlated with age, gender or histological subtype. In 22.5 % of cases simultaneous expression of pmTOR and p-S6 was shown. Kaplan-Meier analysis showed inferior survival of patients expressing both marker proteins, but it was not statistically significant, probably due to low case number. UW228–2 cells had greater sensitivity to mTOR inhibitors, possibly due to its higher mTORC1 specific protein expression levels, compared to Daoy cells. In both cell lines antiproliferative effect of cytostatic drugs was significantly enhanced by mTOR inhibitors (p<0.05). Based on our in vitro and clinicopathological studies mTOR inhibitors may have a role in the future treatment of a subset of patients with medulloblastoma. C1 [Pocza, Timea] Semmelweis University, 2nd Department of Pediatrics, Tuzolto utca 7-9, H-1094 Budapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Turanyi, Eszter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Mark, Agnes] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Sticz, Bela Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Jakab, Zsuzsanna] Semmelweis University, 2nd Department of Pediatrics, Tuzolto utca 7-9, H-1094 Budapest, Hungary. [Hauser, Peter] Semmelweis University, 2nd Department of Pediatrics, Tuzolto utca 7-9, H-1094 Budapest, Hungary. RP Hauser, P (reprint author), Semmelweis University, 2nd Department of Pediatrics, H-1094 Budapest, Hungary. EM hauserpeti@yahoo.com CR Hatten ME, Roussel MF, 2011, Development and Cancer of the Cerebellum. Trends Neurosci 34:134–142 Roussel MF, Hatten ME, 2011, Cerebellum Development and Medulloblastoma. Curr Top Dev Biol 94:235–282 Crawford JR, MacDonald TJ, Packer RJ, 2007, Medulloblastoma in Childhood: new Biological Advances. Lancet Neurol 6:1073–1085 Massimino M, Giangaspero F, Garre ML, Gandola L, Poggi G, Biassoni V, Gatta G, Rutkowski S, 2011, Childhood Medulloblastoma. Crit Rev Oncol Hematol 79:65–83 Gottardo NG, Gajjar A, 2008, Chemotherapy for Malignant Brain Tumors of Childhood. J Child Neurol 23:1149–1159 Packer RJ, Zhou T, Holmes E, Vezina G, Gajjar A, 2013, Survival and Secondary Tumors in Children with Medulloblastoma Receiving Radiotherapy and Adjuvant Chemotherapy: Results of Children’s Oncology Group trial A9961. Neuro Oncol 15:97–103 Dazert E, Hall MN, 2011, mTOR Signaling in Disease. Curr Opin Cell Biol 23:744–755 Willems L, Tamburini J, Chapuis N, Lacombe C, Mayeux P, Bouscary D, 2012, PI3K and mTOR Signaling Pathways in Cancer: New Data on Targeted Therapies. Curr Oncol Rep 14:129– 138 Guertin DA, Sabatini DM, 2007, Defining the Role of mTOR in Cancer. Cancer Cell 12:9–22 Dunlop EA, Tee AR, 2009, Mammalian target of Rapamycin Complex 1: Signalling Inputs, Substrates and Feedback Mechanisms. Cell Signal 21:827–835 Akcakanat A, Singh G, Hung MC, Meric-Bernstam F, 2007, Rapamycin Regulates the Phosphorylation of Rictor. Biochem Biophys Res Commun 362:330–333 Fouladi M, Laningham F, Wu J, O’Shaughnessy MA, Molina K, Broniscer A, Spunt SL, Luckett I, Stewart CF, Houghton PJ, Gilbertson RJ, Furman WL, 2007, Phase I Study of Everolimus in Pediatric Patients with Refractory Solid Tumors. J Clin Oncol 25: 4806–4812 Jozwiak J, Sontowska I, Bikowska B, Grajkowska W, Galus R, Roszkowski M, 2011, Favourable Prognosis in Medulloblastoma with Extensive Nodularity is Associated with Mitogen-activated Protein Kinase Upregulation. Folia Neuropathol 49:257–261 Wlodarski P, Grajkowska W, Lojek M, Rainko K, Jozwiak J, 2006, Activation of Akt and Erk Pathways in Medulloblastoma. Folia Neuropathol 44:214–220 Wlodarski PK, Boszczyk A, Grajkowska W, Roszkowski M, Jozwiak J, 2008, Implication of Active Erk in the Classic Type of Human Medulloblastoma. Folia Neuropathol 46:117–122 Bhatia B, Northcott PA, Hambardzumyan D, Govindarajan B, Brat DJ, Arbiser JL, Holland EC, Taylor MD, Kenney AM, 2009, Tuberous Sclerosis Complex Suppression in Cerebellar Development and Medulloblastoma: Separate Regulation of Mammalian Target of Rapamycin Activity and p27 Kip1 Localization. Cancer Res 69:7224–7234 Bhatia B, Nahle Z, Kenney AM, 2010, Double Trouble: when Sonic Hedgehog Signaling Meets TSC Inactivation. Cell Cycle 9:456–459 Mainwaring LA, Kenney AM, 2011, Divergent Functions for eIF4E and S6 Kinase by Sonic Hedgehog Mitogenic Signaling in the Developing Cerebellum. Oncogene 30:1784–1797 Parathath SR, Mainwaring LA, Fernandez-L A, Campbell DO, Kenney AM, 2008, Insulin Receptor Substrate 1 is an Effector of Sonic Hedgehog Mitogenic Signaling in Cerebellar Neural Precursors. Development 135:3291–3300 Buonamici S, Williams J, Morrissey M, Wang A, Guo R, Vattay A, Hsiao K, Yuan J, Green J, Ospina B, Yu Q, Ostrom L, Fordjour P, Anderson DL, Monahan JE, Kelleher JF, Peukert S, Pan S, Wu X, Maira SM, Garcia-Echeverria C, Briggs KJ, Watkins DN, Yao YM, Lengauer C, Warmuth M, Sellers WR, Dorsch M, 2010, Interfering with resistance to smoothened antagonists by inhibition of the PI3K pathway in medulloblastoma. Sci Transl Med 2:51ra70 Geoerger B, Kerr K, Tang CB, Fung KM, Powell B, Sutton LN, Phillips PC, Janss AJ, 2001, Antitumor Activity of the Rapamycin Analog CCI-779 in Human Primitive Neuroectodermal Tumor/ Medulloblastoma Models as Single Agent and in Combination Chemotherapy. Cancer Res 61:1527–1532 Pei Y, Moore CE,Wang J, Tewari AK, Eroshkin A, Cho YJ,Witt H, Korshunov A, Read TA, Sun JL, Schmitt EM, Miller CR, Buckley AF,McLendon RE,Westbrook TF, Northcott PA, TaylorMD, Pfister SM, Febbo PG, Wechsler-Reya RJ, 2012, An animal Model of MYC-driven Medulloblastoma. Cancer Cell 21:155–167 Hauser P, Jakab Z, Kiss C, Szegedi I, Bardi E, Batyik K, Ottoffy G, Kajtar P, Szucs R, Nagy K, Cservak J, Masat P, Balint K, Kordas M, Bognar L, Kocsis B, Vizkeleti J, Krivan G, Kallay K, Benyo G, Schuler D, Garami M, 2009, Elozetes eredmenyek a medulloblastoma/primitiv neuroektodermalis tumor, PNET, kezeleseben a magyar MBL 2004 kezelesi semaval. Magyar Belorvosi Archivum, pp 196–201 Back JH, Kim AL, 2011, The expanding Relevance of Nuclear mTOR in Carcinogenesis. Cell Cycle 10:3849–3852 Malik AR, Urbanska M, Macias M, Skalecka A, Jaworski J, 2013, Beyond Control of Protein Translation: what we Have Learned About the Non-canonical Regulation and Function of Mammalian Target of Rapamycin, mTOR). Biochim Biophys Acta 1834:1434–1448 Rosner M, Hengstschlager M, 2008, Cytoplasmic and Nuclear Distribution of the Protein complexes mTORC1 and mTORC2: Rapamycin Triggers Dephosphorylation and Delocalization of the mTORC2 Components riCtor and Sin1. Hum Mol Genet 17:2934– 2948 Jozwiak J, Grajkowska W, Wlodarski P, 2007, Pathogenesis of Medulloblastoma and Current Treatment Outlook. Med Res Rev 27:869–890 Yecies JL, Manning BD, 2011, mTOR links Oncogenic Signaling to Tumor Cell Metabolism. J Mol Med, Berl, 89:221–228 Del Valle L, EnamS, Lassak A,Wang JY, Croul S, Khalili K, Reiss K, 2002, Insulin-like Growth Factor I Receptor Activity in Human Medulloblastomas. Clin Cancer Res 8:1822–1830 Yang J, Liao D, Wang Z, Liu F, Wu G, 2011, Mammalian Target of Rapamycin Signaling Pathway Contributes to Glioma Progression and Patients’ Prognosis. J Surg Res 168:97–102 Pelloski CE, Lin E, Zhang L, YungWK, Colman H, Liu JL,Woo SY, Heimberger AB, Suki D, PradosM, Chang S, Barker FG, Fuller GN, Aldape KD, 2006, Prognostic Associations of Activated Mitogenactivated Protein Kinase and Akt Pathways in Glioblastoma. Clin Cancer Res 12:3935–3941 Setsu N, Kohashi K, EndoM, Yamamoto H, Tamiya S, Takahashi Y, Yamada Y, Ishii T, Matsuda S, Yokoyama R, Iwamoto Y, Oda Y, 2013, Phosphorylation of Signal Transducer and Activator of Transcription 3 in Soft Tissue Leiomyosarcoma is Associated with a Better Prognosis. Int J Cancer 132:109–115 Sebestyen A, Sticz TB, Mark A, HajduM, Timar B, Nemes K, Nagy N, Varadi Z, Kopper L, 2012, Activity and Complexes of mTOR in Diffuse large B-cell Lymphomas–a Tissue Microarray Study. Mod Pathol 25:1623–1628 Tampellini M, Longo M, Cappia S, Bacillo E, Alabiso I, Volante M, Dogliotti L, Papotti M, 2007, Co-expression of EGF Receptor, TGFalpha and S6 Kinase is Significantly Associated with Colorectal Carcinomas with Distant Metastases at Diagnosis. Virchows Arch 450:321–328 Chaisuparat R, Rojanawatsirivej S, Yodsanga S, 2013, Ribosomal Protein S6 Phosphorylation is Associated with Epithelial Dysplasia and Squamous Cell Carcinoma of the Oral Cavity. Pathol Oncol Res 19:189–193 Baryawno N, Sveinbjornsson B, Eksborg S, Chen CS, Kogner P, Johnsen JI, 2010, Small-molecule Inhibitors of Phosphatidylinositol 3-Kinase/Akt Signaling Inhibit Wnt/beta-Catenin Pathway Crosstalk and SuppressMedulloblastoma Growth. Cancer Res 70:266–276 Noh WC, Mondesire WH, Peng J, Jian W, Zhang H, Dong J, Mills GB, Hung MC, Meric-Bernstam F, 2004, Determinants of Rapamycin Sensitivity in Breast Cancer Cells. Clin Cancer Res 10: 1013–1023 Maira SM, Stauffer F, Brueggen J, Furet P, Schnell C, Fritsch C, Brachmann S, Chene P, De Pover A, Schoemaker K, Fabbro D, Gabriel D, Simonen M, Murphy L, Finan P, Sellers W, Garcia- Echeverria C, 2008, Identification and Characterization of NVPBEZ235, A New Orally Available Dual phosphatidylinositol 3- Kinase/Mammalian Target of Rapamycin Inhibitor with Potent in vivo Antitumor Activity. Mol Cancer Ther 7:1851–1863 Wolff JE, Brown RE, Buryanek J, Pfister S, Vats TS, Rytting ME, 2012, Preliminary Experience with Personalized and Targeted Therapy for Pediatric Brain Tumors. Pediatr Blood Cancer 59:27–33 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 893 EP 900 DI 10.1007/s12253-014-9771-0 PG 8 ER PT J AU Wrobel, T Pogrzeba, J Stefanko, E Wojtowicz, M Jazwiec, B Dzietczenia, J Mazur, G Kuliczkowski, K AF Wrobel, Tomasz Pogrzeba, Joanna Stefanko, Ewa Wojtowicz, Marcin Jazwiec, Bozena Dzietczenia, Justyna Mazur, Grzegorz Kuliczkowski, Kazimierz TI Expression of Eph A4, Eph B2 and Eph B4 Receptors in AML SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acutemyeloid leukemia; EphA4-Eph B2-Eph B4; Prognostic impact ID Acutemyeloid leukemia; EphA4-Eph B2-Eph B4; Prognostic impact AB Eph receptors represent the largest subfamily of receptor tyrosine kinases (RTKs). The up- regulation of Eph receptors has been documented in various solid tumors, where it often correlates with poor prognosis. Their significance in hematologic malignancies is still unclear. This study aimed to investigate the expression of Eph A4, Eph B2, and Eph B4 mRNA in non - M3 AML patients and determine their prognostic significance. Bone marrow samples from 101 newly diagnosed non -M3 AML patients and 26 healthy controls for comparison were quantified by real time reverse transcriptase polymerase chain reaction (RT-PCR), and the comparative cycle threshold (Ct) method was used to determine their relative expression levels to GUS control gene. The results showed that expression of all selected Eph receptors was significantly lower in AML patients comparing to controls. It also differed according to FAB subtypes. The decreased expression levels of Eph A4 were associated with higher leukocytes (p=0.022) and blast cell counts (p=0.001), and unfavorable FLT3-ITD mutation. Our study revealed significant correlation between lower EphB2 expression levels, and higher complete remission rate (p=0.009724) and longer overall survival. Additionally, we found that patients with shorter RFS had decreased EphB4 expression (p=0.00). In conclusion, the results suggest the prognostic impact of decreased expression levels of some Eph receptors in AML patients. C1 [Wrobel, Tomasz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, L. Pasteura 4 Street, 50-367 Wroclaw, Poland. [Pogrzeba, Joanna] Regional Hospital in Opole, Department of Hematology, ul. Katowicka 64Opole, Poland. [Stefanko, Ewa] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, L. Pasteura 4 Street, 50-367 Wroclaw, Poland. [Wojtowicz, Marcin] Regional Hospital in Opole, Department of Hematology, ul. Katowicka 64Opole, Poland. [Jazwiec, Bozena] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, L. Pasteura 4 Street, 50-367 Wroclaw, Poland. [Dzietczenia, Justyna] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, L. Pasteura 4 Street, 50-367 Wroclaw, Poland. [Mazur, Grzegorz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, L. Pasteura 4 Street, 50-367 Wroclaw, Poland. [Kuliczkowski, Kazimierz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, L. Pasteura 4 Street, 50-367 Wroclaw, Poland. RP Pogrzeba, J (reprint author), Regional Hospital in Opole, Department of Hematology, Opole, Poland. EM joannapog@interia.pl CR Eph Nomenclature Committee, 1997, Unified nomenclature for Eph family receptors and their ligands, the ephrins. Cell 90:403–404 Gale NW, Holland SJ, Valenzuela DM, 1996, Eph receptors and ligands comprise two major specificity subclasses and are reciprocally compartmentalized during embryogenesis. Neuron 17:9–19 Pasquale EB, 2005, Eph receptor signaling casts a wide net on cell behavior. Nat Rev Mol Cell Biol 6:462–475 Himanen JP, Nicolov DB, 2003, Eph receptors and ephrins. IJBCB 35:130–134 Bruckner K, Klein R, 1998, Signaling by Eph receptors and their ephrin ligands. Curr Opin Neurobiol 8:75–82 Cowan CA, Henkemeyer M, 2002, Ephrins in reverse, park and drive. Trends Cell Biol 12:339–346 Wang HU et al, 1998, Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor EphB4. Cell 93:741–753 Flanagan JG, Vanderhaegen P, 1998, The ephrins and Eph receptors in neural development. Annu Rev Neurosci 21:309–345 Andeas AC, Ziemiecki A, 2003, Eph and ephrin signaling in mammary gland morphogenesis and cancer. J Mammary Gland Biol Neoplasia 8:475–485 Ji XD, Li G, Feng YX, 2011, EphB3 is overexpressed in non-smallcell lung cancer and promotes tumor metastasis by enhancing cell survival and migration. Cancer Res 71(3):1156–1166 Kinch MS,MooreMB, Harpole DH Jr, 2003, Predictive value of the EphA2 receptor tyrosine kinase in lung cancer recurrence and survival. Clin Cancer Res 9(2):613–618 Stephenson SA, Slomka S, Douglas EL, 2001, Receptor protein tyrosine kinase EphB4 is up-regulated in colon cancer. BMC Mol Biol 2:15 Yuan W, Ge J, Chen Z, 2009, Over-expression of EphA2 and EphrinA-1 in human gastric adenocarcinoma and is prognostic value for postoperative patients. Dig Dis Sci 54:2410–2417 YuanW, Chen Z,Wu S, 2009, Expression of EphA2 and E-cadherin in gastric cancer: correlated with tumor progression and lymphogenous metastasis. Pathol Oncol Res 15(3):473–478 Tachibana M, Tonomoto Y, Hyakudomi M, 2007, Expression and prognostic significance of EFNB2 and EphB4 genes in patients with oesophageal squamous cell carcinoma. Dig Liver Dis 39(8):725–732 Wang J, Dong Y, Wang X, 2010, Expression of EphA1 in gastric carcinomas is associated with metastasis and survival. Oncol Rep 24(6):1577–1584 Yu G, Gao Y, Ni C, 2011, Reduced expression of EphB2 is significantly associated with nodal metastasis in Chinese patients with gastric cancer. J Cancer Res Clin Oncol 137(1):73–80 Alazzouzi H, Davalos V, Kokko A, 2005, Mechanisms of inactivation of the receptor tyrosine kinase EPHB2 in colorectal tumors. Cancer Res 65(22):10170–10173 Huusko P, Ponciano-Jackson D, Wolf M, 2004, Nonsense-mediated decay microarray analysis identifies mutations of EPHB2 in human prostate cancer. Nat Genet 36(9):979–983 Guan M, Liu L, Zhao X, 2011, Copy number variations of EphA3 are associated with multiple types of hematologic malignancies. Clin Lymphoma Myeloma Leuk 11(1):50–53 Bennett JM, Catovsky D, Daniel MT et al, 1985, Proposals for the classification of the acute leukaemias. A report of the French- American-British, FAB, co-operative group. Ann Intern Med 103: 620–625 Dohner H, Estey EH, Amadori S et al, 2010, Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European Leukemia Net. Blood 115:453–474 Holowiecki J, Grosicki S, Robak T et al, 2004, Polish Adult Leukemia Group, PALG). Addition of cladribine to daunorubicin and cytarabine increases complete remission rate after a single course of induction treatment in acute myeloid leukemia. Multicenter, phase III Study. Leukemia 18:989–997 Holowiecki J, Grosicki S, Kyrcz-Krzemien S et al, 2008, Daunorubicin, cytarabine and fludarabine, DAF, for remission induction in relapsed or refractory acute myeloid leukemia. Evaluation of safety, tolerance and early outcome–Polish Adult Leukemia Group, PALG, pilot study. Ann Hematol 81:361–367 Holowiecki J, Grosicki S, Robak T et al, 2001, Daunorubicin, cytarabine, and 2CdA, DAC-7, for remission indiction in “de novo” Adult acute myeloid leukemia patients. Acta Haematol Pol 2002: 839–847 Wrzesien-Kus A, Robak T, Jamroziak K et al, 2002, The treatment of acute myeloid leukemia with mitoxantrone, etoposide and low-dose cytarabine in elderly patients - a report of Polish Acute Leukemia Group, PALG, phase II study. Neoplasma 49:405–411 Yates J, Glidewell O, Wiernik P et al, 1982, Cytosine arabinoside with daunorubicin or adriamycin for therapy of acute myelocytic leukemia: a CALGB study. Blood 60: 454–462 Christ O, Feuring-Buske M, Hiddemann W et al, 2007, Pathobiology of acute myeloid leukemia. Med Clin 102: 290–295 Sekeres MA, 2008, Treatment of older adults with acute myeloid leukemia: state of the art and current perspectives. Haematologica 93: 1769–1772 Kuang SQ, Bai H, Fang ZH, 2010, Aberrant DNA methylation and epigenetic inactivation of Eph receptor tyrosine kinases and ephrin ligands in acute lymphoblastic leukemia. Blood 115(12):2412–2419 Davalos V, Dopeso H, Velho S, 2007, High EPHB2 mutation rate in gastric but not endometrial tumors with microsatellite instability. Oncogene 26(2):308–311 Oba SM, Wang YJ, Song JP, Li ZY, Kobayashi K, Tsugane S, Hamada GS, Tanaka M, Sugimura H, 2001, Genomic structure and loss of heterozygosity of EPHB2 in colorectal cancer. Cancer Lett 164(1):97–104 Alonso-C LM, TrinidadEM, deGarcillan B, Ballesteros M, Castellanos M, Cotillo I, Munoz JJ, Zapata AG, 2009, Expression profile of Eph receptors and ephrin ligands in healthy human B lymphocytes and chronic lymphocytic leukemia B-cells. Leuk Res 33(3):395–406 Oshima T, Akaike M, Yoshihara K, 2008, Overexpression of EphA4 gene and reduced expression of EphB2 gene correlates with liver metastasis in colorectal cancer. Int J Oncol 33(3): 573–577 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 901 EP 907 DI 10.1007/s12253-014-9767-9 PG 7 ER PT J AU Kalfert, D Celakovsky, P Laco, J Ludvikova, M AF Kalfert, David Celakovsky, Petr Laco, Jan Ludvikova, Marie TI The Role of Protein p16INK4a in Glottic Laryngeal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Laryngeal cancer; Squamous cell carcinoma; p16 protein; Prognostic marker ID Laryngeal cancer; Squamous cell carcinoma; p16 protein; Prognostic marker AB Head and neck squamous cell cancer (HNSCC) includes tumors of various anatomical sites sharing the common etiological factors. However some differences in pathogenesis and prognosis of HNSCC have been hitherto documented. Laryngeal squamous cell carcinoma (LSCC) is one the most common type of the head and neck cancer. The majority of laryngeal cancers are located in the glottic area. P16 was recently documented to be important prognostic marker in many tumors including HNSCC. The aim of our study was to assess the significance of p16 expression in glottic LSCC. Fifty eight patients after surgical treatment of the glottic LSCC were enrolled in the retrospective study. The p16 expression was immunohistochemically detected and semiquantitatively evaluated in tumor tissue. The results were statistically correlated with clinical and pathological parameters. Protein p16 was expressed in glottic LSCC of 15 patients (25.9 %). Statistically significant higher p16 overexpression was proven in non-smokers in comparison with smokers (75 % versus 18 %; p =0.003). Recurrent cancer was diagnosed in 8 patients (13.8 %), and all these tumors were p16 negative. Our study shows, that p16 expression in glottic LSCC especially in subgroup of non-smokers might be promising prognosticator of better clinical outcome in routine practice. The p16 status did not statistically correlate with cervical lymph nodemetastases orwith grading and staging of cancers, respectively. The preliminary results suggest that p16 overexpression in glottic LSCC may identify patients at low risk of disease recurrence. However, the pathobiology of this tumor as well as predictive role of p16 expression in laryngeal cancer still remains to be better elucidated. C1 [Kalfert, David] University Hospital Hradec Kralove, Department of Otorhinolaryngology and Head and Neck SurgeryPrague, Czech Republic. [Celakovsky, Petr] University Hospital Hradec Kralove, Department of Otorhinolaryngology and Head and Neck SurgeryPrague, Czech Republic. [Laco, Jan] Charles University, 1st Faculty of Medicine and General Teaching HospitalPrague, Czech Republic. [Ludvikova, Marie] Charles University in Prague, Faculty of Medicine in Pilsen, Institute of Biology, Karlovarska 48, 30166 Pilsen, Czech Republic. RP Ludvikova, M (reprint author), Charles University in Prague, Faculty of Medicine in Pilsen, Institute of Biology, 30166 Pilsen, Czech Republic. EM ludvikova.m@email.cz CR Ferlay J, Shin HR, Bray F, Forman D,Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127(12):2893–2917., DOI 10.1002/ijc.25516 de Oliveira DE, Bacchi MM, Macarenco RS, Tagliarini JV, Cordeiro RC, Bacchi CE, 2006)Human papillomavirus and Epstein-Barr virus infection, p53 expression, and cellular proliferation in laryngeal carcinoma. Am J Clin Pathol 126(2):284–293., DOI 10.1309/UU2JADUE- HDWA-TVM9 Stasikowska-Kanicka O, Wagrowska-Danilewicz M, Danilewicz M, 2011, Effect of human papillomavirus on cell cycle-related proteins p16INK4A, p21waf1/cip1, p53 and cyclin D1 in sinonasal inverted papilloma and laryngeal carcinoma.an in situ hybridization study. Folia Histochem Cytobiol 49(1):34–40 Sudhoff HH, Schwarze HP, Winder D, Steinstraesser L, Gorner M, Stanley M, Goon PK, 2011, Evidence for a causal association for HPV in head and neck cancers. Eur Arch Otorhinolaryngol 268(11): 1541–1547., DOI 10.1007/s00405-011-1714-8 TorrenteMC, Rodrigo JP, HaigentzMJr, Dikkers FG, Rinaldo A, Takes RP, Olofsson J, Ferlito A, 2011, Human papillomavirus infections in laryngeal cancer. Head Neck 33(4):581–586., DOI 10.1002/hed.21421 Ribeiro KB, Levi JE, Pawlita M, Koifman S, Matos E, Eluf-Neto J, Wunsch-Filho V, Curado MP, Shangina O, Zaridze D, Szeszenia- Dabrowska N, Lissowska J, Daudt A, Menezes A, Bencko V, Mates D, Fernandez L, Fabianova E, Gheit T, Tommasino M, Boffetta P, Brennan P, Waterboer T, 2011, Low human papillomavirus prevalence in head and neck cancer: results from two large case–control studies in high-incidence regions. Int J Epidemiol 40(2):489–502., DOI 10.1093/ije/dyq249 Konig F, Krekeler G, Honig JF, Cordon-Cardo C, Fischer G, Korabiowska M, 2007, Relation between human papillomavirus positivity and p16 expression in head and neck carcinomas–a tissue microarray study. Anticancer Res 27(1A):283–288 Thomas J, Primeaux T, 2012, Is p16 immunohistochemistry a more cost-effective method for identification of human papilloma virusassociated head and neck squamous cell carcinoma? Ann Diagn Pathol 16(2):91–99., DOI 10.1016/j.anndiagpath.2011.09.002 Canzonieri V, Barzan L, Franchin G, Vaccher E, Talamini R, Sulfaro S, Baldassarre G, 2012, Alteration of G1/S transition regulators influences recurrences in head and neck squamous carcinomas. J Cell Physiol 227(1):233–238., DOI 10.1002/jcp.22723 Krecicki T, Smigiel R, Fraczek M, Kowalczyk M, Sasiadek MM, 2004, Studies of the cell cycle regulatory proteins P16, cyclin D1 and retinoblastoma protein in laryngeal carcinoma tissue. J Laryngol Otol 118(9):676–680., DOI 10.1258/0022215042244769 Nadal A, CardesaA(2003)Molecular biology of laryngeal squamous cell carcinoma. Virchows Arch 442(1):1–7., DOI 10.1007/s00428- 002-0726-6 Tamas L, Szentkuti G, Eros M, Danos K, Brauswetter D, Szende B, Zsakovics I, Krenacs T, 2011, Differential biomarker expression in head and neck cancer correlates with anatomical localization. Pathol Oncol Res 17(3):721–727., DOI 10.1007/s12253-011-9376-9 Sobin LH, Wittekind C, International Union against Cancer, 2002, TNM classification of malignant tumours, 6th edn. Wiley-Liss, New York Laco J, Nekvindova J, Novakova V, Celakovsky P, Dolezalova H, Tucek L, Vosmikova H, Vosmik M, Neskudlova T, Cermakova E, Hacova M, Sobande FA, Ryska A, 2012, Biologic importance and prognostic significance of selected clinicopathological parameters in patients with oral and oropharyngeal squamous cell carcinoma, with emphasis on smoking, protein p16(INK4a, expression, and HPV status. Neoplasma 59(4):398–408., DOI 10.4149/neo_2012_052 Laco J, Slaninka I, Jirasek M, Celakovsky P, Vosmikova H, Ryska A, 2008, High-risk human papillomavirus infection and p16INK4a protein expression in laryngeal lesions. Pathol Res Pract 204(8): 545–552 Silva SD, Nonogaki S, Soares FA, Kowalski LP, 2012, p16, INK4a, has clinicopathological and prognostic impact on oropharynx and larynx squamous cell carcinoma. Braz J Med Biol Res 45(12):1327– 1333 Baumann JL, Cohen S, Evjen AN, Law JH, Vadivelu S, Attia A, Schindler JS, Chung CH, Wirth PS, Meijer CJ, Snijders PJ, Yarbrough WG, Slebos RJ, 2009, Human papillomavirus in early laryngeal carcinoma. Laryngoscope 119(8):1531–1537., DOI 10.1002/ lary.20509 Garcia-Milian R, Hernandez H, Panade L, Rodriguez C, Gonzalez N, Valenzuela C, Arana MD, Perea SE, 1998, Detection and typing of human papillomavirus DNA in benign and malignant tumours of laryngeal epithelium. Acta Otolaryngol 118(5):754–758 Rihkanen H, Peltomaa J, Syrjanen S, 1994, Prevalence of human papillomavirus, HPV, DNA in vocal cords without laryngeal papillomas. Acta Otolaryngol 114(3):348–351 Hobbs CG, Sterne JA, Bailey M, Heyderman RS, Birchall MA, Thomas SJ, 2006, Human papillomavirus and head and neck cancer: a systematic review and meta-analysis. Clin Otolaryngol 31(4):259– 266., DOI 10.1111/j.1749-4486.2006.01246.x Lewis JS Jr, Thorstad WL, Chernock RD, Haughey BH, Yip JH, Zhang Q, El-Mofty SK, 2010, p16 positive oropharyngeal squamous cell carcinoma:an entity with a favorable prognosis regardless of tumor HPV status. Am J Surg Pathol 34(8):1088–1096., DOI 10. 1097/PAS.0b013e3181e84652 Lewis JS Jr, Ukpo OC, Ma XJ, Flanagan JJ, Luo Y, Thorstad WL, Chernock RD, 2012, Transcriptionally-active high-risk human papillomavirus is rare in oral cavity and laryngeal/hypopharyngeal squamous cell carcinomas–a tissue microarray study utilizing E6/E7 mRNA in situ hybridization. Histopathology 60(6):982–991., DOI 10.1111/j.1365-2559.2011.04169.x Ukpo OC, Flanagan JJ,Ma XJ, Luo Y, Thorstad WL, Lewis JS Jr, 2011, High-risk human papillomavirus E6/E7 mRNA detection by a novel in situ hybridization assay strongly correlates with p16 expression and patient outcomes in oropharyngeal squamous cell carcinoma. Am J Surg Pathol 35(9):1343–1350., DOI 10.1097/PAS.0b013e318220e59d Sano T, Oyama T, Kashiwabara K, Fukuda T, Nakajima T, 1998, Expression status of p16 protein is associated with human papillomavirus oncogenic potential in cervical and genital lesions. Am J Pathol 153(6):1741–1748., DOI 10.1016/S0002-9440(10)65689-1 Hafkamp HC, Mooren JJ, Claessen SM, Klingenberg B, Voogd AC, Bot FJ, Klussmann JP, Hopman AH, Manni JJ, Kremer B, Ramaekers FC, Speel EJ, 2009, P21 Cip1/WAF1 expression is strongly associated with HPV-positive tonsillar carcinoma and a favorable prognosis. Mod Pathol 22(5):686–698., DOI 10.1038/ modpathol.2009.23 Broglie MA, Soltermann A, Haile SR, Roosli C, Huber GF, Schmid S, Stoeckli SJ, 2013, Quality of life of oropharyngeal cancer patients with respect to treatment strategy and p16-positivity. Laryngoscope 123(1):164–170., DOI 10.1002/lary.23622 Stephen JK, Chen KM, Shah V, Havard S, Lu M, Schweitzer VP, Gardner G,Worsham MJ, 2012, Human papillomavirus outcomes in an access-to-care laryngeal cancer cohort. Otolaryngol Head Neck Surg 146(5):730–738., DOI 10.1177/0194599811434482 Vidal L, Gillison ML, 2008, Human papillomavirus in HNSCC: recognition of a distinct disease type. Hematol Oncol Clin N Am 22(6):1125–1142., DOI 10.1016/j.hoc.2008.08.006, vii Zandberg DP, Bhargava R, Badin S, Cullen KJ, 2013, The role of human papillomavirus in nongenital cancers. CA Cancer J Clin 63(1):57–81., DOI 10.3322/caac.21167 Vairaktaris E,Yapijakis C, PsyrriA, Spyridonidou S,Yannopoulos A, Lazaris A, Vassiliou S, Ferekidis E, Vylliotis A, Nkenke E, Patsouris E, 2007, Loss of tumour suppressor p16 expression in initial stages of oral oncogenesis. Anticancer Res 27(2):979–984 Morshed K, 2010, Association between human papillomavirus infection and laryngeal squamous cell carcinoma. J Med Virol 82(6): 1017–1023., DOI 10.1002/jmv.21749 Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tan PF, Westra WH, Chung CH, Jordan RC, Lu C, Kim H, Axelrod R, Silverman CC, Redmond KP, Gillison ML, 2010, Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 363(1):24–35., DOI 10.1056/NEJMoa0912217 Fischer CA, Kampmann M, Zlobec I, Green E, Tornillo L, Lugli A, WolfensbergerM, Terracciano LM, 2010, p16 expression in oropharyngeal cancer: its impact on staging and prognosis compared with the conventional clinical staging parameters. Ann Oncol 21(10): 1961–1966., DOI 10.1093/annonc/mdq210 Lewis JS Jr, Carpenter DH, Thorstad WL, Zhang Q, Haughey BH, 2011, Extracapsular extension is a poor predictor of disease recurrence in surgically treated oropharyngeal squamous cell carcinoma. Mod Pathol 24(11):1413–1420., DOI 10.1038/modpathol.2011.105 Haughey BH, Sinha P, 2012, Prognostic factors and survival unique to surgically treated p16+ oropharyngeal cancer. Laryngoscope 122(Suppl 2):S13–S33., DOI 10.1002/lary.23493 Chernock RD,Wang X, Gao G, Lewis JS Jr, Zhang Q, ThorstadWL, El-Mofty SK, 2013, Detection and significance of human papillomavirus, CDKN2A(p16, and CDKN1A(p21, expression in squamous cell carcinoma of the larynx. Mod Pathol 26(2):223–231., DOI 10.1038/modpathol.2012.159 Lukas J, Sorensen CS, Lukas C, Santoni-Rugiu E, Bartek J, 1999, p16INK4a, but not constitutively active pRb, can impose a sustained G1 arrest: molecular mechanisms and implications for oncogenesis. Oncogene 18(27):3930–3935., DOI 10.1038/sj.onc.1202777 Swellam M, El-Arab LR, Adly A, 2008, Prognostic value of cellcycle regulators and cellular biomarkers in laryngeal squamous cell carcinoma. Clin Biochem 41(13):1059–1066., DOI 10.1016/j. clinbiochem.2008.06.001 Rother J, Jones D, 2009, Molecular markers of tumor progression in melanoma. Curr Genomics 10(4):231–239., DOI 10.2174/ 138920209788488526 Sanchez-Fayos Calabuig P, Martin Relloso MJ, Porres Cubero JC, 2007, Genetic profile and molecular bases of pancreatic carcinogenesis. Gastroenterol Hepatol 30(10):592–596 Ye Y,WangD, Su C, Rong T,GuoA(2009, Combined detection of p53, p16, Rb, and EGFR mutations in lung cancer by suspension microarray. Genet Mol Res 8(4):1509–1518., DOI 10.4238/vol8-4gmr627 Orlow I, Lacombe L, Hannon GJ, SerranoM, Pellicer I, Dalbagni G, Reuter VE, Zhang ZF, Beach D, Cordon-Cardo C, 1995, Deletion of the p16 and p15 genes in human bladder tumors. J Natl Cancer Inst 87(20):1524–1529 Rocco JW, Sidransky D, 2001, p16(MTS-1/CDKN2/INK4a, in cancer progression. Exp Cell Res 264(1):42–55., DOI 10.1006/excr.2000. 5149 Tarakji B, Kujan O, Nassani MZ, 2010, An immunohistochemical study of the distribution of p 16 protein in oral mucosa in smokers, non-smokers and in frictional keratosis. Med Oral Patol Oral Cir Bucal 15(5):e681–e684 Abrahao AC, Bonelli BV, Nunes FD, Dias EP, Cabral MG, 2011, Immunohistochemical expression of p53, p16 and hTERT in oral squamous cell carcinoma and potentially malignant disorders. Braz Oral Res 25(1):34–41 Koscielny S, Dahse R, Ernst G, von Eggeling F, 2007, The prognostic relevance of p16 inactivation in head and neck cancer. ORL J Otorhinolaryngol Relat Spec 69(1):30–36., DOI 10.1159/000096714 van der Riet P, Nawroz H, Hruban RH, Corio R, Tokino K, KochW, Sidransky D, 1994, Frequent loss of chromosome 9p21-22 early in head and neck cancer progression. Cancer Res 54(5):1156–1158 Gonzalez MV, Pello MF, Lopez-Larrea C, Suarez C, Menendez MJ, Coto E, 1997, Deletion and methylation of the tumour suppressor gene p16/CDKN2 in primary head and neck squamous cell carcinoma. J Clin Pathol 50(6):509–512 El-Naggar AK, Lai S, Clayman G, Lee JK, Luna MA, Goepfert H, Batsakis JG, 1997, Methylation, a major mechanism of p16/CDKN2 gene inactivation in head and neck squamous carcinoma.AmJ Pathol 151(6):1767–1774 Wong TS, Gao W, Li ZH, Chan JY, Ho WK, 2012, Epigenetic dysregulation in laryngeal squamous cell carcinoma. J Oncol 2012: 73946110., DOI 10.1155/2012/739461 Mendelsohn AH, Lai CK, Shintaku IP, Elashoff DA, Dubinett SM, Abemayor E, St John MA, 2010, Histopathologic findings of HPV and p16 positive HNSCC. Laryngoscope 120(9):1788–1794., DOI 10. 1002/lary.21044 Fu ZJ, Ma ZY, Wang QR, Lei DP, Wang R, Liu CX, Pan XL, 2008, Overexpression of CyclinD1 and underexpression of p16 correlate with lymph node metastases in laryngeal squamous cell carcinoma in Chinese patients. Clin Exp Metastasis 25(8):887–892., DOI 10.1007/ s10585-008-9207-x Duray A, Descamps G, Arafa M, Decaestecker C, Remmelink M, Sirtaine N, Ernoux-Neufcoeur P, Mutijima E, Somja J, Depuydt CE, Delvenne P, Saussez S, 2011, High incidence of high-risk HPV in benign and malignant lesions of the larynx. Int J Oncol 39(1):51–59., DOI 10.3892/ijo.2011.1031 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 909 EP 915 DI 10.1007/s12253-014-9773-y PG 7 ER PT J AU Hong, Z Bi, A Chen, D Gao, L Yin, Z Luo, L AF Hong, Zhuan Bi, Aijing Chen, Dan Gao, Li Yin, Zhimin Luo, Lan TI Activation of Hedgehog Signaling Pathway in Human Non-small Cell Lung Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Shh; Ptch-1; Gli-1; Non-small cell lung cancer; Survival ID Shh; Ptch-1; Gli-1; Non-small cell lung cancer; Survival AB The activation of the hedgehog pathway, which is an important signaling mechanism crucial in embryogenesis, has strong links to carcinogenesis. Aberrant regulation of this pathway can result in the development of tumors. The present study was designed to investigate Hh related protein expression in non-small cell lung cancers. Fifty five non-small cell lung cancers samples were used in the study. By reverse transcription-polymerase chain reaction (RT-PCR), the expression of Shh, Ptch-1, and Gli-1 in tumor and adjacent normal tissues was examined and associated to clinical pathologic features. The expression levels of Shh, Ptch-1, Gli-1 in non-small cell lung cancer tissues were 63.64, 69.09, 43.64%, respectively, higher than that in the adjacent normal tissues. Survival analysis showed that both Ptch-1 and Gli-1 expression were associated with poor survival (both P<0.05, logrank test). Shh and Ptch-1 expression were correlated with lymph node metastasis. These results suggest that dysregulation of Hh signaling pathway plays an important role in the development of human NSCLCs. The expression of Ptch-1 and Gli-1 is possibly involved in NSCLCs progression, which may be a useful prognostic indicator of NSCLCs. C1 [Hong, Zhuan] Nanjing Normal University, College of Life Science, Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, 22 Hankou Road, 210093 Nanjing, China. [Bi, Aijing] Nanjing Normal University, College of Life Science, Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, 22 Hankou Road, 210093 Nanjing, China. [Chen, Dan] Nanjing Normal University, College of Life Science, Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, 22 Hankou Road, 210093 Nanjing, China. [Gao, Li] Nanjing Normal University, College of Life Science, Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, 22 Hankou Road, 210093 Nanjing, China. [Yin, Zhimin] Nanjing Normal University, College of Life Science, Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, No. 1 Wenyuan Road, 210046 Nanjing, China. [Luo, Lan] Nanjing Normal University, College of Life Science, Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, 22 Hankou Road, 210093 Nanjing, China. RP Luo, L (reprint author), Nanjing Normal University, College of Life Science, Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, 210093 Nanjing, China. EM lanluo@nju.edu.cn CR Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60:277–300 Herbst RS, Heymach JV, Lippman SM(2008, Lung cancer. N Engl J Med 359:1367–1380 Jemal A, Murray T, Ward E, Samuels A, TIWARI RC et al, 2005, Cancer statistics, 2005. CA Cancer J Clin 55:10–30 Pasca di Magliano M, Hebrok M, 2003, Hedgehog signaling in cancer formation and maintenance. Nat Rev Cancer 3:903–911 Taipale J, Beachy PA, 2001, The hedgehog and Wnt signaling pathways in cancer. Nature 411:349–354 Berman DM, Karhadkar SS, Hallahan AR, Pritchard JI, Eberhart CG et al, 2002, Medulloblastoma growth inhibition by hedgehog pathway blockade. Science 297:1559–1561 Ingham PW, Mcmahon AP, 2001, Hedgehog signaling in animal development: paradigms and principles. Genes Dev 15:3059–3087 Chari NS, McDonnell TJ, 2007, The sonic hedgehog signaling network in development and neoplasia. Adv Anat Pathol 14:344–352 Katoh Y, Katoh M, 2009, Hedgehog target genes: mechanisms of carcinogenesis induced by aberrant hedgehog signaling activation. Curr Mol Med 9:873–886 Rubin LL, de Sauvage FJ, 2006, Targeting the hedgehog pathway in cancer. Nat Rev Drug Discov 5:1026–1033 Evangelista M, Tian H, de Sauvage FJ, 2006, The hedgehog signaling pathway in cancer. Clin Cancer Res 12:5924–5928 Watkins DN, Berman DM, Burkholder SG,Wang B, Beachy PA et al, 2003, Hedgehog signaling within airway epithelial progenitors and in small-cell lung cancer. Nature 422:313–317 Schneider S, Thurnher D, Kloimstein P, Leitner V, Petzelbauer P et al, 2011, Expression of the sonic hedgehog pathway in squamous cell carcinoma of the skin and the mucosa of the head and neck. Head Neck 33:244–250 Altaba AR, Sanchez P, Dahmane N, 2002, Gli and hedgehog in cancer: tumours, embryos and stem cell. Nat Rev Cancer 2:361–372 Athar M, Tang X, Lee JL, Kopelovich L, Kim AL, 2006, Hedgehog signaling in skin development and cancer. Exp Dermatol 15:667–677 Dahman N, Lee J, Robins P, Heller P, Ruiz I, Altaba A, 1997, Activation of the transcription factor Gli-1 and the sonic hedgehog signaling pathway in skin tumours. Nature 389:876–881 Xie J, Murone M, Luoh SM, Ryan A, Gu Q et al, 1998, Activating smoothened mutations in sporadic basal-cell carcinoma. Nature 391: 90–92 Chi S, Huang S, Li C, Zhang X, He N et al, 2006, Activation of the hedgehog pathway in a subset of lung cancers. Cancer Lett 244:53– 60 Hahn H, Wicking C, Zaphiropoulous PG, Gailani MR, Shanley S et al, 1996, Mutations of the human homolog of drosophila patched in the nevoid basal cell carcinoma syndrome. Cell 85:841–851 Lauth M, Toftgard R, 2007, Non-canonical activation of GLI transcription factors: implications for targeted anti-cancer therapy. Cell Cycle 6:2458–2463 He HC, Chen JH, Chen XB, Qin GQ, Cai C et al, 2012, Expression of hedgehog pathway components is associated with bladder cancer progression and clinical outcome. Pathol Oncol Res 18:349–355 ten Haaf A, Bektas N, von Serenyi S, Losen I, Arweiler EC et al, 2009, Expression of the glioma-associated oncogene homolog, GLI, 1 in human breast cancer is associated with unfavourable overall survival. BMC Cancer 9:298 Che L, Yuan YH, Jia J, Ren J, 2012, Activation of sonic hedgehog signaling pathway is an independent potential prognosis predictor in human hepatocellular carcinoma patients. Chin J Cancer Res 24:323– 331 Liao X, Siu MK, Au CW, Wong ES, Chan HYet al, 2009, Aberrant activation of hedgehog signaling pathway in ovarian cancers: effect on prognosis, cell invasion and differentiation. Carcinogenesis 30: 131–140 High A, Zedan W, 2005, Basal cell nevus syndrome. Curr Opin Oncol 17:160–166 Wetmore C, 2003, Sonic hedgehog in normal and neoplastic proliferation: insight gained from human tumors and animal models. Curr Opin Genet Dev 13:34–42 KasperM, Regl G, Frischauf AM, Aberger F, 2006, Gli transcription factor: mediators of oncogenic hedgehog signaling. Eur J Cancer 4: 437–445 Bailey JM, Mohr AM, Hollingsworth MA, 2009, Sonic hedgehog paracrine signaling regulates metastases and lymphangiogenesis in pancreatic cancer. Oncogene 28:3513–3525 Sanchez P, Hernandez AM, Stecca B, Kahler AJ, Degueme AM et al, 2004, Inhibition of prostate cancer proliferation by interference with SONIC HEDGEHOG-GLI1 signaling. Proc Natl Acad Sci U S A 101:12561–12366 Yoo YA, Kang MH, Lee HJ, Kim BH, Park JK et al, 2011, Sonic hedgehog pathway promotes metastasis and lymphangiogenesis via activation of Akt, EMT, and MMP-9 pathway in gastric cancer. Cancer Res 71:7061–7070 Bulk E, Sargin B, Krug U, Hascher A, Jun Y et al, 2009, S100A2 inducesmetastasis inNon-small cell lung cancer. Clin Cancer Res 15: 22–29 Pignon JP, Tribodet H, Scagliotti GV, Douillard JY, Shepherd FA et al, 2008, Lung adjuvant cisplatin evaluation: a pooled analysis by the LACE collaborative group. J Clin Oncol 26:3552–3559 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 917 EP 922 DI 10.1007/s12253-014-9774-x PG 6 ER PT J AU Kis, A Tatar, ZsT Gall, T Boda, R Tar, I Major, T Redl, P Gergely, L Szarka, K AF Kis, Andrea Tatar, Zsofia Timea Gall, Tamas Boda, Robert Tar, Ildiko Major, Tamas Redl, Pal Gergely, Lajos Szarka, Krisztina TI Frequency of Genetic and Epigenetic Alterations of p14ARF and p16INK4A in Head and Neck Cancer in a Hungarian Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral squamous cell cancer; Laryngeal squamous cell cancer; Tumour suppressor gene; Promoter methylation ID Oral squamous cell cancer; Laryngeal squamous cell cancer; Tumour suppressor gene; Promoter methylation AB Occurrence of genetic and epigenetic alterations affecting p14ARF and p16INK4A were investigated in tumour samples of 37 oral (OSCC) and 28 laryngeal squamous cell cancer (LSCC) patients, and compared to exfoliated buccal epithelial cells of 68 healthy controls. Presence of deletions andmutations/polymorphisms affecting exons were examined using sequencing. Methylation status of promoters was assessed by methylation-specific PCR. Chi-square and Fisher’s exact tests were used to compare frequency of events. Exon deletions were found in four controls, one OSCC and 22 LSCC patients; the latter significantly differed from controls (p<0.001). Only two mutations (T24610A and C24702A) were in p16 exon 1 of two OSCC patients. Polymorphisms G28575A (Ala140Thr), G31292C (C540G) and G28608A were found in both patient groups. The p14 promoter was unmethylated in 86.7 % of OSCC and in 85.7 % of LSCC patients; for the p16 promoter these rates were 69.0 % and 76.2 % for OSCC and LSCC patients, respectively. Combining the two patient groups, unmethylated promoter was significantly less frequent in case of both p14 and p16 (p=0.043 and p=0.001, respectively) compared to the control group. In summary, exon deletion may be important in LSCC, while promoter methylation was relatively frequent in both patient groups. C1 [Kis, Andrea] University of Debrecen, Faculty of Medicine, Department of Medical Microbiology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Tatar, Zsofia Timea] University of Debrecen, Faculty of Medicine, Department of Medical Microbiology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Gall, Tamas] University of Debrecen, Faculty of Medicine, Department of Medical Microbiology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Boda, Robert] University of Debrecen, Faculty of Dentistry, Department of Oral and Maxillofacial Surgery, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Tar, Ildiko] University of Debrecen, Faculty of Dentistry, Department of Periodontology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Major, Tamas] University of Debrecen, Faculty of Medicine, Department of Medical Microbiology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Redl, Pal] University of Debrecen, Faculty of Dentistry, Department of Oral and Maxillofacial Surgery, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Gergely, Lajos] University of Debrecen, Faculty of Medicine, Department of Medical Microbiology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Szarka, Krisztina] University of Debrecen, Faculty of Medicine, Department of Medical Microbiology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. RP Szarka, K (reprint author), University of Debrecen, Faculty of Medicine, Department of Medical Microbiology, H-4032 Debrecen, Hungary. EM szkrisz@med.unideb.hu CR Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global Cancer Statistics, 2002. CA Cancer J Clin 55:74–108 Calmon MF, Colombo J, Carvalho F, Souza FP, Filho JF et al, 2007, Methylation Profile of Genes CDKN2A, p14 and p16), DAPK1, CDH1, and ADAM23 in Head and Neck Cancer. Cancer Genet Cytogenet 173:31–37 Demokan S, Dalay N, 2011, Role of DNA Methylation in Head and Neck Cancer. Clin Epigenet 2:123–150 Warnakulasuriya S, Sutherland G, Scully C, 2005, Tobacco, Oral Cancer, and Treatment of Dependence. Oral Oncol 41:244–260 Freedman ND, Abnet CC, Leitzmann MF, Hollenbeck AR, Schatzkin A, 2007, Prospective Investigation of the Cigarette Smoking-Head and Neck Cancer Association by Sex. Cancer 110: 1593–1601 Curado MP, HashibeM(2009, Recent Changes in the Epidemiology of Head and Neck Cancer. Curr Opin Oncol 21:195–200 Scėsnaitė A, Gudlevicienė Z, Jarmalaitė S, 2008, Promoter Hypermethylation of Tumour Suppressor Genes in Tumour Cells from Patients with Head and Neck Cancer. Biologija 54: 161–166 Andrews E, SeamanWT,Webster-Cyriaque J, 2009, Oropharyngeal Carcinoma in non-Smokers and non-Drinkers: A Role for HPV. Oral Oncol 45:486–491 Sand LP, Jalouli J, Larsson PA, Hirsch JM, 2002, Prevalence of Epstein-Barr Virus in Oral Cell Carcinoma, Oral Lichen Planus, and Normal Oral Mucosa. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 93:586–592 Shimakage M, Horii K, Tempaku A, Kakudo K, Shirasaka T et al, 2002, Association of Epstein-Barr Virus with Oral Cancers. Hum Pathol 33:608–614 Mao L, Hong WK, Papadimitrakopoulou VA, 2004, Focus on Head and Neck Cancer. Cancer Cell 5:311–316 Hansson BG, Rosenquist K, Antonsson A,Wennerberg J, Schildt EB et al, 2005, Strong Association Between Infection with Human Papillomavirus and Oral and Oropharyngeal Squamous Cell Carcinoma: A Population-Based Case–Control Study in Southern Sweden. Acta Otolaryngol 125:1337–1344 Kreimer AR, Clifford GM, Boyle P, Franceschi S, 2005, Human Papillomavirus Types in Head and Neck Squamous Cell Carcinomas Worldwide: A Systematic Review. Cancer Epidemiol Biomarkers Prev 14:467–475 Sherr CJ, 2001, The INK4a/ARF Network in Tumour Suppression. Nat Rev Mol Cell Biol 2:731–737 Sharpless NE, DePinho RA, 1999, The INK4A/ARF Locus and its two Gene Products. Curr Opin Genet Dev 9:22–30 Gallagher SJ, Kefford RF, Rizos H, 2006, The ARF Tumour Suppressor. Int J Biochem Cell Biol 38:1637–1641 Belinsky SA, Nikula KJ, Palmisano WA, Mchels R, Saccomanno G et al, 1998, Aberrant Methylation of p16INK4a in an Early Event in Lung Cancer and a Potential Biomarker for Early Diagnosis. Proc Natl Acad Sci 95:11891–11896 Shintani S, Nakahara Y, Mihara M, Ueyama Y, Matsumura T, 2001, Inactivation of the p14ARF, p15INK4B and p16INK4A Genes is a Frequent Event in Human Oral Squamous Cell Carcinomas. Oral Oncol 37:498–504 Kim BN, Yamamoto H, Ikeda K, Damdinsuren B, Sugita Y et al, 2005, Methylation and Expression of p16INK4 Tumor Suppressor Gene in Primary Colorectal Cancer Tissues. Int J Oncol 26: 1217–1226 Sailasree R, Abhilash A, Sathyan KM, Nalinakumari KR, Thomas S et al, 2008, Differential Roles of p16INK4A and p14ARF Genes in Prognosis of Oral Carcinoma. Cancer Epidemiol Biomarkers Prev 17:414–420 Jaffrain-Rea ML, Ferretti E, Toniato E, Cannita K, Santoro A et al, 1999, P16, INK4a, MTS-1, Gene Polymorphism and Methylation Status in Human Pituitary Tumours. Clin Endocrinol 51:317–325 Yan L, Na W, Shank K, Xiao-Wei M, Wei G et al, 2008, P16, CDKN2, Gene Polymorphism: Association with Histologic Subtypes of Epithelial Ovarian Cancer in China. Int J Gynecol Cancer 18:30–35 Thakur N, Hussain S, Nasare V, Das BC, Basir SF et al, 2012, Association Analysis of p16, CDKN2A, and RB1 Polymorphisms with Susceptibility to Cervical Cancer in Indian Population.Mol Biol Rep 39:407–414 Chen J, Li D, Wei C, Sen S, Killary AM et al, 2007, Aurora-A and p16 Polymorphisms Contribute to an Earlier age at Diagnosis of Pancreatic Cancer in Caucasians. Clin Cancer Res 13:3100–3104 Chen J, Li D, Killary AM, Sen S, Amos CI et al, 2009, Polymorphisms of p16, p27, p73, and MDM2 Modulate Response and Survival of Pancreatic Cancer Patients Treated with Preoperative Chemoradiation. Ann Surg Oncol 16:431–439 Sauroja I, Smeds J, Vlaykova T, Kumar R, Talve L et al, 2000, Analysis of G1/S Checkpoint Regulators in Metastatic Melanoma. Genes Chromosomes Cancer 28:404–414 Zheng Y, Shen H, Sturgis EM, Wang LE, Shete S et al, 2002, Haplotypes of two Variants in p16, CDKN2/MTS-1/INK4a, Exon 3 and Risk of Squamous Cell Carcinoma of the Head and Neck: A Case–Control Study. Cancer Epidemiol Biomarkers Prev 11: 640–645 Kis A, Feher E, Gall T, Tar I, Boda R et al, 2009, Epstein-Barr Virus Prevalence in Oral Squamous Cell Cancer and in Potentially Malignant Disorders in an Eastern Hungarian Population. Eur J Oral Sci 117:536–540 Szarka K, Tar I, Feher E, Gall T, Kis A et al, 2009, Progressive Increase of Human Papillomavirus Carriage Rates in Potentially Malignant and Malignant Oral Disorders with Increasing Malignant Potential. Oral Microbiol Immunol 24:314–318 Baur AS, Shaw P, Burri N, Delacretaz F, Bosman FT et al, 1999, Frequent Methylation Silencing of p15INK4b, MTS2, and p16INK4a, MTS1, in B-Cell and T-Cell Lymphomas. Blood 94: 1773–1781 Chen TC, Hsieh LL, Kuo TT, Ng KF,Wu Chou YH et al, 2000, P16 INK4 Gene Mutation and Allelic Loss of Chromosome 9p21-22 in Taiwanese Hepatocellular Carcinoma. Anticancer Res 20:1621–1626 Nagy E, Beck Z, Kiss A, Csoma E, Telek B et al, 2003, Frequent Methylation of p16INK4A and p14ARF Genes Implicated in the Evolution of Chronic Myeloid Leukaemia from its Chronic to Accelerated Phase. Eur J Cancer 39:2298–2305 Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB, 1996, Methylation-Specific PCR: A Novel PCR Assay for Methylation Status of CpG Islands. Proc Natl Acad Sci 93:9821–9826 Ruesga MT, Acha-Sagredo A, Rodriguez MJ, Aguirregaviria JI, Videgain J et al, 2007, P16INK4a Promoter Hypermethylation in Oral Scrapings of Oral Squamous Cell Carcinoma Risk Patients. Cancer Lett 250:140–145 Reed AL, Califano J, Cairns P, Westra WH, Jones RM et al, 1996, High Frequency of p16, CDKN2/MTS-1/INK4A, Inactivation in Head and Neck Squamous Cell Carcinoma. Cancer Res 56:3630–3633 Nakahara Y, Shintani S, Mihara M, Ueyama Y, Matsumura T, 2001, High Frequency of Homozygous Deletion and Methylation of p16INK4A Gene in Oral Squamous Cell Carcinomas. Cancer Lett 163:221–228 Bradley G, Irish J, MacMillan C, Mancer K, Witterick I et al, 2001, Abnormalities of the ARF-p53 Pathway in Oral Squamous Cell Carcinoma. Oncogene 20:654–658 Lin SC, Chang KW, Chang CS, Liu TY, Tzeng YS et al, 2000, Alterations of p16/MTS1 Gene in Oral Squamous Cell Carcinomas from Taiwanese. J oral Pathol Med 29:159–166 Ohta S, Uemura H, Matsui Y, Ishiguro H, Fujinami K et al, 2009, Alterations of p16 and p14ARF Genes and Their 9p21 Locus in Oral Squamous Cell Carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 107:81–91 Weber A, Bellmann U, Bootz F, Wittekind C, Tannapfel A, 2002, INK4a-ARF Alterations and p53 Mutations in Primary and Consecutive Squamous Cell Carcinoma of the Head and Neck. Virchows Arch 441:133–142 Kulkarni V, Saranath D, 2004, Concurrent Hypermethylation of Multiple Regulatory Genes in Chewing Tobacco Associated Oral Squamous Cell Carcinomas and Adjacent Normal Tissues. Oral Oncol 40:145–153 Salam I, Hussain S, Mir MM, Dar NA, Abdullah S et al, 2009, Aberrant Promoter Methylation and Reduced Expression of p16 Gene in Esophageal Squamous Cell Carcinoma from Kashmir Valley: A High-Risk Area. Mol Cell Biochem 332:51–58 Hall GL, Shaw RJ, Field EA, Rogers SN, Sutton DN et al, 2008, P16 Promoter Methylation is a Potential Predictor of Malignant Transformation in Oral Epithelial Dysplasia. Cancer Epidemiol Biomarkers Prev 17:2174–2179 Kondo Y, Shen L, Issa JP, 2003, Critical Role of Histone Methylation in Tumor Suppressor Gene Silencing in Colorectal Cancer. Mol Cell Biol 23:206–215 Suzuki H, Maruyama R, Yamamoto E, Kai M, 2013, Epigenetic Alteration and microRNA Dysregulation in Cancer. Front Genet 4:258 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 923 EP 929 DI 10.1007/s12253-014-9775-9 PG 7 ER PT J AU Ye, Q Chen, L Yin, X Liu, JChY Ji, Q Zhao, E AF Ye, Qingqing Chen, Li Yin, Xiaolu Liu, Jie Charles Yuan Ji, Qunsheng Zhao, Enfeng TI Development of Serous Ovarian Cancer is Associated with the Expression of Homologous Recombination Pathway Proteins SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Serous ovarian cancer (SOC); Homologous recombination (HR); Immunohistochemistry (IHC); Meiotic recombination 11 (Mre11); Mediator of DNA damage checkpoint protein 1 (MDC1); Ataxia telangiectasia mutated (ATM); ATM-Rad3-related (ATR); Breast cancer susceptibility gene 1 (BRCA1) ID Serous ovarian cancer (SOC); Homologous recombination (HR); Immunohistochemistry (IHC); Meiotic recombination 11 (Mre11); Mediator of DNA damage checkpoint protein 1 (MDC1); Ataxia telangiectasia mutated (ATM); ATM-Rad3-related (ATR); Breast cancer susceptibility gene 1 (BRCA1) AB To investigate the expressions of key markers in the homologous recombination (HR) pathway and the correlation with clinicopathological parameters in serous ovarian cancer (SOC). We analyzed the protein expression of MRE11, MDC1, ATM, ATR and BRCA1 by immunohistochemistry (IHC) in 97 SOC samples, and correlated with clinical parameters including age, tumor grades, clinical stage, status of menstruation and chemotherapy. Low expression of MRE11 and MDC1 was detected in 14.4 % and 3.1 % of the patient samples, and negative expression of ATM, ATR and BRCA1 was found in 11.3 %, 6.3 % and 29.9 % of the patient samples, respectively. ATR deficiency was significantly associated with menopause (P=0.025), strong expression of ATM (P=0.017) and MRE11 (P=0.040) with premenopausal SOC, strong expression of MRE11 (P=0.016) with low tumor grade, and strong expression of BRCA1 (P=0.015) with early clinical stage. In addition, low expression of MRE11 was strongly associated with negativity of ATR (P<0.001) and BRCA1 (P= 0.004) Furthermore, ATR deficiency was associated with low expression of ATM (P=0.028) and loss expression of BRCA1 (P=0.009). Our results suggest that reduced expression or loss of proteins in HR pathway is associated with SOC development. Abnormality of MRE11 and BRCA1 are strongly associated with late clinical stage in SOC patients. C1 [Ye, Qingqing] AstraZeneca Global R&D, Innovation Center ChinaShanghai, China. [Chen, Li] Chinese PLA General Hospital, Department of Obstetrics & GynecologyBeijing, China. [Yin, Xiaolu] AstraZeneca Global R&D, Innovation Center ChinaShanghai, China. [Liu, Jie Charles Yuan] AstraZeneca Global R&D, Innovation Center ChinaShanghai, China. [Ji, Qunsheng] AstraZeneca Global R&D, Innovation Center ChinaShanghai, China. [Zhao, Enfeng] Hainan Branch of Chinese PLA General Hospital, Department of Obstetrics & Gynecology, 572014 Sanya, Hainan, China. RP Zhao, E (reprint author), Hainan Branch of Chinese PLA General Hospital, Department of Obstetrics & Gynecology, 572014 Sanya, China. EM 301.zhao@163.com CR Van Gent DC, Hoeijmakers JHJ, Kanaar R, 2001, Chromosomal stability and the DNA double-stranded break connection. Nat Rev Genet 2(3):196–206 Watson JD, 1970, Molecular biology of the gene.Molecular biology of the gene, 2nd edn) Haber JE, 2000, Partners and pathways: repairing a double-strand break. Trends Genet 16(6):259–264 ShrivastavM, De Haro LP, Nickoloff JA, 2007, Regulation of DNA double-strand break repair pathway choice. Cell Res 18(1):134–147 Carson CT, Schwartz RA, Stracker TH, Lilley CE, Lee DV, Weitzman MD, 2003, The Mre11 complex is required for ATM activation and the G2/M checkpoint. The EMBO J 22(24):6610–6620 Goldberg M, Stucki M, Falck J, D’Amours D, Rahman D, Pappin D, Bartek J, Jackson SP, 2003, MDC1 is required for the intra-S-phase DNA damage checkpoint. Nature 421(6926): 952–956 Stewart GS, Wang B, Bignell CR, Taylor AMR, Elledge SJ, 2003, MDC1 is a mediator of the mammalian DNA damage checkpoint. Nature 421(6926):961–966 Abraham RT, 2001, Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev 15(17):2177–2196 Cortez D, Wang Y, Qin J, Elledge SJ, 1999, Requirement of ATM-dependent phosphorylation of brca1 in the DNA damage response to double-strand breaks. Science 286(5442): 1162–1166 Helleday T Homologous recombination in cancer development, treatment and development of drug resistance. Carcinogenesis 31, 6): 955–960 Kastan MB, Bartek J, 2004, Cell-cycle checkpoints and cancer. Nature 432(7015):316–323 McCabe N, Turner NC, Lord CJ, Kluzek K, Bialkowska A, Swift S, Giavara S, O’Connor MJ, Tutt AN, Zdzienicka MZ, 2006, Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly, ADP-ribose, polymerase inhibition. Cancer Res 66(16):8109 Hsu HM, Wang HC, Chen ST, Hsu GC, Shen CY, Yu JC, 2007, Breast cancer risk is associated with the genes encoding the DNA double-strand break repair Mre11/Rad50/Nbs1 complex. Cancer Epidemiol Biomark Prev 16(10):2024–2032 Bartkova ZH J, 2007, DNA damage response mediators MDC1 and 53BP1: constitutive activation and aberrant loss in breast and lung cancer, but not in testicular germ cell tumours. Oncogene 26(53): 7414–7422 Giannini G, Rinaldi C, Ristori E, Ambrosini MI, Cerignoli F, Viel A, Bidoli E, Berni S, D’Amati G, Scambia G, 2004, Mutations of an intronic repeat induce impaired MRE11 expression in primary human cancer with microsatellite instability. Oncogene 23(15):2640–2647 Giannini G, Ristori E, Cerignoli F, Rinaldi C, Zani M, Viel A, Ottini L, Crescenzi M, Martinotti S, Bignami M, 2002, Human MRE11 is inactivated in mismatch repair-deficient cancers. EMBO Rep 3(3): 248–254 Liu ZK, Zhu SC, Wang YX, 2007, Expression of DNA damage checkpoint mediator 1 and p53-binding protein 1 in human esophageal cancer cell lines TE-1, TE-13, and Eca109]. Nan fang yi ke da xue xue bao=. J of South Med Univ 27(9):1314 Shiloh Y, Kastan MB, 2001, ATM: genome stability, neuronal development, and cancer cross paths. Adv Cancer Res 83:209–254 Chenevix-Trench G, Spurdle AB, Gatei M, Kelly H, Marsh A, Chen X, Donn K, Cummings M, Nyholt D, Jenkins MA, 2002, Dominant negative ATMmutations in breast cancer families. J Natl Cancer Inst 94(3):205–215 Thorstenson YR, Roxas A, Kroiss R, JenkinsMA, Yu KM, Bachrich T, Muhr D, Wayne TL, Chu G, Davis RW, 2003, Contributions of ATM mutations to familial breast and ovarian cancer. Cancer Res 63(12):3325 Durocher F, Labrie Y, Soucy P, Sinilnikova O, Labuda D, Bessette P, Chiquette J, Laframboise R, Lepine J, Lesperance B, 2006)Mutation analysis and characterization of ATR sequence variants in breast cancer cases from high-risk French Canadian breast/ovarian cancer families. BMC Cancer 6(1):230 Geisler JP, Hatterman-Zogg MA, Rathe JA, Buller RE, 2002, Frequency of BRCA1 dysfunction in ovarian cancer. J Natl Cancer Inst 94(1):61–67 Welcsh PL, King MC, 2001, BRCA1 and BRCA2 and the genetics of breast and ovarian cancer. Hum Mol Genet 10(7): 705–713 Jemal A, Siegel R, Ward E, Murray T, Xu J, Smigal C, Thun MJ, 2006, Cancer statistics, 2006. CA Cancer J clin 56(2):106–130 Auranen A, Song H, Waterfall C, DiCioccio RA, Kuschel B, Kjaer SK, Hogdall E, Hogdall C, Stratton J, Whittemore AS, 2005, Polymorphisms in DNA repair genes and epithelial ovarian cancer risk. Int J Cancer 117(4):611–618 Powell SN, Willers H, Xia F, 2002, BRCA2 Keeps Rad51 in Line: High-Fidelity Homologous Recombination Prevents Breast and Ovarian Cancer? Mol Cell 10(6):1262–1263 Heikkinen K, Mansikka V, Karppinen SM, Rapakko K, Winqvist R, 2005, Mutation analysis of the ATR gene in breast and ovarian cancer families. Breast Cancer Res 7(4):R495–501 Frank TS, Manley SA, Olopade OI, Cummings S, Garber JE, Bernhardt B, Antman K, Russo D, Wood ME, Mullineau L, 1998, Sequence analysis of BRCA1 and BRCA2: correlation of mutations with family history and ovarian cancer risk. J Clin Oncol 16(7): 2417–2425 Brown PO, Palmer C, 2009, The preclinical natural history of serous ovarian cancer: defining the target for early detection. PLoS Med 6(7):e1000114 Ries LAG, Young JL, Keel GE, Eisner MP, Lin YD, Horner MJ, 2007, SEER Survival Monograph: Cancer Survival Among Adults: US SEER Program, 1988–2001, Patient and Tumor Characteristics. National Cancer Institute, SEER Program, NIH Pub, 07–6215) Esteban Cardenosa E, Bolufer Gilabert P, de Juan Jimenez I, Palanca Suela S, Barragan Gonzalez E, Gonzalez Anguix V, Lerma Alejos E, Chirivella Gonzalez I, Segura Huerta A, Guillen Ponce C, Relationship of BRCA1 and BRCA2 mutations with cancer burden in the family and tumor incidence. Familial cancer 9, 3):291–295 Lux MP, Fasching PA, Beckmann MW, 2006, Hereditary breast and ovarian cancer: review and future perspectives. J MolMed 84(1):16–28 Kadouri L, Hubert A, Rotenberg Y, Hamburger T, Sagi M, Nechushtan C, Abeliovich D, Peretz T, 2007, Cancer risks in carriers of the BRCA1/2 Ashkenazi founder mutations. J Med Genet 44(7): 467–471 Thompson D, Easton DF, 2002, Cancer incidence in BRCA1 mutation carriers. J Natl Cancer Inst 94(18):1358–1365 W-b L, Ao L, Z-h C, Z-y Z, Y-h Z, X-y Y, Y-l X, Cao J, J-y L, 2011, Molecular analysis of DNA repair gene methylation and protein expression during chemical-induced rat lung carcinogenesis. Biochem Biophys Res Commun 408(4):595–601 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 931 EP 938 DI 10.1007/s12253-014-9776-8 PG 8 ER PT J AU Kovacs, P Panczel, G Borbola, K Juhasz, G Liszkay, G AF Kovacs, Peter Panczel, Gitta Borbola, Kinga Juhasz, Gabriella Liszkay, Gabriella TI Psychological Changes in Melanoma Patients During Ipilimumab Treatment Compared to Low-Dose Interferon Alpha Therapy—A Follow-Up Study of First Experiences SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ipilimumab; Interferon; Malignant melanoma; Depression; Anxiety ID Ipilimumab; Interferon; Malignant melanoma; Depression; Anxiety AB Immuntherapies are frequently accompanied by psychological side effects. Our goals were to detect the changes of psychological factors (depression, anxiety) among melanoma patients during ipilimumab treatment. Ten ipilimumab treated melanoma patients (Group 1.) and 18 low-dose interferon-alpha treated patients (Group 2.) were compared. In our longitudinal study we measured depression (Zung Self- Rating Depression Scale) and anxiety (State-Trait Anxiety Inventory, STAI). Psychological status was tested four times: in every 3 week during ipilimumab treatment according to the relevant treatment protocol and at baseline, 1st, 3rd and 6th month of interferon therapy. No significant differences were detected at different timepoints in the level of depression or in the anxiety scale in Group 1. However significant increase of depression was found in Group 2 during the 6 months of the study. Increased levels of anxiety were found in the second timepoint in both treatment groups. This increase was only temporary and the level of anxiety returned to the baseline. In our sample no measurable psychological differences were detectable during the 12 weeks treatment period of ipilimumab. Ipilimumab seems to have fewer psychological side-effects compared to other immune therapies. C1 [Kovacs, Peter] National Institute of Oncology, 7-9. Gyorgy Rath Street, 1122 Budapest, Hungary. [Panczel, Gitta] National Institute of Oncology, 7-9. Gyorgy Rath Street, 1122 Budapest, Hungary. [Borbola, Kinga] National Institute of Oncology, 7-9. Gyorgy Rath Street, 1122 Budapest, Hungary. [Juhasz, Gabriella] Semmelweis University, Department of Pharmacology and PharmacotherapyBudapest, Hungary. [Liszkay, Gabriella] National Institute of Oncology, 7-9. Gyorgy Rath Street, 1122 Budapest, Hungary. RP Kovacs, P (reprint author), National Institute of Oncology, 1122 Budapest, Hungary. EM kovacs.peter@oncol.hu CR Petrella T, Verma S, Spithoff K, McCready D, 2012, Adjuvant interferon therapy for patients at high risk for recurrent melanoma: an updated systematic review and practice guideline. Clin Oncol 26(4):413–423 Gray-Schopfer V, Wellbrock C, Marais R, 2007, Melanoma biology and new targeted therapy. Nature 445(85):1–7 Mocellin S, Pasquali S, Rossi CR, Nitti D, 2010, Interferon alpha adjuvant therapy in patients with high-risk melanoma: a systematic review and meta-analysis. J Natl Cancer Inst 102:493–501 Garbe C, Leiter U, 2009, Melanoma epidemiology and trends. Clin Dermatol 27(1):3–9 Tsao H, Atkins MB, Sober AJ, 2004, Management of cutaneous melanoma. N Engl J Med 351:998–1012 Agarwala SS, 2009, Current systemic therapy for metastatic melanoma. Expert Rev Anticancer Ther 9:587–595 Rusthoven JJ, Quirt IC, Iscoe NA et al, 1996, Randomized, double-blind, placebo-controlled trial comparing the response rates of carmustine, dacarbazine, and cisplatin with and without tamoxifen in patients with metastatic melanoma. J Clin Oncol 14:2083–2090 MiddletonMR, Grob JJ, Aaronson N et al, 2000, Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma. J Clin Oncol 18:158–166 Verma S, Quirt I, McCready D, Bak K, Charrette M, Iscoe N, 2006, Systematic review of systemic adjuvant therapy for patients at high risk for recurrent melanoma. Cancer 106:1431–1442 Robert C, Thomas L, Bondarenko I et al, 2011, Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 364(26):2517–2526 Hodi FS, O’Day SJ, McDermott DF et al, 2010, Improved survival with ipilimumab in patients with metastatic melanoma. N Engl JMed 363:711–723 Garbe C, Peris K, Hauschild A et al, 2012, Diagnosis and treatment of melanoma. European consensus-based interdisciplinary guideline– update. Eur J Cancer 48:2375–2390 Wheatley K, Ives N, Eggermont A, on behalf of International Malignant Melanoma Collaborative Group et al, 2007, Interferon- α as adjuvant therapy for melanoma: an individual patient data metaanalysis of randomised trials. J Clin Oncol 25(suppl 18):8526 Friebe A, Horn M, Schmidt F et al, 2010, Dose-dependent development of depressive symptoms during adjuvant interferon treatment of patients with malignant melanoma. Psychosomatics 51:466–473 Bonaccorso S, Marino V, Biondi M et al, 2002, Depression induced by treatment with interferon- in patients affected by hepatitis C virus. J Affect Disord 72:237–241 Borden EC, Parkinson D, 1998, A perspective on the clinical effectiveness and tolerance of interferon-alpha. Semin Oncol 25:3–8 Dieperink E, Willenbring M, Ho SB, 2000, Neuropsychiatric symptoms associated with hepatitis C and interferon: a review. Am J Psychiatry 157:867–876 Dafny N, Prieto-Gomez B, Reyes-Vazquez C, 1985, Does the immune system communicate with the central nervous system? Interferon modifies central nervous activity. J Neuroimmunol 9:1–12 Schaefer M, Engelbrecht MA, Gut O et al, 2002, Interferon, IFN, and psychiatric syndromes: a review. Prog Neuropsychopharmacol Biol Psychiatry 26:731–746 Zung WWK, 1965, A self-rating depression scale. Arch Gen Psychiatry 12:63–70 Simon A, 1994, A Zung-fele Onertekelo Depresszio Skala. In:Merei F, Szakacs F, eds, Pszichodiagnosztikai Vademecum. Nemzeti Tankonyvkiado, Budapest, pp 180–185 Spielberger CD, Gorsuch RL, Lushene RE, Vagg PR, Jacobs GA, 1983, Manual for the state-trait anxiety inventory. Consulting Psychologists Press, Palo Alto Sipos K, Sipos M, 1983, The development and validation of the Hungarian formof the STAI. In: Spielberger CD, DiazGuerro R, eds, Cross cultural anxiety, 2. Hemisphere Publishing Corporation, Washington-London, pp 51–61 Caldwell RA, Pearson JL, Chin RJ, 1987, Stress-moderating effects: social support in the context of gender and locus of control. Personal Soc Psychol Bull 13(1):5–17 Rychetnick L, McCaffery K, Morton R, Irwing L, 2012, Psychosocial aspects of post-treatment follow-up for stage I/II melanoma: a systematic review of the literature. Psychooncology 22(4): 721–736 Kasparian AN, Sansom-Daly U, McDonald RP, Meiser B, Butow PN, Mann GJ, 2012, The nature and structure of psychological distress in people at high risk for melanoma: a factor analytic study. Psychooncology 21:845–856 Meyer F, Ehrlich M, Peteet J, 2009, Psycho-oncology: a review for the general psychiatrist. FOCUS 7:317–331 Satin JR, Linden W, Phillips MJ, 2009, Depression as a predictor of disease progression and mortality in cancer patients. Cancer 115: 5349–5361 Fawzy IF, Fawzy NW, Hyun CS et al, 1993, Malignant melanoma: effects of an early structured psychiatric intervention, coping, and affective state on recurrence and survival 6 years later. Arch Gen Psychiatry 50:681–689 Eggermont AM, Suciu S, SantinamiM, EORTC Melanoma Group et al, 2008, Adjuvant therapy with pegylated interferon alfa-2b versus observation alone in resected stage III melanoma: final results of EORTC 18991, a randomized phase III trial. Lancet 372(9633):117–126 Trask P, Paterson A, Esper P et al, 2004, Longitudinal course of depression, fatigue, and quality of life in patients with high-risk melanoma receiving adjuvant interferon. Psychooncology 13: 526–536 Eggermont AMM, Suciu S, MacKie R, for the EORTC Melanoma Group et al, 2005, Post-surgery adjuvant therapy with intermediate doses of interferon alfa 2b versus observation in patients with stage IIb/III melanoma, EORTC 18952): randomised controlled trial. Lancet 366:1189–1196 Grob JJ, Dreno B, de la Salmoniere P et al, 1998, Randomised trial of interferon alpha-2a as adjuvant therapy in resected primary melanoma thicker than 1,5 mm without clinically detectable node metastases. Lancet 351:1905–1910 Muller N, Schwarz MJ, 2007, The immune-mediated alteration of serotonin and glutamate: towards an integrated view of depression. Mol Psychiatry 12:988–1000 Licinio J, Kling MA, Hauser P, 1998, Cytokines and brain function: relevance to interferon-alpha induced mood and cognitive changes. Semin Oncol 25(suppl 1):S30–S38 Reiche EMV, Nunes SOV, Morimoto HK, 2004, Stress, depression, the immune system and cancer. Lancet 5:617–625 Musselmann DL, Lawson DH, Gumnick JF et al, 2001, Paroxetine for the prevention of depression induced by highdose interferon alpha. N Engl J Med 344:961–966 Almeida AG, Guindalini C, Batista-Neves S et al, 2010, Can antidepressants prevent interferon-alpha-induced depression? A review of the literature. Gen Hosp Psychiatry 32:401–405 Raison CL, Capuron L, Miller AH, 2006, Cytokines sing the blues: inflammation and the pathogenesis of major depression. Trends Immunol 27:24–31 Heinze S, Egberts F, Rotzer S et al, 2010, Depressive mood changes and psychiatric symptoms. J Immunother 33:106–114 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 939 EP 944 DI 10.1007/s12253-014-9777-7 PG 6 ER PT J AU Powrozek, T Krawczyk, P Jarosz, B Mlak, R Wojas-Krawczyk, K Sawicki, M Stencel, D Trojanowski, T Milanowski, J AF Powrozek, Tomasz Krawczyk, Pawel Jarosz, Bozena Mlak, Radoslaw Wojas-Krawczyk, Kamila Sawicki, Marek Stencel, Dariusz Trojanowski, Tomasz Milanowski, Janusz TI The Application of Real-Time PCR Technique to Detect Rare Cell Clones with Primary T790M Substitution of EGFR Gene in Metastases of Non-small Cell Lung Cancer to Central Nervous System in Chemotherapy Naive Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; EGFR gene; T790M mutation; Metastases to central nervous system; EGFR tyrosine kinase inhibitor ID Non-small cell lung cancer; EGFR gene; T790M mutation; Metastases to central nervous system; EGFR tyrosine kinase inhibitor AB The time-limited efficacy of reversible EGFR-TKIs in patients with advanced non-small cell lung cancer (NSCLC) with EGFR gene activating mutations is associated with development of treatment resistance after some period of therapy. This resistance predominantly results from secondary mutations located in EGFR gene, especially T790M substitution. There is limited information available concerning the prevalence of primary T790M mutations in patients with metastatic NSCLC tumors before treatment with EGFR-TKIs. The aim of work was to assess the prevalence of de novo T790M mutations in EGFR gene in tissue samples from NSCLC metastatases in central nervous system (CNS) in both chemotherapy and EGFR-TKI naive NSCLC patients. We analyzed DNA samples isolated from paraffin-embedded tissue from CNS metastases for T790M mutations using realtime PCR and TaqMan probe against the T790M mutant sequence. The tissue samples were taken during palliative neurosurgery in 143 NSCLC patients. Amplification of the T790M-specific sequence was detected in 25 patients (17.5 %). The quantity of mutated DNA was less than 1 % in all samples with amplification, and in vast majority (20 patients, 14 % of all samples) it was even less that 0.1 %. In 5 patients (3.5 %) quantity of mutated DNA ranged from 0.1 to 1 % and true positive results of T790M mutation presence in these patients were most possible. Amplification of this sequence was not concurrent with common EGFR mutations and was not associated with sex, smoking status and pathological type of cancer. There is a possibility to detect the primary T790M mutation in brain metastases of NSCLC in EGFR-TKIs naive patients. C1 [Powrozek, Tomasz] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Krawczyk, Pawel] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Jarosz, Bozena] Medical University of Lublin, Neurosurgery and Pediatric Neurosurgery DepartmentLublin, Poland. [Mlak, Radoslaw] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Wojas-Krawczyk, Kamila] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Sawicki, Marek] Medical University of Lublin, Thoracic Surgery DepartmentLublin, Poland. [Stencel, Dariusz] Boehringer Ingelheim PolandWarsaw, Poland. [Trojanowski, Tomasz] Medical University of Lublin, Neurosurgery and Pediatric Neurosurgery DepartmentLublin, Poland. [Milanowski, Janusz] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. RP Krawczyk, P (reprint author), Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, 20-954 Lublin, Poland. EM krapa@poczta.onet.pl CR Zhou W, Ercan B, Janne PA, Gray NS, 2011, Discovery of selective irreversible inhibitors for EGFR-T790M. Bioorg Med Chem Lett 21(2):638–643 Ma C, Wei S, Song Y, 2011, T790M and acquired resistance of EGFRTKI: a literature review of clinical reports. J Thorac Dis 3:10– 18 Krawczyk P, Mlak R, Powrozek T, Nicos M, Kowalski DM, Wojas- Krawczyk K et al, 2012, Mechanisms of resistance to reversible inhibitors of EGFR tyrosine kinase in non-small cell lung cancer. Wspolczesna Onkol 16(5):401–406 Maheswaran S, Sequist LV, Nagrath S, Ulkus L, Brannigan B, Collura CVet al, 2008, Detection of mutations in EGFR in circulating lung-cancer cells. N Engl J Med 359:366–377 Su K-Y, Chen H-Y, Li K-C, Kuo M-L, Yang JC-H, Chan W-K et al, 2012, Pretreatment epidermal growth factor receptor, EGFR, T790M mutation predicts shorter EGFR tyrosine kinase inhibitor response duration in patients with non–small-cell lung cancer. J Clin Oncol 30:433–440 Oh JE, An CH, Yoo NJ, Lee SH, 2011, Detection of low-level EGFR T790M mutation in lung cancer tissues. APMIS 119(7):403–411 Asano H, Toyooka S, Tokumo M, Ichimura K, Aoe K, Ito S et al, 2006, Detection of EGFR gene mutation in lung cancer by mutantenriched polymerase chain reaction assay. Clin Cancer Res 12:43–48 Matsumoto S, Takahashi K, Iwakawa R, Matsuno Y, Nakanishi Y, Kohno T et al, 2006, Frequent EGFR mutations in brain metastases of lung adenocarcinoma. Int J Cancer 119(6):1491–1494 Han HS, Eom DW, Kim JH, Kim KH, Shin HM, An JYet al, 2011, EGFR mutation status in primary lung adenocarcinomas and corresponding metastatic lesions: discordance in pleural metastases. Clin Lung Cancer 12(6):380–386 Inukai M, Toyooka S, Ito S, Asano H, Ichihara S, Soh J et al, 2006, Presence of epidermal growth factor receptor gene T790M mutation as aminor clone in non–small cell lung cancer. Cancer Res 66:7854–7858 Miller VA, Hirsh V, Cadranel J, Chen YM, Park K, Kim SW et al, 2012, Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy, LUX-Lung 1): a phase 2b/3 randomised trial. Lancet Oncol 13(5):528–538 Hirsh V, Cadranel J, Cong XJ, Fairclough D, Finnern HW, Lorence RM et al, 2013, Symptom and quality of life benefit of afatinib in advanced non-small-cell lung cancer patients previously treated with erlotinib or gefitinib: results of a randomized phase IIb/III trial, LUXLung 1). J Thorac Oncol 8(2):229–237 Janjigian YY, Groen HJ, Horn L, Smit EF, Fu Y,Wang F et al., 2011, Activity and tolerability of afatinib, BIBW 2992, and cetuximab in NSCLC patients with acquired resistance to erlotinib or gefitinib. J Clin Oncol 29(suppl): abstr 7525 Fujita Y, Suda K, Kimura H, Matsumoto K, Arao T, Nagai T et al, 2012, Highly sensitive detection of EGFR T790M mutation using colony hybridization predicts favorable prognosis of patients with lung cancer harboring activating EGFR mutation. J Thorac Oncol 7(11):1640–1644 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 945 EP 951 DI 10.1007/s12253-014-9778-6 PG 7 ER PT J AU Ding, W Wang, G Shao, K Wang, F Huang, H Ju, Sh Cong, H Wang, H AF Ding, Weifeng Wang, Guihua Shao, Keke Wang, Feng Huang, Hua Ju, Shaoqing Cong, Hui Wang, Huimin TI Amelioration of Colorectal Cancer Using Negative Lipidoid Nanoparticles to Encapsulate siRNA Against APRIL by Enema Delivery Mode SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; A proliferation-inducing ligand; Negative lipidoid nanoparticles; siRNA; Enema; Small-molecule drugs ID Colorectal cancer; A proliferation-inducing ligand; Negative lipidoid nanoparticles; siRNA; Enema; Small-molecule drugs AB A proliferation-inducing ligand (APRIL) is a key cell proliferation-regulatory molecule and have been investigated well enough in immunity regulation and a few of immune diseases. APRIL can stimulate tumor cell growth and is up-expressed in cancer tissues, especially in CRC (colorectal cancer). However, whether inhibition of APRIL can regulate tumor-relative genes expression in vivo and subsequently ameliorate the pathological progress of CRC remains obscure. To address this question, we developed a novel negative lipidoid nanoparticles (NLNs) encapsulating small interference RNA (siRNA) for selectively silencing APRIL in the parenchyma of CRC focus in vivo, which uptake proceeded through a lipid raft endocytotic pathway. Local enema delivery of APRIL-NLNs silenced APRIL in CRC cells and animal models, and then ameliorated experimentally the progress of CRC by suppressing CRC cell proliferation, metastasis, and apoptosis-related cytokine expression and did not affect the function of liver and kidneys and not trigger the immune response of CRC models. This study reveals APRIL to be a potential anti-CRC target by in vivo experiments, and suggests that the application of similar modes of siRNA delivery may be feasible in other therapeutic settings. C1 [Ding, Weifeng] Affiliated Hospital of Nantong University, Department of Laboratory MedicineNantong, Jiangsu Province, China. [Wang, Guihua] Affiliated Hospital of Nantong University, Department of Laboratory MedicineNantong, Jiangsu Province, China. [Shao, Keke] The First People’s Hospital of Yancheng City, Medical Laboratory DepartmentYancheng, Jiangsu Province, China. [Wang, Feng] Affiliated Hospital of Nantong University, Department of Laboratory MedicineNantong, Jiangsu Province, China. [Huang, Hua] Affiliated Hospital of Nantong University, Pathology Medicine CenterNantong, Jiangsu Province, China. [Ju, Shaoqing] Affiliated Hospital of Nantong University, Department of Laboratory MedicineNantong, Jiangsu Province, China. [Cong, Hui] Affiliated Hospital of Nantong University, Department of Laboratory MedicineNantong, Jiangsu Province, China. [Wang, Huimin] Affiliated Hospital of Nantong University, Department of Laboratory MedicineNantong, Jiangsu Province, China. RP Cong, H (reprint author), Affiliated Hospital of Nantong University, Department of Laboratory Medicine, Nantong, China. EM dwfnt@hotmail.com CR Siegel R,Ward E, Brawley O, Jemal A, 2011, Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 61:212–236 Ding W, Wang J, Wang F, Wang G, Wu Q, Ju S, Cong H, Wang H, 2013, Serum sAPRIL: a potential tumor-associated biomarker to colorectal cancer. Clin Biochem 46(15):1590–1594 Sun B, Wang H, Wang X, Huang H, Ding W, Jing R, Shi G, Zhu L, 2009, A proliferation- inducing ligand: a new biomarker for Non- Small Cell Lung Cancer. Exp Lung Res 35:486–500 Wang F, Chen L, Ding W, Wang G, Wu Y, Wang J, Luo L, Cong H, Wang Y, Ju S, Shao J, Wang H, 2011, Serum APRIL, a potential tumor marker in pancreatic cancer. Clin Chem Lab Med 49:1715– 1719 Hahne M, Kataoka T, Schroter M, Hofmann K, Irmler M, Bodmer JL, Schneider P, Bornand T, Holler N, French LE, Sordat B, Rimoldi D, Tschopp J, 1998, APRIL, a new ligand of the tumor necrosis factor family, stimulates tumor cell growth. J Exp Med 188:1185– 1190 Petty RD, Samuel LM, Murray GI, MacDonald G, O’Kelly T, Loudon M, Binnie N, Aly E, McKinlay A, Wang W, Gilbert F, Semple S, Collie-Duguid ES, 2009, APRIL is a novel clinical chemo-resistance biomarker in colorectal adenocarcinoma identified by gene expression profiling. BMC Cancer 11:434 Wang G, Wang F, Ding W, Wang J, Jing R, Li H, Wang X, Wang Y, Ju S,Wang H, 2013, APRIL induces tumorigenesis andmetastasis of colorectal cancer cells via activation of the PI3K/Akt pathway. PLoS One 8:e55298 Lascano V, Zabalegui LF, Cameron K, Guadagnoli M, Jansen M, BurggraafM, VerslootM, Rodermond H, van der Loos C, Carvalho- Pinto CE, Kalthoff H, Medema JP, HahneM(2012, The TNF family member APRIL promotes colorectal tumorigenesis. Cell Death Differ 19:1826–1835 Dillon SR, Gross JA, Ansell SM, Novak AJ, 2006, An APRIL to remember: novel TNF ligands as therapeutic targets. Nat Rev Drug Discov 5:235–246 Ding W, Wang J, Sun B, Ju S, Yuan H, Wang X, Wang Y, Wang H, 2009, APRIL knockdown suppresses migration and invasion of human colon carcinoma cells. Clin Biochem 42:1694–1698 Wang F, Chen L, Mao ZB, Shao JG, Tan C, Huang WD, 2008, Lentivirus-mediated short hairpin RNA targeting APRIL gene suppresses growth of pancreatic cancer cells in vitro and in vivo. Oncol Rep 20:135–139 Wang F, Ding W, Wang J, Jing R, Wang X, Cong H, Wang Y, Ju S, Wang H, 2011, Identification of microRNA-target interaction in APRIL-knockdown colorectal cancer cells. Cancer Gene Ther 18: 500–509 Akinc A, Zumbuehl A, Goldberg M, Leshchiner ES, Busini V, Hossain N, Bacallado SA, Nguyen DN, Fuller J, Alvarez R, Borodovsky A, Borland T, Constien R, de Fougerolles A, Dorkin JR, Narayanannair Jayaprakash K, Jayaraman M, John M, Koteliansky V, Manoharan M, Nechev L, Qin J, Racie T, Raitcheva D, Rajeev KG, Sah DW, Soutschek J, Toudjarska I, Vornlocher HP, Zimmermann TS, Langer R, Anderson DG, 2008, A combinatorial library of lipid-like materials for delivery of RNAi therapeutics. Nat Biotechnol 26:561–569 Naahidi S, Jafari M, Edalat F, Raymond K, Khademhosseini A, Chen P, 2013, Biocompatibility of engineered nanoparticles for drug delivery. J Control Release 166:182–194 Rao NM, 2010, Cationic lipid-mediated nucleic acid delivery: beyond being cationic. Chem Phys Lipids 163:245–252 Williams DF, 2008, On the mechanisms of biocompatibility. Biomaterials 29:2941–2953 DingW,Wang F, Zhang J, Guo Y, Ju S,WangH, 2013, A novel local anti-colorectal cancer drugs delivery system: negative lipidoid nanoparticles with passive target via a size-dependent pattern. Nanotechnology 24:375101 Allred DC, Harvey JM, Berardo M, Clark GM(1998, Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11:155–168 Zhou X, Wang JL, Lu J, Song Y, Kwak KS, Jiao Q, Rosenfeld R, Chen Q, Boone T, Simonet WS, Lacey DL, Goldberg AL, Han HQ, 2010, Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival. Cell 142:531–543 de Fougerolles A, Vornlocher HP,Maraganore J, Lieberman J, 2007, Interfering with disease: a progress report on siRNA-based therapeutics. Nat Rev Drug Discov 6:443–453 McNamara JO 2nd, Andrechek ER,Wang Y, Viles KD, Rempel RE, Gilboa E, Sullenger BA, Giangrande PH, 2006, Cell type-specific delivery of siRNAs with aptamer-siRNA chimeras. Nat Biotechnol 24:1005–1015 Palliser D, Chowdhury D, Wang QY, Lee SJ, Bronson RT, Knipe DM, Lieberman J, 2006, An siRNA-based microbicide protects mice from lethal herpes simplex virus 2 infection. Nature 439:89–94 Zimmermann TS, Lee AC, Akinc A, Bramlage B, Bumcrot D, Fedoruk MN, Harborth J, Heyes JA, Jeffs LB, John M, Judge AD, Lam K, McClintock K, Nechev LV, Palmer LR, Racie T, Rohl I, Seiffert S, Shanmugam S, Sood V, Soutschek J, Toudjarska I,Wheat AJ, Yaworski E, Zedalis W, Koteliansky V, Manoharan M, Vornlocher HP,MacLachlan I, 2006, RNAi-mediated gene silencing in non-human primates. Nature 441:111–114 Heidel JD, Yu Z, Liu JY, Rele SM, Liang Y, Zeidan RK, Kornbrust DJ, Davis ME, 2007, Administration in non-human primates of escalating intravenous doses of targeted nanoparticles containing ribonucleotide reductase subunit M2 siRNA. Proc Natl Acad Sci U S A 104:5715–5721 Howard KA, Rahbek UL, Liu X, Damgaard CK, Glud SZ, Andersen MO, Hovgaard MB, Schmitz A, Nyengaard JR, Besenbacher F, Kjems J, 2006, RNA interference in vitro and in vivo using a novel chitosan/siRNA nanoparticle system. Mol Ther 14:476–484 Kim WJ, Christensen LV, Jo S, Yockman JW, Jeong JH, Kim YH, Kim SW, 2006, Cholesteryl oligoarginine delivering vascular endothelial growth factor siRNA effectively inhibits tumor growth in colon adenocarcinoma. Mol Ther 14:343–350 Peer D, Zhu P, Carman C, Lieberman J, Shimaoka M, 2007, Selective gene silencing in activated leukocytes by targeting siRNAs to the integrin lymphocyte function-associated antigen-1. Proc Natl Acad Sci U S A 104:4095–4100 Peer D, Park EJ, Morishita Y, Carman CV, Shimaoka M, 2008, Systemic leukocyte-directed siRNA delivery revealing Cyclin D1 as an anti-inflammatory target. Science 319:627–630 Jackson AL, Bartz SR, Schelter J, Kobayashi SV, Burchard J, Mao M, Li B, Cavet G, Linsley PS, 2003, Expression profiling reveals offtarget gene regulation by RNAi. Nat Biotechnol 21:635–637 Fedorov Y, Anderson EM, Birmingham A, Reynolds A, Karpilow J, Robinson K, Leake D, Marshall WS, Khvorova A, 2006, Off-target effects by siRNA can induce toxic phenotype. RNA 12:1188–1196 Judge AD, Sood V, Shaw JR, Fang D, McClintock K, MacLachlan I, 2005, Sequence-dependent stimulation of the mammalian innate immune response by synthetic siRNA. Nat Biotechnol 23:457–462 Takahashi N, Tamagawa K, Shimizu K, Fukui T, Maitani Y, 2003, Effects on M5076-hepatic metastasis of retinoic acid and N-(4- hydroxyphenyl, retinamide, fenretinide entrapped in SG-liposomes. Biol Pharm Bull 26:1060–1063 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 953 EP 964 DI 10.1007/s12253-014-9779-5 PG 12 ER PT J AU Choi, JY Kim, SM An, HCh Yoo, JN Lee, HS AF Choi, Jin Youn Kim, Sung Min An, Hyeok Chang Yoo, Jin Nam Lee, Hyung Sug TI Regional Bias of Intratumoral Genetic Heterogeneity of Nucleotide Repeats in Colon Cancers with Microsatellite Instability SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bethesda panel; Colorectal cancer; Dinucleotide repeats; Intratumoral heterogeneity; Microsatellite instability; Mononucleotide repeats; Promega panel ID Bethesda panel; Colorectal cancer; Dinucleotide repeats; Intratumoral heterogeneity; Microsatellite instability; Mononucleotide repeats; Promega panel AB Intratumoral heterogeneity (ITH) may produce regional biases in genotype and phenotype evaluation in a single tumor and may impede proper cancer diagnosis. To evaluate the extent of ITH in colorectal cancer (CRC) with microsatellite instability (MSI), we obtained 4–7 biopsies from39CRCs followed by MSI analysis either using the Bethesda MSI evaluation system or Promega system with 5 mononucleotide markers. We found decreased prevalence of MSI (+) by the Promega system compared to the Bethesda system. The overall discordance between the two systems was 54%. In contrast to the previous studies that had shown discordance only in low MSI (MSI-L), our results showed the discordance not only in MSI-L, but also in high MSI (MSI-H) cases. Among the MSI (+) CRCs, ITH of MSI status was identified in 41.7%of CRC by the Bethesda systemand 22.2%by the Promega system. In terms of MSI markers, the ITH originated from dinucleotide markers in most cases (69 %), but it originated from mononucleotide markers (31 %) as well. Pooling of DNA from a regional biopsy with MSI (+) with additional biopsies from stable MSI (MSS) showed that this approach was beneficial to increase the sensitivity of MSI detection. Our results indicate that ITH of MSI phenotype by the Bethesda system is more overestimated than previously identified. However, because there was considerable ITH of MSI subtypes and markers even by the Promega system, our data suggest that analysis of MSI status in multiple regional biopsies is needed for a better evaluation of MSI status in CRC. C1 [Choi, Jin Youn] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [An, Hyeok Chang] The Catholic University of Korea, College of Medicine, Department of General Surgery, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Wainscoat JS, Fey MF, 1990, Assessment of clonality in human tumors: a review. Cancer Res 50:1355–60 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–34 Duffy M, Crown J, 2013, Companion biomarkers: paving the pathway to personalized treatment for cancer. Clin Chem 59:1447–56 Chen ZY, ZhongWZ, Zhang XC, Su J, Yang XN, Chen ZH, Yang JJ, Zhou Q, Yan HH, An SJ, Chen HJ, Jiang BY, Mok TS, Wu YL, 2012, EGFR mutation heterogeneity and the mixed response to EGFR tyrosine kinase inhibitors of lung adenocarcinomas. Oncologist 17:978–85 Loukola A, Eklin K, Laiho P, Salovaara R, Kristo P, Jarvinen H, Mecklin JP, Launonen V, Aaltonen LA, 2001, Microsatellite marker analysis in screening for hereditary nonpolyposis colorectal cancer, HNPCC). Cancer Res 61:4545–9 Goel A, Arnold CN, Niedzwiecki D, Chang DK, Ricciardiello L, Carethers JM, Dowell JM, Wasserman L, Compton C, Mayer RJ, Bertagnolli MM, Boland CR, 2003, Characterization of sporadic colon cancer by patterns of genomic instability. Cancer Res 63: 1608–14 Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutterbaugh J, Fan RS, Zborowska E, Kinzler KW, Vogelstein B, Brattain M, Wilson JKV, 1995, Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science 268:1336–8 Calin GA, Gaf R, Tibiletti MG, Herlea V, Becheanu G, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression in microsatellite instability high, MSI-H, colon cancers correlates with clinico-pathological parameters: a study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89:230–5 Duval A, Gayet J, Zhou XP, Iacopetta B, Thomas G, Hamelin R, 1999, Frequent frameshiftmutations of the TCF-4 gene in colorectal cancers with microsatellite instability. Cancer Res 59:4213–5 Barnetson R, Jass J, Tse R, Eckstein R, Robinson B, Schnitzler M, 2000, Mutations associated with microsatellite unstable colorectal carcinomas exhibit widespread intratumoral heterogeneity. Gene Chromosome Cancer 29:130–6 Duval A, Hamelin R, 2002, Mutations at coding repeat sequences in mismatch repair-deficient human cancers: toward a new concept of target genes for instability. Cancer Res 62:2447–54 Chung YJ, Kim KM, Choi JR, Choi SW, Rhyu MG, 1998, Relationship between intratumor histological heterogeneity and genetic abnormalities in gastric carcinoma with microsatellite instability. Int J Cancer 82:782–8 Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, Meltzer SJ, Rodriguez-Bigas MA, Fodde R, Ranzani GN, Srivastava S, 1998, A National Cancer Institute Workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58:5248–57 Patil DT, Bronner MP, Portier BP, Fraser CR, Plesec TP, LiuX, 2012, A five-marker panel in a multiplex PCR accurately detects microsatellite instability-high colorectal tumors without control DNA. Diagn Mol Pathol 21:127–33 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. JMol Diagn 8:305–11 Tomlinson I, Halford S, Aaltonen L, Hawkins N, Ward R, 2002, Does MSI-low exist? J Pathol 197:6–13 Xicola RM, Llor X, Pons E, Castells A, Alenda C, Pinol V, Andreu M, Castellvi-Bel S, Paya A, Jover R, Bessa X, Giros A, Duque JM, Nicolas-Perez D, Garcia AM, Rigau J, Gassull MA; Gastrointestinal Oncology Group of the Spanish Gastroenterological Association, 2007, Performance of different microsatellite marker panels for detection of mismatch repair-deficient colorectal tumors. J Natl Cancer Inst 99:244–52 Buhard O, Suraweera N, Lectard A, Duval A, Hamelin R, 2004, Quasimonomorphic mononucleotide repeats for high-level microsatellite instability analysis. Dis Markers 20:251–7 Suraweera N, Duval A, Reperant M, Vaury C, Furlan D, Leroy K, Seruca R, Iacopetta B, Hamelin R, 2002, Evaluation of tumor microsatellite instability using five quasimonomorphic mononucleotide repeats and pentaplex PCR. Gastroenterology 123:1804–11 Danjoux M, Guimbaud R, Al Saati T,Meggetto F, Carrere N, Portier G, Delsol G, Selves J, 2006, Contribution of microdissection for the detection ofmicrosatellite instability in colorectal cancer. Hum Pathol 37:361–8 Popat S, Houlston RS, 2005, A systematic review and meta-analysis of the relationship between chromosome 18q genotype, DCC status and colorectal cancer prognosis. Eur J Cancer 41:2060–70 Carethers JM, Smith EJ, Behling CA, Nguyen L, Tajima A, Doctolero RT, Cabrera BL, Goel A, Arnold CA, Miyai K, Boland CR, 2004, Use of 5-fluorouracil and survival in patients with microsatellite-unstable colorectal cancer. Gastroenterology 126: 394–401 Fallik D, Borrini F, Boige V, Viguier J, Jacob S, Miquel C, Sabourin JC, Ducreux M, Praz F, 2003)Microsatellite instability is a predictive factor of the tumor response to irinotecan in patients with advanced colorectal cancer. Cancer Res 63:5738–44 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 965 EP 971 DI 10.1007/s12253-014-9781-y PG 7 ER PT J AU Sari, E Nagy, GyZs Baghy, K Rajnai, H Bodor, Cs Csomor, J Barna, G Rudas, G Kovalszky, I Demeter, J AF Sari, Eszter Nagy, Gyorgy Zsolt Baghy, Kornelia Rajnai, Hajnalka Bodor, Csaba Csomor, Judit Barna, Gabor Rudas, Gabor Kovalszky, Ilona Demeter, Judit TI Treatment of Refractory Hairy Cell Leukemia with a BRAF-inhibitor: Lessons to be Learnt SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hairy cell leukemia; Refractory; BRAF V600E; Vemurafenib ID Hairy cell leukemia; Refractory; BRAF V600E; Vemurafenib AB Hairy cell leukemia is a rare chronic lymphoproliferative disorder with indolent but progressive clinical course. Patients require treatment when they have significant cytopenia or recurrent infections. The gold standard treatment are purine nucleoside analogues (cladribine and pentostatine), with these agents the rate of complete remission can approach even 95 %. The differential diagnosis between classical hairy cell leukemia and other, rare splenic lymphomas that can mimic this disease might be really challenging. Splenic lymphoma with villous lymphocytes and other new, provisional WHO entities share some, but not all immunophenotypical features with hairy cell leukemia. The correct diagnosis is of an extreme importance as these entities require different treatment. Thus further investigation in the pathogenesis of hairy cell leukemia is required in order to solve this challenge. Discovery of the BRAF V600E mutation as a diseasedefining genetic event in hairy cell leukemia can be helpful in both differential diagnosis and treatment of this disease.We report the case of three hairy cell leukemia patients, whose diagnosis or treatment was based on this newly discovered somatic mutation, but the treatment results and side effects were individual. C1 [Sari, Eszter] Semmelweis University, 1st Department of Internal Medicine, Koranyi S. Str. 2/a, H-1083 Budapest, Hungary. [Nagy, Gyorgy Zsolt] Semmelweis University, 1st Department of Internal Medicine, Koranyi S. Str. 2/a, H-1083 Budapest, Hungary. [Baghy, Kornelia] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Str. 26, H-1085 Budapest, Hungary. [Rajnai, Hajnalka] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Str. 26, H-1085 Budapest, Hungary. [Bodor, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Str. 26, H-1085 Budapest, Hungary. [Csomor, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Str. 26, H-1085 Budapest, Hungary. [Barna, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Str. 26, H-1085 Budapest, Hungary. [Rudas, Gabor] Semmelweis Egyetem, MR Kutatokozpont, Balassa Str. 6, H-1083 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Str. 26, H-1085 Budapest, Hungary. [Demeter, Judit] Semmelweis University, 1st Department of Internal Medicine, Koranyi S. Str. 2/a, H-1083 Budapest, Hungary. RP Demeter, J (reprint author), Semmelweis University, 1st Department of Internal Medicine, H-1083 Budapest, Hungary. EM demjud@bel1.sote.hu CR GreverMA, 2010, How I Treat Hairy Cell Leukemia. Blood 115:21– 28 Krenacs L, Toth-Liptak J, Demeter J et al, 2013, Monoclonal Antibody HBME-1 Reacts with a Minor Subset of B cells with Villous Surface and can be Useful in the Diagnosis of Hairy Cell Leukemia and other Indolent Lymphoproliferations of Villous B Lymphocytes. Virchows Arch 463(6):787–794 Naik RR, Saven A, 2012, My Treatment Approach to Hairy Cell Leukemia. Mayo Clin Proc 87(1):67–76 Tiacci E, Trifonov V, Schiavoni G et al, 2011, BRAF Mutations in Hairy Cell Leukemia. N Engl J Med 364:2305–2315 Davies H, Bignell GR, Cox C et al, 2002, Mutations of the BRAF Gene in Human Cancer. Nature 417:949–954 Tiacci E, Schiavoni G, Forconi F et al, 2012, Simple Genetic Diagnosis of Hairy cell Leukemia by Sensitive Detection of the BRAF-V600E Mutation. Blood 119:192–195 Blombery PA, Wong SQ, Hewitt CA et al, 2012, Detection of BRAF Mutations in Patients with Hairy Cell Leukemia and Related Lymphoproliferative Disorders. Haematologica 97: 780–783 Boyd EM, Bench AJ, Veer MB V’t et al, 2011, High Resolution Melting Analysis for Detection of BRAF exon 15Mutations in Hairy Cell Leukaemia and Other LymphoidMalignancies. Br J Haem 155: 609–612 Arcaini L, Zibellini S, Boveri E et al, 2012, The BRAF V600E Mutation in Hairy Cell Leukemia and other Mature B-cell Neoplasms. Blood 119:188–191 Lennerz JK, Klaus BM, Marienfeld RB et al, 2012, Pyrosequencing of BRAF V600E in Routine Samples of Hairy Cell Leukaemia Identifies CD5+ variant Hairy Cell Leukaemia that Lacks V600E. Br J Haem 157:267–269 Schnittger S, Bacher U, Haferlach T et al, 2012, Development and Validation of a Real-Time Quantification Assay to Detect and Monitor BRAFV600E Mutations in Hairy Cell Leukemia. Blood 119:3151–3154 Xi L, Arons E, Navarro Wet al, 2012, Both variant and IGHV4-34- Expressing Hairy Cell Leukemia Lack the BRAF V600E Mutation. Blood 119:3330–3332 Langabeer SE, O’Brien D, Liptrot S et al, 2012, Correlation of the BRAF V600E Mutation in Hairy Cell Leukaemia with Morphology, Cytochemistry and Immunophenotype. Int J Lab Hem 34:417–421 Andrulis M, Penzel R, Weichert W et al, 2012, Application of a BRAF V600E Mutation-Specific Antibody for the Diagnosis of Hairy Cell Leukemia. Am J SurgPathol 36(12):1796–1800 Laurini JA, Aoun P, Iqbaal J et al, 2012, Investigation of the BRAF V600E Mutation by Pyrosequencing in Lymphoproliferative Disorders. Am J Clin Pathol 138:877–883 Dietrich S, Glimm H, Andrulis M et al, 2012, BRAF Inhibition in Refractory Hairy-Cell Leukemia. N Engl J Med 366:2038–2040 Follows GA, Sims H, Bloxham DM et al, 2013, Rapid Response of Biallelic BRAF V600E Mutated Hairy Cell Leukemia to Low Dose Vemurafenib. Br J Haem 161(1):150–153 Peyrade F, Re D, Ginet C et al, 2013, Low-Dose Vemurafenib Induces Complete Remission in a Case of Hairy-Cell Leukemia with a V600E Mutation. Haematologica 98(2):e20–e22 Munoz J, Schlette E, Kurzrock R, 2013, Rapid Response to Vemurafenib in a Heavily Pretreated Patient with Hairy Cell Leukemia and a BRAF Mutation. J Clin Oncol 31(20):e351–e352 Maurer H, Haas P, Wengenmayer T et al, 2013, Successful Vemurafenib Salvage Treatment in a Patient with Primary Refractory Hairy Cell Leukemia and Pulmonary Aspergillosis. Ann Hematol DOI., DOI 10.1007/s.00277–013–1987–7 Urnova EC, Al-Radi LS, Kuzmina LA et al, 2013, Successful use of Vemurafenib in a Patient with a Resistant Hairy Cell Leukemia. Ter Arkh 85(7):76–78 Samuel J, Macip S, Dyer MJS, 2014, Efficacy of Vemurafenib in Hairy-Cell Leukemia. N Engl J Med 370:286–288 Dietrich S, Hullein J, HundemerM et al, 2013, Continued Response off Treatment after BRAF Inhibition in Refractory Hairy Cell Leukemia. J Clin Oncol 31(19):e300–e303 Cantwell-Dorris ER, O’Leary JJ, Sheils OM, 2011, BRAF V600E: Implications for Carcinogenesis and Molecular Therapy. Mol Canc Ther 3:385–394 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 973 EP 980 DI 10.1007/s12253-014-9783-9 PG 8 ER PT J AU Ladanyi, A Sebestyen, T Mohos, A Liszkay, G Somlai, B Toth, E Timar, J AF Ladanyi, Andrea Sebestyen, Timea Mohos, Anita Liszkay, Gabriella Somlai, Beata Toth, Erika Timar, Jozsef TI Ectopic lymphoid structures in primary cutaneous melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ectopic lymphoid structures; Primary melanoma; B cells; T cells; Follicular dendritic cells; High endothelial venules ID Ectopic lymphoid structures; Primary melanoma; B cells; T cells; Follicular dendritic cells; High endothelial venules AB Ectopic lymphoid structures have been described in several tumor types including metastatic lesions, but not primary tumors, of patients with melanoma. Here we present evidence of B-cell follicles in primary cutaneous melanomas, being present in 39 of 147 cases (27 %). B-cell clusters were associated with T lymphocytes, most of which belonging to CD45RO+ memory T cells. A network of CD21+ follicular dendritic cells was demonstrated in 8 of 22 cases studied (36 %). MECA-79+ HEV-like venules were observed in the neighborhood of the follicles in the majority of cases, however, their presence was not confined to tumors hosting ectopic lymphoid structures. The appearance of B-cell aggregates did not show association with the outcome of the disease, although a trend for their higher prevalence was observed in thicker tumors. Our results show that neogenesis of lymphoid structures does occur in primary melanomas, albeit with lower frequency compared to that reported in metastases. C1 [Ladanyi, Andrea] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, 7-9. Rath Gyorgy u., H-1122 Budapest, Hungary. [Sebestyen, Timea] St John's Hospital, Department of Pathology, 1-3. Dios arok, H-1125 Budapest, Hungary. [Mohos, Anita] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 26. Ulloi ut, H-1085 Budapest, Hungary. [Liszkay, Gabriella] National Institute of Oncology, Department of Dermatology, 7-9. Rath Gyorgy u., H-1122 Budapest, Hungary. [Somlai, Beata] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, 41. Maria u, H-1085 Budapest, Hungary. [Toth, Erika] National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, 7-9. Rath Gyorgy u., H-1122 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, 93. Ulloi ut, H-1091 Budapest, Hungary. RP Ladanyi, A (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, H-1122 Budapest, Hungary. EM ladanyi@oncol.hu CR Bell D, Chomarat P, Broyles D, Netto G, Moumneh Harb G, Lebecque S, Valladeau J, Davoust J, Palucka KA, Banchereau J, 1999, In breast carcinoma tissue, immature dendritic cells reside within the tumor, whereas mature dendritic cells are located in peritumoral areas. J Exp Med 10:1417–1425 BombardieriM, Barone F, Humby F, Kelly S,McGurkM,Morgan P, Challacombe S, De Vita S, Valesini G, Spencer J, Pitzalis C, 2007, Activation-induced cytidine deaminase expression in follicular dendritic cell networks and interfollicular large B cells supports functionality of ectopic lymphoid neogenesis in autoimmune sialoadenitis andMALT lymphoma in Sjogren’s syndrome. J Immunol 179:4929– 4938 Cipponi A, Mercier M, Seremet T, Baurain JF, Theate I, van den Oord J, Stas M, Boon T, Coulie PG, van Baren N, 2012, Neogenesis of lymphoid structures and antibody responses occur in human melanoma metastases. Cancer Res 72:3997–4007 Coppola D, Nebozhyn M, Khalil F, Dai H, Yeatman T, Loboda A, Mule JJ, 2011, Unique ectopic lymph node-like structures present in human primary colorectal carcinoma are identified by immune gene array profiling. Am J Pathol 179:37–45 Coronella-Wood JA, Hersh EM, 2003, Naturally occurring B-cell responses to breast cancer. Cancer Immunol Immunother 52:715– 738 de Chaisemartin L, Goc J, Damotte D, Validire P, Magdeleinat P, Alifano M, Cremer I, Fridman WH, Sautes-Fridman C, Dieu- Nosjean MC, 2011, Characterization of chemokines and adhesion molecules associated with T cell presence in tertiary lymphoid structures in human lung cancer. Cancer Res 71:6391–6399 Dieu-Nosjean MC, Antoine M, Danel C, Heudes D, Wislez M, Poulot V, Rabbe N, Laurans L, Tartour E, de Chaisemartin L, Lebecque S, Fridman WH, Cadranel J, 2008, Long-term survival for patients with non–small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol 26:4410–4417 Iwamoto M, Shinohara H, Miyamoto A, Okuzawa M, Mabuchi H, Nohara T, GonG, ToyodaM, TanigawaN, 2003, Prognostic value of tumor-infiltrating dendritic cells expressing CD83 in human breast carcinomas. Int J Cancer 104:92–97 Jensen TO, Schmidt H, Moller HJ, Donskov F, HoyerM, Sjoegren P, Christensen IJ, Steiniche T, 2012, Intratumoral neutrophils and plasmacytoid dendritic cells indicate poor prognosis and are associated with pSTAT3 expression in AJCC stage I/II melanoma. Cancer 118:2476–2485 Kirk CJ, Hartigan-O’Connor D, Mule JJ, 2001, The dynamics of the T-cell antitumor response: Chemokine-secreting dendritic cells can prime tumor-reactive T cells extranodally. Cancer Res 61:8794–8802 Ladanyi A, Kiss J, Mohos A, Somlai B, Liszkay G, Gilde K, Fejos Z, Gaudi I, Dobos J, Timar J, 2011, Prognostic impact of B-cell density in cutaneous melanoma. Cancer Immunol Immunother 60:1729– 1738 Ladanyi A, Kiss J, Somlai B, Gilde K, Fejos Z, Mohos A, Gaudi I, Timar J, 2007, Density of DC-LAMP+ mature dendritic cells in combination with activated T lymphocytes infiltrating primary cutaneous melanoma is a strong independent prognostic factor. Cancer Immunol Immunother 56:1459–1469 Martinet L, Le Guellec S, Filleron T, Lamant L, Meyer N, Rochaix P, Garrido I, Girard JP, 2012, High endothelial venules, HEVs, in human melanoma lesions. Major gateways for tumor-infiltrating lymphocytes. OncoImmunology 1:829–839 Messina JL, Fenstermacher DA, Eschrich S, Qu X, Berglund AE, Lloyd MC, Schell MJ, Sondak VK, Weber JS, Mule JJ, 2012, 12- chemokine gene signature identifies lymph node-like structures in melanoma: Potential for patient selection for immunotherapy? Sci Rep 2:765 Miyagawa S, Soeda J, Takagi S,Miwa S, Ichikawa E,Noike T, 2004, Prognostic significance of mature dendritic cells and factors associated with their accumulation in metastatic liver tumors from colorectal cancer. Hum Pathol 35:1392–1396 Moldenhauer G, Popov SW, Wotschke B, Bruderlein S, Riedl P, Fissolo N, Schirmbeck R, Ritz O, Moller P, Leithauser F, 2006, AID expression identifies interfollicular large B cells as putative precursors of mature B-cell malignancies. Blood 107:2470–2473 Nzula S, Going JJ, Stott DI, 2003, Antigen-driven clonal proliferation, somatic hypermutation, and selection of B lymphocytes infiltrating human ductal breast carcinomas. Cancer Res 63:3275–3280 O’Brien PM, Tsirimonaki E, Coomber DW, Millan DW, Davis JA, Campo MS, 2001, Immunoglobulin genes expressed by Blymphocytes infiltrating cervical carcinoma show evidence of antigen-driven selection. Cancer Immunol Immunother 50:523–532 Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA, Frosina D, Gnjatic S, Ambrosone C, Kepner J, Odunsi T, Ritter G, Lele S, Chen Y-T, Ohtani H, Old LJ, Odunsi K, 2005, Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A 102:18538–18543 Suzuki A, Masuda A, Nagata H, Kameoka S, Kikawada Y, Yamakawa M, Kasajima T, 2002, Mature dendritic cells make clusters with T cells in the invasive margin of colorectal carcinoma. J Pathol 196:37–43 Thompson ED, Enriquez HL, Fu YX, Engelhard VH, 2010, Tumor masses support naive Tcell infiltration, activation, and differentiation into effectors. J Exp Med 207:1791–1804 Tsukayama S, Omura K, Yoshida K, Tanaka Y, Watanabe G, 2005, Prognostic value of CD83-positive mature dendritic cells and their relation to vascular endothelial growth factor in advanced human gastric cancer. Oncol Rep 14:369–375 van Baren N, Baurain JF, Coulie PG, 2013, Lymphoid neogenesis in melanoma. What does it tell us? OncoImmunology 2:e22505 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 981 EP 985 DI 10.1007/s12253-014-9784-8 PG 5 ER PT J AU Zhang, Y Liu, H Chen, X Bai, Q Liang, R Shi, B Liu, L Tian, D Liu, M AF Zhang, Yongqing Liu, Hongjuan Chen, Xiequn Bai, Qingxian Liang, Rong Shi, Bing Liu, Lihui Tian, DengMei Liu, Mingjuan TI Modified Bortezomib, Adriamycin and Dexamethasone (PAD) Regimen in Advanced Multiple Myeloma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Modified PAD regimen; Bortezomib; Advanced multiple myeloma; Efficacy and safety ID Modified PAD regimen; Bortezomib; Advanced multiple myeloma; Efficacy and safety AB The PAD regime, composed of bortezomib, adriamycin and dexamethasone, improves the outcomes of patients with advanced multiple myeloma (MM), but at the same time produces high frequency of serious toxic side effects. For the first time, we evaluated the efficacy and safety of a bortezomib-dose-reduced PAD regime in the treatment of relapsed/refractory MM in this clinical study. Forty-five patients were treated with two to six 21-day cycles of PAD, comprising bortezomib at 1.3 mg/m2 (P1AD, n=21) or 1.0 mg/m2 (P2AD, n=24) (days 1, 4, 8, 11), adriamycin at 9 mg/m2 (days 1–4) and dexamethasone at 40 mg/day (days 1–4). Overall, 36 patients (80 %) showed at least partial remission (PR), in which 9 cases (20 %) showed complete remission (CR) and 10 cases (22 %) showed very good partial remission (VGPR). The efficacy of PAD regimen in advanced MM patients was not related to the traditional prognostic factors. There was no significant difference between P1AD and P2AD in the rates of PR, CR or VGPR, 1.5-year progression-free survival (PFS), and overall survival (OS) (81 % vs. 79%, 48 % vs. 38%, 64% vs. 59%, and 85% vs. 73 %, respectively). However, the grade 3–4 toxic effects, including thrombocytopenia (13 % vs. 38 %), peripheral neuropathy (8 % vs. 33%) and 3–4 grade gastrointestinal reaction (13%vs. 43%), were markedly inhibited after P2AD compared to P1AD (P<0.05). The bortezomib-dose-reduced PAD regime reduced the incidence of adverse reactions without affecting the treatment efficacy in patients with advanced MM. C1 [Zhang, Yongqing] the 309th Hospital of Chinese People’s Liberation Army, Department of Hematology, 17 Heishanhu Road, 100091 Beijing, China. [Liu, Hongjuan] the 309th Hospital of Chinese People’s Liberation Army, Department of Geriatrics, 100091 Beijing, China. [Chen, Xiequn] the Fourth Military Medical University, Xijing Hospital, Department of Hematology, 710032 Xi’an, China. [Bai, Qingxian] the Fourth Military Medical University, Xijing Hospital, Department of Hematology, 710032 Xi’an, China. [Liang, Rong] the Fourth Military Medical University, Xijing Hospital, Department of Hematology, 710032 Xi’an, China. [Shi, Bing] the 309th Hospital of Chinese People’s Liberation Army, Department of Hematology, 17 Heishanhu Road, 100091 Beijing, China. [Liu, Lihui] the 309th Hospital of Chinese People’s Liberation Army, Department of Hematology, 17 Heishanhu Road, 100091 Beijing, China. [Tian, DengMei] the 309th Hospital of Chinese People’s Liberation Army, Department of Hematology, 17 Heishanhu Road, 100091 Beijing, China. [Liu, Mingjuan] the 309th Hospital of Chinese People’s Liberation Army, Department of Hematology, 17 Heishanhu Road, 100091 Beijing, China. RP Zhang, Y (reprint author), the 309th Hospital of Chinese People’s Liberation Army, Department of Hematology, 100091 Beijing, China. EM zhangyongqing0725@163.com CR Rajkumar SV, Blood E, Vesole D, Fonseca R, Greipp PR, 2006, Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group. J Clin Oncol 24(3):431–436 Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D, Rajkumar SV, Srkalovic G, Alsina M, Alexanian R, Siegel D, Orlowski RZ, Kuter D, Limentani SA, Lee S, Hideshima T, Esseltine DL, Kauffman M, Adams J, Schenkein DP, Anderson KC, 2003, A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med 348(26):2609–2617 Mohty B, El-Cheikh J, Yakoub-Agha I, Avet-Loiseau H, Moreau P, Mohty M, 2012, Treatment strategies in relapsed and refractory multiple myeloma: a focus on drug sequencing and ‘retreatment’ approaches in the era of novel agents. Leukemia 26(1):73–85 Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, Facon T, Harousseau JL, Ben-Yehuda D, Lonial S, Goldschmidt H, Reece D, San-Miguel JF, Blade J, Boccadoro M, Cavenagh J, Dalton WS, Boral AL, Esseltine DL, Porter JB, Schenkein D, Anderson KC, 2005, Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med 352(24):2487–2498 Chanan-Khan A, Miller KC, Musial L, Padmanabhan S, Yu J, Ailawadhi S, Sher T, Mohr A, Bernstein ZP, Barcos M, Patel M, Iancu D, Lee K, Czuczman MS, 2009, Bortezomib in combination with pegylated liposomal doxorubicin and thalidomide is an effective steroid independent salvage regimen for patients with relapsed or refractory multiple myeloma: results of a phase II clinical trial. Leuk Lymphoma 50(7):1096–1101 Popat R, Oakervee H, Williams C, Cook M, Craddock C, Basu S, Singer C, Harding S, Foot N, Hallam S, Odeh L, Joel S, Cavenagh J, 2009, Bortezomib, low-dose intravenous melphalan, and dexamethasone for patients with relapsed multiple myeloma. Br J Haematol 144(6):887–894 Benevolo G, Larocca A, GentileM, Pregno P, Gay F, Botto B, Frairia C, Evangelista A,Morabito F, BoccadoroM(2011, The efficacy and safety of bortezomib and dexamethasone as a maintenance therapy in patients with advanced multiple myeloma who are responsive to salvage bortezomib‐containing regimens. Cancer 117(9):1884–1890 Waterman GN, Yellin O, Swift RA,Mapes R, Eades B, Ackerman E, Berenson JR, 2011, A modified regimen of pegylated liposomal doxorubicin, bortezomib, and dexamethasone is effective and well tolerated in the treatment of relapsed or refractory multiple myeloma. Ann Hematol 90(2):193–200 Romano A, Chiarenza A, Consoli U, Conticello C, Forte S, Uccello G, Vetro C, Cavalli M, Coppolino F, Palumbo GA, Di Raimondo F, 2013, Intravenous injection of bortezomib, melphalan and dexamethasone in refractory and relapsed multiple myeloma. Ann Oncol 24(4):1038–1044 Pantani L, Zamagni E, Zannetti BA, Pezzi A, Tacchetti P, Brioli A, Mancuso K, Perrone G, Rocchi S, Tosi P, CavoM(2013, Bortezomib and dexamethasone as salvage therapy in patients with relapsed/ refractory multiple myeloma: analysis of long-term clinical outcomes. Annals of Hematology: Epub ahead of print. http://www. bioportfolio.com/resources/pmarticle/464488/Bortezomib-anddexamethasone- as-salvage-therapy-in-patients-with-relapsedrefractory- multiple.html Richardson P, Jagannath S, Colson K, 2006, Optimizing the efficacy and safety of bortezomib in relapsed multiple myeloma. Clin Adv Hematol Oncol 4(5):1 Palumbo A, Gay F, Bringhen S, Falcone A, Pescosta N, Callea V, Caravita T, Morabito F, Magarotto V, Ruggeri M, Avonto I, Musto P, Cascavilla N, Bruno B, Boccadoro M, 2008, Bortezomib, doxorubicin and dexamethasone in advanced multiple myeloma. Ann Oncol 19(6):1160–1165 Telek B,Mehes L, Batar P, Kiss A, UdvardyM(2008, Effective PAD, bortezomib, doxorubicin, dexamethasone, treatment of a patient with plasma cell leukaemia that has developed after autologous stem cell transplantation. Orv Hetil 149(41):1957–1959 Oakervee HE, Popat R, Curry N, Smith P, Morris C, Drake M, Agrawal S, Stec J, Schenkein D, Esseltine DL, Cavenagh JD, 2005, PAD combination therapy, PS-341/bortezomib, doxorubicin and dexamethasone, for previously untreated patients with multiple myeloma. Br J Haematol 129(6):755–762 Popat R, Oakervee HE, Hallam S, Curry N, Odeh L, Foot N, Esseltine DL, Drake M, Morris C, Cavenagh JD, 2008, Bortezomib, doxorubicin and dexamethasone, PAD, front-line treatment ofmultiplemyeloma: updated results after long-term follow-up. Br J Haematol 141(4):512–516 Greipp PR, SanMiguel J,Durie BG, Crowley JJ, Barlogie B, Blade J, Boccadoro M, Child JA, Avet-Loiseau H, Kyle RA, Lahuerta JJ, LudwigH, Morgan G, Powles R, ShimizuK, Shustik C, Sonneveld P, Tosi P, Turesson I, Westin J, 2005, International staging system for multiple myeloma. J Clin Oncol 23(15):3412–3420 Durie BG, Harousseau JL,Miguel JS, Blade J, Barlogie B, Anderson K, Gertz M, Dimopoulos M, Westin J, Sonneveld P, Ludwig H, Gahrton G, Beksac M, Crowley J, Belch A, Boccadaro M, Cavo M, Turesson I, Joshua D, Vesole D, Kyle R, Alexanian R, Tricot G, Attal M, Merlini G, Powles R, Richardson P, Shimizu K, Tosi P, Morgan G, Rajkumar SV, 2006, International uniform response criteria for multiple myeloma. Leukemia 20(9):1467–1473 Min H, Hui W, Ji P, 2006, Genetic Polymorphism of humanCYP3A4gene in population of Han nationality in China. Chin J Clin Pharmacol Ther 11(3):300–304 Hsieh KP, Lin YY, Cheng CL, Lai ML, Lin MS, Siest JP, Huang JD, 2001, Novel mutations of CYP3A4 in Chinese. Drug Metab Dispos 29(3):268–273 Wei JY, Tong HY, Zhu WF, Liu H, Zhang FJ, Yu WJ, Jin J, 2009, Bortezomib in treatment of extramedullary plasmacytoma of the pancreas. Hepatobiliary Pancreat Dis Int 8(3):329–331 Obeng EA, Carlson LM, Gutman DM, Harrington WJ Jr, Lee KP, Boise LH, 2006, Proteasome inhibitors induce a terminal unfolded protein response inmultiple myeloma cells. Blood 107(12):4907–4916 Meister S, Schubert U, Neubert K, Herrmann K, Burger R, Gramatzki M, Hahn S, Schreiber S, Wilhelm S, Herrmann M, Jack HM, Voll RE, 2007, Extensive immunoglobulin production sensitizes myeloma cells for proteasome inhibition. Cancer Res 67(4): 1783–1792 Gotoh A, Ohyashiki K, Oshimi K, Usui N, Hotta T, Dan K, Ikeda Y, 2006, Lung injury associated with bortezomib therapy in relapsed/ refractory multiple myeloma in Japan: a questionnaire-based report from the “lung injury by bortezomib” joint committee of the Japanese society of hematology and the Japanese society of clinical hematology. Int J Hematol 84(5):406–412 Miyakoshi S, Kami M, Yuji K, Matsumura T, TakatokuM, SasakiM, Narimatsu H, Fujii T, KawabataM, Taniguchi S, Ozawa K, Oshimi K, 2006, Severe pulmonary complications in Japanese patients after bortezomib treatment for refractory multiplemyeloma. Blood 107(9): 3492–3494 Tong Y, Qian J, Li Y, Meng H, Jin J, 2007, The high incidence of varicella herpes zoster with the use of bortezomib in 10 patients. Am J Hematol 82(5):403–404 Reece DE, Sullivan D, Lonial S, Mohrbacher AF, Chatta G, Shustik C, Burris H 3rd, Venkatakrishnan K, Neuwirth R, RiordanWJ, Karol M, von Moltke LL, Acharya M, Zannikos P, Keith Stewart A, 2011, Pharmacokinetic and pharmacodynamic study of two doses of bortezomib in patients with relapsed multiple myeloma. Cancer Chemother Pharmacol 67(1):57–67 Yuan ZG, Jin J, Huang XJ, Li Y, ChenWM, Liu ZG, Chen XQ, Shen ZX, Hou J, 2011, Different dose combinations of bortezomib and dexamethasone in the treatment of relapsed or refractory myeloma: an open-label, observational,multi-center study in China. Chin Med J 124(19):2969–2974 Petrucci M, Blau I, Corradini P, Dimopoulos M, Drach J, Giraldo P, Teixeira A, Blade J, 2010, Efficacy and safety of retreatment with bortezomib in patients with multiple myeloma: interim results from RETRIEVE, a prospective international phase 2 study. Haematologica 95(s2):152 Lonial S, Waller EK, Richardson PG, Jagannath S, Orlowski RZ, Giver CR, Jaye DL, Francis D, Giusti S, Torre C, Barlogie B, Berenson JR, Singhal S, Schenkein DP, Esseltine DL, Anderson J, Xiao H, Heffner LT, Anderson KC, 2005, Risk factors and kinetics of thrombocytopenia associated with bortezomib for relapsed, refractory multiple myeloma. Blood 106(12):3777– 3784 Berges C, Haberstock H, Fuchs D, Miltz M, Sadeghi M, Opelz G, Daniel V, Naujokat C, 2008, Proteasome inhibition suppresses essential immune functions of human CD4+ T cells. Immunology 124(2):234–246 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2014 VL 20 IS 4 BP 987 EP 995 DI 10.1007/s12253-014-9785-7 PG 9 ER PT J AU Sulyok, M Makara, M Ujhelyi, E Valyi-Nagy, I AF Sulyok, Mihaly Makara, Mihaly Ujhelyi, Eszter Valyi-Nagy, Istvan TI Non-Hodgkin Lymphoma and Hepatitis C: Where We are and What Next? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Hepatitis C; Cryoglobulinemia; Non-Hodgkin lymphoma; Direct acting antivirals; Interferon ID Hepatitis C; Cryoglobulinemia; Non-Hodgkin lymphoma; Direct acting antivirals; Interferon AB The association between hepatitis C virus and certain B-cell non-Hodgkin lymphomas, such as marginal zone lymphomas, is supported by epidemiological studies. The exact pathogenetic mechanism is still unknown but both chronic antigenic stimulation and viral lymphotropism may contribute to the evolution of the malignant clone. Furthermore, the hematologic response following hepatitis C antiviral treatment suggests that the virus may have an etiologic role. Interferon and ribavirin based treatment proved to be successful in small case series of hepatitis C virus associated splenic lymphoma with villous lymphocytes, therefore, it is suggested that antiviral treatment could be an alternative to chemo-immunotherapy. In the near future new more potent direct acting antivirals will make interferon free treatments possible. It is still an open question whether these new shortcourse regimens are also effective in the treatment of associated lymphomas and what is the importance of the lymphoid reservoir in eliminating HCV. C1 [Sulyok, Mihaly] St. Istvan and St Laszlo Hospital, Hepatology Center, Albert Florian str. 5-7, 1097 Budapest, Hungary. [Makara, Mihaly] St. Istvan and St Laszlo Hospital, Hepatology Center, Albert Florian str. 5-7, 1097 Budapest, Hungary. [Ujhelyi, Eszter] St. Istvan and St Laszlo Hospital, Immunology LaboratoryBudapest, Hungary. [Valyi-Nagy, Istvan] St. Istvan and St Laszlo Hospital, Hepatology Center, Albert Florian str. 5-7, 1097 Budapest, Hungary. RP Sulyok, M (reprint author), St. Istvan and St Laszlo Hospital, Hepatology Center, 1097 Budapest, Hungary. EM sulyok.mihaly@gmail.com CR Lavanchy D, 2011, Evolving epidemiology of hepatitis C virus. Clin Microbiol Infect Off Publ Eur Soc Clin Microbiol Infect Dis 17(2): 107–115., DOI 10.1111/j.1469-0691.2010.03432.x European Association for the Study of the L, 2011, EASL Clinical Practice Guidelines: management of hepatitis C virus infection. J Hepatol 55(2):245–264., DOI 10.1016/j.jhep.2011.02.023 Monti G, Pioltelli P, Saccardo F, Campanini M, Candela M, Cavallero G, De Vita S, Ferri C, Mazzaro C, Migliaresi S, Ossi E, Pietrogrande M, Gabrielli A, Galli M, Invernizzi F, 2005, Incidence and characteristics of non-Hodgkin lymphomas in a multicenter case file of patients with hepatitis C virus-related symptomatic mixed cryoglobulinemias. Arch Intern Med 165(1):101–105., DOI 10.1001/ archinte.165.1.101 Suarez F, Lortholary O, Hermine O, Lecuit M, 2006, Infectionassociated lymphomas derived from marginal zone B cells: a model of antigen-driven lymphoproliferation. Blood 107(8):3034–3044., DOI 10.1182/blood-2005-09-3679 Arcaini L, Lazzarino M, Colombo N, Burcheri S, Boveri E, Paulli M, Morra E, GambacortaM, Cortelazzo S, Tucci A, Ungari M, Ambrosetti A, Menestrina F, Orsucci L, Novero D, Pulsoni A, FrezzatoM, Gaidano G, Vallisa D, Minardi V, Tripodo C, Callea V, Baldini L, Merli F, Federico M, Franco V, Iannitto E, Integruppo Italiano L, 2006, Splenic marginal zone lymphoma: a prognostic model for clinical use. Blood 107(12):4643–4649., DOI 10.1182/blood-2005-11-4659 Saadoun D, Suarez F, Lefrere F, Valensi F, Mariette X, Aouba A, Besson C, Varet B, Troussard X, Cacoub P, Hermine O, 2005, Splenic lymphoma with villous lymphocytes, associated with type II cryoglobulinemia and HCVinfection: a new entity? Blood 105(1): 74–76., DOI 10.1182/blood-2004-05-1711 Arcaini L, Paulli M, Burcheri S, Rossi A, Spina M, Passamonti F, Lucioni M, Motta T, Canzonieri V, Montanari M, Bonoldi E, Gallamini A, Uziel L, Crugnola M, Ramponi A, Montanari F, Pascutto C, Morra E, Lazzarino M, Intergruppo Italiano L, 2007, Primary nodal marginal zone B-cell lymphoma: clinical features and prognostic assessment of a rare disease. Br J Haematol 136(2):301– 304., DOI 10.1111/j.1365-2141.2006.06437.x Cavalli F, Isaacson PG, Gascoyne RD, Zucca E, 2001, MALT lymphomas. Hematol/Educ Program Am Soc Hematol Am Soc Hematol Educ Program 2001(1):241–58., DOI 10.1182/ asheducation202001.1.241 Arcaini L, Burcheri S, Rossi A, Paulli M, Bruno R, Passamonti F, Brusamolino E,Molteni A, PulsoniA, CoxMC, Orsucci L, Fabbri A, Frezzato M, Voso MT, Zaja F, Montanari F, Merli M, Pascutto C, Morra E, Cortelazzo S, Lazzarino M, 2007, Prevalence of HCV infection in nongastric marginal zone B-cell lymphoma of MALT. Ann Oncol Off J Eur Soc Med Oncol / ESMO 18(2):346–350., DOI 1093/annonc/mdl388 10. Ambrosetti A, Zanotti R, Pattaro C, Lenzi L, Chilosi M, Caramaschi P, Arcaini L, Pasini F, Biasi D, Orlandi E, D’Adda M, Lucioni M, Pizzolo G, 2004, Most cases of primary salivary mucosa-associated lymphoid tissue lymphoma are associated either with Sjoegren syndrome or hepatitis C virus infection. Br J Haematol 126(1):43–49., DOI 10.1111/j.1365-2141.2004.04993.x Ferreri AJ, Viale E, GuidoboniM, Resti AG, De Conciliis C, Politi L, Lettini AA, Sacchetti F, Dolcetti R, Doglioni C, Ponzoni M, 2006, Clinical implications of hepatitis C virus infection in MALT-type lymphoma of the ocular adnexa. Ann Oncol Off J Eur Soc Med Oncol / ESMO 17(5):769–772., DOI 10.1093/annonc/mdl027 Paulli M, Arcaini L, Lucioni M, Boveri E, Capello D, Passamonti F, MerliM, Rattotti S, Rossi D, Riboni R, Berti E,Magrini U, Bruno R, Gaidano G, Lazzarino M, 2010, Subcutaneous ‘lipoma-like’ B-cell lymphoma associated with HCV infection: a new presentation of primary extranodal marginal zone B-cell lymphoma of MALT. Ann Oncol Off J Eur Soc Med Oncol / ESMO 21(6):1189–1195., DOI 10. 1093/annonc/mdp454 Leleu X, O’Connor K, Ho AW, Santos DD, Manning R, Xu L, Hatjiharissi E, Moreau AS, Branagan AR, Hunter ZR, Dimmock EA, Soumerai J, Patterson C, Ghobrial I, Treon SP, 2007, Hepatitis C viral infection is not associated with Waldenstrom’s macroglobulinemia. Am J Hematol 82(1):83–84., DOI 10.1002/ajh.20724 Arcaini L, Merli M, Volpetti S, Rattotti S, Gotti M, Zaja F, 2012, Indolent B-cell lymphomas associated with HCV infection: clinical and virological features and role of antiviral therapy. Clin Dev Immunol 2012:638185., DOI 10.1155/2012/638185 Pellicelli AM,MarignaniM, Zoli V, RomanoM,Morrone A, Nosotti L, Barbaro G, Picardi A, Gentilucci UV, Remotti D, D’Ambrosio C, Furlan C,Mecenate F, Mazzoni E,Majolino I, Villani R, Andreoli A, Barbarini G, 2011, Hepatitis C virus-related B cell subtypes in non Hodgkin’s lymphoma.World J Hepatol 3(11):278–284., DOI 10.4254/ wjh.v3.i11.278 Besson C, Canioni D, Lepage E, Pol S, Morel P, Lederlin P, Van Hoof A, Tilly H, Gaulard P, Coiffier B, Gisselbrecht C, Brousse N, Reyes F, Hermine O, Groupe d’Etude des Lymphomes de l’Adulte P, 2006, Characteristics and outcome of diffuse large B-cell lymphoma in hepatitis C virus-positive patients in LNH 93 and LNH 98 Groupe d’Etude des Lymphomes de l’Adulte programs. J Clin Oncol Off J Am Soc Clin Oncol 24(6):953–960., DOI 10.1200/JCO. 2005.01.5016 Visco C, Arcaini L, Brusamolino E, Burcheri S, Ambrosetti A, Merli M, Bonoldi E, Chilosi M, Viglio A, Lazzarino M, Pizzolo G, Rodeghiero F, 2006, Distinctive natural history in hepatitis C virus positive diffuse large B-cell lymphoma: analysis of 156 patients from northern Italy. Ann Oncol Off J Eur Soc Med Oncol / ESMO 17(9): 1434–1440., DOI 10.1093/annonc/mdl131 Talamini R, Montella M, Crovatto M, Dal Maso L, Crispo A, Negri E, Spina M, Pinto A, Carbone A, Franceschi S, 2004, Non- Hodgkin’s lymphoma and hepatitis C virus: a case-control study from northern and southern Italy. Int J Cancer J Int Cancer 110(3): 380–385., DOI 10.1002/ijc.20137 GuadagninoV, Stroffolini T, RapicettaM, Costantino A, Kondili LA, Menniti-Ippolito F, Caroleo B, Costa C, Griffo G, Loiacono L, Pisani V, Foca A, Piazza M, 1997, Prevalence, risk factors, and genotype distribution of hepatitis C virus infection in the general population: a community-based survey in southern Italy. Hepatology 26(4):1006– 1011., DOI 10.1002/hep.510260431 Collier JD, Zanke B, Moore M, Kessler G, Krajden M, Shepherd F, Heathcote J, 1999, No association between hepatitis C and B-cell lymphoma. Hepatology 29(4):1259–1261., DOI 10.1002/hep. 510290422 Gisbert JP, Garcia-Buey L, Pajares JM, Moreno-Otero R, 2003, Prevalence of hepatitis C virus infection in porphyria cutanea tarda: systematic review and meta-analysis. J Hepatol 39(4):620–627 Matsuo K, Kusano A, Sugumar A, Nakamura S, Tajima K, Mueller NE, 2004, Effect of hepatitis C virus infection on the risk of non- Hodgkin’s lymphoma: a meta-analysis of epidemiological studies. Cancer Sci 95(9):745–752 Dal Maso L, Franceschi S, 2006, Hepatitis C virus and risk of lymphoma and other lymphoid neoplasms: a meta-analysis of epidemiologic studies. Cancer Epidemiol Biomarkers Prev 15(11):2078– 2085., DOI 10.1158/1055-9965.EPI-06-0308 Nieters A, Kallinowski B, Brennan P, Ott M, Maynadie M, Benavente Y, Foretova L, Cocco PL, Staines A, Vornanen M, Whitby D, Boffetta P, Becker N, De Sanjose S, 2006, Hepatitis C and risk of lymphoma: results of the European multicenter casecontrol study EPILYMPH. Gastroenterology 131(6):1879–1886., DOI 10.1053/j.gastro.2006.09.019 de Sanjose S, Benavente Y, Vajdic CM, Engels EA, Morton LM, Bracci PM, Spinelli JJ, Zheng T, Zhang Y, Franceschi S, Talamini R, Holly EA, Grulich AE, Cerhan JR, Hartge P, Cozen W, Boffetta P, Brennan P, Maynadie M, Cocco P, Bosch R, Foretova L, Staines A, Becker N, Nieters A, 2008, Hepatitis C and non-Hodgkin lymphoma among 4784 cases and 6269 controls from the International Lymphoma Epidemiology Consortium. Clin Gastroenterol Hepat Off Clin Pract J Am Gastroenterol Assoc 6(4):451–458., DOI 10. 1016/j.cgh.2008.02.011 Hermine O, Lefrere F, Bronowicki JP, Mariette X, Jondeau K, Eclache-Saudreau V, Delmas B, Valensi F, Cacoub P, Brechot C, Varet B, Troussard X, 2002, Regression of splenic lymphoma with villous lymphocytes after treatment of hepatitis C virus infection. N Engl J Med 347(2):89–94., DOI 10.1056/NEJMoa013376 Kelaidi C, Rollot F, Park S, Tulliez M, Christoforov B, Calmus Y, Podevin P, Bouscary D, Sogni P, Blanche P, Dreyfus F, 2004, Response to antiviral treatment in hepatitis C virus-associated marginal zone lymphomas. Leukemia 18(10):1711–1716., DOI 10.1038/ sj.leu.2403443 Tursi A, Brandimarte G, Torello M, 2004, Disappearance of gastric mucosa-associated lymphoid tissue in hepatitis C virus-positive patients after anti-hepatitis C virus therapy. J Clin Gastroenterol 38(4): 360–363 Mazzaro C, De Re V, Spina M, Dal Maso L, Festini G, Comar C, Tirelli U, Pozzato G, 2009, Pegylated-interferon plus ribavirin for HCV-positive indolent non-Hodgkin lymphomas. Br J Haematol 145(2):255–257., DOI 10.1111/j.1365-2141.2008.07565.x Peveling-Oberhag J, Arcaini L, Hansmann ML, Zeuzem S, 2013, Hepatitis C-associated B-cell non-Hodgkin lymphomas. Epidemiology, molecular signature and clinical management. J Hepatol 59(1):169–177., DOI 10.1016/j.jhep.2013.03.018 Vallisa D, Bernuzzi P, Arcaini L, Sacchi S, Callea V, Marasca R, Lazzaro A, Trabacchi E, Anselmi E, Arcari AL, Moroni C, Berte R, LazzarinoM, Cavanna L, 2005, Role of anti-hepatitis C virus, HCV, treatment in HCV-related, low-grade, B-cell, non-Hodgkin’s lymphoma: a multicenter Italian experience. J Clin Oncol Off J Am Soc Clin Oncol 23(3):468–473., DOI 10.1200/JCO.2005.06.008 Patriarca F, Silvestri F, Fanin R, Zaja F, Sperotto A, Baccarani M, 2001, Long-lasting complete remission of hepatitis C virus, HCV, infection and HCV-associated immunocytoma with alpha-interferon treatment. Br J Haematol 112(2):370–372 Casato M, Mecucci C, Agnello V, Fiorilli M, Knight GB, Matteucci C, Gao L, Kay J, 2002, Regression of lymphoproliferative disorder after treatment for hepatitis C virus infection in a patient with partial trisomy 3, Bcl-2 overexpression, and type II cryoglobulinemia. Blood 99(6):2259–2261 Caramaschi P, Biasi D, Carletto A, Ambrosetti A,Mocella S, Randon M, Bambara LM, 1999, MALT lymphomas of the salivary glands. Review of the literature apropos of a case in a patient with hepatitis C virus infection. Recenti Prog Med 90(11):585–591 Bauduer F, 1996, MALT non-Hodgkin’s lymphoma associated with hepatitis C virus infection treated by interferon alpha. Am J Hematol 53(3):209., DOI 10.1002/(SICI)1096-8652(199611)53:3<209::AIDAJH18> 3.0.CO;2-8 Pitini V, Arrigo C, Righi M, Scaffidi M, Sturniolo G, 2004, Systematic screening for HCV infection should be performed in patients with splenic marginal zone lymphoma. Br J Haematol 124(2):252–253 Moccia F, Tognoni E, Boccaccio P, 1999, The relationship between splenic marginal zone B-cell lymphoma and chronic liver disease associated with hepatitis C virus infection. Ann Ital Med Int Organo Uff Soc Ital Med Int 14(4):288–293 OdaY,Kou T, WatanabeM, Sakuma Y, TaguchiN,KatoY,KudoY, Yamauchi A, Sugiura Y, Ohashi S, Asada M, Fukunaga T, Kawaguchi K, Ito H, Nakamura T, Yazumi S, 2010, Regression of B-cell lymphoma of the liver with hepatitis C virus infection after treatment with pegylated interferon-alpha and ribavirin. Dig Dis Sci 55(6):1791–1793., DOI 10.1007/s10620-009-0902-5 Mauro E, Pedata M, Ermacora A, Mazzaro C, 2012, An additional line of therapy with pegylated interferon and ribavirin after rituximab in a patient with hepatitis C virus-related mixed cryoglobulinaemia and indolent non-Hodgkin’s lymphoma previously treated with interferon. Blood Transfus Trasfus Sang 10(1):101–103., DOI 10.2450/ 2011.0006-11 Takahashi K, Nishida N, Kawabata H, Haga H, Chiba T, 2012, Regression of Hodgkin lymphoma in response to antiviral therapy for hepatitis C virus infection. Intern Med 51(19):2745–2747 Svoboda J, Andreadis C, Downs LH, Miller WT Jr, Tsai DE, Schuster SJ, 2005, Regression of advanced non-splenic marginal zone lymphoma after treatment of hepatitis C virus infection. Leuk Lymphoma 46(9):1365–1368., DOI 10.1080/ 104281905001028289 Iannitto E, Ammatuna E, Tripodo C,Marino C, CalvarusoG, Florena AM,MontaltoG, FrancoV(2004, Long-lasting remission of primary hepatic lymphoma and hepatitis C virus infection achieved by the alpha-interferon treatment. Hematol J Off J Eur Haematol Assoc / EHA 5(6):530–533., DOI 10.1038/sj.thj.6200408 La Mura V, De Renzo A, Perna F, D’Agostino D, Masarone M, Romano M, Bruno S, Torella R, Persico M, 2008, Antiviral therapy after complete response to chemotherapy could be efficacious in HCV-positive non-Hodgkin’s lymphoma. J Hepatol 49(4):557–563., DOI 10.1016/j.jhep.2008.06.025 De Re V, De Vita S, Marzotto A, Gloghini A, Pivetta B, Gasparotto D, Cannizzaro R, Carbone A, Boiocchi M, 2000, Pre-malignant and malignant lymphoproliferations in an HCV-infected type II mixed cryoglobulinemic patient are sequential phases of an antigen-driven pathological process. Int J Cancer J Int Cancer 87(2):211–216 Quinn ER, Chan CH, Hadlock KG, Foung SK, Flint M, Levy S, 2001, The B-cell receptor of a hepatitis C virus, HCV)-associated non-Hodgkin lymphoma binds the viral E2 envelope protein, implicating HCV in lymphomagenesis. Blood 98(13):3745–3749 Flint M, McKeating JA, 2000, The role of the hepatitis C virus glycoproteins in infection. Rev Med Virol 10(2):101–117 Peveling-Oberhag J, Crisman G, Schmidt A, Doring C, Lucioni M, Arcaini L, Rattotti S, Hartmann S, Piiper A, Hofmann WP, Paulli M, Kuppers R, Zeuzem S, Hansmann ML, 2012, Dysregulation of global microRNA expression in splenic marginal zone lymphoma and influence of chronic hepatitis C virus infection. Leukemia 26(7): 1654–1662., DOI 10.1038/leu.2012.29 Schneider P, MacKay F, Steiner V, Hofmann K, Bodmer JL, Holler N, Ambrose C, Lawton P, Bixler S, Acha-Orbea H, Valmori D, Romero P, Werner-Favre C, Zubler RH, Browning JL, Tschopp J, 1999, BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth. J Exp Med 189(11):1747–1756 Mackay F, Woodcock SA, Lawton P, Ambrose C, Baetscher M, Schneider P, Tschopp J, Browning JL, 1999, Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med 190(11):1697–1710 FabrisM, Quartuccio L, Sacco S, De Marchi G, Pozzato G,Mazzaro C, Ferraccioli G, Migone TS, De Vita S, 2007, B-Lymphocyte stimulator, BLyS, up-regulation in mixed cryoglobulinaemia syndrome and hepatitis-C virus infection. Rheumatology 46(1):37–43., DOI 10.1093/rheumatology/kel174 Sene D, Limal N, Ghillani-Dalbin P, Saadoun D, Piette JC, Cacoub P, 2007, Hepatitis C virus-associated B-cell proliferation–the role of serumB lymphocyte stimulator, BLyS/BAFF). Rheumatology 46(1): 65–69., DOI 10.1093/rheumatology/kel177 Rowan AG, Fletcher JM, Ryan EJ, Moran B, Hegarty JE, O’Farrelly C, Mills KH, 2008, Hepatitis C virus-specific Th17 cells are suppressed by virus-induced TGF-beta. J Immunol 181(7):4485–4494 Forghieri F, Luppi M, Barozzi P, Maffei R, Potenza L, Narni F, Marasca R, 2012, Pathogenetic mechanisms of hepatitis C virus-induced B-cell lymphomagenesis. Clin Dev Immunol 2012:807351., DOI 10.1155/2012/807351 Zuckerman E, Zuckerman T, Sahar D, Streichman S, Attias D, Sabo E, Yeshurun D, Rowe J, 2001, bcl-2 and immunoglobulin gene rearrangement in patients with hepatitis C virus infection. Br J Haematol 112(2):364–369 Zuckerman E, Zuckerman T, Sahar D, Streichman S, Attias D, Sabo E, Yeshurun D, Rowe JM, 2001, The effect of antiviral therapy on t(14;18, translocation and immunoglobulin gene rearrangement in patients with chronic hepatitis C virus infection. Blood 97(6):1555–1559 Machida K, Cheng KT, Sung VM, Lee KJ, Levine AM, Lai MM, 2004, Hepatitis C virus infection activates the immunologic, type II, isoform of nitric oxide synthase and thereby enhances DNA damage and mutations of cellular genes. J Virol 78(16):8835–8843., DOI 10. 1128/JVI.78.16.8835-8843.2004 Karavattathayyil SJ, Kalkeri G, Liu HJ, Gaglio P, Garry RF, Krause JR, Dash S, 2000, Detection of hepatitis C virus RNA sequences in B-cell non-Hodgkin lymphoma. Am J Clin Pathol 113(3):391–398., DOI 10.1309/REV9-FDTM-5NGC-HBWY Machida K, Cheng KT, Sung VM, Shimodaira S, Lindsay KL, Levine AM, Lai MY, Lai MM, 2004, Hepatitis C virus induces a mutator phenotype: enhanced mutations of immunoglobulin and protooncogenes. Proc Natl Acad Sci U S A 101(12):4262–4267., DOI 10.1073/pnas.0303971101 Machida K, ChengKT, Pavio N, Sung VM, LaiMM(2005)Hepatitis C virus E2-CD81 interaction induces hypermutation of the immunoglobulin gene in B cells. J Virol 79(13):8079–8089., DOI 10.1128/JVI. 79.13.8079-8089.2005 Bachy E, Besson C, Suarez F, Hermine O, 2010, Hepatitis C virus infection and lymphoma. Mediterr J Hematol Infect Dis 2(1): e2010004., DOI 10.4084/MJHID.2010.004 Manns MP, McHutchison JG, Gordon SC, Rustgi VK, Shiffman M, Reindollar R, Goodman ZD, Koury K, Ling M, Albrecht JK, 2001, Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomised trial. Lancet 358(9286):958–965 Jacobson IM, Gordon SC, Kowdley KV, Yoshida EM, Rodriguez- Torres M, Sulkowski MS, Shiffman ML, Lawitz E, Everson G, Bennett M, Schiff E, Al-Assi MT, Subramanian GM, An D, Lin M, McNally J, Brainard D, Symonds WT, McHutchison JG, Patel K, Feld J, Pianko S, Nelson DR, Study P, Study F, 2013, Sofosbuvir for hepatitis C genotype 2 or 3 in patients without treatment options. N Engl J Med 368(20):1867–1877., DOI 10.1056/NEJMoa1214854 Lawitz E, Mangia A, Wyles D, Rodriguez-Torres M, Hassanein T, Gordon SC, Schultz M, Davis MN, Kayali Z, Reddy KR, Jacobson IM, Kowdley KV, Nyberg L, Subramanian GM, Hyland RH, Arterburn S, Jiang D, McNally J, Brainard D, Symonds WT, McHutchison JG, Sheikh AM, Younossi Z, Gane EJ, 2013, Sofosbuvir for previously untreated chronic hepatitis C infection. N Engl J Med 368(20):1878–1887., DOI 10.1056/NEJMoa1214853 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 1 EP 7 DI 10.1007/s12253-014-9845-z PG 7 ER PT J AU Sikalidis, KA AF Sikalidis, K Angelos TI Amino Acids and Immune Response: A Role for Cysteine, Glutamine, Phenylalanine, Tryptophan and Arginine in T-cell Function and Cancer? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE T-cell; Amino acids; Cysteine; Glutamine; Phenylalanine; Tryptophan; Arginine; Immune response; Signaling; Immunomodulation; Avidity ID T-cell; Amino acids; Cysteine; Glutamine; Phenylalanine; Tryptophan; Arginine; Immune response; Signaling; Immunomodulation; Avidity AB While proteins are critical for immunity, T-cells constitute a critical component of adaptive immunity by clearing cancerous cells among other abnormal cells. However, cancer cells exhibit a potential to escape T-cell control by employing mechanisms not completely delineated. Interesting work has investigated how certain amino acids affect the proliferation rate of T-cells as well as their effectiveness in clearing tumors. The role of amino acids cysteine, glutamine, phenylalanine, tryptophan and arginine in immunomodulation and particularly regarding T-cell proliferation and activation is discussed. The redox balance is reported to affect T-cell proliferation via modulation of cysteine availability. In addition antigen presenting cells (APCs), similar to myeloid cells determine the availability of amino acids in the extracellular microenvironment affecting T-cell proliferation and activation. A better mechanistic understanding of T-cell function modulation via amino acid signaling or metabolic properties may be helpful towards optimization of adaptive immunity with implications for cancer prognosis and treatment. C1 [Sikalidis, K Angelos] Harvard Medical School, Harvard Institutes of Medicine, Dana-Farber Cancer Institute, Immunobiology Laboratory, 77Avenue Louis Pasteur, 02115 Boston, MA, USA. RP Sikalidis, KA (reprint author), Harvard Medical School, Harvard Institutes of Medicine, Dana-Farber Cancer Institute, Immunobiology Laboratory, 02115 Boston, USA. EM angelos_sikalidis@dfci.harvard.edu CR Steinke JW, Lawrence MG, 2014, T-cell biology in immunotherapy. Ann Allergy Asthma Immunol 112:195–199 Smith KA, 2014, Revisiting the first long-term culture of antigenspecific cytotoxic T cells. Front Immunol 5:194 Viret C, Janeway CA Jr, 1999, MHC and T cell development. Rev Immunogenet 1:91–104 von Essen MR, Kongsbak M, Geisler C, 2012, Mechanisms behind functional avidity maturation in T cells. Clin Dev Immunol 2012: 163453 RobinsonAT,Miller N,Alexander DR, 1993, CD3 antigen-mediated calcium signals and protein kinase C activation are higher in CD45RO+ than in CD45RA+ human T lymphocyte subsets. Eur J Immunol 23(1):61–68 von Essen MR, Kongsbak M, Schjerling P, Olgaard K, Odum N, Geisler C, 2010, Vitamin D controls Tcell antigen receptor signaling and activation of human T cells. Nat Immunol 11(4):344–349 Ericsson PO, Orchansky PL, Carlow DA, Teh HS, 1996, Differential activation of phospholipase C-γ1 and mitogen-activated protein kinase in naive and antigen-primed CD4 T cells by the peptide/MHC ligand. J Immunol 156(6):2045–2053 Vigano S, Utzschneider DT, Perreau M, Pantaleo G, Zehn D, Harari A, 2012, Functional avidity: a measure to predict the efficacy of effector T cells? Clin Dev Immunol 153863 Ebert PJ, Jiang S, Xie J, Li QJ, Davis MM, 2009, An endogenous positively selecting peptide enhances mature T cell responses and becomes an autoantigen in the absence of microRNA miR-181a. Nat Immunol 10(11):1162–1169 Garcia Z, Pradelli E, Celli S, Beuneu H, Simon A, Bousso P, 2007, Competition for antigen determines the stability of T cell-dentritic cell interactions during clonal expansion. Proc Natl Acad Sci U S A 104(11):4553–4558 McMahan RH, Slansky JE, 2007, mobilizing the low-avidity T cell repertoire to kill tumors. Semin Cancer Biol 17(4):317–329 Choi EML, Chen JL, Wooldridge et al, 2003, High avidity antigenspecific CTL identified by CD8-idependent tetramer staining. J Immunol 171(10):5116–5123 Xiao Z, Mescher MF, Jameson SC, 2007, Detuning CD8 T cells: down-regulation of CD8 expression, tetramer binding, and response during CTL activation. J Exp Med 294(11):2667–2677 Slifka MK, Whitton JL, 2001, Functional avidity maturation of CD8+ T cells without selection of higher affinity TCR. Nat Immunol 2(8):711–717 Rechtsteiner G,Warger T, Hofmann M, Rammensee HG, Schild HJ, Radsak MP, 2006, Precursor frequency can compensate for lower TCR expression in T cell competition during priming in vivo. Eur J Immunol 36(10):2613–2623 Sikalidis AK, Lee J-I, Stipanuk MH, 2011, Cellular responses to indispensable amino acid deficiency are mediated through the integrated stress response pathway. Amino Acids 41(1):159–171 Sikalidis AK, Stipanuk MH, 2010, Growing rats respond to a sulfur amino acid-deficient diet by phosphorylation of eIF2α and induction of adaptive components of the integrated stress. J Nutr 140(6):1080–1085 Bella DL, Hirschberger LL, Hosokawa Y, Stipanuk MH, 1999, The mechanisms involved in the regulation of key enzymes of cysteine metabolism in rat liver in vivo. Am J Physiol 276, Endocrin. Metab. 39): E326–E335 Bella DL, Hahn C, Stipanuk MH, 1999, The effects of non-sulfur amino acids and of sulfur amino acids on the regulation of hepatic enzymes of cysteine metabolism. Am J Phsyiol 277(Endocrin. Metab. 40): E144-E153 Cresenzi CL, Lee JI, Stipanuk MH, 2003, Cysteine is the metabolic signal responsible for dietary regulation of hepatic cysteine dioxygenase and glutamate cysteine ligase in intact rats. J Nutr 133(9):2697–2702 Yang BH,Wang X, Ren X, 2012, Amino acid metabolism related to immune tolerance by MDSCs. Int Rev Immunol 31(3):177–183 Rodriguez PC, Ochoa AC, 2006, Tcell dysfunction in cancer: role of myeloid cells and tumor cells regulating amino acid availability and oxidative stress. Semin Cancer Biol 16(1):66–72 Levring TB, Hansen AK, Nielsen BL, Kongsbak M, von Essen MR, Woetmann A, Odum N, Bonefeld CM, Geisler C, 2012, Activated human CD4+ T cells express transporters for both cysteine and cystine. Sci Rep 2:266 Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand- Rosenberg S, 2010, Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res 70(1):68–77 Yan Z, Banerjee R, 2010, Redox remodeling as an immunoregulatory strategy. Biochemistry 49(6):1059–1066 Sido B, Lasitschka F, Giese T, Gassler N, Funke B, Schroder- Braunstein J, Brunnemer U, Meuer SC, Autschbach F, 2008, A prominent role for mucosal cystine/cysteine metabolism in intestinal immunoregulation. Gastroenterology 134(1):179–191 Yan Z, Garg SK, Banerjee R, 2010, Regulatory T cells interfere with glutathione metabolism in dendritic cells and T cells. J Biol Chem 285(53):41525–41532 Edinger AL, Thompson CB, 2002, Antigen-presenting cells control T cell proliferation by regulating amino acid availability. Proc Natl Acad Sci U S A 99(3):1107–1109 Droge W, Eck HP, Gmunder H, Mihm S, 1991, Dysregulation of plasma amino acid levels in HIV-infection and cancer and its relevance for the immune system. Amino Acids 1(2):193–198 Mihm S, Galter D, DrogeW(1995)Modulation of transcription factor NF kappa B activity by intracellular glutathione levels and by variations of the extracellular cysteine supply. FASEB J 9(2):246–252 Droge W, Eck HP, Gmunder H, Mihm S, 1991, Modulation of lymphocyte functions and immune response by cysteine and cysteine derivatives. Am J Med 91(3C):140S–144S Grimble RF, 2006, The effects of sulfur amino acid intake on immune function in humans. J Nutr 136(6 Suppl):1660S–1665S Murphy G, Fan JH,Mark SD, Dawsey SM, Selhub J,Wang J, Taylor PR, Qiao YL, Abnet CC, 2011, Prospective study of serum cysteine levels and oesophageal and gastric cancers in China. Gut 60(5):618– 623 Gostner JM, Becker K, Fuchs D, Sucher R, 2013, Redox regulation of the immune response. Redox Rep 18(3):88–94 Yan Z, Garg SK, Kipnis J, Banerjee R, 2009, Extracellular redox modulation by regulatory T cells. Nat Chem Biol 5(10):721–723 Matsue H, Edelbaum D, Shalhevet D, Mizumoto N, Yang C, Mummert ME et al, 2003, Generation and function of reactive oxygen species in dendritic cells during antigen presentation. J Immunol 171(6):3010–3018 Los M, Schenk H, Hexel K, Baeuerle PA, DrogeW, Schulze-Osthoff K, 1995, IL-2 gene expression and NF-kappa B activation through CD28 requires reactive oxygen production by 5-lipoxygenase. EMBO J 14(15):3731–3740 Miller TW, Wang EA, Gould S, Stein EV, Kaur S, Lim L, Amarnath S, Fowler DH, Roberts DD, 2012, Hydrogen sulfide is an endogenous potentiator of T cell activation. J Biol Chem 287(6):4211–4221 Kontny E, Szczepanska K, Kowalczewski J, KurowskaM, Janicka I, Marcinklewicz J, Maslinski W, 2000, The mechanism of taurine chloramine inhibition of cytokine IL-6, IL-8 production by rheumatoid arthritis fibroblast-like synoviocytes. Arthritis Rheum 43(10): 2169–2177 Grimble RF, 1994, Sulphur amino acids and the metabolic response to cytokines. Adv Exp Med Biol 359:41–49 Huxtable RJ, 1996, Taurine past, present and future. Adv Exp Med Biol 403:641–650 Grimble RF, 1996, Theory and efficacy of antioxidant therapy. Curr Opin Crit Care 2:260–266 Chorazy M, Kontny E, Marcinklewicz J,MaslinskiW(2002, Taurine chloramine modulates cytokine production by human peripheral blood mononuclear cells. Amino Acids 23(4):407–413 Winder B, Lebilhuber F, Frick B, Laich A, Artner-Dworzak E, Fuchs D, 2002, Moderate hyperchromocysteinaemia ad immune activation in Parkinson’s disease. J Neural Transm 109(12):1445–1452 Gottfried E, Kreutz M, Mackensen A, 2012, Tumor metabolism as modulator of immune response and tumor progression. Semin Cancer Biol 22:335–341 Newsholme P, 2001, Why is L-glutamine metabolism important to cells of the immune system in health, postinjury, surgery or infection? J Nutr 131(9 Suppl):2512–2522 Wilmore DW, Shabert JK, 1998, The role of glutamine in immunologic responses. Nutrition 14(4):618–626 Hammami I, Chen J, Bronte V, DeCrescenzo G, Jolicoeur M, 2012, L-glutamine is a key parameter in the immunosuppression phenomenon. Biochem Biophys Res Commun 425(4):724–729 Raber P, Ochoa AC, Paulo C, 2012, Metabolism of L-arginine by myeloid-derived suppressor cells in cancer: mechanisms of T cell suppression and therapeutic perspectives. Immunol Investig 41(6–7): 614–634 Barbul A, 1990, Arginine and immune function. Nutrition 6(6–7): 53–58 Taheri F, Ochoa JB, Faghiri Z, Culotta K, Park HJ, Lan MS et al, 2001, L-Arginine regulates the expression of the T-cell receptor zeta chain, CD3zeta, in Jurkat cells. Clin Cancer Res 7(3 Suppl):958s– 965s Rodrigez PC, Zen AH, Culotta KS, Zabaleta J, Ochoa JB, Ochoa AC, 2002, Regulation of Tcell receptor CD3 zeta chain expression by Larginine. J Biol Chem 277(24):21123–21129 Albina JE, Caldwell MD, Henry WL Jr,Mills CD, 1989, Regulation of macrophage functions by L-arginine. J Exp Med 169(3):1021– 1029 Popovic PJ, Zeh HJ 3rd, Ochoa JB, 2007, Arginine and immunity. J Nutr 137(6 Suppl 2):1681S–1686S DeSanctis F, Sandri S, Ferrarini G, Pagliarello I, Sartoris S, Ugel S, Marigo I, Molon B, Bronte V, 2014, The emerging immunological role of post-translational modifications by reactive nitrogen species in cancer microenvironment. Front Immunol 5:1–16 Terabe M, Swann J, Ambrosino E et al, 2005, A nonclassical non- Valpha14Jalpha18 CD1d-restricted, type II, NKT cells is sufficient for down-regulation of tumor immunosurveillance. J Exp Med 202(12):1627–1633 Takahashi A, Hanson MGV, Norell HR et al, 2005, Preferential cell death of CD8+ effector memory, CCR7−CD45RA−, T cells by hudrogen peroxide-induced oxidative stress. J Immunol, Baltimore, 174(10):6080–6087 Schroder AJ, Pavlidis P, Arimura A, Capece D, Rothman PB, 2002, Cutting edge: STAT6 serves as a positive and negative regulator of gene expression in IL-4-stimulated B lymphocytes. J Immunol 168(3):996–1000 Godin-Ethier J, Hanafi LA, Piccirillo CA, Lapointe R, 2011, Indoleamine 2,3-dioxygenase expression in human cancers: clinical and immunologic perspectives. Clin Cancer Res 17(22):6985–6991 Mellor AL, Munn DH, 2001, Tryptophan catabolism prevents maternal T cells from activating lethal anti-fetal immune responses. J Reprod Immunol 52(1–2):5–13 MunnDH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A,Mellor AL, 1999, Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med 189(9):1363–1372 Lee GK, Park HJ, Macleod M, Chandler P, Munn DH, Mellor AL, 2002, Tryptophan deprivation sensitizes activated T cells to apoptosis prior to cell division. Immunology 107(4):452–460 Weinlich G, Murr C, Richardsen L, Winkler C, Fuchs D, 2007, Decreased serum tryptophan concentration predicts poor prognosis in malignant melanoma patients. Dermatology 214(1):8–14 Brandacher G, Perathoner A, Ladurner R, Schneeberger S, Obrist P, Winkler C, Werner ER, Werner-Felmayer G, Weiss HG, Gobel G, Margreiter R, Konigsrainer A, Fuchs D, Amberger A, 2006, Prognostic value of indoleamine 2,3-dioxygenase expression in colorectal cancer: effect on tumor-infiltrating T cells. Clin Cancer Res 12(4):1144–1151 Ramanathan A, Wang C, Schreiber SL, 2005, Perturbational profiling of a cell-line model of tumorigenesis by using metabolic measurements. Proc Natl Acad Sci U S A 102(17):5992–5997 Elstrom RL, Bauer DE, Buzzai M, Karnauskas R, Harris MH, Plas DR et al, 2004, Akt stimulates aerobic glycolysis in cancer cells. Cancer Res 64(11):3892–3899 Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, Schumacher T, Jestaedt L, Schrenk D, Weller M, Jugold M, Guillemin GJ, Miller CL, Lutz C, Radlwimmer B, Lehmann I, von Deimling A, Wick W, Platten M, 2011, An endogenous tumourpromoting ligand of the human aryl hydrocarbon receptor. Nature 478(7368):197–203 Pilotte L, Larrieu P, Stroobant V, Colau D, Dolusic E, Frederick R, De Plaen E, Uyttenhove C, Wouters J, Masereel B, Van den Eynde BJ, 2012, Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase. Proc Natl Acad Sci U S A 109(7):2497–2502 Platten M,WickW,Van den Eynde BJ, 2012, Tryptophan catabolism in cancer: beyond IDO and tryptophan depletion. Cancer Res 72(21): 5435–5440 Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD, 2002, Cancer immunoediting: from immunosurveillence to tumor escape. Nat Immunol 3(11):991–998 Mizoguchi H, O’Shea JJ, Longo DL, Loeffler CM, McVicar DW, Ochoa AC, 1992, Alterations in signal transduction molecules in T lymphocytes from tumor-bearing mice. Science 258(5089):1795– 1798 Lee J-I, Dominy JE Jr, Sikalidis AK, Hirschberger LL, Wang W, Stipanuk MH, 2008, HepG2/C3A cells respond to cysteinedeprivation by induction of the amino acid deprivation/integrated stress response pathway. Physiol Genomics 33(2):218–229 Sikalidis AK, Mazor KM, Kang M, Liu H, Stipanuk MH, 2013, Total 4EBP1 is elevated in liver of rats in response to low sulfur amino acid intake. J Amino Acids 2013:864757 Sikalidis AK, 2013, Cellular and animal indispensable amino acid limitation responses and health promotion. Can the two be linked? A critical review. Int J Food Sci Nutr 64(3):300–311 Maciolek JA, Pasternak JA, Wilson HL, 2014, Metabolism of activated T lymphocytes. Curr Opin Immunol 27:60–74 Baracos VE,MackenzieML, 2006, Investigations of branched-chain amino acids and their metabolites in animal models of cancer. J Nutr 136(1 Suppl):237S–242S NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 9 EP 17 DI 10.1007/s12253-014-9860-0 PG 9 ER PT J AU Wang, XT Li, DG Li, L Kong, FB Pang, LM Mai, W AF Wang, Xiao-Tong Li, De-Gang Li, Lei Kong, Fan-Biao Pang, Li-Ming Mai, Wei TI Meta-Analysis of Oncological Outcome After Abdominoperineal Resection or Low Anterior Resection for Lower Rectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Low anterior resection; Abdominoperineal resection; Lower rectal cancer; Prognosis ID Low anterior resection; Abdominoperineal resection; Lower rectal cancer; Prognosis AB In lower rectal cancer, postoperative outcome is still subject of controversy between the advocates of abdominoperineal resection (APR) and low anterior resection (LAR). Reports suggest that low anterior resection may be oncologically superior to abdominoperineal excision, although no good evidence exists to support this. Publications were identified which assessed the differences comparing 5- year survival, local recurrence, circumferential resection margin rate, complications and so on. A meta-analysis was performed to clarify the safety and feasibility of the two procedures with several types of outcome measures. A total of 13 studies met the inclusion criteria, and comprised 6,850 cases. Analysis of these data showed that LAR group was highly correlated with 5-year survival (pooled OR=1.73, 95%CI: 1.30–2.29, P=0.0002 random-effect). And local recurrence rate of APR group was significantly higher than that in LAR group (pooled OR=0.63, 95%CI: 0.53–0.75, P<0.00001 fixed-effect). Also, the circumferential resection margin (CRM) were high involved in APR group than in LAR group. (5 trials reported the data, pooled OR=0.43, 95%CI: 0.36– 0.52, P<0.00001 fixed-effect). Besides, the incidents of overall complications of APR group was higher compared with LAR group (pooled OR=0.52, 95%CI: 0.29–0.92, P=0.03 random-effect). Patients treated by APR have a higher rate of CRM involvement, a higher local recurrence, and poorer prognosis than LAR. And there is evidence that in selected low rectal cancer patients, LAR can be used safely with a better oncological outcome than APR. due to the inherent limitations of the present study, for example, the trails available for this systematic review are limited and the finite retrospective data, future prospective randomized controlled trials will be useful to fully investigate these outcome measures and to confirm this conclusion. C1 [Wang, Xiao-Tong] People’s Hospital of Guangxi Zhuang Autonomous Region, Departments of Gastrointestinal and Peripheral Vascular SurgeryNanning, China. [Li, De-Gang] The First Affiliated Hospital of Guangxi University of Chinese Medicine, Departments of SurgeryNanning, China. [Li, Lei] People’s Hospital of Guangxi Zhuang Autonomous Region, Departments of Gastrointestinal and Peripheral Vascular SurgeryNanning, China. [Kong, Fan-Biao] The First Affiliated Hospital of Guangxi University of Chinese Medicine, Departments of SurgeryNanning, China. [Pang, Li-Ming] The First Affiliated Hospital of Guangxi University of Chinese Medicine, Departments of SurgeryNanning, China. [Mai, Wei] People’s Hospital of Guangxi Zhuang Autonomous Region, Departments of Gastrointestinal and Peripheral Vascular SurgeryNanning, China. RP Mai, W (reprint author), People’s Hospital of Guangxi Zhuang Autonomous Region, Departments of Gastrointestinal and Peripheral Vascular Surgery, Nanning, China. EM 13977154858@139.com CR Varpe P, Huhtinen H, Rantala A, Salminen P, Rautava P, Hurme S, Gronroos J, 2011, Quality of life after surgery for rectal cancer with special reference to pelvic floor dysfunction. Color Dis 13(4):399– 405., DOI 10.1111/j.1463-1318.2009.02165.x Bujko K, Rutkowski A, Chang GJ, Michalski W, Chmielik E, Kusnierz J, 2012, Is the 1-cm rule of distal bowel resection margin in rectal cancer based on clinical evidence?A systematic review. Ann Surg Oncol 19(3):801–808., DOI 10.1245/s10434-011-2035-2 Schoetz DJ Jr, 2006, Evolving practice patterns in colon and rectal surgery. J Am Coll Surg 203(3):322–327., DOI 10.1016/j.jamcollsurg. 2006.05.302 Perry WB, Connaughton JC, 2007, Abdominoperineal resection: how is it done and what are the results? Clin Colon Rectal Surg 20(3):213–220., DOI 10.1055/s-2007-984865 Nagtegaal ID, van de Velde CJ, Marijnen CA, van Krieken JH, Quirke P, 2005, Low rectal cancer: a call for a change of approach in abdominoperineal resection. J Clin Oncol 23(36):9257–9264., DOI 10.1200/JCO.2005.02.9231 Li CS,Wan DS, Pan ZZ, Zhou ZW, Chen G, Wu XJ, Li LR, Lu ZH, Ding PR, Li Y, 2006, Multivariate prognostic analysis of patients with low and middle rectal cancer after curative resection. Ai Zheng 25(5):587–590., DOI 10.3321/j.issn:1000-467X.2006.05.013 Shihab OC, Brown G, Daniels IR, Heald RJ, Quirke P, Moran BJ, 2010, Patients with low rectal cancer treated by abdominoperineal excision have worse tumors and higher involved margin rates compared with patients treated by anterior resection. Dis Colon Rectum 53(1):53–56., DOI 10.1007/ DCR. 0b013e3181c70465 ChambersW, Khan A,Waters R, Lindsey I, George B,Mortensen N, Cunningham C, 2010, Examination of outcome following abdominoperineal resection for adenocarcinoma in Oxford. Color Dis 12(12):1192–1197., DOI 10.1111/j.1463-1318.2009.01939.x Marr R, Birbeck K, Garvican J, Macklin CP, Tiffin NJ, Parsons WJ, DixonMF, Mapstone NP, Sebag-Montefiore D, Scott N, Johnston D, Sagar P, Finan P, Quirke P, 2005, The modern abdominoperineal excision: the next challenge after total mesorectal excision. Ann Surg 242(1):74–82., DOI 10.1097/01.sla.0000167926.60908.15 Campos-Lobato LF, Alves-Ferreira PC, Lavery IC, Kiran RP, 2011, Abdominoperineal resection does not decrease quality of life in patients with low rectal cancer. Clin, Sao Paulo, 66(6):1035–1040., DOI 10.1590/S1807-59322011000600019 Chuwa EW, Seow-Choen F, 2006, Outcomes for abdominoperineal resections are not worse than those of anterior resections. Dis Colon Rectum 49(1):41–49., DOI 10.1007/s10350-005-0227-1 Kim JC, Yu CS, Lim SB, Kim CW, Kim JH, Kim TW, 2013, Abdominoperineal resection and low anterior resection: comparison of long-term oncologic outcome in matched patients with lower rectal cancer. Int J Color Dis 28(4):493–501., DOI 10.1007/s00384-012-1590-8 Ramsay G, Parnaby C, Mackay C, Hanlon P, Ong S, Loudon M, 2013, Analysis of outcome using a levator sparing technique of abdominoperineal excision of rectum and anus. Cylindrical ELAPE is not necessary in all patients. Eur J Surg Oncol 39(11):1219–1224., DOI 10.1016/j.ejso.2013.08.004 Keating JP, 2004, Sexual function after rectal excision. ANZ J Surg 74(4):248–259., DOI 10.1111/j.1445-2197.2004.02954.x Wibe A, Syse A, Andersen E, Tretli S, Myrvold HE, Soreide O, 2004, Oncological outcomes after total mesorectal excision for cure for cancer of the lower rectum: anterior vs. abdominoperineal resection. Dis Colon Rectum 47(1):48–58., DOI 10.1007/s10350-003-0012-y Heald RJ, Smedh RK, Kald A, Sexton R, Moran BJ, 1997, Abdominoperineal excision of the rectum–an endangered operation.Norman Nigro Lectureship. Dis Colon Rectum 40(7): 747–751., DOI 10.1007/BF02055425 LawWL, Chu KW(2001, Impact of total mesorectal excision on the results of surgery of distal rectal cancer. Br J Surg 88(12):1607–1612., DOI 10.1046/j.0007-1323.2001.01929.x HeWJ,Wang L, Hu H, Kang SY, Qian HX, Xu FM(2002, Correlation of invasion,metastasis, and prognosis in low and middle rectal cancer.Ai Zheng 21(11):1222–1225., DOI 10.3321/j.issn:1000-467X.2002.11.011 Li CH, 2009, Clinical comparision of two operative methods of adenocatcinoma of middle/low rectum. JMod ClinMed 35(1):61–62 Zolfaghari S, Williams LJ, Moloo H, Boushey RP, 2010, Rectal cancer: current surgical management. Minerva Chir 65(2):197–211 Holm T, Ljung A, Haggmark T, Jurell G, Lagergren J, 2007, Extended abdominoperineal resection with gluteus maximus flap reconstruction of the pelvic floor for rectal cancer. Br J Surg 94(2): 232–238., DOI 10.1002/bjs.5489 Huang LN,Wang DS, Chen YQ, LiW, Hu FD, 2012)Meta-analysis for cyclin E in lung cancer survival. Clin Chim Acta 413:663–668., DOI 10.1016/j.cca.2011.12.020 Fan J, Wang L, Jiang GN, Gao W, 2012, Sublobectomy versus lobectomy for stage I non-small-cell lung cancer, a meta-analysis of published studies. Ann Surg Oncol 19:661–668., DOI 10.1245/ s10434-011-1931-9 Christian P, Tielsch JM, 2012, Evidence for multiple micronutrient effects based on randomized controlled trials and meta-analyses in developing countries. J Nutr 142:173S–177S., DOI 10.3945/jn.111. 149898 Glynne-Jones R,Mawdsley S, Pearce T, BuyseM(2006, Alternative clinical end points in rectal cancer–are we getting closer? Ann Oncol 17(8):1239–1248., DOI 10.1093/annonc/mdl173 Nagtegaal ID, Marijnen CA, Kranenbarg EK, van de Velde CJ, van Krieken JH, 2002, Circumferential margin involvement is still an important predictor of local recurrence in rectal carcinoma: not one millimeter but two millimeters is the limit. Am J Surg Pathol 26(3): 350–357., DOI 10.1097/00000478-200203000-00009 Den Dulk M, Marijnen CA, Putter H, Rutten HJ, Beets GL, Wiggers T, Nagtegaal ID, van de Velde CJ, 2007, Risk factors for adverse outcome in patients with rectal cancer treated with an abdominoperineal resection in the total mesorectal excision trial. Ann Surg 246(1):83–90., DOI 10. 1097/01.sla.0000259432.29056.9d Jorge JM, Wexner SD, 1993, Etiology and management of fecal incontinence. Dis Colon Rectum 36(1):77–97., DOI 10.1007/ BF02050307 Hozo SP, Djulbegovic B, Hozo I, 2005, Estimating the mean and variance from themedian, range, and the size of a sample.BMCMed Res Methodol 5:13., DOI 10.1186/1471-2288-5-13 Takase S, Kamigaki T, Yamashita K et al, 2009, Preoperative chemoradiotherapy for resectable lower rectal cancer. Gan To Kagaku Ryoho 36(12):2006–2008, PMID: 20037306 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 19 EP 27 DI 10.1007/s12253-014-9863-x PG 9 ER PT J AU Kiss, O Tokes, AM Spisak, S Szilagyi, A Lippai, N Szekely, B Szasz, AM Kulka, J AF Kiss, Orsolya Tokes, Anna-Maria Spisak, Sandor Szilagyi, Anna Lippai, Norbert Szekely, Borbala Szasz, Attila Marcell Kulka, Janina TI Breast- and Salivary Gland-Derived Adenoid Cystic Carcinomas: Potential Post-Transcriptional Divergencies. A Pilot Study Based on miRNA Expression Profiling of Four Cases and Review of the Potential Relevance of the Findings SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adenoid cystic carcinoma; microRNA; Breast; Salivary gland ID Adenoid cystic carcinoma; microRNA; Breast; Salivary gland AB Adenoid cystic carcinoma (ACC) is a malignant tumor of the salivary glands but identical tumors can also arise from the breast. Despite their similar histomorphological appearance the salivary gland- and the breast-derived forms differ in their clinical features: while ACC of the salivary glands (sACC) have an agressive clinical course, the breastderived form (bACC) shows a very favourable clinical outcome. To date no exact molecular alterations have yet been identified which would explain the diverse clinical features of the ACCs of different origin. In our pilot experiment we investigated the post-transcriptional features of ACC cases by performing microRNA-profiling on 2-2 bACC and sACC tissues and on 1-1 normal breast and salivary gland tissue. By comparing the microRNA-profiles of the investigated samples we identified microRNAs which were expressed differently in bACC and sACC cases according to their normal controls: 7 microRNAs were overexpressed in sACC cases and downexpressed in bACC tumors (let-7b, let-7c, miR-17, miR-20a, miR-24, miR-195, miR-768-3) while 9 microRNAs were downexpressed in sACC cases and overexpressed in bACC tissues (let-7e, miR-23b, miR-27b, miR-193b, miR-320a, miR-320c, miR-768-5p, miR-1280 and miR-1826) relative to their controls. We also identified 8 microRNAs which were only expressed in sACCs and one microRNA (miR-1234) which was only absent in sACC cases. By target predictor online databases potential targets of the these microRNAs were detected to identify genes that may play central role in the diverse cinical outcome of bACC and sACC cases. C1 [Kiss, Orsolya] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Tokes, Anna-Maria] Hungarian Academy of Sciences, MTA-SE Tumor Progression Research GroupBudapest, Hungary. [Spisak, Sandor] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Szilagyi, Anna] Fejer County Szent Gyorgy HospitalSzekesfehervar, Hungary. [Lippai, Norbert] Jasz-Nagykun-Szolnok County Hetenyi Geza HospitalSzolnok, Hungary. [Szekely, Borbala] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Szasz, Attila Marcell] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Kulka, J (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM kulka.janina@med.semmelweis-univ.hu CR Nkanza NK, 1988, Adenoid cystic carcinoma of the prostate. Cent Afr J Med 34:166–168 Stefani M, Speranza N, 1970, A case of cylindroma of the vagina. Riv Anat Patol Oncol 36:77–105 Tchertkoff V, Sedlis A, 1962, Cylindroma of the cervix. Am J Obstet Gynecol 84:749–752 Paulino AF, Huvos AG, 1999, Epithelial tumors of the lacrimal glands: a clinicopathologic study. Ann Diagn Pathol 3:199–204 Billroth T, 1859, Beobachtungen Uber Geschwulste der Speicheldrusen. Arch Path Anat 17:357–375 Mc LP, Tennant R, Sarokhan J, 1953, Adenoid cystic carcinoma of the breast; report of a case with unusual features. Surgery 33:905–908 Dent R, TrudeauM, Pritchard KI et al, 2007, Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res 13:4429–4434 Ghabach B, Anderson WF, Curtis RE et al, 2010, Adenoid cystic carcinoma of the breast in the United States, 1977 to 2006): a population-based cohort study. Breast Cancer Res 12:R54 Ciccolallo L, Licitra L, Cantu G et al, 2009, Survival from salivary glands adenoid cystic carcinoma in European populations. Oral Oncol 45:669–674 Persson M, Andren Y,Mark J et al, 2009, Recurrent fusion ofMYB and NFIB transcription factor genes in carcinomas of the breast and head and neck. Proc Natl Acad Sci U S A 106:18740–18744 Marchio C, Weigelt B, Reis-Filho JS, 2010, Adenoid cystic carcinomas of the breast and salivary glands, or ‘The strange case of Dr Jekyll and Mr Hyde’ of exocrine gland carcinomas). J Clin Pathol 63:220–228 Lai EC, 2002)Micro RNAs are complementary to 3′ UTR sequence motifs that mediate negative post-transcriptional regulation. Nat Genet 30:363–364 Stark A, Brennecke J, Russell RB et al, 2003, Identification of Drosophila MicroRNA targets. PLoS Biol 1:E60 Vasudevan S, Tong Y, Steitz JA, 2007, Switching from repression to activation: microRNAs can up-regulate translation. Science 318: 1931–1934 Place RF, Li LC, Pookot D et al, 2008, MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci U S A 105:1608–1613 Alves Vidigal J and Ventura A, 2012, Embryonic stem cell miRNAs and their roles in development and disease. Semin Cancer Biol 22(5-6):428–36 Houbaviy HB, Murray MF, Sharp PA, 2003, Embryonic stem cellspecific MicroRNAs. Dev Cell 5:351–358 Poy MN, Spranger M, Stoffel M, 2007, microRNAs and the regulation of glucose and lipid metabolism. Diabetes Obes Metab 9(Suppl 2):67–73 Schroen B, Heymans S, 2012, Small but smart–microRNAs in the centre of inflammatory processes during cardiovascular diseases, the metabolic syndrome, and ageing. Cardiovasc Res 93:605–613 Slaby O, Svoboda M, Michalek J et al, 2009, MicroRNAs in colorectal cancer: translation of molecular biology into clinical application. Mol Cancer 8:102 Tseng CW, Lin CC, Chen CN et al, 2011, Integrative network analysis reveals active microRNAs and their functions in gastric cancer. BMC Syst Biol 5:99 Zhou SL, Wang LD, 2010, Circulating microRNAs: novel biomarkers for esophageal cancer. World J Gastroenterol 16:2348– 2354 Paranjape T, Slack FJ, Weidhaas JB, 2009, MicroRNAs: tools for cancer diagnostics. Gut 58:1546–1554 Yeung ML, Jeang KT, 2011, MicroRNAs and cancer therapeutics. Pharm Res 28:3043–3049 Xiao F, Zuo Z, Cai G et al, 2009, miRecords: an integrated resource for microRNA-target interactions. Nucleic Acids Res 37:D105–D110 Vergoulis T, Vlachos IS, Alexiou P et al, 2012, TarBase 6.0: capturing the exponential growth of miRNA targets with experimental support. Nucleic Acids Res 40:D222–D229 Lewis BP, Burge CB, Bartel DP, 2005, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120:15–20 Friedman RC, Farh KK, Burge CB et al, 2009, Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19:92–105 Grimson A, Farh KK, Johnston WK et al, 2007, MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell 27:91–105 Garcia DM, Baek D, Shin C et al, 2011)Weak seed-pairing stability and high target-site abundance decrease the proficiency of lsy-6 and other microRNAs. Nat Struct Mol Biol 18:1139–1146 Wang X, 2008, miRDB: a microRNA target prediction and functional annotation database with a wiki interface. RNA 14:1012–1017 Wang X, El Naqa IM, 2008, Prediction of both conserved and nonconserved microRNA targets in animals. Bioinformatics 24:325– 332 Vranic S, Bilalovic N, Lee LM et al, 2007, PIK3CA and PTEN mutations in adenoid cystic carcinoma of the breast metastatic to kidney. Hum Pathol 38:1425–1431 Vranic S, Frkovic-Grazio S, Lamovec J et al, 2010, Adenoid cystic carcinomas of the breast have low Topo IIalpha expression but frequently overexpress EGFR protein without EGFR gene amplification. Hum Pathol 41:1617–1623 Sung H, Jeon S, Lee KM et al, 2012, Common genetic polymorphisms of microRNA biogenesis pathway genes and breast cancer survival. BMC Cancer 12:195 Han J, Lee Y, Yeom KH et al, 2004, The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev 18:3016–3027 Noh H, Hong S, Dong Z et al, 2011, ImpairedMicroRNA processing facilitates breast cancer cell invasion by upregulating urokinasetype plasminogen activator expression. Genes Cancer 2:140–150 Zhang X, Cairns M, Rose B et al, 2009, Alterations in miRNA processing and expression in pleomorphic adenomas of the salivary gland. Int J Cancer 124:2855–2863 Falbo V, Floridia G, Censi F et al, 2011, Three cases of rare salivary gland tumours: a molecular study of TP53, CDKN2A/ARF, RAS, BRAF, PTEN, MAPK2 and EGFR genes. Oncol Rep 26:3–11 Gomes CC, Diniz MG, Orsine LA et al, 2012, Assessment of TP53 mutations in benign and malignant salivary gland neoplasms. PLoS One 7:e41261 Matizonkas-Antonio LF, de Mesquita RA, de Souza SC et al, 2005, TP53 mutations in salivary gland neoplasms. Braz Dent J 16:162–166 Kazakov DV,Grossmann P, SpagnoloDVet al, 2010, Expression of p53 and TP53mutational analysis inmalignant neoplasms arising in preexisting spiradenoma, cylindroma, and spiradenocylindroma, sporadic or associated with Brooke-Spiegler syndrome. Am J Dermatopathol 32:215–221 de Lima MD, Marques YM, Alves Sde M Jr et al, 2009, MDM2, P53, P21WAF1 and pAKT protein levels in genesis and behaviour of adenoid cystic carcinoma. Cancer Epidemiol 33:142–146 Martin JL, Baxter RC, 2007, Expression of insulin-like growth factor binding protein-2 by MCF-7 breast cancer cells is regulated through the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin pathway. Endocrinology 148:2532–2541 Lee AV, Jackson JG, Gooch JL et al, 1999, Enhancement of insulinlike growth factor signaling in human breast cancer: estrogen regulation of insulin receptor substrate-1 expression in vitro and in vivo. Mol Endocrinol 13:787–796 Zeng Z, Lin H, Zhao X et al, 2012, Overexpression of GOLPH3 promotes proliferation and tumorigenicity in breast cancer via suppression of the FOXO1 transcription factor. Clin Cancer Res 18: 4059–4069 Li J, Yang L, Song L et al, 2009, Astrocyte elevated gene-1 is a proliferation promoter in breast cancer via suppressing transcriptional factor FOXO1. Oncogene 28:3188–3196 Dote H, Toyooka S, Tsukuda K et al, 2004, Aberrant promoter methylation in human DAB2 interactive protein, hDAB2IP, gene in breast cancer. Clin Cancer Res 10:2082–2089 Chen H, Toyooka S, Gazdar AF et al, 2003, Epigenetic regulation of a novel tumor suppressor gene, hDAB2IP, in prostate cancer cell lines. J Biol Chem 278:3121–3130 Dote H, Toyooka S, Tsukuda K et al, 2005, Aberrant promoter methylation in human DAB2 interactive protein, hDAB2IP, gene in gastrointestinal tumour. Br J Cancer 92:1117–1125 Duan YF, Li DF, Liu YH et al, 2013, Decreased expression of DAB2IP in pancreatic cancer with wild-type KRAS. Hepatobiliary Pancreat Dis Int 12:204–209 Liu T, Li Y, Gu H et al, 2013, p21-Activated kinase 6, PAK6, inhibits prostate cancer growth via phosphorylation of androgen receptor and tumorigenic E3 ligase murine double minute-2, Mdm2). J Biol Chem 288:3359–3369 Minden A, 2012, PAK4-6 in cancer and neuronal development. Cell Logist 2:95–104 Lee SR, RamosSM,KoAet al, 2002, ARand ER interaction with a p21-activated kinase, PAK6). Mol Endocrinol 16:85–99 VozML,AstromAK, Kas K et al, 1998, The recurrent translocation t(5;8)(p13;q12, in pleomorphic adenomas results in upregulation of PLAG1 gene expression under control of the LIFR promoter. Oncogene 16:1409–1416 Hibbard MK, Kozakewich HP, Dal Cin P et al, 2000, PLAG1 fusion oncogenes in lipoblastoma. Cancer Res 60:4869–4872 Debiec-Rychter M, Van Valckenborgh I, Van den Broeck C et al, 2001, Histologic localization of PLAG1, pleomorphic adenoma gene 1, in pleomorphic adenoma of the salivary gland: cytogenetic evidence of common origin of phenotypically diverse cells. Lab Invest 81:1289–1297 Daa T, Nakamura I, Yada N et al, 2013, PLAG1 and CYLD do not play a role in the tumorigenesis of adenoid cystic carcinoma. Mol Med Rep 7:1086–1090 Gross S, Krause Y, Wuelling M et al, 2012, Hoxa11 and Hoxd11 regulate chondrocyte differentiation upstream of Runx2 and Shox2 in mice. PLoS One 7:e43553 Bouba I, Siomou E, Stefanidis CJ et al, 2009, Absence ofmutations in the HOXA11 and HOXD11 genes in children with congenital renal malformations. Pediatr Nephrol 24:1569–1572 Schwab K, Hartman HA, Liang HC et al, 2006, Comprehensive microarray analysis of Hoxa11/Hoxd11 mutant kidney development. Dev Biol 293:540–554 Turbendian HK, Gordillo M, Tsai SY et al, 2013, GATA factors efficiently direct cardiac fate from embryonic stem cells. Development 140:1639–1644 Terada T, 2012, Pigmented adenoid cystic carcinoma of the ear skin arising from the epidermis: a case report with immunohistochemical studies. Int J Clin Exp Pathol 5:254–259 El Ayoubi A, Poizat F, Garrel R et al, 2009, Sinonasal adenocarcinomas reviewed. Prognostic value of WHO 2005 histological classification. Ann Otolaryngol Chir Cervicofac 126:175–181 Takata T, Kudo Y, Zhao M et al, 1999, Reduced expression of p27(Kip1, protein in relation to salivary adenoid cystic carcinoma metastasis. Cancer 86:928–935 Shahsavari F, Eslami M, Baghaie F e t a l, 2005, Immunohistochemical evaluation of p27, kip1, in pleomorphic adenomas and adenoid cystic carcinomas of the minor salivary glands. Asian Pac J Cancer Prev 6:527–530 Daa T, Kashima K, Kondo Y et al, 2008, Aberrant methylation in promoter regions of cyclin-dependent kinase inhibitor genes in adenoid cystic carcinoma of the salivary gland. APMIS 116:21–26 Ivanov SV, Panaccione A, Brown B et al, 2012, TrkC signaling is activated in adenoid cystic carcinoma and requires NT-3 to stimulate invasive behavior. Oncogene 32(32):3698–710 Katsushima K, Shinjo K, Natsume A et al, 2012, Contribution of microRNA-1275 to Claudin11 suppression via a polycombmediated silencing mechanism in human glioma stem-like cells. J Biol Chem 287(33):27396–406 Hashiguchi Y, Nishida N, Mimori K et al, 2012, Down-regulation of miR-125a-3p in human gastric cancer and its clinicopathological significance. Int J Oncol 40:1477–1482 Jiang L, Huang Q, Zhang S et al, 2010, Hsa-miR-125a-3p and hsamiR- 125a-5p are downregulated in non-small cell lung cancer and have inverse effects on invasion and migration of lung cancer cells. BMC Cancer 10:318 Zhang X, Ladd A, Dragoescu E et al, 2009, MicroRNA-17-3p is a prostate tumor suppressor in vitro and in vivo, and is decreased in high grade prostate tumors analyzed by laser capture microdissection. Clin Exp Metastasis 26:965–979 Nohata N, Hanazawa T, Enokida H et al, 2012, microRNA-1/133a and microRNA-206/133b clusters: dysregulation and functional roles in human cancers. Oncotarget 3:9–21 Suzuki K, Cheng J, Watanabe Y, 2003, Hepatocyte growth factor and c-Met, HGF/c-Met, in adenoid cystic carcinoma of the human salivary gland. J Oral Pathol Med 32:84–89 Chen J, Shi Y, Li Z et al, 2011, A functional variant of IC53 correlates with the late onset of colorectal cancer. Mol Med 17: 607–618 Wetterskog D, Lopez-Garcia MA, Lambros MB et al, 2012, Adenoid cystic carcinomas constitute a genomically distinct subgroup of triplenegative and basal-like breast cancers. J Pathol 226:84–96 Vincent-Salomon A,Macgrogan G, Charaffe-Jauffret E et al, 2010, Identification of basal-like carcinomas in clinical practice: “triple zero/BRCA1-like” carcinomas. Bull Cancer 97:357–363 Ding L, Zhu S, Xie S et al, 2008, Effect of exogenous bFGF on the proliferation of human adenoid cystic carcinoma ACC-2 cells. J Huazhong Univ Sci Technol Med Sci 28:227–229 Hu K, Gan YH, Li SL et al, 2009, Relationship of activated extracellular signal-regulated kinase 1/2 with lung metastasis in salivary adenoid cystic carcinoma. Oncol Rep 21:137–143 Bell A, Bell D, Weber RS et al, 2011, CpG island methylation profiling in human salivary gland adenoid cystic carcinoma. Cancer 117:2898–2909 Tanzer A, Stadler PF, 2004, Molecular evolution of a microRNA cluster. J Mol Biol 339:327–335 Dews M, Homayouni A, Yu D et al, 2006, Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster. NatGenet 38:1060– 1065 Sareeboot T, Punyarit P, Petmitr S, 2011, DNA amplification on chromosome 13q31.1 correlated with poor prognosis in colorectal cancer. Clin Exp Med 11:97–103 Reichek JL, Duan F, Smith LM et al, 2011, Genomic and clinical analysis of amplification of the 13q31 chromosomal region in alveolar rhabdomyosarcoma: a report from the Children’s Oncology Group. Clin Cancer Res 17:1463–1473 Eiriksdottir G, Johannesdottir G, Ingvarsson S et al, 1998)Mapping loss of heterozygosity at chromosome 13q: loss at 13q12-q13 is associated with breast tumour progression and poor prognosis. Eur J Cancer 34:2076–2081 Lin YW, Sheu JC, Liu LY et al, 1999, Loss of heterozygosity at chromosome 13q in hepatocellular carcinoma: identification of three independent regions. Eur J Cancer 35:1730–1734 He L, Thomson JM, Hemann MT et al, 2005, A microRNA polycistron as a potential human oncogene. Nature 435:828–833 Tsuchida A, Ohno S,WuWet al, 2011, miR-92 is a key oncogenic component of the miR-17-92 cluster in colon cancer. Cancer Sci 102:2264–2271 Hayashita Y, Osada H, Tatematsu Y et al, 2005, A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res 65:9628–9632 Huang G, Nishimoto K, Zhou Z et al, 2012, miR-20a encoded by the miR-17-92 cluster increases the metastatic potential of osteosarcoma cells by regulating Fas expression. Cancer Res 72:908–916 Italiano A, Thomas R, BreenM et al, 2012, The miR-17-92 cluster and its target THBS1 are differentially expressed in angiosarcomas dependent on MYC amplification. Genes Chromosomes Cancer 51:569–578 Ota A, Tagawa H, Karnan S et al, 2004, Identification and characterization of a novel gene, C13orf25, as a target for 13q31-q32 amplification in malignant lymphoma. Cancer Res 64:3087–3095 Tagawa H, Seto M, 2005, A microRNA cluster as a target of genomic amplification in malignant lymphoma. Leukemia 19: 2013–2016 Gordon AT, Brinkschmidt C, Anderson J et al, 2000, A novel and consistent amplicon at 13q31 associated with alveolar rhabdomyosarcoma. Genes Chromosomes Cancer 28:220–226 Li H, Bian C, Liao L et al, 2011, miR-17-5p promotes human breast cancer cell migration and invasion through suppression of HBP1. Breast Cancer Res Treat 126:565–575 KangHW,Wang F,Wei Q et al, 2012, miR-20a promotesmigration and invasion by regulating TNKS2 in human cervical cancer cells. FEBS Lett 586:897–904 Doebele C, Bonauer A, Fischer A et al, 2010, Members of the microRNA-17-92 cluster exhibit a cell-intrinsic antiangiogenic function in endothelial cells. Blood 115:4944–4950 Gottardo F, Liu CG, Ferracin M et al, 2007, Micro-RNA profiling in kidney and bladder cancers. Urol Oncol 25:387–392 Volinia S, Calin GA, Liu CG et al, 2006, A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 103:2257–2261 O’Donnell KA,Wentzel EA, Zeller KI et al, 2005, c-Myc-regulated microRNAs modulate E2F1 expression. Nature 435:839–843 Vekony H, Raaphorst FM, Otte AP et al, 2008, High expression of Polycomb group protein EZH2 predicts poor survival in salivary gland adenoid cystic carcinoma. J Clin Pathol 61:744–749 Shirasaki T, Maruya S, Mizukami H et al, 2008, Effects of small interfering RNA targeting thymidylate synthase on survival of ACC3 cells from salivary adenoid cystic carcinoma. BMC Cancer 8:348 Sequeiros-Santiago G, Garcia-Carracedo D, FresnoMF et al, 2009, Oncogene amplification pattern in adenoid cystic carcinoma of the salivary glands. Oncol Rep 21:1215–1222 Hoffmann TK, Sonkoly E, Homey B et al, 2007, Aberrant cytokine expression in serum of patients with adenoid cystic carcinoma and squamous cell carcinoma of the head and neck. Head Neck 29:472– 478 Zhao X, Chao YL, Wan QB et al, 2011, Flavokawain B induces apoptosis of human oral adenoid cystic cancer ACC-2 cells via upregulation of Bim and down-regulation of Bcl-2 expression. Can J Physiol Pharmacol 89(12):875–83 Hao L, Xiao-lin N, Qi C et al, 2010, Nerve growth factor and vascular endothelial growth factor: retrospective analysis of 63 patients with salivary adenoid cystic carcinoma. Int J Oral Sci 2: 35–44 Zhang J, Peng B, 2007, In vitro angiogenesis and expression of nuclear factor kappaB and VEGF in high and low metastasis cell lines of salivary gland Adenoid Cystic Carcinoma. BMC Cancer 7: 95 Bernheim A, Toujani S, Saulnier P et al, 2008, Highresolution array comparative genomic hybridization analysis of human bronchial and salivary adenoid cystic carcinoma. Lab Invest 88:464–473 Porkka KP, Pfeiffer MJ, Waltering KK et al, 2007, MicroRNA expression profiling in prostate cancer. Cancer Res 67:6130–6135 Fuse M, Kojima S, Enokida H et al, 2012, Tumor suppressive microRNAs, miR-222 and miR-31, regulate molecular pathways based on microRNA expression signature in prostate cancer. J Hum Genet 57:691–699 Ishteiwy RA, Ward TM, Dykxhoorn DM et al, 2012, The microRNA -23b/-27b cluster suppresses the metastatic phenotype of castration-resistant prostate cancer cells. PLoS One 7:e52106 Hu H, Li S, Liu J et al, 2012, MicroRNA-193b modulates proliferation, migration, and invasion of non-small cell lung cancer cells. Acta Biochim Biophys Sin, Shanghai, 44:424–430 Chen J, Abi-DaoudM,WangA et al, 2012, Stathmin 1 is a potential novel oncogene in melanoma. Oncogene 32(10):1330–7 Rauhala HE, Jalava SE, Isotalo J et al, 2010, miR-193b is an epigenetically regulated putative tumor suppressor in prostate cancer. Int J Cancer 127:1363–1372 Desmet CJ, Gallenne T, Prieur A et al, 2013, Identification of a pharmacologically tractable Fra-1/ADORA2B axis promoting breast cancer metastasis. Proc Natl Acad Sci U S A 110:5139–5144 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 29 EP 44 DI 10.1007/s12253-014-9770-1 PG 16 ER PT J AU Zhang, G Liu, X Huang, W Li, X Johnstone, M Deng, Y Ke, Y Nunes, MQ Wang, H Wang, Y Zhang, X AF Zhang, Guanjun Liu, Xi Huang, Wei Li, Xiaofeng Johnstone, Marianne Deng, Yuan Ke, Yongqiang Nunes, M Quentin Wang, Hongyan Wang, Yili Zhang, Xuebin TI Carcinoma Showing Thymus-Like Elements of the Thyroid Gland: Report of Three Cases Including One Case with Breast Cancer History SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Carcinoma showing thymus-like elements; Breast cancer; Second primary tumor; Solid cell nests; Differential diagnosis; Histogenesis ID Carcinoma showing thymus-like elements; Breast cancer; Second primary tumor; Solid cell nests; Differential diagnosis; Histogenesis AB Carcinoma showing thymus-like elements (CASTLE) is a raremalignant tumor of the thyroid or adjacent neck soft tissues, whose histogenesis is still debated. It may resemble other primary or metastatic poorly differentiated tumors histologically and the differential diagnosis is crucial for CASTLE has a better prognosis. However, CASTLE as a second primary tumor has not been reported in the literature. We report three cases of thyroid CASTLE, including a unique tumor following breast-conserving surgery for early-stage breast invasive carcinoma. There were two female and one male. All three tumors were located in the right lobe of the thyroid, and one tumor showed extension into the surrounding soft tissue. Histologically, all tumors showed expansive growth and consisted of cords, nests or sheets of epithelial cells divided into irregularly shaped lobules by fibrous connective tissue with lymphoplasmacytic infiltration. Focal squamous differentiation resembling Hassall’s corpuscles were observed. All cases stained positively for CD5, CD117, high molecular weight cytokeratin, cytokeratin, P63, carcinoembryonic antigen and epithelial membrane antigen. Positive staining for Bcl-2 in two cases and chromogranin A in one case was noted. Ki-67 expression ranged from 15 to 25 %. Thyroid transcription factor and CD3 were negative. There was no evidence of recurrent or metastatic disease at following surgery. These features demonstrated CASTLE may arise from branchial pouch remnants, the thyroid solid cell nests. CASTLE is a rare entity, awareness of its occurrence as a second primary tumor is important to avoid overtreatment because it is associated with a favorable prognosis. C1 [Zhang, Guanjun] Medical College of Xian Jiaotong University, First Affiliated Hospital, Department of Pathology, 710061 Xi’an, China. [Liu, Xi] Medical College of Xian Jiaotong University, First Affiliated Hospital, Department of Pathology, 710061 Xi’an, China. [Huang, Wei] Royal Liverpool University Hospital, Division of Surgery and OncologyLiverpool, UK. [Li, Xiaofeng] Medical College of Xian Jiaotong University, First Affiliated Hospital, Department of Pathology, 710061 Xi’an, China. [Johnstone, Marianne] Royal Liverpool University Hospital, Division of Surgery and OncologyLiverpool, UK. [Deng, Yuan] Medical College of Xian Jiaotong University, First Affiliated Hospital, Department of Pathology, 710061 Xi’an, China. [Ke, Yongqiang] Royal Liverpool University Hospital, Division of Surgery and OncologyLiverpool, UK. [Nunes, M Quentin] Royal Liverpool University Hospital, Division of Surgery and OncologyLiverpool, UK. [Wang, Hongyan] Medical College of Xian Jiaotong University, First Affiliated Hospital, Department of Pathology, 710061 Xi’an, China. [Wang, Yili] Medical College of Xian Jiaotong University, First Affiliated Hospital, Department of Pathology, 710061 Xi’an, China. [Zhang, Xuebin] Medical College of Xian Jiaotong University, First Affiliated Hospital, Department of Pathology, 710061 Xi’an, China. RP Liu, X (reprint author), Medical College of Xian Jiaotong University, First Affiliated Hospital, Department of Pathology, 710061 Xi’an, China. EM liuxizgq@gmail.com CR Miyauchi A, Kuma K, Matsuzuka F, Matsubayashi S, Kobayashi A, Tamai H, Katayama S, 1985, Intrathyroidal Epithelial Thymoma: An Entity Distinct from Squamous Cell Carcinoma of the Thyroid. World J Surg 9:128–135 Chan JK, Rosai J, 1991, Tumors of the Neck Showing Thymic or Related Branchial Pouch Differentiation: A Unifying Concept. Hum Pathol 22:349–367 Ito Y, Miyauchi A, Nakamura Y, Miya A, Kobayashi K, Kakudo K, 2007, Clinicopathologic Significance of Intrathyroidal Epithelial Thymoma/Carcinoma Showing Thymus-Like Differentiation: A Collaborative Study with Member Institutes of The Japanese Society of Thyroid Surgery. Am J Clin Pathol 127:230–236 Reimann JD, Dorfman DM, Nose V, 2006, Carcinoma Showing Thymus-Like Differentiation of the Thyroid, CASTLE): a Comparative Study: Evidence of Thymic Differentiation and Solid Cell Nest Origin. Am J Surg Pathol 30:994–1001 Preto A, Cameselle-Teijeiro J, Moldes-Boullosa J, Soares P, Cameselle-Teijeiro JF, Silva P, Reis-Filho JS, Reyes-Santias RM, Alfonsin-Barreiro N, Forteza J, Sobrinho-Simoes M, 2004, Telomerase Expression and Proliferative Activity Suggest a Stem Cell Role for Thyroid Solid Cell Nests. Mod Pathol 17:819–826 Reis-Filho JS, Preto A, Soares P, Ricardo S, Cameselle-Teijeiro J, Sobrinho-SimoesM(2003, P63 Expression in Solid Cell Nests of the Thyroid: Further Evidence for a Stem Cell Origin. Mod Pathol 16: 43–48 Pusztaszeri M, Yerly S, Lobrinus JA, Bongiovanni M, Becker M, Zare M, Granger P, 2013, Carcinoma Showing Thymus-Like Elements of the Thyroid, CASTLE, Arising in Close Association with Solid Cell Nests. Evidence for a Precursor Lesion? Thyroid 23: 511–516 Kakudo K, Mori I, Tamaoki N, Watanabe K, 1988, Carcinoma of Possible Thymic Origin Presenting as a Thyroid Mass: A new Subgroup of Squamous Cell Carcinoma of the Thyroid. J Surg Oncol 38:187–192 AhujaAT, Chan ES, AllenPW, LauKY, KingW,Metreweli C, 1998, Carcinoma Showing Thymiclike Differentiation, CASTLE Tumor). AJNR Am J Neuroradiol 19:1225–1228 Luo CM, Hsueh C, Chen TM, 2005, Extrathyroid Carcinoma Showing Thymus-Like Differentiation, CASTLE, Tumor–A New Case Report and Review of Literature. Head Neck 27:927–933 Kusada N, Hara Y, Kobayashi S,Weihua T, Nakamura Y, Kakudo K, Yuasa H, 2005, A Case of Aggressive Carcinoma Showing Thymus- Like Differentiation with Distant Metastases. Thyroid 15:1383–1388 Yamazaki M, Fujii S, Daiko H, Hayashi R, Ochiai A, 2008, Carcinoma Showing Thymus-Like Differentiation, CASTLE, with Neuroendocrine Differentiation. Pathol Int 58:775–779 Cappelli C, Tironi A,Marchetti GP, Pirola I, DeMartino E, Delbarba A, Castellano M, Rosei EA, 2008, Aggressive Thyroid Carcinoma Showing Thymic-Like Differentiation, CASTLE): Case Report and Review of the Literature. Endocr J 55:685–690 Youens KE, Bean SM, Dodd LG, Jones CK, 2011, Thyroid Carcinoma Showing Thymus-Like Differentiation, CASTLE): Case Report with Cytomorphology and Review of the Literature. Diagn Cytopathol 39:204–209 Sun T, Wang Z, Wang J, Wu Y, Li D, Ying H, 2011, Outcome of Radical Resection and Postoperative Radiotherapy for Thyroid Carcinoma Showing Thymus-Like Differentiation. World J Surg 35:1840–1846 Pan Y, Zhao X, Yang J, Deng J, Zhan Z, Luo Y, Li Q, Guo Y, Zhai Q, Sun B, 2012)Absence ofGeneMutations inKIT-Positive Carcinoma Showing Thymus-Like Elements of the Thyroid. Hum Pathol 43: 350–355 Chang S, Joo M, Kim H, 2012, Cytologic Findings of Thyroid Carcinoma Showing Thymus-like Differentiation: A Case Report. Korean J Pathol 46:302–305 Hirokawa M, Kuma S, Miyauchi A, 2012, Cytological Findings of Intrathyroidal Epithelial Thymoma/Carcinoma Showing Thymus- Like Differentiation: A Study of Eight Cases. Diagn Cytopathol 40(Suppl 1):E16–20 Veits L, Mechtersheimer G, Steger C, Freitag J, Mikuz G, Schmid KW, Hofmann W, Schirmacher P, Hartmann A, Rieker RJ, 2011, Chromosomal Imbalances in Carcinoma Showing Thymus-Like Elements, CASTLE). Virchows Arch 459:221–226 Kakudo K, Bai Y, Ozaki T, Homma K, Ito Y, Miyauchi A, 2013, Intrathyroid Epithelial Thymoma, ITET, and Carcinoma Showing Thymus-Like Differentiation, CASTLE): CD5-Positive Neoplasms Mimicking Squamous Cell Carcinoma of the Thyroid. Histol Histopathol 28:543–556 HirokawaM,Miyauchi A,Minato H, Yokoyama S, Kuma S, Kojima M, 2013, Intrathyroidal Epithelial Thymoma/Carcinoma Showing Thymus-Like Differentiation; Comparison with Thymic Lymphoepithelioma-Like Carcinoma and a Possibility of Development from aMultipotential Stem Cell. APMIS 121:523–530 Liu Z, Teng XY, Sun DX, Xu WX, Sun SL, 2013, Clinical Analysis of Thyroid Carcinoma Showing Thymus-Like Differentiation: Report of 8 Cases. Int Surg 98:95–100 Tsutsui H, Hoshi M, Kubota M, Suzuki A, Nakamura N, Usuda J, Shibuya H,Miyajima K, Ohira T, Ito K, Ikeda N, 2013)Management of Thyroid Carcinoma Showing Thymus-Like Differentiation, CASTLE, Invading the Trachea. Surg Today., DOI 10.1007/s00595- 013-0560-2 Cheuk W, Chan JKC, Dorfman DM, Giordano T, 2004, Carcinoma Showing Thymus-Like Differentiation. In: DeLellis RA, Lloyd RV, Heitz PU, Eng C, eds, World Health Organization Classification of Tumours. Pathology and Genetics of Tumours of Endocrine Organs, 3rd edn. IARC Press, Lyon, pp 96–97 Torres-Cabala CA,WangWL, Trent J, Yang D, Chen S, Galbincea J, Kim KB, Woodman S, Davies M, Plaza JA, Nash JW, Prieto VG, Lazar AJ, Ivan D, 2009, Correlation Between KIT Expression and KITMutation inMelanoma: A Study of 173 Cases with Emphasis on the Acral-Lentiginous/Mucosal Type. Mod Pathol 22:1446–1456 Lakhani SR, Ellis IO, Schnitt SJ, Tan PH, van de Vijver MJ, 2012, WHO Classification of Tumours of the Breast, 4th edn. IARC Press, Lyon Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, 2010, AJCC Cancer Staging Manual, 7th edn. Springer, New York Chan JKC, 2007, Tumors of the Thyroid and Parathyroid Glands. In: Fletcher CDM, ed, Diagnostic Histopathology of Tumors, 3rd edn. Churchill Livingstone, Philadelphia, pp 1048–1049 Dorfman DM, Shahsafaei A, Miyauchi A, 1998, Intrathyroidal Epithelial Thymoma, ITET)/Carcinoma Showing Thymus-Like Differentiation, CASTLE, Exhibits CD5 Immunoreactivity: new Evidence for Thymic Differentiation. Histopathology 32:104–109 Kornstein MJ, Rosai J, 1998, CD5 Labeling of Thymic Carcinomas and Other Nonlymphoid Neoplasms. Am J Clin Pathol 109:722–726 Lemkjaer L, Friis S, Olsen JH, Scelo G, Hemminki K, Tracey E, Andersen A, Brewster DH, Pukkala E, McBride ML, Kliewer EV, Tonita JM, Kee-Seng C, Pompe-Kirn V, Martos C, Jonasson JG, Boffetta P, Brennan P, 2006, Risk of Second Cancer Among Women with Breast Cancer. Int J Cancer 118:2285–2292 Rubino C, de Vathaire F, Diallo I, Shamsaldin A, Le MG, 2000, Increased Risk of Second Cancers Following Breast Cancer: Role of the Initial Treatment. Breast Cancer Res Treat 61:183–195 Langballe R, Olsen JH, AnderssonM,Mellemkjaer L, 2011, Risk for Second Primary non-Breast Cancer in pre- and Postmenopausal Women with Breast Cancer not Treated with Chemotherapy, Radiotherapy or Endocrine Therapy. Eur J Cancer 47:946–952 Li CI, Rossing MA, Voigt LF, Daling JR, 2000, Multiple Primary Breast and Thyroid Cancers: Role of age at Diagnosis and Cancer Treatments, United States). Cancer Causes Control 11:805–811 Mamounas EP, Perez-Mesa C, Penetrante RB, Driscoll DL, Blumenson LE, Tsangaris TN, 1993, Patterns of Occurrence of Second Primary non-Mammary Malignancies in Breast Cancer Patients: Results from 1382 Consecutive Autopsies. Surg Oncol 2: 175–185 Smyth PP, 1997, The Thyroid and Breast Cancer: A Significant Association? Ann Med 29:189–191 Cameselle-Teijeiro J, Febles-Perez C, Sobrinho-Simoes M, 1995, Papillary and Mucoepidermoid Carcinoma of the Thyroid with Anaplastic Transformation: A Case Report with Histologic and Immunohistochemical Findings That Support a Provocative Histogenetic Hypothesis. Pathol Res Pract 191:1214–1221 Rios Moreno MJ, Galera-Ruiz H, De Miguel M, Lopez MI, Illanes M, Galera-Davidson H, 2011, Inmunohistochemical Profile of Solid Cell Nest of Thyroid Gland. Endocr Pathol 22:35–39 Cameselle-Teijeiro J, Abdulkader I, Perez-Becerra R, Vazquez- Boquete A, Alberte-Lista L, Ruiz-Ponte C, Forteza J, Sobrinho- Simoes M, 2009, BRAF Mutation in Solid Cell Nest Hyperplasia Associated with Papillary Thyroid Carcinoma. A Precursor Lesion? Hum Pathol 40:1029–1035 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 45 EP 51 DI 10.1007/s12253-014-9772-z PG 7 ER PT J AU Ottoffy, G Szigeti, E Bartyik, K Nyari, Cs Parker, L McNally, JQR Nyari, AT AF Ottoffy, Gabor Szigeti, Erika Bartyik, Katalin Nyari, Csaba Parker, Louise McNally, J Q Richard Nyari, Andras Tibor TI Investigating the Relationship between Mortality from Respiratory Diseases and Childhood Acute Lymphoblastic Leukaemia in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Childhood acute lymphoblastic leukaemia; Gender-specific difference; Deaths from respiratory infections; Poisson regression ID Childhood acute lymphoblastic leukaemia; Gender-specific difference; Deaths from respiratory infections; Poisson regression AB Our aim was to investigate the ecological association between death from infectious disease of the respiratory system and the risk of acute lymphoid leukaemia (ALL) in children aged less than 7 years. Poisson regression analyses were carried out using overall data and gender-specific models. The study included 176 cases (92(52.3 %) boys and 84 (47.7 %) girls) of ALL in those aged 0–6 years in South Hungary. Eight cases were diagnosed before the age of 1 year. A significant risk of ALL disease was observed with higher levels of mortality from the chronic respiratory diseases (p=0.035) and pneumonia (p=0.010) among children aged 2–5 years (Odds Ratio for trend was 1.001 and 95%CI [1.000–1.002] and Odds ratio for trend was 1.013 and 95%CI [1.003–1.023], respectively). Significantly increased risk of childhood ALL was detected among children under 1 year of age residing in areas around birth with higher levels of mortality from influenza (Odds Ratio (OR) for trend was 1.05; 95%CI [1.01–1.09]; p=0.012). This risk was also detected in girls (p<0.001), but not in boys (p=0.43). Our findings provide new evidence that will help to understand the different pattern of female and male childhood ALL occurrence , but further studies are needed using detailed individual medical history to clarify the role of influenza and other infectious diseases in the etiology of childhood ALL and to explain gender-specific effects. C1 [Ottoffy, Gabor] University of Pecs, Department of PediatricsPecs, Hungary. [Szigeti, Erika] University of Szeged, Department of Medical Informatics, H-6701 Szeged, Hungary. [Bartyik, Katalin] University of Szeged, Department of PediatricsSzeged, Hungary. [Nyari, Csaba] Saint Stephen University, Faculty of Economics, Agriculture and Health StudiesBekescsaba, Hungary. [Parker, Louise] Dalhousie University, IWK Health Centre, Department of PediatricsHalifax, Canada. [McNally, J Q Richard] Newcastle University, Institute of Health and SocietyNewcastle upon Tyne, UK. [Nyari, Andras Tibor] University of Szeged, Department of Medical Informatics, H-6701 Szeged, Hungary. RP Nyari, AT (reprint author), University of Szeged, Department of Medical Informatics, H-6701 Szeged, Hungary. EM nyari.tibor@med.u-szeged.hu CR Kinlen LJ, 1995, Epidemiological evidence for an infective basis in childhood leukaemia. Br J Cancer 71:1–5 McNally RJQ, Eden TOB, 2004, An infectious aetiology for childhood acute leukaemia: a review of the evidence. Br J Haematol 127: 243–263 Hakulinen T, Hovi L, Karkinen-Jaaskelainen, Penttinen K, Saxen L, 1973, Association between influenza during pregnancy and childhood leukaemia. Br Med J 4(5887):265–267 Bithell JF, Draper GJ, Gorbach PD, 1973, Association between malignant disease in children and maternal virus infections. Br Med J 1(5855):706–708 Randolph VL, Heath CW Jr, 1974, Influenza during pregnancy in relation to subsequent childhood leukemia and lymphoma. Am J Epidemiol 100(5):399–409 Curnen MG, Varma AA, Christine BW, Turgeon LR, 1974, Childhood leukemia and maternal infectious diseases during pregnancy. J Natl Cancer Inst 53(4):943–947 Kwan ML, Metayer C, Crouse V, Buffler PA, 2007, Maternal illness and drug/medication use during the period surrounding pregnancy and risk of childhood leukemia among offspring. Am J Epidemiol 165(1):27–35 Parodi S, Santi I,Marani E et al, 2012, Infectious diseases and risk of leukemia and non-Hodgkin’s lymphoma: a case–control study. Leuk Res 36:1354–1358 Nyari TA, Dickinson HO, Parker L, 2003, Childhood cancer in relation to infections in the community during pregnancy and around the time of birth. Int J Cancer 104(6):772–777 The Hungarian Central Statistical Office. Demographic Yearbook, 1981 – 2001. Budapest, KSH Nyari TA, Kajtar P, Bartyik K, Thurzo L, McNally R, Parker L, 2008, Seasonal variation of childhood acute lymphoblastic leukaemia is different between girls and boys. Pathol Oncol Res 14: 423–428 Nyari TA, Kajtar P, Bartyik K, Thurzo L, Parker L, 2006, Childhood acute lymphoblastic leukaemia in relation to population mixing around the time of birth in South Hungary. Pediatr Blood Cancer 47: 944–948 Nyari TA, Ottoffy G, Bartyik K et al, 2013, Spatial clustering of childhood acute lymphoblastic leukaemia in Hungary. Pathol Oncol Res 19:297–302 Cotterill SJ, Parker L, Malcolm AJ, Reid M, More L, Craft AW, 2000, Incidence and survival for cancer in children and young adults in the North of England, 1968–1995: a report from the Northern Region Young Persons’ Malignant Disease Registry. Br J Cancer 83: 397–403 Dickinson HO, Parker L, 2002, Leukaemia and non-Hodgkin’s lymphoma in children of male Sellafield radiation workers. Int J Cancer 99:437–444 Feltbower RG, Pearce MS, Dickinson HO, Parker L, McKinney PA, 2001, Seasonality of birth for cancer in Northern England, UK. Paediatr Perinat Epidemiol 15(4):338–345 Kroll ME, Draper GJ, Stiller CA, Murphy MF, 2006, Childhood leukemia incidence in Britain, 1974–2000: time trends and possible relation to influenza epidemics. J Natl Cancer Inst 98:417–420 Shore RE, Pasternack BS, Curnen MG, 1976, Relating influenza epidemics to childhood leukemia in tumor registries without a defined population base: a critique with suggestions for improved methods. Am J Epidemiol 103(6):527–535 Dockerty JD, Skegg DC, Elwood JM, Herbison GP, Becroft DM, Lewis ME, 1999, Infections, vaccinations, and the risk of childhood leukaemia. Br J Cancer 80:1483–1489 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 53 EP 57 DI 10.1007/s12253-014-9786-6 PG 5 ER PT J AU Girardi, MF Barra, BM Zettler, GC AF Girardi, Muradas Fabio Barra, Bizarro Marinez Zettler, Galleano Claudio TI Predictive Factors for Lymph Node Metastasis in Solitary Papillary Thyroid Carcinomas: A Retrospective Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thyroid gland; Thyroid neoplasms; Carcinoma; Papillary; Lymphatic metastasis ID Thyroid gland; Thyroid neoplasms; Carcinoma; Papillary; Lymphatic metastasis AB Identifying risk factors for neck lymph node metastasis (LNM) in papillary thyroid carcinoma (PTC) is important for patient prognosis establishment.We conducted a retrospective study among 317 patients with solitary PTC. Factors associated with clinically evident LNM were evaluated. LNM were identified in 69 (21.7%) patients. Central compartment (17.3%) and lateral compartment (9.4 %) were involved. Thyroid capsule invasion and extrathyroidal extension were found to be independent risk factors for both central and lateral compartment metastasis in multivariate analysis. Larger diameter was associated with central compartment metastasis in logistic regression model, whereas male gender only with lateral compartment metastasis. As closer tumors were positioned in relation to glandular capsule we expected rising rates of nodal spread. It was unlikely to find clinically evident neck LNM among patients with centrally located tumors. C1 [Girardi, Muradas Fabio] Santa Casa Hospital Complex, Santa Rita Hospital, Department of Head and Neck SurgeryPorto Alegre, RS, Brazil. [Barra, Bizarro Marinez] Santa Casa Hospital Complex, Santa Rita Hospital, Department of PathologyPorto Alegre, RS, Brazil. [Zettler, Galleano Claudio] Santa Casa Hospital Complex, Santa Rita Hospital, Department of PathologyPorto Alegre, RS, Brazil. RP Girardi, MF (reprint author), Santa Casa Hospital Complex, Santa Rita Hospital, Department of Head and Neck Surgery, Porto Alegre, Brazil. EM fabiomgirardi@gmail.com CR Sherman SI, 2003, Thyroid carcinoma. Lancet 361(9356):501–11 Davies L,Welch HG, 2006, Increasing incidence of thyroid cancer in the United States, 1973-2002. JAMA 295(18):2164–7 Baek SK, Jung KY, Kang SM, Kwon SY, Woo JS, Cho SH et al, 2010, Clinical risk factors associated with cervical lymph node recurrence in papillary thyroid carcinoma. Thyroid 20(2):147–52 Beasley NJ, Lee J, Eski S, Walfsh P, Witterick I, 2002, Impact of nodal metastases on prognosis in patients with well-differentiated thyroid cancer. Arch Otolaryngol Head Neck Surg 128(7):825–8 So YK, Son YI, Hong SD, SeoMY, Baek CH, Jeong HS et al, 2010, Subclinical lymph node metastasis in papillary thyroid microcarcinoma: a study of 551 resections. Surgery 148(3):526–31 Sugitani I, Toda K, Yamada K, Yamamoto N, Ikenaga M, Fugimoto Y, 2010, Three distinctly different kinds of papillary thyroid microcarcinoma should be recognized: Our treatment strategies and outcomes. World J Surg 34(6):1222–31 Roti E, degli Uberti EC, Bondanelli M, Braverman LE, 2008, Thyroid papillary microcarcinoma: a descriptive and meta-analysis study. Eur J Endocrinol 159(6):659–73 Ito Y, Tomoda C, Uruno T, Takamura Y, Miya A, Kobayashi K et al, 2004, Papillary microcarcinoma of the thyroid: how should it be treated? World J Surg 28(11):1115–21 Ito Y, Tomoda C, Uruno T, Takamura Y, Miya A, Kobayashi K et al, 2004, Preoperative ultrasonographic examination for lymph node metastasis: usefulness when designing lymph node dissection for papillary microcarcinoma of the thyroid. World J Surg 28(5):498–501 Wada N, Duh QY, Sugino K, Iwasaki H, Kameyama K, Mimura T et al, 2003, Lymph node metastasis from 259 papillary thyroid Microcarcinomas: Frequency, Pattern of Occurrence and Recurrence, andOptimal Strategy forNeckDissection. Ann Surg 237(3):399–407 Hughes CJ, Shaha AR, Shah JP, Loree TR, 1996, Impact of lymph nodemetastasis in differentiated carcinoma of the thyroid: amatchedpair analysis. Head Neck 18(2):127–32 Sarvanan K, Bapuraj JR, Sharma SC, Radotra BD, Khandelwal N, Suri S, 2002, Computed tomography and ultrasonographic evaluation ofmetastatic cervical lymph nodes with surgicoclinicopathologic correlation. J Laryngol Otol 116(3):194–9 Lee DW, Ji YB, Sung ES, Park JS, Lee YJ, Park DW et al, 2013, Roles of ultrasonography and computed tomography in the surgical management of cervical lymph node metastases in papillary thyroid carcinoma. Eur J Surg Oncol 39(2):191–6 Jeong HS, Baek CH, Son YI, Choi JY, Kim HJ, Ko YH et al, 2006, Integrated 18 F-FDG PET/CT for the initial evaluation of cervical node level of patients with papillary thyroid carcinoma: comparison with ultrasound and contrast-enhanced CT. Clin Endocrinol, Oxf, 65(3):402–7 Henry JF, Gramatica L, Denizot A, Kvachenyuk A, Puccini M, Defechereux T, 1998, Morbidity of prophylactic lymph node dissection in the central neck area in patients with papillary thyroid carcinoma. Langenbecks Arch Surg 383(2):167–9 Pereira JA, Jimeno J,Miquel J, IglesiasM,Munne A, Sancho JJ et al, 2005, Nodal yield, morbidity, and recurrence after central neck dissection for papillary thyroid carcinoma. Surgery 138(6):1095–100 Edge SE, Byrd DR, Carducci MA, Compton CA, 2010, AJCC cancer staging manual. Springer, New York Shah JP, 1990, Cervical lymph node metastases—diagnostic, therapeutic, and prognostic implications.Oncology, Williston Park, 4(10): 61–9 CooperDS, Doherty GM, Haugen BR, Kloos RT, Lee SL,Mandel SJ et al, 2009, Revised american thyroid association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid 19(11):1167–214 Cho JK, Kim JY, Jeong CY, Jung EJ, Park ST, Jeong SH et al, 2012, Clinical features and prognostic factors in papillary thyroid microcarcinoma depends on age. J Korean Surg Soc 82(5):281–7 Choi SJ, Kim TY, Lee JC, Shong YK, Cho KJ, Ryu JS et al, 2008, Is routine central neck dissection necessary for the treatment of papillary thyroid microcarcinoma? Clin Exp Otorhinolayngol 1(1):41–5 Zuniga S, Sanabria A, 2009, Prophylactic central neck dissection in stage N0 papillary thyroid carcinoma. Arch Otolaryngol Head Neck Surg 135(11):1087–91 Nam IC, Park JO, Joo YH, Cho KJ, Kim MS, 2013, Pattern and predictive factors of regional lymph node metastasis in papillary thyroid carcinoma: A prospective study. Head Neck 35(1):40–45 Jeong JJ, Lee YS, Lee SC, Kang SW, Chung WY, Chang HS et al, 2011, A scoring system for prediction of lateral neck node metastasis from papillary thyroid cancer. J Korean Med Sci 26(8): 996–1000 Gulben K, Berberoglu U, Celen O, Mersin HH, 2008, Incidental papillary microcarcinoma of the thyroid - factors affecting lymph node metastasis. Langenbecks Arch Surg 393(1):25–9 Choi Y, Park KJ, Ryu S, Kim DH, Yun J, Kang DK et al, 2012, Papillary thyroid carcinoma involving cervical neck lymph nodes: correlations with lymphangiogenesis and ultrasound features. Endocr J 59(10):941–8 Hunt JP, Buchmann LO, Wang L, Abraham D, 2011, An analysis of factors predicting lateral cervical nodal metastases in papillary carcinoma of the thyroid. Arch Otolaryngol Head Neck Surg 137(11): 1141–5 Zhang L, Wei WJ, Ji QH, Zhu YX, Wang ZY, Wang Y et al, 2012, Risk factors for neck nodal metastasis in papillary thyroid microcarcinoma: a study of 1066 patients. J Clin Endocrinol Metab 97(4):1250–7 Mirallie E, Sagan C, Hamy A, Paineau J, Kahn X, Le Neel JC et al, 1999, Predictive factors for node involvement in papillary thyroid carcinoma. Univariate and multivariate analyses. Eur J Cancer 35(3): 420–3 Ortiz S, Rodriguez JM, Soria T, Perez-Flores D, Pinero A, Moreno J et al, 2001, Extrathyroid spread in papillary carcinoma of the thyroid: clinicopathological and prognostic study. Otolaryngol Head Neck Surg 124(3):261–5 Niemeier LA, Kuffner Akatsu H, Song C, Carty SE, Hodak SP, Yip L et al, 2012, A combined molecular-pathologic score improves risk stratification of thyroid papillary microcarcinoma. Cancer 118(8): 2069–77 Kim SS, Lee BJ, Lee JC, Kim SJ, Lee SH, Jeon YK et al, 2011, Preoperative ultrasonographic tumor characteristics as a predictive factor of tumor stage in papillary thyroid carcinoma. Head Neck 33(12):1719–26 Foschini MP, Papotti M, Parmeggiani A, Tallini G, Castaldini L, Meringolo D et al, 2004, Three-dimensional reconstruction of vessel distribution in benign and malignant lesions of thyroid. Virchows Arch 445(2):189–98 Pissas A, Papamiltiades M, 1981, Our experience on anatomical injections of the lymphatic vessels. Lymphology 14(4): 145–8 Ito Y, Tomoda C, Uruno T, Takamura Y, Miya A, Kobayashi K et al, 2006, Minimal extrathyroid extension does Not affect the relapsefree survival of patients with papillary thyroid carcinoma measuring 4 cm or less over the Age of 45 years. Surg Today 36(1):12–8 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 59 EP 64 DI 10.1007/s12253-014-9788-4 PG 6 ER PT J AU Alsaegh, AM Miyashita, H Zhu, RSh AF Alsaegh, Amjed Mohammed Miyashita, Hitoshi Zhu, Rong Sheng TI Expression of Human Papillomavirus is Correlated with Ki-67 and COX-2 Expressions in Keratocystic Odontogenic Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human papillomavirus (HPV); Keratocyst; Odontogenic; Ki-67; Cyclooxygenase-2 (COX-2) ID Human papillomavirus (HPV); Keratocyst; Odontogenic; Ki-67; Cyclooxygenase-2 (COX-2) AB The aim of the current study was to investigate the presence of human papillomavirus (HPV) and evaluate its association with Ki-67 and cyclooxygenase-2 (COX-2) expressions in keratocystic odontogenic tumor (KCOT).Nineteen cases were included in the present study. Conventional PCR method and immunohistochemical analysis were performed for the detection of HPV-DNA and HPV-L1 capsid protein. Moreover, the expressions of Ki-67 and COX-2 proteins were analyzed immunohistochemically. HPV-DNA was detected in 36.8 % (7/19) of tumor samples, whilst HPV-L1 protein was identified in 68.4 % (13/19) of them. The Kappa coefficient statistical test showed a moderate agreement (κ0.424) between PCR and IHC assays for HPV detection. Expression of HPV-DNA was positively correlated with Ki-67 and COX-2 expressions (p<0.05), whereas HPV-L1 positive staining was positively correlated with COX-2 (p<0.05) and highly associated with those of Ki-67 (p< 0.01). There was no significant correlation between the presence of HPV and the recurrence of the studied lesions. The results of the current study showed that active HPV infection was present in the odontogenic epithelium of KCOT, and it was associated with increased proliferation rate and COX-2 expression. These findings suggest that HPV may have a role in the pathogenesis and aggressiveness of KCOT. Based on these conclusions, we recommend further investigations of HPV vaccine or antiviral therapy and COX-2 inhibitors as nonsurgical options in the prevention and management of KCOT. C1 [Alsaegh, Amjed Mohammed] Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Department of Stomatology, 430030 Wuhan, China. [Miyashita, Hitoshi] Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Department of Stomatology, 430030 Wuhan, China. [Zhu, Rong Sheng] Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Department of Stomatology, 430030 Wuhan, China. RP Zhu, RSh (reprint author), Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Department of Stomatology, 430030 Wuhan, China. EM tongji36@yahoo.com CR Barnes L, Eveson JW, Reichart P, Sidransky D, 2005, World Health Organization classification of tumours. Pathology and genetics of head and neck tumours. IARC, Lyon Stoelinga PJ, Bronkhorst FB, 1988, The incidence, multiple presentation and recurrence of aggressive cysts of the jaws. J Craniomaxillofac Surg 16:184–195 Lund VJ, 1985, Odontogenic keratocyst of the maxilla: a case report. Br J Oral Maxillofac Surg 23:210–215 Madras J, Lapointe H, 2008, Keratocystic odontogenic tumour: reclassification of the odontogenic keratocyst from cyst to tumour. J Can Dent Assoc 74:165–165h Shear M, Speight PM, 2007, Odontogenic keratocyst. In: Shear M, Speight PM, eds, Cysts of the Oral and Maxillofacial Regions, 4th edn. Blackwell Munksgaard, Oxford, pp 6–58 Scholzen T, Gerdes J, 2000, The Ki-67 protein: from the known and the unknown. J Cell Physiol 182:311–322 Gurgel CA, Ramos EA, Azevedo RA, Sarmento VA, da Silva Carvalho AM, dos Santos JN, 2008, Expression of Ki-67, p53 and p63 proteins in keratocyst odontogenic tumours: an immunohistochemistry study. J Mol Histol 39:311–316 Gadbail AR, Chaudhary M, Patil S, Gawande M, 2009, Actual Proliferating Index and p53 protein expression as prognostic marker in odontogenic cysts. Oral Dis 15:490–498 de Oliveira MG, Lauxen Ida S, Chaves AC, Rados PV, Sant'Ana Filho M, 2011, Odontogenic epithelium: immunolabeling of Ki-67, EGFR and survivin in pericoronal follicles, dentigerous cysts and keratocystic odontogenic tumors. Head Neck Pathol 5:1–7 Smith WL, DeWitt DL, Garavito RM, 2000, Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem 69: 145–182 Marnett LJ, DuBois RN, 2002, COX-2: a target for colon cancer prevention. Annu Rev Pharmacol Toxicol 42:55–80 Kurihara Y, Hatori M, Ando Y, Ito D, Toyoshima T, Tanaka M, Shintani S, 2009, Inhibition of cyclooxygenase-2 suppresses the invasiveness of oral squamous cell carcinoma cell lines via downregulation of matrix metalloproteinase-2 production and activation. Clin Exp Metastasis 26:425–432 Chang F, Syrjanen S, Kellokoski J, Syrjanen K, 1991, Human papillomavirus, HPV, infections and their associations with oral disease. J Oral Pathol Med 20:305–317 Psyrri A, Boutati E, Karageorgopoulou S, 2011, Human papillomavirus in head and neck cancers: biology, prognosis, hope of treatment, and vaccines. Anticancer Drugs 22:586–590 Kahn MA, 1989, Ameloblastoma in young persons: a clinicopathologic analysis and etiologic investigation. Oral Surg Oral Med Oral Pathol 67:706–715 KahnMA, 1992, Demonstration of human papillomavirus DNA in a peripheral ameloblastoma by in situ hybridization. Hum Pathol 23: 188–191 Namin AK, Azad TM, Eslami B, Sarkarat F, Shahrokhi M, Kashanian F, 2003, A study of the relationship between ameloblastoma and human papilloma virus. J Oral Maxillofac Surg 61:467–470 Cox M, Eveson J, Scully C, 1991, Human papillomavirus type 16 DNA in an odontogenic keratocyst. J Oral Pathol Med 20: 143–145 Gonzalez-Moles MA, Mosqueda-Taylor A, Delgado-Rodriguez M et al, 2006, Analysis of p53 protein by PAb240, Ki-67 expression and human papillomavirus DNA detection in different types of odontogenic keratocyst. Anticancer Res 26:175–181 Kleter B, van Doorn LJ, ter Schegget J et al, 1998, Novel shortfragment PCR assay for highly sensitive broad-spectrum detection of anogenital human papillomaviruses. Am J Pathol 153: 1731–1739 Syrjanen S, Lodi G, von Bultzingslowen I et al, 2011, Human papillomaviruses in oral carcinoma and oral potentially malignant disorders: a systematic review. Oral Dis 17(Suppl 1):58–72 Miller CS, Johnstone BM, 2001, Human papillomavirus as a risk factor for oral squamous cell carcinoma: ameta-analysis, 1982–1997. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 91:622–635 Montaldo C, Mastinu A, Quartuccio M et al, 2007, Detection and genotyping of human papillomavirus DNA in samples from healthy Sardinian patients: a preliminary study. J Oral Pathol Med 36:482– 487 SahebJamee M, Boorghani M, Ghaffari SR, AtarbashiMoghadam F, Keyhani A, 2009, Human papillomavirus in saliva of patients with oral squamous cell carcinoma. Med Oral Patol Oral Cir Bucal 14: e525–528 Adelstein DJ, Ridge JA, Gillison ML et al, 2009, Head and neck squamous cell cancer and the human papillomavirus: summary of a National Cancer Institute State of the Science Meeting, November 9– 10, 2008, 31. Head Neck, Washington DC, pp 1393–1422 Balan R, Simion N, Giusca SE et al, 2011, Immunohistochemical assessment of p16, COX-2 and EGFR in HPV-positive cervical squamous intraepithelial lesions. Rom J Morphol Embryol 52: 1187–1194 Ozbun MA, Meyers C, 1997, Characterization of late gene transcripts expressed during vegetative replication of human papillomavirus type 31b. J Virol 71:5161–5172 Correnti M, Rossi M, Avila M, Perrone M, Rivera H, 2010, Human papillomavirus in ameloblastoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 110:e20–24 van Heerden WF, van Rensburg EJ, Raubenheimer EJ, Venter EH, 1993, Detection of human papillomavirus DNA in an ameloblastoma using the in situ hybridization technique. J Oral Pathol Med 22:109–112 Sand L, Jalouli J, Larsson PA, Magnusson B, Hirsch JM, 2000, Presence of human papilloma viruses in intraosseous ameloblastoma. J Oral Maxillofac Surg 58:1129–1134 Migaldi M, PecorariM, Rossi G et al, 2005, Does HPV play a role in the etiopathogenesis of ameloblastoma? An immunohistochemical, in situ hybridization and polymerase chain reaction study of 18 cases using laser capture microdissection. Mod Pathol 18:283–289 Dalgleish AG, 1991, Viruses and cancer. Br Med Bull 47:21–46 Stubenrauch F, Laimins LA, 1999, Human papillomavirus life cycle: active and latent phases. Semin Cancer Biol 9:379–386 Garner-Hamrick PA, Fostel JM, Chien WM et al, 2004, Global effects of human papillomavirus type 18 E6/E7 in an organotypic keratinocyte culture system. J Virol 78:9041–9050 Samir R, Asplund A, Tot T, Pekar G, Hellberg D, 2011, High-risk HPV infection and CIN grade correlates to the expression of c-myc, CD4+, FHIT, E-cadherin, Ki-67, and p16INK4a. J Low Genit Tract Dis 15:280–286 de Vicente JC, Torre-Iturraspe A, Gutierrez AM, Lequerica- Fernandez P, 2010, Immunohistochemical comparative study of the odontogenic keratocysts and other odontogenic lesions. Med Oral Patol Oral Cir Bucal 15:e709–715 Gonzalez-Alva P, Tanaka A, Oku Y, Yoshizawa D, Itoh S, Sakashita H, Ide F, Tajima Y, Kusama K, 2008, Keratocystic odontogenic tumor: a retrospective study of 183 cases. J Oral Sci 50:205–212 Kim SH, Oh JM, No JH, Bang YJ, Juhnn YS, Song YS, 2009, Involvement of NF-kappaB and AP-1 in COX-2 upregulation by human papillomavirus 16 E5 oncoprotein. Carcinogenesis 30:753–757 Wu R, Coniglio SJ, Chan A, Symons MH, Steinberg BM, 2007, Upregulation of Rac1 by epidermal growth factor mediates COX-2 expression in recurrent respiratory papillomas.MolMed 13:143–150 Subbaramaiah K, Dannenberg AJ, 2007, Cyclooxygenase-2 transcription is regulated by human papillomavirus 16 E6 and E7 oncoproteins: evidence of a corepressor/coactivator exchange. Cancer Res 67:3976–3985 Zhuang ZH, Tsao SW, Deng W et al, 2008, Early upregulation of cyclooxygenase-2 in human papillomavirus type 16 and telomeraseinduced immortalization of human esophageal epithelial cells. J Gastroenterol Hepatol 23:1613–1620 Prescott SM, Fitzpatrick FA, 2000, Cyclooxygenase-2 and carcinogenesis. Biochim Biophys Acta 1470:M69–78 Mohan S, Epstein JB, 2003, Carcinogenesis and cyclooxygenase: the potential role of COX-2 inhibition in upper aerodigestive tract cancer. Oral Oncol 39:537–546 Mestre JR, Subbaramaiah K, Sacks PG et al, 1997, Retinoids suppress epidermal growth factor-induced transcription of cyclooxygenase-2 in human oral squamous carcinoma cells. Cancer Res 57:2890–2895 Mendes RA, Carvalho JF,Waal I, 2009, An overview on the expression of cyclooxygenase-2 in tumors of the head and neck. Oral Oncol 45:e124–128 Zhang Y, McCluskey K, Fujii K, Wahl LM, 1998, Differential regulation of monocyte matrix metalloproteinase and TIMP-1 production by TNF-α, granulocyte-macrophage CSF and IL-1β through prostaglandin-dependent and -independent mechanisms. J Immunol 161:3071–3076 Callejas NA, Casado M, Diaz-Guerra MJ, Bosca L, Martin-Sanz P, 2001, Expression of cyclooxygenase-2 promotes the release of matrix metalloproteinase-2 and −9 in fetal rat hepatocytes. Hepatology 33:860–867 Ottino P, Bazan HE, 2001, Corneal stimulation of MMP-1, −9 and uPA by platelet-activating factor is mediated by cyclooxygenase-2 metabolites. Curr Eye Res 23:77–85 Gomes CC, Duarte AP, DinizMG, Gomez RS, 2010, Review article: Current concepts of ameloblastoma pathogenesis. J Oral Pathol Med 39:585–591 HarrisM(1978, Odontogenic cyst growth and prostaglandin-induced bone resorption. Ann R Coll Surg Engl 60:85–91 Fracon RN, Teofilo JM, Satin RB, Lamano T, 2008, Prostaglandins and bone: potential risks and benefits related to the use of nonsteroidal anti-inflammatory drugs in clinical dentistry. J Oral Sci 50:247– 252 Gao L, Li TJ, 2005, In vitro study on bone resorption of odontogenic cysts and ameloblastomas. Zhonghua Kou Qiang Yi Xue Za Zhi 40: 233–236 Mendes RA, Carvalho JF, van der Waal I, 2011, Potential relevance of cyclooxygenase-2 expression in keratocystic odontogenic tumours - an immunohistochemical study. J Oral Pathol Med 40:497–503 Tsai CH, Huang FM, Yang LC, Chou MY, Chang YC, 2002, Immunohistochemical localization of cyclooxygenase-2 in radicular cysts. Int Endod J 35:854–858 Johnson RL, Rothman AL, Xie J et al, 1996, Human homolog of patched, a candidate gene for the basal cell nevus syndrome. Science 272:1668–1671 Ren C, Amm HM, DeVilliers P et al, 2012, Targeting the sonic hedgehog pathway in keratocystic odontogenic tumor. J Biol Chem 287:27117–27125 Bhargava D, Deshpande A, Pogrel MA, 2012, Keratocystic odontogenic tumour, KCOT)–a cyst to a tumour. Oral Maxillofac Surg 16:163–170 Xuan YH, Li GL, Jiang HY, Lin ZH, 2009, Relationship between hedgehog signaling pathway molecules and HPV16 infection in uterine cervical cancers. Zhonghua Bing Li Xue Za Zhi 38:178–182 Samarzija I, Beard P, 2012, Hedgehog pathway regulators influence cervical cancer cell proliferation, survival and migration. Biochem Biophys Res Commun 425:64–69 Yagyuu T, Kirita T, Sasahira T,Moriwaka Y, Yamamoto K, Kuniyasu H, 2008, Recurrence of keratocystic odontogenic tumor: clinicopathological features and immunohistochemical study of the Hedgehog signaling pathway. Pathobiology 75:171–176 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 65 EP 71 DI 10.1007/s12253-014-9789-3 PG 7 ER PT J AU Gurgel, CD Valenca-Junior, TJ Dornelas, AC Vieira, BR Maia-Filho, TAJ Lima-Junior, CPR Ribeiro, AR Almeida, RCP AF Gurgel, Cordeiro Daniel Valenca-Junior, Telmo Jose Dornelas, Aparecida Conceicao Vieira, Braga Renato Maia-Filho, Tarcisio Alves Joao Lima-Junior, Cesar Pereira Roberto Ribeiro, Albuquerque Ronaldo Almeida, Roberto Carvalho Paulo TI Immunoexpression of Metalloproteinases 2 and 14 and TIMP-2 Inhibitor in Main Types of Primary Gastric Carcinomas and Lymph Node Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Immunohistochemistry; Macrophage; Metalloproteinase; Metastasis ID Gastric cancer; Immunohistochemistry; Macrophage; Metalloproteinase; Metastasis AB Metalloproteinase-2 (MMP-2) and −14 (MMP-14) and the tissue inhibitor of metalloproteinases type 2 (TIMP-2) participate in epithelial-mesenchymal transition and tumor progression in many cancers. However, the correlation between these enzymes in gastric cancer and the metastatic potential to their respective lymph node needs to be determined. Here, we evaluated the expression of these enzymes in gastric carcinoma and lymph node metastases and their possible involvement in tumor progression. Histological samples from 83 patients with gastric cancer and their respective lymph nodes were used. MMP-2,MMP-14 and TIMP-2 immunoexpression was scored. TIMP-2 expression in tumor-associated macrophages occurred more frequently than in normal mucosa (P=0.0128). Female tumor samples presented higher MMP-2 expression (P= 0.0248), while TIMP-2 occurred mainly in patients over 50 years old (P=0.0034). MMP-2 was higher expressed in primary tumor macrophages than in neoplastic cells (P=0.0118), and was also seen in macrophages from metastatic-affected lymph nodes of intestinal and diffuse histotypes (P=0.0006). MMP-2, MMP-14 and TIMP-2 expression in mononuclear cells might be correlated with progression of gastric cancer. MMP-14 production by macrophages appears to be more involved in diffuse gastric cancer progression. C1 [Gurgel, Cordeiro Daniel] Federal University of Ceara, Faculty of Medicine, Department of Pathology and Forensic Medicine, Rua Monsenhor Furtado S/N, Rodolfo Teofilo, 60430-350 Fortaleza, Ceara, Brazil. [Valenca-Junior, Telmo Jose] Federal University of Ceara, Faculty of Medicine, Department of Pathology and Forensic Medicine, Rua Monsenhor Furtado S/N, Rodolfo Teofilo, 60430-350 Fortaleza, Ceara, Brazil. [Dornelas, Aparecida Conceicao] Federal University of Ceara, Faculty of Medicine, Department of Pathology and Forensic Medicine, Rua Monsenhor Furtado S/N, Rodolfo Teofilo, 60430-350 Fortaleza, Ceara, Brazil. [Vieira, Braga Renato] Federal University of Ceara, Faculty of Medicine, Department of Pathology and Forensic Medicine, Rua Monsenhor Furtado S/N, Rodolfo Teofilo, 60430-350 Fortaleza, Ceara, Brazil. [Maia-Filho, Tarcisio Alves Joao] Federal University of Ceara, Faculty of Medicine, Department of Pathology and Forensic Medicine, Rua Monsenhor Furtado S/N, Rodolfo Teofilo, 60430-350 Fortaleza, Ceara, Brazil. [Lima-Junior, Cesar Pereira Roberto] Federal University of Ceara, Faculty of Medicine, Department of Physiology and PharmacologyFortaleza, Ceara, Brazil. [Ribeiro, Albuquerque Ronaldo] Federal University of Ceara, Faculty of Medicine, Department of Physiology and PharmacologyFortaleza, Ceara, Brazil. [Almeida, Roberto Carvalho Paulo] Federal University of Ceara, Faculty of Medicine, Department of Pathology and Forensic Medicine, Rua Monsenhor Furtado S/N, Rodolfo Teofilo, 60430-350 Fortaleza, Ceara, Brazil. RP Almeida, RCP (reprint author), Federal University of Ceara, Faculty of Medicine, Department of Pathology and Forensic Medicine, 60430-350 Fortaleza, Brazil. EM paulorcalmeida@uol.com.br;paulo.almeida@ufc.br CR Fitzgerald R, Hardwick R, Huntsman D et al, 2010, Hereditary diffuse gastric cancer: update consensus guidelines for clinical management and directions for future research. JMed Genet 47:436–444 Instituto Nacional de Cancer, Brasil,, 2012, Estimativa 2012: incidencia de cancer no Brasil. INCA, Rio de Janeiro, http:// www.inca.gov.br/estimativa/2012/ Rodrigues M, Queiroz D, Rodrigues R et al, 2005, Prevalence of Helicobacter pylori infection in Fortaleza, Northeastern Brazil. Rev Saude Publica 39:847–849 Gao D, Vahdat L, Wong S et al, 2012, Microenvironmental regulation of epithelial-mesenchymal transitions in cancer. Cancer Res 72(19):4883–4889 Talbot L, Bhattacharya S, Kuo P, 2012, Epithelial-mesenchymal transition, the tumor microenvironment, and metastatic behavior of epithelial malignancies. Int J Biochem Mol Biol 3(2):117–136 Munshi H, StackM(2006, Reciprocal interactions between adhesion receptor signaling and MMP regulation. Cancer Metastasis Rev 25: 45–56 Bourboulia D, Stetler-Stevenson W, 2010, Matrix MetalloProteinases and Tissue Inhibitors of Metalloproteinases: positive and negative regulators in tumor cell adhesion. Semin Cancer Biol 20(3):161–168 Al-Dasooqi N, Gibson R, Bowen J, Keefe D, 2009, Matrix metalloproteinases: key regulators in the pathogenesis of chemoterapyinduced mucositis? Cancer Chemother Pharmacol 64:1–9 Ries C, Egea V, Karow M et al, 2007, MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: differential regulation by inflammatory cytokines. Blood 109(9):4055–4063 Azar D, Casanova F,Mimura Tet al, 2010, Corneal epithelialMMP- 14 inhibits vascular endothelial cell proliferation and migration. Cornea 29:321–330 Brew K, Nagase H, 2010, The tissue inhibitors of metalloproteinases, TIMPs): an ancient family with structural and functional diversity. Biochim Biophys Acta 1803:55–71 Mrena J,Wiksten J, Nordling S et al, 2006, MMP-2 but not MMP-9 associated with COX-2 and survival in gastric cancer. J Clin Pathol 59:618–623 Ra H, Parks W, 2007, Control of matrix metalloproteinase catalytic activity. Matrix Biol 26:587–596 Kubben F, Sier C, Van DuijnWet al, 2006)Matrix metalloproteinase-2 is a consistent prognostic factor in gastric cancer. Brit J Cancer 95:1035– 1040 Yoshikawa T, Yanoma S, Tsuburaya A et al, 2006, Expression of MMP-7 and MMP-14 in peritoneal dissemination of gastric cancer. Hepatogastroenterology 53:964–967 Hong S, Park I, Hong W et al, 1996, Overexpression of tissue inhibitors of metalloproteinase −1 and −2 in the stroma of gastric cancer. J Korean Med Sci 11:474–479 Sobin L, Gospodarowicz M,Wittekind C, 2010, TNM classification of malignant tumours. International union against cancer, 7th edn. Wiley-Blackwell, New York Lauren P, 1965, The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand 64:31–49 Gurgel D, Dornelas C, Lima-Junior R et al, 2012, An adapted tissue microarray for the development of a matrix arrangement of tissue samples. Pathol Res Pract 208:167–168 Rojiani M, Alidina J, Esposito N, Rojiani A, 2010, Expression of MMP-2 correlates with increased angiogenesis in CNS metastasis of lung carcinoma. Int J Clin Exp Pathol 3(8):775–781 Bonfil R, Dong Z, Trindade-Filho J et al, 2007, Prostate cancerassociated membrane type 1 – matrix metalloproteinase. A pivotal role in bone response and intraosseous tumor growth. Am J Pathol 170:2100–2111 Dong B, Sato M, Sakurada A et al, 2005, Computed tomographic images reflect the biologic behavior of small lung adenocarcinoma: they correlate with cell proliferation, microvascularization, cell adhesion, degradation of extracellular matrix, and K-ras mutation. J Thorac Cardiovasc Surg 130:733–739 Buskens C, Sivula A, Van Rees B et al, 2003, Comparison of cyclooxygenase 2 expression in adenocarcinomas of the gastric cardia and distal oesophagus. Gut 52:1678–1683 Hwang TL, Lee LY, Wang CC et al, 2010, Claudin-4 expression is associated with tumor invasion, MMP-2 and MMP-9 expression in gastric cancer. Exp Ther Med 1(5):789–797 Zhou Y, Li G, Wu J et al, 2010, Clinicopathological significance of Ecadherin, VEGF, and MMPs in gastric cancer. Tumour Biol 31(6):549– 558 Wu ZY, Li JH, Zhan WH, He YL, 2006, Lymph node micrometastasis and its correlation with MMP-2 expression in gastric carcinoma. World J Gastroenterol 12(18):2941–2944 Kubben FJ, Sier CF, Meijer MJ et al, 2006, Clinical impact of MMP and TIMP gene polymorphisms in gastric cancer. Br J Cancer 95(6): 744–751 Alakus H, Afriani N,Warnecke-Eberz U et al, 2010, Clinical impact of MMP and TIMP gene polymorphisms in gastric cancer. World J Surg 34(12):2853–2859 Joo YE, Seo KS, Kim HS et al, 2000, Expression of tissue inhibitors of metalloproteinases, TIMPs, in gastric cancer. Dig Dis Sci 45(1): 114–121 Zhang JF, Zhang YP, Hao FYet al, 2005, DNA ploidy analysis and expression ofMMP-9, TIMP-2, and E-cadherin in gastric carcinoma. World J Gastroenterol 11(36):5592–5600 Zheng H, Takahashi H,Murai Yet al, 2006, Expressions ofMMP-2, MMP-9 and VEGF are closely linked to growth, invasion, metastasis and angiogenesis of gastric carcinoma. Anticancer Res 26(5A):3579– 3583 Alakus H, Grass G, Hennecken JK et al, 2008, Clinicopathological significance of MMP-2 and its specific inhibitor TIMP-2 in gastric cancer. Histol Histopathol 23(8):917–923 He L, Chu D, Li X et al, 2013, Matrix metalloproteinase-14 is a negative prognostic marker for patients with gastric cancer. Dig Dis Sci 58(5):1264–1270 Dicken B, Graham K, Hamiltom S et al, 2006, Lymphovascular invasion is associated with poor survival in gastric cancer: an application of gene-expression and tissue array techniques. Ann Surg 243:64–73 Gu Q, Wang D, Gao Yet al, 2002, Expression of MMP1 in surgical and radiation-impaired wound healing and its effects on the healing process. J Environ Pathol Toxicol Oncol 21(1):71–78 Murnane M, Cai J, Shuja S et al, 2009, Active MMP-2 effectively identifies the presence of colorectal cancer. Int J Cancer 125:2893–2902 Wang LB, Jiang ZN, Fan MYet al, 2008, Changes of histology and expression ofMMP-2 and nm23-H1 in primary andmetastatic gastric cancer. World J Gastroenterol 14(10):1612–1616 Monig S, Baldus S, Hennecken J et al, 2001, Expression of MMP-2 is associated with progression and lymph node metastasis of gastric carcinoma. Histopathology 39:597–602 Lee LY, Wu CM, Wang CC et al, 2008, Expression of matrix metalloproteinases MMP-2 and MMP-9 in gastric cancer and their relation to claudin-4 expression. Histol Histopathol 23(5):515–521 Freudenberg J, Chen W, 2007, Induction of Smad1 by MMP-14 contributes to tumor growth. Int J Cancer 121:966–977 Seiki M,Yana I, 2003, Roles of pericellular proteolysis bymembrane type-1 matrix metalloproteinase in cancer invasion and angiogenesis. Cancer Sci 94(7):569–574 Allavena P, Sica A, Solinas G et al, 2008, The inflammatory microenvironment in tumor progression: the role of tumor-associated macrophages. Crit Rev Oncol Hematol 66:1–9 Kang J, Chen J, Lee C et al, 2010, Intratumoral macrophage counts correlate with tumor progression in colorectal cancer. J Surg Oncol 102:242–248 Roa M, Asbun J, Ruiz A et al, 2008, Expression of MMP-1 and MMP-11 in squamous cell carcinoma of the nasal cavity and paranasal sinuses. Patol Rev Latinoam 46:209–214 Ribeiro B, Iglesias D, Nascimento G et al, 2009, Immunoexpression of MMPs-1, −2 and −9 in ameloblastoma and odontogenic adenomatoid tumor. Oral Dis 15:472–477 Ishigaki S, ToiM, Ueno Tet al, 1999, Significance ofmembrane type 1 matrix metalloproteinase expression in breast cancer. Jpn J Cancer Res 90:516–522 Mylona E, Nomikos A, Magkou C et al, 2007, The clinicopathological and prognostic significance of membrane type 1 matrix metalloproteinase, MT1-MMP, and MMP-9 according to their localization in invasive breast carcinoma. Histopathology 50(3):338–347 Karadag N, Kirimlioglu H, Isik B et al, 2008, Expression of matrix metalloproteinases in gallbladder carcinoma and their significance in carcinogenesis. Appl Immunohistochem Mol Morphol 16(2):148–152 Markovic D, Vinnakota K, Chirasani S et al, 2009, Gliomas induce and exploit microglial MMP-14 expression for tumor expansion. PNAS 106:12530–12535 Mori M, Mimori K, ShiraishiTet al, 1997)Analysis ofMT1-MMP and MMP2 expression in human gastric cancers. Int J Cancer 74(3):316–321 Murray GI, Duncan ME, Arbuckle E et al, 1998, Matrix metalloproteinases and their inhibitors in gastric cancer. Gut 43(6):791–797 SternlichtM,Werb Z, 2001, How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 17:463–516 Munshi H, Wu Y, Mukhopadhyay S et al, 2004, Differential regulation of membrane type 1-matrix metalloproteinase activity by ERK 1/2- and p38MAPK-modulated tissue inhibitor ofmetalloproteinases 2 expression controls transforming growth factor-β1-induced pericelular collagenolysis. J Biol Chem 279:39042–39050 Sounni N, Rozanov D, Remacle A et al, 2010, TIMP-2 binding with cellular MMP-14 stimulates invasion-promoting MEK/ERK signaling in cancer cells. Int J Cancer 126:1067–1078 Tetu B, Brisson J, Wang CS et al, 2006, The influence of MMP-14, TIMP-2 and MMP-2 expression on breast cancer prognosis. Breast Cancer Res 8(3), 1-9-R28 Reis ST, Antunes AA, Pontes J Jr et al, 2012, Underexpression of MMP-2 and its regulators, TIMP2, MT1-MMP and IL-8, is Associated with Prostate Cancer. Int Braz J Urol 38:167–174 Kikuchi R, Noguchi T, Takeno S et al, 2000, Immunohistochemical detection of membrane-type-1-matrix metalloproteinase in colorectal carcinoma. Br J Cancer 83(2):215–218 Rodriguez D, Morrison CJ, Overall CM, 2010, Matrix metalloproteinases: what do they not do? New substrates and biological roles identified by murine models and proteomics. Biochim Biophys Acta 1803(1):39–54 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 73 EP 81 DI 10.1007/s12253-014-9790-x PG 9 ER PT J AU Zhong, W Yu, Z Zhan, J Yu, T Lin, Y Xia, ZSh Yuan, YH Chen, QK AF Zhong, Wa Yu, Zhong Zhan, Jun Yu, Tao Lin, Ying Xia, Zhong-Sheng Yuan, Yu-Hong Chen, Qi-Kui TI Association of Serum Levels of CEA, CA199, CA125, CYFRA21-1 and CA72-4 and Disease Characteristics in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Cancer marker; TNM stage; Duke stage; Overall survival ID Colorectal cancer; Cancer marker; TNM stage; Duke stage; Overall survival AB Identifying predictive biomarkers for colorectal cancer would facilitate diagnosis and treatment of the disease. This study aimed to investigate the association of the serological biomarkers CEA, CA19–9, CA125, CYFRA21–1 and CA72–4 with patient characteristics and disease outcomes in colorectal cancer. Patients (N=373) with colorectal cancer were evaluated for the association of CEA, CA19–9, CA125, CYFRA21–1, and CA72–4 pre and post-surgery and at disease recurrence with demographics, disease characteristics including pathological types, degree of differentiation, invasion depth, abdominal lymph node metastasis, TMN stage, Dukes stage, location of cancer and metastasis, and disease outcomes. It was more common for a patient to express these markers prior to surgery and at disease recurrence than following surgery. Overall, the serum levels of CEA, CA19–9, CA125, CYFRA21–1, and CA72–4 were not associatedwith age, gender, pathological type and location of cancer (all P-values >0.05), but were associated with the poor tumor differentiation, higher tumor invasion, greater degree of abdominal lymph node metastasis, and higher TNM and Duke stage tumors (all P-values<0.01). CEA expressionwas associatedwith older ages (median age 65 years). Multivariate analysis indicated that CEA was correlated with overall survival and none of the markers correlated with disease recurrence. The expression of CEA, CA19–9, CA125, CYFRA21–1, and CA72–4 was associated with specific disease characteristics which tended to indicated more advanced disease and disease recurrence consistent with these biomarkers being useful for detecting colorectal cancer. C1 [Zhong, Wa] Sun Yat-Sen University, Sun Yat-Sen Memorial Hospital, Department of Gastroenterology, No.107 Yanjiang Xi Road, Yuexiu District, 510120 Guangzhou, Guangdong, China. [Yu, Zhong] Sun Yat-Sen University, Sun Yat-Sen Memorial Hospital, Department of Gastroenterology, No.107 Yanjiang Xi Road, Yuexiu District, 510120 Guangzhou, Guangdong, China. [Zhan, Jun] Sun Yat-Sen University, Sun Yat-Sen Memorial Hospital, Department of Gastroenterology, No.107 Yanjiang Xi Road, Yuexiu District, 510120 Guangzhou, Guangdong, China. [Yu, Tao] Sun Yat-Sen University, Sun Yat-Sen Memorial Hospital, Department of Gastroenterology, No.107 Yanjiang Xi Road, Yuexiu District, 510120 Guangzhou, Guangdong, China. [Lin, Ying] Sun Yat-Sen University, Sun Yat-Sen Memorial Hospital, Department of Gastroenterology, No.107 Yanjiang Xi Road, Yuexiu District, 510120 Guangzhou, Guangdong, China. [Xia, Zhong-Sheng] Sun Yat-Sen University, Sun Yat-Sen Memorial Hospital, Department of Gastroenterology, No.107 Yanjiang Xi Road, Yuexiu District, 510120 Guangzhou, Guangdong, China. [Yuan, Yu-Hong] Sun Yat-Sen University, Sun Yat-Sen Memorial Hospital, Department of Gastroenterology, No.107 Yanjiang Xi Road, Yuexiu District, 510120 Guangzhou, Guangdong, China. [Chen, Qi-Kui] Sun Yat-Sen University, Sun Yat-Sen Memorial Hospital, Department of Gastroenterology, No.107 Yanjiang Xi Road, Yuexiu District, 510120 Guangzhou, Guangdong, China. RP Chen, QK (reprint author), Sun Yat-Sen University, Sun Yat-Sen Memorial Hospital, Department of Gastroenterology, 510120 Guangzhou, China. EM qikuichen@yahoo.com CR Jemal A, Bray F, CenterMMet al, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Colorectal cancerfacts & figures: 2010–2013. http://www.cancer.org/ acs/groups/content/@epidemiologysurveilance/documents/ document/acspc-028323.pdf. Accessed: August 16 2013 Liu X, Cai H, Wang Y, 2012, Prognostic significance of tumour markers in chinese patients with gastric cancer. ANZ J Surg., DOI 10. 1111/j.1445-2197.2012.06287.x McKeown E, Nelson DW, Johnson EK et al, 2014, Current approaches and challenges for monitoring treatment response in colon and rectal cancer. J Cancer 5(1):31–43 Perkins GL, Slater ED, Sanders GK et al, 2003, Serum tumor markers. Am Fam Physician 68(6):1075–1082 Goldberg EM, Simunovic LM, Drake SL et al, 1989, Comparison of serum ca 19–9 and cea levels in a population at high risk for colorectal cancer. Hybridoma 8(5):569–575 Bast RC Jr, Klug TL, St John E et al, 1983, A radioimmunoassay using a monoclonal antibody to monitor the course of epithelial ovarian cancer. N Engl J Med 309(15):883–887 Zhang D, Yu M, Xu T et al, 2013, Predictive value of serum cea, ca19–9 and ca125 in diagnosis of colorectal liver metastasis in chinese population. Hepatogastroenterology 60(126):1297–1301 Ohuchi N, Takahashi K,Matoba N et al, 1989, Comparison of serum assays for tag-72, ca19–9 and cea in gastrointestinal carcinoma patients. Jpn J Clin Oncol 19(3):242–248 Shitara M, Yoshioka H, Sakurabayashi I et al, 1988, fundamental studies and the reference values of cancer associated antigen tag-72, ca72–4). Rinsho Byori 36(10):1177–1181 KobayashiH, 1989, The clinical usefulness of serumca72–4 analysis in patients with ovarian cancer. Nihon Sanka Fujinka Gakkai Zasshi 41(5):585–589 Guadagni F, Roselli M, Cosimelli M et al, 1995, Ca 72–4 serum marker–a new tool in the management of carcinoma patients. Cancer Investig 13(2):227–238 Kosacka M, Jankowska R, 2009, Comparison of cytokeratin 19 expression in tumor tissue and serum cyfra 21–1 levels in non-small cell lung cancer. Pol Arch Med Wewn 119(1–2):33– 37 Pang L, Wang J, Jiang Y et al, 2013, Decreased levels of serum cytokeratin 19 fragment cyfra 21–1 predict objective response to chemotherapy in patients with non-small cell lung cancer. Exp Ther Med 6(2):355–360 Pujol JL, Grenier J, Daures JP et al, 1993, Serum fragment of cytokeratin subunit 19 measured by cyfra 21–1 immunoradiometric assay as a marker of lung cancer. Cancer Res 53(1):61–66 Holdenrieder S, Stieber P, Liska V et al, 2012, Cytokeratin serum biomarkers in patients with colorectal cancer. Anticancer Res 32(5): 1971–1976 Wild N, Andres H, RollingerWet al, 2010, A combination of serum markers for the early detection of colorectal cancer. Clin Cancer Res 16(24):6111–6121 Kornek GV, Depisch D, Rosen HR et al, 1992, Comparative analysis of ca72–4, ca195 and carcinoembryonic antigen in patients with gastrointestinal malignancies. J Cancer Res Clin Oncol 118(4):318– 320 Posner MR, Mayer RJ, 1994, The use of serologic tumor markers in gastrointestinal malignancies. Hematol Oncol Clin North Am 8(3): 533–553 Ueda T, Shimada E, Urakawa T, 1994, The clinicopathologic features of serum ca 19–9-positive colorectal cancers. Surg Today 24(6): 518–525 Filella X, Molina R, Grau JJ et al, 1992, Prognostic value of ca 19.9 levels in colorectal cancer. Ann Surg 216(1):55–59 Zheng CX, Zhan WH, Zhao JZ et al, 2001, The prognostic value of preoperative serum levels of cea, ca19–9 and ca72–4 in patientswith colorectal cancer. World J Gastroenterol 7(3):431–434 Streppel MM, Vincent A, Mukherjee R et al, 2012, Mucin 16, cancer antigen 125, expression in human tissues and cell lines and correlation with clinical outcome in adenocarcinomas of the pancreas, esophagus, stomach, and colon. Hum Pathol 43(10):1755–1763 Zhang D, Yu M, Xu T et al, 2013, Predictive value of serum cea, ca19–9 and ca125 in diagnosis of colorectal liver metastasis in chinese population. Hepatogastroenterology 60(126):1301 Yang XQ, Chen C, Wang FB et al, 2011, Preoperative serum carcinoembryonic antigen, carbohydrate antigen19–9 and carbohydrate antigen 125 as prognostic factors for recurrence-free survival in colorectal cancer. Asian Pac J Cancer Prev 12(5): 1251–1256 Yang XQ, Li Y, Chen C et al, 2011, Preoperative serum carbohydrate antigen 125 level is an independent negative prognostic marker for overall survival in colorectal cancer. Med Oncol 28(3):789–795 Forones NM, Tanaka M, 1999, Cea and ca 19–9 as prognostic indexes in colorectal cancer. Hepatogastroenterology 46(26):905– 908 Nakayama T, Watanabe M, Teramoto T et al, 1997, Ca19–9 as a predictor of recurrence in patients with colorectal cancer. J Surg Oncol 66(4):238–243 Shimono R,Mori M, Akazawa K et al, 1994, Immunohistochemical expression of carbohydrate antigen 19–9 in colorectal carcinoma.Am J Gastroenterol 89(1):101–105 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 83 EP 95 DI 10.1007/s12253-014-9791-9 PG 13 ER PT J AU Moatter, T Aban, M Iqbal, W Pervez, Sh AF Moatter, Tariq Aban, Muniba Iqbal, Waseem Pervez, Shahid TI Cyclooxygenase-2 Polymorphisms and Breast Cancer Associated Risk in Pakistani Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cyclooxygenase; Cox-2; Breast Cancer; Polymorphism; Haplotypes; RFLP ID Cyclooxygenase; Cox-2; Breast Cancer; Polymorphism; Haplotypes; RFLP AB Prostaglandins produced by Cyclooxygenase-2 enzyme have been implicated to have a role in breast carcinogenesis. Several single nucleotide polymorphisms (SNPs) linked to COX-2 enzyme are reported to modulate its expression. The aim of the present study was to examine association of these SNPs to breast cancer risk in Pakistani patients. Methods In this case–control study, three sequence variants rs689465, rs689466, rs20417 in the promoter region of COX- 2 were screened to evaluate the association with breast cancer risk. A total of 150 breast cancer patients and 101 healthy control genomic DNA were genotyped for rs689456, rs689466, rs20417 and their genotypes distribution in cases and control were compared using Pearson chi square test. Risk association was analyzed through odd ratio calculated by logistic regression. Results A screening analysis of COX-2 SNPs in 101 healthy controls showed distribution of Minor allelic frequency distribution of SNPs as follows : rs689465 (0.12), rs689466 (0.15), rs20417 (0.23). Further analyses revealed that their observed genotype frequencies were consistent with Hardy Weinberg equilibrium and strong linkage disequilibrium was identified between rs20417, rs689465 and rs689466. The Combined allele variants analysis showed that Haplotype rs68965G- 689466A-20417C (OR 2.909; CI 95 %1.3776.327; P=0.007) was significantly associated with breast cancer. Conclusions Our results indicate no strong association between three most frequent COX-2 SNPs rs689465 rs689466, rs20417 studied with breast cancer risk in the single locus analysis. However, our data suggested that combined COX-2 SNP haplotype have a role in breast cancer associated risk in Pakistani patients. C1 [Moatter, Tariq] The Aga Khan University, Department of Pathology and Microbiology, Stadium Road, 74800 Karachi, Pakistan. [Aban, Muniba] Baqai Medical University, Department of PathologyKarachi, Pakistan. [Iqbal, Waseem] Baqai Medical University, Department of PathologyKarachi, Pakistan. [Pervez, Shahid] The Aga Khan University, Department of Pathology and Microbiology, Stadium Road, 74800 Karachi, Pakistan. RP Pervez, Sh (reprint author), The Aga Khan University, Department of Pathology and Microbiology, 74800 Karachi, Pakistan. EM shahid.pervez@aku.edu CR Rolland PH, Martin PM, Jacquemier J, Rolland AM, Toga M, 1980, Prostaglandin in human breast cancer: Evidence suggesting that an elevated prostaglandin production is a marker of high metastatic potential for neoplastic cells. J Natl Cancer Inst 64:1061–1070 Williams CS, DuBois RN, 1996, Prostaglandin endoperoxide synthase: why two isoforms? Am J Physiol 270:G393–400 O’Neill GP, Ford-Hutchinson AW, 1993, Expression of mRNA for cyclooxygenase-1 and cyclooxygenase-2 in human tissues. FEBS Lett 330:156–160 Herschman HR, 1994, Regulation of prostaglandin synthase-1 and prostaglandin synthase-2. Cancer Metastasis Rev 13:241–256 Bennett A, Charlier EM, McDonald AM, Simpson JS, Stamford IF, Zebro T, 1977, Prostaglandins and breast cancer. Lancet 2:624–626 Half E, TangXM, Gwyn K, Sahin A,Wathen K, Sinicrope FA, 2002, Cyclooxygenase-2 expression in human breast cancers and adjacent ductal carcinoma in situ. Cancer Res 62:1676–1681 Hwang D, Scollard D, Byrne J, Levine E, 1998, Expression of cyclooxygenase-1 and cyclooxygenase-2 in human breast cancer. J Natl Cancer Inst 90:455–460 Gong Z, Bostick RM, Xie D, Hurley TG, Deng Z, Dixon DA et al, 2009, Genetic polymorphisms in the cyclooxygenase-1 and cyclooxygenase-2 genes and risk of colorectal adenoma. Int J Colorectal Dis 24:647–654 Hu HM, Kuo CH, Lee CH, Wu IC, Lee KW, Lee JM, et al, 2009, Polymorphism in COX-2 modifies the inverse association between Helicobacter pylori seropositivity and esophageal squamous cell carcinoma risk in Taiwan: a case control study. BMC Gastroenterol 9:37 Hamajima N, Takezaki T,Matsuo K, Saito T, InoueM, Hirai T, Kato T, Ozeki J, Tajima K, 2001, Genotype Frequencies of Cyclooxygenase 2, COX2, Rare Polymorphisms for Japanese with and without Colorectal Cancer. Asian Pac J Cancer Prev 2:57–62 Appleby SB, Ristimaki A, Neilson K, Narko K, Hla T, 1994, Structure of the human cyclo-oxy genase-2 gene. Biochem J 302:723–727 Papafili A, Hill MR, Brull DJ, McAnulty RJ, Marshall RP, Humphries SE et al, 2002, Common promoter variant in cyclooxygenase-2 represses gene expression: evidence of role in acute-phase inflammatory response. Arterioscler Thromb Vasc Biol 22:1631–1636 Zhang X, Miao X, Tan W, Ning B, Liu Z, Hong Y et al, 2005, Identification of functional genetic variants in cyclooxygenase-2 and their association with risk of esophageal cancer. Gastroenterology 129:565–576 Szezekilik W, Sanak M, Szezeklik A, 2004, Functional effects and gender association of COX-2 gene polymorphism G-765C in bronchial asthama. J Allergy Clin Immunol 114:248–253 Zang X,Miao X, TanW, Ning B, Liu Z, HongY, 2005, Identification of functional genetic variants in cyclooxygenase-2 and their association with risk of oseophageal cancer. Gastroenterolgy 129:565–576 Bhurgri Y, 2004, The role of cancer registration in National Cancer Control Pakistan. J Pak Med Assoc 54:402–404 Pereira C, Sousa H, Ferreira P, Fragoso M, Moreira-Dias L, Lopes C et al, 2006, 765G>C COX-2 polymorphism may be a susceptibility marker for gastric adenocarcinoma in patients with atrophy or intestinal metaplasia. World J Gastroenterol 12:5473–5478 Upadhyay R, Jain M, Kumar S, Ghoshal UC, Mittal B, 2009, Functional polymorphisms of cyclooxygenase-2, COX-2, gene and risk for esophageal squamous cell carcinoma. Mutat Res 663:52–59 Piranda DN, Festa-Vasconcellos JS, Amaral LM, Bergmann A, Vianna-Jorge R, 2010, Polymorphisms in regulatory regions of cyclooxygenase-2 gene and breast cancer risk in Brazilians: a case– control study. BMC Cancer 10:613 Ke-Da Y, Chen AX, Yang C, Qiu LX, Fan L, Xu WH et al, 2010, Current evidence on the relationship between polymorphisms in the COX-2 gene and breast cancer risk: a meta-analysis. Breast Cancer Res Treat 122:251–257 Zhu W, Wei BB, Shan X, Liu P, 2010, 765G>C and 8473 T>C polymorphisms of COX-2 and cancer risk: a meta-analysis based on 33 case–control studies. Mol Biol Rep 37:277–288 Ahmed F, 1996, Pakistan: Ethnic fragmentation or national integration. Pak Dev Rev 35:631–645 Gao J, Ke Q, Ma HX,Wang Y, Zhou Y, Hu ZB, Zhai XJ,Wang XC, Qing JW, Chen WS et al, 2007, Functional polymorphisms in the cyclooxygenase 2, COX-2, gene and risk of breast cancer in a Chinese population. J Toxicol Environ Health A 70:908–915 Dossus L, Kaaks R, Canzian F, Albanes D, Berndt SI, Boeing H et al, 2010, PTGS2 and IL6 genetic variation and risk of breast and prostate cancer: results from the Breast and Prostate Cancer Cohort Consortium, BPC3). Carcinogenesis 31:455–461 Wang X-f, Huang M-z, Zhang X-w, Hua R-x, Guo W-j, COX-2- 765G, 2013, C Polymorphism Increases the Risk of Cancer: AMeta- Analysis. PLoS ONE 8(9):e73213., DOI 10.1371/journal.pone. 0073213 Brasky TM, Bonner MR, Moysich KB, Ambrosone CB, Nie J, Tao MH et al, 2011, Genetic variant in Cox-2, Non-steroidal antiinflammatory drug and breast cancer risk in the Western New York Exposures and Breast Cancer, WEB, Study. Breast Cancer Res Treat 126:157–165 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 97 EP 101 DI 10.1007/s12253-014-9792-8 PG 5 ER PT J AU Pereira, LD dos Santos Ferreira, CA Faria, PdG Kwee, KJ AF Pereira, Lima Debora dos Santos Ferreira, Carolina Ana Faria, Pinto de Giselle Kwee, Kiemlian Jolie TI Autophagy Interplays with Apoptosis and Cell Cycle Regulation in the Growth Inhibiting Effect of Trisenox in HEP-2, a Laryngeal Squamous Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Laryngeal squamous cancer; Trisenox; L-buthionine sulfoximine; Autophagy; Apoptosis ID Laryngeal squamous cancer; Trisenox; L-buthionine sulfoximine; Autophagy; Apoptosis AB Laryngeal squamous cell carcinoma (LSCC) is the most common among several types of head and neck cancers. Current treatments have a poor effect on early and advanced cases, and further investigations for novel agents against LSCCs are desirable. In this study, we elucidate the cytotoxic enhancing effect of arsenic trioxide (As2O3) combined with L-buthionine sulfoximine (BSO) in LSCC. The effect of BSO with As2O3 or Cisplatin (CDDP) on cell viability was examined using 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). The reactive oxygen species (ROS) levels, cell cycle, and apoptosis were measured by flow cytometry using 2′,7′-dichlorodihydrofluorescein diacetate (DCFH-DA), propidium iodide (PI) and annexin V/PI. The acidic vacuolar organelles were visualized by fluorescence microscope and quantified using flow cytometry. Neither CDDP nor As2O3 when used alone reduced the cell viability. BSO was found to enhance only As2O3 sensitivity, leading to G2/M arrest and autophagy with no correlation of ROS induction. This result suggests that modulation of glutathione enhances autophagy, which interplays with apoptosis. In this study, we obtained initial preclinical evidence for the potential efficacy of these drugs in a combined therapy protocol. C1 [Pereira, Lima Debora] A. C. Camargo Cancer Hospital, Department of StomatologySao Paulo, Brazil. [dos Santos Ferreira, Carolina Ana] Instituto Nacional de Cancer Jose Alencar Gomes da Silva (INCA), Programa de Pos-Graduacao em Oncologia do Instituto Nacional de Cancer (PPGO-INCA)Rio de Janeiro, Brazil. [Faria, Pinto de Giselle] Universidade Federal da Bahia (UFBA), Instituto de Ciencias da Saude (ICS), Departamento de BiorregulacaoBahia, Brazil. [Kwee, Kiemlian Jolie] INCA, Coordenacao de PesquisaRio de Janeiro, Brazil. RP Kwee, KJ (reprint author), INCA, Coordenacao de Pesquisa, Rio de Janeiro, Brazil. EM jkwee@inca.gov.br CR Khariwala SS, Hatsukami D, Hecht SS, 2012, Tobacco carcinogen metabolites and DNA adducts as biomarkers in head and neck cancer: Potential screening tools and prognostic indicators. Head and Neck 34:441–447 Marur S, Forastiere AA, 2010, Challenges of integrating chemotherapy and targeted therapy with radiation in locally advanced head and neck squamous cell cancer. Curr Opin Oncol 22:206–211 CancelaMC,Voti L, Guerra-YiM, Chapuis F,Mazuir M, CuradoMP, 2010, Oral cavity cancer in developed and in developing countries: Population-Based Incidence. Head and Neck 32:357–367 Schmitz S, Machiels JP, 2010, Molecular biology of squamous cell carcinoma of the head and neck: relevance and therapeutic implications. Expert Rev. Anticancer Ther 10:1471–1484 GendenEM, Ferlito A, RinaldoA, Silver CE, Fagan JJ, Suarez C et al, 2008, Recent changes in the treatment of patients with advanced laryngeal cancer. Head and Neck 30:103–110 Ma X, Herbert Y, 2007, Global Burden of Cancer. Yale J Biol Med 79:85–94 Holsinger FC, Lin HY, Bassot V, Laccourreye O, 2009, Platin-based exclusive chemotherapy for selected patients with squamous cell carcinoma of the larynx and pharynx. Cancer 115:3909–3918 HoffmanHT, Porter K,Karnell LH, Cooper JS,Weber RS, Langer CJ et al, 2006, Laryngeal cancer in the United States: Changes in demographics, patterns of care, and survival. Laryngoscope 116:1–13 Shi N, Han X, Yu W, Wang L, Lu A, 2012, Adoption in China of clinical practice guidelines for hypertension using traditional Chinese medical approaches: a literature review based on clinical studies. J Altern Complement Med 18:1–8 Chen SJ, Zhou GB, Zhang XW, Mao JH, de The H, Chen Z, 2011, From an old remedy to a magic bullet: molecular mechanisms underlying the therapeutic effects of arsenic in fighting leukemia. Blood 117:6425–6437 Soignet SL, Maslak P,Wang ZG, Jhanwar S, Calleja E, Dardashti LJ et al, 1998, Complete remission after treatment of acute promyelocytic leukemia with arsenic trioxide. N Engl J Med 339: 1341–1348 Zhang TD, Chen GQ,Wang ZG,Wang ZY, Chen SJ, Chen Z, 2001, Arsenic trioxide, a therapeutic agent for APL. Oncogene 20:7146– 7153 Sahu GR, Jena RK, 2005, Significance of intracellular arsenic trioxide for therapeutic response in acute promyelocytic leukemia. Am J Hematol 78:113–116 Konig H, Hartel N, Schultheis B, SchatzM, Lorentz C,Melo JVet al, 2007, Enhanced Bcr-Abl-specific antileukemic activity of arsenic trioxide through glutathione-depletion in imatinib-resistant cells. Haematologica 92:838–841 Chiu HW, Ho YS,Wang YJ, 2011, Arsenic trioxide induces autophagy and apoptosis in human glioma cells in vitro and in vivo through downregulation of survivin. J Mol Med 89:927–941 Dizaji MZ, Malehmir M, Ghavamzadeh A, Alimoghaddam K, Ghaffari SH, 2012, Synergistic effects of arsenic trioxide and silibinin on apoptosis and invasion in human glioblastoma U87MG cell line. Neurochem Res 37:370–380 Smith D, Patel S, Raffoul F, Haller E, MillsGB, NanjundanM(2010, Arsenic trioxide induces a beclin-1 independent autophagic pathway via modulation of snon/skil expression in ovarian carcinoma cells. Cell Death Differ 17:1867–1881 Liu N, Tai S, Ding B, Thor RK, Bhuta S, Sun Yet al, 2012, Arsenic trioxide synergizes with everolimus, Rad001, to induce cytotoxicity of ovarian cancer cells through increased autophagy and apoptosis. Endocrine-Related Cancer 19:711–723 Kumar P, Gao Q, Ning Y, Wang Z, Krebsbach PH, Polverini PJ, 2008, Arsenic trioxide enhances the therapeutic efficacy of radiation treatment of oral squamous carcinoma while protecting bone. Mol Cancer Ther 7:2060–2069 Zhang X, Su Y, Zhang M, Sun Z, 2012, Opposite effects of arsenic trioxide on the Nrf2 pathway in oral squamous cell carcinoma in vitro and in vivo. Cancer Lett 318:93–98 Diepart C, Karroum O,Magat J, Feron O, Verrax J, Calderon PB et al, 2012, Arsenic trioxide treatment decreases the oxygen consumption rate of tumor cells and radiosensitizes solid tumors. Cancer Res 72: 482–490 Chen G, Wang K, Yang BY, Tang B, Chen JX, Hua ZC, 2012, Synergistic antitumor activity of oridonin and arsenic trioxide on hepatocellular carcinoma cells. Int J Oncol 40:139–147 Li H, Zhu X, Zhang Y, Chen H, 2009, Arsenic trioxide exerts synergistic effects with cisplatin on non-small cell lung cancer cells via apoptosis induction. J Exp Clin Cancer Res 28:110 Brown M, Bellon M, Nicot C, 2007, Emodin and DHA potently increase arsenic trioxide interferon-α-induced cell death of HTLV-Itransformed cells by generation of reactive oxygen species and inhibition of AKT and AP-1. Blood 109:1653–1659 You BR, Park WH, 2012, Arsenic trioxide induces human pulmonary fibroblast cell death via increasing ROS levels and GSH depletion. Oncol Rep 28:749–757 Sun RC, Board PG, Blackburn AC, 2011, Targetingmetabolismwith arsenic trioxide and dichloroacetate in breast cancer cells. Mol Cancer 10:142 Xie LX, Lin XH, Li DR, Chen JY, Hong CQ, Du CW, 2007, Synergistic therapeutic effect of arsenic trioxide and radiotherapy in BALB/C nude mice bearing nasopharyngeal carcinoma xenografts. Exp Oncol 29:45–48 Sides MD, Sosulski ML, Luo F, Lin Z, Flemington EK, Lasky JA, 2013, Co-treatment with arsenic trioxide and ganciclovir reduces tumor volume in murine xenograft model of nasopharyngeal carcinoma. Virol J 10:1–7 Kotowski U, Heiduschka G, Brunner M, Erovic BM, Martinek H, Thurnher D, 2012, Arsenic trioxide enhances the cytotoxic effect of cisplatin in head and neck squamous cell carcinoma cell lines. Oncol Lett 3:1326–1330 Brozovic A, Majhen D, Roje V, Mikac N, Jakopec S, Fritz G et al, 2008, αvβ3 Integrin-mediated drug resistance in human laryngeal carcinoma cells is caused by glutathione-dependent elimination of drug-induced reactive oxidative species.Mol Pharmacol 74:298–306 Yang X, 2012, Proteosome inhibitor bortezomi-induced the apoptosis of laryngeal squamous cell carcinoma Hep-2 cell line disrupting redox equilibrium. Biomed Pharmacother 66:607–611 SobhakumariA, Love-Homan L, Fletcher EVM,Martin SM, Parsons AD, Spitz DR, 2012, Susceptibility of human head and neck cancer cells to combined inhibition of glutathione and thioredoxin metabolism. PLoS ONE 7:e48175 Mosmann T, 1983, Rapid colorimetric assay for cellular growth and survival: application to proliferation and citotoxicity assays. J Immunol Methods 65(1–2):55–63 Griffith OW, Meister A, 1979, Potent and specific inhibition of glutathione synthesis by buthionine sulfoximine, S-n-butyl homocysteine sulfoximine). J Biol Chem 254(16):7558–7560 Fischel J-L, Formento P, Milano G, 2005, Epidermal growth factor receptor double targeting by a tyrosine kinase inhibitor, Iressa, and a monoclonal antibody, Cetuximab). Impact on cell growth and molecular factors. Br J Cancer 92(6):1063–1068 Chou T, Talalay P, 1984, Quantitative analysis of dose-effects relationships: the combined effects of multiple drugs or enzyme inhibtors. Adv Enzym Regul 22:27–55 Kwee JK, Luque DG, Ferreira ACS, Vasconcelos FC, Silva KL, Klumb CE, Maia RC, 2008, Modulation of reactive oxygen species by antioxidants in chronic myeloid leukemia cells enhances imatinib sensitivity through surviving downregulation. Anti-Cancer Drugs 19: 975–981 Dos Santos Ferreira AC, Fernandes RA, Kwee JK, Klumb CE, 2012, Histone deacetylase inhibitor potentiates chemotherapy-induced apoptosis through Bim upregulation in Burkitt’s lymphoma cells. J Cancer Res Clin Oncol 138:317–325 Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo- Arozena A, Adeli K et al, 2012, Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy 8:445–544 Jiang H, White EJ, Conrad C, Gomez-Manzano C, Fueyo J, 2009, Autophagy pathways in glioblastoma. Methods Enzymol 453:273– 286 Burtness B, Goldwasser MA, Flood W, Mattar B, Forastiere AA, 2005, Phase III randomized trial of cisplatin plus placebo compared with cisplatin plus cetuximab in metastatic/recurrent head and neck cancer: an Eastern Cooperative Oncology Group study. J Clin Oncol 23:8646–8654 Razak AR, Siu LL, Le Tourneau C, 2010, Molecular targeted therapies in all histologies of head and neck cancers: an update. Curr Opin Oncol 22:212–220 Specenier P, Vermorken JB, 2013, Cetuximab: its unique place in head and neck cancer treatment. Biologics 7:77–90 Bailey HH, Mulcahy RT, Tutsch KD, Arzoomanian RZ, Alberti D, Tombes MB et al, 1994, Phase 1 clinical trial of intravenous Lbuthionine sulfoximine and melphalan: an attempt at modulation of glutathione. J Clin Oncol 12:194–205 O’Dwyer PJ, Hamilton TC, LaCreta FP, Gallo JM, Kilpatrick D, Halbherr T et al, 1996, Phase I trial of buthionine sulfoximine in combination with melphalan in patients with cancer. J Clin Oncol 14: 249–256 Bailey HH, Ripple G, Tutsch KD, Arzoomanian RZ, Alberti D, Feierabend C et al, 1997, Phase I study of continuous-infusion L-S, R-buthionine sulfoximine with intravenous melphalan. J Natl Cancer Inst 89:1789–1796 Bailey HH, 1998, L-S, R-Buthionine sulfoximine: historical development and clinical issues. Chem Biol Interact 111–112: 239–254 Wu JH, Batist G, 2013, Glutathione and glutathione analogues; therapeutic potentials. Biochim Biophys Acta 1830:3350–3353 Ramanathan B, Jan KY, Chen CH, Hour TC, Yu HJ, Pu YS, 2005, Resistance to paclitaxel is proportional to cellular total antioxidant capacity. Cancer Res 65:8455–8460 Ong PS, Chan SY, Ho PC, 2011, Differential augmentative effects of buthionine sulfoximine and ascorbic acid in As2O3-induced ovarian cancer cell death: oxidative stress-independent and –dependent cytotoxic potentiation. Int J Oncol 38:1731–1739 Giansanti V, Torriglia A, Scovassi AI, 2011, Conversation between apoptosis and autophagy: “Is it your turn or mine?”. Apoptosis 16: 321–333 Huang L,Wang S, Li SS,YangXM(2013, Prognostic significance of beclin-1 expression in laryngeal squamous cell carcinoma. Pathol Oncol Res 2013 in press Church DN, Talbot DC, 2012, Survivin in solid tumors: rationale for development of inhibitors. Curr Oncol Rep 14:120–128 Thomas DJ, 2010, Arsenolysis and thiol-dependent arsenate reduction. Toxicol Sci 117:249–252 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 103 EP 111 DI 10.1007/s12253-014-9794-6 PG 9 ER PT J AU Basaran, D Salman, CM Boyraz, G Selcuk, I Usubutun, A Ozgul, N Yuce, K AF Basaran, Derman Salman, Coskun Mehmet Boyraz, Gokhan Selcuk, Ilker Usubutun, Alp Ozgul, Nejat Yuce, Kunter TI Accuracy of Intraoperative Frozen Section in the Evaluation of Patients with Adnexal Mass: Retrospective Analysis of 748 Cases with Multivariate Regression Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Frozen section; Ovarian tumor; Adnexalmass; Ovarian cancer; Borderline ovarian tumor ID Frozen section; Ovarian tumor; Adnexalmass; Ovarian cancer; Borderline ovarian tumor AB Objective: To evaluate the accuracy of intraoperative frozen section in the evaluation of patients with adnexal mass and to define the clinicopathological factors associated with misdiagnosis during frozen section evaluation. Methods: The clinicopathological data of patients who underwent exploratory laparotomy for adnexal mass were reviewed. Results of the intraoperative frozen section and permanent histology reports were compared. Univariate and multivariate analyses were performed to reveal factors associated with misdiagnosis. Results: The study group consisted of 748 patients. Of these patients, 509 (68.0 %) had benign, 43 (5.7 %) had borderline, 196 (26.2 %) had malignant histological diagnosis at permanent section. The overall agreement between intraoperative frozen section and permanent pathology was 96.8 %. Twenty four out of 745 cases (3.8 %) were misdiagnosed by frozen section. Univariate analysis showed that borderline histology (p<0.0001) and tumor size larger than 10 cm (p=0.012) were associated with misdiagnosis. According to multivariate analysis, borderline histology (OR: 22.6, p<0.0001) was the only independent predictor for misdiagnosis during frozen examination. Conclusion: The frozen section evaluation of the adnexal mass is highly accurate. However, tumor size greater than 10 cm and borderline histology are the factors that adversely influence the accuracy of intraoperative frozen section. Clinicians must be aware of these pitfalls during intraoperative decision making following frozen section report. C1 [Basaran, Derman] Hacettepe University, Faculty of Medicine, Department of Obstetrics and Gynecology, Sihhiye, 06100 Ankara, Turkey. [Salman, Coskun Mehmet] Hacettepe University, Faculty of Medicine, Department of Obstetrics and Gynecology, Sihhiye, 06100 Ankara, Turkey. [Boyraz, Gokhan] Hacettepe University, Faculty of Medicine, Department of Obstetrics and Gynecology, Sihhiye, 06100 Ankara, Turkey. [Selcuk, Ilker] Hacettepe University, Faculty of Medicine, Department of Obstetrics and Gynecology, Sihhiye, 06100 Ankara, Turkey. [Usubutun, Alp] Hacettepe University, Faculty of Medicine, Department of PathologyAnkara, Turkey. [Ozgul, Nejat] Hacettepe University, Faculty of Medicine, Department of Obstetrics and Gynecology, Sihhiye, 06100 Ankara, Turkey. [Yuce, Kunter] Hacettepe University, Faculty of Medicine, Department of Obstetrics and Gynecology, Sihhiye, 06100 Ankara, Turkey. RP Salman, CM (reprint author), Hacettepe University, Faculty of Medicine, Department of Obstetrics and Gynecology, 06100 Ankara, Turkey. EM csalman@hacettepe.edu.tr CR Myers ER, Bastian LA, Havrilesky LJ, Kulasingam SL, Terplan MS, Cline KE, Gray RN, McCrory DC, 2006, Management of adnexal mass. Evid Rep Technol Assess, Full Rep, 130:1–145 Geomini P, Bremer G, Kruitwagen R, Mol BW, 2005, Diagnostic accuracy of frozen section diagnosis of the adnexal mass: a metaanalysis. Gynecol Oncol 96(1):1–9 Brun JL, Cortez A, Rouzier R, Callard P, Bazot M, Uzan S et al, 2008, Factors influencing the use and accuracy of frozen section diagnosis of epithelial ovarian tumors. Am J Obstet Gynecol 199(3): 244 e1–244 e7 Usubutun A, Altinok G, Kucukali T, 1998, The value of intraoperative consultation, frozen section, in the diagnosis of ovarian neoplasms. Acta Obstet Gynecol Scand 77(10):1013–1016 HeatleyMK(2012, A systematic review of papers examining the use of intraoperative frozen section in predicting the final diagnosis of ovarian lesions. Int J Gynecol Pathol 31(2):111–115 Medeiros LR, Rosa DD, Edelweiss MI, Stein AT, Bozzetti MC, Zelmanowicz A et al, 2005, Accuracy of frozen-section analysis in the diagnosis of ovarian tumors: a systematic quantitative review. Int J Gynecol Cancer 15(2):192–202 Geomini PM, Zuurendonk LD, Bremer GL, de Graaff J, Kruitwagen RF, Mol BW, 2005, The impact of size of the adnexal mass on the accuracy of frozen section diagnosis. Gynecol Oncol 99(2):362–366 Bige O, Demir A, Saygili U, Gode F, Uslu T, KoyuncuogluM(2011, Frozen section diagnoses of 578 ovarian tumors made by pathologists with and without expertise on gynecologic pathology. Gynecol Oncol 123(1):43–46 Taskiran C, Erdem O, Onan A, Bozkurt N, Yaman-Tunc S, Ataoglu Oet al, 2008, The role of frozen section evaluation in the diagnosis of adnexal mass. Int J Gynecol Cancer 18(2):235–240 Tangjitgamol S, Jesadapatrakul S, Manusirivithaya S, Sheanakul C, 2004, Accuracy of frozen section in diagnosis of ovarian mass. Int J Gynecol Cancer 14(2):212–219 Song T, Choi CH, Kim HJ, Kim MK, Kim TJ, Lee JW et al, 2011, Accuracy of frozen section diagnosis of borderline ovarian tumors. Gynecol Oncol 122(1):127–131 Menzin AW, Rubin SC, Noumoff JS, LiVolsi VA, 1995, The accuracy of a frozen section diagnosis of borderline ovarian malignancy. Gynecol Oncol 59(2):183–185 Houck K, Nikrui N, Duska L, Chang Y, Fuller AF, Bell D et al, 2000, Borderline tumors of the ovary: correlation of frozen and permanent histopathologic diagnosis. Obstet Gynecol 95(6 Pt 1):839–843 Ilvan S, Ramazanoglu R, Ulker Akyildiz E, Calay Z, Bese T, Oruc N, 2005, The accuracy of frozen section, intraoperative consultation, in the diagnosis of ovarian masses. Gynecol Oncol 97(2):395–399 Stewart CJ, Brennan BA, Hammond IG, Leung YC, McCartney AJ, 2006, Intraoperative assessment of ovarian tumors: a 5-year review with assessment of discrepant diagnostic cases. Int J Gynecol Pathol 25(3):216–222 Baker P, Oliva E, 2008, A practical approach to intraoperative consultation in gynecological pathology. Int J Gynecol Pathol 27, 3):353–365 Stewart CJ, Brennan BA, Koay E, Naran A, Ruba S, 2010, Value of cytology in the intraoperative assessment of ovarian tumors: a review of 402 cases and comparison with frozen section diagnosis. Cancer Cytopathol 118(3):127–136 Tempfer CB, Polterauer S, Bentz EK, Reinthaller A, Hefler LA, 2007, Accuracy of intraoperative frozen section analysis in borderline tumors of the ovary: a retrospective analysis of 96 cases and review of the literature. Gynecol Oncol 107(2):248–252 Puls L, Heidtman E, Hunter JE, Crane M, Stafford J, 1997, The accuracy of frozen section by tumor weight for ovarian epithelial neoplasms. Gynecol Oncol 67(1):16–19 WangKG, Chen TC,Wang TY, Yang YC, Su TH, 1998, Accuracy of frozen section diagnosis in gynecology. Gynecol Oncol 70(1):105– 110 Gol M, Baloglu A, Yigit S, Dogan M, Aydin C, Yensel U, 2003, Accuracy of frozen section diagnosis in ovarian tumors: is there a change in the course of time? Int J Gynecol Cancer 13(5):593–597 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 113 EP 118 DI 10.1007/s12253-014-9795-5 PG 6 ER PT J AU Acikalin, A Gumurdulu, D Bagir, KE Torun, G Guzel, BA Zeren, H Vardar, AM AF Acikalin, Arbil Gumurdulu, Derya Bagir, Kilic Emine Torun, Goncagul Guzel, Baris Ahmet Zeren, Handan Vardar, Ali Mehmet TI The Guidance of Intraoperative Frozen Section For Staging Surgery in Endometrial Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial cancer; Intraoperative frozen section; Staging surgery ID Endometrial cancer; Intraoperative frozen section; Staging surgery AB The objective of this study was to evaluate the reliability of an intraoperative frozen section during the endometrial carcinoma staging surgery procedure. The paraffin section reports of 291 cases with endometrial carcinoma were compared with intraoperative frozen section reports, which were diagnosed in the Pathology Department of Cukurova University, Medical Faculty between June 2006 and December 2012. The reports were reviewed for diagnostic accuracy of the frozen section in terms of histological subtype, grade, and myometrial invasion. Concordance values between frozen and paraffin section reports were 86, 84.3, and 91.6 % for histological subtype, grade, and myometrial invasion, respectively. When collectively evaluated, two (0.7 %) of 291 patients were inappropriately operated on due to frozen section reports. Intraoperative frozen section is a reliable guide for surgeons to evaluate the risk group of patients with endometrial cancer and prevent an unnecessary staging surgery operation. C1 [Acikalin, Arbil] Cukurova University, School of Medicine, Pathology Department, Saricam, 01330 Adana, Turkey. [Gumurdulu, Derya] Cukurova University, School of Medicine, Pathology Department, Saricam, 01330 Adana, Turkey. [Bagir, Kilic Emine] Cukurova University, School of Medicine, Pathology Department, Saricam, 01330 Adana, Turkey. [Torun, Goncagul] Cukurova University, School of Medicine, Pathology Department, Saricam, 01330 Adana, Turkey. [Guzel, Baris Ahmet] Cukurova University, School of Medicine, Obstetrics and Gynecology DepartmentAdana, Turkey. [Zeren, Handan] Acibadem University, School of Medicine, Pathology DepartmentIstanbul, Turkey. [Vardar, Ali Mehmet] Cukurova University, School of Medicine, Obstetrics and Gynecology DepartmentAdana, Turkey. RP Acikalin, A (reprint author), Cukurova University, School of Medicine, Pathology Department, 01330 Adana, Turkey. EM arbilavci@yahoo.com CR Creasman WT, Morrow CP, Bundy BN, Homesley HD, Graham JE, Heller PB, 1987, Surgical pathologic spread patterns of endometrial cancer. A Gynecologic Oncology Group study. Cancer 60(8 Suppl): 2035–2041 Ugaki H, Kimura T, Miyatake T, Ueda Y, Yoshino K, Matsuzaki S, Fujita M, Kimura T, Morii E, Enomoto T, 2011, Intraoperative frozen section assessment of myometrial invasion and histology of endometrial cancer using the revised FIGO staging system. Int J Gynecol Cancer 21(7):1180–1184 ArkoD, Takac I, 2000, High frequency transvaginal ultrasonography in preoperative assessment of myometrial invasion in endometrial cancer. J Ultras Med 19(9):639–643 Kinkel K, Kaji Y, Yu KK, Segal MR, Lu Y, Powell CB, Hricak H, 1999, Radiologic staging in patients with endometrial cancer: a meta-analysis. Radiology 212(3):711–718 Connor JP, Andrews JI, Anderson B, Buller RE, 2000, Computed tomography in endometrial carcinoma. Obstet Gynecol 95(5):692– 696., DOI 10.1016/S0029-7844(99)00626-2 Quinlivan JA, Petersen RW, Nicklin JL, 2001, Accuracy of frozen section for the operative management of endometrial cancer. Brit J Obstet Gynaecol 108(8):798–803., DOI 10.1111/J.1471-0528.2001. 00196.X Case AS, Rocconi RP, Straughn JM Jr, Conner M, Novak L, Wang W, HuhWK, 2006, A prospective blinded evaluation of the accuracy of frozen section for the surgical management of endometrial cancer. Obstet Gynecol 108(6):1375–1379., DOI 10.1097/01.AOG. 0000245444.14015.00 Frumovitz M, Slomovitz BM, Singh DK, Broaddus RR, Abrams J, Sun CC, Bevers M, Bodurka DC, 2004, Frozen section analyses as predictors of lymphatic spread in patients with early-stage uterine cancer. J Am Coll Surg 199(3):388–393., DOI 10.1016/J.Jamcollsurg. 2004.05.258 Kumar S, Bandyopadhyay S, Semaan A, Shah JP, Mahdi H, Morris R, Munkarah A, Ali-Fehmi R, 2011, The role of frozen section in surgical staging of low risk endometrial cancer. Plos One 6(9)., DOI ARTN e21912 DOI 10.1371/journal.pone.0021912 Kumar S, Medeiros F, Dowdy SC, Keeney GL, Bakkum-Gamez JN, Podratz KC, ClibyWA, Mariani A, 2012, A prospective assessment of the reliability of frozen section to direct intraoperative decision making in endometrial cancer. Gynecol Oncol 127(3):525–531., DOI 10.1016/j.ygyno.2012.08.024 Turan T, Oguz E, Unlubilgin E, Tulunay G, Boran N, Demir OF, Kose MF, 2013, Accuracy of frozen-section examination for myometrial invasion and grade in endometrial cancer. Eur J Obstet Gyn R B 167(1):90–95., DOI Doi 10.1016/J.Ejogrb.2012.11.004 Sanjuan A, Cobo T, Pahisa J, Escaramis G, Ordi J, Ayuso JR, Garcia S, Hernandez S, Torne A, Roman SM, Lejarcegui JA, Vanrell JA, 2006, Preoperative and intraoperative assessment of myometrial invasion and histologic grade in endometrial cancer: role of magnetic resonance imaging and frozen section. Int J Gynecol Cancer 16(1):385–390., DOI 10.1111/J.1525- 1438.2006.00414.X Wang XP, Zhang H, Di W, Li WP, 2009, Clinical factors affecting the diagnostic accuracy of assessing dilation and curettage vs frozen section specimens for histologic grade and depth of myometrial invasion in endometrial carcinoma. Am J Obstet Gynecol 201(2)., DOI ARTN 194.e1 DOI 10.1016/ j.ajog.2009.05.003 Ozdemir S, Celik C, Emlik D, Kiresi D, Esen H, 2009, Assessment of myometrial invasion in endometrial cancer by transvaginal sonography, doppler ultrasonography, magnetic resonance imaging and frozen section. Int J Gynecol Cancer 19(6):1085–1090., DOI 10. 1111/Igc.0b013e3181ad3eb6 Maneschi F, Nardi S, Sarno M, Manicone AM, Perugini A, Partenzi A, Lambiase A, Pericoli N, Maiorca A, Travaini S, 2007, Endometrial carcinoma. Intraoperative evaluation of myometrial invasion. A prospective study. Int J Gynecol Cancer 17(4):940 Obrzut B, Obrzut M, Skret-Magierlo J, Skret A, Ulman D, Krol P, Zmuda M, 2008, Value of the intraoperative assessment of the depth ofmyometrial invasion in endometrial carcinoma. Ginekol Pol 79(6): 404–409 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 119 EP 122 DI 10.1007/s12253-014-9796-4 PG 4 ER PT J AU Jenner, S Wiedorn, HK Techel, D AF Jenner, Stefan Wiedorn, Herrmann Klaus Techel, Dieter TI Development of a DUSP9 Methylation Screening Assay SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DUSP9; Colorectal cancer; Promoter methylation; CIMP; Pyrosequencing ID DUSP9; Colorectal cancer; Promoter methylation; CIMP; Pyrosequencing AB A methylation screening assay for DUSP9 (dualspecificity phosphatase 9) has been developed and applied on 79 FFPE samples from patients with colorectal cancer (CRC) and 22 corresponding tumor free colon samples in this study. Quantitative pyrosequencing was used for the determination of the methylation in the promoter CpG island, including 83 CpG motifs. In this way, the methylation pattern of the 11 tumor samples with the weakest and the strongest methylation could be identified and were compared to their corresponding tumor free colon samples. Forty six percent of the weakly methylated samples showed no significant difference to their tumor free counterparts, whereas in 27 % of the cases an increased or reduced methylation was detectable. For the strongly methylated tumor samples only 18 % showed no significant difference to their tumor free counterparts, whereas 82 % were significantly stronger methylated. In CRC, the aberrant promoter methylation of tumor suppressor genes is one aspect that defines the CpG island methylator phenoptype (CIMP) and is frequently observed in a subpopulation of cases. Patients harboring a CIMP phenotype often show additional clinicopathological characteristics, the so called CIMP features. Interestingly, no CIMP features were found for the weakly methylated samples analyzed in this study but could be seen in 82 % of the strongly methylated cases, indicating a possible use for DUSP9 as CIMP marker. C1 [Jenner, Stefan] Katharinen Hospital Stuttgart, Department of Pathology, Division of Molecular Pathology, Kriegsbergstr. 60, 70174 Stuttgart, Germany. [Wiedorn, Herrmann Klaus] Katharinen Hospital Stuttgart, Department of Pathology, Division of Molecular Pathology, Kriegsbergstr. 60, 70174 Stuttgart, Germany. [Techel, Dieter] Katharinen Hospital Stuttgart, Department of Pathology, Division of Molecular Pathology, Kriegsbergstr. 60, 70174 Stuttgart, Germany. RP Jenner, S (reprint author), Katharinen Hospital Stuttgart, Department of Pathology, Division of Molecular Pathology, 70174 Stuttgart, Germany. EM S.Jenner@Klinikum-Stuttgart.de CR Ferlay J, Shin HR, Bray F, Forman D,Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127(12):2893–2917 Cappell MS, 2005, The pathophysiology, clinical presentation, and diagnosis of colon cancer and adenomatous polyps. Med Clin N Am 89(1):1–42 Wong JJ, Hawkins NJ, Ward RL, 2007, Colorectal cancer: a model for epigenetic tumorigenesis. Gut 56(1):140–148 Feinberg AP, Vogelstein B, 1983, Hypomethylation of ras oncogenes in primary human cancers. Biochem Biophys Res Commun 111(1): 47–54 Rivera RM, Bennett LB, 2010, Epigenetics in humans: an overview. Curr Opin Endocrinol Diabetes Obes 17(6):493–499 Esteller MD, 2008, Epigenetics in cancer. N Engl J Med 358(11): 1148–1159 Tommasi S, Pinto R, Petriella D, Pilato B, Lacalamita R, Santini D, Zito F et al, 2011)Oncosuppressormethylation: a possible key role in colon metastatic progression. J Cell Physiol 226(7):1934–1939 Ottini L, Falchetti M, Lupi R, Rizzolo P, Agnese V, Colucci G et al, 2006, Patterns of genomic instability in gastric cancer: clinical implications and perspectives. Ann Oncol 17(Suppl 7):vii97–vii102 Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP, 1999, CpG island methylator phenotype in colorectal cancers. Proc Natl Acad Sci U S A 96(15):8681–8686 Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA et al, 2006, CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 38(7):787–793 Ogino S, Odze RD, Kawasaki T, BrahmandamM, Kirkner GJ, Laird PWet al, 2006, Correlation of pathologic features with CpG island methylator phenotype, CIMP, by quantitative DNA methylation analysis in colorectal carcinoma. Am J Surg Pathol 30(9):1175–1183 Grady WM, 2007, CIMP and colon cancer gets more complicated. Gut 56(11):1498–1500 Hinoue T,Weisenberger DJ, Pan F, CampanM, KimM, Young J et al, 2009, Analysis of the association between CIMP and BRAF in colorectal cancer by DNA methylation profiling. PLoS One 4(12): e8357 Ogino S, Goel A, 2008, Molecular classification and correlates in colorectal cancer. J Mol Diagn 10(1):13–27 Kambara T, Simms LA,Whitehall VL, Spring KJ,Wynter CV,Walsh MDet al, 2004, BRAF mutation is associated with DNAmethylation in serrated polyps and cancers of the colorectum. Gut 53(8):1137– 1144 MudaM, Boschert U, Smith A, Antonsson B, Gillieron C, Chabert C et al, 1997, Molecular cloning and functional characterization of a novel mitogen-activated protein kinase phosphatase, MKP-4. J Biol Chem 272(8):5141–5151 Bhaduri A, Sowdhamini R, 2003, A genome-wide survey of human tyrosine phosphatases. Protein Eng 16(12):881–888 Jeffrey KL, Camps M, Rommel C, Mackay CR, 2007, Targeting dual-specificity phosphatases: manipulating MAP kinase signalling and immune responses. Nat Rev Drug Discov 6(5):391ff Patterson KI, Brummer T, O’Brien PM, Daly RJ, 2009, Dualspecificity phosphatases: critical regulators with diverse cellular targets. Biochem J 418(3):475–489 Jeong DG, Yoon TS, Jung SK, Park BC, Park H, Ryu SE et al, 2011, Exploring binding sites other than the catalytoc core in the crystal structure of catalytic domain of MKP-4. Acta Crystallogr D Biol Crystallogr 67(Pt 1):25–31 Nunes-Xavier C, Roma-Mateo C, Rios P, Tarrega C, Cejudo-Marin R, Tabernero L et al, 2011, Dual-specificity MAP kinase phosphatases as targets of cancer treatment. Anticancer Agents Med Chem 11(1):109–132 Bermudez O, Pages G, Gimond C, 2010, The dual-specificity MAP kinase phosphatases: critical roles in development and cancer. Am J Physiol Cell Physiol 299(2):C189–C202 Liu Y, Lagowski J, Sundholm A, Sundberg A, Kulesz-Martin M, 2007, Microtubule disruption and tumor suppression by mitogenactivated protein kinase phosphatase 4. Cancer Res 67(22):10711– 10719 Wu S, Wang Y, Sun L, Zhang Z, Jiang Z, Qin Z et al, 2011, Decreased expression of dual-specificity phosphatase 9 is associated with poor prognosis in clear cell renal cell carcinoma. BMC Cancer 11:413 Liu J, Ni W, Xiao M, Jiang F, Ni R, 2013, Decreased expression and prognostic role of mitogen-activated protein kinase phosphatase 4 in hepatocellular carcinoma. J Gastrointest Surg 17(4):756–765 Keyse SM, 2008, Dual-specificity MAP kinase phosphatases, MKPs, and cancer. Cancer Metastasis Rev 27(2):253–261 Sansom OJ, Meniel V, Wilkins JA, Cole AM, Oien KA, Marsh V et al, 2006, Loss of Apc allows phenotypic manifestation of the transforming properties of an endogenous Kras oncogene in vivo. Proc Natl Acad Sci U S A 103(38): 14122–14127 Baldus SE, Schaefer KL, Engers R, Hartleb D, Stoecklein NH, Gabbert HE, 2010, Prevalence and heterogeneity of KRAS, BRAF, and PIK3CA mutations in primary colorectal adenocarcinomas and their corresponding metastases. Clin Cancer Res 16(3):790–799 Dunn J, Baborie A, Alam F, Joyce K, Moxham M, Sibson R et al, 2009, Extent of MGMT promoter methylation correlates with outcome in glioblastomas given temozolomide and radiotherapy. Br J Cancer 101(1):124–131 Kondoh K, Nishida E, 2007, Regulation of MAP kinases by MAP kinase phosphatases. Biochim Biophys Acta 1773(8):1227–1237 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 123 EP 130 DI 10.1007/s12253-014-9797-3 PG 8 ER PT J AU Zhou, L Rui, JA Wang, ShB Chen, ShG Qu, Q AF Zhou, Li Rui, Jing-An Wang, Shao-Bin Chen, Shu-Guang Qu, Qiang TI Clinicopathological Predictors of Poor Survival and Recurrence After Curative Resection in Hepatocellular Carcinoma Without Portal Vein Tumor Thrombosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Portal vein tumor thrombosis; Survival; Recurrence ID Hepatocellular carcinoma; Portal vein tumor thrombosis; Survival; Recurrence AB Many factors associated with long-term outcome in hepatocellular carcinoma (HCC) with portal vein tumor thrombosis (PVTT) were previously identified. However, those in HCC without PVTT have not been elucidated. This study was designed to define the risk factors of poor postsurgical survival and recurrence in this subgroup of HCC. Medical records and follow-up data of consecutive 152 patients with PVTT-absent HCC underwent curative resection were reviewed. The impacts of clinical and pathological variables on patient survival and recurrence were evaluated by univariate and multivariate analyses. It was shown that Edmondson-Steiner grade, TNM stage, microvascular invasion (MVI), satellite nodule, serum AFP level, tumor size and number were significant for tumor-specific and/or tumor-free survival in univariate analysis. Among them, Edmondson- Steiner grade and TNM stage were of independent significances for both, whereas satellite nodule independently predicted tumor-free survival. In Chi-square test, Edmondson- Steiner grade, TNM stage and MVI were significantly related to overall as well as early recurrence. Stepwise logistic regression identified Edmondson-Steiner grade as the single independent predictor of both. To be summarized, variables that are associated with poor prognosis and recurrence in HCC without PVTT are all tumor-related ones. Of these, differentiation degree might be of particular importance. C1 [Zhou, Li] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. [Rui, Jing-An] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. [Wang, Shao-Bin] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. [Chen, Shu-Guang] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. [Qu, Qiang] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. RP Zhou, L (reprint author), Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. EM lizhou02@hotmail.com CR Parkin DM, Pisani P, Ferlay J, 1993, Estimates of the worldwide incidence of eighteen major cancers in 1985. Int J Cancer 54:594– 606 Pisani P, Parkin DM, Bray F, Ferlay J, 1999, Estimates of the worldwide mortality from 25 cancers in 1990. Int J Cancer 83:18–29 Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Poon RT, Fan ST, Lo CM, Ng IO, Liu CL, Lam CM,Wong J, 2001, Improving survival results after resection of hepatocellular carcinoma: a prospective study of 377 patients over 10 years. Ann Surg 234: 63–70 Shimozawa N, Hanazaki K, 2004, Longterm prognosis after hepatic resection for small hepatocellular carcinoma. J Am Coll Surg 198: 356–365 Verhoef C, deMan RA, Zondervan PE, Eijkemans MJ, Tilanus HW, Ijzermans JN, 2004, Good outcomes after resection of large hepatocellular carcinoma in the non-cirrhotic liver. Dig Surg 21:380–386 Tandon P, Garcia-Tsao G, 2009, Prognostic indicators in hepatocellular carcinoma: a systematic review of 72 studies. Liver Int 29:502– 510 Sugimoto C, Saito A, Kikuzato M, Hayashi N, 2002, Angioechographic evaluation of tumor thrombi and the effect of transcatheter arterial embolization in patients with hepatocellular carcinoma. J Gastroenterol 37:363–368 Ando E, Tanaka M, Yamashita F, Kuromatsu R, Yutani S, Fukumori K, Sumie S, Yano Y, Okuda K, Sata M, 2002, Hepatic arterial infusion chemotherapy for advanced hepatocellular carcinoma with portal vein tumor thrombosis: analysis of 48 cases. Cancer 95:588– 595 Lai YC, Shih CY, Jeng CM, Yang SS, Hu JT, Sung YC, Liu HT, Hou SM, Wu CH, Chen TK, 2003, Hepatic arterial infusion chemotherapy for hepatocellular carcinoma with portal vein tumor thrombosis. World J Gastroenterol 9:2666–2670 Fan J, Zhou J, Wu ZQ, Qiu SJ, Wang XY, Shi YH, Tang ZY, 2005, Efficacy of different treatment strategies for hepatocellular carcinoma with portal vein tumor thrombosis. World J Gastroenterol 11:1215– 1219 Li Q, Wang J, Sun Y, Cui YL, Juzi JT, Li HX, Qian BY, Hao XS, 2006, Efficacy of postoperative transarterial chemoembolization and portal vein chemotherapy for patients with hepatocellular carcinoma complicated by portal vein tumor thrombosis–a randomized study. World J Surg 30:2004–2011 Takizawa D, Kakizaki S, Sohara N, Sato K, Takagi H, Arai H, Katakai K, Kojima A, Matsuzaki Y, Mori M, 2007, Hepatocellular carcinoma with portal vein tumor thrombosis: clinical characteristics, prognosis, and patient survival analysis. Dig Dis Sci 52: 3290–3295 Toya R, Murakami R, Baba Y, Nishimura R, Morishita S, Ikeda O, Kawanaka K, Beppu T, Sugiyama S, Sakamoto T, Yamashita Y, Oya N, 2007, Conformal radiation therapy for portal vein tumor thrombosis of hepatocellular carcinoma. Radiother Oncol 84:266–271 Kamiyama T, Nakanishi K, Yokoo H, Tahara M, Nakagawa T, Kamachi H, Taguchi H, Shirato H, Matsushita M, Todo S, 2007, Efficacy of preoperative radiotherapy to portal vein tumor thrombus in the main trunk or first branch in patients with hepatocellular carcinoma. Int J Clin Oncol 12:363–368 Liang LJ, HuWJ, Yin XY, Zhou Q, Peng BG, Li DM, LuMD(2008, Adjuvant intraportal venous chemotherapy for patients with hepatocellular carcinoma and portal vein tumor thrombi following hepatectomy plus portal thrombectomy. World J Surg 32:627–631 Lin DX, Zhang QY, Li X, Ye QW, Lin F, Li LL, 2011, An aggressive approach leads to improved survival in hepatocellular carcinoma patients with portal vein tumor thrombus. J Cancer Res Clin Oncol 137:139–149 Luo J, Guo RP, Lai EC, Zhang YJ, LauWY, Chen MS, ShiM(2011, Transarterial chemoembolization for unresectable hepatocellular carcinoma with portal vein tumor thrombosis: a prospective comparative study. Ann Surg Oncol 18:413–420 Chen JS,Wang Q, Chen XL, Huang XH, Liang LJ, Lei J, Huang JQ, Li DM, Cheng ZX, 2012, Clinicopathologic characteristics and surgical outcomes of hepatocellular carcinoma with portal vein tumor thrombosis. J Surg Res 175:243–250 Yoon SM, LimYS,WonHJ, KimJH, KimKM, LeeHC, ChungYH, Lee YS, Lee SG, Park JH, Suh DJ, 2012, Radiotherapy plus transarterial chemoembolization for hepatocellular carcinoma invading the portal vein: long-term patient outcomes. Int J Radiat Oncol Biol Phys 82:2004–2011 Peng ZW, Guo RP, Zhang YJ, Lin XJ, Chen MS, Lau WY, 2012, Hepatic resection versus transcatheter arterial chemoembolization for the treatment of hepatocellular carcinoma with portal vein tumor thrombus. Cancer 118:4725–4736 Jia L, Kiryu S, Watadani T, Akai H, Yamashita H, Akahane M, Ohtomo K, 2012, Prognosis of hepatocellular carcinoma with portal vein tumor thrombus:assessment based on clinical and computer tomography characteristics. Acta Med Okayama 66:131–141 Roayaie S, Blume IN, Thung SN, GuidoM, FielMI, Hiotis S, Labow DM, Llovet JM, Schwartz ME, 2009, A system of classifying microvascular invasion to predict outcome after resection in patients with hepatocellular carcinoma. Gastroenterology 137:850–855 Okusaka T, Okada S, Ueno H, Ikeda M, Shimada K, Yamamoto J, Kosuge T, Yamasaki S, Fukushima N, Sakamoto M, 2002, Satellite lesions in patients with small hepatocellular carcinoma with reference to clinicopathologic features. Cancer 95:1931–1937 Edmondson HA, Steiner PE, 1954, Primary carcinoma of the liver: a study of 100 cases among 48,900 necropsies. Cancer 7:462–503 Cheng CH, Lee CF, Wu TH, Chan KM, Chou HS, Wu TJ, Yu MC, Chen TC, Lee WC, Chen MF, 2011, Evaluation of the new AJCC staging system for resectable hepatocellular carcinoma.World J Surg Oncol 9:114 Terminology Committee of the International Hepato-Pancreato- Biliary Association, 2000, The Brisbane 2000 terminology of liver anatomy and resections. HPB 2:333–339 Wang T, Hu HS, Feng YX, Shi J, Li N, GuoWX, Xue J, Xie D, Liu SR, Wu MC, Cheng SQ, 2010, Characterisation of a novel cell line, CSQT-2, with high metastatic activity derived from portal vein tumour thrombus of hepatocellular carcinoma. Br J Cancer 102: 1618–1626 Minagawa M, Makuuchi M, 2006, Treatment of hepatocellular carcinoma accompanied by portal vein tumor thrombus. World J Gastroenterol 12:7561–7567 El-Serag HB, 2011, Hepatocellular carcinoma. N Engl J Med 365: 1118–1127 Poon RT, Fan ST, Ng IO, Lo CM, Liu CL, Wong J, 2000, Different risk factors and prognosis for early and late intrahepatic recurrence after resection of hepatocellular carcinoma. Cancer 89: 500–507 Shah SA, Greig PD, Gallinger S, Cattral MS, Dixon E, Kim RD, Taylor BR, Grant DR, Vollmer CM, 2006, Factors associated with early recurrence after resection for hepatocellular carcinoma and outcomes. J Am Coll Surg 202:275–283 Chun JM, Kwon HJ, Sohn J, Kim SG, Park JY, Bae HI, Yun YK, Hwang YJ, 2011, Prognostic factors after early recurrence in patients who underwent curative resection for hepatocellular carcinoma. J Surg Oncol 103:148–151 Lim KC, Chow PK, Allen JC, Chia GS, Lim M, Cheow PC, Chung AY, Ooi LL, Tan SB, 2011, Microvascular invasion is a better predictor of tumor recurrence and overall survival following surgical resection for hepatocellular carcinoma compared to theMilan criteria. Ann Surg 254:108–113 Kaido T, Arii S, Oe H,Mori A, ImamuraM(2005, Predictive factors affecting early recurrence after hepatectomy for hepatocellular carcinoma in 5-year survivors. Hepatogastroenterology 52:1484– 1487 Muscari F, Foppa B, Carrere N, Kamar N, Peron JM, Suc B, 2011, Resection of a transplantable single-nodule hepatocellular carcinoma in Child-Pugh class A cirrhosis: factors affecting survival and recurrence. World J Surg 35:1055–1062 Arii S, Tanaka J, Yamazoe Y,Minematsu S, Morino T, Fujita K, Maetani S, Tobe T, 1992, Predictive factors for intrahepatic recurrence of hepatocellular carcinoma after partial hepatectomy. Cancer 69:913–919 Kim BK, Han KH, Park YN, ParkMS, Kim KS, Choi JS, Moon BS, Chon CY, Moon YM, Ahn SH, 2008, Prediction of microvascular invasion before curative resection of hepatocellular carcinoma. J Surg Oncol 97:246–252 Yu GR, Kim SH, Park SH, Cui XD, Xu DY, Yu HC, Cho BH, Yeom YI, Kim SS, Kim SB, Chu IS, Kim DG, 2007, Identification of molecular markers for the oncogenic differentiation of hepatocellular carcinoma. Exp Mol Med 39:641–652 Xiang ZL, Zeng ZC, Tang ZY, Fan J, Sun HC, Tan YS, 2011, Expression of cytokeratin 19 and matrix metalloproteinase 2 predicts lymph node metastasis in hepatocellular carcinoma. Mol Biol Rep 38:3531–3539 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 131 EP 138 DI 10.1007/s12253-014-9798-2 PG 8 ER PT J AU Cui, G Yang, H Zhao, J Yuan, A Florholmen, J AF Cui, Guanglin Yang, Hang Zhao, Jianbo Yuan, Aping Florholmen, Jon TI Elevated Proinflammatory Cytokine IL-17A in the Adjacent Tissues Along the Adenoma-Carcinoma Sequence SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE IL-17A; Adjacent mucosa; Colorectum; Cancer ID IL-17A; Adjacent mucosa; Colorectum; Cancer AB Considerable evidence has suggested that chronic inflammation is a causative factor in the development of human colorectal cancer (CRC). Interleukin (IL)-17A produced mainly by Th17 cells is a novel proinflammatory cytokine and increased IL-17A is associated with colorectal neoplastic transformation. In this study, we have evaluated the expression of IL-17A in the adjacent tissues along the colorectal adenoma-carcinoma sequence. The expression of IL- 17A in the adjacent tissues of colorectal adenoma (adenomaadjacent, n=32) and sporadic CRC (CRC-adjacent, n=45) was examined. In addition, the expression pattern of Th17 cell differentiation stimulators (IL-1β, IL-6 and IL-23A) in the adjacent tissues were also examined. The results showed that the expression level of IL-17A mRNA was non-statistically increased (4-fold higher) in the adenoma-adjacent tissues and it became significantly increased (9-fold higher) in the CRC-adjacent tissues as compared with the control. The expression level of IL-17A in the CRC-adjacent tissues was not associated with CRC clinicopathological parameters and overall survival. Immunohistochemistry confirmed an increased density of intraepithelial IL-17A expressing cells in the CRC-adjacent tissues. The Th17 cell differentiation simulators IL- 1β and IL-6 were also shown in an increase trend from the adenoma-adjacent to CRC-adjacent tissues. These results provide evidence that IL-17A/Th17 response is enhanced in the adjacent tissues during the colorectal neoplastic transformation. C1 [Cui, Guanglin] The Second Affiliated Hospital of Zhengzhou University, Department of GastroenterologyZhengzhou, Henan, China. [Yang, Hang] The Second Affiliated Hospital of Zhengzhou University, Department of GastroenterologyZhengzhou, Henan, China. [Zhao, Jianbo] The Second Affiliated Hospital of Zhengzhou University, Department of GastroenterologyZhengzhou, Henan, China. [Yuan, Aping] The Second Affiliated Hospital of Zhengzhou University, Department of GastroenterologyZhengzhou, Henan, China. [Florholmen, Jon] University of Tromso, Faculty of Medicine, Institute of Clinical Medicine, Laboratory of Gastroenterology & NutritionTromso, Norway. RP Cui, G (reprint author), The Second Affiliated Hospital of Zhengzhou University, Department of Gastroenterology, Zhengzhou, China. CR O’Byrne KJ, Dalgleish AG, 2001, Chronic immune activation and inflammation as the cause of malignancy. Br J Cancer 85:473–483 Balkwill FR, Mantovani A, 2012, Cancer-related inflammation: common themes and therapeutic opportunities. Semin Cancer Biol 22:33–40 Lakatos PL, DavidG, Pandur T, Erdelyi Z, MesterG et al, 2011, Risk of colorectal cancer and small bowel adenocarcinoma in Crohn’s disease: a population-based study from western Hungary 1977– 2008. J Crohns Colitis 5:122–128 Lakatos PL, Lakatos L, 2008, Risk for colorectal cancer in ulcerative colitis: changes, causes and management strategies. World J Gastroenterol 14:3937–3947 Triantafillidis JK, Nasioulas G, Kosmidis PA, 2009, Colorectal cancer and inflammatory bowel disease: epidemiology, risk factors, mechanisms of carcinogenesis and prevention strategies. Anticancer Res 29:2727–2737 Kraus S, Arber N, 2009, Inflammation and colorectal cancer. Curr Opin Pharmacol 9:405–410 Brigati C, Noonan DM, Albini A, Benelli R, 2002, Tumors and inflammatory infiltrates: friends or foes? Clin Exp Metastasis 19: 247–258 Rhodes JM, Campbell BJ, 2002, Inflammation and colorectal cancer: IBD-associated and sporadic cancer compared. Trends Mol Med 8: 10–16 O’Sullivan C, Lewis CE, 1994, Tumour-associated leucocytes: friends or foes in breast carcinoma. J Pathol 172:229–235 Yu P, Fu YX, 2006, Tumor-infiltrating T lymphocytes: friends or foes? Lab Invest 86:231–245 Shurin GV, Ma Y, Shurin MR, 2013, Immunosuppressive mechanisms of regulatory dendritic cells in cancer. Cancer Microenviron 6: 159–167 Moossavi S, Bishehsari F, 2012, Inflammation in sporadic colorectal cancer. Arch of Iran med 15:166–170 Cui G, Yuan A, Vonen B, Florholmen J, 2009, Progressive cellular response in the lamina propria of the colorectal adenoma-carcinoma sequence. Histopathology 54:550–560 Banner BF, Savas L, Baker S, Woda BA, 1993, Characterization of the inflammatory cell populations in normal colon and colonic carcinomas. Virchows Arch B Cell Pathol Incl Mol Pathol 64:213–220 Banner BF, Sonmez-Alpan E, Yousem SA, 1993, An immunophenotypic study of the inflammatory cell populations in colon adenomas and carcinomas. Mod Pathol 6:295–301 Cui G, Goll G, Olsen T, Steigen S, Husebekk A et al, 2007, Reduced expression of microenvironmental Th1 cytokines accompanies adenomas-carcinomas sequence of colorectum. Cancer Immunol & Immunother 56:985–995 Cui G, Yuan A, Goll R, Florholmen J, 2012, IL-17A in the tumor microenvironment of the human colorectal adenoma-carcinoma sequence. Scand J Gastroenterol 47:1304–1312 Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini PL, 2011, 2011: the immune hallmarks of cancer. Cancer Immunol Immunother 60: 319–326 Ito R, Kita M, Shin-Ya M, Kishida T, Urano A et al, 2008, Involvement of IL-17A in the pathogenesis of DSS-induced colitis in mice. Biochem Biophys Res Commun 377:12–16 Ogawa A, Andoh A, Araki Y, Bamba T, Fujiyama Y, 2004, Neutralization of interleukin-17 aggravates dextran sulfate sodiuminduced colitis in mice. Clin Immunol 110:55–62 Monteleone I, Sarra M, Pallone F,MonteleoneG(2012, Th17-related cytokines in inflammatory bowel diseases: friends or foes? CurrMol Med 12:592–597 Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X et al, 2009, A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med 15:1016–1022 Nam JS, Terabe M, Kang MJ, Chae H, Voong N et al, 2008, Transforming growth factor beta subverts the immune system into directly promoting tumor growth through interleukin-17. Cancer Res 68:3915–3923 Le Gouvello S, Bastuji-Garin S, Aloulou N, Mansour H, Chaumette MT et al, 2008, High prevalence of Foxp3 and IL17 in MMRproficient colorectal carcinomas. Gut 57:772–779 Langley RR, Fidler IJ, 2007, Tumor cell-organ microenvironment interactions in the pathogenesis of cancer metastasis. Endocr Rev 28: 297–321 Shi Y, Lin H, Cui J, Qi H, Florholmen J et al, 2013, The role of interleukin-17A in colorectal tumorigenesis. Cancer Biother Radiopharm 28:429–432 Radosavljevic G, Ljujic B, Jovanovic I, Srzentic Z, Pavlovic S et al, 2010, Interleukin-17 may be a valuable serum tumor marker in patients with colorectal carcinoma. Neoplasma 57:135–144 Wang J, Xu K, Wu J, Luo C, Li Yet al, 2012, The changes of Th17 cells and the related cytokines in the progression of human colorectal cancers. BMC Cancer 12:418 Liu J, Duan Y, Cheng X, Chen X, Xie W et al, 2011, IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma. Biochem Biophys Res Commun 407:348–354 Oshiro K, Kohama H, Umemura M, Uyttenhove C, Inagaki- Ohara K et al, 2012, Interleukin-17A is involved in enhancement of tumor progression in murine intestine. Immunobiology 217:54–60 Yang S,Wang B,Guan C,Wu B, Cai C et al, 2011, Foxp3+IL-17+ T cells promote development of cancer-initiating cells in colorectal cancer. J Leukoc Biol 89:85–91 Hyun YS, Han DS, Lee AR, Eun CS, Youn J et al, 2012, Role of IL- 17A in the development of colitis-associated cancer. Carcinogenesis 33:931–936 ChaeWJ, Gibson TF, Zelterman D, Hao L, Henegariu O et al, 2010, Ablation of IL-17A abrogates progression of spontaneous intestinal tumorigenesis. Proc Natl Acad Sci U S A 107:5540–5544 Fearon ER, 1994, Molecular genetic studies of the adenomacarcinoma sequence. Adv Intern Med 39:123–147 Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC et al, 1988, Genetic alterations during colorectal-tumor development. N Engl J Med 319:525–532 Rabinovich GA, Gabrilovich D, Sotomayor EM, 2007, Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol 25:267–296 Almand B, Resser JR, Lindman B, Nadaf S, Clark JI et al, 2000, Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res 6:1755–1766 Jiang B,Mason J, Jewett A, Qian J, Ding Yet al, 2013, Cell budding from normal appearing epithelia: a predictor of colorectal cancer metastasis? Int J Biol Sci 9:119–133 Kuniyasu H, Yasui W, Shinohara H, Yano S, Ellis LM et al, 2000, Induction of angiogenesis by hyperplastic colonicmucosa adjacent to colon cancer. Am J Pathol 157:1523–1535 Kuniyasu H, Ohmori H, Sasaki T, Sasahira T, Yoshida K et al, 2003, Production of interleukin 15 by human colon cancer cells is associated with induction of mucosal hyperplasia, angiogenesis, and metastasis. Clin Cancer Res 9:4802–4810 Hao CY, Moore DH, Wong P, Bennington JL, Lee NM et al, 2005, Alteration of gene expression in macroscopically normal colonic mucosa from individuals with a family history of sporadic colon cancer. Clin Cancer Res 11:1400–1407 Cui G, Yuan A, Goll R, Olsen T, Husebekk A et al, 2007, Distinct changes of dendritic cell number and IL-12 mRNA level in adjacent mucosa throughout the colorectal adenoma-carcinoma sequence. Cancer immunol immunother: CII 56:1993–2001 Korn T, Bettelli E, Oukka M, Kuchroo VK, 2009, IL-17 and Th17 Cells. Annu Rev Immunol 27:485–517 Cui G, Olsen T, Christiansen I, Vonen B, Florholmen J et al, 2006, Improvement of real-time PCR for quantifying TNF-a mRNA expression in inflamed colorectal mucosa-an approach to optimize procedures for clinical use. The Scand J of Clin and Lab Investig 66:249–259 Cui G, Yuan A, Goll R, Vonen B, Florholmen J, 2009, Dynamic changes of interleukin-8 network along the colorectal adenomacarcinoma sequence. Cancer immunol immunother: CII 58:1897– 1905 Yuan A, Steigen SE, Goll R, Vonen B, Husbekk A et al, 2008, Dendritic cell infiltration pattern along the colorectal adenomacarcinoma sequence. APMIS : acta pathologica, microbiologica, et immunologica Scandinavica 116:445–456 Miki C, Tonouchi H,Wakuda R, Hatada T, Inoue Yet al, 2002, Intratumoral interleukin-6 down-regulation system and genetic mutations of tumor suppressor genes in colorectal carcinoma. Cancer 94:1584– 1592 Chung YC, Chang YF, 2003, Serum interleukin-6 levels reflect the disease status of colorectal cancer. J Surg Oncol 83:222–226 Lan F, Zhang L, Wu J, Zhang J, Zhang S et al, 2011, IL-23/IL-23R: potential mediator of intestinal tumor progression from adenomatous polyps to colorectal carcinoma. Int J Colorectal Dis 26:1511–1518 Suzuki H, Ogawa H, Miura K, Haneda S, Watanabe K et al, 2012, IL-23 directly enhances the proliferative and invasive activities of colorectal carcinoma. Oncol Lett 4:199–204 Kusmartsev S, Gabrilovich DI, 2006, Effect of tumor-derived cytokines and growth factors on differentiation and immune suppressive features of myeloid cells in cancer. Cancer Metastasis Rev 25:323–331 Dome B, Hendrix MJ, Paku S, Tovari J, Timar J, 2007, Alternative vascularization mechanisms in cancer: pathology and therapeutic implications. Am J Pathol 170:1–15 NumasakiM, Fukushi J, OnoM, Narula SK, Zavodny PJ et al, 2003, Interleukin-17 promotes angiogenesis and tumor growth. Blood 101: 2620–2627 Silva-Santos B, 2010, Promoting angiogenesis within the tumor microenvironment: the secret life of murine lymphoid IL-17- producing gammadelta T cells. Eur J Immunol 40:1873–1876 Liu J, Duan Y, Cheng X, Chen X, Xie W et al, 2011, IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma. Biochem Biophys Res Commun 407:348–354 Pasche N, Frey K, Neri D, 2012, The targeted delivery of IL17 to the mouse tumor neo-vasculature enhances angiogenesis but does not reduce tumor growth rate. Angiogenesis 15:165–169 Fox SH, Whalen GF, Sanders MM, Burleson JA, Jennings K et al, 1998, Angiogenesis in normal tissue adjacent to colon cancer. J Surg Oncol 69:230–234 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 139 EP 146 DI 10.1007/s12253-014-9799-1 PG 8 ER PT J AU Voros, A Csorgo, E Kovari, B Lazar, P Kelemen, Gy Rusz, O Nyari, T Cserni, G AF Voros, Andras Csorgo, Erika Kovari, Bence Lazar, Peter Kelemen, Gyongyi Rusz, Orsolya Nyari, Tibor Cserni, Gabor TI Different Methods of Pretreatment Ki-67 Labeling Index Evaluation in Core Biopsies of Breast Cancer Patients Treated with Neoadjuvant Chemotherapy and Their Relation to Response to Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast carcinoma; Evaluation methods; Ki-67; Neoadjuvant therapy; Tumor regression ID Breast carcinoma; Evaluation methods; Ki-67; Neoadjuvant therapy; Tumor regression AB Increased proliferation activity of breast cancer cells evaluated by Ki-67 immunohistochemistry, i.e. a high Ki-67 labeling index (Ki-67 LI), may predict better tumor regression in case of neoadjuvant chemotherapy. Despite recommendations for the evaluation of Ki-67 LI, there are variations in methodology. We assessed the effect of different evaluation methods on the Ki-67 LI in patients with different response to neoadjuvant chemotherapy. Thirty pretreatment core-biopsy samples of patients receiving neoadjuvant docetaxel-epirubicin chemotherapy with or without capecitabine were evaluated for their Ki-67 LI. Pathologic regression was categorized as no regression, partial regression and complete regression, with 10 cases in each category. Three antibodies (MIB1, B56, SP6), 4 observers and 4 methods (counting or estimating on glass slides and counting or estimating on representative digital images) were compared. The Kruskal-Wallis test and analyses of variance were performed to investigate the differences in Ki-67 LIs between different clinical outcomes (tumor regression categories). Breast carcinomas with pathological complete regression had a higher mean Ki-67 LI than tumors not achieving complete regression with any methods, observers and antibodies investigated, although there was a variation between different evaluations in what may represent high proliferation. Estimating the Ki-67 LI on digital images representing the highest proliferation in the core biopsy seemed the best in separating complete responders from non-responders. High Ki-67 LI values were more likely associated with pathological complete regression independently of the method of evaluation used, although the definition of high proliferation is problematic. Estimating the Ki-67 LI may be an adequate method of evaluation. C1 [Voros, Andras] University of Szeged, Department of Pathology, Allomas u 2, 6725 Szeged, Hungary. [Csorgo, Erika] University of Szeged, Department of Pathology, Allomas u 2, 6725 Szeged, Hungary. [Kovari, Bence] University of Szeged, Department of Pathology, Allomas u 2, 6725 Szeged, Hungary. [Lazar, Peter] University of Szeged, Department of Pathology, Allomas u 2, 6725 Szeged, Hungary. [Kelemen, Gyongyi] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Rusz, Orsolya] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Nyari, Tibor] University of Szeged, Department of Medical Informatics, Koranyi fasor 9, 6720 Szeged, Hungary. [Cserni, Gabor] University of Szeged, Department of Pathology, Allomas u 2, 6725 Szeged, Hungary. RP Voros, A (reprint author), University of Szeged, Department of Pathology, 6725 Szeged, Hungary. EM andrasvoros@libero.it CR Stuart-Harris R, Caldas C, Pinder SE, Pharoah P, 2009, Proliferation markers and survival in early breast cancer: a systematic review and meta-analysis of 85 studies in 32,825 patients. Breast 17(4):323–334 de Azambuja E, Cardoso F, de Castro G, Jr CM, Mano DV et al, 2007, Ki-67 as prognostic marker in early breast cancer: a metaanalysis of published studies involving 12,155 patients. Br J Cancer 96:1504–1513 Urruticoechea A, Smith IE, Dowsett M, 2005, Proliferation marker Ki-67 in early breast cancer. J Clin Oncol 23:7212–7220 Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA, 2010, Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol 11:174–183 Boom HJ, Richardson WW, 1957, Histological grading and prognosis in breast cancer; a study of 1409 cases of which 359 have been followed for 15 years. Br J Cancer 11(3):359–377 Beresford MJ,Wilson GD, Makris A, 2006, Measuring proliferation in breast cancer: practicalities and applications. Breast Cancer Res 8: 216 Gerdes J, Li L, Schlueter C, Duchrow M, Wohlenberg C, Gerlach C et al, 1991, Immunobiochemical and molecular biologic characterization of the cell proliferation-associated nuclear antigen that is defined by monoclonal antibody Ki-67. Am J Pathol 138:867–873 Gerdes J, Schwab U, Lemke H, Stein H, 1983, Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer 31:13–20 Fasching PA, Heusinger K, Haeberle L, Niklos M, Hein A, Bayer CM et al, 2011, Ki67, chemotherapy response, and prognosis in breast cancer patients receiving neoadjuvant treatment. BMC Cancer 11:486 Bozzetti C, Musolino A, Camisa R, Bisagni G, Flora M, Bassano C et al, 2006, Evaluation of HER-2/neu amplification and other biological markers as predictors of response to neoadjuvant anthracycline-based chemotherapy in primary breast cancer: the role of anthracycline dose intensity. Am J Clin Oncol 29(2):171–177 MacGrogan G, Mauriac L, Durand M, Bonichon F, Trojani M, de Mascarel I, Coindre JM, 1996, Primary chemotherapy in breast invasive carcinoma: predictive value of the immunohistochemical detection of hormonal receptors, p53, c-erbB-2, MiB1, pS2 and GST pi. Br J Cancer 74(9):1458–1465 Faneyte IF, Schrama JG, Peterse JL, Remijnse PL, Rodenhuis S, van de Vijver MJ, 2003, Breast cancer response to neoadjuvant chemotherapy: predictive markers and relation with outcome. Br J Cancer 88(3):406–412 Penault-Llorca F, Abrial C, Raoelfils I, Chollet P, Cayre A, Mouret- Reynier MA et al, 2008, Changes and predictive and prognostic value of the mitotic index, Ki-67, cyclin D1, and cyclo-oxygenase- 2 in 710 operable breast cancer patients treated with neoadjuvant chemotherapy. Oncologist 13(12):1235–1245 Petit T, Wilt M, Velten M, Millon R, Rodier JF, Borel C et al, 2004, Comparative value of tumour grade, hormonal receptors, Ki-67, HER-2 and topoisomerase II alpha status as predictive markers in breast cancer patients treated with neoadjuvant anthracycline-based chemotherapy. Eur J Cancer 40(2):205–211 Pohl G, Rudas M, Taucher S, Stranzl T, Steger GG, Jakesz R et al, 2003, Expression of cell cycle regulatory proteins in breast carcinomas before and after preoperative chemotherapy. Breast Cancer Res Treat 78(1):97–103 Luporsi E, Andre F, Spyratos F, Martin PM, Jacquemier J, Penault-Llorca F et al, 2012, Ki-67: level of evidence and methodological considerations for its role in the clinical management of breast cancer: analytical and critical review. Breast Cancer Res Treat 132:895–915 Goldhirsch A, Ingle JN, Gelber RD, Coates AS, Thurlimann B, Senn HJ, 2009, Thresholds for therapies: highlights of the St Gallen International Expert Consensus on the primary therapy of early breast cancer 2009. Ann Oncol 20:1319–1329 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, 2011, Strategies for subtypes–dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol 22:1736–1747 GnantM, Harbeck N, Thomssen C, 2011, St. Gallen, 2011: summary of the consensus discussion. Breast Care, Basel, 6:136–141 Curigliano G, Criscitiello C, Andre F, Colleoni M, Di Leo A, 2013, Highlights from the 13th St Gallen International Breast Cancer Conference 2013. Access to innovation for patients with breast cancer: how to speed it up? Ecancermedicalscience 7:299 Fasanella S, Leonardi E, Cantaloni C, Eccher C, Bazzanella I, Aldovini D et al, 2011, Proliferative activity in human breast cancer: Ki-67 automated evaluation and the influence of different Ki-67 equivalent antibodies. Diagn Pathol 6 DowsettM, Nielsen TO,A’Hern R, Bartlett J, Coombes RC, Cuzick J et al, 2011, Assessment of Ki67 in breast cancer: recommendations from the International Ki67 in Breast Cancer Working Group. J Natl Cancer Inst 103:1–9 Voros A, Csorgo E, Nyari T, Cserni G, 2012, An intra- and interobserver reproducibility analysis of the Ki-67 proliferation marker assessment on core biopsies of breast cancer patients and its potential clinical implications. Pathobiology 80:111–118 Voros A, Csorgo E, Kovari B, Lazar P, Kelemen G, Cserni G., 2013, The Use of Digital Images Improves Reproducibility of the Ki-67 Labeling Index as a Proliferation Marker in Breast Cancer. Pathol Oncol Res. 2013 Nov 8 Nikolenyi A, Sukosd F, Kaizer L, Csorgo E, Voros A, Uhercsak G et al, 2011, Tumor topoisomerase II alpha status and response to anthracycline-based neoadjuvant chemotherapy in breast cancer. Oncology 80(3–4):269–277 Wells CA, Amendoeira I, Bellocq JP, Bianchi S, BoeckerW, Borisch B et al, 2012, S2: Pathology update. Quality assurance guidelines for pathology. In: Perry N, BroedersM, deWolf C, Tornberg S, Holland R, von Karsa L, eds, European guidelines for quality assurance in breast cancer screening and diagnosis. Fourth edition, Supplements. European Commission, Office for Official Publications of the European Union, Luxembourg, pp 73–120 Chang J, OrmerodM, Powles TJ, Allred DC, Ashley SE, DowsettM, 2000, Apoptosis and proliferation as predictors of chemotherapy response in patients with breast carcinoma. Cancer 89:2145–2152 Faneyte IF, Schrama JG, Peterse JL, Remijnse PL, Rodenhuis S, van de Vijver MJ, 2003, Breast cancer response to neoadjuvant chemotherapy: predictive markers and relation with outcome. Br J Cancer 88:406–412 Keam B, Im SA, Park S, Nam BH, Han SW, Oh DY et al, 2011, Nomogram predicting clinical outcomes in breast cancer patients treated with neoadjuvant chemotherapy. J Cancer Res Clin Oncol 137:1301–1308 Makris A, Powles TJ, Dowsett M, Osborne CK, Trott PA, Fernando IN et al, 1997, Prediction of response to neoadjuvant chemoendocrine therapy in primary breast carcinomas. Clin Cancer Res 3:593–600 Dowsett M, Smith IE, Ebbs SR, Dixon JM, Skene A, A’Hern R et al, 2007, Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer. J Natl Cancer Inst 99(2):167–170 Zs V, Diebold J, Dommann-Scherrer C, Frick H, Kaup D, Noske A et al, 2012, How reliable is Ki-67 immunohistochemistry in grade 2 breast carcinomas? A QA study of the Swiss Working Group of Breast- and Gynecopathologists. PLoS ONE 7:e37379 Cserni G, Voros A, Liepniece-Karele I, Bianchi S, Vezzosi V, Grabau D et al, 2014, Distribution pattern of the Ki67 labelling index in breast cancer and its implications for choosing cut-off values. Breast 2014 Mar 6 Valero V, Buzdar AU, McNeese M, Singletary E, Hortobagyi GN, 2002, Primary chemotherapy in the treatment of breast cancer: the University of Texas M. D. Anderson Cancer Center experience. Clin Breast Cancer 3(Suppl 2):S63–S68 Montagna E, Bagnardi V, Rotmensz N, Viale G, Pruneri G, Veronesi P et al, 2010, Pathological complete response after preoperative systemic therapy and outcome: relevance of clinical and biologic baseline features. Breast Cancer Res Treat 124(3):689–699 Cheang MCU, Chia SK, Voduc D, Gao D, Leung S, Snider J et al, 2009, Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 101:736–750 Houssami N, Macaskill P, von Minckwitz G, Marinovich ML, Mamounas E, 2012)Meta-analysis of the association of breast cancer subtype and pathologic complete response to neoadjuvant chemotherapy. Eur J Cancer 48:3342–3354 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 147 EP 155 DI 10.1007/s12253-014-9800-z PG 9 ER PT J AU Omran, MO Al Sheeha, M AF Omran, Mahmoud Ola Al Sheeha, Muneera TI Human Papilloma Virus Early Proteins E6 (HPV16/18-E6) and the Cell Cycle Marker P16 (INK4a) are Useful Prognostic Markers in Uterine Cervical Carcinomas in Qassim Region- Saudi Arabia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Immunohistochemistry; Cervical carcinoma; HPV16/18E6; P16 (INK4a) ID Immunohistochemistry; Cervical carcinoma; HPV16/18E6; P16 (INK4a) AB Cervical cancer is a common and an important public health problem for adult women in developing countries. In contrast, cervical cancer incidence is low in Saudi Arabia. High-risk types of human papilloma viruses (HPV16 and HPV18) are the most significant risk factors for cervical cancer. HPV16/18-E6 oncoprotein is associated with HPV etiology, viral persistence and epithelial transformation. Cell cycle protein p16 INK4a (p16) plays an important role in the pathophysiology of cervical carcinomas. The aims of this study were to investigate the expression of HPV16/18-E6 and p16 in uterine cervical carcinomas in Qassim Region - Saudi Arabia, and to relate the results to the established clinicopathological prognostic parameters (age of the patient, educational level, birth control methods, number of pregnancy, smoking status, degree of histological differentiation, clinical stage, and lymph node metastasis) The study included 40 specimens of uterine cervical squamous cell carcinomas diagnosed and confirmed by biopsy. Histopathological classification of cervical tumors cases was performed according to the International Federation of Gynecology and Obstetrics (FIGO). Immunohistochemical analysis for HPV16/18-E6 and p16 were carried out on formalin-fixed paraffinembedded sections of cervical tissues using avidin-biotin peroxidase method. There was a significant statistical correlation between HPV16/18-E6 expression in cervical carcinoma and nationality, smoking status and size of the tumor. HPV16/ 18-E6 oncoprotein expression in normal lymphocytes and endothelial cells in the tumor tissues and the adjacent normal cervical tissues suggest the possibility that HPV infection might spread to other organs through blood circulation. P16 expression has been correlated with high grade, stage of cervical SCC and HPV16/18-E6 expression. The current study supports the critical function of p16 and HPV16/18-E6 as specific markers for cervical carcinoma. However the potential for usage of p16 and HPV16/18-E6 as prognostic markers will require detailed follow data for a larger group of patients. C1 [Omran, Mahmoud Ola] Asyut University, Faculty of Medicine, Pathology DepartmentAssiut, Egypt. [Al Sheeha, Muneera] Qassim University, College of Medicine, Department of Obstetrics and GynacologyBuridah, Qassim Region, Saudi Arabia. RP Omran, MO (reprint author), Asyut University, Faculty of Medicine, Pathology Department, Assiut, Egypt. EM ola67oh@yahoo.com CR Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics 2002. CA Cancer J Clin 55:74–108 Sankaranarayanan R, Madhukar A, Rajkumar R, 2001, Effective screening programmes for cervical cancer in low- and middleincome developing countries. Bull World Health Organ 79(10): 954–962 GLOBOCAN 2000. Cancer incidence, mortality and prevalence worldwide, version 10. IARC Cancer Base No 5 Lyon, IARC Press. 2001 Bosch FX, de Sanjose S, 2003, Human papillomavirus and cervical cancer – burden and assessment of causality. J Natl Cancer Inst Monogr 31:3–13 Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, Snijders PJ, Peto J, Meijer CJ, Munoz N, 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189(1):12–19 de Sanjose S,DiazM, CastellsagueX, Clifford G, Bruni L,MunozN, Bosch FX(2007)Worldwide prevalence and genotype distribution of cervical human papillomavirus DNA in women with normal cytology: a meta-analysis. Lancet Infect Dis 7(7):453–459 Wright TC Jr, Schiffman M, 2003, Adding a test for human papillomavirus DNA to cervical- cancer screening. N Engl J Med 348:489 What Causes Cancer of the Cervix?. American Cancer Society, 2014-01-31). RashedMM, Bekele A, 2011, The prevalence and pattern ofHPV-16 immunostaining in uterine cervical carcinomas in ethiopian women: a pilot study. Pan Afr Med J 8:21 Missaoui N, Hmissa S, Frappart L, Trabelsi A, Ben Abdelkader A, Traore C, Mokni M, Yaacoubi MT, Korbi S, 2006, p16INK4A overexpression and HPVinfection in uterine cervix adenocarcinoma. Virchows Arch 448(5):597–603 Bhurgri Y, Bhurgri A, Rahim A, Bhutto K, Pinjani PK, Usman A, Hasan SH, 1999, The pattern of malignancies in Karachi, 1995 to 1996). J Pak Med Assoc 49:157–161 Altaf F, 2001, Pattern of cervical smear cytology inwestern region of Saudi Arabia. Ann Saudi Med 21:94–96 Jamal A, Al-Maghrabi JA, 2003, Profile of pap smear cytology in the Western region of Saudi Arabia. Saudi Med J 24:1225–1229 Raza SA, Franceschi S, Pallardy S, Malik FR, Avan BI, Zafar A, Ali SH, Pervez S, Serajuddaula S, Snijders PJ et al, 2010, Human papillomavirus infection in women with and without cervical cancer in Karachi, Pakistan. Br J Cancer 102:1657–1660 Castellsague X, Bosch FX, Munoz N, Meijer CJ, Shah KV, de Sanjose S, Eluf-Neto J, Ngelangel CA, Chichareon S, Smith JS et al, 2002, Male circumcision, penile human papillomavirus infection, and cervical cancer in female partners. N Engl JMed 346:1105– 1112 Hammouda D, Clifford GM, Pallardy S, Ayyach G, Chekiri A, Boudrich A, Snijders PJ, van Kemenade FJ, Meijer CJ, Bouhadef A et al, 2011, Human papillomavirus infection in a population-based sample of women in Algiers, Algeria. Int J Cancer 128:2224–2229 Alsbeih G, Ahmed R, Al-Harbi N, Venturina LA, Tulbah A, Balaraj K, 2011, Prevalence and genotypes’ distribution of human papillomavirus in invasive cervical cancer in Saudi Arabia. Gynecol Oncol 121:522–526 Al-Badawi IA, Al-Suwaine A, Al-Aker M, Asaad L, Alaidan A, Tulbah A, Fe BoholM,Munkarah AR, 2011)Detection and genotyping of human papiloma virus in cervical cancer specimens from Saudi patients. Int J Gynecol Cancer 21:907–910 Khorasanizadeh F, Hassanloo J, Khaksar N, Taheri SM, Marzaban M, Rashidi BH, Sari AA, Zendehdel K, 2012, Epidemiology of cervical cancer and human papilloma virus infection among Iranian women - analyses of national data and systematic review of the literature. Gynecol Oncol 128:277–281 Hussain S, Bharadwaj M, Nasare V, Kumari M, Sharma S, Hedau S, Das BC, 2012, Human papillomavirus infection among young adolescents in India: impact of vaccination. J Med Virol 84:298–305 Al-Muammar T, Al-AhdalMN, Hassan A, Kessie G, Dela Cruz DM, Mohamed GE, 2007, Human papilloma virus-16/18 cervical infection among women attending a family medical clinic in Riyadh. Ann Saudi Med 27:1–5 Duttagupta C, Sengupta S, Roy M, Sengupta D, Bhattacharya P, Laikangbam P, Roy S, Ghosh S, Das R, 2004, Are Muslim women less susceptible to oncogenic human papillomavirus infection? a study from rural Eastern India. Int J Gynecol Cancer 14:293–303 de BoerMA,Vet JN, AzizMF, Cornain S, Purwoto G, van den Akker BE, Dijkman A, Peters AA, Fleuren GJ, 2006, Human papillomavirus type 18 and other risk factors for cervicacancer in Jakarta, Indonesia. Int J Gynecol Cancer 16:1809–1814 Alsbeih G, Al-Harbi N, El-Sebaie M, Al-Badawi I, 2013, HPV prevalence and genetic predisposition to cervical cancer in Saudi Arabia. Infect Agent Cancer 8(1):15 Ren C, Cheng X, Lu B, Yang G. Activation of interleukin-6/ signal transducer and activator of transcription 3 by human papillomavirus early proteins 6 induces fibroblast senescence to promote cervical tumourigenesis through autocrine and paracrine pathways in tumour microenvironment. Eur J Cancer. 2013 Aug 14. Kaewprag J, Umnajvijit W, Ngamkham J, Ponglikitmongkol M, 2013, HPV16 oncoproteins promote cervical cancer invasiveness by upregulating specific matrix metalloproteinases. PLoS One 8(8): e71611 Sano T, Oyama T, Kashiwabara K, Fukuda T, Nakajima T, 1998, Expression status of p16 protein is associated with human papillomavirus oncogenic potential in cervical and genital lesions. Am J Pathol 153(6):1741–1748 Bringold F, Serrano M, 2000, Tumor suppressors and oncogenes in cellular senescence. Exp Gerontol 35(3):317–329 Lin HP, Wang YP, Chiang CP, 2011, Expression of p53, MDM2, p21, heat shock protein 70, and HPV 16/18 E6 proteins in oral verrucous carcinoma and oral verrucous hyperplasia. Head Neck 33:334–340 Amaro-Filho SM, Golub JE, Nuovo GJ, Cunha CB, Levi JE, Villa LL, Andrade CV, Russomano FB, Tristao A, Pires A, Nicol AF, 2013, A comparative analysis of clinical and molecular factors with the stage of cervical cancer in a Brazilian cohort. PLoS One 8(3): e57810 Mendoza L, Mongelos P, Paez M, Castro A, Rodriguez-Riveros I, Gimenez G, Araujo P, Echague G, Diaz V, Laspina F, Castro W, Jimenez R, Marecos R, Ever S, Deluca G, Picconi MA, 2013, Human papillomavirus and other genital infections in indigenous women from Paraguay: a cross-sectional analytical study. BMC Infect Dis 13(1):531 Simonella LM, Lewis H, Smith M, Neal H, Bromhead C, Canfell K, 2013, Type-specific oncogenic human papillomavirus infection in high grade cervical disease in New Zealand. BMC Infect Dis 13:114 Cornet I, Gheit T, Iannacone MR, Vignat J, Sylla BS, Del Mistro A, Franceschi S, Tommasino M, Clifford GM, 2013, HPV16 genetic variation and the development of cervical cancer worldwide. Br J Cancer 108(1):240–244 Tungteakkhun SS, Duerksen-Hughes PJ, 2008, Cellular binding partners of the human papillomavirus E6 protein. Arch Virol 153(3):397–408 Togtema M, Pichardo S, Jackson R, Lambert PF, Curiel L, Zehbe I, 2012, Sonoporation delivery of monoclonal antibodies against human papillomavirus 16 E6 restores p53 expression in transformed cervical keratinocytes. PLoS One 7(11):e50730 Lin HP, Wang YP, Chiang CP, 2011, Expression of p53, MDM2, p21, heat shock protein 70, and HPV 16/18 E6 proteins in oral verrucous carcinoma and oral verrucous hyperplasia. Head Neck 33(3):334–340 Doorbar J, Quint W, Banks L, Bravo IG, Stoler M, Broker TR, Stanley MA, 2012, The biology and life-cycle of human papillomaviruses. Vaccine 30(Suppl 5):F55–F70 Cuschieri K, Wentzensen N, 2008, Human papillomavirus mRNA and p16 detection as biomarkers for the improved diagnosis of cervical neoplasia. Cancer Epidemiol Biomarkers Prev 17(10): 2536–2545 Iijima N, Goodwin EC, Dimaio D, Iwasaki A, 2013, High-risk human papillomavirus E6 inhibits monocyte differentiation to Langerhans cells. Virology 444(1-2):257–262 Offringa RI, de Jong A, Toes RE, van der Burg SH, Melief CJ, 2003, Interplay between human papillomaviruses and dendritic cells. Curr Top Microbiol Immunol 276:215–240 Sasagawa T, Takagi H, Makinoda S, 2012, Immune responses against human papillomavirus, HPV, infection and evasion of host defense in cervical cancer. J Infect Chemother 18(6):807– 815 Niebler M, Qian X, Hofler D, Kogosov V, Kaewprag J, Kaufmann AM, Ly R, Bohmer G, Zawatzky R, Rosl F, Rincon-Orozco B, 2013, Post-translational control of IL-1β via the human papillomavirus type 16 E6 oncoprotein: a novel mechanismof innate immune escape mediated by the E3-ubiquitin ligase E6-AP and p53. PLoS Pathog 9(8):e1003536 Bodaghi S, Wood LV, Roby G, Ryder C, Steinberg SM, Zheng ZM, 2005, Could human papilloma viruses be spread through blood? J Clin Microbiol 43(11):5428–5434 Chen AC, Keleher A, Kedda MA, Spurdle AB, McMillan NA, Antonsson A, 2009, Human papillomavirus DNA detected in peripheral blood samples from healthy Australian male blood donors. J Med Virol 81(10):1792–1796 Foresta C, Bertoldo A, Garolla A, Pizzol D, Mason S, Lenzi A, De Toni L, 2013)Human papillomavirus proteins are found in peripheral blood and semen Cd20+ and Cd56+ cells during Hpv-16 semen infection. BMC Infect Dis 13:593 Reijmers RM, Spaargaren M, Pals ST, 2013, Heparan sulfate proteoglycans in the control of B cell development and the pathogenesis of multiple myeloma. FEBS J 280(10):2180–2193 Leemans CR, Braakhuis BJ, Brakenhoff RH, 2011, The molecular biology of head and neck cancer. Nat Rev Cancer 11:9–22 Carozzi F, Gillio-Tos A, Confortini M, Del Mistro A, Sani C, De Marco L, Girlando S, Rosso S, Naldoni C, Dalla Palma P, Zorzi M, Giorgi-Rossi P, Segnan N, Cuzick J, Ronco G, 2013, NTCC working group. risk of high-grade cervical intraepithelial neoplasia during follow-up in HPV-positive women according to baseline p16- INK4A results: a prospective analysis of a nested substudy of the NTCC randomised controlled trial. Lancet Oncol 14(2):168–176 Chiou HL, Wu MF, Liaw YC, Cheng YWSchiffman M, Castle PE, Jeronimo J, Rodriguez AC, Wacholder S, 2007, Human papillomavirus and cervical cancer. Lancet 370(9590):890–907 Chen TH, Huang CC, Yeh KT, Chang SH, Chang SW, Sung WW, Cheng YW, Lee H, 2012, Human papilloma virus 16 E6 oncoprotein associated with p53 inactivation in colorectal cancer. World J Gastroenterol 18(30):4051–8.14 Roncaglia MT, Fregnani JH, Tacla M, DE Campos SG, Caiaffa HH, Ab’saber A, DAMotta EV, Alves VA, Baracat EC, Longatto Filho A, 2013, Characterization of p16 and E6 HPV-related proteins in uterine cervix high-grade lesions of patients treated by conization with large loop excision. Oncol Lett 6(1):63–68 Giarnieri E, Mancini R, Pisani T, Alderisio M, Vecchione A, 2000, Msh2, Mlh1, Fhit, p53, Bcl-2, and Bax expression in invasive and in situ squamous cell carcinoma of the uterine cervix. Clin Cancer Res 6(9):3600–3606 Volgareva G, Zavalishina L, Andreeva Y, Frank G, Krutikova E, Golovina D, Bliev A, Spitkovsky D, Ermilova V, Kisseljov F, 2004, Protein p16 as a marker of dysplastic and neoplastic alterations in cervical epithelial cells. BMC Cancer 4:58 Hampl M, Wentzensen N, Vinokurova S, von Knebel-Doeberitz M, Poremba C, Bender HG, Kueppers V, 2007, Comprehensive analysis of 130 multicentric intraepithelial female lower genital tract lesions by HPV typing and p16 expression profile. J Cancer Res Clin Oncol 133(4):235–245 MimicaM, Tomic S, Kardum G, Hofman ID, Kaliterna V, Pejkovic L, 2010, Ki-67 quantitative evaluation as a marker of cervical intraepithelial neoplasia and human papillomavirus infection. Int J Gynecol Cancer 20(1):116–119 Wang JL, Zheng BY, Li XD, Nokelainen K, Angstrom T, Lindstrom MS, Wallin KL, 2005, p16INK4A and p14ARF expression pattern by immunohistochemistry in human papillomavirus-related cervical neoplasia. Mod Pathol 18(5):629–637 Murphy N, RingM, Killalea AG et al, 2003, p16INK4A as a marker for cervical dyskaryosis: CIN and cGIN in cervical biopsies and ThinPrep smears. J Clin Pathol 56:56–63 Jung YS, Kato I, Kim HR, 2013, A novel function of HPV16-E6/E7 in epithelial-mesenchymal transition. Biochem Biophys Res Commun 435(3):339–344 Qi ZL, Huo X, Xu XJ, Zhang B, DuMG, Yang HW, Zheng LK, Li J, Shen ZY, 2006, Relationship between HPV16/18 E6 and 53, 21WAF1, MDM2, Ki67 and cyclin D1 expression in esophageal squamous cell carcinoma: comparative study by using tissue microarray technology. Exp Oncol 28(3):235–240 Galgano MT, Castle PE, Atkins KA, Brix WK, Nassau SR, Stoler MH, 2010, Using biomarkers as objective standards in the diagnosis of cervical biopsies. Am J Surg Pathol 34(8):1077–1087 Nicol AF, Golub JE, e Silva JR, Cunha CB, Amaro-Filho SM, Oliveira NS, Menezes W, Andrade CV, Russomano F, Tristao A, Grinsztejn B, Friedman RK, Oliveira MP, Pires A, Nuovo GJ, 2012, An evaluation of p16, INK4a, expression in cervical intraepithelial neoplasia specimens, including women with HIV-1. Mem Inst Oswaldo Cruz 107(5):571–577 Cheah PL, Looi LM, Teoh KH, Mun KS, Nazarina AR, 2012, p16, INK4a, is a useful marker of human papillomavirus integration allowing risk stratification for cervical malignancies. Asian Pac J Cancer Prev 13(2):469–472 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 157 EP 166 DI 10.1007/s12253-014-9801-y PG 10 ER PT J AU Celik, B Khoor, A Bulut, T Nassar, A AF Celik, Betul Khoor, Andras Bulut, Tangul Nassar, Aziza TI Rapid On-Site Evaluation has High Diagnostic Yield Differentiating Adenocarcinoma vs Squamous Cell Carcinoma of Non–Small Cell Lung Carcinoma, not Otherwise Specified Subgroup SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cytology; Lung carcinoma; Non-small cell lung carcinoma; On-site cytologic evaluation; Subclassification ID Cytology; Lung carcinoma; Non-small cell lung carcinoma; On-site cytologic evaluation; Subclassification AB Our objective was to evaluate the diagnostic yield of rapid on-site evaluation (ROSE) on the differential diagnosis of non–small cell lung carcinoma, not otherwise specified (NSCLC-NOS). Biopsied cases diagnosed as NSCLC-NOS with ROSE during 2004 through 2008 were retrieved. Diagnostic confirmation was done with immunohistochemistry (IHC) involving thyroid transcription factor-1 and p63 immunostains. For the study, 106 cases were available. The final diagnoses rendered were squamous cell carcinoma (SqCC) (n=39) and adenocarcinoma (AC) (n=67). Cytologic, histologic, and IHC concordance for these diagnoses occurred in 75 cases (70.8 %), of which 56 (52.8 %) were AC and 19 (17.9 %) were SqCC. Cytologic, histologic, and IHC discordance was found in 31 cases (29.2 %). Of these 31 cases, 11 NSCLC-NOS diagnoses histologically corresponded to 1 SqCC plus 4 ACs, and 4 favor SqCC plus 2 ACs; the former 5 NSCLC-NOS cases classified correctly through cytology, as well as IHC. However, IHC was not available for the latter 6 NSCLC-NOS cases that were also classified correctly through cytology. In addition, only 3 NSCLC-NOS diagnoses cytologically corresponded to 3 favor SqCC histologically, in which IHC was not available, and for 2 cases that both corresponded to favor SqCC and favor AC histologically and cytologically. In the other 15 cases, histology labeled 4 cases NSCLC-NOS and misclassified 2 cases; cytology labeled 1 case NSCLC-NOS and misclassified 13 cases. ROSE has high diagnostic yield over subclassification of NSCLC-NOS. We recommend allocating a cytotechnologist for specimen adequacy and a cytopathologist for cytologic diagnosis. C1 [Celik, Betul] Mayo Clinic, Department of Laboratory Medicine and Pathology, 4500 San Pablo Rd, 32224 Jacksonville, FL, USA. [Khoor, Andras] Mayo Clinic, Department of Laboratory Medicine and Pathology, 4500 San Pablo Rd, 32224 Jacksonville, FL, USA. [Bulut, Tangul] Antalya Training and Research Hospital, Department of PathologyAntalya, Turkey. [Nassar, Aziza] Mayo Clinic, Department of Laboratory Medicine and Pathology, 4500 San Pablo Rd, 32224 Jacksonville, FL, USA. RP Nassar, A (reprint author), Mayo Clinic, Department of Laboratory Medicine and Pathology, 32224 Jacksonville, USA. EM nassar.aziza@mayo.edu CR Travis WD, Rekhtman N, Riley GJ, Geisinger KR, Asamura H, Brambilla E et al, 2010, Pathologic diagnosis of advanced lung cancer based on small biopsies and cytology: a paradigm shift. J Thorac Oncol 5(4):411–414 Agackiran Y, Ozcan A, Akyurek N, Memis L, Findik G, Kaya S, 2012, Desmoglein-3 and napsin a double stain, a useful immunohistochemical marker for differentiation of lung squamous cell carcinoma and adenocarcinoma from other subtypes. Appl Immunohistochem Mol Morphol 20(4):350–355 da Cunha Santos G, Ko HM, Saieg MA, Geddie WR, 2013, “The petals and thorns” of ROSE, rapid on-site evaluation). Cancer Cytopathol 121(1):4–8, Epub 2012 Jul 3 Collins BT, Chen AC, Wang JF, Bernadt CT, Sanati S, 2013, Improved laboratory resource utilization and patient care with the use of rapid on-site evaluation for endobronchial ultrasound fineneedle aspiration biopsy. Cancer Cytopathol 121(10):544–551, Epub 2013 Jul 3 TravisWD, Brambilla E, NoguchiM, Nicholson AG, Geisinger KR, Yatabe Y et al, 2011, International association for the study of lung cancer/american thoracic society/european respiratory society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol 6(2):244–285 Whitson BA, Groth SS, Odell DD, Briones EP, Maddaus MA, D’Cunha J et al, 2013, True negative predictive value of endobronchial ultrasound in lung cancer: are we being conservative enough? Ann Thorac Surg 95(5):1689–1694, Epub 2012 Dec 13 Dumonceau JM, Koessler T, van Hooft JE, Fockens P, 2012, Endoscopic ultrasonography-guided fine needle aspiration: relatively low sensitivity in the endosonographer population. World J Gastroenterol 18(19):2357–2363 Oki M, Saka H, Kitagawa C, Kogure Y, Murata N, Adachi T et al, 2013, Rapid on-site cytologic evaluation during endobronchial ultrasound-guided transbronchial needle aspiration for diagnosing lung cancer: a randomized study. Respiration 85(6):486–492, Epub 2013 Apr 3 Schmidt RL, Howard K, Hall BJ, Layfield LJ, 2012, The comparative effectiveness of fine-needle aspiration cytology sampling policies: a simulation study. Am J Clin Pathol 138(6):823–830 Khurana KK, Kovalovsky A,Wang D, Lenox R, 2013, Feasibility of dynamic telecytopathology for rapid on-site evaluation of endobronchial ultrasound-guided transbronchial fine needle aspiration. Telemed J E Health 19(4):265–271 Karnak D, Ciledag A, Ceyhan K, Atasoy C, Akyar S, Kayacan O, 2013, Rapid on-site evaluation and low registration error enhance the success of electromagnetic navigation bronchoscopy. Ann Thorac Med 8(1):28–32 Koegelenberg CF, Diacon AH, Irusen EM, von Groote-Bidlingmaier F, Mowlana A, Wright CA et al, 2011, The diagnostic yield and safety of ultrasound-assisted transthoracic biopsy of mediastinal masses. Respiration 81(2):134–141, Epub 2010 Dec 2 Trisolini R, Cancellieri A, Tinelli C, Paioli D, Scudeller L, Casadei GP et al, 2011, Rapid on-site evaluation of transbronchial aspirates in the diagnosis of hilar and mediastinal adenopathy: a randomized trial. Chest 139(2):395–401, Epub 2010 Oct 28 Griffin AC, Schwartz LE, Baloch ZW, 2011, Utility of on-site evaluation of endobronchial ultrasound-guided transbronchial needle aspiration specimens. Cytojournal 8:20, Epub 2011 Nov 21 Hebert-Magee S, Bae S, Varadarajulu S, Ramesh J, Frost AR, Eloubeidi MA et al, 2013, The presence of a cytopathologist increases the diagnostic accuracy of endoscopic ultrasound-guided fine needle aspiration cytology for pancreatic adenocarcinoma: a meta-analysis. Cytopathology 24(3):159–171 Ecka RS, SharmaM(2013, Rapid on-site evaluation of EUS-FNAby cytopathologist: an experience of a tertiary hospital. Diagn Cytopathol 41(12):1075–1080, Epub 2013 Oct 25 NicholsonAG,GonzalezD, Shah P, PynegarMJ,DeshmukhM, Rice A et al, 2010, Refining the diagnosis and EGFR status of non-small cell lung carcinoma in biopsy and cytologicmaterial, using a panel of mucin staining, TTF-1, cytokeratin 5/6, and P63, and EGFRmutation analysis. J Thorac Oncol 5(4):436–441 Hu Y, Puri V, Crabtree TD, Kreisel D, Krupnick AS, Patterson AG et al, 2013, Attaining proficiency with endobronchial ultrasoundguided transbronchial needle aspiration. J Thorac Cardiovasc Surg 146(6):1387–1392.e1, Epub 2013 Sep 24 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 167 EP 172 DI 10.1007/s12253-014-9802-x PG 6 ER PT J AU Dede, K Salamon, F Landherr, L Jakab, F Bursics, A AF Dede, Kristof Salamon, Ferenc Landherr, Laszlo Jakab, Ferenc Bursics, Attila TI Pathologic Assessment of Response to Chemotherapy in Colorectal Cancer Liver Metastases after Hepatic Resection: Which Method to Use? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal cancer liver metastases; Preoperative chemotherapy; Pathologic response ID colorectal cancer liver metastases; Preoperative chemotherapy; Pathologic response AB Background: Patients with metastatic colorectal cancer receive chemotherapy prior liver resection more and more frequently. Histopathologic assessmentmethods of the resected specimen could evaluate the response to chemotherapy. In this study it is analyzed if these histopathologic changes are specific to preoperative chemotherapy and if these methods have correlation with survival. Methods: Sixty three patients with available pathology slides, resected for colorectal cancer liver metastases were enrolled in this study. 46 patients (73 %) received neoadjuvant chemotherapy. Five pathological evaluation methods were compared according to the literature: [1] residual tumor cell ratio, [2] tumor regression grade (TRG) scoring system, [3] modified tumor regression grade (mTRG) scoring system with the type of necrosis, [4] pattern of tumor regression and [5] the tumor thickness at the tumor-normal interface (TNI). Results: Analyzing the pathological methods between the chemotherapy (CTX) and the non-chemotherapy group (NC), we found that that four evaluation methods showed significant and one showed strong correlation with the use of chemotherapy. (Residual tumor cell ratio: p=0.08; TRG: p <0.01; mTRG: p=0.03; pattern of tumor regression: p <0.01; TNI: p=0.02). In the chemotherapy group none of the analyzed pathological methods showed significant correlation with progression free survival (PFS) or with overall survival (OS). Residual tumor cell ratio, TRG and the pattern of tumor cells showed positive but not significant correlation with OS and PFS and a slight difference in the group of patients with TNI <2 mm could be documented. Conclusions: Tumor regression grade (TRG) and tumor thickness at the tumor-normal interface (TNI) were the most useful methods for pathological response evaluation and these methods had some correlation with survival. According to these data, authors concluded, that a reproducible and well defined scoring system, based on different histopathological evaluation methods should be developed to predict more accurately the effect of neoadjuvant chemotherapy in CRCLM patients. C1 [Dede, Kristof] Fovarosi Onkormanyzat Uzsoki utcai Korhaz, Sebeszeti-Onkosebeszeti Osztaly, Uzsoki utca 29, 1145 Budapest, Hungary. [Salamon, Ferenc] Fovarosi Uzsoki utcai Korhaz, PathologiaBudapest, Hungary. [Landherr, Laszlo] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai KozpontBudapest, Hungary. [Jakab, Ferenc] Fovarosi Onkormanyzat Uzsoki utcai Korhaz, Sebeszeti-Onkosebeszeti Osztaly, Uzsoki utca 29, 1145 Budapest, Hungary. [Bursics, Attila] Fovarosi Onkormanyzat Uzsoki utcai Korhaz, Sebeszeti-Onkosebeszeti Osztaly, Uzsoki utca 29, 1145 Budapest, Hungary. RP Dede, K (reprint author), Fovarosi Onkormanyzat Uzsoki utcai Korhaz, Sebeszeti-Onkosebeszeti Osztaly, 1145 Budapest, Hungary. EM dede.kristof@gmail.com CR Nordlinger B, Vauthey J-N, Poston G et al, 2010, The timing of chemotherapy and surgery for the treatment of colorectal liver metastases. Clin Colorectal Cancer 9(4):212–218 Chua T. C., Saxena A., Liauw A., et al.: Systematic review of randomized and nonrandomized trials of the clinical response and outcomes of neoadjuvant systemic chemotherapy for resectable colorectal liver metastases Ann Surg Oncol. 2010, 2):492–501., 17. Gruenberger B, Tamandl D, Schueller J et al, 2008, Bevacizumab, capecitabine, and oxaliplatin as neoadjuvant therapy for patients with potentially curable metastatic colorectal cancer. J Clin Oncol 26(11): 1830–1835 Nordlinger B, Van Cutsem E, Gruenberger T et al, 2009, Combination of surgery and chemotherapy and the role of targeted agents in the treatment of patients with colorectal liver metastases: recommendations from an expert panel. Ann Oncol 20(6):985–92 Adam R, Delvart V, Pascal G et al, 2004, Rescue surgery for unresectable colorectal liver metastases downstaged by chemotherapy: A model to predict long-term survival. Ann Surg 240:644–657 Dede K, Mersich T, Besznyak I et al, 2013, Bevacizumab treatment before resection of colorectal liver metastases: safety, recovery of liver function, pathologic assesment. Pathol Oncol Res 19:501–508 Scoggins CR, Campbell ML, Landry CS et al, 2009, Preoperative chemotherapy does not increase morbidity or mortality of hepatic resection for colorectal cancer metastases. Ann Surg Oncol 16(1):35–41 Therasse P, Arbuck SG, Eisenhauer EA et al, 2000, New guidelines to evaluate the response to treatment is solid tumors. J Natl Cancer Inst 92:205–216 ChunYS, Vauthey JN, Boonsirikamchai P et al. Association of Computed Tomography Morphologic Criteria With Pathologic Response and Survival in Patients Treated With Bevacizumab for Colorectal LiverMetastases JAMA. 2 [Epub ahead of print].009 302, 21):2338–2344. Dan G, Blazer III, Yoji K et al, 2008, Pathologic response to preoperative chemotherapy: a New outcome End point after resection of hepatic colorectal metastases. J Clin Onc 25(33):5344–5351 Rubbia-Brandt L, Giostra E, Brezault C et al, 2007, Importance of histological tumor response assessment in predicting the outcome in patients with colorectal liver metastases treated with neoadjuvant chemotherapy followed by liver surgery. Ann Oncol 18:299–304 Chang HHI, LeeperWR, Chan G et al, 2012, Infarct-like necrosis: a distinct formof necrosis seen in colorectal carcinoma liver metastases treated with perioperative chemotherapy. Am J Surg Pathol 36:570–576 Ribero D, Wang H, DonadonM et al, 2007, Bevacizumab improves pathologic response and protects against hepatic injury in patients treated with oxaliplatin-based chemotherapy for colorectal liver metastases. Cancer 110(12):2761–2767 Maru DM, Kopetz S, Boonsirikamchai P et al, 2010, Tumor thickness at the tumor-normal interface: a novel pathologic indicator of chemotherapy response in hepatic colorectal metastases. Am J Surg Pathol 34:1287–129 Gomez Dorronsoro ML, Vera R, Ortega L, et al. Recommendations of a group of experts for the pathological assessment of tumour regression of liver metastases of colorectal cancer and damage of non-tumour liver tissue after neoadjuvant therapy Clin Transl Oncol. [Epub ahead of print]. Knijn N, de Ridder JA, Punt CJ et al, 2013, Histopathological evaluation of resected colorectal cancer liver metastases: what should be done? Histopathology 63(2):149–56 Ng JKS, Urbanski SJ, Mangat N et al, 2008, Colorectal liver metastases contract centripetally with a response to chemotherapy. Cancer 112:362–71 Dede K, Lang I, Porneczi B et al, 2013, Preoperative chemotherapy in the surgical treatment of colorectal liver metastases. Magy Seb 66(6):325–30 Shindoh J, Loyer EM, Kopetz S et al, 2012, Optimal morphologic response to preoperative chemotherapy: an alternate outcome End point before resection of hepatic colorectal metastases. J Clin Oncol 30:4566–4572 Egger ME, Cannon RM, Metzger TL et al, 2013, Assessment of chemotherapy response in colorectal liver metastases in patients undergoing hepatic resection and the correlation to pathologic residual viable tumor. J Am Coll Surg 216:845–56 Poultsides GA, Bao F, Servais EL et al, 2012, Pathologic response to preoperative chemotherapy in colorectal liver metastases: fibrosis, not necrosis. Predicts outcome. Ann Surg Oncol 19:2797–2804 Gruenberger T, Arnold D, Rubbia-Brandt L, 2012, Pathologic response to bevacizumab-containing chemotherapy in patients with colorectal liver metastases and its correlation with survival. Surg Oncol 21:309–15 Loupakis F, Schirripa M, CaparelloC et al.: Histopathologic evaluation of liver metastases from colorectal cancer in patients treated with FOLFOXIRI plus bevacizumab Br J Cancer. 2013. [Epub ahead of print]. Klinger M, Tamandl D, Eipeldauer S et al, 2010, Bevacizumab improves pathological response of colorectal cancer liver metastases treated with XELOX/FOLFOX. Ann Surg Oncol 17(8):2059–65 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 173 EP 179 DI 10.1007/s12253-014-9803-9 PG 7 ER PT J AU An, HCh Je, ME Yoo, JN Lee, HS AF An, Hyeok Chang Je, Mi Eun Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutations of Cadherin Genes DCHS2, CDH10 and CDH24 Genes in Gastric and Colorectal Cancers with High Microsatellite Instability SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DHCS2; CDH10; CDH24; Cadherin; Mutation; Cancer; Microsatellite instability ID DHCS2; CDH10; CDH24; Cadherin; Mutation; Cancer; Microsatellite instability AB Cadherins (CDHs) are important in maintenance of cell adhesion and polarity, alterations of which contribute to tumorigenesis. Alterations of E-cadherin, a prototype CDH, have been reported in many cancers. However, alterations of unconventional CDHs, including CDH10, CDH24 and DCHS2 are largely unknown in cancers. Aim of this study was to explore whether CDH10, CDH24 and DCHS2 genes are mutated in gastric (GC) and colorectal cancers (CRC). In a public database, we found that CDH10, CDH24 and DCHS2 genes had mononucleotide repeats in the coding sequences that might be mutation targets in the cancers with microsatellite instability (MSI).We analyzed the mutations in 89GC and 131 CRC (high MSI (MSI-H) or stable MSI/low MSI (MSS/ MSI-L)) by single-strand conformation polymorphism analysis and DNA sequencing. We found six DCHS2, one CDH10 and one CDH24 frameshift mutations in them. All of the mutations were detected in cancers with MSI-H and there was a statistical difference in the frameshift mutation frequencies between the cancers with MSI-H (8/105) and MSS/MSIL (0/115). The DCHS2 frameshift mutations were found in 8.8 % and 4.2 % of GC and CRC with MSI-H respectively. Our results show that unconventional CDH10, CDH24 and DCHS2 genes harbored frameshift mutations. These mutations might inactivate the cell adhesion-related functions and could be a feature of GC and CRC with MSI-H. C1 [An, Hyeok Chang] The Catholic University of Korea, College of Medicine, Department of General SurgerySeoul, South Korea. [Je, Mi Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Hulpiau P, van Roy F, 2009, Molecular evolution of the cadherin superfamily. Int J Biochem Cell Biol 41:349–369 Angst BD, Marcozzi C, Magee AI, 2001, The cadherin superfamily: diversity in form and function. J Cell Sci 114(Pt 4):629–941 Gooding JM, Yap KL, IkuraM(2004, The cadherin-catenin complex as a focal point of cell adhesion and signalling: new insights from three-dimensional structures. Bioessays 26:497–511 Kalluri R, Weinberg RA, 2009, The basics of epithelialmesenchymal transition. J Clin Invest 119:1420–1428 Tepass U, Truong K, GodtD et al, 2000, Cadherins in embryonic and neural morphogenesis. Nat Rev Mol Cell Biol 1:91–100 Gottardi CJ, Wong E, Gumbiner BM, 2001, E-cadherin suppresses cellular transformation by inhibiting beta-catenin signaling in an adhesion-independent manner. J Cell Biol 153:1049–1060 Hirohashi S, 1998, Inactivation of the E-cadherin-mediated cell adhesion system in human cancers. Am J Pathol 153:333–339 Muta H, NoguchiM, Kanai Yet al, 1996, E-cadherin genemutations in signet ring cell carcinoma of the stomach. Jpn J Cancer Res 87: 843–848 Williams MJ, Lowrie MB, Bennett JP et al, 2005, Cadherin-10 is a novel blood–brain barrier adhesion molecule in human and mouse. Brain Res 1058:62–72 Walker MM, Ellis SM, Auza MJ et al, 2008, The intercellular adhesion molecule, cadherin-10, is a marker for human prostate luminal epithelial cells that is not expressed in prostate cancer. Mod Pathol 21:85–95 Katafiasz BJ, Nieman MT, Wheelock MJ et al, 2003, Characterization of cadherin-24, a novel alternatively spliced type II cadherin. J Biol Chem 278:27513–27519 Hong JC, Ivanov NV, Hodor P et al, 2004, Identification of new human cadherin genes using a combination of protein motif search and gene finding methods. J Mol Biol 337:307–317 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Murphy K, Zhang S, Geiger T et al, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8:305–311 Kim YR, Oh JE, KimMS et al, 2010, Oncogenic NRF2 mutations in squamous cell carcinomas of oesophagus and skin. J Pathol 220:446– 451 Yoo NJ, Kim HR, Kim YR et al, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60: 943–952 Je EM, Kim MR, Min KO et al, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–1047 BirchmeierW, Behrens J, 1994, Cadherin expression in carcinomas: role in the formation of cell junctions and the prevention of invasiveness. Biochim Biophys Acta 1198:11–26 Sato Y, Yoshizato T, Shiraishi Y et al, 2013, Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet 45:860–867 Barbieri CE, Baca SC, LawrenceMS et al, 2012, Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet 44:685–689 Cancer Genome Atlas Network, 2012, Comprehensive molecular characterization of human colon and rectal cancer. Nature 487:330- –337 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 181 EP 185 DI 10.1007/s12253-014-9804-8 PG 5 ER PT J AU Pula, B Tazbierski, T Zamirska, A Werynska, B Bieniek, A Szepietowski, J Rys, J Dziegiel, P Podhorska-Okolow, M AF Pula, Bartosz Tazbierski, Tadeusz Zamirska, Aleksandra Werynska, Bozena Bieniek, Andrzej Szepietowski, Jacek Rys, Janusz Dziegiel, Piotr Podhorska-Okolow, Marzena TI Metallothionein 3 Expression in Normal Skin and Malignant Skin Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Metallothionein-3; Skin cancer; Squamous cell carcinoma; Basal cell carcinoma ID Metallothionein-3; Skin cancer; Squamous cell carcinoma; Basal cell carcinoma AB Metallothionein-3 (MT-3) has been shown to be expressed in several malignancies and to have an impact on patients’ survival in breast and urinary bladder cancer cases. However, its expression has not been determined in normal skin or in its malignant lesions. MT-3 expression was studied using immunohistochemistry in 17 cases of normal skin, 18 of actinic keratosis (AK), 39 of squamous cell carcinoma (SCC), and 23 of basal cell carcinoma (BCC). Low MT-3 expression was observed in normal skin epidermis with faint or no expression in the epidermis basal layer. Significantly higher MT-3 expression was noted in AK (P=0.007) and SCC (P<0.0001), as compared with normal skin epidermis. BCC cases were characterized by the lowest MT-3 expression of all the examined groups, which was significantly lower in comparison to normal skin epidermis, AK, and SCC (P=0.009; P<0.0001 and P<0.0001, respectively). In conclusion, MT-3 may be involved in the development of SCC. C1 [Pula, Bartosz] Wroclaw Medical University, Department of Human Morphology and Embryology, Chalubinskiego 6a, 50-368 Wroclaw, Poland. [Tazbierski, Tadeusz] Wroclaw Medical University, Department of Human Morphology and Embryology, Chalubinskiego 6a, 50-368 Wroclaw, Poland. [Zamirska, Aleksandra] Wroclaw Medical University, Department of Dermatology, Venereology and AllergologyWroclaw, Poland. [Werynska, Bozena] Medical University of Wroclaw, Department of Pulmonology and Pulmonary TumorsWroclaw, Poland. [Bieniek, Andrzej] Wroclaw Medical University, Department of Dermatology, Venereology and AllergologyWroclaw, Poland. [Szepietowski, Jacek] Wroclaw Medical University, Department of Dermatology, Venereology and AllergologyWroclaw, Poland. [Rys, Janusz] Maria Sklodowska-Curie Memorial Institute Oncology Center, Department of Tumor PathologyKrakow, Poland. [Dziegiel, Piotr] Wroclaw Medical University, Department of Human Morphology and Embryology, Chalubinskiego 6a, 50-368 Wroclaw, Poland. [Podhorska-Okolow, Marzena] Wroclaw Medical University, Department of Human Morphology and Embryology, Chalubinskiego 6a, 50-368 Wroclaw, Poland. RP Podhorska-Okolow, M (reprint author), Wroclaw Medical University, Department of Human Morphology and Embryology, 50-368 Wroclaw, Poland. EM marzenna.podhorska-okolow@umed.wroc.pl CR Marks R, 1995, The epidemiology of non-melanoma skin cancer: who, why and what can we do about it. J Dermatol 22(11):853–857 Tufaro AP, Chuang JC, Prasad N, Chuang A, Chuang TC, Fischer AC, 2011, Molecular markers in cutaneous squamous cell carcinoma. Int J Surg Oncol 2011:231475., DOI 10.1155/2011/231475 Zalaudek I, Whiteman D, Rosendahl C, Menzies SW, Green AC, Hersey P, Argenziano G, 2011, Update on melanoma and nonmelanoma skin cancer. Annual Skin Cancer Conference 2011, Hamilton Island, Australia, 5–6 August 2011. Expert Rev Anticancer Ther 11(12):1829–1832., DOI 10.1586/era.11.180 Rubin AI, Chen EH, Ratner D, 2005, Basal-cell carcinoma. N Engl J Med 353(21):2262–2269., DOI 10.1056/NEJMra044151 Crowson AN, 2006, Basal cell carcinoma: biology, morphology and clinical implications. Mod Pathol 19(Suppl 2):S127–147., DOI 10. 1038/modpathol.3800512 Goppner D, Leverkus M, 2011, Basal cell carcinoma: from the molecular understanding of the pathogenesis to targeted therapy of progressive disease. J Skin Cancer 2011:650258., DOI 10.1155/2011/650258 Coyle P, Philcox JC, Carey LC, Rofe AM, 2002, Metallothionein: the multipurpose protein. Cell Mol Life Sci 59(4):627–647 Dziegiel P, 2004, Expression ofmetallothioneins in tumor cells. Pol J Pathol 55(1):3–12 Nielsen AE, Bohr A, Penkowa M, 2007, The Balance between Life and Death of Cells: Roles of Metallothioneins. Biomark Insights 1:99–111 Pula B, Domoslawski P, Podhorska-Okolow M, Dziegiel P, 2012, Role of metallothioneins in benign and malignant thyroid lesions. Thyroid Res 5(1):26., DOI 10.1186/1756-6614-5-26 Thirumoorthy N, Shyam Sunder A, Manisenthil Kumar K, Senthil Kumar M, Ganesh G, Chatterjee M, 2011, A review of metallothionein isoforms and their role in pathophysiology. World J Surg Oncol 9:54., DOI 10.1186/1477-7819-9-54 Dziegiel P, Salwa-Zurawska W, Zurawski J, Wojnar A, Zabel M, 2005, Prognostic significance of augmented metallothionein, MT, expression correlated with Ki-67 antigen expression in selected soft tissue sarcomas. Histol Histopathol 20(1):83–89 Surowiak P, Matkowski R, Materna V, Gyorffy B, Wojnar A, Pudelko M, Dziegiel P, Kornafel J, Zabel M, 2005, Elevated metallothionein, MT, expression in invasive ductal breast cancers predicts tamoxifen resistance. Histol Histopathol 20(4):1037–1044 Wojnar A, Pula B, Piotrowska A, Jethon A, Kujawa K, Kobierzycki C, Rys J, Podhorska-Okolow M, Dziegiel P, 2011, Correlation of intensity of MT-I/II expression with Ki-67 and MCM-2 proteins in invasive ductal breast carcinoma. Anticancer Res 31(9):3027–3033 Surowiak P, Materna V, Kaplenko I, Spaczynski M, Dietel M, Lage H, Zabel M, 2005, Augmented expression of metallothionein and glutathione S-transferase pi as unfavourable prognostic factors in cisplatin-treated ovarian cancer patients. Virchows Arch 447(3): 626–633., DOI 10.1007/s00428-005-1228-0 Surowiak P, Materna V, Maciejczyk A, Pudelko M, Markwitz E, Spaczynski M, Dietel M, Zabel M, Lage H, 2007, Nuclear metallothionein expression correlates with cisplatin resistance of ovarian cancer cells and poor clinical outcome. Virchows Arch 450(3):279– 285., DOI 10.1007/s00428-006-0362-7 Tuzel E, Kirkali Z, Yorukoglu K, Mungan MU, Sade M, 2001, Metallothionein expression in renal cell carcinoma: subcellular localization and prognostic significance. J Urol 165(5):1710–1713 Dziegiel P, Jelen M, Muszczynska B, Maciejczyk A, Szulc A, Podhorska-Okolow M, Cegielski M, Zabel M, 2004, Role of metallothionein expression in non-small cell lung carcinomas. Rocz Akad Med Bialymst 49(Suppl 1):43–45 Werynska B, Pula B, Muszczynska-Bernhard B, Gomulkiewicz A, Piotrowska A, Prus R, Podhorska-OkolowM, Jankowska R,Dziegiel P, 2013, Metallothionein 1 F and 2A overexpression predicts poor outcome of non-small cell lung cancer patients. Exp Mol Pathol 94(1):301–308., DOI 10.1016/j.yexmp.2012.10.006 Dziegiel P, Forgacz J, Suder E, Surowiak P, Kornafel J, Zabel M, 2003, Prognostic significance of metallothionein expression in correlation with Ki-67 expression in adenocarcinomas of large intestine. Histol Histopathol 18(2):401–407 Uchida Y, Tomonaga M, 1989, Neurotrophic action of Alzheimer’s disease brain extract is due to the loss of inhibitory factors for survival and neurite formation of cerebral cortical neurons. Brain Res 481(1): 190–193 Somji S, Garrett SH, Zhou XD, Zheng Y, Sens DA, SensMA, 2010, Absence ofMetallothionein 3 Expression in Breast Cancer is a Rare, But Favorable Marker of Outcome that is Under Epigenetic Control. Toxicol Environ Chem 92(9):1673–1695., DOI 10.1080/ 02772241003711274 Deng D, El-RifaiW, Ji J, Zhu B, Trampont P, Li J, SmithMF, Powel SM, 2003, Hypermethylation of metallothionein-3 CpG island in gastric carcinoma. Carcinogenesis 24(1):25–29 Garrett SH, Sens MA, Shukla D, Nestor S, Somji S, Todd JH, Sens DA, 1999, Metallothionein isoform 3 expression in the human prostate and cancer-derived cell lines. Prostate 41(3):196–202 Garrett SH, SensMA, Todd JH, Somji S, Sens DA(1999, Expression of MT-3 protein in the human kidney. Toxicol Lett 105(3):207–214 Kim D, Garrett SH, Sens MA, Somji S, Sens DA, 2002, Metallothionein isoform 3 and proximal tubule vectorial active transport. Kidney Int 61(2):464–472., DOI 10.1046/j.1523-1755. 2002.00153.x Sens MA, Somji S, Lamm DL, Garrett SH, Slovinsky F, Todd JH, Sens DA, 2000, Metallothionein isoform 3 as a potential biomarker for human bladder cancer. Environ Health Perspect 108(5):413– 418 Somji S, Garrett SH, SensMA, Gurel V, Sens DA, 2004, Expression of metallothionein isoform 3, MT-3, determines the choice between apoptotic or necrotic cell death in Cd+2-exposed human proximal tubule cells. Toxicol Sci 80(2):358–366., DOI 10.1093/toxsci/kfh158 Smith E, Drew PA, Tian ZQ, De Young NJ, Liu JF, Mayne GC, Ruszkiewicz AR,Watson DI, JamiesonGG, 2005)Metallothionien 3 expression is frequently down-regulated in oesophageal squamous cell carcinoma by DNA methylation. Mol Cancer 4:42., DOI 10.1186/ 1476-4598-4-42 PengD, Hu TL, Jiang A,WashingtonMK,Moskaluk CA, Schneider- Stock R, El-Rifai W, 2011, Location-specific epigenetic regulation of the metallothionein 3 gene in esophageal adenocarcinomas. PLoS One 6(7):e22009., DOI 10.1371/journal.pone.0022009 Bieniek A, Pula B, Piotrowska A, Podhorska-Okolow M, Salwa A, Koziol M, Dziegiel P, 2012, Expression of metallothionein I/II and Ki-67 antigen in various histological types of basal cell carcinoma. Folia Histochem Cytobiol 50(3):352– 357., DOI 10.5603/19744 Zamirska A, Matusiak L, Dziegiel P, Szybejko-Machaj G, Szepietowski JC, 2012, Expression ofmetallothioneins in cutaneous squamous cell carcinoma and actinic keratosis. Pathol Oncol Res 18(4):849–855., DOI 10.1007/s12253-012-9513-0 AnnerothG, Batsakis J, LunaM(1987, Review of the literature and a recommended system of malignancy grading in oral squamous cell carcinomas. Scand J Dent Res 95(3):229–249 Remmele W, Stegner HE, 1987, Recommendation for uniform definition of an immunoreactive score, IRS, for immunohistochemical estrogen receptor detection, ER-ICA, in breast cancer tissue. Pathologe 8(3):138–140 Palmiter RD, Findley SD, Whitmore TE, Durnam DM, 1992, MTIII, a brain-specific member of the metallothionein gene family. Proc Natl Acad Sci U S A 89(14):6333–6337 Uchida Y, Takio K, Titani K, Ihara Y, TomonagaM(1991, The growth inhibitory factor that is deficient in the Alzheimer’s disease brain is a 68 amino acid metallothionein-like protein. Neuron 7(2):337–347 Sens MA, Somji S, Garrett SH, Beall CL, Sens DA, 2001, Metallothionein isoform 3 overexpression is associated with breast cancers having a poor prognosis. Am J Pathol 159(1):21–26., DOI 10. 1016/S0002-9440(10)61668-9 Werynska B, Pula B, Muszczynska-Bernhard B, Gomulkiewicz A, Jethon A, Podhorska-Okolow M, Jankowska R, Dziegiel P, 2013, Expression of metallothionein-III in patients with non-small cell lung cancer. Anticancer Res 33(3):965–974 Quaife CJ, Findley SD, Erickson JC, Froelick GJ, Kelly EJ, Zambrowicz BP, Palmiter RD, 1994, Induction of a new metallothionein isoform, MT-IV, occurs during differentiation of stratified squamous epithelia. Biochemistry 33(23):7250–7259 Lamore SD, Wondrak GT, 2011, Zinc pyrithione impairs zinc homeostasis and upregulates stress response gene expression in reconstructed human epidermis. Biometals 24(5):875–890., DOI 10.1007/ s10534-011-9441-6 Kumar P, Lal NR, Mondal AK, Mondal A, Gharami RC, Maiti A, 2012, Zinc and skin: a brief summary. Dermatol Online J 18(3):1 Prasad AS, 1998, Zinc deficiency in humans: a neglected problem. J Am Coll Nutr 17(6):542–543 PedersenMO, Larsen A, StoltenbergM, PenkowaM(2009, The role of metallothionein in oncogenesis and cancer prognosis. Prog Histochem Cytochem 44(1):29–64., DOI 10.1016/j.proghi.2008.10. 001 Lee SJ, Koh JY, 2010, Roles of zinc and metallothionein-3 in oxidative stress-induced lysosomal dysfunction, cell death, and autophagy in neurons and astrocytes. Mol Brain 3(1):30., DOI 10.1186/1756-6606-3-30 Lee SJ, Park MH, Kim HJ, Koh JY, 2010, Metallothionein-3 regulates lysosomal function in cultured astrocytes under both normal and oxidative conditions. Glia 58(10):1186–1196., DOI 10.1002/glia.20998 Kim HG, Hwang YP, Han EH, Choi CY, Yeo CY, Kim JY, Lee KY, Jeong HG, 2009, Metallothionein-III provides neuronal protection through activation of nuclear factor-kappaB via the TrkA/phosphatidylinositol-3 kinase/Akt signaling pathway. Toxicol Sci 112(2):435–449., DOI 10.1093/toxsci/kfp230 Bito T, Sumita N, Ashida M, Budiyanto A, Ueda M, Ichihashi M, Tokura Y, Nishigori C, 2011, Inhibition of Epidermal Growth Factor Receptor and PI3K/Akt Signaling Suppresses Cell Proliferation and Survival through Regulation of Stat3 Activation in Human Cutaneous Squamous Cell Carcinoma. J Skin Cancer 2011:874571., DOI 10.1155/2011/874571 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 187 EP 193 DI 10.1007/s12253-014-9805-7 PG 7 ER PT J AU Tian, X Wang, Q Li, Y Hu, J Wu, L Ding, Q Zhang, Ch AF Tian, Xiangguo Wang, Qizhi Li, Yan Hu, Jinhua Wu, Lei Ding, Qian Zhang, Chunqing TI The Expression of S100A4 Protein in Human Intrahepatic Cholangiocarcinoma: Clinicopathologic Significance and Prognostic Value SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intrahepatic cholangiocarcinoma; Prognosis; S100A4; Matrix metalloproteinase-9 (MMP-9); Immunohistochemistry ID Intrahepatic cholangiocarcinoma; Prognosis; S100A4; Matrix metalloproteinase-9 (MMP-9); Immunohistochemistry AB Intrahepatic cholangiocarcinoma(ICC) is a highly malignant adenocarcinoma arising from bile duct epithelial cells of the intrahepatic biliary system with early hematogenous and lymphatic extrahepatic spread. The current treatment methods for ICC are far fromideal. Identifying novel effective prognostic biomarkers which might be related to the development and progression of ICC may help provide new therapeutic strategies. Both calcium-binding protein S100A4 and Matrix metalloproteinase-9(MMP-9) are correlated with development and progression of many carcinomas. In the present study, we investigated expression of S100A4 as well as MMP-9 in ICC tissues from 65 patients using immunohistochemistry. The correlation of S100A4 and MMP-9 expression with clinicopathological features and prognosis of patients were analyzed. S100A4 and MMP-9 were positively expressed in 32(49.2 %) and 35(53.8 %) patients, respectively. The positive correlation between S100A4 and MMP-9 expression was statistically significant (P=0.018). S100A4 positive expression was significantly correlated with vascular invasion (P=0.008), lymph node metastasis (P=0.029) and the TNM stage (P=0.008). MMP-9 expression was not found to be correlated with any clinicopathological parameter. Patients with S100A4 positive expression had a significantly poorer overall survival rate than those with S100A4 negative expression (P=0.000). MMP-9 positive expression was also correlated with poor survival (P=0.044). However, only S100A4 expression (P=0.004) and the surgical margin (P=0.024) were significantly independent prognostic predictors by multivariate analysis. In conclusion, expression of S100A4 is correlated with MMP-9 expression and it could be a useful marker for predicting the progression, metastasis and prognosis of ICC. C1 [Tian, Xiangguo] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. [Wang, Qizhi] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. [Li, Yan] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. [Hu, Jinhua] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. [Wu, Lei] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. [Ding, Qian] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. [Zhang, Chunqing] Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 324 Jingwu Weiqi Road, 250021 Jinan, China. RP Zhang, Ch (reprint author), Provincial Hospital Affiliated to Shandong University, Department of Gastroenterology, 250021 Jinan, China. EM zhangchunqing_sdu@163.com CR Rizvi S, Gores GJ, 2013, Pathogenesis, diagnosis, and management of cholangiocarcinoma. Gastroenterology 145:1215–1229 Gatto M, Bragaz z i MC, Semeraro R et al, 2010, Cholangiocarcinoma: update and future perspectives. Dig Liver Dis 42:253–260 Khan SA, Thomas HC, Davidson BR et al, 2005, Cholangiocarcinoma. Lancet 366:1303–1314 Dhanasekaran R, Hemming AW, Zendejas I et al, 2013, Treatment outcomes and prognostic factors of intrahepatic cholangiocarcinoma. Oncol Rep 29:1259–1267 Nagorney DM, Kendrick ML, 2006, Hepatic resection in the treatment of hilar cholangiocarcinoma. Adv Surg 40:159–171 Jarnagin WR, Shoup M, 2004, Surgical management of cholangiocarcinoma. Semin Liver Dis 24:189–199 Sirica AE, Dumur CI, Campbell DJ et al, 2009, Intrahepatic cholangiocarcinoma progression: prognostic factors and basic mechanisms. Clin Gastroenterol Hepatol 7:S68–S78 Mazzucchelli L, 2002, Protein S100A4: too long overlooked by pathologists? Am J Pathol 160:7–13 Boye K, Maelandsmo GM, 2010, S100A4 and metastasis: a small actor playing many roles. Am J Pathol 176:528–535 Huang L, Xu Y, Cai G et al, 2012, Downregulation of S100A4 expression by RNA interference suppresses cell growth and invasion in human colorectal cancer cells. Oncol Rep 27:917–922 Orre LM, Panizza E, Kaminskyy VO et al, 2013, S100A4 interacts with p53 in the nucleus and promotes p53 degradation. Oncogene 32: 5531–5540 Jia W, Gao XJ, Zhang ZD et al, 2013, S100A4 silencing suppresses proliferation, angiogenesis and invasion of thyroid cancer cells through downregulation of MMP-9 and VEGF. Eur Rev Med Pharmacol Sci 17:1495–1508 Ambartsumian N, Klingelhofer J, Grigorian M et al, 2001, The metastasis-associated Mts1(S100A4, protein could act as an angiogenic factor. Oncogene 20:4685–4695 Hemandas AK, Salto-Tellez M, Maricar SH et al, 2006, Metastasisassociated protein S100A4–a potential prognostic marker for colorectal cancer. J Surg Oncol 93:498–503 Ninomiya I, Ohta T, Fushida S et al, 2001, Increased expression of S100A4 and its prognostic significance in esophageal squamous cell carcinoma. Int J Oncol 18:715–720 Tsukamoto N, Egawa S, Akada M et al, 2013, The expression of S100A4 in human pancreatic cancer is associated with invasion. Pancreas 42:1027–1033 Matsumoto K, Irie A, Satoh Tet al, 2007, Expression of S100A2 and S100A4 predicts for disease progression and patient survival in bladder cancer. Urology 70:602–607 Wang YY, Ye ZY, Zhao ZS et al, 2010, High-level expression of S100A4 correlates with lymph nodemetastasis and poor prognosis in patients with gastric cancer. Ann Surg Oncol 17:89–97 Platt-Higgins AM, Renshaw CA, West CR et al, 2000, Comparison of the metastasis-inducing protein S100A4, p9ka, with other prognostic markers in human breast cancer. Int J Cancer 89:198–208 Liu Z, Liu H, Pan H et al, 2013, Clinicopathological significance of S100A4 expression in human hepatocellular carcinoma. J Int Med Res 41:457–462 Fabris L, CadamuroM,Moserle L et al, 2011, Nuclear expression of S100A4 calcium-binding protein increases cholangiocarcinoma invasiveness and metastasization. Hepatology 54:890–899 Gialeli C, Theocharis AD, Karamanos NK, 2011, Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting. FEBS J 278:16–27 Kessenbrock K, Plaks V, Werb Z, 2010, Matrix metalloproteinases: regulators of the tumor microenvironment. Cell 141:52–67 Shirabe K, ShimadaM, Kajiyama Ket al, 1999, Expression of matrix metalloproteinase-9 in surgically resected intrahepatic cholangiocarcinoma. Surgery 126:842–846 Zhang HY, Zheng XZ, Wang XH et al, 2012, S100A4 mediated cell invasion and metastasis of esophageal squamous cell carcinoma via the regulation of MMP-2 and E-cadherin activity. Mol Biol Rep 39: 199–208 Schmidt-Hansen B, Ornas D, Grigorian M et al, 2004, Extracellular S100A4(mts1, stimulates invasive growth of mouse endothelial cells and modulatesMMP-13 matrixmetalloproteinase activity. Oncogene 23:5487–5495 Bjornland K, Winberg JO, Odegaard OT et al, 1999, S100A4 involvement in metastasis: deregulation of matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases in osteosarcoma cells transfected with an anti-S100A4 ribozyme. Cancer Res 59: 4702–4708 Zhang G, Li M, Jin J et al, 2011, Knockdown of S100A4 decreases tumorigenesis and metastasis in osteosarcoma cells by repression of matrix metalloproteinase-9. Asian Pac J Cancer Prev 12:2075–2080 Saleem M, Kweon MH, Johnson JJ et al, 2006, S100A4 accelerates tumorigenesis and invasion of human prostate cancer through the transcriptional regulation of matrix metalloproteinase 9. Proc Natl Acad Sci U S A 103:14825–14830 Nakanuma Y, Sripa B, Vatanasapt V et al, 2000, Intrahepatic cholangiocarcinoma. In: Hamilton SR, Aaltonen LA, eds, World health organization classification of tumours: pathology and genetics of tumours of the digestive system. IARC Press, Lyon, pp 173–180 Cardinale V, Semeraro R, Torrice A et al, 2010, Intra-hepatic and extra-hepatic cholangiocarcinoma: new insight into epidemiology and risk factors. World J Gastrointest Oncol 2:407– 416 Schmidt-Hansen B, Klingelhofer J, Grum-Schwensen B et al, 2004, Functional significance of metastasis-inducing S100A4(Mts1, in tumor-stroma interplay. J Biol Chem 279: 24498–24504 Sato Y, Harada K, Sasaki M et al, 2013, Clinicopathological significance of S100 protein expression in cholangiocarcinoma. J Gastroenterol Hepatol 28:1422–1429 Shibahara H, Tamada S, Higashi M et al, 2004, MUC4 is a novel prognostic factor of intrahepatic cholangiocarcinoma-mass forming type. Hepatology 39:220–229 Sotiropoulos GC, Miyazaki M, Konstadoulakis MM et al, 2010, Multicentric evaluation of a clinical and prognostic scoring system predictive of survival after resection of intrahepatic cholangiocarcinoma. Liver Int 30:996–1002 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 195 EP 201 DI 10.1007/s12253-014-9806-6 PG 7 ER PT J AU Toth-Liptak, J Piukovics, K Borbenyi, Z Demeter, J Bagdi, E Krenacs, L AF Toth-Liptak, Judit Piukovics, Klara Borbenyi, Zita Demeter, Judit Bagdi, Eniko Krenacs, Laszlo TI A Comprehensive Immunophenotypic Marker Analysis of Hairy Cell Leukemia in Paraffin-Embedded Bone Marrow Trephine Biopsies–A Tissue Microarray Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hairy cell leukemia; Bone marrow trephine biopsy; Tissue microarray; Immunohistochemistry ID Hairy cell leukemia; Bone marrow trephine biopsy; Tissue microarray; Immunohistochemistry AB Hairy cell leukemia (HCL) is an uncommon B cell lymphoproliferation characterized by a unique immunophenotype. Due to low number of circulating neoplastic cells and ‘dry tap’ aspiration, the diagnosis is often based on BM trephine biopsy. We have performed a consecutive immunohistochemical analysis to evaluate diagnostic usefulness of various HCL markers (CD11c, CD25, CD68, CD103, CD123, CD200, annexin A1, cyclin D1, DBA.44, HBME-1, phospho-ERK1/2, TRAP, and T-bet) currently available against fixation resistant epitopes. We analyzed tissue microarrays consisting of samples gained from 73 small B-cell lymphoma cases, including hairy cell leukemia (HCL) (n=32), HCL variant (HCL-v) (n=4), B-cell chronic lymphocytic leukemia (B-CLL) (n=11), lymphoplasmacytic lymphoma (LPL) (n=3), mantle cell lymphoma (MCL) (n=10), splenic diffuse red pulp small B cell lymphoma (SDRPL) (n=2), splenic B cell marginal zone lymphoma (SMZL) (n=8), and splenic B cell lymphoma/leukemia, unclassifiable (SBCL) (n=3) cases. The HCL cases were 100 % positive for all but 2 (DBA.44 and CD123) of these markers. Annexin A1 showed 100 % specificity and accuracy, which was followed by CD123, pERK, CD103, HBME-1, CD11c, CD25, CD68, cyclin D1, CD200, T-bet, DBA.44, and TRAP, in decreasing order. In conclusion, our results reassured the high specificity of annexin A1 and pERK, as well as the diagnostic value of standard HCL markers of CD11c, CD25, CD103, and CD123 also in paraffin-embedded BM samples. Additional markers, including HBME-1, cyclin D1, CD200, and T-bet also represent valuable tools in the differential diagnosis of HCL and its mimics. C1 [Toth-Liptak, Judit] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular Diagnostics, Jobb fasor 23/B, 6726 Szeged, Hungary. [Piukovics, Klara] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Borbenyi, Zita] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Demeter, Judit] Semmelweis University, 1st Department of Internal MedicineBudapest, Hungary. [Bagdi, Eniko] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular Diagnostics, Jobb fasor 23/B, 6726 Szeged, Hungary. [Krenacs, Laszlo] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular Diagnostics, Jobb fasor 23/B, 6726 Szeged, Hungary. RP Krenacs, L (reprint author), Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular Diagnostics, 6726 Szeged, Hungary. EM krenacsl@vipmail.hu CR Foucar K, Falini B, Catovsky D, Stein H, 2008, Hairy cell leukemia. In: Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Vardiman JW, eds, WHO classification of tumours of haematopoietic and lymphoid tissues, vol 2, 4th edn. WHO classification of tumours. IARC, Lyon, pp 188–190 Robbins BA, Ellison DJ, Spinosa JC, Carey CA, Lukes RJ, Poppema S, Saven A, Piro LD, 1993, Diagnostic application of two-color flow cytometry in 161 cases of hairy cell leukemia. Blood 82:1277–1287 Sanchez ML, Almeida J, Vidriales B, Lopez-Berges MC, Garcia- Marcos MA, Moro MJ, Corrales A, Calmuntia MJ, San Miguel JF, Orfao A, 2002, Incidence of phenotypic aberrations in a series of 467 patients with B chronic lymphoproliferative disorders: basis for the design of specific four-color stainings to be used for minimal residual disease investigation. Leukemia 16:1460–1469 Del Guidice I, Matutes E, Morilla R, Owusu-Ankomah K, Rafiq F, A’Hern R, Delgado J, Bazerbashi MB, Catovsky D, 2004, The diagnostic value of CD123 in B cell disorders with hairy or villous lymphocytes. Haematologica 89:303–308 Chen YH, Tallman MS, Goolsby C, Peterson L, 2006, Immunophenotypic variations in hairy cell leukemia. Am J Clin Pathol 125:251–259., DOI 10.1309/PMQXVY619Q8Y43AR Dong HY,Weisberger J, Liu Z, Tugulea S, 2009, Immunophenotypic analysis of CD103+ B-lymphoproliferative disorders: hairy cell leukemia and its mimics. Am J Clin Pathol 131:586–595., DOI 10.1309/ AJCPL13YDUHFKPJU Venkataraman G, Aguhar C, Kreitman RJ, Yuan CM, Stetler- Stevenson M, 2011, Characteristic CD103 and CD123 expression pattern defines hairy cell leukemia: usefulness of CD123 and CD103 in the diagnosis ofmature B-cell lymphoproliferative disorders. AmJ Clin Pathol 136:625–630., DOI 10.1309/AJCPKUM9J4IXCWEU Matutes E, Wotherspoon A, Brito-Babapulle V, Catovsky D, 2001, The natural history and clinico-pathological features of the variant form of hairy cell leukemia. Leukemia 15:184–186 Cessna MH, Hartung L, Tripp S, Perkins SL, Bahler DW, 2005, Hairy cell leukemia variant: fact or fiction. Am J Clin Pathol 123: 132–138., DOI 10.1309/8QYTYQ1CLQMHQ9CL Piris M, Foucar K, Mollejo M, Campo E, Falini B, 2008, Splenic Bcell lymphoma/leukaemia, unclassifiable. In: Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Vardiman JW, eds, WHO classification of tumours of haematopoietic and lymphoid tissues, WHO classification of tumours, vol 2, 4th edn. IARC Press, Lyon, pp 191–193 Kanellis G, Mollejo M, Montes-Moreno S, Rodriguez-Pinilla SM, Cigudosa JC, Algara P, Montalban C, Matutes E, Wotherspoon A, Piris MA, 2009, Splenic diffuse red pulp small B-cell lymphoma: revision of a series of cases reveals characteristic clinico-pathological features. Haematologica 95:1122–1129., DOI 10.3324/haematol.2009. 013714 Isaacson PG, Piris MA, Berger F, Swerdlow SH, Thieblemont C, Pittaluga S, Harris NL, 2008, Splenic B-cell marginal zone lymphoma. In: SwerdlowSH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Vardiman JW, eds, WHO classification of tumours of haematopoietic and lymphoid tissues, 4th edn,WHO classification of tumours, vol 2. IARC Press, Lyon, pp 185–187 Else M, Ruchlemer R, Osuji N, Del Giudice I,Matutes E,Woodman A, Wotherspoon A, Swansbury J, Dearden C, Catovsky D, 2005, Long remissions in hairy cell leukemia with purine analogs: a report of 219 patients with a median follow-up of 12.5 years. Cancer 104: 2442–2448 Tiacci E, Liso A, Piris M, Falini B, 2006, Evolving concepts in the pathogenesis of hairy-cell leukemia. Nat Rev Cancer 6:437–448., DOI 10.1038/nrc1888 Hounieu H, Chittal SM, al Saati T, deMascarel A, Sabattini E, Pileri S, Falini B, Ralfkiaer E, Le Tourneau A, Selves J et al, 1992, Hairy cell leukemia. Diagnosis of bone marrow involvement in paraffinembedded sections with monoclonal antibody DBA.44. Am J Clin Pathol 98:26–33 Janckila AJ, Cardwell EM, Yam LT, Li CY, 1995, Hairy cell identification by immunohistochemistry of tartrate-resistant acid phosphatase. Blood 85:2839–2844 Hoyer JD, Li CY, Yam LT, Hanson CA, Kurtin PJ, 1997, Immunohistochemical demonstration of acid phosphatase isoenzyme 5, tartrate-resistant, in paraffin sections of hairy cell leukemia and other hematologic disorders. Am J Clin Pathol 108:308–315 Went PT, Zimpfer A, Pehrs AC, Sabattini E, Pileri SA, Maurer R, Terracciano L, Tzankov A, Sauter G, Dirnhofer S, 2005, High specificity of combined TRAP and DBA.44 expression for hairy cell leukemia. Am J Surg Pathol 29:474–478 Bosch F, Jares P, Campo E, Lopez-Guillermo A, Piris MA, Villamor N, Tassies D, Jaffe ES, Montserrat E, Rozman C, Cardesa A, 1994, PRAD-1/cyclin D1 gene overexpression in chronic lymphoproliferative disorders: a highly specific marker of mantle cell lymphoma. Blood 84:2726–2732 Miranda RN, Briggs RC, Kinney MC, Veno PA, Hammer RD, Cousar JB, 2000, Immunohistochemical detection of cyclin D1 using optimized conditions is highly specific for mantle cell lymphoma and hairy cell leukemia. Mod Pathol 13:1308–1314 Sherman MJ, Hanson CA, Hoyer JD, 2011, An assessment of the usefulness of immunohistochemical stains in the diagnosis of hairy cell leukemia. Am J Clin Pathol 136:390–399., DOI 10.1309/ AJCP5GE1PSBMBZTW Johrens K, Happerfield LC, Brown JP, Erber WN, Stein H, Anagnostopoulos I, 2008, A novel CD11c monoclonal antibody effective in formalin-fixed tissue for the diagnosis of hairy cell leukemia. Pathobiology 75:252–256., DOI 10.1159/000132386 Johrens K, Stein H, Anagnostopoulos I, 2007, T-bet transcription factor detection facilitates the diagnosis of minimal hairy cell leukemia infiltrates in bone marrow trephines. Am J Surg Pathol 31:1181– 1185., DOI 10.1080/10428190903161042 Sadik W, Coupland S, Thachil J, 2010, Hairy cell leukaemia, negative for conventional cell markers, diagnosed using antibodies to annexin A1 and T-bet. Br J Haematol 151:207., DOI 10.1111/j.1365- 2141.2010.08359.x Falini B, Tiacci E, Liso A, Basso K, Sabattini E, Pacini R, Foa R, Pulsoni A, Dalla Favera R, Pileri S, 2004, Simple diagnostic assay for hairy cell leukaemia by immunocytochemical detection of annexin A1, ANXA1). Lancet 363:1869–1870 Andrulis M, Penzel R, Weichert W, von Deimling A, Capper D, 2012, Application of a BRAF V600E mutation-specific antibody for the diagnosis of hairy cell leukemia. Am J Surg Pathol 36:1796– 1800., DOI 10.1097/PAS.0b013e3182549b50 Tiacci E, Schiavoni G, Martelli MP, Boveri E, Pacini R, Tabarrini A, Zibellini S, Santi A, Pettirossi V, Fortini E, Ascani S, Arcaini L, Inghirami G, Paulli M, Falini B, 2013, Constant activation of the RAF-MEK-ERK pathway as a diagnostic and therapeutic target in hairy cell leukemia. Haematologica 98:635–639., DOI 10.3324/ haematol.2012.078071 Morgan EA, Yu H, Pinkus JL, Pinkus GS, 2013, Immunohistochemical detection of hairy cell leukemia in paraffin sections using a highly effective CD103 rabbit monoclonal antibody. Am J Clin Pathol 139:220–230. doi :10.1309/ AJCPHW7RULIZT2GB Krenacs L, Toth-Liptak J, Demeter J, Piukovics K, Borbenyi Z, Gogolak P, Sari E, Bagdi E, 2013, Monoclonal antibody HBME-1 reacts with a minor subset of B cells with villous surface and can be useful in the diagnosis of hairy cell leukemia and other indolent lymphoproliferations of villous B lymphocytes. Virchows Arch 463:787–794., DOI 10.1007/s00428-013-1490-52013 Frisch B, Bartl R, eds,, 1999, Biopsy interpretation of bone and bone marrow, 2nd edn. Arnold, London-Sidney-Auckland, p 350 Erber WN, Mason DY, 1988, Expression of the interleukin-2 receptor, Tac antigen/CD25, in hematologic neoplasms. Am J Clin Pathol 89:645–648 Micklem KJ, Dong Y, Willis A, Pulford KA, Visser L, Durkop H, Poppema S, Stein H, Mason DY, 1991, HML-1 antigen on mucosa-associated T cells, activated cells, and hairy leukemic cells is a new integrin containing the beta 7 subunit. Am J Pathol 139: 1297–1301 Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, Jones CM, Marshall CJ, Springer CJ, Barford D, Marais R, 2004, Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 116:855–867 Dorfman DM, Hwang ES, Shahsafaei A, Glimcher LH, 2004, T-bet, a T-cell-associated transcription factor, is expressed in a subset of Bcell lymphoproliferative disorders. J Clin Pathol 122:292–297., DOI 10.1309/AQQ2DVM75DVY0PWP Munoz L, Nomdedeu JF, Lopez O, Carnicer MJ, BellidoM, Aventin A, Brunet S, Sierra J, 2001, Interleukin-3 receptor alpha chain, CD123, is widely expressed in hematologic malignancies. Haematologica 86:1261–1269 Li WV, Kapadia SB, Sonmez-Alpan E, Swerdlow SH, 1996, Immunohistochemical characterization of mast cell disease in paraffin sections using tryptase, CD68, myeloperoxidase, lysozyme, and CD20 antibodies. Mod Pathol 9:982–988 Dorfman DM, Shahsafaei A, 2010, CD200, OX-2 membrane glycoprotein, expression in B cell-derived neoplasms. Am J Clin Pathol 134:726–733., DOI 10.1309/AJCP38XRRUGSQOVC Sainati L, Matutes E, Mulligan S, de Oliveira MP, Rani S, Lampert IA, Catovsky D, 1990, Avariant form of hairy cell leukemia resistant to alpha-interferon: clinical and phenotypic characteristics of 17 patients. Blood 76:157–162 Basso K, Liso A, Tiacci E, Benedetti R, Pulsoni A, Foa R, Di Raimondo F, Ambrosetti A, Califano A, Klein U, Dalla Favera R, Falini B, 2004, Gene expression profiling of hairy cell leukemia reveals a phenotype related to memory B cells with altered expression of chemokine and adhesion receptors. J Exp Med 199:59–68 Tiacci E, Schiavoni G, Forconi F, Santi A, Trentin L, Ambrosetti A, Cecchini D, Sozzi E, di Francia CP, Di Bello C, Pulsoni A, Foa R, Inghirami G, Falini B, 2012, Simple genetic diagnosis of hairy cell leukemia by sensitive detection of the BRAF-V600E mutation. Blood 119:192–195., DOI 10.1182/blood-2011-08-371179 Visser L, Shaw A, Slupsky J, Vos H, Poppema S, 1989)Monoclonal antibodies reactive with hairy cell leukemia. Blood 74:320–325 Schwarting R, Stein H,Wang CY, 1985, The monoclonal antibodies alpha S-HCL 1, alpha Leu-14, and alpha S-HCL 3, alpha Leu-M5, allow the diagnosis of hairy cell leukemia. Blood 65:974–983 Armitage RJ, Cawley JC, 1986, Normal and certain leukaemic B cells express IL-2 receptors without in vitro activation. Clin Exp Immunol 63:298–302 SheibaniK,Winberg CD, van deVelde S, BlayneyDW, Rappaport H, 1987, Distribution of lymphocytes with interleukin-2 receptors, TAC antigens, in reactive lymphoproliferative processes, Hodgkin’s disease, and non-Hodgkin’s lymphomas. An immunohistologic study of 300 cases. Am J Pathol 127:27–37 De Boer CJ, Kluin-Nelemans JC, Dreef E, Kester MGD, Kluin PM, Schuuring E, van Krieken JHJM, 1996, Involvement of the CCND1 gene in hairy cell Leukemia. Ann Oncol 7:251–256 Kreft A, Busche G, Bernhards J, Georgi i A, 1997, Immunophenotype of hairy-cell leukaemia after cold polymerization of methyl-methacrylate embeddings from 50 diagnostic bone marrow biopsies. Histopathology 30:145–151 Brunetti L, Di Noto R, Abate G, Gorrese M, Gravetti A, Raia M, Scalia G, Pascariello C, Camera A, Del Vecchio L, 2009, CD200/ OX2, a cell surface molecule with immuno-regulatory function, is consistently expressed on hairy cell leukemia neoplastic cells. Br J Haematol 145:665–667., DOI 10.1111/j.1365-2141.2009.07644.x Palumbo GA, Parrinello N, Fargione G, Cardillo K, Chiarenza A, Berretta S, Conticello C, Villari L, Di Raimondo F, 2009, CD200 expression may help in differential diagnosis between mantle cell lymphoma and B-cell chronic lymphocytic leukemia. Leuk Res 33: 1212–1216., DOI 10.1016/j.leukres.2009.01.017 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 203 EP 211 DI 10.1007/s12253-014-9807-5 PG 9 ER PT J AU Magyari, F Barna, S Miltenyi, Zs Rajnai, H Csomor, J Udvardy, M Illes, Varoczy, L AF Magyari, Ferenc Barna, Sandor Miltenyi, Zsofia Rajnai, Hajnalka Csomor, Judit Udvardy, Miklos Illes, Arpad Varoczy, Laszlo TI Histopathological Difficulties in an Adolescent Lymphoma Patient SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE B-cell lymphoma intermediate beetwen DLBCL and classical HL; 18FDG-PET/CT; R-CHOP; Agressive lymphomas ID B-cell lymphoma intermediate beetwen DLBCL and classical HL; 18FDG-PET/CT; R-CHOP; Agressive lymphomas AB The B-cell lymphoma, unclassifiable, showing intermediate features typical for both diffuse large B-cell lymphoma (DLBCL) and classical Hodgkin lymphoma (HL) is a novel category of diffuse large B-cell lymphomas (DLBCL/ HL), which has been described by the WHO classification in 2008. This rare type of lymphomas, previously called as gray zone lymphoma presents peculiar clinical, morphological and immunophenotypical patterns. In December 2011 a 17-year old male was diagnosed with mixed cellularity subtype of classical HL. His clinical stage was IV/BXS (abdominal bulky) with unfavourable prognosis. Because of the unusally extended disease (nodal-extranodal-bulky) a histological revision was performed. After an incomplete course of ABVD chemotherapy the patient’s symptoms disappeared and regression was detected in the size of peripheral lymph nodes. The diagnosis changed into DLBCL/HL, so the treatment was modified to R-CHOP-14 regimen. After the administration of 3 cycles of R-CHOP-14, he achieved a complete metabolic remission (CMR), which was confirmed by a 18FDG-PET/CT scan. Receiving further 4 cycles of R-CHOP-14 therapy the patient was still in CMR, but a PET negative large mass (70× 30 mm) still remained visible in the abdominal region. Considering this residuum and the agressive subtype of lymphoma he was referred for an autologous hemopoietic stem cell transplantation (AHSCT). After 2 cycles of R-DHAP regimen, successful CD34 positive stem cell collection was performed in August 2012. In September 2012, he underwent a R-BEAM conditioning followed by AHSCT. The next 18FDG-PET/CT still detected CMR 100 days after the AHSCT. The patient was in excellent clinical condition and also in complete remission 15 months after the AHSCT. Upon this case, it should be underlined that the diagnosis may need revision if a patient represents atypical clinical signs and behavior, and the importance of cooperation between clinicians and pathologists is also strongly emphasized. C1 [Magyari, Ferenc] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Barna, Sandor] Scanomed Kft.Debrecen, Hungary. [Miltenyi, Zsofia] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Rajnai, Hajnalka] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Csomor, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Udvardy, Miklos] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Varoczy, Laszlo] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. RP Magyari, F (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, H-4032 Debrecen, Hungary. EM magyarif13@gmail.com CR Swerdlow S, Campo E, Harris NL et al, 2008, WHO classification of tumors of hematopoietic and lymphoid tissues. IARC, Lyon, pp 267–268 Grant C, Dunleavy K, Eberle FC et al, 2011, Primary mediastinal large B-cell lymphoma, classic hodgkin lymphoma presenting in the mediastinum, and mediastinal gray zone lymphoma: what is the oncologist to do? Curr Hematol Malig Rep 6(3):157–163 Rosenwald A,Wright G, Leroy K et al, 2003)Molecular diagnosis of primarymediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma. J Exp Med 198:851–862 Hasserjian RP, Ott G, Elenitoba-Johnson KS et al, 2009, Commentary on the WHO classification of tumors of lymphoid tissues, 2008)“Gray zone” lymphomas overlapping with Burkitt lymphoma or classicalHodgkin lymphoma. J Hematopathol 2:89–95 Dunleavy K, Grant C, Eberle FC et al, 2012, Gray-zone lymphoma: better treated like hodgkin-lymphoma or mediastinal large B-cell lymphoma. Curr Hematol Malig Rep 7:241–247 Cabanillas F, 2011, Non-Hodgkin’s lymphoma: the old and the new. Clin Lymphoma Myeloma Leuk., DOI 10.1016/j.clml.2011.03.029, Epub 2011 Apr 28 Dunleavy K, Pittaluga S, Shovlin M et al, 2011, Untreated primary mediastinal B-cell, PMBL, and mediastinal grey zone, MGZL, lymphomas: comparison of biological features and clinical outcome following DA-EPOCH-R without radiation. Ann Oncol 22(suppl 4):134 Rieger M, Osterborg A, Pettengell R, White D, Gill D, Walewski J, Kuhnt E, Loeffler M, Pfreundschuh M, Ho D, A, and for the MabThera International Trial, MInT, Group, 2011, Primary mediastinal B-cell lymphoma treated with CHOP-like chemotherapy with or without rituximab: results of the Mabthera International Trial Group study. Ann Oncol, DOI 10.1093/annonc/mdq418 Zinzani PL, Martelli M, Poletti Vet al., 2008, Practice guidelines for the management of extranodal non- Hodgkin’s lymphomas of adult non-immunodeficient patients. Part I: primary lung and mediastinal lymphomas. A project of the Italian Society of Hematology, the Italian Society of Experimental Hematology and the Italian Group for Bone Marrow Transplantation. Haematologica., DOI 10.3324/ haematol.12742. Epub 2008 Jul 4 Cazals-Hatem D, Andre M, Munier N et al, 2001, Pathologic and clinical features of 77 Hodgkin’s lymphoma patients treated in a lymphoma protocol, LNH87, a GELA study. Am J Surg Pathol 25(3):297–306 Iwaki N, Sato Y, Kurakowa T et al, 2013, B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and classical Hodgkin lymphoma without mediastinal disease: mimicking nodular sclerosis classical Hodgkin lymphoma. Med Mol Morphol., DOI 10.1007/s00795-013-0038-8. Epub 2013 Mar 20 Vassilakopuolos PT, Pangalis AG, Katsigiannis A et al, 2012, Rituximab, cyclophosphamide, doxorubicin, vincristin, and prednisone with or without radiotherapy in primarymediastinal large B-cell lymphoma: the emerging standard of care. Oncologist., DOI 10.1634/ theoncologist Han HS, Escalo MP, Hsiao B et al, 2010, High incidence of falsepositive PET scans in patients with aggressive non Hodgkin lymphoma treated with rituximab containing regimen. Ann Oncol 20:309–318 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 213 EP 217 DI 10.1007/s12253-014-9810-x PG 5 ER PT J AU Sun, G Wang, Y Liu, Q Hu, W AF Sun, Guogui Wang, Yadi Liu, Q Hu, Wanning TI Expression of Wip1 in Kidney Carcinoma and its Correlation with Tumor Metastasis and Clinical Significance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Wip1; Kidney cancer; Cell proliferation; Cell apoptosis; Prognosis ID Wip1; Kidney cancer; Cell proliferation; Cell apoptosis; Prognosis AB This study aimed to analyze the expression, clinical significance of proto-oncogene in kidney carcinoma and the biological effect in its cell line by siRNA targeting wild-type p53-induced phosphatase 1 (Wip1). Immunohistochemistry and western blot were respectively used to analyze Wip1 protein expression in 78 cases of kidney cancer and normal tissues to study the relationship between Wip1 expression and clinical factors. Wip1 siRNA was transiently transfected into papillary kidney carcinoma cell by liposome-mediated method and was detected by Quantitative real-time RT-PCR (qRTPCR) and western blot.MTT assay, cell apoptosis, cell migration and invasion were also conducted as to the influence of the down-regulated expression of Wip1 that might be found on ACHN cells biological effect. The level of Wip1 protein expression was found to be significantly higher in kidney cancer tissue than normal tissues (P<0.05). There were significant differences between Wip1 expression and lymph node metastasis, clinical stages and tumor differentiation (P<0.05). Meanwhile, Increased expression of Wip1 was significantly with poor overall survival time by Kaplan-Meier analysis (P<0.05). qRT-PCR and Western blot showed that ACHN cell transfected Wip1 siRNA had a lower relative expressive content than normal cell (P<0.05).MTT assay, cell apoptosis, cell cycles demonstrated that ACHN cell transfected Wip1 siRNA had a lower survival fraction, higher cell apoptosis, more percentage of the G0/G1 phases, significant decrease in migration and invasion, and higher P53 and P16 protein expression compared with ACHN cell untransfected Wip1 siRNA (P<0.05). Wip1 protein was increased in kidney carcinoma, specifically in T stages, lymph node metastasis, clinical stages and tumor differentiation. Wip1 may involved in the biological processes of kidney cancer cell proliferation, apoptosis, and migration and invasion by regulation P53 and P16 protein expression. C1 [Sun, Guogui] Tangshan People’s Hospital, Department of Chemoradiotherapy, NO.65, Shengli Road, Lunan District, 063000 Tangshan, Hebei Province, China. [Wang, Yadi] The Military General Hospital of Beijing PLA, Department of Radiotherapy, 100700 Beijing, China. [Liu, Q] The Fourth Hospital of Hebei Medical University, Department of Radiotherapy, 050017 Shijiazhuang, China. [Hu, Wanning] Tangshan People’s Hospital, Department of Chemoradiotherapy, NO.65, Shengli Road, Lunan District, 063000 Tangshan, Hebei Province, China. RP Hu, W (reprint author), Tangshan People’s Hospital, Department of Chemoradiotherapy, 063000 Tangshan, China. EM guogui_sun@hotmail.com CR Tan X, He S, Han Y, Yu Y, Xiao J, Xu D, Wang G, Du Y, Chang W, Yin J, Su T, Hou J, Cao G, 2013, Establishment and characterization of clear cell renal cell carcinoma cell lines with different metastatic potential from Chinese patients. Cancer Cell Int 13:20 StadlerWM, Figlin RA,McDermott DF, Dutcher JP, Knox JJ,Miller WH Jr, Hainsworth JD, Henderson CA, George JR, Hajdenberg J, Kindwall-Keller TL, Ernstoff MS, Drabkin HA, Curti BD, Chu L, Ryan CW, Hotte SJ, Xia C, Cupit L, Bukowski RM, 2010, ARCCS Study Investigators: safety and efficacy results of the advanced renal cell carcinoma sorafenib expanded access programinNorthAmerica. Cancer 116:1272–1280 Fuku T, Semba S, Yutori H, Yokozaki H, 2007, Increased wild-type p53-induced phosphatase 1, Wip1 or PPM1D, expression correlated with downregulation of checkpoint kinase 2 in human gastric carcinoma. Pathol Int 57:566–571 Li J, Yang Y, Peng Y, Austin RJ, van Eyndhoven WG, Nguyen KC, Gabriele T,McCurrachME,Marks JR, Hoey T, Lowe SW, Powers S, 2002, Oncogenic properties of PPM1D located within a breast cancer amplification epicenter at 17q23. Nat Genet 31:133–134 Saito-Ohara F, Imoto I, Inoue J, Inoue J, Hosoi H, Nakagawara A, Sugimoto T, Inazawa J, 2003, PPM1D is a potential target for 17q gain in neuroblastoma. Cancer Res 63:1876–1883 Mendrzyk F, Radlwimmer B, Joos S, Kokocinski F, Benner A, Stange DE, Neben K, Fiegler H, Carter NP, Reifenberger G, Korshunov A, Lichter P, 2005, Enomic and protein expression profiling identifies CDK6 as novel in dependent prognostic marker in medulloblastoma. J Clin Oncol 23:8853–8862 Yang DH, He JA, Li J, Ma WF, Hu XH, Xin SJ, Duan ZQ, 2010, Expression of proto-oncogene Wip1 in breast cancer and its clinical significance. Natl Med J Chin 90:519–522 Liang CH, Jiao BH, Lu SK, Guo EK, Zhang GY, 2011, The expression of proto-oncogeneWip1 in human glioblastoma multiforme and cell lines. Chin J Neuro-Oncol 9:1–6 Liang CH, Jiao BH, Guo EK, Lu SK, 2011, Over-expression of proto-oncogene Wip1 in intracranial ependymomas association with P53. Basic Clin Med 31:430–434 Yang DH, Zhang H, Hu XH, Xin SJ, Duan ZQ, 2011, Abnormality of p16 / p38MAPK / p53 /Wip1 pathway in papillary thyroid cancer and its significance. Chin J Gen Surg 20:1199–1202 Oliva TM, Berthonaud V, Chevalier A, Ducrot C, Marsolier-Kergoat MC, Mann C, Leteurtre F, 2007, The Wip1 phosphatase, PPM1D, antagonizes activation of the Chk2 tumour suppressor kinase. Oncogene 26:1449–1458 Fall B, Diao B, Sow Y, Sarr A, Thiam A, Fall PA, Ndoye AK, Sylla C, Ba M, Mendes V, Diagne BA, 2011, Adult renal cancer in Senegal: current epidemiological, clinical features, profile’s evolution over the two past decades. Prog Urol 21:521–526 Frommhold J, Jocham D, Doehn C, 2011, How accurate is the correlation between clinical and pathological TNM stages in renal tumours? Aktuelle Urol 42:247–251 Holley JL, 2011, The importance of prognosis in cancer screening in patients with chronic kidney disease. Semin Dial 24:16–17 Baxter EW, Milner J, 2010, p53 Regulates LIF expression in human medulloblastoma cells. J Neuro Oncol 97:373–382 Lowe JM, Cha H, Yang Q, Fornace AJ Jr, 2010, Nuclear factorkappaB, NF-kappaB, is a novel positive transcriptional regulator of the oncogenic Wip1 phosphatase. J Biol Chem 285:5249–5257 Fu L, ChenW, GuoW,Wang J, Tian Y, Shi D, Zhang X, Qiu H, Xiao X, Kang T, HuangW,Wang S, DengW(2013, Berberine targets AP- 2/hTERT, NF-κB/COX-2, HIF-1α/VEGF and cytochrome-c/caspase signaling to suppress human cancer cell growth. PLoS One 8:e69240 Armstrong NJ, Fagotto F, Pthmann C, Rupp RA, 2012, Maternal Wnt /β-catenin signaling coactivates transcription through NF-κB binding sites during Xenopus axis ormation. PLoS One 7:e36136 Pan H, ZhouW, HeW, LiuX,DingQ, Ling L, Zha X,Wang S, 2012, Genistein inhibits MDA-MB-231 triple- negative breast cancer cell growth by inhibiting NF-kappaB activity via the Notch-1 pathway. Int J Mol Med 30:337–343 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 219 EP 224 DI 10.1007/s12253-014-9811-9 PG 6 ER PT J AU Je, ME Choi, JY Yoo, JN Lee, HS AF Je, Mi Eun Choi, Jin Youn Yoo, Jin Nam Lee, Hyung Sug TI Oncogenic PTPN11 Mutations are Rare in Solid Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Je, Mi Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Choi, Jin Youn] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Pugh TJ,Morozova O, Attiyeh EF, Asgharzadeh S,Wei JS, Auclair D, Carter SL, Cibulskis K, Hanna M, Kiezun A, Kim J, Lawrence MS, Lichenstein L, McKenna A, Pedamallu CS, Ramos AH, Shefler E, Sivachenko A, Sougnez C, Stewart C, Ally A, Birol I, Chiu R, Corbett RD, Hirst M, Jackman SD, Kamoh B, Khodabakshi AH, Krzywinski M, Lo A, Moore RA, Mungall KL, Qian J, Tam A, Thiessen N, Zhao Y, Cole KA, Diamond M, Diskin SJ, Mosse YP, Wood AC, Ji L, Sposto R, Badgett T, LondonWB,Moyer Y, Gastier-Foster JM, Smith MA, Guidry Auvil JM, Gerhard DS, Hogarty MD, Jones SJ, Lander ES, Gabriel SB, Getz G, Seeger RC, Khan J,MarraMA, Meyerson M, Maris JM, 2013, The genetic landscape of high-risk neuroblastoma. Nat Genet 45:279–84 Chan G, Kalaitzidis D, Neel BG, 2008, The tyrosine phosphatase Shp2, PTPN11, in cancer. Cancer Metastasis Rev 27:179–92 Tartaglia M, Niemeyer CM, Fragale A, Song X, Buechner J, Jung A, Hahlen K, Hasle H, Licht JD, Gelb BD, 2003, Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia. Nat Genet 34:148–50 YooNJ, KimHR, KimYR, An CH, Lee SH, 2012, Somaticmutations of the KEAP1 gene in common solid cancers. Histopathology 60:943– 52 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–7 Bentires-Alj M, Paez JG, David FS, Keilhack H, Halmos B, Naoki K, Maris JM, Richardson A, Bardelli A, Sugarbaker DJ, Richards WG, Du J, Girard L, Minna JD, Loh ML, Fisher DE, Velculescu VE, Vogelstein B, Meyerson M, Sellers WR, Neel BG, 2004, Activating mutations of the noonan syndrome-associated SHP2/PTPN11 gene in human solid tumors and adult acute myelogenous leukemia. Cancer Res 64:8816–20 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2015 VL 21 IS 1 BP 225 EP 227 DI 10.1007/s12253-014-9780-z PG 3 ER PT J AU Bianco, MA Uno, M Oba-Shinjo, MS Clara, AC de Almeida Galatro, FTh Rosemberg, S Teixeira, JM Nagahashi Marie, KS AF Bianco, Macedo Andre Uno, Miyuki Oba-Shinjo, Mieko Sueli Clara, Afonso Carlos de Almeida Galatro, Fernanda Thais Rosemberg, Sergio Teixeira, Jacobsen Manoel Nagahashi Marie, Kazue Suely TI CXCR7 and CXCR4 Expressions in Infiltrative Astrocytomas and Their Interactions with HIF1α Expression and IDH1 Mutation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chemokine; Astrocytoma; CXCR7; CXCR4; HIF1α; IDH1 ID Chemokine; Astrocytoma; CXCR7; CXCR4; HIF1α; IDH1 AB The CXCR7, a new receptor for CXCL12 with higher affinity than CXCR4 has raised key issues on glioma cell migration. The aim of this study is to investigate the CXCR7 mRNA expression in diffuse astrocytomas tissues and to evaluate its interactions with CXCR4 and HIF1α expression and IDH1 mutation. CXCR7, CXCR4 and HIF1α mRNA expression were evaluated in 129 frozen samples of astrocytomas. IDH1 mutation status was analyzed with gene expressions, matched with clinicopathological parameters and overall survival time. Protein expression was analyzed by immunohistochemistry in different grades of astrocytoma and in glioma cell line (U87MG) by confocal microscopy. There was significant difference in the expression levels of the genes studied between astrocytomas and non-neoplasic (NN) controls (p<0.001). AGII showed no significant correlation between CXCR7/HIF1α (p=0.548); there was significant correlation between CXCR7/CXCR4 (p=0.042) and CXCR7/IDH1 (p=0.008). GBM showed significant correlations between CXCR7/CXCR4 (p = 0.002), CXCR7/IDH1 (p<0.001) and CXCR7/HIF1α (p=0.008). HIF1α overexpression was associated with higher expressions of CXCR7 (p=0.01) and CXCR4 (p<0.0001), while IDH1 mutation was associated with lower CXCR7 (p=0.009) and CXCR4 (p=0.0005) mRNA expressions. Protein expression increased with malignancy and in U87MG cell line was mainly localized in the cellular membrane. CXCR7 was overexpressed in astrocytoma and correlates with CXCR4 and IDH1 in AGII and CXCR4, IDH1 and HIF1α in GBM. Overexpression HIF1α was related with higher expressions of CXCR7 and CXCR4, otherwise IDH1 mutation related with lower expression of both genes. No association between CXCR7 and CXCR4 expression and survival data was related. C1 [Bianco, Macedo Andre] University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell Biology, LIM15 Av. Dr. Arnaldo, 455, 4th floor, r.4110, 01246-903 Sao Paulo, SP, Brazil. [Uno, Miyuki] University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell Biology, LIM15 Av. Dr. Arnaldo, 455, 4th floor, r.4110, 01246-903 Sao Paulo, SP, Brazil. [Oba-Shinjo, Mieko Sueli] University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell Biology, LIM15 Av. Dr. Arnaldo, 455, 4th floor, r.4110, 01246-903 Sao Paulo, SP, Brazil. [Clara, Afonso Carlos] Barretos Cancer Hospital, Rua Antenos Duarte Villela, 1331, 14784-400 Barretos, SP, Brazil. [de Almeida Galatro, Fernanda Thais] University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell Biology, LIM15 Av. Dr. Arnaldo, 455, 4th floor, r.4110, 01246-903 Sao Paulo, SP, Brazil. [Rosemberg, Sergio] Sao Paulo University School of Medicine, Department of Pathology, Av. Dr. Arnaldo, 455, 01246-903 Sao Paulo, SP, Brazil. [Teixeira, Jacobsen Manoel] University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell Biology, LIM15 Av. Dr. Arnaldo, 455, 4th floor, r.4110, 01246-903 Sao Paulo, SP, Brazil. [Nagahashi Marie, Kazue Suely] University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell Biology, LIM15 Av. Dr. Arnaldo, 455, 4th floor, r.4110, 01246-903 Sao Paulo, SP, Brazil. RP Bianco, MA (reprint author), University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell Biology, 01246-903 Sao Paulo, Brazil. EM abianco@usp.br;amb0973@gmail.com CR Dolecek TA, Propp JM, Stroup NE, Kruchko C, 2012, CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005–2009. Neuro Oncol 14(Suppl 5):v1–v49., DOI 10.1093/neuonc/nos218 Kleihues P, Louis DN, Scheithauer BW et al, 2002, The WHO classification of tumors of the nervous system. J Neuropathol Exp Neurol 61:215–225, discussion 226–229 Janny P, Cure H, Mohr M et al, 1994, Low grade supratentorial astrocytomas. Manag Prognostic Factors Cancer 73:1937–1945 Keles GE, Lamborn KR, Berger MS, 2001, Low-grade hemispheric gliomas in adults: a critical review of extent of resection as a factor influencing outcome. J Neurosurg 95:735–745., DOI 10.3171/jns. 2001.95.5.0735 Leighton C, Fisher B, Bauman G et al, 1997, Supratentorial lowgrade glioma in adults: an analysis of prognostic factors and timing of radiation. J Clin Oncol 15:1294–1301 Schomas DA, Laack NNI, Rao RD et al, 2009, Intracranial lowgrade gliomas in adults: 30-year experience with long-termfollow-up at Mayo clinic. Neuro Oncol 11:437–445., DOI 10.1215/15228517- 2008-102 Chang EF, Smith JS, Chang SM et al, 2008, Preoperative prognostic classification system for hemispheric low-grade gliomas in adults. J Neurosurg 109:817–824., DOI 10.3171/JNS/2008/109/11/0817 Van den Bent MJ, Afra D, deWitte O et al, 2005, Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: the EORTC 22845 randomised trial. Lancet 366:985–990., DOI 10.1016/S0140-6736(05)67070-5 Stupp R,Mason WP, van den BentMJ et al, 2005, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352:987–996., DOI 10.1056/NEJMoa043330 Balkwill F, 2004, Cancer and the chemokine network. Nat Rev Cancer 4:540–550., DOI 10.1038/nrc1388 Gerber PA, Hippe A, Buhren BA et al, 2009, Chemokines in tumorassociated angiogenesis. Biol Chem 390:1213–1223., DOI 10.1515/ BC.2009.144 Kakinuma T, Hwang ST, 2006, Chemokines, chemokine receptors, and cancer metastasis. J Leukoc Biol 79:639–651., DOI 10.1189/jlb. 1105633 Calatozzolo C, Canazza A, Pollo B et al, 2011, Expression of the new CXCL12 receptor, CXCR7, in gliomas. Cancer Biol Ther 11:242– 253 D’Alterio C, Consales C, Polimeno M et al, 2010, Concomitant CXCR4 and CXCR7 expression predicts poor prognosis in renal cancer. Curr Cancer Drug Targets 10:772–781 Gebauer F, TachezyM, Effenberger K et al, 2011, Prognostic impact of CXCR4 and CXCR7 expression in pancreatic adenocarcinoma. J Surg Oncol 104:140–145., DOI 10.1002/jso.21957 Hattermann K, Held-Feindt J, Lucius R et al, 2010, The chemokine receptor CXCR7 is highly expressed in human glioma cells and mediates antiapoptotic effects. Cancer Res 70:3299–3308., DOI 10. 1158/0008-5472.CAN-09-3642 Balabanian K, Lagane B, Infantino S et al, 2005, The chemokine SDF-1/CXCL12 binds to and signals through the orphan receptor RDC1 in T lymphocytes. J Biol Chem 280:35760–35766., DOI 10. 1074/jbc.M508234200 Burns JM, Summers BC, Wang Y et al, 2006, A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. J Exp Med 203:2201–2213., DOI 10. 1084/jem.20052144 Arvidsson Y, Bergstrom A, Arvidsson L et al, 2010, Hypoxia stimulates CXCR4 signalling in ileal carcinoids. Endocr Relat Cancer 17: 303–316., DOI 10.1677/ERC-09-0085 Liu H, Xue W, Ge G et al, 2010, Hypoxic preconditioning advances CXCR4 and CXCR7 expression by activating HIF-1α in MSCs. Biochem Biophys Res Commun 401:509–515., DOI 10.1016/j.bbrc. 2010.09.076 Liu Y-L, Yu J-M, Song X-R et al, 2006, Regulation of the chemokine receptor CXCR4 and metastasis by hypoxiainducible factor in non small cell lung cancer cell lines. Cancer Biol Ther 5:1320–1326 Sun X, Wei L, Chen Q, Terek RM, 2010, CXCR4/SDF1 mediate hypoxia induced chondrosarcoma cell invasion through ERK signaling and increased MMP1 expression. Mol Cancer 9:17., DOI 10.1186/ 1476-4598-9-17 Schioppa T, Uranchimeg B, Saccani A et al, 2003, Regulation of the chemokine receptor CXCR4 by hypoxia. J Exp Med 198:1391–1402., DOI 10.1084/jem.20030267 Parsons DW, Jones S, Zhang X et al, 2008, An integrated genomic analysis of human glioblastoma multiforme. Science 321:1807– 1812., DOI 10.1126/science.1164382 Dang L, White DW, Gross S et al, 2009, Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 462:739–744., DOI 10. 1038/nature08617 Gottle P, Kremer D, Jander S et al, 2010, Activation of CXCR7 receptor promotes oligodendroglial cell maturation. Ann Neurol 68: 915–924., DOI 10.1002/ana.22214 Valente V, Teixeira SA, Neder L et al, 2009, Selection of suitable housekeeping genes for expression analysis in glioblastoma using quantitative RT-PCR. BMC Mol Biol 10:17., DOI 10.1186/1471- 2199-10-17 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), Method. Methods 25:402–408., DOI 10.1006/meth.2001.1262 UnoM, Oba-Shinjo SM, da Silva R et al, 2011, IDH1 mutations in a Brazilian series of Glioblastoma. Clinics, Sao Paulo, 66:163–165 Hermann PC,Huber SL,Herrler Tet al, 2007, Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1:313–323., DOI 10.1016/j. stem.2007.06.002 Ehtesham M, Mapara KY, Stevenson CB, Thompson RC, 2009, CXCR4 mediates the proliferation of glioblastoma progenitor cells. Cancer Lett 274:305–312., DOI 10.1016/j.canlet.2008.09.034 Salmaggi A, Maderna E, Calatozzolo C et al, 2009, CXCL12, CXCR4 and CXCR7 expression in brain metastases. Cancer Biol Ther 8:1608–1614 Hartmann TN, Burger M, Burger JA, 2004, The role of adhesion molecules and chemokine receptor CXCR4, CD184, in small cell lung cancer. J Biol Regul Homeost Agents 18:126–130 Wang J, Shiozawa Y,Wang J et al, 2008, The role of CXCR7/RDC1 as a chemokine receptor for CXCL12/SDF-1 in prostate cancer. J Biol Chem 283:4283–4294., DOI 10.1074/jbc.M707465200 Miao Z, Luker KE, Summers BC et al, 2007, CXCR7, RDC1, promotes breast and lung tumor growth in vivo and is expressed on tumor-associated vasculature. Proc Natl Acad Sci U S A 104:15735– 15740., DOI 10.1073/pnas.0610444104 Iwakiri S, Mino N, Takahashi T et al, 2009, Higher expression of chemokine receptor CXCR7 is linked to early and metastatic recurrence in pathological stage I nonsmall cell lung cancer. Cancer 115: 2580–2593., DOI 10.1002/cncr.24281 Zheng K, Li H-Y, Su X-L et al, 2010, Chemokine receptor CXCR7 regulates the invasion, angiogenesis and tumor growth of human hepatocellular carcinoma cells. J Exp Clin Cancer Res 29:31., DOI 10.1186/1756-9966-29-31 Liang JJ, Zhu S, Bruggeman R et al, 2010, High levels of expression of human stromal cell-derived factor-1 are associated with worse prognosis in patients with stage II pancreatic ductal adenocarcinoma. Cancer Epidemiol Biomarkers Prev 19:2598–2604., DOI 10.1158/ 1055-9965.EPI-10-0405 Wang L, Chen W, Gao L et al, 2012, High expression of CXCR4, CXCR7 and SDF-1 predicts poor survival in renal cell carcinoma. World J Surg Oncol 10:212., DOI 10.1186/1477-7819-10-212 Hao M, Zheng J, Hou K et al, 2012, Role of chemokine receptor CXCR7 in bladder cancer progression. Biochem Pharmacol 84:204– 214., DOI 10.1016/j.bcp.2012.04.007 Maishi N, Ohga N, Hida Y et al, 2012, CXCR7: a novel tumor endothelial marker in renal cell carcinoma. Pathol Int 62:309–317., DOI 10.1111/j.1440-1827.2012.02792.x SchrevelM, Karim R, ter Haar NTet al, 2012, CXCR7 expression is associated with disease-free and disease-specific survival in cervical cancer patients. Br J Cancer 106:1520–1525., DOI 10.1038/bjc.2012. 110 Takano S, Yamashita T, Ohneda O, 2010, Molecular therapeutic targets for glioma angiogenesis. J Oncol 2010:351908., DOI 10.1155/ 2010/351908 Wang Y-N, Hung M-C, 2012, Nuclear functions and subcellular trafficking mechanisms of the epidermal growth factor receptor family. Cell Biosci 2:13., DOI 10.1186/2045-3701-2-13 Brand TM, Iida M, Li C, Wheeler DL, 2011, The nuclear epidermal growth factor receptor signaling network and its role in cancer. Discov Med 12:419–432 Wang S-C, Hung M-C, 2009, Nuclear translocation of the epidermal growth factor receptor family membrane tyrosine kinase receptors. Clin Cancer Res 15:6484–6489., DOI 10.1158/1078-0432.CCR-08- 2813 Rong Y, Durden DL, VanMeir EG, Brat DJ, 2006, “Pseudopalisading” necrosis in glioblastoma: a familiar morphologic feature that links vascular pathology, hypoxia, and angiogenesis. J Neuropathol Exp Neurol 65:529–539 Kioi M, Vogel H, Schultz G et al, 2010, Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J Clin Invest 120:694–705., DOI 10.1172/JCI40283 Ceradini DJ, Kulkarni AR, CallaghanMJ et al, 2004, Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 10:858–864., DOI 10.1038/nm1075 Pugh CW, Ratcliffe PJ, 2003, Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med 9:677–684., DOI 10.1038/ nm0603-677 Zagzag D, Lukyanov Y, Lan L et al, 2006, Hypoxia-inducible factor 1 and VEGF upregulate CXCR4 in glioblastoma: implications for angiogenesis and glioma cell invasion. Lab Invest 86:1221–1232., DOI 10.1038/labinvest.3700482 Schutyser E, Su Y, Yu Y et al, 2007, Hypoxia enhances CXCR4 expression in human microvascular endothelial cells and human melanoma cells. Eur Cytokine Netw 18:59–70., DOI 10.1684/ecn. 2007.0087 Mendez O, Zavadil J, EsencayMet al, 2010, Knock down of HIF-1? in glioma cells reduces migration in vitro and invasion in vivo and impairs their ability to form tumor spheres. Mol Cancer 9:133., DOI 10.1186/1476-4598-9-133 Metellus P, Colin C, Taieb D et al, 2011, IDH mutation status impact on in vivo hypoxia biomarkers expression: new insights from a clinical, nuclear imaging and immunohistochemical study in 33 glioma patients. J Neurooncol 105:591–600., DOI 10.1007/s11060- 011-0625-2 Williams SC, Karajannis MA, Chiriboga L et al, 2011, R132Hmutation of isocitrate dehydrogenase-1 is not sufficient for HIF-1α upregulation in adult glioma. Acta Neuropathol 121:279–281., DOI 10.1007/s00401-010-0790-y Noushmehr H,Weisenberger DJ,Diefes Ket al, 2010, Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17:510–522., DOI 10.1016/j.ccr.2010.03.017 Turcan S, Rohle D, Goenka A et al, 2012, IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483: 479–483., DOI 10.1038/nature10866 HegiME, Diserens A-C, Gorlia Tet al, 2005)MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 352: 997–1003., DOI 10.1056/NEJMoa043331 Leu S, von Felten S, Frank S et al, 2013, IDH/MGMT-driven molecular classification of low-grade glioma is a strong predictor for long-term survival. Neuro Oncol 15:469–479., DOI 10.1093/ neuonc/nos317 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 229 EP 240 DI 10.1007/s12253-014-9813-7 PG 12 ER PT J AU Ascencio-Cedillo, R Lopez-Pulido, IE Munoz-Valle, FJ Villegas-Sepulveda, N Del Toro-Arreola, S Estrada-Chavez, C Daneri-Navarro, A Franco-Topete, R Perez-Montiel, D Garcia-Carranca, A Pereira-Suarez, LA AF Ascencio-Cedillo, Rafael Lopez-Pulido, Ivan Edgar Munoz-Valle, Francisco Jose Villegas-Sepulveda, Nicolas Del Toro-Arreola, Susana Estrada-Chavez, Ciro Daneri-Navarro, Adrian Franco-Topete, Ramon Perez-Montiel, Delia Garcia-Carranca, Alejandro Pereira-Suarez, Laura Ana TI Prolactin and Prolactin Receptor Expression in Cervical Intraepithelial Neoplasia and Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; Prolactin; Prolactin receptor; Expression ID Cervical cancer; Prolactin; Prolactin receptor; Expression AB Prolactin receptor (PRLR) overexpression could play a role in tumorigenesis. The aim of this study was to determine prolactin (PRL) and PRLR expression in biopsies from patients with precursor lesions and uterine cervical cancer. PRLR expression was analyzed in 63 paraffin-embedded biopsies of uterine cervical tissue. In total, eleven low-grade squamous intraepithelial lesions (LSIL), 23 high-grade squamous intraepithelial lesions (HSIL), 21 uterine cervical cancers (UCC) and 8 normal epithelium (NE) were examined using immunoperoxidase staining and Western blot analysis. Additionally, PRL expression was identified in human cervical cancer serumand tissues. The PRLR expressionwas found to be significantly increased in cervical cancer in comparison with normal tissue and precursor lesions (P<0.0003). The presence of the long isoform of the PRLR was observed only in cervical cancer tissues. Serum PRL levels were normal in all samples and local prolactin expression was similar in precursor lesions and cervical cancer by Western blot analysis. Our data suggest a possible role for PRLR in the progression of cervical cancer. C1 [Ascencio-Cedillo, Rafael] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Departamento de FisiologiaGuadalajara, Jalisco, Mexico. [Lopez-Pulido, Ivan Edgar] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Departamento de FisiologiaGuadalajara, Jalisco, Mexico. [Munoz-Valle, Francisco Jose] Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, Instituto de Investigacion en Ciencias BiomedicasGuadalajara, Jalisco, Mexico. [Villegas-Sepulveda, Nicolas] Unidad Zacatenco, Centro de Investigacion y de Estudios Avanzados-IPN, Departamento de Biomedicina Molecular, 07360 Mexico City, DF, Mexico. [Del Toro-Arreola, Susana] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Departamento de FisiologiaGuadalajara, Jalisco, Mexico. [Estrada-Chavez, Ciro] Centro de Investigacion y Asistencia en Tecnologia y diseno del Estado de Jalisco A.C., 44270 Guadalajara, Jalisco, Mexico. [Daneri-Navarro, Adrian] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Departamento de FisiologiaGuadalajara, Jalisco, Mexico. [Franco-Topete, Ramon] Nuevo Hospital Civil de Guadalajara, Departamento de PatologiaGuadalajara, Jalisco, Mexico. [Perez-Montiel, Delia] Instituto de Investigaciones Biomedicas UNAM, Departamento de Biologia Molecular y BiotecnologiaMexico City, Mexico. [Garcia-Carranca, Alejandro] Instituto de Investigaciones Biomedicas UNAM, Departamento de Biologia Molecular y BiotecnologiaMexico City, Mexico. [Pereira-Suarez, Laura Ana] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Departamento de FisiologiaGuadalajara, Jalisco, Mexico. CR zur Hausen H. Papillomavirus infections–a major cause of human cancers. Biochimica et biophysica acta. 1996;1288, 2):F55-78. Epub 1996/10/09. NevilleMC, McFadden TB, Forsyth I, 2002, Hormonal regulation of mammary differentiation and milk secretion. J Mammary Gland Biol Neoplasia 7(1):49–66, Epub 2002/08/06 Rui H, Kirken RA, Farrar WL, 1994, Activation of receptorassociated tyrosine kinase JAK2 by prolactin. J Biol Chem 269(7): 5364–8, Epub 1994/02/18 Al-Sakkaf KA, Dobson PR, Brown BL, 1997, Prolactin induced tyrosine phosphorylation of p59fyn may mediate phosphatidylinositol 3-kinase activation in Nb2 cells. J Mol Endocrinol 19(3):347–50, Epub 1998/02/14 Das R, Vonderhaar BK, 1996, Activation of raf-1, MEK, and MAP kinase in prolactin responsive mammary cells. Breast Cancer Res Treat 40(2):141–9, Epub 1996/01/01 Nohara A, OhmichiM, Koike K, Jikihara H, Kimura A, Masuhara K et al, 1997, Prolactin stimulates mitogen-activated protein kinase in human leiomyoma cells. Biochem Biophys Res Commun 238(2): 473–7, Epub 1997/09/23 Ben-Jonathan N, Lapensee CR, Lapensee EW. What Can We Learn from Rodents about Prolactin in Humans? Endocr Rev. 2007. McHale K, Tomaszewski JE, Puthiyaveettil R, Livolsi VA, Clevenger CV, 2008, Altered expression of prolactin receptorassociated signaling proteins in human breast carcinoma. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 21(5):565–71, Epub 2008/02/05 Leav I, Merk FB, Lee KF, Loda M, Mandoki M, McNeal JE et al, 1999, Prolactin receptor expression in the developing human prostate and in hyperplastic, dysplastic, and neoplastic lesions. Am J Pathol 154(3):863–70, Epub 1999/03/18 Levina VV, Nolen B, Su Y, Godwin AK, Fishman D, Liu J et al, 2009, Biological significance of prolactin in gynecologic cancers. Cancer Res 69(12):5226–33, Epub 2009/06/06 Gonzalez-Lucano LR, Munoz-Valle JF, Ascencio-Cedillo R, Dominguez-Rosales JA, Lopez-Rincon G, Del Toro-Arreola S et al, 2012, Increased expression of the prolactin receptor is associated with malignant laryngeal tumors. Experimental and therapeutic medicine 3(4):603–7, Epub 2012/09/13 Macfee MS, McQueen J, Strayer DE, 1987, Immunocytochemical localization of prolactin in carcinoma of the cervix. Gynecol Oncol 26(3):314–8, Epub 1987/03/01 Perumal BG, Moodley J, Pegoraro RJ, 1998, Prolactin as a tumour marker in cancer of the cervix. J Obstet Gynaecol 18(3):260–2, Epub 2004/10/30 Harbaum L, Pollheimer MJ, Bauernhofer T,Kornprat P, Lindtner RA, Schlemmer A et al, 2010, Clinicopathological significance of prolactin receptor expression in colorectal carcinoma and corresponding metastases. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 23(7):961–71, Epub 2010/05/11 Strukov EL, Safronnikova NR, Bobrov Iu F, Gamaiunova VB, Bokhman Ia V, Dil'man VM. [Hyperprolactinemia as a marker of the progression of cervical cancer]. Voprosy onkologii. 1990;36, 7): 831–5. Epub 1990/01/01. Giperprolaktinemiia kak marker progressii raka sheiki matki. Hsu CT, Yu MH, Lee CY, Jong HL, Yeh MY, 1992, Ectopic production of prolactin in uterine cervical carcinoma. Gynecol Oncol 44(2):166–71, Epub 1992/02/01 Shoupe D, Montz FJ, Kletzky OA, DiZerega GS, 1983, Prolactin molecular heterogeneity. Response to thyrotropin-releasing hormone stimulation of concanavalin A-bound and -unbound immunoassayable prolactin during human pregnancy. Am J Obstet Gynecol 147(5):482–7, Epub 1983/11/01 Pellegrini I, Lebrun JJ, Ali S, Kelly PA, 1992, Expression of prolactin and its receptor in human lymphoid cells. Mol Endocrinol 6(7): 1023–31, Epub 1992/07/01 Champier J, Claustrat B, Sassolas G, BergerM(1987, Detection and enzymatic deglycosylation of a glycosylated variant of prolactin in human plasma. FEBS Lett 212(2):220–4, Epub 1987/02/23 Sabharwal P, Glaser R, Lafuse W, Varma S, Liu Q, Arkins S et al, 1992, Prolactin synthesized and secreted by human peripheral blood mononuclear cells: an autocrine growth factor for lymphoproliferation. Proc Natl Acad Sci U S A 89(16):7713–6, Epub 1992/08/15 Paraiba DB, Soares CR, Bartolini P, Arthuso FS, Borba EF, Bonfa E et al, 2010, Lymphocytic prolactin does not contribute to systemic lupus erythematosus hyperprolactinemia. Clin Exp Rheumatol 28(6): 866–72, Epub 2011/01/06 Lopez-Rincon G, Pereira-Suarez AL, Del Toro-Arreola S, Sanchez- Hernandez PE, Ochoa-Zarzosa A, Munoz-Valle JF et al, 2013, Lipopolysaccharide induces the expression of an autocrine prolactin loop enhancing inflammatory response in monocytes. J Inflamm, Lond, 10(1):24, Epub 2013/06/05 Ginsburg E, Vonderhaar BK, 1995, Prolactin synthesis and secretion by human breast cancer cells. Cancer Res 55(12):2591–5, Epub 1995/06/15 Dagvadorj A, Collins S, Jomain JB, Abdulghani J, Karras J, Zellweger T et al, 2007, Autocrine prolactin promotes prostate cancer cell growth via Janus kinase-2-signal transducer and activator of transcription-5a/b signaling pathway. Endocrinology 148(7): 3089–101, Epub 2007/04/07 Lopez-Pulido EI, Munoz-Valle JF, Del Toro-Arreola S, Jave-Suarez LF, Bueno-TopeteMR, Estrada-Chavez C, Pereira-Suarez AL, 2013, High expression of prolactin receptor is associated with cell survival in cervical cancer cells. Cancer Cell Int 13(1):103 Kossiakoff AA, 2004, The structural basis for biological signaling, regulation, and specificity in the growth hormone-prolactin systemof hormones and receptors. Adv Protein Chem 68:147–69, Epub 2004/10/27 Meng J, Tsai-Morris CH, Dufau ML, 2004, Human prolactin receptor variants in breast cancer: low ratio of short forms to the long-form human prolactin receptor associated with mammary carcinoma. Cancer Res 64(16):5677–82, Epub 2004/08/ 18 Huang KT,WalkerAM(2010, Long termincreased expression of the short form 1b prolactin receptor in PC-3 human prostate cancer cells decreases cell growth and migration, and causes multiple changes in gene expression consistent with reduced invasive capacity. Prostate 70(1):37–47, Epub 2009/09/10 Varghese B, Barriere H, Carbone CJ, Banerjee A, Swaminathan G, Plotnikov A et al, 2008, Polyubiquitination of prolactin receptor stimulates its internalization, postinternalization sorting, and degradation via the lysosomal pathway. Mol Cell Biol 28(17):5275–87, Epub 2008/06/25 Plotnikov A, Li Y, Tran TH, Tang W, Palazzo JP, Rui H et al, 2008, Oncogene-mediated inhibition of glycogen synthase kinase 3 beta impairs degradation of prolactin receptor. Cancer Res 68(5):1354–61, Epub 2008/03/05 Li Y, Clevenger CV, Minkovsky N, Kumar KG, Raghunath PN, Tomaszewski JE et al, 2006, Stabilization of prolactin receptor in breast cancer cells. Oncogene 25(13):1896–902, Epub 2005/11/10 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 241 EP 246 DI 10.1007/s12253-014-9814-6 PG 6 ER PT J AU Vizkeleti, J Vereczkey, I Frohlich, G Varga, Sz Horvath, K Pulay, T Pete, I Nemeskeri, Cs Mayer, Sipos, N Kasler, M Polgar, Cs AF Vizkeleti, Julia Vereczkey, Ildiko Frohlich, Georgina Varga, Szilvia Horvath, Katalin Pulay, Tamas Pete, Imre Nemeskeri, Csaba Mayer, Arpad Sipos, Norbert Kasler, Miklos Polgar, Csaba TI Pathologic Complete Remission after Preoperative High-Dose-Rate Brachytherapy in Patients with Operable Cervical Cancer: Preliminary Results of a Prospective Randomized Multicenter Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; Preoperative brachytherapy; Pathologic complete remission; Surgical margins ID Cervical cancer; Preoperative brachytherapy; Pathologic complete remission; Surgical margins AB The role of preoperative intrauterine brachytherapy (BT) in the multidisciplinary treatment of early stage cervical carcinoma (ESCC) is controversial. In 2005, a prospective randomized multicenter study was initiated in Hungary in order to explore the potential advantages of preoperative high-dose-rate (HDR) BT. In this article we evaluate the efficiency of preoperative HDR BT by the rate of pathologic complete remission (pCR) in the first 185 patients enrolled in the study at the National Institute of Oncology and at the Uzsoki Municipal Cancer Center in collaboration with the 1st Department of Gynaecology and Obstetrics of Semmelweis University, Budapest, Hungary. In arm A, patients received 2x8Gy preoperative intracavitary HDR BT, while in arm B no preoperative treatment was given. In both arms patients underwent radical Wertheim (Piver III) hysterectomy. The pCR rate was 25.7% after preoperative HDR BT, while it was only 11.2% with surgery alone (p=0.03), in these cases the tumor was eliminated during the diagnostic excision or conisation. The rate of positive surgical margins was 1.5% after preoperative BT, while it was as high as 11.4% without preoperative RT (p=0.02). There was no significant difference in the local tumor control (LTC), distant metastases free survival (DMFS) and overall survival (OS) between the two arms. According to our preliminary results preoperative intracavitary HDR BT significantly increases the rate of pCR and decreases the rate of positive surgical margins in patients with ESCC. Longer follow-up is required to establish the possible impact of pCR on the ultimate LTC and OS. C1 [Vizkeleti, Julia] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Vereczkey, Ildiko] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Frohlich, Georgina] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Varga, Szilvia] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Horvath, Katalin] National Institute of Oncology, Center of RadiologyBudapest, Hungary. [Pulay, Tamas] National Institute of Oncology, Center of GynaecologyBudapest, Hungary. [Pete, Imre] National Institute of Oncology, Center of GynaecologyBudapest, Hungary. [Nemeskeri, Csaba] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai KozpontBudapest, Hungary. [Mayer, Arpad] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai KozpontBudapest, Hungary. [Sipos, Norbert] Semmelweis Medical University, Ist Department of Gynaecology and ObstetricsBudapest, Hungary. [Kasler, Miklos] National Institute of OncologyBudapest, Hungary. [Polgar, Csaba] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. RP Vizkeleti, J (reprint author), National Institute of Oncology, Center of Radiotherapy, Budapest, Hungary. EM juliavizkeleti@yahoo.com CR Practice Bulletin ACOG, 2002, Diagnosis and treatment of cervical carcinomas. Int J Gynec Obst 78:79–91 NCCN Clinical Practice Guidelines in Oncology-Cervical Cancer, 2011, www.nccn.org/ Haie-Meder C,Morice P, CastiglioneM; ESMO GuidelinesWorking Group, 2008, Cervical cancer: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol 19 Suppl 2:ii17-8., DOI 10.1093./annonc/mdn112 Atlan D, Touboul E, Deniaud-Alexandre E et al, 2002, Operable stages IB and II cervical carcinomas: A retrospective study comparing preoperative uterovaginal brachytherapy and postoperative radiotherapy. Int J Radiat Oncol Biol Phys 54:780–793 Beskow C, Agren-Cronqvist AK, Granath F et al, 2002, Pathologic complete remission after preoperative intracavitary radiotherapy of cervical cancer stage Ib and IIa is a strong prognostic factor for longterm survival: Analysis of the Radiumhemmet data 1989–1991. Int J Gynecol Cancer 12:158–170 Gerbaulet AL, Kunkler IH, Kerr GR et al, 1992, Combined radiotherapy and surgery: local control and complications in early carcinoma of the uterine cervix – the Villejuif experience, 1975–1984. Radiother Oncol 23:66–73 Huguet F, Cojocariu OM, Levy P et al, 2008, Preoperative concurrent radiation therapy and chemotherapy for bulky stage IB2, IIA, and IIB carcinoma of the uterine cervix with proximal parametrial invasion. Int J Radiat Oncol Biol Phys 72:1508–1515 Muschitz S, Petrow P, Briot E et al, 2004, Correlation between the treated volume, the GTV and the CTV at the time of brachytherapy and the histopathologic findings in 33 patients with operable cervix carcinoma. Radiother Oncol 73:187–194 Resbeut MR, Alzieu C, Gonzague-Casabianca L et al, 2001, Combined brachytherapy and surgery for early carcinoma of the uterine cervix: Analysis of extent of surgery on outcome. Int J Radiat Oncol Biol Phys 50:873–881 Mayer A, Nemeskeri C, Poti Z, 2004, Evaluation of high-doserate brachytherapy in the preoperative treatment of FIGO stage IB cervical carcinoma. Magyar Onkologia [In Hungarian] 48: 141–144 Nemeth G, Naszaly A, Takacsi-Nagy L, 1979, Results of preoperative radiotherapy in cancer of the uterine cervix and corpus regarding tumor regression and 5 year survival. MagyNoorvLapja [in Hungarian] 42:526–528 Nemeth G, 1998, A comparative study on the therapeutic effectiveness of different preoperative high-dose-rate after loading brachytherapies in stage IB cancer of the cervix uteri in relation to the tumour-free state of the surgical preparation. Magyar Onkologia [In Hungarian] 42:169–170 Papp Z, Csapo Z, Mayer A et al, 2006, Wertheim-operation: 5-year survival of 501 consecutive cases of cervical cancer. OrvHetil [in Hungarian] 147:537–545 Poka R, Szluha K, Hernadi Z et al, 1995, Analysis of survival in stage IB and IIA cervical cancer treated with or without preoperative local radiotherapy. Eur J Gynaecol Oncol 16:208–211 Grigsby PW, Perez CA, Chao KSC et al, 1999, Lack of effect of tumor size on the prognosis of carcinoma of the uterine cervix stage IB and IIA treated with preoperative irradiation and surgery. Int J Radiat Oncol Biol Phys 45:645–651 Haie-Meder C, Chargari C, Rey A et al, 2009, DVH parameters and outcome for patients with early-stage cervical cancer treated with preoperative MRI-based low dose rate brachytherapy followed by surgery. Radiother Oncol 93:316–321 Hannoun-Levi JM, Chand-Fouche ME, Gautier M et al, 2013, Interstitial preoperative high-dose-rate brachytherapy for early stage cervical cancer: Dose-volume histogram parameters, pathologic response and early clinical outcome. Brachytherapy 12(2): 148–155 Bialas B, Kellas-Sleczka S, FijalkowskiMet al, 2009, Tolerance and efficacy of preoperative intracavitary HDR brachytherapy in IB and IIA cervical cancer. J Contemp Brachyther 1:38–44 Jacinto AA, Castilho MS, Novaes PE et al, 2007, Preoperative external beam radiotherapy and reduced dose brachytherapy for carcinoma of the cervix: Survival and pathological response. Radiat Oncol 2:9–15 Mundt AJ,Waggoner S, Herbst A et al, 1999, Preoperative intracavitary brachytherapy in early-stage cervical carcinoma. Am J Clin Oncol 22:73–77 Chung CK, Nahhas WA, Stryker JA et al, 1980, Analysis of factors contributing to treatment failures in stages IB and IIA carcinoma of the cervix. Am J Obstet Gynecol 138:550–555 Look KY, Rocereto TF, 1990, Relapse patterns in FIGO IB carcinoma of the cervix. Gynecol Oncol 38:114–120 Larson DM, Copeland LJ, Stringer CA et al, 1988, Recurrent cervical carcinoma after radical hysterectomy. Gynecol Oncol 30:381–387 Uzan C, Merlot B, Gouy S et al, 2013, Laparoscopic Radical Hysterectomy after Preoperative Brachytherapy for stage IB1 cervical cancer: feasibility, results, and surgical implications in a large bicentric study of 162 consecutive cases. Ann Surg Oncol 20:872–880 Pellegrino A, Vizza E, Fruscia R et al, 2009, Total laparoscopic radical hysterectomy and pelvic lymphadenectomy in patients with IB1 stage cervical cancer: analysis of surgical and oncological outcome. Eur J Surg Oncol 35:98–103 Puntambekar SP, Palep RJ, Puntambekar SS et al, 2007, Laparoscopic total radical hysterectomy by the Pune technique: our experience of 248 cases. J Minim Invasive Gynecol 14:682–689 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 247 EP 256 DI 10.1007/s12253-014-9815-5 PG 10 ER PT J AU Hu, YM Li, J Yu, LCh Shi, ShB Du, YJ Wu, JN Shi, LW AF Hu, Yi-Ming Li, Jing Yu, Li-Chao Shi, Shun-Bing Du, Yong-Jie Wu, Jian-Nong Shi, Lin Wei TI Survivin mRNA Level in Blood Predict the Efficacy of Neoadjuvant Chemotherapy in Patients with Stage IIIA-N2 Non-Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; Neoadjuvant chemotherapy; Survivin mRNA; Prognosis; Prediction ID Non-small cell lung cancer; Neoadjuvant chemotherapy; Survivin mRNA; Prognosis; Prediction AB In a previous study, survivin mRNA expression in non-small cell lung cancer (NSCLC) tissue had been demonstrated to be associated with unfavorable prognosis of patients treated with chemotherapy. In this study, we investigated the survivin mRNA levels in blood of patients with stage IIIA-N2 NSCLC and their association with the efficacy of neoadjuvant chemotherapy (NCT) and disease-free survival (DFS) and overall survival (OS). Blood specimens were collected from 56 patients with stage IIIA-N2 NSCLC before (N0) and after the complete of NCT (N1). Survivin mRNA was measured by realtime quantitative-PCR assay. Receiver operating characteristics curve analysis was undertaken to determine the best cutoff value for survivin mRNA. Results showed that high blood survivin mRNA levels at N0 and N1 were significantly associated with clinical (P=0.01 and P=0.008, respectively) and pathologic response (both P=0.004, respectively). Moreover, the change of blood survivin mRNA levels in these NSCLC patients is associated with the clinical and pathologic response to NCT. Patients with high survivin mRNA levels at N0 and N1 had significantly shorter DFS and OS than those with low survivin mRNA levels (P=0.021 and P=0.014, respectively for DFS; P=0.009 and P=0.005, respectively for OS). Multivariate analysis demonstrated that high blood survivin mRNA level was an independent predictor for worse DFS and OS in the NSCLC patients receiving NCT. In conclusion, survivin mRNA level in blood from stage IIIA-N2 NSCLC patients receiving NCT is predictive of cancer outcome. C1 [Hu, Yi-Ming] Affiliated Hospital of Jiangsu University, Department of Pulmonary Medicine, 438 North Jiefang Street, 212001 Zhenjiang, China. [Li, Jing] Affiliated Hospital of Jiangsu University, Department of Pulmonary Medicine, 438 North Jiefang Street, 212001 Zhenjiang, China. [Yu, Li-Chao] Affiliated Hospital of Jiangsu University, Department of Thoracic Surgery, 212001 Zhenjiang, China. [Shi, Shun-Bing] Affiliated Hospital of Jiangsu University, Department of Thoracic Surgery, 212001 Zhenjiang, China. [Du, Yong-Jie] Affiliated Hospital of Jiangsu University, Department of Pulmonary Medicine, 438 North Jiefang Street, 212001 Zhenjiang, China. [Wu, Jian-Nong] Affiliated Hospital of Jiangsu University, Department of Pathology, 212001 Zhenjiang, China. [Shi, Lin Wei] Affiliated Hospital of Jiangsu University, Department of Pulmonary Medicine, 438 North Jiefang Street, 212001 Zhenjiang, China. RP Li, J (reprint author), Affiliated Hospital of Jiangsu University, Department of Pulmonary Medicine, 212001 Zhenjiang, China. EM lijian541226@163.com CR Li J, Dai C-H, Yu L-C et al, 2009, Results of trimodality therapy in patients with stage IIIA, N2-Bulky, and stage IIIB non-small cell lung cancer. Clin Lung Cancer 10:353–359 Roth JA, Atkinson EN, Fossella F et al, 1998, Long-term follow-up of patients enrolled in a randomized trial comparing perioperative chemotherapy and surgery with surgery alone in resectable stage IIIA non-small-cell lung cancer. Lung Cancer 21:1–6 Stefani A, Alifano M, Bobbio A et al, 2010, Which patients should be operated after induction chemotherapy for N2 non-small cell lung cancer? Analysis of a 7 year experience in 175 patients. J Thorac Cardiovase Surg 140:356–363 Rosell R, Gomez-Codina J, Camps C et al, 1999, Preresectional chemotherapy in stage IIIA non-small-cell lung cancer: a 7-year assessment of a randomized clinical trial. Lung Cancer 26:7–14 Depierre A, Milleron B, Moro-Sibilot D et al, 2002, Preoperative chemotherapy followed by surgery compared with primary surgery in resectable stage I, except T1N0), II, and IIIa non-small-cell lung cancer. J Clin Oncol 2:247–253 Song W-A, Zhou N-K, Wang W et al, 2010, Survival benefit of neoadjuvant chemotherapy in non-small cell lung cancer. An updated meta-analysis of 13 randomized control trials. J Thorac Oncol 5:510–516 Detterbeck FC, Boffa DJ, Tanoue LT, 2009, The new lung cancer staging system. Chest 36:260–271 Felip E, Stahel RA, Pavlidis N, 2005, ESMO minimum clinical recommendation for diagnosis, treatment and follow-up of nonsmall- cell lung cancer, NSCLC). Ann Oncol 16:128–129 Ramnath N, Dilling TJ, Harris LJ et al, 2013, Treatment of stage IIIA non-small cell lung cancer: diagnosis and management of lung cancer, 3rd: American College of Chest physicians evidence-based clinical practice guidelines. Chest 143(5 suppl): e 314s–e 340s StraussGM(1999, Role of chemotherapy in stage I-III non-small cell lung cancer. Chest 116(6 suppl):509s–516s Bueno R, Richards WG, Swanson SJ et al, 2000, Nodal stage after induction therapy for stage IIIA lung cancer determines patient survival. Ann Thorac Surg 70:1826–1831 Bepler G, Sommers KE, Cantor A et al, 2008, Clinical efficacy and predictive molecular markers of neoadjuvant gemcitabine and pemetrexed in resectable non-small-cell lung cancer. J Thorax Oncol 3:1112–1118 John T, Starmans MHW, Chen Y-T et al, 2013, The role of cancertestis antigens as predictive and prognostic markers in non-small cell lung cancer. PLoS ONE 8:e67876 Altieri DC, 2003, Validating survivin as a cancer therapeutic target. Nat Rev Cancer 3:46–54 Ambrosini G, Adida C, Alfieri DC, 1997, A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 3:917–921 Velculescu VE, Madden SL, Zhang L et al, 1999, Analysis of human transcriptomes. Nat Genet 23:387–388 Fukud S, Pelus LM(2006, Survivin, a cancer target with an emerging role in normal adult tissue. Mol Cancer Ther 5:1087–1098 Dai C-H, Li J, Shi S-B et al, 2010, Survivin and smac gene expressions but not livin are predictors of prognosis in non-small cell lung cancer patients treated with adjuvant chemotherapy following surgery. Jpn J Clin Oncol 40:327–335 Chen P, Li J, Ge L-P et al, 2010, Prognostic value of survivin, Xlinked inhibitor of apoptosis protein and second mitochondriaderived activator of caspases expression in advanced non-small-cell lung cancer patients. Respirology 15:501–509 Tang X-P, Li J, Yu L-C et al, 2013, Clinical significance of survivin and VEGF mRNA detection in the cell fraction of the peripheral blood in non-small cell lung cancer patients before and after surgery. Lung Cancer 8:273–279 Therasse P, Arbuck SG, Eisenhauer EA et al, 2000, New guideline to evaluate the response to treatment in solid tumor. European Organization for Research and Treatment for Cancer, National Cancer Institute the United State, National Cancer Institute of Canada. J Natl Cancer Inst 92:205–216 de Bore RH, Snith IE, Pastorino U et al, 1999, Pre-operative chemotherapy in early stage resectable non-small-cell lung cancer: a randomized feasibility study justifying a multicenter phase III trial. Br J Cancer 79:1514–1518 Livak KJ, Schimittgen TD, 2001, Analysis of reactive gene expression data using real-time quantitative PCR and 2-△△Ct method. Method 25:402–408 Goldstraw P, Crowley J, Chansky K et al, 2007, The IASLC Lung Cancer Staging Project: proposals for the revision of the TNM stage groupings in the forthcoming, Seventh, edition of the TNM classification of malignant tumors. J Thorac Oncol 2:706–714 Junker K, Langner K, Klinke F et al, 2001, Grading of tumor regression in non-small cell lung cancer: morphology and prognosis. Chest 120:1584–1591 Betticher DC, Schmitz S-FH, Totsch M et al, 2003, Mediastinal lymph node clearance after docetaxel-cisplatin neoadjuvant chemotherapy is prognostic of survival in patients with stage IIIA pN2 nonsmall- cell lung cancer: a multicenter phase II trial. J Clin Oncol 21: 1752–1759 Pisters KM, Ginsbery RJ, Giroux DJ et al, 2000, Induction chemotherapy before surgery for early-stage lung cancer: a novel approach. Bimodality Lung Oncology Team. J Thorac Cardiovase Surg 119: 429–439 Betticher DC, Schmitz S-FH, Totsch M et al, 2006, Prognostic factors affecting long-term outcomes in patients with resected stage IIIA non-small-cell lung cancer: 5 year follow-up of a phase II study. Br J Cancer 94:1099–1106 Singhal S, Vachani A, Antin-Ozerkis D et al, 2005, Prognostic implications of cell cycle, apoptosis, and angiogenesis biomarkers in nonsmall cell lung cancer: a review. Clin Cancer Res 11:3974–3986 Crino L, Weder W, van Meerbeeck J et al, 2010, On behalf of the ESMO GuidelinesWorking Group. Early stage and locally advanced, non-metastatic, non-small-cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 21(supp1 5):v103–v115 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 257 EP 265 DI 10.1007/s12253-014-9816-4 PG 9 ER PT J AU Shen, CC Cui, XY He, Y Kang, YH Yi, Ch Yang, JL Gou, LT AF Shen, Cong-Cong Cui, Xin-Yi He, Yi Kang, Yu-Huan Yi, Cheng Yang, Jin-Liang Gou, Lan-Tu TI High Phosphorylation Status of AKT/mTOR Signal in DESI2-Reduced Pancreatic Ductal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DESI2; AKT; mTOR; Pancreatic ductal adenocarcinoma ID DESI2; AKT; mTOR; Pancreatic ductal adenocarcinoma AB Desumoylating isopeptidase 2 (DESI2) is a recently identified protein with unclear functions. In this study, a total of 132 tissue samples of pancreatic ductal adenocarcinoma and 73 samples of pancreatic normal tissues were explored to assess DESI2 expression and its implications to AKT/ mTOR signal. Immunohistochemistry showed DESI2 expression is significantly decreased in cancer tissues versus normal tissues, presenting lowest level in poorly differentiated cancer. Unlike DESI2, the key factors in AKT/mTOR pathway including p-AKT, mTOR, p-mTOR and p-P70S6K present high expression in pancreatic cancer. It is notable that p-mTOR is significantly increased in DESI2-lower cancer compared with DESI2-higher cancer, although mTOR presents no difference in the two groups. The relative p-mTOR/mTOR ratio is also significantly elevated in DESI2-lower cancer. Moreover, the samples whose p-AKT and p-mTOR scores both exceed the median are obviously increased in DESI2-lower cancer compared with DESI2-higher cancer. As a downstream molecule of AKT/mTOR pathway, p-P70S6K was found to display higher level in DESI2-lower pancreatic cancer. High phosphorylation status of those proteins in DESI2-reduced pancreatic cancer indicates that there is high activity of AKT/mTOR signal in condition of DESI2 reduction, which could provide clues to reveal the implications of DESI2 in carcinogenesis. C1 [Shen, Cong-Cong] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, No. 1, Keyuan Road 4, Gaopeng Street, 610041 Chengdu, Sichuan, China. [Cui, Xin-Yi] Sichuan University, West China Hospital, Division of Abdominal Cancer, Cancer Center, 610041 Chengdu, Sichuan, China. [He, Yi] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, No. 1, Keyuan Road 4, Gaopeng Street, 610041 Chengdu, Sichuan, China. [Kang, Yu-Huan] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, No. 1, Keyuan Road 4, Gaopeng Street, 610041 Chengdu, Sichuan, China. [Yi, Cheng] Sichuan University, West China Hospital, Division of Abdominal Cancer, Cancer Center, 610041 Chengdu, Sichuan, China. [Yang, Jin-Liang] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, No. 1, Keyuan Road 4, Gaopeng Street, 610041 Chengdu, Sichuan, China. [Gou, Lan-Tu] Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, No. 1, Keyuan Road 4, Gaopeng Street, 610041 Chengdu, Sichuan, China. RP Gou, LT (reprint author), Sichuan University, West China Hospital, State Key Laboratory of Biotherapy, 610041 Chengdu, China. EM goulantu@gmail.com CR Gregory SG, Barlow KF,McLay KE, Kaul R, Swarbreck D,Dunham A, 2006, The DNA sequence and biological annotation of human chromosome 1. Nature 441:315–321 MGC Project Team, 2004, The status, quality, and expansion of the NIH full-length cDNA project: the Mammalian Gene Collection, MGC). Genome Res 14:2121–2127 Mammalian Gene Collection Program Team, 2002, Generation and initial analysis of more than 15,000 full-length human and mouse cDNA sequences. Proc Natl Acad Sci U S A 99: 16899–16903 Iyer LM, Koonin EV, Aravind L, 2004, Novel predicted peptidases with a potential role in the ubiquitin signaling pathway. Cell Cycle 3: 1440–1450 Lai CH, Chou CY, Ch’ang LY, Liu CS, Lin W, 2000, Identification of novel human genes evolutionarily conserved in Caenorhabditis elegans by comparative proteomics. Genome Res 10:703–713 Shin EJ, Shin HM, Nam E et al, 2012, DeSUMOylating isopeptidase: a second class of SUMO protease. EMBO Rep 13: 339–346 Yao S, Xie L, Qian M et al, 2008, Pnas4 is a novel regulator for convergence and extension during vertebrate gastrulation. FEBS Lett 582:2325–2332 Yan F,Gou L,Yang J et al, 2009)Anovel pro-apoptosis gene PNAS4 that induces apoptosis in A549 human lung adenocarcinoma cells and inhibits tumor growth in mice. Biochimie 91:502–507 Gou LT, Tong AP, Yan F et al, 2009, Altered protein-expressing profile in hPNAS4-induced apoptosis in A549 human lung adenocarcinoma cells. J Cell Biochem 108:1211–1219 He Y, Wang J, Gou L et al, 2013, Comprehensive analysis of expression profile reveals the ubiquitous distribution of PPPDE peptidase domain 1, a Golgi apparatus component, and its implications in clinical cancer. Biochimie 95:1466– 1475 Hidalgo M, 2010, Pancreatic cancer. N Engl J Med 362:1605–1617 Di Marco M, Di Cicilia R, Macchini M et al, 2010, Metastatic pancreatic cancer: is gemcitabine still the best standard treatment? Oncol Rep 23:1183–1192 Wijesekara N, Krishnamurthy M, Bhattacharjee A, Suhail A, Sweeney G, Wheeler MB, 2010, Adiponectin-induced ERK and Akt phosphorylation protects against pancreatic beta cell apoptosis and increases insulin gene expression and secretion. J Biol Chem 285:33623–33631 Li J, Liang X, Yang X, 2012, Ursolic acid inhibits growth and induces apoptosis in gemcitabine-resistant human pancreatic cancer via the JNK and PI3K/Akt/NF-κB pathways. Oncol Rep 28:501–510 Javle MM, Shroff RT, Xiong H et al, 2010, Inhibition of the mammalian target of rapamycin, mTOR, in advanced pancreatic cancer: results of two phase II studies. BMC Cancer 10:368 Wolpin BM, Hezel AF, Abrams T et al, 2009, Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol 27:193–198 Sabatini DM, 2006, mTOR and cancer: insights into a complex relationship. Nat Rev Cancer 6:729–734 O’Reilly KE, Rojo F, She QB et al, 2006, mTOR inhibition induces upstreamreceptor tyrosine kinase signaling and activates Akt. Cancer Res 66:1500–1508 Kreisberg JI, Malik SN, Prihoda TJ et al, 2004, Phosphorylation of Akt, Ser473, is an excellent predictor of poor clinical outcome in prostate cancer. Cancer Res 64:5232–5236 Feligioni M, Brambilla E, Camassa A et al, 2011, Crosstalk between JNK and SUMO signaling pathways: deSUMOylation is protective against H2O2-induced cell injury. PLoS One 6: e28185 Dou H, Huang C, SinghM, Carpenter PB, Yeh ET, 2010, Regulation of DNA repair through deSUMOylation and SUMOylation of replication protein A complex. Mol Cell 39:333–345 Wang Y, Dasso M, 2009, SUMOylation and deSUMOylation at a glance. J Cell Sci 122:4249–4252 Baker SP, Phillips J, Anderson S et al, 2010, Histone H3 Thr 45 phosphorylation is a replication-associated post-translational modification in S. cerevisiae. Nat Cell Biol 12:294–298 Ma J, Sawai H, Ochi N et al, 2009, PTEN regulates angiogenesis through PI3K/Akt/VEGF signaling pathway in human pancreatic cancer cells. Mol Cell Biochem 331:161–171 Michl P, Gress TM, 2013, Current concepts and novel targets in advanced pancreatic cancer. Gut 62:317–326 Cao W, Manicassamy S, Tang H et al, 2008, Toll-like receptormediated induction of type I interferon in plasmacytoid dendritic cells requires the rapamycin-sensitive PI(3)K-mTOR-p70S6K pathway. Nat Immunol 9:1157–1164 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 267 EP 272 DI 10.1007/s12253-014-9817-3 PG 6 ER PT J AU Pavlakis, K Bobos, M Batistatou, A Kotoula, V Eleftheraki, GA Stofas, A Timotheadou, E Pentheroudakis, G Psyrri, A Koutras, A Pectasides, D Papakostas, P Razis, E Christodoulou, Ch Kalogeras, TK Fountzilas, G AF Pavlakis, Kitty Bobos, Mattheos Batistatou, Anna Kotoula, Vassiliki Eleftheraki, G Anastasia Stofas, Anastasios Timotheadou, Eleni Pentheroudakis, George Psyrri, Amanda Koutras, Angelos Pectasides, Dimitrios Papakostas, Pavlos Razis, Evangelia Christodoulou, Christos Kalogeras, T Konstantine Fountzilas, George TI p85 Protein Expression is Associated with Poor Survival in HER2-Positive Patients with Advanced Breast Cancer Treated with Trastuzumab SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p85; Prognostic factors; Breast carcinoma; Immunohistochemistry; HER2 status; Trastuzumab ID p85; Prognostic factors; Breast carcinoma; Immunohistochemistry; HER2 status; Trastuzumab AB To investigate the immunohistochemical expression of p85 in a cohort of trastuzumab-treated HER2- positive and HER2-negative metastatic breast cancer patients. The medical records of all patients with metastatic breast cancer treated with trastuzumab-based regimens between 1998 and 2010 were reviewed and clinical information was obtained. Formalin-fixed paraffin-embedded tumor tissue samples with adequate material were retrospectively collected from 183 patients. Samples were evaluated by immunohistochemistry for p85, estrogen receptors (ER), progesterone receptors (PgR), HER2, Ki67, PTEN and phosphorylated Akt (S473 and T308). HER2 status was studied by fluorescence in situ hybridization, as well. PIK3CA mutational status was also evaluated. Median follow-up for all patients was 72 months. Central re-evaluation for HER2 revealed only 111 HER2- positive cases, with the remaining 72 patients being HER2- negative. Median survival was longer in HER2-positive patients (50.7 months) compared to HER2-negative patients (36.6 months) both treated with trastuzumab, but this difference has not reached significance (p=0.068). In total, 62 % of the patients were found positive for p85, however the p85 protein was not found to be differentially expressed in HER2- positive versus HER2-negative cases. There were no significant associations between protein expression of p85 and any of the markers under study, or with time to progression. Positive p85 protein expression was however associated with poor survival in trastuzumab-treated HER2-positive patients. In our cohort of trastuzumab-treated HER2-positive breast cancer patients, positive p85 protein expression appears to be a prognostic factor of poor survival and, if validated, might have important implications in the treatment of such patients. C1 [Pavlakis, Kitty] University of Athens, Medical School, Department of PathologyAthens, Greece. [Bobos, Mattheos] Aristotle University of Thessaloniki School of Medicine, Hellenic Foundation for Cancer Research, Laboratory of Molecular OncologyThessaloniki, Greece. [Batistatou, Anna] Ioannina University Hospital, Department of PathologyIoannina, Greece. [Kotoula, Vassiliki] Aristotle University of Thessaloniki, AHEPA University Hospital, Pathology DepartmentThessaloniki, Greece. [Eleftheraki, G Anastasia] Data Office, Hellenic Cooperative Oncology Group, Section of BiostatisticsAthens, Greece. [Stofas, Anastasios] University of Athens, Medical School, Department of PathologyAthens, Greece. [Timotheadou, Eleni] Aristotle University of Thessaloniki School of Medicine, Department of Medical OncologyThessaloniki, Greece. [Pentheroudakis, George] Ioannina University Hospital, Department of Medical OncologyIoannina, Greece. [Psyrri, Amanda] Attikon University Hospital, Second Department of Internal MedicineAthens, Greece. [Koutras, Angelos] University of Patras Medical School, University Hospital, Division of Oncology, Department of MedicinePatras, Greece. [Pectasides, Dimitrios] Attikon University Hospital, Second Department of Internal MedicineAthens, Greece. [Papakostas, Pavlos] University of Athens, 3rd Department of Medicine, Oncology UnitAthens, Greece. [Razis, Evangelia] University of Athens, 3rd Department of Medicine, Oncology UnitAthens, Greece. [Christodoulou, Christos] Second Department of Medical OncologyPiraeus, Greece. [Kalogeras, T Konstantine] Aristotle University of Thessaloniki School of Medicine, Department of Medical OncologyThessaloniki, Greece. [Fountzilas, George] Aristotle University of Thessaloniki School of Medicine, Hellenic Foundation for Cancer Research, Laboratory of Molecular OncologyThessaloniki, Greece. RP Pavlakis, K (reprint author), University of Athens, Medical School, Department of Pathology, Athens, Greece. EM epavlaki@med.uoa.gr CR Geering B, Cutillas PR, Nock G, 2007, Class IA phosphoinositide 3- kinases are obligate p86-p110 heterodimers. PNAS 104(19):7809– 7814 Cantrell DA, 2001, Phosphoinositide 3-kinase signaling pathways. J Cell Sci 114:1439–1445 Katso R, Okkenhaug K, Ahmadi K et al, 2001, Cellular function of phosphoinositide 3-kinases: implications for development, homeostasis and cancer. Annu Rev Cell Dev Biol 17: 615–675 Domin J, Waterfield MD, 1997, Using structure to define the function of phosphoinositide 3-kinase family members. FEBS Lett 410: 91–95 Zito CR, Jilaveanou LB, Anagnostou V et al, 2012, Multi-level targeting of the phosphatidylinositol-3-kinase pathway in non-small cell lung cancer cells. PLoS One 7(2):e31331 Cantley LC, 2002, The phosphoinositide 3-kinase pathway. Science 31(296):1655–7 Vanhaesebroeck B, Waterfield MD, 1999, Signaling by distinct classes of phosphoinisitide 3-kinases. Exp Cell Res 253: 239–254 Stambolic V, Suzuki A, de la Pompa JL et al, 1998, Negative regulation of PKB/Akt dependent survival by the tumor suppressor PTEN. Cell 95:29–39 Chagpar RB, Links PH, Pastor MC et al, 2010, Direst positive regulation of PTEN by the p85 subunit of phosphatidylinositol 3- kinase. PNAS 23(107):5471–5476 Huang CH, Mandelker D, Gabelli SB et al, 2008, Insights into the oncogenic effects of PIK3CA mutations from the structure of p110α/ p85α. Cell Cycle 7(9):1151–1156 Razis E, Bobos M, Kotoula V et al, 2011, Evaluation of the association of PIK3CA mutations and PTEN loss with efficacy of trastuzumab therapy in metastatic breast cancer. Breast Cancer Res Treat 128:447–456 Skarlos P, Christodoulou C, Kalogeras KT et al, 2012, Triplenegative phenotype is of adverse prognostic value in patients treated with dose-dense sequential adjuvant chemotherapy: a translational research analysis in the context of a hellenic cooperative oncology group, HeCOG, randomized phase III trial. Cancer Chemother Pharmacol 69(2):533–546 Bamias A, Karina M, Papakostas P et al, 2010, A randomized phase III study of adjuvant platinum/docetaxel chemotherapy with or without radiation therapy in patients with gastric cancer. Cancer Chemother Pharmacol 65(6):1009– 1021 Magkou C, Mylona E, Theohari I et al, 2007, An immunohistochemical evaluation of phosphorylated Akt at threonine 308 [pAkt, Thr308)] in invasive breast cancer. In Vivo 21(6): 967–972 Rojo F, Najera L, Likola J et al, 2007, 4E-binding protein 1, a cell signaling hallmark in breast cancer that correlates with pathologic grade and prognosis. Clin Cancer Res 13(1):81–89 Hammond ME, Hayes DF, Dowsett M et al, 2010, American society of clinical oncology/college of american pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol 28:2784–2795 Wolff AC, Hammond ME, Schwartz JN et al, 2007, American society of clinical oncology/college of American pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol 25(1):118– 145 CheangMC, Chia SK,VoducD et al, 2009)Ki67 index,HER2 status and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 101(10):736–750 McShane LM, Altman DG, Sauerbrei W et al, 2005, Statistics subcommittee of the NCI-EORTC working group on cancer diagnostics. Reporting recommendations for tumor marker prognostic studies. J Clin Oncol 23:9067–9072 Chamberlain MD, Berry TR, Pastor MC et al, 2004, The p85α subunit of phosphatidylinositol 3-kinase binds to and stimulates the GTPase activity of Rab protein. J Biol Chem 279(47):48607–48614 De Santis G, Miotti S, Mazzi M et al, 2009, E-cadherin directly contributes to PI3K/AKT activation by engaging the PI3K-p85 regulatory subunit to adherens junctions of ovarian carcinoma cells. Oncogene 28(9):1206–1217 Luo J, Sobkiw CL, Logsdon NM et al, 2005, Modulation of epithelial neoplasia and lymphoid hyperplasia in PTEN+/− mice by the p85 regulatory subunits of phosphoinisitide 3-kinase. Proc Natl Acad Sci U S A 102(29):10238–10243 Aziz SA, Davies M, Pick E et al, 2009, Phosphatidylinositol 3-kinase as a therapeutic target in melanoma. Clin Cancer Res 15(9):3029–3036 Lin X, Bohle AS, Dohrmann P et al, 2001, Overexpression of phosphatidylinositol 3-kinase in human lung cancer. Langenbecks Arch Surg 386(4):293–301 El Sheikh SS, Domin J, Tomtitchong P et al, 2003, Topographical expression of class IA and class II phosphoinositide 3-kinase enzymes in normal human tissues is consistent with a role in differentiation. BMC Clin Pathol 3(1):4 Gershtein ES, Scherbakov AM, Shatskaya VA et al, 2007, Phosphatidylinositol 3-kinase/AKT signaling pathway components in human breast cancer: clinicopathological correlations. Anticancer Res 27:1777–1782 Yonezawa M,Wada K, Tatsuguchi A et al, 2009, Heregulin-induced VEGF expression via the ErbB3 signaling pathway in colon cancer. Digestion 80(4):215–225 Nagata R, Lan KH, Zhou X et al, 2004, PTEN activation contributes to tumor inhibition by transtuzumab and loss of PTEN predicts transtuzumab resistance in patients. Cancer Cell 6:117–127 Chan TW, Pollak M, Huynh H, 2001, Inhibition of insulin-like growth factor signaling pathways in mammary gland by pure antiestrogen ICI 182,780. Clin Cancer Res 7(8):2545–2554 Ritter CA, Perez-TorresM, Rinehart C et al, 2007, Human breast cancer cells selected for resistance to transtuzumab in vivo overexpress epidermal growth factor receptor and ErbB ligands and remain dependent on the ErbB receptor network. Clin Cancer Res 13(16):4909–4919 Kakarala M, Wicha MS, 2008, Implications of the cancer stem-cell hypothesis for breast cancer prevention and therapy. J Clin Oncol 26(17):2813–2820 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 273 EP 282 DI 10.1007/s12253-014-9818-2 PG 10 ER PT J AU Sun, G Zhang, J Ma, BX Wang, Y Cheng, JY Hu, W AF Sun, Guogui Zhang, Jianguo Ma, B X Wang, Yadi Cheng, J Y Hu, Wanning TI Overexpression of Wild-Type p53-Induced Phosphatase 1 Confers Poor Prognosis of Patients with Nasopharyngeal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Wip1; Nasopharyngeal cancer; Cell proliferation; Cell apoptosis; Prognosis ID Wip1; Nasopharyngeal cancer; Cell proliferation; Cell apoptosis; Prognosis AB This study aimed to analyze the expression, clinical significance of proto-oncogene in nasopharyngeal carcinoma and the biological effect in its cell line by siRNA targeting wild-type p53-induced phosphatase 1 (Wip1). Immunohistochemistry and western blot were respectively used to analyze Wip1 protein expression in 85 cases of nasopharyngeal cancer and normal tissues to study the relationship between Wip1 expression and clinical factors. Wip1 siRNA was transiently transfected into papillary nasopharyngeal carcinoma cell by liposome-mediated method and was detected by Quantitative real-time RT-PCR (qRT-PCR) and western blot. MTT-assay, cell apoptosis, migration and invasion were also conducted as to the influence of the down-regulated expression of Wip1 that might be found on CNE2 cells biological effect. The level of Wip1 protein expression was found to be significantly higher in nasopharyngeal cancer tissue than normal tissues (P <0.05). There were significant differences between Wip1 expression and T stages, lymph node metastasis, clinical stages, tumor differentiation and radiotherapy response (P<0.05), regardless of age, gender (P>0.05). Meanwhile, Increased expression of Wip1 was significantly with poor overall survival time by Kaplan- Meier analysis (P<0.05). Wip1 expression deletion determines independent risk factors for prognosis of patients with nasopharyngeal carcinoma in addition to tumor T stage, clinical stage, histological grade and lymph node metastasis outside by Cox-2 in the regression analysis (P<0.05). qRT-PCR and Western blot showed that CNE2 cell transfected Wip1 siRNA had a lower relative expressive content than normal cell (P<0.05). MTT assay, cell apoptosis, cell cycles demonstrated that CNE2 cell transfected Wip1 siRNA had a lower survival fraction, higher cell apoptosis, more percentage of the G0/G1 phases, significant decrease in migration and invasion, and higher P53 and P16 protein expression compared with CNE2 cell untransfected Wip1 siRNA (P<0.05).Wip1 protein was increased in nasopharyngeal carcinoma, specifically in T stages, lymph node metastasis, clinical stages and tumor differentiation. Wip1 may involved in the biological processes of nasopharyngeal cancer cell proliferation, apoptosis, and migration and invasion by regulation P53 and P16 protein expression. C1 [Sun, Guogui] Tangshan People’s Hospital, Department of Chemoradiotherapy, NO.65, Shengli road, Lunan district, 063000 Tangshan, Hebei province, China. [Zhang, Jianguo] Tangshan People’s Hospital, Department of Pathology, 063000 Tangshan, Hebei province, China. [Ma, B X] Tangshan People’s Hospital, Department of Pathology, 063000 Tangshan, Hebei province, China. [Wang, Yadi] The Military General Hospital of Beijing PLA, Department of Radiotherapy, 100700 Beijing, China. [Cheng, J Y] The Fourth Hospital of Hebei Medical University, Department of Radiotherapy, 050017 Shijiazhuang, China. [Hu, Wanning] Tangshan People’s Hospital, Department of Chemoradiotherapy, NO.65, Shengli road, Lunan district, 063000 Tangshan, Hebei province, China. RP Hu, W (reprint author), Tangshan People’s Hospital, Department of Chemoradiotherapy, 063000 Tangshan, China. EM wanning_hu2008@sina.com CR Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, 2012, Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010:a systematic analysisfor the global burden of disease study 2010. Lancet 380(9859):2095–128 Wei WI, Sham JS, 2005, Nasopharyngeal carcinoma. Lancet 365(9476):2041–2054 Chang ET, Adami HO, 2006, The enigmatic epidemiology of nasopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev 15(10): 1765–1777 Lo KW, To KF, Huang DP, 2004, Focus on nasopharyngeal carcinoma. Cancer Cell 5(5):423–428 Fuku T, Semba S, Yutori H, Yokozaki H, 2007, Increased wild-type p53-induced phosphatase 1, Wip1 or PPM1D, expression correlated with downregulation of checkpoint kinase 2 in human gastric carcinoma. Pathol Int 57(9):566–571 Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT, Christian MC, Gwyther SG, 2000, New guidelines to evaluate the response to treatment in solid tumors. European organization for research and treatment of cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 92(3):205–216 McCarty KS Jr, Szabo E, Flowers JL, Cox EB, Leight GS, Miller L, Konrath J, Soper JT, Budwit DA, Creasman WT, 1986, Use of a monoclonal anti-estrogen receptor antibody in the immunohistochemical evaluation of human tumors. Cancer Res 46(8 Suppl): 4244s–4248s Turashvili G, Bouchal J, Ehrmann J, Fridman E, Skarda J, Kolar Z, 2007, Novel immunohistochemical markers for the differentiation of lobular and ductal invasive breast carcinomas. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 151(1):59–64 Rai-el-Balhaa G, Pellerin JL, Bodin G, Abdullah A, Hiron H, 1985, Lymphoblastic transformation assay of sheep peripheral blood lymphocytes: a new rapid and easy-to-read technique. Comp Immunol Microbiol Infect Dis 8(3–4):311–318 Vermes I, Haanen C, Steffens-Nakken H, Reutelingsperger C, 1995, Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V. J Immunol Methods 184(1):39–51, A novel assay for apoptosis Li J, Yang Y, Peng Y, Austin RJ, van Eyndhoven WG, Nguyen KC, Gabriele T,McCurrachME, Marks JR, Hoey T, Lowe SW, Powers S, 2002, Oncogenic properties of PPM1D located within a breast cancer amplification epicenter at 17q23. Nat Genet 31(2):133–134 Saito-Ohara F, Imoto I, Inoue J, Inoue J, Hosoi H, Nakagawara A, Sugimoto T, Inazawa J, 2003, PPM1D is a potential target for 17q gain in neuroblastoma. Cancer Res 63(8):1876–1883 Mendrzyk F, Radlwimmer B, Joos S, Kokocinski F, Benner A, Stange DE, Neben K, Fiegler H, Carter NP, Reifenberger G, Korshunov A, Lichter P, 2005, enomic and protein expression profiling identifies CDK6 as novel in dependent prognostic marker in medulloblastoma. J Clin Oncol 23(34):8853–8862 Yang DH, He JA, Li J, Ma WF, Hu XH, Xin SJ, Duan ZQ, 2010, Expression of proto-oncogene Wip1 in breast cancer and its clinical significance. Nat Med J Chin 90(8):519–522 Ling CH, Jiao BH, Lu SK, Guo EK, Zhang GY, 2011, The Expression of proto-oncogene Wip1 in human glioblastoma multiforme and cell lines. Chin J Neurol Oncol 9(1):1–6 Ling CH, Jiao BH, Guo EK, Lu SK, 2011, Over-expression of protooncogene Wip1 in intracranial ependymomas association with P53. Basic Clin Med 31(4):430–434 Yang DH, Zhang H, Hu XH, Xin SJ, Duan ZQ, 2011, Abnormality of p16/p38MAPK/p53/Wip1 pathway in papillary thyroid cancer and its significance. Chin J Gen Surg 20(11):1199–1202 Oliva TM, Erthonaud BV, Chevalier A, Ducrot C, Marsolier-Kergoat MC, Mann C, Leteurtre F, 2007, The Wip1 phosphatase, PPM1D, antagonizes activation of the Chk2 tumour suppressor kinase. Oncogene 26(10):1449–1458 Liao Q, Guo X, Li X, XiongW, Li X, Yang J, Chen P, ZhangW, Yu H, Tang H, DengM, Liang F,Wu M, Luo Z,Wang R, Zeng X, Zeng Z, Li G, 2013, Prohibitin is an important biomarker for nasopharyngeal carcinoma progression and prognosis. Eur J Cancer Prev 22(1): 68–76 Zhou JY, Chong VF, Khoo JB, Chan KL, Huang J, 2007, The relationship between nasopharyngeal carcinoma tumor volume and TNM T- classification:a quantitativeanalysis. Eur Arch Otorhinolaryngol 264(2):169–174 Wu Z, Zeng RF, Su Y, Gu MF, Huang SM, 2013, Prognostic significance of tumor volume in patients with nasopharyngeal carcinoma undergoing intensity-modulated radiation therapy. Head Neck 35(5):689–694 Wang P, Rao J, Yang HF, Zhao HY, Yang L, 2010, Inhibitory effects of lentivirus mediated RNA interference targeting humanWip1 gene on the prolif-eration of human glioma U251 cells. Chin J Exp Surg 27(4):76–79 Baxter EW, Milner J, 2010, p53 Regulates LIF expression in human medulloblastoma cells. J Neuro-Oncol 97(3):373–382 Lowe JM, Cha H, Yang Q, Fornace AJ Jr, 2010, Nuclear factor- kappaB, NF-kappaB, is a novel positive transcriptional regulator of the oncogenic Wip1 phosphatase. J Biol Chem 285(8):5249–5257 Armstrong NJ, Fagotto F, Pthmann C, Rupp RA, 2012, Maternal Wnt/β-catenin signaling coactivates transcription through NF-κB binding sites during Xenopus axis ormation. PLoS One 7(5):e36136 Pan H, ZhouW, HeW, LiuX,DingQ, Ling L, Zha X,Wang S, 2012, Genistein inhibits MDA-MB-231 triple- negative breast cancer cell growth by inhibiting NF-kappaB activity via the Notch-1 pathway. Int J Mol Med 30(2):337–343 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 283 EP 291 DI 10.1007/s12253-014-9819-1 PG 9 ER PT J AU Nourashrafeddin, S Aarabi, M Modarressi, HM Rahmati, M Nouri, M AF Nourashrafeddin, Seyedmehdi Aarabi, Mahmoud Modarressi, Hosein Mohammad Rahmati, Marveh Nouri, Mohammad TI The Evaluation of WBP2NL-Related Genes Expression in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; WBP2NL-related genes; qRT-PCR ID Breast cancer; WBP2NL-related genes; qRT-PCR AB Breast cancer is the most frequent cause of mortality in women all around the world; therefore, study on molecular aspects of breast cancer is necessary for finding new biomarkers. Recent studies have shown that WW Binding Protein 2 (WBP2) is an important protein for the oncogenic property of cancer. We have previously evaluated the WW Binding Protein 2 N-Terminal Like (WBP2NL) gene expression in cancerous cell line and breast tumor tissues, and reported changes in expression, which could increase tumorigenic cell growth. However, the molecular mechanisms of WBP2NL and its clinical relevance have not been investigated. In this study, the expression of WBP2NL-related genes in the invasive breast carcinoma and normal breast tissues was evaluated for the first time. Analysis of WBP2NL-related genes expression was performed with reverse transcription-PCR and real time-PCR detection method. The target genes studied were as follow: WW domain containing E3 ubiquitin protein ligase 1(WWP1),membrane associated guanylatekinase containing WW and PDZ domain-1 (MAGI1), neural precursor cell expressed developmentally down-regulated 4 (NEDD4), formin binding protein-4 (FNBP4), BCL2-associated athanogene-3 (BAG3), WW domain-containing oxidoreductase (WWOX), yes-associated protein-1 (YAP1), WW domain containing transcription regulator (WWTR1), member RAS oncogene family (RAB2A), and small G protein signaling modulator 3 (SGSM3). The expression of WWP1, BAG3, and WWTR1 was significantly increased in breast cancer. In contrast, the expression of WWOX, YAP1, RAB2A, and SGSM3 was significantly decreased. The MAGI1 and NEDD4 expression was increased, while the expression of FNBP4 was unchanged. These findings lead us to suggest that WBP2NL might play roles as an antiapoptotic factor or co-activator to promote breast cancer cell survival and proliferation. C1 [Nourashrafeddin, Seyedmehdi] Tabriz University of Medical Sciences, Women’s Reproductive Health Research CenterTabriz, Iran. [Aarabi, Mahmoud] Queen’s University, Department of Anatomy and Cell BiologyKingston, Canada. [Modarressi, Hosein Mohammad] Tehran University of Medical Sciences, Faculty of Medicine, Department of Medical GeneticsTehran, Iran. [Rahmati, Marveh] Tabriz University of Medical Science, Faculty of Medicine, Department of BiochemistryTabriz, Iran. [Nouri, Mohammad] Tabriz University of Medical Sciences, Women’s Reproductive Health Research CenterTabriz, Iran. CR Patsialou A, Wang Y, Lin J, Whitney K, Goswami S, Kenny PA, Condeelis JS, 2012, Selective gene-expression profiling of migratory tumor cells in vivo predictsclinical outcome in breast cancer patients. Breast Cancer Res 14(5):R139 Key TJ, Verkasalo PK, Banks E, 2001, Epidemiology of breast cancer. The Lancet Oncology 2(3):133–140 Nishidate T, Katagiri T, Lin ML, Mano Y, Miki Y, Kasumi F, Yoshimoto M, Tsunoda T, Hirata K, Nakamura Y, 2004, Genomewide gene-expression profiles of breast-cancer cells purified with laser microbeam microdissection: identification of genes associated with progression and metastasis. Int J Oncol 25(4):797–819 Chen HI, SudolM(1995, TheWWdomain ofYes-associated protein binds a proline-rich ligand that differs from the consensus established for Src homology 3-binding modules. Proc Natl Acad Sci U S A 92(17):7819–23 Sudol M, Chen HI, Bougeret C, Einbond A, Bork P, 1995, Characterization of a novel protein-binding module—the WW domain. FEBS Lett 369:67–71 Komuro A, Nagai M, Navin NE, Sudol M, 2003, WW domaincontaining protein YAP associates with ErbB-4 and acts as a cotranscriptional activator for the carboxyl-terminal fragment of ErbB-4 that translocates to the nucleus. J Biol Chem 278:33334–33341 OverholtzerM, Zhang J, Smolen GA,Muir B, LiW, Sgroi DC, Deng CX, Brugge JS, Haber DA, 2006, Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon. Proc Natl Acad Sci USA 103:12405–12410 Sudol M, 1996, Structure and function of the WW domain. Prog Biophys Mol Biol 65:113–132 Sudol M, Hunter T, 2000, NeW wrinkles for an old domain. Cell 103(7):1001–4 Macias MJ,Wiesner S, Sudol M, 2002)WWand SH3 domains, two different scaffolds to recognize proline-rich ligands. FEBS Lett 513(1):30–7 McDonald CB, McIntosh SK, Mikles DC, Bhat V, Deegan BJ, Seldeen KL, Saeed AM, Buffa L, Sudol M, Nawaz Z, Farooq A, 2011, Biophysical analysis of binding ofWWdomains of the YAP2 transcriptional regulator to PPXY motifs within WBP1 and WBP2 adaptors. Biochemistry 50:9616–9627 Wang K, Degerny C, XuM, Yang XJ, 2009, YAP, TAZ, and Yorkie: a conserved family of signal-responsive transcriptional coregulators in animal development and human disease. Biochem Cell Biol 87(1): 77–91 Chan SW, Lim C, Huang C, Chong YF, Gunaratne HJ, Hogue KA, Blackstock WP, Harvey KF, Hong W, 2011, WW domain-mediated interaction with Wbp2 is important for the oncogenic property of TAZ. Oncogene 30:600–610 de Cristofaro T, Di Palma T, Ferraro A, Corrado A, Lucci V, Franco R, Fusco A, Zannini M, 2011, TAZ/ WWTR1 is overexpressed in papillary thyroid carcinoma. Eur J Cancer 47:926–33 15- Wang L, Chen Z, Wang Y, Chang D, Su L, Guo Y, Liu C, 2013, WWTR1 promotes cell proliferation and inhibits apoptosis through cyclin A and CTGF regulation in non-small cell lung cancer. Tumour Biol. [Epub ahead of print] Sudol M, 2010, Newcomers to the WW domain-mediated network of the hippo tumor suppressor pathway. Genes Cancer 1(11):1115–8 Zhang X, Milton CC, Poon CL, Hong W, Harvey KF, 2011, Wbp2 cooperates with yorkie to drive tissue growth downstream of the Salvador-warts-hippo pathway. Cell Death Differ 18(8):1346–55 Wu AT, Sutovsky P, Xu W, Vander Spoel AC, Platt FM, Oko R, 2007, The postacrosomal assembly of sperm head protein, PAWP, is independent of acrosome formation and dependent on microtubular manchette transport. Dev Biol 312(2):471–83 Sutovsky P, Manandhar G, Wu A, Oko R, 2003, Interactions of sperm perinuclear theca with the oocyte: implications for oocyte activation, anti-polyspermy defense, and assisted reproduction. Microsc Res Tech 61(4):362–78 Wu AT, Sutovsky P, Manandhar G, Xu W, Katayama M, Day BN, Park KW, Yi YJ, Xi YW, Prather RS, Oko R, 2007, PAWP, A sperm specific ww-domain binding protein, promotes meiotic resumption and pronuclear development during fertilization. J Biol Chem 282(16):12164–75 Oko R, Sutovsky P, 2009, Biogenesis of sperm perinuclear theca and its role in sperm functional competence and fertilization. J Reprod Immunol 83(1–2):2–7 Aarabi M, Qin Z, Xu W, Mewburn J, Oko R, 2010, Sperm-borne protein, PAWP, initiates zygotic development in Xenopus laevis by eliciting intracellular calciumrelease.Mol Reprod Dev 77(3):249–56 Nourashrafeddin S, Aarabi M, Miryounesi M, Ebrahimzadeh-Vesal R, Zarghami N, Modarressi MH, Nouri M, 2014, Expression analysis of PAWP during mouse embryonic stem cell-based spermatogenesis in vitro. Vitro Cell Dev Biol Anim 50(5):475–81 Gyorffy B, Lanczky A, Eklund AC, Denkert C, Budczies J, Li Q, Szallasi Z, 2010, An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Res Treat 123:725–731 von Mering C, Jensen LJ, KuhnM, Chaffron S, Doerks T, Kruger B, Snel B, Bork P, 2007, STRING 7 - recent developments in the integration and prediction of protein interactions. Nucleic Acids Res 35:D358–D362 Dhananjayan SC, Ramamoorthy S, Khan OY, Ismail A, Sun J, Slingerland J, O’Malley BW, Nawaz Z, 2006, WW domain binding protein-2, an E6-assoiated protein interacting protein, acts as a coactivator of estrogen and progesterone recep- tors. Mol Endocrinol 20: 2343–54 Lim SK, Orhant-Prioux M, ToyW, Tan KY, Lim YP, 2011, Tyrosine phosphorylation of transcriptional coactivator WW-domain binding protein 2 regulates estrogen receptor α function in breast cancer via the Wnt pathway. FASEB J 25(9):3004–18 Buffa L, Saeed AM, Nawaz Z, 2013)Molecular mechanism ofWWdomain binding protein-2 coactivation function in estrogen receptor signaling. IUBMB Life 65(1):76–84 Xie M, Zhang L, He CS, Hou JH, Lin SX, Hu ZH, Xu F, Zhao HY, 2012, Prognostic significance of TAZ expression in resected nonsmall cell lung cancer. J Thorac Oncol 7(5):799–807 Zhou Z, Hao Y, Liu N, Raptis L, Tsao MS, Yang X, 2011, TAZ is a novel oncogene in non-small cell lung cancer. Oncogene 30(18):2181–6 Chan SW, Lim CJ, Guo K, Ng CP, Lee I, HunzikerW, Zeng Q, Hong W, 2008, A role for TAZ in migration, invasion, and tumorigenesis of breast cancer cells. Cancer Res 68(8):2592–8 Chen C, Sun X, Guo P, Dong XY, Sethi P, Zhou W et al, 2007, Ubiquitin E3 ligaseWWP1 as an oncogenic factor in human prostate cancer. Oncogene 26:2386–2394 Li Y, Zhou Z, Alimandi M, Chen C, 2009, WW domain containing E3 ubiquitin protein ligase 1 targets the full-length ErbB4 for ubiquitin-mediated degradation in breast cancer. Oncogene 28(33): 2948–58 Zhao D, Zhi X, Zhou Z, Chen C, 2012, TAZ antagonizes theWWP1- mediated KLF5 degradation and promotes breast cell proliferation and tumorigenesis. Carcinogenesis 33(1):59–67 Rosati A, Graziano V, De Laurenzi V, Pascale M, Turco MC, 2011, BAG3: amultifaceted protein that regulatesmajor cell pathways. Cell Death Dis 2:e141 Chen Y, Yang LN, Cheng L, Tu S, Guo SJ, Le HY, Xiong Q, Mo R, Li CY, Jeong JS, Jiang L, Blackshaw S, Bi LJ, Zhu H, Tao SC, Ge F, 2013, BAG3 interactome analysis reveals a new role in modulating proteasome activity. Mol Cell Proteomics 12(10):2804–19 Boiani M, Daniel C, Liu X, Hogarty MD, Marnett LJ, 2013, The stress protein BAG3 stabilizes Mcl-1 protein and promotes survival of cancer cells and resistance to antagonist ABT-737. J Biol Chem 288(10):6980–90 Qu J, Lu W, Li B, Lu C, Wan X, 2013, WWOX induces apoptosis and inhibits proliferation in cervical cancer and cell lines. Int J Mol Med 31(5):1139–47 Gothlin Eremo A, Wegman P, Stal O, Nordenskjold B, Fornander T, Wingren S, 2013, Wwox expression may predict benefit from adjuvant tamoxifen in randomized breast cancer patients. Oncol Rep 29(4):1467–74 Yang H, Sasaki T, Minoshima S, Shimizu N, 2007, Identification of three novel proteins, SGSM1, 2, 3, which modulate small G protein, RAP and RAB)-mediated signaling pathway. Genomics 90(2):249– 60 ChiaWJ, Tang BL, 2009, Emerging roles for Rab family GTPases in human cancer. Biochim Biophys Acta 1795:110–6 Diep CH, Zucker KM, Hostetter G,Watanabe A, Hu C, Munoz RM, Von Hoff DD, Han H, 2012, Down-regulation of yes associated protein 1 expression reduces cell proliferation and clonogenicity of pancreatic cancer cells. PLoS One 7(3):e32783 Yuan M, Tomlinson V, Lara R, Holliday D, Chelala C, Harada T, Gangeswaran R, Manson-Bishop C, Smith P, Danovi SA, Pardo O, Crook T, Mein CA, Lemoine NR, Jones LJ, Basu S, 2008, Yesassociated protein, YAP, functions as a tumor suppressor in breast. Cell Death Differ 15:1752–1759 Ma S, Huang J, Xie Y, Yi N, 2012, Identification of breast cancer prognosis markers using integrative sparse boosting. Methods Inf Med 51(2):152–61 Stenmark H, 2009, Rab GTPases as coordinators of vesicle traffic. Nature Rev Mol Cell Biol 10:513–525 Mosesson Y, Mills GB, Yarden Y, 2008, Derailed endocytosis: an emerging feature of cancer. Nature Rev Cancer 8(11):835–850 Mountjoy JR, Xu W, McLeod D, Hyndman D, Oko R, 2008, RAB2A: a major subacrosomal protein of bovine spermatozoa implicated in acrosomal biogenesis. Biol Reprod 79(2):223–32 WangX, Trotman LC, TKoppie T,Alimonti A, Chen Z, Gao Z,Wang J, Erdjument-Bromage H, Tempst P, Cordon Cardo C, Pandolfi PP, Jiang X, 2007, NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell 128(1):129–39 AmodioN, ScrimaM, Palaia L, SalmanAN,Quintiero A, FrancoR, Botti G, Pirozzi P, Rocco G, De Rosa N, Viglietto G, 2010, Oncogenic role of the E3 ubiquitin ligase NEDD4–1, a PTEN negative regulator, in non-small-cell lung carcinomas. Am J Pathol 177(5):2622–34 Wong KA, Wilson J, Russo A, Wang L, Okur MN, Wang X, Martin NP, Scappini E, Carnegie GK, O’Bryan JP, 2012, Intersectin, ITSN, family of scaffolds function as molecular hubs in protein interaction networks. PLoS One 7(4):e36023 Kondo Y, Koshimizu E, Megarbane A, Hamanoue H, Okada I, Nishiyama K, Kodera H, Miyatake S, Tsurusaki Y, Nakashima M, Doi H, Miyake N, Saitsu H, Matsumoto N, 2013, Whole-exome sequencing identified a homozygous FNBP4 mutation in a family with a condition similar to microphthalmia with limb anomalies. Am J Med Genet A 161A(7):1543–6 Zhang G, Wang Z, 2011, MAGI1 inhibits cancer cell migration and invasion of hepatocellular carcinoma via regulating PTEN. Zhong Nan Da Xue Xue Bao Yi Xue Ban 36(5):381–5 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 293 EP 300 DI 10.1007/s12253-014-9820-8 PG 8 ER PT J AU Tang, JY Hsi, E Huang, YCh Hsu, ChHN Yang, WCh Chang, HW Chai, ChY Chu, PY AF Tang, Jen-Yang Hsi, Edward Huang, Ya-Chun Hsu, Chung-Heng Nicholas Yang, Wen-Chi Chang, Hsueh-Wei Chai, Chee-Yin Chu, Pei-Yi TI Overexpression of Autophagy-Related 16-Like 1 in Patients with Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Autophagy; Oral cancer; Immunohistochemistry; Autophagy-related 16-like 1 ID Autophagy; Oral cancer; Immunohistochemistry; Autophagy-related 16-like 1 AB Dyregulation of autophagy has been reported in various human cancers including oral squamous cell carcinoma (OSCC). The objective of this study was to link expression of autophagy-related 16-like 1 (ATG16L1), a protein essential for autophagosome formation, to clinical outcome in a cohort of 90 OSCC patients. Expression level of ATG16L1 was assessed by immunohistochemistry and an immunoreactivity score (IRS), ranging from 0 to 9, was assigned to each case. The results were correlated with clinicopathological parameters and outcome of patients. Twenty-seven patients (30 %) exhibited ATG16L1 overexpression as indicated by an IRS of 9. Overexpression of ATG16L1 was significantly associated with disease stage (p=0.001), size (p=0.031) of the tumor, lymph node metastasis (p=0.004), and histological grade (p=0.038). ATG16L1 overexpression significantly affected the overall survival (p=0.020) and time to recurrence (p=0.031) of OSCC patients in Kaplan- Meier analysis. The present study suggested that ATG16L1 may be used as a biomarker for selecting OSCC patients with a more aggressive phenotype. C1 [Tang, Jen-Yang] Kaohsiung Medical University, Faculty of Medicine, College of Medicine, Department of Radiation OncologyKaohsiung, Taiwan, Republic of China. [Hsi, Edward] Kaohsiung Medical University, College of Medicine, Graduate Institute of Clinical MedicineKaohsiung, Taiwan, Republic of China. [Huang, Ya-Chun] Kaohsiung Medical University, College of Medicine, Department of Pathology, No. 100, Tzyou 1st RoadKaohsiung, Taiwan, Republic of China. [Hsu, Chung-Heng Nicholas] Kaohsiung Medical University, College of Medicine, Graduate Institute of Clinical MedicineKaohsiung, Taiwan, Republic of China. [Yang, Wen-Chi] Kaohsiung Medical University, College of Medicine, Graduate Institute of Clinical MedicineKaohsiung, Taiwan, Republic of China. [Chang, Hsueh-Wei] Kaohsiung Medical University, College of Medicine, Research Center for Environmental MedicineKaohsiung, Taiwan, Republic of China. [Chai, Chee-Yin] Kaohsiung Medical University, College of Medicine, Department of Pathology, No. 100, Tzyou 1st RoadKaohsiung, Taiwan, Republic of China. [Chu, Pei-Yi] St. Martin De Porres Hospital, Department of Pathology, No. 565, Sec. 2, Daya RoadNew Taipei City, Taiwan, Republic of China. RP Chai, ChY (reprint author), Kaohsiung Medical University, College of Medicine, Department of Pathology, Kaohsiung, Taiwan, Republic of China. EM cychai@kmu.edu.tw CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90., DOI 10.3322/ caac.20107 Ranadive KJ, Ranadive SN, Shivapurkar NM, Gothoskar SV, 1979, Betel quid chewing and oral cancer: experimental studies on hamsters. International Journal of Cancer Journal International du Cancer 24(6):835–843 Mashberg A, Boffetta P, Winkelman R, Garfinkel L, 1993, Tobacco smoking, alcohol drinking, and cancer of the oral cavity and oropharynx among U.S. veterans. Cancer 72(4):1369–1375 Pindborg JJ, Reibel J, Roed-Peterson B, Mehta FS, 1980, Tobaccoinduced changes in oral leukoplakic epithelium. Cancer 45(9):2330– 2336 Paterson IC, Eveson JW, Prime SS, 1996, Molecular changes in oral cancer may reflect aetiology and ethnic origin. Eur J Cancer B Oral Oncol 32B(3):150–153 Park JY, Muscat JE, Ren Q, Schantz SP, Harwick RD, Stern JC, Pike V, Richie JP Jr, Lazarus P, 1997, CYP1A1 and GSTM1 polymorphisms and oral cancer risk. Cancer Epidemiol Biomarkers Prev: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology 6(10): 791–797 Elwood JM, Pearson JC, Skippen DH, Jackson SM, 1984, Alcohol, smoking, social and occupational factors in the aetiology of cancer of the oral cavity, pharynx and larynx. International Journal of Cancer Journal International du Cancer 34(5):603–612 Klionsky DJ, Emr SD, 2000, Autophagy as a regulated pathway of cellular degradation. Science 290(5497):1717–1721 Mizushima N, 2007, Autophagy: process and function. Genes Dev 21(22):2861–2873., DOI 10.1101/gad.1599207 MathewR,Karantza-WadsworthV,White E, 2007, Role of autophagy in cancer. Nat Rev Cancer 7(12):961–967., DOI 10.1038/nrc2254 Mizushima N, Noda T, Ohsumi Y, 1999, Apg16p is required for the function of the Apg12p-Apg5p conjugate in the yeast autophagy pathway. EMBO J 18(14):3888–3896., DOI 10.1093/emboj/18.14. 3888 Nomura H, Uzawa K, Yamano Y, Fushimi K, Ishigami T, Kouzu Y, Koike H, Siiba M, Bukawa H, Yokoe H, Kubosawa H, Tanzawa H, 2009, Overexpression and altered subcellular localization of autophagy-related 16-like 1 in human oral squamous-cell carcinoma: correlation with lymphovascular invasion and lymph-node metastasis. Hum Pathol 40(1):83–91., DOI 10.1016/j.humpath.2008. 06.018 Hippert MM, O’Toole PS, Thorburn A, 2006, Autophagy in cancer: good, bad, or both? Cancer Res 66(19):9349–9351., DOI 10.1158/ 0008-5472.CAN-06-1597 Kimmelman AC, 2011, The dynamic nature of autophagy in cancer. Genes Dev 25(19):1999–2010., DOI 10.1101/gad.17558811 Tang JY, Hsi E, Huang YC, Hsu NC, Chu PY, Chai CY, 2013, High LC3 expression correlates with poor survival in patients with oral squamous cell carcinoma. Hum Pathol 44(11):2558–2562., DOI 10. 1016/j.humpath.2013.06.017 Wu WK, Coffelt SB, Cho CH, Wang XJ, Lee CW, Chan FK, Yu J, Sung JJ, 2012, The autophagic paradox in cancer therapy. Oncogene 31(8):939–953., DOI 10.1038/onc.2011.295 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 301 EP 305 DI 10.1007/s12253-014-9821-7 PG 5 ER PT J AU Mostowska, A Sajdak, S Pawlik, P Markowska, J Pawalowska, M Lianeri, M Jagodzinski, PP AF Mostowska, Adrianna Sajdak, Stefan Pawlik, Piotr Markowska, Janina Pawalowska, Monika Lianeri, Margarita Jagodzinski, P Pawel TI Replication Study for the Association of Seven Genome- Gwas-Identified Loci With Susceptibility to Ovarian Cancer in the Polish Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ovarian cancer; Single nucleotide polymorphisms; Genome-wide association studies ID Ovarian cancer; Single nucleotide polymorphisms; Genome-wide association studies AB We investigated the previously-demonstrated association of seven genome-wide association studies (GWAS) single nucleotide polymorphisms (SNPs), including rs2072590 (HOXD-AS1), rs2665390 (TIPARP), rs10088218 and rs10098821 (8q24), rs3814113 (9p22), rs9303542 (SKAP1) and rs2363956 (ANKLE1), as risk factors of epithelial ovarian tumors (EOTs). These SNPs were genotyped in two hundred seventy three patients with EOTs and four hundred sixty four unrelated healthy females from the Polish population. We observed the lowest p values of the trend test for the 9p22 rs3814113 and 8q24 rs10098821 SNPs in patients with all subtypes of ovarian cancer (ptrend=0.010 and ptrend=0.014, respectively). There were also significant p values for the trend of the 9p22 rs3814113 and the 8q24 rs10098821 SNPs for serous histological subtypes of ovarian cancer (ptrend=0.006, ptrend=0.033, respectively). Moreover, stratification of the patients based on their histological type of cancer demonstrated, in the dominant hereditary model, a significant association of the 9p22 rs3814113 SNP with serous ovarian carcinoma OR=0.532 (95 % CI=0.342 - 0.827, p=0.005, pcorr=0.035). Despite the relatively small sample size of cases and controls, our studies confirmed some of the previously-demonstrated GWAS SNPs as genetic risk factors for EOTs. C1 [Mostowska, Adrianna] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. [Sajdak, Stefan] Poznan University of Medical Sciences, Clinic of Gynecological SurgeryPoznan, Poland. [Pawlik, Piotr] Poznan University of Medical Sciences, Clinic of Gynecological SurgeryPoznan, Poland. [Markowska, Janina] Poznan University of Medical Sciences, Chair of Gynecologic OncologyPoznan, Poland. [Pawalowska, Monika] Poznan University of Medical Sciences, Chair of Gynecologic OncologyPoznan, Poland. [Lianeri, Margarita] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. [Jagodzinski, P Pawel] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. RP Jagodzinski, PP (reprint author), Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 60-781 Poznan, Poland. EM pjagodzi@am.poznan.pl CR Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics, 2012. CA Cancer J Clin 62:10–29 Siegel R, Naishadham D, Jemal A, Jemal A, Siegel R, Ward E, Hao Y, Xu J et al, 2008, Cancer statistics. CA Cancer J Clin 58:71–96 Gurung A, Hung T, Morin J et al, 2013, Molecular abnormalities in ovarian carcinoma: clinical, morphological and therapeutic correlates. Histopathology 62:59–70 Danforth KN, Tworoger SS, Hecht JL et al, 2007, Breastfeeding and risk of ovarian cancer in two prospective cohorts. Cancer Causes Control 18:517–523 Whittemore AS,Harris R, Itnyre J et al, 1992, Characteristics relating to ovarian cancer risk: collaborative analysis of 12 US case–control studies. I. Methods. Collaborative Ovarian Cancer Group. Am J Epidemiol 136:1175–1183 Cibula D,WidschwendterM,Majek O et al, 2011, Tubal ligation and the risk of ovarian cancer: review and meta-analysis. Hum Reprod Update 17:55–67 Havrilesky LJ, Moorman PG, Lowery WJ et al, 2013, Oral contraceptive pills as primary prevention for ovarian cancer: a systematic review and meta-analysis. Obstet Gynecol 122:139–147 TsilidisKK,Allen NE,Key TJ et al, 2011, Oral contraceptive use and reproductive factors and risk of ovarian cancer in the European Prospective Investigation into Cancer and Nutrition. Br J Cancer 105:1436–1442 Lacey JV Jr, Mink PJ, Lubin JH et al, 2002, Menopausal hormone replacement therapy and risk of ovarian cancer. JAMA 288:334–341 Trabert B, Wentzensen N, Yang HP et al, 2012, Ovarian cancer and menopausal hormone therapy in the NIH-AARP diet and health study. Br J Cancer 25:1181–1187 Pieta B, Chmaj-Wierzchowska K, Opala T, 2012, Past obstetric history and risk of ovarian cancer. Ann Agric Environ Med 19: 385–388 Bodelon C, Wentzensen N, Schonfeld SJ et al, 2013, Hormonal risk factors and invasive epithelial ovarian cancer risk by parity. Br J Cancer., DOI 10.1038/bjc.2013.344 Salehi F, Dunfield L, Phillips KP et al, 2008, Risk factors for ovarian cancer: an overview with emphasis on hormonal factors. J Toxicol Environ Health B Crit Rev 11:301–321 Sayasneh A, Tsivos D, Crawford R, 2011, Endometriosis and ovarian cancer: a systematic review. ISRN Obstet Gynecol 2011:140310 LinHW, Tu YY, Lin SYet al, 2011, Risk of ovarian cancer in women with pelvic inflammatory disease: a population-based study. Lancet Oncol 12:900–904 Garland CF,Mohr SB, Gorham ED et al, 2006, Role of ultraviolet B irradiance and vitamin D in prevention of ovarian cancer. Am J Prev Med 31:512–514 Lichtenstein P, Holm NV, Verkasalo PK et al, 2000, Environmental and heritable factors in the causation of cancer—analyses of cohorts of twins fromSweden, Denmark and Finland. N Engl J Med 343:78– 85 Stratton JF, Pharoah P, Smith SK et al, 1998, A systematic review and meta-analysis of family history and risk of ovarian cancer. Br J Obstet Gynaecol 105:493–499 Berchuck A, Schildkraut JM, Marks JR et al, 1999, Managing hereditary ovarian cancer risk. Cancer 86:2517–2524 Frank TS, Manley SA, Olopade OI et al, 1998, Sequence analysis of BRCA1 and BRCA2: correlation of mutations with family history and ovarian cancer risk. J Clin Oncol 16:2417– 2425 Holschneider CH, Berek JS, 2000, Ovarian cancer: epidemiology, biology, and prognostic factors. Semin Surg Oncol 19:3–10 Song H, Ramus SJ, Tyrer J et al, 2009, A genome-wide association study identifies a new ovarian cancer susceptibility locus on 9p22.2. Nat Genet 41:996–1000 Goode EL, Chenevix-Trench G, Song H et al, 2010, A genome-wide association study identifies susceptibility loci for ovarian cancer at 2q31 and 8q24. Nat Genet 42:874–879 Couch FJ, Gaudet MM, Antoniou AC et al, 2010, Common variants at 19p13 are associated with susceptibility to ovarian cancer. Nat Genet 42:880–884 Hahn LW, RitchieMD, Moore JH, 2003)Multifactor dimensionality reduction software for detecting gene-gene and gene-environment interactions. Bioinformatics 19:376–382 Ramus SJ, Kartsonaki C, Gayther SA et al, 2011, Consortium of Investigators of Modifiers of BRCA1/2.Genetic variation at 9p22.2 and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 103:105–116 Shen J, Wang D, Gregory SR et al, 2012, Evaluation of microRNA expression profiles and their associations with risk alleles in lymphoblastoid cell lines of familial ovarian cancer. Carcinogenesis 33:604–612 Zhao H, Shen J, Wang D et al, 2012, Associations between gene expression variations and ovarian cancer risk alleles identified from genome wide association studies. PLoS One 7:e47962 Ghoussaini M, Song H, Koessler T et al, 2008, Multiple loci with different cancer specificities within the 8q24 gene desert. J Natl Cancer Inst 100:962–966 Gong WF, Zhong JH, Xiang BD et al, 2013, Single nucleotide polymorphism 8q24 rs13281615 and risk of breast cancer: metaanalysis of more than 100,000 cases. PLoS One 8:e60108 Grisanzio C, Freedman ML, 2010, Chromosome 8q24-associated cancers and MYC. Genes Cancer 1:555–559 Hawken SJ, Greenwood CM, Hudson TJ et al, 2010, The utility andpredictive value of combinations of low penetrance genes forscreening and risk prediction of colorectal cancer. Hum Genet 128:89–101 Park JH,Wacholder S, GailMH et al, 2010, Estimation of effect size distribution from genome-wide association studies an dimplicationsfor future discoveries. NatGenet 42:570–575 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 307 EP 313 DI 10.1007/s12253-014-9822-6 PG 7 ER PT J AU Dedic Plavetic, N Jakic-Razumovic, J Kulic, A Sirotkovic-Skerlev, M Baric, M Vrbanec, D AF Dedic Plavetic, Natalija Jakic-Razumovic, Jasminka Kulic, Ana Sirotkovic-Skerlev, Maja Baric, Marina Vrbanec, Damir TI Prognostic Value of Ki-67 in Breast Carcinoma: Tissue Microarray Method Versus Whole Section Analysis- Potentials and Pitfalls SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ki-67; Breast cancer; Proliferation; Prognostic factor; Tissue microarrays ID Ki-67; Breast cancer; Proliferation; Prognostic factor; Tissue microarrays AB In our study we have compared the prognostic value of two distinct methods of immunohistochemical Ki- 67 determination, tissue microarray (TMA) and classical whole section analysis. "Cut-off" values were used according to the 2009 St. Gallen Consensus. Tissue specimens were obtained from a consecutive retrospective series of 215 female patients with primary invasive tumours. Two hundred and thirteen patients were included in the study. Data on Ki-67 was collected by both tissue microarray (TMA) and whole section analysis. Follow up data on overall (OS) and diseasefree survival (DFS) were collected. Median follow-up was 95 months (range from 7.8 through 107 months). Mutual correlation of two Ki-67 determination methods was nonsignificant (Person’s r=0.13417; p=0.0528). There was statistically significant association of whole section Ki-67 expression with histological and nuclear grade, progesterone receptor and HER2/neu status. The expression of Ki-67 protein in TMAs correlated only with histological and nuclear grade, but not with other traditional clinicopathological factors. Statistically significant differences in DFS (p=0.0156) and OS (p=0.0028) were confirmed between subgroups with low and high whole section Ki-67 expression. When subgroups with high and intermediate expression were compared, significant difference was found in DFS (p=0.0272), but not in OS (p=0.0624). On the other hand, there was no statistically significant difference either in DFS, or in OS, according to the expression of Ki-67 in TMAs (p=0.6529; p= 0.7883; p=0.7966 for DFS, and p=0.8917; p=0.6448; p=0.4323 for OS, respectively). In our study, classical whole section was superior to TMA analysis in terms of prognosis and clinicopathological correlation. Our results indicate that the method used may have impact on prognostic significance of Ki-67. Further studies are needed, covering a greater number of patients and including a precisely defined stage and treatment patient cohorts, in order to solve controversies in Ki-67 assessment methodology. C1 [Dedic Plavetic, Natalija] University Hospital Center Zagreb and Zagreb Medical School, Department of Medical Oncology, Kispaticeva 12, 10 000 Zagreb, Croatia. [Jakic-Razumovic, Jasminka] University Hospital Center Zagreb, Department of PathologyZagreb, Croatia. [Kulic, Ana] University Hospital Center Zagreb and Zagreb Medical School, Department of PathophysiologyZagreb, Croatia. [Sirotkovic-Skerlev, Maja] University Hospital Center Zagreb and Zagreb Medical School, Department of PathophysiologyZagreb, Croatia. [Baric, Marina] University Hospital Center Zagreb and Zagreb Medical School, Department of Medical Oncology, Kispaticeva 12, 10 000 Zagreb, Croatia. [Vrbanec, Damir] University Hospital Center Zagreb and Zagreb Medical School, Department of Medical Oncology, Kispaticeva 12, 10 000 Zagreb, Croatia. RP Dedic Plavetic, N (reprint author), University Hospital Center Zagreb and Zagreb Medical School, Department of Medical Oncology, 10 000 Zagreb, Croatia. EM natalija.dedic-plavetic@zg.t-com.hr CR Gerdes J, Schwab U, Lemke H, Stein H, 1983, Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer 31:13–20 Bullwinkel J, Baron-Luhr B, Ludemann A,Wohlenberg C, Gerdes J, Scholze T, 2006, Ki67 protein is associated with ribosomal RNA transcription in quiescent and proliferating cells. J Cell Physiol 206: 624–635 Rahmanzadeh R, Huttmann G, Gerdes J, Scholzen T, 2007, Chromophore-assisted light inactivation of pKi67 leads to inhibition of ribosomal RNA synthesis. Cell Prolif 40:422–430 Scholzen T, Gerdes J, 2000, The Ki- 67 protein: from the known and the unknown. J Cell Physiol 182:311–322 Brown DC, Gatter KC, 2000, Ki67 protein: the immaculate deception? Histopathology 40:2–11 Lopez F, Belloc F, Lacombe F et al, 1991)Modalities of synthesis of Ki67 antigen during the stimulation of lymphocytes. Cytometry 12: 2–49 Cattoretti G, Becker MH, Key G et al, 1992, Monoclonal antibodies against recombinant parts of the Ki-67 antigen, MIB 1 and MIB 3, detect proliferating cells in microwave-processed formalin-fixed paraffin sections. J Pathol 168:357–363 Veronese SM, Maisano C, Scibilia J, 1995, Comparative prognostic value of Ki-67 and MIB-1 proliferation indices in breast cancer. Anticancer Res 15(6B):2717–2722 Urruticoechea A, Smith IE, Dowsett M, 2005, Proliferation marker Ki-67 in early breast cancer. J Clin Oncol 23:7212–7220 Shoker BS, Jarvis C, Clarke RB et al, 1999, estrogen receptorpositive proliferative cells in normal and precancerous breast. Am J Pathol 155:1811–1815 Okumura Y, Yamamoto Y, Zhang Z et al, 2008, Identification of biomarkers in ductal carcinoma in situ of the breast with microinvasion. BMC Cancer 8:287 Wintzer HO, Zipfel I, Schulte-Monting J et al, 1991, Ki-67 immunostaining in human breast tumours and its relationships to prognosis. Cancer 67:421–428 Spyratos F, Ferrero-Pous M, Trassard M et al, 2002, Correlation betweenMIB-1 and other proliferation markers: clinical implications of the MIB-1 cutoff value. Cancer 94:2151–2159 Rudolph P, Olsson H, BonatzG et al, 1999, Correlation between p53, c-erbB-2, and topoisomerase II alpha expression, DNA ploidy, hormonal receptor status and proliferation in 356 node-negative breast carcinomas: prognostic implications. J Pathol 187:207–216 Trihia H, Murray S, Price K et al, 2003, Ki-67 expression in breast carcinoma: its association with grading systems, clinical parameters, and other prognostic factors – a surrogate marker? Cancer 97:1321– 1331 Tan PH, Bay BH, Yip G, Selvarajan S, Tan P, Wu J, Lee CH, Li KB, 2005, Immunohistochemical detection of Ki67 in breast cancer correlates with transcriptional regulation of genes related to apoptosis and cell death. Mod Pathol 18(3):374–381 Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA, 2010, Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol 11(2):174–183 Wiesner FG, Magener A, Fasching PA, Wesse J, Bani MR, Rauh C, Jud S, Schrauder M, Loehberg CR, Beckmann MW, Hartmann A, Lux MP, 2009, Ki-67 as a prognostic molecular marker in routine clinical use in breast cancer patients. Breast 18:135–141 Neri A,Marrelli D, Pedrazzani C, Caruso S,De StefanoA,Mariani F, Megha T, De Marco G, Corso G, Pinto E, Roviello F, 2008, Prognostic relevance of proliferative activity evaluated by Mib-1 immunostaining in node negative breast cancer. EJSO 34:1299–1303 Goldhirsch A, Ingle JN, Gelber RD, Coates AS, Thurlimann B, 2009, Senn HJ & Panel members 2009 thresholds for therapies: highlights of the St Gallen International expert consensus on the primary therapy of early breast cancer. Ann Oncol 20:1319–1329 Elston CW, Ellis IO, 1987, Pathological prognostic factors in breast cancer. I. the value of histological grade in long- term prognosis of breast cancer: a study of 1010 patients, uniformly treated at the institute Gustave-Roussy. J Clin Oncol 5:1378–1386 Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19: 403–410 Carreno G, del Casar JM, Corte D et al, 2007, Local recurrence after mastectomy for breast cancer: analysis of clinicopathological, biological and prognostic characteristics. Breast Cancer Res Treat 102: 61–73 Ruiz C, Seibt S, Al Kuraya K et al, 2006, Tissue microarrays for comparing molecular features with proliferation activity in breast cancer. Int J Cancer 118:2190–2194 DowsettM, Nielsen TO, A’Hern R et al, 2011, Assessment of ki67 in breast cancer. J Natl Cancer Inst 103(22):1656–1664 Honma N, Horii R, Iwase T et al, 2013, Ki-67 evaluation at the hottest spot predicts clinical outcome of patients with hormone receptor- positive/HER2-negative breast cancer treated with adjuvant tamoxifen monotherapy. Breast Cancer., DOI 10.1007/s12282-013- 0455-5 Wersto RP, Liblit RL, Deitch D, Koss LG, 1991, Variability in DNA measurements in multiple tumour samples of human colonic carcinoma. Cancer 67:106–115 GerlingerM, Rowan AJ, Horswell S et al, 2012, Intratumour heterogeneity and branched evolution revealed by multiregion sequencing. NEJM 366:883–892 Jones RL, Salter J, A’Hern R et al, 2010, Relationship between oestrogen receptor status and proliferation in predicting response and long-term outcome to neoadjuvant chemotherapy for breast cancer. Breast Cancer Res Treat 119(2):315–323 Fasanella S, Leonardi E, Cantaneli C et al, 2011, Proliferative activity in human breast cancer: Ki-67 automated evaluation and the influence of different Ki-67 equivalent antibodies. Diagn Pathol 6(Suppl 1):S7 Mohammed ZMA, McMillan DC, Elsberger B et al, 2012, Comparison of visual and automated assessment of Ki-67 proliferative activity and their impact on outcome in primary operable invasive ductal breast cancer. Br J Cancer 106:383–388 Cheang MC, Chia SK, Voduc D, Gao D, Leung S, Snider J, 2009, Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 101:736–750 Stuart-Harris R, Caldas C, Pinder SE, Pharoah P, 2008, Proliferation markers and survival in early breast cancer: a systematic review and meta-analysis of 85 studies in 32,825 patients. Breast 17:323–334 de Azambuja E, Cardoso F, de Castro JG et al, 2007, Ki-67 as prognostic marker in early breast cancer: ameta-analysis of published studies involving 12,155 patients. Br J Cancer 96:1504–1513 Colozza M, Azambuja E, Cardoso F, Sotiriou C, Larsimont D, Piccart MJ, 2005, Proliferative markers as prognostic and predictive tools in early breast cancer: where are we now? Ann Oncol 16:1723–1739 CuzickJ DM, Pineda S et al, 2011, Prognostic value of a combined estrogen receptor, progesterone receptor, ki-67, and human epidermal growth factor receptor 2 immunohistochemical score and comparison with the genomic health recurrence score in early breast cancer. J Clin Oncol 29:4273–4278 Ishihara M, Mukai H, Nagai S et al, 2013, Retrospective analysis of risk factors for central nervous system metastases in operable breast cancer: effects of biologic subtype and ki67 overexpression on survival. Oncology 84:135–140 Penault-Llorca F, Andre F, Sagan C et al, 2009, Ki67 expression and docetaxel efficacy in patients with estrogen receptor–positive breast cancer. J Clin Oncol 27:2809–2815 Viale G, ReganMM, MastropasquaMGet al, 2008, Predictive value of tumour ki-67 expression in two randomized trials of adjuvant chemoendocrine therapy for node-negative breast cancer. J Natl Cancer Inst 100:207–212 Viale G, Giobbie-Hurder A, Regan MM et al, 2008, Prognostic and predictive value of centrally reviewed Ki-67 labeling index in postmenopausal women with endocrine responsive breast cancer. results from breast international group trial 1–98 comparing adjuvant tamoxifen with letrozole. J Clin Oncol 26:5569–5575 Dowsett M, Dunbier AK, 2008, Emerging biomarkers and new understanding of traditional markers in personalized therapy for breast cancer. Clin Cancer Res 14:8019–8026 DowsettM, Smith IE, Ebbs SR et al, 2007, Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer. J Natl Cancer Inst 99(2):167–170 Rimm DL, Camp RL, Charette LA et al, 2001, Tissue microarray: a new technology for amplification of tissue resources. Cancer J 7:24– 31 Eerola H, Heikkila P, Tamminen A, Aittomaki K, Blomqvist C, Nevanlinna H, 2005, Relationship of patients’ age to histopathological features of breast tumours in BRCA1 and BRCA2 and mutationnegative breast cancer families. Breast Cancer Res 7:R465–R469 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 315 EP 324 DI 10.1007/s12253-014-9823-5 PG 10 ER PT J AU Han, BE Chang, YB Jung, SY Kim, YS AF Han, Byeol Eun Chang, Yoon Bo Jung, Suk Young Kim, Yeon Sung TI Lantana camara Induces Apoptosis by Bcl-2 Family and Caspases Activation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Apoptosis; Bcl-2 family; Breast cancer; Caspase; Lantana camara ID Apoptosis; Bcl-2 family; Breast cancer; Caspase; Lantana camara AB Breast cancer is one of the most common cancers worldwide, and the second most fatal cancer in women after lung cancer. Because there are instances of cancer resistance to existing therapies, studies focused on the identification of novel therapeutic drugs are very important. In this study, we identified a natural anticancer agent from Lantana camara, a flowering plant species of the genus Verbena. The extract obtained from the L. camara exhibited cell death properties in the human breast cancer cell line,MCF-7.We found that the apoptosis induced by treatment with the L. camara extract was regulated by the Bcl-2 family. Bid and Bax was increased and Bcl-2 was decreased by L. camara extract. L. camara extract modulated cleavage of caspase-8, and caspase-9, as well as poly (ADP-ribose) polymerase (PARP). Our results support the potential use of the L. camara extract as an anti-breast cancer drug. C1 [Han, Byeol Eun] Wonkwang University, College of Pharmacy, Institute of Pharmaceutical Research and Development, 460 Ikandae-ro, 570-749 Iksan, Jeonbuk, South Korea. [Chang, Yoon Bo] Wonkwang University, College of Pharmacy, Institute of Pharmaceutical Research and Development, 460 Ikandae-ro, 570-749 Iksan, Jeonbuk, South Korea. [Jung, Suk Young] Pusan National University, College of PharmacyBusan, South Korea. [Kim, Yeon Sung] Wonkwang University, College of Pharmacy, Institute of Pharmaceutical Research and Development, 460 Ikandae-ro, 570-749 Iksan, Jeonbuk, South Korea. RP Kim, YS (reprint author), Wonkwang University, College of Pharmacy, Institute of Pharmaceutical Research and Development, 570-749 Iksan, South Korea. EM sungykim@wonkwang.ac.kr CR Adams JM, Cory S, 2007, The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene 26(9):1324–1337., DOI 10.1038/ sj.onc.1210220 Almubarak H, Jones A, Chaisuparat R, Zhang M, Meiller TF, Scheper MA, 2011, Zoledronic acid directly suppresses cell proliferation and induces apoptosis in highly tumorigenic prostate and breast cancers. J of carcinog 10:2., DOI 10.4103/1477-3163.75723 Bentzon N, During M, Rasmussen BB, Mouridsen H, Kroman N, 2008, Prognostic effect of estrogen receptor status across age in primary breast cancer. Int j of cancer J int du cancer 122(5):1089– 1094., DOI 10.1002/ijc.22892 Chen YH, Yeh CW, Lo HC, Su SL, Hseu YC, Hsu LS, 2012, Generation of reactive oxygen species mediates butein-induced apoptosis in neuroblastoma cells. Oncol Rep 27(4):1233–1237., DOI 10. 3892/or.2012.1632 Cory S, Huang DC, Adams JM(2003, The Bcl-2 family: roles in cell survival and oncogenesis. Oncogene 22(53):8590–8607., DOI 10. 1038/sj.onc.1207102 Darzynkiewicz Z, Bruno S, Del Bino G, Gorczyca W, Hotz MA, Lassota P, Traganos F, 1992, Features of apoptotic cells measured by flow cytometry. Cytometry 13(8):795–808., DOI 10.1002/cyto. 990130802 Deena MJ, Thoppil JE, 2000, Antimicrobial activity of the essential oil of Lantana camara. Fitoterapia 71(4):453–455 Dunnwald LK, Rossing MA, Li CI, 2007, Hormone receptor status, tumor characteristics, and prognosis: a prospective cohort of breast cancer patients. Breast cancer res : BCR 9(1):R6., DOI 10.1186/ bcr1639 Fan TJ, Han LH, Cong RS, Liang J, 2005, Caspase family proteases and apoptosis. Acta Biochim Biophys Sin 37(11):719–727 Fulda S, 2010, Modulation of apoptosis by natural products for cancer therapy. Planta Med 76(11):1075–1079., DOI 10.1055/s-0030- 1249961 Ghisalberti EL, 2000, Lantana camara L., Verbenaceae). Fitoterapia 71(5):467–486 Ghobrial IM, Witzig TE, Adjei AA, 2005, Targeting apoptosis pathways in cancer therapy. CA: a cancer j for clin 55(3):178–194 Heeres JT, Hergenrother PJ, 2007, Poly(ADP-ribose, makes a date with death. Curr Opin Chem Biol 11(6):644–653., DOI 10.1016/j. cbpa.2007.08.038 Jonville MC, Kodja H, Humeau L, Fournel J, De Mol P, Cao M, Angenot L, Frederich M, 2008, Screening of medicinal plants from Reunion Island for antimalarial and cytotoxic activity. J Ethnopharmacol 120(3):382–386., DOI 10.1016/j.jep.2008.09.005 Kasibhatla S, Tseng B, 2003, Why target apoptosis in cancer treatment? Mol Cancer Ther 2(6):573–580 Kaufmann SH, Desnoyers S, Ottaviano Y, Davidson NE, Poirier GG, 1993, Specific proteolytic cleavage of poly(ADP-ribose, polymerase: an early marker of chemotherapy-induced apoptosis. Cancer Res 53(17):3976–3985 Kumar VP, Chauhan NS, Padh H, Rajani M, 2006, Search for antibacterial and antifungal agents from selected Indian medicinal plants. J Ethnopharmacol 107(2):182–188., DOI 10.1016/j.jep.2006.03.013 Nayak BS, Raju SS, Eversley M, Ramsubhag A, 2009, Evaluation of wound healing activity of Lantana camara L. - a preclinical study. Phytother res : PTR 23(2):241–245., DOI 10.1002/ptr.2599 Nguewa PA, Fuertes MA, Alonso C, Perez JM, 2003, Pharmacological modulation of Poly(ADP-ribose, polymerasemediated cell death: exploitation in cancer chemotherapy. Mol Pharmacol 64(5):1007–1014., DOI 10.1124/mol.64.5.1007 Nita ME, Nagawa H, Tominaga O, Tsuno N, Fujii S, Sasaki S, Fu CG, Takenoue T, Tsuruo T, Muto T, 1998, 5-Fluorouracil induces apoptosis in human colon cancer cell lines with modulation of Bcl-2 family proteins. Br J Cancer 78(8):986–992 Niu M, Sun Y, Liu X, Tang L, Qiu R, 2013, Tautomycetin induces apoptosis by inactivating Akt through a PP1-independent signaling pathway in human breast cancer cells. J Pharmacol Sci 121(1):17–24 Oliver FJ, de la Rubia G, Rolli V, Ruiz-Ruiz MC, de Murcia G, Murcia JM, 1998, Importance of poly(ADP-ribose, polymerase and its cleavage in apoptosis. Lesson from an uncleavable mutant. The J of biol chem 273(50):33533–33539 Partridge AH, Burstein HJ, Winer EP, 2001, Side effects of chemotherapy and combined chemohormonal therapy in women with earlystage breast cancer. J Natl Cancer Inst Monogr 30:135–142 Shapiro CL, Recht A, 2001, Side effects of adjuvant treatment of breast cancer. N Engl J Med 344(26):1997–2008., DOI 10.1056/ NEJM200106283442607 Sharma M, Sharma PD, Bansal MP, 2008, Lantadenes and their esters as potential antitumor agents. J Nat Prod 71(7):1222–1227., DOI 10.1021/np800167x Sharma M, Sharma PD, Bansal MP, Singh J, 2007, Synthesis, cytotoxicity, and antitumor activity of lantadene-A congeners. Chem & biodivers 4(5):932–939., DOI 10.1002/cbdv.200790082 Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics, 2012. CA: a cancer j for clin 62(1):10–29., DOI 10.3322/caac.20138 Sun SN, Jia WD, Chen H, Ma JL, Ge YS, Yu JH, Li JS, 2013, Docosahexaenoic acid, DHA, induces apoptosis in human hepatocellular carcinoma cells. Int j of clin and exp pathol 6(2):281–289 Tempfer CB, Froese G, Heinze G, Bentz EK, Hefler LA, Huber JC, 2009, Side effects of phytoestrogens: a meta-analysis of randomized trials. The Am j of med 122(10):939–946., DOI 10.1016/j.amjmed. 2009.04.018 Tsujimoto Y, 1998, Role of Bcl-2 family proteins in apoptosis: apoptosomes or mitochondria? Genes to cells : devoted to mol & cell mech 3(11):697–707 Weis J, Poppelreuter M, Bartsch HH, 2009, Cognitive deficits as long-term side-effects of adjuvant therapy in breast cancer patients: ‘subjective’ complaints and ‘objective’ neuropsychological test results. Psycho-Oncology 18(7):775–782., DOI 10.1002/ pon.1472 Yang L, Wu D, Luo K, Wu S, Wu P, 2009, Andrographolide enhances 5-fluorouracil-induced apoptosis via caspase-8-dependent mitochondrial pathway involving p53 participation in hepatocellular carcinoma, SMMC-7721, cells. Cancer Lett 276(2):180–188., DOI 10. 1016/j.canlet.2008.11.015 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 325 EP 331 DI 10.1007/s12253-014-9824-4 PG 7 ER PT J AU Cincin, BZ Unlu, M Kiran, B Bireller, SE Baran, Y Cakmakoglu, B AF Cincin, Birsu Zeynep Unlu, Miray Kiran, Bayram Bireller, Sinem Elif Baran, Yusuf Cakmakoglu, Bedia TI Apoptotic Effects of Quercitrin on DLD-1 Colon Cancer Cell Line SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon Cancer; Quercitrin; Antiproliferative; Apoptosis ID Colon Cancer; Quercitrin; Antiproliferative; Apoptosis AB Quercetin, which is the most abundant bioflavonoid compound, is mainly present in the glycoside form of quercitrin. Although different studies indicated that quercitrin is a potent antioxidant, the action of this compound is not well understood. In this study, we investigated whether quercitrin has apoptotic and antiproliferative effects in DLD-1 colon cancer cell lines. Time and dose dependent antiproliferative and apoptotic effects of quercitrin were subsequently determined by WST-1 cell proliferation assay, lactate dehydrogenase (LDH) cytotoxicity assay, detection of nucleosome enrichment factor, changes in caspase-3 activity, loss of mitochondrial membrane potential (MMP) and also the localization of phosphatidylserine (PS) in the plasma membrane. There were significant increases in caspase-3 activity, loss of MMP, and increases in the apoptotic cell population in response to quercitrin in DLD-1 colon cancer cells in a time- and dose-dependent manner. These results revealed that quercitrin has antiproliferative and apoptotic effects on colon cancer cells. Quercitrin activity supported with in vivo analyses could be a biomarker candicate for early colorectal carcinoma. C1 [Cincin, Birsu Zeynep] Istanbul University, Institute of Experimental Medical Research, CapaIstanbul, Turkey. [Unlu, Miray] Izmir Institute of Technology, Science Faculty, Department of Molecular Biology and Genetics, Urla, 35430 Izmir, Turkey. [Kiran, Bayram] Kastamonu University, Department of BiologyKastamonu, Turkey. [Bireller, Sinem Elif] Istanbul University, Institute of Experimental Medical Research, CapaIstanbul, Turkey. [Baran, Yusuf] Izmir Institute of Technology, Science Faculty, Department of Molecular Biology and Genetics, Urla, 35430 Izmir, Turkey. [Cakmakoglu, Bedia] Istanbul University, Institute of Experimental Medical Research, CapaIstanbul, Turkey. RP Cakmakoglu, B (reprint author), Istanbul University, Institute of Experimental Medical Research, Istanbul, Turkey. EM bedia@istanbul.edu.tr CR Jemal A, Bray F, CenterMMet al, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90 Akao Y, Nakagawa Y, Naoe T, 2006, MicroRNAs 143 and 145 are possible common onco-microRNAs in human cancers. Oncol Rep 16:845–850 Valeri N, Gasparini P, FabbriMet al, 2010)Modulation of mismatch repair and genomic stability by miR-155. Proc Natl Acad Sci U S A 107:6982–6987 Wattenberg LW, 1985, Chemoprevention of cancer. Cancer Res 45: 1–8 Boone CW, Kelloff GJ, Malone WE, 1990, Identification of candidate cancer chemopreventive agents and theirevaluation in animal models and human clinical trials: a review. Cancer Res 50:2–9 Reddy BS, 2000, The Fourth DeWitt S. Goodman lecture. Novel approaches to the prevention of colon cancer bynutritional manipulation and chemoprevention. CancerEpidemiol. Biomark Prev 9: 239–247 Cody V, 1988, Crystal and molecular structures of flavonoids. Prog Clin Biol Res 280:29–44 Pietta PG, 2000, Flavonoids as antioxidants. J Nat Prod 63: 1035–1042 Tripoli E, GuardiaML, Giammanco S et al, 2007, Citrus flavonoids: molecular structure, biological activity and nutritional properties: a review. Food Chem 104:466–479 Pilsakova L, Riecansky I, Jagla F, 2010, The physiological actions of isoflavone phytoestrogens. Physiol Res 59:651–664 Domitrovic R, 2011, The molecular basis for the pharmacological activity of anthocyans. Curr Med Chem 18:4454–4469 Tanaka T, Takahashi R, 2013, Flavonoids and asthma.Nutrition 5(6): 2128–43 Ding M, Zhao J, Bowman L et al, 2010, Inhibition of AP-1 and MAPK signaling and activation of Nrf2/ARE pathway by quercitrin. Int J Oncol 36(1):59–67 Sa’nchez de Medina F, Vera B, Ga’lvez J et al, 2002, Effect of quercitrin on the early stages of hapten induced colonic inflammation in the rat. Life Sci 70:3097–3108 Gee JM, DuPont MS, Rhodes MJ et al, 1998, Quercetin glucosides interact with the intestinal glucose transport pathway. Free Radic Biol Med 25:19–25 Wagner C, Fachinetto R, Dalla Corte CL et al, 2006, Quercitrin, a glycoside form of quercetin, prevents lipid peroxidation in vitro. Brain Res 1107:192–198 Cruz EA, Da-Silva SA, Muzitano MF et al, 2008, Immunomodulatory pretreatment with Kalanchoe pinnata extract and its quercitrin flavonoid effectively protects mice against fatal anaphylactic shock. Int Immunopharmacol 8(12): 1616–21 Yang HM. Ham YM. Yoon WJ. Roh SW. Jeon YJ. et al., 2012, Quercitrin protects against ultraviolet B-induced cell death in vitro and in an in vivo zebrafish model. J. Photochem. Photobiol. B Sep 3;114:126–31 Camuesco D, Comalada M, Rodrguez-Cabezas ME et al, 2004, The intestinal anti-inflammatory effect of quercitrin is associated with an inhibition in iNOS expression. Br J Pharmacol 143: 908–918 Comalada M, Camuesco D, Sierra S et al, 2005, In vivo quercitrin anti-inflammatory effect involves release of quercetin, which inhibits inflammation through down-regulation of the NF-kappaB pathway. Eur J Immunol 35(2):584–92 Sanchez deMedina F, Vera B, Galvez J, Zarzuelo A, 2002, Effect of quercitrin on theearly stages of hapten induced colonic inflammation in the rat. Life Sci 70(26):3097–108 Browning AM, Walle UK, Walle T, 2005, Flavonoid glycosides inhibit oral cancer cell proliferation–role of cellular uptake and hydrolysis to the aglycones. J Pharm Pharmacol 57, 8):1037–42 Xu R, Zhang Y, Ye X, Xue S, Shi J et al, 2013, Inhibition effects and induction of apoptosis of flavonoids on the prostate cancer cell line PC-3 in vitro. Food Chem 138(1):48–53 Ly JD, Grubb DR, Lawen A, 2003, The mitochondrial membrane potential, deltapsi(m))in apoptosis; an update. Apoptosis 8(2): 115–28 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 333 EP 338 DI 10.1007/s12253-014-9825-3 PG 6 ER PT J AU Zidi, S Stayoussef, M Zouidi, F Benali, S Gazouani, E Mezlini, A Yacoubi-Loueslati, B AF Zidi, Sabrina Stayoussef, Mouna Zouidi, Ferjeni Benali, Samir Gazouani, Ezzedine Mezlini, Amel Yacoubi-Loueslati, Besma TI Tumor Necrosis Factor Alpha (−238 / −308) and TNFRII-VNTR (−322) Polymorphisms as Genetic Biomarkers of Susceptibility to Develop Cervical Cancer Among Tunisians SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tumor necrosis factor; Polymorphisms; Cervical cancer; Tunisians; Haplotypes ID Tumor necrosis factor; Polymorphisms; Cervical cancer; Tunisians; Haplotypes AB Host genetic factors may confer susceptibility to Cervical Cancer. TNF-α as pro-inflammatory cytokine participates in the maintenance of immune homeostasis. Allelic variation of immuno-modulatory genes is associated with alteration in immune function. This study investigated the associations between TNF-α-308G>A, −238G>A, and TNFRII - VNTR-322 and cervical cancer in Tunisian women. Genotypes of those polymorphisms were detected in 130 cases and 260 controls. The variant heterozygote −308 G/A was associated with a 41 % decreased risk of cervical cancer (GG vs A/A; p=0.002; OR = 0.41; 95 % CI =0.23–0.76). Furthermore, compared with dominant variant G/G, the (G/A+A/A) genotypes was significantly associated with a decreased risk of CC (GG vs G/A+A/A; p=0.026; OR = 0.62; 95 % CI = 0.40–0.97). The FIGO stratified analysis showed that the minor variant A/A and combined G/A+A/A of TNFα-238 G>A and TNFα-308 G>A increased the risk of the tumor evolution, respectively, (P=0.011; OR = 2.98; 95% CI = 1.16–7.72) (P=0.008; OR = 2.76; 95%CI = 1.20–6.41), (P=0.000; OR = 16.33; 95 % CI = (5.10–55.23) (P=0.000; OR = 7.54; 95 % CI = 2.68–22.29). There was statistically significant relationship between the incidence of the TNF-α mutations and the clinical progression of cancer according to the FIGO classification. In our study, the haploview analysis revealed no LD between rs1800629 and rs361525. TNF-α and TNFRII polymorphisms might be genetic risk factors for cervical cancer in Tunisian population C1 [Zidi, Sabrina] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 2092 El MANAR I, 1092 Tunis, Tunisia. [Stayoussef, Mouna] University of Monastir, Faculty of Pharmacy, Research Unit of Hematological and Autoimmune DiseasesMonastir, Tunisia. [Zouidi, Ferjeni] Habib Bourguiba University, Hospital of Sfax, Immunology DepartmentSfax, Tunisia. [Benali, Samir] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 2092 El MANAR I, 1092 Tunis, Tunisia. [Gazouani, Ezzedine] Military Hospital of Tunis, Laboratory of ImmunologyTunis, Tunisia. [Mezlini, Amel] Salah Azeiz Oncology InstituteTunis, Tunisia. [Yacoubi-Loueslati, Besma] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 2092 El MANAR I, 1092 Tunis, Tunisia. RP Zidi, S (reprint author), El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 1092 Tunis, Tunisia. EM ZIDISABRINA86@gmail.com CR AhmedMI, Salahy EE, Fayed STet al, 2001, Human papillomavirus infection among Egyptian females with cervical carcinoma: relationship to spontaneous apoptosis and TNF-alpha. Clin Biochem 34(6): 491–498 Allendoerfer R, MageeDM, Smith JGet al, 1993, Induction of tumor necrosis factor-α in murine Candida albicans infection. Oxf J Med J Infect Dis 167(5):1168–1172 Ardizzoia A, Lissoni P, Brivio F et al, 1992, Tumor necrosis factor in solid tumors: increased blood levels in the metastatic disease. J Biol Regul Homeost Agents 6(3):103–107 Asghar T, Yoshida S, Kennedy S et al, 2004, The tumor necrosis factor-apromoter 1031C polymorphism is associated with decreased risk of endometriosis in a Japanese population. Hum Reprod 19: 2509–2514 Aukrust P, Sandberg WJ, Otterdal K et al, 2011, Tumor necrosis factor superfamily molecules in acute coronary syndromes. Ann Med 43:90–103 Badano I, Stietz SM, Schurr TG et al, 2012, Analysis of TNFalpha promoter SNPs and the risk of cervical cancer in urban populations of Posadas, Misiones, Argentina). J Clin Virol 53:54–59 Balkwill F, Mantovani A, 2001, Inflammation and cancer: back to Virchow. Lancet 17:539–545 Barbisan G, Perez LO, Contreras A et al, 2012, TNF-alpha and IL-10 promoter polymorphisms, HPV infection, and cervical cancer risk. Tumour Biol 33:1549–1556 Bazzoni F, Beutler B, 1996, The tumor necrosis factor ligand and receptor families. N Engl J Med 334:1717 Bernard HU, Burk RD, Chen Z et al, 2010, Classification of papillomaviruses, PVs, based on 189 PV types and proposal of taxonomic amendments. Virology 401:70–79 Burkly LC, Michaelson JS, Zheng TS, 2011, TWEAK/Fn14 pathway: an immunological switch for shaping tissue responses. Immunol Rev 244:99–114 Calhoun ES, Mcgovern RM, Janney CA et al, 2002, Host genetic polymorphism analysis in cervical cancer. Clin Chem 48(8):1218– 1224 Chopra V, Dinh TV, Hannigan EV, 1998, Circulating serum levels of cytokines and angiogenic factors in patients with cervical cancer. Cancer Investig 16:152–159 Craveiro R, Costa S, Pinto D et al, 2004, TP73 alterations in cervical carcinoma. Cancer Genet Cytogenet 150:116–121 De Freitas AC, Gurgel AP, Chagas BS et al, 2012, Susceptibility to cervical cancer: an overview. Gynecol Oncol 126:304–311 De Giovine FS, Camp NJ, Cox A et al, 2000, Detection and population analysis of IL-1 and 430 TNF gene polymorphisms in cytokine molecular biology. Oxford University, Oxford, pp 21–46 Deshpande A, Nolan JP, White PS et al, 2005, TNF-alpha promoter polymorphisms and susceptibility to human papillomavirus 16- associated cervical cancer. J Infect Dis 191(6):969–976 Ding B, Shilong F, Meilin Wet al, 2012, Tumor Necrosis Factor α −308 G>A polymorphisms and cervical cancer risk: a meta-analysis. Int J Gynecol Cancer 22(2):213–219 Dranoff G, 2004, Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer 4:11–22 Duarte I, Santos A, Sousa H et al, 2005, G-308A TNF-alpha polymorphism is associated with an increased risk of invasive cervical cancer. Biochem Biophys Res Commun 334:588–592 Fong CL, Siddiqui AH, Mark DF, 1994, Identification and characterization of a novel repressor site in the human tumor necrosis factor ogene. Nucleic Acids Res 22(6):1108–1114 Gostout BS, Poland GA, Calhoun ES et al, 2003, TAP1, TAP2, and HLA-DR2 alleles are predictors of cervical cancer risk. Gynecol Oncol 88:326–323 Govan VA, Constant D, Hoffman M et al, 2006, The allelic distribution of −308 tumor necrosis factor-alpha gene polymorphism in south African women with cervical cancer and control women. BMC Cancer 6:24 Hallermalm K, Seki K,Wei CH et al, 2001, Tumor necrosis factor–a induces coordinated changes in major histocompatibility class I presentation pathway, resulting in increased stability of class I complexes at the cell surface. Blood 98:1108–1115 Ibeanu OA, 2011)Molecular pathogenesis of cervical cancer. Cancer Biol Ther 11:295–306 Jacob CO, Fronek Z, Lewis GD, Koo M, Hansen JA, McDevitt HO, 1990, Heritable major histocompatibility complex class II-associated differences in production of tumor necrosis factor a: prevalence to genetic predisposition to systemic lupus erythematosus. Proc Natl Acad Sci USA 87:1233 Jang WH, Yang YI, Yea SS et al, 2001, The −238 tumor necrosis factor-alpha promoter polymorphism is associated with decreased susceptibility to cancers. Cancer Lett 166(1):41–46 Jemal A, Bray F, CenterMMet al, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Keen L, Wood N, Olomolaiye O et al, 1999, A bi-allelic VNTR in the human TNFR2, p75, gene promoter. Genes Immunol 1:164 Kohaar I, Thakur N, Salhan S et al, 2007, TNFalpha-308 G/A polymorphism as a risk factor for HPVassociated cervical cancer in Indian population. Cell Oncol 29:249–256 Liu L, Yang X, Chen X et al, 2012, Association between TNF-α polymorphisms and cervical cancer risk: a meta-analysis. Mol Biol Rep 39:2683–2689 Ljunggren G, Anderson DJ, 1998, Cytokine-induced modulation of MHC class-I and class-II molecules on human cervical epithelial cells. J Reprod Immunol 38:123–138 Mekinian A, Tamouza R, Pavy S et al, 2011, Functional study of TNF-alpha promoter polymorphisms: literature review and metaanalysis. Eur Cytokine Netw 22:88–102 Naylor MS, Stamp GW, Foulkes WD et al, 1993, Tumor necrosis factor and its receptors in human ovarian cancer. Potential role in disease progression. J Clin Invest 91:2194–2206 Pan F, Tian J, Ji C-S et al, 2012, Association of TNF-α-308 and-238 polymorphisms with risk of cervical cancer: a meta-analysis. Asian Pac J Cancer Prev 13(11):5777–5783 Patel R, Cafardi JM, Patel N et al, 2011, Tumor necrosis factor biologics beyond psoriasis in dermatology. Expert Opin Biol Ther 11:1341–1359 Perrey C, Turner SJ, Pravica V et al, 1999, ARMS-PCR methodologies to determine IL-10, TNF-α, TNF-β and TGF-β1 gene polymorphisms. Transpl Immunol 7:127–128 Qidwai T, Khan F, 2011, Tumour necrosis factor gene polymorphism and disease prevalence. Scand J Immunol 74:522–547 Santos AM, Sousa H, Catarino R et al, 2005, TP53 codon 72 polymorphism and risk for cervical cancer in Portugal. Cancer Genet Cytogenet 159:143–147 Shepperd JH, 1995, FIGO staging of gynecologic cancers; cervical and vulva. Int J Gynecol Cancer 5:319–323 Shishodia S, Majumdar S, Banerjee S et al, 2003, Ursolic acid inhibits nuclear factor-κB activation induced by carcinogenic agents through suppression of IκBα kinase and p65 phosporylation: Correlation with down-regulation of cyclooxygenase 2, matrix metalloproteinase 9, and cyclin D1. Cancer Res 63:4375–4383 Stanczuk GA, Sibanda EN, Tswana SA et al, 2003, Polymorphism at the −308-promoter position of the tumor necrosis factoralpha, TNFalpha, gene and cervical cancer. Int J Gynecol Cancer 13:148–153 Steinshamn S, Bemelmans MH, Van Tits LJ et al, 1996, TNF receptors in murine Candida Albicans infection: evidence for an important role of TNF receptor p55 in antifungal defence. J Immunol 157:2155 ThomasM, Narayan N, Pim D et al, 2008, Human papillomaviruses, cervical cancer and cell polarity. Oncogene 27:7018–7030 Upasna G, Aggarwal BB, 2003, Regulation of proliferation, survival and apoptosis by members of the TNF superfamily. Biochem Pharmacol 66(8):1403–1408 Van Heel DA, Udalova IA, De Silva AP et al, 2002, Inflammatory bowel disease is associated with a TNF polymorphism that affects an interaction between the OCT1 and NF-κB transcription factors. Hum Mol Genet 11:1281–1289 Wang N, Yin D, Zhang S et al, 2012, TNFalpha rs1800629 polymorphism is not associated with HPV infection or cervical cancer in the Chinese population. PLoS One 7:e45246 Wang SS, BrattiMC, Rodriguez AC et al, 2009, Common variants in immune and DNA repair genes and risk for human papillomavirus persistence and progression to cervical cancer. J InfectDis 199:20–30 Wilson AG, De Giovine FS, Duff GW, 1995, Genetics of TNF-α in autoimmune, infectious and neoplastic diseases. J Inflamm 45:1 Wilson AG, Symons JA, McDowell TL et al, 1997, Effects of a polymorphism in the human tumor necrosis factor a promoter on transcriptional activation. Proc Natl Acad Sci U S A 94:3195–3199 Winkles JA, 2008, The TWEAK-Fn14 cytokine-receptor axis: discovery, biology and therapeutic targeting. Nat Rev Drug Discov 7:411–425 Zhang H-L, Zhang Y-J, 2013, A systemic assessment of the association between tumor necrosis factor alpha 308 G/A polymorphism and risk of cervical cancer. Tumor Biol 34(3):1659–1665 Zidi S, Verdi H, Yilmaz-Yalcin Y et al, 2014, Impact of Toll-Like Receptors 2/3/4/9, IL-1-a/b and TNF-a Polymorphisms in Cervical Cancer Susceptibility in Tunisia Pathol Oncol Res, DOI 10.1007/ s12253-014-9793-7. Zuo F, Liang W, Ouyang Yet al, 2010, Association of TNF-α gene promoterpoly-morphisms with susceptibility of cervical cancer in Southwest China. Lab Med 42:287–290 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 339 EP 345 DI 10.1007/s12253-014-9826-2 PG 7 ER PT J AU Mise, PB Telesmanic, DV Tomic, S Sundov, D Capkun, V Vrdoljak, E AF Mise, Petric Branka Telesmanic, Dobric Vesna Tomic, Snjezana Sundov, Dinka Capkun, Vesna Vrdoljak, Eduard TI Correlation Between E-cadherin Immunoexpression and Efficacy of First Line Platinum-Based Chemotherapy in Advanced High Grade Serous Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ovarian cancer; E-cadherin; Immunohistochemistry; Response to platinum-based chemotherapy; Platinum sensitivity; Prognosis ID Ovarian cancer; E-cadherin; Immunohistochemistry; Response to platinum-based chemotherapy; Platinum sensitivity; Prognosis AB To analyze correlation between immunoexpression of E-cadherin and efficacy of first line platinum-based chemotherapy in patients with advanced-stage high-grade serous ovarian carcinoma. The expression of E-cadherin was analyzed immunohistochemically in formalin-fixed, paraffinembedded samples from 98 patients with advanced-stage high-grade serous ovarian cancer and related to clinical features (stage according to the International Federation of Gynecology and Obstetrics (FIGO) and residual tumors after initial cytoreductive surgery), response to platinum-based chemotherapy (according to Response Evaluation Criteria in Solid tumors (RECIST 1.1 criteria)), platinum sensitivity (according to platinum free interval (PFI) as platinum-refractory, platinum-resistant and platinum-sensitive) and patients progression free survival (PFS) and overall survival (OS). Ecadherin immunostaining was positive in 74 and negative in 24 serous ovarian carcinomas. E-cadherin immunoreactivity was not associated with FIGO stage, residual tumor after initial cytoreductive surgery and number of chemotherapy cycles. Positive E-cadherin expression predict significantly better response to first line platinum-based chemotherapy (p<0.001) and platinum sensitivity (p<0.001). Moreover, positive E-cadherin expression predict significantly longer PFS (p<0.001) and OS (p<0.001). The multivariate analysis for OS showed that positive E-cadherin expression is predictor to platinum sensitivity (p<0.001) and longer OS (p=0.01). Positive E-cadherin expression seems to be a predictor of better response to first line platinum-based chemotherapy, platinum sensitivity and favorable clinical outcome in patients with advanced-stage serous ovarian cancer. Negative Ecadherin expression was shown to be significant, independent predictor of poorer PFS and OS. E-cadherin as a marker has predictive and prognostic value. C1 [Mise, Petric Branka] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Oncology, Spinciceva 1, 21000 Split, Croatia. [Telesmanic, Dobric Vesna] General Hospital Zadar, Department of Oncology, Boze Pericica 5, 23000 Zadar, Croatia. [Tomic, Snjezana] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Pathology, Forensic Medicine and Cytology, Spinciceva 1, 21000 Split, Croatia. [Sundov, Dinka] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Pathology, Forensic Medicine and Cytology, Spinciceva 1, 21000 Split, Croatia. [Capkun, Vesna] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Nuclear Medicine, Spinciceva 1, 21000 Split, Croatia. [Vrdoljak, Eduard] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Oncology, Spinciceva 1, 21000 Split, Croatia. RP Mise, PB (reprint author), University of Split, School of Medicine, Clinical Hospital Center Split, Department of Oncology, 21000 Split, Croatia. EM brapemi@gmail.com CR Ferlay J, Soerjomataram I, Ervik M et al. GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11 [Internet]. Lyon, France: International Agency for Research on Cancer; 2013. Available from: http://globocan.iarc.fr. Accessed on 26/May/2014 Carter JS, Downs LS Jr, 2011, Ovarian cancer test and treatment. Female Patient, Parsippany, 36:30–35 SEER Cancer Statistics Factsheets: Ovary Cancer. National Cancer Institute. Bethesda, MD,http://seer.cancer.gov/statfacts/html/ovary. html, http://seer.cancer.gov/statfacts/html/ovary.html). Date of access: 26/May/2014 Gilks CB, Prat J, 2009, Ovarian carcinoma pathology and genetics: resent advances. Hum Pathol 40:1213–1223., DOI 10.1016/j.humpath. 2009.04.017 Helleman J, van Staveren IL, Dinjens WN et al, 2006, Mismatch repair and treatment resistance in ovarian cancer.BMC Cancer 6:201., DOI 10.1186/1471-2407-6-201 Kim A, Ueda Y, Naka T et al, 2012, Therapeutic strategies in epithelial ovarian cancer. J Exp Clin Cancer Res 31:14., DOI 10. 1186/1756-9966-31-14 Bagnato A, Rosano L, 2012, Understanding and overcoming chemoresistance in ovarian cancer: emerging role of the endothelin axis. Curr Oncol 19:36–38., DOI 10.3747/co.19.895 Bast RC Jr, Hennessy B, Mills GB, 2009, The biology of ovarian cancer: newopportunities for translation. Nat Rev Cancer 9:415–428., DOI 10.1038/nrc2644 Martin LP, Hamilton TC, Schilder RJ et al, 2008, Platinum resistance: the role of DNA repair pathways. Clin Cancer Res 14:1291– 1295., DOI 10.1158/1078-0432.CCR-07-2238 Stewart DJ, 2007, Mechanisms of resistance to cisplatin and carboplatin. Crit Rev Oncol Hematol 63:12–31., DOI 10.1016/j. critrevonc.2007.02.001 Rosano L, Cianfrocca R, Spinella F, 2011, Acquisition of chemoresistance and EMT phenotype is linked with activation of the endothelin A receptor pathway in ovarian carcinoma cells. Clin Cancer Res 17:2350–2360., DOI 10.1158/1078-0432.CCR-10-2325 Baribeau S, Chaudhry P, Parent S et al, 2014, Resveratrol inhibits cisplatin-induced epithelial to mesenchymal transition in ovarian cancer cells lines. PLoS One 9:e86987., DOI 10.1371/journal.pone. 0086987 Pecina-Slaus N, 2003, Tumor suppressor gene E-cadherin and its role in normal andmalignant cells. Cancer Cell Int 3:17., DOI 10.1186/ 1475-2867-3-17 PutzkeAP,VenturaAP, BaileyAMet al, 2011, Metastatic progression of prostate cancer and E-cadherin regulation by zeb1 and SRC family kinases. Am J Pathol 179:400–410., DOI 10.1016/j.ajpath.2011.03.028 Luber B, Deplazes J, Keller G et al, 2011, Biomarker analysis of cetuximab plus oxaliplatin/leucovorin/5-fluorouracil in first-line metastatic gastric and oesophago-gastric junction cancer: results from a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie, AIO). BMC Cancer 11:509., DOI 10.1186/1471-2407-11-509 Dong HM, Liu G, Hou YF et al, 2007, Dominant negative Ecadherin inhibits the invasiveness of inflammatory breast cancer cells in vitro. J Cancer Res Clin Oncol 133:83–92., DOI 10.1007/s00432- 006-0140-6 Ye Y, Tellez JD, Durazo M et al, 2010, E-cadherin accumulation within the lymphovascular embolus of inflammatory breast cancer due to altered trafficking. Anticancer Res 30:3903–3910 Bacic B, Haller H,Mrklic I et al, 2013, Prognostic role of E-cadherin in patients with advanced serous ovarian cancer. Arch Gynecol Obstet 287:1219–1224., DOI 10.1007/s00404-012-2684-9 Kurman RJ, Shih IeM, 2010, The origin and pathogenesis of epithelial ovarian cancer: a proposed uniflying theory. AmJ Surg Pathol 34: 433–443., DOI 10.1097/PAS.0b013e3181cf3d79 Singer G, Oldt R 3rd, Cohen Yet al, 2003, Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst 95:484–486., DOI 10.1093/jnci/95.6.484 Kurman RJ, VisvanathanK, Roden R et al, 2008, Early detection and treatment of ovarian cancer: shifting from early stage to minimal volume of disease based on a new model of carcinogenesis. Am J Obstet Gynecol 198:351–356., DOI 10.1016/j.ajog.2008.01.005 Cho KR, Shih IeM, 2009, Ovarian cancer. Annu Rev Pathol 4:287– 313., DOI 10.1146/annurev.pathol.4.110807.092246 Prat J, FIGO Committee on Gynecologic Oncology, 2014, Staging classification for cancer of the ovary, fallopian tube, and peritoneum. Int J Gynaecol Obstet 124:1–5., DOI 10.1016/j.ijgo.2013.10.001 Malpica A, Deavers MT, Lu K et al, 2004, Grading ovarian serous carcinoma using a two-tier system. Am J Surg Pathol 28:496–504 Wimberger P, Wehling M, Lehmann N et al, 2010, Influence of residual tumor on outcome in ovarian cancer patients with FIGO stage IV disease: an explratory analysis of the AGO-OVAR, Arbeitsgemeinschaft Gynaekologische Onkologie Ovarian Cancer Study Group). Ann Surg Oncol 17:1642–1648., DOI 10.1245/s10434- 010-0964-9 Eisenhauer EA, Therasse P, Bogaerts J et al, 2009, New response evaluation criteria in solid tumours: revised RECIST guideline, version 1.1). Eur J Cancer 45:228–247., DOI 10.1016/j.ejca.2008.10.026 Peng HL, He L, Zhao X, 2012, Association of reduced immunohistochemical expression of E-cadherin with a poor ovarian cancer prognosis – results of meta-analysis. Asian Pac J Cancer Prev 13: 2003–2007., DOI 10.7314/APJCP.2012.13.5.2003 Blechschmidt K, Sassen S, Schmalfeldt B et al, 2008, The Ecadherin repressor Snail is associated with lower overall survival of ovarian cancer patients. Br J Cancer 98:489–495., DOI 10.1038/sj.bjc. 6604115 Cho EY, Choi Y, Chae SWet al, 2006, Immunohistochemical study of the expression of adhesionmolecules in ovarian serous neoplasms. Pathol Int 56:62–70., DOI 10.1111/j.1440-1827.2006.01925.x Darai E, Scoazec JY, Walker-Combrouze F et al, 1997, Expression of cadherins in benign, borderline, and malignant ovarian epithelial tumors: a clinicopathologic study of 60 cases. Hum Pathol 28:922–928 Dian D, Bruning A, Mylonas I, 2011, E-cadherin as prognostic marker in human serous carcinomas of the ovary: an immunohistichemical analysis. Arch Gynecol Obstet 284:437–443., DOI 10.1007/s00404-010-1657-0 Faleiro-Rodrigues C, Macedo-Pinto I, Pereira D et al, 2004, Prognostic value of E-cadherin immunoexpression in patients with primary ovarian carcinomas. Ann Oncol 15:1535–1542., DOI 10. 1093/annonc/mdh387 Ho CM, Cheng WF, Lin MC et al, 2010, Prognostic and predictive values of E-cadherin for patients of ovarian clear cell adenocarcinoma. Int J Gynecol Cancer 20:1490–1497., DOI 10.1111/IGC. 0b013e3181e68a4d Bodnar L, Stanczak A, Cierniak S et al, 2014, WNT/β-catenin pathway as a potential prognostic and predictive marker in patients with advanced ovarian cancer. J Ovar Res 7:16., DOI 10.1186/1757- 2215-7-16 Ozols RF, Bundy BN, Greer BE et al, 2003, Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol 21:3194–3200., DOI 10.1200/JCO.2003.02.153 du Bois A, Luck HJ, MeierWet al, 2003, A randomised clinical trial of cisplatin/paclitaxel versus carboplatin/paclitacel as first-line treatment of ovarian cancer. J Natl Cancer Inst 95:1320–1329., DOI 10. 1093/jnci/djg036 McGuire WP, Hoskins WJ, Brady MF et al, 1996, Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IVovarian cancer. N Engl J Med 334:1–6., DOI 10.1056/NEMJ199601043340101 Cardoso F, Piccart-Gebhart M, Van’t Veer L et al, 2007, The MINDAC trial: the first prospective clinical validation of a genomic tool. Mol Oncol 1:246–251., DOI 10.1016/jmolonc.2007.10.004 Chang JC, Makris A, Gutierrez MC et al, 2008, Gene expression patterns in formalin-fixed, paraffin-embedded core biopsies predict docetaxel chemosensitivity in brest cancer patients. Breast Cancer Res Treat 108:233–240., DOI 10.1007/s10549-007-9590-2 Garber K(2010, ALK, lung cancer, and personalized therapy: portent of the future? J Natl Cancer Inst 102:672–675., DOI 10.1093/jnci/ djq184 Lin CC, Yang LC, 2011, Optimal management with patients with non-small-cell lung cancer and epidermal growth receptor mutations 71:79–88., DOI 10.2165/11587560-000000000-00000 Berx G, van Roy F, 2009, Involvement of members of cadherin superfamily in cancer. Cold Spring Harb Perspect Biol 1:a003129., DOI 10.1101/cshperspect.a003129 Rodriguez FJ, Lewis-Tuffin L, Anastasiadis PZ, 2012, E-cadherin’s dark side: possible role in tumor progression. Biochim Biophys Acta 1826:23–31., DOI 10.1016/j.bbcan.2012.03.002 Koo JS, JungW, Jeong J, 2009, The predictive role of E-cadherin and androgen receptor on in vitro chemosensitivity in triple negative breast cancer. Jpn J ClinOncol 39:560–568., DOI 10.1093/jjco/hyp065 Fricke E, Hermannstadter C, Keller G et al, 2004, Effect of wild-type and mutant E-cadherin on cell proliferation and responsiveness to the chemotherapeutic agents cisplatin, etoposide and 5-fluorouracile. Oncology 66:105–109., DOI 10.1159/000077442 Hofmann G, Balic M, Dandachi N et al, 2013, The predictive value of serum soluble E-cadherin levels in breast cancer patients undergoing preoperative systemic chemotherapy. Clin Biochem 46:1585– 1589., DOI 10.1016/j.clinbiochem.2013.06.010 Nakamoto K, Nagahara H, Maeda K et al, 2013, Expression of Ecadherin and KRASmutationmay serve as biomarkers in cetuximabbased therapy in metastatic colorectal cancer. Oncol Lett 5:1295– 1300., DOI 10.3892/ol.2013.1187 Witta SE, Gemmill RM, Hirsch FR et al, 2006, Restoring E-cadherin expression increases sensitivity to epidermal growth factor receptor inhibitors in lung cancer cell lines. Cancer Res 66:944–950., DOI 10. 1158/0008-5472.CAN-05-1988 Ravi V, Yang J, Araujo DM et al., 2011, The role of E-cadherin expression in response and outcome in patients with leiomyosarcoma treated with chemotherapy. J Clin Oncol 29 suppl:abstr 10096 Graziano F, Mandolesi A, Ruzzo A et al, 2004, Predictive and prognostic role of E-cadherin protein expression in patients with advanced gastric carcinomas treated with palliative chemotherapy. Tumour Biol 25:106–110 Spreafico A, Gregorc V, Dziadziuszko R et al., 2007, Serum Ecadherin does not associate with response to gefitinib in patients with non-small cell lung cancer, NSCLC): P3-088. J Thorac Oncol 2: pS716., DOI 10.1097/01.JTO.0000284064.64727.66 Shim HS, Yoon BS, Cho NH, 2009, Prognostic significance of paired epithelial cell adhesion molecule and E-cadherin in ovarian serous carcinoma. Hum Pathol 40:693–698., DOI 10.1016/j.humpath. 2008.10.013 Voutilainen KA, Anttila MA, Sillanpaa SM et al, 2006, Prognostic significance od E-cadherin-catenin complex in epithelial ovarian cancer. J Clin Pathol 59:460–467., DOI 10.1136/jcp.2005.029876 Huang KJ, Sui LH, 2012, The relevance and role of vascular endothelial growth factor C, matrix metalloproteinase-2 and E-cadherin in epithelial ovarian cancer. Med Oncol 29:318–323., DOI 10.1007/ s12032-010-9817-4 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 347 EP 356 DI 10.1007/s12253-014-9827-1 PG 10 ER PT J AU Coban, I Cakir, A Unal, DKT Bassullu, N Karpuz, V Dogusoy, BG Alper, M AF Coban, Ipek Cakir, Asli Unal, Dilay Kokenek Tuba Bassullu, Nuray Karpuz, Vildan Dogusoy, Bulbul Gulen Alper, Murat TI Emerin Expression in Well Differentiated Epithelial Lesions of Thyroid: Implications in Papillary Thyroid Carcinoma Diagnosis and Predicting Malignant Behavior SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Emerin; Thyroid; Nucleus; Membrane ID Emerin; Thyroid; Nucleus; Membrane AB Recently, it has been reported that identifying nuclear membrane irregularities with anti-emerin antibody is useful for papillary thyroid carcinoma diagnosis. However, literature regarding the significance of emerin immunohistochemistry in thyroid is limited. We evaluated the diagnostic accuracy of the well-established nuclear alterations, nuclear protrusions and recently described nuclear shapes (garlands and star-like shapes) with emerin immunohistochemistry and hematoxylin- eosin stain in thyroid lesions. We further evaluated the diagnostic accuracy measures of tissue microarrays evaluated with both stains, to detect whether emerin immunohistochemistry improves the diagnostic accuracy for papillary thyroid carcinoma. For papillary thyroid carcinoma, pseudo- inclusions were best performers with emerin (diagnostic accuracy: 0.91), whereas with hematoxylin- eosin diagnostic accuracy of grooves was the highest (0.92). For follicular variant of papillary thyroid carcinoma, with both stains, predominately oval nuclear shape had the best diagnostic performance (diagnostic accuracy: 0.95). Nuclear protrusions were poor identifiers for papillary thyroid carcinoma. However, with emerin immunohistochemistry, they could successfully identify malignancy in 83 % of the cases. Using emerin immunohistochemistry, in addition to hematoxylin- eosin improved the diagnostic accuracy for papillary thyroid carcinoma when compared to hematoxylin- eosin evaluation only (sensitivity: 0.70 vs 0.86, negative predictive value: 0.81 vs. 0.94, diagnostic accuracy: 0.87 vs. 0.94). Consistent with the previous literature, our findings indicate that emerin immunohistochemistry may be used as an adjunct diagnostic method to identify papillary thyroid carcinoma. Additionally, we suggest that nuclear protrusions detected with emerin imunohistochemistry may be used as indicators of malignant behavior in small tissue samples of thyroid. C1 [Coban, Ipek] Istanbul Bilim University, School of Medicine, Department of Pathology, Cemil Aslan Guder Sok. No: 8 Gayrettepe, 30342 Istanbul, Turkey. [Cakir, Asli] Medipol University School of Medicine, Department of PathologyIstanbul, Turkey. [Unal, Dilay Kokenek Tuba] Diskapi Yildirim Beyazit Education and Research Hospital, Department of PathologyAnkara, Turkey. [Bassullu, Nuray] Istanbul Bilim University, School of Medicine, Department of PathologyIstanbul, Turkey. [Karpuz, Vildan] Istanbul Bilim University, School of Medicine, Department of PathologyIstanbul, Turkey. [Dogusoy, Bulbul Gulen] Istanbul Bilim University, School of Medicine, Department of PathologyIstanbul, Turkey. [Alper, Murat] Diskapi Yildirim Beyazit Education and Research Hospital, Department of PathologyAnkara, Turkey. RP Coban, I (reprint author), Istanbul Bilim University, School of Medicine, Department of Pathology, 30342 Istanbul, Turkey. EM ipekco@gmail.com;ipek.coban@florence.com.tr CR Clary KM, Condel JL, Liu Y, Johnson DR, Grzybicki DM, Raab SS, 2005, Interobserver variability in the fine needle aspiration biopsy diagnosis of follicular lesions of the thyroid gland. Acta Cytol 49(4): 378–382 Elsheikh TM, Asa SL, Chan JK, DeLellis RA, Heffess CS, LiVolsi VA, Wenig BM, 2008, Interobserver and intraobserver variation among experts in the diagnosis of thyroid follicular lesions with borderline nuclear features of papillary carcinoma. Am J Clin Pathol 130(5):736–744., DOI 10.1309/AJCPKP2QUVN4RCCP Rosai J, 2008, Papillary thyroid carcinoma: a root-and-branch rethink. Am J Clin Pathol 130(5):683–686., DOI 10.1309/ AJCPBF63BWMCYSLW Bussolati G, 2008, Proper detection of the nuclear shape: ways and significance. Rom J Morphol Embryol 49(4):435–439 Beesley MF, McLaren KM, 2002, Cytokeratin 19 and galectin-3 immunohistochemistry in the differential diagnosis of solitary thyroid nodules. Histopathology 41(3):236–243 Mai KT, Bokhary R, Yazdi HM, Thomas J, Commons AS, 2002, Reduced HBME-1 immunoreactivity of papillary thyroid carcinoma and papillary thyroid carcinoma-related neoplastic lesions with Hurthle cell and/or apocrine-like changes. Histopathology 40(2): 133–142 Menon MP, Khan A, 2009, Micro-RNAs in thyroid neoplasms: molecular, diagnostic and therapeutic implications. J Clin Pathol 62(11):978–985., DOI 10.1136/jcp.2008.063909 Nikiforov YE, 2002, RET/PTC rearrangement in thyroid tumors. Endocr Pathol 13(1):3–16 Nikiforova MN, Biddinger PW, Caudill CM, Kroll TG, Nikiforov YE, 2002, PAX8-PPARgamma rearrangement in thyroid tumors: RT-PCR and immunohistochemical analyses. Am J Surg Pathol 26(8):1016–1023 Rosai J, 2003, Immunohistochemical markers of thyroid tumors: significance and diagnostic applications. Tumori 89(5):517–519 Sack MJ, Astengo-Osuna C, Lin BT, Battifora H, LiVolsi VA, 1997, HBME-1 immunostaining in thyroid fine-needle aspirations: a useful marker in the diagnosis of carcinoma. Mod Pathol 10(7):668–674 Xu XC, el-Naggar AK, Lotan R, 1995, Differential expression of galectin-1 and galectin-3 in thyroid tumors. Potential diagnostic implications. Am J Pathol 147(3):815–822 Sobrinho-Simoes M, Eloy C, Magalhaes J, Lobo C, Amaro T. Follicular thyroid carcinoma. Mod Pathol 24(Suppl 2):S10–S18., DOI 10.1038/modpathol.2010.133 Kakudo K, Bai Y, Katayama S, Hirokawa M, Ito Y, Miyauchi A, Kuma K, 2009, Classification of follicular cell tumors of the thyroid gland: analysis involving Japanese patients from one institute. Pathol Int 59(6):359–367., DOI 10.1111/j.1440-1827. 2009.02378.x Holaska JM, Kowalski AK, Wilson KL, 2004, Emerin caps the pointed end of actin filaments: evidence for an actin cortical network at the nuclear inner membrane. PLoS Biol 2(9):E231., DOI 10.1371/ journal.pbio.0020231 Wilson KL, Holaska JM, Montes de Oca R, Tifft K, Zastrow M, Segura-Totten M, Mansharamani M, Bengtsson L, 2005, Nuclear membrane protein emerin: roles in gene regulation, actin dynamics and human disease. Novartis Found Symp 264:51–58, discussion 58–62, 227–230 Fischer AH, Taysavang P, Weber CJ, Wilson KL, 2001, Nuclear envelope organization in papillary thyroid carcinoma. Histol Histopathol 16(1):1–14 Asioli S, Bussolati G, 2009, Emerin immunohistochemistry reveals diagnostic features of nuclear membrane arrangement in thyroid lesions. Histopathology 54(5):571–579., DOI 10.1111/j.1365-2559. 2009.03259.x Asioli S, Maletta F, Pacchioni D, Lupo R, Bussolati G. Cytological detection of papillary thyroid carcinomas by nuclear membrane decoration with emerin staining. Virchows Arch 457(1):43–51., DOI 10.1007/s00428-010-0910-z Kinsella MD, Hinrichs B, Cohen C, Siddiqui MT. Highlighting nuclear membrane staining in thyroid neoplasms with emerin: review and diagnostic utility. Diagn Cytopathol 41(6):497–504., DOI 10. 1002/dc.22870 Rosai J, 2011, Thyroid gland. In: Rosai J, ed, Rosai and Ackerman’s surgical pathology, 10th edn. Elsevier/Saunders, Philadelphia, PA, p 505 Lloyd RV, Erickson LA, Casey MB, Lam KY, Lohse CM, Asa SL, Chan JK, DeLellis RA, Harach HR, Kakudo K, LiVolsi VA, Rosai J, Sebo TJ, Sobrinho-Simoes M, Wenig BM, Lae ME, 2004, Observer variation in the diagnosis of follicular variant of papillary thyroid carcinoma. Am J Surg Pathol 28(10):1336–1340 Batistatou A, Scopa CD, 2009, Pathogenesis and diagnostic significance of nuclear grooves in thyroid and other sites. Int J Surg Pathol 17(2):107–110., DOI 10.1177/1066896908316071 Rosai J, Kuhn E, Carcangiu ML, 2006, Pitfalls in thyroid tumour pathology. Histopathology 49(2):107–120., DOI 10.1111/j.1365-2559. 2006.02451.x Scopa CD,Melachrinou M, Saradopoulou C,MerinoMJ, 1993, The significance of the grooved nucleus in thyroid lesions. Mod Pathol 6(6):691–694 Fenech M, Kirsch-Volders M, Natarajan AT, Surralles J, Crott JW, Parry J, Norppa H, Eastmond DA, Tucker JD, Thomas P. Molecular mechanisms of micronucleus, nucleoplasmic bridge and nuclear bud formation in mammalian and human cells. Mutagenesis 26(1):125– 132., DOI 10.1093/mutage/geq052 Samanta S, Dey P. Micronucleus and its applications. Diagn Cytopathol 40(1):84–90., DOI 10.1002/dc.21592 Lee YH, Oh BK, Yoo JE, Yoon SM, Choi J, Kim KS, Park YN, 2009, Chromosomal instability, telomere shortening, and inactivation of p21(WAF1/CIP1, in dysplastic nodules of hepatitis B virusassociated multistep hepatocarcinogenesis. Mod Pathol 22(8):1121– 1131., DOI 10.1038/modpathol.2009.76 Caruso RA, Fedele F, Crisafulli C, Paparo D, Parisi A, Luciano R, Cavallari V. Abnormal nuclear structures, micronuclei, nuclear blebs, strings, and pockets, in a case of anaplastic giant cell carcinoma of the thyroid: an immunohistochemical and ultrastructural study. Ultrastruct Pathol 35(1):14–18., DOI 10.3109/01913123.2010.517899 Nilsson G, 1978, Micronuclei studied in fine needle goitre aspirates. Acta Pathol Microbiol Scand A 86(3):201–204 Pavlov AV, Aleksandrov Iu K, Beliakov IE, Korableva TV, 2007, Morphological analysis of genetically damaged thyrocytes in nodular pathology of the thyroid gland. Vestn Khir Im I I Grek 166(2):58–61 Zaharopoulos P,Wong J,Wen JW(1998, Nuclear protrusions in cells from cytologic specimens. Mechanisms of formation. Acta Cytol 42(2):317–329 Jankovic B, Le KT, Hershman JM Clinical Review: Hashimoto’s thyroiditis and papillary thyroid carcinoma: is there a correlation?. J Clin Endocrinol Metab 98(2):474–482., DOI 10.1210/jc.2012-2978 ChuiMH, Cassol CA, Asa SL,Mete O. Follicular epithelial dysplasia of the thyroid: morphological and immunohistochemical characterization of a putative preneoplastic lesion to papillary thyroid carcinoma in chronic lymphocytic thyroiditis. Virchows Arch 462(5):557– 563., DOI 10.1007/s00428-013-1397-1 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 357 EP 366 DI 10.1007/s12253-014-9828-0 PG 10 ER PT J AU Hu, Q Luo, T He, P Zhong, X Tian, T Lv, Q Yan, X Zheng, H AF Hu, Qiancheng Luo, Ting He, Ping Zhong, Xiaorong Tian, Tinglun Lv, Qing Yan, Xi Zheng, Hong TI Trends and Present Treatment Patterns of Early Breast Cancer in Southwest China SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Early breast cancer; Southwest China; Trends; Present treatment ID Early breast cancer; Southwest China; Trends; Present treatment AB To analyze the changing patterns of treatment and to explore the current treatment approaches for breast cancer in Southwest China, we conducted a population-based retrospective cohort study of early breast cancer cases. The data of patients who registered in the information management system for breast cancer in Huaxi Hospital, Sichuan University from 1989 to 2012 were extracted. Nearly all patients underwent surgery, among whom radical mastectomy was the predominant option. Chemotherapy (88.7 %) was the most predominant adjuvant therapy approach. The percentage of patients receiving radiation therapy displayed fluctuant increase, which was 37.1 % in 2001 and reached up to 67.6 % in 2011. Besides, the endocrinetherapy became more and more popular in the hormone-receptor positive patients and the percentage of endocrinetherapywas increased from54.1 at 2001 to 85.6 % at 2011. However, more than 10 % of hormone-receptor positive patients still did not receive endocrinetherapy annually. The hormone-receptor positive patients who received endocrinetherapy had better 5-year disease free survival (DFS) and overall survival (OS) compared to those without endocrinetherapy (5-y DFS: 88.4% vs. 75.1 %, P<0.001; 5-y OS: 95.7 % vs. 88.4 %, P<0.001). N stage appeared to have greater impact on the 5-year DFS and OS than molecular subtyping. The treatment for breast cancer in China has been significantly improved but more attentions should be paid to radiotherapy and endocrine therapy. In addition, the value of N stage in the prognosis of breast cancer should not be ignored when the molecular typing draws more and more attentions. C1 [Hu, Qiancheng] West China Hospital, Sichuan University, Cancer Center and State Key Laboratory of Biotherapy, Laboratory of Molecular Diagnosis of Cancer, Department of Head & Neck and Mammary Oncology and Department of Medical Oncology, No.37 Guo Xue Xiang, 610041 Chengdu, China. [Luo, Ting] West China Hospital, Sichuan University, Cancer Center and State Key Laboratory of Biotherapy, Laboratory of Molecular Diagnosis of Cancer, Department of Head & Neck and Mammary Oncology and Department of Medical Oncology, No.37 Guo Xue Xiang, 610041 Chengdu, China. [He, Ping] West China Hospital, Sichuan University, Cancer Center and State Key Laboratory of Biotherapy, Laboratory of Molecular Diagnosis of Cancer, Department of Head & Neck and Mammary Oncology and Department of Medical Oncology, No.37 Guo Xue Xiang, 610041 Chengdu, China. [Zhong, Xiaorong] West China Hospital, Sichuan University, Cancer Center and State Key Laboratory of Biotherapy, Laboratory of Molecular Diagnosis of Cancer, Department of Head & Neck and Mammary Oncology and Department of Medical Oncology, No.37 Guo Xue Xiang, 610041 Chengdu, China. [Tian, Tinglun] West China Hospital, Sichuan University, Cancer Center and State Key Laboratory of Biotherapy, Laboratory of Molecular Diagnosis of Cancer, Department of Head & Neck and Mammary Oncology and Department of Medical Oncology, No.37 Guo Xue Xiang, 610041 Chengdu, China. [Lv, Qing] Sichuan University, West China Hospital, Department of Breast and Thyroid Surgery, 610041 Chengdu, China. [Yan, Xi] West China Hospital, Sichuan University, Cancer Center and State Key Laboratory of Biotherapy, Laboratory of Molecular Diagnosis of Cancer, Department of Head & Neck and Mammary Oncology and Department of Medical Oncology, No.37 Guo Xue Xiang, 610041 Chengdu, China. [Zheng, Hong] West China Hospital, Sichuan University, Cancer Center and State Key Laboratory of Biotherapy, Laboratory of Molecular Diagnosis of Cancer, Department of Head & Neck and Mammary Oncology and Department of Medical Oncology, No.37 Guo Xue Xiang, 610041 Chengdu, China. RP Zheng, H (reprint author), West China Hospital, Sichuan University, Cancer Center and State Key Laboratory of Biotherapy, Laboratory of Molecular Diagnosis of Cancer, Department of Head & Neck and Mammary Oncology and Department of Medical Oncology, 610041 Chengdu, China. EM hongzheng11@gmail.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90 Huang X-E, Hirose K, Wakai K et al, 2004, Comparison of lifestyle risk factors by family history for gastric, breast, lung and colorectal cancer. Asian Pac J Cancer Prev 5:419–427 Jemal A, Center MM, Desantis C, Ward EM, 2010, Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol Biomarkers Prev 19:1893–1907 Fan L, Zheng Y, Yu KD et al, 2009, Breast cancer in a transitional society over 18 years: trends and present status in Shanghai, China. Breast Cancer Res Treat 117:409–416 Berry DA, Cronin KA, Plevritis SK et al, 2005, Effect of screening and adjuvant therapy on mortality from breast cancer. N Engl J Med 353:1784–1792 Fisher B, Anderson S, Bryant J et al, 2002, Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 347:1233–1241 Ctsu RI, 2005, Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet 365:1687–1717 Howell A, Cuzick J, Baum M et al, 2005, Results of the ATAC, Arimidex, tamoxifen, alone or in combination, trial after completion of 5 years’ adjuvant treatment for breast cancer. Lancet 365:60–62 Thurlimann B, Keshaviah A, Coates AS et al, 2005, A comparison of letrozole and tamoxifen in postmenopausal women with early breast cancer. N Engl J Med 353:2747–2757 Coombes RC, Hall E, Gibson LJ et al, 2004, A randomized trial of exemestane after two to three years of tamoxifen therapy in postmenopausal women with primary breast cancer. N Engl JMed 350:1081– 1092 Eggemann H, Ignatov A, Smith BJ et al, 2013, Adjuvant therapy with tamoxifen compared to aromatase inhibitors for 257 male breast cancer patients. Breast Cancer Res Treat 137:465–470 Perez EA, Romond EH, Suman VJ et al, 2011, Four-year follow-up of trastuzumab plus adjuvant chemotherapy for operable human epidermal growth factor receptor 2–positive breast cancer: Joint analysis of data from NCCTG N9831 and NSABP B-31. J Clin Oncol 29:3366–3373 Welslau M, Dieras V, Sohn JH et al, 2014, Patient-reported outcomes from EMILIA, a randomized phase 3 study of Trastuzumab Emtansine, T-DM1, versus capecitabine and lapatinib in human epidermal growth factor receptor 2-positive locally advanced or metastatic breast cancer. Cancer-Am Cancer Soc 120:642–651 Goldhirsch A, Wood WC, Senn HJ, Glick JH, Gelber RD, 1995, Meeting highlights: international consensus panel on the treatment of primary breast cancer. J Natl Cancer Inst 87:1441–1445 Goldhirsch A, Glick JH, Gelber RD, Senn HJ, 1998, Meeting highlights: international consensus panel on the treatment of primary breast cancer. J Natl Cancer Inst 90:1601–1608 Goldhirsch A, Glick JH, Gelber RD, Coates AS, Senn HJ, 2001, Meeting highlights: international consensus panel on the treatment of primary breast cancer. Seventh international conference on adjuvant therapy of primary breast cancer. J Clin Oncol 19:3817–3827 Goldhirsch A, Wood WC, Gelber RD, Coates AS, Thurlimann B, Senn HJ, 2003, Meeting highlights: updated international expert consensus on the primary therapy of early breast cancer. J Clin Oncol 21:3357–3365 Goldhirsch A, Glick JH, Gelber RD et al, 2005, Meeting highlights: international expert consensus on the primary therapy of early breast cancer 2005. Ann Oncol 16:1569–1583 Goldhirsch A, Ingle JN, Gelber RD et al, 2009, Thresholds for therapies: highlights of the St Gallen international expert consensus on the primary therapy of early breast cancer 2009. Ann Oncol 20: 1319–1329 Hammond ME, Hayes DF, DowsettMet al, 2010, American society of clinical oncology/College of American pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer, unabridged version). Arch Pathol Lab Med 134:e48–72 Goldhirsch A, Wood WC, Coates AS et al, 2011, Strategies for subtypes–dealing with the diversity of breast cancer: highlights of the St. Gallen international expert consensus on the primary therapy of early breast cancer 2011. Ann Oncol 22:1736–1747 Network NCC, NCCN clinical practice guidelines in oncology: breast cancer, 2007, National comprehensive cancer network Harbeck N, Jakesz R, 2007, St. Gallen 2007: breast cancer treatment consensus report. Breast Care 2:130–134 Parkin DM, Fernandez LM, 2006, Use of statistics to assess the global burden of breast cancer. Breast J 12:S70–S80 Carey LA, Perou CM, Livasy CA et al, 2006, Race, breast cancer subtypes, and survival in the Carolina breast cancer study. JAMA 295:2492–2502 Xue C, Wang X, Peng R et al, 2012, Distribution, clinicopathologic features and survival of breast cancer subtypes in Southern China. Cancer Sci 103:1679–1687 Sorlie T, Perou CM, Tibshirani R et al, 2001, Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98:10869–10874 Kurebayashi J, Moriya T, Ishida T et al, 2007, The prevalence of intrinsic subtypes and prognosis in breast cancer patients of different races. The Breast 16:72–77 Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V, 2007, Descriptive analysis of estrogen receptor, ER)‐negative, progesterone receptor, PR)‐negative, and HER2‐negative invasive breast cancer, the so‐called triple‐negative phenotype. Cancer 109:1721–1728 Haffty BG, Yang Q, Reiss M et al, 2006, Locoregional relapse and distant metastasis in conservatively managed triple negative earlystage breast cancer. J Clin Oncol 24:5652–5657 Zhao J, Liu H,WangMet al, 2009, Characteristics and prognosis for molecular breast cancer subtypes in Chinese women. J Clin Oncol 100:89–94 Panjwani P, Epari S, Karpate A et al., 2010, Assessment of HER-2/ neu status in breast cancer using fluorescence in situ hybridization & immunohistochemistry: experience of a tertiary cancer referral centre in India PressMF, Sauter G, Bernstein L et al, 2005, Diagnostic evaluation of HER-2 as a molecular target: an assessment of accuracy and reproducibility of laboratory testing in large, prospective, randomized clinical trials. Clin Cancer Res 11:6598–6607 Yu K-D, Li S, Shao Z-M, 2011, Different annual recurrence pattern between lumpectomy and mastectomy: implication for breast cancer surveillance after breast-conserving surgery. Oncologist 16:1101– 1110 Smith GL,XuY, Shih Y-CTet al, 2009, Breast-conserving surgery in older patients with invasive breast cancer: current patterns of treatment across the United States. J Am Coll Surg 209:425–433. e422 Li J, Zhang B-N, Fan J-H et al, 2011, A Nation-Wide multicenter 10- year, 1999-2008, retrospective clinical epidemiological study of female breast cancer in china. BMC Cancer 11:364 Slamon D, EiermannW, Robert N et al, 2011, Adjuvant trastuzumab in HER2-positive breast cancer. N Engl J Med 365:1273–1283 Hershman DL, Shao T, Kushi LH et al, 2011, Early discontinuation and non-adherence to adjuvant hormonal therapy are associated with increased mortality in women with breast cancer. Breast Cancer Res Treat 126:529–537 Barron TI, Connolly R, Bennett K, Feely J, Kennedy MJ, 2007, Early discontinuation of tamoxifen. Cancer 109:832–839 Baum M, Budzar A, Cuzick J et al, 2002, Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomised trial. Lancet 359:2131–2139 Ellis MJ, Coop A, Singh B et al, 2001, Letrozole is more effective neoadjuvant endocrine therapy than tamoxifen for ErbB-1- and/or ErbB-2-positive, estrogen receptor-positive primary breast cancer: Evidence from a phase III randomized trial. J Clin Oncol 19:3808– 3816 Dixon JM, Jackson J, Hills M et al, 2004, Anastrozole demonstrates clinical and biological effectiveness in oestrogen receptor-positive breast cancers, irrespective of the erbB2 status. Eur J Cancer 40: 2742–2747 Ihemelandu CU, Leffall LD Jr, Dewitty RL et al, 2007, Molecular breast cancer subtypes in premenopausal and postmenopausal African-American women: age-specific prevalence and survival. J Surg Oncol 143:109–118 Wang GS, Zhu H, Bi SJ, 2012, Pathological features and prognosis of different molecular subtypes of breast cancer. Mol Med Report 6: 779–782 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 367 EP 378 DI 10.1007/s12253-014-9829-z PG 12 ER PT J AU Ibrahim, RT Abdel-Raouf, MS AF Ibrahim, R Taiseer Abdel-Raouf, M Samar TI Immunohistochemical Study of Glypican-3 and HepPar-1 in Differentiating Hepatocellular Carcinoma from Metastatic Carcinomas in FNA of the Liver SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HCC; MA; Fine needle aspiration; Glypican-3; GPC-3; HepPar-1; Immunohistochemistry ID HCC; MA; Fine needle aspiration; Glypican-3; GPC-3; HepPar-1; Immunohistochemistry AB Hepatocellular carcinoma (HCC) remains a common malignant cancer worldwide, it is considered the fifth most common malignant cancer. On the other hand,metastatic tumors are widespread in the liver , with metastatic adenocarcinoma (MA) constituting the greatest part, therefore differentiation of HCC from MA is a frequent problem facing the pathologist especially in liver fine-needle aspiration biopsies. Evaluating the diagnostic value of glypican-3 (GPC-3) and HepPar-1 immunostaining in differentiating hepatocellular carcinoma from metastatic tumors in liver cell block material. Fourty eight cell blocks prepared from FNA from the liver ( 30 cases HCC, 18 cases metastatic carcinoma in liver ) stained by Glypican -3 and HepPar-1 immunohistochemical markers. Glypican-3 was immunoexpressed in 97 % of cases of HCC while all cases of metastatic carcinoma were negative . HepPar-1 was expressed in 93 % of cases of HCC and 11 % of metastatic carcinoma of the liver . In this study the sensitivity of GPC3 in the diagnosis of HCC in cytological material was 96.7% and the specificity was 100% while the sensitivity and specificity of HepPar-1 was 93.3 % and 88.9 % respectively. Immunohistochemical staining for GPC-3 in cell block material of the liver is highly sensitive and specific and it is a valuable tool capable of differentiating HCC from most of metastatic tumors of the liver. C1 [Ibrahim, R Taiseer] Zagazig University, Faculty of Medicine, Department of PathologyAl Sharqiyah, Egypt. [Abdel-Raouf, M Samar] Zagazig University, Faculty of Medicine, Department of PathologyAl Sharqiyah, Egypt. RP Abdel-Raouf, MS (reprint author), Zagazig University, Faculty of Medicine, Department of Pathology, Al Sharqiyah, Egypt. EM samar_foda2010@yahoo.com CR El Serag HB, 2002, Hepatocellullar carcinoma:an epidemiologic view. J Clin Gastroenterol 35(5 suppl 2):S72–S78 Waly Raphael S, Yangde Z and Yuxiang C, 2012)Hepatocellular carcinom :focus on different aspects of management.ISRN Oncol;2012: 421673 National Cancer Instiute Cairo, 2010, Cancer registry 2002-2007. Website: www.Nci.edu.eg.data Anwar AA, Khaled HM, Amra H et al, 2008, Changing pattern of hepatocellullar carcinoma, HCC, and its risk factors in Egypt: possibilities for prevention. Mutat Res 659:176–184 Aleem E, Elshayeb A, Elhabschi N,Mansour AR, Gowily A, Hela A, 2012, Serum IGFBP-1 is a more effective predictor than IGF-1 and IGF-2 for the development of hepatocellular carcinoma in patients with chronic HCV infection. Oncol Lett 3(3):704–712 Choi BI, 2004, The current status of imaging diagnosis of hepatocellullar carcinoma. Liver Transpl 10(2 suppl 1):S20–S25 Ligato S, Mandich D, Richard W, 2008, Utility of glypican-3 in differentiating hepatocellular carcinoma from other primary and metastatic lesions in FNA of the liver: an immunocytochemical study. Mod Pathol 21:626–631 Kakar S, Gown AM, Goodman ZD, Ferrell LD, 2007, Best practices in diagnostic immunohistochemistry: hepatocellular carcinoma versus metastatic neoplasms. Archit Pathol Lab 131:1648–1654 Kandil D, Cooper K, 2009, Glypican-3 a novel diagnosticmarker for hepatocellular carcinoma and more. Adv Anat Pathol 16:125–129 Yamauchi N,Watanabe A, HishinumaMet al, 2005, The glypican 3 oncofetal protein is a promising diagnostic marker for hepatocellular carcinoma. Mod Pathol 18:1591–1598 Suriawinata A, Thung SN, 2010, Molecular signature of early hepatocellular carcinoma. Oncol 78:36–39 Anatelli F, Chuang S, Yang X, Wang H, 2008, Value of glypican 3 immunostaining in the diagnosis of hepatocellullar carcinoma on needle biopsy. Am J Clin Pathol 130:219–223 Di Tommaso L, Franchi G, Park YN et al, 2007, Diagnostic value of HSP70, glypican 3 and glutamine synthetase in hepatocellullar nodules in cirrhosis. Hepatology 45:725–734 Shafizadeh N, Ferrell LD, Kakar S, 2008, Utility and limitations of glypican-3 expression for the diagnosis of hepatocellular carcinoma at both ends of the differentiation spectrum. Mod Pathol 21:1011– 1018 Yan BC, Gong C, Song J et al, 2010, Arginase-1: a new immunohistochemical marker of hepatocytes and hepatocellular neoplasms. Am J Surg Pathol 34:1147–1154 Hajosi-Kalcakosz S, Dezso K, Buggik E et al, 2012, Enhancer of zeste homologue 2(EZH2, is a reliable immunohistochemical marker to differentiate malignant and bengin hepatic tumors. Diagn Pathol 7:86 Thiese ND,Curado MP and Franceschi S, 2010, Hepatocellullar carcinoma .In WHO classification of tumors of the digestive system .4th edition .Edited by Bosman FT, Carneiro F,Hruban RH, Thiese ND. Lyon, France IARC Press:205-216 Ishak KG,Goodman ZD and Stocker JT, 2001, Tumor of liver and intrahepatic bile ducts: in :Atlas of tumor pathology, 3rd series,Fascicle 31. Washington, DC: Armed Forces Institute of Pathology; 9-48 Sternberg SS, Mills SE, Carter D, 2010, Sternberg s diagnostic surgical pathology, 5th edn. Wolter Kluwer Lippincott Williams& Wilkins, Philadelphia Wee A, 2011, Fine needle aspiration biopsy of hepatocellullar carcinoma and hepatocellular nodular lesions: role, controversies and approach to diagnosis. Cytopathology 22:287–305 Ceyhan K, Kupana SA, Bektas M et al, 2007, The diagnostic value of on-site cytopathological evaluation and cell block preparation in fine needle aspiration cytology of liver masses. Cytopathology 17: 267–274 Shiran MS, Isa MR, Sherina MS, Rampal L, Hairuszah I, Sabariah AR, 2006, The utility of hepatocyte paraffin-1 antibody in the immunohistochemical distinction of hepatocellular carcinoma from cholangiocarcinoma and metastatic carcinoma. Malays J Pathol 28(2):87–92 Shirakawa H, Kuronuma T, Nishimura Y, 2009, Glypican-3 as a useful diagnostic marker for a component of hepatocellullar carcinoma in human liver cancer. Int J Oncol 34:649–656 Abdelgawad IA, Mossallam GI, Radwan NH, Elzawahry HM, Elhifnawy NM, 2013, Can glypican-3 be diagnostic for early hepatocellular carcinoma among Egyptian patients. Asian Pac J Cancer Prev 14:7345–7349 Zaakook M, Ayoub M, Sinna EA, El-Sheik S, 2013, Role of glypican-3 immunocytochemistry in differentiatating hepatocellullar carcinoma from metastatic carcinima of the liver utilizing fine needle aspiration cytology. J Egypt Nati Inst Dec 25(4):173–80 Nasser A, Cohen C, Siddiqui MT, 2009, Utility of glypican-3 and survivin in differentiating hepatocellular carcinoma from benign and preneoplastic hepatic lesions and metastatic carcinomas in liver fineneedle aspiration biopsies. Diagn Cytopathol 37(9):629–35 McKnight R, Nassar A, Cohen C, Siddiqui MT, 2012, Arginase-1: a novel immunohistochemical marker of hepatocellullar differentiation in fine needle aspiration cytology. Cancer Cytopathol 120:223–229 Fujiwara M, Kwok S, Yano H, Pai RK, 2012, Arginase-1 is a more sensitivemarker of hepatic differentiation than HepPar-1 and Glypican-3 in fine needle aspiration biopsies. Cancer Cytopathol 120:230–237 Timek DT, Shi J, Liu H, Lin F, 2012, Arginase-1, HepPar-1 and Glypican-3 are the most effective panel of markers in differentiating hepatocellular carcinoma from metastatic tumor on fine needle aspiration specimens. Am J Clin Pathol 138:203–210 Radwan NA, Ahmed NS, 2012, The diagnostic value of arginase-1 immunostaining in differentiating hepatocellular carcinoma from metastatic carcinoma and cholangiocarcinoma as compared to HepPar-1. Diagn Pathol 7:149 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 379 EP 387 DI 10.1007/s12253-014-9830-6 PG 9 ER PT J AU Yong, Z Zhang, Q Fan, Z Sun, J Liu, X Cheng, L Li, A Xu, J AF Yong, Zhang Zhang, Qiang Fan, Zhongze Sun, Jue Liu, Xulin Cheng, Lingling Li, Ao Xu, Jianhua TI A Chinese herbal Formula, Chang-Wei-Qin, Synergistically Enhances Antitumor Effect of Oxaliplatin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Xenograft model; Changweiqing; Platinum drugs; Copper transporters ID Colorectal cancer; Xenograft model; Changweiqing; Platinum drugs; Copper transporters AB Chang-Wei-Qing (CWQ), a Chinese herbal formula, has long been employed clinically to treat cancers. In this study, we investigated the synergistic effect of CWQ with oxaliplatin (OXA) on the tumor growth inhibition of orthotopic transplanted colon cancer and explored the underlying mechanism. By generating the orthotopic transplanted nude mouse model of human colon carcinoma, we found that (1) CWQ enhanced OXA-mediated tumor suppression by 4.25-fold. (2) The body weights of nude mice in CWQ group and combination group were increased. (3) The survival time of tumor-bearing nude mice was dramatically improved in CWQ and CWQ/OXA group. (4) CWQ could restore OXA-mediated deregulation of copper transporter genes, hCTR1, ATP7A and ATP7B. (5) OXA-induced drug resistance index for OXA, 5-FU, HCPT and THP were 7.59, 4.28, 5.78 and 4.50 respectively, while the reversal index by combined CWQ treatment were 6.57, 2.61, 4.97 and 3.10, respectively. Our study demonstrates that the repeated intraperitoneal injection of OXA can induce multi-drug resistance of orthotopic transplanted nude mouse model of human colon carcinoma. The CWQ treatment can alleviate OXA-triggered side effects and reverse platinum drug resistance via up-regulation of hCTR1 expression and down-regulation of ATP7A and ATP7B levels. C1 [Yong, Zhang] Shanghai University of Traditional Chinese Medicine, Putuo Hospital, Department of Medical Oncology, 164 Lanxi Road, Putuo District, 200062 Shanghai, China. [Zhang, Qiang] Shanghai University of Traditional Chinese Medicine, Putuo Hospital, Department of Medical Oncology, 164 Lanxi Road, Putuo District, 200062 Shanghai, China. [Fan, Zhongze] Shanghai University of Traditional Chinese Medicine, Putuo Hospital, Department of Medical Oncology, 164 Lanxi Road, Putuo District, 200062 Shanghai, China. [Sun, Jue] Shanghai University of Traditional Chinese Medicine, Putuo Hospital, Department of Medical Oncology, 164 Lanxi Road, Putuo District, 200062 Shanghai, China. [Liu, Xulin] Shanghai University of Traditional Chinese Medicine, Putuo Hospital, Department of Medical Oncology, 164 Lanxi Road, Putuo District, 200062 Shanghai, China. [Cheng, Lingling] Shanghai University of Traditional Chinese Medicine, Putuo Hospital, Department of Medical Oncology, 164 Lanxi Road, Putuo District, 200062 Shanghai, China. [Li, Ao] Shanghai University of Traditional Chinese Medicine, Putuo Hospital, Department of Medical Oncology, 164 Lanxi Road, Putuo District, 200062 Shanghai, China. [Xu, Jianhua] Shanghai University of Traditional Chinese Medicine, Putuo Hospital, Department of Medical Oncology, 164 Lanxi Road, Putuo District, 200062 Shanghai, China. RP Xu, J (reprint author), Shanghai University of Traditional Chinese Medicine, Putuo Hospital, Department of Medical Oncology, 200062 Shanghai, China. EM xujianhua_2013@126.com;xujianhua50@126.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA. Cancer J Clin 61(2):69–90 Yang Y, Cai, Q, Zou D, Yang L, 2013, Advances of molecular targeted drugs in the treatment of advanced colorectal cancer. Chinese J Cancer Biother 20(2):247–250 Shiragami R,Murata S, Kosugi C, Tezuka T, YamazakiM, Hirano A, Yoshimura Y, Suzuki M, Shuto K, Koda K, 2013, Enhanced antitumor activity of cerulenin combined with oxaliplatin in human colon cancer cells. Int J Oncol 43(2):431–438 McCleary NJ, Odejide O, Szymonifka J, Ryan D, Hezel A, Meyerhardt JA, 2013, Safety and effectiveness of oxaliplatin-based chemotherapy regimens in adults 75 years and older with colorectal cancer. Clin Colorectal Cancer 12(1):62–69 Gordon MA, ZhangW, Yang D, Iqbal S, El-Khouiery A, Nagashima F, Lurje G, Labonte M, Wilson P, Sherrod A et al, 2011, Genderspecific genomic profiling in metastatic colorectal cancer patients treated with 5-fluorouracil and oxaliplatin. Pharmacogenomics 12(1):27–39 Inoue Y, Matsumoto H, Yamada S, Kawai K, Suemizu H, Gika M, Takanami I, Iwazaki M, Nakamura M, 2010, Association of ATP7A expression and in vitro sensitivity to cisplatin in non-small cell lung cancer. Oncol Lett 1(5):837–840 Dmitriev OY, 2011, Mechanism of tumor resistance to cisplatin mediated by the copper transporter ATP7B. Biochem Cell biol= Biochimie et biologie cellulaire 89(2):138––147 Chen HH, Yan JJ, ChenWC, KuoMT, Lai YH, LaiWW, Liu HS, Su WC, 2012, Predictive and prognostic value of human copper transporter 1, hCtr1, in patients with stage III non-small-cell lung cancer receiving first-line platinum-based doublet chemotherapy. Lung Cancer 75(2):228–234 KuoMT, Fu S, Savaraj N, Chen HH, 2012, Role of the human highaffinity copper transporter in copper homeostasis regulation and cisplatin sensitivity in cancer chemotherapy. Cancer Res 72(18): 4616–4621 Yoshida H, Teramae M, Yamauchi M, Fukuda T, Yasui T, Sumi T, Honda K, Ishiko O, 2013, Association of copper transporter expression with platinum resistance in epithelial ovarian cancer. Anticancer Res 33(4):1409–1414 Howell SB, Safaei R, Larson CA, Sailor MJ, 2010, Copper transporters and the cellular pharmacology of the platinum-containing cancer drugs. Mol Pharmacol 77(6):887–894 Komatsu M, Sumizawa T, Mutoh M, Chen ZS, Terada K, Furukawa T, Yang XL, Gao H, Miura N, Sugiyama T et al, 2000, Copper-transporting P-type adenosine triphosphatase, ATP7B, is associated with cisplatin resistance. Cancer Res 60(5):1312–1316 Katano K, Safaei R, Samimi G, Holzer A, Rochdi M, Howell SB, 2003, The copper export pump ATP7B modulates the cellular pharmacology of carboplatin in ovarian carcinoma cells. Mol Pharmacol 64(2):466–473 Kummar S, Copur MS, Rose M, Wadler S, Stephenson J, O’Rourke M, Brenckman W, Tilton R, Liu SH, Jiang Z et al, 2011, A phase I study of the chinese herbal medicine PHY906 as a modulator of irinotecan-based chemotherapy in patients with advanced colorectal cancer. Clin Colorectal Cancer 10(2):85–96 Wang TC, Fang CN, Shen CC, Wei HY, Weng YP, Lin JY, Hsieh-Li HM, Lee CY, 2012, Yang-Dan-Tang, Identified from 15 Chinese Herbal Formulae. Inhibits Human Lung Cancer Cell Proliferation via Cell Cycle Arrest Evidence-based complementary and alternative medicine : eCAM 2012:276032 Zheng Z, Cho WC, Xu L, Wang J, Sze DM, 2013, Lessons learnt from evidence-based approach of using chinese herbal medicines in liver cancer. Evid-Based Complement Alternat Med : eCAM 2013: 656351 Maimon Y, Karaush V, Yaal-Hahoshen N, Ben-Yosef R, Ron I, Vexler A, Lev-Ari S, 2010, Effect of Chinese herbal therapy on breast cancer adenocarcinoma cell lines. J Int Med Res 38(6):2033– 2039 Shi Y, Jin, H, Wang Y, Yi J, Liu C, 2013, Rat Serum Containing of Buzhong Yi q i Decoction Reverse Chemotherapeutic Resistance in A549/DDP Cells by Downregulating Expression Levels of mTOR. Chinese J Exp Tradit Med Formulae 19(9):215–219 Jia QA, Ren ZG, Bu Y, Wang ZM, Zhang QB, Liang L, Jiang XM, Tang ZY, 2012, Herbal Compound “Songyou Yin” Renders Hepatocellular Carcinoma Sensitive to Oxaliplatin through Inhibition of Stemness. Evid-Based Complement Alternat Med : eCAM 2012:908601 Zhang Y, Sun XW, Xu JH, Lu H, Fan ZZ, Sun J, Zhang XX, 2012, 2012. Zhong xi yi jie he xue bao=J Chin Integr Med 10(8):901–910 Gao QT, Cheung JK, Li J, Chu GK, Duan R, Cheung AW, Zhao KJ, Dong TT, Tsim KW, 2006, A Chinese herbal decoction, Danggui Buxue Tang, prepared from Radix Astragali and Radix Angelicae Sinensis stimulates the immune responses. PlantaMed 72(13):1227– 1231 Ruan WJ, Lai MD, Zhou JG, 2006, Anticancer effects of Chinese herbal medicine, science or myth? J Zhejiang Univ Sci B 7(12): 1006–1014 Xu J, Fan Z, Sun J, ZhuM, Fei R,Han J, 2007, Effects of“ChangWei Qing”in Treating Advanced Gastrointestinal Cancer and Its Effects on Periphereal Blood MDR1 mRNA. Shanghai J Tradit Chin Med 41(5):40–42 Zhang Y, Xu J, Sun J, 2010, Clinical study of Jianpijiedu-Decoction plus FOLFOX4 regimen in the treatm ent of advanced colorectal cancer. Glob Tradit Chin Med 3(2):117–120 Nakagawa T, Inoue Y, Kodama H, Yamazaki H, Kawai K, Suemizu H, Masuda R, Iwazaki M, Yamada S, Ueyama Y et al, 2008, Expression of copper-transporting P-type adenosine triphosphatase, ATP7B, correlates with cisplatin resistance in human non-small cell lung cancer xenografts. Oncol Rep 20(2):265–270 Li J, Fan ZZ, Sun J, Xu JH, 2011, In vitro antimetastatic effect of Changweiqing through antiinvasion of hypoxic colorectal carcinoma LoVo cells. Chin J Integr Med 17(7): 517–524 Xu JH, Deng WL, Fan ZZ, 2010, [Effects of changwelqing on nuclear translocation of Y-box binding protein-1 and expression of P-glycoprotein in human colon cancer cell line with drug-resistance induced by vincristine]. Zhongguo Zhong xi yi jie he za zhi Zhongguo Zhongxiyi jiehe zazhi=Chin J Integr Tradit West Med / Zhongguo Zhong xi yi jie he xue hui, Zhongguo Zhong yi yan jiu yuan zhu ban 30(7):743–747 Abraham EH, Prat AG, Gerweck L, Seneveratne T, Arceci RJ, Kramer R, Guidotti G, Cantiello HF, 1993, The multidrug resistance, mdr1, gene product functions as an ATP channel. Proc Natl Acad Sci U S A 90(1):312–316 Mishima M, Samimi G, Kondo A, Lin X, Howell SB, 2002, The cellular pharmacology of oxaliplatin resistance. Eur J Cancer 38(10): 1405–1412 Rabik CA, Dolan ME, 2007, Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat Rev 33(1):9–23 Martin LP, Hamilton TC, Schilder RJ, 2008, Platinum resistance: the role of DNA repair pathways. Clin Cancer Res: Off J Am Assoc Cancer Res 14(5):1291–1295 Lin H, Sun G, Qin F, Cao Y, Wang X, Chen J, Wang X, Huang H, 2013, A Randomized, Double-blinded, Drugcontrolled and Multicentre Clinical Trial of Chemotherapy Assisted with Jinlong Capsule on Gastric Cancer. Cancer Res Prev Treat 40(1):12–15 Hu J, Feng Y, Cao K, 2011, Immunoregulation of Yiyuan Huoxue Tang in Malignant Tumor Patients. Chinese Journal of Experimental Traditional Medical Formulae 17(21):245–247 Martinez-Balibrea E, Martinez-Cardus A, Musulen E, Gines A, Manzano JL, Aranda E, Plasencia C, Neamati N, Abad A, 2009, Increased levels of copper efflux transporter ATP7B are associated with poor outcome in colorectal cancer patients receiving oxaliplatin-based chemotherapy. Int J Cancer J Int du cancer 124(12):2905–2910 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 389 EP 397 DI 10.1007/s12253-014-9831-5 PG 9 ER PT J AU Docs, O Fazakas, F Horvath, LN Toth, L Andras, Cs Horvath, Zs Mehes, G AF Docs, Otto Fazakas, Ferenc Horvath, Lugosine Nora Toth, Laszlo Andras, Csilla Horvath, Zsolt Mehes, Gabor TI Changes of KRAS Exon 2 Codon 12/13 Mutation Status in Recurrent Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE KRAS; Colorectal carcinoma; Heterogeneity; Multiple tumors ID KRAS; Colorectal carcinoma; Heterogeneity; Multiple tumors AB Colorectal cancer (CRC) is a heterogeneous disease presenting with a wide spectrum of morphological and molecular characteristics sometimes even within the same patient. To understand the mechanisms of oscillations in the KRAS status we evaluated the collective of CRC patients tested using allele-specific PCR and Sanger-sequencing. Mutant KRAS allele was observed in 43.3 % of cases. Repeated analysis of KRAS status in recurrent tumors ormetastases was performed in 18/665 cases and a total of 6 cases with different KRAS status was found. In three cases the histological pattern of the tumor was identical. In one patient different histology and molecular status was seen between the primary and the recurrent tumor samples. In two further cases localization, histological type and KRAS mutational status all supported the occurrence of synchron/metachron colorectal tumors. In conclusion, both the progression of the original disease but also multiple tumor formation may contribute to mutation status differences during the course of colorectal carcinoma. C1 [Docs, Otto] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Fazakas, Ferenc] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Horvath, Lugosine Nora] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Toth, Laszlo] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Andras, Csilla] University of Debrecen, Department of Oncology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Horvath, Zsolt] University of Debrecen, Department of Oncology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. RP Mehes, G (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, H-4032 Debrecen, Hungary. EM gabor.mehes@med.unideb.hu;gabor.mehes@dote.hu CR Baselga J, 2001, The EGFR as a target for anticancer therapy – focus on cetuximab. Eur J Cancer 37:16–22 Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, LeppertM, Nakamura Y,White R, Smits AM, Bos JL, 1988, Genetic alterations during colorectal-tumor development. N Engl J Med 319(9):525–532 Vaughn CP, Zobell SD, Furtado LV, Baker CL, SamowitzWS, 2011, Frequency of KRAS, BRAF, and NRAS mutations in colorectal cancer. Gene Chromosom Cancer 50(5):307–312 Bamford S, Dawson E, Forbes S, Clements J, Pettett R, Dogan A, Flanagan A, Teague J, Futreal PA, Stratton MR, Wooster R, 2004, The COSMIC, catalogue of somatic mutations in cancer, database and website. Br J Cancer 91(2):355–358 Tanaka M, Omura K, Watanabe Y, Oda Y, Nakanishi I, 1994, Prognostic factors of colorectal cancer: K-ras mutation, overexpression of the p53 protein, and cell proliferative activity. J Surg Oncol 57(1):57–64 Andreyev HJ, Norman AR, Cunningham D, Oates JR, Clarke PA, 1998, Kirsten ras mutations in patients with colorectal cancer: the multicenter “RASCAL” study. J Natl Cancer Inst 90(9):675–684 Lievre A, Bachet JB, Le Corre D, Boige V, Landi B, Emile JF, Cote JF, Tomasic G, Penna C, Ducreux M, Rougier P, Penault-Llorca F, Laurent-Puig P, 2006, KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 66(8): 3992–3995 Losi L, Baisse B, Bouzourene H, Benhattar J, 2005, Evolution of intratumoral genetic heterogeneity during colorectal cancer progression. Carcinogenesis 26(5):916–922 Velho S, Oliveira C, Seruca R, 2009, KRAS mutations and antiepidermal growth factor receptor therapy in colorectal cancer with lymph node metastases. J Clin Oncol 27(1):158–159, author reply 159 Oltedal S, Aasprong OG,Moller JH, Korner H, Gilje B, Tjensvoll K, Birkemeyer EM, Heikkila R, Smaaland R, Nordgard O, 2011, Heterogeneous distribution of K-ras mutations in primary colon carcinomas: implications for EGFR-directed therapy. Int J Color Dis 26(10):1271–1277 Knijn N, Mekenkamp LJ, Klomp M, Vink-Borger ME, Tol J, Teerenstra S, Meijer JW, Tebar M, Riemersma S, van Krieken JH, Punt CJ, Nagtegaal ID, 2011, KRAS mutation analysis: a comparison between primary tumours and matched liver metastases in 305 colorectal cancer patients. Br J Cancer 104(6):1020–1026 Kimura K, Nagasaka T, Hoshizima N, Sasamoto H, Notohara K, Takeda M, Kominami K, Iishii T, Tanaka N, Matsubara N, 2007, No duplicate KRAS mutation is identified on the same allele in gastric or colorectal cancer cells with multiple KRAS mutations. J IntMed Res 35(4):450–457 Gasch C, Bauernhofer T, Pichler M, Langer-Freitag S, Reeh M, Seifert AM, Mauermann O, Izbicki JR, Pantel K, Riethdorf S, 2013, Heterogeneity of epidermal growth factor receptor status and mutations of KRAS/PIK3CA in circulating tumor cells of patients with colorectal cancer. Clin Chem 59(1):252–260 Coghlin C, Murray GI, 2010, Current and emerging concepts in tumour metastasis. J Pathol 222(1):1–15 Tziris N, Dokmetzioglou J, Giannoulis K, Kesisoglou I, Sapalidis K, Kotidis E, Gambros O, 2008, Synchronous and metachronous adenocarcinomas of the large intestine. Hippokratia 12(3):150–152 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 399 EP 404 DI 10.1007/s12253-014-9834-2 PG 6 ER PT J AU Satapathy, RSh Mohapatra, P Das, D Siddharth, S Kundu, NCh AF Satapathy, Ranjan Shakti Mohapatra, Purusottam Das, Dipon Siddharth, Sumit Kundu, Nath Chanakya TI The Apoptotic Effect of Plant Based Nanosilver in Colon Cancer Cells is a p53 Dependent Process Involving ROS and JNK Cascade SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Silver nanoparticle; Periwinkle; p53; MAPK; MMP; ROS; Colon cancer ID Silver nanoparticle; Periwinkle; p53; MAPK; MMP; ROS; Colon cancer AB Here, we report the p53 dependent mitochondriamediated apoptotic mechanism of plant derived silvernanoparticle (PD-AgNPs) in colorectal cancer cells (CRCs). PD-AgNPs was synthesized by reduction of AgNO3 with leaf extract of a medicinal plant periwinkle and characterized. Uptake of PD-AgNPs (ξ - 2.52±4.31 mV) in HCT116 cells was 3 fold higher in comparison to synthetic AgNPs (ξ + 2.293±5.1 mV). A dose dependent increase in ROS production, activated JNK and decreased mitochondrial membrane potential (MMP) were noted in HCT116 but not in HCT116 p53−/− cells after PD-AgNP exposure. PD-AgNP-mediated apoptosis in CRCs is a p53 dependent process involving ROS and JNK cascade. C1 [Satapathy, Ranjan Shakti] KIIT University, KIIT School of Biotechnology, Cancer Biology Division, Campus-11, Patia, Bhubaneswar, 751024 Orissa, India. [Mohapatra, Purusottam] KIIT University, KIIT School of Biotechnology, Cancer Biology Division, Campus-11, Patia, Bhubaneswar, 751024 Orissa, India. [Das, Dipon] KIIT University, KIIT School of Biotechnology, Cancer Biology Division, Campus-11, Patia, Bhubaneswar, 751024 Orissa, India. [Siddharth, Sumit] KIIT University, KIIT School of Biotechnology, Cancer Biology Division, Campus-11, Patia, Bhubaneswar, 751024 Orissa, India. [Kundu, Nath Chanakya] KIIT University, KIIT School of Biotechnology, Cancer Biology Division, Campus-11, Patia, Bhubaneswar, 751024 Orissa, India. RP Kundu, NCh (reprint author), KIIT University, KIIT School of Biotechnology, Cancer Biology Division, 751024 Orissa, India. EM cnkundu@gmail.com CR Satapathy SR, Mohapatra PM, Preet R, Das D, Sarkar B, Choudhuri T,WyattMD, Kundu CN, 2013, Silver nanoparticles induce apoptosis in human colon cancer cellsmediated through p53. Nanomedicine 8:1307–1322., DOI 10.2217/nnm.12.176 Tao A, Sinsermsuksaku P, Yang P, 2006, Polyhedral silver nanocrystals with distinct scattering signatures. Angew Chem Int Ed 45:4561–4597., DOI 10.1002/anie.200601277 AshaRani PV, Low Kah Mun G, Hande MP, Valiyaveettil S, 2009, Cytotoxicity and genotoxicity of silver nanoparticles in human cells. ACS Nano 3:279–290., DOI 10.1021/nn800596w Raveendran P, Fu J,Wallen SL, 2003, Completely “green” synthesis and stabilization of metal nanoparticles. J AmChem Soc 125:13940– 13951., DOI 10.1021/ja029267j Jha AK, Prasad K, Kulkarni AR, 2009, Synthesis of TiO2 nanoparticles using microorganisms. Colloids Surf B: Biointerfaces 71:226– 229., DOI 10.1016/j.colsurfb.2009.02.007 Dhillon GS, Brar SK, Kaur S, Verma M, 2012, Green approach for nanoparticle biosynthesis by fungi: current trends and applications. Crit Rev Biotechnol 32:49–73., DOI 10.3109/07388551.2010.550568 Iravani S, 2011, Green synthesis of metal nanoparticles using plants. Green Chem 13:2638–2650., DOI 10.1039/C1GC15386B Mukunthan K, Balaji S, 2012, Cashew apple juice, Anacardium occidentale L., speeds up the synthesis of silver nanoparticles. Int J Green Nanotechnol 4:71–79., DOI 10.1080/19430892.2012.676900 Kasthuri J, Veerapandian S, Rajendran N, 2009, Biological synthesis of silver and gold nanoparticles using apiin as reducing agent. Colloids Surf B: Biointerfaces 68:55–60., DOI 10.1016/j,colsurfb. 2008.09.021 Goyal P, 2008, In vitro evaluation of crude extracts of Catharanthus roseus for potential antibacterial activity. Int J Green Pharm 2:176– 181., DOI 10.4103/0973-8258.42739 Rasineni K, Bellamkonda R, Singareddy SR, Desireddy S, 2010, Antihyperglycemic activity of Catharanthus roseus leaf powder in streptozotocin-induced diabetic rats. Pharmacogn Res 2:195–201., DOI 10.4103/0974-8490.65523 Ponarulselvam S, Panneerselvam C, Murugan K, Aarthi N, KalimuthuK, Thangamani S, 2012, Synthesis of silver nanoparticles using leaves of Catharanthus roseus Linn. G. Don and their antiplasmodial activities. Asian Pac J Trop Biomed 2:574–580., DOI 10.1016/S2221-1691(12)60100-2 Jacob SJ, Finub JS, Narayanan A, 2012, Synthesis of silver nanoparticles using Piper longum leaf extracts and its cytotoxic activity against Hep-2 cell line. Colloids Surf B: Biointerfaces 91:212–214., DOI 10.1016/j.colsurfb.2011.11.001 Suzuki H, Toyooka T, Ibuki Y, 2007, Simple and easy method to evaluate uptake potential of nanoparticles in mammalian cells using a flow cytometric light scatter analysis. Environ Sci Technol 41:3018– 24., DOI 10.1021/es0625632 Parang Z, Keshavarz A, Farahi S, Elahi SM, Ghoranneviss M, Parhoodeh S, 2012, Fluorescence emission spectra of silver and silver/cobalt nanoparticles. Scientia Iranica F 19:943–47., DOI 10. 1016/j.scient.2012.02.026 Chairuangkitti P, Lawanprasert S, Roytrakul S, Aueviriyavit S, Phummiratch D, Kulthong K, Chanvorachote P, Maniratanachote R, 2013, Silver nanoparticles induce toxicity in A549 cells via ROSdependent and ROS-independent pathways. Toxicol Vitro 27:330– 338., DOI 10.1016/j.tiv.2012.08.021 Kumar S, Sitasawad SL, 2009, N-acetylcysteine prevents glucose/ glucose oxidase-induced oxidative stress, mitochondrial damage and apoptosis in h9c2 cells. Life Sci 84:328–36., DOI 10.1002/jctb.2023 Wu GS, 2004, The functional interactions between the p53 and MAPK signaling pathways. Cancer Biol Ther 3:156–161., DOI 10. 4161/cbt.3.2.614 Saha MN, Jiang H, Yang Y, Zhu X, Wang X, Schimmer AD, Qiu L, Chang H, 2012, Targeting p53 via JNK Pathway: a novel role of RITA for apoptotic signaling in multiple myeloma. PLoS ONE 7: e30215., DOI 10.1371/journal.pone.0030215 Taylor CA, Zheng Q, Liu Z, Thompson JE, 2013, Role of p38 and JNK MAPK signaling pathways and tumor suppressor p53 on induction of apoptosis in response to AdeIF5A1 in A549 lung cancer cells. Mol Cancer 12:35–45., DOI 10.1186/1476-4598-12-35 Wilhelm C, Billotey C, Roger J, Pons JN, Bacri JC, Gazeau F, 2003, Intracellular uptake of anionic superparamagnetic nanoparticles as a function of their surface coating. Biomaterials 24:1001–1011., DOI 10. 1016/S0142-9612(02)00440-4 Patil S, Sandberg A, Heckert E, Self W, Seal S, 2007, Protein adsorption and cellular uptake of cerium oxide nanoparticles as a function of zeta potential. Biomaterials 28:4600–4607., DOI 10.1016/ j.biomaterials.2007.07.029 Lim HK, Asharani PV, Hande MP, 2012, Enhanced genotoxicity of silver nanoparticles in DNA repair deficient Mammalian cells. Front Genet 3:104–116., DOI 10.3389/fgene.2012.00104 Derfus AM, Chan WCW, Bhatia SN, 2004, Intracellular delivery of quantum dots for live cell labeling and organelle tracking. Adv Mater 16:961–966., DOI 10.1021/nl102172j Xia T, Kovochich M, Brant J, Hotze M, Sempf J, Oberley T, Sioutas C, Yeh JI,WiesnerMR, Nel AE, 2006, Comparison of the abilities of ambient and manufactured nanoparticles to induce cellular toxicity according to an oxidative stress paradigm. Nano Lett 6:1794–1807., DOI 10.1021/nl061025k Zhivotovsky B, Orrenius S, Brustugus OT, Doskeland SO, 1998, Injected cytochrome c induces apoptosis. Nature 391:449–50., DOI 10. 1038/35060 JohnsonBW, Cepero E, Boise LH(2000, Bcl-xL inhibits cytochrome c release but notmitochondrial depolarization during the activation of multiple death pathways by tumor necrosis factor-alpha. J Biol Chem 275:31546–31553., DOI 10.1074/jbc.M001363200 Piao MJ, Kang KA, Lee IK, Kim HS, Kim S, Choi JY, Choi J, Hyun JW, 2011, Silver nanoparticles induce oxidative cell damage in human liver cells through inhibition of reduced glutathione and induction of mitochondria-involved apoptosis. Toxicol Lett 201:92– 100., DOI 10.1016/j.toxlet.2010.12.010 Cargnello M, Roux PP, 2011, Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev 75:50–83., DOI 10.1128/ MMBR.00031-10 Hsin YH, Chen CF, Huang S, Shih TS, Lai PS, Chueh PJ, 2008, The apoptotic effect of nanosilver is mediated by a ROS and JNK dependent mechanism involving the mitochondrial pathway in NIH3T3 cells. Toxicol Lett 179:130–139., DOI 10.1016/j.toxlet.2008.04.015 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 405 EP 411 DI 10.1007/s12253-014-9835-1 PG 7 ER PT J AU Todorova, AT Jordanov, HS Stancheva, SG Chalakov, JI Melnicharov, BM Kunev, VK Mitev, IV Kaneva, PR Goranova, ET AF Todorova, A Teodora Jordanov, H Stanislav Stancheva, S Gergana Chalakov, J Ivan Melnicharov, B Mincho Kunev, V Kuncho Mitev, I Vanio Kaneva, P Radka Goranova, E Teodora TI Mutational Status of CDKN2A and TP53 Genes in Laryngeal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CDKN2A; TP53; Mutations; Sequencing; Laryngeal squamous cell carcinoma ID CDKN2A; TP53; Mutations; Sequencing; Laryngeal squamous cell carcinoma AB Laryngeal squamous cell carcinoma (LSCC) is the second most common tumour of the head and neck. It is characterized by frequent aberrations in two cell-cycle regulators— CDKN2A and TP53. However, LSCC has been often studied as a part of the group of head and neck cancers and not as an individual entity. In the current study we aimed to examine mutation status of CDKN2A and TP53 genes in 108 LSCC patients. DNA was extracted from fresh-frozen tumour tissues; exons 1–3 of CDKN2A and exons 5–8 of TP53 were screened for mutations by direct sequencing. Genetic aberrations in CDKN2A were found in 16 (14.2 %) and those in TP53—in 56/108 (51.9 %) tumours. Seven mutations (two insertions, three deletions, one missense and one silent) detected in CDKN2Awere not described previously. Also, we found seven novel deletions and a novel indel in TP53. No significant associations with clinical features were found. However, TP53 mutations were predominantly observed in smokers with advanced stage tumours. Screening for genetic aberrations in a defined group of LSCC contributes to the knowledge about laryngeal carcinogenesis. Further investigations are required to confirm the observed trends in associations with clinical features. C1 [Todorova, A Teodora] Medical University of Sofia, Molecular Medicine Center, 2 Zdrave street, 1431 Sofia, Bulgaria. [Jordanov, H Stanislav] Medical University - Sofia, Clinic of Otorhinolaryngology, UMHAT, 8 Byalo More Street, 1527 Sofia, Bulgaria. [Stancheva, S Gergana] Medical University of Sofia, Molecular Medicine Center, 2 Zdrave street, 1431 Sofia, Bulgaria. [Chalakov, J Ivan] Medical University - Sofia, Clinic of Otorhinolaryngology, UMHAT, 8 Byalo More Street, 1527 Sofia, Bulgaria. [Melnicharov, B Mincho] Medical University - Sofia, Clinic of Otorhinolaryngology, UMHAT, 8 Byalo More Street, 1527 Sofia, Bulgaria. [Kunev, V Kuncho] Medical University - Sofia, Clinic of Otorhinolaryngology, UMHAT, 8 Byalo More Street, 1527 Sofia, Bulgaria. [Mitev, I Vanio] Medical University of Sofia, Molecular Medicine Center, 2 Zdrave street, 1431 Sofia, Bulgaria. [Kaneva, P Radka] Medical University of Sofia, Molecular Medicine Center, 2 Zdrave street, 1431 Sofia, Bulgaria. [Goranova, E Teodora] Medical University of Sofia, Molecular Medicine Center, 2 Zdrave street, 1431 Sofia, Bulgaria. RP Goranova, ET (reprint author), Medical University of Sofia, Molecular Medicine Center, 1431 Sofia, Bulgaria. EM goranova@mmcbg.org CR Leemans CR, Braakhuis BJM, Brakenhoff RH, 2011, The molecular biology of head and neck cancer. Nat Rev Cancer 11:9–22 Temam S, Koka V,Mamelle G et al, 2005, Treatment of the N0 neck during salvage surgery after radiotherapy of head and neck squamous cell carcinoma. Head Neck 27:653–658 Almadori G, Bussu F, Cadoni G et al, 2004, Multistep laryngeal carcinogenesis helps our understanding of the field cancerisation phenomenon: a review. Eur J Cancer 40:2383–2388 Perez-Ordonez B, Beauchemin M, Jordan RCK, 2006, Molecular biology of squamous cell carcinoma of the head and neck. J Clin Pathol 59:445–453 Kim MM, Califano JA, 2004, Molecular pathology of head-andneck cancer. Int J Cancer 112:545–553 Loyo M, Pai SI, 2008, The molecular genetics of laryngeal cancer. Otolaryngol Clin N Am 41:657–672 Longworth MS, Laimins LA, 2004, Pathogenesis of human papillomaviruses in differentiating epithelia. Microbiol Mol Biol Rev 68: 362–372 Van der Riet P, Nawroz H, Hruban RH et al, 1994, Frequent loss of chromosome 9p21-22 early in head and neck cancer progression. Cancer Res 54:1156–1158 Nobori T, Miura K, Wu DJ et al, 1994, Deletions of the cyclindependent kinase-4 inhibitor gene in multiple human cancers. Nature 368:753–756 Caldas C, Hahn SA, da Costa LT et al, 1994, Frequent somatic mutations and homozygous deletions of the p16, MTS1, gene in pancreatic adenocarcinoma. Nat Genet 8:27–32 Agnese V, Corsale S, Calo Vet al, 2006, Significance of P16INK4A hypermethylation gene in primary head/neck and colorectal tumors: it is a specific tissue event? Results of a 3-year GOIM, Gruppo Oncologico dell’Italia Meridionale, prospective study. Ann Oncol 17(Suppl 7):vii137–vii141 Pierini S, Jordanov SH, Mitkova AV et al, 2013, Promoter hypermethylation of CDKN2A, MGMT, MLH1 and DAPK genes in laryngeal squamous cell carcinoma and their associations with clinical profiles of the patients. Head Neck., DOI 10.1002/hed.23413 Kamijo T, Zindy F, RousselMF et al, 1997, Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19 ARF. Cell 91:649–659 Rocco JW, Sidransky D, 2001, p16(MTS-1/CDKN2/INK4a, in cancer progression. Exp Cell Res 264:42–55 Califano J, van der Riet P,WestraWet al, 1996, Genetic progression model for head and neck cancer: implications for field cancerization. Cancer Res 56:2488–2492 Mountzios G, Rampias T, Psyrri A, 2014, The mutational spectrum of squamous-cell carcinoma of the head and neck: Targetable genetic events and clinical impact. Ann Oncol., DOI 10.1093/annonc/mdu143 Xu J, Reumers J, Couceiro JR et al, 2011, Gain of function of mutant p53 by coaggregation with multiple tumor suppressors. Nat Chem Biol 7:285–295 Sanger F, Air GM, Barrell BG et al, 1997, Nucleotide sequence of bacteriophage phi X174 DNA. Nature 265:687–695 Liu L, Dilworth D, Gao L et al, 1999, Mutation of the CDKN2A 5’ UTR creates an aberrant initiation codon and predisposes to melanoma. Nat Genet 21:128–132 Debniak T, Scott RJ, Huzarski T et al, 2005, CDKN2A common variants and their association withmelanoma risk: a population-based study. Cancer Res 65:835–839 Nakagawa K, Conrad NK, Williams JP, Johnson BE, Kelley MJ, 1995, Mechanism of inactivation of CDKN2 and MTS2 in nonsmall cell lung cancer and association with advanced stage. Oncogene 11:1843–1851 Zhang SY, Klein-Szanto AJ, Sauter ER et al, 1994, Higher frequency of alterations in the p16/CDKN2 gene in squamous cell carcinoma cell lines than in primary tumors of the head and neck. Cancer Res 54: 5050–5053 Ohta M, Nagai H, Shimizu M et al, 1994, Rarity of somatic and germline mutations of the cyclin-dependent kinase 4 inhibitor gene, CDK4I, in melanoma. Cancer Res 54:5269–5272 Zhao Y, Zhang S, Fu B, Xiao C, 1999, Abnormalities of tumor suppressor genes P16 and P15 in primary maxillofacial squamous cell carcinomas. Cancer Genet Cytogenet 112:26–33 CasulaM, Colombino M, SattaMP et al, 2007, Factors predicting the occurrence of germline mutations in candidate genes among patients with cutaneous malignant melanoma from South Italy. Eur J Cancer 43:137–143 Jares P, Nadal A, Fernandez PL et al, 1999, Disregulation of p16MTS1/CDK4I protein and mRNA expression is associated with gene alterations in squamous-cell carcinoma of the larynx. Int J Cancer 81:705–711 Agarwalla PK, Dunn IF, Turner CD et al, 2008, A novel TP53 germline mutation in a family with a history of multiple malignancies: case report and review of the literature. Pediatr Neurosurg 44:501–508 Goi K, Takagi M, Iwata S et al, 1997, DNA damage-associated dysregulation of the cell cycle and apoptosis control in cells with germ-line p53 mutation. Cancer Res 57:1895–1902 Varley JM, McGown G, Thorncroft M et al, 1997, Germ-line mutations of TP53 in Li-Fraumeni families: an extended study of 39 families. Cancer Res 57:3245–3252 Wolf P, Kreimer-Erlacher H, Seidl H et al, 2004, The ultraviolet fingerprint dominates the mutational spectrum of the p53 and Ha-ras genes in psoralen+ultraviolet A keratoses from psoriasis patients. J Invest Dermatol 122:190–200 Kyritsis APP, Bondy MLL, Xiao M et al, 1994, Germline p53 gene mutations in subsets of glioma patients. J Natl Cancer Inst 86:344–349 Salani R, Kurman RJ, Giuntoli R et al, 2008, Assessment of TP53 mutation using purified tissue samples of ovarian serous carcinomas reveals a higher mutation rate than previously reported and does not correlate with drug resistance. Int J Gynecol Cancer 18:487–491 Oda H, Nakatsuru Y, Imai Y, Sugimura H, Ishikawa T, 1995, A mutational hot spot in the p53 gene is associated with hepatoblastomas. Int J Cancer 60:786–790 Chen C-H, Dickman KG, Moriya M et al, 2012, Aristolochic acidassociated urothelial cancer in Taiwan. PNAS 109:8241–8246 Sheu JC, Huang GT, Lee PH et al, 1992, Mutation of p53 gene in hepatocellular carcinoma in Taiwan. Cancer Res 52: 6098–6100 McIntyre JFF, Smith-Sorensen B, Friend SHH et al, 1994, Germline mutations of the p53 tumor suppressor gene in children with osteosarcoma. J Clin Oncol 12:925–930 Gleeson CMM, Sloan JMM, McGuigan JAA, Ritchie AJJ, Russell SEE, 1995, Base transitions at CpG dinucleotides in the p53 gene are common in esophageal adenocarcinoma. Cancer Res 55:3406–3411 Imazeki F, Omata M, Nose H, Ohto M, Isono K, 1992, p53 gene mutations in gastric and esophageal cancers. Gastroenterology 103: 892–896 Cabelguenne A, Blons H, de Waziers I et al, 2000, p53 alterations predict tumor response to neoadjuvant chemotherapy in head and neck squamous cell carcinoma: a prospective series. J Clin Oncol 18: 1465–1473 Frebourg T, Barbier N, Yan YXX et al, 1995, Germ-line p53 mutations in 15 families with Li-Fraumeni syndrome. Am J Hum Genet 56:608–615 Andersen TI, Holm R, Nesland JM et al, 1993, Prognostic significance of TP53 alterations in breast carcinoma. Br J Cancer 68:540–548 Matsushita K, Kobayashi S, KatoMet al, 1996, Reduced messenger RNA expression level of p21 CIP1 in human colorectal carcinoma tissues and its association with p53 gene mutation. Int J Cancer 69: 259–264 Horio Y, Suzuki H, Ueda R et al, 1994, Predominantly tumor-limited expression of a mutant allele in a Japanese family carrying a germline p53 mutation. Oncogene 9:1231–1235 James CDD, Galanis E, Frederick L et al, 1999, Tumor suppressor gene alterations in malignant gliomas: histopathological associations and prognostic evaluation. Int J Oncol 15: 547–553 Birch JMM, Hartley ALL, Tricker KJJ et al, 1994, Prevalence and diversity of constitutional mutations in the p53 gene among 21 Li- Fraumeni families. Cancer Res 54:1298–1304 Tenti P, Pavanello S, Padovan L et al, 1998, Analysis and clinical implications of p53 gene mutations and human papillomavirus type 16 and 18 infection in primary adenocarcinoma of the uterine cervix. Am J Pathol 152:1057–1063 Diller L, Sexsmith E, Gottlieb A, Li FP, Malkin D, 1995, Germline p53 mutations are frequently detected in young children with rhabdomyosarcoma. J Clin Invest 95:1606–1611 McGregor JM, Berkhout RJ, Rozycka M et al, 1997, p53 mutations implicate sunlight in post-transplant skin cancer irrespective of human papillomavirus status. Oncogene 15: 1737–1740 Naito M, Satake M, Sakai E et al, 1992, Detection of p53 gene mutations in human ovarian and endometrial cancers by polymerase chain reaction single strand conformation polymorphism analysis. Japan J Cancer Res 83:1030–1036 Meinhold-Heerlein I, Ninci E, Ikenberg H et al, 2001, Evaluation of methods to detect p53 mutations in ovarian cancer. Oncology 60: 176–188 Mok CH, Tsao SW, Knapp RC, Fishbaugh PM, Lau CC, 1992, Unifocal origin of advanced human epithelial ovarian cancers. Cancer Res 52:5119–5122 Malkin D, Li FPP, Strong LCC et al, 1990, Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science 250:1233–1238 Milner BJJ, Allan LAA, Eccles DMM et al, 1993, p53 mutation is a common genetic event in ovarian carcinoma. Cancer Res 53: 2128–2132 Chompret A, Brugieres L, Ronsin M et al, 2000, P53 germline mutations in childhood cancers and cancer risk for carrier individuals. Br J Cancer 82:1932–1937 Trkova M, Prochazkova K, Krutilkova V, Sumerauer D, Sedlacek Z, 2007, Telomere length in peripheral blood cells of germline TP53 mutation carriers is shorter than that of normal individuals of corresponding age. Cancer 110:694–702 Toguchida J, Yamaguchi T, Dayton SH et al, 1992, Prevalence and spectrum of germline mutations of the p53 gene among patients with sarcoma. N Engl J Med 326:1301–1308 Shibata A, Parsonnet J, Longacre TA et al, 2002, CagA status of Helicobacter pylori infection and p53 gene mutations in gastric adenocarcinoma. Carcinogenesis 23:419–424 Pang A, Ng IOO, Fan STT, Kwong YLL, 2003, Clinicopathologic significance of genetic alterations in hepatocellular carcinoma. Cancer Genet Cytogenet 146:8–15 Boyle JO, Hakim J, Koch W et al, 1993, The incidence of p53 mutations increases with progression of head and neck cancer. Cancer Res 53:4477–4480 Nishida N, Fukuda Y, Kokuryu H et al, 1993, Role and mutational heterogeneity of the p53 gene in hepatocellular carcinoma. Cancer Res 53:368–372 Hwang S-J, Lozano G, Amos CI, Strong LC, 2003, Germline p53 mutations in a cohort with childhood sarcoma: sex differences in cancer risk. Am J Hum Genet 72:975–983 Kiwerska K, Rydzanicz M, Kram A et al, 2010, Mutational analysis of CDKN2A gene in a group of 390 larynx cancer patients.Mol Biol Rep 37:325–332 Gleich LL, Salamone FN, 2002, Molecular genetics of head and neck cancer. Cancer Control 9:369–378 Agrawal N, FrederickMJ, Pickering CR et al, 2011, Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 333:1154–1157 Smigiel R, Sasiadek M, Krecicki T et al, 2004, Inactivation of the cyclin-dependent kinase inhibitor 2A, CDKN2A, gene in squamous cell carcinoma of the larynx. Mol Carcinog 39: 147–154 Soussi T, 2007, p53 alterations in human cancer:more questions than answers. Oncogene 26:2145–2156 Cho Y, Gorina S, Jeffrey PD, Pavletich NP, 1994, Crystal structure of a p53 tumor suppressor-DNA complex: understanding tumorigenic mutations. Science 265:346–355 Bosch FX, Ritter D, Enders C et al, 2004, Head and neck tumor sites differ in prevalence and spectrum of p53 alterations but these have limited prognostic value. Int J Cancer 111:530–538 Partridge M, Costea DE, Huang X, 2007, The changing face of p53 in head and neck cancer. Int J Oral Maxillofac Surg 36:1123–1138 Poeta ML, Manola J, Goldwasser MA et al, 2007, TP53 mutations and survival in squamous-cell carcinoma of the head and neck. N Engl J Med 357:2552–2561 Van Houten VMM, Tabor MP, van den Brekel MWM et al, 2002, Mutated p53 as a molecular marker for the diagnosis of head and neck cancer. J Pathol 198:476–486 Balz V, Scheckenbach K, Gotte K et al, 2003, Is the p53 inactivation frequency in squamous cell carcinomas of the head and neck underestimated? Analysis of p53 exons 2–11 and human papillomavirus 16/18 E6 transcripts in 123 unselected tumor specimens. Cancer Res 63:1188–1191 Tabor MP, Brakenhoff RH, van Houten VM et al, 2001, Persistence of genetically altered fields in head and neck cancer patients: biological and clinical implications. Clin Cancer Res 7:1523–1532 Brennan JA, Boyle JO, Koch WM et al, 1995, Association between cigarette smoking and mutation of the p53 gene in squamous-cell carcinoma of the head and neck. N Engl J Med 332:712–717 Blons H, Laurent-Puig P, 2003, TP53 and head and neck neoplasms. Hum Mutat 21:252–257 Kress S, Sutter C, Strickland PT et al, 1992, Carcinogenspecific mutational pattern in the p53 gene in ultraviolet B radiation-induced squamous cell carcinomas of mouse skin. Cancer Res 52:6400–6403 Paget V, Lechevrel M, Andre V et al, 2012, Benzo[a]pyrene, aflatoxine B1 and acetaldehyde mutational patterns in TP53 gene using a functional assay: relevance to human cancer aetiology. PLoS One 7:e30921 Urashima M, Hama T, Suda T et al, 2013, Distinct effects of alcohol consumption and smoking on genetic alterations in head and neck carcinoma. PLoS One 8:e80828 Sigal A, Rotter V, 2000, Oncogenic mutations of the p53 tumor suppressor: the demons of the guardian of the genome. Cancer Res 60:6788–6793 Bullock AN, Henckel J, DeDecker BS et al, 1997, Thermodynamic stability of wild-type and mutant p53 core domain. PNAS 94:14338– 14342 Suzuki HI, Yamagata K, Sugimoto K et al, 2009, Modulation of microRNA processing by p53. Nature 460: 529–533 Hermeking H, 2012, MicroRNAs in the p53 network: micromanagement of tumour suppression. Nat Rev Cancer 12:613–626 Vogelstein B, Lane D, Levine AJ, 2000, Surfing the p53 network. Nature 408:307–310 Hermeking H, 2007, p53 enters the microRNA world. Cancer Cell 12:414–418 Hermeking H, 2010, The miR-34 family in cancer and apoptosis. Cell Death Differ 17:193–199 MenendezD, IngaA, ResnickMA(2009, The expanding universe of p53 targets. Nat Rev Cancer 9:724–737 Vousden KH, Ryan KM(2009, p53 andmetabolism. Nat Rev Cancer 9:691–700 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 413 EP 421 DI 10.1007/s12253-014-9836-0 PG 9 ER PT J AU Podmaniczky, E Fabian, K Papay, J Puskas, R Gyulai, M Furak, J Tiszlavicz, L Losonczy, Gy Timar, J Moldvay, J AF Podmaniczky, Eszter Fabian, Katalin Papay, Judit Puskas, Rita Gyulai, Marton Furak, Jozsef Tiszlavicz, Laszlo Losonczy, Gyorgy Timar, Jozsef Moldvay, Judit TI Decreased ERCC1 Expression After Platinum-Based Neoadjuvant Chemotherapy in non-Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ERCC1; Lung cancer; Platinumbased chemotherapy; Immunohistochemistry; Gender differences ID ERCC1; Lung cancer; Platinumbased chemotherapy; Immunohistochemistry; Gender differences AB We have already demonstrated in a small cohort of 17 non-small cell lung cancer patients that ERCC1 (excision repair cross-complementation group 1) protein expression decreased after platinum-based treatment, however, certain clinicopathological parameters, such as histologic subtypes, ERCC1 expression scores, chemotherapeutic combinations, response rate, gender and smoking history were not analyzed. The aim of our present study was to extend the studied cohort and analyze those parameters. ERCC1 protein expression was examined in 46 patients treated with neoadjuvant chemotherapy. 46 bronchoscopic biopsy samples (27 squamous cell carcinomas /SCC/ and 19 adenocarcinomas /ADC/) together with their corresponding surgical biopsies were studied. ERCC1 immunohistochemistry was performed on formalinfixed, paraffin-embedded tissues. Staining scores were calculated by multiplying the percentage of positive tumor cells (0–100) by the staining intensity (0–3). 24/27 bronchoscopic SCC tissues expressed ERCC1. Thirteen of these cases became negative after neoadjuvant therapy and the expression differences between pre- and postchemotherapy samples were highly significant (p<0.001). 11/19 bronchoscopic ADC tissues expressed ERCC1. Six of these cases became negative after neoadjuvant therapy and the expression differences were significant (p<0.010). There was no newly expressed ERCC1 postoperatively. Comparison of staining score changes revealed more pronounced decrease in SCC (p=0.032). We observed no correlation between initial ERCC1 level or ERCC1 decrease and overall survival, but we demonstrated correlations between decrease in ERCC1 expression and histologic subtypes of tumors and gender. We could confirm our previous data in a larger cohort that platinum-based chemotherapy affects the ERCC1 expression probably referring to an induction of tumor cell selection. C1 [Podmaniczky, Eszter] Semmelweis University, Department of Pulmonology, Diosarok u. 1/c, H-1125 Budapest, Hungary. [Fabian, Katalin] Semmelweis University, Department of Pulmonology, Diosarok u. 1/c, H-1125 Budapest, Hungary. [Papay, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Puskas, Rita] Semmelweis University, Department of Pulmonology, Diosarok u. 1/c, H-1125 Budapest, Hungary. [Gyulai, Marton] County Hospital of PulmonologyTorokbalint, Hungary. [Furak, Jozsef] Szegedi Tudomanyegyetem, AOK, Mellkassebeszeti TanszekSzeged, Hungary. [Tiszlavicz, Laszlo] University of Szeged, Department of PathologySzeged, Hungary. [Losonczy, Gyorgy] Semmelweis University, Department of Pulmonology, Diosarok u. 1/c, H-1125 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Moldvay, Judit] Semmelweis University, Department of Pulmonology, Diosarok u. 1/c, H-1125 Budapest, Hungary. RP Fabian, K (reprint author), Semmelweis University, Department of Pulmonology, H-1125 Budapest, Hungary. EM drfabian.katalin@gmail.com CR Ferlay J, Shin HR, Bray F, Forman D,Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127(12):2893–2917., DOI 10.1002/ijc.25516 Carbone DP, Felip E, 2011, Adjuvant therapy in non-small cell lung cancer: future treatment prospects and paradigms. Clin Lung Cancer 12(5):261–271., DOI 10.1016/j.cllc.2011.06.002 Le Chevalier T, 2010, Adjuvant chemotherapy for resectable nonsmall- cell lung cancer:where is it going?AnnOncol 21(7):196–-198., DOI 10.1093/annonc/mdq376 SongWA, Zhou NK,WangW, Chu XY, Liang CY, Tian XD,Guo JT, Liu X, Liu Y, Dai WM, 2010, Survival benefit of neoadjuvant chemotherapy in non-small cell lung cancer: an updated metaanalysis of 13 randomized control trials. J Thorac Oncol 5(4):510– 516., DOI 10.1097/JTO.0b013e3181cd3345 Mouillet G,Monnet E, Milleron B, Puyraveau M, Quoix E, David P, Ducolone A, Molinier O, Zalcman G, Depierre A,Westeel V, 2012, Pathologic complete response to preoperative chemotherapy predicts cure in early-stage non-small-cell lung cancer: combined analysis of two IFCT randomized trials. J Thorac Oncol 7(5):841–849., DOI 10. 1097/JTO.0b013e31824c7d92 Zheng Z, Chen T, Li X, Haura E, Sharma A, Bepler G, 2007, DNA synthesis and repair genes RRM1 and ERCC1 in lung cancer. N Engl J Med 356(8):800–808., DOI 10.1056/NEJMoa065411 Kang CH, Jang BG, Kim DW, Chung DH, Kim YT, Jheon S, Sung SW, Kim JH, 2010, The prognostic significance of ERCC1, BRCA1, XRCC1, and betaIII-tubulin expression in patients with non-small cell lung cancer treated by platinum- and taxane-based neoadjuvant chemotherapy and surgical resection. Lung Cancer 68(3):478–483., DOI 10.1016/j.lungcan.2009.07.004 Soria JC, 2007, ERCC1-tailored chemotherapy in lung cancer: the first prospective randomized trial. J Clin Oncol 25(19):2648–2649., DOI 10.1200/jco.2007.11.3167 Papay J, Sapi Z, Egri G, Gyulai M, Szende B, Losonczy G, Timar J, Moldvay J, 2009, Platinum-based chemotherapy in lung cancer affects the expression of certain biomarkers including ERCC1. Pathol Oncol Res 15(3):445–450., DOI 10.1007/s12253-009-9155-z Rami-Porta R, Crowley JJ, Goldstraw P, 2009, The revised TNM staging system for lung cancer. Ann Thorac Cardiovasc Surg 15(1): 4–9 Kerr KM, 2012, Personalized medicine for lung cancer: new challenges for pathology. Histopathology 60(4):531–546., DOI 10.1111/j. 1365-2559.2011.03854.x Pomerri F, Pucciarelli S, Gennaro G, Maretto I, Nitti D, Muzzio PC, 2012, Comparison between CT volumemeasurement and histopathological assessment of response to neoadjuvant therapy in rectal cancer. Eur J Radiol 81(12):3918–3924., DOI 10.1016/j.ejrad.2012. 04.038 Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J, 2009, New response evaluation criteria in solid tumours: revised RECIST guideline, version 1.1). Eur J Cancer 45(2):228–247., DOI 10.1016/j. ejca.2008.10.026 NishinoM, Jackman DM, Hatabu H, Yeap BY, Cioffredi LA, Yap JT, Janne PA, Johnson BE, Van den Abbeele AD, 2010, New response evaluation criteria in solid tumors, RECIST, guidelines for advanced non-small cell lung cancer: comparison with original RECIST and impact on assessment of tumor response to targeted therapy. AJR Am J Roentgenol 195(3):W221–228., DOI 10.2214/ajr.09.3928 Lee HY, Lee KS, AhnMJ, Hwang HS, Lee JW, Park K, Ahn JS, Kim TS, Yi CA, ChungMJ, 2011, New CT response criteria in non-small cell lung cancer: proposal and application in EGFR tyrosine kinase inhibitor therapy. Lung Cancer 73(1):63–69., DOI 10.1016/j.lungcan. 2010.10.019 Burger IA, Schwarz EI, Samarin A, Breitenstein S, Weber A, Hany TF, 2013, Correlation between therapy response assessment using FDG PET/CT and histopathologic tumor regression grade in hepatic metastasis of colorectal carcinoma after neoadjuvant therapy. Ann Nucl Med 27(2):177–183., DOI 10.1007/s12149-012-0670-8 Pataer A, Kalhor N, Correa AM, Raso MG, Erasmus JJ, Kim ES, Behrens C, Lee JJ, Roth JA, Stewart DJ, Vaporciyan AA,Wistuba II, Swisher SG, 2012, Histopathologic response criteria predict survival of patients with resected lung cancer after neoadjuvant chemotherapy. J Thorac Oncol 7(5):825–832., DOI 10.1097/JTO.0b013e318247504a Sun Y, Campisi J, Higano C, Beer TM, Porter P, Coleman I, True L, Nelson PS, 2012, Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat Med 18(9):1359–1368., DOI 10.1038/nm.2890 Xiang F, Yu W, Shen Y, Wu C, Wang Y, 2007, Effects of neoadjuvant chemotherapy on the quantitative expression of P-gp, LRP, MRP, GST-pi in NSCLC and its clinical significance. Zhongguo Fei Ai Za Zhi 10(5):398–405., DOI 10.3779/j.issn.1009-3419.2007. 05.11 Liu YP, Yang CJ, Huang MS, Yeh CT,Wu AT, Lee YC, Lai TC, Lee CH, Hsiao YW, Lu J, Shen CN, Lu PJ, Hsiao M, 2013, Cisplatin selects for multidrug-resistant CD133+ cells in lung adenocarcinoma by activating Notch signaling. Cancer Res 73(1):406–416., DOI 10. 1158/0008-5472.can-12-1733 Meert AP, Martin B, Verdebout JM, Paesmans M, Berghmans T, Ninane V, Sculier JP, 2004, Correlation of different markers, p53, EGF-R, c-erbB-2, Ki-67, expression in the diagnostic biopsies and the corresponding resected tumors in non-small cell lung cancer. Lung Cancer 44(3):295–301., DOI 10.1016/j.lungcan.2003.12.009 Choi CM, Yang SC, Jo HJ, Song SY, Jeon YJ, Jang TW, Kim DJ, Jang SH, Yang SH, Kim YD, Lee KH, Jang SJ, Kim YT, Kim DK, Chung DH, Kim L, Nam HS, Cho JH, Kim HJ, Ryu JS, 2012, Proteins involved in DNA damage response pathways and survival of stage I non-small-cell lung cancer patients. Ann Oncol 23(8): 2088–2093., DOI 10.1093/annonc/mdr606 Kang CH, Jang BG, Kim DW, Chung DH, Kim YT, Jheon S, Sung SW, Kim JH, 2009, Differences in the expression profiles of excision repair cross complementation group 1, x-ray repair cross complementation group 1, and betaIII-tubulin between primary non-small cell lung cancer and metastatic lymph nodes and the significance in mid-term survival. J Thorac Oncol 4(11):1307–1312., DOI 10.1097/ JTO.0b013e3181b9f236 Schneider S, Uchida K, Brabender J, Baldus SE, Yochim J, Danenberg KD, Salonga D, Chen P, Tsao-Wei D, Groshen S, Hoelscher AH, Schneider PM, Danenberg PV, 2005, Down regulation of TS, DPD, ERCC1, GST-Pi, EGFR, and HER2 gene expression after neoadjuvant three-modality treatment in patients with esophageal cancer. J Am Coll Surg 200(3):336–344., DOI 10.1016/j. jamcollsurg.2004.10.035 Xia Y, Hu C, Zhang M, Yang H, Zhou D, Liang S, 2007, Relationship between ERCC1 expression and cisplatin intervention in human lung adenocarcinoma cell lines. Zhongguo Fei Ai Za Zhi 10(5):362–365., DOI 10.3779/j.issn.1009-3419.2007.05.03 Baba H, Watanabe M, Okabe H, Miyamoto Y, Sakamoto Y, Baba Y, IwatsukiM, Chikamoto A, Beppu T, 2012, Up regulation of ERCC1 and DPD expressions after oxaliplatin-based first-line chemotherapy formetastatic colorectal cancer. Br J Cancer 107(12):1950–1955., DOI 10.1038/bjc.2012.502 Olaussen KA, Dunant A, Fouret P, Brambilla E, Andre F, Haddad V, Taranchon E, Filipits M, Pirker R, Popper HH, Stahel R, Sabatier L, Pignon JP, Tursz T, Le Chevalier T, Soria JC, 2006, DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N Engl J Med 355(10):983–91 Schneider JG1, Farhadfar N, Sivapiragasam A, et al. Commercial Laboratory Testing of Excision Repair Cross-Complementation Group 1 Expression in Non-Small Cell Lung Cancer. Oncologist. 2014 Apr 4. [Epub ahead of print] Pierceall WE, Olaussen KA, Rousseau V, Brambilla E, Sprott KM, Andre F, Pignon JP, Le Chevalier T, Pirker R, Jiang C, Filipits M, Chen Y, Kutok JL, Weaver DT, Ward BE, Soria JC, 2012, Cisplatin benefit is predicted by immunohistochemical analysis of DNA repair proteins in squamous cell carcinoma but not adenocarcinoma: theranostic modeling by NSCLC constituent histological subclasses. Ann Oncol 23(9):2245–52., DOI 10.1093/annonc/mdr624 Olaussen KA, Adam J, Vanhecke E, Vielh P, Pirker R, Friboulet L, Popper H, Robin A, Commo F, Thomale J, Kayitalire L, Filipits M, Le Chevalier T,Andre F, Brambilla E, Soria JC, 2013, PARP1 impact on DNA repair of platinum adducts: preclinical and clinical readouts. Lung Cancer 80(2):216–22., DOI 10.1016/j.lungcan.2013.01. 014 Yoon YK, Im SA,Min A, Kim HP, Hur HS, Lee KH, Han SW, Song SH, Youn Oh D, Kim TY, Kim WH, Bang YJ, 2012, Sunitinib synergizes the antitumor effect of cisplatin via modulation of ERCC1 expression in models of gastric cancer. Cancer Lett 321(2): 128–136., DOI 10.1016/j.canlet.2012.01.019 Guarneri V, DieciMV, Barbieri E, Piacentini F, Omarini C, Ficarra G, Bettelli S, Conte PF, 2013, Loss of HER2 positivity and prognosis after neoadjuvant therapy in HER2-positive breast cancer patients. Ann Oncol., DOI 10.1093/annonc/mdt364 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 423 EP 431 DI 10.1007/s12253-014-9839-x PG 9 ER PT J AU Saadat, M Saadat, Sh AF Saadat, Mostafa Saadat, Shekoofeh TI Genetic Polymorphism of CAT C-262 T and Susceptibility to Breast Cancer, a Case–Control Study and Meta-Analysis of the Literatures SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; CAT; Genetic polymorphism; Meta-analysis ID Breast cancer; CAT; Genetic polymorphism; Meta-analysis AB Catalase (CAT) activity is likely to be affected by functional polymorphism of C-262 T (rs1001179) in the CAT gene (OMIM: 115500). It is hypothesized that individuals with the lower expressing forms of the CAT polymorphism may be more susceptible to breast cancer. Therefore, the present case–control study and meta-analysis were carried out. The present case–control study consisted of 407 females with breast cancer and a total of 395 healthy female from population matched with patients according to age. Genotypic analysis for the CAT C-262 T polymorphism was determined by PCR. We identified 7 eligible studies, including 10,471 subjects (4,959 patients, and 5,512 healthy controls) in relation to the CAT C-262 T polymorphism and breast cancer risk. Based on the present case–control study, the CT (OR=0.90, 95 % CI: 0.66–1.22, P=0.484) and TT (OR=0.68, 95 % CI: 0.35–1.30, P=0.245) genotypes were not associated with breast cancer risk compared to the CC genotype. For metaanalysis including all studies, there was significant heterogeneity between studies. The overall ORs of the breast cancer risk were not associatedwith the CT (Q-statistic=14.90, df=6, P<0.05; OR=1.01, 95 % CI: 0.92–1.09, P=0.862) and TT (Q-statistic=2.57, df=6, P>0.05; OR=1.03, 95 % CI: 0.85– 1.24, P=0.770) genotypes. There was no association between C-262 T polymorphism of the CAT and risk of breast cancer. C1 [Saadat, Mostafa] Shiraz University, College of Sciences, Department of Biology, 71454 Shiraz, Iran. [Saadat, Shekoofeh] Shiraz University, College of Sciences, Department of Biology, 71454 Shiraz, Iran. RP Saadat, M (reprint author), Shiraz University, College of Sciences, Department of Biology, 71454 Shiraz, Iran. EM saadat@shirazu.ac.ir;msaadat41@yahoo.com CR Klaunig JE, Kamendulis LM, 2004, The role of oxidative stress in carcinogenesis. Annu Rev Pharmacol Toxicol 44:239–267 Kang DH, 2002, Oxidative stress, DNA damage, and breast cancer. AACN Clin Issues 13:540–549 Ambrosone CB, 2000, Oxidants and antioxidants in breast cancer. Antioxid Redox Signal 2:903–917 Emerit I, 1994, Reactive oxygen species, chromosome mutation, and cancer: possible role of clastogenic factors in carcinogenesis. Free Radic Biol Med 16:99–109 Loft S, Olsen A, Moller P, Poulsen HE, Tjonneland A, 2013, Association between 8-oxo-7,8-dihydro-2′-deoxyguanosine excretion and risk of postmenopausal breast cancer: nested case–control study. Cancer Epidemiol Biomarkers Prev 22:1289–1296 Chang D, Wang F, Zhao YS, Pan HZ, 2008, Evaluation of oxygen stress in colorectal cancer patients. Biomed Environ Sci 21:286–289 Ahn J, Nowell S,McCann SE, Yu J, Carter L, Lang NP, Kadlubar FF, Ratnasinghe LD, Ambrosone CB, 2006, Associations between catalase phenotype and genotype: modification by epidemiologic factors. Cancer Epidemiol Biomarkers Prev 15:1217–1222 Nadif R, Mintz M, Jedlicka A, Bertrand JP, Kleeberger SR, Kauffmann F, 2005, Association of CAT polymorphisms with catalase activity and exposure to environmental oxidative stimuli. Free Radic Res 39:1345–1350 Ahn J, Gammon MD, Santella RM, Gaudet MM, Britton JA, Teitelbaum SL, Terry MB, Nowell S, Davis W, Garza C, Neugut AI, Ambrosone CB, 2005, Associations between breast cancer risk and the catalase genotype, fruit and vegetable consumption, and supplement use. Am J Epidemiol 162:943–952 Cebrian A, Pharoah PD, Ahmed S, Smith PL, Luccarini C, Luben R, Redman K,Munday H, Easton DF, Dunning AM, Ponder BA, 2006, Tagging single-nucleotide polymorphisms in antioxidant defense enzymes and susceptibility to breast cancer. Cancer Res 66:1225– 1233 Quick SK, Shields PG, Nie J, Platek ME, McCann SE, Hutson AD, Trevisan M, Vito D, Modali R, Lehman TA, Seddon M, Edge SB, Marian C, Muti P, Freudenheim JL, 2008, Effect modification by catalase genotype suggests a role for oxidative stress in the association of hormone replacement therapy with postmenopausal breast cancer risk. Cancer Epidemiol Biomarkers Prev 17:1082–1087 Li Y, Ambrosone CB, McCullough MJ, Ahn J, Stevens VL, Thun MJ, Hong CC, 2009, Oxidative stress-related genotypes, fruit and vegetable consumption and breast cancer risk. Carcinog 30:777–784 Tsai S,Wu S, HouM, Chen Y,Ma H, Tsai L, 2012, Oxidative stressrelated enzyme gene polymorphisms and susceptibility to breast cancer in non-smoking, non-alcohol consuming Taiwanese women: a case–control study. Ann Clin Biochem 49:152–158 McCullough LE, SantellaRM, Cleveland RJ, Bradshaw PT, Millikan RC, North KE, Olshan AF, Eng SM, Ambrosone CB, Ahn J, Steck SE, Teitelbaum SL, Neugut AI, Gammon MD, 2012, Polymorphisms in oxidative stress genes, physical activity, and breast cancer risk. Cancer Causes Control 23:1949–1958 Abu-Amero KK, Azad TA, Mousa A, Osman EA, Sultan T, Al- Obeidan SA, 2013, A catalase promoter variant rs1001179 is associated with visual acuity but not with primary angle closure glaucoma in Saudi patients. BMC Med Genet 14:84 Babusikova E, JesenakM, EvinovaA, Banovcin P, Dobrota D, 2013, Frequency of polymorphism -262 C/T in catalase gene and oxidative damage in Slovak children with bronchial asthma. Arch Bronconeumol 49:507–512 Tefik T, Kucukgergin C, Sanli O, Oktar T, Seckin S, Ozsoy C, 2013, Manganese superoxide dismutase Ile58Thr, catalase C-262T and myeloperoxidase G-463A gene polymorphisms in patients with prostate cancer: relation to advanced and metastatic disease. BJU Int 112: E406–E414 Khodayari S, Salehi Z, Fakhrieh Asl S, Aminian K, Mirzaei-Gisomi N, Torabi-Dalivandan S, 2013, Catalase gene C-262T polymorphism: importance in ulcerative colitis. J Gastroenterol Hepatol 28: 819–822 Karunasinghe N, Han DY, Goudie M, Zhu S, Bishop K, Wang A, Duan H, Lange K, Ko S, Medhora R, Kan ST, Masters J, Ferguson LR, 2012, Prostate disease risk factors among a New Zealand cohort. J Nutrigenet Nutrigenomics 5:339–351 Komina AV, Korostileva KA, Gyrylova SN, Belonogov RN, Ruksha TG, 2012, Interaction between single nucleotide polymorphism in catalase gene and catalase activity under the conditions of oxidative stress. Physiol Res 61:655–658 Chang D, Hu ZL, Zhang L, Zhao YS, Meng QH, Guan QB, Zhou J, Pan HZ, 2012, Association of catalase genotype with oxidative stress in the predication of colorectal cancer: modification by epidemiological factors. Biomed Environ Sci 25:156–162 Ghaly MS, Ghattas MH, Labib SM, 2012, Association of catalase gene polymorphisms with catalase activity and susceptibility to systemic lupus erythematosus in the Suez Canal area, Egypt. Lupus 21: 1244–1249 Goth L, Nagy T, Kosa Z, Fejes Z, Bhattoa HP, Paragh G, Kaplar M, 2012, Effects of rs769217 and rs1001179 polymorphisms of catalase gene on blood catalase, carbohydrate and lipid biomarkers in diabetes mellitus. Free Radic Res 46:1249–1257 Dutkiewicz G, Domanski L, Binczak-Kuleta A, Pawlik A, Safranow K, Ciechanowicz A, Dziedziejko V, Ciechanowski K, 2010, The association of -262C/T polymorphism in the catalase gene and delayed graft function of kidney allografts. Nephrol, Carlton, 15:587– 591 Mohamadynejad P, Saadat M, 2008, Genetic polymorphisms of XRCC1, at codons 194 and 399, in Shiraz population, Fars province, southern Iran). Mol Biol Rep 35:669–672 Rafiee L, Saadat I, SaadatM(2010, Glutathione S-transferase genetic polymorphisms, GSTM1, GSTT1 and GSTO2, in three Iranian populations. Mol Biol Rep 37:155–158 Zarbock R, Hendig D, Szliska C, Kleesiek K, Gotting C, 2007, Pseudoxanthoma elasticum: genetic variations in antioxidant genes are risk factors for early disease onset. Clin Chem 53:1734–1740 DerSimonian R, Laird N, 1986, Meta-analysis in clinical trials. Control Clin Trials 7:177–188 Mantel N, Haenszel W, 1959, Statistical aspects of the analysis of data from retrospective studies of disease. J Natl Cancer Inst 22:719– 748 Chang D, Hu ZL, Zhang L, Zhao YS, Meng QH, Guan QB, Zhou J, Pan HZ, 2012, Association of catalase genotype with oxidative stress in the prediction of colorectal cancer: Modification by epidemiological factors. Biomed Environ Sci 25:156–162 Komina AV, Korostileva KA, Gyrylova SN, Benlonogov RN, Ruksha TG, 2012, Interaction between single nucleotide polymorphism in catalase gene and catalase acticity under the conditions of oxidative etress. Physiol Res 61:655–658 SaadatM(2006, Genetic polymorphisms of glutathione S-transferase T1, GSTT1, and susceptibility to gastric cancer: a meta-analysis. Cancer Sci 97:505–509 Kiyohara C, TakayamaK,Nakanishi Y, 2006, Association of genetic polymorphisms in the base excision repair pathway with lung cancer risk: a meta-analysis. Lung Cancer 54:267–283 Saadat M, Ansari-Lari M, 2007, Genetic polymorphism of glutathione S-transferase T1, M1 and asthma, a meta-analysis of the literature. Pak J Biol Sci 10:4183–4189 SaadatM, Ansari-LariM(2009, Polymorphism of XRCC1, at codon 399, and susceptibility to breast cancer, a meta-analysis of the literatures. Breast Cancer Res Treat 115:137–144 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 433 EP 437 DI 10.1007/s12253-014-9840-4 PG 5 ER PT J AU Kusters-Vandevelde, VNH van Engen- van Grunsven, ACHI Coupland, ES Lake, LS Rijntjes, J Pfundt, R Kusters, B Wesseling, P Blokx, AMW Groenen, JTAP AF Kusters-Vandevelde, V N Heidi van Engen- van Grunsven, A C H Ilse Coupland, E Sarah Lake, L Sarah Rijntjes, Jos Pfundt, Rolph Kusters, Benno Wesseling, Pieter Blokx, A M Willeke Groenen, J T A Patricia TI Mutations in G Protein Encoding Genes and Chromosomal Alterations in Primary Leptomeningeal Melanocytic Neoplasms SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Leptomeningeal melanocytic neoplasm; Melanocytoma; Melanoma; Central nervous system; GNAQ; GNA11 ID Leptomeningeal melanocytic neoplasm; Melanocytoma; Melanoma; Central nervous system; GNAQ; GNA11 AB Limited data is available on the genetic features of primary leptomeningeal melanocytic neoplasms (LMNs). Similarities with uveal melanoma were recently suggested as both entities harbor oncogenic mutations in GNAQ and GNA11. Whether primary LMNs share additional genetic alterations with uveal melanoma including copy number variations is unknown. Twenty primary LMNs ranging from benign and intermediate-grade melanocytomas to melanomas were tested by direct sequencing for hotspot mutations in the genes GNA11, GNAQ, BRAF, NRAS and HRAS. Furthermore, the lesions were tested for copy number variations of chromosomes frequently present in uveal melanoma (1p, 3, 6 and 8q) by multiplex ligation-dependent probe amplification (MLPA). Genome-wide analyses of copy number alterations of two leptomeningeal melanocytic neoplasms were performed using the OncoScan SNP-array. GNAQQ209 mutations were present in eleven LMNs, while two of 20 cases carried a GNA11Q209 mutation. No BRAF, HRAS or NRAS hotspot mutations were detected. Monosomy 3 and gain of 8q were present in one leptomeningeal melanoma, and one intermediate-grade melanocytoma harbored a gain of chromosome 6. With MLPA, the melanocytomas did not show any further gross chromosomal variations. Our data shows that primary LMNs, like uveal melanoma, harbor oncogenic mutations in GNAQ and GNA11 but lack mutations in BRAF, NRAS and HRAS. This finding may help in the differential diagnosis between a primary LMN and a metastasis from a cutaneous melanoma to the central nervous system. Copy number variations in some aggressive LMNs resemble those present in uveal melanoma but their prognostic significance is unclear. C1 [Kusters-Vandevelde, V N Heidi] Canisius Wilhelmina Hospital, Department of Pathology, C66, 6500 GS Nijmegen, The Netherlands. [van Engen- van Grunsven, A C H Ilse] Radboud University Nijmegen, Medical Centre, Department of Pathology, 6500 HB Nijmegen, The Netherlands. [Coupland, E Sarah] University of Liverpool, Department of Molecular and Clinical Cancer Medicine, Pathology, Daulby Street, L69 3GA Liverpool, UK. [Lake, L Sarah] University of Liverpool, Department of Molecular and Clinical Cancer Medicine, Pathology, Daulby Street, L69 3GA Liverpool, UK. [Rijntjes, Jos] Radboud University Nijmegen, Medical Centre, Department of Pathology, 6500 HB Nijmegen, The Netherlands. [Pfundt, Rolph] Radboud University Medical Center, Department of Human Genetics, 6500 HB Nijmegen, The Netherlands. [Kusters, Benno] Radboud University Nijmegen, Medical Centre, Department of Pathology, 6500 HB Nijmegen, The Netherlands. [Wesseling, Pieter] Canisius Wilhelmina Hospital, Department of Pathology, C66, 6500 GS Nijmegen, The Netherlands. [Blokx, A M Willeke] Radboud University Nijmegen, Medical Centre, Department of Pathology, 6500 HB Nijmegen, The Netherlands. [Groenen, J T A Patricia] Radboud University Nijmegen, Medical Centre, Department of Pathology, 6500 HB Nijmegen, The Netherlands. RP Kusters-Vandevelde, VNH (reprint author), Canisius Wilhelmina Hospital, Department of Pathology, C66, 6500 GS Nijmegen, The Netherlands. EM h.kusters@cwz.nl CR Brat DJ, Perry A, 2007, Melanocytic lesions. In: Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, eds, WHO classification of tumours of the central nervous system. IARC, Lyon, pp 181–183 Bydon A, Gutierrez JA, Mahmood A, 2003, Meningeal melanocytoma: an aggressive course for a benign tumor. J Neurooncol 64:259–263 Liubinas SV, Maartens N, Drummond KJ, 2010, Primary melanocytic neoplasms of the central nervous system. J Clin Neurosci 17:1227–1232 Perrini P, Caniglia M, Pieroni M, Castagna M, Parenti GF, 2010, Malignant transformation of intramedullary melanocytoma: case report. Neurosurg 67:867–869 Uozumi Y, Kawano T, Kawaguchi T, KanekoY, Ooasa T, Ogasawara S, Yoshida H, Yoshida T, 2003, Malignant transformation of meningeal melanocytoma: a case report. Brain Tumor Pathol 20:21–5 Brat DJ, Giannini C, Scheithauer BW, Burger PC, 1999, Primary melanocytic neoplasms of the central nervous systems. Am J Surg Pathol 23:745–754 Arunkumar MJ, Ranjan A, Jacob M, Rajshekhar V, 2001, Neurocutaneous melanosis: a case of primary intracranial melanoma with metastasis. Clin Oncol, R Coll Radiol, 13:52–54 Koenigsmann M, Jautzke G, Unger M, Theallier-Janko A,Wiegel T, Stoltenburg-Didinger G, 2002, 57-year-old male with leptomeningeal and liver tumors. Brain Pathol 12:519–521 Kusters-Vandevelde HV, Klaasen A, Kusters B, Groenen PJ, van Engen-van Grunsven IA, van Dijk MR, Reifenberger G, Wesseling P, Blokx WA, 2009, Activating mutations of the GNAQ gene: a frequent event in primary melanocytic neoplasms of the central nervous system. Acta Neuropathol 119: 317–23 Gessi M, Hammes J, Lauriola L, Dorner E, Kirfel J, Kristiansen G, Zur Muehlen A, Denkhaus D, Waha A, Pietsch T, 2012, GNA11 and N-RAS mutations: alternatives for MAPK pathway activating GNAQ mutations in primary melanocytic tumours of the central nervous system. Neuropathol Appl Neurobiol 39:417– 25 Fuld AD, SpeckME, Harris BT, Simmons NE, Corless CL, Tsongalis GJ, Pastel DA, Hartford AC, Ernstoff MS, 2011, Primary melanoma of the spinal cord: a case report, molecular footprint, and review of the literature. J Clin Oncol 29:499–502 Onken MD, Worley LA, Long MD, Duan S, Council ML, Bowcock AM, Harbour JW, 2008, Oncogenic mutations in GNAQ occur early in uveal melanoma. Invest Ophthalmol Vis Sci 49:5230–5234 Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L, O'Brien JM, Simpson EM, Barsh GS, Bastian BC, 2009, Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature 457:599–602 Van Raamsdonk CD, Griewank KG, Crosby MB, Garrido MC, Vemula S, Wiesner T, Obenauf AC A, Wackernagel W, Green G, Bouvier N, SozenMM, Baimukanova G, Roy R, Heguy A, Dolgalev I et al, 2010, Mutations in GNA11 in Uveal Melanoma. N Engl J Med 363:2191–9 Lamba S, Felicioni L, Buttitta F, Bleeker FE, Malatesta S, Corbo V, Scarpa A, Rodolfo M, Knowles M, Frattini M, Marchetti A, Bardell A, 2009, Mutational profile of GNAQQ209 in human tumors. PLoS One 4:e6833 Hurowitz EH, Melnyk JM, Chen YJ, Kouros-Mehr H, Simon MI, Shizuya H, 2000, Genomic characterization of the human heterotrimeric G protein alpha, beta, and gamma subunit genes. DNA Res 7:111–120 Markby DW, Onrust R, Bourne HR, 1993, Separate GTP binding and GTPase activating domains of a G alpha subunit. Science 262: 1895–1901 Radhika V, Dhanasekaran N, 2001, Transforming G proteins. Oncogene 20:1607–1614 Murali R, Wiesner T, Rosenblum MK, Bastian BC, 2012, GNAQ and GNA11 mutations in melanocytomas of the central nervous system. Acta Neuropathol 123:457–9 Strom RG, Shvartsbeyn M, Rosenblum MK, Hameed MR, Nafa K, Mikolaenko I, Babu RP, 2012, Melanocytic tumor with GNA11 p.Q209L mutation mimicking a foramen magnum meningioma. Clin Neurol Neurosurg 114:1197–200 Damato B,Dopierala JA, Coupland SE, 2010, Genotypic profiling of 452 choroidal melanomas with multiplex ligation-dependent probe amplification. Clin Cancer Res 16:6083–6092 Prescher G, Bornfeld N, Hirche H, Horsthemke B, Jockel KH, Becher R, 1996, Prognostic implications of monosomy 3 in uveal melanoma. Lancet 347:1222–1225 van Dongen JJ, Langerak AW, Bruggemann M, Evans PA, Hummel M, Lavender FL, Delabesse E, Davi F, Schuuring E, Garcia-Sanz R, van Krieken JH, Droese J, Gonzalez D, Bastard C, White HEHE et al, 2003, Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cel l receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936. Leukemia 17:2257–2317 Wang Y, Carlton VE, Karlin-Neumann G, Sapolsky R, Zhang L, Moorhead M, Wang ZC, Richardson AL, Warren R, Walther A, Bondy M, Sahin A, Krahe R, Tuna M, Thompson PA et al, 2009, High quality copy number and genotype data from FFPE samples using molecular inversion probe, MIP, microarrays. BMC Med Genomics 2:8 Bauer J, Kilic E, Vaarwater J, Bastian BC, Garbe C, de Klein A, 2009, Oncogenic GNAQ mutations are not correlated with diseasefree survival in uveal melanoma. Br J Cancer 101:813–815 Koopmans AE, Vaarwater J, Paridaens D, Naus NC, Kilic E, de Klein A, 2013, Patient survival in uveal melanoma is not affected by oncogenic mutations in GNAQ and GNA11. Br J Cancer 109:493–496 Mitsiades N, Chew SA, He B, Riechardt AI, Karadedou T, Kotoula V, Poulaki V, 2011, Genotype-dependent sensitivity of uveal melanoma cell lines to inhibition of B-Raf, MEK, and Akt kinases: rationale for personalized therapy. Invest Ophthalmol Vis Sci 52: 7248–7255 Wu X, Zhu M, Fletcher JA, Giobbie-Hurder A, Hodi FS, 2012, The protein kinase C inhibitor enzastaurin exhibits antitumor activity against Uveal Melanoma. PLoS One 7:e29622 von Euw E, Atefi M, Attar N, Chu C, Zachariah S, Burgess BL,Mok S, Ng C,Wong DJ, Chmielowski B, Lichter DI, Koya RC,McCannel TA, Izmailova E, Ribas A, 2012, Antitumor effects of the investigational selective MEK inhibitor TAK733 against cutaneous and uveal melanoma cell lines. Mol Cancer 11:22 Khalili JS, Yu X, Wang J, Hayes BC, Davies MA, Lizee G, Esmaeli B, Woodman SE, 2012, Combination small molecule MEK and PI3K inhibition enhances uveal melanoma cell death in a mutant GNAQand GNA11-dependent manner. Clin Canc Res 18:4345–4355 Ho AL, Musi E, Ambrosini G, Nair JS, Deraje Vasudeva S, de Stanchina E, Schwartz GK, 2012, Impact of combined mTOR and MEK inhibition in uveal melanoma is driven by tumor genotype. PLoS One 7:e40439 Carjaval RD. Quevedo F. et al., 2013, Phase II study of selumetinib versus temozolomide in gnaq/gna11 mutation uveal melanoma. In: 2013 ASCO Annual Meeting Chen X.Wu Q. Tan L. Porter D. JagerMJ. C. Emery C. B.C. Bastian BC, 2013, Combined PKC and MEK inhibition in uveal melanoma with GNAQ and GNA11 mutations. Oncogene, DOI 10.1038/ onc.2013.418 Wang H, Zhang S,Wu C, Zhang Z, Qin T, 2013)Melanocytomas of the central nervous system: a clinicopathological and molecular study. Eur J Clin Invest 43:809–15 PedersenM,Kusters-VandeveldeHV, VirosA,Groenen PJ, Sanchez- Laorden B, Gilhuis JH, van Engen-van Grunsven IA, Renier W, Schieving J, Niculescu-Duvaz I, Springer CJ, Kusters B, Wesseling P, Blokx WA, Marais R, 2013, Primary melanoma of the CNS in children is driven by congenital expression of oncogenic NRAS in melanocytes. Cancer Discov 3:458–469 Kinsler VA, Thomas AC, IshidaM, Bulstrode NW, Loughlin S, Hing S, Chalker J, McKenzie K, Abu-Amero S, Slater O, Chanudet E, Palmer R, Morrogh D, Stanier P, Healy E, Sebire NJ, Moore GE, 2013, Multiple congenital melanocytic nevi and neurocutaneous melanosis are caused by postzygotic mutations in codon 61 of NRAS. J Invest Dermatol 133:2229–36 Bafaloukos D, Gogas H, 2004, The treatment of brain metastases in melanoma patients. Cancer Treat Rev 30:515–520 Greco Crasto S, Soffietti R, Bradac GB, Boldorini R, 2001, Primitive cerebral melanoma: case report and review of the literature. Surg Neurol 55:163–168 Theunissen P, Spincemaille G, Pannebakker M, Lambers J, 1993, Meningeal melanoma associated with nevus of Ota: case report and review. Clin Neuropathol 12:125–129 Rodriguez y Baena R, Gaetani P, Danova M, Bosi F, Zappoli F, 1992, Primary solitary intracranial melanoma: case report and review of the literature. Surgical Neurol 38:26–37 Skarli SO, Wolf AL, Kristt DA, Numaguch Y, 1994, Melanoma arising in a cervical spinal nerve root: report of a case with a benign course and malignant features. Neurosurg 34:533–537 Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J,Woffendin H, GarnettMJ, BottomleyW, Davis N, Dicks E, Ewing R, Floyd Y, Gray K et al, 2002, Mutations of the BRAF gene in human cancer. Nature 417:949–954 Broekaert SM, Roy R, Okamoto I, van den Oord J, Bauer J, Garbe C, Barnhill RL, Busam KJ, Cochran AJ, Cook MG, Elder DE, McCarthy SW, Mihm MC, Schadendorf D, Scolyer RA, Spatz A, Bastian BC, 2010, Genetic and morphologic features for melanoma classification. Pigment Cell Melanoma Res 23:763–770 van den Bosch BT, Kilic E, Paridaens D, de Klein A, 2010, Genetics of uveal melanoma and cutaneous melanoma: two of a kind? Dermatol Res Pract 360136 Parrella P, Sidransky D, Merbs SL, 1999, Allelotype of posterior uveal melanoma: implications for a bifurcated tumor progression pathway. Cancer Res 59:3032–3037 Cruz F, Rubin BP, Wilson D, Town A, Schroeder A, Haley A, Bainbridge T, Heinrich MC, Corless CL, 2003, Absence of BRAF and NRAS mutations in uveal melanoma. Cancer Res 63:5761–5766 Kilic E, Bruggenwirth HT, Verbiest MM, Zwarthoff EC, Mooy NM, Luyten GP, deKleinA, 2004, The RAS-BRAF kinase pathway is not involved in uveal melanoma. Melanoma Res 14:203–205 4 8 . Weber A, Hengge UR, Urbanik D, Markwart A, Mirmohammadsaegh A, Reichel MB, Wittekind C, Wiedemann P, Tannapfel A, 2003, Absence of mutations of the BRAF gene and constitutive activation of extracellular-regulated kinase in malignant melanomas of the uvea. Lab Invest 83:1771–1776 49. Janssen CS, Sibbett R, Henriquez FL, McKay IC, Kemp EG, Roberts F, 2008, The T1799A point mutation is present in posterior uveal melanoma. Br J Cancer 99:1673–1677 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 439 EP 447 DI 10.1007/s12253-014-9841-3 PG 9 ER PT J AU Dehghan, R Hosseinpour Feizi, AM Pouladi, N Babaei, E Montazeri, V Fakhrjoo, A Sedaei, A Azarfam, P Nemati, M AF Dehghan, Roghayeh Hosseinpour Feizi, Ali Mohammad Pouladi, Nasser Babaei, Esmaeil Montazeri, Vahid Fakhrjoo, Ashraf Sedaei, Ayda Azarfam, Parvin Nemati, Masoumeh TI Association of P53 (−16ins-Pro) Haplotype with the Decreased Risk of Differentiated Thyroid Carcinoma in Iranian-Azeri Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thyroid; Neoplasms; P53 tumor suppressor protein; Polymorphism; Haplotype; Molecular marker ID Thyroid; Neoplasms; P53 tumor suppressor protein; Polymorphism; Haplotype; Molecular marker AB Association of P53 polymorphisms with the increased risk of various cancers has been investigated in numerous studies. However, the results were conflicting and no polymorphism has been determined as a definite risk factor. It is likely that the study of P53 combined genotypes and haplotypes may be more useful than individual polymorphisms. Thus, in this study, we analyzed the associations of intron 3 Ins16bp and exon 4 Arg72Pro polymorphisms, aswell as their combined genotypes and haplotypes with the risk of differentiated thyroid carcinoma in Iranian-Azeri patients. This case– control study was performed on 84 Iranian Azeri patients with differentiated thyroid carcinoma and 150 healthy subjects. Intron 3 genotype was determined using PCR products analysis on polyacrylamide gels and AS-PCR was used for genotyping Arg72Pro polymorphism. The javastat online statistics package software and SHEsis program were applied for data analysis. There was no significant difference in genotype frequencies of both two polymorphisms between cases and controls. However, the (−16ins/−16ins) (Arg/Pro) genotype combination had a noticeable but not significant association with decreased risk of thyroid cancer development (OR= 0.497 95%CI: 0.209–1.168 P=0.080) and also the frequency of (−16ins-Pro) haplotype was significantly higher in controls rather than patients (OR=0.543 95%CI: 0.326–0.903 P=0.018). In our study, there was association between (−16ins-Pro) haplotype with decreased risk of differentiated thyroid carcinoma development in Iranian-Azeri patients. C1 [Dehghan, Roghayeh] Tabriz University, Department of BiologyTabriz, Iran. [Hosseinpour Feizi, Ali Mohammad] Tabriz University, Department of BiologyTabriz, Iran. [Pouladi, Nasser] Azarbaijan Shahid Madani University, Department of BiologyTabriz, Iran. [Babaei, Esmaeil] Tabriz University, Department of BiologyTabriz, Iran. [Montazeri, Vahid] Noor-E-Nejat Hospital, Department of Thorax SurgeryTabriz, Iran. [Fakhrjoo, Ashraf] Tabriz University of Medical Sciences, Department of PathologyTabriz, Iran. [Sedaei, Ayda] Tabriz University, Department of BiologyTabriz, Iran. [Azarfam, Parvin] Tabriz University, Department of BiologyTabriz, Iran. [Nemati, Masoumeh] Tabriz University, Department of BiologyTabriz, Iran. RP Hosseinpour Feizi, AM (reprint author), Tabriz University, Department of Biology, Tabriz, Iran. EM pourfeiz@eastp.ir CR Stegh AH, 2012, Targeting the P53 signaling pathway in cancer therapy—The promises, challenges, and perils. Expert Opin Ther Targets 16(1):67–83 Machado-Silva A, Perrier S, Bourdon JC, 2010, P53 family members in cancer diagnosis and treatment. Semin Cancer Biol 20:57–62 Whibley C, Pharoah PD, Hollstein M, 2009, P53 polymorphisms: cancer implications. Nat Rev Cancer 9:95–107 Wu X, Zhao H, Amos CI, Shete S, Makan N, Hong WK, Kadlubar FF, Spitz MR, 2002, P53 Genotypes and haplotypes associated with lung cancer susceptibility and ethnicity. J Natl Cancer Inst 94:681– 690 Gemignani F,Moreno V, Landi S, Moullan N, Chabrier A, Gutierrez- Enriquez S, Hall J, Guino E, Peinado MA, Capella G, Canzian F, 2004, A TP53 polymorphism is associated with increased risk of colorectal cancer and with reduced levels of TP53mRNA. Oncogene 23:1954–1956 FaghaniM,GhasemiFM, Nikhbakht M, SalehiM(2011, TP53 PIN3 polymorphism associated with breast cancer risk in Iranian women. Indian J Cancer 48(3):298–302 Hu Z, Li X, Qu X, He Y, Ring BZ, Song E, Su L, 2010, Intron 3 16 bp duplication polymorphism of TP53 contributes to cancer susceptibility: a meta-analysis. Carcinogenesis 31(4):643–647 Runnebaum IB, Tong XW, Konig R, Zhao H, Korner K, Atkinson EN, Kreienberg R, Kieback DG, Hong Z, 1995, P53- based blood test for P53PIN3 and risk for sporadic ovarian cancer. Lancet 1345(8955):994–994 Alawadi S, Ghabreau L, AlsalehM, Abdulaziz Z, RafeekM, Akil N, Alkhalaf M, 2011, 53 gene polymorphisms and breast cancer risk in Arab women. Med Oncol 28(3):709–715 Sagne C,MarcelV,AmadouA, Hainaut P, OlivierM, Hall J, 2013, A meta-analysis of cancer risk associated with the TP53 intron 3 duplication polymorphism, rs17878362): geographic and tumor-specific effects. Cell Death Dis 14(4):e492 Dumont P, Leu JI, Della Pietra AC, George DL 3rd, Murphy M, 2003, The codon 72 polymorphic variants of P53 have markedly different apoptotic potential. Nat Genet 33:357–365 Papadakis EN, Dokianakis DN, Spandidos DA, 2000, p53 codon 72 polymorphism as a risk factor in the development of breast cancer. Mol Cell Biol Res Commun 3:389–92 Brenna SM, Silva ID, Zeferino LC, Pereira JS, Martinez EZ, Syrjanen KJ, 2004, Prognostic value of P53 codon 72 polymorphism in invasive cervical cancer in Brazil. Gynecol Oncol 93:374–380 Matakidou A, Eisen T, Houlston RS, 2003, TP53 polymorphisms and lung cancer risk: a systematic review and meta-analysis. Mutagenesis 18:377–385 Hou J, Jiang Y, TangW, Jia S, 2013, P53 codon 72 polymorphism and breast cancer risk: a meta-analysis. Exp Ther Med 5(5):1397–1402 Sipos JA, Mazzaferri EL, 2010, Thyroid cancer epidemiology and prognostic variables. Clin Oncol 22:395–404 Legakis I, Syrigos K, 2011, Recent advances in molecular diagnosis of thyroid cancer. J Thyroid Res 2011:384213 Antico Arciuch VG, Russo MA, Dima M, Kang KS, Dasrath F, Liao XH, Refetoff S, Montagna C, Di Cristofano A, 2011, Thyrocytespecific inactivation of P53 and Pten results in anaplastic thyroid carcinomas faithfully recapitulating human tumors. Oncotarget 2(12):1109–26 Donghi R, Longoni A, Pilotti S, Michieli P, Della Porta G, Pierotti MA(1993, Gene P53 mutations are restricted to poorly differentiated and undifferentiated carcinomas of the thyroid gland. J Clin Invest 91(4):1753–60 Marcel V, Tran PL, Sagne C, Martel-Planche G, Vaslin L, Teulade- Fichou MP, Hall J, Mergny JL, Hainaut P, Van Dyck E, 2011, G-Gquadruplex structures in TP53 intron 3: role in alternative splicing and in production of p53 mRNA isoforms. Carcinogenesis 32(3): 271–8 Dai S,Mao C, Jiang L,Wang G, Cheng H, 2009, P53 polymorphism and lung cancer susceptibility: a pooled analysis of 32 case–control studies. Hum Genet 125:633–8 Soulitzis N, Sourvinos G,Dokianakis DN, Spandidos DA, 2002, p53 codon 72 polymorphism and its association with bladder cancer. Cancer Lett 179(2):175–83 de Souza LG, de Lima JM, da Silva IDCG, Forones NM, 2009, P53 Arg72Pro polymorphism in gastric cancer patients. J Gastrointest Cancer 40:41–5 Reis AA, Silva DM, CuradoMP, da Cruz AD, 2010, Involvement of CYP1A1, GST, 72TP53 polymorphisms in the pathogenesis of thyroid nodules. Genet Mol Res 9(4):2222–9 Aral C, Caglayan S, Ozisik G,Massoumilary S, Sonmez O, Akkiprik M, Baloglu H, OzataM, Ozer A, 2007, The association of P53 codon 72 polymorphism with thyroid cancer in turkish patients. Marmara Med J 20:1–5 Boltze C, Roessner A, Landt O, Szibor R, Peters B, Schneider-Stock R, 2002, Homozygous proline at codon 72 of p53 as a potential risk factor favoring the development of undiferentiated thyroid carcinoma. Int J Oncol 21:1151–1154 Wu B, Guo D, Guo Y, 2014, Association between p53 Arg72Pro polymorphism and thyroid cancer risk: a meta-analysis. Tumour Biol 35(1):561–5 Trifa F, Karray-Chouayekh S, Mabrouk I, Baccouche S, Khabir A, Sellami-Boudawara T, Gargouri A, Mokdad-Gargouri R, 2010, Haplotype analysis of P53 polymorphisms: Arg72Pro, Ins16bp and G13964C inTunisian patients with familial or sporadic breast cancer. Cancer Epidemiol 34:184–188 Patel KR, Vajaria BN, Begum R, Shah FD, Patel JB, Shukla SN, Patel PS, 2013, Association between p53 gene variants and oral cancer susceptibility in population from Gujarat, west India. Asian Pac J Cancer Prev 14(2):1093–1100 Costa S, Pinto D, Pereira D, Rodrigues H, Cameselle-Teijeiro J, Medeiros R, Schmitt F, 2008, Importance of TP53 codon 72 and intron 3 duplication 16 bp polymorphisms in prediction of susceptibility on breast cancer. BMC Cancer 8:32 Buyru N, Altinisik J, Demokan S, DalayN, 2007, P53 genotypes and haplotypes associated with risk of breast cancer. Cancer Detect Prev 31:207–13 Sjalander A, Birgander R, Hallmans G, Cajander S, Lenner P, Athlin L, Beckman G, Beckman L, 1996, p53 polymorphisms and haplotypes in breast cancer. Carcinogenesis 17(6):1313– 1316 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 449 EP 454 DI 10.1007/s12253-014-9846-y PG 6 ER PT J AU Pan, W Li, G Yang, X Miao, J AF Pan, Weiran Li, Gang Yang, Xiaoxiao Miao, Jinming TI Revealing the Potential Pathogenesis of Glioma by Utilizing a Glioma Associated Protein-Protein Interaction Network SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; Differentially expressed genes; Glioma associated protein-protein interaction network; Functional enrichment analysis ID Glioma; Differentially expressed genes; Glioma associated protein-protein interaction network; Functional enrichment analysis AB This study aims to explore the potential mechanism of glioma through bioinformatic approaches. The gene expression profile (GSE4290) of glioma tumor and non-tumor samples was downloaded from Gene Expression Omnibus database. A total of 180 samples were available, including 23 non-tumor and 157 tumor samples. Then the raw data were preprocessed using robust multiarray analysis, and 8,890 differentially expressed genes (DEGs) were identified by using ttest (false discovery rate<0.0005). Furthermore, 16 known glioma related genes were abstracted from Genetic Association Database. After mapping 8,890 DEGs and 16 known glioma related genes to Human Protein Reference Database, a glioma associated protein-protein interaction network (GAPN) was constructed. In addition, 51 sub-networks in GAPN were screened out through Molecular Complex Detection (score≥1), and sub-network 1 was found to have the closest interaction (score=3). What’ more, for the top 10 sub-networks, Gene Ontology (GO) enrichment analysis (p value<0.05) was performed, and DEGs involved in subnetwork 1 and 2, such as BRMS1L and CCNA1, were predicted to regulate cell growth, cell cycle, and DNA replication via interacting with known glioma related genes. Finally, the overlaps of DEGs and human essential, housekeeping, tissue-specific genes were calculated (p value=1.0, 1.0, and 0.00014, respectively) and visualized by Venn Diagram package in R. About 61 % of human tissue-specific genes were DEGs as well. This research shed new light on the pathogenesis of glioma based on DEGs and GAPN, and our findings might provide potential targets for clinical glioma treatment. C1 [Pan, Weiran] Shengjing Hospital of China Medical University, Department of Neurosurgery, NO. 36 Sanhaojie Street, Heping District, 110004 Shenyang, China. [Li, Gang] Shengjing Hospital of China Medical University, Department of Neurosurgery, NO. 36 Sanhaojie Street, Heping District, 110004 Shenyang, China. [Yang, Xiaoxiao] Shengjing Hospital of China Medical University, Department of Neurosurgery, NO. 36 Sanhaojie Street, Heping District, 110004 Shenyang, China. [Miao, Jinming] Shengjing Hospital of China Medical University, Department of Neurosurgery, NO. 36 Sanhaojie Street, Heping District, 110004 Shenyang, China. RP Pan, W (reprint author), Shengjing Hospital of China Medical University, Department of Neurosurgery, 110004 Shenyang, China. EM weiranpan2@hotmail.com CR Parney IF, 2012, Basic concepts in glioma immunology. Glioma. Springer New York,42–52 Knizhnik AV, Roos WP, Nikolova T et al, 2013, Survival and death strategies in glioma cells: autophagy, senescence and apoptosis triggered by a single type of temozolomide-inducedDNAdamage. PLoS One 8:e55665 Wang Y, Jiang T, 2013, Understanding high grade glioma: molecular mechanism, therapy and comprehensive management. Cancer Lett 331: 139–146. Heim S, Mitelman F, 2011, Cancer cytogenetics: chromosomal and molecular genetic abberations of tumor cells. John Wiley & Sons Furnari FB, Fenton T, Bachoo RM et al, 2007, Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev 21: 2683–2710 Kapoor GS, O’rourke DM, 2003, Mitogenic signaling cascades in glial tumors. Neurosurgery 52:1425–1435 Milinkovic V, Bankovic J, Rakic M et al, 2012, Genomic instability and p53 alterations in patients with malignant glioma. Exp Mol Pathol 93:200–206 Bansal K, Liang ML, Rutka JT, 2006, Molecular biology of human gliomas. Technol Cancer Res Treat 5:185–194 Sun L, Hui A-M, Su Q et al, 2006, Neuronal and glioma-derived stem cell factor induces angiogenesis within the brain. Cancer Cell 9: 287–300 Barrett T, TroupDB,Wilhite SE et al, 2007, NCBI GEO:mining tens of millions of expression profiles–database and tools update. Nucleic Acids Res 35:D760–765 Barrett T, Troup DB,Wilhite SE et al, 2011, NCBI GEO: archive for functional genomics data sets—10 years on. Nucleic Acids Res 39: D1005–D1010 Mccall MN, Bolstad BM, Irizarry RA, 2010, Frozen robust multiarray analysis, fRMA). Biostatistics 11:242–253 Gautier L, Irizarry R, Cope L, and Bolstad B, 2012, Description of Affy Falcon S, Gentleman R, 2007, Using GOstats to test gene lists for GO term association. Bioinformatics 23:257–258 Abbas A, Kong X-B, Liu Z, Jing B-Y, Gao X, 2013, Automatic peak selection by a Benjamini-Hochberg-based algorithm. PLoS One 8: e53112 Becker KG, Barnes KC, Bright TJ, Wang SA, 2004, The genetic association database. Nat Genet 36:431–432 Peri S, Navarro JD, Amanchy R et al, 2003, Development of human protein reference database as an initial platform for approaching systems biology in humans. Genome Res 13: 2363–2371 Kohl M, Wiese S, Warscheid B, 2011, Cytoscape: software for visualization and analysis of biological networks, in data mining in proteomics. Humana Press. 291–303 Assenov Y, Ramirez F, Schelhorn SE, Lengauer T, Albrecht M, 2008, Computing topological parameters of biological networks. Bioinformatics 24:282–284 Zhou T, Yan G, Wang B-H, 2005, Maximal planar networks with large clustering coefficient and power-law degree distribution. Phys Rev E 71:046141 Bader GD, Hogue CW, 2003, An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinforma 4:2 Mi G, Di Y, Emerson S, Cumbie JS, Chang JH, 2012, Length bias correction in gene ontology enrichment analysis using logistic regression. PLoS One 7:e46128 Maere S, Heymans K, KuiperM(2005, BiNGO: a Cytoscape plugin to assess overrepresentation of gene ontology categories in biological networks. Bioinformatics 21:3448–3449 Li X, Li C, Shang D et al, 2011, The implications of relationships between human diseases and metabolic subpathways. PLoS One 6: e21131 Chen H, Boutros P, 2011, VennDiagram: a package for the generation of highly-customizable Venn and Euler diagrams in R. BMC Bioinforma 12:35 Hurst DR, Mehta A, Moore BP et al, 2006, Breast cancer metastasis suppressor 1, BRMS1, is stabilized by the Hsp90 chaperone. Biochem Biophys Res Commun 348:1429–1435 Mei P, Bai J, Shi M et al, 2014, BRMS1 suppresses glioma progression by regulating invasion, migration and adhesion of glioma cells. PLoS One 9:e98544 Hurst DR, Welch DR, 2011, Unraveling the enigmatic complexities of BRMS1-mediated metastasis suppression. FEBS Lett 585:3185– 3190 Nikolaev AY, Papanikolaou NA, Li M, Qin J, Gu W, 2004, Identification of a novel BRMS1-homologue protein p40 as a component of the mSin3A/p33ING1b/HDAC1 deacetylase complex. Biochem Biophys Res Commun 323:1216–1222 Liu D, Liao C, Wolgemuth DJ, 2000, A role for cyclin A1 in the activation of MPF and G2–Mtransition during meiosis of male germ cells in mice. Dev Biol 224:388–400 Ji T, Liu D, Shao W, Yang W, Wu H, Bian X, 2012, Decreased expression of LATS1 is correlated with the progression and prognosis of glioma. J Exp Clin Cancer Res 31: 67 Watanabe T, Yokoo H,YokooM, Yonekawa Y, Kleihues P, Ohgaki H, 2001, Concurrent inactivation of RB1 and TP53 pathways in anaplastic oligodendrogliomas. J Neuropathol Exp Neurol 60:1181– 1189 Kanter DM, Bruck I, Kaplan DL, 2008)Mcm subunits can assemble into two different active unwinding complexes. J Biol Chem 283: 31172–31182 Van Den BoomJ,WolterM, Kuick R et al, 2003, Characterization of gene expression profiles associated with glioma progression using oligonucleotide-based microarray analysis and real-time reverse transcription-polymerase chain reaction. Am J Pathol 163:1033– 1043 Dickerson JE, Zhu A, Robertson DL, Hentges KE, 2011, Defining the role of essential genes in human disease. PLoS One 6:e27368 Tunbridge EM, Eastwood SL, Harrison PJ, 2011, Changed relative to what? Housekeeping genes and normalization strategies in human brain gene expression studies. Biol Psychiatry 69:173–179 Xiao S-J, Zhang C, Zou Q, Ji Z-L, 2010, TiSGeD: a database for tissue-specific genes. Bioinformatics 26:1273–1275 Gong X-J, Yu H, Yang C-B, Li Y-F, 2012, Knowledge enrichment analysis for human tissue-specific genes uncover new biological insights. J Integr Bioinform 9:194 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 455 EP 462 DI 10.1007/s12253-014-9848-9 PG 8 ER PT J AU Forghanifard, MM Taleb, Sh Abbaszadegan, RM AF Forghanifard, Mahdi Mohammad Taleb, Shaghayegh Abbaszadegan, Reza Mohammad TI Notch Signaling Target Genes are Directly Correlated to Esophageal Squamous Cell Carcinoma Tumorigenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Notch signaling pathway; Esophageal squamous cell carcinoma; Real time PCR; HEY1; HEY2 ID Notch signaling pathway; Esophageal squamous cell carcinoma; Real time PCR; HEY1; HEY2 AB Notch signaling is an important cellular pathway which affects the development and function ofmany organs. It plays critical roles in maintaining of progenitor stem cell population as well as balancing cell proliferation, survival, differentiation and apoptosis. It has been shown that notch signaling is aberrantly activated during the carcinogenesis of a variety of human cancers. In this study we aimed to explore activation of this signaling pathway in esophageal squamous cell carcinoma (ESCC) through expressional analysis of notch signaling target genes. The mRNA expression of HEY1and HEY2 was comparatively analyzed by real-time PCR in tumor and related margin normal tissues of 50 ESCC patients. Comparative quantitative real-time PCR indicates the overexpression of HEY1 and HEY2 in 54 and 30 % of ESCC samples, respectively. Overexpression of HEY1 was significantly associated with stage of the tumor (p=0.048) and tumor location (p=0.008). HEY2 overexpression was also significantly correlated to node metastasis of tumor cells (p=0.043). Overexpression of HEY1 and HEY2 in ESCC is correlated to different indices of poor prognosis and it is extrapolated that such overexpression is important in progression and development of ESCC tumorigenesis. To the best of our knowledge, this is the first report introducing aberrant activation of notch signaling target genes in ESCC, where it plays roles in development and progression of the malignancy and may be considered in therapeutic modalities to restrict ESCC progression. C1 [Forghanifard, Mahdi Mohammad] Islamic Azad University, Damghan Branch, Department of Biology, Cheshmeh-Ali Boulevard, Sa’dei Square, 3671639998 Damghan, Iran. [Taleb, Shaghayegh] Mashhad University of Medical Sciences, Medical School, Medical Genetics Research CenterMashhad, Iran. [Abbaszadegan, Reza Mohammad] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, Bu-Ali square, 9196773117 Mashhad, Iran. RP Abbaszadegan, RM (reprint author), Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, 9196773117 Mashhad, Iran. EM Abbaszadeganmr@mums.ac.ir CR Wang Z, Li Y, Banerjee S et al, 2009, Emerging role of notch in stem cells and cancer. Cancer Lett 279(1):8–12 Chiba S, 2006, Notch signaling in stem cell systems. Stem Cells 24(11):2437–2447 Leong KG, Karsan A, 2006, Recent insights into the role of Notch signaling in tumorigenesis. Blood 107(6):2223–2233 Carlson ME, Conboy IM, 2007, Regulating the Notch pathway in embryonic, adult and old stem cells. Curr Opin Pharmacol 7(3):303– 309 Radtke F, Clevers H, Riccio O, 2006, From gut homeostasis to cancer. Curr Mol Med 6(3):275–289 Katoh M, 2002, WNT and FGF gene clusters, review). Int J Oncol 21(6):1269–1273 Katoh M, Katoh M, 2007, Notch signaling in gastrointestinal tract, review). Int J Oncol 30(1):247–251 Artavanis-Tsakonas S, Rand MD, Lake RJ, 1999, Notch signaling: cell fate control and signal integration in development. Science 284(5415):770–776 Scoville DH, Sato T, HeXC et al, 2008, Current view: intestinal stem cells and signaling. Gastroenterology 134(3):849–864 Wilson A, Radtke F, 2006, Multiple functions of Notch signaling in self-renewing organs and cancer. FEBS Lett 580(12):2860–2868 Adepoju O, Wong A, Kitajewski A et al, 2011, Expression of HES and HEY genes in infantile hemangiomas. Vasc Cell 3:19 Katoh M, Katoh M, 2007, Integrative genomic analyses on HES/ HEY family: Notch-independent HES1, HES3 transcription in undifferentiated ES cells, and Notch-dependent HES1, HES5, HEY1, HEY2, HEYL transcription in fetal tissues, adult tissues, or cancer. Int J Oncol 31(2):461–466 Kageyama R, Ohtsuka T, Kobayashi T, 2007, The Hes gene family: repressors and oscillators that orchestrate embryogenesis. Development 134(7):1243–1251 ForghanifardMM, Moaven O, FarshchianMet al, 2012, Expression analysis elucidates the roles of MAML1 and Twist1 in esophageal squamous cell carcinoma aggressiveness and metastasis. Ann Surg Oncol 19(3):743–749 Sobin LH, Fleming ID, 1997, TNM classification of malignant tumors, fifth edition, 1997). union internationale contre le cancer and the american joint committee on cancer. Cancer 80(9):1803– 1804 Moghbeli M, Abbaszadegan MR, Farshchian M et al, 2013, Association of PYGO2 and EGFR in esophageal squamous cell carcinoma. Med Oncol 30(2):516 ForghanifardMM, MoghbeliM, Raeisossadati R et al, 2013, Role of SALL4 in the progression and metastasis of colorectal cancer. J Biomed Sci 20:6 Liu J, Sato C, CerlettiMet al, 2010)Notch signaling in the regulation of stem cell self-renewal and differentiation. Curr Top Dev Biol 92: 367–409 Koch U, Radtke F, 2007, Notch and cancer: a double-edged sword. Cell Mol Life Sci 64(21):2746–2762 Stylianou S, Clarke RB, Brennan K, 2006, Aberrant activation of notch signaling in human breast cancer. Cancer Res 66(3):1517–1525 Ohashi S, Natsuizaka M, Yashiro-Ohtani Y et al, 2010, NOTCH1 and NOTCH3 coordinate esophageal squamous differentiation through a CSL-dependent transcriptional network. Gastroenterology 139(6):2113–2123 Gaetani P, Hulleman E, Levi D et al, 2010, Expression of the transcription factor HEY1 in glioblastoma: a preliminary clinical study. Tumori 96(1):97–102 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 463 EP 467 DI 10.1007/s12253-014-9849-8 PG 5 ER PT J AU Yuan, P Liu, D Deng, M Liu, J Wang, J Zhang, L Liu, Q Zhang, T Chen, Y Jin, G AF Yuan, Pengfei Liu, Dechun Deng, Miao Liu, Jiangbo Wang, Jianguang Zhang, Like Liu, Qipeng Zhang, Ting Chen, Yanbin Jin, Gaoyuan TI Identification of Differently Expressed Genes with Specific SNP Loci for Breast Cancer by the Integration of SNP and Gene Expression Profiling Analyses SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Single nucleotide polymorphisms; Gene expression profile; Fisher’s combined probability test; Correlation analysis; Transcription factor ID Breast cancer; Single nucleotide polymorphisms; Gene expression profile; Fisher’s combined probability test; Correlation analysis; Transcription factor AB This study aims to explore the relationship between gene polymorphism and breast cancer, and to screen DEGs (differentially expressed genes) with SNPs (single nucleotide polymorphisms) related to breast cancer. The SNPs of 17 patients and the preprocessed SNP profiling GSE 32258 (38 cases of normal breast cells) were combined to identify their correlation with breast cancer using chi-square test. The gene expression profiling batch8_9 (38 cases of patients and 8 cases of normal tissue) was preprocessed with limma package, and the DEGs were filtered out. Then fisher’s method was applied to integrate DEGs and SNPs associated with breast cancer. With NetBox software, TRED (Transcriptional Regulatory Element Database) and UCSC (University of California Santa Cruz) database, genes-associated network and transcriptional regulatory network were constructed using cytoscape software. Further, GO (Gene Ontology) and KEGG analyses were performed for genes in the networks by using siggenes. In total, 332 DEGs were identified. There were 160 breast cancer-related SNPs related to 106 genes of gene expression profiling (19 were significant DEGs). Finally, 11co-correlated DEGs were selected. In genes-associated network, 9 significant DEGs were correlated to 23 LINKER genes while, in transcriptional regulatory network, E2F1 had regulatory relationships with 7 DEGs including MTUS1, CD44, CCNB1 and CCND2. KRAS with SNP locus of rs1137282 was involved in 35 KEGG pathways. The genes of MTUS1, CD44, CCNB1, CCND2 and KRAS with specific SNP loci may be used as biomarkers for diagnosis of breast cancer. Besides, E2F1 was recognized as the transcription factor of 7 DEGs including MTUS1, CD44, CCNB1 and CCND2. C1 [Yuan, Pengfei] The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, Jinghua Road No. 24, Jianxi District, 471003 Luoyang, China. [Liu, Dechun] The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, Jinghua Road No. 24, Jianxi District, 471003 Luoyang, China. [Deng, Miao] The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, Jinghua Road No. 24, Jianxi District, 471003 Luoyang, China. [Liu, Jiangbo] The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, Jinghua Road No. 24, Jianxi District, 471003 Luoyang, China. [Wang, Jianguang] The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, Jinghua Road No. 24, Jianxi District, 471003 Luoyang, China. [Zhang, Like] The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, Jinghua Road No. 24, Jianxi District, 471003 Luoyang, China. [Liu, Qipeng] The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, Jinghua Road No. 24, Jianxi District, 471003 Luoyang, China. [Zhang, Ting] The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, Jinghua Road No. 24, Jianxi District, 471003 Luoyang, China. [Chen, Yanbin] The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, Jinghua Road No. 24, Jianxi District, 471003 Luoyang, China. [Jin, Gaoyuan] The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, Jinghua Road No. 24, Jianxi District, 471003 Luoyang, China. RP Liu, D (reprint author), The First Affiliated Hospital of Henan University Science and Technology, Department of Breast Surgery, 471003 Luoyang, China. EM liudechun2008@126.com CR Sariego J, 2010, Breast cancer in the young patient. Am Surg 76(12): 1397–1400 Boyle P, Levin B, 2008, World Cancer Report 2008. IARC Press, International Agency for Research on Cancer, Lyon Siegel R, Ma J, Zou Z, Jemal A, 2014, Cancer statistics, 2014. CA Cancer J Clin 64(1):9–29 Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF, 2003, Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci 100(7):3983 Sant M, Allemani C, Capocaccia R, Hakulinen T, Aareleid T, Coebergh JW, Coleman MP, Grosclaude P, Martinez C, Bell J, 2003, Stage at diagnosis is a key explanation of differences in breast cancer survival across Europe. Int J Cancer 106(3):416–422 Turnbull C, Ahmed S, Morrison J, Pernet D, Renwick A, Maranian M, Seal S, Ghoussaini M, Hines S, Healey CS, 2010, Genome-wide association study identifies five new breast cancer susceptibility loci. Nat Genet 42(6):504–507 Easton DF, Pooley KA, Dunning AM, Pharoah PD, Thompson D, Ballinger DG, Struewing JP, Morrison J, Field H, Luben R, 2007, Genome-wide association study identifies novel breast cancer susceptibility loci. Nature 447(7148):1087–1093 Zardawi SJ, Zardawi I, McNeil CM, Millar EK, McLeod D, Morey AL, Crea P, Murphy NC, Pinese M, Lopez‐Knowles E, 2010, High Notch1 protein expression is an early event in breast cancer development and is associated with the HER‐2 molecular subtype. Histopathology 56(3):286–296 Veronesi A, de Giacomi C, Magri MD, Lombardi D, Zanetti M, Scuderi C, Dolcetti R, Viel A, Crivellari D, Bidoli E, 2005, Familial breast cancer: characteristics and outcome of BRCA 1–2 positive and negative cases. BMC Cancer 5(1):70 Satorra A, Bentler PM, 2001, A scaled difference chi-square test statistic for moment structure analysis. Psychometrika 66(4):507–514 Sanges R, Cordero F, Calogero RA, 2007, oneChannelGUI: a graphical interface to Bioconductor tools, designed for life scientists who are not familiar with R language. Bioinformatics 23(24):3406–3408 Whitlock M, 2005, Combining probability from independent tests: the weighted Z‐method is superior to Fisher’s approach. J Evol Biol 18(5):1368–1373 Kim H-Y, Byun M-J, Kim H, 2011, A replication study of genome-wide CNVassociation for hepatic biomarkers identifies nine genes associated with liver function. Biochem Mol Biol Rep 44(9): 578–583 Yang J, Chen L, Wang L, Zhang W, Liu T, Jin Q, 2007, TrED: the Trichophyton rubrum expression database. BMC Genomics 8(1):250 Wang Y, Wang DD, 2013, University of California, Santa Cruz Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T, 2003, Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13(11):2498–2504 Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, 2000, Gene Ontology: tool for the unification of biology. Nat Genet 25(1):25–29 Nakaya A, Katayama T, Itoh M, Hiranuka K, Kawashima S, Moriya Y, Okuda S, Tanaka M, Tokimatsu T, Yamanishi Y, 2013, KEGG OC: a large-scale automatic construction of taxonomy-based ortholog clusters. Nucleic Acids Res 41(D1):D353–D357 Schwender H, Krause A, Ickstadt K, 2006, Identifying interesting genes with siggenes. The Newsletter of the R Project Volume 6/5, December 2006 34:45 Voduc KD, Cheang MC, Tyldesley S, Gelmon K, Nielsen TO, Kennecke H, 2010, Breast cancer subtypes and the risk of local and regional relapse. J Clin Oncol 28(10):1684–1691 Camphausen F, Kohl M, Ruckdeschel P, Stabla T, Ruckdeschel MP, 2007, The distr Package Irizarry RA, Hobbs B, Collin F, Beazer‐Barclay YD, Antonellis KJ, Scherf U, Speed TP, 2003, Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4(2):249–264 Tavassoli FA, Devilee P, 2003, Pathology and genetics of tumours of the breast and female genital organs, vol 4. World Health Organization Zuern C, Heimrich J, Kaufmann R, Richter KK, Settmacher U, Wanner C, Galle J, Seibold S, 2010, Down-regulation of MTUS1 in human colon tumors. Oncol Rep 23(1):183–189 FrankB, Bermejo JL, HemminkiK, Sutter C,Wappenschmidt B, Meindl A, Kiechle-Bahat M, Bugert P, Schmutzler RK, Bartram CR, 2007, Copy number variant in the candidate tumor suppressor gene MTUS1 and familial breast cancer risk. Carcinogenesis 28(7):1442–1445 Ye H, Pungpravat N, Huang B-L,Muzio LL, Mariggio MA, Chen Z, Wong DT, Zhou X, 2007, Genomic assessments of the frequent loss of heterozygosity region on 8p21. 3∼p22 in head and neck squamous cell carcinoma. Cancer Genet Cytogenet 176(2):100–106 Rodrigues-Ferreira S, Di Tommaso A, Dimitrov A, Cazaubon S, Gruel N, Colasson H, Nicolas A, Chaverot N, Molinie V, Reyal F, 2009, 8p22 MTUS1 gene product ATIP3 is a novel anti-mitotic protein underexpressed in invasive breast carcinoma of poor prognosis. PLoS One 4(10):e7239 Bajorath J, 2000, Molecular organization, structural features, and ligand binding characteristics of CD44, a highly variable cell surface glycoprotein with multiple functions. Proteins Struct Funct Bioinforma 39(2):103–111 Mayer B, Jauch K, Schildberg F, Funke I, Gunthert U, Figdor C, Johnson J, 1993, De-novo expression of CD44 and survival in gastric cancer. Lancet 342(8878):1019–1022 Du L, Wang H, He L, Zhang J, Ni B, Wang X, Jin H, Cahuzac N, Mehrpour M, Lu Y, 2008, CD44 is of functional importance for colorectal cancer stem cells. Clin Cancer Res 14(21):6751–6760 Alvero AB, Chen R, Fu H-H, Montagna M, Schwartz PE, Rutherford T, Silasi D-A, Steffensen KD, Waldstrom M, Visintin I, 2009, Molecular phenotyping of human ovarian cancer stem cells unravel the mechanisms for repair and chemo-resistance. Cell Cycle, Georgetown, Tex, 8(1):158 Herrera-Gayol A, Jothy S, 1999, Adhesion proteins in the biology of breast cancer: contribution of CD44. Exp Mol Pathol 66(2):149–156 Li Y, Chen Y-L, Xie Y-T, Zheng L-Y, Han J-Y, Wang H, Tian X-X, FangW-G, 2013, Association study of germline variants in CCNB1 and CDK1 with breast cancer susceptibility, progression, and survival among Chinese Han women. PLoS One 8(12):e84489 French JD, Ghoussaini M, Edwards SL, Meyer KB, Michailidou K, Ahmed S, Khan S, MaranianMJ, O’Reilly M, Hillman KM, 2013, Functional variants at the 11q13 risk locus for breast cancer regulate cyclin D1 expression through long-range enhancers. Am J Hum Genet 92(4):489–503 Song H, Hogdall E, Ramus SJ, DiCioccio RA, Hogdall C, Quaye L, McGuire V,Whittemore AS, Shah M, Greenberg D, 2008, Effects of common germ-line genetic variation in cell cycle genes on ovarian cancer survival. Clin Cancer Res 14(4):1090–1095 Driver KE, Song H, Lesueur F, Ahmed S, Barbosa-Morais NL, Tyrer JP, Ponder BA, Easton DF, Pharoah PD, Dunning AM, 2008, Association of single-nucleotide polymorphisms in the cell cycle genes with breast cancer in the British population. Carcinogenesis 29(2):333–341 Johnson DG, 2000, The paradox of E2F1: oncogene and tumor suppressor gene. Mol Carcinog 27(3):151–157 Abdraboh ME, Gaur RL, Hollenbach AD, Sandquist D, Raj MH, Ouhtit A, 2011, Survivin is a novel target of CD44-promoted breast tumor invasion. Am J Pathol 179(2):555–563 Huang L, 2008, Carrot and stick: HIF-α engages c-Myc in hypoxic adaptation. Cell Death Differ 15(4):672–677 Wang WY, Chien YC, Wong YK, Lin YL, Lin JC, 2012, Effects of KRAS mutation and polymorphism on the risk and prognosis of oral squamous cell carcinoma. Head Neck 34(5):663–666 Yen L-C, Yeh Y-S, Chen C-W,Wang H-M, Tsai H-L, Lu C-Y, Chang Y-T, Chu K-S, Lin S-R, Wang J-Y, 2009, Detection of KRAS oncogene in peripheral blood as a predictor of the response to cetuximab plus chemotherapy in patients with metastatic colorectal cancer. Clin Cancer Res 15(13):4508–4513 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 469 EP 475 DI 10.1007/s12253-014-9851-1 PG 7 ER PT J AU Bianchi, S Caini, S Paglierani, M Saieva, C Vezzosi, V Baroni, G Simoni, A Palli, D AF Bianchi, Simonetta Caini, Saverio Paglierani, Milena Saieva, Calogero Vezzosi, Vania Baroni, Gianna Simoni, Antonella Palli, Domenico TI Accuracy and Reproducibility of HER2 Status in Breast Cancer Using Immunohistochemistry: A Quality Control Study in Tuscany Evaluating the Impact of Updated 2013 ASCO/CAP Recommendations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Human epidermal growth factor receptor 2; Immunohistochemistry; Fluorescence in situ hybridization; Accuracy; Reproducibility ID Breast cancer; Human epidermal growth factor receptor 2; Immunohistochemistry; Fluorescence in situ hybridization; Accuracy; Reproducibility AB The correct identification of HER2-positive cases is a key point to provide the most appropriate therapy to breast cancer (BC) patients.We aimed at investigating the reproducibility and accuracy of HER2 expression by immunohistochemistry (IHC) in a selected series of 35 invasive BC cases across the pathological anatomy laboratories in Tuscany, Italy. Unstained sections of each BC case were sent to 12 participating laboratories. Pathologists were required to score according to the Food and Drug Administration (FDA) four-tier scoring system (0, 1+, 2+, 3+). Sixteen and nineteen cases were HER2 non-amplified and amplified respectively on fluorescence in situ hybridization. Among 192 readings of the 16 HER2 non-amplified samples, 153 (79.7 %) were coded as 0 or 1+, 39 (20.3 %) were 2+, and none was 3+ (false positive rate 0 %). Among 228 readings of the 19 HER2 amplified samples, 56 (24.6 %) were scored 0 or 1+, 79 (34.6 %) were 2+, and 93 (40.8 %) were 3+. The average sensitivity was 75.4 %, ranging between 47 % and 100 %, and the overall false negative rate was 24.6 %. Participation of pathological anatomy laboratories performing HER2 testing by IHC in external quality assurance programs should be made mandatory, as the system is able to identify laboratories with suboptimal performance that may need technical advice. Updated 2013 ASCO/CAP recommendations should be adopted as the widening of IHC 2+ "equivocal" category would improve overall accuracy of HER2 testing, as more cases would be classified in this category and, consequently, tested with an in situ hybridisation method. C1 [Bianchi, Simonetta] AOU Careggi, Department of Surgery and Translational MedicineFlorence, Italy. [Caini, Saverio] Cancer Research and Prevention Institute (ISPO), Molecular and Nutritional Epidemiology UnitFlorence, Italy. [Paglierani, Milena] AOU Careggi, Department of Surgery and Translational MedicineFlorence, Italy. [Saieva, Calogero] Cancer Research and Prevention Institute (ISPO), Molecular and Nutritional Epidemiology UnitFlorence, Italy. [Vezzosi, Vania] AOU Careggi, Department of Surgery and Translational MedicineFlorence, Italy. [Baroni, Gianna] AOU Careggi, Department of Surgery and Translational MedicineFlorence, Italy. [Simoni, Antonella] AOU Careggi, Department of Surgery and Translational MedicineFlorence, Italy. [Palli, Domenico] Cancer Research and Prevention Institute (ISPO), Molecular and Nutritional Epidemiology UnitFlorence, Italy. RP Bianchi, S (reprint author), AOU Careggi, Department of Surgery and Translational Medicine, Florence, Italy. EM simonetta.bianchi@unifi.it CR Popescu NC, King CR, Kraus MH, 1989, Localization of the human erbB-2 gene on normal and rearranged chromosomes 17 to bands q12-21.32. Genomics 4:362–366 Perez EA, Cortes J, Gonzalez-Angulo AM, JMS et al, 2014, HER2 testing: current status and future directions. Cancer Treat Rev 40: 276–284 Hanna WM, Ruschoff J, Bilous M et al, 2014, HER2 in situ hybridization in breast cancer: clinical implications of polysomy 17 and genetic heterogeneity. Mod Pathol 27:4–18 Wolff AC, Hammond EH, Schwartz JN et al, 2007, American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol 25:118–145 Provenzano E, Johnson N, 2009, Overview of recommendations of HER2 testing in breast cancer. Diagn Histopathol 15:478–484 Walker RA, Bartlett JM, DowsettMet al, 2008)HER2 testing inUK: further update to recommendations. J Clin Pathol 61:818–824 Wolff AC, Hammond EH, Hicks DG et al, 2013, Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol 31: 3997–4013 Hanna W, O’Malley FP, Barnes P et al, 2007, Updated recommendations from the Canadian National Consensus Meeting on HER2/neu testing in breast cancer. Curr Oncol 4:149–153 Paik S, Bryant J, Tan-Chiu E et al, 2002, Real-world performance of HER2 testing-National Surgical adjuvant Breast and Bowel Project Experience. J Natl Cancer Inst 94:852–854 Vogel CL, Bloom K, Burris H et al, 2011, Discordance between central and local laboratory Her2 testing from a large HER2-negative population in VIRGO, a metastatic breast cancer registry. Cancer Res 71(24):188s Reiner-Concin A, 2008, External quality assurance in immunohistochemistry-is it the solution to a complex problem? Breast Care 3:78–79 MirlacherM, KasperM, StorzMet al, 2004, Influence of slide aging on results of translational research studies using immunohistochemistry. Mod Pathol 17:1414–1420 Marchio C, LambrosMB, Gugliotta P et al, 2008, Does chromosome 17 centromere copy number predict polysomy in breast cancer? A fluorescence in situ hybridization and microarray-based CGH analysis. J Pathol 219:16–24 Vance GH, Barry TS, Bloom KJ et al, 2009, Genetic heterogeneity in HER2 testing in breast cancer: panel summary and guidelines. Arch Pathol Lab Med 133:611–612 Vandem Bempt I, Van Loo P, DrijkoningenMet al, 2008, Polysomy 17 in breast cancer: clinicopathologic significance and impact on HER-2 testing. J Clin Oncol 26:4869–4874 Cohen J, 1968, Weighted kappa: nominal scale agreement with provision for scaled disagreement or partial credit. Psychol Bull 70: 213–220 Landis JR,KochGG(1977, Themeasurement of observer agreement for categorical data. Biometrics 33:159–174 Ellis IO, Dowsett M, Bartlett J et al, 2000, Recommendations for HER2 testing in the UK. J Clin Pathol 53:890–892 von Wasielewski R, Krusche CA, Ruschoff J et al, 2008, Implementation of external quality assurance trials for immunohistochemically determined breast cancer biomarkers in Germany. Breast Care 3:128–133 Schrohl AS, Pedersen HC, Jonsen SS et al, 2011, Human epidermal growth factors receptors 2, HER2, immunoreactivity: specificity of three pharmacodiagnostic antibodies. Histopathology 59: 975–983 Leong TY-M, Cooper K, Leong AS-Y, 2010, Immnunohistology - past, present and future. Adv Anat Pathol 17:404–418 Reiner-Concin A, Regitnig P, Dinges HP et al, 2008, Practice of HER2 immunohistochemistry in breast carcinoma in Austria. Pathol Oncol Res 14:253–259 Bartlett JM, Going JJ, Mallon EA et al, 2001, Evaluating HER2 amplification and overexpression in breast cancer. J Pathol 195:422– 428 Rhodes A, Jasani B, Anderson E et al, 2002, Evaluation of HER-2/ neu immunohistochemical assay sensitivity and scoring on formalinfixed and paraffin-processed cell lines and breast tumors: a comparative study involving results from laboratories in 21 countries. Am J Clin Pathol 118:408–417 Sauter G, Lee J, Bartlett JMS et al, 2009, Guidelines for human epidermal growth factor receptor 2 testing: biologic and methodologic considerations. J Clin Oncol 27:1323–1333 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 477 EP 485 DI 10.1007/s12253-014-9852-0 PG 9 ER PT J AU Togami, Sh Sasajima, Y Kasamatsu, T Oda-Otomo, R Okada, S Ishikawa, M Ikeda, Shi Kato, T Tsuda, H AF Togami, Shinichi Sasajima, Yuko Kasamatsu, Takahiro Oda-Otomo, Rie Okada, Satoshi Ishikawa, Mitsuya Ikeda, Shun-ichi Kato, Tomoyasu Tsuda, Hitoshi TI Immunophenotype and Human Papillomavirus Status of Serous Adenocarcinoma of the Uterine Cervix SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Serous adenocarcinoma; Uterine cervix; Immunohistochemical features; Human papillomavirus ID Serous adenocarcinoma; Uterine cervix; Immunohistochemical features; Human papillomavirus AB Serous adenocarcinoma of the cervix (SACC) is a very rare tumor. Our study aimed to characterize the immune profile and human papillomavirus (HPV) status of SACC, in comparison with other serous adenocarcinomas arising in the female genital tract. The pathological specimens obtained from 81 patients with serous carcinoma of the uterine cervix (n=12), 29 endometrium, 20 ovary and 20 patients with mucinous carcinoma of the uterine cervix were reviewed. We assessed the expression of WT-1, p53, p16, HER2, CEA, and CA125 by immunohistochemistry and HPV DNA by PCR in 12 SACC samples. Their immune profile was compared with that of uterine papillary serous carcinoma (UPSC), ovarian serous adenocarcinoma (OSA), and mucinous endocervical adenocarcinoma (MEA). WT-1 and HER2 were expressed in very few SACC samples (0 and 0 %, respectively), but p16, CA125, CEA and p53 were present in 100, 92, 58 and 50 %, respectively. The difference in WT-1 expression between SACC and UPSC, MEA is not significant, but SACC differ significantly from OSA (p<0.01).HPV DNA (type 16 or 18) was detected in 4 of the 12 SACC. The immunophenotype of SACC was similar to UPSC, whereas the frequency of expression of WT-1 was significantly lower in SACC than OSA. It appeared that p53 expression was associated with worse clinical outcome in patients with SACC, and that HPV infection was related to its occurrence. C1 [Togami, Shinichi] National Cancer Center Hospital, Department of Gynecology, 5-1–1 Tsukiji, Chuo-ku, 104-0045 Tokyo, Japan. [Sasajima, Yuko] National Cancer Center Hospital, Department of Pathology and Clinical Laboratories, Chuo-kuTokyo, Japan. [Kasamatsu, Takahiro] National Cancer Center Hospital, Department of Gynecology, 5-1–1 Tsukiji, Chuo-ku, 104-0045 Tokyo, Japan. [Oda-Otomo, Rie] National Cancer Center Hospital, Department of Pathology and Clinical Laboratories, Chuo-kuTokyo, Japan. [Okada, Satoshi] National Cancer Center Hospital, Department of Gynecology, 5-1–1 Tsukiji, Chuo-ku, 104-0045 Tokyo, Japan. [Ishikawa, Mitsuya] National Cancer Center Hospital, Department of Gynecology, 5-1–1 Tsukiji, Chuo-ku, 104-0045 Tokyo, Japan. [Ikeda, Shun-ichi] National Cancer Center Hospital, Department of Gynecology, 5-1–1 Tsukiji, Chuo-ku, 104-0045 Tokyo, Japan. [Kato, Tomoyasu] National Cancer Center Hospital, Department of Gynecology, 5-1–1 Tsukiji, Chuo-ku, 104-0045 Tokyo, Japan. [Tsuda, Hitoshi] National Cancer Center Hospital, Department of Pathology and Clinical Laboratories, Chuo-kuTokyo, Japan. RP Togami, Sh (reprint author), National Cancer Center Hospital, Department of Gynecology, 104-0045 Tokyo, Japan. EM togami@m3.kufm.kagoshima-u.ac.jp CR Olawaiye AB, Rauh-Hain JA, Withiam-Leitch M, Rueda B, Goodman A, del Carmen MG, 2008, Utility of pre-operative serum CA-125 in the management of uterine papillary serous carcinoma. Gynecol Oncol 110(3):293–298 Zhou C, Gilks CB, Hayes M, Clement PB, 1998, Papillary serous carcinoma of the uterine cervix: a clinicopathologic study of 17 cases. Am J Surg Pathol 22(1):113–120 Togami S, Kasamatsu T, Sasajima Y, Onda T, Ishikawa M, Ikeda SI, Kato T, 2012, Tsuda H serous adenocarcinoma of the uterine cervix: a clinicopathological study of 12 cases and a review of the literature. Gynecol Obstet Investig 73(1):26–31 BockerW(2002)WHO classification of breast tumors and tumors of the female genital organs: pathology and genetics. Verh Dtsch Ges Pathol 86:116–119 Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, McShane LM, Paik S, Pegram MD, Perez EA, Press MF, Rhodes A, Sturgeon C, Taube SE, Tubbs R, Vance GH, van de Vijver M, Wheeler TM, Hayes DF, 2007, American society of clinical oncology/college of american pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 131(1):18–43 de Roda Husman AM, Walboomers JM, van den Brule AJ, Meijer CJ, Snijders PJ, 1995, The use of general primers GP5 and GP6 elongated at their 3′ ends with adjacent highly conserved sequences improves human papillomavirus detection by PCR. J Gen Virol 76(Pt 4):1057–1062 Baalbergen A, Ewing-Graham PC, Eijkemans MJ, Helmerhorst TJ, 2007, Prognosis of adenocarcinoma of the uterine cervix: p53 expression correlates with higher incidence of mortality. Int J Cancer 121(1):106–110 Hunt CR, Hale RJ, Buckley CH, Hunt J, 1996, p53 expression in carcinoma of the cervix. J Clin Pathol 49(12):971–974 Tsuda H, Jiko K, Tsugane S, Yajima M, Yamada T, Tanemura K, Tsunematsu R, Ohmi K, Sonoda T, Hirohashi S, 1995, Prognostic value of p53 protein accumulation in cancer cell nuclei in adenocarcinoma of the uterine cervix. Jpn J Cancer Res 86(11):1049–1053 Batistatou A, Zolota V, Tzoracoleftherakis E, Scopa CD, 2000, Papillary serous adenocarcinoma of the endocervix: a rare neoplasm. Immunohistochemical profile. Int J Gynecol Cancer 10(4):336–339 Nofech-Mozes S, Rasty G, Ismiil N, Covens A, Khalifa MA, 2006, Immunohistochemical characterization of endocervical papillary serous carcinoma. Int J Gynecol Cancer 16(Suppl 1):286–292 Broker TR, Jin G, Croom-Rivers A, BraggSM, RichardsonM, Chow LT, Vermund SH, Alvarez RD, Pappas PG, Squires KE, Hoesley CJ, 2001, Viral latency–the papillomavirus model. Dev Biol 106:443– 51, discussion 452–3, 465–475 Fontaine J, Hankins C, Mayrand MH, Lefevre J, Money D, Gagnon S, Rachlis A, Pourreaux K, Ferenczy A, Coutlee F, 2005, High levels of HPV-16 DNA are associated with high-grade cervical lesions in women at risk or infected with HIV. AIDS, London, England, 19(8): 785–794 Riethdorf L, Riethdorf S, Lee KR, Cviko A, Loning T, Crum CP, 2002, Human papillomaviruses, expression of p16, and early endocervical glandular neoplasia. Hum Pathol 33(9):899–904 Swan DC, Tucker RA, Tortolero-Luna G, Mitchell MF, Wideroff L, Unger ER, Nisenbaum RA, Reeves WC, Icenogle JP, 1999, Human papillomavirus, HPV, DNA copy number is dependent on grade of cervical disease and HPV type. J Clin Microbiol 37(4):1030–1034 Andersson S, Rylander E, Larsson B, Strand A, Silfversvard C, Wilander E, 2001, The role of human papillomavirus in cervical adenocarcinoma carcinogenesis. Eur J Cancer 37(2):246–250 Missaoui N, Hmissa S, Frappart L, Trabelsi A, Ben Abdelkader A, Traore C, Mokni M, Yaacoubi MT, Korbi S, 2006, p16INK4A overexpression and HPVinfection in uterine cervix adenocarcinoma. Virchows Arch 448(5):597–603 Pirog EC, Kleter B, Olgac S, Bobkiewicz P, Lindeman J, QuintWG, Richart RM, Isacson C, 2000, Prevalence of human papillomavirus DNA in different histological subtypes of cervical adenocarcinoma. Am J Pathol 157(4):1055–1062 Chiesa-Vottero AG, Malpica A, Deavers MT, Broaddus R, Nuovo GJ, Silva EG, 2007, Immunohistochemical overexpression of p16 and p53 in uterine serous carcinoma and ovarian high-grade serous carcinoma. Int J Gynecol Pathol 26(3):328–333 Clifford GM, Smith JS, Plummer M, Munoz N, Franceschi S, 2003, Human papillomavirus types in invasive cervical cancer worldwide: a meta-analysis. Br J Cancer 88(1):63–73 Franco EL, Rohan TE, Villa LL, 1999, Epidemiologic evidence and human papillomavirus infection as a necessary cause of cervical cancer. J Natl Cancer Inst 91(6):506–511 Nofech-Mozes S, Khalifa MM, Ismiil N, Dube V, Saad RS, Sun P, Seth A, 2010, Ghorab Z Detection of HPV-DNA by a PCR-based method in formalin-fixed, paraffin-embedded tissue from rare endocervical carcinoma types. Appl Immunohistochem Mol Morphol 18(1):80–85 Hadzisejdc I, Krasevic M, Haller H, Grahovac B, 2007, Distribution of human papillomavirus types in different histological subtypes of cervical adenocarcinoma. Coll Anthropol 31(Suppl 2):97–102 Acs G, Pasha T, Zhang PJ, 2004, WT1 is differentially expressed in serous, endometrioid, clear cell, and mucinous carcinomas of the peritoneum, fallopian tube, ovary, and endometrium. Int J Gynecol Pathol 23(2):110–118 Hashi A, Yuminamochi T, Murata S, Iwamoto H, Honda T, Hoshi K, 2003)Wilms tumor gene immunoreactivity in primary serous carcinomas of the fallopian tube, ovary, endometrium, and peritoneum. Int J Gynecol Pathol 22(4):374–377 Hylander B, Repasky E, Shrikant P, IntenganM, Beck A, Driscoll D, Singhal P, Lele S, Odunsi K, 2006, Expression ofWilms tumor gene, WT1, in epithelial ovarian cancer. Gynecol Oncol 101(1):12–17 Lin X, Lindner JL, Silverman JF, Liu Y, 2008, Intestinal type and endocervical-like ovarian mucinous neoplasms are immunophenotypically distinct entities. Appl Immunohistochem Mol Morphol 16(5):453–458 Nofech-Mozes S, Khalifa MA, Ismiil N, Saad RS, Hanna WM, Covens A, Ghorab Z, 2008, Immunophenotyping of serous carcinoma of the female genital tract. Mod Pathol 21(9):1147–1155 2 9. De a v e r s MT, M a l p i c a A, S i l v a EG, 2 0 0 3 , Immunohistochemistry in gynecological pathology. Int J Gynecol Cancer 13(5):567–579 30. Alkushi A, Irving J, Hsu F, Dupuis B, Liu CL, Rijn M, Gilks CB, 2003, Immunoprofile of cervical and endometrial adenocarcinomas using a tissue microarray. Virchows Arch 442(3):271–277 31. Santin AD, Bellone S, Van Stedum S, BushenW, PalmieriM, Siegel ER, De Las Casas LE, Roman JJ, Burnett A, Pecorelli S, 2005, Amplification of c-erbB2 oncogene: a major prognostic indicator in uterine serous papillary carcinoma. Cancer 104(7):1391–1397 32. Slomovitz BM, Broaddus RR, Burke TW, Sneige N, Soliman PT,Wu W, Sun CC, MunsellMF, Gershenson DM, Lu KH, 2004, Her-2/neu overexpression and amplification in uterine papillary serous carcinoma. J Clin Oncol 22(15):3126–3132 33. Togami S, Sasajima Y, Oi T, Ishikawa M, Onda T, Ikeda S, Kato T, Tsuda H, Kasamatsu T, 2012, Clinicopathological and prognostic impact of human epidermal growth factor receptor type 2, HER2, and hormone receptor expression in uterine papillary serous carcinoma. Cancer Sci 103(5):926–932 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 487 EP 494 DI 10.1007/s12253-014-9854-y PG 8 ER PT J AU Nemeth, H Kuronya, Zs Biro, K Kormosoi-Toth, K Horvath, Sz Geczi, L AF Nemeth, Hajnalka Kuronya, Zsofia Biro, Krisztina Kormosoi-Toth, Krisztina Horvath, Szabolcs Geczi, Lajos TI Pericardial Tamponade Caused by Tumor Hemorrhage – a Rare Complication of Metastatic Testicular Choriocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Nemeth, Hajnalka] National Institute of Oncology, Department of Chemotherapy and ClinicopharmacologyBudapest, Hungary. [Kuronya, Zsofia] National Institute of Oncology, Department of Chemotherapy and ClinicopharmacologyBudapest, Hungary. [Biro, Krisztina] National Institute of Oncology, Department of Chemotherapy and ClinicopharmacologyBudapest, Hungary. [Kormosoi-Toth, Krisztina] National Institute of Oncology, Department of Anesthesiology and Intensive TherapyBudapest, Hungary. [Horvath, Szabolcs] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Geczi, Lajos] National Institute of Oncology, Department of Chemotherapy and ClinicopharmacologyBudapest, Hungary. RP Nemeth, H (reprint author), National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Budapest, Hungary. EM nemethhajnalka@hotmail.com CR Williams MB, Schwartz BF, 2012, Testicular choriocarcinoma. http://emedicine.medscape.com/article/435577-overview . Accessed 26 Dec 2013 Logothetis CJ, SamuelsML, Selig DE, Ogden S, Dexeus F, Swanson D, Johnson D, von Eschenbach A, 1986, Cyclic chemotherapy with cyclophosphamide, doxorubicin, and cisplatin plus vinblastine and bleomycin in advanced germinal tumors. Results with 100 patients. Am J Med 81(2):219–228 Motzer RJ, BoslGJ, 1987)Hemorrhage: a complication ofmetastatic testicular choriocarcinoma. Urology 30(2):119–122 Tatokoro M, Kawakami S, Sakura M, Kobayashi T, Kihara K, Akamatsu H, 2008, Successful management of life-threatening choriocarcinoma syndrome with rupture of pulmonary metastatic foci causing hemorrhagic shock. Int J Urol 15(3):263–264., DOI 10.1111/j. 1442-2042.2007.01979.x Ruan Z, Wang S, Wang Z, Jing Y, 2012, A rare case of bilateral massive hemothorax from spontaneous rupture of a primary mediastinal mixed germ cell tumor. Ann Thorac Surg 93(2):664–666., DOI 10.1016/j.athoracsur.2011.07.018 Moore K, Imbeault A, Roy G, Bolduc S, 2010)Massive hemorrhage from spontaneous rupture of a retroperitoneal lymph node in patient with metastatic mixed germ cell tumor. Urology 76(1):159–161., DOI 10.1016/j.urology.2009.10.001 Baagar K, Khan FY, Alkuwari E, 2013, Choriocarcinoma syndrome: a case report and a literature review. Case Rep Oncol Med 2013: 697251., DOI 10.1155/2013/697251 Shariat SF, Duchene D, Kabbani W, Mucher Z, Lotan Y, 2005, Gastrointestinal hemorrhage as first manifestation of metastatic testicular tumor. Urology 66(6):1319 Lal A, Singhal M, Kumar S, Bag S, Singh SK, Khandelwal N, 2009, Bilateral renal and jejunal metastasis of choriocarcinoma presenting as spontaneous renal hemorrhage. Cancer Imaging 9:56–58., DOI 10. 1102/1470-7330.2009.0010 Wheater MJ, Mead GM, Bhandari S, Fennell J, 2008, Recombinant factor VIIa in the management of pulmonary hemorrhage associated with metastatic choriocarcinoma. J Clin Oncol 26(6):1008–1010., DOI 10.1200/JCO.2007.14.8486 Johnson DE, Appelt G, Samuels ML, Luna M, 1976, Metastases fromtesticular carcinoma. Study of 78 autopsied cases. Urology 8(3): 234–239 Allan N, Siller C, Breen A, 2012, Anaesthetic implications of chemotherapy. Cont Edu Anaesth Crit Care Pain 12(2):52– 56 Gersak B, Lakic N, Gorjup V, Gulic T, Berden P, Cernic NS, 2002, Right ventricular metastatic choriocarcinoma obstructing inflow and outflow tract. Ann Thorac Surg 73(5):1631–1633 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 495 EP 499 DI 10.1007/s12253-014-9842-2 PG 5 ER PT J AU Choudhari, KSh Gadbail, RA AF Choudhari, Korde Sheetal Gadbail, R Amol TI Hybrid Odontogenic Tumors: A Controversy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Choudhari, Korde Sheetal] Yerala Dental College and Hospital, Department of Oral Pathology & Microbiology, B-15, Mandovi, Chheda Nagar, Chembur(W), 4400089 Mumbai, Maharashtra, India. [Gadbail, R Amol] Sharad Pawar Dental College, Department of Oral Pathology & Microbiology, Sawangi, 442001 Wardha, Maharashatra, India. RP Choudhari, KSh (reprint author), Yerala Dental College and Hospital, Department of Oral Pathology & Microbiology, 4400089 Mumbai, India. EM kordesheetal@yahoo.co.in CR Meyer R, 1919, Beitrag zur verstaninbung uber dieNamengebung in der geschwulstlehrle. Zentralbl Pathol 30:391–6 Seifert G, Donath K, 1996, Hybrid tumours of salivary glands. Definition and classification of five rare cases. Eur J Cancer B Oral Oncol 32B:251–9 Ide F, Horie N, Shimoyama T, Sakashita H, Kusama K, 2001, So called hybrid odontogenic tumors: do they really exist. Oral Med Pathol 6:13–21 Takeda Y, Kudo K, 1986, Adenomatoid odontogenic tumor associated with calcifying odontogenic tumor. Int J Oral Maxillofac Surg 15:469–73 Seim P, Regezi JA, O’Ryan F, 2005, Hybrid ameloblastoma and calcifying epithelial odontogenic tumor: case report. J Oral Maxillofac Surg 63(6):852–5 Economopoulou P, Soliriadou S, 1998, An unusual tumor of the mandible with features of unicystic ameloblastoma and ameloblastic fibroma. J Oral Maxillofac Surg 56:1196–1200 Hisatomi M, Asaumi J, Konouchi H, Yanagi Y, Kishi K, 2000, A case of glandular odontogenic cyst associated with ameloblastoma: correlation of diagnostic imaging with histopathological features. Dentomaxillofacial Radiol 29:249–253 Slabbert H, Altini M, Crooks J, Uys P, 1992, Ameloblastoma with dentinoid induction: dentinoameloblastoma. J Oral Pathol Med 74: 776–9 Allen CM, Neville BW, Hammond HL, 1998, Adenomatoid dentinoma. report of four cases of an unusual odontogenic lesion. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 86:313–7 Hg KH, Siar CH, 2003, Odontogenic keratocyst with dentinoid formation. Oral Surg Oral Med Oral Pathol 95:601–6 MiyauchiM, Ogawa I, Takata T, Ito H, Nikai H, Ijuhhin N, Tanimoto K, 1998, Itoh Y, 1998, Clear cell odontogenic tumor: a case with induction of dentin like structures? J Oral Pathol Med 27:220–4 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2015 VL 21 IS 2 BP 501 EP 502 DI 10.1007/s12253-014-9844-0 PG 2 ER PT J AU Galamb, Benczik, M Zinner, B Vigh, E Baghy, K Jeney, Cs Kiss, A Lendvai, G Sobel, G AF Galamb, Adam Benczik, Marta Zinner, Balazs Vigh, Eszter Baghy, Kornelia Jeney, Csaba Kiss, Andras Lendvai, Gabor Sobel, Gabor TI Dysregulation of microRNA Expression in Human Cervical Preneoplastic and Neoplastic Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Cervix; Cervical cancer; microRNA; Biomarker; Screening ID Cervix; Cervical cancer; microRNA; Biomarker; Screening AB Data discussed in recent reviews demonstrated that dysregulation of microRNA (miRNA) expression profiles occurs during cervical carcinogenesis and characteristic upor downregulation of certain miRNAs might be used as biomarkers. The majority of altered miRNAs, however were found to be inconsistent upon comparison with cancerous and normal cervical epithelia in the discussed studies due to several reasons. The results obtained in this present review suggest the need for further investigations on miRNAs on larger sample sizes in order to indicate sensitivity and specificity by means of well defined, "unified" methods. In addition, obtaining further data on the clinical course and outcome of patients in comparison to the dysregulation of miRNA expression profile could turn miRNAs into prognostic and/or progression markers. Inhibition of overexpressed miRNAs, as suggested by some authors, might even serve as target for cancer therapy. C1 [Galamb, Adam] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi ut 78/a, 1082 Budapest, Hungary. [Benczik, Marta] CellCall Ltd, Roppentyu u. 48, 1134 Budapest, Hungary. [Zinner, Balazs] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi ut 78/a, 1082 Budapest, Hungary. [Vigh, Eszter] Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. [Baghy, Kornelia] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Jeney, Csaba] CellCall Ltd, Roppentyu u. 48, 1134 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. [Lendvai, Gabor] Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. [Sobel, Gabor] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi ut 78/a, 1082 Budapest, Hungary. RP Sobel, G (reprint author), Semmelweis University, 2nd Department of Obstetrics and Gynecology, 1082 Budapest, Hungary. EM sobelg@gmail.com CR Ferlay J, Shin HR, Bray F, Forman D,Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer J Int Du Cancer 127(12):2893–2917., DOI 10. 1002/ijc.25516 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90., DOI 10.3322/ caac.20107 Safaeian M, Sherman ME, 2013, From Papanicolaou to papillomaviruses: evolving challenges in cervical cancer screening in the era of human papillomavirus vaccination. J Natl Cancer Inst 105(20):1524– 1526., DOI 10.1093/jnci/djt267 Solomon D, Davey D, Kurman R, Moriarty A, O’Connor D, PreyM, Raab S, Sherman M, Wilbur D, Wright T Jr, Young N, Bethesda W, Forum GroupM(2002, The 2001 Bethesda System: terminology for reporting results of cervical cytology. JAMA J Am Med Assoc 287(16):2114–2119 Crosbie EJ, EinsteinMH, Franceschi S, KitchenerHC, 2013, Human papillomavirus and cervical cancer. Lancet 382(9895):889–899., DOI 10.1016/S0140-6736(13)60022-7 Isidean SD, Franco EL, 2014, Embracing a new era in cervical cancer screening. Lancet 383(9916):493–494., DOI 10.1016/S0140- 6736(13)62028-0 Ronco G, Dillner J, Elfstrom KM, Tunesi S, Snijders PJ, Arbyn M, Kitchener H, Segnan N, Gilham C, Giorgi-Rossi P, Berkhof J, Peto J, Meijer CJ, International HPVswg, 2014, Efficacy of HPV-based screening for prevention of invasive cervical cancer: follow-up of four European randomised controlled trials. Lancet 383(9916):524– 532., DOI 10.1016/S0140-6736(13)62218-7 de Freitas AC, Coimbra EC, Leitao MD, 2014, Molecular targets of HPVoncoproteins: potential biomarkers for cervical carcinogenesis. Biochim Biophys Acta 1845(2):91–103., DOI 10.1016/j.bbcan.2013. 12.004 TorneselloML, Buonaguro L, Giorgi-Rossi P, Buonaguro FM(2013, Viral and cellular biomarkers in the diagnosis of cervical intraepithelial neoplasia and cancer. Biomed Res Int 2013:519619., DOI 10.1155/2013/519619 ArbynM(2012, Evidence regarding human papillomavirus testing in secondary prevention of cervical cancer. Vaccine Zinner B, Gyongyosi B, Babarczi E, Kiss A, Sobel G, 2013, Claudin 1 expression characterizes human uterine cervical reserve cells. J Histochem Cytochem Off J Histochem Soc 61(12):880–888., DOI 10.1369/0022155413501324 Hu X, Schwarz JK, Lewis JS Jr, Huettner PC, Rader JS, Deasy JO, Grigsby PW, Wang X, 2010, A microRNA expression signature for cervical cancer prognosis. Cancer Res 70(4):1441–1448., DOI 10. 1158/0008-5472.CAN-09-3289 LuoM, Shen D, ZhouX, Chen X,WangW(2013)MicroRNA-497 is a potential prognostic marker in human cervical cancer and functions as a tumor suppressor by targeting the insulin-like growth factor 1 receptor. Surgery 153(6):836–847., DOI 10.1016/j.surg.2012.12.004 Reshmi G, Chandra SS, Babu VJ, Babu PS, Santhi WS, Ramachandran S, Lakshmi S, Nair AS, Pillai MR, 2011, Identification and analysis of novel microRNAs from fragile sites of human cervical cancer: computational and experimental approach. Genomics 97(6):333–340., DOI 10.1016/j.ygeno.2011.02.010 Reshmi G, Pillai MR, 2008, Beyond HPV: oncomirs as new players in cervical cancer. FEBS Lett 582(30):4113–4116., DOI 10.1016/j. febslet.2008.11.011 Pereira PM, Marques JP, Soares AR, Carreto L, Santos MA, 2010, MicroRNA expression variability in human cervical tissues. PLoS One 5(7):e11780., DOI 10.1371/journal.pone.0011780 Rao Q, Shen Q, Zhou H, Peng Y, Li J, Lin Z, 2012, Aberrant microRNA expression in human cervical carcinomas. Med Oncol 29(2):1242–1248., DOI 10.1007/s12032-011-9830-2 Cheung TH, Man KN, Yu MY, Yim SF, Siu NS, Lo KW, Doran G, Wong RR, Wang VW, Smith DI, Worley MJ Jr, Berkowitz RS, Chung TK, Wong YF, 2012, Dysregulated microRNAs in the pathogenesis and progression of cervical neoplasm. Cell Cycle 11(15): 2876–2884., DOI 10.4161/cc.21278 Lee JW, Choi CH, Choi JJ, Park YA, Kim SJ, Hwang SY, Kim WY, Kim TJ, Lee JH, Kim BG, Bae DS, 2008, Altered MicroRNA expression in cervical carcinomas. Clin Cancer Res Off J Am Assoc Cancer Res 14(9):2535–2542., DOI 10.1158/1078-0432.CCR- 07-1231 Lui WO, Pourmand N, Patterson BK, Fire A, 2007, Patterns of known and novel small RNAs in human cervical cancer. Cancer Res 67(13):6031–6043., DOI 10.1158/0008-5472.CAN-06-0561 Wang X, Tang S, Le SY, Lu R, Rader JS, Meyers C, Zheng ZM, 2008, Aberrant expression of oncogenic and tumor-suppressive microRNAs in cervical cancer is required for cancer cell growth. PLoS One 3(7):e2557., DOI 10.1371/journal.pone.0002557 Bartel DP, 2009, MicroRNAs: target recognition and regulatory functions. Cell 136(2):215–233., DOI 10.1016/j.cell.2009.01.002 Lee RC, Feinbaum RL, Ambros V, 1993, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75(5):843–854 Lee RC, Ambros V, 2001, An extensive class of small RNAs in caenorhabditis elegans. Science 294(5543):862–864., DOI 10.1126/ science.1065329 Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T, 2001, Identification of novel genes coding for small expressed RNAs. Science 294(5543):853–858., DOI 10.1126/science.1064921 Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR, 2005, MicroRNA expression profiles classify human cancers. Nature 435(7043):834–838., DOI 10. 1038/nature03702 Deng S, Calin GA, Croce CM, Coukos G, Zhang L, 2008, Mechanisms of microRNA deregulation in human cancer. Cell Cycle 7(17):2643–2646 Zheng ZM, Wang X, 2011, Regulation of cellular miRNA expression by human papillomaviruses. Biochim Biophys Acta 1809(11– 12):668–677., DOI 10.1016/j.bbagrm.2011.05.005 Farazi TA, Spitzer JI,Morozov P, Tuschl T, 2011, miRNAs in human cancer. J Pathol 223(2):102–115., DOI 10.1002/path.2806 Lendvai G, Kiss A, Kovalszky I, Schaff Z, 2010, Alterations in microRNA expression patterns in liver diseases. Orv Hetil 151(45): 1843–1853., DOI 10.1556/OH.2010.28985 Tang T, Wong HK, Gu W, Yu MY, To KF, Wang CC, Wong YF, Cheung TH, Chung TK, Choy KW(2013, MicroRNA-182 plays an onco-miRNA role in cervical cancer. Gynecol Oncol 129(1):199– 208., DOI 10.1016/j.ygyno.2012.12.043 Ribeiro J, Sousa H, 2014, MicroRNAs as biomarkers of cervical cancer development: a literature review on miR-125b and miR-34a. Mol Biol Rep 41(3):1525–1531., DOI 10.1007/s11033-013-2998-0 Deftereos G, Corrie SR, Feng Q, Morihara J, Stern J, Hawes SE, Kiviat NB, 2011, Expression of mir-21 and mir-143 in cervical specimens ranging from histologically normal through to invasive cervical cancer. PLoS One 6(12):e28423., DOI 10.1371/journal.pone. 0028423 Gocze K, Gombos K, Juhasz K, Kovacs K, Kajtar B, Benczik M, Gocze P, Patczai B, Arany I, Ember I, 2013, Unique microRNA expression profiles in cervical cancer. Anticancer Res 33(6):2561– 2567 Saavedra KP, Brebi PM, Roa JC, 2012, Epigenetic alterations in preneoplastic and neoplastic lesions of the cervix. Clin Epigenetics 4(1):13., DOI 10.1186/1868-7083-4-13 Liu L,Yu X, Guo X, Tian Z, SuM, Long Y, Huang C, Zhou F, LiuM, Wu X, Wang X, 2012, miR-143 is downregulated in cervical cancer and promotes apoptosis and inhibits tumor formation by targeting Bcl-2. Mol Med Rep 5(3):753–760., DOI 10.3892/mmr.2011.696 Slaby O, Svoboda M, Fabian P, Smerdova T, Knoflickova D, Bednarikova M, Nenutil R, Vyzula R, 2007, Altered expression of miR-21, miR-31, miR-143 and miR-145 is related to clinicopathologic features of colorectal cancer. Oncology 72(5–6):397–402., DOI 10.1159/000113489 Murakami Y, Yasuda T, Saigo K, Urashima T, Toyoda H, Okanoue T, Shimotohno K, 2006, Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene 25(17):2537–2545., DOI 10.1038/sj.onc.1209283 Yu Y, Zhang Y, Zhang S, 2013, MicroRNA-92 regulates cervical tumorigenesis and its expression is upregulated by human papillomavirus-16 E6 in cervical cancer cells. Oncol Lett 6(2):468– 474., DOI 10.3892/ol.2013.1404 Scheffner M, Huibregtse JM, Vierstra RD, Howley PM, 1993, The HPV-16 E6 and E6-AP complex functions as a ubiquitin-protein ligase in the ubiquitination of p53. Cell 75(3):495–505 LiY,Wang F, Xu J,Ye F, ShenY, Zhou J, LuW,Wan X, Ma D, Xie X, 2011, Progressive miRNA expression profiles in cervical carcinogenesis and identification of HPV-related target genes for miR-29. J Pathol 224(4):484–495., DOI 10.1002/path.2873 Martinez I, Gardiner AS, Board KF, Monzon FA, Edwards RP, Khan SA, 2008, Human papillomavirus type 16 reduces the expression of microRNA-218 in cervical carcinoma cells. Oncogene 27(18):2575– 2582., DOI 10.1038/sj.onc.1210919 Liu L,Wang YL,Wang JF, 2012, Differential expression of miR-21, miR-126, miR-143, miR-373 in normal cervical tissue, cervical cancer tissue and Hela cell. Sichuan Da Xue Xue Bao Yi Xue Ban 43(4): 536–539 Huang L, Lin JX, Yu YH, Zhang MY, Wang HY, Zheng M, 2012, Downregulation of six microRNAs is associated with advanced stage, lymph node metastasis and poor prognosis in small cell carcinoma of the cervix. PLoS One 7(3):e33762., DOI 10.1371/journal. pone.0033762 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 503 EP 508 DI 10.1007/s12253-014-9871-x PG 6 ER PT J AU Bal, MM Ramadwar, M Deodhar, K Shrikhande, Sh AF Bal, Meenu Munita Ramadwar, Mukta Deodhar, Kedar Shrikhande, Shailesh TI Pathology of Gallbladder Carcinoma: Current Understanding and New Perspectives SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Gallbladder carcinoma; Epidemiology; Pathways; Terminology; Precancerous lesions; Histologic subtypes; Molecular pathology ID Gallbladder carcinoma; Epidemiology; Pathways; Terminology; Precancerous lesions; Histologic subtypes; Molecular pathology AB Gallbladder carcinoma is a rare and highly lethal malignancy. It stands out from amongst the other GI tract malignancies for its unique epidemiological profile, proclivity for female gender, definitional ambiguities, ability to escape early diagnosis, and absence of effective treatment. Pathobiology of gallbladder carcinoma continues to remain poorly understood. Recently, better characterization of the precursor lesions and elucidation of underlying molecular pathways has enhanced our understanding of gallbladder tumorigenesis. Proposal of a unified terminology and evolving consensus in classifying gallbladder pre-invasive neoplasia offers hope of better assimilation of rare data from diverse parts of the world. Identifying biomarkers and cancer specific cellular targets that will pave the way for novel therapeutic approaches for gallbladder carcinoma is urgently needed. In this review we delve into the epidemiologic, genetic and pathologic characteristics of this enigmatic disease with a special focus on the recent advancements in the field of gallbladder pathology. C1 [Bal, Meenu Munita] Tata Memorial Hospital, Department of Pathology, 400012 Parel, Mumbai, India. [Ramadwar, Mukta] Tata Memorial Hospital, Department of Pathology, 400012 Parel, Mumbai, India. [Deodhar, Kedar] Tata Memorial Hospital, Department of Pathology, 400012 Parel, Mumbai, India. [Shrikhande, Shailesh] Tata Memorial Hospital, Department of Hepato-Pancreato-Biliary Surgical Oncology, 400012 Parel, Mumbai, India. RP Bal, MM (reprint author), Tata Memorial Hospital, Department of Pathology, 400012 Parel, India. EM munitamenon@gmail.com CR Misra S, Chaturvedi A,Misra NC, Sharma ID, 2003, Carcinoma of the gallbladder. Lancet Oncol 4:167–176 Kumar JR, Tewari M, Rai A, Sinha R, Mohapatra SC, Shukla HS, 2006, An objective assessment of demography of gallbladder cancer. J Surg Oncol 93:610–614 Randi G, Franceschi S, La Vecchia C, 2006, Gallbladder cancer worldwide: geographical distribution and risk factors. Int J Cancer 118:1591–1602 Lazcano-Ponce EC, Miquel JF,MunozN et al, 2001, Epidemiology and molecular pathology of gallbladder cancer. CA Cancer J Clin 51:349–364 Hsing AW, Bai Y, Andreotti G et al, 2007, Family history of gallstones and the risk of biliary tract cancer and gallstones: a population-based study in Shanghai, China. Int J Cancer 121:832– 838 AndiaME, Hsing AW, Andreotti G, Ferreccio C, 2008, Geographic variation of gallbladder cancer mortality and risk factors in Chile: a population-based ecologic study. Int J Cancer 123:1411–1416 Serra I, Calvo A, Csendes A, Sharp A, 1989, Gastric and gallbladder carcinoma in Chile: epidemiological changes and control programs. Rev Med Chil 17:834–836 Reid KM, Ramos-De la Medina A, Donohue JH, 2007, Diagnosis and surgical management of gallbladder cancer: a review. J Gastrointest Surg 11:671–681 Nath G, Gulati AK, Shukla VK, 2010, Role of bacteria in carcinogenesis, with special reference to carcinoma of the gallbladder. World J Gastroenterol 16:5395–5404 Stinton LM, Shaffer EA, 2012, Epidemiology of gallbladder disease: cholelithiasis and cancer. Gut Liver 6:172–187 Diehl AK, Beral V, 1981, Cholecystectomy and changingmortality from gallbladder cancer. Lancet 2:187–189 Lowenfels AB,Walker AM, Althaus DP, Townsend G, Domello FL, 1989, Gallstone growth, size, and risk of gallbladder cancer: an interracial study. Int J Epidemiol 18:50–54 Diehl AK, 1983, Gallstone size and the risk of gallbladder cancer. JAMA 250:2323–2326 Albores-Saavedra J, Alcantra-Vazguez A, Cruz-Ortiz H, Herra- Goepfert R, 1980, The precursor lesions of invasive gallbladder carcinoma: hyperplasia, atypical hyperplasia and carcinoma in situ. Cancer 45:919–927 Wistuba II, Gazdar AF, 2004, Gallbladder cancer: lessons from a rare tumour. Nat Rev Cancer 4:695–706 Shrikhande SV, Barreto SG, Singh S, Udwadia TE, Agarwal AK, 2010, Cholelithiasis in gallbladder cancer: coincidence, cofactor, or cause! Eur J Surg Oncol 36:514–519 Tewari M, 2006, Contribution of silent gallstones in gallbladder cancer. J Surg Oncol 93:629–632 Spina GP, Pagani M, 1998, Prophylactic cholecystectomy for gallbladder calculosis. Ann Ital Chir 69:713–717 DiehlAK(1980, Epidemiology of gallbladder cancer: a synthesis of recent data. J Natl Cancer Int 65:1209–1214 Axelrod L, Munster AM, O’Brien TF, 1971, Typhoid cholecystitis and gallbladder carcinoma after interval of 67 years. JAMA 217:83 Caygill CP, HillMJ, BraddickM, Sharp JC, 1994, Cancer mortality in chronic typhoid and paratyphoid carriers. Lancet 343:83–84 Mellemgaard A, Gaarslev K, 1988, Risk of hepatobiliary cancer in carriers of Salmonella typhi. J Natl Cancer Inst 80:288 el Zayadi A, Ghoneim M, Kabil SM, el Tawil A, Sherif A, Selim O, 1991, Bile duct carcinoma in Egypt: possible etiological factors. Hepatogastroenterology 38:337–340 Strom BL, Soloway RD, Rios-Dalenz JL et al, 1995, Risk factors for gallbladder cancer. An international collaborative case–control study. Cancer 76:1747–1756 Shukla VK, Tiwari SC, Roy SK, 1993, Biliary bile acids in cholelithiasis and carcinoma of the gall bladder. Eur J Cancer Prev 2:155– 160 Nath G, Singh YK, Maurya P, Gulati AK, Srivastava RC, Tripathi SK, 2010, Does Salmonella Typhi primarily reside in the liver of chronic typhoid carriers? J Infect Dev Ctries 4:259–261 Shenker BJ, McKay T, Datar S, Miller M, Chowhan R, Demuth D, 1999, Actinobacillus actinomycetemcomitans immunosuppressive protein is a member of the family of Cytolethal distending toxins capable of causing a G2 arrest in human T cells. J Immunol 162: 4773–4780 Kumar S, 2006, Infection as a risk factor for gallbladder cancer. J Surg Oncol 93:633–639 Goldberg MS, Theriault G, 1994, Retrospective cohort study of workers of a synthetic textiles plant in Quebec: II. Colorectal cancer mortality and incidence. Am J Ind Med 25:909–922 Kowalewski K, Todd EF, 1971, Carcinoma of the gallbladder induced in hamsters by insertion of cholesterol pellets and feeding dimethylnitrosamine. Proc Soc Exp Biol Med 136:482–486 Cunningham SC, Alexander HR, 2007, Porcelain gallbladder and cancer: ethnicity explains a discrepant literature? Am J Med 120: 17–18 Stephen AE, BergerDL, 2001, Carcinoma in the porcelain gallbladder: a relationship revisited. Surgery 129:699–703 Towfigh S, McFadden DW, Cortina GR et al, 2001, Porcelain gallbladder is not associated with gallbladder carcinoma. Am Surg 67: 7–10 Kim JH, KimWH, Yoo BM, Kim JH, Kim MW, 2009, Should we perform surgical management in all patients with suspected porcelain gallbladder? Hepatogastroenterology 56:943–945 Seth S, Bedford A, Chopra C, 2000, Primary gallbladder cancer: recognition of risk factors and the role of prophylactic cholecystectomy. Am J Gastroenterol 95:1402–1410 Pandey M, 2003, Risk factors for gallbladder cancer: a reappraisal. Eur J Cancer Prev 12:15–24 Chijiiwa K, Kimura H, TanakaM(1995)Malignant potential of the gallbladder in patients with anomalous pancreaticobiliary ductal junction. Int Surg 80:61–64 Kang CM, Kim KS, Choi JS, Lee WJ, Kim BR, 2007, Gallbladder carcinoma associated with anomalous pancreaticobiliary duct junction. Can J Gastroenterol 21:383–387 Tomono H, Nimura Y, Aono K, Nakashima I, Iwamoto T, Nakashima N, 1996, Point mutations of the c-Ki-ras gene in carcinoma and atypical epithelium associated with congenital biliary dilation. Am J Gastroenterol 91:1211–1214 Lewis JT, Talwalkar JA, Rosen CB, Smyrk TC,AbrahamSC, 2007, Prevalence and risk factors for gallbladder neoplasia in patients with primary sclerosing cholangitis: evidence for a metaplasia-dysplasiacarcinoma sequence. Am J Surg Pathol 31:907–913 Parker RG, Kendall EJ, 1954, The liver in ulcerative colitis. BrMed J 2:1030–1032 Ritchie JK, Allan RN, Macartney J, Thompson H, Hawley PR, CookeWT, 1974, Biliary tract carcinoma associated with ulcerative colitis. Q J Med 43:263–279 Pandey M, 2003, Risk factors for gallbladder cancer: a reappraisal. Eur J Cancer Prev 12:15–24 Pandey SN, Srivastava A, Dixit M, Choudhuri G, Mittal B, 2007, Haplotype analysis of signal peptide, insertion/deletion, and XbaI polymorphisms of the APOB gene in gallbladder cancer. Liver Int 27:1008–1015 Buch S, Schafmayer C, Volzke H et al, 2007, A genome-wide association scan identifies the hepatic cholesterol transporter ABCG8 as a susceptibility factor for human gallstone disease. Nat Genet 39:995–999 Park SK, Andreotti G, Sakoda LC et al, 2009, Variants in hormonerelated genes and the risk of biliary tract cancers and stones: a population-based study in China. Carcinogenesis 30:606–614 Shaffer EA, 2008, Gallbladder cancer: the basics. Gastroenterol Hepatol, NY, 4:737–741 Srivastava A, Mittal B, 2009, Complement receptor 1, A3650G RsaI and intron 27 HindIII, polymorphisms and risk of gallbladder cancer in north Indian population. Scand J Immunol 70:614–620 Srivastava A, Choudhuri G, Mittal B, 2010, CYP7A1, −204 A>C; rs3808607 and −469 T>C; rs3824260, promoter polymorphisms and risk of gallbladder cancer in North Indian population. Metabolism 59:767–773 Pandey SN, Dixit M, Choudhuri G, Mittal B, 2006, Lipoprotein receptor associated protein, LRPAP1, insertion/deletion polymorphism: association with gallbladder cancer susceptibility. Int J Gastrointest Cancer 37:124–128 Srivastava A, Pandey SN, Choudhuri G, Mittal B, 2008, CCR5 Delta32 polymorphism: associated with gallbladder cancer susceptibility. Scand J Immunol 67:516–522 Vishnoi M, Pandey SN, Modi DR, Kumar A, Mittal B, 2008, Genetic susceptibility of epidermal growth factor +61A>G and transforming growth factor beta1–509C>T gene polymorphisms with gallbladder cancer. Hum Immunol 69:360–367 Vishnoi M, Pandey SN, Choudhuri G, Mittal B, 2008, IL-1 gene polymorphisms and genetic susceptibility of gallbladder cancer in a north Indian population. Cancer Genet Cytogenet 186:63–68 Shukla PJ, Barreto SG, Gupta P, et al., 2007, Is there a role for estrogen and progesterone receptors in gall bladder cancer? HPB, Oxford 9:285–288 Dixit R, Srivastava P, Basu S, Srivastava P, Mishra PK, Shukla VK, 2013, Association ofmustard oil as cooking media with carcinoma of the gallbladder. J Gastrointest Cancer 44:177–181 Rai A,Mohapatra SC, Shukla HS, 2004, A review of association of dietary factors in gallbladder cancer. Indian J Cancer 41:147–151 Rai A, Mohapatra SC, Shukla HS, 2006, Correlates between vegetable consumption and gallbladder cancer. Eur J Cancer Prev 15: 134–137 Myers RP, Shaffer EA, Beck PL, 2002, Gallbladder polyps: epidemiology, natural history and management. Can J Gastroenterol 16: 187–194 Roa I, de Aretxabala X, Araya JC, Roa J, 2006, Preneoplastic lesions in gallbladder cancer. J Surg Oncol 93:615–623 Albores-Saavedra J, Henson DE, 2000, Tumors of gallbladder and extrahepatic bile ducts. Fascicle 23, 3rd edn. Armed Forces Institute of Pathology, Washington Roa I, Araya JC, Villaseca M et al, 1996, Preneoplastic lesions and gallbladder cancer: an estimate of the period required for progression. Gastroenterology 111:232–236 Lee SH, Lee DS, You IY et al, 2010, Histopathologic analysis of adenoma and adenoma-related lesions of the gallbladder. Korean J Gastroenterol 55:119–1126 Goldin RD, Roa JC, 2009)Gallbladder cancer: amorphological and molecular update. Histopathology 55:218–229 Roa I, de Aretxabala X, Morgan R et al, 2004, Clinicopathological features of gallbladder polyps and adenomas. Rev Med Chil 132: 673–679 Kozuka S, Tsubone N, Yasui A, Hachisuka K, 1982, Relation of adenoma to carcinoma in the gallbladder. Cancer 50:2226–2234 Adsay V, Jang KT, Roa JC et al, 2012, Intracholecystic papillarytubular neoplasms, ICPN, of the gallbladder, neoplastic polyps, adenomas, and papillary neoplasms that are ≥1.0 cm): clinicopathologic and immunohistochemical analysis of 123 cases. Am J Surg Pathol 36:1279–301 Dursun N, Roa JC, Tapia O, et al., 2010, Intravesicular papillarytubular neoplasm, IVPN, as a unifying category for mass forming preinvasive neoplasms of the gallbladder: an analysis of 87 cases. Mod Pathol. 2010;23:144A [Abstract] Albores-Saavedra J, Adsay NV, Crawford JM, et al., 2010, Carcinoma of the gallbladder and extrahepatic bile ducts. In: Bosman FT, Carneiro F, Hruban R, et al., eds. World Health Organisation Classification of Tumors of Digestive system. 4th ed. Lyon: IARC Press266-274 YamamotoM, Nakajo S, Tahara E, 1988)Histological classification of epithelial polypoid lesions of the gallbladder. Acta Pathol Jpn 38: 181–192 Adsay NV, Klimstra DS, 2009, Benign and malignant tumors of the gallbladder and extrahepatic biliary tract. In: Odze RD, Goldblum JR, eds, Surgical Pathology of the GI tract, Liver, Biliary Tract, and Pancreasedn, Vol. Elsevier, Philadelphia, pp 845–875 Albores-Saavedra J, Tuck M, McLaren BK et al, 2005, Papillary carcinomas of the gallbladder: analysis of noninvasive and invasive types. Arch Pathol Lab Med 129:905–909 Albores-Saavedra J, Vardaman CJ, Vuitch F, 1993, Non-neoplastic polypoid lesions and adenomas of the gallbladder. Pathol Annu 28: 145–177 Escalona A, Leon F, Bellolio F et al, 2006, Gallbladder polyps: correlation between ultrasonographic and histopathological findings. Rev Med Chil 134:1237–1242 Albores-Saavedra J, Henson DE, 1993, Gallbladder and extrahepatic bile ducts. W.B. Saunders, Philadelphia, pp 167–181 Kato S, 1995, Morphological analysis of the gallbladder elevated lesions—macroscopic, stereoscopic, and histological study. Nippon Shokakibyo Gakkai Zasshi 92:1149–1160 Laitio M, Histogenesis of epithelial neoplasms of human gallbladder II, 1983, Classification of carcinoma on the basis of morphological features. Pathol Res Pract 178:57–66 Nagata S, Ajioka Y, Nishikura K et al, 2007, Co-expression of gastric and biliary phenotype in pyloric-gland type adenoma of the gallbladder: immunohistochemical analysis of mucin profile and CD10. Oncol Rep 17:721–729 Nakajo S, Yamamoto M, Tahara E, 1990, Morphometrical analysis of gall-bladder adenoma and adenocarcinoma with reference to histogenesis and adenoma-carcinoma sequence. Virchows Arch A Pathol Anat Histopathol 417:49–56 Tanous JH, Ilano PC, Cruz A, 1960, Adenoma of the gallbladder. J Int Coll Surg 34:298–301 Yamagiwa H, 1986, Clinico-pathological study of gallbladder diseases— V. Adenoma of the gallbladder. Rinsho Byori 34:729–732 ZhouMJ, 1985, Adenoma of the gallbladder–pathologic analysis of 14 cases. Zhonghua Zhong Liu Za Zhi 7:448–450 Christensen AH, Ishak KG, 1970, Benign tumors and pseudotumors of the gallbladder. Report of 180 cases. Arch Pathol 90:423–432 Zhang ZX, Yin WH, Zhu ZY, 1994, Adenoma of the gallbladder and its canceration: an analysis of 88 cases. Zhonghua Wai Ke Za Zhi 32:400–402 Roa JC, Katabi N, Adsay NV, 2011, Molecular pathology of liver diseases. In: Monga P, ed, Neoplasms of the Gallbladder edn, Vol. Springer, New York, pp 891–905 Adsay NV, 2010, Sternberg’s diagnostic surgical pathology. In: Mills SE, ed, Gallbladder, Extrahepatic Biliary Tree, and Ampulla, vol 2, 5th edn. Lippincott Williams and Wilkins, Philadelphia, pp 1600–1663 Mayo C. Papillomas of the gallbladder, 1915, Collected papers of the Mayo Clinic 249 Hruban RH, Pitman MB, Klimstra DS, eds, 2007, Tumors of the pancreas. In: Atlas of Tumor Pathology, 4th Series. American Registry of Pathology and Armed Forces Institute of Pathology, Washington, DC Zen Y, Adsay NV, Bardadin K et al, 2007, Biliary intraepithelial neoplasia: an international interobserver agreement study and proposal for diagnostic criteria. Mod Pathol 20:701–709 Adsay NV, Conlon KC, Zee SY et al, 2002, Intraductal papillary mucinous neoplasms of the pancreas: an analysis of in situ and invasive carcinomas in 28 patients. Cancer 94:62–77 Yamaguchi H, Shimizu M, Ban S et al, 2009, Intraductal tubulopapillary neoplasms of the pancreas distinct from pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasms. Am J Surg Pathol 33:1164–1172 Kloek JJ, van der Gaag NA, Erdogan D et al, 2011, A comparative study of intraductal papillary neoplasia of the biliary tract and pancreas. Hum Pathol 42:824–832 Ji Y, Fan J, Zhou J et al, 2008, Intraductal papillary neoplasms of bile duct. A distinct entity like its counterpart in pancreas. Histol Histopathol 23:41–50 YamamotoM, Nakajo S, Tahara E, 1989, Dysplasia of the gallbladder. Its histogenesis and correlation to gallbladder adenocarcinoma. Pathol Res Pract 185:454–460 Yamamoto M, Nakajo S, Tahara E, 1989, Carcinoma of the gallbladder: the correlation between histogenesis and prognosis. Virchows Arch A Pathol Anat Histopathol 414:83–90 Adsay NV, 2007, Neoplastic precursors of the gallbladder and extrahepatic biliary system. Gastroenterol Clin North Am 36:889–900 Ojeda VJ, Shilkin KB, Walters MN, 1985, Premalignant epithelial lesions of the gall bladder: a prospective study of 120 cholecystectomy specimens. Pathology 17:451–454 Chan KW(1988, Review of 253 cases of significant pathology in 7, 910 cholecystectomies in Hong Kong. Pathology 20:20–23 Duarte I, Llanos O, Domke H, Harz C, Valdivieso V, 1993, Metaplasia and precursor lesions of gallbladder carcinoma. Frequency, distribution, and probability of detection in routine histologic samples. Cancer 72:1878–1884 Bergquist A, Glaumann H, Stal P, Wang GS, Broome U, 2001, Biliary dysplasia, cell proliferation and nuclear DNAfragmentation in primary sclerosing cholangitis with and without cholangiocarcinoma. J Intern Med 249:69–75 Boberg KM, Jebsen P, Clausen OP, Foss A, Aabakken L, Schrumpf E, 2006, Diagnostic benefit of biliary brush cytology in cholangiocarcinoma in primary sclerosing cholangitis. J Hepatol 45:568–574 Lal A,OkonkwoA, Schindler S, de Frias D, Nayar R, 2004, Role of biliary brush cytology in primary sclerosing cholangitis. Acta Cytol 48:9–12 Ludwig J, Wahlstrom HE, Batts KP, Wiesner RH, 1992, Papillary bile duct dysplasia in primary sclerosing cholangitis. Gastroenterology 102:2134–2138 Narayanan Menon KV, 2006, Primary sclerosing cholangitis, chronic ulcerative colitis, and bile duct dysplasia–Case presentation. Liver Transplantation: Official publication of the american association for the study of liver diseases and the international liver transplantation society 12:S14 Kloppel G, Adsay V, Konukiewitz B, Kleeff J, Schlitter AM, Esposito I, 2013, Precancerous lesions of the biliary tree. Best Pract Res Clin Gastroenterol 27:285–297 Zen Y, Aishima S, Ajioka Y et al, 2005, Proposal of histological criteria for intraepithelial atypical/proliferative biliary epithelial lesions of the bile duct in hepatolithiasis with respect to cholangiocarcinoma: preliminary report based on interobserver agreement. Pathol Int 55:180–188 Albores-Saavedra J, NadjiM, Henson DE, Intestinal-type adenocarcinoma of the gallbladder, 1986, A clinicopathologic study of seven cases. Am J Surg Pathol 10:19–25 Bagci P, Saka B, Erbarut I, et al., 2013, Growth patterns of highgrade gallbladder dysplasia: clinicopathologic associations and diagnostic implications in an analysis of 318 cases.Modern Pathology [Abstract confirmation number: 1567] Zhao H, Davydova L, Mandich D, Cartun RW, Ligato S, 2007, S100A4 protein and mesothelin expression in dysplasia and carcinoma of the extrahepatic bile duct. Am J Clin Pathol 127:374–379 Wistuba II, Gazdar AF, Roa I, Albores-Saavedra J, 1996, p53 protein overexpression in gallbladder carcinoma and its precursor lesions: an immunohistochemical study. Hum Pathol 27:360–365 Parwani A, Geradts J, Caspers E et al, 2003, Immunohistochemical and genetic analysis of non-small cell and small cell gallbladder carcinoma and their precursor lesions. Mod Pathol 16:299–308 Albores-Saavedra J, Murakata L, Krueger JE et al, 2000, Noninvasive and minimally invasive papillary carcinomas of the extrahepatic bile ducts. Cancer 89:508–515 Foster DR, Foster DB, 1980, Gall-bladder polyps in Peutz-Jeghers syndrome. Postgrad Med J 56:373–376 Tantachamrun T, Borvonsombat S, Theetranont C, 1979, Gardner’s syndrome associated with adenomatous polyp of gall bladder: report of a case. J Med Assoc Thail ¼ Chotmaihet Thangphaet 62:441– 447 Walsh N, Qizilbash A, Banerjee R, Waugh GA, 1987, Biliary neoplasia in Gardner’s syndrome. Arch Pathol Lab Med 111:76–77 Roa I, Araya JC, Villaseca M et al, 1999, Gallbladder cancer in a high risk area: morphological features and spread patterns. Hepatogastroenterology 46:1540–1546 Romero-Gonzalez RJ, Garza-Flores A, Martinez-PerezMaldonado L, Diaz-Elizondo JA, Muniz-Eguia JJ, Barbosa-Quintana A, 2012, Gallbladder selection for histopathological analysis based on a simple method: a prospective comparative study. Ann R Coll Surg Engl 94:159–64 Darmas B, Mahmud S, Abbas A, Baker AL, 2007, Is there any justification for the routine histological examination of straightforward cholecystectomy specimens? Ann R Coll Surg Engl 89:238– 241 Siddiqui FG, Memon AA, Abro AH, Sasoli NA, Ahmad L, 2013, Routine histopathology of gallbladder after elective cholecystectomy for gallstones: waste of resources or a justified act? BMC Surg 8: 13:26 Shrestha R, Tiwari M, Ranabhat SK, Aryal G, Rauniyar SK, Shrestha HG, 2010, Incidental gallbladder carcinoma: value of routine histological examination of cholecystectomy specimens. Nepal Med Coll J 12:90–94 Adsay V, Saka B, Basturk O, Roa JC, 2013, Criteria for pathologic sampling of gallbladder specimens. Am J Clin Pathol 140:278–280 Gourgiotis S, Kocher HM, Solaini L, Yarollahi A, Tsiambas E, Salemis NS, 2008, Gallbladder cancer. Am J Surg 196:252–264 Levin B, 1999, Gallbladder carcinoma. Ann Oncol 10:S129–30 Mukhopadhyay S, Landas SK, 2005, Putative precursors of gallbladder dysplasia: a review of 400 routinely resected specimens. Arch Pathol Lab Med 129:386–390 Henson DE, Albores-Saavedra J, Corle D, 1992, Carcinoma of the gallbladder: histologic grade, stage of disease, grade and survival. Cancer 70:1493–1497 Vardaman C, Albores-Saavedra J, 1995, Clear cell carcinomas of the gallbladder and extrahepatic bile ducts.AmJ Surg Pathol 19:91– 99 Ghaouti M, Znati K, Jahid A et al, 2013, A gallbladder tumor revealing metastatic clear cell renal carcinoma: report of case and review of literature. Diagn Pathol 8:4 Albores-Saavedra J, Molberg K, Henson DE, 1996, Unusual malignant epithelial tumors of the gallbladder. Semin Diagn Pathol 13: 326–38 Konishi E, Nakashima Y, Smyrk TC, Masuda S, 2003, Clear cell carcinoid tumor of the gallbladder. A case without von Hippel- Lindau disease. Arch Pathol Lab Med 127:745–747 Dursun N, Escalona OT, Roa JC, Basturk O, Bagci P, Cakir A et al, 2012, Mucinous carcinomas of the gallbladder: clinicopathologic analysis of 15 cases identified in 606 carcinomas. Arch Pathol Lab Med 136:1347–1358 Roa JC, Tapia O, Cakir A et al, 2011, Squamous cell and adenosquamous carcinomas of the gallbladder: clinicopathological analysis of 34 cases identified in 606 carcinomas. Mod Pathol 24: 1069–1078 Kondo M, Dono K, Sakon M et al, 2002, Adenosquamous carcinoma of the gallbladder. Hepatogastroenterology 49:1230–1234 Chan KM, Yu MC, Lee WC et al, 2007, Adenosquamous/ squamous cell carcinoma of the gallbladder. J Surg Oncol 95: 129–134 Roa JC, Anabalon RL, Tapia EO,Melo AA, de Aretxabala UX, Roa EI, 2005, Frequency of K-ras mutation in biliary and pancreatic tumors. Rev Med Chil 133:1434–1440 NagahashiM, Ajioka Y, Lang I, Szentirmay Z, KaslerM, Nakadaira H et al, 2008, Genetic changes of p53, K-ras, and microsatellite instability in gallbladder carcinoma in high-incidence areas of Japan and Hungary. World J Gastroenterol 14:70–75 SinghMK, Chetri K, Pandey UB, Kapoor VK, Mittal B, Choudhuri G, 2004, Mutational spectrum of K-ras oncogene among Indian patients with gallbladder cancer. J Gastroenterol Hepatol 19:916– 921 Saetta AA, 2006, K-ras, p53 mutations, and microsatellite instability, MSI, in gallbladder cancer. J Surg Oncol 93:644–649 Wistuba II, Jaclyn Hung KS et al, 1995, Allele-specific mutations involved in the pathogenesis of endemic gallbladder carcinoma in Chile. Cancer Res 55:2511–2515 Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M(1988)Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell 53:549–554 Tada M, Yokosuka O, Omata M, Ohto M, Isono K, 1990, Analysis of ras gene mutations in biliary and pancreatic tumors by polymerase chain reaction and direct sequencing. Cancer 66:930–935 Yoshida S, Todoroki T, Ichikawa Y et al, 1995, Mutations of p16Ink4/CDKN2 and p15Ink4B/MTS2 genes in biliary tract cancers. Cancer Res 55:2756–2760 Ajiki T, Fujimori T, Onoyama H et al, 1996, K-ras gene mutation in gall bladder carcinomas and dysplasia. Gut 38:426–429 Nakazawa K, Dobashi Y, Suzuki S, Fujii H, Takeda Y, Ooi A, 2005, Amplification and overexpression of c-erbB-2, epidermal growth factor receptor, and c-met in biliary tract cancers. J Pathol 206: 356–365 Chaube A, Tewari M, Garbyal RS, Singh U, Shukla HS, 2006, Preliminary study of p53 and c-erbB-2 expression in gallbladder cancer in Indian patients. BMC Cancer 6:126 Suzuki T, Takano Y, Kakita A, Okudaira M, 1993, An immunohistochemical and molecular biological study of c-erbB-2 amplification and prognostic relevance in gallbladder cancer. Pathol Res Pract 189:283–292 Shafizadeh N, Grenert JP, Sahai V, Kakar S, 2010, Epidermal growth factor receptor andHER-2/neu status by immunohistochemistry and fluorescence in situ hybridization in adenocarcinomas of the biliary tree and gallbladder. Hum Pathol 41:485–492 Leone F, Cavalloni G, Pignochino Yet al, 2006, Somatic mutations of epidermal growth factor receptor in bile duct and gallbladder carcinoma. Clin Cancer Res 12:1680–1685 Harder J, Waiz O, Otto F et al, 2009, EGFR and HER2 expression in advanced biliary tract cancer. World J Gastroenterol 15:4511– 4517 Zhou YM, Li YM, Cao N, Feng Y, Zeng F, 2003, Significance of expression of epidermal growth factor, EGF, and its receptor, EGFR, in chronic cholecystitis and gallbladder carcinoma. Ai Zheng 22:262–265 Hui AM, Li X, Shi YZ, Takayama T, Torzilli G, Makuuchi M, 2000, Cyclin D1 overexpression is a critical event in gallbladder carcinogenesis and independently predicts decreased survival for patients with gallbladder carcinoma. Clin Cancer Res 6:4272–4277 Eguchi N, Fujii K, Tsuchida A, Yamamoto S, Sasaki T, Kajiyama G, 1999, Cyclin E overexpression in human gallbladder carcinomas. Oncol Rep 6:93–96 Hui AM, Xin L, Shi YZ, Takayama T, Torzilli G, Makuuchi M, 2000, Cyclin D1 overexpression is a critical event in gallbladder carcinogenesis and independently predicts decreased survival for patients with gallbladder carcinoma. Clin Cancer Res 6:4272–4277 Hanada K, Itoh M, Fujii K, Tsuchida A, Ooishi H, Kajiyama G, 1996, K-ras and p53 mutations in stage I gallbladder carcinoma with an anomalous junction of the pancreaticobiliary duct. Cancer 77:452–458 Fujii K, Yokozaki H, Yasui Wet al, 1996, High frequency of p53 gene mutation in adenocarcinomas of the gallbladder. Cancer Epidemiol Biomarkers Prev 5:461–466 Hanada K, Itoh M, Fujii K et al, 1997, TP53 mutations in stage I gallbladder carcinoma with special attention to growth patterns. Eur J Cancer 33:1136–1140 Itoi T, Watanabe H, Yoshida M, Ajioka Y, Nishikura K, Saito T, 1997, Correlation of p53 protein expression with gene mutation in gall-bladder carcinomas. Pathol Int 47:525–530 Yokoyama N, Hitomi J, Watanabe H et al, 1998, Mutations of p53 in gallbladder carcinomas in high-incidence areas of Japan and Chile. Cancer Epidemiol Biomarkers Prev 7:297–301 Pfeifer GP, 2000, p53 mutational spectra and the role of methylated CpG sequences. Mutat Res 450:155–166 Xuan YH, ChoiYL, ShinYKet al, 2005)An immunohistochemical study of the expression of cell-cycle-regulated proteins p53, cyclin D1, RB, p27, Ki67 and MSH2 in GBC and its precursor lesions. Histol Histopathol 20:59–66 Wistuba II, Albores-Saavedra J, 1999, Genetic abnormalities involved in the pathogenesis of gallbladder carcinoma. J Hepatobiliary Pancreat Surg 6:237–244 Campomenosi P, Conio M, BoglioloMet al, 1996, p53 is frequently mutated in Barrett’s metaplasia of the intestinal type. Cancer Epidemiol Biomarkers Prev 5:559–565 Hussain SP, Amstad P, Raja K et al, 2000, Increased p53 mutation load in noncancerous colon tissue from ulcerative colitis: a cancerprone chronic inflammatory disease. Cancer Res 60:3333–3337 Roa I,Villaseca M, Araya J et al, 1997, p53 tumour suppressor gene protein expression in early and advanced gallbladder carcinoma. Histopathology 31:226–230 Billo P, Marchegiani C, Capella C, Sessa F, 2000, Expression of p53 in GBC and in dysplastic and metaplastic lesions of the surrounding mucosa. Pathologica 92:249–256 Roa JC, Vo Q, Araya JC, Villaseca M, Guzman P, Ibacache GS, de Aretxabala X, Roa I, 2004, Inactivation of CDKN2A gene, p16, in gallbladder carcinoma]. Rev Med Chil 132:1369–1376 Quan ZW, Wu K, Wang J, Shi W, Zhang Z, Merrell RC, 2001, Association of p53, p16, and vascular endothelial growth factor protein expressions with the prognosis and metastasis of gallbladder cancer. J Am Coll Surg 193:380–383 Shi YZ, Hui AM, Li X, Takayama T, Makuuchi M, 2000, Overexpression of retinoblastoma protein predicts decreased survival and correlates with loss of p16INK4 protein in gallbladder carcinomas. Clin Cancer Res 6:4096–4100 Li X, Hui AM, Shi YZ, Takayama T, Makuuchi M, 2001, Reduced p21, WAF1/CIP1, expression is an early event in gallbladder carcinogenesis and is of prognostic significance for patients with carcinomas of the gallbladder. Hum Pathol 32:771–777 Puhalla H, Wrba F, Kandioler D et al, 2007, Expression of p21(Wafl/Cip1), p57(Kip2, and HER2/neu in patients with gallbladder cancer. Anticancer Res 27:1679–1684 Wistuba II,Ashfaq R, Maitra A,AlvarezH, Riquelme E,Gazdar AF et al, 2002, Fragile histidine triad gene abnormalities in the pathogenesis of gallbladder carcinoma. Am J Pathol 160:2073–2079 Koda M, Yashima K, Kawaguchi K et al, 2003, Expression of Fhit, Mlh1, and P53 protein in human gallbladder carcinoma. Cancer Lett 199:131–138 TangM, Baez S, Pruyas M, Diaz A, Calvo A, Riquelme E,Wistuba II et al, 2004, Mitochondrial DNA mutation at the D310, displacement loop, mononucleotide sequence in the pathogenesis of gallbladder carcinoma. Clin Cancer Res 10:1041–1046 Asano T, Shoda J, Ueda T et al, 2002, Expressions of cyclooxygenase-2 and prostaglandin E-receptors in the gallbladder: crucial role of arachidonate metabolism in growth and progression. Clin Cancer Res 8:1157–1167 Zhi YH, Liu RS, Song MM et al, 2005, Cyclooxygenase-2 promotes angiogenesis by increasing vascular growth factor and predicts prognosis in gallbladder carcinoma.World J Gastroenterol 11: 3724–3728 Zhang M, Pan JW, Ren TR, Zhu YF, Han YJ, Kuhnel W, 2003, Correlated expression of inducible nitric oxide synthase and P53, Bax in benign and malignant diseased gallbladder. Ann Anat 185:549–554 Holczbauer A, Gyongyosi B, Lotz G, Torzsok P, Kaposi-Novak P, Szijarto A et al, 2014, Increased expression of claudin-1 and claudin-7 in liver cirrhosis and hepatocellular carcinoma. Pathol Oncol Res 20:493–502 Nemeth Z, Szasz AM, Tatrai P, Nemeth J, Gyorffy H, Somoracz A et al, 2009, Claudin-1, −2, −3, −4, −7, −8, and −10 protein expression in biliary tract cancers. J Histochem Cytochem 57:113–21 Roa I, Villaseca M, Araya J, Roa J, de Aretxabala X, Ibacache G et al, 2001, CD44, HCAM, expression in subserous gallbladder carcinoma. Rev Med Chil 129:727–734 Ylagan LR, Scholes J, Demopoulos R, 2000, Cd44: a marker of squamous differentiation in adenosquamous neoplasms. Arch Pathol Lab Med 124:212–215 Chang HJ, Jee CD, Kim WH, 2002, Mutation and altered expression of beta-catenin during gallbladder carcinogenesis. Am J Surg Pathol 26:758–766 Yanagisawa N, Mikami T, Saegusa M, Okayasu I, 2001, More frequent beta-catenin exon 3 mutations in gallbladder adenomas than in carcinomas indicate different lineages. Cancer Res 61:19–22 Choi YL,XuanYH, ShinYKet al, 2004)An immunohistochemical study of the expression of adhesion gallbladder lesions. J Histochem Cytochem 52:591–601 Sasaki M, Yamato T, Nakanuma Y, Ho SB, Kim YS, 1999, Expression of MUC2, MUC5AC and MUC6 apomucins in carcinoma, dysplasia and non-dysplastic epithelia of the gallbladder. Pathol Int 49:38–44 Kashiwagi H, Kijima H, Dowaki S et al, 2001, MUC1 and MUC2 expression in human gallbladder carcinoma: a clinicopathological study and relationship with prognosis. Oncol Rep 8:485–489 GhoshM, Kamma H, Kawamoto T, Koike N,MiwaM, Kapoor VK et al, 2005, MUC 1 core protein as a marker of gallbladder malignancy. Eur J Surg Oncol 31:891–896 Lee HK, Cho M, Kim TH, 2012, Prognostic significance of muc4 expression in gallbladder carcinoma. World J Surg Oncol 10:224 Garcia P,Manterola C, Araya JC, Villaseca M, Guzman P, Sanhueza A et al, 2009, Promoter methylation profile in preneoplastic and neoplastic gallbladder lesions. Mol Carcinog 48:79–89 Luzar B, Poljak M, Cor A, Klopcic U, Ferlan-Marolt V, 2005, Expression of human telomerase catalytic protein in gallbladder carcinogenesis. J Clin Pathol 58:820–825 Wistuba II, Maitra A, Carrasco R et al, 2002, High resolution chromosome 3p, 8p, 9q and 22q allelotyping analysis in the pathogenesis of gallbladder carcinoma. Br J Cancer 87:432–440 Saetta AA, Papanastasiou P, Michalopoulos NV, Gigelou F, Korkolopoulou P, Bei T et al, 2004, Mutational analysis of BRAF in gallbladder carcinomas in association with K-ras and p53 mutations and microsatellite instability. Virchows Arch 445:179–82 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 509 EP 525 DI 10.1007/s12253-014-9886-3 PG 17 ER PT J AU Fernandes, VJ Cobucci, NOR Jatoba, ANC de Medeiros Fernandes, AATh de Azevedo, WVJ de Araujo, MGJ AF Fernandes, Verissimo Jose Cobucci, Ney Oliveira Ricardo Jatoba, Andre Nunes Carlos de Medeiros Fernandes, Allyrio Araujo Thales de Azevedo, Welber Verissimo Judson de Araujo, Maria Galvao Joselio TI The Role of the Mediators of Inflammation in Cancer Development SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Inflammation and cancer; Inflammation mediators; Mechanisms of tumorigenesis ID Inflammation and cancer; Inflammation mediators; Mechanisms of tumorigenesis AB Epigenetic disorders such as point mutations in cellular tumor suppressor genes, DNA methylation and posttranslational modifications are needed to transformation of normal cells into cancer cells. These events result in alterations in critical pathways responsible for maintaining the normal cellular homeostasis, triggering to an inflammatory response which can lead the development of cancer. The inflammatory response is a universal defense mechanism activated in response to an injury tissue, of any nature, that involves both innate and adaptive immune responses, through the collective action of a variety of soluble mediators. Many inflammatory signaling pathways are activated in several types of cancer, linking chronic inflammation to tumorigenesis process. Thus, Inflammatory responses play decisive roles at different stages of tumor development, including initiation, promotion, growth, invasion, and metastasis, affecting also the immune surveillance. Immune cells that infiltrate tumors engage in an extensive and dynamic crosstalk with cancer cells, and some of the molecular events that mediate this dialog have been revealed. A range of inflammation mediators, including cytokines, chemokines, free radicals, prostaglandins, growth and transcription factors, microRNAs, and enzymes as, cyclooxygenase and matrix metalloproteinase, collectively acts to create a favorable microenvironment for the development of tumors. In this review are presented the main mediators of the inflammatory response and discussed the likely mechanisms through which, they interact with each other to create a condition favorable to development of cancer. C1 [Fernandes, Verissimo Jose] Federal University of Rio Grande do Norte, Post-Graduate Program in Biological SciencesNatal, RN, Brazil. [Cobucci, Ney Oliveira Ricardo] Federal University of Rio Grande do Norte, Maternidade Escola Januario CiccoNatal, RN, Brazil. [Jatoba, Andre Nunes Carlos] Federal University of Rio Grande do Norte, Department of PathologyNatal, RN, Brazil. [de Medeiros Fernandes, Allyrio Araujo Thales] University of Rio Grande do Norte State, Department of Biomedical SciencesMossoro, RN, Brazil. [de Azevedo, Welber Verissimo Judson] Federal University of Triangulo Mineiro, Hospital UniversitarioMina Gerais, Brazil. [de Araujo, Maria Galvao Joselio] Federal University of Rio Grande do Norte, Post-Graduate Program in Biological SciencesNatal, RN, Brazil. RP Fernandes, VJ (reprint author), Federal University of Rio Grande do Norte, Post-Graduate Program in Biological Sciences, Natal, Brazil. EM joseverissimo1951@gmail.com CR Martinon F,Mayor A, Tschopp J, 2009, The inflammasomes: guardians of the body. Annu Rev Immunol 27:229–265 Ashley NT,Weil ZM, Nelson RJ, 2012, Inflammation: mechanisms, costs, and natural variation. Annu Rev Ecol Evol Syst 43:385–406 Bermejo-Martin JF, Martin-Loeches I, Bosinger S, 2014, Inflammation and infection in critical care medicine. Mediat Inflamm 2014:456256., DOI 10.1155/2014/456256 Eiro N, Vizoso FJ, 2012, Inflammation and cancer. World J Gastrointest Surg 4(3):62–72 Campbell LM, Maxwell PJ, Waugh DJ, 2013, Rationale and means to target pro-inflammatory interleukin-8, CXCL8, signaling in cancer. Pharmaceuticals 6(8):929–959 Kaushal N, Kudva AK, 2013, Oxidative stress and inflammation: the lesser of two evils in carcinogenesis. PostDoc J 1(2):89–101 Parks WC, Wilson CL, Lopez-Boado YS, 2004, Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol 4(8):617–629 Hussain SP, Harris CC, 2007, Inflammation and cancer: an ancient link with novel potentials. Int J Cancer 121(11):2373–2380 Adefuye A, Sales K, 2012, Regulation of inflammatory pathways in cancer and infectious disease of the cervix. Scientifica, Cairo, 2012: 548150 Allavena P, Germano G, Marchesi F, Mantovani A, 2011, Chemokines in cancer related inflammation. Exp Cell Res 317(5): 664–673 Raman D, Baugher PJ, Thu YM, Richmond A, 2007, Role of chemokines in tumor growth. Cancer Lett 256(2):137–165 Hoesel B, Schmid JA, 2013, The complexity of NF-κB signaling in inflammation and cancer. Mol Cancer 12:86., DOI 10.1186/1476- 4598-12-86 Liang Y, Zhou Y, Shen P, 2004, NF-kappaB and its regulation on the immune system. Cell Mol Immunol 1(5):343–350 Wu LJ, Li HX, Luo XT et al, 2014, STAT3 activation in tumor cellfree lymph nodes predicts a poor prognosis for gastric cancer. Int J Clin Exp Pathol 7(3):1140–1146 Fan Y,Mao R, Yang J, 2013, NF-κB and STAT3 signaling pathways collaboratively link inflammation to cancer. Protein Cell 4(3):176– 185 Gomis-Ruth FX, 2009, Catalytic domain architecture of metzincin metalloproteases. J Biol Chem 284(23):15353–15357 Loffek S, Schilling O, Franzke CW, 2011, Biological role of matrix metalloproteinases: a critical balance. Eur Respir J 38:191–208 Walter L, Harper C, Gar P, 2013, Role of matrix metalloproteinases in inflammation/colitis-associated colon cancer. Immuno- Gastroenterology 2(1):22–28 Chen Q, Jin M, Yang F, Zhu J, Xiao Q, Zhang L, 2013, Matrix metalloproteinases: inflammatory regulators of cell behaviors in vascular formation and remodeling. Mediat Inflamm 2013:928315., DOI 10.1155/2013/928315 Butler GS, 2000, Overall CM, 2013, Matrix metalloproteinase processing of signaling molecules to regulate inflammation. Periodontol 63(1):123–148 Said AH, Raufman JP, Xie G, 2014, The role of matrix metalloproteinases in colorectal cancer. Cancers 6(1):366–375 Zyada MM, Shamaa AA, 2008, Is collagenase-3, MMP-13, expression in chondrosarcoma of the jaws a true marker for tumor aggressiveness? Diagn Pathol 3:26., DOI 10.1186/1746-1596-3-26 Zha S, Yegnasubramanian V, NelsonWG, IsaacsWB, DeMarzo AM, 2004, Cyclooxygenases in cancer: progress and perspective. Cancer Lett 215(1):1–20 Cathcart MC, O’Byrne KJ, Reynolds JV, O’Sullivan J, Pidgeon GP, 2012, COX-derived prostanoid pathways in gastrointestinal cancer development and progression: novel targets for prevention and intervention. Biochim Biophys Acta 1825(1):49–63 Asting AG, Caren H, Andersson M, Lonnroth C, Lagerstedt K, Lundholm K, 2011, COX-2 gene expression in colon cancer tissue related to regulating factors and promoter methylation status. BMC Cancer 11:238., DOI 10.1186/1471-2407-11-238 Wang D, Dubois RN, 2010, The Role of COX-2 in intestinal inflammation and colorectal cancer. Oncogene 29(6):781–788 Sharkey DJ, Tremellen KP, Jasper MJ, Gemzell-Danielsson K, Robertson SA, 2012, Seminal fluid induces leukocyte recruitment and cytokine and chemokine mRNA expression in the human cervix after coitus. J Immunol 188(5):2445–2454 Sales KJ, Katz AA, Howard B et al, 2002, Cyclooxygenase-1 is upregulated in cervical carcinomas: autocrine/paracrine regulation of cyclooxygenase-2, prostaglandin e receptors, and angiogenic factors by cyclooxygenase-1. Cancer Res 62:424–432 zur Hausen H, 2009, Papillomaviruses in the causation of human cancers - a brief historical account. Virology 384(2):260–265 Ahmad J, Hasnain SE, Siddiqui MA, Ahamed M, Musarrat J, Al- Khedhairy AA, 2013, MicroRNA in carcinogenesis & cancer diagnostics: a new paradigm. Indian J Med Res 137(4):680–694 Croce CM, 2009, Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet 10:704–714 Ostermann E, Laventie JR, Pfeffer S, Bahram S, Sprauel PS, Georgel P, 2013, MicroRNAs: Fine-turners of type-I interferon-dependent inflammatory responses. Curr Trends Immunol 14:35–44 Tili E,Michaille JJ, CroceCM(2013)MicroRNAs play a central role in molecular dysfunctions linking inflammation with cancer. Immunol Rev 253(1):167–184 Shivdasani RA, 2006, MicroRNAs: regulators of gene expression and cell differentiation. Blood 108(12):3646–3653 Yeung ML, Jeang KT, 2011, MicroRNAs and cancer therapeutics. Pharm Res 28(12):3043–3049 O’Connell RM, Rao DS, Baltimore D, 2012, MicroRNA regulation of inflammatory responses. Annu Rev Immunol 30:295–312 Chiba T, Marusawa H, Ushijima T, 2012, Inflammation-associated cancer development in digestive organs: mechanisms and roles for genetic and epigenetic modulation. Gastroenterology 143(3):550– 563 Schetter AJ, Okayama H, Harris CC, 2012, The role of microRNAs in colorectal cancer. Cancer J 18:244–252 Tang T,Wong HK, GuWet al, 2013)MicroRNA-182 plays an oncomiRNA role in cervical cancer. Gynecol Oncol 129(1):199–208 Zhou Q, Haupt S, Kreuzer JT et al, 2014, Decreased expression of miR-146a and miR-155 contributes to an abnormal Treg phenotype in patients with rheumatoid arthritis. Ann Rheum Dis., DOI 10.1136/ annrheumdis-2013-204377 Iliopoulos D, 2014, MicroRNA circuits regulate the cancerinflammation link. Sci Signal 7(318):pe8., DOI 10.1126/scisignal. 2005053 Josse C, Bouznad N, Geurts P et al, 2014, Identification of a microRNA landscape targeting the PI3K/Akt signaling pathway in inflammation-induced colorectal carcinogenesis. Am J Physiol Gastrointest Liver Physiol 306(3):G229–G243 Schauer IG, Zhang J, Xing Z, Guo X, Mercado-Uribe I, Sood AK, Huang P, Liu J, 2013, Interleukin-1β promotes ovarian tumorigenesis through a p53/NF-κB-mediated inflammatory response in stromal fibroblasts. Neoplasia 15(4):409–420 Lou JL, Maeda S, Hsu LC, Yagita H, Karin M, 2004, Inhibition of NF-kappaB in cancer cells converts inflammation-induced tumor growth mediated by TNF-α to TRAIL-mediated tumor regression. Cancer Cell 6(3):297–305 Waters JP, Pober JS, Bradley JR, 2013, Tumour necrosis factor and cancer. J Pathol 230(3):241–248 Hussain SP, Hofseth LJ, Harris CC, 2003, Radical causes of cancer. Nat Rev Cancer 3(4):276–285 Lebrun JJ, 2012, The dual role of TGF-β in human cancer: from tumor suppression to cancer metastasis. ISRN Mol Biol 2012: 381428., DOI 10.5402/2012/ 381428 Elliott RL, Blobe GC, 2005, Role of transforming growth factor beta in human cancer. J Clin Oncol 23(9):2078–2093 ChenW, Konkel JE, 2010, TGF-beta and ‘adaptive’ Foxp3(+, regulatory T cells. J Mol Cell Biol 2(1):30–36 Visperas A, Do JS, Bulek K, Li X, Min B, 2014, IL-27, targeting antigen-presenting cells, promotes Th17 differentiation and colitis in mice. Mucosal Immunol 7(3):625–633 Guo Y, Xu F, Lu T, Duan Z, Zhang Z, 2012, Interleukin-6 signaling pathway in targeted therapy for cancer. Cancer Treat Rev 38(7):904– 910 Vicari AP, Trinchieri G, 2004, Interleukin-10 in viral diseases and cancer: exiting the labyrinth? Immunol Rev 202:223–236 Mocelin S, Marincola FM, Young HA, 2005, Interleukin-10 and the immune response against cancer: a counterpoint. J Leukoc Biol 78: 1043–1051 Driessler F, Venstrom K, Sabat R, Asadullah K, Schottelius AJ, 2004, Molecular mechanisms of interleukin-10-mediated inhibition of NF-kappaB activity: a role for p50. Clin Exp Immunol 135(1):64– 73 Pattison MJ, MacKenzie KF, Arthur JS, 2012, Inhibition of JAKs in macrophages increases lipopolysaccharide-induced cytokine production by blocking IL-10-mediated feedback. J Immunol 189(6):2784– 2792 Sato T, Terai M, Tamura Y, Alexeev V, Mastrangelo MJ, Selvan SR, 2011, Interleukin 10 in the tumor microenvironment: a target for anticancer immunotherapy. Immunol Res 51(2–3):170–82 Cope A, Le Friec G, Cardone J, Kemper C, 2011, The Th1 life cycle: molecular control of IFN-γ to IL-10 switching. Trends Immunol 32(6):278–286 Stewart CA,Metheny H, Iida N et al, 2013, Interferon-dependent IL- 10 production by Tregs limits tumor Th17 inflammation. J Clin Invest 123(11):4859–4874 Shigehara K, Shijubo N, OhmichiMet al, 2001, IL-12 and IL-18 are increased and stimulate IFN-γ production in sarcoid lungs. J Immunol 166(1):642–649 Wong JL, Berk E, Edwards RP, Kalinski P, 2013, IL-18-primed helper NK cells collaborate with dendritic cells to promote recruitment of effector CD8+ T cells to the tumor microenvironment. Cancer Res 73(15):4653–4662 Srivastava S, Salim N, Robertson MJ, 2010, Interleukin-18: biology and role in the immunotherapy of cancer. Curr Med Chem 17(29): 3353–3357 Palma G, Barbieri A, Bimonte S, Palla M, Zappavigna S, Caraglia M, Ascierto PA, Ciliberto G, Arra C, 2013, Interleukin 18: friend or foe in cancer. Biochim Biophys Acta 1836:296–303 Miossec P, 2009, IL-17 and Th17 cells in human inflammatory diseases. Microbes Infect 11(5):625–630 Xiang T, Long H, He L, Han X, Lin K, Liang Z, ZhuoW, Xie R, Zhu B, 2013, Interleukin-17 produced by tumor microenvironment promotes self-renewal of CD133+ cancer stem-like cells in ovarian cancer. Oncogene., DOI 10.1038/onc.2013.537 Chen WC, Lai YH, Chen HY, Guo HR, Su IJ, Chen HH, 2013, Interleukin-17-producing cell infiltration in the breast cancer tumour microenvironment is a poor prognostic factor. Histopathology 63(2): 225–233 Chen Z, Ding J, Pang N, Du R, Meng W, Zhu Y, Zhang Y, Ma C, Ding Y, 2013, The Th17/Treg balance and the expression of related cytokines in Uygur cervical cancer patients. Diagn Pathol 8:61., DOI 10.1186/1746-1596-8-61 Ngiow SF, Teng MW, Smyth MJ, 2013, Balance of interleukin-12 and −23 in cancer. Trends Immunol 34(11):548–555 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 527 EP 534 DI 10.1007/s12253-015-9913-z PG 8 ER PT J AU Bianchi, S Bendinelli, B Saladino, V Vezzosi, V Brancato, B Nori, J Palli, D AF Bianchi, Simonetta Bendinelli, Benedetta Saladino, Valeria Vezzosi, Vania Brancato, Beniamino Nori, Jacopo Palli, Domenico TI Non-Malignant Breast Papillary Lesions - B3 Diagnosed on Ultrasound - Guided 14-Gauge Needle Core Biopsy: Analysis of 114 Cases from a Single Institution and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary lesions; 14-gauge needle core biopsy; Epithelial atypia; Excision; Associated carcinoma ID Papillary lesions; 14-gauge needle core biopsy; Epithelial atypia; Excision; Associated carcinoma AB One-hundred-fourteen consecutive cases of breast ultrasound-guided 14-gauge needle core biopsy (14G NCB) performed from January 2001 to June 2013 and diagnosed as non-malignant papillary lesion (PL)-B3, were reviewed and compared with definitive histological diagnosis on surgical excision (SE) to evaluate the diagnostic accuracy of ultrasound-guided 14G NCB. PL with epithelial atypia on 14G NCB were associated to malignancy on definitive histological diagnosis on SE in 22 (7 DCIS and 15 invasive carcinomas) of 46 cases with an underestimation rate of 47.8 %, while 9 (4 DCIS and 5 invasive carcinomas) cases out of 68 cases of PL without epithelial atypia were upgraded to carcinoma with an underestimation rate of 13.2 %. In cases of PL with epithelial atypia on ultrasound-guided 14G NCB, SE appears mandatory due to the high risk of associated malignancy. The diagnosis of PL without epithelial atypia on ultrasound-guided 14G NCB does not exclude malignancy at subsequent SE, consequently further assessment (by surgical or vacuum-assisted excision) is recommended to avoid the risk of delaying a diagnosis of malignancy, although this tends to be lower (1 in 8 patients). C1 [Bianchi, Simonetta] AOU Careggi, Department of Surgery and Translational Medicine, Largo G.A. Brambilla 3, 50134 Florence, Italy. [Bendinelli, Benedetta] Cancer Research and Prevention Institute (ISPO), Molecular and Nutritional Epidemiology UnitFlorence, Italy. [Saladino, Valeria] AOU Careggi, Department of Surgery and Translational Medicine, Largo G.A. Brambilla 3, 50134 Florence, Italy. [Vezzosi, Vania] AOU Careggi, Department of Surgery and Translational Medicine, Largo G.A. Brambilla 3, 50134 Florence, Italy. [Brancato, Beniamino] Cancer Research and Prevention Institute (ISPO), Senology UnitFlorence, Italy. [Nori, Jacopo] Careggi University Hospital, Diagnostic Senology UnitFlorence, Italy. [Palli, Domenico] Cancer Research and Prevention Institute (ISPO), Molecular and Nutritional Epidemiology UnitFlorence, Italy. RP Bianchi, S (reprint author), AOU Careggi, Department of Surgery and Translational Medicine, 50134 Florence, Italy. EM simonetta.bianchi@unifi.it CR Lakhani SR, Ellis IO, Schnitt SJ, Hoon Tan P, van de VijverMJ, eds,, 2012, WHO classification of tumours of the breast. International Agency for Research on Cancer, Lyon Ouchi N, Abe R, Kasai M, 1984, Possible cancerous change of intraductal papillomas of the breast. A 3- D reconstruction study of 25 cases. Cancer 54:605–611 NHS Breast Screening Programme and the Royal College of Pathologists., 2005, Guidelines for pathology reporting in breast disease. NHSBSP Publication No 58. www.cancerscreening.nhs.uk European Commission Working Group on Breast Screening Pathology, 2006, Quality assurance guidelines for pathology. In: Perry N, Broeders M, de Wolf C, et al. editors. European guidelines for quality assurance in breast cancer screening and diagnosis. Luxemburg: Office for Official publications of the European Communities; Fourth Edition Puglisi F, Zuiani C, BazzocchiM, Valent F, Aprile G, Pertoldi B et al, 2003, Role of mammography ultrasound and large core biopsy in the diagnostic evaluation of papillary breast lesions. Oncology 65: 311–315 Maxwell AJ, Makata G, Pearson JM, 2013, Benign papilloma diagnosed on image-guide 14 G core biopsy of the breast: effect of lesion type on likelihood of malignancy at excision. Clin Radiol 68:383– 387 Koo JS, Han K, Kim MJ, Moon HJ, Kim EK, Park BW, 2013, Can additional immunohistochemistry staining replace the surgical excision for the diagnosis of papillary breast lesions classified as benign on 14-gauge core needle biopsy? Breast Cancer Res Treat 137:797– 806 Liberman L, Bracero N, Vuolo MA, Dershaw DD, Morris EA, Abramson AF et al, 1999, Percutaneous large-core biopsy of papillary breast lesions. AJR Am J Roentgenol 172:331–337 IoffeO, BergWA, Silverberg SG, 2000, Analysis of papillary lesions diagnosed on core needle biopsy: management implications. Mod Pathol 13:23A Philpotts LE, Shaheen NA, Jain KS, Carter D, Lee CH, 2000, Uncommon high-risk lesions of the breast diagnosed at stereotactic core-needle biopsy:clinical importance. Radiology 216:831–837 Mercado CL, Hamele-Bena D, Singer C, Koenigsberg T, Pile- Spellman E, Higgins H et al, 2001, Papillary lesions of the breast: evaluation with stereotactic directional vacuum-assisted biopsy. Radiology 221:650–655 Rajendiran S, Gupta D, Sumkin J, Johnsonn R, Nath M, 2001, Correlation of image-guided core and excision biopsy of papillary lesions of the breast, abstract). Mod Pathol 14:34A Rosen EL, Bentley RC, Baker JA, Soo MS, 2002, Imaging-guided core needle biopsy of papillary lesions of the breast. Am J Roentgenol 179:1185–1192 Masood S, Loya A, Khalbuss W, 2003, Is core needle biopsy superior to fine-needle aspiration biopsy in the diagnosis of papillary breast lesions? Diagn Cytopathol 28:329–334 Agoff SN, Lawton TJ, 2004, Papillary lesions of the breast with and without atypical ductal hyperplasia. Am J Clin Pathol 122:440–443 Ivan D, Selinko V, Sahin AA, Sneige N, Middleton LP, 2004, Accurancy of core needle biopsy diagnosis in assessing papillary breast lesions: histologic predictors of malignancy. Mod Pathol 17: 165–171 Renshaw AA, Derhagopian RP, Tizol-Blanco DM, Gould EW(2004, Papillomas and atypical papillomas in breast core needle biopsy specimens: risk of carcinoma in subsequent excision. Am J Clin Pathol 122:217–221 Carder PJ, Garvican J, Haigh I, Liston JS, 2005, Needle core biopsy can reliably distinguish between benign and malignant papillary lesions of the breast. Histopathology 46:320–327 Shah VI, Flowers CI, Douglas-Jones AG, Dallimore NS, Rashid M, 2006, Immunohistochemistry increases the accuracy of diagnosis of benign papillary lesions in breast core needle biopsy specimens. Histopathology 48:683–691 Arora N, Hill C, Hoda SA, Rosenblatt R, Pigalarga R, Tousimis EA, 2007, Clinicopathologic features of papillary lesions on core needle biopsy of the breast predictive of malignancy. Am J Surg 194:444– 449 Ashkenazi I, Ferrer K, Sekosan M, Marcus E, Bork J, Aiti T et al, 2007, Papillary lesions of the breast discovered on percutaneous large core and vacuum-assisted biopsies: reliability of clinical and pathological parameters in identifying benign lesions. Am J Surg 194:183–188 Ko ES, Cho N, Cha JH, Park JS, Kim SM, Moon WK, 2007, Sonographically-guided 14-gauge core needle biopsy for papillary lesions of the breast. Korean J Radiol 8:206–211 Sohn V, Keilock J, Arthus Z, Wilson A, Herbert G, Perry J et al, 2002, Breast papilloma in the era of percutaneous needle biopsy. Surg Oncol 14:2979–2984 Sydnor MK, Wilson JD, Hijaz TA, Massey HD, de Paredes ES S, 2007, Underestimation of the presence of breast carcinoma in papillary lesions initially diagnosed at core-needle biopsy. Radiology 242:58–62 Kil WH, Cho EY, Kim JH, Nam SJ, Yang JH, 2008, Is surgical excision necessary in benign papillary lesions initially diagnosed at core biopsy? Breast 17:258–262 RizzoM, Lund MJ, Oprea G, Schniederjan M,WoodWC,Mosunjac M, 2008, Surgical follow-up and clinical presentation of 142 breast papillary lesions diagnosed by ultrasound-guided core needle biopsy. Ann Surg Oncol 15:1040–1047 Sakr R, Rouzier R, Salem C, Antoine M, Choipier J, Darai E et al, 2008, Risk of breast cancer associated with papilloma. J Cancer Surg 34:1304–1308 Shin HJ, Kim HH, Kim SM, Yang HR, Sohn JH, Kwon GY et al, 2008, Papillary lesions of the breast diagnosed at percutaneous sonographically guided biopsy: comparison of sonographic features and biopsy methods. Am J Roentgenol 190:630–636 Ahmadiyeh N, Stoleru MA, Raza S, Lester SC, Golshan M, 2009, Management of intraductal papillomas of the breast: an analysis of 129 cases and their outcome. Ann Surg Oncol 16:2264–2269 Bernik SF, Troob S, Ying BL, Simpson SA, Axelord DM, Siegel B et al, 2009, Papillary lesions of the breast diagnosed by core needle biopsy: 71 cases with surgical follow-up. Am J Surg 197:473–478 BodeMK, Rissanen T, Apaja-SarkkinenM(2009, Ultrasonographyguided core needle biopsy in differential diagnosis of papillary breast tumors. Acta Radiol 7:722–729 Cheng TY, Chen CM, Lee MY, Lin KJ, Hung CF, Yang PS et al, 2009, Risk factors associated with conversion from nonmalignant to malignant diagnosis after surgical excision of breast papillary. Ann Surg Oncol 16:3375–3379 Tseng HS, Chen YL, Chen ST,Wu YC, Kuo SJ, Chen LS et al, 2009, The management of papillary lesion of the breast by core needle biopsy. J Cancer Surg 35:21–24 Bennet LE, Ghate SV, Bentley R, Baker JA, 2010, Is surgical excision of core biopsy proven benign papillomas of the breast necessary? Acad Radiol 17:553–557 Tse GM, Tan PH, LacambraMD, Jara-Lazaro AR, Chan SK, Lui PC et al, 2009, Papillary lesions of the breast accuracy of core biopsy. Histopathology 56:481–488 Youk JH, Kim EK, Kwak JY, Son EJ, Park BW, Kim SII, 2011, Benign papilloma without atypia diagnosed at us-guided 14-gauge core-needle biopsy: clinical and us features predictive of upgrade to malignancy. Radiology 258:81–88 Chang JM, HanW, MoonWK, Cho N, Noh DY, Park IA et al, 2011, Papillary lesions initially diagnosed at ultrasound-guided vacuumassisted breast biopsy: rate of malignancy based on subsequent surgical excision. Ann Surg Oncol 18:2506–2514 KimMJ,Kim SI,Youk JH, Moon HJ,Kwak JY, ParkBWet al, 2011, The diagnosis of non malignant papillary lesions of the breast: comparison of ultrasound-guided automated gun biopsy and vacuumassisted removal. Clin Radiol 66:530–535 Rakha EA, Lee AHS, Jenkins JA, Murphy AE, Hamilton LJ, Ellis IO, 2011, Characterization and outcome of breast needle core biopsy diagnoses of lesions of uncertain malignant potential, B3, in abnormalities detected by mammographic screening. Int J Cancer 129: 1417–1424 Richter-Ehrenstein C, Tombokan F, Fallenberg EM, Schneider A, Denkert C, 2011, Intraductal papillomas of the breast: diagnosis and management of 151 patients. Breast 20:501–504 Destounis S, Seifert P, Somerville P,Murphy P,Morgan R, Arieno A, Young WL, 2011, Underestimation of papillary breast lesions by core biopsy: correlation to surgical excision. Breast Cancer 18:42–50 Fu CY, Chen TW, Hong ZJ, Chan DC, Young CY, Chen CJ et al, 2012, Papillary breast lesions diagnosed by core biopsy require complete excision. EJSO 38:1029–1035 Holley SO, Appleton CM, Farria DM, Reichert VC, Warrik J, Craig Allred D et al, 2012, Patologic outcomes of nonmalignant papillary breast lesions diagnosed at imaging-guided core needle biopsy. Radiology 265:379–384 Lu Q, Tan EY, Ho B, Chen JJC, Chan PMY, 2012, Surgical excision of intraductal breast papilloma diagnosed on core biosy. ANZ J Surg 82:168–172 Rizzo M, Linebarger J, Lowe MC, Pan L, Gabram SG, Vasquez L et al, 2012, Management of papillary breast lesions diagnosed on core-needle biopsy:clinical pathologic and radiologic analysis of 276 cases with surgical follow-up. J Am Coll Surg 214:280–287 Al Hassan T, Delli Fraine P, El-Khoury M, Joseph L, Zheng J, Mesurolle B, 2013, Accuracy of percutaneous core needle biopsy in diagnosing papillary breast lesions and potential impact of sonographic features on their management. J Clin Ultrasound 41:1–9 Wiratkapun C, Keeratitragoon T, Lertsithichai P, Chanplakorn N, 2013, Upgrading rate of papillary breast lesions diagnosed by core-needle biopsy. Diagn Interv Radiol 19:361–367 BazzocchiM, Berra I, Francescutti GE, Del Frate C, Zuiani C, Puglisi F et al, 2001, Papillary lesions of the breast: diagnostic imaging and contribution of percutaneous needle biopsy with 14G needle. Radiol Med 101:424–431 Irfan K, Brem RF, 2002, Surgical and mammographic follow-up of papillary lesions and atypical lobular hyperplasia diagnosed with stereotactic vacuum-assisted biopsy. Breast J 8:230–233 Jacobs TW, Connoly JL, Shnitt SJ, 2002, Nonmalignant lesions in a breast core needle biopsies: to excise or not excise? Am J Surg Pathol 26:1095–1110 Gendler LS, Feldman SM, Balassanian R, Riker MA, Frencher SK, Whelan DB et al, 2004, Association of breast cancer with papillary lesions identified at percutaneous image-guided breast biopsy. Am J Surg 188:365–370 LamWW, ChuWC, Tang AP, Tse G, Ma TK, 2006, Role of radiologic features in the management of papillary lesions of the breast. Am J Roentgenol 196:1322–1327 Libermann L, Tornos C, Huzjan R, Bartella L, Morris EA, Dershaw DD, 2006, Is surgical excision warranted after benign, concordant diagnosis of papilloma at percutaneous breast biopsy? AJR Am J Roentgenol 186:1328–1334 Mercado CL, Hamele-Bena D, Oken SM, Singer CI, Ciangiarella J, 2006, Papillary lesions of the breast at percutaneus core needle biopsy. Radiology 238:801–808 Skandarajah AR, Field L, Yuen Larn Mou A, Buchanan M, Evans J, Hart S et al, 2008, Benign papilloma on core biopsy requires surgical excision. Ann Surg Oncol 15:2272–2277 Jaffer S, Nagi C, Bleiweiss IJ, 2009, Excision is indicated for intaductal papilloma of the breast diagnosed on core needle biopsy. Cancer 115:2837–2843 Jung SY, Kang HS, Kwon Y, Min SY, Kim EA, Ko KL et al, 2010, Risk factors for malignancy in benign papillomas of the breast on core needle biopsy. World J Surg 34:261–265 Chang JM,MoonWK, Cho N, HanW, Noh DY, Park IA et al, 2010, Risk of carcinoma after subsequent excision of benign papilloma initially diagnosed with an ultrasound, US)-guided 14-gauge core needle biopsy: a prospective observational study. Eur Radiol 20: 1093–1100 Chang JM, Moon WK, Cho N, Han W, Noh DY, Park IA et al, 2011, Management of ultrasonogaphically detected benign papillomas of the breast at core needle biopsy. Am J Roentgenol 196:723–729 Shouhed D, Amersi FF, Spurrier R, Dang C, Astvatsturyan K, Bose S et al, 2012, Intraductal papillary lesions of the breast: clinical and pathological correlation. Am Surg 78:1161–1165 Wen X, Cheng W, 2013, Nonmalignant breast papillary lesions at core-needle biopsy: a meta-analysis of underestimation and influencing factors. Ann Surg Oncol 20:94–101 Rajan S, Shaabab AM, Dall BJ, Sharma N, 2012, New patient pathway using vacuum-assisted biopsy reduces diagnostic surgery for B3 lesions. Clin Radiol 67:244–249 Tennant SL, Evans A, Hamilton LJ, James J, Lee AH, Hodi Z et al, 2008, Vacuum-assisted excision of breast lesions of uncertain malignant potential, B3)- an alternative to surgery in selected cases. Breast 17:546–549 Wilkinson LS, Wells C, The W, Desai A, Wilson R, 2012, The management of indeterminate breast lesions-a clinicians guide. London Breast Screening Quality Assurance Reference Centre, London NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 535 EP 546 DI 10.1007/s12253-014-9882-7 PG 12 ER PT J AU Chen, YY Li, FYA Huang, KH Lan, YT Chen, MH Chao, Y Lo, SSh Wu, ChW Shyr, YM Fang, WL AF Chen, Yin-Yin Li, Fen-Yau Anna Huang, Kuo-Hung Lan, Yuan-Tzu Chen, Ming-Huang Chao, Yee Lo, Su-Shun Wu, Chew-Wun Shyr, Yi-Ming Fang, Wen-Liang TI Adenosquamous Carcinoma of the Stomach and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adenosquamous carcinoma; Adenocarcinoma; Squamous cell carcinoma; Metastatic lymph node ID Adenosquamous carcinoma; Adenocarcinoma; Squamous cell carcinoma; Metastatic lymph node AB Adenosquamous carcinoma of the stomach is a very rare disease, consisting of less than 0.4 % of all stomach cancer. From 1991 to 2013, a total of 2800 patients received gastrectomy for gastric cancer at Taipei Veterans General hospital. Among them, seven patients (0.25 %) diagnosed as adenosquamous carcinoma were enrolled. The clinicopathologic characteristics and prognosis were analyzed. The mean age of the seven patients was 62.3 years-old. There were 5 males and 2 females. Six patients were stage III disease and one patient was stage IV disease. Four patients finally died of gastric cancer. Only one patient had no recurrence until death. Among the seven patients, adenocarcinoma component comprises the majority of the metastatic lymph node in 6 patients (85.7 %). The only one patient with major squamous cell carcinoma component in metastatic lymph node had no tumor recurrence till death. Adenosquamous carcinoma of stomach is a rare disease and is associated with a poor prognosis. The component of adenocarcinoma and squamous cell carcinoma in the metastatic lymph node may influence the prognosis. C1 [Chen, Yin-Yin] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd., Beitou District, 11217 Taipei, Taiwan, Republic of China. [Li, Fen-Yau Anna] Taipei Veterans General Hospital, Department of PathologyTaipei, Taiwan, Republic of China. [Huang, Kuo-Hung] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd., Beitou District, 11217 Taipei, Taiwan, Republic of China. [Lan, Yuan-Tzu] Taipei Veterans General Hospital, Department of Surgery, Division of Colon & Rectal SurgeryTaipei, Taiwan, Republic of China. [Chen, Ming-Huang] Taipei Veterans General Hospital, Department of Medicine, Division of Hematology and OncologyTaipei, Taiwan, Republic of China. [Chao, Yee] Taipei Veterans General Hospital, Department of Medicine, Division of Hematology and OncologyTaipei, Taiwan, Republic of China. [Lo, Su-Shun] National Yang-Ming University, School of MedicineTaipei, Taiwan, Republic of China. [Wu, Chew-Wun] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd., Beitou District, 11217 Taipei, Taiwan, Republic of China. [Shyr, Yi-Ming] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd., Beitou District, 11217 Taipei, Taiwan, Republic of China. [Fang, Wen-Liang] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd., Beitou District, 11217 Taipei, Taiwan, Republic of China. RP Fang, WL (reprint author), Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, 11217 Taipei, Taiwan, Republic of China. EM s821094@hotmail.com CR Mori E, Watanabe A, Maekawa S et al, 2000, Adenosquamous carcinoma of the remnant stomach: report of a case. Surg Today 30:643– 646 Toyota N, Minagi S, Takeuchi T et al, 1996, Adenosquamous carcinoma of the stomach associated with separate early gastric cancer, type IIc). J Gastroenterol 31:105–108 Tohma T, Yamamoto Y, Seki Yet al, 2009)Weekly paclitaxel therapy is effective for gastric adenosquamous carcinoma: a case report. Hepatogastroenterology 56:568–570 Samejima Y, Mizuno T, Sasakawa M et al, 1974, A case report of early gastric adenoacanthoma and brief review of its literature. I To Cho 9:783–788, in Japanese) Watanabe Y, Nakamura N, Iwasaki N et al, 1976, Early gastric adenoacanthoma of superficial spreading type diagnosed by gastroscopy, reported of a case. I To Cho 11:347–352, in Japanese) Hirai S, Koyama S, Ebihara T et al, 1986, Two cases of the gastric adenosquamous cell carcinoma diagnosed by gastroscopic biopsy. Shokaki Naishikyo 28:3139–3145, in Japanese) Horikawa M, Miyashiro N, Nakatani M et al, 1987, A case of early gastric adenosquamous carcinoma. Gan No Rinsho 33:305–310, in Japanese) Johzaki H, Murayama H, Maekawa T et al, 1988, A case of small squamous cell carcinoma with a grading of Ila + IIc, an early gastric cancer type. Gan No Rinsho 34:1025–1030, in Japanese) Yasuda K, Daibo M, Urabe M et al, 1991, A case of early gastric adenosquamous carcinoma. Shokaki Naishikyo No Shinpo 38:335– 338, in Japanese) Nishidoi H, IshiguroM, Kudoh H et al, 1993, A case of early gastric adenosquamous carcinoma, type lie). Nippon Rinsho Geka Igakkai Zasshi 54:430, in Japanese) Yoshida K, Manabe T, Tsunoda T et al, 1996, Early gastric cancer of adenosquamous carcinoma type: report of a case and review of literature. Jpn J Clin Oncol 26:252–257 Faria GR, Eloy C, Preto JR et al, 2010, Primary gastric adenosquamous carcinoma in a Caucasian woman: a case report. J Med Case Rep 4: 351–5 Saito S, Hosoya Y, Morishima K et al, 2012, A clinicopathological and immunohistochemical study of gastric cancer with squamous cell carcinoma components: a clinically aggressive tumor. J Dig Dis 13: 407–413 Sobin L, Gospodarowicz M, Wittekind C, eds,, 2009, TNM classification of malignant tumours, UICC International Union Against Cancer), 7th edn. New York, Wiley-Blackwell NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 547 EP 551 DI 10.1007/s12253-014-9890-7 PG 5 ER PT J AU Ben Abdelkrim, S Fathallah, K Rouatbi, R Ayachi, M Hmissa, S Mokni, M AF Ben Abdelkrim, Soumaya Fathallah, Khadija Rouatbi, Rim Ayachi, Malak Hmissa, Sihem Mokni, Moncef TI OM.Breast Cancer in Very Young Women Aged 25 Year-Old or Below in the Center of Tunisia and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Young women; Tunisia; 25 years-old ID Breast cancer; Young women; Tunisia; 25 years-old AB Breast cancer in very young women under 40 or 35 years attracted a widespread attention. Few studies have focused on women aged below 25 years. The aim of this study was to evaluate the situation of breast cancer in women ≤25 years in the center of Tunisia. Retrospective review from 1993 to 2013. Clinical, histopathological, therapeutic and outcome data were recorded. Cases were classified into different molecular subtypes based on the immunohistochemistrybased definitions. The series included 25 patients. The mean duration of symptoms was 7.5 months. The most common presenting symptom was a palpable mass. Four patients had at least one relative diagnosed with breast cancer. Mammography combined with ultrasound was suggestive of malignancy in 60 % of cases. Curative surgical treatment could be offered in 19 cases. The mean tumor size was 39 mm. Nodal metastases were detected in 9/18 cases. Twenty cases could be classified into: luminal A (5 cases), luminal B (6 cases), Her-2 (1 case), triple negative (6 cases) and unclassified (2 cases). Two women experienced locoregional recurrence and 6 had distant recurrence. Asynchronous contralateral breast cancer occurred in one case. The overall survival at 5 and 10 years was 85 and 75 % respectively. The survival was significantly lower in grade III tumors (p=0.04) and triple negative tumors (p= 0.03). Breast cancer in women ≤25 years is uncommon. An adequate medical education of young women and physicians is necessary. C1 [Ben Abdelkrim, Soumaya] Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. [Fathallah, Khadija] Farhat Hached University Hospital, Department of GynecologySousse, Tunisia. [Rouatbi, Rim] Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. [Ayachi, Malak] Cancer RegistrySousse, Tunisia. [Hmissa, Sihem] Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. [Mokni, Moncef] Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. RP Ben Abdelkrim, S (reprint author), Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. EM benabdelkrims@voila.fr CR Missaoui N, Jaidene L, Ben Abdelkrim S, Ben Abdelkader A, Beizig N, Ben Yaacoub L, Yaacoubi MT, Hmissa S, 2011, Breast cancer in Tunisia: clinical and pathological findings. Asian Pac J Cancer Prev 12(1):169–172 Missaoui N, Landolsi H, Jaidaine L, Ben Abdelkader A, Yaacoubi MT,Hmissa S, 2012, Breast cancer in central Tunisia: an earlier age at diagnosis and incidence increase over a 15-year period. Breast J 18(3):289–291 Narod SA, 2012, Breast cancer in young women. Nat Rev Clin Oncol 9:460–470 Kothari AS, Beechey-Newman N, D’Arrigo C, Hanby AM, Ryder K, Hamed H, Fentiman IS, 2002, Breast carcinoma in women age 25 years or less. Cancer 94(3):606–614 Khanfir A, Frikha M, Kallel F, Meziou M, Trabelsi K, Boudawara T, Mnif J, Daoud J, 2006, Breast cancer in young women in the south of Tunisia. Cancer Radiother 10(8):565–571 Dimitrakakis C, Tsigginou A, Zagouri F, Marinopoulos S, Sergentanis TN, Keramopoulos A, Liakou P, Zografos GC, Papadimitriou CA, Dimopoulos MA, Antsaklis A, 2013, Breast cancer in women aged 25 years and younger. Obstet Gynecol 121(6):1235–1240 Paillocher N, Lacourtoisie SA, Fondrinier E, Catala L, Morand C, Boursier J, Guerin O, Descamps P, 2006, Infiltrating breast cancer in women younger than 25 years: 13 cases. PresseMed 35(11 Pt 1): 1618–1624 Yao S, Xu B, Ma F, Liao Y, Fan Y, 2009, Breast cancer in women younger than 25: clinicopathological features and prognostic factors. Ann Oncol 20(2):387–389 Alipour S, Omranipour R, Jahanzad I, Bagheri K, 2013, Very young breast cancer in a referral center in Tehran, Iran; review of 55 cases aged 25 or less throughout 33 years. Asian Pac J Cancer Prev 14(11):6529–6532 Lakhani SR, Ellis IO, Schnitt SJ, Tan PH, van de Vijver MJ, eds,, 2012, WHO classification of tumours of the breast, fourth edition. IARC Press, Lyon Elston CW, Ellis IO, 1991, Pathological prognostic factors in breast cancer.I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19:403–410 Breast. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer StagingManual. 7th ed. New York, NY: Springer, 2010, pp 347–76 Lin CH, Liau JY, Lu YS, Huang CS, Lee WC, Kuo KT, Shen YC, Kuo SH, Lan C, Liu JM, Kuo WH, Chang KJ, Cheng AL, 2009, Molecular subtypes of breast cancer emerging in young women in Taiwan: evidence for more than just westernization as a reason for the disease in Asia. Cancer Epidemiol Biomarkers Prev 18(6): 1807–1814 Allred DC, Harvey JM, Berardo M, Clark GM, 1998, Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11(2):155–168 Khurshid A, Faridi N, Arif AM, Naqvi H, Tahir M, 2013, Breast lesions in adolescents and young women in Pakistan: a 5 year study of significance of early recognition. Asian Pac J Cancer Prev 14(6): 3465–3467 Ntekim A, Nufu FT, Campbell OB, 2009, Breast cancer in young women in Ibadan, Nigeria. Afr Health Sci 9(4):242–246 Keegan TH, DeRouen MC, Press DJ, Kurian AW, Clarke CA, 2012, Occurrence of breast cancer subtypes in adolescent and young adult women. Breast Cancer Res 14(2):R55 Gabriel CA, Domchek SM, 2010, Breast cancer in young women. Breast Cancer Res 12(5):212 Mintzer D, Glassburn J, Mason BA, Sataloff D, 2002, Breast cancer in the very young patient: a multidisciplinary case presentation. Oncologist 7(6):547–554 Chan A, Pintilie M, Vallis K, Girourd C, Goss P, 2000, Breast cancer in women < or = 35 years: review of 1002 cases from a single institution. Ann Oncol 11(10):1255–1262 Colditz GA, Willett WC, Hunter DJ, Stampfer MJ, Manson JE, Hennekens CH, Rosner BA, 1993, Family history, age, and risk of breast cancer. Prospective data from the nurses’ health study. JAMA 270(3):338–343 Assi HA, Khoury KE, Dbouk H, Khalil LE, Mouhieddine TH, El Saghir NS, 2013, Epidemiology and prognosis of breast cancer in young women. J Thorac Dis 5(S1):S2–S8 Thapa B, Singh Y, Sayami P, Shrestha UK, Sapkota R, Sayami G, 2013, Breast cancer in young women from a low risk population in Nepal. Asian Pac J Cancer Prev 14(9):5095–5099 Shannon C, Smith IE, 2003, Breast cancer in adolescents and young women. Eur J Cancer 39(18):2632–2642 ColleoniM, Rotmensz N, Robertson C, Orlando L, Viale G, Renne G, Luini A, Veronesi P, Intra M, Orecchia R, Catalano G, Galimberti V, Nole F, Martinelli G, Goldhirsch A, 2002, Very young women, <35 years, with operable breast cancer: features of disease at presentation. Ann Oncol 13(2):273–279 Tichy JR, Lim E, Anders CK, 2013, Breast cancer in adolescents and young adults: a review with a focus on biology. J Natl Compr Canc Netw 11(9):1060–1069 Bertheau P, Steinberg SM, Merino MJ, 1998, C-erbB-2, p53 and nm23 gene product expression in breast cancer in young women: immunohistochemical analysis and clinicopathologic correlation. Hum Pathol 29(4):323–329 Andre F, Domont J, Delaloge S, 2007, What can breast cancer molecular sub-classification add to conventional diagnostic tools? Ann Oncol 18(Suppl 9):ix33–6 Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D, 2000, Molecular portraits of human breast tumours. Nature 17;406(6797):747–752 Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H, Pesich R, Geisler S, Demeter J, Perou CM, Lonning PE, Brown PO, Borresen-Dale AL, Botstein D, 2003, Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A 100(14):8418–8423 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, 2011, Strategies for subtypes–dealing with the diversity of breast cancer: highlights of the St. Gallen international expert consensus on the primary therapy of early breast cancer. Ann Oncol 22(8):1736–1747 Keegan TH, Press DJ, Tao L, Derouen MC, Kurian AW, Clarke CA, Gomez SL, 2013, Impact of breast cancer subtypes on threeyear survival among adolescent and young adult women. Breast Cancer Res 15(5):R95 Ben Abdelkrim S, Trabelsi A, Missaoui N, Beizig N, Bdioui A, Anjorin A, Jomaa W, Mokni M, 2010, Distribution of molecular breast cancer subtypes among Tunisianwomen and correlation with histopathological parameters: a study of 194 patients. Pathol Res Pract 206(11):772–775 Madaras L, Szasz MA, Baranyak Z, Tokes AM, Szittya L, Lotz G, Szekely B, Szentmartoni G, Dank M, Baranyai Z, Kulka J, 2012, Morphological and immunophenotypical heterogeneity in breast cancers of young and elderly women. Magy Onkol 56(2):75–78 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 553 EP 561 DI 10.1007/s12253-015-9944-5 PG 9 ER PT J AU Zhou, X Zheng, R Zhang, H He, T AF Zhou, Xiaodong Zheng, Ruiguo Zhang, Huifang He, Tianlin TI Pathway Crosstalk Analysis of Microarray Gene Expression Profile in Human Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human hepatocellular carcinoma; Pathway crosstalk; Protein-protein interaction network ID Human hepatocellular carcinoma; Pathway crosstalk; Protein-protein interaction network AB Liver cancer is the third most common cause of cancer death in the world. Hepatocellular carcinoma (HCC) is the main pathological types in liver cancer, which amounts to 70–85 % of primary liver cancer in the world and 90 % in China. The aim of this study was to establish a PPI network and a pathway crosstalk network to isolate important dysfunctional pathways which play an important role in the pathogenesis of HCC. System biology approach was used in this research. A PPI network was firstly built and then a dysfunctional crosstalk network of HCC related pathways was constructed. Several important significant dysfunctional crosstalk pathways were identified. Basal transcription factors (hsa03022), Glycerophospholipid metabolism (hsa00564) and Metabolism of xenobiotics by cytochrome P450 (hsa00980) were significantly interact with Pathway in cancer (hsa05200). Besides, pathway Axon guidance (hsa04360) was also dysfunctional crosstalk with Pathway in cancer (hsa05200). The crosstalks among these pathways reveal some evidence that the pathways closely cooperated and play important tasks in HCC progression. Besides, the pathway hsa04360 dysfunctional crosstalk with the hsa05200 indicates there would be a same mechanism for HCC invasion and migration. C1 [Zhou, Xiaodong] PLA 535 Hospital, Department of General Surgery, 418008 Huaihua, China. [Zheng, Ruiguo] PLA 535 Hospital, Department of General Surgery, 418008 Huaihua, China. [Zhang, Huifang] PLA 535 Hospital, Department of Emergency, 418008 Huaihua, China. [He, Tianlin] Changhai Hospital, Department of General Surgery, No.168 Changhai Road, Yangpu District, 200433 Shanghai, China. RP He, T (reprint author), Changhai Hospital, Department of General Surgery, 200433 Shanghai, China. EM Skyrainhe@163.com CR Parkin DM, Bray F, Ferlay J, Pisani P, 2001, Estimating the world cancer burden: globocan 2000. Int J Cancer 94(2):153–156 Perz JF, Armstrong GL, Farrington LA, Hutin YJ, Bell BP, 2006, The contributions of hepatitis B virus and hepatitis C virus infections to cirrhosis and primary liver cancer worldwide. J Hepatol 45(4): 529–538 Tang Z-Y, Ye S-L, Liu Y-K, Qin L-X, Sun H-C, Ye Q-H, Wang L, Zhou J, Qiu S-J, Li Y, 2004, A decade’s studies on metastasis of hepatocellular carcinoma. J Cancer Res Clin Oncol 130(4):187–196 Berger JC, Vander Griend D, Stadler WM, Rinker-Schaeffer C, 2004, Metastasis suppressor genes: signal transduction, cross-talk and the potential for modulating the behavior of metastatic cells. Anti-Cancer Drugs 15(6):559–568 Berasain C, Ujue Latasa M, Urtasun R, Goni S, Elizalde M, Garcia- Irigoyen O, Azcona M, Prieto J, AvilaMA, 2011, Epidermal growth factor receptor, EGFR, crosstalks in liver cancer. Cancer 3(2):2444– 2461 Dai R, Chen R, Li H, 2009, Cross-talk between PI3K/Akt andMEK/ ERK pathways mediates endoplasmic reticulum stress-induced cell cycle progression and cell death in human hepatocellular carcinoma cells. Int J Oncol 34(6):1749–1757 Berasain C, Castillo J, Prieto J, Avila MA, 2007, New molecular targets for hepatocellular carcinoma: the ErbB1 signaling system. Liver Int 27(2):174–185 Chen L, Wang R-S, Zhang X-S, 2009, Biomolecular networks: methods and applications in systems biology, vol 10. Wiley Zhao X-M, Wang R-S, Chen L, Aihara K, 2008, Uncovering signal transduction networks from high-throughput data by integer linear programming. Nucleic Acids Res 36(9):e48–e48 Ideker T, Sharan R, 2008, Protein networks in disease. Genome Res 18(4):644–652 Wang T, Gu J, Yuan J, Tao R, Li Y, Li S, 2013, Inferring pathway crosstalk networks using gene set co-expression signatures. Mol BioSyst Stefanska B, Huang J, Bhattacharyya B, Suderman M, Hallett M, Han Z-G, Szyf M, 2011, Definition of the landscape of promoter DNA hypomethylation in liver cancer. Cancer Res 71(17):5891– 5903 R TRDC, 2008, A language and environment for statistical computing. R foundation for statistical computing Irizarry RA, Hobbs B, Collin F, Beazer‐Barclay YD, Antonellis KJ, Scherf U, Speed TP, 2003, Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4(2):249–264 Smyth GK, 2004, Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 3(1):3 BenjaminiY, HochbergY, 1995, Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society Series B, Methodological):289–300 Kanehisa M The KEGG database. In: Novartis Found Symp, 2002. pp 91–101 Zanzoni A, Montecchi-Palazzi L, Quondam M, Ausiello G, Helmer- Citterich M, Cesareni G, 2002, MINT: a Molecular INTeraction database. FEBS Lett 513(1):135–140 Mishra GR, SureshM, Kumaran K, Kannabiran N, Suresh S, Bala P, Shivakumar K, Anuradha N, Reddy R, Raghavan TM(2006, Human protein reference database—2006 update. Nucleic Acids Res 34(suppl 1):D411–D414 Stark C, Breitkreutz B-J, Chatr-Aryamontri A, Boucher L, Oughtred R, Livstone MS, Nixon J, Van Auken K,Wang X, Shi X, 2011, The BioGRID interaction database: 2011 update. Nucleic Acids Res 39(suppl 1):D698–D704 Birnbaum A, 1954, Combining independent tests of significance*. J Am Stat Assoc 49(267):559–574 Liu Z-P, Wang Y, Zhang X-S, Chen L, 2010, Identifying dysfunctional crosstalk of pathways in various regions ofAlzheimer’s disease brains. BMC Syst Biol 4(Suppl 2):S11 Panani AD, Roussos C, 2006, Cytogenetic and molecular aspects of lung cancer. Cancer Lett 239(1):1–9 Breuer R, Postmus P, Smit E, 2005, Molecular pathology of nonsmall- cell lung cancer. Respiration 72(3):313–330 Lowy D, Willumsen B, 1993, Function and regulation of ras. Annu Rev Biochem 62(1):851–891 Giatromanolaki A, Koukourakis M, O’Byrne K, Kaklamanis L, Dicoglou C, Trichia E, Whitehouse R, Harris A, Gatter K, 1996, Non-small cell lung cancer: c-erbB-2 overexpression correlates with low angiogenesis and poor prognosis. Anticancer Res 16(6B):3819 Dobbelstein M, Wienzek S, Koenig C, Roth J, 1999, Inactivation of the p53-homologue p73 by the mdm2-oncoprotein. Oncogene 18(12):2101 Ortega S, Malumbres M, Barbacid M, 2002, Cyclin D-dependent kinases, INK4 inhibitors and cancer. Biochim Biophys Acta, BBA)- Rev Cancer 1602(1):73–87 Roy B, Taneja R, Chambon P, 1995, Synergistic activation of retinoic acid, RA)-responsive genes and induction of embryonal carcinoma cell differentiation by an RA receptor alpha, RAR alpha)-, RAR beta-, or RAR gamma-selective ligand in combination with a retinoid X receptor-specific ligand.Mol Cell Biol 15(12):6481–6487 Agathanggelou A, Honorio S, Macartney DP, Martinez A, Dallol A, Rader J, Fullwood P, Chauhan A, Walker R, Shaw JA, 2001, Methylation associated inactivation of RASSF1A from region 3p21. 3 in lung, breast and ovarian tumours. Oncogene 20(12):1509 Darnell JE, 2002, Transcription factors as targets for cancer therapy. Nat Rev Cancer 2(10):740–749 Safe S, Abdelrahim M, 2005, Sp transcription factor family and its role in cancer. Eur J Cancer 41(16):2438–2448 Gordon S, Akopyan G, Garban H, Bonavida B, 2006, Transcription factor YY1: structure, function, and therapeutic implications in cancer biology. Oncogene 25(8):1125–1142., DOI 10.1038/sj.onc. 1209080 Scott KE, Wheeler FB, Davis AL, Thomas MJ, Ntambi JM, Seals DF, Kridel SJ, 2012, Metabolic regulation of invadopodia and invasion by acetyl-CoA carboxylase 1 and de novo lipogenesis. PLoS One 7(1):e29761 Pyragius CE, Fuller M, Ricciardelli C, Oehler MK, 2013, Aberrant lipid metabolism: an emerging diagnostic and therapeutic target in ovarian cancer. Int J Mol Sci 14(4):7742–7756 Menendez JA, Lupu R, 2007, Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat Rev Cancer 7(10):763–777 Rodriguez-Antona C, Ingelman-Sundberg M, 2006, Cytochrome P450 pharmacogenetics and cancer. Oncogene 25(11):1679–1691 Agundez JA, 2004, Cytochrome P450 gene polymorphism and cancer. Curr Drug Metab 5(3):211–224 Bailey LR, Roodi N, Dupont WD, Parl FF, 1998, Association of cytochrome P450 1B1, CYP1B1, polymorphism with steroid receptor status in breast cancer. Cancer Res 58(22):5038–5041 Ishibe N, Hankinson SE, Colditz GA, Spiegelman D, Willett WC, Speizer FE, Kelsey KT, Hunter DJ, 1998, Cigarette smoking, cytochrome P450 1A1 polymorphisms, and breast cancer risk in the Nurses’ Health Study. Cancer Res 58(4):667–671 Sadanandam A, Varney ML, Singh S, Ashour AE, Moniaux N, Deb S, Lele SM, Batra SK, Singh RK, 2010, High gene expression of semaphorin 5A in pancreatic cancer is associated with tumor growth, invasion and metastasis. Int J Cancer 127(6):1373–1383 Pan G, Zhang X, Ren J, Lu J, Li W, Fu H, Zhang S, Li J, 2013, Semaphorin 5A, an axon guidance molecule, enhances the invasion and metastasis of human gastric cancer through activation ofMMP9. Pathol Oncol Res 19(1):11–18., DOI 10.1007/s12253-012-9550-8 Pan G, Zhu Z, Huang J, Yang C, Yang Y, Wang Y, Tuo X, Su G, Zhang X, Yang Z, 2013, Semaphorin 5A Promotes Gastric Cancer Invasion/Metastasis via Urokinase-Type Plasminogen Activator/ Phosphoinositide 3-Kinase/Protein Kinase B. Digestive diseases and sciences: 1–8 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 563 EP 569 DI 10.1007/s12253-014-9855-x PG 7 ER PT J AU Jederan, Lovey, J Szentirmai, Z Hitre, E Lerant, G Horvath, K Godeny, M AF Jederan, Eva Lovey, Jozsef Szentirmai, Zoltan Hitre, Erika Lerant, Gergely Horvath, Katalin Godeny, Maria TI The Role of MRI in the Assessment of the Local Status of Anal Carcinomas and in Their Management SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MRI; Anal cancer; Staging of anal cancer; Chemoradiation therapy ID MRI; Anal cancer; Staging of anal cancer; Chemoradiation therapy AB This study aims to define the role of Magnetic Resonance (MR) examinations in the assessment and therapy of anal cancer (AC), and to present the main features of the MR examinations and the typical tumor spread pattern. The MR examinations of 67 anal cancer patients with histologically confirmed planocellular cancer were analyzed retrospectively. The tumor size and the signal intensity, the nodal status were examined before and after the treatment, and in recidive tumors (N=13). At the time of the diagnosis the primary tumor was in early stage (Tis, T1, T2) in 71.5%of the cases, and it was localized in 97 %. In 97.4 % of the cases the tumor had relatively increased signal intensities compared to the adjacent muscles. Patients received chemo-radiotherapy (CRT). After CRT in 26 out of 39 patients (66.7%) the size of the tumor decreased (in 75 %), and the signal intensity decreased on the T2 weighted (T2w) images. In the residual tumor cases (19/39) verified 6 patients out of 19 had further decrease in size, and signal intensity a year after the end of the therapy. The MR examination plays a key role in the therapy of AC, by assessing the precise local status, the possible recidive tumors, and monitoring the therapy. C1 [Jederan, Eva] National Institute of Oncology, Center of RadiologyBudapest, Hungary. [Lovey, Jozsef] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Szentirmai, Zoltan] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Hitre, Erika] National Institute of Oncology, Department of Chemotherapy and ClinicopharmacologyBudapest, Hungary. [Lerant, Gergely] National Institute of Oncology, Center of RadiologyBudapest, Hungary. [Horvath, Katalin] National Institute of Oncology, Center of RadiologyBudapest, Hungary. [Godeny, Maria] National Institute of Oncology, Center of RadiologyBudapest, Hungary. RP Jederan, (reprint author), National Institute of Oncology, Center of Radiology, Budapest, Hungary. EM jederan@t-online.hu;jederan@oncol.hu NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 571 EP 579 DI 10.1007/s12253-014-9857-8 PG 9 ER PT J AU Wu, H Gu, Yh Wei, L Guo, Tk Zhao, Y Su, G Li, J Xie, Xd AF Wu, Hua Gu, Yuan-hui Wei, Li Guo, Tian-kang Zhao, Yong Su, Gang Li, Jiong Xie, Xiao-dong TI Association of Romo1 Gene Genetic Polymorphisms with Risk of Gastric Cancer in Northwestern Chinese Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Romo1 gene; Single nucleotide polymorphisms; DHPLC ID Gastric cancer; Romo1 gene; Single nucleotide polymorphisms; DHPLC AB Increased expression of reactive oxygen species modulator 1 protein-triggered reactive oxygen species production was reported in the mitochondria of various cancer cell lines. To date there is no report on association between Romo1 gene polymorphisms and gastric cancer risk. To investigate the relationship between Romo1 gene polymorphisms and GC risk, we conducted a case-control study in a population from northwest China (358 GC patients and 412 healthy controls). The genotypes of two SNPs were determined with PCR– denaturing high-performance liquid chromatography and direct DNA sequencing. We found that the genotype and allele distributions of two polymorphisms were significantly different in GC patients compared with controls, When the wild type of two loci were served as the reference group, respectively, significantly increased risk for gastric cancer were associated with rs6060566 TC genotype (Adjusted OR= 1.525, 95 % CI =1.126–2.138), rs6060567 GC genotype (Adjusted OR=1.641, 95 % CI =1.238–2.291) and CC genotype (Adjusted OR=1.594, 95%CI =1.102–2.973). This effect was more pronounced in patients with smoking, alcohol consumption, H.pylori infection,and male patients subgroups. Haplotypes analysis of two genetic variants showed that the most common haplotype TG displayed the strongest evidence of association with GC (corrected P=9.30×10−5), and was associated with protection against GC (OR=0.584). Whereas the CC haplotypes had significant correlation with GC risk (OR=1.732). These findings suggested genetic polymorphisms of Romo1 gene were associated with significant risk of GC in Northwestern Chinese population, which is strengthened by alcohol use, smoking, H.pylori infection or male patients. C1 [Wu, Hua] Lanzhou University, School of Basic Medical Science, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 730000 Lanzhou, Gansu Province, China. [Gu, Yuan-hui] People’s Hospital of Gansu Province, Surgical Department, 730000 Lanzhou, Gansu Province, China. [Wei, Li] People’s Hospital of Gansu Province, Surgical Department, 730000 Lanzhou, Gansu Province, China. [Guo, Tian-kang] People’s Hospital of Gansu Province, Surgical Department, 730000 Lanzhou, Gansu Province, China. [Zhao, Yong] General Hospital of Lanzhou Military Region, 730050 Lanzhou, Gansu Province, China. [Su, Gang] Lanzhou University, School of Basic Medical Science, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 730000 Lanzhou, Gansu Province, China. [Li, Jiong] Lanzhou University, School of Basic Medical Science, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 730000 Lanzhou, Gansu Province, China. [Xie, Xiao-dong] Lanzhou University, School of Basic Medical Science, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 730000 Lanzhou, Gansu Province, China. RP Xie, Xd (reprint author), Lanzhou University, School of Basic Medical Science, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 730000 Lanzhou, China. EM xdxie@lzu.edu.cn;xdxiegenetics@outlook.com CR Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics,, 2002, CA Cancer J Clin 55:74–108 Sun XD, Mu R, Zhou YS, Dai XD, Zhang SW, Lu FZ, Qiao YL et al, 2004, Analysis of mortality rate of stomach caner and its trend in twenty years in China. Zhonghua Zhong Liu Za Zhi 26(1):4–9, Article in Chinese) Ferlay J, Bray F, Pisani P, Parkin DM, 2004, GLOBOCAN 2002 cancer incidence, mortality and prevalence worldwide. IARC Cancer Base Press No. 5, version 2.0., Lyon, France Barber M, Fitzgerald RC, Caldas C, 2006, Familial gastric canceraetiology and pathogenesis. Best Pract Res Clin Gastroenterol 207: 21–34 Brenner C, Kroemer G, 2000, Apoptosis, mitochondria, the death signal integrators. Science 289:1150–1151 Modica-Napolitano JS, Kulaviec M, Singh KK, 2007, Mitochondria and human cancer. Curr Mol Med 7:121–131 Martindale JL, Holbrook NJ, 2002, Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol 192:1–15 Toyokuni S, Okamoto K, Yodoi J, Hiai H, 1995, Persistent oxidative stress in cancer. FEBS Lett 358:1–3 Chung YM, Yoo YD et al, 2006, A novel protein, Romo1, induces ROS production in the mitochondria. Biochem Biophys Res Commun 347:649–655 Na AR, Chung YM, Lee SB, Park SH, Lee M-S, Yoo YD, 2008, A critical role for Romo1-derived ROS in cell proliferation. Biochem Biophys Res Commun 369(2):672–678 Chung JS, Lee SB, Park SH, Yoo YD et al, 2009, Mitochondrial reactive oxygen species originating from Romo1 exert an important role in normal cell cycle progression by regulating p27Kip1 expression. Free Radic Res 43:729–737 Deloukas P,Matthews LH, Ashurst J et al, 2001, The DNA sequence and comparative analysis of human chromosome 20. Nature 414: 865–871 Giordano C, Sebastiani M, d’Amati G, Carelli V et al, 2008, Gastrointestinal dysmotility in mitochondrial neurogastrointestinal encepha-lomyopathy is caused by mitochondrial DNA depletion. Am J Pathol 173:1121–1128 Solaini G, Harris DA, Lenaz G, Sgarbi G, Baracca A, 2008, The study of the pathogenic mechanism of mitochondrial diseases provides information on basic bioenergetics. Biochim Biophys Acta 1777(7–8):941–945 Montoya J, Lopez-Gallardo E, Diez-Sanchez C, Lopez-Perez MJ, 2009, Twenty years of human mtDNA pathologic point mutations: carefully reading the pathogenicity criteria. Biochim Biophys Acta 1787(5):476–483 Broder AC, 1925, The grading of carcinoma. Minn Med 8:726–730 Shi YY, He L, 2005, SHEsis, a powerful software platform for analyses of linkage disequilibrium, haplotype construction, and genetic association at polymorphism loci. Cell Res 15:97–98 Hwang IT, Chung YM, Kim JJ, Chung JS, Kim BS, Kim HJ, Kim JS, Yoo YD, 2007, Drug resistance to 5-FU linked to reactive oxygen species modulator 1. Biochem Biophys Res Commun 359:304–310 Chung YM, Lee SB, Kim HJ, Park SH, Kim JJ, Chung JS, Yoo YD, 2008, Replicative senescence induced by Romo1-derived reactive oxygen species. J Biol Chem 283:33763–33771 Lee SB, Kim JJ, Kim TW, Kim BS, LeeM-S, Yoo YD, 2010, Serum deprivation-induced reactive oxygen species production is mediated by Romo1. Apoptosis 15(2):204–218 Chung JS, Park S, Park SH, Park ER, Cha PH, Kim BY, Chung YM, Woo SR, Han CJ, Kim SB, Suh KS, Jang JJ, Lee K, Choi DW, Lee S, Lee GY, Hahm KB, Shin JA, Kim BS, Noh KH, Kim TW, Lee KH, Yoo YD, 2012, Overexpression of romo1 promotes production of reactive oxygen species and invasiveness of hepatic tumor cells. Gastroenterology 143(4):1084–1094 Zhao J, Liu T, Jin S-B, Tomilin N, Castro J, Shupliakov O, Lendahl U, Nister M, 2009, The novel conserved mitochondrial innermembrane protein MTGM regulates mitochondrial morphology and cell proliferation. J Cell Sci 122:2252–2262 KosakiK,Udaka T, Okuyama T, 2005)DHPLC in clinicalmolecular diagnostic services. Mol Genet Metab 86(1–2):117–123 Guo L, Li L, Wang W, Pan Z, Zhou Q, Wu Z, 2012, Mitochondrial reactive oxygen species mediates nicotine-induced hypoxia-inducible factor-1α expression in human non-small cell lung cancer cells. Biochim Biophys Acta 1822(6):852–861 Liao D, Corle C, Seagroves TN, Johnson RS, 2007, Hypoxiainducible factor-1alpha is a key regulator of metastasis in a transgenic model of cancer initiation and progression. Cancer Res 67(2):563–572 Ding WQ, Fried U, Larsson C, Alling C, 1998, Ethanol exposure potentiates fosB and junB expression induced bymuscarinic receptor stimulation in neuroblastoma SH-SY5Y cells. Alcohol Clin Exp Res 22:225–230 Huang B, Liu B, Yang L, Li Y, Cheng M et al, 2012, Functional genetic variants of c-Jun and their interaction with smoking and drinking increase the susceptibility to lung cancer in so in southern and eastern Chinese. Int J Cancer 131:E744–E758 IARC-International Agency for Research on Cancer, 1994, Infection with helicobacter pylori. IARC Monogr Eval Carcinog Risks Hum 61:177–240 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 581 EP 587 DI 10.1007/s12253-014-9858-7 PG 7 ER PT J AU Baric, M Kulic, A Sirotkovic-Skerlev, M Dedic Plavetic, N Vidovic, M Horvatic-Herceg, G Vrbanec, D AF Baric, Marina Kulic, Ana Sirotkovic-Skerlev, Maja Dedic Plavetic, Natalija Vidovic, Marina Horvatic-Herceg, Gordana Vrbanec, Damir TI Circulating Her-2/Neu Extracellular Domain in Breast Cancer Patients-Correlation with Prognosis and Clinicopathological Parameters Including Steroid Receptor, Her-2/Neu Receptor Coexpression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HER-2/neu ECD; Breast cancer; Prognosis; Steroid receptor/HER-2/neu receptor coexpression ID HER-2/neu ECD; Breast cancer; Prognosis; Steroid receptor/HER-2/neu receptor coexpression AB HER-2/neu extracellular domain (ECD) can be detected in blood as a soluble circulating protein. The aim of this study was to analyze the relationship between HER-2/neu extracellular domain in the serum and the prognosis in breast cancer patients. We also correlated HER-2/neu ECD with various clinicopathological factors including steroid receptor, HER-2/neu receptor coexpression. The serum from seventy nine patients with invasive breast cancer and twenty individuals without malignancy was analyzed using the enzymelinked immune adsorbent assay method. The cut-off value was estimated by the ROC curve analysis (15.86 μg/L). HER-2/neu ECD values in the serum of patients with breast cancer were significantly higher than in control subjects. Circulating HER-2/neu ECD was significantly associated with the histological grade of tumors and the status of axillary lymph nodes. Negative correlation was observed between HER-2/neu ECD in the serumand estrogen receptor positivity. When we analyzed HER-2/neu ECD in relation with coexpression of steroid receptor and HER-2/neu receptor in tissue, statistically higher values were found in the subgroup of patients with steroid receptor negative, HER-2/neu negative tumors than in the other subgroups. HER-2/neu ECD was not an independent factor in the univariate and multivariate analysis. However, elevated HER-2/neu ECD levels were found in patients with breast cancer possessing more aggressive phenotype. C1 [Baric, Marina] University Hospital Center Zagreb and Zagreb Medical School, Department of Medical Oncology, Kispaticeva 12, 10 000 Zagreb, Croatia. [Kulic, Ana] University Hospital Center Zagreb and Zagreb Medical School, Department of Medical OncologyZagreb, Croatia. [Sirotkovic-Skerlev, Maja] University Hospital Center Zagreb and Zagreb Medical School, Department of Medical OncologyZagreb, Croatia. [Dedic Plavetic, Natalija] University Hospital Center Zagreb and Zagreb Medical School, Department of Medical Oncology, Kispaticeva 12, 10 000 Zagreb, Croatia. [Vidovic, Marina] University Hospital Center Zagreb and Zagreb Medical School, Department of Medical Oncology, Kispaticeva 12, 10 000 Zagreb, Croatia. [Horvatic-Herceg, Gordana] University Hospital Center Zagreb, Clinical Department of Nuclear Medicine and Radiation ProtectionZagreb, Croatia. [Vrbanec, Damir] University Hospital Center Zagreb and Zagreb Medical School, Department of Medical Oncology, Kispaticeva 12, 10 000 Zagreb, Croatia. RP Baric, M (reprint author), University Hospital Center Zagreb and Zagreb Medical School, Department of Medical Oncology, 10 000 Zagreb, Croatia. EM marina.baric11@gmail.com CR Slamon DJ, ClarkGM,Wong SG, LevinWJ, Ullrich A,McguireWL, 1987, Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235:177–182 Ross JS, Slodkowska EA, Symmans WF, Pusztai L, Ravdin PM, Hortobagyi GN, 2009, The HER-2 receptor and breast cancer: 10 years of targeted anti-HER-2 therapy and personalized medicine. Oncologist 4:320–368 Suzuki M, Shigematsu H, Hiroshima K, Iizasa T, Nakatani Y, Minna JD et al, 2005, Epidermal growth factor receptor expression status in lung cancer correlates with its mutation. Hum Pathol 36:1127–1134 Zeng S, Yang Y, Tan Y, Lu C, Pan Y, Chen L et al, 2012, ERBB2- induced inflammation in lung carcinogenesis. Mol Biol Rep 39: 7911–7917 Minner S, Jessen B, Stiedenroth L, Burandt E, Kollermann J, Mirlacher M et al, 2010, Low level HER2 overexpression is associated with rapid tumor cell proliferation and poor prognosis in prostate cancer. Clin Cancer Res 16:1553–1560 Bookman MA, Darcy KM, Clarke-Pearson D, Boothby RA, Horowitz IR, 2003, Evaluation of monoclonal humanized anti- HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the gynecologic oncology group. J Clin Oncol 21:283–290 Giuffre G, Ieni A, Barresi V, Caruso RA, Tuccari G, 2012, HER2 status in unusual histological variants of gastric adenocarcinomas. J Clin Pathol 65:237–241 Tse C, Gauchez AS, Jacot W, Lamy PJ, 2012, HER2 shedding and serum HER2 extracellular domain: biology and clinical utility in breast cancer. Cancer Treat Rev 38:133–142 Sasso M, Bianchi F, Ciravolo V, Tagliabue E, Campiglio M, 2011, HER2 splice variants and their relevance in breast cancer. J Nucleic Acids Investig 2:e9 Horwitz KB, Mcguire WL, 1977, Progesterone and progesterone receptors in experimental breast cancer. Cancer Res 37:1733–1738 Bloom HJ, Richardson WW, 1957, Histological grading and prognosis in breast cancer; a study of 1409 cases of which 359 have been followed for 15 years. Br J Cancer 11(3):359–377 CarneyWP, Neumann R, Lipton A, Leitzel K, Ali S, Price CP, 2003, Potential clinical utility of serum HER-2/neu oncoprotein concentrations in patients with breast cancer. Clin Chem 49:1579–1598 DresseMMD,Heinemann V, Kahlert S, Bauerfeind I,Nagel D, et al.., 2009, HER-2/neu in tissue and serum at time of primary breast cancer. J Clin Oncol Abstract #11500. Leary AF, HannaWM, van de Vijver MJ, Penault-Llorca F, Ruschoff J, Osamura RYet al, 2009, Value and limitations of measuring HER- 2 extracellular domain in the serum of breast cancer patients. J Clin Oncol 27:1694–1705 Carney WP, Bernhardt D, Jasani B, 2013, Circulating HER2 extracellular domain: a specific and quantitative biomarker of prognostic value in all breast cancer patients? Biomark Cancer 5:31–39 Thriveni K, Deshmane V, Bapsy PP, Krishnamoorthy L, Ramaswamy G, 2007, Clinical utility of serum human epidermal receptor-2/neu detection in breast cancer patients. Indian J Med Res 125:137–142 Garoufali A, Kyriakou F, Kountourakis P, Yioti I, Malliou S, Nikaki A et al, 2008, Extracellular domain of HER2: a useful marker for the initial workup and follow-up of HER2-positive breast cancer. J BUON 13:409–413 Pallud C, Guinebretiere JM, Guepratte S, Hacene K, Neumann R, Carney W et al, 2005, Tissue expression and serum levels of the oncoprotein HER-2/neu in 157 primary breast tumours. Anticancer Res 25:1433–1440 Saghatchian M, Guepratte S, Hacene K, Neumann R, Floiras JL, Pichon MF, 2004, Serum HER-2 extracellular domain: relationship with clinicobiological presentation and prognostic value before and after primary treatment in 701 breast cancer patients. Int J Biol Markers 19:14–22 Kong Y, Dai S, Xie X, Xiao X, Lv N, Guo J et al, 2012, High serum HER2 extracellular domain levels: correlation with a worse diseasefree survival and overall survival in primary operable breast cancer patients. J Cancer Res Clin Oncol 138:275–284 Molina MA, Codony-Servat J, Albanell J, Rojo F, Arribas J, Baselga J, 2001, Trastuzumab, herceptin), a humanized anti- Her2 receptor monoclonal antibody, inhibits basal and activated Her2 ectodomain cleavage in breast cancer cells. Cancer Res 61: 4744–4749 Codony-Servat J, Albanell J, Lopez-Talavera J, Arribas J, Baselga J, 1999, Cleavage of the HER2 ectodomain is a pervanadate-activable process that is inhibited by the tissue inhibitor of metalloproteases-1 in breast cancer cells. Cancer Res 59:1196–1201 Quaranta M, Daniele A, Coviello M, Savonarola A, Abbate I, Venneri MT et al, 2006, c-erbB-2 protein level in tissue and sera of breast cancer patients: a possibly useful clinical correlation. Tumori 92:311–317 Ludovini V, Gori S, Colozza M, Pistola L, Rulli E, Floriani I et al, 2008, Evaluation of serum HER2 extracellular domain in early breast cancer patients: correlation with clinicopathological parameters and survival. Ann Oncol 19:883–890 Ma L, Yang HY, Han XH, Li J, Wang F, Zhang CL et al, 2012, Relationship between serum HER2 extracellular domain levels, tissue HER2 expression, and clinico-pathological parameters in early stage breast cancer. Chin Med J 125:4104–4110 Molina R, Auge JM, Escudero JM, Filella X, Zanon G, Pahisa J et al, 2010, Evaluation of tumor markers, HER-2/neuoncoprotein, CEA, and CA 15.3, in patients with locoregional breast cancer: prognostic value. Tumour Biol 31:171–180 Duru N, Candas D, Jiang G, Li JJ, 2014, Breast cancer adaptive resistance: HER2 and cancer stem cell repopulation in a heterogeneous tumor society. J Cancer Res Clin Oncol 140(1):1–14 Lee CY, Lin Y, Bratman SV, Feng W, Kuo AH, Scheeren FA et al, 2014, Neuregulin autocrine signaling promotes self-renewal of breast tumor-initiating cells by triggering HER2/HER3 activation. Cancer Res 74(1):341–352 Samy N, Ragab HM, ElMaksoud NA, ShaalanM(2010, Prognostic significance of serum Her2/neu, BCL2, CA15-3 and CEA in breast cancer patients: a short follow-up. Cancer Biomark 6:63–72 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 589 EP 595 DI 10.1007/s12253-014-9859-6 PG 7 ER PT J AU Nemes, K Csoka, M Nagy, N Mark, Varadi, Zs Danko, T Kovacs, G Kopper, L Sebestyen, A AF Nemes, Karolina Csoka, Monika Nagy, Noemi Mark, Agnes Varadi, Zsofia Danko, Titanilla Kovacs, Gabor Kopper, Laszlo Sebestyen, Anna TI Expression of Certain Leukemia/Lymphoma Related microRNAs and its Correlation with Prognosis in Childhood Acute Lymphoblastic Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE miRNA-128b miRNA-223; Childhood ALL; Prognosis; Relapse ID miRNA-128b miRNA-223; Childhood ALL; Prognosis; Relapse AB In spite of the improved efficacy of therapy, it still fails in 15–20 % of childhood acute lymphoblastic leukemia (ALL) patients. Recently, altered expression of certain miRNAs (miRs) have been described in ALL with potential effect on prognosis. Presence of certain miRs (miRNA-16, −21, −24, −29b, −128b, −142-3p, −155, −223) was characterized in human lymphoma and leukemia cells by real-time PCR. Expression of miRs in pediatric ALL patients (n=24) was measured before chemotherapy, at conventional response checkpoints and at relapse. Correlation between altered miR expression and response to prednisolone at day 8 of therapy and long term prognosis was statistically analysed. Overexpression of "oncomiR/inflammamiR"-21 – which is characteristic in different tumors—was missing in human ALL cells. However, higher expression of miR-128b and lower expression of miR-223 is generally characteristic for human ALL cell lines and ALL cells isolated from pediatric patients. Correlation was shown between miR-128b expression and prognosis, prednisolone response and survival data in childhood ALL. Expression of miR-128b and miR-223—both are leukemia specific—changed in parallel with percentage of bone marrow blasts in remission and during relapse. Therefore, we suggest that overexpression of miR-128b and downregulation of miR-223 shows a significant correlation with treatment response and prognosis in childhood ALL. C1 [Nemes, Karolina] Semmelweis University, 2nd Department of Pediatrics, Tuzolto u. 7-9., 1094 Budapest, Hungary. [Csoka, Monika] Semmelweis University, 2nd Department of Pediatrics, Tuzolto u. 7-9., 1094 Budapest, Hungary. [Nagy, Noemi] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., 1085 Budapest, Hungary. [Mark, Agnes] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., 1085 Budapest, Hungary. [Varadi, Zsofia] Semmelweis University, 2nd Department of Pediatrics, Tuzolto u. 7-9., 1094 Budapest, Hungary. [Danko, Titanilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., 1085 Budapest, Hungary. [Kovacs, Gabor] Semmelweis University, 2nd Department of Pediatrics, Tuzolto u. 7-9., 1094 Budapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., 1085 Budapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26., 1085 Budapest, Hungary. RP Csoka, M (reprint author), Semmelweis University, 2nd Department of Pediatrics, 1094 Budapest, Hungary. EM dr.csoka.monika@gmail.com CR Kaatsch P, 2010, Epidemiology of childhood cancer. Cancer Treat Rev 36:277–285 Pui CH, Evans WE, 2006, Treatment of acute lymphoblastic leukemia. N Engl J Med 354:166–178 Baranwal S, Alahari SK, 2010, miRNA control of tumor cell invasion and metastasis. Int J Cancer 126:1283–1290 Shin VY, Chu KM, 2014, MiRNA as potential biomarkers and therapeutic targets for gastric cancer. World J Gastroenterol 20: 10432–10439 Zhu J, Zheng Z, Wang J, Sun J, Wang P, Cheng X, Fu L, Zhang L, Wang Z, Li Z, 2014, Different miRNA expression profiles between human breast cancer tumors and serum. Front Genet 5:149 Vasilatou D, Papageorgiou S, Pappa V, Papageorgiou E, Dervenoulas J, 2010, The role of microRNAs in normal and malignant hematopoiesis. Eur J Haematol 84:1–16 Lawrie CH, 2013, MicroRNAs in hematological malignancies. Blood Rev 27:143–154 Wang Y, Lee CG, 2009)MicroRNA and cancer-focus on apoptosis. J Cell Mol Med 13:12–23 Dong C, Ji M, Ji C, 2009, MicroRNAs and their potential target genes in leukemia pathogenesis. Cancer Biol Ther 8:200–205 So AY, Zhao JL, Baltimore D, 2013, The Yin and Yang of microRNAs: leukemia and immunity. Immunol Rev 253:129–145 Schotte D, Akbari Moqadam F, Lange-Turenhout EA, Chen C, van Ijcken WF, Pieters R, den Boer ML, 2011, Discovery of new microRNAs by small RNAome deep sequencing in childhood acute lymphoblastic leukemia. Leukemia 25:1389–1399 Rainer J, Ploner C, Jesacher S, Ploner A, Eduardoff M, Mansha M, WasimM, Panzer-Grumayer R, Trajanoski Z, Niederegger H, Kofler R, 2009, Glucocorticoid-regulated microRNAs and mirtrons in acute lymphoblastic leukemia. Leukemia 23:746–752 Mi S, Lu J, SunM, Li Z, Zhang H, NeillyMB,Wang Y, Qian Z, Jin J, Zhang Y, Bohlander SK, Le Beau MM, Larson RA, Golub TR, Rowley JD, Chen J, 2007, MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia. Proc Natl Acad Sci U S A 104:19971–19976 de Oliveira JC, Scrideli CA, Brassesco MS, Morales AG, Pezuk JA, QueirozRdeP YJA, Brandalise SR, Tone LG, 2012, Differential miRNA expression in childhood acute lymphoblastic leukemia and association with clinical and biological features. Leuk Res 36:293– 298 WangY, Gao X,Wei F, ZhangX, Yu J, Zhao H, Sun Q, Yan F, Yan C, Li H, Ren X, 2014, Diagnostic and prognostic value of circulating miR-21 for cancer: a systematic review and meta-analysis. Gene 533: 389–397 Mirnezami AH, Pickard K, Zhamg L, Primrose JN, Packham G, 2009, MicroRNAs: key players in carcinogenesis and novel therapeutic targets. Eur J Surg Oncol 35:339–347 Loffler D, Brocke-Heinrich K, Pfeifer G, Stocsits C, Hackemuller J, Kretzschmar AK, Burger R, Gramatzki M, Blumert C, Bauer K, Cvijic H, Ullmann AK, Stadler PF, Horn F, 2007, Interleukin-6 dependent survival of multiple myeloma cells involves the Stat3- mediated induction of microRNA-21 through a highly conserved enhancer. Blood 110:1330–1333 Jurkovicova D, Magyerkova M, Kulcsar L, Krivjanska M, Krivjansky V, Gibadulinova A, Oveckova I, Chovanec M, 2014, miR-155 as a diagnostic and prognostic marker in hematological and solid malignancies. Neoplasma 61:241–251 Seddiki N, Brezar V, Ruffin N, Levy Y, Swaminathan S, 2014, Role of miR-155 in the regulation of lymphocyte immune function and disease. Immunology 142:32–38 Huang X, Shen Y, Liu M, Bi C, Jiang C, Igbal J, McKeithan TW, ChanWC, Ding SJ, Fu K, 2012, Quantitative proteomics reveals that miR-155 regulates the PI3K-AKT pathway in diffuse large B-cell lymphoma. Am J Pathol 181:26–33 Pedersen IM, Otero D, Kao E, Miletic AV, Hother C, Ralfkiaer E, Rickert RC, Gronbaek K, David M, 2009, Onco-miR-155 targets SHIP1 to promote TNFalpha-dependent growth of B cell lymphomas. EMBO Mol Med 1:288–295 Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M, Taccioli C, Zanesi N, Garzon R, Aqeilan RI, Alder RI, Alder H, Volinia S, Rassenti L, Liu X, Liu CG, Kipps TJ, Negrini M, Croce CM, 2008, MiR-15a and miR-16-1 cluster functions in human leukemia. Proc Natl Acad Sci U S A 105:5166–5171 Zaidi SK, Dowdy CR, van Wijnen AJ, Lian JB, Raza A, Stein JL, Croce CM, Stein GS, 2009, Altered Runx1 subnuclear targeting enhances myeloid cell proliferation and blocks differentiation by activating a miR-24/MKP-7/MAPK network. Cancer Res 69:8249– 8255 Garzon R, Heaphy CE, Havelange V, Fabbri M, Volinia S, Tsao T, Zanesi N, Kornblau SM, Marcucci G, Calin GA, Andreeff M, Croce CM, 2009, MicroRNA 29b functions in acute myeloid leukemia. Blood 114:5331–5341 Zhang YK,Wang H, Leng Y, Li ZL, Yang YF, Xiao FJ, Li QF, Chen XQ, Wang LS, 2011, Overexpression of microRNA-29b induces apoptosis of multiple myeloma cells through down regulating Mcl- 1. Biochem Biophys Res Commun 414:233–239 LiY,WangH, TaoK, Xiao Q, Huang Z, Zhong L, CaoW,Wen J, Fen W, 2013, miR-29b suppresses CML cell proliferation and induces apoptosis via regulation of BCR/ABL1 protein. Exp Cell Res 319: 1094–1101 Lv M, Zhang X, Jia H, Li D, Zhang B, Zhang H, Hong M, Jiang T, Jiang Q, Lu J, Huang X, Huang B, 2012, An oncogenic role of miR- 142-3p in human T-cell acute lymphoblastic leukemia, T-ALL, by targeting glucocorticoid receptor-alpha and cAMP/PKA pathways. Leukemia 26:769–777 Wang XS, Gong JN, Yu J,Wang F, Zhang XH, Yin XL, Tan ZQ, Luo ZM, Yang GH, Shen C, Zhang JW, 2012, MicroRNA-29a and microRNA-142-3p are regulators of myeloid differentiation and acute myeloid leukemia. Blood 119:4992–5004 Dou L, Li J, Zheng D, Li Y, Gao X, Xu C, Gao L, Wang L, Yu L, 2013, MicroRNA-142-3p inhibits cell proliferation in human acute lymphoblastic leukemia by targeting the MLL-AF4 oncogene. Mol Biol Rep 40:6811–6819 Palumbo T, Faucz FR, Azevedo M, Xekouki P, Iliopoulos D, Stratakis CA, 2013, Functional screen analysis reveals miR-26b and miR-128 as central regulators of pituitary somatomammotrophic tumor growth through activation of the PTEN-AKT pathway. Oncogene 32:1651–1659 Fazi F, Rosa A, Fatica A, Gelmetti V, De Marchis ML, Nervi C, Bozzoni I, 2005, A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPalpha regulates human granulopoiesis. Cell 123:819–831 Liu TY, Chen SU, Kuo SH, Cheng AL, Lin CW(2010, E2A-positive gastric MALT lymphoma has weaker plasmacytoid infiltrates and stronger expression of the memory B-cell-associated miR-223: possible correlation with stage and treatment response. Mod Pathol 23: 1507–1517 Mosakhani N, Sarhadi VK, Usvasalo A, Karjalainen-Lindsberg ML, Lahti L, Tuononen K, Saarinen-Pihkala UM, Knuutila S, 2012, MicroRNA profiling in pediatric acute lymphoblastic leukemia: novel prognostic tools. Leuk Lymphoma 53:2517–2520 de Oliveira JC, Brassesco MS, Scrideli CA, Tone LG, Narendran A, 2012)MicroRNA expression and activity in pediatric acute lymphoblastic leukemia, ALL). Pediatr Blood Cancer 59:599–604 Vigorito E, Kohlhaas S, Lu D, Leyland R, 2013, miR-155: an ancient regulator of the immune system. Immunol Rev 253:146–157 Akbari Moqadam F, Boer JM, Lange-Turenhout EA, Pieters R, den Boer ML, 2014, Altered expression of miR-24, miR-126 and miR- 365 does not affect viability of childhood TCF3-rearranged leukemia cells. Leukemia 28:1008–1014 Kaddar T, Chien WW, Bertrand Y, Pages MP, Rouault JP, Salles G, Ffrench M, Magaud JP, 2009, Prognostic value of miR-16 expression in childhood acute lymphoblastic leukemia relationships to normal and malignant lymphocyte proliferation. Leuk Res 33: 1217–1223 Mott JL, Kurita S, Cazanave SC, Bronk SF, Werneburg NW, Fernandez-Zapico ME, 2010, Transcriptional suppression of mir- 29b-1/mir-29a promoter by c-Myc, hedgehog, and NF-kappaB. J Cell Biochem 110:1155–1164 Mraz M, Kipps TJ, 2013, MicroRNAs and B cell receptor signaling in chronic lymphocytic leukemia. Leuk Lymphoma 54:1836–1839 Xi Y, Li J, Zan L, Wang J, Wang G, Ning Y, 2013, Micro-RNA-16 expression in paraffin-embedded specimen correlateswith overall survival of T-lymphoblastic lymphoma/leukemia. Hum Pathol 44:1011–1016 Mishra PJ, Humeniuk R, Mishra PJ, Longo-SorbelloGS, Banerjee D, Bertino JR, 2007, A miR-24 microRNA binding-site polymorphism in dihydrofolate reductase gene leads tomethotrexate resistance. Proc Natl Acad Sci U S A 104:13513–13518 Han BW, Feng DD, Li ZG, Luo XQ, Zhang H, Li XJ, Zhang XJ, Zheng LL, Zheng CW, Lin KY, Zhang P, Xu L, Chen YQ, 2011, A set of miRNAs that involve in the pathways of drug resistance and leukemic stem-cell differentiation is associated with the risk of relapse and glucocorticoid response in childhood ALL. Hum Mol Genet 20:4903–4915 Zhang H, Luo XQ, Zhang P, Huang LB, Zheng YS, Wu J, Zhou H, Qu LH, Xu L, Chen YQ, 2009, MicroRNA patterns associated with clinical prognostic parameters and CNS relapse prediction in pediatric acute leukemia. PLoS One 4:e7826 Alqurashi N, Hashimi SM, Wei MQ, 2013, Chemical Inhibitors and microRNAs, miRNA, Targeting the Mammalian Target of Rapamycin, mTOR, Pathway: Potential for Novel Anticancer Therapeutics. Int J Mol Sci 14:3874–3900 Jia CY, Li HH, Zhu XC, Dong YW, Fu D, Zhao QL,Wu W,Wu XZ, 2011, MiR-223 suppresses cell proliferation by targeting IGF-1R. PLoS One 6:e27008 Wang Y, Li Z, He C, DongmeiW, Xianggui Y, Jianjun C, Jie J, 2010, MicroRNAs expression signatures are associated with lineage and survival in acute leukemias. Blood Cells Mol Dis 44:191–197 Kotani A, Ha D, Hsieh J, Rao PK, Schotte D, den Boer ML, Armstrong SA, Lodish HF, 2009, miR-128b is a potent glucocorticoid sensitizer in MLL-AF4 acute lymphocytic leukemia cells and exerts cooperative effects with miR-221. Blood 114:4169–4178 Nemes K, Sebestyen A,Mark A, HajduM, Kenessey I, Sticz T, Nagy E, Barna G, Varadi Z, Kovacs G, Kopper L, Csoka M, 2013, Mammalian target of rapamycin, mTOR, activity dependent phospho-protein expression in childhood acute lymphoblastic leukemia, ALL). PLoS One 8:e59335 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 597 EP 604 DI 10.1007/s12253-014-9861-z PG 8 ER PT J AU Damjanovic, A Matic, ZI Ðordic, M Ðurovic, NM Nikolic, S Roki, K Milovanovic, Z Antic-Stankovic, J Dzodic, R Damjanovic, S Kanjer, K Abu Rabi, Z Juranic, Z AF Damjanovic, Ana Matic, Z Ivana Ðordic, Marija Ðurovic, Nikolic Marina Nikolic, Srdan Roki, Ksenija Milovanovic, Zorka Antic-Stankovic, Jelena Dzodic, Radan Damjanovic, Svetozar Kanjer, Ksenija Abu Rabi, Zaki Juranic, Zorica TI Metformin Effects on Malignant Cells and Healthy PBMC; The Influence of Metformin on the Phenotype of Breast Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer; Herceptin; Metformin; PBMC ID Cancer; Herceptin; Metformin; PBMC AB The aim of research was to determine the effects of maximally therapeutically achievable concentrations of metformin on malignant cells and healthy peripheral blood mononuclear cells (PBMC). Eight patients with T2D or hyperglycemia and nine healthy volunteers were included in the study. For determination of the influence of metformin on the phenotype of breast carcinoma, 1,410 patients with surgically removed tumors were included. From this group 37 breast cancer patients had DM type 2 or hyperglycemia and were pretreated with metformin alone or sometimes in combination with other antidiabetic drugs. Our results proved that metformin at low concentrations inducedmild decrease in survival of malignant cells and PBMC stimulated for proliferation, but it didn’t affect survival of resting PBMC. The effects of plasma of hyperglycemic patients who were under metformin therapy on autologous PBMC-induced decrease in survival of MDAMB- 361 cells, was noticeable in some patients. Metformin pretreatment for 24 h of HER2+ MDA-MB-361 cells, which were subsequently treated for 48 h with Herceptin, induced additional decline in cell survival. The analysis of influence of metformin on phenotype of breast cancer cells revealed significantly lower number of diabetic cancer patients treated with metformin with overexpressed HER2+ tumors (p<0.013), while the number of patientswith ER+PR+ tumors was not significantly changed (p<0.832). In conclusion, therapeutically used concentrations of metformin exhibit mild cytotoxic action on malignant and dividing normal cells pointing to its preferred role in malignant and autoimmune diseases. The use of metformin was associated with pronounced decrease in HER2 overexpressing tumors. C1 [Damjanovic, Ana] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Matic, Z Ivana] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Ðordic, Marija] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Ðurovic, Nikolic Marina] Clinical Center of Serbia, Clinic of Endocrinology, Diabetes and Metabolic Diseases, dr Subotica 13, 11000 Belgrade, Serbia. [Nikolic, Srdan] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Roki, Ksenija] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Milovanovic, Zorka] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Antic-Stankovic, Jelena] University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia. [Dzodic, Radan] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Damjanovic, Svetozar] Clinical Center of Serbia, Clinic of Endocrinology, Diabetes and Metabolic Diseases, dr Subotica 13, 11000 Belgrade, Serbia. [Kanjer, Ksenija] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Abu Rabi, Zaki] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Juranic, Zorica] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. RP Juranic, Z (reprint author), Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia. EM juranicz@ncrc.ac.rs CR Wang P, Kang D, Cao W, Wang Y, Liu Z, 2012, Diabetes mellitus and risk of hepatocellular carcinoma: a systematic review and metaanalysis. Diabetes Metab Res Rev 28:109–122., DOI 10.1002/dmrr. 1291 Hense HW, Kajuter H, Wellmann J, Batzler WU, 2011, Cancer incidence in type 2 diabetes patients—first results from a feasibility study of the D2C cohort. Diabetol Metab Syndr 3:15., DOI 10.1186/ 1758-5996-3-15 Tseng CH, 2011, Diabetes and risk of bladder cancer: a study using the National Health Insurance database in Taiwan. Diabetologia 54: 2009–2015., DOI 10.1007/s00125-011-2171-z Svacina S, 2011, Colorectal cancer and diabetes. Vnitr Lek 57:378– 380 Papa V, Belfiore A, 1996, Insulin receptors in breast cancer: biological and clinical role. J Endocrinol Investig 19:324–333 ShawRJ, LamiaKA,Vasquez D, Koo SH, Bardeesy N, Depinho RA, Montminy M, Cantley LC, 2005, The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science 310:1642–1646., DOI 10.1126/science.1120781 Algire C, Amrein L, Bazile M, David S, Zakikhani M, Pollak M, 2011, Diet and tumor LKB1 expression interact to determine sensitivity to anti-neoplastic effects of metformin in vivo. Oncogene 30: 1174–1182., DOI 10.1038/onc.2010.483 Liu B, Fan Z, Edgerton SM, Deng XS, Alimova IN, Lind SE, Thor AD, 2009, Metformin induces unique biological and molecular responses in triple negative breast cancer cells. Cell Cycle 8:2031– 2040., DOI 10.4161/cc.8.13.8814 Janjetovic K, Harhaji-Trajkovic L, Misirkic-Marjanovic M, Vucicevic L, Stevanovic D, Zogovic N, Sumarac-Dumanovic M, Micic D, Trajkovic V, 2011, In vitro and in vivo anti-melanoma action of metformin. Eur J Pharmacol 668:373–382., DOI 10.1016/j. ejphar.2011.07.004 Jalving M, Gietema JA, Lefrandt JD, de Jong S, Reyners AK, Gans RO, de Vries EG, 2010, Metformin: taking away the candy for cancer? Eur J Cancer 46:2369–2380., DOI 10.1016/j.ejca.2010.06.012 Sahra IB, Regazzetti C, Robert G, Laurent K, Le Marchand-Brustel Y, Auberger P, Tanti JF, Giorgetti-Peraldi S, Bost F, 2011, Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res 71:4366–4372., DOI 10. 1158/0008-5472.CAN-10-1769 Rocha GZ, Dias MM, Ropelle ER, Osorio-Costa F, Rossato FA, Vercesi AE, Saad MJ, Carvalheira JB, 2011, Metformin amplifies chemotherapy-induced AMPK activation and antitumoral growth. Clin Cancer Res 17:3993–4005., DOI 10.1158/1078-0432.CCR-10- 2243 Janjetovic K,Vucicevic L,MisirkicM, Vilimanovich U, TovilovicG, Zogovic N, Nikolic Z, Jovanovic S, Bumbasirevic V, Trajkovic V, Harhaji-Trajkovic L, 2011, Metformin reduces cisplatin-mediated apoptotic death of cancer cells through AMPK-independent activation of Akt. Eur J Pharmacol 651:41–50., DOI 10.1016/j.ejphar.2010. 11.005 Allavena P, Introna M, Sessa C, Mangioni C, Mantovani A, 1982, Interferon effect on cytotoxicity of peripheral blood and tumor-associated lymphocytes against human ovarian carcinoma cells. J Natl Cancer Inst 68:555–562 Introna M, Mantovani A, 1983, Natural killer cells in human solid tumors. Cancer Metastasis Rev 2:337–350 Liu F, Lang R, Zhao J, ZhangX, PringleGA, FanY,YinD,Gu F,Yao Z, Fu L, 2011, CD8+ cytotoxic T cell and FOXP3+ regulatory T cell infiltration in relation to breast cancer cell survival and molecular subtypes. Breast Cancer Res Treat 130:645–655., DOI 10.1007/ s10549-011-1647-3 Juranic ZD, Neskovic-Konstantinovic Z, Stanojkovic TP, Zizak Z, Srdic T, Stanojevic-Bakic N, Milosevic D, Jovanovic D, 2005, The antitumor immune response in HER-2 positive, metastatic breast cancer patients. J Transl Med 3:13., DOI 10.1186/1479-5876-3-13 Sliwkowski MX, Lofgren JA, Lewis GD, Hotaling TE, Fendly BM, Fox JA, 1999, Nonclinical studies addressing the mechanism of action of trastuzumab, Herceptin). Semin Oncol 26:60–70 Webster RM, Abraham J, Palaniappan N, Caley A, Jasani B, Barrett- Lee P, 2012, Exploring the use and impact of adjuvant Trastuzumab for HER2-positive breast cancer patients in a large UK cancer network. Do the results of international clinical trials translate into a similar benefit for patients in South EastWales? Br J Cancer 106:32– 38., DOI 10.1038/bjc.2011.506 Disis ML, Calenoff E,McLaughlin G,Murphy AE, ChenW, Groner B, Jeschke M, Lydon N, McGlynn E, Livingston RB, Moe R, Cheever MA, 1994, Existent T cell and antibody immunity to HER-2/neu protein in patients with breast cancer. Cancer Res 54: 16–20 Disis ML, Pupa SM, Gralow JR, Dittadi R, Menard S, Cheever MA, 1997, High titer HER-2/neu protein specific antibody immunity can be detected in patients with early stage breast cancer. J Clin Oncol 15: 3363–3367 Hou G, Zhang S, Zhang X,Wang P, Hao X, Zhang J, 2013, Clinical pathological characteristics and prognostic analysis of 1,013 breast cancer patients with diabetes. Breast Cancer Res Treat 137:807–816., DOI 10.1007/s10549-012-2404-y Besic N, Satej N, Ratosa I, Horvat AG,Marinko T, Gazic B, Petric R, 2014, Long-term use of metformin and the molecular subtype in invasive breast carcinoma patients—a retrospective study of clinical and tumor characteristics. BMC Cancer 14:298., DOI 10.1186/1471- 2407-14-298 Vazquez-Martin A, Oliveras-Ferraros C, Menendez JA, 2009, The antidiabetic drug metformin suppresses HER2, erbB-2, oncoprotein overexpression via inhibition of the mTOR effector p70S6K1 in human breast carcinoma cells. Cell Cycle 8:88–96., DOI 10.4161/cc. 8.1.7499 Chen TW, Liang YN, Feng D, Tao LY, Qi K, Zhang HY, Wang HX, Lin QS, Kong H, 2013, Metformin inhibits proliferation and promotes apoptosis of HER2 positive breast cancer cells by downregulating HSP90. J BUON 18:51–56 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 605 EP 612 DI 10.1007/s12253-014-9864-9 PG 8 ER PT J AU Banovic, M Mahovlic, V Salopek, MK Banovic, V Babic, I Oreskovic, S Babic, D AF Banovic, Maja Mahovlic, Vesna Salopek, Meljanac Kristina Banovic, Vladimir Babic, Ivan Oreskovic, Slavko Babic, Damir TI The Value of HPV-HR DNA Testing During the Follow-Up After Treatment of CIN3/AIS SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human papillomavirus; Preinvasive cervical disease; Postconisation; HPV-HR DNA test; Follow-up ID Human papillomavirus; Preinvasive cervical disease; Postconisation; HPV-HR DNA test; Follow-up C1 [Banovic, Maja] University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Obstetrics and Gynecology, Petrova 13, 10000 Zagreb, Croatia. [Mahovlic, Vesna] University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Pathology and CytologyZagreb, Croatia. [Salopek, Meljanac Kristina] University of Zagreb, School of Medicine, Department of PathologyZagreb, Croatia. [Banovic, Vladimir] University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Obstetrics and Gynecology, Petrova 13, 10000 Zagreb, Croatia. [Babic, Ivan] University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Obstetrics and Gynecology, Petrova 13, 10000 Zagreb, Croatia. [Oreskovic, Slavko] University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Obstetrics and Gynecology, Petrova 13, 10000 Zagreb, Croatia. [Babic, Damir] University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Pathology and CytologyZagreb, Croatia. RP Banovic, M (reprint author), University of Zagreb, School of Medicine, Clinical Hospital Center Zagreb, Department of Obstetrics and Gynecology, 10000 Zagreb, Croatia. EM mzanko03@gmail.com CR Nuovo J, Melnikow J, Willan AR, Chan BK, 2000, Treatment outcomes for squamous intraepithelial lesions. Int J Gynaecol Obstet 68(1):25–33 Arbyn M, Ronco G, Anttila A, Meijer CJ, Poljak M, Ogilvie G et al, 2012, Evidence regarding human papillomavirus testing in secondary prevention of cervical cancer. Vaccine 20(30 Suppl 5):F88–99 MartinHirsch PL, Paraskevaidis E, Kitchener H, 2000, Surgery for cervical intraepithelial neoplasia. Cochrane Database Syst Rev 2000;(2):CD001318 Bae JH, KimCJ, Park TC, Namkoong SE, Park JS, 2007, Persistence of human papillomavirus as a predictor for treatment failure after loop electrosurgical excision procedure. Int J Gynecol Cancer 17(6): 1271–7 Prato B, Ghelardi A, Gadducci A, Marchetti I, Di Cristofano C, Di Coscio G et al, 2008, Correlation of recurrence rates and times with posttreatment human papillomavirus status in patients treated with loop electrosurgical excision procedure conization for cervical squamous intraepithelial lesions. Int J Gynecol Cancer 18(1):90–4 Costa S, De Simone P, Venturoli S, Cricca M, Zerbini ML, Musiani M et al, 2003, Factors predicting human papillomavirus clearance in cervical intraepithelial neoplasia lesions treated by conization. Gynecol Oncol 90(2):358–65 Ovanin-Rakic A, Pajtler M, Stankovic T et al, 2003, Classification of cytologic cervical smears ‘’Zagreb 2002”.Modification of classifications ‘’Zagreb 1991 and NCI Bethesda system 2001”. Gynaecol Perinatol 12: 148–53 Katki HA, Schiffman M, Castle PE, Fetterman B, Poitras NE, Lorey T, Cheung LC, Raine-Bennett T, Gage JC, Kinney WK, 2013, Fiveyear risk of recurrence after treatment of CIN 2, CIN 3, or AIS: performance of HPVand Pap cotesting in posttreatment management. J Low Genit Tract Dis 17(5 Suppl 1):S78–84., DOI 10.1097/LGT. 0b013e31828543c5 Almog B, Gamzu R, Bornstein J, Levin I, Fainaru O, Niv J, Lessing JB, Bar-Am A, 2003, Clinical and economic benefit of HPV-load testing in follow-up and management of women postcone biopsy for CIN2-3. Br J Cancer Jul 7;89(1):109–12. Coupe VM, Berkhof J, Verheijen RH, Meijer CJ, 2007, Costeffectiveness of human papillomavirus testing after treatment for cervical intraepithelial neoplasia. BJOG 114(4):416–24 Gok M, Coupe VM, Berkhof J, Verheijen RH, Helmerhorst TJ, Hogewoning CJ et al, 2007, HPV16 and increased risk of recurrence after treatment for CIN. Gynecol Oncol 104(2):273–5 Stanley M, 2010, Pathology and epidemiology of HPV infection in females. Gynecol Oncol 117(2 Suppl):S5–10., DOI 10.1016/j.ygyno. 2010.01.024 Du R, Meng W, Chen ZF, Zhang Y, Chen SY, Ding Y, 2013, Posttreatment human papillomavirus status and recurrence rates in patients treated with loop electrosurgical excision procedure conization for cervical intraepithelial neoplasia. Eur J Gynaecol Oncol 34(6):548–51 Zielinski GD, Bais AG, Helmerhorst TJ, Verheijen RH, de Schipper FA, Snijders PJ, Voorhorst FJ, van Kemenade FJ, Rozendaal L, Meijer CJ, 2004, HPV testing and monitoring of women after treatment of CIN 3: review of the literature and meta-analysis. Obstet Gynecol Surv 59(7):543–53 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 613 EP 617 DI 10.1007/s12253-014-9865-8 PG 5 ER PT J AU Hever, VN Pentek, M Ballo, A Gulacsi, L Baji, P Brodszky, V Damasdi, M Bognar, Z Toth, Gy Buzogany, I Szanto, AF Hever, V Noemi Pentek, Marta Ballo, Andras Gulacsi, Laszlo Baji, Petra Brodszky, Valentin Damasdi, Miklos Bognar, Zita Toth, Gyorgy Buzogany, Istvan Szanto, Arpad TI Health Related Quality of Life in Patients with Bladder Cancer: A Cross-Sectional Survey and Validation Study of the Hungarian Version of the Bladder Cancer Index SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; Quality of life; EQ-5D; FACT-Bl; Bladder Cancer Index; Validation ID Bladder cancer; Quality of life; EQ-5D; FACT-Bl; Bladder Cancer Index; Validation AB Health-related quality of life (HRQoL) is an important outcome in oncology care although an underexplored area in bladder cancer (BC). Our aims were to assess HRQoL of patients with BC, analyse relationships between diverse HRQoL measures and validate the Hungarian version of the Bladder Cancer Index (BCI) questionnaire. A cross-sectional survey was performed among patients with BC (N=151). Validated Hungarian versions of the FACT-Bl, SF-36 and EQ-5D were applied and SF-6D was derived. Psychometric analysis of the Hungarian BCI was performed. Pearson correlations between the five measures were analysed. Deterioration in SF-36 Physical Functioning was detected among patients aged 45–64 years. The EQ-5D score did not differ significantly from the age-matched population norm. Correlations between the FACT-Bl, EQ-5D and SF-6D utility measures were strong (r>0.6). Cronbach alpha coefficients of the Hungarian BCI ranged from 0.75 to 0.97 and factor analysis confirmed that data fit to the six predefined subdomains. Test-retest correlations (reliability, N=50) ranged from 0.67 to 0.87 and interscale correlations between urinary, bowel and sexual BCI domains were weak or moderate (r=0.29 to 0.49). Convergent validity revealed a stronger correlation with FACT-Bl (r=0.126 to 0.719) than with generic health state scores (r=0.096 to 0.584). Results of divergent validity of the Hungarian BCI by treatment groups by Kruskal Wallis test were promising although limited by low sample sizes in cystectomy subgroups. Generic health state measures have limited capacity to capture HRQoL impact of BC. Validity tests yielded favourable results for the Hungarian BCI. Mapping studies to estimate utility scores from FACT-Bl are encouraged but less recommendable with the BCI. C1 [Hever, V Noemi] Corvinus University of Budapest, Department of Health Economics, Fovam ter 8., 1093 Budapest, Hungary. [Pentek, Marta] Corvinus University of Budapest, Department of Health Economics, Fovam ter 8., 1093 Budapest, Hungary. [Ballo, Andras] University of Pecs, Department of Urology, Munkacsy Mihaly utca 2, 7621 Pecs, Hungary. [Gulacsi, Laszlo] Corvinus University of Budapest, Department of Health Economics, Fovam ter 8., 1093 Budapest, Hungary. [Baji, Petra] Corvinus University of Budapest, Department of Health Economics, Fovam ter 8., 1093 Budapest, Hungary. [Brodszky, Valentin] Corvinus University of Budapest, Department of Health Economics, Fovam ter 8., 1093 Budapest, Hungary. [Damasdi, Miklos] University of Pecs, Department of Urology, Munkacsy Mihaly utca 2, 7621 Pecs, Hungary. [Bognar, Zita] University of Pecs, Institute of Biochemistry and Medical Chemistry, Szigeti ut 12, 7624 Pecs, Hungary. [Toth, Gyorgy] Josa Andras Hospital, Department of Urology, Szent Istvan ut 68, 4400 Nyiregyhaza, Hungary. [Buzogany, Istvan] Peterfy Hospital, Department of Urology, Peterfy S u. 8-20, 1076 Budapest, Hungary. [Szanto, Arpad] University of Pecs, Department of Urology, Munkacsy Mihaly utca 2, 7621 Pecs, Hungary. RP Pentek, M (reprint author), Corvinus University of Budapest, Department of Health Economics, 1093 Budapest, Hungary. EM marta.pentek@uni-corvinus.hu CR Ploeg M, Aben KK, Kiemeney LA, 2009, The present and future burden of urinary bladder cancer in the world. World J Urol 27(3): 289–293., DOI 10.1007/s00345-009-0383-3 BottemanMF, Pashos CL, Hauser RS, Laskin BL, Redaelli A, 2003, Quality of life aspects of bladder cancer: a review of the literature. Qual Life Res 12(6):675–688 Ali AS, Hayes MC, Birch B, Dudderidge T, Somani BK, 2014, Health related quality of life, HRQoL, after cystectomy: comparison between orthotopic neobladder and ileal conduit diversion. Eur J Surg Oncol., DOI 10.1016/j.ejso.2014.05.006 Gilbert SM, Dunn RL, Hollenbeck BK, Montie JE, Lee CT, Wood DP,Wei JT, 2010, Development and validation of the bladder cancer index: a comprehensive, disease specific measure of health related quality of life in patients with localized bladder cancer. J Urol 183(5): 1764–1769., DOI 10.1016/j.juro.2010.01.013 Hedgepeth RC, Gilbert SM, He C, Lee CT,Wood DP Jr, 2010, Body image and bladder cancer specific quality of life in patients with ileal conduit and neobladder urinary diversions. Urology 76(3):671–675., DOI 10.1016/j.urology.2010.01.087 Bartsch G, Daneshmand S, Skinner EC, Syan S, Skinner DG, Penson DF, 2014, Urinary functional outcomes in female neobladder patients. World J Urol 32(1):221–228., DOI 10.1007/s00345-013-1219-8 Poch MA, Stegemann AP, Rehman S, Sharif MA, Hussain A, Consiglio JD, Wilding GE, Guru KA, 2014, Short-term patient reported health-related quality of life, HRQL, outcomes after robotassisted radical cystectomy, RARC). BJU Int 113(2):260–265., DOI 10.1111/bju.12162 Aboumohamed AA, Raza SJ, Al-Daghmin A, Tallman C, Creighton T, Crossley H, Dailey S, Khan A, Din R,MehedintD,Wang K, ShiY, Sharif M, Wilding G, Weizer A, Guru KA, 2014, Health-related quality of life outcomes after robot-assisted and open radical cystectomy using a validated bladder-specific instrument: a multiinstitutional study. Urology., DOI 10.1016/j.urology.2014.02.024 Gaunez N, Larre S, Pires C, Dore B, Wei J, Pfister C, Irani J, 2010, French translation and linguistic validation of the questionnaire bladder cancer index, BCI). Prog Urol 22(6):350–353., DOI 10.1016/j. purol.2011.12.004 Schmidt S, Riel R, Frances A, Lorente Garin JA, BonfillX,Martinez- Zapata MJ, Morales Suarez-Varela M, dela Cruz J, Emparanza JI, SanchezMJ, Zamora J, Goni JM, Alonso J, FerrerM(2014, Bladder cancer index: cross-cultural adaptation into Spanish and psychometric evaluation. Health Qual Life Outcomes 12:20., DOI 10.1186/1477- 7525-12-20 Yeung C, Dinh T, Lee J, 2014, The health economics of bladder cancer: an updated review of the published literature. Pharmacoeconomics., DOI 10.1007/s40273-014-0194-2 Torrance GW, 1986, Measurement of health state utilities for economic appraisal. J Health Econ 5(1):1–30 Svatek RS, Hollenbeck BK, Holmang S, Lee R, Kim SP, Stenzl A, Lotan Y, 2014, The economics of bladder cancer: costs and considerations of caring for this disease. Eur Urol., DOI 10.1016/j.eururo. 2014.01.006 McTaggart-Cowan H, Teckle P, Peacock S, 2013, Mapping utilities from cancer-specific health-related quality of life instruments: a review of the literature. Expert Rev Pharmacoecon Outcomes Res 13(6):753–765., DOI 10.1586/14737167.2013.850420 Tejido-Sanchez A,Garcia-Gonzalez L, Jimenez-Alcaide E,Arrebola- Pajares A, Medina-Polo J, Villacampa-Auba F, Diaz-Gonzalez R, 2014, Quality of life in patients with ileal conduit cystectomy due to bladder cancer. Actas Urol Esp 38(2):90–95., DOI 10.1016/j.acuro. 2013.04.006 Li X, Fang Q, Ji H, Pan J, Zhang J, Li Z, Chen M, Wu X, Zhou Z, Chen Z, 2014, Use of urostomy bags in the management of perioperative urine leakage after radical cystectomy. Cancer Nurs 37(3): 170–174., DOI 10.1097/NCC.0b013e318277db29 Kulkarni GS, Alibhai SM, Finelli A, Fleshner NE, Jewett MA, Lopushinsky SR, Bayoumi AM, 2009, Cost-effectiveness analysis of immediate radical cystectomy versus intravesical bacillus Calmette-Guerin therapy for high-risk, high-grade, T1G3, bladder cancer. Cancer 115(23):5450–5459., DOI 10. 1002/cncr.24634 Wong KA, Zisengwe G, Athanasiou T, O’Brien T, Thomas K, 2013, Outpatient laser ablation of non-muscle-invasive bladder cancer: is it safe, tolerable and cost-effective? BJU Int 112(5):561–567., DOI 10. 1111/bju.12216 Cella DF, Tulsky DS, Gray G, Sarafian B, Linn E, Bonomi A, Silberman M, Yellen SB, Winicour P, Brannon J et al, 1993, The functional assessment of cancer therapy scale: development and validation of the general measure. J Clin Oncol 11(3):570– 579 Rabin R, de Charro F, 2001, EQ-5D: a measure of health status from the EuroQol group. Ann Med 33(5):337–343 McHorney CA,Ware JE Jr, Lu JF, Sherbourne CD, 1994, TheMOS 36-item short-form health survey, SF-36): III. Tests of data quality, scaling assumptions, and reliability across diverse patient groups. Med Care 32(1):40–66 Brazier J, Usherwood T, Harper R, Thomas K, 1998, Deriving a preference-based single index from the UK SF-36 health survey. J Clin Epidemiol 51(11):1115–1128 Terwee CB, Bot SD, de Boer MR, van der Windt DA, Knol DL, Dekker J, Bouter LM, de Vet HC, 2007, Quality criteria were proposed for measurement properties of health status questionnaires. J Clin Epidemiol 60(1):34–42., DOI 10.1016/j.jclinepi.2006.03.012 Cohen J, 1992, A power primer. Psychol Bull 112(1):155–159 Szende A, Nemeth R, 2003, Health-related quality of life of the Hungarian population. Orv Hetil 144(34):1667–1674 Czimbalmos A, Nagy Z, Varga Z, Hustik P, 1999, Paciens megelegedettsegi vizsgalat SF-36 kerdoivvel, a magyarorszagi normalertekek meghatarozasa. Nepegeszsegugy 80(1):4–19 Sousa VD, Rojjanasrirat W, 2011, Translation, adaptation and validation of instruments or scales for use in cross-cultural health care research: a clear and user-friendly guideline. J Eval Clin Pract 17(2): 268–274., DOI 10.1111/j.1365-2753.2010.01434.x NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 619 EP 627 DI 10.1007/s12253-014-9866-7 PG 9 ER PT J AU Abderrahmane, R Louhibi, L Moghtit, ZF Boubekeur, A Benseddik, K Boudjema, A Benrrahal, F Aberkane, M Fodil, M Saidi-Mehtar, N AF Abderrahmane, Rym Louhibi, Lotfi Moghtit, Zohra Fatima Boubekeur, Amina Benseddik, Khedidja Boudjema, Abdellah Benrrahal, Fouzia Aberkane, Meriem Fodil, Mostefa Saidi-Mehtar, Nadhira TI TP53 Arg 72Pro and MDM2 SNP309 Polymorphisms and Colorectal Cancer Risk: A West Algerian Population Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Polymorphism; TP53 Arg72Pro; MDM2 SNP309; CRC; Algerian population; Case/Control study ID Polymorphism; TP53 Arg72Pro; MDM2 SNP309; CRC; Algerian population; Case/Control study AB The tumor suppressor gene TP53 and its regulator MDM2 are both key players involved in multiple pathways including apoptosis, cellular transcriptional control and cell cycle regulation. Common germline polymorphisms in these genes may affect colorectal cancer (CRC) susceptibility. An arginine-to-proline substitution at codon 72 in the TP53 gene is reported to decrease apoptotic potential, while a thymine-toguanine polymorphism at nucleotide 309 (named SNP309) of murine double minute 2 MDM2 gene increases its transcription. These two polymorphisms therefore may be of importance in colorectal carcinogenesis. The relation of these polymorphisms to colorectal cancer in the Algerian population was addressed in this study. DNA samples from 121 controls and 116 cases were genotyped for these two polymorphisms by PCR/RFLP then confirmed by sequencing. Unexpectedly no significant association was found between this potential marker TP53 Arg72Pro and CRC (p>0.05). However, our findings reveal that individuals with the MDM2 SNP309 GG genotype have a low risk of CRC as compared to the TT genotype (OR=0.49; 95%CI: 0.24–0.98, p=0.04),withmore significance for females (OR=0.16; 95 % CI: 0.06–0.41, p<0.05). Moreover, no significant association was observed between the combined TP53 and MDM2 genotypes and CRC. Contrary to initial expectations that the GG genotype with high MDM2 levels would increase cancer risk, our results demonstrate that the MDM2 SNP309 GG genotype is associated with decreased risk of colorectal cancer. This is suggesting that other mechanisms independent of increased MDM2 levels can influence cancer susceptibility. C1 [Abderrahmane, Rym] Universite des Sciences et de la Technologie d’Oran- Mohamed BOUDIAF-USTOMB, Faculte des sciences de la nature et de la vie, Departement de Genetique Moleculaire Appliquee, Laboratoire de Genetique Moleculaire et Cellulaire, BP 1505 El M’naouer, 31000 Oran, Algeria. [Louhibi, Lotfi] Universite des Sciences et de la Technologie d’Oran- Mohamed BOUDIAF-USTOMB, Faculte des sciences de la nature et de la vie, Departement de Genetique Moleculaire Appliquee, Laboratoire de Genetique Moleculaire et Cellulaire, BP 1505 El M’naouer, 31000 Oran, Algeria. [Moghtit, Zohra Fatima] Universite des Sciences et de la Technologie d’Oran- Mohamed BOUDIAF-USTOMB, Faculte des sciences de la nature et de la vie, Departement de Genetique Moleculaire Appliquee, Laboratoire de Genetique Moleculaire et Cellulaire, BP 1505 El M’naouer, 31000 Oran, Algeria. [Boubekeur, Amina] Universite des Sciences et de la Technologie d’Oran- Mohamed BOUDIAF-USTOMB, Faculte des sciences de la nature et de la vie, Departement de Genetique Moleculaire Appliquee, Laboratoire de Genetique Moleculaire et Cellulaire, BP 1505 El M’naouer, 31000 Oran, Algeria. [Benseddik, Khedidja] Centre de Recherche en Cancerologie de MarseilleMarseille, France. [Boudjema, Abdellah] Universite des Sciences et de la Technologie d’Oran- Mohamed BOUDIAF-USTOMB, Faculte des sciences de la nature et de la vie, Departement de Genetique Moleculaire Appliquee, Laboratoire de Genetique Moleculaire et Cellulaire, BP 1505 El M’naouer, 31000 Oran, Algeria. [Benrrahal, Fouzia] Centre Hospitalier d’Oran, 76, Bd Dr Benzerdjeb - PlateauOran, Algeria. [Aberkane, Meriem] Universite d’Oran, Faculte de Medecine, Departement de Pharmacie, BP 1524 El M’naouer, 31000 Oran, Algeria. [Fodil, Mostefa] Universite des Sciences et de la Technologie d’Oran- Mohamed BOUDIAF-USTOMB, Faculte des sciences de la nature et de la vie, Departement de Genetique Moleculaire Appliquee, Laboratoire de Genetique Moleculaire et Cellulaire, BP 1505 El M’naouer, 31000 Oran, Algeria. [Saidi-Mehtar, Nadhira] Universite des Sciences et de la Technologie d’Oran- Mohamed BOUDIAF-USTOMB, Faculte des sciences de la nature et de la vie, Departement de Genetique Moleculaire Appliquee, Laboratoire de Genetique Moleculaire et Cellulaire, BP 1505 El M’naouer, 31000 Oran, Algeria. RP Abderrahmane, R (reprint author), Universite des Sciences et de la Technologie d’Oran- Mohamed BOUDIAF-USTOMB, Faculte des sciences de la nature et de la vie, Departement de Genetique Moleculaire Appliquee, Laboratoire de Genetique Moleculaire et Cellulaire, 31000 Oran, Algeria. EM abderrahmane_rym@yahoo.fr CR Ballinger AB, Anggiansah C, 2007, Colorectal cancer. BMJ 335(7622):715–718., DOI 10.1136/bmj.39321.527384.BE Meddah D, Meddah B, Tir Touil A, Ghalek M, Sahraoui T, 2009, Etude epidemiologique du cancer du colon chez des patients de l’Westalgerien. J Afr Cancer 1(1):31–35., DOI 10.1007/s12558-008- 0006-8 Honda R, Tanaka H, Yasuda H, 1997, Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett 420(1): 25–27., DOI 10.1016/S0014-5793(97)01480-4 Kubbutat MH, Jones SN, Vousden KH, 1997, Regulation of p53 stability by Mdm2. Nature 387(6630):299–303 Whibley C, Pharoah PD, Hollstein M, 2009, p53 polymorphisms: cancer implications. Nat Rev Cancer 9(2):95–107 SjalanderA, Birgander R, Athlin L, Stenling R, Rutegard J, Beckman L, Beckman G, 1995, P53 germ line haplotypes associated with increased risk for colorectal cancer. Carcinogenesis 16(7):1461–1464 Gemignani F,Moreno V, Landi S, Moullan N, Chabrier A, Gutierrez- Enriquez S, Hall J, Guino E, Peinado MA, Capella G, Canzian F, 2004, A TP53 polymorphism is associated with increased risk of colorectal cancer and with reduced levels of TP53mRNA. Oncogene 23(10):1954–1956 Zhu ZZ, Wang AZ, Jia HR, Jin XX, He XL, Hou LF, Zhu G, 2007, Association of the TP53 codon 72 polymorphism with colorectal cancer in a Chinese population. Jpn J Clin Oncol 37(5):385–390 Cao Z, Song JH, Park YK, Maeng EJ, Nam SW, Lee JY, Park WS, 2009, The p53 codon 72 polymorphism and susceptibility to colorectal cancer in Korean patients. Neoplasma 56(2):114–118 Olschwang S, Laurent-Puig P, Vassal A, Salmon RJ, Thomas G, 1991, Characterization of a frequent polymorphism in the coding sequence of the Tp53 gene in colonic cancer patients and a control population. Hum Genet 86(4):369–370 Kawajiri K, Nakachi K, Imai K,Watanabe J, Hayashi S, 1993, Germ line polymorphisms of p53 and CYP1A1 genes involved in human lung cancer. Carcinogenesis 14(6):1085–1089 Murata M, Tagawa M, Kimura M, Kimura H,Watanabe S, Saisho H, 1996, Analysis of a germ line polymorphismof the p53 gene in lung cancer patients; discrete results with smoking history. Carcinogenesis 17(2):261–264 Sayhan N, Yazici H, BudakM, Bitisik O, Dalay N, 2001, P53 codon 72 genotypes in colon cancer. Association with human papillomavirus infection. Res Commun Mol Pathol Pharmacol 109(1–2):25–34 Hamajima N, Matsuo K, Suzuki T, Nakamura T, Matsuura A, Hatooka S, Shinoda M, Kodera Y, Yamamura Y, Hirai T, Kato T, Tajima K, 2002, No associations of p73 G4C14-to-A4T14 at exon 2 and p53 Arg72Pro polymorphisms with the risk of digestive tract cancers in Japanese. Cancer Lett 181(1):81–85 Perez LO, Abba MC, Dulout FN, Golijow CD, 2006, Evaluation of p53 codon 72 polymorphism in adenocarcinomas of the colon and rectum in La Plata, Argentina. World J Gastroenterol 12(9): 1426–1429 Koushik A, Tranah GJ, Ma J, Stampfer MJ, Sesso HD, Fuchs CS, Giovannucci EL, Hunter DJ, 2006, p53 Arg72Pro polymorphism and risk of colorectal adenoma and cancer. Int J Cancer 119(8): 1863–1868 Dakouras A, Nikiteas N, Papadakis E, Perakis M, Valis D, Rallis G, Tzanakis N, Peros G, Tsigkris C, Kittas C, Karakitsos P, 2008, P53Arg72 homozygosity and its increased incidence in left-sided sporadic colorectal adenocarcinomas, in a Greek-Caucasian population. Anticancer Res 28(2A):1039–1043 Csejtei A, Tibold A, Varga Z, Koltai K, Ember A, Orsos Z, Feher G, Horvath OP, Ember I, Kiss I, 2008, GSTM, GSTT and p53 polymorphisms asmodifiers of clinical outcome in colorectal cancer.Anticancer Res 28(3B):1917–1922 Mammano E, Belluco C, Bonafe M, Olivieri F, Mugianesi E, Barbi C, Mishto M, Cosci M, Franceschi C, Lise M, Nitti D, 2009, Association of p53 polymorphisms and colorectal cancer:modulation of risk and progression. Eur J Surg Oncol 35(4):415–419 Bond GL, Hu W, Bond EE, Robins H, Lutzker SG, Arva NC, Bargonetti J, Bartel F, Taubert H, Wuerl P, Onel K, Yip L, Hwang SJ, Strong LC, Lozano G, Levine AJ, 2004, A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell 119(5):591–602 Bond GL, Hu W, Levine A, 2005, A Single nucleotide polymorphism in theMDM2 Gene: froma molecular and cellular explanation to clinical effect. Cancer Res 65(13):5481–5484., DOI 10.1158/0008- 5472.can-05-0825 BondGL, Levine AJ, 2007)Asingle nucleotide polymorphismin the p53 pathway interacts with gender, environmental stresses and tumor genetics to influence cancer in humans. Oncogene 26(9):1317–1323 Hong Y,Miao X, Zhang X, Ding F, Luo A, Guo Y, TanW, Liu Z, Lin D, 2005, The role of P53 and MDM2 polymorphisms in the risk of esophageal squamous cell carcinoma. Cancer Res 65(20):9582–9587 Schmidt MK, Reincke S, Broeks A, Braaf LM, Hogervorst FB, Tollenaar RA, Johnson N, Fletcher O, Peto J, Tommiska J, Blomqvist C, Nevanlinna HA, Healey CS, Dunning AM, Pharoah PD, Easton DF, Dork T, Van’t Veer LJ, 2007, Do MDM2 SNP309 and TP53 R72P interact in breast cancer susceptibility? A large pooled series from the breast cancer association consortium. Cancer Res 67(19):9584–9590 Horikawa Y, Nadaoka J, Saito M, Kumazawa T, Inoue T, Yuasa T, Tsuchiya N, Nishiyama H, Ogawa O, Habuchi T, 2008, Clinical implications of theMDM2 SNP309 and p53Arg72Pro polymorphisms in transitional cell carcinoma of the bladder. Oncol Rep 20(1):49–55 Talseth BA,Meldrum C, Suchy J, Kurzawski G, Lubinski J, Scott RJ, 2007)MDM2 SNP309 T>G alone or in combination with the TP53 R72P polymorphism does not appear to influence disease expression and age of diagnosis of colorectal cancer in HNPCC patients. Int J Cancer 120(3):563–565 Miller SA, Dykes DD, Polesky HF, 1988, A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16(3):1215 Dokianakis DN, Koumantaki E, Billiri K, Spandidos DA, 2000, P53 codon 72 polymorphism as a risk factor in the development of HPVassociated non-melanoma skin cancers in immunocompetent hosts. Int J Mol Med 5(4):405–409 Matlashewski GJ, Tuck S, Pim D, Lamb P, Schneider J, Crawford LV, 1987, Primary structure polymorphism at amino acid residue 72 of human p53. Mol Cell Biol 7(2):961–963 Mabrouk I, Baccouche S, El-Abed R, Mokdad-Gargouri R, Mosbah A, Said S, Daoud J, Frikha M, Jlidi R, Gargouri A, 2003, No evidence of correlation between p53 codon 72 polymorphism and risk of bladder or breast carcinoma in Tunisian patients. Ann N Y Acad Sci, :764–770 Zehbe I, Voglino G, Wilander E, Genta F, 1999, Tommasino M Codon 72 polymorphism of p53 and its association with cervical cancer. Lancet 354(9174):218–219 Noma C, Miyoshi Y, Taguchi T, Tamaki Y, Noguchi S, 2004, Association of p53 genetic polymorphism, Arg72Pro, with estrogen receptor positive breast cancer risk in Japanese women. Cancer Lett 210(2):197–203 Tornesello ML, Biryahwaho B, Downing R, Hatzakis A, Alessi E, Cusini M, Ruocco V, Katongole-Mbidde E, Buonaguro L, Buonaguro FM, 2009, TP53 codon 72 polymorphism in classic, endemic and epidemic Kaposi’s sarcoma in African and Caucasian patients. Oncology 77(5):328–334 Pegoraro R,Moodley J, Naiker S, Lanning P, Rom L, 2000, The p53 codon 72 polymorphism in black South African women and the risk of cervical cancer. BJOG: Int J Obstet Gynaecol 107(9):1164–1165., DOI 10.1111/j.1471-0528.2000.tb11118.x Peixoto Guimaraes D, Hsin Lu S, Snijders P,Wilmotte R, Herrero R, Lenoir G,Montesano R,Meijer CJ,Walboomers J, Hainaut P, 2001, Absence of association between HPV DNA, TP53 codon 72 polymorphism, and risk of oesophageal cancer in a high-risk area of China. Cancer Lett 162(2):231–235 Economopoulos KP, Sergentanis TN, Zagouri F, Zografos GC, 2010, Association between p53 Arg72Pro polymorphism and colorectal cancer risk: a meta-analysis. Onkologie 33(12):666–674 Cao X, Zhang T, Zhao Z, Zhao T, 2012, MDM2 SNP309 polymorphism and colorectal cancer risk: a meta-analysis. DNA Cell Biol 31(3):355–359 Fang F, Yu XJ, Yu L,Yao L, 2011)MDM2 309 T/G polymorphism is associated with colorectal cancer risk especially in Asians: a metaanalysis. Med Oncol 28(4):981–985 Qin X, Peng Q, TangW, Lao X, Chen Z, Lai H, Deng Y, Mo C, Sui J, Wu J, Zhai L, Yang S, Li S, Zhao J, 2013, An updated meta-analysis on the association of MDM2 SNP309 polymorphism with colorectal cancer risk. PLoS ONE 8(9):0076031 Chaar I, Arfaoui TA, el HO EA,Mahmoud LB, KhiariM, Sammoud S, Lounis A, Amara S, Gharbi L, Hmida AB, Mzabi S, Bouraoui S, 2012, Impact of MDM2 polymorphism: increased risk of developing colorectal cancer and a poor prognosis in the Tunisian population. Eur J Gastroenterol Hepatol 24(3):320–327 Brekman A, Singh KE, Polotskaia A, Kundu N, Bargonetti J, 2011, A p53-independent role of Mdm2 in estrogen-mediated activation of breast cancer cell proliferation. Breast Cancer Res 13, 1) Bond GL, Hirshfield KM, Kirchhoff T, Alexe G, Bond EE, Robins H, Bartel F, Taubert H,Wuerl P, HaitW, Toppmeyer D, Offit K, Levine AJ, 2006)MDM2SNP309 accelerates tumor formation in a gender-specific and hormone-dependent manner. Cancer Res 66(10):5104–5110 Gu L, Findley HW, Zhou M(2002)MDM2 induces NF-kappaB/p65 expression transcriptionally through Sp1-binding sites: a novel, p53- independent role of MDM2 in doxorubicin resistance in acute lymphoblastic leukemia. Blood 99(9):3367–3375 Hu W, Feng Z, Ma L, Wagner J, Rice JJ, Stolovitzky G, Levine AJ, 2007, A single nucleotide polymorphism in the MDM2 gene disrupts the oscillation of p53 and MDM2 levels in cells. Cancer Res 67(6):2757–2765 Petz LN, Ziegler YS, Schultz JR, Kim H, Kemper JK, Nardulli AM, 2004, Differential regulation of the human progesterone receptor gene through an estrogen response element half site and Sp1 sites. J Steroid Biochem Mol Biol 88(2):113–122 Knappskog S, Bjornslett M, Myklebust LM, Huijts PE, Vreeswijk MP, Edvardsen H, Guo Y, Zhang X, Yang M, Ylisaukko-Oja SK, Alhopuro P, Arola J, Tollenaar RA, van Asperen CJ, Seynaeve C, Staalesen V, Chrisanthar R, Lokkevik E, Salvesen HB, Evans DG, Newman WG, Lin D, Aaltonen LA, Borresen-Dale AL, Tell GS, Stoltenberg C, Romundstad P, Hveem K, Lillehaug JR, Vatten L, Devilee P, Dorum A, Lonning PE, 2011, The MDM2 promoter SNP285C/309G haplotype diminishes Sp1 transcription factor binding and reduces risk for breast and ovarian cancer in Caucasians. Cancer Cell 19(2):273–282 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 629 EP 635 DI 10.1007/s12253-014-9867-6 PG 7 ER PT J AU Wu, J Chen, P Xie, Yg Gong, Nm Sun, Ll Sun, Chf AF Wu, Jing Chen, Ping Xie, Yang-gui Gong, Nian-mei Sun, Lin-lin Sun, Chun-feng TI Comparison of the Effectiveness and Safety of Ultrasoundand CT-Guided Percutaneous Radiofrequency Ablation of Non-Operation Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE US guidance; CT guidance; Radiofrequency ablation; Hepatocellular carcinoma ID US guidance; CT guidance; Radiofrequency ablation; Hepatocellular carcinoma AB To retrospectively compare the effectiveness and safety of ultrasound (US)- and computer tomography (CT)- guided percutaneous radiofrequency ablation (PRFA) in treating patients with non-operation hepatocellular carcinoma (HCC). Forty patients with non-operation HCC who were treated with US-guided PRFA (20 patients with 24 HCC lesions) or CT-guided PRFA (20 patients with 27 HCC lesions) were enrolled in this study. Follow-up was performed with US and CT/MRI. Complete ablation rate, local recurrence rate, and overall survival rate were used to evaluate the efficacy of the two therapeutic choices. The PRFA-related complications including hilar bile duct injury, sepsis, liver failure, renal dysfunction, peritoneal hemorrhage, and skin burn were assessed. The operation time of CT-guided group was significantly longer than that of the US-guided group (P<0.05). The single ablation times for tumors with similar size showed no significant difference between the two groups (P>0.05). The differences in complete ablation rate (79.2 vs. 88.9 %, P>0.05) and local recurrence rate (16.7 vs. 14.8 %, P>0.05) between US- and CT-guided groups were not statistically significant. In the US-guided group, the 1-, 2-, and 3- year overall survival rates were 85, 74, and 68%, respectively, while they were 84, 72, and 58%in the CT-guided group. The differences were not statistically significant (P>0.05). No severe complications were found in the two groups. Both US- and CT-guided PRFA are safe and effective therapies for patients with HCC when surgical options are precluded. C1 [Wu, Jing] The Third Hospital Affiliated to Nantong University, Department of Ultrasound, 226006 Nantong, China. [Chen, Ping] The Third Hospital Affiliated to Nantong University, Department of Oncology, 226006 Nantong, China. [Xie, Yang-gui] The Hospital Affiliated to Nantong University, Department of Ultrasound, Xisi Road No. 20, 226006 Nantong, China. [Gong, Nian-mei] The Third Hospital Affiliated to Nantong University, Department of Ultrasound, 226006 Nantong, China. [Sun, Lin-lin] The Third Hospital Affiliated to Nantong University, Department of Ultrasound, 226006 Nantong, China. [Sun, Chun-feng] The Third Hospital Affiliated to Nantong University, Department of Medical Imaging, 226006 Nantong, China. RP Xie, Yg (reprint author), The Hospital Affiliated to Nantong University, Department of Ultrasound, 226006 Nantong, China. EM xieyanggui34@hotmail.com CR Llovet JM, Burroughs A, Bruix J, 2003, Hepatocellular carcinoma. Lancet 362(9399):1907–1917., DOI 10.1016/S0140-6736(03)14964-1 Shiina S, 2009, Image-guided percutaneous ablation therapies for hepatocellular carcinoma. J Gastroenterol 44(Suppl 19):122–131., DOI 10.1007/s00535-008-2263-9 Lencioni R, Crocetti L, 2013, Image-guided ablation for hepatocellular carcinoma. Recent Results Cancer Res Fortschr Der Krebsforschung Progres Dans Les Rech Sur Le Cancer 190:181– 194., DOI 10.1007/978-3-642-16037-0_12 Kim PN, Choi D, Rhim H, Rha SE, Hong HP, Lee J, Choi JI, Kim JW, Seo JW, Lee EJ, Lim HK, 2012, Planning ultrasound for percutaneous radiofrequency ablation to treat small, 0.05). C1 [Mate, Miklos] Saint Emerich Hospital Budapest, Surgical DepartmentBudapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. RP Mate, M (reprint author), Saint Emerich Hospital Budapest, Surgical Department, Budapest, Hungary. EM miklos.mate@gmail.com CR Croft J, Parry E, Jenkins GJ, Doak SH, Baxter JN, Griffiths AP, Brownn TH, Parry JM, 2002, Analysis of the premalignant stages of Barrett’s oesophagus through to adenocarcinoma by comparative genomic hybridization. Eur Gastroenterol Hepatology 14:1179–1186 Souza RF,Morales CP, Spechler SJ, 2001, A conceptual approach to understanding the molecular mechanism of cancer development in Barrett’s esophagus, Review, alimentary pharmacology. Therapy 15: 1087–1100 Spechler SJ, 2002, Barrett’s esophagus. N Engl J Med 346:836–842 Lagergren J, Bergstorm R, Lindgren A, Nyren O, 1999, Symptomatic gastroesophageal reflux as a risk factor for oesophageal adenocarcinoma. N Engl J Med 340:825–831 Moyes LH, Going JJ, 2011, Still waiting for predictive biomarkers in Barrett’s oesophagus. J Clin Pathol 64:742–750 Flejou JF, 2005, Barrett’s esophagus: from metaplasia to dysplasia and cancer. Gut 54(Suppl. 1):6–12 Ong C-AJ, Chin-Ann J, Fitzgerald RC, 2010, Biomarkers in Barrett’s esophagus and esophageal adenocarcinoma: predictors of progression and prognosis. World J Gastroenterol 16(45):5669–5681 Jengins GJ, DOak SH, Parry JM, D’Souza FR, Baxter JN, 2002, Genetic pathways involved int he progression of Barrett’s metaplasia to adenocarcinoma, Review). Br J Surg, 89):824–837 Scholzen T, Gerdes J, 2000, The Ki-67 protein: from the known and the unknown. J Cell Physiol 182:311–322 Sipos F, Zagoni T,Molnar B et al, 2002, Changes int he proliferation and apoptosis of colonis epithelial cells in correlation with histologic activity of ulcerative colitis. Orv Hetil 143:2485–2488 Geddert H, Zeriouh M, Wolter M, Heise JW, Gabbert HE, Sarbia M, 2002, Gene amplification and proteins overexpression of c-erb-b2 in Barrett’s carcinoma and its precursor laesions A. J Clin Pathol 117: 558–566 Egashira A, Morita M, Yoshida R, Saeki H, Oki E, Sadanaga N, Kakeji Y, Tsujitani S-I, Maehara Y, 2011, Loss of p53 in esophageal squamosus cell carcinoma and the correlation with survival: analyses of gene mutations, protein expression, and loss of heterozygosity in Japanese patients. J Surg Oncol 104:169–175 Ramel S, 2002, Barrett’s Esophagus: model of neoplastic progression. World J Surg, 73):1697–1703 Schneider PM, Stoeltzing O, Roth JA, Hoelscher AH, Wegerer S, Mizumoto S et al, 2000, p53 mutational status improves estimation fo prognosis in patients with curatively rsected adenocarcinoma in Barrett’s esophagus. Clin Cancer Res 6:3153–3158 Neuhaus H, Terheggen G, Rutz EM, Vieth M, Schumacher B, 2012, Endoscopic submucosal dissection plus radiofrequency ablation of neoplastic Barrett’s esophagus. Endocopy 44:1105–1113 Seery JP, 2002, Stem cells of the oesophageal epithelium. J Cell Sci 115:1783–1789 Sarosi G, Brown G, Jaiswal K, Feagins LA, Lee E, Crook TW, Souza RF, Zou YS, Shay JW, Spechler SJ, 2008, Bone marrow progenitor cells contribute to esophageal regeneration and metaplasia in a rat model of Barrett’s esophagus. Dis Esophagus 21:43–50 Arul GS, Moorghen M, Myerscough N, Alderson DA, Spicer RD, Corfield AP, 2000)Mucin gene expression in Barrett’s esophagus: an in situ hybridization and immunohistochemical study. Gut 47:753– 761 Burjonrappa SC, Reddimasu S, Nawaz Z, Gao X, Sharma P, Loggie B, 2007, Mucin expression profile in Barrett’s, dysplasia, adenocarcinoma sequence int he esophagus. Indian J Cancer Year 44(1):1–5 Rahn JJ, Shen Q, Mah BK, Hugh JC, 2004, MUC1 initiates a calcium signal after ligation by intercellular adhesion molecule-1. J Biol Chem 279:29386–29390 Sharma P, Sidorenko EI, 2005, Are screening and surveillance for Barrett’s esophagus really worthhile ? Gut, 54):27–32 Cameron AJ, 2000, Oesophageal cancer: epidemiological overview. J Roy Coll Surgery Edinborogh 45:259–260 Lagergren J, 2005, Adenocarcinoma of esophagus: what exactly is the size of the problem and who is at risk? Gut, 54)1–5 Galipeau PC, Li X, Blount PL, Maley CC, Sanchez CA, Odze RD, Ayub K, Rabinovitch PS, Vaughan TL, Reid BJ, 2007, NSAIDs modulate CDKN2A, TP53, and DNA content risk for progression to esophageal adenocarcinoma. PLoS Med 4:e67 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 669 EP 673 DI 10.1007/s12253-014-9873-8 PG 5 ER PT J AU Das, RB Bhaumik, S Firoz, A Mandsaurwala, A Satam, H AF Das, Ranjan Bibhu Bhaumik, Sangeet Firoz, Ahmad Mandsaurwala, Aziz Satam, Heena TI Molecular Spectrum of Somatic EGFR and KRAS Gene Mutations in non Small Cell Lung Carcinoma: Determination of Frequency, Distribution Pattern and Identification of Novel Variations in Indian Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR mutation; Lung cancer; Novel mutation; India ID EGFR mutation; Lung cancer; Novel mutation; India AB Somatic mutations of EGFR and KRAS gene represent the most common alterations currently known in NSCLC patients. This study explored the frequency, distribution pattern of EGFR and KRAS mutations in Indian patients. The frequencies of EGFR and KRAS mutations were 29 % (116/400) and 4.5 % (6/132) respectively. Both EGFR and KRAS mutations were prevalent in females, and a trend towards higher mutation frequency was seen in patients under≥ 60 years age. The presence of EGFR and KRAS mutations were higher in adenocarcinomas in comparison to other histological subtype. Sequencing analysis of EGFR exon 18 revealed Inframe deletion (G709_T710>A) and missense mutation (K713R). Among exon 19 positive cases, 49.3 % (37/75) were in-frame deletions, of which E746_A750del was frequent. Similarly, ~47 % (35/75) cases showed complex mutation involving indel. Among mutations in exon 20 (N= 9), 8 were substitutions, one showed duplication, while all exon 21 mutations were of the missense types with L858R as the most recurrent type. Sequencing analysis of KRAS exon 1 revealed three different types codon 12 substitutions resulting in c34G>T (G12C) (n=4), c.35G>A (G12D) (n=1), and c.35G>T (G12V) (n=1). In conclusion, the present study is an example of molecular diversity of EGFR and KRAS gene in Indian patients and further confirms that the frequency of EGFR and KRAS mutations varies considerably globally. To the best of our knowledge, this is the first Indian study to evaluate KRAS mutation. The current study also served to identify novel variations that added new insights into the genetic heterogeneity of NSCLC. C1 [Das, Ranjan Bibhu] SRL Ltd, Research and Development, Plot No.1, Prime square building S.V.Road, Goregaon (W), 400062 Mumbai, India. [Bhaumik, Sangeet] SRL Ltd, Research and Development, Plot No.1, Prime square building S.V.Road, Goregaon (W), 400062 Mumbai, India. [Firoz, Ahmad] SRL Ltd, Research and Development, Plot No.1, Prime square building S.V.Road, Goregaon (W), 400062 Mumbai, India. [Mandsaurwala, Aziz] SRL Ltd, Research and Development, Plot No.1, Prime square building S.V.Road, Goregaon (W), 400062 Mumbai, India. [Satam, Heena] SRL Ltd, Research and Development, Plot No.1, Prime square building S.V.Road, Goregaon (W), 400062 Mumbai, India. RP Das, RB (reprint author), SRL Ltd, Research and Development, 400062 Mumbai, India. EM brdas@srl.in CR Noronha V, Dikshit R, Raut N et al, 2012, Epidemiology of lung cancer in India: focus on the differences between non-smokers and smokers: a single-centre experience. Indian J Cancer 49:74–81 Ferlay J SH, Bray F, Forman D et al., 2010, GLOBOCAN 2008 v2.0, Cancer Incidence and Mortality Worldwide: IARC Cancer Base No. 10 [Internet]. Lyon, France: International Agency for Research on Cancer Available: http://globocan.iarc.fr Behera D, Balamugesh T, 2004, Lung cancer in India. Indian J Chest Dis Allied Sci 46:269–281 Behera D, Balamugesh T, Aggarwal AN et al, 2003, Docetaxel and cisplatin combination chemotherapy in advanced NSCL: A followup study from India. Lung Cancer 419(Suppl 2):S99 An X, Tiwari AK, Sun Y et al, 2010, BCR-ABL tyrosine kinase inhibitors in the treatment of Philadelphia chromosome positive chronic myeloid leukemia: a review. Leuk Res 10:1255–1268 Berghoff AS, Bago-Horvath Z, Dubsky P et al, 2013, Impact of her- 2-targeted therapy on overall survival in patients with her-2 positive metastatic breast cancer. Breast J 19:149–155 Ciardiello F, Tortora G, 2008, EGFR antagonists in cancer treatment. N Engl J Med 358:1160–1174 Lynch TJ, Bell DW, Sordella R et al, 2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350:2129– 2139 Shigematsu H, Lin L, Takahashi Tet al, 2005, Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancer. J Natl Cancer Inst 97:339–346 Shigematsu H, Gazdar AF, 2006, Somatic mutations of epidermal growth factor receptor signaling pathway in lung cancer. Int J Cancer 118:257–262 Rosell R,Moran T, Queralt C et al, 2009, Spanish lung cancer group. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med 361:958–967 Paez JG, Janne PA, Lee JC et al, 2004, EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science 304:1497–1500 Sharma SV, Bell DW, Settleman J et al, 2007, Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer 7:169–181 Han B, Zhou X, Zhang RX et al, 2011, Mutations of the epidermal growth factor receptor gene inNSCLC patients.Oncol Lett 2:1233–1237 Keedy VL, Temin S, SomerfieldMRet al, 2011)American society of clinical oncology provisional clinical opinion: epidermal growth factor receptor, EGFR, mutation testing for patients with advanced non-small-cell lung cancer considering first-line EGFR tyrosine kinase inhibitor therapy. J Clin Oncol 29:2121–2127 Forbes S, Clements J, Dawson E et al, 2006, COSMIC 2005. Br J Cancer 94:318–322 PaoW,Wang TY, Riely GJ et al, 2005, KRASmutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib. PLoS Med 2:e17 Eberhard DA, Johnson BE, Amler LC et al, 2005, Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol 23:5900–5909 Pesek M, Benesova L, Belsanova B et al, 2009)Dominance of EGFR and insignificant KRAS mutations in prediction of tyrosine-kinase therapy for NSCLC patients stratified by tumor subtype and smoking status. Anticancer Res 29:2767–2773 Socinski MA, 2010, The emerging role of biomarkers in advanced non-small-cell lung cancer. Clin Lung Cancer 11:149–159 Kato Y, Peled N, Wynes MWet al, 2010, Novel epidermal growth factor receptor mutation-specific antibodies for non-small cell lung cancer: immunohistochemistry as a possible screening method for epidermal growth factor receptor mutations. J Thorac Oncol 5:1551– 1558 Marchetti A, Martella C, Felicioni L et al, 2005, EGFR mutations in non-small-cell lung cancer: analysis of a large series of cases and development of a rapid and sensitivemethod for diagnostic screening with potential cations on pharmacologic treatment. J Clin Oncol 23: 857–865 Righi L, Cuccurullo A, Vatrano S et al, 2013, Detection and characterization of classical and 'uncommon" exon 19 Epidermal Growth Factor Receptor mutations in lung cancer by pyrosequencing. BMC Cancer 13:114 Pan ZK, Zhang L, Zhang X et al, 2005, Epidermal growth factor receptormutation in Chinese patientswith non-small cell lung cancer. Ai Zheng 24:919–923 Okami J, Taniguchi K, Higashiyama M et al, 2007, Prognostic factors for gefitinib-treated postoperative recurrence in non-small cell lung cancer. Oncology 72:234–242 Kim YT, Kim T, Lee DS et al, 2008)Molecular changes of epidermal growth factor receptor, EGFR, and KRAS and their impact on the clinical outcomes in surgically resected adenocarcinoma of the lung. Lung Cancer 59:111–118 Chougule A, Prabhash K, Noronha V et al, 2013, Frequency of EGFR Mutations in 907 Lung Adenocarcioma Patients of Indian Ethnicity. PLoS One., DOI 10.1371/journal.pone.0076164 Bhatt AD, Pai R, Rebekah G et al, 2013, Clinicopathologic features of non-small cell lung cancer in India and correlation with epidermal growth factor receptor mutational status. Indian J Cancer 50:94–101 Doval DC, Azam S, Batra U et al, 2013, Epidermal growth factor receptor mutation in lung adenocarcinoma in India: a single center study. J Carcinog 12:1477–3163 Noronha V, Prabhash K, Thavamani A et al, 2013, EGFR mutations in Indian lung cancer patients: clinical correlation and outcome to EGFR targeted therapy. PLoS One 8:e61561 Willmore–Payne C, Holden JA, Layfield LJ, 2006, Detection of EGFR- and HER2-activating mutations in squamous cell carcinoma involving the head and neck. Mod Pathol 9:634–640 Kalikaki A, Koutsopoulos A, Trypaki M et al, 2008, Comparison of EGFR and K-RAS gene status between primary tumours and corresponding metastases in NSCLC. Br J Cancer 99:923–929 American Cancer Society: Cancer Facts & Figures 2010. Atlanta: American Cancer Society. 2010 Liu Y, Liu B, Li XYet al, 2011, A comparison of ARMS and direct sequencing for EGFR mutation analysis and tyrosine kinase inhibitors treatment prediction in body fluid samples of Non-small-cell lung cancer patients. J Exp Clin Cancer Res 30:111 Eberhard DA, Johnson BE, Amler LC et al, 2005, Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol 23:5900–5909 Tsao AS, Tang XM, Sabloff B et al, 2006, Clinicopathologic characteristics of the EGFR genemutation in non-small cell lung cancer. J Thorac Oncol 6:231–239 Massarelli E, Varella-GarciaM, Tang X et al, 2007, KRAS mutation is an important predictor of resistance to therapy with epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. Clin Cancer Res 13:2890–2896 Zhu CQ, da Cunha SG, Ding K et al, 2008, Role of KRAS and EGFR as biomarkers of response to erlotinib in national cancer institute of canada clinical trials group study BR.21. J Clin Oncol 26:4268–4275 Schmid K, Oehl N, Wrba F et al, 2009, EGFR/KRAS/BRAF mutations in primary lung adenocarcinomas and corresponding locoregional lymph node metastases. Clin Cancer Res 15:4554–4560 Douillard JY, Shepherd FA, Hirsh Vet al, 2010, Molecular predictors of outcome with gefitinib and docetaxel in previously treated nonsmall- cell lung cancer: data from the randomized phase III INTEREST trial. J Clin Oncol 28:744–752 Bonanno L, Schiavon M, Nardo G et al, 2010, Prognostic and predictive implications of EGFR mutations, EGFR copy number and KRAS mutations in advanced stage lung adenocarcinoma. Anticancer Res 30:5121–5128 Santis G, Angell R, Nickless G et al, 2011, Screening for EGFR and KRAS mutations in endobronchial ultrasound derived transbronchial needle aspirates in non-small cell lung cancer using COLD-PCR. PLoS One., DOI 10.1371/journal.pone.0025191 Arrieta O, Cardona AF, Federico GB et al, 2011, Genotyping nonsmall cell lung cancer, NSCLC, in Latin America. J Thorac Oncol 6: 1955–1959 Billah S, Stewart J, Staerkel G et al, 2011, EGFR and KRAS mutations in lung. Carcinoma molecular testing by using cytology specimens. Cancer Cytopathol 19:111–117 Brugger W, Triller N, Blasinska-Morawiec M et al, 2011, Prospective molecular marker analyses of EGFR and KRAS from a randomized, placebo-controlled study of erlotinib maintenance therapy in advanced non-small-cell lung cancer. J Clin Oncol 29:4113– 4120 Cardarella S, Ortiz TM, Joshi VA et al, 2012, The introduction of systematic genomic testing for patients with non-small-cell lung cancer. J Thorac Oncol 7:1767–1774 Cadranel J,Mauguen A, FallerM(2012, Impact of systematic EGFR and KRAS mutation evaluation on progression-free survival and overall survival in patients with advanced non-small-cell lung cancer treated by erlotinib in a French prospective cohort, ERMETIC project–part 2. J Thorac Oncol 7:1490–1502 Cai G, Wong R, Chhieng D et al, 2013, Identification of EGFR mutation, KRAS mutation, and ALK gene rearrangement in cytological specimens of primary and metastatic lung /adenocarcinoma. Cancer Cytopathol 121:500–507 Fiala O, Pesek M, Finek J et al, 2013, Gene mutations in squamous cell NSCLC: insignificance of EGFR, KRAS and PIK3CA mutations in prediction of EGFR-TKI treatment efficacy. Anticancer Res 33: 1705–1711 Kerner GS, Schuuring E, Sietsma J et al, 2013, Common and rare EGFR and KRAS mutations in a Dutch non-small-cell lung cancer population and their clinical outcome. PloS one., DOI 10.1371/journal. pone.0070346 Carneiro JG, Couto PG, Bastos-Rodrigues L et al, 2014, Spectrum of somatic EGFR, KRAS, BRAF, PTEN mutations and TTF-1 expression in Brazilian lung cancer patients. Genet Res, Camb)., DOI 10. 1017/S0016672314000032 Kosaka T, Yatabe Y, Endoh H et al, 2004)Mutations of the epidermal growth factor receptor gene in lung cancer: biological and clinical implications. Cancer Res 64:8919–8923 Shigematsu H, Lin L, Takahashi Tet al, 2005, Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancer. J Natl Cancer Inst 97:339–346 Tam IY, Chung LP, Suen WS et al, 2006, Distinct epidermal growth factor receptor and KRAS mutation patterns in Non–small cell lung cancer patients with different tobacco exposure and clinicopathologic features. Clin Cancer Res 12:1647–1653 Bae NC, ChaeMH, LeeMH et al, 2007, EGFR, ERBB2, and KRAS mutations in Korean non-small cell lung cancer patients. Cancer Genet Cytogenet 173:107–113 Na II, Rho JK, Choi YJ et al, 2007, Clinical features reflect exon sites of EGFR mutations in patients with resected non-small-cell lung cancer. J Korean Med Sci 22:393–399 Wu CC, Hsu HY, Liu HP et al, 2008, Reversed mutation rates of KRAS and EGFR genes in adenocarcinoma of the lung in Taiwan and their implications. Cancer 113:3199–3208 Jang TW, Oak CH, Chang HK et al, 2009, EGFR and KRAS mutations in patients with adenocarcinoma of the lung. Korean J Intern Med 24:48–54 Yamaguchi F, Kugawa S, Tateno H et al, 2012, Analysis of EGFR, KRAS and P53 mutations in lung cancer using cells in the curette lavage fluid obtained by bronchoscopy. Lung Cancer 78:201–206 Kim HR, Shim HS, Chung JH et al, 2012, Distinct clinical features and outcomes in never-smokers with nonsmall cell lung cancer who harbor EGFR or KRAS mutations or ALK rearrangement. Cancer 118:729–39 Liu Y, Wu BQ, Zhong HH, 2013, Screening for EGFR and KRAS mutations in non-small cell lung carcinomas using DNA extraction by hydrothermal pressure coupled with PCR-based direct sequencing. Int J Clin Exp Pathol 6:1880–1889 Ohtaki Y, Shimizu K, Kakegawa S et al, 2014, Postrecurrence survival of surgically resected pulmonary adenocarcinoma patients according to EGFR and KRAS mutation status. Mol Clin Oncol 2:187–196 Serizawa M, Koh Y, Kenmotsu H et al, 2014, Assessment of mutational profile of Japanese lung adenocarcinoma patients by multitarget assays: A prospective, single-institute study. Cancer., DOI 10.1002/ cncr.28604 Sahoo R, Harini VV, Babu VC et al, 2011, Screening for EGFR mutations in lung cancer, a report from India. Lung Cancer 73:316–319 Mehta J, 2013, Molecular epidemiology of epidermal growth factor receptor mutations in lung cancers in Indian population. Indian J Cancer 50:102–106 Choughule A, Noronha V, Joshi A et al, 2013, Epidermal growth factor receptor mutation subtypes and geographical distribution among Indian non-small cell lung cancer patients. Indian J Cancer 50:107–111 Veldore VH, Rao RM, Kakara S et al, 2013, Epidermal growth factor receptor mutation in non-small-cell lung carcinomas: A retrospective analysis of 1036 lung cancer specimens from a network of tertiary cancer care centers in India. Indian J Cancer 50:87–93 Boch C, Kollmeier J, Roth A et al, 2013, The frequency of EGFR and KRASmutations in non-small cell lung cancer, NSCLC): routine screening data for central Europe from a cohort study. BMJ Open 3: e002560., DOI 10.1136/bmjopen-2013-002560 Sasaki H, Shimizu S, Endo K et al, 2006, EGFR and erbB2 mutation status in Japanese lung cancer patients. Int J Cancer 118:180–184 Shiozawa T, Ishii G, Goto K et al, 2013, Clinicopathological characteristics of EGFR mutated adenosquamous carcinoma of the lung. Pathol Int 63:77–84 Kang SM, Kang HJ, Shin JH et al, 2007, Identical epidermal growth factor receptor mutations in adenocarcinomatous and squamous cell carcinomatous components of adenosquamous carcinoma of the lung. Cancer 109:581–587 Network CC, 2011, NCCN clinical practice guidelines in oncology: Non-small cell lung cancer. Version 1, Available: http://www.nccn. org/professionals/physician_gls/pdf/nscl.pdf Choi YL, Sun JM, Cho J et al, 2013, EGFR mutation testing in patients with advanced non-small cell lung cancer: a comprehensive evaluation of real-world practice in an East Asian tertiary hospital. PLoS One., DOI 10.1371/journal.pone.0056011 Yin XW, Jiang XT, Yuan YT et al, 2010, Influence of mutations in epidermal growth factor receptor gene on growth, metastasis and survival rate of non-small cell lung carcinoma. Zhonghua Yi Xue Za Zhi 90:1808–12 Jang TW, Oak CH, Chang HK et al, 2009, EGFR and KRAS mutations in patients with adenocarcinoma of the lung. Korean J Intern Med 24:48–54 Feng Q, Li XH, Chen Z, He JS et al, 2011, Epidermal growth factor receptor gene mutations and clinicopathologic correlation in 309 patients with non-small cell lung cancer. Zhonghua Bing Li Xue Za Zhi 40:660–663 Hung MS, Lin CK, Leu SW et al, 2006, Epidermal growth factor receptor mutations in cells from non-small cell lung cancer malignant pleural effusions. Chang Gung Med J 29: 373–379 Sueoka N, Sato A, Eguchi H, Komiya K et al, 2007, Mutation profile of EGFR gene detected by denaturing high-performance liquid chromatography in Japanese lung cancer patients. J Cancer Res Clin Oncol 133:93–102 Sun MH, Yang F, Shen L et al, 2011, Detection of epidermal growth factor receptor mutations in non-small-cell lung carcinoma by direct sequencing and correlations with clinicopathological characteristics and sample types. Zhonghua Bing Li Xue Za Zhi 40:655–659 Asano H, Toyooka S, Tokumo M et al, 2006, Detection of EGFR gene mutation in lung cancer by mutant-enriched polymerase chain reaction assay. Clin Cancer Res 12:43–48 Sone T, Kasahara K, Kimura H et al, 2007, Comparative analysis of epidermal growth factor receptormutations and gene amplification as predictors of gefitinib efficacy in Japanese patients with nonsmall cell lung cancer. Cancer 109:1836–1844 Huncharek M, Muscat J, Geschwind JF, 1999, K-ras oncogene mutation as a prognostic marker in non-small cell lung cancer: a combined analysis of 881 cases. Carcinogenesis 20:1507–1510 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 675 EP 687 DI 10.1007/s12253-014-9874-7 PG 13 ER PT J AU Saadat, M Pashaei, S Amerizade, F AF Saadat, Mostafa Pashaei, Samira Amerizade, Foroozan TI Susceptibility to Gastric Cancer and Polymorphisms of Insertion/Deletion at the Intron 3 of the XRCC4 and VNTR at the Promoter Region of the XRCC5 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Polymorphism; Ins/Del; VNTR; XRCC4; XRCC5 ID Gastric cancer; Polymorphism; Ins/Del; VNTR; XRCC4; XRCC5 AB The genes encoding X-ray repair crosscomplementing group 4 (XRCC4; OMIM: 194363) and 5 (XRCC5; OMIM: 194364) are involved in repair of DNA double-strand breaks. To investigating the associations between polymorphisms of Insertion/Deletion (I/D, rs28360071) in the intron 3 of the XRCC4 and VNTR in the promoter region of the XRCC5 and risk of gastric cancer, the present study was carried out. We included 159 (56 females, 103 males) with gastric cancer and 242 (75 females, 167 males) healthy blood donors frequency matched for age and gender. Using PCR-basedmethods, the genotypes of the study polymorphisms were determined. The alleles of VNTR XRCC5 polymorphism divided into two groups: L (0 and 1 repeats) and H (2 and 3 repeats) alleles. For the I/D XRCC4 polymorphism, after stratification of the subjects according to their family history (FH) of cancer, either the ID (OR=3.19, 95%CI: 1.35–7.50, P=0.008) or the DD genotypes (OR= 4.62, 95%CI: 1.63–13.0, P=0.004) among positive FH persons, increased the risk of gastric cancer compared with the reference group (persons who have negative FH and II genotype). For the VNTR XRCC5 polymorphism, the LH+HH genotypes among positive FH persons, increased the risk of gastric cancer compared with the reference group (persons who have negative FH and LL genotype) (OR=2.88, 95%CI: 1.34–6.18, P=0.006). Sensitivity analysis showed that the above mentioned associations were not occurred due to the maldistribution of the genotypes among missing data. The present study suggests that both polymorphisms of the XRCC4 and XRCC5 might be risk factors for gastric cancer development especially among persons with positive FH. C1 [Saadat, Mostafa] Shiraz University, College of Sciences, Department of Biology, 71454 Shiraz, Iran. [Pashaei, Samira] Shiraz University, College of Sciences, Department of Biology, 71454 Shiraz, Iran. [Amerizade, Foroozan] Shiraz University, College of Sciences, Department of Biology, 71454 Shiraz, Iran. RP Saadat, M (reprint author), Shiraz University, College of Sciences, Department of Biology, 71454 Shiraz, Iran. EM saadat@shirazu.ac.ir;msaadat41@yahoo.com CR Gao S, Zhang X, Wang P, Dai L, Zhang J, Wang K, 2011, Genetic epidemiological analysis reveals a multi-gene additive model for gastric cancer. Fam Cancer 10:119–125 Draghicescu T, Roman IC, Paltanea L, 1998, Epidemiologic research on the genetic risk factors in gastric and colorectal cancer. Rom J Morphol Embryol 44:149–152 Gilley D, Tanaka H, Hande MP, Kurimasa A, Li GC, Oshimura M, Chen DJ, 2001, DNA-PKcs is critical for telomere capping. Proc Natl Acad Sci U S A 98:15084–15088 Difilippantonio MJ, Zhu J, Chen HT, Meffre E, Nussenzweig MC, Max EE et al, 2000, DNA repair protein Ku80 suppresses chromosomal aberrations and malignant transformation. Nature 404:510–514 Ferguson DO, Alt FW, 2001, DNA double strand break repair and chromosomal translocation: lessons from animal models. Oncogene 20:5572–5579 Gao Y, Ferguson DO, XieW, Manis JP, Sekiguchi J, Frank KM et al, 2000, Interplay of p53 and DNA-repair protein XRCC4 in tumorigenesis, genomic stability and development. Nature 404:897–900 Zhu C,Mills KD, Ferguson DO, Lee C,Manis J, Fleming J et al, 2002, Unrepaired DNA breaks in p53-deficient cells lead to oncogenic gene amplification subsequent to translocations. Cell 109:811–821 Pfeiffer P, Goedecke W, Kuhfittig-Kulle S, Obe G, 2004, Pathways of DNA double-strand break repair and their impact on the prevention and formation of chromosomal aberrations. Cytogenet Genome Res 104:7–13 Kuschel B, Auranen A, McBride S, Novik KL, Antoniou A, Lipscombe JM et al, 2002, Variants in DNA double-strand break repair genes and breast cancer susceptibility. Hum Mol Genet 11: 1399–1407 Li Z, Otevrel T, Gao Y, Cheng HL, Seed B, Stamato TD et al, 1995, The XRCC4 gene encodes a novel protein involved in DNA doublestrand break repair and V(D)J recombination. Cell 83:1079–1089 Gao Y, Sun Y, FrankKM, Dikkes P, Fujiwara Y, Seidl KJ et al, 1998, A critical role for DNA end-joining proteins in both lymphogenesis and neurogenesis. Cell 95:891–902 Chang CH, Chiu CF, Wu HC, Tseng HC, Wang CH, Lin CC et al, 2008, Significant association of XRCC4 single nucleotide polymorphisms with prostate cancer susceptibility in Taiwanese males. Mol Med Rep 1:525–530 Mandal RK, Singh V, Kapoor R, Mittal RD, 2011, Do polymorphisms in XRCC4 influence prostate cancer susceptibility in North Indian population? Biomarkers 16:236–242 Chiu CF, Tsai MH, Tseng HC, Wang CL, Wang CH, Wu CN et al, 2008, A novel single nucleotide polymorphism in XRCC4 gene is associated with oral cancer susceptibility in Taiwanese patients. Oral Oncol 44:898–902 Wu KH,Wang CH, Yang YL, Peng CT, LinWD, Tsai FJ et al, 2010, Significant association of XRCC4 single nucleotide polymorphisms with childhood leukemia in Taiwan. Anticancer Res 30:529–533 RajaeiM, Saadat I, SaadatM(2012, Introducing a novel allele for the polymorphism of variable number of tandem repeats in the promoter region of XRCC5. Biochem Biophys Res Commun 427:503–505 Rajaei M, Saadat I, Saadat M, 2013, The novel allele, 3R, of the VNTR polymorphism in the XRCC5 promoter region dramatically decreases the gene expression. Biochem Biophys Res Commun 430: 640–641 Lim JW, Kim H, Kim KH, 2002, Expression of Ku70 and Ku80 mediated by NF-kappa B and cyclooxygenase-2 is related to proliferation of human gastric cancer cells. J Biol Chem 277:46093–46100 Hosoi Y, Watanabe T, Nakagawa K, Matsumoto Y, Enomoto A, Morita A et al, 2004, Up-regulation of DNA-dependent protein kinase activity and Sp1 in colorectal cancer. Int J Oncol 25:461–468 Chang HW, Kim SY, Yi SL, Son SH, Song do Y, Moon SY et al, 2006, Expression of Ku80 correlates with sensitivities to radiation in cancer cell lines of the head and neck. Oral Oncol 42:979–986 Gorre M,Mohandas PE, Kagita S, Annamaneni S, Digumarti R, Satti V, 2014, Association of XRCC5 VNTR polymorphism with the development of chronic myeloid leukemia. Tumour Biol 35:923–927 Wang S, Wang M, Yin S, Fu G, Li C, Chen R et al, 2008, A novel variable number of tandem repeats, VNTR, polymorphism containing Sp1 binding elements in the promoter of XRCC5 is a risk factor for human bladder cancer. Mutat Res 638:26–36 Dong XJ, Wang N, Guo W, Zhou RM, Zhang XJ, Li Y, 2007, Correlations of XRCC5 polymorphisms to genetic susceptibility to esophageal squamous cell carcinoma and gastric cardiac adenocarcinoma in a high incidence region. Ai Zhengzhou 26:280–284 [Chinese] Li R, Yang Y, An Y, Zhou Y, Liu Y, Yu Q et al, 2011, Genetic polymorphisms in DNA double-strand break repair genes XRCC5, XRCC6 and susceptibility to hepatocellular carcinoma. Carcinogenesis 32:530–536 Yang MD, Tsai CW, Chang WS, Tsou YA, Wu CN, Bau DT, 2011, Predictive role of XRCC5/XRCC6 genotypes in digestive system cancers. World J Gastrointest Oncol 3:175–181 Rajaei M, Saadat I, Omidvari S, Saadat M, 2014, Association between polymorphisms at promoters of XRCC5 and XRCC6 genes and risk of breast cancer. Med Oncol 31:885 Rafiee L, Saadat I, SaadatM(2010, Glutathione S-transferase genetic polymorphisms, GSTM1, GSTT1 and GSTO2, in three Iranian populations. Mol Biol Rep 37:155–158 Mohamadynejad P, Saadat M, 2008, Genetic polymorphisms of XRCC1, at codons 194 and 399, in Shiraz population, Fars province, southern Iran). Mol Biol Rep 35:669–672 RajaeiM, Saadat I, SaadatM(2012, High resolution melting analysis for detection of variable number of tandem repeats polymorphism of XRCC5. Biochem Biophys Res Commun 425:398–400 Liu J, Shu T, Chang S, Sun P, Zhu H, LiH, 2014, Risk ofmalignancy associated with a maternal family history of cancer. Asian Pac J Cancer Prev 15:2039–2044 Jiang X, Tseng CC, Bernstein L, Wu AH, 2014, Family history of cancer and gastroesophageal disorders and risk of esophageal and gastric adenocarcinomas: a case-control study. BMC Cancer 14:60 Turati F, Negri E, La Vecchia C, 2014, Family history and the risk of cancer: genetic factors influencing multiple cancer sites. Expert Rev Anticancer Ther 14:1–4 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 689 EP 693 DI 10.1007/s12253-014-9875-6 PG 5 ER PT J AU Wu, H Sun, Y Ye, H Yang, Sh Lee, LS de las Morenas, A AF Wu, Hao Sun, Yue Ye, Huihui Yang, Shi Lee, L Stephanie de las Morenas, Antonio TI Anaplastic Thyroid Cancer: Outcome and the Mutation/Expression Profiles of Potential Targets SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Anaplastic thyroid cancer; Targeted therapy; PDL1; BRAF; c-KIT ID Anaplastic thyroid cancer; Targeted therapy; PDL1; BRAF; c-KIT AB Anaplastic thyroid cancer (ATC) is a rare but aggressive malignancy of the thyroid. No effective treatment modalities are currently available. Targeted therapy against protein kinases showed promising results in preclinical studies. Our goal was to assess the mutational status of potential therapeutic targets, as well as the biomarker for immunotherapy in the clinical context. Using allele specific PCR, Sanger sequencing, fragment analysis and immunohistochemistry, we assessed BRAF, KRAS, EGFR mutations and protein overexpression of C-KIT and PDL1 in anaplastic thyroid cancer specimens. Results were compared to clinical information and patient outcome to assess the utility of these biomarkers. There were 13 patients in our study with a median overall survival of 19 weeks. Of the 13 ATC patients, 3 (23 %) had BRAF V600E mutation. C-KIT overexpression was found in 1 (8 %) patient who responded well to a tyrosine kinase inhibitor. PDL1 expression was seen in 3 (23 %) patients, none of them were surgical candidates due to unresectability and poor performance status. KRAS codon 12/13 and EGFR exon 18, 19, 20 and 21 were all wild type in our patients. Protein kinase inhibitors and immunotherapy may be useful adjuvant therapies for ATC. C1 [Wu, Hao] Baylor College of Medicine, Texas Children’s Hospital, Department of Pathology, AB190.11, 6621 Fannin Street, 77030 Houston, TX, USA. [Sun, Yue] Boston University Medical Center, Department of Pathology and Laboratory Medicine, 02118 Boston, MA, USA. [Ye, Huihui] Harvard Medical School, Beth Israel Deaconess Medical Center, Department of Pathology and Laboratory Medicine, 02115 Boston, MA, USA. [Yang, Shi] Boston University Medical Center, Department of Pathology and Laboratory Medicine, 02118 Boston, MA, USA. [Lee, L Stephanie] Boston University Medical Center, Department of Medicine, 02118 Boston, MA, USA. [de las Morenas, Antonio] Boston University Medical Center, Department of Pathology and Laboratory Medicine, 02118 Boston, MA, USA. RP Wu, H (reprint author), Baylor College of Medicine, Texas Children’s Hospital, Department of Pathology, 77030 Houston, USA. EM hao.wu@bcm.edu CR Carling T, Udelsma R, 2008, Thyroid tumors. In: DeVita VT, Lawrence TS, Rosenberg SA, eds, Devita, Hellman & Rosenberg’s cancer [electronic resource]: principles and practice of oncology. Wolters Kluwer/Lippincott Williams & Wilkins, Philadelphia, pp 1663–1682 Edge SB, Byrd DR, Compton CC et al., eds,, 2010, AJCC cancer staging handbook. 7th ed. New York, NY: Springer New York Sugitani I, Miyauchi A, Sugino K et al, 2012, Prognostic factors and treatment outcomes for anaplastic thyroid carcinoma: ATC research consortium of Japan cohort Study of 677 patients. World J Surg 36: 1247–1254 Kebebew E, Greenspan FS, Clark OH et al, 2005, Anaplastic thyroid carcinoma. Treatment outcome and prognostic factors. Cancer 103: 1330–1335 Busnardo B, Daniele O, Pelizzo MR et al, 2000, A multimodality therapeutic approach in anaplastic thyroid carcinoma: study on 39 patients. J Endocrinol Investig 23:755–761 Pierie J-PEN, Muzikansky A, Gaz RD et al, 2002, The effect of surgery and radiotherapy on outcome of anaplastic thyroid carcinoma. Ann Surg Oncol 9:57–64 Haigh PI, Ituarte PH, Wu HS et al, 2001, Completely resected anaplastic thyroid carcinoma combined with adjuvant chemotherapy and irradiation is associated with prolonged survival. Cancer 91: 2335–2342 Smallridge RC, 2012, Approach to the patient with anaplastic thyroid carcinoma. J Clin Endocrinol Metab 97:2566–2572 Akaishi J, Sugino K, KitagawaWet al, 2011, Prognostic factors and treatment outcomes of 100 cases of anaplastic thyroid carcinoma. Thyroid : Off J Am Thyroid Assoc 21:1183–1189 Nehs MA, Nagarkatti S, Nucera C et al, 2010, Thyroidectomy with neoadjuvant PLX4720 extends survival and decreases tumor burden in an orthotopic mouse model of anaplastic thyroid cancer. Surgery 148:1154–1162, discussion 1162 GuleMK, Chen Y, Sano D et al, 2011, Targeted therapy of VEGFR2 and EGFR significantly inhibits growth of anaplastic thyroid cancer in an orthotopic murine model. Clin Cancer Res 17:2281–2291 Nobuhara Y, Onoda N, Yamashita Yet al, 2005, Efficacy of epidermal growth factor receptor-targeted molecular therapy in anaplastic thyroid cancer cell lines. Br J Cancer 92:1110–1116 Kondo T, Ezzat S, Asa SL, 2006, Pathogenetic mechanisms in thyroid follicular-cell neoplasia. Nat Rev Cancer 6:292–306 Sipos JA, Shah MH, 2010, Thyroid cancer: emerging role for targeted therapies. Ther Adv Med Oncol 2:3–16 Antonelli A, Fallahi P, Ferrari SM et al, 2012, RET TKI: potential role in thyroid cancers. Curr Oncol Rep 14:97–104 Keir ME, Butte MJ, Freeman GJ et al, 2008, PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 26:677–704 Brahmer JR, Tykodi SS, Chow LQMet al, 2012, Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366:2455–2465 Topalian SL, Hodi FS, Brahmer JR et al, 2012, Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366:1–12 Hong DS, Kurzrock R, Supko JG et al, 2012, A phase I first-inhuman trial of bardoxolone methyl in patients with advanced solid tumors and lymphomas. Clin Cancer Res 18:3396–3406 Yang S, Farraye FA, Mack C et al, 2004, BRAF and KRAS Mutations in hyperplastic polyps and serrated adenomas of the colorectum: relationship to histology and CpG island methylation status. Am J Surg Pathol 28:1452–1459 Davies H, Bignell GR, Cox C et al, 2002, Mutations of the BRAF gene in human cancer. Nature 417:949–954 Pollock PM, Harper UL, Hansen KS et al, 2003, High frequency of BRAF mutations in nevi. Nat Genet 33:19–20 Amado RG, Wolf M, Peeters M et al., 2008, Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer Khambata-Ford S, Garrett CR,MeropolNJ et al, 2007, Expression of epiregulin and amphiregulin and K-ras mutation status predict disease control in metastatic colorectal cancer patients treated with cetuximab. J Clin Oncol 25:3230–3237 Pan Q, Pao W, Ladanyi M, 2005, Rapid polymerase chain reactionbased detection of epidermal growth factor receptor gene mutations in lung adenocarcinomas. J Mol Diagn JMD 7:396–403 Pinter F, Papay J, Almasi A et al, 2008, Epidermal growth factor receptor, EGFR, high gene copy number and activating mutations in lung adenocarcinomas are not consistently accompanied by positivity for EGFR protein by standard immunohistochemistry. J Mol Diagn JMD 10:160–168 Oh Y-H, Kim Y, Kim Y-P et al, 2010, Rapid detection of the epidermal growth factor receptor mutation in non-small-cell lung cancer for analysis of acquired resistance using molecular beacons. J Mol Diagn JMD 12:644–652 Gupta-Abramson V, Troxel AB, Nellore A et al, 2008, Phase II trial of sorafenib in advanced thyroid cancer. J Clin Oncol 26:4714–4719 Kloos RT, Ringel MD, Knopp MV et al, 2009, Phase II trial of Sorafenib in metastatic thyroid cancer. J Clin Oncol 27:1675–1684 Pennell NA, Daniels GH, Haddad RI et al., 2008, A phase II study of gefitinib in patients with advanced thyroid cancer Elliott DD, Sherman SI, Busaidy NL et al, 2008, Growth factor receptors expression in anaplastic thyroid carcinoma: potential markers for therapeutic stratification. Hum Pathol 39:15–20 Lee DH, Lee GK, Kong S et al, 2007, Epidermal growth factor receptor status in anaplastic thyroid carcinoma. J Clin Pathol 60: 881–884 Fisher KE, Jani JC, Fisher SB et al, 2013, Epidermal growth factor receptor overexpression is a marker for adverse pathologic features in papillary thyroid carcinoma. J Surg Res., DOI 10.1016/j.jss.2013.05. 003 Schoenfeld JD, Odejide OO, Wirth LJ et al, 2012, Survival of a patient with anaplastic thyroid cancer following intensity-modulated radiotherapy and sunitinib—a case report. Anticancer Res 32:1743– 1746 Bible KC, Suman VJ, Molina JR et al., 2010, Efficacy of pazopanib in progressive, radioiodine-refractory, metastatic differentiated thyroid cancers: results of a phase 2 consortium study Bible KC, Suman VJ, Menefee ME et al, 2012, A multiinstitutional phase 2 trial of pazopanib monotherapy in advanced anaplastic thyroid cancer. J Clin Endocrinol Metab 97:3179–3184 Ha HT, Lee JS, Urba S et al, 2010, A phase II study of imatinib in patients with advanced anaplastic thyroid cancer. Thyroid Off J Am Thyroid Assoc 20:975–980 Kimura H, Nakajima T, Takeuchi K et al, 2012, ALK fusion gene positive lung cancer and 3 cases treated with an inhibitor for ALK kinase activity. Lung Cancer 75:66–72 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 695 EP 701 DI 10.1007/s12253-014-9876-5 PG 7 ER PT J AU Nowakowski, A de Souza, CS Jach, R Rosillon, D Ksiazek, A Holl, K AF Nowakowski, Andrzej de Souza, Collas Sabrina Jach, Robert Rosillon, Dominique Ksiazek, Alicja Holl, Katsiaryna TI HPV-Type Distribution and Reproducibility of Histological Diagnosis in Cervical Neoplasia in Poland SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; CIN; Diagnosis reproducibility; HPV; Poland ID Cervical cancer; CIN; Diagnosis reproducibility; HPV; Poland AB This study was performed to assess attribution of high grade cervical intraepithelial neoplasia (HG-CIN) and invasive cervical cancer (ICC) to human papillomavirus (HPV) genotypes and secondarily to assess reproducibility of HG-CIN/ICC diagnosis obtained in Poland. Formaldehyde fixed, paraffin embedded blocks of HG-CIN/ICC from two distant institutions were sent to a central laboratory together with original histological diagnoses. Central/ expert review of histopathological specimens was performed and agreement between local and central/expert diagnoses was calculated. HPV detection and genotyping in the samples was carried out with the use of SPF10- LiPA25 technology. Results were analyzed for 205 HGCIN and 193 ICC cases with centrally confirmed diagnoses. Kappa coefficients and 95 % confidence intervals for HG-CIN and ICC diagnoses were: 0.13 (0.09;0.17) and 0.19 (0.11;0.26) respectively. Cohen’s kappa coefficients for lesions with representative number of samples ranged from 0.01 for cervical intraepithelial neoplasia grade 2 to 0.75 for adenocarcinoma. HPV DNA was detected in 96.1 and 91.2 % of the confirmed HG-CIN and ICC specimens respectively. HPV positive HG-CIN was most commonly attributed to HPV types: 16 (62.8), 33 (7.8), 31 (6.6), 52 (3.7), 45 (2.6) and 58 (2.6 %). HPV positive ICC was most commonly attributed to HPV types: 16 (72.1), 18 (10.8), 33 (5.7), 45 (3.4) and 31 (1.7 %). Reproducibility of histological diagnosis of HG-CIN/ICC obtained in Poland generally increases with the severity of lesion and is lowest for cervical intraepithelial neoplasia grade 2 and highest for adenocarcinoma. Over 80 % of ICC cases are vaccinepreventable in Poland. C1 [Nowakowski, Andrzej] Medical University of Lublin, Department of Oncologic Gynecology and Gynecology, ul. Staszica 16, 20-081 Lublin, Poland. [de Souza, Collas Sabrina] 4Clinics, 8 rue de la Terrasse, F-75017 Paris, France. [Jach, Robert] Krakow University Hos, Medical College of Jagiellonian University, Department of Obstetrics and Perinatology, ul. Kopernika 23, 31-501 Krakow, Poland. [Rosillon, Dominique] GlaxoSmithKline Vaccines, Rue Fleming 20, 1300 Wavre, Belgium. [Ksiazek, Alicja] GlaxoSmithKline, ul. Rzymowskiego 53, 02-697 Warsaw, Poland. [Holl, Katsiaryna] GlaxoSmithKline Vaccines, Rue Fleming 20, 1300 Wavre, Belgium. RP Nowakowski, A (reprint author), Medical University of Lublin, Department of Oncologic Gynecology and Gynecology, 20-081 Lublin, Poland. EM andrzejmnowakowski@poczta.onet.pl CR Spaczynski M, Karowicz-Bilinska A, Rokita W et al, 2010, Attendance rate in the polish cervical cancer screening program in the years 2007–2009. Ginekol Pol 81:655–663 IARC. Globocan 2008, Section of Cancer Information. Available at: http://globocan.iarc.fr/factsheets/cancers/cervix.asp. Accessed on December 11, 2012. Nowakowski A, Jackowska T, Oszukowski P, Radowicki S,Wysocki J, Zatonski W, 2013, Prevention of cervical cancer – an interdisciplinary problem. Can we improve the situation in Poland and how? Pediatr Pol 88:340–346 The communicate of Chief Sanitary Inspector for Poland on the Programme of Immunization for the year 2013. Available at: http:// dziennikmz.mz.gov.pl/DUM_MZ/2012/78/akt.pdfa Kedzia W, Pruski D, Jozefiak A, Rokita W, Spaczynski M, 2010, Genotyping of oncogenic human papilloma viruses in women with HG SIL diagnosis. Ginekol Pol 81:740–744 Bardin A, Vaccarella S, Clifford GM et al, 2008, Human papillomavirus infection inwomen with and without cervical cancer inWarsaw, Poland. Eur J Cancer 44:557–564 Liss J, Lukaszuk K, Gulczynski J et al, 2002, The incidence of human papilloma virus, HPV, DNA in patients with cervical carcinoma from Gdansk region. Ginekol Pol 73:740–744 Tjalma WA, Fiander A, Reich O et al, 2013, Differences in human papillomavirus type distribution in high-grade cervical intraepithelial neoplasia and invasive cervical cancer in Europe. Int J Cancer 132: 854–867 Tavassoli FA, Devilee P, 2003)World health organization classification of tumours. Pathology and genetics of tumours of the breast and female genital organs. IARC Press, Lyon Insinga RP, Liaw KL, Johnson LG, Madeleine MM, 2008, A systematic review of the prevalence and attribution of human papillomavirus types among cervical, vaginal, and vulvar precancers and cancers in the United States. Cancer Epidemiol Biomarkers Prev 17: 1611–1622 Wentzensen N, Schiffman M, Dunn T et al, 2009, Multiple human papillomavirus genotype infections in cervical cancer progression in the study to understand cervical cancer early endpoints and determinants. Int J Cancer 125:2151–2158 de Sanjose S, QuintWG, Alemany L et al, 2010, Human papillomavirus genotype attribution in invasive cervical cancer: a retrospective cross-sectional worldwide study. Lancet Oncol 11:1048–1056 Saslow D, Solomon D, Lawson HW et al, 2012, ACS-ASCCPASCP cervical cancer guideline committee. American cancer society, american society for colposcopy and cervical pathology, and american society for clinical pathology screening guidelines for the prevention and early detection of cervical cancer. CA Cancer J Clin 62: 147–172 Arbyn M, Kyrgiou M, Simoens C et al, 2008, Perinatal mortality and other severe adverse pregnancy outcomes associated with treatment of cervical intraepithelial neoplasia: meta-analysis. BMJ 337:a1284 Dalla Palma P, Giorgi Rossi P, Collina G et al, 2009, The reproducibility of CIN diagnoses among different pathologists: data from histology reviews from a multicenter randomized study. Am J Clin Pathol 132:125–132 Castle PE, Stoler MH, Solomon D, Schiffman M, 2007, The relationship of community iopsy-diagnosed cervical intraepithelial neoplasia grade 2 to the quality control pathology-reviewed diagnoses: an ALTS report. Am J Clin Pathol 127:805–815 Kato I, SantamariaM,De Ruiz PAet al, 1995, Inter-observer variation in cytological and histological diagnoses of cervical neoplasia and its epidemiologic implication. J Clin Epidemiol 48:1167–1174 ICD10 WHO webpage available at: http://apps.who.int/classifications/ icd10/browse/2010/en#/N80-N98. Accessed on 1st May 2013. Data from Polish National Cancer Registry webpage available at: http://epid.coi.waw.pl/krn/. Accessed on 1 May 2013. Klaes R, Benner A, Friedrich T et al, 2002, p16INK4a immunohistochemistry improves interobserver agreement in the diagnosis of cervical intraepithelial neoplasia. Am J Surg Pathol 26:1389–1399 Pinto AP, Schlecht NF, Woo TY, Crum CP, Cibas ES, 2008, Biomarker, ProEx C, p16, INK4A), and MiB-1, distinction of high-grade squamous intraepithelial lesion from its mimics. Mod Pathol 21:1067–1074 Dijkstra MG, Heideman DA, de Roy SC et al, 2010, p16, INK4a, immunostaining as an alternative to histology review for reliable grading of cervical intraepithelial lesions. J Clin Pathol 63:972– 977 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 703 EP 711 DI 10.1007/s12253-014-9877-4 PG 9 ER PT J AU Oh, RH An, HCh Yoo, JN Lee, HS AF Oh, Rim Hye An, Hyeok Chang Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutations of MUC15 Gene in Gastric and its Regional Heterogeneity in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MUC15; MUC7; Heterogeneity; Mutation; Cancer; Microsatellite instability ID MUC15; MUC7; Heterogeneity; Mutation; Cancer; Microsatellite instability AB Mucins are important in tumorigenesis and expressional alterations of mucins are common in human cancers. A membrane-bound mucin MUC15 and secreted mucins MUC4 and MUC7 are known to involve in tumorigenesis, but their mutation status in cancers remains unknown. Aim of this study was to explore whether MUC4, MUC7 and MUC15 genes are mutated and expressionally altered in gastric (GC) and colorectal cancers (CRC). In a public database, we found that MUC15 and MUC7 genes had mononucleotide repeats in the coding sequences that might be mutation targets in the cancers with microsatellite instability (MSI). We analyzed the mutations in 90 GC and 141 CRC (high MSI (MSI-H) or stable MSI/low MSI (MSS/MSI-L)) by single-strand conformation polymorphism analysis and DNA sequencing. In the present study, we found MUC15 frameshift mutations (14.7% of GC and 15.2% of CRC with MSI-H), MUC 7 frameshift mutations (2.9% of GC with MSI-H) and MUC4 frameshift mutations (8.8% of GC and 3.8% of CRC with MSI-H). These mutations were not found in in MSS/MSI-L (0/118). Additionally, we analyzed intratumoral heterogeneity (ITH) of MUC15 mutation in 16 CRC and found that seven CRC (43.8%) harbored regional ITH of MUC15. We also analyzed MUC15 expression in GC and CRC by immunohistochemistry. Negative MUC15 expression was identified in 15–41% of the GC and CRC irrespective of MSI status. Of note, the negative expression was more common in those with MUC15 mutations.We identified alterations of MUC genes at various levels (frameshift mutations, genetic ITH and expression loss), which together might play a role in tumorigenesis of GC and CRC with MSI-H. Our data suggest that mutation analysis in multiple regions is needed for a better evaluation of mutation status in CRC with MSI-H. C1 [Oh, Rim Hye] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [An, Hyeok Chang] The Catholic University of Korea, College of Medicine, Department of General Surgery, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Kufe DW, 2009, Mucins in cancer: function, prognosis and therapy. Nat Rev Cancer 9:874–885 Rachagani S, TorresMP,Moniaux N, Batra SK, 2009, Current status of mucins in the diagnosis and therapy of cancer. Biofactors 35:509– 527 Bafna S, Kaur S, Batra SK, 2010, Membrane-bound mucins: the mechanistic basis for alterations in the growth and survival of cancer cells. Oncogene 29:2893–2894 Irimura T, Denda K, Si I, Takeuchi H, Kato K, 1999, Themembranebound mucins: From cell signalling to transcriptional regulation and expression in epithelial cancers. J Biochem 126:975–985 Byrd JC, Bresalier RS, 2004, Mucins and mucin binding proteins in colorectal cancer. Cancer Metastasis Rev 23:77–99 Van Klinken BJ, Van derWal JW, Einerhand AW, Buller HA, Dekker J, 1999, Sulphation and secretion of the predominant secretory human colonic mucin MUC2 in ulcerative colitis. Gut 44:387–393 Ogata S, Uehara H, Chen A, Itzkowitz SH, 1992, Mucin gene expression in colonic tissues and cell lines. Cancer Res 52:5971–5978 Williams SJ, McGuckin MA, Gotley DC, Eyre HJ, Sutherland GR, Antalis TM, 1999, Two novel mucin genes down-regulated in colorectal cancer identified by differential display. Cancer Res 59:4083– 4089 Williams SJ, Wreschner DH, Tran M, Eyre HJ, Sutherland GR, McGuckin MA, 2001, Muc13, a novel human cell surface mucin expressed by epithelial and hemopoietic cells. J Biol Chem 276: 18327–18336 Nakamori S, Ota DM, Cleary KR, Shirotani K, Irimura T, 1994, MUC1 mucin expression as a marker of progression and metastasis of human colorectal carcinoma. Gastroenterology 106:353–361 Pallesen LT, Berglund L, Rasmussen LK, Petersen TE, Rasmussen JT, 2002, Isolation and characterization of MUC15, a novel cell membrane-associated mucin. Eur J Biochem 269:2755–2763 Shyu MK, Lin MC, Shih JC, Lee CN, Huang J, Liao CH, Huang IF, Chen HY, Huang MC, Hsieh FJ, 2007, Mucin 15 is expressed in human placenta and suppresses invasion of trophoblast-like cells in vitro. Hum Reprod 22:2723–2732 Huang J, Che MI, Huang YT, Shyu MK, Huang YM, Wu YM, Lin WC, Huang PH, Liang JT, Lee PH, Huang MC, 2009, Overexpression of MUC15 activates extracellular signal-regulated kinase 1/2 and promotes the oncogenic potential of human colon cancer cells. Carcinogenesis 30:1452–1458 Bobek LA, Tsai H, Biesbrock AR, Levine MJ, 1993, Molecular cloning, sequence, and specificity of expression of the gene encoding the low molecular weight human salivary mucin, MUC7). J Biol Chem 268:20563–20569 Sharma P, Dudus L, Nielsen PA, Clausen H, Yankaskas JR, Hollingsworth MA, Engelhardt JF, 1998, MUC5B and MUC7 are differentially expressed in mucous and serous cells of submucosal glands in human bronchial airways. Am J Respir Cell Mol Biol 19: 30–37 Retz M, Lehmann J, Roder C, Plotz B, Harder J, Eggers J, Pauluschke J, Kalthoff H, Stockle M, 1998, Differential mucin MUC7 gene expression in invasive bladder carcinoma in contrast to uniform MUC1 and MUC2 gene expression in both normal urothelium and bladder carcinoma. Cancer Res 58:5662–5666 Retz M, Lehmann J, Szysnik C, Zwank S, Venzke T, Roder C, Kalthoff H, Basbaum C, StockleM(2004, Detection of occult tumor cells in lymph nodes from bladder cancer patients by MUC7 nested RT-PCR. Eur Urol 45:314–319 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8:305–311 Lee JW, Soung YH, Kim SY, Lee HW, ParkWS, Nam SW, Kim SH, Lee JY, Yoo NJ, Lee SH, 2005, PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas. Oncogene 24: 1477–1480 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Kim MS, Hur SY, Yoo NJ, Lee SH, 2010, Mutational analysis of FOXL2 codon 134 in granulosa cell tumour of ovary and other human cancers. J Pathol 221:147–152 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 Wang RY, Chen L, Chen HY, Hu L, Li L, Sun HY, Jiang F, Zhao J, Liu GM, Tang J, Chen CY, Yang YC, Chang YX, Liu H, Zhang J, Yang Y, Huang G, Shen F, Wu MC, Zhou WP, Wang HY, 2013, MUC15 inhibits dimerization of EGFR and PI3K-AKTsignaling and is associated with aggressive hepatocellular carcinomas in patients. Gastroenterology 145:1436–1448 Pino MS, Kikuchi H, Zeng M, Herraiz MT, Sperduti I, Berger D, Park DY, Iafrate AJ, Zukerberg LR, Chung DC, 2010, Epithelial to mesenchymal transition is impaired in colon cancer cells with microsatellite instability. Gastroenterology 138:1406–1417 Calin GA, Gafa R, TibilettiMG, Herlea V, Becheanu G, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression in microsatellite instability high, MSI-H, colon cancers correlates with clinico-pathological parameters: a study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89:230–235 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 713 EP 718 DI 10.1007/s12253-014-9878-3 PG 6 ER PT J AU Chen, G Li, H Niu, X Li, G Han, N Li, X Li, G Liu, Y Sun, G Wang, Y Li, Z Li, Q AF Chen, Guoting Li, Hengping Niu, Xianping Li, Guofeng Han, Ning Li, Xin Li, Guang Liu, Yangzhou Sun, Guixin Wang, Yong Li, Zengchun Li, Qinchuan TI Identification of Key Genes Associated with Colorectal Cancer Based on the Transcriptional Network SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Differentially expressed genes; Transcriptional network; Module selection; Functional analysis ID Colorectal cancer; Differentially expressed genes; Transcriptional network; Module selection; Functional analysis AB Colorectal cancer (CRC) is among the most lethal human cancers, but the mechanism of the cancer is still unclear enough. We aimed to explore the key genes in CRC progression. The gene expression profile (GSE4183) of CRC was obtained from Gene Expression Omnibus database which included 8 normal samples, 15 adenoma samples, 15 CRC samples and 15 inflammatory bowel disease (IBD) samples. Thereinto, 8 normal, 15 adenoma, and 15 CRC samples were chosen for our research. The differentially expressed genes (DEGs) in normal vs. adenoma, normal vs. CRC, and adenoma vs. CRC, were identified using the Wilcoxon test method in R respectively. The interactive network of DEGs was constructed to select the significant modules using the Pearson’s correlation. Meanwhile, transcriptional network of DEGs was also constructed using the g: Profiler. Totally, 2, 741 DEGs in normal vs. adenoma, 1,484 DEGs in normal vs. CRC, and 396 DEGs in adenoma vs. CRC were identified. Moreover, function analysis of DEGs in each group showed FcR-mediated phagocytosis pathway in module 1, cardiac muscle contraction pathway in module 6, and Jak-STAT signaling pathway in module 19 were also enriched. Furthermore, MZF1 and AP2 were the transcription factor in module 6, with the target SP1, while SP1 was also a transcription in module 20. DEGs like NCF1, AKT, SP1, AP2, MZF1, and TPM might be used as specific biomarkers in CRC development. Therapy targeting on the functions of these key genes might provide novel perspective for CRC treatment. C1 [Chen, Guoting] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. [Li, Hengping] Shandong Laigang Hoaspital, Department of General Surgery, 271126 Laiwu, Shandong Province, China. [Niu, Xianping] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. [Li, Guofeng] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. [Han, Ning] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. [Li, Xin] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. [Li, Guang] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. [Liu, Yangzhou] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. [Sun, Guixin] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. [Wang, Yong] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. [Li, Zengchun] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. [Li, Qinchuan] Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, No. 150, Jimo Road, 200120 Shanghai, China. RP Li, Q (reprint author), Tongji University School of Medicine, East Hospital, Department of Emergency Surgery, 200120 Shanghai, China. EM li.qinchuan@163.com CR Rebecca SM, Deepa NaishadhamMA, Ahmedin Jemal DVM(2013, Cancer statistics, 2013. CA Cancer J Clin 63:11–30 ChenW-Q, Zeng H-M, Zheng R-S et al, 2012, Cancer incidence and mortality in China, 2007. Chin J Cancer Res 24:1–8 Goel A, Boland CR, 2012, Epigenetics of colorectal cancer. Gastroenterology 143:1442–1460 Garagnani P, Pirazzini C, Franceschi C, 2013, Colorectal cancer microenvironment: among nutrition, gut microbiota, inflammation and epigenetics. Curr Pharm Des 19:765–778 Vinci S, Gelmini S, Mancini I et al, 2013, Genetic and epigenetic factors in regulation ofmicroRNA in colorectal cancers.Methods 59: 138–146 Wilkinson J, 2012, Epigenome enhancer profiling reveals signature for colon cancer. Epigenomics 4(3):248 Schweiger MR, Hussong M, Rohr C et al, 2013, Genomics and epigenomics of colorectal cancer. Wiley Interdiscip Rev Syst Biol Med 5:205–219 Han Y, Kuang Y, Xue X et al, 2014, NLK, a novel target of miR- 199a-3p, functions as a tumor suppressor in colorectal cancer. Biomed Pharmacother 9:003 Zhang Y, Tian X, Ji H et al., 2014, Expression of SATB1 Promotes the Growth and Metastasis of Colorectal Cancer. PloS one 9 Van Cutsem E, Kohne C-H, Lang I et al, 2011, Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. JCO 2010(2033):5091 Misale S, Yaeger R, Hobor S et al., 2012, Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature Galamb O, Sipos F, Solymosi N et al, 2008, Diagnostic mRNA expression patterns of inflamed, benign, and malignant colorectal biopsy specimen and their correlation with peripheral blood results. Cancer Epidemiol Biomarkers Prev 17:2835–2845 Nam S, Park T, 2012, Pathway-based evaluation in early onset colorectal cancer suggests focal adhesion and immunosuppression along with Epithelial-Mesenchymal transition. PLoS ONE 7:e31685 Heijink D, Fehrmann R, de Vries E et al, 2011)A bioinformatical and functional approach to identify novel strategies for chemoprevention of colorectal cancer. Oncogene 30:2026–2036 Barrett T, TroupDB,Wilhite SE et al, 2007, NCBI GEO:mining tens of millions of expression profiles—database and tools update. Nucleic Acids Res 35:D760–D765 Irizarry RA, Hobbs B, Collin F et al, 2003, Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4:249–264 Gehan EA, 1965, A generalized Wilcoxon test for comparing arbitrarily singly-censored samples. Biometrika 52:203–223 Adler J, Parmryd I, 2010, Quantifying colocalization by correlation: the pearson correlation coefficient is superior to theMander’s overlap coefficient. J Int Soc Anal Cytol 77:733–742., DOI 10.1002/cyto.a. 20896 Collobert R, Bengio S,Mariethoz J, 2002, Torch: a modular machine learning software library. Technical Report IDIAP-RR 02-46, IDIAP Reimand J, Kull M, Peterson H et al, 2007, g: Profiler—a web-based toolset for functional profiling of gene lists from large-scale experiments. Nucleic Acids Res 35:W193–W200 Alvord G, Roayaei J, Stephens R et al, 2007, The DAVID gene functional classification tool: a novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol 8:R183 AshburnerM, Ball CA, Blake JA et al, 2000, Gene ontology: tool for the unification of biology. Nat Genet 25:25 Ogata H, Goto S, Sato K et al, 1999, KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res 27:29–34 Matys V, Fricke E, Geffers R et al, 2003, TRANSFAC: transcriptional regulation, from patterns to profiles. Nucleic Acids Res 31: 374–378 van Spriel AB, van Ojik HH, Bakker A et al, 2003, Mac-1, CD11b/ CD18, is crucial for effective Fc receptor–mediated immunity to melanoma. Blood 101:253–258 Roos D, de Boer M, Koker MYet al, 2006, Chronic granulomatous disease caused by mutations other than the common GT deletion in NCF1, the gene encoding the p47phox component of the phagocyte NADPH oxidase. Hum Mutat 27:1218–1229 Suraweera N, Zampeli E, Rogers P et al, 2004, NCF1, p47phox, and ncf1 pseudogenes are not associated with inflammatory bowel disease. Inflamm Bowel Dis 10:758–762 Parry D, Squire J, 1973, Structural role of tropomyosin in muscle regulation: analysis of the x-ray diffraction patterns from relaxed and contracting muscles. J Mol Biol 75:33–55 Luo YX, Cui J, Wang L et al, 2009, Identification of cancerassociated proteins by proteomics and downregulation of betatropomyosin expression in colorectal adenoma and cancer. Proteomics Clin Appl 3:1397–1406 Mlakar V, Berginc G, VolavsekMet al, 2009, Presence of activating KRAS mutations correlates significantly with expression of tumour suppressor genes DCN and TPM1 in colorectal cancer. BMC Cancer 9:1471–2407 Franke TF, Yang S-I, Chan TO et al, 1995, The protein kinase encoded by theAktproto-oncogene is a target of the PDGF-activated phosphatidylinositol 3-kinase. Cell 81:727–736 Baba Y, Nosho K, Shima K et al, 2011, Phosphorylated AKT expression is associated with PIK3CA mutation, low stage, and favorable outcome in 717 colorectal cancers. Cancer 117:1399–1408 Agarwal E, Brattain MG, Chowdhury S, 2013, Cell survival and metastasis regulation by Akt signaling in colorectal cancer. Cell Signal 25:1711–1719 PIM JS JAK-STAT signaling pathway. Diaz T, Navarro A, Ferrer G et al, 2011, Lestaurtinib inhibition of the Jak/STAT signaling pathway in hodgkin lymphoma inhibits proliferation and induces apoptosis. PLoS ONE 6:0018856 Yue CH, ChiuYW, Tung JN et al, 2012, Expression of protein kinase C alpha and the MZF-1 and Elk-1 transcription factors in human breast cancer cells. Chin J Physiol 55:31–36 Hsieh YH, Wu TT, Huang CYet al, 2007, Suppression of tumorigenicity of human hepatocellular carcinoma cells by antisense oligonucleotide MZF-1. Chin J Physiol 50:9–15 Mudduluru G, Vajkoczy P, Allgayer H, 2010, Myeloid zinc finger 1 induces migration, invasion, and in vivo metastasis through Axl gene expression in solid cancer. Mol Cancer Res 8:159–169 Deng Y, Wang J, Wang G et al, 2013, p55PIK transcriptionally activated by MZF1 promotes colorectal cancer cell proliferation. Biomed Res Int 868131:23 Raman D, Sai J, Hawkins O et al, 2014, Adaptor protein2, AP2, orchestrates CXCR2-mediated cell migration. Traffic 15:451–469 Kadonaga JT, Carner KR,Masiarz FR et al, 1987, Isolation of cDNA encoding transcription factor Sp1 and functional analysis of the DNA binding domain. Cell 51:1079–1090 Lam AK, Chan SS, 2014, Leung M Synchronous colorectal cancer: clinical, pathological and molecular implications. World J Gastroenterol 20(22):6815–6820 Stabach PR, Thiyagarajan MM, Woodfield GW et al, 2006, AP2alpha alters the transcriptional activity and stability of p53. Oncogene 25:2148–2159 Banerjee S, Sangwan V, McGinn O et al, 2013, Triptolideinduced cell death in pancreatic cancer is mediated by OGlcNAc modification of transcription factor Sp1. J Biol Chem 288:33927–33938 Li H, Zhang Y, Strose A et al, 2014, Integrated high-throughput analysis identifies Sp1 as a crucial determinant of p53-mediated apoptosis. Cell Death Differ 27:69 Zhao Y, Zhang W, Guo Z et al, 2013, Inhibition of the transcription factor Sp1 suppresses colon cancer stem cell growth and induces apoptosis in vitro and in nude mouse xenografts. Oncol Rep 30: 1782–1792 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 719 EP 725 DI 10.1007/s12253-014-9880-9 PG 7 ER PT J AU Chen, B Hu, KW Zhang, JW Wei, ZJ Meng, XL Xiong, MM AF Chen, Bo Hu, Kong-Wang Zhang, Jia-Wei Wei, Zhi-Jian Meng, Xiang-Ling Xiong, Mao-Ming TI A critical Analysis of the Relationship Between Aldehyde dehydrogenases-2 Glu487Lys Polymorphism and Colorectal Cancer Susceptibility SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Aldehyde dehydrogenases-2; ALDH-2; Polymorphism; Risk factor ID Colorectal cancer; Aldehyde dehydrogenases-2; ALDH-2; Polymorphism; Risk factor AB Studies investigating the association between genetic polymorphism of aldehyde dehydrogenases-2 (ALDH- 2) Glu487Lys and colorectal cancer (CRC) risk have reported conflicting results. Given this uncertainty, we carried out a critical analysis of published case-control studies to derive a more precise estimation of this relationship. Published literature from PubMed, EMBASE and China Knowledge Resource Integrated Database were retrieved, and the literature search was updated in June 2014. Eleven studies comprising 6965 subjects were selected (2300 cases and 4665 controls). Overall, our study showed no statistical significance for CRC risk associated with any of the genetic models of ALDH-2 Glu487Lys polymorphism. When studies were stratified for control source, a decreased risk of CRC for participants with Lys/Lys was observed in population based case-control studies [Lys/Lys vs. (Glu/Lys + Glu/Glu): odds ratio (OR)=0.57, 95% confidence interval (CI)=0.38–0.87]. Furthermore, we also confirmed the significant correlation between Glu487Lys polymorphism and the influence on the risk of rectal cancer in males [Glu/Glu vs. (Glu/Lys + Lys/Lys): OR=1.52, 95%CI= 1.10–2.08]. The combined effects of the two gene polymorphisms [ALDH-2 and alcohol dehydrogenase 1B (ADH-1B)] were also studied. Compared with subjects having ALDH-2 Lys+ with ADH-1B His/His, ORs and 95%CIs for those with ALDH-2 Glu/Glu and ADH-1B His/His was 3.42(0.57– 20.38). Similar trends were observed for the other two types of comparisons. Our study supports that ALDH-2 Glu487Lys polymorphism is associated with significant reduced risks of CRC in population-based samples, and of rectal cancer in males. C1 [Chen, Bo] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, Anhui, China. [Hu, Kong-Wang] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, Anhui, China. [Zhang, Jia-Wei] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, Anhui, China. [Wei, Zhi-Jian] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, Anhui, China. [Meng, Xiang-Ling] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, Anhui, China. [Xiong, Mao-Ming] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, Anhui, China. RP Xiong, MM (reprint author), The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, China. EM anyixmm@163.com CR Siegel R, Desantis C, Jemal A, 2014, Colorectal cancer statistics, 2014. CA Cancer J Clin 64:104–117 Durko L, Malecka-Panas E, 2014, Lifestyle modifications and colorectal cancer. Curr Colorectal Cancer Rep 10:45–54 Zhang A, Sun H, Yan G, Wang P, Han Y, Wang X, 2014, Metabolomics in diagnosis and biomarker discovery of colorectal cancer. Cancer Lett 345:17–20 Church D, Midgley R, Kerr D, 2012, Biomarkers in early-stage colorectal cancer: ready for prime time? Dig Dis 30(Suppl 2):27–33 Haas SL, YeW, Lohr JM(2012, Alcohol consumption and digestive tract cancer. Curr Opin Clin Nutr Metab Care 15:457–467 Cho E, Smith-Warner SA, Ritz J, van den Brandt PA, Colditz GA, Folsom AR, Freudenheim JL, Giovannucci E, Goldbohm RA, Graham S, Holmberg L, Kim DH, Malila N, Miller AB, Pietinen P, Rohan TE, Sellers TA, Speizer FE, Willett WC, Wolk A, Hunter DJ, 2004, Alcohol intake and colorectal cancer: a pooled analysis of 8 cohort studies. Ann Intern Med 140:603–613 Jarosz M, Sekula W, Rychlik E, 2013, Trends in dietary patterns, alcohol intake, tobacco smoking, and colorectal cancer in polish population in 1960–2008. Biomed Res Int 2013:183204 Bongaerts BW, de Goeij AF, Wouters KA, van Engeland M, Gottschalk RW, Van Schooten FJ, Goldbohm RA, van den Brandt PA, Weijenberg MP, 2011, Alcohol consumption, alcohol dehydrogenase 1c, adh1c, genotype, and risk of colorectal cancer in the netherlands cohort study on diet and cancer. Alcohol, Fayetteville, NY, 45:217–225 Kim J, Cho YA, Kim DH, Lee BH, Hwang DY, Jeong J, Lee HJ, Matsuo K, Tajima K, Ahn YO, 2012, Dietary intake of folate and alcohol, mthfr c677t polymorphism, and colorectal cancer risk in korea. Am J Clin Nutr 95:405–412 Yin G, Morita M, Ohnaka K, Toyomura K, Hamajima N, Mizoue T, Ueki T, Tanaka M, Kakeji Y, Maehara Y, Okamura T, Ikejiri K, Futami K, Yasunami Y, Maekawa T, Takenaka K, Ichimiya H, Terasaka R, 2012, Genetic polymorphisms of xrcc1, alcohol consumption, and the risk of colorectal cancer in japan. J Epidemiol / Jpn Epidemiol Assoc 22:64–71 Chen K, Jiang Q,Ma X, Li Q, Yao K, YuW, Zheng S, 2005, Alcohol drinking and colorectal cancer: a population-based prospective cohort study in china. Eur J Epidemiol 20:149–154 Baan R, Straif K, Grosse Y, Secretan B, El Ghissassi F, Bouvard V, Altieri A, Cogliano V, Group WHOIAfRoCMW, 2007, Carcinogenicity of alcoholic beverages. Lancet Oncol 8:292–293 Secretan B, Straif K, Baan R, Grosse Y, El Ghissassi F, Bouvard V, Benbrahim-Tallaa L, Guha N, Freeman C, Galichet L, Cogliano V, Group WHOIAfRoCMW, 2009, A review of human carcinogens– part e: tobacco, areca nut, alcohol, coal smoke, and salted fish. Lancet Oncol 10:1033–1034 Yoshida A, Rzhetsky A, Hsu LC, Chang C, 1998, Human aldehyde dehydrogenase gene family. Eur J Biochem / FEBS 251:549–557 Crous-Bou M, Rennert G, Cuadras D, Salazar R, Cordero D, Saltz Rennert H, Lejbkowicz F, Kopelovich L, Monroe Lipkin S, Bernard Gruber S, Moreno V, 2013, Polymorphisms in alcohol metabolism genes adh1b and aldh2, alcohol consumption and colorectal cancer. PLoS One 8:e80158 WuM, Chang SC, Kampman E, Yang J,Wang XS, Gu XP, Han RQ, Liu AM, Wallar G, Zhou JY, Kok FJ, Zhao JK, Zhang ZF, 2013, Single nucleotide polymorphisms of adh1b, adh1c and aldh2 genes and esophageal cancer: a population-based case-control study in china. Int J Cancer J Int Cancer 132:1868–1877 Wang HL, Zhou PY, Liu P, Zhang Y, 2014, Aldh2 and adh1 genetic polymorphisms may contribute to the risk of gastric cancer: a metaanalysis. PLoS One 9:e88779 Zhao H, Liu KJ, Lei ZD, Lei SL, Tian YQ, 2014, Meta-analysis of the aldehyde dehydrogenases-2, aldh2, glu487lys polymorphism and colorectal cancer risk. PLoS One 9:e88656 Guo XF, Wang J, Yu SJ, Song J, Ji MY, Zhang JX, Cao Z, Wang J, Dong WG, 2013, Meta-analysis of the adh1b and aldh2 polymorphisms and the risk of colorectal cancer in east asians. InternMed 52: 2693–2699 Tsutsumi M, Takase S, Takada A, 1993, Genetic factors related to the development of carcinoma in digestive organs in alcoholics. Alcohol and alcoholism, Oxford, Oxfordshire). Supplement 1B:21–26 Yokoyama A, Muramatsu T, Ohmori T, Yokoyama T, Okuyama K, Takahashi H, Hasegawa Y, Higuchi S, Maruyama K, Shirakura K, Ishii H, 1998, Alcohol-related cancers and aldehyde dehydrogenase-2 in japanese alcoholics. Carcinogenesis 19:1383–1387 Murata M, Tagawa M, Watanabe S, Kimura H, Takeshita T, Morimoto K, 1999, Genotype difference of aldehyde dehydrogenase 2 gene in alcohol drinkers influences the incidence of japanese colorectal cancer patients. Japan J Cancer Res: Gann 90: 711–719 Matsuo K, Hamajima N, Hirai T, Kato T, Koike K, Inoue M, Takezaki T, Tajima K, 2002, Aldehyde dehydrogenase 2, aldh2, genotype affects rectal cancer susceptibility due to alcohol consumption. J Epidemiol / Jpn Epidemiol Assoc 12:70–76 Otani T, IwasakiM, Hanaoka T, KobayashiM, Ishihara J, Natsukawa S, Shaura K, Koizumi Y, Kasuga Y, Yoshimura K, Yoshida T, Tsugane S, 2005, Folate, vitamin b6, vitamin b12, and vitamin b2 intake, genetic polymorphisms of related enzymes, and risk of colorectal cancer in a hospital-based case-control study in japan. Nutr Cancer 53:42–50 Matsuo K,Wakai K, Hirose K, Ito H, Saito T, Suzuki T, Kato T, Hirai T, Kanemitsu Y, Hamajima H, Tajima K, 2006, A gene-gene interaction between aldh2 glu487lys and adh2 his47arg polymorphisms regarding the risk of colorectal cancer in japan. Carcinogenesis 27: 1018–1023 Jiang A, Gao C, Wu J, Takezaki T, Cao J, Tajima K, Shen H, 2007, Association between adh2/aldh2 genetic polymorphism and habit of alcohol drinking and the susceptibility of rectal cancer. Chin J Cancer Prev Treat 14:1445–1449 Yin G, Kono S, Toyomura K,MooreMA, Nagano J,Mizoue T, Mibu R, TanakaM, Kakeji Y, Maehara Y, Okamura T, Ikejiri K, Futami K, Yasunami Y, Maekawa T, Takenaka K, Ichimiya H, Imaizumi N, 2007, Alcohol dehydrogenase and aldehyde dehydrogenase polymorphisms and colorectal cancer: the fukuoka colorectal cancer study. Cancer Sci 98:1248–1253 Yang H, Zhou Y, Zhou Z, Liu J, Yuan X, Matsuo K, Takezaki T, Tajima K, Cao J, 2009, A novel polymorphism rs1329149 of cyp2e1 and a known polymorphism rs671 of aldh2 of alcohol metabolizing enzymes are associated with colorectal cancer in a southwestern chinese population. Cancer Epidemiol Biomarkers Prev 18:2522–2527 Miyasaka K, Hosoya H, Tanaka Y, Uegaki S, Kino K, Shimokata H, Kawanami T, Funakoshi A, 2010, Association of aldehyde dehydrogenase 2 gene polymorphism with pancreatic cancer but not colon cancer. Geriatr Gerontol Int 10(Suppl 1):S120–S126 Chiang CP, Jao SW, Lee SP, Chen PC, Chung CC, Lee SL, Nieh S, Yin SJ, 2012, Expression pattern, ethanol-metabolizing activities, and cellular localization of alcohol and aldehyde dehydrogenases in human large bowel: association of the functional polymorphisms of adh and aldh genes with hemorrhoids and colorectal cancer. Alcohol, Fayetteville, NY, 46:37–49 Chen B, Cao L, Yang P, Zhou Y, Wu XT, 2012, Cyclin d1, ccnd1, g870a gene polymorphism is an ethnicity-dependent risk factor for digestive tract cancers: a meta-analysis comprising 20,271 subjects. Cancer Epidemiol 36:106–115 Zakhari S, 2006, Overview: how is alcohol metabolized by the body? Alcohol Res Health: J Natl Inst Alcohol Abus Alcohol 29:245–254 Druesne-Pecollo N, Tehard B,MalletY, GerberM, Norat T, Hercberg S, Latino-Martel P, 2009, Alcohol and genetic polymorphisms: effect on risk of alcohol-related cancer. Lancet Oncol 10:173–180 Gao CM, Takezaki T,Wu JZ, Zhang XM, Cao HX, Ding JH, Liu YT, Li SP, Cao J, Matsuo K, Hamajima N, Tajima K, 2008, Polymorphisms of alcohol dehydrogenase 2 and aldehyde dehydrogenase 2 and colorectal cancer risk in chinese males. World J Gastroenterol : WJG 14:5078–5083 Mulligan CJ, Robin RW, Osier MV, Sambuughin N, Goldfarb LG, Kittles RA, Hesselbrock D, Goldman D, Long JC, 2003, Allelic variation at alcohol metabolism genes, adh1b, adh1c, aldh2, and alcohol dependence in an american indian population. Hum Genet 113:325–336 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 727 EP 733 DI 10.1007/s12253-014-9881-8 PG 7 ER PT J AU Isic Dencic, MT Savin, BS Selemetjev, AS Paskas, DS Zivaljevic, RV Bozic, DV Cvejic, SD AF Isic Dencic, M Tijana Savin, B Svetlana Selemetjev, A Sonja Paskas, D Svetlana Zivaljevic, R Vladan Bozic, D Vesna Cvejic, S Dubravka TI Strong Expression of HBME-1 Associates with High-Risk Clinicopathological Factors of Papillary Thyroid Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary thyroid carcinoma; HBME-1; Diagnosis; Prognosis; Lymph node metastasis; pT; pTNM ID Papillary thyroid carcinoma; HBME-1; Diagnosis; Prognosis; Lymph node metastasis; pT; pTNM AB Thyroid cancer comprises a heterogeneous group of lesions with great diversity of biological behaviour. Markers which could help clinicians to identify high-risk patients for tailored optimization of clinical management are of crucial importance. HBME-1 protein level was analysed immunohistochemically using routinely prepared archival tissue sections of a broad range of papillary thyroid carcinoma (PTC) variants and in corresponding lymph node metastases (LNM). The results were evaluated in comparison with clinicopathological features of PTC. Positive immunoreaction was noticed in most classical (83/92; 90.2 %), follicular (60/71; 84.5 %) and trabecular (4/5; 80.0 %) variants of PTC. All cases of macrofollicular, Warthin-like and diffuse sclerosing PTC variants were HBME-1 positive (4/4, 3/3, 2/2; 100 % respectively). Tall cell and solid PTC variants showed diversity of staining (2/3; 66.67 % and 13/23; 56.52 % respectively), while PTCs with mixed histological pattern containing insular areas were mainly weakly positive (2/5; 40.0 %). A single case of clear cell PTC variant showed no reaction. Moreover, all matched metastatic PTC into lymph nodes (LNM) were HBME-1 positive (17/17; 100 %) and expressed HBME-1 in a similar pattern to the matched primary tumour. We also found a statistically significant association between high HBME-1 expression and the presence of lymph node metastasis, advanced pT status and pTNM stage (P<0.05), but only a tendency for association with extrathyroidal invasion of the tumour (P=0.058). Therefore, we recommend using immunoexpression of HBME-1 as useful mean to increase the likelihood of detecting most PTC variants and to predict some unfavourable clinical parameters in these patients. C1 [Isic Dencic, M Tijana] University of Belgrade, Institute for the Application of Nuclear Energy - INEP, Banatska 31b, Zemun, 11080 Belgrade, Serbia. [Savin, B Svetlana] University of Belgrade, Institute for the Application of Nuclear Energy - INEP, Banatska 31b, Zemun, 11080 Belgrade, Serbia. [Selemetjev, A Sonja] University of Belgrade, Institute for the Application of Nuclear Energy - INEP, Banatska 31b, Zemun, 11080 Belgrade, Serbia. [Paskas, D Svetlana] University of Belgrade, Institute for the Application of Nuclear Energy - INEP, Banatska 31b, Zemun, 11080 Belgrade, Serbia. [Zivaljevic, R Vladan] Clinical Center of Serbia, Clinic of Endocrinology, Diabetes and Metabolic Diseases, 11000 Belgrade, Serbia. [Bozic, D Vesna] University of Belgrade, Faculty of Medicine, Institute for Pathology, 11000 Belgrade, Serbia. [Cvejic, S Dubravka] University of Belgrade, Institute for the Application of Nuclear Energy - INEP, Banatska 31b, Zemun, 11080 Belgrade, Serbia. RP Isic Dencic, MT (reprint author), University of Belgrade, Institute for the Application of Nuclear Energy - INEP, 11080 Belgrade, Serbia. EM tijana@inep.co.rs CR Siegel R, DeSantis C, Virgo K, Stein K, Mariotto A, Smith T, Cooper D, Gansler T, Lerro C, Fedewa S, Lin C, Leach C, Cannady RS, Cho H, Scoppa S, Hachey M, Kirch R, Jemal A, Ward E, 2012, Cancer treatment and survivorship statistics, 2012. CA Cancer J Clin 62: 220–241., DOI 10.3322/caac.21149 Treseler PA, Clark OH, 1997, Prognostic factors in thyroid carcinoma. Surg Oncol Clin N Am 6:555–598 Gilliland FD, Hunt WC, Morris DM, Key CR, 1997, Prognostic factors for thyroid carcinoma. A population-based study of 15,698 cases from surveillance, epidemiology and END results, SEER, program 1973–1991. Cancer 79:564–573 Sipos JA, Mazzaferri EL, 2010, Thyroid cancer epidemiology and prognostic variables. Clin Oncol, R Coll Radiol, 22:395–404., DOI 10. 1016/j.clon.2010.05.004 Podnos YD, Smith D,Wagman LD, Ellenhorn JD, 2005, The implication of lymph node metastasis on survival in patients with welldifferentiated thyroid cancer. Am Surg 71:731–734 Fischer S, Asa SL, 2008, Application of immunohistochemistry to thyroid neoplasms.Arch Pathol Lab Med 132:359–372., DOI 10.1043/ 1543-2165(2008)132[359:AOITTN]2.0.CO;2 Barut F, Onak Kandemir N, Bektas S, Bahadir B, Keser S, Ozdamar S, 2010, Universal markers of thyroid malignancies: galectin-3, HBME-1, and cytokeratin-19. Endocr Pathol 21:80–89., DOI 10. 1007/s12022-010-9114-y Sethi K, Sarkar S, Das S, Mohanty B, Mandal M, 2010, Biomarkers for the diagnosis of thyroid cancer. J Exp Ther Oncol 8:341–352 Cheng S, Serra S, Mercado M, Ezzat S, Asa S, 2011, A highthroughput proteomic approach provides distinct signatures for thyroid cancer behavior. Clin Cancer Res 17:2385–2394., DOI 10.1158/ 1078-0432.CCR-10-2837 Paunovic I, Isic T, Havelka M, Tatic S, Cvejic D, Savin S, 2012, Combined immunohistochemistry for thyroid peroxidase, galectin-3, CK19 and HBME-1 in differential diagnosis of thyroid tumors. APMIS 120:368–379., DOI 10.1111/j.1600-0463.2011.02842.x Raphael SJ, 2002, The meanings of markers: ancillary techniques in diagnosis of thyroid neoplasia. Endocr Pathol 13:301–311 deMatos PS, Ferreira AP, de Oliveira F, Assumpcao LVM,Matze K, Ward LS, 2005, Usefulness of HBME-1, cytokeratin 19 and galectin- 3 immunostaining in the diagnosis of thyroid malignancy. Histopathol 47:391–401 Saleh HA, Jin B, Barnwell J, Alzohaili O, 2010, Utility of immunohistochemical markers in differentiating benign from malignant follicular-derived thyroid nodules. Diagn Pathol 5:9., DOI 10.1186/ 1746-1596-5-9 Rossi ED, Raffaelli M, Mule A, Miraglia A, Lombardi CP, Vecchio FM, Fadda G, 2006, Simultaneous immunohistochemical expression of HBME-1 and galectin-3 differentiates papillary carcinomas from hyperfunctioning lesions of the thyroid. Histopathology 48:795–800., DOI 10.1111/j.1365-2559.2006.02428.x Cheung CC, Ezzat S, Freeman JL, Rosen IB, Asa SL, 2001, Immunohistochemical diagnosis of papillary thyroid carcinoma. Mod Pathol 14:338–342 Prasad M, Pellegata N, Huang Y, Nagaraja H, de la Chapelle A, Kloos R, 2005, Galectin-3, fibronectin-1, CITED-1, HBME-1 and cytokeratin-19 immunohistochemistry is useful for the differential diagnosis of thyroid tumors. Mod Pathol 18:48–57 Scognamiglio T, Hyjek E, Kao J, Chen YT, 2006, Diagnostic usefulness ofHBME1, galectin-3, CK19, and CITED1 and evaluation of their expression in encapsulated lesions with questionable features of papillary thyroid carcinoma. Am J Clin Pathol 126:700–708 NgaME, Lim GS, Soh CH, KumarasingheMP, 2008, HBME-1 and CK19 are highly discriminatory in the cytological diagnosis of papillary thyroid carcinoma. Diagn Cytopathol 36:550–556., DOI 10. 1002/dc.20841 Choi YL, Kim MK, Suh JW, Han J, Kim JH, Yang JH, Nam SJ, 2005, Immunoexpression of HBME-1, high molecular weight cytokeratin, cytokeratin 19, thyroid transcription factor-1, and Ecadherin in thyroid carcinomas. J Korean Med Sci 20:853–859 Rossi ED, Straccia P, Palumbo M, Stigliano E, Revelli L, Lombardi CP, Santeusanio G, Pontecorvi A, Fadda G, 2013, Diagnostic and prognostic role of HBME-1, galectin-3, and β-catenin in poorly differenti a ted and anaplast ic thyroid carcinomas. Appl Immunohistochem Mol Morphol 21:237–241., DOI 10.1097/PAI. 0b013e3182688d0f Torregrossa L, Faviana P, Camacci T, Materazzi G, Berti P, Minuto M, Elisei R, Vitti P, Miccoli P, Basolo F, 2007, Galectin-3 is highly expressed in nonencapsulated papillary thyroid carcinoma but weakly expressed in encapsulated type; comparison with Hector Battifora mesothelial cell 1 immunoreactivity. Hum Pathol 38:1482–1488 Hirokawa M, Horiguchi H,Wakatsuki S, Miki H, Sonoo H, Manabe T, Sano T, 2001, Intranodal benign thyroid tissue: significance of HBME-1 in differentiation from metastatic papillary thyroid carcinoma. APMIS 109:875–880 Liang H, Zhong Y, Luo Z, Huang Y, Lin H, Zhan S, Xie K, Li QQ, 2011, Diagnostic value of 16 cellular tumor markers for metastatic thyroid cancer: an immunohistochemical study. Anticancer Res 31: 3433–3440 CuiW, SangW, Zheng S, Ma Y, Liu X, ZhangW(2012, Usefulness of cytokeratin-19, galectin-3, and Hector Battifora mesothelial-1 in the diagnosis of benign and malignant thyroid nodules. Clin Lab 58: 673–680 LiVolsi VA, Albores-Saavedra J, Asa SL et al, 2004, Papillary carcinoma. In: DeLellis RA, Lloyd R, Heitz PU, Eng C, eds)World health organization classification of tumors. Pathology and genetics of tumors of endocrine organs. IARC Press, Lyon, pp 50–66 Volante M, Collini P, Nikiforov YE, Sakamoto A, Kakudo K, Katoh R, Lloyd RV, LiVolsi VA, Papotti M, Sobrinho-Simoes M, Bussolati G, Rosai J, 2007, Poorly differentiated thyroid carcinoma: the Turin proposal for the use of uniform diagnostic criteria and an algorithmic diagnostic approach. Am J Surg Pathol 31:1256–1264 Edge SB, Byrd DR, Compton CC et al., 2010,, ed). AJCC cancer staging manual. In: Thyroid, 7thedn. Springer, New York, pp 87–96 Park YJ, Kwak SH, Kim DC, Kim H, Choe G, do Park J, Jang HC, Park SH, Cho BY, Park SY, 2007, Diagnostic value of galectin-3, HBME-1, cytokeratin 19, high molecular weight cytokeratin, cyclin D1 and p27, kip1, in the differential diagnosis of thyroid nodules. J Korean Med Sci 22:621–628., DOI 10.3346/jkms.2007.22.4.621 Zhu X, Sun T, Lu H, Zhou X, Lu Y, Cai X, 2010, Diagnostic significance of CK19, RET, galectin-3 and HBME-1 expression for papillary thyroid carcinoma. J Clin Pathol 63:786–789., DOI 10.1136/jcp. 2010.076901 de Matos LL, Del Giglio AB, Matsubayashi CO, de Lima FM, Del Giglio A, da Silva PinhalMA, 2012, Expression of CK-19, galectin- 3 and HBME-1 in the differentiation of thyroid lesions: systematic review and diagnostic meta-analysis. Diagn Pathol 7:97., DOI 10. 1186/1746-1596-7-97, Review Emad R, Maha A, Kfoury HK, Al-Sheikh AM, Zaidi SN, 2011, Three cases of macrofollicular variant of papillary thyroid carcinoma. Ann Saudi Med 31:644–647., DOI 10.4103/0256-4947. 87104 CardenasMG, Kini S,WisgerhofM(2009, Two patients with highly aggressive macrofollicular variant of papillary thyroid carcinoma. Thyroid 19:413–416., DOI 10.1089/thy.2008.0178 Ghossein R, Leboeuf R, Patel KN, RiveraM, Katabi N, Carlson DL, Tallini G, Shaha A, Singh B, Tuttle RM, 2007, Tall cell variant of papillary thyroid carcinoma without extrathyroid extension: biologic behavior and clinical implications. Thyroid 17:655–661 Rigau V, Martel B, Evrard C, Rousselot P, Galateau-Salle F, 2001, HBME-1 immunostaining in thyroid pathology. Ann Pathol 21:15– 20 Ziadi S, Trimeche M, Zermani R, Elmay A, Baltagi-Ben Jilani S, 2005, Diagnostic contribution of HBME-1 and anti-cytokeratin-19 antibodies in thyroid pathology: a retrospective study of 163 cases. Tunis Med 83:274–278 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 735 EP 742 DI 10.1007/s12253-014-9883-6 PG 8 ER PT J AU Sumegi, K Jaromi, L Magyari, L Kovesdi, E Duga, B Szalai, R Maasz, A Matyas, P Janicsek, I Melegh, B AF Sumegi, Katalin Jaromi, Luca Magyari, Lili Kovesdi, Erzsebet Duga, Balazs Szalai, Renata Maasz, Anita Matyas, Petra Janicsek, Ingrid Melegh, Bela TI Functional Variants of Lipid Level Modifier MLXIPL, GCKR, GALNT2, CILP2, ANGPTL3 and TRIB1 Genes in Healthy Roma and Hungarian Populations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Polymorphisms; Triglyceride; Roma; Hungarian ID Polymorphisms; Triglyceride; Roma; Hungarian AB The role of triglyceride metabolism in different diseases, such as cardiovascular or cerebrovascular diseases is still under extensive investigations. In genome-wide studies several polymorphisms have been reported, which are highly associated with plasma lipid level changes. Our goal was to examine eight variants: rs12130333 at the ANGPTL3, rs16996148 at the CILP2, rs17321515 at the TRIB1, rs17145738 and rs3812316 of the MLXIPL, rs4846914 at GALNT2, rs1260326 and rs780094 residing at the GCKR loci. A total of 399 Roma (Gypsy) and 404 Hungarian population samples were genotyped using PCR-RFLP method. Significant differences were found between Roma and Hungarian population samples in both MLXIPL variants (C allele frequency of rs17145738: 94.1% vs. 85.6%, C allele frequency of rs3812316: 94.2% vs. 86.8% in Romas vs. in Hungarians, p<0.05), in ANGPTL3 (Tallele frequency of rs1213033: 12.2% vs. 18.5% in Romas vs. Hungarians, p<0.05) and GALNT2 (G allele frequency of rs4846914: 46.6% vs. 54.5% Romas vs. in Hungarians, p<0.05), while no differences over SNPs could be verified and the known minor alleles showed no correlation with triglyceride levels in any population samples. The current study revealed fundamental differences of known triglyceride modifying SNPs in Roma population. Failure of finding evidence for affected triglyceride metabolismshows that these susceptibility genes aremuch less effective compared for example to the apolipoprotein A5 gene. C1 [Sumegi, Katalin] University of Pecs, Department of Medical Genetics and Child Development, Szigeti u. 12, H-7624 Pecs, Hungary. [Jaromi, Luca] University of Pecs, Department of Medical Genetics and Child Development, Szigeti u. 12, H-7624 Pecs, Hungary. [Magyari, Lili] University of Pecs, Department of Medical Genetics and Child Development, Szigeti u. 12, H-7624 Pecs, Hungary. [Kovesdi, Erzsebet] University of Pecs, Department of Medical Genetics and Child Development, Szigeti u. 12, H-7624 Pecs, Hungary. [Duga, Balazs] University of Pecs, Department of Medical Genetics and Child Development, Szigeti u. 12, H-7624 Pecs, Hungary. [Szalai, Renata] University of Pecs, Department of Medical Genetics and Child Development, Szigeti u. 12, H-7624 Pecs, Hungary. [Maasz, Anita] University of Pecs, Department of Medical Genetics and Child Development, Szigeti u. 12, H-7624 Pecs, Hungary. [Matyas, Petra] University of Pecs, Department of Medical Genetics and Child Development, Szigeti u. 12, H-7624 Pecs, Hungary. [Janicsek, Ingrid] University of Pecs, Department of Medical Genetics and Child Development, Szigeti u. 12, H-7624 Pecs, Hungary. [Melegh, Bela] University of Pecs, Department of Medical Genetics and Child Development, Szigeti u. 12, H-7624 Pecs, Hungary. RP Melegh, B (reprint author), University of Pecs, Department of Medical Genetics and Child Development, H-7624 Pecs, Hungary. EM melegh.bela@pte.hu CR Szalai C, Keszei M, Duba J, Prohaszka Z, Kozma GT, Csaszar A, Balogh S, Almassy Z, Fust G, Czinner A, 2004, Polymorphismin the promoter region of the apolipoprotein A5 gene is associated with an increased susceptibility for coronary artery disease. Atherosclerosis 173(1):109–114 Vaessen SF, Schaap FG, Kuivenhoven JA, Groen AK, Hutten BA, Boekholdt SM, Hattori H, Sandhu MS, Bingham SA, Luben R, Palmen JA, Wareham NJ, Humphries SE, Kastelein JJ, Talmud PJ, Khaw KT, 2006, Apolipoprotein A-V, triglycerides and risk of coronary artery disease: the prospective Epic-Norfolk Population Study. J Lipid Res 47(9):2064–2070 Martinelli N, Girelli D, Ferraresi P, Olivieri O, Lunghi B,Manzato F, Corrocher R, Bernardi F, 2007, Increased factor VIII coagulant activity levels in male carriers of the factor V R2 polymorphism. Blood Coagul Fibrinolysis 18(2):125–129 Maasz A, Kisfali P, Jaromi L, Horvatovich K, Szolnoki Z, Csongei V, Safrany E, Sipeky C, Hadarits F, Melegh B, 2008, Apolipoprotein A5 gene IVS3+G476A allelic variant confers susceptibility for development of ischemic stroke. Circ J 72(7):1065–1070 Maasz A, Kisfali P, Szolnoki Z, Hadarits F, Melegh B, 2008, Apolipoprotein A5 gene C56G variant confers risk for the development of large-vessel associated ischemic stroke. J Neurol 255(5): 649–654 Feitosa MF, An P, Ordovas JM, Ketkar S, Hopkins PN, Straka RJ, Arnett DK, Borecki IB, 2011, Association of gene variants with lipid levels in response to fenofibrate is influenced bymetabolic syndrome status. Atherosclerosis 215(2):435–439 Garelnabi M, Lor K, Jin J, Chai F, Santanam N, 2012, The paradox of ApoA5 modulation of triglycerides: Evidence from clinical and basic research. Clin Biochem Mohas M, Kisfali P, Jaromi L, Maasz A, Feher E, Csongei V, Polgar N, Safrany E, Cseh J, Sumegi K, Hetyesy K, Wittmann I, Melegh B, 2010, GCKR gene functional variants in type 2 diabetes and metabolic syndrome: do the rare variants associate with increased carotid intima-media thickness? Cardiovasc Diabetol 9:79 Jaromi L, Csongei V, Polgar N, Szolnoki Z, Maasz A, Horvatovich K, Farago B, Sipeky C, Safrany E, Magyari L, Kisfali P, Mohas M, Janicsek I, Lakner L, Melegh B, 2010, Functional variants of glucokinase regulatory protein and apolipoprotein A5 genes in ischemic stroke. J Mol Neurosci 41(1):121–128 Jaromi L, Csongei V, Polgar N, Rappai G, Szolnoki Z, Maasz A, Horvatovich K, Safrany E, Sipeky C, Magyari L, Melegh B, 2011, Triglyceride level-influencing functional variants of the ANGPTL3, CILP2, and TRIB1 loci in ischemic stroke. Neuromolecular Med 13(3):179–186 Johansen CT, Hegele RA, 2012, Allelic and phenotypic spectrum of plasma triglycerides. Biochim Biophys Acta 1821(5):833–842 Souverein OW, Jukema JW, Boekholdt SM, Zwinderman AH, Tanck MW, 2005, Polymorphisms in APOA1 and LPL genes are statistically independently associated with fasting TG in men with CAD. Eur J Hum Genet 13(4):445–451 Havasi V, Szolnoki Z, Talian G, Bene J, Komlosi K, Maasz A, Somogyvari F, Kondacs A, Szabo M, Fodor L, Bodor A, Melegh B, 2006, Apolipoprotein A5 gene promoter region T-1131C polymorphism associates with elevated circulating triglyceride levels and confers susceptibility for development of ischemic stroke. J Mol Neurosci 29(2):177–183 Freiberg JJ, Tybjaerg-Hansen A, Jensen JS, Nordestgaard BG, 2008, Nonfasting triglycerides and risk of ischemic stroke in the general population. JAMA 300(18):2142–2152 Willer CJ, Sanna S, Jackson AU, Scuteri A, Bonnycastle LL, Clarke R, Heath SC, Timpson NJ, Najjar SS, Stringham HM, Strait J, Duren WL, Maschio A, Busonero F, Mulas A, Albai G, Swift AJ, Morken MA, Narisu N, Bennett D, Parish S, Shen H, Galan P, Meneton P, Hercberg S, Zelenika D, Chen WM, Li Y, Scott LJ, Scheet PA, Sundvall J, Watanabe RM, Nagaraja R, Ebrahim S, Lawlor DA, Ben-Shlomo Y, Davey-Smith G, Shuldiner AR, Collins R, Bergman RN, Uda M, Tuomilehto J, Cao A, Collins FS, Lakatta E, Lathrop GM, BoehnkeM, Schlessinger D,Mohlke KL, Abecasis GR, 2008, Newly identified loci that influence lipid concentrations and risk of coronary artery disease. Nat Genet 40(2):161–169 Kathiresan S, Willer CJ, Peloso GM, Demissie S, Musunuru K, Schadt EE, Kaplan L, Bennett D, Li Y, Tanaka T, Voight BF, Bonnycastle LL, Jackson AU, Crawford G, Surti A, Guiducci C, Burtt NP, Parish S, Clarke R, Zelenika D, Kubalanza KA, Morken MA, Scott LJ, Stringham HM, Galan P, Swift AJ, Kuusisto J, Bergman RN, Sundvall J, Laakso M, Ferrucci L, Scheet P, Sanna S, Uda M, Yang Q, Lunetta KL, Dupuis J, de Bakker PI, O’Donnell CJ, Chambers JC, Kooner JS, Hercberg S, Meneton P, Lakatta EG, Scuteri A, Schlessinger D, Tuomilehto J, Collins FS, Groop L, Altshuler D, Collins R, Lathrop GM, Melander O, Salomaa V, Peltonen L, Orho-Melander M, Ordovas JM, Boehnke M, Abecasis GR,Mohlke KL, Cupples LA, 2009, Common variants at 30 loci contribute to polygenic dyslipidemia. Nat Genet 41(1):56– 65 Labreuche J, Touboul PJ, Amarenco P, 2009, Plasma triglyceride levels and risk of stroke and carotid atherosclerosis: a systematic review of the epidemiological studies. Atherosclerosis 203(2):331– 345 Perez-Martinez P, Garcia-Rios A, Delgado-Lista J, Perez-Jimenez F, Lopez-Miranda J, 2011, Nutrigenetics of the postprandial lipoprotein metabolism: evidences from human intervention studies. Curr Vasc Pharmacol 9(3):287–291 Aslibekyan S, Goodarzi MO, Frazier-Wood AC, Yan X, Irvin MR, Kim E, Tiwari HK, Guo X, Straka RJ, Taylor KD, TsaiMY, Hopkins PN, Korenman SG, Borecki IB, Chen YD, Ordovas JM, Rotter JI, Arnett DK, 2012, Variants identified in a GWAS meta-analysis for blood lipids are associated with the lipid response to fenofibrate. PLoS One 7(10):e48663 Grundy SM, 2002, Approach to lipoprotein management in 2001 National Cholesterol Guidelines. Am J Cardiol 90(8A):11i–21i Brautbar A, Covarrubias D, Belmont J, Lara-Garduno F, Virani SS, Jones PH, Leal SM, Ballantyne CM, 2011, Variants in the APOA5 gene region and the response to combination therapy with statins and fenofibric acid in a randomized clinical trial of individuals with mixed dyslipidemia. Atherosclerosis 219(2):737–742 Kisfali P, Mohas M, Maasz A, Hadarits F, Marko L, Horvatovich K, Oroszlan T, Bagosi Z, Bujtor Z, Gasztonyi B,Wittmann I,Melegh B, 2008, Apolipoprotein A5 IVS3+476Aallelic variant associates with increased trigliceride levels and confers risk for development of metabolic syndrome in Hungarians. Circ J 72(1):40–43 Kathiresan S,Melander O, Guiducci C, Surti A, Burtt NP, Rieder MJ, Cooper GM, Roos C, Voight BF, Havulinna AS, Wahlstrand B, Hedner T, Corella D, Tai ES, Ordovas JM, Berglund G, Vartiainen E, Jousilahti P, Hedblad B, Taskinen MR, Newton-Cheh C, Salomaa V, Peltonen L, Groop L,AltshulerDM, Orho-MelanderM(2008, Six new loci associated with blood low-density lipoprotein cholesterol, high-density lipoprotein cholesterol or triglycerides in humans. Nat Genet 40(2):189–197 Shimizugawa T, Ono M, Shimamura M, Yoshida K, Ando Y, Koishi R, Ueda K, Inaba T, Minekura H, Kohama T, Furukawa H, 2002, ANGPTL3 decreases very low density lipoprotein triglyceride clearance by inhibition of lipoprotein lipase. J Biol Chem 277(37):33742– 33748 Seidelmann SB, Li L, Shen GQ, Topol EJ, Wang QK, 2008, Identification of a novel locus for triglyceride on chromosome 1p31-32 in families with premature CAD and MI. J Lipid Res 49(5):1034–1038 Izar MC, Fonseca FA, Ihara SS, Kasinski N, Sang WH, Lopes IE, Pinto Ldo E, Relvas WG, Lourenco D, Tufik S, de Paola AA, Carvalho AC, 2003, Risk factors, biochemical markers, and genetic polymorphisms in early coronary artery disease. Arq Bras Cardiol 80(4):379–395 Thomsen SB, Rathcke CN, Skaaby T, Linneberg A, Vestergaard H, 2012, The association between genetic variations of CHI3L1, levels of the encoded glycoprotein YKL-40 and the lipid profile in a Danish population. PLoS One 7(10):e47094 Baroni MG, Berni A, Romeo S, Arca M, Tesorio T, Sorropago G, Di Mario U, Galton DJ, 2003, Genetic study of common variants at the Apo E, Apo AI, Apo CIII, Apo B, lipoprotein lipase, LPL, and hepatic lipase, LIPC, genes and coronary artery disease, CAD): variation in LIPC gene associates with clinical outcomes in patients with established CAD. BMC Med Genet 4:8 Meigs JB, Nathan DM, D’Agostino RB Sr, Wilson PW, 2002, Fasting and postchallenge glycemia and cardiovascular disease risk: the Framingham offspring study. Diabetes Care 25(10):1845–1850 Pennacchio LA, Rubin EM(2003, Apolipoprotein A5, a newly identified gene that affects plasma triglyceride levels in humans and mice. Arterioscler Thromb Vasc Biol 23(4):529–534 Ma X, Bacci S, MlynarskiW, Gottardo L, Soccio T,Menzaghi C, Iori E, Lager RA, Shroff AR, Gervino EV, Nesto RW, Johnstone MT, Abumrad NA, Avogaro A, Trischitta V, Doria A, 2004, A common haplotype at the CD36 locus is associated with high free fatty acid levels and increased cardiovascular risk in Caucasians. Hum Mol Genet 13(19):2197–2205 Saidi S, Slamia LB, Ammou SB, Mahjoub T, Almawi WY, 2007, Association of apolipoprotein E gene polymorphism with ischemic stroke involving large-vessel disease and its relation to serum lipid levels. J Stroke Cerebrovasc Dis 16(4):160–166 Moorjani P, Patterson N, Loh P, Lipson M, Kisfali P, Melegh B, Bonin M, Kadasi L, Rieß O, Berger B, Reich D, Melegh B, 2013, Reconstructing Roma history from genome-wide data. . American Journal of Human Genetics Miller SA, Dykes DD, Polesky HF, 1988, A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 16(3):1215 Martinelli N, Trabetti E, Bassi A, Girelli D, Friso S, Pizzolo F, Sandri M, Malerba G, Pignatti PF, Corrocher R, Olivieri O, 2007, The −1131 T>C and S19WAPOA5 gene polymorphisms are associated with high levels of triglycerides and apolipoprotein C-III, but notwith coronary artery disease: an angiographic study. Atherosclerosis 191(2):409–417 Vaxillaire M, Cavalcanti-Proenca C, Dechaume A, Tichet J, Marre M, Balkau B, Froguel P, 2008, The common P446L polymorphism in GCKR inversely modulates fasting glucose and triglyceride levels and reduces type 2 diabetes risk in the DESIR prospective general French population. Diabetes 57(8):2253–2257 Hishida A, Morita E, Naito M, Okada R, Wakai K, Matsuo K, Nakamura K, Takashima N, Suzuki S, Takezaki T, Mikami H, Ohnaka K, Watanabe Y, Uemura H, Kubo M, Tanaka H, Hamajima N, 2012, Associations of apolipoprotein A5, APOA5), glucokinase, GCK, and glucokinase regulatory protein, GCKR, polymorphisms and lifestyle factors with the risk of dyslipidemia and dysglycemia in Japanese—a cross-sectional data from the J-MICC Study. Endocr J 59(7):589–599 Dussaillant C, Serrano V, Maiz A, Eyheramendy S, Cataldo LR, Chavez M, Smalley SV, Fuentes M, Rigotti A, Rubio L, Lagos CF, Martinez JA, Santos JL, 2012, APOA5 Q97X mutation identified through homozygosity mapping causes severe hypertriglyceridemia in a Chilean consanguineous family. BMC Med Genet 13:106 Warner JP, Leek JP, Intody S, Markham AF, Bonthron DT, 1995, Human glucokinase regulatory protein, GCKR): cDNA and genomic cloning, complete primary structure, and chromosomal localization. Mamm Genome 6(8):532–536 Hayward BE, Dunlop N, Intody S, Leek JP,MarkhamAF,Warner JP, Bonthron DT, 1998, Organization of the human glucokinase regulator gene GCKR. Genomics 49(1):137–142 Orho-Melander M, Melander O, Guiducci C, Perez-Martinez P, Corella D, Roos C, Tewhey R, Rieder MJ, Hall J, Abecasis G, Tai ES, Welch C, Arnett DK, Lyssenko V, Lindholm E, Saxena R, de Bakker PI, Burtt N, Voight BF, Hirschhorn JN, Tucker KL, Hedner T, Tuomi T, Isomaa B, Eriksson KF, Taskinen MR, Wahlstrand B, Hughes TE, Parnell LD, Lai CQ, Berglund G, Peltonen L, Vartiainen E, Jousilahti P, Havulinna AS, Salomaa V, Nilsson P, Groop L, Altshuler D, Ordovas JM, Kathiresan S, 2008, Common missense variant in the glucokinase regulatory protein gene is associated with increased plasma triglyceride and C-reactive protein but lower fasting glucose concentrations. Diabetes 57(11):3112–3121 Rafiq S, Venkata KK, Gupta V, Guru VD, Spurgeon CJ, Parameshwaran S, Madana SN, Kinra S, Bowen L, Timpson NJ, Smith GD, Dudbridge F, Prabhakaran D, Ben-Shlomo Y, Reddy KS, Ebrahim S, Chandak GR, 2012, Evaluation of seven common lipid associated loci in a large Indian sib pair study. Lipids Health Dis 11(1):155 Veiga-da-CunhaM, Delplanque J, Gillain A, Bonthron DT, Boutin P, Van Schaftingen E, Froguel P, 2003, Mutations in the glucokinase regulatory protein gene in 2p23 in obese French caucasians. Diabetologia 46(5):704–711 Santoro N, Zhang CK, Zhao H, Pakstis AJ, Kim G, Kursawe R, Dykas DJ, Bale AE, Giannini C, Pierpont B, Shaw MM, Groop L, Caprio S, 2012, Variant in the glucokinase regulatory protein, GCKR, gene is associated with fatty liver in obese children and adolescents. Hepatology 55(3):781–789 Pisciotta L, Favari E, Magnolo L, Simonelli S, Adorni MP, Sallo R, Fancello T, Zavaroni I, Ardigo D, Bernini F, Calabresi L, Franceschini G, Tarugi P, Calandra S, Bertolini S, 2012, Characterization of three kindreds with familial combined hypolipidemia caused by loss-of-function mutations of ANGPTL3. Circ Cardiovasc Genet 5(1):42–50 Liu Y, Zhou D, Zhang Z, Song Y, Zhang D, Zhao T, Chen Z, Sun Y, Yang Y, Xing Q, Zhao X, Xu H, He L, 2010, Effects of genetic variants on lipid parameters and dyslipidemia in a Chinese population. J Lipid Res 52(2):354–360 Vrablik M, Hubacek JA, Dlouha D, Lanska V, Rynekrova J, Zlatohlavek L, Prusikova M, Ceska R, Adamkova V, 2012, Impact of variants within seven candidate genes on statin treatment efficacy. Physiol Res Tai ES, Sim XL, Ong TH,Wong TY, Saw SM, Aung T, Kathiresan S, Orho-Melander M, Ordovas JM, Tan JT, Seielstad M, 2009, Polymorphisms at newly identified lipid-associated loci are associated with blood lipids and cardiovascular disease in an Asian Malay population. J Lipid Res 50(3):514–520 Maasz A, Kisfali P, Horvatovich K, Mohas M, Marko L, Csongei V, Farago B, Jaromi L, Magyari L, Safrany E, Sipeky C, Wittmann I, Melegh B, 2007, Apolipoprotein A5 T-1131C variant confers risk for metabolic syndrome. Pathol Oncol Res 13(3):243–247 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 743 EP 749 DI 10.1007/s12253-014-9884-5 PG 7 ER PT J AU Posfai, Marton, I Kiraly, AP Kotosz, B Kiss-Laszlo, Zs Szell, M Borbenyi, Z AF Posfai, Eva Marton, Imelda Kiraly, Attila Peter Kotosz, Balazs Kiss-Laszlo, Zsuzsanna Szell, Marta Borbenyi, Zita TI JAK2 V617F, MPL, and CALR Mutations in Essential Thrombocythaemia and Major Thrombotic Complications: A Single-Institute Retrospective Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Myeloproliferative neoplasms; Essential thrombocythaemia; JAK2 V617F mutation; MPL mutations; CALR mutations; Thrombosis ID Myeloproliferative neoplasms; Essential thrombocythaemia; JAK2 V617F mutation; MPL mutations; CALR mutations; Thrombosis AB Thrombo-haemorrhagic events are the main cause of morbidity and mortality in essential thrombocythemia. The aim of this study was to estimate the incidence of thrombotic events and the impact of the JAK2V617F, MPL (W515L, W515K, W515R, W515A and S505N) and CALR (type-1, type-2) mutations on 101 essential thrombocythaemia patients (72 females and 29 males with a mean age of 61 years) diagnosed in a Southern Hungarian regional academic centre. The incidence of major thrombosis was 13.86 %. Sixty percent of the patients carried the JAK2V617F mutation. The MPL mutations were analysed by sequencing and the W515L was the only one we could identify with an incidence of 3.96 %. Type-2 CALR mutation could be identified in 3 cases among the patients who had JAK2/MPL-unmutated ET. Statistical analyses revealed that the JAK2V617F mutation was associated with significantly increased levels of platelet (p=0.042), haemoglobin (p=0.000), red blood cell (p=0.000) and haematocrit (p=0.000) and hepatomegaly (p=0.045) at diagnosis compared to JAK2V617F negative counterparts, however there was no significant association between the JAK2V617F mutation status (relative risk: 1.297, 95 % CI 0.395–4.258; p=0.668) and subsequent thrombotic complications. The impact of JAK2V617F, MPL W515L and CALR mutations on the clinical findings at the diagnosis of ET was obvious, but their statistically significant role in the prediction of thrombotic events could not be proven in this study. Our results indirectly support the concept that, besides the quantitative and qualitative changes in the platelets, the mechanisms leading to thrombosis are more complex and multifactorial. C1 [Posfai, Eva] University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, Koranyi fasor 6, H-6720 Szeged, Hungary. [Marton, Imelda] University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, Koranyi fasor 6, H-6720 Szeged, Hungary. [Kiraly, Attila Peter] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kotosz, Balazs] University of Szeged, Institute of Economics and Rural DevelopmentSzeged, Hungary. [Kiss-Laszlo, Zsuzsanna] University of Szeged, Department of Medical GeneticsSzeged, Hungary. [Szell, Marta] University of Szeged, Department of Medical GeneticsSzeged, Hungary. [Borbenyi, Zita] University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, Koranyi fasor 6, H-6720 Szeged, Hungary. RP Posfai, (reprint author), University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, H-6720 Szeged, Hungary. EM evaposfay@gmail.com CR Tefferi A, Vardiman JW, 2008, Classification and diagnosis of myeloproliferative neoplasms: the 2008 World Health Organization criteria and point-of-care diagnostic algorithms. Leukemia 22(1): 14–22., DOI 10.1038/sj.leu.2404955 Falanga A,MarchettiM(2012, Thrombotic disease in the myeloproliferative neoplasms. Hematology Am Soc Hematol Educ Program 2012:571–581., DOI 10.1182/asheducation-2012.1.571 Tefferi A, 2012, Polycythemia vera and essential thrombocythemia: 2012 update on diagnosis, risk stratification, and management. Am J Hematol 87(3):285–293., DOI 10.1002/ajh.23135 Tefferi A, 2013, Polycythemia vera and essential thrombocythemia: 2013 update on diagnosis, risk-stratification, and management. Am J Hematol 88(6):507–516., DOI 10.1002/ajh.23417 Landolfi R, Cipriani MC, Novarese L, 2006, Thrombosis and bleeding in polycythemia vera and essential thrombocythemia: pathogenetic mechanisms and prevention. Best Pract Res Clin Haematol 19, 3):617–633., DOI 10.1016/j.beha.2005.07.011 Michiels JJ, Abels J, Steketee J, van Vliet HH, Vuzevski VD, 1985, Erythromelalgia caused by platelet-mediated arteriolar inflammation and thrombosis in thrombocythemia. Ann InternMed 102(4):466–471 Mignon I, Grand F, Boyer F, Hunault-BergerM, Hamel JF,Macchi L, 2013, Thrombin generation and procoagulant phospholipids in patients with essential thrombocythemia and reactive thrombocytosis. Am J Hematol., DOI 10.1002/ajh.23553 Posfai E, Marton I, Szoke A, Borbenyi Z, Vecsei L, Csomor A, Sas K, 2013, Stroke in essential thrombocythemia. J Neurol Sci., DOI 10. 1016/j.jns.2013.10.016 Andrikovics H, Nahajevszky S, Koszarska M, Meggyesi N, Bors A, Halm G, Lueff S, Lovas N, Matrai Z, Csomor J, Rasonyi R, Egyed M, Varkonyi J, Mikala G, Sipos A, Kozma A, Adam E, Fekete S, Masszi T, Tordai A, 2010, JAK2 46/1 haplotype analysis in myeloproliferative neoplasms and acute myeloid leukemia. Leukemia 24, 10):1809–1813., DOI 10.1038/leu.2010.172 Andrikovics H, Szilvasi A, Meggyesi N, Kiraly V, Halm G, Lueff S, Nahajevszky S, Mikala G, Sipos A, Lovas N, Csukly Z, Matrai Z, Tamaska J, TordaiA,Masszi T, 2007, Role of the activatingmutation Val617Phe of Janus kinase 2 gene in myeloproliferative diseases and significance of its detection. Orv Hetil 148(5):203–210., DOI 10.1556/ OH.2007.27860 Carobbio A, Thiele J, Passamonti F, Rumi E, Ruggeri M, Rodeghiero F, Randi ML, Bertozzi I, Vannucchi AM, Antonioli E, Gisslinger H, Buxhofer-Ausch V, Finazzi G, Gangat N, Tefferi A, Barbui T, 2011, Risk factors for arterial and venous thrombosis in WHO-defined essential thrombocythemia: an international study of 891 patients. Blood 117(22):5857–5859., DOI 10.1182/blood-2011-02-339002 Tefferi A, 2012, Polycythemia vera and essential thrombocythemia: 2012 update on diagnosis, risk stratification, and management. American Journal of Hematology 87(3):284–293., DOI 10.1002/ajh. 23135 Tefferi A, Elliott M, 2007, Thrombosis in myeloproliferative disorders: prevalence, prognostic factors, and the role of leukocytes and JAK2V617F. Semin Thromb Hemost 33(4):313–320., DOI 10.1055/s- 2007-976165 Barbui T, Finazzi G, Carobbio A, Thiele J, Passamonti F, Rumi E, Ruggeri M, Rodeghiero F, Randi ML, Bertozzi I, Gisslinger H, Buxhofer-Ausch V, De Stefano V, Betti S, Rambaldi A, Vannucchi AM, TefferiA, 2012, Development and validation of an International Prognostic Score of thrombosis in World Health Organizationessential thrombocythemia, IPSET-thrombosis). Blood 120(26): 5128–5133., DOI 10.1182/blood-2012-07-444067, quiz 5252 Tefferi A, Barbui T, 2013, Personalized management of essential thrombocythemia-application of recent evidence to clinical practice. Leukemia., DOI 10.1038/leu.2013.99 Lee HS, Park LC, Lee EM, Lee SJ, Shin SH, Im H, Do KM, Kim EJ, Ye BJ, Song MK, Kim SH, Lee SM, Lee WS, Kim YS, 2012, Incidence rates and risk factors for vascular events in patients with essential thrombocythemia: a multicenter study from Korea. Clin Lymphoma Myeloma Leuk 12(1):70–75., DOI 10.1016/j.clml.2011. 10.002 Matsumura I, Horikawa Y, Kanakura Y, 1999, Functional roles of thrombopoietin-c-mpl system in essential thrombocythemia. Leuk Lymphoma 32(3–4):351–358., DOI 10.3109/10428199909167396 Craig S. Kitchens BAK, Craig M. Kessler, ed,, 2013, Consultative hemostasis and thrombosis. Elsevier Health Sciences Tefferi A, 2010, Novel mutations and their functional and clinical relevance in myeloproliferative neoplasms: JAK2, MPL, TET2, ASXL1, CBL, IDH and IKZF1. Leukemia 24(6):1128–1138., DOI 10.1038/leu.2010.69 Teofili L, Giona F, Torti L, Cenci T, Ricerca BM, Rumi C, Nunes V, Foa R, Leone G, Martini M, Larocca LM, 2010, Hereditary thrombocytosis caused by MPLSer505Asn is associated with a high thrombotic risk, splenomegaly and progression to bone marrow fibrosis. Haematologica 95(1):65–70., DOI 10.3324/haematol.2009. 007542 Pikman Y, Lee BH, Mercher T, McDowell E, Ebert BL, Gozo M, Cuker A, Wernig G, Moore S, Galinsky I, DeAngelo DJ, Clark JJ, Lee SJ, Golub TR, Wadleigh M, Gilliland DG, Levine RL, 2006, MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med 3(7):e270., DOI 10.1371/journal. pmed.0030270 Pardanani AD, Levine RL, Lasho T, Pikman Y, Mesa RA,Wadleigh M, Steensma DP, Elliott MA, Wolanskyj AP, Hogan WJ, McClure RF, Litzow MR, Gilliland DG, Tefferi A, 2006, MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood 108(10):3472–3476., DOI 10.1182/blood-2006-04- 018879 Klampfl T, Gisslinger H, Harutyunyan AS, Nivarthi H, Rumi E, Milosevic JD, Them NC, Berg T, Gisslinger B, Pietra D, Chen D, Vladimer GI, Bagienski K, Milanesi C, Casetti IC, Sant’Antonio E, Ferretti V, Elena C, Schischlik F, Cleary C, Six M, Schalling M, Schonegger A, Bock C, Malcovati L, Pascutto C, Superti-Furga G, Cazzola M, Kralovics R, 2013, Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 369(25):2379–2390., DOI 10.1056/NEJMoa1311347 Tefferi A, Wassie EA, Guglielmelli P, Gangat N, Belachew AA, Lasho TL, Finke C, Ketterling RP, Hanson CA, Pardanani A, Wolanskyj AP, Maffioli M, Casalone R, Pacilli A, Vannucchi AM, Passamonti F, 2014, Type 1 versus Type 2 calreticulin mutations in essential thrombocythemia: a collaborative study of 1027 patients. Am J Hematol 89(8):E121–E124., DOI 10.1002/ajh.23743 Chen CC, Gau JP, Chou HJ, You JY, Huang CE, Chen YY, Lung J, Chou YS, Leu YW, Lu CH, Lee KD, Tsai YH, 2014, Frequencies, clinical characteristics, and outcome of somatic CALR mutations in JAK2-unmutated essential thrombocythemia. Ann Hematol., DOI 10. 1007/s00277-014-2151-8 Hungarian Central Statistical Office -Population census., 2011). Regional data-Csongrad county http://www.ksh.hu/nepszamlalas/ tables_regional_03; Regional data- Bekes county http://www.ksh. hu/nepszamlalas/tables_regional_04; Regional data - Csongrad county http://www.ksh.hu/nepszamlalas/tables_regional_06 Barbui T, BarosiG,Grossi A,Gugliotta L, Liberato LN,Marchetti M, Mazzucconi MG, Rodeghiero F, Tura S, 2004, Practice guidelines for the therapy of essential thrombocythemia. A statement from the Italian Society of Hematology, the Italian Society of Experimental Hematology and the Italian Group for BoneMarrow Transplantation. Haematologica 89(2):215–232 Finazzi G, Barbui T, 2005, Risk-adapted therapy in essential thrombocythemia and polycythemia vera. Blood Rev 19(5):243– 252., DOI 10.1016/j.blre.2005.01.001 Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin N, Scott MA, ErberWN, Green AR, Cancer Genome P, 2005, Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 365(9464):1054–1061., DOI 10.1016/S0140-6736(05)71142-9 Bergamaschi GM, Primignani M, Barosi G, Fabris FM, Villani L, Reati R, Dell’era A, Mannucci PM, 2008, MPL and JAK2 exon 12 mutations in patients with the Budd-Chiari syndrome or extrahepatic portal vein obstruction. Blood 111(8):4418., DOI 10.1182/blood-2008- 02-137687 AB.Hill, ed,, 1984: 170, A short textbook of medical statistics. Hodder and Stoughton, London Beer PA, Campbell PJ, Scott LM, Bench AJ, ErberWN, Bareford D, Wilkins BS, Reilly JT, Hasselbalch HC, Bowman R, Wheatley K, Buck G, Harrison CN, Green AR, 2008, MPL mutations in myeloproliferative disorders: analysis of the PT-1 cohort. Blood 112(1): 141–149., DOI 10.1182/blood-2008-01-131664 Hsiao HH, YangMY, Liu YC, Lee CP, YangWC, Liu TC, Chang CS, Lin SF, 2007, The association of JAK2V617F mutation and leukocytosis with thrombotic events in essential thrombocythemia. Exp Hematol 35(11):1704–1707., DOI 10.1016/j.exphem.2007.08.011 Hobbs CM, Manning H, Bennett C, Vasquez L, Severin S, Brain L, Mazharian A, Guerrero JA, Li J, Soranzo N, Green AR, Watson SP, Ghevaert C, 2013, JAK2V617F leads to intrinsic changes in platelet formation and reactivity in a knock-in mouse model of essential thrombocythemia. Blood., DOI 10.1182/blood-2013-06-501452 Vannucchi AM, Barbui T, 2007, Thrombocytosis and thrombosis. Hematology Am Soc Hematol Educ Program:363–370., DOI 10.1182/ asheducation-2007.1.363 Ivanyi JL, Marton E, Plander M, 2011, Significance of the JAK2V617F mutation in patients with chronic myeloproliferative neoplasia. Orv Hetil 152(45):1795–1803., DOI 10.1556/OH.2011. 29226 Cho YU, Chi HS, Lee EH, Jang S, Park CJ, Seo EJ, 2009, Comparison of clinicopathologic findings according to JAK2 V617F mutation in patients with essential thrombocythemia. Int J Hematol 89(1):39–44., DOI 10.1007/s12185-008-0222-0 Andrikovics H, Meggyesi N, Szilvasi A, Tamaska J, Halm G, Lueff S, Nahajevszky S, Egyed M, Varkonyi J, Mikala G, Sipos A, Kalasz L, Masszi T, Tordai A, 2009, HFE C282Y mutation as a genetic modifier influencing disease susceptibility for chronic myeloproliferative disease. Cancer Epidemiol Biomarkers Prev 18(3):929–934., DOI 10.1158/1055-9965.EPI-08-0359 Tefferi A, Wassie EA, Lasho TL, Finke C, Belachew AA, Ketterling RP, Hanson CA, Pardanani A, Gangat N, Wolanskyj AP, 2014, Calreticulin mutations and long-term survival in essential thrombocythemia. Leukemia., DOI 10.1038/leu.2014.148 Andrikovics H, Krahling T, Balassa K, Halm G, Bors A, Koszarska M, Batai A, Dolgos J, Csomor J, Egyed M, Sipos A, Remenyi P, Tordai A, Masszi T, 2014, Distinct clinical characteristics of myeloproliferative neoplasms with calreticulin mutations. Haematologica 99(7):1184–1190., DOI 10.3324/haematol.2014.107482 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 751 EP 758 DI 10.1007/s12253-014-9885-4 PG 8 ER PT J AU Shinmura, K Kato, H Igarashi, H Inoue, Y Nakamura, S Du, Ch Kurachi, K Nakamura, T Ogawa, H Tanahashi, M Niwa, H Sugimura, H AF Shinmura, Kazuya Kato, Hisami Igarashi, Hisaki Inoue, Yusuke Nakamura, Satoki Du, Chunping Kurachi, Kiyotaka Nakamura, Toshio Ogawa, Hiroshi Tanahashi, Masayuki Niwa, Hiroshi Sugimura, Haruhiko TI CD44-SLC1A2 Fusion Transcripts in Primary Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD44-SLC1A2; Fusion transcript; Colorectal cancer ID CD44-SLC1A2; Fusion transcript; Colorectal cancer AB A CD44-SLC1A2 fusion has recently been discovered in a subset of primary gastric cancers, and an APIPSLC1A2 fusion has been described in a colon cancer cell line (SNU-C1); however, whether such SLC1A2 fusions occur in primary colorectal cancer (CRC) and whether such fusions are specific for gastrointestinal cancers remain uncertain. In the present study, we examined 90 primary CRCs and 112 primary non-small cell lung cancers (NSCLCs) for CD44-SLC1A2 and APIP-SLC1A2 fusion transcripts using RT-PCR and subsequent sequencing analyses. Although the expression of both types of SLC1A2 fusion transcripts was not detected in any of the NSCLCs, the expression of CD44-SLC1A2, but not the APIP-SLC1A2 fusion transcript, was detected in one (1.1 %) CRC. The CD44-SLC1A2 fusion transcript was expressed in cancerous tissue but not in corresponding non-cancerous tissue, and the fusion occurred between exon 1 of CD44 and exon 2 of SLC1A2; it was expected that a slightly truncated but functional SLC1A2 protein would be produced under the CD44 promoter. A quantitative RT-PCR analysis revealed that SLC1A2 mRNA expression was upregulated in CRC containing SLC1A2 fusion transcripts, while it was downregulated in most other CRCs. The SLC1A2 fusion-positive carcinoma was located on the right-side of colon,was amucinous adenocarcinoma, was immunohistochemically negative for MSH2 mismatch repair protein, and contained no APC or KRAS mutations. Together, these results suggest that the expression of SLC1A2 fusion transcripts is related to a subset of primary CRCs and may contribute to the elucidation of the characteristics of SLC1A2 fusion-positive CRCs in the future. C1 [Shinmura, Kazuya] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Shizuoka, Japan. [Kato, Hisami] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Shizuoka, Japan. [Igarashi, Hisaki] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Shizuoka, Japan. [Inoue, Yusuke] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Shizuoka, Japan. [Nakamura, Satoki] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Shizuoka, Japan. [Du, Chunping] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Shizuoka, Japan. [Kurachi, Kiyotaka] Hamamatsu University School of Medicine, Department of Surgery 2Hamamatsu, Japan. [Nakamura, Toshio] Hamamatsu University School of Medicine, Department of Surgery 2Hamamatsu, Japan. [Ogawa, Hiroshi] Seirei Mikatahara General Hospital, Division of PathologyHamamatsu, Japan. [Tanahashi, Masayuki] Seirei Mikatahara General Hospital, Respiratory Disease Center, Division of Thoracic SurgeryHamamatsu, Japan. [Niwa, Hiroshi] Seirei Mikatahara General Hospital, Respiratory Disease Center, Division of Thoracic SurgeryHamamatsu, Japan. [Sugimura, Haruhiko] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Shizuoka, Japan. RP Shinmura, K (reprint author), Hamamatsu University School of Medicine, Department of Tumor Pathology, 431-3192 Hamamatsu, Japan. EM kzshinmu@hama-med.ac.jp CR Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S,Watanabe H, Kurashina K, Hatanaka H, BandoM, Ohno S, Ishikawa Y, Aburatani H, Niki T, Sohara Y, Sugiyama Y, Mano H, 2007, Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 448:561–566 Rikova K, Guo A, Zeng Q, Possemato A, Yu J, Haack H, Nardone J, Lee K, Reeves C, Li Y, Hu Y, Tan Z, Stokes M, Sullivan L, Mitchell J, Wetzel R, Macneill J, Ren JM, Yuan J, Bakalarski CE, Villen J, Kornhauser JM, Smith B, Li D, Zhou X, Gygi SP, Gu TL, Polakiewicz RD, Rush J, Comb MJ, 2007, Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 131:1190–1203 Shinmura K, Kageyama S, Tao H, Bunai T, Suzuki M, Kamo T, Takamochi K, Suzuki K, Tanahashi M, Niwa H, Ogawa H, Sugimura H, 2008, EML4-ALK fusion transcripts, but no NPM-, TPM3-, CLTC-, ATIC-, or TFG-ALK fusion transcripts, in nonsmall cell lung carcinomas. Lung Cancer 61:163–169 Matsuura S, Shinmura K, Kamo T, Igarashi H,Maruyama K, Tajima M, Ogawa H, Tanahashi M, Niwa H, Funai K, Kohno T, Suda T, Sugimura H, 2013, CD74-ROS1 fusion transcripts in resected nonsmall cell lung carcinoma. Oncol Rep 30:1675–1680 Zheng J, 2013, Oncogenic chromosomal translocations and human cancer. Oncol Rep 30:2011–2019 Bittner N, Ostoros G, Geczi L, 2014, New treatment options for lung adenocarcinoma–in view of molecular background. Pathol Oncol Res 20:11–25 Mitsudomi T, Suda K, Yatabe Y, 2013, Surgery for NSCLC in the era of personalized medicine. Nat Rev Clin Oncol 10:235–244 Tao J, Deng NT, Ramnarayanan K, Huang B, Oh HK, Leong SH, LimSS, Tan IB,Ooi CH,Wu J, LeeM, Zhang S, Rha SY, Chung HC, Smoot DT, Ashktorab H, Kon OL, Cacheux V, Yap C, Palanisamy N, Tan P, 2011, CD44-SLC1A2 gene fusions in gastric cancer. Sci Transl Med 3:77ra30 Giacomini CP, Sun S, Varma S, Shain AH, GiacominiMM, Balagtas J, Sweeney RT, Lai E, Del Vecchio CA, Forster AD, Clarke N, Montgomery KD, Zhu S, Wong AJ, van de Rijn M, West RB, Pollack JR, 2013, Breakpoint analysis of transcriptional and genomic profiles uncovers novel gene fusions spanning multiple human cancer types. PLoS Genet 9:e1003464 Shinmura K, Goto M, Suzuki M, Tao H, Yamada H, Igarashi H, Matsuura S, Maeda M, Konno H, Matsuda T, Sugimura H, 2011, Reduced expression of MUTYH with suppressive activity against mutations caused by 8-hydroxyguanine is a novel predictor of a poor prognosis in human gastric cancer. J Pathol 225:414–423 Tao H, Shinmura K, Yamada H, Maekawa M, Osawa S, Takayanagi Y, Okamoto K, Terai T, Mori H, Nakamura T, Sugimura H, 2010, Identification of 5 novel germline APC mutations and characterization of clinical phenotypes in Japanese patients with classical and attenuated familial adenomatous polyposis. BMC Res Notes 3:305 Fujisaki T, Tanaka Y, Fujii K, Mine S, Saito K, Yamada S, Yamashita U, Irimura T, Eto S, 1999, CD44 stimulation induces integrinmediated adhesion of colon cancer cell lines to endothelial cells by up-regulation of integrins and c-Met and activation of integrins. Cancer Res 59:4427–4434 Lugli A, Tzankov A, Zlobec I, Terracciano LM, 2008, Differential diagnostic and functional role of the multi-marker phenotype CDX2/CK20/CK7 in colorectal cancer stratified by mismatch repair status. Mod Pathol 21:1403–1412 Marcus VA, Madlensky L, Gryfe R, Kim H, So K, Millar A, Temple LK, Hsieh E, Hiruki T, Narod S, Bapat BV, Gallinger S, Redston M, 1999, Immunohistochemistry for hMLH1 and hMSH2: a practical test for DNA mismatch repair-deficient tumors. Am J Surg Pathol 23: 1248–1255 Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, Nakagawa H, Sotamaa K, Prior TW,Westman J, Panescu J, Fix D, Lockman J, Comeras I, de la Chapelle A, 2005, Screening for the Lynch syndrome, hereditary nonpolyposis colorectal cancer). N Engl J Med 352:1851–1860 Kelleher FC, McDermott R, 2010, The emerging pathogenic and therapeutic importance of the anaplastic lymphoma kinase gene. Eur J Cancer 46:2357–2368 Takeuchi K, Soda M, Togashi Y, Suzuki R, Sakata S, Hatano S, Asaka R, Hamanaka W, Ninomiya H, Uehara H, Lim Choi Y, Satoh Y, Okumura S, Nakagawa K, Mano H, Ishikawa Y, 2012, RET, ROS1 and ALK fusions in lung cancer. Nat Med 18:378–381 Dang CV, 2010, Rethinking the Warburg effect with Myc micromanaging glutamine metabolism. Cancer Res 70:859–862 Willard SS, Koochekpour S, 2013, Glutamate signaling in benign and malignant disorders: current status, future perspectives, and therapeutic implications. Int J Biol Sci 9:728–742 Kakar S, Aksoy S, Burgart LJ, Smyrk TC, 2004, Mucinous carcinoma of the colon: correlation of loss of mismatch repair enzymes with clinicopathologic features and survival. Mod Pathol 17:696–700 Terui H, Tachikawa T, Kakuta M, Nishimura Y, Yatsuoka T, Yamaguchi K, Yura K, Akagi K, 2013, Molecular and clinical characteristics of MSH6 germline variants detected in colorectal cancer patients. Oncol Rep 30:2909–2916 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 759 EP 764 DI 10.1007/s12253-014-9887-2 PG 6 ER PT J AU Naruse, T Yanamoto, S Yamada, Shi Rokutanda, S Kawakita, A Kawasaki, G Umeda, M AF Naruse, Tomofumi Yanamoto, Souichi Yamada, Shin-ichi Rokutanda, Satoshi Kawakita, Akiko Kawasaki, Goro Umeda, Masahiro TI Anti-Tumor Effect of the Mammalian Target of Rapamycin Inhibitor Everolimus in Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral cancer; mTOR; Everolimus; Hypoxia ID Oral cancer; mTOR; Everolimus; Hypoxia AB The mammalian target of rapamycin (mTOR) has recently emerged as a promising target for therapeutic anti-cancer interventions in several human tumors. In present study, we investigated the expression of mTOR, and subsequently examined its relationship with clinicopathological factors and the anti-tumor effect of everolimus (also known as RAD001) in oral squamous cell carcinoma (OSCC). The expression of phosphorylated mTOR (p-mTOR) was immunohistochemically evaluated in specimens obtained from 70 OSCC patients who underwent radical surgery. The relationships between the expression of p-mTOR and clinicopathological factors and survival were determined. We also investigated the effect of everolimus on the OSCC cell lines, SAS, HSC- 2, HSC-3, HSC-4, OSC-20, SCC25 and Ca9-22 by the MTT assay. We further evaluated whether mTOR contributed to cell functions by blocking its activity with everolimus, and confirmed the direct target by the Matrigel invasion assay, wound healing assay and Western blotting. p-mTOR was overexpressed in 37 tumors (52.8 %), and correlated with the T classification, N classification, and survival rate (P<0.05). The treatment with everolimus significantly inhibited cell growth, and significantly reduced the expression of p-mTOR, downstream signaling proteins, and hypoxic related proteins as well as invasion and migration potentials (P<0.05). The results of the present study suggest that everolimus may represent an attractive approach for the future treatment of OSCC. C1 [Naruse, Tomofumi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yanamoto, Souichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yamada, Shin-ichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Rokutanda, Satoshi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Kawakita, Akiko] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Kawasaki, Goro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Umeda, Masahiro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. RP Naruse, T (reprint author), Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 852-8588 Nagasaki, Japan. EM naruse12@nagasaki-u.ac.jp CR Mao L, Hong WK, Papadimitrakopoulou VA, 2004, Focus on head and neck cancer. Cancer Cell 5:311–6 Seiwert TY, Cohen EE, 2005, State-of-the-artmanagement of locally advanced head and neck cancer. Br J Cancer 92(8):1341–8 Rogers SN, Brown JS, Woolgar JA et al, 2009, Survival following primary surgery for oral cancer. Oral Oncol 45(3):201–11 Shaw RJ, Cantley LC, 2006, Ras, PI, 3, K and mTOR signaling controls tumor cell growth. Nature 441(7092):424–30 Liu FY, Zhao ZJ, Li P et al, 2010, Mammalian target of rapamycin, mTOR, is involved in the survival of cells mediated by chemokine receptor 7 through PI3K/Akt in metastatic squamous cell carcinoma of the head and neck. Br J Oral Maxillofac Surg 48:291–6 Semenza GL, 2003, Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3:721–32 Laplante M, Sabatini DM, 2012, mTOR signaling in growth control and disease. Cell 149(2):274–93 Pouyssegur J, Dayan F, Mazure NM, 2006, Hypoxia signaling in cancer and approaches to enforce tumour regression. Nature 441: 437–43 Hohwer N, Cramer T, 2011, Hypoxia-mediated drug resistance: novel insights on the functional interaction of HIFs and cell death pathways. Drug Resist Updat 14:191–201 Brown LM, Cowen RL, Debray C et al, 2006, Reversing hypoxic cell chemoresistance in vitro using genetic and small molecule approaches targeting hypoxia inducible factor-1. Mol Pharmacol 69: 411–8 Liu L, Ning X, Sun L et al, 2008, Hypoxia-inducible factor-1 alpha contributes to hypoxia-induced chemoresistance in gastric cancer. Cancer Sci 99:121–8 Yoshiba S, Ito D, Nagumo T et al, 2009, Hypoxia induces resistance to 5-fluorouracil in oral cancer cells via G, 1, phase cell cycle arrest. Oral Oncol 45:109–15 Monteiro LS, Delgado ML, Ricardo S et al, 2013, Phosphorylated mammalian target of rapamycin is associated with an adverse outcome in oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 115(5):638–45 Hirashima K, Baba Y, Watanabe M et al, 2010, Phosphorylated mTOR expression is associated with poor prognosis for patients with esophageal squamous cell carcinoma. Ann Surg Oncol 17(9):2486– 93 Bakarakos P, Theohari I , Nomikos A e t a l, 2010, Immunohistochemical study of PTEN and phosphorylated mTOR proteins in familial and sporadic invasive breast carcinomas. Histopathology 56(7):876–82 Xu DZ, Geng QR, Tian Yet al, 2010, Activated mammalian target of rapamycin is a potential therapeutic target in gastric cancer. BMC Cancer 10:536 Del Bufalo D, Ciuffreda L, Trisciuoqlio D et al, 2006, Antiangiogenic potential of the Mammalian target of rapamycin inhibitor temsirolimus. Cancer Res 66:5549–54 Matsumoto K, Arao T, Tanaka K et al, 2009, mTOR signal and hypoxia-inducible factor-1 alpha regulate CD133 expression in cancer cells. Cancer Res 69:7160–4 Okui T, Shimo T, Fukazawa T et al, 2010, Antitumor effect of temsirolimus against oral squamous cell carcinoma associated with bone destruction. Mol Cancer Ther 9:2960–9 Okui T, Shimo T, Fukazawa T et al, 2013, Novel HSP90 inhibitor NVP-AUY922 enhances the anti-tumor effect of temsirolimus against oral squamous cell carcinoma. Curr Cancer Drug Targets 13:289–99 Huang S, Houghton PJ, 2003, Targeting mTOR signaling for cancer therapy. Curr Opin Pharmacol 3:371–7 Motzer RJ, Escudier B, Oudard S et al, 2008, Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomized, placebo-controlled phase III trial. Lancet 372:449–56 Pavel ME, Hainsworth JD, Baudin E et al, 2011, Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome, RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 378:2005–12 Burris HA 3rd, Lebrun F, RugoHS et al, 2013, Health-related quality of life of patients with advanced breast cancer treatedwith everolimus plus exmestane versus placebo plus exemestane in the phase 3, randomizd, controlled, BOLERO-2 trial. Cancer 119(10):1908–15 Fury MG, Lee NY, Sherman E et al, 2013, A phase 1 study of everolimus+weekly cisplatin+intensity modulated radiation therapy in head and neck cancer. Int J Radiat Oncol Biol Phys 87(3):479–86 Clark C, Shah S, Herman-Ferdinandez L et al, 2010, Teasing out the best molecular marker in the AKT/mTOR pathway in head and neck squamous cell cancer patients. Laryngoscope 20(6):1159–65 Brown RE, Zhang PL, Lun M et al, 2006, Morphoproteomic and pharmacoproteomic rationale for mTOR effectors as therapeutic targets in head and neck squamous cell carcinoma. Ann Clin Lab Sci 36(3):273–82 Myoung H, Kim MJ, Lee JH et al, 2006, Correlation of proliferative markers, Ki-67 and PCNA, with survival and lymph node metastasis in oral squamous cell carcinoma: a clinical and histopathological analysis of 113 patients. Int J Oral Maxillofac Surg 35(11):1005–10 Dorevic G, Matusan-Ilijas K, Babarovic E et al, 2009, Hypoxia inducible factor-1alpha correlates with vascular endothelial growth factor A and C indicating worse prognosis in clear cell renal cell carcinoma. J Exp Clin Cancer Res 20:28–40 Cao D, Hou M, Guan YS et al, 2009, Expression of HIF-1alpha and VEGF in colorectal cancer: association with clinical outcomes and prognostic implications. BMC Cancer 9:432 Oh SY, Kwon HC, Kim SH et al, 2008, Clinicopathologic significance of HIF-1alpha, p53, and VEGF expression and preoperative serum VEGF level in gastric cancer. BMC Cancer 8:123 Juengel E, Engler J, Natsheh I et al, 2009, Combining the receptor tyrosine kinase inhibitor AEE788 and the mammalian target of rapamycin, mTOR, inhibitor RAD001 strongly inhibits adhesion and growth of renal cell carcinoma cells. BMC Cancer 9:161 Zitzmann K, De Toni EN, Brand S et al, 2007, The novel mTOR inhibitor RAD001, everolimus, induces antiproliferative effects in human pancreatic neuroendocrine tumor cells. Neuroendocrinology 85(1):54–60 Grozinsky-Glasberg S, Rubinfeld H, Nordenberg Yet al, 2010, The rapamycin-derivative RAD001, everolimus, inhibits cell viability and interacts with the Akt-mTOR-p70S6K pathway in human medullary thyroid carcinoma cells. Mol Cell Endocrinol 315:87–94 Nishi T, IwasakiK,OhashiNet al, 2013, Phosphorylation of 4E-BP1 predicts sensitivity to everolimus in gastric cancer cells. Cancer Lett 331(2):220–9 Miyazawa M, Yasuda M, Fujita M et al, 2009, Therapeutic strategy targeting the mTOR-HIF-1alpha-VEGF pathway in ovarian clear cell adenocarcinoma. Pathol Int 59:19–27 Naruse T, Kawasak i G, Yanamoto S et al, 2011, Immunohistochemical study of VEGF expression in oral squamous cell carcinomas: correlation with the mTOR- HIF-1α pathway. Anticancer Res 12:4429–37 Otrock ZK,Makarem JA, Shamseddine AI, 2007, Vascular endothelial growth factor family of ligands and receptors: review. Blood Cells Mol Dis 38(3):258–68 Sobin LH, Wittekind C, 2002, UICC TNM classification of malignant tumours, 6th edn. Wiley, New York Pinborg JJ, Reichart PA, Smith CJ et al, eds,, 1997, World health organization histological typing of cancer and precancer of the oral mucosa, 2nd edn. Springer, New York Bryne M, Boysen M, Alfsen CG et al, 1998, The invasive front of carcinomas. the most important area for tumour prognosis? Anticancer Res 18(6B):4757–64 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 765 EP 773 DI 10.1007/s12253-014-9888-1 PG 9 ER PT J AU Vrdoljak, E Geczi, L Mardiak, J Ciuleanu, TE Leyman, S Zhang, K Sajben, P Torday, L AF Vrdoljak, Eduard Geczi, Lajos Mardiak, Jozef Ciuleanu, Tudor-Eliade Leyman, Sophie Zhang, Ke Sajben, Peter Torday, Laszlo TI Central and Eastern European Experience with Sunitinib in Metastatic Renal Cell Carcinoma: A Sub-analysis of the Global Expanded-Access Trial SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Renal cell carcinoma; Metastatic; Sunitinib; Expanded-access ID Renal cell carcinoma; Metastatic; Sunitinib; Expanded-access AB A global, open-label, expanded-access trial (EAT) provided sunitinib treatment on a compassionate-use basis to patients with metastatic renal cell carcinoma (mRCC) between 2005 and 2011. This retrospective analysis examines outcomes in patients from Central and East European (CEE) countries participating in the global EAT. Sunitinib (starting dose 50 mg orally once daily, with dose reduction for toxicity) was administered in repeated 6-week cycles (4 weeks on and 2 weeks off) until occurrence of disease progression or unacceptable toxicity. Tumor assessments were guided by Response Evaluation Criteria in Solid Tumors (RECIST) criteria but were performed according to local standards of care. In total, 401 CEE patients received sunitinib (median treatment duration 9.6 months), of whom 378 were evaluable for tumor response. The most frequent grade ≥3 toxicities were fatigue (7.5 %), hypertension (7.0 %), thrombocytopenia (6.5 %), diarrhea (4.2 %), nausea and hand-foot syndrome (both 3.7 %) and neutropenia (3.0 %). Median overall survival was 30.7 months (95 % CI 23.3, months). Overall survival tended to be longer in cytokine-naive than cytokineexperienced patients (median 60.8 vs. 27.5 months; P= 0.1324). Among patients with evaluable tumors, 4.0 % achieved a complete and 14.6 % a partial response [objective response rate (ORR) 18.5 % (95 % CI 14.7, 22.8 %)].Median progression-free survival was 11.6 months (95 % CI 10.3, 12.8 months). Sunitinib demonstrates safety and effectiveness in real-world mRCC patients in CEE countries. Expandedaccess program patients showed a lower tumor response rate but similar survival outcomes to patients in the pivotal Phase III clinical trial of sunitinib in mRCC. C1 [Vrdoljak, Eduard] University of Split, School of Medicine, Clinical Hospital Center Split, Department of Oncology, Spinciceva 1, 21000 Split, Croatia. [Geczi, Lajos] National Institute of OncologyBudapest, Hungary. [Mardiak, Jozef] National Cancer Institute, Department of Medical OncologyBratislava, Slovakia. [Ciuleanu, Tudor-Eliade] University of Medicine and Pharmacy Iuliu Hatieganu, Institute of Oncology Ion ChiricutaCluj-Napoca, Romania. [Leyman, Sophie] Pfizer Inc.Brussels, Belgium. [Zhang, Ke] Pfizer Inc.La Jolla, CA, USA. [Sajben, Peter] Pfizer Inc.New York, NY, USA. [Torday, Laszlo] University of Szeged, Department of OncotherapySzeged, Hungary. RP Vrdoljak, E (reprint author), University of Split, School of Medicine, Clinical Hospital Center Split, Department of Oncology, 21000 Split, Croatia. EM edo.vrdoljak@gmail.com CR Mena AC, Pulido EG, Guillen-Ponce C. Understanding the molecular-based mechanism of action of the tyrosine kinase inhibitor: sunitinib. Anti-Cancer Drugs 21 Suppl. 1: S3-S11., DOI 10.1097/ 01.cad.0000361534.44052.c5 Escudier B, Eisen T, Porta C, Patard JJ, KhooV, Algaba F,Mulders P, Kataja V, 2012, Renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 23(7):vii65–vii71., DOI 10.1093/annonc/mds227 Patard JJ, Pignot G, Escudier B, Eisen T, Bex A, Sternberg C, Rini B, Roigas J, Choueiri T, Bukowski R, Motzer R, Kirkali Z, Mulders P, Bellmunt J, 2011, ICUD-EAU International consultation on kidney cancer 2010: treatment of metastatic disease. Eur Urol 60(4):684– 690., DOI 10.1016/j.eururo.2011.06.017 Motzer RJ, Hutson TE, Tomczak P,MichaelsonMD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen I, Bycott PW, Baum CM, Figlin RA, 2007, Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 356(2):115–124., DOI 10.1056/NEJMoa065044 Motzer RJ, Hutson TE, Tomczak P,MichaelsonMD, Bukowski RM, Oudard S, Negrier S, Szczylik C, Pili R, Bjarnason GA, Garcia-del- Muro X, Sosman JA, Solska E, Wilding G, Thompson JA, Kim ST, Chen I, Huang X, Figlin RA, 2009, Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J Clin Oncol 27(22):3584–3590., DOI 10.1200/JCO.2008.20.1293 GoreME, Szczylik C, Porta C, Bracarda S, BjarnasonGA, Oudard S, Hariharan S, Lee SH, Haanen J, Castellano D, Vrdoljak E, Schoffski P, Mainwaring P, Nieto A, Yuan J, Bukowski R, 2009, Safety and efficacy of sunitinib for metastatic renal-cell carcinoma: an expanded-access trial. Lancet Oncol 10(8):757–763., DOI 10.1016/ S1470-2045(09)70162-7 Gore ME, Porta C, Bracarda S, Bjarnason GA, Oudard S, Lee S, Crino L, Kim TM, Fly K, Szczylik C, 2012, Sunitinib global expanded-access trial in metastatic renal cell carcinoma, mRCC): final results. Ann Oncol 23(9):271, Abstract 820P Curado MP, Edwards B, Shin HR, Storm H, Ferlay J, Heanue M, Boyle P, 2007). In: Cancer Incidence in Five Continents Vol. IX, vol Scientific Publication No. 160. IARC, Lyon, France Levi F, Lucchini F, Negri E, Boyle P, La Vecchia C, 2004, Cancer mortality in Europe, 1995-1999, and an overview of trends since 1960. Int J Cancer 110(2):155–169., DOI 10.1002/ijc.20097 Levi F, Lucchini F, Negri E, Zatonski W, Boyle P, La Vecchia C, 2004, Trends in cancer mortality in the European Union and accession countries, 1980-2000. Ann Oncol 15(9):1425–1431., DOI 10. 1093/annonc/mdh346 Berrino F, De Angelis R, Sant M, Rosso S, Bielska-Lasota M, Coebergh JW, Santaquilani M, 2007, Survival for eight major cancers and all cancers combined for European adults diagnosed in 1995-99: results of the EUROCARE-4 study. Lancet Oncol 8(9): 773–783., DOI 10.1016/S1470-2045(07)70245-0 Castellano D, DuhMS, Korves C, Suthoff ED, Neary M, Hernandez Pastor LJ, Bellmunt J, 2013, Safety and treatment patterns of angiogenesis inhibitors in patients with advanced renal cell carcinoma in Spain. Expert Opin Drug Saf 12(4):455–463., DOI 10.1517/14740338. 2013.781581 Porta C, Paglino C, Imarisio I, Canipari C, Chen K, Neary M, Duh MS, 2011, Safety and treatment patterns of multikinase inhibitors in patients with metastatic renal cell carcinoma at a tertiary oncology center in Italy. BMC Cancer 11:105., DOI 10.1186/1471-2407-11-105 Ansari J, Fatima A, Fernando K, Collins S, James ND, Porfiri E, 2010, Sunitinib in patients with metastatic renal cell carcinoma: Birmingham experience. Oncol Rep 24(2):507–510 Hess G, Borker R, Fonseca E, 2013, Treatment patterns: targeted therapies indicated for first-line management of metastatic renal cell carcinoma in a real-world setting. Clin Genitourin Cancer 11(2):161– 167., DOI 10.1016/j.clgc.2012.10.003 Filson CP, Redman BG, Dunn RL, Miller DC, 2011, Initial patterns of care with oral targeted therapies for patients with renal cell carcinoma. Urology 77(4):825–830., DOI 10.1016/j.urology.2010.11.003 Choueiri TK, Duh MS, Clement J, Brick AJ, Rogers MJ, Kwabi C, Shah K, Percy AG, Antras L, Jayawant SS, Chen K,Wang ST, Luka A, Neary MP, McDermott D, OhWK, 2010, Angiogenesis inhibitor therapies for metastatic renal cell carcinoma: effectiveness, safety and treatment patterns in clinical practice-based on medical chart review. BJU Int 105(9):1247–1254., DOI 10.1111/j.1464-410X.2009.08972.x Feinberg BA, Jolly P,Wang ST, Fortner B, Scott J, Gilmore J, Neary MP, Duh MS, 2012, Safety and treatment patterns of angiogenesis inhibitors in patients with metastatic renal cell carcinoma: evidence from US community oncology clinics. Med Oncol 29(2):786–794., DOI 10.1007/s12032-011-9922-z Hong MH, Kim HS, Kim C, Ahn JR, Chon HJ, Shin SJ, Ahn JB, Chung HC, Rha SY, 2009, Treatment outcomes of sunitinib treatment in advanced renal cell carcinoma patients: a single cancer center experience in Korea. Cancer Res Treat 41(2):67–72., DOI 10. 4143/crt.2009.41.2.67 Schmidinger M, Larkin J, Ravaud A, 2012, Experience with sunitinib in the treatment ofmetastatic renal cell carcinoma. Ther Adv Urol 4(5):253–265., DOI 10.1177/1756287212454933 Heck JE, Charbotel B, Moore LE, Karami S, Zaridze DG, Matveev V, Janout V, Kollarova H, Foretova L, Bencko V, Szeszenia- Dabrowska N, Lissowska J, Mates D, Ferro G, Chow WH, Rothman N, Stewart P, Brennan P, Boffetta P, 2010, Occupation and renal cell cancer in central and Eastern Europe. Occup Environ Med 67(1):47–53., DOI 10.1136/oem.2009.046250 Moore LE, Hung R, Karami S, Boffetta P, Berndt S, Hsu CC, Zaridze D, Janout V, Kollarova H, Bencko V, Navratilova M, Szeszenia- Dabrowska N, Mates D, Mukeria A, Holcatova I, Yeager M, Chanock S, Garcia-Closas M, Rothman N, Chow WH, Brennan P, 2008, Folate metabolism genes, vegetable intake and renal cancer risk in central Europe. Int J Cancer 122(8):1710–1715., DOI 10.1002/ ijc.23318 Fabianova E, Szeszenia-Dabrowska N, Kjaerheim K, Boffetta P, 1999, Occupational cancer in central European countries. Environ Health Perspect 107(Suppl 2):279–282 Dobrossy L, 2002, Cancer mortality in central-eastern Europe: facts behind the figures. Lancet Oncol 3(6):374–381 Antunes JL, Toporcov TN, de Andrade FP, 2003, Trends and patterns of cancer mortality in European countries. Eur J Cancer Prev 12(5):367–372., DOI 10.1097/01.cej.0000090182.08740.97 Zatonski W, Didkowska J, 2008, Closing the gap: cancer in Central and Eastern Europe, CEE). Eur J Cancer 44(10):1425–1437., DOI 10. 1016/j.ejca.2008.02.014 Vrdoljak E,WojtukiewiczMZ, Pienkowski T, Bodoky G, Berzinec P, Finek J, Todorovic V, Borojevic N, Croitoru A, 2011, Cancer epidemiology in Central, South and Eastern European countries. Croat Med J 52(4):478–487., DOI 10.3325/cmj.2011.52.478 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 775 EP 782 DI 10.1007/s12253-014-9889-0 PG 8 ER PT J AU Imayama, N Yamada, Shi Yanamoto, S Naruse, T Matsushita, Y Takahashi, H Seki, S Fujita, Sh Ikeda, T Umeda, M AF Imayama, Naomi Yamada, Shin-ichi Yanamoto, Souichi Naruse, Tomofumi Matsushita, Yuki Takahashi, Hidenori Seki, Sachiko Fujita, Shuichi Ikeda, Tohru Umeda, Masahiro TI FOXC2 Expression is Associated with Tumor Proliferation and Invasion Potential in Oral Tongue Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE FOXC2; Invasion; Metastasis; Proliferation; Oral tongue squamous cell carcinoma ID FOXC2; Invasion; Metastasis; Proliferation; Oral tongue squamous cell carcinoma AB Forkhead box protein C2 (FOXC2) is a gene encoding a transcription factor that controls the generation of mesodermal tissue including vascular and lymphatic tissues. FOXC2 has previously been associated with EMT and tumor angiogenesis in various cancers. Moreover, a relationship between the expression of FOXC2 and poor prognosis has been reported in various cancers.We herein examined the clinicopathological significance of FOXC2 in oral tongue squamous cell carcinoma (OTSCC) and attempted to clarify the function of FOXC2 in OTSCC cell lines in vitro. The overexpression of FOXC2 was more frequent in cancers with higher grades according to the pattern of invasion (grade 4 vs. 1–3; p<0.05).A correlationwas observed between the expression of FOXC2 and that of VEGF-A and -C (VEGF-A; p<0.05, VEGF-C; p<0.001). The high-FOXC2 expression group had a significantly poorer prognosis than that of the low-expression group (p<0.001). Multivariate analysis indicated that the overexpression of FOXC2 may also be an independent prognostic factor, similar to N classification (N0 vs 1/2; p<0.05), stage classification (stage I/II vs III/IV; p<0.05), pattern of invasion (grade 1-3vs 4; p<0.05), local recurrence (local recurrence (+) vs (−); p<0.01), and the overexpression of FOXC2 (FOXC2 overexpression (−) vs.(+); p<0.05). In the OTSCC cell line analysis, the expression of FOXC2 was also associated with proliferation and invasion potential. These results strongly suggest that the overexpression of FOXC2 may be a potent predictor of survival in OTSCC patients. C1 [Imayama, Naomi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yamada, Shin-ichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yanamoto, Souichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Naruse, Tomofumi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Matsushita, Yuki] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Takahashi, Hidenori] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Seki, Sachiko] Nagasaki University Graduate School of Biomedical Sciences, Department of Oral Pathology and Bone MetabolismNagasaki, Japan. [Fujita, Shuichi] Nagasaki University Graduate School of Biomedical Sciences, Department of Oral Pathology and Bone MetabolismNagasaki, Japan. [Ikeda, Tohru] Nagasaki University Graduate School of Biomedical Sciences, Department of Oral Pathology and Bone MetabolismNagasaki, Japan. [Umeda, Masahiro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. RP Yamada, Shi (reprint author), Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 852-8588 Nagasaki, Japan. EM shinshin@nagasaki-u.ac.jp CR Johnson NW, Jayasekara D, Amarasinghe AA, 2011, Squamous cell carcinoma and precursor lesions of the oral cavity: epidemiology and aetiology. Periodontol 57(1):19–37 HicksWL Jr, North JH Jr, Loree TR, Maamoun S, Mullins A, Orner JB, Bakamjian VY, Shedd DP, 1998, Surgery as a single modality therapy for squamous cell carcinoma of the oral tongue. Am J Otolaryngol 19(1):24–28 Sessions DG, Spector GJ, Lenox J, Parriott S, Haughey B, Chao C, Marks J, Perez C, 2003, Analysis of treatment results for base of tongue cancer. Laryngoscope 113(7):1252–1261 Gonzalez-Garcia R, Naval-Gias L, Rodriguez-Campo FJ, Sastre- Perez J, Munoz-Guerra MF, Gil-Diez Usandizaga JL, 2008, Contralateral lymph neck node metastasis of squamous cell carcinoma of the oral cavity: a retrospective analytic study in 315 patients. J Oral Maxillofac Surg 66(7):1390–1398 Ziober BL, Silverman SS Jr, Kramer RH, 2001, Adhesive mechanisms regulating invasion andmetastasis in oral cancer. Crit Rev Oral Biol Med 12(6):499–510 Bankfalvi A, Krassort M, Buchwalow IB, Vegh A, Felszeghy E, Piffko J, 2002, Gains and losses of adhesion molecules, CD44, Ecadherin, and beta-catenin, during oral carcinogenesis and tumour progression. J Pathol 198(3):343–351 Arora S, Kaur J, Sharma C, Mathur M, Bahadur S, Shukla NK, Deo SV, Ralhan R, 2005, Stromelysin 3, Ets-1, and vascular endothelial growth factor expression in oral precancerous and cancerous lesions: correlation with microvessel density, progression, and prognosis. Clin Cancer Res 11(6):2272–2284 Yanamoto S,KawasakiG,Yoshitomi I, Iwamoto T,Hirata K,Mizuno A, 2007, Clinicopathologic significance of EpCAM expression in squamous cell carcinoma of the tongue and its possibility as a potential target for tongue cancer gene therapy. Oral Oncol 43(9):869–877 BrinkmanBM,WongDT, 2006)Disease mechanismand biomarkers of oral squamous cell carcinoma. Curr Opin Oncol 18(2):228–233 Sasahira T, Kirita T, Kurihara M, Yamamoto K, Bhawal UK, Bosserhoff AK, Kuniyasu H, 2010, MIA-dependent angiogenesis and lymphangiogenesis are closely associated with progression, nodal metastasis and poor prognosis in tongue squamous cell carcinoma. Eur J Cancer 46(12):2285–2294 Sakamoto K, Imanishi Y, Tomita T, Shimoda M, Kameyama K, Shibata K, Sakai N, Ozawa H, Shigetomi S, Fujii R, Fujii M, Ogawa K, 2012, Overexpression of SIP1 and downregulation of Ecadherin predict delayed neck metastasis in stage I/II oral tongue squamous cell carcinoma after partial glossectomy. Ann Surg Oncol 19(2):612–619 Berx G, Raspe E, Christofori G, Thiery JP, Sleeman JP, 2007, Pre- EMTing metastasis? Recapitulation of morphogenetic processes in cancer. Clin Exp Metastasis 24(8):587–597 Kalluri R, Weinberg RA, 2009, The basics of epithelialmesenchymal transition. J Clin Invest 119(6):1420–1428 Baranwal S, Alahari SK, 2009, Molecular mechanisms controlling E-cadherin expression in breast cancer. Biochem Biophys Res Commun 384(1):6–11 Kopfstein L, Veikkola T, Djonov VG, Baeriswyl V, Schomber T, Strittmatter K, Stacker SA, Achen MG, Alitalo K, Christofori G, 2007, Distinct roles of vascular endothelial growth factor-D in lymphangiogenesis and metastasis. Am J Pathol 170(4):1348–1361 Lohela M, Bry M, Tammela T, Alitalo K, 2009, VEGFs and receptors involved in angiogenesis versus lymphangiogenesis. Curr Opin Cell Biol 21(2):154–165 Kume T, 2008, Foxc2 transcription factor: a newly described regulator of angiogenesis. Trends Cardiovasc Med 18(6):224–228 Kriederman BM, Myloyde TL, Witte MH, Dagenais SL, Witte CL, RennelsM, BernasMJ, LynchMT, Erickson RP, Caulder MS,Miura N, Jackson D, Brooks BP, Glover TW, 2003, FOXC2 haploinsufficient mice are a model for human autosomal dominant lymphedema-distichiasis syndrome. Hum Mol Genet 12(10):1179– 1185 Mani SA, Yang J, Brooks M, Schwaninger G, Zhou A, Miura N, Kutok JL, Hartwell K, Richardson AL, Weinberg RA, 2007, Mesenchyme Forkhead 1, FOXC2, plays a key role in metastasis and is associated with aggressive basal-like breast cancers. Proc Natl Acad Sci U S A 104(24):10069–10074 Mortazavi F, An J, Dubinett S, Rettig M, 2010, p120-catenin is transcriptionally downregulated by FOXC2 in non-small cell lung cancer cells. Mol Cancer Res 8(5):762–774 Watanabe A, Suzuki H, Yokobori T, Altan B, Kubo N, Araki K, Wada S, Mochida Y, Sasaki S, Kashiwabara K, Hosouchi Y, Kuwano H, 2013, Forkhead box protein C2 contributes to invasion and metastasis of extrahepatic cholangiocarcinoma, resulting in a poor prognosis. Cancer Sci 104(11):1427–1432 Hollier BG, Tinnirello AA,Werden SJ, EvansKW, Taube JH, Sarkar TR, Sphyris N, Shariati M, Kumar SV, Battula VL, Herschkowitz JI, Guerra R, Chang JT, Miura N, Rosen JM, Mani SA, 2013, FOXC2 expression links epithelial-mesenchymal transition and stem cell properties in breast cancer. Cancer Res 73(6):1981–1992 Liu B, Han SM, Tang XY, Han L, Li CZ, 2014, Overexpressed FOXC2 in ovarian cancer enhances the epithelial-to-mesenchymal transition and invasion of ovarian cancer cells. Oncol Rep 31(6): 2545–2554 Kume T, 2012, The role of FoxC2 transcription factor in tumor angiogenesis. J Oncol., DOI 10.1155/2012/204593 Sano H, Leboeuf JP, Novitskiy SV, Seo S, Zaja-Milatovic S, Dikov MM, Kume T, 2010, The Foxc2 transcription factor regulates tumor angiogenesis. Biochem Biophys Res Commun 392(2):201–206 Nishida N, Mimori K, Yokobori T, Sudo T, Tanaka F, Shibata K, Ishii H, Doki Y, Mori M, 2011, FOXC2 is a novel prognostic factor in human esophageal squamous cell carcinoma. Ann Surg Oncol 18(2): 535–542 Jiang W, Pang XG, Wang Q, Shen YX, Chen XK, Xi JJ, 2012, Prognostic role of Twist, Slug, and Foxc2 expression in stage I non-small-cell lung cancer after curative resection. Clin Lung Cancer 13(4):280–287 Zhu JL, Song YX, Wang ZN, Gao P, Wang MX, Dong YL, Xing CZ, Xu HM, 2013, The clinical significance of mesenchyme forkhead 1, FoxC2, in gastric carcinoma. Histopathology 62(7):1038–1048 Sasahira T, Ueda N, Yamamoto K, Kurihara M, Matsushima S, Bhawal UK, Kirita T, Kuniyasu H, 2014, Prox1 and FOXC2 act as regulators of lymphangiogenesis and angiogenesis in oral squamous cell carcinoma. PLoS One., DOI 10.1371/journal.pone.0092534 Yamamoto E, Kohama G, Sunakawa H, Iwai M, Hiratsuka H, 1983, Mode of invasion, bleomycin sensitivity, and clinical course in squamous cell carcinoma of the oral cavity. Cancer 51(12):2175–2180 McCarthy KS Jr, Szabo E, Flowers JL, Cox EB, Leight GS, Miller L, Konrath J, Soper JT, Budwit DA, Creasmna WT, Seigler HF, McCarthy KS Sr, 1986, Use of a monoclonal anti-estrogen receptor antibody in the immunohistochemical evaluation of human tumors. Cancer Res 46(8):4244–4268 Otrock ZK,Makarem JA, Shamseddine AI, 2007, Vascular endothelial growth factor family of ligands and receptors: review. Blood Cells Mol Dis 38(3):258–268 Ding MX, Lin XQ, Fu XY, Zhang N, Li JC, 2006, Expression of vascular endothelial growth factor-C and angiogenesis in esophageal squamous cell carcinoma. World J Gastroenterol 12(28):4582–4585 Watanabe T, Kobunai T, Yamamoto Y, Matsuda K, Ishihara S, Nozawa K, Iinuma H, Kanazawa T, Tanaka T, Konishi T, Ikeuchi H, Eshima K, Muto T, Nagawa H, 2011, Gene expression of mesenchyme forkhead 1, FOXC2, significantly correlates with the degree of lymph node metastasis in colorectal cancer. Int Surg 96(3): 207–216 LiW, Fu X, Liu R,Wu C, Bai J, Xu Y, Zhao Y, Xu Y, 2014, FOXC2 often overexpressed in glioblastoma enhances proliferation and invasion in glioblastoma cells. Oncol Res 21(2):111–120 Naruse T, Kawasaki G, Yanamoto S, Mizuno A, Umeda M, 2011, Immunohistochemical study of VEGF expression in oral squamous cell carcinomas: correlation with the mTOR-HIF-1α pathway. Anticancer Res 31(12):4429–4437 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 783 EP 791 DI 10.1007/s12253-014-9891-6 PG 9 ER PT J AU Ahmed, AR Aboelnaga, ME AF Ahmed, Allah Rehab Aboelnaga, M Engy TI Thyroid Cancer in Egypt: Histopathological Criteria, Correlation With Survival and Oestrogen Receptor Protein Expression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thyroid cancer; Immunohistochemistry; Papillary carcinoma; Oestrogen receptor; Egypt ID Thyroid cancer; Immunohistochemistry; Papillary carcinoma; Oestrogen receptor; Egypt AB Thyroid cancer represents approximately 1% of new cancer and oestrogen may play a role in the pathogenesis of thyroid neoplasm. We aimed to study the clinicopathological criteria and ER expression of thyroid cancer in Mansoura University (Egypt), and to correlate the survival to these clinicopathological data and ER expression. This retrospective study reviewed 644 patients with histologically proven thyroid carcinoma during the period from 2003 to 2011. 152 cases during the period between 2008 and 2011 were retrieved from the archive and examined by immunohistochemistry for oestrogen receptor-α (ER) expression. ER-α expression is significantly associated with the female sex, lymph node metastasis, TNM stage, extrathyroid extension, multifocality disease and recurrence and in the whole series (p<0.5). The same was noticed in papillary carcinoma (PTC) except the gender of the patient. Tumour type, extrathyroid extension and ER expression were the independent prognostic factors of DFS, while in PTC, only ER expression was the independent one. The histological type was the only independent prognostic factor for OAS in the series were studied for ER expression, while extrathyroid extension was the only one that affected OAS of PTC. There was significant positive correlation with lymph node metastasis and ER expression in whole patient and PTC cases. No difference in survival between the low and high ranges of positive oestrogen expression. The prognosis of thyroid carcinoma in Egypt is similar to that occurs worldwide. ER-α expression was a significant prognostic marker for DFS in thyroid cancer and can be used as a predictive factor of lymph node metastasis. C1 [Ahmed, Allah Rehab] Mansoura University, Faculty of Medicine, Department of PathologyMansoura, Egypt. [Aboelnaga, M Engy] Mansoura University, Faculty of Medicine, Clinical Oncology and Nuclear Medicine DepartmentMansoura, Egypt. RP Aboelnaga, ME (reprint author), Mansoura University, Faculty of Medicine, Clinical Oncology and Nuclear Medicine Department, Mansoura, Egypt. EM engyms2007@gmail.com CR Kilfoy BA, Zheng T, Holford TR et al, 2009, International patterns and trends in thyroid cancer incidence, 1973–2002. Cancer Causes Control 20(5):525–531 Enewold L, Zhu K, Ron E, 2009, Rising thyroid cancer incidence in the United States by demographic and tumour characteristics 1980– 2005. Cancer Epidemiol Biomarkers Prev 18(3):784–791 Yu GP, Li JC, Branovan D et al, 2010, Thyroid cancer incidence and survival in the national cancer institute surveillance, epidemiology, and end results race/ethnicity groups. Thyroid 20(5):465–473 Freedman LS, Edwards BK, Ries LAG et al., 2006, Cancer incidence in four member countries, Cyprus, Egypt, Israel and Jordan, of the Middle East cancer consortium, MECC, compared with US SEER National cancer institute, NIHPub. No 06-5873. Bethesda, MD. Chapter title; Thyroid cancer, 13, pp 120-131. Edited by Ronckers C and Ron E Livolsi VA, Albores-Saavedra J, Asa SL, 2004, Papillary carcinoma. In:De Lellis RA, Lloyd RV, Heitz PU, eds, Pathology and genetics of tumours of the endocrine organs. World Health Organization classification of tumours. IARC Press, Lyon, pp 57–66 Hundahl SA, Fleming ID, Fremgen AM et al, 1998, A National Cancer Data Base report on 53,856 cases of thyroid carcinoma treated in the U.S., 1985–1995, see comments). Cancer 83(12):2638–2648 Kebebew E, Greenspan FS, Clark OH et al, 2005, Anaplastic thyroid carcinoma. treatment outcome and prognostic factors. Cancer 103(7): 1330–1335 Carter WB, Tourtelot JB, Savell JG et al, 2011, New treatments and shifting paradigms in differentiated thyroid cancer management. Cancer Control 18(2):96–103 SantinAP and Furlanetto TW,, 2011, Role of estrogen in thyroid function and growth regulation. J Thyroid Res; Volume 2011, Article ID 875125, 7 pages Tavangar SM, Monajemzadeh M, Larijani B et al, 2007, Immunohistochemical study to oestrogen receptors in 351 human thyroid glands. Singapore Med J 48(8):744–747 Osborne CK, Schiff R, 2005, Estrogen-receptor biology: continuing progress and therapeutic implications. J Clin Oncol 23(8):1616–1622 Rossing MA, Voigt LF, Wicklund KG et al, 2000, Reproductive factors and risk of papillary thyroid cancer in women. Am J Epidemiol 151:765–772 Kishino T, Watanabe M, Kimura M, 1997, Anti-proliferative effect of toremifene and tamoxifen on oestrogen receptor-lacking anaplastic thyroid carcinoma cell lines. Biol Pharm Bull 20:1257–1260 Delellis RA, Lloyd RV, Heitz PU et al.,, 2004, WHO histological classification of thyroid and parathyroid tumours; pp50. In: Chapter 2 pp49-135. In: World Health Organization classification of tumours. Pathology and genetics of tumours of endocrine organs. ARC Press. Lyon Edge SB, Byrd DR, Compton CC et al, 2010, AJCC cancer staging manual. 7thed. Springer, New York Allred DC, Harvey JM, Berardo M et al, 1998, Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11:155–168 Tai P, Mould RF, Aye P et al, 2003, Descriptive epidemiology of thyroid carcinoma. Curr Oncol 10:54–65 Ito Y, McIver B, Besic N et al.,, 2012, Prognostic factors for thyroid carcinoma originating from follicular cells. J Thyroid Res., DOI 10.1155 Altekruse SF, Kosary CL, Krapcho M et al.,, 2010, SEER Cancer Statistics Review, 1975-2007, National Cancer Institute. Bethesda, MD, http://seer.cancer.gov/csr/1975_2007/, based on November 2009 SEER data submission, posted to the SEER web site, 2010 Williams D, 2008, Radiation carcinogenesis: lessons from chernobyl radiation carcinogenesis: lessons from chernobyl.Oncogene 27:S9–S18 Chen GG, Vlantis AC, Zeng Q et al, 2008, Regulation of cell growth by estrogen signaling and potential targets in thyroid cancer. Curr Cancer Drug Targets 8(5):367–377 Preston-Martin S, Franceschi S, Ron E et al, 2003, Thyroid cancer pooled analysis from 14 case control studies: what have we learned? Cancer Causes Control 14:787–789 Chunxia Q, Wei C, Yongxue Z et al, 2012, Correlation of clinicopathological features and expression ofmolecular markers with prognosis after 131i treatment of differentiated thyroid. Clin Nucl Med 37(3):e40–e46 Shah JP, Loree TR, Dharker D et al, 1992, Prognostic factors in differentiated carcinoma of the thyroid gland. Am J Surg 164(6):658–661 Cushing SL, Palme CE, Audet N et al, 2004, Prognostic factors in welldifferentiated thyroid carcinoma. Laryngoscope 114(12):2110–2115 Kavanagh D, Marie McIlroy M, Myers E et al, 2010, The role of oestrogen receptor a in human thyroid cancer: contributions from coregulatory proteins and the tyrosine Kinase receptor HER2. Endocrine-Relat Cancer 17:255–264 Cameselle-Teijeiro J, Alberte-Lista L and Peteiro-Gonzalez D,, 2011, Estrogen receptors and progesterone receptor expression and their relationship to types of thyroid cancer. Endocrine Abstracts 26; P454. 13th European Congress of Endocrinology Cho M, Kyung Lee M, Nam K, 2007, Expression and role of estrogen receptor α and b in medullary thyroid carcinoma: different roles in cancer growth and apoptosis. J Endocrinol 195:255–263 Passler C, Scheuba C, Prager G et al, 2004, Prognostic factors of papillary and follicular thyroid cancer: differences in an iodinereplete endemic goiter region. Endocrine-Relat Cancer 11:131–139 Ito Y, Ichihara K, Masuoka H et al, 2010, Establishment of an intraoperative staging system, istage, by improving UICC TNM classification system for papillary thyroid carcinoma. World J Surg 34(11):2570–2580 ItoY, Hirokawa M, Higashiyama Tet al, 2007, Prognosis and prognostic factors of follicular carcinoma in Japan: importance of postoperative pathological examination. World J Surg 31(7):1417–1424 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 793 EP 802 DI 10.1007/s12253-014-9892-5 PG 10 ER PT J AU Alay, I Turan, T Ureyen, I Karalok, A Tasci, T Ozfuttu, A Kose, FM Tulunay, G AF Alay, Ismail Turan, Taner Ureyen, Isin Karalok, Alper Tasci, Tolga Ozfuttu, Ahmet Kose, Faruk Mehmet Tulunay, Gokhan TI Lymphadenectomy Should Be Performed Up to the Renal Vein in Patients with Intermediate-High Risk Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial cancer; Supramesenteric lymph node; Lymphatic spread; Left renal vein ID Endometrial cancer; Supramesenteric lymph node; Lymphatic spread; Left renal vein AB We aimed to evaluate para-aortic metastases relative to the level of inferior mesenteric artery (IMA) and to discuss the clinico-pathological features of these patients. A total of 204 patients who underwent systematic pelvic and para-aortic lymphadenectomy up to the level of renal veins for endometrial cancer between January 2007 and August 2013 were included in this study. Of these 204 patients, 44 (21.6 %) had lymph node involvement. From a total of 27 patients with paraaortic lymph node (PALN) metastasis, 11 had only supramesenteric and 4 had only inframesenteric nodal involvement, while 12 had both supramesenteric and inframesenteric metastases. Supramesenteric lymph node metastases were detected in 85.2 % of patients who have paraaortic metastases and in 11.3 % of all patients. Additionally, 5 patients had only supramesenteric lymphatic metastasis. The surgico-pathological characteristics of patients with isolated supramesenteric and inframesenteric metastasis were similar. However, the patients with lymphatic spread in both regions were found to have pelvic lymphatic metastasis and cervical invasion more commonly compared to patients with only supramesenteric or only inframesenteric metastasis. The site of metastatic lymph nodes wasn’t associated with the likelihood and site of recurrence. Lymphadenectomy should be performed up to the level of renal vein in case of the presence of indication for lymphadenectomy in patients with endometrial cancer. Additionally, it is not possible to predict the patients with supramesenteric lymph node involvement by tumor grade, histological type and myometrial invasion. C1 [Alay, Ismail] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology Department, Etlik Street, Kecioren, 06010 Ankara, Turkey. [Turan, Taner] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology Department, Etlik Street, Kecioren, 06010 Ankara, Turkey. [Ureyen, Isin] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology Department, Etlik Street, Kecioren, 06010 Ankara, Turkey. [Karalok, Alper] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology Department, Etlik Street, Kecioren, 06010 Ankara, Turkey. [Tasci, Tolga] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology Department, Etlik Street, Kecioren, 06010 Ankara, Turkey. [Ozfuttu, Ahmet] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Pathology Division, KeciorenAnkara, Turkey. [Kose, Faruk Mehmet] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology Department, Etlik Street, Kecioren, 06010 Ankara, Turkey. [Tulunay, Gokhan] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology Department, Etlik Street, Kecioren, 06010 Ankara, Turkey. RP Ureyen, I (reprint author), Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology Department, 06010 Ankara, Turkey. EM isin.ureyen@gmail.com CR Announcements, 1989, FIGO, the International Federation of Obstetricians and Gynecologists, stages: 1988 revision. Gynecol Oncol 35:125–126 Hoekstra AV, Kim RJ, Small W Jr et al, 2009, FIGO stage IIIC endometrial carcinoma: prognostic factors and outcomes. Gynecol Oncol 114(2):273–278 McMeekin DS, Lashbrook D, Gold M, Johnson G, Walker JL, Mannel R, 2001, Analysis of FIGO Stage IIIC endometrial cancer patients. Gynecol Oncol 81(2):273–278 Creasman WT, Morrow CP, Bundy BN et al, 1987, Surgical pathologic spread patterns of endometrial cancer. A Gynecologic Oncology Group study. Cancer 60:2035–2041 Hirahatake K, Hareyama H, Sakuragi N et al, 1997, A clinical and pathologic study on para-aortic lymph nodemetastasis in endometrial carcinoma. J Surg Oncol 65:82–87 Mariani A, Keeney GL, Aletti G et al, 2004, Endometrial carcinoma: paraaortic dissemination. Gynecol Oncol 92:833–838 Turan T, Hizli D, Sarici S et al, 2011, Is it possible to predict paraaortic lymph node metastasis in endometrial cancer? Eur J Obstet Gynecol Reprod Biol 158(2):274–279 National Comprehensive Cancer Network, 2006, Clinical practice guidelines in oncology, uterine cancers. Version 1. National Comprehensive Cancer Network, Washington, DC ASTEC study group, Kitchener H, Swart AM, Qian Q, Amos C, Parmar MK, 2009, Efficacy of systematic pelvic lymphadenectomy in endometrial cancer, MRC ASTEC trial): a randomized study. Lancet 373(9658):125–136 Chan JK, Cheung MK, Huh WK et al, 2006, Therapeutic role of lymph node resection in endometrioid corpus cancer: a study of 12, 333 patients. Cancer 107(8):1823–1830 Fotopoulou C, Savvatis K, Kraetschell R, Schefold JC, Lichtenegger W, Sehouli J, 2010, Systematic pelvic and aortic lymphadenectomy in intermediate and high-risk endometrial cancer: lymph-node mapping and identification of predictive factors for lymph-node status. Eur J Obstet Gynecol Reprod Biol 149(2):199–203 Kumar S, Podratz KC, Bakkum-Gamez JN et al, 2014, Prospective assessment of the prevalence of pelvic, paraaortic and high paraaortic lymph nodemetastasis in endometrial cancer. Gynecol Oncol 132(1): 38–43 Nomura H, Aoki D, Suzuki N et al, 2006, Analysis of clinicopathologic factors predicting para-aortic lymph node metastasis in endometrial cancer. Int J Gynecol Cancer 16(2):799–804 Odagiri T,Watari H, Kato T, et al, 2014, Distribution of lymph node metastasis sites in endometrial cancer undergoing systematic pelvic and para-aortic lymphadenectomy: a proposal of optimal lymphadenectomy for future clinical trials. Ann Surg Oncol Chang SJ, Kim WY, Yoon JH, Yoo SC, Chang KH, Ryu HS, 2008, Para-aortic lymphadenectomy improves survival in patients with intermediate to high-risk endometrial carcinoma. Acta Obstet Gynecol Scand 87(12):1361–1369 Mariani A,Webb MJ, Galli L, Podratz KC, 2000, Potential therapeutic role of para-aortic lymphadenectomy in node-positive endometrial cancer. Gynecol Oncol 76(3):348–356 Todo Y, Kato H, Kaneuchi M, Watari H, Takeda M, Sakuragi N, 2010, Survival effect of para-aortic lymphadenectomy in endometrial cancer, SEPAL study): a retrospective cohort analysis. Lancet 375(9721):1165–1172 Meeting Report, 2009, The new FIGO staging system for cancers of the vulva, cervix, endometrium and sarcomas. Gynecol Oncol 115: 325–328 Turan T, Yilmaz SS, Hizli D et al, 2011, A prospective evaluation of lymphatic dissemination in endometrial cancer: is it adequate to perform lymph node dissection up to the inferior mesenteric artery? Int J Gynecol Cancer 21(5):864–869 Creutzberg CL, van Putten WL, Koper PC et al, 2000, Surgery and postoperative radiotherapy versus surgery alone for patients with stage-1 endometrial carcinoma: multicentre randomized trial. PORTEC Study Group. Post Operative Radiation Therapy in Endometrial Carcinoma. Lancet 355(9213):1404–1411 National Comprehensive Cancer Network, 2014, Clinical practice guidelines in oncology, uterine cancers. Version 1. National Comprehensive Cancer Network, Washington, DC Yokoyama Y, Maruyama H, Sato S et al, 1997, Indispensability of pelvic and paraaortic lymphadenectomy in endometrial cancers. Gynecol Oncol 64:411–417 Mariani A, Dowdy SC, Cliby WA et al, 2008, Prospective assessment of lymphatic dissemination in endometrial cancer: a paradigm shift in surgical staging. Gynecol Oncol 109:11– 18 Burke TW, Levenback C, Tornos C et al, 1996, Intraabdominal lymphatic mapping to direct selective pelvic and paraaortic lymphadenectomy in women with high-risk endometrial cancer: results of a pilot study. Gynecol Oncol 62:169–173 Maccauro M, Lucignani G, Aliberti G et al, 2005, Sentinel lymph node detection following the hysteroscopic peritumoural injection of 99mTc-labelled albumin nanocolloid in endometrial cancer. Eur J Nucl Med Mol Imaging 32:569–574 Turan T, Hizli D, Sarici S et al, 2012, What is the impact of cervical invasion on lymph node metastasis in patients with stage IIIC endometrial cancer? Arch Gynecol Obstet 285(4):1119–1124 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 803 EP 810 DI 10.1007/s12253-014-9893-4 PG 8 ER PT J AU Berretta, R Gizzo, S Noventa, M Marrazzo, V Franchi, L Migliavacca, C Michela, M Merisio, C Modena, BA Patrelli, ST AF Berretta, Roberto Gizzo, Salvatore Noventa, Marco Marrazzo, Vivienne Franchi, Laura Migliavacca, Costanza Michela, Monica Merisio, Carla Modena, Bacchi Alberto Patrelli, Silvio Tito TI Quality of Life in Patients Affected by Endometrial Cancer: Comparison Among Laparotomy, Laparoscopy and Vaginal Approach SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial cancer; Laparotomic surgery; Laparoscopic surgery; Vaginal surgery; Quality of life; Complications ID Endometrial cancer; Laparotomic surgery; Laparoscopic surgery; Vaginal surgery; Quality of life; Complications AB The aim of this study is to verify if the surgical approach (laparoscopy/laparotomy/vaginal) in stage-I endometrial cancer treatment, may have effects on intra- and postoperative outcomes and on the patient’s quality of life. The study group consisted of patients with histological diagnosis of type-I endometrial adenocarcinoma, stage-I. They were divided into three groups according to surgical approach chosen (laparotomic/laparoscopic/vaginal). Every patient answered a telephone health survey (SF-36) at 30 and 180 days post-surgery. Surgical-operating times, hospitalization length and short/long-term complications after surgery were also compared. The SF-36 survey revealed a better performance status in patients who underwent laparoscopy as compared to those who received laparotomy or vaginal surgery. We found significantly better results considering General Health, Physical Functioning, Role-Physical and Bodily Pain in the laparoscopy group after 30 and 180 days. Patients who underwent laparoscopy had significantly shorter hospitalization and less post-operative complications even if laparoscopy required significantly longer surgical-operating times compared to vaginal surgery. Our data confirm the superiority of the laparoscopic approach respect to the laparotomic and vaginal ones both in term of hospitalization length and post-operative complications. C1 [Berretta, Roberto] University of Parma, Department of Surgical SciencesParma, Italy. [Gizzo, Salvatore] University of Padua, Department of Woman and Child Health, Via Giustiniani 3, 35128 Padova, Italy. [Noventa, Marco] University of Padua, Department of Woman and Child Health, Via Giustiniani 3, 35128 Padova, Italy. [Marrazzo, Vivienne] University of Parma, Department of Surgical SciencesParma, Italy. [Franchi, Laura] University of Parma, Department of Surgical SciencesParma, Italy. [Migliavacca, Costanza] University of Parma, Department of Surgical SciencesParma, Italy. [Michela, Monica] University of Parma, Department of Surgical SciencesParma, Italy. [Merisio, Carla] University of Parma, Department of Surgical SciencesParma, Italy. [Modena, Bacchi Alberto] University of Parma, Department of Surgical SciencesParma, Italy. [Patrelli, Silvio Tito] University of Parma, Department of Surgical SciencesParma, Italy. RP Gizzo, S (reprint author), University of Padua, Department of Woman and Child Health, 35128 Padova, Italy. EM ginecologia_padova@libero.it CR Berretta R, Merisio C, Melpignano M et al, 2008, Vaginal versus abdominal hysterectomy in endometrial cancer: a retrospective study in a selective population. Int J Gynecol Cancer 18:797–802 Gizzo S, Di Gangi S, Bertocco A et al, 2014, Levonorgestrel intrauterine system in adjuvant tamoxifen treatment: balance of breast risks and endometrial benefits–systematic review of literature. Reprod Sci 21(4):423–431 Saccardi C, Gizzo S, Patrelli TS et al, 2013, Endometrial surveillance in tamoxifen users: role, timing and accuracy of hysteroscopic investigation: observational longitudinal cohort study. Endocr Relat Cancer 20(4):455–462 Patrelli TS, Berretta R, RollaMet al, 2009, Pelvic lymphadenectomy in endometrial cancer: our current experience. Eur J Gynaecol Oncol 30:536–538 Brazier JE, Harper R, Jones NM et al, 1992, Validating the SF-36 health survey questionnaire: new outcome measure for primary care. BMJ 305(6846):160–164 Pecorelli S, 2009, Revised FIGO staging for carcinoma of the vulva, cervix, and endometrium. Int J Gynaecol Obstet 105:103–104 Mikuta JJ, 1993, International Federation of Gynecology and Obstetrics staging of endometrial cancer 1988. Cancer 71(4 Suppl): 1460–1463 Berretta R, Patrelli TS, Migliavacca C et al, 2014, Assessment of tumor size as a useful marker for the surgical staging of endometrial cancer. Oncol Rep 31(5):2407–2412 Gizzo S, Burul G, Di Gangi S et al, 2013, LigaSure vessel sealing system in vaginal hysterectomy: safety, efficacy and limitations.Arch Gynecol Obstet 288(5):1067–1074 Eltabbakh GH, 2002, Analysis of survival after laparoscopy in women with endometrial carcinoma. Cancer 95(9):1894–1901 Litta P, Fracas M, Pozzan C et al, 2003, Laparoscopicmanagement of early stage endometrial cancer. Eur J Gynaecol Oncol 24(1):41–44 Berretta R, Patrelli TS, Faioli R et al, 2013, Dedifferentiated endometrial cancer: an atypical case diagnosed from cerebellar and adrenal metastasis: case presentation and review of literature. Int J Clin Exp Pathol 6(8):1652–1657 Patrelli TS, Gizzo S, Di Gangi S et al, 2011, Cervical Mullerian adenosarcoma with heterologous sarcomatous overgrowth: a fourth case and review of literature. BMC Cancer 11:236 Patrelli TS, Silini EM, Gizzo S et al, 2011, Extragenital Mullerian adenosarcoma with pouch of Douglas location. BMC Cancer 11:171 Magrina JF, Weaver AL, 2004, Laparoscopic treatment of endometrial cancer: five-year recurrence and survival rates. Eur J Gynaecol Oncol 25(4):439–441 Malur S, Possover M,MichelsW, Schneider A, 2001, Laparoscopicassisted vaginal versus abdominal surgery in patients with endometrial cancer–a prospective randomized trial. Gynecol Oncol 80(2): 239–244 Holub Z, Jabor A, Bartos P et al, 2002, Laparoscopic surgery for endometrial cancer: long-term results of a multicentric study. Eur J Gynaecol Oncol 23(4):305–310 Zullo F, Palomba S, Russo T et al, 2005, A prospective randomized comparison between laparoscopic and laparotomic approaches in women with early stage endometrial cancer: a focus on the quality of life. Am J Obstet Gynecol 193(4):1344–1352 Nieboer TE, Hendriks JC, Bongers MY et al, 2012, Quality of life after laparoscopic and abdominal hysterectomy: a randomized controlled trial. Obstet Gynecol 119(1):85–91 Janda M, Gebski V, Brand A et al, 2010, Quality of life after total laparoscopic hysterectomy versus total abdominal hysterectomy for stage I endometrial cancer, LACE): a randomised trial. Lancet Oncol 11(8):772–780 Capelli G, De Vincenzo RI, Addamo A et al, 2002, Which dimensions of health-related quality of life are altered in patients attending the different gynecologic oncology health care settings? Cancer 95(12):2500–2507 Kornblith AB, Huang HQ, Walker JL et al, 2009, Quality of life of patients with endometrial cancer undergoing laparoscopic international federation of gynecology and obstetrics staging compared with laparotomy: a Gynecologic Oncology Group study. J Clin Oncol 27(32):5337–5342, Erratum in: J Clin Oncol. 2010 Jun 1;28(16): 2805 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 811 EP 816 DI 10.1007/s12253-014-9895-2 PG 6 ER PT J AU Terada, T AF Terada, Tadashi TI Expression of Cytokeratins in Glioblastoma Multiforme SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma multiforme; CK; Immunohistochemistry ID Glioblastoma multiforme; CK; Immunohistochemistry AB Little is known about the cytokeratin (CK) expressions in glioblastoma multiforme (GBM). The aim is to explore the CK expression in GM using immunohistochemistry (IHC). IHC study in 30 cases (median=68 years, SE=12.6) of GBM in brain. CK expression using AE1/3 antidody was seen in 29/30 (97 %) cases. There were no expressions of CK34BE12, CK5, CK6, CK7, CK8, CK14, CK 18, CK19, and CK20. Expression of p53 and glial fibrillary acidic protein (GFAP) were recognized in all 30 cases (100 %). All cases showed Ki-67 antigen labeling, index of which ranged from 6 to 43 % (m±SD=24+16). The IHC using CKAE1/3 in GBM very frequently shows positive reaction. The expression may cause difficulty in pathologic diagnosis in GBM, particularly in discrimination between GBM and metastatic carcinoma. The CK positivity in GM may be due to CK molecules other than CK34BE12, CK5, CK6, CK7, CK8, CK14, CK18, CK19 and CK20. GBM frequently expression p53 and high Ki-67 labeling. C1 [Terada, Tadashi] Shizuoka City Shimizu Hospital, Department of Pathology, Miyakami 1231, Shimizu-Ku, 424-8636 Shizuoka, Japan. RP Terada, T (reprint author), Shizuoka City Shimizu Hospital, Department of Pathology, 424-8636 Shizuoka, Japan. EM piyo0111jp@yahoo.co.jp CR CosgroveM, Fitzgibbons PL, Sherrod A, Chandrasoma PT,Martin SE, 1989, Intermediate filament expression in astrocytic neoplasms.AmJ Surg Pathol 13:141–145 Ng HK, Lo ST, 1989, Cytokeratin immunoreactivity in gliomas. Histopathology 14:359–368 Cosgrove MM, Rich KA, Kunin SA, Sherrod AE, Martin SE, 1993, Keratin intermediate filament expression in astrocytic neoplasms: analysis by immunocytochemistry, western blot, and northern hybridization. Mod Pathol 6:342–347 Hirato J, Nakazato Y, Ogawa A, 1994, Expression of non-glial intermediate filament proteins in gliomas. Clin Neuropathol 13:1–11 Oh D, Prayson RA, 1999, Evaluation of epithelial and keratin markers in glioblastoma multiforme: an immunohistochemical study. Arch Pathol Lab Med 123:917–920 Goswami C, Chatterjee U, Sen S, Chatterjee S, Sarkar S, 2007, Expression of cytokeratins in gliomas. Indian J Pathol Microbiol 50: 478–481 Terada T, Kawaguchi M, Furukawa K, Sekido Y, Osamura Y, 2002, Minute mixed ductal-endocrine carcinoma of the pancreas with predominant intraductal growth. Pathol Int 52:740–746 Masica DL, Karchin R, 2011, Correlation of somatic mutation and expression identifies genes important in human glioblastoma progression and survival. Cancer Res 71:4550–4561 Jung TY, Jung S, Kim IY,Moon KS, JangWY, Park SJ, Kim YH, Kim HS, Min JJ, Kim J, 2012, Pathologic analysis of glioblastoma via multiple stereotactic biopsies of active tumor and necrosis. Oncol Rep 27:707–713 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 817 EP 819 DI 10.1007/s12253-015-9896-9 PG 3 ER PT J AU Marosvari, D Teglasi, V Csala, I Marschalko, M Bodor, Cs Timar, B Csomor, J Harsing, J Reiniger, L AF Marosvari, Dora Teglasi, Vanda Csala, Iren Marschalko, Marta Bodor, Csaba Timar, Botond Csomor, Judit Harsing, Judit Reiniger, Lilla TI Altered MicroRNA Expression in Folliculotropic and Transformed Mycosis Fungoides SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MicroRNA; Mycosis fungoides; Folliculotropic mycosis fungoides; Transformed mycosis fungoides ID MicroRNA; Mycosis fungoides; Folliculotropic mycosis fungoides; Transformed mycosis fungoides AB Mycosis fungoides (MF) is a common, indolent primary cutaneous T-cell lymphoma (CTCL), with rare, more aggressive variants, such as folliculotropic MF (FMF). A minority of the MF cases may undergo large cell transformation (T-MF) associated with poor prognosis. A selection of microRNAs (miRs) contribute to the pathogenesis and progression of classic MF, and may also be useful in differential diagnostics. However, the molecular background of FMF and the mechanisms involved in large cell transformation are obscure. We analyzed the expression of 11 miRs in 9 FMF and 7 T-MF cases. Three miRs, including miR-93-5p, miR-181a and miR-34a were significantly upregulated in both FMF and T-MF. FMF also showed overexpression of miR-155 and miR-223, while miR-181b and miR-326 were overexpressed in T-MF cases compared to controls. These results by identifying a number of differentially expressed microRNAs add further insight into the molecular pathogenesis of folliculotropic MF and large cell transformation of MF. C1 [Marosvari, Dora] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Teglasi, Vanda] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Csala, Iren] Semmelweis University, Institute of Behavioural SciencesBudapest, Hungary. [Marschalko, Marta] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Bodor, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Timar, Botond] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Csomor, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Harsing, Judit] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Reiniger, Lilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Reiniger, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM lillareiniger@yahoo.com CR Swerdlow SH, International Agency for Research on Cancer., World Health Organization. WHO classification of tumours of haematopoietic and lymphoid tissues. 4th ed. Lyon, France: International Agency for Research on Cancer, 2008 Willemze R, Jaffe ES, Burg G et al, 2005)WHO-EORTC classification for cutaneous lymphomas. Blood 105:3768–85 Scarisbrick JJ, Kim YH,Whittaker SJ et al, 2014, Prognostic factors, prognostic indices and staging in mycosis fungoides and sezary syndrome: where are we now? Br J Dermatol 170:1226–36 Gerami P, Rosen S, Kuzel T, Boone SL, Guitart J, 2008, Folliculotropic mycosis fungoides: an aggressive variant of cutaneous T-cell lymphoma. Arch Dermatol 144:738–46 Vergier B, de Muret A, Beylot-Barry M et al, 2000, Transformation of mycosis fungoides: clinicopathological and prognostic features of 45 cases french study group of cutaneious lymphomas. Blood 95: 2212–8 BennerMF, Jansen PM,VermeerMH,Willemze R, 2012, Prognostic factors in transformed mycosis fungoides: a retrospective analysis of 100 cases. Blood 119:1643–9 Wong HK, Mishra A, Hake T, Porcu P, 2011, Evolving insights in the pathogenesis and therapy of cutaneous T-cell lymphoma, mycosis fungoides and Sezary syndrome). Br J Haematol 155:150–66 van Doorn R, Zoutman WH, Dijkman R et al, 2005, Epigenetic profiling of cutaneous T-cell lymphoma: promoter hypermethylation of multiple tumor suppressor genes including BCL7a, PTPRG, and p73. J Clin Oncol 23:3886–96 Garzon R, Croce CM, 2008, MicroRNAs in normal and malignant hematopoiesis. Curr Opin Hematol 15:352–8 Calore F, Lovat F, GarofaloM(2013, Non-coding RNAs and cancer. Int J Mol Sci 14:17085–110 van Kester MS, Ballabio E, Benner MF et al, 2011, miRNA expression profiling of mycosis fungoides. Mol Oncol 5:273–80 Maj J, Jankowska-Konsur A, Sadakierska-Chudy A, Noga L, Reich A, 2011, Altered microRNA expression in mycosis fungoides. Br J Dermatol 166:331–6 Ralfkiaer U, Hagedorn PH, Bangsgaard N et al, 2011, Diagnostic microRNA profiling in cutaneous T-cell lymphoma, CTCL). Blood 118:5891–900 Moyal L, Barzilai A, Gorovitz B et al, 2013, miR-155 is involved in tumor progression of mycosis fungoides. Exp Dermatol 22:431–3 Contassot E, Kerl K, Roques S et al, 2008, Resistance to FasL and tumor necrosis factor-related apoptosis-inducing ligand-mediated apoptosis in Sezary syndrome T-cells associated with impaired death receptor and FLICE-inhibitory protein expression. Blood 111:4780–7 Dereure O, Levi E, Vonderheid EC, KadinME, 2002, Infrequent Fas mutations but no Bax or p53 mutations in early mycosis fungoides: a possible mechanism for the accumulation of malignant T lymphocytes in the skin. J Invest Dermatol 118:949–56 Wu J, Nihal M, Siddiqui J, Vonderheid EC, Wood GS, 2009, Low FAS/CD95 expression by CTCL correlates with reduced sensitivity to apoptosis that can be restored by FAS upregulation. J Invest Dermatol 129:1165–73 Nielsen M, Kaestel CG, Eriksen KWet al, 1999, Inhibition of constitutively activated Stat3 correlates with altered Bcl-2/Bax expression and induction of apoptosis in mycosis fungoides tumor cells. Leukemia 13:735–8 Sommer VH, Clemmensen OJ, Nielsen O et al, 2004, In vivo activation of STAT3 in cutaneous T-cell lymphoma. Evidence for an antiapoptotic function of STAT3. Leukemia 18:1288–95 Mao X, Orchard G, Lillington DM et al, 2004, BCL2 and JUNB abnormalities in primary cutaneous lymphomas. Br J Dermatol 151: 546–56 Mao X, Orchard G, Mitchell TJ et al, 2008, A genomic and expression study of AP-1 in primary cutaneous T-cell lymphoma: evidence for dysregulated expression of JUNB and JUND in MF and SS. J Cutan Pathol 35:899–910 Mao X, Lillington D, Scarisbrick JJ et al, 2002, Molecular cytogenetic analysis of cutaneous T-cell lymphomas: identification of common genetic alterations in sezary syndrome and mycosis fungoides. Br J Dermatol 147:464–75 Benner MF, Ballabio E, van Kester MS et al, 2012, Primary cutaneous anaplastic large cell lymphoma shows a distinct miRNA expression profile and reveals differences from tumor-stage mycosis fungoides. Exp Dermatol 21:632–4 Prochazkova M, Chevret E, Mainhaguiet G et al, 2007, Common chromosomal abnormalities in mycosis fungoides transformation. Genes, Chromosom Cancer 46:828–38 Laharanne E, Chevret E, Idrissi Y et al, 2010, CDKN2A-CDKN2B deletion defines an aggressive subset of cutaneous T-cell lymphoma. Mod Pathol 23:547–58 Manfe V, Biskup E, Rosbjerg A et al, 2012, miR-122 regulates p53/ Akt signalling and the chemotherapy-induced apoptosis in cutaneous T-cell lymphoma. PLoS One 7:e29541 McGirt LY, Adams CM, Baerenwald DA, Zwerner JP, Zic JA, Eischen CM, 2013, miR-223 regulates cell growth and targets proto-oncogenes in mycosis fungoides/cutaneous T-cell lymphoma. J Investi Dermatol 134:1101–7 Mi QS, Xu YP,Wang H, Qi RQ, Dong Z, Zhou L, 2013, Deletion of microRNA miR-223 increases Langerhans cell cross-presentation. Int J Biochem Cell Biol 45:395–400 Sotillo E, Laver T, Mellert H et al, 2011, Myc overexpression brings out unexpected antiapoptotic effects of miR-34a. Oncogene 30: 2587–94 Rizzo M, Mariani L, Cavallini S, Simili M, Rainaldi G, 2012, The over-expression of miR-34a fails to block DoHH2 lymphoma cell proliferation by reducing p53 via c-MYC down-regulation. Nucleic acid therapeutics 22:283–8 Kanavaros P, Ioannidou D, TzardiMet al, 1994)Mycosis fungoides: expression of C-myc p62 p53, bcl-2 and PCNA proteins and absence of association with Epstein-Barr virus. Pathol Res Pract 190:767–74 Salgado R, Servitje O, Gallardo F et al, 2010, Oligonucleotide array- CGH identifies genomic subgroups and prognosticmarkers for tumor stage mycosis fungoides. J invest Dermatol 130:1126–35 Fang L,DuWW,YangWet al, 2012)MiR-93 enhances angiogenesis and metastasis by targeting LATS2. Cell cycle, Georgetown. Tex 11: 4352–65 Xi Y, Formentini A, Chien M et al, 2006, Prognostic values of microRNAs in colorectal cancer. Biomark Insights 2:113–21 Mansueto G, Forzati F, Ferraro A et al, 2010, Identification of a New pathway for tumor progression: MicroRNA-181b Up-regulation and CBX7 down-regulation by HMGA1 protein. Genes & cancer 1:210– 24 Taylor MA, Sossey-Alaoui K, Thompson CL, Danielpour D, Schiemann WP, 2013, TGF-beta upregulates miR-181a expression to promote breast cancer metastasis. J Clin Invest 123:150–63 Fang L, Deng Z, Shatseva T et al, 2010, MicroRNA miR-93 promotes tumor growth and angiogenesis by targeting integrin-beta8. Oncogene 30:806–21 Williams A, Henao-Mejia J, Harman CC, Flavell RA, 2013, miR- 181 and metabolic regulation in the immune system. Cold Spring Harb Symp Quant Biol 78:223–30 Hsu SD, Tseng YT, Shrestha S et al, 2014, miRTarBase update 2014: an information resource for experimentally validated miRNA-target interactions. Nucleic Acids Res 42:D78–85 Scarisbrick JJ, Woolford AJ, Russell-Jones R, Whittaker SJ, 2000, Loss of heterozygosity on 10q and microsatellite instability in advanced stages of primary cutaneous T-cell lymphoma and possible association with homozygous deletion of PTEN. Blood 95:2937–42 Katona TM, Smoller BR,Webb AL, Hattab EM, Khalil A, Hiatt KM, 2013, Expression of PTEN in mycosis fungoides and correlation with loss of heterozygosity. Am J Dermatopathol 35:555–60 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 821 EP 825 DI 10.1007/s12253-015-9897-8 PG 5 ER PT J AU Ivanov, BI Gritsenko, AV Miroshnikov, AS AF Ivanov, B Iuri Gritsenko, A Viktor Miroshnikov, A Sergey TI Evaluation of Antitumor Activity of Platelet Microbicidal Protein on the Model of Transplanted Breast Cancer in CBRB-Rb(8.17) 1Iem Mice SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Platelet microbicidal protein; Antitumor activity; Breast cancer; Mouse model ID Platelet microbicidal protein; Antitumor activity; Breast cancer; Mouse model AB Breast cancer is the most common women’s cancer in the world. There is considerable current interest in developing anticancer agents with a new mode of action because of the development of resistance by cancer cells towards current anticancer drugs. Mamalian cells have been shown to contain small, cationic, microbicidal peptides. Antimicrobial peptides have drawn attention as a promising alternative to current antitumor agents. Such peptides have been isolated both from animal and human platelets and have been termed platelets microbicidal proteins (PMP). The aim of this work was to study antitumor activity of PMP in vivo on the model of mouse breast cancer in comparison with antitumor hexapeptide Arg-alpha-Asp-Lys-Val-Tyr-Arg (Immunofan). We demonstrated that the tumors treated with PMP were significant smaller than the control groups (P<0.05). In experiments in vivo using CBRB-Rb(8.17)1Iem mice with transplanted tumors PMP inhibited tumor growth during the treatments and after its discontinuation. These findings indicate that PMP can exert antitumor effects. Therefore, PMP may be used for the development of therapy for the intervention of breast cancer. C1 [Ivanov, B Iuri] Russian Academy of Sciences, Institute of Cellular and Intracellular SymbiosisOrenburg, Russian Federation. [Gritsenko, A Viktor] Russian Academy of Sciences, Institute of Cellular and Intracellular SymbiosisOrenburg, Russian Federation. [Miroshnikov, A Sergey] Russian Academy of Agricultural Sciences, All-Russian Institute of Meat Cattle BreedingOrenburg, Russian Federation. RP Ivanov, BI (reprint author), Russian Academy of Sciences, Institute of Cellular and Intracellular Symbiosis, Orenburg, Russian Federation. EM ubi2010@rambler.ru CR Biersack B, Ahmad A, Sarkar FH, Schobert R, 2012, Coinage metal complexes against breast cancer. Curr Med Chem 19:3949–3956 Trape AP, Gonzalez-Angulo AM, 2012, Breast cancer and metastasis: on the way toward individualized therapy. Cancer Genomics Proteomics 9:297–310 Caffarel MM, Andradas C, Perez-Gomez E, Guzman M, Sanchez C, 2012, Cannabinoids: a new hope for breast cancer therapy? Cancer Treat Rev 38:911–918 Hou L, Zhao X, Wang P, Ning Q, Meng M, Caigang L, 2013, Antitumor activity of antimicrobial peptides containing CisoDGRC in CD13 negative breast cancer cells. PLoS ONE 8:e53491., DOI 10. 1371/journal.pone.0053491 Shamova OV, Sakuta GA, Orlov DS, Zenin VV, Shteĭn GI,Kolodkin NI, Afonina IV, Kokriakov VN, 2007, Effects of antimicrobial peptides of neutrophils on tumor and normal cells in culture. Tsitologiia 49:1000–1010 Kedzierska M, Czernek U, Szydlowska-Pazera K, Potemski P, Piekarski J, Jeziorski A, Olas B, 2013, The changes of blood platelet activation in breast cancer patients before surgery, after surgery, and in various phases of the chemotherapy. Platelets 24(6):462–468 Timar J, Tovari J, Raso E, Meszaros L, Bereczky B, Lapis K, 2005, Platelet-mimicry of cancer cells: epiphenomenon with clinical significance. Oncology 69:185–201 Yeaman MR, 2010, Platelets in defense against bacterial pathogens. Cell Mol Life Sci 67:525–544 Nurden AT, 2011, Platelets, inflammation and tissue regeneration. Thromb Haemost 105(Suppl 1):S13–S33 Ivanov IB, Miroshnikov SA, Lebedev SV, Notova SV, Sipailova OI, Gritsenko VA, 2011, Antitumor medication RU Patent No. 2419451 Vodovozova EL,Moiseeva EV,GrechkoGK,Gayenko GP,Nifant’ev NE, Bovin NV, Molotkovsky JG, 2000, Antitumor activity of cytotoxic liposomes, equipped with selectin ligand SiaLeX, in mouse mammary adenocarcinoma. Eur J Cancer 36:942–949 Maas RA, Dullens HFJ, De Jong WH, Den Otter W, 1989, Immunotherapy of mice with a large burden of disseminated lymphoma with low-dose interleukin-2. Cancer Res 49:7037–7040 Ivanov IB, Gritsenko VA, 2009, Comparative activities of cattle and swine platelet microbicidal proteins. Probiotics Antimicrob Proteins 1:148–151 Korystov YN, Lebedev VV, Shaposhnikova VV, Ermakova NV, Kublik LN, Chailakhyan LM, 2004, The hexapeptide immunofan inhibits the MRP-dependent multidrug resistance of tumor cells. Dokl Biol Sci 395:177–180 Mei HF, Jin XB, Zhu JY, Zeng AH, Wu Q, Lu XM, Li XB, Shen J, 2012, β-defensin 2 as an adjuvant promotes anti-melanoma immune responses and inhibits the growth of implanted murine melanoma in vivo. PLoS One 7(2):e31328., DOI 10.1371/journal.pone.0031328 Papo N, Shai Y, 2007, Host defense peptides as new weapons in cancer treatment. Cell Mol Life Sci 62:784–790 Struyf S, Van Damme J, 2007, The role of plasma chemokines in cancer. Verh K Acad Geneeskd Belg 69(3):149–165 Coller BS, 2012, A brief history of ideas about platelets in health and disease. In: Michelson AD, ed, Platelets. Academic, New York, pp xix–liv Yeaman MR, Yount NY, Waring AJ, Gank KD, Kupferwasser D, Wiese R, Bayer AS, Welch WH, 2007, Modular determinants of antimicrobial activity in platelet factor-4 family kinocidins. Biochim Biophys Acta 1768:609–619 Krijgsveld J, Zaat SA, Meeldijk J, van Veelen PA, Fang G, Poolman B, Brandt E, Ehlert JE, Kuijpers AJ, Engbers GH, Feijen J, Dankert J, 2000, Thrombocidins, microbicidal proteins from human blood platelets, are C-terminal deletion products of CXC chemokines. J Biol Chem 275:20374–20381 Vandercappellen J, Van Damme J, Struyf S, 2011, The role of the CXC chemokines platelet factor-4, CXCL4/PF-4, and its variant, CXCL4L1/PF-4var, in inflammation, angiogenesis and cancer. Cytokine Growth Factor Rev 22(1):1–18., DOI 10.1016/j.cytogfr. 2010.10.011 Lippi G, Favaloro EJ, 2010, Recombinant platelet factor 4: a therapeutic, anti-neoplastic chimera? Semin Thromb Hemost 36(5):558– 569., DOI 10.1055/s-0030-1255450 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 827 EP 830 DI 10.1007/s12253-014-9812-8 PG 4 ER PT J AU Ata, A Polat, A Serinsoz, E Sungur, AM Arican, A AF Ata, Alper Polat, Ayse Serinsoz, Ebru Sungur, Ali Mehmet Arican, Ali TI Prognostic Value of Increased her2 Expression in Cancers of Pancreas and Biliary Tree SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HER2; Cancers of pancreas and biliary tract; Survival ID HER2; Cancers of pancreas and biliary tract; Survival AB Increased HER2 expression has a prognostic, and predictive value in many solid cancer types, predominantly in breast cancer. However the effects of HER2 on survival from cancers of pancreas, gall bladder, cholangiocellular, and ampullary region are not known. In this study, the effects of increased HER2 expression on these types of cancer have been analyzed. Immunohistochemical HER2 staining was performed in 31 (44.9 %) female, and 38 (55.1 %) male patients with a mean age of 65±10 years, and various parameters, mostly survival rates of patients with pancreas (n=30; 43.5 %), gall bladder (n=17; 24.6 %), cholangiocellular (n= 12; 17.4%), and ampullary region (n=10; 14.5 %) carcinomas were evaluated. Strong (3 +) membranous staining for HER2 was observed in 2 patients with gall bladder cancers (11.76 % of all gall bladder cancers). In 2.90 % of all cases strong membranous staining (2+ or 3+) was observed. Weak (1+) membranous staining was noted in one (3.33 %) pancreatic, and one cholangiocellular (8.33 %) cancer patient, and in none of the ampullary region patient membranous staining for HER2 was observed. Since only scarce number of patients demonstrated membranous staining for HER2, survival analysis was not performed on these patients. Based on cytoplasmic HER2 staining scores, the patients were divided into weakly (0–3 pts; n=17 patients; 24.66 %), moderate (4–5 pts; n=22; 31.88 %), and strongly (6–7 pts; n=30; 43.46 %) stained groups. Patients whose specimens demonstrated borderline statistical significant (p=0.052) low staining for HER2 had higher survival rates when compared with other cases. Increased HER2 expression has no prognostic, and predictive value in cancers of pancreas, biliary tract, and ampulla vateri. If HER2 will be evaluated in these types of cancer, membranous, as well as cytoplasmic staining properties should be taken into account. C1 [Ata, Alper] Mersin University School of Medicine, Department of Medical OncologyMersin, Turkey. [Polat, Ayse] Mersin University School of Medicine, Department of PathologyMersin, Turkey. [Serinsoz, Ebru] Mersin University School of Medicine, Department of PathologyMersin, Turkey. [Sungur, Ali Mehmet] Mersin University School of Medicine, Department of BiostatisticsMersin, Turkey. [Arican, Ali] Mersin University School of Medicine, Department of Medical OncologyMersin, Turkey. RP Ata, A (reprint author), Mersin University School of Medicine, Department of Medical Oncology, Mersin, Turkey. EM dralperata@yahoo.com CR Brenner H, Gondos A, Arndt V, 2007, Recent major progress in long-term cancer patient survival disclosed by modeled period analysis. J Clin Oncol 25:3274–3280 Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55(2):74 Klapper LN, Glathe S, Vaisman N et al, 1999, The ErbB-2/HER2 oncoprotein of human carcinomas may function solely as a shared coreceptor for multiple stroma-derived growth factors. Proc Natl Acad Sci U S A 96:4995 Giatromanolaki A, Koukourakis MI, Simopoulos C et al, 2004, cerbB- 2 related aggressiveness in breast cancer is hypoxia inducible factor-1alpha dependent. Clin Cancer Res 10:7972 Desmedt C, Sperinde J, Piette F et al, 2009, Quantitation of HER2 expression or HER2:HER2 dimers and differential survival in a cohort of metastatic breast cancer patients carefully selected for trastuzumab treatment primarily by FISH. Diagn Mol Pathol 18(1):22–29 Hudis CA, 2007, Trastuzumab—mechanism of action and use in clinical practice. N Engl J Med 357(1):39–51 Blackwell KL, Burstein HJ, Storniolo AM et al, 2010, Randomized study of lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J Clin Oncol 28(7):1124–1130 Saxby AJ, Nielsen A, Scarlett CJ, Clarkson A, Morey A, Gill A, Smith RC, 2005, Assessment of HER-2 status in pancreatic adenocarcinoma: correlation of immunohistochemistry, quantitative realtime RT-PCR, and FISH with aneuploidy and survival. Am J Surg Pathol 29:1125–1134 Lei S, Appert HE, Nakata B, Domenico DR, Kim K, Howard JM, 1995, Overexpression of HER2/neu oncogene in pancreatic cancer correlates with shortened survival. Int J Pancreatol 17:15–21 Koka V, Potti A, KochM, Fraiman G,Mehdi S, Levitt R, 2002, Role of immunohistochemical identification of Her-2/neu and detection of variability in overexpression in pancreatic carcinoma. Anticancer Res 22:1593–1597 Komoto M, Nakata B, Amano R, Yamada N, Yashiro M, Ohira M, WakasaK, Hirakawa K., 2009 Jul)HER2 overexpression correlateswith survival after curative resection of pancreatic cancer. Cancer Sci 100 Yoshimi N, DiGiovanni J, 2001, Constitutive expression of ErbB-2 in gallbladder epithelium results in development of adenocarcinoma. Cancer Res 61:6971–6976 Puhalla H,Wrba F, Kandioler D, Lehnert M, Huynh A, Gruenberger T, Tamandl D, Filipits M, 2007, Expression of p21(Wafl/Cip1), p57(Kip2, and HER2/neu in patients with gallbladder cancer. Anticancer Res 27(3B):1679–1684 Yoshikawa D, Ojima H, Iwasaki M, Hiraoka N, Kosuge T, Kasai S, Hirohashi S, Shibata T, 2008 Jan 29, Clinicopathological and prognostic significance of EGFR, VEGF, and HER2 expression in cholangiocarcinoma. Br J Cancer 98(2):418–25. Epub 2007 Dec 18 Harder J, Waiz O, Otto F, Geissler M, Olschewski M, Weinhold B, Blum HE, Schmitt-Graeff A, Opitz OG, 2009, EGFR and HER2 expression in advanced biliary tract cancer. World J Gastroenterol 15(36):4511–4517 Baumhoer D, Zlobec I, Tornillo L, Dietmaier W, Wuensch PH, Hartmann A, Sessa F, Ruemmele P, Terracciano LM, 2008 Dec, Immunophenotyping and oncogene amplifications in tumors of the papilla of Vater. Virchows Arch 453(6):579–88. Epub 2008 Oct 21 Albarello L, Pecciarini L, Doglioni C, 2011, HER2 testing in gastric cancer. Adv Anat Pathol 18(1):53–59 Wolff AC, Hammond ME, Schwartz JN, 2007, American Society of Clinical Oncology/Collage of American Pathologist Guideline recommendations for human epidermal factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 131:18–43 Tetu B, Brisson J, 1994, Prognostic significance of HER-2/neu oncoprotein expression in node-positive breast cancer. The influence of the pattern of immunostaining and adjuvant therapy. Cancer 73: 2359–2365 Coombs LM, Pigott DA, Sweeney E et al, 1991, Amplification and over-expression of c-erbB-2 in transitional cell carcinoma of the urinary bladder. Br J Cancer 63:601–608 Kay EW, Mulcahy H, Walsh CB et al, 1994, Cytoplasmic c-erbB-2 protein expression correlates with survival in Dukes’ B colorectal carcinoma. Histopathology 25:455–461 Roychowdhury DF, Tseng A, Fu KK et al, 1996, New prognostic factors in nasopharyngeal carcinoma. Tumor angiogenesis and cerbB- 2 expression. Cancer 77:1419–1426 Hall PA, Hughes CM, Staddon SL et al, 1990, The c-erb B-2 protooncogene in human pancreatic cancer. J Pathol 161:195–200 Sugg SL, Ezzat S, Zheng L et al, 1998, Cytoplasmic staining of erbB-2 but not mRNA levels correlates with differentiation in human thyroid neoplasia. Clin Endocrinol, Oxf, 49:629–637 Hornick JL, Lauwers GY, Odze RD, 2005, Immunohistochemistry can help distinguish metastatic pancreatic adenocarcinomas from bile duct adenomas and hamartomas of the liver. Am J Surg Pathol 29(3): 381–389 Moriya T, Kimura W, Semba S, Sakurai F, Hirai I, Ma J, Fuse A, Maeda K, Yamakawa M, 2005, Biological similarities and differences between pancreatic intraepithelial neoplasias and intraductal papillary mucinous neoplasms. Int J Gastrointest Cancer 35(2):111–119 Bergmann F, Moldenhauer G, Herpel E, Gaida MM, Strobel O, Werner J, Esposito I, Muerkoster SS, Schirmacher P, Kern MA, 2010, Expression of L1CAM, COX-2, EGFR, c-KIT and Her2/neu in anaplastic pancreatic cancer: putative therapeutic targets? Histopathology 56(4):440–448 Chaturvedi P, Singh AP, Chakraborty S, Chauhan SC, Bafna S,Meza JL, Singh PK, HollingsworthMA,Mehta PP, Batra SK, 2008 Apr 1, MUC4 mucin interacts with and stabilizes the HER2 oncoprotein in human pancreatic cancer cells. Cancer Res 68(7):2065–70. Erratum in: Cancer Res 2008 May 1;68(9):3550 Tsiambas E, Karameris A, Dervenis C, Lazaris AC, Giannakou N, Gerontopoulos K, Patsouris E, 2006, HER2/neu expression and gene alterations in pancreatic ductal adenocarcinoma: a comparative immunohistochemistry and chromogenic in situ hybridization study based on tissue microarrays and computerized image analysis. JOP 7(3):283–294 Sharif S, Ramanathan RK, Potter D, Cieply K, KrasinskasAM(2008 Nov, HER2 gene amplification and chromosome 17 copy number do not predict survival of patients with resected pancreatic adenocarcinoma. Dig Dis Sci 53(11):3026–32. Epub 2008 May 8 Kaufman M, Mehrotra B, Limaye S,White S, Fuchs A, Lebowicz Y, Nissel-Horowitz S, Thomas A, 2008, EGFR expression in gallbladder carcinoma in North America. Int J Med Sci 5(5):285–291 Su WC, Shiesh SC, Liu HS, Chen CY, Chow NH, Lin XZ, 2001, Expression of oncogene products HER2/Neu and Ras and fibrosisrelated growth factors bFGF, TGF-beta, and PDGF in bile from biliary malignancies and inflammatory disorders. Dig Dis Sci 46(7): 1387–1392 Endo K, Yoon BI, Pairojkul C, Demetris AJ, Sirica AE, 2002, ERBB- 2 overexpression and cyclooxygenase-2 up-regulation in human cholangiocarcinoma and risk conditions. Hepatology 36:439–450 Nakazawa K, Dobashi Y, Suzuki S, Fujii H, Takeda Y, Ooi A, 2005, Amplification and overexpression of c-erbB-2, epidermal growth factor receptor, and c-met in biliary tract cancers. J Pathol 206(3): 356–365 PignochinoY, Sarotto I, Peraldo-Neia C, Penachioni JY, Cavalloni G, MigliardiG, Casorzo L, Chiorino G, RisioM, BardelliA,AgliettaM, Leone F, 2010, Targeting EGFR/HER2 pathways enhances the antiproliferative effect of gemcitabine in biliary tract and gallbladder carcinomas. BMC Cancer 10:631 Haerslev T, Jacobsen GK, 1993, Microwave processing of formalinfixed and paraffin-embedded sections improves the immunoreactivity of c-erbB-2 oncoprotein in breast carcinoma. Appl Immunohistochem 1:223–226 Keshgegian AA, Cnaan Aet al, 1997, erbB-2 oncoprotein expression in breast carcinoma. Poor prognosis associated with high degree of cytoplasmic positivity using CB-11 antibody. Am J Clin Pathol 108: 456–463 Shafizadeh N, Grenert JP, Sahai V, Kakar S, 2010, Apr Epidermal growth factor receptor and HER-2/neu status by immunohistochemistry and fluorescence in situ hybridization in adenocarcinomas of the biliary tree and gallbladder. Hum Pathol 41(4):485–92. Epub 2009 Dec 29 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 831 EP 838 DI 10.1007/s12253-014-9847-x PG 8 ER PT J AU Hata, N Suzuki, OS Murata, H Hatae, R Akagi, Y Sangatsuda, Y Amano, T Yoshimoto, K Tahira, T Mizoguchi, M AF Hata, Nobuhiro Suzuki, O Satoshi Murata, Hideki Hatae, Ryusuke Akagi, Yojiro Sangatsuda, Yuhei Amano, Toshiyuki Yoshimoto, Koji Tahira, Tomoko Mizoguchi, Masahiro TI Genetic Analysis of a Case of Glioblastoma with Oligodendroglial Component Arising During the Progression of Diffuse Astrocytoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE GBMO; IDH1; Loss of heterozygosity; Secondary glioblastoma ID GBMO; IDH1; Loss of heterozygosity; Secondary glioblastoma AB The most recent definition of glioblastoma with oligodendroglioma component (GBMO) assigned clinical significance to the observation of oligodendroglial foci within glioblastomas. However, the pathological mechanism of its histogenesis has not yet been determined. We report the genetic analysis of a GBMO case that evolved from an astrocyte lineage. A 37-year-old male underwent a third craniotomy for the removal of recurrent lesions of a secondary glioblastoma originating from a previous diffuse astrocytoma. The lesion in the right frontal lobe contained oligodendroglial foci within a glioblastoma background, while the remaining lesions showed only classic glioblastoma histology. Genetic analyses revealed distal 10q loss of heterozygosity (LOH) occurring de novo in the oligodendroglial tissue, as well as 10p, 17p LOH, and isocitrate dehydrogenase-1 gene (IDH1) mutations inherited from the previous lesions. The final recurrent glioblastoma underwent LOH on almost the entire of chromosome 10. Based on these results, the importance of an oligodendroglial component in glioblastomas may be limited. C1 [Hata, Nobuhiro] Kyushu University, Graduate School of Medical Sciences, Department of NeurosurgeryFukuoka, Japan. [Suzuki, O Satoshi] Kyushu University, Graduate School of Medical Sciences, Department of NeuropathologyFukuoka, Japan. [Murata, Hideki] Kyushu University, Graduate School of Medical Sciences, Department of NeurosurgeryFukuoka, Japan. [Hatae, Ryusuke] Kyushu University, Graduate School of Medical Sciences, Department of NeurosurgeryFukuoka, Japan. [Akagi, Yojiro] Kyushu University, Graduate School of Medical Sciences, Department of NeurosurgeryFukuoka, Japan. [Sangatsuda, Yuhei] Kyushu University, Graduate School of Medical Sciences, Department of NeurosurgeryFukuoka, Japan. [Amano, Toshiyuki] Kyushu University, Graduate School of Medical Sciences, Department of NeurosurgeryFukuoka, Japan. [Yoshimoto, Koji] Kyushu University, Graduate School of Medical Sciences, Department of NeurosurgeryFukuoka, Japan. [Tahira, Tomoko] Kyushu University, Medical Institute of Bioregulation, Division of Genome Analysis, Research Center for Genetic Information, 3-1-1 Maidashi, Higashi-kuFukuoka, Japan. [Mizoguchi, Masahiro] Kyushu University, Graduate School of Medical Sciences, Department of NeurosurgeryFukuoka, Japan. RP Hata, N (reprint author), Kyushu University, Graduate School of Medical Sciences, Department of Neurosurgery, Fukuoka, Japan. EM hatanobu@kyumed.jp CR Louis DN, Ohgaki H,Wiestler OD, CaveneeWK(eds,, 2007)World health organization classification of tumours of the central nervous system. IARC Press, Lyon Yoshimoto K, Iwaki T, Inamura T, Fukui M, Tahira T, Hayashi K, 2002, Multiplexed analysis of post-PCR fluorescence-labeled microsatellite alleles and statistical evaluation of their imbalance in brain tumors. Jpn J Cancer Res 93:284–290 Hartmann C, Meyer J, Balss J et al, 2009, Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas. Acta Neuropathol 118:469–474 SalvatiM, Formichella AI, D’EliaA, Brogna C, Frati A, Giangaspero F, Delfini R, Santoro A, 2009, Cerebral glioblastoma with oligodendrogliomal component: analysis of 36 cases. J Neuro- Oncol 94:129–134 Cairncross JG, Ueki K, Zlatescu MC et al, 1998, Specific genetic predictors of chemotherapeutic response and survival in patients with anaplastic oligodendrogliomas. J Natl Cancer Inst 90:1473–1479 Nakamura H, Makino K, Kuratsu J-I, 2011, Molecular and clinical analysis of glioblastoma with an oligodendroglial component, GBMO). Brain Tumor Pathol 28:185–190 Hegi ME, Janzer R-C, Lambiv WL et al, 2012, Presence of an oligodendroglioma-like component in newly diagnosed glioblastoma identifies a pathogenetically heterogeneous subgroup and lacks prognostic value: central pathology review of the EORTC_26981/ NCIC_CE.3 trial. Acta Neuropathol 123:841–852 Kraus JA, Lamszus K, Glesmann N et al, 2001, Molecular genetic alterations in glioblastomas with oligodendroglial component. Acta Neuropathol 101:311–320 Jeuken JWM, Sijben A, Bleeker FE, Boots-Sprenger SHE et al, 2011, The nature and timing of specific copy number changes in the course of molecular progression in diffuse gliomas: further elucidation of their genetic “life story”. Brain Pathol 21:308–320 ScheithauerBW, Fuller GN, Vanden Berg SR, 2008, The 2007WHO classification of tumors of the nervous system: controversies in surgical neuropathology. Brain Pathol 18:307–316 Watanabe T, Nobusawa S, Kleihues P, Ohgaki H, 2008, The 2007 WHO classification of tumors of the nervous system: controversies in surgical neuropathology. Brain Pathol 18:307–316 Ichimura K, Schmidt EE, Miyakawa A, Goike HM, Collins VP, 1998, Distinct patterns of deletion on 10p and 10q suggest involvement of multiple tumor suppressor genes in the development of astrocytic gliomas of different malignancy grades. Genes Chromosome Cancer 22:9–15 Hata N, Yoshimoto K, Yokoyama N et al, 2006, Allelic losses of chromosome 10 in glioma tissues detected by quantitative singlestrand conformation polymorphism analysis. Clin Chem 52:370–378 Sasaki H, Betensky RA, Cairncross JG, Louis DN, 2002, DMBT1 polymorphisms: relationship to malignant glioma tumorigenesis. Cancer Res 62:1790–1796 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 839 EP 843 DI 10.1007/s12253-014-9850-2 PG 5 ER PT J AU Jargin, VS AF Jargin, V Sergei TI Renal Cell Carcinoma after Chernobyl: on the Role of Radiation vs. Late Detection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Jargin, V Sergei] Peoples’ Friendship University of Russia, Clementovski per 6-82, 115184 Moscow, Russian Federation. RP Jargin, VS (reprint author), Peoples’ Friendship University of Russia, 115184 Moscow, Russian Federation. EM sjargin@mail.ru CR UNSCEAR, 2008, Report to the General Assembly. Sources and effects of ionizing radiation. Annex D. Health effects due to radiation from the Chernobyl accident. New York, United Nations UNSCEAR, 2010, Report to the General Assembly. Summary of low-dose radiation effects on health. United Nations, New York Mould RF, 2000, The Chernobyl record. The definite history of Chernobyl catastrophe. Institute of Physics, Bristol & Philadelphia Romanenko AM, Ruiz-Sauri A, Morell-Quadreny L, Valencia G, Vozianov AF, Llombart-Bosch A, 2012, Microvessel Density is High in Clear-Cell Renal Cell Carcinomas of Ukrainian Patients Exposed to Chronic Persistent Low-Dose Ionizing Radiation After the Chernobyl Accident. Virchows Arch 460:611–619., DOI 10.1007/ s00428–012–1243-x Romanenko A, Morell-Quadreny L, Ramos D, Nepomnyaschiy V, Vozianov A, Llombart-Bosch A, 2006, Extracellular Matrix Alterations in Conventional Renal Cell Carcinomas by Tissue Microarray Profiling Influenced by the Persistent, Long-Term, low- Dose Ionizing Radiation Exposure in Humans. Virchows Arch 448: 584–590., DOI 10.1007/s00428–006–0160–2 Romanenko A, Morell-Quadreny L, Nepomnyaschy V, Vozianov A, Llombart-Bosch A, 2000, Pathology and Proliferative Activity of Renal-Cell Carcinomas, RCCS, and Renal Oncocytomas in Patients With Different Radiation Exposure After the Chernobyl Accident in Ukraine. Int J Cancer 87:880–883., DOI 10.1002/1097– 0215(20000915)87:6<880::AID-IJC19>3.0.CO;2-J Romanenko A, Morell-Quadreny L, Nepomnyaschy V, Vozianov A, Llombart-Bosch A, 2001, Radiation Sclerosing Proliferative Atypical Nephropathy of Peritumoral Tissue of Renal-Cell Carcinomas After the Chernobyl Accident in Ukraine. Virchows Arch 438:146–153., DOI 10.1007/s004280000334 Morell-Quadreny L, Romanenko A, Lopez-Guerrero JA, Calabuig S, Vozianov A, Llombart-Bosch A, 2011, Alterations of Ubiquitylation and Sumoylation in Conventional Renal Cell Carcinomas After the Chernobyl Accident: a Comparison with Spanish Cases. Virchows Arch 459:307–313., DOI 10.1007/s00428–011–1124–8 Mettler FA Jr, Huda W, Yoshizumi TT, Mahesh M, 2008, Effective Doses in Radiology and Diagnostic Nuclear Medicine: a Catalog. Radiology 248:254–263., DOI 10.1148/radiol.2481071451 Kavantzas N, Paraskevakou H, Tseleni-Balafouta S, Aroni K, Athanassiades P, Agrogiannis G, Patsouris E, 2007, Association Between Microvessel Density and Histologic Grade in Renal Cell Carcinomas. Pathol Oncol Res 13:145–148, PMID: 17607376 Jargin SV, 2007, Over-Estimation of Radiation-InducedMalignancy After the Chernobyl Accident. Virchows Arch 451:105–106., DOI 10. 1007/s00428–007–0428–1 Jargin SV, 2010, Chernobyl-Related Cancer: Re-Evaluation Needed. Turkish J Pathol 26:177–1., DOI 10.5146/tjpath.2010.01021 Romanenko A, Morell-Quadreny L, Ramos D, Nepomnyaschiy V, Vozianov A, Llombart-Bosch A, 2007, Author Reply to: Over-Estimation of Radiation-Induced Malignancy after the Chernobyl Accident. Virchows Arch 451:107–108., DOI 10. 1007/s00428–007–0442–3 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 845 EP 846 DI 10.1007/s12253-014-9787-5 PG 2 ER PT J AU Lee, HS Je, ME Yoo, JN Lee, HS AF Lee, Hak Sung Je, Mi Eun Yoo, Jin Nam Lee, Hyung Sug TI HMCN1, a cell polarity-related gene, is somatically mutated in gastric and colorectal cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Lee, Hak Sung] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong Socho-gu, 137-701 Seoul, South Korea. [Je, Mi Eun] The Catholic University of Korea, College of Medicine, Department of PathologySeoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of PathologySeoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of PathologySeoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, Seoul, South Korea. EM suhulee@catholic.ac.kr CR Vogel BE, Muriel JM, Dong C, Xu X, 2006, Hemicentins: what have we learned from worms? Cell Res 16:872–878 Vogel BE, Hedgecock EM, 2001, Hemicentin, a conserved extracellular member of the immunoglobulin superfamily, organizes epithelial and other cell attachments into oriented line-shaped junctions. Development 128:883–894 Schultz DW, KleinML, Humpert AJ, LuzierCW, Persun V, SchainM, Mahan A, Runckel C, Cassera M, Vittal V, Doyle TM, Martin TM, Weleber RG, Francis PJ, Acott TS, 2003, Analysis of the ARMD1 locus: evidence that a mutation in HEMICENTIN-1 is associated with age-related macular degeneration in a large family. Hum Mol Genet 12:3315–3323 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–1047 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of theKEAP1 gene in common solid cancers. Histopathology 60:943–952 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 847 EP 848 DI 10.1007/s12253-014-9809-3 PG 2 ER PT J AU Oh, RH An, HCh Yoo, JN Lee, HS AF Oh, Rim Hye An, Hyeok Chang Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutations of TAF7L Gene, a Core Component for Transcription by RNA Polymerase II, in Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Oh, Rim Hye] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [An, Hyeok Chang] The Catholic University of Korea, College of Medicine, Department of General SurgerySeoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Gegonne A, Devaiah BN, Singer DS, 2013, TAF7: traffic controller in transcription initiation. Transcription 4:29–33 Kloet SL, Whiting JL, Gafken P, Ranish J, Wang EH, 2012, Phosphorylation-dependent regulation of cyclin D1 and cyclin A gene transcription by TFIID subunits TAF1 and TAF7. Mol Cell Biol 32: 3358–3369 Fukuchi J, Hiipakka RA, Kokontis JM, Nishimura K, Igarashi K, Liao S, 2004, TATA-binding protein-associated factor 7 regulates polyamine transport activity and polyamine analog-induced apoptosis. J Biol Chem 279:29921–29929 Yazarloo F, Shirkoohi R, Mobasheri MB, Emami A, Modarressi MH, 2013, Expression analysis of four testis-specific genes AURKC, OIP5, PIWIL2 and TAF7L in acute myeloid leukemia: a genderdependent expression pattern. Med Oncol 30:368 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 YooNJ, KimHR, KimYR, An CH, Lee SH, 2012, Somaticmutations of the KEAP1 gene in common solid cancers. Histopathology 60:943– 952 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 849 EP 850 DI 10.1007/s12253-014-9832-4 PG 2 ER PT J AU Choi, JY Yoo, JN Lee, HS AF Choi, Jin Youn Yoo, Jin Nam Lee, Hyung Sug TI Mutation of HELLS, a Chromatin Remodeling Gene, Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Choi, Jin Youn] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Becker PB, 2002, HorzW(2002, ATP-dependent nucleosome remodeling. Annu Rev Biochem 71:247–273 Burrage J, Termanis A, Geissner A, Myant K, Gordon K, Stancheva I, 2012, The SNF2 family ATPase LSH promotes phosphorylation of H2AX and efficient repair of DNA double-strand breaks in mammalian cells. J Cell Sci 125:5524–5534 MaQC, EnnisCA, Aparicio S, 2012, Opening Pandora’s box–the new biology of driver mutations and clonal evolution in cancer as revealed by next generation sequencing. Curr Opin Genet Dev 22:3–9 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 YooNJ, KimHR, KimYR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Lee DW, Zhang K, Ning ZQ, Raabe EH, Tintner S, Wieland R, Wilkins BJ, Kim JM, Blough RI, Arceci RJ, 2000, Proliferationassociated SNF2-like gene, PASG): a SNF2 family member altered in leukemia. Cancer Res 60:3612–3622 GoelA, ArnoldCN,NiedzwieckiD,Carethers JM,Dowell JM,Wasserman L, Compton C, Mayer RJ, Bertagnolli MM, Boland CR, 2004, Frequent inactivation of PTEN by promoter hypermethylation in microsatellite instability-high sporadic colorectal cancers. Cancer Res 64:3014–3021 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 851 EP 852 DI 10.1007/s12253-014-9862-y PG 2 ER PT J AU Choi, RM Yoo, JN Lee, HS An, HCh AF Choi, Ryoung Mi Yoo, Jin Nam Lee, Hyung Sug An, Hyeok Chang TI Frameshift Mutation of an Angiogenesis Factor VEGFB and its Mutational Heterogeneity in Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Choi, Ryoung Mi] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [An, Hyeok Chang] The Catholic University of Korea, College of Medicine, Department of General Surgery, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP An, HCh (reprint author), The Catholic University of Korea, College of Medicine, Department of General Surgery, 137-701 Seoul, South Korea. EM achcolo@catholic.ac.kr CR Hanahan D,Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Silvestre JS, Tamarat R, Ebrahimian TG, Le-Roux A, Clergue M, Emmanuel F, Duriez M, Schwartz B, Branellec D, Levy BI, 2003, Vascular endothelial growth factor-B promotes in vivo angiogenesis. Circ Res 93:114–123 Olofsson B, Pajusola K, Kaipainen A, von Euler G, Joukov V, Saksela O, Orpana A, Pettersson RF, Alitalo K, Eriksson U, 1996, Vascular endothelial growth factor B, a novel growth factor for endothelial cells. Proc Natl Acad Sci U S A 93:2576–2581 Dai W, Zeller C, Masrour N, Siddiqui N, Paul J, Brown R, 2013, Promoter CpG island methylation of genes in key cancer pathways associates with clinical outcome in high-grade serous ovarian cancer. Clin Cancer Res 19:5788–5797 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Gylfe AE, Kondelin J, Turunen M, Ristolainen H, Katainen R, Pitkanen E, Kaasinen E, Rantanen V, Tanskanen T, Varjosalo M, Lehtonen H, Palin K, Taipale M, Taipale J, Renkonen-Sinisalo L, Jarvinen H, Bohm J, Mecklin JP, Ristimaki A, Kilpivaara O, Tuupanen S, Karhu A, Vahteristo P, Aaltonen LA, 2013, Identification of candidate oncogenes in human colorectal cancers with microsatellite instability. Gastroenterology 145(3):540–543 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 Calin GA, Gafa R, TibilettiMG, Herlea V, Becheanu G, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression in microsatellite instability high, MSI-H, colon cancers correlates with clinico-pathological parameters: a study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89:230–235 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 853 EP 855 DI 10.1007/s12253-015-9900-4 PG 3 ER PT J AU Choi, JY Yoo, JN Lee, HS AF Choi, Jin Youn Yoo, Jin Nam Lee, Hyung Sug TI Mutation and Expression of a Methyl-Binding Protein 6 (MBD6) in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Choi, Jin Youn] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR WeberM, Schubeler D, 2007, Genomic patterns of DNA methylation: targets and function of an epigenetic mark. Curr Opin Cell Biol 19: 273–280 SuzukiMM, Bird A, 2008, DNAmethylation landscapes: provocative insights from epigenomics. Nat Rev Genet 9:465–476 Jung JS, Jee MK, Cho HT, Choi JI, Im YB, Kwon OH, Kang SK, 2013, MBD6 is a direct target of Oct4 and controls the stemness and differentiation of adipose tissue-derived stem cells. Cell Mol Life Sci 70:711–728 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 YooNJ, KimHR, KimYR, An CH, Lee SH, 2012, Somaticmutations of the KEAP1 gene in common solid cancers. Histopathology 60:943– 952 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 857 EP 858 DI 10.1007/s12253-015-9904-0 PG 2 ER PT J AU Je, ME An, HCh Chung, JY Yoo, JN Lee, HS AF Je, Mi Eun An, Hyeok Chang Chung, Jun Yeun Yoo, Jin Nam Lee, Hyung Sug TI GNAS Mutation Affecting Codon 201 is Rare in Most Human Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE GNAS; Mutation; Cancers ID GNAS; Mutation; Cancers C1 [Je, Mi Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [An, Hyeok Chang] The Catholic University of Korea, College of Medicine, Department of General Surgery, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Chung, Jun Yeun] The Catholic University of Korea, College of Medicine, Department of Microbiology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Landis CA, Masters SB, Spada A, Pace AM, Bourne HR, Vallar L, 1989, GTPase inhibiting mutations activate the alpha chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature 340:692–696 Aldred MA, Trembath RC, 2000, Activating and inactivating mutations in the human GNAS1 gene. Hum Mutat 16:183–189 Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, Mandelker D, Leary RJ, Ptak J, Silliman N, Szabo S, Buckhaults P, Farrell C, Meeh P, Markowitz SD, Willis J, Dawson D, Willson JK, Gazdar AF, Hartigan J, Wu L, Liu C, Parmigiani G, Park BH, Bachman KE, Papadopoulos N, Vogelstein B, Kinzler KW, Velculescu VE, 2006, The consensus coding sequences of human breast and colorectal cancers. Science 314:268–274 Nishikawa G, Sekine S, Ogawa R, Matsubara A, Mori T, Taniguchi H, Kushima R, Hiraoka N, Tsuta K, Tsuda H, Kanai Y, 2013, Frequent GNAS mutations in low-grade appendiceal mucinous neoplasms. Br J Cancer 108:951–958 Nault JC, FabreM, Couchy G, Pilati C, Jeannot E, Tran Van Nhieu J, Saint-Paul MC, De Muret A, Redon MJ, Buffet C, Salenave S, Balabaud C, Prevot S, Labrune P, Bioulac-Sage P, Scoazec JY, Chanson P, Zucman-Rossi J, 2012, GNAS-activating mutations define a rare subgroup of inflammatory liver tumors characterized by STAT3 activation. J Hepatol 56:184–191 Kalfa N, Lumbroso S, Boulle N, Guiter J, Soustelle L, Costa P, Chapuis H, Baldet P, Sultan C, 2006, Activating mutations of Gsalpha in kidney cancer. J Urol 176:891–895 Furukawa T, Kuboki Y, Tanji E, Yoshida S, Hatori T, Yamamoto M, Shibata N, Shimizu K, Kamatani N, Shiratori K, 2011, Wholeexome sequencing uncovers frequent GNAS mutations in intraductal papillary mucinous neoplasms of the pancreas. Sci Rep 1:161 Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia- Manero G, Kantarjian H, Raza A, Levine RL, Neuberg D, Ebert BL, 2011, Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med 364:2496–2506 Ong CK, Subimerb C, Pairojkul C, Wongkham S, Cutcutache I, Yu W, McPherson JR, Allen GE, Ng CC, Wong BH, Myint SS, Rajasegaran V, Heng HL, Gan A, Zang ZJ, Wu Y, Wu J, Lee MH, Huang D, Ong P, Chan-on W, Cao Y, Qian CN, Lim KH, Ooi A, Dykema K, Furge K, Kukongviriyapan V, Sripa B, Wongkham C, Yongvanit P, Futreal PA, Bhudhisawasdi V, Rozen S, Tan P, Teh BT, 2012, Exome sequencing of liver fluke-associated cholangiocarcinoma. Nat Genet 44:690–693 Jung SW, Lee SY, JekarlDW, KimM, LimJ, KimY, Han K, KimYJ, Cho SG, Song J, 2011, Cytogenetic characteristics and prognosis analysis in 231 myelodysplastic syndrome patients from a single institution. Leuk Res 35:735–740 Lee SH, Jeong EG, Soung YH, Lee JW, Yoo NJ, Lee SH, 2008, Absence of GNAS and EGFL6 mutations in common human cancers. Pathology 40:95–97 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2015 VL 21 IS 3 BP 859 EP 860 DI 10.1007/s12253-015-9919-6 PG 2 ER PT J AU Cserni, G Maraz, R AF Cserni, Gabor Maraz, Robert TI Regional Disease Control in Selected Patients with Sentinel Lymph Node Involvement and Omission of Axillary Lymph Node Dissection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Axillary lymph node dissection; Sentinel lymph node; Micrometastasis; Isolated tumour cells/clusters; Follow-up ID Axillary lymph node dissection; Sentinel lymph node; Micrometastasis; Isolated tumour cells/clusters; Follow-up AB Whether an axillary lymph node dissection (ALND) is needed for breast cancer patients with minimal sentinel lymph node (SLN) involvement is arguable despite recent data supporting the omission of axillary clearance in these patients. Data on disease recurrence of 111 patients with SLN involvement and no ALND were analysed. Patients with minimal SLN involvement were assessed for their risk of non- SLN metastasis by means of several nomograms. The series included patients with isolated tumour cells (n=76), micrmetastasis (n=33) and macrometastasis (n=2) who were followed for a median of 37 months (range 12–148 months). Six patients died, 3 of disease and 3 of unrelated causes. Eight further patients had breast cancer related events: 1 local breast recurrence and seven distant metastases. No axillary regional recurrence was detected. Disease related events were not associated with the risk of non-SLN metastasis. The presented data suggest that omitting ALND in patients with low volume SLN metastasis may be a safe procedure, and support the observation that systemic disease recurrence may not be associated with axillary recurrence or the risk of NSLN involvement predicted by nomograms. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, H-6000 Kecskemet, Hungary. [Maraz, Robert] Bacs-Kiskun County Hospital, Department of Surgery, Nyiri ut 38, H-6000 Kecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.hu CR Veronesi U, Viale G, Paganelli G, Zurrida S, Luini A, Galimberti V et al, 2010, Sentinel lymph node biopsy in breast cancer: ten-year results of a randomized controlled study. Ann Surg 251:595–600 Zavagno G, De Salvo GL, Scalco G, Bozza F, Barutta L, Del Bianco P et al, 2008, A randomized clinical trial on sentinel lymph node biopsy versus axillary lymph node dissection in breast cancer: results of the Sentinella/GIVOM Trial. Ann Surg 247:207–213 Andersson Y, de Boniface J, Jonsson PE, Ingvar C, Liljegren G, Bergkvist L et al, 2012, Axillary recurrence rate 5 years after negative sentinel node biopsy for breast cancer. Br J Surg 99:226–231 Hunt KK, Ballman KV, McCall LM, Boughey JC, Mittendorf EA, Cox CE et al, 2012, Factors associated with local-regional recurrence after a negative sentinel node dissection: results of the ACOSOG Z0010 trial. Ann Surg 256:428–436 Krag DN, Anderson SJ, Julian TB, Brown AM, Harlow SP, Costantino JP et al, 2010, Sentinel-lymph-node resection compared with conventional axillary-lymph-node dissection in clinically nodenegative patients with breast cancer: overall survival findings from the NSABP B-32 randomised phase 3 trial. Lancet Oncol 11:927– 933 Cserni G, Burzykowski T, Vinh-Hung V, Kocsis L, Boross G, Sinko M et al, 2004, Axillary sentinel node and tumour-related factors associated with non-sentinel node involvement in breast cancer. Jpn J Clin Oncol 34:519–524 Viale G, Maiorano E, Pruneri G, Mastropasqua MG, Valentini S, Galimberti V et al, 2005, Predicting the risk for additional axillary metastases in patients with breast carcinoma and positive sentinel lymph node biopsy. Ann Surg 241:319–325 Giuliano AE, McCall L, Beitsch P, Whitworth PW, Blumencranz P, Leitch AM et al, 2010, Locoregional recurrence after sentinel lymph node dissection with or without axillary dissection in patients with sentinel lymph node metastases: the American College of Surgeons Oncology Group Z0011 randomized trial. Ann Surg 252:426–432 Sola M, Alberro JA, Fraile M, Santesteban P, Ramos M, Fabregas R et al, 2013, Complete axillary lymph node dissection versus clinical follow-up in breast cancer patients with sentinel node micrometastasis: final results from the multicenter clinical trial AATRM 048/13/2000. Ann Surg Oncol 20:120–127 Galimberti V, Cole BF, Zurrida S, Viale G, Luini A, Veronesi P et al, 2013, Axillary dissection versus no axillary dissection in patients with sentinel-node micrometastases, IBCSG 23-01): a phase 3 randomised controlled trial. Lancet Oncol 14:297–305 Park J, Fey JV, Naik AM, Borgen PI, Van Zee KJ, Cody HS 3rd, 2007, A declining rate of completion axillary dissection in sentinel lymph node-positive breast cancer patients is associated with the use of a multivariate nomogram. Ann Surg 245:462–468 Bilimoria KY, Bentrem DJ, Hansen NM, Bethke KP, Rademaker AW, Ko CYet al, 2009, Comparison of sentinel lymph node biopsy alone and completion axillary lymph node dissection for nodepositive breast cancer. J Clin Oncol 27:2946–2953 Wasif N, Maggard MA, Ko CY, Giuliano AE, 2010, Underuse of axillary dissection for the management of sentinel node micrometastases in breast cancer. Arch Surg 145: 161–166 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ et al, 2011, Strategies for subtypes-dealing with the diversity of breast cancer: highlights of the St. Gallen International expert consensus on the primary therapy of early breast cancer 2011. Ann Oncol 22:1736–1747 AGO, DGS, SGS, OGS, Panelists, Executive BoardMembers, 2013, German, Austrian and Swiss consensus conference on the diagnosis and local treatment of the axilla in breast cancer. Eur J Cancer 49: 2277–2283 Tvedskov TF, JensenMB, Lisse IM, Ejlertsen B, Balslev E, Kroman N, 2012, High risk of non-sentinel node metastases in a group of breast cancer patients with micrometastases in the sentinel node. Int J Cancer 131:2367–2375 Meretoja TJ, Strien L, Heikkila PS, Leidenius MH, 2012, A simple nomogram to evaluate the risk of nonsentinel node metastases in breast cancer patients with minimal sentinel node involvement. Ann Surg Oncol 19:567–576 Houvenaeghel G, Nos C, Giard S,Mignotte H, Esterni B, Jacquemier J et al, 2009, A nomogram predictive of non-sentinel lymph node involvement in breast cancer patients with a sentinel lymph node micrometastasis. Eur J Surg Oncol 35:690–695 Houvenaeghel G, Bannier M, Nos C, Giard S, Mignotte H, Jacquemier J et al, 2012, Non sentinel node involvement prediction for sentinel node micrometastases in breast cancer: nomogram validation and comparison with other models. Breast 21:204–209 Grabau D, Dihge L, FernoM, Ingvar C, Ryden L, 2013, Completion axillary dissection can safely be omitted in screen detected breast cancer patients with micrometastases. A decade’s experience from a single institution. EJSO 39:601–607 Cserni G, Rajtar M, Boross G, Sinko M, Svebis M, Baltas B, 2002, Comparison of vital dye-guided lymphatic mapping and dye plus gamma probe-guided sentinel node biopsy in breast cancer. World J Surg 26:592–597 Cserni G, 2001, The potential value of intraoperative imprint cytology of axillary sentinel lymph nodes in breast cancer patients. Am Surg 67:86–91 Cserni G, 2002, Complete step sectioning of axillary sentinel lymph nodes in patients with breast cancer. Analysis of two different step sectioning and immunohistochemistry protocols in 246 patients. J Clin Pathol 55:926–931 Cserni G, Bezsenyi I, Marko L, 2013, Patients′ choice on axillary lymph node dissection following sentinel lymph node micrometastasis–first report on prospective use of a nomogram in very low risk patients. Pathol Oncol Res 19:211–216 Cserni G, Gregori D, Merletti F, Sapino A, Mano MP, Ponti A et al, 2004)Meta-analysis of non-sentinel nodemetastases associated with micrometastatic sentinel nodes in breast cancer. Br J Surg 91:1245– 1252 van Deurzen CH, de Boer M, Monninkhof EM, Bult P, van der Wall E, Tjan-Heijnen VC et al, 2008)Non-sentinel lymph nodemetastases associated with isolated breast cancer cells in the sentinel node. J Natl Cancer Inst 100:1574–1580 De Mascarel I, MacGrogan G, Debled M, Brouste V, Mauriac L, 2008, Distinction between isolated tumor cells and micrometastases in breast cancer: is it reliable and useful? Cancer 112:1672–1678 Cserni G, Bianchi S, Vezzosi V, van Diest P, van Deurzen C, Sejben I et al, 2008, Variations in sentinel node isolated tumour cells / micrometastasis and non-sentinel node involvement rates according to different interpretations of the TNM definitions. Eur J Cancer 44: 2185–2191 Meretoja TJ, Vironen JH, Heikkila PS, Leidenius MH, 2010, Outcome of selected breast cancer patients with micrometastasis or isolated tumor cells in sentinel node biopsy and no completion axillary lymph node dissection. J Surg Oncol 102: 215–219 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 861 EP 866 DI 10.1007/s12253-015-9899-6 PG 6 ER PT J AU Zhong, Dt Wu, Rp Wang, Xl Huang, XB Lin, Mx Lan, Yq Chen, Q AF Zhong, Dong-ta Wu, Ri-ping Wang, Xin-li Huang, Xiao-Bing Lin, Meng-xin Lan, Yan-qin Chen, Qiang TI Combination Chemotherapy with S-1 and Oxaliplatin (SOX) as First-Line Treatment in Elderly Patients with Advanced Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Advanced gastric cancer; S-1; Oxaliplatin; Elderly; Combination chemotherapy ID Advanced gastric cancer; S-1; Oxaliplatin; Elderly; Combination chemotherapy AB This study is a retrospective analysis evaluating the efficacy and toxicity of combination chemotherapy with S-1 and oxaliplatin (SOX) as first-line treatment in elderly patients with advanced gastric cancer. One hundred and twenty-nine patients with recurrent or metastatic gastric adenocarcinoma were treated with SOX; S-1 (40–60 mg depending on patient’s body surface area) was given orally, twice daily on days 1 to 14 followed by a 7-day rest period, 130 mg/m2 oxaliplatin was given as an intravenous infusion over 2-hours on day one. The cycle was repeated every three weeks. All of the patients were older than 65 years. Among 129 patients enrolled, nine patients could not be evaluated for responses because of the absence of any measurable lesions or early discontinuation of therapy. Assessment of the response of 120 patients was made. The overall objective response rate was 54.2 % (95 %CI, 45.3–63.1 %), with three complete responses and 62 partial responses. The disease control rate was 80.8 % (95 %CI, 73.8–87.8 %). The median follow-up period was 23 months (range, 5–42 months). The median time to progression was 6.9 months (95 %CI, 5.5–8.3 months) and the median overall survival was 12.8 months (95 %CI, 11.4– 14.2 months). The one-year survival rate was 57.5 % (95 %CI, 48.7–66.3 %). In 129 patients assessed safety, grade 3 and 4 toxicities included leucopenia (20.9 %), neutropenia (24.0 %), anemia (10.9 %), thrombocytopenia (10.1 %), anorexia (3.1 %), peripheral neurotoxicity (15.5 %), and fatigue (12.4 %). No treatment-related deaths occurred. Combination chemotherapy with SOX offers an effective, safe and welltolerated regimen for elderly patients with advanced gastric cancer. C1 [Zhong, Dong-ta] Fujian Medical University Union Hospital, Department of Medical Oncology, No. 29 Xinquan Road, 350001 Fuzhou, Fujian, China. [Wu, Ri-ping] Fujian Medical University Union Hospital, Department of Medical Oncology, No. 29 Xinquan Road, 350001 Fuzhou, Fujian, China. [Wang, Xin-li] Fujian Medical University Union Hospital, Department of Medical Oncology, No. 29 Xinquan Road, 350001 Fuzhou, Fujian, China. [Huang, Xiao-Bing] Fujian Medical University Union Hospital, Department of Medical Oncology, No. 29 Xinquan Road, 350001 Fuzhou, Fujian, China. [Lin, Meng-xin] Fujian Medical University Union Hospital, Department of Medical Oncology, No. 29 Xinquan Road, 350001 Fuzhou, Fujian, China. [Lan, Yan-qin] Fujian Medical University Union Hospital, Department of Medical Oncology, No. 29 Xinquan Road, 350001 Fuzhou, Fujian, China. [Chen, Qiang] Fujian Medical University Union Hospital, Department of Medical Oncology, No. 29 Xinquan Road, 350001 Fuzhou, Fujian, China. RP Chen, Q (reprint author), Fujian Medical University Union Hospital, Department of Medical Oncology, 350001 Fuzhou, China. EM cqiang8@189.cn CR Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics, 2009. CA Cancer J Clin 59:225–249 Saito H, Osaki T, Murakami D, Sakamoto T, Kanaji S, Tatebe S, Tsujitani S, Ikeguchi M, 2006, Effect of age on prognosis in patients with gastric cancer. ANZ J Surg 76:458–461 Glimelius B, Ekstrom K, Hoffman K, Graf W, Sjoden PO, Haglund U, Svensson C, Enander LK, Linne T, Sellstrom H, Heuman R, 1997, Randomized comparison between chemotherapy plus best supportive care with best supportive care in advanced gastric cancer. Ann Oncol 8:163–168 Kato T, Shimamoto Y, Uchida J, Ohshimo H, Abe M, Shirasaka T, Fukushima M, 2001, Possible regulation of 5-fluorouracil-induced neuro- and oral toxicities by two biochemical modulators consisting of S-1, a new oral formulation of 5-fluorouracil. Anticancer Res 21: 1705–1712 Koizumi W, Kurihara M, Nakano S, Hasegawa K, 2000, Phase II study of S-1, a novel oral derivative of 5-fluorouracil, in advanced gastric cancer. For the S-1 Cooperative Gastric Cancer Study Group. Oncology 58:191–197 Lee JL, Kang YK, Kang HJ, Lee KH, Zang DY, Ryoo BY, Kim JG, Park SR, KangWK, Shin DB, Ryu MH, Chang HM, Kim TW, Baek JH, Min YJ, 2008, A randomised multicentre phase II trial of capecitabine vs S-1 as first-line treatment in elderly patients with metastatic or recurrent unresectable gastric cancer. Br J Cancer 99:584–590 Rosati G, Ferrara D, Manzione L, 2009, New perspectives in the treatment of advanced or metastatic gastric cancer. World J Gastroenterol 15:2689–2692 Koizumi W, Takiuchi H, Yamada Y, Boku N, Fuse N, Muro K, Komatsu Y, Tsuburaya A, 2010, Phase II study of oxaliplatin plus S-1 as first-line treatment for advanced gastric cancer, G-SOX study). Ann Oncol 21:1001–1005 Park I, Lee JL, RyuMH, Chang HM, Kim TW, Sym SJ, Lee SS, Jang G, Yoo C, Bae KS, Kang YK, 2010, Phase I/II and pharmacokinetic study of S-1 and oxaliplatin in previously untreated advanced gastric cancer. Cancer Chemother Pharmacol 65:473–480 van Cutsem E, Moiseyenko VM, Tjulandin S, Majlis A, Constenla M, Boni C, Rodrigues A, Fodor M, Chao Y, Voznyi E, Risse ML, Ajani JA, 2006, Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group. J Clin Oncol 24:4991–4997 Kang YK, KangWK, Shin DB, Chen J, Xiong J,Wang J, Lichinitser M, Guan Z, Khasanov R, Zheng L, Philco-Salas M, Suarez T, Santamaria J, Forster G, McCloud PI, 2009, Capecitabine/cisplatin versus 5-fluorouracil/cisplatin as first-line therapy in patients with advanced gastric cancer: a randomized phase III noninferiority trial. Ann Oncol 20(4):666–673 Cunningham D, Starling N, Rao S, Iveson T, Nicolson M, Coxon F, Middleton G, Daniel F, Oates J, Norman AR, 2008, Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med 358:36–46 Al-Batran SE, Hartmann JT, Probst S, Schmalenberg H, Hollerbach S, Hofheinz R, Rethwisch V, Seipelt G, Homann N, Wilhelm G, Schuch G, Stoehlmacher J, Derigs HG, Hegewisch-Becker S, Grossmann J, Pauligk C, Atmaca A, Bokemeyer C, Knuth A, Jager E, 2008, Phase III trial in metastatic gastroesophageal adenocarcinoma with fluorouracil, leucovorin plus either oxaliplatin or cisplatin: a study of the Arbeitsgemeinschaft Internistische Onkologie. J Clin Oncol 26:1435–1442 Hirata K, Horikoshi N, Tominaga K, Sohma K, Yamaguchi K, Okazaki M, Furuhata T, Sasaki K, Nakano Y, Ishizuka H, Yamada Y, Uno S, Taguchi T, Yamamitsu S, Shirasaka T, 2006, Pharmacokinetics of S-1. Gan To Kagaku Ryoho 33: 27–35 Koizumi W, Narahara H, Hara T, Takagane A, Akiya T, Takagi M, Miyashita K, Nishizaki T, Kobayashi O, Takiyama W, Toh Y, Nagaie T, Takagi S, Yamamura Y, Yanaoka K, Orita H, Takeuchi M, 2008, S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer, SPIRITS trial): a phase III trial. Lancet Oncol 9:215–221 Boku N, Yamamoto S, Fukuda H, Shirao K, Doi T, Sawaki A, Koizumi W, Saito H, Yamaguchi K, Takiuchi H, Nasu J, Ohtsu A, 2009, Fluorouracil versus combination of irinotecan plus cisplatin versus S-1 in metastatic gastric cancer: a randomised phase 3 study. Lancet Oncol 10:1063–1069 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 867 EP 873 DI 10.1007/s12253-015-9903-1 PG 7 ER PT J AU Quan, L Qiu, T Liang, J Li, M Zhang, Y Tao, K AF Quan, Liangliang Qiu, Tao Liang, Jiulong Li, Miao Zhang, Yu Tao, Kai TI Identification of Target Genes Regulated by KSHV miRNAs in KSHV-Infected Lymphoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Kaposi’s sarcoma-associated herpesvirus; Gene; microRNAs; Functional annotation ID Kaposi’s sarcoma-associated herpesvirus; Gene; microRNAs; Functional annotation AB This study aimed to identify target genes regulated by KSHV miRNAs in KSHV-infected lymphoma cells. Original Ago HITS-CLIP data of BC-3 and BCBL-1 cell lines were downloaded from SRA database in NCBI, including mRNA and miRNA samples. The raw mRNA reads were mapped into human reference genome hg19 via TopHat for read alignment. PCR duplicates were removed via the SAM tool and the peaks of reads were analyzed via Cufflinks. For miRNA data, the raw data were mapped to the mature miRNA sequences based on miRBase via Bowtie. Peak intersection was computed by using intersectBed in BEDtools. Then, the mature miRNA seeds were identified and then were aligned with 3’ UTR merged peaks. The regulationships of miRNAs and their corresponding genes were analyzed based on the signal of RNA-induced silencing complex. Totally, 7 KSHV-related genes regulated by KSHV miRNAs were identified, including IPO5, EDA, NT5C3, WSB1, KCNS1, PRAM1 and MTRNR2L6. Among them, EDA, MTRNR2L6 and IPO5 were regulated by multiple KSHV miRNAs, such as kshv-miR-K12-1-5p, kshv-miR-K12-4-3p and kshv-miR-K12-3-5p, respectively. Furthermore, expression of kshv-miR-K12-1-5p and kshv-miR-K12-3-5p in BCBL-1 cell line were lower than that in BC-3 cell line, conversely, expression of kshv-miR-K12-4-3p in BCBL-1 cell line were higher than that in BC-3 cell line. In conclusion, potential target genes regulated by KSHV miRNAs in KSHV-infected lymphoma cells might play key roles in the nosogenesis of this disease. These findings might provide the basis for deep understanding of KSHV-infected tumors and further molecular experiments. C1 [Quan, Liangliang] General Hospital of Shenyang Military Area Command, Department of Plastic Surgery, No. 83 Wenhua Road, Shenhe District, 110016 Shenyang, Liaoning Province, China. [Qiu, Tao] General Hospital of Shenyang Military Area Command, Department of Plastic Surgery, No. 83 Wenhua Road, Shenhe District, 110016 Shenyang, Liaoning Province, China. [Liang, Jiulong] General Hospital of Shenyang Military Area Command, Department of Plastic Surgery, No. 83 Wenhua Road, Shenhe District, 110016 Shenyang, Liaoning Province, China. [Li, Miao] General Hospital of Shenyang Military Area Command, Department of Plastic Surgery, No. 83 Wenhua Road, Shenhe District, 110016 Shenyang, Liaoning Province, China. [Zhang, Yu] General Hospital of Shenyang Military Area Command, Department of Plastic Surgery, No. 83 Wenhua Road, Shenhe District, 110016 Shenyang, Liaoning Province, China. [Tao, Kai] General Hospital of Shenyang Military Area Command, Department of Plastic Surgery, No. 83 Wenhua Road, Shenhe District, 110016 Shenyang, Liaoning Province, China. RP Tao, K (reprint author), General Hospital of Shenyang Military Area Command, Department of Plastic Surgery, 110016 Shenyang, China. EM kaitaodr@163.com CR Cesarman E, Chang Y, Moore PS, Said JW, Knowles DM, 1995, Kaposi’s sarcoma–associated herpesvirus-like DNA sequences in AIDS-related body-cavity–based lymphomas. N Engl J Med 332: 1186–1191 Lin X, Li X, Liang D, Lan K, 2012, MicroRNAs and unusual small RNAs discovered in Kaposi’s sarcoma-associated herpesvirus virions. J Virol 86:12717–12730 Gottwein E, Mukherjee N, Sachse C et al, 2007, A viral microRNA functions as an orthologue of cellular miR-155. Nature 450:1096– 1099 Hansen A, Henderson S, Lagos D et al, 2010, KSHV-encoded miRNAs target MAF to induce endothelial cell reprogramming. Genes Dev 24:195–205 Gottwein E, Cullen BR, 2010, A human herpesvirus microRNA inhibits p21 expression and attenuates p21-mediated cell cycle arrest. J Virol 84:5229–5237 Haecker I, Gay LA, Yang Y et al, 2012, Ago HITS-CLIP expands understanding of Kaposi’s sarcoma-associated herpesvirus miRNA function in primary effusion lymphomas. PLoS Pathog 8:23 Trapnell C, Pachter L, Salzberg SL, 2009, TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25:1105–1111 Li H, Handsaker B,Wysoker A et al, 2009, The sequence alignment/ map format and SAMtools. Bioinformatics 25:2078–2079 Trapnell C, Williams BA, Pertea G et al, 2010, Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoformswitching during cell differentiation. Nat Biotechnol 28:511– 515 Langmead B, Trapnell C, Pop M, Salzberg SL, 2009, Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 10:R25 Quinlan AR, Hall IM, 2010, BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26:841–842 DaWei Huang BTS, Lempicki RA, 2008, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4:44–57 Ashburner M, Ball CA, Blake JA et al, 2000, Gene Ontology: tool for the unification of biology. Nat Genet 25:25–29 Punj V,Matta H, Chaudhary PM(2010, X-linked ectodermal dysplasia receptor is downregulated in breast cancer via promoter methylation. Clin Cancer Res 16:1140–1148 Keller SA, Schattner EJ, Cesarman E, 2000, Inhibition of NF-κB induces apoptosis of KSHV-infected primary effusion lymphoma cells. Blood 96:2537–2542 De Oliveira DE, Ballon G, Cesarman E, 2010, NF-κB signaling modulation by EBVand KSHV. Trends Microbiol 18:248–257 Aggarwal BB, 2000, Tumour necrosis factors receptor associated signalling molecules and their role in activation of apoptosis, JNK and NF-κB. Ann Rheum Dis 59:i6–i16 Lei X, Bai Z, Ye F et al, 2010, Regulation of NF-κB inhibitor IκBα and viral replication by a KSHV microRNA. Nat Cell Biol 12:193– 199 Bodzioch M, Lapicka-Bodzioch K, Zapala B, KamyszW, Kiec-Wilk B, Dembinska-Kiec A, 2009, Evidence for potential functionality of nuclearly-encoded humanin isoforms. Genomics 94:247–256 Hashimoto Y, Niikura T, Tajima H et al, 2001, A rescue factor abolishing neuronal cell death by a wide spectrum of familial Alzheimer’s disease genes and Aβ. Proc Natl Acad Sci U S A 98: 6336–6341 Guo B, Zhai D, Cabezas E et al, 2003, Humanin peptide suppresses apoptosis by interfering with Bax activation. Nature 423:456–461 Luciano F, Zhai D, Zhu X et al, 2005, Cytoprotective peptide humanin binds and inhibits proapoptotic Bcl-2/Bax family protein BimEL. J Biol Chem 280:15825–15835 Skalsky RL, Samols MA, Plaisance KB et al, 2007, Kaposi’s sarcoma-associated herpesvirus encodes an ortholog of miR-155. J Virol 81:12836–12845 Gorlich D, Mattaj IW, 1996, Nucleocytoplasmic transport. Science 271:1513–1519 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 875 EP 880 DI 10.1007/s12253-015-9902-2 PG 6 ER PT J AU Emami, N Saadat, I Omidvari, Sh AF Emami, Naghmeh Saadat, Iraj Omidvari, Shahpour TI Susceptibility to Colorectal Cancer and Two Genetic Polymorphisms of XRCC4 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Polymorphisms; XRCC4 ID Colorectal cancer; Polymorphisms; XRCC4 AB The X-ray complementing group 4 (XRCC4, OMIM: 194363) plays a key role in non-homologous endjoining DNA repair pathway in mammalian cells. This pathway is believed to help maintain genomic stability. In the present study, it is hypothesized that genetic polymorphisms in the NHEJ repair XRCC4 gene may be associated with an increased risk in developing colorectal cancer (CRC). We genotyped two polymorphisms of XRCC4, G-1394T (rs6869366) and intron 3 insertion/deletion (I/D; rs28360071) in 200 colorectal cancer patients as well as 256 healthy individuals, and evaluated their association with CRC. We found that in G-1394T polymorphism, neither the TG nor the GG genotypes (versus the TT genotype) were associated with the risk of developing CRC. The results of our study indicate that in comparison with the II genotype, ID and DD genotypes had no significant association with the risk of developing CRC. Subjects with TT genotype and positive family history in colorectal cancer were found to be at a much lower risk of developing CRC in comparison with the reference group (OR=0.31, 95%CI: 0.11–0.85, P=0.023). It should be noted that participants having at least one G allele (TG+GG genotypes) were at a significantly higher risk to develop the disease compared with the reference group (OR=9.10, 95%CI: 2.00–41.3, P=0.004). In relation to I/D polymorphism, among participants, those with positive family history, either with ID (OR=4.78, 95%CI: 2.26–10.0, P<0.001) or DD genotypes (OR=5.73, 95%CI: 1.99–16.4, P=0.001) had a significantly association with the disease. Among participants with a positive family history in CRC, the haplotype GD dramatically increased the risk of developing CRC (OR=10.2, 95%CI: 2.28–46, P=0.002). The results of this study indicate that G-1394T and I/D polymorphisms of XRCC4 among individuals with positive family history for colorectal cancer substantially increase the risk factor for developing colorectal cancers. C1 [Emami, Naghmeh] Shiraz University, College of Sciences, Department of Biology, 71454 Shiraz, Iran. [Saadat, Iraj] Shiraz University, College of Sciences, Department of Biology, 71454 Shiraz, Iran. [Omidvari, Shahpour] Shiraz University of Medical Sciences, Department of ChemotherapyShiraz, Iran. RP Saadat, I (reprint author), Shiraz University, College of Sciences, Department of Biology, 71454 Shiraz, Iran. EM isaadat@shirazu.ac.ir CR Doll R, Peto R, 1981, The causes of cancer: quantitative estimates of avoidable risks of cancer in the United States today. J Natl Cancer Inst 66:1191–1308 Thomas HJ, 1993, Familial colorectal cancer. BMJ 307:277–278 Giovannucci E, Colditz GA, Stampfer MJ, Hunter D, Rosner BA, Willett WC, Speizer FE, 1994, A prospective study of cigarette smoking and risk of colorectal adenoma and colorectal cancer in U.S. women. J Natl Cancer Inst 86:192–199 Giovannucci E, Rimm EB, Stampfer MJ, Colditz GA, Ascherio A, Kearney J, Willett WC, 1994, A prospective study of cigarette smoking and risk of colorectal adenoma and colorectal cancer in U.S. men. J Natl Cancer Inst 86:183–191 Van Gent DC, Hoeijmakers JH, Kanaar R, 2001, Chromosomal stability and the DNA double-stranded break connection. Nat Rev Genet 2:196–206 West SC, 2003, Molecular views of recombination proteins and their control. Nat Rev Mol Cell Biol 4:435–445 YanoK,Morotomi-Yano K, Adachi N, Akiyama H, 2009, Molecular mechanism of protein assembly on DNA double-strand breaks in the non-homologous end-joining pathway. J Radiat Res 50:97–108 Li Z, Otevrel T, GaoY, Cheng HL, Seed B, Stamato TD, Taccioli GE, Alt FW, 1995, The XRCC4 gene encodes a novel protein involved in DNA double-strand break repair and V, D, J recombination. Cell 83: 1079–1089 Critchlow SE, Bowater RP, Jackson SP, 1997, Mammalian DNA double-strand break repair protein XRCC4 interacts with DNA ligase IV. Curr Biol 7:588–598 Chen L, Trujillo K, Sung P, Tomkinson AE, 2000, Interactions of the DNA ligase IV-XRCC4 complex with DNA ends and the DNAdependent protein kinase. J Biol Chem 275:26196–26205 Chang CH, Chiu CF, Wang HC, Wu HC, Tsai RY, Tsai CW, Wang RF, Wang CH, Tsou YA, Bau DT, 2009, Significant association of ERCC6 single nucleotide polymorphisms with bladder cancer susceptibility in Taiwan. Anticancer Res 29: 5121–5124 Bau DT, Fu YP, Chen ST, Cheng TC,Yu JC,Wu PE, Shen CY, 2004, Breast cancer risk and the DNA double-strand break end joining capacity of non homologous end-joining genes are affected by BRCA1. Cancer Res 64:5013–5019 Bau DT, Mau YC, Ding SL, Wu PE, Shen CY, 2007, DNA double strand break repair capacity and risk of breast cancer. Carcinogenesis 28:1726–1730 Chiu CF,Wang HC,Wang CH,Wang CL, Lin CC, Shen CY, Chiang SY, Bau DT, 2008, A new single nucleotide polymorphism in XRCC4 gene is associated with breast cancer susceptibility in Taiwanese patients. Anticancer Res 28:267–270 Chiu CF, Wang CH, Wang CL, Lin CC, Hsu NY, Weng JR, Bau DT, 2008, A novel single nucleotide polymorphism in XRCC4 gene is associated with gastric cancer susceptibility in Taiwan. Ann Surg Oncol 15:514–518 Chiu CF, Tsai MH, Tseng HC, Wang CL, Wang CH, Wu CN, Lin CC, Bau DT, 2008, A novel single nucleotide polymorphism in XRCC4 gene is associated with oral cancer susceptibility in Taiwanese patients. Oral Oncol 44:898–902 Bau DT, Yang MD, Tsou YA, Lin SS, Wu CN, Hsieh HH, Wang RF, Tsai CW, Chang WS, Hsieh HM, Sun SS, Tsai RY, 2010, Colorectal cancer and genetic polymorphism of DNA doublestrand break repair gene XRCC4 in Taiwan. Anticancer Res 30: 2727–2730 Rafiee L, Saadat I, Saadat M, 2010, Glutathione S-transferase genetic polymorphisms, GSTM1, GSTT1 and GSTO2, in three Iranian populations. Mol Biol Rep 37:155–158 Mohamadynejad P, Saadat M, 2008, Genetic polymorphisms of XRCC1, at codons 194 and 399, in Shiraz population, Fars province, southern Iran). Mol Biol Rep 35:669–672 Bazrgar M, Karimi M, Fathzadeh M, Senemar S, Peiravian F, Shojaee A, Saadat M, 2008, Apolipoprotein E polymorphism in Southern Iran: E4 allele in the lowest reported amounts. Mol Biol Rep 35:495–499 Wu KH, Wang CH, Yang YL, Peng CT, Lin WD, Tsai FJ, Lin DT, Bau DT, 2010, Significant association of XRCC4 single nucleotide polymorphisms with childhood leukemia in Taiwan. Anticancer Res 30:529–533 Chang CH, Chiu CF, Wu HC, Tseng HC, Wang CH, Lin CC, Tsai CW, Liang SY, Wang CL, Bau DT, 2008, Significant association of XRCC4 single nucleotide polymorphisms with prostate cancer susceptibility in Taiwanese males. Mol Med Rep 1:525–530 Chang CH, Chang CL, Tsai CW, Wu HC, Chiu CF, Wang RF, Liu CS, Lin CC, Bau DT, 2009, Significant association of an XRCC4 single nucleotide polymorphism with bladder cancer susceptibility in Taiwan. Anticancer Res 29:1777–1782 Hsu NY, Wang HC, Wang CH, Chang CL, Chiu CF, Lee HZ, Tsai CW, Bau DT, 2009, Lung cancer susceptibility and genetic polymorphism of DNA repair gene XRCC4 in Taiwan. Cancer Biomark 5: 159–165 Mandal RK, Singh V, Kapoor R, Mittal RD, 2011, Do polymorphisms in XRCC4 influence prostate cancer susceptibility in North Indian population? Biomarkers 16:236–242 Lovett E, 1976, Family studies in cancer of the colon and rectum. Br J Surg 63:13–18 Fuchs CS, Giovannucci EL, Colditz GA, Hunter DJ, Speizer FE, Willett WC, 1994, A prospective study of family history and the risk of colorectal cancer. N Engl J Med 331:1669–1674 St John DJ, McDermott FT, Hopper JL, Debney EA, Johnson WR, Hughes ES, 1993, Cancer risk in relatives of patients with common colorectal cancer. Ann Intern Med 118:785–790 Taber JM, Aspinwall LG, Heichman KA, Kinney AY, 2014, Preferences for blood-based colon cancer screening differ by race/ethnicity. Am J Health Behav 38:351–361 Harmon BE, Little MA, Woekel ED, Ettienne R, Long CR, Wilkens LR, Le Marchand L, Henderson BE, Kolonel LN, Maskarinec G, 2014, Ethnic differences and predictors of colonoscopy, prostate-specific antigen, and mammography screening participation in the multiethnic cohort. Cancer Epidemiol 38: 162–167 Saadat M, Ansari-Lari M, 2009, Polymorphism of XRCC1, at codon 399, and susceptibility to breast cancer, a meta-analysis of the literatures. Breast Cancer Res Treat 115:137–144 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 881 EP 885 DI 10.1007/s12253-015-9905-z PG 5 ER PT J AU Eduardo, GDPM Campaner, BA Silva, ALGM AF Eduardo, Gamba De Paula Mariana Campaner, Bittencourt Adriana Silva, Antonieta Longo Galvao Maria TI Apoptosis Phenomena in Squamous Cell Carcinomas and Adenocarcinomas of the Uterine Cervix SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Apoptosis; Uterine cervix; Squamous cell carcinoma; Adenocarcinoma; Bcl-2 protein ID Apoptosis; Uterine cervix; Squamous cell carcinoma; Adenocarcinoma; Bcl-2 protein AB To investigate the relationship between apoptosis and histologic types in invasive squamous cell carcinoma and adenocarcinoma of the uterine cervix. The present study involved the assessment of surgical specimens from 74 women with cervical carcinomas FIGO stage IB1 (54 squamous cell carcinomas and 20 adenocarcinomas). The study samples were obtained from selected paraffin blocks containing specimens from patients submitted to surgical procedures. The respective medical charts of patients were reviewed and epidemiologic, clinical and disease-related data were collected. Cervical specimens were assessed by the immunohistochemistry technique using the Bcl-2 protein as a marker. The reactions were considered positive when the cells became stained in brown color. Bcl-2 positive cells were counted in 10 fields under a high magnification (400x) using light microscopy, in the slides area containing squamous carcinoma and adenocarcinoma of the cervix. The total cell count was expressed as the number of positive Bcl-2 cells per mm2. No significant difference in the number of cells marked by the Bcl-2 protein was found for the variables age, tumor diameter, angiolymphatic invasion or number of lymph nodes affected. Comparison of the number of cells marked by the Bcl-2 protein in the two histological groups revealed a statistically significant difference, with squamous tumors presenting a greater number of marked cells. Squamous cervical tumors present a greater number of positive Bcl-2 cells per mm2, suggesting that that the rate of cell death in squamous cell carcinomas of the cervix is lower than in adenocarcinomas. C1 [Eduardo, Gamba De Paula Mariana] Santa Casa de Sao Paulo, School of Medicine, Department of Obstetrics and Gynecology, R. Cesario Mota Jr, 112 - Vila Buarque, 01221-020 Sao Paulo, SP, Brazil. [Campaner, Bittencourt Adriana] Santa Casa de Sao Paulo, School of Medicine, Department of Obstetrics and Gynecology, R. Cesario Mota Jr, 112 - Vila Buarque, 01221-020 Sao Paulo, SP, Brazil. [Silva, Antonieta Longo Galvao Maria] Santa Casa de Sao Paulo, School of Medicine, Department of Pathology, Rua Cesario Mota Jr 112 - Vila Buarque, 01221-020 Sao Paulo, SP, Brazil. RP Campaner, BA (reprint author), Santa Casa de Sao Paulo, School of Medicine, Department of Obstetrics and Gynecology, 01221-020 Sao Paulo, Brazil. EM abcampaner@terra.com.br CR Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55(2):74–108 Maucort-Boulch D, Franceschi S, Plummer M, IARC HPV Prevalence Surveys Study Group, 2008, International correlation between human papillomavirus prevalence and cervical cancer incidence. Cancer Epidemiol Biomarkers Prev 17(3):717–720 World Health Organization, 2008, WHO. World Cancer Report, 2008. International Agency for Research on Cancer, Lyon Smith HO, Tiffany MF, Qualis CR, Key CR, 2000, The rising incidence of adenocarcinoma relative to squamous cell carcinoma of the uterine cervix in the United States- a 24-year population-based study. Gynecol Oncol 78(2):97–105 Tjalma WA, Van Waes TR, Van den Eeden LEM, Bogers JJPM, 2005, Role of human papillomavirus in the carcinogenesis of squamous cell carcinoma and adenocarcinoma of the cervix. Best Pract Res Clin Obstet Gynaecol 19(4):469–483 Vinh-Hung V, Bourgain C, Vlastos G, Cserni G, De Ridder M, Storme G, Vlastos AT, 2007, Prognostic value of histopathology and trends in cervical cancer: a SEER population study. BMC Cancer 7:164 Mathew A, George PS, 2009, Trends in incidence and mortality rates of squamous cell carcinoma and adenocarcinoma of cervix–worldwide. Asian Pac J Cancer Prev 10(4):645–650 Berrington deGonzalez A, Green J, 2007, Comparison of risk factors for invasive squamous cell carcinoma and adenocarcinoma of the cervix: collaborative reanalysis of individual data on 8,097 women with squamous cell carcinoma and 1,374 women with adenocarcinoma from 12 epidemiological studies. Int J Cancer 120(11):2525 Bosch FX, de Sanjose S, 2007, The epidemiology of human papillomavirus infection and cervical cancer. Dis Markers 23(4):213–227 Fregnani JH, Soares FA, Novik PR, Lopes A, Latorre MR, 2008, Comparison of biological behavior between early-stage adenocarcinoma and squamous cell carcinoma of the uterine cervix. Eur J Obstet Gynecol Reprod Biol 136(2):215–223 Hopkins MP, Morley GW, 1991, A comparison of adenocarcinoma and squamous cell carcinoma of the cervix. Obstet Gynecol 77:912– 917 Shingleton HM, Bell MC, Fremgen A et al, 1995, Is there really a difference in survival of women with squamous cell carcinoma, adenocarcinoma, and adenosquamous cell carcinoma of the cervix? Cancer 76(10 Suppl):1948–1955 Eifel PJ, Burke TW,Morris M, Smith TL, 1995, Adenocarcinoma as an independent risk factor for disease recurrence in patients with stage IB cervical carcinoma. Gynecol Oncol 59:38–44 Look KY, Brunetto VL, Clarke-Pearson DL et al, 1996, An analysis of cell type in patients with surgically staged stage IB carcinoma of the cervix: a Gynecologic Oncology Group study. Gynecol Oncol 63: 304–311 Ayhan A, Al RA, Baykal C, Demirtas E, Yuce K, Ayhan A, 2004, A comparison of prognoses of FIGO stage IB adenocarcinoma and squamous cell carcinoma. Int J Gynecol Cancer 14:279–285 Elmore S, 2007, Apoptosis: a review of programmed cell death. Toxicol Pathol 35(4):495–516 Fulda S, Gorman AM, Hori O, Samali A, 2010, Cellular stress responses: cell survival and cell death. Int J Cell Biol 2010:214074 FIGO, 2009, Committee on Gynecologic Oncology. Revised FIGO staging for carcinoma of the vulva, cervix, and endometrium. Int J Gynecol Obstet 105:103–104 Campaner AB, Longo GalvaoMA, 2009, Application of an easy and useful morphometric technique for immunohistochemistry counting. Gynecol Oncol 112(1):282–283 D’Amelio M, Tino E, Cecconi F, 2008, The apoptosome: emerging and new potential targets for drug design. Pharm Res 25:740–751 Okun I, Balakin KV, Tkachenko SE, Ivachtchenko AV, 2008, Caspase activity modulators as anticancer agents. Anticancer Agents Med Chem 8(3):322–341 Gewies A, 2003, Introduction of apoptosis. ApoReview. [serial online] 1–26. Available from: http://www.celldeath.de/encyclo/aporev/ aporev.htm [2006 June 7] KangMH, Reynolds CP, 2009, Bcl-2 inhibitors: targeting mitochondrial apoptotic pathways in cancer therapy. Clin Cancer Res 15(4): 1126–1132 Tjalma WA, Weyler JJ, Bogers JJ, Pollefliet C, Baay M, Goovaerts GC, Vermorken JB, van Dam PA, van Marck EA, Buytaert PM, 2001, The importance of biological factors, bcl-2, bax, p53, PCNA, MI, HPVand angiogenesis, in invasive cervical cancer. Eur J Obstet Gynecol Reprod Biol 97(2):223–230 Borner C, 2003, The Bcl-2 protein family: sensors and checkpoints for life-or-death decisions. Mol Immunol 39(11):615–647 Kallel-Bayoudh I, Hassen HB, Khabir A, Boujelbene N, Daoud J, Frikha M, Sallemi-Boudawara T, Aifa S, Rebai A, 2010, Bcl-2 expression and triple negative profile in breast carcinoma. Med Oncol. [epub ahead of print] Kokawa K, Shikone T, Otani T, Nakano R, 1999, Apoptosis and the expression of Bax and Bcl-2 in squamous cell carcinoma and adenocarcinoma of the uterine cervix. Cancer 85(8):1799–1809 Dimitrakakis C, Kymionis G, Diakomanolis E, Papaspyrou I, Rodolakis A, Arzimanoglou I, Leandros E, Michalas S, 2000, The possible role of p53 and bcl-2 expression in cervical carcinomas and their premalignant lesions. Gynecol Oncol 77(1):129–136 Yao JF, Zhou CY, Wei LF, Wang SY, Shi YF, 2008, Expression of aquaporin-8 and bcl-2 protein in human cervical carcinoma and their correlations. Zhonghua Fu Chan Ke Za Zhi 43(3):205–208 Stergiou L, Hengartner MO, 2004, Death and more: DNA damage response pathways in the nematode C. elegans. Cell Death Differ 11(1):21–28 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 887 EP 892 DI 10.1007/s12253-015-9906-y PG 6 ER PT J AU Alshenawy, AH AF Alshenawy, AlSaeid Hanan TI Immunohistochemical Panel for Differentiating Renal Cell Carcinoma with Clear and Papillary Features SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Renal cell carcinoma; CK7; Carbonic anhydrase IX; α-methylacyl-CoA-racemase; TFE-3 ID Renal cell carcinoma; CK7; Carbonic anhydrase IX; α-methylacyl-CoA-racemase; TFE-3 AB Renal cell carcinoma (RCC) in which clear cells with papillary architecture are present is a difficult diagnostic challenge. The most common type, clear cell RCC, only rarely has papillary architecture. The second most common one, papillary RCC, only rarely contains clear cells. However, two recently described less-common types, clear cell papillary and Xp11 translocation RCC characteristically feature both papillary architecture and cells with clear cytoplasm. Accurate diagnosis has both prognostic and therapeutic implications. This study aims to highlight the helpful cytomorphologic and immunohistochemical features of each of these entities to enable reproducible classification. Sixty RCC cases with clear cells and papillary architecture were selected and classified according to The International Society of Urological Pathology (ISUP) Vancouver Classification of Renal Neoplasia and graded according to The International Society of Urological Pathology (ISUP) grading system for renal cell carcinoma then stained for CK7, carbonic anhydrase IX (CA IX), α- methylacyl-CoA-racemase (AMACR) and TFE-3. The characteristic immunoprofile of Clear RCC is CK7-, AMACR-, CA IX+ and TFE3-, papillary RCC is CK7+, AMACR+, CAIX- and TFE3-, while for clear cell papillary RCC it is CK7+, AMACR-, CAIX+ and TFE3- and lastly Xp11translocation RCC is CK7-, AMACR+, CAIX- and TFE3+. Immunohistochemical staining for CA IX, CK7, AMACR and TFE3 comprises a concise panel for distinguishing RCC with papillary and clear pattern. C1 [Alshenawy, AlSaeid Hanan] Tanta University, Faculty of Medicine, Department of Pathology, 25 Hamdy Gado streetTanta, Egypt. RP Alshenawy, AH (reprint author), Tanta University, Faculty of Medicine, Department of Pathology, Tanta, Egypt. EM hanan_alshenawy@yahoo.com CR Deng F, Melamed J, 2012, Histologic variants of renal cell carcinoma: does tumor type influence outcome? Urol Clin N Am 39:119–132 Zhanyong B, Priti L, Song L et al, 2013, Role of carbonic anhydrase IX, α-methylacyl coenzyme a racemase, cytokeratin 7, and galectin-3 in the evaluation of renal neoplasms: a tissue microarray immunohistochemical study. Ann Diagn Pathol 17:58–62 Sean R, John N, Liang C et al, 2013, Clear cell papillary renal cell carcinoma: differential diagnosis and extended immunohistochemical profile. Mod Pathol 26:697–708 Hillary R, Guido M, Pedram A, 2012, Renal cell carcinoma with clear cell and papillary features. Arch Pathol Lab Med 136:391–399 5 . Wal t e r B, Hartmann A, Hofstadter F et al, 2012, Immunohistochemical marker panel differentiates between the three most common subtypes of renal cell carcinoma independent from histomorphologic criteria. Virchows Arch 460:343–352 6. Borislav A, Cinthia B, 2014, Clear cell papillary renal cell carcinoma: incidence, morphological features, immunohistochemical profile, and biologic behavior: a single institution study. Pathol Res Pract 210:234–241 7. Rajen G, Elizabeth G, Ximing J et al, 2013, Differential diagnosis of renal tumors with clear cytoplasm: clinical relevance of renal tumor subclassification in the era of targeted therapies and personalized medicine. Arch Pathol Lab Med 137:467–480 8. Park J, Lee C, Suh J et al, 2012, Clear cell papillary renal cell carcinoma: a report of 15 cases including three cases of concurrent othertype renal cell carcinomas. Korean J Pathol 46:541–547 9. Saba K, Ozden T, Sumer B et al, 2008, Diagnostic utility of cytokeratins 7, 10 and 20 in renal cell carcinoma and oncocytoma. Turk J Pathol 24:140–146 10. Michelle P, Amy Z, Zhanyong B, 2013, Useful immunohistochemical panel for differentiating clear cell papillary renal cell carcinoma from its mimics. Ann Diagn Pathol 17:437–440 11. Elizabeth M, Musie G, Robert N et al, 2010, Carbonic anhydrase IX expression in renal neoplasms: correlation with tumor type and grade. Am J Clin Pathol 134:873–879 12. John RS, Brett D, John NE et al, 2013, The International Society of Urological Pathology, ISUP, Vancouver classification of renal neoplasia. Am J Surg Pathol 37:1209–1220 13. Brett D, John CC, Guido M et al, 2013, The International Society of Urological Pathology, ISUP, grading system for renal cell carcinoma and other prognostic parameters. Am J Surg Pathol 37(10):1490– 1504 14. Edge S, Byrd D, Compton C et al, eds,, 2010, AJCC cancer staging manual, 7th edn. Springer, New York, pp 479–489 15. Stephen M, Yonghong X, Yupu L et al, 2011, Clear-cell papillary renal cell carcinoma: molecular and immunohistochemical analysis with emphasis on the von Hippel–Lindau gene and hypoxia-inducible factor pathway-related proteins. Mod Pathol 24:1207–1220 16. Brannon A, Haake S, Hacker K et al, 2012, Meta-analysis of clear cell renal cell carcinoma gene expression defines a variant subgroup and identifies gender influences on tumor biology. Eur Urol 61:258– 268 17. Aydin H, Chen L, Cheng L et al, 2010, Clear cell tubulopapillary renal cell carcinoma: a study of 36 distinctive low-grade epithelial tumors of the kidney. Am J Surg Pathol 34:608–1621 18. Duan L, Youseff R, Margulis Vet al, 2011, Clear cell papillary renal cell carcinoma: clinicopathologic, immunohistochemical, andmolecular analysis. Mod Pathol 24:189–193 19. Brunelli M, Menestrina F, Segala D et al, 2009, Renal cell carcinoma with prominent leiomyomatous proliferation appears not to be a variant of clear cell renal cell carcinoma. Mod Pathol 22:160– 165 20. Shen S, Truong L, Scarpelli M et al, 2012, Role of immunohistochemistry in diagnosing renal neoplasms: when is it really useful? Arch Pathol Lab Med 136:410–417 21. Al-Ahmadie H, Alden D et al, 2011, Role of immunohistochemistry in the evaluation of needle core biopsies in adult renal cortical tumors: an ex vivo study. Am J Surg Pathol 36:949–961 22. Tickoo S, Reuter V, 2011, Differential diagnosis of renal tumors with papillary architecture. Adv Anat Pathol 18:120–131 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 893 EP 899 DI 10.1007/s12253-015-9898-7 PG 7 ER PT J AU Al-Bahrani, R Nagamori, S Leng, R Petryk, A Sergi, C AF Al-Bahrani, Redha Nagamori, Seishi Leng, Roger Petryk, Anna Sergi, Consolato TI Differential Expression of Sonic Hedgehog Protein in Human Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Cholangiocarcinoma; Sonic hedgehog; Bone morphogenetic protein 4; Immunohistochemistry ID Hepatocellular carcinoma; Cholangiocarcinoma; Sonic hedgehog; Bone morphogenetic protein 4; Immunohistochemistry AB Hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (CCA) are the two most common primary liver malignancies in adult patients. The molecular mechanisms underlying the pathogenesis of HCC and CCA are still poorly understood. Sonic hedgehog (SHH) signaling plays an essential role during mammalian development, i.e., promoting organ growth, tissue differentiation, and cell polarity. The upregulation of SHH has been observed during carcinogenesis, including colorectal carcinoma. Our aimwas to investigate the expression pattern of SHH in HCC and CCA.We investigated 40 malignant tumors of the liver, including 21 HCC and 19 of intrahepatic CCA cases by immunohistochemistry (IHC) using a polyclonal antibody against SHH and Avidin-Biotin Complex method. We also investigated the co-localization of SHH and Bone morphogenetic protein 4 (BMP4) in CCA using indirect double IHC. Moreover, we examined whether SHH is expressed in two HCC cell lines HepG2 and HuH-7 and three CCA cell lines OZ, HuCCT1 and HuH28.We found that SHH was expressed in 15 out of 21 cases (71.4 %) of HCC and 100 % of CCA cases by immunohistochemistry. SHH expression showed a positive trend in liver tumors (HCC, CCA) with high grade (G2-G3). SHH localized to the epithelial cells, while BMP4 was expressed in the stromal cells in CCA by double IHC. However, both HCC and CCA cell lines showed SHH expression by Western blot analysis. In conclusion, SHH seems to be an interesting marker of dedifferentiation in liver tumors and the simultaneous epithelial-mesenchymal expression may be an intriguing prompt to investigate cross-talks between SHH and BMP4. C1 [Al-Bahrani, Redha] University of Alberta, Department of Laboratory Medicine and Pathology, 8440-112 Street, T6G 2B7 Edmonton, AB, Canada. [Nagamori, Seishi] National Institute of Infectious Diseases, Department of Virology IITokyo, Japan. [Leng, Roger] University of Alberta, Department of Laboratory Medicine and Pathology, 8440-112 Street, T6G 2B7 Edmonton, AB, Canada. [Petryk, Anna] University of Minnesota, Department of PediatricsMinneapolis, MN, USA. [Sergi, Consolato] University of Alberta, Department of Laboratory Medicine and Pathology, 8440-112 Street, T6G 2B7 Edmonton, AB, Canada. RP Sergi, C (reprint author), University of Alberta, Department of Laboratory Medicine and Pathology, T6G 2B7 Edmonton, Canada. EM sergi@ualberta.ca CR BlonskiW, Kotlyar DS, Forde KA, 2010, Non-viral causes of hepatocellular carcinoma. World J Gastroenterol 16:3603–3615 Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Motola-Kuba D, Zamora-Valdes D, Uribe M, Mendez-Sanchez N, 2006, Hepatocellular carcinoma. An overview. Ann Hepatol 5:16– 24 Al-Bahrani R, Abuetabh Y, Zeitouni N, Sergi C, 2013, Cholangiocarcinoma: risk factors, environmental influences and oncogenesis. Ann Clin Lab Sci. Spring;43(2):195–210 de Martel C, Plummer M, Franceschi S, 2010, Cholangiocarcinoma: descriptive epidemiology and risk factors. Gastroenterol Clin Biol 34:173–180 Nusslein-Volhard C, Wieschaus E, 1980, Mutations affecting segment number and polarity in Drosophila. Nature 287:795–801 Wong SY, Reiter JF, 2008, The primary cilium at the crossroads of mammalian hedgehog signaling. Curr Top Dev Biol 85:225–260 Litingtung Y, Lei L,Westphal H, Chiang C, 1998, Sonic hedgehog is essential to foregut development. Nat Genet 20:58–61 Dessaud E, McMahon AP, Briscoe J, 2008, Pattern formation in the vertebrate neural tube: a sonic hedgehog morphogen-regulated transcriptional network. Development 135:2489–2503 Chiang C, Litingtung Y, Lee E, Young KE, Corden JL, Westphal H, Beachy PA, 1996, Cyclopia and defective axial patterning in mice lacking sonic hedgehog gene function. Nature 383:407–413 Huang S, Yang L, An Y, Ma X, Zhang C, Xie G, Chen ZY, Xie J, Zhang H, 2011, Expression of hedgehog signaling molecules in lung cancer. Acta Histochem 113:564–569 Shahi MH, Afzal M, Sinha S, Eberhart CG, Rey JA, Fan X, Castresana JS, 2010, Regulation of sonic hedgehog-GLI1 downstream target genes PTCH1, Cyclin D2, Plakoglobin, PAX6 and NKX2.2 and their epigenetic status in medulloblastoma and astrocytoma. BMC Cancer 10:614-2407-10-614 Kawai S, Sugiura T, 2001, Characterization of human bone morphogenetic protein, BMP)-4 and -7 gene promoters: activation of BMP promoters by Gli, a sonic hedgehog mediator. Bone 29:54–61 Gekas J, Sergi C, KamnasaranD, 2012)Molecular prenatal diagnosis of a sporadic alobar holoprosencephalic fetus: genotype-phenotype correlations. J Prenat Med 6:36–39 Roessler E, Belloni E, Gaudenz K, Jay P, Berta P, Scherer SW, Tsui LC,MuenkeM(1996)Mutations in the human sonic hedgehog gene cause holoprosencephaly. Nat Genet 14:357–360 Yoshikawa K, Shimada M, Miyamoto H, Higashijima J, Miyatani T, Nishioka M, Kurita N, Iwata T, Uehara H, 2009, Sonic hedgehog relates to colorectal carcinogenesis. J Gastroenterol 44:1113–1117 Hahn H, Wicking C, Zaphiropoulous PG, Gailani MR, Shanley S, Chidambaram A, Vorechovsky I, Holmberg E, Unden AB, Gillies S, Negus K, Smyth I, Pressman C, Leffell DJ, Gerrard B, Goldstein AM, Dean M, Toftgard R, Chenevix-Trench G, Wainwright B, Bale AE, 1996, Mutations of the human homolog of Drosophila patched in the nevoid basal cell carcinoma syndrome. Cell 85:841–851 Reifenberger J, Wolter M, Weber RG, Megahed M, Ruzicka T, Lichter P, Reifenberger G, 1998, Missense mutations in SMOH in sporadic basal cell carcinomas of the skin and primitive neuroectodermal tumors of the central nervous system. Cancer Res 58:1798–1803 Regateiro FS, Howie D, Cobbold SP,Waldmann H, 2011, TGF-beta in transplantation tolerance. Curr Opin Immunol 23:660–669 Fan Q, He M, Sheng T, Zhang X, Sinha M, Luxon B, Zhao X, Xie J, 2010, Requirement of TGFbeta signaling for SMO-mediated carcinogenesis. J Biol Chem 285:36570–36576 Liem KF Jr, Tremml G, Roelink H, Jessell TM, 1995, Dorsal differentiation of neural plate cells induced byBMP-mediated signals from epidermal ectoderm. Cell 82:969–979 Jensen ED, Pham L, Billington CJ Jr, Espe K, Carlson AE, Westendorf JJ, Petryk A, Gopalakrishnan R, Mansky K, 2010, Bone morphogenic protein 2 directly enhances differentiation of murine osteoclast precursors. J Cell Biochem 109:672–682 Alarmo EL, Huhtala H, Korhonen T, Pylkkanen L, Holli K, Kuukasjarvi T, Parkkila S, Kallioniemi A, 2013, Bone morphogenetic protein 4 expression in multiple normal and tumor tissues reveals its importance beyond development. Mod Pathol 26:10–21 WangDH, Clemons NJ, Miyashita T, Dupuy AJ, ZhangW, Szczepny A, Corcoran-Schwartz IM,Wilburn DL,Montgomery EA,Wang JS, Jenkins NA, Copeland NA, Harmon JW, Phillips WA, Watkins DN, 2010, Aberrant epithelial-mesenchymal hedgehog signaling characterizes Barrett’s metaplasia. Gastroenterology 138:1810–1822 AbuetabhY, Persad S, Nagamori S, Huggins J, Al-Bahrani R, Sergi C, 2011, Expression of E-cadherin and beta-catenin in two cholangiocarcinoma cell lines, OZ and HuCCT1, with different degree of invasiveness of the primary tumor. Ann Clin Lab Sci 41:217– 223 Sergi C, Kahl P, Otto HF, 2000, Contribution of apoptosis and apoptosis-related proteins to the malformation of the primitive intrahepatic biliary system in Meckel syndrome. Am J Pathol 156: 1589–1598 Morton JP, Mongeau ME, Klimstra DS, Morris JP, Lee YC, Kawaguchi Y, Wright CV, Hebrok M, Lewis BC, 2007, Sonic hedgehog acts at multiple stages during pancreatic tumorigenesis. Proc Natl Acad Sci U S A 104:5103–5108 Ma XL, Sun HJ, Wang YS, Huang SH, Xie JW, Zhang HW, 2006, Study of sonic hedgehog signaling pathway related molecules in gastric carcinoma. World J Gastroenterol 12:3965–3969 Omenetti A, Diehl AM, 2008, The adventures of sonic hedgehog in development and repair. II. Sonic hedgehog and liver development, inflammation, and cancer. Am J Physiol Gastrointest Liver Physiol 294:G595–G598 Pereira Tde A,Witek RP, SynWK, Choi SS, Bradrick S, Karaca GF, Agboola KM, Jung Y, Omenetti A, Moylan CA, Yang L, Fernandez- ZapicoME, Jhaveri R, Shah VH, Pereira FE, Diehl AM, 2010, Viral factors induce hedgehog pathway activation in humans with viral hepatitis, cirrhosis, and hepatocellular carcinoma. Lab Invest 90: 1690–1703 Marcelle C, Stark MR, Bronner-Fraser M, 1997, Coordinate actions of BMPs, Wnts, Shh and noggin mediate patterning of the dorsal somite. Development 124:3955–3963 van den Brink GR, Bleuming SA, Hardwick JC, Schepman BL, Offerhaus GJ, Keller JJ, Nielsen C, Gaffield W, van Deventer SJ, Roberts DJ, PeppelenboschMP, 2004, Indian Hedgehog is an antagonist of Wnt signaling in colonic epithelial cell differentiation. Nat Genet 36:277–282 Johnston J, Al-Bahrani R, Abuetabh Y, Chiu B, Forsman CL, Nagamori S, Leng R, Petryk A, Sergi C, 2012, Twisted gastrulation expression in cholangiocellular and hepatocellular carcinoma. J Clin Pathol 65:945–948 Eiro N, Vizoso FJ, 2014, Importance of tumor/stroma interactions in prognosis of hepatocellular carcinoma. Hepatobiliary Surg Nutr 3(2): 98–101 Virchow R, 1863, Die krankhaften Geschwulste. August Hirschwald, Berlin NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 901 EP 908 DI 10.1007/s12253-015-9918-7 PG 8 ER PT J AU Ramya, NL Pulicherla, KK AF Ramya, N L Pulicherla, Kanth Krishna TI Studies on Deimmunization of Antileukaemic L-Asparaginase to have Reduced Clinical Immunogenicity- An in silico Approach SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE L- Asparaginase; Hypersensitive reactions; Neutralizing antibodies; Anaphylactic shock; Pectobacterium carotovorum; Immunogenicity; Acute lymphoblastic leukaemia ID L- Asparaginase; Hypersensitive reactions; Neutralizing antibodies; Anaphylactic shock; Pectobacterium carotovorum; Immunogenicity; Acute lymphoblastic leukaemia AB Protein therapeutics, particularly of heterologous origin are shown to elicit immunogenic responses which result in adverse allergic reactions in spite of their promising clinical benefit. L-Asparaginase is one such well known chemotherapeutic agent that has enhanced the survival rates to 90%in the treatment of acute lymphoblastic leukaemia for past 30 years. But the use of this enzyme is accompanied by hypersensitive reactions ranging from allergy to anaphylactic shock which have a drastic influence in treatment outcomes. Numerous attempts have been made to minimize the problems of immunogenicity, which remained as a major bottleneck in the treatment protocols. Conjugating the enzyme L- Asparaginase with PEG was successful as it has reduced the complications in therapy and frequency of injections (dosages), and thus became prominent in reducing the immunogenicity up to a certain extent. Keeping the bottlenecks in consideration during the development of therapeutics, the present study concentrates on engineering of protein as an alternative to the PEGylated enzyme, having reduced immunogenicity as an inbuilt character of protein by using in silico approaches. L-Asparaginase from Escherichia coli and Pectobacterium carotovorum were selected for the present study. The methodology consists of (i) locating the B and CD4+ T cell epitopes of enzyme by in silico tools (ii) generating point mutations of these epitopes to alter or reduce the immunogenicity of protein (iii) generating enzyme models by molecular modelling (iv) assessing the binding affinity of the substrate with L-Asparaginase variants by in silico docking methods using Autodock 4.2 and (v) validating the mutated model for stability by molecular dynamics simulation studies using Gromacs. C1 [Ramya, N L] Acharya Nagarjuna University, Department of Biotechnology, Nagarjuna Nagar, 522510 Guntur, AP, India. [Pulicherla, Kanth Krishna] VIT University, Center for Bioseparation Technology, 632014 Vellore, TN, India. RP Pulicherla, KK (reprint author), VIT University, Center for Bioseparation Technology, 632014 Vellore, India. EM kkpulicherla@gmail.com CR De Duve C, 1966, The significance of lysosome in pathology and medicine. Proc Inst Med Chic 26:73–76 Offman MN, Krol M, Patel N, Krishnan S, Liu JZ, Saha V, Bates PA, 2011, Rational engineering of L- asparaginase reveals importance of dual activity for cancer cell toxicity. Blood 117:1614–1621 Wai Kin Chan, Philip L. Lorenzi, Andriy Anishkin, Preeti Purwaha, David M. Rogers, Sergei, Sukharev, Susan B. Rempe, and John N. Weinstein., 2014). The glutaminase activity of L-asparaginase is not required for anticancer activity against ASNS-negative cells. Blood., DOI 10.1182/blood-2013-10-535112. StamsWAG, den BoerML, Holleman A, Appel IM, Beverloo B, van Wering ER, Janka-Schaub GE, Evans WE, Pieters R, 2005, Asparagine synthetase expression is linked with l-asparaginase resistance in TEL-AML1-negative but not TEL-AML1-positive pediatric acute lymphoblastic leukemia. Blood 105:4223–4225 Narta UK, Kanwar SS, AzmiW(2007, Pharmacological and clinical evaluation of L-asparaginase in the treatment if leukaemia. Crit Rev Oncol/Hematol 61:208–221 Pieters R, Hunger S, Boos J et al, 2011, L-asparaginase treatment in acute lymphoblastic leukemia. Cancer 117:238–249 Avramis VI, Sencer S, Periclou AP et al, 2002, A randomized comparison of native escherichia coli asparaginase and polyethylene glycol conjugated asparaginase for treatment of children with newly diagnosed standard-risk acute lymphoblastic leukemia: a children’s cancer group study. Blood 99:1986–1994 Zalewska-Szewczyk B, Gach A, Wyka K et al, 2009, The crossreactivity of antiasparaginase antibodies against different L-asparaginase preparations. Clin Exp Med 9:113–116 Raetz EA, Salzer WL, 2010, Tolerability and efficacy of L-asparaginase therapy in pediatric patients with acute lymphoblastic leukemia. J Pediatr Oncol 32:554–563 Rizzari C, Citterio M, Zucchetti M et al, 2006, A pharmacological study on pegylated asparaginase used in front-line treatment of children with acute lymphoblastic leukemia. Haematologica 91:24–31 Abuchowski A, Van Es T, Palczuk NC, 1979, Treatment of L5178Y tumor bearing BDf mice with a non-immunogenic lglutaminaseasparaginase. Cancer Treat Rep 63:1127–1129 Hawkins DS, Park JR, Thomson BG et al, 2004, Asparaginase pharmacokinetics after intensive polyethylene glycol-conjugated L-asparaginase therapy for children with relapsed acute lymphoblastic leukemia. Clin Cancer Res 10:5335–5341 Albertsen BK, Schroder H, Jakobsen P et al, 2002, Antibody formation during intravenous and intramuscular therapy with Erwinia asparaginase. Med Pediatr Oncol 38:310–316 Klug Albertsen B, Schmiegelow K, Schroder H et al, 2002, Anti- Erwinia asparaginase antibodies during treatment of childhood acute lymphoblastic leukemia and their relationship to outcome: a case– control study. Cancer Chemother Pharmacol 50:117–120 Vrooman LM, Supko JG, Neuberg DS et al, 2010, Erwinia asparaginase after allergy to E. coli asparaginase in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 54:199–205 Kumar GT, Prakash NJ, Laxmi N, Krishna TD, 2011, Biotechnology based durg delivery by PEGylation method. IJRAP 2(1):95–102 Kumar S, Pakshirajan K, Venkata Dasu V, 2009, Development of medium for enhanced production of glutaminase-free L-asparaginase from Pectobacterium carotovorum MTCC 1428. Appl Microbiol Biotechnol 84:477–486 Kumar S, Pakshirajan K, Venkata Dasu V, 2010, Assessment of physical process conditions for enhanced production of novel glutaminase-free L-Asparaginase from Pectobacterium carotovorum MTCC 1428. Appl Biochem Biotechnol 163:327–337 Tamura K, Peterson D, Peterson N, Stecher G, Nei M, Kumar S, 2001, MEGA5: molecular evolutionary genetics analysis using maximum likelihood, evolutionary distance, and maximum parsimony methods. Mol Biol Evol 28:2731–2739 Bailey T L, Elkan C, 1994, "Fitting a mixture model by expectation maximization to discover motifs in biopolymers," in Proceedings of the 2nd International Conference on Intelligent Systems for Molecular Biology. AAAI Press, pp 28–36 Punta M, Coggill PC, Eberhardt RY, Mistry J, Tate J, Boursnell C, Pang N, Forslund K, Ceric G, Clements J, Heger A, Holm L, Sonnhammer ELL, Eddy SR, Bateman A, Finn RD, 2012, Nucleic acids research. Database Issue 40:D290–D301 Zhang Q, Wang P, Kim Y, 2008, Immune epitope database analysis resource, IEDB-AR). Nucleic Acids Res 36(Web Server issue): W513–W518 Emini EA, Hughes JV, Perlow DS, Boger J, 1985, Induction of hepatitis A virus-neutralizing antibody by a virus-specific synthetic peptide. J Virol 55:836–839, PMID: 2991600 Parker JM, Guo D, Hodges RS, 1986, New hydrophilicity scale derived from high-performance liquid chromatography peptide retention data: correlation of predicted surface residues with immunogenicity and X-ray-derived accessible sites. Biochemistry 25:5425– 5432, PMID: 2430611 Nielsen M, Lundegaard C, LundO, 2007, Prediction ofMHC class II binding affinity using SMM-align, a novel stabilization matrix alignment method. BMC Bioinf 8:238 Cantor JR,Yoo TH, Dixit A, Iverson BL, Forsthuber TG, GeorgiouG, 2011, Therapeutic enzyme deimmunization by combinatorial T-cell epitope removal using neutral drift. PNAS 108(4):1272–1277 Hete’nyi C, Spoel VD, 2002, Protein Sci 11:1729 [PMID:12070326] Sousa SF, Fernandes PA, Ramos MJ et al, 2006, Proteins 65:15, PMID: 16862531 Huey R, Morris GM, Olson AJ, Goodsell DS, 2007, A semi empirical free energy force field with charge-based desolvation. J Comput Chem 28:145–152 Wang B, Relling MV, Storm MC et al, 2003, Evaluation of immunologic crossreaction of antiasparaginase antibodies in acute lymphoblastic leukemia, ALL, and lymphoma patients. Leukemia 17:1583– 1588 Berendsen HJC, van der Spoel D, van Drunen R, 1995, GROMACS: a message-passing parallel molecular dynamics implementation. Comput Phys Commun 91:43–56 Erik L, Berk H, van der Spoel D, 2001, GROMACS 3.0: a package for molecular simulation and trajectory analysis. J Mol Model 7:306– 317 Van der Spoel D, Lindahl E, Hess B, van Buuren AR, Apol E, Meulenhoff PJ, Tieleman DP, Sijbers ALTM, Feenstra KA, van Drunen R, Berendsen HJC, 2005, Gromacs User Manual version 4.0. www.gromacs.org Schuttelkopf AW, van Aalten DMF, 2004, PRODRG - a tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr D60:1355–1363 Kaushik Ramakrishanan S, Krishna V, Kumar SV, Lakshmi BS, Anishetty S, Gautham P, 2008, Molecular dynamics simulation of lipases. Int J Integr Biol 2:204–213 Clementi A, 1922, Desamidation enzymatique de l’ L-Asparagine. Archives Internationales de Physiologie 19:369 Ramya LN, Mukesh D, Rekha VPB, Pulicherla KK, 2012, L-Asparaginase as potent anti-leukemic agent and its significance of having reduced glutaminase side activity for the better treatment of acute lymphoblastic leukaemia. Appl Biochem Biotechnol 167(8):2144–2159 Kidd JG, 1953, Regression of transplanted lymphomas induced in vivo by means of normal guinea pig serum—course of transplanted cancers of various kinds in mice and rats given guinea pig serum or rabbit serum. J Exp Med 98:565–582 Wade HE, Elsworth R, Herbert D, 1968, A new L-asparaginase with anti-tumor activity. Lancet 2:776–777 Manna S, Sinha A, Sadhukhan R, Chakrabarty SL, 1995, Purification, characterization and antitumor activity of L-asparaginase isolated from Pseudomonas stutzeri MB-405. Curr Microbiol 30:291–298 Bander NH et al, 2005, Phase I trial of 177lutetium-labeled J591, a monoclonal antibody to prostate-specific membrane antigen, in patients with androgen-independent prostate cancer. J Clin Oncol 23: 4591–4601 Holgate RG, Baker MP, 2009, Circumventing immunogenicity in the development of therapeutic antibodies. IDrugs 12:233–237 Macfarlane DJ et al, 2006, Safety, pharmacokinetic and dosimetry evaluation of the proposed thrombus imaging agent 99mTc-DI-DD- 3B6/22-80B3 Fab 0. Eur J Nucl Med Mol Imaging 33:648–656 Wang P, Sidney J, Dow C, Mothe B, Sette A, Peters B, 2008, A systematic assessment of MHC class II peptide binding predictions and evaluation of a consensus approach. PLoS Comput Biol 4(4): e1000048 Wang P et al, 2008, A systematic assessment ofMHC class II peptide binding predictions and evaluation of a consensus approach. PLoS Comput Biol 4:e1000048 Moola ZB, Scawen MD, Atkinson T, Nicholls DJ, 1994, Erwinia chrysanthemi L-asparaginase: epitope mapping and production of antigenically modified enzymes. Biochem J 302:921–927 Jianhua C, YujunW, Ruibo J, MinW,WutongW(2006, Probing the antigenicity of E. coli L-asparaginase by mutational analysis. Mol Biotechnol 33(1):57–65 Ramya LN, Doble M, Rekha VPB, Pulicherla KK, 2011, In silico engineering of L-Asparaginase to have reduced glutaminase side activity for effective treatment of acute lymphoblastic leukaemia. J Pediatr Hematol Oncol 33(8):617–621 Derst C, Henseling J, Rohm KH, 1992, Probing the role of threonine and serine residues of E.coli asparaginase II by site-specific mutagenesis. Protein Eng 5:785–789, PubMed: 1287659 Patil R, Das S, Stanley A et al, 2010, Optimized hydrophobic interactions and hydrogen bonding at the target-ligand interface leads the pathways of drug-designing. PLoS ONE 5(8):1–10 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 909 EP 920 DI 10.1007/s12253-015-9912-0 PG 12 ER PT J AU Mostafavi-Pour, Z Kianpour, S Dehghani, M Mokarram, P Torabinejad, S Monabati, A AF Mostafavi-Pour, Zoherh Kianpour, Sedigheh Dehghani, Mehdi Mokarram, Pooneh Torabinejad, Simin Monabati, Ahmad TI Methylation of Integrin α4 and E-Cadherin Genes in Human Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Integrin α4; E-Cadherin; DU145; PC3; Prostate cancer; DNA methylation ID Integrin α4; E-Cadherin; DU145; PC3; Prostate cancer; DNA methylation AB Prostate cancer is the second most common malignancy in men worldwide. Abnormal epigenetic alterations such as DNA methylation and histone modification play an important role in tumor initiation, progression and regulation of cancer-related genes such as integrin α4 and E-cadherin. Expression of these genes was determined by semiquantitative reverse transcriptase-PCR in prostate cancer cell lines, DU145 and PC3, before and after treatment with 5-aza- 2-deoxycytidine and trichostatin A. Laser capture microdissection microscopy was used to obtain exclusively affected epithelial cells from prostate gland biopsies of 30 patients with prostate cancer and 40 with benign prostate hyperplasia. DNA bisulfite modifications followed by methylation-specific PCR were used to evaluate the promoter methylation status of Ecadherin and α4 integrin genes in extracted DNA from patients and aforementioned cell lines. The integrin α4 promoter in DU145 was fully methylated, whereas in PC3 cells, partial methylation was detected. E-cadherin was expressed in both cell lines; trichostatin A and 5-aza-2-deoxycytidine treatment had no effect on E-cadherin expression, however the combined treatment of both drugs or 5-aza-2-deoxycytidine alone increased integrin α4 expression. Integrin α4 and E-cadherin were hypermethylated in 66.6 % and 6.6 % of prostate cancer cases, respectively; no hypermethylation was observed in patients with benign prostate hyperplasia. These results together suggest that aberrant DNA methylation is one of the mechanisms involved in integrin α4 expression and may play an important role in human prostate carcinogenesis. In addition, the higher rate of integrin α4 gene methylation in prostate cancer patients elects it as a potential molecular tumor marker. C1 [Mostafavi-Pour, Zoherh] Shiraz University of Medical Sciences, Department of BiochemistryShiraz, Iran. [Kianpour, Sedigheh] Shiraz University of Medical Sciences, Department of BiochemistryShiraz, Iran. [Dehghani, Mehdi] University of Texas Health Science Center, Department of Internal Medicine, Division of Oncology, 77030 Houston, TX, USA. [Mokarram, Pooneh] Shiraz University of Medical Sciences, Department of BiochemistryShiraz, Iran. [Torabinejad, Simin] Shiraz University of Medical Sciences, Shiraz Nephrourology Research CenterShiraz, Iran. [Monabati, Ahmad] Medical School, Shiraz University of Medical Sciences, Department of PathologyShiraz, Iran. RP Mostafavi-Pour, Z (reprint author), Shiraz University of Medical Sciences, Department of Biochemistry, Shiraz, Iran. EM zmostafavipour88@yahoo.co.uk CR Stewart DA, Cooper CR, Sikes RA, 2004, Changes in extracellular matrix, ECM, and ECM-associated proteins in the metastatic progression of prostate cancer. Reprod Biol Endocrinol 2:2., DOI 10. 1186/1477-7827-2-2 Zhu Y, Wang J, He Q et al, 2011, Association of p53 codon 72 polymorphism with prostate cancer: a meta-analysis. Mol Biol Rep 38(3):1603–7 Tran NL, Nagle RB, Cress AE et al, 1999, N-cadherin expression in human prostate carcinoma cell lines. An epithelial-mesenchymal transformation mediating adhesion with stromal cells. Am J Pathol 155(3):787–798 Liu A, Wei L, Gardner WA et al, 2009, Correlated alterations in prostate basal cell layer and basement membrane. Int J Biol Sci 5(3):276–285 Tai KY, Shiah SG, Shieh YS et al, 2007, DNA methylation and histone modification regulate silencing of epithelial cell adhesion molecule for tumor invasion and progression. Oncogene 26(27): 3989–97 Corn PG, Smith BD, Ruckdeschel ES et al, 2000, E-cadherin expression is silenced by 5′ CpG island methylation in acute leukemia. Clin Cancer Res 6(11):4243–4248 Goel HL, Li J, Kogan S et al, 2008, Integrins in prostate cancer progression. Endocr Relat Cancer 15(3):657–64 Felding-Habermann B, 2003, Integrin adhesion receptors in tumor metastasis. Clin Exp Metastasis 20(3):203–13 Jia Y, Zeng ZZ, Markwart SM et al, 2004, Integrin fibronectin receptors in matrix metalloproteinase-1–dependent invasion by breast cancer and mammary epithelial cells. Cancer Res 64(23):8674–8681 Qian F, Vaux DL, Weissman IL, 1994, Expression of the integrin alpha 4 beta 1 on melanoma cells can inhibit the invasive stage of metastasis formation. Cell 77(3):335–47 Ren JZ, Huo JR, 2010, Effect of 5-Aza-CdR on expression and methylation of E-cadherin gene in human colon carcinoma cells. Chin J Cancer 29(1):38–42 Park J, Song SH, Kim TY et al, 2004, Aberrant methylation of integrin alpha4 gene in human gastric cancer cells. Oncogene 23(19):3474–80 Naghibalhossaini F, Zamani M, Mokaram P et al, 2011, Epigenetic and genetic analysis of WNT signaling pathway in sporadic colorectal cancer patients from Iran. Mol Biol Rep 39(5):6171–8 Nam JS, Ino Y, Kanai Yet al, 2004, 5-aza-2′-deoxycytidine restores the E-cadherin system in E-cadherin-silenced cancer cells and reduces cancer metastasis. Clin Exp Metastasis 21(1):49–56 Katto J, Mahlknecht U, 2011, Epigenetic regulation of cellular adhesion in cancer. Carcinogenesis 32(10):1414–8 Cheng JC, Auersperg N, Leung CK, 2011, Inhibition of p53 represses E-cadherin expres s ion by inc re a s ing DNA methyltransferase-1 and promoter methylation in serous borderline ovarian tumor cells. Oncogene 30(37):3930–42 Li LC, Zhao H, Nakajima K et al, 2001, Methylation of the Ecadherin gene promoter correlates with progression of prostate cancer. J Urol 166(2):705–709 Lee EJ, Lee BB, Han J et al, 2008, CpG island hypermethylation of E-cadherin, CDH1, and integrin alpha4 is associated with recurrence of early stage esophageal squamous cell carcinoma. Int J Cancer 123(9):2073–9 Nan X, Ng HH, Johnson CA et al, 1998, Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 393(6683):386–389 KirschnerKM,Wagner N,WagnerKD et al, 2006, TheWilms tumor suppressor Wt1 promotes cell adhesion through transcriptional activation of the alpha4integrin gene. J Biol Chem 281(42):31930–9 Li LC, Carroll PR, Dahiya R, 2005, Epigenetic changes in prostate cancer: implication for diagnosis and treatment. J Natl Cancer Inst 97(2):103–15 Al-Rayyan N, Litchfield LM, Ivanova MM et al, 2014, 5-Aza-2- deoxycytidine and trichostatin A increase COUP-TFII expression in antiestrogen-resistant breast cancer cell lines. Cancer Lett 347(1): 139–50 Dehghani M, Mostafavi-Pour Z et al, 2009, Evaluation of plasma interleukin-8 concentration in patients with prostate cancer and benign prostate hyperplasia. Iran J Immunol 6(2):92–98 Engl T, Makarevic J, Relja B et al, 2005, Mycophenolate mofetil modulates adhesion receptors of the beta1 integrin family on tumor cells: impact on tumor recurrence and malignancy. BMC Cancer 5:4 Mostafavi-Pour Z, Keihani S, Talaei-Khozani T et al, 2012, Expression of α2, α5 and α6 subunits of integrin in dedifferentiated NIH3T3 cells by cell-free extract of embryonic stem cells.Mol Biol Rep 39(7):7339–7346 Siegel R, Ma J, Zou Z et al, 2014, Cancer statistics. CA Cancer J Clin 64(1):9–29 Liu H-H, Tsai Y-S, Lai C-L et al, 2014, Evolving personalized therapy for castration-resistant prostate cancer. Biomedicine 4(1):7–15 Kong A-N T, Zhang C, Su Z-Y, 2013, Targeting epigenetics for cancer prevention by dietary cancer preventive compounds—the case of miRNA. Cancer Prev Res 6(7):622–4 Kang GH, Lee S, Kim J-S et al, 2003, Profile of aberrant CpG island methylation along the multistep pathway of gastric carcinogenesis. Lab Investig 83(5):635–41 Gosslar U, Jonas P, Luz A et al, 1996, Predominant role of alpha 4- integrins for distinct steps of lymphoma metastasis. Proc Natl Acad Sci U S A 93(10):4821–6 Phe V, Cussenot O, Roupret M, 2010, Methylated genes as potential biomarkers in prostate cancer. BJU Int 105(10):1364–70 Majumdar S, Buckles E, Estrada J et al, 2011, Aberrant DNA methylation and prostate cancer. Curr Genomics 12(7):486–505 Keil KP, Abler LL, Mehta V et al, 2014, DNA methylation of Ecadherin is a priming mechanism for prostate development. Dev Biol 387(2):142–53 SchmidMC,Avraamides CJ, Foubert P et al, 2011, Combined blockade of integrin-α4β1 plus cytokines SDF-1α or IL-1β potently inhibits tumor inflammation and growth. Cancer Res 71(22):6965–75 Portela A, Liz J, Nogales Vet al, 2013, DNA methylation determines nucleosome occupancy in the 50-CpG islands of tumor suppressor genes. Oncogene 32(47):5421–8 Kim H, Lapointe J, Kaygusuz G, 2005, The retinoic acid synthesis gene ALDH1a2 is a candidate tumor suppressor in prostate cancer. Cancer Res 65(18):8118–24 Hood JD, Cheresh DA, 2002, Role of integrins in cell invasion and migration. Nat Rev Cancer 2(2):91–100 Guo W, Giancotti FG, 2004, Integrin signalling during tumour progression. Nat Rev Mol Cell Biol 5(10):816–26 Graff JR, Herman JG, Lapidus RG et al, 1995, E-cadherin expression is silenced by DNA hypermethylation in human breast and prostate carcinomas. Cancer Res 55(22):5195–9 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 921 EP 927 DI 10.1007/s12253-015-9917-8 PG 7 ER PT J AU Alexandrakis, GM Goulidaki, N Pappa, AC Boula, A Psarakis, F Neonakis, I Tsirakis, G AF Alexandrakis, G Michael Goulidaki, Nektaria Pappa, A Constantina Boula, Anna Psarakis, Fotios Neonakis, Ioannis Tsirakis, George TI Interleukin-10 Induces Both Plasma Cell Proliferation and Angiogenesis in Multiple Myeloma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Angiogenesis; Cytokines; Growth factors; Multiple myeloma ID Angiogenesis; Cytokines; Growth factors; Multiple myeloma AB In multiple myeloma the angiogenic process is enhanced by various mediators.Among them interleukin-10 (IL- 10), secreted mainly by myeloma-associated macrophages seems to participate in myeloma progression with variable manners. The aim of the study was to measure serum levels of IL-10 in various stages of MM patients and to correlate them with various angiogenic cytokines, such as vascular endothelial growth factor and angiopoietin-2 and with known proliferation parameters, such as serum levels of B-cell activating factor and bone marrow infiltration by myeloma plasma cells, in order to explore their clinical significance. We measured serum levels of the above parameters by ELISA in 54 newly diagnosed MM patients. All of them were higher in MM patients and were increasing in parallel with disease progression. Furthermore, IL-10 correlated positively with both angiogenic cytokines and proliferation markers. This correlation of IL-10 with both angiogenic cytokines and markers of disease activity implicates that they all have an important role in MM pathogenesis and progression. C1 [Alexandrakis, G Michael] University Hospital of Heraklion, Hematology Department, Stavrakia, 71110 Heraklion, Crete, Greece. [Goulidaki, Nektaria] University Hospital of Heraklion, Hematology DepartmentHeraklion, Greece. [Pappa, A Constantina] University Hospital of Heraklion, Hematology DepartmentHeraklion, Greece. [Boula, Anna] University Hospital of Heraklion, Hematology DepartmentHeraklion, Greece. [Psarakis, Fotios] University Hospital of Heraklion, Hematology DepartmentHeraklion, Greece. [Neonakis, Ioannis] University Hospital of Heraklion, Hematology DepartmentHeraklion, Greece. [Tsirakis, George] University Hospital of Heraklion, Hematology Department, Stavrakia, 71110 Heraklion, Crete, Greece. RP Tsirakis, G (reprint author), University Hospital of Heraklion, Hematology Department, 71110 Heraklion, Greece. EM georgetsirakis@gmail.com CR Kyle RA, Rajkumar SV, 2004, Multiple myeloma. N Engl J Med 351:1860–1873 Coussens LM,Werb Z, 2002, Inflammation and cancer. Nature 420: 860–867 Benjamin D, Park CD, Sharma V, 1994, Human B cell interleukin 10. Leuk Lymphoma 12:205–210 Lauta VM, 2003, A review of the cytokine network in multiple myeloma: diagnostic, prognostic, and therapeutic implications. Cancer 97:2440–2452 Nacinovic-Duletic A, Stifter S, Dvornik S, Skunca Z, Jonjic N, 2008, Correlation of serum IL-6, IL-8 and IL-10 levels with clinicopathological features and prognosis in patients with diffuse large B-cell lymphoma. Int J Lab Hematol 30:230–239 Gupta M, Han JJ, Stenson M,Maurer M,Wellik L, Hu G, Ziesmer S, Dogan A, 2012, Witzig TE, 2012, Elevated serum IL-10 levels in diffuse large B-cell lymphoma: a mechanism of aberrant JAK2 activation. Blood 119:2844–2853 Fayad L, Keating MJ, Reuben JM, O’Brien S, Lee BN, Lerner S, Kurzrock R, 2001, Interleukin-6 and interleukin-10 levels in chronic lymphocytic leukemia: correlation with phenotypic characteristics and outcome. Blood 97:256–263 Kovacs E, 2010, Interleukin-6 leads to interleukin-10 production in several human multiple myeloma cell lines. Does interleukin-10 enhance the proliferation of these cells? Leuk Res 34:912–916 AlexandrakisMG, Roussou P, Pappa CA, Messaritakis I, Xekalou A, Goulidaki N, Boula A, Tsirakis G, 2013, Relationship between circulating BAFF serum levels with proliferating markers in patients with multiple myeloma. Biomed Res Int 2013:389579 Vacca A, Ria R, Ribatti D, Semeraro F, Djonov V, Di Raimondo F, Dammacco F, 2003, A paracrine loop in the vascular endothelial growth factor pathway triggers tumor angiogenesis and growth in multiple myeloma. Haematologica 88:176–185 Dankbar B, Padro T, Leo R, Feldmann B, Kropff M, Mesters RM, Serve H, Berdel WE, Kienast J, 2000, Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma. Blood 95:2630–2636 Davis S, Aldrich TH, Jones PF, Acheson A, Compton DL, Jain V, Ryan TE, Bruno J, Radziejewski C, Maisonpierre PC, Yancopoulos GD, 1996, Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expression cloning. Cell 87:1161–1169 Scharpfenecker M, Fiedler U, Reiss Y, Augustin HG, 2005, The Tie- 2 ligand angiopoietin-2 destabilizes quiescent endothelium through an internal autocrine loop mechanism. J Cell Sci 118:771–780 Holash J,Wiegand SJ, Yancopoulos GD, 1999, New model of tumor angiogenesis: dynamic balance between vessel regression and growth mediated by angiopoietins and VEGF. Oncogene 18:5356–5362 Papetti M, Herman IM, 2002, Mechanisms of normal and tumorderived angiogenesis. Am J Physiol Cell Physiol 282:C947–970 Giuliani N, Colla S, Lazzaretti M, Sala R, Roti G, Mancini C, Bonomini S, Lunghi P, Hojden M, Genestreti G, Svaldi M, Coser P, Fattori PP, Sammarelli G, Gazzola GC, Bataille R, Almici C, Caramatti C, Mangoni L, Rizzoli V, 2003, Proangiogenic properties of human myeloma cells: Production of angiopoietin-1 and its potential relationship to myeloma-induced angiogenesis. Blood 102:638– 645 Fragioudaki M, Boula A, Tsirakis G, Psarakis F, Spanoudakis M, Papadakis IS, Pappa CA, Alexandrakis MG, 2012, B cellactivating factor: Its clinical significance in multiple myeloma patients. Ann Hematol 91:1413–1418 Ria R, Reale A, De Luisi A, Ferrucci A, Moschetta M, Vacca A, 2011, Bone marrow angiogenesis and progression in multiple myeloma. Am J Blood Res 1:76–89 Hao NB, Lu MH, Fan YH, Cao YL, Zhang ZR, Yang SM, 2012, Macrophages in tumor microenvironments and the progression of tumors. Clin Dev Immunol 2012:948098 Sica A, Saccani A, Mantovani A, 2002, Tumor-associated macrophages: a molecular perspective. Int Immunopharmacol 2:1045– 1054 Mantovani A, Sica A, Allavena P, Garlanda C, Locati M, 2009, Tumor-associated macrophages and the related myeloid-derived suppressor cells as a paradigm of the diversity of macrophage activation. Hum Immunol 70:325–330 Sindrilaru A, Peters T, Wieschalka S, Baican C, Baican A, Peter H, Hainzl A, Schatz S, Qi Y, Schlecht A, Weiss JM, Wlaschek M, Sunderkotter C, Scharffetter-Kochanek K, 2011, An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J Clin Invest 121:985–997 Dace DS, Khan AA, Kelly J, Apte RS, 2008, Interleukin-10 promotes pathological angiogenesis by regulating macrophage response to hypoxia during development. PLoS One 3:e3381 Lewis C, Murdoch C, 2005, Macrophage responses to hypoxia: Implications for tumor progression and anti-cancer therapies. Am J Pathol 167:627–635 Solinas G, Germano G, Mantovani A, Allavena P, 2009, Tumorassociated macrophages, TAM, as major players of the cancerrelated inflammation. J Leukoc Biol 86:1065–1073 Siveen KS, Kuttan G, 2009, Role ofmacrophages in tumour progression. Immunol Let 123:97–102 Tanaka Y, Kobayashi H, Suzuki M, Kanayama N, Suzuki M, Terao T, 2002, Thymidine phosphorylase expression in tumor-infiltrating macrophages may be correlated with poor prognosis in uterine endometrial cancer. Hum Pathol 33:1105–1113 Matsuda M, Salazar F, Petersson M, Masucci G, Hansson J, Pisa P, Zhang QJ, Masucci MG, Kiessling R, 1994, Interleukin 10 pretreatment protects target cells from tumor- and allospecific cytotoxic T cells and downregulates HLA class I expression. J Exp Med 180: 2371–2376 Miotto D, Lo Cascio N, Stendardo M, Querzoli P, Pedriali M, De Rosa E, Fabbri LM, Mapp CE, Boschetto P, 2010, CD8+ T cells expressing IL-10 are associated with a favourable prognosis in lung cancer. Lung Cancer 69:355–360 Lopez MV, Adris SK, Bravo AI, Chernajovsky Y, Podhajcer OL, 2005, IL-12 and IL-10 expression synergize to induce the immune-mediated eradication of established colon and mammary tumors and lung metastasis. J Immunol 175:5885–5894 Mocellin S,Marincola FM, Young HA, 2005, Interleukin-10 and the immune response against cancer: a counterpoint. J Leukoc Biol 78: 1043–1051 Berardi S, Ria R, Reale A, De Luisi A, Catacchio I, Moschetta M, Vacca A, 2013, Multiple myeloma macrophages: pivotal players in the tumor microenvironment. J Oncol 183602 Vacca A, Ribatti D, Presta M,Minischetti M, IurlaroM, Ria R,Albini A, Bussolino F, Dammacco F, 1999, Bone marrow neovascularization, plasma cell angiogenic potential, and matrix metalloproteinase- 2 secretion parallel progression of human multiple myeloma. Blood 93:3064–3073 Pappa CA, Tsirakis G, Samiotakis P, Tsigaridaki M, Alegakis A, Goulidaki N, Alexandrakis MG, 2013, Serum levels of angiopoietin-2 are associated with the growth of multiple myeloma. Cancer Invest 31:385–389 Uneda S, Matsuno F, Sonoki T, Tniguchi I, Kawano F, Hata H, 2003, Expressions of vascular endothelial growth factor and angiopoietin-2 in myeloma cells. Haematologica 88:113–115 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 929 EP 934 DI 10.1007/s12253-015-9921-z PG 6 ER PT J AU Michalska, MM Samulak, D Romanowicz, H Smolarz, B AF Michalska, M Magdalena Samulak, Dariusz Romanowicz, Hanna Smolarz, Beata TI Single Nucleotide Polymorphisms (SNPs) of RAD51-G172T and XRCC2-41657C/T Homologous Recombination Repair Genes and the Risk of Triple- Negative Breast Cancer in Polish Women SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE RAD51; XRCC2; Triple negative breast cancer; Gene polymorphism ID RAD51; XRCC2; Triple negative breast cancer; Gene polymorphism AB Double strand DNA breaks are the most dangerous DNA damage which, if non-repaired or misrepaired, may result in genomic instability, cancer transformation or cell death. RAD51 and XRCC2 encode proteins that are important for the repair of double-strand DNA breaks by homologous recombination. Therefore, genetic variability in these genes may contribute to the occurrence and progression of triple-negative breast cancer. The polymorphisms of the XRCC2 gene - 41657C/T (rs718282) and of the RAD51 gene, −172G/T (rs1801321), were investigated by PCR-RFLP in 70 patients with triple-negative breast cancer and 70 age- and sex matched non-cancer controls. The obtained results demonstrated a significant positive association between the RAD51 T/T genotype and TNBC, with an adjusted odds ratio (OR) of 4.94 (p=0.001). The homozygous T/T genotype was found in 60%of TNBC cases and in 14% of the used controls. Variant 172 Tallele of RAD51 increased cancer risk (OR=2.81 (1.72– 4.58), p<.0001). No significant associations were observed between -41657C/T genotype of XRCC2 and the incidence of TNBC. There were no significant differences between the distribution of XRCC2 -41657C/T genotypes in the subgroups assigned to histological grades. The obtained results indicate that the polymorphism of RAD51, but not of XRCC2 gene, may be positively associated with the incidence of triplenegative breast carcinoma in the population of Polish women. C1 [Michalska, M Magdalena] Regional Hospital in Kalisz, Department of Obstetrics and GynaecologyKalisz, Poland. [Samulak, Dariusz] Regional Hospital in Kalisz, Department of Obstetrics and GynaecologyKalisz, Poland. [Romanowicz, Hanna] Regional Hospital in Glowno, High School of InformaticsGlowno, Poland. [Smolarz, Beata] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. RP Smolarz, B (reprint author), Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, 93-338 Lodz, Poland. EM smolbea@wp.pl CR Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V, 2007, Descriptive analysis of estrogen receptor, ER)-negative, progesterone receptor, PR)-negative, and HER2-negative invasive breast cancer, the socalled triple-negative phenotype: a population-based study from the California Cancer Registry. Cancer 109:1721–1728 Chacon RD, Costanzo MV, 2010, Triple-negative breast cancer. Breast Cancer Res 12:S3 Cleator S, HellerW, Coombes RC, 2007, Triple-negative breast cancer: therapeutic options. Lancet Oncol 8:235–244 Dawson SJ, Provenzano E, Caldas C, 2009, Triple negative breast cancers: clinical and prognostic implications. Eur J Cancer 45:27–40 de Ruijter TC, Veeck J, de Hoon JP, van Engeland M, Tjan-Heijnen VC, 2011, Characteristics of triple-negative breast cancer. J Cancer Res Clin Oncol 137:183–192 Jackson SP, 2002, Sensing and repairing DNA double-strand breaks. Carcinogenesis 23:687–696 Helleday T, 2003, Pathways for mitotic homologous recombination in mammalian cells. Mutat Res 532:103–115 Thacker J, 2005, The RAD51 gene family, genetic instability and cancer. Cancer Lett 219:125–135 Hasselbach L, Haase S, Fischer D, Kolberg HC, Sturzbecher HW, 2005, Characterisation of the promoter region of the human DNArepair gene RAD51. Eur J Gynaecol Oncol 26:589–598 Antoniou AC, SinilnikovaOM, Simard J et al, 2007, RAD51 135G– >C modifies breast cancer risk among BRCA2 mutation carriers: results from a combined analysis of 19 studies. Am J Hum Genet 81:1186–1200 Krivokuca AM, Malisic EJ, Dobricic JD, Brotto KV, Cavic MR, Jankovic RN, Tomasevic ZI, Brankovic-Magic MV, 2014, RAD51 135G>C and TP53 Arg72Pro polymorphisms and susceptibility to breast cancer in Serbian women. Fam Cancer.13: 173–80 Wang W, Li JL, He XF, Li AP, Cai YL, Xu N, Sun SM, Wu BY, 2013, Association between the RAD51 135 G>C polymorphism and risk of cancer: a meta-analysis of 19,068 cases and 22,630 controls. PLoS ONE 8:e75153 Romanowicz-Makowska H, Smolarz B, Zadrozny M, Westfa B, Baszczynski J, Kokolaszwili G, Burzyfiski M, Polac I, Sporny S, 2012, The association between polymorphisms of the RAD51- G135C, XRCC2-Arg188His and XRCC3-Thr241Met genes and clinico-pathologic features in breast cancer in Poland. Eur J Gynaecol Oncol 33:145–150 Romanowicz-Makowska H, Smolarz B, Zadrozny M, Westfal B, Baszczynski J, Polac I, Sporny S, 2011, Single nucleotide polymorphisms in the homologous recombination repair genes and breast cancer risk in Polish women. Tohoku J Exp Med 224:201–208 Lee KM, Choi JY, Kang C, Kang CP, Park SK, Cho H, Cho DY, Yoo KY, Noh DY, Ahn SH, Park CG, Wei Q, Kang D, 2005, Genetic polymorphisms of selected DNA repair genes, estrogen and progesterone receptor status, and breast cancer risk. Clin Cancer Res 11: 4620–4626 Shin A, Lee KM, Ahn B, Park CG, Noh SK, Park DY, Ahn SH, Yoo KY, Kang D, 2008, Genotype-phenotype relationship between DNA repair gene genetic polymorphisms and DNA repair capacity. Asian Pac J Cancer Prev 9:501–505 Kuschel B, Auranen A, McBride S, 2002, Variants in DNA doublestrand break repair genes and breast cancer susceptibility. Hum Mol Genet 11:1399–40722 Thacker J, ZdzienickaMZ, 2004, The XRCC genes: expanding roles in DNA double-strand break repair. DNA Repair, Amst, 3:1081– 1090 Silva SN, Tomar M, Paulo C, Gomes BC, Azevedo AP, Teixeira V, Pina JE, Rueff J, Gaspar JF, 2010, Breast cancer risk and common single nucleotide polymorphisms in homologous recombination DNA repair pathway genes XRCC2, XRCC3, NBS1 and RAD51. Cancer Epidemiol 34:85–92 Lin WY, Camp NJ, Cannon-Albright LA et al, 2011, A role for XRCC2 gene polymorphisms in breast cancer risk and survival. J Med Genet 48:477–484 Sliwinski T, Krupa R, Majsterek I et al, 2005, Polymorphisms of the BRCA2 and RAD51 genes in breast cancer. Breast Cancer Res Treat 94:105–109 Wang N, Xiu-juan D, Rong-miao Z, Wei G, Xiao-juan Z, Yan L, 2009, An investigation on the polymorphisms of two DNA repair genes and susceptibility to ESCC and GCA of high-incidence region in northern China. Mol Biol Rep 36:357–364 Lu J, Wang LE, Xiong P, Sturgis EM, Spitz MR, Wei Q, 2007, 172G>T variant in the 5′ untranslated region of DNA repair gene RAD51 reduces risk of squamous cell carcinoma of the head and neck and interacts with a P53 codon 72 variant. Carcinogenesis 28: 988–994 Davidson JM, Gorringe KL, Chin SF, Orsetti B, Besret C, Courtay- Cahen C, Roberts I, Theillet C, Caldas C, Edwards PA, 2000, Molecular cytogenetic analysis of breast cancer cell lines. Br J Cancer 83:1309–1317 Loveday RL, Greenman J, Simcox DL, Speirs V, Drew PJ, Monson JR, Kerin MJ, 2000, Genetic changes in breast cancer detected by comparative genomic hybridisation. Int J Cancer 86:494–500 Zhao M, Chen P, Dong Y, Zhu X, Zhang X, 2014, Relationship between Rad51 G135C and G172T variants and the susceptibility to cancer: a meta-analysis involving 54 case–control studies. PLoS ONE 9:e87259 Smolarz B, Zadrozny M, Duda-Szymanska J, Makowska M, Samulak D, Michalska MM, Mojs E, Brys M, Forma E, Romanowicz-Makowska H, 2013, RAD51 genotype and triplenegative breast cancer, TNBC, risk in Polish women. Pol J Pathol 64:39–43 Hosseini M, Houshmand M, Ebrahimi A, 2013, RAD51 polymorphisms and breast cancer risk. Mol Biol Rep 40:665–668 Romanowicz-Makowska H, Smolarz B, Gajecka M, Kiwerska K, Rydzanicz M, Kaczmarczyk D, Olszewski J, Szyfter K, Blasiak J, Morawiec-Sztandera A, 2012, Polymorphism of the DNA repair genes RAD51 and XRCC2 in smoking- and drinking-related laryngeal cancer in a Polish population. Arch Med Sci 8:1065–1075 Improta G, Sgambato A, Bianchino G, Zupa A, Grieco V, La Torre G, Traficante A, Cittadini A, 2008, Polymorphisms of the DNA repair genes XRCC1 and XRCC3 and risk of lung and colorectal cancer: a case–control study in a Southern Italian population. Anticancer Res 28:2941–2946 Lu PH, Li C, Zhou HY, Wang T, Chu CQ, Shen W, Tao GQ, 2011, Same data sources, different pooled analysis result: the ongoing uncertainty in the subgroup analysis of RAD51 135G/C polymorphism and breast cancer risk. Breast Cancer Res Treat 125:299–300 Sliwinski T, Walczak A, Przybylowska K et al, 2010, Polymorphisms of theXRCC3 C722Tand the RAD51G135C genes and the risk of head and neck cancer in a polish population. Exp Mol Pathol 89:358–366 Li F, Li C, Jiang Z, Ma N, Gao X, 2011, XRCC3 T241M polymorphism and bladder cancer risk: a meta-analysis. Urology 77:511–515 Smolarz B, Samulak D, MichalskaM, Goralczyk B, Szyllo K, Lewy J, Sporny S, Kokolaszwili G, Burzynski M, Romanowicz-Makowska H, 2011, 135G>C and 172G>T polymorphism in the 5′ untranslated region of RAD51 and sporadic endometrial cancer risk in Polish women. Pol J Pathol 62:157–162 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 935 EP 940 DI 10.1007/s12253-015-9922-y PG 6 ER PT J AU Fang, WL Huang, KH Lan, YT Chen, MH Chao, Y Lo, SSh Wu, ChW Shyr, YM Li, FYA AF Fang, Wen-Liang Huang, Kuo-Hung Lan, Yuan-Tzu Chen, Ming-Huang Chao, Yee Lo, Su-Shun Wu, Chew-Wun Shyr, Yi-Ming Li, Fen-Yau Anna TI The Risk Factors of Lymph Node Metastasis in Early Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Early gastric cancer; Lymph node metastasis; Lymphatic invasion ID Early gastric cancer; Lymph node metastasis; Lymphatic invasion AB Endoscopic mucosal resection (EMR) or endoscopic submucosal dissection (ESD) is an effective alternative treatment for early gastric cancer. However, a major concern is the likelihood of lymph node metastasis. From December 1987 to December 2006, 391 patients who underwent curative surgery for gastric cancer with mucosal (T1a, n=265) or submucosal (T1b, n=126) invasion and a retrieved lymph node number≧15 were enrolled. The frequency and risk factors of lymph node metastasis were analyzed. The frequency of lymph node metastasis was 4.9 % in T1a lesions and 21.4 % in T1b lesions. Although the depth of submucosal tumor invasion was<2 mm, there was a 28.6 % chance of lymph node metastasis. A T1b lesion, i.e., the width of the submucosal tumor invasion was<5 mm, resulted in fewer lymph node metastases than lesions>5 mm in width. Multivariate analysis demonstrated that Lauren’s diffuse type and lymphatic invasion were independent risk factors for lymph node metastasis in T1a lesions, while lymphatic invasion was the strongest risk factor for lymph node metastasis in T1b lesions. EMR/ESD is a good alternative for T1a intestinal type adenocarcinoma without lymphatic invasion. Surgical resection is necessary for patients with T1b gastric cancer with lymphatic invasion. C1 [Fang, Wen-Liang] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd, Beitou District, 11217 Taipei, Taiwan, Republic of China. [Huang, Kuo-Hung] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd, Beitou District, 11217 Taipei, Taiwan, Republic of China. [Lan, Yuan-Tzu] Taipei Veterans General Hospital, Department of Surgery, Division of Colon & Rectal SurgeryTaipei, Taiwan, Republic of China. [Chen, Ming-Huang] Taipei Veterans General Hospital, Department of Medicine, Division of Hematology and OncologyTaipei, Taiwan, Republic of China. [Chao, Yee] Taipei Veterans General Hospital, Department of Medicine, Division of Hematology and OncologyTaipei, Taiwan, Republic of China. [Lo, Su-Shun] National Yang-Ming University, School of MedicineTaipei, Taiwan, Republic of China. [Wu, Chew-Wun] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd, Beitou District, 11217 Taipei, Taiwan, Republic of China. [Shyr, Yi-Ming] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd, Beitou District, 11217 Taipei, Taiwan, Republic of China. [Li, Fen-Yau Anna] Taipei Veterans General Hospital, Department of Pathology, No. 201, Sec. 2, Shipai Rd, Beitou District, 11217 Taipei, Taiwan, Republic of China. RP Fang, WL (reprint author), Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, 11217 Taipei, Taiwan, Republic of China. EM s821094@hotmail.com CR Japanese Gastric Cancer Association, 2011, Japanese classification of gastric carcinoma: 3rd English edition. Gastric Cancer 14:101–112 Gotoda T, 2007, Endoscopic resection of early gastric cancer. Gastric Cancer 10:1–11 Takenaka R, Kawahara Y, Okada H et al, 2008, Risk factors associated with local recurrence of early gastric cancers after endoscopic submucosal dissection. Gastrointest Endosc 68:887–894 Holscher AH, Drebber U, Monig SP et al, 2009, Early gastric cancer: lymph node metastasis starts with deep mucosal infiltration. Ann Surg 250:791–797 Sobin L, Gospodarowicz M, Wittekind C, eds, 2009, TNM Classification of Malignant Tumours In: 7th ed. International Union Against Cancer, UICC). New York: Wiley Sano T, Kobori O,Muto T, 1992, Lymph node metastasis from early gastric cancer: endoscopic resection of tumour. Br J Surg 79:241–244 An JY, Baik YH, Choi MG et al, 2007, Predictive factors for lymph node metastasis in early gastric cancer with submucosal invasion. Ann Surg 246:749 Ye BD, Kim SG, Lee JY et al, 2008, Predictive factors for lymph node metastasis and endoscopic treatment strategies for undifferentiated early gastric cancer. J Gastroenterol Hepatol 23:46–50 Folli S, Morgagni P, Roviello F et al, 2001, Risk factors for lymph node metastases and their prognostic significance in early gastric cancer, EGC, for the Italian Research Group for Gastric Cancer, IRGGC). Jpn J Clin Oncol 31:495–499 Japanese gastric cancer treatment guidelines 2010, ver.3,, 2001, Gastric Cancer. 14: 113–23. Soetikno R, Kaltenbach T, Yeh R et al, 2005, Endoscopic mucosal resection for early cancers of the upper gastrointestinal tract. J Clin Oncol 23:4490–4498 Hirasawa T, Gotoda T, Miyata S et al, 2009, Incidence of lymph node metastasis and the feasibility of endoscopic resection for undifferentiated-type early gastric cancer. Gastric Cancer 12:148–152 Bravo Neto GP, dos Santos EG, Victer FC et al, 2014, Lymph node metastasis in early gastric cancer. Rev Col Bras Cir 41:11–17 Ishigami S, Hokita S, Natsugoe S et al, 1998, Carcinomatous infiltration into the submucosa as a predictor of lymph node involvement in early gastric cancer. World J Surg 22:1056–1059 Yamada T, Sugiyama H, Ochi D et al, 2014, Risk factors for submucosal and lymphovascular invasion in gastric cancer looking indicative for endoscopic submucosal dissection. Gastric Cancer 17:692– 696 Kim H, Kim JH, Park JC et al, 2011, Lymphovascular invasion is an important predictor of lymph node metastasis in endoscopically resected early gastric cancers. Oncol Rep 25:1589–1595 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 941 EP 946 DI 10.1007/s12253-015-9920-0 PG 6 ER PT J AU Chen, H Ye, Q Lv, J Ye, P Sun, Y Fan, Sh Su, X Gavine, P Yin, X AF Chen, Hao Ye, Qingqing Lv, Jing Ye, Peng Sun, Yun Fan, Shuqiong Su, Xinying Gavine, Paul Yin, Xiaolu TI Evaluation of Trastuzumab Anti-Tumor Efficacy and its Correlation with HER-2 Status in Patient-Derived Gastric Adenocarcinoma Xenograft Models SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HER2; Gastric carcinoma; Trastuzumab; Xenograft model antitumor assays; Fluorescence in situ hybridization; Immunohistochemistry ID HER2; Gastric carcinoma; Trastuzumab; Xenograft model antitumor assays; Fluorescence in situ hybridization; Immunohistochemistry AB The aimof the study was to investigate trastuzumab anti-tumor efficacy and its correlation with HER-2 status in primary xenograft models derived from Chinese patients with gastric adenocarcinoma. Patient-derived gastric adenocarcinoma xenograft (PDGAX) mouse models were firstly generated by implanting gastric adenocarcinoma tissues from patients into immune deficient mice. A high degree of histological and molecular similarity between the PDGAX mouse models and their corresponding patients’ gastric adenocarcinoma tissues was shown by pathological observation, HER-2 expression, HER-2 gene copy number, and mutation detection. Based on Hoffmann’s criteria in gastric cancer, three models (PDGAX001, PDGAX003 and PDGAX005) were defined as HER-2 positive with fluorescence in situ hybridization (FISH) amplification or immunohistochemistry (IHC) 2+/ 3+, while two models (PDGAX002, PDGAX004) were defined as HER-2 negative. Upon trastuzumab treatment, significant tumor regression (105 % TGI) was observed in model PDGAX005 (TP53 wt), while moderate sensitivity (26 % TGI) was observed in PDGAX003, and resistance was observed in PDGAX001, 002 and 004. A significant increase in HER-2 gene copy number was only observed in PDGAX005 (TP53 wt). Interestingly, trastuzumab showed no efficacy in PDGAX001 (HER2 IHC 3+ and FISH amplification, but with mutant TP53). Consistent with this finding, phosphor-HER2 modulation by trastuzumab was observed in model PDGAX005, but not in PDGAX001. C1 [Chen, Hao] Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital, Department of General Surgery, No. 160 Pujian Road, 200127 Shanghai, China. [Ye, Qingqing] AstraZeneca R&D, Asia & Emerging Market iMed, 199 Liangjing Road, 201203 Shanghai, China. [Lv, Jing] AstraZeneca R&D, Asia & Emerging Market iMed, 199 Liangjing Road, 201203 Shanghai, China. [Ye, Peng] AstraZeneca R&D, Asia & Emerging Market iMed, 199 Liangjing Road, 201203 Shanghai, China. [Sun, Yun] AstraZeneca R&D, Asia & Emerging Market iMed, 199 Liangjing Road, 201203 Shanghai, China. [Fan, Shuqiong] AstraZeneca R&D, Asia & Emerging Market iMed, 199 Liangjing Road, 201203 Shanghai, China. [Su, Xinying] AstraZeneca R&D, Asia & Emerging Market iMed, 199 Liangjing Road, 201203 Shanghai, China. [Gavine, Paul] AstraZeneca R&D, Asia & Emerging Market iMed, 199 Liangjing Road, 201203 Shanghai, China. [Yin, Xiaolu] AstraZeneca R&D, Asia & Emerging Market iMed, 199 Liangjing Road, 201203 Shanghai, China. RP Yin, X (reprint author), AstraZeneca R&D, Asia & Emerging Market iMed, 201203 Shanghai, China. EM lucy.yin@astrazeneca.com CR Kaur A, Dasanu CA, 2011, Targeting the her2 pathway for the therapy of lower esophageal and gastric adenocarcinoma. Expert Opin Pharmacother 12:2493–2503 Power DG, Kelsen DP, Shah MA, 2010, Advanced gastric cancer– slow but steady progress. Cancer Treat Rev 36:384–392 Kelley JR, Duggan JM, 2003, Gastric cancer epidemiology and risk factors. J Clin Epidemiol 56:1–9 Shah MA, Kelsen DP, 2010, Gastric cancer: a primer on the epidemiology and biology of the disease and an overview of the medical management of advanced disease. J Natl Compr Canc Netw 8:437– 447 Lordick F, 2011, Trastuzumab: a new treatment option for her2- positive metastatic gastric and gastroesophageal junction cancer. Future Oncol 7:187–199 Kamangar F, Dores GM, Anderson WF, 2006, Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol 24:2137–2150 Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, Scarffe JH, Lofts FJ, Falk SJ, Iveson TJ, Smith DB, Langley RE, Verma M, Weeden S, Chua YJ, Participants MT, 2006, Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med 355:11– 20 Lorenzen S, Lordick F, 2011, How will human epidermal growth factor receptor 2-neu data impact clinical management of gastric cancer? Curr Opin Oncol 23:396–402 Gravalos C, Jimeno A, 2008, Her2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol 19:1523–1529 Sakai K, Mori S, Kawamoto T, Taniguchi S, Kobori O, Morioka Y, Kuroki T, Kano K, 1986, Expression of epidermal growth factor receptors on normal human gastric epithelia and gastric carcinomas. J Natl Cancer Inst 77:1047–1052 TaiW, Mahato R, Cheng K, 2010, The role of her2 in cancer therapy and targeted drug delivery. J Control Release 146:264–275 Meza-Junco J, Au HJ, Sawyer MB, 2009, Trastuzumab for gastric cancer. Expert Opin Biol Ther 9:1543–1551 BangYJ,Van CutsemE, Feyereislova A, ChungHC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T, Aprile G, Kulikov E, Hill J, Lehle M, Ruschoff J, Kang YK, 2010, Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of her2-positive advanced gastric or gastro-oesophageal junction cancer, toga): A phase 3, open-label, randomised controlled trial. Lancet 376:687–697 Albarello L, Pecciarini L, Doglioni C, 2011, Her2 testing in gastric cancer. Adv Anat Pathol 18:53–59 Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, McShane LM, Paik S, Pegram MD, Perez EA, Press MF, Rhodes A, Sturgeon C, Taube SE, Tubbs R, Vance GH, van de Vijver M, Wheeler TM, Hayes DF, 2007, American society of clinical oncology/college of american pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 131:18–43 Hofmann M, Stoss O, Shi D, Buttner R, van de Vijver M, Kim W, Ochiai A, Ruschoff J, Henkel T, 2008, Assessment of a her2 scoring system for gastric cancer: results from a validation study. Histopathology 52:797–805 Kurosumi M, 2009, Recent trends of her-2 testing and trastuzumab therapy for breast cancer. Breast Cancer 16:284–287 Sato-Kuwabara Y, Neves JI, Fregnani JH, Sallum RA, Soares FA, 2009, Evaluation of gene amplification and protein expression of her-2/neu in esophageal squamous cell carcinoma using fluorescence in situ hybridization, fish, and immunohistochemistry. BMC Cancer 9:6 Wu S, Zhu Z, He J, Luo X, Xu J, Ren-Heidenreich L, 2010, A novel mutant-enriched liquidchip technology for the qualitative detection of somatic mutations in kras gene from both serum and tissue samples. Clin Chem Lab Med 48:1103–1106 Furukawa T, Fu X, Kubota T, Watanabe M, Kitajima M, Hoffman RM(1993, Nude mousemetastatic models of human stomach cancer constructed using orthotopic implantation of histologically intact tissue. Cancer Res 53:1204–1208 Fidler IJ, 1990, Critical factors in the biology of human cancer metastasis: twenty-eighth g.H.A. Clowes memorial award lecture. Cancer Res 50:6130–6138 Garber K, 2009, From human to mouse and back: ‘tumorgraft’ models surge in popularity. J Natl Cancer Inst 101:6–8 Yano T, Doi T, Ohtsu A, Boku N, Hashizume K, Nakanishi M, Ochiai A, 2006, Comparison of her2 gene amplification assessed by fluorescence in situ hybridization and her2 protein expression assessed by immunohistochemistry in gastric cancer. Oncol Rep 15:65–71 Mackenzie M, Spithoff K, Jonker D, 2011, Systemic therapy for advanced gastric cancer: a clinical practice guideline. Curr Oncol 18:e202–209 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 947 EP 955 DI 10.1007/s12253-015-9909-8 PG 9 ER PT J AU Garay, T Molnar, E Juhasz, Laszlo, V Barbai, T Dobos, J Schelch, K Pirker, Ch Grusch, M Berger, W Timar, J Hegedus, B AF Garay, Tamas Molnar, Eszter Juhasz, Eva Laszlo, Viktoria Barbai, Tamas Dobos, Judit Schelch, Karin Pirker, Christine Grusch, Michael Berger, Walter Timar, Jozsef Hegedus, Balazs TI Sensitivity of Melanoma Cells to EGFR and FGFR Activation but Not Inhibition is Influenced by Oncogenic BRAF and NRAS Mutations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Melanoma; BRAF; NRAS; Mutation; EGF; FGF2; EGFR inhibitor; FGFR inhibitor ID Melanoma; BRAF; NRAS; Mutation; EGF; FGF2; EGFR inhibitor; FGFR inhibitor AB BRAF and NRAS are the two most frequent oncogenic driver mutations in melanoma and are pivotal components of both the EGF and FGF signaling network. Accordingly, we investigated the effect of BRAF and NRAS oncogenic mutation on the response to the stimulation and inhibition of epidermal and fibroblast growth factor receptors in melanoma cells. In the three BRAF mutant, two NRAS mutant and two double wild-type cell lines growth factor receptor expression had been verified by qRT-PCR. Cell proliferation and migration were determined by the analysis of 3- days-long time-lapse videomicroscopic recordings. Of note, a more profound response was found in motility as compared to proliferation and double wild-type cells displayed a higher sensitivity to EGF and FGF2 treatment when compared to mutant cells. Both baseline and induced activation of the growth factor signaling was assessed by immunoblot analysis of the phosphorylation of the downstream effectors Erk1/2. Low baseline and higher inducibility of the signaling pathway was characteristic in double wild-type cells. In contrast, oncogenic BRAF or NRAS mutation did not influence the response to EGF or FGF receptor inhibitors in vitro. Our findings demonstrate that the oncogenic mutations in melanoma have a profound impact on the motogenic effect of the activation of growth factor receptor signaling. Since emerging molecularly targeted therapies aim at the growth factor receptor signaling, the appropriate mutational analysis of individual melanoma cases is essential in both preclinical studies and in the clinical trials and practice. C1 [Garay, Tamas] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Molnar, Eszter] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Juhasz, Eva] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Laszlo, Viktoria] Medical University of Vienna, Department of Thoracic Surgery, Spitalgasse 23, A-1090 Vienna, Austria. [Barbai, Tamas] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Dobos, Judit] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Schelch, Karin] Medical University of Vienna, Department of Experimental Pathology and Laboratory Animal Pathology, Borschkegasse 8a, A-1090 Vienna, Austria. [Pirker, Christine] Medical University of Vienna, Department of Experimental Pathology and Laboratory Animal Pathology, Borschkegasse 8a, A-1090 Vienna, Austria. [Grusch, Michael] Medical University of Vienna, Department of Experimental Pathology and Laboratory Animal Pathology, Borschkegasse 8a, A-1090 Vienna, Austria. [Berger, Walter] Medical University of Vienna, Department of Experimental Pathology and Laboratory Animal Pathology, Borschkegasse 8a, A-1090 Vienna, Austria. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Hegedus, Balazs] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. RP Hegedus, B (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. CR Zhang X, Ibrahimi OA, Olsen SK, Umemori H, Mohammadi M, Ornitz DM(2006, Receptor specificity of the fibroblast growth factor family. The complete mammalian FGF family. J Biol Chem 281(23): 15694–15700., DOI 10.1074/jbc.M601252200 Ornitz DM, Xu J, Colvin JS, McEwen DG, MacArthur CA, Coulier F, Gao G, Goldfarb M, 1996, Receptor specificity of the fibroblast growth factor family. J Biol Chem 271(25):15292–15297 Heinzle C, Sutterluty H, Grusch M, Grasl-Kraupp B, Berger W, Marian B, 2011, Targeting fibroblast-growth-factor-receptordependent signaling for cancer therapy. Expert Opin Ther Targets 15(7):829–846., DOI 10.1517/14728222.2011.566217 Harris RC, Chung E, Coffey RJ, 2003, EGF receptor ligands. Exp Cell Res 284(1):2–13 Dreux AC, Lamb DJ, Modjtahedi H, FernsGA(2006, The epidermal growth factor receptors and their family of ligands: their putative role in atherogenesis. Atherosclerosis 186(1):38–53., DOI 10.1016/j. atherosclerosis.2005.06.038 Cotton LM, O’Bryan MK, Hinton BT, 2008, Cellular signaling by fibroblast growth factors, FGFs, and their receptors, FGFRs, in male reproduction. Endocr Rev 29(2):193–216., DOI 10.1210/er.2007-0028 Maruta H, Burgess AW, 1994, Regulation of the Ras signalling network. BioEssays: News Rev Mol Cell Dev Biol 16(7):489–496., DOI 10.1002/bies.950160708 Liang G, Liu Z, Wu J, Cai Y, Li X, 2012, Anticancer molecules targeting fibroblast growth factor receptors. Trends Pharmacol Sci 33(10):531–541., DOI 10.1016/j.tips.2012.07.001 Ghosh P, Chin L, 2009, Genetics and genomics ofmelanoma. Expert Rev Dermatol 4(2):131., DOI 10.1586/edm.09.2 Dutta PR, Maity A, 2007, Cellular responses to EGFR inhibitors and their relevance to cancer therapy. Cancer Lett 254(2):165–177., DOI 10.1016/j.canlet.2007.02.006 Curtin JA, Fridlyand J, Kageshita T, PatelHN, BusamKJ, Kutzner H, Cho KH, Aiba S, Brocker EB, LeBoit PE, Pinkel D, Bastian BC, 2005, Distinct sets of genetic alterations in melanoma. N Engl J Med 353(20):2135–2147., DOI 10.1056/NEJMoa050092 Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J,Woffendin H, GarnettMJ, BottomleyW, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA, 2002, Mutations of the BRAF gene in human cancer. Nature 417(6892):949–954., DOI 10.1038/nature00766 Demunter A, Stas M, Degreef H, DeWolf-Peeters C, van den Oord JJ, 2001, Analysis of N- and K-ras mutations in the distinctive tumor progression phases of melanoma. J Invest Dermatol 117(6):1483– 1489., DOI 10.1046/j.0022-202x.2001.01601.x Houben R, Becker JC, Kappel A, Terheyden P, Brocker EB, Goetz R, Rapp UR, 2004, Constitutive activation of the Ras-Raf signaling pathway in metastatic melanoma is associated with poor prognosis. J Carcinog 3(1):6., DOI 10.1186/1477-3163-3-6 Kumar R, Angelini S, Czene K, Sauroja I, Hahka-Kemppinen M, Pyrhonen S, Hemminki K, 2003, BRAF mutations in metastatic melanoma: a possible association with clinical outcome. Clin Cancer Res 9(9):3362–3368 Maldonado JL, Fridlyand J, Patel H, Jain AN, BusamK, Kageshita T, Ono T, Albertson DG, Pinkel D, Bastian BC, 2003, Determinants of BRAF mutations in primary melanomas. J Natl Cancer Inst 95(24): 1878–1890 Tsao H, Goel V,Wu H, Yang G, Haluska FG, 2004, Genetic interaction between NRAS and BRAF mutations and PTEN/MMAC1 inactivation in melanoma. J Invest Dermatol 122(2):337–341., DOI 10. 1046/j.0022-202X.2004.22243.x Meier F, Schittek B, Busch S, Garbe C, Smalley K, Satyamoorthy K, Li G, Herlyn M, 2005, The RAS/RAF/MEK/ERK and PI3K/AKT signaling pathways present molecular targets for the effective treatment of advanced melanoma. Front Biosci 10:2986–3001 Sinnberg T, Lasithiotakis K, Niessner H, Schittek B, Flaherty KT, Kulms D, Maczey E, Campos M, Gogel J, Garbe C, Meier F, 2009, Inhibition of PI3K-AKT-mTOR signaling sensitizes melanoma cells to cisplatin and temozolomide. J Invest Dermatol 129(6): 1500–1515., DOI 10.1038/jid.2008.379 Collisson EA, De A, Suzuki H, Gambhir SS, Kolodney MS, 2003, Treatment of metastatic melanoma with an orally available inhibitor of the Ras-Raf-MAPK cascade. Cancer Res 63(18):5669–5673 Carragher NO,Westhoff MA, Fincham VJ, Schaller MD, FrameMC, 2003, A novel role for FAK as a protease-targeting adaptor protein: regulation by p42 ERK and Src. Curr Biol 13(16):1442–1450 Lazar-Molnar E, Hegyesi H, Toth S, Falus A, 2000, Autocrine and paracrine regulation by cytokines and growth factors in melanoma. Cytokine 12(6):547–554., DOI 10.1006/cyto.1999.0614 Feinmesser M, Veltman V, Morgenstern S, Tobar A, Gutman H, Kaganovsky E, Tzabari C, Sulkes J,Okon E, 2010, Different patterns of expression of the erbB family of receptor tyrosine kinases in common nevi, dysplastic nevi, and primary malignant melanomas: an immunohistochemical study. Am J Dermatopathol 32(7):665–675., DOI 10.1097/DAD.0b013e3181d1e6f0 Boone B, Jacobs K, Ferdinande L, Taildeman J, Lambert J, Peeters M, Bracke M, Pauwels P, Brochez L, 2011, EGFR in melanoma: clinical significance and potential therapeutic target. J Cutan Pathol 38(6):492–502., DOI 10.1111/j.1600-0560.2011.01673.x Rakosy Z, Vizkeleti L, Ecsedi S, Voko Z, Begany A, Barok M, Krekk Z, Gallai M, Szentirmay Z, Adany R, Balazs M, 2007, EGFR gene copy number alterations in primary cutaneous malignant melanomas are associated with poor prognosis. Int J Cancer 121(8):1729–1737., DOI 10.1002/ijc.22928 Gordon-Thomson C, Mason RS, Moore GP, 2001, Regulation of epidermal growth factor receptor expression in human melanocytes. Exp Dermatol 10(5):321–328 Timar J, Gyorffy B, Raso E, 2010, Gene signature of the metastatic potential of cutaneous melanoma: too much for too little? Clin Exp Metastasis 27(6):371–387., DOI 10.1007/s10585-010-9307-2 Bracher A, Cardona AS, Tauber S, FinkAM, Steiner A, Pehamberger H, Niederleithner H, Petzelbauer P, Groger M, Loewe R, 2013, Epidermal growth factor facilitates melanoma lymph node metastasis by influencing tumor lymphangiogenesis. J Invest Dermatol 133(1): 230–238., DOI 10.1038/jid.2012.272 Becker D, Lee PL, Rodeck U, Herlyn M, 1992, Inhibition of the fibroblast growth factor receptor 1, FGFR-1, gene in human melanocytes and malignant melanomas leads to inhibition of proliferation and signs indicative of differentiation. Oncogene 7(11):2303–2313 Easty DJ, Ganz SE, Farr CJ, Lai C, Herlyn M, Bennett DC, 1993, Novel and known protein tyrosine kinases and their abnormal expression in human melanoma. J Invest Dermatol 101(5):679–684 Yayon A, Ma YS, Safran M, Klagsbrun M, Halaban R, 1997, Suppression of autocrine cell proliferation and tumorigenesis of human melanoma cells and fibroblast growth factor transformed fibroblasts by a kinase-deficient FGF receptor 1: evidence for the involvement of Src-family kinases. Oncogene 14(25):2999–3009., DOI 10. 1038/sj.onc.1201159 Streit S,Mestel DS, Schmidt M, Ullrich A, Berking C, 2006, FGFR4 Arg388 allele correlates with tumour thickness and FGFR4 protein expression with survival of melanoma patients. Br J Cancer 94(12): 1879–1886., DOI 10.1038/sj.bjc.6603181 Gartside MG, Chen H, Ibrahimi OA, Byron SA, Curtis AV, Wellens CL, Bengston A, Yudt LM, Eliseenkova AV, Ma J, Curtin JA, Hyder P, Harper UL, Riedesel E, Mann GJ, Trent JM, Bastian BC, Meltzer PS, Mohammadi M, Pollock PM, 2009, Loss-of-function fibroblast growth factor receptor-2 mutations in melanoma. Mol Cancer Res 7(1):41–54., DOI 10.1158/1541-7786.mcr-08-0021 Halaban R, Langdon R, Birchall N, Cuono C, Baird A, Scott G, Moellmann G,McGuire J, 1988, Basic fibroblast growth factor from human keratinocytes is a naturalmitogen for melanocytes. J Cell Biol 107(4):1611–1619 Dotto GP, Moellmann G, Ghosh S, Edwards M, Halaban R, 1989, Transformation of murine melanocytes by basic fibroblast growth factor cDNA and oncogenes and selective suppression of the transformed phenotype in a reconstituted cutaneous environment. J Cell Biol 109(6 Pt 1):3115–3128 Nesbit M, Nesbit HK, Bennett J, Andl T, Hsu MY, Dejesus E, McBrian M, Gupta AR, Eck SL, Herlyn M, 1999, Basic fibroblast growth factor induces a transformed phenotype in normal human melanocytes. Oncogene 18(47):6469–6476., DOI 10.1038/sj.onc. 1203066 Wang Y, Becker D, 1997, Antisense targeting of basic fibroblast growth factor and fibroblast growth factor receptor-1 in human melanomas blocks intratumoral angiogenesis and tumor growth. Nat Med 3(8):887–893 Chalkiadaki G, Nikitovic D, Berdiaki A, Sifaki M, Krasagakis K, Katonis P, Karamanos NK, Tzanakakis GN, 2009, Fibroblast growth factor-2 modulates melanoma adhesion and migration through a syndecan-4-dependent mechanism. Int J Biochem Cell Biol 41(6): 1323–1331., DOI 10.1016/j.biocel.2008.11.008 Becker D, Meier CB, Herlyn M, 1989, Proliferation of human malignant melanomas is inhibited by antisense oligodeoxynucleotides targeted against basic fibroblast growth factor. EMBO J 8(12):3685– 3691 Ozen M, Medrano EE, Ittmann M, 2004, Inhibition of proliferation and survival of melanoma cells by adenoviral-mediated expression of dominant negative fibroblast growth factor receptor. Melanoma Res 14(1):13–21 Li D, Wang H, Xiang JJ, Deng N, Wang PP, Kang YL, Tao J, Xu M, 2010, Monoclonal antibodies targeting basic fibroblast growth factor inhibit the growth of B16 melanoma in vivo and in vitro. Oncol Rep 24(2):457–463 Blanco Codesido M, Tesainer Brunetto A, Frentzas S, Moreno Garcia V, Papadatos-Pastos D, Pedersen JV, Trani L, Puglisi M, Molife LR, Banerji U, 2011, Outcomes of patients with metastatic melanoma treated with molecularly targeted agents in phase I clinical trials. Oncology 81(2):135–140., DOI 10.1159/000330206 Ji Z, Flaherty KT, Tsao H, 2012, Targeting the RAS pathway in melanoma. Trends Mol Med 18(1):27–35., DOI 10.1016/j.molmed. 2011.08.001 Djerf EA, Trinks C, Abdiu A, Thunell LK, Hallbeck AL, Walz TM, 2009, ErbB receptor tyrosine kinases contribute to proliferation of malignant melanoma cells: inhibition by gefitinib, ZD1839). Melanoma Res 19(3):156–166 . d o i :10.1097/CMR. 0b013e32832c6339 Patel SP, Kim KB, Papadopoulos NE, Hwu WJ, Hwu P, Prieto VG, Bar-Eli M, Zigler M, Dobroff A, Bronstein Y, Bassett RL, Vardeleon AG, Bedikian AY, 2011)A phase II study of gefitinib in patients with metastatic melanoma. Melanoma Res 21(4):357–363., DOI 10.1097/ CMR.0b013e3283471073 Schicher N, Paulitschke V, Swoboda A, Kunstfeld R, Loewe R, Pilarski P, Pehamberger H, Hoeller C, 2009, Erlotinib and bevacizumab have synergistic activity against melanoma. Clin Cancer Res 15(10):3495–3502., DOI 10.1158/1078-0432.ccr-08-2407 Djerf Severinsson EA, Trinks C, Green H, Abdiu A, Hallbeck AL, Stal O, Walz TM, 2011, The pan-ErbB receptor tyrosine kinase inhibitor canertinib promotes apoptosis of malignantmelanoma in vitro and displays anti-tumor activity in vivo. Biochem Biophys Res Commun 414(3):563–568., DOI 10.1016/j.bbrc.2011.09.118 Kim H, Lim HY, 2011, Novel EGFR-TK inhibitor EKB-569 inhibits hepatocellular carcinoma cell proliferation by AKT andMAPK pathways. J KoreanMed Sci 26(12):1563–1568., DOI 10.3346/jkms.2011. 26.12.1563 Kwak EL, Sordella R, Bell DW, Godin-Heymann N, Okimoto RA, Brannigan BW, Harris PL, Driscoll DR, Fidias P, Lynch TJ, Rabindran SK, McGinnis JP, Wissner A, Sharma SV, Isselbacher KJ, Settleman J, Haber DA, 2005, Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proc Natl Acad Sci U S A 102(21):7665–7670., DOI 10.1073/pnas. 2860102 50. Bryce AH, Rao R, Sarkaria J, Reid JM, Qi Y, Qin R, James CD, Jenkins RB, Boni J, Erlichman C, Haluska P, 2012, Phase I study of temsirolimus in combination with EKB-569 in patients with advanced solid tumors. Investig New Drugs 30(5):1934–1941., DOI 10. 1007/s10637-011-9742-1 Metzner T, Bedeir A, Held G, Peter-Vorosmarty B, Ghassemi S, Heinzle C, Spiegl-Kreinecker S, Marian B, Holzmann K, Grasl- Kraupp B, Pirker C, Micksche M, Berger W, Heffeter P, Grusch M, 2011, Fibroblast growth factor receptors as therapeutic targets in human melanoma: synergism with BRAF inhibition. J Invest Dermatol 131(10):2087–2095., DOI 10.1038/jid.2011.177 Torok S, Cserepes TM, Renyi-Vamos F, Dome B, 2012, Nintedanib, BIBF 1120, in the treatment of solid cancers: an overview of biological and clinical aspects. Magy Onkol 56(3):199–208,, DOI MagyOnkol.2012.56.3.199 Katoh M, Nakagama H, 2013, FGF receptors: cancer biology and therapeutics. Med Res Rev., DOI 10.1002/med.21288 Hilberg F, Roth GJ, Krssak M, Kautschitsch S, Sommergruber W, Tontsch-Grunt U, Garin-Chesa P, Bader G, Zoephel A, Quant J, Heckel A, Rettig WJ, 2008, BIBF 1120: triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res 68(12):4774–4782., DOI 10.1158/0008-5472.can-07-6307 O’Hare T, Shakespeare WC, Zhu X, Eide CA, Rivera VM, Wang F, Adrian LT, Zhou T, Huang WS, Xu Q, Metcalf CA 3rd, Tyner JW, Loriaux MM, Corbin AS, Wardwell S, Ning Y, Keats JA, Wang Y, Sundaramoorthi R, ThomasM, Zhou D, Snodgrass J, Commodore L, Sawyer TK, Dalgarno DC, Deininger MW, Druker BJ, Clackson T, 2009, AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell 16(5):401–412., DOI 10. 1016/j.ccr.2009.09.028 Gozgit JM, Wong MJ, Moran L, Wardwell S, Mohemmad QK, Narasimhan NI, Shakespeare WC, Wang F, Clackson T, Rivera VM, 2012, Ponatinib, AP24534), a multitargeted pan-FGFR inhibitor with activity in multiple FGFR-amplified or mutated cancer models. Mol Cancer Ther 11(3):690–699., DOI 10.1158/1535-7163. mct-11-0450 Guagnano V, Furet P, Spanka C, Bordas V, Le Douget M, Stamm C, Brueggen J, Jensen MR, Schnell C, Schmid H, Wartmann M, Berghausen J, Drueckes P, Zimmerlin A, Bussiere D, Murray J, Graus Porta D, 2011, Discovery of 3-(2,6-dichloro-3,5-dimethoxyphenyl)- 1-{6-[4-(4-ethyl-piperazin-1-yl)-phenylamin o]-pyrimidin- 4-yl}-1-methyl-urea, NVP-BGJ398), a potent and selective inhibitor of the fibroblast growth factor receptor family of receptor tyrosine kinase. J Med Chem 54(20):7066–7083., DOI 10.1021/jm2006222 Cheng T, Roth B, Choi W, Black PC, Dinney C, McConkey DJ, 2013, Fibroblast growth factor receptors-1 and -3 play distinct roles in the regulation of bladder cancer growth and metastasis: implications for therapeutic targeting. PLoS One 8(2):e57284., DOI 10.1371/ journal.pone.0057284 Gavine PR,Mooney L, Kilgour E, Thomas AP, Al-Kadhimi K, Beck S, Rooney C, Coleman T, Baker D, Mellor MJ, Brooks AN, Klinowska T, 2012, AZD4547: an orally bioavailable, potent, and selective inhibitor of the fibroblast growth factor receptor tyrosine kinase family. Cancer Res 72(8):2045–2056., DOI 10.1158/0008- 5472.can-11-3034 Yadav V, Zhang X, Liu J, Estrem S, Li S, Gong XQ, Buchanan S, Henry JR, Starling JJ, Peng SB, 2012, Reactivation of mitogenactivated protein kinase, MAPK, pathway by FGF receptor 3, FGFR3)/Ras mediates resistance to vemurafenib in human B-RAF V600E mutant melanoma. J Biol Chem 287(33):28087–28098., DOI 10.1074/jbc.M112.377218 Girotti MR, Pedersen M, Sanchez-Laorden B, Viros A, Turajlic S, Niculescu-Duvaz D, Zambon A, Sinclair J, Hayes A, Gore M, Lorigan P, Springer C, Larkin J, Jorgensen C, Marais R, 2013, Inhibiting EGF receptor or SRC family kinase signaling overcomes BRAF inhibitor resistance in melanoma. Cancer discovery 3(2):158– 167., DOI 10.1158/2159-8290.cd-12-0386 Mueller BM, Romerdahl CA, Trent JM, Reisfeld RA, 1991, Suppression of spontaneous melanoma metastasis in scid mice with an antibody to the epidermal growth factor receptor. Cancer Res 51(8):2193–2198 Ladanyi A, Gallai M, Paku S, Nagy JO, Dudas J, Timar J, Kovalszky I, 2001, Expression of a decorin-like moleculein human melanoma. Pathol Oncol Res 7(4):260–266 Berger W, Elbling L, Minai-Pour M, Vetterlein M, Pirker R, Kokoschka EM, Micksche M, 1994, Intrinsic MDR-1 gene and Pglycoprotein expression in human melanoma cell lines. Int J Cancer 59(5):717–723 Garay T, Juhasz E, Molnar E, Eisenbauer M, Czirok A, Dekan B, Laszlo V, Hoda MA, Dome B, Timar J, Klepetko W, Berger W, Hegedus B, 2013, Cell migration or cytokinesis and proliferation? - revisiting the "go or grow" hypothesis in cancer cells in vitro. Exp Cell Res., DOI 10.1016/j.yexcr.2013.08.018 Hegedus B, Zach J, Czirok A, Lovey J, Vicsek T, 2004, Irradiation and Taxol treatment result in non-monotonous, dose-dependent changes in the motility of glioblastoma cells. J Neuro-Oncol 67(1– 2):147–157 Easty DJ, Gray SG, O’Byrne KJ, O’Donnell D, Bennett DC, 2011, Receptor tyrosine kinases and their activation in melanoma. Pigment Cell Melanoma Res 24(3):446–461., DOI 10.1111/j.1755-148X.2011. 00836.x Jakob JA, Bassett RL Jr, Ng CS, Curry JL, Joseph RW, Alvarado GC, Rohlfs ML, Richard J, Gershenwald JE, Kim KB, Lazar AJ, Hwu P, Davies MA, 2012, NRAS mutation status is an independent prognostic factor in metastatic melanoma. Cancer 118(16):4014–4023., DOI 10.1002/cncr.26724 Safaee Ardekani G, Jafarnejad SM, Tan L, Saeedi A, LiG, 2012, The prognostic value of BRAF mutation in colorectal cancer and melanoma: a systematic review and meta-analysis. PLoS One 7(10): e47054., DOI 10.1371/journal.pone.0047054 Houben R, Vetter-Kauczok CS, Ortmann S, Rapp UR, Broecker EB, Becker JC, 2008, Phospho-ERK staining is a poor indicator of the mutational status of BRAF and NRAS in human melanoma. J Invest Dermatol 128(8):2003–2012., DOI 10.1038/jid.2008.30 Yazdi AS, Ghoreschi K, Sander CA, Rocken M(2010, Activation of the mitogen-activated protein kinase pathway in malignant melanoma can occur independently of the BRAF T1799A mutation. Eur J Dermatol EJD 20(5):575–579., DOI 10.1684/ejd.2010.1011 Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, KimS,Wilson CJ, Lehar J,KryukovGV, Sonkin D, ReddyA, Liu M, Murray L, BergerMF,Monahan JE, Morais P,Meltzer J, Korejwa A, Jane-Valbuena J, Mapa FA, Thibault J, Bric-Furlong E, Raman P, Shipway A, Engels IH, Cheng J, Yu GK, Yu J, Aspesi P Jr, de Silva M, Jagtap K, Jones MD, Wang L, Hatton C, Palescandolo E, Gupta S, Mahan S, Sougnez C, Onofrio RC, Liefeld T, MacConaill L, WincklerW, Reich M, Li N, Mesirov JP, Gabriel SB, Getz G, Ardlie K, Chan V, Myer VE, Weber BL, Porter J, Warmuth M, Finan P, Harris JL, Meyerson M, Golub TR, Morrissey MP, Sellers WR, Schlegel R, Garraway LA, 2012, The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483(7391):603–607., DOI 10.1038/nature11003 Kim KB, Chesney J, Robinson D, Gardner H, Shi MM, Kirkwood JM, 2011, Phase I/II and pharmacodynamic study of dovitinib, TKI258), an inhibitor of fibroblast growth factor receptors and VEGF receptors, in patients with advanced melanoma. Clin Cancer Res 17(23):7451–7461., DOI 10.1158/1078-0432.ccr-11-1747 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 957 EP 968 DI 10.1007/s12253-015-9916-9 PG 12 ER PT J AU Tsvetkova, A Todorova, A Todorov, T Georgiev, G Drandarska, I Mitev, V AF Tsvetkova, Anita Todorova, Albena Todorov, Tihomir Georgiev, Georgi Drandarska, Ivanka Mitev, Vanyo TI Molecular and Clinicopathological Aspects of Prostate Cancer in Bulgarian Probands SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CRPC; Molecular subtyping; Prostate cancer; PCA3; TMPRSS2:ERG gene fusions; Prognosis ID CRPC; Molecular subtyping; Prostate cancer; PCA3; TMPRSS2:ERG gene fusions; Prognosis AB To correlate the molecular data to the clinicopathological parameters in Bulgarian prostate cancer patients. PCA3 overexpression, TMPRSS2-ERG gene fusion, GSTP1 promoter hypermethylation, somatic mutations in the AR gene and the IVS1-27G > A polymorphism in the KLF6 gene were studied. A total of 148 patients were analyzed: 16 aggressive PCa, 83 non-aggressive PCa, 25 BPH and 24 chronic inflammatory diseases. Real-time RT-PCR, DNA sequencing, and bisulfite conversion of DNA, were applied. All cases with aggressive PCa before treatment were tested positive for PCA3 overexpression, expression of a T2-ERG gene fusion product and GSTP1 promoter hypermethylation. No somatic mutations were detected in the AR gene and all patients showed normal KLF6-IVS1-27G > A genotype. The TMPRSS2-ERG positive status correlates with moderate to poorly differentiated prostate tumors and it is considered as unfavorable disease predictor. Positive GSTP1 promoter hypermethylation seems to be highly specific and the earliest epigenetic change in the prostate gland, which indicates the beginning of the pathological process. The appearance of positive molecular markers in blood was considered as a predictor of PCa dissemination. GSTP1 promoter hypermethylation was found as the earliest and a long-lasting epigenetic marker in blood samples of PCa patients, which makes it suitable as a marker for treatment follow-up. The molecular profile of prostate cancer needs to be strictly monitored during the course of disease treatment, which is of a great help in determining the patient’s individual therapy response. C1 [Tsvetkova, Anita] Medical University - Sofia, Department of Medical Chemistry and BiochemistrySofia, Bulgaria. [Todorova, Albena] Medical University - Sofia, Department of Medical Chemistry and BiochemistrySofia, Bulgaria. [Todorov, Tihomir] Genetic Medico-Diagnostic LaboratorySofia, Bulgaria. [Georgiev, Georgi] Military Medical Academy, Department of UrologySofia, Bulgaria. [Drandarska, Ivanka] Military Medical Academy, Department of PathologySofia, Bulgaria. [Mitev, Vanyo] Medical University - Sofia, Department of Medical Chemistry and BiochemistrySofia, Bulgaria. RP Tsvetkova, A (reprint author), Medical University - Sofia, Department of Medical Chemistry and Biochemistry, Sofia, Bulgaria. EM annita_25@abv.bg CR Salagierski M, Schalken JA, 2010, PCA3 and TMPRSS2-ERG: promising biomarkers in prostate cancer diagnosis. Cancers 2(3): 1432–1440., DOI 10.3390/cancers2031432 Heidenreich A, Aus G, Bolla M, Joniau S, Matveev VB, Schmid HP et al, 2008, EAUguidelines on prostate cancer. Eur Urol 53(1):68–80 de Kok JB, Verhaegh GW, Roelofs RW, Hessels D, Kiemeney LA, Aalders TWet al, 2002, DD3PCA3, a very sensitive and specific marker to detect prostate tumors. Cancer Res 62(9):2695–8 Tomlins SA, Rhodes DR, Perner S,Dhanasekaran SM, Mehra R, Sun XWet al, 2005, Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310(5748):644–8 Gonzalgo ML, Pavlovich CP, Lee SM, Nelson WG, 2003, Prostate cancer detection by GSTP1 methylation analysis of postbiopsy urine specimens. Clin Cancer Res 9(7):2673–7 Thomas G, Jacobs KB, Yeager M, Kraft P, Wacholder S, Orr N et al, 2008, Multiple loci identified in a genome-wide association study of prostate cancer. Nat Genet 40:310–315., DOI 10.1038/ng.91 Eeles RA, Kote-Jarai Z, Giles GG, Olama AA, Guy M, Jugurnauth SK et al, 2008, Multiple newly identified loci associated with prostate cancer susceptibility. Nat Genet 40(3):316–21., DOI 10.1038/ng. 90 Mitelman F, Johansson B, Mertens F, 2007, The impact of translocations and gene fusions on cancer causation. Nat Rev Cancer 7(4): 233–45 Morris DS, Tomlins SA, Montie JE, Chinnaiyan AM, 2008, The discovery and application of gene fusions in prostate cancer. BJU Int 102(3):276–82 Barry M, Perner S, Demichelis F, Rubin A, 2007, TMPRSS2-ERG fusion heterogeneity in multifocal prostate cancer: clinical and biologic implications. Urology 70(4):630–3 Demichelis F, Fall K, Perner S, Andren O, Schmidt F, Setlur SR et al, 2007, TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene 26(31):4596–9 Clark JP, Munson KW, Gu JW, Lamparska-Kupsik K, Chan KG, Yoshida JS et al, 2008, Performance of a single assay for both type III and type VI TMPRSS2:ERG fusions in noninvasive prediction of prostate biopsy outcome. Clin Chem 54(12):2007–17 Wang J, Cai Y, Ren C, Ittmann M, 2006, Expression of variant TMPRSS2/ERG fusion messenger RNAs is associated with aggressive prostate cancer. Cancer Res 66(17):8347–51 Hessels D, Smit FP, Verhaegh GW, Witjes JA, Cornel EB, Schalken JA, 2007, Detection of TMPRSS2-ERG fusion transcripts and prostate cancer antigen 3 in urinary sediments may improve diagnosis of prostate cancer. Clin Cancer Res 13(17):5103–8 Kirby RS, Christmas TJ, Brawer MK, 2001, Prostate Cancer. London Schroder FH, 2005, Detection of prostate cancer: the impact of the European Randomized Study of Screening for Prostate Cancer, ERSPC). Can J Urol 1:2–6, 92–3 Yoshimoto M, Joshua AM, Cunha IW, Coudry RA, Fonseca FP, Ludkovski O et al, 2008, Absence of TMPRSS2:ERG fusions and PTEN losses in prostate cancer is associated with a favorable outcome. Mod Pathol 21(12):1451–60 ShenMM, Abate-Shen C, 2010, Molecular genetics of prostate cancer: new prospects for old challenges genes. Genes Dev 24(18): 1967–2000 Tomlins SA, Laxman B, Dhanasekaran SM, Helgeson BE, Cao X, Morris DS et al, 2007, Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature 448(7153):595–9 Rajput AB, Miller MA, De Luca A, Boyd N, Leung S, Hurtado-Coll A et al, 2007, Frequency of the TMPRSS2:ERG gene fusion is increased in moderate to poorly differentiated prostate cancers. J Clin Pathol 60(11):1238–1243 Eisermann K,Wang D, Jing Y, Pascal LE,Wang Z, 2013, Androgen receptor gene mutation, rearrangement, polymorphism. Transl Androl Urol 2(3):137–147 Narla G, Difeo A, Reeves HL, Schaid DJ, Hirshfeld J, Hod E et al, 2005)A germline DNA polymorphism enhances alternative splicing of the KLF6 tumor suppressor gene and is associated with increased prostate cancer risk. Cancer Res 65(4):1213–22 Narla G, DiFeo A, Fernandez Y, Dhanasekaran S, Huang F, Sangodkar J et al, 2008, KLF6-SV1 overexpression accelerates human and mouse prostate cancer progression and metastasis. Clin Invest 118(8):2711–21 BussemakersMJ, van Bokhoven A, Verhaegh GWet al, 1999, DD3: a new prostate-specific gene, highly overexpressed in prostate cancer. Cancer Res 59(23):5975–9 Brooks JD,Weinstein M, Lin X, Sun Y, Pin SS, Bova GS et al, 1998, CG island methylation changes near the GSTP1 gene in prostatic intraepithelial neoplasia. Cancer Epidemiol Biomarkers Prev 7(6): 531–6 Nakayama M, Bennett CJ, Hicks JL, Epstein JI, Platz EA, Nelson WG et al, 2003, Hypermethylation of the human glutathione Stransferase- gene, GSTP1, CpG island is present in a subset of proliferative inflammatory atrophy lesions but not in normal or hyperplastic epithelium of the prostate: a detailed studyusing laser-capture microdissection. Am J Pathol 163(3):923–933 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 969 EP 976 DI 10.1007/s12253-015-9915-x PG 8 ER PT J AU Rusz, O Voros, A Varga, Z Kelemen, Gy Uhercsak, G Nikolenyi, A Ormandi, K Simonka, Zs Kahan, Zs AF Rusz, Orsolya Voros, Andras Varga, Zoltan Kelemen, Gyongyi Uhercsak, Gabriella Nikolenyi, Aliz Ormandi, Katalin Simonka, Zsolt Kahan, Zsuzsanna TI One-Year Neoadjuvant Endocrine Therapy in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Hormone sensitivity; Letrozole; Tamoxifen; Neoadjuvant endocrine therapy; Predictive factors ID Breast cancer; Hormone sensitivity; Letrozole; Tamoxifen; Neoadjuvant endocrine therapy; Predictive factors AB The evaluation of the effects of 1-year endocrine therapy (NET) was aimed at. A retrospective analysis of 42 cases with 46 stage II–III invasive, hormone receptor-positive, HER2-negative breast cancers was performed. One-year NET was planned with letrozole (n=33, postmenopausal group), or with goserelin plus letrozole (n=7) or with goserelin plus tamoxifen (n=2) (premenopausal group). Surgery was performed in accordance with the initial stage and the response to therapy. With regard to the tumor remaining in the surgical specimen, risk groups were constructed: Group 1: stage 0, pathological complete regression (pCR); Group 2: stages IAIIA; Group 3: stages ≥IIB + cases with clinical progression. Due to local progression, NET was replaced by neoadjuvant chemotherapy in three patients (four tumors). In two postmenopausal patients, letrozole was replaced by tamoxifen because of the insufficient treatment effect. In 19/42 cases, breastconserving surgery was performed. Within Group 1, there was no cancer in four cases, while only DCIS remained in 2 (pCR: 13 %); Groups 2 and 3 comprised 25 and 15 cases, respectively. The likeliness of a good response (Groups 1 and 2 vs. Group 3) to NET was increased by 7 % for every 1 % increase of the expression of ER (OR=1.070; 95 % CI: 1.007–1.138, p=0.029). Progression-free survival differed according to treatment response (p=0.001). The post-therapy Ki67 value of ≤15 % had only a marginal effect on survival. No other associations were detected between the tumor characteristics and the therapeutic response or survival. Longduration NET is effective and safe in cases of hormonesensitive breast cancer. C1 [Rusz, Orsolya] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Voros, Andras] University of Szeged, Department of PathologySzeged, Hungary. [Varga, Zoltan] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Kelemen, Gyongyi] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Uhercsak, Gabriella] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Nikolenyi, Aliz] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Ormandi, Katalin] University of Szeged, Department of RadiologySzeged, Hungary. [Simonka, Zsolt] University of Szeged, Department of SurgerySzeged, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. RP Kahan, Zs (reprint author), University of Szeged, Department of Oncotherapy, 6720 Szeged, Hungary. EM kahan.zsuzsanna@med.u-szeged.hu CR Kaufmann M, von Minckwitz G, Mamounas EP, Cameron D, Carey LA, Cristofanilli M, Denkert C, Eiermann W, Gnant M, Harris JR, Karn T, Liedtke C, Mauri D, Rouzier R, Ruckhaeberle E, Semiglazov V, Symmans WF, Tutt A, Pusztai L, 2012, Recommendations from an international consensus conference on the current status and future of neoadjuvant systemic therapy in primary breast cancer. Ann Surg Oncol 19:1508–1516 Fumagalli D, Bedard PL, Nahleh Z, Michiels S, Sotiriou C, Loi S, Sparano JA, Ellis M, Hylton N, Zujewski JA, Hudis C, Esserman L, Piccart M, BIG-NABCG collaboration, 2012, A common language in neoadjuvant breast cancer clinical trials: proposals for standard definitions and endpoints. Lancet Oncol 13:e240–e248 Hoffman KE, Mittendorf EA, Buchholz TA, 2012, Optimising radiation treatment decisions for patients who receive neoadjuvant chemotherapy and mastectomy. Lancet Oncol 13:e270–e276 von Minckwitz G, Blohmer JU, Costa SD, Denkert C, Eidtmann H, EiermannW, Gerber B, Hanusch C, Hilfrich J, Huober J, Jackisch C, Kaufmann M, Kummel S, Paepke S, Schneeweiss A, Untch M, Zahm DM, Mehta K, Loibl S, 2013, Response-guided neoadjuvant chemotherapy for breast cancer. J Clin Oncol 31:3623–3630 Prowell TM, Pazdur R, 2012, Pathological complete response and accelerated drug approval in early breast cancer. N Engl J Med 366: 2438–2441 Cortazar P, Zhang L, Untch M,Mehta K, Costantino JP,Wolmark N, Bonnefoi H, Cameron D, Gianni L, Valagussa P, Swain SM, Prowell T, Loibl S, Wickerham DL, Bogaerts J, Baselga J, Perou C, Blumenthal G, Blohmer J, Mamounas EP, Bergh J, Semiglazov V, Justice R, Eidtmann H, Paik S, Piccart M, Sridhara R, Fasching PA, Slaets L, Tang S, Gerber B, Geyer CE Jr, Pazdur R, Ditsch N, Rastogi P, Eiermann W, von Minckwitz G, 2014, Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet., DOI 10.1016/S0140-6736(13)62422-8 Geisler J, Smith I, Miller W, 2012, Presurgical, neoadjuvant, endocrine therapy is a useful model to predict response and outcome to endocrine treatment in breast cancer patients. J Steroid Biochem Mol Biol 131:93–100 Goldhirsch A,Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, Senn HJ, Panel members, 2013, Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol 24:2206–2223 Charehbili A, Fontein DB, Kroep JR, Liefers GJ, Mieog JS, Nortier JW, van de Velde CJ, 2014, Neoadjuvant hormonal therapy for endocrine sensitive breast cancer: a systematic review. Cancer Treat Rev 40:86–92 Chia YH, EllisMJ, Ma CX, 2010, Neoadjuvant endocrine therapy in primary breast cancer: indications and use as a research tool. Br J Cancer 103:759–764 Barnadas A, Gil M, Gonzalez S, Tusquets I, Munoz M, Arcusa A, Prieto L, Margeli-Vila M, Moreno A, 2009, Exemestane as primary treatment of oestrogen receptor-positive breast cancer in postmenopausal women: a phase II trial. Br J Cancer 100:442–449 Toi M, Saji S, Masuda N, Kuroi K, Sato N, Takei H, Yamamoto Y, Ohno S, Yamashita H, Hisamatsu K, Aogi K, Iwata H, Takada M, Ueno T, Saji S, Chanplakorn N, Suzuki T, Sasano H, 2011, Ki67 index changes, pathological response and clinical benefits in primary breast cancer patients treated with 24 weeks of aromatase inhibition. Cancer Sci 102:858–865 Dixon JM, Renshaw L, Macaskill EJ, Young O, Murray J, Cameron D, Kerr GR, Evans DB, Miller WR, 2009, Increase in response rate by prolonged treatment with neoadjuvant letrozole. Breast Cancer Res Treat 113:145–151 Allevi G, Strina C, Andreis D, Zanoni V, Bazzola L, Bonardi S, Foroni C, Milani M, Cappelletti MR, Gussago F, Aguggini S, Giardini R, Martinotti M, Fox SB, Harris AL, Bottini A, Berruti A, Generali D, 2013, Increased pathological complete response rate after a long-term neoadjuvant letrozole treatment in postmenopausal oestrogen and/or progesterone receptor-positive breast cancer. Br J Cancer 108:1587–1592 Hojo T, Kinoshita T, Imoto S, Shimizu C, Isaka H, Ito H, Imi K, Wada N, Ando M, Fujiwara Y, 2013, Use of the neo-adjuvant exemestane in post-menopausal estrogen receptor-positive breast cancer: a randomized phase II trial, PTEX46, to investigate the optimal duration of preoperative endocrine therapy. Breast 22:263–267 Montagna E, Cancello G, Colleoni M, 2013, The aromatase inhibitors, plus ovarian function suppression, in premenopausal breast cancer patients: ready for prime time? Cancer Treat Rev 39:886–890 Torrisi R, Bagnardi V, Rotmensz N, Scarano E, IorfidaM, Veronesi P, Luini A, Viale G, Santoro A, Colleoni M, Goldhirsch A, 2011, Letrozole plus GnRH analogue as preoperative and adjuvant therapy in premenopausal women with ER positive locally advanced breast cancer. Breast Cancer Res Treat 126:431–441 Miller AB, Hoogstraten B, Staquet M, Winkler A, 1981, Reporting results of cancer treatment. Cancer 47:207–214 Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, McShane LM, Paik S, Pegram MD, Perez EA, Press MF, Rhodes A, Sturgeon C, Taube SE, Tubbs R, Vance GH, van de Vijver M, Wheeler TM, Hayes DF, American Society of Clinical Oncology, College of American Pathologists, 2007, American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol 25:118–145 Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, Fitzgibbons PL, Francis G, Goldstein NS, Hayes M, Hicks DG, Lester S, Love R, Mangu PB, McShane L, Miller K, Osborne CK, Paik S, Perlmutter J, Rhodes A, Sasano H, Schwartz JN, Sweep FC, Taube S, Torlakovic EE, Valenstein P, Viale G, Visscher D, Wheeler T, Williams RB, Wittliff JL, Wolff AC, American Society of Clinical Oncology, College of American Pathologists, 2010, American Society of Clinical Oncology/ College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. Arch Pathol Lab Med 134:e48–e72 Dowsett M, Nielsen TO, A’Hern R, Bartlett J, Coombes RC, Cuzick J, Ellis M, Henry NL, Hugh JC, Lively T, McShane L, Paik S, Penault-Llorca F, Prudkin L, Regan M, Salter J, Sotiriou C, Smith IE, Viale G, Zujewski JA, Hayes DF, International Ki-67 in Breast CancerWorking Group, 2011, Assessment of Ki67 in breast cancer: recommendations from the International Ki67 in Breast Cancer working group. J Natl Cancer Inst 103:1656–1664 Ellis MJ, Tao Y, Luo J, A’Hern R, Evans DB, Bhatnagar AS, Chaudri Ross HA, von Kameke A,MillerWR, Smith I, EiermannW, Dowsett M, 2008, Outcome prediction for estrogen receptor-positive breast cancer based on postneoadjuvant endocrine therapy tumor characteristics. J Natl Cancer Inst 100:1380–1388 Smith IE, Dowsett M, Ebbs SR, Dixon JM, Skene A, Blohmer JU, Ashley SE, Francis S, Boeddinghaus I, Walsh G, IMPACT Trialists Group, 2005, Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen, or both in combination: the Immediate Preoperative Anastrozole, Tamoxifen, or Combined with Tamoxifen, IMPACT, multicenter double-blind randomized trial. J Clin Oncol 23:5108–5116 DeCensi A, Guerrieri-Gonzaga A, Gandini S, Serrano D, Cazzaniga M, Mora S, Johansson H, Lien EA, Pruneri G, Viale G, Bonanni B, 2011, Prognostic significance of Ki-67 labeling index after shortterm presurgical tamoxifen in women with ER-positive breast cancer. Ann Oncol 22:582–587 Ellis MJ, Coop A, Singh B, Mauriac L, Llombert-Cussac A, Janicke F, Miller WR, Evans DB, Dugan M, Brady C, Quebe-Fehling E, Borgs M, 2001, Letrozole is more effective neoadjuvant endocrine therapy than tamoxifen for ErbB-1- and/or ErbB-2-positive, estrogen receptor-positive primary breast cancer: evidence from a phase III randomized trial. J Clin Oncol 19:3808–3816 Dowsett M, Smith IE, Ebbs SR, Dixon JM, Skene A, Griffith C, Boeddinghaus I, Salter J, Detre S, Hills M, Ashley S, Francis S, Walsh G, A’Hern R, 2008, Proliferation and apoptosis as markers of benefit in neoadjuvant endocrine therapy of breast cancer. Clin Cancer Res 12:1024s–1030s Mello-Grand M, Singh V, Ghimenti C, Scatolini M, Regolo L, Grosso E, Zambelli A, Da Prada GA, Villani L, Fregoni V, Baiardi P, Marsoni S, Miller WR, Costa A, Chiorino G, 2010, Gene expression profiling and prediction of response to hormonal neoadjuvant treatment with anastrozole in surgically resectable breast cancer. Breast Cancer Res Treat 121:399–411 Ueno T, Masuda N, Yamanaka T, Saji S, Kuroi K, Sato N, Takei H, Yamamoto Y, Ohno S, Yamashita H, Hisamatsu K, Aogi K, Iwata H, Sasano H, Toi M, 2014, Evaluating the 21-gene assay Recurrence Score® as a predictor of clinical response to 24 weeks of neoadjuvant exemestane in estrogen receptor-positive breast cancer. Int J Clin Oncol 19:607–613 Krainick-Strobel UE, Lichtenegger W, Wallwiener D, Tulusan AH, Janicke F, Bastert G, Kiesel L, Wackwitz B, Paepke S, 2008, Neoadjuvant letrozole in postmenopausal estrogen and/or progesterone receptor positive breast cancer: a phase IIb/III trial to investigate optimal duration of preoperative endocrine therapy. BMC Cancer 8: 62 Carpenter R, Doughty JC, Cordiner C, Moss N, Gandhi A,Wilson C, Andrews C, Ellis G, Gui G, Skene AI, 2014, Optimum duration of neoadjuvant letrozole to permit breast conserving surgery. Breast Cancer Res Treat 144:569–576 Alba E, Calvo L, Albanell J, De la Haba JR, Arcusa Lanza A, Chacon JI, Sanchez-Rovira P, Plazaola A, Lopez Garcia- Asenjo JA, Bermejo B, Carrasco E, Lluch A, 2012, GEIC AM: Chemotherapy, CT, and hormonotherapy, HT, as neoadjuvant treatment in luminal breast cancer patients: results from the GEICAM/2006-03, a multicenter, randomized, phase-II study. Ann Oncol 23:3069–3074 Gnant M, Mlineritsch B, Stoeger H, Luschin-Ebengreuth G, Heck D, Menzel C, Jakesz R, Seifert M, Hubalek M, Pristauz G, Bauernhofer T, Eidtmann H, Eiermann W, Steger G, Kwasny W, Dubsky P, Hochreiner G, Forsthuber EP, Fesl C, Greil R, Austrian Breast and Colorectal Cancer Study Group, 2011, Adjuvant endocrine therapy plus zoledronic acid in premenopausal women with early-stage breast cancer: 62-month follow-up from the ABCSG-12 randomised trial. Lancet Oncol 12:631–641 Pagani O, ReganMM,Walley BA, Fleming GF, Colleoni M, Lang I, Gomez HL, Tondini C, Burstein HJ, Perez EA, Ciruelos E, Stearns V, Bonnefoi HR,Martino S, Geyer CE Jr, Pinotti G, Puglisi F, Crivellari D, Ruhstaller T,Winer EP, Rabaglio-Poretti M, Maibach R, Ruepp B, Giobbie-Hurder A, Price KN, Bernhard J, Luo W, Ribi K, Viale G, Coates AS, Gelber RD, Goldhirsch A, Francis PA, TEXT and SOFT Investigators, International Breast Cancer Study Group: the TEXT and SOFT Investigators and the International Breast Cancer Study Group, 2014, Adjuvant exemestane with ovarian suppression in premenopausal breast cancer. N Engl J Med 371:107–118 Debled M, Auxepaules G, de Lara CT, Garbay D, Brouste V, Bussieres E, Mauriac L, Macgrogan G, 2014, Neoadjuvant endocrine treatment in breast cancer: analysis of daily practice in large cancer center to facilitate decision making. AmJ Surg., DOI 10.1016/j. amjsurg.2013.12.032 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 977 EP 984 DI 10.1007/s12253-015-9911-1 PG 8 ER PT J AU Kravchick, S Lobik, L Cytron, Sh Kravchenko, Y Dor, BD Peled, R AF Kravchick, Sergey Lobik, Leonid Cytron, Shmuel Kravchenko, Yakov Dor, Ben David Peled, Ronit TI Patients with Persistently Elevated PSA and Negative Results of TRUS-Biopsy: Does 6-Month Treatment with Dutasteride can Indicate Candidates for Re-Biopsy. What is the Best of Saturation Schemes: Transrectal or Transperineal Approach? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Biopsy; Elevated PSA; Dutasteride ID Prostate cancer; Biopsy; Elevated PSA; Dutasteride AB To identify patients who actually need a re - biopsy, based on alterations in PSA readings after 6-month treatment with Dutasteride. We also sought to bring out the most beneficial re-biopsy scheme. We have reviewed the records of patients with persistently elevated PSA and at least one set of TRUS biopsies. Patients who were treated with alpha - blockers/Dutasteride combination were considered as the study group, while patients in control received alphablockers alone. Patients in both groups underwent re-biopsy 6 months later. The two protocols of re-biopsies were used at that time: 20-24 cores saturation transrectal (ST)) and ≥40 cores saturation transperineal template-guided (STT) biopsies. One hundred thirty-three patients were included in this study. In 86.7 % of the patients in the study group mean PSA decreased from 7.4±2.69 to 4.037±1.53 (p-0.001). The overall cancer detection rate was 29 % (n-39: 19 v/s 20, control and study groups, respectively). In the study group PSA decreased to 26.73±11.26 % in patients with cancer, compared with 40.54±13.3 % in patients without. It must be emphasized that STT-biopsies detected significantly more cancers (38.46 v/s 20.59 %, p- 0.005). Mean cores number got to 21±2.45 and 45±5.65 in ST and STT biopsies, respectively. Six-month treatment with Dutasteride decreases PSA readings in 86.7 % of the patients. A PSA decline of less than 40% (cutoff) should be considered as an indicator for re-biopsy. Transperineal template-guided biopsies had a higher cancer detection rate. C1 [Kravchick, Sergey] Barzialy Medical Center, Urology DepartmentAshkelon, Israel. [Lobik, Leonid] Barzialy Medical Center, Urology DepartmentAshkelon, Israel. [Cytron, Shmuel] Barzialy Medical Center, Urology DepartmentAshkelon, Israel. [Kravchenko, Yakov] Barzialy Medical Center, Pathology DepartmentAshkelon, Israel. [Dor, Ben David] Barzialy Medical Center, Pathology DepartmentAshkelon, Israel. [Peled, Ronit] Ben Gurion University, Epidemiology and StatisticsBeer-Sheva, Israel. RP Kravchick, S (reprint author), Barzialy Medical Center, Urology Department, Ashkelon, Israel. EM kravchick.sergey@gmail.com CR Presti JC Jr, 2009, Repeat prostate biopsy - when, where, and how. Urol Oncol 27:312–314 Theoret MR, Ning Y-M, Zhang JJ et al, 2011, The risk and benefits of 5α-Reductase inhibitors for prostate-cancer prevention. N Engl J Med 365(2):97–99 Kaplan SA, Ghafar MA, Volpe MA et al, 2002, PSA response to finasteride challenge in men with a serum PSA greater than 4 ng/ml and previous negative prostate biopsy: preliminary study. Urology 60(3):464–468 Kaplan SA, Lee RK, Chung DE et al, 2012, Prostate biopsy in response to a change in nadir prostate specific antigen of 0.4 ng/ml after treatment with 5α-reductase inhibitors markedly enhances the detection rate of prostate cancer. J Urol 188(3):757–761 Handel LN, Agarwal S, Schiff SF et al, 2006, Can effect of finasteride on prostate-specific antigen be used to decrease repeat prostate biopsy? Urology 68(6):1220–1223 Ekwueme K, Simpson H, Zakhour H et al, 2013, Transperineal template-guided saturation biopsy using a modified technique: outcome of 270 cases requiring repeat prostate biopsy. BJU Int 111(8): 365–373 Simon J, Kuefer R, Bartsch G Jr et al, 2008, Intensifying the saturation biopsy technique for detecting prostate cancer after previous negative biopsies: a step in the wrong direction. BJU Int 102(4): 459–462 Ukimura O, Coleman JA, de la Taille A et al, 2013, Contemporary role of systematic prostate biopsies: indications, techniques, and implications for patient care. Eur Urol 63(2):214–230 ZaytounOM,Moussa AS, Gao Tet al, 2011, Office based transrectal saturation biopsy improves prostate cancer detection compared to extended biopsy in the repeat biopsy population. J Urol 186(3): 850–854 Abdollah F, Novara G, Briganti A et al, 2011, Trans-rectal versus trans-perineal saturation rebiopsy of the prostate: is there a difference in cancer detection rate? Urology 77(4):921–925 D'Amico AV,Whittington R, Malkowicz SB et al, 1995, A multivariate analysis of clinical and pathological factors that predict for prostate specific antigen failure after radical prostatectomy for prostate cancer. J Urol 154:131–138 Epstein JI, 2011, Prognostic significance of tumor volume in radical prostatectomy and needle biopsy specimens. J Urol 186:790–797 Kranse R, Roobol M, Schro¨der FH, 2008, A graphical device to represent the outcomes of a logistic regression analysis. Prostate 68: 1674–1680 Steyerberg EW, Roobol MJ, Kattan MW et al, 2007, Prediction of indolent prostate cancer: validation and updating of a prognostic nomogram. J Urol 177:107–112 RoobolMJ, Zhu X, Schroder FH et al, 2013, Acalculator for prostate cancer risk 4 years after an initially negative screen: findings from ERSPC rotterdam. Eur Urol 63(4):627–633 Thompson IM, Ankerst DP, Chi C et al, 2005, Operating characteristics of prostate-specific antigen in men with an initial PSA level of 3.0 ng/ml or lower. JAMA 294:66–70 Andriole GL, Bostwick D, Brawley OW, REDUCE Study Group et al, 2011, The effect of dutasteride on the usefulness of prostate specific antigen for the diagnosis of high grade and clinically relevant prostate cancer in men with a previous negative biopsy: results from the REDUCE study. J Urol 185(1):126–131 Roehrborn CG, Andriole GL, Wilson TH et al, 2011, Effect of dutasteride on prostate biopsy rates and the diagnosis of prostate cancer in men with lower urinary tract symptoms and enlarged prostates in the combination of avodart and tamsulosin trial. Eur Urol 59(2):244–249 Buschemeyer WC 3rd, Freedland SJ, 2007, Obesity and prostate cancer: epidemiology and clinical implications. Eur Urol 52(2): 331–343 Allott EH, Masko EM, Freedland SJ, 2013, Obesity and prostate cancer: weighing the evidence. Eur Urol 63(5):800–809 Andriole GL, Bostwick DG, Brawley OW et al, 2010, Effect of dutasteride on the risk of prostate cancer. N Engl J Med 362:1192– 1202 Thompson IM, Goodman PJ, Tangen CM et al, 2003, The influence of finasteride on the development of prostate cancer. N Engl J Med 349(3):215–224 Evans HC, Goa KL, 2003, Dutasteride. Drugs Aging 20:905–916 Rosario DJ, Lane JA, Metcalfe C et al, 2012, Short term outcomes of prostate biopsy in men tested for cancer by prostate specific antigen: prospective evaluation within ProtecT study. BMJ 344:d7894 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 985 EP 989 DI 10.1007/s12253-015-9910-2 PG 5 ER PT J AU Josa, V Krzystanek, M Vass, T Lang, T Juhasz, V Szilagyi, K Tihanyi, B Harsanyi, L Szallasi, Z Salamon, F Baranyai, Zs AF Josa, Valeria Krzystanek, Marcin Vass, Tamas Lang, Tamas Juhasz, Viktoria Szilagyi, Kamilla Tihanyi, Balazs Harsanyi, Laszlo Szallasi, Zoltan Salamon, Ferenc Baranyai, Zsolt TI Thrombocytosis of Liver Metastasis from Colorectal Cancer as Predictive Factor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thrombocytosis; Liver metastasis of colorectal cancer; Prognostic factor; Survival ID Thrombocytosis; Liver metastasis of colorectal cancer; Prognostic factor; Survival AB There is increasing evidence that thrombocytosis is associated with tumor invasion and metastasis formation. It was shown in several solid tumor types that thrombocytosis prognosticates cancer progression. The aim of this study was to evaluate preoperative thrombocytosis as a potential prognostic biomarker in isolated metastases, in patients with liver metastasis of colorectal cancer (mCRC). Clinicopathological data of 166 patients with mCRC who had surgical resection between 2001 and 2011 were collected retrospectively. All primary tumors have been already resected. The platelet count was evaluated based on the standard preoperative blood profile. The patients were followed-up on average for 28 months. Overall survival (OS) of patients with thrombocytosis was significantly worse both in univariate (HR=3.00, p=0.03) and in multivariate analysis (HR=4.68, p=0.056) when adjusted for gender, age, tumor size and surgical margin. Thrombocytosis was also a good prognosticator of diseasefree survival (DFS) with HR=2.7, p=0.018 and nearly significant in multivariate setting (HR=2.26, p=0.073). The platelet count is a valuable prognostic marker for the survival in patients with mCRC. C1 [Josa, Valeria] Tumorgenetika Human Biospecimen Collection and Research, Kerekgyarto u. 36, 1147 Budapest, Hungary. [Krzystanek, Marcin] Technical University of Denmark, Department of Systems Biology, Kemitorvet Building 208, 2800 Kgs. Lyngby, Denmark. [Vass, Tamas] Semmelweis University, 1st Department of Surgery, Ulloi ut 78, 1082 Budapest, Hungary. [Lang, Tamas] Szent Istvan Hospital, Department of General Surgery, Nagyvarad ter 1, 1097 Budapest, Hungary. [Juhasz, Viktoria] Bajcsy Hospital, Department of Surgery, Maglodi ut 89-91, 1106 Budapest, Hungary. [Szilagyi, Kamilla] HDF Medical Centre, Department of Otorhinolaryngology and Head and Neck Surgery, Podmaniczky utca 109-111, 1062 Budapest, Hungary. [Tihanyi, Balazs] Semmelweis University, 1st Department of Surgery, Ulloi ut 78, 1082 Budapest, Hungary. [Harsanyi, Laszlo] Semmelweis University, 1st Department of Surgery, Ulloi ut 78, 1082 Budapest, Hungary. [Szallasi, Zoltan] Technical University of Denmark, Department of Systems Biology, Kemitorvet Building 208, 2800 Kgs. Lyngby, Denmark. [Salamon, Ferenc] Fovarosi Uzsoki utcai Korhaz, Pathologia, Uzsoki u. 29-41, 1145 Budapest, Hungary. [Baranyai, Zsolt] Semmelweis University, 1st Department of Surgery, Ulloi ut 78, 1082 Budapest, Hungary. RP Baranyai, Zs (reprint author), Semmelweis University, 1st Department of Surgery, 1082 Budapest, Hungary. EM barazso@gmail.com CR Kanavos P, Schurer W, 2010, The dynamics of colorectal cancer management in 17 countries. Eur J Health Econ 10(Suppl 1):S115– S129 McArdle CS, Hole DJ, 2002, Outcome following surgery for colorectal cancer: analysis by hospital after adjustment for case-mix and deprivation. Br J Cancer 86:331–335 Riess L, 1872, Zur pathologischen Anatomie des Blutes. Arch Anat Physiol Wissensch Med 39:237–249 Hernandez E, LavineM, Dunton CJ, Gracely E, Parker J, 1992, Poor prognosis associated with thrombocytosis in patients with cervical cancer. Cancer 69:2975–2977 Pedersen LM, Milman N, 1996, Prognostic significance of thrombocytosis in patients with primary lung cancer. Eur Respir J 9:1826–1830 Taucher S, Salat A, GnantM, KwasnyW,Mlineritsch B,Menzel RC, Schmid M, Smola MG, Stierer M, Tausch C, Galid A, Steger G, Jakesz R, 2003, Impact of pretreatment thrombocytosis on survival in primary breast cancer. Thromb Haemost 89:1098–1106 Wosnitzer M, Polland A, Hai Q, Hruby G, McKiernan J, 2011, Role of preoperative platelet level in clinical and pathological outcomes after surgery for renal cortical malignancies. BJU Int 108:73–79 Feng JF, Huang Y, Lu WS, Chen QX, 2013, Preoperative platelet count in esophageal squamous cell carcinoma: is it a prognostic factor? Langenbecks Arch Surg 398(8):1115–22 Heras P, Hatzopoulos A, Kritikos N, Kritikos K, 2010, Platelet count and tumor progression in gastric cancer patients. Scand J Gastroenterol 45:1005–1006 Brown KM, Domin C, Aranha GV, Yong S, Shoup M, 2005, Increased preoperative platelet count is associated with decreased survival after resection for adenocarcinoma of the pancreas. Am J Surg 189:278–282 Buergy D, Wenz F, Groden C, Brockmann MA, 2012, Tumorplatelet interaction in solid tumors. Int J Cancer 130:2747–2760 Gay LJ, Felding-Habermann B, 2011, Contribution of platelets to tumour metastasis. Nat Rev Cancer 11:123–134 Nieswandt B, Hafner M, Echtenacher B, Mannel DN, 1999, Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res 59:1295–1300 Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, Kombrinck KW, Jirouskova M, Degen JL, 2005, Platelets and fibrin( ogen, increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood 105:178–185 Wang D, Huang HJ, Kazlauskas A, CaveneeWK(1999, Induction of vascular endothelial growth factor expression in endothelial cells by platelet-derived growth factor through the activation of phosphatidylinositol 3-kinase. Cancer Res 59:1464–1472 Ho-Tin-Noe B, Goerge T, Wagner DD, 2009, Platelets: guardians of tumor vasculature. Cancer Res 69:5623–5626 Stone RL, Nick AM, McNeish IA, Balkwill F, Han HD, Bottsford- Miller J, Rupairmoole R, Armaiz-Pena GN, Pecot CV, Coward J, Deavers MT, Vasquez HG, Urbauer D, Landen CN, Hu W, Gershenson H, Matsuo K, Shahzad MM, King ER, Tekedereli I, Ozpolat B, Ahn EH, Bond VK, Wang R, Drew AF, Gushiken F, Lamkin D, Collins K, DeGeest K, Lutgendorf SK, Chiu W, Lopez- Berestein G, Afshar-Kharghan V, Sood AK, 2012, Paraneoplastic thrombocytosis in ovarian cancer. N Engl J Med 366:610–618 Qiu MZ, Yuan ZY, Luo HY, Ruan DY,Wang ZQ,Wang FH, Li YH, Xu RH, 2010, Impact of pretreatment hematologic profile on survival of colorectal cancer patients. Tumour Biol 31:255–260 Sasaki K, Kawai K, Tsuno NH, Sunami E, Kitayama J, 2012, Impact of preoperative thrombocytosis on the survival of patients with primary colorectal cancer. World J Surg 36:192–200 Lin MS, Huang JX, Zhu J, Shen HZ, 2012, Elevation of platelet count in patients with colorectal cancer predicts tendency to metastases and poor prognosis. Hepatogastroenterology 59:1687–1690 Baranyai Z, Krzystanek M, Josa V, Dede K, Agoston E, Szasz AM, Sinko D, Szarvas V, Salamon F, Eklund AC, Szallasi Z, Jakab F, 2014, The comparison of thrombocytosis and platelet-lymphocyte ratio as potential prognostic markers in colorectal cancer. Thromb Haemost 111:483–490 Griesshammer M, Bangerter M, Sauer T, Wennauer R, Bergmann L, Heimpel H, 1999, Aetiology and clinical significance of thrombocytosis: analysis of 732 patients with an elevated platelet count. J Intern Med 245:295–300 Buss DH, Cashell AW, O’Connor ML, Richards F 2nd, Case LD, 1994, Occurrence, etiology, and clinical significance of extreme thrombocytosis: a study of 280 cases. Am J Med 96:247–253 Schafer AI, 2004, Thrombocytosis. N Engl J Med 350:1211–1219 Kulnigg-Dabsch S, Schmid W, Howaldt S, Stein J, Mickisch O, Waldhor T, Evstatiev R, Kamali H, Volf I, Gasche C, 2013, Iron deficiency generates secondary thrombocytosis and platelet activation in IBD: the randomized, controlled thromboVIT trial. Inflamm Bowel Dis 19:1609–1616 Choi SI, Simone JV, 1973, Platelet production in experimental iron deficiency anemia. Blood 42:219–228 Choi SI, Simone JV, Jackson CW, 1974, Megakaryocytopoiesis in experimental iron deficiency anemia. Blood 43:111–120 Simanek R, Vormittag R, Ay C, Alguel G, Dunkler D, Schwarzinger I, Steger G, Jaeger U, Zielinski C, Pabinger I, 2010, High platelet count associated with venous thromboembolism in cancer patients: results from the Vienna Cancer and Thrombosis Study, CATS). J Thromb Haemost 8:114–120 Khorana AA, Kuderer NM, Culakova E, Lyman GH, Francis CW, 2008, Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood 111:4902–4907 Evstatiev R, Bukaty A, Jimenez K, Kulnigg-Dabsch S, Surman L, Schmid W, Eferl R, Lippert K, Scheiber-Mojdehkar B, Michael Kvasnicka H, Khare V, Gasche C, 2014, Iron deficiency alters megakaryopoiesis and platelet phenotype independent of thrombopoietin. Am J Hematol 89:524–529 Erpenbeck L, Schon MP, 2010, Deadly allies: the fatal interplay between platelets and metastasizing cancer cells. Blood 115:3427– 3436 Wang D, Huang HJ, Kazlauskas A, CaveneeWK(1999, Induction of vascular endothelial growth factor expression in endothelial cells by platelet-derived growth factor through the activation of phosphatidylinositol 3-kinase. Cancer Res 59:1464–1472 Ho-Tin-Noe B, Goerge T, Wagner DD, 2009, Platelets: guardians of tumor vasculature. Cancer Res 69:5623–5626 Trikha M, Zhou Z, Timar J, Raso E, Kennel M, Emmell E, Nakada MT, 2002, Multiple roles for platelet GPIIb/IIIa and alphavbeta3 integrins in tumor growth, angiogenesis, and metastasis. Cancer Res 62:2824–2833 Stone RL, Nick AM, McNeish IA, Balkwill F, Han HD, Bottsford- Miller J, Rupairmoole R, Armaiz-Pena GN, Pecot CV, Coward J, Deavers MT, Vasquez HG, Urbauer D, Landen CN, Hu W, Gershenson H, Matsuo K, Shahzad MM, King ER, Tekedereli I, Ozpolat B, Ahn EH, Bond VK, Wang R, Drew AF, Gushiken F, Lamkin D, Collins K, DeGeest K, Lutgendorf SK, Chiu W, Lopez- Berestein G, Afshar-Kharghan V, Sood AK, 2012, Paraneoplastic thrombocytosis in ovarian cancer. N Engl J Med 366:610–618 De Vita F, Romano C, Orditura M, Galizia G, Martinelli E, Lieto E, Catalano G, 2001, Interleukin-6 serum level correlates with survival in advanced gastrointestinal cancer patients but is not an independent prognostic indicator. J Interferon Cytokine Res 21:45–52 Paule B, Belot J, Rudant C, Coulombel C, Abbou CC, 2000, The importance of IL-6 protein expression in primary human renal cell carcinoma: an immunohistochemical study. J Clin Pathol 53:388– 390 Nakashima J, Tachibana M, Horiguchi Y, Oya M, Ohigashi T, Asakura H, Murai M, 2000, Serum interleukin 6 as a prognostic factor in patients with prostate cancer. Clin Cancer Res 6:2702–2706 Plante M, Rubin SC,Wong GY, Federici MG, Finstad CL, Gastl GA, 1994, Interleukin-6 level in serum and ascites as a prognostic factor in patients with epithelial ovarian cancer. Cancer 73:1882–1888 Takeuchi E, Ito M, Mori M, Yamaguchi T, Nakagawa M, Yokota S, Nishikawa H, Sakuma-Mochizuki J, Hayashi S, Ogura T, 1996, Lung cancer producing interleukin-6. Intern Med 35:212–214 Corbeil J, Evans LA, Vasak E, Cooper DA, Penny R, 1991, Culture and properties of cells derived from Kaposi sarcoma. J Immunol 146: 2972–2976 Chen Q, Solar G, Eaton DL, de Sauvage FJ, 1998, IL-3 does not contribute to platelet production in c-Mpl-deficient mice. Stem Cells 16(Suppl 2):31–36 Gainsford T, Roberts AW, Kimura S, Metcalf D, Dranoff G, Mulligan RC, Begley CG, Robb L, Alexander WS(1998, Cytokine production and function in c-mpl-deficient mice: no physiologic role for interleukin- 3 in residual megakaryocyte and platelet production. Blood 91:2745–2752 Kaser A, Brandacher G, Steurer W, Kaser S, Offner FA, Zoller H, Theurl I,WidderW, Molnar C, Ludwiczek O, Atkins MB, Mier JW, Tilg H, 2001, Interleukin-6 stimulates thrombopoiesis through thrombopoietin: role in inflammatory thrombocytosis. Blood 98: 2720–2725 D’Hondt V, Humblet Y, Guillaume T, Baatout S, Chatelain C, Berliere M, Longueville J, Feyens AM, de Greve J, Van Oosterom A et al, 1995, Thrombopoietic effects and toxicity of interleukin-6 in patients with ovarian cancer before and after chemotherapy: a multicentric placebo-controlled, randomized phase Ib study. Blood 85:2347–2353 van Gameren MM, Willemse PH, Mulder NH, Limburg PC, Groen HJ, Vellenga E, de Vries EG, 1994, Effects of recombinant human interleukin-6 in cancer patients: a phase I-II study. Blood 84:1434– 1441 Griesshammer M, Bangerter M, Sauer T, Wennauer R, Bergmann L, Heimpel H, 1999, Aetiology and clinical significance of thrombocytosis: analysis of 732 patients with an elevated platelet count. J Intern Med 245:295–300 Buss DH, Cashell AW, O’Connor ML, Richards F 2nd, Case LD, 1994, Occurrence, etiology, and clinical significance of extreme thrombocytosis: a study of 280 cases. Am J Med 96:247–253 Kulnigg-Dabsch S, Schmid W, Howaldt S, Stein J, Mickisch O, Waldhor T, Evstatiev R, Kamali H, Volf I, Gasche C, 2013, Iron deficiency generates secondary thrombocytosis and platelet activation in IBD: the randomized, controlled thromboVIT trial. Inflamm Bowel Dis 19:1609–1616 Choi SI, Simone JV, 1973, Platelet production in experimental iron deficiency anemia. Blood 42:219–228 Choi SI, Simone JV, Jackson CW, 1974, Megakaryocytopoiesis in experimental iron deficiency anemia. Blood 43:111–120 Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A, 2009, Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 30:1073–1081 Hanahan D,Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Balkwill F, Mantovani A, 2001, Inflammation and cancer: back to Virchow? Lancet 357:539–545 Mantovani A, Allavena P, Sica A, Balkwill F, 2008, Cancer-related inflammation. Nature 454:436–444 Thompson CB, 1995, Apoptosis in the pathogenesis and treatment of disease. Science 267:1456–1462 Vakkila J, Lotze MT, 2004, Inflammation and necrosis promote tumour growth. Nat Rev Immunol 4:641–648 McMillan DC, Crozier JE, Canna K, Angerson WJ, McArdle CS, 2007, Evaluation of an inflammation-based prognostic score, GPS, in patients undergoing resection for colon and rectal cancer. Int J Colorectal Dis 22:881–886 Crumley AB, McMillan DC, McKernan M, Going JJ, Shearer CJ, Stuart RC, 2006, An elevated C-reactive protein concentration, prior to surgery, predicts poor cancer-specific survival in patients undergoing resection for gastro-oesophageal cancer. Br J Cancer 94:1568– 1571 Jamieson NB, Glen P, McMillan DC, McKay CJ, Foulis AK, Carter R, Imrie CW, 2005, Systemic inflammatory response predicts outcome in patients undergoing resection for ductal adenocarcinoma head of pancreas. Br J Cancer 92:21–23 Karakiewicz PI, Hutterer GC, TrinhQD, Jeldres C, Perrotte P, Gallina A, Tostain J, Patard JJ, 2007, C-reactive protein is an informative predictor of renal cell carcinoma-specificmortality: a European study of 313 patients. Cancer 110:1241–1247 Hilmy M, Campbell R, Bartlett JM, McNicol AM, Underwood MA, McMillan DC, 2006, The relationship between the systemic inflammatory response, tumour proliferative activity, T-lymphocytic infiltration and COX-2 expression and survival in patients with transitional cell carcinoma of the urinary bladder. Br J Cancer 95:1234– 1238 Hara M, Matsuzaki Y, Shimuzu T, Tomita M, Ayabe T, Enomoto Y, Onitsuka T, 2007, Preoperative serum C-reactive protein level in non-small cell lung cancer. Anticancer Res 27: 3001–3004 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 991 EP 997 DI 10.1007/s12253-015-9925-8 PG 7 ER PT J AU Wieczorek, A Balwierz, W AF Wieczorek, Aleksandra Balwierz, Walentyna TI The Role of Id2 Protein in Neuroblatoma in Children SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Neuroblastoma; Id2 protein; Prognostic factor ID Neuroblastoma; Id2 protein; Prognostic factor AB Id (DNA binding and/or differentiation) proteins occur physiologically during ontogenesis and negatively regulate the activity of other helix-loop-helix (HLH) proteins. Id2 protein causes block of cells differentiation in the S phase of the cell cycle and regulates the activity of Rb protein. The role of Id2 protein in physiological cell cycle progression and in neuroblastoma (NBL) pathogenesis was proposed by Lasorella. The aim of the study was evaluation of Id2 expression and its prognostic significance in NBL cells coming from primary tumors and evaluation of its prognostic significance, and correlation of Id2 expression with known prognostic factors. Sixty patients with primary NBL treated from 1991 to 2005 were included in the analysis.We found 50 patients with high and 10 patients with low intensity of Id2 expression. The median percentage of NBL cells with Id2 expression was 88 %. We found no correlation between the number of NBL cells or the intensity of Id2 expression and OS and DFS. In patients with stage 4 NBL, almost all patients had high expression of Id2 and it was significantly more common than in other disease stages (p=0,03). We found no correlation between Id2 expression and other known prognostic factor in NBL patients. We assume that Id2 is not prognostic factor. However, due to its abundant expression in most of NBL cells and its role in cell cycle, it may be potential therapeutic target. Exact knowledge of expression time may be helpful in explaining mechanisms of oncogenesis. C1 [Wieczorek, Aleksandra] Jagiellonian University Medical College, Polish-American Institute of Pediatrics, Department of Pediatric Oncology and HematologyKrakow, Poland. [Balwierz, Walentyna] Jagiellonian University Medical College, Polish-American Institute of Pediatrics, Department of Pediatric Oncology and HematologyKrakow, Poland. RP Wieczorek, A (reprint author), Jagiellonian University Medical College, Polish-American Institute of Pediatrics, Department of Pediatric Oncology and Hematology, Krakow, Poland. EM a.wieczorek@uj.edu.pl CR Benezra R, Davis RL, Lockshon D, Turner DL,Weintraub H, 1990, The protein Id: a negative regulator of helix-hoop-helixDNA binding proteins. Cell 61(1):49–59 Iavarone A, Garg A, Lasorella A, Hsu J, Israel M, 1994, The helixloop- helix protein Id2 enhances cell proliferation and binds to retinoblastoma protein. Genes Dev 8:270–284 Lasorella A, Noseda M, Beyna M, Yokota Y, Iavarone A, 2000, Id2 is a retinoblastoma protein target and mediates signaling by Myc oncoproteins. Nature 407:592–598 Liu Y, Encinas M, Comella JX, Aldea M, Gallego C, 2004, Basic helix-loop-helix proteins bind to TrkB and p21cip promoters linking dedifferentiation of hypoxic neuroblastoma cells. J Bioch Chem 24(7):2662–2672 Norton JD, Deed RW, Crags G, Sablitzky F, 1998, Id helix-loophelix proteins in cell growth and differentiation. Trends Cell Biol 8: 58–65 Yokota Y, 2001, Id and development. Oncogene 20:8290–8298 Iavarone A, Lasorella A, 2004, Id2 proteins in neural cancer. Cancer Lett 204:270–284 Lasorella A, Iavarone A, Israel MA, 1996, Id2 specificity alters regulation of the cell cycle by tumor suppressor proteins. Mol Cell Biol 16(6):2570–2578 Riechmann V, Sablitzky F, 1995, Mutually exclusive expression of two dominant-negative helix-loop-helix, dnHLH, genes, Id4 and Id3, in the developing brain of the mouse suggests distinct regulatory roles of these dnHLH proteins during cellular proliferation and differentiation of the nervous system. Cell Growth Differ 6(7):837–843 ZhuW, Dahmen J, Bulfone A, Rigolet M, Hernandez MC, KuoWL, Rubenstein JRL, Israel MA, 1995, Id gene expression during development and molecular cloning of the human Id-1 gene. Mol Brain Res 30(2):312–326 Biggs J, Murphy EV, Israel MA, 1992, A human Id-like helix-loophelix protein expressed during early development. Proc Natl Acad Scie USA 89:1512–1516 Toma JG, El-Bizri H, Barnabe-Heider F, Aloyz R, Miler FD, 2000, Evidence that helix-loop-helix proteins collaborate with retinoblastoma tumor suppressor protein to regulate cortical neurogenesis. J Neurosc 20(20):7648–7656 Lasorella A, Iavarone A, 2006, The protein ENH is a cytoplasmic sequestration factor for Id2 in normal and tumor cells from the nervous system. Proc Natl Acad Sci U S A 103(13):4976–4981 Lasorella A, Uo T, Iavarone A, 2001, Id proteins at the cross-road of development and cancer. Oncogene 20:8326–8333 Lasorella A, Boldrini R, Dominici C, Donfrancesco A, Yokota Y, Inserra A, Iavarone A, 2002, Id2 is critical for cellular proliferation and is the oncogenic effector of N-Myc in human neuroblastoma. Cancer Res 63:301–306 Tzeng SF, 2003, Inhibitors of DNA binding in neural proliferation and differentiation. Neurochem Res 28(10):45–52 Yokota Y,Mori S, Narumi O, Kitajima K, 2001, In vivo function of a differentiation inhibitor Id2. Life 51:207–214 Gleichmann M, Buchheim G, El-Bizri H, Yokota Y, Klockgether T, Kuegler S, Bahr M, Weller M, Schulz JB, 2002, Identification of inhibitor-of-differentiation 2, Id2, as a modular of neuronal apoptosis. J Neurochem 80:755–762 Lee EY, Hu N, Yuan SS, Cox LA, Bradley A, Lee WH, Herrup K, 1994, Dual roles of the retinoblastoma protein in cell cycle regulation and neuron differentiation. Genes Dev 8:2008–2021 Chen XS, Zhang YH, Cai QY, Zyao ZX, 2012, ID2: a negative transcription factor regulating oligodnedroglia differentiation. J Neurosc Res 90:925–932 Al-Haji M, Clarke MFL, 2004, Self-renewal and solid tumor stem cells. Oncogene 24:7273–7282 Freidenraich D, Stilwell D, Romero E,Wilkes D,Manova K, Basson C, Benezra R, 2004, Rescue of cardiac defects in ID knockout embryos by injection of embryonic stem cells. Science 306:247–252 Lasorella A, Rothschild G, Yokota Y, Russel RG, Iavarone A, 2005, Id2 mediates tumor initiation, proliferation and angiogenesis in Rb mutant mice. Mol Cell Biol 25(9):3563–3574 BalwierzW(2004, Strategie postepowania w nerwiaku zarodkowym wspolczulnym. Przegl Lek 61(2):3–8 Berthold F, Kassenbohmer R, Zieschang J, 1994, Multivariate evaluation of prognostic factors in localized neuroblastoma. Am J Pediatr Hematol Oncol 16:107–115 Brodeur GM, Maris JM, Jamashiro DM, 1997, Biology and genetics of human neuroblastomas. J Pediatr Hematol Oncol 19:93–101 Brodeur GM, Seeger RC, Schwabb M, Varmus HE, Bishop JM, 1984, Amplifiaction of Myc-N in untreated human neuroblastoma correlates with advanced disease stage. Science 224:1121–1124 Caron H, 1995, Allelic loss of chromosome 1 and additional chromosome 17 material are both unfavorable prognostic markers in neuroblastoma. Med Ped Oncol 24(4):215–221 Cohn SL, London WB, Huang D, Katzenstien HM, Salwen HR, Reinhart T, Madafiglio J, Marshall GM, Norris MD, Haber M, 2000, MYCN expression is not prognostic of adverse outcome in advance-stage neuroblastoma with nonamplified MYCN. J Clin Oncol 18(21):3604–3613 Oude Luttikhuis MEM, Powell JA, Rees SA, Genus T, Chughtai S, Ramani P,Mann JR,McCoville CM, 2001, Neuroblastoma with 11q loss and singleMYCN copy comprise a biologically distinct group of tumours with adverse prognosis. Brit J Cancer 85(4):531–537 Vandesompele J, Speleman F, Van Roy N, Laureys G, Brinkschmidt C, Christiansen H, Lampert F, Lastowska M, Bown N, Pearson A, Nicolsson JC, Ross F, Combarett V, Delattre O, Feuerstein DG, Plantaz D, 2001, Multicentre analysis of patterns of DNA gains and loses in 204 neuroblastoma tumors: howmany genetic subgroups are there? Med Ped Oncol 36:5–10 Iavarone A, Lasorella A, 2006, ID proteins as targets in cancer and tools in neurobiology. Trend Mol Med 12:588–594 Vandesompele J, Edsjo D, De Preter K, Axelson H, Speleman F, Pahlman S, 2003, ID2 expression in neuroblastoma does not correlate to MYCN levels. Oncogene 22:456–460 Alaminos M, Gerald WL, Cheung NKV, 2005, Prognostic value of MYCN and ID2 overexpression in neuroblastoma. Pediatr Blood Cancer 45:909–915 Wang Q, Guo C, Hii G, Shusterman S, Mosse Y, Zhao H, Cnaan A, Rappaport E, Hogarty MD, Marris JM, 2003, ID2 expression is not associated with MYCN amplification or expression in human neuroblastoma. Cancer Res 63:1631–1635 Gebauer S, Yu AI, Omura-Minamisawa M, Batova A, Dicciani M, 2004, Expression profiles and clinical relationship of ID2, CDKN1B and CDKN2B in primary neuroblastoma. Genes Chromosomes Cancer 41:297–308 Raetz EA, Kim MK, Moos P, Carslon M, Bruggers C, Hooper DK, Foot L, Liu T, Seeger R, Carroll WL, 2003, Identification of genes that are regulated transcriptionally by Myc in childhood tumors. Cancer 98(4):841–853 Lofstedt T, Jogi A, Sigvardsson A, Gradin K, Poellinger L, Pahlman S, Axelson H, 2004, Indcution of ID2 expression by hypoxiainducible factor-1. A role in dedifferentiation of hypoxic neuroblastoma cells. J Biol Chem 279(38):39223–39231 Perk J, Iavarone A, Benezra R, 2005, Id family of helix-loop-helix protein in cancer. Nat Rev Cancer 5(8):603–614 Ciarapica R, Annibali L, Raimondi L, Savino M, Nasi S, Rota R, 2009, Targeting Id protein interactions by an engineered HLH domains induces human neuroblastoma cell differentiation 13I and neuroblastoma differentiation. Oncogene 28:1881–1891 Han W, Wu Z, Zhao Y, Meng Y, Si Y, Yang J, Fu X, Yu L, 2009, FHL2 interacts with and acts as a functional repressor of Id2 in human neuroblastoma cells. Nucleic Acid Research 12(37):3996–4009 Hu JG, Wu XJ, Feng YF, Xi GM, Deng LX, Wang ZH, Wang R, Shen L, Zhou JS, Lu HZ, 2013, The molecular events involved in oligodendrocyte precursor cell proliferation induced by the conditioned medium from B104 neuroblastoma cells. Neurochem Res 38:601–609 Annibali D, Gioia U, Savino M, Laneve P, Caffarelli E, Nasi S, 2012, A new module in neural differentiation control: two microRNAs upregulated by retinoic acid, miR-9 and −103, target the differentiation inhibitor ID2., DOI 10.1371/journal. pone.0040269 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 999 EP 1004 DI 10.1007/s12253-015-9908-9 PG 6 ER PT J AU Micsik, T Kiszler, G Szabo, D Krecsak, L Hegedus, Cs Krenacs, T Molnar, B AF Micsik, Tamas Kiszler, Gabor Szabo, Daniel Krecsak, Laszlo Hegedus, Csaba Krenacs, Tibor Molnar, Bela TI Computer Aided Semi-Automated Evaluation of HER2 Immunodetection—A Robust Solution for Supporting the Accuracy of Anti HER2 Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Digital pathology; Validation; Image analysis; HER2; Immunohistochemistry ID Breast cancer; Digital pathology; Validation; Image analysis; HER2; Immunohistochemistry AB HER2-positive breast cancers usually benefit from anti-HER2 therapy, thus, HER2 evaluation became inevitable for patient selection. HER2-negative (IHC 0, 1+) and strong positive (IHC 3+) cases can easily be interpreted with immunohistochemistry, but equivocal (IHC 2+) cases require further analysis of HER2 gene amplification using in situ hybridization. Our study aimed to validate digital pathology and automated image analysis for unbiased evaluation of HER2 immunostains. We developed an image segmentation algorithm for analyzing HER2-immunostaining (4B5 clone) in tissue microarrays of breast cancers. Two pathologists assessed 309 microscopic regions of at least 100 tumor cells each— representing all HER2 positivity groups—according to international guidelines either semi-quantitatively or by using the MembraneQuant software. Scoring results were statistically correlated with each other and with FISH data, and almost perfect agreement was found (inter-method Cohen’s kappa=0.872, Spearman-rho=0.928). When clinical relevance (scoring disagreement that may define erroneous treatment selection) was examined high agreement was found (quadratic weighted kappa=0.967). Image analysis classified cases with excellent correlation with visual evaluation, therefore, MembraneQuant software proved to be a reliable tool for assessing HER2 immunoreactions and supporting better targeting anti-HER2 therapy. As digital analysis of immunomorphological markers allows permanent archiving, standardization and accurate reviewing of results, it supports quality assurance initiatives in diagnostic pathology—especially of equivocal cases which are hard to interpret. C1 [Micsik, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi u. 26, 1085 Budapest, Hungary. [Kiszler, Gabor] 3D HISTECH KftBudapest, Hungary. [Szabo, Daniel] 3D HISTECH KftBudapest, Hungary. [Krecsak, Laszlo] 3D HISTECH KftBudapest, Hungary. [Hegedus, Csaba] 3D HISTECH KftBudapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi u. 26, 1085 Budapest, Hungary. [Molnar, Bela] 3D HISTECH KftBudapest, Hungary. RP Micsik, T (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM micsikt@gmail.com;micsikt@mail.korb1.sote.hu CR Nahta R, Shabaya S, Ozbay T, Rowe DL, 2009, Personalizing HER2-targeted therapy in metastatic breast cancer beyond HER2 status: what we have learned from clinical specimens. Curr Pharmacogenomics Pers Med 7(4):263–274 Shah S, Chen B, 2011, Testing for HER2 in breast cancer: a continuing evolution. Pathol Res Int 2011:903202., DOI 10.4061/2011/ 903202 Bartlett JM, Starczynski J, Atkey N, Kay E, O’Grady A, Gandy M, Ibrahim M, Jasani B, Ellis IO, Pinder SE, Walker RA, 2011, HER2 testing in the UK: recommendations for breast and gastric in-situ hybridisation methods. J Clin Pathol 64(8):649–653., DOI 10.1136/ jcp.2011.089847 Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, McShane LM, Paik S, Pegram MD, Perez EA, Press MF, Rhodes A, Sturgeon C, Taube SE, Tubbs R, Vance GH, van de Vijver M, Wheeler TM, Hayes DF, 2007, American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 131(1):18–43., DOI 10.1043/1543- 2165(2007)131[18:ASOCCO]2.0.CO;2 Arnould L, Roger P, Macgrogan G, Chenard MP, Balaton A, Beauclair S, Penault-Llorca F, 2012, Accuracy of HER2 status determination on breast core-needle biopsies, immunohistochemistry, FISH, CISH and SISH vs FISH).Mod Pathol 25(5):675–682., DOI 10. 1038/modpathol.2011.201 Rhodes A, Jasani B, Anderson E, Dodson AR, Balaton AJ, 2002, Evaluation of HER-2/neu immunohistochemical assay sensitivity and scoring on formalin-fixed and paraffin-processed cell lines and breast tumors: a comparative study involving results from laboratories in 21 countries. Am J Clin Pathol 118(3):408–417., DOI 10.1309/ 97WN-W6UX-XJWT-02H2 Mulrane L, Rexhepaj E, Penney S, Callanan JJ, Gallagher WM, 2008, Automated image analysis in histopathology: a valuable tool in medical diagnostics. Expert Rev Mol Diagn 8(6):707–725., DOI 10. 1586/14737159.8.6.707 Yang YL, Fan Y, Lang RG, Gu F, Ren MJ, Zhang XM, Yin D, Fu L, 2012, Genetic heterogeneity of HER2 in breast cancer: impact on HER2 testing and its clinicopathologic significance. Breast Cancer Res Treat 134(3):1095–1102., DOI 10.1007/s10549-012-2046-0 Murphy CG, Modi S, 2009, HER2 breast cancer therapies: a review. Biologics 3:289–301 Choritz H, Busche G, Kreipe H, 2011, Quality assessment of HER2 testing by monitoring of positivity rates. Virchows Arch 459(3):283– 289., DOI 10.1007/s00428-011-1132-8 Dobson L, Conway C, Hanley A, Johnson A, Costello S, O’Grady A, Connolly Y, Magee H, O’Shea D, Jeffers M, Kay E, 2010, Image analysis as an adjunct to manual HER-2 immunohistochemical review: a diagnostic tool to standardize interpretation. Histopathology 57(1):27–38., DOI 10.1111/j.1365-2559.2010.03577.x Gavrielides MA, Gallas BD, Lenz P, Badano A, Hewitt SM, 2011, Observer variability in the interpretation of HER2/neu immunohistochemical expressionwith unaided and computer-aided digitalmicroscopy. Arch Pathol Lab Med 135(2):233–242., DOI 10.1043/1543- 2165-135.2.233 Tuominen VJ, Tolonen TT, Isola J, 2012, ImmunoMembrane: a publicly available web application for digital image analysis of HER2 immunohistochemistry. Histopathology 60(5):758–767., DOI 10.1111/ j.1365-2559.2011.04142.x Brey EM, Lalani Z, Johnston C, Wong M, McIntire LV, Duke PJ, Patrick CW Jr, 2003, Automated selection of DAB-labeled tissue for immunohistochemical quantification. J Histochem Cytochem 51(5): 575–584 Bloom K, Harrington D, 2004, Enhanced accuracy and reliability of HER-2/neu immunohistochemical scoring using digital microscopy. Am J Clin Pathol 121(5):620–630., DOI 10.1309/y73u-8x72-b68tmgh5 Lehr HA, Jacobs TW, Yaziji H, Schnitt SJ, Gown AM, 2001, Quantitative evaluation of HER-2/neu status in breast cancer by fluorescence in situ hybridization and by immunohistochemistry with image analysis. Am J Clin Pathol 115(6):814–822., DOI 10.1309/ aj84-50ak-1x1b-1q4c Ciampa A, Xu B, Ayata G, Baiyee D, Wallace J, Wertheimer M, Edmiston K, Khan A, 2006, HER-2 status in breast cancer: correlation of gene amplification by FISH with immunohistochemistry expression using advanced cellular imaging system. Appl Immunohistochem Mol Morphol 14(2):132–137., DOI 10.1097/01. pai.0000150516.75567.13 Keller B, Chen W, Gavrielides MA, 2012, Quantitative assessment and classification of tissue-based biomarker expression with color content analysis. Arch Pathol Lab Med 136(5):539–550., DOI 10. 5858/arpa.2011-0195-OA Krecsak L, Micsik T, Kiszler G, Krenacs T, Szabo D, Jonas V, Csaszar G, Czuni L, Gurzo P, Ficsor L, Molnar B, 2011, Technical note on the validation of a semi-automated image analysis software application for estrogen and progesterone receptor detection in breast cancer. Diagn Pathol 6:6., DOI 10.1186/1746-1596-6-6 Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JM, BilousM, Fitzgibbons P, Hanna W, Jenkins RB, Mangu PB, Paik S, Perez EA, Press MF, Spears PA, Vance GH, Viale G, Hayes DF, 2013, Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol 31(31):3997–4013., DOI 10.1200/jco.2013.50.9984 Landis JR,KochGG(1977, Themeasurement of observer agreement for categorical data. Biometrics 33(1):159–174 Fitzgibbons PL, Murphy DA, Dorfman DM, Roche PC, Tubbs RR, 2006, Interlaboratory comparison of immunohistochemical testing for HER2: results of the 2004 and 2005 College of American Pathologists HER2 Immunohistochemistry Tissue Microarray Survey. Arch Pathol Lab Med 130(10):1440–1445., DOI 10.1043/ 1543-2165(2006)130[1440:ICOITF]2.0.CO;2 Martin V, Camponovo A, Ghisletta M, Bongiovanni M, Mazzucchelli L, 2012, Internal Quality Assurance Program for ERBB2, HER2, testing improves the selection of breast cancer patients for treatment with Trastuzumab. Pathol Res Int 2012:261857., DOI 10.1155/2012/261857 Roche PC, Suman VJ, Jenkins RB, Davidson NE, Martino S, Kaufman PA, Addo FK, Murphy B, Ingle JN, Perez EA, 2002, Concordance between local and central laboratory HER2 testing in the breast intergroup trial N9831. J Natl Cancer Inst 94(11):855–857 Grimm EE, Schmidt RA, Swanson PE, Dintzis SM, Allison KH, 2010, Achieving 95% cross-methodological concordance in HER2 testing: causes and implications of discordant cases. AmJ Clin Pathol 134(2):284–292., DOI 10.1309/AJCPUQB18XZOHHBJ Andersson J, Linderholm B, Bergh J, Elmberger G, 2004, HER-2/ neu, c-erbB-2, evaluation in primary breast carcinoma by fluorescent in situ hybridization and immunohistochemistry with special focus on intratumor heterogeneity and comparison of invasive and in situ components. Appl Immunohistochem Mol Morphol 12(1):14–20 Shin SJ, Hyjek E, Early E, Knowles DM, 2006, Intratumoral heterogeneity of her-2/neu in invasive mammary carcinomas using fluorescence in-situ hybridization and tissue microarray. Int J Surg Pathol 14(4):279–284., DOI 10.1177/1066896906293055 Joshi AS, Sharangpani GM, Porter K, Keyhani S, Morrison C, Basu AS, Gholap GA, Gholap AS, Barsky SH, 2007, Semi-automated imaging system to quantitate Her-2/neu membrane receptor immunoreactivity in human breast cancer. Cytometry A 71(5):273–285., DOI 10.1002/cyto.a.20374 Health CfDaR, 2007, 510(k, substantial equivalence determination decision summary for Aperio Technologies, Inc. ScanScope® XT System, IHC HER2/neu Image Analysis Application. Food and Drug Administration. http://www.accessdata.fda.gov/cdrh_docs/ reviews/K071128.pdf. Accessed 28 Mar 2013 Health CfDaR, 2004, 510(k, substantial equivalence determination decision summary for Applied Imaging Corporation, Applied Imaging Ariol™ with HER2 Application. Food and Drug Administration. http://www.accessdata.fda.gov/cdrh_docs/pdf3/ k032113.pdf. Accessed 28 Mar 2013 Health CfDaR, 2003, 510(k, substantial equivalence determination decision summary for ChromaVision Medical Systems Inc. Automated Cellular Imaging System. . http://www.accessdata.fda. gov/cdrh_docs/reviews/K031715.pdf. Accessed 28 Mar 2012 Micsik TKG, Szabo D, Krecsak L, Krenacs T, Molnar B, 2013, Is HER2 amplification predictable by digital immunohistochemistry? Diagn Pathol 8(Suppl 1):S14 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1005 EP 1011 DI 10.1007/s12253-015-9927-6 PG 7 ER PT J AU Li, J Hu, G Kong, F Wu, K Song, K He, J Sun, W AF Li, Jian Hu, Guohuang Kong, Fujiao Wu, Kemin Song, Kun He, Jianfeng Sun, Weijia TI Elevated STMN1 Expression Correlates with Poor Prognosis in Patients with Pancreatic Ductal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic ductal adenocarcinoma (PDAC); STMN1; Prognosis ID Pancreatic ductal adenocarcinoma (PDAC); STMN1; Prognosis AB STMN1 is a cytosolic phosphoprotein that not only participates in cell division, but also plays an important role in other microtubule-dependent processes, such as cell motility. Furthermore, STMN1 acts as a "relay protein" in several intracellular signaling pathways that influence cell growth and differentiation. Thus, STMN1 is likely to support cellular processes essential for tumor progression: survival and migration. Indeed, elevated STMN1 expression has been reported in various types of human malignancies and is correlated with poor prognosis in these human malignancies. However, the clinical and prognostic significance of STMN1 in pancreatic ductal adenocarcinoma (PDAC) remains unknown. Thus, we assessed STMN1 in PDAC in this retrospective study.We first examined STMN1 expression in PDAC tissues from 27 cases and matched adjacent non-cancerous tissues by quantitative polymerase chain reaction (PCR) and western blot analyses. Next, immunohistochemistry was used to evaluate STMN1 expression in 87 archived paraffin-embedded PDAC specimens. STMN1 mRNA and protein expression levels were to a large extent up-regulated in PDAC tissue compared with their adjacent non-cancerous tissues. Moreover, STMN1 expression was closely correlated with histological differentiation, lymphatic metastasis, and TNM stage (P= 0.023, 0.047, and 0.014, respectively). In addition, PDAC patients with higher STMN1 expression died sooner than those with lower STMN1 expression (P<0.01). Multivariate analysis demonstrated that STMN1 expression was an independent prognostic factor for PDAC patients (P<0.01). Herein, we provide the first evidence that up-regulated STMN1 may contribute to tumor progression and poor prognosis in PDAC patients and may serve as a novel prognostic marker. C1 [Li, Jian] Central South University, Xiangya Hospital, Department of PET centerChangsha, China. [Hu, Guohuang] The fourth Hospital of Changsha, Department of General SurgeryChangsha, China. [Kong, Fujiao] Central South University, Xiangya Hospital, Department of AnesthesiologyChangsha, China. [Wu, Kemin] Central South University, Xiangya Hospital, Department of General SurgeryChangsha, China. [Song, Kun] Central South University, Xiangya Hospital, Department of General SurgeryChangsha, China. [He, Jianfeng] Central South University, Xiangya Hospital, Department of General SurgeryChangsha, China. [Sun, Weijia] Central South University, Xiangya Hospital, Department of General SurgeryChangsha, China. RP Sun, W (reprint author), Central South University, Xiangya Hospital, Department of General Surgery, Changsha, China. EM sunweijia2013@126.com CR Ferlay J, Shin HR, Bray F, Forman D,Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127:2893–2917 Malik NK, Malik NK, May KS, Chandrasekhar R, Wee W, Flaherty L, Iyer R, Gibbs J, Kuvshinoff B, Wilding G, Warren G, Yang GY, 2012, Treatment of locally advanced unresectable pancreatic cancer: a 10-year experience. J Gastrointest Oncol 3:326–334 Kaur S, Baine MJ, Jain M, Sasson AR, Batra SK, 2012, Early diagnosis of pancreatic cancer: challenges and new developments. Biomark Med 6:597–612 McCleary-Wheeler AL, Lomberk GA, Weiss FU, Schneider G, Fabbri M, Poshusta TL, Dusetti NJ, Baumgart S, Iovanna JL, Ellenrieder V, Urrutia R, Fernandez-Zapico ME, 2012, Insights into the epigenetic mechanisms controlling pancreatic carcinogenesis. Cancer Lett 328:212–221 Sun C, Ansari D, Andersson R, Wu DQ, 2012, Does gemcitabinebased combination therapy improve the prognosis of unresectable pancreatic cancer? World J Gastroenterol 18:4944–4958 Howell B, Larsson N, GullbergM, Cassimeris L, 1999, Dissociation of the tubulin-sequestering and microtubule catastrophe-promoting activities of oncoprotein 18/stathmin. Mol Biol Cell 10:105–118 Belmont LD, Mitchison TJ, 1999, Identification of a protein that interacts with tubulin dimers and increases the catastrophe rate of microtubules. Cell 84:623–631 Iancu-Rubin C, Nasrallah CA, Atweh GF, 2005, Stathmin prevents the transition from a normal to an endomitotic cell cycle during megakaryocytic differentiation. Cell Cycle 4:1774–1782 Sobel A, 1991, Stathmin: a relay phosphoprotein for multiple signal transduction? Trends Biochem Sci 16:301–305 Saal LH, Johansson P, Holm K, Gruvberger-Saal SK, She QB, Maurer M, Koujak S, Ferrando AA, Malmstrom P, Memeo L, Isola J, Bendahl PO, Rosen N, Hibshoosh H, Ringner M, Borg A, Parsons R, 2007, Poor prognosis in carcinoma is associated with a gene expression signature of aberrant PTEN tumor suppressor pathway activity. Proc Natl Acad Sci U S A 104:7564–7569 Chung MK, Kim HJ, Lee YS, Han ME, Yoon S, Baek SY, Kim BS, Kim JB, Oh SO, 2010, Hedgehog signaling regulates proliferation of prostate cancer cells via stathmin1. Clin Exp Med 10:51–57 Karst AM, Levanon K, Duraisamy S, Liu JF, Hirsch MS, Hecht JL, Drapkin R, 2011, Stathmin 1, a marker of PI3K pathway activation and regulator of microtubule dynamics, is expressed in early pelvic serous carcinomas. Gynecol Oncol 123:5–12 Bieche I, Lachkar S, Becette V, Cifuentes-Diaz C, Sobel A, Lidereau R, Crumi PA, 1998, Overexpression of the stathmin gene in a subset of human breast cancer. Br J Cancer 78:701–709 Singer S, Ehemann V, Brauckhoff A, Keith M, Vreden S, Schirmacher P, Breuhahn K, 2007, Protumorigenic overexpression of stathmin/Op18 by gain-of-function mutation in p53 in human hepatocarcinogenesis. Hepatology 46:759–768 Kouzu Y, Uzawa K, Koike H, Saito K, Nakashima D, Higo M, Endo Y, Kasamatsu A, Shiiba M, Bukawa H, Yokoe H, Tanzawa H, 2006, Overexpression of stathmin in oral squamous-cell carcinoma: correlation with tumour progression and poor prognosis. Br J Cancer 94: 717–723 Yuan RH, Jeng YM, Chen HL, Lai PL, Pan HW, Hsieh FJ, Lin CY, Lee PH, Hsu HC, 2006, Stathmin overexpression cooperates with p53 mutation and osteopontin overexpression, and is associated with tumour progression, early recurrence, and poor prognosis in hepatocellular carcinoma. J Pathol 209:549–558 Jeon TY, Han ME, Lee YW, Kim GH, Song GA, Hur GY, Kim JY, Kim HJ, Yoon S, Baek SY, Kim BS, Kim JB, Oh SO, 2010, Overexpression of stathmin1 in the diffuse type of gastric cancer and its roles in proliferation and migration of gastric cancer cells. Br J Cancer 102:710–718 Chen G,Wang H,Gharib TG, Huang CC, Thomas DG, Shedden KA, Kuick R, Taylor JM, Kardia SL, Misek DE, Giordano TJ, Iannettoni MD, Orringer MB, Hanash SM, Beer DG, 2003, Overexpression of oncoprotein 18 correlates with poor differentiation in lung adenocarcinomas. Mol Cell Proteomics 2:107–116 Ghosh R, Gu G, Tillman E, Yuan J, Wang Y, Fazli L, Rennie PS, Kasper S, 2007, Increased expression and differential phosphorylation of stathmin may promote prostate cancer progression. Prostate 67:1038–1052 Gan L, Guo K, Li Y, Kang X, Sun L, Shu H, Liu Y, 2010, Upregulated expression of stathmin may be associated with hepatocarcinogenesis. Oncol Rep 23:1037–1043 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), Method. Method 25:402–408 Chen HT, Cai QC, Zheng JM,Man XH, Jiang H, Song B, Jin G, Zhu W, Li ZS, 2012, High expression of delta-like ligand 4 predicts poor prognosis after curative resection for pancreatic cancer. Ann Surg Oncol 19(Suppl 3):S464–S474 de JongMC, Li F, Cameron JL,Wolfqang CL, Edil BH, Herman JM, Choti MA, Eckhauser F, Hirose K, Schulick RD, Pawlik TM, 2011, Re-evaluating the impact of tumor size on survival following pancreaticoduodenectomy for pancreatic adenocarcinoma. J Surg Oncol 103:656–662 Ueda M, Endo I, Nakashima M, Minami Y, Takeda K, Naqano Y, Tanaka K, Ichikawa Y, Toqo S, Kunisaki C, Shimada H, 2009, Prognostic factors after resection of pancreatic cancer. World J Surg 33:104–110 Gleason MX, Mdzinarishvili T, Are C, Sasson A, Sherman A, Shats O, Sherman S, 2013, Prognostic estimator of survival for patients with localized and extended pancreatic ductal adenocarcinoma. Cancer Informat 12:103–114 Perini MV, Montaqnini AL, Jukemura J, Penteado S, Adbo EE, Patzina R, Cecconello I, Cunha JE, 2008, Clinical and pathologic prognostic factors for curative resection for pancreatic cancer. HPB, Oxford, 10:356–362 Sobel A, Tashjian AH Jr, 1983, Distinct patterns of cytoplasmic protein phosphorylation related to regulation of synthesis and release of prolactin by GH cells. J Biol Chem 258:10312–10324 Mistry SJ, Atweh GF, 2001, Stathmin inhibition enhances okadaic acid-induced mitotic arrest: a potential role for stathmin in mitotic exit. J Biol Chem 276:31209–31215 Mistry SJ, Atweh GF, 2006, Therapeutic interactions between stathmin inhibition and chemotherapeutic agents in prostate cancer. Mol Cancer Ther 5:3248–3257 Curmi PA, Noques C, Lachkar S, Carelle N, Gonthier MP, Sobel A, Lidereau R, Bieche I, 2000, Overexpression of stathmin in breast carcinomas points out to highly proliferative tumours. Br J Cancer 82:142–150 Zheng P, Liu YX, Chen L, Liu XH, Xiao ZQ, Zhao L, Li GQ, Zhou J, Ding YQ, Li JM, 2010, Stathmin, a new target of PRL-3 identified by proteomic methods, plays a key role in progression and metastasis of colorectal cancer. J Proteome Res 9:4897–4905 Tan HT, Wu W, Nq YZ, Zhang X, Yan B, Ong CW, Tan S, Salto- Tellez M, Hooi SC, Chung MC, 2012, Proteomic analysis of colorectal cancer metastasis: stathmin-1 revealed as a player in cancer cell migration and prognostic marker. J Proteome Res 11:1433–1445 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1013 EP 1020 DI 10.1007/s12253-015-9930-y PG 8 ER PT J AU Wang, XX Liu, BB Wu, X Su, D Zhu, Z Fu, L AF Wang, Xin-Xin Liu, Bing-Bing Wu, Xiao Su, Dan Zhu, Zhengmao Fu, Li TI Loss of Leucine Zipper Putative Tumor Suppressor 1 (LZTS1) Expression Contributes to Lymph Node Metastasis of Breast Invasive Micropapillary Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast IMPC; LZTS1; Gene expression and promoter methylation ID Breast IMPC; LZTS1; Gene expression and promoter methylation AB Breast invasive micropapillary carcinoma (IMPC) is a rare subtype of breast cancer with a high potential of lymph node metastasis, aggressive clinical behavior, and poor disease-free or overall survival. Expression of leucine zipper putative tumor suppressor 1 (LZTS1) was frequently lost or reduced in breast cancer tissues. This study investigated the expression of LZTS1 protein in breast IMPC tissues using immunohistochemistry. In addition, somatic LZTS1 mutations and promoter methylation were assessed to determine an association with clinicopathological data from IMPC patients. LZTS1 protein was downregulated in 62 (62 %) of 100 IMPC tissue samples and was significantly associated with lymph node metastasis (P<0.05). A LZTS1 exon mutation occurred in one of the 53 IMPC cases analyzed, whereas a LZTS1 intron mutation occurred in 26 of 53 cases. Moreover, LZTS1 promoter was frequently methylated in IMPC samples and was associated with reduced LZTS1 expression levels in IMPC tissues. These data demonstrated that the loss of LZTS1 expression was associated with lymph node metastasis in patients with IMPC, and LZTS1 promoter methylation could be responsible for the loss of LZTS1 expression. C1 [Wang, Xin-Xin] Capital Medical University, Beijing Youan Hospital, Department of Pathology, 100069 Beijing, China. [Liu, Bing-Bing] Key Laboratory of Cancer Prevention and Therapy Tianjin, Tianjin Medical University Cancer Institute and Hospital, Huan Hu Xi Road, 300060 Tianjin, China. [Wu, Xiao] Nankai University, College of Life Sciences, Department of Genetics and Cell Biology, 300071 Tianjin, China. [Su, Dan] Nankai University, College of Life Sciences, Department of Genetics and Cell Biology, 300071 Tianjin, China. [Zhu, Zhengmao] Nankai University, College of Life Sciences, Department of Genetics and Cell Biology, 300071 Tianjin, China. [Fu, Li] Key Laboratory of Cancer Prevention and Therapy Tianjin, Tianjin Medical University Cancer Institute and Hospital, Huan Hu Xi Road, 300060 Tianjin, China. RP Fu, L (reprint author), Key Laboratory of Cancer Prevention and Therapy Tianjin, Tianjin Medical University Cancer Institute and Hospital, 300060 Tianjin, China. EM tj.lzts1@gmail.com CR Lakhani SR, Ellis IO, Schnitt SJ, Tan PH, van de Vijver MJ, 2011, WHO classification of tumors of the breast. IARC Press, Lyon, pp 14–17 Tavassoli FA, Devilee P, 2003, WHO classification of tumors. Pathology & genetics, tumours of the breast and female genital organs. IARC Press, Lyon, p 10 Nassar H, Wallis T, Andea A, Dey J, Adsay V, Visscher D, 2001, Clinicopathologic analysis of invasive micropapillary differentiation in breast carcinoma. Mod Pathol 14:836–841 Walsh MM, Bleiweiss IJ, 2001, Invasive micropapillary carcinoma of the breast: eighty cases of an underrecognized entity. Hum Pathol 32:583–589 Pettinato G, Manivel CJ, Panico L, Sparano L, Petrella G, 2004, Invasive micropapillary carcinoma of the breast: clinicopathologic study of 62 cases of a poorly recognized variant with highly aggressive behavior. Am J Clin Pathol 121:857–866 Ishii H, Baffa R, Numata SI, Murakumo Y, Rattan S, Inoue H, Mori M, Fidanza V, Alder H, Croce CM, 1999, The FEZ1 gene at chromosome 8p22 encodes a leucine-zipper protein, and its expression is altered in multiple human tumors. Proc Natl Acad Sci U S A 96: 3928–3933 Toyooka S, Fukuyama Y, Wistuba II, Tockman MS, Minna JD, Gazdar AF, 2002, Differential expression of FEZ1/LZTS1 gene in lung cancers and their cell cultures. Clin Cancer Res 8:2292–2297 Vecchione A, Ishii H, Shiao YH, Trapasso F, Rugge M, Tamburrino JF, Murakumo Y, Alder H, Croce CM, Baffa R, 2001, Fez1/lzts1 alterations in gastric carcinoma. Clin Cancer Res 7:1546–1552 Vecchione A, Ishii H, Baldassarre G, Bassi P, Trapasso F, Alder H, Pagano F, Gomella LG, Croce CM, Baffa R, 2002, FEZ1/LZTS1 is down-regulated in high-grade bladder cancer, and its restoration suppresses tumorigenicity in transitional cell carcinoma cells. Am J Pathol 160:1345–1352 Nonaka D, Fabbri A, Roz L, Mariani L, Vecchione A, Moore GW, Tavecchio L, Croce CM, Sozzi G, 2005, Reduced FEZ1/LZTS1 expression and outcome prediction in lung cancer. Cancer Res 65: 1207–1212 Knowles MA, Aveyard JS, Taylor CF, Harnden P, Bass S, 2005, Mutation analysis of the 8p candidate tumour suppressor genes DBC2, RHOBTB2, and LZTS1 in bladder cancer. Cancer Lett 225: 121–130 Ono K, Uzawa K, Nakatsuru M, Shiiba M, Mochida Y, Tada A, Bukawa H, Miyakawa A, Yokoe H, Tanzawa H, 2003, Downregulation of FEZ1/LZTS1 gene with frequent loss of heterozygosity in oral squamous cell carcinomas. Int J Oncol 23:297–302 Vecchione A, Galetti TP, GardimanM, Ishii H, Giarnieri E, Pagano F, Gomella LG, Croce CM, Baffa R, 2004, Collecting duct carcinoma of the kidney: an immunohistochemical study of 11 cases.BMC Urol 4:11 Ishii H, Vecchione A, Murakumo Y, Baldassarre G, Numata S, Trapasso F, Alder H, Baffa R, Croce CM, 2001, FEZ1/LZTS1 gene at 8p22 suppresses cancer cell growth and regulates mitosis. Proc Natl Acad Sci U S A 98:10374–10379 Chen L, Zhu Z, Sun X, Dong XY,Wei J, Gu F, Sun YL, Zhou J,Dong JT, Fu L, 2009, Down-regulation of tumor suppressor gene FEZ1/ LZTS1 in breast carcinoma involves promoter methylation and associates with metastasis. Breast Cancer Res Treat 116:471–478 Guo X, Chen L, Lang R, Fan Y, Zhang X, Fu L, 2006, Invasive micropapillary carcinoma of the breast: association of pathologic features with lymph node metastasis. Am J Clin Pathol 126:740–746 Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB, 1996, Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S A 93:9821–9826 Arnold JM, Choong DY, Lai J, Campbell IG, Chenevix-Trench G, 2006, Mutation and expression analysis of LZTS1 in ovarian cancer. Cancer Lett 233:151–157 Baylin SB, Herman JG, Graff JR, Vertino PM, Issa JP, 1998, Alterations in DNA methylation: a fundamental aspect of neoplasia. Adv Cancer Res 72:141–196 Wang XX, Zhu Z, Su D, Lei T, Wu X, Fan Y, Li X, Zhao J, Fu L, Dong JT, Fu L, 2011, Down-regulation of leucine zipper putative tumor suppressor 1 is associated with poor prognosis, increased cell motility and invasion, and pithelial-to-mesenchymal transition characteristics in human breast carcinoma. Hum Pathol 42:1410–1419 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1021 EP 1026 DI 10.1007/s12253-015-9923-x PG 6 ER PT J AU Zavesky, L Jandakova, E Turyna, R Langmeierova, L Weinberger, V Zaveska Drabkova, L Hulkova, M Horinek, A Duskova, D Feyereisl, J Minar, L Kohoutova, M AF Zavesky, Ludek Jandakova, Eva Turyna, Radovan Langmeierova, Lucie Weinberger, Vit Zaveska Drabkova, Lenka Hulkova, Martina Horinek, Ales Duskova, Daniela Feyereisl, Jaroslav Minar, Lubos Kohoutova, Milada TI Evaluation of Cell-Free Urine microRNAs Expression for the Use in Diagnosis of Ovarian and Endometrial Cancers. A Pilot Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ovarian cancer; Endometrial cancer; Urine; microRNA; miR-92a; miR-106b ID Ovarian cancer; Endometrial cancer; Urine; microRNA; miR-92a; miR-106b AB Among gynaecological cancers, epithelial ovarian cancers are the most deadly cancers while endometrial cancers are the most common diseases. Efforts to establish relevant novel diagnostic, screening and prognostic markers are aimed to help reduce the high level of mortality, chemoresistance and recurrence, particularly in ovarian cancer. MicroRNAs, the class of post-transcriptional regulators, have emerged as the promising diagnostic and prognostic markers associated with various diseased states recently. Urine has been shown as the source of microRNAs several years ago; however, there has been lack of information on urine microRNA expression in ovarian and endometrial cancers till now. In this pilot study, we examined the expression of candidate cell-free urine microRNAs in ovarian cancer and endometrial cancer patients using quantitative real-time PCR. We compared the expression between pre- and post-surgery ovarian cancer samples, and between patients with ovarian and endometrial cancers and healthy controls, within three types of experiments. These experiments evaluated three different isolation methods of urine RNA, representing two supernatant and one exosome fractions of extracellular microRNA. In ovarian cancer, we found miR-92a significantly up-regulated, and miR-106b significantly down-regulated in comparison with control samples. In endometrial cancer, only miR-106b was found down-regulated significantly compared to control samples. Using exosome RNA, no significant de-regulations in microRNAs expression could be found in either of the cancers investigated.We propose that more research should now focus on confirming the diagnostic potential of urine microRNAs in gynaecological cancers using more clinical samples and largescale expression profiling methods. C1 [Zavesky, Ludek] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Institute of Biology and Medical Genetics, Albertov 4, CZ-128 00 Prague, Czech Republic. [Jandakova, Eva] University Hospital Brno, Department of Pathology, Obilni trh 11, CZ-625 00 Brno, Czech Republic. [Turyna, Radovan] Institute for the Care of Mother and Child, Podolske nabrezi 157, CZ-147 00 Prague, Czech Republic. [Langmeierova, Lucie] General University Hospital in Prague, Faculty Transfusion Centre, U Nemocnice 2, CZ-128 08 Prague, Czech Republic. [Weinberger, Vit] University Hospital Brno, Department of Obstetrics and Gynaecology, Obilni trh 11, CZ-625 00 Brno, Czech Republic. [Zaveska Drabkova, Lenka] Academy of Sciences of the Czech Republic, Institute of Botany of the ASCR, v. v. i., Zamek 1, CZ-252 43 Prague, Czech Republic. [Hulkova, Martina] General University Hospital in Prague, Department of Pediatrics and Adolescent Medicine, Ke Karlovu 2, CZ-121 00 Prague, Czech Republic. [Horinek, Ales] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Institute of Biology and Medical Genetics, Albertov 4, CZ-128 00 Prague, Czech Republic. [Duskova, Daniela] General University Hospital in Prague, Faculty Transfusion Centre, U Nemocnice 2, CZ-128 08 Prague, Czech Republic. [Feyereisl, Jaroslav] Institute for the Care of Mother and Child, Podolske nabrezi 157, CZ-147 00 Prague, Czech Republic. [Minar, Lubos] University Hospital Brno, Department of Obstetrics and Gynaecology, Obilni trh 11, CZ-625 00 Brno, Czech Republic. [Kohoutova, Milada] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Institute of Biology and Medical Genetics, Albertov 4, CZ-128 00 Prague, Czech Republic. RP Zavesky, L (reprint author), Charles University, 1st Faculty of Medicine and General Teaching Hospital, Institute of Biology and Medical Genetics, CZ-128 00 Prague, Czech Republic. EM ludek.zavesky@gmail.com CR Siegel R, Ma J, Zou Z, Jemal A, 2014, Cancer statistics, 2014. Ca- Cancer J Clin 64:9–29., DOI 10.3322/caac.21208 Howlader N, Noone AM, Krapcho M et al., eds,, 2013, SEER Cancer Statistics Review, 1975–2010, National Cancer Institute. Bethesda, MD. [http://seer.cancer.gov/csr/1975_2010/, based on November 2012 SEER data submission, posted to the SEER web site, April 2013]. Zavesky L, Jancarkova N, Kohoutova M, 2011, Ovarian cancer: origin and factors involved in carcinogenesis with potential use in diagnosis, treatment and prognosis of the disease. Neoplasma 58: 457–468., DOI 10.4149/neo_2011_06_457 Devor EJ, Hovey AM, GoodheartMJ et al, 2011, microRNA expression profiling of endometrial endometrioid adenocarcinomas and serous adenocarcinomas reveals profiles containing shared, unique and differentiating groups of microRNAs. Oncol Rep 26:995–1002., DOI 10.3892/or.2011.1372 Banno K, Nogami Y, Kisu I et al, 2013, Candidate biomarkers for genetic and clinicopathological diagnosis of endometrial cancer. Int J Mol Sci 14(6):12123–12137., DOI 10.3390/ijms140612123 Iorio MV, Croce CM, 2012, MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med 4:143–159., DOI 10.1002/emmm.201100209 Zhao YN, Chen GS, Hong SJ, 2014, Circulating MicroRNAs in gynecological malignancies: from detection to prediction. Exp Hematol Oncol 3:14., DOI 10.1186/2162-3619-3-14 Weber JA, Baxter DH, Zhang SL et al, 2010, The MicroRNA Spectrum in 12 Body Fluids. Clin Chem, Washington, DC, U S, 56:1733–1741., DOI 10.1373/clinchem.2010.147405 Yamada Y, Enokida H, Kojima S et al, 2011, MiR-96 and miR-183 detection in urine serve as potential tumor markers of urothelial carcinoma: correlation with stage and grade, and comparison with urinary cytology. Cancer Sci 102:522–529., DOI 10.1111/j.1349-7006. 2010.01816.x Bryant RJ, Pawlowski T, Catto JWF et al, 2012, Changes in circulating microRNA levels associated with prostate cancer. Br J Cancer 106:768–774., DOI 10.1038/bjc.2011.595 Mengual L, Loyano JJ, Ingelmo-Torres M et al, 2013, Using microRNA profiling in urine samples to develop a non-invasive test for bladder cancer. Int J Cancer 133(11):2631–2641., DOI 10.1002/ijc. 28274 Hanke M, Hoefig K, Merz H et al, 2010, A robust methodology to study urine microRNA as tumor marker: microRNA-126 and microRNA-182 are related to urinary bladder cancer. Urol Oncol Semin Orig Invest 28:655–661., DOI 10.1016/j.urolonc.2009.01.027 Haj-Ahmad TA, Abdalla MAK, Haj-Ahmad Y, 2014, Potential urinary miRNA biomarker candidates for the accurate detection of prostate cancer among benign prostatic hyperplasia patients. J Cancer 5(3):182–191., DOI 10.7150/jca.6799 Wang G, Chan ESY, Kwan BCH et al, 2012, Expression of microRNAs in the urine of patients with bladder cancer. Clin Genitourin Cancer 10(2):106–113., DOI 10.1016/j.clgc.2012.01.001 Zhao S, Fernald RD, 2005, Comprehensive algorithm for quantitative real-time polymerase chain reaction. J Comput Biol 12:1047– 1064., DOI 10.1089/cmb.2005.12.1047 Guescini M, Sisti D, Rocchi MBL et al, 2008, A new real-time PCR method to overcome significant quantitative inaccuracy due to slight amplification inhibition. BMC Bioinf 9:326., DOI 10.1186/1471- 2105-9-326 Pfaffl MW, Tichopad A, Prgomet C, Neuvians TP, 2004, Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper - excel-based tool using pair-wise correlations. Biotechnol Lett 26:509–515., DOI 10. 1023/B:BILE.0000019559.84305.47 Andersen CL, Jensen JL, Orntoft TF, 2004, Normalization of realtime quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res 64:5245– 5250., DOI 10.1158/0008-5472.CAN-04-0496 Wyman SK, Parkin RK, Mitchell PS et al, 2009, Repertoire of microRNAs in epithelial ovarian cancer as determined by next generation sequencing of small RNA cDNA libraries. Plos One 4:e5311., DOI 10.1371/journal.pone.0005311 Resnick KE, Alder H, Hagan JP et al, 2009, The detection of differentially expressed microRNAs from the serum of ovarian cancer patients using a novel real-time PCR platform. Gynecol Oncol 112: 55–59., DOI 10.1016/j.ygyno.2008.08.036 Dahiya N, Sherman-Baust CA, Wang TL et al, 2008, MicroRNA expression and identification of putative miRNA targets in ovarian cancer. Plos One 3(6):e2436., DOI 10.1371/journal.pone.0002436 Hausler SFM, Keller A, Chandran PA et al, 2010, Whole bloodderived miRNA profiles as potential new tools for ovarian cancer screening. Br J Cancer 103:693–700., DOI 10.1038/sj.bjc.6605833 Shapira I, Oswald M, Lovecchio J et al, 2014, Circulating biomarkers for detection of ovarian cancer and predicting cancer outcomes. Br J Cancer 110:976–983., DOI 10.1038/bjc.2013.795 Laios A, O’Toole S, Flavin R et al, 2008, Potential role of miR-9 and miR-223 in recurrent ovarian cancer. Mol Cancer 7:35., DOI 10.1186/ 1476-4598-7-35 Dong P, Kaneuchi M, Watari H et al, 2014, MicroRNA-106b modulates epithelial-mesenchymal transition by targeting TWIST1 in invasive endometrial cancer cell lines. Mol Carcinog 53:349–359., DOI 10.1002/mc.21983 Ni X, Xia T, Zhao T et al, 2014, Downregulation of miR-106b induced breast cancer cell invasion and motility in association with overexpression of matrix metalloproteinase 2. Cancer Sci 105:18– 25., DOI 10.1111/cas.12309 Hiroki E, Akahira J, Suzuki F et al, 2010, Changes in microRNA expression levels correlate with clinicopathological features and prognoses in endometrial serous adenocarcinomas. Cancer Sci 101: 241–249., DOI 10.1111/j.1349-7006.2009.01385.x Kan CWS, Hahn MA, Gard GB et al, 2012, Elevated levels of circulating microRNA-200 family members correlate with serous epithelial ovarian cancer. BMC Cancer 12:627., DOI 10.1186/1471-2407- 12-627 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1027 EP 1035 DI 10.1007/s12253-015-9914-y PG 9 ER PT J AU Oshikawa, G Yoshioka, K Takahashi, Y Shingai, N Ikegawa, Sh Kobayashil, T Doki, N Kakihana, K Ohashi, K Sakamaki, H AF Oshikawa, Gaku Yoshioka, Kousuke Takahashi, Yukie Shingai, Naoki Ikegawa, Shuntaro Kobayashil, Takeshi Doki, Noriko Kakihana, Kazuhiko Ohashi, Kazuteru Sakamaki, Hisashi TI Impact of prior azacitidine on the outcome of allogeneic hematopoietic transplantation for myelodysplastic syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Myelodysplastic syndrome; Allogeneic hematopoietic stem cell transplantation; Azacitidine ID Myelodysplastic syndrome; Allogeneic hematopoietic stem cell transplantation; Azacitidine AB To clarify the clinical impact of prior use of azacitidine (AZA) on outcomes of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for myelodysplastic syndrome (MDS), we retrospectively reviewed the clinical outcomes of 15 MDS patients who were treated with AZA before allo-HSCT (AZA group). We compared the outcomes of these 15 patients with 52 MDS patients who were solely given the best supportive care (BSC) before allo-HSCT (BSC group). Although patients in the AZA group were older with higher International Prognostic Scoring System (IPSS) scores compared to patients in the BSC group, no significant differences were found between the two groups in overall survival (OS), disease-free survival (DFS), cumulative incidence of relapse (CIR) or non-relapse mortality. However, in patients with a higher IPSS score (Int-2/High), pre-transplant AZA may provide better OS and DFS and lower CIR. Acute graft-versus-host disease rates were similar between the two groups. These results should be reassuring to patients with high-risk MDS receiving AZA before allo-HSCT. C1 [Oshikawa, Gaku] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Yoshioka, Kousuke] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Takahashi, Yukie] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Shingai, Naoki] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Ikegawa, Shuntaro] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kobayashil, Takeshi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Doki, Noriko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kakihana, Kazuhiko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Ohashi, Kazuteru] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Sakamaki, Hisashi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. RP Oshikawa, G (reprint author), Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 113-8677 Tokyo, Japan. EM gaku.oshikawa@gmail.com CR Gyurkocza B, Deeg HJ, 2012, Allogeneic hematopoietic cell transplantation forMDS: forwhom, when and how? Blood Rev 26:247– 254 Silverman LR, Demakos EP, Peterson BL, Kornblith AB, Holland JC, Odchimar-Reissig R et al, 2002, Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol 20:2429–2440 Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A et al, 2009, Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol 10:223–232 Kim DY, Lee JH, Park YH, Kim SD, Choi Y, Lee SB et al, 2012, Feasibility of hypomethylating agents followed by allogeneic hematopoietic cell transplantation in patients with myelodysplastic syndrome. Bone Marrow Transplant 47:374–379 Damaj G, Duhamel A, Robin M, Beguin Y, Michallet M,MohtyM et al, 2012, Impact of azacitidine before allogeneic stem-cell transplantation for myelodysplastic syndromes: a study by the Societe Francaise de Greffe de Moelle et de Therapie-Cellulaire and the Groupe-Francophone des Myelodysplasies. J Clin Oncol 20: 4533–4540 Gerds AT, Gooley TA, Estey EH, Appelbaum FR, Deeg HJ, Scott BL, 2012, Pretransplantation therapy with azacitidine vs induction chemotherapy and posttransplantation outcome in patients with MDS. Biol Blood Marrow Transplant 18:1211–1218 Field T, Perkins J, Huang Y, Kharfan-DabajaMA, Alsina M, Ayala E et al, 2010, 5-Azacitidine for myelodysplasia before allogeneic hematopoietic cell transplantation. Bone Marrow Transplant 45: 255–260 Cogle CR, Imanirad I, Wiggins LE, Hsu J, Brown R, Scornik JC et al, 2010, Hypomethylating agent induction therapy followed by hematopoietic cell transplantation is feasible in patients with myelodysplastic syndromes. Clin Adv Hematol Oncol 8:40– 46 Yahng SA, Yoon JH, Shin SH, Lee SE, Cho BS, Lee DG et al, 2013, Response to pretransplant hypomethylating agents influences the outcome of allogeneic hematopoietic stem cell transplantation in adults with myelodysplastic syndromes. Eur J Haematol 90:111–120 Damaj G, Duhamel A, Robin M, Milpied N, Michallet M, Chevallier P et al, 2013, Azacitidine versus best supportive care before non-myeloablative allogeneic stem cell transplantation for MDS: a study by the SFGM-TC. Leukemia Res 37(Suppl 1):S14– S15 Cheson BD, Greenberg PL, Bennett JM, Lowenberg B,Wijermans PW, Nimer SD et al, 2006, Clinical application and proposal for modification of the International Working Group, IWG, response criteria in myelodysplasia. Blood 108:419–425 Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR et al, 1976, Proposals for the classification of the acute leukaemias. French-American-British, FAB, co-operative group. Br J Haematol 33:451–458 Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G et al, 1997, International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 89:2079–2088 Giralt S, 2005, Reduced-intensity conditioning regimens for hematologic malignancies: what have we learned over the last 10 years? Hematol Am Soc Hematol Educ Program 2005:384–389 Kanda Y, 2013, Investigation of the freely available easy-to-use software ‘EZR’ for medical statistics. Bone Marrow Transplant 48(3):452–458 NCCN, 2012, Clinical practice guidelines in oncology: Myelodysplastic syndromes version 2.2014. National Comprehensive Cancer Network, Washington Sanchez-Abarca LI, Gutierrez-Cosio S, Santamaria C, Caballero- Velazquez T, Blanco B, Herrero-Sanchez C et al, 2010, Immunomodulatory effect of 5-azacytidine, 5-azaC): potential role in the transplantation setting. Blood 115:107–121 Choi J, Ritchey J, Prior JL, Holt M, Shannon WD, Deych E et al, 2010, In vivo administration of hypomethylating agents mitigate graft-versus-host disease without sacrificing graft-versus-leukemia. Blood 116:129–139 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1037 EP 1043 DI 10.1007/s12253-015-9933-8 PG 7 ER PT J AU Marrero-Rodriguez, D Taniguchi-Ponciano, K Lopez-Sleman, J Romero-Morelos, P Mendoza-Rodriguez, M Garcia, I Huerta-Padilla, V Mantilla, A Duarte, A Pina, P Rodriguez-Esquivel, M Lopez-Romero, R Parrazal-Romero, J Tobias-Alonso, S Jimenez-Vega, F Alvarez-Blanco, M Salcedo, M AF Marrero-Rodriguez, Daniel Taniguchi-Ponciano, Keiko Lopez-Sleman, Julio Romero-Morelos, Pablo Mendoza-Rodriguez, Monica Garcia, Israel Huerta-Padilla, Victor Mantilla, Alejandra Duarte, Armando Pina, Patricia Rodriguez-Esquivel, Miriam Lopez-Romero, Ricardo Parrazal-Romero, Jorge Tobias-Alonso, Salvador Jimenez-Vega, Florinda Alvarez-Blanco, Mario Salcedo, Mauricio TI Thymopoietin Beta and Gamma Isoforms as a Potential Diagnostic Molecular Marker for Breast Cancer: Preliminary Data SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thymopoietin; Isoforms; Molecular marker; Breast cancer ID Thymopoietin; Isoforms; Molecular marker; Breast cancer AB Thymopoietin (TMPO) is an inner nuclear membrane protein, the coding gene named equally, can give arise to six isoforms by alternative splicing. This gene has been found up regulated in several types of cancer. At present work, we evaluated the TMPO isoforms generated by alternative splicing as well as the protein signal detection in breast cancer samples. TMPO expression was analyzed by immunohistochemistry in tissue microarray containing 46 breast tissue samples including normal (n=6), benign lesions (n=18) (fibroadenomas (n=6), fibrocystic changes (n=6), ductal hyperplasias (n=6)) and breast carcinoma (n=22). Isoforms -α, −β and -γ of TMPO were evaluated using RT-PCR; clinical-pathological correlation analysis were done by mean of X2. Neither the normal nor the benign lesions of the breas t showed positive TMPO immunodetection, whilst 45 % of the breast carcinomas were immunopositive (p=0.000), nine of ten positives carcinomas correspond to the Luminal A subtype. Further, alpha isoform was present in all breast samples analyzed; however, beta and gamma isoforms were only present in ten (p=0.003) and 17 (p=0.000), respectively, in the breast cancer samples. According with the present data, we suggest that TMPOβ and -γ isoforms could provide a potential reliable diagnostic marker for breast cancer. C1 [Marrero-Rodriguez, Daniel] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. [Taniguchi-Ponciano, Keiko] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. [Lopez-Sleman, Julio] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. [Romero-Morelos, Pablo] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. [Mendoza-Rodriguez, Monica] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. [Garcia, Israel] Hospital de Oncologia CMN-SXXI, IMSS, Servicio de Ginecologia OncologicaMexico City, Mexico. [Huerta-Padilla, Victor] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. [Mantilla, Alejandra] Hospital de Oncologia CMN-SXXI, IMSS, Servicio de PatologiaMexico City, Mexico. [Duarte, Armando] Hospital General de Zona #35, IMSS, Cd. Juarez, Clinica de MamaChihuahua, Mexico. [Pina, Patricia] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. [Rodriguez-Esquivel, Miriam] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. [Lopez-Romero, Ricardo] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. [Parrazal-Romero, Jorge] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. [Tobias-Alonso, Salvador] Hopital General, Cd. Juarez, Departamento de PatologiaChihuahua, Mexico. [Jimenez-Vega, Florinda] Universidad Autonoma de Ciudad Juarez, Laboratorio de BiotecnologiaChihuahua, Mexico. [Alvarez-Blanco, Mario] Hospital General de Mexico, S.S., Servicio de Oncologia, Unidad de QuimioterapiaMexico City, Mexico. [Salcedo, Mauricio] Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, Av. Cuauhtemoc 330, Col. Doctores, DF 06720 Mexico City, Mexico. RP Salcedo, M (reprint author), Hospital de Oncologia CMN-SXXI. IMSS, Unidad de Investigacion Medica en Enfermedades Oncologicas, Laboratorio de Oncologia genomica, DF 06720 Mexico City, Mexico. EM maosal89@yahoo.com CR Knaul FM, Nigenda B, Lozano R, Arreola H, Langer A, Frenk J, 2008, Breast cancer in Mexico: a pressing priority. Reprod Health Matters 16:113–123 Cantile M, Pettinato G, Procino A, Feliciello I, Cindolo L, Cillo C, 2003, In vivo expression of the whole HOX gene network in human breast cancer. Eur J Cancer 39:257–264 Harris C, Andryuk P, Cline S, Mathew S, Siekerka J, Goldstein G, 1995, Structure and mapping of the human thymopoietin, TMPO, gene and relationship of human TMPO B to rat lamin-associated polypeptide 2. Genomics 28:198 Ward M, van der Watt P, Tzoneva G, Leaner V, 2011, Deregulated LAP2a expression in cervical cancer associates with aberrant E2F and p53 activities. IUBMB Life 11:1018–1026 Parise P, Finocchiaro G, Masciadri B, Quarto M, Francois S, Mancuso F et al, 2006, Lap2α expression is controlled by E2F and deregulated in various human tumors. Cell Cycle 12:1331–1341 LaTulippe E, Satagopan J, Smith A, Scher H, Scardino P, Reuter V et al, 2002, Comprehensive gene expression analysis of prostate cancer reveals distinct transcriptional programs associated with metastatic disease. Cancer Res 62:4499–506 Agrawal D, Chen T, Irby R, Quackenbush J, Chambers A, Szabo M et al, 2002, Osteopontin identified as lead marker of colon cancer progression, using pooled sample expression profiling. J Natl Cancer Inst 94:513–521 Marrero D, Peralta R, Valdivia A, De la Mora A, Romero P, Parra M et al, 2012, The neurofibromin 1 type I isoform predominance characterises female population affected by sporadic breast cancer: preliminary data. J Clin Pathol 5:419–423 Hugh J, Hanson J, Chon U, Nielsen T, Perou C, Dumontet C et al, 2009, Breast cancer subtypes and response to docetaxel in nodepositive breast cancer: use of an immunohistochemical definition in the BCIRG 001 trial. J Clin Oncol 8:1168–1176 Dorner D, Vlcek S, Foeger N, Gajewski A, Makolm C, Gotzmann J et al, 2006, Lamina-associated polypeptide 2α regulates cell cycle progression and differentiation via the retinoblastoma–E2F pathway. J Cell Biol 1:83–93 Kim H, Hwang H, HanM, Baek S, Sim H, Yoon S et al, 2012, LAP2 Is widely overexpressed in diverse digestive tract cancers and regulates motility of cancer cells. Plos One 6:e39482 Taylor MR, Slavov D, Gajewski A, Vlcek S, Ku L, Fain PR et al, 2005, Thymopoietin, Lamina-Associated Polypeptide 2, gene mutation associated with dilated cardiomyopathy. Hum Mutat 6:566– 574 Dechat T, Gotzmann J, Stockinger A, Harris C, Talle M, Siekierka J et al, 1998, Detergent-salt resistance of LAP2α in interphase nuclei and phosphorylation-dependent association with chromosomes early in nuclear assembly implies functions in nuclear structure dinamics. EMBO J 17:4887–4902 McCafferty M, Healy N, KerinM(2009, Breast cancer subtypes and molecular biomarkers. Diagn Histopathol 15:485–489 Staples E, Gasiewicz T, Fiore N, Lubahn D, Korach K, Silverstone A, 1999, Estrogen receptor is necessary in thymic development and estradiol-induced thymic alterations. J Anat 163:4168–4174 Ertel A, Dean J, Rui H, Liu C, Witkiewicz A, Knudsen KE et al, 2010, RB-pathway disruption in breast cancer: differential association with disease subtypes, disease-specific prognosis and therapeuti responsse. Cell Cycle 20:4153–4163 Zhang S, Liu S, Al-Saleem L, Holloran D, Babb J, GuoX et al, 2000, E2F-1: a proliferative marker of breast neoplasia. Cancer Epidemiol Biomarkers Prev 9:395–401 Moroy T, Heyd F, 2007, The impact of alternative splicing in vivo: mouse models show the way. RNA 13:1155–1171 Venables J, Klinck R, Bramard A, Inkel L, Dufresne-Martin G, Koh C et al, 2008, Identification of alternative splicing markers for breast cancer. Cancer Res 22:9525–9531 Vlcek S, Korbei B, Foisner R, 2002, Distinct functions of the unique C terminus of LAP2 in cell proliferation and nuclear assembly. J Biol Chem 21:18898–18907 Martins S, Eikvar S, Furukawa K, Collas P, 2003, HA95 and LAP2 mediate a novel chromatin–nuclear envelope interaction implicated in initiation of DNA replication. J Cell Biol 2:177–188 Gant T, Harris C,Wilson K, 1999, Roles of LAP2 proteins in nuclear assembly and DNA replication: truncated LAP2b proteins alter lamina assembly, envelope formation, nuclear size, and DNA replication efficiency in Xenopus laevis extracts. J Cell Biol 6:1083–1096 Dechat T, Vlcek S, Foisner R, 2000, Review: lamina-associated polypeptide 2 isoforms and related proteins in cell cycle-dependent nuclear structure dynamics. J Struct Biol 129:335–345 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1045 EP 1050 DI 10.1007/s12253-015-9907-x PG 6 ER PT J AU Lakosi, F Gulyban, Janvary, ZsL Simoni, BMS Jansen, N Seidel, L Kovacs, Vavassis, P Coucke, P AF Lakosi, Ferenc Gulyban, Akos Janvary, Zsolt Levente Simoni, Ben-Mustapha Selma Jansen, Nicolas Seidel, Laurence Kovacs, Arpad Vavassis, Peter Coucke, Philippe TI Respiratory Motion, Anterior Heart Displacement and Heart Dosimetry: Comparison Between Prone (Pr) and Supine (Su) Whole Breast Irradiation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Prone breast radiotherapy; 4D-computed tomography; Respiratory motion; Left-anterior-descending coronary artery ID Breast cancer; Prone breast radiotherapy; 4D-computed tomography; Respiratory motion; Left-anterior-descending coronary artery AB To analyze respiratory motion of surgical clips, chest wall (CW) and the anterior displacement of the heart and its impact on heart dosimetry between prone (Pr) and supine (Su) positions during whole breast radiotherapy after breast conserving surgery. Sixteen patients underwent 4D-CT for radiotherapy planning in Pr and Su positions. Maximum inhale and maximum exhale phases were analyzed. Mean 3D vectorial displacements±standard deviations (SD) of the surgical clips were measured. Volumetric changes of the CW were recorded and compared. Cardiac displacement was assessed by a volume between the inner surface of CW and the myocardium of the heart (CW/H-V). For left-sided cases, comparative dosimetry was performed in each position simulating no- (Pr-noC, Su-noC) versus daily correction protocols (Pr-C, Su-C). The movements of 81 surgical clips were analyzed. Prone positioning significantly reduced both the mean 3D vectorial displacements (1.1±0.6 (Pr) vs. 2.0±0.9 mm (Su), p<0.01) and their variability (0.3±0.2 vs. 0.5±0.3 mm, p=0.01). Respiration-induced volumetric changes of CW were also significantly lower in Pr (2.3±4.9 vs. 9.6± 7.1 cm3 , p<0.01). The CW/H-V was significantly smaller in Pr than in Su (39.9±14.6 vs. 64.3±28.2 cm3 , p<0.01). Besides identical target coverage heart, left-anterior-descending coronary artery (LADCA) and ipsilateral lung dose parameters were lowered with Pr-C compared to Pr-noC, Su-C and Su-noC. Prone position significantly reduced respirationrelated surgical clip movements, their variability as well as CW movements. Significant anterior heart displacement was observed in Pr. Prone position with daily online correction could maximize the heart and LADCA protection. C1 [Lakosi, Ferenc] University Hospital of Liege, Department of Radiation Oncology, Avenue de l’hopital, B.35, 4000 Liege, Belgium. [Gulyban, Akos] University Hospital of Liege, Department of Radiation Oncology, Avenue de l’hopital, B.35, 4000 Liege, Belgium. [Janvary, Zsolt Levente] University Hospital of Liege, Department of Radiation Oncology, Avenue de l’hopital, B.35, 4000 Liege, Belgium. [Simoni, Ben-Mustapha Selma] University Hospital of Liege, Department of Radiation Oncology, Avenue de l’hopital, B.35, 4000 Liege, Belgium. [Jansen, Nicolas] University Hospital of Liege, Department of Radiation Oncology, Avenue de l’hopital, B.35, 4000 Liege, Belgium. [Seidel, Laurence] University Hospital of Liege, Department of BiostatisticsLiege, Belgium. [Kovacs, Arpad] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. [Vavassis, Peter] Hopital Maisonneuve- RosemontQuebec, Canada. [Coucke, Philippe] University Hospital of Liege, Department of Radiation Oncology, Avenue de l’hopital, B.35, 4000 Liege, Belgium. RP Lakosi, F (reprint author), University Hospital of Liege, Department of Radiation Oncology, 4000 Liege, Belgium. EM lakosiferenc@yahoo.com;flakosi@chu.ulg.ac.be CR Darby S, McGale P, Correa C, Taylor C, Arriagada R, Clarke M et al, 2011, Effect of radiotherapy after breast-conserving surgery on 10-year recurrence and 15-year breast cancer death: metaanalysis of individual patient data for 10,801 women in 17 randomised trials. Lancet 378:1707–1716., DOI 10.1016/S0140- 6736(11)61629-2 Kovacs A, Lakosi F, Liposits G, Toller G, Hadjiev J, Vandulek C et al, 2011, 3-D conformal photon boost in the treatment of early stage breast cancer: 4 years follow up results. Pathol Oncol Res 17: 17–23., DOI 10.1007/s12253-010-9264-8 Haviland JS, Owen JR, Dewar JA, Agrawal RK, Barrett J, Barrett- Lee PJ et al, 2013, The UK Standardisation of Breast Radiotherapy, START, trials of radiotherapy hypofractionation for treatment of early breast cancer: 10-year follow-up results of two randomised controlled trials. Lancet Oncol 14:1086–1094., DOI 10.1016/S1470- 2045(13)70386-3 Polgar C, Van Limbergen E, Potter R, Kovacs G, Polo A, Lyczek J et al, 2010, Patient selection for accelerated partial-breast irradiation, APBI, after breast-conserving surgery: recommendations of the Groupe Europeen de Curietherapie-European Society for Therapeutic Radiology and Oncology, GEC-ESTRO, breast cancer working group ba. Radiother Oncol 94:264–273., DOI 10.1016/j. radonc.2010.01.014 Smith BD, Arthur DW, Buchholz TA, Haffty BG, Hahn CA, Hardenbergh PH et al, 2009, Accelerated partial breast irradiation consensus statement from the American Society for Radiation Oncology, ASTRO). Int J Radiat Oncol Biol Phys 74:987–1001., DOI 10.1016/j.ijrobp.2009.02.031 Whelan TJ, Pignol J-P, Levine MN, Julian JA, MacKenzie R, Parpia S et al, 2010, Long-term results of hypofractionated radiation therapy for breast cancer. N Engl J Med 362:513–520., DOI 10. 1056/NEJMoa0906260 FreedmanGM,White JR, ArthurDW, Allen Li X,Vicini FA, 2013, Accelerated fractionation with a concurrent boost for early stage breast cancer. Radiother Oncol 106:15–20., DOI 10.1016/j.radonc. 2012.12.001 FreedmanGM,White JR, ArthurDW, Allen Li X,Vicini FA, 2013, Accelerated fractionation with a concurrent boost for early stage breast cancer. Radiother Oncol 106:15–20., DOI 10.1016/j.radonc. 2012.12.001 Kim LH, Goyal S, Haffty BG, Taunk NK, Yue NJ, 2012, Using respiratory motion to guide planning target volume margins for external beam partial breast irradiation. Int J Radiat Oncol Biol Phys 82:1303–1306., DOI 10.1016/j.ijrobp.2011.11.048 Kirby AM, Evans PM, Donovan EM, Convery HM, Haviland JS, Yarnold JR, 2010, Prone versus supine positioning for whole and partial-breast radiotherapy: a comparison of non-target tissue dosimetry. Radiother Oncol 96:178–184., DOI 10.1016/j.radonc.2010. 05.014 Kirby AM, Evans PM, Helyer SJ, Donovan EM, Convery HM, Yarnold JR, 2011, A randomised trial of supine versus prone breast radiotherapy, SuPr study): comparing set-up errors and respiratory motion. Radiother Oncol 100:221–226., DOI 10.1016/j.radonc.2010. 11.005 Krengli M, Masini L, Caltavuturo T, Pisani C, Apicella G, Negri E et al, 2013, Prone versus supine position for adjuvant breast radiotherapy: a prospective study in patients with pendulous breasts. Radiat Oncol 8:232., DOI 10.1186/1748-717X-8-232 Lymberis SC, de Wyngaert JK, Parhar P, Chhabra AM, Fenton- Kerimian M, Chang J et al, 2012, Prospective assessment of optimal individual position, prone versus supine, for breast radiotherapy: volumetric and dosimetric correlations in 100 patients. Int J Radiat Oncol Biol Phys 84:902–909., DOI 10.1016/j.ijrobp.2012. 01.040 Mitchell J, Formenti SC, DeWyngaert JK, 2010, Interfraction and intrafraction setup variability for prone breast radiation therapy. Int J Radiat Oncol Biol Phys 76:1571–1577., DOI 10.1016/j.ijrobp. 2009.07.1683 Morrow NV, Stepaniak C,White J, Wilson JF, Li XA, 2007, Intraand interfractional variations for prone breast irradiation: an indication for image-guided radiotherapy. Int J Radiat Oncol Biol Phys 69:910–917., DOI 10.1016/j.ijrobp.2007.06.056 Mulliez T, Veldeman L, van Greveling A, Speleers B, Sadeghi S, Berwouts D et al, 2013, Hypofractionated whole breast irradiation for patients with large breasts: a randomized trial comparing prone and supine positions. Radiother Oncol 108:203–208., DOI 10.1016/j. radonc.2013.08.040 Nissen HD, Appelt AL, 2013, Improved heart, lung and target dose with deep inspiration breath hold in a large clinical series of breast cancer patients. Radiother Oncol 106:28–32., DOI 10.1016/j.radonc. 2012.10.016 Veldeman L, De GersemW, Speleers B, Truyens B, Van Greveling A, Van den Broecke R et al, 2012, Alternated prone and supine whole-breast irradiation using IMRT: setup precision, respiratory movement and treatment time. Int J Radiat Oncol Biol Phys 82: 2055–2064., DOI 10.1016/j.ijrobp.2010.10.070 Darby SC, Ewertz M, McGale P, Bennet AM, Blom-Goldman U, Bronnum D et al, 2013, Risk of ischemic heart disease in women after radiotherapy for breast cancer. N Engl J Med 368:987–998., DOI 10.1056/NEJMoa1209825 Wurschmidt F, Stoltenberg S, Kretschmer M, Petersen C, 2014, Incidental dose to coronary arteries is higher in prone than in supine whole breast irradiation. A dosimetric comparison in adjuvant radiotherapy of early stage breast cancer. Strahlenther Onkol 190: 563–568., DOI 10.1007/s00066-014-0606-4 Chino JP, Marks LB, 2008, Prone positioning causes the heart to be displaced anteriorly within the thorax: implications for breast cancer treatment. Int J Radiat Oncol Biol Phys 70:916–920., DOI 10. 1016/j.ijrobp.2007.11.001 Z.Varga, A. Cserhati, F. Rarosi,K. Boda, G. Gulyas,Z. Egyud, et al, 2013, Individualized positioning for maximum heart protection during breast irradiation. Acta Oncol. 1–7., DOI 10.3109/ 0284186X.2013.781674 van Herk M, Remeijer P, Rasch C, Lebesque JV, 2000, The probability of correct target dosage: dose-population histograms for deriving treatment margins in radiotherapy. Int J Radiat Oncol Biol Phys 47:1121–1135 FengM, Moran JM, Koelling T, Chughtai A, Chan JL, Freedman L et al, 2011, Development and validation of a heart atlas to study cardiac exposure to radiation following treatment for breast cancer. Int J Radiat Oncol Biol Phys 79:10–18., DOI 10.1016/j.ijrobp.2009. 10.058 Gulyban A, Kovacs P, Sebestyen Z, Farkas R, Csere T, Karacsonyi G et al, 2008, Multisegmented tangential breast fields: a rational way to treat breast cancer. Strahlenther Onkol 184:262–269., DOI 10. 1007/s00066-008-1770-1 Price GJ, Sharrock PJ, Marchant TE, Parkhurst JM, Burton D, Jain P et al, 2009, An analysis of breast motion using high-frequency, dense surface points captured by an optical sensor during radiotherapy treatment delivery. Phys Med Biol 54:6515–6533., DOI 10.1088/ 0031-9155/54/21/005 Kim LH, Goyal S, Haffty BG, Taunk NK, Yue NJ, 2012, Using respiratory motion to guide planning target volume margins for external beam partial breast irradiation. Int J Radiat Oncol Biol Phys 82:1303–1306., DOI 10.1016/j.ijrobp.2011.11.048 Price GJ, Sharrock PJ, Marchant TE, Parkhurst JM, Burton D, Jain P et al, 2009, An analysis of breast motion using high-frequency, dense surface points captured by an optical sensor during radiotherapy treatment delivery. Phys Med Biol 54:6515–6533., DOI 10.1088/ 0031-9155/54/21/005 Lakosi F, Ben Mustapha S, Gulyban A, Cucchiaro S, Ernst C, Martin N et al, 2014, Hypofractionated whole prone breast RT using Sagittilt system: patient comfort, setup accuracy and acute toxicity. Radiat Oncol ESTRO 33:EP–1204 Chino JP, Marks LB, 2008, Prone positioning causes the heart to be displaced anteriorly within the thorax: implications for breast cancer treatment. Int J Radiat Oncol Biol Phys 70:916–920., DOI 10. 1016/j.ijrobp.2007.11.001 Hall EJ, Wuu C-S, 2003, Radiation-induced second cancers: the impact of 3D-CRT and IMRT. Int J Radiat Oncol 56:83–88., DOI 10. 1016/S0360-3016(03)00073-7 de Puysseleyr A, Mulliez T, Gulyban A, Bogaert E, Vercauteren T, Van Hoof T et al, 2013, Improved cone-beam computed tomography in supine and prone breast radiotherapy. Surface reconstruction, radiation exposure, and clinical workflow., Strahlenther. Onkol 189:945–950., DOI 10.1007/s00066-013-0435-x NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1051 EP 1058 DI 10.1007/s12253-015-9932-9 PG 8 ER PT J AU Erdelyi-Belle, B Torok, Gy Apati, Sarkadi, B Schaff, Zs Kiss, A Homolya, L AF Erdelyi-Belle, Boglarka Torok, Gyorgy Apati, Agota Sarkadi, Balazs Schaff, Zsuzsa Kiss, Andras Homolya, Laszlo TI Expression of Tight Junction Components in Hepatocyte-Like Cells Differentiated from Human Embryonic Stem Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human embryonic stem cells; Stem cell differentiation; Cytokeratins; Tight junctions; Claudins; Tricellulin; Agrin ID Human embryonic stem cells; Stem cell differentiation; Cytokeratins; Tight junctions; Claudins; Tricellulin; Agrin AB Human embryonic stem cells can be differentiated in vitro into a wide variety of progeny cells by addition of different morphogens and growth factors. Our aim was to monitor the expression pattern of tight junction (TJ) components and various cellular markers during differentiation of stem cell lines toward the hepatic lineage. Human embryonic stem cell lines (HUES1, HUES9) were differentiated into endoderm-like cells, and further differentiated to hepatocytelike cells. Gene expressions of Oct3/4, Nanog, alpha-fetoprotein, albumin, cytokeratins (CK-7, CK-8, CK-18, CK-19), ATP-binding cassette (ABC) transporters (ABCC2, ABCC7, ABCG2), and various TJ components, including claudin-1, claudin-4, claudin-5, claudin-7, and tricellulin, as well as an extracellular matrix component, agrin were monitored during hepatic differentiation by real-time quantitative PCR. The differentiated cells exhibit epithelial morphology and functional assessments similar to that of hepatocytes. The expression level of stem cell marker genes (Oct3/4 and Nanog) significantly and gradually decreased, while liver-associated genes (alpha-fetoprotein, albumin) reached their highest expression at the end of the differentiation. The endoderm-like cells expressed claudin-1, which declined eventually. The expression levels of cholangiocyte markers including claudin-4, CK-7, CK-19, and agrin gradually increased and reached their highest level at the final stage of differentiation. In contrast, these cells did not express notable level of claudin- 7, CK-8 and tricellulin. The marker set used for monitoring differentiation revealed both hepatocyte and cholangiocyte characteristics of the differentiated cells at the final stage. This is the first report describing the expression level changes of various TJ components, and underlining their importance in hepatic differentiation. C1 [Erdelyi-Belle, Boglarka] Semmelweis University, 2nd Department of Pathology, Ulloi Street 93, 1091 Budapest, Hungary. [Torok, Gyorgy] Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute of Enzymology, Magyar tudosok korutja 2, 1117 Budapest, Hungary. [Apati, Agota] Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute of Enzymology, Magyar tudosok korutja 2, 1117 Budapest, Hungary. [Sarkadi, Balazs] Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute of Enzymology, Magyar tudosok korutja 2, 1117 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi Street 93, 1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi Street 93, 1091 Budapest, Hungary. [Homolya, Laszlo] Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute of Enzymology, Magyar tudosok korutja 2, 1117 Budapest, Hungary. RP Homolya, L (reprint author), Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary. EM homolya.laszlo@ttk.mta.hu CR Schulz TC et al, 2012, A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS One 7(5):e37004 Kirkeby A et al, 2012, Generation of regionally specified neural progenitors and functional neurons from human embryonic stem cells under defined conditions. Cell Rep 1(6):703–714 Nicoleau C et al, 2011, Human pluripotent stem cell therapy for Huntington’s disease: technical, immunological, and safety challenges human pluripotent stem cell therapy for Huntington’s disease: technical, immunological, and safety challenges. Neurotherapeutics 8(4):562–576 Moon SH et al, 2011, A system for treating ischemic disease using human embryonic stem cell-derived endothelial cells without direct incorporation. Biomaterials 32(27):6445–6455 Murry CE, Keller G, 2008, Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell 132(4):661–680 Vosough M et al, 2011, Cell-based therapeutics for liver disorders. Br Med Bull 100:157–172 Lodi C et al, 2006, Claudin-4 differentiates biliary tract cancers from hepatocellular carcinomas. Mod Pathol 19(3):460–469 Nemeth Z et al, 2009, Claudin-1, -2, -3, -4, -7, -8, and -10 protein expression in biliary tract cancers. J Histochem Cytochem 57(2): 113–121 Kinugasa T et al, 2012, Increased claudin-1 protein expression in hepatic metastatic lesions of colorectal cancer. Anticancer Res 32(6):2309–2314 Van Eyken P et al, 1988, The development of the intrahepatic bile ducts in man: a keratin-immunohistochemical study. Hepatology 8(6):1586–1595 Paku S et al, 2005, Immunohistochemical analysis of cytokeratin 7 expression in resting and proliferating biliary structures of rat liver. Hepatology 42(4):863–870 Vestentoft PS et al, 2011, Three-dimensional reconstructions of intrahepatic bile duct tubulogenesis in human liver. BMC Dev Biol 11:56 Dezso K et al, 2009, Architectural and immunohistochemical characterization of biliary ductules in normal human liver. Stem Cells Dev 18(10):1417–1422 Mayer R et al, 1995, Expression of the MRP gene-encoded conjugate export pump in liver and its selective absence from the canalicular membrane in transport-deficient mutant hepatocytes. J Cell Biol 131(1):137–150 Bodo A et al, 2003, Differential modulation of the human liver conjugate transporters MRP2 and MRP3 by bile acids and organic anions. J Biol Chem 278(26):23529–23537 Fitz JG et al, 1993, Regulation ofmembrane chloride currents in rat bile duct epithelial cells. J Clin Invest 91(1):319–328 Guo Y, Lubbert M, Engelhardt M, 2003, CD34- hematopoietic stem cells: current concepts and controversies. Stem Cells 21(1): 15–20 Apati A et al, 2008, High level functional expression of the ABCG2 multidrug transporter in undifferentiated human embryonic stem cells. Biochim Biophys Acta 1778(12):2700–2709 Sarkadi B et al, 2010, Evaluation of ABCG2 expression in human embryonic stem cells: crossing the same river twice? Stem Cells 28(1):174–176 Padmanabhan R et al, 2012, Regulation and expression of the ATPbinding cassette transporter ABCG2 in human embryonic stem cells. Stem Cells 30(10):2175–2187 Furuse M, Sasaki H, Tsukita S, 1999, Manner of interaction of heterogeneous claudin species within and between tight junction strands. J Cell Biol 147(4):891–903 Amasheh S et al, 2005, Contribution of claudin-5 to barrier properties in tight junctions of epithelial cells. Cell Tissue Res 321(1): 89–96 Hewitt KJ, Agarwal R, Morin PJ, 2006, The claudin gene family: expression in normal and neoplastic tissues. BMC Cancer 6:186 Mineta K et al, 2011, Predicted expansion of the claudin multigene family. FEBS Lett 585(4):606–612 Furuse M et al, 1998, Claudin-1 and -2: novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol 141(7):1539–1550 Riazuddin S et al, 2006, Tricellulin is a tight-junction protein necessary for hearing. Am J Hum Genet 79(6):1040–1051 Patonai A et al, 2011, Claudins and tricellulin in fibrolamellar hepatocellular carcinoma. Virchows Arch 458(6):679–688 Grosse B et al, 2012, Claudin-1 involved in neonatal ichthyosis sclerosing cholangitis syndrome regulates hepatic paracellular permeability. Hepatology 55(4):1249–1259 Bunthot S et al, 2012, Overexpression of claudin-4 in cholangiocarcinoma tissues and its possible role in tumor metastasis. Asian Pac J Cancer Prev 13(Suppl):71–76 Jakab C et al, 2010, Claudin-7 protein differentiates canine cholangiocarcinoma from hepatocellular carcinoma. Histol Histopathol 25(7):857–864 Schulzke JD, FrommM(2009, Tight junctions: molecular structure meets function. Ann N YAcad Sci 1165:1–6 Forster C, 2008, Tight junctions and the modulation of barrier function in disease. Histochem Cell Biol 130(1):55–70 Tatrai P et al, 2006, Agrin, a novel basementmembrane component in human and rat liver, accumulates in cirrhosis and hepatocellular carcinoma. Lab Invest 86(11):1149–1160 Batmunkh E et al, 2007, Comparison of the expression of agrin, a basement membrane heparan sulfate proteoglycan, in cholangiocarcinoma and hepatocellular carcinoma. Hum Pathol 38(10):1508– 1515 Higashi Yet al, 2007, Loss of claudin-1 expression correlates with malignancy of hepatocellular carcinoma. J Surg Res 139(1):68–76 Sobel G et al, 2005, Increased expression of claudins in cervical squamous intraepithelial neoplasia and invasive carcinoma. Hum Pathol 36(2):162–169 Jakab C et al, 2010, Expression of claudin-1, -3, -4, -5 and -7 proteins in low grade colorectal carcinoma of canines. Histol Histopathol 25(1):55–62 Miettinen M, Sarlomo-Rikala M,Wang ZF, 2011, Claudin-5 as an immunohistochemical marker for angiosarcoma and hemangioendotheliomas. Am J Surg Pathol 35(12):1848–1856 Brokalaki EI et al, 2012, Claudin-7 expression in hepatocellular carcinoma. Transplant Proc 44(9):2737–2740 Ersoz S et al, 2011, Prognostic importance of Claudin-1 and Claudin-4 expression in colon carcinomas. Pathol Res Pract 207(5):285–289 Tsutsumi K et al, 2012, Claudin-4 expression predicts survival in pancreatic ductal adenocarcinoma. Ann Surg Oncol 19(Suppl 3): S491–S499 Agarwal R, D’Souza T, Morin PJ, 2005, Claudin-3 and claudin-4 expression in ovarian epithelial cells enhances invasion and is associated with increased matrix metalloproteinase-2 activity. Cancer Res 65(16):7378–7385 Lappi-Blanco E et al, 2013, Divergence of tight and adherens junction factors in alveolar epithelium in pulmonary fibrosis. Hum Pathol 44(5):895–907 Resnick MB et al, 2005, Claudin-1 is a strong prognostic indicator in stage II colonic cancer: a tissue microarray study. Mod Pathol 18(4):511–518 Kominsky SL, 2006, Claudins: emerging targets for cancer therapy. Expert Rev Mol Med 8(18):1–11 Dezso K et al, 2012, Structural analysis of oval-cell-mediated liver regeneration in rats. Hepatology 56(4):1457–1467 Mateizel I et al, 2012, Establishment of hESC lines from the inner cell mass of blastocyst-stage embryos and single blastomeres of 4- cell stage embryos. Methods Mol Biol 873:81–112 Xu C et al, 2001, Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol 19(10):971–974 Chen Yet al, 2006, Instant hepatic differentiation of human embryonic stem cells using activin A and a deleted variant of HGF. Cell Transplant 15(10):865–871 Cai J et al, 2007, Directed differentiation of human embryonic stem cells into functional hepatic cells. Hepatology 45(5):1229–1239 Baharvand H et al, 2006, Differentiation of human embryonic stem cells into hepatocytes in 2D and 3D culture systems in vitro. Int J Dev Biol 50(7):645–652 Vosough M et al, 2013, Generation of functional hepatocyte-like cells from human pluripotent stem cells in a scalable suspension culture. Stem Cells Dev 22(20):2693–2705 Rambhatla L et al, 2003, Generation of hepatocyte-like cells from human embryonic stem cells. Cell Transplant 12(1):1–11 Turksen K, 2011, Claudins and cancer stem cells. Stem Cell Rev 7(4):797–798 Sell S, Leffert HL, 2008, Liver cancer stem cells. J Clin Oncol 26(17):2800–2805 Halasz J et al, 2006, Claudin-1 and claudin-2 differentiate fetal and embryonal components in human hepatoblastoma. Hum Pathol 37(5):555–561 Adewumi O et al, 2007, Characterization of human embryonic stem cell lines by the international stem cell initiative. Nat Biotechnol 25(7):803–816 Erdei Z et al, 2013, Dynamic ABCG2 expression in human embryonic stem cells provides the basis for stress response. Eur Biophys J 42(2–3):169–179 Papp V et al, 2013, Expansion of hepatic stem cell compartment boosts liver regeneration. Stem Cells Dev Basma H et al, 2009, Differentiation and transplantation of human embryonic stem cell-derived hepatocytes. Gastroenterology 136(3): 990–999 Thomson JA et al, 1998, Embryonic stem cell lines derived from human blastocysts. Science 282(5391):1145–1147 Silva J, Smith A, 2008, Capturing pluripotency. Cell 132(4):532– 536 Osafune K et al, 2008, Marked differences in differentiation propensity among human embryonic stem cell lines. Nat Biotechnol 26(3):313–315 Turanyi E et al, 2010, Immunohistochemical classification of ductular reactions in human liver. Histopathology 57(4):607–614 Tsukita S et al, 2008, Tight junction-based epithelial microenvironment and cell proliferation. Oncogene 27(55):6930–6938 Zadori G et al, 2011, Examination of claudin-1 expression in patients undergoing liver transplantation owing to hepatitis C virus cirrhosis. Transplant Proc 43(4):1267–1271 Farquhar MJ et al, 2012, Hepatitis C virus induces CD81 and claudin-1 endocytosis. J Virol 86(8):4305–4316 Jarmund T, Telle W, 1982, Binding of Clostridium perfringens enterotoxin to hepatocytes, small intestinal epithelial cells and Vero cells.Acta PatholMicrobiol Immunol ScandB 90(5):377–381 Korompay A et al, 2012, Tricellulin expression in normal and neoplastic human pancreas. Histopathology 60(6B):E76–E86 Tatrai P et al, 2009, Agrin and CD34 immunohistochemistry for the discrimination of benign versus malignant hepatocellular lesions. Am J Surg Pathol 33(6):874–885 Somoracz A et al, 2010, Agrin immunohistochemistry facilitates the determination of primary versus metastatic origin of liver carcinomas. Hum Pathol 41(9):1310–1319 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1059 EP 1070 DI 10.1007/s12253-015-9936-5 PG 12 ER PT J AU Carmignani, L Macchi, A Ratti, D Finkelberg, E Casellato, S Bozzini, G Maruccia, S Marenghi, C Picozzi, S AF Carmignani, Luca Macchi, Alberto Ratti, Dario Finkelberg, Elisabetta Casellato, Stefano Bozzini, Giorgio Maruccia, Serena Marenghi, Carlo Picozzi, Stefano TI Are Histological Findings of Thulium Laser Vapo-Enucleation Versus Transurethral Resection of the Prostate Comparable? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate; Benign prostatic hypertrophy; Transurethral; Thulium; Laser ID Prostate; Benign prostatic hypertrophy; Transurethral; Thulium; Laser AB We investigated if an adequate histological diagnosis can be made from tissue after Thulium laser vapoenucleation of the prostate (ThuVEP) and whether it is comparable to transurethral prostate resection (TURP) tissue findings in patients with symptomatic benign prostatic hyperplasia. We analyzed 350 ThuLEP and 100 matched TURP tissue specimens from patients who underwent one of the two procedures between January 2009 and June 2014. Thulium Laser Enucleation of Prostate (ThuVEP) was combined with mechanical morcellation of the resected lobe. Each histological specimen was reviewed by two pathologists. Preoperative prostate ultrasound volume, total serum prostatic specific antigen and postoperative tissue weight were evaluated. Microscopic histological diagnosis was assessed by standard histological techniques and immunohistochemical evaluation. Patients were comparable in terms of age and preoperative total serum prostate specific antigen. Incidental adenocarcinoma and high grade PIN of the prostate were diagnosed in a comparable percent of specimens in the 2 groups (2.5 % in the ThuVEP group versus 3 % in the TURP group). Tissue thermal artifacts induced by the Thulium laser are mostly due to coagulation as that of the conventional monopolar diathermy in TURP. Tissue quality was maintained in the ThuVEP histological specimens. Tissue maintain histological characteristics and proprieties without modification for successive immunoistochemical analysis. The pathologist ability to detect incidental prostate cancer and PIN was maintained even if there is a quoted of vaporized tissue. C1 [Carmignani, Luca] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Macchi, Alberto] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Ratti, Dario] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Finkelberg, Elisabetta] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Casellato, Stefano] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Bozzini, Giorgio] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Maruccia, Serena] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Marenghi, Carlo] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. [Picozzi, Stefano] IRCCS Policlinico San Donato, University of Milan, Urology Department, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy. RP Picozzi, S (reprint author), IRCCS Policlinico San Donato, University of Milan, Urology Department, 20097 San Donato Milanese, Italy. EM stepico@tin.it CR Suhani, Gupta S, Gupta A, Saha S, Mahapatra L, Srivastava U, 2013, Outcome of surgery for benign prostatic hyperplasia-is it predictable? J Clin Diagn Res 7:2859–2862 Oelke M, Bachmann A, Descazeaud A, Emberton M, Gravas S, Michel MC, N’dow J, Nordling J, de la Rosette JJ, European Association of Urology, 2013, EAU guidelines on the treatment and follow-up of non-neurogenic male lower urinary tract symptoms including benignprostatic obstruction. Eur Urol 64:118–140 Herrmann TR, Liatsikos EN, Nagele U, Traxer O, Merseburger AS, EAU Guidelines Panel onLasers, Technologies, 2012, EAU guidelines on laser technologies. Eur Urol 61:783–795 Xu Y, Sun D, Wei Z, Hong B, Yang Y, 2013, Clinical study on the application of a 2-μ m continuous wave laser in transurethral vaporesection of the prostate. Exp Ther Med 5:1097–1100 Herrmann TR, Bach T, Imkamp F, Georgiou A, Burchardt M, Oelke M, Gross AJ, 2010, Thulium laser enucleation of the prostate, ThuLEP): transurethral anatomical prostatectomy withlaser support. Introduction of a novel technique for the treatment of benign prostatic obstruction. World J Urol 28: 45–51 Mebust WK, Holtgrewe HL, Cockett ATK, Peters PC, and Writing Committee, 1989, Transurethral prostatectomy: immediate and postoperative complications. A cooperative study of 13 participating institutions evaluating 3,885 patients. J Urol 141:243 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1071 EP 1075 DI 10.1007/s12253-015-9931-x PG 5 ER PT J AU Liu, P Jiang, W Ren, H Zhang, H Hao, J AF Liu, Pengfei Jiang, Wenhua Ren, He Zhang, Huilai Hao, Jihui TI Exploring the Molecular Mechanism and Biomakers of Liver Cancer Based on Gene Expression Microarray SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Liver cancer; Bioinformatics; Biomarker; Pathway ID Liver cancer; Bioinformatics; Biomarker; Pathway AB Liver cancer is one of the most common cancers worldwide with high morbidity and mortality. Its molecular mechanism hasn’t been fully understood though many studies have been conducted and thus further researches are still needed to improve the prognosis of liver cancer. Firstly, differentially expressed genes (DEGs) between six Mdr2-knockout (Mdr2- KO) mutant mice samples (3-month-old and 12-month-old) and six control mice samples were identified. Then, the enriched GO terms and KEGG pathways of those DEGs were obtained using the Database for Annotation, Visualization and Integrated Discovery (DAVID, http://david.abcc.ncifcrf.gov/). Finally, protein-protein interactions (PPI) network of those DEGs were constructed using STRING database (http://www.string-db.org/) and visualized by Cytoscape software, at the same time, genes with high degree were selected out. Several novel biomarkers that might play important roles in liver cancer were identified through the analysis of gene microarray in GEO. Also, some genes such as Tyrobp, Ctss and pathways such as Pathways in cancer, ECM-receptor interaction that had been researched previously were further confirmed in this study. Through the bioinformatics analysis of the gene microarray in GEO, we found some novel biomarkers of liver cancer and further confirmed some known biomarkers. C1 [Liu, Pengfei] National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Department of Lymphoma, Sino-US Center of Lymphoma and LeukemiaTianjin, China. [Jiang, Wenhua] The Second Hospital of Tianjin Medical University, Department of RadiotherapyTianjin, China. [Ren, He] National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Department of Pancreatic CancerTianjin, China. [Zhang, Huilai] National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Department of Lymphoma, Sino-US Center of Lymphoma and LeukemiaTianjin, China. [Hao, Jihui] National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Department of Pancreatic CancerTianjin, China. RP Hao, J (reprint author), National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Department of Pancreatic Cancer, Tianjin, China. EM haojihui@tjmuch.com CR Bertino G, Demma S, Ardiri A, Proiti M, Gruttadauria S, Toro A, Malaguarnera G, Bertino N, Malaguarnera M, Di Carlo I, 2014, Hepatocellular carcinoma: novel molecular targets in carcinogenesis for future therapies. Biomed Res Int 2014, 203693 JianXin J, Cha Y, ZhiPeng L, Jie X, Hao Z, Meiyuan C, ChengYi S, 2014, GOLP3 is a predictor of survival in patients with hepatocellular carcinoma. Clin Invest Med 37(4):E233 MahWC, Thurnherr T, Chow PK, Chung AY, Ooi LL, Toh HC, Teh BT, Saunthararajah Y, Lee CG, 2014, Methylation profiles reveal distinct subgroup of hepatocellular carcinoma patients with poor prognosis. PLoS One 9(8):e104158 Smit JJ, Schinkel AH, Oude Elferink RP, Groen AK, Wagenaar E, van Deemter L, Mol CA, Ottenhoff R, van der Lugt NM, van Roon MA et al, 1993, Homozygous disruption of the murine mdr2 Pglycoprotein gene leads to a complete absence of phospholipid from bile and to liver disease. Cell 75(3):451–462 Katzenellenbogen M, Mizrahi L, Pappo O, Klopstock N, Olam D, Barash H, Domany E, Galun E, Goldenberg D, 2007, Molecular mechanisms of the chemopreventive effect on hepatocellular carcinoma development in Mdr2 knockout mice. Mol Cancer Ther 6(4): 1283–1291 Yang YM, Lee WH, Lee CG, An J, Kim ES, Kim SH, Lee SK, Lee CH, Dhanasekaran DN, Moon A, Hwang S, Lee SJ, Park JW, Kim KM, Kim SG, 2014, Galpha gep oncogene deregulation of p53- responsive microRNAs promotes epithelial-mesenchymal transition of hepatocellular carcinoma. Oncogene 0 Xu X, Huang P, Yang B, Wang X, Xia J, 2014, Roles of CXCL5 on migration and invasion of liver cancer cells. J Transl Med 12:193 Katzenellenbogen M, Pappo O, Barash H, Klopstock N, Mizrahi L, Olam D, Jacob-Hirsch J, Amariglio N, Rechavi G, Mitchell LA, Kohen R, Domany E, Galun E, Goldenberg D, 2006, Multiple adaptive mechanisms to chronic liver disease revealed at early stages of liver carcinogenesis in the Mdr2-knockout mice. Cancer Res 66(8): 4001–4010 Gautier L, Cope L, Bolstad BM, Irizarry RA, 2004, Affy–analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 20(3): 307–315 Diboun I, Wernisch L, Orengo CA, Koltzenburg M, 2006, Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma. BMC Genomics 7:252 Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T, 2003, Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13(11):2498–2504 Chefetz I, Alvero AB, Holmberg JC, Lebowitz N, Craveiro V, Yang- Hartwich Y, Yin G, Squillace L, Gurrea Soteras M, Aldo P, Mor G, 2013, TLR2 enhances ovarian cancer stem cell self-renewal and promotes tumor repair and recurrence. Cell Cycle 12(3):511–521 Usami Y, Ishida K, Sato S, Kishino M, KiryuM, Ogawa Y, Okura M, Fukuda Y, Toyosawa S, 2013, Intercellular adhesion molecule-1, ICAM-1, expression correlates with oral cancer progression and induces macrophage/cancer cell adhesion. Int J Cancer 133(3):568– 578 Ciftci R, Tas F, Yasasever CT, Aksit E, Karabulut S, Sen F, Keskin S, Kilic L, Yildiz I, Bozbey HU, Duranyildiz D, Vatansever S, 2014, High serum transforming growth factor beta 1, TGFB1, level predicts better survival in breast cancer. Tumour Biol 35(7):6941–6948 Liu YY, Han JY, Lin SC, Liu ZY, Jiang WT, 2014, Effect of CDH1 gene methylation on transforming growth factor, TGF-beta)-induced epithelial-mesenchymal transition in alveolar epithelial cell line A549. Genet Mol Res 13(4):8568–8576 Huang CY, FongYC, Lee CY, ChenMY, Tsai HC,HsuHC, TangCH, 2009, CCL5 increases lung cancer migration via PI3K, Akt and NFkappaB pathways. Biochem Pharmacol 77(5):794–803 Leavy O, 2014, Innate immune signalling: TIRAP diversifies the sites of TLR signalling. Nat Rev Immunol 14(4):211 Medeiros MC, Frasnelli SC, Bastos Ade S, Orrico SR, Rossa C, 2014)Modulation of cell proliferation, survival and gene expression by RAGE and TLR signaling in cells of the innate and adaptive immune response: role of p38 MAPK and NF-KB. J Appl Oral Sci 22(3):185–193 Kos J, Sekirnik A, Kopitar G, Cimerman N, Kayser K, Stremmer A, Fiehn W, Werle B, 2001, Cathepsin S in tumours, regional lymph nodes and sera of patients with lung cancer: relation to prognosis. Br J Cancer 85(8):1193–1200 Lee TK, Cheung VC, Lu P, Lau EY, Ma S, Tang KH, Tong M, Lo J, Ng IO, 2014, Blockade of CD47-mediated cathepsin S/proteaseactivated receptor 2 signaling provides a therapeutic target for hepatocellular carcinoma. Hepatology 60(1):179–191 Shabo I, Svanvik J, 2011, Expression of macrophage antigens by tumor cells. Adv Exp Med Biol 714:141–150 Shabo I, Olsson H, Stal O, Svanvik J, 2013, Breast cancer expression of DAP12 is associated with skeletal and liver metastases and poor survival. Clin Breast Cancer 13(5):371–377 Costa VL, Henrique R, Danielsen SA, Duarte-Pereira S, Eknaes M, Skotheim RI, Rodrigues A, Magalhaes JS, Oliveira J, Lothe RA, Teixeira MR, Jeronimo C, Lind GE, 2010, Three epigenetic biomarkers, GDF15, TMEFF2, and VIM, accurately predict bladder cancer from DNA-based analyses of urine samples. Clin Cancer Res 16(23):5842–5851 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1077 EP 1083 DI 10.1007/s12253-015-9926-7 PG 7 ER PT J AU Cates, MMJ AF Cates, M M Justin TI Prognostic factors for second recurrence after surgical resection of recurrent desmoid-type fibromatosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Aggressive fibromatosis; Local recurrence; Prognosis; Second recurrence ID Aggressive fibromatosis; Local recurrence; Prognosis; Second recurrence AB The optimal management of recurrent desmoidtype fibromatosis is undefined. This study was performed to determine what factors, if any, predict second recurrence after surgical resection of recurrent desmoid tumors. Of 41 patients with recurrent desmoid-type fibromatosis, 29 underwent surgical resection and 8 were followed by observation. Four others received adjuvant chemo/pharmacotherapy. Clinicopathologic features were assessed as prognostic factors for second recurrence after surgical resection of recurrent desmoid tumors by Cox proportional hazards regression. Nine of 29 patients who underwent surgical resection of recurrent desmoid tumor developed a second recurrence. Larger size of recurrent tumor was associated with increased risk of second recurrence (hazard ratio, 1.09; P=0.006). Treatment of the primary tumor with adjuvant radiation therapy also increased risk of re-recurrence (3.41; P=0.032). Second recurrence of desmoid-type fibromatosis after surgical resection is, much like recurrence of primary desmoid tumor, difficult to predict with current prognostic indicators. C1 [Cates, M M Justin] Vanderbilt University Medical Center and Vanderbilt University, School of Medicine, Department of Pathology, CC-3322, 1161 21st Ave. South, 37232 Nashville, TN, USA. RP Cates, MMJ (reprint author), Vanderbilt University Medical Center and Vanderbilt University, School of Medicine, Department of Pathology, 37232 Nashville, USA. EM justin.m.cates@vanderbilt.edu CR National Comprehensive Cancer Network Soft Tissue Sarcoma, Version 2.2014). http://www.Nccn.Org/Professionals/Physician_ Gls/Pdf/Sarcoma.Pdf. Accessed 20 March 2014 Ballo MT, Zagars GK, Pollack A, Pisters PW, Pollack RA, 1999, Desmoid tumor: prognostic factors and outcome after surgery, radiation therapy, or combined surgery and radiation therapy. J Clin Oncol 17:158–167 Lev D, Kotilingam D, Wei C, Ballo MT, Zagars GK, Pisters PW, Lazar AA, Patel SR, Benjamin RS, Pollock RE, 2007, Optimizing treatment of desmoid tumors. J Clin Oncol 25:1785–1791 Gluck I, Griffith KA, Biermann JS, Feng FY, Lucas DR, Ben-Josef E, 2011, Role of radiotherapy in the management of desmoid tumors. Int J Radiat Oncol Biol Phys 80:787–792 Prodinger PM, Rechl H, Keller M, Pilge H, Salzmann M, Von Eisenhart-Rothe R, Holzapfel BM, 2013, Surgical resection and radiation therapy of desmoid tumours of the extremities: results of a supra-regional tumour centre. Int Orthop 37:1987–1993 Mullen JT, Delaney TF, Kobayashi WK, Szymonifka J, Yeap BY, Chen YL, Rosenberg AE, Harmon DC, Choy E, Yoon SS, Raskin KA, Nielsen GP, Hornicek FJ, 2012, Desmoid tumor: analysis of prognostic factors and outcomes in a surgical series. Ann Surg Oncol 19:4028–4035 Nuyttens JJ, Rust PF, Thomas CR Jr, Turrisi AT, 2000, Surgery versus radiation therapy for patients with aggressive fibromatosis or desmoid tumors: a comparative review of 22 articles. Cancer 88: 1517–1523 Bonvalot S, Eldweny H, Haddad V, Rimareix F, Missenard G, Oberlin O, Vanel D, Terrier P, Blay JY, Le Cesne A, Le Pechoux C, 2008, Extra-abdominal primary fibromatosis: aggressive management could be avoided in a subgroup of patients. Eur J Surg Oncol 34:462–468 Gronchi A, Casali PG, Mariani L, Lo Vullo S, Colecchia M, Lozza L, Bertulli R, Fiore M,Olmi P, SantinamiM, Rosai J, 2003, Quality of surgery and outcome in extra-abdominal aggressive fibromatosis: a series of patients surgically treated at a single institution. J Clin Oncol 21:1390–1397 Peng PD, Hyder O, Mavros MN, Turley R, Groeschl R, Firoozmand A, Lidsky M, Herman JM, Choti M, Ahuja N, Anders R, Blazer DG III, Gamblin TC, Pawlik TM, 2012, Management and recurrence patterns of desmoids tumors: a multi-institutional analysis of 211 patients. Ann Surg Oncol 19: 4036–4042 Fiore M, Rimareix F, Mariani L, Domont J, Collini P, Le Pechoux C, Casali PG, Le Cesne A, Gronchi A, Bonvalot S, 2009, Desmoid-type fibromatosis: a front-line conservative approach to select patients for surgical treatment. Ann Surg Oncol 16:2587–2593 Huber PJ, 1967, The behavior of maximum likelihood estimates under nonstandard conditions, vol 1. Proceedings of the Fifth Berkeley Symposium on Mathematical Statistics and Probability. University Of California Press, Berkeley White HL Jr, 1980, A heteroskedasticity-consistent covariance matrix estimator and a direct test for heteroskedasticity. Econometrica 48:817–838 Cates JM, Stricker TP, 2014, Surgical resection margins in desmoid-type fibromatosis: a critical reassessment. Am J Surg Pathol 38:1707–1714 Crago AM, Denton B, Salas S, Dufresne A, Mezhir JJ, Hameed M, Gonen M, Singer S, Brennan MF, 2013, A prognostic nomogram for prediction of recurrence in desmoid fibromatosis. Ann Surg 258: 347–353 Cates JM, Stricker TP, Sturgeon D, Coffin CM, 2014, Desmoidtype fibromatosis-associated gardner fibromas: prevalence and impact on local recurrence. Cancer Lett 353:176–181 Salas S, Dufresne A, Bui B, Blay JY, Terrier P, Ranchere-Vince D, Bonvalot S, Stoeckle E, Guillou L, Le Cesne A, Oberlin O, Brouste V, Coindre JM, 2011, Prognostic factors influencing progressionfree survival determined from a series of sporadic desmoid tumors: a wait-and-see policy according to tumor presentation. J Clin Oncol 29:3553–3558 Lewis JJ, Boland PJ, Leung DH, Woodruff JM, Brennan MF, 1999, The enigma of desmoid tumors. Ann Surg 229:866– 872 Stoeckle E, Coindre JM, Longy M, Binh MB, Kantor G, Kind M, De Lara CT, Avril A, Bonichon F, Bui BN, 2009, A critical analysis of treatment strategies in desmoid tumours: a review of a series of 106 cases. Eur J Surg Oncol 35:129–134 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1085 EP 1090 DI 10.1007/s12253-015-9939-2 PG 6 ER PT J AU Lerant, G Sarkozy, P Takacsi-Nagy, Z Polony, G Tamas, L Toth, E Boer, A Javor, L Godeny, M AF Lerant, Gergely Sarkozy, Peter Takacsi-Nagy, Zoltan Polony, Gabor Tamas, Laszlo Toth, Erika Boer, Andras Javor, Laszlo Godeny, Maria TI Dynamic Contrast-Enhanced MRI Parameters as Biomarkers in Assessing Head and Neck Lesions After Chemoradiotherapy Using a Wide-Bore 3 Tesla Scanner SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DCE-MRI; Head-and neck tumors; Post-treatment imaging; Chemoradiotherapy; Time intensity curve; Biomarker ID DCE-MRI; Head-and neck tumors; Post-treatment imaging; Chemoradiotherapy; Time intensity curve; Biomarker AB Pilot studies have shown promising results in characterizing head and neck tumors (HNT) using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), differentiating between malignant and benign lesions and evaluating changes in response to chemoradiotherapy (CRT). Our aim was to find DCE-MRI parameters, biomarkers in evaluating the post-CRT status. Two hundred and five patients with head and neck lesions were examined with DCE-MRI sequences. The time intensity curves (TIC) were extracted and processed to acquire time-to-peak (TTP), relative maximum enhancement (RME), relative wash-out (RWO), and two new parameters attack and decay. These parameters were analyzed using univariate tests in SPSS (Statistical Package for the Social Sciences, version 17, SPSS Inc. Chicago, USA) to identify parameters that could be used to infer tumor malignancy and post-CRT changes. Multiple parameters of curve characteristics were significantly different between malignant tumors after CRT (MACRT) and changes caused by CRT. The best-performing biomarkers were the attack and the decay. We also found multiple significant (p<0.05) parameters for both the benign and malignant status as well as pre- and post-CRT status. Our large cohort of data supports the increasing role of DCE-MRI in HNT differentiation, particularly for the assessment of post-CRT status along with accurate morphological imaging. C1 [Lerant, Gergely] National Institute of Oncology, Center of Radiology, Rath Gyorgy Street 7-9, 1122 Budapest, Hungary. [Sarkozy, Peter] Budapest University of Technology, Department of Measurement and Information systemsBudapest, Hungary. [Takacsi-Nagy, Zoltan] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Polony, Gabor] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Tamas, Laszlo] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Toth, Erika] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Boer, Andras] National Institute of Oncology, Multidisciplinary Center of Head and Neck OncologyBudapest, Hungary. [Javor, Laszlo] National Institute of Oncology, Center of Radiology, Rath Gyorgy Street 7-9, 1122 Budapest, Hungary. [Godeny, Maria] National Institute of Oncology, Center of Radiology, Rath Gyorgy Street 7-9, 1122 Budapest, Hungary. RP Lerant, G (reprint author), National Institute of Oncology, Center of Radiology, 1122 Budapest, Hungary. EM lerger@freemail.hu CR Furukawa M, Parvathaneni U, Maravilla K, Richards TL, Anzai Y, 2013, Dynamic contrast-enhanced MR perfusion imaging of head and neck tumors at 3 Tesla. Head Neck 35:923–929 Yuan Y, Yue XH, Tao XF, 2012, The diagnostic value of dynamic contrast-enhanced MRI for thyroid tumors. Eur J Radiol 81:3313– 3318 Kim S, Loevner LA, Quon H, Kilger A, Sherman E, Weinstein G, Chalian A, Poptani H, 2010, Prediction of response to chemoradiation therapy in squamous cell carcinomas of the head and neck using dynamic contrast-enhanced MR imaging. AJNR Am J Neuroradiol 31:262–268 Godeny M, 2014, Prognostic factors in advanced pharyngeal and oral cavity cancer; significance of multimodality imaging in terms of 7th edition of TNM. Cancer Imaging 14:1–13 Gregorie V, Lefebvre J, Licitra L, Felip E, 2010, Squamous cell carcinoma of the head and neck: EHNS-ESMO-ESTRO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 21(5):v184–v186 Hermans R, De Keyzer F, Vandecaveye V, Carp L, 2012, Head and neck cancer imaging. Springer, Berlin/Heidelberg, Germany Licitra L, Felip E, ESMO Guidelines Working Group, 2009, Squamous cell carcinoma of the head and neck: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol 20:iv121–iv122 Hermans R, 2006, Head and neck cancer: how imaging predicts treatment outcome. Cancer Imaging 6:S145–S153 Hermans R, 2008, Posttreatment imaging in head and neck cancer. Eur J Radiol 66:501–511 Saito N, Nadgir RN, Nakahira M, TakahashiM, Uchino A, Kimura F, Truong MT, Sakai O, 2012, Posttreatment CT and MR imaging in head and neck cancer: what the radiologist needs to know. Radiographics 32:1261–1282 Baba Y, Furusawa M, Murakami R, Yokoyama T, Sakamoto Y, Nishimura R, Yamashita Y, Takahashi M, Ishikawa T, 1997, Role of dynamic MRI in the evaluation of head and neck cancers treated with radiation therapy. Int J Radiat Oncol Biol Phys 37:783–787 Baba Y, Yamashita Y, Onomichi M, 2002, Dynamic MR imaging and radiotherapy. Magn Reson Med Sci 1:32–37 Semiz Oysu A, Ayanoglu E, Kodalli N, Oysu C, Uneri C, Erzen C, 2005, Dynamic contrast-enhanced MRI in the differentiation of posttreatment fibrosis from recurrent carcinoma of the head and neck. Clin Imaging 29:307–312 Rashed L et al., 2010, The added value of DWI MRI in characterization of tissue in treated head and neck tumors. Egypt J Ear Nose Throat 11 Chikui T et al., 2012, The principal of dynamic contrast enhanced MRI, themethod of pharmacokinetic analysis, and its application in the head and neck region Int J Dent Article ID 480659 http:// dx.doi.org/10.1155/2012/480659 Sumi M, Nakamura T, 2014, Head and neck tumours: combined MRI assessment based on IVIM and TIC analyses for the differentiation of tumors of different histological types. Eur Radiol 24:223– 231 Andrade RS, Heron DE, Degirmenci B, Filho PA, Branstetter BF, Seethala RR, Ferris RL, Avril N, 2006, Posttreatment assessment of response using FDG-PET/CT for patients treated with definitive radiation therapy forhead and neck cancers. Int J Radiat Oncol Biol Phys 65:1315–1322 Dunsky KA, Wehrmann DJ, Osman MM, Thornberry BM, Varvares MA, 2013, PET-CT and the detection of the asymptomatic recurrence or second primary lesions in the treated head and neck cancer patient. Laryngoscope 123:2161–2164 Escott EJ, 2013, Role of positron emission tomography/computed tomography, PET/CT, in head and neck cancer. Radiol Clin N Am 51:881–893 Tantiwongkosi B, Yu F, Kanard A, Miller FR, 2014, Role of, 18)FFDGPET/ CTin pre and post treatment evaluation in head and neck carcinoma. World J Radiol 6:177–191 QueirozMA, Hullner M, Kuhn F, Huber G,Meerwein C, Kollias S, von Schulthess G, Veit-Haibach P, 2014, PET/MRI and PET/CT in follow-up of head and neck cancer patients. Eur J Nucl Med Mol Imaging 41:1066–1075 Romboldt Z, Al–Okaili R, Deveikis JP, 2005, Perfusion CT for head and neck tumors: pilot study. AJNR Am J Neuroradiol 26: 1178–1185 Vandecaveye V, de Keyzer F, Vander Poorten V, Deraedt K, Alaerts H, LanduytW, Nuyts S, Hermans R, 2006, Evaluation of the larynx for tumour recurrence by diffusion-weighted MRI after radiotherapy: initial experience in four cases. Br J Radiol 79:681–687 Vandecaveye V, De Keyzer F, Nuyts S, Deraedt K, Dirix P, Hamaekers P, Vander Poorten V, Delaere P, Hermans R, 2007, Detection of head and neck squamous cell carcinoma with diffusion weighted MRI after, chemo, radiotherapy: correlation between radiologic and histopathologic findings. Int J Radiat Oncol Biol Phys 67:960–971 Vandecaveye V, De Keyzer F, Dirix P, Lambrecht M, Nuyts S, Hermans R, 2010, Applications of diffusion-weighted magnetic resonance imaging in head and neck squamous cell carcinoma. Neuroradiology 52:773–784 Vandecaveye V, Dirix P, de Keyzer F, de Beeck KO, Vander Poorten V, Roebben I, Nuyts S, Hermans R, 2010, Predictive value of diffusion-weighted magnetic resonance imaging during chemoradiotherapy for head and neck squamous cell carcinoma. Eur Radiol 20:1703–1715 Vandecaveye V, Dirix P, De Keyzer F, de Op Beeck K, Vander Poorten V, Hauben E, Lambrecht M, Nuyts S, Hermans R, 2012, Diffusion-weighted magnetic resonance imaging early after chemoradiotherapy to monitor treatment response in head-and-neck squamous cell carcinoma. Int J Radiat Oncol Biol Phys 82:1098–1107 Yuan J, Chow SK, Yeung DK, King AD, 2012, A five-colour colour-coded mapping method for DCE-MRI analysis of head and neck tumours. Clin Radiol 67:216–223 Lee FK, King AD, Ma BB, Yeung DK, 2012, Dynamic contrast enhancement magnetic resonance imaging, DCE-MRI, for differential diagnosis in head and neck cancers. Eur J Radiol 81:784–788 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1091 EP 1099 DI 10.1007/s12253-015-9942-7 PG 9 ER PT J AU Zidi, S Sghaier, I Zouidi, F Benahmed, A Stayoussef, M Kochkar, R Gazouani, E Mezlini, A Yacoubi-Loueslati, B AF Zidi, Sabrina Sghaier, Ikram Zouidi, Ferjeni Benahmed, Amira Stayoussef, Mouna Kochkar, Radhia Gazouani, Ezzedine Mezlini, Amel Yacoubi-Loueslati, Besma TI Interleukin-1 Gene Cluster Polymorphisms and its Haplotypes may Predict the Risk to Develop Cervical Cancer in Tunisia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Polymorphism; Haplotype; Interleukin-1beta; Interleukin-1alpha; Interleukin-1receptor antagonist; Cervical cancer; Tunisians ID Polymorphism; Haplotype; Interleukin-1beta; Interleukin-1alpha; Interleukin-1receptor antagonist; Cervical cancer; Tunisians AB Our study aimed to evaluate the association between IL-1α (4845 G/T), IL-1β (-511C/T) and IL-1RN (VNTR) polymorphisms and risk of cervical cancer. This case-control study investigates three polymorphisms in 130 patients and 260 controls by PCR-restriction fragment length polymorphism (RFLP). The IL-1RN (VNTR) A1/A3 genotype appear as a cervical cancer risk factor (p=0.048; OR= 2.92; 95 % CI=1.00–8.74), moreover, the L/2* decreased the risk (p=0.011; OR=0.47; 95 % CI=0.25–0.88) and may be a protective factor against this pathology. Stratified analysis according to the FIGO stage subgroup revealed that the IL-1β- 511 T/T genotype and T allele may be a protective factors against cervical cancer development for patients with early stage (p=0.030; OR=0.46; 95 % CI=0.22–0.96) (p=0.020; OR=0.68; 95 % CI=0.48–0.97). However, for the patients with advanced FIGO stage, IL-1RN-VNTR L/2* genotype appear as a protective factor for this pathology (p=0.023; OR=0.29; 95 % CI=0.08–0.99). The (G-T-L) haplotype showed a significant decreased frequency in cervical cancer patients as compared to controls (p=0.032; OR=0.53; 95 % CI=0.29–0.95). In contrast, the (T-T-2*) combination appear a risk factor for the development of cervical cancer (p=0.018; OR=1.57; 95%CI=1.07–2.30).Our study suggested that IL1 cluster polymorphisms and haplotypes may be a genetic risk factor for cervical cancer. C1 [Zidi, Sabrina] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 2092 El MANAR I, 1092 Tunis, Tunisia. [Sghaier, Ikram] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 2092 El MANAR I, 1092 Tunis, Tunisia. [Zouidi, Ferjeni] Habib Bourguiba UniversitySfax, Tunisia. [Benahmed, Amira] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 2092 El MANAR I, 1092 Tunis, Tunisia. [Stayoussef, Mouna] University of Monastir, Faculty of Pharmacy, Research Unit of Hematological and Autoimmune DiseasesMonastir, Tunisia. [Kochkar, Radhia] Military Hospital of Tunis, Laboratory of ImmunologyTunis, Tunisia. [Gazouani, Ezzedine] Military Hospital of Tunis, Laboratory of ImmunologyTunis, Tunisia. [Mezlini, Amel] Salah Azeiz Oncology InstituteTunis, Tunisia. [Yacoubi-Loueslati, Besma] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 2092 El MANAR I, 1092 Tunis, Tunisia. RP Zidi, S (reprint author), El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 1092 Tunis, Tunisia. EM ZIDISABRINA86@gmail.com CR Coussens LM, Werb Z, 2002, Inflammation and cancer. Nature 420:860–867 International Agency for Research on Cancer, 2008, GLOBOCAN v2.0: CancerIncidence and Mortality Worldwide; http://globocan. iarc.fr. Accessed 13.03.13 Maxwell PD, Bray F, Ferlay J, Jemal A, 2014, Cancer in Africa 2012. Cancer Epidemiol Biomarkers Prev 23:953–966 Castle PE, Hillier SL, Rabe LK, Hildesheim A, Herrero R, Bratti MC, Sherman ME, Burk RD, Rodriguez AC, Alfaro M, Hutchinson ML, Morales J, Schiffman M, 2001, An association of cervical inflammation with high-grade cervical neoplasia in women infected with oncogenic human papillomavirus, HPV). Cancer Epidemiol Biomarkers Prev 10:1021–1027 Smith JS, Herrero R, Bosetti C, Munoz N, Bosch FX, Eluf-Neto J, Castellsague X, Meijer CJ, Van den Brule AJ, Franceschi S, Ashley R, 2002, Herpes simplex virus-2 as a human papillomavirus cofactor in the etiology of invasive cervical cancer. J Natl Cancer Inst 94: 1604–1613 van der Poll T, van Deventer SJ, 1999, Cytokines and anticytokines in the pathogenesis of sepsis. Infect Dis Clin N Am 13:413–426 Cohen J, 2002, The immunopathogenesis of sepsis. Nature 420: 885–891 Riedemann NC, Guo RF, Ward PA, 2003, The enigma of sepsis. J Clin Invest 112:460–467 Dinarello CA, 1996, Biologic bases for interleukin-1 in disease. Blood 87(6):2095–2147 Bird S, Zou J, Wang T, Munday B, Cunningham C, Secombes CJ, 2002, Evolution of interleukin-1beta. Cytokine Growth Factor Rev 13:483–502 Nicklin MJH, Weith A, Duff GW, 1994, A physical map of the region encompassing the human interleukin-1a, interleukin-1b, and interleukin-1 receptor antagonist genes. Genomics 19:382–384 Dinarello C, 1998, Interleukin-1, interleukin-1 receptors and interleukin-1receptor antagonist. Int Rev Immunol 16:457–499 Majeed GS, Glew S, Bidwell J, 1999, An association between LSIL and the high secretor phenotype of IL-1beta. Gynecol Oncol 73:359–361 Tarlow JK, Blakemore AI, Lennard A, Solari R, Hughes HN, Steinkasserer A et al, 1993, Polymorphism in human IL-1 receptor antagonist gene intron 2 is caused by variable numbers of an 86-bp tandem repeat. Hum Genet 91(4):403–404 Kinane DF, Shiba H, Hart TC, 2005, The genetic basis of periodontitis. Periodontol 2000(39):91–117 Marth C, ZeimetAG, HeroldM, BrummC,Windbichler G,Muller- Holzner E et al, 1996, Different effects of interferons, interleukin- 1ß and tumor necrosis factor- in normal, OSE, and malignant human ovarian epithelial cells. Int J Cancer 67:826–830 Grimm C,Watrowski R, Baumuhlner K, Natter C, Tong D,Wolf A, Zeillinger R, Leodolter S, Reinthaller A, Hefler L, 2011, Genetic variations of interleukin-1 and -6 genes and risk of cervical intraepithelial neoplasia. Gynecol Oncol 121:537–541 Tarlow JK, Blakemore AIF, Lennard A et al, 1993, Polymorphism in human IL-1 receptor antagonist gene intron 2 is caused by variable numbers of an 86-bp tandem repeat. Hum Genet 91:403–404 Clay FE, Tarlow JK, Cork MJ, Cox A, Nicklin MJH, Duff GW, 1996, Novel interleukin-1 receptor antagonist exon polymorphisms and their use in allele-specific mRNA assessment. Hum Genet 97:723–726 Carter MJ, di Giovine FS, Jones S et al, 2001, Association of the interleukin-1 receptor antagonist gene with ulcerative colitis in Northern European Caucasians. Gut 48:461–467 Mustea A, Sehouli J, Konsgen D, Stengel D, Sofroni D, Lichtenegger W, 2003, Interleukin 1 receptor antagonist, IL- 1RA, polymorphism in women with cervical cancer. Anticancer Res 23:1099–1102 Sehouli J, Mustea A, 2002, Interleukin-1 receptor antagonist gene polymorphism and cancer. Clin Infect Dis 34:1535–1536 Kang S, Kim JW, Park NH, Song YS, Park SY, Kang SB et al, 2007, Interleukin-1 beta-511 polymorphism and risk of cervical cancer. J Korean Med Sci 22:110–113 Zidi S, Verdi H, Yilmaz-Yalcin Y et al, 2014, Impact of toll-like receptors 2/3/4/9, IL-1-a/b and TNF-a Polymorphisms in cervical cancer susceptibility in Tunisia. Pathol Oncol Res., DOI 10.1007/ s12253-014-9793-7 Achyut BR, Srivastava A, Bhattacharya S, Mittal B, 2007, Genetic association of interleukin-1beta, -511C/T, and interleukin-1 receptor antagonist, 86 bp repeat, polymorphisms with Type 2 diabetes mellitus in North Indians. Clin Chim Acta 377:163–169 Eisenberg SP, BrewerMT, Verderber E, Heimdal P, Brandhuber BJ, Thompson RC, 1991, Interleukin 1 receptor antagonist is amember of the interleukin 1 gene family: evolution of a cytokine control mechanism. Proc Natl Acad Sci U S A 88:5232–5236 Qian N, Chen X, Han S, Qiang F, Jin G, Zhou X, Dong J,Wang X, Shen H, Hu Z, 2010, Circulating IL-1beta levels, polymorphisms of IL-1B, and risk of cervical cancer in Chinese women. J Cancer Res Clin Oncol 136(5):709–716 El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA et al, 2000, Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature 404:398–402 He JF, Jia WH, Fan Q, Zhou XX, Qin HD, Shugart YY, Zeng YX, 2007, Genetic polymorphisms of TLR3 are associated with nasopharyngeal carcinoma risk in Cantonese population. Bio Med Cent Cancer 7:194 Shin WG, Jang JS, Kim HS, Kim SJ, Kim KH, Jang MK, Lee JH, Kim HJ, Kim HY, 2008, Polymorphisms of interleukin-1 and interleukin-2 genes in patients with gastric cancer in Korea. J Gastroenterol Hepatol 23(10):1567–1573 McIntyre KW, Stepan GJ, Kolinsky KD, Benjamin WR, Plocinski JM, Kaffka KL, Campen CA, Chizzonite RA, Kilian PL, 1991, Inhibition of interleukin 1, IL-1, binding and bioactivity in vitro and modulation of acute inflammation in vivo by IL-1 receptor antagonist and anti-IL-1 receptor monoclonal antibody. J Exp Med 173(4):931–939 Maruta Y, Okayama N, Hiura M, Suehiro Y, Hirai H, Hinoda Y, 2008, Determination of ancestral allele for possible human cancerassociated polymorphisms. Cancer Genet Cytogenet 180(1):24–29 Kapoor S, 2009, Role of polymorphisms of interleukin-1 receptor antagonist gene in systemic oncogenesis: significant etiopathologic connections besides in benign prostatic hyperplasia. Urology 73(1): 215 Tamandani DM, Sobti RC, Shekari M, Kaur S, Huria A, 2008, Impact of polymorphism in IL-1RA gene on the risk of cervical cancer. Arch Gynecol Obstet 277(6):527–533 Singh H, Sachan R, Goel H, Mittal B, 2008, Genetic variants of interleukin-1RN and interleukin-1beta genes and risk of cervical cancer. BJOG 115(5):633–638 Shepperd JH, 1995, FIGO staging of gynecologic cancers; cervical and vulva. Int J Gynecol Cancer 5:319–323 Hugo S, Santos AM, Catarino R, Pinto D, Moutinho J, Canedo P, Machado JC, Medeiros R, 2012, IL-1RN VNTR polymorphism and genetic susceptibility to cervical cancer in Portugal. Mol Biol Rep 39:10837–10842 Sobti RC, Kordi Tamandani DM, Shekari M, Kaur P, Malekzadeh K, Suri V, 2008, Interleukin 1 beta gene polymorphism and risk of cervical cancer. Int J Gynecol Obstet 101:47–52 Shimu W, Guiping H, Chen J, Xie G, 2014, Interleukin 1β and interleukin 1 receptor antagonist gene polymorphisms and cervical cancer: a meta-analysis. Int J Gynecol Cancer 24(6):984–990 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1101 EP 1107 DI 10.1007/s12253-015-9941-8 PG 7 ER PT J AU Vuletic, A Jovanic, I Jurisic, V Milovanovic, Z Nikolic, S Spurnic, I Konjevic, G AF Vuletic, Ana Jovanic, Irena Jurisic, Vladimir Milovanovic, Zorka Nikolic, Srdan Spurnic, Igor Konjevic, Gordana TI Decreased Interferon γ Production in CD3+ and CD3−CD56+ Lymphocyte Subsets in Metastatic Regional Lymph Nodes of Melanoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Regional lymph nodes; Interferon γ; T cells; NK cells ID Regional lymph nodes; Interferon γ; T cells; NK cells AB As lymphogenic dissemination is very common in melanoma, regional lymph nodes (LN)s represent first immunological barriers to tumor invasion and play a complex role in antitumor immune defense. In this sense, their most prominent role is the presentation of tumor-derived antigens to naive T cells and generation of cell-mediated adaptive immune response. Since tumor micro-environment affects immune cell function in this study we have evaluated the ability of T cells and NK cells in metastatic (involved) and non-metastatic regional LNs to produce interferon γ (IFNγ), a pleiotropic cytokine that regulates adaptive antitumor immune response. Our results show reduced IFNγ production in both T and NK lymphocyte subsets and decreased prevalence of T cells in metastatic regional LNs of melanoma patients. The decrease of IFNγ production in T cells was more pronounced with increased number of involved regional LNs indicating tumor-induced functional impairment of both T and NK cell lymphocyte subsets in involved regional LNs. Therefore, shown low IFNγ production in metastatic LNs may represent an obstacle in adaptive cell-mediated antitumor immune response and hence may enable tumor progression. C1 [Vuletic, Ana] Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia. [Jovanic, Irena] Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia. [Jurisic, Vladimir] University of Kragujevac, Faculty of MedicineKragujevac, Serbia. [Milovanovic, Zorka] Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia. [Nikolic, Srdan] Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia. [Spurnic, Igor] Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia. [Konjevic, Gordana] Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia. RP Vuletic, A (reprint author), Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia. EM radovanovica@ncrc.ac.rs CR Cochran AJ, Huang RR, Lee J, Itakura E, Leong SP, Essner R, 2006, Tumour-induced immune modulation of sentinel lymph nodes. Nat Rev Immunol 6:659–670 Seliger B, Ruiz-Cabello F, Garrido F, 2008, IFN inducibility of major histocompatibility antigens in tumors. Adv Cancer Res 101:249–276 Zaidi MR, Merlino G, 2011, The two faces of interferon-γ in cancer. Clin Cancer Res 17:6118–6124 Carrega P, Ferlazzo G, 2012, Natural killer cell distribution and trafficking in human tissues. Front Immunol 3:347 Martin-Fontecha A, Lanzavecchia A, Sallusto F, 2009, Dendritic cell migration to peripheral lymph nodes. Handb Exp Pharmacol 188:31–49 Ferlazzo G, Morandi B, 2014, Cross-talks between natural killer cells and distinct subsets of dendritic cells. Front Immunol 5:159 Fehniger TA, Cooper MA, Nuovo GJ, Cella M, Facchetti F, Colonna M, Caligiuri MA, 2003, CD56bright natural killer cells are present in human lymph nodes and are activated by T cellderived IL-2: a potential new link between adaptive and innate immunity. Blood 101:3052–3057 Morandi B, Bougras G, Muller WA, Ferlazzo G, Munz C, 2006, NK cells of human secondary lymphoid tissues enhance T cell polarization via IFN-gamma secretion. Eur J Immunol 36:2394– 2400 LucasM, SchachterleW, OberleK, Aichele P, DiefenbachA, 2007, Dendritic cells prime natural killer cells by trans-presenting interleukin 15. Immunity 26:503–517 Morandi B, Mortara L, Carrega P, Cantoni C, Costa G, Accolla RS, Mingari MC, Ferrini S, Moretta L, Ferlazzo G, 2009, NK cells provide helper signal for CD8+ T cells by inducing the expression of membrane-bound IL-15 on DCs. Int Immunol 21:599–606 Jackson A, Warner N, 1986, Preparation, staining and analysis by flow cytometry of peripheral blood leukocytes. In: Rose N, Friedman H, Fahey J, eds, Manual of clinical laboratory immunology, 3rd edn. American Society for Microbiology, Washington, pp 226–235 Farzad Z, Cochran AJ, McBrideWH, Gray JD,Wong V, Morton L, 1990, Lymphocyte subset alterations in nodes regional to human melanoma. Cancer Res 50:3585–35888 Vuletic A, Jurisic V, Jovanic I, Milovanovic Z, Nikolic S, Konjevic G, 2013, Distribution of several activating and inhibitory receptors onCD3(−)CD56(+, NKcells in regional lymph nodes ofmelanoma patients. J Surg Res 183:860–868 Morton BA, RameyWG, PaderonH, Miller RE, 1986)Monoclonal antibody-defined phenotypes of regional lymph node and peripheral blood lymphocyte subpopulations in early breast cancer. Cancer Res 46(4 Pt 2):2121–2126 Nandakumar S, Woolard SN, Yuan D, Rouse BT, Kumaraguru U, 2008, Natural killer cells as novel helpers in anti-herpes simplex virus immune response. J Virol 82:10820–10831 Adam C, King S, Allgeier T, Braumuller H, Luking C, Mysliwietz J, Kriegeskorte A, Busch DH, Rocken M, Mocikat R, 2005, DCNK cell cross talk as a novel CD4+ T-cell-independent pathway for antitumor CTL induction. Blood 106:338–344 De Maria A, Bozzano F, Cantoni C, Moretta L, 2011, Revisiting human natural killer cell subset function revealed cytolytic CD56(dim)CD16+ NK cells as rapid producers of abundant IFNgamma on activation. Proc Natl Acad Sci U S A 108:728–732 Carrega P, Bonaccorsi I, Di Carlo E, Morandi B, Paul P, Rizzello V, Cipollone G, Navarra G, Mingari MC, Moretta L, Ferlazzo G, 2014, CD56brightperforinlow noncytotoxic human NK cells are abundant in both healthy and neoplastic solid tissues and recirculate to secondary lymphoid organs via afferent lymph. J Immunol 192: 3805–3815 Umansky V, Sevko A, 2012, Melanoma-induced immunosuppression and its neutralization. Semin Cancer Biol 22:319–326 Talmadge JE, 2011, Immune cell infiltration of primary and metastatic lesions: mechanisms and clinical impact. Semin Cancer Biol 21:131–138 Nevala WK, Vachon CM, Leontovich AA, Scott CG, Thompson MA, Markovic SN, 2009, Melanoma Study Group of the Mayo Clinic Cancer Center. Evidence of systemic Th2-driven chronic inflammation in patients with metastatic melanoma. Clin Cancer Res 15:1931–1939 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1109 EP 1114 DI 10.1007/s12253-015-9938-3 PG 6 ER PT J AU Lorincz, T Jemnitz, K Kardon, T Mandl, J Szarka, A AF Lorincz, Tamas Jemnitz, Katalin Kardon, Tamas Mandl, Jozsef Szarka, Andras TI Ferroptosis is Involved in Acetaminophen Induced Cell Death SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acetaminophen; Hepatotoxicity; Ferroptosis; Necroptosis; Glutathione ID Acetaminophen; Hepatotoxicity; Ferroptosis; Necroptosis; Glutathione AB The recently described form of programmed cell death, ferroptosis can be induced by agents causing GSH depletion or the inhibition of GPX4. Ferroptosis clearly shows distinct morphologic, biochemical and genetic features from apoptosis, necrosis and autophagy. Since NAPQI the highly reactive metabolite of the widely applied analgesic and antipyretic, acetaminophen induces a cell death which can be characterized by GSH depletion, GPX inhibition and caspase independency the involvement of ferroptosis in acetaminophen induced cell death has been investigated. The specific ferroptosis inhibitor ferrostatin-1 failed to elevate the viability of acetaminophen treated HepG2 cells. It should be noticed that these cells do not form NAPQI due to the lack of phase I enzyme expression therefore GSH depletion cannot be observed. However in the case of acetaminophen treated primary mouse hepatocytes the significant elevation of cell viability could be observed upon ferrostatin-1 treatment. Similar to ferrostatin-1 treatment, the addition of the RIP1 kinase inhibitor necrostatin-1 could also elevate the viability of acetaminophen treated primary hepatocytes. Ferrostatin-1 has no influence on the expression of CYP2E1 or on the cellular GSH level which suggest that the protective effect of ferrostatin-1 in APAP induced cell death is not based on the reduced metabolism of APAP to NAPQI or on altered NAPQI conjugation by cellular GSH. Our results suggest that beyond necroptosis and apoptosis a third programmed cell death, ferroptosis is also involved in acetaminophen induced cell death in primary hepatocytes. C1 [Lorincz, Tamas] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, Szent Gellert ter 4, 1111 Budapest, Hungary. [Jemnitz, Katalin] Research Centre for Natural Sciences, Hungarian Academy of Sciences, Institute of Molecular PharmacologyBudapest, Hungary. [Kardon, Tamas] Hungarian Academy of Sciences and Semmelweis University, Pathobiochemistry Research Group, 1444 Budapest, Hungary. [Mandl, Jozsef] Hungarian Academy of Sciences and Semmelweis University, Pathobiochemistry Research Group, 1444 Budapest, Hungary. [Szarka, Andras] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, Szent Gellert ter 4, 1111 Budapest, Hungary. RP Szarka, A (reprint author), Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, 1111 Budapest, Hungary. EM szarka@mail.bme.hu CR Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, Morrison B 3rd, Stockwell BR, 2012, Ferroptosis: an irondependent form of nonapoptotic cell death. Cell 149(5):1060– 1072., DOI 10.1016/j.cell.2012.03.042 Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, Brown LM, Girotti AW, Cornish VW, Schreiber SL, Stockwell BR, 2014, Regulation of ferroptotic cancer cell death by GPX4. Cell 156(1–2):317–331., DOI 10.1016/j.cell.2013.12.010 Friedmann Angeli JP, SchneiderM, Proneth B, Tyurina YY, Tyurin VA, HammondVJ, Herbach N, AichlerM,WalchA, Eggenhofer E, Basavarajappa D, Radmark O, Kobayashi S, Seibt T, Beck H, Neff F, Esposito I,Wanke R, Forster H,Yefremova O, Heinrichmeyer M, Bornkamm GW, Geissler EK, Thomas SB, Stockwell BR, O’Donnell VB, Kagan VE, Schick JA, Conrad M, 2014, Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol 16(12):1180–1191., DOI 10.1038/ ncb3064 Murphy TH, Miyamoto M, Sastre A, Schnaar RL, Coyle JT, 1989, Glutamate toxicity in a neuronal cell line involves inhibition of cystine transport leading to oxidative stress. Neuron 2(6):1547– 1558., DOI 10.1016/0896-6273(89)90043-3 McGill MR, Lebofsky M, Norris HR, Slawson MH, Bajt ML, Xie Y, Williams CD, Wilkins DG, Rollins DE, Jaeschke H, 2013, Plasma and liver acetaminophen-protein adduct levels in mice after acetaminophen treatment: dose–response,mechanisms, and clinical implications. Toxicol Appl Pharmacol 269(3):240–249., DOI 10. 1016/j.taap.2013.03.026 Mitchell JR, Jollow DJ, Potter WZ, Davis DC, Gillette JR, Brodie BB, 1973, Acetaminophen-induced hepatic necrosis. I. Role of drug metabolism. J Pharmacol Exp Ther 187:185–194 Mitchell JR, Jollow DJ, Potter WZ, Gillette JR, Brodie BB, 1973, Acetaminophen-induced hepatic necrosis. IV. Protective role of glutathione. J Pharmacol Exp Ther 187:211–217 Jaeschke H, McGill MR, Williams CD, Ramachandran A, 2011, Current issues with acetaminophen hepatotoxicity–a clinically relevant model to test the efficacy of natural products. Life Sci 88(17– 18):737–745., DOI 10.1016/j.lfs.2011.01.025 Tirmenstein MA, Nelson SD, 1990, Acetaminophen-induced oxidation of protein thiols. Contribution of impaired thiolmetabolizing enzymes and the breakdown of adenine nucleotides. J Biol Chem 265(6):3059–3065 Lawson JA, Fisher MA, Simmons CA, Farhood A, Jaeschke H, 1999, Inhibition of Fas receptor, CD95)-induced hepatic caspase activation and apoptosis by acetaminophen in mice. Toxicol Appl Pharmacol 156:179–186 Gujral JS, Knight TR, Farhood A, Bajt ML, Jaeschke H, 2002, Mode of cell death after acetaminophen overdose inmice: apoptosis or oncotic necrosis? Toxicol Sci 67:322–328 Jaeschke H, McGill MR, Ramachandran A, 2012, Oxidant stress, mitochondria, and cell death mechanisms in drug-induced liver injury: lessons learned from acetaminophen hepatotoxicity. Drug Metab Rev 44(1):88–106., DOI 10.3109/03602532.2011.602688 Williams CD, Farhood A, Jaeschke H, 2010, Role of caspase-1 and interleukin-1beta in acetaminophen-induced hepatic inflammation and liver injury. Toxicol Appl Pharmacol 247:169–178., DOI 10. 1016/j.taap.2010.07.004 Jemnitz K, Veres Z, Monostory K, Kobori L, Vereczkey L, 2008, Interspecies differences in acetaminophen sensitivity of human, rat, and mouse primary hepatocytes. Toxicol In Vitro 22(4):961–967., DOI 10.1016/j.tiv.2008.02.001 Washko PW, Wang Y, Levine M, 1993, Ascorbic acid recycling in human neutrophils. J Biol Chem 268(21):15531–15535 Sakurai J, Funae Y, Nemoto N, 1996, Maintenance and activation of Cyp2e-1 gene expression in mouse hepatocytes in primary culture. Biochim Biophys Acta 1313(1):35–40 Nagy G, Szarka A, Lotz G, Doczi J,Wunderlich L, Kiss A, Jemnitz K,Veres Z, Banhegyi G, Schaff Z, Sumegi B,Mandl J, 2010, BGP- 15 inhibits caspase-independent programmed cell death in acetaminophen-induced liver injury. Toxicol Appl Pharmacol 243(1):96–103 Roe AL, Snawder JE, Benson RW, Roberts DW, Casciano DA, 1993, HepG2 cells: an in vitro model for P450-dependent metabolism of acetaminophen. Biochem Biophys Res Commun 190(1): 15–19 McGill MR, Yan HM, Ramachandran A, Murray GJ, Rollins DE, Jaeschke H, 2011, HepaRG cells: a human model to study mechanisms of acetaminophen hepatotoxicity. Hepatology 53(3):974– 982., DOI 10.1002/hep.24132 McGill MR, Williams CD, Xie Y, Ramachandran A, Jaeschke H, 2012, Acetaminophen-induced liver injury in rats and mice: comparison of protein adducts, mitochondrial dysfunction, and oxidative stress in the mechanism of toxicity. Toxicol Appl Pharmacol 264(3):387–394., DOI 10.1016/j.taap.2012.08.015 Degterev A, Hitomi J, Germscheid M, Ch’en IL, Korkina O, Teng X, Abbott D, Cuny GD, Yuan C, Wagner G, Hedrick SM, Gerber SA, Lugovskoy A, Yuan J, 2008, Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol 4(5):313– 321., DOI 10.1038/nchembio.83 Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G, 2010, Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol 11(10):700–714., DOI 10.1038/ nrm2970 Nagy G, Kardon T, Wunderlich L, Szarka A, Kiss A, Schaff Z, Banhegyi G, Mandl J, 2007, Acetaminophen induces ER dependent signaling in mouse liver. Arch Biochem Biophys 459(2):273– 279 Manov I, Hirsh M, Iancu TC, 2004, N-acetylcysteine does not protect HepG2 cells against acetaminophen-induced apoptosis. Basic Clin Pharmacol Toxicol 94(5):213–225 Macanas-Pirard P, Yaacob NS, Lee PC, Holder JC, Hinton RH, Kass GE, 2005, Glycogen synthase kinase-3 mediates acetaminophen-induced apoptosis in human hepatoma cells. J Pharmacol Exp Ther 313(2):780–789 Takemoto K, Hatano E, Iwaisako K, Takeiri M, Noma N, Ohmae S, Toriguchi K, Tanabe K, Tanaka H, Seo S, Taura K, Machida K, Takeda N, Saji S, Uemoto S, Asagiri M, 2014, Necrostatin-1 protects against reactive oxygen species, ROS)-induced hepatotoxicity in acetaminophen-induced acute liver failure. FEBS Open Bio 4: 777–787., DOI 10.1016/j.fob.2014.08.007 Zhang YF, He W, Zhang C, Liu XJ, Lu Y, Wang H, Zhang ZH, Chen X, Xu DX, 2014, Role of receptor interacting protein, RIP)1 on apoptosis-inducing factor-mediated necroptosis during acetaminophen-evoked acute liver failure in mice. Toxicol Lett 225(3):445–453., DOI 10.1016/j.toxlet.2014.01.005 Meister A, 1994, Glutathione, ascorbate, and cellular protection. Cancer Res 54(7 Suppl):1969s–1975s May JM, Qu ZC, Morrow JD, 1996, Interaction of ascorbate and alpha-tocopherol in resealed human erythrocyte ghosts. Transmembrane electron transfer and protection from lipid peroxidation. J Biol Chem 271(18):10577–10582 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1115 EP 1121 DI 10.1007/s12253-015-9946-3 PG 7 ER PT J AU Byatnal, AA Byatnal, A Sen, S Guddattu, V Solomon, ChM AF Byatnal, Amit Aditi Byatnal, Amit Sen, Subhalakshmi Guddattu, Vasudev Solomon, Charlotte Monica TI Cyclooxygenase-2 – An Imperative Prognostic Biomarker in Oral Squamous Cell Carcinoma- An Immunohistochemical Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Carcinoma; Squamous cell; Cyclooxygenase 2; Immunohistochemistry ID Carcinoma; Squamous cell; Cyclooxygenase 2; Immunohistochemistry AB Oral squamous cell carcinoma (OSCC) is the most common head and neck squamous cell carcinoma (HNSCC) with metastasis and tumor recurrence resulting in 90 % of cancer associated mortality. COX-2, an inflammatory biomarker, has been shown to play a significant role in tumorigenesis of OSCC. To study the expression of COX-2 in OSCC by immunohistochemistry and investigate its association with the clinicopathological parameters including patient survival. A cross sectional study was carried out in 75 histologically confirmed cases of OSCC. COX-2 expression was evaluated by indirect streptavidin biotin method. The expression was semi-quantitatively assessed using established criteria. The expression profile of COX-2 was correlated with the clinicopathological details like tumor size, regional lymphnode metastasis, distant metastasis, clinical stage, local recurrence of tumor, histological grade, and survival of patient. Chi square and Kaplan Meier statistical tests were applied for assessing this association. COX-2 expression was absent in normal oral mucosa. Over expression of COX-2 was seen in 58 out of 75 specimens of OSCC. Overexpression of COX-2 was significantly associated with the lymphnode involvement, histological grade, local recurrence of tumor and patient survival. COX-2 expression represents an important biomarker of prognostic significance that may be used to identify a subset of patients at high risk and to predict patient survival. C1 [Byatnal, Amit Aditi] Oral Pathology and MicrobiologyHubli, Karnataka, India. [Byatnal, Amit] AMES Dental College and Hospital, Department of Oral Medicine and RadiologyRiachur, Karnataka, India. [Sen, Subhalakshmi] Manipal University, Manipal College of Dental Sciences, Department of Oral Pathology and Microbiology, 576104 Manipal, Karnataka, India. [Guddattu, Vasudev] Manipal University, Department of BiostatisticsManipal, Karnataka, India. [Solomon, Charlotte Monica] Manipal University, Manipal College of Dental Sciences, Department of Oral Pathology and Microbiology, 576104 Manipal, Karnataka, India. RP Solomon, ChM (reprint author), Manipal University, Manipal College of Dental Sciences, Department of Oral Pathology and Microbiology, 576104 Manipal, India. EM solomonmc@gmail.com CR Werning JW, 2007, Oral cancer: diagnosis, management, and rehabilitation. In: Werning JW, ed). New York Hamada GS, Bos AJ, Kasuga H, Hirayama T, 1991, Comparative epidemiology of oral cancer in Brazil & India. Tokai J Exp Clin Med 16(1):63–72 Vane JR, Mitchell JA, Appleton I, Tomlinson A, Bishop-Bailey D, Croxtall X et al, 1994, Inducible isoforms of cyclooxygenase and nitric-oxide synthase in inflammation. Proc Natl Acad Sci U S A 91(16):2046–2050 Broders AC, 1942, The microscopic significance of histological grading of cancer. Surg Clin North Am 21:947–62 Itoh S,Matsui K, Furuta I, Takano Y, 2003, Immunohistochemical study on overexpression of cyclooxygenase-2 in squamous cell carcinoma of the oral cavity: its importance as a prognostic predictor. Oral Oncol 39:829–835 Jemal A, Thomas A, Murray T, Thun M, 2002, Cancer statistics 2002. CA Cancer J Clin 52(1):23–47 Yoshikawa R, Sano H, Masuda C, Kawamura M, Tsubouchi Y, Chargui J et al, 2000, Expression of cyclooxygenase-2 in prostate carcinoma. Cancer 89(3):589–596 Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM et al, 1999, Cyclooxygenase-2 expression is up-regulated in human pancreatic cancer. Cancer Res 59(5):987–990 Sano H, Kawahito Y,Wilder RL, Hashiramoto A,Mukai S, Asai K et al, 1995, Expression of cyclooxgenase-1 and-2 in human colorectal cancer. Cancer Res 55(17):3785–3789 Huang M, Stolina M, Sharma S, Mao JT, Zhu L, Miller PW et al, 1998, Non-small cell lung cancer cyclooxygenase-2-dependent regulation of cytokine balance in lymphocytes and macrophage: up-regulation of interleukin 10 and down-regulation of interleukin 12 production. Cancer Res 58(6):1208–1216 Chan G, Boyle JO, Yang EK, Zhang F, Sacks PG, Shah JP et al, 1999, Cyclooxygenase-2 expression is up-regulated in squamous cell carcinoma of the head and neck. Cancer Res 59(5):991–994 Fujiwaki R, Iida K, Kanasaki H, Ozaki T, Hata K, Miyazaki K, 2002, Cyclooxygenase-2 expression in endometrial cancer: correlation with microvessel count and expression of vascular endothelial growth factor and thymidine phosphorylase. Hum Pathol 33(2): 213–219 Tsujii M, DuBois RN, 1995, Alterations in cellular adhesion and apoptosis in epithelial cells overexpressing prostaglandin endoperoxide synthase 2. Cell 83(3):493–501 Tsujii M, Kawano S, Tuji S, Sawaoka H, Hori M, DuBois RN, 1998, Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell 93(5):705–716 Snyderman CH, Milanovich M, Wagner RL, Johnson JT, 1995, Prognostic significance of prostaglandin E2 production in fresh tissues of head and neck cancer patients. Head Neck 17(2):108–113 Renkonen J, Wolff H, Paavonen T, 2002, Expression of cyclooxygenase- 2 in human tongue carcinoma and its precursor lesions. Virchows Arch 440(6):594–597 Segawa E, Sakurai K, Kishimoto H, Takaoka K, Noguchi K, Hashitani S et al, 2008, Expression of cyclooxygenase-2 and DNA topoisomerase II alpha in precancerous and cancerous lesions of the oral mucosa. Oral Oncol 44(7):664–671 Mauro A, Lipari L, Leone A, Tortorici S, Burruano F, Provenzano S et al, 2010, Expression of cyclooxygenase-1 and cyclooxygenase-2 in normal and pathological human oral mucosa. Folia Histochem Cytobiol 48(4):555–563 Svensson CI, Yaksh TL, 2002, The spinal phospholipasecyclooxygenase- prostanoid cascade in nociceptive processing. Annu Rev Pharmacol Toxicol 42:553–583 Kirschenbaum A, Liotta DR, Yao S, Liu XH, Klausner AP, Unger P et al, 2000, Immunohistochemical localization of cyclooxygenase- 1 and cyclooxygenase-2 in the human foetal and adult male reproductive tracts. J Clin Endocrinol Metab 85(9):3436–3441 Xie WL, Chipman JG, Robertson DL, Erikson RL, Simmons DL, 1991, Expression of a mitogen-responsive gene encoding prostaglandin synthase is regulated by mRNA splicing. Proc Natl Acad Sci U S A 88(7):2692–2696 Kujubu DA, Fletcher BS, Varnum BC, Lim RW, Herschman HR, 1991, TIS10, a phorbol ester tumor promoter-inducible mRNA from Swiss 3 T3 cells, encodes a novel prostaglandin synthase/ cyclooxygenase homologue. J Biol Chem 266(20):12866–12872 Li WZ, Ding YQ, Li ZG, Zhang JH, 2008, Expression of COX-2 and VEGF-C in squamous cell carcinoma of the tongue and its correlation to lymph node metastasis. Nan Fang Yi Ke Da Xue Xue Bao 28(2):180–183 Nagatsuka H, Siar CH, Tsujigiwa H, Naomoto Y, Han PP, Gunduz M et al, 2012, Heparanase and cyclooxygenase-2 gene and protein expressions during progression of oral epithelial dysplasia to carcinoma. Ann Diagn Pathol 16(5):354–361 Xiao Y, Teng Y, Zhang R, Luo L, 2012, Antitumor effect of the selective COX-2 inhibitor celecoxib on endometrial adenocarcinoma in vitro and in vivo. Oncol Lett 4(6):1219–1224 Soland TM, Husvik C, Koppang HS, Boysen M, Sandvik L, Clausen OP et al, 2008, A study of phosphorylated ERK1/2 and COX-2 in early stage, T1-T2, oral squamous cell carcinomas. J Oral Pathol Med 37(9):535–542 Grivennikov SI, Greten FR, Karin M, 2010, Immunity, inflammation, and cancer. Cell 140(6):883–899 Zamarron BF, ChenW(2011, Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci 7(5): 651–658 Sheng H, Shao J,Morrow JD, Beauchamp RD, DuBois RN, 1998, Modulation of apoptosis and Bcl-2 expression by prostaglandin E2 in human colon cancer cells. Cancer Res 58(2):362–366 Reya T, Morrison SJ, Clarke MF, Weissman IL, 2001, Stem cells, cancer, and cancer stem cells. Nature 414(6859):105–111 Feinberg AP, Ohlsson R, Henikoff S, 2006, The epigenetic progenitor origin of human cancer. Nat Rev Genet 7(1):21–33 Hanahan D, Folkman J, 1996, Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86(3):353–364 Dormond O, Foletti A, Paroz C, Ruegg C, 2001, NSAIDs inhibit alpha V beta 3 integrin-mediated and Cdc42/Rac-dependent endothelial-cell spreading, migration and angiogenesis. Nat Med 7(9):1041–1047 Yoshimura R, Sano H, Masuda C, Kawamura M, Tsubouchi Y, Chargui J et al, 2000, Expression of cyclooxygenase-2 in prostate carcinoma. Cancer 89(3):589–596 Bandyopadhyay R, Chatterjee U, Mondal SK, Nag D, Sinha SK, 2011, A study on expression pattern of cyclooxygenase-2 in carcinoma of cervix. Indian J Pathol Microbiol 54(4):695–699 Lipari L, Mauro A, Gallina S, Tortorici S, Buscemi M et al, 2012, Expression of gelatinases, MMP-2, MMP-9, and cyclooxygenases, COX-1, COX-2, in some benign salivary gland tumors. Int J Immunopathol Pharmacol 25(1):107–115 Sappayatosok K, Maneerat Y, Swasdison S, Viriyavejakul P, Dhanuthai K, Zwang J et al, 2009, Expression of proinflammatory protein, iNOS, VEGF and COX-2 in oral squamous cell carcinoma, OSCC), relationship with angiogenesis and their clinico-pathological correlation. Med Oral Patol Oral Cir Bucal 14(7):E319–E324 Wang YH, Wu MW, Yang AK, Zhang WD, Sun J, Liu TR et al, 2011, COX-2 Gene increases tongue cancer cell proliferation and invasion through VEGF-C pathway. Med Oncol Suppl 1:S360–S366 Morita Y, Hata K, Nakanishi M, Nishisho T, Yura Y, Yoneda T, 2012, Cyclooxygenase-2 promotes tumor lymphangiogenesis and lymph node metastasis in oral squamous cell carcinoma. Int J Oncol 41(3):885–892 Tsujii M, Kawano S, DuBois RN, 1997, Cyclooxygenase-2 expression in human colon cancer cells increasesmetastatic potential. Proc Natl Acad Sci U S A 94(7):3336–3340 Buchanan FG, Wang D, Bargiacchi F, DuBois RN, 2003, Prostaglandin E2 regulates cell migration via the intracellular activation of the epidermal growth factor receptor. J Biol Chem 278(37):35451–35457 Birchmeier C, BirchmeierW, Gherardi E, VandeWoude GF, 2003, Met, metastasis, motility and more. Nat Rev Mol Cell Biol 4(12): 915–925 Greenhough A, Smartt JM, Moore AE, Roberts HR, Williams AC, Paraskeva C et al, 2009, The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation of tumor microenvironment. Carcinogenesis 30(3):377–386 Gallo O, Masini E, Bianchi B, Bruschini L, Paglierani M, FranchiA, 2002, Prognostic significance of cyclooxygenase-2 pathway and angiogenesis in head and neck squamous cell carcinoma. Hum Pathol 33:708–714 Prins MJ, Verhage RJ, ten Kate FJ, van Hillegersberg R, 2012, Cyclooxygenase isoenzyme-2 and vascular endothelial growth factor are associated with poor prognosis in oesophageal adenocarcinoma. J Gastrointest Surg 16(5):956–966 Yan YX, LiWZ, Huang YQ, LiaoWX(2012, The COX-2 inhibitor Celecoxib enhances the sensitivity of KB/VCR oral cancer cell lines to Vincristine by down-regulating P-glycoprotein expression and function. Prostaglandins Other Lipid Mediat 97(1–2):29–35 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1123 EP 1131 DI 10.1007/s12253-015-9940-9 PG 9 ER PT J AU Chen, Px Li, Qy Yang, Z AF Chen, Pu-xiang Li, Qiao-yan Yang, Zhulin TI Musashi-1 Expression is a Prognostic Factor in Ovarian Adenocarcinoma and Correlates with ALDH-1 Expression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ovarian cancer; Musashi-1; Immunohistochemistry ID Ovarian cancer; Musashi-1; Immunohistochemistry AB The presence of cancer stem-like cells (CSCs) has been demonstrated to be associated with tumor metastasis, chemoresistance, and rapid recurrence of various tumors. The impact of CSC-related markers in the metastasis and prognosis of ovarian cancer has not been well established. In this study, the protein expression of musashi-1 and ALDH1 was measured using immunohistochemistry. Results demonstrated that the percentage of positive musashi-1 and ALDH1 expression were significantly higher in ovarian serous adenocarcinomas, mucinous adenocarcinomas and clear cell adenocarcinomas than in cystadenomas and normal tissues. The percentage of positive musashi-1 and ALDH1 expression were significantly lower in patients identifiedwith clinical stage I or II ovarian adenocarcinomas without lymph node metastasis compared to patients with clinical stage III or IV tumors and lymph node metastasis. The expression of musashi-1 and ALDH1 was found to be highly consistent in ovarian adenocarcinomas. Univariate Kaplan-Meier analysis showed a negative correlation between musashi-1 or ALDH1 expression and overall survival. Multivariate Cox regression analysis showed that positive expression of musashi-1 or ALDH1 in ovarian adenocarcinoma was an independent predictor of poor prognosis. Our study suggested that musashi-1 and ALDH1 expression are closely related to metastasis of ovarian adenocarcinoma. The positive expression of musashi-1 and ALDH1 might be a poor-prognostic factor of ovarian adenocarcinoma. C1 [Chen, Pu-xiang] Second Xiangya Hospital, Department of Obstetrics and Gynaecology, 410011 Changsha, Hunan, China. [Li, Qiao-yan] Hunan Provincial People’s Hospital, 410011 Changsha, Hunan, China. [Yang, Zhulin] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, Hunan, China. RP Yang, Z (reprint author), Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, China. EM zhulinyang@yahoo.com CR Van Simaeys D, Lopez-Colon D, Sefah K, Sutphen R, Jimenez E, Tan W, 2010, Study of the molecular recognition of aptamers selected through ovarian cancer cell-SELEX. PLoS One 5:e13770 Hartge P, 2010, Designing early detection programs for ovarian cancer. J Natl Cancer Inst 102:3–4 Willmott LJ, Fruehauf JP, 2010, Targeted therapy in ovarian cancer. J Oncol 2010:740472 Hassan MK, Watari H, Christenson L, Bettuzzi S, Sakuragi N, 2011, Intracellular clusterin negatively regulates ovarian chemoresistance: compromised expression sensitizes ovarian cancer cells to paclitaxel. Tumour Biol 32:1031–1047 Reya T, Morrison SJ, Clarke MF, Weissman IL, 2001, Stem cells, cancer, and cancer stem cells. Nature 414:105–111 Yen TH, Wright NA, 2006, The gastrointestinal tract stem cell niche. Stem Cell Rev 2:203–212 Dave B, Chang J, 2009, Treatment resistance in stem cells and breast cancer. J Mammary Gland Biol Neoplasia 14:79–82 Douville J, Beaulieu R, Balicki D, 2009, ALDH1 as a functional marker of cancer stem and progenitor cells. Stem Cells Dev 18: 17–25 Benson DM Jr, Panzner K, Hamadani M, Hofmeister CC, Bakan CE, SmithMK, Elder P,KrughD, O’Donnell L, Devine SM, 2010, Effects of induction with novel agents versus conventional chemotherapy on mobilization and autologous stem cell transplant outcomes in multiple myeloma. Leuk Lymphoma 51:243–251 Riveros-Rosas H, Julian-Sanchez A, Pina E, 1997, Enzymology of ethanol and acetaldehyde metabolism in mammals. Arch Med Res 28:453–471 Ma I, Allan AL, 2011, The role of human aldehyde dehydrogenase in normal and cancer stem cells. Stem Cell Rev 7:292–306 Sladek NE, Kollander R, Sreerama L, Kiang DT, 2002, Cellular levels of aldehyde dehydrogenases, ALDH1A1 and ALDH3A1, as predictors of therapeutic responses to cyclophosphamide-based chemotherapy of breast cancer: a retrospective study, rational individualization of oxazaphosphorine-based cancer chemotherapeutic regimens. Cancer Chemoth Pharm 49:309–321 Tanei T, Morimoto K, Shimazu K, Kim SJ, Tanji Y, Taguchi T, TamakiY, Noguchi S, 2009, Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential Paclitaxel and epirubicin-based chemotherapy for breast cancers. Clin Cancer Res 15:4234–4241 Alison MR, Guppy NJ, Lim SM, Nicholson LJ, 2010, Finding cancer stem cells: are aldehyde dehydrogenases fit for purpose. J Pathol 222:335–354 Charafe-Jauffret E, Ginestier C, Iovino F, Tarpin C, Diebel M, Esterni B, Houvenaeghel G, Extra JM, Bertucci F, Jacquemier J, Xerri L, Dontu G, Stassi G, Xiao Y, Barsky SH, Birnbaum D, Viens P, Wicha MS, 2010, Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer. Clin Cancer Res 16:45–55 Chang B, Liu G, Xue F, Rosen DG, Xiao L, Wang X, Liu J, 2009, ALDH1 expression correlates with favorable prognosis in ovarian cancers. Mod Pathol 22:817–823 Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, Bronner-Fraser M, Kornblum HI, 2003, Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci U S A 100:15178–15183 Okabe M, Imai T, Kurusu M, Hiromi Y, Okano H, 2001, Translational repression determines a neuronal potential in Drosophila asymmetric cell division. Nature 411:94–98 Potten CS, Booth C, Tudor GL, Booth D, Brady G, Hurley P, Ashton G, Clarke R, Sakakibara S, Okano H, 2003, Identification of a putative intestinal stem cell and early lineage marker; musashi- 1. Differentiation 71:28–41 Nishimura S, Wakabayashi N, Toyoda K, Kashima K, Mitsufuji S, 2003, Expression of musashi-1 in human normal colon crypt cells: a possible stem cell marker of human colon epithelium. Dig Dis Sci 48:1523–1529 Gotte M, Greve B, Kelsch R, Muller-Uthoff H, Weiss K, Kharabi Masouleh B, SibrowskiW, Kiesel L, Buchweitz O, 2011, The adult stem cell marker musashi-1 modulates endometrial carcinoma cell cycle progression and apoptosis via notch-1 and p21(WAF1/CIP1). Int J Cancer 129:2042–2049 Okano H, Imai T, OkabeM, 2002, Musashi: a translational regulator of cell fate. J Cell Sci 115:1355–1359 Jiang F, Qiu Q, Khanna A, Todd NW, Deepak J, Xing L, Wang H, Liu Z, Su Y, Stass SA, Katz RL, 2009, Aldehyde dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer. Mol Cancer Res 7:330–338 Penumatsa K, Edassery SL, Barua A, Bradaric MJ, Luborsky JL, 2010, Differential expression of aldehyde dehydrogenase 1a1, ALDH1, in normal ovary and serous ovarian tumors. J Ovarian Res 3:28 Wang YC, Yo YT, Lee HY, Liao YP, Chao TK, Su PH, Lai HC, 2012, ALDH1-bright epithelial ovarian cancer cells are associated with CD44 expression, drug resistance, and poor clinical outcome. Am J Pathol 180:1159–1169 Deng S, Yang X, Lassus H, Liang S, Kaur S, Ye Q, Li C,Wang LP, Roby KF, Orsulic S, Connolly DC, Zhang Y, Montone K, Butzow R, Coukos G, Zhang L, 2010, Distinct expression levels and patterns of stem cell marker, aldehyde dehydrogenase isoform 1, ALDH1), in human epithelial cancers. PLoS One 5:e10277 Kuroda T, Hirohashi Y, Torigoe T, Yasuda K, Takahashi A, Asanuma H, Morita R, Mariya T, Asano T, Mizuuchi M, Saito T, Sato N, 2013, ALDH1-high ovarian cancer stem-like cells can be isolated from serous and clear cell adenocarcinoma cells, and ALDH1 high expression is associated with poor prognosis. PLoS One 8:e65158 Rahadiani N, Ikeda J, Mamat S,Matsuzaki S, Ueda Y, Umehara R, Tian T, Wang Y, Enomoto T, Kimura T, Aozasa K, Morii E, 2011, Expression of aldehyde dehydrogenase 1, ALDH1, in endometrioid adenocarcinoma and its clinical implications. Cancer Sci 102:903–908 Todaro M, Francipane MG, Medema JP, Stassi G, 2010, Colon cancer stem cells: promise of targeted therapy. Gastroenterology 138:2151–2162 TodaroM, Perez Alea M, Scopelliti A,Medema JP, Stassi G, 2008, IL-4-mediated drug resistance in colon cancer stem cells. Cell Cycle 7:309–313 Ma YH, Mentlein R, Knerlich F, Kruse ML, Mehdorn HM, Held- Feindt J, 2008, Expression of stem cell markers in human astrocytomas of different WHO grades. J Neurooncol 86:31–45 Moreira AL, Gonen M, Rekhtman N, Downey RJ, 2010, Progenitor stem cell marker expression by pulmonary carcinomas. Mod Pathol 23:889–895 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1133 EP 1140 DI 10.1007/s12253-015-9943-6 PG 8 ER PT J AU Zhou, L Yu, L Ding, G Chen, W Zheng, S Cao, L AF Zhou, Liangjing Yu, Leilai Ding, Guoping Chen, Wenchao Zheng, Sixin Cao, Liping TI Overexpressions of DLL4 and CD105 are Associated with Poor Prognosis of Patients with Pancreatic Ductal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DLL4 protein; Endoglin protein; Pancreatic neoplasms; Prognosis ID DLL4 protein; Endoglin protein; Pancreatic neoplasms; Prognosis AB The aim of this study was to investigate the expression of Delta-like ligand 4(DLL4) and Endoglin(CD105) labeled microvessel density(MVD) in pancreatic ductal adenocarcinoma (PDAC) and evaluate their correlation with major clinicopathologic features and patients’ survival. Forty-two pancreatic cancer and 20 normal pancreatic tissues were included in the study. Immunohistochemical staining was employed to assess the expression level of DLL4 both in tumor cells and stromal vascular endothelial cells, as well as CD105 which was used to determine MVD. The relationships of DLL4 and CD105 expression with clinicopathologic parameters and clinical outcome were evaluated. Both DLL4 and CD105-labeled microvessel were observed highly immunostained in PDAC cases, and high expression of DLL4 was positively correlated with MVD. Moreover, the high expression of DLL4 was significantly associated with histological grade, node stage and TNM stage in not only the cancer cells but also stroma; while high expression of CD105 was associated with histological grade, TNM stage, node stage and distant metastasis. In univariant analysis, patients with high expression of DLL4 and CD105 tended to significantly poorer overall survival. Both DLL4 and CD105 were overexpressed in a large proportion of patients with PDAC. The expression of DLL4 was positively correlated with CD105-labeled MVD, indicating DLL4 may involved in angiogenesis. In addition, high DLL4 and CD105 expression correlated with the poor clinical outcome and overall survival in patients with PDAC. C1 [Zhou, Liangjing] School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. [Yu, Leilai] School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. [Ding, Guoping] School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. [Chen, Wenchao] School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. [Zheng, Sixin] School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. [Cao, Liping] School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. RP Cao, L (reprint author), School of Medicine, Zhejiang University, Second Affiliated Hospital, Department of Hepatobiliary Surgery, 310006 Hangzhou, China. EM cao@zju.edu.cn CR Sarkar FH, Banerjee S, Li Y, 2007, Pancreatic cancer: pathogenesis, prevention and treatment. Toxicol Appl Pharmacol 224(3):326– 336 Jemal A, Bray F, Center MM et al, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Folkman J, 1995, Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med 1(1):27–31 Svagzdys S, Lesauskaite V, Pavalkis D et al, 2009, Microvessel density as a new prognostic marker after radiotherapy in rectal cancer. BMC Cancer 9:95 Uzzan B, Nicolas P, CucheratMet al, 2004)Microvessel density as a prognostic factor in womenwith breast cancer: a systematic reviewof the literature and meta-analysis. Cancer Res 64(9):2941– 2955 Zhao HC, Qin R, Chen XX et al, 2006, Microvessel density is a prognostic marker of human gastric cancer. World J Gastroenterol 12(47):7598–7603 Han H, Silverman JF, Santucci TS et al, 2001, Vascular endothelial growth factor expression in stage I non-small cell lung cancer correlates with neoangiogenesis and a poor prognosis. Ann Surg Oncol 8(1):72–79 Barau A, Ruiz-Sauri A, Valencia G et al, 2013, High microvessel density in pancreatic ductal adenocarcinoma is associated with high grade. Virchows Arch 462:541–546 van der Zee JA, van Eijck CH, HopWC et al, 2011, Angiogenesis: a prognostic determinant in pancreatic cancer? Eur J Cancer 47(17): 2576–2584 Kuiper P, Hawinkels LJAC, de Jonge-Muller ESM et al, 2011, Angiogenic markers endoglin and vascular endothelial growth factor in gastroenteropancreatic neuroendocrine tumors. World J Gastroenterol 17(2):219–225 Zhou W, Wang G, Guo S, 2013, Regulation of angiogenesis via Notch signaling in breast cancer and cancer stem cells. Biochim Biophys Acta 1836(2):304–320 Liu Z, Fan F, Wang A et al, 2014, Dll4-Notch signaling in regulation of tumor angiogenesis. J Cancer Res Clin Oncol 140(4):525– 536 Chen HT, Cai QC, Zheng JM et al, 2012, High expression of deltalike ligand 4 predicts poor prognosis after curative resection for pancreatic cancer. Ann Surg Oncol 19(Suppl 3):S464–S474 JubbAM, Soilleux EJ, Turley H et al, 2010, Expression of vascular notch ligand delta-like 4 and inflammatory markers in breast cancer. Am J Pathol 176(4):2019–2028 Linder S, Blasjo M, von Rosen A et al, 2001, Pattern of distribution and prognostic value of angiogenesis in pancreatic duct carcinoma: a semiquantitative immunohistochemical study of 45 patients. Pancreas 22(3):240–247 Mullendore ME, Koorstra JB, Li YM et al, 2009, Liganddependent Notch signaling is involved in tumor initiation and tumor maintenance in pancreatic cancer. Clin Cancer Res 15(7): 2291–2301 Ishigami S, Arigami T, Uenosono Y et al, 2013, Clinical implications of DLL4 expression in gastric cancer. J Exp Clin Cancer Res 32:46 Donnem T, Andersen S, Al-Shibli K et al, 2010, Prognostic impact of Notch ligands and receptors in non-small cell lung cancer: coexpression of Notch-1 and vascular endothelial growth factor-A predicts poor survival. Cancer 116:5676–5685 Kofler NM, Shawber CJ, Kangsamaksin T et al, 2011, Notch signaling in developmental and tumor angiogenesis. Genes Cancer 2(12):1106–1116 Oishi H, SunamuraM, Egawa S et al, 2010, Blockade of Delta-like ligand 4 signaling inhibits both growth and angiogenesis of pancreatic cancer. Pancreas 39(6):897–903 Sheldon H, Heikamp E, Turley H et al, 2010, New mechanism for Notch signaling to endothelium at a distance by Delta-like 4 incorporation into exosomes. Blood 116(13):2385–2394 Jacobsen TL, Brennan K, Arias AM et al, 1998, Cis-interactions between Delta and Notch modulate neurogenic signalling in Drosophila. Development 125(22):4531–4540 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1141 EP 1147 DI 10.1007/s12253-015-9937-4 PG 7 ER PT J AU Kalmar, A Peterfia, B Hollosi, P Wichmann, B Bodor, A Patai, V Scholler, A Krenacs, T Tulassay, Zs Molnar, B AF Kalmar, Alexandra Peterfia, Balint Hollosi, Peter Wichmann, Barnabas Bodor, Andras Patai, V Arpad Scholler, Andrea Krenacs, Tibor Tulassay, Zsolt Molnar, Bela TI Bisulfite-Based DNA Methylation Analysis from Recent and Archived Formalin-Fixed, Paraffin Embedded Colorectal Tissue Samples SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DNA methylation; Colorectal cancer; Genomic DNA isolation; Methylation-sensitive high-resolution melting analysis; Pyrosequencing ID DNA methylation; Colorectal cancer; Genomic DNA isolation; Methylation-sensitive high-resolution melting analysis; Pyrosequencing AB We aimed to test the applicability of formalin-fixed and paraffin-embedded (FFPE) tissue samples for gene specific DNA methylation analysis after using two commercially available DNA isolation kits. Genomic DNA was isolated from 5 colorectal adenocarcinomas and 5 normal adjacent tissues from "recent", collected within 6 months, and "archived", collected more than 5 years ago, FFPE tissues using either High Pure FFPET DNA Isolation kit or QIAamp DNA FFPE Tissue kit. DNA methylation analysis of MAL, SFRP1 and SFRP2 genes, known to be hypermethylated in CRC, was performed using methylation-sensitive high resolution melting (MS-HRM) analysis and sequencing. QIAamp (Q) method resulted in slightly higher recovery in archived (HP: 1.22± 3.18μg DNA; Q: 3.00±4.04μg DNA) and significantly (p<0.05) higher recovery in recent samples compared to High Pure method (HP) (HP: 4.10±2.91μg DNA; Q: 11.51 ±7.50μg DNA). Both OD260/280 and OD260/230 ratios were lower, but still high in the High Pure isolated archived and recent samples compared to those isolated with QIAamp. Identical DNA methylation patterns were detected for all 3 genes tested by MS-HRM with both isolation kits in the recent group. However, despite of higher DNA recovery in QIAamp slightly more reproducible methylation results were obtained from High Pure isolated archived samples. Sequencing confirmed DNA hypermethylation in CRCs. In conclusion, reproducible DNA methylation patterns were obtained from recent samples using both isolation kits. However, long term storage may affect the reliability of the results leading to moderate differences between the efficiency of isolation kits. C1 [Kalmar, Alexandra] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Peterfia, Balint] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Hollosi, Peter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Wichmann, Barnabas] Hungarian Academy of Sciences, Molecular Medicine Research Group, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Bodor, Andras] Eotvos Lorand University, Department of Physics of Complex Systems, Pazmany Peter setany 1/A, 1117 Budapest, Hungary. [Patai, V Arpad] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Scholler, Andrea] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. RP Kalmar, A (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM alexandra.kalmar@gmail.com CR Krijgsman O, Israeli D, Haan JC et al, 2012, CGH arrays compared for DNA isolated from formalin-fixed, paraffin-embeddedmaterial. Genes Chromosom Cancer 51(4):344–352., DOI 10.1002/gcc.21920 Lyons-Weiler M, Hagenkord J, Sciulli C et al, 2008, Optimization of the affymetrix GeneChip mapping 10K 2.0 assay for routine clinical use on formalin-fixed paraffin-embedded tissues. Diagn Mol Pathol Am J Sur Pathol Part B 17(1):3–13., DOI 10.1097/ PDM.0b013e31815aca30 Schweiger MR, Kerick M, Timmermann B et al, 2009, Genomewide massively parallel sequencing of formaldehyde fixed-paraffin embedded, FFPE, tumor tissues for copy-number- and mutationanalysis. PLoS One 4(5):e5548., DOI 10.1371/journal.pone.0005548 Kalmar A, Wichmann B, Galamb O et al, 2013, Gene expression analysis of normal and colorectal cancer tissue samples from fresh frozen and matched formalin-fixed, paraffin-embedded, FFPE, specimens after manual and automated RNA isolation. Methods 59(1):S16–19., DOI 10.1016/j.ymeth.2012.09.011 von Ahlfen S, Missel A, Bendrat K et al, 2007, Determinants of RNA quality from FFPE samples. PLoS One 2(12):e1261., DOI 10. 1371/journal.pone.0001261 Macabeo-Ong M, Ginzinger DG, Dekker N et al, 2002, Effect of duration of fixation on quantitative reverse transcription polymerase chain reaction analyses. Modern Pathol Off J US Canadian Acad Pathol Inc 15(9):979–987., DOI 10.1097/01.MP.0000026054. 62220.FC Solassol J, Ramos J, Crapez E et al, 2011, KRAS mutation detection in paired frozen and formalin-fixed paraffin-embedded, FFPE, colorectal cancer tissues. Int J Mol Sci 12(5):3191–3204., DOI 10. 3390/ijms12053191 Jemal A, v F, Center MM et al, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90., DOI 10.3322/caac.20107 Cancer Genome Atlas N, 2012, Comprehensive molecular characterization of human colon and rectal cancer. Nature 487(7407):330– 337., DOI 10.1038/nature11252 Caldwell GM, Jones C, Gensberg K et al, 2004, The Wnt antagonist sFRP1 in colorectal tumorigenesis. Cancer Res 64(3):883–888 Oberwalder M, Zitt M,Wontner C et al, 2008, SFRP2 methylation in fecal DNA–a marker for colorectal polyps. Int J Color Dis 23(1): 15–19., DOI 10.1007/s00384-007-0355-2 Wang DR, Tang D, 2008, Hypermethylated SFRP2 gene in fecal DNA is a high potential biomarker for colorectal cancer noninvasive screening. World J Gastroenterol WJG 14(4):524–531 Takeda M, Nagasaka T, Dong-Sheng S et al, 2011, Expansion of CpG methylation in the SFRP2 promoter region during colorectal tumorigenesis. Acta Med Okayama 65(3):169–177 Lind GE, Ahlquist T, Kolberg M et al, 2008, Hypermethylated MAL gene - a silent marker of early colon tumorigenesis. J Transl Med 6:13., DOI 10.1186/1479-5876-6-13 Wojdacz TK, Dobrovic A, 2007, Methylation-sensitive high resolution melting, MS-HRM): a new approach for sensitive and high-throughput assessment of methylation. Nucleic Acids Res 35(6): e41., DOI 10.1093/nar/gkm013 Kalmar A, Peterfia B, Wichmann B et al, 2015, Comparison of automated and manual DNA isolation methods for DNA methylation analysis of biopsy, fresh frozen, and formalin-fixed paraffinembedded colorectal cancer samples. Journal of laboratory automation., DOI 10.1177/2211068214565903 Smith TF, Waterman MS, 1981, Identification of common molecular subsequences. J Mol Biol 147(1):195–197 Gotoh O, 1982, An improved algorithm for matching biological sequences. J Mol Biol 162(3):705–708 Luo C, Tsementzi D, Kyrpides N et al, 2012, Direct comparisons of illumina vs. Roche 454 sequencing technologies on the same microbial community DNA sample. PLoS One 7(2):e30087., DOI 10. 1371/journal.pone.0030087 TCGA Research Network: http://cancergenome.nih.gov/ NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1149 EP 1156 DI 10.1007/s12253-015-9945-4 PG 8 ER PT J AU Korkmaz, TD Demirhan, O Abat, D Demirberk, B Tunc, E Kuleci, S AF Korkmaz, Tastemir Deniz Demirhan, Osman Abat, Deniz Demirberk, Bulent Tunc, Erdal Kuleci, Sedat TI Microchimeric Cells, Sex Chromosome Aneuploidies and Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sex chromosome aneuploidies; Microchimeric cells; Lung cancer; Bladder cancer ID Sex chromosome aneuploidies; Microchimeric cells; Lung cancer; Bladder cancer AB The phenomenon of feta-maternal microchimerisms inspires numerous questions. Many questions remain to be answered regarding this new avenue of genetics. The X and Y chromosomes have been associated with malignancy in different types of human tumors.We aimed to investigate the numerical aberrations of chromosomes X and Y in lung cancer (LC) and bladder cancer (BC) and review recent evidence for possible roles of microchimeric cells (McCs) in these cancers. We carried out cytogenetic analysis of the tumor and blood sampling in 52 cases of people with BC and LC, and also with 30 healthy people. A total of 48 (92.3 %) of the patients revealed sex chromosome aneuploidies (SCAs). A total SCAs was found in 9.8 % of 2282 cells that were analyzed as one or more cells in each case. The 68 and 95 SCAs were found in the 1952 (8.4 %) cells in peripheral blood, and 41 and 19 SCAs in the 330 (18.2 %) cells in the tumoral tissues respectively. There was a significant difference in the frequencies of SCAs between the patients and the control groups determined by the Fischer’s Exact Test (p<0.0001). The frequencies of SCAs were higher in the tumoral tissues than in the blood (p<0.0001). There was a significant difference in the frequencies of SCAs between the tumor and blood tissues, and this was higher in the tumor tissue (p<0.0001). In general, 78.9 % (41) of the 52 patients with LC and BC had X and Y chromosome monosomies. Largely a Y chromosome loss was present in 77.8 % of the men, and the 47, XXY karyotype was found in 33.3 % of them. The second most common SCA was monosomy X, and was found in 71.4 % of the women. McCs were observed in 26.9 % of the 52 patients, and the frequencies of McCs were higher in the blood than in the tissues (p<0.0001). XY cells were identified in the lung and bladder tissues of the women who had been pregnant with boys, but not in those who had not. There was a significant difference in the frequencies of McCs between the LC and BC patients (p<0.0005). We speculate that the microchimerism could have a general beneficial role in cancer, in which some sites may not be evident because of an allogeneic maternal immune reaction that hastens cancer development. A further understanding of McCs may help in anticipating its implications in cancer. Our results may suggest that SCAs may be contributing factors in the development of LC and BC, and aneuploidies of X and Y chromosomes play a role in the pathogenesis of cancers. C1 [Korkmaz, Tastemir Deniz] Adiyaman University, Vocational School of Health Services, AltinsehirAdiyaman, Turkey. [Demirhan, Osman] Cukurova University, Faculty of Medicine, Department of Medical Biology and Genetics, 01330 Adana, Turkey. [Abat, Deniz] Cukurova University, Faculty of Medicine, Department of UrologyAdana, Turkey. [Demirberk, Bulent] Adana Mental and Nervous diseases’ HospitalAdana, Turkey. [Tunc, Erdal] Cukurova University, Faculty of Medicine, Department of Medical Biology and Genetics, 01330 Adana, Turkey. [Kuleci, Sedat] Cukurova University, Faculty of Medicine, Department of Chest DiseasesAdana, Turkey. RP Demirhan, O (reprint author), Cukurova University, Faculty of Medicine, Department of Medical Biology and Genetics, 01330 Adana, Turkey. EM osdemir@cu.edu.tr CR Scheike O, Visfeldt J, Petersen B, 1973, Male breast cancer. 3. Breast cancer in association with Klinefelter syndrome. Acta Path Microbiol Scand A 81(3):352–358 Krasna I, Lee H, Sinilow ML, Sciorra P, Eierman L, 1992, Risk of malignancy in bilateral streak gonads: the role of the Y chromosome. J Pediatr Surg 27:1376–1380 Jacobs PA, Maloney V, Cooke R, Crolla JA, Ashworth A, Swerdlow AJ, 2013, Male breast cancer, age and sex chromosome aneuploidy. Br J Cancer 108:959–963 Riske CB, Morgan R, Ondreyco S, Sandberg AA, 1994, X and Y chromosome loss as sole abnormality in acute non-lymphocytic leukemia, ANLL). Cancer Genet Cytogenet 72:44–47 Khosrotehrani K, Bianchi DW, 2003, Fetal cell microchimerism: helpful or harmful to the parous woman? Curr Opin Obstet Gynecol 15:195–199 O’Donoghue K, Chan J, de la Fuente J, Kennea N, Sandison A, Anderson JR, Roberts IAG, Fisk NM, 2004, Microchimerism in female bone marrow and bone decades after fetal mesenchymal stem cell trafficking in pregnancy. Lancet 364:179–182 Sawicki JA, 2008, Fetal microchimerism and cancer. Cancer Res 68:9567–9569 Grigorova M, Lyman RC, Caldas C, Edwards PAW, 2005, Chromosome abnormalities in 10 lung cancer cell lines of the NCI-H series analyzed with spectral karyotyping. Cancer Genet Cytogenet 162:1–9 Watters AD, Going JJ, Grigor KM, Bartlett JM, 2002, Progression to detrusor-muscle invasion in bladder carcinoma is associated with polysomy of chromosomes 1 and 8 in recurrent pTa/pT1 tumors. Eur J Cancer 38:1593–1599 Ribal MJ, Alcaraz A, Mengual L, Carrio A, Lopez-Guillermo A, Mallofre C, Palou J, Gelabert A, Villavicencio H, 2004, Chromosomal high-polysomies predict tumor progression in T1 transitional cell carcinoma of the bladder. Eur Urol 45:593–599 RossMT, Grafham DV, Coffey AJ, Scherer S, McLay K,Muzny D, PlatzerM, Howell GR, Burrows C, Bird CP, Frankish A, Lovell FL, Howe KL, Ashurst JL, Fulton RS, Sudbrak R, Wen G, Jones MC, Hurles ME, Andrews TD et al, 2005, The DNA sequence of the human X chromosome. Nature 434:325–337 Piao Z, Malkhosyan SR, 2002, Frequent loss Xq25 on the inactive X chromosome in primary breast carcinomas is associated with tumor grade and axillary lymph node metastasis. Gene Chromosome Cancer 33:262–269 Choi C, Cho S, Horikawa I, Berchuck A, Wang N, Cedrone E, Jhung SW, Lee JB, Kerr J, Chenevix-Trench G, Kim S, Barrett JC, Koi M, 1997, Loss of heterozygosity at chromosome segment Xq25-26.1 in advanced human ovarian carcinomas. Gene Chromosome Cancer 20:234–242 Kersemaekers AM, van de Vijver MJ, Kenter GG, Fleuren GJ, 1999, Genetic alterations during the progression of squamous cell carcinomas of the uterine cervix. Gene Chromosome Cancer 26: 346–354 Sandberg AA, 1983, The X chromosome in human neoplasia, including sex chromatin and congenital conditions with Xchromosome anomalies. In: Sandberg AA, ed, Cytogenetics of the mammalian X chromosome, part B: X chromosome anomalies and their clinical manifestations. Alan R. Liss, New York, pp 459– 498 Visakorpi T, Hyytinen E, Kallioniemi A, Isola J, Kallioniemi OP, 1994, Sensitive detection of chromosome copy number aberrations in prostate cancer by fluorescence in situ hybridization. AmJ Pathol 145:624–630 Wang MG, Zakut R, Yi H, Rosenberg S, McBride OW, 1994, Localization of the MAGE1 gene encoding a human melanoma antigen to chromosome Xq28. Cytogenet Cell Genet 67:116–119 Gough NM, Gearing DP, Nicola NA, Baker E, Pritchard M, Callen DF, Sutherland GR, 1990, Localization of the human GM-CSF receptor gene to the X-Y pseudoautosomal region. Nature 345: 734–736 Beck TW, Huleihel M, Gunnell M, Bonner TI, Rapp UR, 1987, The complete coding sequence of the human A-raf-1 oncogene and transforming activity of ahuman A-raf carrying retrovirus. Nucleic Acids Res 15:595–609 Rao VN, Huebner K, IsobeM, ar-Rushdi A, Croce CM, Reddy ES, 1989, Elk, tissue-specific ets-related genes on chromosomes X and 14 near translocation breakpoints. Science 244:66–70 Noguchi T, Mattei MG, Oberle I, Planche J, Imbert J, Pelassy C, Birg F, Birnbaum D, 1987, Localization of the mcf.2 transforming sequence to the X chromosome. EMBO J 6:1301–1307 Brothman AR, Maxwell TM, Cui J, Deubler DA, Zhu XL, 1999, Chromosomal clues to the development of prostate tumors. Prostate 38:303–312 Jordan JJ, Hanlon AL, Greenberg RE, Al-Saleem TI, Tricoli JV, 2001, Loss of the short arm of the Y chromosome in human prostate carcinoma. Cancer Genet Cytogenet 124:122–126 Vijayakumar S, Garcia D, Hensel CH, Banerjee M, Bracht T, Xiang R, Kagan J, Naylor SL, 2005, The human Y chromosome suppresses the tumorigenicity of PC-3, a human prostate cancer cell line, in a thymic nude mice. Gene Chromosome Cancer 44:365– 372 TeixeiraMR, Pandis N, Dietrich CU, ReedW, Andersen J, Qvist H, Heim S, 1998, Chromosome banding analysis of gynecomastias and breast carcinomas in men. Gene Chromosome Cancer 23:16– 20 Rudas M, Schmidinger M, Wenzel C, Okamoto I, Budinsky A, Fazeny B, Marosi C, 2000, Karyotypic findings in two cases of male breast cancer. Cancer Genet Cytogenet 121:190–193 Wallrapp C, Hahnel S, Boeck W, Soder A, Mincheva A, Lichter P, Leder G, Gansauge F, Sorio C, Scarpa A, Gress TM(2001, Loss of the Y chromosome is a frequent chromosomal imbalance in pancreatic cancer and allows differentiation to chronic pancreatitis. Int J Cancer 91:340–344 Powell I, Tyrkus M, Kleer E, 1990, Apparent correlation of sex chromosome loss and disease course in urothelial cancer. Cancer Genet Cytogenet 50:97–101 Mohanty D, 2004, Sex chromosome loss and malignancy: Does a relationship established?.Indian. J Hum Genet 10:3–4 Betz J, Meloni AM, Sandberg AA, 1996, FISH studies on the Y chromosome in male urinary cells. Cancer Genet Cytogenet 88: 155–157 Sauter G, Moch H,Wagner U, Novotna H, Gasser TC,Mattarelli G, Mihatsch MJ, Waldman FM, 1995, Y chromosome loss detected by FISH in bladder cancer. Cancer Genet Cytogenet 82:163–169 Sauter G, Moch H, Gasser TC, Mihatsch MJ,Waldman FM, 1995, Heterogeneity of chromosome 17 and erbB-2 gene copy number in primary and metastatic bladder cancer. Cytometry 21:40–46 Pierre RV, Hoagland HC, 1972, Age-associated aneuploidy: Loss of Y chromosome from human bonemarrow cells with aging. Cancer 30:889–894 Sakurai M, Sandberg AA, 1976, The chromosomes and causation of human caner and leukemia XVIII. The missing Y in acute myeloblastic leukemia and Ph1-positive chronic myelocytic leukemia [CML]. Cancer 38:762–769 United Kingdom Cancer Cytogenetics Group, UK CCG,, 1992, Loss of the Y chromosome from normal and neoplastic bonemarrows. Gene Chromosome Cancer 5:83–88 Mertens F, Johansson B, Hoglund M, Mitelman F, 1997, Chromosomal imbalance maps of malignant solid tumors: a cytogenetic survey of 3185 neoplasms. Cancer Res 57:2765–2780 El-Naggar A, Dinh M, Tucker SL, Swanson D, Steck K, Vielh P, 1999, Numerical chromosomal changes in DNA hypodiploid solid tumors: restricted loss and gain of certain chromosomes. Cytometry 37:107–112 Nilbert M, Heim S, 1990, Uterine leiomyoma cytogenetics. Gene Chromosome Cancer 2:3–13 Mark J, Havel G, Grepp C, Dahlenfors R, Wedell B, 1990, Chromosomal patterns in human benign uterine leiomyomas. Cancer Genet Cytogenet 44:1–13 Vanni R, Lecca U, Faa G, 1991, Uterine leiomyoma cytogenetics. II. Report of forty cases. Cancer Genet Cytogenet 53:247–256 Witek A, Skalba P, ZiebaM(2001, Pituitary tumor in a woman with a 47, XXX karyotype–case report. Med Sci Monit 7:304–330 Gul D, Akin R, Kismet E, 2003, Neuroblastoma in a patient with 47, XXX karyotype. Cancer Genet Cytogenet 146:84–85 Swerdlow AJ, Schoemaker MJ, Higgins C,Wright AF, Jacobs PA, UK Clinical Cytogenetics Group, 2005, Cancer incidence andmortality in patients with Klinefelter’s syndrome: a cohort study. J Natl Cancer Inst 97:1204–1210 Brown CJ, Greally JM, 2003, A stain upon the silence: genes escaping X inactivation. Trends Genet 19:432–438 Nelson JL, 2001, Microchimerism: expanding new horizon in human health or incidental remnant of pregnancy? Lancet 358:2011– 2012 Gadi VK, Nelson JL, 2007, Fetal microchimerism in women with breast cancer. Cancer Res 67:9035–9038 O’Donoghue K, Sultan HA, Al-Allaf FA, Anderson JR, Wyatt- Ashmead J, Fisk NM, 2008, Microchimeric fetal cells cluster at sites of tissue injury in lung decades after pregnancy. Reprod Biomed Online 16:382–390 Li L, Neaves WB, 2006, Normal stem cells and cancer stem cells: the niche matters. Cancer Res 66:4553–4557 Cha DH, Khosrotehrani K, Kim Y, Stroh H, Bianchi DW, Johnson CL, 2003, Cervical cancer and microchimerism. Obstet Gynecol 102:774–781 Kamper-Jorgensen M, Biggar RJ, Tjonneland A, Hjalgrim H, Kroman N, Rostgaard K, Stamper CL, Olsen A, Andersen AM, Gadi VK, 2012, Opposite effects of microchimerism on breast and colon cancer. Eur J Cancer 48:2227–2235 Srivatsa B, Srivatsa S, Johnson KL, Bianchi DW, 2003, Maternal cell microchimerism in newborn tissues. J Pediatr 142:31–35 BianchiDW,Williams JM, Sullivan LM, Hanson FW, KlingerKW, Shuber AP, 1997, PCR quantitation of fetal cells in maternal blood in normal and aneuploid pregnancies. Am J Hum Genet 61:822– 882 Srivatsa B, Srivatsa S, Johnson KL, Samura O, Lee SL, Bianchi DW, 2001, Microchimerism of presumed fetal origin in thyroid specimens from women: a case-control study. Lancet 358:2034– 2038 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1157 EP 1165 DI 10.1007/s12253-015-9934-7 PG 9 ER PT J AU Moizs, M Bajzik, G Lelovics, Zs Strausz, J Rakvacs, M Zadori, P Kovacs, Repa, I AF Moizs, Mariann Bajzik, Gabor Lelovics, Zsuzsanna Strausz, Janos Rakvacs, Marianna Zadori, Peter Kovacs, Arpad Repa, Imre TI Characterization of Individuals Taking Part in Low Dose Computed Tomography (LDCT) Screening Program SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Lung screening; Participation rate; Hungary ID Lung cancer; Lung screening; Participation rate; Hungary AB In the past years the participation rate in conventional voluntary x-ray lung screening has been around 22%in Somogy County in Hungary. Due to the high morbidity and mortality rates of lung cancer, low participation rate of the high risk individuals on the screening is a primary question in Hungary. To obtain an effectively high level of participation in our ongoing low dose CT screening program, we had to emphasize the benefits of participation for the targeted individuals. As a first step, our aim was to gather information on the aspects affecting the individuals’ will for participation.We used the most accessible source of information: individuals over the age of 50, who attended the conventional voluntary lung screening, were approached to fill a questionnaire on their habits relating to smoking, health issues and their prior participation of lung screening. 1080 adults anonymously completed the questionnaire. Analyzing the results, beside other findings, we found a unique variable factor, which altered negatively the compliance for the screening: older individuals, who started participating in the screening in obligation to the health regulations, took part in the voluntary screening programs at a significantly lower rate. Our findings led us to better understanding the complexity of decision making affecting the individual’s participation and attitudes toward health issues. Trial registration: IG/03833/2012. C1 [Moizs, Mariann] Kaposi Mor Teaching Hospital, Tallian Gyula u. 20-32, H-7400 Kaposvar, Hungary. [Bajzik, Gabor] Kaposvar University, Department of Radiation Oncology, Guba Sandor u. 40, H-7400 Kaposvar, Hungary. [Lelovics, Zsuzsanna] Kaposi Mor Teaching Hospital, Tallian Gyula u. 20-32, H-7400 Kaposvar, Hungary. [Strausz, Janos] Kaposi Mor Teaching Hospital, Tallian Gyula u. 20-32, H-7400 Kaposvar, Hungary. [Rakvacs, Marianna] Kaposi Mor Teaching Hospital, Tallian Gyula u. 20-32, H-7400 Kaposvar, Hungary. [Zadori, Peter] Kaposvar University, Department of Radiation Oncology, Guba Sandor u. 40, H-7400 Kaposvar, Hungary. [Kovacs, Arpad] Kaposvar University, Department of Radiation Oncology, Guba Sandor u. 40, H-7400 Kaposvar, Hungary. [Repa, Imre] Kaposi Mor Teaching Hospital, Tallian Gyula u. 20-32, H-7400 Kaposvar, Hungary. RP Repa, I (reprint author), Kaposi Mor Teaching Hospital, H-7400 Kaposvar, Hungary. EM repa.imre@sic.ke.hu CR NCCN, 2012, National Comprehensive Cancer Network: Lung Cancer Screening Guidelines. Ver. 1 IARC, 2008, Fact Sheet, 2008. IOP Publishing PhysicsWeb. http:// globocan.iarc.fr/factsheet.asp. Accessed 23 Oct 2013 OECD, 2012, Health at a Glance: Europe 2012. IOP Publishing PhysicsWeb. http://www.oecd-ilibrary.org/sites/9789264183896- en/01/05/index.html?contentType=/ns/Chapter, / n s/ StatisticalPublication&itemId=/content/chapter/9789264183896-8- en&containerItemId=/content/serial/23056088&accessItemIds= &mimeType=text/html. Accessed 23 Oct 2013 La Vecchia C, Bosetti C, Lucchini F, Bertuccio P, Negri E, Boyle P, Levi F, 2010, Cancer mortality in Europe, 2000–2004, and an overview of trends since 1975. Ann Oncol 21:1323–1360 Ostoros G, 2013, Lung cancer. Koranyi Bull 1:24–31, In Hung Kovacs G, 2008, The 50 years of the chest X-ray screeing in Hungary. Med Thorac 61:76–89, In Hung McLoud TC, 2011, Initial results of the national lung cancer screening trial. Cancer Imaging 11:S85 Rampinelli C, Preda L, Maniglio M, Sirica L, Travaini LL,Veronesi G, Bellomi M, 2011, Extrapulmonary malignancies detected at lung cancer screening. Radiology 261:293–299 Pastorino U, Rossi M, Rosato V, Marchiano A, Sverzellati N, Morosi C, Fabbri A, Galeone C, Negri E, Sozzi G, Pelosi G, La Vecchia C, 2012, Annual or biennial CTscreening versus observation in heavy smokers: 5-year results of the MILD trial. Eur J Cancer Prev 21:308–315 Moizs M, Bajzik G, Lelovics Z, Rakvacs M, Strausz J, Repa I, 2013, First result of differentiated communication—to smokers and non-smokers—in order to increase the voluntary participation rate in lung screening. BMC Public Health 13:914 Moizs M, Bajzik G, Lelovics ZS, Rakvacs M, Strausz J, Repa I, 2014, Preliminary experiences of introducing low dose CT lung cancer screening in Hungary. Orv Hetil 155:383–388, In Hung Callol L, Roig F, Cuevas A, de Granda JI, Villegas F, Jareno J et al, 2007, Low-dose CT: a useful and accessible tool for the early diagnosis of lung cancer in selected populations. Lung Cancer 56: 217–221 Menezes RJ, Roberts HC, Paul NS, McGregor M, Chung TB, Patsios D, Weisbrod G, Herman S, Pereira A, McGregor A, Dong Z, Sitartchouk I, Boerner S, Tsao MS, Keshavjee S, Shepherd FA, 2010, Lung cancer screening using low-dose computed tomography in at-risk individuals: the Toronto experience. Lung Cancer 67: 177–183 US Centers for Disease Control and Prevention, 2010). Health behaviors of adults: United States, 2005-2007. Vital and Health Statistics, Series 10, Number 245, Appendix II, p. 80 National Lung Screening Trial Research Team, Aberle DR, Adams AM, Berg CD, Clapp JD, Clingan KL, Gareen IF, Lynch DA, Marcus PM, Pinsky PF, 2010, Baseline characteristics of participants in the randomized national lung screening trial. J Natl Cancer Inst 102:1771–1779 Bach PB, Mirkin JN, Oliver TK, Azzoli CG, Berry DA, Brawley OW, Byers T, Colditz GA, Gould MK, Jett JR, Sabichi AL, Smith- Bindman R, Wood DE, Qaseem A, Detterbeck FC, 2012, Benefits and harms of CT screening for lung cancer. A systematic review. JAMA 307:2418–2429 NLST, 2011, The national lung screening trial research team: reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med 365:395–409 Wender R, Fontham ETH, Barrera E, Colditz GA, Church TR, Ettinger DS, Etzioni R, Flowers CR, Gazelle GS, Kelsey DK, LaMonte SJ, Michaelson JS, Oeffinger KC, Shih YC, Sullivan DC, Travis W, Walter L, Wolf AM, Brawley OW, Smith RA, 2013, American cancer society lung cancer screening guidelines. CA Cancer J Clin 63:107–117 Marshall HM, Bowman RV, Yang IA, Fong KM, Berg CD, 2013, Screening for lung cancer with low-dose computed tomography: a review of current status. J Thorac Dis 5:S524–S539 Tammemagi MC, Lam S, 2014, Screening for lung cancer using low dose computed tomography. BMJ 348:g2253 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1167 EP 1173 DI 10.1007/s12253-015-9929-4 PG 7 ER PT J AU Nikkuni, O Kaira, K Toyoda, M Shino, M Sakakura, K Takahashi, K Tominaga, H Oriuchi, N Suzuki, M Iijima, M Asao, T Nishiyama, M Nagamori, Sh Kanai, Y Oyama, T Chikamatsu, K AF Nikkuni, Osamu Kaira, Kyoichi Toyoda, Minoru Shino, Masato Sakakura, Koichi Takahashi, Katsumasa Tominaga, Hideyuki Oriuchi, Noboru Suzuki, Masami Iijima, Misa Asao, Takayuki Nishiyama, Masahiko Nagamori, Shushi Kanai, Yoshikatsu Oyama, Tetsunari Chikamatsu, Kazuaki TI Expression of Amino Acid Transporters (LAT1 and ASCT2) in Patients with Stage III/IV Laryngeal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Larynx; LAT1; CD98; Imunohistochemistry; ASCT2; Prognostic factor ID Larynx; LAT1; CD98; Imunohistochemistry; ASCT2; Prognostic factor AB The aim of this study is to evaluate the clinicopathological significance of L-type amino acid transporter 1 (LAT1) expression in patients with advanced laryngeal squamous cell carcinoma (LSCC). A total of 73 patients with advanced LSCC were retrospectively reviewed. Tumor sections were stained by immunohistochemistry for LAT1, 4F2hc, system ASC amino acid transporter-2 (ASCT2), cell proliferation by Ki-67, microvessel density (MVD) determined by CD34 and p53. A positive LAT1, 4F2hc and ASCT2 expression (staining more than a quarter) in the primary sites were recognized in 85, 80 and 45 %, respectively, and a high LAT1, 4F2hc and ASCT2 expression (staining more than a half) yielded 48, 31 and 18 %, respectively. High expression of LAT1 was significantly associated with lymph node metastasis, 4F2hc, ASCT2, Ki-67 and p53. The expression of LAT1 was significantly correlated with ASCT2, 4F2hc, cell proliferation, and MVD. By univariate analysis, there was no statistically significant relationship between LAT1 expression and prognosis in advanced LSCC. LAT1, 4F2hc and ASCT2 were highly expressed in patients with advanced laryngeal cancer. Our study suggests that the expression of LAT1 plays a crucial role in the metastasis and tumor progression in advanced LSCC. C1 [Nikkuni, Osamu] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Japan. [Kaira, Kyoichi] Gunma University, Graduate School of Medicine, Department of Oncology Clinical Development, Showa-machi, 371-8511 Maebashi, Japan. [Toyoda, Minoru] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Japan. [Shino, Masato] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Japan. [Sakakura, Koichi] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Japan. [Takahashi, Katsumasa] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Japan. [Tominaga, Hideyuki] Fukushima Medical University, Advanced Clinical Research CenterFukushima, Japan. [Oriuchi, Noboru] Gunma University, Graduate School of Medicine, Department of Diagnostic Radiology and Nuclear medicineMaebashi, Japan. [Suzuki, Masami] Gunma Prefectural Cancer Center, Department of Head & Neck SurgeryOta, Japan. [Iijima, Misa] Gunma Prefectural Cancer Center, Department of Pathology and Clinical LaboratoriesOta, Japan. [Asao, Takayuki] Gunma University, Graduate School of Medicine, Department of Oncology Clinical Development, Showa-machi, 371-8511 Maebashi, Japan. [Nishiyama, Masahiko] Gunma University, Graduate School of Medicine, Department of Molecular Pharmacology and OncologyMaebashi, Japan. [Nagamori, Shushi] Osaka University, Graduate School of Medicine, Division of Bio-system PharmacologyOsaka, Japan. [Kanai, Yoshikatsu] Osaka University, Graduate School of Medicine, Division of Bio-system PharmacologyOsaka, Japan. [Oyama, Tetsunari] Gunma University, Graduate School of Medicine, Department of Diagnostic PathologyMaebashi, Japan. [Chikamatsu, Kazuaki] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck SurgeryMaebashi, Japan. RP Kaira, K (reprint author), Gunma University, Graduate School of Medicine, Department of Oncology Clinical Development, 371-8511 Maebashi, Japan. EM kkaira1970@yahoo.co.jp CR Ragin CC, Modugno F, Gollin SM, 2007, The epidemiology and risk factors of head and neck cancer: a focus on human papillomavirus. J Dent Res 86:104–114 Thephamongkhol K, Browman G, Hodson I, Oliver T, Zuraw L, members of the Head and Neck Cancer Disease Site Group, 2004, Chemotherapy with radiotherapy for nasopharygeal cancer: a clinical practice guidelineevidence-based Series #5-7. Cancer Care Ontario, Program in Evidence-Based Care, Toronto Christensen HN, 1990, Role of amino acid transport and countertransport in nutrition and metabolism. Physiol Rev 70:43– 77 Fuchs BC, Bode BP, 2006, Amino acid transporters ASCT2 and LAT1 in cancer: partners in crime? Semin Cancer Biol 15:254–266 Kanai Y, Segawa H, Miyamoto K et al, 1998, Expression cloning and characterization of a transporter for large neutral amino acids activated by the heavy chain of 4F2 antigen, CD98). J Biol Chem 273:23629–23632 Yanagida O, Kanai Y, Chairoungdua A et al, 2001, Human L-type amino acid transporter 1, LAT 1): characterization of function and expression in tumor cell lines. BiochimBiophys Acta 1514:291–302 Nawashiro H, Otani N, ShinomiyaN et al, 2006, L-type amino acid transporter 1 as a potential molecular target in human astrocytic tumors. Int J Cancer 119:484–492 Kim CS, Cho SH, Chun HS et al, 2008, BCH, an inhibitor of system L amino acid transporters, induces apoptosis in cancer cells. Biol Pharm Bull 31:1096–1100 Kim CS, Moon IS, Park JH et al, 2010, Inhibition of L-type amino acid transporter modulates the expression of cell cycle regulatory factors in KB oral cancer cells. Biol Pharm Bull 33:1117–1121 Kekuda R, Prasad PD, Fei YJ et al, 1996, Cloning of the sodiumdependent, broad-scope, neutral amino acid transporter Bo from a human placental choriocarcinoma cell line. J Biol Chem 271: 18657–18661 Fuch BC, Finger RE, Onan MC et al, 2007, ASCT2 silencing regulates mammalian target of rapamycin growth and survival signaling in human hepatoma cells. Am J Physiol Cell Physiol 293: C55–C63 Kobayashi H, Ishii Y, Takayama T, 2005, Expression of L-type amino acid transporter 1, LAT1, in esophageal carcinoma. J Surg Oncol 90:233–238 Kaira K, Oriuchi N, Imai H et al, 2008, Prognostic significance of L-type amino acid transporter 1 expression in resectable stage I-III nonsmall cell lung cancer. Br J Cancer 98:742–748 Nakanishi K, Ogata S, Matsuo H et al, 2007, Expression of LAT1 predicts risk of progression of transitional cell carcinoma of the upper urinary tract. Virchows Arch 451:681–690 Sakata T, Ferdous G, Tsuruta T et al, 2009, L-type amino acid transporter 1 as a novel biomarker for high-grade malignancy in prostate cancer. Pathol Int 59:7–18 IchinoeM, Mikami T, Yoshida T et al, 2011, High expression of Ltype amino-acid transporter 1, LAT1, in gastric carcinomas: comparison with non-cancerous lesions. Pathol Int 61:281–289 Furuya M, Horiguchi J, Nakajima H et al, 2012, Correlation of Ltype amino acid transporter 1 and CD98 expression with triple negative breast cancer prognosis. Cancer Sci 103:382–389 Kaira K, Oriuchi N, Takahashi T et al, 2011, LAT1 expression is closely associated with hypoxic markers and mTOR in resected non-small cell lung cancer. Am J Transl Res 3:468–478 Kaira K, Sunose Y, Arakawa K et al, 2012, Prognostic significance of L-type amino acid transporter 1 expression in surgically resected pancreatic cancer. Br J Cancer 107:632–638 Shimizu K, Kaira K, Tomizawa Y et al, 2014, ASC amino acid transporter 2, ASCT2, as a novel prognostic marker in non-small cell lung cancer. Br J Cancer 110:2030–2039 ToyodaM, Kaira K, OhshimaYet al, 2014, Prognostic significance of amino acid transporter expression, LAT1, ASCT2 and xCT, in surgically resected tongue cancer. Br J Cancer 110:2506–2513 Li R, Younes M, Frolov A et al, 2003, Expression of neutral amino acid transporter ASCT2 in human prostate. Anticancer Res 23: 3413–3418 Whitte D, Ali N, Carlson N et al, 2002, Overexpression of the neutral amino acid transporter ASCT2 in human colorectal adenocarcinoma. Anticancer Res 22:2555–2557 Kaira K, Oriuchi N, Imai H et al, 2009, CD98 expression is associated with poor prognosis in resected non-small-cell lung cancer with lymph node metastases. Ann Surg Oncol 16:3473–3481 Kaira K, Oriuchi N, Imai H et al, 2009, Prognostic significance of L-type amino acid transporter 1, LAT1, and 4F2 heavy chain, CD98, expression in stage I pulmonary adenocarcinoma. Lung Cancer 66:120–126 Namikawa M, Kakizaki S, Kaira K, et al, 2014, Expression of amino acid transporters, LAT1, ASCT2 and xCT, as clinical significance in hepatocellular carcinoma. Hepatol Res in press Hassanein M, Hoeksema MD, Shiota M et al, 2013, SLC1A5 mediates glutamine transport required for lung cancer cell growth and survival. Clin Cancer Res 19:560–570 Imai H, Kaira K, Oriuchi N et al, 2010, Inhibition of L-type amino acid transporter 1 has antitumor activity in non-small cell lung cancer. Anticancer Res 30:4819–4828 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1175 EP 1181 DI 10.1007/s12253-015-9954-3 PG 7 ER PT J AU Stahl, RP Schnellert, J Koop, Ch Simon, R Marx, A Izbicki, RJ Sauter, G Quaas, A AF Stahl, R Phillip Schnellert, Jessica Koop, Christina Simon, Ronald Marx, Andreas Izbicki, R Jakob Sauter, Guido Quaas, Alexander TI Determination of Tumor Heterogeneity in Colorectal Cancers Using Heterogeneity Tissue Microarrays SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tissue microarrays; Colorectal cancer; Heterogeneity; p53; HER2 ID Tissue microarrays; Colorectal cancer; Heterogeneity; p53; HER2 AB Cancer is often heterogeneous both on a morphological and on a genetic level. Though resected tumors are often large, molecular tumor analysis is usually restricted to one tissue block. In this project we introduce a new tool for a highthroughput heterogeneity analysis of colorectal cancer. A heterogeneity tissuemicroarray (TMA) wasmanufactured fromtissues of 340 patients with colorectal cancer. For this purpose 8 different tissue spots were taken from as many different cancer blocks per patient as possible (at least 4 different blocks). Additional tissue samples from 1 to 4 corresponding lymph node metastases were added from 134 patients. The systemwas then validated by analysing one parameter each known for minimal (p53) or substantial (HER2) heterogeneity in colorectal cancer. P53 alterations as detected by immunohistochemistry were seen in 174 (51.3%) of 339 analyzable primary tumors of which 23 (13.2% of positive cases) showed a heterogeneous distribution pattern. HER2 overexpression was seen in 18 (5.4 %) of 336 evaluable tumors. HER2 amplification occurred in 6 (33.3 %) of the 18 cases with HER2 overexpression. Genomic heterogeneity was more prevalent for HER2 alterations than for p53 alterations. For immunohistochemical expression analysis, 16 of 18 positive cases were heterogeneous (88.9 %) and for amplification 3 of 6 cases (50 %) were heterogeneous. Large section validation revealed, however a considerable fraction of heterogeneous cases were due to technical artifacts. In summary, our data suggest, that heterogeneity TMAs are a powerful tool to rapidly screen for molecular heterogeneity in colorectal cancer. C1 [Stahl, R Phillip] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. [Schnellert, Jessica] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. [Koop, Christina] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. [Simon, Ronald] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. [Marx, Andreas] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. [Izbicki, R Jakob] University Medical Center Hamburg-Eppendorf, General, Visceral and Thoracic Surgery Department, Martinistrasse 52, 20246 Hamburg, Germany. [Sauter, Guido] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. [Quaas, Alexander] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. RP Stahl, RP (reprint author), University Medical Center Hamburg Eppendorf, Pathology, 20246 Hamburg, Germany. EM p.stahl@uke.de CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90 Lombardi L, Morelli F, Cinieri S et al, 2010, Adjuvant colon cancer chemotherapy: where we are and where we’ll go. Cancer Treat Rev 36:S34–S41 Van Cutsem E, Kohne CH, Hitre E et al, 2009, Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med 360:1408–1417 Fearon ER, Vogelstein B, 1990, A genetic model for colorectal tumorigenesis. Cell 61:759–767 Vousden KH, Prives C, 2009, Blinded by the light: the growing complexity of p53. Cell 137:413–431 Levine AJ, Oren M, 2009, The first 30 years of p53: growing ever more complex. Nat Rev Cancer 9:749–758 Russo A, Bazan V, Iacopetta B et al, 2005, The TP53 colorectal cancer international collaborative study on the prognostic and predictive significance of p53mutation: influence of tumor site, type of mutation, and adjuvant treatment. J Clin Oncol 23:7518–7528 Munro AJ, Lain S, Lane DP, 2005, P53 abnormalities and outcomes in colorectal cancer: a systematic review. Br J Cancer 92: 434–444 Jourdan F, Sebbagh N, Comperat E et al, 2003, Tissue microarray technology: validation in colorectal carcinoma and analysis of p53, hMLH1, and hMSH2 immunohistochemical expression. Virchows Arch 443:115–121 Scott N, Sagar P, Stewart J, Blair GE, Dixon MF, Quirke P, 1991, p53 in colorectal cancer: clinicopathological correlation and prognostic significance. Br J Cancer 63:317–319 Rubin I, Yarden Y, 2001, The basic biology of HER2. Ann Oncol 12:S3–S8 Slamon DJ, Leyland-Jones B, Shak S et al, 2001, Use of chemotherapy and a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2.NEngl JMed 344:783–792 Bang YJ, Van Cutsem E, Feyereislova A et al, 2010, Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer, ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376:687–697, Epub 2010 Aug 19. Erratum in: Lancet. Oct 16;376(9749):1302 Tapia C, Glatz K, Novotny H et al, 2007, Close association between HER-2 amplification and overexpression in human tumors of non-breast origin. Mod Pathol 20:192–198 Hu Y, Bandla S, Godfrey TE et al, 2011, HER2 amplification, overexpression and score criteria in esophageal adenocarcinoma. Mod Pathol 24:899–907 WilliamsMD, Roberts DB, KiesMS,Mao L,Weber RS, El-Naggar AK, 2010, Genetic and expression analysis of HER-2 and EGFR genes in salivary duct carcinoma: empirical and therapeutic significance. Clin Cancer Res 16:2266–2274 Burandt E, Schreiber M, Stein A et al, 2014, Continuous tissue microarray based identification of cancers with homogeneous target expression for successful targeted therapy in clinical routine practice. Genes Chromosomes Cancer 53:228–239 Ramanathan RK, Hwang JJ, Zamboni WC et al, 2004, Low overexpression of HER-2/neu in advanced colorectal cancer limits the usefulness of trastuzumab, Herceptin, and irinotecan as therapy. A phase II trial. Cancer Investig 22:858–865 Saunders NA, Simpson F, Thompson EW et al, 2012, Role of intratumoural heterogeneity in cancer drug resistance: molecular and clinical perspectives. EMBO Mol Med 4:675–684 Minner S, Gartner R, Freudenthaler F et al, 2013, Marked heterogeneity of ERG expression in large primary prostate cancers. Mod Pathol 26(1):106–116 Simon R, Mirlacher M, Sauter G, 2005, Tissue microarrays. Methods Mol Med 114:257–268 Rosen DG, Huang X, Deavers MT, Malpica A, Silva EG, Liu J, 2004, Validation of tissue microarray technology in ovarian carcinoma. Mod Pathol 17:790–797 Graham AD, Faratian D, Rae F, Thomas JS, 2008, Tissue microarray technology in the routine assessment of HER-2 status in invasive breast cancer: a prospective study of the use of immunohistochemistry and fluorescence in situ hybridization. Histopathology 52:847–855 Bhargava R, Lal P, Chen B, 2004, Feasibility of using tissue microarrays for the assessment of HER-2 gene amplification by fluorescence in situ hybridization in breast carcinoma. Diagn Mol Pathol 13:213–216 Sauter G, Lee J, Barlett JM, Slamon DJ, Press MF, 2009, Guidelines for human epidermal growth factor receptor 2 testing: biologic and methodologic considerations. J Clin Oncol 27:1323– 1333 Fernebro E, Dictor M, Bendahl PO, Ferno M, Nilbert M, 2002, Evaluation of the tissue microarray technique for immunohistochemical analysis in rectal cancer. Arch Pathol Lab Med 126: 702–705 Kountourakis P, Pavlakis K, Psyrri A et al, 2006, Clinicopathologic significance of EGFR and Her-2/neu in colorectal adenocarcinomas. Cancer J 12:229–236 Nathanson DR, Culliford AT, Shia J et al, 2003, HER 2/neu expression and gene amplification in colon cancer. Int J Cancer 105:796–802 Marx AH, Burandt EC, Choschzick M et al, 2010, Heterogenous high-level HER-2 amplification in a small subset of colorectal cancers. Hum Pathol 41:1577–1585 McKay JA, Loane JF, Ross VG et al, 2002, c-erbB-2 is not a major factor in the development of colorectal cancer. Br JCancer 86:568–573 Maurer CA, Friess H, Kretschmann B et al, 1998, Increased expression of erbB3 in colorectal cancer is associated with concomitant increase in the level of erbB2. Hum Pathol 29:771–777 Seidal T, Balaton AJ, Battifora H, 2001, Interpretation and quantification of immunostains. Am J Surg Pathol 25:1204–1207 Prat A, Parera M, Reyes V et al, 2008, Successful treatment of pulmonary metastatic salivary ductal carcinoma with trastuzumabbased therapy. Head Neck 30:680–683 Cappuzzo F, Bemis L, Varella-Garcia M, 2006, HER2 mutation and response to trastuzumab therapy in non-small-cell lung cancer. N Engl J Med 354:2619–2621 Santin AD, Bellone S, Roman JJ, McKenney JK, Pecorelli S, 2008, Trastuzumab treatment in patients with advanced or recurrent endometrial carcinoma overexpressing HER2/neu. Int J Gynaecol Obstet 102:128–131 Karam A, Berek JS, Stenson A, Rao J, Dorigo O, 2008, HER-2/neu targeting for recurrent vulvar Paget’s disease A case report and literature review. Gynecol Oncol 111:568–571 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1183 EP 1189 DI 10.1007/s12253-015-9953-4 PG 7 ER PT J AU San-Miguel, T Pinto, S Navarro, L Callaghan, CR Monteagudo, C Lopez-Gines, C Cerda-Nicolas, M Gil-Benso, R AF San-Miguel, Teresa Pinto, Sandra Navarro, Lara Callaghan, C Robert Monteagudo, Carlos Lopez-Gines, Concha Cerda-Nicolas, Miguel Gil-Benso, Rosario TI Expression of the Chemokine Receptors CXCR3, CXCR4, CXCR7 and Their Ligands in Rhabdomyosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Alveolar Rhabdomyosarcoma; Chemokines; Embryonal Rhabdomyosarcoma; Flow cytometry ID Alveolar Rhabdomyosarcoma; Chemokines; Embryonal Rhabdomyosarcoma; Flow cytometry AB Rhabdomyosarcomas (RMS) are soft tissue malignant tumors of childhood and adolescents. The mechanisms underlying their aggressiveness are still poorly understood. Chemokines are chemotactic proteins involved in pathological processes that have been intensely studied in several types of cancers because of their influence in migration, angiogenesis, or metastases.We analyzed the expression of the chemokine receptors CXCR3, CXCR4 and CXCR7 and their ligands CXCL9, CXCL10, CXCL11 and CXCL12, in 15 RMS samples derived from nine patients. Expression was measured in tumors and primary cultures of RMS by Real-Time Polymerase Chain Reaction, immunostaining and flow cytometry. Our results show that these receptors are widely expressed in RMS. A significant difference between CXCL12/CXCR4, CXCL12/CXCR7, CXCL11/CXCR7 expression ratios was found in alveolar versus embryonal RMS and similarly between CXCL12/CXCR4 and CXCL11/CXCR3 ratios in primary versus recurrent tumors. These findings suggest a possible association between the interrelation of chemokine/ chemokine-receptor and an aggressive biological behavior in RMS. C1 [San-Miguel, Teresa] University of Valencia, Department of Pathology, Blasco Ibanez, 15, 46010 Valencia, Spain. [Pinto, Sandra] University of Coimbra, Center for Neuroscience and Cell Biology – CNC.IBILI, Blasco Ibanez,17, 3004-517 Coimbra, Portugal. [Navarro, Lara] University of Valencia, Department of Pathology, Blasco Ibanez, 15, 46010 Valencia, Spain. [Callaghan, C Robert] University of Valencia, Department of Pathology, Blasco Ibanez, 15, 46010 Valencia, Spain. [Monteagudo, Carlos] University of Valencia, Department of Pathology, Blasco Ibanez, 15, 46010 Valencia, Spain. [Lopez-Gines, Concha] University of Valencia, Department of Pathology, Blasco Ibanez, 15, 46010 Valencia, Spain. [Cerda-Nicolas, Miguel] University of Valencia, Department of Pathology, Blasco Ibanez, 15, 46010 Valencia, Spain. [Gil-Benso, Rosario] University of Valencia, Department of Pathology, Blasco Ibanez, 15, 46010 Valencia, Spain. RP San-Miguel, T (reprint author), University of Valencia, Department of Pathology, 46010 Valencia, Spain. EM teconsan@uv.es CR Barr FG, 2009, Sot tissue tumores: alverolar rhabdomyosarcoma: Atlas Genet Cytogenet Oncol Haematol Meyer WH, Spunt SL, 2004, Soft tissue sarcomas of childhood. Cancer Treat Rev 30:269–280 Xia SJ, Pressey JG, Barr FG, 2002, Molecular pathogenesis of rhabdomyosarcoma. Cancer Biol Ther 1:97–104 Barber TD, Barber MC, Tomescu O, Barr FG, Ruben S, Friedman TB, 2002, Identification of target genes regulated by PAX3 and PAX3-FKHR in embryogenesis and alveolar rhabdomyosarcoma. Genomics 79:278–284 Bridge JA, Liu J, Qualman SJ, Suijkerbuijk R,Wenger G, Zhang J, Wan X, Baker KS, Sorensen P, Barr FG, 2002, Genomic gains and losses are similar in genetic and histologic subsets of rhabdomyosarcoma, whereas amplification predominates in embryonal with anaplasia and alveolar subtypes. Genes Chromosomes Cancer 33: 310–321 Gil-Benso R, Lopez-Gines C, Carda C, Lopez-Guerrero JA, Ferrer J, Pellin-Perez A, Llombart-Bosch A, 2003, Cytogenetic and molecular findings related to rhabdomyosarcoma.An analysis of seven cases. Cancer Genet Cytogenet 144:125–133 Gil-Benso R, San-Miguel T, Callaghan RC, Bataller-Calatayud A, Caballero J, Pellin-Carcelen A, Donat J, Navarro S, Peris T, Cerda- Nicolas M, Lopez-Gines C, 2013, Chromosomal and genetic changes produced in tumoral progression of embryonal rhabdomyosarcoma. Histopathology 62:816–819 Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegui E, Zlotnik A, 2001, Involvement of chemokine receptors in breast cancer metastasis. Nature 410:50–56 Pinto S, Martinez-Romero A, O’Connor JE, Gil-Benso R, San- Miguel T, Terradez L, Monteagudo C, Callaghan RC, 2014, Intracellular coexpression of CXC- and CC- chemokine receptors and their ligands in human melanoma cell lines and dynamic variations after xenotransplantation. BMC Cancer 14:118 Gasparinho MG, Morgado S, Fonseca R, Chaves P, 2012, Collision metastases of breast and rectal carcinoma–a possible role for chemokines receptors expression. Pathol Oncol Res 18:729– 732 Rubin JB, Kung AL, Klein RS, Chan JA, Sun Y, Schmidt K, Kieran MW, Luster AD, Segal RA, 2003, A small-molecule antagonist of CXCR4 inhibits intracranial growth of primary brain tumors. Proc Natl Acad Sci U S A 100:13513–13518 Kucia M, Jankowski K, Reca R, Wysoczynski M, Bandura L, Allendorf DJ, Zhang J, Ratajczak J, Ratajczak MZ, 2004, CXCR4-SDF-1 signalling, locomotion, chemotaxis and adhesion. J Mol Histol 35:233–245 Burger JA, Kipps TJ, 2006, CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood 107:1761– 1767 Kakinuma T, Hwang ST, 2006, Chemokines, chemokine receptors, and cancer metastasis. J Leukoc Biol 79:639–651 Vandercappellen J, Van Damme J, Struyf S, 2008, The role of CXC chemokines and their receptors in cancer. Cancer Lett 267:226–244 Balabanian K, Lagane B, Infantino S, Chow KY, Harriague J, Moepps B, Arenzana-Seisdedos F, Thelen M, Bachelerie F, 2005, The chemokine SDF-1/CXCL12 binds to and signals through the orphan receptor RDC1 in T lymphocytes. J Biol Chem 280:35760– 35766 Burns JM, Summers BC,WangY,Melikian A, Berahovich R, Miao Z, Penfold ME, Sunshine MJ, Littman DR, Kuo CJ, Wei K, McMaster BE, Wright K, Howard MC, Schall TJ, 2006, A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. J Exp Med 203:2201–2213 Miao Z, Luker KE, Summers BC, Berahovich R, Bhojani MS, Rehemtulla A, Kleer CG, Essner JJ, Nasevicius A, Luker GD, Howard MC, Schall TJ, 2007, CXCR7, RDC1, promotes breast and lung tumor growth in vivo and is expressed on tumorassociated vasculature. Proc Natl Acad Sci U S A 104:15735– 15740 Juarez J, Bradstock KF, Gottlieb DJ, Bendall LJ, 2003, Effects of inhibitors of the chemokine receptor CXCR4 on acute lymphoblastic leukemia cells in vitro. Leukemia 17:1294–1300 Marchesi F, Monti P, Leone BE, Zerbi A, Vecchi A, Piemonti L, Mantovani A, Allavena P, 2004, Increased survival, proliferation, and migration in metastatic human pancreatic tumor cells expressing functional CXCR4. Cancer Res 64:8420–8427 Miekus K, Jarocha D, Trzyna E, Majka M, 2010, Role of I-TACbinding receptors CXCR3 and CXCR7 in proliferation, activation of intracellular signaling pathways and migration of various tumor cell lines. Folia Histochem Cytobiol 48:104–111 Sauty A, Dziejman M, Taha RA, Iarossi AS, Neote K, Garcia- Zepeda EA, Hamid Q, Luster AD, 1999, The T cell-specific CXC chemokines IP-10, Mig, and I-TAC are expressed by activated human bronchial epithelial cells. J Immunol 162:3549–3558 Struyf S, Burdick MD, Peeters E, Van den Broeck K, Dillen C, Proost P, Van Damme J, StrieterRM(2007, Platelet factor-4 variant chemokine CXCL4L1 inhibits melanoma and lung carcinoma growth and metastasis by preventing angiogenesis. Cancer Res 67:5940–5948 Libura J, Drukala J, Majka M, Tomescu O, Navenot JM, Kucia M, Marquez L, Peiper SC, Barr FG, Janowska-Wieczorek A, Ratajczak MZ, 2002, CXCR4-SDF-1 signaling is active in rhabdomyosarcoma cells and regulates locomotion, chemotaxis, and adhesion. Blood 100:2597–2606 Jankowski K, Kucia M, Wysoczynski M, Reca R, Zhao D, Trzyna E, Trent J, Peiper S, Zembala M, Ratajczak J, Houghton P, Janowska-Wieczorek A, Ratajczak MZ, 2003, Both hepatocyte growth factor, HGF, and stromal-derived factor-1 regulate the metastatic behavior of human rhabdomyosarcoma cells, but only HGF enhances their resistance to radiochemotherapy. Cancer Res 63: 7926–7935 Tomescu O, Xia SJ, Strezlecki D, Bennicelli JL, Ginsberg J, Pawel B, Barr FG, 2004, Inducible short-term and stable long-term cell culture systems reveal that the PAX3-FKHR fusion oncoprotein regulates CXCR4, PAX3, and PAX7 expression. Lab Investig 84: 1060–1070 Strahm B, Durbin AD, Sexsmith E, Malkin D, 2008, The CXCR4- SDF1alpha axis is a critical mediator of rhabdomyosarcoma metastatic signaling induced by bone marrow stroma. Clin Exp Metastasis 25:1–10 Diomedi-Camassei F, McDowell HP, De Ioris MA, Uccini S, Altavista P, Raschella G, Vitali R, Mannarino O, De Sio L, Cozzi DA, Donfrancesco A, Inserra A, Callea F, Dominici C, 2008, Clinical significance of CXC chemokine receptor-4 and c-Met in childhood rhabdomyosarcoma. Clin Cancer Res 14:4119–4127 Tarnowski M, Grymula K, Liu R, Tarnowska J, Drukala J, Ratajczak J, Mitchell RA, Ratajczak MZ, Kucia M, 2010, Macrophage migration inhibitory factor is secreted by rhabdomyosarcoma cells, modulates tumor metastasis by binding to CXCR4 and CXCR7 receptors and inhibits recruitment of cancer-associated fibroblasts. Mol Cancer Res 8:1328–1343 Grymula K, Tarnowski M, Wysoczynski M, Drukala J, Barr FG, Ratajczak J, Kucia M, Ratajczak MZ, 2010, Overlapping and distinct role of CXCR7-SDF-1/ITAC and CXCR4-SDF-1 axes in regulating metastatic behavior of human rhabdomyosarcomas. Int J Cancer 127:2554–2568 Monteagudo C, Ramos D, Pellin-Carcelen A, Gil R, Callaghan RC, Martin JM, Alonso V, Murgui A, Navarro L, Calabuig S, Lopez- Guerrero JA, Jorda E, Pellin A, 2012, CCL27-CCR10 and CXCL12-CXCR4 chemokine ligand-receptor mRNA expression ratio: new predictive factors of tumor progression in cutaneous malignant melanoma. Clin Exp Metastasis 29:625–637 Gil-Benso R, Monteagudo C, Cerda-Nicolas M, Callaghan RC, Pinto S, Martinez-Romero A, Pellin-Carcelen A, San-Miguel T, Cigudosa JC, Lopez-Gines C, 2012, Characterization of a new human melanoma cell line with CD133 expression. Hum Cell 25: 61–67 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T), method. Methods 25:402–408 CristW, Gehan EA, Ragab AH, Dickman PS, Donaldson SS, Fryer C, Hammond D, Hays DM, Herrmann J, Heyn R et al, 1995, The third intergroup Rhabdomyosarcoma study. J Clin Oncol 13:610– 630 ParhamDM, EllisonDA(2006, Rhabdomyosarcomas in adults and children: an update. Arch Pathol Lab Med 130:1454–1465 Tarnowski M, Grymula K, Reca R, Jankowski K, Maksym R, Tarnowska J, Przybylski G, Barr FG, Kucia M, Ratajczak MZ, 2010, Regulation of expression of stromal-derived factor-1 receptors: CXCR4 and CXCR7 in human rhabdomyosarcomas. Mol Cancer Res 8:1–14 Wysoczynski M,Miekus K, Jankowski K,Wanzeck J, Bertolone S, Janowska-Wieczorek A, Ratajczak J, Ratajczak MZ, 2007, Leukemia inhibitory factor: a newly identified metastatic factor in rhabdomyosarcomas. Cancer Res 67:2131–2140 Masuda T, Nakashima Y, Ando K, Yoshinaga K, Saeki H, Oki E, Morita M, Oda Y, Maehara Y, 2014, Nuclear expression of chemokine receptor CXCR4 indicates poorer prognosis in gastric cancer. Anticancer Res 34:6397–6403 Yao X, Zhou L, Han S, Chen Y, 2011, High expression of CXCR4 and CXCR7 predicts poor survival in gallbladder cancer. J Int Med Res 39:1253–1264 Tamas K, Domanska UM, van Dijk TH, Timmer-Bosscha H, Havenga K, Karrenbeld A, Sluiter WJ, Beukema JC, van Vugt MA, de Vries EG, Hospers GA, Walenkamp AM, 2015, Expression in rectal tumors of stage IV patients before and after local radiotherapy and systemic neoadjuvant treatment. Curr Pharm Des Bar M, Bar D, Lehmann B, 2009, Selection and validation of candidate housekeeping genes for studies of human keratinocytes–review and recommendations. J Invest Dermatol 129:535–537 Speetjens FM, Liefers GJ, Korbee CJ, Mesker WE, van de Velde CJ, van Vlierberghe RL, Morreau H, Tollenaar RA, Kuppen PJ, 2009, Nuclear localization of CXCR4 determines prognosis for colorectal cancer patients. Cancer Microenviron 2:1–7 Wang SC, Lin JK, Wang HS, Yang SH, Li AF, Chang SC, 2010, Nuclear expression of CXCR4 is associated with advanced colorectal cancer. Int J Colorectal Dis 25:1185–1191 Wang L, Wang Z, Yang B, Yang Q, Sun Y, 2009, CXCR4 nuclear localization follows binding of its ligand SDF-1 and occurs in metastatic but not primary renal cell carcinoma. Oncol Rep 22:1333– 1339 Na IK, Scheibenbogen C, Adam C, Stroux A, Ghadjar P, Thiel E, Keilholz U, Coupland SE, 2008, Nuclear expression of CXCR4 in tumor cells of non-small cell lung cancer is correlated with lymph node metastasis. Hum Pathol 39:1751–1755 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1191 EP 1199 DI 10.1007/s12253-015-9947-2 PG 9 ER PT J AU Chen, YT Tsao, ShCh Yuan, FShSh Tsai, HP Chai, ChY AF Chen, Yi-Ting Tsao, Shu-Chuan Yuan, F Shyng-Shiou Tsai, Hung-Pei Chai, Chee-Yin TI Serine Protease Inhibitor Kazal Type 1 (SPINK1) Promotes Proliferation of Colorectal Cancer Through the Epidermal Growth Factor as a Prognostic Marker SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Epidermal growth factor receptor (EGFR); Serine protease inhibitor Kazal type-1 (SPINK1); Tumor-associated trypsin inhibitor (TATI) ID Colorectal cancer; Epidermal growth factor receptor (EGFR); Serine protease inhibitor Kazal type-1 (SPINK1); Tumor-associated trypsin inhibitor (TATI) AB Serine protease inhibitor Kazal type-1 (SPINK1), a trypsin kinase inhibitor, is involved in inflammation, cell proliferation and carcinogenesis. The role and association between SPINK1, EGFR and Ki-67 in colorectal adenoma (CRA) and colorectal cancer (CRC) are still unknown. In this study, we used immunohistochemical stain to evaluate expression of SPINK1, EGFR and Ki-67 proteins in 30 CRA and 53 CRC patients semiquantitatively, and then analyzed their correlation with clinicopathologic parameters. Our results revealed that SPINK1 expression was noted in the upper and basal parts of the crypts in CRA and was more intensely related with cellular atypia. EGFR expression was found in 13 out of 30 adenomas, including 9 out of 15 adenomas with dysplasia or synchronous CRC (60 %), and 4 out of 15 adenomas without dysplasia (26.7 %). In CRC, high SPINK1 expression was significantly associated with males (p= 0.041) and advanced disease stage (p=0.015). EGFR positivity was significantly correlated with higher T stage (p=0.004) and disease stage (stage I-IV, p=0.017; early vs. late, p=0.015). Pearson’s correlation showed positive correlation between the SPINK1 intensity and EGFR immunoreactivity (p=0.011), and Ki-67 and SPINK1 intensity or percentage (p=0.017 and p=0.039 respectively). In Kaplan-Meier analyses, patients with high SPINK1 intensity tended to have shorter overall survival (p=0.03). Concomitant expression of high SPINK1 intensity and EGFR was also identified as being associated with poor prognosis (p=0.015). In conclusion, high SPINK1 expression is associated with advanced stage and poor prognosis. There is positive correlation between high SPINK1 expression, EGFR immunoreactivity, and high Ki- 67 labeling index. The SPINK1 protein seems to play a role in tumor proliferation and malignant transformation through the EGFR pathway. SPINK1 may serve as a prognostic biomarker in therapeutic targeting in the future. C1 [Chen, Yi-Ting] Kaohsiung Medical University, College of Medicine, Department of Pathology, No. 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. [Tsao, Shu-Chuan] Kaohsiung Medical University, College of Medicine, Department of Pathology, No. 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. [Yuan, F Shyng-Shiou] Kaohsiung Medical University, Cancer Center, Department of Medical Research, Translational Research CenterKaohsiung, Taiwan, Republic of China. [Tsai, Hung-Pei] Kaohsiung Medical University, College of Medicine, Graduate Institute of Clinical MedicineKaohsiung, Taiwan, Republic of China. [Chai, Chee-Yin] Kaohsiung Medical University, College of Medicine, Department of Pathology, No. 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. RP Chai, ChY (reprint author), Kaohsiung Medical University, College of Medicine, Department of Pathology, 807 Kaohsiung, Taiwan, Republic of China. EM cychai@kmu.edu.tw CR Ferlay J, Soerjomataram I, Ervik M, 2012, GLOBOCAN: Estimated cancer incidence, mortality and prevalence worldwide in 2012. International Agency for Research on Cancer Yamaji Y,Mitsushima T, Yoshida H,Watabe H, OkamotoM,Wada R, Ikuma H, Kawabe T, Omata M, 2006, The malignant potential of freshly developed colorectal polyps according to age. Cancer Epidemiol Biomarkers Prev 15(12):2418–21 Kazal LA, Spicer DS, Brahinsky RA, 1948, Isolation of a crystalline trypsin inhibitor-anticoagulant protein from pancreas. J Am Chem Soc 70(9):3034–40 Masamune A, Mizutamari H, Kume K, Asakura T, Satoh K, Shimosegawa T, 2004, Hereditary pancreatitis as the premalignant disease: a Japanese case of pancreatic cancer involving the SPINK1 gene mutation N34S. Pancreas 28(3):305–10 Stenman UH, 2011, SPINK1: a new therapeutic target in cancer? Clin Chem 57(11):1474–5 Ateeq B, Tomlins SA, Laxman B, Asangani IA, Cao Q, Cao X, Li Y, Wang X, Feng FY, Pienta KJ, Varambally S, Chinnaiyan AM, 2011, Therapeutic targeting of SPINK1-positive prostate cancer. Sci Transl Med 3(72):72ra17 Lenz HJ, 2006, Anti-EGFR mechanism of action: antitumor effect and underlying cause of adverse events. Oncology, Williston Park, 20(5 Suppl 2):5–13 Nicholson RI, Gee JM, Harper ME, 2001, EGFR and cancer prognosis. Eur J Cancer 37(Suppl 4):S9–15 Vokes EE, Chu E, 2006, Anti-EGFR therapies: clinical experience in colorectal, lung, and head and neck cancers. Oncology, Williston Park, 20(5 Suppl 2):15–25 Fukuoka S, Fushiki T, Kitagawa Y, Sugimoto E, Iwai K, 1987, Competition of a growth stimulating-/cholecystokinin, CCK, releasing-peptide, monitor peptide, with epidermal growth factor for binding to 3T3 fibroblasts. Biochem Biophys Res Commun 145(2):646–50 Ozaki N, Ohmuraya M, Hirota M, Ida S, Wang J, Takamori H, Higashiyama S, Baba H, Yamamura K, 2009, Serine protease inhibitor Kazal type 1 promotes proliferation of pancreatic cancer cells through the epidermal growth factor receptor. Mol Cancer Res 7(9):1572–81 Ozaki N, OhmurayaM, Ida S, Hashimoto D, Ikuta Y, Chikamoto A, Hirota M, Baba H, 2013, Serine protease inhibitor Kazal type 1 and epidermal growth factor receptor are expressed in pancreatic tubular adenocarcinoma, intraductal papillary mucinous neoplasm, and pancreatic intraepithelial neoplasia. J Hepatobiliary Pancreat Sci Scholzen T, Gerdes J, 2000, The Ki-67 protein: from the known and the unknown. J Cell Physiol 182(3):311–22 Edge SB, Byrd DR, Compton CC, 2010, American Joint Committee on Cancer, AJCC, Cancer Staging Manual, 7th edn. Springer, New York Rink M, Park K, Volkmer BG, Xylinas E, Hansen J, Cha EK, Robinson BD, Hautmann R, Kufer R, Engel O, Chun FK, Dahlem R, Rubin MA, Shariat SF, Mosquera JM, 2012, Loss of SPINK1 expression is associated with unfavorable outcomes in urothelial carcinoma of the bladder after radical cystectomy. Urol Oncol Koskensalo S, Hagstrom J, Louhimo J, Stenman UH, Haglund C, 2012, Tumour-associated trypsin inhibitor TATI is a prognostic marker in colorectal cancer. Oncology 82(4):234–41 Gaber A, Johansson M, Stenman UH, Hotakainen K, Ponten F, Glimelius B, Bjartell A, Jirstrom K, Birgisson H, 2009, High expression of tumour-associated trypsin inhibitor correlates with liver metastasis and poor prognosis in colorectal cancer. Br J Cancer 100(10):1540–8 Atkins D, Reiffen KA, Tegtmeier CL, Winther H, Bonato MS, Storkel S, 2004, Immunohistochemical detection of EGFR in paraffin-embedded tumor tissues: variation in staining intensity due to choice of fixative and storage time of tissue sections. J Histochem Cytochem 52(7):893–901 Allegra CJ, Paik S, Colangelo LH, Parr AL, Kirsch I, Kim G, Klein P, Johnston PG,Wolmark N,WieandHS, 2003, Prognostic value of thymidylate synthase, Ki-67, and p53 in patients with Dukes’ B and C colon cancer: a National Cancer Institute-National Surgical Adjuvant Breast and Bowel Project collaborative study. J Clin Oncol 21(2):241–50 Fluge O, Gravdal K, Carlsen E, Vonen B, Kjellevold K, Refsum S, Lilleng R, Eide TJ, Halvorsen TB, Tveit KM, Otte AP, Akslen LA, Dahl O, 2009, Expression of EZH2 and Ki-67 in colorectal cancer and associations with treatment response and prognosis. Br J Cancer 101(8):1282–9 Peng Y, Wang L, Gu J, 2013, Elevated preoperative carcinoembryonic antigen, CEA, and Ki67 is predictor of decreased survival in IIA stage colon cancer. World J Surg 37(1): 208–13 Ohmachi Y, Murata A, Matsuura N, Yasuda T, Uda K, Mori T, 1994, Overexpression of pancreatic secretory trypsin inhibitor in pancreatic cancer. Evaluation of its biological function as a growth factor. Int J Pancreatol 15(1):65–73 Paju A, Hotakainen K, Cao Y, Laurila T, Gadaleanu V, Hemminki A, Stenman UH, Bjartell A, 2007, Increased expression of tumorassociated trypsin inhibitor, TATI, in prostate cancer and in androgen-independent 22Rv1 cells. Eur Urol 52(6):1670–9 Bohe H, Bohe M, Lindstrom C, Ohlsson K, 1990, Immunohistochemical demonstration of pancreatic secretory trypsin inhibitor in normal and neoplastic colonic mucosa. J Clin Pathol 43(11):901–4 Higashiyama M, Monden T, Tomita N, Murotani M, Kawasaki Y, Morimoto H, Murata A, Shimano T, Ogawa M, Mori T, 1990, Expression of pancreatic secretory trypsin inhibitor, PSTI, in colorectal cancer. Br J Cancer 62(6):954–8 Gaber A, Nodin B, Hotakainen K, Nilsson E, Stenman UH, Bjartell A, Birgisson H, Jirstrom K, 2010, Increased serum levels of tumour-associated trypsin inhibitor independently predict a poor prognosis in colorectal cancer patients. BMC Cancer 10(498 Gouyer V, Fontaine D, Dumont P, de Wever O, Fontayne-Devaud H, Leteurtre E, Truant S, Delacour D, Drobecq H, Kerckaert JP, de Launoit Y, Bracke M, Gespach C, Desseyn JL, Huet G, 2008, Autocrine induction of invasion and metastasis by tumorassociated trypsin inhibitor in human colon cancer cells. Oncogene 27(29):4024–33 Marchbank T, Mahmood A, Playford RJ, 2013, Pancreatic secretory trypsin inhibitor causes autocrine-mediated migration and invasion in bladder cancer and phosphorylates the EGF receptor, Akt2 and Akt3, and ERK1 and ERK2. Am J Physiol Renal Physiol 305(3):F382–9 Gaber A, Stene C, Hotakainen K, Nodin B, Palmquist I, Bjartell A, Stenman UH, Jeppsson B, Johnson LB, Jirstrom K, 2011, Effects of radiation therapy on tissue and serum concentrations of tumour associated trypsin inhibitor and their prognostic significance in rectal cancer patients. Radiat Oncol 6(100 Rego RL, Foster NR, Smyrk TC, Le M, O’ConnellMJ, Sargent DJ, Windschitl H, Sinicrope FA, 2010, Prognostic effect of activated EGFR expression in human colon carcinomas: comparison with EGFR status. Br J Cancer 102(1):165–72 Zhang W, Gordon M, Lenz HJ, 2006, Novel approaches to treatment of advanced colorectal cancer with anti-EGFR monoclonal antibodies. Ann Med 38(8):545–51 Bansal A, Liu X, McGregor DH, Singh V, Hall S, 2010, Correlation of epidermal growth factor receptor with morphological features of colorectal advanced adenomas: a pilot correlative case series. Am J Med Sci 340(4):296–300 Rokita M, Stec R, Bodnar L, Charkiewicz R, Korniluk J, Smoter M, Cichowicz M, Chyczewski L, Niklinski J, Kozlowski W, Szczylik C, 2013, Overexpression of epidermal growth factor receptor as a prognostic factor in colorectal cancer on the basis of the Allred scoring system. Onco Targets Ther 6(967-76) Roberts RB, Min L, Washington MK, Olsen SJ, Settle SH, Coffey RJ, Threadgill DW, 2002, Importance of epidermal growth factor receptor signaling in establishment of adenomas and maintenance of carcinomas during intestinal tumorigenesis. Proc Natl Acad Sci U S A 99(3):1521–6 Ogawa M, Tsushima T, Ohba Y, Ogawa N, Tanaka S, Ishida M, Mori T, 1985, Stimulation of DNA synthesis in human fibroblasts by human pancreatic secretory trypsin inhibitor. Res Commun Chem Pathol Pharmacol 50(1):155–8 Koskensalo S, Louhimo J, Hagstrom J, Lundin M, Stenman UH, Haglund C, 2013, Concomitant tumor expression of EGFR and TATI/SPINK1 associates with better prognosis in colorectal cancer. PLoS One 8(10), e76906 Grondahl-Hansen J, Christensen IJ, Rosenquist C, Brunner N, Mouridsen HT, Dano K, Blichert-Toft M, 1993, High levels of urokinase-type plasminogen activator and its inhibitor PAI-1 in cytosolic extracts of breast carcinomas are associated with poor prognosis. Cancer Res 53(11): 2513–21 Schrohl AS, Holten-Andersen MN, Peters HA, Look MP, Meijervan Gelder ME, Klijn JG, Brunner N, Foekens JA, 2004, Tumor tissue levels of tissue inhibitor of metalloproteinase-1 as a prognostic marker in primary breast cancer. Clin Cancer Res 10(7):2289–98 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1201 EP 1208 DI 10.1007/s12253-015-9949-0 PG 8 ER PT J AU Ipekci, T Ozden, F Unal, B Saygin, C Uzunaslan, D Ates, E AF Ipekci, Tumay Ozden, Ferhat Unal, Betul Saygin, Caner Uzunaslan, Didem Ates, Erhan TI Epithelial-Mesenchymal Transition Markers β-catenin, Snail, and E-Cadherin do not Predict Disease Free Survival in Prostate Adenocarcinoma: a Prospective Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Epithelial-mesenchymal transition; β-catenin; Snail; E-cadherin; Survival ID Prostate cancer; Epithelial-mesenchymal transition; β-catenin; Snail; E-cadherin; Survival AB Current methods for diagnosis and staging of prostate adenocarcinoma are not sensitive enough to distinguish between patients with indolent disease and those that should receive radical treatment. Epithelial-mesencyhmal transition (EMT) is a well-characterized process involved in tumor invasion and metastasis. The aim of this study is to analyze the expression of β-catenin, Snail, and E-cadherin in prostate cancer patients with prospective evaluation of their value in predicting disease-free survival (DFS). One-hundred-andthree consecutive prostate carcinoma patients who underwent radical prostatectomy and 35 patients with benign prostate hyperplasia (BPH) were enrolled. Age, initial PSA level, tumor size and clinical stage were documented for adenocarcinoma patients and they were enrolled in active surveillance with serum PSA levels. Recurrence was defined as PSA level of ≥0.2 ng/ml on at least 2 occasions over a 2-month period. Immunohistochemical staining intensity was scored as negative, weakly positive, moderately positive, and strongly positive. For Snail and β-catenin immunoreaction, the tumors were considered nuclear positive when more than 5 % of the nuclei of tumor cells were positively stained. Patients with prostate cancer had weaker β-catenin (p<0.0001), Snail (p= 0.006), and E-cadherin (p=0.02) staining when compared to BPH patients and the frequency of nuclear positivity for β- catenin and Snail were higher in adenocarcinoma group (p< 0.0001). Increased expression and nuclear positivity of β- catenin were associated with advanced stage (p=0.012 and p=0.003) and higher tumor volume (p=0.013 and p=0.002). Additionally, patients with increased Snail expression had higher Gleason scores and tumor volume at presentation (p= 0.008 and p=0.004). However, there were no significant DFS differences in adenocarcinoma patients who did and did not have β-catenin, Snail, and E-cadherin expression as assessed with log-rank test. Expressions of β-catenin, Snail, and Ecadherin were significantly lower in prostate cancer patients compared to BPH patients and both β-catenin and Snail had nuclear staining pattern in patients with adenocarcinoma. However, none of these markers predicted DFS in 36-month follow up of our cohort. C1 [Ipekci, Tumay] Antalya Training and Research Hospital, Department of UrologyAntalya, Turkey. [Ozden, Ferhat] Van Regional Training and Research Hospital, Department of PathologyVan, Turkey. [Unal, Betul] Antalya Training and Research Hospital, Department of PathologyAntalya, Turkey. [Saygin, Caner] Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, Kocamustafapasa, Fatih, 0340090 Istanbul, Turkey. [Uzunaslan, Didem] Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, Kocamustafapasa, Fatih, 0340090 Istanbul, Turkey. [Ates, Erhan] Antalya Training and Research Hospital, Department of UrologyAntalya, Turkey. RP Saygin, C (reprint author), Cerrahpasa Medical School, Istanbul University, Department of Internal Medicine, 0340090 Istanbul, Turkey. EM csaygin@yahoo.com CR Siegel R, Miller KD, Jemal A, 2015, Cancer statistics, 2015. CA Cancer J Clin; In Press Marta GN, Hanna SA, da Silva JLF, 2013, Carvalho HdA. Screening for prostate cancer: an updated review. Expert Rev Anticancer Ther 13:101–108 Trantham LC, Nielsen ME, Mobley LR, Wheeler SB, Carpenter WR, Biddle AK, 2013, Use of prostate-specific antigen testing as a disease surveillance tool following radical prostatectomy. Cancer 119:3523–3530 NCCN. Clinical practice guidelines in oncology: prostate cancer. National comprehensive cancer network 2014; Version 1: Available from: NCCN.org. Detailed guidelines on the detection, prevention, risk stratification and treatment of patients Celia-Terrassa T, Meca-Cortes O, Mateo F, de Paz AM, Rubio N, Arnal-Estape A, Ell BJ, Bermudo R, Diaz A, Guerra-Rebollo M, Lozano JJ, Estaras C, Ulloa C, Alvarez-Simon D,Mila J, Vilella R, Paciucci R, Martinez-BalbasM, de Herreros AG, Gomis RR, Kang Y, Blanco J, Fernandez PL, Thomson TM, 2012, Epithelialmesenchymal transition can suppress major attributes of human epithelial tumor-initiating cells. J Clin Invest 122(5):1849–1868 Cano A, Perez-MorenoMA, Rodrigo I, Locascio A, Blanco MJ, del Barrio MG et al, 2000, The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol 2:76–83 Ohkubo T, Ozawa M, 2004, The transcription factor snail downregulates the tight junction components independently of Ecadherin downregulation. J Cell Sci 117:1675–1685 Emadi Baygi M, Soheili ZS, Schmitz I, Sameie S, Schulz WA, 2010, Snail regulates cell survival and inhibits cellular senescence in human metastatic prostate cancer cell lines. Cell Biol Toxicol 26: 553–567 Smith BN, Odero-Marah VA, 2012, The role of snail in prostate cancer. Cell Adh Migr 6(5):433–441 Davies G, Jiang WG, Mason MD, 2000, Cell-cell adhesion molecules and signaling intermediates and their role in the invasive potential of prostate cancer cells. J Urol 163:985–992 Nowicki A, Sporny S, Duda-Szymanska J, 2012, β-catenin as a prognostic factor for prostate cancer, PCa). Cent Eur J Urol 65(3): 119–123 Poblete CE, Fulla J, Gallardo M,Munoz V, Castellon EA, Gallegos I, Contreras HR, 2014, Increased SNAIL expression and low syndecan levels are associated with high Gleason grade in prostate cancer. Int J Oncol 44(3):647–654 Heeboll S, BorreM, Ottosen PD, Dyrskjot L, Orntoft TF, Torring N, 2009, Snail1 is over-expressed in prostate cancer. APMIS 117(3): 196–204 Whiteland H, Spencer-Harty S, Thomas DH, Davies C, Morgan C, Kynaston H, Bose P, Fenn N, Lewis PD, Bodger O, Jenkins S, Doak SH, 2013, Putative prognostic epithelial-to mesenchymal transition biomarkers for aggressive prostate cancer. Exp Mol Pathol 95(2):220–226 Wen YC, ChenWY, Lee WJ, Yang SF, Lee LM, Chien MH, 2014, Snail as a potential marker for predicting the recurrence of prostate cancer in patients at stage T2 after radical prostatectomy. Clin Chim Acta 431:169–173 Freedland SJ, SutterME, Dorey F, Aronson WJ, 2003, Defining the ideal cutpoint for determining PSA recurrence after radical prostatectomy. Prostate-specific antigen. Urology 61:365–369 Kypta RM, Waxman J, 2012, Wnt/beta-catenin signalling in prostate cancer. Nat Rev Urol 9:418–428 Song LN, Gelmann EP, 2005, Interaction of beta-catenin and TIF2/ GRIP1 in transcriptional activation by the androgen receptor. J Biol Chem 280(45):37853–37867 Yang X, Chen MW, Terry S, Vacherot F, Bemis DL, Capodice J, Kitajewski J, de la Taille A, Benson MC, Guo Y, Buttyan R, 2006, Complex regulation of human androgen receptor expression by Wnt signaling in prostate cancer cells. Oncogene 25(24):3436– 3444 Lee E, Madar A, David G, Garabedian MJ, Dasgupta R, Logan SK, 2013, Inhibition of androgen receptor and β-catenin activity in prostate cancer. Proc Natl Acad Sci U S A 110(39):15710–15715 Jung SJ, Oh S, Lee GT, Chung J, Min K, Yoon J, KimW, Ryu DS, Kim IY, Kang DI, 2013, Clinical significance of Wnt/β-catenin signalling and androgen receptor expression in prostate cancer. World J Mens Health 31(1):36–46 Micalizzi DS, Farabaugh SM, Ford HL, 2010, Epithelialmesenchymal transition in cancer: parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia 15:117–134 Talbot L, Bhattacharya S, Kuo P, 2012, Epithelial-mesenchymal transition, the tumor microenvironment, and metastatic behavior of epithelial malignancies. Int J Biochem Mol Biol 3:117–136 Onder TT, Gupta PB, Mani SA, Yang J, Lander ES, Weinberg RA, 2008, Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res 68:3645–3654 Lazari P, Poulias H, Gakiopoulou H, Thomopoulou GH, Barbatis C, Lazaris AC, 2013, Differential immunohistochemical expression of CD44s, E-cadherin and β-catenin among hyperplastic and neoplastic lesions of the prostate gland. Urol Int 90(1):109–116 Pontes J Jr, Srougi M, Borra PM, Dall’ Oglio MF, Ribeiro-Filho LA, Leite KR, 2010, E-cadherin and beta-catenin loss of expression related to bone metastasis in prostate cancer. Appl Immunohistochem Mol Morphol 18(2):179–184 Uchikado Y, Okumura H, Ishigami S, Setoyama T, Matsumoto M, Owaki T, Kita Y, Natsugoe S, 2011, Increased slug and decreased E-cadherin expression is related to poor prognosis in patients with gastric cancer. Gastric Cancer 14(1):41–49 Meng Z, Cao R, Yang Z, Liu T, Wang Y, Wang X, 2013, Inhibitor of 5-lipoxygenase, zileuton, suppresses prostate cancer metastasis by upregulating E-cadherin and paxillin. Urology 82(6):1452–e7- 14 Krawczyk N, Meier-Stiegen F, Banys M, Neubauer H, Ruckhaeberle E, Fehm T, 2014, Expression of stem cell and epithelial-mesenchymal transition markers in circulating tumor cells of breast cancer patients. Biomed Res Int 2014:415721 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1209 EP 1216 DI 10.1007/s12253-015-9958-z PG 8 ER PT J AU Bittner, N Baliko, Z Sarosi, V Laszlo, T Toth, E Kasler, M Geczi, L AF Bittner, Nora Baliko, Zoltan Sarosi, Veronika Laszlo, Terezia Toth, Erika Kasler, Miklos Geczi, Lajos TI Bone Metastases and the EGFR and KRAS Mutation Status in Lung Adenocarcinoma - The Results of Three Year Retrospective Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung adenocarcinoma; Skeletal related events; Tyrosine kinase inhibitors; EGFR mutation; KRAS mutation ID Lung adenocarcinoma; Skeletal related events; Tyrosine kinase inhibitors; EGFR mutation; KRAS mutation AB Lung cancer is a heterogeneous group of disease and mutational profiling of lung adenocarcinomas is a routine practice in thoracic oncology. Kirsten-RAS (KRAS) and EGFR mutations play an important role in the carcinogenesis of a subset of lung adenocarcinomas. Our aim was to investigate the correlation between bone metastases and EGFR and KRAS mutation status in lung adenocarcinoma patients. Retrospectively we analysed 224 patients with recurrent or metastatic lung adenocarcinomas. Patients were treated with standard chemotherapy as first line therapy and with EGFRTK inhibitors as a second or third line therapy. 72 of 224 patients (32 %) had verified bone metastases. Bone metastases and Skeletal Related Events (SRE) were more frequent in men, heavy smokers and without treatment of EGFR TK inhibitors. We have found that EGFR and KRAS mutation status are both predictive factors for the treatment efficacy and prognostic factors for the disease progression. However there were no significant correlation between mutation status and the presence of bone metastases (P=0, 59). In our study the presence of bone metastases proved to be an independent prognostic factor related to poor performance status and worse Quality of Life (QL). C1 [Bittner, Nora] National Institute of OncologyBudapest, Hungary. [Baliko, Zoltan] University of Pecs, Faculty of Medicine, Department of PulmonologyPecs, Hungary. [Sarosi, Veronika] University of Pecs, Faculty of Medicine, Department of PulmonologyPecs, Hungary. [Laszlo, Terezia] University of Pecs, Faculty of Medicine, Department of PulmonologyPecs, Hungary. [Toth, Erika] National Institute of OncologyBudapest, Hungary. [Kasler, Miklos] National Institute of OncologyBudapest, Hungary. [Geczi, Lajos] National Institute of OncologyBudapest, Hungary. RP Bittner, N (reprint author), National Institute of Oncology, Budapest, Hungary. EM nora_bittner@yahoo.ca CR Jemal A, Siegel R, Ward E, Hao Y, 2009, Cancer statistic, 2009. CA Cancer J Clin 59:225–249 Travis WD, Brambilla E, Noguchi M, Nicholson AG, Geisinger KR, 2011, International Association for the Study of Lung Cancer /American Thoracic Society / European Respiratory Society International multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol 6:2244–2285 Oxnard GR, Binder A, Janne PA, 2013, New targetable oncogenes in non-small-cell lung cancer. J Clin Oncol 31:1097–1104 Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, 2002, Comparison of four chemotherapy regimens for advanced nonsmall- cell lung cancer. N Engl J Med 346:92–98 NSCLC Meta-Analyses Collaborative Group, 2008, Chemotherapy in addition to supportive care improves survival in advanced non-small-cell lung cancer: a systematic review and meta-analysis of individual patient data from 16 randomized controlled trials. J Clin Oncol 26:4617–4625 Azolli CG, Baker S Jr, Temin S, Pao W, Aliff T, Brahmer J, 2009, American Society of Clinical Oncology Clinical Practice Guideline update on chemotherapy for stage IV non-small-cell lung cancer. J Clin Oncol 36:6251–6266 Paz-Ares L, 2012, PARAMOUNT final overall survival, OS, result of the phase III study of maintenance pemetrexed, pem, plus best supportive care, BSC, versus placebo, plb, plus BSC immediatly following induction treatment with pem plus cisplatin, cis, for advanced nonsquamosus, NS, non-small cell lung cancer, NSCLC). J Clin Oncol 30:(Suppl, abstr LBA 7507) Gazdar AF, 2009, Activating and resistance mutations of EGFR in nonsmall- cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene 28(Suppl 1):S24– S31 Fukuoka M, Wu Y, 2011, Biomarker Analyses and Final Overall Survival results From a Phase III, randomised, open an label, Firstline Study of Gefitinib versus carboplatin/Paclitaxel in Clinically Selected Patients with Advanced Non-Small-Cell Lung cancer in Asia, IPASS). J Clin Oncol 29:2866–2874 Bittner N, Ostoros G, Geczi L, 2014, New treatment options for lung adenocarcinoma- in view of molecular background. Pathol Oncol Res 20:11–25 Cappuzzo F, Ciuleanu T, Stelmakh L, Cicenas S, 2010, Erlotinib as maintenance treatment in advanced non-small-cell lung cancer: a multicentre, randomised, placebo-controlled phase 3 study. Lancet Oncol 11:521–529 Alice Tsang S, Mehra R, 2013, Clinical activity of the ALK inhibitor LDK378 in advanced, ALK-positive NSCLC. J Clin Oncol 31 Delea TE, McKiernan J, Brandman J, Edelsberg J, Sung J, 2006, Impact of skeletal complications on total medical care costs among patients with bone metastases of lung cancer. J Thorac Oncol 1: 571–576 Sun JM, Ahn JS, Lee S, Kim JA, Lee J, 2011, Predictors of skeletalrelated events in non-small cell lung cancer patients with bone metastases. Lung Cancer 71:89–93 Reinersman JM, Johnson ML, Riely GJ, Chitale DA, Nicastri AD, 2011, Frequency of EGFR and KRAS mutations in lung adenocarcinomas in African Americans. J Thorac Oncol 6:28–31 Roberts PJ, Stinchcombe TE, 2013, KRAS mutation: should we test for it, and does it matter? J Clin Oncol 31:1112–1121 Cserepes M, Ostoros G, Lohinai Z, Raso E, Barbai T, 2014, Subtype-specific KRAS mutations in advanced lung adenocarcinoma: a retrospective study of patients treated with platinum-based chemotherapy. Eur J Cancer 50:1819–1828 Normanno N, Luca D, Aldinucci D, Maiello MR, Mancino M, 2005, Gefitinib inhibits the ability of human bone marrow stromal cells to induce osteoclast differentiation: implications for the pathogenesis and treatment of bonemetastasis. Endocr Relat Cancer 12: 471–482 Lu X, Wang Q, Hu G, Van Poznak C, Fleisher M, 2009, ADAM TS1 and MMP1 proteolytically engage EGF-like ligands in an osteolytic signaling cascade for bone metastasis. Genes Dev 23: 1882–1894 Nagata M, Kudoh S, Mitsuoka S, Suzumura T, Umekawa K, 2013, Skeletal-related events in advanced lung adenocarcinoma patients evaluated EGFR mutations. Osaka City Med J 59:45–52 Bae HM, Lee SH, Kim TM, Kim DW, Yang SC, 2012, Prognostic factor for non-small cell lung cancer with bone metastases at the time of diagnosis. Letters to the editor. Lung Cancer 78:167–170 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1217 EP 1221 DI 10.1007/s12253-015-9955-2 PG 5 ER PT J AU Pala, EE Bayol, U Keskin, UE Ozguzer, A Kucuk, U Ozer, O Koc, A AF Pala, Ebru Emel Bayol, Umit Keskin, Usturali Elif Ozguzer, Alp Kucuk, Ulku Ozer, Ozge Koc, Altug TI Determination of HER2 and p53 Mutations by Sequence Analysis Method and EGFR/Chromosome 7 Gene Status by Fluorescence in Situ Hybridization for the Predilection of Targeted Therapy Modalities in Immunohistochemically Triple Negative Breast Carcinomas in Turkish Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Triple negative breast cancer; EGFR; p53; HER2 ID Triple negative breast cancer; EGFR; p53; HER2 AB Triple negative breast cancer (TNBC), an agressive subtype accounts nearly 15 % of all breast carcinomas. Conventional chemotherapy is the only treatment modality thus new, effective targeted therapy methods have been investigated. Epidermal growth factor receptor (EGFR) inhibitors give hope according to the recent studies results. Also therapeutic agents have been tried against aberrant p53 signal activity as TNBC show high p53 mutation rates. Our aim was to detect the incidence of mutations/amplifications identified in TNBC in our population. Here we used sequence analysis to detect HER2 (exon 18–23), p53 (exon 5–8) mutations; fluorescence in situ hybridization (FISH) method to analyse EGFR/chromosome 7 centromere gene status in 82 immunohistochemically TNBC. Basaloid phenotype was identified in 49 (59.8 %) patients. EGFR amplification was noted in 5 cases (6.1 %). All EGFR amplified cases showed EGFR overexpression by immunohistochemistry (IHC). p53 mutations were identified in 33 (40.2 %) cases. Almost 60 % of the basal like breast cancer cases showed p53 mutation. Only one case showed HER2 mutation (exon 20:g.36830_3). Our results showed that gene amplification is not the unique mechanism in EGFR overexpression. IHC might be used in the decision of anti-EGFR therapy in routine practice. p53 mutation rate was lower than the rates reported in the literature probably due to ethnic differences and low sensitivity of sanger sequences in general mutation screening.We also established the rarity of HER2 mutation in TNBC. In conclusion EGFR and p53 are the major targets in TNBC also for our population. C1 [Pala, Ebru Emel] Tepecik Education and Research Hospital, Pathology Department, Gaziler Caddesi, Yenisehir, 35000 Izmir, Turkey. [Bayol, Umit] Tepecik Education and Research Hospital, Pathology Department, Gaziler Caddesi, Yenisehir, 35000 Izmir, Turkey. [Keskin, Usturali Elif] Tepecik Education and Research Hospital, Pathology Department, Gaziler Caddesi, Yenisehir, 35000 Izmir, Turkey. [Ozguzer, Alp] Tepecik Education and Research Hospital, Pathology Department, Gaziler Caddesi, Yenisehir, 35000 Izmir, Turkey. [Kucuk, Ulku] Tepecik Education and Research Hospital, Pathology Department, Gaziler Caddesi, Yenisehir, 35000 Izmir, Turkey. [Ozer, Ozge] Genetic DepartmentIzmir, Turkey. [Koc, Altug] Genetic DepartmentIzmir, Turkey. RP Pala, EE (reprint author), Tepecik Education and Research Hospital, Pathology Department, 35000 Izmir, Turkey. EM emelozkok@yahoo.com CR Sorlie T, Perou CM, Tibshirani R et al, 2001, Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98(19):10869– 10874 Carey LA, Perou CM, Livasy CA et al, 2006, Race, breast cancer subtypes, and survival in the carolina breast cancer study. JAMA 295(21):2492–2502, PMID:16757721 Oakman C, Viale G, Di Leo A, 2010, Management of triple negative breast cancer. Breast 19(5):312–321., DOI 10.1016/j.breast. 2010.03.026, PMID:20382530 Carey LA, Rugo HS, Marcom PK et al, 2012, TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol 30(21):2615–2623., DOI 10.1200/JCO.2010.34.5579 O’Shaughnessy J, Osborne C, Pippen JE, Yoffe M, Patt D, Rocha C, Koo IC, Sherman BM, Bradley C, 2011, Iniparib plus chemotherapy in metastatic triple-negative breast cancer. N Engl J Med 364(3):205–214., DOI 10.1056/NEJMoa1011418 Langerod A, Zhao H, Borgan O, Nesland JM, Bukholm IR, Ikdahl T, Karesen R, Borresen-Dale AL, Jeffrey SS, 2007, TP53 mutation status and gene expression profiles are powerful prognosticmarkers of breast cancer. Breast Cancer Res 9(3):R30, PMID:17504517 Turner N, Moretti E, Siclari O, Migliaccio I, Santarpia L, D’Incalci M, Piccolo S, Veronesi A, Zambelli A, Del Sal G, Di Leo A, 2013, Targeting triple negative breast cancer: is p53 the answer? Cancer Treat Rev 39(5):541–550., DOI 10.1016/j.ctrv.2012.12.001 Bhargava R, GeraldWL, Li AR, Pan Q, Lal P, Ladanyi M, Chen B, 2005, EGFR gene amplification in breast cancer: correlation with epidermal growth factor receptor mRNA and protein expression and HER-2 status and absence of EGFR-activating mutations. Mod Pathol 18(8):1027–1033, PMID:15920544 Hirsch FR, Varella-Garcia M, Bunn PA Jr, Di Maria MV, Veve R, Bremmes RM, Baron AE, Zeng C, FranklinWA, 2003, Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol 21(20):3798–3807, PMID:12953099 Sorlie T, Tibshirani R, Parker J et al, 2003, Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A 100(14):8418–8423, PMID:12829800 Bertucci F, Finetti P, Cervera N, Esterni B, Hermitte F, Viens P, Birnbaum D, 2008, How basal are triple-negative breast cancers? Int J Cancer 123(1):236–240., DOI 10.1002/ijc.23518, PMID:18398844 PiresMM, HopkinsBD, Saal LH, Parsons RE, 2013, Alterations of EGFR, p53 and PTEN that mimic changes found in basal-like breast cancer promote transformation of human mammary epithelial cells. Cancer Biol Ther 14(3):246–253., DOI 10.4161/cbt.23297 Network CGA, 2012, Comprehensive molecular portraits of human breast tumours. Nature 490(7418):61–70., DOI 10.1038/ nature11412, PMID:23000897 Dumay A, Feugeas JP, Wittmer E et al, 2013, Distinct tumor protein p53 mutants in breast cancer subgroups. Int J Cancer 132(5): 1227–1231., DOI 10.1002/ijc.27767, PMID:22886769 Shah SP, Roth A, Goya R et al, 2012, The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature 486(7403):395–399., DOI 10.1038/nature10933, PMID:22495314 Grob TJ, Heilenkotter U, Geist S, Paluchowski P, Wilke C, Jaenicke F, Quaas A, Wilczak W, Choschzick M, Sauter G, Lebeau A, 2012, Rare oncogenic mutations of predictive markers for targeted therapy in triple-negative breast cancer. Breast Cancer Res Treat 134(2):561–567., DOI 10.1007/s10549- 012-2092-7, PMID:22610646 Burness ML, Grushko TA, Olopade OI, 2010, Epidermal growth factor receptor in triple-negative and basal-like breast cancer: promising clinical target or only a marker? Cancer J 16(1):23–32., DOI 10. 1097/PPO.0b013e3181d24fc1, PMID:20164687 Gumuskaya B, Alper M, Hucumenoglu S, Altundag K, Uner A, Guler G, 2010, EGFR expression and gene copy number in triplenegative breast carcinoma. Cancer Genet Cytogenet 203(2):222– 229., DOI 10.1016/j.cancergencyto.2010.07.118 Ryden L, Jirstrom K, Haglund M, Stal O, Ferno M, 2010, Epidermal growth factor receptor and vascular endothelial growth factor receptor 2 are specific biomarkers in triple-negative breast cancer. Results from a controlled randomized trial with long-term follow-up. Breast Cancer Res Treat 120(2):491–498., DOI 10.1007/ s10549-010-0758-6, PMID:20135347 Corkery B, Crown J, Clynes M, O’Donovan N, 2009, Epidermal growth factor receptor as a potential therapeutic target in triplenegative breast cancer. Ann Oncol 20(5):862–867., DOI 10.1093/ annonc/mdn710, PMID:19150933 Nogi H, Kobayashi T, Suzuki M, Tabei I, Kawase K, Toriumi Y, Fukushima H, Uchida K, 2009, EGFR as paradoxical predictor of chemosensitivity and outcome among triple-negative breast cancer. Oncol Rep 21(2):413–417, PMID:19148516 Siziopikou KP, Cobleigh M, 2007, The basal subtype of breast carcinomas may represent the group of breast tumors that could benefit from EGFR-targeted therapies. Breast 16(1):104–107, PMID:17097880 Shao MM, Zhang F, Meng G, Wang XX, Xu H, Yu XW, Chen LY, Tse GM, 2011, Epidermal growth factor receptor gene amplification and protein overexpression in basal-like carcinoma of the breast. Histopathology 59(2):264–273., DOI 10.1111/j.1365-2559. 2011.03921.x, PMID:21884205 Viale G, Rotmensz N, Maisonneuve P et al, 2009, Invasive ductal carcinoma of the breast with the "triple-negative" phenotype: prognostic implications of EGFR immunoreactivity. Breast Cancer Res Treat 116(2):317–328., DOI 10.1007/s10549-008-0206-z, PMID:18839307 Tang Y, Zhu L, Li Y, Ji J, Li J, Yuan F,Wang D, ChenW, Huang O, Chen X,Wu J, Shen K, LooWT, Chow LW(2012, Overexpression of epithelial growth factor receptor, EGFR, predicts better response to neo-adjuvant chemotherapy in patients with triple-negative breast cancer. J Transl Med 10(1):S4., DOI 10.1186/1479-5876-10- S1-S4, PMID:23046633 Personeni N, Fieuws S, Piessevaux H et al, 2008, Clinical usefulness of EGFR gene copy number as a predictive marker in colorectal cancer patients treated with cetuximab: a fluorescent in situ hybridization study. Clin Cancer Res 14(18): 5869–5876., DOI 10.1158/1078-0432.CCR-08-0449, PMID:18794099 Li C, Hao L, Li Y,Wang S, Chen H, Zhang L, Ke B, Yin Y, Suo H, Sun B, Zhang B, Wang C, 2014, Prognostic value analysis of mutational and clinicopathological factors in non-small cell lung cancer. PLoS ONE 9(9):e107276., DOI 10.1371/journal.pone.0107276, PMID:25198510 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1223 EP 1227 DI 10.1007/s12253-015-9956-1 PG 5 ER PT J AU Winczura, P Sosinska-Mielcarek, K Duchnowska, R Badzio, A Lakomy, J Majewska, H Peksa, R Pieczynska, B Radecka, B Debska-Szmich, S Adamowicz, K Biernat, W Jassem, J AF Winczura, Piotr Sosinska-Mielcarek, Katarzyna Duchnowska, Renata Badzio, Andrzej Lakomy, Joanna Majewska, Hanna Peksa, Rafal Pieczynska, Beata Radecka, Barbara Debska-Szmich, Sylwia Adamowicz, Krzysztof Biernat, Wojciech Jassem, Jacek TI Immunohistochemical Predictors of Bone Metastases in Breast Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Bone metastases; Predictive factors; Immunohistochemistry ID Breast cancer; Bone metastases; Predictive factors; Immunohistochemistry AB Bones are the most common metastatic site of relapse in breast cancer patients and the prediction of bone metastases (BM) risk might prompt developing preventive and therapeutic strategies. The aim of the study was to correlate imumohistochemical (IHC) expression of selected proteins in primary breast cancer with the occurrence of BM. We analyzed expression of proteins potentially associated with BM in primary tumors of 184 patients with metastatic breast cancer (113 with- and 71 without BM). Expression of estrogen receptor (ER) in primary tumor was more common in patients with- compared to those without BM (74 vs. 45 % respectively, p=0.0001), whereas in this subset less common was expression of parathyroid hormone related protein receptor type 1 (16 vs. 34 %, respectively, p=0.007) and cytoplasmic expression of osteopontin (OPNcyt; 1.9 vs. 14 %, respectively, p=0.002). The relationship between expression of ER and OPNcyt and the occurrence of BM was confirmed in the multivariate analysis. The ER-positive/OPNcyt negative phenotype was significantly more common in patients with- compared to those without BM (75 and 25 %, p<0.0001, respectively; HR 1.79, p=0.013). Luminal A (43 vs. 23 % respectively, p=0.009) and luminal B/HER2-positive (16 vs. 4.9 % respectively, p=0.032) subtypes were more common in patients with- compared to those without BM, whereas triple negative breast cancer subtype was less common (16 vs. 38 %, p=0.002). C1 [Winczura, Piotr] Medical University of GdanskGdansk, Poland. [Sosinska-Mielcarek, Katarzyna] Medical University of Gdansk, Department of Oncology and RadiotherapyGdansk, Poland. [Duchnowska, Renata] Military Institute of MedicineWarsaw, Poland. [Badzio, Andrzej] Medical University of GdanskGdansk, Poland. [Lakomy, Joanna] Medical University of GdanskGdansk, Poland. [Majewska, Hanna] Medical University of GdanskGdansk, Poland. [Peksa, Rafal] Medical University of GdanskGdansk, Poland. [Pieczynska, Beata] Medical University of GdanskGdansk, Poland. [Radecka, Barbara] Regional Oncology CenterOpole, Poland. [Debska-Szmich, Sylwia] Medical University of LodzLodz, Poland. [Adamowicz, Krzysztof] Medical University of Gdansk, Department of Oncology and RadiotherapyGdansk, Poland. [Biernat, Wojciech] Medical University of GdanskGdansk, Poland. [Jassem, Jacek] Medical University of GdanskGdansk, Poland. RP Winczura, P (reprint author), Medical University of Gdansk, Gdansk, Poland. EM pwinczura@gmail.com CR Tubiana-Hulin M, 1991, Incidence prevalence and distribution of bone metastases. Bone, Suppl. 1):9–10 Coleman RE, Smith P, Rubens RD, 1998, Clinical course and prognostic factors following bone recurrence from breast cancer. Br J Cancer 77:336–340 Plunkett TA, Smith P, Rubens RD, 2000, Risk of complications from bone metastases in breast cancer. Implications for management. Eur J Cancer 36:476–482 Guise TA, Mohammad KS, Clines G, Stebbins EG, Wong DH, Higgins LS et al, 2006, Basic mechanisms responsible for osteolytic and osteoblastic bone metastases. Clin Cancer Res 12: 6213–6216 Kennecke H, Yerushalmi R, Woods R, Cheang MC, Voduc D, Speers CH et al, 2010, Metastatic behavior of breast cancer subtypes. J Clin Oncol 28:3271–3277 Hess KR, Pusztai L, Buzdar AU, Hortobagyi GN, 2003, Estrogen receptors and distinct patterns of breast cancer relapse. Breast Cancer Res Treat 78:105–118 Sihto H, Lundin J, Lundin M, Lehtimaki T, Ristimaki A, Holli K et al, 2011, Breast cancer biological subtypes and protein expression predict for the preferential distantmetastasis sites: a nationwide cohort study. Breast Cancer Res 13:R87 Gnant M, Mlineritsch B, Stoeger H, Luschin-Ebengreuth G, Heck D, Menzel C et al, 2011, Adjuvant endocrine therapy plus zoledronic acid in premenopausal women with earlystage breast cancer: 62-month follow-up from the ABCSG- 12 randomised trial. Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria. Lancet Oncol 12:631–641 Coleman RE, Marshall H, Cameron D, Dodwell D, Burkinshaw R, KeaneM et al, 2011, Breast cancer adjuvant therapy with zoledronic acid. N Engl J Med 365:1396–1405 Singh B, Berry JA, Shoher A, Lucci A, 2006, COX-2 induces IL- 11 production in human breast cancer cells. J Surg Res 131:267– 275 Andre F, Xia W, Conforti R, Wei Y, Boulet T, Tomasic G et al, 2009, CXCR4 expression in early breast cancer and risk of distant recurrence. Oncologist 14:1182–1188 Linforth R, Anderson N, Hoey R, Nolan T, Downey S, Brady G et al, 2002, Coexpression of parathyroid hormone related protein and its receptor in early breast cancer predicts poor patient survival. Clin Cancer Res 8:3172–3177 Hoey RP, Sanderson C, Iddon J, Brady G, Bundred NJ, Anderson NG, 2003, The parathyroid hormone-related protein receptor is expressed in breast cancer bone metastases and promotes autocrine proliferation in breast carcinoma cells. Br J Cancer 88:567–573 Asou Y, Rittling SR, Yoshitake H, Tsuji K, Shinomiya K, Nifuji A et al, 2001, Osteopontin facilitates angiogenesis, accumulation of osteoclasts, and resorption in ectopic bone. Endocrinology 142: 1325–1332 Patani N, Jouhra F, JiangW,Mokbel K, 2008, Osteopontin expression profiles predict pathological and clinical outcome in breast cancer. Anticancer Res 28:4105–4110 Mihai R, Stevens J,McKinney C, IbrahimNB, 2006, Expression of the calcium receptor in human breast cancer—a potential new marker predicting the risk of bone metastases. Eur J Surg Oncol 32:511–515 Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S et al, 2010, American Society of Clinical Oncology/ College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer, unabridged version). American Society of Clinical Oncology; College of American Pathologists. Arch Pathol Lab Med 134:48–72 Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ et al, 2007, American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. American Society of Clinical Oncology/ College of American Pathologists. Arch Pathol Lab Med 131: 18–43 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ et al, 2011, Strategies for subtypes—dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol 22:1736–1747 Dowsett M, Nielsen TO, A’Hern R, Bartlett J, Coombes RC, Cuzick J et al, 2011, Assessment of Ki67 in breast cancer: recommendations from the International Ki67 in Breast Cancer working group. International Ki-67 in Breast CancerWorking Group. J Natl Cancer Inst 103:1656–1664 Diallo-Danebrock R, Ting E, Gluz O, Herr A, Mohrmann S, Geddert H et al, 2007, Protein expression profiling in highrisk breast cancer patients treated with high-dose or conventional dose-dense chemotherapy. Clin Cancer Res 13:488– 497 Ristimaki A, Sivula A, Lundin J, Lundin M, Salminen T, Haglund C et al, 2002, Prognostic significance of elevated cyclooxygenase-2 expression in breast cancer. J Cancer Res 62:632–635 Yasuoka H, Tsujimoto M, Yoshidome K, Nakahara M, Kodama R, Sanke T et al, 2008, Cytoplasmic CXCR4 expression in breast cancer: induction by nitric oxide and correlation with lymph node metastasis and poor prognosis. BMC Cancer 8:340 Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordon-Cardo C et al, 2003, A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3:537–549 Ibrahim T, Sacanna E, Gaudio M, Mercatali L, Scarpi E, Zoli W et al, 2011, Role of RANK, RANKL, OPG, and CXCR4 tissue markers in predicting bone metastases in breast cancer patients. Clin Breast Cancer 11:369–375 MacLeod RJ, Yano S, Chattopadhyay N, 2004, Extracellular calcium-sensing receptor transactivates the epidermal growth factor receptor by a triple-membrane-spanning signaling mechanism. Biochem Biophys Res Commun 320:455–460 Craig AM, Bowden GT, Chambers AF, Spearman MA, Greenberg AH, Wright JA et al, 1990, Secreted phosphoprotein mRNA is induced during multi-stage carcinogenesis in mouse skin and correlates with the metastatic potential of murine fibroblasts. Int J Cancer 46:133–137 Junaid A, Moon MC, Harding GE, Zahradka P, 2007, Osteopontin localizes to the nucleus of 293 cells and associates with polo-like kinase-1. Am J Physiol Cell Physiol 292:919–926 Luparello C, Burtis WJ, Raue F, Birch MA, Gallagher JA, 1995, Parathyroid hormone-related peptide and 8701-BC breast cancer cell growth and invasion in vitro: evidence for growth-inhibiting and invasion-promoting effects. Mol Cell Endocrinol 111:225–232 Akino K, Ohtsuru A, Kanda K, Yasuda A, Yamamoto T, Akino Y et al, 2000, Parathyroid hormone-related peptide is a potent tumor angiogenic factor. Endocrinology 141:4313–4316 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1229 EP 1236 DI 10.1007/s12253-015-9957-0 PG 8 ER PT J AU Singh, KM Singh, L Pushker, N Sen, S Sharma, A Chauhan, AF Kashyap, S AF Singh, Kumar Mithalesh Singh, Lata Pushker, Neelam Sen, Seema Sharma, Anjana Chauhan, Ahamad Feeroj Kashyap, Seema TI Correlation of High Mobility Group Box-1 Protein (HMGB1) with Clinicopathological Parameters in Primary Retinoblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Retinoblastoma; Immunohistochemistry; HMGB1; RT-PCR ID Retinoblastoma; Immunohistochemistry; HMGB1; RT-PCR AB HMGB1 is considered to be DNA chaperone as it binds without any specificity. It is the structural protein which alters nuclear homeostasis and genomic stability of chromatin. Its role in retinoblastoma (Rb) remains unclear. The aim of the present study was to evaluate the expression of HMGB1 protein in primary enucleated retinoblastomas. Expression of HMGB1 in 69 prospective cases of primary retinoblastoma were assessed by immunohistochemistry and reverse transcriptase PCR (RT-PCR) technique and correlated with clinicopathological parameters. Immunohistochemical staining revealed expression of HMGB1 in 55.07%(38/69) cases. Semiquantitative RT-PCR was performed on 31 fresh tumor tissues. mRNA expression was observed in 77.41 % (24/31) cases. Expression of HMGB1 was statistically significant with poor tumor differentiation (p=0.0440) & optic nerve invasion (p=0.0128).HMGB1 expression was frequently seen in poorly differentiated tumors and those with histopathological high risk factors. Therefore, HMGB1 may contribute to tumor invasiveness and could serve as a poor prognostic marker in Rb. C1 [Singh, Kumar Mithalesh] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular PathologyNew Delhi, India. [Singh, Lata] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular PathologyNew Delhi, India. [Pushker, Neelam] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of OphthalmologyNew Delhi, India. [Sen, Seema] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular PathologyNew Delhi, India. [Sharma, Anjana] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular MicrobiologyNew Delhi, India. [Chauhan, Ahamad Feeroj] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular PathologyNew Delhi, India. [Kashyap, Seema] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular PathologyNew Delhi, India. RP Kashyap, S (reprint author), All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular Pathology, New Delhi, India. EM dr_skashyap@hotmail.com CR Kivela T, 2009, The epidemiological challenge of the most frequent eye cancer: retinoblastoma, an issue of birth and death. Br J Ophthalmol 93:1129–1131 Comings DE, 2009, A general theory of carcinogenesis. Proc Natl Acad Sci U S A 70:3324–3328 Friend SH, Bernards R, Rogelj S, Weinberg RA, Rapaport JM, Albert DM, Dryja TP, 1986, A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature 323:643–646 Knudson AG Jr, 1971, Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci U S A 68:820–823 Hock R, Furusawa T, Ueda T, BustinM(2007)HMG chromosomal proteins in development and disease. Trends Cell Biol 17:72–79 Grosschedl R, Giese K, Pagel J, 1994, HMG domain proteins: architectural elements in the assembly of nucleoprotein structures. Trends Genet 10:94–100 Bustin M, 2001, Revised nomenclature for high mobility group, HMG, chromosomal proteins. Trends Biochem Sci 3:152–153 Goodwin GH, Johns EW, 1977, The isolation and purification of the high mobility group, HMG, nonhistone chromosomal proteins. Methods Cell Biol 16:257–267 Cui XS, Shen XH, Kim NH, 2007, High mobility group box 1, HMGB1, is implicated in preimplantation embryo development in the mouse. Mol Reprod Dev 75:1290–1299 Lange SS, Vasquez KM(2009)HMGB1: the jack-of-all-trades protein is a master DNA repair mechanic. Mol Carcinog 48:571–580 Ferrari S, Finelli P, Rocchi M, Bianchi ME, 1996, The active gene that encodes human high mobility group 1 protein, HMG1, contains introns and maps to chromosome 13. Genomics 35:367–371 Tang D, Kang R, Zeh HJ 3rd, Lotze MT, 1799, High-mobility group box 1 and cancer. Biochim Biophys Acta 2010:131 Flohr AM, Rogalla P, Meiboom M, Borrmann L, Krohn M, Thode- Halle B, Bullerdiek J, 2001, Variation of HMGB1 expression in breast cancer. Anticancer Res 21:3881–3885 Poser I, Bosserhoff AK, 2004, Transcription factors involved in development and progression of malignant melanoma. Histol Histopathol 19:173–178 Choi YR, Kim H, Kang HJ, Kim NG, Kim JJ, Park KS, Paik YK, KimHO, KimH(2003, Overexpression of high mobility group box 1 in gastrointestinal stromal tumors with KIT mutation. Cancer Res 63:2188–2193 Meyer A, Staratschek-Jox A, Springwald A, Wenk H, Wolf J, Wickenhauser C, Bullerdiek J, 2008, Non-Hodgkin lymphoma expressing high levels of the danger-signalling protein HMGB1. Leuk Lymphoma 49:1184–1189 Mu G, Liu H, Zhou F, Xu X, Jiang H, Wang Y, Qu Y, 2010, Correlation of overexpression of HMGA1 and HMGA2 with poor tumor differentiation, invasion, and proliferation associated with let-7 down-regulation in retinoblastomas. Hum Pathol 41:493–502 Venkatesan N, Kandalam M, Pasricha G, Sumantran V, Manfioletti G, Ono SJ, ReddyMA, Krishnakumar S, 2009, Expression of high mobility group A2 protein in retinoblastoma and its association with clinicopathologic features. J Pediatr Hematol Oncol 31:209– 214 Finger PT, Harbour JW, Murphree AL, 2009, Retinoblastoma. In: Edge DR, Byrd DR, Compton CC, eds, AJCC cancer staging manual, vol 7. Springer, New York, pp 561–566 Jiao Y,Wang HC, Fan SJ, 2007, Growth suppression and radiosensitivity increase by HMGB1 in breast cancer. Acta Pharmacol Sin 28:1957–1967 Liu K, Huang J, XieM, Yu Y, Zhu S, Kang R, Cao L, Tang D, Duan X, 2014, MIR34A regulates autophagy and apoptosis by targeting HMGB1 in the retinoblastoma cell. Autophagy 10:442–452 Yang GL, Zhang LH, Bo JJ, Huo XJ, Chen HG, Cao M, Liu DM, Huang YR, 2012, Increased expression of HMGB1 is associated with poor prognosis in human bladder cancer. J Surg Oncol 106:57–61 Bao G, Qiao Q, Zhao H, He X, 2010, Prognostic value of HMGB1 overexpression in resectable gastric adenocarcinomas.World J Surg Oncol 8:52–58 Hao Q, Du XQ, Fu X, Tian J, 2008, Expression and clinical significance of HMGB1 and RAGE in cervical squamous cell carcinoma. Zhonghua Zhong Liu Za Zhi 30:292–295 Yao X, Zhao G, Yang H, Hong X, Bie L, Liu G, 2010, Overexpression of highmobility group box 1 correlates with tumor progression and poor prognosis in human colorectal carcinoma. J Cancer Res Clin Oncol 136:677–684 Wu D, Ding Y, Wang S, Zhang Q, Liu L, 2008, Increased expression of high mobility group box 1, HMGB1, is associated with progression and poor prognosis in human nasopharyngeal carcinoma. J Pathol 216:167–175 Cheng BQ, Jia CQ, Liu CT, Lu XF, Zhong N, Zhang ZL, FanW, Li YQ, 2008, Serum high mobility group box chromosomal protein 1 is associated with clinicopathologic features in patients with hepatocellular carcinoma. Dig Liver Dis 40:446–452 Liu F, Zhang Y, Peng Z, Gao H, Xu L, Chen M, 2012, High expression of high mobility group box 1, hmgb1, predicts poor prognosis for hepatocellular carcinoma after curative hepatectomy. J Transl Med 10:135–144 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1237 EP 1242 DI 10.1007/s12253-015-9951-6 PG 6 ER PT J AU Avci, N Ture, M Deligonul, A Cubukcu, E Olmez, FO Sahinturk, S Topak, A Kurt, E Evrensel, T Sahin, BA Yakut, T AF Avci, Nilufer Ture, Mehmet Deligonul, Adem Cubukcu, Erdem Olmez, Fatih Omer Sahinturk, Serdar Topak, Ali Kurt, Ender Evrensel, Turkkan Sahin, Bilgehan Ahmet Yakut, Tahsin TI Association and Prognostic Significance of the Functional −1562C/T Polymorphism in the Promoter Region of MMP-9 in Turkish Patients with Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Matrix metalloproteinases; Polymorphism; Association study; Prognosis ID Gastric cancer; Matrix metalloproteinases; Polymorphism; Association study; Prognosis AB Matrix metalloproteinases (MMPs) are a group of zinc-dependent peptidases that participate in matrix turnover in solid malignancies. The aim of this study was twofold. First, we sought to investigate under a case–control design the association between the functional −1562C/T polymorphism in the promoter region of MMP-9 and gastric cancer (GC) in a Turkish sample. Second, we examined its prognostic significance in GC patients. A total of 144 subjects were enrolled in the case–control study (79 GC cases and 65 controls). Overall survival (OS) and progression-free survival (PFS) served as the main outcome measures in the longitudinal study. The MMP-9 −1562C/T polymorphism was genotyped using a polymerase chain reaction-restriction fragment length polymorphism method. The odds ratio (OR) of GC for the CC genotype relative to the CT+TT genotypes was not significant (OR=0.89, 95 % confidence interval [CI]=0.44–1.82, P=0.75). These results did not change after allowance for age and sex in multivariable regression analysis (OR=0.81, 95 % CI=0.40–1.94, P=0.84). When the MMP-9 −1562C/T polymorphism was analyzed among GC patients in relation to OS and PFS, we found no significant differences between subjectswith the CC and CT+TT genotypes. In conclusion, the results of our study did not point toward a major role of the MMP-9−1562C/T polymorphism in the pathogenesis and clinical course of GC in Turkish subjects. C1 [Avci, Nilufer] Ali Osman Sonmez Oncology Hospital, Department of Medical OncologyBursa, Turkey. [Ture, Mehmet] Uludag University Medicine Faculty, Department of Medical GeneticsBursa, Turkey. [Deligonul, Adem] Uludag University Medicine Faculty, Department of Medical OncologyBursa, Turkey. [Cubukcu, Erdem] Ali Osman Sonmez Oncology Hospital, Department of Medical OncologyBursa, Turkey. [Olmez, Fatih Omer] Medipol University School of Medicine, Department of Medical OncologyIstanbul, Turkey. [Sahinturk, Serdar] Uludag University Medicine Faculty, Department of Medical GeneticsBursa, Turkey. [Topak, Ali] Uludag University Medicine Faculty, Department of Medical GeneticsBursa, Turkey. [Kurt, Ender] Uludag University Medicine Faculty, Department of Medical OncologyBursa, Turkey. [Evrensel, Turkkan] Uludag University Medicine Faculty, Department of Medical OncologyBursa, Turkey. [Sahin, Bilgehan Ahmet] Uludag University Medicine Faculty, Department of Internal MedicineBursa, Turkey. [Yakut, Tahsin] Uludag University Medicine Faculty, Department of Medical GeneticsBursa, Turkey. RP Avci, N (reprint author), Ali Osman Sonmez Oncology Hospital, Department of Medical Oncology, Bursa, Turkey. EM drniluferavci@hotmail.com CR Fock KM, 2014, Review article: the epidemiology and prevention of gastric cancer. Aliment Pharmacol Ther 40:250–260 de Martel C, Forman D, Plummer M, 2013, Gastric cancer: epidemiology and risk factors. Gastroenterol Clin N Am 42:219–240 Lordick F, Allum W, Carneiro F, Mitry E, Tabernero J, Tan P, Van Cutsem E, van de Velde C, Cervantes A, 2014, Unmet needs and challenges in gastric cancer: the way forward. Cancer Treat Rev 40: 692–700 CrewKD, Neugut AI, 2006, Epidemiology of gastric cancer.World J Gastroenterol 12:354–362 Shi J, Qu YP, Hou P, 2014, Pathogenetic mechanisms in gastric cancer. World J Gastroenterol 20:13804–13819 McLean MH, El-Omar EM, 2014, Genetics of gastric cancer. Nat Rev Gastroenterol Hepatol 11:664–674 Yadav L, Puri N, Rastogi V, Satpute P, Ahmad R, Kaur G, 2014, Matrix metalloproteinases and cancer—roles in threat and therapy. Asian Pac J Cancer Prev 15:1085–1091 Nissinen L, Kahari VM, 2014, Matrix metalloproteinases in inflammation. Biochim Biophys Acta 1840:2571–2580 Verma S, Kesh K, Ganguly N, Jana S, Swarnakar S, 2014, Matrix metalloproteinases and gastrointestinal cancers: impacts of dietary antioxidants. World J Biol Chem 5:355–376 Sampieri CL, Leon-Cordoba K, Remes-Troche JM, 2013, Matrix metalloproteinases and their tissue inhibitors in gastric cancer as molecular markers. J Cancer Res Ther 9:356–363 Chen J, Chen LJ, Zhou HC, Yang RB, Lu Y, Xia YL, Wu W, Hu LW, 2014, Prognostic value of matrix metalloproteinase-9 in gastric cancer: a meta-analysis. Hepatogastroenterology 61:518–524 ZhangQW, Liu L, Chen R,Wei YQ, Li P, Shi HS, ZhaoYW(2012, Matrix metalloproteinase-9 as a prognostic factor in gastric cancer: a meta-analysis. Asian Pac J Cancer Prev 13:2903–2908 Singh K, Nair RR, Khanna A, 2012, Functional SNP -1562C/T in the promoter region of MMP9 and recurrent early pregnancy loss. Reprod Biomed Online 24:61–65 Li J, Lu H, Tao F, Zhou H, Feng G, He L, Zhou L, 2013, Metaanalysis of MMP9-1562C/T and the risk of coronary heart disease. Cardiology 124:53–59 Gong LL, Liu H, Liu LH, 2014, Lack of association between matrix metalloproteinase-9 gene-1562C/T polymorphism and preeclampsia: a meta-analysis. Hypertens Pregnancy 33:389–394 Yang TF, Guo L, Wang Q, 2014, Meta-analysis of associations between four polymorphisms in the matrix metalloproteinases gene and gastric cancer risk. Asian Pac J Cancer Prev 15:1263–1267 Hirschhorn JN, Lohmueller K, Byrne E, Hirschhorn K, 2002, A comprehensive review of genetic association studies. GenetMed 4: 45–61 Wang L, Ma YT, Xie X, Yang YN, Fu ZY, Liu F, Li XM, Chen BD, 2011, Association ofMMP-9 gene polymorphisms with acute coronary syndrome in the Uygur population of China. World J Emerg Med 2:104–110 Emanuele E, Lista S, Ghidoni R, Binetti G, Cereda C, Benussi L, Maletta R, Bruni AC, Politi P, 2011, Chromosome 9p21.3 genotype is associated with vascular dementia and Alzheimer’s disease. Neurobiol Aging 32:1231–1235 Emanuele E, Bertona M, Geroldi D, 2010, A multilocus candidate approach identifies ACE and HIF1A as susceptibility genes for cellulite. J Eur Acad Dermatol Venereol 24:930–935 Schveigert D, Valuckas KP, Kovalcis V, Ulys A, Chvatovic G, Didziapetriene J, 2013, Significance of MMP-9 expression and MMP-9 polymorphism in prostate cancer. Tumori 99:523–529 Chiranjeevi P, Spurthi KM, Rani NS, Kumar GR, Aiyengar TM, Saraswati M, Srilatha G, Kumar GK, Sinha S, Kumari CS, Reddy BN,Vishnupriya S, Rani HS, 2014, Gelatinase B, −1562C/T, polymorphism in tumor progression and invasion of breast cancer. Tumour Biol 35:1351–1356 El Samanoudy A, Monir R, Badawy A, Ibrahim L, Farag K, El Baz S, Alenizi D, Alenezy A, 2014, Matrix metalloproteinase-9 gene polymorphism in hepatocellular carcinoma patients with hepatitis B and C viruses. Genet Mol Res 13:8025–8034 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1243 EP 1247 DI 10.1007/s12253-015-9950-7 PG 5 ER PT J AU Mallick, S Breta, M Gupta, DS Dinda, KA Mohanty, KB Singh, KM AF Mallick, Saumyaranjan Breta, Monika Gupta, Datta Siddhartha Dinda, Kumar Amit Mohanty, K Biddhu Singh, K Manoj TI Angiogenesis, Proliferative Activity and DNA Ploidy in Oral Verrucous Carcinoma: A Comparative Study Including Verrucous Hyperplasia and Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Angiogenesis; Proliferative activity; Ploidy; Verrucous carcinoma ID Angiogenesis; Proliferative activity; Ploidy; Verrucous carcinoma AB Verrucous carcinoma (VC) is a rare and distinct clinicopathologic variant of well-differentiated squamous cell carcinoma (SCC). This study aims to evaluate the histomorphology, proliferative activity, level of angiogenesis, and DNA ploidy of these pathological entities. This was a retrospective-prospective study of 18 cases of verrucous hyperplasia (VH), 41 cases of VC, and 44 cases of SCC. Immunohistochemical analysis for Ki-67 (MIB-1) and CD34 were performed. The tumor proliferative index, endothelial proliferative index and microvascular density were calculated. DNA ploidy was determined using image cytometry. The age range and gender ratio were similar in all three groups. The differences in MIB-1 labeling index (p=0.0001), microvascular density (p=0.01), and endothelial proliferative index (p= 0.001) between VC and SCC were found to be statistically significant. A non-significant increasing trend was observed in all of these parameters between VH and VC. On ploidy analysis, 100 % of SCC cases were aneuploid, compared to 39 % of VH and 86 % of VC cases. Our study demonstrates a significant difference in tumor proliferation, microvessel density, and ploidy between VC and SCC while increasing trend between VH and VC. These parameters, along with morphological findings, may be useful in differentiating these entities in small mucosal biopsies. C1 [Mallick, Saumyaranjan] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Breta, Monika] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Gupta, Datta Siddhartha] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Dinda, Kumar Amit] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Mohanty, K Biddhu] All India Institute of Medical Sciences, Department of Radiation oncology, 110029 New Delhi, India. [Singh, K Manoj] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. RP Singh, KM (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM makusi@hotmail.com CR Ackerman LV, 1948, Verrucous carcinoma of oral cavity. Surgery 23:670–81 McCoy JM, Waldron CA, 1981, Verrucous carcinoma of the oral cavity: a review of 49 cases. Oral Surg 52:623–9 Eisenberg E, Rosenberg B, Krutchkoff DJ, 1985, Verrucous Carcinoma: a possible viral pathogenesis. Oral Surg Oral Med Oral Pathol 59:52–7 Goethals PL, Harrison EG, Devine KD, 1963, Verrucous squamous cell carcinoma of oral canity. Am J Surg 106:845–51 Gassenmaier A, Hornstein OP, 1988, Presence of papilloma virus DNA in benign and precancerous oral leukoplakias and squamous cell carcinoma. Dermatologica 176:224–33 Trivedy CR, Craig G, Warnakulasuriya S, 2002, The oral health consequences of chewing areca nut. Addict Biol 7:115–25 Fujita S, Senba M, Kumatori A et al, 2008, Human papillomavirus infection in oral verrucous carcinoma: genotyping analysis and inverse correlation with p53 expression. Pathobiology 75:257–64 Medina JE, DichtelW, Luna MA, 1984, Verrucous squamous carcinoma of the oral cavity: a clinicopathological study of 104 cases. Arch Otolaryngol 110:437–40 Rekha KP, Angadi PV, 2010, Verrucous carcinoma of the oral cavity: a clinico-pathologic appraisal of 133 cases in Indians. Oral Maxillofac Surg 14:211–8 Kraus FT, Perz-mesa C, 1966, Verrucous carcinoma clinical and pathologic study of 105 cases involving oral cavity, larynx genitalia. Cancer 19:26–33 Fonts EA, Greenlaw RH, Rush BF et al, 1969, Verrucous squamous cell carcinoma of the oral cavity. Cancer 23:152–9 Ali Sbai M, Balti W, Dhahak S et al, 2009, Buschke Lowenstein tumor: unusual bilateral localization. Tunis Med 87:627–9 Pattee SF, Bordeaux J, Mahalingam M et al, 2007, Verrucous carcinoma of the scalp. J Am Acad Dermatol 56:506–17 Chiheb S, Bouziane K, Azzouzi S et al, 2010, Verrucous carcinoma of the toe. Ann Dermatol Venereol 137:169–70 Ackerman LV, McGavran, 1958, Proliferating benign and malignant potential lesion of the oral cavity. J Oral Surg 16:400–13 Kang CJ, Chang JT, Chen TM et al, 2003, Surgical treatment of oral verrucous carcinoma. Chang Gung Med J 26:807–12 Sakurai K, Urade M, Takahashi Y et al, 2000, Increased expression of c-erb- 3 protein and proliferating cell nuclear antigen during development of verrucous carcinoma of oral mucosa. Cancer 89: 2597–605 Theegarten D, Barfuss A, Gellirich NC et al, 1997, Expression of proliferation markers PCNA and MIB1 in verrucous squamous epithelial carcinoma of oral cavity. Mund Kiefer Gesichtschir 1:133–6 Ogawa A, Fukuta Y, Nakajima Tet al, 2004, Treatment results of oral verrucous carcinoma and its biological behavior. Oral Oncol 40:793– 7 Angadi PV, Krishnapillai R, 2007, Cyclin D1 expression in oral squamous cellcarcinoma and verrucous carcinoma: correlation with histological differentiation. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 103:e30–5 Hemmer J, Kraft K, 2000, High resolution DNA flow cytometry in oral verrucous carcinoma. Oncol Rep 7:433–35 Fitzpatrick SG, Neuman AN, CohenDMet al, 2012, The clinical and histologic presentation of gingival squamous cell carcinoma: a study of 519 cases. Oral Surg Oral Med Oral Pathol Oral Radiol 114:509– 15 Winn DM, Blot WV, Shy CM et al, 1981, Snuff dipping and oral cancer among women in the Southern United States. N Engl J Med 304:745–9 Shear M, Pindborg JJ, Odont D, 1980, Verrucous hyperplasia of the oral mucosa. Cancer 46:1855–62 Folkman J, 1995, Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med 1:27–31 Folkman J, 1971, Tumor angiogenesis: therapeutic implications. N Engl J Med 285:1182–6 Weidner N, Semple JP, Welch WR et al, 1991, Tumor angiogenesis and metastasis- correlation in invasive breast carcinoma. N Engl J Med 3(324):1–8 Vermeulen PB, Gasparini G, Fox SB et al, 1996, Quantification of angiogenesis in solid human tumor: an international consensus on methodology and criteria of evaluation. Eur J Cancer 32: 2472–84 Hannen EMJ, Riediger D, 2004, The quantification of angiogenesis in relation to metastatic oral cancer: a review. Int J Oral Maxillofac Surg 33:2–7 Lorincz T, Toth J, Szendroi M et al, 2005, Microvascular density of breast cancer in bone metastasis: influence of therapy. Anticancer Res 25:3075–81 Macchiarini P, Fontanini G, Hardin MJ et al, 1992, Relation of neovascularisation to metastasis of non-small-cell lung cancer. Lancet 340:145–46 Srivastava A, Laidler P, Davies RP et al, 1988, The prognostic significance of tumor vascularity in intermediate-thickness, 0.76– 4.0 mm thick, skin melanoma. A quantitative histologic study. Am J Pathol 133:419–23 Oliveira-Neto HH, Gleber-Netto FO, de Sousa SF et al, 2012, A comparative study of microvessel density in squamous cell carcinoma of the oral cavity and lip. Oral Surg Oral Med Oral Pathol Oral Radiol 113:391–8 El-Rouby DH, 2010, Association of macrophages with angiogenesis in oral verrucous and squamous cell carcinomas. J Oral Pathol Med 39:559–64 Abbas NF, Labib El-Sharkawy S, Abbas EA et al, 2007, Immunohistochemical study of p53 and angiogenesis in benign and preneoplastic oral lesions and oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 103:385–90 Mashhadiabbas F, Mahjour F, Mahjour SB et al, 2012, The immunohistochemical characterization of MMP-2, MMP-10, TIMP-1, TIMP-2, and podoplanin in oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol 114:240–50 Matsumoto Y, Ishiko O, Nishimura S et al, 2000, Angiogenesis in metastatic verrucous carcinoma of the uterine cervix. Oncol Rep 7: 1079–82 Macluskey M, Chandrachud LM, Pazouki S et al, 2000, Apoptosis, proliferation, and angiogenesis in oral tissues: possible relevance to tumour progression. J Pathol 191:368–75 Maraki D, Boecking A, Pomjanski N et al, 2006, Verrucous carcinoma of the buccalmucosa: histopathological, cytological and DNAcytometric features. J Oral Pathol Med 35:633–5 Ng SP, Mann IS, Zed C et al, 2012, The use of quantitative cytology in identifying high-risk oral lesions in community practice. Oral Surg Oral Med Oral Pathol Oral Radiol 114:358–64 Kahn MA, Dockter ME, Hermann-Petrin JM, 1994, Proliferative verrucous leukoplakia. Four cases with flow cytometric analysis. Oral Surg Oral Med Oral Pathol 78:469–75 Klijanienko J, el-Naggar AK, de Braud F et al, 1995, Tumor vascularization, mitotic index, histopathologic grade, and DNA ploidy in the assessment of 114 head and neck squamous cell carcinomas. Cancer 75:1649–56 Cristi E, Perrone G, Toscano G et al, 2005, Tumour proliferation, angiogenesis, and ploidy status in human colon cancer. J Clin Pathol 58:1170–4 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1249 EP 1257 DI 10.1007/s12253-014-9856-9 PG 9 ER PT J AU Zhu, L Liu, J Shenglin, M Zhang, Sh AF Zhu, Lucheng Liu, Jihong Shenglin, Ma Zhang, Shirong TI Long Noncoding RNA MALAT-1 Can Predict Metastasis and a Poor Prognosis: a Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MALAT-1; LncRNA; Metastasis; Prognosis; Survival; Meta-analysis ID MALAT-1; LncRNA; Metastasis; Prognosis; Survival; Meta-analysis AB Elevated expression of MALAT-1 was found in various cancers, and correlated with metastasis and prognostic. This meta-analysis collected all relevant articles and explored correlation of MALAT-1 with lymph node metastasis (LNM), distant metastasis (DM), and overall survival (OS). A quantitative meta-analysis was performed through a systematic search in PubMed,Web of Science, Medline, CNKI, CBM, and the Cochrane Library. The odds ratios (OR) of LNM and DM and hazard ratio (HR) of OS were calculated to assess the association strength. Eight studies with a total of 845 patients were included in the meta-analysis. Six different types of cancer were evaluated, with 2 non-small cell lung cancer (NSCLC), 1 colorectal cancer (CRC), 1 gastric cancer (GC), 2 pancreatic cancer (PC), 1 clear cell renal cell carcinoma (ccRCC), and 1 osteosarcoma (OSA). Compared with low MALAT-1 expression, high MALAT-1 expression correlated with more LNM (OR=2.08, 95 %CI: 1.00-4.32, p=0.05) by a random-effects model (I2=71 %, p=0.004). A similar result was seen between MALAT-1 expression and DM, the OR was 3.52 (95 %CI: 1.06–11.71, p=0.04) adopting a randomeffects model (I2=59 %, p=0.04). Additionally, our analysis showed a poorer OS in patients with high MALAT-1 expression than those with low MALAT-1 expression (HR=2.12, 95 %CI: 1.60–2.82, p<0.001) adopting a random-effects model (I2=56 %, p=0.04). MALAT-1 may serve as a molecular marker for cancer metastasis and prognosis. C1 [Zhu, Lucheng] Hangzhou First People’s Hospital, Department of Medical Oncology, No.261, huansha Road, shangcheng District, 310006 Hangzhou, China. [Liu, Jihong] Hangzhou First People’s Hospital, Department of Medical Oncology, No.261, huansha Road, shangcheng District, 310006 Hangzhou, China. [Shenglin, Ma] Hangzhou First People’s Hospital, Department of Medical Oncology, No.261, huansha Road, shangcheng District, 310006 Hangzhou, China. [Zhang, Shirong] Hangzhou First People’s Hospital, Department of Medical Oncology, No.261, huansha Road, shangcheng District, 310006 Hangzhou, China. RP Shenglin, M (reprint author), Hangzhou First People’s Hospital, Department of Medical Oncology, 310006 Hangzhou, China. EM mashenglin@outlook.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2014, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer., DOI 10.1002/ijc.29210 Pereira A, Perez-Medina T, Magrina JF, Magtibay PM, Rodriguez- Tapia A, de Leon J, Peregrin I, Ortiz-Quintana L, 2014, Correlation between the extent of intraperitoneal disease and nodal metastasis in node-positive ovarian cancer patients. Eur J Surg Oncol 40(8):917– 924., DOI 10.1016/j.ejso.2014.04.001 Lee T, Tanaka H, Ohira M, Okita Y, Yoshii M, Sakurai K, Toyokawa T, Kubo N, Muguruma K, Tanaka S, Ohsawa M, Hirakawa K, 2014, Clinical impact of the extent of lymph node micrometastasis in undifferentiated-type early gastric cancer. Oncology 86(4):244–252., DOI 10.1159/000358803 Treilleux I, Arnedos M, Cropet C, Wang Q, Ferrero JM, Abadie- Lacourtoisie S, Levy C, Legouffe E, Lortholary A, Pujade-Lauraine E, Bourcier AV, Eymard JC, Spaeth D, Bachelot T, 2014, Translational studies within the TAMRAD randomized GINECO trial: evidence for mTORC1 activation marker as a predictive factor for everolimus efficacy in advanced breast cancer. Ann Oncol., DOI 10.1093/annonc/mdu497 Hu A, Huang JJ, Xu WH, Jin XJ, Li JP, Tang YJ, Huang XF, Cui HJ, Sun GB, 2014, miR-21 and miR-375 microRNAs as candidate diagnostic biomarkers in squamous cell carcinoma of the larynx: association with patient survival. Am J Transl Res 6(5):604–613 Ponting CP, Oliver PL, Reik W, 2009, Evolution and functions of long noncoding RNAs. Cell 136(4):629–641., DOI 10.1016/j.cell. 2009.02.006 Kaneko S, Li G, Son J, Xu CF, Margueron R, Neubert TA, Reinberg D, 2010, Phosphorylation of the PRC2 component Ezh2 is cell cycle-regulated and up-regulates its binding to ncRNA. Genes Dev 24(23):2615–2620., DOI 10.1101/gad.1983810 Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, Shi Y, Segal E, Chang HY, 2010, Long noncoding RNA as modular scaffold of histone modification complexes. Science 329(5992):689–693., DOI 10.1126/science.1192002 Wilusz JE, Sunwoo H, Spector DL, 2009, Long noncoding RNAs: functional surprises from the RNAworld. Genes Dev 23(13):1494– 1504., DOI 10.1101/gad.1800909 WangX, Song X, Glass CK, RosenfeldMG(2011, The long arm of long noncoding RNAs: roles as sensors regulating gene transcriptional programs. Cold Spring Harb Perspect Biol 3(1):a003756., DOI 10.1101/cshperspect.a003756 Ji Q, Zhang L, Liu X, Zhou L, Wang W, Han Z, Sui H, Tang Y, Wang Y, Liu N, Ren J, Hou F, Li Q, 2014, Long non-coding RNA MALAT1 promotes tumour growth and metastasis in colorectal cancer through binding to SFPQ and releasing oncogene PTBP2 from SFPQ/PTBP2 complex. Br J Cancer 111(4):736–748., DOI 10. 1038/bjc.2014.383 Shen L, Chen L, Wang Y, Jiang X, Xia H, Zhuang Z, 2014, Long noncoding RNA MALAT1 promotes brain metastasis by inducing epithelial-mesenchymal transition in lung cancer. J Neurooncol., DOI 10.1007/s11060-014-1613-0 Cai B, Wu Z, Liao K, Zhang S, 2014, Long noncoding RNA HOTAIR can serve as a common molecular marker for lymph node metastasis: ameta-analysis. Tumour Biol 35(9):8445–8450., DOI 10. 1007/s13277-014-2311-4 Akhbari P, Whitehouse A, Boyne JR, 2014, Long noncoding RNAs drive metastatic progression in melanoma, Review). Int J Oncol 45(6):2181–2186., DOI 10.3892/ijo.2014.2691 Ji P, Diederichs S, Wang W, Boing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, Thomas M, Berdel WE, Serve H, Muller-Tidow C, 2003, MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene 22(39):8031– 8041., DOI 10.1038/sj.onc.1206928 Liu JH, Chen G, Dang YW, Li CJ, Luo DZ, 2014, Expression and prognostic significance of lncRNA MALAT1 in pancreatic cancer tissues. Asian Pac J Cancer Prev 15(7):2971–2977 Xu C, YangM, Tian J,Wang X, Li Z, 2011)MALAT-1: a long noncoding RNA and its important 3′ end functional motif in colorectal cancer metastasis. Int J Oncol 39(1):169–175., DOI 10.3892/ijo. 2011.1007 Tano K, Akimitsu N, 2012, Long non-coding RNAs in cancer progression. Front Genet 3:219., DOI 10.3389/fgene.2012.00219 Eissmann M, Gutschner T, Hammerle M, Gunther S, Caudron- Herger M, Gross M, Schirmacher P, Rippe K, Braun T, Zornig M, Diederichs S, 2012, Loss of the abundant nuclear non-coding RNA MALAT1 is compatible with life and development. RNA Biol 9(8): 1076–1087., DOI 10.4161/rna.21089 Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR, 2007, Practical methods for incorporating summary time-to-event data into meta-analysis. Trials 8:16., DOI 10.1186/1745-6215-8-16 21. Zheng HT, Shi DB, Wang YW, Li XX, Xu Y, Tripathi P, Gu WL, Cai GX, Cai SJ, 2014, High expression of lncRNA MALAT1 suggests a biomarker of poor prognosis in colorectal cancer. Int J Clin Exp Pathol 7(6):3174–3181 22. Zhang HM, Yang FQ, Chen SJ, Che J, Zheng JH, 2014, Upregulation of long non-coding RNA MALAT1 correlates with tumor progression and poor prognosis in clear cell renal cell carcinoma. Tumour Biol., DOI 10.1007/s13277-014-2925-6 23. Pang EJ, Yang R, Fu XB, Liu YF, 2014, Overexpression of long non-coding RNA MALAT1 is correlated with clinical progression and unfavorable prognosis in pancreatic cancer. Tumour Biol., DOI 10.1007/s13277-014-2850-8 24. Okugawa Y, Toiyama Y, Hur K, Toden S, Saigusa S, Tanaka K, Inoue Y, Mohri Y, Kusunoki M, Boland CR, Goel A, 2014, Metastasis-associated long non-coding RNA drives gastric cancer development and promotes peritoneal metastasis. Carcinogenesis., DOI 10.1093/carcin/bgu200 25. Dong Y, Liang G, Yuan B, Yang C, Gao R, Zhou X, 2014)MALA T1 promotes the proliferation and metastasis of osteosarcoma cells by activating the PI3K/Akt pathway. Tumour Biol., DOI 10.1007/ s13277-014-2631-4 26. Schmidt LH, Spieker T, Koschmieder S, Schaffers S, Humberg J, Jungen D, Bulk E, Hascher A, Wittmer D, Marra A, Hillejan L, Wiebe K, Berdel WE, Wiewrodt R, Muller-Tidow C, 2011, The long noncoding MALAT-1 RNA indicates a poor prognosis in nonsmall cell lung cancer and induces migration and tumor growth. J Thorac Oncol 6(12):1984–1992. doi :10.1097/JTO. 0b013e3182307eac 27. Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q,Watt AT, Freier SM, Bennett CF, Sharma A, Bubulya PA, Blencowe BJ, Prasanth SG, Prasanth KV, 2010, The nuclear-retained noncoding RNA MALA T1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol Cell 39(6):925–938., DOI 10.1016/j.molcel. 2010.08.011 28. Yang L, Lin C, LiuW, Zhang J, Ohgi KA, Grinstein JD, Dorrestein PC, RosenfeldMG(2011, ncRNA- and Pc2 methylation-dependent gene relocation between nuclear structuresmediates gene activation programs. Cell 147(4):773–788., DOI 10.1016/j.cell.2011.08.054 29. Gutschner T, Hammerle M, Eissmann M, Hsu J, Kim Y, Hung G, Revenko A, Arun G, Stentrup M, Gross M, Zornig M, MacLeod AR, Spector DL, Diederichs S, 2013, The noncoding RNA MALA T1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res 73(3):1180–1189., DOI 10.1158/0008-5472.can- 12-2850 30. Hellerbrand C, Amann T, Schlegel J, Wild P, Bataille F, Spruss T, Hartmann A, Bosserhoff AK, 2008, The novel geneMIA2 acts as a tumour suppressor in hepatocellular carcinoma. Gut 57(2):243– 251., DOI 10.1136/gut.2007.129544 31. Xu Y, Li WL, Fu L, Gu F, Ma YJ, 2010, Slit2/Robo1 signaling in glioma migration and invasion. Neurosci Bull 26(6):474–478 32. Legg JA, Herbert JM, Clissold P, Bicknell R, 2008, Slits and Roundabouts in cancer, tumour angiogenesis and endothelial cell migration. Angiogenesis 11(1):13–21., DOI 10.1007/s10456-008- 9100-x 33. Yiu GK, Kaunisto A, Chin YR, Toker A, 2011, NFAT promotes carcinoma invasive migration through glypican-6. Biochem J 440(1):157–166., DOI 10.1042/bj20110530 34. Sekine Y, Demosky SJ, Stonik JA, Furuya Y, Koike H, Suzuki K, Remaley AT, 2010, High-density lipoprotein induces proliferation and migration of human prostate androgen-independent cancer cells by an ABCA1-dependent mechanism. Mol Cancer Res 8(9):1284– 1294., DOI 10.1158/1541-7786.mcr-10-0008 35. Toh B, Wang X, Keeble J, Sim WJ, Khoo K, Wong WC, Kato M, Prevost-Blondel A, Thiery JP, Abastado JP, 2011, Mesenchymal transition and dissemination of cancer cells is driven by myeloidderived suppressor cells infiltrating the primary tumor. PLoS Biol 9(9):e1001162., DOI 10.1371/journal.pbio.1001162 36. Gutschner T, Hammerle M, Diederichs S, 2013, MALAT1—a paradigm for long noncoding RNA function in cancer. J Mol Med, Berl, 91(7):791–801., DOI 10.1007/s00109-013-1028-y 37. Wu XS,Wang XA,WuWG, Hu YP, LiML, Ding Q,Weng H, Shu YJ, Liu TY, Jiang L, Cao Y, Bao RF, Mu JS, Tan ZJ, Tao F, Liu YB, 2014, MALAT1 promotes the proliferation and metastasis of gallbladder cancer cells by activating the ERK/MAPKpathway. Cancer Biol Ther 15(6):806–814., DOI 10.4161/cbt.28584 38. LiN,Wang HX, Zhang J, Ye YP, HeGY, 2012)KISS-1 inhibits the proliferation and invasion of gastric carcinoma cells. World J Gastroenterol 18(15):1827–1833., DOI 10.3748/wjg.v18.i15.1827 39. Hong SK, Yoon S, Moelling C, Arthan D, Park JI, 2009, Noncatalytic function of ERK1/2 can promote Raf/MEK/ERK-mediated growth arrest signaling. J Biol Chem 284(48):33006–33018., DOI 10.1074/jbc.M109.012591 40. Xander P, Brito RR, Perez EC, Pozzibon JM, de Souza CF, Pellegrino R, Bernardo V, Jasiulionis MG, Mariano M, Lopes JD, 2013, Crosstalk between B16 melanoma cells and B-1 lymphocytes induces global changes in tumor cell gene expression. Immunobiology 218(10):1293–1303., DOI 10.1016/j.imbio.2013. 04.017 41. Fan Y, Shen B, Tan M, Mu X, Qin Y, Zhang F, Liu Y, 2014, TGFbeta- induced upregulation of malat1 promotes bladder cancer metastasis by associating with suz12. Clin Cancer Res 20(6):1531– 1541., DOI 10.1158/1078-0432.ccr-13-1455 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1259 EP 1264 DI 10.1007/s12253-015-9960-5 PG 6 ER PT J AU Fabian, M Toth, V Somlai, B Harsing, J Kuroli, E Rencz, F Kuzmanovszki, D Szakonyi, J Toth, B Karpati, S AF Fabian, Melinda Toth, Veronika Somlai, Beata Harsing, Judit Kuroli, Eniko Rencz, Fanni Kuzmanovszki, Daniella Szakonyi, Jozsef Toth, Bela Karpati, Sarolta TI Retrospective Analysis of Clinicopathological Characteristics of Pregnancy Associated Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Melanoma; Pregnancy; Postpartum; Young women ID Melanoma; Pregnancy; Postpartum; Young women AB Pregnancy associated melanoma (PAM) by definition appears during pregnancy or within 1 year after delivery. In this retrospective study we analysed the pathological characteristics and survival rate of PAM and matched the data with non-pregnant age- and stage-matched control patients. Between 2003 and 2014, 34 pregnant women (aged 32.5±5.6 years) were diagnosed with melanoma at the Department of Dermatology, Venereology and Dermatooncology of the Semmelweis University. During the pathological process histologic subtype, Breslow thickness and Clark level, tumor cell type, mitotic rate, peritumoral inflammation, as well as ulceration, regression, necrosis, vascular invasion and presence of satellitewere analyzed and related to clinical data. Primary tumor location and clinical staging, disease course, local recurrence and metastases, 5-year survival rate, other tumor development before or after the diagnosis of melanoma have also been documented. We found no difference in all parameters between pregnant and non-pregnant melanoma cases except peritumoral inflammation which was higher in PAM group, moreover the presence of mild inflammation was significantly higher in PAM group compared to non-pregnancy associated melanoma (NPAM) women group. C1 [Fabian, Melinda] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Toth, Veronika] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Somlai, Beata] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Harsing, Judit] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Kuroli, Eniko] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Rencz, Fanni] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Kuzmanovszki, Daniella] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Szakonyi, Jozsef] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Toth, Bela] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. [Karpati, Sarolta] Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Maria street 41, 1085 Budapest, Hungary. RP Karpati, S (reprint author), Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, 1085 Budapest, Hungary. EM karpatsar@gmail.com CR Tomao F, Papa A, Lo Russo G, Zuber S, Spinelli GP, Rossi L, Caruso D, Prinzi N, Stati V, Benedetti Panici P, Tomao S, 2014, Correlation between fertility drugs use and malignant melanoma incidence: the state of the art. Tumour Biol 35(9):8415–8424 Driscoll MS, Grant-Kels JM, 2007, Hormones, nevi, and melanoma: an approach to the patient. J Am Acad Dermatol 57(6):919– 931, quiz 932–6 Smith LH, Dalrymple JL, Leiserowitz GS, Danielsen B, Gilbert WM, 2001, Obstetrical deliveries associated with maternal malignancy in California, 1992 through 1997. Am J Obstet Gynecol 184(7):1504–1512, discussion 1512–3 Zick CD, Smith KR, Mayer RN, Taylor LB, 2014, Family, frailty, and fatal futures? Own-health and family-health predictors of subjective life expectancy. Res Aging 36(2):244–266 Pentheroudakis G, Orecchia R, Hoekstra HJ, Pavlidis N, Group EGW, 2010, Cancer, fertility and pregnancy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 21(Suppl 5):v266–v273 Andersson TM, Johansson AL, Fredriksson I, Lambe M, 2015, Cancer during pregnancy and the postpartum period: a population-based study. Cancer Zhou JH, Kim KB, Myers JN, Fox PS, Ning J, Bassett RL, Hasanein H, Prieto VG, 2014, Immunohistochemical expression of hormone receptors in melanoma of pregnant women, nonpregnant women, and men. Am J Dermatopathol 36(1):74–79 Stensheim H, Moller B, van Dijk T, Fossa SD, 2009, Causespecific survival for women diagnosed with cancer during pregnancy or lactation: a registry-based cohort study. J Clin Oncol 27(1): 45–51 Eibye S, Kjaer SK, Mellemkjaer L, 2013, Incidence of pregnancyassociated cancer in Denmark, 1977–2006. Obstet Gynecol 122(3): 608–617 Lens MB, Rosdahl I, Ahlbom A, Farahmand BY, Synnerstad I, Boeryd B, Newton Bishop JA, 2004, Effect of pregnancy on survival in women with cutaneous malignant melanoma. J Clin Oncol 22(21):4369–4375 Frishberg DP, Balch C, Balzer BL, Crowson AN, Didolkar M, McNiff JM, Perry RR, Prieto VG, Rao P, Smith MT, Smoller BR, Wick MR, C.o.A.P. Members of the Cancer Committee, 2009, Protocol for the examination of specimens from patients with melanoma of the skin. Arch Pathol Lab Med 133(10):1560–1567 Clayton SG, 1946, Melanoma of the vulva with pregnancy. Proc R Soc Med 39(9):578–579 Moller H, Purushotham A, Linklater KM, Garmo H, Holmberg L, Lambe M, Yallop D, Devereux S, 2013, Recent childbirth is an adverse prognostic factor in breast cancer and melanoma, but not in Hodgkin lymphoma. Eur J Cancer 49(17):3686–3693 Scoggins CR, Ross MI, Reintgen DS, Noyes RD, Goydos JS, Beitsch PD, Urist MM, Ariyan S, Sussman JJ, Edwards MJ, Chagpar AB, Martin RC, Stromberg AJ, Hagendoorn L, McMasters KM, Sunbelt Melanoma T, 2006, Gender-related differences in outcome for melanoma patients. Ann Surg 243(5):693– 698, discussion 698–700 Sanlorenzo M, Ribero S, Osella-Abate S, Zugna D, Marenco F, Macripo G, Fierro MT, Bernengo MG, Quaglino P, 2014, Prognostic differences across sexes in melanoma patients: what has changed from the past? Melanoma Res 24(6):568–576 Gamba CS, Clarke CA, Keegan TH, Tao L, Swetter SM, 2013, Melanoma survival disadvantage in young, non-Hispanic white males compared with females. JAMA Dermatol 149(8):912–920 Nosrati A, Wei ML, 2014, Sex disparities in melanoma outcomes: the role of biology. Arch Biochem Biophys 563C:42–50 Joosse A, deVries E, Eckel R,Nijsten T, EggermontAM, Holzel D, Coebergh JW, Engel J, MunichMelanoma G, 2011, Gender differences in melanoma survival: female patients have a decreased risk of metastasis. J Invest Dermatol 131(3):719–726 Kanda N, Watanabe S, 2001, 17beta-estradiol, progesterone, and dihydrotestosterone suppress the growth of human melanoma by inhibiting interleukin-8 production. J Invest Dermatol 117(2):274– 283 O’Meara AT, Cress R, Xing G, Danielsen B, Smith LH, 2005, Malignant melanoma in pregnancy. A population-based evaluation. Cancer 103(6):1217–1226 Daryanani D, Plukker JT, De Hullu JA, Kuiper H, Nap RE, Hoekstra HJ, 2003, Pregnancy and early-stage melanoma. Cancer 97(9):2248–2253 Johansson AL, Andersson TM, Plym A, Ullenhag GJ, Moller H, Lambe M, 2014, Mortality in women with pregnancy-associated malignant melanoma. J Am Acad Dermatol 71(6):1093–1101 Byrom L, Olsen C, Knight L, Khosrotehrani K, Green AC, 2015, Increased mortality for pregnancy-associated melanoma: systematic review and meta-analysis. J Eur Acad Dermatol Venereol Travers RL, Sober AJ, Berwick M, Mihm MC Jr, Barnhill RL, Duncan LM, 1995, Increased thickness of pregnancy-associated melanoma. Br J Dermatol 132(6):876–883 Massi D, Franchi A, Borgognoni L, Reali UM, Santucci M, 1999, Thin cutaneous malignant melanomas, < or =1.5 mm): identification of risk factors indicative of progression. Cancer 85(5):1067– 1076 Ladanyi A, Somlai B, Gilde K, Fejos Z, Gaudi I, Timar J, 2004, Tcell activation marker expression on tumor-infiltrating lymphocytes as prognostic factor in cutaneousmalignantmelanoma. Clin Cancer Res 10(2):521–530 Fortes C, Mastroeni S, Manooranparanpampil T, Passarelli F, Zappala A, Marino C, Russo N, Michelozzi P, 2015, Tumorinfiltrating lymphocytes predict cutaneous melanoma survival. Melanoma Res. 25(4)306–11 Toth V, Somlai B, Harsing J, Hatvani Z, Karpati S, 2013, Stage distribution of malignant melanomas in a Hungarian centre. Orv Hetil 154(25):969–976 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1265 EP 1271 DI 10.1007/s12253-015-9961-4 PG 7 ER PT J AU Dong, F Liu, F Yan, S Liu, X Jiang, Z Liu, J AF Dong, Fengyun Liu, Fuhong Yan, Suhua Liu, Xiaochun Jiang, Zhongmin Liu, Ju TI Elevated Expression of CD109 in Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Dong, Fengyun] Qianfoshan Hospital Affiliated to Shandong University, 16766 Jingshi Road, 250014 Jinan, Shandong, China. [Liu, Fuhong] Qianfoshan Hospital Affiliated to Shandong University, 16766 Jingshi Road, 250014 Jinan, Shandong, China. [Yan, Suhua] Qianfoshan Hospital Affiliated to Shandong University, 16766 Jingshi Road, 250014 Jinan, Shandong, China. [Liu, Xiaochun] The University of Texas, MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, 1515 Holcombe Blvd, 77030 Houston, TX, USA. [Jiang, Zhongmin] Qianfoshan Hospital Affiliated to Shandong University, 16766 Jingshi Road, 250014 Jinan, Shandong, China. [Liu, Ju] Qianfoshan Hospital Affiliated to Shandong University, 16766 Jingshi Road, 250014 Jinan, Shandong, China. RP Jiang, Z (reprint author), Qianfoshan Hospital Affiliated to Shandong University, 250014 Jinan, China. EM qyjzm@sina.com.cn CR Stoner GD, Wang LS, Chen T, 2007, Chemoprevention of esophageal squamous cell carcinoma. Toxicol Appl Pharmacol 224(3):337– 349., DOI 10.1016/j.taap.2007.01.030 Osawa H, Nakajima M, Kato H, Fukuchi M, Kuwano H, 2004, Prognostic value of the expression of Smad6 and Smad7, as inhibitory Smads of the TGF-beta superfamily, in esophageal squamous cell carcinoma. Anticancer Res 24(6):3703–3709 Megumi K, Ishigami S, UchikadoY, Kita Y, Okumura H,Matsumoto M, Uenosono Y, Arigami T, Kijima Y, Kitazono M, Shinchi H, Ueno S, Natsugoe S, 2012, Clinicopathological significance of BMP7 expression in esophageal squamous cell carcinoma. Ann Surg Oncol 19(6):2066–2071., DOI 10.1245/s10434-011-2024-5 Finnson KW, Tam BY, Liu K, Marcoux A, Lepage P, Roy S, Bizet AA, Philip A, 2006, Identification of CD109 as part of the TGF-beta receptor system in human keratinocytes. FASEB J Off Publ Fed Am Soc Exp Biol 20(9):1525–1527., DOI 10. 1096/fj.05-5229fje Hashimoto M, Ichihara M, Watanabe T, Kawai K, Koshikawa K, Yuasa N, Takahashi T, Yatabe Y, Murakumo Y, Zhang JM, Nimura Y, Takahashi M, 2004, Expression of CD109 in human cancer. Oncogene 23(20):3716–3720., DOI 10.1038/sj.onc.1207418 Ozbay PO, Ekinci T, Yigit S, Yavuzcan A, Uysal S, Soylu F, Cakalagaoglu F, 2013, Investigation of prognostic significance of CD109 expression in women with vulvar squamous cell carcinoma. Onco Targets Ther 6:621–627., DOI 10.2147/OTT.S41069 Zhang JM, Hashimoto M, Kawai K, Murakumo Y, Sato T, Ichihara M, Nakamura S, Takahashi M, 2005, CD109 expression in squamous cell carcinoma of the uterine cervix. Pathol Int 55(4):165– 169., DOI 10.1111/j.1440-1827.2005.01807.x Duff B, Weigel JA, Bourne P, Weigel PH, McGary CT, 2002, Endothelium in hepatic cavernous hemangiomas does not express the hyaluronan receptor for endocytosis. Hum Pathol 33(3):265–269 Cuppini L, Calleri A, BruzzoneMG, Prodi E, Anghileri E, Pellegatta S, Mancuso P, Porrati P, Di Stefano AL, Ceroni M, Bertolini F, Finocchiaro G, Eoli M, 2013, Prognostic value of CD109+ circulating endothelial cells in recurrent glioblastomas treated with bevacizumab and irinotecan. PLoS One 8(9):e74345., DOI 10.1371/ journal.pone.0074345 Hagiwara S, Murakumo Y, Sato T, Shigetomi T,Mitsudo K, Tohnai I, UedaM, Takahashi M, 2008, Up-regulation of CD109 expression is associated with carcinogenesis of the squamous epithelium of the oral cavity. Cancer Sci 99(10):1916–1923., DOI 10.1111/j.1349-7006. 2008.00949.x NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1273 EP 1275 DI 10.1007/s12253-014-9894-3 PG 3 ER PT J AU Kim, JCh Shin, WJ Jung, WS Park, RB Park, HN AF Kim, Jae Chang Shin, Woo Jung Jung, Won Seok Park, Ryung Bo Park, Hwa Neung TI Somatic Mutation of ARHI Gene in Hepatocellular Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Kim, Jae Chang] University of Ulsan, College of Medicine, Ulsan University Hospital, Biomedical Research Center, 682-714 Ulsan, South Korea. [Shin, Woo Jung] University of Ulsan, College of Medicine, Ulsan University Hospital, Biomedical Research Center, 682-714 Ulsan, South Korea. [Jung, Won Seok] University of Ulsan, College of Medicine, Ulsan University Hospital, Biomedical Research Center, 682-714 Ulsan, South Korea. [Park, Ryung Bo] University of Ulsan, College of Medicine, Ulsan University Hospital, Biomedical Research Center, 682-714 Ulsan, South Korea. [Park, Hwa Neung] University of Ulsan, College of Medicine, Ulsan University Hospital, Biomedical Research Center, 682-714 Ulsan, South Korea. RP Park, HN (reprint author), University of Ulsan, College of Medicine, Ulsan University Hospital, Biomedical Research Center, 682-714 Ulsan, South Korea. EM airhiro@naver.com CR Ahn J, Flamm SL, 2004, Hepatocellular carcinoma. Dis Mon 50: 556–573 Bosch FX, Ribes J, Diaz M, Cleries R, 2004, Primary liver cancer: worldwide incidence and trends. Gastroenterology 127:S5–S16 Yu Y, Xu F, Peng H, Fang X, Zhao S, Li Y, Cuevas B, KuoWL, Gray JW, Siciliano M, Mills GB, Bast RC Jr, 1999, NOEY2, ARHI), an imprinted putative tumor suppressor gene in ovarian and breast carcinomas. Proc Natl Acad Sci U S A 96:214–219 Luo RZ, Fang X, Marquez R, Liu SY,Mills GB, LiaoWS,Yu Y, Bast RC, 2003, ARHI is a Ras-related small G-protein with a novel Nterminal extension that inhibits growth of ovarian and breast cancers. Oncogene 22:2897–2909 Yu Y, Luo R, Lu Z,Wei FengW, Badgwell D, Issa JP, Rosen DG, Liu J, Bast RC Jr, 2006, Biochemistry and biology of ARHI, DIRAS3), an imprinted tumor suppressor gene whose expression is lost in ovarian and breast cancers. Methods Enzymol 407:455–468 Wang L, Hoque A, Luo RZ, Yuan J, Lu Z, Nishimoto A, Liu J, Sahin AA, Lippman SM, Bast RC Jr, Yu Y, 2003, Loss of the expression of the tumor suppressor gene ARHI is associated with progression of breast cancer. Clin Cancer Res 9:3660–3666 Rosen DG,Wang L, Jain AN, Lu KH, Luo RZ, Yu Y, Liu J, Bast RC Jr, 2004, Expression of the tumor suppressor gene ARHI in epithelial ovarian cancer is associated with increased expression of p21WAF1/ CIP1 and prolonged progression-free survival. Clin Cancer Res 10: 6559–6566 Weber F, Aldred MA, Morrison CD, Plass C, Frilling A, Broelsch CE, Waite KA, Eng C, 2005, Silencing of the maternally imprinted tumor suppressor ARHI contributes to follicular thyroid carcinogenesis. J Clin Endocrinol Metab 90:1149–1155 Hisatomi H, Nagao K, Wakita K, Kohno N, 2002, ARHI/NOEY2 inactivation may be important in breast tumor pathogenesis. Oncology 62:136–140 FengW, Marquez RT, Lu Z, Liu J, Lu KH, Issa JP, Fishman DM, Yu Y, Bast RC Jr, 2008, Imprinted tumor suppressor genes ARHI and PEG3 are the most frequently down-regulated in human ovarian cancers by loss of heterozygosity and promoter methylation. Cancer 112: 1489–1502 Dalai I, Missiaglia E, Barbi S, Butturini G, Doglioni C, Falconi M, Scarpa A, 2007, Low expression of ARHI is associated with shorter progression-free survival in pancreatic endocrine tumors. Neoplasia 9:181–183 Huang J, Lin Y, Li L, Qing D, Teng XM, Zhang YL, Hu X, Hu Y, Yang P, Han ZG, 2009, ARHI, as a novel suppressor of cell growth and downregulated in human hepatocellular carcinoma, could contribute to hepatocarcinogenesis. Mol Carcinog 48:130–140 Yang J, Hu A, Wang L, Li B, Chen Y, ZhaoW, XuW, Li T, 2009, NOEY2 mutations in primary breast cancers and breast hyperplasia. Breast 18:197–203 Janssen EA, Ovestad IT, Skaland I, Soiland H, Gudlaugsson E, Kjellevold KH, Nysted A, Soreide JA, Baak JP, 2009, LOH at 1p31, ARHI, and proliferation in lymph node-negative breast cancer. Cell Oncol 31:335–343 Yu Y, Fujii S, Yuan J, Luo RZ,Wang L, Bao J, KadotaM, Oshimura M, Dent SR, Issa JP, Bast RC Jr, 2003, Epigenetic regulation of ARHI in breast and ovarian cancer cells. Ann N Y Acad Sci 983: 268–277 Pei XH, Yang Z, Liu HX, Qiao SS, 2011, Aplasia Ras homologue member I overexpression induces apoptosis through inhibition of survival pathways in human hepatocellular carcinoma cells in culture and in xenograft. Cell Biol Int 35:1019–1024 Zhao X, Li J, Zhuo J, Cai L, 2010, Reexpression of ARHI inhibits tumor growth and angiogenesis and impairs the mTOR/VEGF pathway in hepatocellular carcinoma. Biochem Biophys Res Commun 403:417–421 Park WS, Cho YG, Kim CJ, Song JH, Lee YS, Kim SY, Nam SW, Lee SH, Yoo NJ, Lee JY, 2005, Hypermethylation of the RUNX3 gene in hepatocellular carcinoma. Exp Mol Med 37:276–281 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1277 EP 1279 DI 10.1007/s12253-015-9924-9 PG 3 ER PT J AU Jo, SY Kim, SM Lee, HS Yoo, JN AF Jo, Sol Yun Kim, Sung Min Lee, Hyung Sug Yoo, Jin Nam TI Mutational Heterogeneity of MED23 Gene in Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Yoo, JN (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM goldfish@catholic.ac.kr CR Poss ZC, Ebmeier CC, Taatjes DJ, 2013, TheMediator complex and transcription regulation. Crit Rev Biochem Mol Biol 48:575–608 Makinen N, Mehine M, Tolvanen J, Kaasinen E, Li Y, Lehtonen HJ, Gentile M, Yan J, Enge M, Taipale M, Aavikko M, Katainen R, Virolainen E, Bohling T, Koski TA, Launonen V, Sjoberg J, Taipale J, Vahteristo P, Aaltonen LA, 2011, MED12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas. Science 334:252–255 LimWK, Ong CK, Tan J, Thike AA, Ng CC, Rajasegaran V, Myint SS, Nagarajan S, Nasir ND,McPherson JR, Cutcutache I, Poore G, Tay ST, Ooi WS, Tan VK, Hartman M, Ong KW, Tan BK, Rozen SG, Tan PH, Tan P, Teh BT, 2014, Exome sequencing identifies highly recurrentMED12 somatic mutations in breast fibroadenoma. Nat Genet 46:877–880 Yang X, Zhao M, Xia M, Liu Y, Yan J, Ji H, Wang G, 2012, Selective requirement forMediator MED23 in Ras-active lung cancer. Proc Natl Acad Sci U S A 109:E2813–E2822 Guo Y,Wang J, Li H, Liu W, Chen D, Zhao K, Liang X, Zhang Q, Yang Y, Chen G, 2015, MED23 overexpression as a novel target for suppressing proliferation and tumorigenesis in hepatocellular carcinoma. J Gastroenterol Hepatol 30:1094–1103 Shi J, Han Q, Zhao H, Zhong C, Yao F, 2014, Downregulation of MED23 promoted the tumorigenecity of esophageal squamous cell carcinoma. Mol Carcinog 53:833–840 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Calin GA, Gafa R, Tibiletti MG, Herlea V, Becheanu G, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression in microsatellite instability high, MSI-H, colon cancers correlates with clinicopathological parameters: a study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89: 230–235 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2015 VL 21 IS 4 BP 1281 EP 1282 DI 10.1007/s12253-015-9959-y PG 2 ER PT J AU Halasz, T Horvath, G Kiss, A Par, G Szombati, A Gelley, F Nemes, B Kenessey, I Piurko, V Schaff, Zs AF Halasz, Tunde Horvath, Gabor Kiss, Andras Par, Gabriella Szombati, Andrea Gelley, Fanni Nemes, Balazs Kenessey, Istvan Piurko, Violetta Schaff, Zsuzsa TI Evaluation of Histological and non-Invasive Methods for the Detection of Liver Fibrosis: The Values of Histological and Digital Morphometric Analysis, Liver Stiffness Measurement and APRI Score SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Liver fibrosis; Liver stiffness; Digital morphometric analysis; Liver biopsy ID Liver fibrosis; Liver stiffness; Digital morphometric analysis; Liver biopsy AB Prognosis and treatment of liver diseases mainly depend on the precise evaluation of the fibrosis. Comparisons were made between the results of Metavir fibrosis scores and digital morphometric analyses (DMA), liver stiffness (LS) values and aminotransferase-platelet ratio (APRI) scores, respectively. Liver biopsy specimens stained with Sirius red and analysed by morphometry, LS and APRI measurements were taken from 96 patients with chronic liver diseases (56 cases of viral hepatitis, 22 cases of autoimmune- and 18 of mixed origin). The strongest correlation was observed between Metavir score and DMA (r=0.75 p<0.05), followed in decreasing order by LS and Metavir (r=0.61), LS and DMA (r=0.47) LS and APRI (r=0.35) and Metavir and APRI (r= 0.24), respectively. DMA is a helpful additional tool for the histopathological evaluation of fibrosis, even when the sample size is small and especially in case of advanced fibrosis. The non-invasive methods showed good correlation with the histopathological methods; LS proved to be more accurate than APRI. The stronger correlation between LS values and Metavir scores, as well as the results of DMA in case of appropriate sample size were remarkable. C1 [Halasz, Tunde] Semmelweis University, 2nd Department of Pathology, Ulloi str 93, 1091 Budapest, Hungary. [Horvath, Gabor] Hepatology Center of BudaBudapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi str 93, 1091 Budapest, Hungary. [Par, Gabriella] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Szombati, Andrea] Del-Pesti CentrumkorhazBudapest, Hungary. [Gelley, Fanni] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Nemes, Balazs] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Kenessey, Istvan] Semmelweis University, 2nd Department of Pathology, Ulloi str 93, 1091 Budapest, Hungary. [Piurko, Violetta] Semmelweis University, 2nd Department of Pathology, Ulloi str 93, 1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi str 93, 1091 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM schaff.zsuzsa@med.semmelweis-univ.hu CR Bedossa P, Dargere D, Paradis V, 2003, Sampling variability of liver fibrosis in chronic hepatitis C. Hepatology 38:1449–1457 Afdhal NH, 2004, Biopsy or biomarkers: is there a gold standard for diagnosis of liver fibrosis? Clin Chem 50:1299–1300 Desmet VJ, Gerber M, Hoofnagle JH et al, 1994, Classification of chronic hepatitis: diagnosis, grading and staging. Hepatology 19: 1513–1520 Germani G, Burroughs AK, Duillon AP, 2010, The relationship between liver disease stage and liver fibrosis: a tangled web. Histopathology 57:773–784 Huang Y, de Boer WB, Adams LA et al, 2014, Image analysis of liver biopsy samples measures fibrosis and predicts clinical outcome. J Hepatol 61:22–27 Hall AR, Tsochatzis E, Morris R et al, 2013, Sample size requirement for digital image analysis of collagen prportionate area in cirrhotic livers. Histopathology 62:421–430 Isgro G, Calvaruso V, Andreana L et al, 2013, The relationship between transient elastograpy and histological collagen proportionate area for assessing fibrosis in chronic viral hepatitis. J Gastroenterol 48:921–929 Huang Y, de Boer WB, Adams LA et al, 2013, Image analysis of liver collagen using sirius red is more accurate and correlates better with serum fibrosis markers than trichrome. Liver Int 338:1249– 1256 Poynard T, Bedossa P, Opolon P, 1997, Natural history of liver fibrosis progression in patient with chronic hepatitis C. The OBSVIRC, METAVIR, CLINVIR and DOSVIRCgroups. Lancet 349:825–832 Corpechot C, El Naggar A, Poujol-Robert A et al, 2006, Assessment of biliary fibrosis by transient elastography in patients with PBC and PSC. Hepatology 43:1118–1124 Brunt M, Janney CG, Di Bisceglie AM et al, 1999, Nonacoholic steatohepatitis: a proposal for grading and staging the histological lesions. Am J Gastroenterol 94:2467–2474 Ishak K, Baptista L, Bianchi L et al, 1995, Histological grading and staging of chronic hepatitis. J Hepatol 22:696–699 Wai CT, Greenson JK, Fontana RJ, 2003, A simple noninvasive index can predict both significant fibrosis and cirrhosis in patient with chronic hepatitis C. Hepatology 38:518–526 Dahab GM, Kheriza MM, El-Beltagi HM et al, 2004, Digital quatification of fibrosis in liver biopsy sections: description of a new method by photoshop software. J Gastroenterol Hepatol 19: 78–85 Goodman ZD, Stoddard AM, Bonkowsky HL et al, 2009, Fibrosis progression in chronic hepatitis C: morphometric image analysis in the HALT-C trial. Hepatology 50:1738–1749 Huss S, Schmitz J, Goltz D et al, 2010, Development and evaluation of an open source Delphi-based software for morphometric quantification of liver fibrosis. Fibrogenesis Tissue Repair 3:10 Wright M, Thursz M, Pullen R et al, 2003, Quantitative versus morphological assessment of liver fibrosis: semi-quantitative scores aremore robust than digital image fibrosis area estimation. Liver Int 23:28–34 Maduli E, Andorno S, Rigamonti C et al, 2002, Evaluation of liver fibrosis in chronic hepatits C with a computer-assisted morphometric method. Ann Ital Med Int 17:242–247 Arima M, Terao H, Kashima K et al, 2004, Regression of liver fibrosis in cases of chronic liver disease type C: quantitative evaluation by using computed image analysis. Intern Med 43:902–910 FriedenbergMA,Miller L, Chung CYet al, 2005, Simplified method of hepatic fibrosis quantification: design of a new morphometric analysis application. Liver Int 25:1156–1161 O’BrienMJ, KeatingNM, Elderiny S et al, 2000, An assessment of digital image analysis to measure fibrosis in liver biopsy specimens of patients with chronic hepatitis C. Am J Clin Pathol 114:712–718 Pilette C, Rousselet C, Bedossa P et al, 1998, Histopathological evaluation of liver fibrosis: quantitative image analysis vs semiquantitative scores. Comparison with serum markers J Hepatol 28:439–446 Ziol M, Kettaneh A, Ganne-Carrie N et al, 2009, Relationships between fibrosis amounts assessed by morphometry and liver stiffness measurements in chronic hepatitis or steatohepatitis. Eur J Gastroenterol Hepatol 21:1261–1268 Lazzarini AL, Levine RA, Ploutz-Synder J et al, 2005, Advances in digital quantification technique enhance discrimination between mild and advanced liver fibrosis in chronic hepatitis C. Liver Int 25:1142–1149 Cho HJ, Seo YS, Lee KG et al, 2011, Serum aminotransferase levels instead of etiology affects the accuracy of transient elastography in chronic viral hepatitis patient. Gastroenterol Hepatol 26:492–500 Yasuda M, Shimizu I, Shiba M et al, 1999, Supressive efects of estradiol on dimetylnitrosamine-induced fibrosis of the liver in rats. Hepatology 29:719–727 Fraquelli M, Rigamonti C, Casazza G et al, 2007, Reproducibility of transient elastograpy in the evaluation of liver fibrosis in patient with chronic liver disease. Gut 56:968–973 Arena U, Vizutti F, Abraldes JG et al, 2008, Reliability of transient elastography for the diagnosis of advanced fibrosis in chronic hepatits C. Gut 57:1288–1293 Tapper EB, Cohen EB, Afdhal N et al, 2012, Levels of alanine aminotransferase confound use of transient elastography to diagnose fibrosis in patients with chronic HCV infection. Clin Gastroenterol Hepatol 10:932–937 McCormick SE, Goodman ZD, Maydonovitch CC et al, 1996, Evaluation of liver histology ALT elevation and HCV RNA titer in patients with chronic hepatitis C. Am J Gastroenterol 91:1516–1522 Poynard T, Ngo Y, Perazzo H et al, 2011, Prognostic value of liver fibrosis biomarkers. Ameta-analysis. Gastroenterol Hepatol 7:445– 454 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 1 EP 6 DI 10.1007/s12253-015-9964-1 PG 6 ER PT J AU Zhou, L Li, J Shao, QQ Guo, JCh Liang, ZY Zhou, WX Zhang, TP You, L Zhao, YP AF Zhou, Li Li, Jian Shao, Qian-Qian Guo, Jun-Chao Liang, Zhi-Yong Zhou, Wei-Xun Zhang, Tai-Ping You, Lei Zhao, Yu-Pei TI Expression and Significances of MTSS1 in Pancreatic Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Metastasis suppressor 1; Pancreatic cancer; Lymph node metastasis; Prognosis ID Metastasis suppressor 1; Pancreatic cancer; Lymph node metastasis; Prognosis AB Thus far, expression of metastasis suppressor 1 (MTSS1), its clinicopathologic and prognostic significances in pancreatic cancer (PC) remain unknown. Expression of MTSS1 was detected by Western blotting in PC cell lines, and by tissue microarray-based immunohistochemical staining in paired tumor and non-tumor samples from 242 patients with PC. Furthermore, the correlations between MTSS1 expression and clinicopathologic variables as well as overall survival were evaluated. In PC cell lines, MTSS1 was differentially expressed. In addition, MTSS1 expression was significantly lower in tumor than in non-tumor tissues (P<0.001 in both McNemar and Mann–Whitney U tests). High tumoral expression of MTSS1 was closely associated with absence of lymph node metastasis (P=0.023). Univariate analysis found that high MTSS1 expression in tumor tissues was a strong predictor of favorable overall survival in the whole cohort (P<0.001). Besides, its impacts on prognosis were also observed in nine out of fourteen subgroups. Finally, MTSS1 expression was identified as an independent prognostic marker in the whole cohort (P=0.031) as well as in six subgroups (P<0.05), as shown by multivariate Cox regression test. Down-regulation of MTSS1 expression is evident in PC, and is associated with lymph node metastasis and poor prognosis. C1 [Zhou, Li] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. [Li, Jian] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. [Shao, Qian-Qian] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. [Guo, Jun-Chao] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. [Liang, Zhi-Yong] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of Pathology, 100730 Beijing, China. [Zhou, Wei-Xun] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of Pathology, 100730 Beijing, China. [Zhang, Tai-Ping] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. [You, Lei] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. [Zhao, Yu-Pei] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. RP Guo, JCh (reprint author), Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. EM gjcpumch@163.com CR HidalgoM(2010, Pancreatic cancer. N Engl JMed 362:1605–1617 Egawa S, Toma H, Ohigashi H, Okusaka T, Nakao A, Hatori T, Maguchi H, Yanagisawa A, Tanaka M, 2012, Japan pancreatic cancer registry; 30th year anniversary: Japan pancreas society. Pancreas 41:985–992 Lee SR, Kim HO, Son BH, Yoo CH, Shin JH, 2013, Prognostic factors associated with long-term survival and recurrence in pancreatic adenocarcinoma. Hepatogastroenterology 60:358–362 Pongprasobchai S, Pannala R, Smyrk TC, BamletW, Pitchumoni S, Ougolkov A, de Andrade M, Petersen GM, Chari ST, 2008, Longterm survival and prognostic indicators in small, 25 % of cases. The E3 ubiquitin ligase NEDD4 (also known as NEDD4-1) has been reported to negatively regulate PTEN protein levels through poly-ubiquitination and proteolysis in carcinomas of the prostate, lung, and bladder, but its effect on PTEN in the breast has not been studied extensively. To investigate whether NEDD4 contributes to low PTEN levels in human breast cancer, we analyzed the expression of these proteins by immunohistochemistry across a large Swedish cohort of breast tumor specimens, and their transcript expression levels by microarrays. For both NEDD4 and PTEN, their transcript expression was significantly correlated to their protein expression. However, comparing NEDD4 expression to PTEN expression, either no association or a positive correlation was observed at the protein and transcript levels. This unexpected observation was further corroborated in two independent breast cancer cohorts from The Netherlands Cancer Institute and The Cancer Genome Atlas. Our results suggest that NEDD4 is not responsible for the frequent down-regulation of the PTEN protein in human breast carcinoma. C1 [Chen, Yilun] Lund University, Department of Clinical Sciences, Division of Oncology and PathologyLund, Sweden. [van de Vijver, J Marc] Academic Medical Center, Department of PathologyAmsterdam, The Netherlands. [Hibshoosh, Hanina] Columbia University Medical CenterNew York, NY, USA. [Parsons, Ramon] Icahn School of Medicine at Mount Sinai, Department of Oncological SciencesNew York, NY, USA. [Saal, H Lao] Lund University, Department of Clinical Sciences, Division of Oncology and PathologyLund, Sweden. RP Saal, HL (reprint author), Lund University, Department of Clinical Sciences, Division of Oncology and Pathology, Lund, Sweden. EM lao.saal@med.lu.se CR Maehama T, Dixon JE, 1998, The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J Biolumin Chemilumin 273:13375–13378 Arch EM, Goodman BK, Van Wesep RA, Liaw D, Clarke K, Parsons R, et al, 1997, Deletion of PTEN in a patient with bannayan-riley-ruvalcaba syndrome suggests allelism with cowden disease. Am J Med Genet 71:489–493 Liaw D, Marsh DJ, Li J, Dahia PL,Wang SI, Zheng Z, et al, 1997, Germline mutations of the PTEN gene in cowden disease, an inherited breast and thyroid cancer syndrome. Nat Genet 16:64–67 Hobert JA, Eng C, 2009, PTEN hamartoma tumor syndrome: an overview. Genitourin Med 11:687–694 Li J, Yen C, Liaw D, Podsypanina K, Bose S,Wang SI, et al, 1997, PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 275:1943–1947 Steck PA, PershouseMA, Jasser SA, Yung WK, Lin H, Ligon AH, et al, 1997, Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet 15:356–362 Shaw RJ, Cantley LC, 2006, Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature 441:424–430 Forbes SA, Beare D, Gunasekaran P, Leung K, Bindal N, Boutselakis H, et al, 2015, COSMIC: exploring the world’s knowledge of somatic mutations in human cancer. Nucleic Acids Res 43: D805–811 Saal LH, Holm K,Maurer M, Memeo L, Su T,Wang X, et al, 2005, PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma. Cancer Res 65:2554–2559 The Cancer Genome Atlas Network, 2012, Comprehensive molecular portraits of human breast tumours. Nature 490:61–70 Simpson L, Parsons R, 2001, PTEN: life as a tumor suppressor. Exp Cell Res 264:29–41 Wang X, Jiang X, 2008, Post-translational regulation of PTEN. Oncogene 27:5454–5463 Saal LH, Gruvberger-Saal SK, Persson C, Lovgren K, Jumppanen M, Staaf J, et al, 2008, Recurrent gross mutations of the PTEN tumor suppressor gene in breast cancers with deficient DSB repair. Nat Genet 40:102–107 Wang X, Trotman LC, Koppie T, Alimonti A, Chen Z, Gao Z, et al, 2007, NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell 128:129–139 Drinjakovic J, Jung H, Campbell DS, Strochlic L, Dwivedy A, Holt CE, 2010, E3 ligase Nedd4 promotes axon branching by downregulating PTEN. Neuron 65:341–357 Goh CP, Low LH, Putz U, Gunnersen J, Hammond V, Howitt J, et al, 2013, Ndfip1 expression in developing neurons indicates a role for protein ubiquitination by Nedd4 E3 ligases during cortical development. Neurosci Lett 555:225–230 Guo H, Qiao G, Ying H, Li Z, Zhao Y, Liang Y, et al, 2012, E3 ubiquitin ligase Cbl-b regulates pten via Nedd4 in T cells independently of its ubiquitin ligase activity. Cell Rep 1:472–482 Chung S, Nakashima M, Zembutsu H, Nakamura Y, 2011, Possible involvement of NEDD4 in keloid formation; its critical role in fibroblast proliferation and collagen production. Proc Jpn Acad Ser B Phys Biol Sci 87:563–573 Shi Y, Wang J, Chandarlapaty S, Cross J, Thompson C, Rosen N, et al, 2014, PTEN is a protein tyrosine phosphatase for IRS1. Nat Struct Mol Biol 21:522–527 Amodio N, Scrima M, Palaia L, Salman AN, Quintiero A, Franco R, et al, 2010, Oncogenic role of the E3 ubiquitin ligase NEDD4-1, a PTEN negative regulator, in non-small-cell lung carcinomas. Am J Pathol 177:2622–2634 Hong SW, Moon JH, Kim JS, Shin JS, Jung KA, Lee WK, et al, 2014, p34 is a novel regulator of the oncogenic behavior of NEDD4-1 and PTEN. Cell Death Differ 21:146–160 Trotman LC, Wang X, Alimonti A, Chen Z, Teruya-Feldstein J, Yang H, et al, 2007, Ubiquitination regulates PTEN nuclear import and tumor suppression. Cell 128:141–156 Fouladkou F, Landry T, Kawabe H, Neeb A, Lu C, Brose N, et al, 2008, The ubiquitin ligase Nedd4-1 is dispensable for the regulation of PTEN stability and localization. Proc Natl Acad Sci U S A 105:8585–8590 Maddika S, Kavela S, Rani N, Palicharla VR, Pokorny JL, Sarkaria JN, et al, 2011)WWP2 is an E3 ubiquitin ligase for PTEN. Nat Cell Biol 13:728–733 Yang Z, Yuan XG, Chen J, Lu NH, 2012, Is NEDD4-1 a negative regulator of phosphatase and tensin homolog in gastric carcinogenesis? World J Gastroenterol 18:6345–6348 Eide PW, Cekaite L, Danielsen SA, Eilertsen IA, Kjenseth A, Fykerud TA, et al, 2013, NEDD4 is overexpressed in colorectal cancer and promotes colonic cell growth independently of the PI3K/PTEN/AKT pathway. Cell Signal 25:12–18 Saal LH, Johansson P, Holm K, Gruvberger-Saal SK, She QB, Maurer M, et al, 2007, Poor prognosis in carcinoma is associated with a gene expression signature of aberrant PTEN tumor suppressor pathway activity. Proc Natl Acad Sci U S A 104:7564–7569 van de Vijver MJ, He YD, van't Veer LJ, Dai H, Hart AA, Voskuil DW, et al, 2002, A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 347:1999–2009 van 't Veer, LJ, Dai, H, van de Vijver, MJ, He, YD, Hart, AA,Mao, M et al, 2002, Gene expression profiling predicts clinical outcome of breast cancer. Nature 415:530–536 Ahn Y, Hwang CY, Lee SR, Kwon KS, Lee C, 2008, The tumour suppressor PTEN mediates a negative regulation of the E3 ubiquitin-protein ligase Nedd4. Biochem J 412:331–338 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 41 EP 47 DI 10.1007/s12253-015-9971-2 PG 7 ER PT J AU Kucharczyk, T Krawczyk, P Powrozek, T Kowalski, MD Ramlau, R Kalinka-Warzocha, E Knetki-Wroblewska, M Winiarczyk, K Krzakowski, M Milanowski, J AF Kucharczyk, Tomasz Krawczyk, Pawel Powrozek, Tomasz Kowalski, M Dariusz Ramlau, Rodryg Kalinka-Warzocha, Ewa Knetki-Wroblewska, Magdalena Winiarczyk, Kinga Krzakowski, Maciej Milanowski, Janusz TI The Effectiveness of Pemetrexed Monotherapy Depending on Polymorphisms in TS and MTHFR Genes as Well as Clinical Factors in Advanced NSCLC Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; Pemetrexed monotherapy; TS; MTHFR; Polymorphism ID Non-small cell lung cancer; Pemetrexed monotherapy; TS; MTHFR; Polymorphism AB In NSCLC, second-line chemotherapy using pemetrexed or docetaxel has limited efficacy and should be dedicated to selected groups of patients. Pemetrexed is an antifolate compound with the ability to inhibit enzymes (TS, DHFR and GARFT) involved in pyrimidine and purine synthesis. The objective of this study was to evaluate the association between polymorphisms of TS and MHFR genes and clinical outcomes in NSCLC patients treated with pemetrexed monotherapy. DNA was isolated from peripheral blood of 72 non-squamous NSCLC patients treated with pemetrexed. Using PCR and RFLP methods, the variable number of tandem repeats (VNTR), the G > C SNP in these repeats and insertion/deletion polymorphism of TS gene as well as 677C > T SNP in MTHFR gene were analyzed and correlated with disease control rate, progression-free survival and overall survival (OS) of NSCLC patients. Carriers of 2R/3R(G), 3R(C)/3R(G), 3R(G)/3R(G) genotypes showed significantly more frequent early progression than carriers of 2R/2R, 2R/ 3R(C), 3R(C)/3R(C) genotypes of TS gene (p < 0.05). Among carriers of triple 28 bp tandem repeats (3R) in TS gene and C/ C genotype of MTHFR gene a significantly shorter OS was observed (HR = 3.07; p = 0.003). In multivariate analysis, significantly higher risk of death was observed in carriers of both 3R/3R genotype in TS and C/C genotype in 677C > T SNP in MTHFR (HR = 3.85; p < 0.005) aswell as in patients with short duration of response to first-line chemotherapy (HR = 2.09; p < 0.005). Results of our study suggested that genetic factors may have a high predictive and prognostic value (even greater than clinical factors) for patients treated with pemetrexed monotherapy. C1 [Kucharczyk, Tomasz] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Krawczyk, Pawel] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Powrozek, Tomasz] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Kowalski, M Dariusz] M. Sklodowska-Curie in Warsaw, Oncology Centre-Institute, Department of Lung and Chest Cancer, W. K. Roentgena 5, 02-781 Warsaw, Poland. [Ramlau, Rodryg] Greater Poland Center of Pulmonology and Thoracic Surgery of Eugenia and Janusz ZeylandPoznan, Poland. [Kalinka-Warzocha, Ewa] Regional Centre of Oncology in Lodz, Ignacego Paderewskiego 4, 90-993 Lodz, Poland. [Knetki-Wroblewska, Magdalena] M. Sklodowska-Curie in Warsaw, Oncology Centre-Institute, Department of Lung and Chest Cancer, W. K. Roentgena 5, 02-781 Warsaw, Poland. [Winiarczyk, Kinga] M. Sklodowska-Curie in Warsaw, Oncology Centre-Institute, Department of Lung and Chest Cancer, W. K. Roentgena 5, 02-781 Warsaw, Poland. [Krzakowski, Maciej] M. Sklodowska-Curie in Warsaw, Oncology Centre-Institute, Department of Lung and Chest Cancer, W. K. Roentgena 5, 02-781 Warsaw, Poland. [Milanowski, Janusz] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. RP Krawczyk, P (reprint author), Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, 20-954 Lublin, Poland. EM krapa@poczta.onet.pl CR Dela Cruz CS, Tanoue LT, Matthay RA, 2011, Lung cancer: epidemiology, etiology, and prevention. Clin Chest Med 32:605–644 Scagliotti GV, Parikh P, von Pawel J, Biesma B, Vansteenkiste J, Manegold C, et al., 2008, Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol 26:3543–3551 Hanna N, Shepherd FA, Fossella FV, Pereira JR, DeMarinis F, von Pawel J, et al., 2004, Randomized phase III trial of pemetrexed versus docetaxel in patients with non–small-cell lung cancer previously treated with chemotherapy. J Clin Oncol. 22:1589–1597 Paz-Ares LG, de Marinis F, Dediu M, Thomas M, Pujol JL, Bidoli P, et al., 2013, PARAMOUNT: final overall survival results of the phase III study of maintenance pemetrexed versus placebo immediately after induction treatment with pemetrexed plus cisplatin for advanced nonsquamous non-small-cell lung cancer. J Clin Oncol. 31:2895–2902 Chen CY, Chang YL, Shih JY, Lin JW, Chen KY, Yang CH, et al., 2011, Thymidylate synthase and dihydrofolate reductase expression in non-small cell lung carcinoma: the association with treatment efficacy of pemetrexed. Lung Cancer 74:132–138 Buque A, Aresti U, Calvo B, Sh Muhialdin J, Munoz A, Carerra S, et al., 2013, Thymidylate synthase expression determines pemetrexed targets and resistance development in tumour cells. PLoS One 8:e63338 Wang T, Chuan Pan C, Rui Yu J, Long Y, Hong Cai X, De Yin X, et al., 2013, Association between TYMSexpression and efficacy of pemetrexed-based chemotherapy in advanced non-small cell lung cancer: ameta-analysis. PLoS One 10(8):e74284 Ceppi P, Volante M, Saviozzi S, Rapa I, Nevello S, Cambieri A, et al., 2006, Squamous cell carcinoma of the lung compared with other histotypes shows higher messenger RNA and protein levels for thymidylate synthase. Cancer 107:1589–1596 Tanaka F, Wada H, Fukui Y, Fukushima M, 2011, Thymidylate synthase, TS, gene expression in primary lung cancer patients: a large-scale study in Japanese population. AnnOncol 22:1791–1797 Tiseo M, Giovannetti E, Tibaldi C, Camerini A, Di Costanzo F, Barbieri F, et al., 2012, Pharmacogenetic study of patients with advanced non-small cell lung cancer, NSCLC, treated with second-line pemetrexed or pemetrexed-carboplatin. Lung Cancer 78:92–99 Horie N, Aiba H, Oguro K, Hojo H, Takeishi K, 1995, Functional analysis and DNA polymorphism of the tandemly repeated sequences in the 5’–terminal regulatory region for the human gene thymidylate synthase. Cell Struct Funct 20:191–197 Mandola MV, Stoehlmacher J, Muller-Weeks S, Cesarone G, Yu MC, Lenz HJ, et al., 2003, A novel single nucleotide polymorphism within the 5’ tandem repeat polymorphism of the thymidylate synthase gene abolishes USF-1 binding and alters transcriptional activity. Cancer Res 63:2898–2904 Mandola MV, Stoehlmacher J, ZhangW, Groshen S, Yu MC, Iqbal S, et al., 2004, A 6 bp polymorphism in the thymidylate synthase gene causes message instability and is associated with decreased intratumoral TS mRNA levels. Pharmacogenetics 14:319–327 Sulzyc-Bielicka V, Bielicki D, Binczak-Kuleta A, Kaczmarczyk M, Pioch W, Machoy-Mokrzynska M, et al., 2013, Thymidylate synthase gene polymorphism and survival of colorectal cancer patients receiving adjuvant 5-fluorouracil. Gent Test Mol Biomarkers 17: 799–780 Sohn KJ, Croxford R, Yates Z, LucockM, Kim YI, 2004, Effect of the methylenetetrahydrofolate reductase C677T polymorphism on chemosensitivity of colon and breast cancer cells to 5-fluorouracil and methotrexate. J Natl Cancer Inst 96:134–144 Iacopetta B, Grieu F, Joseph D, Elsaleh H, 2001, A polymorphism in the enhancer region of the thymidylate synthase promoter influences the survival of colorectal cancer patients treated with 5-fluorouracil. Br J Cancer 85:827–830 Dotor E, Cuatrecases M, Martinez-Iniesta M, Navarro M, Vilardell F, Guino E, et al., 2006, Tumor thymidylate synthase 1494del6 genotype as a prognostic factor in colorectal cancer patients receiving fluorouracil-based adjuvant treatment. J Clin Oncol 24:1603– 1611 Frosst P, Blom HJ, Milos R, Goyette P, Sheppard CA, Matthews RG, et al., 1995, A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet 10:111–113 Ardizzoni A, TiseoM, Boni L, Vincent AD, Passalacqua R, Buti S, et al., 2012, Pemetrexed versus pemetrexed and carboplatin as second-line chemotherapy in advanced non-small-cell lung cancer: results of the GOIRC 02–2006 randomized phase II study and pooled analysis with the NVALT7 trial. J Clin Oncol 30:4501–4507 Paz-Ares L, de Marinis F, Dediu M, Thomas M, Pujol JL, Bidoli P, et al., 2012, Maintenance therapy with pemetrexed plus best supportive care versus placebo plus best supportive care after induction therapy with pemetrexed plus cisplatin for advanced non-squamous non-small-cell lung cancer, PARAMOUNT): a double-blind, phase 3, randomized controlled trial. Lancet Oncol 13:247–255 Chang MH, Ahn JS, Lee J, Kim KH, Park YH, Han J, et al., 2010, The efficacy of pemetrexed as a third- or fourth-line therapy and the significance of thymidylate synthase expression in patients with advanced non-small cell lung cancer. Lung Cancer 69:323–329 JungM, Lee CH, Park HS, Lee JH, KangYA, KimSK, et al., 2013, Pharmacogenomic assessment of outcomes of pemetrexed-treated patients with adenocarcinoma of the lung. Yonsei Med J 54:854– 864 Smit EF, Burgers SA, Biesma B, Smit HJ, Eppinga P, Dingemans AM, et al., 2009, Randomized phase II and pharmacogenetic study of pemetrexed compared with pemetrexed plus carboplatin in pretreated patients with advanced non-small-cell lung cancer. J Clin Oncol 27:2038–2045 Tiseo M, Giovannetti E, Tibaldi C, Camerini A, Di Costanzo F, Barbieri F, et al., 2012, Pharmacogenetic study of patients with advanced non-small cell lung cancer, NSCLC, treated with second-line pemetrexed or pemetrexed–carboplatin. Lung Cancer 78:92–99 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 49 EP 56 DI 10.1007/s12253-015-9966-z PG 8 ER PT J AU Sudsarn, P Boonmars, Th Ruangjirachuporn, W Namwat, N Loilome, W Sriraj, P Aukkanimart, R Wonkchalee, N Jiraporn, S AF Sudsarn, Pakkayanee Boonmars, Thidarut Ruangjirachuporn, Wipaporn Namwat, Nisana Loilome, Watcharin Sriraj, Pranee Aukkanimart, Ratchadawan Wonkchalee, Nadchanan Jiraporn, Songsri TI Combination of Praziquantel and Aspirin Minimizes Liver Pathology of Hamster Opisthorchis viverrini Infection Associated Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Praziquantel; Aspirin; Opisthorchis viverrini; N-nitrosodimethylamine; Cholangiocarcinoma; Syrian hamster ID Praziquantel; Aspirin; Opisthorchis viverrini; N-nitrosodimethylamine; Cholangiocarcinoma; Syrian hamster AB Opisthorchiasis is one of the major risk factors for cholangiocarcinoma (CCA) in northeastern Thailand. An effective drug for killing this parasite is praziquantel. Recently, several reports have shown that with frequent use, praziquantel may itself be a CCA risk and can cause liver cell damage from an immunopathological response after parasite death. Aspirin has many properties including antiinflammation and anti-cancer. Therefore, we use of aspirin (As) and praziquantel (Pz) to improve hepatobiliary system function in hamsters infected with Opisthorchis viverrini (OV) and or administered N-nitrosodimethylamine (ND). Livers of OVNDAsPz, appeared healthy macroscopically, suggesting slow progression of cholangiocarcinoma evident by extent of fibrosis and bile duct cell proliferation was less than OVND although aggregations of inflammatory cells remained. Proliferating cell nuclear antigen (PCNA), cytokeratin 19 (CK19), and cancer antigen (CA19-9) staining were strongly positive in OVND, but were only slight in OVNDAs. Moreover, OVNDAsPz, appeared a few inflammatory infiltrations, bile duct proliferation, fibrosis and CCA area than the OVNDAs group. Thirty seven point five percent of hamster in this group could not develop CCA. These findings suggest that using aspirin combination with praziquantel treatment can improve the hepatobiliary system after O. viverrini infection and reduce the risk of CCA. C1 [Sudsarn, Pakkayanee] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Boonmars, Thidarut] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Ruangjirachuporn, Wipaporn] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. [Namwat, Nisana] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Loilome, Watcharin] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Sriraj, Pranee] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Aukkanimart, Ratchadawan] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Wonkchalee, Nadchanan] Khon Kaen University, Liver Fluke and Cholangiocarcinoma Research Center, 40002 Khon Kaen, Thailand. [Jiraporn, Songsri] Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. RP Boonmars, Th (reprint author), Khon Kaen University, Faculty of Medicine, Department of Parasitology, 40002 Khon Kaen, Thailand. EM bthida@kku.ac.th;boonmars@yahoo.com CR IARC, 2011, A review of human carcinogens part B: biological agents, Opisthorchis viverrini and Clonorchis sinensis). IARC Monogr Eval Carcinog Risks Hum 100:347–3762 Sripa B, Kaewkes S, Sithithaworn P,Mairiang E, Laha T, Smout M, Pairojkul C, Bhudhisawasdi V, Tesana S, Thinkamrop B, Bethony JM, Loukas A, Brindley PJ, 2007, Liver fluke induces cholangiocarcinoma. PLoS Med 4(7):e201 Boonmars T, Srirach P, Kaewsamut B, Srisawangwong T, Pinlaor S, Pinlaor P, Yongvanit P, Sithithaworn P, 2008, Apoptosis-related gene expression in hamster opisthorchiasis post praziquantel treatment. Parasitol Res 102(3):447–455 Pinlaor S, Prakobwong S,HirakuY,Kaewsamut B, Dechakhamphu S, Boonmars T, Sithithaworn P, Pinlaor P, Ma N, Yongvanit P, Kawanishi S, 2008, Oxidative and nitrative stress in Opisthorchis viverrini-infected hamsters: an indirect effect after praziquantel treatment. AmJTrop Med Hyg 78(4):564–573 Boonjaraspinyo S, Boonmars T, Aromdee C, Kaewsamut B, 2010, Effect of fingerroot on reducing inflammatory cells in hamster infected with Opisthorchis viverrini and N-nitrosodimethylamine administration. Parasitol Res 106(6):1485–1489 Jongthawin J, Techasen A, Loilome W, Yongvanit P, Namwat N, 2012, Anti-inflammatory agents suppress the prostaglandin E2 production and migration ability of cholangiocarcinoma cell lines. Asian Pac J Cancer Prev 13(Suppl):47–51 Plengsuriyakarn T, Viyanant V, Eursitthichai V, Picha P, Kupradinun P, Itharat A, Na-Bangchang K, 2012, Anticancer activities against cholangiocarcinoma, toxicity and pharmacological activities of Thai medicinal plants in animal models. BMC Complement Altern Med 12:23 Boonjaraspinyo S, Boonmars T, Aromdee C, Srisawangwong T, Kaewsamut B, Pinlaor S, Yongvanit P, Puapairoj A, 2009, Turmeric reduces inflammatory cells in hamster opisthorchiasis. Parasitol Res 105(5):1459–1463 Pinlaor S, Prakobwong S, Hiraku Y, Pinlaor P, Laothong U, Yongvanit P, 2010, Reduction of periductal fibrosis in liver fluke-infected hamsters after long-term curcumin treatment. Eur J Pharmacol 638(1–3):134–141 Prakobwong S, Khoontawad J, Yongvanit P, Pairojkul C, Hiraku Y, Sithithaworn P, Pinlaor P, Aggarwal BB, Pinlaor S, 2011, Curcumin decreases cholangiocarcinogenesis in hamsters by suppressing inflammation-mediated molecular events related to multistep carcinogenesis. Int J Cancer 129(1):88–100 Wonkchalee O, Boonmars T, Aromdee C, Laummaunwai P, Khunkitti W, Vaeteewoottacharn K, Sriraj P, Aukkanimart R, Loilome W, Chamgramol Y, Pairojkul C, Wu Z, Juasook A, Sudsarn P, 2012, Anti-inflammatory, antioxidant and hepatoprotective effects of Thunbergia laurifolia Linn. On experimental opisthorchiasis. Parasitol Res 111(1):353–359 Gu Q, Wang JD, Xia HH, Lin MC, He H, Zou B, Tu SP, Yang Y, Liu XG, Lam SK, Wong WM, Chan AO, Yuen MF, Kung HF, Wong BC, 2005, Activation of the caspase-8/bid and Bax pathways in aspirin-induced apoptosis in gastric cancer. Carcinogenesis 26(3):541–546 Redlak MJ, Power JJ, Miller TA, 2005, Role of mitochondria in aspirin-induced apoptosis in human gastric epithelial cells. Am J Physiol Gastrointest Liver Physiol 289(4):G731–G738 Zimmermann KC,Waterhouse NJ, Goldstein JC, Schuler M, Green DR, 2000, Aspirin induces apoptosis through release of cytochrome c from mitochondria. Neoplasia 2(6):505–513 Bousserouel S, Gosse F, Bouhadjar M, Soler L, Marescaux J, Raul F, 2010, Long-term administration of aspirin inhibits tumour formation and triggers anti-neoplastic molecular changes in a preclinical model of colon carcinogenesis. Oncol Rep 23(2):511–517 Cooper K, Squires H, Carroll C, Papaioannou D, Booth A, Logan RF, Maguire C, Hind D, Tappenden P, 2010, Chemoprevention of colorectal cancer: systematic review and economic evaluation. Health Technol Assess 14(32):1–206 Larkins TL, Nowell M, Singh S, Sanford GL, 2006, Inhibition of cyclooxygenase-2 decreases breast cancer cell motility, invasion and matrix metalloproteinase expression. BMC Cancer 6:181 ThunMJ, NamboodiriMM, Calle EE, FlandersWD, HeathCW, Jr., 1993, Aspirin use and risk of fatal cancer. Cancer Res 53(6):1322– 1327 Boonjaraspinyo S, Boonmars T, Aromdee C, Puapairoj A, Wu Z, 2011, Indirect effect of a turmeric diet: enhanced bile duct proliferation in Syrian hamsters with a combination of partial obstruction by Opisthorchis viverrini infection and inflammation by Nnitrosodimethylamine administration. Parasitol Res 108(1):7–14 Juasook A, Boonmars T, Wu Z, Loilome W, Veteewuthacharn K, Namwat N, Sudsarn P, Wonkchalee O, Sriraj P, Aukkanimart R, 2013, Immunosuppressive prednisolone enhances early cholangiocarcinoma in Syrian hamsters with liver fluke infection and administration of N-nitrosodimethylamine. Pathol Oncol Res 19(1): 55–62 Thamavit W, Pairojkul C, Tiwawech D, Shirai T, Ito N, 1994, Strong promoting effect of Opisthorchis viverrini infection on dimethylnitrosamine-initiated hamster liver. Cancer Lett 78(1–3): 121–125 Miliaras S, Miliaras D, Vrettou E, Zavitsanakis A, Kiskinis D, 2004, The effect of aspirin and high fibre diet on colorectal carcinoma: a comparative experimental study. Tech Coloproctol 8(Suppl 1):s59–s61 Wonkchalee O, Boonmars T, Kaewkes S, Chamgramol Y, Pairojkul C, Wu Z, Juasook A, Sudsarn P, Boonjaraspinyo S, 2011, Opisthorchis viverrini infection causes liver and biliary cirrhosis in gerbils. Parasitol Res 109(3):545–551 Muerkoster S,Wegehenkel K, Arlt A,Witt M, Sipos B, Kruse ML, Sebens T, Kloppel G, Kalthoff H, Folsch UR, Schafer H, 2004, Tumor stroma interactions induce chemoresistance in pancreatic ductal carcinoma cells involving increased secretion and paracrine effects of nitric oxide and interleukin-1beta. Cancer Res 64(4): 1331–1337 IARC, 1994, Infection with liver flukes, Opisthorchis viverrini, Opisthorchis felineus and Clonorchis sinensis). IARC Monogr Eval Carcinog Risks Hum 61:121–175 Haswell-Elkins MR, Mairiang E, Mairiang P, Chaiyakum J, Chamadol N, Loapaiboon V, Sithithaworn P, Elkins DB, 1994, Cross-sectional study of Opisthorchis viverrini infection and cholangiocarcinoma in communities within a high-risk area in northeast Thailand. Int J Cancer 59(4):505–509 Sripa B, Pairojkul C, 2008, Cholangiocarcinoma: lessons from Thailand. Curr Opin Gastroenterol 24(3):349–356 Srivatanakul P, 2001, Epidemiology of liver cancer in Thailand. Asian Pac J Cancer Prev 2(2):117–121 Honjo S, Srivatanakul P, Sriplung H, Kikukawa H, Hanai S, Uchida K, Todoroki T, Jedpiyawongse A, Kittiwatanachot P, Sripa B, Deerasamee S, Miwa M, 2005, Genetic and environmental determinants of risk for cholangiocarcinoma via Opisthorchis viverrini in a densely infested area in Nakhon Phanom, northeast Thailand. Int J Cancer 117(5):854–860 Songserm N, Promthet S, Sithithaworn P, Pientong C, Ekalaksananan T, Chopjitt P, Parkin DM, 2012, Risk factors for cholangiocarcinoma in high-risk area of Thailand: role of lifestyle, diet and methylenetetrahydrofolate reductase polymorphisms. Cancer Epidemiol 36(2):e89–e94 Sriamporn S, Pisani P, Pipitgool V, Suwanrungruang K, Kamsa-ard S, Parkin DM(2004, Prevalence of Opisthorchis viverrini infection and incidence of cholangiocarcinoma in Khon Kaen, northeast Thailand. Tropical Med Int Health 9(5):588–594 Harris RE, Namboodiri KK, Farrar WB, 1996, Nonsteroidal antiinflammatory drugs and breast cancer. Epidemiology 7(2): 203–205 Bardia A, Olson JE, Vachon CM, Lazovich D, Vierkant RA,Wang AH, Limburg PJ, Anderson KE, Cerhan JR, 2011, Effect of aspirin and other NSAIDs on postmenopausal breast cancer incidence by hormone receptor status: results from a prospective cohort study. Breast Cancer Res Treat 126(1):149–155 Bhattacharyya M, Girish GV, Ghosh R, Chakraborty S, Sinha AK, 2010, Acetyl salicylic acid, aspirin, improves synthesis of maspin and lowers incidence of metastasis in breast cancer patients [corrected]. Cancer Sci 101(10):2105–2109 Holmes MD, Chen WY, Li L, Hertzmark E, Spiegelman D, Hankinson SE, 2010, Aspirin intake and survival after breast cancer. J Clin Oncol 28(9):1467–1472 Benamouzig R, Uzzan B, Deyra J, Martin A, Girard B, Little J, Chaussade S, 2012, Prevention by daily soluble aspirin of colorectal adenoma recurrence: 4-year results of the APACC randomised trial. Gut 61(2):255–261 Chan AT, Ogino S, Fuchs CS, 2009, Aspirin use and survival after diagnosis of colorectal cancer. JAMA 302(6):649–658 Din FV, Theodoratou E, Farrington SM, Tenesa A, Barnetson RA, Cetnarskyj R, Stark L, Porteous ME, Campbell H, Dunlop MG, 2010, Effect of aspirin and NSAIDs on risk and survival from colorectal cancer. Gut 59(12):1670–1679 Fendrich V, Chen NM, Neef M, Waldmann J, Buchholz M, Feldmann G, Slater EP, Maitra A, Bartsch DK, 2010, The angiotensin-I-converting enzyme inhibitor enalapril and aspirin delay progression of pancreatic intraepithelial neoplasia and cancer formation in a genetically engineered mouse model of pancreatic cancer. Gut 59(5):630–637 Sclabas GM, Uwagawa T, Schmidt C, Hess KR, Evans DB, Abbruzzese JL, Chiao PJ, 2005, Nuclear factor kappa B activation is a potential target for preventing pancreatic carcinoma by aspirin. Cancer 103(12):2485–2490 Goonetilleke KS, Siriwardena AK, 2007, Systematic review of carbohydrate antigen, CA 19–9, as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol 33(3):266–270 Wang Z, Tian YP, 2014, Clinical value of serum tumor markers CA19-9, CA125 and CA72-4 in the diagnosis of pancreatic carcinoma. Mol Clin Oncol 2, 2):265–268 Cai WK, Lin JJ, He GH, Wang H, Lu JH, Yang GS, 2014, Preoperative serum CA19-9 levels is an independent prognostic factor in patients with resected hilar cholangiocarcinoma. Int J Clin Exp Pathol 7(11):7890–7898 Lumachi F, Lo Re G, Tozzoli R, D'Aurizio F, Facomer F, Chiara GB, Basso SM, 2014, Measurement of serum carcinoembryonic antigen, carbohydrate antigen 19–9, cytokeratin-19 fragment and matrix metalloproteinase-7 for detecting cholangiocarcinoma: a preliminary case–control study. Anticancer Res 34(11):6663–6667 Juasook A, Aukkanimart R, Boonmars T, Sudsarn P, Wonkchalee N, Laummaunwai P, Sriraj P, 2013, Tumor-related gene changes in immunosuppressive Syrian hamster cholangiocarcinoma. Pathol Oncol Res 19(4):785–794 Assy N, Hussein O, Khalil A, Luder A, Szvalb S, Paizi M, Spira G, 2007, The beneficial effect of aspirin and enoxaparin on fibrosis progression and regenerative activity in a ratmodel of cirrhosis. Dig Dis Sci 52(5):1187–1193 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 57 EP 65 DI 10.1007/s12253-015-9967-y PG 9 ER PT J AU Wang, Qh Ji, ZG Li, Hz Fan, H Chen, Zg Shi, Bb Fang, Y AF Wang, Qing-hai Ji, Zhi-Gang Li, Han-zhong Fan, Hua Chen, Zhi-gang Shi, Bing-bing Fang, Yujiang TI Clinicopathologic Comparison of Urothelial Bladder Carcinoma in Young and Elder Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Urothelial bladder cancer; Young; Pathology ID Urothelial bladder cancer; Young; Pathology AB Bladder cancer (BC) is a common aggressive malignancy and Urothelial bladder cancer (UBC) consists of the majority of BC. There is uncertainty regarding the clinicopathologic characteristics of UBCs in younger patients. To investigate the clinicopathologic features of young patients with UBCs. A total of 2825 pathological records of UBC patients, including 42 young patients (≤ 30 years old) and 2783 elder patients (> 30 years old), were retrospectively studied. The stage distribution classified was statistically significant (Χ2 = 12.25, P = 0.02) between young and old patients; superficial tumors was far more in young patients than in old patients. More young patients tended to be low- and moderate-grade UBCs (Χ2 = 6.75, P = 0.009). Young patients with superficial UBCs also showed lower recurrence rate, compared to elder patients (Χ2 = 5.77, P = 0.02). For 5-year survival rate, young patients (93.8 %) showed better than elder patients (85.1 %) (Χ2 = 4.01, P = 0.045). Patients younger than 30 years old with UBCs had low-grade and low-stage tumors and exhibited better prognosis than elder patients. C1 [Wang, Qing-hai] the Affiliated Hospital of Qingdao University, Department of Kidney Transplantation, 1677 Wutaishan Road, 266000 Qingdao, China. [Ji, Zhi-Gang] Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Department of Urology, 1 Shuaifuyuan, Wangfujing, 100730 Beijing, China. [Li, Han-zhong] Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Department of Urology, 1 Shuaifuyuan, Wangfujing, 100730 Beijing, China. [Fan, Hua] Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Department of Urology, 1 Shuaifuyuan, Wangfujing, 100730 Beijing, China. [Chen, Zhi-gang] Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Department of Urology, 1 Shuaifuyuan, Wangfujing, 100730 Beijing, China. [Shi, Bing-bing] Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Department of Urology, 1 Shuaifuyuan, Wangfujing, 100730 Beijing, China. [Fang, Yujiang] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. RP Shi, Bb (reprint author), Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Department of Urology, 100730 Beijing, China. EM shibbpumch@126.com CR Siegel R et al., 2014, Cancer statistics, 2014. CA Cancer J Clin 64(1):9–29 Shakhssalim N et al., 2010, Prominent bladder cancer risk factors in Iran. Asian Pac J Cancer Prev 11(3):601–606 Olfert SM, Felknor SA, Delclos GL, 2006, An updated review of the literature: risk factors for bladder cancer with focus on occupational exposures. South Med J 99(11):1256–1263 Tajuddin SM et al., 2014, LINE-1 methylation in leukocyte DNA, interaction with phosphatidylethanolamine N-methyltransferase variants and bladder cancer risk. Br J Cancer 110(8):2123–2130 Liu C et al., 2013, XRCC1 Arg194Trp and Arg280His polymorphisms in bladder cancer susceptibility: a meta-analysis. Crit Rev Eukaryot Gene Expr 23(4):339–354 Kirkali Z et al., 2005, Bladder cancer: epidemiology, staging and grading, and diagnosis. Urology 66(6 Suppl 1):4–34 Jemal A et al., 2010, Cancer statistics, 2010. CA Cancer J Clin 60(5):277–300 Migaldi M et al., 2004, Superficial papillary urothelial carcinomas in young and elderly patients: a comparative study. BJU Int 94(3): 311–316 Kutarski PW, Padwell A, 1993, Transitional cell carcinoma of the bladder in young adults. Br J Urol 72(5 Pt 2):749–755 Fitzpatrick JM, Reda M, 1986, Bladder carcinoma in patients 40 years old or less. J Urol 135(1):53–54 Yossepowitch O, Dalbagni G, 2002, Transitional cell carcinoma of the bladder in young adults: presentation, natural history and outcome. J Urol 168(1):61–66 Chang SY, Ma CP, 1987, Transitional cell carcinoma of the urinary bladder in patients under 40 years of age. Br J Urol 60(4):343–344 Fine SW et al., 2005, Urothelial neoplasms in patients 20 years or younger: a clinicopathological analysis using the world health organization 2004 bladder consensus classification. J Urol 174(5):1976–1980 Keetch DW, Manley CB, Catalona WJ, 1993, Transitional cell carcinoma of bladder in children and adolescents. Urology 42(4):447–449 StantonML et al., 2013, Urothelial tumors of the urinary bladder in young patients: a clinicopathologic study of 59 cases. Arch Pathol Lab Med 137(10):1337–1341 Edge SB, Compton CC, 2010, The American joint committee on cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol 17(6):1471–1474 Poletajew S et al., 2012, Urothelial bladder carcinoma in young patients is characterized by a relatively good prognosis. Ups J Med Sci 117(1):47–51 Aboutaieb R et al., 1998, Bladder tumors in young patients. Prog Urol 8(1):43–46 Lerena J et al., 2010, Transitional cell carcinoma of the bladder in children and adolescents: six-case series and review of the literature. J Pediatr Urol 6(5):481–485 Iori F et al., 2001, Superficial bladder tumors in patients under 40 years of age: clinical, prognostic and cytogenetic aspects. Urol Int 67(3):224–227 Wen YC et al., 2005, Urothelial carcinoma of the urinary bladder in young adults–clinical experience at Taipei veterans general hospital. J Chin Med Assoc 68(6):272–275 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 67 EP 70 DI 10.1007/s12253-015-9968-x PG 4 ER PT J AU Horvath, BK Pankovics, P Kalman, E Kadar, Zs Battyani, Z Lengyel, Zs Reuter, G AF Horvath, Barbara Katalin Pankovics, Peter Kalman, Endre Kadar, Zsolt Battyani, Zita Lengyel, Zsuzsanna Reuter, Gabor TI Epidemiological, Clinicopathological and Virological Features of Merkel Cell Carcinomas in Medical Center of University of Pecs, Hungary (2007–2012) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Merkel cell polyomavirus; Merkel cell carcinoma; Skin tumour; DNA tumour virus ID Merkel cell polyomavirus; Merkel cell carcinoma; Skin tumour; DNA tumour virus AB Merkel cell carcinoma (MCC) is a rare, highly aggressive skin tumour. In 2008, a Merkel cell polyomavirus (MC) was identified in MCCs as a potential etiological factor of MCC. The aims of this retrospective study were to investigate the epidemiological, clinicopathological and virological features of MCCs. Between 2007 and 2012, 11 patients had been diagnosed with MCC by histological methods in University of Pecs, Hungary. In eight MCC cases MC was tested by PCR (in primary skin lesions, lymph nodes/cutan metastases, MCC neighboring carcinomas). Clinicopathological characteristics (age, histological pattern, lymphovascular invasion, co-morbidities) of MC-positive and MC-negative cases were compared. MC was detected in three (37.5 %) out of eight patients’ primary tumour or metastasis. The average age was 73.8 (64.3 in MC-positive group). Except the youngest, 55 year-old patient (the primary tumour appeared on his leg), all tumours were found at the head and neck region. Immunosuppression (steroid therapy, chronic lymphoid leukaemia, chronic obstructive pulmonary disease) and/or old age were characteristic for all cases. Histological pattern was different in MC-positive and in MC-negative groups: MCCs with MC showed more homogeneous histological pattern, lack of lymphovascular invasion and were associated with better prognosis (mortality rate: 33 % versus 80 %). MCC associated with oncogenic virus is a newly recognized clinical entity. However, MC could not be detected in all histologically proven MCCs. The well-defined selection of patients/ disease groups and better characterization of differences between MC-positive and negative cases is an important step towards the recognition of the etiology and pathogenesis of all MCCs. C1 [Horvath, Barbara Katalin] ANTSZ Regional Institute of State Public Health Service, Regional Laboratory of Virology, Szabadsag ut 7, H-7623 Pecs, Hungary. [Pankovics, Peter] ANTSZ Regional Institute of State Public Health Service, Regional Laboratory of Virology, Szabadsag ut 7, H-7623 Pecs, Hungary. [Kalman, Endre] University of Pecs, Department of PathologyPecs, Hungary. [Kadar, Zsolt] University of Pecs, Department of Dermatology, Venereology and OncodermatologyPecs, Hungary. [Battyani, Zita] University of Pecs, Department of Dermatology, Venereology and OncodermatologyPecs, Hungary. [Lengyel, Zsuzsanna] University of Pecs, Department of Dermatology, Venereology and OncodermatologyPecs, Hungary. [Reuter, Gabor] ANTSZ Regional Institute of State Public Health Service, Regional Laboratory of Virology, Szabadsag ut 7, H-7623 Pecs, Hungary. RP Reuter, G (reprint author), ANTSZ Regional Institute of State Public Health Service, Regional Laboratory of Virology, H-7623 Pecs, Hungary. EM reuter.gabor@gmail.com CR Woo SH, Stumpfova M, Jensen UB, Lumpkin EA, Owens DM, 2010, Identification of epidermal progenitors for the Merkel cell lineage. Development 137(23):3965–3971 Van Keymeulen A, Mascre G, Youseff KK, Harel I, Michaux C, De Geest N, Szpalski C, Achouri Y, BlochW, Hassan BA, Blanpain C, 2009, Epidermal progenitors give rise to Merkel cells during embryonic development and adult homeostasis. J Cell Biol 187(1):91– 100 HeathM, Jaimes N, Lemos B,Mostaghimi A,Wang LC, Penas PF, Nghiem P, 2008, Clinical characteristics of Merkel cell carcinoma at diagnosis in 195 patients: the AEIOU features. J Am Acad Dermatol 58(3):375–381 Feng H, ShudaM, Chang Y, Moore PS, 2008, Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science 319(5866):1096–1100 Lemos BD, Storer BE, Iyer JG, Phillips JL, Bichakjian CK, Fang LC, Johnson TM, Liegeois-Kwon NJ, Otley CC, Paulson KG, Ross MI, Yu SS, Zeitouni NC, Byrd DR, Sondak VK, Gershenwald JE, Sober AJ, Nghiem P, 2010, Pathologic nodal evaluation improves prognostic accuracy in Merkel cell carcinoma: analysis of 5823 cases as the basis of the first consensus staging system. J Am Acad Dermatol 63(5):751–761 Becker JC, Houben R, Uqurel S, Trefzer U, Pfohler C, Schrama D, 2009, MC polyomavirus is frequently present in Merkel cell carcinoma of European patients. J Invest Dermatol 129(1):248–250 Varga E, Kiss M, Szabo K, Kemeny L, 2009, Detection of Merkel cell polyomavirus DNA in Merkel cell carcinomas. Br J Dermatol 161(4):930–932 Pilloni L, Manieli C, Senes G, Ribuffo D, Faa G, 2009, Merkel cell carcinoma with an unusual immunohistochemical profile. Eur J Histochem 53(4):e33 Bagasra O, 2007, Protocols for the in situ PCR-amplification and detection of mRNA and DNA sequences. Nat Protoc 2(11):2782– 2795 SchowalterRM, Pastrana DV, Pumphrey KA, Moyer AL, Buck CB, 2010, Merkel cell polyomavirus and two previously unknown polyomaviruses are chronically shed from human skin. Cell Host Microbe 7(6):509–515 van der Meijden E, Janssens RW, Lauber C, Bouwes Bavinck JN, Gorbalenya AE, Feltkamp MC, 2010, Discovery of a new human polyomavirus associated with trichodysplasia spinulosa in an immunocompromized patient. PLoS Pathog 6(7): e1001024 Scuda N, Hofmann J, Calvignac-Spencer S, Ruprecht K, Liman P, Kuhn J, Hengel H, Ehlers B, 2011, A novel human polyomavirus closely related to the African green monkey-derived lymphotropic polyomavirus. J Virol 85(9):4586–4590 Toker C, 1972, Trabecular carcinoma of the skin. Arch Dermatol 105(1):107–110 Hodgson NC, 2005, Merkel cell carcinoma: changing incidence trends. J Surg Oncol 89(1):1–4 Lunder EJ, Stern RS, 1998, Merkel-cell carcinomas in patients treated with methoxsalen and ultraviolet a radiation. N Engl J Med 339(17):1247–1248 Sihto H, Kukko H, Koljonen V, Sankila R, Bohling T, Joensuu H, 2009, Clinical factors associated with Merkel cell polyomavirus infection in Merkel cell carcinoma. J Natl Cancer Inst 101(13):938– 945 Laude HC, Jonchere B, Maubec E, Carlotti A, Marinho E, Couturaud B, Peter M, Sastre-Garau X, Avril MF, Duprin N, Rozenberg F, 2010, Distinct merkel cell polyomavirus molecular features in tumour and non tumour specimens from patients with merkel cell carcinoma. PLoS Pathog 6(8):e1001076 Bhatia K, Goedert JJ, Modali R, Preiss L, Ayers LW, 2010, Immunological detection of viral large T antigen identifies a subset of Merkel cell carcinoma tumors with higher viral abundance and better clinical outcome. Int J Cancer 127(6):1493–1496 Waltari M, Sihto H, Kukko H, Koljonen V, Sankila R, Bohling T, Joensuu H, 2011, Association of Merkel cell polyomavirus infection with tumor p53, KIT, stem cell factor, PDGFR-alpha and survival in Merkel cell carcinoma. Int J Cancer 129(3):619–628 Busam KJ, Jungbluth AA, Rekthman N, Coit D, PulitzerM, Bini J, Arora R, Hanson NC, Tassello JA, Frosina D, Moore P, Chang Y, 2009)Merkel cell polyomavirus expression in merkel cell carcinomas and its absence in combined tumors and pulmonary neuroendocrine carcinomas. Am J Surg Pathol 33(9):1378–1385 Martel-Jantin C, Filippone C, Cassar O, Peter M, Tomasic G, Vielh P, Briere J, Petrella T, Aubriot-Lorton MH, Mortier L, Jouvion G, Sastre-GarauX, Robert C,GessainA(2012, Genetic variability and integration of Merkel cell polyomavirus in Merkel cell carcinoma. Virology 426(2):134–142 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 71 EP 77 DI 10.1007/s12253-015-9974-z PG 7 ER PT J AU Tarnoki, LD Tarnoki, DA Ohlmann-Knafo, S Pickuth, D AF Tarnoki, Laszlo David Tarnoki, Domonkos Adam Ohlmann-Knafo, Susanne Pickuth, Dirk TI Pattern of Tumour Spread of Common Primary Tumours as Seen on Magnetic Resonance Imaging SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Breast tumor; Vertebral metastasis; Spine; Pedicle ID Lung cancer; Breast tumor; Vertebral metastasis; Spine; Pedicle AB Although some reports with computed tomography and bone scintigraphy are available in the literature, the distinct epidemiologic description of skeletal metastatic pattern of various tumors is still lacking. This study uses a novel approach to identify skeletal metastases from magnetic resonance imaging (MRI) data to describe metastatic pattern in common malignancies. A retrospective analysis of 130 cancer patients (42 lung, 56 breast, 11 prostate cancers; 21 multiple myeloma) with vertebral metastases and without disseminated disease, and whom underwent a whole body 3Tesla MRI investigation (Discovery MR750w), was carried out. Multiple myeloma had the most commonly disseminated metastatic disease (95 %) compared to lung (28 %), breast (44 %) and prostate (71 %) cancers. Lung cancer was related to more frequent pedicle involvement compared to breast or prostate cancer (29, 9 and 0 %, p<0.05). Pathologic fracture was mainly associated with multiple myeloma (43 %). The prevalence of lung cancer metastases was more frequent in the lumbal spine (81 %), as well as particular in C7, D7, D8, D9 and L1, compared to breast cancers. Most differences among tumors were detected in the extravertebral osseous metastatic pattern (p<0.05). The highest frequency of extravertebral skeletal metastases was present in multiple myeloma (28 to 76 %). Brain metastasis was more frequent in lung cancer compared to breast cancers (35 % vs. 17 %, p<0.05). Significant differences in the skeletal metastatic pattern among common malignancies were demonstrated with MRI. C1 [Tarnoki, Laszlo David] Caritasklinikum Saarbrucken St. Theresia, Academic Teaching Hospital of Saarland University, Department of Diagnostic and Interventional Radiology, Rheinstraße 2, D-66113 Saarbrucken, Germany. [Tarnoki, Domonkos Adam] Caritasklinikum Saarbrucken St. Theresia, Academic Teaching Hospital of Saarland University, Department of Diagnostic and Interventional Radiology, Rheinstraße 2, D-66113 Saarbrucken, Germany. [Ohlmann-Knafo, Susanne] Caritasklinikum Saarbrucken St. Theresia, Academic Teaching Hospital of Saarland University, Department of Diagnostic and Interventional Radiology, Rheinstraße 2, D-66113 Saarbrucken, Germany. [Pickuth, Dirk] Caritasklinikum Saarbrucken St. Theresia, Academic Teaching Hospital of Saarland University, Department of Diagnostic and Interventional Radiology, Rheinstraße 2, D-66113 Saarbrucken, Germany. RP Tarnoki, LD (reprint author), Caritasklinikum Saarbrucken St. Theresia, Academic Teaching Hospital of Saarland University, Department of Diagnostic and Interventional Radiology, D-66113 Saarbrucken, Germany. EM tarnoki4@gmail.com CR Vineis P, Wild CP, 2014, Global cancer patterns: causes and prevention. Lancet 383:549–557 Sciubba DM, Gokaslan ZL, 2006, Diagnosis and management of metastatic spine disease. Surg Oncol 15:141–151 Sundaresan N, Boriani S, Rothman A, Holtzman R, 2004, Tumors of the osseous spine. J Neurooncol 69:273–290 White AP, Kwon BK, Lindskog DM, Friedlaender GE, Grauer JN, 2006, Metastatic disease of the spine. J Am Acad Orthop Surg 14: 587–598 Witham TF, Khavkin YA, Gallia GL, Wolinsky JP, Gokaslan ZL, 2006, Surgery insight: current management of epidural spinal cord compression frommetastatic spine disease. Nat Clin Pract Neurol 2: 87–94 Memon AG, Jaleel A, Aftab J, 2006, Pattern of prostatic carcinoma metastases in bones detected by bone scans using Technitium 99m methyl dipohsphate, Tc99m MDP, imaging technique. Pak J Med Sci 22:180–183 Morgan JWM, Adcock KM, Donohue RE, 1990, Distribution of skeletal metastases in prostatic and lung cancer. Urology 36:31–34 Coleman RE, Rubens RD, 1987, The clinical course of bone metastases from breast cancer. Br J Cancer 55:61–66 Coleman RE, 1997, Skeletal complications of malignancy. Cancer 80:1588–1594 Bubendorf L, Schopfer A, Wagner U, Sauter G, Moch H, Willi N, Gasser TC, Mihatsch MJ, 2000, Metastatic patterns of prostate cancer: an autopsy study of 1,589 patients. HumPathol 31:578–583 Marulanda GA, Mont MA, Lucci A, Letson GD, Khakpour N, 2008, Orthopedic surgery implications of breast cancer. Expert Rev Anticancer Ther 8:949–956 Black P, 1979, Spinal metastasis: current status and recommended guidelines for management. Neurosurgery 5:726–746 Scutellari PN, Addonisio G, Righi R, Giganti M, 2000, Diagnostic imaging of bone metastases. Radiol Med 100:429–435 Sciubba DM, Petteys RJ, Dekutoski MB, Fisher CG, Fehlings MG, Ondra SL, Rhines LD, Gokaslan ZL, 2010, Diagnosis and management of metastatic spine disease. A review. J Neurosurg Spine 13:94– 108 Khanna AJ, Shindle MK, Wasserman BA, Gokaslan ZL, Gonzales RA, Buchowski JM, Riley LH 3rd, 2005, Use of magnetic resonance imaging in differentiating compartmental location of spinal tumors. Am J Orthop, Belle Mead NJ, 34:472–476 Kakhki VR, Anvari K, Sadeghi R, Mahmoudian AS, Torabian- Kakhki M, 2013, Pattern and distribution of bone metastases in common malignant tumors. NuclMed Rev Cent East Eur 16:66–69 Cossigny D, Quan GM, 2012, In vivo animal models of spinal metastasis. Cancer Metastasis Rev 31:99–108 Schaberg J, Gainor BJ, 1985, A profile of metastatic carcinoma of the spine. Spine, Phila Pa 1976, 10:19–20 Weigert F, Reiser M, Pfandner K, 1987, Imaging of neoplastic changes in the spine using MR tomography]. Rofo 146:123–30 Bubendorf L, Schopfer A, Wagner U, Sauter G, Moch H, Willi N, Gasser TC, Mihatsch MJ, 2000, Metastatic patterns of prostate cancer: an autopsy study of 1,589 patients. HumPathol 31:578–583 Bohlman HH, Sachs BL, Carter JR, Riley L, Robinson RA, 1986, Primary neoplasms of the cervical spine. Diagnosis and treatment of twenty-three patients. J Bone Joint Surg Am 68:483–494 Hart RA, Boriani S, Biagini R, Currier B, Weinstein JN, 1997, A system for surgical staging and management of spine tumors. A clinical outcome study of giant cell tumors of the spine. Spine, Phila Pa 1976, 22:1773–1782 Lee C, Jung C, 2012, Metastatic spinal tumor. Asian Spine J 6:71–87 Shackman R, Harrison CV, 1948, Occult bone metastases. Br J Surg 35:385–389 Algra PR, Heimans JJ, Valk J, Nauta JJ, Lachniet M, Van Kooten B, 1992, Dometastases in vertebrae begin in the body or the pedicles? Imaging study in 45 patients. AJR Am J Roentgenol 158:1275– 1279 Abdel-Wanis ME, Solyman MT, Hasan NM, 2011, Sensitivity, specificity and accuracy of magnetic resonance imaging for differentiating vertebral compression fractures caused by malignancy, osteoporosis, and infections. J Orthop Surg, Hong Kong, 19:145– 150 Arguello F, Baggs RB, Duerst RE, Johnstone L, McQueen K, Frantz CN, 1990, Pathogenesis of vertebral metastasis and epidural spinal cord compression. Cancer 65:98–106 Asdourian PL, Weidenbaum M, DeWald RL, Hammerberg KW, Ramsey RG, 1990, The pattern of vertebral involvement in metastatic vertebral breast cancer. Clin Orthop Relat Res 250:164–170 Coleman RE, 2006, Clinical features ofmetastatic bone disease and risk of skeletal morbidity. Clin Cancer Res 12:6243–6249 Bundred N, Walker RA, Ratcliffe WA, Warwick J, Morrison JM, Ratcliffe JG, 1992, Parathyroid hormone related protein and skeletal morbidity in breast cancer. Eur J Cancer 28:690–692 Koenders P, Beex LV, Langens R, Kloppenborg PW, Smals AG, Benraad TJ, 1991, Steroid hormone receptor activity of primary human breast cancer and pattern of first metastasis. Breast Cancer Res Treat 18:27–32 YuhWT, Quets JP, Lee HJ, Simonson TM, Michalson LS, Nguyen PT, Sato Y, Mayr NA, Berbaum KS, 1996, Anatomic distribution of metastases in the vertebral body and modes of hematogenous spread. Spine, Phila Pa 1976, 21:2243–2250 Zhou Q, Zu L, Li L, Chen X, Chen X, Li Y, Liu H, Sun Z, 2014, Screening and establishment of human lung cancer cell lineswith organspecific metastasis potential. Zhongguo Fei Ai Za Zhi 17:175–182 Melton LJ 3rd, Kyle RA, Achenbach SJ, Oberg AL, Rajkumar SV, 2005, Fracture risk with multiple myeloma: a population-based study. J Bone Miner Res 20:487–493 Song IC, Kim JN, Choi YS, Ryu H, LeeMW, Lee HJ, Yun HJ, Kim S, Kwon ST, Jo DY, 2014, Diagnostic and prognostic implications of spine magnetic resonance imaging at diagnosis in patients with multiple myeloma. Cancer Res Treat., DOI 10.4143/crt.2014.010 Greenspan SL, Wagner J, Nelson JB, Perera S, Britton C, Resnick NM(2013, Vertebral fractures and trabecular microstructure in men with prostate cancer on androgen deprivation therapy. J Bone Miner Res 28:325–332 Klimo P Jr, Schmidt MH, 2004, Surgical management of spinal metastases. Oncologist 9:188–196 Schirrmeister H, Guhlmann A, Elsner K, Kotzerke J, Glatting G, Rentschler M, Neumaier B, Trager H, Nussle K, Reske SN, 1999, Sensitivity in detecting osseous lesions depends on anatomic localization: planar bone scintigraphy versus 18F PET. J Nucl Med 40:1623– 1629 Aydinli U, Ozturk C, Bayram S, Sarihan S, Evrensel T, Yilmaz HS, 2006, Evaluation of lung cancer metastases to the spine. Acta Orthop Belg 72:592–597 Batson OV, 1940, The function of vertebral veins and their role in the spread of metastases. Ann Surg 112:38–149 Bubendorf L, Schopfer A, Wagner U, Sauter G, Moch H, Willi N, Gasser TC, Mihatsch MJ, 2000, Metastatic patterns of prostate cancer: an autopsy study of 1,589 patients. HumPathol 31:578–583 Moreno A, Albiach C, Soria R, Vidal V, Gomez R, Antequera M, 2014, Oligometastases in prostate cancer: restaging stage IV cancers and new radiotherapy options. Radiat Oncol 9:258 Saitoh H, Yoshida K, Uchijima Y, Kobayashi N, Suwata J, Kamata S, 1990, Two different lymph node metastatic patterns of a prostate cancer. Cancer 65:1843–1846 Fabozzi SJ, Schellhammer PF, El-Mahdi AM, 1995, Pulmonary metastases from prostate cancer. Cancer 75:2706–2709 Maccauro G, Spinelli MS, Mauro S, Perisano C, Graci C, RosaMA, 2011, Physiopathology of spine metastasis. Int J Surg Oncol 2011: 107969 Sugiura H, Yamada K, Sugiura T, Hida T, Mitsudomi T, 2008, Predictors of survival in patients with bone metastasis of lung cancer. Clin Orthop Relat Res 466:729–736 Sorensen JB, Hansen HH, Hansen M, Dombernowsky P, 1988, Brain metastases in adenocarcinoma of the lung: frequency, risk groups, and prognosis. J Clin Oncol 6:1474–1480 Lagerwaard FJ, Levendag PC, Nowak PJ, Eijkenboom WM, Hanssens PE, Schmitz PI, 1999, Identification of prognostic factors in patients with brain metastases: a review of 1292 patients. Int J Radiat Oncol Biol Phys 43:795–803 Riihimaki M, Hemminki A, Fallah M, Thomsen H, Sundquist K, Sundquist J, Hemminki K, 2014, Metastatic sites and survival in lung cancer. Lung Cancer 86:78–84 Petren-MallminM, Nordstrom B, Andreasson I, Nyman R, Jonsson H Jr, Rauschning W, Hemmingsson A, 1992, MR imaging with histopathological correlation in vertebral metastases of breast cancer. Acta Radiol 33:213–220 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 79 EP 85 DI 10.1007/s12253-015-9975-y PG 7 ER PT J AU Soylu, H Acar, N Ozbey, O Unal, B Koksal, TI Bassorgun, I Ciftcioglu, A Ustunel, I AF Soylu, Hakan Acar, Nuray Ozbey, Ozlem Unal, Betul Koksal, Turker Ismail Bassorgun, Ibrahim Ciftcioglu, Akif Ustunel, Ismail TI Characterization of Notch Signalling Pathway Members in Normal Prostate, Prostatic Intraepithelial Neoplasia (PIN) and Prostatic Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Immunohistochemistry; Notch signalling pathway; Prostate cancer; Prostatic intraepithelial neoplasia ID Immunohistochemistry; Notch signalling pathway; Prostate cancer; Prostatic intraepithelial neoplasia AB Prostate Cancer (PCa) holds the second place in terms of cancer-related mortality rate among men. The Notch signalling pathway regulates the proliferation and differentiation in embryonic and adult tissues and determines the cell fate. The body of knowledge in the present literature is currently controversial about the effect of the Notch pathway on prostatic cancer. Therefore, the present study aimed to examine the immunolocalization and expression levels of Notch1- 4, Jagged1-2, Delta, HES1 and HES5 from among the members of the Notch signalling pathway in tissues of normal, prostatic intraepithelial neoplasia (PIN) and malignant prostate. The current study included a sample of 20 patients with localised prostatic adenocarcinoma, 18 patients with high grade PIN (H-PIN) and 18 normal prostatic tissue. Immunolocalisations of Notch1, 2, 3, 4, Jagged1, 2, Delta, HES1 and HES5 were identified through the immunohistochemical method. The findings of the present study showed that all in-scope members of the Notch signalling pathway were localised in PIN structures to a greater extent than in other tissues and from amongst these members, specifically Notch1, Notch4, Jagged1 and HES1 were at more significant levels. Consequently, the findings of the present study may indicate that the Notch signalling pathway can play a role especially in the formation of PIN structures. C1 [Soylu, Hakan] Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, 07070 Antalya, Turkey. [Acar, Nuray] Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, 07070 Antalya, Turkey. [Ozbey, Ozlem] Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, 07070 Antalya, Turkey. [Unal, Betul] Akdeniz University, Faculty of Medicine, Department of Pathology, 07070 Antalya, Turkey. [Koksal, Turker Ismail] Akdeniz University, Faculty of Medicine, Department of Urology, 07070 Antalya, Turkey. [Bassorgun, Ibrahim] Akdeniz University, Faculty of Medicine, Department of Pathology, 07070 Antalya, Turkey. [Ciftcioglu, Akif] Akdeniz University, Faculty of Medicine, Department of Pathology, 07070 Antalya, Turkey. [Ustunel, Ismail] Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, 07070 Antalya, Turkey. RP Ustunel, I (reprint author), Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, 07070 Antalya, Turkey. EM iustunel@akdeniz.edu.tr CR Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics, 2012. CA Cancer J Clin 62(1):10–29., DOI 10.3322/caac.20138 Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, Feuer EJ, Thun MJ, 2005, Cancer statistics, 2005. CA Cancer J Clin 55(1):10–30 Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics, 2009. CA Cancer J Clin 59(4):225–249., DOI 10.3322/ caac.20006 Ferlay J, Parkin DM, Steliarova-Foucher E, 2010, Estimates of cancer incidence and mortality in europe in 2008. Eur J Cancer 46(4):765–781., DOI 10.1016/j.ejca.2009.12.014 Baade PD, Coory MD, Aitken JF, 2004, International trends in prostate-cancer mortality: the decrease is continuing and spreading. Cancer Causes Control : CCC 15(3):237–241., DOI 10.1023/ B:CACO.0000024212.66334.26 Gronberg H, 2003, Prostate cancer epidemiology. Lancet 361(9360):859–864., DOI 10.1016/S0140-6736(03)12713-4 Greene KL, Li L.C., Okino S.T., Carroll P.R., 2008, Molecular basis of prostate cancer. In: Mendelson J, Howley, P.M., Israel M.A., Gray, J.W., Thompson, C.B., ed, The Molecular Basis of Cancer. Third edn., pp 431–441 Zhu H, Zhou X, Redfield S, Lewin J, Miele L, 2013, Elevated jagged-1 and notch-1 expression in high grade and metastatic prostate cancers. Am J Transl Res 5(3):368–378 Danza G, Di Serio C, Rosati F, Lonetto G, Sturli N, Kacer D, Pennella A, Ventimiglia G, Barucci R, Piscazzi A, Prudovsky I, Landriscina M, Marchionni N, Tarantini F, 2012, Notch signaling modulates hypoxia-induced neuroendocrine differentiation of human prostate cancer cells. Molecular Cancer Research : MCR 10(2):230–238., DOI 10.1158/1541-7786.MCR-11-0296 Gridley T, 2007, Notch signaling in vascular development and physiology. Development 134(15):2709–2718., DOI 10.1242/dev.004184 Fortini ME, 2009, Notch signaling: the core pathway and its posttranslational regulation. Dev Cell 16(5):633–647., DOI 10.1016/j. devcel.2009.03.010 Blaumueller CM, Qi H, Zagouras P, Artavanis-Tsakonas S, 1997, Intracellular cleavage of notch leads to a heterodimeric receptor on the plasma membrane. Cell 90(2):281–291 Miele L, 2006, Notch signaling. Clin Cancer Res : Off J Am Assoc Cancer Res 12(4):1074–1079., DOI 10.1158/1078-0432.CCR-05-2570 Miele L,Miao H,Nickoloff BJ, 2006, NOTCH signaling as a novel cancer therapeutic target. Curr Cancer Drug Targets 6(4):313–323 Dunwoodie SL, Henrique D, Harrison SM, Beddington RS, 1997, Mouse Dll3: a novel divergent delta gene which may complement the function of other delta homologues during early pattern formation in the mouse embryo. Dev 124(16):3065–3076 Lindsell CE, Shawber CJ, Boulter J,Weinmaster G, 1995, Jagged: a mammalian ligand that activates Notch1. Cell 80(6):909–917 Shawber C, Boulter J, Lindsell CE,Weinmaster G, 1996, Jagged2: a serrate-like gene expressed during rat embryogenesis. Dev Biol 180(1):370–376., DOI 10.1006/dbio.1996.0310 Maier MM, Gessler M, 2000, Comparative analysis of the human and mouse Hey1 promoter: Hey genes are new notch target genes. Biochem Biophys Res Commun 275(2):652–660., DOI 10.1006/ bbrc.2000.3354 Grishina IB, Kim SY, Ferrara C, Makarenkova HP, Walden PD, 2005, BMP7 inhibits branching morphogenesis in the prostate gland and interferes with notch signaling. Dev Biol 288(2):334– 347., DOI 10.1016/j.ydbio.2005.08.018 Wang XD, Leow CC, Zha J, Tang Z,Modrusan Z, Radtke F, Aguet M, de Sauvage FJ, Gao WQ, 2006, Notch signaling is required for normal prostatic epithelial cell proliferation and differentiation. Dev Biol 290(1):66–80., DOI 10.1016/j.ydbio.2005.11.009 Shou J, Ross S, Koeppen H, de Sauvage FJ, Gao WQ, 2001, Dynamics of notch expression during murine prostate development and tumorigenesis. Cancer Res 61(19):7291–7297 Scorey N, Fraser SP, Patel P, Pridgeon C, Dallman MJ, Djamgoz MB, 2006, Notch signalling and voltage-gated Na + channel activity in human prostate cancer cells: independent modulation of in vitro motility. Prostate Cancer Prostatic Dis 9(4):399–406., DOI 10.1038/sj.pcan.4500894 BrownMD,Gilmore PE, HartCA, Samuel JD, RamaniVA,George NJ, Clarke NW, 2007, Characterization of benign and malignant prostate epithelial hoechst 33342 side populations. Prostate 67(13): 1384–1396., DOI 10.1002/pros.20620 Santagata S, Demichelis F, Riva A, Varambally S, Hofer MD, Kutok JL, Kim R, Tang J, Montie JE, Chinnaiyan AM, Rubin MA, Aster JC, 2004, JAGGED1 expression is associated with prostate cancer metastasis and recurrence. Cancer Res 64(19): 6854–6857., DOI 10.1158/0008-5472.CAN-04-2500 Kopan R, Ilagan MX, 2009, The canonical notch signaling pathway: unfolding the activation mechanism. Cell 137(2):216–233., DOI 10.1016/j.cell.2009.03.045 Yoon HA, Noh MH, Kim BG, Han JS, Jang JS, Choi SR, Jeong JS, Chun JH, 2011, Clinicopathological significance of altered notch signaling in extrahepatic cholangiocarcinoma and gallbladder carcinoma. World J Gastroenterol : WJG 17(35):4023–4030., DOI 10. 3748/wjg.v17.i35.4023 Callahan R, Raafat A, 2001, Notch signaling in mammary gland tumorigenesis. J Mammary Gland Biol Neoplasia 6(1):23–36 Harrison H, Farnie G, Howell SJ, Rock RE, Stylianou S, Brennan KR, Bundred NJ, Clarke RB, 2010, Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor. Cancer Res 70(2):709–718., DOI 10.1158/0008-5472.CAN-09-1681 ZayzafoonM, Abdulkadir SA, McDonald JM, 2004, Notch signaling and ERK activation are important for the osteomimetic properties of prostate cancer bone metastatic cell lines. The J Biol Chem 279(5):3662–3670., DOI 10.1074/jbc.M308158200 WangX, FuY, ChenX, Ye J, LuB, Ye F, LuW, XieX(2010, The expressions of bHLH gene HES1 and HES5 in advanced ovarian serous adenocarcinomas and their prognostic significance: a retrospective clinical study. J Cancer Res Clin Oncol 136(7):989–996., DOI 10.1007/s00432-009-0744-8 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 87 EP 94 DI 10.1007/s12253-015-9983-y PG 8 ER PT J AU Melling, N Grimm, N Simon, R Stahl, P Bokemeyer, C Terracciano, L Sauter, G Izbicki, RJ Marx, HA AF Melling, Nathaniel Grimm, Norbert Simon, Ronald Stahl, Philip Bokemeyer, Carsten Terracciano, Luigi Sauter, Guido Izbicki, R Jakob Marx, H Andreas TI Loss of H2Bub1 Expression is Linked to Poor Prognosis in Nodal Negative Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tissue microarray; Gastrointestinal cancer; Colorectal cancer; Biomarker ID Tissue microarray; Gastrointestinal cancer; Colorectal cancer; Biomarker AB To correlate H2Bub1 expression with outcome in colorectal cancer, H2Bub1 expression was analyzed by immunohistochemistry on a tissue microarray containing 1800 colorectal cancers. Results were compared to clinicopathological parameters. H2Bub1 IHC was seen in 1256 (79.3 %) of 1584 interpretable CRC and was considered weak in 26.2 % and strong in 53.1% of cancers. H2Bub1 expression was completely lost in 20.7 % of the cases. Loss of H2Bub1 expression was associated with high tumor grade (p = 0.0211), high tumor stage (p = 0.0003), positive nodal status (p = 0.0139) and histological tumor type (p = 0.0202). No link was found between H2Bub1 expression and tumor localization (p = 0.1262), peritumoral lymphocytic infiltration (p = 0.2523) or vascular invasion (p = 0.5970). Loss of H2Bub1 expression in CRC was strongly associated with poor patient survival (p = 0.0006). This observation held true also in a subset survival analysis of nodal negative (N0) and nodal positive (N1) cancers (p = 0.0296 and p = 0.0197, respectively). In the subgroup of p53 negative cancers no prognostic impact of H2Bub1 staining was seen (p = 0.1924), whereas in p53 positive CRC H2Bub1 expression loss was associated with poor prognosis (p = 0.0031). Strikingly worsened outcome was found for nodal negative cancers presenting with accumulation of p53 when H2Bub1 expression was lost (p = 0.0006). Our data demonstrate that a reduced H2Bub1 expression is a strong prognostic biomarker both in nodal negative and nodal positive CRC. H2Bub1 expression measurement might help to select nodal negative CRC patients that may benefit from adjuvant therapy. C1 [Melling, Nathaniel] University Medical Center Hamburg-Eppendorf, Department of Surgery, 20246 Hamburg, Germany. [Grimm, Norbert] Regio Hosptital Pinneberg, Department of Surgery, 25421 Pinneberg, Germany. [Simon, Ronald] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. [Stahl, Philip] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. [Bokemeyer, Carsten] University Medical Center Hamburg-Eppendorf, Hubertus Wald Cancer Center, Department of Oncology, Hematology, BMT with section Pneumology, 20246 Hamburg, Germany. [Terracciano, Luigi] University Hospital Basel, Institute of Pathology, 4001 Basel, Switzerland. [Sauter, Guido] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. [Izbicki, R Jakob] University Medical Center Hamburg-Eppendorf, Department of Surgery, 20246 Hamburg, Germany. [Marx, H Andreas] University Medical Center Hamburg Eppendorf, Pathology, Martinistrasse 52, 20246 Hamburg, Germany. RP Marx, HA (reprint author), University Medical Center Hamburg Eppendorf, Pathology, 20246 Hamburg, Germany. EM a.marx@uke.de CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90., DOI 10. 3322/caac.20107 Bilchik AJ, Nora DT, Saha S, Turner R, Wiese D, Kuo C, Ye X, Morton DL, Hoon DS, 2002, The use of molecular profiling of early colorectal cancer to predict micrometastases. Arch Surg 137(12):1377–1383 Kim J, Hake SB, Roeder RG, 2005, The human homolog of yeast BRE1 functions as a transcriptional coactivator through direct activator interactions. Mol Cell 20(5):759–770., DOI 10.1016/j.molcel. 2005.11.012 Pavri R, Zhu B, Li G, Trojer P,Mandal S, Shilatifard A, Reinberg D, 2006, Histone H2B monoubiquitination functions cooperatively with FACT to regulate elongation by RNA polymerase II. Cell 125(4):703–717., DOI 10.1016/j.cell.2006.04.029 Minsky N, Shema E, Field Y, Schuster M, Segal E, Oren M, 2008, Monoubiquitinated H2B is associated with the transcribed region of highly expressed genes in human cells. Nat Cell Biol 10(4):483– 488., DOI 10.1038/ncb1712 Johnsen SA, 2012, The enigmatic role of H2Bub1 in cancer. FEBS Lett 586(11):1592–1601., DOI 10.1016/j.febslet.2012.04.002 Pirngruber J, Shchebet A, Schreiber L, Shema E, Minsky N, Chapman RD, Eick D, Aylon Y, Oren M, Johnsen SA, 2009, CDK9 directs H2B monoubiquitination and controls replicationdependent histone mRNA 3′-end processing. EMBO Rep 10(8): 894–900., DOI 10.1038/embor.2009.108 Kari V, Shchebet A, Neumann H, Johnsen SA, 2011, The H2B ubiquitin ligase RNF40 cooperates with SUPT16H to induce dynamic changes in chromatin structure during DNA double-strand break repair. Cell Cycle 10(20):3495–3504., DOI 10.4161/cc.10.20.17769 Moyal L, Lerenthal Y, Gana-Weisz M, Mass G, So S, Wang SY, Eppink B, Chung YM, Shalev G, Shema E, Shkedy D, Smorodinsky NI, van Vliet N, Kuster B, Mann M, Ciechanover A, Dahm-Daphi J, Kanaar R, Hu MC, Chen DJ, Oren M, Shiloh Y, 2011, Requirement of ATM-dependent monoubiquitylation of histone H2B for timely repair of DNA double-strand breaks. Mol Cell 41(5):529–542., DOI 10.1016/j.molcel.2011.02.015 Karpiuk O, Najafova Z, Kramer F, HennionM, Galonska C, Konig A, Snaidero N, Vogel T, Shchebet A, Begus-Nahrmann Y, Kassem M, Simons M, Shcherbata H, Beissbarth T, Johnsen SA, 2012, The histone H2B monoubiquitination regulatory pathway is required for differentiation of multipotent stem cells. Mol Cell 46(5):705–713., DOI 10.1016/j.molcel.2012.05.022 Trujillo KM, Osley MA, 2012, A role for H2B ubiquitylation in DNA replication. Mol Cell 48(5):734–746., DOI 10.1016/j.molcel. 2012.09.019 Sadeghi L, Siggens L, Svensson JP, Ekwall K, 2014, Centromeric histone H2B monoubiquitination promotes noncoding transcription and chromatin integrity. Nat Struct Mol Biol 21(3):236–243., DOI 10.1038/nsmb.2776 Espinosa JM, 2008, Histone H2B ubiquitination: the cancer connection. Genes Dev 22(20):2743–2749., DOI 10.1101/gad.1732108 Prenzel T, Begus-Nahrmann Y, Kramer F, Hennion M, Hsu C, Gorsler T, Hintermair C, Eick D, Kremmer E, Simons M, Beissbarth T, Johnsen SA, 2011, Estrogen-dependent gene transcription in human breast cancer cells relies upon proteasomedependent monoubiquitination of histone H2B. Cancer Res 71(17):5739–5753., DOI 10.1158/0008-5472.CAN-11-1896 HahnMA, Dickson KA, Jackson S, Clarkson A, Gill AJ,Marsh DJ, 2012, The tumor suppressor CDC73 interacts with the ring finger proteins RNF20 and RNF40 and is required for the maintenance of histone 2B monoubiquitination. Hum Mol Genet 21(3):559–568., DOI 10.1093/hmg/ddr490 Urasaki Y, Heath L, Xu CW, 2012, Coupling of glucose deprivation with impaired histone H2B monoubiquitination in tumors. PLoS One 7, 5):e36775., DOI 10.1371/journal.pone.0036775 Mirlacher M, Simon R, 2010, Recipient block TMA technique. Methods Mol Biol 664:37–44., DOI 10.1007/978-1-60761-806-5_4 Marx A, Simon P, Simon R, Mirlacher M, Izbicki JR, Yekebas E, Kaifi JT, Terracciano L, Sauter G, 2008, AMACR expression in colorectal cancer is associated with left-sided tumor localization. Virchows Archiv : An Int J Pathol 453(3):243–248., DOI 10.1007/ s00428-008-0646-1 Yemelyanova A, Vang R, Kshirsagar M, Lu D, Marks MA, Shih Ie M, Kurman RJ, 2011, Immunohistochemical staining patterns of p53 can serve as a surrogate marker for TP53 mutations in ovarian carcinoma: an immunohistochemical and nucleotide sequencing analysis. Modern Pathology : an Official Journal of the United States and Canadian Academy of Pathology, Inc 24(9):1248– 1253., DOI 10.1038/modpathol.2011.85 Zhang F, Yu X, 2011, WAC, a functional partner of RNF20/40, regulates histone H2B ubiquitination and gene transcription. Mol Cell 41(4):384–397., DOI 10.1016/j.molcel.2011.01.024 Chernikova SB, Razorenova OV, Higgins JP, Sishc BJ, Nicolau M, Dorth JA, Chernikova DA, Kwok S, Brooks JD, Bailey SM, Game JC, Brown JM, 2012, Deficiency in mammalian histone H2B ubiquitin ligase Bre1, Rnf20/ Rnf40, leads to replication stress and chromosomal instability. Cancer Res 72(8):2111–2119., DOI 10.1158/0008-5472. CAN-11-2209 Nakamura K, Kato A, Kobayashi J, Yanagihara H, Sakamoto S, Oliveira DV, Shimada M, Tauchi H, Suzuki H, Tashiro S, Zou L, Komatsu K, 2011, Regulation of homologous recombination by RNF20-dependent H2B ubiquitination. Mol Cell 41(5):515–528., DOI 10.1016/j.molcel.2011.02.002 Shema E, Tirosh I, Aylon Y, Huang J, Ye C, Moskovits N, Raver-Shapira N, Minsky N, Pirngruber J, Tarcic G, Hublarova P, Moyal L, Gana-Weisz M, Shiloh Y, Yarden Y, Johnsen SA, Vojtesek B, Berger SL, Oren M, 2008, The histone H2B-specific ubiquitin ligase RNF20/hBRE1 acts as a putative tumor suppressor through selective regulation of gene expression. Genes Dev 22(19):2664–2676., DOI 10.1101/gad.1703008 Barber TD, McManus K, Yuen KW, Reis M, Parmigiani G, Shen D, Barrett I, Nouhi Y, Spencer F, Markowitz S, Velculescu VE, Kinzler KW, Vogelstein B, Lengauer C, Hieter P, 2008, Chromatid cohesion defects may underlie chromosome instability in human colorectal cancers. Proc Natl Acad Sci U S A 105(9):3443–3448., DOI 10.1073/pnas. 0712384105 Chernikova SB, Dorth JA, Razorenova OV, Game JC, Brown JM, 2010, Deficiency in Bre1 impairs homologous recombination repair and cell cycle checkpoint response to radiation damage in mammalian cells. Radiat Res 174(5):558–565., DOI 10.1667/ RR2184.1 Baker SJ, Fearon ER,Nigro JM, Hamilton SR, Preisinger AC, Jessup JM, vanTuinen P, Ledbetter DH, Barker DF, Nakamura Y, White R, Vogelstein B, 1989, Chromosome 17 deletions and p53 gene mutations in colorectal carcinomas. Science 244(4901):217–221 Rinaldo C, Moncada A, Gradi A, Ciuffini L, D’Eliseo D, Siepi F, Prodosmo A, Giorgi A, Pierantoni GM, Trapasso F, Guarguaglini G, Bartolazzi A, Cundari E, Schinina ME, Fusco A, Soddu S, 2012, HIPK2 controls cytokinesis and prevents tetraploidization by phosphorylating histone H2B at the midbody. Mol Cell 47(1):87–98., DOI 10.1016/j.molcel. 2012.04.029 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 95 EP 102 DI 10.1007/s12253-015-9977-9 PG 8 ER PT J AU Qingxu, G Yan, Z Jiannan, X Yunlong, L AF Qingxu, Guo Yan, Zhang Jiannan, Xu Yunlong, Liu TI Association Between the Gene Polymorphisms of HDAC9 and the Risk of Atherosclerosis and Ischemic Stroke SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Histone deacetylase 9; Atherosclerosis; Ischemic stroke; Polymorphism ID Histone deacetylase 9; Atherosclerosis; Ischemic stroke; Polymorphism AB Genome-wide association studies have demonstrated various polymorphisms of histone deacetylase 9 (HDAC9) gene was strong risk locus for large-vessel stroke, but the results were controversial. This study aims to replicate the association between the previous detected SNPs of HDAC9 and the susceptibility of ischemic stroke. The study population consisted of 262 consecutive patients diagnosed with ischemic stroke and 300 age and gender matched unrelated controls between October 2012 and October 2014. Rs11984041, rs2389995, and rs2240419 of HDAC9 were genotyped and compared between the cases and controls. The SNP rs11984041 of HDAC9 was found nonpolymorphic in the population involved. The G allele of rs2389995 was found significantly associated with decreased risk of ischemic stroke, no matter with the codominant (AG v.s AA, 0.53 (0.36–0.77), P < 0.001; GG v.s AA, 0.63 (0.27–1.43), P < 0.001), dominant (AG + GG v.s AA, 0.54 (0.38–0.78), P < 0.001), or the recessive model (GG vs AA + AG, 0.75 (0.33–1.71), P < 0.001). On the other hand, The T allele of rs2240419 was found significantly associated with increased risk of ischemic stroke, no matter with the codominant (CT v.s CC, 1.75 (1.22–2.51), P < 0.001; TT v.s CC, 2.67 (1.55–4.61), P < 0.001), dominant (CT + TT v.s CC, 1.93 (1.38–2.71), P < 0.001), or the recessive model (TT vs CC + CT, 2.07 (1.23–3.47), P < 0.001). No linkage disequilibrium was found between rs2389995 and rs2240419 of HDAC9. In conclusion, the present study demonstrated the SNP rs11984041 of HDAC9 was nonpolymorphic in Chinese Han population. The minor G allele of rs2389995 significantly decreased and the minor Tallele of rs2240419 significantly increased the risk of ischemic stroke. C1 [Qingxu, Guo] The General Hospital of Beijing Military Region, Department of Vascular Surgery, No.5 NancangmenBeijing, China. [Yan, Zhang] The General Hospital of Beijing Military Region, Ophthalmology DepartmentBeijing, China. [Jiannan, Xu] Yanjiao People’s Hospital, Department of CardiologyHebei, China. [Yunlong, Liu] The General Hospital of Beijing Military Region, Department of Vascular Surgery, No.5 NancangmenBeijing, China. RP Yunlong, L (reprint author), The General Hospital of Beijing Military Region, Department of Vascular Surgery, Beijing, China. EM liuyunlong_dragon@163.com CR Lozano R, Naghavi M, Foreman K, et al., 2012, Global and regional mortality from235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the global burden of disease study 2010. Lancet 380(9859):2095–2128., DOI 10.1016/S0140-6736(12)61728-0 Ross R, 1999, Atherosclerosis–an inflammatory disease. N Engl J Med 340(2):115–126., DOI 10.1056/NEJM199901143400207 Weber C, Noels H, 2011, Atherosclerosis: current pathogenesis and therapeutic options. Nat Med 17(11):1410–1422., DOI 10.1038/nm. 2538 International Stroke Genetics C, Wellcome Trust Case Control C, Bellenguez C et al. Genome-wide association study identifies a variant in HDAC9 associated with large vessel ischemic stroke. Nat Genet. 2012;44(3):328–33., DOI 10.1038/ng.1081 Consortium CAD, Deloukas P, Kanoni S, et al., 2013, Large-scale association analysis identifies new risk loci for coronary artery disease. Nat Genet 45(1):25–33., DOI 10.1038/ng.2480 Markus HS, Makela KM, Bevan S et al. Evidence HDAC9 genetic variant associated with ischemic stroke increases risk via promoting carotid atherosclerosis. Stroke; J. Cereb. Circ. 2013;44(5):1220–5., DOI 10.1161/STROKEAHA.111.000217 Han Y, SunW,Wang L et al. HDAC9 gene is associated with stroke risk in a Chinese population. Exp. Biol. Med.. 2013;238(7):842–7., DOI 10.1177/1535370213494650. Jia Q, Liu L, Wang Y, 2011, Risk factors and prevention of stroke in the Chinese population. J. Stroke Cereb. Dis. : Off. J. Natl. Stroke Assoc 20(5):395–400., DOI 10.1016/j.jstrokecerebrovasdis.2010.02. 008 Kim YD, Cha MJ, Kim J, et al., 2013, Long-term mortality in patients with coexisting potential causes of ischemic stroke. Int. J. Stroke : Off. J. Int. Stroke Soc., DOI 10.1111/ijs.12013 Dichgans M, Malik R, Konig IR, et al., 2014, Shared genetic susceptibility to ischemic stroke and coronary artery disease: a genome-wide analysis of common variants. Stroke; J. Cereb. Circ 45(1):24–36., DOI 10.1161/STROKEAHA.113.002707 Traylor M, Farrall M, Holliday EG et al. Genetic risk factors for ischaemic stroke and its subtypes, the METASTROKE collaboration): a meta-analysis of genome-wide association studies. Lancet Neurol. 2012;11(11):951–962., DOI 10.1016/S1474-4422(12, 70234-X. Lee JD, Lee TH, Huang YC, et al., 2011, ALOX5AP genetic variants and risk of atherothrombotic stroke in the Taiwanese population. J. Clin. Neurosci.: Off. J. Neurosurg. Soc. Aust 18(12):1634– 1638., DOI 10.1016/j.jocn.2011.03.035 Groenen PM, Vanderlinden G, Devriendt K, Fryns JP, Van de Ven WJ, 1998, Rearrangement of the human CDC5L gene by a t(6;19)(p21;q13.1, in a patient with multicystic renal dysplasia. Genomics 49(2):218–229., DOI 10.1006/geno.1998.5254 Markus HS, 2013, WellcomeTrust genome-wide association study of ischemic stroke. Stroke; J. Cereb. Circ 44(6 Suppl 1):S20–S22., DOI 10.1161/STROKEAHA.112.680652 Haberland M, Montgomery RL, Olson EN, 2009, The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet 10(1):32–42., DOI 10. 1038/nrg2485 Chang S, McKinsey TA, Zhang CL, et al., 2004, Histone deacetylases 5 and 9 govern responsiveness of the heart to a subset of stress signals and play redundant roles in heart development. Mol Cell Biol 24(19):8467–8476., DOI 10.1128/MCB.24.19.8467- 8476.2004 Langley B, Brochier C, Rivieccio MA, 2009, Targeting histone deacetylases as a multifaceted approach to treat the diverse outcomes of stroke. Stroke; J. Cereb. Circ. 40(8):2899–2905., DOI 10. 1161/STROKEAHA.108.540229 Holliday EG, Maguire JM, Evans TJ, et al., 2012, Common variants at 6p21.1 are associated with large artery atherosclerotic stroke. Nat Genet 44(10):1147–1151., DOI 10.1038/ng.2397 Cao Q, Rong S, Repa JJ et al. Histone deacetylase 9 represses cholesterol efflux and alternatively activated macrophages in atherosclerosis development. Arterioscler. Thromb. Vasc. Biol.. 2014;34(9):1871–9., DOI 10.1161/ATVBAHA.114.303393 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 103 EP 107 DI 10.1007/s12253-015-9978-8 PG 5 ER PT J AU Huang, CF Deng, WW Zhang, L Zhang, WF Sun, ZJ AF Huang, Cong-Fa Deng, Wei-Wei Zhang, Lu Zhang, Wen-Feng Sun, Zhi-Jun TI Expression of LC3, LAMP2, KEAP1 and NRF2 in Salivary Adenoid Cystic Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE LC3; LAMP2; KEAP1; NRF2; Salivary adenoid cystic carcinoma ID LC3; LAMP2; KEAP1; NRF2; Salivary adenoid cystic carcinoma AB Salivary Adenoid Cystic Carcinoma (SACC) is a tumor characterized by inevitable local progression and terminal hematogenous metastasis. This study aimed to investigate the expression of LC3, LAMP2, KEAP1 and NRF2 in SACC. Human salivary gland tissue microarray which contains 74 SACC, 12 pleomorphic adenoma and 18 normal salivary gland specimens. High expression of LC3, LAMP2, KEAP1 and NRF2 were found in SACC patients, and LC3, LAMP2, KEAP1 and NRF2 expression were significantly higher in SACC than as compared with pleomorphic adenoma and (or) normal salivary gland. The expression of NRF2 was correlated with pathological type of human SACC (P < 0.05). Moreover, the high-expression of KEAP1 had significant correlations with LC3 (P < 0.001, R = 0.3195), and LAMP2 (P < 0.001, R = 0.3346) and NRF2 (P < 0.05, R = 0.2246) by using the Pearson correlation coefficient test. Our findings demonstrated that up-regulation of LC3, LAMP2, KEAP1 and NRF2 were associated with carcinogenesis and progression of SACC patients, suggesting that they may be useful molecular targets in salivary adenoid cystic carcinoma. C1 [Huang, Cong-Fa] Wuhan University, School and Hospital of Stomatology, The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, 37 Luoyu Road, 430079 Wuhan, China. [Deng, Wei-Wei] Wuhan University, School and Hospital of Stomatology, The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, 37 Luoyu Road, 430079 Wuhan, China. [Zhang, Lu] Wuhan University, School and Hospital of Stomatology, The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, 37 Luoyu Road, 430079 Wuhan, China. [Zhang, Wen-Feng] Wuhan University, School and Hospital of Stomatology, The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, 37 Luoyu Road, 430079 Wuhan, China. [Sun, Zhi-Jun] Wuhan University, School and Hospital of Stomatology, The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, 37 Luoyu Road, 430079 Wuhan, China. RP Zhang, WF (reprint author), Wuhan University, School and Hospital of Stomatology, The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, 430079 Wuhan, China. EM zhangwf59@163.com CR Wang YF, ZhangW, He KF, Liu B, Zhang L, et al., 2014, Induction of autophagy-dependent cell death by the survivin suppressant YM155 in salivary adenoid cystic carcinoma. Apoptosis 19:748– 758 Tang QL, Fan S, Li HG, Chen WL, Shen XM, et al., 2011, Expression of Cyr61 in primary salivary adenoid cystic carcinoma and its relation to Ki-67 and prognosis. Oral Oncol 47:365–370 Munafo DB, Colombo MI, 2001, A novel assay to study autophagy: regulation of autophagosome vacuole size by amino acid deprivation. J Cell Sci 114:3619–3629 Meijer AJ, Dubbelhuis PF, 2004, Amino acid signalling and the integration of metabolism. Biochem Biophys Res Commun 313: 397–403 Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, et al., 2008, Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy 4:151–175 Saha T, 2012, LAMP2A overexpression in breast tumors promotes cancer cell survival via chaperone-mediated autophagy. Autophagy 8:1643–1656 Fehrenbacher N, Bastholm L, Kirkegaard-Sorensen T, Rafn B, Bottzauw T, et al., 2008, Sensitization to the lysosomal cell death pathway by oncogene-induced down-regulation of lysosomeassociated membrane proteins 1 and 2. Cancer Res 68:6623–6633 Miracco C, Cevenini G, Franchi A, Luzi P, Cosci E, et al., 2010, Beclin 1 and LC3 autophagic gene expression in cutaneous melanocytic lesions. Hum Pathol 41:503–512 Tang JY, Hsi E, Huang YC, Hsu NC, Chu PY, et al., 2013, High LC3 expression correlates with poor survival in patients with oral squamous cell carcinoma. Hum Pathol 44:2558–2562 Eskelinen EL, 2011, The dual role of autophagy in cancer. Curr Opin Pharmacol 11:294–300 Valko M, Rhodes CJ, Moncol J, IzakovicM, MazurM, 2006, Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem Biol Interact 160:1–40 Takaya K, Suzuki T, Motohashi H, Onodera K, Satomi S, et al., 2012, Validation of the multiple sensor mechanism of the Keap1- Nrf2 system. Free Radic Biol Med 53:817–827 Leinonen HM, Kansanen E, Polonen P, Heinaniemi M, Levonen AL, 2014, Role of the keap1-nrf2 pathway in cancer. Adv Cancer Res 122:281–320 Huang CF, Zhang L, Ma SR, Zhao ZL, Wang WM, et al., 2013, Clinical significance of Keap1 and Nrf2 in oral squamous cell carcinoma. PLoS One 8: e83479 Liu C, Xu P, Chen D, Fan X, Xu Y, et al., 2013, Roles of autophagy-related genes beclin-1 and LC3 in the development and progression of prostate cancer and benign prostatic hyperplasia. Biomed Rep 1:855–860 Jiang L, Huang S, LiW, ZhangD, Zhang S, et al., 2012, Expression of autophagy and ER stress-related proteins in primary salivary adenoid cystic carcinoma. Pathol Res Pract 208:635–641 Mello AS, Goldim MP, Mezzalira J, Garcia CS, Daitx VV, et al., 2014, LAMP2 as a marker of EBV-mediated B lymphocyte transformation in the study of lysosomal storage diseases. Mol Cell Biochem 385:1–6 Kansanen E, Kuosmanen SM, Leinonen H, Levonen AL, 2013, The Keap1-Nrf2 pathway: mechanisms of activation and dysregulation in cancer. Redox Biol 1:45–49 Suzuki T, Motohashi H, Yamamoto M, 2013, Toward clinical application of the Keap1-Nrf2 pathway. Trends Pharmacol Sci 34: 340–346 Lister A, Nedjadi T, Kitteringham NR, Campbell F, Costello E, et al., 2011, Nrf2 is overexpressed in pancreatic cancer: implications for cell proliferation and therapy. Mol Cancer 10:37 Gonzalez-Polo RA, Boya P, Pauleau AL, Jalil A, Larochette N, et al., 2005, The apoptosis/autophagy paradox: autophagic vacuolization before apoptotic death. J Cell Sci 118:3091–3102 Tanida I, Tanida-Miyake E, Ueno T, Kominami E, 2001, The human homolog of Saccharomyces cerevisiae Apg7p is a proteinactivating enzyme for multiple substrates including human Apg12p, GATE-16, GABARAP, and MAP-LC3. J Biol Chem 276:1701–1706 Gonzalez Y, Aryal B, Chehab L, Rao VA, 2014, Atg7- and Keap1- dependent autophagy protects breast cancer cell lines against mitoquinone-induced oxidative stress. Oncotarget 5:1526–1537 Huber S, Valente S, Chaimbault P, Schohn H, 2014, Evaluation of 2-pioglitazone, an analogue of pioglitazone, on colon cancer cell survival: evidence of drug treatment association with autophagy and activation of the Nrf2/Keap1 pathway. Int J Oncol 45:426–438 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 109 EP 114 DI 10.1007/s12253-015-9981-0 PG 6 ER PT J AU Markow, M Bui, MM Lin, HY Lloyd, M Sexton, JW Dhillon, J AF Markow, Michael Bui, M Marilyn Lin, Hui-Yi Lloyd, Mark Sexton, J Wade Dhillon, Jasreman TI Evaluation of PAX8 Expression and Its Potential Diagnostic and Prognostic Value in Renal and Extra-Renal Ewing Sarcomas/PNETs SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PAX8; PNET; Renal PNET ID PAX8; PNET; Renal PNET AB PAX8 is a transcription factor involved in the regulation of organogenesis of the thyroid gland, kidney, and Mullerian system. It is commonly expressed in epithelial tumors of thyroid and parathyroid glands, kidney, thymus, and female genital tract. PAX8 is increasingly used in the establishment of tissue of origin in carcinomas and has recently been identified in a subset of small blue round cell tumors including Ewing sarcomas/PNETs. However, it is unclear if this association in ES/PNETs is due to renal origin or is PNET specific. In this study we investigated the PAX8 staining pattern of primary renal and extra-renal ES/PNETs to explore its potential diagnostic and prognostic role. A tissue microarray (TMA) of 22 cases of extra-renal Ewing/PNETs and two separate cases of primary renal PNET whole slide sections were immunohistochemically stained with rabbit polyclonal PAX8 antibody. PAX8 was positive in 2 of 2 primary renal PNETs and in 14 (64 %) cases of the extra renal PNETs. The association between PAX8 immunoreactivity and Ewing/PNET was identified in both primary renal and extra-renal Ewing/PNETs for the first time. Further studies are warranted to verify these findings and to shed light in the tumorigenesis of Ewing/ PNET. However, PAX8 is not useful in establishing a diagnosis of Ewing/PNET due to its presence in different tumors like carcinomas, lymphomas and sarcomas. PAX8 does not seem to have prognostic value. C1 [Markow, Michael] University of South Florida, College of Medicine, Department of Pathology and Cell Biology, 12902 Magnolia Drive, 33612 Tampa, FL, USA. [Bui, M Marilyn] University of South Florida, College of Medicine, Department of Pathology and Cell Biology, 12902 Magnolia Drive, 33612 Tampa, FL, USA. [Lin, Hui-Yi] Moffitt Cancer Center, Department of BiostatisticsTampa, FL, USA. [Lloyd, Mark] H. Lee Moffitt Cancer Center and Research Institute, Analytic Microscopy CoreTampa, FL, USA. [Sexton, J Wade] Moffitt Cancer Center and Research Institute, Department of Genitourinary OncologyTampa, FL, USA. [Dhillon, Jasreman] University of South Florida, College of Medicine, Department of Pathology and Cell Biology, 12902 Magnolia Drive, 33612 Tampa, FL, USA. RP Dhillon, J (reprint author), University of South Florida, College of Medicine, Department of Pathology and Cell Biology, 33612 Tampa, USA. EM jasreman.dhillon@moffitt.org CR Plachov D, Chowdhury K, Walther C, Simon D, Guenet JL, Gruss P, 1990, PAX8, a murine paired box gene expressed in the developing excretory system and thyroid gland. Development 110(2): 643–651 Tacha D, Zhou D, Cheng L, 2011, Expression of PAX8 in normal and neoplastic tissues: a comprehensive immunohistochemical study. Appl Immunohistochem Mol Morphol 19(4):293–299 Laury AR, Perets R, Piao H, Krane JF, Barletta JA, French C, Chirieac LR, Lis R, Loda M, Hornick JL, Drapkin R, Hirsch MS, 2011, A comprehensive analysis of PAX8 expression in human epithelial tumors. Am J Surg Pathol 35(6):816–826 Koo J, Mertens RB, Mirocha JM,Wang HL, Dhall D, 2012, Value of islet 1 and PAX8 in identifying metastatic neuroendocrine tumors of pancreatic origin. Mod Pathol 25(6):893–901 Ordonez NG, 2012, Value of PAX8 immunostaining in tumor diagnosis: a review and update. Adv Anat Pathol 19(3):140–151 Tan PH, Cheng L, Rioux-Leclercq N, Merino MJ, Netto G, Reuter VE, Shen SS, Grignon DJ, Montironi R, Eqevad L, Srigley JR, Delahunt B, Moch H, ISUP Renal Tumor Panel, 2013, ISUP renal tumor panel. Renal tumors: diagnostic and prognostic biomarkers. Am J Surg Pathol 37(10):1518–1531 Chang A, Brimo F, Montgomery EA, Epstein JI, 2013, Use of PAX8 and GATA3 in diagnosing sarcomatoid renal cell carcinoma and sarcomatoid urothelial carcinoma. Hum Pathol 44(8):1563– 1568 Carney EM, Banerjee P, Ellis CL, Albadine R, Sharma R, Chaux AM, Burger PC, Netto GJ, 2011, PAX2(−)/PAX8(−)/inhibin A(+, immunoprofile in hemangioblastoma: a helpful combination in the differential diagnosis with metastatic clear cell renal cell carcinoma to the central nervous system. Am J Surg Pathol 35(2):262–267 Doyle LA, Fletcher CD, 2014, Peripheral hemangioblastoma: clinicopathologic characterization in a series of 22 cases. Am J Surg Pathol 38(1):119–127 Fan R, 2014, PAX immunoreactivity in poorly differentiated small round cell tumors of childhood. Fetal Pediatr Pathol 33(4):244–252 Zhao M,Williamson SR, Yu J, XiaW, Li C, Zheng J, ZhuY, Sun K, Wang Z, Cheng L, 2013, PAX8 expression in sporadic hemangioblastoma of the kidney supports a primary renal cell lineage: implications for differential diagnosis. Hum Pathol 44(10): 2247–2255 Surdez D, Benetkiewicz M, Perrin V, Han ZY, Pierron G, Ballet S, Lamoureux F, Redini F, Decouvelaere AV, Daudigeos-Dubus E, Geoerger B, de Pinieux G, Delattre O, Tirode F, 2012, Targeting the EWSR1-FLI1 oncogene-induced protein kinase PKC-β abolishes Ewing sarcoma growth. Cancer Res 72(17):4494–4503 Holmes BJ, Gown AM, Vang R, Ronnett BM, Yemelyanova A, 2014, PAX8 expression in uterine malignant mesodermal mixed tumor, carcinosarcoma). Int J Gynecol Pathol 33(4):42–31 Sangoi AR, Cassarino DS, 2013, PAX-8 expression in primary and metastatic Merkel cell carcinoma: an immunohistochemical analysis. Am J Dermatopathol 35(4):448–451 Morgan EA, Pozdnyakova O, Nascimento AF, Hirsch MS, 2013, PAX8 and PAX5 are differentially expressed in B-cell and T-cell lymphomas. Histopathology 62(3):406–413 Fletcher CD, Bridge JA, Hogendoorn P, Mertens F, 2013, WHO classification of tumours of soft tissue and bone. Fourth Edition. World Health Organization; 468 pp. BuiMM, Han G, Geza A, Reed D, Gonzalez R, Pasha TL, Zhang P, 2011, Connexin 43 is a prognostic biomarker for Ewing Sarcoma, EWS)/primitive neuroectodermal tumor, PNET). Sarcoma 2011: 971050 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 115 EP 120 DI 10.1007/s12253-015-9986-8 PG 6 ER PT J AU Li, H Jiang, Y Pei, F Li, L Yan, B Geng, X Liu, B AF Li, Hui Jiang, Yang Pei, Fenghua Li, Lu Yan, Bingzhu Geng, Xinyu Liu, Bingrong TI Aldehyde Dehydragenase 1 and Nodal as Significant Prognostic Markers in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ALDH1; Nodal; Colorectal cancer; Predictive factors ID ALDH1; Nodal; Colorectal cancer; Predictive factors AB This study aimed to analyze prognostic significance of aldehyde dehydragenase 1 (ALDH1) and Nodal expression in patients with colorectal cancer. ALDH1 and Nodal expressions were observed based on the immunohistochemistry staining from 108 colorectal cancer patients. Scores were given to the staining intensity and percentage of positive cells, and sum of two scores for each case was used to define the groups of ALDH1 and Nodal.We also investigated the protein and mRNA levels of ALDH1 and Nodal by Western blot and qRT-PCR assays. The results were analyzed with the clinicopathologic parameters of these patients. The results indicated that expressions of ALDH1 and Nodal were significantly correlated with the differentiation degree, metastasis, number of tumor positive lymph nodes and AJCC stage. ALDH1 was inclined to express more in the worse differentiated degrees, lymph node metastasis, and worse AJCC stage of colorectal cancer patients. And the expression of Nodal was inversely compared with ALDH1.While the expression of ALDH1 was inversely correlated with the Nodal (r = −0.709, P < 0.01). C1 [Li, Hui] The Second Affiliated Hospital of Harbin Medical University, Department of Gastroenterology, Xuefu Road 246, Nangang District, 150001 Harbin, Heilongjiang, China. [Jiang, Yang] Harbin Medical University, Department of Pathology, Xuefu Road 157, 150081 Harbin, Heilongjiang, China. [Pei, Fenghua] The Second Affiliated Hospital of Harbin Medical University, Department of Gastroenterology, Xuefu Road 246, Nangang District, 150001 Harbin, Heilongjiang, China. [Li, Lu] The Second Affiliated Hospital of Harbin Medical University, Department of Gastroenterology, Xuefu Road 246, Nangang District, 150001 Harbin, Heilongjiang, China. [Yan, Bingzhu] The Second Affiliated Hospital of Harbin Medical University, Department of Infectious Disease, Xuefu Road 246, 150001 Harbin, Heilongjiang, China. [Geng, Xinyu] The Second Affiliated Hospital of Harbin Medical University, Department of Gastroenterology, Xuefu Road 246, Nangang District, 150001 Harbin, Heilongjiang, China. [Liu, Bingrong] The Second Affiliated Hospital of Harbin Medical University, Department of Gastroenterology, Xuefu Road 246, Nangang District, 150001 Harbin, Heilongjiang, China. RP Liu, B (reprint author), The Second Affiliated Hospital of Harbin Medical University, Department of Gastroenterology, 150001 Harbin, China. EM bingrongliu1963@126.com CR Saika K, Sobue T, 2013, Cancer statistics in the world. Gan to Kagaku ryoho 40:2475–2480 Giuliani F, De Vita F, Colucci G, Pisconti S, 2010, Maintenance therapy in colon cancer. Cancer Treat Rev 36(Suppl 3):S42–S45 Hess DA,Wirthlin L, Craft TP, Herrbrich PE, Hohm SA, Lahey R, EadesWC, Creer MH, Nolta JA, 2006, Selection based on CD133 and high aldehyde dehydrogenase activity isolates long-term reconstituting human hematopoietic stem cells. Blood 107:2162– 2169 Dave B, Chang J, 2009, Treatment resistance in stem cells and breast cancer. J Mammary Gland Biol Neoplasia 14:79–82 Jiang F, Qiu Q, Khanna A, Todd NW, Deepak J, Xing L, Wang H, Liu Z, Su Y, Stass SA, Katz RL, 2009, Aldehyde dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer. Mol Cancer Res 7:330–338 Li T, Su Y, Mei Y, Leng Q, Leng B, Liu Z, Stass SA, Jiang F, 2010, ALDH1A1 is a marker for malignant prostate stem cells and predictor of prostate cancer patients' outcome. Lab Investig 90: 234–244 Huang EH, Hynes MJ, Zhang T, Ginestier C, Dontu G, Appelman H, Fields JZ, Wicha MS, Boman BM, 2009, ALDH1 is a marker for normal and malignant human colonic stem cells and track SC overpolution during cancer tumorigenesis. Cancer Res 69:3382– 3389 Schier AF, Shen MM, 2000, Nodal signalling in vertebrate development. Nature 403:385–389 Strizzi L, HardyKM, Kirschmann DA, Ahrlund-Richter L, Hendrix MJ, 2012, Nodal expression and detection in cancer: experience and challenges. Cancer Res 72:1915–1920 Hamilton SR, Vogelstein B,Kudo S, Nakamura S, Hainaut P, Rubio AC, Sobin LH, Fogt F, Winawer SJ, Goldgar DE, Jass JR, 2000, Carcinoma of the colon and rectum. In: Hamilton SR, Aaltonen LA, eds, World Health Organization Classification of Tumors, International Agency for Research on Cancer, IARC). Press, Lyon, pp. 110–111 Li FY, Lai MD, 2009, Colorectal cancer, one entity or three. J Zhejiang Univ Sci B 10:219–229 Ma I, AllanAL, 2011, The role of human aldehyde dehydrogenases in normal and cancer stem cells. Stem Cell Rev 7:292–306 Yasmeen R, Jeyakumar SM, Reichert B, Yang F, Ziouzenkova O, 2012, The contribution of vitamin a to autocrine regulation of fat depots. Biochim Biophys Acta 1821:190–197 Goossens-Beumer IJ, Zeestraten EC, Benard A, Christen T, Reimers MS, Keijzer R, Sier CF, Liefers GJ, Morreau H, Putter H, Vahrmeijer AL, van de Velde CJ, Kuppen PJ, 2014, Clinical prognostic value of combined analysis of Aldh1, survivin, and EpCAM expression in colorectal cancer. Br J Cancer 110:2935–2944 Fitzgerald TL, Rangan S, Dobbs L, Starr S, Sigounas G, 2014, The impact of aldehyde dehydrogenase 1 expression on prognosis for metastatic colon cancer. J Surg Res 192:82–89 Oh SY, Sohn SH, Yim H, Lee D, SuhKW, KimYB, 2015, ALDH1 is a prognostic factor for patients treated with neoadjuvant chemoradiotherapy and radical resection for stage III rectal cancer. J Surg Oncol 111:243–247 Zhou F, Mu YD, Liang J, Liu ZX, Zhou D, NingWL, Li YZ, Ding D, Zhang JF, 2015, Aldehyde dehydrogenase 1: a specific cancer stemcellmarker for human colorectal carcinoma.MolMed Rep 11: 3894–3899 Hou Y, Liu YY, Zhao XK, 2013, Expression of aldehyde dehydrogenase 1 in colon cancer. Asian Pac J Trop Med 6:574–577 Juan H, Hamada H, 2001, Roles of nodal-lefty regulatory loops in embryonic pattering of vertebrates. Genes Cells 6:923–930 Zarzynska JM, 2014, Two faces of TGF-beta1 in breast cancer. Mediat Inflamm 2014:141747 Xu G, Zhong Y, Munir S, Yang BB, Tsang BK, Peng C, 2004, Nodal induces apoptosis and inhibits proliferation in human epithelial ovarian cancer cells via activin receptor-like kinase 7. J Clin Endocrinol Metab 89:5523–5534 Zhong Y, Xu G, Ye G, Lee D, Modica-Amore J, Peng C, 2009, Nodal and activin receptor-like kinase 7 induce apoptosis in human breast cancer cell lines: role of caspase 3. Int J Physiol Pathophysiol Pharmacol 1:83–96 De Silva T, Ye G, Liang YY, Fu G, Xu G, Peng C, 2012, Nodal promotes glioblastoma cell growth. Front Endocrinol, Lausanne, 3:59 GongY, GuoY, Hai Y, YangH, LiuY, YangS,ZhangZ,MaM,Liu L, Li Z, He Z, 2014, Nodal promotes the self-renewal of human colon cancer stem cells via an autocrine manner through Smad2/ 3signaling pathway. Biomed Res Int 2014:364134 Katsuno Y, Ehata S, Yashiro M, Yanagihara K, Hirakawa K, MiyazonoK(2012, Coordinated expression of REG4 and aldehyde dehydrogenase 1 regulating tumourigenic capacity of diffuse-type gastric carcinoma-initiating cells is inhibited by TGF-b. J Pathol 228:391–404 Wang Y, Jiang Y, Tian T, Hori Y, Wada N, Ikeda J, Morii E, 2013, Inhibitory effect of nodal on the expression of aldehyde dehydrogenase 1 in endometrioid adenocarcinoma of uterus. Biochem Biophys Res Commun 440:731–736 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 121 EP 127 DI 10.1007/s12253-015-9984-x PG 7 ER PT J AU Xu, H Seifert, PR Niu, X Li, Y Bui, MM AF Xu, Hairong Seifert, P Robert Niu, Xiaohui Li, Yuan Bui, M Marilyn TI The Establishment and Utility of a Free Online Database of Primary Bone Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Online database; Bone tumors; Establishment; Epidemiology; Tool ID Online database; Bone tumors; Establishment; Epidemiology; Tool AB Primary bone tumors are rare. It is estimated that 3010 individuals (1680 male and 1330 female) were diagnosed with malignancies of bone and joints in 2013 in the United States. [1] The yearly incidence is about 0.9 per 100,000. In comparison, the 2013 incidence of prostate and breast cancer was estimated to be 238,590 and 234,580 respectively. [1] The rarity of this entity together with the non-specific clinical symptoms and complex imaging findings, make this a challenging diagnosis for clinicians. [2–4] However, early and accurate diagnosis is the key to appropriate treatment and better clinical outcome. Primary bone tumors follow a somewhat rigid age-specific distribution. [5, 6] For instance, between the ages of 10– 20 years, there is an incidence peak for osteochondroma and osteosarcoma. [7] Beyond age 40 years, there is a steady increase in chondrosarcoma, myeloma and lymphoma. [7] The use of epidemiological data, including the patient’s age, sex and tumor location, has been the standard 1st approach to establishing a differential diagnosis. This information is augmented by radiological and histological studies to render an accurate diagnosis. However, in many instances, comprehensive epidemiology data can indicate a definite diagnosis before obtaining diagnostic tissue via an invasive procedure. [8, 9] Although epidemiological information on bone and soft tissue tumors can be gathered from literature searches and published sources, an online source would provide greater convenience. In addition, a patient database could provide a real-time and up-to-date source unlike literature and textbook searches. [10–13] To the best of our knowledge, based on Chinese and English literature searches, no such database has so far been described. A database is defined as a data structure composed of organized and interrelated data. They are designed to facilitate the collection of data and support the acquisition of relevant information. [14] Here, we describe our recently built, free, online database of primary bone tumors, including its construction and the main practical utility. This may serve as a reference for users interested in constructing a free online database. It will also be useful for sarcoma physicians and researchers alike. C1 [Xu, Hairong] Peking University, Beijing Ji Shui Tan Hospital, Department of Orthopedic Oncology SurgeryBeijing, China. [Seifert, P Robert] University of South Florida, College of Medicine, Department of Pathology and Cell BiologyTampa, FL, USA. [Niu, Xiaohui] Peking University, Beijing Ji Shui Tan Hospital, Department of Orthopedic Oncology SurgeryBeijing, China. [Li, Yuan] Peking University, Beijing Ji Shui Tan Hospital, Department of Orthopedic Oncology SurgeryBeijing, China. [Bui, M Marilyn] University of South Florida, College of Medicine, Department of Pathology and Cell BiologyTampa, FL, USA. RP Bui, MM (reprint author), University of South Florida, College of Medicine, Department of Pathology and Cell Biology, Tampa, USA. EM Marilyn.Bui@moffitt.org CR Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63:11–30 Franchi A, 2012, Epidemiology and classification of bone tumors. Clin Cases Miner Bone Metab 9:92–95 Limb D, Dreghorn C, Murphy JK, Mannion R, 1994, Primary lymphoma of bone. Int Orthop 18:180–183 Widhe B,Widhe T, 2000, Initial Symptoms and Clinical Features in Osteosarcoma and Ewing Sarcoma*. J Bone Joint Surg Am 82:667 Campanacci M, 1999, Bone and soft tissue tumors: clinical features, imaging, pathology and treatment. Springer Verlag, Wien Unni KK, Inwards CY, 2010, Dahlin's bone tumors: general aspects and data on 10,165 cases. Lippincott Williams & Wilkins, Philadelphia Anfinsen KP, Devesa SS, Bray F, et al., 2011, Age-period-cohort analysis of primary bone cancer incidence rates in the United States, 1976–2005). Cancer Epidemiol Biomark Prev 20:1770–1777 Wu JS and Hochman M. Bone Tumors: A Practical Guide to Imaging. Springer: New York, 2012, p.414. Davies AM, Sundaram M, James SJ, 2009, Imaging of bone tumors and tumor-like lesions: techniques and applications. Springer, New York, p 716 Higgs BW, Elashoff M, Richman S, Barci B, 2006, An online database for brain disease research. BMC Genomics 7:70 Rainville I, Garber JE, 2008, Familial cancer database online. Lancet Oncol 9:925–926 Khan AM, Zaidi SM, Zaffar Bharati MS, 2009, Use of on-line databases by faculty members and research scholars of jawaharlal nehru university, JNU, and jamia millia islamia, JMI), new Delhi, India): a survey. The International Information &. Libr Rev 41:71–78 Castaldi PJ, Cho MH, Cohn M, et al., 2010, The COPD genetic association compendium: a comprehensive online database of COPD genetic associations. Hum Mol Genet 19:526–534 NeufeldML, CornogM(1986, Database history: from dinosaurs to compact discs. J Am Soc Inf Sci 37:183–190 Fletcher CD, Unni KK, Mertens F, 2002, Pathology & genetics: tumours of soft tissue and bone. World Health Organization, Geneva Baena-Ocampo LC, 2009, Ramirez-perez E, linares-gonzalez LM and delgado-chavez R. Epidemiology of bone tumors in Mexico city: retrospective clinicopathologic study of 566 patients at a referral institution. Ann Diagn Pathol 13:16–21 Hogendoorn PCW, Athanasou N, Bielack S, et al., 2010, Bone sarcomas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 21:v204–vv13 Kaatsch P, 2010, Epidemiology of childhood cancer. Cancer Treat Rev 36:277–285 Iarc.Who Classification of Tumours of Soft Tissue and BoneIARC WHO Classification of Tumours SeriesVolume 5 of World Health Organization Classification of Tumours. 4 ed.: World Health Organization: Geneva, 2013, p.427 Chen X, Choi JH, 2010, Designing online collaborative locationaware platform for history learning. J Educ Technol Dev Exch 3: 13–26 Williams B, Damstra D, Stern H, 2012, Professional WordPress: Design and Development. John Wiley & Sons, Hoboken Niu XH, Xu HR, Inwards CY, Li Y, Zhang Q, Ding Y, Letson GD, BuiMM(2015, Primary bone tumors: epidemiological comparison of 9,200 patients treated at Beijing Ji Shui Tan Hospital, Beijing, China with 10, 165 patients at Mayo Clinic, Rochester, MN, USA. Arch Pathol Lab Med 139(9):1149–1155 Junghans TB, Sevin IF, Ionin B, Seifried H, 2004, Cancer information resources: digital and online sources. Toxicology 198:177–193 Lawrence DW, Laflamme L, 2009, Using online databases to find journal articles on injury prevention and safety promotion topics: how do SafetyLit subscribers use other databases? Saf Sci 47:1–8 Blamey RW, Hornmark-Stenstam B, Ball G, et al., 2010, ONCOPOOL - a european database for 16,944 cases of breast cancer. Eur J Cancer 46:56–71 Saracco CM, Roth MA, Wolfson DC, 2004, Enabling distributed enterprise integration with WebSphere and DB2 information integrator. IBM Syst J 43:255–269 Taylor B, Adurty N, Bradley S, Taylor CK, 2010, The official new features guide to sybase ASE 15. Wordware Publishing, Inc., USA Baranyi J, Tamplin ML, 2004, ComBase: a common database on microbial responses to food environments. J Food Prot® 67:1967– 1971 Zawodny JD and Balling DJ. High performance MySQL: optimization, backups, replication, load balancing & more o'reilly: sebastopol; 2008 Jones KM, Farrington P-A, 2011, Using WordPress as a library content management system. American Library Association, Chicago Marcotte E, 2011, Responsive web design, Editions Eyrolles Frain B, 2012, Responsive web design with HTML5 and CSS3. Packt Publishing, UK NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 129 EP 133 DI 10.1007/s12253-015-9982-z PG 5 ER PT J AU Bienkiewicz, J Smolarz, B Malinowski, A AF Bienkiewicz, Jan Smolarz, Beata Malinowski, Andrzej TI Association Between Single Nucleotide Polymorphism +276G > T (rs1501299) in ADIPOQ and Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial cancer; Single nucleotide polymorphism; + 276G>T; Obesity; Adiponectin; ADIPOQ ID Endometrial cancer; Single nucleotide polymorphism; + 276G>T; Obesity; Adiponectin; ADIPOQ AB Current literature gives evidence of an indisputable role adiponectin plays in adipose tissue metabolism and obesity-related diseases. Moreover, latest research efforts focus on linking genetic markers of this adipocytokine’s gene (ADIPOQ) with cancer. Aim of this study was to determine the genotype distribution of single nucleotide polymorphism +276G > T (rs1501299) in ADIPOQ and an attempt to identify the impact this polymorphism exerts on endometrial cancer risk in obese females. The test group comprised 90 women treated surgically for endometrial cancer between 2000 and 2012 in the Department of Surgical & Endoscopic Gynecology and Gynecologic Oncology, Polish Mothers’ Memorial Hospital - Research Institute, Lodz, Poland. 90 individuals treated in the parallel period for uterine fibroids constituted the control group. Patients within both groups were stratified according to BMI into: lean, overweight and obese subjects. Statistical analysis was performed between two major groups and, furthermore, within the abovementioned subgroups. The analysis revealed that allele G of the investigated polymorphism in obese women with endometrial cancer is significantly more frequent, and allele T is significantly less frequent than in lean controls. However, no significant correlation was observed between the polymorphism and endometrial cancer in lean and overweight females. Single nucleotide polymorphism +276G > T (rs1501299) in ADIPOQmay be considered to be a risk factor of endometrial cancer. Further research on SNP in EC is warranted to obtain more conclusive outcomes. C1 [Bienkiewicz, Jan] Polish Mothers’ Memorial Hospital-Research Institute, Department of Surgical & Endoscopic Gynecology and Gynecologic OncologyLodz, Poland. [Smolarz, Beata] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular GeneticsLodz, Poland. [Malinowski, Andrzej] Polish Mothers’ Memorial Hospital-Research Institute, Department of Surgical & Endoscopic Gynecology and Gynecologic OncologyLodz, Poland. RP Bienkiewicz, J (reprint author), Polish Mothers’ Memorial Hospital-Research Institute, Department of Surgical & Endoscopic Gynecology and Gynecologic Oncology, Lodz, Poland. EM jan.a.bienkiewicz@gmail.com CR Siegel R, Naishadham D, Jemal A, et al., 2012, Cancer statistics. CA Cancer J Clin 62(1):10–29 Bokhman JV, 1983, Two pathogenetic types of endometrial carcinoma. Gynecol Oncol 15(1):10–17 Crosbie EJ, Zwahlen M, Kitchener HC, et al., 2010, Body mass index, hormone replacement therapy, and endometrial cancer risk: a meta-analysis. Cancer Epidemiol Biomark Prev 19(12):3119–3130 Bhaskaran K, Douglas I, Forbes H, et al. Body-mass index and risk of 22 specific cancers: a population-based cohort study of 5·24 million UK adults. Lancet. 2014 Aug 30;384(9945):755–765. SGO Clinical Practice Endometrial Cancer Working Group, Burke WM, Orr J, et al., 2014, Endometrial cancer: a review and current management strategies: part I. Gynecol Oncol 134(2):385–392 Calle EE, Rodriguez C, Walker-Thurmond K, et al., 2003, Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med 348(17):1625–1638 Friberg E, Mantzoros CS, Wolk A, 2007, Diabetes and risk of endometrial cancer: a population-based prospective cohort study. Cancer Epidemiol Biomark Prev 16(2):276–280 Hecht J, Mutter GL, 2006, Molecular and pathologic aspects of endometrial carcinogenesis. J Clin Oncol 24(29):4783–4791 Tilg H, 2006)Moschen AR adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol 6(10):772–783 Nigro E, Scudiero O, Monaco ML, et al., 2014, New insight into adiponectin role in obesity and obesity-related diseases. Biomed Res Int 2014:658913 Fisman EZ, Tenenbaum A, 2014, Adiponectin: a manifold therapeutic target for metabolic syndrome, diabetes, and coronary Disease? Cardiovasc Diabetol 13:103 Di Chiara T, Argano C, Corrao S, e t a l ., 2012, Hypoadiponectinemia: a link between visceral obesity and metabolic syndrome. J Nutr Metab 2012:175245 Chandran M, Phillips SA, Ciaraldi T, et al., 2003, Adiponectin: more than just another fat cell hormone? Diabetes Care 26(8): 2442–2450 Gu HF, 2009, Biomarkers of adiponectin: plasma protein variation and genomic DNA polymorphisms. Biomark Insights 4:123–133 Melistas L, Mantzoros CS, Kontogianni M, et al., 2009, Association of the +45 T > G and +276G > T polymorphisms in the adiponectin gene with insulin resistance in nondiabetic Greek women. Eur J Endocrinol 161(6):845–852 Stumvoll M, Tschritter O, Fritsche A, et al., 2002, Association of the T-G polymorphism in adiponectin, exon 2, with obesity and insulin sensitivity: interaction with family history of type 2 diabetes. Diabetes 51(1):37–41 Chu H, Wang M, Zhong D, et al., 2013, AdipoQ polymorphisms are associated with type 2 diabetes mellitus: a meta-analysis study. Diabetes Metab Res Rev 29(7):532–545 Teras LR, Goodman M, Patel AV, et al., 2009, No association between polymorphisms in LEP, LEPR, ADIPOQ, ADIPOR1, or ADIPOR2 and postmenopausal breast cancer risk. Cancer Epidemiol Biomark Prev 18(9):2553–2557 Ye C,Wang J, Tan, et al., 2013)Meta-analysis of adiponectin polymorphisms and colorectal cancer risk. Int J Med Sci 10(9):1113– 1120 Dhillon PK, Penney KL, Schumacher F, et al., 2011, Common polymorphisms in the adiponectin and its receptor genes, adiponectin levels and the risk of prostate cancer. Cancer Epidemiol Biomark Prev 20(12):2618–2627 Chen X, Xiang YB, Long JR, et al., 2012, Genetic polymorphisms in obesity-related genes and endometrial cancer risk. Cancer 118(13):3356–3364 Meyer LA, Westin SN, Lu KH, et al., 2008, Genetic polymorphisms and endometrial cancer risk. Expert Rev Anticancer Ther 8(7):1159–1167 Fan HJ, Wen ZF, Xu BL, et al., 2013, Three adiponectin rs1501299G/T, rs822395A/C, and rs822396A/G polymorphisms and risk of cancer development: a meta-analysis. Tumour Biol 34(2):769–778 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 135 EP 138 DI 10.1007/s12253-015-9985-9 PG 4 ER PT J AU Fekete, F Fadgyas, B Papp, Szilagyi, Prohaszka, Z Muller, B Kovacs, G AF Fekete, Ferenc Fadgyas, Balazs Papp, Eva Szilagyi, Agnes Prohaszka, Zoltan Muller, Brigitta Kovacs, Gabor TI The role of mannose binding lectin on fever episodes in pediatric oncology patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MBL; Polymorphism; Febrile neutropenia; Oncohematology ID MBL; Polymorphism; Febrile neutropenia; Oncohematology AB Despite significant changes in pediatric oncological therapy, mortality is still high, mainly due to infections. Complement system as an ancient immune defense against microorganisms plays a significant role in surmounting infections, therefore, deficiency of its components may have particular importance in malignancies. The present paper assesses the effect of promoter (X/Y) and exon 1 (A/0) polymorphisms of the MBL2 gene altering mannose binding lectin (MBL) serum level in pediatric oncological patients with febrile neutropenia. Furthermore, frequency distribution of MBL2 alleles in children with malignancies and age-matched controls was analysed. Fifty-four oncohematological patients and 53 children who had undergone pediatric surgery were enrolled into this retrospective study. No significant differences were found in the frequency of MBL2 alleles between the hematooncologic and control group. The average duration of fever episodes was significantly shorter (p = 0.035) in patients carrying genotypes (AY/AY and AY/AX) that encode normal MBL level, compared to individuals with genotypes associated with lower functional MBL level (AX/AX, AY/0, AX/0, or 0/0) (days, median (IQ range) 3.7(0–5.4) vs. 5.0(3.8–6.6), respectively). In conclusion, our data suggest that MBL2 genotypes may influence the course of febrile neutropenia in pediatric patients with malignancies, and may contribute to clarification of the importance of MBL in infections. C1 [Fekete, Ferenc] Heim Pal Children's Hospital, Department of Hematology, Madarasz Street Building, Madarasz utca 22-24, 1131 Budapest, Hungary. [Fadgyas, Balazs] Heim Pal Children’s Hospital, Department of Pediatric Surgery and TraumatologyBudapest, Hungary. [Papp, Eva] Gyula Nyiro Hospital, National Institute of Psychiatry and AddictionsBudapest, Hungary. [Szilagyi, Agnes] Semmelweis University, 3rd Department of Internal MedicineBudapest, Hungary. [Prohaszka, Zoltan] Semmelweis University, 3rd Department of Internal MedicineBudapest, Hungary. [Muller, Brigitta] Heim Pal Children's Hospital, Department of Hematology, Madarasz Street Building, Madarasz utca 22-24, 1131 Budapest, Hungary. [Kovacs, Gabor] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. RP Fekete, F (reprint author), Heim Pal Children's Hospital, Department of Hematology, 1131 Budapest, Hungary. EM efekete@heimpalkorhaz.hu CR Walport MJ, 2001, Complement Second of two parts. N Engl J Med 344:1140–1144 Walport MJ, 2001, Complement First of two parts. N Engl J Med 344:1058–1066 Neth O, Jack DL, Dodds AW, et al., 2000, Mannose-binding lectin binds to a range of clinically relevantmicroorganisms and promotes complement deposition. Infect Immun 68:688–693 Garred P, 2008, Mannose-binding lectin genetics: from a to Z. Biochem Soc Trans 36:1461–1466 Garred P, Larsen F, Madsen HO, et al., 2003, Mannose-binding lectin deficiency–revisited. Mol Immunol 40:73–84 Bouwman, LH, Roep, BO & Roos,, 2006, A Mannose-binding lectin: clinical implications for infection, transplantation, and autoimmunity. Hum Immunol. 67:247–256. Ruskamp JM, Hoekstra MO, Rovers MM, et al., 2006,, 2006, mannose-binding lectin and upper respiratory tract infections in children and adolescents: a review. Arch Otolaryngol Head Neck Surg 132:482–486 Koutsounaki E, Goulielmos GN, Koulentaki M, et al., 2008, Mannose-binding lectin MBL2 gene polymorphisms and outcome of hepatitis C virus-infected patients. J Clin Immunol 28:495–500 Garred P, S J,J, Quist L, et al, 2003, Association of mannosebinding lectin polymorphisms with sepsis and fatal outcome, in patients with systemic inflammatory response syndrome. J Infect Dis 188:1394–1403 Schmiegelow K, Garred P, Lausen B, et al., 2002, Increased frequency of mannose-binding lectin insufficiency among children with acute lymphoblastic leukemia. Blood 100:3757–3760 Neth O, Hann I, Turner MW, et al., 2001, Deficiency of mannosebinding lectin and burden of infection in children with malignancy: a prospective study. Lancet 358:614–618 Ghazi M, Isadyar M, Gachkar L, et al., 2012, Serum levels of mannose-binding lectin and the risk of infection in pediatric oncology patients with chemotherapy. J Pediatr Hematol Oncol 34:128– 130 Horiuchi T, Gondo H, Miyagawa H, et al., 2005, Association of MBL gene polymorphismswithmajor bacterial infection in patients treated with high-dose chemotherapy and autologous PBSCT. Genes Immun 6:162–166 Peterslund NA, Koch C, Jensenius JC, et al., 2001, Association between deficiency ofmannose-binding lectin and severe infections after chemotherapy. Lancet 358:637–638 Vekemans M, Robinson J, Georgala A, et al., 2007, Low mannosebinding lectin concentration is associated with severe infection in patients with hematological cancer who are undergoing chemotherapy. Clin Infect Dis 44:1593–1601 Frakking FN, Brouwer N, Dolman KM, et al., 2011, Mannosebinding lectin, MBL, as prognostic factor in paediatric oncology patients. Clin Exp Immunol 165:51–59 Bergmann OJ, Christiansen M, Laursen I, et al., 2003, Low levels of mannose-binding lectin do not affect occurrence of severe infections or duration of fever in acute myeloid leukaemia during remission induction therapy. Eur J Haematol 70:91–97 Lausen B, Schmiegelow K, Andreassen B, et al., 2006, Infections during induction therapy of childhood acute lymphoblastic leukemia– no association to mannose-binding lectin deficiency. Eur J Haematol 76:481–487 Martinez-Lopez J, Rivero A, Rapado I, et al., 2009, Influence of MBL-2 mutations in the infection risk of patients with follicular lymphoma treated with rituximab, fludarabine, and cyclophosphamide. Leuk Lymphoma 50:1283–1289 Rubnitz JE, Howard SC, Willis J, et al., 2008, Baseline mannose binding lectin levels may not predict infection among children with leukemia. Pediatr Blood Cancer 50:866–868 Zehnder, A, Fisch, U, Hirt, A, et al., 2009, prognosis in pediatric hematologic malignancies is associated with serum concentration of mannose-binding lectin-associated serine protease-2, MASP-2). Pediatr Blood Cancer. 2009;53: 53–57. Frakking FN, Israels J, Kremer LC, et al., 2011, Mannose-binding lectin, MBL, and the risk for febrile neutropenia and infection in pediatric oncology patients with chemotherapy. Pediatr Blood Cancer 57:89–96 Bang P, Laursen I, Thornberg K, et al., 2008,, 2008, the pharmacokinetic profile of plasma-derived mannan-binding lectin in healthy adult volunteers and patients with Staphylococcus aureus septicaemia. Scand J Infect Dis 40: 44–48 Frakking FN, Brouwer N, van de Wetering MD, et al., 2009, Safety and pharmacokinetics of plasma-derived mannose-binding lectin, MBL, substitution in children with chemotherapy-induced neutropaenia. Eur J Cancer 45:505–512 Valdimarsson H, 2003, Infusion of plasma-derived mannan-binding lectin, MBL, into MBL-deficient humans. Biochem Soc Trans 31:768–769 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 139 EP 143 DI 10.1007/s12253-015-9992-x PG 5 ER PT J AU Jamdade, SV Mundhe, AN Kumar, P Tadla, V Lahkar, M AF Jamdade, Sudhir Vinayak Mundhe, A Nitin Kumar, Parveen Tadla, Venkatesh Lahkar, Mangala TI Raloxifene Inhibits NF-kB Pathway and Potentiates Anti-Tumour Activity of Cisplatin with Simultaneous Reduction in its Nephrotoxictiy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cisplatin; Raloxifene; Nephrotoxicity; Inflammation; Pro-inflammatory cytokines ID Cisplatin; Raloxifene; Nephrotoxicity; Inflammation; Pro-inflammatory cytokines AB Cisplatin induced nephrotoxicity is the chief obstacle in the use of cisplatin as chemotherapeutic agent. However, it remains as most widely employed anticancer agent to treat various solid tumours like head-neck, testicular, ovarian and mammary gland cancer. Raloxifene is claimed to be potent anti-inflammatory as well as anti-cancer agent. The present study was carried out to explore the effect of pre-treatment of raloxifene on cisplatin induced nephrotoxicity and its antitumour activity in 7, 12 dimethyl benz [a] anthracene induced mammary tumour in animal model. Renal damage was accessed by measuring serum level of creatinine, blood urea nitrogen and albumin whereas systemic inflammation was accessed by measuring level of pro-inflammatory cytokines like tumour necrosis factor alpha (TNF-α), interleukin 6 (IL-6), interleukin 10 (IL-10) and nuclear factor kappa B (NFκB). Moreover, assessment of tumour reduction was done by measuring tumour volume and percentage tumour reduction. A single dose of cisplatin (7.5 mg/kg) resulted in significant increase in serum creatinine, blood urea nitrogen, NF-kB, TNF-α and IL-6 levels along with decrease in albumin and IL-10 levels. However, there were no significant changes in raloxifene (8 mg/kg) treated group. Pre-treatment of raloxifene (8 mg/kg) caused marked decrease in serum creatinine, blood urea nitrogen, TNF-α and IL-6 levels whereas increase in albumin and IL-10 levels. However, pre-treatment of raloxifene showed maximum tumour reduction as compared to cisplatin and raloxifene treated groups. The present study demonstrates that raloxifene potentiates anti-tumour activity of cisplatin with simultaneous reduction in its nephrotoxicity, and this effect is attributed to its direct anti-inflammatory activity. C1 [Jamdade, Sudhir Vinayak] Gauhati Medical College, National Institute of Pharmaceutical Education and Research (NIPER), Department of Pharmacology and Toxicology, Laboratory of Molecular Pharmacology and Toxicology, 781032 Guwahati, Assam, India. [Mundhe, A Nitin] Gauhati Medical College, National Institute of Pharmaceutical Education and Research (NIPER), Department of Pharmacology and Toxicology, Laboratory of Molecular Pharmacology and Toxicology, 781032 Guwahati, Assam, India. [Kumar, Parveen] Gauhati Medical College, National Institute of Pharmaceutical Education and Research (NIPER), Department of Pharmacology and Toxicology, Laboratory of Molecular Pharmacology and Toxicology, 781032 Guwahati, Assam, India. [Tadla, Venkatesh] Gauhati Medical College, National Institute of Pharmaceutical Education and Research (NIPER), Department of Pharmacology and Toxicology, Laboratory of Molecular Pharmacology and Toxicology, 781032 Guwahati, Assam, India. [Lahkar, Mangala] Gauhati Medical College, National Institute of Pharmaceutical Education and Research (NIPER), Department of Pharmacology and Toxicology, Laboratory of Molecular Pharmacology and Toxicology, 781032 Guwahati, Assam, India. RP Jamdade, SV (reprint author), Gauhati Medical College, National Institute of Pharmaceutical Education and Research (NIPER), Department of Pharmacology and Toxicology, Laboratory of Molecular Pharmacology and Toxicology, 781032 Guwahati, India. EM vinayak757@gmail.com CR Bogdanovic G, Kojic V, Srdic T, Jakimov D, Djuran MI, Bugarcic ZD, Baltic M, Baltic VV, 2002, Growth effects of some platinum(II, complexes with sulfur-containing carrier ligands on MCF7 human breast cancer cell line upon simultaneous administration with taxol. Met Based Drugs 9:33–43 Aisner J, Jacobs M, Sinabaldi V, Gray W, Eisenberger M, 1994, Chemoradiotherapy for the treatment of regionally advanced head and neck cancers. Semin Oncol 21:35–44 Kelland L, 2007, The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer 7:573–584 Ali BH, Al Moundhri MS, 2006, Agents ameliorating or augmenting the nephrotoxicity of cisplatin and other platinum compounds: a review of some recent research. Food Chem Toxicol 44:1173–1183 Ramesh G, Kimball SR, Jefferson LS, Reeves WB, 2007, Endotoxin and cisplatin synergistically stimulate TNF-alpha production by renal epithelial cells. Am J Physiol Renal Physiol 292: F812–F819 Basnakian AG, Apostolov EO, Yin X, Napirei M, Mannherz HG, Shah SV, 2005, Cisplatin nephrotoxicity is mediated by deoxyribonuclease I. J Am Soc Nephrol 16:697–702 Arany I, Safirstein RL, 2003, Cisplatin nephrotoxicity. Semin Nephrol 23:460–464 Faubel S, Lewis EC, Reznikov L, Ljubanovic D, Hoke TS, Somerset H, Oh DJ, Lu L, Klein CL, Dinarello CA, Edelstein CL, 2007, Cisplatin-induced acute renal failure is associated with an increase in the cytokines interleukin, IL)-1beta, IL-18, IL-6, and neutrophil infiltration in the kidney. J Pharmacol Exp Ther 322:8– 15 Ramesh G, Brian Reeves W, 2006, Cisplatin increases TNF-alpha mRNA stability in kidney proximal tubule cells. Ren Fail 28:583– 592 Yao X, Panichpisal K, Kurtzman N, Nugent K, 2007, Cisplatin nephrotoxicity: a review. Am J Med Sci 334:115–124 Ramesh G, ReevesWB(2002, TNF-alphamediates chemokine and cytokine expression and renal injury in cisplatin nephrotoxicity. J Clin Invest 110:835–842 Ramesh G, Reeves WB, 2003, TNFR2-mediated apoptosis and necrosis in cisplatin-induced acute renal failure. Am J Physiol Renal Physiol 285:F610–F618 Ramesh G, ReevesWB, 2004, Salicylate reduces cisplatin nephrotoxicity by inhibition of tumour necrosis factor-alpha. Kidney Int 65:490–499 Dong Z, Atherton SS, 2007, Tumour necrosis factor-alpha in cisplatin nephrotoxicity: a homebred foe? Kidney Int 72:5–7 Kaya H, Ozkaya O, SezikM, Arslanoglu E,Yilmaztepe A, Ulukaya E, 2005, Effects of raloxifene on serum malondialdehyde, erythrocyte superoxide dismutase, and erythrocyte glutathione peroxidase levels in healthy postmenopausal women. Maturitas 50:182–188 Lippuner K, Buchard PA, De Geyter C, Imthurn B, Lamy O, Litschgi M, Luzuy F, Schiessl K, Stute P, Birkhauser M, 2012, Recommendations for raloxifene use in daily clinical practice in the Swiss setting. Eur Spine J 21:2407–2417 Shibata MA, Morimoto J, Shibata E, Kurose H, Akamatsu K, Li ZL, Kusakabe M, Ohmichi M, Otsuki Y, 2010, Raloxifene inhibits tumour growth and lymph node metastasis in a xenograft model of metastatic mammary cancer. BMC Cancer 10:566 Galien R, Garcia T, 1997, Estrogen receptor impairs interleukin-6 expression by preventing protein binding on the NF-kappaB site. Nucleic Acids Res 25:2424–2429 Olivier S, Close P, Castermans E, de Leval L, Tabruyn S, Chariot A, Malaise M, Merville MP, Bours V, Franchimont N, 2006, Raloxifene-induced myeloma cell apoptosis: a study of nuclear factor-kappaB inhibition and gene expression signature. Mol Pharmacol 69:1615–1623 K.K. Thakur, N.B. Bolshette, C. Trandafir, V.S. Jamdade, A. Istrate, R. Gogoi, A. Cucuianu, Role of toll-like receptors in multiple myeloma and recent advances. Exp Hematol, 2014, 43(3):158–167 Kumar P, Bolshette NB, Jamdade VS, Mundhe NA, Thakur KK, Saikia KK, LahkarM(2013, Breast cancer status in India: an overview. J Carcinog 3:177–183 Cheung J, Mak YT, Papaioannou S, Evans BA, Fogelman I, Hampson G, 2003, Interleukin-6, IL-6), IL-1, receptor activator of nuclear factor kappaB ligand, RANKL, and osteoprotegerin production by human osteoblastic cells: comparison of the effects of 17-beta oestradiol and raloxifene. J Endocrinol 177:423–433 Deng J, Kohda Y, Chiao H, Wang Y, Hu X, Hewitt SM, Miyaji T, McLeroy P, Nibhanupudy B, Li S, Star RA, 2001, Interleukin-10 inhibits ischemic and cisplatin-induced acute renal injury. Kidney Int 60:2118–2128 Kalaitzidis D, Gilmore TD, 2005, Transcription factor cross-talk: the estrogen receptor and NF-kappaB. Trends Endocrinol Metab 16:46–52 Kumar P, Kadakol A, Shasthrula P, Mundhe NA, Jamdade VS, Barua CC, Gaikwad AB, 2015, Curcumin as an adjuvant to breast cancer treatment. Anti Cancer Agents Med Chem 15:647–656 T. Whitsett, M. Carpinter, C.A. Lamartiniere, 2006, Resveratrol, but not EGCG, in the diet suppresses DMBA-induced mammary cancer in rats. J Carcinog 5 15. Stuhr LE, Iversen VV, Straume O, Maehle BO, Reed RK, 2004, Hyperbaric oxygen alone or combined with 5-FU attenuates growth ofDMBA-induced ratmammary tumors. Cancer Lett 210(1):35–40 Tsuruya K, Ninomiya T, Tokumoto M, Hirakawa M, Masutani K, TaniguchiM, Fukuda K, Kanai H, Kishihara K, Hirakata H, IidaM, 2003, Direct involvement of the receptor-mediated apoptotic pathways in cisplatin-induced renal tubular cell death. Kidney Int 63: 72–82 Liu Y, Webb HK, Fukushima H, Micheli J, Markova S, Olson JL, Kroetz DL, 2012, Attenuation of cisplatin-induced renal injury by inhibition of soluble epoxide hydrolase involves nuclear factor kappaB signaling. J Pharmacol Exp Ther 341:725–734 Miller RP, Tadagavadi RK, Ramesh G, Reeves WB, 2010, Mechanisms of cisplatin nephrotoxicity. Toxins, Basel, 2:2490– 2518 Schrier RW, 2002, Cancer therapy and renal injury. J Clin Invest 110:743–745 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 145 EP 153 DI 10.1007/s12253-015-9988-6 PG 9 ER PT J AU Hutoczki, G Bognar, L Toth, J Scholtz, B Zahuczky, G Hanzely, Z Csosz, Remenyi-Puskar, J Kallo, G Hortobagyi, T Klekner, AF Hutoczki, Gabor Bognar, Laszlo Toth, Judit Scholtz, Beata Zahuczky, Gabor Hanzely, Zoltan Csosz, Eva Remenyi-Puskar, Judit Kallo, Gergo Hortobagyi, Tibor Klekner, Almos TI Effect of Concomitant Radiochemotherapy on Invasion Potential of Glioblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Radiochemotherapy; Temozolomide; Extracellular matrix; Invasion; Glioblastoma ID Radiochemotherapy; Temozolomide; Extracellular matrix; Invasion; Glioblastoma AB Glioblastoma (GBM) is the most common primary brain tumor in adults with inevitable recurrence after oncotherapy. The insufficient effect of "gold standard" temozolomide-based concomitant radiochemotherapy may be due to the inability to prevent tumor cell invasion. Peritumoral infiltration depends mainly on the interaction between extracellular matrix (ECM) components and cell membrane receptors. Changes in invasive behaviour after oncotherapy can be evaluated at the molecular level by determining the RNA expression and protein levels of the invasionrelated ECM components. The expression of nineteen ECM molecules was determined at both RNA and protein levels in thirty-one GBM samples. Fifteen GBM samples originated from the first surgical procedure on patients before oncotherapy, and sixteen GBM samples were collected at the second surgery due to local recurrence after concomitant chemoirradiation. RNA expressions were measured with qRTPCR, and protein levels were determined by quantitative analysis of Western blots. Only MMP-9 RNA transcript level was reduced (p < 0.05) whereas at protein level, eight molecules showed changes concordant with RNA expression with significant decrease in brevican only. The results suggest that concomitant radiochemotherapy does not have sufficient impact on the expression of invasion-related ECM components of glioblastoma, oncotherapy does not significantly affect its invasive behavior. To avoid the spread of tumors into the brain parenchyma, supplementation of antiproliferative treatment with anti-invasive agents may be worth consideration in oncotherapy for glioblastoma. C1 [Hutoczki, Gabor] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Bognar, Laszlo] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Toth, Judit] University of Debrecen, Department of Oncology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Scholtz, Beata] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular Biology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Zahuczky, Gabor] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular Biology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Hanzely, Zoltan] National Institute of Clinical Neurosciences, Amerikai ut 57, 1145 Budapest, Hungary. [Csosz, Eva] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular Biology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Remenyi-Puskar, Judit] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Kallo, Gergo] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular Biology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Hortobagyi, Tibor] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Klekner, Almos] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98, 4032 Debrecen, Hungary. RP Bognar, L (reprint author), University of Debrecen, Clinical Center, Department of Neurosurgery, 4032 Debrecen, Hungary. EM neurosurgery.debrecen@freemail.hu CR Murnyak B, Csonka T, Hegyi K, Mehes G, Klekner A, Hortobagyi T, 2013, Occurrence and molecular pathology of high grade gliomas., in Hungarian). Ideggyogy Sz 66:312–321 Gurney JG, Kadan-Lottick N, 2001, Brain and other central nervous system tumors: rates, trends, and epidemiology. Curr Opin Oncol 13(3):160–166 Chaichana KL, Zadnik P, Weingart JD, Olivi A, Gallia GL, Blakeley J, Lim M, Brem H, Quinones-Hinojosa A, 2013, Multiple resections for patients with glioblastoma: prolonging survival. J Neurosurg 118(4):812–820 Stupp R, Mason WP, van den Bent MJ, 2005, Radiotherapy plus concomitant and adjuvant temozolimide for glioblastoma. N Engl J Med 352:987–996 Jain RK, 2013, Normalizing tumor microenvironment to treat cancer: bench to bedside to biomarkers. J Clin Oncol 31(17):2205– 2218 Petras M, Hutoczki G, Varga I, Vereb G, Szollosi J, Bognar L, Ruszthi P, Kenyeres A, Toth J, Hanzely Z, Scholtz B, Klekner A, 2009, Expression pattern of invasion-related molecules in brain tumors of different origin. Magy Onkol 53(3):253–258 Klekner A, Varga I, Bognar L, Hutoczki G, Kenyeres A, Toth J, Hanzely Z, Scholtz B, 2010, Extracellular matrix of cerebral tumors with different invasiveness. Ideggyogy Sz 63(1–2):38–43 Varga I, Hutoczki G, Petras M, Scholtz B, Miko E, Kenyeres A, Toth J, Zahuczky G, Bognar L, Hanzely Z, Klekner A, 2010, Expression of invasion-related extracellular matrix molecules in human glioblastoma versus intracerebral lung adenocarcinoma metastasis. Cen Eur Neurosurg 71(4):173–180 Varga I, Hutoczki G, Szemcsak CD, Zahucky G, Toth J, Adamecz Z, Kenyeres A, Bognar L, Hanzely Z, Klekner A, 2012, Brevican, neurocan, tenascin-C and versican are mainly responsible for the invasiveness of low-grade astrocytoma. Pathol Oncol Res 18(2): 413–420 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−delta delta C(T), method. Methods 25:402–408 Bradford MM, 1976, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72:248–254 Iuga C, Seicean A, Iancu C, Buiga R, Sappa PK, Volker U, Hammer E, 2014, Proteomic identification of potential prognostic biomarkers in resectable pancreatic ductal adenocarcinoma. Proteomics 14(7–8):945–955 Dogan A, 2014, Advances in clinical applications of tissue proteomics: opportunities and challenges. Expert Rev Proteomics 11(5): 531–533 Lange V, Picotti P, Domon B, Aebersold R, 2008, Selected reaction monitoring for quantitative proteomics: a tutorial. Mol Syst Biol 4: 222 Blankley RT, Fisher C, Westwood M, North R, Baker PN, Walker MJ,Williamson A,Whetton AD, LinW, McCowan L, Roberts CT, Cooper GJ, Unwin RD, Myers JE, 2013, A label-free selected reaction monitoring workflow identifies a subset of pregnancy specific glycoproteins as potential predictive markers of early-onset pre-eclampsia. Mol Cell Proteomics 12(11):3148–3159 Martinez-Aguilar J,Molloy MP, 2013, Label-free selected reaction monitoring enables multiplexed quantitation of S100 protein isoforms in cancer cells. J Proteome Res 12(8):3679–3688 Andersen AP, 1978, Postoperative irradiation of glioblastomas. Results in a randomized series. Acta Radiol Oncol Radiat Phys Biol 17(6):475–484 Nieder C, Andratschke N, Wiedenmann N, Busch R, Grosu AL, MollsM(2004, Radiotherapy for high-grade Gliomas. Does altered fractionation improve the Outcome? Strahlenther Onkol 180(7): 401–7. Souhami L, Seiferheld W, Brachman D, Podgorsak EB, Werner- Wasik M, Lustig R, Schultz CJ, Sause W, Okunieff P, Buckner J, Zamorano L, Mehta MP, Curran Jr WJ, 2004, Randomized comparison of stereotactic radiosurgery followed by conventional radiotherapy with carmustine to conventional radiotherapy with carmustine for patients with glioblastoma multiforme: report of radiation therapy oncology group 93–05 protocol. Int J Radiat Oncol Biol Phys 60(3):853–860 Shirazi HA, Grimm S, Raizer J, Mehta MP, 2011, Combined modality approaches in the management of adult glioblastoma. Front Oncol 1:36 Newlands ES, StevensMFG,Wedge SR, Wheelhouse RT, Brock C, 1997, Temozolomide: a review of its discovery, chemical properties, pre-clinical development and clinical trials. Cancer Treat Rev 23:35–61 Mannas JP, Lightner DD, Defrates SR, Pittman T, Villano JL, 2014, Long-term treatment with temozolomide in malignant glioma. J Clin Neurosci 21(1):121–123 Garside R, Pitt M, Anderson R, Rogers G, Dyer M, Mealing S, Somerville M, Price A, Stein K, 2007, The effectiveness and cost-effectiveness of carmustine implants and temozolomide for the treatment of newly diagnosed high-grade glioma: a systematic review and economic evaluation. Health Technol Assess 11(45):IIIIV– IX-221 Koukourakis GV, Kouloulias V, Zacharias G, Papadimitriou C, Pantelakos P, Maravelis G, Fotineas A, Beli I, Chaldeopoulos D, Kouvaris J, 2009, Temozolomide with radiation therapy in high grade brain gliomas: pharmaceuticals considerations and efficacy; a review article. Molecules 14(4):1561–1577 Bobola MS, Kolstoe DD, Blank A, Silber JR, 2010, Minimally cytotoxic doses of temozolomide produce radiosensitization in human glioblastoma cells regardless of MGMT expression. Mol Cancer Ther 9(5):1208–1218 Vehlow A, Cordes N, 2013, Invasion as target for therapy of glioblastoma multiforme. Biochim Biophys Acta 1836(2):236–244 Zhao WJ, Zhang W, Li GL, Cui Y, Shi ZF, Yuan F, 2012, Differential expression of MMP-9 and AQP4 in human glioma samples. Folia Neuropathol 50(2):176–186 Trog D, Yeghiazaryan K, Fountoulakis M, Friedlein A, Moenkemann H, Haertel N, Schueller H, Breipohl W, Schild H, Leppert D, Golubnitschaja O, 2006, Pro-invasive gene regulating effect of irradiation and combined temozolomide-radiation treatment on surviving human malignant glioma cells. Eur J Pharmacol 542(1–3):8–15 Gary SC, Kelly GM, Hockfield S, 1998, BEHAB/brevican: a brain-specific lectican implicated in gliomas and glial cell motility. Curr Opin Neurobiol 8(5):576–581 Viapiano MS, Bi WL, Piepmeier J, et al, 2005, Novel tumorspecific isoforms of BEHAB/brevican identified in human malignant gliomas. Cancer Res 65(15):6726–6733 Nutt CL, Matthews RT, Hockfield S, 2001, Glial tumor invasion: a role for the upregulation and cleavage of BEHAB/brevican. Neuroscientist 7(2):113–122 Gary SC, Hockfield S, 2000, BEHAB/brevican: an extracellular matrix component associated with invasive glioma. Clin Neurosurg 47:72–82 Hu B, Kong LL, Matthews RT, et al, 2008, The proteoglycan brevican binds to fibronectin after proteolytic cleavage and promotes glioma cell motility. J Biol Chem 283(36):24848–24859 NakadaM,Miyamori H, Kita D, Takahashi T, Yamashita J, Sato H, Miura R, Yamaguchi Y, Okada Y, 2005, Human glioblastomas overexpress ADAMTS-5 that degrades brevican. Acta Neuropathol 110(3):239–246 Held-Feindt J, Paredes EB, Blomer U, Seidenbecher C, Stark AM, Mehdorn HM, Mentlein R, 2006, Matrix-degrading proteases ADAMTS4 and ADAMTS5, disintegrins and metalloproteinases with thrombospondin motifs 4 and 5, are expressed in human glioblastomas. Int J Cancer 118(1):55–61 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 155 EP 160 DI 10.1007/s12253-015-9989-5 PG 6 ER PT J AU Tian, L Shan, W Zhang, Y Lv, X Li, X Wei, C AF Tian, Lei Shan, Weiyu Zhang, Yufei Lv, Xuejun Li, Xuehua Wei, Caiyun TI Up-Regulation of miR-21 Expression Predicate Advanced Clinicopathological Features and Poor Prognosis in Patients with Non-Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; miR-21; Prognosis ID Lung cancer; miR-21; Prognosis AB MicroRNAs (miRNAs) are endogenous small (19– 24 nt long) noncoding RNAs that regulate gene expression in a sequence specific manner. An increasing association between miRNA and cancer has been recently reported. Lung cancer is globally responsible for 1.4 million deaths annually and is the leading cause of cancer-related deaths in both women and men. In this study, we investigated the miR-21 expression in non-small cell lung cancer (NSCLC) to evaluate their value in prognosis of this tumor. Here, we assess miR-21 expression in NSCLC and its clinical significance including survival analysis. The expression of miR-21 in matched normal and tumor tissues of NSCLC was evaluated using a quantitative real-time RT-PCR. A Kaplan–Meier survival curve was generated following a logrank test. It was observed that miR-21 expression was up-regulated in NSCLC tissues compared with noncancerous lung tissues (mean ± SD: 6.7 ± 2.3 vs. 3.7 ± 1.5, P < 0.001). The up-regulation of miR-21 in NSCLC cancer tissues was also significantly correlated with aggressive clinicopathological features.We found that the patients with high miR-21 expression have a higher tumor grade (P = 0.027) and are in higher risk of lymph node metastasis (P = 0.021). Moreover, the results of Kaplan–Meier analyses showed that NSCLC patients with the high miR-21 expression tend to have shorter overall survival and progression free survival (P < 0.001). The multivariate analysis clearly indicated that the high miR-21 expression in biopsy samples may be considered as an independent prognostic factor in NSCLC for decreased survival (RR 3.88; 95%CI, 2.47–6.11). Our data indicate the potential of miR-21 as a novel prognostic biomarker for NSCLC. Large well-designed studies with diverse populations and functional evaluations are warranted to confirm and extend our findings. C1 [Tian, Lei] No.88 Hospital of People’s Republic of China, Department of RespirationGuangzhou, China. [Shan, Weiyu] No.88 Hospital of People’s Republic of China, Department of RespirationGuangzhou, China. [Zhang, Yufei] No.88 Hospital of People’s Republic of China, Department of RespirationGuangzhou, China. [Lv, Xuejun] No.88 Hospital of People’s Republic of China, Department of RespirationGuangzhou, China. [Li, Xuehua] No.88 Hospital of People’s Republic of China, Department of RespirationGuangzhou, China. [Wei, Caiyun] No.88 Hospital of People’s Republic of China, Department of RespirationGuangzhou, China. RP Tian, L (reprint author), No.88 Hospital of People’s Republic of China, Department of Respiration, Guangzhou, China. EM drtianlei@163.com CR Zycinska K, Kostrzewa-Janicka J, Nitsch-Osuch A, Wardyn K, 2013, Cancer incidence in pulmonary vasculitis. Adv Exp Med Biol 788:349–353 Hata A, Katakami N, Yoshioka H, Takeshita J, Tanaka K, Nanjo S, Fujita S, et al., 2013, Rebiopsy of non-small cell lung cancer patients with acquired resistance to epidermal growth factor receptortyrosine kinase inhibitor: comparison between T790M mutationpositive and mutation-negative populations. Cancer 119:4325– 4323 Spigel DR, Ervin TJ, Ramlau RA, Daniel DB, Goldschmidt Jr JH, Blumenschein Jr GR, Krzakowski MJ, et al., 2013, Randomized phase II trial of onartuzumab in combination with erlotinib in patients with advanced Non-small-cell lung cancer. J Clin Oncol 31: 4105–4114 Shapiro M, Kadakia S, Lim J, Breglio A, Wisnivesky JP, Kaufman A, Lee DS, et al., 2013, Lobe-specific mediastinal nodal dissection is sufficient during lobectomy by video-assisted thoracic surgery or thoracotomy for early-stage lung cancer. Chest 144:1615–1621 Dhawan P, Singh AB, Ellis DL, Richmond A, 2002, Constitutive activation of Akt/protein kinase B in melanoma leads to upregulation of nuclear factor-kappaB and tumor progression. Cancer Res 62:7335–7342 Rodriguez A, Griffiths-Jones S, Ashurst JL, Bradley A, 2004, Identification of mammalian microRNA host genes and transcription units. Genome Res 14:1902–1910 Esquela-Kerscher A, Slack FJ, 2006, Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer 6:259–269 de Yebenes VG, Ramiro AR, 2010, MicroRNA activity in B lymphocytes. Methods Mol Biol 667:177–192 Pasquinelli AE, Reinhart BJ, Slack F, Martindale MQ, Kuroda MI, Maller B, Hayward DC, et al., 2000, Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature 408:86–89 Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T, 2001, Identification of novel genes coding for small expressed RNAs. Science 294:853–858 Cai X, Hagedorn CH, Cullen BR, 2004, Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA 10:1957–1966 Chan JA, Krichevsky AM, Kosik KS, 2005, MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res 65: 6029–6033 Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S, Allgayer H, 2008, MicroRNA-21, miR-21, posttranscriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 27:2128–2136 Gao W, Yu Y, Cao H, Shen H, Li X, Pan S, Shu Y, 2010, Deregulated expression of miR-21, miR-143 and miR-181a in non small cell lung cancer is related to clinicopathologic characteristics or patient prognosis. Biomed Pharmacother 64:399–408 Lo TF, Tsai WC, Chen ST, 2013, MicroRNA-21-3p, a berberineinduced miRNA, directly down-regulates human methionine adenosyltransferases 2A and 2B and inhibits hepatoma cell growth. PLoS One 8:e75628 WangY, Gao X,Wei F, Zhang X, Yu J, Zhao H, Sun Q, et al., 2013, Diagnostic and prognostic value of circulating miR-21 for cancer: a systematic review and meta-analysis. Gene 533(1):389–397 Ferraro A, Kontos C, Boni T, Bantounas I, Siakouli D, Kosmidou V, Vlassi M, et al., 2013, Epigenetic regulation of miR-21 in colorectal cancer: ITGB4 as a novel miR-21 target and a three-gene network, miR-21-ITGBeta4-PCDC4, as predictor of metastatic tumor potential. Epigenetics 9:129–141 Brito JA, Gomes CC, Guimaraes AL, Campos K, Gomez RS, 2014, Relationship between microRNA expression levels and histopathological features of dysplasia in oral leukoplakia. J Oral Pathol Med 43:211–216 Chen TH, Chang SW, Huang CC,Wang KL, Yeh KT, Liu CN, Lee H, et al., 2013, The prognostic significance of APC gene mutation and miR-21 expression in advanced stage colorectal cancer. Color Dis 15:1367–1374 Li Z, Li N,Wu M, Li X, Luo Z,Wang X, 2013, Expression ofmiR- 126 suppresses migration and invasion of colon cancer cells by targeting CXCR4. Mol Cell Biochem 381:233–242 Jia LF,Wei SB, Gong K, Gan YH, Yu GY. Prognostic implications of micoRNA miR-195 expression in human tongue squamous cell carcinoma. PLoS One 2013;8:e56634. Pelosi A, Careccia S, Lulli V, Romania P, Marziali G, Testa U, Lavorgna S, et al., 2013, MiRNA let-7c promotes granulocytic differentiation in acute myeloid leukemia. Oncogene 32:3648– 3654 Zhu S,Wu H,Wu F, NieD, Sheng S,Mo YY(2008)MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res 18:350–359 Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, et al., 2006, A microRNAexpression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 103:2257–2261 Selcuklu SD, Donoghue MT, Spillane C, 2009, miR-21 as a key regulator of oncogenic processes. Biochem Soc Trans 37:918–925 Loffler D, Brocke-Heidrich K, Pfeifer G, Stocsits C, Hackermuller J, Kretzschmar AK, Burger R, et al., 2007, Interleukin-6 dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer. Blood 110:1330–1333 SaitoM, Schetter AJ, Mollerup S, Kohno T, Skaug V, Bowman ED, Mathe EA, et al., 2011, The association of microRNA expression with prognosis and progression in early-stage, non-small cell lung adenocarcinoma: a retrospective analysis of three cohorts. Clin Cancer Res 17:1875–1882 Schmittgen TD, Jiang J, Liu Q, Yang L. A high-throughput method to monitor the expression of microRNA precursors. Nucleic Acids Res 2004;32:e43. Ozgun A, Karagoz B, Bilgi O, Tuncel T, Baloglu H, Kandemir EG, 2013, MicroRNA-21 as an indicator of aggressive phenotype in breast cancer. Onkologie 36:115–118 Lu Z, LiuM, Stribinskis V, KlingeCM, Ramos KS, ColburnNH, Li Y, 2008, MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene 27:4373–4379 Komatsu S, Ichikawa D, Tsujiura M, Konishi H, Takeshita H, Nagata H, Kawaguchi T, et al., 2013, Prognostic impact of circulating miR-21 in the plasma of patients with gastric carcinoma. Anticancer Res 33:271–276 Hermansen SK, Dahlrot RH, Nielsen BS, Hansen S, Kristensen BW, 2013, MiR-21 expression in the tumor cell compartment holds unfavorable prognostic value in gliomas. J Neuro-Oncol 111:71–81 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 161 EP 167 DI 10.1007/s12253-015-9979-7 PG 7 ER PT J AU Lim, ChL Looi, LM Zakaria, ZSS Sagap, I Rose, MI Chin, SF Jamal, R AF Lim, Cheng Lay Looi, Lee Mee Zakaria, Zulkifli Syed Syed Sagap, Ismail Rose, Mohammed Isa Chin, Siok-Fong Jamal, Rahman TI Identification of Differentially Expressed Proteins in the Serum of Colorectal Cancer Patients Using 2D-DIGE Proteomics Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Proteomics; 2D-DIGE; LC-MS/MS; Apolipoprotein A1 ID Colorectal cancer; Proteomics; 2D-DIGE; LC-MS/MS; Apolipoprotein A1 AB Early detection of colorectal cancer (CRC) is vital for the improvement of disease prognosis. However to date there are no blood-based biomarkers sensitive and specific enough for early diagnosis. We analysed the differences in serum protein expression of early stage CRC (Dukes’ A and B) and late stage CRC (Dukes’ C and D) against normal controls using 2D Fluorescence Difference Gel Electrophoresis (2D-DIGE). Analysis of the 2D maps showed that 23 proteins were differentially expressed between groups (p≤0.05) and these proteins were identified with LC-MS/MS. Eight proteins were up-regulated and 2 down-regulated in patients with early CRC, whereas 14 proteins were up-regulated and 4 downregulated in those with late CRC compared to normal controls (p≤0.05). Five proteins, namely apolipoprotein A1 (APOA1), apolipoprotein E (APOE), complement factor H (CFH), galectin-7 (GAL7) and synaptojanin-2 (SYNJ2) were validated using ELISA and only APOA1 and GAL-7 showed consistent findings. Further validation using immunohistochemistry showed negative immunoreactivity for GAL-7 in CRC tissues, suggesting that GAL-7 detected in the serum did not originate from the CRC tumour. APOA1 showed positive immunoreactivity but its expression did not correlate with Dukes’ staging (p=0.314), tumour grading (p=0.880) and lymph node involvement (p=0.108). Differences in APOA1 isoforms and/or conformation between serum and tissue samples as well as tumour heterogeneity may explain for the discrepancies between DIGE and ELISA when compared to immunohistochemistry. Structural and functional studies of APOA1 in future would best describe the role of APOA1 in CRC. C1 [Lim, Cheng Lay] Universiti Kebangsaan Malaysia, UKM Medical Molecular Biology Institute (UMBI), Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia. [Looi, Lee Mee] Taylor’s University, Lakeside Campus, School of Biosciences, Subang JayaSelangor, Malaysia. [Zakaria, Zulkifli Syed Syed] Universiti Kebangsaan Malaysia, UKM Medical Molecular Biology Institute (UMBI), Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia. [Sagap, Ismail] Universiti Kebangsaan Malaysia, Faculty of Medicine, Department of SurgeryKuala Lumpur, Malaysia. [Rose, Mohammed Isa] Universiti Kebangsaan Malaysia, Faculty of Medicine, Department of PathologyKuala Lumpur, Malaysia. [Chin, Siok-Fong] Universiti Kebangsaan Malaysia, UKM Medical Molecular Biology Institute (UMBI), Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia. [Jamal, Rahman] Universiti Kebangsaan Malaysia, UKM Medical Molecular Biology Institute (UMBI), Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia. RP Jamal, R (reprint author), Universiti Kebangsaan Malaysia, UKM Medical Molecular Biology Institute (UMBI), 56000 Kuala Lumpur, Malaysia. EM rahmanj@ppukm.ukm.edu.my CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin M, 2010, GLOBOCAN 2008, cancer incidence and mortality worldwide: IARC cancer base no.10. International Agency for Research on Cancer, Lyon, France Zainal AO, Zainudin MA, Saleha IT, 2006, Malaysian cancer statistics-data and figure peninsular Malaysia 2006. National Cancer Registry, Ministry of Health Malaysia, Kuala Lumpur American Cancer Society, 2007, Cancer facts and figures 2007. Atlanta: American Cancer Society Rex DK, 2007, Colorectal cancer screening. Am Soc Gastrointest Endosc 14(4):1–4 World Gastroenterology Organisation/International Digestive Cancer Alliance, 2007, Practice guidelines for colorectal cancer screening. World Gastroenterology Organisation Anderson NL, Anderson NG, 2002, The human plasma proteome: history, character and diagnostic prospect. Mol Cell Proteomics 1(11):845–867 Tanaka T, Tanaka M, Ishigamori R, 2010, Biomarkers for colorectal cancer. Int J Mol Sci 11(9):3209–3225., DOI 10.3390/ ijms11093209 GoldsteinMJ, Mitchell EP, 2005, Carcinoembryonic antigen in the staging and follow up of patients with colorectal cancer. Cancer Investig 23:338–351 Hammarstrom S, 1999, The carcinoembryonic antigen, CEA, family: structures, suggested functions and expression in normal and malignant tissues. Cancer Biol 9:67–81 Marshall KW, Mohr S, Khettabi FE, Nossova N, Chao S, BaoWS, Ma J, Li XJ, Liew CC, 2010, A blood-based biomarker panel for stratifying current risk for colorectal cancer. Int J Cancer 126:1177– 1186 Yip KT, Das PK, Suria D, Lim CR, Ng GH, Liew CC, 2010, A case-controlled validation study of a blood-based seven-gene biomarker panel for colorectal cancer in Malaysia. J Exp Clin Cancer Res 29(128):1–7 Guo YF, Xiao P, Lei SF, Deng FY, Xiao GS, Liu YZ, Chen XD, Li LM, Wu S, Chen Y, Jiang H, Tan LJ, Xie JY, Zhu XZ, Liang SP, Deng HW, 2008, How is mRNA expression predictive for protein expression? A correlation study on human circulating monocytes. Acta Biochim Biophys Sin 40:426–436 Kim KH, Kim SH, Back JH, Park MJ, Kim JM, 2006, Cyclooxygenase-2 and inducible nitric oxide synthase expression in thyroid neoplasms and their clinicopathological correlation. J Korean Med Sci 21(6):1064–1069 Malecz N, McCabe PC, Spaargaren C, Qiu R-G, Chuang Y-Y, Symons M, 2000, Synaptojanin 2, a novel Rac1 effector that regulates clathrin-mediated endocytosis. Curr Biol 10(21):1383–1386., DOI 10.1016/S0960-9822(00)00778-8 Vial E, Sahai E, Marshall CJ, 2003, ERK-MAPK signaling coordinately regulates activity of Rac1 and RhoA for tumor cell motility. Cancer Cell 4(1):67–79., DOI 10.1016/S1535-6108(03)00162-4 Wortzel I, Seger R, 2011, The ERK cascade: distinct functions within various subcellular organelles. Genes Cancer 2(3):195–209 Chuang YY, Tran NL, Rusk N, Nakada M, Berens ME, Symons M, 2004, Role of synaptojanin 2 in glioma cell migration and invasion. Cancer Res 64:8271–8275 Rodríguez de Cordoba S, Esparza-Gordillo J, Goicoechea de Jorge E, Lopez-Trascasa M, Sanchez-Corral P, 2004, The human complement factor H: functional roles, genetic variations and disease associations.Mol Immunol 41(4):355–367., DOI 10.1016/j.molimm. 2004.02.005 Nguyen VT, Arredondo J, Chernyavsky AI, Kitajima Y, Pittelkow M, Grando SA, 2004, Pemphigus vulgaris IgG and methylprednisolone exhibit reciprocal effects on keratinocytes. J Biol Chem 279(3):2135–2146., DOI 10.1074/jbc.M309000200 Demers M, Magnaldo T, St-Pierre Y, 2005, A novel function for galectin-7: promoting tumorigenesis by up-regulatingMMP-9 gene expression. Cancer Res 65(12):5205–5210., DOI 10.1158/0008- 5472.CAN-05-0134 Kuwabara I, Kuwabara Y, Yang RY, Schuler M, Green DR, Zuraw BL, Hsu DK, Liu FT, 2002, Galectin-7, PIG1, exhibits proapoptotic function through JNK activation and mitochondrial cytochrome c release. J Biol Chem 277(5):3487–3497 Saussez S, Cludts S, Capouillez A, Mortuaire G, Smetana K, Kaltner H, Andre S, Leroy X, Gabius HJ, Decaestecker C, 2009, Identification of matrix metalloproteinase-9 as an independent prognostic marker in laryngeal and hypopharyngeal cancer with opposite correlations to adhesion/growth regulatory galectins-1 and -7. Int J Oncol 34:433–439 Park JE, Chang WY, Cho MJ, 2009, Induction of matrix metalloproteinase-9 by galectin-7 through p38 MAPK signaling in HeLa human cervical epithelial adenocarcinoma cells. Oncol Rep 22:1373–1379 Aggarwal BB, Shishodia S, Sandur SK, Pandey MK, Sethi G, 2006, Inflammation and cancer: how hot is the link? Biochem Pharmacol 72(11):1605–1621 KhovidhunkitW, KimMS, Memon RA, Shigenaga JK,Moser AH, Feingold KR, Grunfeld C, 2004, Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. J Lipid Res 45(7):1169–1196 Beigneux AP, Moser AH, Shigenaga JK, Grunfeld C, Feingold KR, 2000, The acute phase response is associated with retinoid X receptor repression in rodent liver. J Biol Chem 275(21):16390– 16399 Laffitte BA, Repa JJ, Joseph SB, Wilpitz DC, Kast HR, Mangelsdorf DJ, Tontonoz P, 2001, LXRs control lipid-inducible expression of the apolipoprotein E gene in macrophages and adipocytes. ProcNatl Acad Sci U S A98(2):507–512., DOI 10.1073/ pnas.021488798 Schwartz K, Lawn RM, Wade DP, 2000, ABC1 gene expression and APOA1 mediated cholesterol efflux are regulated by LXR. Biochem Biophys Res Commun 274:794–802 Kim W-K, Meliton V, Park KW, Hong C, Tontonoz P, Niewiadomski P, Waschek JA, Tetradis S, Parhami F, 2009, Negative regulation of hedgehog signaling by liver X receptors. Mol Endocrinol 23(10):1532–1543., DOI 10.1210/me.2008-0453 Tachibana M, Ohkura Y, Kobayashi Y, Sakamoto H, Tanaka Y, Watanabe J, Amikura K, Nishimura Y, Akagi K, 2003, Expression of apolipoprotein A1 in colonic adenocarcinoma. Anticancer Res 23(5b):4161–4167 Van Duijnhoven FJB, Bueno-De-Mesquita HB, CalligaroM, Jenab M, Pischon T, Jansen EHJM, Frohlich J, Ayyobi A, Overvad K, Toft-Petersen AP, Tjonneland A, Hansen L, Boutron-Ruault MC, Clavel-Chapelon F, Cottet V, Palli D, Tagliabue G, Panico S, Tumino R, Vineis P, Kaaks R, Teucher B, Boeing H, Drogan D, Trichopoulou A, Lagiou P, Dilis V, Peeters PHM, Siersema PD, Rodriguez L, Gonzalez CA, Molina-Montes E, Dorronsoro M, Tormo MJ, Barricarte A, Palmqvist R, Hallmans G, Khaw KT, Tsilidis KK, Crowe FL, Chajes V, Fedirko V, Rinaldi S, Norat T, Riboli E, 2011, Blood lipid and lipoprotein concentrations and colorectal cancer risk in the european propective investigation into cancer and nutrition. Gut 60:1094–1102 Opstal-van Winden AWJ, Krop EJM, Karedal MH, Gast MCW, Lindh CH, Jeppsson MC, Jonsson BAG, Grobbee DE, Peeters PHM, Beijnen JH, van Gils CH, Vermeulen RCH, 2011, Searching for early breast cancer biomarkers by serum protein profiling of pre-diagnostic serum; a nested case-control study. BMC Cancer 11., DOI 10.1186/1471-2407-11-381 Engwegen JYMN, Helgason HH, Cats A, Harris N, Bonfrer JMG, Schellens JHM, Beijnen JH, 2006, Identification of serum proteins discriminating colorectal cancer patients and healthy controls using surface-enhanced laser desorption ionisation-time of flight mass spectrometry. World J Gastroenterol 12(10):1536–1544 Zhu CS, Pinsky PF, Cramer DW, Ransohoff DF, Hartge P, Pfeiffer RM, Urban N,Mor G, Bast RC, Moore LE, Lokshin AE, McIntosh MW, Skates SJ, Vitonis A, Zhang Z, Ward DC, Symanowski JT, Lomakin A, Fung ET, Sluss PM, Scholler N, Lu KH, Marrangoni AM, Patriotis C, Srivastava S, Buys SS, Berg CD, Team PP, 2011, A framework for evaluating biomarkers for early detection: validation of biomarker panels for ovarian cancer. Cancer Prev Res 4(3): 375–383., DOI 10.1158/1940-6207.Capr-10-0193 Chen Y-T, Chen C-L, Chen H-W, Chung T, Wu C-C, Chen C-D, Hsu C-W, Chen M-C, Tsui K-H, Chang P-L, Chang Y-S, Yu J-S, 2010, Discovery of novel bladder cancer biomarkers by comparative urine proteomics using iTRAQ technology. J Proteome Res 9(11):5803–5815., DOI 10.1021/pr100576x Li H, Li C, Wu H, Zhang T, Wang J, Wang S, Chang J, 2011, Identification of APO-A1 as a biomarker for early diagnosis of bladder transitional cell carcinoma. Proteome Sci 9:21 Hamrita B, Nasr HB, Gabbouj S, Bouaouina N, Chouchane L, ChahedK(2011)Apolipoprotein A1-75G/A and +83C/T polymorphisms: susceptibility and prognostic implications in breast cancer. Mol Biol Rep 38:1637–1643 Fung ET, 2010, A recipe for proteomics diagnostic test development: the OVA1 test, from biomarker discovery to FDA clearance. Clin Chem 56(2):327–329 Sparks DL, Philips MC, Lund-Katz S, 1992, The conformation of apolipoprotein A-I in discoidal and spherical recombinant high density lipoprotein particles. J Biol Chem 267(36): 25830–25838 Heppner GH, 1984, Tumor heterogeneity. Cancer Res 44(6):2259– 2265 Permuth-Wey J, Boulware D, Valkov N, Livingston S, Nicosia S, Lee JH, Sutphen R, Schildkraut J, Narod S, Parker A, Coppola D, Sellers T, Pal T, 2009, Sampling strategies for tissue microarrays to evaluate biomarkers in ovarian cancer. Cancer Epidemiol Biomarkers Prev 18(1):28–34., DOI 10.1158/1055-9965.EPI-08- 0713 Bentzen SM, Buffa FM, Wilson GD, 2008, Multiple biomarker tissue microarrays: bioinformatics and practical approaches. Cancer Metastasis Rev 27(3):481–494., DOI 10.1007/s10555-008- 9145-8 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 169 EP 177 DI 10.1007/s12253-015-9991-y PG 9 ER PT J AU Benczik, M Galamb, Koiss, R Kovacs, A Jaray, B Szekely, T Szekerczes, T Schaff, Zs Sobel, G Jeney, Cs AF Benczik, Marta Galamb, Adam Koiss, Robert Kovacs, Attila Jaray, Balazs Szekely, Tamas Szekerczes, Timea Schaff, Zsuzsa Sobel, Gabor Jeney, Csaba TI Claudin-1 as a Biomarker of Cervical Cytology and Histology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; Biomarker; Claudin-1; Immunocytochemistry ID Cervical cancer; Biomarker; Claudin-1; Immunocytochemistry AB Several immunochemistry tests are used for triaging human papilloma virus (HPV) and cytology positive cases in cervical cancer screening and as an adjunct test to diagnose cervical cancer. Claudin-1 (CLDN1) protein is a major component of the tight junction, shown to have altered expression in cervical cancer. In this study, value of CLDN1 was analysed as a screening and triage immunochemistry test compared to cytology and HPV testing. A population of 352 women attending colposcopic referral visits resulting in cervical conisation and a second population of 150 women attending routine gynaecological visits with negative cervical cytology were enrolled in a multi-centre clinical study in Hungary. Cytology and HPV (Genoid Full Spectrum HPV Amplification and Detection System) testing were carried out along with immunocytochemistry for CLDN1, and as a reference, using CINtec p16 Cytology Kit. Three different evaluation protocols were used which assessed immunostaining characteristics with or without cytological readings. High correlation observable between p16INK4a and CLDN1 established CLDN1 as a competing marker in cervical cancer. Concordance of CLDN1 immunostaining of cervical intraepithelial neoplasia 2 and above (CIN2+) positives was 84.0 % (73.8–89.3); concordance of CIN2+ negatives was 69.0 % (59.6–75.8). In conclusion, CLDN1 has similar diagnostic potential as p16INK4a, our results established it as a histological and cytological biomarker with the potential to improve the clinical performance of cervical cytology and histology. C1 [Benczik, Marta] CellCall Ltd, Roppentyu u. 48, 1134 Budapest, Hungary. [Galamb, Adam] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi ut 78/a, 1082 Budapest, Hungary. [Koiss, Robert] Del-Pesti Centrumkorhaz, Nagyvarad ter 1, 1097 Budapest, Hungary. [Kovacs, Attila] CellCall Ltd, Roppentyu u. 48, 1134 Budapest, Hungary. [Jaray, Balazs] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Szekely, Tamas] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Szekerczes, Timea] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Sobel, Gabor] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi ut 78/a, 1082 Budapest, Hungary. [Jeney, Csaba] CellCall Ltd, Roppentyu u. 48, 1134 Budapest, Hungary. RP Jeney, Cs (reprint author), CellCall Ltd, 1134 Budapest, Hungary. EM csjeney@gmail.com CR Forouzanfar MH, Foreman KJ, Delossantos AM, et al., 2011, Breast and cervical cancer in 187 countries between 1980 and 2010: a systematic analysis. Lancet 378:1461–1484., DOI 10.1016/ S0140-6736(11)61351-2 Iarc, 2003, Colposcopy and treatment of cervical intraepithelial neoplasia: A beginners' manual. 1–132. Wright TC, Massad LS, Dunton CJ, et al., 2007, 2006 consensus guidelines for the management of women with abnormal cervical cancer screening tests. Am J Obstet Gynecol 197:346–355., DOI 10. 1016/j.ajog.2007.07.047 Gustafsson L, Ponten J, Bergstrom R, Adami HO, 1997, International incidence rates of invasive cervical cancer before cytological screening. Int J Cancer 71:159–165 Walboomers JM, Jacobs MV, Manos MM, et al., 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189:12–19., DOI 10.1002/(SICI)1096- 9896(199909)189:1<12::AID-PATH431>3.0.CO;2-F Cuzick J, Clavel C, Petry K-U, et al., 2006, Overview of the European and north American studies on HPV testing in primary cervical cancer screening. Int J Cancer 119:1095–1101., DOI 10. 1002/ijc.21955 Ronco G,Giorgi-Rossi P, Carozzi F, et al., 2010, Efficacy of human papillomavirus testing for the detection of invasive cervical cancers and cervical intraepithelial neoplasia: a randomised controlled trial. Lancet Oncol 11:249–257., DOI 10.1016/S1470-2045(09)70360-2 Anttila A, Kotaniemi-Talonen L, Leinonen M, et al., 2010, Rate of cervical cancer, severe intraepithelial neoplasia, and adenocarcinoma in situ in primary HPV DNA screening with cytology triage: randomised study within organised screening programme. BMJ 340:c1804 Plummer M, Schiffman M, Castle PE, et al., 2007, A 2-year prospective study of human papillomavirus persistence among women with a cytological diagnosis of atypical squamous cells of undetermined significance or low-grade squamous intraepithelial lesion. J Infect Dis 195:1582–1589., DOI 10.1086/516784 Rodriguez AC, Schiffman M, Herrero R, et al., 2008, Rapid clearance of human papillomavirus and implications for clinical focus on persistent infections. J Natl Cancer Inst 100:513–517., DOI 10.1093/ jnci/djn044 Saslow D, Solomon D, Lawson HW, et al. American Cancer Society, American Society for Colposcopy and Cervical Pathology, and American Society for Clinical Pathology screening guidelines for the prevention and early detection of cervical cancer. CA Cancer J Clin 62:147–172., DOI 10.3322/caac.21139 Naucler P, RydW, Tornberg S, et al., 2009, Efficacy of HPV DNA testing with cytology triage and/or repeat HPV DNA testing in primary cervical cancer screening. J Natl Cancer Inst 101:88–99., DOI 10.1093/jnci/djn444 Rijkaart DC, Berkhof J, van Kemenade FJ, et al., 2012, HPV DNA testing in population-based cervical screening, VUSA-screen study): results and implications. Br J Cancer 106:975–981., DOI 10.1038/bjc.2011.581 Castle PE, Stoler MH, Wright TC, et al., 2011, Performance of carcinogenic human papillomavirus, HPV, testing and HPV16 or HPV18 genotyping for cervical cancer screening of women aged 25 years and older: a subanalysis of the ATHENA study. Lancet Oncol 12:880–890., DOI 10.1016/S1470-2045(11)70188-7 Overmeer RM, Louwers JA,Meijer CJLM, et al., 2011, Combined CADM1 and MAL promoter methylation analysis to detect, pre- )malignant cervical lesions in high-risk HPV-positive women. Int J Cancer 129:2218–2225., DOI 10.1002/ijc.25890 Depuydt CE, Makar AP, Ruymbeke MJ, et al., 2011, BD-ProExC as adjunct molecular marker for improved detection of CIN2+ after HPV primary screening. Cancer Epidemiol Biomark Prev 20:628– 637., DOI 10.1158/1055-9965.EPI-10-0818 Patel DA, Rozek LS, Colacino JA, et al., 2012, Patterns of cellular and HPV 16 methylation as biomarkers for cervical neoplasia. J Virol Methods 184:84–92., DOI 10.1016/j.jviromet.2012.05.022 De Strooper LMA, Hesselink AT, Berkhof J, et al., 2014, Combined CADM1/MAL methylation and cytology testing for colposcopy triage of high-risk HPV-positive women. Cancer Epidemiol Biomark Prev 23:1933–1937., DOI 10.1158/1055-9965. EPI-14-0347 BibboM, DeCecco J, Kovatich AJ, 2003, P16INK4A as an adjunct test in liquid-based cytology. Anal Quant Cytol Histol 25:8–11 Galamb A, Benczik M, Zinner B, et al., 2015, Dysregulation of microRNA expression in human cervical preneoplastic and neoplastic lesions. Pathol Oncol Res., DOI 10.1007/s12253-014-9871-x Tsoumpou I, Arbyn M, Kyrgiou M, et al., 2009, p16(INK4a, immunostaining in cytological and histological specimens from the uterine cervix: a systematic review and meta-analysis. Cancer Treat Rev 35:210–220., DOI 10.1016/j.ctrv.2008.10.005 Galgano MT, Castle PE, Atkins KA, et al., 2010, Using biomarkers as objective standards in the diagnosis of cervical biopsies. Am J Surg Pathol 34:1077–1087., DOI 10.1097/PAS.0b013e3181e8b2c4 Denton KJ, Bergeron C, Klement P, et al., 2010, The sensitivity and specificity of p16(INK4a, cytology vs HPV testing for detecting high-grade cervical disease in the triage of ASC-US and LSIL pap cytology results. Am J Clin Pathol 134:12–21., DOI 10.1309/ AJCP3CD9YKYFJDQL Van Bogaert L-J, 2012, P16INK4a immunocytochemistry/immunohistochemistry: need for scoring uniformization to be clinically useful in gynecological pathology. Ann Diagn Pathol 16:422–426., DOI 10.1016/j.anndiagpath.2012.03.006 Lal-Nag M, Morin PJ, 2009, The claudins. Genome Biol 10:235., DOI 10.1186/gb-2009-10-8-235 Tsukita S, Yamazaki Y, Katsuno T, Tamura A, 2008, Tight junction-based epithelial microenvironment and cell proliferation. Oncogene 27:6930–6938., DOI 10.1038/onc.2008.344 Morin PJ, 2005, Claudin proteins in human cancer: promising new targets for diagnosis and therapy. Cancer Res 65:9603–9606., DOI 10.1158/0008-5472.CAN-05-2782 Sobel G, Nemeth J, Kiss A, et al., 2006, Claudin 1 differentiates endometrioid and serous papillary endometrial adenocarcinoma. Gynecol Oncol 103:591–598., DOI 10.1016/j.ygyno.2006.04.005 Sobel G, Paska C, Szabo I, et al., 2005, Increased expression of claudins in cervical squamous intraepithelial neoplasia and invasive carcinoma. Hum Pathol 36:162–169., DOI 10.1016/j.humpath.2004. 12.001 Szabo I, Kiss A, Schaff Z, Sobel G, 2009, Claudins as diagnostic and prognostic markers in gynecological cancer. Histol Histopathol 24:1607–1615 Lee J-W, Lee S-J, Seo J, et al., 2005, Increased expressions of claudin-1 and claudin-7 during the progression of cervical neoplasia. Gynecol Oncol 97:53–59., DOI 10.1016/j.ygyno.2004.11.058 Wentzensen N, von Knebel DM, 2007, Biomarkers in cervical cancer screening. Dis Markers 23:315–330 Tornesello ML, Buonaguro L, Giorgi-Rossi P, Buonaguro FM, 2013, Viral and cellular biomarkers in the diagnosis of cervical intraepithelial neoplasia and cancer. Biomed Res Int 2013: 519619., DOI 10.1155/2013/519619 Vazquez-Ortiz G, Ciudad CJ, Pina P, et al. Gene identification by cDNA arrays in HPV-positive cervical cancer. Arch Med Res 36: 448–458., DOI 10.1016/j.arcmed.2005.04.016 Vrdoljak-Mozetic D, Krasevic M, Versa Ostojic D, et al., 2015, HPV16 genotype, p16/Ki-67 dual staining and koilocytic morphology as potential predictors of the clinical outcome for cervical lowgrade squamous intraepithelial lesions. Cytopathology 26:10–18., DOI 10.1111/cyt.12121 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 179 EP 188 DI 10.1007/s12253-015-9990-z PG 10 ER PT J AU Giaginis, C Alexandrou, P Delladetsima, I Karavokyros, I Danas, E Giagini, A Patsouris, E Theocharis, S AF Giaginis, Constantinos Alexandrou, Paraskevi Delladetsima, Ioanna Karavokyros, Ioannis Danas, Eugene Giagini, Athina Patsouris, Efstratios Theocharis, Stamatios TI Clinical Significance of Hu-Antigen Receptor (HuR) and Cyclooxygenase-2 (COX-2) Expression in Human Malignant and Benign Thyroid Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thyroid malignancy; Hu-antigen R; Cyclooxygenase-2; Clinicopathological parameters; Immunohistochemistry ID Thyroid malignancy; Hu-antigen R; Cyclooxygenase-2; Clinicopathological parameters; Immunohistochemistry AB Hu-antigen R (HuR) is considered to play a crucial role in tumor formation and growth by binding to mRNAs encoding proteins such as Cyclooxygenase-2 (COX-2) and inducing their expression via mRNA stabilization and/or altered translation. The present study aimed to evaluate the clinical significance of HuR and COX-2 proteins’ expression in human benign and malignant thyroid lesions. HuR and COX-2 proteins’ expression was assessed immunohistochemically on paraffin-embedded thyroid tissues obtained from 98 patients with benign (n = 48) and malignant (n = 50) lesions and was statistically analyzed with clinicopathological parameters, follicular cells’ proliferative capacity and recurrence risk rate. Enhanced HuR and COX-2 expression was significantly more frequently observed in malignant compared to benign thyroid lesions (p = 0.0073 and p = 0.0016, respectively), as well as in papillary carcinomas compared to hyperplastic nodules (p = 0.0039 and p = 0.0009, respectively). Positive associations of both HuR and COX-2 expression with follicular cells’ proliferation rate were also noted (p = 0.0087 and p = 0.0127, respectively). In malignant thyroid lesions, elevated COX-2 expression was significantly associated with female patients’ gender (p = 0.0381) and the presence of lymph node metastases (p = 0.0296). The present data support evidence that both HuR and COX-2 may be involved in the malignant state of thyroid neoplasia and may be utilized in the diagnosis of malignant thyroid tumors. C1 [Giaginis, Constantinos] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str, GR11527 Goudi, Athens, Greece. [Alexandrou, Paraskevi] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str, GR11527 Goudi, Athens, Greece. [Delladetsima, Ioanna] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str, GR11527 Goudi, Athens, Greece. [Karavokyros, Ioannis] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str, GR11527 Goudi, Athens, Greece. [Danas, Eugene] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str, GR11527 Goudi, Athens, Greece. [Giagini, Athina] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str, GR11527 Goudi, Athens, Greece. [Patsouris, Efstratios] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str, GR11527 Goudi, Athens, Greece. [Theocharis, Stamatios] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str, GR11527 Goudi, Athens, Greece. RP Theocharis, S (reprint author), University of Athens, Medical School, First Department of Pathology, GR11527 Goudi, Greece. EM statheocharis@yahoo.com CR Ma WJ, Cheng S, Campbell C, Wright A, Furneaux H, 1996, Cloning and characterization of HuR, a ubiquitously expressed elav-like protein. J Biol Chem 271:8144–8151 MaWJ, Furneaux H, 1997, Localization of the human HuR gene to chromosome 19p13.2. Hum Genet 99:32–33 Burd CG, Dreyfuss G, 1994, Conserved structures and diversity of functions of RNA-binding proteins. Science 265:615–621 Lopez de Silanes I, Zhan M, Lal A, Yang X, Gorospe M, 2004, Identification of a target RNA motif for RNA-binding protein HuR. Proc Natl Acad Sci U S A 101:2987–2992 Fan XC, Steitz JA, 1998, HNS, a nuclear-cytoplasmic shuttling sequence in HuR. Proc Natl Acad Sci U S A 95:15293–11528 Abdelmohsen K, Lal A, Kim HH, Gorospe M, 2007, Posttranscriptional orchestration of an anti-apoptotic program by HuR. Cell Cycle 6:1288–1292 Yiakouvaki A, Dimitriou M, Karakasiliotis I, Eftychi C, Theocharis S, Kontoyiannis DL, 2012, Myeloid cell expression of the RNAbinding protein HuR protects mice from pathologic inflammation and colorectal carcinogenesis. J Clin Invest 122:48–61 Khan Z, Khan N, Tiwari RP, Sah NK, Prasad GB, Bisen PS, 2011, Biology of Cox-2: an application in cancer therapeutics. Curr Drug Targets 12:1082–1093 GhoshN, Chaki R,Mandal V,Mandal SC, 2010, COX-2 as a target for cancer chemotherapy. Pharmacol Rep 62:233–244 Leenhardt L, Grosclaude P, Cherie-Challine L, 2004, Increased incidence of thyroid carcinoma in France: a true epidemic or thyroid nodule management effects? Report from the French thyroid cancer committee. Thyroid 14:1056–1060 Ward EM, Jemal A, Chen A, 2010, Increasing incidence of thyroid cancer: is diagnostic scrutiny the sole explanation? Future Oncol 6: 185–188 Hundahl SA, Fleming ID, Fremgen AM, et al., 1998, A national cancer data base report on 53,856 cases of thyroid carcinoma treated in the U.S., 1985–1995. Cancer 83:2638–2648 Gharib H, Papini E, 2007, Thyroid nodules: clinical importance, assessment, and treatment. Endocrinol Metab Clin N Am 36:707–735 Schlumberger M, Lacroix L, Russo D, Filetti S, Bidart JM, 2007, Defects in iodide metabolism in thyroid cancer and implications for the follow-up and treatment of patients. Nat Clin Pract Endocrinol Metab 3:260–269 Rhee WJ, Ni CW, Zheng Z, Chang K, Jo H, Bao G, 2010, HuR regulates the expression of stress-sensitive genes and mediates inflammatory response in human umbilical vein endothelial cells. Proc Natl Acad Sci U S A 107:6858–6863 Zhang J, Modi Y, Yarovinsky T, et al., 2012, Macrophage beta2 integrin-mediated, HuR-dependent stabilization of angiogenic factor-encoding mRNAs in inflammatory angiogenesis. Am J Pathol 180:1751–1760 Lopez de Silanes I, Lal A, Gorospe M, 2005, HuR: posttranscriptional paths to malignancy. RNA Biol 2:11–13 Kotta-Loizou I, Giaginis C, Theocharis S, 2014, Clinical significance of HuR expression in human malignancy. Med Oncol 31:161 Giaginis C, Alexandrou P, Tsoukalas N, et al., 2015, Hu-antigen receptor, HuR, and cyclooxygenase-2, COX-2, expression in human non-small cell lung carcinoma: associations with clinicopathological parameters, tumor proliferative capacity and patients’ survival. Tumour 36:315–327 Abdelmohsen K, Srikantan S, Kuwano Y, GorospeM, 2008, miR- 519 reduces cell proliferation by lowering RNA-binding protein HuR levels. Proc Natl Acad Sci U S A 105:20297–20302 Abdelmohsen K, Kim MM, Srikantan S, et al., 2010, miR-519 suppresses tumor growth by reducing HuR levels. Cell Cycle 9: 1354–1359 Srikantan S, Gorospe M, 2012, HuR function in disease. Front Biosci, Landmark Ed, 17:189–205 Rosai J, 2004, Appendix C. Staging of cancer. In: Houston M, ed, Rosai and Ackerman’s Surgical Pathology, 9th edn. Mosby, London, pp. 2809–2810 Tuttle RM, Tala H, Shah J, et al., 2010, Estimating risk of recurrence in differentiated thyroid cancer after total thyroidectomy and radioactive iodine remnant ablation: using response to therapy variables to modify the initial risk estimates predicted by the new American thyroid association staging system. Thyroid 20:1341 Michailidi C, Giaginis C, Stolakis V, et al., 2010, Evaluation of FAK and Src expression in human benign and malignant thyroid lesions. Pathol Oncol Res 16:497–507 Giaginis C, Alexandrou P, Delladetsima I, Giannopoulou I, Patsouris E, Theocharis S, 2014, Clinical significance of histone deacetylase, HDAC)-1, HDAC-2, HDAC-4, and HDAC-6 expression in human malignant and benign thyroid lesions. Tumour Biol 35:61–71 Karidis NP, Giaginis C, Tsourouflis G, et al., 2011, Eph-A2 and Eph-A4 expression in human benign andmalignant thyroid lesions: an immunohistochemical study. Med Sci Monit 17:BR257–BR265 Cha JD, Li S, Cha IH, 2011, Association between expression of embryonic lethal abnormal vision-like protein HuR and cyclooxygenase-2 in oral squamous cell carcinoma. Head Neck 33:627–637 Zhang C, Xue G, Bi J, et al., 2014, Cytoplasmic expression of the ELAV-like protein HuR as a potential prognostic marker in esophageal squamous cell carcinoma. Tumour Biol 35:73–80 Wang J, Wang B, Bi J, Zhang C, 2011, Cytoplasmic HuR expression correlates with angiogenesis, lymphangiogenesis, and poor outcome in lung cancer. Medical Oncol 28:S577–S585 Lim SJ, Kim HJ, Kim JY, Park K, Lee CM, 2007, Expression of HuR is associated with increased cyclooxygenase-2 expression in uterine cervical carcinoma. Int J Gynecol Pathol 26:229–234 Liang PI, Li WM, Wang YH, et al., 2012, HuR cytoplasmic expression is associated with increased cyclin A expression and poor outcome with upper urinary tract urothelial carcinoma. BMC Cancer 12:611 Sun DP, Lin CY, Tian YF, et al., 2013, Clinicopathological significance of HuR expression in gallbladder carcinoma: with special emphasis on the implications of its nuclear and cytoplasmic expression. Tumour Biol 34:3059–3069 Mrena J,Wiksten JP, KokkolaA, Nordling S,Haglund C, Ristimaki A, 2006, Prognostic significance of cyclin a in gastric cancer. Int J Cancer 119:1897–1901 Denkert C, Koch I, von Keyserlingk N, et al., 2006, Expression of the ELAV-like protein HuR in human colon cancer: association with tumor stage and cyclooxygenase-2. Mod Pathol 19: 1261–1269 Zhu Z, Wang B, Bi J, et al., 2013, Cytoplasmic HuR expression correlates with P-gp, HER-2 positivity, and poor outcome in breast cancer. Tumour Biol 34:2299–22308 Erkinheimo TL, Lassus H, Sivula A, et al., 2003, CytoplasmicHuR expression correlates with poor outcome and with cyclooxygenase 2 expression in serous ovarian carcinoma. Cancer Res 63: 7591–7594 Marechal R, Van Laethem JL, 2009, HuR modulates gemcitabine efficacy: new perspectives in pancreatic cancer treatment. Expert Rev Anticancer Ther 9:1439–1441 Krawczyk-Rusiecka K, Lewinski A, 2010, Cyclooxygenase-2 expression and its association with thyroid lesions. Arch Med Sci 6: 653–657 Casey MB, Zhang S, Jin L, Kajita S, Lloyd RV, 2004, Expression of cyclooxygenase-2 and thromboxane synthase in non-neoplastic and neoplastic thyroid lesions. Endocr Pathol 15:107–116 Lee KJ, Jung YS, Kim WH, Yoon TI, Joo HJ, Soh EY, 2008, Cyclooxygenase-2 expression in human thyroid disease. J Endocrinol Investig 31:111–118 Lee HM, Baek SK,Kwon SY, et al., 2006, Cyclooxygenase 1 and 2 expressions in the human thyroid gland. Eur Arch Otorhinolaryngol 263:199–204 Garcia-Gonzalez M, Abdulkader I, Boquete AV, Neo XM, Forteza J, Cameselle-Teijeiro J, 2005, Cyclooxygenase-2 in normal, hyperplastic and neoplastic follicular cells of the human thyroid gland. Virchows Arch 447:12–17 Ito Y, Yoshida H, Nakano K, et al., 2003, Cyclooxygenase-2 expression in thyroid neoplasms. Histopathology 42:492–497 Siironen P, Ristimaki A, Nordling S, Louhimo J, Haapiainen R, Haglund C, 2004, Expression of COX-2 is increased with age in papillary thyroid cancer. Histopathology 44:490–497 Filippova N, Yang X, Wang Y, et al., 2011, The RNA-binding protein HuR promotes glioma growth and treatment resistance. Mol Cancer Res 9:648–659 Hostetter C, Licata LA, Witkiewicz A, et al., 2008, Cytoplasmic accumulation of the RNA binding protein HuR is central to tamoxifen resistance in estrogen receptor positive breast cancer cells. Cancer Biol Ther 7:1496–1506 Costantino CL, Witkiewicz AK, Kuwano Y, et al., 2009, The role of HuR in gemcitabine efficacy in pancreatic cancer: HuR upregulates the expression of the gemcitabine metabolizing enzyme deoxycytidine kinase. Cancer Res 69:4567–4572 Williams TK, Costantino CL, Bildzukewicz NA, et al., 2010, pp32, ANP32A, expression inhibits pancreatic cancer cell growth and induces gemcitabine resistance by disrupting HuR binding to mRNAs. PLoS One 5:e15455 McAllister F, Pineda DM, Jimbo M, et al., 2014, dCK expression correlates with 5-fluorouracil efficacy and HuR cytoplasmic expression in pancreatic cancer: a dual-institutional follow-up with the RTOG 9704 trial. Cancer Biol Ther 15:688–698 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 189 EP 196 DI 10.1007/s12253-015-9997-5 PG 8 ER PT J AU Chen, YCh Fang, WL Wang, RF Liu, ChA Yang, MH Lo, ShSh Wu, ChW Li, FYA Shyr, YM Huang, KH AF Chen, Yun-Chi Fang, Wen-Liang Wang, Ruei-Fang Liu, Chien-An Yang, Muh-Hwa Lo, Shu-Shun Wu, Chew-Wen Li, Fen-Yau Anna Shyr, Yi-Ming Huang, Kuo-Hung TI Clinicopathological Variation of Lauren Classification in Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Gastrecotomy; Lauren’s classification ID Gastric cancer; Gastrecotomy; Lauren’s classification AB The investigation of prognostic factor for gastric cancer is still desirable because of dismal prognosis in gastric cancer. Lauren’s classification is currently a useful histological classification. There are few large series evaluating the prognostic significance of Lauren’s classification in gastric cancer. From January 1987 to December 2013, a total of 3071 patients received gastrectomy for gastric cancer. According Lauren’s classification, 1423(46.3 %) patients were intestinal type, 1000 patients (32.6 %) were diffuse type, and 648 patients (21.1 %) were mixed type. The clinicopathological characteristics and prognosis in Lauren’s classification were analyzed in these patients. Our results showed that patients with intestinal type gastric cancer (57.7%) had a better 5-year overall survival than diffuse type (45.6 %) and mixed type (43.4 %, P<0.001). The clinicopathological characteristics showed that gastric cancer patients with intestinal type were older (P<0.001), male predominant (P<0.001), smaller tumor size (P<0.001), distal stomach predominant (P<0.001), relative well differentiated (P<0.001), less advanced Borrmann type (P<0.001), less scirrhous type stromal reaction(P<0.001), less infiltrating type of Ming’s histology type(P<0.001), less tumor invasion depth and less lymphovascular invasion (P<0.001). Multivariate analysis with overall survival as an endpoint showed that age (P=0.005), Borrmann classification (P<0.001), pathological T category (P=0.023), pathological N category (P<0.001) and Lauren’s classification (P=0.003) were significant correlated in gastric cancer. Lauren’s classification is an independent prognostic factor in gastric cancer patient undergoing gastrectomy. Lauren’s classification can serve as a prognostic marker for gastric cancer patient receiving gastrectomy. The clinicopathological appearance and prognosis of mixed type gastric cancer is similar to diffuse type gastric cancer. C1 [Chen, Yun-Chi] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No.201, Sec. 2, Shipai Rd., Beitou District, 11217 Taipei, Taiwan, Republic of China. [Fang, Wen-Liang] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No.201, Sec. 2, Shipai Rd., Beitou District, 11217 Taipei, Taiwan, Republic of China. [Wang, Ruei-Fang] Shin Kong Wu Ho-Su Memorial Hospital, Department of Emergency MedicineTaipei, Taiwan, Republic of China. [Liu, Chien-An] National Yang-Ming University, School of MedicineTaipei, Taiwan, Republic of China. [Yang, Muh-Hwa] National Yang-Ming University, School of Medicine, Institute of Clinical MedicineTaipei, Taiwan, Republic of China. [Lo, Shu-Shun] National Yang-Ming University, School of MedicineTaipei, Taiwan, Republic of China. [Wu, Chew-Wen] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No.201, Sec. 2, Shipai Rd., Beitou District, 11217 Taipei, Taiwan, Republic of China. [Li, Fen-Yau Anna] National Yang-Ming University, School of MedicineTaipei, Taiwan, Republic of China. [Shyr, Yi-Ming] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No.201, Sec. 2, Shipai Rd., Beitou District, 11217 Taipei, Taiwan, Republic of China. [Huang, Kuo-Hung] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No.201, Sec. 2, Shipai Rd., Beitou District, 11217 Taipei, Taiwan, Republic of China. RP Huang, KH (reprint author), Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, 11217 Taipei, Taiwan, Republic of China. EM khhuang@vghtpe.gov.tw CR Taiwan Public Health Report 2012, Department of Health, Executive Yuan, R.O.C., Taiwan) Wu CW, Hsiung CA, Lo SS et al, 2006, Nodal dissection for patients with gastric cancer: a randomized controlled trial. Lancet Oncol 7:309–315 Hartgrink HH, van de Velde CJH, Putter H et al, 2004, Extended lymph-node dissection for gastric cancer: who may benefit? final results of the randomized Dutch gastric cancer group trial. J Clin Oncol 22:2069–2077 Lauren P, 1965, The two histologic main types of gastric carcinoma: diffuse and so-called intestinal type carcinoma, an attempt at a histo-clinicalclassification. Acta Parhol Microb Scan 64:31–49 Qiu MZ, Cai MY, Zhang DS et al, 2013, Clinicopathological characteristics and prognostic analysis of lauren classification in gastric adenocarcinoma in China. J Transl Med 11:58 Berlth F, Bollschweiler E, Drebber U et al, 2014, Pathohistological classification systems in gastric cancer: diagnostic relevance and prognostic value. World J Gastroenterol 20(19):5679–5684 Ming SC, 1977, Gastric carcinoma: a pathological classification. Cancer 39(6):2475–2485 American Joint Committee on Cancer, 2010, AJCC cancer staging manual, 7th edn. Springer, New York Komatsu S, Ichikawa D, Miyamae M et al, 2015, Histological mixed-type as an independent prognostic factor in stage I gastric carcinoma. World J Gastroenterol 21(2):549–555 Zheng HC, Li XH, Hara T et al, 2008, Mixed type gastric carcinoma exhibit more aggressive features and indicate the histogenesis of carcinomas. Virchows Arch 452(5):525–534 GravalosC, JimenoA(2008)HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol 19(9):1523–1529 Qiu M, Zhou Y, Zhong X et al, 2014, Lauren classification combined with HER2 status is a better prognostic factor in Chinese gastric cancer patients. BMC Cancer 14:823 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 197 EP 202 DI 10.1007/s12253-015-9996-6 PG 6 ER PT J AU Glowacka, I Nowikiewicz, T Siedlecki, Z Hagner, W Nowacka, K Zegarski, W AF Glowacka, Iwona Nowikiewicz, Tomasz Siedlecki, Zygmunt Hagner, Wojciech Nowacka, Krystyna Zegarski, Wojciech TI The Assessment of the Magnitude of Frontal Plane Postural Changes in Breast Cancer Patients After Breast-Conserving Therapy or Mastectomy – Follow-up Results 1 Year After the Surgical Procedure SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast conserving therapy; Mastectomy; Computed-assisted postural assessment; Body posture; Frontal plane ID Breast conserving therapy; Mastectomy; Computed-assisted postural assessment; Body posture; Frontal plane AB Breast cancer is the most common malignancy in Polish women. Management of breast cancer includes surgical treatment as well as adjuvant chemotherapy, radiotherapy, hormonal therapy, and combination regimens. One of the adverse consequences of oncological management of breast cancer may involve changes in frontal plane body posture. The objective of the study was to assess the frontal plane body posture changes in women treated for breast cancer. A prospective study including 101 of female breast cancer patients subjected to surgical treatment in the period from October 2011 to October 2012 (mastectomy was performed in 51 cases while breast conserving therapy was administered in the remaining 50 cases). The body posture in the frontal plane was assessed using the computerassisted postural assessment system with Moire fringe analysis. No statistically significant differences were observed in pre-operational postural parameters of interest. Exam II revealed highly significant differences in SLA values; results suggesting more pronounced dysfunction were observed in the MAS group. Exam III revealed highly significant differences in PIA, SH, SD and SLA values; results suggesting more pronounced dysfunction were observed in the MAS group. Undesirable postural changes occur both in women who were treated with radical mastectomy and in those who underwent breast-conserving surgery; breast-conserving surgery is associated with decreased severity in postural abnormalities; C1 [Glowacka, Iwona] Collegium Medicum of the Nicolaus Copernicus University in Torun, Department of RehabilitationBydgoszcz, Poland. [Nowikiewicz, Tomasz] Bydgoszcz Oncology Center, Clinical Department of Breast Cancers and Breast Reconstruction SurgeryBydgoszcz, Poland. [Siedlecki, Zygmunt] Collegium Medicum of the Nicolaus Copernicus University in Torun, Department of NeurosurgeryBydgoszcz, Poland. [Hagner, Wojciech] Collegium Medicum of the Nicolaus Copernicus University in Torun, Department of RehabilitationBydgoszcz, Poland. [Nowacka, Krystyna] Collegium Medicum of the Nicolaus Copernicus University in Torun, Department of RehabilitationBydgoszcz, Poland. [Zegarski, Wojciech] Medical College of the Nicolaus Copernicus University in Torun, Clinic and Chair of Oncological SurgeryBydgoszcz, Poland. RP Glowacka, I (reprint author), Collegium Medicum of the Nicolaus Copernicus University in Torun, Department of Rehabilitation, Bydgoszcz, Poland. EM iwona.glowacka@cm.umk.pl CR Devoogdt N, Van Kampen M, Christiaens MR et al, 2011, Shortand long-term recovery of upper limb function after axillary lymph node dissection. Eur J Cancer Care, Engl, 20:77–86 Favarao KU, Mantese JC, Barros AC, 2010, Shoulder mobility after axillary sentinel node biopsy for early infiltrating breast cancer treatment. Eur J Gynaecol Oncol 31:23–26 Yang EJ, Park WB, Seo KS et al, 2010, Longitudinal change of treatment-related upper limb dysfunction and its impact on late dysfunction in breast cancer surviviors: a prospective cohort study. J Surg Oncol 101:84–91 Ferrell BR, Grant MM, Funk B et al, 1997, Quality of life in breast cancer survivors as identified focus groups. Psychooncology 6:13–23 Crosible J, Kilbreath SL, Dylke E et al, 2010, Effect ofmastectomy on shoulder and spinal kinematics during bilateral upper-limb movement. Phys Ther 90:679–692 Crosible J, Kilbreath SL, Hollman L et al, 2008, Scapulohumeral rhythm and associated spinal motion. Clin Biomech 23:184–192 Keramopoulos A, Tsionou C, Minaretzis D et al, 1993, Arm morbidity following treatment of breast cancer with total axillary dissection: a multivariated approach. Oncology 50:445–449 McClure PW, Michener LA, Sennett BJ et al, 2001, Direct 3- dimensional measurement of scapular kinematics during dynamic movements in vivo. J Shoulder Elbow Surg 10:269–277 Bentzen SM, Dische S, 2000, Morbidity related to axillary irradiation in the treatment of breast cancer. Acta Oncol 39:337–347 Tanis PJ, Nieweg OE, Valdes Olmos RA, Kroon BB, 2001, Anatomy and physiology of lymphatic drainage of the breast from the perspective of sentinel node biopsy. J Am Coll Surg 192:399– 409 Tanis PJ, Nieweg OE, Valdes Olmos RA, 2001, History of sentinel node and validation of the technique. Breast Cancer Res 3:109–112 Zavagno G, Rubello D, Franchini Z et al, 2005, Axillary sentinel lymph nodes in breast cancer: a single lymphatic pathway drains the en tire mammary gland. Eur J Surg Oncol 31:479–484 Rietman JS, Dikstra PU, Hoekstra HJ et al, 2003, Late morbidity after treatment of breast cancer in relation to daily activities and quality of life: a systemic review. Eur J Surg Oncol 29:229–238 Hojan K, Wruk B, Stryla W, 2011, Wplyw leczenia raka piersi na dolegliwosci bolowe kregoslupa w odcinku ledzwiowokrzyzowym. Onkologia Polska 13:177–184 Ciesla S, Polom K, 2010, The effect of immediate breast reconstruction with Becker-25 prosthesis on the preservation of proper body posture in patients after mastectomy. Eur J Surg Oncol 36(7): 625–631 Bak M, Rostkowska E, 2000, The influence of using breast prothesis during the night on the changes of body posture among women after mastectomy. Fizjoterapia 8(4):11–15 Adderley-Kelly B, Williams-Stephens E, 2003, The relationsship between obesity and breast cancer. ABNF Journal 14(3):61–65 Segal R, Evans W, Johnson D et al, 2001, Structured exercise improves physicalfunctioning in women with stages I and II breast cancer: results of a randomized controlled trial. J Clin Oncol 19: 657–665 Fisher B, Redmond C, Poisson R et al, 1989, Eight-year results of a randomised trial comparing total mastectomy and lumpectomy with or without irradiation in the treatment of breast cancer. N Engl J Med 320:822–828 Liljegren G, Holmberg M, Adami HO et al, 1994, Sector resection with or without postoperative radiotherapy for stage I breast cancer: five-year results of a randomised trial. J Natl Cancer Inst 86:717– 722 Clark AC, McColloch PB, Levine MN et al, 1992, Randomised clinical trial to assess the effectiveness of breast irradiation following lumpectomy and axillary dissection for node-negative breast cancer. J Natl Cancer Inst 84:683–689 Veronesi U, Luini A, Del VecchioMet al, 1993, Radiotherapy after breast-preserving surgery in women with localized cancer of the breast. N Engl J Med 328:1587–1589 Ragaz J, Jackson SM, Le N et al, 1997, Adjuvant radiotherapy and chemotherapy in node-positive premenopausal women with breast cancer. N Engl J Med 337:956–962 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 203 EP 208 DI 10.1007/s12253-015-9995-7 PG 6 ER PT J AU Toth, D Varga, Zs Sebo, Torok, M Kovacs, I AF Toth, Dezso Varga, Zsolt Sebo, Eva Torok, Miklos Kovacs, Ilona TI Predictive Factors for Positive Margin and the Surgical Learning Curve in Non-Palpable Breast Cancer After Wire-Guided Localization – Prospective Study of 214 Consecutive Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Surgery; Non-palpable; Wire-guided localization; Learning curve ID Breast cancer; Surgery; Non-palpable; Wire-guided localization; Learning curve AB To investigate the most commonly used technique, the wire-guided localization (WGL) in non-palpable breast cancer. To analyze the effective factors on positive surgical margins in our practice and determine the surgical learning curve of this method. Prospective consecutive study was performed from January 2005 to December 2011. Inclusion criteria was a non-palpable breast lesion with malignancy on preoperative histology. All lesions were localized by ultrasound or stereotactic guided wire placement. Margins 1 mm or closer were accepted as positive margins which required reexcision. To determine the learning curve of WGL method we investigated the change in the reoperation rate after primary procedure performed by "high-volume" surgeon. Two hundred and fourteen consecutive patients were enrolled. In 23 patients (10.7 %) reexcision was needed. Positive surgical margins were significantly influenced by the patient’s age (p=0.03), tumor volume (p<=0.001), proportion of tumor volume/specimen volume (p<0.001), presence of DCIS (p<0.001), multifocality (p=0.03) and the learning curve (p=0.006) with univariate analysis. Only the tumor volume, presence of DCIS and the learning curve were proved as independent prognostic factor for reoperation by multivariate analysis. The reoperation rate decreased below 20 % after the fortieth operation. Results of our single institutional study suggest, that this localization technique can be performed safely with very good results after 40 procedures as a learning curve for surgeons. C1 [Toth, Dezso] Kenezy Teaching Hospital, Department of General Surgery, 2-26 Bartok Street, 4031 Debrecen, Hungary. [Varga, Zsolt] Kenezy Teaching Hospital, Department of General Surgery, 2-26 Bartok Street, 4031 Debrecen, Hungary. [Sebo, Eva] Kenezy Teaching Hospital, Kenezy Breast Center, 21 Jeriko Street, 4032 Debrecen, Hungary. [Torok, Miklos] Kenezy Teaching Hospital, Department of Pathology, 2-26 Bartok Street, 4031 Debrecen, Hungary. [Kovacs, Ilona] Kenezy Teaching Hospital, Department of Pathology, 2-26 Bartok Street, 4031 Debrecen, Hungary. RP Toth, D (reprint author), Kenezy Teaching Hospital, Department of General Surgery, 4031 Debrecen, Hungary. EM detoth@gmail.com CR ParkinM, Bray F, Ferlay J, Pisani P, 2002, Global cancer statistics. Cancer J Clin 55:74–108 Skinner KA, Silberman H, Sposto R, Silverstein MJ, 2001, Palpable breast cancers are inherently different from nonpalpable breast cancers. Ann Surg Oncol 8(9):705–710 Blanks RG, Moss SM, McGahan CE, Quinn MJ, Babb PJ, 2000, Effect of NHS breast screening programme on mortality from breast cancer in England and Wales, 1990–8: comparison of observed with predicted mortality. BMJ 321:665–669 Gray RJ, Salud C, Nguyen K et al, 2001, Randomized prospective evaluation of a novel technique for biopsy or lumpectomy of nonpalpable breast lesions: radioactive seed versus wire localization. Ann Surg Oncol 8(9):711–715 Medina-Franco H, Abarca-Perez L, Garcia-Alvarez MN, Ulloa- Gomez JL, Romero-Trejo C, Sepulveda-Mendez J, 2008, Radioguided occult lesion localization, ROLL, versus wireguided lumpectomy for non-palpable breast lesions: a randomized prospective evaluation. J Surg Oncol 97(2):108–111 Ocal K, Dag A, Turkmenoglu O, Gunay EC, Yucel E, Duce MN, 2011, Radioguided occult lesion localization versus wire-guided localization for non-palpable breast lesions: randomized controlled trial. Clinics, Sao Paulo, 66(6):1003–1007 Lovrics PJ, Cornacchi SD, Farrokhyar F et al, 2009, The relationship between surgical factors and margin status after breastconservation surgery for early stage breast cancer. Am J Surg 197(6):740–746 Toth D, Sebo E, Sarkadi L, Kovacs I, Kiss C, Damjanovich L, 2012, Role of core needle biopsy in the treatment of radial scar. Breast 21(6):761–763 Postma EL,Witkamp AJ, van den BoschMA, Verkooijen HM, van Hillegersberg R, 2011, Localization of nonpalpable breast lesions. Expert Rev Anticancer Ther 11(8):1295–1302 Besic N, Zgajnar J, HocevarMet al, 2002, Breast biopsy with wire localization: factors influencing complete excision of nonpalpable carcinoma. Eur Radiol 12(11):2684–2689 Davis PS,Wechsler RJ, Feig SA, MarchDE, 1988)Migration of breast biopsy localization wire. AJR Am J Roentgenol 150(4):787–788 Rahusen FD, Bremers AJ, Fabry HF, van Amerongen AH, Boom RP, Meijer S, 2002, Ultrasound-guided lumpectomy of nonpalpable breast cancer versus wire-guided resection: a randomized clinical trial. Ann Surg Oncol 9:994–998 Homer MJ, 1983, Transection of the localization hooked wire during breast biopsy. Am J Roentgenol 141:929–930 Tykka H, Castren-Person M, Sjoblom M, 1993, Pneumothorax caused by hooked wire localization of an impalpable breast lesion detected by mammography. Breast 2:52–53 Jeevan R, Cromwell DA, Trivella M et al, 2012, Reoperation rates after breast conserving surgery for breast cancer among women in England: retrospective study of hospital episode statistics. BMJ 345:e4505 Takacs T, Paszt A, Simonka Z et al, 2013, Radioguided occult lesion localisation versus wire-guided lumpectomy in the treatment of non-palpable breast lesions. Pathol Oncol Res 19(2):267–273 Bernardi S, Bertozzi S, Londero AP, Gentile G, Giacomuzzi F, Carbone A, 2012, Incidence and risk factors of the intraoperative localization failure of nonpalpable breast lesions by radio-guided occult lesion localization: a retrospective analysis of 579 cases. World J Surg 36(8):1915–1921 Wazer DE, Schmidt-Ullrich RK, Ruthazer R et al, 1999, The influence of age and extensive intraductal component histology upon breast lumpectomy margin assessment as a predictor of residual tumor. Int J Radiat Oncol Biol Phys 45(4):885–891 Smitt MC, Horst K, 2007, Association of clinical and pathologic variables with lumpectomy surgicalmargin status after preoperative diagnosis or excisional biopsy of invasive breast cancer. Ann Surg Oncol 14(3):1040–1044 Sanchez C, Brem RF, McSwain AP, Rapelyea JA, Torrente J, Teal CB, 2010, Factors associated with re-excision in patients with early-stage breast cancer treated with breast conservation therapy. Am Surg 76(3):331–334 Lovrics PJ, Cornacchi SD, Vora R, Goldsmith CH, Kahnamoui K, 2011, Systematic review of radioguided surgery for non-palpable breast cancer. Eur J Surg Oncol 37(5):388–397 Del Turco MR, Ponti A, Bick U, Biganzoli L et al, 2010, Quality indicators in breast cancer care. Eur J Cancer 46(13):2344–2356 AhmedM, DouekM(2013, Radioactive seed localisation, RSL, in the treatment of non-palpable breast cancers: systematic review and meta-analysis. Breast 22(4):383–388 Postma EL, Verkooijen HM, van Esser S et al, 2012, Efficacy of ‘radioguided occult lesion localisation’, ROLL, versus ‘wire-guided localisation’, WGL, in breast conserving surgery for nonpalpable breast cancer: a randomised controlled multicentre trial. Breast Cancer Res Treat 136(2):469–478 Cleffken B, Postelmans J, Olde Damink S, Nap M, Schreutelkamp I, van der Bijl H, 2007, Breast-conserving therapy for palpable and nonpalpable breast cancer: can surgical residents do the job irrespective of experience? World J Surg 31(9):1731–1736 Landheer ML, Hoorntje LE, Klinkenbijl JH, Borel Rinkes IH, 2004, The surgical treatment of nonpalpable breast carcinoma in a university teaching hospital and a general teaching hospital by residents-in-training and surgeons; comparable results. Ned Tijdschr Geneeskd 148(35):1724–1727 Edge SB, Byrd DR, Compton CC et al, eds,, 2010, AJCC cancer staging manual, 7th edn. Springer, New York, pp 347–376 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 209 EP 215 DI 10.1007/s12253-015-9999-3 PG 7 ER PT J AU Ujj, Zs Buglyo, G Udvardy, M Beyer, D Vargha, Gy Biro, S Rejto, L AF Ujj, Zsofia Buglyo, Gergely Udvardy, Miklos Beyer, Daniel Vargha, Gyorgy Biro, Sandor Rejto, Laszlo TI WT1 Expression in Adult Acute Myeloid Leukemia: Assessing its Presence, Magnitude and Temporal Changes as Prognostic Factors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE WT1 gene; Overexpression; Quantitative real-time PCR; Acute myeloid leukemia ID WT1 gene; Overexpression; Quantitative real-time PCR; Acute myeloid leukemia AB Expression of the gene Wilms tumor 1 (WT1) has been suggested as a marker of minimal residual disease in acute myeloid leukemia (AML), but literature data are not without controversy. Our aimwas to assess the presence, magnitude and temporal changes of WT1 expression as prognostic factors. 60 AML patients were followed until death or the end of the 6-year observation period. Blood samples were taken at diagnosis, post-induction, during remission and in case of a relapse. Using quantitative real-time PCR, we determined WT1 expression from each sample, normalized it against the endogenous control gene glyceraldehyde 3-phosphate dehydrogenase (GAPDH), and classified samples as negative, moderately positive or highly positive. We divided the patients into groups based on detected WT1 expression values, illustrated overall and disease-free survival on Kaplan-Meier curves, and compared differences between each group by the logrank test. Disappearance of WT1-positivity during chemotherapy had a favorable effect on survival. Interestingly, no difference was seen between the survivals of WT1-positive subgroups that expressed moderate or high levels of WT1 mRNA. A 1-log decrease in WT1 expression without becoming negative did not affect prognosis, either. Our results suggest that defining a cut-off value for WT1-positivity, rather than just using logarithmic figures of changes in gene expression, might have prognostic use in post-induction AML patients. We encourage further, larger-scale studies. C1 [Ujj, Zsofia] University of Debrecen, Faculty of Medicine, Department of Hematology, 98 Nagyerdei korut, 4028 Debrecen, Hungary. [Buglyo, Gergely] University of Debrecen, Department of Human Genetics, 98 Nagyerdei korut, 4028 Debrecen, Hungary. [Udvardy, Miklos] University of Debrecen, Faculty of Medicine, Department of Hematology, 98 Nagyerdei korut, 4028 Debrecen, Hungary. [Beyer, Daniel] University of Debrecen, Department of Human Genetics, 98 Nagyerdei korut, 4028 Debrecen, Hungary. [Vargha, Gyorgy] University of Debrecen, Department of Human Genetics, 98 Nagyerdei korut, 4028 Debrecen, Hungary. [Biro, Sandor] University of Debrecen, Department of Human Genetics, 98 Nagyerdei korut, 4028 Debrecen, Hungary. [Rejto, Laszlo] University of Debrecen, Faculty of Medicine, Department of Hematology, 98 Nagyerdei korut, 4028 Debrecen, Hungary. RP Buglyo, G (reprint author), University of Debrecen, Department of Human Genetics, 4028 Debrecen, Hungary. EM gbuglyo@hotmail.com CR Rose EA, Glaser T, Jones C, Smith CL, Lewis WH, Call KM, Minden M, Champagne E, Bonetta L, Yeger H, Housman DE, 1990, Complete physical map of the WAGR region of 11p13 localizes a candidate Wilms' tumor gene. Cell 60:495–508 CallKM, Glaser T, Ito CY, Buckler AJ, Pelletier J, Haber DA, Rose EA, Kral A, Yeger H, Lewis WH, 1990, Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms' tumor locus. Cell 60:509–520 Essafi A, Webb A, Berry RL, Slight J, Burn SF, Spraggon L, Velecela V, Martinez-Estrada OM, Wiltshire JH, Roberts SG, Brownstein D, Davies JA, Hastie ND, Hohenstein P, 2011, A wt1-controlled chromatin switching mechanism underpins tissuespecific wnt4 activation and repression. Dev Cell 21:559–574 Morrison AA, Viney RL, Ladomery MR, 2008, The posttranscriptional roles of WT1, a multifunctional zinc-finger protein. Biochim Biophys Acta 1785:55–62 Haber DA, Sohn RL, Buckler AJ, Pelletier J, Call KM, Housman DE, 1991, Alternative splicing and genomic structure of theWilms tumor gene WT1. Proc Natl Acad Sci U S A 88:9618–9622 Miller-Hodges E, Hohenstein P, 2012)WT1 in disease: shifting the epithelial-mesenchymal balance. J Pathol 226:229–240 Miwa H, Beran M, Aunders GF, 1992, Expression of the Wilms tumor gene, WT1, in human leukemias. Leukemia 6:405–409 Vidovic K, Svensson E, Nilsson B, Thuresson B, Olofsson T, Lennartsson A, Gullberg U, 2010, Wilms' tumor gene 1 protein represses the expression of the tumor suppressor interferon regulatory factor 8 in human hematopoietic progenitors and in leukemic cells. Leukemia 24:992–1000 Bergmann L, Miething C, Maurer U, Brieger J, Karakas T, Weidmann E, Hoelzer D, 1997, High levels of Wilms' tumor gene, wt1, mRNA in acute myeloid leukemias are associated with a worse long-term outcome. Blood 90:1217–1225 Gaiger A, Schmid D, Heinze G, Linnerth B, Greinix H, Kalhs P, Tisljar K, Priglinger S, Laczika K, Mitterbauer M, Novak M, Mitterbauer G,Mannhalter C, Haas OA, Lechner K, Jager U, 1998, Detection of the WT1 transcript by RT-PCR in complete remission has no prognostic relevance in de novo acute myeloid leukemia. Leukemia 12:1886–1894 Noronha SA, Farrar JE, Alonzo TA, Gerbing RB, Lacayo NJ, Dahl GV, Ravindranath Y, Arceci RJ, LoebDM(2009)WT1 expression at diagnosis does not predict survival in pediatric AML: a report from the children's oncology group. Pediatr Blood Cancer 53:1136–1139 Mossallam GI, Hamid TM, Mahmoud HK, 2013, Prognostic significance of WT1 expression at diagnosis and end of induction in Egyptian adult acute myeloid leukemia patients. Hematology 18: 69–73 Gray JX, McMillen L, Mollee P, Paul S, Lane S, Bird R, Gill D, Saal R, Marlton P, 2012, WT1 expression as a marker of minimal residual disease predicts outcome in acute myeloid leukemia when measured post-consolidation. Leuk Res 36:453–458 Andersson C, Li X, Lorenz F, Golovleva I, Wahlin A, Li A, 2012, Reduction in WT1 gene expression during early treatment predicts the outcome in patients with acute myeloid leukemia. Diagn Mol Pathol 21:225–233 Polak J, Hajkova H, Haskovec C, Cechova H, Marinov I, Mikulenkova D, Markova J, Markova M, Vitek A, Valkova V, 2013, Quantitative monitoring of WT1 expression in peripheral blood before and after allogeneic stem cell transplantation for acute myeloid leukemia - a useful tool for early detection of minimal residual disease. Neoplasma 60:74–82 Cilloni D, Messa F, Arruga F, Defilippi I, Gottardi E, Fava M, Carturan S, Catalano R, Bracco E, Messa E, Nicoli P, Diverio D, Sanz MA, Martinelli G, Lo-Coco F, Saglio G, 2008, Early prediction of treatment outcome in acute myeloid leukemia by measurement of WT1 transcript levels in peripheral blood samples collected after chemotherapy. Haematologica 93:921–924 Cilloni D, Renneville A, Hermitte F, Hills RK, Daly S, Jovanovic JV, Gottardi E, FavaM, Schnittger S,Weiss T, Izzo B, Nomdedeu J, van der Heijden A, van der Reijden BA, Jansen JH, van der Velden VH, Ommen H, Preudhomme C, Saglio G, Grimwade D, 2009, Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: a European LeukemiaNet study. J Clin Oncol 27:5195–5201 Cilloni D, Saglio G, 2004)WT1 as a universal marker for minimal residual disease detection and quantification in myeloid leukemias and in myelodysplastic syndrome. Acta Haematol 112:79–84 Ogawa H, Ikegame K, Kawakami M, Tamaki H, 2004, WT1 gene transcript assay for relapse in acute leukemia after transplantation. Leuk Lymphoma 45:1747–1753 Gianfaldoni G, Mannelli F, Ponziani V, Longo G, Bencini S, Bosi A, Vannucchi AM, 2010, Early reduction of WT1 transcripts during induction chemotherapy predicts for longer disease free and overall survival in acute myeloid leukemia. Haematologica 95: 833–836 Sugiyama H, 2010, WT1, Wilms' Tumor gene 1): biology and cancer immunotherapy. Jpn J Clin Oncol 40:377–387 Renneville A, Boissel N, Zurawski V, Llopis L, Biggio V, Nibourel O, Philippe N, Thomas X, Dombret H, Preudhomme C, 2009, Wilms tumor 1 gene mutations are associated with a higher risk of recurrence in young adults with acute myeloid leukemia: a study from the acute leukemia French association. Cancer 115:3719– 3727 Hou HA, Huang TC, Lin LI, Liu CY, Chen CY, Chou WC, Tang JL, Tseng MH, Huang CF, Chiang YC, Lee FY, Liu MC, Yao M, Huang SY, Ko BS, Hsu SC, Wu SJ, Tsay W, Chen YC, Tien HF, 2010, WT1 mutation in 470 adult patients with acute myeloid leukemia: stability during disease evolution and implication of its incorporation into a survival scoring system. Blood 115:5222–5231 Huff V, 2011)Wilms' tumours: about tumour suppressor genes, an oncogene and a chameleon gene. Nat Rev Cancer 11:111–121 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 217 EP 221 DI 10.1007/s12253-015-0002-0 PG 5 ER PT J AU Oh, RH Choi, JY Yoo, JN Lee, HS AF Oh, Rim Hye Choi, Jin Youn Yoo, Jin Nam Lee, Hyung Sug TI Leukemia Relapse-Associated Mutation of NT5C2 Gene is Rare in de Novo Acute Leukemias and Solid Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Oh, Rim Hye] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Choi, Jin Youn] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Tozzi MG, Camici M, Pesi R, Allegrini S, Sgarrella F, Ipata PL, 1991, Nucleoside phosphotransferase activity of human colon carcinoma cytosolic 5′-nucleotidase. Arch Biochem Biophys 291:212– 217 Meyer JA, Wang J, Hogan LE, Yang JJ, Dandekar S, Patel JP, Tang Z, Zumbo P, Li S, Zavadil J, Levine RL, Cardozo T, Hunger SP, Raetz EA, Evans WE, Morrison DJ, Mason CE, Carroll WL, 2013, Relapse-specific mutations in NT5C2 in childhood acute lymphoblastic leukemia. Nat Genet 45:290–294 Tzoneva G, Perez-Garcia A, Carpenter Z, Khiabanian H, Tosello V, Allegretta M, Paietta E, Racevskis J, Rowe JM, Tallman MS, Paganin M, Basso G, Hof J, Kirschner-Schwabe R, Palomero T, Rabadan R, Ferrando A, 2013, Activating mutations in the NT5C2 nucleotidase gene drive chemotherapy resistance in relapsed ALL. Nat Med 19:368–371 Oh JE, An CH, Yoo NJ, Lee SH, 2011, Detection of lowlevel EGFR T790M mutation in lung cancer tissues. APMIS 119:403–411 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 223 EP 224 DI 10.1007/s12253-015-9965-0 PG 2 ER PT J AU Choi, RM An, HCh Yoo, JN Lee, HS AF Choi, Ryoung Mi An, Hyeok Chang Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutations of CAB39L, an Activator of LKB1 Tumor Suppressor, in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Choi, Ryoung Mi] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-, 137-701 Seoul, South Korea. [An, Hyeok Chang] The Catholic University of Korea, College of Medicine, Department of General SurgerySeoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Alessi DR, Sakamoto K, Bayascas JR, 2006, LKB1-dependent signaling pathways. Transcription 4:29–33 Hemminki A, Tomlinson I, Markie D, Jarvinen H, Sistonen P, Bjorkqvist AM, Knuutila S, Salovaara R, Bodmer W, Shibata D, de la Chapelle A, Aaltonen LA, 1997, Localization of a susceptibility locus for Peutz-Jeghers syndrome to 19p using comparative genomic hybridization and targeted linkage analysis. Nat Genet 15: 87–90 Sanchez-Cespedes M, 2007, A role for LKB1 gene in human cancer beyond the Peutz-Jeghers syndrome. Oncogene 26:7825–7832 Mehellou Y, Alessi DR, Macartney TJ, SzklarzM, Knapp S, Elkins JM, 2013, Structural insights into the activation of MST3 by MO25. Biochem Biophys Res Commun 431:604–609 Ling P, Lu TJ, Yuan CJ, Lai MD, 2008, Biosignaling of mammalian Ste20-related kinases. Cell Signal 20:1237–1247 Imai K, 2008, Yamamoto H, 2008, carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Yoo NJ, Kim HR, Kim YR, An CH, 2012, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 Vogelstein B, Papadopoulos N,Velculescu VE, Zhou S, Diaz Jr LA, Kinzler KW(2013, Cancer genome landscapes. Science 339:1546– 1558 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 225 EP 226 DI 10.1007/s12253-015-9973-0 PG 2 ER PT J AU Lee, HJ Kim, SM Yoo, JN Lee, HS AF Lee, Hwa Ju Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Absence of KNSTRN Mutation, a Cutaneous Squamous Carcinoma-Specific Mutation, in Other Solid Tumors and Leukemias SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Lee, Hwa Ju] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Fang L, Seki A, Fang G, 2009, SKAP associates with kinetochores and promotes the metaphase-to-anaphase transition. Cell Cycle 8: 2819–2827. Lee CS, Bhaduri A, Mah A, JohnsonWL, Ungewickell A, Aros CJ, Nguyen CB, Rios EJ, Siprashvili Z, Straight A, Kim J, Aasi SZ, Khavari PA, 2014, Recurrent point mutations in the kinetochore gene KNSTRN in cutaneous squamous cell carcinoma. Nat Genet 46:1060–1062. Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044-E1047. Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers Histopathology 60:943–952. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 227 EP 228 DI 10.1007/s12253-015-9993-9 PG 2 ER PT J AU Goodyer, M Langabeer, ES Haslam, K Murphy, K AF Goodyer, Matthew Langabeer, E Stephen Haslam, Karl Murphy, Karen TI The JAK2 V617F Allele Burden in Latent Myeloproliferative Neoplasms Presenting with Splanchnic Vein Thrombosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Goodyer, Matthew] St. Vincent’s University Hospital, Department of HaematologyDublin, Ireland. [Langabeer, E Stephen] St. James’s Hospital, Central Pathology Laboratory, Cancer Molecular DiagnosticsDublin, Ireland. [Haslam, Karl] St. James’s Hospital, Central Pathology Laboratory, Cancer Molecular DiagnosticsDublin, Ireland. [Murphy, Karen] St. Vincent’s University Hospital, Department of HaematologyDublin, Ireland. RP Langabeer, ES (reprint author), St. James’s Hospital, Central Pathology Laboratory, Cancer Molecular Diagnostics, Dublin, Ireland. EM slangabeer@stjames.ie CR Sekhar M, McVinnie K, Burroughs AK, 2013, Splanchnic vein thrombosis in myeloproliferative neoplasms. Br J Haematol 162: 730–747 Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Vardiman JW, eds,, 2008)WHO classification of tumours of haematopoietic and lymphoid tissue, 4th ed. International Agency for Research on Cancer, Lyon Langabeer SE, Andrikovics H, Asp J, et al, 2015, Molecular diagnostics of myeloproliferative neoplasms. Eur J Haematol 95:270– 279 Dentali F, Squizzato A, Brivio L, et al, 2009, JAK2 V617F mutation for the early diagnosis of Ph- myeloproliferative neoplasms in patients with venous thromboembolism: a meta-analysis. Blood 113:5617–5623 Pardanani A, Lasho T, Schwager S, et al, 2007, JAK2 V617F prevalence and allele burden in non-splanchnic venous thrombosis in the absence of overt myeloproliferative disorder. Leukemia 21: 1828–1829 Toyama K, Karasawa M, Yamane A, et al, 2009, Low allele burden of a JAK2-V617F mutated clone in monoclonal haematopoiesis in a Japanese woman with Budd-Chiari syndrome. Int J Hematol 89: 517–522 Colaizzo D, Amitrano L, Guardascione MA, et al, 2013, Outcome for patients with splanchnic venous thrombosis presenting without overt MPN: a role for the JAK2 V617F mutation re-evaluation. Thromb Res 132:e99–e104 Haslam K, Molloy KM, Conneally E, Langabeer S, 2014, Evaluation of a JAK2 V617F quantitative PCR to monitor residual disease post-allogeneic hematopoietic stem cell transplantation for myeloproliferative neoplasms. Clin Chem Lab Med 52:e29–e31 Nielsen C, Bojesen SE, Nordestgaard BG, Kofoed KF, Birgens HS, 2014, JAK2 V617F somatic mutation in the general population: myeloproliferative neoplasms development and progression rate. Haematologica 99:1448–1455 Lippert E, Mansier O, Migeon M, et al, 2014, Clinical and biological characterization of patients with low, 0.1–2 %, JAK2 V617F allele burden at diagnosis. Haematologica 99:e98–e101 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 229 EP 230 DI 10.1007/s12253-015-9994-8 PG 2 ER PT J AU Jo, SY Kim, SM Yoo, JN Lee, HS AF Jo, Sol Yun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Absence of PRKD1 Mutation, a Salivary Tumor-Specific Mutation, in Solid Tumors and Leukemias SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Weinreb I, Piscuoglio S, Martelotto LG, Waggott D, Ng CK, Perez- Ordonez B, Harding NJ, Alfaro J, Chu KC, Viale A, Fusco N, da Cruz PA, Marchio C, Sakr RA, Lim R, Thompson LD, Chiosea SI, Seethala RR, Skalova A, Stelow EB, Fonseca I, Assaad A, How C, Wang J, de Borja R, Chan-Seng-Yue M, Howlett CJ, Nichols AC, Wen YH, Katabi N, Buchner N, Mullen L, Kislinger T,Wouters BG, Liu FF, Norton L, McPherson JD, Rubin BP, Clarke BA,Weigelt B, Boutros PC, Reis-Filho JS, 2014, Hotspot activating PRKD1 somatic mutations in polymorphous low-grade adenocarcinomas of the salivary glands. Nat Genet 46:1166–1169 Rozenrurt E, 2011, Protein kinase D signaling: multiple biological functions in health and disease. Physiology, Bethesda, 26:23–33 Debnath J, Brugge JS, 2005, Modelling glandular epithelial cancers in three-dimensional cultures. Nat Rev Cancer 5:675– 688 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2016 VL 22 IS 1 BP 231 EP 232 DI 10.1007/s12253-015-0001-1 PG 1 ER PT J AU Jarosova, M Kriegova, E Schneiderova, P Fillerova, R Prochazka, V Mikesova, M Flodr, P Indrak, K Papajik, T AF Jarosova, Marie Kriegova, Eva Schneiderova, Petra Fillerova, Regina Prochazka, Vit Mikesova, Michaela Flodr, Patrik Indrak, Karel Papajik, Tomas TI A Novel Non-Immunoglobulin (non-Ig)/BCL6 Translocation in Diffuse Large B-Cell Lymphoma Involving Chromosome 10q11.21 Loci and Review on Clinical Consequences of BCL6 Rearrangements SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Diffuse large B-cell lymphoma; BCL6 rearrangements; BCL2 rearrangements; Non-immunoglobulin gene translocations; Complex chromosomal changes ID Diffuse large B-cell lymphoma; BCL6 rearrangements; BCL2 rearrangements; Non-immunoglobulin gene translocations; Complex chromosomal changes AB BCL6 rearrangements (3q27) are the most common chromosomal abnormalities in diffuse large B-cell lymphoma (DLBCL), with numerous immunoglobulin (Ig) and non-Ig genes as partners. In DLBCL, the translocations occur predominantly in the "major breakpoint region" encompassing the first noncoding exon and a part of the first intron of BCL6; few cases with "alternative breakpoint cluster" located 245– 285 kb 5′ BCL6 were also described. The regulatory sequences of known Ig and non-Ig partners replace the 5′ untranslated region of the BCL6 in the same transcriptional orientation. Contrary to Ig/BCL6 fusions typical by high BCL6 gene expression, in non-Ig/BCL6 translocations were observed unexpectedly low BCL6 mRNA levels. From the clinical point of view, the survival rate of DLBCL patients with non-Ig partners is inferior to those with Ig/BCL6 translocations, suggesting that non-Ig/BCL6 fusion is a poor prognostic indicator. Hereby we provide comprehensive information about known non-Ig translocation partners and clinical consequences of BCL6 rearrangements in DLBCL. Moreover, we describe a novel reciprocal translocation t(3;10) in refractory patient with DLBCL with the breaking points at 5′ untranslated region of BCL6 and 5′ untranslated region of the RASGEF1A gene on chromosome 10q11.21 loci; this rearrangement was associated with low BCL6 and RASGEF1A gene expressions. Our patient harbouring dual chromosomal rearrangement involving BCL2 and BCL6 genes relapsed three-times and died soon; thus, further supporting the notion that non-Ig/BCL6 fusion is a poor prognostic indicator of DLBCL. There is evidence of prognostic value of BCL6 rearrangements also in rituximab era. C1 [Jarosova, Marie] Palacky University and University Hospital, Medical School, Department of Hemato-oncology, Hnevotinska 3, 77900 Olomouc, Czech Republic. [Kriegova, Eva] Palacky University Olomouc and the University Hospital Olomouc, Faculty of Medicine and Dentistry, Department of Immunology, Hnevotinska 3, 77900 Olomouc, Czech Republic. [Schneiderova, Petra] Palacky University Olomouc and the University Hospital Olomouc, Faculty of Medicine and Dentistry, Department of Immunology, Hnevotinska 3, 77900 Olomouc, Czech Republic. [Fillerova, Regina] Palacky University Olomouc and the University Hospital Olomouc, Faculty of Medicine and Dentistry, Department of Immunology, Hnevotinska 3, 77900 Olomouc, Czech Republic. [Prochazka, Vit] Palacky University and University Hospital, Medical School, Department of Hemato-oncology, Hnevotinska 3, 77900 Olomouc, Czech Republic. [Mikesova, Michaela] Palacky University and University Hospital, Medical School, Department of Hemato-oncology, Hnevotinska 3, 77900 Olomouc, Czech Republic. [Flodr, Patrik] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular Pathology, Hnevotinska 3, 77900 Olomouc, Czech Republic. [Indrak, Karel] Palacky University and University Hospital, Medical School, Department of Hemato-oncology, Hnevotinska 3, 77900 Olomouc, Czech Republic. [Papajik, Tomas] Palacky University and University Hospital, Medical School, Department of Hemato-oncology, Hnevotinska 3, 77900 Olomouc, Czech Republic. RP Jarosova, M (reprint author), Palacky University and University Hospital, Medical School, Department of Hemato-oncology, 77900 Olomouc, Czech Republic. EM marie.jarosova@fnol.cz CR Zhang J, Grubor V, Love CL, Banerjee A, Richards KL, Mieczkowski PA, Dunphy C, Choi W, Au WY, Srivastava G, Lugar PL, Rizzieri DA, Lagoo AS, Bernal-Mizrachi L, Mann KP, Flowers C, Naresh K, Evens A, Gordon LI, Czader M, Gill JI, Hsi ED, Liu Q, Fan A, Walsh K, Jima D, Smith LL, Johnson AJ, Byrd JC, LuftigMA, Ni T, Zhu J, Chadburn A, Levy S, Dunson D, Dave SS, 2013, Genetic heterogeneity of diffuse large B-cell lymphoma. Proc Natl Acad Sci U S A 110(4):1398–1403 Tirado CA, Chen W, Garcia R, Kohlman KA, Rao N, 2012, Genomic profiling using array comparative genomic hybridization define distinct subtypes of diffuse large B-cell lymphoma: a review of the literature. J Hematol Oncol 5:54 Ohno H, 2006, Pathogenetic and clinical implications of nonimmunoglobulin; BCL6 translocations in B-cell non-Hodgkin’s lymphoma. J Clin Exp Hematop 46(2):43–53 Shustik J, Han G, Farinha P, Johnson NA, Ben Neriah S, Connors JM, Sehn LH, Horsman DE, Gascoyne RD, Steidl C, 2010, Correlations between BCL6 rearrangement and outcome in patients with diffuse large B-cell lymphoma treated with CHOP or RCHOP. Haematologica 95(1):96–101 Flodr P, Latalova P, Tichy T, Kubova Z, Papajik T, Svachova M, Vrzalikova K, Radova L, Jarosova M, Murray PG, 2014, Diffuse large B-cell lymphoma—the history, current view and new perspectives. Neoplasma 61(5):491–504 Nedomova R, Papajik T, ProchazkaV, Indrak K, JarosovaM(2013, Cytogenetics and molecular cytogenetics in diffuse large B-cell lymphoma, DLBCL). Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 157(3):239–247 Lossos IS, Czerwinski DK, Alizadeh AA,Wachser MA, Tibshirani R, Botstein D, Levy R, 2004, Prediction of survival in diffuse large- B-cell lymphoma based on the expression of six genes. N Engl J Med 350(18):1828–1837 Chang CC, Ye BH, Chaganti RS, Dalla-Favera R, 1996, BCL-6, a POZ/zinc-finger protein, is a sequence-specific transcriptional repressor. Proc Natl Acad Sci U S A 93(14):6947–6952 Barros P, Lam EW, Jordan P,Matos P, 2012, Rac1 signalling modulates a STAT5/BCL-6 transcriptional switch on cell-cycleassociated target gene promoters. Nucleic Acids Res 40(16): 7776–7787 Siggs OM, Beutler B, 2012, The BTB-ZF transcription factors. Cell Cycle 11(18):3358–3369 Ueda Y, Nishida K, Miki T, Horiike S, Kaneko H, Yokota S, Misawa S, Abe T, Kashima K, Taniwaki M, 1997, Interphase detection of BCL6/IgH fusion gene in non-Hodgkin lymphoma by fluorescence in situ hybridization. Cancer Genet Cytogenet 99(2): 102–107 Yoshida S, KaneitaY, AokiY, SetoM,Mori S,MoriyamaM(1999, Identification of heterologous translocation partner genes fused to the BCL6 gene in diffuse large B-cell lymphomas: 5′-RACE and LA - PCR analyses of biopsy samples. Oncogene 18(56):7994– 7999 Akasaka H, Akasaka T, Kurata M, Ueda C, Shimizu A, Uchiyama T, Ohno H, 2000, Molecular anatomy of BCL6 translocations revealed by long-distance polymerase chain reaction-based assays. Cancer Res 60(9):2335–2341 Jardin F, Ruminy P, Bastard C, Tilly H, 2007, The BCL6 protooncogene: a leading role during germinal center development and lymphomagenesis. Pathol Biol 55(1):73–83 Iqbal J, Greiner TC, Patel K, Dave BJ, Smith L, Ji J, Wright G, Sanger WG, Pickering DL, Jain S, Horsman DE, Shen Y, Fu K, Weisenburger DD, Hans CP, Campo E, Gascoyne RD, Rosenwald A, Jaffe ES, Delabie J, Rimsza L, Ott G, Muller-Hermelink HK, Connors JM, Vose JM, McKeithan T, Staudt LM, ChanWC, 2007, Distinctive patterns of BCL6 molecular alterations and their functional consequences in different subgroups of diffuse large B-cell lymphoma. Leukemia 21(11):2332–2343 Barrans SL, O’Connor SJ, Evans PA, Davies FE, Owen RG, Haynes AP, Morgan GJ, Jack AS, 2002, Rearrangement of the BCL6 locus at 3q27 is an independent poor prognostic factor in nodal diffuse large B-cell lymphoma. Br J Haematol 117(2):322– 332 Akasaka T, Ueda C, Kurata M, Akasaka H, Yamabe H, Uchiyama T, OhnoH, 2000, Nonimmunoglobulin, non-Ig)/BCL6 gene fusion in diffuse large B-cell lymphoma results in worse prognosis than Ig/ BCL6. Blood 96(8):2907–2909 Keller CE, Nandula S, Vakiani E, Alobeid B, Murty VV, Bhagat G, 2006, Intrachromosomal rearrangement of chromosome 3q27: an under recognized mechanism of BCL6 translocation in B-cell non- Hodgkin lymphoma. Hum Pathol 37(8):1093–1099 Prochazka V, Papajik T, Faber E, Raida L, Kapitanova Z, Langova K, Prouzova Z, Jarosova M, Indrak K, 2014, Soluble interleukin-2 receptor level predicts survival in patients with follicular lymphoma treated with CHOP chemotherapy in the rituximab era. Leuk Lymphoma 55(7):1584–1590 Niitsu N, Nakamine H, Hagiwara Y, Tanae K, Miura I, 2008, The t(1;3)(p34;q27, translocation: a nonrandomBCL6 rearrangement in diffuse large B cell lymphoma. Ann Hematol 87(2):151–153 Nakamura Y, Takahashi N, Kakegawa E, Yoshida K, Ito Y, Kayano H, Niitsu N, Jinnai I, Bessho M, 2008, The GAS5, growth arrestspecific transcript 5, gene fuses to BCL6 as a result of t(1;3)(q25;q27, in a patient with B-cell lymphoma. Cancer Genet Cytogenet 182(2):144–149 Bloomfield CD, Arthur DC, Frizzera G, Levine EG, Peterson BA, Gajl-Peczalska KJ, 1983, Nonrandom chromosome abnormalities in lymphoma. Cancer Res 43(6):2975–2984 Chenevix-Trench G, Brown JA, Tyler GB, Behm FG, 1988, Chromosome analysis of 30 cases of non-Hodgkin’s lymphoma. Med Oncol Tumor Pharmacother 5(1):17–32 Martin-Subero JI, Odero MD, Hernandez R, Cigudosa JC, Agirre X, Saez B, Sanz-Garcia E, Ardanaz MT, Novo FJ, Gascoyne RD, Calasanz MJ, Siebert R, 2005, Amplification of IGH/MYC fusion in clinically aggressive IGH/BCL2-positive germinal center B-cell lymphomas. Genes Chromosomes Cancer 43(4):414–423 Jerkeman M, Johansson B, Akerman M, Cavallin-Stahl E, Kristoffersson U, Mitelman F, 1999, Prognostic implications of cytogenetic aberrations in diffuse large B-cell lymphomas. Eur J Haematol 62(3):184–190 Utsumi S, Miura I, Takahashi N, Ohshima A, Nimura T, Hashimoto K, Chubachi A, Saito M, Takatsu H, Yamaguchi A et al, 1995, Bone and brain involvement in a case of diffuse large B cell lymphoma associated with t(2;3)(p12;q27). Int J Hematol 62(4):247– 252 Dave BJ, Weisenburger DD, Higgins CM, Pickering DL, Hess MM, ChanWC, Sanger WG(2004, Cytogenetics and fluorescence in situ hybridization studies of diffuse large B-cell lymphoma in children and young adults. Cancer Genet Cytogenet 153(2):115– 121 Sanchez-Izquierdo D, Siebert R, Harder L, Marugan I, Gozzetti A, Price HP, Gesk S, Hernandez-Rivas JM, Benet I, Sole F, Sonoki T, Le Beau MM, Schlegelberger B, Dyer MJ, Garcia-Conde J, Martinez-Climent JA, 2001, Detection of translocations affecting the BCL6 locus in B cell non-Hodgkin’s lymphoma by interphase fluorescence in situ hybridization. Leukemia 15(9):1475–1484 Van Roosbroeck K, Cools J, Dierickx D, Thomas J, Vandenberghe P, Stul M, Delabie J, De Wolf-Peeters C, Marynen P, Wlodarska I, 2010, ALK-positive large B-cell lymphomas with cryptic SEC31A-ALK and NPM1-ALK fusions. Haematologica 95(3): 509–513 Yoshioka T, Miura I, Kume M, Takahashi N, Okamoto M, Ichinohasama R, Yoshino T, Yamaguchi M, Hirokawa M, Sawada K, Nakamura S, 2005, Cytogenetic features of de novo CD5-positive diffuse large B-cell lymphoma: chromosome aberrations affecting 8p21 and 11q13 constitute major subgroups with different overall survival. Genes Chromosomes Cancer 42(2): 149–157 Akasaka T, Lossos IS, Levy R, 2003, BCL6 gene translocation in follicular lymphoma: a harbinger of eventual transformation to diffuse aggressive lymphoma. Blood 102(4):1443–1448 Hashimoto K, Miura I, Chyubachi A, Saito M, Miura AB, 1995, Correlations of chromosome abnormalities with histologic and immunologic characteristics in 49 patients from Akita, Japan with non-Hodgkin lymphoma. Cancer Genet Cytogenet 81(1):56–65 Wlodarska I, Stul M, De Wolf-Peeters C, Hagemeijer A, 2004, Heterogeneity of BCL6 rearrangements in nodular lymphocyte predominant Hodgkin’s lymphoma. Haematologica 89(8):965–972 Pasqualucci L, Neumeister P, Goossens T, Nanjangud G, Chaganti RS, Kuppers R, Dalla-Favera R, 2001, Hypermutation of multiple proto-oncogenes in B-cell diffuse large-cell lymphomas. Nature 412(6844):341–346 Dallery E, Galiegue-Zouitina S, Collyn-d’Hooghe M, Quief S, Denis C, Hildebrand MP, Lantoine D, Deweindt C, Tilly H, Bastard C et al, 1995, TTF, a gene encoding a novel small G protein, fuses to the lymphoma-associated LAZ3 gene by t(3;4, chromosomal translocation. Oncogene 10(11):2171–2178 Chaganti SR, Rao PH, ChenW, Dyomin V, Jhanwar SC, Parsa NZ, Dalla-Favera R, Chaganti RS, 1998, Deregulation of BCL6 in non- Hodgkin lymphoma by insertion of IGH sequences in complex translocations involving band 3q27. Genes Chromosomes Cancer 23(4):328–336 Yeh YM, Chang KC, Chen YP, Kao LY, Tsai HP, Ho CL,Wang JR, Jones D, Chen TY, 2010, Large B cell lymphoma presenting initially in bone marrow, liver and spleen: an aggressive entity associated frequently with haemophagocytic syndrome. Histopathology 57(6):785–795 Tanaka R, Satoh H,MoriyamaM, Satoh K, Morishita Y, Yoshida S, Watanabe T, Nakamura Y, Mori S, 2000, Intronic U50 smallnucleolar- RNA, snoRNA, host gene of no protein-coding potential is mapped at the chromosome breakpoint t(3;6)(q27;q15, of human B-cell lymphoma. Genes Cells 5(4):277–287 ChenW, Itoyama T, Chaganti RS, 2001, Splicing factor SRP20 is a novel partner of BCL6 in a t(3;6)(q27;p21, translocation in transformed follicular lymphoma. Genes Chromosomes Cancer 32(3): 281–284 KurataM, Maesako Y, Ueda C, NishikoriM, Akasaka T, Uchiyama T, Ohno H, 2002, Characterization of t(3;6)(q27;p21, breakpoints in B-cell non-Hodgkin’s lymphoma and construction of the histone H4/BCL6 fusion gene, leading to altered expression of Bcl-6. Cancer Res 62(21):6224–6230 Akasaka T,Miura I, Takahashi N, Akasaka H,Yonetani N, Ohno H, Fukuhara S, Okuma M, 1997, A recurring translocation, t(3;6)(q27;p21), in non-Hodgkin’s lymphoma results in replacement of the 5′ regulatory region of BCL6 with a novel H4 histone gene. Cancer Res 57(1):7–12 Katsura Y, Ohta I, Yoshida C, Ohtani H, Komeno T, 2011, Diffuse large B-cell lymphoma carrying both t(3;7)(q27;p12, and t(8;14)(q24;q32). Intern Med 50(8):905–908 Ichinohasama R, Miura I, Funato T, Sato I, Suzuki C, Saito Y, Decoteau JF, Myers JB, Kadin ME, Sawai T, Ooya K, 1998, A recurrent nonrandom translocation, 3;7)(q27;p12, associated with BCL-6 gene rearrangement in B-cell diffuse large cell lymphoma. Cancer Genet Cytogenet 104(1):19–27 HosokawaY,MaedaY, Ichinohasama R, Miura I, TaniwakiM, Seto M, 2000, The Ikaros gene, a central regulator of lymphoid differentiation, fuses to the BCL6 gene as a result of t(3;7)(q27;p12, translocation in a patient with diffuse large B-cell lymphoma. Blood 95(8):2719–2721 Kumari P, Mukherjee G, Rao CR, Devi MG, Biswas S, Appaji L, Arunakumari BS, Padma M, Bapsy PP, Sundareshan TS, 2003, Cytogenetic study of non-Hodgkin lymphoma from South India. histologic and geographic correlations. Cancer Genet Cytogenet 141(1):14–19 Schneider B, Nagel S, Kaufmann M, Winkelmann S, Bode J, Drexler HG, MacLeod RA, 2008, T(3;7)(q27;q32, fuses BCL6 to a non-coding region at FRA7H near miR-29. Leukemia 22(6): 1262–1266 Bertrand P, Bastard C, Maingonnat C, Jardin F, Maisonneuve C, Courel MN, Ruminy P, Picquenot JM, Tilly H, 2007, Mapping of MYC breakpoints in 8q24 rearrangements involving nonimmunoglobulin partners in B-cell lymphomas. Leukemia 21(3): 515–523 Johnson NA, Savage KJ, Ludkovski O, Ben-Neriah S, Woods R, Steidl C, Dyer MJ, Siebert R, Kuruvilla J, Klasa R, Connors JM, Gascoyne RD, Horsman DE, 2009, Lymphomas with concurrent BCL2 and MYC translocations: the critical factors associated with survival. Blood 114(8):2273–2279 Wang HY, Bossler AD, Schaffer A, Tomczak E, DiPatri D, Frank DM, Nowell PC, Bagg A, 2007, A novel t(3;8)(q27;q24.1, simultaneously involving both the BCL6 and MYC genes in a diffuse large B-cell lymphoma. Cancer Genet Cytogenet 172(1):45–53 Bacher U, Haferlach T, Alpermann T, Kern W, Schnittger S, Haferlach C, 2011, Several lymphoma-specific genetic events in parallel can be found in mature B-cell neoplasms. Genes Chromosomes Cancer 50(1):43–50 Nordgren A, Sorensen AG, Tinggaard-Pedersen N, Blennow E, Larsson C, Lagercrantz S, 2000, New chromosomal breakpoints in non-Hodgkin’s lymphomas revealed by spectral karyotyping and G-banding. Int J Mol Med 5(5):485–492 Iqbal J, Gupta S, Chen QH, Brody JP, Koduru P, 2007, Diffuse large B-cell lymphoma with a novel translocation involving BCL6. Cancer Genet Cytogenet 178(1):73–76 Galieque Zouitina S, Quief S, Hildebrand MP, Denis C, Lecocq G, Collyn-d’Hooghe M, Bastard C, Yuille M, Dyer MJ, Kerckaert JP, 1996, The B cell transcriptional coactivator BOB1/OBF1 gene fuses to the LAZ3/BCL6 gene by t(3;11)(q27;q23.1, chromosomal translocation in a B cell leukemia line, Karpas 231). Leukemia 10(4):579–587 Mikraki V, Jhanwar SC, Filippa DA, Wollner N, Chaganti RS, 1992, Distinct patterns of chromosome abnormalities characterize childhood non-Hodgkin’s lymphoma. Br J Haematol 80(1):15–20 Montesinos-Rongen M, Akasaka T, Zuhlke-Jenisch R, Schaller C, Van Roost D,Wiestler OD, Siebert R, DeckertM(2003, Molecular characterization of BCL6 breakpoints in primary diffuse large Bcell lymphomas of the central nervous system identifies GAPD as novel translocation partner. Brain Pathol 13(4):534–538 Lai JL, Daudignon A, Kerckaert JP, Galiegue-Zouitina S, Detourmignies L, Morel P, Bauters F, Fenaux P, 1998, Translocation, 3;13)(q27;q14): a nonrandom and probably secondary structural change in non-Hodgkin lymphomas. Cancer Genet Cytogenet 103(2):140–143 Johnson NA, Al-Tourah A, Brown CJ, Connors JM, Gascoyne RD, Horsman DE, 2008, Prognostic significance of secondary cytogenetic alterations in follicular lymphomas. Genes Chromosomes Cancer 47(12):1038–1048 Au WY, Gascoyne RD, Viswanatha DS, Skinnider BF, Connors JM, Klasa RJ, Horsman DE, 1999, Concurrent chromosomal alterations at 3q27, 8q24 and 18q21 in B-cell lymphomas. Br J Haematol 105(2):437–440 Ueda C, Akasaka T, Kurata M, Maesako Y, Nishikori M, Ichinohasama R, Imada K, Uchiyama T, Ohno H, 2002, The gene for interleukin-21 receptor is the partner of BCL6 in t(3;16)(q27;p11), which is recurrently observed in diffuse large B-cell lymphoma. Oncogene 21(3):368–376 Ichinohasama R, Miura I, Takahashi N, Sugawara T, Tamate E, Endoh K, Endoh F, Naganuma H, DeCoteau JF, Griffin JD, Kadin ME, Ooya K, 2000, Ph-negative non-Hodgkin’s lymphoma occurring in chronic phase of Ph-positive chronic myelogenous leukemia is defined as a genetically different neoplasm from extramedullary localized blast crisis: report of two cases and review of the literature. Leukemia 14(1):169–182 Yunis JJ, OkenMM, Theologides A, Howe RB, KaplanME, 1984, Recurrent chromosomal defects are found in most patients with non-Hodgkin’s-lymphoma. Cancer Genet Cytogenet 13(1):17–28 Kriegova E, Arakelyan A, Fillerova R, Zatloukal J, Mrazek F, Navratilova Z, Kolek V, du Bois RM, Petrek M, 2008, PSMB2 and RPL32 are suitable denominators to normalize gene expression profiles in bronchoalveolar cells. BMC Mol Biol 9:69 Falkenberg VR, Whistler T, Murray JR, Unger ER, Rajeevan MS, 2011, Identification of Phosphoglycerate Kinase 1, PGK1, as a reference gene for quantitative gene expression measurements in human blood RNA. BMC Res Notes 4:324 Saito M, Novak U, Piovan E, Basso K, Sumazin P, Schneider C, Crespo M, Shen Q, Bhagat G, Califano A, Chadburn A, Pasqualucci L, Dalla-Favera R, 2009, BCL6 suppression of BCL2 viaMiz1 and its disruption in diffuse large B cell lymphoma. Proc Natl Acad Sci U S A 106(27):11294–11299 Juszczynski P, Chen L, O’Donnell E, Polo JM, Ranuncolo SM, Dalla-Favera R, Melnick A, Shipp MA, 2009, BCL6 modulates tonic BCR signaling in diffuse large B-cell lymphomas by repressing the SYK phosphatase, PTPROt. Blood 114(26):5315–5321 Basso K, Dalla-Favera R, 2012, Roles of BCL6 in normal and transformed germinal center B cells. Immunol Rev Immunol Rev 247(1):172–183 Basso K, Saito M, Sumazin P, Margolin AA, Wang K, Lim WK, Kitagawa Y, Schneider C, Alvarez MJ, Califano A, Dalla-Favera R, 2010, Integrated biochemical and computational approach identifies BCL6 direct target genes controlling multiple pathways in normal germinal center B cells. Blood 115(5):975–984 CiW, Polo JM, Cerchietti L, Shaknovich R,Wang L, Yang SN, Ye K, Farinha P, Horsman DE, Gascoyne RD, Elemento O,Melnick A, 2009, The BCL6 transcriptional program features repression of multiple oncogenes in primary B cells and is deregulated in DLBCL. Blood 113(22):5536–5548 Ranuncolo SM, Polo JM, Dierov J, Singer M, Kuo T, Greally J, Green R, Carroll M, Melnick A, 2007, Bcl-6 mediates the germinal center B cell phenotype and lymphomagenesis through transcriptional repression of the DNA-damage sensor ATR. Nat Immunol 8(7):705–714 Phan RT, Dalla-Favera R, 2004, The BCL6 proto-oncogene suppresses p53 expression in germinal-centre B cells. Nature 432(7017):635–639 Tunyaplin C, Shaffer AL, Angelin-Duclos CD, Yu X, Staudt LM, Calame KL, 2004, Direct repression of prdm1 by Bcl-6 inhibits plasmacytic differentiation. J Immunol 173(2):1158–1165 KikuchiM, Miki T, Kumagai T, Fukuda T, Kamiyama R,Miyasaka N, Hirosawa S, 2000, Identification of negative regulatory regions within the first exon and intron of the BCL6 gene. Oncogene 19(42):4941–4945 Bea S, Colomo L, Lopez-Guillermo A, Salaverria I, Puig X, Pinyol M, Rives S, Montserrat E, Campo E, 2004, Clinicopathologic significance and prognostic value of chromosomal imbalances in diffuse large B-cell lymphomas. J Clin Oncol 22(17):3498–3506 Jardin F, Gaulard P, Buchonnet G, Contentin N, Lepretre S, Lenain P, Stamatoullas A, Picquenot JM, Duval C, Parmentier F, Tilly H, Bastard C, 2002, Follicular lymphoma without t(14;18, and with BCL-6 rearrangement: a lymphoma subtype with distinct pathological, molecular and clinical characteristics. Leukemia 16(11):2309– 2317 Miura I,Ohshima A, ChubachiA, Nimura T,Komatsuda A, Utsumi S, Saito M, Machii T, Nakamura S, Seto M, Miura AB, 1997, BCL6 rearrangement in a patient with mantle cell lymphoma. Ann Hematol 74(5):247–250 Yaman E, Gasper R, Koerner C, Wittinghofer A, Tazebay UH, 2009, RasGEF1A and RasGEF1B are guanine nucleotide exchange factors that discriminate between Rap GTP-binding proteins and mediate Rap2-specific nucleotide exchange. FEBS J 276(16):4607–4616 Ratajczak MZ, Kucia M, Liu R, Shin DM, Bryndza E, Masternak MM, Tarnowski M, Ratajczak J, Bartke A, 2011, RasGrf1: genomic imprinting, VSELs, and aging. Aging, Albany NY, 3(7):692– 697 Borras C, Monleon D, Lopez-Grueso R, Gambini J, Orlando L, Pallardo FV, Santos E, Vina J, Font de Mora J, 2011, RasGrf1 deficiency delays aging in mice. Aging, Albany NY, 3(3):262–276 Ura K, Obama K, Satoh S, Sakai Y, Nakamura Y, Furukawa Y, 2006, Enhanced RASGEF1A expression is involved in the growth and migration of intrahepatic cholangiocarcinoma. Clin Cancer Res 12(22):6611–6616 Font de Mora J, Esteban LM, Burks DJ, Nunez A, Garces C, Garcia‐Barrado MJ, Iglesias‐Osma MC, Moratinos J, Ward JM, Santos E, 2003, Ras‐GRF1 signaling is required for normal betacell development and glucose homeostasis. Embo J 22(12):3039– 3049 Pannekoek WJ, Linnemann JR, Brouwer PM, Bos JL, Rehmann H, 2013, Rap1 and Rap2 antagonistically control endothelial barrier resistance. PLoS One 8(2):e57903 Tarnowski M, Schneider G, Amann G, Clark G, Houghton P, Barr FG, Kenner L, Ratajczak MZ, KuciaM, 2012, RasGRF1 regulates proliferation and metastatic behavior of human alveolar rhabdomyosarcomas. Int J Oncol 41(3):995–1004 Lu Z, Tsai AG, Akasaka T, Ohno H, Jiang Y, Melnick AM, Greisman HA, Lieber MR, 2013, BCL6 breaks occur at different AID sequence motifs in Ig-BCL6 and non-Ig-BCL6 rearrangements. Blood 121(22):4551–4554 Lohr JG, Stojanov P, Lawrence MS, Auclair D, Chapuy B, Sougnez C, Cruz-Gordillo P, Knoechel B, Asmann YW, Slager SL, Novak AJ, Dogan A, Ansell SM, Link BK, Zou L, Gould J, Saksena G, Stransky N, Rangel-Escareno C, Fernandez-Lopez JC, Hidalgo- Miranda A, Melendez-Zajgla J, Hernandez-Lemus E, Schwarz- Cruz y Celis A, Imaz-Rosshandler I, Ojesina AI, Jung J, Pedamallu CS, Lander ES, Habermann TM, Cerhan JR, Shipp MA, Getz G, Golub TR, 2012, Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma, DLBCL, by whole-exome sequencing. Proc Natl Acad Sci U S A 109(10): 3879–3884 Lossos IS, Jones CD, Warnke R, Natkunam Y, Kaizer H, Zehnder JL, Tibshirani R, Levy R, 2001, Expression of a single gene, BCL- 6, strongly predicts survival in patients with diffuse large B-cell lymphoma. Blood 98(4):945–951 Ueda C, Akasaka T, Ohno H, 2002, Non-immunoglobulin/BCL6 gene fusion in diffuse large B-cell lymphoma: prognostic implications. Leuk Lymphoma 43(7):1375–1381 Ueda C, Uchiyama T, Ohno H, 2002, Immunoglobulin, Ig)/BCL6 versus non-Ig/BCL6 gene fusion in diffuse large B-cell lymphoma corresponds to a high- versus low-level expression of BCL6 mRNA. Blood 99(7):2624–2625 Cheema FN, Agloria M, Koshy N, Hildebrandt GC, 2014, Double hit lymphoma—a case of unusual response after sequential aggressive chemotherapy and review of the literature. Clin Med Insights Case Rep 7:117–121 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 233 EP 243 DI 10.1007/s12253-015-9972-1 PG 11 ER PT J AU Shao, QQ Zhang, TP Zhao, WJ Liu, ZW You, L Zhou, L Guo, JCh Zhao, YP AF Shao, Qian-Qian Zhang, Tai-Ping Zhao, Wen-Jing Liu, Zi-Wen You, Lei Zhou, Li Guo, Jun-Chao Zhao, Yu-Pei TI Filamin A: Insights into its Exact Role in Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Filamin A; Cancer metastasis; Localization; Nucleus; Cytoplasm ID Filamin A; Cancer metastasis; Localization; Nucleus; Cytoplasm AB Filamin A (FlnA) is a well-known actin crosslinking protein. It serves as a scaffold for over 90 binding partners and involves in multiple cell functions, of which cell migration and adhesion is especially critical. Recently, its role in the cell has come under scrutiny for FlnA’s involvement in cancer development. Originally revealed as a cancerpromoting protein, FlnA actually plays a dual role in cancers. When localized to the cytoplasm, FlnA has a tumorpromoting effect by interacting with signaling molecules. While once localized to the nucleus, it may act to suppress tumor growth and inhibit metastasis by interacting with transcription factors. Thus drugs that can cause FlnA to transpose from cytoplasm to nucleus could be a promising treatment for cancers. Study to this end is on the way in prostate cancer and the results are encouraging. FlnA has been studied in large categories of cancers, such as prostate cancer, breast cancer, melanoma, lung cancer, etc. However, most studies did not evaluate the differences that arise from the localization of the protein, which was a great pity! What’s more, although FlnA’s is undoubtedly important in cancer invasion and metastasis, both preclinical and clinical researches are very rare in some highly metastatic cancers, such as pancreatic cancer. In this mini-review, we give a comprehensive summary of FlnA’ s expression in cancers. Where available, we also indicate the correlation of FlnA with cancer stages and patient prognosis, and clarify its localization (nucleus/cytoplasm) and its dual role (promote/suppress) in different cancers. C1 [Shao, Qian-Qian] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuaifuyuan Wangfujing, 100730 Beijing, China. [Zhang, Tai-Ping] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuaifuyuan Wangfujing, 100730 Beijing, China. [Zhao, Wen-Jing] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuaifuyuan Wangfujing, 100730 Beijing, China. [Liu, Zi-Wen] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuaifuyuan Wangfujing, 100730 Beijing, China. [You, Lei] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuaifuyuan Wangfujing, 100730 Beijing, China. [Zhou, Li] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuaifuyuan Wangfujing, 100730 Beijing, China. [Guo, Jun-Chao] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuaifuyuan Wangfujing, 100730 Beijing, China. [Zhao, Yu-Pei] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuaifuyuan Wangfujing, 100730 Beijing, China. RP Guo, JCh (reprint author), Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. EM gjcpumch@163.com CR Hartwig JH, Stossel TP, 1975, Isolation and properties of actin, myosin and a new actinbinding protein in rabbit alveolar macrophages. J Biol Chem 250:5696–5705 Nakamura F, Stossel TP, 2011, The filamins: organizers of cell structure and function. Cell Adhes Migr 5:160–169 Robertson SP, 2005, Filamin A: phenotypic diversity. Curr Opin Genet Dev 15:301–307. Stossel TP, Condeelis J, Cooley L, Hartwig JH, Noegel A, SchleicherM, et al., 2001, Filamins as integrators of cellmechanics and signalling. Nat Rev Mol Cell Biol 2:138–145 Savoy RM, Ghosh PM, 2013, The dual role of filamin A in cancer: can’t live with, too much of, it, can’t live without it. Endocr Relat Cancer 20:R341–R356. Fucini P, Koppel B, Schleicher M, Lustig A, Holak TA, Muller R, et al., 1999, Molecular architecture of the rod domain of the Dictyosteliumgelation factor, ABP120). J Mol Biol 291:1017– 1023 Gardel ML, Nakamura F, Hartwig JH, Crocker JC, Stossel TP, Weitz DA, 2006, Prestressed F-actin networks cross-linked by hinged filamins replicate mechanical properties of cells. Proc Natl Acad Sci U S A 103:1762–1767 Kasza KE, Koenderink GH, Lin YC, Broedersz CP, Messner W, Nakamura F, Stossel TP, et al, 2009, Nonlinear elasticity of stiff biopolymers connected by flexible linkers. Phys Rev E Stat Nonlinear Soft Matter Phys 79:41928 Kasza KE, Nakamura F, Hu S, Kollmannsberger P, Bonakdar N, Fabry B, Stossel TP, et al, 2009, Filamin A is essential for active cell stiffening but not passive stiffening under external force. Biophys J 96:4326–4335 Tseng Y, An KM, Esue O, Wirtz D, 2004, The bimodal role of filamin in controlling the architecture of F-actin networks. J Biol Chem 279:1819–1826 Cunningham CC, Gorlin JB, Kwiatkowski DJ, Hartwig JH, Janmey PA, Byers HR, et al., 1992, Actin-binding protein requirement for cortical stability and efficient locomotion. Science 255:325–327 Simpson KJ, Selfors LM, Bui J, Reynolds A, Leake D, Khvorova A, et al., 2008, Identification of genes that regulate epithelial cell migration using an siRNA screening approach. Nat Cell Biol 10: 1027–1038 Flevaris P, Stojanovic A, GongH, Chishti A,Welch E, Du X, 2007, A molecular switch that controls cell spreading and retraction. J Cell Biol 179:553–565 Fox JW, Lamperti ED, Eksioglu YZ, Hong SE, Feng Y, Graham DA, et al., 1998, Mutations in filamin 1 prevent migration of cerebral cortical neurons in human periventricular heterotopia. Neuron 21:1315–1325 Sarkisian MR, Bartley CM, Chi H, Nakamura F, Hashimoto-Torii K, Torii M, et al., 2006, MEKK4 signaling regulates filamin expression and neuronal migration. Neuron 52:789–801 Jurak Begonja A, Hoffmeister KM, Hartwig JH, Falet H, 2011, FlnA-null megakaryocytes prematurely release large and fragile platelets that circulate poorly. Blood 118:2285–2295 Hart AW,Morgan JE, Schneider J,West K, McKie L, Bhattacharya S, et al., 2006, Cardiac malformations and midline skeletal defects in mice lacking filamin A. Hum Mol Genet 15:2457–2467 deWitMC, Kros JM, Halley DJ, de Coo IF, Verdijk R, Jacobs BC, et al., 2009, Filamin A mutation, a common cause for periventricular heterotopia, aneurysms and cardiac defects. J Neurol Neurosurg Psychiatry 80:426–428 Masruha MR, Caboclo LO, Carrete Jr H, Cendes IL, Rodrigues MG, Garzon E, et al., 2006, Mutation in filamin A causes periventricular heterotopia, developmental regression, and west syndrome in males. Epilepsia 47:211–214 Sheen VL, Dixon PH, Fox JW, Hong SE, Kinton L, Sisodiya SM, et al., 2001, Mutations in the X-linked filamin 1 gene cause periventricular nodular heterotopia in males as well as in females. Hum Mol Genet 10:1775–1783 Parrini E, Ramazzotti A, Dobyns WB, Mei D, Moro F, Veggiotti P, et al., 2006, Periventricular heterotopia: phenotypic heterogeneity and correlation with Filamin A mutations. Brain 129:1892–1906 Kyndt F, Gueffet JP, Probst V, Jaafar P, Legendre A, Le Bouffant F, et al., 2007)Mutations in the gene encoding filaminAas a cause for familial cardiac valvular dystrophy. Circulation 115:40–49 Zhou AX, Hartwig JH, Akyurek LM, 2010, Filamins in cell signaling, transcription and organ development. Trends Cell Biol 20: 113–123 KimH, Sengupta A, GlogauerM,McCulloch CA(2008, FilaminA regulates cell spreading and survival viab1 integrins. Exp Cell Res 314:834–846 Kim H, McCulloch CA, 2011, Filamin A mediates interactions between cytoskeletal proteins that control cell adhesion. FEBS Lett 585:18–22 Maceyka M, Alvarez SE, Milstien S, Spiegel S, 2008, Filamin A links sphingosine kinase 1 and sphingosine-1-phosphate receptor 1 at lamellipodia to orchestrate cell migration. Mol Cell Biol 28: 5687–5697 Takabayashi T, Xie MJ, Takeuchi S, Kawasaki M, Yagi H, Okamoto M, et al., 2010, LL5bdirects the translocation of filamin A and SHIP2 to sites of phosphatidylinositol 3,4,5-triphosphate, PtdIns(3,4,5)P3, accumulation, and PtdIns(3,4,5)P3 localization is mutually modified by co-recruited SHIP2. J Biol Chem 285: 16155–16165 Gawecka JE, Griffiths GS, Ek-Rylander B, Ramos JW, Matter ML, 2010, R-Ras regulates migration through an interaction with filamin A in melanoma cells. PLoS ONE 5:e11269. Bedolla RG, Wang Y, Asuncion A, Chamie K, Siddiqui S, Mudryj MM, et al., 2009, Nuclear versus cytoplasmic localization of filamin A in prostate cancer: immunohistochemical correlation with metastases. Clin Cancer Res 15:788–796 Browne KA, Johnstone RW, Jans DA, Trapani JA, 2000, Filamin, 280-kDa actin-binding protein, is a caspase substrate and is also cleaved directly by the cytotoxic T lymphocyte protease granzyme B during apoptosis. J Biol Chem 275:39262–39266 Zhou AX, Toylu A, Nallapalli RK, Nilsson G, Atabey N, Heldin CH, et al., 2011, Filamin A mediates HGF/c-MET signaling in tumor cell migration. Int J Cancer 128:839–846 Silver DA, Pellicer I, Fair WR, Heston WD, Cordon-Cardo C, 1997, Prostate-specific membrane antigen expression in normal and malignant human tissues. Clin Cancer Res 3:81–85 Ghosh A, Heston WD, 2004, Tumor target prostate specific membrane antigen, PSMA, and its regulation in prostate cancer. J Cell Biochem 91:528–539 Rajasekaran AK, Anilkumar G, Christiansen JJ, 2005, Is prostatespecific membrane antigen a multifunctional protein? Am J Physiol 288:C975–C981 Gopalakrishnapillai Anilkumar, Sigrid A. Rajasekaran, SongWang, Oliver Hankinson, Neil H. Bander, Rajasekaran AK et al., 2003, Prostate-specific membrane antigen association with filamin A modulates its internalization and NAALADase activity. Cancer Res 63:2645–2648. Ozanne DM, Brady ME, Cook S, Gaughan L, Neal DE, Robson CN, 2000, Androgen receptor nuclear translocation is facilitated by the f-actin cross-linking protein filamin. Mol Endocrinol 14:1618– 1626 Wang Y, Kreisberg JI, Bedolla RG, Mikhailova M, de Vere White RW, Ghosh PM(2007, A 90MkDa fragment of filamin A promotes Casodex-induced growth inhibition in Casodex-resistant androgen receptor positiveC4-2 prostate cancer cells. Oncogene 26:6061– 6070 Loy CJ, Sim KS, Yong EL, 2003, Filamin-A fragment localizes to the nucleus to regulate androgen receptor and coactivator functions. PNAS 100:4562–4567 Giovannelli P, Di Donato M, Auricchio F, Castoria G, 2014, Analysis of the androgen receptor/filamin A complex in stromal cells. Methods Mol Biol 1204:109–121 Sun GG, Lu YF, Zhang J, Hu WN, 2014, Filamin A regulates MMP-9 expression and suppresses prostate cancer cell migration and invasion. Tumor Biol 35:3819–3826 CatalonaWJ, 1994, Management of cancer of the prostate. N Engl J Med 331:996–1004 Gustavsson H, Welen K, Damber JE, 2005, Transition of an androgen-dependent human prostate cancer cell line into an androgen-independent subline is associated with increased angiogenesis. Prostate 62:364–373 Liu T, Mendes DE, Berkman CE, 2014, Prolonged androgen deprivation leads to overexpression of calpain 2: implications for prostate cancer progression. Int J Oncol 44:467–472 Mooso BA,Vinall RL, Tepper CG, SavoyRM, Cheung JP, Singh S, et al., 2012, Enhancing the effectiveness of androgen deprivation in prostate cancer by inducing filamin A nuclear localization. Endocr Relat Cancer 19:795–777 Sun G-G, Wei C-D, Jing S-W, Wan-Ning H, 2014, Interactions between filamin A and MMP-9 regulate proliferation and invasion in renal cell carcinoma. Asian Pac J Cancer Prev 15:3789–3795 Xu Y, Bismar TA, Su J, Xu B, Kristiansen G, Varga Z, et al., 2010, Filamin A regulates focal adhesion disassembly and suppresses breast cancer cell migration and invasion. J Exp Med 207:2421– 2437 Ravid D, Maor S, Werner H, Liscovitch M, 2005, Caveolin-1 inhibits cell detachment-induced p53 activation and anoikis by upregulation of insulin-like growth factor-I receptors and signaling. Oncogene 24:1338–1347 Ravid D, Chuderland D, Landsman L, Lavie Y, Reich R, Liscovitch M, 2008, Filamin A is a novel caveolin-1-dependent target in IGFI- stimulated cancer cell migration. Exp Cell Res 314:2762–2773 Zhong Z, Yeow WS, Zou C,Wassell R,Wang C, Pestell RG, et al., 2010, Cyclin D1/cyclin-dependent kinase 4 interacts with filamin A and affects the migration and invasion potential of breast cancer cells. Cancer Res 70:2105–2114 Jiang X, Yue J, Lu H, Campbell N, Yang Q, Lan S, et al., 2013, Inhibition of filamin-A reduces cancer metastatic potential. Int J Biol Sci 9:67–77 Hammer A, Rider L, Oladimeji P, Cook L, Li Q, Mattingly RR, Diakonova M, 2013, Tyrosyl phosphorylated PAK1 regulates breast cancer cell motility in response to prolactin through filamin A. Mol Endocrinol 27:455–465 Alper O, Stetler-Stevenson WG, Harris LN, Leitner WW, Ozdemirli M, Hartmann D, et al., 2009, Novel anti-filamin-A antibody detects a secreted variant of filamin-A in plasma from patients with breast carcinoma and high-grade astrocytoma. Cancer Sci 100:1748–1756 Tian H-M, Liu X-H, Han W, Zhao L-L, Yuan B, Yuan C-J, 2013, Differential expression of filamin A and its clinical significance in breast cancer. Oncol Lett 6:6681–6686. Sun GG, Sheng SH, Jing SW, Hu WN, 2014, An antiproliferative gene FLNA regulates migration and invasion of gastric carcinoma cell in vitro and its clinical significance. Tumor Biol 35:2641–2648 Tian ZQ, Shi JW, Wang XR, Li Z, Wang GY, 2015, New cancer suppressor gene for colorectal adenocarcinoma: filamin A.World J Gastroenterol 21:2199–2205 Li C, Yu S, Nakamura F, Yin S, Xu J, Petrolla AA, et al., 2009, Binding of pro-prion to filamin A disrupts cytoskeleton and correlates with poor prognosis in pancreatic cancer. J Clin Invest 119: 2725–2736 Sy MS, Li C, Yu S, XinW(2010, The fatal attraction between proprion and filamin A: prion as a marker in human cancers. Biomark Med 4:453–464 Keshamouni VG, Michailidis G, Grasso CS, Anthwal S, Strahler JR,Walker A, et al., 2006, Differential protein expression profiling by iTRAQ-2DLC-MS/MS of lung cancer cells undergoing epithelial-mesenchymal transition reveals amigratory/invasive phenotype. J Proteome Res 5:1143–1154 Nallapalli RK, Ibrahim MX, Zhou AX, Bandaru S, Sunkara SN, Redfors B, et al., 2012, Targeting filamin A reduces K-RAS–induced lung adenocarcinomas and endothelial response to tumor growth in mice. Mol Cancer 11:50. Uramoto H, Akyurek LM, Hanagiri T, 2010, A positive relationship between filamin and VEGF in patients with lung cancer. Anticancer Res 30:3939–3944 Sun GG, Lu YF, Cheng YJ, Hu WN, 2014, Absent expression of FLNA is correlated with poor prognosis of nasopharyngeal cancer. Tumor Biol 35:2967–2974 Kamochi N, Nakashima M, Aoki S, Uchihashi K, Sugihara H, Toda S, et al., 2008, Irradiated fibroblast-induced bystander effects on invasive growth of squamous cell carcinoma under cancer–stromal cell interaction. Cancer Sci 99:2417–2427 Zhang K, Zhu T, Gao D, ZhangY, Zhao Q, Liu S, Su T, et al., 2014, Filamin A expression correlates with proliferation and invasive properties of human metastatic melanoma tumors: implications for survival in patients. J Cancer Res Clin Oncol 140:1913–1926 Li C, Yu S, Nakamura F, Pentikainen OT, Singh N, Yin S, et al., 2010, Pro-prion binds filamin A, facilitating its interaction with integrin1, and contributes to melanomagenesis. J Biol Chem 285: 30328–30339 HosakaM,Murase N, Orito T, MoriM(1985, Immunohistochemical evaluation of factor VIII related antigen, filament proteins and lectin binding in haemangiomas.VirchowsArch APathol Anat Histopathol 407:237–247 Bachmann AS, Howard JP, Vogel CW, 2006, Actin-binding protein filamin A is displayed on the surface of human neuroblastoma cells. Cancer Sci 97:1359–1365 Porter RM, Holme TC, Newman EL, Hopwood D, Wilkinson JM, Cuschieri A, 1993, Monoclonal antibodies to cytoskeletal proteins: an immunohistochemical investigation of human colon cancer. J Pathol 170:435–440 Larriba MJ, Martin-Villar E, Garcia JM, Pereira F, Pena C, de Herreros AG, et al., 2009, Snail2 cooperates with Snail1 in the repression of vitamin D receptor in colon cancer. Carcinogenesis 30:1459–1468 Guedj N, Zhan Q, Perigny M, Rautou PE, Degos F, Belghiti J, et al., 2009, Comparative protein expression profiles of hilar and peripheral hepatic cholangiocarcinomas. J Hepatol 5:193–101 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 245 EP 252 DI 10.1007/s12253-015-9980-1 PG 8 ER PT J AU Zeng, H Zhang, Z Dai, X Chen, Y Ye, J Jin, Z AF Zeng, Haibin Zhang, Zhong Dai, Xiaoming Chen, Yongzhong Ye, Jianhua Jin, Zhixin TI Increased Expression of microRNA-199b-5p Associates with Poor Prognosis Through Promoting Cell Proliferation, Invasion and Migration Abilities of Human Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; microRNA-199b-5p; Clinicopathological characteristics; Prognosis; Carcinogenesis ID Osteosarcoma; microRNA-199b-5p; Clinicopathological characteristics; Prognosis; Carcinogenesis AB MicroRNA (miR)-199b-5p has been reported to be upregulated in human osteosarcoma tissues and participate in the Notch signaling in osteosarcoma cells. This study was aimed to investigate the associations of miR-199b-5p expression with tumor progression of primary osteosarcoma, and to deepen the understanding of its involvement in carcinogenesis. Quantitative real-time reverse transcriptase-polymerase chain reaction was performed to detect expression levels of miR-199b-5p in 98 osteosarcoma and corresponding adjacent normal tissues. Then, the correlations of its expression with clinicopathological characteristics and patient prognosis were statistically analyzed. Moreover, in vitro assays were performed to assess the effects of miR-199b-5p on the proliferation, migration and invasion of two human osteosarcoma cell lines MG63 and U2OS. Compared to normal controls, miR- 199b-5p expression was significantly upregulated in osteosarcoma tissues (P<0.001). In addition, the expression levels of miR-199b-5p in osteosarcoma patients with high tumor grade (P=0.008), positive metastasis (P=0.001) and positive recurrence (P=0.001) were markedly higher than those with low tumor grade, negative metastasis and negative recurrence. Moreover, osteosarcoma patients with high miR-199b-5p expression showed shorter overall survival (P<.001) and shorter disease-free survival (P<0.001) than those with low expression. Furthermore, the inhibition of miR-199b-5p significantly suppressed cell proliferation, and reduced the migratory and invasive abilities of osteosarcoma cells. This study offer the convincing evidence for the first time that the increased expression of miR-199b-5p may play crucial roles in aggressive progression and poor prognosis of human osteosarcoma. miR- 199b-5p may function as an oncogene by positively regulating the malignant potentials of this neoplasm. C1 [Zeng, Haibin] Fuzhou General Hospital of Nanjing Military Region, 476 Clinical Division, Department of Orthopedics, 350002 Fuzhou, China. [Zhang, Zhong] Fuzhou General Hospital of Nanjing Military Region, 476 Clinical Division, Department of Orthopedics, 350002 Fuzhou, China. [Dai, Xiaoming] Fuzhou General Hospital of Nanjing Military Region, 476 Clinical Division, Department of Orthopedics, 350002 Fuzhou, China. [Chen, Yongzhong] Fuzhou General Hospital of Nanjing Military Region, 476 Clinical Division, Department of Orthopedics, 350002 Fuzhou, China. [Ye, Jianhua] Fuzhou General Hospital of Nanjing Military Region, 476 Clinical Division, Department of Orthopedics, 350002 Fuzhou, China. [Jin, Zhixin] Fuzhou General Hospital of Nanjing Military Region, 476 Clinical Division, Department of Orthopedics, 350002 Fuzhou, China. RP Zeng, H (reprint author), Fuzhou General Hospital of Nanjing Military Region, 476 Clinical Division, Department of Orthopedics, 350002 Fuzhou, China. EM Zenghaib139@163.com CR Marina N, Gebhardt M, Teot L, Gorlick R, 2004, Biology and therapeutic advances for pediatric osteosarcoma. Oncologist 9:422–441 Longhi A, Errani C, De Paolis M, Mercuri M, Bacci G, 2006, Primary bone osteosarcoma in the pediatric age: state of the art. Cancer Treat Rev 32:423–436 Bielack SS, Marina N, Ferrari S, 2008, Osteosarcoma: the same old drugs or more? J Clin Oncol 26:3102–3103 Broadhead ML, Clark JC, Myers DE, Dass CR, Choong PF, 2011, The molecular pathogenesis of osteosarcoma: a review. Sarcoma 2011:959248 Kansara M, ThomasDM(2007, Molecular pathogenesis of osteosarcoma. DNA Cell Biol 26:1–18 Carrington JC, Ambros V, 2003, Role of microRNAs in plant and animal development. Science 301:336–338 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281–297 Brodersen P, Voinnet O, 2009, Revisiting the principles of microRNA target recognition and mode of action. Nat Rev Mol Cell Biol 10:141–148 Garzon R, Marcucci G, Croce CM, 2010, Targeting microRNAs in cancer: rationale, strategies and challenges. Nat Rev Drug Discov 9: 775–789 Calin GA, Croce CM, 2006, MicroRNA signatures in human cancers. Nat Rev Cancer 6:857–866 Ji F, Zhang H, Wang Y, 2013, MicroRNA-133a, downregulated in osteosarcoma, suppresses proliferation and promotes apoptosis by targeting Bcl-xL and Mcl-1. Bone 56:220–226 Pan W, Wang H, Jianwei R, Ye Z, 2014, MicroRNA-27a promotes proliferation, migration and invasion by targeting MAP2K4 in human osteosarcoma cells. Cell Physiol Biochem 33:402–412 Novello C, Pazzaglia L, Cingolani C, 2013, miRNA expression profile in human osteosarcoma: role of miR-1 and miR-133b in proliferation and cell cycle control. Int J Oncol 42:667–675 Zhang K, Zhang C, Liu L, Zhou J, 2014, A key role of microRNA-29b in suppression of osteosarcoma cell proliferation and migration via modulation of VEGF. Int J Clin Exp Pathol 7:5701–5708 Shen L, Wang P, Yang J, Li X, 2014, MicroRNA-217 regulates WASF3 expression and suppresses tumor growth and metastasis in osteosarcoma. PLoS ONE 9:e109138 Geng S, Zhang X, Chen J, Liu X, Zhang H, Xu X,Ma Y, Li B, Zhang Y, Bi Z, Yang C, 2014, The tumor suppressor role of miR-124 in osteosarcoma. PLoS ONE 9:e91566 Flamant S, Ritchie W, Guilhot J, 2010, Micro-RNA response to imatinib mesylate in patients with chronic myeloid leukemia. Haematologica 95:1325–1333 Garzia L, Andolfo I, Cusanelli E, 2009, MicroRNA-199b-5p impairs cancer stem cells through negative regulation of HES1 in medulloblastoma. PLoS ONE 4:e4998 Won KY, Kim YW, Kim HS, Lee SK, Jung WW, Park YK, 2013, MicroRNA-199b-5p is involved in the Notch signaling pathway in osteosarcoma. Hum Pathol 44:1648–1655 Fang C, Zhao Y, Guo B, 2013, MiR-199b-5p targets HER2 in breast cancer cells. J Cell Biochem 114:1457–1463 Liu MX, Siu MK, Liu SS, Yam JW, Ngan HY, Chan DW, 2014, Epigenetic silencing of microRNA-199b-5p is associated with acquired chemoresistance via activation of JAG1-Notch1 signaling in ovarian cancer. Oncotarget 5:944–958 Li Z, Gu X, Fang Y, Xiang J, Chen Z, 2012, microRNA expression profiles in human colorectal cancers with brain metastases. Oncol Lett 3:346–350 Li Y, Bai H, Zhang Z, 2014, The up-regulation of miR-199b-5p in erythroid differentiation is associated with GATA-1 and NF-E2. Mol Cell 37:213–219 Bacci G, Bertoni F, Longhi A, 2003, Neoadjuvant chemotherapy for high-grade central osteosarcoma of the extremity. Histologic response to preoperative chemotherapy correlates with histologic subtype of the tumor. Cancer 97:3068–3075 Favreau AJ, Cross EL, Sathyanarayana P, 2012, miR-199b-5p directly targets PODXL and DDR1 and decreased levels of miR-199b-5p correlate with elevated expressions of PODXL and DDR1 in acute myeloid leukemia. Am J Hematol 87: 442–446 Zhang H, Cai X, Wang Y, Tang H, Tong D, Ji F, 2010, MicroRNA- 143, down-regulated in osteosarcoma, promotes apoptosis and suppresses tumorigenicity by targeting Bcl-2. Oncol Rep 24:1363–1369 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using realtime quantitative PCR and the 2−ΔΔCt method. Methods 25:402–408 Andolfo I, Liguori L, De Antonellis P, 2012, The micro-RNA 199b-5p regulatory circuit involves Hes1, CD15, and epigenetic modifications in medulloblastoma. Neuro Oncol 14:596– 612 Lauvrak SU, Munthe E, Kresse SH, Stratford EW, Namlos HM, Meza-Zepeda LA, Myklebost O, 2013, Functional characterisation of osteosarcoma cell lines and identification ofmRNAs and miRNAs associated with aggressive cancer phenotypes. Br J Cancer 109: 2228–2236 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 253 EP 260 DI 10.1007/s12253-015-9901-3 PG 8 ER PT J AU Xu, L Tang, W AF Xu, Liping Tang, Wenru TI Associations of Polymorphisms in mir-196a2, mir-146a and mir-149 with Colorectal Cancer Risk: A Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mir-196a2; mir-146a; mir-149; CRC; SNP ID mir-196a2; mir-146a; mir-149; CRC; SNP AB MicroRNAs (miRNAs) are non-coding RNAs which act as tumor suppressors or oncogenes. And single nucleotide polymorphism (SNP) in miRNA regions is one type of genetic variations in human genome. Various studies have investigated the associations of miRNAs SNP and kinds of cancers. In this article, we searched eligible studies to explore the relationships between mir-196a2 /mir-146a /mir- 149 polymorphisms and colorectal cancer (CRC). A literature search of PubMed, Web of Science and ScienceDirect was conducted to identify all relevant studies. Three genetic models with pooled ratio and 95 % confidence interval were used to evaluate the associations. We found that mir-196a2 polymorphism was significantly associated with CRC in Asian group (additive model: OR=1.197, 95 %CI 1.084~ 1.32, P<0.001; dominant model: OR=1.247, 95 %CI 1.065 ~1.46, P=0.006; recessive model: OR=1.298, 95 %CI 1.101 ~1.531, P=0.002). And no associations were observed between SNPs of mir-146a, mir-149 and CRC in three genetic models.We also found CRC risk was not associated with mir- 146a and mir-149 polymorphisms in population subgroup analysis. The current meta-analysis suggests that mir-196a2 polymorphism is associated with CRC, especially in Asian group. While, no associations have been found between mir- 146a /mir-149 polymorphisms and CRC. C1 [Xu, Liping] Kunming University of Science and Technology, Medical Faculty, Laboratory of Molecular Genetics of Aging & Tumor, Chenggong Campus, 727 South Jingming Road, 650500 Kunming, Yunnan, China. [Tang, Wenru] Kunming University of Science and Technology, Medical Faculty, Laboratory of Molecular Genetics of Aging & Tumor, Chenggong Campus, 727 South Jingming Road, 650500 Kunming, Yunnan, China. RP Tang, W (reprint author), Kunming University of Science and Technology, Medical Faculty, Laboratory of Molecular Genetics of Aging & Tumor, 650500 Kunming, China. EM 531081047@qq.com CR Ambros V, 2004, The functions of animal microRNAs. Nature 431: 350–355 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281–297 Bartels CL, Tsongalis GJ, 2009, MicroRNAs: novel biomarkers for human cancer. Clin Chem 55:623–631 Rosenfeld N, Aharonov R, Meiri E et al, 2008, MicroRNAs accurately identify cancer tissue origin. Nat Biotechnol 26:462–469 Bentwich I, 2005, Prediction and validation of microRNAs and their targets. FEBS Lett 579:5904–5910 Brennecke J, Stark A, Russell RB et al, 2005, Principles of microRNA-target recognition. PLoS Biol 3:e85 Yang W, Chendrimada TP, Wang Q et al, 2006, Modulation of microRNA processing and expression through RNA editing by ADAR deaminases. Nat Struct Mol Biol 13:13–21 Wu M, Jolicoeur N, Li Z et al, 2008, Genetic variations of microRNAs in human cancer and their effects on the expression of miRNAs. Carcinogenesis 29:1710–1716 Jemal A, Siegel R, Ward E et al, 2009, Cancer statistics, 2009. CA Cancer J Clin 59:225–249 Yang L, Parkin DM, Li L et al, 2003, Time trends in cancer mortality in China: 1987–1999. Int J Cancer 106:771–783 Center MM, Jemal A,Ward E, 2009, International trends in colorectal cancer incidence rates. Cancer Epidemiol Biomarkers Prev 18: 1688–1694 Zhan JF, Chen LH, Chen ZX et al, 2011, A functional variant in microRNA-196a2 is associated with susceptibility of colorectal cancer in a Chinese population. Arch Med Res 42:144– 148 Tian T, Shu Y, Chen J et al, 2009, A functional genetic variant in microRNA-196a2 is associated with increased susceptibility of lung cancer in Chinese. Cancer Epidemiol Biomarkers Prev 18:1183– 1187 Kim MJ, Yoo SS, Choi YYet al, 2010, A functional polymorphism in the pre-microRNA-196a2 and the risk of lung cancer in a Korean population. Lung Cancer 69:127–129 Hu Z, Liang J,Wang Z et al, 2009, Common genetic variants in premicroRNAs were associated with increased risk of breast cancer in Chinese women. Hum Mutat 30:79–84 Hoffman AE, Zheng T, Yi C et al, 2009, microRNAmiR-196a-2 and breast cancer: a genetic and epigenetic association study and functional analysis. Cancer Res 69:5970–5977 Qi P, Dou TH, Geng L et al, 2010, Association of a variant in MIR 196A2 with susceptibility to hepatocellular carcinoma in male Chinese patients with chronic hepatitis B virus infection. Hum Immunol 71:621–626 Egger M, Davey SG, SchneiderM et al, 1997, Bias in meta-analysis detected by a simple, graphical test. BMJ 315:629–634 Chen H, Sun LY, Chen LL et al, 2012, Avariant inmicroRNA-196a2 is not associated with susceptibility to and progression of colorectal cancer in Chinese. Intern Med J 42:e115–e119 Min KT, Kim JW, Jeon YJ et al, 2012, Association of the miR- 146aC>G, 149C>T, 196a2C>T, and 499A>G polymorphisms with colorectal cancer in the Korean population. Mol Carcinog 51(Suppl 1):E65–E73 Zhu L, Chu H, Gu D et al, 2012, A functional polymorphism in miRNA-196a2 is associated with colorectal cancer risk in a Chinese population. DNA Cell Biol 31:350–354 Hezova R, Kovarikova A, Bienertova-Vasku J et al, 2012, Evaluation of SNPs in miR-196-a2, miR-27a and miR-146a as risk factors of colorectal cancer. World J Gastroenterol 18:2827–2831 Vinci S, Gelmini S, Mancini I et al, 2013, Genetic and epigenetic factors in regulation ofmicroRNA in colorectal cancers.Methods 59: 138–146 Zhang MW, Jin MJ, Yu YX et al, 2012, Associations of lifestylerelated factors, hsa-miR-149 and hsa-miR-605 gene polymorphisms with gastrointestinal cancer risk. Mol Carcinog 51(Suppl 1):E21–E31 Lv M, Dong W, Li L et al, 2013, Association between genetic variants in pre-miRNA and colorectal cancer risk in a Chinese population. J Cancer Res Clin Oncol 139:1405–1410 Chae YS, Kim JG, Lee SJ et al, 2013, A miR-146a polymorphism, rs2910164, predicts risk of and survival from colorectal cancer. Anticancer Res 33:3233–3239 Ma L, Zhu L, Gu D et al, 2013, A genetic variant in miR-146a modifies colorectal cancer susceptibility in a Chinese population. Arch Toxicol 87:825–833 Vogelstein B, Kinzler KW, 2004, Cancer genes and the pathways they control. Nat Med 10:789–799 Mueller DW, Bosserhoff AK, 2011, MicroRNA miR-196a controls melanoma-associated genes by regulating HOX-C8 expression. Int J Cancer 129:1064–1074 Liu XH, Lu KH, Wang KM et al, 2012, MicroRNA-196a promotes non-small cell lung cancer cell proliferation and invasion through targeting HOXA5. BMC Cancer 12:348 Yuan Z, Zeng X, Yang D et al, 2013, Effects of common polymorphism rs11614913 in Hsa-miR-196a2 on lung cancer risk. PLoS One 8:e61047 Xu J, Hu Z, Xu Z et al, 2009, Functional variant in microRNA-196a2 contributes to the susceptibility of congenital heart disease in a Chinese population. Hum Mutat 30:1231–1236 Hu Z, Chen J, Tian T et al, 2008, Genetic variants of miRNA sequences and non-small cell lung cancer survival. J Clin Invest 118:2600–2608 Wei J, Zheng L, Liu S et al, 2013, MiR-196a2 rs11614913 T>C polymorphism and risk of esophageal cancer in a Chinese population. Hum Immunol 74:1199–1205 Wang S, Tao G, Wu D et al, 2013, A functional polymorphism in MIR196A2 is associated with risk and prognosis of gastric cancer. Mol Carcinog 52(Suppl 1):E87–E95 Zhang H, Su YL, Yu H et al, 2012, Meta-analysis of the association between Mir-196a-2 polymorphism and cancer susceptibility. Cancer Biol Med 9:63–72 Li YJ, Zhang ZY,MaoYYet al, 2014)Agenetic variant in MiR-146a modifies digestive system cancer risk: a meta-analysis. Asian Pac J Cancer Prev 15:145–150 Taganov KD, Boldin MP, Chang KJ et al, 2006, NF-kappaBdependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci U S A 103:12481–12486 Pastrello C, Polesel J, Della PL et al, 2010, Association between hsamir- 146a genotype and tumor age-of-onset in BRCA1/BRCA2- negative familial breast and ovarian cancer patients. Carcinogenesis 31:2124–2126 Wang PY, Gao ZH, Jiang ZH et al, 2013, The associations of single nucleotide polymorphisms in miR-146a, miR-196a and miR-499 with breast cancer susceptibility. PLoS One 8:e70656 Zhou B, Dong LP, Jing XY et al, 2014, Association between miR- 146aG>C and miR-196a2C>T polymorphisms and the risk of hepatocellular carcinoma in a Chinese population. Biol, Tumour Lin RJ, Lin YC, Yu AL, 2010, miR-149* induces apoptosis by inhibiting Akt1 and E2F1 in human cancer cells. Mol Carcinog 49: 719–727 Oster B, Linnet L, Christensen LL et al, 2013, Non-CpG island promoter hypomethylation and miR-149 regulate the expression of SRPX2 in colorectal cancer. Int J Cancer 132:2303–2315 Zhang J, Liu YF, Gan Y, 2012, Lack of association between miR- 149 C>T polymorphism and cancer susceptibility: a meta-analysis based on 4,677 cases and 4,830 controls. Mol Biol Rep 39:8749– 8753 Zhang YG, Shi JX, Song CH et al, 2013, Association of mir-499 and mir-149 polymorphisms with cancer risk in the Chinese population: evidence from published studies. Asian Pac J Cancer Prev 14:2337– 2342 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 261 EP 267 DI 10.1007/s12253-014-9843-1 PG 7 ER PT J AU Giaginis, C Alexandrou, P Poulaki, E Delladetsima, I Troungos, C Patsouris, E Theocharis, S AF Giaginis, Constantinos Alexandrou, Paraskevi Poulaki, Elpida Delladetsima, Ioanna Troungos, Constantinos Patsouris, Efstratios Theocharis, Stamatios TI Clinical Significance of EphB4 and EphB6 Expression in Human Malignant and Benign Thyroid Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ephrin receptors; Thyroid malignancy; Clinicopathological parameters; Immunohistochemistry; Papillary carcinoma; Hyperplastic nodule ID Ephrin receptors; Thyroid malignancy; Clinicopathological parameters; Immunohistochemistry; Papillary carcinoma; Hyperplastic nodule AB Ephrin receptors (Ephs) are frequently overexpressed in a wide variety of human malignant tumors, being associated with tumor growth, invasion, angiogenesis and metastasis. The present study aimed to evaluate the clinical significance of EphB4 and EphB6 protein expression in human malignant and benign thyroid lesions. EphB4 and EphB6 protein expression was assessed immunohistochemically on paraffinembedded thyroid tissues obtained from 127 patients with benign (n=71) and malignant (n=56) thyroid lesions. Enhanced EphB4 and EphB6 expression was more frequently observed in malignant compared to benign thyroid lesions (p=0.0508 and p=0.0006, respectively). EphB4 and EphB6 expression also provided a distinct discrimination between papillary carcinoma and hyperplastic nodules (p=0.0302 and p=0.0013, respectively). In malignant thyroid lesions, enhanced EphB4 expression was significantly associated with larger tumor size (p=0.0366). Enhanced EphB6 expression was significantly associated with larger tumor size (p=0.0366), the presence of lymph node metastases (p=0.0023), the presence of capsular (p=0.0038), lymphatic (p=0.0053) and vascular invasion (p=0.0018) and increased risk of recurrence rate (p=0.0038). The present study supported evidence that EphB4 and mainly EphB6 may participate in the malignant thyroid transformation, reinforcing their utility as useful biomarkers and possible therapeutic targets in this type of neoplasia. C1 [Giaginis, Constantinos] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str., 11527 Goudi, Athens, Greece. [Alexandrou, Paraskevi] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str., 11527 Goudi, Athens, Greece. [Poulaki, Elpida] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str., 11527 Goudi, Athens, Greece. [Delladetsima, Ioanna] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str., 11527 Goudi, Athens, Greece. [Troungos, Constantinos] University of Athens, Medical School, Department of BiochemistryAthens, Greece. [Patsouris, Efstratios] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str., 11527 Goudi, Athens, Greece. [Theocharis, Stamatios] University of Athens, Medical School, First Department of Pathology, 75 M. Asias str., 11527 Goudi, Athens, Greece. RP Theocharis, S (reprint author), University of Athens, Medical School, First Department of Pathology, 11527 Goudi, Greece. EM stamtheo@med.uoa.gr CR Zhang J, Hughes SE, 2006, Role of the ephrin and Ephrin receptor tyrosine kinase families in angiogenesis and development of the cardiovascular system. J Pathol 208:453–461 Pasquale EB, 2008, Eph-ephrin bidirectional signaling in physiology and disease. Cell 133:38–52 Nakamoto M, Bergemann AD, 2002, Diverse role for the Eph family of receptor tyrosine kinases in carcinogenesis. Microscopy Res Technique 59:58–67 Surawska H, Ma PC, Salgia R, 2004, The role of ephrins and Eph receptors in cancer. Cytokine Growth Factor Rev 15:419–433 Cheng N, Brantley DM, Chen J, 2002, The ephrins and Eph receptors in angiogenesis. Cytokine Growth Factor Rev 13:75–85 Brandley-SiedersDM, Chen J, 2007, Eph receptor tyrosine kinase in angiogenesis: from development to disease. Angiogenesis 7:17–28 Castano J, DavalosV, Schwartz S Jr, Arango D, 2008, EPH receptors in cancer. Histol Histopathol 23:1011–1023 Leenhardt L, Grosclaude P, Cherie-Challine L, 2004, Increased incidence of thyroid carcinoma in France: a true epidemic or thyroid nodule management effects? Report from the French Thyroid Cancer Committee. Thyroid 14:1056–1060 Jemal A, Siegel R, Ward E et al, 2009, Cancer statistics, 2009. CA Cancer J Clin 59:225–249 Hundahl SA, Fleming ID, Fremgen AM et al, 1998, A National Cancer Data Base report on 53,856 cases of thyroid carcinoma treated in the U.S., 1985–1995. Cancer 83:2638–2648 Gharib H, Papini E, 2007, Thyroid nodules: clinical importance, assessment, and treatment. Endocrinol Metab Clin North Am 36: 707–735 Schlumberger M, Lacroix L, Russo D, Filetti S, Bidart JM, 2007, Defects in iodide metabolism in thyroid cancer and implications for the follow-up and treatment of patients. Nat Clin Pract Endocrinol Metab 3:260–269 Stang MT, Carty SE, 2009, Recent developments in predicting thyroid malignancy. Curr Opin Oncol 21:11–17 Fischer S, Asa L, 2008, Application of immunohistochemistry to thyroid neoplasms. Arch Pathol Lab Med 132:359–372 Vriens MR, Schreinemakers JM, Suh I et al, 2009, Diagnostic markers and prognostic factors in thyroid cancer. Future Oncol 5: 1283–1293 Aisner DL, Sams SB, 2012, The role of cytology specimens in molecular testing of solid tumors: techniques, limitations, and opportunities. Diagn Cytopathol 40:511–524 Giaginis C, Tsourouflis G, Zizi-Serbetzoglou A, Kouraklis G, Chatzopoulou E, Theocharis S, 2010, Clinical significance of Ephrin, Eph)-A1, −A2, −A4, −A5 and -A7 receptors in pancreatic ductal adenocarcinoma. Pathol Oncol Res 16:267–276 Giaginis C, Tsoukalas N, Bournakis E et al, 2014, Ephrin, Eph, receptor A1, A4, A5 and A7 expression in human non-small cell lung carcinoma: associations with clinicopathological parameters, tumor proliferative capacity and patients’ survival. BMC Clin Pathol 14:8 Karidis NP, Giaginis C, Tsourouflis G, Alexandrou P, Delladetsima I, Theocharis S, 2011, Eph-A2 and Eph-A4 expression in human benign and malignant thyroid lesions: an immunohistochemical study. Med Sci Monit 17:BR257–BR265 Theocharis S, Klijanienko J, Giaginis C, Alexandrou P, Patsouris E, Sastre-Garau X, 2013, Ephrin receptor, Eph, -A1, −A2, −A4 and - A7 expression in mobile tongue squamous cell carcinoma: associations with clinicopathological parameters and patients survival. Pathol Oncol Res., DOI 10.1007/s12253-013-9692-3 Yin H, Lu C, Tang Y, Wang H, Wang H, Wang J, 2013, Enhanced expression of EphrinB1 is associated with lymph node metastasis and poor prognosis in breast cancer. Cancer Biomark 13:261–267 Ji XD, Li G, Feng YX et al, 2011, EphB3 is overexpressed in nonsmall- cell lung cancer and promotes tumor metastasis by enhancing cell survival and migration. Cancer Res 71:1156–1166 LiM, Zhao ZW, Zhang Y, Xin Y, 2011, Over-expression of Ephb4 is associated with carcinogenesis of gastric cancer. Dig Dis Sci 56:698– 706 Lu Z, Zhang Y, Li Z et al, 2012, Overexpression of the B-type Eph and ephrin genes correlates with progression and pain in human pancreatic cancer. Oncol Lett 3:1207–1212 Oshima T, Akaike M, Yoshihara K et al, 2008, Overexpression of EphA4 gene and reduced expression of EphB2 gene correlates with liver metastasis in colorectal cancer. Int J Oncol 33:573–577 Wang H, Wen J, Wang H et al, 2013, Loss of expression of EphB1 protein in serous carcinoma of ovary associated with metastasis and poor survival. Int J Clin Exp Pathol 7:313–321 XuqingW, Lei C, ZhengfaMet al, 2012, EphB4 is overexpressed in papillary thyroid carcinoma and promotes the migration of papillary thyroid cancer cells. Tumour Biol 33:1419–1427 Tang XX, Zhao H, Robinson ME et al, 2000, Implications of EPHB6, EFNB2, and EFNB3 expressions in human neuroblastoma. Proc Natl Acad Sci U S A 97:10936–10941 Hafner C, Bataille F, Meyer S et al, 2003, Loss of EphB6 expression in metastatic melanoma. Int J Oncol 23:1553–1559 Rosai J, Appendix C, 2004, Staging of cancer. In: Houston M, ed, Rosai and Ackerman’s surgical pathology, 9th edn. Mosby, London, pp 2809–2810 Tuttle RM, Tala H, Shah J et al, 2010, Estimating risk of recurrence in differentiated thyroid cancer after total thyroidectomy and radioactive iodine remnant ablation: using response to therapy variables to modify the initial risk estimates predicted by the new American Thyroid Association staging system. Thyroid 20:1341 Tu Y, He S, Fu J et al, 2012, Expression of EphrinB2 and EphB4 in glioma tissues correlated to the progression of glioma and the prognosis of glioblastoma patients. Clin Transl Oncol 14:214–220 Alam SM, Fujimoto J, Jahan I, Sato E, Tamaya T, 2009, Coexpression of EphB4 and ephrinB2 in tumor advancement of uterine cervical cancers. Gynecol Oncol 114:84–98 Zheng MF, Ji Y, Wu XB, Ye SG, Chen JY, 2012, EphB4 gene polymorphism and protein expression in non-small-cell lung cancer. Mol Med Rep 6:405–408 Sinha UK,Mazhar K, Chinn SB et al, 2006, The association between elevated EphB4 expression, smoking status, and advanced-stage disease in patients with head and neck squamous cell carcinoma. Arch Otolaryngol Head Neck Surg 132:1053–1059 Davalos V, Dopeso H, Castano J et al, 2006, EPHB4 and survival of colorectal cancer patients. Cancer Res 66:8943–8948 Takai N, Miyazaki T, Fujisawa K, Nasu K, Miyakawa I, 2001, Expression of receptor tyrosine kinase EphB4 and its ligand ephrin- B2 is associated with malignant potential in endometrial cancer. Oncol Rep 8:567–573 Alam SM, Fujimoto J, Jahan I, Sato E, Tamaya T, 2008, Coexpression of EphB4 and ephrinB2 in tumour advancement of ovarian cancers. Br J Cancer 98:845–851 Tang XX, Evans AE, Zhao H et al, 1999, High-level expression of EPHB6, EFNB2, and EFNB3 is associated with low tumor stage and high TrkA expression in human neuroblastomas. Clin Cancer Res 5: 1491–1496 Xi HQ, Wu XS,Wei B, Chen L, 2012, Eph receptors and ephrins as targets for cancer therapy. J Cell Mol Med 16:2894–2909 Ferguson BD, Liu R, Rolle CE et al, 2013, The EphB4 receptor tyrosine kinase promotes lung cancer growth: a potential novel therapeutic target. PLoS One 8:e67668 Hasina R, Mollberg N, Kawada I et al, 2013, Critical role for the receptor tyrosine kinase EPHB4 in esophageal cancers. Cancer Res 73:184–194 Spannuth WA, Mangala LS, Stone RL et al, 2010, Converging evidence for efficacy from parallel EphB4-targeted approaches in ovarian carcinoma. Mol Cancer Ther 9:2377–2388 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 269 EP 275 DI 10.1007/s12253-014-9879-2 PG 7 ER PT J AU Li, Y Dong, M Sheng, W Huang, L AF Li, Yuji Dong, Ming Sheng, Weiwei Huang, Longping TI Roles of Carbonic Anhydrase IX in Development of Pancreatic Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Carbonic anhydrase IX; Pancreatic cancer; Invasion; Migration ID Carbonic anhydrase IX; Pancreatic cancer; Invasion; Migration AB The aim of this study was to study the effects of carbonic anhydrase IX (CA IX) towards the invasion and metastasis of pancreatic cancer. The expressions of CA IX in 58 cases of pancreatic cancer and paired paracancerous normal tissues, obtained from 2005 to 2012 in the first Affiliated Hospital of China Medical University, were detected, as well as its expressions in different pancreatic cancer cell lines, aiming to detect the impacts of CA IX silencing towards the invasion and metastasis of pancreatic cancer cells. The CA IX expressions in 58 pancreatic cancer cases were higher than those in the paired paracancerous normal tissues (P<0.01), and positively correlated with the tumor size and the UICC staging UICC (P<0.05), the multivariate analysis showed that the high expression of CA IX was the independent risk factor towards the prognosis of pancreatic cancer (P<0.05). The CA IX was highly expressed in AxPC–1 and Miapaca–2, and the interference effects were significant. CA IX silencing could significantly inhibit the invasion and metastasis of AxPC-1 and Miapaca. We support a pro-tumor role of CA IX in the development and progression of pancreatic cancer. C1 [Li, Yuji] China Medical University, The First Affiliated Hospital, Department of General Surgery, 110001 Shenyang, China. [Dong, Ming] China Medical University, The First Affiliated Hospital, Department of General Surgery, 110001 Shenyang, China. [Sheng, Weiwei] China Medical University, The First Affiliated Hospital, Department of General Surgery, 110001 Shenyang, China. [Huang, Longping] China Medical University, The First Affiliated Hospital, Department of General Surgery, 110001 Shenyang, China. RP Dong, M (reprint author), China Medical University, The First Affiliated Hospital, Department of General Surgery, 110001 Shenyang, China. EM mingdongcn@163.com CR Jemal A, Siegel R, Xu J, Ward E, 2010, Cancer statistics. CA Cancer J Clin 60:277–300., DOI 10.3322/caac.20073 Philip PA, Mooney M, Jaffe D, Eckhardt G,Moore M,Meropol N, Emens L, O’Reilly E, Korc M, Ellis L, Benedetti J, Rothenberg M, Willett C, Tempero M, Lowy A, Abbruzzese J, Simeone D, Hingorani S, Berlin J, Tepper J, 2009, Consensus report of the national cancer institute clinical trials planning meeting on pancreas cancer treatment. J Clin Oncol 27:5660–5669., DOI 10.1200/JCO. 2009.21.9022 Ivanov S, Liao SY, Ivanova A, Danilkovitch-Miagkova A, Tarasova N, Weirich G, Merrill MJ, Proescholdt MA, Oldfield EH, Lee J, Zavada J, Waheed A, Sly W, Lerman MI, Stanbridge EJ, 2001, Expression of hypoxia-induc-ible cell-surface transmembrane carbonic anhydrase in human cancer. Am J Pathol 158:905– 919., DOI 10.1016/S0002-9440(10)64038-2 Sundfor K, LyngH REK, 1998, Tumour hypoxia and vascular density as predictors of metastasis in squamouscell carcinoma of the uterine cervix. Br J Cancer 78:822–827 Brizel DM, ScullySP HJM, Layfield LJ, Bean JM, Prosnitz LR, Dewhirst MW, 1996, Tumor oxygenation predicts for the likelihood of distant metastases in humans of tissue sarcoma. Cancer Res 56:941–943 Kim SJ, Rabbani ZN, Dewhirst MW, Vujaskovic Z, Vollmer RT, Schreiber EG, Oosterwijk E, KelleyMJ, 2005, Expression of HIF- 1, CA IX, VEGF, and MMP-9 in surgically resected non-small cell lung cancer. Lung Cancer 49:325–335., DOI 10.1016/j.lungcan. 2005.03.036 Klatte T, Seligson DB, Rao JY, Yu H, de Martino M, Kawaoka K, Wong SG, Belldegrun AS, Pantuck AJ, 2009, Carbonic anhydrase IX in bladder cancer: a diagnostic, prognostic, and the rapeutic moleeular marker. Cancer 115:1448–1458., DOI 10.1002/cncr.24163 Rasheed S, Harris AL, Tekkis PP, Turley H, Silver A, McDonald PJ, Talbot IC, Glynne-Jones R, Northover JM, Guenther T, 2009, Assessment of microvessel density and carbonic anhydrase-9, CA- 9, expression in rectal cancer. Pathol Res Pract 205:1–9., DOI 10. 1016/j.prp.2008.08.008 Choueiri TK, Regan MM, Rosenberg JE, Oh WK, Clement J, Amato AM, McDermott D, Cho DC, Atkins MB, Signoretti S, 2010, Carbonic anhydrase IX and pathological features as predictors of outcome in patients with metastatic clear- cell renal cell carcinoma receiving vascular endothelia growth factor- targeted therapy. BJU Int 106:772–778., DOI 10.1111/j.1464-410X.2010. 09218.x Robertson N, Potter C, Harris AL, 2004, Role of carbonic anhydrase IX in human tumor cell growth, survival, and invasion. Cancer Res 64:6160–6165., DOI 10.1158/0008-5472.CAN-03-2224 Juhasz M, Chen J, Lendeckel U, Kellner U, KasperHU, Tulassay Z, Pastorekova S, Malfertheiner P, Ebert MP D2003] Expression of carbonic anhydrase IX in human pancreatic cancer. Aliment Pharmacol Ther 18:837–846, pii:1738 ShengW, Dong M, Zhou J, Li X, Dong Q, 2013, Down regulation of CAII is associated with tumor differentiation and poor prognosis in patients with pancreatic cancer. J Surg Oncol 107:536–543., DOI 10.1002/jso.23282 Masunaga R, Kohno H, Dhar DK, Ohno S, Shibakita M, Kinugasa S, Yoshimura H, Tachibana M, Kubota H, Nagasue N, 2000, Cyclooxygenase-2 expression correlates with tumor neovascularization and prognosis in human colorectal carcinoma patients. Clin Cancer Res 6:4064–4068 Stathis A, Moore MJ, 2010, Advanced pancreatic carcinoma: current treatment and future challenges. Nat Rev Clin Oncol 7:163– 172., DOI 10.1038/nrclinonc.2009.236 Kivela AJ, Parkkila S, Saarnio J, Karttunen TJ, Kivela J, Parkkila AK, Pastorekova S, Pastorek J, Waheed A, Sly WS, Rajaniemi H, 2000, Expression of transmembrane carbonic anhydrase isoenzymes IX and XII in normal human pancreas and pancreatic tumours. Histochem Cell Biol 114:197–204 Kivela AJ, Saarnio J, Karttunen TJ, Kivela J, Parkkila AK, Pastorekova S, Pastorek J, Waheed A, Sly WS, Parkkila TS, Rajaniemi H, 2001, Differential expression of cytoplasmic carbonic anhydrases, CA I and II, and membrane-associated isozymes, CA IX and XII, in normal mucosa of large intestine and in colorectal tumors. Dig Dis Sci 46:2179–2186 Kim SJ, Rabbani ZN, Vollmer RT, Schreiber EG, Oosterwijk E, Dewhirst MW, Vujaskovic Z, Kelley MJ, 2004, Carbonic anhydrase IX in early-stage non small cell lung cancer. Clin Cancer Res 10:7925–7933., DOI 10.1158/1078-0432.CCR-04-0636 Choschzick M, Oosterwijk E, Muller V, Woelber L, Simon R, Moch H, Tennstedt P, 2011, Overexpression of carbonic anhydrase IX, CA IX, is an independent unfavorable prognostic marker in endometrioid ovarian cancer. Virchows Arch 459:193–200., DOI 10.1007/s00428-011-1105-y Schutze D, Milde-Langosch K, Witzel I, Rody A, Karn T, Schmidt M, Choschzick M, Janicke F, MullerV, 2013, Relevance of cellular and serum carbonic anhydrase IX in primary breast cancer. J Cancer Res Clin Oncol 139:747–754., DOI 10.1007/s00432-013-1378-4 Li MP, Ren LF, Cai HG, Yang HY, Lu B, Zhang P, Bao L, 2013, Significance of carbonic anhydrase IX protein expression in molecular subtyping of breast cancers. Zhonghua Bing Li Xue Za Zhi 42:182–185., DOI 10.3760/cma.j.issn. 0529-5807.2013.03.009 En-ping BA, Ya-li LV, Lin LIU, Bo ZHAO, 2011, Expression of carbonic anhydrase-9 correlates with malignant phenotypes of invasive breast ductal carcinoma. Chin J Clinicians, Electronic Edition, 5:4656–4660., DOI 10.3877/cma.j.issn. 1674-0785.2011. 16.083 Simi L, Venturini G, Malentacchi F, Gelmini S, Andreani M, Janni A, Pastorekova S, Supuran CT, Pazzagli M, Orlando C, 2006, Quantitative analysis of carbonic anhydrase IXmRNA in human non-small cell lung cancer. Lung Cancer 52:59–66., DOI 10.1016/j. lungcan.2005.11.017 Klimowicz AC, Bose P, Petrillo SK, Magliocco AM, Dort JC, BrocktonNT, 2013, The prognostic impact of a combined carbonic anhydrase IX and Ki67 signature in oral squamous cell carcinoma. Br J Cancer 109:1859–1866., DOI 10.1038/bjc.2013.533 Kim SJ, Shin HJ, Jung KY, Baek SK, Shin BK, Choi J, Kim BS, Shin SW, Kim YH, Kim JS, Oosterwijk E, 2007, Prognostic value of carbonic anhydrase IX and Ki-67 exression in squamous cell carcinoma of the tongue. J Clin Onco 37:812–819., DOI 10.1093/ jjco/hym121 Shin HJ, Rho SB, Jung DC, Han IO, Oh ES, Kim JY, 2011, Carbonic anhydrase IX, CA9, modulates tumor-associated cell migration and invasion. J Cell Sci 124:1077–1087., DOI 10.1242/jcs. 072207 Lou Y, McDonald PC, Oloumi A, Chia S, Ostlund C, Ahmadi A, Kyle A, Auf dem Keller U, Leung S, Huntsman D, Clarke B, Sutherland BW, Waterhouse D, Bally M, Roskelley C, Overall CM, Minchinton A, Pacchiano F, Carta F, Scozzafava A, Touisni N,Winum JY, Supuran CT, Dedhar S, 2011, Targeting tumor hypoxia: suppression of breast tumor growth and metastasis by novel carbonic anhydrase IX inhibitors. Cancer Res 71:3364–3376., DOI 10.1158/0008-5472.CAN-10-4261 De Craene B, Berx G, 2013, Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer 13:97– 110., DOI 10.1038/nrc3447 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 277 EP 286 DI 10.1007/s12253-015-9935-6 PG 10 ER PT J AU Shastry, HA Thota, B Mallavarapu, RS Arivazhagan, A Santosh, V AF Shastry, H Arun Thota, Balaram Mallavarapu, R Srividya Arivazhagan, Arimappamagan Santosh, Vani TI Nuclear Protein Phosphatase 1 α (PP1A) Expression is Associated with Poor Prognosis in p53 Expressing Glioblastomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma; Prognosis; TP53; Protein phosphatase 1 α; PPP1CA ID Glioblastoma; Prognosis; TP53; Protein phosphatase 1 α; PPP1CA AB Background: Protein phosphatase 1 α (PP1A) is an enzyme intimately associated with cell cycle, the over expression of which has been demonstrated in glioblastoma (GBM). Further, the nuclear expression of PP1A has been shown to be highly specific to GBM. In addition, PP1A has been shown to be a connecting molecule in the p53 containing GBM sub network. In view of these, we evaluated the prognostic relevance of PP1A. Methods: GBM tissues were examined for protein expression of PP1A by immunohistochemistry (IHC). Nuclear expression of PP1A was scored in all tumor tissue samples. Survival analyses were performed by Cox-Regression and Kaplan-Meier survival analysis with Log Rank tests. IDH1, ATRX and p53 IHC and stratification of all GBM cases were performed and subgroup specific evaluation of nuclear PP1A correlation with overall and progression free survival was performed. Results: PP1A protein expression showed no correlation with prognosis in all cases of GBM or on stratification based on IDH1 or ATRX expression. However on p53 stratification nuclear PP1A expression emerged as strong independent predictor of poor overall survival only in p53 positive GBMs both in univariate and multivariate analysis. Conclusions: While PP1A expression uniquely associates with poor prognosis only in p53 expressing GBMs, there is a notable absence of such correlation in p53 negative GBMs; thus skewing the overall relation of this molecule with prognosis in GBM. PP1A emerging as a strong prognostic marker in p53 expressing GBMs, enables us to foresee this molecule as a potential therapeutic target. C1 [Shastry, H Arun] National Institute of Mental Health and Neurosciences (NIMHANS), Department of Clinical Neurosciences, Hosur RoadBangalore, India. [Thota, Balaram] National Institute of Mental Health and Neurosciences, Department of Neuropathology, Hosur RoadBangalore, India. [Mallavarapu, R Srividya] National Institute of Mental Health and Neurosciences, Department of Neuropathology, Hosur RoadBangalore, India. [Arivazhagan, Arimappamagan] National Institute of Mental Health and Neurosciences, Department of Neurosurgery, Hosur RoadBangalore, India. [Santosh, Vani] National Institute of Mental Health and Neurosciences (NIMHANS), Department of Clinical Neurosciences, Hosur RoadBangalore, India. RP Santosh, V (reprint author), National Institute of Mental Health and Neurosciences (NIMHANS), Department of Clinical Neurosciences, Bangalore, India. EM vani.santosh@gmail.com CR Omuro A, DeAngelis LM, 2013, Glioblastoma and other malignant gliomas: a clinical review. JAMA 310:1842–50., DOI 10.1001/jama. 2013.280319 Ladha J, Donakonda S, Agrawal S et al, 2010, Glioblastoma-specific protein interaction network identifies PP1A and CSK21 as connecting molecules between cell cycle-associated genes. Cancer Res 70:6437–47., DOI 10.1158/0008-5472.CAN-10-0819 Ceulemans H, BollenM(2004, Functional diversity of protein phosphatase- 1, a cellular economizer and reset button. Physiol Rev 84:1– 39., DOI 10.1152/physrev.00013.2003 Wu JQ, Guo JY, Tang Wet al, 2009, PP1-mediated dephosphorylation of phosphoproteins atmitotic exit is controlled by inhibitor-1 and PP1 phosphorylation. Nat Cell Biol 11:644–51., DOI 10.1038/ ncb1871 Wang RH, Liu CW, Avramis VI, Berndt N, 2001, Protein phosphatase 1alpha-mediated stimulation of apoptosis is associated with dephosphorylation of the retinoblastoma protein. Oncogene 20:6111– 22., DOI 10.1038/sj.onc.1204829 Castro ME, Ferrer I, Cascon A et al, 2008, PPP1CA contributes to the senescence program induced by oncogenic Ras. Carcinogenesis 29:491–9., DOI 10.1093/carcin/bgm246 Lu Z,Wan G, Guo H et al, 2013, Protein phosphatase 1 inhibits p53 signaling by dephosphorylating and stabilizing Mdmx. Cell Signal 25:796–804., DOI 10.1016/j.cellsig.2012.12.014 Ruiz L, Traskine M, Ferrer I et al, 2008, Characterization of the p53 response to oncogene-induced senescence. PLoS One 3:e3230., DOI 10.1371/journal.pone.0003230 Thota B, Shukla SK, Srividya MR et al, 2012, IDH1 mutations in diffusely infiltrating astrocytomas: grade specificity, association with protein expression, and clinical relevance. Am J Clin Pathol 138: 177–84., DOI 10.1309/AJCPZOIY3WY4KIKE Haberler C, Wohrer A, 2014, Clinical Neuropathology practice news 2–2014: ATRX, a new candidate biomarker in gliomas. Clin Neuropathol 33:108–111., DOI 10.5414/NP300758 Hou LC, Veeravagu A, Hsu AR, Tse VCK, 2006, Recurrent glioblastoma multiforme: a review of natural history and management options. Neurosurg Focus 20:E5 Lamborn KR, Yung WKA, Chang SM et al, 2008, Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol 10:162–70., DOI 10.1215/ 15228517-2007-062 Weller M, Felsberg J, Hartmann C et al, 2009, Molecular predictors of progression-free and overall survival in patients with newly diagnosed glioblastoma: a prospective translational study of the German Glioma Network. J Clin Oncol 27:5743–50., DOI 10.1200/JCO.2009. 23.0805 Srividya MR, Thota B, Arivazhagan v et al, 2010, Age-dependent prognostic effects of EGFR/p53 alterations in glioblastoma: study on a prospective cohort of 140 uniformly treated adult patients. J Clin Pathol 63:687–91., DOI 10.1136/jcp.2009.074898 Hofstetter CP, Burkhardt J-K, Shin BJ et al, 2012, Protein phosphatase 2A mediates dormancy of glioblastoma multiforme-derived tumor stem-like cells during hypoxia. PLoS One 7:e30059., DOI 10. 1371/journal.pone.0030059 Lu J, Kovach JS, Johnson F et al, 2009, Inhibition of serine/ threonine phosphatase PP2A enhances cancer chemotherapy by blocking DNA damage induced defense mechanisms. Proc Natl Acad Sci U S A 106:11697–702., DOI 10.1073/ pnas.0905930106 Li J, Huang J, Li M et al, 2012, Anisomycin induces glioma cell death via down-regulation of PP2A catalytic subunit in vitro. Acta Pharmacol Sin 33:935–40., DOI 10.1038/aps.2012.46 Tsaytler P, Bertolotti A, 2013, Exploiting the selectivity of protein phosphatase 1 for pharmacological intervention. FEBS J 280:766– 70., DOI 10.1111/j.1742-4658.2012.08535.x Cohen PTW, 2002, Protein phosphatase 1–targeted in many directions. J Cell Sci 115:241–56 Chatterjee J, Kohn M, 2013, Targeting the untargetable: recent advances in the selective chemical modulation of protein phosphatase-1 activity. Curr Opin Chem Biol 17:361–8., DOI 10.1016/j.cbpa.2013. 04.008 Reddy SP, Britto R, Vinnakota K et al, 2008, Novel glioblastoma markers with diagnostic and prognostic value identified through transcriptome analysis. Clin Cancer Res 14:2978–87., DOI 10.1158/1078- 0432.CCR-07-4821 Pich A, Margaria E, Chiusa L, 2000, Oncogenes and male breast carcinoma: c-erbB-2 and p53 coexpression predicts a poor survival. J Clin Oncol 18:2948–56 Nakopoulou LL, Alexiadou A, Theodoropoulos GE et al, 1996, Prognostic significance of the co-expression of p53 and c-erbB-2 proteins in breast cancer. J Pathol 179:31–8. doi :10.1002/(SICI)1096-9896(199605)179:1<31::AIDPATH523> 3.0.CO;2-O Park YB, Kim HS, Oh JH, Lee SH, 2001, The co-expression of p53 protein and P-glycoprotein is correlated to a poor prognosis in osteosarcoma. Int Orthop 24:307–10 Xie Y, BulbulMA, Ji L et al, 2014, p53 expression is a strong marker of inferior survival in de novo diffuse large B-cell lymphoma and may have enhanced negative effect with MYC coexpression: a single institutional clinicopathologic study. Am J Clin Pathol 141:593–604., DOI 10.1309/AJCPPHMZ6VHF0WQV Takami H, Yoshida A, Fukushima S et al, 2014, Revisiting TP53 mutations and immunohistochemistry—a comparative study in 157 diffuse gliomas. Brain Pathol., DOI 10.1111/bpa.12173 Liu CWY, Wang R-H, Berndt N, 2006, Protein phosphatase 1alpha activity prevents oncogenic transformation. Mol Carcinog 45:648– 56., DOI 10.1002/mc.20191 Hsu L-C, Huang X, Seasholtz S et al, 2006, Gene amplification and overexpression of protein phosphatase 1alpha in oral squamous cell carcinoma cell lines. Oncogene 25:5517–26., DOI 10.1038/sj.onc. 1209563 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 287 EP 292 DI 10.1007/s12253-015-9928-5 PG 6 ER PT J AU Marton, I Krenacs, L Bagdi, E Bakos, A Demeter, J Borbenyi, Z AF Marton, Imelda Krenacs, Laszlo Bagdi, Eniko Bakos, Annamaria Demeter, Judit Borbenyi, Zita TI Clinical and Molecular Diagnostic Evaluation of Systemic Mastocytosis in the South-Eastern Hungarian Population Between 2001–2013 – A Single Centre Experience SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Myeloproliferative neoplasm; Mastocytosis; KIT D816V mutation; Orphan disease ID Myeloproliferative neoplasm; Mastocytosis; KIT D816V mutation; Orphan disease AB Systemic mastocytosis (SM) is a rare chronic myeloproliferative neoplasm with only limited epidemiologic data published so far. We aimed to analyze the clinical and molecular diagnostic features, and the prognosis and cumulative incidence of SM cases in a cohort of south-eastern Hungarian patients of 13 year follow up. In the period 2001–2013, 35 consecutive SM cases were diagnosed in our regional centre. Immunophenotype, KIT D816V mutation frequency and clinical characteristics, and the prognosis impact of clinical subtypes were tested and compared with published data. Indolent SM (ISM) was diagnosed in 14 patients, SM with an associated clonal hematologic non-mast cell lineage disease (SM-AHNMD) in 15 patients and aggressive SM (ASM) in 6 patients. The KIT D816V mutation was found in 11/14 (78 %) of the ISM cases, in 12/15 (80 %) of the SMAHNMD cases and in 5/6 (83 %) of the ASM cases. The life expectancy of ISM patients was better, whereas the SMAHNMD and ASM groups exhibited a reduced median survival. The cumulative incidence for 13 year of the SM was 0.27/10,000. We detected lower 13 year cumulative SM incidence than of published epidemiologic data due to in our analyses involved only those patients who had bone marrow biopsy and histopathologically confirmed SM. This clinical overview clearly showed that the clinical characteristics differ between ISM (UP, anaphylaxis and osteoporosis) and SMAHNMD/ ASM (cytopenia, eosinophilia and splenomegaly). C1 [Marton, Imelda] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Krenacs, Laszlo] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Bagdi, Eniko] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Bakos, Annamaria] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Demeter, Judit] Semmelweis University, 1st Department of Internal MedicineBudapest, Hungary. [Borbenyi, Zita] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. RP Marton, I (reprint author), University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, Szeged, Hungary. EM imeldamarton@gmail.com CR Brunning RD, McKenna RW, Rosai J, Parkin JL, Risdall R, 1983, Systemic mastocytosis. Extracutaneous manifestations. Am J Surg Pathol 7(5):425–438 Stevens EC, Rosenthal NS, 2001, Bone marrow mast cell morphologic features and hematopoietic dyspoiesis in systemic mast cell disease. Am J Clin Pathol 116(2):177–182., DOI 10.1309/Q2WJ- 46CL-YRFT-M5JF Gotlib J, Pardanani A, Akin C, Reiter A, George T, Hermine O, Kluin-Nelemans H, Hartmann K, SperrWR, Brockow K, Schwartz LB, Orfao A,Deangelo DJ,ArockM, Sotlar K, HornyHP, Metcalfe DD, Escribano L, Verstovsek S, Tefferi A, Valent P, 2013, International Working Group-Myeloproliferative Neoplasms Research and Treatment, IWG-MRT, & European Competence Network on Mastocytosis, ECNM, consensus response criteria in advanced systemic mastocytosis. Blood 121(13):2393–2401., DOI 10.1182/blood-2012-09-458521 Wadleigh M, Tefferi A, 2010, Classification and diagnosis of myeloproliferative neoplasms according to the 2008 World Health Organization criteria. Int J Hematol 91(2):174–179., DOI 10.1007/ s12185-010-0529-5 Valent P, 2013, Mastocytosis: a paradigmatic example of a rare disease with complex biology and pathology. Am J Cancer Res 3(2):159–172 van Doormaal JJ, Arends S, Brunekreeft KL, van der Wal VB, Sietsma J, van Voorst Vader PC, Oude Elberink JN, Kluin- Nelemans JC, van der Veer E, de Monchy JG, 2013, Prevalence of indolent systemic mastocytosis in a Dutch region. J Allergy Clin Immunol 131(5):1429–1431 e1421., DOI 10.1016/j.jaci.2012.10. 015 Cohen SS, Skovbo S, Vestergaard H, Kristensen T, Moller M, Bindslev-Jensen C, Fryzek JP, Broesby-Olsen S, 2014, Epidemiology of systemic mastocytosis in Denmark. Br J Haematol., DOI 10.1111/bjh.12916 Valent P, Arock M, Bonadonna P, Brockow K, Broesby-Olsen S, Escribano L, Gleixner KV, Grattan C, Hadzijusufovic E, Hagglund H, Hermine O, Horny HP, Kluin-Nelemans HC, Maurer M, Niedoszytko M, Nedoszytko B, Nilsson G, Oude-Elberink HN, Orfao A, Radia D, Reiter A, Siebenhaar F, Sotlar K, Sperr WR, Triggiani M, VanDoormaal JJ, Varkonyi J, Yavuz S, Hartmann K, 2012, European Competence Network on Mastocytosis, ECNM): 10-year jubilee, update, and future perspectives. Wien Klin Wochenschr 124(23–24):807–814., DOI 10.1007/s00508-012-0293-z Valent P, Akin C, Escribano L, Fodinger M, Hartmann K, Brockow K, Castells M, Sperr WR, Kluin-Nelemans HC, Hamdy NA, Lortholary O, Robyn J, van Doormaal J, Sotlar K, Hauswirth AW, Arock M, Hermine O, Hellmann A, Triggiani M, Niedoszytko M, Schwartz LB, Orfao A, Horny HP, Metcalfe DD, 2007, Standards and standardization in mastocytosis: consensus statements on diagnostics, treatment recommendations and response criteria. Eur J Clin Invest 37(6):435–453., DOI 10.1111/j.1365-2362.2007.01807.x Valent P, Akin C, ArockM, Brockow K, Butterfield JH, CarterMC, Castells M, Escribano L, Hartmann K, Lieberman P, Nedoszytko B, Orfao A, Schwartz LB, Sotlar K, Sperr WR, Triggiani M, Valenta R, Horny HP, Metcalfe DD, 2012, Definitions, criteria and global classification of mast cell disorders with special reference to mast cell activation syndromes: a consensus proposal. Int Arch Allergy Immunol 157(3):215–225., DOI 10.1159/000328760 Pardanani A, 2013, Systemic mastocytosis in adults: 2013 update on diagnosis, risk stratification, and management. Am J Hematol., DOI 10.1002/ajh.23459 Garcia-Montero AC, Jara-Acevedo M, Teodosio C, Sanchez ML, Nunez R, Prados A, Aldanondo I, Sanchez L, Dominguez M, Botana LM, Sanchez-Jimenez F, Sotlar K, Almeida J, Escribano L, Orfao A, 2006, KIT mutation in mast cells and other bone marrow hematopoietic cell lineages in systemic mast cell disorders: a prospective study of the Spanish Network on Mastocytosis, REMA, in a series of 113 patients. Blood 108(7):2366–2372., DOI 10.1182/blood-2006-04-015545 Soucie E, Hanssens K, Mercher T, Georgin-Lavialle S, Damaj G, Livideanu C, Chandesris MO, Acin Y, Letard S, de Sepulveda P, Hermine O, Bernard OA, Dubreuil P, 2012, In aggressive forms of mastocytosis, TET2 loss cooperates with c-KITD816V to transform mast cells. Blood 120(24):4846–4849., DOI 10.1182/blood-2011-12- 397588 Tay CM, Ong CW, Lee VK, Pang B, 2013, KIT gene mutation analysis in solid tumours: biology, clincial applications and trends in diagnostic reporting. Pathology 45(2):127–137., DOI 10.1097/ PAT.0b013e32835c7645 Beghini A, Peterlongo P, Ripamonti CB, Larizza L, Cairoli R, Morra E, Mecucci C, 2000, C-kit mutations in core binding factor leukemias. Blood 95(2):726–727 Lim KH, Tefferi A, Lasho TL, Finke C, Patnaik M, Butterfield JH, McClure RF, Li CY, Pardanani A, 2009, Systemic mastocytosis in 342 consecutive adults: survival studies and prognostic factors. Blood 113(23):5727–5736., DOI 10.1182/blood-2009-02-205237 Pardanani A, Tefferi A, 2010, Systemic mastocytosis in adults: a review on prognosis and treatment based on 342 Mayo Clinic patients and current literature. Curr Opin Hematol 17(2):125–132., DOI 10.1097/MOH.0b013e3283366c59 Verstovsek S, 2013, Advanced systemic mastocytosis: the impact of KIT mutations in diagnosis, treatment, and progression. Eur J Haematol 90(2):89–98., DOI 10.1111/ejh.12043 Escribano L, Alvarez-Twose I, Sanchez-Munoz L, Garcia-Montero A, Nunez R, Almeida J, Jara-Acevedo M, Teodosio C, Garcia- Cosio M, Bellas C, Orfao A, 2009, Prognosis in adult indolent systemic mastocytosis: a long-term study of the Spanish Network on Mastocytosis in a series of 145 patients. J Allergy Clin Immunol 124(3):514–521., DOI 10.1016/j.jaci.2009.05.003 Ustun C, DeRemer DL, Akin C, 2011, Tyrosine kinase inhibitors in the treatment of systemic mastocytosis. Leuk Res 35(9):1143– 1152., DOI 10.1016/j.leukres.2011.05.006 Akin C, Fumo G, Yavuz AS, Lipsky PE, Neckers L, Metcalfe DD, 2004, A novel form of mastocytosis associated with a transmembrane c-kit mutation and response to imatinib. Blood 103(8):3222– 3225., DOI 10.1182/blood-2003-11-3816 Hungarian Central Statistical Office-Population census, 2011). Regional data-Bacs-Kiskun county http://www.ksh.hu/ nepszamlalas/tables_regional_03; Regional data- Bekes county http://www.ksh.hu/nepszamlalas/tables_regional_04; Regional data - Csongrad county http://www.ksh.hu/nepszamlalas/tables_ regional_06 Valent P, Sperr WR, Akin C, 2010, How I treat patients with advanced systemic mastocytosis. Blood 116(26):5812–5817., DOI 10. 1182/blood-2010-08-292144 Lim KH, Pardanani A, Butterfield JH, Li CY, Tefferi A, 2009, Cytoreductive therapy in 108 adults with systemic mastocytosis: Outcome analysis and response prediction during treatment with interferon-alpha, hydroxyurea, imatinib mesylate or 2- chlorodeoxyadenosine. Am J Hematol 84(12):790–794., DOI 10. 1002/ajh.21561 Pardanani A, 2013, How I treat patients with indolent and smoldering mastocytosis, rare conditions but difficult tomanage). Blood 121(16):3085–3094., DOI 10.1182/blood-2013-01-453183 Valent P, 1996, Biology, classification and treatment of human mastocytosis. Wien Klin Wochenschr 108(13):385–397 Escribano L, Akin C, Castells M, Orfao A, Metcalfe DD, 2002, Mastocytosis: current concepts in diagnosis and treatment. Ann Hematol 81(12):677–690., DOI 10.1007/s00277-002-0575-z Valent P, Akin C, Sperr WR, Horny HP, Arock M, Lechner K, Bennett JM, Metcalfe DD, 2003, Diagnosis and treatment of systemic mastocytosis: state of the art. Br J Haematol 122(5):695–717 Schumacher JA, Elenitoba-Johnson KS, Lim MS, 2008, Detection of the c-kit D816V mutation in systemic mastocytosis by allelespecific PCR. J Clin Pathol 61(1):109–114., DOI 10.1136/jcp.2007. 047928 Akin C, 2005, Clonality and molecular pathogenesis of mastocytosis. Acta Haematol 114(1):61–69., DOI 10.1159/ 000085563 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 293 EP 299 DI 10.1007/s12253-015-9948-1 PG 7 ER PT J AU Mendes, J Goncalves, CA Alves, R Jorge, J Pires, A Ribeiro, A Sarmento-Ribeiro, BA AF Mendes, Jose Goncalves, Cristina Ana Alves, Raquel Jorge, Joana Pires, Ana Ribeiro, Ana Sarmento-Ribeiro, Bela Ana TI L744,832 and Everolimus Induce Cytotoxic and Cytostatic Effects in Non-Hodgkin Lymphoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Diffuse Large B-cell Lymphoma; Burkitt’s lymphoma; Everolimus; L744; 832; Apoptosis ID Diffuse Large B-cell Lymphoma; Burkitt’s lymphoma; Everolimus; L744; 832; Apoptosis AB Non-Hodgkin Lymphoma (NHL) constitutes a very heterogeneous group of diseases with different aggressiveness. Diffuse large B-cell lymphoma (DLBCL) and Burkitt’s lymphoma (BL) are two clinically aggressive lymphomas from the germinal center, very heterogeneous and with different genetic signatures. Several intracellular pathways are involved in lymphomagenesis, being BCR/PI3K/AKT/mTOR and RAS/RAF pathways the most frequently ones. In this context the therapeutic potential of a mTOR inhibitor – everolimus – and a RAS/RAF pathway inhibitor – L744,832 – was evaluated in two NHL cell lines. Farage and Raji cells were cultured in the absence and presence of several concentrations of everolimus and L744,832 in monotherapy and in combination with each other, as well as in association with the conventional chemotherapy drug vincristine. Our results show that everolimus and L744,832 induce antiproliferative and cytotoxic effect in a time-, dose-, and cell line-dependent manner, inducing cell death mainly by apoptosis. A potentiation effect was observed when the drugs were used in combination. In conclusion, the results suggest that everolimus and L744, 832, alone or in combination, could provide therapeutic benefits in these subtypes of NHL. C1 [Mendes, Jose] Faculty of Medicine University of Coimbra – FMUC, University Clinic of Hematology and Applied Molecular Biology, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal. [Goncalves, Cristina Ana] Faculty of Medicine University of Coimbra – FMUC, University Clinic of Hematology and Applied Molecular Biology, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal. [Alves, Raquel] Faculty of Medicine University of Coimbra – FMUC, University Clinic of Hematology and Applied Molecular Biology, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal. [Jorge, Joana] Faculty of Medicine University of Coimbra – FMUC, University Clinic of Hematology and Applied Molecular Biology, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal. [Pires, Ana] Faculty of Medicine University of Coimbra – FMUC, University Clinic of Hematology and Applied Molecular Biology, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal. [Ribeiro, Ana] Faculty of Medicine University of Coimbra – FMUC, University Clinic of Hematology and Applied Molecular Biology, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal. [Sarmento-Ribeiro, Bela Ana] Faculty of Medicine University of Coimbra – FMUC, University Clinic of Hematology and Applied Molecular Biology, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal. RP Sarmento-Ribeiro, BA (reprint author), Faculty of Medicine University of Coimbra – FMUC, University Clinic of Hematology and Applied Molecular Biology, 3000-548 Coimbra, Portugal. EM absarmento@fmed.uc.pt CR Shankland KR, Armitage JO, Hancock BW, 2012, Non-Hodgkin lymphoma. Lancet 380(9844):848–57 Stein H, Warnke RA, Chan WC, Jaffe ES, Chan JKC, Gatter KC, Campo E, 2008, Diffuse large B-cell lymphoma, not otherwise specified. In: Swerdlow S, Campo E, Harris N, Jaffe E, Pileri S, Stein H et al, eds, WHO classification of tumours of haematopoietic and lymphoid tissues. IARC, Lyon, pp 233–237 Schneider C, Pasqualucci L, Dalla-Favera R, 2011)Molecular pathogenesis of diffuse large B-cell lymphoma. Semin Diagn Pathol 28(2):167–177 Lenz G, Staudt LM, 2010, Aggressive lymphomas. N Engl J Med 362(15):1417–1429 Kenkre VP, Stock W, 2009, Burkitt lymphoma/leukemia: improving prognosis. Clin Lymphoma Myeloma 9(Suppl 3):S231–8 Bornkamm GW, 2009, Epstein-Barr virus and its role in the pathogenesis of Burkitt’s lymphoma: an unresolved issue. Semin Cancer Biol 19(6):351–65 Vose JM, Chiu BC, Cheson BD, Dancey J,Wright J, 2002, Update on epidemiology and therapeutics for non-Hodgkin’s lymphoma. Hematol Am Soc Hematol Educ Prog 2002:241–262 Hennessy BT, Hanrahan EO, Daly PA, 2004, Non-Hodgkin lymphoma: an update. Lancet Oncol 5(6):341–353 Castagna L, Magagnoli M, Demarco M, Santoro A, 2007, Lymphomas. Updat Cancer Ther 2(2):101–110 Michallet AS, Coiffier B, 2009, Recent developments in the treatment of aggressive non-Hodgkin lymphoma. Blood Rev 23(1):11– 23 Murawski N, Pfreundschuh M, 2010, New drugs for aggressive Bcell and T-cell lymphomas. Lancet Oncol 11(11):1074–85 Parcells BW, Ikeda AK, Simms-Waldrip T, Moore TB, Sakamoto KM, 2006, FMS-like tyrosine kinase 3 in normal hematopoiesis and acute myeloid leukemia. Stem Cells 24(5):1174–1184 Small D, 2006, FLT3 mutations: biology and treatment. Hematol Am Soc Hematol Educ Prog 2006:178–184 Morschhauser F, Bezombes C, Jardin F, 2010, Targeting molecular pathways. Education Program for the 15th Congress of European Hematology Association 4(1):118–123 Hachem A, Gartenhaus RB, 2005, Oncogenes as molecular targets in lymphoma. Blood 106(6):1911–1923 Easton JB, Houghton PJ, 2006, MTOR and cancer therapy. Oncogene 25(48):6436–6446 Polivka J Jr, Janku F, 2014)Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacol Ther 142(2):164–175 Niemann CU, Wiestner A, 2013, B-cell receptor signaling as a driver of lymphoma development and evolution. Semin Cancer Biol 23P:410–421 Bojarczuk K, Bobrowicz M, DwojakM, Miazek N, Zapala P, Bunes A, Siernicka M, Rozanska M, Winiarska M, 2015, B-cell receptor signaling in the pathogenesis of lymphoid malignancies. Blood Cells Mol Dis 55(3):255–65., DOI 10.1016/j.bcmd.2015.06.016 Costa CB, Casalta-Lopes J, Andrade C, Moreira D, Oliveira A, Goncalves AC, Alves V, Silva T, Dourado M, Nascimento-Costa JM, Sarmento-Ribeiro AB, 2012, Farnesyltransferase inhibitors:molecular evidence of therapeutic efficacy in acute lymphoblastic leukemia through cyclin D1 inhibition. Anticancer Res 32(3):831–838 Song SY,Meszoely IM, Coffey RJ, Pietenpol JA, Leach SD, 2000, K-Ras-independent effects of the farnesyl transferase inhibitor L744,832 on cyclin B1/Cdc2 kinase activity, G2/M cell cycle progression and apoptosis in human pancreatic ductal adenocarcinoma cells. Neoplasia 2(3):261–272 Adjei AA, Davis JN, Erlichman C, Svingen PA, Kaufmann SH, 2000, Comparison of potential markers of farnesyltransferase inhibition. Clin Cancer Res 6(6):2318–2325 Beck LA, Hosick TJ, SinenskyM(1990, Isoprenylation is required for the processing of the lamin a precursor. J Cell Biol 110(5):1489– 1499 Kilic F, Salas-Marco J, Garland J, SinenskyM(1997, Regulation of prelaminA endoprotease activity by prelaminA. FEBS Lett 414(1): 65–68 Brunner TB, Hahn SM, Gupta AK, Muschel RJ, McKenna WG, Bernhard EJ, 2003, Farnesyltransferase inhibitors: an overview of the results of preclinical and clinical investigations. Cancer Res 63(18):5656–5668 Appels NM, Beijnen JH, Schellens JH, 2005, Development of farnesyl transferase inhibitors: a review. Oncologist 10(8):565–578 Chan S, 2004, Targeting the mammalian target of rapamycin, mTOR): a new approach to treating cancer. Br J Cancer 91(8): 1420–1424 Wolpin BM, Hezel AF, Abrams T, Blaszkowsky LS, Meyerhardt JA, Chan JA, Enzinger PC, Allen B, Clark JW, Ryan DP, Fuchs CS, 2009, Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol 27(2):193–198 Frost P, Moatamed F, Hoang B, Shi Y, Gera J, Yan H, Frost F, Gibbons J, Lichtenstein A, 2004, In vivo antitumor effects of the mTOR inhibitor CCI-779 against human multiple myeloma cells in a xenograft model. Blood 104(13):4181–4187 Wendel HG, De Stanchina E, Fridman JS, Malina A, Ray S, Kogan S, Cordon-Cardo C, Pelletier J, Lowe SW, 2004, Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy. Nature 428(6980):332–337 Houghton PJ, 2010, Everolimus. Clin Cancer Res 16(5):1368– 1372 Scholzen T, Gerdes J, 2000, The Ki-67 protein: from the known and the unknown. J Cell Physiol 182(3):311–322 Yang Q, Guan KL, 2007, Expanding mTOR signaling. Cell Res 17(8):666–681 Kelly KR, Rowe JH, Padmanabhan S, Nawrocki ST, Carew JS, 2011)Mammalian target of rapamycin as a target in hematological malignancies. Target Oncol 6(1):53–61 Recher C, Dos Santos C, Demur C, Payrastre B, 2005, MTOR, a new therapeutic target in acute myeloid leukemia. Cell Cycle 4(11): 1540–1549 Sander S, Calado DP, Srinivasan L, Kochert K, Zhang B, Rosolowski M, Rodig SJ, Holzmann K, Stilgenbauer S, Siebert R, Bullinger L, Rajewsky K, 2012, Synergy between PI3K signaling and MYC in Burkitt lymphomagenesis. Cancer Cell 22:167– 179 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 301 EP 309 DI 10.1007/s12253-015-9998-4 PG 9 ER PT J AU Nabi, Sh Kahlon, P Bozorgnia, F Arshad, A Mikkelsen, T Donthireddy, V AF Nabi, Shahzaib Kahlon, Pushpinderdeep Bozorgnia, Farshid Arshad, Adeel Mikkelsen, Tom Donthireddy, Vijayalakshmi TI Predictors of Venous Thromboembolism in Patients with Glioblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE GBM; Venous thromboembolism; KPS scale; Bevacizumab ID GBM; Venous thromboembolism; KPS scale; Bevacizumab AB To evaluate different risk factors associated with development of venous thromboembolism (VTE) in patients with Glioblastoma (GBM). A retrospective chart review was performed to include patients diagnosed with GBM from 2001 to 2011. Cases (n = 162) were defined as patients with GBM who developed VTE after diagnosis of GBM. Controls (n = 840) were defined as patients with GBM with no history of VTE. Data was collected for multiple variables including age, gender, race, length of hospital stay after brain biopsy, total number of hospital admissions unrelated to VTE, Karnofsky Performance Status (KPS), use of Bevacizumab and any bleeding episodes. Patients with GBM who had VTE had poorer KPS scores, with the majority (57 %) being in between 40 and 70, as compared to the controls where majority (82 %) had better performance (KPS 80-100). For every one year increase in age, the odds of developing VTE increased by 3%(OR 1.03, 95%CI 1.02-1.04, p < 0.001) with the mean age being 61.8 ± 11.4 years. GBM patients who developed a VTE were found to have greater number of hospital admissions (OR 1.43, 95%CI 1.33-1.53, p < 0.001) and longer stays in hospital after GBM biopsy (OR 1.14, 95%CI 1.09-1.18, p < 0.001). Patients receiving Bevacizumab were more likely to develop VTE (OR 1.79, 95%CI 1.21-2.64, p < 0.001) and were more likely to have a bleed (OR 3.78, 95 % CI 2.70-5.30, p < 0.001). Patients with GBM are at a higher risk of developing VTE. The risk is higher in older patients who require multiple hospital admissions, longer duration of hospital stays related to GBM biopsy, and in patients with lower KPS scores. Bevacizumab use is related to a higher incidence of VTE as well as bleeds. This study suggests that a more aggressive strategy for VTE prophylaxis should be considered in GBM patients with risk factors for VTE. C1 [Nabi, Shahzaib] Henry Ford Health System / Wayne State University, Department of Internal Medicine, 2799 West Grand Boulevard, CFP 1, 48202 Detroit, MI, USA. [Kahlon, Pushpinderdeep] Henry Ford Health System / Wayne State University, Department of Internal Medicine, 2799 West Grand Boulevard, CFP 1, 48202 Detroit, MI, USA. [Bozorgnia, Farshid] Wayne State University, School of MedicineDetroit, MI, USA. [Arshad, Adeel] Weill Cornell University, Hamad Medical Corporation, Department of Internal MedicineDoha, Qatar. [Mikkelsen, Tom] Henry Ford Health System / Wayne State University, Hermelin Brain Tumor CenterDetroit, MI, USA. [Donthireddy, Vijayalakshmi] Henry Ford Health System / Wayne State University, Department of Hematology-OncologyDetroit, MI, USA. RP Nabi, Sh (reprint author), Henry Ford Health System / Wayne State University, Department of Internal Medicine, 48202 Detroit, USA. EM snabi1@hfhs.org CR Thaler J, Ay C, Kaider A, Reitter EM, Haselbock J, Mannhalter C, Zielinski C, Marosi C, Pabinger I, 2014, Biomarkers predictive of venous thromboembolism in patients with newly diagnosed high-grade gliomas. Neuro-Oncology 16(12):1645– 1651., DOI 10.1093/neuonc/nou106 Khorana AA, Francis CW, Culakova E, Kuderer NM, Lyman GH, 2007, Thromboembolism is a leading cause of death in cancer patients receiving outpatient chemotherapy. J Thromb Haemost 5(3):632–634., DOI 10.1111/j.1538-7836.2007.02374.x Magnus N, D'Asti E, Garnier D, Meehan B, Rak J, 2013, Brain neoplasms and coagulation. Semin Thromb Hemost 39(8):881– 895., DOI 10.1055/s-0033-1357483 Khorana AA, Kuderer NM, Culakova E, Lyman GH, Francis CW, 2008, Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood 111(10):4902–4907., DOI 10.1182/blood-2007-10-116327 Connolly GC, Khorana AA, Kuderer NM, Culakova E, Francis CW, Lyman GH, 2010, Leukocytosis, thrombosis and early mortality in cancer patients initiating chemotherapy. Thromb Res 126(2):113–118., DOI 10.1016/j.thromres.2010.05.012 Semrad TJ, O'Donnell R, Wun T, Chew H, Harvey D, Zhou H, White RH, 2007, Epidemiology of venous thromboembolism in 9489 patients with malignant glioma. J Neurosurg 106(4):601– 608., DOI 10.3171/jns.2007.106.4.601 Chaichana KL, Pendleton C, Jackson C, Martinez-Gutierrez JC, Diaz-Stransky A, Aguayo J, Olivi A, Weingart J, Gallia G, Lim M, Brem H, Quinones-Hinojosa A, 2013, Deep venous thrombosis and pulmonary embolisms in adult patients undergoing craniotomy for brain tumors. Neurol Res 35(2):206–211., DOI 10.1179/1743132812Y.0000000126 Seidel C, Hentschel B, Simon M, Schnell O, Heese O, Tatagiba M, Krex D, Reithmeier T, Kowoll A, Weller M, Wick W, 2013, A comprehensive analysis of vascular complications in 3,889 glioma patients from the German glioma network. J Neurol 260(3):847– 855., DOI 10.1007/s00415-012-6718-9 Jenkins EO, Schiff D, Mackman N, Key NS, 2010, Venous thromboembolism in malignant gliomas. J Thromb Haemost 8(2):221– 227., DOI 10.1111/j.1538-7836.2009.03690.x Lyman GH, Khorana AA, Falanga A, Clarke-Pearson D, Flowers C, Jahanzeb M, Kakkar A, Kuderer NM, Levine MN, Liebman H, Mendelson D, Raskob G, Somerfield MR, Thodiyil P, Trent D, Francis CW, American Society of Clinical O, 2007, American society of clinical oncology guideline: recommendations for venous thromboembolism prophylaxis and treatment in patients with cancer. J Clin Oncol : Off J Am Soc Clin Oncol 25(34):5490–5505., DOI 10.1200/JCO.2007.14.1283 Gallus AS, 1997, Prevention of post-operative deep leg vein thrombosis in patients with cancer. Thromb Haemost 78(1):126–132 Sartori MT, Della Puppa A, Ballin A, Campello E, Radu CM, Saggiorato G, d’Avella D, Scienza R, Cella G, Simioni P, 2013, Circulatingmicroparticles of glial origin and tissue factor bearing in high-grade glioma: a potential prothrombotic role. Thromb Haemost 110(2):378–385., DOI 10.1160/TH12-12-0957 Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, Hahn WC, Ligon KL, Louis DN, Brennan C, Chin L, DePinho RA, Cavenee WK, 2007, Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev 21(21):2683–2710., DOI 10.1101/gad.1596707 Sathornsumetee S, Reardon DA, Desjardins A, Quinn JA, Vredenburgh JJ, Rich JN, 2007, Molecularly targeted therapy for malignant glioma. Cancer 110(1):13–24., DOI 10.1002/cncr.22741 Fine HA, 2014, Bevacizumab in glioblastoma–still much to learn. N Engl JMed 370(8):764–765., DOI 10.1056/NEJMe1313309 Koukourakis GV, 2012, Has bevacizumab, avastin, given extra therapeutic gain in metastatic colorectal cancer and malignant brain gliomas? Systematic review answering this question. Recent Patents Inflamm Allergy Drug Discov 6(1):70–77 Kreisl TN, Kim L, Moore K, Duic P, Royce C, Stroud I, Garren N, Mackey M, Butman JA, Camphausen K, Park J, Albert PS, Fine HA, 2009, Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol : Off J Am Soc Clin Oncol 27(5):740–745., DOI 10.1200/JCO.2008.16.3055 Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE, Yung WK, Paleologos N, Nicholas MK, Jensen R, Vredenburgh J, Huang J, Zheng M, Cloughesy T, 2009, Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol : Off J Am Soc Clin Oncol 27(28): 4733–4740., DOI 10.1200/JCO.2008.19.8721 Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, Colman H, Chakravarti A, Pugh S, Won M, Jeraj R, Brown PD, Jaeckle KA, Schiff D, Stieber VW, Brachman DG, Werner-Wasik M, Tremont-Lukats IW, Sulman EP, Aldape KD, Curran Jr WJ, Mehta MP, 2014, A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med 370(8):699–708., DOI 10.1056/NEJMoa1308573 Chinot OL, Wick W, MasonW, Henriksson R, Saran F, Nishikawa R, Carpentier AF, Hoang-Xuan K, Kavan P, Cernea D, Brandes AA, Hilton M, Abrey L, Cloughesy T, 2014, Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med 370(8):709–722., DOI 10.1056/NEJMoa1308345 Simonetti G, Trevisan E, Silvani A, Gaviani P, Botturi A, Lamperti E, Beecher D, Bertero L, Bosa C, Salmaggi A, 2014, Safety of bevacizumab in patients with malignant gliomas: a systematic review. Neurol Sci 35(1):83–89., DOI 10.1007/s10072- 013-1583-6 Hapani S, Sher A, Chu D, Wu S, 2010, Increased risk of serious hemorrhage with bevacizumab in cancer patients: a meta-analysis. Oncology 79(1–2):27–38., DOI 10.1159/000314980 Narita Y, 2013, Drug review: safety and efficacy of bevacizumab for glioblastoma and other brain tumors. Jpn J Clin Oncol 43(6): 587–595., DOI 10.1093/jjco/hyt051 Wen PY, Schiff D, Kesari S, Drappatz J, Gigas DC, Doherty L, 2006, Medical management of patients with brain tumors. J Neuro-Oncol 80(3):313–332., DOI 10.1007/s11060-006- 9193-2 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 311 EP 316 DI 10.1007/s12253-015-0008-7 PG 6 ER PT J AU Burada, F Ciurea, EM Nicoli, R Streata, I Vilcea, DI Rogoveanu, I Ioana, M AF Burada, Florin Ciurea, Eugen Marius Nicoli, Raluca Streata, Ioana Vilcea, Dan Ionica Rogoveanu, Ion Ioana, Mihai TI ATG16L1 T300A Polymorphism is Correlated with Gastric Cancer Susceptibility SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Autophagy; ATG16L1 gene; Polymorphism; Gene expression ID Gastric cancer; Autophagy; ATG16L1 gene; Polymorphism; Gene expression AB Gastric cancer is a major leading cause of cancerrelated death in both sexes in Europe. The role of autophagy process in carcinogenesis remains unclear and there is increasing evidence that Helicobacter pylori is a key player in modulating autophagy in gastric carcinogenesis. The aim of this studywas to assess the potential association of ATG16L1 T300A polymorphismwith susceptibility of gastric cancer, and further to analyze the expression profile of ATG16L1 gene in paired tumoral and peritumoral gastric tissue. A total of 108 patients diagnosed with gastric cancer and 242 healthy controlswere enrolled.ATG16L1 T300A polymorphism was detected using TaqMan genotyping assay containing primers and specific probes for A and G allele, respectively. ATG16L1 mRNA level was evaluated in 34 paired tumoral and peritumoral tissues using qRT-PCR. We found a significant association for both carriers of AG (OR 0.52, 95 % CI: 0.30–0.91, p = 0.02) and GG genotype (OR 0.53, 95 % CI: 0.28–0.98, p = 0.043), these were at a lower risk for gastric cancer when compared with the wild-type AA genotype. The strongest association was observed in a dominant model, the carriers of G allele were protected against gastric cancer (OR 0.52, 95 % CI: 0.13–0.88, p = 0.013). In a stratified analyse, the association was limited to non-cardia type and intestinal type. ATG16L1 gene expression was detected in both tumor and peritumoral tissues, with the mRNA-ATG16L1 levels significantly higher in tumor sample. Our results suggest that ATG16L1 T300A polymorphismmay be associatedwith gastric carcinogenesis. C1 [Burada, Florin] University of Medicine and Pharmacy of Craiova, Research Center of Gastroenterology and Hepatology, 1 May street no 66Craiova, Romania. [Ciurea, Eugen Marius] University of Medicine and Pharmacy of Craiova, Research Center of Gastroenterology and Hepatology, 1 May street no 66Craiova, Romania. [Nicoli, Raluca] University of Medicine and Pharmacy of Craiova, Research Center of Gastroenterology and Hepatology, 1 May street no 66Craiova, Romania. [Streata, Ioana] University of Medicine and Pharmacy of Craiova, Research Center of Gastroenterology and Hepatology, 1 May street no 66Craiova, Romania. [Vilcea, Dan Ionica] University of Medicine and Pharmacy of Craiova, Department of Surgery, Petru Rares street no 2-4, 200349 Craiova, Romania. [Rogoveanu, Ion] University of Medicine and Pharmacy of Craiova, Research Center of Gastroenterology and Hepatology, 1 May street no 66Craiova, Romania. [Ioana, Mihai] University of Medicine and Pharmacy of Craiova, Research Center of Gastroenterology and Hepatology, 1 May street no 66Craiova, Romania. RP Burada, F (reprint author), University of Medicine and Pharmacy of Craiova, Research Center of Gastroenterology and Hepatology, Craiova, Romania. EM buradaflorin@gmail.com CR Arnold M, Karim-Kos HE, Coebergh JW, Byrnes G, Antilla A, Ferlay J, Renehan AG, Forman D, Soerjomataram I, 2015, Recent trends in incidence of five common cancers in 26 European countries since 1988: analysis of the European cancer observatory. Eur J Cancer 51(9):1164–1187., DOI 10.1016/j.ejca. 2013.09.002 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D, Bray F, 2013, Cancer incidence andmortality patterns in europe: estimates for 40 countries in 2012. Eur J Cancer 49(6):1374–1403., DOI 10.1016/j.ejca.2012.12. 027 Polk DB, Peek Jr RM, 2010, Helicobacter pylori: gastric cancer and beyond. Nat Rev Cancer 10(6):403–414., DOI 10.1038/nrc2857 Biological agents. Volume 100 B. A review of human carcinogens, 2012). IARCMonogr Eval Carcinog Risks Hum 100, Pt B):1–441 Sepulveda AR, 2013, Helicobacter, inflammation, and gastric cancer. Curr Pathobiol Rep 1(1):9–18., DOI 10.1007/ s40139-013-0009-8 Cid TP, Fernandez MC, Benito Martinez S, Jones NL, 2013, Pathogenesis of Helicobacter pylori infection. Helicobacter 18(Suppl 1):12–17., DOI 10.1111/hel.12076 Levine B, Kroemer G, 2008, Autophagy in the pathogenesis of disease. Cell 132(1):27–42., DOI 10.1016/j.cell.2007.12.018 Weidberg H, Shvets E, Elazar Z, 2011, Biogenesis and cargo selectivity of autophagosomes. Annu Rev Biochem 80:125–156., DOI 10.1146/annurev-biochem-052709-094552 Levine B, Mizushima N, Virgin HW, 2011, Autophagy in immunity and inflammation. Nature 469(7330):323–335., DOI 10.1038/ nature09782 Fujita N, Itoh T, Omori H, Fukuda M, Noda T, Yoshimori T, 2008, The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol Biol Cell 19(5):2092–2100., DOI 10.1091/mbc.E07-12-1257 Brest P, Corcelle EA, Cesaro A, Chargui A, Belaid A, Klionsky DJ, Vouret-Craviari V, Hebuterne X, Hofman P, Mograbi B, 2010, Autophagy and crohn’s disease: at the crossroads of infection, inflammation, immunity, and cancer. Curr Mol Med 10(5):486–502 Zheng H, Ji C, Li J, Jiang H, Ren M, Lu Q, Gu S, Mao Y, Xie Y, 2004, Cloning and analysis of human Apg16L. DNA Seq 15(4): 303–305 Cooney R, Baker J, Brain O, Danis B, Pichulik T, Allan P, Ferguson DJ, Campbell BJ, Jewell D, Simmons A, 2010, NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat Med 16(1):90–97., DOI 10.1038/nm. 2069 Hampe J, Franke A, Rosenstiel P, Till A, Teuber M, Huse K, Albrecht M, Mayr G, De La Vega FM, Briggs J, Gunther S, Prescott NJ, Onnie CM, Hasler R, Sipos B, Folsch UR, Lengauer T, Platzer M, Mathew CG, Krawczak M, Schreiber S, 2007, A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for crohn disease in ATG16L1. Nat Genet 39(2):207–211., DOI 10.1038/ng1954 Rioux JD, Xavier RJ, Taylor KD, Silverberg MS, Goyette P, Huett A, Green T, Kuballa P, Barmada MM, Datta LW, Shugart YY, Griffiths AM, Targan SR, Ippoliti AF, Bernard EJ, Mei L, Nicolae DL, Regueiro M, Schumm LP, Steinhart AH, Rotter JI, Duerr RH, Cho JH, Daly MJ, Brant SR, 2007, Genome-wide association study identifies new susceptibility loci for crohn disease and implicates autophagy in disease pathogenesis. Nat Genet 39(5):596–604., DOI 10.1038/ng2032 Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls, 2007). Nature 447, 7145):661– 678., DOI 10.1038/nature05911 Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, Mukherjee C, Shi Y, Gelinas C, Fan Y, Nelson DA, Jin S,White E, 2006, Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 10(1):51– 64., DOI 10.1016/j.ccr.2006.06.001 Greenfield LK, Jones NL, 2013, Modulation of autophagy by Helicobacter pylori and its role in gastric carcinogenesis. Trends Microbiol 21(11):602–612., DOI 10.1016/j.tim.2013.09.004 Terebiznik MR, Raju D, Vazquez CL, Torbricki K, Kulkarni R, Blanke SR, Yoshimori T, Colombo MI, Jones NL, 2009, Effect of Helicobacter pylori’s vacuolating cytotoxin on the autophagy pathway in gastric epithelial cells. Autophagy 5(3):370–379 Raju D, Hussey S, Ang M, Terebiznik MR, Sibony M, Galindo- Mata E, Gupta V, Blanke SR, Delgado A, Romero-Gallo J, Ramjeet MS, Mascarenhas H, Peek RM, Correa P, Streutker C, Hold G, Kunstmann E, Yoshimori T, Silverberg MS, Girardin SE, Philpott DJ, ElOmar E, Jones NL, 2012)Vacuolating cytotoxin and variants in Atg16L1 that disrupt autophagy promote Helicobacter pylori infection in humans. Gastroenterology 142(5):1160–1171., DOI 10. 1053/j.gastro.2012.01.043 Saitoh T, FujitaN, JangMH,Uematsu S, YangBG, Satoh T,Omori H, Noda T, Yamamoto N, Komatsu M, Tanaka K, Kawai T, Tsujimura T, Takeuchi O, Yoshimori T, Akira S, 2008, Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL- 1beta production. Nature 456(7219):264–268., DOI 10.1038/ nature07383 Huijbers A, Plantinga TS, Joosten LA, Aben KK, Gudmundsson J, den HeijerM, Kiemeney LA, NeteaMG, Hermus AR, Netea-Maier RT, 2012, The effect of the ATG16L1 Thr300Ala polymorphism on susceptibility and outcome of patients with epithelial cellderived thyroid carcinoma. Endocr Relat Cancer 19(3):L15–L18., DOI 10.1530/erc-11-0302 Lassen KG, Kuballa P, Conway KL, Patel KK, Becker CE, Peloquin JM, Villablanca EJ, Norman JM, Liu TC, Heath RJ, Becker ML, Fagbami L, Horn H, Mercer J, Yilmaz OH, Jaffe JD, Shamji AF, Bhan AK, Carr SA, Daly MJ, Virgin HW, Schreiber SL, Stappenbeck TS, Xavier RJ, 2014, Atg16L1 T300A variant decreases selective autophagy resulting in altered cytokine signaling and decreased antibacterial defense. Proc Natl Acad Sci U S A 111(21):7741–7746., DOI 10.1073/pnas.1407001111 Kuballa P, Huett A, Rioux JD, DalyMJ, Xavier RJ, 2008, Impaired autophagy of an intracellular pathogen induced by a crohn’s disease associated ATG16L1 variant. PLoS One 3(10):e3391., DOI 10.1371/ journal.pone.0003391 Grimm WA, Messer JS, Murphy SF, Nero T, Lodolce JP, Weber CR, Logsdon MF, Bartulis S, Sylvester BE, Springer A, Dougherty U, Niewold TB, Kupfer SS, Ellis N, Huo D, Bissonnette M, Boone DL, 2015, The Thr300Ala variant in ATG16L1 is associated with improved survival in human colorectal cancer and enhanced production of type I interferon. Gut., DOI 10.1136/gutjnl-2014-308735 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 317 EP 322 DI 10.1007/s12253-015-0006-9 PG 6 ER PT J AU Zidi, S Sghaier, I Gazouani, E Mezlini, A Yacoubi-Loueslati, B AF Zidi, Sabrina Sghaier, Ikram Gazouani, Ezzedine Mezlini, Amel Yacoubi-Loueslati, Besma TI Evaluation of Toll-Like Receptors 2/3/4/9 Gene Polymorphisms in Cervical Cancer Evolution SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Toll-like receptor; Cervical cancer; Gene polymorphism; FIGO stage ID Toll-like receptor; Cervical cancer; Gene polymorphism; FIGO stage AB Accumulative epidemiological evidence suggests that polymorphisms of Toll-like receptors signaling pathway elucidated the cellular and molecular mechanisms of human diseases whose gaining a primordial importance. The aim of our study is to identify the role of TLR 2 (−196 to −174 del), TLR 3 (1377 C>T), TLR 4 (Asp299Gly) and TLR 9 (G2848A) gene polymorphisms with the evolution of cervical cancer in Tunisian women. Blood samples were collected from histopathologically confirmed patients with cervical cancer and unrelated healthy female controls of similar ethnicity. Genotyping of the analyzed polymorphisms were done using Polymerase Chain Reaction and Restriction Fragment Length Polymorphism. For the TLR 2, Ins/Ins genotype is a protector factor [p = 0.006; OR: 0.35(0.16–0.73)] and the dominant genotype of TLR 3 increased the risk of CC in stage (III+ IV); C/C versuss C/T [p = 0.033; OR: 2.03(1.00–4.13)] and C/C versus C/T+T/T [p = 0.036; OR: 1.93(1.00–3.74)]. For TLR 4, the dominant genotype Asp/Asp is implicated in the occurrence of CC in stage (I+II) [p = 0.000; OR: 4.55(1.58– 13.06)], [p = 0.001; OR: 3.49(1.44–8.45)] and in stage (III+ IV) [p = 0.038; OR: 3.77(0.87–16.29)], [p = 0.007; OR: 5.21(1.65–16.46)] and the major allele Asp is a risk factor for the development of tumor in stage (I+II). The TLR2 Ins/ Del genotype is associated with tumor evolution to stage (III+ IV) [p = 0.003; OR: 3.00 (1.22–7.35)] and the genotypes Gly/ Gly and Asp/Gly+Gly/Gly and Gly allele of TLR 4 are implicated in tumor evolution to the advanced stages. Further, TLR 2, TLR 3, TLR 4 and TLR 9 gene polymorphisms are implicated in the modulation of CC risk due to tobacco usage and statue of menopause among cases. Our study suggests a relationship between the incidence of the TLR2, TLR 3, TLR 4 and TLR9 mutations and the clinical progression of CC according to the FIGO classification. However, future studies with different demographic and clinical characteristics in ethnically diverse populations may provide a more comprehensive involvement of innate immunity in cervical cancer etiology in women worldwide. C1 [Zidi, Sabrina] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 2092 El MANAR I, 1092 Tunis, Tunisia. [Sghaier, Ikram] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 2092 El MANAR I, 1092 Tunis, Tunisia. [Gazouani, Ezzedine] Military Hospital of Tunis, Laboratory of ImmunologyTunis, Tunisia. [Mezlini, Amel] Salah Azeiz Oncology InstituteTunis, Tunisia. [Yacoubi-Loueslati, Besma] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 2092 El MANAR I, 1092 Tunis, Tunisia. RP Zidi, S (reprint author), El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 1092 Tunis, Tunisia. EM ZIDISABRINA86@gmail.com CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127:2893–2917 Bernard HU, Burk RD, Chen Z, vanDoorslaer K, Hausen H, De Villiers EM, 2010, Classification of papillomaviruses, PVs, based on 189 PV types and proposal of taxonomic amendments. Virology 401:70–79 Giuliano AR, Harris R, Sedjo RL, Baldwin S, Roe D, Papenfuss MR, et al., 2002, Incidence, prevalence, and clearance of typespecific human papillomavirus infections: the Young Women’s Health Study. J Infect Dis 186:462–469 Akira S, Takeda K, Kaisho T, 2001, Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol 2: 675–680 DeCarlo CA, Rosa B, Jackson R, Niccoli S, Escott NG, Zehbe I, 2011, Toll-like receptor transcriptome in the HPV-positive cervical cancer microenvironment clinical. Dev Immunol 2012:785–825 Pandey S, Mittal B, Srivastava M, Singh S, Srivastava K, Lal P, Mittal RD, 2011, Evaluation of toll-like receptors 3, c 1377C/T, and 9, G2848A, gene polymorphisms in cervical cancer susceptibility. Mol Biol Rep 38:4715–4721 Pandey S, Mittal RD, Srivastava M, Srivastava K, Singh S, Srivastava S, Mittal B, 2009, Impact of toll-like receptors [TLR] 2, −196 to −174 del, and TLR 4, Asp299Gly, Thr399Ile, in cervical cancer susceptibility in North Indian women. Gynecol Oncol 114:501–505 Roszak A, Lianeri M, Sowin’ska A, Jagodzin’ski PP, 2012, Involvement of toll-like receptor 9 polymorphismin cervical cancer development. Mol Biol Rep 39:8425–8430 Chen X, Wang S, Liu L, Chen Z, Qiang F, Kan Y, Shen Y, Wu J, Shen H, Hu Z, 2012, A genetic variant in the promoter region of toll-like receptor 9 and cervical cancer susceptibility. DNA Cell Biol 31:766–771 Shepperd JH, 1995, FIGO staging of gynecologic cancers; cervical and vulva. Int J Gynecol Cancer 5:319–323 Fajardo-Dolci G, Solorio-Abreu J, Romero-Alvarez JC, Zavaleta- Villa B, Cerezo-Camacho O, Jimenez-Lucio R, Olivo-Diaz A, 2006, DQA1 and DQB1 association and nasal polyposis. Otolaryngol Head Neck Surg 135:243–247 Lorenz E, Frees KL, Schwartz DA, 2001, Determination of the TLR4 genotype using allele-specific PCR. Biotechniques 31:22–24 Tahara T, Arisawa T, Wang F, Shibata T, Nakamura M, Sakata M, Hirata I, Nakano H, 2007, Toll-like receptor 2–196 to 174del polymorphism influences the susceptibility of Japanese people to gastric cancer. Cancer Sci 98:1790–1794 Cheng PL, Eng HL, Chou MH, Chou MH, You HL, Lin TM, 2007, Genetic polymorphisms of viral infection-associated toll-like receptors in Chinese population. Transl Res 150: 311–318 De Giovine FS, Camp NJ, Cox A, Chaudhary AG, Sorrell JA, Crane A, Duff GW, 2000, Detection and population analysis of IL-1 and TNF gene polymorphisms. In: Balkwill F, ed, Cytokine molecular biology. Oxford University, Oxford, pp. 21–46 Zidi S, Verdi H, Yilmaz-Yalcin Y, Yazici AC, Gazouani E, Mezlini A, Atac FB, Yacoubi-Loueslati B, 2014, Involvement of toll-like receptors in cervical cancer susceptibility among Tunisian women. Bull Cancer 101:E31–EE5 Etem EO, Elyas H, Ozgocmen S, Yildirim A, Godekmerdan A, 2010, The investigation of toll-like receptor 3, 9 and 10 gene polymorphisms in Turkish rheumatoid arthritis patients. Rheumatol Int 31:1369–1374 He JF, Jia WH, Fan Q, Zhou XX, Qin HD, Shugart YY, Zeng YX, 2007, Genetic polymorphisms of TLR3 are associated with nasopharyngeal carcinoma risk in Cantonese population. Bio Med Cent Cancer 7:194 Salome GR, LauraM, Lucia G, Luis OG, Jose MC,Maria LL, Jose MGQ, Francisco JV, 2010, Study of TLR3, TLR4 and TLR9 in breast carcinomas and their associationwithmetastasis. BMC Cancer 10:665 El-Omar EM, Ng MT, Hold GL, 2008, Polymorphisms in toll-like receptor genes and risk of cancer. Oncogene 27:244–252 Jing JJ, Li M, Yuan Y, 2012, Toll-like receptor 4 Asp299Gly and Thr399Ile polymorphisms in cancer: a meta-analysis. Gene 499: 237–242 Noguchi E, Nishimura F, Fukai H, et al., 2004, An association study of asthma and total serum immunoglobin E levels for tolllike receptor polymorphisms in a Japanese population. Clin Exp Allergy 34:177–183., DOI 10.1111/j.1365-2222.2004.01839.x Berghofer B, Frommer T, Konig IR, et al., 2005, Common human toll-like receptor 9 polymorphisms and haplotypes: association with atopy and functional relevance. Clin Exp Allergy 35:1147–1154., DOI 10.1111/j.1365-2222.2005.02325.x DeCarlo CA, Rosa B, Jackson R, Niccoli S, Escott NG, Zehbe I, 2012, Toll-like receptor transcriptome in the hpv-positive cervical cancer microenvironment. Clin Dev Immunol 2012(ID 785825):9 Hindawi Publishing Corporation NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 323 EP 330 DI 10.1007/s12253-015-0009-6 PG 8 ER PT J AU Zhu, L Jing, S Wang, B Wu, K Shenglin, M Zhang, Sh AF Zhu, Lucheng Jing, Saisai Wang, Bing Wu, Kan Shenglin, Ma Zhang, Shirong TI Anti-PD-1/PD-L1 Therapy as a Promising Option for Non-Small Cell Lung Cancer: a Single arm Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PD-1; PD-L1; Non-small cell lung cancer; Checkpoint inhibitor; Meta-analysis ID PD-1; PD-L1; Non-small cell lung cancer; Checkpoint inhibitor; Meta-analysis AB Anti-PD-1/PD-L1 antibodies showed satisfactory efficacy in treating non-small-cell lung cancer. We conducted this meta-analysis to explore the advantage subtypes and best therapeutic modalities of Anti-PD-1/PD-L1 therapy on NSCLC. A quantitative meta-analysis was performed through a systematic search in PubMed, Web of Science, and the Cochrane Library. The pooled ORR, 6-month progressionfree survival rate (PFSR6m), and 1-year overall survival rate (OSR1y) were calculated and compared. 15 trials were included in this meta-analysis. Our analyses demonstrated the pooled ORR of 1st line and 2nd or more line anti-PD-1/PDL1 therapy were 36.5 % (21.9–51.0 %) and 17.0 % (14.3– 19.7 %), respectively. While the difference was significant (Z = 3.31, p < 0.001). The pooled ORR for non-squamous and squamous cell lung cancer were 18.5 % (16.0–21.1 %) and 17.9 % (14.4–21.5 %), respectively. The difference was not significant (Z = 0.27, p = 0.791). The pooled ORR for PDL1 positive and negative patients were 29.6 % (21.6–37.6 %) and 13.5 % (10.6–16.3 %), respectively. The difference was significant (Z = 4.39, p < 0.001). The PFSR6m for PD-L1 positive and negative NSCLC were 50.0 % (40.5–62.3 %) and 27.0 % (19.2–34.7 %). The difference was significant (Z = 3.72, p < 0.001). The OSR1y for PD-L1 positive and negative NSCLC were 66.8 % (44.8 %-88.9 %) and 54.0 % (32.6–75.3 %). The difference was not significant (Z = 0.77, p = 0.441). Anti-PD-1/PD-L1 antibody can serve as a promising treatment option for NSCLC. Patients with positive PD-L1 expression may benefit more from anti-PD-1/PD-L1 therapy. 1st-line anti-PD-1/ PD-L1 therapy can be chosen as the best modality. Squamous cell lung cancer also benefit from anti-PD-1/ PD-L1 therapy. C1 [Zhu, Lucheng] Hangzhou First People’s Hospital, No.261, Huansha Road, Shangcheng District, 310006 Hangzhou, China. [Jing, Saisai] Hangzhou First People’s Hospital, No.261, Huansha Road, Shangcheng District, 310006 Hangzhou, China. [Wang, Bing] Hangzhou First People’s Hospital, No.261, Huansha Road, Shangcheng District, 310006 Hangzhou, China. [Wu, Kan] Hangzhou First People’s Hospital, No.261, Huansha Road, Shangcheng District, 310006 Hangzhou, China. [Shenglin, Ma] Hangzhou First People’s Hospital, No.261, Huansha Road, Shangcheng District, 310006 Hangzhou, China. [Zhang, Shirong] Hangzhou First People’s Hospital, No.261, Huansha Road, Shangcheng District, 310006 Hangzhou, China. RP Shenglin, M (reprint author), Hangzhou First People’s Hospital, 310006 Hangzhou, China. EM mashenglin@outlook.com CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127(12):2893–2917 Topalian SL, Weiner GJ, Pardoll DM, 2011, Cancer immunotherapy comes of age. J Clin Oncol 29(36):4828–4836., DOI 10.1200/ jco.2011.38.0899 Drake CG, Jaffee E, Pardoll DM, 2006, Mechanisms of immune evasion by tumors. Adv Immunol 90:51–81., DOI 10.1016/s0065- 2776(06)90002-9 Mellman I, Coukos G, Dranoff G, 2011, Cancer immunotherapy comes of age. Nature 480(7378):480–489., DOI 10.1038/ nature10673 Reck M, Bondarenko I, Luft A, Serwatowski P, Barlesi F, Chacko R, Sebastian M, Lu H, Cuillerot JM, Lynch TJ, 2013, Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial. Ann Oncol 24(1):75– 83., DOI 10.1093/annonc/mds213 Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI,Wolchok JD,Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, UrbaWJ, 2010, Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363(8):711– 723., DOI 10.1056/NEJMoa1003466 Ribas A, Kefford R, Marshall MA, Punt CJA, Haanen JB, Marmol M,Garbe C, Gogas H, Schachter J, LinetteG, Lorigan P, KendraKL, Maio M, Trefzer U, Smylie M, McArthur GA, Dreno B, Nathan PD, Mackiewicz J, Kirkwood JM, Gomez-Navarro J, Huang B, Pavlov D, Hauschild A, 2013, Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol, Meeting Abstracts, 31, 5):616– 622., DOI 10.1200/jco.2012.44.6112 Zatloukal P, Heo DS, Park K, Kang J, Butts C, Bradford D, Graziano S, Huang B, Healey D, 2009, Randomized phase II clinical trial comparing tremelimumab, CP-675,206, with best supportive care, BSC, following first-line platinum-based therapy in patients, pts, with advanced non-small cell lung cancer, NSCLC). J Clin Oncol, Meeting Abstracts, 27, 15S):8071 Attia P, Phan GQ, Maker AV, Robinson MR, Quezado MM, Yang JC, Sherry RM, Topalian SL, Kammula US, Royal RE, Restifo NP, Haworth LR, Levy C, Mavroukakis SA, Nichol G, Yellin MJ, Rosenberg SA, 2005, Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anticytotoxic T-lymphocyte antigen-4. J Clin Oncol 23(25): 6043–6053., DOI 10.1200/jco.2005.06.205 IshidaY, AgataY, Shibahara K, Honjo T, 1992, Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. Embo J 11(11):3887–3895 Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, Bowman MR, Carreno BM, Collins M, Wood CR, Honjo T, 2000, Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 192(7): 1027–1034 Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, Albacker LA, Koulmanda M, Freeman GJ, Sayegh MH, Sharpe AH, 2006, Tissue expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med 203(4):883–895., DOI 10.1084/jem.20051776 Mu CY, Huang JA, Chen Y, Chen C, Zhang XG, 2011, High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation. Med Oncol 28(3):682– 688., DOI 10.1007/s12032-010-9515-2 Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, Martins R, Eaton K, Chen S, Salay TM, Alaparthy S, Grosso JF, Korman AJ, Parker SM, Agrawal S, Goldberg SM, Pardoll DM, Gupta A, Wigginton JM, 2012, Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366(26):2455–2465., DOI 10.1056/NEJMoa1200694 Rizvi NA, Mazieres J, Planchard D, Stinchcombe TE, Dy GK, Antonia SJ, Horn L, Lena H, Minenza E, Mennecier B, Otterson GA, Campos LT, GandaraDR, Levy BP,Nair SG, Zalcman G,Wolf J, Souquet PJ, Baldini E, Cappuzzo F, Chouaid C, Dowlati A, Sanborn R, Lopez-Chavez A, Grohe C, Huber RM, Harbison CT, Baudelet C, Lestini BJ, Ramalingam SS, 2015, Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer, CheckMate 063): a phase 2, single-arm trial. Lancet Oncol 16(3):257–265., DOI 10.1016/s1470-2045(15)70054-9 Wells G, Shea B, O’Connell D, Petersen J, Welch V, Losos M, Tugwell P, 2011, The Newcastle-Ottawa Scale, NOS, for assessing the quality of nonrandomized studies in meta-analyses. Department of Epidemiology and Community Medicine, University of Ottawa, Canada. http://www.ohrica/programs/clinical_epidemiology/ oxfordasp Spira AI, Park K, Mazieres J, Vansteenkiste JF, Rittmeyer A, Ballinger M, Waterkamp D, Kowanetz M, Mokatrin A, Fehrenbacher L Efficacy, safety and predictive biomarker results from a randomized phase II study comparing MPDL3280A vs docetaxel in 2 L/3 L NSCLC, POPLAR). In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p 8010 Spigel DR, Chaft JE, Gettinger SN, Chao BH, Dirix LY, Schmid P, Chow LQM, Chappey C, Kowanetz M, Sandler A Clinical activity and safety from a phase II study, FIR, ofMPDL3280A, anti-PDL1, in PD-L1-selected patients with non-small cell lung cancer, NSCLC). In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p 8028 Paz-Ares L, Horn L, Borghaei H, Spigel DR, Steins M, Ready N, Chow LQM, Vokes EE, Felip E, Holgado E Phase III, randomized trial, CheckMate 057, of nivolumab, NIVO, versus docetaxel, DOC, in advanced non-squamous cell, non-SQ, non-small cell lung cancer, NSCLC). In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p LBA109 Patnaik A, Socinski MA, Gubens MA, Gandhi L, Stevenson J, Bachman RD, Bourque J, Yang Ge J, Im E, Gadgeel SM Phase 1 study of pembrolizumab, pembro;MK-3475, plus ipilimumab, IPI, as second-line therapy for advanced non-small cell lung cancer, NSCLC): KEYNOTE-021 cohort D. In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p 8011 Papadimitrakopoulou V, Patnaik A, Borghaei H, Stevenson J, Gandhi L, Gubens MA, Yang JC-H, Sequist LV, Yang Ge J, Bourque J Pembrolizumab, pembro; MK-3475, plus platinum doublet chemotherapy, PDC, as front-line therapy for advanced non-small cell lung cancer, NSCLC): KEYNOTE- 021 Cohorts A and C. In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p 8031 Nishio M, Hida T, Nakagawa K, Sakai H, Nogami N, Atagi S, Takahashi T, Nokihara H, Saka H, Takenoyama M Phase II studies of nivolumab, anti-PD-1, BMS-936558, ONO-4538, in patients with advanced squamous, sq, or nonsquamous, non-sq, nonsmall cell lung cancer, NSCLC). In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p 8027 Liu SV, Powderly JD, Camidge DR, Ready N, Heist RS, Hodi FS, Giaccone G, Liu B, Wallin J, Funke RP Safety and efficacy of MPDL3280A, anti-PDL1, in combination with platinum-based doublet chemotherapy in patients with advanced non-small cell lung cancer, NSCLC). In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p 8030 Horn L, Spigel DR, Gettinger SN, Antonia SJ, Gordon MS, Herbst RS, Sequist LV, Chappey C, Kowanetz M, Sandler A Clinical activity, safety and predictive biomarkers of the engineered antibody MPDL3280A, anti-PDL1, in non-small cell lung cancer, NSCLC): update from a phase Ia study. In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p 8029 Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, RizviNA, Powderly JD, Heist RS, Carvajal RD, Jackman DM, Sequist LV, Smith DC, Leming P, Carbone DP, Pinder-Schenck MC, Topalian SL, Hodi FS, Sosman JA, Sznol M, McDermott DF, Pardoll DM, Sankar V, Ahlers CM, Salvati M, Wigginton JM, Hellmann MD, Kollia GD, Gupta AK, Brahmer JR, 2015, Overall survival and long-term safety of nivolumab, anti-programmed death 1 antibody, BMS-936558, ONO-4538, in patients with previously treated advanced Non-small-cell lung cancer. J Clin Oncol., DOI 10.1200/jco. 2014.58.3708 Gettinger SN, Hellmann MD, Shepherd FA, Antonia SJ, Brahmer JR, Chow LQM, Goldman JW, Juergens RA, Borghaei H, Ready N First-line monotherapy with nivolumab, NIVO; anti-programmed death-1 [PD-1], in advanced non-small cell lung cancer, NSCLC): Safety, efficacy and correlation of outcomes with PD-1 ligand, PDL1, expression. In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p 8025 Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, GubensM, Horn L, Carcereny E, Ahn MJ, Felip E, Lee JS, Hellmann MD, Hamid O, Goldman JW, Soria JC, Dolled-Filhart M, Rutledge RZ, Zhang J, Lunceford JK, Rangwala R, Lubiniecki GM, Roach C, Emancipator K, Gandhi L, 2015, Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 372(21):2018–2028., DOI 10.1056/NEJMoa1501824 Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, Waterhouse D, Ready N, Gainor J, Aren Frontera O, Havel L, Steins M, Garassino MC, Aerts JG, Domine M, Paz-Ares L, Reck M, Baudelet C, Harbison CT, Lestini B, Spigel DR, 2015, Nivolumab versus docetaxel in advanced squamous-cell Non-small-cell lung cancer. N Engl J Med., DOI 10.1056/NEJMoa1504627 Bauer TM, McCleod M, Chandler JC, Blumenschein GR, Schwartzberg LS, Burris H, Waterhouse D, Jotte RM, Hussein M, Spigel DR An ongoing phase IIIb/IV safety trial of nivolumab, NIVO, in patients, pts, with advanced or metastatic non-small-cell lung cancer, NSCLC, who progressed after receiving 1 or more prior systemic regimens. In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p 3013 Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, Hassel JC, Rutkowski P, McNeil C, Kalinka-Warzocha E, Savage KJ, Hernberg MM, Lebbe C, Charles J, Mihalcioiu C, Chiarion- Sileni V, Mauch C, Cognetti F, Arance A, Schmidt H, Schadendorf D, Gogas H, Lundgren-Eriksson L, Horak C, Sharkey B, Waxman IM, Atkinson V, Ascierto PA, 2015, Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 372(4): 320–330., DOI 10.1056/NEJMoa1412082 NCCN, 2015, NCCN Clinical Practice Guideline in Oncology: Non-Small Cell Lung Cancer, Version 6.2015). http://www. nccnorg/professionals/physician_gls/f_guidelinesasp#nscl Wang A,Wang HY, Liu Y, Zhao MC, Zhang HJ, Lu ZY, Fang YC, Chen XF, LiuGT, 2015, The prognostic value of PD-L1 expression for non-small cell lung cancer patients: a meta-analysis. Eur J Surg Oncol 41(4):450–456., DOI 10.1016/j.ejso.2015.01.020 Zhang Y, Kang S, Shen J, He J, Jiang L,Wang W, Guo Z, Peng G, Chen G, He J, Liang W, 2015, Prognostic significance of programmed cell death 1, PD-1, or PD-1 ligand 1, PD-L1, expression in epithelial-originated cancer: a meta-analysis. Medicine 94(6): e515., DOI 10.1097/md.0000000000000515 Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M, 2012, Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366(26):2443–2454., DOI 10.1056/NEJMoa1200690 Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, Chen L, Pardoll DM, Topalian SL, Anders RA, 2014, Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 20(19):5064–5074., DOI 10.1158/1078-0432.ccr-13-3271 Jia M, Feng W, Kang S, Zhang Y, Shen J, He J, Jiang L, Wang W, Guo Z, Peng G, Chen G, He J, Liang W, 2015, Evaluation of the efficacy and safety of anti-PD-1 and anti-PD-L1 antibody in the treatment of non-small cell lung cancer, NSCLC): a meta-analysis. J Thorac Dis 7(3):455–461., DOI 10.3978/j.issn.2072-1439.2015.02.06 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 331 EP 339 DI 10.1007/s12253-015-0011-z PG 9 ER PT J AU Mallak, JA Abbaszadegan, RM Khorasanizadeh, NP Forghanifard, MM AF Mallak, Javdani Afsaneh Abbaszadegan, Reza Mohammad Khorasanizadeh, Naeemi Pegah Forghanifard, Mahdi Mohammad TI Contribution of EVX1 in Aggressiveness of Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EVX1; BMP signaling pathway; Epithelial mesenchymal transition; Escc ID EVX1; BMP signaling pathway; Epithelial mesenchymal transition; Escc AB Homeobox genes play an overruling role in the regional cell fate determination during development. EVX1 is known as a new target gene of BMP signaling pathway, a group of morphogens which are making the largest subset within the transformation growth factor beta (TGF-β) superfamily. In this study, we aimed to enlighten the expression level of EVX1 in esophageal squamous cell carcinoma (ESCC) and to disclose its apparent roles in maintenance and progression of the disease. The expression level of EVX1 was analyzed in fresh tumoral tissues in comparison with distant tumor-free tissues of 50 ESCC patients using relative comparative real-time PCR. The importance of EVX1 in development and cancer was also reviewed. EVX1 was underexpressed in 70 % of tumor samples. There was a significant correlation between down-regulation of EVX1 and lymph node metastasis of tumor cells (p = 0.027). Furthermore, EVX1 underexpression was significantly correlated with depth of tumor cell invasion (P = 0.037). To the best of our knowledge, this is the first report highlighting EVX1 expression in ESCC to date. The clinicopathological relevance of EVX1 mRNA expression in ESCC targeted this gene as a new independent molecular marker for advanced tumor, which determine the characteristics and behavior of aggressive ESCC. C1 [Mallak, Javdani Afsaneh] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human GeneticsMashhad, Iran. [Abbaszadegan, Reza Mohammad] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human GeneticsMashhad, Iran. [Khorasanizadeh, Naeemi Pegah] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human GeneticsMashhad, Iran. [Forghanifard, Mahdi Mohammad] Islamic Azad University, Damghan Branch, Department of Biology, Cheshmeh-Ali Boulevard, Sa’dei SquareDamghan, Iran. RP Forghanifard, MM (reprint author), Islamic Azad University, Damghan Branch, Department of Biology, Damghan, Iran. EM forghanifard@gmail.com CR Ruiz i Altaba A, Choi T, Melton DA, 1991, Expression of the Xhox3 homeobox protein in xenopus embryos: blocking its early function suggests the requirement of Xhox3 for normal posterior development. Develop Growth Differ 33:651–669 Ruiz i Altaba A, Melton DA, 1989a, Bimodal and graded expression of the xenopus homeobox gene Xhox3 during embryonic development. Development 106:173–183 Ruiz i Altaba A, Melton DA, 1989b, Involvement of the xenopus homeobox gene Xhox3 in pattern formation along the anterior– posterior axis. Cell 57:317–326 Briata P, Van De Werken R, Airoldi I, Ilengo C, Di Blas E, Boncinelli E, Corte G, 1995, Transcriptional repression by the human homeobox protein EVX1 in transfected mammalian cells. J Biophys Chem 270:27695–27701., DOI 10.1074/ jbc.270.46.27695 Hui X, Long-jiang L, Fang X, Bo H, Zhuang Z, Jiang W, 2012, Differentially expressed homeobox genes in salivary adenoid cystic carcinoma versus normal salivary gland tissue. Life Sci J 9(2):292– 296 Guo X, Wang XF, 2009, Signaling cross-talk between TGF-beta/ BMP and other pathways. Cell Res 19(1):71–88., DOI 10.1038/cr. 2008.302 Blanco Calvo M, Bolos Fernandez V, Medina Villaamil V, Aparicio Gallego G, Diaz Prado S, Grande Pulido E, 2009, Biology of BMP signalling and cancer. Clin Transl Oncol 11(3):126–137., DOI 10. 1007/S12094-009-0328-8 Zhang G, Mai R, Huang B, 2010, ADH1B Arg47His polymorphism is associated with esophageal cancer risk in high-incidence Asian population: evidence from a meta-analysis. PLoS One 5(10): e13679., DOI 10.1371/journal.pone.0013679 Baldwin, K., 2015). Esophageal Cancer. Retrieved from http:// emedicine.medscape.com/article/277930-overview Reya T, Clevers H, 2005, Wnt signalling in stem cells and cancer. Nature 434(7035):843–850., DOI 10.1038/nature03319 Taipale J, Beachy PA, 2001, The hedgehog and Wnt signalling pathways in cancer. Nature 411(6835):349–354., DOI 10.1038/ 35077219 Grier DG, Thompson A, Kwasniewska A, McGonigle GJ, Halliday HL, Lappin TR, 2005, The pathophysiology of HOX genes and their role in cancer. J Pathol 205(2):154–171., DOI 10.1002/path. 1710 Zhai Y, Kuick R, Nan B, Ota I, Weiss SJ, Trimble CL, Fearon ER, Cho KR, 2007, Gene expression analysis of preinvasive and invasive cervical squamous cell carcinomas identifies HOXC10 as a key mediator of invasion. Cancer Res 67(21):10163–10172., DOI 10. 1158/0008-5472.CAN-07-2056 Sobin LH, Gospodarowicz MK,Wittekind C, 2011, TNM classification of malignant tumours. Wiley, London Garcia-Fernandez J, 2005, The genesis and evolution of homeobox gene clusters. Nat Rev Genet 6(12):881–892., DOI 10.1038/nrg1723 Rauch T, Wang Z, Zhang X, Zhong X, Wu X, Lau SK, Kernstine KH, Riggs AD, Pfeifer GP, 2007, Homeobox gene methylation in lung cancer studied by genome-wide analysis with a microarraybased methylated CpG island recovery assay. Proc Natl Acad Sci U S A 104(13):5527–5532., DOI 10.1073/pnas.0701059104 Osada S, Ohmori SY, Taira M, 2003, XMAN1, an inner nuclear membrane protein, antagonizes BMP signaling by interacting with Smad1 in xenopus embryos. Development 130(9):1783–1794., DOI 10.1242/dev.00401 Takebayashi-Suzuki K, Funami J, Tokumori D, Saito A, Watabe T, Miyazono K, Kanda A, Suzuki A, 2003, Interplay between the tumor suppressor p53 and TGF beta signaling shapes embryonic body axes in xenopus. Development 130(17):3929–3939., DOI 10. 1242/dev.00615 Spyropoulos DD, Capecchi MR, 1994, Targeted disruption of the even-skipped gene, evx1, causes early postimplantation lethality of the mouse conceptus. Genes Dev 8(16):1949–1961., DOI 10.1101/ gad.8.16.1949 Alev C, Wu Y, Kasukawa T, Jakt LM, Ueda HR, Sheng G, 2010, Transcriptomic landscape of the primitive streak. Development 137, 17):2863–2874., DOI 10.1242/dev.053462 Bastian H, Gruss P, 1990, A murine even-skipped homologue, Evx 1, is expressed during early embryogenesis and neurogenesis in a biphasic manner. EMBO J 9(6):1839–1852 Dush MK, Martin GR, 1992, Analysis of mouse Evx genes: Evx-1 displays graded expression in the primitive streak. Dev Biol 151(1): 273–287., DOI 10.1016/0168-9525(92)90245-Y Fuchtbauer EM, 1995, Expression of M-twist during postimplantation development of the mouse. Dev Dyn 204(3):316–322., DOI 10.1002/aja.1002040309 Wolf C, Thisse C, Stoetzel C, Thisse B, Gerlinger P, Perrin-Schmitt F, 1991, The M-twist gene of Mus is expressed in subsets of mesodermal cells and is closely related to the xenopus X-twi and the drosophila twist genes. Dev Biol 143(2):363–373., DOI 10.1016/ 0012-1606(91)90086-I Kanai-Azuma M, Kanai Y, Gad JM, Tajima Y, Taya C, Kurohmaru M, Sanai Y, Yonekawa H, Yazaki K, Tam PP, Hayashi Y, 2002, Depletion of definitive gut endoderm in Sox17-null mutant mice. Development 129(10):2367–2379 Yasunaga M, Tada S, Torikai-Nishikawa S, Nakano Y, Okada M, Jakt LM, Nishikawa S, Chiba T, Era T, Nishikawa S, 2005, Induction and monitoring of definitive and visceral endoderm differentiation of mouse ES cells. Nat Biotechnol 23(12):1542–1550., DOI 10.1038/nbt1167 Jones CM, Dale L, Hogan BL, Wright CV, Smith JC, 1996, Bone morphogenetic protein-4, BMP-4, acts during gastrula stages to cause ventralization of xenopus embryos. Development 122(5): 1545–1554 Nostro MC, Cheng X, Keller GM, Gadue P, 2008, Wnt, activin, and BMP signaling regulate distinct stages in the developmental pathway from embryonic stem cells to blood. Cell Stem Cell 2(1): 60–71., DOI 10.1016/j.stem.2007.10.011 Kalisz M, Winzi M, Bisgaard HC, Serup P, 2012, Even-skipped homeobox 1 controls human ES cell differentiation by directly repressing goosecoid expression. Dev Biol 362:94–103., DOI 10. 1016/j.ydbio.2011.11.017 Yasuo H, Lemaire P, 2001, Role of goosecoid, xnot and Wnt antagonists in the maintenance of the notochord genetic programme in xenopus gastrulae. Development 128(19):3783–3793 Luu O, Nagel M, Wacker S, Lemaire P, Winklbauer R, 2008, Control of gastrula cell motility by the goosecoid/mix.1/siamois network: basic patterns and paradoxical effects. Dev Dyn 237(5): 1307–1320., DOI 10.1002/dvdy.21522 Hartwell KA, Muir B, Reinhardt F, Carpenter AE, Sgroi DC, Weinberg RA, 2006, The Spemann organizer gene, goosecoid, promotes tumor metastasis. Proc Natl Acad Sci U S A 103(50): 18969–18974., DOI 10.1073/pnas.0608636103 Forghanifard MM, Moaven O, Farshchian M, Montazer M, Raeisossadati R, Abdollahi A, Moghbeli M, Nejadsattari T, Parivar K, Abbaszadegan MR, 2012, Expression analysis elucidates the roles ofMAML1 and Twist1 in esophageal squamous cell carcinoma aggressiveness and metastasis. Ann Surg Oncol 19(3): 743–749., DOI 10.1245/s10434-011-2074-8 Ansieau S, Morel AP, Hinkal G, Bastid J, Puisieux A, 2010, TWISTing an embryonic transcription factor into an oncoprotein. Oncogene 29(22):3173–3184., DOI 10.1038/onc.2010.92 Christiansen JJ, Rajasekaran AK, 2006, Reassessing epithelial to mesenchymal transition as a prerequisite for carcinoma invasion and metastasis. Cancer Res 66(17):8319–8326., DOI 10.1158/0008- 5472.CAN-06-0410 Klymkowsky MW, Savagner P, 2009, Epithelial - mesenchymal transition: a cancer researcher’s conceptual friend and foe. Am J Pathol 174(5):1588–1593., DOI 10.2353/ ajpath.2009.080545 Yang J,Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, Savagner P, Gitelman I, Richardson A, Weinberg RA, 2004, Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117(7):927–939., DOI 10.1016/j.cell.2004. 06.006 Sasaki K, Natsugoe S, Ishigami S, Matsumoto M, Okumura H, Setoyama T, Uchikado Y, Kita Y, Tamotsu K, Sakamoto A, Owaki T, Aikou T, 2009, Significance of twist expression and its association with E-cadherin in esophageal squamous cell carcinoma. J Exp Clin Cancer Res 28:158., DOI 10.1186/ 1756-9966-28-158 Yuen HF, Chan YP, Wong ML, Kwok WK, Chan KK, Lee PY, Srivastava G, Law SY, Wong YC, Wang X, Chan KW, 2007, Upregulation of twist in oesophageal squamous cell carcinoma is associated with neoplastic transformation and distant metastasis. J Clin Pathol 60(5):510–514., DOI 10. 1136/jcp.2006.039099 Du YC, Oshima H, Oguma K, Kitamura T, Itadani H, Fujimura T, Piao YS, Yoshimoto T, Minamoto T, Kotani H, Taketo MM, Oshima M, 2009, Induction and down-regulation of Sox17 and its possible roles during the course of gastrointestinal tumorigenesis. Gastroenterology 137(4):1346–1357., DOI 10. 1053/j.gastro.2009.06.041 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 341 EP 347 DI 10.1007/s12253-015-0005-x PG 7 ER PT J AU Helpap, B Ringli, D Tonhauser, J Poser, I Breul, J Gevensleben, H Seifert, HH AF Helpap, Burkhard Ringli, Daniel Tonhauser, Jens Poser, Immanuel Breul, Jurgen Gevensleben, Heidrun Seifert, Hans-Helge TI The Significance of Accurate Determination of Gleason Score for Therapeutic Options and Prognosis of Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate; Carcinoma; Gleason and combigrading; Prognosis; PSA progress; Survival ID Prostate; Carcinoma; Gleason and combigrading; Prognosis; PSA progress; Survival AB The Gleason score (GS) to date remains one of the most reliable prognostic predictors in prostate cancer (PCa). However, the majority of studies supporting its prognostic relevance were performed prior to its modification by the International Society of Urological Pathology (ISUP) in 2005. Furthermore, the combination of Gleason grading and nuclear/nucleolar subgrading (Helpap score) has been shown to essentially improve grading concordance between biopsy and radical prostatectomy (RP) specimens. This prompted us to investigate the modified GS and combigrading (Gleason/Helpap score) in association with clinicopathological features, biochemical recurrence (BCR), and survival. Core needle biopsies and corresponding RP specimens from 580 patients diagnosed with PCa between 2005 and 2010 were evaluated. According to the modified GS, the comparison between biopsy and RP samples resulted in an upgrading from GS 6 to GS 7a and GS 7b in 65%and 19 %, respectively. Combigrading further resulted in an upgrading from low grade (GS 6/2a) to intermediate grade PCa (GS 6/2b) in 11.1 % and from intermediate grade (GS 6/2b) to high grade PCa (GS 7b/2b) in 22.6 %. Overall, well-differentiated PCa (GS 6/2a) was detected in 2.8 % of RP specimens, while intermediate grade (GS 6/2b and GS 7a/2b) and high grade cancers (≥ GS 7b) accounted for 39.5 % and 57.4 % of cases, respectively. At a mean follow-up of 3.9 years, BCR was observed in 17.6 % of patients with intermediate (9.8 %) or high grade PCa (30.2 %), while PSA relapse did not occur in GS 6/2a PCa. In conclusion, adding nuclear/nucleolar subgrading to the modified GS allowed for a more accurate distinction between low and intermediate grade PCa, therefore offering a valuable tool for the identification of patients eligible for active surveillance (AS). C1 [Helpap, Burkhard] Hegau-Bodensee Hospital of Singen, Department of Pathology, 78207 Singen, Germany. [Ringli, Daniel] Hegau-Bodensee Hospital of Singen, Department of Pathology, 78207 Singen, Germany. [Tonhauser, Jens] Hegau-Bodensee Hospital of Singen, Department of UrologySingen, Germany. [Poser, Immanuel] Loretto Hospital, Department of Urology and Urologic OncologyFreiburg, Germany. [Breul, Jurgen] Loretto Hospital, Department of Urology and Urologic OncologyFreiburg, Germany. [Gevensleben, Heidrun] University of Bonn, Institute of PathologyBonn, Germany. [Seifert, Hans-Helge] Hegau-Bodensee Hospital of Singen, Department of UrologySingen, Germany. RP Helpap, B (reprint author), Hegau-Bodensee Hospital of Singen, Department of Pathology, 78207 Singen, Germany. EM burkhard.helpap@hbh-kliniken.de CR Tolonen TT, Kujala PM, Tammela TL, Tuominen VJ, Isola JJ, Visakorpi T, 2011, Overall and worst Gleason scores are equally good predictors of prostate cancer progression. BMC Urol 11:21. doi 10.1186/1471-2490-11-21 Epstein JI, Allsbrook SC, Amin MB, Egevad L, and the ISUP Grading Committee, 2005, The 2005 international society of urological pathology ISUP consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol 29:1228–1242. Epstein JI, 2010, An update of the Gleason grading system. J Urol 183:433–440 Vira MA, Guzzo T, Heitjan DF, Tomaszewski JE, D'Amico A, Wein AJ, Malkowicz SB, 2008, Is the biopsy Gleason score important in predicting outcomes for patients after radical prostatectomy once the pathological Gleason score is known? BJU Int 101:1232–1236 Lau WK, Blute MI, Bostwick DG,Weaver AL, Sebo TI, Zincke H, 2001, Prognostic factors for survival of patients with pathological Gleason score 7 prostate cancer: differences in outcome between primary Gleason grades 3 and 4. J Urol 166:1692–1697 Gleason DE, 1966, Classification of prostatic carcinomas. Cancer Chemother Rep 50:125–128 Roder MA, Berg KD, Gruschy L, Brasso K, Iversen P, 2011, First Danish single-institution experience with radical prostatectomy: biochemical outcome in 1200 consecutive patients. Prostate Cancer 2011:236357., DOI 10.1155/2011/236357 Epub 2010 Dec 22 Amin A, Partin A, Epstein JI, 2011, Gleason score 7 prostate cancer on needle biopsy: relation of primary pattern 3 or 4 to pathological stage and progression after radical prostatectomy. J Urol 186:1286– 1290 Dong F,Wang C, Farris AB,Wu S, Lee H, Olumi AF, DouglasWS, Young RH, Wu CL, 2012, Impact on the clinical outcome of prostate cancer by the 2005 international society of urological pathology modified Gleason grading system. Am J Surg Pathol 36:838–843 Helpap B, Egevad L, 2009)Modified Gleason grading. An updated review. Histol Histopathol 24:661–666 Makarov DV, Sanderson H, Partin AW, Epstein JI, 2002, Gleason score 7 prostate cancer on needle biopsy. Is the prognostic difference in Gleason score 4 + 3 and 3 + 4 independent of the number of involved cores? J Urol 167:2440–2442 Rasiah KK, Stricker PD, Haynes A-M, Delprado W, Turner JJ, Golovsky D, Brenner PC, Kooner R, O’Neill GF, Grygiel JJ, Sutherland RL, Henshall SM, 2003, Prognostic significance of Gleason pattern in patients with Gleason score 7 prostate carcinoma. Cancer 98:2560–2565 Chan TY, Partin AW, Walsh PC, Epstein JI, 2000, Prognostic significance of Gleason score 3 + 4 versus Gleason score 4 + 3 tumor at radical prostatectomy. Urology 56:823–827 SakrWA, Tefilli MV, Grignon DJ, Banerjee M, Dey J, Gheiler EL, Tiguert R, Powell IJ, Wood DP, 2000, Gleason score 7 prostate cancer: a heterogeneous entity? Correlation with pathologic parameters and disease-free survival. Urology 56:730–734 Lavery HJ, Droller MJ, 2012, Do Gleason patterns 3 and 4 prostate cancer represent separate disease states? J Urol 188:1667–1675 Stark JR, Perner S, Stampfer MJ, Sinnott JA, Finn S, Eisenstein AS, Ma J, Fiorentino M, Kurth T, Loda M, Giovannucci EL, Rubin M, Mucci LA, 2009, Gleason score and lethal prostate cancer: does 3 + 4 = 4 + 3? J Clin Oncol 27:3459–3464 Helpap B, Egevad L, 2006, The significance of modified Gleason grading of prostatic carcinoma in biopsy and radical prostatectomy specimens. Virchows Arch 449:622–627 Fanning DM, Kay E, Fan Y, Fitzpatrick JM, Watson RW, 2010, Prostate cancer grading: the effect of stratification of needle biopsy Gleason score 4 + 3 as high or intermediate grade. BJU Int 105: 631–635 Nayyar R, Singh P, Gupta NP, Hemal AK, Dogra PN, Seth A, Kumar R, 2010, Upgrading of Gleason score on radical prostatectomy specimen compared to the pre-operative needle core biopsy: an Indian experience. Indian J Urol 26:56–59 Egevad L, Mazzucchelli R, Montironi R, 2012, Implications of the international society of urological pathology modified Gleason grading system. Arch Pathol Lab Med 136:426–434 Zareba P, Zhang J, Yilmaz A, Trpkov K, 2009, The impact of the 2005 international society of urological pathology, ISUP, consensus on Gleason grading in contemporary practice. Histopathology 55:384–391 Epstein JI, Feng Z, Trock BJ, Pierorazio PM, 2012, Upgrading and downgrading of prostate cancer from biopsy to radical prostatectomy: incidence and predictive factors using the modified Gleason grading system and factoring in tertiary grades. Eur Urol 61:1019–1024 Uemura H, Hoshino K, Sasaki T, Miyoshi Y, Ishiguro H, Inayama Y, Kubota Y, 2009, Usefulness of the 2005 international society of urologic pathology Gleason grading system in prostate biopsy and radical prostatectomy specimens. BJU Int 103:1190–1194 Billis A, Guimaraes MS, Freitas LL, Meirelles L, Magna LA, Ferreira U, 2008, The impact of the 2005 international society of urological pathology consensus conference on standard Gleason grading of prostatic carcinoma in needle biopsies. J Urol 180: 548–552 Berney DM, Algaba F, Camparo P, Comperat E, Griffiths D, Kristiansen G, Lopez-Beltran A, Montironi R, Varma M, Egevad L, 2013, The reasons behind variation in Gleason grading of prostatic biopsies: areas of agreement and misconception among 266 European pathologists. Histopathology 64:405–411 Helpap B, Kristiansen G, Kollermann J, Shaikhibrahim Z, Wernert N,OehlerU, FellbaumC, 2013, Significance ofGleason grading of low-grade carcinoma of the prostate with therapeutic option of active surveillance. Urol Int 90:17–23 Helpap B, Kollermann J, 2012, Combined histoarchitectural and cytological biopsy grading improves grading accuracy in low-grade prostate cancer. Int J Urol 19:126–133 Bostwick DG, Foster CS, Algaba F, Hutter RVP, Montironi R, Mostofi FK, et al., 2000, Prostate tissue factor. In: Murphy G, Khoury S, Partin A, Denis L, eds, Prostate cancer. Second international consultation on prostate cancer. Plymbridge Distributors Ltd, Paris, pp. 162–201 Mostofi FK, Sesterhenn IA, Davis CJ, in collaboration with LH Sobin and pathologists from 10 countries, 2002, Histological typing of prostate tumours in WHO international histological classification of tumours. Springer Berlin-Heidelberg, pp. 14–16. Van Veggel BAMH, von Ort IM, Witjes JA, Kiemeney LALM, Hulsbergen-van de Kaa CA, 2011, Quantification of extraprostatic extension in prostate cancer: different parameters correlated to biochemical recurrence after radical prostatectomy. Histopathology 59: 692–702 Han JS, Toll AD, Amin A, Carter HB, Landis P, Lee S, Epstein JI, 2012, Low prostate-specific antigen and no Gleason score upgrade despite more extensive cancer during active surveillance predicts insignificant prostate cancer at radical prostatectomy. Urology 80: 883–888 Epstein JI, 2011, Prognostic significance of tumor volume in radical prostatectomy and needle biopsy specimens. J Urol 186:790–797 Cooperberg MR, Broering JM, Kantoff PW, Carroll PR, 2007, Contemporary trends in low risk prostate cancer: risk assessment and treatment. J Urol 178:14–19 Epub 2007 Jul 20 Cooperberg MR, Broering JM, Carroll PR, 2010, time trends and local variation in primary treatment of localized prostate cancer. J Clin Oncol 28:1117–1123., DOI 10.1200/JCO.2009.26.0133. Epub 2010 Feb 1. Helpap B, Ringli D, Shaikhibrahim Z, Wernert N, Kristiansen G, 2013, The heterogeneous gleason 7 carcinoma of the prostate: analyses of low and high grade, risk, carcinomas with criteria of the international society of urological pathology, ISUP). Pathol Res Pract 209:190–194 Cheng L, Montironi R, Bostwick DG, Lopez-Beltran A, Berney DM, 2012, Staging of prostate cancer. Histopathology 60:87–117 Muntener M, Epstein JI, HernandezDJ, Gonzalgo ML,Mangold LO, Hymphreys E, Walsh PC, Partin AW, 2008, Prognostic significance of Gleason score discrepancies between needle biopsy and radical prostatectomy. Eur Urol 53:767–776 Ganz PA, Barry JM, Burke W, Col NF, Corso PS, Dodson E, Hammond ME, Kogan BA, Lynch CF, Newcomer L, Seifter EJ, Tooze JA, Viswanath K, Wessells H, 2012, National institutes of health state-of-the-science conference: role of active surveillance in the management of men with localized prostate cancer. Ann Intern Med 156:591–595 Shapiro RH, Johnstone PA, 2012, Risk of Gleason grade inaccuracies in prostate cancer patients eligible for active surveillance. Urology 80:661–666 Kristiansen G, Stockle M, Albers P, Schmidberger H, Martus P, Wellek S, Harter M, Bussar-Maatz R, Wiegel T, 2013, The importance of pathology in the German cancer study PREFERE. Pathologe 34:449–462 Carter HB, Partin AW, Walsh PC, Trock BJ, Veltri RW, Nelson WG, Coffey DS, Singer EA, Epstein JI, 2012, Gleason score 6 adenocarcinoma: should it be labeled as cancer? J Clin Oncol 35: 4294–4296., DOI 10.1200/JCO.2012.44.0586 Epub 2012 Oct 1 Hernandez DJ, Nielsen ME, Han M, Trock BJ, Partin AW, Walsh PC, Epstein JI, 2008, Natural history of pathologically organconfined, pT2, Gleason score 6 or less, prostate cancer after radical prostatectomy. Urology 72:172–176 Han M, Partin AW, Zahurak M, Piantalosi S, Epstein JL,Walsh PC, 2003, Biochemical, prostate specific antigen, recurrence probability following radical prostatectomy for clinical localized prostate cancer. J Urol 169:517–523 Pierorazio PM,Walsh PC, Partin AW, Epstein JI, 2013, Prognostic Gleason grade grouping: data based on the modified Gleason scoring system. BJU Int 111:753–760 Isbarn H, Karakiewicz PI, Ahyai SA, Chun FK, Jeldres C, Haese A, Heinzer H, Zacharias M, Heuer R, Eichelberg C, Steuber T, Budaus L, Kollermann J, Salomon G, Schlomm T, Perrotte P, Fisch M, Huland H, Graefen M, 2010, Differences in histopathological and biochemical outcomes in patients with low Gleason score prostate cancer. BJU Int 105:818–823 Bill-Axelson A, Holmberg L, Garmo H, et al., 2014, Radical prostatectomy or watchful waiting in early prostate cancer. N Engl J Med 370:932–940 Isbarn H, Huland H, Graefen M, 2013, Results of radical prostatectomy in newly diagnosed prostate cancer – long term survival rates in locally advanced and high risk cancers. Dtsch Arztebl Int 110:497–503 Van der Kwast T, Bubendorf L, Mazerolles C, Raspollini MR, Van Leenders GJ, Pihl CG, Kujala P, 2013, Guidelines on processing and reporting of prostate biopsies: the 2013 update of the pathology committee of the European randomized study of screening for prostate cancer, ERSPC). Virchows Arch 463:367–377., DOI 10.1007/ s00428-013-1466-5 Epub 2013 Aug 6 Andreoiu M, Cheng L, 2010, Multifocal prostate cancer: biologic, prognostic, and therapeutic implications. Hum Pathol 41:781–793 Montironi R, Hammond EH, Lin DW, Gore JL, Srigley JR, Samaratunga H, Evevad L, Rubin MA, Nacey J, Klotz L, Sandler H, Zietman AL, Holden S, Humphrey PA, Evans AJ, Delahunt B, McKenney JK, Berney D, Wheeler TM, Chinnaiyan A, True L, Knudsen B, Epstein JI,AminMB(2014, Consensus statement with recommendations on active surveillance inclusion criteria and definition of progression in men with localized prostate cancer: the critical role of the pathologist. Virchows Arch 465:623–628 Amin M, Lin DW, Core L, et al., 2014, The critical role of the pathologist in determining eligibility for active surveillance as a management option in patients with prostate cancer. Arch Pathol Lab Med 138:1387–1405 Chun FK, Steuber T, Erbersdobler A, Currlin E, Walz J, Schlomm T, Haese A, Heinzer H, McCormack M, Huland H, Graefen M, Karakiewicz PI, 2006, Development and internal validation of a nomogram predicting the probability of prostate cancer Gleason sum upgrading between biopsy and radical prostatectomy pathology. Eur Urol 49:820–826 Ward JF, Slezak JM, Blute ML, Bergstralh EJ, Zincke H, 2005, Radical prostatectomy for clinically advanced, cT3, prostate cancer since the advent of prostate-specific antigen testing: 15-year outcome. BJU Int 95:751–756 Pierorazio PM, Guzzo TJ, Han M, Bivalacqua TJ, Epstein JI, Schaeffer EM, Schoenberg M, Walsh PC, Partin AW, 2010, Long-term survival after radical prostatectomy for men with high Gleason sum in pathologic specimen. Urology 76:715–721 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 349 EP 356 DI 10.1007/s12253-015-0013-x PG 8 ER PT J AU Kaabi, B Belaaloui, G Benbrahim, W Hamizi, K Sadelaoud, M Toumi, W Bounecer, H AF Kaabi, Batoul Belaaloui, Ghania Benbrahim, Wassila Hamizi, Kamel Sadelaoud, Mourad Toumi, Wided Bounecer, Hocine TI ADRA2A Germline Gene Polymorphism is Associated to the Severity, but not to the Risk, of Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ADRA2A; Single nucleotide polymorphism (SNP); Breast cancer; Risk; Prognosis; Case-control study ID ADRA2A; Single nucleotide polymorphism (SNP); Breast cancer; Risk; Prognosis; Case-control study AB Breast cancer (BC) prognosis and risk were associated to obesity, metabolic syndrome and type 2 diabetes mellitus. Two Single Nucleotide Polymorphisms (SNPs) of the adrenergic receptor-2a gene (ADRA2A): rs1800544 and rs553668, have been associated to these metabolic disorders. We investigated these SNPs in BC risk and prognosis.A total of 102 BC patients and 102 healthy controls were included. The rs1800544 and rs553668 were determined by real-time PCR. Genotypes and haplotypes frequencies between patients and controls, and for different clinico-pathologic parameters were compared. We found a significant association of rs1800544 GG genotype with young age at diagnosis, premenopausal status, higher tumor size, metastasis in lymph nodes, advanced TNM stages and higher Nottingham Prognosis Indicator (NPI) (p < 0.05). There was no association between rs1800544 and SBR stages, Her2, ER and PR statuses and the molecular classification. The rs553668 AA genotype was associated to young age at diagnosis and premenopausal status (p < 0.05). The haplotype GA was associated to the early age of diagnosis (p = 0.03), and the haplotype GG to higher tumor size, lymph node involvement, advanced TNM stages and Her2 positive status (p < 0.05). There was no polymorphism or haplotype association with BC risk (p > 0.05). ADRA2A polymorphismis associated with indicators BC poor prognosis but not with BC susceptibility. This is the first report suggesting that ADRA2A germline gene polymorphismcould represent a predictor factor for BC outcome. Further investigation of other ADRA2A polymorphisms in BC risk or prognosis are needed and may lead to a genotype-based therapy. C1 [Kaabi, Batoul] Batna 1 University, Faculty of SciencesBatna, Algeria. [Belaaloui, Ghania] Batna 2 University, Faculty of Medicine, Citee Ezzouhour, 05000 Batna, Algeria. [Benbrahim, Wassila] Batna 2 University, Faculty of Medicine, Citee Ezzouhour, 05000 Batna, Algeria. [Hamizi, Kamel] Batna 2 University, Faculty of Medicine, Citee Ezzouhour, 05000 Batna, Algeria. [Sadelaoud, Mourad] LAM LaboratoryBatna, Algeria. [Toumi, Wided] LAM LaboratoryBatna, Algeria. [Bounecer, Hocine] Batna 2 University, Faculty of Medicine, Citee Ezzouhour, 05000 Batna, Algeria. RP Belaaloui, G (reprint author), Batna 2 University, Faculty of Medicine, 05000 Batna, Algeria. EM gbelaaloui@yahoo.fr CR Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2013, GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide. IARC CancerBase 2012; No. 11 [Internet]. Lyon, France: International Agency for Research on Cancer. Available from: http://globocan. iarc.fr. Accessed on 2019th Dec 2014 Pusztai L, 2011, Anatomy and biology: two complementary sides of breast cancer prognostication. J Clin Oncol 29(33):4347–4348 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, et al., 2011, Strategies for subtypes–dealing with the diversity of breast cancer: highlights of the St. Gallen international expert consensus on the primary therapy of early breast cancer 2011. Ann Oncol 22(8):1736–1747 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, et al., 2013, Personalizing the treatment of women with early breast cancer: highlights of the St gallen international expert consensus on the primary therapy of early breast cancer 2013. Ann Oncol 24(9):2206–2223 Zhang B, Beeghly-Fadiel A, Long J, Zheng W, 2011, Genetic variants associated with breast-cancer risk: comprehensive research synopsis, meta-analysis, and epidemiological evidence. Lancet Oncol 12(5):477–488 Rafiq S, Khan S, Tapper W, Collins A, Upstill-Goddard R, et al., 2014, A genome wide meta-analysis study for identification of common variation associated with breast cancer prognosis. PLoS One 9(12):e101488 Sapkota Y, 2014, Germline DNA variations in breast cancer predisposition and prognosis: a systematic review of the literature. Cytogenet Genome Res 144(2):77–91 Ribelles N, Santonja A, Pajares B, Llacer C, Alba E, 2014, The seed and soil hypothesis revisited: current state of knowledge of inherited genes on prognosis in breast cancer. Cancer Treat Rev 40(2):293–299 Kobilka BK, Matsui H, Kobilka TS, Yang-Feng TL, Francke U, et al., 1987, Cloning, sequencing, and expression of the gene coding for the human platelet alpha 2-adrenergic receptor. Science 238(4827):650–656 Hoffman BB, Taylor P, 2001, Neurotransmission: the autonomic and somatic motor nervous systems. In: Hardman JG, Limbird LE, Gilman AG, eds, Goodman & Gilman’s the pharmacological basis of therapeutics. McGraw-Hill, New York, pp. 115–153 Lario S, Calls J, Cases A, Oriola J, Torras A, et al., 1997, MspI identifies a biallelic polymorphism in the promoter region of the alpha 2A-adrenergic receptor gene. Clin Genet 51(2):129–130 Lima JJ, Feng H, Duckworth L,Wang J, Sylvester JE, et al., 2007, Association analyses of adrenergic receptor polymorphisms with obesity and metabolic alterations. Metabolism 56(6):757–765 Garenc C, Perusse L, Chagnon YC, Rankinen T, Gagnon J, et al., 2002, The alpha 2-adrenergic receptor gene and body fat content and distribution: the HERITAGE family study. Mol Med 8(2): 88–94 Kurnik D, Muszkat M, Li C, Sofowora GG, Friedman EA, et al., 2011, Genetic variations in the alpha(2A)-adrenoreceptor are associated with blood pressure response to the agonist dexmedetomidine. Circ Cardiovasc Genet 4(2):179–187 Oppert JM, Tourville J, Chagnon M, Mauriege P, Dionne FT, et al., 1995, DNA polymorphisms in the alpha 2- and beta 2-adrenoceptor genes and regional fat distribution in humans: association and linkage studies. Obes Res 3(3):249–255 Ukkola O, Rankinen T,Weisnagel SJ, Sun G, Perusse L, et al., 2000, Interactions among the alpha2-, beta2-, and beta3-adrenergic receptor genes and obesity-related phenotypes in the Quebec family study. Metabolism 49(8):1063–1070 Rosengren AH, Jokubka R, Tojjar D, Granhall C, Hansson O, et al., 2010, Overexpression of alpha2A-adrenergic receptors contributes to type 2 diabetes. Science 327(5962):217–220 Bo S, Cassader M, Cavallo-Perin P, Durazzo M, Rosato R, et al., 2012, The rs553668 polymorphism of the ADRA2A gene predicts the worsening of fasting glucose values in a cohort of subjects without diabetes. A population-based study. Diabet Med 29(4): 549–552 Talmud PJ, Cooper JA, Gaunt T, Holmes MV, Shah S, et al., 2011, Variants of ADRA2A are associated with fasting glucose, blood pressure, body mass index and type 2 diabetes risk: meta-analysis of four prospective studies. Diabetologia 54(7):1710–1719 Trentham-Dietz A, Newcomb PA, Storer BE, Longnecker MP, Baron J, et al., 1997, Body size and risk of breast cancer. Am J Epidemiol 145(11):1011–1019 van den Brandt PA, Spiegelman D, Yaun SS, Adami HO, Beeson L, et al., 2000, Pooled analysis of prospective cohort studies on height, weight, and breast cancer risk. Am J Epidemiol 152(6):514–527 Key TJ, Appleby PN, Reeves GK, Roddam A, Dorgan JF, et al., 2003, Body mass index, serum sex hormones, and breast cancer risk in postmenopausal women. J Natl Cancer Inst 95(16):1218– 1226 Sinicrope FA, Dannenberg AJ, 2011, Obesity and breast cancer prognosis: weight of the evidence. J Clin Oncol 29(1):4–7 DeCensi A, Gennari A, 2011, Insulin breast cancer connection: confirmatory data set the stage for better care. J Clin Oncol 29(1): 7–10 Vazquez SM, Mladovan AG, Perez C, Bruzzone A, Baldi A, et al., 2006, Human breast cell lines exhibit functional alpha2-adrenoceptors. Cancer Chemother Pharmacol 58(1):50–61 Perez Pinero C, Bruzzone A, Sarappa MG, Castillo LF, Luthy IA, 2012, Involvement of alpha2- and beta2-adrenoceptors on breast cancer cell proliferation and tumour growth regulation. Br J Pharmacol 166(2):721–736 Vazquez SM, Pignataro O, Luthy IA, 1999, Alpha2-adrenergic effect on human breast cancer MCF-7 cells. Breast Cancer Res Treat 55(1):41–49 Chiesa IJ, Castillo LF, Luthy IA, 2008, Contribution of alpha2- adrenoceptors to the mitogenic effect of catecholestrogen in human breast cancer MCF-7 cells. J Steroid Biochem Mol Biol 110(1–2): 170–175 Bruzzone A, Pinero CP, Castillo LF, Sarappa MG, Rojas P, et al., 2008, Alpha2-adrenoceptor action on cell proliferation and mammary tumour growth in mice. Br J Pharmacol 155(4):494–504 Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, et al., 2000, Molecular portraits of human breast tumours. Nature 406(6797):747–752 Todd JH, Dowle C,WilliamsMR, ElstonCW, Ellis IO, et al., 1987, Confirmation of a prognostic index in primary breast cancer. Br J Cancer 56(4):489–492 Sole X, Guino E, Valls J, Iniesta R, Moreno V, 2006, SNPStats: a web tool for the analysis of association studies. Bioinformatics 22(15):1928–1929 Wirapati P, Sotiriou C, Kunkel S, Farmer P, Pradervand S, et al., 2008, Meta-analysis of gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures. Breast Cancer Res 10(4):R65 El Saghir NS, Seoud M, Khalil MK, Charafeddine M, Salem ZK, et al., 2006, Effects of young age at presentation on survival in breast cancer. BMC Cancer 6:194 Bharat A, Aft RL, Gao F, Margenthaler JA, 2009, Patient and tumor characteristics associated with increased mortality in young women, < or =40 years, with breast cancer. J Surg Oncol 100(3): 248–251 Anders CK, Johnson R, Litton J, Phillips M, Bleyer A, 2009, Breast cancer before age 40 years. Semin Oncol 36(3):237–249 Le Doussal V, Tubiana-Hulin M, Friedman S, Hacene K, Spyratos F, et al., 1989, Prognostic value of histologic grade nuclear components of scarff-bloom-Richardson, SBR). an improved score modification based on a multivariate analysis of 1262 invasive ductal breast carcinomas. Cancer 64(9):1914–1921 Osborne CK, Yochmowitz MG, Knight 3rd WA, McGuire WL, 1980, The value of estrogen and progesterone receptors in the treatment of breast cancer. Cancer 1980 46(12 Suppl):2884–2888 McGuire WL, Osborne CK, Clark GM, Knight 3rd WA, 1982, Steroid hormone receptors and carcinoma of the breast. Am J Phys 243(2):E99–102 Clark GM, McGuire WL, 1988, Steroid receptors and other prognostic factors in primary breast cancer. Semin Oncol 15(2 Suppl 1): 20–25 Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, et al., 1987, Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235(4785): 177–182 Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, et al., 2001, Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98(19):10869–10874 Hugh J, Hanson J, Cheang MC, Nielsen TO, Perou CM, et al., 2009, Breast cancer subtypes and response to docetaxel in nodepositive breast cancer: use of an immunohistochemical definition in the BCIRG 001 trial. J Clin Oncol 27(8):1168–1176 Boyle DP, McCourt CM, Matchett KB, Salto-Tellez M, 2013, Molecular and clinicopathological markers of prognosis in breast cancer. Expert Rev Mol Diagn 13(5):481–498 Small KM, Brown KM, Seman CA, Theiss CT, Liggett SB, 2006, Complex haplotypes derived from noncoding polymorphisms of the intronless alpha2A-adrenergic gene diversify receptor expression. Proc Natl Acad Sci U S A 103(14): 5472–5477 Lim E, Vaillant F, Wu D, Forrest NC, Pal B, et al., 2009, Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat Med 15(8): 907–913 Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, et al., 2008, Stromal gene expression predicts clinical outcome in breast cancer. Nat Med 14(5):518–527 Sotiriou C, Neo SY, McShane LM, Korn EL, Long PM, et al., 2003, Breast cancer classification and prognosis based on gene expression profiles from a population-based study. Proc Natl Acad Sci U S A 100(18):10393–10398 Shkurnikov MY, Galatenko VV, Lebedev AE, Podol’skii VE, Tonevitskii EA, et al., 2014, On statistical relationship between ADRA2A expression and the risk of breast cancer relapse. Bull Exp Biol Med 157(4):454–458 Du Y, Zhou L,Wang Y, Yan T, Jiang Y, et al., 2014, Association of alpha2a and beta2 adrenoceptor expression with clinical outcome in breast cancer. Curr Med Res Opin 30(7):1337–1344 Lutgendorf SK, Sood AK, Antoni MH, 2010, Host factors and cancer progression: biobehavioral signaling pathways and interventions. J Clin Oncol 28(26):4094–4099 Docherty JR, 1998, Subtypes of functional alpha1- and alpha2- adrenoceptors. Eur J Pharmacol 361(1):1–15 Giessler C, Wangemann T, Silber RE, Dhein S, Brodde OE, 2002, Noradrenaline-induced contraction of human saphenous vein and human internal mammary artery: involvement of different alphaadrenoceptor subtypes. Naunyn Schmiedeberg's Arch Pharmacol 366(2):104–109 Bruckmaier RM, Wellnitz O, Blum JW, 1997, Inhibition of milk ejection in cows by oxytocin receptor blockade, alpha-adrenergic receptor stimulation and in unfamiliar surroundings. J Dairy Res 64(3):315–325 Su F, Ouyang N, Zhu P, JiaW, Gong C, et al., 2005, Psychological stress induces chemoresistance in breast cancer by upregulating mdr1. Biochem Biophys Res Commun 329(3):888–897 Flint MS, Kim G, Hood BL, Bateman NW, Stewart NA, et al., 2009, Stress hormones mediate drug resistance to paclitaxel in human breast cancer cells through a CDK-1-dependent pathway. Psychoneuroendocrinology 34(10):1533–1541 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 357 EP 365 DI 10.1007/s12253-015-0010-0 PG 9 ER PT J AU Zhang, Y Liu, P Jiang, Y Dou, X Yan, J Ma, Ch Fan, Q Wang, W Su, F Tang, H Su, X AF Zhang, Yafei Liu, Peng Jiang, Yizhen Dou, Xiaofeng Yan, Jianghua Ma, Chao Fan, Qun Wang, Weixing Su, Fu Tang, Hui Su, Xinhui TI High Expression of Neuropilin-1 Associates with Unfavorable Clinicopathological Features in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Neuropilin-1; Expression; Clinicopathology ID Hepatocellular carcinoma; Neuropilin-1; Expression; Clinicopathology AB As a co-receptor for vascular endothelial growth factor (VEGF), Neuropilin-1 (NRP-1) plays an important role in angiogenesis and malignant progression of many human cancers. However, the role of NRP-1 in hepatocellular carcinoma (HCC) is not well understood. The study aimed to detected the expression of Neuropilin-1 in HCC and investigate the association between its expression and the clinicopathological characteristics and prognosis of HCC. Quantitative real-time PCR (qRT-PCR), Western blot, Immunofluorescence and immunohistochemistry (IHC) analyses were performed to characterize the expression of NRP-1 in HCC cell lines and tissues. The association of NRP-1 expression with the clinicopathological characteristics and the prognosis was subsequently assessed. qRT-PCR and Western blot assays revealed that the expression of NRP-1 in HCC was significantly increased relative to that of normal live cells and tissues (P < 0.05,and <0.001, respectively). In addition, high expression of NRP-1 was significantly associated with intrahepatic metastasis (P = 0.036), Edmondson grade (P = 0.007), TNM classification (P = 0.0031), and portal vein invasion (P = 0.004). Furthermore, the HCC patients with high NRP- 1 expression had shorter overall survival (OS), and recurrence-free survival (RFS), whereas, patients with low NRP-1 expression had better OS and RFS (P = 0.0035, and 0.0048, respectively). These data indicate that NRP-1 expression may play an important role in the progression of HCC, and that high NRP-1 expression suggests unfavorable clinicopathological characteristics and survival in HCC patients. C1 [Zhang, Yafei] Xiamen University, Zhongshan Hospital, Department of Nuclear Medicine, No.201 Hubin South Road, 361004 Xiamen, China. [Liu, Peng] Xiamen University, Zhongshan Hospital, Department of Nuclear Medicine, No.201 Hubin South Road, 361004 Xiamen, China. [Jiang, Yizhen] Xiamen University, Zhongshan Hospital, Department of Nuclear Medicine, No.201 Hubin South Road, 361004 Xiamen, China. [Dou, Xiaofeng] Zhejiang University School of Medicine, The Second Affiliated Hospital, Department of Nuclear Medicine, No.88 Jiefang Road, 310009 Hangzhou, China. [Yan, Jianghua] Xiamen University, Medical School, Cancer Research Center, Xiangan South Road, 361102 Xiamen, China. [Ma, Chao] Xiamen University, Zhongshan Hospital, Department of Nuclear Medicine, No.201 Hubin South Road, 361004 Xiamen, China. [Fan, Qun] Xiamen University, Zhongshan Hospital, Department of Nuclear Medicine, No.201 Hubin South Road, 361004 Xiamen, China. [Wang, Weixing] Xiamen University, Zhongshan Hospital, Department of Nuclear Medicine, No.201 Hubin South Road, 361004 Xiamen, China. [Su, Fu] Xiamen University, Zhongshan Hospital, Department of Nuclear Medicine, No.201 Hubin South Road, 361004 Xiamen, China. [Tang, Hui] Xiamen University, Zhongshan Hospital, Department of Nuclear Medicine, No.201 Hubin South Road, 361004 Xiamen, China. [Su, Xinhui] Xiamen University, Zhongshan Hospital, Department of Nuclear Medicine, No.201 Hubin South Road, 361004 Xiamen, China. RP Su, X (reprint author), Xiamen University, Zhongshan Hospital, Department of Nuclear Medicine, 361004 Xiamen, China. EM suxinhuii@163.com CR Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63(1):11–30 Qu LS, Liu JX, Liu TT, Shen XZ, Chen TY, Ni ZP, Lu CH, 2014, Association of hepatitis B virus pres- deletions with the development of hepatocellular carcinoma in qidong, China. PLoS One 9(5): e98257 Fong ZV, Tanabe KK, 2014, The clinical management of hepatocellular carcinoma in the United States, Europe, and Asia: a comprehensive and evidence-based comparison and review. Cancer 120(18):2824–2830 Wong RJ, Devaki P, Nguyen L, Cheung R, Nguyen MH, 2014, Ethnic disparities and liver transplantation rates in hepatocellular carcinoma patients in the recent era: results from the surveillance, epidemiology, and end results registry. Liver Transpl 20(5):528– 535 Marrero JA, El-Serag HB, 2011, Alpha-fetoprotein should be included in the hepatocellular carcinoma surveillance guidelines of theAmerican association for the study of liver diseases. Hepatology 53(3):1060–1061 author reply 1061-1062 He Z, Tessier-Lavigne M, 1997, Neuropilin is a receptor for the axonal chemorepellent semaphorin III. Cell 90(4):739–751 Oh H, Takagi H, Otani A, Koyama S, Kemmochi S, Uemura A, Honda Y, 2002, Selective induction of neuropilin-1 by vascular endothelial growth factor, VEGF): a mechanism contributing to VEGF -induced angiogenesis. Proc Natl Acad Sci U S A 99(1): 383–388 Xu Y, Li P, Zhang X, Wang J, Gu D, Wang Y, 2013, Prognostic implication of neuropilin-1 up- regulation in human nasopharyngeal carcinoma. Diagn Pathol 8:155 ChengW, Fu D,Wei ZF, Xu F, Xu XF, Liu YH, et al., 2014, NRP-1 expression in bladder cancer and its implications for tumor progression. Tumour Biol 36(6):6089–6094 Pan Q, Chanthery Y, Liang WC, Stawicki S, Mak J, Rathore N, et al., 2007, Blocking neuropilin-1 function has an additive effect with anti-VEGF to inhibit tumor growth. Cancer Cell 11(1):53–67 Berge M, Allanic D, Bonnin P, de Montrion C, Richard J, Suc M, et al., 2011, Neuropilin-1 is upre- gulated in hepatocellular carcinoma and contributes to tumour growth and vascular remodelling. J Hepatol 55(4):866–875 Chen L, Miao W, Tang X, Zhang H, Wang S, Luo F, Yan J, 2013, The expression and significan- ce of neuropilin-1, NRP-1, on glioma cell lines and glioma tissues. J Biomed Nanotechnol 9(4):559– 563 Li X, Luo F,Wang S, Ni E, Tang X, Lv H, et al., 2011, Monoclonal antibody against NRP-1 b1b2. Hybridoma, Larchmt, 30(4):369– 373 Varotti G, Ramacciato G, Ercolani G, Grazi GL, Vetrone G, et al., 2005, Comparison between the fifth and sixth editions of the AJCC/UICC TNM staging systems for hepatocellular carcinoma: multicentric study on 393 cirrhotic resected patients. Eur J Surg Oncol 31(7):760–767 Kamiya T, Kawakami T, Abe Y, Nishi M, Onoda N, Miyazaki N, et al., 2006, The preserved expre- ssion of neuropilin, NRP, 1 contributes to a better prognosis in colon cancer. Oncol Rep 15(2):369–373 Su X, Chen Q, Chen W, Chen T, Li W, Li Y, et al., 2014, Mycoepoxydiene inhibits activation of BV2 microglia stimulated by lipopolysaccharide through suppressing NF-κB, ERK 1/2 and toll-like receptor pathways. Int Immunopharmacol 19(1):88–93 Li W, Li M, Su X, Qin L, Miao M, Yu C, et al., 2014, Mycoepoxydiene induces apoptosis and inhibits TPA-induced invasion in human cholangiocarcinoma cells via blocking NF-κB pathway. Bio- Chimie 101:183–191 Gu X, Fu M, Ge Z, Zhan F, Ding Y, Ni H, et al., 2014, High expression of MAGE-A9 correlates with unfavorable survival in hepatocellular carcinoma. Sci Rep. 4:6625 Herzog Y, Kalcheim C, Kahane N, Reshef R, Neufeld G, 2001, Differential expression of neuropilin-1 and neuropilin-2 in arteries and veins. Mech Dev 109(1):115–119 Zhu H, Cai H, Tang M, Tang J, 2014, Neuropilin-1 is overexpressed in osteosarcoma and contributes to tumor progression and poor prognosis. Clin Transl Oncol 16(8):732–738 Zhuang PY, Wang JD, Tang ZH, Zhou XP, Yang Y, Quan ZW, et al., 2014, Peritumoral neuropilin- 1 and VEGF receptor-2 expression increases time to recurrence in hepatocellularcarcinoma patients undergoing curative hepatectomy. Oncotarget 5(22):11121–11121 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 367 EP 375 DI 10.1007/s12253-015-0003-z PG 9 ER PT J AU Fernandez-Acenero, JM Cortes, D Gomez del Pulgar, T Cebrian, A Estrada, L Martinez-Useros, J Celdran, A Garcia-Foncillas, J Pastor, C AF Fernandez-Acenero, Jesus Maria Cortes, Delia Gomez del Pulgar, Teresa Cebrian, Arancha Estrada, Lourdes Martinez-Useros, Javier Celdran, Angel Garcia-Foncillas, Jesus Pastor, Carlos TI PLK-1 Expression is Associated with Histopathological Response to Neoadjuvant Therapy of Hepatic Metastasis of Colorectal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatic metastasis; Colorectal carcinoma; Neoadjuvant therapy; PLK1; Histopathological response; Targeted drugs ID Hepatic metastasis; Colorectal carcinoma; Neoadjuvant therapy; PLK1; Histopathological response; Targeted drugs AB Polo-like kinase 1 (PLK1) is a serine/threonineprotein kinase expressed during mitosis and overexpressed in multiple human cancers, including leukemia and also many solid tumors. PLK1 knockdown has been shown to block proliferation of leukemic cell lines and the clonogenic potential of tumor cells grown from patients with cancer. PLK1 inhibition is a promising strategy for the treatment of some tumors. We aim to analyze expression of PLK1 in metastatic colorectal carcinoma. Retrospective analysis of colorectal carcinomas with hepatic metastasis during follow-up receiving neoadjuvant chemotherapy (NAC), based on oxaliplatin. Immunohistochemistry for PLK-1 in paraffin-embedded tissue from the primary and also from the metastasis. 50 patients. 32 % showed good histopathological response. 43 % of the primaries were positive for PLK1, as opposed to 23.5%of the metastasis. Expression of PLK1 was significantly reduced in metastasis compared with the primaries (p = 0.05),what could be due to therapy or to a phenotypic change of the metastatic nodule. Analysis of the prognostic influence of PLK1 expression showed significant association between PLK1 expression in metastasis and lower overall survival (p = 0.000). We have also found a significant association between PLK1 expression and histopathological response (p = 0.02). All the tumors with high expression of PLK1 showed minor response (11/11). This study shows the association between survival and poor histopathological response to therapy and high expression of PLK1 in metastasis. Our results could open a new therapeutic approach through the inhibition of PLK1. C1 [Fernandez-Acenero, Jesus Maria] Hospital Clinico San Carlos, Department of Surgical Pathology, C/ Profesor Martin Lagos s/n, 28040 Madrid, Spain. [Cortes, Delia] University Hospital Fundacion Jimenez Diaz, Health Research Institute FJD-UAM, Department of SurgeryMadrid, Spain. [Gomez del Pulgar, Teresa] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology DivisionMadrid, Spain. [Cebrian, Arancha] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology DivisionMadrid, Spain. [Estrada, Lourdes] Hospital Clinico San Carlos, Department of Surgical Pathology, C/ Profesor Martin Lagos s/n, 28040 Madrid, Spain. [Martinez-Useros, Javier] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology DivisionMadrid, Spain. [Celdran, Angel] University Hospital Fundacion Jimenez Diaz, Health Research Institute FJD-UAM, Department of SurgeryMadrid, Spain. [Garcia-Foncillas, Jesus] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology DivisionMadrid, Spain. [Pastor, Carlos] University Hospital Fundacion Jimenez Diaz, Health Research Institute FJD-UAM, Department of SurgeryMadrid, Spain. RP Fernandez-Acenero, JM (reprint author), Hospital Clinico San Carlos, Department of Surgical Pathology, 28040 Madrid, Spain. EM mgg10167@gmail.com CR Siegel R, Ward E, Brawley O, Jemal A, 2011, Cancer statistics 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 61:212–236 Ribero D, Wang H, Donadon M, et al., 2007, Bevacizumab improves pathologic response and protects against hepatic injury in patients treated with oxaliplatin-based chemotherapy for colorectal liver metastases. Cancer 110:2761–2767 Pawlik TM, Scoggins CR, Zorzi D, et al., 2005, Effect of surgical margin status on survival and site of recurrence after hepatic resection for colorectal metastases. Ann Surg 241:715–722 Benoist S, BrouquetA, Penna C, et al., 2006, Complete response of colorectal liver metastases after chemotherapy: does it mean cure? J Clin Oncol 24:3939–3945 Rubbia-Brandt L, Giostra E, Brezault C, et al., 2007, Importance of histological tumor response assessment in predicting the outcome in patients with colorectal liver metastases treated with neoadjuvant chemotherapy followed by liver surgery. Ann Oncol 18:299–304 Blazer DG, Kishi J, Maru DM, et al., 2008, Pathologic response to preoperative chemotherapy: a new outcome end point after resection of hepatic colorectal metastases. J Clin Oncol 26:5344–5351 Benada J, Burdov K, Lidak T, von Morgen P, Macurek L, 2015, Polo-like kinase 1 inhibits DNA damage response during mitosis. Cell Cycle 14:219–231 Zhang Z, Zhang G, Kong C, 2013, High expression of polo-like kinase 1 is associated with the metastasis and recurrence in urothelial carcinoma of bladder. Urol Oncol 31:1222–1230 Zhang G, Zhang Z, Liu Z, 2013, Polo-like kinase 1 is overexpressed in renal cancer and participates in the proliferation and invasion of renal cancer cells. Tumour Biol 34:1887–1894 Maire V, Nemati F, Richardson M, et al., 2013, Polo-like kinase 1: a potential therapeutic option in combination with conventional chemotherapy for the management of patients with triple-negative breast cancer. Cancer Res 73:813–823 Ackermann S, Goeser F, Schulte JH, et al., 2011, Polo-like kinase 1 is a therapeutic target in high-risk neuroblastoma. Clin Cancer Res 17:731–741 Pellegrino R, Calvisi DF, Ladu S, et al., 2010, Oncogenic and tumor suppressive roles of polo-like kinases in human hepatocellular carcinoma. Hepatology 51:857–868 Zhang Y, Liu Y, Yang YX, et al., 2009, The expression of PLK-1 in cervical carcinoma: a possible target for enhancing chemosensitivity. J Exp Clin Cancer Res 28:130–138 WaneboHJ, LeGolvan M, Paty PB, et al., 2012, Meeting the biologic challenge of colorectal metastases. Clin Exp Metastasis 29:821–839 Rothe JH, Grieser C, Lehmkuhl L, et al., 2013, Size determination and response assessment of liver metastases with computed tomography– comparison of RECIST and volumetric algorithms. Eur J Radiol 82:1831–1839 Shindoh J, Loyer EM, Kopetz S, et al., 2012, Optimal morphologic response to preoperative chemotherapy: an alternate outcome end point before resection of hepatic colorectal metastases. J Clin Oncol. 30:4566–4572 ChunYS,Vauthey JN, Boonsirikamchai P, et al., 2009)Association of computed tomography morphologic criteria with pathologic response and survival in patients treated with bevacizumab for colorectal liver metastases. J Am Med Assoc 302:2338–2344 van Vugt MA, Gardino AK, Linding R, et al., 2010, A mitotic phosphorylation feedback network connects Cdk1, Plk1, 53BP1, and Chk2 to inactivate the G2/M DNA damage checkpoint. PLoS Biol 8:e1000287 Chen S, Bartkovitz D, Cai J, et al., 2012, Identification of novel, potent and selective inhibitors of polo-like kinase 1. Bioorg Med Chem Lett 22:1247–1250 Berg T, Bug G,Ottmann OG, Strebhardt K, 2012, Polo-like kinases in AML. Expert Opin Investig Drugs 21:1069–1074 StadlerWM, Vaughn DJ, Sonpavde G, et al., 2014, An open-label, single-arm, phase 2 trial of the polo-like kinase inhibitor volasertib, BI6727, in patients with locally advanced or metastatic urothelial cancer. Cancer 120:976–982 Lin CC, SuWC, Yen CJ, et al., 2014, A phase I study of two dosing schedules of volasertib, BI6727), an intravenous polo-like kinase inhibitor, in patients with advanced solid malignancies. Br J Cancer 110:2434–2440 Bowles DW, Diamond JR, Lam ET, et al., 2014, Phase I study of oral rigosertib, ON 01910. Na), a dual inhibitor of the PI3K and PLK1 pathways, in adult patients with advanced solid malignancies. Clin Cancer Res 20:1656–1665 Wan NN, Li ZH, Zhao H, et al., 2015, Molecular targeting of the oncoprotein PLK1 in pediatric acute myeloid leukaemia: RO3280, a novel PLK1 inhibitor, induces apoptosis in leukaemia cells. Int J Mol Sci 16:1266–1292 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 377 EP 383 DI 10.1007/s12253-015-0015-8 PG 7 ER PT J AU Rakesh, N Iyengar, A Majumdar, K Vidya, ShG Shantha Kumar, SS AF Rakesh, Nagaraju Iyengar, Asha Majumdar, Kuhu Vidya, Shankar Gurram Shantha Kumar, S S TI Quantitative Evaluation of Tumour - Associated Tissue Eosinophilia and Cyclo-oxegenase-2 Gene in Oral Cancer Patients with Assessment of Long Term Outcomes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral squamous cell carcinoma; Cyclooxygenase-2; COX-2; Tumour associated tissue eosinophilia; TATE; Locoregional recurrence ID Oral squamous cell carcinoma; Cyclooxygenase-2; COX-2; Tumour associated tissue eosinophilia; TATE; Locoregional recurrence AB Various histopathological parameters have been extensively studied for prognostication of oral cancer but the focus is now getting diverted towards the role of inflammatory mediators in cancer progression. The present study was undertaken to evaluate two such components of the inflammatory milieu, tumor-associated tissue eosinophilia (TATE) as well as Cyclo-oxygenase-2 (COX-2) gene expression, quantitatively in oral squamous cell carcinoma (OSCC) patients in relation to treatment outcomes and patterns of recurrence. A total of forty five patients with primary OSCC matching our inclusion criteria were selected for the study and followed up over a five year period. TATE was evaluated from the invasive front of the tumor using Haematoxylin and eosin (H & E) stained sections of histopathological specimens and graded as mild, moderate or intense. COX-2 gene expression was obtained from specimens using the reverse transcriptase - polymerase chain reaction (RT-PCR) method. A statistically significant association was observed between degree of TATE and locoregional recurrence (P < 0.001). The expression of COX-2 gene ranged from 0.4326 to 0.9998 and a highermean COX-2 score was recorded in samples with intense degree of TATE followed by moderate and mild TATE. (P < 0.001). Using the t-test, the difference in mean COX-2 was found to be statistically significant (P < 0.001) between patients who developed locoregional recurrence and those who did not. The analysis of TATE may provide an indication of future recurrence at the time of diagnosis of OSCC. Also, the increased expression of COX-2 gene in OSCC strongly suggests its possible use as a chemopreventive/chemotherapeutic target. C1 [Rakesh, Nagaraju] M.S. Ramaiah Dental College & Hospital, Department of Oral Medicine, Diagnosis and Radiology, MSRIT Post, New BEL RoadBangalore, India. [Iyengar, Asha] M.S. Ramaiah Dental College & Hospital, Department of Oral Medicine, Diagnosis and RadiologyBangalore, India. [Majumdar, Kuhu] M.S. Ramaiah Dental College & Hospital, Department of Oral Medicine, Diagnosis and Radiology, MSRIT Post, New BEL RoadBangalore, India. [Vidya, Shankar Gurram] Ramaiah University of Applied Sciences, Faculty of Dental Sciences, Department of Oral Pathology and MicrobiologyBangalore, India. [Shantha Kumar, S S] M.S. Ramaiah Institute of Technology, Department of BiotechnologyBangalore, India. RP Rakesh, N (reprint author), M.S. Ramaiah Dental College & Hospital, Department of Oral Medicine, Diagnosis and Radiology, Bangalore, India. EM drnrakesh@gmail.com CR Warnakulasuriya S, 2009, Global epidemiology of oral and oropharyngeal cancer. Oral Oncol 45:309–316 Mignogna MD, Fedele S, 2005, Oral cancer screening: 5 minutes to save a life. Lancet 365:1905–1906 Fedele S, 2009, Diagnostic aids in the screening of oral cancer. Head Neck Oncol 1:5., DOI 10.1186/1758-3284-1-5 Massano J, Regateiro FS, Januario G, Ferreira A, 2006, Oral squamous cell carcinoma: review of prognostic and predictive factors. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 102:67–76 Warnakulasuriya S, 2010, Living with oral cancer: epidemiology with particular reference to prevalence and life-style changes that influence survival. Oral Oncol 46(6):407–410 Jerjes W, Upile T, Petrie A, et al., 2010, Clinicopathological parameters, recurrence, locoregional and distant metastasis in 115 T1- T2 oral squamous cell carcinoma patients. Head Neck Oncol. 20:2– 9., DOI 10.1186/1758-3284-2-9 Ford PJ, Farah CS, 2013, Early detection and diagnosis of oral cancer: strategies for improvement. J Cancer Policy 1:e2–e7 Bankfalvi A, Piffko J, 2000, Prognostic and predictive factors in oral cancer: the role of the invasive tumour front. J Oral Pathol Med 29(7):291–298 Woolgar JA, 2000, Histopathological prognosticators in oral and oropharyngeal squamous cell carcinoma. Oral Oncol 42(3):229– 39F Balkwill F, Mantovani A, 2001, Inflammation and cancer: back to Virchow? Lancet 357:539–545 MignognaMD, Fedele S, Lo Russo L, LoMuzio L, Bucci E, 2004, Immune activation and chronic inflammation as the cause of malignancy in oral lichen planus: is there any evidence? Oral Oncol 40(2):120–130 Tepper RI, Coffman RL, Leder P, 1992, An eosinophil dependent mechanism for the antitumor effect of interleukin- 4. Science 257(5069):548–551 Astigiano S, Morandi B, Costa R, et al., 2005, Eosinophil granulocytes account for indoleamine 2,3-dioxygenase-mediated immune escape in human non-small cell lung cancer. Neoplasia 7(4):390–396 Fernandez-Acenero MJ, Galindo-GallegoM SJ, Aljama A, 2000, Prognostic influence of tumor-associated eosinophilic infiltrate in colorectal carcinoma. Cancer 88(7):1544–1548 Kapp DS, LiVolsi VA, 1983, Intense eosinophilic stromal infiltration in carcinoma of the uterine cervix: a clinicopathologic study of 14 cases. Gynecol Oncol 16(1):19–30 Cuschieri A, Talbot IC,Weeden S, 2002, Influence of pathological tumour variables on long-term survival in resectable gastric cancer. Br J Cancer 86(5):674–679 Pretlow TP, Keith EF, Cryar AK, et al., 1983, Eosinophil infiltration of human colonic carcinomas as a prognostic indicator. Cancer Res 43(6):2997–3000 Spiegel GW, Ashraf M, Brooks JJS, 2002, Eosinophils as a marker for invasion in cervical squamous neoplastic lesions. Int J Gynecol Pathol 21(2):117–124 Quaedvlieg PJF, Creytens DH, Epping GG, et al., 2006, Histopathological characteristics of metastasizing squamous cell carcinoma of the skin and lips. Histopathology 49(3):256–264 Agarwal S,WadhwaN, Gupta G, 2002, Eosinophils as amarker for invasion in cervical squamous neoplastic lesions. Int J Gynecol Pathol 21(2):117–124 Dorta RG, Landman G, Kowalski LP, Lauris JR, Latorre MR, Oliveira DT, 2002, Tumour-associated tissue eosinophilia as a prognostic factor in oral squamous cel l carcinomas. Histopathology 41:152–157 Horiuchi K, Mishima K, Ohsawa M, Sugimura M, Aozasa K, 1993, Prognostic factors for well-differentiated squamous cell carcinoma in the oral cavity with emphasis on immunohistochemical evaluation. J Surg Oncol 53:92–96A Tadbir AA, Ashraf MJ, Sardari Y, 2009, Prognostic significance of stromal eosinophilic infiltration in oral squamous cell carcinoma. J Craniofac Surg. 20:287–289 Ercan I, Cakir B, Basak T, Ozdemir T, Sayin I, Turgut S, 2005, Prognostic significance of stromal eosinophilic infiltration in cancer of the larynx. Otolaryngol Head Neck Surg. 132:869–873 Alrawi SJ, Tan D, Stoler DL, et al., 2005, Tissue eosinophilic infiltration: a useful marker for assessing stromal invasion, survival and locoregional recurrence in head and neck squamous neoplasia. Cancer J 11:217–225 Said M, Wiseman S, Yang J, et al., 2005, Tissue eosinophilia: a morphologic marker for assessing stromal invasion in laryngeal squamous neoplasms. BMC Clin Pathol 5:1–8 Sassler AM, McClatchey KD, Wolf GT, Fisher SG, 1995, Eosinophilic infiltration in advanced laryngeal squamous cell carcinoma. Laryngoscope 105:413–416 Lowe D, Fletcher CDM, 1984, Eosinophilia in squamous cell carcinoma of the oral cavity, external genitalia and anusclinical correlations. Histopathology 8(4):627–632 Falconieri G, Luna MA, Pizzolitto S, DeMaglio G, Angione V, Rocco M, 2008, Eosinophil-rich squamous carcinoma of the oral cavity: a study of 13 cases and delineation of a possible new microscopic entity. Ann Diagn Pathol 12:322–327 Goldsmith MM, Belchis DA, Cresson DH, Merritt WD, Askin FB, 1992, The importance of the eosinophil in head and neck cancer. Otolaryngol Head Neck Surg. 106:27–33 Wong DTW, Bowen SM, Elovic A, Gallagher GT, Weller PF, 1999, Eosinophil ablation and tumor development. Oral Oncol 35:496–501 Oliveira DT, Biassi TP, Faustino SE, Carvalho AL, Landman G, Kowalski LP, 2012, Eosinophils may predict occult lymph node metastasis in early oral cancer. Clin Oral Investig 16(6):1523–1528 Davoine F, Sim A, Tang C, et al., 2013, Eosinophils in human oral squamous carcinoma; role of prostaglandin D2. J Inflamm, Lond, 10(1):4 Mohammad S, Ram H, Gupta PN, Husain N, Bhatt MLB, 2011, Overexpression of COX-2 in oral squamous cell carcinoma patients undergoing chemoradiotherapy. Natl J Maxillofac Surg 2(1):17–21 Itoh S, Matsui K, Furuta I, Takano Y, 2003, Immunohistochemical study on over expression of cyclooxygenase-2 in squamous cell carcinoma of the oral cavity: its importance as a prognostic predictor. Oral Oncol 39:829–835 PandeyM, Prakash O, SanthiWS, Soumithran S, Pillai RM, 2008, Overexpression of COX-2 gene in oral cancer is independent of stage of disease and degree of differentiation. Int J Oral Maxillofac Surg 37:379–383 Sharma M, Sah P, Sharma SS, Radhakrishnan R, 2013, Molecular changes in invasive front of oral cancer. J Oral Maxillofac Pathol 17(2):240–247 Oliveira DT, Tjioe KC, Assao A, et al., 2009, Tissue eosinophilia and its association with tumoral invasion of oral cancer. Int J Surg Pathol 17(3):244–249 Lorena SC, Dorta RG, Landman G, Nonogaki S, Oliveira DT, 2003)Morphometric analysis of the tumor associated tissue eosinophilia in the oral squamous cell carcinoma using different staining techniques. Histol Histopathol 18(3):709–713 Tortora G, Caputo R, Damiano V, et al., 2003, Combination of a selective COX-2 inhibitor with epidermal growth factor receptor, tyrosine kinase inhibitor ZD1839 and protein kinase A antisense causes cooperative antitumor and antiangiogenic effect. Clin Cancer Res 9:1566–1572 Kao J, Genden EM, Chen CT, et al., 2011, Phase 1 trial of concurrent erlotinib, celecoxib, and reirradiation for recurrent head and neck cancer. Cancer 117(14):3173–3181 Uddin MJ, Crews BC, Ghebreselasie K, et al., 2011, Fluorinated COX-2 inhibitors as agents in PET imaging of inflammation and cancer. Cancer Prev Res, Phila, 4(10):1536–1545 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 385 EP 392 DI 10.1007/s12253-015-0016-7 PG 8 ER PT J AU Wang, DY Zou, LP Liu, XJ Zhu, HG Zhu, R AF Wang, Di-Yi Zou, Li-Ping Liu, Xiao-Jia Zhu, Hong-Guang Zhu, Rong TI Chemokine Expression Profiles of Human Hepatoma Cell Lines Mediated by Hepatitis B Virus X Protein SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatitis B virus X protein; Chemokine antibody array; Chemokines; Interleukin-8 ID Hepatitis B virus X protein; Chemokine antibody array; Chemokines; Interleukin-8 AB The hepatitis B virus X protein (HBx), which is encoded by hepatitis B virus (HBV), plays crucial roles in the tumorigenesis of HBV associated hepatocellular carcinoma (HCC). Recent studies suggest that the HBx is involved in regulation of host immune cytokines and chemokines in HBV-associated HCC patients. However, effects of the HBx on autocrine chemokine expression profiles of hepatoma cells, which were shown in modulation of tumor-immune cell interactions, have not been investigated comprehensively. In the present study, human hepatoma cell lines SMMC-7721 and HepG2 were transfected with HBx-expressing plasmid. Human chemokine antibody array 1 (RayBio®), which simultaneously detects 38 chemokine factors, was used to determine chemokine expression profiles. Real-time polymerase chain reaction (real-time PCR) was used to further confirm the differential expression of chemokines. Chemokine antibody array revealed that all 38 chomekines were found to be expressed by SMMC-7721 and HepG2 cell lines. Interleukin-8 (IL-8) was obviously up-regulated, and epithelial neutrophil-activating protein 78 (ENA78), eosinophil chemotactic protein-1 (Eotaxin-1), monocyte chemotactic protein-1 (MCP-1), MCP-2, MCP-3 and macrophage inflammatory protein-3β (MIP-3β) were significantly declined in both cell lines following transfection of HBx-expressing plasmid. Other chemokines showed little or no significant changes. HBx-induced differential chemokine expression levels were validated by real-time PCR. Hierarchical cluster analysis identified a distinction of chomekine expression profiles between HBX-expressing hepatoma cell lines and controls. Our findings provide new evidence that HBx is able to selectively regulate chomekines in hepatoma cells that may be involved in the regulation of tumor-immune cell interactions. C1 [Wang, Di-Yi] Affiliated Hospital of Taishan Medical University, Department of Pathology, 271000 Taian, China. [Zou, Li-Ping] Fudan University, Huashan Hospital, Department of Pathology, 200040 Shanghai, China. [Liu, Xiao-Jia] Fudan University, Shanghai Medical College, Department of Oncology, 200032 Shanghai, China. [Zhu, Hong-Guang] Fudan University, Shanghai Medical College, Department of Oncology, 200032 Shanghai, China. [Zhu, Rong] Fudan University, Shanghai Medical College, Department of Oncology, 200032 Shanghai, China. RP Wang, DY (reprint author), Affiliated Hospital of Taishan Medical University, Department of Pathology, 271000 Taian, China. EM xilisa@163.com CR Bosch FX, Ribes J, Diaz M, Cleries R, 2004, Primary liver cancer: worldwide incidence and trends. Gastroenterology 127:S5–S16 Chen L, Zhao H, Yang X, Gao JY, Cheng J, 2014, HBsAg-negative hepatitis B virus infection and hepatocellular carcinoma. Discov Med 18:189–193 Andrisani OM, 2013, Deregulation of epigenetic mechanisms by the hepatitis B virus X protein in hepatocarcinogenesis. Viruses 5: 858–872 Almajhdi FN, Al-Qudari AY, Hussain Z, 2013, Differential expression of transforming growth factor-β1 and HBx enhances hepatitis B virus replication and augments host immune cytokines and chemokines. Ann Hepatol 12:408–415 Cho HK, Kim SY, Seong JK, Cheong J, 2014, Hepatitis B virus X increases immune cell recruitment by induction of chemokine SDF- 1. FEBS Lett 588:733–739 Johrer K, Pleyer L, Olivier A, Maizner E, Zelle-Rieser C, Greil R, 2008, Tumour-immune cell interactions modulated by chemokines. Expert Opin Biol Ther 8:269–290 O’Hayre M, Salanga CL, Handel TM, Allen SJ, 2008, Chemokines and cancer: migration, intracellular signalling and intercellular communication in the microenvironment. Biochem J 409:635–649 Lopez-Lago MA, Posner S, Thodima VJ, Molina AM, Motzer RJ, Chaganti RS, 2013, Neutrophil chemokines secreted by tumor cells mount a lung antimetastatic response during renal cell carcinoma progression. Oncogene 32:1752–1760 Roth MD, Harui A, 2015, Human tumor infiltrating lymphocytes cooperatively regulate prostate tumor growth in a humanized mouse model. J Immunother Cancer 3:12 Verbeke H, Geboes K, Van Damme J, Struyf S, 2012, The role of CXC chemokines in the transition of chronic inflammation to esophageal and gastric cancer. Biochim Biophys Acta 1825:117– 129 Ben-Baruch A, 2006, The multifaceted roles of chemokines in malignancy. Cancer Metastasis Rev 25:357–371 Park MH, Lee JS, Yoon JH, 2012, High expression of CX3CL1 by tumor cells correlates with a good prognosis and increased tumorinfiltrating CD8+ T cells, natural killer cells, and dendritic cells in breast carcinoma. J Surg Oncol 106:386–392 Hirano S, Iwashita Y, Sasaki A, Kai S, Ohta M, Kitano S, 2007, Increased mRNA expression of chemokines in hepatocellular carcinoma with tumor-infiltrating lymphocytes. J Gastroenterol Hepatol 22:690–696 Pollicino T, Bellinghieri L, Restuccia A, Raffa G, Musolino C, Alibrandi A, et al., 2013, Hepatitis B virus, HBV, induces the expression of interleukin-8 that in turn reduces HBV sensitivity to interferon-alpha. Virology 444:317–328 Chen HY, Tang NH, Li XJ, Zhang SJ, Chen ZX, Wang XZ, 2004, Transfection and expression of hepatitis B virus x gene and its effect on apoptosis in HL-7702 cells. World J Gastroenterol 10:959–964 Luo MX, Wong SH, Chan MT, Yu L, Yu SS, Wu F, et al., 2015, Autophagymediates HBx-induced nuclear factor-κB activation and release of IL-6, IL-8, and CXCL2 in hepatocytes. J Cell Physiol 230:2382–2389 Sun LF, Shi C, Yuan L, Sun Y, Yao XX, Ma JW, et al., 2013, Expression of cytokines in mouse hepatitis B virus X genetransfected model. J Huazhong Univ Sci Technolog Med Sci 33: 172–177 Han J, Yoo HY, Choi BH, Rho HM(2000, Selective transcriptional regulations in the human liver cell by hepatitis B viral X protein. Biochem Biophys Res Commun 272:525–530 Miyagawa T, Sugaya M, Okada A, Asano Y, Kadono T, Sato S, 2014, Neutrophil-rich anaplastic large cell lymphoma expressing interleukin-8. Acta Derm Venereol 94:741–742 Cougot D, Wu Y, Cairo S, Caramel J, Renard CA, Levy L, et al., 2007, The hepatitis B virus X protein functionally interacts with CREB-binding protein/p300 in the regulation of CREB-mediated transcription. J Biol Chem 282:4277–4287 Ehling J, Tacke F, 2015, Role of chemokine pathways in hepatobiliary cancer. Cancer Lett., DOI 10.1016/j.canlet.2015.06. 017 Li XP, Yang XY, Biskup E, Zhou J, Li HL,Wu YF, et al, 2015, Coexpression of CXCL8 and HIF-1α is associated withmetastasis and poor prognosis in hepatocellular carcinoma. Oncotarget 6:22880– 22889 Perez LE, Parquet N, Meads M, Anasetti C, Dalton W, 2010, Bortezomib restores stroma-mediated APO2L/TRAIL apoptosis resistance in multiple myeloma. Eur J Haematol 84:212–222 Taub DD, Proost P, Murphy WJ, AnverM, Longo DL, van Damme J, et al., 1995, Monocyte chemotactic protein-1, MCP-1), -2, and -3 are chemotactic for human T lymphocytes. J Clin Invest 95:1370– 1376 Wang Q, Shu C, Su J, Li X, 2015, A crosstalk triggered by hypoxia and maintained by MCP-1/miR-98/IL-6/p38 regulatory loop between human aortic smooth muscle cells and macrophages leads to aortic smooth muscle cells apoptosis via Stat1 activation. Int J Clin Exp Pathol 8:2670–2679 Luo KQ, Shi YN, Peng JC, 2014, The effect of chemokine CC motif ligand 19 on the proliferation and migration of hepatocellular carcinoma. Tumour Biol 35:12575–12581 Lu J, Ma J, Cai W, Wangpu X, Feng H, Zhao J, et al., 2015, CC motif chemokine ligand 19 suppressed colorectal cancer in vivo accompanied by an increase in IL-12 and IFN-γ. Biomed Pharmacother 69:374–379 Lotfi R, Lee JJ, Lotze MTJ, 2007, Eosinophilic granulocytes and damage-associated molecular pattern molecules, DAMPs): role in the inflammatory response within tumors. J Immunother 30:16–28 Arenberg DA, Keane MP, DiGiovine B, Kunkel SL, Morris SB, Xue YY, et al., 1998, Epithelial-neutrophil activating peptide, ENA-78, is an important angiogenic factor in non-small cell lung cancer. J Clin Invest 102:465–472 Lim JB, Chung HW, 2015, Serum ENA78/CXCL5, SDF-1/ CXCL12, and their combinations as potential biomarkers for prediction of the presence and distant metastasis of primary gastric cancer. Cytokine 7:16–22 Zhu F, Liu P, Li J, Zhang Y, 2014, Eotaxin-1 promotes prostate cancer cell invasion via activation of the CCR3-ERK pathway and upregulation of MMP-3 expression. Oncol Rep 31:2049–2054 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 393 EP 399 DI 10.1007/s12253-015-0014-9 PG 7 ER PT J AU Patrikidou, A Valeri, MR Kitikidou, K Destouni, Ch Vahtsevanos, K AF Patrikidou, Anna Valeri, Maria Rosalia Kitikidou, Kyriaki Destouni, Charikleia Vahtsevanos, Konstantinos TI Introducing Cytology-Based Theranostics in Oral Squamous Cell Carcinoma: A Pilot Program SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cytology; Biomarker; Oral squamous cell carcinoma; Angiogenesis; Theranostics ID Cytology; Biomarker; Oral squamous cell carcinoma; Angiogenesis; Theranostics AB We aimed to evaluate the feasibility and reliability of brush cytology in the biomarker expression profiling of oral squamous cell carcinomas within the concept of theranostics, and to correlate this biomarker profile with patient measurable outcomes. Markers representative of prognostic gene expression changes in oral squamous cell carcinoma was selected. These markers were also selected to involve pathways for which commercially available or investigational agents exist for clinical application. A set of 7 markers were analysed by immunocytochemistry on the archival primary tumour material of 99 oral squamous cell carcinoma patients. We confirmed the feasibility of the technique for the expression profiling of oral squamous cell carcinomas. Furthermore, our results affirm the prognostic significance of the epidermal growth factor receptor (EGFR) family and the angiogenic pathway in oral squamous cell carcinoma, confirming their interest for targeted therapy. Brush cytology appears feasible and applicable for the expression profiling of oral squamous cell carcinoma within the concept of theranostics, according to sample availability. C1 [Patrikidou, Anna] Institute Gustave Roussy, 114 rue Edouard Vaillant, 94805 Villejuif, France. [Valeri, Maria Rosalia] Theagenio Cancer Hospital, Department of CytopathologyThessaloniki, Greece. [Kitikidou, Kyriaki] Democritus University, School of Agricultural Sciences and Forestry, Department of Forestry and Management of the Environment and Natural ResourcesAlexandroupolis, Greece. [Destouni, Charikleia] Theagenio Cancer Hospital, Department of CytopathologyThessaloniki, Greece. [Vahtsevanos, Konstantinos] Theagenio Cancer Hospital, Department of Oral and Maxillofacial SurgeryThessaloniki, Greece. RP Patrikidou, A (reprint author), Institute Gustave Roussy, 94805 Villejuif, France. EM anna.patrikidou@gustaveroussy.fr CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, 2011, Global cancer statistics, 2008. CA Cancer J Clin 61:69–90 Siegel R, Naishadham N, Jemal A, 2012, Cancer statistics, 2012. CA Cancer J Clin 62:10–29 Funkhouser J, 2002, Reinventing pharma: the theranostic revolution. Current Drugs Discov 2:17–19 Yu YH, Kuo HK, ChangKW(2008, The evolving transcriptome of head and neck squamous cell carcinoma: a systematic review. PLoS One 3:e3215 Oliveira LR, Ribeiro-Silva A, 2011, Prognostic significance of immunohistochemical biomarkers in oral squamous cell carcinoma. Int J Oral Maxillofac Surg 40:298–307 Smith BD, Smith GL, Carter D, Sasaki CT, Haffty BG, 2000, Prognostic significance of vascular endothelial growth factor protein levels in oral and oropharyngeal squamous cell carcinoma. J Clin Oncol 18:2046–2052 Uehara M, Sano K, Ikeda H, Sekine J, Irie A, Yokota T, Tobita T, Ohba S, Inokuchi T, 2004, Expression of vascular endothelial growth factor and prognosis of oral squamous cell carcinoma. Oral Oncol 40:321–325 Kyzas PA, Cunha IW, Ioannidis JP, 2005, Prognostic significance of vascular endothelial growth factor immunohistochemical expression in head and neck squamous cell carcinoma: a meta-analysis. Clin Cancer Res 11:1434–1440 Shao Z, Zhang WF, Chen XM, Shang ZJ, 2008, Expression of EphA2 and VEGF in squamous cell carcinoma of the tongue: correlation with the angiogenesis and clinical outcome. Oral Oncol 44: 1110–1117 Argiris A, Kotsakis AP, Hoang T, Worden FP, Savvides P, Gibson MK, Gyanchandani R, Blumenschein Jr GR, Chen HX, Grandis JR, Harari PM, Kies MS, Kim S, 2013, Cetuximab and bevacizumab: preclinical data and phase II trial in recurrent or metastatic squamous cell carcinoma of the head and neck. Ann Oncol 24:220–225 Redman MW, Crowley JJ, Herbst RS, Hirsch FR, Gandara DR, 2012, Design of a phase III clinical trial with prospective biomarker validation: SWOG S0819. Clin Cancer Res 18:4004–4012 Cohen EE, Davis DW, Karrison TG, Seiwert TY,Wong SJ, Nattam S, Kozloff MF, Clark JI, Yan DH, Liu W, Pierce C, Dancey JE, Stenson K, Blair E, Dekker A, Vokes EE, 2009, Erlotinib and bevacizumab in patients with recurrent or metastatic squamouscell carcinoma of the head and neck: a phase I/II study. Lancet Oncol 10:204–205 Argiris A, KaramouzisMV, GoodingWE, Branstetter BF, Zhong S, Raez LE, Savvides P, Romkes M, 2011, Phase II trial of pemetrexed and bevacizumab in patients with recurrent or metastatic head and neck cancer. J Clin Oncol 29:1140–1145 Nagatsuka H, Hibi K, Gunduz M, Tsujigiwa H, Tamamura R, Sugahara T, Sasaki A, Nagai N, 2005, Various immunostaining patterns of CD31, CD34 and endoglin and their relationship with lymph node metastasis in oral squamous cell carcinomas. J Oral Pathol Med 34:70–76 Fujieda S, Sunaga H, Tsuzuki H, Sugimoto C, Tanaka N, Saito H, 1998, Expression of platelet-derived endothelial cell growth factor in oral and oropharyngeal carcinoma. Clin Cancer Res 4:1583–1590 Barth PJ, Schweinsberg TS, Ramaswamy A,Moll R, 2004, CD34+ fibrocytes, α-smooth muscle antigen-positive myofibroblasts, and CD117 expression in the stroma of invasive squamous cell carcinomas of the oral cavity, pharynx, and larynx. Virchows Arch 444:231–234 Kademani D, Lewis JT, Lamb DH, Rallis DJ, Harrington JR, 2009, Angiogenesis and CD34 expression as a predictor of recurrence in oral squamous cell carcinoma. J Oral Maxillofac Surg 67:1800– 1805 Ascani G, Balercia P, Messi M, Lupi L, Goteri G, Filosa A, Stramazzotti D, Pieramici T, Rubini C, 2005, Angiogenesis in oral squamous cell carcinoma. Acta Otorhinolaryngol Ital 25:13–17 Alcalde RE, TerakadoN,OtsukiK,Matsumura T, 1997)Angiogenesis and expression of platelet-derived endothelial cell growth factor in oral squamous cell carcinoma. Oncology 54:324–328 Xia W, Lau YK, Zhang HZ, Liu AR, Li L, Kiyokawa N, Clayman GL, Katz RL, Hung MC, 1997, Strong correlation between c-erb-2 overexpression and overall patients with oral squamous cell carcinoma. Clin Cancer Res 3:3–9 Xia W, Lau YK, Zhang HZ, Xiao FY, Johnston DA, Liu AR, Li L, Katz RL, Hung MC, 1999, Combination of EGFR, HER- 2/neu and HER-3 is a stronger predictor for the outcome of oral squamous cell carcinoma than any individual family members. Clin Cancer Res 5:4164–4174 Ang KK, Berkey BA, Tu X, Zhang HZ, Katz R, Hammond EH, Fu KK, Milas L, 2002, Impact of epidermal growth factor receptor expression on survival and pattern of relapse in patients with advanced head and neck carcinoma. Cancer Res 62:7350–7356 Kong A, Leboucher P, Leek R, CallejaV,Winter S, Harris A, Parker PJ, Larijani B, 2006, Prognostic value of an activation state marker for epidermal growth factor receptor in tissue microarrays of head and neck cancer. Cancer Res 66:2834–2843 Albuquerque Jr RL, Miguel MC, Costa AL, Souza LB, 2003, Correlation of c-erbB-2 and S-100 expression with the malignancy grading and anatomical site in oral squamous cell carcinoma. Int J Exp Pathol 84:259–265 Ulanovski D, Stern Y, Roizman P, Shpitzer T, Popovtzer A, Feinmesser R, 2004, Expression of EGFR and cerb-B2 as prognostic factors in cancer of the tongue. Oral Oncol 40:532–537 Ekberg T, Nestor M, Engstrom M, Nordgren H,Wester K, Carlsson J, Anniko M, 2005, Expression of EGFR, HER2, HER3, and HER4 in metastatic squamous cell carcinomas of the oral cavity and base of tongue. Int J Oncol 26:1177–1185 Kuropkat C, Venkatesan TK, Caldarelli DD, Panje WR, Hutchinson J, Preisler HD, Coon JS, Werner JA, 2002, Abnormalities of molecular regulators of proliferation and apoptosis in carcinoma of the oral cavity and oropharynx. Auris Nasus Larynx 29:165–174 Angiero F, Sordo RD, Dessy E, Rossi E, Berenzi A, Stefani M, Sidoni A, 2008, Comparative analysis of c-erbB-2, HER-2/neu, in squamous cell carcinoma of the tongue: does over-expression exist? And what is its correlation with traditional diagnostic parameters? J Oral Pathol Med 37:145–150 Freier K, Bosch FX, Flechtenmacher C, Devens F, Benner A, Lichter P, Joos S, Hofele C, 2003, Distinct site-specific oncoprotein overexpression in head and neck squamous cell carcinoma: a tissue microarray analysis. Anticancer Res 23:3971–3977 Erjala K, Sundvall M, Junttila TT, Zhang N, Savisalo M, Mali P, Kulmala J, Pulkkinen J, Grenman R, Elenius K, 2006, Signaling via ErbB2 and ErbB3 associates with resistance and epidermal growth factor receptor, EGFR, amplification with sensitivity to EGFR inhibitor gefitinib in head and neck squamous cell carcinoma cells. Clin Cancer Res 12:4103–4111 OngkekoWM, Altuna X,Weisman RA,Wang-Rodriguez J, 2005, Expression of protein tyrosine kinases in head and neck squamous cell carcinomas. Am J Clin Pathol 124:71–76 Margaritescu C, Pirici D, Simionescu C, Stepan A, 2011, The utility of CD44, CD117 and CD133 in identification of cancer stem cells, CSC, in oral squamous cell carcinomas, OSCC). Romanian J Morphol Embryol 52(3 Suppl):985–993 Chang BW, Kim DH, Kowalski DP, Burleson JA, Son YH,Wilson LD, Haffty BG(2004, Prognostic significance of cyclooxygenase-2 in oropharyngeal squamous cell carcinoma. Clin Cancer Res 10: 1678–1684 Pannone G, Sanguedolce F, De Maria S, Farina E, Lo Muzio L, Serpico R, Emanuelli M, Rubini C, De Rosa G, Staibano S, Macchia L, Bufo P, 2007, Cyclooxygenase isozymes in oral squamous cell carcinoma: a real-time RT-PCR study with clinic pathological correlations. Int J Immunopathol Pharmacol 20:317–324 Atula T, Hedstrom J, Ristimaki A, Finne P, Leivo I, Markkanen- Leppanen M, Haglund C, 2006, Cyclooxygenase-2 expression in squamous cell carcinoma of the oral cavity and pharynx: association to p53 and clinical outcome. Oncol Rep 16:485–490 RyottM, Marklund L,Wangsa D, Elmberger G,Munck-Wikland E, 2011, Cyclooxygenase-2 expression in oral tongue squamous cell carcinoma. J Oral Pathol Med 40:385–389 Lang S, Tiwari S, Andratschke M, Loehr I, Lauffer L, Bergmann C, Mack B, Lebeau A, Moosmann A,Whiteside TL, Zeidler R, 2007, Immune restoration in head and neck cancer patients after in vivo COX-2 inhibition. Cancer Immunol Immunother 56:1645–1652 Saba NF, Hurwitz SJ, Kono SA, Yang CS, Zhao Y, Chen Z, Sica G, Muller S, Moreno-Williams R, et al., 2014, Chemoprevention of head and neck cancer with celecoxib and erlotinib: results of a phase Ib and pharmacokinetic study. Cancer Prev Res 7:283–291 Gross NB, Bauman JE, GoddingWE, DenqW, Thomas SM,Wang L, Chiosea S, Hood BL, Flint MS, Sun M, et al., 2014, Erlotiniberlotinib- sulindac vs placebo: a randomised, double-blind, placebocontrolled window trial in operable head and neck cancer. Clin Cancer Res 20:3289–3298 De May RM, 1996, The Art and Science of Cytopathology. ASCP Press 1st edition, USA, pp 329–330. Navone R, Pentenero M, Gandolfo S, 2011, Liquid-based cytology in oral cavity squamous cell cancer. Curr Opin Otolaryngol Head Neck Surg 19:77–81 Malle D, Valeri RM, Photiou C, Kaplanis K, Andreadis C, Tsavdaridis D, Destouni C, 2005, Significance of immunocytochemical expression of E-cadherin, N-cadherin and CD44 in serous effusions using liquid-based cytology. Acta Cytol 49:11–16 Schwartz JL, Panda S, Beam C, Bach LE, Adami GR, 2008, RNA from brush oral cytology to measure squamous cell carcinoma gene expression. J Oral Pathol Med 37:70–77 Lothaire P, de Azambuja E, Dequanter D, Lalami Y, Sotiriou C, Andry G, Castro Jr G, Awada A, 2006, Molecular markers of head and neck squamous cell carcinoma Promising signs in need of prospective evaluation. Head Neck 28:256–269 Simon RM, Paik S, Hayes DF, 2009, Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst 101:1446–1452 Maltby S, Freeman S, Gold MJ, Baker JHE, Minchinton AI, Gold MR, Roskelley CD, McNagny KM, 2011, Opposing roles for CD34 in B16 melanoma tumor growth alter early stage vasculature and late stage immune cell infiltration. PLoS One 6(4):e18160 Saura C, Baselga J, Herbst R, del Campo J, Marotti M, Tessier J, Collins B, Heymach J, 2009, Antitumor activity of cediranib in patients with metastatic or recurrent head and neck cancer, HNC, or recurrent non-small cell lung cancer, NSCLC). an open-label exploratory study. J Clin Oncol 27(15S):6023 Machiels J, Haddad RI, Fayette J, Licitra L, Tahara M, Vermorken JB, Clement PM,Gauler T, CupissolD,Grau JJ, Guigay J, Caponigro F, De Castro Jr G, De Souza VL, Keilholz U, Del Campo J, Cong X, Ehmrooth E, Cohen EE, 2015, Afatinib versus methotrexate as second-line treatment for patients with recurrent or metastatic squamous-cell carcinoma of the head and neck progressing on or after platinum-based therapy, LUX-Head & Neck 1): an open-label, randomised phase 3 trial. Lancet Oncol 16:583–594 de Souza JA, Davis DW, Zhang Y, Khattri A, Seiwert TY, Aktolga S, Wong SJ, Kozloff MF, Nattam S, Lingen MW, Kunnavakkam R, Stenson KM, Blair EA, Bozeman J, Dancey JE,Vokes EE, Cohen EE, 2012, A phase II study of lapatinib in recurrent/metastatic squamous cell carcinoma of the head and neck. Clin Cancer Res 18:2336–2243 Vermorken JB, Peyrade F, Krauss J, Mesia R, Remenar E, Gauler TC, Keilholz U, Delord JP, Schafhausen P, Erfan J, Brummendorf TH, Iglesias L, Bethe U, Hicking C, Clement PM, 2014, Cisplatin, 5-fluorouracil, and cetuximab, PFE, with or without cilengitide in recurrent/metastatic squamous cell carcinoma of the head and neck: results of the randomized phase I/II ADVANTAGE trial, phase II part). Ann Oncol 25:682–688 Fruh M, Pless M, 2012, EGFR IHC score for selection of cetuximab treatment: ready for clinical practice? Transl Lung Cancer Res 1:145–146 Lynch T, Bhagavatheeswaran P, Mukhopadyay P, 2011, A retrospective subgroup analysis of EGFR immunohistochemistry expression by histo-score correlated to outcomes from the BMS099 1st line phase III NSCLC trial of cetuximab plus carboplatin/taxane. Eur J Cancer 47:S591–S592 Han XJ, Sun LF, Nishiyama Y, Feng B, Michiue H, Seno M, Matsui H, Tomizawa K, 2013, Theranostic protein targeting ErbB2 for bioluminescence imaging and therapy for cancer. PLoS One 8:e75288 Muthu MS, Tai Leong D, Mei L, Feng SS, 2014, Nanotheranostics application and further development of nanomedicine strategies for advanced theranostics. Theranostics 4:660–677 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 401 EP 411 DI 10.1007/s12253-015-0017-6 PG 11 ER PT J AU Yip, KW He, YP Maizaton, AA Yusoff, S Seow, FH AF Yip, Kien Wai He, Yuan Pei Maizaton, Atmadini Abdullah Yusoff, Suryati Seow, Fong Heng TI Increased Expression of Phosphatidylinositol 3-Kinase p110α and Gene Amplification of PIK3CA in Nasopharyngeal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PIK3CA; Akt; Nasopharyngeal carcinoma; Immunohistochemistry ID PIK3CA; Akt; Nasopharyngeal carcinoma; Immunohistochemistry AB Molecular alterations in PIK3CA oncogene that encodes the p110α catalytic subunit of phosphatidylinositol 3- kinase (PI3K p110α) are commonly found in human cancers. In this study, we examined the expression of PI3K p110α and PIK3CA gene amplification in 74 nasopharyngeal carcinoma (NPC) cases. Immunohistochemical staining demonstrated overexpression of PI3K p110α protein in 44.6 % (33/74) of NPCs and 4.8 % (2/42) of the adjacent normal nasopharyngeal mucosa. Copy number of PIK3CA gene was successfully analyzed in 51 of the total NPC cases and 19 non-malignant nasopharynx tissues by quantitative real-time PCR. Using mean + 2(standard deviation) of copy numbers in the non-malignant nasopharynx tissues as a cutoff value, PIK3CA copy number gain was found in 10 of 51 (19.6 %) NPC cases. High PI3K p110α expression level was correlated with increased PIK3CA copy number (Spearman’s rho =0.324, P = 0.02). PI3K p110α expression and PIK3CA copy number did not associate with Akt phosphorylation, and patient and tumor variables. This study suggests that PI3K p110α overexpression, which is attributed, at least in part, to PIK3CA gene amplification, may contribute to NPC pathogenesis. However, these molecular aberrations may not be responsible for activation of Akt signaling in NPC. C1 [Yip, Kien Wai] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, Serdang, 43400 Selangor, Malaysia. [He, Yuan Pei] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, Serdang, 43400 Selangor, Malaysia. [Maizaton, Atmadini Abdullah] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, Serdang, 43400 Selangor, Malaysia. [Yusoff, Suryati] Hospital Kuala Lumpur, Department of PathologyKuala Lumpur, Malaysia. [Seow, Fong Heng] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, Serdang, 43400 Selangor, Malaysia. RP Seow, FH (reprint author), Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, 43400 Selangor, Malaysia. EM shf@upm.edu.my CR Altomare DA, Testa JR, 2005, Perturbations of the AKT signaling pathway in human cancer. Oncogene 24(50):7455–7464 Vivanco I, Sawyers CL, 2002, The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat Rev Cancer 2(7):489–501 Bruhn MA, Pearson RB, Hannan RD, Sheppard KE, 2013, AKTindependent PI3-K signaling in cancer - emerging role for SGK3. Cancer Manag Res 5:281–292 Samuels Y,Waldman T, 2010, Oncogenic mutations of PIK3CA in human cancers. Curr Top Microbiol Immunol 347:21–41 Kang S, Bader AG, Vogt PK, 2005, Phosphatidylinositol 3-kinase mutations identified in human cancer are oncogenic. Proc Natl Acad Sci U S A 102(3):802–807 Samuels Y, Diaz Jr LA, Schmidt-Kittler O, Cummins JM, Delong L, Cheong I, Rago C, Huso DL, Lengauer C, Kinzler KW, Vogelstein B, Velculescu VE, 2005, Mutant PIK3CA promotes cell growth and invasion of human cancer cells. Cancer Cell 7(6):561– 573 Engelman JA, 2009, Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer 9(8):550–562 Shayesteh L, Lu Y, Kuo WL, Baldocchi R, Godfrey T, Collins C, Pinkel D, Powell B, Mills GB, Gray JW, 1999, PIK3CA is implicated as an oncogene in ovarian cancer. Nat Genet 21(1):99–102 Wu G, Xing M, Mambo E, Huang X, Liu J, Guo Z, Chatterjee A, Goldenberg D, Gollin SM, Sukumar S, Trink B, Sidransky D, 2005, Somatic mutation and gain of copy number of PIK3CA in human breast cancer. Breast Cancer Res 7(5):R609–R616 Bertelsen BI, Steine SJ, Sandvei R, Molven A, Laerum OD, 2006, Molecular analysis of the PI3K-AKT pathway in uterine cervical neoplasia: frequent PIK3CA amplification and AKT phosphorylation. Int J Cancer 118(8):1877–1883 Chang ET, Adami HO, 2006, The enigmatic epidemiology of nasopharyngeal carcinoma. Cancer Epidemiol Biomark Prev 15(10): 1765–1777 Yip WK, Leong VC, Abdullah MA, Yusoff S, Seow HF, 2008, Overexpression of phospho-Akt correlates with phosphorylation of EGF receptor, FKHR and BAD in nasopharyngeal carcinoma. Oncol Rep 19(2):319–328 Yip WK, Seow HF, 2012, Activation of phosphatidylinositol 3- kinase/Akt signaling by EGF downregulates membranous Ecadherin and beta-catenin and enhances invasion in nasopharyngeal carcinoma cells. Cancer Lett 318(2):162–172 Chou CC, Chou MJ, Tzen CY, 2009, PIK3CA mutation occurs in nasopharyngeal carcinoma but does not significantly influence the disease-specific survival. Med Oncol 26(3):322–326 Liu P, Li DJ, Qin HD, Zhang RH, Chen LZ, Zeng YX, 2007, Screening for mutations in the hotspot mutation regions of PIK3CA gene in nasopharyngeal carcinoma. Ai Zheng 26(1): 15–20 Or YY, Hui AB, To KF, Lam CN, Lo KW, 2006, PIK3CA mutations in nasopharyngeal carcinoma. Int J Cancer 118(4):1065–1067 Fendri A, Khabir A, Mnejja W, Sellami-Boudawara T, Daoud J, Frikha M, Ghorbel A, Gargouri A, Mokdad-Gargouri R, 2009, PIK3CA amplification is predictive of poor prognosis in Tunisian patients with nasopharyngeal carcinoma. Cancer Sci 100(11):2034–2039 Mao C, Zhou J, Yang Z, Huang Y,Wu X, Shen H, Tang J, Chen Q, 2012, KRAS, BRAF and PIK3CA mutations and the loss of PTEN expression in Chinese patients with colorectal cancer. PLoS One 7(5):e36653 Yip WK, Choo CW, Leong VC, Leong PP, Jabar MF, Seow HF, 2013, Molecular alterations of ras-Raf-mitogen-activated protein kinase and phosphatidylinositol 3-kinase-Akt signaling pathways in colorectal cancers from a tertiary hospital at Kuala Lumpur, Malaysia. APMIS 121(10):954–966 Hou P, Liu D, Shan Y, Hu S, Studeman K, Condouris S, Wang Y, Trink A, El-Naggar AK, Tallini G,VaskoV, XingM(2007, Genetic alterations and their relationship in the phosphatidylinositol 3-kinase/Akt pathway in thyroid cancer. Clin Cancer Res 13(4): 1161–1170 Aleskandarany MA, Rakha EA, Ahmed MA, Powe DG, Paish EC, Macmillan RD, Ellis IO, Green AR, 2010, PIK3CA expression in invasive breast cancer: a biomarker of poor prognosis. Breast Cancer Res Treat 122(1):45–53 Jehan Z, Bavi P, SultanaM, Abubaker J, Bu R, Hussain A, Alsbeih G, Al-Sanea N, Abduljabbar A, Ashari LH, Alhomoud S, Al-Dayel F, Uddin S, Al-Kuraya KS, 2009, Frequent PIK3CA gene amplification and its clinical significance in colorectal cancer. J Pathol 219(3):337–346 Woenckhaus J, Steger K, Werner E, Fenic I, Gamerdinger U, Dreyer T, Stahl U, 2002, Genomic gain of PIK3CA and increased expression of p110alpha are associated with progression of dysplasia into invasive squamous cell carcinoma. J Pathol 198(3):335– 342 Akagi I, Miyashita M, Makino H, Nomura T, Hagiwara N, Takahashi K, Cho K, Mishima T, Ishibashi O, Ushijima T, Takizawa T, Tajiri T, 2009, Overexpression of PIK3CA is associated with lymph node metastasis in esophageal squamous cell carcinoma. Int J Oncol 34(3):767–775 Abubaker J, Bavi P, Al-HaqawiW, Jehan Z, Munkarah A, Uddin S, Al-Kuraya KS, 2009, PIK3CA alterations in middle eastern ovarian cancers. Mol Cancer 8:51 Woenckhaus J, Steger K, Sturm K, Munstedt K, Franke FE, Fenic I, 2007, Prognostic value of PIK3CA and phosphorylated AKT expression in ovarian cancer. Virchows Arch 450(4):387–395 LinY, JiangX,ShenY, LiM,MaH,XingM,Lu Y, 2009)Frequent mutations and amplifications of the PIK3CA gene in pituitary tumors. Endocr Relat Cancer 16(1):301–310 Astanehe A, Finkbeiner MR, Hojabrpour P, To K, Fotovati A, Shadeo A, Stratford AL, Lam WL, Berquin IM, Duronio V, Dunn SE, 2009, The transcriptional induction of PIK3CA in tumor cells is dependent on the oncoprotein Y-box binding protein-1. Oncogene 28(25):2406–2418 Kok K, Geering B, Vanhaesebroeck B, 2009, Regulation of phosphoinositide 3-kinase expression in health and disease. Trends Biochem Sci 34(3):115–127 Carden CP, Stewart A, Thavasu P, Kipps E, Pope L, Crespo M, Miranda S, Attard G, Garrett MD, Clarke PA, Workman P, de Bono JS, Gore M, Kaye SB, Banerji U, 2012, The association of PI3 kinase signaling and chemoresistance in advanced ovarian cancer. Mol Cancer Ther 11(7):1609–1617 Fenic I, Steger K, Gruber C, Arens C, Woenckhaus J, 2007, Analysis of PIK3CA and Akt/protein kinase B in head and neck squamous cell carcinoma. Oncol Rep 18(1):253–259 Shi J, Yao D, Liu W, Wang N, Lv H, Zhang G, Ji M, Xu L, He N, Shi B, Hou P, 2012, Highly frequent PIK3CA amplification is associated with poor prognosis in gastric cancer. BMC Cancer 12:50 Gasser JA, Inuzuka H, Lau AW, Wei W, Beroukhim R, Toker A, 2014, SGK3 mediates INPP4B-dependent PI3K signaling in breast cancer. Mol Cell 56(4):595–607 Liu T, Sun Q, Li Q, Yang H, Zhang Y, Wang R, Lin X, Xiao D, Yuan Y, Chen L, Wang W, 2014, Dual PI3K/mTOR inhibitors, GSK2126458 and PKI-587, suppress tumor progression and increase radiosensitivity in nasopharyngeal carcinoma. Mol Cancer Ther 14., DOI 10.1158/1535-7163.MCT-1114-0548 Yang F, QianXJ, QinW, Deng R,Wu XQ, Qin J, FengGK, Zhu XF, 2013, Dual phosphoinositide 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 has a therapeutic potential and sensitizes cisplatin in nasopharyngeal carcinoma. PLoS One 8(3): e59879 Jiang H, Gao M, Shen Z, Luo B, Li R, Jiang X, Ding R, HaY,Wang Z, JieW(2014, Blocking PI3K/Akt signaling attenuates metastasis of nasopharyngeal carcinoma cells through induction of mesenchymal-epithelial reverting transition. Oncol Rep 32(2): 559–566 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 413 EP 419 DI 10.1007/s12253-015-0007-8 PG 7 ER PT J AU Vojkovics, D Kellermayer, Z Heidt, D Mihalj, M Kajtar, B Ernszt, D Kovacs, T Nemeth, P Balogh, P AF Vojkovics, Dora Kellermayer, Zoltan Heidt, Diana Mihalj, Martina Kajtar, Bela Ernszt, David Kovacs, Tamas Nemeth, Peter Balogh, Peter TI Isolation and Characterization of a Murine Spontaneous High-Grade Follicular Lymphoma with Restricted In Vivo Spreading – a Model for Lymphatic Metastasis Via the Mesentery SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mouse; Follicular lymphoma; Lymphatics; Model ID Mouse; Follicular lymphoma; Lymphatics; Model AB Spontaneous or induced malignant lymphomas in mice are valuable tools for studying human lymphoproliferative diseases, including the mechanism of migration between peripheral lymphoid organs and positioning within distinct tissue compartments. Here we report the isolation and characterization of a novel spontaneous lymphoma from BALB/c mice showing restricted tissue distribution and metastasis. The lymphoma cells display CD19, B220, MHC II, surface IgG2a/kappa chain with VH7183 rearrangement of the IgH gene, indicating their B-cell origin. Serial intraperitoneal injection of primary tumor into both BALB/c and RAG-1- deficient hosts led to the successful propagation of lymphoma. Despite the cytological characteristics of high-grade follicular B-cell lymphoma, the tumor cells (denoted as Bc-DLFL.1) showed significantly lesser spreading to extraabdominal locations upon intraperitoneal passage compared to splenic and mesenteric lymph node expansion. In mesenteric lymph nodes the high endothelial venules contained only few tumor cells, while the lymphatic vessels were almost completely filled with lymphoma cells. Similarly, the LYVE-1-positive lymphatic capillaries within the mesentery were packed with lymphoma cells. These findings suggest that Bc-DLFL.1 cells likely propagate primarily via the lymphatic circulation within the mesentery, therefore this tumor may offer an in vivo model to investigate the tumor cell migration via the lymphatic circulation from the peritoneal cavity. C1 [Vojkovics, Dora] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7624 Pecs, Hungary. [Kellermayer, Zoltan] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7624 Pecs, Hungary. [Heidt, Diana] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7624 Pecs, Hungary. [Mihalj, Martina] Josip Juraj Strossmayer University of Osijek, School of Medicine, Department of Physiology and ImmunologyOsijek, Croatia. [Kajtar, Bela] University of Pecs, Department of PathologyPecs, Hungary. [Ernszt, David] Pecsi Tudomanyegyetem, Gyogyszereszeti IntezetPecs, Hungary. [Kovacs, Tamas] Pecsi Tudomanyegyetem, Gyogyszereszeti IntezetPecs, Hungary. [Nemeth, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7624 Pecs, Hungary. [Balogh, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, Szigeti ut 12, H-7624 Pecs, Hungary. RP Balogh, P (reprint author), University of Pecs, Faculty of Medicine, Department of Immunology and Biotechnology, H-7624 Pecs, Hungary. EM balogh.peter@pte.hu CR Deshpande AJ, Buske C, Quintanilla-Martinez L, Fend F, 2010, In: Dunphy CH, ed)Molecular oncogenesis in Molecular pathology of hematolymphoid diseases. Springer, New York Dordrecht Heidelberg London, pp. 3–21 Morse HC 3rd , Anver MR, Fredrickson TN, Haines DC, Harris AW, Harris NL, Jaffe ES, Kogan SC, MacLennan IC, Pattengale PK,Ward JM(2002, Hematopathology subcommittee of themouse models of human cancers consortium. Bethesda Proposals for Classification of Lymphoid Neoplasms in Mice Blood 100:246– 258. Donnou S, Galand C, Touitou V, Sautes-Fridman C, Fabry Z, Fisson S, 2012, Murine models of B-cell lymphomas: promising tools for designing cancer therapies. Adv Hematol 2012:701704., DOI 10.1155/2012/701704 Kovalchuk AL, Qi CF, Torrey TA, Taddesse-Heath L, Feigenbaum L, Park SS, Gerbitz A, Klobeck G, Hoertnagel K, Polack A, Bornkamm GW, Janz S, Morse HC 3rd, 2000, Burkitt lymphoma in the mouse. J Exp Med 192:1183–1190 Strasser A, Harris AW, Vaux DL, Webb E, Bath ML, Adams JM, Cory S, 1990, Abnormalities of the immune system induced by dysregulated bcl-2 expression in transgenic mice. Curr Top Microbiol Immunol 166:175–181 Egle A, Harris AW, Bath ML, O’Reilly L, Cory S, 2004, VavPBcl2 transgenic mice develop follicular lymphoma preceded by germinal center hyperplasia. Blood 103:2276–2283 Balogh P, Labadi P, 2011, Structural evolution of the spleen in man andmouse in developmental biology of peripheral lymphoid organs Ed. P. Balogh springer-verlag. Berlin Heidelberg 2011:121–142 Kim HJ, Kammertoens T, Janke M, Schmetzer O, Qin Z, Berek C, Blankenstein T, 2004, Establishment of early lymphoid organ infrastructure in transplanted tumors mediated by local production of lymphotoxin alpha and in the combined absence of functional B and T cells. J Immunol 172:4037–4047 Hopken UE, Rehm A, 2012, Homeostatic chemokines guide lymphoma cells to tumor growth-promoting niches within secondary lymphoid organs. J Mol Med, Berl, 90:1237–1245 Mombaerts P, Iacomini J, Johnson RS, Herrup K, Tonegawa S, Papaioannou VE, 1992, RAG-1-deficient mice have no mature B and T lymphocytes. Cell 68:869–877 Cobaleda C, JochumW, BusslingerM(2007, Conversion of mature B cells into Tcells by dedifferentiation to uncommitted progenitors. Nature 449:473–477 Frith CG, Wiley LD, 1981, Morphologic classification and correlation of incidence of hyperplastic and neoplastic hematopoietic lesions in mice with age. J Gerontol 36:534–545 Bunting KL, Melnick AM, 2013, New effector functions and regulatory mechanisms of BCL6 in normal and malignant lymphocytes. Curr Opin Immunol 25:339–346 Basso K, Dalla-Favera R, 2012, Roles of BCL6 in normal and transformed germinal center B cells. Immunol Rev 247:172–183 Johnston CM, Wood AL, Bolland DJ, Corcoran AE, 2006, Complete sequence assembly and characterization of the C57BL/ 6 mouse Ig heavy chain V region. J Immunol 176:4221–4234 Fu YX, Huang G, Wang Y, Chaplin DD, 1998, B lymphocytes induce the formation of follicular dendritic cell clusters in a lymphotoxin alpha-dependent fashion. J Exp Med 187:1009–1018 Forster R, Mattis AE, Kremmer E,Wolf E, Brem G, LippM(1996, A putative chemokine receptor, BLR1, directs B cell migration to defined lymphoid organs and specific anatomic compartments of the spleen. Cell 87:1037–1047 Ansel KM, Ngo VN, Hyman PL, Luther SA, Forster R, Sedgwick JD, Browning JL, Lipp M, Cyster JG, 2000, A chemokine-driven positive feedback loop organizes lymphoid follicles. Nature 406: 309–314 Rosen SD, 2004, Ligands for L-selectin: homing, inflammation, and beyond. Annu Rev Immunol 22:129–156 Mahadevan A,Welsh IC, Sivakumar A,GludishDW, Shilvock AR, Noden DM, Huss D, Lansford R, Kurpios NA, 2014, The left-right Pitx2 pathway drives organ-specific arterial and lymphatic development in the intestine. Dev Cell 31:690–706 Ansel KM, Harris RB, Cyster JG, 2002, CXCL13 is required for B1 cell homing, natural antibody production, and body cavity immunity. Immunity 16:67–76 Berberich S, Dahne S, Schippers A, Peters T, Muller W, Kremmer E, Forster R, Pabst O, 2008, Differential molecular and anatomical basis for B cell migration into the peritoneal cavity and omental milky spots. J Immunol 180:2196–2203 Gashev AA, 2010, Basic mechanisms controlling lymph transport in the mesenteric lymphatic net. Ann N YAcad Sci 1207(Suppl 1): E16–E20 Moro K, Yamada T, Tanabe M, Takeuchi T, Ikawa T, Kawamoto H, Furusawa J, OhtaniM, Fujii H, Koyasu S, 2010, Innate production of T(H)2 cytokines by adipose tissue-associated c-kit(+)Sca-1(+, lymphoid cells. Nature 463:540–544 Braun A,Worbs T, Moschovakis GL, Halle S, Hoffmann K, Bolter J, Munk A, Forster R, 2011, Afferent lymph-derived T cells and DCs use different chemokine receptor CCR7-dependent routes for entry into the lymph node and intranodal migration. Nat Immunol 12:879–887 Forster R, Braun A,Worbs T, 2012, Lymph node homing of Tcells and dendritic cells via afferent lymphatics. Trends Immunol 33: 271–280 Miller CN, Hartigan-O’Connor DJ, Lee MS, Laidlaw G, Cornelissen IP, Matloubian M, Coughlin SR, McDonald DM, McCune JM, 2013, IL-7 production in murine lymphatic endothelial cells and induction in the setting of peripheral lymphopenia. Int Immunol 25:471–483 Muppidi JR, Schmitz R, Green JA, Xiao W, Larsen AB, Braun SE, An J, Xu Y, Rosenwald A, Ott G, Gascoyne RD, Rimsza LM, Campo E, Jaffe ES, Delabie J, Smeland EB, Braziel RM, Tubbs RR, Cook JR, Weisenburger DD, Chan WC, Vaidehi N, Staudt LM, Cyster JG, 2014, Loss of signalling via Gα13 in germinal Centre B-cell-derived lymphoma. Nature 516:254–258 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 421 EP 430 DI 10.1007/s12253-015-0025-6 PG 10 ER PT J AU Balogh, T Lorincz, T Stiller, I Mandl, J Banhegyi, G Szarka, A AF Balogh, Tibor Lorincz, Tamas Stiller, Ibolya Mandl, Jozsef Banhegyi, Gabor Szarka, Andras TI The Level of ALR is Regulated by the Quantity of Mitochondrial DNA SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Augmenter of liver regeneration; Mitochondrial DNA; ρ0 cells; Regulation ID Augmenter of liver regeneration; Mitochondrial DNA; ρ0 cells; Regulation AB Augmenter of liver regeneration (ALR) contributes to mitochondrial biogenesis, maintenance and to the physiological operation of mitochondria. The depletion of ALR has been widely studied and had serious consequences on the mitochondrial functions. However the inverse direction, the effect of the depletion of mitochondrial electron transfer chain and mtDNA on ALR expression has not been investigated yet. Thus mtDNA depleted, ρ0 cell line was prepared to investigate the role of mitochondrial electron transfer chain and mtDNA on ALR expression. The depletion of mtDNA has not caused any difference at mRNA level, but at protein level the expression of ALR has been markedly increased. The regulatory role of ATP and ROS levels could be ruled out because the treatment of the parental cell line with different respiratory inhibitors and uncoupling agent could not provoke any changes in the protein level of ALR. The effect of mtDNA depletion on the protein level of ALR has been proved not to be liver specific, since the phenomenon could be observed in the case of two other, non-hepatic cell lines. It seems the level of mtDNA and/or its products may have regulatory role on the protein level of ALR. The up-regulation of ALR can be a part of the adaptive response in ρ0 cells that preserves the structural integrity and the transmembrane potential despite the absence of protein components encoded by the mtDNA. C1 [Balogh, Tibor] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, Szent Gellert ter 4, 1111 Budapest, Hungary. [Lorincz, Tamas] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, Szent Gellert ter 4, 1111 Budapest, Hungary. [Stiller, Ibolya] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, 1444 Budapest, Hungary. [Mandl, Jozsef] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, 1444 Budapest, Hungary. [Banhegyi, Gabor] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, 1444 Budapest, Hungary. [Szarka, Andras] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, Szent Gellert ter 4, 1111 Budapest, Hungary. RP Szarka, A (reprint author), Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, 1111 Budapest, Hungary. EM szarka@mail.bme.hu CR Francavilla A, Hagiya M, Porter KA, et al., 1994, Augmenter of liver regeneration: its place in the universe of hepatic growth factors. Hepatology 20:747–757 Hagiya M, Francavilla A, Polimeno L, et al., 1994, Cloning and sequence analysis of the rat augmenter of liver regeneration, ALR, gene: expression of biologically active recombinant ALR and demonstration of tissue distribution. Proc Natl Acad Sci U S A 91: 8142–8146., DOI 10.1073/pnas.92.7.3076a Giorda R, Hagiya M, Seki T, et al., 1996, Analysis of the structure and expression of the augmenter of liver regeneration, ALR, gene. Mol Med 2:97–108 Lisowsky T, Weinstat-Saslow DL, Barton N, et al., 1995, A new human gene located in the PKD1 region of chromosome 16 is a functional homologue to ERV1 of yeast. Genomics 29:690–697 Lisowsky T, 1992, Dual function of a new nuclear gene for oxidative phosphorylation and vegetative growth in yeast. Mol Gen Genet 232:58–64 Lisowsky T, 1994, ERV1 is involved in the cell-division cycle and the maintenance of mitochondrial genomes in Saccharomyces cerevisiae. Curr Genet 26:15–20 Lisowsky T, 1996, Removal of an intron with unique 3′ branch site creates an amino-terminal protein sequence directing the scERV1 gene product to mitochondria. Yeast 12:1501–1510., DOI 10.1002/, SICI)1097-0061(199612)12:15<1501::AID-YEA40>3.0. CO;2-H Lange H, Lisowsky T, Gerber J, et al., 2001, An essential function of the mitochondrial sulfhydryl oxidase Erv1p/ALR in the maturation of cytosolic Fe/S proteins. EMBO Rep 2:715–720 Lisowsky T, Lee JE, Polimeno L, et al., 2001, Mammalian augmenter of liver regeneration protein is a sulfhydryl oxidase. Dig Liver Dis 33:173–180 Mesecke N, Terziyska N, Kozany C, et al., 2005, A disulfide relay system in the intermembrane space of mitochondria that mediates protein import. Cell 121:1059–1069., DOI 10.1016/j. cell.2005.04.011 Dabir DV, Leverich EP, Kim S-K, et al., 2007, A role for cytochrome c and cytochrome c peroxidase in electron shuttling from Erv1. EMBO J 26:4801–4811., DOI 10.1038/sj.emboj.7601909 Bihlmaier K, Mesecke N, Terziyska N, et al., 2007, The disulfide relay system of mitochondria is connected to the respiratory chain. J Cell Biol 179:389–395., DOI 10.1083/jcb.200707123 Glerum DM, Shtanko A, Tzagoloff A, 1996, Characterization of COX17, a yeast gene involved in copper metabolism and assembly of cytochrome oxidase. J Biol Chem 271:14504–14509 Beers J, Glerum DM, Tzagoloff A, 1997, Purification, characterization, and localization of yeast Cox17p, a mitochondrial copper shuttle. J Biol Chem 272:33191–33196 Szarka A, Banhegyi G, 2011, Oxidative folding: recent developments. Biomol Concepts 2:1–12., DOI 10.1515/BMC.2011.038 Di Fonzo A, Ronchi D, Lodi T, et al., 2009, The mitochondrial disulfide relay system protein GFER is mutated in autosomalrecessive myopathy with cataract and combined respiratory-chain deficiency.AmJ HumGenet 84:594–604., DOI 10.1016/j.ajhg.2009. 04.004 Thirunavukkarasu C, Wang LF, Harvey S a K, et al., 2008, Augmenter of liver regeneration: an important intracellular survival factor for hepatocytes. J Hepatol 48:578–588., DOI 10.1016/j.jhep. 2007.12.010 Gandhi CR, Chaillet JR, Nalesnik M a, et al., 2015, Liver-specific deletion of augmenter of liver regeneration accelerates development of steatohepatitis and hepatocellular carcinoma in mice. Gastroenterology 148:379–391.e4., DOI 10.1053/j.gastro.2014.10. 008 Falkenberg M, Larsson N-G, Gustafsson CM, 2007, DNA replication and transcription in mammalian mitochondria. Annu Rev Biochem 76:679–699., DOI 10.1146/annurev.biochem.76.060305. 152028 King MP, Attardi G, 1996, Isolation of human cell lines lacking mitochondrial DNA. Methods Enzymol 264:304–313 Trimmer P a, Keeney PM, BorlandMK, et al., 2004)Mitochondrial abnormalities in cybrid cell models of sporadic Alzheimer’s disease worsen with passage in culture. Neurobiol Dis 15:29–39., DOI 10. 1016/j.nbd.2003.09.011 Dayoub R, Wagner H, Bataille F, et al., 2011, Liver regeneration associated protein, ALR, exhibits antimetastatic potential in hepatocellular carcinoma. Mol Med 17:221–228., DOI 10.2119/molmed. 2010.00117 Gandhi CR, 2012, Augmenter of liver regeneration. Fibrogenesis Tissue Repair 5:10., DOI 10.1186/1755-1536-5-10 Miranda S, Foncea R, Guerrero J, Leighton F, 1999, Oxidative stress and upregulation of mitochondrial biogenesis genes in mitochondrial DNA-depleted HeLa cells. Biochem Biophys Res Commun 258:44–49., DOI 10.1006/bbrc.1999.0580 Boveris A, Chance B, 1973, The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. Biochem J 134:707–716 Lee MS, Kim JY, Park SY, 2004, Resistance of ρ0 cells against apoptosis. Ann N Y Acad Sci 1011:146–153., DOI 10.1196/annals. 1293.015 Holmuhamedov E, Jahangir A, Bienengraeber M, et al., 2003, Deletion of mtDNA disrupts mitochondrial function and structure, but not biogenesis. Mitochondrion 3:13–19., DOI 10.1016/S1567- 7249(03)00053-9 Li K, Neufer PD, Williams RS, 1995, Nuclear responses to depletion of mitochondrial DNA in human cells. Am J Physiol 269: C1265–C1270 Ilowski M, Kleespies A, de Toni EN, Donabauer B, Jauch KW, Hengstler JG, Thasler WE, 2011, Augmenter of liver regeneration, ALR, protects human hepatocytes against apoptosis. Biochem Biophys Res Commun 404(1):148–152 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 431 EP 437 DI 10.1007/s12253-015-0020-y PG 7 ER PT J AU Tian, L Shan, W Zhang, Y Lv, X Li, X Wei, C AF Tian, Lei Shan, Weiyu Zhang, Yufei Lv, Xuejun Li, Xuehua Wei, Caiyun TI Erratum to: Up-Regulation of miR-21 Expression Predicate Advanced Clinicopathological Features and Poor Prognosis in Patients with Non-Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Erratum to: Pathol. Oncol. Res. DOI 10.1007/s12253-015-9979-7. The original article contained an error. The name of the third author should have been Yufei Zhang not Yufei Zhnag. The corrected author name is shown above. C1 [Tian, Lei] No. 88 Hospital of PLA, Department of Pulmonary MedicineTaian, Shandong Province, China. [Shan, Weiyu] No. 88 Hospital of PLA, Department of Pulmonary MedicineTaian, Shandong Province, China. [Zhang, Yufei] No. 88 Hospital of PLA, Department of Pulmonary MedicineTaian, Shandong Province, China. [Lv, Xuejun] No. 88 Hospital of PLA, Department of Pulmonary MedicineTaian, Shandong Province, China. [Li, Xuehua] No. 88 Hospital of PLA, Department of Pulmonary MedicineTaian, Shandong Province, China. [Wei, Caiyun] No. 88 Hospital of PLA, Department of Pulmonary MedicineTaian, Shandong Province, China. RP Tian, L (reprint author), No. 88 Hospital of PLA, Department of Pulmonary Medicine, Taian, China. EM drtianlei@163.com NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 439 EP 439 DI 10.1007/s12253-015-0004-y PG 1 ER PT J AU Wang, Y Wang, X Yang, Z Zhu, G Chen, D Meng, Z AF Wang, Yongzhi Wang, Xinghuan Yang, Zhonghua Zhu, Guangbin Chen, Dong Meng, Zhe TI Erratum to: Menthol Inhibits the Proliferation and Motility of Prostate Cancer DU145 Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Erratum to: Pathol. Oncol. Res. (2012) 18:903–910 DOI 10.1007/s12253-012-9520-1. The original article contained an error. The corrected Acknowledgments section is shown below. Acknowledgments: This study was supported in part by grants from the National Natural Science Foundation of China (grants 81172434 and 81202027) and Fundamental Research Funds for the Central Universities (grant 2012303020211). C1 [Wang, Yongzhi] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. [Wang, Xinghuan] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. [Yang, Zhonghua] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. [Zhu, Guangbin] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. [Chen, Dong] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. [Meng, Zhe] Wuhan University, Zhongnan Hospital, Department of Urology, Number 169, Donghu Road, 430071 Wuhan, Hubei, China. RP Wang, X (reprint author), Wuhan University, Zhongnan Hospital, Department of Urology, 430071 Wuhan, China. EM wangxinghuan@whu.edu.cn NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2016 VL 22 IS 2 BP 441 EP 441 DI 10.1007/s12253-012-9520-1 PG 1 ER PT J AU Vajaria, NB Patel, RK Begum, R Patel, SP AF Vajaria, N Bhairavi Patel, R Kinjal Begum, Rasheedunnisa Patel, S Prabhudas TI Sialylation: an Avenue to Target Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Sialic acid; Sialidase; Sialyltransferase; Sialoproteins; Sialylation ID Sialic acid; Sialidase; Sialyltransferase; Sialoproteins; Sialylation AB Tumorigenesis and metastasis are frequently associated with altered structure and expression of oligosaccharides on cell surface glycoproteins and glycolipids. The expression of sialylated glycoconjugates has been shown to change during development, differentiation, disease and oncogenic transformation. Abnormal sialylation in cancer cell is a distinctive feature associated with malignant properties including invasiveness and metastatic potential. The alterations in sialylation is accompanied by changes in sialic acid, sialidase activity, sialyltransferase (ST) activity or sialoproteins. The present review summarizes the reports on alterations of sialic acid, linkage specific STs and sialoproteins, sialidase activity together with different subtypes of ST and sialidases mRNA expressions in various cancers like lung, breast, oral, cervical, ovarian, pancreatic etc. Sialic acids are widely distributed in nature as terminal sugars of oligosaccharides attached to proteins or lipids. The increase shedding of sialic acid observed in malignant tumors may be due to different types of sialidases. The amount of sialic acid is governed by levels of sialidases and STs. Various types of STs are also involved in formation of different types sialylated tumor associated carbohydrate antigens which plays important role in metastasis. The alterations associated with sialylation aids in early diagnosis, prognosis and post treatment monitoring in various cancers. Recently newer drugs targeting different interplays of sialylation have been developed, which might have profound effect in inhibiting sialylation and thus cancer metastasis and infiltration. C1 [Vajaria, N Bhairavi] The Gujarat Cancer & Research Institute, Department of Cancer Biology, Biochemistry Research Division, 380 016 Ahmedabad, Gujarat, India. [Patel, R Kinjal] The Gujarat Cancer & Research Institute, Department of Cancer Biology, Biochemistry Research Division, 380 016 Ahmedabad, Gujarat, India. [Begum, Rasheedunnisa] The M. S. University of Baroda, Department of BiochemistryVadodara, Gujarat, India. [Patel, S Prabhudas] The Gujarat Cancer & Research Institute, Department of Cancer Biology, Biochemistry Research Division, 380 016 Ahmedabad, Gujarat, India. RP Patel, SP (reprint author), The Gujarat Cancer & Research Institute, Department of Cancer Biology, Biochemistry Research Division, 380 016 Ahmedabad, India. EM prabhudas_p@hotmail.com CR Vajaria BN, Patel KR, Begum R, Patel JB, Shah FD, Shukla SN, Patel PS, 2012, Glycoprotein electrophoretic patterns have potential to monitor changes associated with neoplastic transformation in oral cancer. Int J Biol Markers 27:e247–e256 Tian Y, Estena FJ, Song J, Zhang H, 2012, Altered expression of sialylated glycoproteins in breast cancer using hydrazide chemistry and mass spectrometry. Mol Cell Proteomics 11:M111.011403 Shah MH, Telang SD, Shah PM, Patel PS, 2008, Tissue and serum alpha 2–3- and alpha 2–6-linkage specific sialylation changes in oral carcinogenesis. Glycoconj J 25:279–290 Cylwik B, Chrostek L, Szmitkowski M, 2005, Diagnostic value of total and lipid bound sialic acid in malignancies. Pol Merkur Lekarski 19:237–241 Bose SC, Gokhale PV, Dwivedi S, Singh M, 2013, Quantitative evaluation and correlation of serum glycoconjugates: protein bound hexoses, sialic acid and fucose in leukoplakia, oral sub mucous fibrosis and oral cancer. J Nat Sci Biol Med 4:122–125 YakobM, Fuentes L,WangMB, Abemayor E,Wong DTW(2014, Salivary biomarkers for detection of oral squamous cell carcinoma. Current state and recent advances. Curr Oral Health Rep 1:133–141 Cheng YSL, Rees T, Wright J, 2014, A review of research on salivary biomarkers for oral cancer detection. Clin Transl Med 3: 1–10 Ozturk LK, Emekli-Alturfan E, Kasikcy E, Demir G, Yarat A, 2011, Salivary total sialic acid levels increase in breast cancer patients: a preliminary study. Med Chem 7:443–447 Vajaria BN, Patel KR, Begum R, Patel JB, Shah FD, Joshi GM, Patel PS, 2014, Salivary glyco-sialylation changes monitors oral carcinogenesis. Glycoconj J 31:649–659 Pelak MJ, Jarosz B, Straczynska-Niemiec A, Krawczyk P, Skoczylas P, Pecka KM, Snietura M, Szumilo J, Trojanowski T, 2014, The presence and clinical implications of α-2,6-galactoselinked sialic acids in non-small-cell lung cancer brain metastases - preliminary study. Folia Histochem Cytobiol 52:104–111 Martinez-Duncker I, Salinas-Marin R, Martinez-Duncker C, 2011, Towards invivo imaging of cancer sialylation. Int J Mol Imaging 2011:283497 Lu DY, Lu TR, 2012, Development of antimetastatic drugs by targeting tumor sialic acids. Sci Pharm 80:497–508 Lu DY, Xu J, Lu TR, Wu HY, Xu B, 2013, Inhibition of several antineoplastic drugs on serum sialic acid levels in mice bearing tumors. Sci Pharm 81:223–231 Miyagi T, Yamaguchi K, 2012, Mammalian sialidase: physiological and pathological roles in cellular functions. Glycobiology 22: 880–896 Uemura T, Shiozaki K, Yamaguchi K, Miyazaki S, Satomi S, Kato K, Sakuraba H, Miyagi T, 2009, Contribution of sialidase NEU1 to suppression of metastasis of human Colon cancer cells through desialylation of integrin beta4. Oncogene 28:1218–1229 Silvestri I, Testa F, Zappasodi R, CairoCW, ZhangY, Lupo B, Galli R, Nicola MD, Venerando B, Tringali C, 2014, Sialidase NEU 4 is involved in glioblastoma stem cell survival. Cell Death Dis 5:e1381 O’Shea LK, Abdulkhalek S, Allison S, Neufeld RJ, SzewczukMR, 2014, Therapeutic targeting of Neu1 sialidase with oseltamivir phosphate, Tamiflu, disables cancer cells survival in human pancreatic cancer with acquired chemoresistance. Onco Targets Ther 7: 117–134 Li X, Zhang L, Shao Y, Liang Z, Shao C, Wang B, Guo B, Li N, Zhao X, Li Y, Xu D, 2011, Effects of human plasma membrane associated sialidase siRNA on prostate cancer invasion. Biochem Biophys Res Commun 416:270–276 Tsai CS, Yen HY, Lin MI, Tsai TI, Wang SY, Huang WI, Hsu TL, Cheng YS, Fang JM, Wong CH, 2013, Cell-permeable probe for identification and imaging of human sialidases. Proc Natl Acad Sci U S A 110:2466–2471 Harduin-Lepers A, Krzewinski-Recchi MA, Colomb F, Foulquier F, Groux-Degrootes DP, 2012, Sialyltransferase functions in cancer. Front Biosci 4:499–515 Hauselmann I, Borsig L, 2014, Altered tumor cell-glycosylation promotes metastasis. Front Oncol 4:28 Hakomori S, 2001, Tumor associated carbohydrate antigens defining tumor malignancy: basis for development of anticancer vaccines. Adv Exp Med Biol 491:369–402 Skropeta D, Schworer R, Haag T, Schmidt RR, 2004, Asymmetric synthesis and affinity of potent sialyltransferase inhibitors based on transition-state analogues. Glycoconj J 21:205–219 Chen JY, Tang YA, Huang SM, Juan HF, Wu LW, Sun YC, Wang SC, Wu KW, Balraj G, Chang TT, Li WS, Cheng HC, Wang YC, 2011, A novel sialyltransferase inhibitor suppresses Fak/paxillin signaling and cancer angiogenesis and metastatic pathways. Cancer Res 71:473–483 Nakano T, Matsui T, Ota T, 1996, Benzyl-alpha-GalNAc inhibits sialylation of O-glycosidic sugar chains on CD44 and enhances experimental metastatic capacity in B16BL6 melanoma cells. Anticancer Res 16:3577–3584 Preidl JJ, Gnanapragassam VS, Lisurek M, Saupe J, Horstkorte R, Rademann J, 2014, Fluorescent mimetics of CMP-Neu5Ac are highly potent, cell-permeable polarization probes of eukaryotic and bacterial sialyltransferases and inhibit cellular sialylation. Anqew Chem Int Ed Engl 53:5700–5705 Park JJ, Yi JY, Jin YB, Lee YJ, Lee JS, Lee YS, Ko YG, Lee M, 2012, Sialylation of epidermal growth factor receptor regulates receptor activity and chemosensitivity to gefitinib in colon cancer cells. Biochem Pharmacol 83:849–857 Rillahan CD, Antonopoulos A, Lefort CT, Sonon R, Azadi P, Ley K, Dell A, Haslam SM, Paulson JC, 2012, Global metabolic inhibitors of sialyl- and fucosyltransferases remodel the glycome. Nat Chem Biol 8:661–668 Brown JR, Crawford BE, Esko JD, 2007, Glycan antagonists and inhibitors: a fount for drug discovery. Crit Rev Biochem Mol Biol 42:481–515 Ko K, Furukawa K, Takahashi T, Urano T, Sanai Y, Nagino M, Nimura Y, Furukawa K, 2006, Fundamental study of small interfering RNAs for ganglioside GD3 synthase gene as a therapeutic target of lung cancers. Oncogene 25:6924–6935 Gu Y, Zhang J,MiW, Yang J, Han F, Lu X, YuW(2008, Silencing of GM3 synthase suppresses lung metastasis of murine breast cancer cells. Breast Cancer Res 10:R1 Zhu Y, Srivatana U, Ullah A, Gagneja H, Berenson CS, Lance P, 2001, Suppression of a sialyltransferase by antisense DNA reduces invasiveness of human colon cancer cells in vitro. BiochimBiophys Acta 1536:148–160 Saldova R, Fan Y, Fitzpatrick JM, Watson RW, Rudd PM, 2011, Core fucosylation and alpha2-3 sialylation in serum N-glycome is significantly increased in prostate cancer comparing to benign prostate hyperplasia. Glycobiology 21:195–205 Almaraz RT, Tian Y, Bhattarcharya R, Tan E, Chen SH, Dallas MR, Chen L, Zhang Z, Zhang H, Konstantopoulos K, Yarema KJ, 2012, Metabolic flux increases glycoprotein sialylation: implications for cell adhesion and cancer metastasis. Mol Cell Proteomics 11: M112.017558 Bull C, Boltje TJ, Wassink M, de Graaf AM, van Delft FL, den Brok MH, Adema GJ, 2013, Targeting aberrant sialylation in cancer cells using a fluorinated sialic acid analog impairs adhesion, migration, and in vivo tumor growth. Mol Cancer Ther 12:1935– 1946 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 443 EP 447 DI 10.1007/s12253-015-0033-6 PG 5 ER PT J AU Roncati, L Barbolini, G Piacentini, F Piscioli, F Pusiol, T Maiorana, A AF Roncati, Luca Barbolini, Giuseppe Piacentini, Federico Piscioli, Francesco Pusiol, Teresa Maiorana, Antonio TI Prognostic Factors for Breast Cancer: an Immunomorphological Update SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Prognostic factors; Histology; Immunohistochemistry; Tumor-infiltrating lymphocytes (TILs); Granulysin ID Breast cancer; Prognostic factors; Histology; Immunohistochemistry; Tumor-infiltrating lymphocytes (TILs); Granulysin AB The prognostic variability recorded within homogeneous groups of patients for anatomo-clinical disease stages has led to a more detailed biological characterization of breast cancer. Recently, the attention of the scientific community has focused on the role of tumor-infiltrating lymphocytes (TILs). Therefore, the need of an in-depth immunomorphological characterization of TILs has been emerged. The presence of TILs has been retrospectively investigated in 113 female cases of ductal carcinoma. An immunohistochemical investigation with CD3, CD4, CD8, CD20, CD56, granulysin, perforin-1, granzyme-B and TIA-1 was performed according to the standard procedures on all 17 cases with TILs evidence. TILs consisted of T and B lymphocytes: the prevalent population showed a T immunoprofile with a CD8-immunopositive killer subpopulation (Tk), close-linked to carcinomatous cells, and a CD4-immunopositive helper subpopulation (Th), inside the tumor. A time sequence (firstly T, then B) has been disclosed. Granulysin, perforin, granzyme-B and TIA-1 were expressed by Tk cells. The activated Tk cells secrete these mediators as a result of the binding to the tumor target cell, causing its lytic planned death. The cytotoxicity supported by Tk cells appears an important favorable prognostic factor. Therefore, a graduation system for TILs in breast cancer has been here proposed (absent, non-brisk, brisk). C1 [Roncati, Luca] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical Pathology, I-41124 Modena, MO, Italy. [Barbolini, Giuseppe] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical Pathology, I-41124 Modena, MO, Italy. [Piacentini, Federico] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and of Public Health, Section of OncologyModena, MO, Italy. [Piscioli, Francesco] Provincial Health Care Services, Santa Maria del Carmine Hospital, Institute of PathologyRovereto, TN, Italy. [Pusiol, Teresa] Provincial Health Care Services, Santa Maria del Carmine Hospital, Institute of PathologyRovereto, TN, Italy. [Maiorana, Antonio] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical Pathology, I-41124 Modena, MO, Italy. RP Roncati, L (reprint author), University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical Pathology, I-41124 Modena, Italy. EM emailmedical@gmail.com CR World Health Organization. International Agency for Research on Cancer, 2014, Breast cancer. Chapter 5.2. In:World Cancer Report 362–373 Denoix PF, 1946, Enquete permanent dans les centres antercancereux. Bull Inst Hyg 1:70 Rathore AS, Kumar S, Konwar R, Srivastava AN,Makker A, Goel MM, 2013, Presence of CD3+ tumor infiltrating lymphocytes is significantly associated with good prognosis in infiltrating ductal carcinoma of breast. Indian J Cancer 50:239–244 Loi S,Michiels S, Salgado R, Sirtaine N, Jose V, Fumagalli D, et al., 2014, Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann Oncol 25:1544– 1550 Dieci MV, Criscitiello C, Goubar A, Viale G, Conte P, Guarneri V, et al., 2014, Prognostic value of tumor-infiltrating lymphocytes on residual disease after primary chemotherapy for triple-negative breast cancer: a retrospective multicenter study. Ann Oncol 25: 611–618 Miller CP, Thorpe JD, Kortum AN, Coy CM, ChengWY, Ou Yang TH, et al., 2014, JAK2 expression is associated with tumorinfiltrating lymphocytes and improved breast cancer outcomes: implications for evaluating JAK2 inhibitors. Cancer Immunol Res 2: 301–306 Seo AN, Lee HJ, Kim EJ, Kim HJ, JangMH, Lee HE, et al., 2013, Tumour-infiltrating CD8+ lymphocytes as an independent predictive factor for pathological complete response to primary systemic therapy in breast cancer. Br J Cancer 109:2705–2713 Chen Z, Chen X, Zhou E, Chen G, Qian K, Wu X, et al., 2014, Intratumoral CD8+ cytotoxic lymphocyte is a favorable prognostic marker in node-negative breast cancer. PLoS One 9:e95475 Melichar B, Studentova H, Kalabova H, Vitaskova D, Cermakova P, Hornychova H, et al., 2014, Predictive and prognostic significance of tumor-infiltrating lymphocytes in patients with breast cancer treated with neoadjuvant systemic therapy. Anticancer Res 34: 1115–1125 Loi S, 2013, Tumor-infiltrating lymphocytes, breast cancer subtypes and therapeutic efficacy. Oncoimmunology 2:e24720 Ruffell B, Au A, Rugo HS, Esserman LJ, Hwang ES, CoussensLM, 2012, Leukocyte composition of human breast cancer. Proc Natl Acad Sci U S A 109:2796–2801 Hussein MR, Hassan HI, 2006, Analysis of the mononuclear inflammatory cell infiltrate in the normal breast, benign proliferative breast disease, in situ and infiltrating ductal breast carcinomas: preliminary observations. J Clin Pathol 59:972–977 Tian Q, Taupin J, Elledge S, RobertsonM, Anderson P, 1995, Fasactivated serine/threonine kinase, FAST, phosphorylates TIA-1 during Fas-mediated apoptosis. J Exp Med 182:865–874 Zhou J, Xu XZ, Hu YR, Hu AR, Zhu CL, Gao GS, 2014, Cryptotanshinone induces inhibition of breast tumor growth by cytotoxic CD4+ T cells through the JAK2/STAT4/perforin pathway. Asian Pac J Cancer Prev 15:2439–2445 Roncati L, Barbolini G, Rivasi F, 2010, Vascular endothelial growth factor/receptor systems expressed by basophils in vaginal angiomyofibroblastoma. Pathologica 102:351 Busam KJ, Antonescu CR, Marghoob AA, Nehal KS, Sachs DL, Shia J, et al., 2001, Histologic classification of tumor-infiltrating lymphocytes in primary cutaneous malignant melanoma.Astudy of interobserver agreement. Am J Clin Pathol 115:856–860 Kuroda H, Tamaru J, Sakamoto G, Ohnisi K, Itoyama S, 2005, Immunophenotype of lymphocytic infiltration in medullary carcinoma of the breast. Virchows Arch 446:10–14 Manenti A, Roncati L, Sighinolfi P, Barbolini G, 2014, Absence of immune response as a sign of tissue tolerance in small-cell lung cancer. Gene Cell Tissue 1:e20330 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 449 EP 452 DI 10.1007/s12253-015-0024-7 PG 4 ER PT J AU Zhao, J Chen, J Lin, H Jin, R Liu, J Liu, X Meng, N Cai, X AF Zhao, Jie Chen, Jiang Lin, Hui Jin, Renan Liu, Jinghua Liu, Xiaolong Meng, Ning Cai, Xiujun TI The Clinicopathologic Significance of BAF250a (ARID1A) Expression in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; ARID1A; BAF250a; Tumor suppressor gene ID Hepatocellular carcinoma; ARID1A; BAF250a; Tumor suppressor gene AB Hepatocellular carcinoma (HCC) is one of the most common and lethal human cancers. Recently, exome sequencing has revealed that mutation of ARID1A is frequent in HCC. Herein, we determined the clinicopathologic significance of ARID1A expression in HCC.We detected the level of mRNA and protein expression of ARID1A in 12 paired HCC tumors and adjacent non-cancerous tissues by quantitative real-time PCR and immunohistochemistry (IHC). In addition, we determined the expression of BAF250a on 121 HCC tumors by IHC and assessed the association between BAF250a expression and clinicopathologic and prognostic features. The levels of ARID1A mRNA were significantly elevated in 10 of 12 HCC tumors compared with adjacent non-cancerous tissues. The level of BAF250a protein expression was higher in 10 of 12 HCC tumors compared with adjacent liver tissues. IHC indicated that 12.17 % of HCC tumors (14/115) were BAF250a-negative. Loss of BAF250a was significantly associated with larger tumor size, but not associated with other clinicopathologic features. There was no significant difference in disease-free or overall survival between BAF250a-positive and BAF250a-negative patients. Most HCCs had an increased level of ARID1A mRNA and BAF250a expression. Loss of BAF250a was significantly more frequent in larger HCC tumors, but had no prognostic significance. C1 [Zhao, Jie] Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Hepatobiliary and Pancreas Surgery, No. 3, East Qinchun Road, 310016 Hangzhou, China. [Chen, Jiang] Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Hepatobiliary and Pancreas Surgery, No. 3, East Qinchun Road, 310016 Hangzhou, China. [Lin, Hui] Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Hepatobiliary and Pancreas Surgery, No. 3, East Qinchun Road, 310016 Hangzhou, China. [Jin, Renan] Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Hepatobiliary and Pancreas Surgery, No. 3, East Qinchun Road, 310016 Hangzhou, China. [Liu, Jinghua] Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Hepatobiliary and Pancreas Surgery, No. 3, East Qinchun Road, 310016 Hangzhou, China. [Liu, Xiaolong] Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Hepatobiliary and Pancreas Surgery, No. 3, East Qinchun Road, 310016 Hangzhou, China. [Meng, Ning] Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Hepatobiliary and Pancreas Surgery, No. 3, East Qinchun Road, 310016 Hangzhou, China. [Cai, Xiujun] Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Hepatobiliary and Pancreas Surgery, No. 3, East Qinchun Road, 310016 Hangzhou, China. RP Cai, X (reprint author), Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Hepatobiliary and Pancreas Surgery, 310016 Hangzhou, China. EM cxjsrrsh@163.com CR Torre LA, Bray F, Siegel RL, et al., 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87–108 Llovet JM, Burroughs A, Bruix J, 2003, Hepatocellular carcinoma. Lancet 362(9399):1907–1917 Huang J, Deng Q, Wang Q, et al., 2012, Exome sequencing of hepatitis B virus-associated hepatocellular carcinoma. Nat Genet 44(10):1117–1121 Guichard C, Amaddeo G, Imbeaud S, et al., 2012, Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet 44, 6):694–698 Fujimoto A, Totoki Y, Abe T, et al., 2012, Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet 44(7):760–764 Narlikar GJ, Fan HY, Kingston RE, 2002, Cooperation between complexes that regulate chromatin structure and transcription. Cell 108(4):475–487 Clapier CR, Cairns BR, 2009, The biology of chromatin remodeling complexes. Annu Rev Biochem 78:273–304 Jones S, Wang TL, Shih Ie M, et al., 2010, Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330(6001):228–231 Wiegand KC, Shah SP, Al-Agha OM, et al., 2010, ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med 363(16):1532–1543 Wiegand KC, LeeAF, Al-AghaOM, et al., 2011, Loss of BAF250a, ARID1A, is frequent in high-grade endometrial carcinomas. J Pathol 224(3):328–333 Lichner Z, Scorilas A, White NM, et al., 2013, The chromatin remodeling gene ARID1A is a new prognostic marker in clear cell renal cell carcinoma. Am J Pathol 182(4):1163–1170 Wang K, Kan J, Yuen ST, et al., 2011, Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat Genet 43(12):1219–1223 Jiao Y, Pawlik TM, Anders RA, et al., 2013, Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nat Genet 45(12): 1470–1473 Cancer Genome Atlas N, 2012, Comprehensive molecular characterization of human colon and rectal cancer. Nature 487(7407):330– 337 Imielinski M, Berger AH, Hammerman PS, et al., 2012, Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150(6):1107–1120 Guan B, Wang TL, Shih IM, 2011, ARID1A, a factor that promotes formation of SWI/SNF-mediated chromatin remodeling, is a tumor suppressor in gynecologic cancers. Cancer Res 71(21): 6718–6727 Yan HB, Wang XF, Zhang Q, et al., 2014, Reduced expression of the chromatin remodeling gene ARID1A enhances gastric cancer cell migration and invasion via downregulation of E-cadherin transcription. Carcinogenesis 35(4):867–876 Luo B, Cheung HW, Subramanian A, et al., 2008, Highly parallel identification of essential genes in cancer cells. Proc Natl Acad Sci U S A 105(51):20380–20385 Dykhuizen EC, Hargreaves DC, Miller EL, et al., 2013, BAF complexes facilitate decatenation of DNA by topoisomerase IIalpha. Nature 497(7451):624–627 Park JH, Park EJ, Lee HS, et al., 2006, Mammalian SWI/SNF complexes facilitate DNA double-strand break repair by promoting gamma-H2AX induction. EMBO J 25(17):3986–3997 Bruix J, Sherman M, 2011, American association for the study of liver D. Management of hepatocellular carcinoma: an update. Hepatology 53(3):1020–1022 Feitelson MA, Sun B, Satiroglu Tufan NL, et al., 2002, Genetic mechanisms of hepatocarcinogenesis. Oncogene 21(16):2593–2604 ZhangX, ZhangY,YangY, et al., 2012, Frequent low expression of chromatin remodeling gene ARID1A in breast cancer and its clinical significance. Cancer Epidemiol 36(3):288–293 Cho H, Kim JS, Chung H, et al., 2013, Loss of ARID1A/BAF250a expression is linked to tumor progression and adverse prognosis in cervical cancer. Hum Pathol 44(7):1365–1374 Zhang ZM, Xiao S, Sun GY, et al., 2014, The clinicopathologic significance of the loss of BAF250a, ARID1A, expression in endometrial carcinoma. Int J Gynecol Cancer 24(3):534–540 Abe H,Maeda D, Hino R, et al., 2012, ARID1A expression loss in gastric cancer: pathway-dependent roles with and without epsteinbarr virus infection and microsatellite instability. Virchows Arch 461(4):367–377 Chou A, Toon CW, Clarkson A, et al., 2014, Loss of ARID1A expression in colorectal carcinoma is strongly associated with mismatch repair deficiency. Hum Pathol 45(8):1697–1703 Katagiri A, Nakayama K, Rahman MT, et al., 2012, Loss of ARID1A expression is related to shorter progression-free survival and chemoresistance in ovarian clear cell carcinoma. Mod Pathol 25(2):282–288 Rahman M, Nakayama K, Rahman MT, et al., 2013, Clinicopathologic analysis of loss of AT-rich interactive domain 1A expression in endometrial cancer. Hum Pathol 44(1):103–109 Fadare O, Gwin K, Desouki MM, et al., 2013, The clinicopathologic significance of p53 and BAF-250a, ARID1A, expression in clear cell carcinoma of the endometrium. Mod Pathol 26(8):1101– 1110 Cho CS, Gonen M, Shia J, et al., 2008, A novel prognostic nomogram is more accurate than conventional staging systems for predicting survival after resection of hepatocellular carcinoma. J Am Coll Surg 206(2):281–291 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 453 EP 459 DI 10.1007/s12253-015-0022-9 PG 7 ER PT J AU El-Gendi, MS Mostafa, FM AF El-Gendi, Mohamed Saba Mostafa, Farouk Mohamed TI Runx2 Expression as a Potential Prognostic Marker in Invasive Ductal Breast Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Runx2; Breast carcinoma; Prognostic factor; Clinical outcome; Survival ID Runx2; Breast carcinoma; Prognostic factor; Clinical outcome; Survival AB The Runx family of transcription factors has been implicated in cancer progression, both positively and negatively. Recent studies assigned a role for Runx2 in promoting breast cancer metastasis. However, the role of Runx2 during the early stage of breast carcinoma and its association with clinical outcomes remain unknown. Assessing the clinicopathological significance of Runx2 expression in a cohort of breast invasive ductal carcinomas (IDC). The correlation of nuclear Runx2 LI with clinicopathological parameters was assessed in 84 IDCs. To study the association of Runx2 with patient outcomes, in addition to treating it as a continuous variable, Runx2 was categorized by its median value (65) and by an additional two cut-off points determined by ROC curve analyses, at 45 for disease free survival (DFS) and 40 for overall survival (OS). Multivariate Cox regression models were also constructed. We used the best subset regression to identify models that predict DFS and OS with as few predictors as possible, and validation was performed. Based on the "Predicted R2", the three best models were identified. Using Cox-regression, the interaction between Runx2 and other clinicopathological terms was tested. Runx2 LI was significantly associated only with positive Her-2 status, and did not correlate significantly with other clinicopathological parameters. Although Runx2 LI, in the continuous form and when categorized by the median, did not correlate significantly with DFS and OS; after it was categorized using the optimal cutoff points determined using ROC curve analysis, the patients with Runx2 LI >45%showed a significantly higher event rate and shorter DFS (P = 0.047), whereas patients with Runx2 LI >40 % showed a significantly shorter OS (P = 0.050). Moreover, Runx2 LI contributed significantly in the models built to predict DFS and OS. For DFS, no interaction terms contributed significantly to the models. However, among stage IV cases, the interaction term between centred Runx2 and ER significantly contributed to the prediction of OS. Runx2 was a significant predictor of OS in this model. Runx2 has a role in biological behaviour and affects the outcome of IDC; therefore, its inhibition may be a new therapeutic strategy. The predictability of Runx2 for OS in stage IV tumours differs with different ER states. The pattern of this difference was not determined because the sample size was not sufficient to allow pattern testing. C1 [El-Gendi, Mohamed Saba] University of Alexandria, Faculty of Medicine, Department of PathologyAlexandria, Egypt. [Mostafa, Farouk Mohamed] Alexandria Faculty of Medicine, Department of Clinical Oncology and Nuclear MedicineAlexandria, Egypt. RP El-Gendi, MS (reprint author), University of Alexandria, Faculty of Medicine, Department of Pathology, Alexandria, Egypt. EM sabaelgendi@yahoo.com CR Onodera Y, Miki Y, Suzuki T, Takagi K, Akahira J, Sakyu T, Watanabe M, Inoue S, Ishida T, Ohuchi N, Sasano H, 2010, Runx2 in human breast carcinoma: its potential roles in cancer progression. Cancer Sci 101:2670–2675 Gnant M, Dubsky P, Fitzal F, Blaha P, Schoppmann S, Steger G, Marth C, Samonigg H, Huttner K, Fohler H, Ruecklinger E, Jakesz R, Greil R, Austrian Breast and Colorectal Cancer Study Group, 2009, Maintaining bone density in patients undergoing treatment for breast cancer: is there an adjuvant Benefit? Clin Breast Cancer 9(Suppl 1):S18–S27 Chimge NO, Baniwal SK, Little GH, Chen YB, Kahn M, Tripathy D, Borok Z, Frenkel B, 2011, Regulation of breast cancer metastasis by Runx2 and estrogen signaling: the role of SNAI2. Breast Cancer Res 13(6):R127 Roodman GD, 2004, Mechanisms of bone metastasis. N Engl J Med 350:1655–1664 Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics. CA Cancer J Clin 59(4):225–249 Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordon- Cardo C, Guise TA, Massague J, 2003, A multigenic program mediating breast cancermetastasis to bone. Cancer Cell 3(6):537– 549 Zhang XH, Wang Q, Gerald W, Hudis CA, Norton L, Smid M, Foekens JA, Massague J, 2009, Latent bone metastasis in breast cancer tied to Src dependent survival signals. Cancer Cell 16(1):67– 78 Henderson IC, 1993, Risk factors for breast cancer development. Cancer Supplement 71:2127–2140 Ma H, Bernstein L, Pike MC, Ursin G, 2006, Reproductive factors and breast cancer risk according to joint estrogen and progesterone receptor status: a meta-analysis of epidemiological studies. Breast Cancer Res 8:R43., DOI 10.1186/bcr1525 Yager JD, Davidson NE, 2006, Estrogen carcinogenesis in breast cancer. N Engl J Med 354(3):270–282 Chlebowski RT, Hendrix SL, Langer RD, Stefanick ML, Gass M, Lane D, Rodabough RJ, Gilligan MA, Cyr MG, Thomson CA, Khandekar J, Petrovitch H, McTiernan A; WHI Investigators, 2003, Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the Women’s Health Initiative Randomized Trial. JAMA, 24, 289:3243–3253. Pike MC, Krailo MD, Henderson BE, Casagrande JT, Hoel DG, 1983, Hormonal’ risk factors, ‘breast tissue age’ and the ageincidence of breast cancer. Nature 303:767–770 Sotiriou C, Piccart MJ, 2007, Taking gene-expression profiling to the clinic: when will molecular signatures become relevant to patient care? Nat Rev Cancer 7:545–553 Chimge NO, Baniwal SK, Luo J, Coetzee S, Omar Khalid O, Berman B, Tripathy D, Ellis M, Frenkel B, 2012, Opposing effects of Runx2 and estradiol on breast cancer cell proliferation: in vitro identification of reciprocally-regulated gene signature related to clinical letrozole responsiveness. Clin Cancer Res 18(3):901–911 Komori T, 2010, Regulation of bone development and extracellular matrix protein genes by RUNX2. Cell Tissue Res 339:189–195 Lo Coco F, Pisegna S, Diverio D, 1997, The AML1 gene: a transcription factor involved in the pathogenesis of myeloid and lymphoid leukemias. Haematologica 82:364–370 Woolf E, Xiao C, Fainaru O, Lotem J, Rosen D, Negreanu V, Bernstein Y, Goldenberg D, Brenner O, Berke G, Levanon D, Groner Y, 2003, Runx3 and Runx1 are required for CD8 T cell development during thymopoiesis. Proc Natl Acad Sci U S A 100: 7731–7736 Levanon D, Glusman G, Bettoun D, Ben-Asher E, Negreanu V, Bernstein Y, Harris-Cerruti C, Brenner O, Eilam R, Lotem J, Fainaru O, Goldenberg D, Pozner A, Woolf E, Xiao C, Yarmus M, Groner Y, 2003, Phylogenesis and regulated expression of the RUNT domain transcription factors RUNX1 and RUNX3. Blood Cells Mol Dis 30(2):161–163 Zaidi SK, Pande S, Pratap J, Gaur T, Grigoriu S, Ali SA, Stein JL, Lian JB, van Wijnen AJ, Stein GS, 2007, Runx2 deficiency and defective subnuclear targeting bypass senescence to promote immortalization and tumorigenic potential. Proc Natl Acad Sci U S A 104:19861–19866 . Blyth K, Vaillant F, Jenkins A, McDonald L, Pringle MA, Huser C, Stein T, Neil J, Cameron ER, 2010, Runx2 in normal tissues and cancer cells: a developing story. Blood Cells, Molecules & Diseases. 45, 2):117–123. Frenkel B, Hong A, Baniwal SK, Coetzee GA, Ohlsson C, Khalid O, Gabet Y, 2010, Regulation of adult bone turnover by sex steroids. J Cell Physiol 224:305–310 Javed A, Barnes GL, Pratap J, Antkowiak T, Gerstenfeld LC, van Wijnen AJ, Stein JL, Lian JB, Stein GS, 2005, Impaired intranuclear trafficking of Runx2, AML3/CBFA1, transcription factors in breast cancer cells inhibits osteolysis in vivo. Proc Natl Acad Sci U S A 102:1454–1459 Pratap J, Imbalzano KM, Underwood JM, Cohet N, Gokul K, Akech J, van Wijnen AJ, Stein JL, Imbalzano AN, Nickerson JA, Lian JB, Stein GS, 2009, Ectopic runx2 expression in mammary epithelial cells disrupts formation of normal acini structure: implications for breast cancer progression. Cancer Res 69:6807–6814 Pratap J, Wixted JJ, Gaur T, Zaidi SK, Dobson J, Gokul KD, Hussain S, van Wijnen AJ, Stein JL, Stein GS, Lian JB, 2008, Runx2 transcriptional activation of Indian hedgehog and a downstream bone metastatic pathway in breast cancer cells. Cancer Res 68:7795–7802 Baniwal SK, Khalid O, Gabet Y, Shah RR, Purcell DJ, Mav D, Kohn-Gabet AE, Shi Y, Coetzee GA, Frenkel B, 2010, Runx2 transcriptome of prostate cancer cells: insights into invasiveness and bone metastasis. Mol Cancer 9:258 Akech J, Wixted JJ, Bedard K, van der Deen M, Hussain S, Guise TA, van Wijnen AJ, Stein JL, Languino LR, Altieri DC, Pratap J, Keller E, Stein GS, Lian JB, 2010, Runx2 association with progression of prostate cancer in patients: mechanisms mediating bone osteolysis and osteoblastic metastatic lesions. Oncogene 29: 811–821 Pratap J, Lian JB, Stein GS, 2011, Metastatic bone disease: role of transcription factors and future targets. Bone 48:30–36 Zelzer E, Glotzer DJ, Hartmann C, Thomas D, Fukai N, Soker S, Olsen BR, 2001, Tissue specific regulation of VEGF expression during bone development requires Cbfa1 ⁄ Runx2. Mech Dev 106, 1–2): 97–106. Selvamurugan N, Kwok S, Partridge NC, 2004, Smad3 interacts with JunB and Cbfa1 ⁄ Runx2 for transforming growth factor-beta1- stimulated collagenase-3 expression in human breast cancer cells. J Biol Chem 279:27764–27773 Barnes GL, Javed A, Waller SM, Kamal MH, Hebert KE, Hassan MQ, Bellahcene A, VanWijnen AJ, YoungMF, Lian JB, Stein GS, Gerstenfeld LC, 2003, Osteoblast-related transcription factors Runx2, Cbfa1 ⁄ AML3, andMSX2 mediate the expression of bone sialoprotein in human metastatic breast cancer cells. Cancer Res 63(10):2631–2637 Das K, Leong DT, Gupta A, Shen L, Putti T, Stein GS, vanWijnen AJ, Salto-Tellez M, 2009, Positive association between nuclear Runx2 and oestrogen-progesterone receptor gene expression characterises a biological subtype of breast cancer. Eur J Cancer 45(13):2239–2248 Page DL, Jensen RA, Simpson JF, 1998, Routinely available indicators of prognosis in breast cancer. Breast Cancer Res Treat 51: 195–208 Fitzgibbons PL1, Page DL,Weaver D, Thor AD, Allred DC, Clark GM, Ruby SG, O'Malley F, Simpson JF, Connolly JL, Hayes DF, Edge SB, Lichter A, Schnitt SJ, 2000, Prognostic factors in breast cancer. College of American pathologists consensus statement 1999. Arch Pathol Lab Med 124, 7):966–978. Ellis IO, Elston CW, 2006, Histologic grade, chapter 19). In: O’Malley FP, Pinder SE, eds, Breast pathology. Elsevier, Philadelphia, PA, pp. 225–233 Ellis IO, Bartlett J, Dowsett M, Humphreys S, Jasani B, Miller K, Pinder SE, Rhodes A, Walker R, 2004, Best practice no. 176: updated recommendation for HER-2 testing in the UK. J Clin Pathol 57(3):322–327. Allred DC, Harvey JM, Berardo M, Clarck GM, 1998, Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11:155–168 Pratap J, Javed A, Languino LR, van Wijnen AJ, Stein JL, Stein GS, Lian JB, 2005, The Runx2 osteogenic transcription factor regulates matrix metalloproteinase 9 in bone metastatic cancer cells and controls cell invasion. Mol Cell Biol 25:8581–8591 RisauW(1997)Mechanisms of angiogenesis. Nature 386:671–674 Bergers G, Brekken R, McMahon G, Vu TH, Itoh T, Tamaki K, Tanzawa K, Thorpe P, Itohara S,Werb Z, Hanahan D, 2000, Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol 2:737–744 Itoh T, Tanioka M, Matsuda H, Nishimoto H, Yoshioka T, Suzuki R, Uehira M, 1999, Experimental metastasis is suppressed in MMP-9-deficient mice. Clin Exp Metastasis 17:177–181 Guise TA, Yin JJ, Taylor SD, Kumagai Y, Dallas M, Boyce BF, Yoneda T, Mundy GR, 1996, Evidence for a causal role of parathyroid hormonerelated protein in the pathogenesis of human breast cancer-mediated osteolysis. J Clin Invest 98:1544–1549 Chang CH, Fan TC,Yu JC, Liao GS, Lin YC, Shih A, LiWH,Yu A, 2014, The prognostic significance of RUNX2 and miR-10a/10b and their inter-relationship in breast cancer. J Transl Med 12:257 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 461 EP 470 DI 10.1007/s12253-015-0018-5 PG 10 ER PT J AU Wu, X Sun, L Wang, X Su, P Li, Z Zhang, Ch Wang, Y Gao, P Ma, R AF Wu, Xiaojuan Sun, Limin Wang, Xiao Su, Peng Li, Zhishuang Zhang, Chunyan Wang, Yan Gao, Peng Ma, Rong TI Breast Cancer Invasion and Metastasis by mPRα Through the PI3K/Akt Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mPRα; p-Akt; MMP-9; Invasive breast cancer; Metastasis ID mPRα; p-Akt; MMP-9; Invasive breast cancer; Metastasis AB Invasive breast cancer is the most common type of malignancy in women worldwide. However, the mechanism responsible for breast cancer metastasis is still unclear and needs further illustration. It has been proven that matrix metallopeptidase 9 (MMP-9) promotes metastasis of the cancer cells. However, the interaction between mPRα and MMP-9 has not been studied. Therefore, in the present research, the effect of MMP-9 on the malignant progression of invasive breast cancer promoted by membrane progesterone receptorα (mPRα) was investigated. The results showed that the protein expression of mPRα, p-Akt and MMP-9 increased in the cancerous tissues compared to that of the noncancerous breast tissue. Furthermore, a positive correlation was found between mPRα and C-erbB-2, as well as the number of involved local lymph nodes. On the other hand, a negative correlation was observed between mPRα and estrogen receptors (ER) along with progesterone receptors (PR). Similarly, a positive association was found between MMP-9 and the number of involved local lymph nodes. Besides, the high expression of MMP-9 also had a positive correlation with the tumor size. However, the high level of MMP-9 had a negative correlation with ER and PR. In addition, there was a positive correlation between mPRα and p-Akt together with MMP-9. The results confirm that mPRα was a major marker of harmful prognosis and it promoted the expression of MMP-9 during invasion to the local lymph nodes through the pathway of PI3K/Akt. The present study provided a novel therapeutic strategy to inhibit breast cancer growth by preventing mPRα signaling pathway. C1 [Wu, Xiaojuan] The Second Hospital of Shandong University, Department of Pathology, 44 Wenhuaxi Road, 250012 Jinan, Shandong, China. [Sun, Limin] Shandong Jiaotong Hospital, Department of OrthopedicsJinan, Shandong, China. [Wang, Xiao] The Second Hospital of Shandong University, Department of Pathology, 44 Wenhuaxi Road, 250012 Jinan, Shandong, China. [Su, Peng] The Second Hospital of Shandong University, Department of Pathology, 44 Wenhuaxi Road, 250012 Jinan, Shandong, China. [Li, Zhishuang] The Second Hospital of Shandong University, Department of Pathology, 44 Wenhuaxi Road, 250012 Jinan, Shandong, China. [Zhang, Chunyan] The Second Hospital of Shandong University, Department of Pathology, 44 Wenhuaxi Road, 250012 Jinan, Shandong, China. [Wang, Yan] The Second Hospital of Shandong University, Department of Pathology, 44 Wenhuaxi Road, 250012 Jinan, Shandong, China. [Gao, Peng] The Second Hospital of Shandong University, Department of Pathology, 44 Wenhuaxi Road, 250012 Jinan, Shandong, China. [Ma, Rong] Shandong Univeristy, QiLu Hospital, Department of Breast SurgeryJinan, Shandong, China. RP Gao, P (reprint author), The Second Hospital of Shandong University, Department of Pathology, 250012 Jinan, China. EM gaopeng@sdu.edu.cn CR Chambers AF, Naumov GN, Varghese HJ, Nadkarni KV, MacDonald IC, Groom AC, 2001, Critical steps in hematogenous metastasis: an overview. Surg Oncol Clin N Am 10(2):243–255 vii Basu A, Rowan BG, 2005, Genes related to estrogen action in reproduction and breast cancer. Front Biosci : J Virtual Libr 10:2346–2372 Carnevale RP, Proietti CJ, Salatino M, Urtreger A, Peluffo G, Edwards DP, et al., 2007, Progestin effects on breast cancer cell proliferation, proteases activation, and in vivo development of metastatic phenotype all depend on progesterone receptor capacity to activate cytoplasmic signaling pathways. Mol Endocrinol 21(6): 1335–1358., DOI 10.1210/me.2006-0304 Karteris E, Zervou S, Pang Y, Dong J, Hillhouse EW, Randeva HS, et al., 2006, Progesterone signaling in human myometrium through two novel membrane G protein-coupled receptors: potential role in functional progesterone withdrawal at term. Mol Endocrinol 20(7): 1519–1534., DOI 10.1210/me.2005-0243 Dressing GE, Alyea R, Pang Y, Thomas P, 2012, Membrane progesterone receptors, mPRs, mediate progestin induced antimorbidity in breast cancer cells and are expressed in human breast tumors. Horm Cancer 3(3):101–112., DOI 10. 1007/s12672-012-0106-x Alizadeh AA, Ross DT, Perou CM, van de Rijn M, 2001, Towards a novel classification of humanmalignancies based on gene expression patterns. J Pathol 195(1):41–52., DOI 10.1002/path.889 Loya A, Guray M, Hennessy BT, Middleton LP, Buchholz TA, Valero V, et al., 2009, Prognostic significance of occult axillary lymph node metastases after chemotherapy-induced pathologic complete response of cytologically proven axillary lymph node metastases from breast cancer. Cancer 115(8): 1605–1612., DOI 10.1002/cncr.24173 Illemann M, Bird N, Majeed A, Sehested M, Laerum OD, Lund LR, et al., 2006, MMP-9 is differentially expressed in primary human colorectal adenocarcinomas and their metastases. Mol Cancer Res : MCR 4(5):293–302., DOI 10.1158/1541-7786.MCR-06-0003 Gompel A, 2012, Micronized progesterone and its impact on the endometrium and breast vs. progestogens. Climacteric : J Int Menopause Soc 15(Suppl 1):18–25., DOI 10.3109/13697137.2012. 669584 L’Hermite M, 2013, HRT optimization, using transdermal estradiol plus micronized progesterone, a safer HRT. Climacteric : J Int Menopause Soc. 16(Suppl 1):44–53., DOI 10.3109/13697137.2013.808563 Kuhl H, Schneider HP, 2013, Progesterone–promoter or inhibitor of breast cancer. Climacteric : J Int Menopause Soc 16(Suppl 1):54– 68., DOI 10.3109/13697137.2013.768806 Zhu Y, Rice CD, Pang Y, Pace M, Thomas P, 2003, Cloning, expression, and characterization of a membrane progestin receptor and evidence it is an intermediary in meiotic maturation of fish oocytes. Proc Natl Acad Sci U S A 100(5):2231– 2236., DOI 10.1073/pnas.0336132100 Zhu Y, Bond J, Thomas P, 2003, Identification, classification, and partial characterization of genes in humans and other vertebrates homologous to a fish membrane progestin receptor. Proc Natl Acad Sci U S A 100(5):2237–2242., DOI 10.1073/pnas.0436133100 Thomas P, Pang Y, Dong J, Groenen P, Kelder J, de Vlieg J, et al., 2007, Steroid and G protein binding characteristics of the seatrout and human progestin membrane receptor alpha subtypes and their evolutionary origins. Endocrinology 148(2):705– 718., DOI 10.1210/en.2006-0974 Lyons TJ, Villa NY, Regalla LM, Kupchak BR, Vagstad A, Eide DJ, 2004, Metalloregulation of yeast membrane steroid receptor homologs. Proc Natl Acad Sci U S A 101(15):5506–5511., DOI 10.1073/pnas.0306324101 van Beijnum JR, Petersen K, Griffioen AW(2009, Tumor endothelium is characterized by a matrix remodeling signature. Front Biosci. 1:216–225 Youssef NS, Hakim SA, 2014, Association of fascin and matrix metalloproteinase-9 expression with poor prognostic parameters in breast carcinoma of egyptian women. Diagn Pathol. 9:136., DOI 10.1186/1746-1596-9-136 Xue B, Huang W, Yuan X, Xu B, Lou Y, Zhou Q, et al., 2015, YSY01A, a novel proteasome inhibitor, induces cell cycle arrest on G2 phase in MCF-7 cells via ERalpha and PI3K/Akt pathways. J Cancer 6(4):319–326., DOI 10.7150/jca.10733 Nagaraj G, Ma C, 2015, Revisiting the estrogen receptor pathway and its role in endocrine therapy for postmenopausal women with estrogen receptor-positive metastatic breast cancer. Breast Cancer Res Treat., DOI 10.1007/s10549-015-3316-4 Paez J, Sellers WR, 2003, PI3K/PTEN/AKT pathway. A critical mediator of oncogenic signaling. Cancer Treat Res 115:145–167 Pu P, Kang C, Li J, Jiang H, 2004, Antisense and dominantnegative AKT2 cDNA inhibits glioma cell invasion. Tumour Biol : J Int Soc Oncodevelopmental Biol Med 25(4):172– 178., DOI 10.1159/000081099 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 471 EP 476 DI 10.1007/s12253-015-0023-8 PG 6 ER PT J AU Zheng, DS Bui, K Chiappori, A Bepler, G Bui, MM AF Zheng, D Sam Bui, Katherine Chiappori, Alberto Bepler, Gerold Bui, M Marilyn TI RRM1, ERCC1 and TS1 Immunofluorescence Expression in Leiomyosarcoma: A Tissue Microarray Study with Clinical Outcome Correlation Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Soft tissue tumor; Leiomyosarcoma; Prognosis; Survival; Tissue microarray; ERCC1; RRM1; TS1 ID Soft tissue tumor; Leiomyosarcoma; Prognosis; Survival; Tissue microarray; ERCC1; RRM1; TS1 AB ERCC1, RRM1 and TS1 are reportedly linked to chemotherapy resistance in lung and other cancers. However, there are currently no studies reporting the relationship between these genes and clinical parameters in leiomyosarcomas. Method: This study investigated the expression pattern of ERCC1, RRM1 and TS1 in forty-four leiomyosarcoma samples by the use of tissue microarray (TMA), immunofluorescence and AQUA methods. The results were then analyzed for expression level and correlations were made with clinical outcome to determine their potential prognostic value in leiomyosarcoma. Results: In the forty-four samples studied, the expression level of these three proteins can be well quantified in the AQUA system and reflected by the AQUA score. RRM1 and ERCC1 expression levels did not show any relationship with overall survival. However, a correlation was found between TS1 expression in the cytoplasm and overall survival. The high expression group had a shorter overall survival time (log-rank p = 0.0498). This trend was confirmed by the Cox proportional hazards model. Discussion: The poor overall survival of leiomyosarcoma is linked to TS1 cytoplasm expression which may be useful in predicting prognoses of this tumor, methods targeting expression of TS1 may lead to improved overall survival in leiomyosarcoma, though more detailed information regarding treatment information and a larger sample size is needed to confirm this phenomenon. C1 [Zheng, D Sam] University of South Florida, College of MedicineTampa, FL, USA. [Bui, Katherine] University of South Florida, College of MedicineTampa, FL, USA. [Chiappori, Alberto] H. Lee Moffitt Cancer Center, Thoracic Oncology, MCC 2nd FL, 12029 Magnolia Drive, 33647 Tampa, FL, USA. [Bepler, Gerold] Detroit Medical Center, Barbara Ann Karmanos Cancer InstituteDetroit, MI, USA. [Bui, M Marilyn] H. Lee Moffitt Cancer Center, Thoracic Oncology, MCC 2nd FL, 12029 Magnolia Drive, 33647 Tampa, FL, USA. RP Bui, MM (reprint author), H. Lee Moffitt Cancer Center, Thoracic Oncology, 33647 Tampa, USA. EM marilyn.bui@moffitt.org CR Damiano G, Di Ganci S, Palumbo VD, Spinelli G, De Luca S, Cudia B, Tomasello G, Lo Monte AI, 2012, Gastric leiomyosarcoma case report and review of literature. Clin Ter 163(4):e181–e184 Yamamoto H, Handa M, Tobo T, Setsu N, Fujita K, Oshiro Y, MiharaY, Yoshikawa Y, Oda Y, 2013, Clinicopathological features of primary leiomyosarcoma of the gastrointestinal tract following recognition of gastrointestinal stromal tumors. Histopathology 63: 194–207 Hadoux J, Morice P, Lhomme C, Duvillard P, Balleyguier C, Haie-Meder C, Gouy S, Uzan C, Mazeron R, Tazi Y, Leary A, Duffaud F, Pautier P, 2013, Uterine leiomyosarcoma: epidemiology, pathology, biology, diagnosis, prognosis and treatment. Bull Cancer. Sep. 100(9):903–915., DOI 10.1684/ bdc.2013.1801 Xu J, Zhang T, Wang T, You L, Zhao Y, 2013, Clinical characteristics and prognosis of primary leiomyosarcoma of the pancreas: a systematic review. World J Surg Oncol. 11:290., DOI 10.1186/1477- 7819-11-290 Nappi L, Mele G, Angioni S, Cicinelli E, Greco P, 2014, Uterine leiomyosarcoma: report of three cases and review of the literature. Eur J Gynaecol Oncol 35:328–331 Agaimy A,Weunsch PH, 2007, True smooth muscle neoplasms of the gastrointestinal tract: morphological spectrum and classification in a series of 85 cases from a single institute. Langenbecks Arch Surg 392:75–81 Itoga M, Ito W, Ueki S, Takeda M, Moritoki Y, Kobayashi Y, Chihara M, Suzuki N, 2013, Sasaki H, 2013, Chihara J. Lung metastasis from perineal leiomyosarcoma: a case report and a review of the Japanese literature. Case Rep Med. 496:304., DOI 10. 1155/2013/496,304 Barwad A, Khandelwal N, Vyas S, Gogoi D, Dey P, 2011, Primary leiomyosarcoma of the prostate with lung metastasis: report of a case diagnosed by fine-needle aspiration cytology. Diagn Cytopathol. 39:700–702., DOI 10.1002/dc.21539 Alonso Gomez J, Arjona Sanchez A,Martinez Cecilia D, Diaz Nieto R, Roldan de la Rua J, Valverde Martinez A, Lizarraga Febres E, Padillo Ruiz J, Rufian Pena S, 2012, Uterine leiomyosarcoma metastasis to the pancreas: report of a case and review of the literature. J Gastrointest Cancer 43:361–363., DOI 10.1007/s12029-010-9172-x Luis J, Ejtehadi F, Howlett DC, Donnellan IM, 2015, Leiomyosarcoma of the small bowel: Report of a case and review of the literature. Int J Surg Case Rep 6C:51–54., DOI 10.1016/j.ijscr. 2014.11.009 Lazar A, Evans HL, Shipley J, 2013, Leiomyosarcoma. In: WHO Classification of Tumours of Soft Tissue and Bone. IARC, France, pp. 111–113 Fernandez-Flores A., 2010, Cutaneous leiomyomas and leiomyosarcomas an immunohistochemical study with p53.Rom J Morphol Embryol 51:295–298. Abdulkader I, Cameselle-Teijeiro J, Gude F, Fraga M,Varela-Duran J, Barreiro F, Forteza J, 2002, Predictors of malignant behaviour in gastrointestinal stromal tumours: a clinicopathological study of 34 cases. Eur J Surg 168:288–296 Bergman AM, Pinedo HM, Jongsma APM, et al., 1999, Decreased resistance to gemcitabine, 2_2_-difluorodeoxycytidine, of cytosine arabinoside-resistant myeloblastic murine and rat leukemia cell lines: role of altered activity and substrate specificity of deoxycytidine kinase. Biochem Pharmacol 57:397–406 Bepler G, Sharma S, Cantor A, et al., 2004, RRM1 and PTEN as prognostic parameters for overall and disease-free survival in patients with non-small-cell lung cancer. J Clin Oncol 22:1878–1886 Bepler G, Kusmartseva I, Sharma S, Gautam A, Cantor A, Sharma A, Simon G, 2006, RRM1 modulated in vitro and in vivo efficacy of gemcitabine and platinum in non-small-cell lung cancer. J Clin Oncol 24:4731–4737 Zheng Z, Chen T, Li X, Haura E, Sharma A, Bepler G, 2007, DNA synthesis and repair genes RRM1 and ERCC1 in lung cancer. N Engl J Med 356:800–808 Simon G, Sharma A, Li X, Hazelton T, Walsh F, Williams C, Chiappori A, Haura E, Tanvetyanon T, Antonia S, Cantor A, Bepler G, 2007). Feasibility and efficacy of molecular analysisdirected individualized therapy in advanced non-small-cell lung cancer. J Clin Oncol. Jul 1;25(19):2741–6. Simon GR, Sharma S, Cantor A, Smith P, Bepler G, 2005, ERCC1 expression is a predictor of survival in resected patients with nonsmall cell lung cancer. Chest 127:978–983 Zheng Z, Li X, Schell MJ, Chen T, Boulware D, Robinson L, Sommers E, Bepler G, 2008, Thymidylate synthase in situ protein expression and survival in stage I nonsmall-cell lung cancer. Cancer 112:2765–2773 Chu E, CallenderMA, Farrell MP, Schmitz JC, 2003, Thymidylate synthase inhibitors as anticancer reagent TS1: from bench to bedside. Cancer Chemother Pharmacol. 52(Suppl 1):S80–S89 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 477 EP 482 DI 10.1007/s12253-015-0021-x PG 6 ER PT J AU Liu, L Yang, ZL Wang, Ch Miao, X Liu, Z Li, D Zou, Q Li, J Liang, L Zeng, G Chen, S AF Liu, Luyao Yang, Zhu-Lin Wang, Chunwei Miao, Xiongying Liu, Zhiyu Li, Daiqiang Zou, Qiong Li, Jinghe Liang, Lufeng Zeng, Guixiang Chen, Senlin TI The Expression of Notch 1 and Notch 3 in Gallbladder Cancer and Their Clinicopathological Significance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gallbladder cancer; Prognosis; Notch 1; Notch 3; Adenocarcinoma; Squamous cell/adenosquamous carcinomas ID Gallbladder cancer; Prognosis; Notch 1; Notch 3; Adenocarcinoma; Squamous cell/adenosquamous carcinomas AB Gallbladder cancers (GBCs) are highly malignant gastrointestinal cancers. The biological makers for the prognosis and targeting therapy of GBCs have not been established. The protein expression of Notch 1 and Notch 3 in 46 squamous cell/adenosquamous carcinomas (SC/ASCs) and 80 adenocarcinomas (AC) was measured using immunohistochemistry. Positive Notch 1 and Notch 3 expression in both SC/ASC and AC was significantly associated with large tumor size, invasion, metastasis, and low surgical curability (P < 0.05 or P < 0.01). Univariate Kaplan-Meier analysis showed that positive Notch 1 and Notch 3 expression was significantly associated with mean survival of SC/ASC and AC patients (P < 0.01 or P < 0.001). Multivariate Cox regression analysis showed that positive Notch 1 and Notch 3 expression, as well as low differentiation, large tumor size, high TNM stage, invasion, lymph node metastasis, and surgical curability are independent poor-prognostic factors in both SC/ASC and AC patients. Positive Notch 1 and Notch 3 expression is closely correlated with severe clinicopathological characteristics and poor prognosis in both SC/ASC and AC patients. C1 [Liu, Luyao] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, Hunan, China. [Yang, Zhu-Lin] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, Hunan, China. [Wang, Chunwei] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, Hunan, China. [Miao, Xiongying] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, Hunan, China. [Liu, Zhiyu] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, Hunan, China. [Li, Daiqiang] Central South University, Second Xiangya Hospital, Department of Pathology, 410011 Changsha, Hunan, China. [Zou, Qiong] Central South University, Third Xiangya Hospital, Department of Pathology, 410013 Changsha, Hunan, China. [Li, Jinghe] Central South University, Xiangya Basic Medical College, Department of Pathology, 410078 Changsha, Hunan, China. [Liang, Lufeng] Hunan Provincial People’s Hospital, Department of Hepatobiliary and Pancreatic Surgery, 410007 Changsha, Hunan, China. [Zeng, Guixiang] Loudi Central Hospital, Department of Pathology, 417011 Loudi, Hunan, China. [Chen, Senlin] Hunan Provincial Tumor Hospital, Department of Pathology, 410013 Changsha, Hunan, China. RP Yang, ZL (reprint author), Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary Diseases, 410011 Changsha, China. EM zhulinyang@yahoo.com CR KimWS, JangKT CSH, et al., 2011, Clinicopathologic analysis of adenosquamous/squamous cell carcinoma of the gallbladder. J Surg Oncol 103:239–242 Chan KM, Yu MC, Lee WC, et al., 2007, Adenosquamous/ squamous cell carcinoma of gallbladder. J Surg Oncol 95:129–134 Mingoli A, Brachini G, Petroni R, et al., 2005, Squamous and adenosquamous cell carcinoma of carcinomas of the gallbladder. J Exp Clin Cancer Res 24:143–150 Kondo M, Dono K, Sakon M, et al., 2002, Adenosquamous caecinoma of the gallbladder. Hepato-Gastroenterology 49:1230– 1234 Oohashi Y, Shirai Y, Wakai T, et al., 2002, Adenosquamous carcinoma of the gallbladder warrants resection only if curative resection is feasible. Cancer 94:3000–3005 Nishihara K, Nagai E, Izumi Y, et al., 1994, Adenosquamous carcinoma of the gallbladder: a clinicopahological, immunohistochemical and flow-cytometric study of twenty cases. Jpn J Cancer Res 85:389–399 Wu K, Xu L, Zhang L, Lin Z, et al., 2011, High Jagged1 expression predicts poor outcome in clear cell renal cell carcinoma. Jpn J Clin Oncol 41:411–416 Aster JC, Blacklow SC, Pear WS, 2011, Notch signalling in T-cell lymphoblastic leukaemia/lymphoma and other haematological malignancies. J Pathol 223:262–273 Chen X, Stoeck A, Lee SJ, ShihM, et al., 2010, Jagged1 expression regulated by Notch3 and Wnt/β-catenin signaling pathways in ovarian Cancer. Oncotarget 1:210–218 Donnem T, Andersen S, Al-Shibli K, et al., 2010, Prognostic impact of notch ligands and receptors in nonsmall cell lung cancer: coexpression of notch-1 and vascular endothelial growth factor-a predicts poor survival. Cancer 116:5676–5685 Ranganathan P,Weaver KL, Capobianco AJ, 2011)Notch signaling in solid tumours: a little bit of everything but not all the time. Nat Rev Cancer 11:338–351 Yao J, Qian C, 2009, Over-activated notch-1 protects gastric carcinoma BGC-823 cells from TNFalpha-induced apoptosis. Dig Liver Dis 41:867–874 Wang Z, Zhang Y, Li Y, et al., 2006, Down-regulation of notch-1 contributes to cell growth inhibition and apoptosis in pancreatic cancer Cells. Mol Cancer Ther 5:483–493 Clay MR, Varma S, West RB, 2013, MAST2 and NOTCH1 translocations in breast carcinoma and associated pre-invasive lesions. Hum Pathol 44:2837–2844 Zhang H, Wang X, Xu J, et al., 2014, Notch1 activation is a poor prognostic factor in patients with gastric cancer. Br J Cancer 110: 2283–2290 Xiong Y, Zhang YY, Wu YY, et al., 2014, Correlation of overexpressions of miR-21 and notch-1 in human colorectal cancer with clinical stages. Life Sci 106:19–24 Du X, Zhao YP, Zhang TP, et al., 2013, Notch1 contributes to chemoresistance to gemcitabine and serves as an unfavorable prognostic indicator in pancreatic cancer. World J Surg 37:1688–1694 Maliekal TT, Bajaj J, Giri V, et al., 2008, The role of notch signaling in human cervical cancer: implications for solid tumors. Oncogene 27:5110–5114 Wang M, Wang J, Wang L, et al., 2010, Notch1 expression correlates with tumor differentiation status in ovarian carcinoma. Med Oncol 27:1329–1335 Abdou AG, El-Wahed MM, Kandil MA, et al., 2013, Immunohistochemical analysis of the role and relationship between notch-1 and Oct-4 expression in urinary bladder Carcinoma. APMIS 121:982–996 Lu Z, Liu H, Xue L, et al., 2008, An activated Notch1 signaling pathway inhibits cell proliferation and induces apoptosis in human esophageal squamous cell carcinoma cell line EC9706. Int J Oncol 32:643–651 Duan L, Yao J,Wu X, et al., 2006, Growth suppression induced by Notch1 activation involves Wnt-beta-catenin down-regulation in human tongue carcinoma cells. Biol Cell 98:479–490 Qi R, An H, Yu Y, et al., 2003, Notch1 signaling inhibits growth of human hepatocellular carcinoma through induction of cell cycle arrest and apoptosis. Cancer Res 63:8323–8329 Bellavia D, Checquolo S, Campese AF, et al., 2008, Notch3: from subtle structural differences to functional diversity. Oncogene 27: 5092–5098 Kang H, An HJ, Song JY, et al., 2012, Notch3 and Jagged2 contribute to gastric cancer development and to glandular differentiation associated with MUC2 and MUC5AC expression. Histopathology 61:576–586 Ozawa T, Kazama S, Akiyoshi T, et al., 2014, Nuclear Notch3 expression is associated with tumor recurrence in patients with stage II and III colorectal cancer. Ann Surg Oncol 21:2650–2658 Hu L, Xue F, Shao M, et al., 2013, Aberrant expression of Notch3 predicts poor survival for hepatocellular Carcinomas. Biosci Trends 7:152–156 Mann CD, Bastianpillai C, Neal CP, et al., 2012, Notch3 and HEY- 1 as prognostic biomarkers in pancreatic Adenocarcinoma. PLoS One 7:e51119 Ye YZ, Zhang ZH, Fan XY, et al., 2013, Notch3 overexpression associates with poor prognosis in human non-small-cell lung cancer. Med Oncol 30:595 Yeasmin S, Nakayama K, Rahman MT, et al., 2010, Expression of nuclear Notch3 in cervical squamous cell carcinomas and its association with adverse clinical Outcomes. Gynecol Oncol 117:409– 416 Rahman MT, Nakayama K, Rahman M, et al., 2012, Notch 3 overexpression as potential therapeutic target in advanced stage chemoresistant ovarian cancer. Am J Clin Pathol 138:535–544 Danza G, Di Serio C, Ambrosio MR, et al., 2013, Notch3 is activated by chronic hypoxia and contributes to the progression of human prostate Cancer. Int J Cancer 133:2577–2586 Ding XY, Ding J,Wu K, et al., 2012, Cross-talk between endothelial cells and tumor via delta-like ligand 4/notch/PTEN signaling inhibits lung cancer growth. Oncogene 31:2899–2906 Liu DC, Yang ZL, 2011, Clinicopathologic significance of minichromosomemaintenance protein 2 and Tat-interacting protein 30 expression in benign and malignant lesions of the gallbladder. Hum Pathol 42:1676–1683 Park HS, Jung CK, Lee SH, et al., 2012, Notch1 receptor as a marker of lymph node metastases in papillary thyroid cancer. Cancer Sci 103:305–309 Sun W, Gaykalova DA, Ochs MF, et al., 2014, Activation of the NOTCH pathway in head and neck cancer. Cancer Res 74:1091– 1104 Alqudah MA, Agarwal S, Al-Keilani MS, et al., 2013, NOTCH3 is a prognostic factor that promotes glioma cell proliferation, migration and invasion via activation of CCND1 and EGFR. PLoS One 8:e77299 Howard JD, Moriarty WF, Park J, et al., 2013, Notch signaling mediatesmelanoma-endothelial cell communication andmelanoma cell migration. Pigment Cell Melanoma Res 26:697–707 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 483 EP 492 DI 10.1007/s12253-015-0019-4 PG 10 ER PT J AU Lakosi, F Gulyban, Simoni, BMS Nguyen, VP Cucchiaro, S Seidel, L Janvary, ZsL Nicolas, S Vavassis, P Coucke, P AF Lakosi, Ferenc Gulyban, Akos Simoni, Ben-Mustapha Selma Nguyen, Viet Paul Cucchiaro, Severine Seidel, Laurence Janvary, Zsolt Levente Nicolas, Sophie Vavassis, Peter Coucke, Philippe TI The Influence of Treatment Position (Prone vs. Supine) on Clip Displacement, Seroma, Tumor Bed and Partial Breast Target Volumes: Comparative Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Prone breast radiotherapy; Partial breast irradiation; Target volume ID Breast cancer; Prone breast radiotherapy; Partial breast irradiation; Target volume AB To analyse the displacement of surgical clips in prone (Pr) position and assess the consequences on target volumes and integral dose of partial breast irradiation (PBI). 30 post-lumpectomy breast cancer patients underwent CT imaging in supine (Su) and Pr. Clip displacements were measured by the distances from the clips to a common fix bony reference point. On each dataset, the tumour bed (TB = clips ± seroma), clinical target volume (CTV = TB + 1.5 cm) and planning target volumes (PTV = CTV + 1 cm) for PBI were determined and the volume pairs were compared. Furthermore estimation of integral dose ratio (IDR) within the breast from tangential treatment was performed as the ratio of the irradiated breast volume and the volume encompassing all clips. Clips close to the chest wall (CW) in Su showed significantly less displacement in Pr. The mean volumes of seroma, CTV and PTV were significantly higher in Pr than in Su. The PTV volume difference (Pr-Su) was significantly higher in patients with presence of seroma, deep clips and TB location in the superior-internal-quadrant (SIQ) and at the junction of superior quadrants (jSQ). In a multivariate analysis two factors remained significant: seroma and TB localization in SIQjSQ. The IDR was significantly larger in Su than in Pr (7.6 vs. 4.1 p <0.01). Clip displacements varied considerably with respect to their relative position to the CW. In selected patients Pr position potentially leads to a significant increase in target volumes of PBI. Tangential beam arrangement for PBI should be avoided, not only in Su but in Pr as well in case of clipbased target volume definition. C1 [Lakosi, Ferenc] University Hospital of Liege, Department of Radiation Oncology, B.35, B-4000 Liege, Belgium. [Gulyban, Akos] University Hospital of Liege, Department of Radiation Oncology, B.35, B-4000 Liege, Belgium. [Simoni, Ben-Mustapha Selma] University Hospital of Liege, Department of Radiation Oncology, B.35, B-4000 Liege, Belgium. [Nguyen, Viet Paul] University Hospital of Liege, Department of Radiation Oncology, B.35, B-4000 Liege, Belgium. [Cucchiaro, Severine] University Hospital of Liege, Department of Radiation Oncology, B.35, B-4000 Liege, Belgium. [Seidel, Laurence] University Hospital of Liege, Department of BiostatisticsLiege, Belgium. [Janvary, Zsolt Levente] University of Debrecen, Medical and Health Science Center, Institute of Nuclear MedicineDebrecen, Hungary. [Nicolas, Sophie] University Hospital of Liege, Department of Radiation Oncology, B.35, B-4000 Liege, Belgium. [Vavassis, Peter] Hopital Maisonneuve-Rosemont, Department of Radiation OncologyMontreal, Quebec, Canada. [Coucke, Philippe] University Hospital of Liege, Department of Radiation Oncology, B.35, B-4000 Liege, Belgium. RP Lakosi, F (reprint author), University Hospital of Liege, Department of Radiation Oncology, B-4000 Liege, Belgium. EM lakosiferenc@yahoo.com CR Darby S, McGale P, Correa C, Taylor C, Arriagada R, Clarke M et al, 2011, Effect of radiotherapy after breast-conserving surgery on 10-year recurrence and 15-year breast cancer death: meta-analysis of individual patient data for 10,801 women in 17 randomised trials. Lancet 378:1707–1716., DOI 10.1016/S0140-6736(11)61629-2 Kovacs A, Lakosi F, Liposits G, Toller G, Hadjiev J, Vandulek C et al, 2011, 3-D conformal photon boost in the treatment of early stage breast cancer: four year follow up results. Pathol Oncol Res 17:17– 23., DOI 10.1007/s12253-010-9264-8 Polgar C, Van Limbergen E, Potter R, Kovacs G, Polo A, Lyczek J et al, 2010, Patient selection for accelerated partial-breast irradiation, APBI, after breast-conserving surgery: recommendations of the Groupe Europeen de Curietherapie-European Society for Therapeutic Radiology and Oncology, GEC-ESTRO, breast cancer working group ba. Radiother Oncol 94:264–273., DOI 10.1016/j. radonc.2010.01.014 Smith BD, Arthur DW, Buchholz TA, Haffty BG, Hahn CA, Hardenbergh PH et al, 2009, Accelerated partial breast irradiation consensus statement from the American Society for Radiation Oncology, ASTRO). Int J Radiat Oncol Biol Phys 74:987–1001., DOI 10.1016/j.ijrobp.2009.02.031 Polgar C, Fodor J, Major T, Sulyok Z, Kasler M, 2013, Breastconserving therapy with partial or whole breast irradiation: tenyear results of the Budapest randomized trial. Radiother Oncol 108:197–202., DOI 10.1016/j.radonc.2013.05.008 Mozsa E, Meszaros N,Major T, Frohlich G, Stelczer G, Sulyok Z et al, 2014, Accelerated partial breast irradiation with external beam three-dimensional conformal radiotherapy : five-year results of a prospective phase II clinical study. Strahlenther Onkol 444–450., DOI 10.1007/s00066-014-0633-1 Formenti SC, Hsu H, Fenton-Kerimian M, Roses D, Guth A, Jozsef G et al, 2012, Prone accelerated partial breast irradiation after breast-conserving surgery: five-year results of 100 patients. Int J Radiat Oncol Biol Phys., DOI 10.1016/j.ijrobp.2012.01.039 Kirby AM, Evans PM, Donovan EM, Convery HM, Haviland JS, Yarnold JR, 2010, Prone versus supine positioning for whole and partial-breast radiotherapy: a comparison of non-target tissue dosimetry. Radiother Oncol 96:178–184., DOI 10.1016/j.radonc.2010. 05.014 Coles CE, Wilson CB, Cumming J, Benson JR, Forouhi P, Wilkinson JS et al, 2009, Titaniumclip placement to allow accurate tumour bed localisation following breast conserving surgery: audit on behalf of the IMPORT Trial Management Group. Eur J Surg Oncol 35:578–582., DOI 10.1016/j.ejso.2008.09.005 Major T, Frohlich G, Lovey K, Fodor J, Polgar C, 2009, Dosimetric experience with accelerated partial breast irradiation using imageguided interstitial brachytherapy. Radiother Oncol 90:48–55., DOI 10.1016/j.radonc.2007.10.027 Ahunbay EE, Robbins J, Christian R, Godley A, White J, Li XA, 2012, Interfractional target variations for partial breast irradiation. Int J Radiat Oncol Biol Phys 82:1594–1604., DOI 10.1016/j.ijrobp. 2011.01.041 Kirby AM, Evans PM, Helyer SJ, Donovan EM, Convery HM, Yarnold JR, 2011, A randomised trial of supine versus prone breast radiotherapy, SuPr study): comparing set-up errors and respiratory motion. Radiother Oncol 100:221–226., DOI 10.1016/j.radonc.2010. 11.005 Mitchell J, Formenti SC, DeWyngaert JK, 2010, Interfraction and intrafraction setup variability for prone breast radiation therapy. Int J Radiat Oncol Biol Phys 76:1571–1577., DOI 10.1016/j.ijrobp. 2009.07.1683 Lakosi F, Gulyban A, Janvary L, Simoni SB-M, Jansen N, Seidel L et al, 2015, Respiratory motion, anterior heart displacement and heart dosimetry: comparison between prone, Pr, and supine, Su, whole breast irradiation. Pathol Oncol Res., DOI 10.1007/s12253- -9932-9 15. MukeshM, Harris E, Jena R, Evans P, Coles C, 2012, Relationship between irradiated breast volume and late normal tissue complications: a systematic review. Radiother Oncol 104:1–10., DOI 10.1016/ j.radonc.2012.04.025 Leonard KL, Hepel JT, Hiatt JR, Dipetrillo TA, Price LL,Wazer DE, 2013, The effect of dose-volume parameters and interfraction interval on cosmetic outcome and toxicity after 3-dimensional conformal accelerated partial breast irradiation. Int J Radiat Oncol Biol Phys 85:623–629., DOI 10.1016/j.ijrobp.2012.06.052 Formenti SC, DeWyngaert JK, Jozsef G, Goldberg JD, 2012, Prone vs supine positioning for breast cancer radiotherapy. JAMA 308: 861–863., DOI 10.1001/2012.jama.10759 Mulliez T, Speleers B, Madani I, De Gersem W, Veldeman L, De Neve W, 2013, Whole breast radiotherapy in prone and supine position: is there a place for multi-beam IMRT? Radiat Oncol 8: 151., DOI 10.1186/1748-717X-8-151 Qiu J-J, Chang Z, Horton JK, Wu Q-RJ, Yoo S, Yin F-F, 2014, Dosimetric comparison of 3D conformal, IMRT, and V-MAT techniques for accelerated partial-breast irradiation, APBI). Med Dosim 39:152–158., DOI 10.1016/j.meddos.2013.12.001 Fahimian B, Yu V, Horst K, Xing L, Hristov D, 2013, Trajectory modulated prone breast irradiation: a LINAC-based technique combining intensity modulated delivery and motion of the couch. Radiother Oncol 109:475–481., DOI 10.1016/j.radonc.2013.10.031 Vicini F, Winter K, Wong J, Pass H, Rabinovitch R, Chafe S et al, 2010, Initial efficacy results of RTOG 0319: three-dimensional conformal radiation therapy, 3D-CRT, confined to the region of the lumpectomy cavity for stage I/ II breast carcinoma. Int J Radiat Oncol Biol Phys 77:1120–1127., DOI 10.1016/j.ijrobp.2009.06.067 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 493 EP 500 DI 10.1007/s12253-015-0028-3 PG 8 ER PT J AU Hashiguchi, Y Kasai, M Fukuda, T Ichimura, T Yasui, T Sumi, T AF Hashiguchi, Yasunori Kasai, Mari Fukuda, Takeshi Ichimura, Tomoyuki Yasui, Tomoyo Sumi, Toshiyuki TI Serum Sialyl-Tn (STN) as a Tumor Marker in Patients with Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Serum sialyl Tn level; Endometrial cancer; Tumormarker ID Serum sialyl Tn level; Endometrial cancer; Tumormarker AB There are no potential tumor markers validated for prognosis of endometrial cancer. However, sialyl Tn (STN) is a carbohydrate antigen that is associated with the production of mucin, which reportedly plays important roles in carcinogenesis. Although STN expression in endometrial cancer has been investigated, its prognostic value remains controversial and no studies have investigated serum STN levels in large case series. In this study, we investigated diagnostic and prognostic applications of serum STN for endometrial cancer. Between January 2006 and December 2012, serum STN levels were examined prospectively in patients with endometrial cancer. A total of 146 patients (stage I, 98; stage II, 15; stage III, 17; stage IV, 16) were treated for endometrial cancer. The median age was 60 years (28–83). Subsequently 29 patients (19.9%) relapsed at the time of the last follow-up and the median follow-up time was 44 months (1–83). Elevated serum STN levels were identified in 36 patients (24.7%) and were associated with histological grade (p =0.02) and lymph node metastasis (p= 0.006). Elevated serum STN levels were not related to histological types, clinical stages, myometrial invasions, distant metastases, age, menopausal status, body mass index, or relapse. Among the 36 patients with elevated serum STN levels, 33 (91.7%) achieved remission and serum STN levels returned to the normal range. Seven patients (21.2%) with elevated serum STN levels at baseline relapsed and their serum STN levels were again elevated. Serum STN levels are a potential prognostic indicator for endometrial cancer. C1 [Hashiguchi, Yasunori] Osaka City University, Graduate School of Medicine, Department of Obstetrics and Gynecology, 1-4-3 Asahimachi, Abeno-ku, 545-8585 Osaka, Japan. [Kasai, Mari] Osaka City University, Graduate School of Medicine, Department of Obstetrics and Gynecology, 1-4-3 Asahimachi, Abeno-ku, 545-8585 Osaka, Japan. [Fukuda, Takeshi] Osaka City University, Graduate School of Medicine, Department of Obstetrics and Gynecology, 1-4-3 Asahimachi, Abeno-ku, 545-8585 Osaka, Japan. [Ichimura, Tomoyuki] Osaka City University, Graduate School of Medicine, Department of Obstetrics and Gynecology, 1-4-3 Asahimachi, Abeno-ku, 545-8585 Osaka, Japan. [Yasui, Tomoyo] Osaka City University, Graduate School of Medicine, Department of Obstetrics and Gynecology, 1-4-3 Asahimachi, Abeno-ku, 545-8585 Osaka, Japan. [Sumi, Toshiyuki] Osaka City University, Graduate School of Medicine, Department of Obstetrics and Gynecology, 1-4-3 Asahimachi, Abeno-ku, 545-8585 Osaka, Japan. RP Hashiguchi, Y (reprint author), Osaka City University, Graduate School of Medicine, Department of Obstetrics and Gynecology, 545-8585 Osaka, Japan. EM cbl37090yh@nifty.com CR Staff AC, Trovik J, Eriksson AGZ et al, 2011, Elevated plasma growth differentiation factor-15 correlates with lymph node metastases and poor survival in endometrial cancer. Clin Cancer Res 17(14):4825–4833 Kurihara T, Mizunuma H, Obara Met al, 1998, Determination of a normal level of serum CA125 in postmenopausal women as a tool for preoperative and postoperative surveillance of endometrial carcinoma. Gynecol Oncol 69:192–196 Lo SS, Cheng DK, Ng TY et al, 1997, Prognostic significance of tumor markers in endometrial cancer. Tumour Biol 18:241–249 Gadducci A, Colsio S, Capri A et al, 2004, Serum tumor markers in the management of ovarian, endometrial and cervical cancer. Biomed Pharmacother 1:24–38 Kaku T, Kamura T, Hirakawa Tet al, 1999, Endometrial carcinoma associated with hyperplasia-immunohistochemical study of angiogenesis and p53 expression. Gynecol Oncol 72:51–55 Ohno S, Ohno Y, Nakada H et al, 2006, Expression of Tn and sialyl-Tn antigen in endometrial cancer: its relationship with tumor-produced cyclooxygenase-2, tumor-infiltrated lymphocytes and patient prognosis. Anticancer Res 26:4047–4053 Sewell R, Backstrom M, Dalziel M et al, 2006, The ST6GaINAc-I sialyltransferase localizes throughout the Golgi and is responsible for the synthesis of the tumor-associated sialyl-Tn O-glycan in human breast cancer. J Biol Chem 281(6):3586–3594 Ogata S, Koganty R, Reddish M et al, 1998, Different models of sialyl-Tn expression during malignant transformation of human colonic mucosa. Glycoconj J 15(1):29–35 Kim GE, Bae HI, Park HU et al, 2002, Aberrant expression of MUC5AC and MUC6 gastric mucins and sialyl Tn antigen in intraepithelial neoplasms of the pancreas. Gastroenterology 123(4):1052–1060 Conze T, Carvalho AS, Landegren U et al, 2010)MUC2 mucin is a major carrier of the cancer-associated sialyl-Tn antigen in intestinal metaplasia and gastric carcinomas. Glycobiology 20(2):199–206 Van Elssen CH, Frings PW, Bot FJ et al, 2010, Expression of aberrantly glycosylated Mucin-1 in ovarian cancer. Histopathology 57(4): 597–606 Semczuk A, Paszkowska A, Miturski R et al, 2002, Sialyl-Tn expression in normal and pathological conditions of human endometrium. An immunohistochemical study. Pathol Res Pract 198(9): 589–595 Numa F, Tsunaga N, Michioka T et al, 1995, Tissue expression of sialyl Tn antigen in gynecologic tumors. J Obstet Gynaecol 21(4): 385–389 InoueM, Ogawa H,NakanishiKet al, 1990, Clinical value of sialyl Tn antigen in patients with gynecologic tumors. Obstet Gynecol 75(6):1032–1036 Pecorelli S, 2010, Revised FIGO staging for carcinoma of the vulva, cervix, and endometrium. Int J Gynaecol Obstet 108(2):176 Duk JM, Aalders JG, Fleuren GJ et al, 1986, CA125: a useful marker in endometrial carcinoma. Am J Obstet Gynecol 155: 1097–1102 Patsner B, Mann WJ, Cohen H et al, 1998, Predictive value of preoperative serum CA 125 levels in clinically localized and advanced endometrial carcinoma. Am J Obstet Gynecol 158:399–402 Bruce P, Yim GW, 2013, Predictive value of preoperative serum CA-125 levels in patients with uterine cancer: the Asian experience 2000 to 2012. Obstet Gynecol Sci 56(5):281–288 An YH, Zhang HF, Sun M et al, 2012, sTn is a novel biomarker for type I endometrial carcinoma. Prog Biochem Biophys 39(6):548–555 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 501 EP 504 DI 10.1007/s12253-015-0030-9 PG 4 ER PT J AU Nagy, BZs Wichmann, B Kalmar, A Bartak, KB Tulassay, Zs Molnar, B AF Nagy, Brigitta Zsofia Wichmann, Barnabas Kalmar, Alexandra Bartak, Kinga Barbara Tulassay, Zsolt Molnar, Bela TI miRNA Isolation from FFPET Specimen: A Technical Comparison of miRNA and Total RNA Isolation Methods SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MiRNA; Isolation kit; Comparison; Reference MiRNA; U6; Normalization; Total RNA; Isolation; Ffpe; Colon; Colorectal cancer; Concentration ID MiRNA; Isolation kit; Comparison; Reference MiRNA; U6; Normalization; Total RNA; Isolation; Ffpe; Colon; Colorectal cancer; Concentration AB MiRNA remain stable for detection and PCR-based amplification in FFPE tissue samples. Several miRNA extraction kits are available, however miRNA fraction, as part of total RNA can be isolated using total RNA purification methods, as well. Our primary aim was to compare four different miRNA and total RNA isolation methods from FFPE tissues. Further purposes were to evaluate quantitatively and qualitatively the yield of the isolated miRNA. MiRNAs were isolated from normal colorectal cancer FFPE specimens from the same patients. Two miRNA isolation kits (High Pure miRNA Isolation Kit, miRCURY™ RNA Isolation Kit) and two total RNA isolation kits were compared (High Pure RNA Paraffin Kit, MagNA Pure 96 Cellular RNA LV Kit). Quantity and quality were determined, expression analysis was performed by realtime PCR using qPCR Human Panel I + II (Exiqon) method detecting 742 human miRNAs in parallel. The yield of total RNA was found to be higher than miRNA purification protocols (in CRC: Ex: 0203 ± 0021 μg; HPm: 1,45 ± 0,8 μg; HPp: 21,36 ± 4,98 μg; MP: 8, 6 ± 5,1 μg). MiRNAs were detected in lower relative quantity of total RNA compared to the miRNA kits. Higher number of miRNAs could be detected by the miRNA isolation kits in comparison to the total RNA isolation methods. (Ex: 497 ± 16; HPm: 542 ± 11; HPp: 332 ± 36; MP: 295 ± 74). Colon specific miRNAs (miR-21-5p;-34-5p) give satisfying results by miRNA isolation kits. Although miRNA can be detected also after total RNA isolation methods, for reliable and reproducible miRNA expression profiling the use of miRNA isolation kits are more suitable. C1 [Nagy, Brigitta Zsofia] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Str. 46, 1088 Budapest, Hungary. [Wichmann, Barnabas] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. [Kalmar, Alexandra] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Str. 46, 1088 Budapest, Hungary. [Bartak, Kinga Barbara] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Str. 46, 1088 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Str. 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi Str. 46, 1088 Budapest, Hungary. RP Nagy, BZs (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM nagyzsofiab@gmail.com CR Lewis BP, Burge CB, Bartel DP, 2005, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120(1):15–20., DOI 10.1016/j. cell.2004.12.035 Aslam MI, Taylor K, Pringle JH, Jameson JS, 2009, MicroRNAs are novel biomarkers of colorectal cancer. The British Journal of Surgery 96(7):702–710., DOI 10.1002/bjs.6628 Agostini M, Pucciarelli S, Calore F, Bedin C, Enzo M, Nitti D, 2010, MiRNAs in colon and rectal cancer: a consensus for their true clinical value. Clinica Chimica Acta; International Journal of Clinical Chemistry 411(17–18):1181–1186., DOI 10.1016/j.cca. 2010.05.002 Wang CJ, Zhou ZG,Wang L, Yang L, Zhou B, Gu J, Chen HY, Sun XF, 2009, Clinicopathological significance of microRNA-31,-143 and-145 expression in colorectal cancer. Dis Markers 26(1):27–34., DOI 10.3233/Dma-2009-0601 Kalmar A, Peterfia B, Wichmann B, Patai AV, Bartak BK, Nagy ZB, Furi I, Tulassay Z, Molnar B, 2015, Comparison of automated and manual DNA isolation methods for DNA methylation analysis of biopsy, fresh frozen, and formalin-fixed. Paraffin-Embedded Colorectal Cancer Samples Journal of Laboratory Automation., DOI 10.1177/2211068214565903 Godfrey TE, Kim SH, Chavira M, Ruff DW,Warren RS, Gray JW, Jensen RH, 2000, Quantitative mRNA expression analysis from formalin-fixed, paraffin-embedded tissues using 5 ' nuclease quantitative reverse transcription-polymerase chain reaction. J Mol Diagn 2(2):84–91., DOI 10.1016/S1525-1578(10)60621-6 Cronin M, Pho M, Dutta D, Stephans JC, Shak S, Kiefer MC, Esteban JM, Baker JB, 2004, Measurement of gene expression in archival paraffin-embedded tissues - development and performance of a 92-gene reverse transcriptase-polymerase chain reaction assay. Am J Pathol 164(1):35–42., DOI 10.1016/S0002-9440(10)63093-3 Kalmar A,Wichmann B, Galamb O, Spisak S, Toth K, Leiszter K, Tulassay Z, Molnar B, 2013, Gene expression analysis of normal and colorectal cancer tissue samples from fresh frozen and matched formalin-fixed, paraffin-embedded, FFPE, specimens after manual and automated RNA isolation. Methods 59(1):S16–S19., DOI 10. 1016/j.ymeth.2012.09.011 Li J, Smyth P, Flavin R, Cahill S, Denning K, Aherne S, Guenther SM, O'Leary JJ, SheilsO(2007, Comparison ofmiRNAexpression patterns using total RNA extracted from matched samples of formalin-fixed paraffin-embedded, FFPE, cells and snap frozen cells. BMC Biotechnol 7:36., DOI 10.1186/1472-6750-7-36 Bovell L, Shanmugam C, Katkoori VR, Zhang B, Vogtmann E, Grizzle WE, Manne U, 2012, MiRNAs are stable in colorectal cancer archival tissue blocks. Front Biosci 4:1937–1940 Podolska A, Kaczkowski B, Litman T, Fredholm M, Cirera S, 2011, How the RNA isolation method can affect microRNA microarray results. Acta Biochim Pol 58(4):535–540 Monleau M, Bonnel S, Gostan T, Blanchard D, Courgnaud V, Lecellier CH, 2014, Comparison of different extraction techniques to profilemicroRNAs from human sera and peripheral bloodmononuclear cells. BMC Genomics 15:395., DOI 10.1186/1471-2164-15- 395 Guo Y, Bosompem A, Zhong X, Clark T, Shyr Y, Kim AS, 2014, A comparison of microRNA sequencing reproducibility and noise reduction using mirVana and TRIzol isolation methods. Int J Comput Biol Drug Des 7(2–3):102–112., DOI 10.1504/IJCBDD. 2014.061642 Oue N, Anami K, Schetter AJ, Moehler M, Okayama H, KhanMA, Bowman ED, Mueller A, Schad A, ShimomuraM, Hinoi T, Aoyagi K, Sasaki H, Okajima M, Ohdan H, Galle PR, YasuiW, Harris CC, 2014, High miR-21 expression from FFPE tissues is associated with poor survival and response to adjuvant chemotherapy in colon cancer. Int J Cancer 134(8):1926–1934., DOI 10.1002/Ijc.28522 Schetter AJ, Nguyen GH, Bowman ED, Mathe EA, Yuen ST, Hawkes JE, Croce CM, Leung SY, Harris CC, 2009, Association of inflammation-related and microRNA gene expression with cancer-specific mortality of colon adenocarcinoma. Clin Can Res: An Official Journal of the American Association for Cancer Research 15(18):5878–5887., DOI 10.1158/1078-0432.CCR-09- 0627 Schetter AJ, Leung SY, Sohn JJ, Zanetti KA, Bowman ED, Yanaihara N, Yuen ST, Chan TL, Kwong DL, Au GK, Liu CG, Calin GA, Croce CM, Harris CC, 2008, MicroRNA expression profiles associated with prognosis and therapeutic outcome in colon adenocarcinoma. JAMA 299(4):425–436., DOI 10.1001/jama.299.4. 425 Wang L, Yu J, Xu J, Zheng C, Li X, Du J, 2015, The analysis of microRNA-34 family expression in human cancer studies comparing cancer tissues with corresponding pericarcinous tissues. Gene 554(1):1–8., DOI 10.1016/j.gene.2014.10.032 Hiyoshi Y, Schetter AJ, Okayama H, Inamura K, Anami K, Nguyen GH, Horikawa I, Hawkes JE, Bowman ED, Leung SY, Harris CC, 2015, Increased MicroRNA-34b and-34c predominantly expressed in stromal tissues is associated with poor prognosis in human colon cancer. PLoS One 10(4)., DOI 10.1371/journal.pone. 0124899 Ach RA, Wang H, Curry B, 2008, Measuring microRNAs: comparisons of microarray and quantitative PCR measurements, and of different total RNA prep methods. BMC Biotechnol 8:69., DOI 10. 1186/1472-6750-8-69 Jung M, Schaefer A, Steiner I, Kempkensteffen C, Stephan C, Erbersdobler A, Jung K, 2010, Robust microRNA stability in degraded RNA preparations from human tissue and cell samples. Clin Chem 56(6):998–1006., DOI 10.1373/clinchem.2009.141580 Kotorashvili A, Ramnauth A, Liu C, Lin J, Ye K, Kim R, Hazan R, Rohan T, Fineberg S, Loudig O, 2012, Effective DNA/RNA Coextraction for analysis of MicroRNAs, mRNAs, and genomic DNA from formalin-fixed paraffin-embedded specimens. PLoS One 7(4)., DOI 10.1371/journal.pone.0034683 Ellis-Connell AL, Iempridee T, Xu I, Mertz JE, 2010, Cellular MicroRNAs 200b and 429 regulate the Epstein-Barr virus switch between latency and lytic replication. J Virol 84(19):10329–10343., DOI 10.1128/Jvi.00923-10 Ulivi P, Foschi G,MengozziM, Scarpi E, Silvestrini R,Amadori D, Zoli W, 2013, Peripheral blood miR-328 expression as a potential biomarker for the early diagnosis of NSCLC. Int J Mol Sci 14(5): 10332–10342., DOI 10.3390/ijms140510332 Gao J, Li N, Dong Y, Li S, Xu L, Li X, Li Y, Li Z, Ng SS, Sung JJ, Shen L, Yu J, 2014, miR-34a-5p suppresses colorectal cancer metastasis and predicts recurrence in patients with stage II/III colorectal cancer. Oncogene., DOI 10.1038/onc.2014.348 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 505 EP 513 DI 10.1007/s12253-015-0027-4 PG 9 ER PT J AU Erol, O Suren, D Tutus, B Toptas, T Gokay, AA Derbent, UA Ozel, KM Sezer, C AF Erol, Onur Suren, Dinc Tutus, Birsel Toptas, Tayfun Gokay, Arda Ahmet Derbent, Uysal Aysel Ozel, Kemal Mustafa Sezer, Cem TI Immunohistochemical Analysis of E-Cadherin, p53 and Inhibin-α Expression in Hydatidiform Mole and Hydropic Abortion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE E-cadherin; p53; Inhibin-α; Hydatidiform mole ID E-cadherin; p53; Inhibin-α; Hydatidiform mole AB The purpose of this study was to investigate the role of E-cadherin, p53, and inhibin-α immunostaining in the differential diagnosis of hydropic abortion (HA), partial hydatidiform mole (PHM), and complete hydatidiform mole (CHM). E-cadherin, p53, and inhibin-α protein expression patterns were investigated immunohistochemically using paraffin -embedded tissue sections from histologically diagnosed cases of HA (n=23), PHM (n= 24), and CHM (n=23). Expression patterns of these markers were scored semi-quantitatively according to the staining intensity, percentage of positive cells, and immunoreactivity score. Classification of cases was established on histologic criteria and supported by the molecular genotyping. Immunostaining allowed the identification of specific cell types with E-cadherin, p53, and inhibin-α expression in all cases. E-cadherin expression was detected on the cell surface of villous cytotrophoblasts. We observed a marked decline in the expression of E-cadherin from HAs to PHMs to CHMs. The p53-positive reaction was restricted to the nucleus of villous cytotrophoblasts. Significantly increased p53 expression was observed in CHMs, compared with HAs and PHMs. The expression of inhibin-α was localised in the cytoplasm of villous syncytiotrophoblasts, and the expression of this marker was significantly higher in PHMs and CHMs than HAs. In conclusion, immunohistochemical analysis of E-cadherin, p53, and inhibin-α expression could serve as a useful adjunct to conventional methods in the differential diagnosis of HA, PHM, and CHM. C1 [Erol, Onur] Antalya Training and Research Hospital, Department of Obstetrics and GynecologyAntalya, Turkey. [Suren, Dinc] Antalya Training and Research Hospital, Department of PathologyAntalya, Turkey. [Tutus, Birsel] Antalya Training and Research Hospital, Department of PathologyAntalya, Turkey. [Toptas, Tayfun] Antalya Training and Research Hospital, Department of Gynecologic OncologyAntalya, Turkey. [Gokay, Arda Ahmet] Antalya Training and Research Hospital, Department of PathologyAntalya, Turkey. [Derbent, Uysal Aysel] Antalya Training and Research Hospital, Department of Obstetrics and GynecologyAntalya, Turkey. [Ozel, Kemal Mustafa] Antalya Training and Research Hospital, Department of Obstetrics and GynecologyAntalya, Turkey. [Sezer, Cem] Antalya Training and Research Hospital, Department of PathologyAntalya, Turkey. RP Erol, O (reprint author), Antalya Training and Research Hospital, Department of Obstetrics and Gynecology, Antalya, Turkey. EM dronurerol@hotmail.com CR Cheung AN, 2003, Pathology of gestational trophoblastic disease. Baillieres Best Pract Res Clin Obstet Gynaecol 17:849–868 Fukunaga M, Katabuchi H, Nagasaka T, Mikami Y, Minamiguchi S, Lage JM(2005, Interobserver and intraobserver variability in the diagnosis of hydatidiform mole. Am J Surg Pathol 29:942–947 Jun SY, Ro JY, Kim KR, 2003, P57kip2 is useful in the classification and differential diagnosis of complete and partial hydatidiform moles. Histopathology 43:17–25 Rowlands TM, Symonds JM, Farookhi R, Blaschuk OW, 2000, Cadherins: crucial regulators of structure and function in reproductive tissues. Rev Reprod 5:53–61 Yuan W, Chen Z, Wu S, Ge J, Chang S, Wang X, Chen J, Chen Z, 2009, Expression of EphA2 and E-cadherin in gastric cancer: correlated with tumor progression and lymphogenous metastasis. Pathol Oncol Res 15:473–478 Soussi T, Lozano G, 2005, p53 mutation heterogeneity in cancer. Biochem Biophys Res Commun 331:834–842 Lapolt PS, Hsueh AJW, 1991, Moleculer basis of inhibin production and action. Mol Cell Neurosci 2:449–463 Matias-Guiu X, Lerma E, Prat J, 1998, Current topics in pathology of gynecologic tumors: a selective review. Int J Surg Pathol 6:121–134 Stenvers KL, Findlay JK, 2010, Inhibins: from reproductive hormones to tumor suppressors. Trends EndocrinolMetab 21:174–180 WellsM(2007, The pathology of gestational trophoblastic disease: recent advances. Pathology 39:88–96 Murphy KM, Mc Connell TG, Hafez MJ, Vang R, Ronnett BM, 2009, Molecular genotyping of hydatidiform moles: analytic validation of a multiplex short tandem repeat assay. J Mol Diagn 11: 598–605 HusseinMR, 2009, Analysis of p53, BCL-2 and epidermal growth factor receptor protein expression in the partial and complete hydatidiform moles. Exp Mol Pathol 87:63–69 Soper JT, 2006, Gestational trophoblastic disease. Obstet Gynecol 108:176–187 Kimber SJ, 2000)Molecular interactions at the maternal-embryonic interface during the early phase of implantation. Semin Reprod Med 18:237–253 Floridon C, Nielsen O, Holund B, Sunde L, Westergaard JG, Thomsen SG, Teisner B, 2000, Localization of E-cadherin in villous, extravillous and vascular trophoblasts during intrauterine, ectopic and molar pregnancy. Mol Hum Reprod 6:943–950 Brown LM, Lacey HA, Baker PN, Crocker IP, 2005, E-cadherin in the assessment of aberrant placental cytotrophoblast turnover in pregnancies complicated by preeclampsia. Histochem Cell Biol 124:499–506 Batistatou A, Makrydimas G, Zagorianakou N, Zagorianakou P, Nakanishi Y, Agnantis NJ, Hirohashi S, Charalabopoulos K, 2007, Expression of dysadherin and E-cadherin in trophoblastic tissue in normal and abnormal pregnancies. Placenta 28:590–592 Shu H, Chen H, Yang B, Chang Z, Xiong M, Chen W, 2013, Aberrant expression of E-cadherin and integrin β-1 in trophoblasts is associated with malignant gestational trophoblastic diseases. Int J Gynecol Cancer 23:749–754 Xue WC, Feng HC, Tsao SW, Chan KY, Ngan HY, Chiu PM, Maccalman CD, CheungAN, 2003, Methylation status and expression of E-cadherin and cadherin-11 in gestational trophoblastic diseases. Int J Gynecol Cancer 13:879–888 Balaram P, Alex S, Panikkar B, Rajalekshmi TN, 2004, Adhesionrelated proteins E-cadherin, P-cadherin, CD44, and CD44v6, and antimetastatic protein nm23H1 in complete hydatidiform moles in relation to invasion potential. Int J Gynecol Cancer 14:532–539 Cohen M, Meisser A, Haenggeli L, Irminger-Finger I, Bischof P, 2007, Status of p53 in first-trimester cytotrophoblastic cells. Mol Hum Reprod 13:111–116 Marzusch K, Ruck P, Horny HP, Dietl J, Kaiserling E, 1995, Expression of the p53 tumour suppressor gene in human placenta: an immunohistochemical study. Placenta 16:101–104 Halperin R, Peller S, Sandbank J, Bukovsky I, Schneider D, 2000, Expression of the p53 gene and apoptosis in gestational trophoblastic disease. Placenta 21:58–62 Kale A, Soylemez F, Ensari A, 2001, Expressions of proliferation markers, Ki-67, proliferating cell nuclear antigen, and silverstaining nucleolar organizer regions, and of p53 tumor protein in gestational trophoblastic disease. Am J Obstet Gynecol 184:567– 574 Petignat P, Laurini R, Goffin F, Bruchim I, Bischof P, 2006, Expression of matrix metalloproteinase-2 and mutant p53 is increased in hydatidiform mole as compared with normal placenta. Int J Gynecol Cancer 16:1679–1684 Al-Bozom IA, 2000, p53 and Bcl-2 oncoprotein expression in placentas with hydropic changes and partial and complete moles. APMIS 108:756–760 Chen Y, Shen D, Gu Y, Zhong P, Xie J, Song Q, 2012, The diagnostic value of Ki-67, P53 and P63 in distinguishing partial hydatidiform mole from hydropic abortion. Wien Klin Wochenschr 124:184–187 Petraglia F, 1997, Inhibin, activin, and follistatin in the human placenta: a new family of regulatory proteins. Placenta 18:3–8 Mylonas I, Jeschke U,Wiest I, Hoeing A, Vogl J, Shabani N, Kuhn C, Schulze S, Kupka MS, Friese K, 2004, Inhibin/activinsubunits alpha, beta-A and beta-B are differentially expressed in normal human endometrium throughout the menstrual cycle. Histochem Cell Biol 122:461–471 de Kretser DM, HedgerMP LKL, Phillips DJ, 2002, Inhibins, activins and follistatin in reproduction. Hum Reprod Update 8: 529–541 Mylonas I, Schiessl B, Jeschke U, Vogl J,Makrigiannakis A, Kuhn C, Schulze S, Kainer F, Friese K, 2006, Expression of inhibin/ activin subunits alpha, −alpha), betaA, −betaA), and betaB, betaB, in placental tissue of normal, preeclamptic, and HELLP pregnancies. Endocr Pathol 17:19–33 Shih IM, Kurman RJ, 1999, Immunohistochemical localization of inhibin-alpha in the placenta and gestational trophoblastic lesions. Int J Gynecol Pathol 18:144–150 Kommoss F, Schmidt D, Coerdt W, Olert J, Muntefering H, 2001, Immunohistochemical expression analysis of inhibin-alpha and - beta subunits in partial and complete moles, trophoblastic tumors, and endometrial decidua. Int J Gynecol Pathol 20:380–385 Mylonas I, Shabani N, Vogl J, Makovitzky J, Kunze S, Kuhn C, Schulze S, Friese K, Jeschke U, 2007, Inhibin/activin subunits are immunohistochemically expressed in complete and partial hydatidiform moles. Anticancer Res 27:1995–2000 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 515 EP 521 DI 10.1007/s12253-015-0031-8 PG 7 ER PT J AU Nemejcova, K Ticha, I Kleiblova, P Bartu, M Cibula, D Jirsova, K Dundr, P AF Nemejcova, Kristyna Ticha, Ivana Kleiblova, Petra Bartu, Michaela Cibula, David Jirsova, Katerina Dundr, Pavel TI Expression, Epigenetic and Genetic Changes of HNF1B in Endometrial Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Clear cell carcinoma; Endometrioid carcinoma; HNF-1-beta; Immunohistochemistry; Mutation analysis; Methylation ID Clear cell carcinoma; Endometrioid carcinoma; HNF-1-beta; Immunohistochemistry; Mutation analysis; Methylation AB Hepatocyte nuclear factor 1-beta (HNF-1-beta) is a transcription factor involved in cancerogenesis of various tumors, including endometrioid carcinoma. We performed comprehensive analysis of HNF-1-beta in lesions of the endometrium, including protein expression and genetic and epigenetic changes. Expression of HNF-1-beta was analyzed immunohistochemically in 320 cases including both tumor and non-tumor endometrial lesions. Promoter methylation and genetic variants were evaluated, using bisulphite and direct sequencing, in 30 (18 fresh frozen, 12 FFPE tumors) endometrioid carcinomas (ECs) and 15 ovarian clear cell carcinomas (OCCCs) as a control group. We detected expression of HNF-1-beta in 28 % of ECs (51/180 cases), 26 % of serous carcinoma (7/27 cases), 83 % of endometrial clear cell carcinoma (15/18 cases), 93 % of hyperplastic polyps with atypias (13/14 cases), 100 % of hyperplastic polyps without atypias (16/16 cases), 88% of hyperplasias with atypias (14/16 cases), 91 %of hyperplasias without atypias (10/11 cases), and in ≥80 % of different normal endometrium samples. The control group of OCCCs showed HNF-1-beta expression in 95 % (18/19 cases). Methylation in promoter region was detected in 13.3 % (4/30) of ECs, but not in corresponding normal tissue where available, nor in OCCCs (0/15 cases). Mutation analysis revealed truncating variant c.454C > T (p.Gln152X) in one EC and missense variant c.848C > T (p.Ala283Val) was detected in one OCCC. In conclusion, expression of HNF-1-beta was detected in various extents in all types of lesions analyzed, nevertheless its strong expressionwasmostly limited to clear cell carcinomas. Biological significance of genetic and epigenetic changes needs further investigation. C1 [Nemejcova, Kristyna] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 2 12800 Prague, Czech Republic. [Ticha, Ivana] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 2 12800 Prague, Czech Republic. [Kleiblova, Petra] Charles University in Prague, First Faculty of Medicine, General University Hospital, Institute of Biochemistry and Experimental OncologyPrague, Czech Republic. [Bartu, Michaela] Charles University, 1st Faculty of Medicine and General Teaching HospitalPrague, Czech Republic. [Cibula, David] Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Obstetrics and GynecologyPrague, Czech Republic. [Jirsova, Katerina] Charles University in Prague, First Faculty of Medicine, Bank of Biological MaterialPrague, Czech Republic. [Dundr, Pavel] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 2 12800 Prague, Czech Republic. RP Dundr, P (reprint author), Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, 2 12800 Prague, Czech Republic. EM pdundr@seznam.cz CR Barbacci E, Chalkiadaki A, Masdeu C, et al., 2004, HNF1β/TCF2 mutations impair transactivation potential through altered coregulator recruitment. Hum Mol Genet 13:3139–3149 Cereghini S, 1996, Liver-enriched transcription factors and hepatocyte differentiation. FASEB J 10:267–282 Bach I, Yaniv M, 1993, More potent transcriptional activators or a transdominant inhibitor of the HNF1 homeoprotein family are generated by alternative RNA processing. EMBO J 12:4229–4242 Erratum in: EMBO J 1994 13:492 Horikawa Y, Iwasaki N, Hara M, et al., 1997, Mutation in hepatocyte nuclear factor-1-beta gene, TCF2, associated withMODY. Nat Genet 17:384–385 Tsuchiya A, Sakamoto M, Yasuda J, et al., 2003, Expression profiling in ovarian clear cell carcinoma: identification of hepatocyte nuclear factor-1 β as a molecular marker and a possible molecular target for therapy of ovarian clear cell carcinoma. Am J Pathol 163: 2503–2512 Suzuki E, Kajita S, Takahashi H, Matsumoto T, Tsuruta T, Saegusa M, 2015, Transcriptional upregulation of HNF-1β by NF-κB in ovarian clear cell carcinoma modulates susceptibility to apoptosis through alteration in bcl-2 expression. Lab Investig 95:962–972., DOI 10.1038/labinvest.2015.73 Pontoglio M, 2000, Hepatocyte nuclear factor 1, a transcription factor at the crossroads of glucose homeostasis. J Am Soc Nephrol 16:140–143 Bellanne-Chantelot C, Chauveau D, Gautier JF, et al., 2004, Clinical spectrum associated with hepatocyte nuclear factor-1β mutations. Ann Intern Med 140:510–517 Lokmane L, Haumaitre C, Garcia-Villalba P, Anselme I, Schneider- Maunoury S, Cereghini S, 2008, Crucial role of vHNF1 in vertebrate hepatic specification. Development 135:2777–2786., DOI 10. 1242/dev.023010 Harries LW, Perry JR, McCullagh P, Crundwell M, 2010, Alterations in LMTK2, MSMB and HNF1B gene expression are associated with the development of prostate cancer. BMC Cancer 10:315., DOI 10.1186/1471-2407-10-315 Shen H, Fridley BL, Song H, et al., 2013, Epigenetic analysis leads to identification of HNF1B as a subtype-specific susceptibility gene for ovarian cancer. Nat Commun 4:1628., DOI 10. 1038/ncomms2629 Terasawa K, Toyota M, Sagae S, et al., 2006, Epigenetic inactivation of TCF2 in ovarian cancer and various cancer cell lines. Br J Cancer 94:914–921 Kato N, Motoyama T, 2009, Hepatocyte nuclear factor-1beta(HNF- 1beta, in human urogenital organs: its expression and role in embryogenesis and tumorigenesis. Histol Histopathol 24:1479–1486 Yamamoto S, Tsuda H, Aida S, Shimazaki H, Tamai S, Matsubara O, 2007, Immunohistochemical detection of hepatocyte nuclear factor 1β in ovarian and endometrial clear-cell adenocarcinomas and nonneoplastic endometrium. Hum Pathol 38:1074–1080 Kato N, Sasou S, Motoyama T, 2006, Expression of hepatocyte nuclear factor-1β, HNF-1β, in clear cell tumors and endometriosis of the ovary. Mod Pathol 19:83–89 Kato N, Tamura G, Motoyama T, 2008, Hypomethylation of hepatocyte nuclear factor-1β, HNF-1β, CpG island in clear cell carcinoma of the ovary. Virchows Arch 452:175–180 Park KJ, Kiyokawa T, Soslow RA, Lamb CA, Oliva E, Zivanovic O, JuretzkaMM, Pirog EC, 2011, Unusual endocervical adenocarcinomas: an immunohistochemical analysis with molecular detection of human papillomavirus. Am J Surg Pathol 35:633–646., DOI 10.1097/PAS.0b013e31821534b9 Kenny SL, McBride HA, Jamison J, McCluggage WG, 2012, Mesonephric adenocarcinomas of the uterine cervix and corpus: HPV-negative neoplasms that are commonly PAX8, CA125, and HMGA2 positive and that may be immunoreactive with TTF1 and hepatocyte nuclear factor 1-β. Am J Surg Pathol 36:799–807., DOI 10.1097/PAS.0b013e31824a72c6 Fadare O, Liang SX, 2012, Diagnostic utility of hepatocyte nuclear factor 1-beta immunoreactivity in endometrial carcinomas: lack of specificity for endometrial clear cell carcinoma. Appl Immunohistochem Mol Morphol 20:580–587., DOI 10.1097/ PAI.0b013e31824973d1 Kalloger SE, Kobel M, Leung S, et al., 2011, Calculator for ovarian carcinoma subtype prediction. Mod Pathol 24:512–521., DOI 10. 1038/modpathol.2010.215 Tomassetti A, De Santis G, Castellano G, Miotti S, Mazzi M, Tomasoni D, Van Roy F, Carcangiu ML, Canevari S, 2008, Variant HNF1 modulates epithelial plasticity of normal and transformed ovary cells. Neoplasia 10:1481–1492 Forbes SA, Bindal N, Bamford S, et al., 2011, COSMIC: mining complete cancer genomes in the Catalogue of SomaticMutations in Cancer. Nucleic Acids Res 39(Database issue):D945–D950., DOI 10. 1093/nar/gkq929 Kim L, Liao J, Zhang M, Talamonti M, Bentrem D, Rao S, Yang GY, 2008, Clear cell carcinoma of the pancreas: histopathologic features and a unique biomarker: hepatocyte nuclear factor-1beta. Mod Pathol 21:1075–1083., DOI 10.1038/modpathol.2008.95 Buchner A, Castro M, Hennig A, Popp T, Assmann G, Stief CG, Zimmermann W, 2010, Downregulation of HNF-1B in renal cell carcinoma is associated with tumor progression and poor prognosis. Urology 76:507e6–50711., DOI 10.1016/j.urology.2010.03.042 Hoang LN,Han G, McConechyM, et al., 2014, Immunohistochemical characterization of prototypical endometrial clear cell carcinoma–diagnostic utility of HNF-1β and oestrogen receptor. Histopathology 64: 585–596., DOI 10.1111/his.12286 Painter JN, O’Mara TA, Batra J, et al., 2015, Fine-mapping of the HNF1B multicancer locus identifies candidate variants that mediate endometrial cancer risk. Hum Mol Genet 24:1478–1492., DOI 10. 1093/hmg/ddu552 Spurdle AB, Thompson DJ, Ahmed S, et al., 2011, Genome-wide association study identifies a common variant associated with risk of endometrial cancer. Nat Genet 43:451–454., DOI 10.1038/ng.812 Mandato VD, Farnetti E, Torricelli F, et al., 2015, HNF1B polymorphism influence the prognosis of endometrial cancer patients: a cohort study. BMC Cancer 15:229., DOI 10.1186/s12885-015-1246-5 Pharoah PD, Tsai YY, Ramus SJ, et al., 2013, GWAS meta-analysis and replication identifies three new susceptibility loci for ovarian cancer. Nat Genet 45:362–370., DOI 10.1038/ng.2564 NotoH,Osame K, Sasazuki T, NodaM(2010, Substantially increased risk of cancer in patients with diabetes mellitus: a systematic review and meta-analysis of epidemiologic evidence in Japan. J Diabetes Complicat 24:345–353., DOI 10.1016/j.jdiacomp.2010.06.004 Rebouissou S, Vasiliu V, Thomas C, et al., 2005, Germline hepatocyte nuclear factor 1alpha and 1beta mutations in renal cell carcinomas. Hum Mol Genet 14:603–614 Nemejcova K, Cibula D, Dundr P, 2015, Expression of HNF-1β in cervical carcinomas: an immunohistochemical study of 155 cases. Diagn Pathol 10:8., DOI 10.1186/s13000-015-0245-9 Nemejcova K, Kenny SL, Laco J, et al., 2015, Atypical Polypoid Adenomyoma of the Uterus: An Immunohistochemical and Molecular Study of 21 Cases. Am J Surg Pathol 39:1148–1155., DOI 10.1097/PAS.0000000000000428 Lim D, Ip PP, Cheung AN, Kiyokawa T, Oliva E, 2015, Immunohistochemical Comparison of Ovarian and Uterine Endometrioid Carcinoma, Endometrioid Carcinoma With Clear Cell Change, and Clear Cell Carcinoma. Am J Surg Pathol 39: 1061–1069., DOI 10.1097/PAS.0000000000000436 Fadare O, Zhao C, Khabele D, et al., 2015, Comparative analysis of Napsin A, alpha-methylacyl-coenzyme A racemase, AMACR, P504S), and hepatocyte nuclear factor 1 beta as diagnostic markers of ovarian clear cell carcinoma: an immunohistochemical study of 279 ovarian tumours. Pathology 47:105–111., DOI 10.1097/PAT. 0000000000000223 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 523 EP 530 DI 10.1007/s12253-015-0037-2 PG 8 ER PT J AU Shen, X Guo, Y Qi, J Shi, W Wu, X Ni, H Ju, Sh AF Shen, Xianjuan Guo, Yuehua Qi, Jing Shi, Wei Wu, Xinhua Ni, Hongbing Ju, Shaoqing TI Study on the Association Between miRNA-202 Expression and Drug Sensitivity in Multiple Myeloma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bcell-activating factor (BAFF); Drug resistance; Multiple myeloma (MM); microRNA; Signaling pathway ID Bcell-activating factor (BAFF); Drug resistance; Multiple myeloma (MM); microRNA; Signaling pathway AB An increasing amount of experimental evidence has shown that miRNAs play a causal role in hematologic tumorigenesis. In this study, we characterized the role of miR-202 in multiple myeloma (MM) drug sensitivity. The potential binding site of miR-202 and B cell-activating factor (BAFF) was confirmed by luciferase reporter assay. MM cells were transfected with miR-202 mimics and inhibitor. Cells growth was measured by WST-1 cell proliferation assay and Annexin V-FLUOS apoptosis assay. BAFF and miR-202 mRNA levels were measured by real-time PCR. Meanwhile, BAFF, Bcl-2 family survival proteins and MAPK pathway proteins were measured by Western blot. It was found that miR-202 was functioned as a modulator of BAFF expression. miR-202 over-expression sensitized MM cells to bortezomib (Bort) but less to Thalidomide (Thal) and dexamethasone (Dex). miR-202 mimics in combination with Bort inhibited MM cell survival more effectively as compared with Bort treatment alone. Our study also provided experimental evidence that JNK/SAPK signaling pathway was involved in the regulatory effect of miR-202 on drug resistance of MM cells. These results suggest that the regulatory mechanism of miR-202 expression may be a promising target for fine-tuning anti-myeloma therapy. C1 [Shen, Xianjuan] Affiliated Hospital of Nantong University, Surgical Comprehensive Laboratory, #20 Xisi Road, 226001 Nantong, JS, China. [Guo, Yuehua] Nantong University, #9 Seyuan Road, 226001 Nantong, JS, China. [Qi, Jing] Affiliated Hospital of Nantong University, Surgical Comprehensive Laboratory, #20 Xisi Road, 226001 Nantong, JS, China. [Shi, Wei] Affiliated Hospital of Nantong University, Surgical Comprehensive Laboratory, #20 Xisi Road, 226001 Nantong, JS, China. [Wu, Xinhua] Affiliated Hospital of Nantong University, Surgical Comprehensive Laboratory, #20 Xisi Road, 226001 Nantong, JS, China. [Ni, Hongbing] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, #20 Xisi Road, 226001 Nantong, JS, China. [Ju, Shaoqing] Affiliated Hospital of Nantong University, Surgical Comprehensive Laboratory, #20 Xisi Road, 226001 Nantong, JS, China. RP Ju, Sh (reprint author), Affiliated Hospital of Nantong University, Surgical Comprehensive Laboratory, 226001 Nantong, China. EM jsq814@hotmail.com;274391898@qq.com CR Comert M, Gunes AE, Sahin F, Saydam G, 2013, Quality of Life and Supportive Care inMultipleMyeloma. Turk J Haematol 30(3): 234–246 Andrews SW, Kabrah S, May JE, Donaldson C, Morse HR, 2013, Multiple myeloma: the bone marrow microenvironment and its relation to treatment. Br J Biomed Sci 70(3):110–120 Terpos E, Christoulas D, 2013, Effects of proteasome inhibitors on bone cancer. Bonekey Rep 2:395 Zheng Y, Cai Z,Wang S, Zhang X, Qian J, Hong S, Li H,Wang M, Yang J, Yi Q, 2009, Macrophages are an abundant component of myeloma microenvironment and protect myeloma cells from chemotherapy drug-induced apoptosis. Blood 114(17):3625–3628 Fuchs O, 2013, Targeting of NF-kappaB signaling pathway, other signaling pathways and epigenetics in therapy of multiple myeloma. Cardiovasc Hematol Disord Drug Targets 13(1):16–34 Calin GA, Croce CM, 2006, MicroRNA signatures in human cancers. Nat Rev Cancer 6(11):857–866 Kong YW, Ferland-McCollough D, Jackson TJ, Jackson TJ, Bushell M, 2012, microRNAs in cancer management. Lancet Oncol 13(6):e249–e258 Corsini LR, Bronte G, Terrasi M, Amodeo V, Fanale D, Fiorentino E, Cicero G, Bazan V, Russo A, 2012, The role of microRNAs in cancer: diagnostic and prognostic biomarkers and targets of therapies. Expert Opin Ther Targets 16(Suppl 2):S103–S109 Pichiorri F, Suh SS, Ladetto M, Kuehl M, Palumbo T, Drandi D, Taccioli C, Zanesi N, Alder H, Hagan JP, Munker R, Volinia S, Boccadoro M, Garzon R, Palumbo A, Aqeilan RI, Croce CM, 2008, MicroRNAs regulate critical genes associated with multiple myeloma pathogenesis. Proc Natl Acad Sci U S A 105(35):12885– 12890 Corthals SL, Sun SM, Kuiper R, de Knegt Y, Broyl A, van der Holt B, Beverloo HB, Peeters JK, el Jarari L, Lokhorst HM, Zweegman S, Jongen-LavrencicM, Sonneveld P, 2011, MicroRNA signatures characterize multiple myeloma patients. Leukemia 25(11):1784– 1789 Gao SM, Xing CY, Chen CQ, Lin SS, Dong PH, Yu FJ, 2011, miR- 15a and miR-16-1 inhibit the proliferation of leukemic cells by down-regulating WT1 protein level. J Exp Clin Cancer Res 30:110 Hao M, Zhang L, An G, Meng H, Han Y, Xie Z, Xu Y, Li C, Yu Z, Chang H, Qiu L, 2011, Bone marrow stromal cells protect myeloma cells from bortezomib induced apoptosis by suppressing microRNA-15a expression. Leuk Lymphoma 52(9):1787–1794 Fisher RI, Bernstein SH, Kahl BS, Djulbegovic B, Robertson MJ, de Vos S, Epner E, Krishnan A, Leonard JP, Lonial S, Stadtmauer EA, O’Connor OA, Shi H, Boral AL, Goy A, 2006, Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma. J Clin Oncol 24:4867–4874 Xu G, Shen XJ, Pu J, Chu SP, Wang XD, Wu XH, Sun CJ, Zhang X, Zhu BL, Ju SQ, 2012, BLyS expression and JNKactivation may form a feedback loop to promote survival and proliferation of multiple myeloma cells. Cytokine 60(2):505–513 Rajkumar SV, 2012, Multiple myeloma: 2012 update on diagnosis, risk-stratification, and management. Am J Hematol 87, 1):78–88 Fonseca R, Monge J, Dimopoulos MA, 2014, Staging and prognostication of multiple myeloma. Expert Rev Hematol 7, 1):21–31 Fragioudaki M, Boula A, Tsirakis G, Psarakis F, Spanoudakis M, Papadakis IS, Pappa CA, Alexandrakis MG, 2012, B cellactivating factor: Its clinical significance in multiple myeloma patients. Ann Hematol 91(9):1413–1418 Fragioudaki M, Tsirakis G, Pappa CA, Aristeidou I, Tsioutis C, Alegakis A, Kyriakou DS, Stathopoulos EN, Alexandrakis MG, 2012, Serum baff levels are related to angiogenesis and prognosis in patients with multiple myeloma. Leuk Res 36(8):1004–1008 Jiang P, Yueguo W, Huiming H, Hongxiang Y, MeiW, Ju S, 2009, B-Lymphocyte stimulator: a new biomarker for multiple myeloma. Eur J Haematol 82(4):267–276 Ju S, Wang Y, Ni H, Wang X, Jiang P, Kong X, Zhong R, 2009, Correlation of expression levels of BLyS and its receptors with multiple myeloma. Clin Biochem 42(4–5):387– 399 Meads MB, Hazlehurst LA, Dalton WS, 2008, The bone marrow microenvironment as a tumor sanctuary and contributor to drug resistance. Clin Cancer Res 14(9):2519–2526 Podar K, Chauhan D, Anderson KC, 2009, Bone marrow microenvironment and the identification of new targets for myeloma therapy. Leukemia 23(1):10–24 Yu J, Qiu X, Shen X, ShiW,Wu X, Gu G, Zhu B, Ju S, 2014)miR- 202 expression concentration and its clinical significance in the serum of multiple myelomapatients. Ann Clin Biochem 51:543– 549 Tai YT, Li XF, Breitkreutz I, Song W, Neri P, Catley L, Podar K, Hideshima T, Chauhan D, Raje N, Schlossman R, Richardson P, Munshi NC, Anderson KC, 2006, Role of B-cell-activating factor in adhesion and growth of human multiple myeloma cells in the bone marrow microenvironment. Cancer Res 66(13):6675–6682 Coppo R, 2014, Proteasome inhibitors in progressive renal diseases. Nephrol Dial Transplant 29(Suppl 1):i25–i30 Wen J, Feng Y, Huang W, Chen H, Liao B, Rice L, Preti HA, Kamble RT, Zu Y, Ballon DJ, Chang CC, 2010, Enhanced antimyeloma cytotoxicity by the combination of arsenic trioxide and bortezomib is further potentiated by p38 MAPK inhibition. Leuk Res 34(1):85–92 Kobayashi T, Kuroda J, Ashihara E, Oomizu S, Terui Y, Taniyama A, Adachi S, Takagi T, Yamamoto M, Sasaki N, Horiike S, Hatake K, Yamauchi A, Hirashima M, Taniwaki M, 2010, Galectin-9 exhibits anti-myeloma activity through JNK and p38 MAP kinase pathways. Leukemia 4:843–850 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 531 EP 539 DI 10.1007/s12253-015-0035-4 PG 9 ER PT J AU Qiao, D Li, M Pu, J Wang, W Zhu, W Liu, H AF Qiao, Dongfeng Li, Ming Pu, Juan Wang, Wanwei Zhu, Weiguo Liu, Haiyan TI Loss of Protein Tyrosine Phosphatase Receptor J Expression Predicts an Aggressive Clinical Course in Patients with Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Protein tyrosine phosphatase receptor J; Esophageal squamous cell carcinoma; Progression; Migration; Invasion ID Protein tyrosine phosphatase receptor J; Esophageal squamous cell carcinoma; Progression; Migration; Invasion AB Protein Tyrosine Phosphatase Receptor J (PTPRJ) has been reported to be a tumor suppressor in various human cancers. The aim of this study was to investigate the clinical significance of PTPRJ in ESCC patients and its effects on biological behaviors of ESCC cells. PTPRJ expression, at mRNA and protein levels, were respectively detected by quantitative real-time PCR, western blot and immunohistochemistry, based on 106 newly diagnosed ESCC patients. The associations between PTPRJ expression and clinicopathological characteristics of ESCC patients were statistically analyzed. Then, the effects of PTPRJ in migration and invasion were determined by wound healing and transwell assays based on ESCC cell line transfected with siRNA or expression vector of PTPRJ. Expression of PTPRJ at mRNA and protein levels were both significantly lower in ESCC tissues than those in normal esophageal mucosa. Immunohistochemistry showed that PTPRJ protein was localized in the cytoplasm of cancer cells in ESCC tissues. In addition, PTPRJ downregulation was found to be closely correlated with advanced tumor stage (P = 0.01) and poor differentiation (P = 0.03).Moreover, knockdown of PTPRJ in KYSE510 cells could significantly promote cell migration and invasion (both P < 0.05), which were reversed by the restoration of PTPRJ expression in vitro (both P < 0.05). Our data offer the convincing evidence that loss of PTPRJ expression may predict an aggressive clinical course in ESCC patients. PTPRJ may function as a tumor suppressor and play an important role in the regulation of ESCC cell motility, suggesting its potentials as a therapeutic agent for human ESCC. C1 [Qiao, Dongfeng] People’s Hospital of Lianshui County, Department of Radiotherapy Pathology, 223400 Lianshui, China. [Li, Ming] People’s Hospital of Xuyi County, Department of Radiotherapy Pathology, 211700 Xuyi, China. [Pu, Juan] People’s Hospital of Lianshui County, Department of Radiotherapy Pathology, 223400 Lianshui, China. [Wang, Wanwei] People’s Hospital of Lianshui County, Department of Radiotherapy Pathology, 223400 Lianshui, China. [Zhu, Weiguo] Nanjing Medical University, Huai’an First People’s Hospital, Department of Radiotherapy Oncology, 223300 Huainan, China. [Liu, Haiyan] Nanjing Medical University, Huai’an First People’s Hospital, Department of Pathology, 223300 Huainan, China. RP Zhu, W (reprint author), Nanjing Medical University, Huai’an First People’s Hospital, Department of Radiotherapy Oncology, 223300 Huainan, China. EM jshazwg@126.com CR Baba Y, Watanabe M, Yoshida N, Baba H, 2014, Neoadjuvant treatment for esophageal squamous cell carcinoma. World J Gastrointest Oncol 6:121–128 Shang L, Wang M, 2013, Molecular alterations and clinical relevance in esophageal squamous cell carcinoma. Front Med 7:401– 410 Li JS, Ying JM, Wang XW, Wang ZH, Tao Q, Li LL, 2013, Promoter methylation of tumor suppressor genes in esophageal squamous cell carcinoma. Chin J Cancer 32:3–11 Kruger J, Brachs S, Trappiel M, Kintscher U, Meyborg H, Wellnhofer E, Thone-Reineke C, Stawowy P, Ostman A, Birkenfeld AL, Bohmer FD, Kappert K, 2015, Enhanced insulin signaling in density-enhanced phosphatase-1, DEP-1, knockout mice. Mol Metab 4:325–336 Petermann A, Stampnik Y, Cui Y, Morrison H, Pachow D, Kliese N, Mawrin C, Bohmer FD, 2015, Deficiency of the proteintyrosine phosphatase DEP-1/PTPRJ promotes matrix metalloproteinase-9 expression in meningioma cells. J Neuro- Oncol 122:451–459 Spring K, Lapointe L, Caron C, Langlois S, Royal I, 2014, Phosphorylation of DEP-1/PTPRJ on threonine 1318 regulates Src activation and endothelial cell permeability induced by vascular endothelial growth factor. Cell Signal 26:1283– 1293 Ortuso F, Paduano F, Carotenuto A, Gomez-Monterrey I, Bilotta A, Gaudio E, Sala M, Artese A, Vernieri E, Dattilo V, Iuliano R, Brancaccio D, Bertamino A, Musella S, Alcaro S, Grieco P, Perrotti N, Croce CM, Novellino E, Fusco A, Campiglia P, Trapasso F, 2013, Discovery of PTPRJ agonist peptides that effectively inhibit in vitro cancer cell proliferation and tube formation. ACS Chem Biol 8:1497–1506 Smart CE, Askarian Amiri ME, Wronski A, Dinger ME, Crawford J, Ovchinnikov DA, Vargas AC, Reid L, Simpson PT, Song S, Wiesner C, French JD, Dave RK, da Silva L, Purdon A, Andrew M, Mattick JS, Lakhani SR, Brown MA, Kellie S, 2012, Expression and function of the protein tyrosine phosphatase receptor J, PTPRJ, in normal mammary epithelial cells and breast tumors. PLoS One 7:e40742 Spring K, Fournier P, Lapointe L, Chabot C, Roussy J, Pommey S, Stagg J, Royal I, 2015, The protein tyrosine phosphatase DEP-1/ PTPRJ promotes breast cancer cell invasion and metastasis. Oncogene In press Yan CM, Zhao YL, Cai HY, Miao GY, Ma W, 2015, Blockage of PTPRJ promotes cell growth and resistance to 5-FU through activation of JAK1/STAT3 in the cervical carcinoma cell line C33A. Oncol Rep 33:1737–1744 Chen G, Peng J, Zhu W, Tao G, Song Y, Zhou X, Wang W, 2014, Combined downregulation of microRNA-133a and microRNA- 133b predicts chemosensitivity of patients with esophageal squamous cell carcinoma undergoing paclitaxel-based chemotherapy. Med Oncol 31:263 Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008. Int J Cancer 127:2893–2917 Xu X, Lan W, Jin X, Wang B, Yan H, Chen X, Lai X, Zhang L, Zhang X, Li Z, 2014, Regulated expression of PTPRJ by COX-2/ PGE2 axis in endothelial cells. PLoS One 9:e114996 Dave RK, Dinger ME, Andrew M, Askarian-Amiri M, Hume DA, Kellie S, 2013, Regulated expression of PTPRJ/CD148 and an antisense long noncoding RNA in macrophages by proinflammatory stimuli. PLoS One 8:e68306 Aya-Bonilla C, Green MR, Camilleri E, Benton M, Keane C, Marlton P, Lea R, Gandhi MK, Griffiths LR, 2013, High-resolution loss of heterozygosity screening implicates PTPRJ as a potential tumor suppressor gene that affects susceptibility to Non- Hodgkin’s lymphoma. Genes Chromosom Cancer 52:467–479 Liotta LA, Kohn EC, 2001, The microenvironment of the tumourhost interface. Nature 411:375–379 WangW, Goswami S, Sahai E, 2005, Tumor cells caught in the act of invading: their strategy for enhanced cell motility. Trends Cell Biol 15:138–145 Ridley AJ, Schwartz MA, Burridge K, 2003, Cell migration: integrating signals from front to back. Science 302:1704–1709 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 541 EP 547 DI 10.1007/s12253-015-0036-3 PG 7 ER PT J AU Guo, Yl You, K Geng, L Qiao, J AF Guo, Yan-li You, Ke Geng, Li Qiao, Jie TI Clinical Performance of APTIMA Human Papillomavirus (HPV) 16 18/45 mRNA Genotyping Testing for the Detection of Cervical Intraepithelial Neoplasia 3 (CIN3) or Cancer in a Select Group of Chinese Women SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human papillomavirus; HPV mRNA genotyping; Cervical cancer; Screening ID Human papillomavirus; HPV mRNA genotyping; Cervical cancer; Screening AB HPV type was evaluated in a select group of Chinese women that were positive with hybrid capture, and correlations were performed between the pathology found, the type of virus and a semiquantitation from the hybrid capture results. Totally 394 referred high-risk-HPV-positive women evaluated by Hybrid Capture 2 (HC-2) assay were enrolled. Before colposcopy, cervical specimens were collected from all participants and suspended into PreservCytcollection medium (Hologic Inc., Marlborough, MA), and tested with the APTIMA HPV16 18/45 mRNA assay. Colposcopy and diagnostic biopsies were done on all participants. Viral load was assessed by HC2 assay. Totally 55 women were diagnosed as CIN 3 plus cancer (≥CIN3), and the prevalence of HPV16/18/ 45 was 65.5 % (95 % confidence interval [CI], 52.9–78.0 %) among these ≥CIN3 women. Compared with the group with positive HC2 but negative HPV16/18/45, the odds ratio (OR) to identify ≥CIN3 was 6.3 (95%CI, 3.2–12.3) for HPV16 and 3.2 (95 % CI, 1.4–7.2) for HPV18/45. When using ≥CIN3 as an endpoint, the sensitivity and specificity was 65.5 % (95 % CI, 52.9–78.0 %) and 72.0 % (95 % CI, 67.2–76.8 %). In the case of HPV16/18/45 negative, no high HPV load had a statistically significant increased risk for the prevalence of ≥CIN3. HPV16, 18 and 45 infection is a major cause for ≥CIN3 in Chinese women. Women with positive HPV16/18/ 45 should be referred to colposcopy immediately. HPV load was not suitable for the further triaged of the HPV16/18/45 negative cases. C1 [Guo, Yan-li] Peking University Third Hospital, Department of Obstetrics and Gynecology, Number 49 Huayuanbei Road Haidian District, 100191 Beijing, China. [You, Ke] Peking University Third Hospital, Department of Obstetrics and Gynecology, Number 49 Huayuanbei Road Haidian District, 100191 Beijing, China. [Geng, Li] Peking University Third Hospital, Department of Obstetrics and Gynecology, Number 49 Huayuanbei Road Haidian District, 100191 Beijing, China. [Qiao, Jie] Peking University Third Hospital, Department of Obstetrics and Gynecology, Number 49 Huayuanbei Road Haidian District, 100191 Beijing, China. RP Geng, L (reprint author), Peking University Third Hospital, Department of Obstetrics and Gynecology, 100191 Beijing, China. EM gengli57@163.com CR Walboomers JM, JacobsMV,ManosMM, Bosch FX, Kummer JA, Shah KVet al, 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189:12–19 Munoz N, Bosch FX, de Sanjose S, Herrero R, Castellsague X, Shah KV et al, 2003, Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med 348:518–527 Schiffman M, Wentzensen N, Wacholder S, Kinney W, Gage JC, Castle PE, 2001, Human papillomavirus testing in the prevention of cervical cancer. J Natl Cancer Inst 103:368–383 Wright TC, Stoler MH, Behrens CM, Sharma A, Zhang G, Wright TL, 2015, Primary cervical cancer screening with human papillomavirus: end of study results from the ATHENA study using HPV as the first-line screening test. Gynecol Oncol 136:189–197 Ronco G, Dillner J, Elfstrom KM, Tunesi S, Snijders PJ, Arbyn M et al, 2014, Efficacy of HPV-based screening for prevention of invasive cervical cancer: follow-up of four European randomised controlled trials. Lancet 383:524–532 Katki HA, Schiffman M, Castle PE, Fetterman B, Poitras NE, Lorey T et al, 2013, Five-year risk of CIN3+ and cervical cancer for women who test Pap-negative, but are HPV-positive. J Low Genit Tract Dis 17:S56–S63 Saslow D, Solomon D, Lawson HW, KillackeyM, Kulasingam SL, Cain J et al, 2012, American Cancer Society, American Society for Colposcopy and Cervical Pathology, and American Society for Clinical Pathology Screening Guidelines for the Prevention and Early Detection of Cervical Cancer. Am J Clin Pathol 137:516–542 Massad LS, Einstein MH, Huh WK, Katki HA, Kinney WK, Schiffman M et al, 2013, 2012 updated consensus guidelines for the management of abnormal cervical cancer screening tests and cancer precursors. Obstet Gynecol 121:829–846 ArbynM, Anttila A, Jordan J, Ronco G, Schenck U, Segnan N et al, 2010, European Guidelines for Quality Assurance in cervical cancer screening. Second edition—summary document. Ann Oncol 21:448–458 de Cremoux P, Coste J, Sastre-Garau X, Thioux M, Bouillac C, Labbe S et al, 2003, Efficiency of the hybrid capture 2 HPV DNA test in cervical cancer screening. A study by the French Society of Clinical Cytology. Am J Clin Pathol 120:492–499 ShermanME, LorinczAT, Scott DR,Wacholder S, Castle PE,Glass AG et al, 2003, Baseline cytology, human papillomavirus testing, and risk for cervical neoplasia: a 10-year cohort analysis. J Natl Cancer Inst 95:46–52 Cuzick J, Clavel C, Petry KU, Meijer CJ, Hoyer H, Ratnam S et al, 2006, Overview of the European and North American studies on HPV testing in primary cervical cancer screening. Int J Cancer 119: 1095–1101 Ronco G, Giorgi-Rossi P, Carozzi F, Confortini M, Dalla PP, Del MA et al, 2008, Results at recruitment from a randomized controlled trial comparing human papillomavirus testing alone with conventional cytology as the primary cervical cancer screening test. J Natl Cancer Inst 100:492–501 Sideri M, Igidbashian S, Boveri S, Radice D, Casadio C, Spolti N et al, 2011, Age distribution of HPV genotypes in cervical intraepithelial neoplasia. Gynecol Oncol 121:510–513 Smith JS, Lindsay L, Hoots B, Keys J, Franceschi S,Winer R et al, 2007, Human papillo-mavirus type distribution in invasive cervical cancer and high-grade cervicallesions: a meta-analysis update. Int J Cancer 121:621–632 Li N, Franceschi S, Howell-Jones R, Snijders PJ, Clifford GM, 2011, Human papillomavirus type distribution in 30,848 invasive cervical cancers worldwide: variation by geographical region, histological type and year of publication. Int J Cancer 128:927–935 Guan P, Howell-Jones R, Li N, Bruni L, de Sanjose S, Franceschi S et al, 2012, Human papillomavirus types in 115,789 HPV-positive women: a meta-analysis from cervical infection to cancer. Int J Cancer 131:2349–2359 Clifford GM, Franceschi S, Diaz M, Munoz N, Villa LL, 2006, HPV type-distribution inwomen with and without cervical neoplastic diseases. Vaccine 24:S326–S334 Luttmer R, Berkhof J, Dijkstra MG, van Kemenade FJ, Snijders PJ, Heideman DA et al, 2015, Comparing triage algorithms using HPV DNA genotyping, HPV E7mRNA detection and cytology in highrisk HPV DNA-positive women. J Clin Virol 67:59–66 Lorincz AT, Castle PE, Sherman ME, Scott DR, Glass AG, Wacholder S et al, 2002, Viral load of human papillomavirus and risk of CIN3 or cervical cancer. Lancet 360:228–229 Castle PE, Schiffman M, Wheeler CMALTS group, 2004, Hybrid capture 2 viral load and the 2-year cumulative risk of cervical intraepithelial neoplasia grade 3 or cancer. Am J Obstet Gynecol 191:1590–1597 Castle PE, Rodriguez AC, Burk RD, Herrero R, Wacholder S, Alfaro M et al, 2009, Short term persistence of human papillomavirus and risk of cervical precancer and cancer: population based cohort study. BMJ 339:b2569–b2581 Ramanakumar AV, Goncalves O, Richardson H, Tellier P, Ferenczy A, Coutlee F et al, 2010, Human papillomavirus, HPV, types 16, 18, 31, 45 DNA loads and HPV-16 integration in persistent and transient infections in young women. BMC Infect Dis 10:326–337 Gravitt PE, Kovacic MB, Herrero R, Schiffman M, Bratti C, Hildesheim A et al, 2007, High load for most high risk human papillomavirus genotypes is associated with prevalent cervical cancer precursors but only HPV16 load predicts the development of incident disease. Int J Cancer 121:2787–2793 Flores R, Papenfuss M, Klimecki WT, Giuliano AR, 2006, Crosssectional analysis of oncogenic HPV viral load and cervical intraepithelial neoplasia. Int J Cancer 118:1187–1193 Wanram S, Limpaiboon T, Leelayuwat C, Yuenyao P, Guiney DG, LulitanondVet al, 2009, The use of viral load as a surrogatemarker in predicting disease progression for patients with early invasive cervical cancer with integrated human papillomavirus type 16. Am J Obstet Gynecol 201(79):e1–e7 Depuydt CE, Criel AM, Benoy IH, Arbyn M, Vereecken AJ, Bogers JJ, 2012, Changes in type-specific human papillomavirus load predict progression to cervical cancer. J Cell Mol Med 16: 3096–3104 Depuydt CE, Jonckheere J, BerthM, SalembierGM,Vereecken AJ, Bogers JJ, 2015, Serial type-specific human papillomavirus, HPV, load measurement allows differentiation between regressing cervical lesions and serial virion productive transient infections. Cancer Med., DOI 10.1002/cam4.473 Castle PE, Wheeler CM, Solomon D, Schiffman M, Peyton CL, ALTS Group, 2004, Interlaboratory reliability of hybrid capture 2. Am J Clin Pathol 122:238–245 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 549 EP 554 DI 10.1007/s12253-015-0029-2 PG 6 ER PT J AU Hu, B Yan, X Liu, F Zhu, Ch Zhou, H Chen, Y Liu, J Gu, X Ni, R Zhang, T AF Hu, Baoying Yan, Xia Liu, Fang Zhu, Changlai Zhou, Huiling Chen, Yuyan Liu, Jinxia Gu, Xingxing Ni, Runzhou Zhang, Tianyi TI Downregulated Expression of PTPN9 Contributes to Human Hepatocellular Carcinoma Growth and Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human hepatocellular carcinoma (HCC); PTPN9; Cell proliferation; Prognosis ID Human hepatocellular carcinoma (HCC); PTPN9; Cell proliferation; Prognosis AB Human hepatocellular carcinoma (HCC) is one of the most common malignant cancers, whose molecular mechanisms is remains largely. PTPN9 has recently been reported to play a critical role in breast cancer development. However, the role of PTPN9 in human HCC remains elusive. The present study aimed at investigating the potential role of PTPN9 in HCC. Western blot and immunohistochemistry were used to examine the expression of PTPN9 protein in HCC and adjacent non-tumorous tissues in 45 patients. Furthermore, Cell Counting Kit-8, flow cytometry and RNA interference experiments were performed to analyze the role of PTPN9 in the regulation of HCC cell proliferation. We showed that the expression level of PTPN9 was significantly reduced in HCC, compared with adjacent non-tumorous tissues. PTPN9 expression was inversely associated with Tumor size (P= 0.014), serum AFP level (P=0.004) and Ki-67 expression. Low expression of PTPN9 predicted poor survival in HCC patients. Moreover, PTPN9 interference assay that PTPN9 inhibited cell proliferation in HepG2 cells. Cell apoptosis assay revealed that, silencing of PTPN9 expression significantly reduced cell apoptosis, compared with control ShRNA treatment group. Our results suggested that PTPN9 expression was down-regulated in HCC tumor tissues, and reduced PTPN9 expression was associated with worsened overall survival in HCC patients. Depletion of PTPN9 inhibits the apoptosis and promotes the proliferation of HCC cells. C1 [Hu, Baoying] Nantong University, Medical College, Basic Medic Research Centre, 226001 Nantong, Jiangsu, China. [Yan, Xia] Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, 226001 Nantong, Jiangsu, China. [Liu, Fang] Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, 226001 Nantong, Jiangsu, China. [Zhu, Changlai] Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, 226001 Nantong, Jiangsu, China. [Zhou, Huiling] Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, 226019 Nantong, Jiangsu, China. [Chen, Yuyan] Nantong University, Medical College, Class 5, Grade 13, Clinical Medicine, 226001 Nantong, Jiangsu, China. [Liu, Jinxia] Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, 226019 Nantong, Jiangsu, China. [Gu, Xingxing] Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, 226001 Nantong, Jiangsu, China. [Ni, Runzhou] Affiliated Hospital of Nantong University, Department of Gastroenterology, 226001 Nantong, Jiangsu, China. [Zhang, Tianyi] Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, 226001 Nantong, Jiangsu, China. RP Zhang, T (reprint author), Nantong University, Jiangsu Province Key Laboratory of Neuroregeneration, 226001 Nantong, China. EM ntdxzty@126.com CR Wenzh El-Serag HB, 2011, Hepatocellular carcinoma. N Engl J Med 365:1118–1127 El-Serag HB, Rudolph KL, 2007, Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 132: 2557–2576 But D-Y-K, Lai C-L, Yuen M-F, 2008, Natural history of hepatitisrelated hepatocellular carcinoma. World J Gastroenterol 14:1652– 1656 Poon RT, Ng IO, Fan ST, Lai EC, Lo CM, Liu CL et al, 2001, Clinicopathologic features of long-term survivors and disease-free survivors after resection of hepatocellular carcinoma: a study of a prospective cohort. J Clin Oncol 19:3037–3044 Frau M, Biasi F, Feo F et al, 2010, Prognostic markers and putative therapeutic targets for hepatocellular carcinoma. Mol Asp Med 31: 179–193 Tonks NK, 2006, Nat Rev Mol Cell Biol 7:833–846 den Hertog J, Ostman A, Bohmer FD, 2008, Protein tyrosine phosphatases: regulatory mechanisms. FEBS J 275:831–847 Glondu-Lassis M, Dromard M, Lacroix-Triki M, Nirde P, Puech C, Knani D, Chalbos D, Freiss G, 2010, PTPL1/PTPN13 regulates breast cancer cell aggressiveness through direct inactivation of Src kinase. Cancer Res 70:5116–5126 Aoki N, Matsuda T, 2002, A nuclear protein tyrosine phosphatase TC-PTP is a potential negative regulator of the PRL-mediated signaling pathway: dephosphorylation and deactivation of signal transducer and activator of transcription 5a and 5b by TC-PTP in nucleus. Mol Endocrinol 16:58–69 Johnson KJ, Peck AR, Liu C, Tran TH, Utama FE, Sjolund AB, Schaber JD,Witkiewicz AK, Rui H, 2010, PTP1B suppresses prolactin activation of Stat5 in breast cancer cells. Am J Pathol Lu X, Malumbres R, Shields B, Jiang X, Sarosiek KA, Natkunam Y, Tiganis T, Lossos IS, 2008, PTP1B is a negative regulator of interleukin 4-induced STAT6 signaling. Blood 112:4098–4108 Tebbutt NC, Giraud AS, Inglese M, Jenkins B, Waring P, Clay FJ, Malki S, Alderman BM, Grail D, Hollande F, Heath JK, Ernst M, 2002, Reciprocal regulation of gastrointestinal homeostasis by SHP2 and STAT-mediated trefoil gene activation in gp130 mutant mice. Nat Med 8:1089–1097 Baron M, Davignon JL, 2008, Inhibition of IFN-gamma-induced STAT1 tyrosine phosphorylation by human CMV is mediated by SHP2. J Immunol 181:5530–5536 Tsai CC, Kai JI, Huang WC, Wang CY, Wang Y, Chen CL, Fang YT, Lin YS, Anderson R, Chen SH, Tsao CW, Lin CF, 2009, Glycogen synthase kinase-3beta facilitates IFN-gamma-induced STAT1 activation by regulating Src homology-2 domain-containing phosphatase 2. J Immunol 183:856–864 ten Hoeve J, de Jesus I-SM, Fu Y, Zhu W, Tremblay M, David M, Shuai K, 2002, Identification of a nuclear Stat1 protein tyrosine phosphatase. Mol Cell Biol 22:5662–5668 Meyer T, Hendry L, Begitt A, John S, Vinkemeier U, 2004, A single residue modulates tyrosine dephosphorylation, oligomerization, and nuclear accumulation of stat transcription factors. J Biol Chem 279:18998–19007 Huynh H, Bottini N,Williams S, Cherepanov V, Musumeci L, Saito K, Bruckner S,Vachon E,Wang X, Kruger J, ChowCW, Pellecchia M, Monosov E, Greer PA, Trimble W, Downey GP, Mustelin T, 2004, Nat Cell Biol 6:831–839 Xu MJ, Sui X, Zhao R, Dai C, Krantz SB, Zhao ZJ, 2003, Blood 102:4354–4360 Wang Y, Vachon E, Zhang J, Cherepanov V, Kruger J, Li J, Saito K, Shannon P, Bottini N, Huynh H, Ni H, Yang H, McKerlie C, Quaggin S, Zhao ZJ, Marsden PA, Mustelin T, Siminovitch KA, Downey GP, 2005, J Exp Med 202:1587–1597 Furth PA, Nakles RE, Millman S, Diaz-Cruz ES, Cabrera MC, 2011, Signal transducer and activator of transcription 5 as a key signaling pathway in normal mammary gland developmental biology and breast cancer. Breast Cancer Res 13:220 Yuan T, Wang Y, Zhao ZJ, Gu H, 2010, Protein-tyrosine phosphatase PTPN9 negatively regulates ErbB2 and epidermal growth factor receptor signaling in breast cancer cells. J Biol Chem 285: 14861–14870 Su F, Ren F et al, 2012, Protein tyrosine phosphatase Meg2 dephosphorylates signal transducer and activator of transcription 3 and suppresses tumor growth in breast cancer. Breast Cancer Res., DOI 10.1186/bcr3134 Xu X, Yamamoto H, Sakon M et al, 2003, Overexpression of CDC25A phosphatase is associated with hypergrowth activity and poor prognosis of human hepatocellular carcinomas. Clin Cancer Res 9:1764–1772 Yu C, Chen K, Zheng H et al, 2009, Overexpression of astrocyte elevated gene-1, AEG-1, is associated with esophageal squamous cell carcinoma, ESCC, progression and pathogenesis. Carcinogenesis 30: 894–901 Xue Q, Lv L, Wan C, Chen B, Li M, Ni T et al, 2013, Expression and clinical role of small glutamine-rich tetratricopeptide repeat, TPR)-containing protein alpha, SGTA, as a novel cell cycle protein in NSCLC. J Cancer Res Clin Oncol 139(9):1539–1549., DOI 10.1007/s00432-013-1474-5 Wan C, Hou S, Shen A et al, 2015, MIF4G domain containing protein regulates cell cycle and hepatic carcinogenesis by antagonizing CDK2-dependent p27 stability. Oncogene 34(2):237–245., DOI 10.1038/onc.2013.536 Sun T, Aceto N, Meerbrey KL, Kessler JD, Zhou C, Migliaccio I, Nguyen DX, Pavlova NN, Botero M, Huang J, Bernardi RJ, Schmitt E, Hu G, Li MZ, Dephoure N, Gygi SP, Rao M, Creighton CJ, Hilsenbeck SG, Shaw CA, Muzny D, Gibbs RA, Wheeler DA, Osborne CK, Schiff R, Bentires-Alj M, Elledge SJ, Westbrook TF, 2011, Activation of multiple proto-oncogenic tyrosinekinases in breast cancer via loss of the PTPN12 phosphatase. Cell 144:703–718 Julien SG, Dube N, Hardy S, Tremblay ML, 2010, Inside the human cancer tyrosine phosphatome. Nat Rev Cancer 11:35–49 Hu TH, Huang CC, Lin PR et al, 2003, Expression and prognostic role of tumor suppressor gene PTEN/MMAC1/TEP1 in hepatocellular carcinoma. Cancer 97:1929–1940 Sanchez A, Nagy P, Thorgeirsson SS, 2003, STAT-3 activity in chemically-induced hepatocellular carcinoma. Eur J Cancer 39: 2093–2098 Fuke H, Shiraki K, Sugimoto K, Tanaka J, Beppu T, Yoneda K, Yamamoto N, Ito K, Masuya M, Takei Y, 2007, Jak inhibitor induces S phase cell-cycle arrest and augments TRAIL-induced apoptosis in human hepatocellular carcinoma cells. Biochem Biophys Res Commun 363:738–744 Leslie K, Lang C, Devgan G, Azare J, Berishaj M, Gerald W, Kim YB, Paz K, Darnell JE, Albanese C, Sakamaki T, Pestell R, Bromberg J, 2006, Cyclin D1 is transcriptionally regulated by and required for transformation by activated signal transducer and activator of transcription 3. Cancer Res 66:2544–2552 Bowman T, Broome MA, Sinibaldi D, Wharton W, Pledger WJ, Sedivy JM, Irby R, Yeatman T, Courtneidge SA, Jove R, 2001, Stat3-mediated Myc expression is required for Src transformation and PDGF-induced mitogenesis. Proc Natl Acad Sci U S A 98: 7319–7324 Yue P, Turkson J, 2009, Targeting STAT3 in cancer: how successful are we? Expert Opin Investig Drugs 18:45–56 Du WW, Fang L, Yang BB et al, 2013, MicroRNA miR-24 enhances tumor invasion and metastasis by targeting PTPN9 and PTPRF to promote EGF signaling. J Cell Sci 126(6):1440–1453., DOI 10.1242/jcs.118299 HuangX, Gschweng E,WitteON et al, 2011, Regulated expression of microRNAs-126/126* inhibits erythropoiesis from human embryonic stem cells. Blood 117(7):2157–2165., DOI 10.1182/blood- 2010-08-302711 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 555 EP 565 DI 10.1007/s12253-015-0038-1 PG 11 ER PT J AU Jelicic, J Balint, TM Jovanovic, PM Boricic, N Micev, M Stojsic, J Antic, D Andjelic, B Bila, J Balint, B Pavlovic, S Mihaljevic, B AF Jelicic, Jelena Balint, Todorovic Milena Jovanovic, Perunicic Maja Boricic, Novica Micev, Marjan Stojsic, Jelena Antic, Darko Andjelic, Bosko Bila, Jelena Balint, Bela Pavlovic, Sonja Mihaljevic, Biljana TI The Role of Lymphocyte to Monocyte Ratio, Microvessel Density and High CD44 Tumor Cell Expression in Non Hodgkin Lymphomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lymphoma; Lymphocyte-to-monocyte count; Adhesion; Angiogenesis ID Lymphoma; Lymphocyte-to-monocyte count; Adhesion; Angiogenesis AB Prognostic significance of immune microenvironment has been emphasized using the most advanced analysis, with consecutive attempts to reveal prognostic impact of this findings. The aim of this study was to compare and define prognostic significance of clinical parameters, microvessel density (MVD) in tumour tissue and expression of CD44s as adhesive molecule on tumour cells in diffuse large B cell lymphoma-DLBCL, primary central nervous system DLBCL-CNS DLBCL and follicular lymphoma-FL. A total of 202 histopathological samples (115 DLBCL/65 FL/22 CNS DLBCL) were evaluated. Overall response (complete/ partial remission) was achieved in 81.3 % DLBCL patients, 81.8 % primary CNS DLBCL and 92.3 % FL. Absolute lymphocyte count-ALC/Absolute monocyte count-AMC >2.6 in DLBCL and ALC/AMC≥ 4.7 in FL were associated with better event-free survival (EFS) and overall survival (OS) (p < 0.05). In DLBCL, MVD> 42 blood vessels/0.36 mm2 correlated with primary resistant disease (p < 0.0001), poorer EFS and OS (p =0.014). High CD44s expression in FL correlated with inferior EFS and OS (p<0.01). In DLBCL, multivariate Cox regression analysis showed that ALC/AMC was independent parameter that affected OS (HR 3.27, 95 % CI 1.51–7.09, p = 0.003) along with the NCCN-IPI (HR 1.39, 95 % CI 1.08–1.79, p=0.01). Furthermore, in FL, ALC/ AMC mostly influenced OS (HR 5.21, 95 % CI 1.17–23.21, p =0.03), followed with the FLIPI (HR 3.98, 95 % CI 1.06– 14.95, p =0.041). In DLBCL and FL, ALC/AMC is simple and robust tool that is, with current prognostic scores, able to define long-term survival and identify patients with inferior outcome. The introduction of immunochemotherapy might altered the prognostic significance of microenvionmental biomarkers (MVD and CD44s). C1 [Jelicic, Jelena] Clinical Center of Serbia, Clinic for Hematology, Koste Todorovica 2 str, 11000 Belgrade, Serbia. [Balint, Todorovic Milena] Clinical Center of Serbia, Clinic for Hematology, Koste Todorovica 2 str, 11000 Belgrade, Serbia. [Jovanovic, Perunicic Maja] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia. [Boricic, Novica] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia. [Micev, Marjan] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia. [Stojsic, Jelena] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia. [Antic, Darko] Clinical Center of Serbia, Clinic for Hematology, Koste Todorovica 2 str, 11000 Belgrade, Serbia. [Andjelic, Bosko] Clinical Center of Serbia, Clinic for Hematology, Koste Todorovica 2 str, 11000 Belgrade, Serbia. [Bila, Jelena] Clinical Center of Serbia, Clinic for Hematology, Koste Todorovica 2 str, 11000 Belgrade, Serbia. [Balint, Bela] MMA, Institute for Transfusiology and HemobiologyBelgrade, Serbia. [Pavlovic, Sonja] University of Belgrade, Institute of Molecular Genetics and Genetic EngineeringBelgrade, Serbia. [Mihaljevic, Biljana] Clinical Center of Serbia, Clinic for Hematology, Koste Todorovica 2 str, 11000 Belgrade, Serbia. RP Balint, TM (reprint author), Clinical Center of Serbia, Clinic for Hematology, 11000 Belgrade, Serbia. EM bb.lena@gmail.com CR Anderson JR, Armitage JO, Weisenburger DD, 1998, Epidemiology of the non-Hodgkin’s lymphomas: distributions of the major subtypes differ by geographic locations. Ann Oncol 9: 717–720 Campo E, Swerdlow SH, Harris NL et al, 2011, The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications. Blood 117:5019–5032 Rosenwald A, Wright G, Chan WC et al, 2002, The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med 346:1937–1947 Dave SS, Wright G, Tan B et al, 2004, Prediction of survival in follicular lymphoma based on molecular features of tumorinfiltrating immune cells. N Engl J Med 351:2159–2169 Lenz G,Wright G, Dave SS et al, 2008, Stromal gene signatures in large-B-cell lymphomas. N Engl J Med 359:2313–2323 Bari A, Marcheselli L, Sacchi S et al, 2009, Prognostic models for diffuse large B-cell lymphoma in the rituximab era: a never-ending story. Ann Oncol 21:1486–1491 Cox MC, Nofroni I, Laverde G et al, 2008, Absolute lymphocyte count is a prognostic factor in diffuse large B-cell lymphoma. Br J Haematol 141:265–268 Kim DH, Baek JH, Chae YS et al, 2007, Absolute lymphocyte counts predicts response to chemotherapy and survival in diffuse large B-cell lymphoma. Leuk Off J Leuk Soc Am Leuk Res Fund UK 21:2227–2230 Oki Y, Yamamoto K, Kato H et al, 2008, Low absolute lymphocyte count is a poor prognostic marker in patients with diffuse large Bcell lymphoma and suggests patients’ survival benefit from rituximab. Eur J Haematol 81:448–453 Marcheselli L, Bari A, Anastasia A et al, 2015, Prognostic roles of absolutemonocyte and absolute lymphocyte counts in patients with advanced-stage follicular lymphoma in the rituximab era: an analysis from the FOLL05 trial of the Fondazione Italiana Linfomi. Br J Haematol 169:544–551 Kumagai S, TashimaM, Fujikawa J et al, 2014, Ratio of peripheral blood absolute lymphocyte count to absolute monocyte count at diagnosis is associated with progression-free survival in follicular lymphoma. Int J Hematol 99:737–742 Camilleri-Broet S, Criniere E, Broet P et al, 2006, A uniform activated B-cell-like immunophenotype might explain the poor prognosis of primary central nervous system lymphomas: analysis of 83 cases. Blood 107:190–196 Lin C-H, Kuo K-T, Chuang S-S et al, 2006, Comparison of the expression and prognostic significance of differentiation markers between diffuse large B-cell lymphoma of central nervous system origin and peripheral nodal origin. Clin Cancer Res 12:1152–1156 Cardesa-Salzmann TM, Colomo L, Gutierrez G et al, 2011, High microvessel density determines a poor outcome in patients with diffuse large B-cell lymphoma treated with rituximab plus chemotherapy. Haematologica 96:996–1001 HiddemannW, Kneba M, DreylingMet al, 2005, Frontline therapy with rituximab added to the combination of cyclophosphamide, doxorubicin, vincristine, and prednisone, CHOP, significantly improves the outcome for patients with advanced-stage follicular lymphoma compared with therapy with CHOP alone: r. Blood 106: 3725–3732 Coiffier B, Thieblemont C,Van Den Neste E et al, 2010, Long-term outcome of patients in the LNH-98.5 trial, the first randomized study comparing rituximab-CHOP to standard CHOP chemotherapy in DLBCL patients: a study by the Groupe d’Etudes des Lymphomes de l’Adulte. Blood 116:2040–2045 He M, Zuo C, Wang J et al, 2013, Prognostic significance of the aggregative perivascular growth pattern of tumor cells in primary central nervous system diffuse large B-cell lymphoma. Neuro Oncol 15:727–734 (1993, A predictive model for aggressive non-Hodgkin’s lymphoma. Int Non-Hodgkin’s Lymphoma Prognostic Factors Project. N Engl J Med 329:987–994 Sehn LH, Berry B, Chhanabhai M et al, 2007, The revised International Prognostic Index, R-IPI, is a better predictor of outcome than the standard IPI for patients with diffuse large B-cell lymphoma treated with R-CHOP. Blood 109:1857–1861 Solal-Celigny P, 2004, Follicular lymphoma international prognostic index. Blood 104:1258–1265 Ruan J, Hajjar K, Rafii S, Leonard JP, 2009, Angiogenesis and antiangiogenic therapy in non-Hodgkin’s lymphoma. Ann Oncol 20:413–424 Ponta H, Sherman L, Herrlich PA, 2003, CD44: from adhesion molecules to signalling regulators. Nat Rev Mol Cell Biol 4:33–45 Yuan J, Gu K, He J, Sharma S, 2013, Preferential up-regulation of osteopontin in primary central nervous system lymphoma does not correlate with putative receptor CD44v6 or CD44H expression. Hum Pathol 44:606–611 Braaten KM, Betensky RA, de Leval L et al, 2003, BCL-6 expression predicts improved survival in patients with primary central nervous system lymphoma. Clin Cancer Res 9:1063–1069 Wei X, Xu M, Wei Y et al, 2014, The addition of rituximab to CHOP therapy alters the prognostic significance of CD44 expression. J Hematol Oncol 7:34 HigashiM, Sugaya Y, Soeta S et al, 2009, CD44 expression during tumor progression of follicular lymphoma. Oncol Rep 22:1135– 1140 Cheson BD, Horning SJ, Coiffier B et al, 1999, Report of an international workshop to standardize response criteria for non- Hodgkin’s lymphomas. NCI Sponsored International Working Group. J Clin Oncol 17:1244 DeAngelis LM, SeiferheldW, Schold SC et al, 2002, Combination chemotherapy and radiotherapy for primary central nervous system lymphoma: Radiation Therapy Oncology Group Study 93–10. J Clin Oncol 20:4643–4648 Schraml P, Kononen J, Bubendorf L et al, 1999, Tissue microarrays for gene amplification surveys in many different tumor types. Clin Cancer Res 5:1966–1975 Visco C, Li Y, Xu-Monette ZY et al, 2012, Comprehensive gene expression profiling and immunohistochemical studies support application of immunophenotypic algorithm for molecular subtype classification in diffuse large B-cell lymphoma: a report from the International DLBCL Rituximab-CHOP Consortiu. Leukemia 26: 2103–2113 Tzankov A, Pehrs A-C, Zimpfer A et al, 2003, Prognostic significance of CD44 expression in diffuse large B cell lymphoma of activated and germinal centre B cell-like types: a tissue microarray analysis of 90 cases. J Clin Pathol 56:747–752 Li Z-M, Huang J-J, Xia Y et al, 2012, Blood lymphocyte-tomonocyte ratio identifies high-risk patients in diffuse large B-cell lymphoma treated with R-CHOP. PLoS One 7:e41658 Belotti A, Doni E, Bolis S et al, 2015, Peripheral blood lymphocyte/monocyte ratio predicts outcome in follicular lymphoma and in diffuse large B-cell lymphoma patients in the rituximab era. Clin Lymphoma Myeloma Leuk 15:208–213 Barrans SL, Fenton JAL, Banham A et al, 2004, Strong expression of FOXP1 identifies a distinct subset of diffuse large B-cell lymphoma, DLBCL, patients with poor outcome. Blood 104:2933– 2935 Rubenstein JL, Shen A, Batchelor TT et al, 2009, Differential gene expression in central nervous system lymphoma. Blood 113:266– 267 Lin B, Chen C, Qian Y, Feng J, 2015, Prognostic role of peripheral blood lymphocyte/monocyte ratio at diagnosis in diffuse large Bcell lymphoma: a meta-analysis. Leuk Lymphoma 1–6 Behl D, Ristow K, Markovic SN et al, 2007, Absolute lymphocyte count predicts therapeutic efficacy of rituximab therapy in follicular lymphomas. Br J Haematol 137:409–415 Siddiqui M, Ristow K,Markovic SN et al, 2006, Absolute lymphocyte count predicts overall survival in follicular lymphomas. Br J Haematol 134:596–601 Watanabe R, Tomita N, Kishimoto K et al, 2013, Absolute monocyte count in follicular lymphoma patients treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. Leuk Res 37:1208–1212 Wilcox RA, Ristow K, Habermann TM et al, 2012, The absolute monocyte count is associated with overall survival in patients newly diagnosed with follicular lymphoma. Leuk Lymphoma 53:575–580 Folkman J, 1995)Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med 1:27–31 Ribatti D, 2014, History of research on angiogenesis. Chem Immunol Allergy 99:1–14 Norrby K, Ridell B, 2003, Tumour-type-specific capillary endothelial cell stainability inmalignant B-cell lymphomas using antibodies against CD31, CD34 and factor VIII. APMIS 111:483–489 Jorgensen JM, Sorensen FB, Bendix K et al, 2007, Angiogenesis in non-Hodgkin’s lymphoma: clinico-pathological correlations and prognostic significance in specific subtypes. Leuk Lymphoma 48: 584–595 Mazur G, Wrobel T, Dziegiel P et al, 2004, Angiogenesis measured by expression of CD34 antigen in lymph nodes of patients with non-Hodgkin’s lymphoma. Folia Histochem Cytobiol 42:241–243 Bairey O, Zimra Y, Kaganovsky E et al, 2000)Microvessel density in chemosensitive and chemoresistant diffuse large B-cell lymphomas. Med Oncol 17:314–318 Ribatti D,Nico B, Ranieri G et al, 2013, The role of angiogenesis in human non-Hodgkin lymphomas. Neoplasia 15:231–238 Nagel S, Hirschmann P, Dirnhofer S et al, 2010, Coexpression of CD44 variant isoforms and receptor for hyaluronic acid-mediated motility, RHAMM, CD168, is an International Prognostic Index and C-MYC gene status-independent predictor of poor outcome in diffuse large B-cell lymphomas. Exp Hematol 38:38–45 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 567 EP 577 DI 10.1007/s12253-015-0032-7 PG 11 ER PT J AU Szanto, A Szabo, K Nagy, G Molnar, Cs Zeher, M AF Szanto, Antonia Szabo, Katalin Nagy, Gabor Molnar, Csaba Zeher, Margit TI Characterization and Comparison of Patient Subgroups Suspicious for IgG4-Related Disease and Malignant Lymphoma in Patients Followed-up for Sjogren’s Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE IgG4-related disease; Malignant lymphoma; Sjogren’s syndrome ID IgG4-related disease; Malignant lymphoma; Sjogren’s syndrome AB Differential diagnosis of patients with Sjogren’s syndrome (SS), IgG4-related disease (IgG4-RD) and SS patients having high risk for lymphoma (LHR) can be challenging. Some patients with IgG4-RD might be misdiagnosed as having SS. There are special symptoms of SS that raise the possibility of IgG4-RD whereas other symptoms identify patients as having LHR. The purpose of this study was to characterize and compare patients with SS, possible IgG4-RD and SS patients with LHR. Sixty-five SS patients were divided into 4 subgroups according to having possible IgG4-RD (n = 15), LHR (n = 16), eligible for both aforementioned groups (n = 20) and not eligible for either group (n = 14), respectively. Four patients fulfilled the diagnostic criteria for IgG4-RD. The serum levels of IgG4 were significantly higher in patients suspicious for IgG4-RD compared to that of LHR patients (0.46 g/l vs. 0.12 g/l, p = 0.032). Shared features of the patient groups (salivary gland swelling (SGS) and lymphadenopathy), were separately analysed: SGS patients had higher IgG4/IgG ratio (p = 0.036), lymphadenopathic patients had higher IgG4 levels (p = 0.042). Some patients may be "hidden" under the diagnosis of SS. Although patients with LHR and patients with possible IgG4-RD share some symptoms, they differ significantly regarding IgG4 levels and IgG4/IgG ratio. C1 [Szanto, Antonia] University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, 22 Moricz Zs. Krt, H-4032 Debrecen, Hungary. [Szabo, Katalin] Josa Andras County Hospital, Department of Pediatrics, 68 Szent Istvan Street, H-4400 Nyiregyhaza, Hungary. [Nagy, Gabor] University of Debrecen, Faculty of Medicine, Department of Laboratory Medicine, 22 Moricz Zs. Krt, H-4032 Debrecen, Hungary. [Molnar, Csaba] University of Debrecen, Faculty of Medicine, Department of Pathology, 98 Nagyerdei Krt, H-4032 Debrecen, Hungary. [Zeher, Margit] University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, 22 Moricz Zs. Krt, H-4032 Debrecen, Hungary. RP Zeher, M (reprint author), University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, H-4032 Debrecen, Hungary. EM zeher@iiibel.dote.hu CR Masaki Y, Kurose N, Umehara H, 2011, IgG4-related disease: a novel lymphoproliferative disorder discovered and established in Japan in the 21st century. J Clin Exp Hematopathol 1:13–20 Okazaki K, Uchida K, Research Committee Members, 2009, Proposal of the Concept and diagnostic criteria of IgG4-related disease, in Japanese). Annual reports of research committee of intractable diseases supported by Ministry of Health, Labour and Welfare of Japan 25–30 Umehara H, Okazaki K, Masaki Y, Kawano M, Yamamoto M, Saeki T, Matsui S, Yoshino T, Nakamura S, Kawa S, Hamano H, Kamisawa T, Shimosegawa T, Shimatsu A, Nakamura S, Ito T, Notohara K, Sumida T, Tanaka Y, Mimori T, Chiba T, Mishima M, Hibi T, Tsubouchi H, Inui K, Ohara H, 2012, Comprehensive diagnostic criteria for IgG4-related disease, IgG4-RD). Mod Rheumatol 22:21–30 Vitali C, Bombardieri S, Jonsson R, HMM, EL A, SE C, TE D, PC F, RI F, SS K, SR P, Talal N, MH W, European Study Group on Classification Criteria for Sjogren's Syndrome, 2002, Classification criteria for sjogren’s syndrome: a revised version of the European criteria proposed by the American-European consensus group. Ann Rheum Dis 61:554–558 Geyer JT, Desphande V, 2011, IgG4-associated sialadenitis. Curr Opin Rheumatol 23:95–101 Ioannidis JP, Vassioliou VA, Moutsopoulos HM, 2002, Long-term risk of mortality and lymphoproliferative disease and predictive classification of primary sjogren’s syndrome. Arthritis Rheum 46:741–747 Seror R, Ravaud P, Bowman S, Baron G, Tzioufas A, Theander E, Gottenberg JE, Bootsma H, Mariette X, Vitali C, Sjogren's Task Force EULAR, 2010, EULAR sjogren’s syndrome disease activity index: development of a consensus systemic disease activity index for primary sjogren’s syndrome. Ann Rheum Dis 69:1103–1109 Szanto A, Nagy G, Cs M, Griger Z, Tarr T, Zeher M, 2014, Description of patients with IgG4-related disease from a Hungarian center. Scand J Rheumatol 43:334–337 Ryu JH, Horie R, Sekiguchi H, Peikert T, Yi ES, 2012, Spectrum of disorders associated with elevated serum IgG4 levels encountered in clinical practice. Int J Rheumatol 232960., DOI 10.1155/2012/ 232960 Moriyama M, Tanaka A, Maehara T, Ohyama Y, Shimizu M, Nakashima H, Hayashida JN, Shinozaki S, Kubo Y, Furukawa S, Kikuta T, Nakamura S, 2013, Clinical characteristics of mikulicz’s disease as an IgG4-related disease. Clin Oral Investig 17:1995–2002 Okazaki K, Uchida K, Miyoshi H, Ikeura T, Takaoka M, Nishio A, 2011, Recent concepts of autoimmune pancreatitis and IgG4- related disease. Clin Rev Allergy Immunol 41:126–138 Shimizu I, Nasu K, Sato K, Ueki H, Akahane D, SumiM, Ueno M, Ichikawa N, Asano N, Kojima M, Kobayashi H, 2010, Lymphadenopathy of IgG4-related sclerosing disease: three case reports and review of literature. Int J Hematol 92:751–756 Quartuccio L, Isola M, Baldini C, Priori R, Bocci EB, Carubbi F, Maset M, Gregoraci G, Della Mea V, Salvin S, De Marchi G, Luciano N, Colafrancesco S, Alunno A, Giacomelli R, Gerli R, Valesini G, Bombardieri S, De Vita S, 2014, Biomarkers of lymphoma in sjogren’s syndrome and evaluation of the lymphoma risk in prelymphomatous conditions: results of a multicenter study. J Autoimmun 51:75–80 Stone JH, ZenY, Desphande V, 2012, IgG4-related disease. N Engl J Med 366:539–551 Laco J, Ryska A, Celakovsky P, Dolezalova H, Mottl R, Tucek L, 2011, Chronic sclerosing sialadenitis as one of the immunoglobulin G4-related diseases: a clinicopathological study of six cases from Central Europe. Histopathology 58:1157–1163 Stone JH, Khosroshahi A, Desphande V, Chan JKC, Heathcote JG, Aalberse R, Azumi A, Bloch DB, Brugge WR, Carruthers MN, Cheuk W, Cornell L, Castillo CF, Ferry JA, Forcione D, Kloppel G, Hamilos DL, Kamisawa T, Kasashima S, Kawa S, Kawano M, MasakiY, Notohara K, Okazaki K, Ryu JK, Saeki T, Sahani D, Sato Y, Smyrk T, Stone JR, Takahira M, Umehara H, Webster G, Yamamoto M, Yi E, Yoshino T, Zamboni G, Zen Y, Chari S, 2012, Recommendations for the nomenclature of IgG4-related disease and its individual organ system manifestations. Arthritis Rheum 64:3061–3067 Moriyama M, Furukawa S, Kawano S, Goto Y, Kiyoshima T, Tanaka A, Maehara T, Hayashida J-N, Ohta M, Nakamura S, 2014, The diagnostic utility of biopsies from the submandibular and labial salivary glands in IgG4-related dacryoadenitis and sialoadenitis, so-called mikulicz’s disease. Int J Oral Maxillofac Surg 43:1276–1281 Shimizu Y, Yamamoto M, Naishiro Y, Sudoh G, Ishigami K, Yajima H, Tabeya T, Matsui M, Suzuki C, Takahashi H, Seki N, Himi T, YamashitaK, Noguchi H,Hasegawa T, Suzuki Y, Honda S, Abe T, Imai K, Shinomura Y, 2013, Necessity of early intervention for IgG4-related disease - delayed treatment induces fibrosis progression. Rheumatology, Oxford, 52:679–683 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 579 EP 585 DI 10.1007/s12253-016-0041-1 PG 7 ER PT J AU Jo, SY Kim, SM Yoo, JN Lee, HS AF Jo, Sol Yun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutations of AKAP9 Gene in Gastric and Colorectal Cancers with High Microsatellite Instability SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE AKAP9; Fameshift mutation; Cancer; Microsatellite instability ID AKAP9; Fameshift mutation; Cancer; Microsatellite instability AB A-kinase-anchoring protein 9 (AKAP9) coordinates the cellular location and function of protein kinase A. AKAP9 plays an important role in centrosome duplication, cell cycle progression and maintenance of cell membrane integrity, alterations of which contribute to tumorigenesis. Somatic mutations of AKAP9 gene have been detected in many cancers including gastric (GC) and colorectal cancers (CRC), but the mutation status with respect to microsatellite instability (MSI) has not been reported. In a public database, we found that AKAP9 gene had mononucleotide repeats in the coding sequences that might be mutation targets in the cancers with MSI. We analyzed the mutations in 79 GCs and 124 CRCs including high MSI (MSI-H) and microsatellite stable/low MSI (MSS/MSI-L) cases by single-strand conformation polymorphism analysis and DNA sequencing. Overall, we found AKAP9 frameshift mutations in 4 (11.7 %) GCs and 20 (17.7 %) CRCs with MSI-H (24/113), but not in MSS/MSI-L cancers (0/90) (p < 0.001). In addition, we analyzed intratumoral heterogeneity (ITH) of AKAP9 frameshift mutations in 16 CRCs and found that five CRCs (31.3 %) harbored regional ITH of the AKAP9 frameshift mutations. Our data indicate that AKAP9 gene harbors not only somatic frameshift mutations but also mutational ITH, which together may be features of GC and CRC with MSI-H. Our results also suggest that regional mutation analysis is needed for a better evaluation of mutation status in these tumors to overcome ITH. C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Colledge M, Scotte JD, 1999, AKAPs: from structure to function. Trends Cell Biol 9:216–221 Rubin CS, 1994, A kinase anchor proteins and the intracellular targeting of signals carried by cyclic AMP. Biochim Biophys Acta 1224:467–479 Schmidt PH, Dransfield DT, Claudio JO, Hawley RG, Trotter KW, Milgram SL, Goldenring JR, 1999, AKAP350, a multiply spliced protein kinase a-anchoring protein associated with centrosomes. J Biol Chem 274(5):3055–3066 Terrin A, Monterisi S, Stangherlin A, Zoccarato A, Koschinski A, Surdo NC, Mongillo M, Sawa A, Jordanides NE, Mountford JC, Zaccolo M, 2012, PKA and PDE4D3 anchoring to AKAP9 provides distinct regulation of cAMP signals at the centrosome. J Cell Biol 198:607–621 Mattaloni SM, Ferretti AC, Tonucci FM, Favre C, Goldenring JR, Larocca MC, 2013, Centrosomal AKAP350 modulates the G1/S transition. Cell Logist 3:e26331 Diviani D, Scott JD, 2001, AKAP signaling complexes at the cytoskeleton. J Cell Sci 114:1431–1437 Scotte JD, 2003, A-kinase-anchoring proteins and cytoskeletal signalling events. Biochem Soc Trans 31(Pt 1):87–89 Gros R, Ding Q, Chorazyczewski J, Pickering JG, Limbird LE, Feldman RD, 2006, Adenylyl cyclase isoform-selective regulation of vascular smooth muscle proliferation and cytoskeletal reorganization. Circ Res 99:845–852 Oldenburger A, Poppinga WJ, Kos F, de Bruin HG, Rijks WF, Heijink IH, Timens W, Meurs H, Maarsingh H, Schmidt M, 2014, A-kinase anchoring proteins contribute to loss of Ecadherin and bronchial epithelial barrier by cigarette smoke. Am J Physiol Cell Physiol 306:C585–C597 Chen L, Marquardt ML, Tester DJ, Sampson KJ, Ackerman MJ, Kass RS, 2007, Mutation of an a-kinase-anchoring protein causes long-QT syndrome. Proc Natl Acad Sci U S A 104:20990–20995 Ciampi R, Knauf JA, Kerler R, Gandhi M, Zhu Z, Nikiforova MN, Rabes HM, Fagin JA, Nikiforov YE, 2005, Oncogenic AKAP9- BRAF fusion is a novel mechanism of MAPK pathway activation in thyroid cancer. J Clin Invest 115:94–101 Onken MD, Winkler AE, Kanchi KL, Chalivendra V, Law JH, Rickert CG, Kallogjeri D, Judd NP, Dunn GP, Piccirillo JF, Lewis JS Jr, Mardis ER, Uppaluri R, 2014, A surprising cross-species conservation in the genomic landscape of mouse and human oral cancer identifies a transcriptional signature predicting metastatic disease. Clin Cancer Res 20:2873–2884 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8: 305–311 Oh HR, An CH, Yoo NJ, Lee SH, 2015, Frameshift mutations of MUC15 gene in gastric and its regional heterogeneity in gastric and colorectal cancers. Pathol Oncol Res 21: 713–718 Choi MR, An CH, Yoo NJ, Lee SH, 2015, Laminin gene LAMB4 is somatically mutated and expressionally altered in gastric and colorectal cancers. APMIS 123:65–71 Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, Xie M, Zhang Q, McMichael JF, Wyczalkowski MA, Leiserson MD, Miller CA, Welch JS, Walter MJ, Wendl MC, Ley TJ, Wilson RK, Raphael BJ, Ding L, 2013, Mutational landscape and significance across 12 major cancer types. Nature 502:333–339 Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, Meyerson M, Gabriel SB, Lander ES, Getz G, 2014, Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505:495–501 Gonzalez-Perez A, Perez-Llamas C, Deu-Pons J, Tamborero D, Schroeder MP, Jene-Sanz A, Santos A, Lopez-Bigas N, 2013, IntOGen-mutations identifies cancer drivers across tumor types. Nat Methods 10:1081–1082 Hayashi K, 1992, PCR-SSCP: a simple and sensitive method for detection of mutations in the genomic DNA. PCR Methods Appl 1: 34–38 Calin GA, Gafa R, Tibiletti MG, Herlea V, Becheanu G, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression inmicrosatellite instability high, MSI-H, colon cancers correlates with clinico-pathological parameters: a study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89:230–235 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 587 EP 592 DI 10.1007/s12253-016-0042-0 PG 6 ER PT J AU Ozdemir, AD Usubutun, A AF Ozdemir, Ates Deniz Usubutun, Alp TI PAX2, PAX8 and CDX2 Expression in Metastatic Mucinous, Primary Ovarian Mucinous and Seromucinous Tumors and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PAX2; PAX8; Mucinous ovarian tumor; Mucinous ovarian carcinoma; Borderline seromucinous tumor ID PAX2; PAX8; Mucinous ovarian tumor; Mucinous ovarian carcinoma; Borderline seromucinous tumor AB Ovarian cancer is the most common cause of gynecologic cancer death. Both morphologically and immunohistochemically, metastatic mucinous tumors are the best mimickers of mucinous ovarian tumors; its pathogenesis s t i l l r e m a i n s a m y s t e r y. PAX2 a n d PAX8 immunohisyochemistries are useful for differentiating numerous primary tumour types from metastatic ones. There are few studies in literature about PAX expressions in mucinous and seromucinous tumors. None of these are takes into account the histologic type (whether it is seromucinous or mucinous) or the metastatic origin. With this purpose hematoxylin and eosine slides of ovarian mucinous and seromucinous tumors were reevaluated and one block was chosen for each case. The study included 76 ovarian mucinous and seromucinous tumors of the ovary reported in Hacettepe University department of pathology between 2000 and 2013. Tissue microarray (TMA) was designed from the chosen blocks, PAX2, PAX8, CDX2 immunostains was preformed to the TMA slides. As a result, most of the metastatic cases were negative for PAX2 (91.2 %) and PAX8 (86.3 %), many were diffusely and strongly positive for CDX2 (68.2 %). Seromucinous tumors were devoid of CDX2 expression; but all cases (except one) displayed strong and diffuse positivity with PAX8. In other words differing from mucinous tumors, seromucinous tumors show strong PAX8 positivity–similar to serous tumors. This study shows that PAX8 and CDX2 could be useful in differentiating primary mucinous from metastatic tumor. Furthermore unlike the homogeneity in seromucinous tumors for PAX8 and CDX2 mucinous tumors shows heterogeneity with different expression patterns. C1 [Ozdemir, Ates Deniz] Hacettepe University, Faculty of Medicine, Department of PathologyAnkara, Turkey. [Usubutun, Alp] Hacettepe University, Faculty of Medicine, Department of PathologyAnkara, Turkey. RP Ozdemir, AD (reprint author), Hacettepe University, Faculty of Medicine, Department of Pathology, Ankara, Turkey. EM denizates010@gmail.com CR Seidman JD, Kurman RJ, Ronnett BM, 2003, Primary and metastatic mucinous adenocarcinomas in the ovaries: incidence in routine practice with a new approach to improve intraoperative diagnosis. Am J Surg Pathol 27(7):985–993 Ulbright TM, Roth LM, Stehman FB, 1984, Secondary ovarian neoplasia. A clinicopathologic study of 35 cases. Cancer 53(5):1164–1174 Lagendijk JH, Mullink H, Van Diest PJ, Meijer GA, Meijer CJ, 1998, Tracing the origin of adenocarcinomaswith unknown primary using immunohistochemistry: differential diagnosis between colonic and ovarian carcinomas as primary sites. Hum Pathol 29(5):491–497 Chu PG, Chung L,Weiss LM, Lau SK, 2011, Determining the site of origin of mucinous adenocarcinoma: an immunohistochemical study of 175 cases. Am J Surg Pathol 35(12):1830–1836 Zhai QJ, Ozcan A, Hamilton C, Shen SS, Coffey D, Krishnan B, Truong LD, 2010, PAX-2 expression in non-neoplastic, primary neoplastic, and metastatic neoplastic tissue: a comprehensive immunohistochemical study. Appl Immunohistochem Mol Morphol 18(4):323–332 Ozcan A, Liles N, Coffey D, Shen SS, Truong LD, 2011, PAX2 and PAX8 expression in primary and metastatic mullerian epithelial tumors: a comprehensive comparison. Am J Surg Pathol 35(12):1837–1847 Ozcan A, Shen SS, Hamilton C, Anjana K, Coffey D, Krishnan B, Truong LD, 2011, PAX 8 expression in non-neoplastic tissues, primary tumors, and metastatic tumors: a comprehensive immunohistochemical study. Mod Pathol 24(6):751–764 Jones JM, Cui XS, Medina D, Donehower LA, 1999, Heterozygosity of p21WAF1/CIP1 enhances tumor cell proliferation and cyclin D1-associated kinase activity in a murine mammary cancer model. Cell Growth Differ 10(4):213–222 Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, Senz J, McConechy MK, Anglesio MS, Kalloger SE, Yang W, Heravi- Moussavi A, Giuliany R, Chow C, Fee J, Zayed A, Prentice L, Melnyk N, Turashvili G, Delaney AD, Madore J, Yip S, McPherson AW, Ha G, Bell L, Fereday S, Tam A, Galletta L, Tonin PN, Provencher D, Miller D, Jones SJ, Moore RA, Morin GB, Oloumi A, Boyd N, Aparicio SA, Shih Ie M, Mes-Masson AM, Bowtell DD, Hirst M, Gilks B, Marra MA, Huntsman DG, 2010, ARID1Amutations in endometriosis-associated ovarian carcinomas. New Engl J Med 363(16):1532–1543 Wu CH,Mao TL, Vang R, Ayhan A,Wang TL, Kurman RJ, Shih Ie M, 2012, Endocervical-type mucinous borderline tumors are related to endometrioid tumors based onmutation and loss of expression of ARID1A. Int J Gynecol Pathol 31(4):297–303 Okada S, Ohaki Y, Ogura J, Ishihara M, Kawamura T, Kumazaki T, 2004, Computed tomography and magnetic resonance imaging findings in cases of dermoid cyst coexisting with surface epithelial tumors in the same ovary. J Comput Assist Tomogr 28(2):169–173 Ueda G, Sawada M, Ogawa H, Tanizawa O, Tsujimoto M, 1993, Immunohistochemical study of cytokeratin 7 for the differential diagnosis of adenocarcinomas in the ovary. Gynecol Oncol 51(2): 219–223., DOI 10.1006/gyno.1993.1276 Kerr SE, Flotte AB, McFalls MJ, Vrana JA, Halling KC, Bell DA, 2013, Matching maternal isodisomy in mucinous carcinomas and associated ovarian teratomas provides evidence of germ cell derivation for some mucinous ovarian tumors. Am J Surg Pathol 37(8): 1229–1235 Seidman JD, Khedmati F, 2008, Exploring the histogenesis of ovarian mucinous and transitional cell, brenner, neoplasms and their relationship with walthard cell nests: a study of 120 tumors. Arch Pathol Lab Med 132(11):1753–1760 Cuatrecasas M, Villanueva A, Matias-Guiu X, Prat J, 1997, K-ras mutations in mucinous ovarian tumors: a clinicopathologic and molecular study of 95 cases. Cancer 79(8):1581–1586 Heinzelmann-Schwarz VA, Gardiner-Garden M, Henshall SM, Scurry JP, Scolyer RA, Smith AN, Bali A, Vanden Bergh P, Baron-Hay S, Scott C, Fink D, Hacker NF, Sutherland RL, O'Brien PM, 2006, A distinct molecular profile associated with mucinous epithelial ovarian cancer. Brit J Cancer 94(6):904–913 Ronnett BM, Kajdacsy-Balla A, Gilks CB, Merino MJ, Silva E, Werness BA, Young RH, 2004, Mucinous borderline ovarian tumors: points of general agreement and persistent controversies regarding nomenclature, diagnostic criteria, and behavior. Hum Pathol 35(8):949–960 Kurman RJ, Shih Ie M, 2010, The origin and pathogenesis of epithelial ovarian cancer: a proposed unifying theory. Am J Surg Pathol 34(3):433–443., DOI 10.1097/PAS.0b013e3181cf3d79 McMahon CP, 2000, Ephraim McDowell, jane todd crawford, and the origins of oophorectomy. N C Med J 61(1):401–402 Pinto PB, Derchain SF, Andrade LA, 2012, Metastatic mucinous carcinomas in the ovary: a practical approach to diagnosis related to gross aspects and to immunohistochemical evaluation. Int J Gynecol Pathol 31(4):313–318., DOI 10.1097/PGP.0b013e31823f844d Laury AR, Perets R, Piao H, Krane JF, Barletta JA, French C, Chirieac LR, Lis R, Loda M, Hornick JL, Drapkin R, Hirsch MS, 2011, A comprehensive analysis of PAX8 expression in human epithelial tumors. Am J Surg Pathol 35(6):816–826., DOI 10.1097/ PAS.0b013e318216c112 Pickel H, Tamussino K, 2003, History of gynecological pathology: XIV. Hermann johannes pfannenstiel. Int J Gynecol Pathol 22(3): 310–314., DOI 10.1097/01.PGP.0000070844.85581.A1 Kobel M, Kalloger SE, Boyd N, McKinney S, Mehl E, Palmer C, Leung S, Bowen NJ, Ionescu DN, Rajput A, Prentice LM, Miller D, Santos J, Swenerton K, Gilks CB, Huntsman D, 2008, Ovarian carcinoma subtypes are different diseases: implications for biomarker studies. PLoS Med 5(12):e232., DOI 10.1371/journal.pmed.0050232 Nonaka D, Chiriboga L, Soslow RA, 2008, Expression of pax8 as a useful marker in distinguishing ovarian carcinomas from mammary carcinomas. Am J Surg Pathol 32(10):1566–1571 Tabrizi AD, Kalloger SE, Kobel M, Cipollone J, Roskelley CD, Mehl E, Gilks CB, 2010, Primary ovarian mucinous carcinoma of intestinal type: significance of pattern of invasion and immunohistochemical expression profile in a series of 31 cases. Int J Gynecol Pathol 29(2):99–107 Maeda D, Shih Ie M, 2013, Pathogenesis and the role of ARID1A mutation in endometriosis-related ovarian neoplasms. Adv Anat Pathol 20(1):45–52 Blaustein A, Kurman RJ, 2011, Blaustein's pathology of the female genital tract, 6th edn. Springer, New York, NYp 736–739 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 593 EP 599 DI 10.1007/s12253-016-0040-2 PG 7 ER PT J AU Sulzyc-Bielicka, V Domagala, P Bielicki, D Safranow, K Rogowski, W Domagala, W AF Sulzyc-Bielicka, Violetta Domagala, Pawel Bielicki, Dariusz Safranow, Krzysztof Rogowski, Wojciech Domagala, Wenancjusz TI E2F1/TS Immunophenotype and Survival of Patients with Colorectal Cancer Treated with 5FU-Based Adjuvant Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; E2F1; Thymidylate synthase; 5FU-based therapy ID Colorectal cancer; E2F1; Thymidylate synthase; 5FU-based therapy AB The predictive value of thymidylate synthase (TS) expression alone for 5FU-based treatment of colorectal cancer (CRC) has not been clinically confirmed. Little is known on the association of expression of E2F1, which controls the transcription of genes encoding proteins engaged in DNA synthesis including TS, and survival of patients with CRC. The purpose of this study is to assess the correlation between expression of both E2F1 and TS in CRCs and survival of patients administered adjuvant 5FU-based chemotherapy, in order to find a better predictor of treatment outcome than expression of TS or E2F1 alone. Nuclear TS and E2F1 were detected by immunohistochemistry in tissue microarrays from 190 CRCs (Astler-Coller stage B2 or C). Multivariate analysis identified significant association of the combined E2F1+TS+ immunophenotype with worse OS (HR= 3,78, P=0,009) and DFS (HR= 2,30, P = 0,03) of patients with colon cancer. There were significant differences between E2F1+TS+ and E2F1-TS- Kaplan-Meier survival curves in relation to DFS (P=0.008) and OS (P=0.01). About 37 and 31 % difference in 3-year DFS and OS respectivelywere seen between patients w i t h E2F1+TS+ v s . E2F1-TS- c o l o n c a n c e r immunophenotype. The E2F1+TS+ immunophenotype may be a marker of poor prognosis (the worst DFS and OS) of patients with colon cancer treated with 5FU-based adjuvant therapy. A subgroup of patients with this immunophenotype may require different and perhaps more aggressive treatment than 5FU-based chemotherapy. Thus, the combined E2F1/TS immunophenotype could be a potential indicator of colon cancer sensitivity to 5FU. C1 [Sulzyc-Bielicka, Violetta] Pomeranian Medical University, Department of Clinical OncologySzczecin, Poland. [Domagala, Pawel] Pomeranian Medical University, Department of Pathology, Unii Lubelskiej 1, 71-252 Szczecin, Poland. [Bielicki, Dariusz] Pomeranian Medical University, Department of GastroenterologySzczecin, Poland. [Safranow, Krzysztof] Pomeranian Medical University, Department of Biochemistry and Medical ChemistrySzczecin, Poland. [Rogowski, Wojciech] University of Warmia and Mazury, Department of OncologyOlsztyn, Poland. [Domagala, Wenancjusz] Pomeranian Medical University, Department of Pathology, Unii Lubelskiej 1, 71-252 Szczecin, Poland. RP Domagala, W (reprint author), Pomeranian Medical University, Department of Pathology, 71-252 Szczecin, Poland. EM wenek@pum.edu.pl CR Wolpin BM, Meyerhardt JA, Mamon HJ, Mayer RJ, 2007, Adjuvant treatment of colorectal cancer. CA Cancer J Clin 57: 168–185 Moertel CG, Fleming TR, Macdonald JS, Haller DG, Laurie JA, Goodman PJ, Ungerleider JS, EmersonWA, Tormey DC, Glick JH, 1990, Levamisole and fluorouracil for adjuvant therapy of resected colon carcinoma. N Engl J Med 322:352–358 Quasar Collaborative Group, Gray R, Barnwell J, McConkey C, Hills RK, Williams NS, Kerr DJ, 2007, Adjuvant chemotherapy versus observation in patients with colorectal cancer: a randomised study. Lancet 370:2020–2029 Advanced Colorectal Cancer Meta-Analysis Project, 1992, Modulation of fluorouracil by leucovorin in patients with advanced colorectal cancer: evidence in terms of response rate. J Clin Oncol 10:896–903 Allegra C, 2002, Thymidylate synthase levels: prognostic, predictive, or both? J Clin Oncol 20:1711–1713 Longley DB, Harkin DP, Johnston PG, 2003, 5-Fluorouracil: mechanisms of action and clinical strategies. Nat Rev Cancer 3: 330–338 De Angelis PM, Svendsrud DH, Kravik KL, Stokke T, 2006, Cellular response to 5-fluorouracil, 5-FU, in 5-FU-resistant colon cancer cell lines during treatment and recovery. Mol Cancer 5:20 Edler D, Glimelius B, Hallstrom M, Jakobsen A, Johnston PG, Magnusson I, Ragnhammar P, Blomgren H, 2002, Thymidylate synthase expression in colorectal cancer: a prognostic and predictive marker of benefit from adjuvant fluorouracil-based chemotherapy. J Clin Oncol 20:1721–1728 Soong R, Shah N, Salto-Tellez M, Tai BC, Soo RA, Han HC, Ng SS, Tan WL, Zeps N, Joseph D, Diasio RB, Iacopetta B, 2008, Prognostic significance of thymidylate synthase, dihydropyrimidine dehydrogenase and thymidine phosphorylase protein expression in colorectal cancer patients treated with or without 5-fluorouracil-based chemotherapy. Ann Oncol 19:915–919 Popat S, Chen Z, Zhao D, Pan H, Hearle N, Chandler I, Shao Y, Aherne W, Houlston R, 2006, A prospective, blinded analysis of thymidylate synthase and p53 expression as prognostic markers in the adjuvant treatment of colorectal cancer. Ann Oncol 17:1810– 1817 Showalter SL, Showalter TN, Witkiewicz A, Havens R, Kennedy EP, Hucl T, Kern SE,Yeo CJ, Brody JR, 2008, Evaluating the drugtarget relationship between thymidylate synthase expression and tumor response to 5-fluorouracil. Is it time to move forward? Cancer Biol Ther 7:986–994 Bunz F, 2008, Thymidylate synthase and 5-fluorouracil: a cautionary tale. Cancer Biol Ther 7:995–996 Sulzyc-Bielicka V, Domagala P, Urasinska E, Bielicki D, Safranow K, Domagala W, 2011, Expression of p21WAF1 in Astler-Coller stage B2 colorectal cancer is associated with survival benefit from 5FU-based adjuvant chemotherapy. Virchows Arch 458:431–438 Sulzyc-Bielicka V, Domagala P, Bielicki D, Safranow K, Domagala W, 2014, Thymidylate synthase expression and p21(WAF1)/p53 phenotype of colon cancers identify patients who may benefit from 5-fluorouracil based therapy. Cell Oncol 37:17–28 Enders GH, 2004, Colon cancer metastasis: is E2F-1 a driving force? Cancer Biol Ther 3:400–401 Banerjee D, Schnieders B, Fu JZ, Adhikari D, Zhao SC, Bertino JR, 1998, Role of E2F-1 in chemosensitivity. Cancer Res 58:4292– 4296 DeGregori J, Kowalik T, Nevins JR, 1995, Cellular targets for activation by the E2F1 transcription factor include DNA synthesis and G1/S-regulatory genes. Mol Cell Biol 15:4215–4224 KasaharaM, Takahashi Y, Nagata T, Asai S, Eguchi T, Ishii Y, Fujii M, Ishikawa K, 2000, Thymidylate synthase expression correlates closely with E2F1 expression in colon cancer. Clin Cancer Res 6: 2707–2711 Bramis J, Zacharatos P, Papaconstantinou I, Kotsinas A, Sigala F, Korkolis DP, Nikiteas N, Pazaiti A, Kittas C, Bastounis E, Gorgoulis VG, 2004, E2F-1 transcription factor immunoexpression is inversely associated with tumor growth in colon adenocarcinomas. Anticancer Res 24:3041–3047 Crosby ME, Almasan A, 2004, Opposing roles of E2Fs in cell proliferation and death. Cancer Biol Ther 3:1208–1211 Muller H, Bracken AP, Vernell R, Moroni MC, Christians F, Grassilli E, Prosperini E, Vigo E, Oliner JD, Helin K, 2001, E2Fs regulate the expression of genes involved in differentiation, development, proliferation, and apoptosis. Genes Dev 15:267–285 Dyson N, 1998, The regulation of E2F by pRB-family proteins. Genes Dev 12:2245–2262 YasuiW, Fujimoto J, Suzuki T, Ono S, Naka K,Yokozaki H, Tahara E, 1999, Expression of cell-cycle-regulating transcription factor E2F-1 in colorectal carcinomas. Pathobiology 67:174–179 Belvedere O, Puglisi F, Di Loreto C, Cataldi P, Guglielmi A, Aschele C, Sobrero A, 2004, Lack of correlation between immunohistochemical expression of E2F-1, thymidylate synthase expression and clinical response to 5-fluorouracil in advanced colorectal cancer. Ann Oncol 15:55–58 Shia J, 2008, Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part I. The utility of immunohistochemistry. J Mol Diagn 10:293–300 Chung GG, Kielhorn EP, Rimm DL, 2002, Subjective differences in outcome are seen as a function of the immunohistochemical method used on a colorectal cancer tissue microarray. Clin Colorectal Cancer 1:237–242 Detre S, Saclani Jotti G, DowsettM(1995, A "quickscore" method for immunohistochemical semiquantitation: validation for oestrogen receptor in breast carcinomas. J Clin Pathol 48:876–878 Sulzyc-Bielicka V, Domagala P, Hybiak J, Majewicz-Broda A, Safranow K, Domagala W, 2012, Colorectal cancers differ in respect of PARP-1 protein expression. Pol J Pathol 63:87–92 Obama K, Kanai M, Kawai Y, Fukushima M, Takabayashi A, 2002, Role of retinoblastoma protein and E2F-1 transcription factor in the acquisition of 5-fluorouracil resistance by colon cancer cells. Int J Oncol 21:309–314 Yoshinare K, Kubota T, Watanabe M, Wada N, Nishibori H, Hasegawa H, Kitajima M, Takechi T, Fukushima M, 2003, Gene expression in colorectal cancer and in vitro chemosensitivity to 5- fluorouracil: a study of 88 surgical specimens. Cancer Sci 94:633– 638 van Triest B, Pinedo HM, Blaauwgeers JL, van Diest PJ, Schoenmakers PS, Voorn DA, Smid K, Hoekman K, Hoitsma HF, Peters GJ, 2000, Prognostic role of thymidylate synthase, thymidine phosphorylase/platelet-derived endothelial cell growth factor, and proliferation markers in colorectal cancer. Clin Cancer Res 6:1063–1072 Araki K, Nakajima Y, Eto K, IkedaMA(2003, Distinct recruitment of E2F family members to specific E2F-binding sites mediates activation and repression of the E2F1 promoter. Oncogene 22:7632– 7641 Yamasaki L, Jacks T, Bronson R, Goillot E, Harlow E, Dyson NJ, 1996, Tumor induction and tissue atrophy in mice lacking E2F-1. Cell 85:537–548 Stanelle J, Stiewe T, Theseling CC, Peter M, Putzer BM, 2002, Gene expression changes in response to E2F1 activation. Nucleic Acids Res 30:1859–1867 Elliott MJ, Dong YB, Yang H, McMasters KM, 2001, E2F-1 upregulates c-Myc and p14(ARF, and induces apoptosis in colon cancer cells. Clin Cancer Res 7:3590–3597 Vorburger SA, PataerA, Yoshida K, Liu Y, Lu X, Swisher SG, Hunt KK, 2003, The mitochondrial apoptosis-inducing factor plays a role in E2F-1-induced apoptosis in human colon cancer cells. Ann Surg Oncol 10:314–322 Zacharatos P, Kotsinas A, Evangelou K, Karakaidos P, Vassiliou LV, Rezaei N, Kyroudi A, Kittas C, Patsouris E, Papavassiliou AG, Gorgoulis VG, 2004, Distinct expression patterns of the transcription factor E2F-1 in relation to tumour growth parameters in common human carcinomas. J Pathol 203:744–753 Iwamoto M, Banerjee D, Menon LG, Jurkiewicz A, Rao PH, Kemeny NE, Fong Y, Jhanwar SC, Gorlick R, Bertino JR, 2004, Overexpression of E2F-1 in lung and liver metastases of human colon cancer is associated with gene amplification. Cancer Biol Ther 3:395–399 Banerjee D, Gorlick R, Liefshitz A, Danenberg K, Danenberg PC, Danenberg PV, Klimstra D, Jhanwar S, Cordon-Cardo C, Fong Y, Kemeny N, Bertino JR, 2000, Levels of E2F-1 expression are higher in lung metastasis of colon cancer as compared with hepatic metastasis and correlate with levels of thymidylate synthase. Cancer Res 60:2365–2367 Brody JR, Hucl T, Costantino CL, Eshleman JR, Gallmeier E, Zhu H, van der Heijden MS, Winter JM, Wikiewicz AK, Yeo CJ, Kern SE, 2009, Limits to thymidylate synthase and TP53 genes as predictive determinants for fluoropyrimidine sensitivity and further evidence for RNA-based toxicity as a major influence. Cancer Res 69:984–991 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 601 EP 608 DI 10.1007/s12253-016-0043-z PG 8 ER PT J AU Chen, J Hu, L Chen, J Pan, Q Ding, H Xu, G Zhu, P Wen, X Huang, K Wang, Y AF Chen, Jie Hu, Lijuan Chen, Jian Pan, Qinshi Ding, Hongyan Xu, Gang Zhu, Peiwu Wen, Xiusu Huang, Keta Wang, Yumin TI Detection and Analysis of Wnt Pathway Related lncRNAs Expression Profile in Lung Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung adenocarcinoma; LncRNAs; Wnt pathway; Expression profile ID Lung adenocarcinoma; LncRNAs; Wnt pathway; Expression profile AB Studies have shown that the expression profile of Wnt signaling pathway is very important in lung adenocarcinoma (LAD) and some lncRNAs can regulate the expression of key molecules of Wnt pathway. However, Wnt pathway related lncRNAs are not systematically analyzed and detected in lung adenocarcinoma. We used a high-throughput microarray to compare the lncRNA expression profiles in LAD and corresponding normal tissue (NT) samples. Several candidate Wnt pathway related lncRNAs were verified by real-time quantitative reverse transcription polymerase chain reaction (PCR) analysis. We found that 232 Wnt pathway related lncRNAs were obviously expressed (≥2-fold change) in lung adenocarcinoma samples and 13 Wnt pathway related lncRNAs were aberrantly expressed in lung adenocarcinoma compared with matched histologically normal lung tissues by qPCR. Among these, RP11-181G12.2 and RP11-89 K21.1 were the most aberrantly expressed lncRNAs. Our study ascertained the expression of Wnt pathway related lncRNAs in lung adenocarcinoma. The results revealed that many Wnt pathway related lncRNAs were differentially expressed in lung adenocarcinoma tissues, suggesting that they may play a key role in tumor development. C1 [Chen, Jie] The First Affiliated Hospital of Wenzhou Medical University, Department of Intensive Care UnitWenzhou, Zhejiang Province, China. [Hu, Lijuan] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang Province, China. [Chen, Jian] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang Province, China. [Pan, Qinshi] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang Province, China. [Ding, Hongyan] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang Province, China. [Xu, Gang] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang Province, China. [Zhu, Peiwu] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang Province, China. [Wen, Xiusu] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang Province, China. [Huang, Keta] The First Affiliated Hospital of Wenzhou Medical University, Department of PathologyWenzhou, Zhejiang Province, China. [Wang, Yumin] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang Province, China. RP Wang, Y (reprint author), The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, China. EM wym0577@163.com CR Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, Feuer EJ, Thun MJ, 2005, Cancer statistics, 2005. CA: a Cancer Journal for Clinicians 55:10–30 Gupta A, Verma A, Mishra AK, Wadhwa G, Sharma SK, Jain CK, 2013, The wnt pathway: emerging anticancer strategies. Recent Patents on Endocrine, Metabolic & Immune Drug Discovery 7: 138–147 Le PN, McDermott JD, Jimeno A, 2015, Targeting the wnt pathway in human cancers: therapeutic targeting with a focus on OMP- 54F28. Pharmacology & Therapeutics. 146:1–11 Stewart DJ, ChangDW, Ye Y, Spitz M, Lu C, Shu X,Wampfler JA, Marks RS, Garces YI, Yang P,Wu X, 2014)Wnt signaling pathway pharmacogenetics in non-small cell lung cancer. The Pharmacogenomics Journal 14:509–522 Stewart DJ, 2014, Wnt signaling pathway in non-small cell lung cancer. J Natl Cancer Inst 106:djt356 Yao L, Sun B, Zhao X, Zhao X, Gu Q, Dong X, Zheng Y, Sun J, Cheng R, Qi H, An J, 2014, Overexpression of Wnt5a promotes angiogenesis in NSCLC. BioMed Research International 2014: 832562 Qiu M, Xu Y, Wang J, Zhang E, Sun M, Zheng Y, Li M, Xia W, Feng D, Yin R, Xu L, 2015, A novel lncRNA, LUADT1, promotes lung adenocarcinoma proliferation via the epigenetic suppression of p27. Cell Death & Disease 6:e1858 Wang Y, ChenW, Chen J, Pan Q, Pan J, 2014, LncRNA expression profiles of EGFR exon 19 deletions in lung adenocarcinoma ascertained by using microarray analysis. Medical Oncology. 31:137 Cai Y, He J, Zhang D, 2015, Long noncoding RNA CCAT2 promotes breast tumor growth by regulating thewnt signaling pathway. OncoTargets and Therapy 8:2657–2664 MaY,YangY,Wang F,MoyerMP,WeiQ, Zhang P,Yang Z, LiuW, Zhang H, Chen N, Wang H, Wang H, Qin H, 2015, Long non-coding RNA CCAL regulates colorectal cancer progression by activating wnt/beta-catenin signalling pathway via suppression of activator protein 2alpha., DOI 10.1136/gutjnl-2014-308392 Wang Y, He L, Du Y, Zhu P, Huang G, Luo J, Yan X, Ye B, Li C, Xia P, Zhang G, Tian Y, Chen R, Fan Z, 2015, The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of wnt signaling. Cell Stem Cell 16:413–425 Carrion K, Dyo J, Patel V, Sasik R, Mohamed SA, Hardiman G, Nigam V, 2014, The long non-coding HOTAIR is modulated by cyclic stretch andWNT/beta-CATENIN in human aortic valve cells and is a novel repressor of calcification genes. PLoS ONE 9:e96577 Wang G, Li Z, Zhao Q, Zhu Y, Zhao C, Li X, Ma Z, Li X, Zhang Y, 2014, LincRNA-p21 enhances the sensitivity of radiotherapy for human colorectal cancer by targeting the wnt/beta-catenin signaling pathway. Oncology Reports. 31:1839–1845 Fan Y, Shen B, Tan M, Mu X, Qin Y, Zhang F, Liu Y, 2014, Long non-coding RNA UCA1 increases chemoresistance of bladder cancer cells by regulating wnt signaling. The FEBS Journal 281:1750– 1758 Ren S, Peng Z,Mao JH, Yu Y, Yin C, Gao X, Cui Z, Zhang J, Yi K, XuW, Chen C,Wang F, Guo X, Lu J, Yang J,WeiM, Tian Z, Guan Y, Tang L, Xu C,Wang L, Gao X, TianW,Wang J, Yang H,Wang J, Sun Y, 2012, RNA-seq analysis of prostate cancer in the Chinese population identifies recurrent gene fusions, cancer-associated long noncoding RNAs and aberrant alternative splicings. Cell Research. 22:806–821 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 609 EP 615 DI 10.1007/s12253-016-0046-9 PG 7 ER PT J AU Kanizsai, Sz Ongradi, J Aradi, J Nagy, K AF Kanizsai, Szilvia Ongradi, Jozsef Aradi, Janos Nagy, Karoly TI New Approach for Inhibition of HIV Entry: Modifying CD4 Binding Sites by Thiolated Pyrimidine Derivatives SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HIV entry; Thiolated pyrimidine derivatives; HIV-1 pseudotypes; Redox processes; CD4 ID HIV entry; Thiolated pyrimidine derivatives; HIV-1 pseudotypes; Redox processes; CD4 AB Thiolated pyrimidine derivatives have been synthetized and their antiretroviral effect against human immunodeficiency virus type 1 (HIV-1IIIB) and HIV-1 chimeric pseudovirions have been quantitatively determined in cellbased viral infectivity assays including syncytium inhibition assay as well as a single-cycle viral infection assay on HeLaCD4- LTR/ß-gal cells. Pseudotype virions prepared bearing HIV-1 envelope preference for CCR5 coreceptor, CXCR4 coreceptor or for both, respectively, with a HIV-1 core containing luciferase reporter gene were able to infect susceptible cells but are replication defective so unable to replicate in the cells . Data indicate that thiolated pyrimidine derivatives inhibited effectively virally induced cell fusion in vitro aswell as infectivity of primary HIV- 1IIIB strain and HIV-1 pseudovirions using chemokine receptors CCR5 or CXCR4 or both for virus entry a dose dependent manner. Inhibition was selective, depended on the pseudovirus coreceptor preference. Our results suggest that some of these sulfur containing pyrimidines interact with redoxactive -SH groups required for successful HIV entry, including a redox active disulfide in the CD4 molecule as well as -SH groups in HIV viral envelope gp120. This mode of action is unique representing a new class of potential HIV entry inhibitors. C1 [Kanizsai, Szilvia] Semmelweis University, Department of Medical MicrobiologyBudapest, Hungary. [Ongradi, Jozsef] Semmelweis University, Department of Medical MicrobiologyBudapest, Hungary. [Aradi, Janos] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Nagy, Karoly] Semmelweis University, Department of Medical MicrobiologyBudapest, Hungary. RP Nagy, K (reprint author), Semmelweis University, Department of Medical Microbiology, Budapest, Hungary. EM nagy.karoly@med.semmelweis-univ.hu CR Kaplan JE, Hanson D, Dworkin MS, Frederick T, Bertolli J, Lindegren ML, Holmberg S, Jones JL, 2000, Epidemiology of human immunodeficiency virus-associated opportunistic infectiopns in the United States in the era of highly active antiretroviral therapy. Clin Infect Dis 30:S5–S14 Baeten JM, Donnell D, Ndase P, Mugo NR, Campbell JD,Wangisi J, Tappero JW, Bukusi EA, Cohen CR, Katabira E et al, 2012, Antiretroviral prophylaxis for HIV prevention in heterosexual men and women. N Engl JMed 367:399–410 Kanizsai S, Ghidan A, Ujhelyi E, Banhegyi D, Nagy K, 2001, Monitoring of drug resistance in therapy-naive HIV infected patients and detection of African HIV subtypes in Hungary. Acta Microbiol Immunol Hung 57(1):55–68 Triant VA, Lee H, Hadigan C, Grinspoon SK, 2007, Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immundiodeficiency virus disease. J Clin Endocrinol Metab 92:2506–25012 Monforte AD, Abrams D, Pradier C, Weber R, Reiss P, Bonnet F, Kirk O, LawM, deWitt S, Friis-Moller N et al, 2008, HIV-induced immunodeficiency and mortality from AIDS-defining and non- AIDS-defining malihgnancies. AIDS 22:2143–2153 Neuhaus J, Jacobs DR, Baker JV, Calmy A, Duprez D, la Rosa A, Kuller LH, Pett SL, Ristola M, Ross MJ, 2010, J Infect Dis 201: 1788–1795 Didigu C, Doms RW, 2014, Gene therapy targeting HIV entry. Viruses 6:1395–1409 Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, di Marzio P, Marmon S, Sutton RE, Hill CM et al, 1996, Identification of major co-receptor for primary isolates of HIV-1. Nature 381:661–666 Kemeny B, Nagy K, Horvath A, 2000, CCR5 and SDF1 gene polymorphism in HIV-infected and healthy individuals in Hungary. Hung Venerol Arch IV(2–3):89–92 Ryser HJ, Levy E, Mandel R, DiSciullo GJ, 1994, Inhibition of human immunodeficiency virus infection by agents that interfere with thiol-disulfide interchange upon virus-receptor interaction. Proc Natl Acad Sci U S A 91:4559–4563 Barbouche R, Miquelist R, Jones IM, Fenouillet E, 2003, Proteindisulphide isomerase-mediated reduction of two disulfide binds of HIVenvelope glycoprotein 120 occurs poet-CXCR4 binding and is required for fusion. J Biol Chem 278(5):3131–3136 Beck Z, Kis A, Berenyi E, Kovacs P, Fesus L, Aradi J, 2009, 4- Thio-uridylate, UD29, interferes with the function of protein –SH and inhibits HIV replication in vitro. Pharmacol Rep 61:343–347 Campbell S, Crowe SM, Mak J, 2001, Lipid refats and HIV-1: from viral entry to assembly of progeny virions. J Clin Virol 22:217–227 Kanizsai S, Ongradi J, Aradi J, Nagy K, 2014, Thiolated pyrimidine derivatives may interfere thiol groups concentrated at lipid rafts of HIV-infected cells. Acta Microbiol Immunol Hung 61(4): 447–458 Kanizsai S, Ghidan A, Ongradi J, Nagy K, 2012, Antiretroviral effect of 4-thio-uridylate against human immunodeficiency virus type. Acta Microbiol Immunol Hung 59(4):499–510 Nagy K, 2007, HIV pseudovirions assay for coreceptor selection and entry fitness. Proc. HIV correlates, Ed:Fust G, p12., Budapest Matthias LJ,Yam PT, JiangXM, Vandegraaff N, Li P, Poumbourios P, Donoghue N, Hogg PJ, 2002, Disulfide exchange in domain 2 of CD4 is required for entry of HIV-1. Nat Immunol 3:727–733 Horvath A, Tokes S, Hartman T, Watson K, Turpin JA, Buckheit RW Jr, Sebestyen Z, Szollosi J, Benko I, Bardos TJ, Dunn J, Fesus L, Toth FD, Aradi J, 2005, Potent inhibition of HIV-1 entry by Suligovir, s4dU)3. Virology 334: 214–223 Jarmy G, Henekelein M, Weissbrich B, Jassoy C, Rethwilm A, 2001, Phenotypic analysis of the sensitivity of HIV-1 to inhibitors of the reverse transcriptase, protease and integrase using a self-inactivating virus vector system. J Med Virol 64(3):223–231 Nagy K, Young M, Baboonian C, Merson J, Whittle P, Oroszlan S, 1994, Antiviral activity of human immunodeficiency virus type 1 protease inhibitors in a single cycle of infection: Evidence for a role of protease in the early phase. J Virol 68:757–765 Nagy K, Clapham P, Cheingsong-Popov R, Weiss RA, 1983, Human-T-cell leukemia virus type I: induction of syncytia and inhibition by patients’ sera. Int J Cancer 32:321–328 Clapham P, Nagy K, Cheingsong-Popov R, Exley M, Weiss RA, 1983, Productive infection and cell-free transmission of human T-cell leukemia virus in a non-lymphoid cell line. Science 222:1125–1127 Didigu C, Doms RW(2012, Novel approaches to inhibit HIV entry. Viruses 4:309–424 Dumas F, Preira P, Salome L, 2014, Membrane organization of virus and target cell plays a role in HIV entry. Biochimie 107:22–27 Zarr M, Siliciano R, 2015, Stoichiometric parameters of HIV-1 entry. Virology 474:1–9 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 617 EP 623 DI 10.1007/s12253-016-0044-y PG 7 ER PT J AU Yamada, Shi Yanamoto, S Naruse, T Matsushita, Y Takahashi, H Umeda, M Nemoto, KT Kurita, H AF Yamada, Shin-ichi Yanamoto, Souichi Naruse, Tomofumi Matsushita, Yuki Takahashi, Hidenori Umeda, Masahiro Nemoto, K Takayuki Kurita, Hiroshi TI Skp2 Regulates the Expression of MMP-2 and MMP-9, and Enhances the Invasion Potential of Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Skp2; invasion; metastasis; migration; oral squamous cell carcinoma ID Skp2; invasion; metastasis; migration; oral squamous cell carcinoma AB Oral squamous cell carcinoma (OSCC) is the most common malignant tumor of the head and neck regions and accounts for more than 90 % of cancers in the oral cavity. Sphase kinase-associated protein-2 (Skp2) is a member of the F-box protein family and the substrate recognition subunit of the Skp1-Cullin-F box protein E3 ubiquitin ligase complex. Skp2 is oncogenic and overexpressed in human cancers. The aims of the present study were to determine the clinicopathological significance of Skp2 in OSCC and clarify its function in OSCC cell lines in vitro. Multiple methods including immunohistochemical staining, RT-PCR, western blotting, migration and invasion assays, and siRNA transfection were employed in order to investigate the clinicopathological significance and molecular function of Skp2 in OSCC. The overexpression of Skp2 was more frequent in OSCC than in the normal oral epithelium. It was also more frequently detected in cancers with higher grades according to the Tclassification, N classification, and pattern of invasion. The high-Skp2 expression group had a significantly poorer prognosis, at 30.1 %, than that of the low-expression group, at 63.0 %. The downregulation of Skp2 decreased migration and invasion potentials in HSC3 cells. Moreover, the suppression of Skp2 reduced the enzyme activities of MMP-2 and MMP-9 via Sp1. Skp2 may be a prognostic factor in OSCC patients, and may also play crucial roles in the migration and invasion potentials of OSCC cells. C1 [Yamada, Shin-ichi] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. [Yanamoto, Souichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral OncologyNagasaki, Japan. [Naruse, Tomofumi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral OncologyNagasaki, Japan. [Matsushita, Yuki] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral OncologyNagasaki, Japan. [Takahashi, Hidenori] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral OncologyNagasaki, Japan. [Umeda, Masahiro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral OncologyNagasaki, Japan. [Nemoto, K Takayuki] Nagasaki University Graduate School of Biomedical Sciences, Unit of Basic Medical Sciences, Course of Medical and Dental Sciences, Department of Oral Molecular BiologyNagasaki, Japan. [Kurita, Hiroshi] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. RP Yamada, Shi (reprint author), Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 390-8621 Matsumoto, Japan. EM yshinshin@shinshu-u.ac.jp CR Johnson NW, Jayasekara D, 2000, Amarasinghe AA, 2011, squamous cell carcinoma and precursor lesions of the oral cavity: epidemiology and aetiology. Periodontol 57(1):19–37 HicksWL Jr, North JH Jr, Loree TR,Maamoun S,Mullins A, Orner JB, Bakamjian VY, Shedd DP, 1998, Surgery as a single modality therapy for squamous cell carcinoma of the oral tongue. Am J Otolaryngol 19(1):24–28 Sessions DG, Spector GJ, Lenox J, Parriott S, Haughey B, Chao C, Marks J, Perez C, 2003, Analysis of treatment results for base of tongue cancer. Laryngoscope 113(7):1252–1261 Gonzalez-Garcia R, Naval-Gias L, Rodriguez-Campo FJ, Sastre- Perez J, Munoz-Guerra MF, Gil-Diez Usandizaga JL, 2008, Contralateral lymph neck node metastasis of squamous cell carcinoma of the oral cavity: a retrospective analytic study in 315 patients. J Oral Maxillofac Surg 66(7):1390–1398 Ziober BL, Silverman SS Jr, Kramer RH, 2001, Adhesive mechanisms regulating invasion and metastasis in oral cancer. Crit Rev Oral Biol Med 12(6):499–510 Bankfalvi A, Krassort M, Buchwalow IB, Vegh A, Felszeghy E, Piffko J, 2002, Gains and losses of adhesion molecules, CD44, Ecadherin, and beta-catenin, during oral carcinogenesis and tumour progression. J Pathol 198(3):343–351 Arora S, Kaur J, Sharma C,MathurM, Bahadur S, Shukla NK, Deo SV, Ralhan R, 2005, Stromelysin 3, ets-1, and vascular endothelial growth factor expression in oral precancerous and cancerous lesions: correlation with microvessel density, progression, and prognosis. Clin Cancer Res 11(6):2272–2284 Yanamoto S, Kawasaki G, Yoshitomi I, Iwamoto T, Hirata K, Mizuno A, 2007, Clinicopathologic significance of EpCAM expression in squamous cell carcinoma of the tongue and its possibility as a potential target for tongue cancer gene therapy. Oral Oncol 43(9):869–877 Brinkman BM, Wong DT, 2006, Disease mechanism and biomarkers of oral squamous cell carcinoma. Curr Opin Oncol 18(2): 228–233 Berx G, Raspe E, Christofori G, Thiery JP, Sleeman JP, 2007, Pre- EMTing metastasis? recapitulation of morphogenetic processes in cancer. Clin Exp Metastasis 24(8):587–597 Kalluri R, Weinberg RA, 2009, The basics of epithelialmesenchymal transition. J Clin Invest 119(6):1420–1428 Baranwal S, Alahari SK, 2009, Molecular mechanisms controlling E-cadherin expression in breast cancer. Biochem Biophys Res Commun 384(1):6–11 WeiW, Ayad NG,Wan Y, Zhang GJ, KirschnerMW, KaelinWGJr, 2004, Degradation of the SCF component Skp2 in cell-cycle phase G1 by the anaphase-promoting complex. Nat 11; 428:194–198 Sutterluty H, Chatelain E, Marti A, Wirbelauer C, Senften M, Muller U, 1999, KrekW(1999, p45SKP2 promotes p27Kip1 degradation and induces S phase in quiescent cells. Nat Cell Biol 1(4): 207–214 Carrano AC, Eytan E, Hershko A, Pagano M, 1999, SKP2 is required for ubiquitin-mediated degradation of the CDK inhibitor p27. Nat Cell Biol 1(4):193–199 Porter PL, Malone KE, Heagerty PJ, Alexander GM, Gatti LA, Firpo EJ, Daling JR, Roberts JM, 1997, Expression of cell-cycle regulators p27Kip1 and cyclin E, alone and in combination, correlate with survival in young breast cancer patients. Nat Med 3(2): 222–225 Latres E, Chiarle R, Schulman BA, Pavletich NP, Pellicer A, Inghirami G, Pagano M, 2001, Role of the F-box protein Skp2 in lymphomagenesis. Proc Natl Acad Sci U S A 98(5):2515–2520 KudoY, Kitajima S, Sato S, Miyauchi M, Ogawa I, Takata T, 2001, High expression of S-phase kinase-interacting protein 2, human Fbox protein, correlates with poor prognosis in oral squamous cell carcinomas. Cancer Res 61(19):7044–7047 Masuda TA, Inoue H, Sonoda H,Mine S, Yoshikawa Y, Nakayama K, Nakayama K, Mori M, 2002, Clinical and biological significance of S-phase kinase-associated protein 2, Skp2, gene expression in gastric carcinoma: modulation of malignant phenotype by Skp2 overexpression, possibly via p27 proteolysis. Cancer Res62(13):3819–3825 Yokoi S, Yasui K, Mori M, Iizasa T, Fujisawa T, Inazawa J, 2004, Amplification and overexpression of SKP2 are associated with metastasis of non-small-cell lung cancers to lymph nodes. Am J Pathol 165(1):175–180 Harada K, SupriatnoKS, Kawashima Y, Itashiki Y, Yoshida H, Sato M, 2005, High expression of S-phase kinase-associated protein 2, Skp2, is a strong prognostic marker in oral squamous cell carcinoma patients treated by UFT in combination with radiation. Anticancer Res 25(3c):2471–2475 Carracedo DG, Astudillo A, Rodrigo JP, Suarez C, Gonzalez MV, 2008, Skp2, p27kip1 and EGFR assessment in head and neck squamous cell carcinoma: prognostic implications. Oncol Rep 20(3):589–595 Wang XC,Wu YP, Ye B, Lin DC, Feng YB, Zhang ZQ, Xu X, Han YL, Cai Y, Dong JT, Zhan QM, Wu M, Wang MR, 2009, Suppression of anoikis by SKP2 amplification and overexpression promotes metastasis of esophageal squamous cell carcinoma. Mol Cancer Res 7(1):12–22 Wang J, Huang Y, Guan Z, Zhang JL, Su HK, Zhang W, Yue CF, YanM, Guan S, Liu QQ, 2014, E3-ligase Skp2 predicts poor prognosis and maintains cancer stem cell pool in nasopharyngeal carcinoma. Oncotarget 5(14):5591–55601 Chan CH, Morrow JK, Li CF, Gao Y, Jin G, Moten A, Stagg LJ, Ladbury JE, Cai Z, Xu D, Logothetis CJ, Hung MC, Zhang S, Lin HK, 2013, Pharmacological inactivation of Skp2 SCF ubiquitin ligase restricts cancer stem cell traits and cancer progression. Cell 154(3):556–568 Yang Q, Huang J, Wu Q, Cai Y, Zhu L, Lu X, Chen S, Chen C, Wang Z, 2014, Acquisition of epithelial-mesenchymal transition is associated with Skp2 expression in paclitaxel-resistant breast cancer cells. Br J Cancer 110(8):1958–1967 Bryne M, Boysen M, Alfsen CG, Abeler VM, Sudbo J, Nesland JM, Kristensen GB, Piffko J, Bankfalvi A, 1998, The invasive front of carcinomas. the most important area for tumour prognosis? Anticancer Res 18(6B):4757–4764 Yamada S, Yanamoto S, Kawasaki G, Mizuno A, Nemoto TK, 2010, Overexpression of cortactin increases invasion potential in oral squamous cell carcinoma. Pathol Oncol Res 16(4):523–531 Yamada S, Yanamoto S, Yoshida H, Yoshitomi I, Kawasaki G, Mizuno A, Nemoto TK, 2010, RNAi-mediated down-regulation of alpha-actinin-4 decreases invasion potential in oral squamous cell carcinoma. Int J Oral Maxillofac Surg 39(1):61–67 Yamada S, Yanamoto S, Kawasaki G, Rokutanda S, Yonezawa H, Kawakita A, Nemoto TK, 2011, Overexpression of CRKII increases migration and invasive potential in oral squamous cell carcinoma. Cancer Lett 303(2):84–91 Noel A, Jost M, Maquoi E, 2008, Matrix metalloproteinases at cancer tumor-host interface. Semin Cell Dev Biol 19(1):52–60 Mook OR, Frederiks WM, Van Noorden CJ, 2004, The role of gelatinases in colorectal cancer progression and metastasis. Biochim Biophys Acta 1705(2):69–89 HungWC, TsengWL, Shiea J, Chang HC, 2010, Skp2 overexpression increases the expression of MMP-2 and MMP-9 and invasion of lung cancer cells. Cancer Lett 288(2):156–161 Lu H, Cao X, Zhang H, Sun G, Fan G, Chen L, Wang S, 2014, Imbalance between MMP-2, 9 and TIMP-1 promote the invasion and metastasis of renal cell carcinoma via SKP2 signaling pathways. Tumour Biol 35(10):9807–9813 Sonoda H, Inoue H, Ogawa K, Utsunomiya T, Masuda TA,MoriM, 2006, Significance of skp2 expression in primary breast cancer. Clin Cancer Res 12(4):1215–1220 Inuzuka H, GaoD, Finley LW,YangW,Wan L, Fukushima H, Chin YR, Zhai B, Shaik S, Lau AW, Wang Z, Gygi SP, Nakayama K, Teruya-Feldstein J, Toker A, Haigis MC, Pandolfi PP, Wei W, 2012, Acetylation-dependent regulation of Skp2 function. Cell 150(1):179–193 Qu X, Shen L, ZhengY, Cui Y, Feng Z, Liu F, Liu J, 2014, A signal transduction pathway from TGF-β1 to SKP2 via Akt1 and c-myc and its correlation with progression in human melanoma. J Invest Dermatol 134(1):159–167 Kajiyama H, Shibata K, Terauchi M, Yamashita M, Ino K, Nawa A, Kikkawa F, 2007, Chemoresistance to paclitaxel induces epithelialmesenchymal transition and enhances metastatic potential for epithelial ovarian carcinoma cells. Int J Oncol 31(2):277–283 Du F,Wu X, Liu Y,Wang T, Qi X, Mao Y, Jiang L, Zhu Y, Chen Y, Zhu R, Han X, Jin J, Ma X, Hua D, 2013, Acquisition of paclitaxel resistance via PI3K-dependent epithelial-mesenchymal transition in A2780 human ovarian cancer cells. Oncol Rep 30(3):1113–1118 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 625 EP 632 DI 10.1007/s12253-016-0049-6 PG 8 ER PT J AU Ferrer-Font, L Alcaraz, E Plana, M Candiota, PA Itarte, E Arus, C AF Ferrer-Font, Laura Alcaraz, Estefania Plana, Maria Candiota, Paula Ana Itarte, Emilio Arus, Carles TI Protein Kinase CK2 Content in GL261 Mouse Glioblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; CK2 content; Preclinical brain tumor; GBM therapeutic targets ID Glioma; CK2 content; Preclinical brain tumor; GBM therapeutic targets AB Glioblastoma (GBM) is the most prevalent and aggressive human glial tumour with a median survival of 14– 15 months. Temozolomide (TMZ) is the standard chemotherapeutic choice for GBM treatment. Unfortunately, chemoresistence always ensues with concomitant tumour regrowth. Protein kinase CK2 (CK2) contributes to tumour development, proliferation, and suppression of apoptosis in cancer and it is overexpressed in human GBM. Targeting CK2 in GBM treatment may benefit patients. With this translational perspective in mind, we have studied the CK2 expression level by Western blot analysis in a preclinical model of GBM: GL261 cells growing orthotopically in C57BL/6 mice. The expression level of the CK2 catalytic subunit (CK2α) was higher in tumour (about 4-fold) and in contralateral brain parenchyma (more than 2-fold) than in normal brain parenchyma (p < 0.05). In contrast, no significant changes were found in CK2 regulatory subunit (CK2β) expression, suggesting an increased unbalance of CK2α/CK2β in GL261 tumours with respect to normal brain parenchyma, in agreement with a differential role of these two subunits in tumours. C1 [Ferrer-Font, Laura] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, 08193 Cerdanyola del Valles, Spain. [Alcaraz, Estefania] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, 08193 Cerdanyola del Valles, Spain. [Plana, Maria] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, 08193 Cerdanyola del Valles, Spain. [Candiota, Paula Ana] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, 08193 Cerdanyola del Valles, Spain. [Itarte, Emilio] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, 08193 Cerdanyola del Valles, Spain. [Arus, Carles] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, 08193 Cerdanyola del Valles, Spain. RP Candiota, PA (reprint author), Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, 08193 Cerdanyola del Valles, Spain. EM AnaPaula.Candiota@uab.cat CR Buckner JC, 2003, Factors influencing survival in high-grade gliomas. Semin Oncol 30(6 Suppl 19):10–14 Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al., 2005, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 10;352(10):987–996. Huang Z, Cheng L, Guryanova OA, Wu Q, Bao S, 2010, Cancer stem cells in glioblastoma - molecular signalling and therapeutic targeting. Protein Cell 1(7):638–655 Ausman JI, ShapiroWR, Rall DP, 1970, Studies on the chemotherapy of experimental brain tumors: development of an experimental model. Cancer Res 30:2394–2400 Szatmari T, Lumniczky K, Desaknai S, Trajcevski S, Hidvegi EJ, Hamada H, et al., 2006, Detailed characterization of the mouse glioma 261 tumor model for experimental glioblastoma therapy. Cancer Sci 97(6):546–553 Delgado-Goni T, Julia-Sape M, Candiota AP, Pumarola M, Arus C, 2014, Molecular imaging coupled to pattern recognition distinguishes response to temozolomide in preclinical glioblastoma. NMR Biomed 27(11):1333–1345 Ducan JS, LitchfieldDW(2008, Too much of a good thing: the role of protein kinase CK2 in tumorigenesis and prospects for therapeutic inhibition of CK2. Biochim Biophys Acta 1784:33–47 Ruzzene M, Pinna LA, 2010, Addiction to protein kinase CK2: a common denominator of diverse cancer Cells? Biochim Biophys Acta 1804(3):499–504 Dixit D, Sharma V, Ghosh S,Mehta VS, Sen E, 2012, Inhibition of Casein kinase-2 induces p53-dependent cell cycle arrest and sensitizes glioblastoma cells to tumor necrosis factor, TNFα)-induced apoptosis through SIRT1 inhibition. Cell Death Dis 3:e271 Ji H, Lu Z, 2013, The role of protein kinase CK2 in glioblastoma development. Clin Cancer Res 19:6335–6337 Munstermann U, Fritz G, Seitz G, Yiping L, Schneider HR, Issinger OG, 1990, Casein kinase II is elevated in solid human tumours and rapidly proliferating non-neoplastic tissue. Eur J Biochem 189:251–257 Cozza G, Girardi C, Ranchio A, Liolli G, Sarno S, Orzeszko A, et al., 2014, Cell-permeable dual inhibitors of protein kinases CK2 and PIM-1: structural features and pharmacological potential. Cell Mol Life Sci 71:3173–3185 Kim J, Hwan KS, 2013, CK2 inhibitor CX-4945 blocks TGFbeta1- induced epithelial-to mesenchymal transition in A549 human lung adenocarcinoma cells. PLoS One 8(9):e74342 Simoes RV, Garcia-Martin ML, Cerdan S, Arus C, 2008, Perturbation of mouse glioma MRS pattern by induced acute hyperglycemia. NMR Biomed Mar;21(3):251–264. Mangiola A, Saulnier N, De Bonis P, Orteschi D, Sica G, Lama G, et al., 2013, Gene expression profile of glioblastoma peritumoral tissue: an ex vivo study. PLoS One 8:e57145. Bradford MM, 1976, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72:248–254 Lama G,Mangiola A, Anile C, Sabatino G, De Bonis P, Lauriola L, et al., 2007, Activated ERK1/2 expression in glioblastoma multiforme and peritumor tissue. Int J Oncol 30:1333–1342 Plotnikov A, Chuderland D, Karamansha Y, Livnah O, Seger R, 2011, Nuclear extracellular signal-regulated kinase 1 and 2 translocation is mediated by casein kinase 2 and accelerated by autophosphorylation. Mol Cell Biol 31(17):3515–3530 Agarwal M, Nitta RT, Li G, 2014, Casein kinase 2: A novel player in glioblastoma therapy and cancer stem cells. Mol Genet Med 8: pii: 1000094. Zheng Y, McFarland BC, Drygin D, Yu H, Bellis SL, Kim H, et al., 2013, Targeting protein kinase CK2 supresses prosurvival signaling pathways and growth of glioblastoma. Clin Cancer Res 19(23): 6484–6494 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 633 EP 637 DI 10.1007/s12253-015-9987-7 PG 5 ER PT J AU Sand, M Bechara, GF Skrygan, M Sand, D Gambichler, Th Bromba, M Stockfleth, E Hessam, Sch AF Sand, Michael Bechara, G Falk Skrygan, Marina Sand, Daniel Gambichler, Thilo Bromba, Michael Stockfleth, Eggert Hessam, Schapoor TI Mutation Scanning of D1705 and D1709 in the RNAse IIIb Domain of MicroRNA Processing Enzyme Dicer in Cutaneous Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Melanoma; Dicer; Mutation; MicroRNA ID Melanoma; Dicer; Mutation; MicroRNA AB Since the discovery of microRNAs (miRNAs) there have been performed several studies showing perturbations in the expression of miRNAs and the miRNA expression machinery in cutaneous melanoma. Dicer, a pivotal cytosolic enzyme of miRNA maturation has shown to be affected by both somatic and germline mutations in a variety of cancers. Recent studies have shown that recurrent somatic mutations of Dicer frequently affect the metal-ion-binding sites D1709 and D1705 of its RNase IIIb domain, therefore called hot spot mutations. The present study investigates metalion- binding sites D1709 and D1705 of the Dicer RNase IIIb domain in cutaneous melanomas and melanoma metastasis by Sanger sequencing. All investigated samples showed wildtype sequence and no single mutation was detected. The miRNA processing enzyme Dicer of melanoma and melanoma metastasis does not appear to be affected by mutation in the metal-ion-binding sites D1709 and D1705 of its RNase IIIb domain. C1 [Sand, Michael] Ruhr-University Bochum, St. Josef Hospital, Department of Dermatology, Venereology and Allergology, Dermatologic Surgery Unit, Gudrunstr. 56, 44791 Bochum, Germany. [Bechara, G Falk] Ruhr-University Bochum, St. Josef Hospital, Department of Dermatology, Venereology and Allergology, Dermatologic Surgery Unit, Gudrunstr. 56, 44791 Bochum, Germany. [Skrygan, Marina] Ruhr-University Bochum, St. Josef Hospital, Department of Dermatology, Venereology and Allergology, Dermatologic Surgery Unit, Gudrunstr. 56, 44791 Bochum, Germany. [Sand, Daniel] University of Michigan, Kellogg Eye Center, 1000 Wall Street, 48105 Ann Arbor, MI, USA. [Gambichler, Thilo] Ruhr-University Bochum, St. Josef Hospital, Department of Dermatology, Venereology and Allergology, Dermatologic Surgery Unit, Gudrunstr. 56, 44791 Bochum, Germany. [Bromba, Michael] Catholic Clinics of the Ruhr Peninsula, St. Josef Hospital, Department of Plastic Surgery, Heidbergweg 22-24, 45257 Essen, Germany. [Stockfleth, Eggert] Ruhr-University Bochum, St. Josef Hospital, Department of Dermatology, Venereology and Allergology, Dermatologic Surgery Unit, Gudrunstr. 56, 44791 Bochum, Germany. [Hessam, Schapoor] Ruhr-University Bochum, St. Josef Hospital, Department of Dermatology, Venereology and Allergology, Dermatologic Surgery Unit, Gudrunstr. 56, 44791 Bochum, Germany. RP Sand, M (reprint author), Ruhr-University Bochum, St. Josef Hospital, Department of Dermatology, Venereology and Allergology, Dermatologic Surgery Unit, 44791 Bochum, Germany. EM michael.sand@ruhr-uni-bochum.de CR Sand M, 2014, The pathway of miRNA maturation. Methods Mol Biol 1095:3–10., DOI 10.1007/978-1-62703-703-7_1 Foulkes WD, Priest JR, Duchaine TF, 2014, DICER1: mutations, microRNAs and mechanisms. Nat Rev Cancer 14(10):662–672., DOI 10.1038/nrc3802 Aksoy BA, Jacobsen A, Fieldhouse RJ, Lee W, Demir E, Ciriello G, Schultz N, Marks DS, Sander C, 2014, Cancer-associated recurrent mutations in RNase III domains of DICER1., DOI 10.1101/ 005686 Harbst K, Lauss M, Cirenajwis H,Winter C, Howlin J, Torngren T, Kvist A, Nodin B, Olsson E, Hakkinen J, Jirstrom K, Staaf J, Lundgren L, Olsson H, Ingvar C, Gruvberger-Saal SK, Saal LH, Jonsson G, 2014, Molecular and genetic diversity in the metastatic process of melanoma. J Pathol 233(1):39–50., DOI 10.1002/path. 4318 Sanger F, Coulson AR, Hong GF, Hill DF, Petersen GB, 1982, Nucleotide sequence of bacteriophage lambda DNA. J Mol Biol 162(4):729–773 Pellegrino L, Jacob J, Roca-Alonso L, Krell J, Castellano L, Frampton AE, 2013, Altered expression of the miRNA processing endoribonuclease dicer has prognostic significance in human cancers. Expert Rev Anticancer Ther 13(1):21–27., DOI 10.1586/era.12. 150 Sand M, Gambichler T, Sand D, Altmeyer P, Stuecker M, Bechara FG, 2011, Immunohistochemical expression patterns of the microRNA-processing enzyme dicer in cutaneous malignant melanomas, benign melanocytic nevi and dysplastic melanocytic nevi. Eur J Dermatol EJD 21(1):18–21., DOI 10. 1684/ejd.2011.1210 Nemlich Y, Greenberg E, Ortenberg R, Besser MJ, Barshack I, Jacob-Hirsch J, Jacoby E, Eyal E, Rivkin L, Prieto VG, Chakravarti N, Duncan LM, Kallenberg DM, Galun E, Bennett DC, Amariglio N, Bar-Eli M, Schachter J, Rechavi G, Markel G, 2013)MicroRNA-mediated loss of ADAR1 in metastatic melanoma promotes tumor growth. J Clin Invest 123(6):2703–2718., DOI 10.1172/JCI62980 Mione M, Bosserhoff A, 2014, MicroRNAs in melanocyte and melanoma biology. Pigment Cell Melanoma Res., DOI 10.1111/ pcmr.12346 Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Giannakakis A, Liang S, Naylor TL, Barchetti A, Ward MR, Yao G, Medina A, O’Brien-Jenkins A, Katsaros D, Hatzigeorgiou A, Gimotty PA, Weber BL, Coukos G, 2006, microRNAs exhibit high frequency genomic alterations in human cancer. Proc Natl Acad Sci U S A 103(24):9136–9141., DOI 10.1073/pnas.0508889103 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 639 EP 641 DI 10.1007/s12253-015-0034-5 PG 3 ER PT J AU Damasdi, M Jakab, F Kovacs, K Oldal, M Kemenesi, G Szabo, E Valyi-Nagy, I Pytel, Farkas, L Szanto, AF Damasdi, Miklos Jakab, Ferenc Kovacs, Krisztina Oldal, Miklos Kemenesi, Gabor Szabo, Eszter Valyi-Nagy, Istvan Pytel, Akos Farkas, Laszlo Szanto, Arpad TI Prevalence and Type Diversity of Human Papillomaviruses in Penile Cancers in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Damasdi, Miklos] University of Pecs, Department of UrologyPecs, Hungary. [Jakab, Ferenc] University of Pecs, Szentagothai Research Center, Ifjusag ut 20, H-7624 Pecs, Hungary. [Kovacs, Krisztina] University of Pecs, Department of PathologyPecs, Hungary. [Oldal, Miklos] University of Pecs, Szentagothai Research Center, Ifjusag ut 20, H-7624 Pecs, Hungary. [Kemenesi, Gabor] University of Pecs, Szentagothai Research Center, Ifjusag ut 20, H-7624 Pecs, Hungary. [Szabo, Eszter] Del-Pesti CentrumkorhazBudapest, Hungary. [Valyi-Nagy, Istvan] Del-Pesti CentrumkorhazBudapest, Hungary. [Pytel, Akos] University of Pecs, Department of UrologyPecs, Hungary. [Farkas, Laszlo] University of Pecs, Department of UrologyPecs, Hungary. [Szanto, Arpad] University of Pecs, Department of UrologyPecs, Hungary. RP Jakab, F (reprint author), University of Pecs, Szentagothai Research Center, H-7624 Pecs, Hungary. EM jakabf@gamma.ttk.pte.hu CR BleekerMCG, Heideman DAM, Snijders PJF, Horenblas S, Dillner J, Meijer CJLM, 2009, Penile cancer: epidemiology, pathogenesis and prevention. World J Urol 27:141–150 Miralles-Guri C, Bruni L, Cubilla AL, Castellsague X, Bosch FX, de Sanjose S, 2009, Human papillomavirus prevalence and type distribution in penile carcinoma. J Clin Pathol 62:870–878 Backes DM, Kurman RJ, Pimenta JM, Smith JS, 2009, Systematic review of human papillomavirus prevalence in invasive penile cancer. Cancer Causes Control 20:449–457 Novara G, Galfano A, De Marco V, Artibani W, Ficarra V, 2007, Prognostic factors in squamous cell carcinoma of the penile. Nat Clin Pract Urol 4:140–146 Lopes A, Hidalgo GS, Kowalski LP, Torloni H, Rossi BM, Fonseca FP, 1996, Prognostic factors in carcinoma of the penile: multivariate analysis of 145 patients treated with amputation and lymphadenectomy. J Urol 156:1637–1642 Rubin MA, Kleter B, Zhou M, Ayala G, Cubilla AL, Quint WGV, et al., 2001, Detection and typing human papillomavirus DNA in penile carcinoma, evidence for multiple independent pathways of penile carcinogenesis). Am J Pathol 159:1211–1218 Curado MP, Edwards B, Shin HR, Storm H, Ferlay J, Heanue M et al., 2007,, Ed(s)., Cancer incidence in five continents. IARC Scientific Publications, Lyon IX: 160 Morris BJ, Rose BR, 2007, Cervical screening in the 21st century: the case for human papillomavirus testing of self-collected specimens. Clin Chem Lab Med 45:577–591 Cubilla AL, Lloveras B, AlejoM, Clavero O, Chaux A, Kasamatsu E, et al., 2010, The basaloid cell is the best tissue marker for human papillomavirus in invasive penile squamous cell carcinoma: a study of 202 cases from Paraguay. Am J Surg Pathol 34:104–114 Lont AP, Kroon BK, Horenblas S, GalleMP, Berkhof J, Meijer CJ, et al., 2006, Presence of high-risk human papillomavirus DNA in penile carcinoma predicts favorable outcome in survival. Int J Cancer 119:1078–1081 Heideman DA, Waterboer T, Pawlita M, 2007, Human papillomavirus 16 is the predominant type etiologically involvedin penile squamous cell carcinoma. J Clin Oncol 25:4550–4556 Madsen BS, van den Bruke AJ, Jensen HL,Wohlfarhrt J, Frisch M, 2008, Risk factors for squamous cell carcinoma of the penilepopulation based case-control study in Denmark. Cancer Epidemiol Biomark Prev 17:2683–2691 Bezerra AL, Lopes A, Santiago GH, Ribeiro KC, LatorreMR, Villa LL, 2001, Human papillomavirus as a prognostic factor in carcinoma of the penis: analysis of 82 patients treated with amputation and bilateral lymphadenectomy. Cancer 91:2315–2321 Wiener JS, Effert PJ, Humphrey PA, Yu L, Liu ET, Walther PJ, 1992, Prevalence of human papillomavirus types 16 and 18 in squamous-cell carcinoma of the penis: a retrospective analysis of primary and metastatic lesions by differential polymerase chain reaction. Int J Cancer 50:694–701 Guerrero D, Guarch R, Ojer A, Casas JM, Ropero S, Mancha A, et al., 2008, Hypermethylation of the thrombospondin-1 gene is associated with poor prognosis in penile squamous cell carcinoma. BJU Int 102:747–755 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 643 EP 646 DI 10.1007/s12253-015-0026-5 PG 4 ER PT J AU Girdhar, Y Singh, N Behera, D Sharma, S AF Girdhar, Yashila Singh, Navneet Behera, Digambar Sharma, Siddharth TI Combinations of the Variant Genotypes of CYP1A1, GSTM1 and GSTT1 are Associated with an Increased Lung Cancer Risk in North Indian Population: a Case-Control Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Girdhar, Yashila] Thapar University, Department of Biotechnology, 147004 Patiala, Punjab, India. [Singh, Navneet] Post Graduate Institute of Medical Education & Research (PGIMER), Sector 14, Department of Pulmonary Medicine, Sector-12, 160012 Chandigarh, India. [Behera, Digambar] Post Graduate Institute of Medical Education & Research (PGIMER), Sector 14, Department of Pulmonary Medicine, Sector-12, 160012 Chandigarh, India. [Sharma, Siddharth] Thapar University, Department of Biotechnology, 147004 Patiala, Punjab, India. RP Sharma, S (reprint author), Thapar University, Department of Biotechnology, 147004 Patiala, India. EM siddharthsharma.phd@thapar.edu CR Shah PP, Singh AP, Singh M, Mathur N, Pant MC, Mishra BN, Parmar D, 2008, Interaction of cytochrome P4501A1 genotypes with other risk factors and susceptibility to lung cancer. Mutat Res 639:1–10 Jancova P, Anzenbacherb P, 2010, Anzenbacherova E, 2010, phase II drug metabolizing enzymes. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 154:103–116 Tefre T, Ryberg D, Haugen A, Nebert DW, Skaug V, Brogger A, Borresen AL, 1991, Human CYP1A1, cytochrome P 1450, gene: lack of association between the Msp I restriction fragment length polymorphism and incidence of lung cancer in a Norwegian population. Pharmacogenet Genomics 1:1 Altinisik J, Balta ZB, Aydin G, Ulutin T, Buyru N, 2010, Investigation of glutathione S-transferase M1 and T1 deletions in lung cancer. Mol Biol Rep 37:263–267 Lee KM, Kang D, Clapper ML, Sundberg MI, Kihara MO, Kiyohara C, Min S, Lan Q, Le Marchand L, Lin P, Lung ML, Pinarbasi H, Pisani P, Srivatanakul P, Seow A, Sugimura H, Tokudome S, Yokota J, Taioli E, 2008, CYP1A1, GSTM1, and GSTT1 polymorphisms, smoking, and lung cancer risk in a pooled analysis among Asian populations. Cancer Epidemiol Biomark Prev 17:5 Cascorbi I, Brockmoller J, Roots I, 1996, A C4887A polymorphism in exon 7 of human CYP1A1: population frequency, mutation linkages and impact on lung cancer susceptibility. Cancer Res 56:4965–4969 Shaffi SM, Shah AM, Bhat IA, Koul P, Ahmad SN, Siddiqi MA, 2009, CYP1A1 polymorphismand risk of lung cancer in the ethnic Kashmiri population. Asian Pacific J Cancer Prev 10:651–656 Kumar V, Singh S, Yadav CS, Ahmed RS, Gupta S, Pasha ST, Tripathi AK, Banerjee BD, 2009, CYP1A1 and CYP3A4 polymorphic variations in Delhi population of North India. Environ Toxicol Pharmacol 29:126–130 Shia X, Zhoua S,Wangb Z, Zhouc Z,Wanga Z, 2007, CYP1A1 and GSTM1 polymorphisms and lung cancer risk in Chinese populations: a meta-analysis. Lung Cancer 59, 155—163. Zhan P,Wang Q, Qian Q,Wei SZ,Yu LK, 2011, CYP1A1 MspI and exon7 gene polymorphisms and lung cancer risk: an updated metaanalysis and review. J Exp Clin Cancer Res 30:99 Sobti RC, Sharma S, Joshi A, Jindal SK, Janmeja A, 2004, Genetic polymorphism of the CYP1A1, CYP 2E1, GSTM1 & GSTT1 genes and lung cancer susceptibility in a north Indian population. Mol Cell Biochem 266:1–9 Liu X, Li Z, Zhang Z, ZhangW, LiW, Xiao Z, Liu H, Jiao H,Wang Y, Li G, 2014, Meta-analysis of GSTM1 null genotype and lung cancer risk in Asians.Med Sci Monit 20:1239–1245., DOI 10.12659/ MSM.890490 B’chir F, Aida T, Maurice JA, Saguem S, 2012, Glutathione Stransferase M1 and T1, CYP1A2-2467 T/delT polymorphisms and non-small cell lung cancer risk in Tunisian sample. Egypt J Med Hum Genet 13:307–312 Saarikoski ST, Voho A, Reinikainen M, Anttila S, Karjalainen A, Malaveille C, et al., 1998, Combined effect of polymorphic GST genes on individual susceptibility to lung cancer. Int J Cancer 77:516–521 Quinones L, Lucas D, Godoy J, Ca′ceres D, Berthou F, Varela N, Lee K, Acevedo C, Marti’nez L, Aguilera AM, Gil L, 2001, CYP1A1, CYP2E1 and GSTM1 genetic polymorphisms. The effect of single and combined genotypes on lung cancer susceptibility in Chilean people. Cancer Lett 174:35–44 Sreeja LK, Syamala V, Hariharan S, Madhavan J, Devan SC, Ankathil R, 2005, Possible risk modification by CYO1A1, GSTM1 and GSTT1 gene polymorphism in lung cancer susceptibility in a south Indian population. J Hum Genet 50:618–662 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 647 EP 652 DI 10.1007/s12253-016-0058-5 PG 6 ER PT J AU Choi, JE Kim, SM Yoo, JN Lee, HS AF Choi, Ji Eun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Inactivating Frameshift Mutation of INPP4B Encoding a PI3K Pathway Phosphatase in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Vanhaesebroeck B, Stephens L, Hawkins P, 2012, PIK3K signaling: the path to discovery and understanding. Nat Rev Mol Cell Biol 13: 195–203 Chen M, Nowak DG, Trotman LC, 2014, Molecular pathways: PI3K pathway phosphatases as biomarkers for cancer prognosis and therapy. Clin Cancer Res 20:3057–3063 Li Chew C, Lunardi A, Gulluni F, Ruan DT, Chen M, Salmena L, Nishino M, Papa A, Ng C, Fung J, Clohessy JG, Sasaki J, Sasaki T, Bronson RT, Hirsch E, Pandolfi PP, 2015, In vivo role of INPP4B in tumor and metastasis suppression through regulation of PI3K–AKT signaling at endosomes. Cancer Discov 5:740–751 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2016 VL 22 IS 3 BP 653 EP 654 DI 10.1007/s12253-016-0062-9 PG 2 ER PT J AU Davidson, TK Zhu, Z Fang, Y AF Davidson, T Kristoffer Zhu, Ziwen Fang, Yujiang TI Phytochemicals in the Fight Against Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Phytochemicals; Apple extract; Cancer ID Phytochemicals; Apple extract; Cancer AB Phytochemicals are chemical compounds from fruits, vegetables, or grains and they have been used to treat various diseases for thousands of years. More than one million people in the United States get cancer each year. Although recent advances in medicine have improved the outcomes for cancer patients, there is still a need for novel approaches in the fight against cancer. One such approach that has shown promise in recent years is the use of phytochemicals alone or as synergistic agents. In this review, we will discuss the use of phytochemicals as therapeutic agents against cancer with an emphasis on apple extract. C1 [Davidson, T Kristoffer] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. [Zhu, Ziwen] University of Missouri, School of Medicine, Department of Surgery, 65212 Columbia, MO, USA. [Fang, Yujiang] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. RP Fang, Y (reprint author), Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, USA. EM yujiang.fang@dmu.edu CR Siegel R, Ma J, Zou Z, Jemal A, 2014, Cancer statistics, 2014. CA Cancer J Clin 64(1):9–29 Wang H, Khor TO, Shu L, Su Z, Fuentes F, Lee J-H, Kong A-NT, 2012, Plants against cancer: A review on natural phytochemicals in preventing and treating cancers and their druggability. Anti Cancer Agents Med Chem 12(10):1281 Halberstein RA, 2005, Medicinal plants: historical and crosscultural usage patterns. Ann Epidemiol 15(9):686–699 Simon JJ, 2002, Phytochemicals & cancer. Journal of Chiropractic Medicine 1(3):91–96 Wahle KW, Brown I, Rotondo D, Heys SD, 2010, Plant phenolics in the prevention and treatment of cancer. Bio-Farms for Nutraceuticals. Springer, In, pp. 36–51 Johnson D, Walker C, 1999, Cyclins and cell cycle checkpoints. Annu Rev Pharmacol Toxicol 39(1):295–312 Boyer J, Liu RH, 2004, Apple phytochemicals and their health benefits. Nutr J 3(5):12 Koch TC, Briviba K, Watzl B, Fahndrich C, Bub A, Rechkemmer G, Barth SW, 2009, Prevention of colon carcinogenesis by apple juice in vivo: impact of juice constituents and obesity. Mol Nutr Food Res 53(10):1289–1302 Kahle K, KrausM, ScheppachW, Richling E, 2005, Colonic availability of apple polyphenols–a study in ileostomy subjects. Mol Nutr Food Res 49(12):1143–1150 Hollman PC, van Trijp JM, Buysman MN, van der Gaag MS, Mengelers MJ, de Vries JH, Katan MB, 1997, Relative bioavailability of the antioxidant flavonoid quercetin from various foods in man. FEBS Lett 418(1):152–156 Gerhauser C, 2008, Cancer chemopreventive potential of apples, apple juice, and apple components. Energy, Kcal/kJ, 54:227 De Mejia EG, Prisecaru VI, 2005, Lectins as bioactive plant proteins: a potential in cancer treatment. Crit Rev Food Sci Nutr 45(6): 425–445 HemaIswarya S, Doble M, 2006, Potential synergism of natural products in the treatment of cancer. Phytother Res 20(4):239–249 Kawasaki BT, Hurt EM, Mistree T, Farrar WL, 2008, Targeting cancer stem cells with phytochemicals. Mol Interv 8(4):174 Li Y, Niu Y, Sun Y,Mei L, Zhang B, Li Q, Liu L, Zhang R, Chen J, Mei Q, 2014, An Apple Oligogalactan Potentiates the growth inhibitory effect of celecoxib on colorectal cancer. Nutr Cancer 66(1): 29–37 Basnet P, Skalko-Basnet N, 2011, Curcumin: an anti-inflammatory molecule from a curry spice on the path to cancer treatment. Molecules 16(6):4567–4598 Moiseeva EP, Almeida GM, Jones GD, Manson MM, 2007, Extended treatment with physiologic concentrations of dietary phytochemicals results in altered gene expression, reduced growth, and apoptosis of cancer cells. Mol Cancer Ther 6(11):3071–3079 Weiss JF, Landauer MR, 2003, Protection against ionizing radiation by antioxidant nutrients and phytochemicals. Toxicology 189(1):1–20 Reagan-Shaw S, Eggert D, Mukhtar H, Ahmad N, 2010, Antiproliferative effects of apple peel extract against cancer cells. Nutr Cancer 62(4):517–524 McCann M, Gill C, O’Brien G, Rao J, McRoberts W, Hughes P, McEntee R, Rowland I, 2007, Anti-cancer properties of phenolics from apple waste on colon carcinogenesis in vitro. Food Chem Toxicol 45(7):1224–1230 Loo G, 2003, Redox-sensitive mechanisms of phytochemicalmediated inhibition of cancer cell proliferation, review). J Nutr Biochem 14(2):64–73 Kern M, Pahlke G, Balavenkatraman KK, Bohmer FD, Marko D, 2007, Apple polyphenols affect protein kinase C activity and the onset of apoptosis in human colon carcinoma cells. J Agric Food Chem 55(13):4999–5006 Li Q, Zhou S, Jing J, Yang T, Duan S,Wang Z,Mei Q, Liu L, 2013, Oligosaccharide from apple induces apoptosis and cell cycle arrest in HT29 human colon cancer cells. Int J Biol Macromol 57:245– 254 Zheng C-Q, Qiao B, Wang M, Tao Q, 2013, Mechanisms of apple polyphenols-induced proliferation inhibiting and apoptosis in a metastatic oral adenoid cystic carcinoma cell line. The Kaohsiung Journal of Medical Sciences 29(5):239–245 Sudan S, Rupasinghe HV, 2014, Flavonoid-enriched Apple Fraction AF4 induces cell cycle arrest, DNATopoisomerase II inhibition, and apoptosis in human liver cancer HepG2 cells. Nutr Cancer 66(7):1237–1246 Sun J, Liu RH, 2006, Cranberry phytochemical extracts induce cell cycle arrest and apoptosis in human MCF-7 breast cancer cells. Cancer Lett 241(1):124–134 Ray RB, RaychoudhuriA, Steele R, Nerurkar P, 2010, Bittermelon, Momordica charantia, extract inhibits breast cancer cell proliferation by modulating cell cycle regulatory genes and promotes apoptosis. Cancer Res 70(5):1925–1931 Yoon H, Liu RH, 2007, Effect of selected phytochemicals and apple extracts on NF-κB activation in human breast cancer MCF- 7 cells. J Agric Food Chem 55(8):3167–3173 Chen M, Chen D, Dou Q, 2004, Inhibition of proteasome activity by various fruits and vegetables is associated with cancer cell death. In vivo Athens, Greece 18(1):73 Ding M, Lu Y, Bowman L, Huang C, Leonard S, Wang L, Vallyathan V, Castranova V, Shi X, 2004, Inhibition of AP-1 and neoplastic transformation by fresh apple peel extract. J Biol Chem 279(11):10670–10676 Sliva D, 2003, Ganoderma lucidum, reishi, in cancer treatment. Integrative Cancer Therapies 2(4):358–364 Grube BJ, Eng ET, Kao Y-C, Kwon A, Chen S, 2001)White button mushroom phytochemicals inhibit aromatase activity and breast cancer cell proliferation. The Journal of Nutrition 131(12):3288– 3293 KernM, Tjaden Z, Ngiewih Y, Puppel N,Will F, Dietrich H, Pahlke G, Marko D, 2005, Inhibitors of the epidermal growth factor receptor in apple juice extract. Mol Nutr Food Res 49(4):317–328 Cochrane CB, Nair PR, Melnick SJ, Resek AP, Ramachandran C, 2008, Anticancer effects of Annona glabra plant extracts in human leukemia cell lines. Anticancer Res 28(2 A):965–971 Veeriah S, Kautenburger T, Habermann N, Sauer J, Dietrich H,Will F, Pool-Zobel BL, 2006, Apple flavonoids inhibit growth of HT29 human colon cancer cells and modulate expression of genes involved in the biotransformation of xenobiotics. Mol Carcinog 45(3):164–174 Fini L, Selgrad M, Fogliano V, Graziani G, Romano M, Hotchkiss E, Daoud YA, Edward B, Boland CR, Ricciardiello L, 2007, Annurca apple polyphenols have potent demethylating activity and can reactivate silenced tumor suppressor genes in colorectal cancer cells. The Journal of Nutrition 137(12):2622–2628 Barnes S, 2001, Role of phytochemicals in prevention and treatment of prostate cancer. Epidemiol Rev 23(1):102–105 Sagar SM, Yance D, Wong R, 2006, Natural health products that inhibit angiogenesis: a potential source for investigational new agents to treat cancer—part 1. Curr Oncol 13(1):14 Weng C-J, Yen G-C, 2012, Chemopreventive effects of dietary phytochemicals against cancer invasion and metastasis: phenolic acids, monophenol, polyphenol, and their derivatives. Cancer Treat Rev 38(1):76–87 Shu L, Cheung K-L, Khor TO, Chen C, Kong A-N, 2010, Phytochemicals: cancer chemoprevention and suppression of tumor onset and metastasis. Cancer Metastasis Rev 29(3):483–502 Liu RH, 2004, Potential synergy of phytochemicals in cancer prevention: mechanism of action. The Journal of Nutrition 134(12): 3479S–3485S Russo GL, 2007, Ins and outs of dietary phytochemicals in cancer chemoprevention. Biochem Pharmacol 74(4):533–544 Fini L, Piazzi G, Daoud Y, Selgrad M, Maegawa S, Garcia M, Fogliano V, Romano M, Graziani G, Vitaglione P, 2011, Chemoprevention of intestinal polyps in ApcMin/+ mice fed with western or balanced diets by drinking annurca apple polyphenol extract. Cancer Prevention Research 4(6):907–915 Wolfe K,Wu X, Liu RH, 2003, Antioxidant activity of apple peels. J Agric Food Chem 51(3):609–614 Cline JM, Hughes CL Jr, 1998, Phytochemicals for the prevention of breast and endometrial cancer. Biological and hormonal therapies of cancer. Springer, In, pp. 107–134 Zessner H, Pan L, Will F, Klimo K, Knauft J, Niewohner R, Hummer W, Owen R, Richling E, Frank N, 2008, Fractionation of polyphenol-enriched apple juice extracts to identify constituents with cancer chemopreventive potential.Mol Nutr Food Res 52(S1): S28–S44 Telang NT, Katdare M, Bradlow HL, Osborne MP, Fishman J, 1997, Inhibition of proliferation and modulation of estradiol metabolism: novel mechanisms for breast cancer prevention by the phytochemical indole-3-carbinol. Experimental Biology and Medicine 216(2):246–252 He X, Liu RH, 2008, Phytochemicals of apple peels: isolation, structure elucidation, and their antiproliferative and antioxidant activities. J Agric Food Chem 56(21):9905–9910 Ribeiro FAP, de Moura CFG, Gollucke APB, Ferreira MS, Catharino RR, Aguiar O, Spadari RC, Barbisan LF, Ribeiro DA, 2014, Chemopreventive activity of apple extract following medium-term oral carcinogenesis assay induced by 4- nitroquinoline-1-oxide. Arch Oral Biol 59(8):815–821 Gallus S, Talamini R, Giacosa A, Montella M, Ramazzotti V, Franceschi S, Negri E, La Vecchia C, 2005, Does an apple a day keep the oncologist away? Ann Oncol 16(11):1841–1844 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 655 EP 660 DI 10.1007/s12253-016-0045-x PG 6 ER PT J AU Benharroch, D Ariad, S Tadmor, N Nalbandyan, K Lazarev, I AF Benharroch, Daniel Ariad, Samuel Tadmor, Noa Nalbandyan, Karen Lazarev, Irena TI Relevance of the Measles Virus Expression in Cancer - an Update SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Measles virus; Hodgkin lymphoma; Lung cancer; Breast cancer; Atypical measles syndrome ID Measles virus; Hodgkin lymphoma; Lung cancer; Breast cancer; Atypical measles syndrome AB Evidence of an association between classical Hodgkin lymphoma and the measles virus has previously been presented by our group. Arguments held against our thesis were reevaluated. Substantiation of a relationship between the measles virus and additional solid tumors was submitted. Moreover, a pathogenic pathway was suggested to support a possible contribution of the measles virus to the development of classical Hodgkin lymphoma. We have chosen to exclude a discussion of measles virotherapy, since this carries distinct implications. We now add new evidence regarding the expression of the measles virus phosphoprotein in a few cancers. We also suggest a role in this context for atypical measles syndrome in malignant tumors. Last, we propose a collaboration which may make the best, on the one hand of our cohort of classical Hodgkin lymphoma, half of which carry the measles virus expression in their tumor cells. The planned study will also look into the patients vaccination records and into a previous history of the measles disease. On the other hand, cohorts of patients diagnosed with late onset measles will be assessed for the eventual diagnosis of atypical measles syndrome and will be followed up for the subsequent development of a malignant tumor. C1 [Benharroch, Daniel] Ben-Gurion University of the Negev, Soroka University Medical Center and Faculty of Health Sciences, Departments of Pathology, 1, Itzhak Rager Boulevard, 84101 Beer-Sheva, Israel. [Ariad, Samuel] Ben-Gurion University of the Negev, Soroka University Medical Center and Faculty of Health Sciences, Departments of OncologyBeer-Sheva, Israel. [Tadmor, Noa] Ben-Gurion University of the Negev, National Institute for Biotechnology in the Negev, Department of Life SciencesBeer-Sheva, Israel. [Nalbandyan, Karen] Ben-Gurion University of the Negev, Soroka University Medical Center and Faculty of Health Sciences, Departments of Pathology, 1, Itzhak Rager Boulevard, 84101 Beer-Sheva, Israel. [Lazarev, Irena] Ben-Gurion University of the Negev, Soroka University Medical Center and Faculty of Health Sciences, Departments of OncologyBeer-Sheva, Israel. RP Benharroch, D (reprint author), Ben-Gurion University of the Negev, Soroka University Medical Center and Faculty of Health Sciences, Departments of Pathology, 84101 Beer-Sheva, Israel. EM dbenharroch@yahoo.com CR Benharroch D, Shemer Avni Y, Myint Y-Y, et al., 2004, Measles virus: evidence of an association with Hodgkin's disease. Brit J Cancer 91:572–579 Maggio E, Benharroch D, Gopas J, Dittmer U, Hansmann M-L, Kuppers R, 2007, Absence of measles virus genome and transcripts in Hodgkin-reed/Sternberg cells of a cohort of Hodgkin lymphoma patients. Int J Cancer 121:448–453 Wilson KS, Freeland JML, Gallagher A, et al., 2007)Measles virus and classical Hodgkin lymphoma: no evidence for a direct association. Int J Cancer 121:442–447 Benharroch D, Klinkovich I, Piura B, Shaco-Levy R, Gopas J, 2009, Evidence of measles virus antigens and RNA in endometrial cancer. Eur J Obstet Gynecol Reprod Biol 147:206–209 Sion-Vardy N, Lasarev I,Delgado B,Gopas J, Benharroch D, Ariad S, 2009, Measles virus: evidence for association with lung cancer. Exp Lung Res 35:701–712 Ariad S, Milk N, Bolotin A, Gopas J, Sion-Vardy N, Benharoch D, 2011, Measles virus antigens in breast cancer. Anticancer Res 31: 913–920 Benharroch D, Einav I, Feldman A, Levy A, Ariad S, Gopas J, 2010, Apoptosis of Hodgkin-reed-Sternberg cells in classical Hodgkin lymphoma revisited. APMIS 118:339–345 Karlin D, Belshaw R, 2012, Detecting remote sequence homology in disordered proteins: discovery of conserved motifs in the Ntermini of Mononrgavirales phosphoproteins. PLoS One 7:e31719 Kruzniar A, van Ham RC, Pongor S, Leunissen JA, 2008, The quest for orthologs: finding the corresponding gene across genomes. Trends Genet 24:539–551 Thorley-Lawson DA, Gross A, 2004, Mechanism of disease: persistence of Epstein–Barr virus and the origin of the associated lymphomas. N Engl J Med 350:1328–1337 Peek RM, Crabtree JE, 2006, Helicobacter infection and gastric neoplasia. J Pathol 208:233–248 Tlsty TD, Coussens LM, 2006, Tumor stroma and regulation of cancer development. Annu Rev Pathol 1:119–150 Benharroch D, Shemer-Avni LA, Myint Y-Y, Ariad S, Rager B, Sacks M, Gopas J, 2003, New candidate virus in association with Hodgkin's disease. Leukemia & Lymphoma 44:605–610 Knecht H, Berger C, Rothenberg S, Odermatt BF, Brousset P, 2001, The role of Epstein–Barr virus in neoplastic transformation. Oncology 60:289–302 Niedobitek G, Meru DHJ, 2001, Epstein–Barr virus infection and human malignancies. Int J Exp Pathol 82:149–170 Jarrett RF, Armstrong AA, Alexander E, 1996, Epidemiology of EBVand Hodgkin lymphoma. Ann Oncol 7(suppl):5–10 Chang KL, Albujar PF, Chen YY, Johnson RM, Weiss LM, 1993, High prevalence of Epstein–Barr virus in reed-Sternberg cells of Hodgkin disease occurring in Peru. Blood 81:496–501 Bargou RC, Emmerich F, Krappmann D, et al., 1997, Constitutive nuclear factor-kappaB-RelA activation is required for proliferation and survival of Hodgkin's disease tumor cells. J Clin Invest 100: 2961–2969 Schuhmann KM, Pfaller CK, Conzelmann KK, 2011, The measles virus V protein binds to p65, RelA, to suppress NF-KappaB activity. J Virol 85:3162–3171 Rodig SJ, Savage KJ, Nguyen V, Pinkus GS, Shipp MA, Aster JC, Kutok JL, 2005, TRAF1 expression and c-Rel activation are useful adjuncts in distinguishing classical Hodgkin lymphoma from a subset of morphologically or immunophenotypically similar lymphomas. Am J Surg Pathol 29:196–203 Izban KF, Ergin M, Huang Q, et al., 2001, Characterization of NFkappaB expression in Hodgkin disease. Mod Pathol 14:297–310 MacMahon B, 1966, Epidemiology of Hodgkin's disease. Cancer Res 26:1189–1201 Gutensohn N, Cole P, 1981, Childhood social environment and Hodgkin's disease. N Engl J Med 304:135–140 Glaser SL, 1990, Spacial clustering of Hodgkin's disease in the San Francisco Bay area. Am J Epidemiol 132(1 Suppl):s167–s177 Westergaard T, Melbye M, Pedersen JB, Frish M, Olsen JH, Andersen PK, 1997, Birth order, sibship size and the risk of Hodgkin's disease in children and young adults: a population based study of 31 million person-years. Int J Cancer 72:977–981 Sleckman BG, Mauch PM, Ambinder RF, et al., 1998, Epstein– Barr virus in Hodgkin's disease: correlation of risk factors and disease characteristics with molecular evidence of viral infection. Cancer Epidemiol Biomark Prev 7:1117–1121 Glaser SL, Clarke CA, Nugent RA, Stearns CB, Dorfman RF, 2002, Social class and risk of Hodgkin's disease in young adult women in 1988-94. Int J Cancer 98:110–117 Urayama KY, Jarrett RF, Hjalgrim H, et al., 2012, Genome-wide study of classical Hodgkin lymphoma and Epstein–Barr virus status-defined subgroups. J Natl Cancer Inst 104:240–253 Veit BC, Schyldower M, McIntyre S, Simmons D, Lampe RM, Fearnow RG, Stewart J, 1991, Serological response to measles vaccination in a highly immunized military dependent adolescent population. J Adolesc Health 12:273–278 Desai VK, Kapadia SJ, Kumar P, Nirupam S, 2002, Impact assessment of mass measles vaccination. Ind J Pediat 69:1037–1040 Dickinson HO, Nyari TA, Parker L, 2002, Childhood solid tumors in relation to infections in the community in Cumbria during pregnancy and around the time of birth. Br J Cancer 87:746–750 Tyari TA, Dikinson HO, Parker L, 2003, Childhood cancer in relation to infections in the community during pregnancy and around the time of birth. Int J Cancer 104:772–777 Zygiert Z, 1971, Hodgkin's disease: remission after measles. Lancet 1(7699):593 Mota HC, 1973, Infantile Hodgkin's disease: remission after measles. Br Med J 2:241 Zwitter M, 1984, Hodgkin's disease: therapeutic role of measles vaccine. Am J Med 77:A49 A52, A64 Schattner A, 1984, Therapeutic role of measles vaccine in Hodgkin's disease. Lancet 1(8369):171 Cozen W, Hamilton AS, Zhao P, et al., 2009, A protective role for early oral exposures in the etiology of young adult Hodgkin lymphoma. Blood 114:4014–4020 Stals A, Mathijs E, Baert L, et al., 2012, Molecular detection and genotyping of noroviruses. Food Environ Virol 4:153–167 Bar M, Bar D, Lehman B, 2009, Selection and validation of candidate housaekeeping genes for studies of human keratinocytes - review and recommendations. J Invest Dermatol 129:535–537 Ariad S, Lipshitz I, Benharroch D, Gopas J, Barchana M, 2009, A sharp rise in the incidence of Hodgkin's lymphoma in young adults in Israel. Isr Med Assoc J 11:453–455 Chen YT, Zheng T, Chou MC, Boyle P, Holford TR, 1997, The increase of Hodgkin's disease incidence among young adults. Experience in Connecticut, 1935-1992. Cancer 79:2209–2218 Liu S, Semenciw R,Waters C,Wen SW, Mao Y, 2000, Time trends and sex patterns in Hodgkin's disease incidence in Canada, 1970- 1995. Can J Public Health 91:188–192 Chen M, Cortay J-C, Logan IR, SapountziV, Robson CN, Gerlier D, 2005, Inhibition of ubiqitination and stabilization of human ubiquitin E3 ligase PIRH2 by measles virus phosphoprotein. J Virol 79: 11824–11836 Duan W, Gao L, Druhon LJ, Zhu WG, Morrison C, Otterson GA, Villalona-Calero MA, 2004, Expression of Pirh2, a newly identified ubiquitin protein ligase in lung cancer. J Natl Cancer Inst 96: 1718–1721 Corcoran CA, Huang Y, Sheikh MS, 2004, The p53 paddy wagon: COP1, Pirh2 and MDM2 are found resisting apoptosis and growth arrest. Cancer Biol Ther 3:721–725 Nihie T, Nagata K, Takeuchi K, 2007, The C protein of wild-type measles virus has the ability to shuttle between the nucleus and the cytoplasm. Microbes Infect 9:344–354 Griffin DE, Pan CH, 2009, Inactivated vaccines. In: Griffin DE, Oldstone MBA, eds, Measles, pathogenesis and control. Springer, Berlin, pp. 194–195 Polack FP, Auwaerter PG, Lee SH, et al., 1999, Production of atypicalmeasles in rhesusmacaques: evidence for diseasemediated by immune complex formation and eosinophils in the presence of fusion-inhibiting antibody. Nat Med 5:629–634 Polack FP, Hoffman SJ, Crujeiras G, Griffin DE, 2003, A role for nonprotective complement fixing antibodies with low avidity for measles virus in atypical measles. Nat Med 9:1209–1213 Hall WJ, Hall CB, 1979, Atypical measles in adolescents: evaluation of clinical and pulmonary function. Ann Intern Med 90:882– 886 Annunziato D, Kaplan MH, HallWW, Ichinose H, Lin JH, Balsam D, Paladino VS, 1982, Atypical measles syndrome: pathologic and serologic findings. Pediatrics 70:203–209 Frey HM, Krugman S, 1981, Atypical measles syndrome: unusual hepatic, pulmonary and immunologic aspects. Am J Med Sci 281: 51–55 Melenotte C, Cassir N, Tessonnier L, Brouqui P, 2015, Atypical measles syndrome in adults: still around. BMJ Case Rep Sept 23: 2015., DOI 10.1136/bcr-2015-211054 Sachan D, 2013, Atypical sclerosing panencephalitis with short onset latency. Indian Pediatr 50:969 Malik B, Sharma FJ, Bhardwaj AK, Sharma A, 2012, Subacute sclerosing panencephalitis despite adequate vaccination. Australas Med J 5:359–361 Fabian VA, Lee HY, Keith-Rokosh JL, de Souza JL, Stewart- Wynne E, 2010, A 22-year-old Australian woman with atypical subacute sclerosing panencephalitis diagnosed at postmortem. J Clin Neurosci 17:1192–1194 Giladi M, Shulman A, Kedem R, Danon YL, 1987, Measles in adults: a prospective study of 291 consecutive cases. BMJ 295: 1314 Levine H, Zarka S, Ankol OE, Rozhavski V, Davidovitch N, Aboudy Y, Balicer RD, 2015, Seroprevalence of measles, mumps and rubella among young adults, after 20 years of universal twodose MMR vaccination in Israel. HumVaccin Immunother 11: 1400–1405 [No authors listed], 2015, Progress toward regional measles elimination, worldwide, 2000–2014. Wkly Epidemiol Rec 90:623–31. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 661 EP 666 DI 10.1007/s12253-016-0080-7 PG 6 ER PT J AU Yahyapour, Y Sadeghi, F Alizadeh, A Rajabnia, R Siadati, S AF Yahyapour, Yousef Sadeghi, Farzin Alizadeh, Ahad Rajabnia, Ramazan Siadati, Sepideh TI Detection of Merkel Cell Polyomavirus and Human Papillomavirus in Esophageal Squamous Cell Carcinomas and Non-Cancerous Esophageal Samples in Northern Iran SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal squamous cell carcinoma; Merkel cell polyomavirus; Human papillomavirus; Oncogenic viruses ID Esophageal squamous cell carcinoma; Merkel cell polyomavirus; Human papillomavirus; Oncogenic viruses AB Human papillomavirus (HPV) infection is one of the hypothesized causes of esophageal squamous cell carcinoma (ESCC), but the etiological association remains uncertain. It was postulated that other infectious agents together with HPV may increase the risk of ESCC. The current investigation aimed to explore the presence of a new human tumor virus, Merkel cell polyomavirus (MCPyV), together with HPV in ESCC tumors and non-cancerous esophageal samples in northern Iran. In total, 96 esophageal samples (51 with ESCC, and 45 without esophageal malignancy) were examined. HPV DNA was detected in esophageal specimens of 16 out of the 51 ESCC cases (31.4 %) and 20 out of the 45 noncancerous samples (44.4 %). Untypable HPV genotypes were recognized in high rates in cancerous (75.0 %) and noncancerous (55.0 %) esophageal specimens. MCPyV DNA was detected in esophageal specimens of 23 out of the 51 ESCC cases (45.1 %) and 16 out of the 45 non-cancerous samples (35.6 %). The mean MCPyV DNA copy number was 1.0 × 10−5 ± 2.4 × 10−5 and 6.0 × 10−6 ± 1.3 × 10−5 per cell in ESCC cases and non-cancerous samples, respectively. There was no statistically significant difference between cancerous and non-cancerous samples regarding mean MCPyV DNA load (P = 0.353). A bayesian logistic regression model adjusted to the location of esophageal specimen and MCPyV infection, revealed a significant association between HPV and odds of ESCC (OR, 2.45; 95 % CI: 1.01–6.16). This study provides the evidence of the detection of the MCPyV DNA at a low viral copy number in cancerous and non- cancerous esophageal samples. C1 [Yahyapour, Yousef] Babol University of Medical Sciences, Infectious Diseases & Tropical Medicine Research CenterBabol, Iran. [Sadeghi, Farzin] Babol University of Medical Sciences, Health Research Institute, Cellular and Molecular Biology Research CenterBabol, Iran. [Alizadeh, Ahad] Royan Institute for Reproductive Biomedicine, ACECR, Reproductive Epidemiology Research Center, Department of Epidemiology and Reproductive HealthTehran, Iran. [Rajabnia, Ramazan] Babol University of Medical Sciences, Infectious Diseases & Tropical Medicine Research CenterBabol, Iran. [Siadati, Sepideh] Babol University of Medical Sciences, Department of PathologyBabol, Iran. RP Sadeghi, F (reprint author), Babol University of Medical Sciences, Health Research Institute, Cellular and Molecular Biology Research Center, Babol, Iran. EM sadeghifarzin6@gmail.com CR Lambert R, Hainaut P, 2007, Esophageal cancer: cases and causes, part I). Endoscopy 39(6):550–555 Holmes RS, Vaughan TL, 2007, Epidemiology and pathogenesis of esophageal cancer. Semin Radiat Oncol 17(1):2–9 WeiWQet al., 2004, Prospective study of serum selenium concentrations and esophageal and gastric cardia cancer, heart disease, stroke, and total death. Am J Clin Nutr 79(1):80–85 Al-Haddad S et al., 2014, Infection and esophageal cancer. Ann N YAcad Sci 1325:187–196 Guo F et al., 2012, Human papillomavirus infection and esophageal squamous cell carcinoma: a case-control study. Cancer Epidemiol Biomarkers Prev 21(5):780–785 Yahyapour Y et al., 2013, High-risk and low-risk human papillomavirus in esophageal squamous cell carcinoma at Mazandaran, Northern Iran. Pathol Oncol Res 19(3):385–391 Cao B et al., 2005, LMP7/TAP2 gene polymorphisms and HPV infection in esophageal carcinoma patients from a high incidence area in China. Carcinogenesis 26(7):1280–1284 Moens U, Van Ghelue M, Ehlers B, 2014, Are human polyomaviruses co-factors for cancers induced by other oncoviruses? Rev Med Virol 24(5):343–360 Salehi-Vaziri M et al., 2015, Merkel cell polyomavirus and human papillomavirus infections in cervical disease in Iranian women. Arch Virol 160(5):1181–1187 Feng H et al., 2008, Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science 319(5866):1096–1100 Tachibana T, 1995, The Merkel cell: recent findings and unresolved problems. Arch Histol Cytol 58(4):379–396 Harmse JL et al., 1999, Merkel cells in the human oesophagus. J Pathol 189(2):176–179 Wieland U et al., 2009, Merkel cell polyomavirus DNA in persons without merkel cell carcinoma. Emerg Infect Dis 15(9):1496–1498 Loyo M et al., 2010, Quantitative detection of Merkel cell virus in human tissues and possible mode of transmission. Int J Cancer 126(12):2991–2996 Iaconelli M et al., 2015, First detection of human papillomaviruses and human polyomaviruses in river waters in Italy. Food Environ Virol 7(4):309–315 Yahyapour Y et al., 2012, Evaluation of human papilloma virus infection in patients with esophageal squamous cell carcinoma from the Caspian Sea area, north of Iran. Asian Pac J Cancer Prev 13(4): 1261–1266 Sadeghi F et al., 2015, Detection of Merkel cell polyomavirus large T-antigen sequences in human central nervous system tumors. J Med Virol 87(7):1241–1247 Sadeghi F. et al. Prevalence of JC polyomavirus large T antigen sequences among Iranian patients with central nervous system tumors. Arch Virol; 160(1): 61–68 Becker JC et al., 2009, MC polyomavirus is frequently present in Merkel cell carcinoma of European patients. J Invest Dermatol 129(1):248–250 Imajoh M et al., 2012, Detection of Merkel cell polyomavirus in cervical squamous cell carcinomas and adenocarcinomas from Japanese patients. Virol J 9:154 R Development Core Team, 2005, R: a language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. http://www.R-project.org Parkin DMet al., 2005, Global cancer statistics, 2002. CA Cancer J Clin 55(2):74–108 Blot WJ, McLaughlin JK, 1999, The changing epidemiology of esophageal cancer. Semin Oncol 26(5 Suppl 15):2–8 Matsha T et al., 2007, Expression of p53 and its homolog, p73, in HPV DNA positive oesophageal squamous cell carcinomas. Virology 369(1):182–190 Li X et al., 2014, Systematic review with meta-analysis: the association between human papillomavirus infection and oesophageal cancer. Aliment Pharmacol Ther 39(3):270–281 Zhang SK et al., 2015, The association between human papillomavirus 16 and esophageal cancer in Chinese population: a meta-analysis. BMC Cancer 15:1096 Sichero L et al., 2013, Broad HPV distribution in the genital region of men from the HPV infection in men, HIM, study. Virology 443(2):214–217 Spinillo A et al., 2014, Untypable human papillomavirus infection and risk of cervical intraepithelial neoplasia among women with abnormal cervical cytology. J Med Virol 86(7):1145–1152 McNees AL et al., 2005, Specific and quantitative detection of human polyomaviruses BKV, JCV, and SV40 by real time PCR. J Clin Virol 34(1):52–62 Moore PS, Chang Y, 2010, Why do viruses cause cancer? Highlights of the first century of human tumour virology. Nat Rev Cancer 10(12):878–889 Shuda M et al., 2008, T antigen mutations are a human tumorspecific signature for Merkel cell polyomavirus. Proc Natl Acad Sci U S A 105(42):16272–16277 Houben R et al., 2010, Merkel cell polyomavirus-infected Merkel cell carcinoma cells require expression of viral T antigens. J Virol 84(14):7064–7072 Sihto H et al., 2011, Merkel cell polyomavirus infection, large T antigen, retinoblastoma protein and outcome in Merkel cell carcinoma. Clin Cancer Res 17(14):4806–4813 Goh S et al., 2009, Merkel cell polyomavirus in respiratory tract secretions. Emerg Infect Dis 15(3):489–491 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 667 EP 672 DI 10.1007/s12253-016-0048-7 PG 6 ER PT J AU Patocs, A Lendvai, KN Butz, H Liko, I Sapi, Z Szucs, N Toth, G Grolmusz, KV Igaz, P Toth, M Racz, K AF Patocs, Attila Lendvai, K Nikoletta Butz, Henriett Liko, Istvan Sapi, Zoltan Szucs, Nikolette Toth, Geza Grolmusz, K Vince Igaz, Peter Toth, Miklos Racz, Karoly TI Novel SDHB and TMEM127 Mutations in Patients with Pheochromocytoma/Paraganglioma Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pheochromocytoma; Paraganglioma; Germline mutation; Genotype-phenotype ID Pheochromocytoma; Paraganglioma; Germline mutation; Genotype-phenotype AB Pheochromocytomas (Pheo) and paragangliomas (PGL) are rare tumors,with heterogeneous genetic background. In up to 30 % of all, apparently sporadic Pheo/PGL cases germline mutations can be identified in one of the 15 genes representing genetic susceptibility for Pheo/PGL. Malignancy is rare but it frequently associates with SDHB mutations. Our aim was to determine the prevalence of germline SDHx, SDHAF2, MAX and TMEM127 mutations in Hungarian patients with apparently sporadic Pheo/PGLs. Mutation screening of the SDHx, SDHAF2, MAX and TMEM127 genes was performed in 82 Hungarian patients with apparently sporadic Pheo/PGL using PCR and bidirectional Sanger sequencing. Disease-causing germline mutations were identified in 11 patients, of which 4 SDHB and 2 TMEM127 mutations were novel. Earlier development of Pheo/PGL, more malignant phenotype and multiple tumors were observed in genetically positive cases especially in those with SDHB mutations. The presence of bilateral or multiple tumors was the most predictive for identification of a pathogenic mutation. Together with cases harboring germline RET, VHL and NF1 mutations, Hungarian patients with Pheo/PGL exhibit a heterogeneousmutation spectrum, indicating that all of the Pheo/PGL susceptibility genes should be tested. Novel genotype-phenotype associations revealed by our study may contribute to improvement of diagnostic approaches and may help to achieve a better clinical follow up for patients with Pheo/PGL. C1 [Patocs, Attila] Hungarian Academy of SciencesBudapest, Hungary. [Lendvai, K Nikoletta] Hungarian Academy of SciencesBudapest, Hungary. [Butz, Henriett] Hungarian Academy of SciencesBudapest, Hungary. [Liko, Istvan] Hungarian Academy of SciencesBudapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Szucs, Nikolette] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Toth, Geza] Ferenc Markhot County HospitalEger, Hungary. [Grolmusz, K Vince] Hungarian Academy of SciencesBudapest, Hungary. [Igaz, Peter] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Toth, Miklos] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Racz, Karoly] Bionics Innovation CenterBudapest, Hungary. RP Patocs, A (reprint author), Hungarian Academy of Sciences, Budapest, Hungary. EM patocs.attila@med.semmelweis-univ.hu CR Toledo RA, Dahia PL, 2015, Next-generation sequencing for the diagnosis of hereditary pheochromocytoma and paraganglioma syndromes. Curr Opin Endocrinol Diabetes Obes 22(3):169–179 Neumann HP, Bausch B, McWhinney SR, Bender BU, Gimm O, Franke G, Schipper J, Klisch J, Altehoefer C, Zerres K, Januszewicz A, Eng C, Smith WM, Munk R, Manz T, Glaesker S, Apel TW, Treier M, Reineke M, Walz MK, Hoang-Vu C, Brauckhoff M, Klein-Franke A, Klose P, Schmidt H, Maier- Woelfle M, Peczkowska M, Szmigielski C, Eng C, 2002, Freiburg-Warsaw-Columbus Pheochromocytoma Study Group germ-line mutations in nonsyndromic pheochromocytoma. N Engl J Med 346:1459–1466 Baysal BE, Ferrell RE, Willett-Brozick JE, Lawrence EC, Myssiorek D, Bosch A, van der Mey A, Taschner PE, Rubinstein WS, Myers EN, Richard CW 3rd, Cornelisse CJ, Devilee P, Devlin B, 2000, Mutations in SDHD, a Mitochondrial Complex II Gene, in Hereditary Paraganglioma Science 287:848 Niemann S, Muller U, 2000, Mutations in SDHC cause autosomal dominant paraganglioma, type 3. Nat Genet 26(3):268–270 Astuti D, Latif F, Dallol A, Dahia PL, Douglas F, George E, Skoldberg F, Husebye ES, Eng C, Maher ER, 2001, Gene mutations in the succinate dehydrogenase subunit SDHB cause susceptibility to familial pheochromocytoma and to familial paraganglioma. Am J Hum Genet 69(1):49–54 Hao HX, Khalimonchuk O, Schraders M, Dephoure N, Bayley JP, Kunst H, Devilee P, Cremers CW, Schiffman JD, Bentz BG, Gygi SP, Winge DR, Kremer H, Rutter J, 2009, SDH5, a gene required for flavination of succinate dehydrogenase, is mutated in paraganglioma. Science 28;:1139–42 Burnichon N, Briere JJ, Libe R, Vescovo L, Riviere J, Tissier F, Jouanno E, Jeunemaitre X, Benit P, Tzagoloff A, Rustin P, Bertherat J, Favier J, Gimenez-roqueplo AP, 2010, SDHA Is a Tumor Suppressor Gene Causing Paraganglioma. Hum Mol Genet 19: 3011–3020 Yeh IT, Lenci RE, Qin Y, Buddavarapu K, Ligon AH, Leteurtre E, Do Cao C, Cardot-Bauters C, Pigny P, Dahia PL, 2008, A germline mutation of the KIF1B beta gene on 1p36 in a family with neural and nonneural tumors. Hum Genet 124(3):279–285 Ladroue C, Carcenac R, LeporrierM, Gad S, Le Hello C, Galateau- Salle F, Feunteun J, Pouyssegur J, Richard S, Gardie B, 2008, PHD2 mutation and congenital erythrocytosis with paraganglioma. N Engl J Med 359(25):2685–2692 Qin Y, Yao L, King EE, Buddavarapu K, Lenci RE, Chocron ES, Lechleiter JD, SassM, Aronin N, Schiavi F, Boaretto F, Opocher G, Toledo RA, Toledo SP, Stiles C, Aguiar RC, Dahia PL, 2010, Germline mutations in TMEM127 confer susceptibility to pheochromocytoma. Nat Genet 42(3):229–233 Yao L, Schiavi F, Cascon A, Qin Y, Inglada-Perez L, King EE, Toledo RA, Ercolino T, Rapizzi E, Ricketts CJ, Mori L, Giacche M, Mendola A, Taschin E, Boaretto F, Loli P, Iacobone M, Rossi GP, Biondi B, Lima-Junior JV, Kater CE, Bex M, Vikkula M, Grossman AB, Gruber SB, Barontini M, Persu A, Castellano M, Toledo SP, Maher ER, Mannelli M, Opocher G, Robledo M, Dahia PL, 2010, Spectrum and prevalence of FP/TMEM127 gene mutations in pheochromocytomas and paragangliomas. JAMA 304: 2611–2619 Comino-Mendez I, Gracia-Aznarez FJ, Schiavi F, Landa I, Leandro-Garcia LJ, Leton R, Honrado E, Ramos-Medina R, Caronia D, Pita G, Gomez-Grana A, de Cubas AA, Inglada-Perez L, Maliszewska A, Taschin E, Bobisse S, Pica G, Loli P, Hernandez-Lavado R, Diaz JA, Gomez-Morales M, Gonzalez- Neira A, Roncador G, Rodriguez-Antona C, Benitez J, Mannelli M, Opocher G, Robledo M, Cascon A, 2011, Exome sequencing identifies MAX mutations as a cause of hereditary pheochromocytoma. Nat Genet 43(7):663–667 Castro-Vega LJ, Buffet A, De Cubas AA, Cascon A, Menara M, Khalifa E, Amar L, Azriel S, Bourdeau I, Chabre O, Curras-Freixes M, Franco-Vidal V, Guillaud-Bataille M, Simian C,Morin A, Leton R, Gomez-Grana A, Pollard PJ, Rustin P, Robledo M, Favier J, Gimenez-Roqueplo AP, 2014, Germline mutations in FH confer predisposition to malignant pheochromocytomas and paragangliomas. Hum Mol Genet 23(9):2440–2446 Cascon A, Comino-Mendez I, Curras-Freixes M, Inglada-Perez L, de Cubas AA, Curras-Freixes M, Tysoe C, Izatt L, Leton R, Gomez-Grana A, Mancikova V, Apellaniz-Ruiz M, Mannelli M, Schiavi F, Favier J, Gimenez-Roqueplo AP, Timmers HJ, Roncador G, Garcia JF, Rodriguez-Antona C, Robledo M, Cascon A, 2015, Whole-exome sequencing identifies MDH2 as a new familial paraganglioma gene. J Natl Cancer Inst 11:107(5)., DOI 10.1093/jnci/djv053 Cao M, Sun F, Huang X, Dai J, Cui B, Ning G, 2013, Analysis of the inhe r itance p a t tern of a Chinese family wit h phaeochromocytomas through whole exome sequencing. Gene 526(2):164–169 McInerney-Leo AM, Marshall MS, Benn DE,McFarlane J, Robinson BG, Brown MA, Leo PJ, Clifton-Bligh RJ, Duncan EL, 2014)Whole exome sequencing is an efficient and sensitive method for detection of germline mutations in patients with phaeochromcytomas and paragangliomas. Clin Endocrinol 80(1):25–33 Crona J, Verdugo AD, Granberg D,Welin S, Stalberg P, Hellman P, Bjorklund P, 2013, Next-generation sequencing in the clinical genetic screening of patients with pheochromocytoma and paraganglioma. Endocr Connect 2(2):104–111 Welander J, Andreasson A, Juhlin CC,Wiseman RW, Backdahl M, Hoog A, Larsson C, Gimm O, Soderkvist P, 2014, Rare germline mutations identified by targeted next-generation sequencing of susceptibility genes in pheochromocytoma and paraganglioma. J Clin Endocrinol Metab 99(7):E1352–E1360 Fishbein L, Khare S, Wubbenhorst B, DeSloover D, D'Andrea K, Merrill S, Cho NW, Greenberg RA, Else T, Montone K, LiVolsi V, Fraker D, Daber R, Cohen DL, Nathanson KL, 2015, Wholeexome sequencing identifies somaticATRXmutations in pheochromocytomas and paragangliomas. Nat Commun 6:6140 Lenders JW, Duh QY, Eisenhofer G, Gimenez-Roqueplo AP, Grebe SK, Murad MH, Naruse M, Pacak K, Young WF Jr; Endocrine Society, 2014, Pheochromocytoma and paraganglioma: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 99(6):1915–1942 Ricketts C,Woodward ER, Killick P, Morris MR, Astuti D, Latif F, Maher ER, 2008, Germline SDHB mutations and familial renal cell carcinoma. J Natl Cancer Inst 100(17):1260–1262 Patocs A, Karadi E, Toth M, Varga I, Szucs N, Balogh K,Majnik J, Glaz E, Racz K, 2004, Clinical and biochemical features of sporadic and hereditary phaeochromocytomas: an analysis of 41 cases investigated in a single endocrine centre. Eur J Cancer Prev 13(5): 403–409 Igaz P, Patocs A, Racz K, 2009, UncommonMEN2A phenotype in a patient with a RET protooncogene exon 10, codon 611 mutation. Clin Endocrinol 71(2):304–305 Gergics P, Patocs A, Toth M, Igaz P, Szucs N, Liko I, Fazakas F, Szabo I, Kovacs B, Glaz E, Racz K, 2009, Germline VHL gene mutations in Hungarian families with von Hippel-Lindau disease and patients with apparently sporadic unilateral pheochromocytomas. Eur J Endocrinol 161:495–502 Fishbein L,Merrill S, Fraker DL, Cohen DL, Nathanson KL, 2013, Inherited mutations in pheochromocytoma and paraganglioma: why all patients should be offered genetic testing. Ann Surg Oncol 20(5):1444–1450 Peczkowska M, Kowalska A, Sygut J, Waligorski D, Malinoc A, Janaszek-Sitkowska H, Prejbisz A, Januszewicz A, Neumann HP, 2013, Testing new susceptibility genes in the cohort of apparently sporadic phaeochromocytoma/paraganglioma patients with clinical characteristics of hereditary syndromes. Clin Endocrinol 79(6): 817–823 Brito JP, Asi N, Bancos I, Gionfriddo MR, Zeballos-Palacios CL, LeppinAL, Undavalli C,Wang Z, Domecq JP, PrustskyG, Elraiyah TA, Prokop LJ, Montori VM, Murad MH, 2015, Testing for germline mutations in sporadic pheochromocytoma/ paraganglioma: a systematic review. Clin Endocrinol 82(3):338– 345 Kanai H, Yamashita N, Hashimoto N, Ogawa K, Suzuki S, 2012, A case of malignant paraganglioma presenting with skull metastases. No Shinkei Geka 40(8):711–716 Williamson SR, Eble JN, Amin MB, Gupta NS, Smith SC, Sholl LM, Montironi R, Hirsch MS, Hornick JL, 2015, Succinate dehydrogenase-deficient renal cell carcinoma: detailed characterization of 11 tumors defining a unique subtype of renal cell carcinoma. Mod Pathol 28:80–94 Toledo SP, Lourenco DM Jr, Sekiya T, Lucon AM, Baena ME, Castro CC, Bortolotto LA, Zerbini MC, Siqueira SA, Toledo RA, Dahia PL, 2015, Penetrance and clinical features of pheochromocytoma in a six-generation family carrying a germline TMEM127 mutation. J Clin Endocrinol Metab 100(2):E308–E318 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 673 EP 679 DI 10.1007/s12253-016-0050-0 PG 7 ER PT J AU Farkas, K Szucs, M Nyari, AT AF Farkas, Klaudia Szucs, Monika Nyari, Andras Tibor TI Trends in Gastrointestinal Cancer Mortality Rate in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Cancer of the gallbladder; Esophageal cancer; Gastric cancers; Hepatic cancer; Pancreatic cancer; Mortality rates; Trend; Epidemiology; Hungary ID Colorectal cancer; Cancer of the gallbladder; Esophageal cancer; Gastric cancers; Hepatic cancer; Pancreatic cancer; Mortality rates; Trend; Epidemiology; Hungary AB The aim of this study was to investigate the annual death trends for gastrointestinal cancer in Hungary between 1963 and 2012. Data on the numbers of cancer deaths were obtained from the published nationwide population register. Numbers of deaths from esophageal, gastric and colorectal cancer were available during the study period. However, the mortality data for hepatic, pancreatic and gallbladder cancer have been published only since 1979. Joinpoint regression was applied to investigate the annual trends in the rates of cancer mortality. The annual mortality rates of gastric and gallbladder cancer decreased throughout the study period. Furthermore, declines in mortality from esophageal and hepatic cancers have been observed since 1998 and 1995, respectively. However, the rates of colorectal and pancreatic cancer mortality have been increasing in the past few years. Nevertheless, the mortality rates of colorectal and pancreatic cancers have increased in males aged 40–59 years during the study period. Moreover, significantly higher risks of gastrointestinal cancer-related deaths have been observed in males as compared with females except for death related to cancer of the gallbladder. The presented data suggest that the Hungarian mortality rates are particularly high. The detection of gastrointestinal cancers at an early stage would significantly improves the outcome of these malignancies. C1 [Farkas, Klaudia] University of Szeged, 1st Department of MedicineSzeged, Hungary. [Szucs, Monika] University of Szeged, Department of Medical Informatics, H-6701 Szeged, Hungary. [Nyari, Andras Tibor] University of Szeged, Department of Medical Informatics, H-6701 Szeged, Hungary. RP Nyari, AT (reprint author), University of Szeged, Department of Medical Informatics, H-6701 Szeged, Hungary. EM nyari.tibor@med.u-szeged.hu CR Allemani C, Weir HK, Carreira H, Harewood R, Spika D, Wang XS, et al., 2015, Global surveillance of cancer survival 1995-2009: analysis of individual data for 25,676,887 patients from 279 population-based registries in 67 countries, CONCORD-2). Lancet 385(9972):977–1010 The Hungarian Central Statistical Office. Demographic Yearbook, 1963-2012, Budapest. KSH 2013 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D, Bray F, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403 Bosetti C, Bertuccio P, Malvezzi M, Levi F, Chatenoud L, Negri E, La Vecchia C, 2013, Cancer mortality in Europe, 2005-2009, and an overview of trends since 1980. Ann Oncol 24:2657–2671 Jemal A, Center MM, DeSantis C, Ward EM, 2010, Global Patterns of Cancer Incidence and Mortality Rates and Trends. Cancer Epidemiol Biomark Prev 19:1893–1907 Szabolcs O, 2003, Cancer epidemiology in Hungary and the Bela Johan National Program for the decade of health. Pathol Oncol Res 9:126–130 Peter A, 2013, Connection between cancer- and alcohol-related mortality in a rural practice of a South-Hungarian village. Orv Hetil 154:700–706 Kim HJ, Fay MP, Feuer EJ,Midthune DN, 2000, Permutation tests for joinpoint regression with applications to cancer rates. Stat Med 19:335–351 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90 Spallek J, Arnold M, Razum O, Juel K, Rey G, Deboosere P, Mackenbach JP, Kunst AE, 2012, Cancer mortality patterns among Turkish immigrants in four European countries and in Turkey. Eur J Epidemiol 27:915–921 Randi G, Franceschi S, La Vecchia C, 2006, Gallbladder cancer worldwide: geographical distribution and risk factors. Int J Cancer 118:1591–1602 Levi F, Lucchini F, Negri E, La Vecchia C, 2004, Trends in Mortality from Major Cancers in The European Union, Including Acceding Countries, in 2004. Cancer 101:2843–2850 Ferlay J, Soerjomataram I, Dikshit R, Eser S,Mathers C, Rebelo M, et al., 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:E359–E386 Bosetti C, Levi F, Rosato V, Bertuccio P, Lucchini F, Negri E, La Vecchia C, 2011, Recent trends in colorectal cancer mortality in Europe. Int J Cancer 129:180–191 Sunkara V, 2015, Hebert JR. The colorectal cancer mortality-toincidence ratio as an indicator of global cancer screening and care Cancer 121:1563–1569 Ll H, Tulassay Z, 2010, Epidemiology of gastrointestinal and liver tumors. Eur Rev Med Pharmacol Sci 14:249–258 Ferro A, Peleteiro B, Malvezzi M, Bosetti C, Bertuccio P, Levi F, Negri E, La Vecchia C, Lunet N, 2014)Worldwide trends in gastric cancer mortality, 1980-2011), with predictions to 2015, and incidence by subtype. Eur J Cancer 50:1330–1344 de Vries E, Arroyave I, Pardo C,Wiesner C,Murillo R, Forman D, Burdorf A, AvendanoM(2015, Trends in inequalities in premature cancer mortality by educational level in Colombia, 1998-2007. J Epidemiol Community Health 69:408–415 Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F, 2014, Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomark Prev 23: 700–713 AEl S, RA D'A Jr, RA B, DC S, 2003, International variation in pancreatic cancer mortality for the period 1955-1998. Eur J Epidemiol 18:801–816 AAl B, LJ S, Volovics A, Dorant E, Den Brandt PA v, 2000, Trends in incidence of adenocarcinoma of the oesophagus and gastric cardia in ten European countries. Int J Epidemiol 29:645–654 Kroep S, Lansdorp-Vogelaar I, Rubenstein JH, Lemmens VE, van Heijningen EB, Aragones N, van Ballegooijen M, Inadomi JM, 2014, Comparing trends in esophageal adenocarcinoma incidence and lifestyle factors between the United States, Spain, and the Netherlands. Am J Gastroenterol 109: 336–343 quiz 335, 344 Cl C, Bosetti C, Malvezzi M, Bertuccio P, Levi F, Negri E, La Vecchia C, LunetN, 2014, Patterns and trends in esophageal cancer mortality and incidence in Europe, 1980–2011, and predictions to 2015. Ann Oncol 25:283–290 Bjerregaard JK, Mortensen MB, Pfeiffer P, Academy of Geriatric Cancer Research, AgeCare,, 2016, Trends in cancer of the liver, gall bladder, bile duct, and pancreas in elderly in Denmark, 1980– 2012. Acta Oncol 55(Suppl 1):40–45 Bosch FX, Ribes J, Diaz M, Cleries R, 2004, Primary liver cancer: worldwide incidence and trends. Gastroenterology 127(5 Suppl 1): S5–S16 Review Center MM, Jemal A, 2011, International trends in liver cancer incidence rates. Cancer Epidemiol Biomark Prev 20:2362–2368 Dobrossy L, Kovacs A, Budai A, Simon J, Horvath AR, Cornides A, Tulassay Z, 2011, Controversial issues in colorectal screening in Hungary: conflict of clinical and public health viewpoints. Orv Hetil 152:1223–1232 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 681 EP 688 DI 10.1007/s12253-016-0052-y PG 8 ER PT J AU Mamede, CA Guerra, S Laranjo, M Santos, K Carvalho, JM Carvalheiro, T Moura, P Paiva, A Abrantes, MA Maia, C Botelho, FM AF Mamede, Catarina Ana Guerra, Sara Laranjo, Mafalda Santos, Kathleen Carvalho, Joao Maria Carvalheiro, Tiago Moura, Paulo Paiva, Artur Abrantes, Margarida Ana Maia, J. Claudio Botelho, Filomena Maria TI Oxidative Stress, DNA, Cell Cycle/Cell Cycle Associated Proteins and Multidrug Resistance Proteins: Targets of Human Amniotic Membrane in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human amniotic membrane; Hepatocellular carcinoma; Protein extracts; hAMPE; Oxidative stress; Cell cycle ID Human amniotic membrane; Hepatocellular carcinoma; Protein extracts; hAMPE; Oxidative stress; Cell cycle AB The anticancer effects of human amniotic membrane (hAM) have been studied over the last decade. However, the action mechanisms responsible for these effects are not fully understood until now. Previously results reported by our team proved that hAM is able to induce cytotoxicity and cell death in hepatocellular carcinoma (HCC), a worldwide high incident and mortal cancer. Therefore, this experimental study aimed to investigate the cellular targets of hAM protein extracts (hAMPE) in HCC through in vitro studies. Our results showed that hAMPE is able to modify oxidative stress environment in all HCC cell lines, as well as its cell cycle. hAMPE differently targets deoxyribonucleic acid (DNA), P21, P53, β-catenin and multidrug resistance (MDR) proteins in HCC cell lines. In conclusion, hAMPE has several targets in HCC, being clear that the success of this treatment depends of a personalized therapy based on the biological and genetic characteristics of the tumor. C1 [Mamede, Catarina Ana] University of Coimbra, Faculty of Medicine, Biophysics Institute, Azinhaga de Santa Comba - Celas, 3000-548 Coimbra, Portugal. [Guerra, Sara] University of Coimbra, Faculty of Medicine, Biophysics Institute, Azinhaga de Santa Comba - Celas, 3000-548 Coimbra, Portugal. [Laranjo, Mafalda] University of Coimbra, Faculty of Medicine, Biophysics Institute, Azinhaga de Santa Comba - Celas, 3000-548 Coimbra, Portugal. [Santos, Kathleen] University of Coimbra, Faculty of Medicine, Biophysics Institute, Azinhaga de Santa Comba - Celas, 3000-548 Coimbra, Portugal. [Carvalho, Joao Maria] University of Coimbra, Faculty of Medicine, Biophysics Institute, Azinhaga de Santa Comba - Celas, 3000-548 Coimbra, Portugal. [Carvalheiro, Tiago] Portuguese Institute of the Blood and Transplantation, Blood and Transplantation Center of CoimbraCoimbra, Portugal. [Moura, Paulo] Coimbra Hospital and University Centre, Obstetrics ServiceCoimbra, Portugal. [Paiva, Artur] Coimbra Hospital and University Centre, Clinical Pathology Department, Cytometry Operational Management UnitCoimbra, Portugal. [Abrantes, Margarida Ana] University of Coimbra, Faculty of Medicine, Biophysics Institute, Azinhaga de Santa Comba - Celas, 3000-548 Coimbra, Portugal. [Maia, J. Claudio] University of Beira Interior, Health Sciences Research Centre, CICS-UBICovilha, Portugal. [Botelho, Filomena Maria] University of Coimbra, Faculty of Medicine, Biophysics Institute, Azinhaga de Santa Comba - Celas, 3000-548 Coimbra, Portugal. RP Mamede, CA (reprint author), University of Coimbra, Faculty of Medicine, Biophysics Institute, 3000-548 Coimbra, Portugal. EM ana_mamede@hotmail.com CR Toda A, Okabe M, Yoshida T, Nikaido T, 2007, The potential of amniotic membrane/amnion-derived cells for regeneration of various tissues. J Pharmacol Sci 105:215–228 Davis J, 1910, Skin transplantation with a review of 550 cases at the Johns Hopkins Hospital. Johns Hopkins Med J 15:307 Gomes J, Romano A, Santos M,DuaH(2005)Amniotic membrane use in ophthalmology. Curr Opin Ophthalmol 16:233–240 Dua H, Azuara-Blanco A, 1999, Amniotic membrane transplantation. Br J Ophthalmol 83:748–752 Sawhney C, 1989, Amniotic membrane as a biological dressing in the management of burns. Burns 15:339–342 Mamede A, Carvalho M, Abrantes A, et al., 2012, Amniotic membrane: from structure and functions to clinical applications. Cell Tissue Res 349:447–458 Magatti M, De Munari S, Vertua E, Parolini O, 2012, Amniotic membrane-derived cells inhibit proliferation of cancer cell lines by inducing cell cycle arrest. J Cell Mol Med 16:2208–2218 Jiao H, Guan F, Yang B, et al., 2012, Human amniotic membrane derived-mesenchymal stem cells induce C6 glioma apoptosis in vivo through the bcl-2/caspase pathways. Mol Biol Rep 39:467– 473 Rolfo A, Giuffrida D, Giuffrida M, et al., 2014, New perspectives for prostate cancer treatment: in vitro inhibition of LNCaP and PC3 cell proliferation by amnion-derived mesenchymal stromal cells conditioned media. Aging Male 17: 94–101 Kang N-H, Yi B-R, Lim S, et al., 2012, Human amniotic membrane-derived epithelial stem cells display anticancer activity in BALB/c female nude mice bearing disseminated breast cancer xenografts. Int J Oncol 40:2022–2028 Niknejad H, Khayat-Khoei M, Peirovi H, Abolghasemi H, 2014, Human amniotic epithelial cells induce apoptosis of cancer cells: a new anti-tumor therapeutic strategy. Cytotherapy 16: 33–40 Mamede A, Laranjo M, CarvalhoM, et al., 2014, Effect of amniotic membrane proteins in human cancer cell lines: an exploratory study. J Membr Biol 247:357–360 Mamede A, Guerra S, Laranjo M, et al., 2015, Selective cytotoxicity and cell death induced by human amniotic membrane in hepatocellular carcinoma. Med Oncol 32:257 Gomes M, Priolli D, Tralhao J, Botelho M, 2013, Hepatocellular carcinoma: epidemiology, biology, diagnosis and therapies. Rev Assoc Med Bras 59:514–524 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, et al., 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374– 1403 Marra M, Sordelli I, Lombardi A, et al., 2011, Molecular targets and oxidative stress biomarkers in hepatocellular carcinoma: an overview. J Transl Med 9:171 Alves R, Alves D, Guz B, et al., 2011, Advanced hepatocellular carcinoma: review of targeted molecular drugs. Ann Hepatol 10: 21–27 Brito A, Abrantes A, Pinto-Costa C, et al., 2013, Hepatocellular carcinoma and chemotherapy: the role of p53. Chemotherapy 58: 381–386 Tralhao J, Abrantes A, Hoti E, et al., 2013, Hepatectomy and liver regeneration: from experimental research to clinical application. ANZ J Surg 84:665–671 Abrantes A, Serra M, Goncalves A, et al., 2010, Hypoxia-induced redox alterations and their correlation with 99mTc-MIBI and 99mTc-HL-91 uptake in colon cancer cells. Nucl Med Biol 37: 125–132 Tralhao J, Abrantes A, Goncalves A, et al., 2013, Study of hepatocellular function in the murine model following hepatic artery selective clamping. Acta Cir Bras 28:657–663 Gomes A, Fernandes E, Lima J, 2005, Fluorescence probes used for detection of reactive oxygen species. J Biochem Biophys Methods 65:45–80 Mamede A, Pires A, Abrantes A, et al., 2012, Cytotoxicity of ascorbic acid in a human colorectal adenocarcinoma cell line, WiDr): in vitro and in vivo studies. Nutr Cancer 64:1049–1057 Larrauri A, Lopez P, Gomez-Lechon M, Castell J, 1987, A cytochemical stain for glutathione in rat hepatocytes cultured on plastic. J Histochem Cytochem 35:271–274 Ioannou Y, Chen F, 1996, Quantitation of DNA fragmentation in apoptosis. Nucleic Acids Res 24:992–993 Santos K, Laranjo M, Abrantes A, et al., 2014, Targeting triple-negative breast cancer cells with 6,7-bis(hydroxymethyl)-1H, 3H-pyrrolo[1,2-c]thiazoles. Eur J Med Chem 79:273–281 Serra A, Am RG, Laranjo M, et al., 2012, Synthesis of new 2- galactosylthiazolidine-4-carboxylic acid amides. antitumor evaluation againstmelanoma and breast cancer cells. Eur J Med Chem 53: 398–402 Brito A, Abrantes A, Ribeiro M, et al., 2015, Fluorine-18 fluorodeoxyglucose uptake in hepatocellular carcinoma: correlation with glucose transporters and p53 expression. J Clin Exp Hepatol 5:183–189 Gomes A, Abrantes A, Brito A, et al., 2015, P53 influence on the radiotherapy response of hepatocellular carcinoma. Clin Mol Hepatol 21:257–267 Brito A, Mendes M, Abrantes A, et al., 2014, Positron emission tomography diagnostic imaging in multidrug-resistant hepatocellular carcinoma: focus on 2-deoxy-2-(18F)fluoro-d-glucose. Mol Diagn Ther 18:495–504 Casalta-Lopes J, Abrantes A, Laranjo M, et al., 2011, Efflux pumps modulation in colorectal adenocarcinoma cell lines: the role of nuclear medicine. J Cancer Ther 02:408–417 Choi K, Kim J, Kim G, Choi C, 2009, Oxidative stress-induced necrotic cell death via mitochondira-dependent burst of reactive oxygen species. Curr Neurovasc Res 6:213–222 Valko M, Leibfritz D, Moncol J, et al., 2007, Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol 39:44–84 LiY, Schellhorn H(2007)New developments and novel therapeutic perspectives for vitamin C. J Nutr 137:2171–2184 Georgiou C, Papapostolou I, Grintzalis K, 2009, Protocol for the quantitative assessment of DNA concentration and damage, fragmentation and nicks). Nat Protoc 4:125–131 Reuter S, Gupta S, Chaturvedi M, Aggarwal B, 2010, Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 49:1603–1616 Oberhammer F, Wilson J, Dive C, et al., 1993, Apoptotic death in epithelial cells: cleavage of DNA to 300 and/or 50 kb fragments prior to or in the absence of internucleosomal fragmentation. EMBO J 12:3679–3684 Zamai L, Falcieri E, Marhefka G, Vitale M, 1996, Supravital exposure to propidium iodide identifies apoptotic cells in the absence of nucleosomal DNA fragmentation. Cytometry 23:303–311 Macleod K, Sherry N, Hannon G, et al., 1995, p53-dependent and independent expression of p21 during cell growth, differentiation, and DNA damage. Genes Dev 9:935–944 Hussain S, Schwank J, Staib F, et al., 2007, TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene 26:2166–2176 Cayrol C, Knibiehler M, Ducommun B, 1998, p21 binding to PCNA causes G1 and G2 cell cycle arrest in p53-deficient cells. Oncogene 16:311–320 Punchihewa C, Inoue A, Hishiki A, et al., 2012, Identification of small molecule proliferating cell nuclear antigen, PCNA, inhibitor that disrupts interactions with PIP-box proteins and inhibits DNA replication. J Biol Chem 287: 14289–14300 Stark G, Taylor W, 2004, Analyzing the G2/M checkpoint. In: Schonthal AH, ed, Checkp. Control. cancer Rev. Model Syst. Humana Press, Totowa, pp. 51–82 Wu G, Xu L, LinN, Liu B, 2013, UCN-01 induces S and G2/Mcell cycle arrest through the p53/p21waf1 or CHK2/CDC25C pathways and can suppress invasion in human hepatoma cell lines. BMC Cancer 13:167 Chan K-T, Lung M, 2004, Mutant p53 expression enhances drug resistance in a hepatocellular carcinoma cell line. Cancer Chemother Pharmacol 53:519–526 Tsang W-P, Chau S, Fung K-P, et al., 2003, Modulation of multidrug resistance-associated protein 1, MRP1, by p53mutant in saos- 2 cells. Cancer Chemother Pharmacol 51:161–166 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 689 EP 697 DI 10.1007/s12253-016-0053-x PG 9 ER PT J AU Chen, HI Tsai, HP Chen, YT Tsao, ShCh Chai, ChY AF Chen, Hsin-I Tsai, Hung-Pei Chen, Yi-Ting Tsao, Shu-Chuan Chai, Chee-Yin TI Autophagy and Apoptosis Play Opposing Roles in Overall Survival of Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Autophagy; Apoptosis; Esophageal squamous cell carcinoma ID Autophagy; Apoptosis; Esophageal squamous cell carcinoma AB Esophageal cancer is among the most aggressive gastrointestinal tract malignancies, and squamous cell carcinoma is the most common subtype. Although both autophagy and apoptosis involve programmed cell death, autophagy also maintains cell survival by recycling cellular waste. The relationship between autophagy and apoptosis in esophageal squamous cell carcinoma (ESCC) is unclear. Autophagic and apoptotic markers of ESCC were detected by immunohistochemical staining (IHC) in 43 ESCC patients treated during 2007–2011. Chi-square test and Kaplan-Meier method were used to determine how clinicopathological parameters were related to IHC results for LC3B, Beclin-1 and caspase-3 (CASP-3). Correlations among Beclin-1, LC3B, and CASP- 3 were analyzed by Spearman rho. The statistical analyses revealed no clinicopathological parameters that significantly correlated with expressions of Beclin-1, LC3B, and CASP-3. However, low CASP-3 expression and high LC3B expression revealed by IHC were predictors of a poor prognosis. Additionally, LC3B expression had a significant negative correlation with CASP-3 expression. Autophagy is antagonistic to apoptosis and predicts poor overall survival in ESCC. C1 [Chen, Hsin-I] Kaohsiung Medical University, College of Medicine, Department of Pathology, 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. [Tsai, Hung-Pei] Kaohsiung Medical University, College of Medicine, Graduate Institute of Clinical MedicineKaohsiung, Taiwan, Republic of China. [Chen, Yi-Ting] Kaohsiung Medical University, College of Medicine, Department of Pathology, 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. [Tsao, Shu-Chuan] Kaohsiung Medical University, College of Medicine, Department of Pathology, 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. [Chai, Chee-Yin] Kaohsiung Medical University, College of Medicine, Department of Pathology, 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. RP Chai, ChY (reprint author), Kaohsiung Medical University, College of Medicine, Department of Pathology, 807 Kaohsiung, Taiwan, Republic of China. EM cychai@kmu.edu.tw CR Bosman FT, Carneiro F, Hruban RH, Theise ND, 2010, World Health Organization classification of tumors of the digestive system. IARC Press, Lyon, France Ina H1 SH, Ohashi I, Kitagawa M, 1994, The frequency of a concomitant early esophageal cancer in male patients with oral and oropharyngeal cancer: screening results using lugol dye endoscopy. Cancer 73(8):2038–2041 Yorimitsu T, Klionsky DJ, 2005, Autophagy: molecular machinery for self-eating. Cell Death Differ 12(Suppl 2):1542–1552 Zheng HC, Sun JM, Wei ZL, Yang XF, Zhang YC, Xin Y, 2003, Expression of Fas ligand and caspase-3 contributes to formation of immune escape in gastric cancer.World J Gastroenterol 9(7): 1415–1420 Choi KS, 2012, Autophagy and cancer. Exp Mol Med 44(2):109–120 Kung CP, Budina A, Balaburski G, Bergenstock MK, Murphy M, 2011, Autophagy in tumor suppression and cancer therapy. Crit Rev Eukaryot Gene Expr 21(1):71–100 Hanna RA, Quinsay MN, Orogo AM, Giang K, Rikka S, Gustafsson AB, 2012)Microtubule-associated protein 1 light chain 3, LC3, interacts with Bnip3 protein to selectively remove endoplasmic reticulum and mitochondria via autophagy. J Biol Chem 287(23):19094–19104 Liang XH, Yu J, Brown K, Levine B, 2001, Beclin 1 contains a leucine-rich nuclear export signal that is required for its autophagy and tumor suppressor function. Cancer Res 61(8):3443–3449 Shrivastava S, Bhanja Chowdhury J, Steele R, Ray R, Ray RB, 2012, Hepatitis C virus upregulates Beclin1 for induction of autophagy and activates mTOR signaling. J Virol 86(16):8705–8712 Chen S, Jiang YZ, Huang L, Zhou RJ, Yu KD, Liu Y, Shao ZM, 2013, The residual tumor autophagy marker LC3B serves as a prognostic marker in local advanced breast cancer after neoadjuvant chemotherapy. Clin Cancer Res 19(24):6853–6862 Huang X, Bai HM, Chen L, Li B, Lu YC, 2010, Reduced expression of LC3B-II and beclin 1 in glioblastoma multiforme indicates a down-regulated autophagic capacity that relates to the progression of astrocytic tumors. J Clin Neurosci 17(12):1515–1519 Melguizo C, Prados J, Gonzalez B, Ortiz R, Concha A, Alvarez PJ, Madeddu R, Perazzoli G, Oliver JA, Lopez R, Rodriguez-Serrano F, Aranega A, 2012, MGMT promoter methylation status and MGMT and CD133 immunohistochemical expression as prognostic markers in glioblastoma patients treated with temozolomide plus radiotherapy. J Transl Med 10:250 Lai SW, Liao KF, Lai HC, Muo CH, Sung FC, 2012, Atorvastatin Correlates with Decreased Risk of Esophageal Cancer: a Population-Based Case-Control Study from Taiwan Libyan J Med 7 Chen Y, Li X,Wu X, He C, Guo L, Zhang S, Xiao Y, GuoW, Tan B, 2013, Autophagy-related proteins LC3 and beclin-1 impact the efficacy of chemoradiation on esophageal squamous cell carcinoma. Pathol Res Pract 209(9):562–567 Liu EY, Ryan KM(2012, Autophagy and cancer–issues we need to digest. J Cell Sci 125(Pt 10):2349–2358 Li J, Hou N, Faried A, Tsutsumi S, Kuwano H, 2010, Inhibition of autophagy augments 5-fluorouracil chemotherapy in human colon cancer in vitro and in vivo model. Eur J Cancer 46(10):1900–1909 Liu D, Yang Y, Liu Q,Wang J, 2011, Inhibition of autophagy by 3- MA potentiates cisplatin-induced apoptosis in esophageal squamous cell carcinoma cells. Med Oncol 28(1):105–111 Apel A, Herr I, Schwarz H, Rodemann HP, Mayer A, 2008, Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res 68(5):1485–1494 Huang H, Zhang XF, ZhouHJ, Xue YH, Dong QZ, Ye QH, Qin LX, 2010, Expression and prognostic significance of osteopontin and caspase-3 in hepatocellular carcinoma patients after curative resection. Cancer Sci 101(5):1314–1319 Li WW, Li J, Bao JK, 2012, Microautophagy: lesser-known selfeating. Cell Mol Life Sci 69(7):1125–1136 Yousefi S, Perozzo R, Schmid I, Ziemiecki A, Schaffner T, Scapozza L, Brunner T, Simon HU, 2006, Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis. Nat Cell Biol 8(10):1124–1132 Del Bello B, Toscano M, Moretti D, Maellaro E, 2013, Cisplatininduced apoptosis inhibits autophagy, which acts as a pro-survival mechanism in human melanoma cells. PLoS One 8(2):e57236 Young MM, Takahashi Y, Khan O, Park S, Hori T, Yun J, Sharma AK, Amin S, Hu CD, Zhang J, Kester M, Wang HG, 2012, Autophagosomal membrane serves as platform for intracellular death-inducing signaling complex, iDISC)-mediated caspase-8 activation and apoptosis. J Biol Chem 287(15):12455–12468 Wirawan E, Vande Walle L, Kersse K, Cornelis S, Claerhout S, Vanoverberghe I, Roelandt R, De Rycke R, Verspurten J, Declercq W, Agostinis P, Vanden Berghe T, Lippens S, Vandenabeele P, 2010, Caspase-mediated cleavage of beclin-1 inactivates beclin-1-induced autophagy and enhances apoptosis by promoting the release of proapoptotic factors from mitochondria. Cell Death Dis 1:e18 Ni Chonghaile T, Letai A, 2011, Who put the "A" in Atg12: autophagy or apoptosis? Mol Cell 44(6):844–845 Hu YL, Jahangiri A, Delay M, Aghi MK, 2012, Tumor cell autophagy as an adaptive response mediating resistance to treatments such as antiangiogenic therapy. Cancer Res 72(17):4294–4299 Su M, Mei Y, Sinha S, 2013, Role of the Crosstalk between Autophagy and Apoptosis in Cancer. J Oncol 2013(102735 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 699 EP 705 DI 10.1007/s12253-016-0051-z PG 7 ER PT J AU Karadima, LM Saetta, AA Chatziandreou, I Lazaris, CA Patsouris, E Tsavaris, N AF Karadima, L Maria Saetta, A Angelica Chatziandreou, Ilenia Lazaris, C Andreas Patsouris, Efstratios Tsavaris, Nikolaos TI The Prognostic Influence of BRAF Mutation and other Molecular, Clinical and Laboratory Parameters in Stage IV Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article AB Our aim was to evaluate the predictive and prognostic influence of BRAF mutation and other molecular, clinical and laboratory parameters in stage IV colorectal cancer (CRC). 60 patients were included in this retrospective analysis, and 17 variables were examined for their relation with treatment response and survival. KRAS mutation was identified in 40.3 % of cases, BRAF and PIK3CA in 8.8 % and 10.5 % respectively. 29.8 % of patients responded to treatment. Median survival time was 14.3 months. Weight loss, fever, abdominal metastases, blood transfusion, hypoalbuminaimia, BRAF and PIK3CA mutations, CRP and DNA Index were associated with survival. In multivariate analysis, male patients had 3.8 times higher probability of response, increased DNA Index was inversely correlated with response and one unit raise of DNA Index augmented 6 times the probability of death. Our findings potentiate the prognostic role of BRAF, PIK3CA mutations and ploidy in advanced CRC. C1 [Karadima, L Maria] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, Department of Pathophysiology, Oncology Unit, Mikras Asias 75 Street, 11527 Athens, Greece. [Saetta, A Angelica] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, First Department of PathologyAthens, Greece. [Chatziandreou, Ilenia] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, First Department of PathologyAthens, Greece. [Lazaris, C Andreas] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, First Department of PathologyAthens, Greece. [Patsouris, Efstratios] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, First Department of PathologyAthens, Greece. [Tsavaris, Nikolaos] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, Department of Pathophysiology, Oncology Unit, Mikras Asias 75 Street, 11527 Athens, Greece. RP Karadima, LM (reprint author), National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, Department of Pathophysiology, Oncology Unit, 11527 Athens, Greece. EM m_karadima@hotmail.com CR Siena S, Sartore-Bianchi A, Di Nicolantonio F, Balfour J, Bardelli A, 2009, Biomarkers predicting clinical outcome of epidermal growth factor receptor-targeted therapy in metastatic colorectal cancer. Journal of the National Cancer Institute 101:1308–1324 Sartore-Bianchi A, Di Nicolantonio F, Nichelatti M, Molinari F, De Dosso S, Saletti P, Martini M, Cipani T, Marrapese G, Mazzucchelli L, Lamba S, Veronese S, et al., 2009, Multi-determinants analysis of molecular alterations for predicting clinical benefit to EGFR-targeted monoclonal antibodies in colorectal cancer. PloS One 4:e7287 Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, Jones CM, Marshall CJ, Springer CJ, Barford D, Marais R, Cancer GP, 2004, Mechanism of activation of the RAFERK signaling pathway by oncogenic mutations of B-RAF. Cell 116:855–867 Wang L, Cunningham JM, Winters JL, Guenther JC, French AJ, Boardman LA, Burgart LJ, McDonnell SK, Schaid DJ, Thibodeau SN, 2003, BRAF mutations in colon cancer are not likely attributable to defective DNA mismatch repair. Cancer Research 63:5209–5212 Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, De Dosso S, Mazzucchelli L, Frattini M, Siena S, Bardelli A, 2008, Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 26:5705–5712 Laurent-Puig P, Cayre A, Manceau G, Buc E, Bachet JB, Lecomte T, Rougier P, Lievre A, Landi B, Boige V, Ducreux M, YchouM, et al., 2009, Analysis of PTEN, BRAF, and EGFR status in determining benefit from cetuximab therapy in wild-type KRAS metastatic colon cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 27:5924–5930 Tol J, Nagtegaal ID, Punt CJ, 2009, BRAF mutation in metastatic colorectal cancer. The New England Journal of Medicine 361:98– 99 Roth AD, Tejpar S, Delorenzi M, Yan P, Fiocca R, Klingbiel D, Dietrich D, Biesmans B, Bodoky G, Barone C, Aranda E, Nordlinger B, et al., 2010, Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 28:466–474 Mao M, Tian F, Mariadason JM, Tsao CC, Lemos R Jr, Dayyani F, Gopal YN, Jiang ZQ,Wistuba II, Tang XM, Bornman WG, Bollag G, et al., 2013, Resistance to BRAF inhibition in BRAF-mutant colon cancer can be overcome with PI3K inhibition or demethylating agents. Clinical cancer research: an official journal of the American Association for Cancer Research 19:657–667 SamuelsY,Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S,Yan H, Gazdar A, Powell SM, Riggins GJ,Willson JK,Markowitz S, et al., 2004, High frequency of mutations of the PIK3CA gene in human cancers. Science 304:554 Liao X, Morikawa T, Lochhead P, Imamura Y, Kuchiba A, Yamauchi M, Nosho K, Qian ZR, Nishihara R, Meyerhardt JA, Fuchs CS, Ogino S, 2012, Prognostic role of PIK3CA mutation in colorectal cancer: cohort study and literature review. Clinical cancer research: an official journal of the American Association for Cancer Research 18:2257–2268 GradyWM, Carethers JM, 2008, Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology 135:1079– 1099 Xynos ID, Kavantzas N, Tsaousi S, Zacharakis M, Agrogiannis G, Kosmas C, Lazaris A, Sarantonis J, Sougioultzis S, Tzivras D, Polyzos A, Patsouris ES, et al., 2013, Factors Influencing survival in stage IV colorectal cancer: the influence of DNA ploidy. ISRN gastroenterology 2013:490578 Korkolopoulou P, Levidou G, Trigka EA, Prekete N, Karlou M, Thymara I, Sakellariou S, Fragkou P, Isaiadis D, Pavlopoulos P, Patsouris E, Saetta AA, 2012, A comprehensive immunohistochemical and molecular approach to the PI3K/AKT/mTOR, phosphoinositide 3-kinase/v-akt murine thymoma viral oncogene/mammalian target of rapamycin, pathway in bladder urothelial carcinoma. BJU International 110:E1237–E1248 Trigka EA, Levidou G, Saetta AA, Chatziandreou I, Tomos P, Thalassinos N, Anastasiou N, Spartalis E, Kavantzas N, Patsouris E, Korkolopoulou P, 2013, A detailed immunohistochemical analysis of the PI3K/AKT/mTOR pathway in lung cancer: correlation with PIK3CA, AKT1, K-RAS or PTEN mutational status and clinicopathological features. Oncology Reports 30:623–636 Syrios J, Sougioultzis S, Xynos ID, Kavantzas N, Kosmas C, Agrogiannis G, Griniatsos J, Karavokyros I, Pikoulis E, Patsouris ES, Tsavaris N, 2012, Survival in patients with stage IV noncardia gastric cancer - the influence of DNA ploidy and Helicobacter pylori infection. BMC Cancer 12:264 Chelidonis G, Kavantzas N, Patsouris E, Pagaki E, Athanasiadou AM, Agrogiannis G, Athanasiadou P. DNA ploidy, E-cadherin, beta-catenin expression and their clinicopathologic significance in imprints of non-small cell lung cancer. Analytical and quantitative cytology and histology/the International Academy of Cytology [and] American Society of Cytology 2009;31: 332–339. Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, Juan T, Sikorski R, Suggs S, Radinsky R, Patterson SD, Chang DD, 2008, Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 26:1626–1634 Allegra CJ, Jessup JM, Somerfield MR, Hamilton SR, Hammond EH, Hayes DF, McAllister PK, Morton RF, Schilsky RL, 2009, American Society of Clinical Oncology provisional clinical opinion: testing for KRAS gene mutations in patients with metastatic colorectal carcinoma to predict response to anti-epidermal growth factor receptor monoclonal antibody therapy. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 27:2091–2096 Herreros-Villanueva M, Gomez-Manero N, Muniz P, Garcia-Giron C, 2011, Coma del Corral MJ. PIK3CA mutations in KRAS and BRAF wild type colorectal cancer patients. A study of Spanish population. Molecular Biology Reports 38:1347–1351 Perrone F, Lampis A, Orsenigo M, Di Bartolomeo M, Gevorgyan A, Losa M, Frattini M, Riva C, Andreola S, Bajetta E, Bertario L, Leo E, et al., 2009, PI3KCA/PTEN deregulation contributes to impaired responses to cetuximab in metastatic colorectal cancer patients. Annals of oncology: official journal of the European Society for Medical Oncology/ESMO 20:84–90 Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, Rivera F, Kocakova I, et al., 2013, Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. The New England Journal of Medicine 369:1023–1034 De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, Kalogeras KT, Kotoula V, Papamichael D, Laurent- Puig P, Penault-Llorca F, Rougier P, et al., 2010, Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. The Lancet Oncology 11:753–762 McMillan DC, Wotherspoon HA, Fearon KC, Sturgeon C, Cooke TG, McArdle CS, 1995, A prospective study of tumor recurrence and the acute-phase response after apparently curative colorectal cancer surgery. American Journal of Surgery 170:319–322 Kohne CH, Cunningham D, Di Costanzo F, Glimelius B, Blijham G, Aranda E, ScheithauerW, Rougier P, PalmerM,Wils J, Baron B, Pignatti F, et al., 2002, Clinical determinants of survival in patients with 5-fluorouracil-based treatment for metastatic colorectal cancer: results of a multivariate analysis of 3825 patients. Annals of oncology: official journal of the European Society for Medical Oncology/ ESMO 13:308–317 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 707 EP 714 DI 10.1007/s12253-016-0056-7 PG 8 ER PT J AU Sam, S Reza Sam, M Esmaeillou, M Safaralizadeh, R AF Sam, Sohrab Reza Sam, Mohammad Esmaeillou, Mohammad Safaralizadeh, Reza TI Effective Targeting Survivin, Caspase-3 and MicroRNA-16-1 Expression by Methyl-3-pentyl-6-methoxyprodigiosene Triggers Apoptosis in Colorectal Cancer Stem-Like Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Survivin; Colorectal cancer stem cells; MicroRNA-16-1; Colorectal cancer; Prodigiosin; Serratia marcescens; Caspase-3; Apoptosis ID Survivin; Colorectal cancer stem cells; MicroRNA-16-1; Colorectal cancer; Prodigiosin; Serratia marcescens; Caspase-3; Apoptosis AB Over-expression of the proto-oncogene survivin in colorectal cancer stem cells (CCSCs) is thought to be one the primary causes for therapy failure. It has also been reported that tumor suppressor miR-16-1 is down-regulated in colorectal cancer (CRC) cells. Therefore, the search for new antiproliferative agents which target survivin or miR-16-1 in CCSCs is warranted. Several studies have shown that prodigiosin isolated from cell wall of Serratia marcescens induces apoptosis in different kinds of cancer cells. Here, we investigated the effects of prodigiosin on HCT-116 cells that serve as a model for CRC initiating cells with stem-like cells properties. HCT-116 cells were treated with 100, 200 and 400 nM prodigiosin after which cell number, viability, growth-rate, survivin and miRNA-16-1 expression, caspase- 3 activation and apoptotic rate were evaluated. Prodigiosin decreased significantly growth-rate in a dose-and time-dependent manner. After a 48 h treatment with 100, 200 and 400 nM prodigiosin, growth-rates were measured to be 84.4 ± 9.2 %, 58 ± 6.5 % and 46.3 ± 5.2 %, respectively, compared to untreated cells. We also found that treatment for 48 h with indicated concentrations of prodigiosin resulted in 41 %, 54.5 % and 63 % decrease in survivin mRNA levels and induced 32 %, 48 % and 61 % decrease in survivin protein levels as well as resulted in 128.3 ± 10 %, 178.7 ± 6.1 % and 205 ± 7.6%increase in caspase-3 activation respectively compared to untreated cells. Prodigiosin caused a significant increase in miRNA-16-1 expression at a concentration of 100 nM and treatment with different concentrations of prodigiosin resulted in 2.2- to 3-fold increase in miRNA-16- 1/survivin ratios compared to untreated cells. An increase in number of apoptotic cells ranging from 28.2 % to 86.8 % was also observed with increasing prodigiosin concentrations. Our results provide the first evidence that survivin and miRNA-16- 1 as potential biomarkers could be targeted in CRC initiating cells with stem-like cells properties by prodigiosin and this compound with high pro-apoptotic capacity represents the possibility of its therapeutic application directed against CCSCs. C1 [Sam, Sohrab] Urmia University, Institute of Biotechnology, Department of Cellular and Molecular BiotechnologyUrmia, Iran. [Reza Sam, Mohammad] Urmia University, Institute of Biotechnology, Department of Cellular and Molecular BiotechnologyUrmia, Iran. [Esmaeillou, Mohammad] Urmia University, Institute of Biotechnology, Department of Cellular and Molecular BiotechnologyUrmia, Iran. [Safaralizadeh, Reza] University of Tabriz, Faculty of Natural Science, Department of Animal BiologyTabriz, Iran. RP Reza Sam, M (reprint author), Urmia University, Institute of Biotechnology, Department of Cellular and Molecular Biotechnology, Urmia, Iran. EM s_mohammadreza@yahoo.com;m.sam@urmia.ac.ir CR Siegel R, Desantis C, Jemal A, 2014, Colorectal cancer statistics. CA Cancer J Clin 64(2):104–117 Safaee A, Fatemi SR, Ashtari S, Vahedi M, Moghimi-Dehkordi B, Zali MR, 2012, Four years incidence rate of colorectal cancer in Iran: a survey of national cancer registry data-implications for screening. Asian Pac J Cancer Prev 13(6):2695–2698 Saunders M, Iveson T, 2006, Management of advanced colorectal cancer: state of the art. Br J Cancer 95(2):131–138 Wilson BJ, Schatton T, Frank MH, Frank NY, 2011, Colorectal cancer stem cells: biology and therapeutic implications. Curr Colorectal Cancer Rep 7(2):128–135 Dalerba P, Cho RW, Clarke MF, 2007, Cancer stem cells: models and concepts. Annu Rev Med 58:267–284 Reya T, Morrison SJ, Clarke MF, Weissman IL, 2001, Stem cells, cancer, and cancer stem cells. Nature 414(6859):105–111 O’Brien CA, Pollett A, Gallinger S, Dick JE, 2007, A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445(7123):106–110 Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R, 2007, Identification and expansion of human colon-cancer-initiating cells. Nature 445(7123):111–115 Deorukhkar AA, Chander R, Ghosh SB, Sainis KB, 2007, Identification of a red-pigmented bacterium producing a potent anti-tumor N-alkylated prodigiosin as Serratia marcescens. Res Microbiol 158(5):399–404 Sumathi C, MohanaPriya D, Swarnalatha S, Dinesh GM, Sekaran G, 2014, Production of prodigiosin using tannery fleshing and evaluating its pharmacological effects. Sci World J 2014:290327 Francisco R, Perez-Tomas R, Gimenez-Bonafe P, Soto-Cerrato V, Gimenez-Xavier P, Ambrosio S, 2007, Mechanisms of prodigiosin cytotoxicity in human neuroblastoma cell lines. Eur J Pharmacol 572(2–3):111–119 Ho TF, Peng YT, Chuang SM, Lin SC, Feng BL, Lu CH, Yu WJ, Chang JS, Chang CC, 2009, Prodigiosin down-regulates survivin to facilitate paclitaxel sensitization in human breast carcinoma cell lines. Toxicol Appl Pharmacol 235(2):253–260 Hassankhani R, Sam MR, Esmaeillou M, Ahangar P, 2015, Prodigiosin isolated from cell wall of Serratia marcescens alters expression of apoptosis-related genes and increases apoptosis in colorectal cancer cells. Med Oncol 32(1):366 Sarela AI, Macadam RC, Farmery SM, Markham AF, Guillou PJ, 2000, Expression of the antiapoptosis gene, survivin, predicts death from recurrent colorectal carcinoma. Gut 46(5):645–650 Violette S, Poulain L, Dussaulx E, Pepin D, Faussat AM, Chambaz J, Lacorte JM, Staedel C, Lesuffleur T, 2002, Resistance of colon cancer cells to long-terms 5-flourouracil exposure is correlated to the relative level of BCL-2 and BCL-XL in addition to BAX and P53 status. Int J Cancer 98(4):498–504 Lee RC, Feinbaum RL, Ambros V, 1993, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75(5):843–854 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116(2):281–297 Bartel DP, 2009, MicroRNAs: target recognition and regulatory functions. Cell 136(2):215–233 Esquela-Kerscher A, Slack FJ, 2006, Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer 6(4):259–269 Huang Q, Gumireddy K, Schrier M, le Sage C, Nagel R, Nair S, Egan DA, Li A, Huang G, Klein-Szanto AJ, et al., 2008, The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat Cell Biol 10(2):202–210 Zhang BG, Li JF, Yu BQ, Zhu ZG, Liu BY, Yan M, 2012, microRNA-21 promotes tumor proliferation and invasion in gastric cancer by targeting PTEN. Oncol Rep 27(4):1019–1026 Lee DY, Deng Z, Wang CH, Yang BB, 2007, MicroRNA-378 promotes cell survival, tumor growth, and angiogenesis by targeting SuFu and Fus-1expression. Proc Natl Acad Sci U S A 104(51):20350–20355 Young LE, Moore AE, Sokol L, Meisner-Kober N, Dixon DA, 2012, The mRNA stability factor HuR inhibits microRNA-16 targeting of COX-2. Mol Cancer Res 10(1):167–180 Ma Q,Wang X, Li Z, Li B, Ma F, Peng L, Zhang Y, Xu A, Jiang B, 2013, microRNA-16 represses colorectal cancer cell growth in vitro by regulating the p53/survivin signaling pathway. Oncol Rep 29(4):1652–1658 Montaner B, Perez-Tomas R, 2002, The cytotoxic prodigiosin induces phosphorylation of p38-MAPK but not of SAPK/JNK. Toxicol Lett 129(1–2):93–98 Yeung TM, Gandhi SC, Wilding JL, Muschel R, Bodmer WF, 2010, Cancer stem cells from colorectal cancer-derived cell lines. Proc Natl Acad Sci U S A 107(8):3722–3727 Kai K, Nagano O, Sugihara E, Arima Y, Sampetrean O, Ishimoto T, Nakanishi M, Ueno NT, Iwase H, Saya H, 2009, Maintenance of HCT116 colon cancer cell line conforms to a stochastic model but not a cancer stem cell model. Cancer Sci 100(12):2275–2282 Roy S, Majumdar AF, 2012, Signaling in colon cancer stem cells. J Mol Signal 7(1):11 Deng YH, Pu XX, Huang MJ, Xiao J, Zhou JM, Lin TY, Lin EH, 2010, 5-Fluorouracil upregulates the activity of Wnt signaling pathway in CD133-positive colon cancer stem-like cells. Chin J Cancer 29(9):810–815 Royds JA, Iacopetta B, 2006, p53 and disease: when the guardian angel fails. Cell Death Differ 13(6):1017–1026 Montaner B, Navarro S, Pique M, Vilaseca M, Martinell M, Giralt E, Gil J, Perez-Tomas R, 2000, Prodigiosin from the supernatant of Serratia marcescens induces apoptosis in hematopoietic cancer cell lines. Br J Pharmacol 131(3):585–593 Montaner B, Perez-Tomas R, 2001, Prodigiosin-induced apoptosis in human colon cancer cells. Life Sci 68(17):2025–2036 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 715 EP 723 DI 10.1007/s12253-016-0055-8 PG 9 ER PT J AU Foda, AMA El-Hawary, KA Hamed, H AF Foda, AlRahman Mohammad Abd El-Hawary, Kamal Amira Hamed, Hazem TI Aberrant Expression of Calretinin, D2–40 and Mesothelin in Mucinous and Non-Mucinous Colorectal Carcinomas and Relation to Clinicopathological Features and Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal; Mucinous; Calretinin; D2–40; Mesothelin ID Colorectal; Mucinous; Calretinin; D2–40; Mesothelin AB CRC is a heterogeneous disease in terms of morphology, invasive behavior, metastatic capacity, and clinical outcome. Recently, many so-called mesothelial markers, including calretinin, D2–40, WT1, thrombomodulin, mesothelin, and others, have been certified. The aim of this study was to assess the immunohistochemical expression of calretinin and other mesothelial markers (D2–40 and mesothelin) in colorectal mucinous adenocarcinoma (MA) and non mucinous adenocarcinoma (NMA) specimens and relation to clinicopathological features and prognosis using manual tissue microarray technique. We studied tumor tissue specimens from 150 patients with colorectal MA and NMA who underwent radical surgery from January 2007 to January 2012. High-density manual tissue microarrays were constructed using a modified mechanical pencil tip technique, and paraffin sections were submitted for immunohistochemistry using Calretinin, D2–40 and mesothelin expressions. We found that NMA showed significantly more calretinin and D2–40 expression than MA In contrast, no statistically significant difference between NMA and MA was detected in mesothelin expression. There were no statistically significant relations between any of the clinicopathological or histological parameters and any of the three markers. In a univariate analysis, neither calretinin nor D2–40 expressions showed any significant relations to DFS or OS. However, mesothelin luminal expression was significantly associated with worse DFS. Multivariate Cox regression analysis proved that luminal mesothelin expression was an independent negative prognostic factor in NMA. In conclusion, Calretinin, D2–40 and mesothelin are aberrantly expressed in a proportion of CRC cases with more expression in NMA than MA. Aberrant expression of these mesothelial markers was not associated with clinicopathological or histological features of CRCs. Only mesothelin expression appears to be a strong predictor of adverse prognosis. C1 [Foda, AlRahman Mohammad Abd] Mansoura University, Faculty of Medicine, Department of Pathology, 35516 Mansoura, Egypt. [El-Hawary, Kamal Amira] Mansoura University, Faculty of Medicine, Department of Pathology, 35516 Mansoura, Egypt. [Hamed, Hazem] Mansoura University, Faculty of Medicine, Department of Pathology, 35516 Mansoura, Egypt. RP El-Hawary, KA (reprint author), Mansoura University, Faculty of Medicine, Department of Pathology, 35516 Mansoura, Egypt. EM amira960@hotmail.com CR Ferlay J, Soerjomataram I, Ervik M, et al.: GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11 [Internet]. Lyon, France: International Agency for Research on Cancer 2013. http://globocan.iarc.fr. Accessed on 23 May 2015. Foda AA, El-Hawary AK, Abdel-Aziz A, 2013, Differential expression of matrix metalloproteinase- 13 in mucinous and nonmucinous colorectal carcinomas. Ann Diagn Pathol 17: 347–353 Hamilton SR, Bosman FT, Boffetta P, et al., 2010, Tumors of the colon and rectum. In: Bosman FT, Carneiro F, Hruban RH, et al., eds, WHO Classification of tumors of the digestive system. IARCPress, Lyon, pp. 132–182 Mekenkamp JM, Heesterbeek KJ, Koopman M, et, 2012, Mucinous adenocarcinomas: Poor prognosis in metastatic colorectal cancer. Eur J Cancer, 48: 501–509. Ordonez NG, 2005, D2–40 and podoplanin are highly specific and sensitive immunohistochemical markers of epithelioid malignant mesothelioma. Hum Pathol 205:372–380 Ordonez NG, 2006, Value of immunohistochemistry in distinguishing peritoneal mesothelioma from serous carcinoma of the ovary and peritoneum: a review and update. Adv Anat Pathol 13:16–25 Rogers JH, 1987, Calretinin: a gene for a novel calcium-binding protein expressed principally in neurons. J Cell Biol 105: 1343–1353 Gander JC, Gotzos V, Fellay B, et al., 1996, Inhibition of the proliferative cycle and apoptotic events in WiDr cells after downregulation of the calcium-binding protein calretinin using antisense oligodeoxynucleotides. Exp Cell Res 225:399–410 Miettinen M, Limon J, Niezabitowski A, et al., 2001, Calretinin and other mesothelioma markers in synovial sarcoma: analysis of antigenic similarities and differences withmalignant mesothelioma. Am J Surg Pathol 25:610–617 Lugli A, Forster Y, Haas P, et al., 2003, Calretinin expression in human normal and neoplastic tissues: a tissue microarray analysis on 5233 tissue samples. Hum Pathol. 34:994–1000 Dent R, Hanna WM, Trudeau M, et al., 2009, Pattern of metastatic spread in triple-negative breast cancer. Breast Cancer Res Treat 115: 423–428 Winn B, Tavares R, Fanion J, et al., 2009, Differentiating the undifferentiated: immunohistochemical profile of medullary carcinoma of the colon with an emphasis on intestinal differentiation. Hum Pathol 40:398–404 Chu AU, Litzky LA, Pasha TL, et al., 2005, Utility of D2–40, a novel mesothelial marker, in the diagnosis of malignant mesothelioma. Mod Pathol 18:105–110 Chang K, Pastan I, 1994, Molecular cloning and expression of a cDNA encoding a protein detected by the K1 antibody from an ovarian carcinoma, OVCAR-3, cell line. Int J Cancer 57:90–97 Ordonez NG, 2003, Application of mesothelin immunostaining in tumor diagnosis. Am J Surg Pathol 27:1418–1428 Cheng YW, Idrees K, Shattock R, et al., 2010, Loss of imprinting and marked gene elevation are 2 forms of aberrant IGF2 expression in colorectal cancer. Int J Cancer 127:568–577 Li H, Zhao P, Lu Y, 2012, Correlation of aberrant expression of CD133 with FHIT and malignant phenotype of colorectal adenocarcinoma. Int J Colorectal Dis 27:1015–1020 Lee SJ, Choi SY, Kim WJ, et al., 2013, Combined aberrant expression of E-cadherin and S100 A4, but not β-catenin is associated with disease-free survival and overall survival in colorectal cancer patients. Diagn Pathol 19(8):99 Foda AA, 2013, No-cost manual method for preparation of tissue microarrays having high quality comparable to semiautomated methods. Appl Immunohistochem Mol Morphol 21:271–274 Takeshima Y, Amatya VJ, Kushitani K, et al., 2008, A Useful antibody panel for differential diagnosis between peritoneal mesothelioma and ovarian serous carcinoma in japanese cases. Am J Clin Pathol 130:771–779 Ordonez NG, 2003, Value of mesothelin immunostaining in the diagnosis of mesothelioma. Mod Pathol 16(3):192–197 Hanna A, Pang Y, Bedrossian CW, et al., 2010, Podoplanin is a useful marker for identifying mesothelioma in malignant effusions. Diagn Cytopathol 38(4):264–269 Cargnello R, Celio MR, Schwaller B, et al., 1996, Change of calretinin expression in the human colon adenocarcinoma cell line HT29 after differentaition. Biochim Biophys Acta 1313:201–208 Taliano RJ, Lu S, Singh K, et al., 2013, Calretinin expression in high-grade invasive ductal carcinoma of the breast is associated with basal-like subtype and unfavorable prognosis. Hum Pathol 44(12):2743–2750 Gotzos V, Wintergerst ES, Musy JP, et al., 1999, Selective distribution of calretinin in adenocarcinomas of the human colon and adjacent tissues. Am J Surg Pathol 23:701–711 Purdie CA, Piris J, 2000, Histopathological grade, mucinous differentiation and DNA ploidy in relation to prognosis in colorectal carcinoma. Histopathol 36:121–126 Niv Y, 2000, Mucin and colorectal cancer. IMAJ 2:775–777 Kahn HJ, Marks A, 2002, A new monoclonal antibody, D2–40, for detection of lymphatic invasion in primary tumors. Lab Invest 82: 1255–1257 Raica M, CimpeanAM, RibattiD(2008, The Role of podoplanin in tumor progression and metastasis. Anticancer Res 28:2997–3006 Kawamata F, Homma S, Kamachi H, et al., 2014, C-ERC/ mesothelin provokes lymphatic invasion of colorectal adenocarcinoma. J Gastroenterol 49:81–92 Liebig B, Brabletz T, Staege MS, et al., 2005, Forced expression of deltaN-TCF-1B in colon cancer derived cell lines is accompanied by the induction of CEACAM5/6 and mesothelin. Cancer Lett 223: 159–167 Foda AA, El-Hawary AK, Aziz AA, 2015, Colorectal adenocarcinoma with mucinous component: relation of MMP-13, EGFR, and E-cadherin expressions to clinicopathological features and prognosis. APMIS., DOI 10.1111/apm.12379 Hennessy BT, Gonzalez-Angulo AM, Stemke-Hale K, et al., 2009, Characterization of a naturally occurring breast cancer subset enriched in epithelial-to-mesenchymal transition and stem cell characteristics. Cancer Res 69:4116–4124 Zhu QC, Gao RY, Wu W, et al., 2013, Epithelial-mesenchymal Transition and Its Role in the Pathogenesis of Colorectal Cancer. Asian Pacific J Cancer Prev 14(5):2689–2698 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 725 EP 732 DI 10.1007/s12253-016-0060-y PG 8 ER PT J AU Zhou, P Sun, L Liu, D Liu, Ch Sun, L AF Zhou, Peng Sun, Lixia Liu, Danfeng Liu, Changkuo Sun, Lei TI Long Non-Coding RNA lincRNA-ROR Promotes the Progression of Colon Cancer and Holds Prognostic Value by Associating with miR-145 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lincRNA-ROR; miR-145; Colon cancer; Oncogene ID lincRNA-ROR; miR-145; Colon cancer; Oncogene AB Large intergenic non-coding RNA ribonucleic acids-ROR (lincRNA-ROR) has been reported to exert impacts on the maintenance of induced pluripotent stem cells and embryonic stem cells, and play important roles in human hepatocellular cancer. It contributes to tumorigenesis and metastasis and functions as a competing endogenous RNA (ceRNA) by sponging miR-145 in breast cancer. However, its clinical significance and prognostic value in colon cancer remain unknown. The aim of the present study was to clarify the clinicopathological role and prognostic value of lincRNA-ROR and miR-145 in colon cancer. In the present study, qRT-PCR was performed to measure the expression levels of lincRNA-ROR in colon cancer tissues and cell lines. Then, the clinicopathological significance and prognostic value of lincRNA-ROR were analyzed. LincRNA-ROR expression correlated with pT stage, pN stage, AJCC stage and vascular invasion. Knockdown of lincRNA-ROR restored the expression of miR-145, and had a significant influence on colon cancer cell proliferation, migration and invasion. Patients of the high lincRNA-ROR/low miR-145 group had significantly poorer outcomes than those of the low lincRNA-ROR/high miR-145 group. Taken together, Overexpression of lincRNA-ROR combined with depletion of miR-145 may exert crucial impact on colon cancer prognosis evaluation and treatment. C1 [Zhou, Peng] The Second People’s Hospital of Wuhu, Department of General Surgery, 259 Jiuhua Middle Road, 241000 Wuhu, China. [Sun, Lixia] The Second People’s Hospital of Wuhu, Department of General Surgery, 259 Jiuhua Middle Road, 241000 Wuhu, China. [Liu, Danfeng] The Second People’s Hospital of Wuhu, Department of General Surgery, 259 Jiuhua Middle Road, 241000 Wuhu, China. [Liu, Changkuo] The Second People’s Hospital of Wuhu, Department of General Surgery, 259 Jiuhua Middle Road, 241000 Wuhu, China. [Sun, Lei] The Second People’s Hospital of Wuhu, Department of General Surgery, 259 Jiuhua Middle Road, 241000 Wuhu, China. RP Zhou, P (reprint author), The Second People’s Hospital of Wuhu, Department of General Surgery, 241000 Wuhu, China. EM doczhoupeng@163.com CR Brenner H, KloorM, Pox CP, 2014, Colorectal cancer. Lancet 383: 1490–1502 Sieqel R, Desantis C, Jemal A, 2014, Colorectal cancer statistics, 2014. CA Cancer J Clin 64:104–117 Edwards BK, Ward E, Kohler BA, Eheman C, Zauber AG, Anderson RN, et al., 2010, Annual report to the nation on the status of cancer, 1975-2006, featuring colorectal cancer trends and impact of interventions, risk factors, screening, and treatment, to reduce future rates. Cancer 116:544–573 Jankowski JA, Odze RD, 2009, Biomarkers in gastroenterology: between hope and hype comes histopathology. Am J Gastroenterol 104:1093–1096 Modjtahedi H, Essapen S, 2009, Epidermal growth factor receptor inhibitors in cancer treatment: advances, challenges and opporunities. Anti-Cancer Drugs 20:851–855 Aslam MI, Taylor K, Pringle JH, Jameson JS, 2009, MicroRNAs are novel biomarkers of colorectal cancer. Br J Surg 96:702–710 KiersemJB, Iksahl T, Lingjaerde OC, Guren T, TveitKM, Kure EH, 2014, Plasma microRNAs predicting clinical outcome in metastatic colorectal cancer patients receiving first-line oxaliplatin-based treatment. Mol Oncol 8:59–67 Yang F, Xue X, Bi J, Zheng L, Zhi K, Gu Y, et al., 2013, Long noncoding RNA CCAT1, which coild be activated by c-Myc, promotes the progression of gastric carcinoma. J Cancer Res Clin Oncol 139:437–445 Zhang EB, Yin DD, SunM, Kong R, Liu XH, You LH, et al., 2014, P53-regulated long non- coding RNA TUG1 affects cell proliferation in human non-small cell lung cancer, partly through epigenetically regulating HOXB7 expression. Cell Death Dis 5:e1243 Wu Y, Zhang L, Zhang L,Wang Y, Li H, Ren X, et al., 2015, Long non-coding RNA HOTAIR promotes tumor cell invasion and metastasis by recruiting EZH2 and repressing E-cadherin in oral squamous cell carcinoma. Int J Oncol 46:2586–2594 Wang L, Cai Y, Zhao X, Jia X, Zhang J, Liu J, et al., 2015, Downregulated long non-coding RNA H19 inhibits carcinogenesis of renal cell carcinoma. Neoplasma 62:412–418 Wapinski O, Chang HY, 2011, Long noncoding RNAs and human disease. Trends Cell Biol 21:354–361 Khalil AM, Guttman M, Huarte M, Garber M, Rai A, Rivea Morales D, et al., 2009, Many human large intergenic noncoding RNAs associate with chromatin modifying complexes and affect gene expression. Proc Natl Acad Sci U S A 106: 11667–11672 Guttman M, Rinn JL, 2012, Modular regulatory principles of large non-coding RNAs. Nature 482:339–346 Wang Y, Xu Z, Jiang J, Xu C, Kang J, Xiao L, et al., 2013, Endogenous miRNA sponge lincRNA-ROR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Dev Cell 25:69–80 Eades G, Wolfson B, Zhang Y, Li Q, Yao Y, Zhou Q, 2015, LincRNA-ROR and miR-145 regulate invasion in triple-negative breast cancer via targeting ARF6. Mol Cancer Res 13:330–338 Zhou X, Gao Q, Wang J, Zhang X, Liu K, Duan Z, 2014, Linc- RNA-ROR acts as a "spong" against mediation of the differentiation of endometrial cancer stem cells by microRNA-145. Gynecol Oncol 133:333–339 Iguchi T, Uchi R, Nambara S, Saito T, Komatsu H, Hirata H, et al., 2015, A long noncoding RNA, lncRNA-ATB, is involved in the progression and prognosis of colorectal cancer. Anticancer Res 35: 1385–1388 Xu TP, Huang MD, Xia R, Liu XX, Sun M, Yin L, et al., 2014, Decreased expression of the long non-coding RNA FENDRR is associated with poor prognosis in gastric cancer and FENDRR regulates gastric cancer cell metastasis by affecting fibronectin1 expression. J Hematol Oncol 7:63 Zhang ZZ, Shen ZY, Shen YY, Zhao EH,WangM,Wang CJ, et al., 2015, HOTAIR long noncoding RNA promotes gastric cancer metastasis through suppression of poly r(c)-binding protein, PCBP, 1. Mo Cancer Ther 14:1162–1170 Wu Y, Liu H, Shi X, Yao Y, YangW, Song Y, 2015, The long noncoding RNA HNF1A-AS1 regulates proliferation and metastasis in lung adenocarcinoma. Oncotarget 6:9160–9172 Lee H, Kim C, Ku JL, Kim W, Yoon SK, Kuh HJ, et al., 2014, A long non-coding RNA snaR contributes to 5- fluorouracil resistance in human colon cancer cells. Mol Cell 37:540–546 Loewer S, CabiliMN, Guttman M, Loh YH, Thomas K, Park IH, et al., 2010, Large intergenic non-coding RNA-ROR modulates reprogramming of human induced pluripotent stem cells. Nat Genet 42:1113–1117 Takahashi K, Yan IK, Koqure T, Haqa H, Patel T, 2014, Exreacellular vesicle-mediated transfer of long non-coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open Bio 4: 458–467 Hou P, Zhao Y, Li Z, Yao R, Ma M, Gao Y, et al., 2014, LincRNA-ROR induces epithelial-to-mesenchymal transition and contributes to breast cancer tumorigenesis and metastasis. Cell Death Dis 5:e1287 Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP, 2011, A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language? Cell 146:353–358 Tay Y, Kats L, Salmena L, Weiss D, Tan SM, Ala U, et al., 2011, Coding-independent regulation of the tumor suppressor PTEN by competing endogenous mRNAs. Cell 147:344–357 Yu CC, Tsai LL, Wang ML, Yu CH, Lo WL, Chang YC, et al., 2013, miR-145 targets the SOX9/ADAM17 axis to inhibit tumor-initiating cells and IL-6-mediated paracrine effects in head and neck cancer. Cancer Res 73:3425–3440 Noh JH, Chang YG, Kim MG, Jung KH, Kim JK, Bae HJ, et al., 2013, MiR-145 functions as a tumor suppressor by directly targeting histone deacetylase 2 in liver cancer. Cancer Lett 335:455–462 Zhang Y, Lin Q, 2015, MicroRNA-145 inhibits migration and invasion by down-regulating FSCN1 in lung cancer. Int J Clin Exp Med 8:8794–8802 Qin J,Wang F, Jiang H, Xu J, Jiang Y,Wang Z, 2015, MicroRNA- 145 suppresses cell migration and invasion by targeting paxillin in human colorectal cancer cells. Int J Clin Exp Pathol 8:1328–1340 Foltran L,Maglio GD, Pella N, Ermacora P, Aprile G,Masiero E, et al., 2015, Prognostic role of KRAS, NRAS, BRAF and PIK3CA mutations in advanced colorectal cancer. Future Oncol 11:629–640 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 733 EP 740 DI 10.1007/s12253-016-0061-x PG 8 ER PT J AU Popov, MT Stancheva, G Goranova, ET Rangachev, J Konov, D Todorov, S Stoyanov, O Kaneva, PR Popova, D AF Popov, M Todor Stancheva, Gergana Goranova, E Teodora Rangachev, Julian Konov, Dimitar Todorov, Spiridon Stoyanov, Orlin Kaneva, P Radka Popova, Diana TI Strong Correlation Between mRNA Expression Levels of HIF-2α, VEGFR1, VEGFR2 and MMP2 in Laryngeal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HIF-1α; HIF-2α; VEGFR1; VEGFR2; MMP2; Flt-1; Flk-1; Laryngeal carcinoma ID HIF-1α; HIF-2α; VEGFR1; VEGFR2; MMP2; Flt-1; Flk-1; Laryngeal carcinoma AB The hypoxia that arises due to the rapid proliferation of tumor cells is a fundamental driving force for the canonical pathway of neovascularization. In the current study we report a very strong correlation between mRNA expression levels of HIF-2α (but not HIF-1α), VEGFR-1, VEGFR-2 and MMP2 in ex vivo samples from laryngeal carcinoma. Sixty-three samples from patients with histopathologically verified carcinoma of the larynx were examined in this study. Total RNA was isolated from both normal and tumor fresh frozen tissues of each patient and real-time quantitative PCR reactions were performed. The mRNA expression levels of HIF-1α, HIF-2α, VEGFR1, VEGFR2 and MMP2 were acquired. We found strong positive correlations between mRNA expression levels of HIF-2α and VEGFR-1, rs(98) = .671, p < .0005; HIF-2α and VEGFR-2, rs(98) = .742, p < .0005; HIF-2α and MMP2, rs(98)= .566, p <.0005; VEGFR-1 and VEGFR-2, rs(98) = .791, p < .0005; VEGFR-1 and MMP2, rs(98)= .709, p <.0005; VEGFR-2 and MMP2, rs(98)= .793, p <.0005. Our results provide evidence for the regulatory connection between HIF-2α and VEGFR-1, VEGFR-2 and MMP2 in the light of ETS1/ HIF-2α regulatory axis on a non-in-vitro level in carcinoma tissue, uncover some of the differences between the homologues HIF-1α and HIF-2α and round up and support the results from different experimental models in this field. C1 [Popov, M Todor] Medical University of Sofia, Department of ENT, blvd., BG-1680 Sofia, Bulgaria. [Stancheva, Gergana] Medical University of Sofia, Molecular Medicine CenterSofia, Bulgaria. [Goranova, E Teodora] Medical University of Sofia, Molecular Medicine CenterSofia, Bulgaria. [Rangachev, Julian] Medical University of Sofia, Department of ENT, blvd., BG-1680 Sofia, Bulgaria. [Konov, Dimitar] Medical University of Sofia, Department of ENT, blvd., BG-1680 Sofia, Bulgaria. [Todorov, Spiridon] Medical University of Sofia, Department of ENT, blvd., BG-1680 Sofia, Bulgaria. [Stoyanov, Orlin] Medical University of Sofia, Department of ENT, blvd., BG-1680 Sofia, Bulgaria. [Kaneva, P Radka] Medical University of Sofia, Molecular Medicine CenterSofia, Bulgaria. [Popova, Diana] Medical University of Sofia, Department of ENT, blvd., BG-1680 Sofia, Bulgaria. RP Popov, MT (reprint author), Medical University of Sofia, Department of ENT, BG-1680 Sofia, Bulgaria. EM popov@todorpopov.com;tmpopov@abv.bg CR Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100:57–70 Fong GH, 2008, Mechanisms of adaptive angiogenesis to tissue hypoxia. Angiogenesis 11:121–140 KohMY, PowisG(2012, Passing the baton: the HIF switch. Trends Biochem Sci 37:364–372 Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N, 1989, Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 246:1306–1309 Shibuya M,Yamaguchi S, Yamane A, Ikeda T, Tojo A, Matsushime H, Sato M, 1990, Nucleotide sequence and expression of a novel human receptor-type tyrosine kinase gene, flt, closely related to the fms family. Oncogene 5:519–524 Park JE, Chen HH,Winer J, Houck KA, Ferrara N, 1994, Placenta growth factor. Potentiation of vascular endothelial growth factor bioactivity, in vitro and in vivo, and high affinity binding to Flt-1 but not to Flk-1/KDR. J Biol Chem 269:25646–25654 Fong GH, Rossant J, Gertsenstein M, Breitman ML, 1995, Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature 376:66–70 Robinson CJ, Stringer SE, 2001, The splice variants of vascular endothelial growth factor, VEGF, and their receptors. J Cell Sci 114:853–865 Quinn TP, Peters KG, de Vries C, Ferrara N, Williams LT, 1993, Fetal liver kinase-1 is a receptor for vascular endothelial growth factor and is selectively expressed in vascular endothelium. Proc Natl Acad Sci U S A 90:7533–7537 Popov TM, Goranova TE, Stancheva G et al, 2015, Relative quantitative expression of HIF-1α, HIF-2α, HIF-3α and VEGF-A in laryngeal carcinoma. Oncol Lett 9:2879–2885 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)). Methods 25:402–408 Scrideli CA, Carlotti CG Jr, Okamoto OK et al, 2008, Gene expression profile analysis of primary glioblastomas and non-neoplastic brain tissue: identification of potential target genes by oligonucleotide microarray and real-time quantitative PCR. J Neurooncol 88: 281–291 Borel F, Han R, Visser A et al, 2012, Adenosine triphosphatebinding cassette transporter genes up-regulation in untreated hepatocellular carcinoma is mediated by cellular microRNAs. Hepatology 55:821–832 Dutta D, Ray S, Vivian JL, Paul S, 2008, Activation of the VEGFR1 chromatin domain: an angiogenic signal-ETS1/HIF-2alpha regulatory axis. J Biol Chem 283:25404–25413 Hahne JC, Kummer S, Heukamp LC et al, 2009, Regulation of protein tyrosine kinases in tumour cells by the transcription factor Ets-1. Int J Oncol 35:989–996 Elvert G, Kappel A, Heidenreich R et al, 2013, Cooperative interaction of hypoxia-inducible factor-2alpha, HIF-2alpha , and Ets-1 in the transcriptional activation of vascular endothelial growth factor receptor-2, Flk-1). J Biol Chem 278:7520–7530 Taki M, Verschueren K, Yokoyama K, Nagayama M, Kamata N, 2006, Involvement of Ets-1 transcription factor in inducing matrix metalloproteinase-2 expression by epithelial-mesenchymal transition in human squamous carcinoma cells. Int J Oncol 28:487–496 Liu DX, Liu XM, Su Y, Zhang XJ, 2011, Renal expression of proto-oncogene Ets-1 on matrix remodeling in experimental diabetic nephropathy. Acta Histochem 113:527–533 Eubank TD, Roda JM, Liu H, O’Neil T, Marsh CB, 2011, Opposing roles for HIF-1α and HIF-2α in the regulation of angiogenesis by mononuclear phagocytes. Blood 117:323–332 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 741 EP 746 DI 10.1007/s12253-016-0059-4 PG 6 ER PT J AU Prabhu, B Sivakumar, A Sundaresan, S AF Prabhu, Bhoopathy Sivakumar, Annamalai Sundaresan, Sivapatham TI Diindolylmethane and Lupeol Modulates Apoptosis and Cell Proliferation in N-Butyl-N-(4-Hydroxybutyl) Nitrosamine Initiated and Dimethylarsinic Acid Promoted rat Bladder Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Apoptosis; Cell proliferation; Diindolylmethane; Lupeol and bladder cancer ID Apoptosis; Cell proliferation; Diindolylmethane; Lupeol and bladder cancer AB Bladder cancer has been shown to resist programmed cell death with altered expression of both proapoptotic and anti-apoptotic proteins. To study is to investigate the apoptotic properties of Diindolylmethane (DIM) and Lupeol on N-Butyl-N-(4-hydroxybutyl) Nitrosamine (BBN) initiated and Dimethylarsinic Acid (DMA) promoted urinary bladder cancer. Sixty male Wistar rats were divided into 6 groups. Group I: Control. Group II: Rats were experimentally developed bladder carcinogenesis with BBN and DMA. Group III and IV: DIM and lupeol were administered after BBN treatment for 28 weeks. Group V and VI: DIM and lupeol alone treatment for 36 weeks. All the experimental rats were maintained and euthanized after 36 weeks protocol. Urinary bladder tissues were collected and processed for further investigations. Apoptotis and cell proliferative marker such as Bax, Bcl-2, caspase-3, caspase-9 and PCNA were quantified using immunohistochemical analysis. The Immunohistochemical expression of Bax, Bcl-2, caspase-3, caspase-9 and PCNA were aberrant in BBN+DMA treated tumor group. Administration of DIM and lupeol inhibited the progression of bladder cancer, induced the expression of apoptotic Bax, caspase-3, caspase-9 and inhibited the expression of anti-apoptotic Bcl-2, PCNA in the urinary bladder of rats. Administration of diindolylmethane and lupeol treatment induces apoptosis and cellular proliferation by its anticarcinogenic properties. From our results DIM and lupeol would be the agent or adjunct for the treatment of bladder carcinogenesis. C1 [Prabhu, Bhoopathy] SRM University, SRM Medical College Hospital Research Centre, Department of Medical Research, Kanchipuram District, 603203 Tamil Nadu, India. [Sivakumar, Annamalai] SRM University, SRM Medical College Hospital Research Centre, Department of Medical Research, Kanchipuram District, 603203 Tamil Nadu, India. [Sundaresan, Sivapatham] SRM University, SRM Medical College Hospital Research Centre, Department of Medical Research, Kanchipuram District, 603203 Tamil Nadu, India. RP Sundaresan, S (reprint author), SRM University, SRM Medical College Hospital Research Centre, Department of Medical Research, 603203 Tamil Nadu, India. EM drssundaresan@hotmail.com CR Chavan S, Bray F, Lortet-Tieulent J, Jemal A, 2014, International variations in bladder cancer incidence and mortality. Eur Urol 66: 59–73 Botteman MF, Pashos CL, Redaelli A, 2015, The health economics of bladder cancer: a comprehensive review of the published literature. Pharm Economics 21:1315–30 Redondo-Gonzalez E, de Castro LN, Moreno-Sierra J, 2015). Bladder Carcinoma Data with Clinical Risk Factors and Molecular Markers: A Cluster Analysis. Biomed. Res. Int. 168682., DOI 10.1155/2015/168682. Elmore S, 2007, Apoptosis: A Review of Programmed Cell Death. Toxicol Pathol 35:495–516 Hassan M, Watari H, AbuAlmaaty A, Ohba Y, 2014). Apoptosis and Molecular Targeting Therapy in Cancer. Biomed. Res. Int. 150845., DOI 10.1155/2014/150845. Kim B, Srivastava SK, Sun SH, 2015, Caspase-9 as a therapeutic target for treating cancer. Expert Opin Ther Targets 19:11–127 Kirkin V, Joos S, Zornig M, 2004, The role of Bcl-2 family members in tumorigenesis. Biochim Biophys Acta 1644:229–249 Sylvester RJ,Van der Meijden AP, OosterlinckW(2006, Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials. Eur Urol 49:466–77 Leonardi E, Girlando S, SerioG(1992, PCNA and Ki67 expression in breast carcinoma: correlations with clinical and biological variables. J Clin Pathol 45:416–419 Al-Dhaheri WS, Hassouna I, Al-Salam S, Karam SM, 2008, Characterization of breast cancer progression in the rat. Ann N Y Acad Sci 1138:121–31 Bravo R, Frank R, Blundell PA, 1987, Cyclin/PCNA is the auxiliary protein of DNA polymerase. Nature 326:515–517 Garikapaty VP, Ashok BT, Tadi K, Mittelman A, Tiwari RK, 2006, 3,3′-Diindolylmethane downregulates prosurvival pathway in hormone independent prostate cancer. Biochem Biophys Res Commun 340:718–725 Nagaraj M, Sunitha S, Varalakshmi P, 2000, Effect of lupeol, a pentacyclic triterpene, on the lipid peroxidation and antioxidant status in rat kidney after chronic cadmium exposure. J Appl Toxicol 20:413–417 Hata K, Hori K, Ogasawara H, Takahashi S, 2003, Anti-leukemia activities of lup-28-al-20(29)-en-3-one, a lupane triterpene. Toxicol Lett 143:1–7 Saleem M, Afaq F, Adhami VM, Mukhtar H, 2004, Lupeol modulates NF-kappaB and PI3K/Akt pathways and inhibits skin cancer in CD-1 mice. Oncogene 23:5203–5214 Prabhu B, Balakrishnan D, Alwin D, Sundaresan S, 2014, Protective Effect of Diindolylmethane against N-Butyl-N-(4- hydroxybutyl, Nitrosamine-induced Bladder Carcinogenesis. J Exp Clin Med 6:132–138 Wanibuchi H, Yamamoto S, Chen H, 1996, Promoting effects of dimethylarsinic acid on N-butyl-N-(4-hydroxybutyl, nitrosamineinduced urinary bladder carcinogenesis in rats. Carcinogenesis 17: 2435–2439 Chen I, McDougal A, Wang F, 1998, Aryl hydrocarbon receptormediated antiestrogenic and antitumorigenic activity of diindolylmethane. Carcinogenesis 19:1631–1639 Sudharsan PT, Mythili Y, Selvakumar E, Varalakshmi P, 2005, Cardioprotective effect of pentacyclic triterpene, lupeol and its ester on cyclophosphamide-induced oxidative stress. Hum Exp Toxicol 24:313–318 Nakagawa K, Yamamura K, Maeda S, 1994, Bcl-2 expression in epidermal keratinocytic diseases. Cancer 74:1720–4 Lee JH, Khor TO, Shu L, 2013, Dietary phytochemicals and cancer prevention: Nrf2 signaling, epigenetics, and cell death mechanisms in blocking cancer initiation and progression. Pharmacol Ther 137: 153–171 McKnight JJ, Gray SB, O Kane HF, 2005, Apoptosis and chemotherapy for bladder cancer. J Urol 173:683–90 Inagaki T, Ebisuno S, Uekad Y, 1997, PCNA and p53 in urinary bladder cancer: correlation with histological findings and prognosis. Int J Urol 4:172–7 Marone M, Scambia G, Mozzetti S, 1998, Bcl-2, bax, bcl-XL, and bcl-XS expression in normal and neoplastic ovarian tissues. Clin Cancer Res 4:517–524 Safe S, Papineni S, Chintharlapalli S, 2008, Cancer chemotherapy with indole-3-carbinol, bis(3′-indolyl)methane and synthetic analogs. Cancer Lett 269:326–338 Rahman KM, 2007, Inactivation of NF-kappaB by 3,3′- diindolylmethane contributes to increased apoptosis induced by chemotherapeutic agent in breast cancer cells. Mol Cancer Ther 6:2757–2765 Cho HJ, Park SY, Kim EJ, 2011, 3,3′-Diindolylmethane inhibits prostate cancer development in the transgenic adenocarcinoma mouse prostate model. Mol Carcinogenesis 50:100–12 Kim YS, Milner JA, 2005, Targets for indole-3-carbinol in cancer prevention. J Nutr Biochem 16:65–73 Goldberg AA, Titorenko VI, Beach A, 2014, Ring-substituted analogs of 3,3′-diindolylmethane, DIM, induce apoptosis and necrosis in androgen-dependent and - independent prostate cancer cells. Invest New Drugs 32:25–36 Prasad S, Nigam N, Kalra N, Shukla Y, 2008, Regulation of signaling pathways involved in lupeol induced inhibition of proliferation and induction of apoptosis in human prostate cancer cells. Mol Carcinogenesis 47:916–24 SaleemM, Kaur S,KweonMH(2005, Lupeol, a fruit and vegetable based triterpene, induces apoptotic death of human pancreatic adenocarcinoma cells via inhibition of Ras signaling pathway. Carcinogenesis 26:1956–1964 Saleem M, Kweon MH, Yun JM, 2005, A novel dietary triterpene Lupeol induces fas-mediated apoptotic death of androgen-sensitive prostate cancer cells and inhibits tumor growth in a xenograft model. Cancer Res 65:11203–11213 Palanimuthu D, Baskaran N, Silvan S, Rajasekaran D, Manoharan S, 2012, Lupeol, a bioactive triterpene, prevents tumor formation during 7,12-dimethylbenz(a)anthracene induced oral carcinogenesis. Pathol Oncol Res 18(4):1029–37 Liu Y, Bi T, Wang G, 2015, Lupeol inhibits proliferation and induces apoptosis of human pancreatic cancer PCNA-1 cells through AKT/ERK pathways. Naunyn Schmiedebergs Arch Pharmacol 388:295–304 Nitta M, Azuma K, Hata K, 2013, Systemic and local injections of lupeol inhibit tumor growth in a melanoma-bearing mouse model. Biomed Rep 1:641–645 Prabhu B, Balakrishnan D, Sundaresan S, 2015, Antiproliferative and anti-inflammatory properties of diindolylmethane and lupeol against N-butyl-N-(4-hydroxybutyl, nitrosamine induced bladder carcinogenesis in experimental rats. Hum Exp Toxicol DOI., DOI 10.1177/0960327115597985 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 747 EP 754 DI 10.1007/s12253-016-0054-9 PG 8 ER PT J AU Sarosi, V Baliko, Z Smuk, G Laszlo, T Szabo, M Ruzsics, I Mezosi, E AF Sarosi, Veronika Baliko, Zoltan Smuk, Gabor Laszlo, Terezia Szabo, Mariann Ruzsics, Istvan Mezosi, Emese TI The Frequency of EGFR Mutation in Lung Adenocarcinoma and the Efficacy of Tyrosine Kinase Inhibitor Therapy in a Hungarian Cohort of Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NSCLC; Adenocarcinoma; EGFR mutation; TKI ID NSCLC; Adenocarcinoma; EGFR mutation; TKI AB In the last decades new therapeutic drugs have been developed for the treatment of non-small cell lung cancer (NSCLC) patients. Tyrosine kinase inhibitors (TKIs) significantly increase the progression free survival (PFS) of patients with NSCLC carrying epidermal growth factor receptor (EGFR) mutations. This type of lung cancer occurs mainly among non-smoking women and Asian origin. However, the new ESMO guideline recommends EGFR mutation analysis in every patient with NSCLC, because in patients with activating EGFR mutation, TKIs should be considered as first line therapy. In our recent work, we analyzed data of patients with EGFR-mutant adenocarcinoma from January 2009. The number of patients investigated was 446, among them 44 cases were positive for EGFR mutation. The ratio of positive cases was 9.86 % that is lower than the average mutation rate in Europe and much lower than that found in Asia. The exon 19 deletion was detected in 61.4 % of the patients, while L858R point mutation in exon 21 was observed in 34.1 % of them. In one subject, both exon 19 and 21 mutations were present simultaneously. A rare mutation located in exon 21 was found in another patient. TKI therapy was conducted in 38 patients. The disease control rate by TKI therapy was 85.7 %; primary resistance was documented in five subjects. Non-smoking patients with EGFR mutant adenocarcinoma had the highest benefit from TKI treatment. Our data support the recommendation that EGFR mutation status should be defined in all cases of locally advanced or metastatic lung adenocarcinoma. C1 [Sarosi, Veronika] University of Pecs, I. Department of Internal Medicine, 13 Ifjusag, H-7624 Pecs, Hungary. [Baliko, Zoltan] University of Pecs, I. Department of Internal Medicine, 13 Ifjusag, H-7624 Pecs, Hungary. [Smuk, Gabor] University of Pecs, Department of PathologyPecs, Hungary. [Laszlo, Terezia] University of Pecs, Department of PathologyPecs, Hungary. [Szabo, Mariann] University of Pecs, I. Department of Internal Medicine, 13 Ifjusag, H-7624 Pecs, Hungary. [Ruzsics, Istvan] University of Pecs, I. Department of Internal Medicine, 13 Ifjusag, H-7624 Pecs, Hungary. [Mezosi, Emese] University of Pecs, I. Department of Internal Medicine, 13 Ifjusag, H-7624 Pecs, Hungary. RP Mezosi, E (reprint author), University of Pecs, I. Department of Internal Medicine, H-7624 Pecs, Hungary. EM mezosi.emese@pte.hu CR Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JWW, Comber H, FormanD BF, 2013, Cancer incidence andmortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403 Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63:11–30 Auerbach O, Stout AP, Hammond EC, GarfinkelL, 1961, Changes in bronchial epithelium in relation to cigarette smoking and in relation to lung cancer. N Engl J Med 265:253–267 Chapman S, Robinson G, Stradling J, West S, 2009, Oxford Handbook of Respiratory Medicine, 2nd ed.). Chapter 31 Ryan DP, Chabner BA, 2000, On receptor inhibitors and chemotherapy. Clin Cancer Res 6:4607–4609 Burgess AW, Cho HS, Eigenbrot C, Ferguson KM, Garrett TP, Leahy DJ, Lemmon MA, Sliwkowski MX, Ward CW, Yokoyama S, 2003, An open-and-shut case? Recent insights into the activation of EGF/ErbB receptors. Mol Cell 12:541–552 Lemmon MA, 2009, Ligand-induced ErbB receptor dimerization. Exp Cell Res 315:638–648 Gan HK, Burgess AW, Clayton AH, Scott AM, 2012, Targeting of a conformationally exposed, tumor-specific epitope of EGFR as a strategy for cancer therapy. Cancer Res 72:2924–2930 Ciardiello F, Tortora G, 2008, EGFR antagonists in cancer treatment. N Engl J Med 358:1160–1174 Raymond E, Faivre S, Armand J, 2000, Epidermal growth factor receptor tyrosine kinase as a target for anticancer therapy. Drugs 60(Suppl 1):15–23 discussion 41-42 Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, OkimotoRA BBW, Harris PL, Haserlat SM, Supko JG, HaluskaFG LDN, Christiani DC, Settleman J, Haber DA, 2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350:2129– 2139 Minkovsky N, Berezov A, 2008, BIBW-2992, a dual receptor tyrosine kinase inhibitor for the treatment of solid tumors. Curr Opin Investig Drugs 9:1336–1346 FA S, Rodrigues Pereira J, Ciuleanu T, EH T, HirshV, Thongprasert S, Campos D, Maoleekoonpiroj S, Smylie M, Martins R, vanKooten M, Dediu M, Findlay B, D T, Johnston D, Bezjak A, Clark G, Santabarbara P, Seymour L, National Cancer Institute of Canada Clinical Trials Group, 2005, Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 353:123–132 Paez JG, Janne PA, Lee JC, Tracy S, Greulich H,Gabriel S,Herman P, Kaye FJ, Lindeman N, Boggon TJ, Naoki K, Sasaki H, Fujii Y, Eck MJ, Sellers WR, Johnson BE, Meyerson M, 2004, EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304:1497–1500 Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, Majem M, Lopez-Vivanco G, Isla D, Provencio M, Insa A, Massuti B, JL G-L, Paz-Ares L, Bover I, Garcia-Campelo R, MA M, Catot S, Rolfo C, Reguart N, Palmero R, JM S, Bastus R, Mayo C, Bertran-Alamillo J,MAM, JJ S, TaronM, Spanish Lung Cancer Group, 2009, Screening for epidermal growth factor receptor mutations in lung cancer. N Engl JMed 361:958–967 D’Angelo SP, Pietanza MC, Johnson ML, Riely GJ, Miller VA, Sima CS, Zakowski MF, Rusch VW, Ladanyi M, Kris MG, 2011, Incidence of EGFR exon 19 deletions and L858R in tumor specimens from men and cigarette smokers with lung adenocarcinomas. J Clin Oncol 29:2066–2070 Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, Sunpaweravong P, Han B, Margono B, Ichinose Y, Nishiwaki Y, Ohe Y, Yang JJ, Chewaskulyong B, Jiang H, Duffield EL,Watkins CL, Armour AA, Fukuoka M, 2009, Gefitinib or carboplatinpaclitaxel in pulmonary adenocarcinoma. N Engl J Med 361:947– 957 Fukuoka M,Wu YL, Thongprasert S, Sunpaweravong P, Leong SS, Sriuranpong V, Chao TY, Nakagawa K, Chu DT, Saijo N, Duffield EL, Rukazenkov Y, Speake G, Jiang H, Armour AA, To KF, Yang JC, Mok TS, 2011, Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia, IPASS). J Clin Oncol 29:2866–2874 Han JY, ParkK, KimSW, LeeDH, KimHY, KimHT, AhnMJ,Yun T, Ahn JS, Suh C, Lee JS, Yoon SJ, Han JH, Lee JW, Jo SJ, Lee JS, 2012, First-SIGNAL: first-line single-agent iressa versus gemcitabine and cisplatin trial in never-smokers with adenocarcinoma of the lung. J Clin Oncol 30:1122–1128 Janne PA,Wang X, SocinskiMA, Crawford J, Stinchcombe TE, Gu L, Capelletti M, Edelman MJ, Villalona-Calero MA, Kratzke R, Vokes EE, Miller VA, 2012, Randomized phase II trial of erlotinib alone or with carboplatin and paclitaxel in patients who were never or light former smokers with advanced lung adenocarcinoma: CALGB 30406 trial. J Clin Oncol 30:2063–2069 Zhou C,Wu YL, Chen G, Feng J, LiuXQ,Wang C, Zhang S,Wang J, Zhou S, Ren S, Lu S, Zhang L, Hu C, Hu C, Luo Y, Chen L, Ye M, Huang J, Zhi X, Zhang Y, Xiu Q, Ma J, Zhang L, You C, 2011, Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer, OPTIMAL, CTONG-0802): a multicentre, open-label, randomized, phase 3 study. Lancet Onco1 12:735–742 Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, Palmero R, Garcia-Gomez R, Pallares C, Sanchez JM, Porta R, Cobo M, Garrido P, Longo F, Moran T, Insa A, De Marinis F, Corre R, Bover I, Illiano A, Dansin E, de Castro J, Milella M, Reguart N, Altavilla G, Jimenez U, Provencio M, Moreno MA, Terrasa J, Munoz-Langa J, Valdivia J, Isla D, Domine M, Molinier O, Mazieres J, Baize N, Garcia-Campelo R, Robinet G, Rodriguez- Abreu D, Lopez-Vivanco G, Gebbia V, Ferrera-Delgado L, Bombaron P, Bernabe R, Bearz A, Artal A, Cortesi E, Rolfo C, Sanchez-Ronco M, Drozdowskyj A, Queralt C, de Aguirre I, Ramirez JL, Sanchez JJ, Molina MA, Taron M, Paz-Ares L, Spanish Lung Cancer Group in collaboration with Groupe Francais de Pneumo-Cancerologie and Associazione Italiana Oncologia Toracica, 2012, Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutationpositive non-small-cell lung cancer, EURTAC): a multicentre, open-label, randomized phase 3 trial. Lancet Oncol 13:239–246 Sequist LV, Yang JC, Yamamoto N, O’Byrne K, Hirsh V, Mok T, Geater SL, Orlov S, Tsai CM, Boyer M, SuWC, Bennouna J, Kato T, Gorbunova V, Lee KH, Shah R, Massey D, Zazulina V, Shahidi M, Schuler M, 2013, Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 31:3327–3334 Wu YL, Zhou C, Hu CP, Feng J, Lu S, Huang Y, LiW, Hou M, Shi JH, Lee KY, Xu CR, Massey D, Kim M, Shi Y, Geater SL, 2014, Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations, LUX-lung 6): an open-label, randomized phase 3 trial. Lancet Oncol 15:213–222 Yang JC,Wu YL, Schuler M, Sebastian M, Popat S, Yamamoto N, Zhou C, Hu CP, O'Byrne K, Feng J, Lu S, Huang Y, Geater SL, Lee KY, Tsai CM, Gorbunova V, Hirsh V, Bennouna J, Orlov S, Mok T, BoyerM, SuWC, Lee KH, Kato T, Massey D, ShahidiM, Zazulina V, Sequist LV, 2015, Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma, LUX-lung 3 and LUX-Lung6): analysis of overall survival data from two randomized, phase 3 trials. Lancet Oncol 16:141–151 Reck M, Popat S, Reinmuth N, De Ruysscher D, Kerr KM, Peters S, ESMO GuidelinesWorking Group, 2014, Metastatic non-smallcell lung cancer, NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 25 Suppl 3:iii27–39 Asano H, Toyooka S, Tokumo M, Ichimura K, Aoe K, Ito S, Tsukuda K, Ouchida M, Aoe M, Katayama H, HirakiA SK, Kiura K, Date H, Shimizu N, 2006, Detection of EGFR gene mutation in lung cancer by mutant-enriched polymerase chain reaction assay. Clin Cancer Res 12:43–48 Timar J, 2014, Criteria of the molecular pathology testing of lung cancer. Hung Oncology 58:139–142 Lohinai Z, HodaMA, Fabian K, et al., 2015, Distinct epidemiology and clinical consequences of classic versus rare EGFR mutations in lung adenocarcinoma. J Thorac Oncol 10:738–746 Gainor JF, Shaw AT, 2013, Emerging paradigms in the development of resistance to tyrosine kinase inhibitors in lung cancer. J Clin Oncol 31:3987–3996 Cortot AB, Janne PA, 2014)Molecular mechanisms of resistance in epidermal growth factor receptor-mutant lung adenocarcinomas. Eur Respir Rev 23:356–366 Wang SE, Narasanna A, Perez-TorresM, Xiang B,Wu FY, Yang S, Carpenter G, Gazdar AF, Muthuswamy SK, Arteaga CL, 2006, HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer Cell 10:25–38 Jackman DM, Miller VA, Cioffredi LA, Yeap BY, JannePA RGJ, Ruiz MG, Giaccone G, Sequist LV, Johnson BE, 2009, Impact of EGFR and KRAS mutations on clinical outcomes in previously untreated NSCLC patients: results of an online tumor registry of clinical trials. Clin Cancer Res 15:5267–5273 Ellis LM, Hicklin DJ, 2009, Resistance to targeted therapies: refining anticancer therapy in the era of molecular oncology. Clin Cancer Res 15:7471–7478 Spaans JN, Goss GD, 2014, Drug resistance to molecular targeted therapy and its consequences for treatment decisions in non-smallcell lung cancer. Front Oncol 4:190 Carrera S, Buque A, Azkona E, Aresti U, Calvo B, Sancho A, Arruti M, Nuno M, Rubio I, de Lobera AR, Lopez C, Vivanco GL, 2014, Epidermal growth factor receptor tyrosine-kinase inhibitor treatment resistance in non-small cell lung cancer: biological basis and therapeutic strategies. Clin Transl Oncol 16:339–350 Yang SH, 2013, Molecular basis of drug resistance: epidermal growth factor receptor tyrosine-kinase inhibitors and anaplastic lymphoma kinase inhibitors. Tuberc Respir Dis 75:188–198 Jackman D, Pao W, Riely GJ, Engelman JA, Kris MG, Janne PA, Lynch T, Johnson BE, Miller VA, 2010, Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol 28:357– 360 Pao W, Miller VA, Politi KA, Riely GJ, SomwarR ZMF, Kris MG, Varmus H, 2005, Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med 2:e73 Yun CH, Mengwasser KE, Toms AV, Woo MS, Greulich H, Wong KK, Meyerson M, Eck MJ, 2008, The T790 M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci U S A 105:2070–2075 Engelman JA, Zejnullahu K,Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, Kosaka T, Holmes AJ, Rogers AM, Cappuzzo F, Mok T, Lee C, Johnson BE, Cantley LC, Janne PA, 2007, MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316:1039–1043 Balak MN, Gong Y, Riely GJ, Somwar R, Li AR, Zakowski MF, Chiang A, Yang G, Ouerfelli O, Kris MG, Ladanyi M, Miller VA, Pao W, 2006, Novel D761Yand common secondary T790 M mutations in epidermal growth factor receptor-mutant lung adenocarcinomas with acquired resistance to kinase inhibitors. Clin Cancer Res 12:6494–6501 Bean J, Riely GJ, Balak M, Marks JL, Ladanyi M, Miller VA, Pao W, 2008, Acquired resistance to epidermal growth factor receptor kinase inhibitors associated with a novel T854 a mutation in a patient with EGFR-mutant lung adenocarcinoma. Clin Cancer Res 14:7519–7525 Costa DB, Halmos B, Kumar A, Schumer ST, Huberman MS, Boggon TJ, Tenen DG, Kobayashi S, 2007, BIM mediates EGFR tyrosine kinase inhibitor-induced apoptosis in lung cancers with oncogenic EGFR mutations. PLoS Med 4:1669–1679 discussion 1680 Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, Bergethon K, Shaw AT, Gettinger S, Cosper AK, Akhavanfard S, Heist RS, Temel J, Christensen JG, Wain JC, Lynch TJ, Vernovsky K, Mark EJ, Lanuti M, Iafrate AJ, Mino- Kenudson M, Engelman JA, 2011, Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med 3:75ra26 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 755 EP 761 DI 10.1007/s12253-016-0063-8 PG 7 ER PT J AU An, Ch Zhang, J Chu, H Gu, Ch Xiao, F Zhu, F Lu, R Shi, H Zhang, H Yi, X AF An, Chaolun Zhang, Jiajun Chu, Hongjun Gu, Chunyan Xiao, Feng Zhu, Fengwei Lu, Rujian Shi, Hai Zhang, Hongfei Yi, Xin TI Study of Gefitinib and Pemetrexed as First-Line Treatment in Patients with Advanced Non-Small Cell Lung Cancer Harboring EGFR Mutation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gefitinib; Pemetrexed; Advanced non-small cell lung cancer; EGFR mutation ID Gefitinib; Pemetrexed; Advanced non-small cell lung cancer; EGFR mutation AB To evaluate the efficacy and safety of a combination regimen of gefitinib and pemetrexed as first-line chemotherapy in advanced EGFR-mutated non-small cell lung cancer (NSCLC) patients. Patients and methods Patients with advanced non-squamous NSCLC harboring asensitive EGFR mutation were included in this study and randomly divided into gefitinib + placebo group and gefitinib + pemetrexed group. Pemetrexed or placebo was administered on day 1 at a dose of 500 mg/m2, and gefitinib was sequentially administered on days 2 ~ 16. This treatment regimen was repeated every 3 weeks until disease progression. All investigators and participants were masked to treatment allocation. The overall response rate (ORR) and disease control rate (DCR) of gefitinib + pemetrexed group were higher than that of gefitinib + placebo group but only the difference of DCR between two groups was statistically significant (P < 0.05). The median progression-free survival (PFS) of gefitinib + placebo group and gefitinib + pemetrexed group were 14.0 months vs. 18 months respectively and the difference was statistically significant (P < 0.05). The 2-year PFS rates of gefitinib + pemetrexed group (20.00 %) was higher than that of gefitinib + placebo group (8.89 %) and the difference was statistically significant (P < 0.05). The median overall survival (OS) of gefitinib + placebo group and gefitinib + pemetrexed group were 32.0 months vs. 34 months respectively and the difference was not statistically significant (P > 0.05). The 3-year OS rates of gefitinib + pemetrexed group (44.44 %) was higher than that of gefitinib + placebo group (35.56 %) but the difference was not statistically significant (P > 0.05). Major grade 3 or 4 hematological toxicities included neutropenia, leukopenia and anemia. The main grade 3 or 4 non-hematological toxicities were infection, increased alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, fatigue, diarrhea and pneumonitis. The difference of toxicities between two groups was not statistically significant (P > 0.05). The combination regimen of gefitinib + pemetrexed used in this study showed a higher ORR and DCR, longer median PFS and acceptable toxicity. C1 [An, Chaolun] The Third People’s Hospital of Nantong, Department of Cardiothoracic SurgeryJiangsu, China. [Zhang, Jiajun] Changhai Hospital of Shanghai, Department of Cardiothoracic SurgeryShanghai, China. [Chu, Hongjun] The Third People’s Hospital of Nantong, Department of Cardiothoracic SurgeryJiangsu, China. [Gu, Chunyan] The Third People’s Hospital of Nantong, Department of PathologyJiangsu, China. [Xiao, Feng] The Third People’s Hospital of Nantong, Department of PathologyJiangsu, China. [Zhu, Fengwei] The Third People’s Hospital of Nantong, Department of Cardiothoracic SurgeryJiangsu, China. [Lu, Rujian] The Third People’s Hospital of Nantong, Department of Cardiothoracic SurgeryJiangsu, China. [Shi, Hai] The Third People’s Hospital of Nantong, Department of Cardiothoracic SurgeryJiangsu, China. [Zhang, Hongfei] The Third People’s Hospital of Nantong, Department of Cardiothoracic SurgeryJiangsu, China. [Yi, Xin] Nantong University, Medical College, Department of Human Anatomy, 19 Qixiu Road, 226001 Nantong, Jiangsu Province, China. RP Yi, X (reprint author), Nantong University, Medical College, Department of Human Anatomy, 226001 Nantong, China. EM ntuyixin@foxmail.com CR Siegel R et al., 2012, Cancer treatment and survivorship statistics, 2012. CA Cancer J Clin 62(4):220–241 Nicholson RI, Gee JM, Harper ME, 2001, EGFR and cancer prognosis. Eur J Cancer 37(Suppl 4):S9–15 Onn A et al., 2004, Synchronous overexpression of epidermal growth factor receptor and HER2-neu protein is a predictor of poor outcome in patients with stage I non-small cell lung cancer. Clin Cancer Res 10(1 Pt 1):136–143 Lonardo F et al., 2002, Evidence for the epidermal growth factor receptor as a target for lung cancer prevention. Clin Cancer Res 8(1):54–60 Larsen AK et al., 2011, Targeting EGFR and VEGF(R, pathway cross-talk in tumor survival and angiogenesis. Pharmacol Ther 131(1):80–90 Lichtenberger BM et al., 2010, Autocrine VEGF signaling synergizes with EGFR in tumor cells to promote epithelial cancer development. Cell 140(2):268–279 Yatabe Y, Takahashi T, Mitsudomi T, 2008, Epidermal growth factor receptor gene amplification is acquired in association with tumor progression of EGFR-mutated lung cancer. Cancer Res 68(7):2106–2111 Barr S et al., 2008, Bypassing cellular EGF receptor dependence through epithelial-to-mesenchymal-like transitions. Clin Exp Metastasis 25(6):685–693 Gazdar AF et al., 2004, Mutations and addiction to EGFR: the Achilles ‘heal’ of lung cancers? Trends Mol Med 10(10):481–486 Pao W, Miller VA, 2005, Epidermal growth factor receptor mutations, small-molecule kinase inhibitors, and non-small-cell lung cancer: current knowledge and future directions. J Clin Oncol 23(11):2556–2568 Mok TS et al., 2009, Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 361(10):947–957 Mitsudomi T et al., 2010, Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor, WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 11(2):121–128 Inoue A et al., 2009, First-line gefitinib for patients with advanced non-small-cell lung cancer harboring epidermal growth factor receptor mutations without indication for chemotherapy. J Clin Oncol 27(9):1394–1400 Morita S et al., 2009, Combined survival analysis of prospective clinical trials of gefitinib for non-small cell lung cancer with EGFR mutations. Clin Cancer Res 15(13):4493–4498 Han JY et al., 2012, First-SIGNAL: first-line single-agent iressa versus gemcitabine and cisplatin trial in never-smokers with adenocarcinoma of the lung. J Clin Oncol 30(10):1122–1128 Paz-Ares L et al., 2010, Clinical outcomes in non-small-cell lung cancer patients with EGFR mutations: pooled analysis. J Cell Mol Med 14(1–2):51–69 Jackman D et al., 2010, Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in nonsmall- cell lung cancer. J Clin Oncol 28(2):357–360 Gainor JF, Shaw AT, 2013, Emerging paradigms in the development of resistance to tyrosine kinase inhibitors in lung cancer. J Clin Oncol 31(31):3987–3996 Goldberg SB et al., 2013, Chemotherapy with erlotinib or chemotherapy alone in advanced non-small cell lung cancer with acquired resistance to EGFR tyrosine kinase inhibitors. Oncologist 18(11): 1214–1220 Okabe T et al., 2008, Synergistic antitumor effect of S-1 and the epidermal growth factor receptor inhibitor gefitinib in non-small cell lung cancer cell lines: role of gefitinib-induced down-regulation of thymidylate synthase. Mol Cancer Ther 7(3):599–606 Chen CY et al., 2011, Thymidylate synthase and dihydrofolate reductase expression in non-small cell lung carcinoma: the association with treatment efficacy of pemetrexed. Lung Cancer 74(1): 132–138 Eisenhauer EA et al., 2009, New response evaluation criteria in solid tumours: Revised RECIST guideline, version 1.1). Eur J Cancer 45(2):228–247 Lynch TJ et al., 2004, Activatingmutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350(21):2129–2139 Paez JG et al., 2004, EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304(5676): 1497–1500 Maemondo M et al., 2010, Gefitinib or chemotherapy for nonsmall- cell lung cancer with mutated EGFR. N Engl J Med 362(25):2380–2388 Zhou C et al., 2011, Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer, OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 12(8):735–742 Rosell R et al., 2012, Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer, EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 13(3):239–246 Li TH et al., 2007, Schedule-dependent cytotoxic synergism of pemetrexed and erlotinib in human non-small cell lung cancer cells. Clin Cancer Res 13(11):3413–3422 Johnson BE et al., 2005, Preliminary phase II safety evaluation of ZD6474, in combination with carboplatin and paclitaxel, as 1(st)-line treatment in patients with NSCLC. J Clin Oncol 23(16):645s–645s Giaccone G et al., 2004, Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial-INTACT1. J Clin Oncol 22(5):777–784 Davies AM et al., 2009, Intermittent erlotinib in combination with pemetrexed phase I schedules designed to achieve Pharmacodynamic separation. J Thorac Oncol 4(7):862–868 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 763 EP 768 DI 10.1007/s12253-016-0067-4 PG 6 ER PT J AU Jo, SY Choi, RM Song, YS Kim, SM Yoo, JN Lee, HS AF Jo, Sol Yun Choi, Ryoung Mi Song, Yong Sang Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutations of HSPA4 and MED13 in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HSPA4; HSP70; MED13; Mutation; Cancers; Microsatellite instability ID HSPA4; HSP70; MED13; Mutation; Cancers; Microsatellite instability AB Frameshift mutation of genes containing mononucleotide repeats is a feature of gastric (GC) and colorectal cancers (CRC) with microsatellite instability (MSI). In the public genome database, we found that human HSPA4 gene encoding a heats hock protein 70 protein (HSP70–4) and MED13 gene had mononucleotide repeats in the coding sequences that could be targets for frameshift mutation in cancers with MSI. HSP70–4 is a member of HSP70 that is known to play a role in cell survival. MED13 is a member of MED genome-wide transcription regulators that function as a regulator for diverse biological processes. In this study, we analyzed the mutations in 79 GCs and 124 CRCs including high MSI (MSI-H) and microsatellite stable/low MSI (MSS/MSIL) cases by single-strand conformation polymorphism analysis and DNA sequencing. We found frameshift mutations of HSPA4 gene in two cancers (one GC and one CRC) and MED13 gene in the other two cancers (one GC and one CRC). The frameshift mutations were deletions of one base (c.2396delA (p.Asn799MetfsX50)) in HSPA4 and (c.2175delA (p.Lys725AsnfsX4)) in MED13. Each of HSPA4 and MED13 mutations were detected in GC with MSI-H (1/34: 2.9 %) and CRC with MSI-H (1/79: 1.3 %), but not in those with MSS. Our data show that unconventional HSPA4 and MED13 genes harbored frameshift mutations in GC and CRC with MSI. These mutations might possibly inactivate their functions and could be a feature of GC and CRC with MSI-H. C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Choi, Ryoung Mi] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Song, Yong Sang] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of PathologySeoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Khalil AA, Kabapy NF, Deraz SF, Smith C, 2011, Heat shock proteins in oncology: diagnostic biomarkers or therapeutic targets? Biochim Biophys Acta 1816:89–104 Saleh A, SrinivasulaSM, Balkir L, Robbins PD,Alnemri ES, 2000, Negative regulation of the Apaf-1 apoptosome by Hsp70. Nat Cell Biol 2:476–483 Aghdassi A, Phillips P, Dudeja V, Dhaulakhandi D, Sharif R, Dawra R, Lerch MM, Saluja A, 2007, Heat shock protein 70 increases tumorigenicity and inhibits apoptosis in pancreatic adenocarcinoma. Cancer Res 67:616–625 Arai E, Sakamoto H, Ichikawa H, Totsuka H, Chiku S, Gotoh M, Mori T, Nakatani T, Ohnami S, Nakagawa T, Fujimoto H,Wang L, Aburatani H, Yoshida T, Kanai Y, 2014, Multilayer-omics analysis of renal cell carcinoma, including thewhole exome,methylome and transcriptome. Int J Cancer 135:1330–1342 Poss ZC, Ebmeier CC, Taatjes DJ, 2013, The mediator complex and transcription regulation. Crit Rev Biochem Mol Biol 48:575–608 Makinen N, MehineM, Tolvanen J, Kaasinen E, LiY, Lehtonen HJ, Gentile M, Yan J, Enge M, Taipale M, Aavikko M, Katainen R, Virolainen E, Bohling T, Koski TA, Launonen V, Sjoberg J, Taipale J, Vahteristo P, Aaltonen LA, 2011)MED12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas. Science 334:252–255 LimWK, Ong CK, Tan J, Thike AA, Ng CC, Rajasegaran V, Myint SS, Nagarajan S, Nasir ND,McPherson JR, Cutcutache I, Poore G, Tay ST, Ooi WS, Tan VK, Hartman M, Ong KW, Tan BK, Rozen SG, Tan PH, Tan P, Teh BT, 2014, Exome sequencing identifies highly recurrentMED12 somatic mutations in breast fibroadenoma. Nat Genet 46:877–880 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8: 305–311 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis ofMED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 Turbett GR, Barnett TC, Dillon EK, Sellner LN, 1996, Single-tube protocol for the extraction of DNA or RNA from paraffinembedded tissues using a starch-based adhesive. Biotechniques 20:846–850 Makinen N, Heinonen HR, Sjoberg J, Taipale J, Vahteristo P, Aaltonen LA, 2014, Mutation analysis of components of the mediator kinase module in MED12 mutation-negative uterineleiomyomas. Br J Cancer 110:2246–2249 Turunen M, Spaeth JM, Keskitalo S, ParkMJ, Kivioja T, Clark AD, Makinen N, Gao F, Palin K, Nurkkala H, Vaharautio A, Aavikko M, Kampjarvi K, Vahteristo P, Kim CA, Aaltonen LA, Varjosalo M, Taipale J, Boyer TG, 2014, Uterine leiomyoma-linked MED12 mutations disrupt mediator-associated CDK activity. Cell Rep 7: 654–660 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 769 EP 772 DI 10.1007/s12253-016-0070-9 PG 4 ER PT J AU Ding, J Li, C Tang, J Yi, Ch Liu, JY Qiu, M AF Ding, Jing Li, Cong Tang, Jie Yi, Cheng Liu, Ji-Yan Qiu, Meng TI Higher Expression of Proteins in IGF/IR Axes in Colorectal Cancer is Associated with Type 2 Diabetes Mellitus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Diabetes mellitus; Insulin-like growth factor; Insulin-like growth factor receptor; Insulin receptor ID Colorectal cancer; Diabetes mellitus; Insulin-like growth factor; Insulin-like growth factor receptor; Insulin receptor AB Preexisting type 2 diabetes mellitus (preDM) increases occurrence and mortality of colorectal cancer (CRC). Insulin growth factor (IGF)/insulin receptor (IR) axes play an important role in the development of both diabetes and CRC. We aimed to explore the characteristics of proteins expression in IGF/IR axes in CRC tissues with preDM. Two hundred fifty CRC patients in West China hospital were included in analysis. Among them, 125 patients had history of diabetes matched by 125 CRC without diabetes at a 1:1 ratio. Immunohistochemical staining was used to detect the expression of proteins in IGF/IR axis. More positive expression of IGF-1, IGF-1R and IR were found in CRC group with diabetes than in non-diabetes group. No difference was detected in the expression of IR substrate-1, IR substrate-2, IGF-2, IGF binding protein 3, and mammalian target of rapamycin between two groups. Multivariate analysis showed that diabetes history was associated with all of the expression of IGF-1, IGF-1R and IR, and higher T staging and lymph node metastasis were respectively independent factors of IGF-1 and IGF-1R expression in CRC patients. Besides, IGF-1 expression was positively associated with IGF-1R and IR expression in all CRC tissues, and the association of IGF-1 and IR expression seemed to be closer in diabetes group than in non-diabetes group. Higher expression of IGF-1, IGF-1R and IR proteins in CRC was associated with diabetes, suggesting IGF-1/IR signaling may play a special part in development of CRC in patients with diabetes. C1 [Ding, Jing] West China Medical School, Sichuan University, Cancer Center, the State Key Laboratory of Biotherapy,West China Hospital, Department of Medical Oncology, No. 37, Guo Xue Xiang, 610041 Chengdu, Sichuan Province, China. [Li, Cong] West China Medical School, Sichuan University, Cancer Center, the State Key Laboratory of Biotherapy,West China Hospital, Department of Medical Oncology, No. 37, Guo Xue Xiang, 610041 Chengdu, Sichuan Province, China. [Tang, Jie] West China Medical School, Sichuan University, Cancer Center, the State Key Laboratory of Biotherapy,West China Hospital, Department of Medical Oncology, No. 37, Guo Xue Xiang, 610041 Chengdu, Sichuan Province, China. [Yi, Cheng] West China Medical School, Sichuan University, Cancer Center, the State Key Laboratory of Biotherapy,West China Hospital, Department of Medical Oncology, No. 37, Guo Xue Xiang, 610041 Chengdu, Sichuan Province, China. [Liu, Ji-Yan] West China Medical School, Sichuan University, Cancer Center, the State Key Laboratory of Biotherapy,West China Hospital, Department of Medical Oncology, No. 37, Guo Xue Xiang, 610041 Chengdu, Sichuan Province, China. [Qiu, Meng] West China Medical School, Sichuan University, Cancer Center, the State Key Laboratory of Biotherapy,West China Hospital, Department of Medical Oncology, No. 37, Guo Xue Xiang, 610041 Chengdu, Sichuan Province, China. RP Qiu, M (reprint author), West China Medical School, Sichuan University, Cancer Center, the State Key Laboratory of Biotherapy,West China Hospital, Department of Medical Oncology, 610041 Chengdu, China. EM qiumeng33@hotmail.com CR Xu CX, Zhu HH, Zhu YM, 2014, Diabetes and cancer: associations, mechanisms, and implications for medical practice. World J Diabetes 5(3):372–380 Joost HG, 2014, Diabetes and cancer: epidemiology and potential mechanisms. Diab Vasc Dis Res 11(6):390–394 Peeters PJ, Bazelier MT, Leufkens HG, de Vries F, De Bruin ML, 2015, The risk of colorectal cancer in patients with type 2 diabetes: associations with treatment stage and obesity. Diabetes Care 38(3): 495–502 Luo J, Lin HC, He K, HendryxM(2014, Diabetes and prognosis in older persons with colorectal cancer. Br J Cancer 110(7):1847– 1854 ZandersMM, Vissers PA, Haak HR, van de Poll-Franse LV, 2014, Colorectal cancer, diabetes and survival: epidemiological insights. Diabetes Metab 40(2):120–127 Mills KT, Bellows CF, Hoffman AE, Kelly TN, 2013, Gagliardi G, 2013, diabetes mellitus and colorectal cancer prognosis: a metaanalysis. Dis Colon Rectum 56(11):1304–1319 Chen KH, Shao YY, Lin ZZ, et al., 2014, Type 2 diabetes mellitus is associated with increased mortality in Chinese patients receiving curative surgery for colon cancer. Oncologist 19(9):951–958 Sehdev A, Shih YC, Vekhter B, Bissonnette MB, Olopade OI, Polite BN, 2015, Metformin for primary colorectal cancer prevention in patients with diabetes: a case-control study in a US population. Cancer 121(7):1071–1078 Cardel M, Jensen SM, Pottegard A, Jorgensen TL, Hallas J, 2014, Long-term use of metformin and colorectal cancer risk in type II diabetics: a population-based case-control study. Cancer Med 3(5): 1458–1466 Mei ZB, Zhang ZJ, Liu CY, et al., 2014, Survival benefits of metformin for colorectal cancer patients with diabetes: a systematic review and meta-analysis. PLoS One 9(3):e91818 Gallagher EJ, LeRoith D, 2010, The proliferating role of insulin and insulin-like growth factors in cancer. Trends EndocrinolMetab 21(10):610–618 Ding J, Tang J, Chen X, et al., 2013, Expression characteristics of proteins of the insulin-like growth factor axis in non-small cell lung cancer patients with preexisting type 2 diabetes mellitus. Asian Pac J Cancer Prev 14(10):5675–5680 Samani AA, Yakar S, LeRoith D, Brodt P, 2007, The role of the IGF system in cancer growth and metastasis: overview and recent insights. Endocr Rev 28(1):20–47 Liu R, Hu LL, Sun A, et al., 2014, mRNA expression of IGF-1 and IGF-1R in patients with colorectal adenocarcinoma and type 2 diabetes. Arch Med Res 45(4):318–324 Zhang H, Fagan DH, Zeng X, Freeman KT, Sachdev D, Yee D, 2010, Inhibition of cancer cell proliferation and metastasis by insulin receptor downregulation. Oncogene 29:2517–2527 Shu S, Yang Y, Li X, et al., 2011, Down-regulation of IGF-1R expression inhibits growth and enhances chemosensitivity of endometrial carcinoma in vitro. Mol Cell Biochem 353(1–2):225–233 LeRoith D, Roberts CT, 2003, The insulin-like growth factor system and cancer. Cancer Lett 195:127–137 Brahmkhatri VP, Prasanna C, Atreya HS, 2015, Insulin-like growth factor system in cancer: novel targeted therapies. Biomed Res Int 2015:538019 Rajpathak SN, Gunter MJ,Wylie-Rosett J, et al., 2009, The role of insulin-like growth factor-I and its binding proteins in glucose homeostasis and type 2 diabetes. Diabetes Metab Res Rev 25:3–12 Rinaldi S, Cleveland R, Norat T, et al., 2010, Serumlevels of IGF-I, IGFBP-3 and colorectal cancer risk: results from the EPIC cohort, plus a meta-analysis of prospective studies. Int J Cancer 126: 1702–1715 Yakar S, Nunez NP, Pennisi P, et al., 2006, Increased tumor growth in mice with dietinduced obesity: impact of ovarian hormones. Endocrinology 147:5826–5834 Ferguson R, Gallagher EJ, Cohen D, et al., 2013, Hyperinsulinemia promotes metastasis to the lung in a mouse model of Her2/Neumediated breast cancer. Endocr Relat Cancer 20:391–401 Shiratsuchi I, Akagi Y, Kawahara A, et al., 2011, Expression of IGF-1 and IGF-1R and their relation to clinicopathological factors in colorectal cancer. Anticancer Res 31(7):2541–2545 Wu Y, Yakar S, Zhao L, Hennighausen L, LeRoith D, 2002, Circulating insulin-like growth factor-i levels regulate colon cancer growth and metastasis. Cancer Res 62:1030–1035 Hakam A, Yeatman TJ, Lu L, et al., 1999, Expression of insulinlike growth factor-1 receptor in human colorectal cancer. Hum Pathol 30:1128–1133 Guler HP, Zapf J, Froesch ER, 1987, Short-term metabolic effects of recombinant human insulin-like growth factor I in healthy adults. N Engl JMed 317:137–140 Sakai K, Lowman HB, Clemmons DR, 2002, Increases in free, unbound insulin-like growth factor I enhance insulin responsiveness in human hepatoma G2 cells in culture. J Biol Chem 277: 13620–13627 Modan-Moses D, JanicotM,McLenithan JC, Lane MD, Casella SJ, 1998, Expression and function of insulin/insulin-like growth factor I hybrid receptors during differentiation of 3 T3-L1 preadipocytes. Biochem J 333(Pt 3):825–831 Xin C, Jing D, Jie T, Wu-Xia L, Meng Q, Ji-Yan L, 2015, The expression difference of insulin-like growth factor 1 receptor in breast cancers with or without diabetes. J Cancer Res Ther 11(2): 295–299 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 773 EP 779 DI 10.1007/s12253-016-0065-6 PG 7 ER PT J AU Stasikowska-Kanicka, O Wagrowska-Danilewicz, M Danilewicz, M AF Stasikowska-Kanicka, Olga Wagrowska-Danilewicz, Malgorzata Danilewicz, Marian TI Immunohistochemical Study EMT-Related Proteins in HPV-, and EBV-Negative Patients with Sinonasal Tumours SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Slug; Sinonasal inverted papilloma; Sinonasal cancer; E-cadherin; Fibronectin ID Slug; Sinonasal inverted papilloma; Sinonasal cancer; E-cadherin; Fibronectin AB Epithelial to mesenchymal transition (EMT) is a biological process in which the epithelial cells, transform to mesenchymal cells via multiple biochemical modifications. Immunohistochemical method was used to examine the expression of EMT-related proteins: Slug, E-cadherin and fibronectin, in 41 cases of sinonasal inverted papilloma (SIP), 33 cases of sinonasal squamous cell carcinoma (SNC), and 22 cases of normal mucosa as a control. In all cases negative viral status was previously confirmed using both in situ hybridization and immunohistochemical method. The immunoexpression of Slug and fibronectin were significantly increased in the SNC group as compared to SIPs and control cases. The immunoexpresssion of Slug was also higher in SIPs as compared to controls. The immunoexpression of Ecadherin was significantly lower in SNCs group as compared with SIPs and controls, but no statistically significant difference in E-cadherin immunoexpression was noted between SIPs and control cases. There were statistically significant negative correlations between immunoexpression of Slug vs E-cadherin, Ecadherin vs fibronectin and positive correlation between Slug vs fibronectin in SNC. Statistically significant correlation between Slug and fibronectin immunoexpression in SIPs was also found. In conclusion, our findings suggest that relationships between Slug, E-cadherin and fibronectin could potentially point to EMT in the sinonasal cancer. Lack of correlation between EMT-related proteins in tested SIPs could reflect a benign nature of those cases. C1 [Stasikowska-Kanicka, Olga] Medical University of Lodz, Department of NephropathologyLodz, Poland. [Wagrowska-Danilewicz, Malgorzata] Medical University of Lodz, Department of NephropathologyLodz, Poland. [Danilewicz, Marian] Medical University of Lodz, Department of NephropathologyLodz, Poland. RP Stasikowska-Kanicka, O (reprint author), Medical University of Lodz, Department of Nephropathology, Lodz, Poland. EM olgastasikowska@umed.lodz.pl CR Caplan LS, Hall I, Levine RS, et al., 2000, Preventable risk factors for nasal cancer. Ann Epidemiol 10:186–191 Rinaldo A, Ferlito A, Shaha AR, et al., 2002, Is elective neck treatment indicated in patients with squamous cell carcinoma of the maxillary sinus? Acta Otolaryngol 122:443–447 Zimmer LA, Carrau RL, 2006, Neoplasms of the nose and paranasal sinuses. In: Bailey BJ, Johnson JT, SD N, eds, Head & Neck Surgery - Otolaryngology, vol 417. 4th. Lippincott,Williams & Wilkins, Philadelphia, p. 1480 D’Errico A, Pasian S, Baratti A, et al., 2009, A case-controlled study on occupational risk factors for sinonasal cancer. Occup Environ Med 66:448–455 Batsakis JG, Suares P, 2001, Schneiderian papillomas and carcinomas: a review. Adv Anat Pathol 8:53–64 Mork J, Lie AK, Glattre E, et al., 2001, Human papillomavirus infection as a risk factor for squamous-cell carcinoma of the head and neck. N Engl J Med 344:1125–1131 Shen J, Tate JE, Crum CP, et al., 1996, Prevalence of human papillomavirus, HPV, in benign and malignant tumors of the upper respiratory tract. Mod Pathol 9:15–20 Barnes L, 2002, Schneiderian papillomas and nonsalivary glandular neoplasms of the head and neck. Mod Pathol 15(3):279–297 Von Buchwald C, Bradley PJ, 2007, Risks of malignancy in inverted papilloma of the nose and paranasal sinuses. Curr Opin Otolaryngol Head Neck Surg 15(2):95–98 Theys J, Jutten B, Habets R, et al., 2011, E-cadherin loss associated with EMT promotes radioresistance in human tumor cells. Radiother Oncol 99(3):392–397., DOI 10.1016/j.radonc.2011.05. 044 Kalluri R, Weinberg RA, 2009, The basics of epithelialmesenchymal transition. J Clin Invest 119:1420–1428 Zeisberg M, Neilson EG, 2009, Biomarkers for epithelialmesenchymal transitions. J Clin Invest 119:1429–1437 Tania M, Khan MA, Fu J, 2014, Epithelial to mesenchymal transition inducing transcription factors and metastatic cancer. Tumour Biol 35(8):7335–7342 Vered M, Dayan D, Yahalom R, et al., 2010, Cancer-associated fibroblasts and epithelial-mesenchymal transition in metastatic oral tongue squamous cell carcinoma. Int J Cancer 127(6):1356–1362 Shapiro L,Weis WI, 2009, Structure and biochemistry of cadherins and catenins. Cold Spring Harb Perspect Biol 1:a003053 Makrilia N, Kollias A, Manolopoulos L, et al., 2009, Cell adhesion molecules: role and clinical significance in cancer. Cancer Investig 27:1023–1037 Thiery JP, Acloque H, Huang RY, et al., 2009, Epithelialmesenchymal transitions in development and disease. Cell 139: 871–890 NietoMA, 2002, The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol 3:155–166 Barrallo-Gimeno A, Nieto MA, 2005, The snail genes as inducers of cell movement and survival: implications in development and cancer. Development 132:3151–3161 Peinado H, Olmeda D, CanoA, 2007, Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 7:415–428 Cobaleda C, Perez-Caro M, Vicente-Duenas C, et al., 2007, Function of the zinc-finger transcription factor SNAI2 in cancer and development. Annu Rev Genet 41:41–61 Come C, Magnino F, Bibeau F, et al., 2006, Snail and slug play distinct roles during breast carcinoma progression. Clin Cancer Res 12:5395–5402 Batlle E, Sancho E, Franci C, et al., 2000, The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol 2:84–89 Gaur N, Gandhi J, Robertson ES, et al., 2015, Epstein-Barr virus latent antigens EBNA3C and EBNA1 modulate epithelial to mesenchymal transition of cancer cells associated with tumor metastasis. Tumour Biol 36(4):3051–3060., DOI 10.1007/s13277- 014-2941-6 Horikawa T, Yoshizaki T, Kondo S, et al., 2011, Epstein-Barr virus latent membrane protein 1 induces snail and epithelialmesenchymal transition in metastatic nasopharyngeal carcinoma. Br J Cancer 104(7):1160–1167 Horikawa T, Yang J, Kondo S, et al., 2007, Twist and epithelialmesenchymal transition are induced by the EBVoncoprotein latent membrane protein 1 and are associated with metastatic nasopharyngeal carcinoma. Cancer Res 67(5):1970–1978 Kaul R,Murakami M, Choudhuri T, et al., 2007, Epstein-Barr virus latent nuclear antigens can induce metastasis in a nude mouse model. J Virol 81(19):10352–10361 Meng X, Wu X, Yuan Y, 2014, Significances of COX-2, p21, Ki- 67 expression and HPV infection in nasal inverted papilloma. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 28(23):1823–7. Abstract. Stasikowska O, Danilewicz M, Wagrowska-Danilewicz M, 2010, Human papillomavirus in sinonasal inverted papillomas and squamous cell carcinoma of the larynx. In situ hybridization with human papillomaviruses DNA probes. Period Biol 112(57):111–115 Major T, Szarka K, Sziklai I, et al., 2005, The characteristic of human papillomavirus DNA in head and neck cancer and papillomas. J Clin Pathol 58:51–55 Syrjanen KJ, 2003, HPV infection in benign and malignant sinonasal lesion. J Clin Pathol 56:174–181 Kim JY, Yoon JK, Citardi MJ, et al., 2007, The prevalence of human papilloma virus infection in sinonasal inverted papilloma specimens classified by histological grade. Am J Rhinol 21(6): 664–669 Pagano JS, 1999, Epstein-Barr virus: the first human tumor virus and its role in cancer. Proc Assoc Am Physicians 111(6):573–580 Christiansen JJ, Rajasekaran AK, 2006, Reassessing epithelial to mesenchymal transition as a prerequisite for carcinoma invasion and metastasis. Cancer Res 66(17):8319–8326 Barnes L, Everson JW, Reichart P, et al., 2005, World Health Organization classification of tumours. Pathology and Genetics Head and Neck Tumours IARC Press Lyon pp 15-17:28–32 Yanagawa J, Walser TC, Zhu LX, et al., 2009, SNAIL promotes CXCR2 ligand-dependent tumor progression in non-small cell lung carcinoma. Clin Cancer Res 15:6820–6829 Come C, Arnoux V, Bibeau F, et al., 2004, Roles of the transcription factors snail and slug during mammary morphogenesis and breast carcinoma progression. J Mammary Gland Biol Neoplasia 9(2):183–193 Jin H, Yu Y, Zhang T, et al., 2010, SNAIL is critical for tumor growth and metastasis of ovarian carcinoma. Int J Cancer 126: 2102–2111 Kosaka T, Kikuchi E,Mikami S, et al., 2010, Expression of SNAIL in upper urinary tract urothelial carcinoma: prognostic significance and implications for tumor invasion. Clin Cancer Res 16:5814– 5823 Yang MH, Chen CL, Chau GY, et al., 2009, Comprehensive analysis of the independent effect of twist and SNAIL in promoting metastasis of hepatocellular carcinoma. Hepatology 50:1464–1474 Christofori G, Semb H, 1999, The role of the cell-adhesion molecule E-cadherin as a tumour-suppressor gene. Trends Biochem Sci 24:73–76 Gilles C, Polette M, Zahm JM, et al., 1999, Vimentin contributes to human mammary epithelial cell migration. J Cell Sci 112:4615– 4625 Parry DA, 2006, Hendecad repeat in segment 2A and linker L2 of intermediate filament chains implies the possibility of a righthanded coiled-coil structure. J Struct Biol 155:370–374 Prasad CP, RathG, Mathur S, et al., 2009, Expression analysis of Ecadherin, SLUG and GSK3beta in invasive ductal carcinoma of breast. BMC Cancer 9:325 Cano A, Perez-Moreno MA, Rodrigo I, et al., 2000, The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol 2:76–83 Burk U, Schubert J, Wellner U, et al., 2008, A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep 9:582–589 Vesuna F, Van Diest P, Chen JH, et al., 2008, Twist is a transcriptional repressor of E-cadherin gene expression in breast cancer. Biochem Biophys Res Commun 367:235–241 Gasparotto D, Powesel J, Marzotto A, et al., 2011, Overexpression of TWIST2 correlates with poor prognosis in head and nech squamous cell carcinomas. Oncotarget 2:1165–1175 Hajra KM, Chen DY, Fearon ER, 2002, The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Res 15(62(6):):1613–1618 Yu Q, Zhang K, Wang X, et al., 2010, Expression of transcription factors snail, slug, and twist in human bladder carcinoma. J Exp Clin Cancer Res 1:29–119., DOI 10.1186/1756-9966-29-119 Savagner P, Yamada KM, Thiery JP, 1997, The zinc-finger protein slug causes desmosome dissociation, an initial and necessary step for growth factor-induced epithelial-mesenchymal transition. J Cell Biol 16;137(6):1403–1419 Hotz B, Arndt M, Dullat S, et al., 2007, Epithelial to mesenchymal transition: expression of the regulators snail, slug, and twist in pancreatic cancer. Clin Cancer Res 15;13(16): 4769–4776 Richter P, Umbreit C, Franz M et al, 2011, EGF/TGFβ1 costimulation of oral squamous cell carcinoma cells causes an epithelial-mesenchymal transition cell phenotype expressing laminin 332. J Oral Pathol Med 40(1):46–54., DOI 10.1111/j.1600-0714. 2010.00936.x. Epub 2010 Aug 31. Koo BS, Jung BJ, Kim SG, et al., 2011, Altered expression of Ecadherin and β-catenin in malignant transformation of sinonasal inverted papillomas. Rhinology 49(4):479–485., DOI 10. 4193/Rhino10.214 Lu SS, Xu JW, Huang KT, et al, 2007, Relationship between the biological behavior of nasal cavity or sinonasal inverted papilloma and infection of human papillomavirus. Zhonghua Yi Xue Za Zhi 87(19):1342–4, Abstract) NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 781 EP 788 DI 10.1007/s12253-016-0068-3 PG 8 ER PT J AU Chang, X Xu, X Xue, X Ma, J Li, Z Deng, P Chen, J Zhang, Sh Zhi, Y Dai, D AF Chang, Xiaojing Xu, Xiaoyang Xue, Xiaoying Ma, Jinguo Li, Zhenhua Deng, Peng Chen, Jing Zhang, Shuanglong Zhi, Yu Dai, Dongqiu TI NDRG1 Controls Gastric Cancer Migration and Invasion through Regulating MMP-9 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; NDRG1; Cell differentiation; MMP-9; Metastasis ID Gastric cancer; NDRG1; Cell differentiation; MMP-9; Metastasis AB The purpose of this study is to detect the clinical significance of NDRG1 and its relationship with MMP-9 in gastric cancer metastatic progression. 101 cases of gastric cancer specimens were utilized to identify the protein expression of NDRG1 and MMP-9 by immunohistochemistry, their clinical significance was also analyzed. The suppression by siRNA-NDRG1 was employed to detect the role of NDRG1 in gastric cancer progression and its relationship with MMP-9. NDRG1 expression was correlated inversely with the degree of tumor cell differentiation (p < 0.01), invasion depth (p < 0.05), lymph node metastasis (p < 0.05) and TNM stage (p < 0.05), whereas MMP-9 was positive correlated with the degree of tumor cell differentiation (p < 0.01), lymph node metastasis (p < 0.05) and TNM stage (p < 0.05), but not correlated with invasion depth (p>0.05). Furthermore, cell proliferation and invasion effect were remarkably enhanced when NDRG1 was silencing, but MMP-9 expression was increased. NDRG1 silencing enhances gastric cancer cells progression through upregulating MMP-9. It suggests that NDRG1 may inhibit the metastasis of gastric cancer via regulating MMP-9. C1 [Chang, Xiaojing] China Medical University, The Fourth Affiliated Hospital, Department of Gastrointestinal SurgeryShenyang, China. [Xu, Xiaoyang] China Medical University, The Fourth Affiliated Hospital, Cancer CenterShenyang, China. [Xue, Xiaoying] Hebei Medical University, The Second Hospital, Department of RadiotherapyShijiazhuang, China. [Ma, Jinguo] China Medical University, The Fourth Affiliated Hospital, Cancer CenterShenyang, China. [Li, Zhenhua] China Medical University, The Fourth Affiliated Hospital, Cancer CenterShenyang, China. [Deng, Peng] China Medical University, The Fourth Affiliated Hospital, Cancer CenterShenyang, China. [Chen, Jing] China Medical University, The Fourth Affiliated Hospital, Department of Gastrointestinal SurgeryShenyang, China. [Zhang, Shuanglong] China Medical University, The Fourth Affiliated Hospital, Cancer CenterShenyang, China. [Zhi, Yu] China Medical University, The Fourth Affiliated Hospital, Cancer CenterShenyang, China. [Dai, Dongqiu] China Medical University, The Fourth Affiliated Hospital, Department of Gastrointestinal SurgeryShenyang, China. RP Dai, D (reprint author), China Medical University, The Fourth Affiliated Hospital, Department of Gastrointestinal Surgery, Shenyang, China. EM daidq63@163.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Melotte V, Qu X, Ongenaert M, van Criekinge W, de Bruine AP, Baldwin HS, van EngelandM(2010, The N-myc downstream regulated gene, NDRG, family: diverse functions, multiple applications. FASEB J 24:4153–4166 Matsugaki T, Zenmyo M, Hiraoka K, Fukushima N, Shoda T, Komiya S, Ono M, Kuwano M, Nagata K, 2010, N-myc downstream- regulated gene1/Cap43 expression promotes cell differentiation of human osteosarcoma cells. Oncol Rep 24:721–725 Fotovati A, Abu-Ali S, Sugita Y, Nakamura Y, 2011, Expression of N-myc downstream regulated gene 1(NDRG1, in central neurocytoma. J Clin Neurosci 18:1383–1385 Maruyama Y, Ono M, Kawahara A, Yokoyama T, Basaki Y, Kage M, Aoyagi S, Kinoshita H, Kuwano M, 2006, Tumor growth suppression in pancreatic cancer by a putative metastasis suppressor gene Cap43/NDRG1/Drg-1 through modulation of angiogenesis. Cancer Res 66:6233–6242 Song Y, Oda Y, Hori M, Kuroiwa K, Ono M, Hosoi F, Basaki Y, Tokunaga S, Kuwano M, Naito S, Tsuneyoshi M, 2010, N-myc downstream regulated gene-1 Cap43 may play an important role in malignant progression of prostate cancer, in its close association with E-cadherin. Hum Pathol 41:214–222 Akiba J,Murakami Y, NodaM,Watari K, Ogasawara S, Yoshida T, Kawahara A, Sanada S, Yasumoto M, Yamaguchi R, Kage M, Kuwano M, Ono M, Yano H, 2011, N-myc downstream regulated gene 1/Cap43 overexpression suppresses tumor growth by hepatic cancer cells through cell cycle arrest at the G0/G1 phase. Cancer Lett 310(1):25–34 Stein S, Thomas EK, Herzog B, Westfall MD, Rocheleau JV, Jackson RS 2nd, Wang M, Liang P, 2004, NDRG1 is necessary for p53-dependent apoptosis. J Biol Chem 279:48930–48940 Lerner A, Grafi-Cohen M, Napso T, Azzam N, Fares F, 2012, The indolic diet-derivative, 3′3-diindolylmethane induced apoptosis in human colon cancer cells through upregulation of NDRG1. J Biomed Biotechnol 2012:256178 Chen Z, Zhang D, Yue F, Zheng M, Kovacevic Z, Richardson DR, 2012, The iron chelators Dp44mT and DFO inhibit TGF-β- induced epithelial-mesenchymal transition via up-regulation of Nmyc downstream-regulated gene 1, NDRG1). J Biol Chem 287: 17016–17028 Richardson A, Kovacevic Z, Richardson DR, 2013, Iron chelation: inhibition of key signaling pathways in the induction of the epithelial mesenchymal transition in pancreatic cancer and other tumors. Crit Rev Oncog 18(5):409–434 Frank B, HoffmeisterM, Klopp N, Illig T, Chang-Claude J, Brenner H, 2010, Single nucleotide polymorphisms in Wnt signaling and cell death pathway genes and susceptibility to colorectal cancer. Carcinogenesis 31:1381–1386 Liu W, Xing F, Iiizumi-Gairani M, Okuda H, Watabe M, Pai SK, Pandey PR, Hirota S, Kobayashi A, Mo YY, Fukuda K, Li Y, Watabe K, 2012, N-myc downstream regulated gene 1 modulates Wnt-β-catenin signaling and pleiotropically suppresses metastasis. EMBO Mol Med 4:93–108 Sun J, Zhang D, Zheng Y, Zhao Q, Zheng M, Kovacevic Z, Richardson DR, 2013, Targeting the metastasis suppressor, NDRG1, using novel iron chelators: regulation of stress fibermediated tumor cell migration via modulation of the ROCK1/ pMLC2 signaling pathway. Mol Pharmacol 83(2):454–469 Kachhap SK, Faith D, Qian DZ, Shabbeer S, Galloway NL, Pili R, Denmeade SR, DeMarzo AM, Carducci MA, 2007, The N-myc down regulated Gene1(NDRG1, is a Rab4a effector involved in vesicular recycling of E-cadherin. PLoS One 2:e844 Liu YL, Bai WT, Luo W, Zhang DX, Yan Y, Xu ZK, Zhang FL, 2011, Downregulation of NDRG1 promotes invasion of human gastric cancer AGS cells through MMP-2. Tumour Biol 32:99–105 Tandon A, Sinha S, 2011, Structural insights into the binding of MMP9 inhibitors. Bioinformation 5(8):310–314 Liu Z, Li L, Yang Z, LuoW, Li X, Yang H, Yao K,Wu B, FangW, 2010, Increased expression ofMMP9 is correlated with poor prognosis of nasopharyngeal carcinoma. BMC Cancer 10:270 Cheng J, Xie HY, Xu X,Wu J,Wei X, Su R, ZhangW, Lv Z, Zheng S, Zhou L, 2011, NDRG1 as a biomarker formetastasis, recurrence and poor prognosis in hepatocellular carcinoma. Cancer Lett 310(1):35–45 Sun B, Chu D, Li W, Chu X, Li Y, Wei D, Li H, 2009, Decreased expression of NDRG1 in glioma is related to tumor progression and survival of patients. J Neuro-Oncol 94(2):213–219 Al-Nasiry S, Geusens N, Hanssens M, Luyten C, Pijnenborg R, 2007, The use of Alamar blue assay for quantitative analysis of viability, migration and invasion of choriocarcinoma cells. Hum Reprod 22(5):1304–1309 Chung LC, Tsui KH, Feng TH, Lee SL, Chang PL, Juang HH, 2012, L-Mimosine block cell proliferation via upregulation of Bcell translocation gene 2 and N-myc downstream regulated gene 1 in prostate carcinoma cells. Am J Phys Cell Phys 302(4):C676– C685 Lai LC, Su YY, Chen KC, Tsai MH, Sher YP, Lu TP, Lee CY, Chuang EY, 2011, Down-regulation of NDRG1 promotes migration of cancer cells during Reoxygenation. PLoS One 6:e24375 Lane DJ, Saletta F, Suryo Rahmanto Y, Kovacevic Z, Richardson DR, 2013, N-myc downstream regulated 1, NDRG1, is regulated by eukaryotic initiation factor 3a, eIF3a, during cellular stress caused by iron depletion. PLoS One 8(2):e57273 Kovacevic Z, Sivagurunathan S, Mangs H, Chikhani S, Zhang D, Richardson DR, 2011, The metastasis suppressor, N-myc downstream regulated gene 1(NDRG1), upregulates p21 via p53- independent mechanisms. Carcinogenesis 32(5):732–740 Li S, Chen J, Yang Z, Lu G, Tang H, Hu H, 2008, N-myc downstream- regulated gene 1 as a downregulated target gene of PTEN in the controlling of tumourigenesis in endometrioid carcinoma. Indian J Med Res 127:453–459 Zhang J, Chen S, ZhangW, Zhang J, LiuX, ShiH, Che H,WangW, Li F, Yao L, 2008, Human differentiation-related gene NDRG1 is a myc downstream-regulated gene that is repressed by myc on the core promoter region. Gene 417:5–12 Sugiki T, Taketomi Y, Kikuchi-Yanoshita R, Murakami M, Kudo I, 2004, Association of N-myc downregulated gene1 with heatshock cognate protein 70 in mast cells. Biol Pharm Bull 27:628– 633 VM B, Medova M, Keogh A, Furer C, Zimmer Y, Candinas D, Stroka D, 2009, Hsp90 transcriptionally and post-translationally regulates the expression of NDRG1 and maintains the stability of modifying kinase GSK3beta. Biochim Biophys Acta 1793:1597– 1603 Lv XH, Chen JW, Zhao G, Feng ZZ, Yang DH, Sun WW, Fan JS, Zhu GH, 2012, N-myc downstream-regulated gene 1/Cap43 may function as tumor suppressor in endometrial cancer. J Cancer Res Clin Oncol 138(10):1703–1715 Matsushita K, Uchida K, Saigusa S, Ide S, Hashimoto K, Koike Y, Otake K, Inoue M, Tanaka K, Kusunoki M, 2013, Low NDRG1 mRNA expression predicts a poor prognosis in neuroblastoma patients. Pediatr Surg Int 29(4):363–368 Wang B, Li J, Ye Z, Li Z, Wu X, 2014, N-myc downstream regulated gene 1 acts as a tumor suppressor in ovarian cancer.Oncol Rep 31(5):2279–2285 Mao Z, Sun J, Feng B, Ma J, Zang L, Dong F, Zhang D, Zheng M, 2013, The metastasis suppressor, N-myc downregulated gene 1(NDRG1), is a prognostic biomarker for human colorectal cacner. PLoS One 8(7):e68206 Wang D, Tian X, Jiang Y, 2012, NDRG1/Cap43 overexpression in tumor tissues and serum from lung cancer patients. J Cancer Res Clin Oncol 138(11):1813–1820 Nagai MA, Gerhard R, Fregnani JH, Nonogaki S, Rierger RB, Netto MM, Soares FA, 2011, Prognostic value of NDRG1 and SPARC protein expression in breast cancer patients. Breast Cancer Res Treat 126:1–14 Chang X, Zhang S, Ma J, Li Z, Zhi Y, Chen J, Lu Y, Dai D, 2013, Association of NDRG1 gene promoter methylation with reduced NDRG1 expression in gastric cancer cells and tissue specimens. Cell Biochem Biophys 66(1):93–101 Chang X, Xu X, Ma J, Xue X, Li Z, Deng P, Zhang S, Zhi Y, Chen J,DaiD, 2014)NDRG1 expression is related to the progression and prognosis of gastric cancer patients through modulating proliferation, invasion and cell cycle of gastric cancer cells. Mol Biol Rep 41(9):6215–6223 Kawahara A, Akiba J, Hattori S, Yamaguchi T, Abe H, Taira T, Ureshino H, Murakami Y, Watari K, Koufuji K, Shirouzu K, Kuwano M, Ono M, Kage M, 2011, Nuclear expression of Nmyc downstream regulated gene 1/Ca(2+)-associated protein 43 is closely correlated with tumor angiogenesis and poor survival in patients with gastric cancer. Exp Ther Med 2(3):471–479 Jiang K, Shen Z, Ye Y, Yang X, Wang S, 2010, A novel molecular marker for early detection and evaluating prognosis of gastric cancer: N-myc downstream regulated gene-1, NDRG1). Scand J Gastroenterol 45(7–8):898–908 Carroll J, Field D, O’Connor PM, Cotter PD, Coffey A, Hill C, Ross RP, O’Mahony J, 2010)Gene encoded antimicrobial peptides, a template for the design of novel anti-mycobacterial drugs. Bioeng Bugs 1:408–412 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 789 EP 796 DI 10.1007/s12253-016-0071-8 PG 8 ER PT J AU Riquelme, I Tapia, O Espinoza, AJ Leal, P Buchegger, K Sandoval, A Bizama, C Araya, CJ Peek, MR Roa, CJ AF Riquelme, Ismael Tapia, Oscar Espinoza, A Jaime Leal, Pamela Buchegger, Kurt Sandoval, Alejandra Bizama, Carolina Araya, Carlos Juan Peek, M Richard Roa, Carlos Juan TI The Gene Expression Status of the PI3K/AKT/mTOR Pathway in Gastric Cancer Tissues and Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PI3K/AKT/mTOR pathway; Gastric cancer; AGS; MKN28 and MKN45 cell lines ID PI3K/AKT/mTOR pathway; Gastric cancer; AGS; MKN28 and MKN45 cell lines AB The PI3K/AKT/mTOR pathway plays a crucial role in the regulation of multiple cellular functions including cell growth, proliferation, metabolism and angiogenesis. Emerging evidence has shown that deregulation of this pathway has a role promoting gastric cancer (GC). The aim was to assess the expression of genes involved in this pathway by qPCR in 23 tumor and 23 non-tumor gastric mucosa samples from advanced GC patients, and in AGS, MKN28 and MKN45 gastric cancer cell lines. Results showed a slight overexpression of PIK3CA, PIK3CB, AKT1, MTOR, RPS6KB1, EIF4EBP1 and EIF4E genes, and a slightly decreased PTEN and TSC1 expression. In AGS, MKN28 and MKN45 cells a significant gene overexpression of PIK3CA, PIK3CB, AKT1, MTOR, RPS6KB1 and EIF4E, and a significant repression of PTEN gene expression were observed. Immunoblotting showed that PI3K-β, AKT, p-AKT, PTEN, mTOR, p-mTOR, P70S6K1, p- P70S6K1, 4E-BP1, p-4E-BP1, eIF4E and p-eIF4E proteins were present in cell lines at different levels, confirming activation of this pathway in vitro. This is the first time this extensive panel of 9 genes within PI3K/AKT/mTOR pathway has been studied in GC to clarify the biological role of this pathway in GC and develop new strategies for this malignancy. C1 [Riquelme, Ismael] Universidad de La Frontera, School of Medicine, Department of Pathological Anatomy, Avenida Alemania 0458, 4810296 Temuco, Chile. [Tapia, Oscar] Universidad de La Frontera, School of Medicine, Department of Pathological Anatomy, Avenida Alemania 0458, 4810296 Temuco, Chile. [Espinoza, A Jaime] Pontificia Universidad Catolica de Chile, Department of Pathology, Marcoleta 377, 7th Floor, 8330024 Santiago, Chile. [Leal, Pamela] Universidad de La Frontera, Scientific and Technological Bioresource Nucleus (BIOREN), Avenida Alemania 0458, 4810296 Temuco, Chile. [Buchegger, Kurt] Universidad de La Frontera, School of Medicine, Department of Pathological Anatomy, Avenida Alemania 0458, 4810296 Temuco, Chile. [Sandoval, Alejandra] Pontificia Universidad Catolica de Chile, School of Medicine, UC Centre for Investigational Oncology (CITO), Portugal 61, 8330034 Santiago, Chile. [Bizama, Carolina] Pontificia Universidad Catolica de Chile, Department of Pathology, Marcoleta 377, 7th Floor, 8330024 Santiago, Chile. [Araya, Carlos Juan] Universidad de La Frontera, School of Medicine, Department of Pathological Anatomy, Avenida Alemania 0458, 4810296 Temuco, Chile. [Peek, M Richard] Vanderbilt University, School of Medicine, Department of Medicine and Cancer Biology, 2215 Garland Avenue, 37232 Nashville, TN, USA. [Roa, Carlos Juan] Pontificia Universidad Catolica de Chile, Department of Pathology, Marcoleta 377, 7th Floor, 8330024 Santiago, Chile. RP Roa, CJ (reprint author), Pontificia Universidad Catolica de Chile, Department of Pathology, 8330024 Santiago, Chile. EM jcroa@med.puc.cl CR Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63:11–30 Brenner H, Rothenbacher D, Arndt V, 2009, Epidemiology of stomach cancer. Methods Mol Biol 472:467–477 Allum WH, Griffin SM, Watson A, Colin-Jones D, 2002, Guidelines for the management of oesophageal and gastric cancer. Gut 50:v1–v23 Nakajima T, 2002, Gastric cancer treatment guidelines in Japan. Gastric Cancer 5:1–5 Zhang W, 2014, TCGA divides gastric cancer into four molecular subtypes: implications for individualized therapeutics. Chin J Cancer 33:469–470 Al-Batran S-E, Ducreux M, Ohtsu A, 2012, mTOR as a therapeutic target in patients with gastric cancer. Int J Cancer 130:491–496 Fan Q-W, Weiss WA, 2010, Targeting the RTK-PI3K-mTOR axis in malignant glioma: overcoming resistance. Curr Top Microbiol Immunol 347:279–296 Matsuoka T, Yashiro M, 2014, The role of PI3K/Akt/mTOR signaling in gastric carcinoma. Cancers, Basel, 6:1441–1463 YangW, Raufi A, Klempner SJ, 2014, Targeted therapy for gastric cancer: molecular pathways and ongoing investigations. Biochim Biophys Acta 1846:232–237 Morgensztern D, McLeod HL, 2005, PI3K/Akt/mTOR pathway as a target for cancer therapy. Anti-Cancer Drugs 16:797–803 Caron E, Ghosh S, Matsuoka Y, Ashton-Beaucage D, Therrien M, et al., 2010, A comprehensive map of the mTOR signaling network. Mol Syst Biol 6:453 Polivka J, Janku F, 2014, Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacol Ther 142:164–175 Tapia O, Riquelme I, Leal P, Sandoval A, Aedo S, et al., 2014, The PI3K/AKT/mTOR pathway is activated in gastric cancer with potential prognostic and predictive significance. Virchows Arch 465: 25–33 Yap TA, Garrett MD, Walton MI, Raynaud F, de Bono JS, Workman P, 2008, Targeting the PI3K-AKT-mTOR pathway: progress, pitfalls, and promises. Curr Opin Pharmacol 8:393–412 Hay N, Sonenberg N, 2004, Upstream and downstream of mTOR. Genes Dev 18:1926–1945 Yang Q, Guan K-L, 2007, Expanding mTOR signaling. Cell Res 17:666–681 Dreesen O, Brivanlou AH, 2007, Signaling pathways in cancer and embryonic stem cells. Stem Cell Rev 3:7–17 Xiao L, Wang YC, Li WS, Du Y, 2009, The role of mTOR and phospho-p70S6K in pathogenesis and progression of gastric carcinomas: an immunohistochemical study on tissue microarray. J Exp Clin Cancer Res 28:152 Chen H, Guan R, Lei Y, Chen J, Ge Q, et al., 2015, Lymphangiogenesis in gastric cancer regulated through Akt/ mTOR-VEGF-C/VEGF-D axis. BMC Cancer 15:103 Greenfield LK, Jones NL, 2013, Modulation of autophagy by helicobacter pylori and its role in gastric carcinogenesis. Trends Microbiol 21:602–612 Liu JF, Zhou XK, Chen JH, Yi G, Chen HG, et al., 2010, Upregulation of PIK3CA promotes metastasis in gastric carcinoma. World J Gastroenterol 16:4986–4991 Ye B, Jiang L-L, Xu H-T, Zhou D-W, Li Z-S Expression of PI3K/ AKT pathway in gastric cancer and its blockade suppresses tumor growth and metastasis. Int J Immunopathol Pharmacol 25:627–636 Cinti C, Vindigni C, Zamparelli A, La Sala D, Epistolato MC, et al., 2008, Activated Akt as an indicator of prognosis in gastric cancer. Virchows Arch 453:449–455 Murayama T, Inokuchi M, Takagi Y, Yamada H, Kojima K, et al., 2009, Relation between outcomes and localisation of p-mTOR expression in gastric cancer. Br J Cancer 100:782–788 Lang SA, Gaumann A, Koehl GE, Seidel U, Bataille F, et al., 2007, Mammalian target of rapamycin is activated in human gastric cancer and serves as a target for therapy in an experimental model. Int J Cancer 120:1803–1810 Sun DF, jie ZY, XQ T, YX C, JY F, 2014, Inhibition of mTOR signalling potentiates the effects of trichostatin a in human gastric cancer cell lines by promoting histone acetylation. Cell Biol Int 38: 50–63 Yang HY, Xue LY, Xing LX, Wang J, Wang JL, et al., 2013, Putative role of the mTOR/4E-BP1 signaling pathway in the carcinogenesis and progression of gastric cardiac adenocarcinoma. Mol Med Rep 7:537–542 Fan S, Ramalingam SS, Kauh J, Xu Z, Khuri FR, Sun S-Y, 2014, Phosphorylated eukaryotic translation initiation factor 4, eIF4E, is elevated in human cancer tissues. Cancer Biol Ther 8:1463–1469 Liang S, Guo R, Zhang Z, Liu D, Xu H, et al., 2013, Upregulation of the eIF4E signaling pathway contributes to the progression of gastric cancer, and targeting eIF4E by perifosine inhibits cell growth. Oncol Rep 29:2422–2430 Chen C-N, Hsieh F-J, Cheng Y-M, Lee P-H, Chang K-J, 2004, Expression of eukaryotic initiation factor 4E in gastric adenocarcinoma and its association with clinical outcome. J Surg Oncol 86: 22–27 Wen Y-G, Wang Q, Zhou C-Z, Qiu G-Q, Peng Z-H, Tang H-M, 2010, Mutation analysis of tumor suppressor gene PTEN in patients with gastric carcinomas and its impact on PI3K/AKT pathway. Oncol Rep 24:89–95 Kang Y-H, Lee HS, Kim WH, 2002, Promoter methylation and silencing of PTEN in gastric carcinoma. Lab Investig 82:285–291 Schwanhausser B, Busse D, Li N, Dittmar G, Schuchhardt J, et al., 2011, Global quantification of mammalian gene expression control. Nature 473:337–342 The Cancer Genome Research Network, 2014, Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513(7517):202–209 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 797 EP 805 DI 10.1007/s12253-016-0066-5 PG 9 ER PT J AU Ruiz-Sauri, A Valencia-Villa, G Romanenko, A Perez, J Garcia, R Garcia, H Benavent, J Sancho-Tello, M Carda, C LLombart-Bosch, A AF Ruiz-Sauri, Amparo Valencia-Villa, Gerardo Romanenko, Alina Perez, Jesus Garcia, Raul Garcia, Heydi Benavent, Jose Sancho-Tello, Maria Carda, Carmen LLombart-Bosch, Antonio TI Influence of Exposure to Chronic Persistent Low-Dose Ionizing Radiation on the Tumor Biology of Clear-Cell Renal-Cell Carcinoma. An Immunohistochemical and Morphometric Study of Angiogenesis and Vascular Related Factors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Clear-cell renal-cell carcinoma (CCRCC); Chronic persistent low-dose ionizing radiation (CPLDIR); Angiogenesis; Morphometry; Angiogenic factors; Microvessel density (MVD); Immunohistochemistry ID Clear-cell renal-cell carcinoma (CCRCC); Chronic persistent low-dose ionizing radiation (CPLDIR); Angiogenesis; Morphometry; Angiogenic factors; Microvessel density (MVD); Immunohistochemistry AB Increased angiogenesis is related to boosted growth and malignancy in carcinomas. "Chronic Persistent Low-Dose Ionizing Radiation" (CPLDIR) exposure increases incidence and aggressive behavior of clear-cell renal-cell carcinoma (CCRCC). The aim was to study the biology of angiogenesis, including microvessel density (MVD), in human clear-cell renalcell carcinomas (CCRCC) originating from a radio-contaminated geographical area (Ukraine) and to compare with similar tumors diagnosed in non-contaminated regions of Europe (Spain, Valencia) and Latin America (Colombia, Barranquilla). MVD was comparatively examined in 124 patients diagnosed with CCRCC from three geographical areas by means of digital micro-imaging and computerized analysis. Additionally, 50 adult normal kidneys were used for controls (autopsy kidneys from Valencia and Barranquilla). Furthermore, an immunohistochemical study of several vascular related growth factors was undertaken using a similar methodology. MVD as well as VEFG are the most discriminating factors associated with an aggressive behavior of CCRCC. Their expression increased in proportion to the level of exposure to chronic low-dose ionizing radiation in Ukrainian patients in the 25 years since the Chernobyl accident substantiated by comparison with the two control groups of renal carcinomas present in non-irradiated areas (Spain and Colombia). No major biological differences relating to angiogenesis appear to exist between the CCRCC diagnosed in two distant geographical areas of the world. HIF-1α expression was similar in all groups, with no statistical significance. Present findings demonstrate the existence of a significant relationship between MVD and VEGF in CCRCC: an increased expression of VEGF is associated with a high level of angiogenesis. C1 [Ruiz-Sauri, Amparo] University of Valencia, Department of Pathology, Avenida Blasco Ibanez, 15, 46010 Valencia, Spain. [Valencia-Villa, Gerardo] University of Valencia, Department of Pathology, Avenida Blasco Ibanez, 15, 46010 Valencia, Spain. [Romanenko, Alina] Academy of Medical Sciences of Ukraine, Institute of Urology, Department of PathologyKiev, Ukraine. [Perez, Jesus] Universidad Libre, Medical SchoolBarranquilla, Colombia. [Garcia, Raul] Universidad del Norte, Medical SchoolBarranquilla, Colombia. [Garcia, Heydi] Department of legal medicineBarranquilla, Colombia. [Benavent, Jose] University of Valencia, Department of Pathology, Avenida Blasco Ibanez, 15, 46010 Valencia, Spain. [Sancho-Tello, Maria] University of Valencia, Department of Pathology, Avenida Blasco Ibanez, 15, 46010 Valencia, Spain. [Carda, Carmen] University of Valencia, Department of Pathology, Avenida Blasco Ibanez, 15, 46010 Valencia, Spain. [LLombart-Bosch, Antonio] University of Valencia, Department of Pathology, Avenida Blasco Ibanez, 15, 46010 Valencia, Spain. RP Ruiz-Sauri, A (reprint author), University of Valencia, Department of Pathology, 46010 Valencia, Spain. EM Amparo.Ruiz-Sauri@uv.es CR Jemal A, Siegel R,Ward E,HaoY, Xu J,Murray T, ThunMJ, 2008, Cancer statistics, 2008. CA Cancer J Clin 58(2):71–96 www.seom.org.es, Sociedad Espanola de Oncologia Medica. Janzen NK, Kim HL, Figlin RA, Belldegrun AS, 2003, Surveillance after radical or partial nephrectomy for localized renal cell carcinoma and management of recurrent disease. Urol Clin North Am 30:843–852 Bergers G, Benjamin LE, 2003, Tumorigenesis and the angiogenic switch. Nat Rev Cancer 3:401–410 Kerbel RS, 2008, Tumor angiogenesis. N Engl J Med 358(19): 2039–2049 ZhangY, Jiang X,Qin X, Ye D,Yi Z, LiuM, Bai O, Fang J, Chen Y, 2010, RKTG inhibits angiogenesis by suppressing MAPK- mediated autocrine VEGF signaling and is down regulated in clear-cell renal cell carcinoma. Oncogene 29:5404–5415 Yhee JY, Yu CH, Kim JH, Im KS, Brodersen BW, Doster AR, Sur HJ, 2012, Angiogenesis and expression of vascular endothelial growth factor, tumour necrosis factor-alpha and hypoxia inducible factor-1alpha in canine renal cell carcinoma. J Comp Pathol 147: 129–138 Dewhirst MW, Cao Y, Moeller B, 2008, Review cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nat Rev Cancer 8(6):425–437 Vaupel P, 2008, Review hypoxia and aggressive tumor phenotype: implications for therapy and prognosis. Oncologist 13(3):21–26 Yang MH,Wu MZ, Chiou SH, Chen PM, Chang SY, Liu CJ, Teng SC, Wu KJ, 2008, Direct regulation of TWIST by HIF-1alpha promotes metastasis. Nat Cell Biol 10(3):295–305 www.nap.edu/. The National Academies Press. Health Risks from Exposure to Low Levels of Ionizing Radiation: BEIR VII Phase 2, 2006). Cap. 2 Molecular and Cellular Responses to Ionizing Radiation, 43–64). Clarke RH, 2001, Control of low-level radiation exposure: what is the problem and how can it be solved? Health Phys 80:391–396 Christensen R, Alsner J, Brandt Sorensen F, Dagnaes-Hansen F, Kolvraa S, Serakinci N, 2008, Transformation of human mesenchymal stem cells in radiation carcinogenesis: long-term effect of ionizing radiation. Regen Med 3(6):849–861 Mullenders L, Atkinson M, Peretzke H, Sabatier L, Bouffler S, 2009, Assessing cancer risks of low-dose radiation. Nat Rev Cancer 9(8):596–604 Romanenko A, Morell-Quadreny L, Nepomnyaschy V, Vozianov A, Llombart-Bosch A, 2000, Pathology and proliferative activity of renal-cell carcinomas and renal oncocytomas in patients with different radiation exposure after the Chernobyl accident in Ukraine. Int J Cancer 87:880–888 Romanenko A, Morell-Quadreny L, Nepomnyaschy V, Vozianov A, Llombart-Bosch A, 2001, Radiation sclerosing proliferative atypical nephropathy of peritumoral tissue of renal-cell carcinomas after the Chernobyl accident in Ukraine. Virchows Arch 438:146– 153 Romanenko A, Morell-Quadreny L, Lopez-Guerrero JA, Pellin A, Nepomnyaschy V, Vozianov A, Llombart-Bosch A, 2004, The INK4a/ARF locus: role in cell cycle control for renal cell epithelial tumor growth after the Chernobyl accident. Virchows Arch 445: 298–304 Romanenko A, Morell-Quadreny L, Ramos D, Vozianov A, Llombart-Bosch A, 2006, Alteration of apoptotic regulatory molecules in conventional renal cell carcinoma influenced by chronic long-term low-dose radiation exposure in humans revealed by tissue microarray. Cancer Genomics Proteomics 3:107–112 Romanenko A, Morell-Quadreny L, Ramos D, Nepomnyaschy V, Vozianov A, Llombart-Bosch A, 2006, Extracellular matrix alterations in conventional renal cell carcinomas by tissue microarray profiling influenced by persistent, long-term, low-dose ionizing radiation exposure in humans. Virchows Arch 448:584–590 Romanenko AM, Ruiz-Sauri A, 2012, Morell-Quadreny L, Valencia G, Vozianov AF, Llombart-Bosch a. Microvessel density is high in clear-cell renal cell carcinomas of Ukrainian patients exposed to chronic persistent low-dose ionizing radiation after the Chernobyl accident. Virchows Arch 460(6):611–619 American Joint Committee on Cancer, AJCC, Cancer. Staging Manual. 7° Edicion. Nueva York: Springer; 2010. Fuhrman SA, Lasky LC, Limas C, 1982, Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol 6(7):655–663 Thoenes W, Storkel ST, Rompelt HJ, 1986, Histopathology and classification of renal cell tumors, adenomas, oncocytomas and carcinomas): the basic cytological and histopathological elements and their use for diagnostic. Pathol Res Pract 181(2):125–143 Folkman J, 1995, Seminars in medicine of the Beth Israel hospital Boston. Clinical applications of research on angiogenesis. N Engl J Med 333:1757–1763 Weidner N, Semple JP, Welch WR, Folkman J, 1991, Tumor angiogenesis and metastasis-correlation in invasive breast carcinoma. N Engl JMed 324:1–8 Chow WH, Devesa SS, Warren JL, Fraumeni JF, 1999, Rising incidence of renal cell cancer in the United States. JAMA 281: 1628–1631 Saydackova NA, Starceva LM, Kravchuk NC, 2007, The state of urological assistance for the population in Ukraine. Annual report. Ministry of Health in Ukraine, Kiev, pp. 146–153 Bazyka DA, Ye PA, AY R, ZP F, NA G, MM F, OM K, NK T, LO G, YL G, YV S, 2012, Cancer incidence and nuclear facilities in Ukraine: a community-based study. Exp Oncol 34(2):116–120 Yao X, Qian C-N, Zhang Z-F, TanM-H, Kort RJ, Yang XJ, Jmes H, 2007, Resau, and bin Tean Teh. Clin Cancer Res 13(1):161–169 Imao T, Egawa M, Takashima H, Kohida K, NamikiM(2004, Int J Urol 11:948–953 Rious-Leclercq N, Epstein JI, Bansard J-Y, Turlin B, Patard J-J, Manunta AA, Chan T, Ramee M-P, Lobel B, Moulinoux J-P, 2001, Clinical significance of cell proliferation, microvessel density, and CD44 adhesion molecule expression in Rena, cell carcinoma. Hum Pathol 32:1209–1215 Sabo E, Boltenko A, Sova Y, Stein A, Kleinhaus S, Murray B, 2001, Resnick Clinical Cancer Research 7:533–537 Schrami P, Struckmann K, Hatz F, Sonnet S, Kully C, Gasser T, Sauter G, Mihatsch MJ, Moch H, 2002, VHL mutations and their correlation with tumour cell proliferation, microvessel density, and patient prognosis in clear cell renal cell carcinoma. J Pathol 196: 186–193 Iakovlev VV, Gabril M, Dubinski W, Scorilas A, Youssef YM, Faragalla H, Kovacs K, Rotondo F, Metias S, Arsanious A, Plotkin A, Girgis AHF, Streutker CJ, Yousef. GM, 2012, Microvascular density as an independent predictor of clinical outcome in renal cell carcinoma: an automated image analysis study. Lab Investig 92:46–56 Nativ O, Sabo E, Reiss A,Wald M, Madjar S, Moskovitz B, 1998, Clinical significance of tumor angiogenesis in patients with localized renal cell carcinoma. Urology 51:693–696 Joo HJ, OH DK, Kim YS, Lee KB, Kim SJ, 2004 Feb, Increased expression of caveolin-1 and microvessel density correlates with metastasis and poor prognosis in clear cell renal cell carcinoma. BJU Int 93(3):291–296 Yoshino S, Kato M, Okada K, 1995, Prognostic significance of microvessel count in low stage renal cell carcinoma. Int J Urol 2: 156–160 Romanenko A, Kakehashi A, Morimura K, Wanibuchi H, Wei M, Vozianov A, Fukushima S, 2009, Urinary bladder carcinogenesis induced by chronic exposure to persistent low-dose ionizing radiation after Chernobyl accident. Carcinogenesis 30(11):1821–1831 Morimura K, Romanenko A, Min W, Salim EI, Kinoshita A, Wanibuchi H, Vozianov A, Fukushima S, 2004, Possible distinct molecular carcinogenic pathways for bladder cancer in Ukraine, before and after the Chernobyl disaster. Oncol Rep 11:881–886 Zablotska LB, Bazyka D, Lubin JH, Gudzenko N, Little MP, Hatch M, Finch S, Dyagil I, Reiss RF, Chumak VV, Bouville A, Drozdovitch V, Kryuchkov VP, Golovanov I, Bakhanova E, Babkina N, Lubarets T, Bebeshko V, Romanenko A, Mabuchi K, 2012, Radiation and the risk of chronic lymphocytic and other Leukemias among Chernobyl cleanup workers. Environ Health Perspect 121(1):59–65 Cote ML, Colt JS, Schwartz KL, Wacholder S, Ruterbusch JJ, Davis F, Purdue M, Graubard BI, Chow W, 2012, Cigarette smoking and renal cell carcinoma risk among black and white Americans: effect modification by hypertension and obesity. Cancer Epidemiol Biomark Prev 21(5):770–779 Llombart-Bosh A, 2002, New trends in cancer for the 21ST century. Adv Exp Med Biol 532:69–85 Folkman J, 2002, Role of angiogenesis in tumor growth and metastasis. Semin Oncol 29(6):15–18 Minardi D, Lucarini G, Filosa A, Milanese G, Zizzi A, Di Primio R, Montironi R, Muzzonigro G, 2008, Prognostic role of tumor necrosis, microvessel density, vascular endothelial growth factor and hypoxia inducible factor-1alpha in patients with clear cell renal carcinoma after radical nephrectomy in a long term follow-up. Int J Immunopathol Pharmacol 21(2):447–455 Tomisawa M, Tokunaga T, Oshika Y, Tsuchida T, Fukushima Y, Sato H, Kijima H, Yamazaki H, Ueyama Y, Tamaoki N, Nakamura M, 1999, Expression pattern of vascular endothelial growth factor isoform is closely correlated with tumour stage and vascularisation in renal cell carcinoma. Eur J Cancer 35(1):133–137 Mizukami Y, 2007, Hypoxia inducible factor-1 independent pathways in tumor angiogenesis. Clin Cancer Res 13(19):5670–5674 Boticario C, Cascales M, 2010, Hipoxia y cancer. Anales Real Academia Nacional Farmacologia 76(3):379–408 http//www.nap.edu/catalog.php?record_id=11340. The National Academies Press. Health Risks from Exposure to Low Levels of Ionizing Radiation: BEIR VII Phase 2, 2006). Vickers MM, Heng DY, 2010, Prognostic and predictive biomarkers in renal cell carcinoma. Target Oncol 5(2):85–94 Lidgren A, Hedberg Y, Grankvist K, Rasmuson T, Vasko J, Ljungberg B, 2005, The expression of hypoxia-inducible factor 1alpha is a favorable independent prognostic factor in renal cell carcinoma. Clin Cancer Res 11:1129–1135 Henze AT, Acker T, 2010, Feedback regulators of hypoxiainducible factors and their role in cancer biology. Cell Cycle 9: 2749–2763 Markovic-Lipkovski J, Brasanac D. Muller GA, Muller CA. Cadherins and integrins in renal cell carcinoma: an immunohistochemical study. Tumori 2001; 87(3):173–178. Meyer M, ClausM, Lepple-Wienhues A,Waltenberger J, Augustin HG, Ziche M, Lanz C, Buttner M, RzihaHJ, Dhio C, 1999, Anovel vascular endothelial growth factor encoded by Orf virus, VEGF-E, mediates angiogenesis via signalling through VEGFR-2, KDR, but not VEGFR-1, Flt-1, receptor tyrosine kinases. EMBO J 18(2): 363–374 Dordevic G, Matusan-LLijas K, Babarovic E, Hadzisejdic I, Grahova M, Grahov B, Jonjic N, 2009, Hypoxia inducible factor- 1alpha correlates with vascular endothelial growth factor a and C indicating worse prognosis in clear cell renal cell carcinoma. J Exp Clin Cancer Res 28(1):40–47 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 807 EP 815 DI 10.1007/s12253-016-0072-7 PG 9 ER PT J AU Mirecka, A Morawiec, Z Wozniak, K AF Mirecka, Alicja Morawiec, Zbigniew Wozniak, Katarzyna TI Genetic Polymorphism of SUMO-Specific Cysteine Proteases − SENP1 and SENP2 in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; SENP1 gene; SENP2 gene; Gene polymorphism; SUMO modification ID Breast cancer; SENP1 gene; SENP2 gene; Gene polymorphism; SUMO modification AB SENP proteases take part in post-translational modification of proteins known as sumoylation. They catalyze three distinct processes during sumoylation: processing of SUMO protein, deconjugation of SUMO from the target protein, and chain editing which mentions to the dismantling of SUMO chain. Many proteins that are involved in the basic processes of cells, such as regulation of transcription, DNA repair or cell cycle control, are sumoylated. The aim of these studies was to investigate an association between polymorphic variants (SNPs) of the SENP1 gene (c.1691 + 36C > T, rs12297820) and SENP2 gene (c.902C > A, p.Thr301Lys, rs6762208) and a risk of breast cancer occurrence. We performed a case-control study in 324 breast cancer cases and 335 controls using PCR-RLFP. In the case of the SENP1 gene polymorphism we did not find any association between this polymorphism and breast cancer risk. In the case of SENP2 gene polymorphism we observed higher risk of breast cancer for carriers of the A allele (OR =1.33; 95 % CI 1.04–1.69). Our analysis also showed the genotype C/C (OR =0.67, 95 % CI 0.48–0.93) and the allele C (OR =0.75, 95%CI 0.59–0.69) of this polymorphism decrease a risk of breast cancer.We also checked the distribution of genotypes and frequency of alleles of the SENP1 and SENP2 genes polymorphisms in groups of patients with different hormone receptor status, patients with positive and negative lymph node status and patients with different tumor grade. Odds ratio analysis showed a higher risk of metastases in women with the genotype C/C (OR =2.07, 95 % CI 1.06–4.05) and allele C (OR =2.10 95 % CI 1.10–4.01) of the c.1691 + 36C > T SENP1 gene polymorphism. Moreover, we observed reduced risk in women with the allele T (OR =0.48, 95 % CI 0.25–0.91) in this polymorphic site. In the case of SENP2 gene polymorphism we observed that the A/A genotype correlated with the lack of estrogen receptor (OR =1.94, 95 % CI 1.04–3.62). Our results suggest that the variability of the SENP1 and SENP2 genes may play a role in breast cancer occurrence. Further studies are needed to clarify their biological functions in breast cancer. C1 [Mirecka, Alicja] University of Lodz, Department of Molecular GeneticsLodz, Poland. [Morawiec, Zbigniew] N. Copernicus Hospital, Department of Surgical OncologyLodz, Poland. [Wozniak, Katarzyna] University of Lodz, Department of Molecular GeneticsLodz, Poland. RP Wozniak, K (reprint author), University of Lodz, Department of Molecular Genetics, Lodz, Poland. EM wozniak@biol.uni.lodz.pl CR Owerbach D, Mckay EM,Yeh ET, GabbayKH, BohrenKM(2005, A proline-90 residue unique to SUMO-4 prevents maturation and sumoylation. Biochem Biophys Res Commun 337:517–520 Bawa-Khalfe T, Yeh ETH, 2010, SUMO losing balance: SUMO proteases disrupt SUMO homeostasis to facilitate cancer development and progression. Genes Cancer 1:748–752 Kim JH, Baek SH, 2009, Emerging roles of desumoylating enzymes. Biochim Biophys Acta 1792:155–162 Itahana Y, Yeh ETH, Zhang Y, 2006, Nucleocytoplasmic shuttling modulates activity and ubiquitination-dependent turnover of SUMO-specific protease 2. Mol Cell Biol 26:4675–4689 Watts FZ, 2013, Starting and stopping SUMOylation. What regulates the regulator? Chromosoma 122:451–463 Ihara M, Koyama H, Uchimura Y, Saitoh H, Kikuchi A, 2007, Noncovalent binding of small ubiquitin-related modifier, SUMO, protease to SUMO is necessary for enzymatic activities and cell growth. J Biol Chem 282:16465–16475 Bettermann K, Benesch M, Weis S, Haybaeck J, 2012, SUMOylation in carcinogenesis. Cancer Lett 316:113–125 Jacques C, Baris O, Prunier-Mirebeau D, Savagner F, Rodien P, Rohmer V, Franc B, Guyetant S, Malthiery Y, Reynier P, 2005, Two-step differential expression analysis reveals a new set of genes involved in thyroid oncocytic tumors. J Clin Endocrinol Metab 90: 2314–2320 Cheng J, Bawa T, Lee P, Gong L, Yeh ET, 2006, Role of desumoylation in the development of prostate cancer. Neoplasia 8:667–676 Bawa-Khalfe T, Cheng J, Lin SH, Ittmann MM, Yeh ETH, 2010, SENP1 induces prostatic intraepithelial neoplasia through multiple mechanisms. J Biol Chem 285:25859–25866 Wang C, Tao W, Ni S, Chen Q, Zhao Z, Ma L, Fu Y, Jiao Z, 2015, Tumor-suppressive microRNA-145 induces growth arrest by targeting SENP1 in human prostate cancer cells. Cancer Sci 106: 375–382 Ma C, Wu B, Huang X, Yuan Z, Nong K, Dong B, Bai Y, Zhu H, Wang W, Ai K, 2014, SUMO-specific protease 1 regulates pancreatic cancer cell proliferation and invasion by targetingMMP- 9. Tumor Biol 35:12729–12735 Wang Q, Xia N, Li T, Xu Y, Zou Y, Zou Y, Fan Q, Bawa-Khalfe T, Yeh ET, Cheng J, 2013, SUMO-specific protease 1 promotes prostate cancer progression and metastasis. Oncogene 32:2493–2498 Xu Y, Li J, Zuo Y, Deng J, Wang LS, Chen GQ, 2011, SUMOspecific protease 1 regulates the in vitro and in vivo growth of colon cancer cells with the upregulated expression of CDK inhibitors. Cancer Lett 309:78–84 Jiang Q-F, Tian Y-W, Shen Q, Xue H-Z, Li K, 2014, SENP2 regulated the stability of β-catenin through WWOX in hepatocellular carcinoma cell. Tumor Biol 35:9677–9682 Tan MY, Mu XY, Liu B, Wang Y, Bao ED, Qiu JX, Fan Y, 2013, SUMO-specific protease 2 suppresses cell migration and invasion through inhibiting the expression of MMP13 in bladder cancer cells. Cell Physiol Biochem 32:542–548 Nait Achour T, Sentis S, Teyssier C, Philippat A, Lucas A, Corbo L, Cavailles V, Jalaguier S, 2014, Transcriptional repression of estrogen receptor α signaling by SENP2 in breast cancer cells. Mol Endocrinol 28:183–196 Goeres J, Chan P-K, Mukhopadhyay D, Zhang H, Raught B, Matunis MJ, 2011, The SUMO-specific isopeptidase SENP2 associates dynamically with nuclear pore complexes through interactions with karyopherins and the Nup107-160 nucleoporin subcomplex. Mol Biol Cell 22:4868–4882 Hang J, Dasso M, 2002, Association of the human SUMO-1 protease SENP2 with the nuclear pore. J Biol Chem 277:19961–19966 Chow KH, Elgort S, DassoM, PowersMA, Ullman KS, 2014, The SUMO proteases SENP1 and SENP2 play a critical role in nucleoporin homeostasis and nuclear pore complex function. Mol Biol Cell 25:160–168 Kumar A, Zhang KYJ, 2015, Advances in the development of SUMO specific protease, SENP, inhibitors. Comput Struct Biotechnol J 13:204–211 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 817 EP 823 DI 10.1007/s12253-016-0064-7 PG 7 ER PT J AU Wang, Hy Zhang, Jj Zheng, Xy Liu, Jh Li, Yw AF Wang, Hai-ying Zhang, Jin-jun Zheng, Xiang-yu Liu, Jian-hua Li, Yong-wei TI Association between IL-6 Gene (−174 & -572 G/C) Polymorphisms and Endometrial Adenocarcinoma Risk SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Interleukin-6; Endometrial adenocarcinoma; Gene polymorphism ID Interleukin-6; Endometrial adenocarcinoma; Gene polymorphism AB We aimed to evaluate the association of IL-6 gene polymorphisms at positions of −174 and −572 and predisposition of endometrial adenocarcinoma (EAC) in a Chinese population. EAC patients have remarkably higher frequency of IL-6 -174 CC genotype [odds ratio (OR) =1.56, 95 % confidence interval (CI) =1.07–2.23; P = 0.03], IL-6 -572 CC genotype (OR =1.93, 95%CI =1.17–3.15; P = 0.01) and IL-6 -174 C allele (OR =1.22, 95 % CI =1.03–1.46; P = 0.04) compared with healthy controls. When stratified with FIGO stage, patients with III-IV EAC have a significantly higher frequency of IL-6 -174 CC genotype (OR =1.66, 95%CI =1.06–2.58; P = 0.02) than healthy controls. The CC genotype of IL-6 gene polymorphisms at positions of −174 and −572 may denote potential high risk of EAC. C1 [Wang, Hai-ying] Henan Province Chinese Medicine Hospital, Department of laboratory medicine, 450002 Zhenzhou, China. [Zhang, Jin-jun] Henan Province Chinese Medicine Hospital, Department of laboratory medicine, 450002 Zhenzhou, China. [Zheng, Xiang-yu] Henan Province Chinese Medicine Hospital, Department of laboratory medicine, 450002 Zhenzhou, China. [Liu, Jian-hua] Henan Province Chinese Medicine Hospital, Department of laboratory medicine, 450002 Zhenzhou, China. [Li, Yong-wei] Henan Province Chinese Medicine Hospital, Department of laboratory medicine, 450002 Zhenzhou, China. RP Liu, Jh (reprint author), Henan Province Chinese Medicine Hospital, Department of laboratory medicine, 450002 Zhenzhou, China. EM LIUJianh68@126.com;LIUJianhua593@gmail.com CR Fowler W, Mutch D, 2008, Management of endometrial cancer. Women's Health, Lond Engl, 4:479–489 Hosoi A, Ueda Y, Shindo M, et al., 2013, Endometrial thickness measured by ultrasonography in postmenopausal patients with endometrial carcinoma has significance, irrespective of histological subtype. Int J Gynecol Cancer 23:1266–1269 Tao QS, Huang HL, Chai Y, et al., 2012, Interleukin-6 up-regulates the expression of interleukin-15 is associated withMAPKs and PI3- K signaling pathways in the human keratinocyte cell line, HaCaT. Mol Biol Rep 39:4201–4205 Bowcock AM, Kidd JR, Lathrop GM, et al., 1988, The human "interferon-beta 2/hepatocyte stimulating factor/interleukin-6" gene: DNA polymorphism studies and localization to chromosome 7p21. Genomics 3:8–16 Huang D, Zheng C, Giscombe R, Matell G, Pirskanen R, Lefvert AK, 1999, Polymorphisms at −174 and in the 3′ flanking region of interleukin-6, IL-6, gene in patients with myasthenia gravis. J Neuroimmunol 101:197–200 Bennermo M, Held C, Green F, et al., 2004, Prognostic value of plasma interleukin-6 concentrations and the −174 G > C and −572 G > C promoter polymorphisms of the interleukin-6 gene in patients with acute myocardial infarction treated with thrombolysis. Atherosclerosis 174:157–163 Magalhaes JF, Cortinhas AJ, Albuquerque CM, et al., 2013, Interleukin-6 gene -174G > C and -636G > C promoter polymorphisms and prostate cancer risk. Mol Biol Rep 40:449–455 Xu B, Niu XB, Wang ZD, et al., 2011, IL-6-174G > C polymorphism and cancer risk: a meta-analysis involving 29, 377 cases and 37, 739 controls. Mol Biol Rep 38:2589–2596 Garg R, Wollan M, Galic V, et al., 2006, Common polymorphism in interleukin 6 influences survival of women with ovarian and peritoneal carcinoma. Gynecol Oncol 103:793–796 Nogueira de Souza NC, Brenna SM, Campos F, Syrjanen KJ, Baracat EC, Silva ID, 2006, Interleukin-6 polymorphisms and the risk of cervical cancer. Int J Gynecol Cancer 16:1278–1282 Godarzi EM, Sarvestani EK, Aflaki E, Amirghofran Z, 2011, Interleukin-6 gene polymorphism in Iranian patients with systemic lupus erythematosus[J]. Clin Rheumatol 30(2):179–184 Prayong P, Mairiang E, Pairojkul C, et al., 2014, An interleukin-6 receptor polymorphism is associated with opisthorchiasis-linked cholangiocarcinoma risk in Thailand. Asian Pac J Cancer Prev 15: 5443–5447 Liu Z,Wang Z, Xiao Y, Lu Y, Lu Y, 2015, Association between the interleukin-6 gene polymorphisms and renal cancer risk. Immunol Lett 164:125–128 Ebadi N, Jahed M, Mivehchi M, Majidizadeh T, Asgary M, Hosseini SA, 2014, Interleukin-12 and interleukin-6 gene polymorphisms and risk of bladder cancer in the Iranian population. Asian Pac J Cancer Prev 15:7869–7873 Totaro F, Cimmino F, Pignataro P, et al., 2013, Impact of interleukin- 6-174 G > C gene promoter polymorphism on neuroblastoma. PLoS One 8:e76810 Pohjanen VM, Koivurova OP,Makinen JM, et al., 2013, Interleukin 6 gene polymorphism −174 is associated with the diffuse type gastric carcinoma. Genes Chromosom Cancer 52:976–982 Tang S, Yuan Y, He Y, et al., 2014, Genetic polymorphism of interleukin-6 influences susceptibility to HBV-related hepatocellular carcinoma in amale Chinese Han population. HumImmunol 75: 297–301 Delahanty RJ, Xiang YB, Spurdle A, et al., 2013, Polymorphisms in inflammation pathway genes and endometrial cancer risk. Cancer Epidemiol Biomark Prev 22:216–223 Srivani R, Nagarajan B, 2003, A prognostic insight on in vivo expression of interleukin-6 in uterine cervical cancer. Int J Gynecol Cancer 13:331–339 Bellone S,Watts K, Cane’ S et al. High serum levels of interleukin- 6 in endometrial carcinoma are associated with uterine serous papillary histology, a highly aggressive and chemotherapyresistant variant of endometrial cancer. Gynecol Oncol 2005; 98: 92–98. Wei LH, KuoML, Chen CA, et al., 2001, The anti-apoptotic role of interleukin-6 in human cervical cancer ismediated by up-regulation of mcl-1 through a PI 3-K/Akt pathway. Oncogene 20:5799–5809 Grimm C, Watrowski R, Baumuhlner K, et al., 2011, Genetic variations of interleukin-1 and −6 genes and risk of cervical intraepithelial neoplasia. Gynecol Oncol 121:537–541 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 825 EP 829 DI 10.1007/s12253-016-0073-6 PG 5 ER PT J AU Pyo, JS Sohn, HJ Kang, G AF Pyo, Jung-Soo Sohn, Hee Jin Kang, Guhyun TI Diagnostic Accuracy of BRAF Immunohistochemistry in Colorectal Cancer: a Meta-Analysis and Diagnostic Test Accuracy Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BRAFV600E immunohistochemistry; Colorectal cancer; Concordance; Diagnostic accuracy; Meta-analysis ID BRAFV600E immunohistochemistry; Colorectal cancer; Concordance; Diagnostic accuracy; Meta-analysis AB The aim of this study was to evaluate the concordance between the BRAFV600E mutation test and immunohistochemistry (IHC) and to evaluate the diagnostic accuracy of BRAF IHC for colorectal cancer (CRC) through a systematic review, meta-analysis, and diagnostic test accuracy review. The current study included 1021 CRCs from eight eligible studies. The concordance rates were investigated between BRAF IHC and the mutation test. In addition, diagnostic test accuracy review was conducted and calculated using the value of area under curve (AUC) on the summary receiver operating characteristic (SROC) curve. The positive rate of BRAF IHC was 30.5 % (range; 13.2–66.2 %), and the BRAF mutation was found in 30.2 % (range; 11.7–66.2 %). The overall concordance rate between BRAF IHC and the mutation test was 0.944 (95 % confidence interval (CI) 0.873–0.977). In the BRAF IHC-positive and -negative groups, the concordance rates between BRAF IHC and the mutation test were 0.895 (95 % CI 0.800–0.945) and 0.956 (95 % CI 0.878–0.985), respectively. The pooled sensitivity and specificity were 0.94 (95 % CI 0.91–0.96) and 0.96 (95 % CI 0.95–0.98), respectively. The diagnostic odds ratio was 272.86 (95 % CI 46.11– 1614.88), and the value of AUC on SROC curve was 0.9846. Taken together, our results suggest that BRAF IHC is strongly concordant with the BRAF mutation test and has high diagnostic accuracy in BRAF mutation analysis of CRCs. Further cumulative studies on detailed evaluation criteria are needed before application in daily practice. C1 [Pyo, Jung-Soo] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 29 Saemunanro, Jongno-gu, 03181 Seoul, South Korea. [Sohn, Hee Jin] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 29 Saemunanro, Jongno-gu, 03181 Seoul, South Korea. [Kang, Guhyun] Inje University Sanggye Paik Hospital, Department of PathologySeoul, South Korea. RP Sohn, HJ (reprint author), Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Pathology, 03181 Seoul, South Korea. EM jhpath.sohn@samsung.com CR Pyo JS, Kang G, Kim DH, Chae SW, Park C, Kim K, et al., 2013, Activation of nuclear factor-κB contributes to growth and aggressiveness of papillary thyroid carcinoma. Pathol Res Pract 209:228– 232 Cantwell-Dorris ER, O’Leary JJ, Sheils OM, 2011, BRAFV600E: implications for carcinogenesis and molecular therapy. Mol Cancer Ther 10:385–394 Rubinstein JC, Sznol M, Pavlick AC, Ariyan S, Cheng E, Bacchiocchi A, et al., 2010, Incidence of the V600 K mutation among melanoma patients with BRAF mutations, and potential therapeutic response to the specific BRAF inhibitor PLX4032. J Transl Med 8:67 Adackapara CA, Sholl LM, Barletta JA, Hornick JL, 2013, Immunohistochemistry using the BRAF V600E mutation-specific monoclonal antibody VE1 is not a useful surrogate for genotyping in colorectal adenocarcinoma. Histopathology 63:187–193 Affolter K, Samowitz W, Tripp S, Bronner MP, 2013, BRAF V600E mutation detection by immunohistochemistry in colorectal carcinoma. Genes Chromosom Cancer 52:748–752 Day F, Muranyi A, Singh S, Shanmugam K,Williams D, Byrne D, et al., 2015, A mutant BRAF V600E-specific immunohistochemical assay: correlation with molecular mutation status and clinical outcome in colorectal cancer. Target Oncol 10:99–109 Dvorak K, Aggeler B, Palting J, McKelvie P, Ruszkiewicz A, Waring P, 2014, Immunohistochemistry with the anti-BRAF V600E, VE1, antibody: impact of pre-analytical conditions and concordance with DNA sequencing in colorectal and papillary thyroid carcinoma. Pathology 46:509–517 Lasota J, Kowalik A, Wasag B, Wang ZF, Felisiak-Golabek A, Coates T, et al., 2014, Detection of the BRAF V600E mutation in colon carcinoma: critical evaluation of the imunohistochemical approach. Am J Surg Pathol 38:1235–1241 Nolan S, Arnason T, Drucker A, Huang WY, 2014, The utility of BRAFV600E mutation-specific antibody for colon cancers with microsatellite instability. Appl Immunohistochem Mol Morphol 22:e8–e13 Roth RM, Hampel H, Arnold CA, Yearsley MM, Marsh WL, Frankel WL, 2015, A modified Lynch syndrome screening algorithm in colon cancer: BRAF immunohistochemistry is efficacious and cost beneficial. Am J Clin Pathol 143:336–343 Sinicrope FA, Smyrk TC, Tougeron D, Thibodeau SN, Singh S, Muranyi A, et al., 2013, Mutation-specific antibody detects mutant BRAFV600E protein expression in human colon carcinomas. Cancer 119:2765–2770 Long GV,Wilmott JS, Capper D, PreusserM, Zhang YE, Thompson JF, et al., 2013, Immunohistochemistry is highly sensitive and specific for the detection of V600E BRAF mutation in melanoma. Am J Surg Pathol 37:61–65 Tiacci E, Trifonov V, Schiavoni G, Holmes A, Kern W, Martelli MP, et al., 2011, BRAF mutations in hairy-cell leukemia. N Engl J Med 364:2305–2315 Pakneshan S, Salajegheh A, Smith RA, Lam AK, 2013, Clinicopathological relevance of BRAF mutations in human cancer. Pathology 45:346–356 Grisham RN, Iyer G, Garg K, DeLair D, Hyman DM, ZhouQ, et al., 2013, BRAF mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer. Cancer 119:548–554 Vaughn CP, Zobell SD, Furtado LV, Baker CL, Samowitz WS, 2011, Frequency of KRAS, BRAF, and NRAS mutations in colorectal cancer. Genes Chromosom Cancer 50:307–312 Toon CW, Walsh MD, Chou A, Capper D, Clarkson A, Sioson L, et al., 2013, BRAFV600E immunohistochemistry facilitates universal screening of colorectal cancers for lynch syndrome. Am J Surg Pathol 37:1592–1602 Barras D, 2015, BRAF mutation in colorectal cancer: an update. Biomark Cancer 7(Suppl 1):9–12 Parsons MT, Buchanan DD, Thompson B, Young JP, Spurdle AB, 2012, Correlation of tumour BRAF mutations and MLH1 methylation with germline mismatch repair, MMR, gene mutation status: a literature review assessing utility of tumour features for MMR variant classification. J Med Genet 49:151–157 Pyo JS, Sohn JH, Kang G, 2015, BRAF immunohistochemistry using clone VE1 is strongly concordant with BRAF, V600E, mutation test in papillary thyroid carcinoma. Endocr Pathol 26:211– 217 Zamora J, Abraira V, Muriel A, Khan K, Coomarasamy A, 2006, Meta-DiSc: a software for meta-analysis of test accuracy data. BMC Med Res Methodol 6:31 Moses LE, Shapiro D, Littenberg B, 1993, Combining independent studies of a diagnostic test into a summary ROC curve: dataanalytic approaches and some additional considerations. Stat Med 12:1293–1316 Bahreini F, Soltanian AR, Mehdipour P, 2015, A meta-analysis on concordance between immunohistochemistry, IHC, and fluorescence in situ hybridization, FISH, to detect HER2 gene overexpression in breast cancer. Breast Cancer 22:615–625 Pyo JS, Sohn JH, Kim WH, 2016, Concordance rate between HER2 immunohistochemistry and in situ hybridization in gastric carcinoma: systematic review and meta-analysis. Int J BiolMarkers 31:e1–10 Vakiani E, Yaeger R, Brooke S, Zhou Y, Klimstra DS, Shia J, 2015, Immunohistochemical detection of the BRAF V600E mutant protein in colorectal neoplasms. Appl ImmunohistochemMolMorphol 23:438–443 Capper D, Voigt A, Bozukova G, Ahadova A, Kickingereder P, von Deimling A, et al., 2013, BRAF V600E-specific immunohistochemistry for the exclusion of lynch syndrome in MSI-H colorectal cancer. Int J Cancer 133:1624–1630 Kim YH, Choi SE, Yoon SO, Hong SW(2014, A testing algorithm for detection of the B-type Raf kinase V600E mutation in papillary thyroid carcinoma. Hum Pathol 45:1483–1488 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 831 EP 837 DI 10.1007/s12253-016-0077-2 PG 7 ER PT J AU Sarsik, B Doganavsargil, B Simsir, A Yazici, A Pehlivanoglu, B Cal, C Sen, S AF Sarsik, Banu Doganavsargil, Basak Simsir, Adnan Yazici, Ayse Pehlivanoglu, Burcin Cal, Cag Sen, Sait TI P21 and p27 Immunoexpression in Upper Urinary Tract Urothelial Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cylin-dependent kinase inhibitors; p21; p27; Prognosis; Upper tract urothelial carcinoma; Urinary tract ID Cylin-dependent kinase inhibitors; p21; p27; Prognosis; Upper tract urothelial carcinoma; Urinary tract AB p21 and p27 are members of cyclin-dependent kinase family, which function as tumor suppressors and they are involved in development and progression of several malignancies. We investigated their expression in upper urinary tract urothelial carcinoma (UUTUC). Radical nephroureterectomy materials of 34 patients were assessed by immunohistochemistry to evaluate expression of p21 and p27 in UUTUC. Results were correlated with various clinicopathological variables as age, gender, tumor grade and stage, tumor architecture, multifocality, subsequent bladder carcinoma development and clinical outcome. p21 and p27 expression was observed in 52.9 % (n = 18) and 88.2 % (n = 30), respectively. A total of 21 tumors (61.7 %) showed either total loss of p21 expression (n = 16, 47 %) or lower expression (n = 5, 14.7 %). No correlation was found between p21 expression and clinicopathologic variables. Cases showing total loss or lower p27 expression (11.7 % and <25.6 %, respectively) (n = 19, 55.8 %) constituted 67.6 % (n = 23) of the cases totally. This loss or lower p27 expression correlated with a shorter overall survival in both univariate and multivariate analysis (p = 0.039 and p = 0.037, respectively). None of the noninvasive tumors (papillary and nodular tumors) showed loss of p27 (p = 0.016) while 33.3 % of invasive ones showed p27 loss. Noninvasive tumor architecture also correlated with subsequent bladder carcinoma development (p = 0.032) while invasive tumor architecture correlated with advanced stage (T3 and T4) (p = 0.003). p27 is widely expressed in UUTUC, while p21 expression is observed in half of the cases. Loss of p27 expression correlated with tumor architecture and overall survival in UUTUC. However, further research is needed to assess their role in UUTUC. C1 [Sarsik, Banu] Ege University, Faculty of Medicine, Department of Pathology, Bornova, 35100 Izmir, Turkey. [Doganavsargil, Basak] Ege University, Faculty of Medicine, Department of Pathology, Bornova, 35100 Izmir, Turkey. [Simsir, Adnan] Ege University, Faculty of Medicine, Department of UrologyIzmir, Turkey. [Yazici, Ayse] Mugla University, Education and Research Hospital, Department of PathologyMugla, Turkey. [Pehlivanoglu, Burcin] Adiyaman University, Training and Research Hospital, Department of PathologyAdiyaman, Turkey. [Cal, Cag] Ege University, Faculty of Medicine, Department of UrologyIzmir, Turkey. [Sen, Sait] Ege University, Faculty of Medicine, Department of Pathology, Bornova, 35100 Izmir, Turkey. RP Sarsik, B (reprint author), Ege University, Faculty of Medicine, Department of Pathology, 35100 Izmir, Turkey. EM bsarsik@yahoo.com CR Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics, 2009. CA Cancer J Clin 59(4):225–249 Novara G, De Marco V, Gottardo F, Dalpiaz O, Bouygues V, Galfano A, et al., 2007, Independent predictors of cancer-specific survival in transitional cell carcinoma of the upper urinary tract: multiinstitutional dataset from 3 European centers. Cancer 110(8): 1715–1722 Chromecki TF, Cha EK, Fajkovic H, Margulis V, Novara G, Scherr DS, et al., 2012, The impact of tumor multifocality on outcomes in patients treated with radical nephroureterectomy. Eur Urol 61(2): 245–253 Verhoest G, Shariat SF, Chromecki TM, Raman JD, Margulis V, Novara G, et al., 2011, Predictive factors of recurrence and survival of upper tract urothelial carcinomas. World J Urol 29(4):495–501 Catzavelos C, Bhattacharya N, Ung YC, Wilson JA, Roncari L, Sandhu C, et al., 1997, Decreased levels of the cell-cycle inhibitor p27Kip1 protein; prognostic implications in primary breast cancer. Nat Med 3(2):227–230 Cote RJ, Susan Y, Groshen S, Alexander GM, Gatti LA, Firpo EJ, et al., 1998, Association of p27Kip1 levels with recurrence and survival in patients with stage C prostate carcinoma. J Natl Cancer Inst 90(12):916–920 Loda M, Cukor B, Tam SW, Lavin P, Fiorentino M, Draetta GF, et al., 1997, Increased proteasomedependent degradation of the cyclin-dependent kinase inhibitor p27 in aggressive colorectal carcinomas. Nat Med 3(2):231–234 Shariat SF, Zlotta AR, Ashfaq R, Sagalowsky AI, Lotan Y, 2007, Cooperative effect of cell-cycle regulators expression on bladder cancer development and biologic aggressiveness. Mod Pathol 20(4):445–459 Lloyd RV, Erickson LA, Jin L, Kulig E, Qian X, Cheville JC, et al., 1999, p27kip1: a multifunctional cyclin-dependent kinase inhibitor with prognostic significance in human cancers. Am J Pathol 154(2):313–323 Korkolopoulou P, Konstantinidou AE, Thomas-Tsagli E, Christodoulou P, Kapralos P, Davaris P, 2000, WAF1/p21 protein expression is an independent prognostic indicator in superficial and invasive bladder cancer. Appl Immunohistochem Mol Morphol 8(4):285–292 Shariat SF, Karakiewicz PI, Ashfaq R, Lerner SP, Palapattu GS, Cote RJ, et al., 2008, Multiple biomarkers improve prediction of bladder cancer recurrence and mortality in patients undergoing cystectomy. Cancer 112(2):315–325 Matsushima H, Sasaki T, Goto T, HosakaY, Homma Y, Kitamura T, et al., 1998, Immunohistochemical study of p21WAF1 and p53 proteins in prostatic cancer and their prognostic significance. Hum Pathol 29(8):778–783 Masuda M, Takano Y, Iki M, Makiyama K, Ikeda I, Noguchi S, et al., 2000, Cyclin-dependent kinase inhibitor p27kip1 expression in transitional cell carcinoma of renal pelvis and ureter. Cancer Lett 150(2):183–189 Kamai T, Takagi K, Asami H, Ito Y, Arai K, Yoshida KI, 2000, Prognostic significance of p27kip1 and Ki-67 expression in carcinoma of the renal pelvis and ureter. BJU Int 86(1):14–19 Fromont G, Roupret M, Amira N, Sibony M, Vallancien G, Validire P, et al., 2005, Tissuemicroarray analysis of the prognostic value of E-cadherin, Ki67, p53, p27, survivin and MSH2 expression in upper urinary tract transitional cell carcinoma. Eur Urol 48(5):764– 770 Gakis G, Schwentner C, Todenhofer T, Stenzl A, 2012, Current status of molecular markers for prognostication and outcome in invasive bladder cancer. BJU Int 110(2):233–237 Simsir A, Sarsik B, Cureklibatir I, Sen S, Gunaydin G, Cal C, 2011, Prognostic factors for upper urinary tract urothelial carcinomas: stage, grade, and smoking status. Int Urol Nephrol 43(4):1039– 1045 Sobin LH, GospodarowiczMK,Wittekind C, eds. TNM classification of malignant tumors, ed. 7. UICC International Union Against Cancer. Hoboken, NJ: Wiley-Blackwell; 2010. p. 258–265. Eble JN, Sauter G, Epstein JI, et al. World Health Organization classification of tumours. Pathology and genetics of tumours of the urinary system and male genital organs. Lyon, France: IARC Press; 2004, p.90. Remzi M, Haitel A, Margulis V, Karakiewicz P, Montorsi F, Kikuchi E, et al., 2009, Tumour architecture is an independent predictor of outcomes after nephroureterectomy: a multiinstitutional analysis of 1363 patients. BJU Int 103(3):307–311 Doganavsargil B, Simsir A, Boyacioglu H, Cal C, Hekimgil M, 2006, A comparison of p21 and p27 immunoexpression in benign glands, prostatic intraepithelial neoplasia and prostate adenocarcinoma. BJU Int 97:644–648 Romics I, Banfi G, Szekely E, Krenacs T, Szende B, 2008, Expression of p21(waf1/cip1), p27, kip1), p63 and androgen receptor in low and high Gleason score prostate cancer. Pathol Oncol Res 14(3):307–311 Huang LW, Seow KM, Lee CC, Lin YH, Pan HS, Chen HJ, 2010, Decreased p21 expression in HPV-18 positive cervical carcinomas. Pathol Oncol Res 16(1):81–86 Fritsche HM, Novara G, Burger M, Gupta A, Matsumoto K, Kassouf W, et al., 2012, Macroscopic sessile tumor architecture is a pathologic feature of biologically aggressive upper tract urothelial carcinoma. Urol Oncol 30(5):666–672 Margulis V, Youssef RF, Karakiewicz PI, Lotan Y, Wood CG, Zigeuner R, et al., 2010, Preoperative multivariable prognostic model for prediction of nonorgan confined urothelial carcinoma of the upper urinary tract. J Urol 184(2):453–458 Franke KH, Miklosi M, Goebell P, Clasen S, Steinhoff C, Anastasiadis AG, et al., 2000, Cyclin-dependent kinase inhibitor P27, KIP1, is expressed preferentially in early stages of urothelial carcinoma. Urology 56(4):689–695 Hisataki T, Miyao N, Masumori N, Takahashi A, Sasai M, Yanase M, et al., 2000, Risk factors for the development of bladder cancer after upper tract urothelial cancer. Urology 55(5):663–667 Koga F, Nagamatsu H, Ishimaru H, Mizuo T, Yoshida K, 2001, Risk factors for the development of bladder transitional cell carcinoma following surgery for transitional cell carcinoma of the upper urinary tract. Urol Int 67(2):135–141 Raman JD, Ng CK, Scherr DS, Margulis V, Lotan Y, Bensalah K, et al., 2010, Impact of tumor location on prognosis for patients with upper tract urothelial carcinoma managed by radical nephroureterectomy. Eur Urol 57(6):1072–1079 Clasen S, Schulz WA, Gerharz CD, Grimm MO, Christoph F, Schmitz-Drager BJ, 1998, Frequent and heterogeneous expression of cyclin-dependent kinase inhibitorWAF1/p21 protein andmRNA in urothelial carcinoma. Br J Cancer 77(4):515–521 Zlotta AR, Noel JC, Fayt I, Drowart A, Van Vooren JP, Huygen K, et al., 1999, Correlation and prognostic significance of p53, p21 WAF1/CIP1 and Ki67 expression in patients with superficial bladder tumors treated with bacillus Calmette-Guerin intravesical therapy. J Urol 161(3):792–798 Stein JP, Ginsberg DA, Grossfeld GD, Chatterjee SJ, Esrig D, Dickinson MG, et al., 1998, Effect of p21 WAF1/CIP1 expression on tumor progression in bladder cancer. J Natl Cancer Inst 90(14): 1072–1079 Philipp-Staheli J, Kim KH, Liggitt D, Gurley KE, Longton G, Kemp CJ, 2004, Distinct roles for p53, 27Kip1, and p21Cip1 during tumor development. Oncogene 23(4):905–913 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 839 EP 845 DI 10.1007/s12253-016-0075-4 PG 7 ER PT J AU Benezech, S Chabaud, S Chambon, F Dijoud, F Chotel, F Marec-Berard, P AF Benezech, Sarah Chabaud, Sylvie Chambon, Fanny Dijoud, Frederique Chotel, Franck Marec-Berard, Perrine TI Prognostic Value of Vascular Invasion in Pediatric Osteosarcomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bone tumor; Vascular invasion; Osteosarcoma; Prognosis ID Bone tumor; Vascular invasion; Osteosarcoma; Prognosis AB Metastatic status, histologic response, and quality of surgical resection are prognostic factors for osteosarcomas. Pathology reports sometimes describe peritumoral vascular invasion on surgical specimens after neoadjuvant chemotherapy but their prognostic significance as an independent parameter has never been reported. The aim of this study was to evaluate how the presence of this peritumoral vascular invasion could influence survival. We retrospectively analyzed histology, demographics, and outcomes of pediatric patients treated for osteosarcoma in our institutions between January 2007 and December 2012. A single pathologist analyzed the resection specimens after neoadjuvant chemotherapy. Fiftyone osteosarcomas were diagnosed over a 6-year period; nine had metastatic disease at diagnosis. Surgery was performed after neoadjuvant chemotherapy in all cases. We identified peritumoral vascular invasion in the surgical specimens in 15 cases. Two-year event-free survival (EFS) was 78 % (CI95%[64;93]) for patients without vascular invasion versus 48 % (CI95% [21;75]) in patients with vascular invasion, and 2-year overall survival (OS) was 94 % (CI95%[86;100]) for those without vascular invasion versus 79%(CI95%[57;100]) for others. Multivariate analysis demonstrated correlation of metastatic status and presence of vascular invasion with survival. The histopathological description of peritumoral vascular invasion in surgical specimens of osteosarcoma after neoadjuvant chemotherapy can be considered a prognostic factor and could indicate modification of the postoperative therapeutic strategy. C1 [Benezech, Sarah] Institut d’Hematologie et Oncologie Pediatrique, 1 Place Joseph Renaut, 69008 Lyon, France. [Chabaud, Sylvie] Centre Regional Leon BerardLyon, France. [Chambon, Fanny] Centre Hospitalier Universitaire de Clermont-FerrandClermont-Ferrand, France. [Dijoud, Frederique] Hopital Femme-Mere-EnfantBron, France. [Chotel, Franck] Hopital Femme-Mere-EnfantBron, France. [Marec-Berard, Perrine] Institut d’Hematologie et Oncologie Pediatrique, 1 Place Joseph Renaut, 69008 Lyon, France. RP Benezech, S (reprint author), Institut d’Hematologie et Oncologie Pediatrique, 69008 Lyon, France. EM benezech.sarah@gmail.com CR Bielack SS, Kempf-Bielack B, Delling G, Exner GU, Flege S, Helmke K, et al., 2002, Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol 20:776–790 Merimsky O, Isakov J, Kollender Y, Nirkin A, Inbar M, Meller I. Vascular invasion in high grade sarcoma of the extremity is associated with short overall survival. Oncol Rep n.d.1998; 5:985–9. Le Deley M-C, Guinebretiere J-M, Gentet J-C, Pacquement H, Pichon F, Marec-Berard P, et al., 2007, SFOP OS94: a randomised trial comparing preoperative high-dose methotrexate plus doxorubicin to high-dose methotrexate plus etoposide and ifosfamide in osteosarcoma patients. Eur J Cancer 43:752–761., DOI 10.1016/j. ejca.2006.10.023 Marroum MC, Huvos AG, Rosen G, 1977, Pathologic aspects of chemotherapy response in the treatment of osteogenic sarcoma. An analysis of two cases. Oncology 34:273–280 Kaplan EL, Meier P, 1958, Nonparametric estimation from incomplete observations. J Am Stat Assoc 53:457–481., DOI 10.2307/ 2281868 Peto R, Pike MC, Armitage P, Breslow NE, Cox DR, Howard SV, et al., 1977, Design and analysis of randomized clinical trials requiring prolonged observation of each patient. II analysis and examples BrJCancer 35:1–39 Cox DR, 1972, Regression models and life tables. J R Stat Soc Ser B 34:187–220 Goorin AM, Perez-Atayde A, Gebhardt M, Andersen JW, Wilkinson RH, Delorey MJ, et al., 1987, Weekly high-dose methotrexate and doxorubicin for osteosarcoma: the Dana-Farber Cancer Institute/the Children’s hospital - study III. J Clin Oncol 5:1178–1184 Provisor AJ, Ettinger LJ, Nachman JB, Krailo MD, Makley JT, Yunis EJ, et al., 1997, Treatment of nonmetastatic osteosarcoma of the extremity with preoperative and postoperative chemotherapy: a report from the Children’s cancer group. J Clin Oncol 15:76–84 Baldini N, Scotlandi K, Barbanti-Brodano G, ManaraMC,Maurici D, Bacci G, et al., 1995, Expression of P-glycoprotein in highgrade osteosarcomas in relation to clinical outcome. N Engl J Med 333:1380–1385., DOI 10.1056/NEJM199511233332103 Arndt CAS, Rose PS, Folpe AL, Laack NN, 2012, Common musculoskeletal tumors of childhood and adolescence. Mayo Clin Proc 87:475–487., DOI 10.1016/j.mayocp.2012.01.015 Davis AM, Bell RS, Goodwin PJ, 1994, Prognostic factors in osteosarcoma: a critical review. J Clin Oncol 12:423–431 Rosen G, Caparros B, Huvos AG, Kosloff C, Nirenberg A, Cacavio A, et al., 1982, Preoperative chemotherapy for osteogenic sarcoma: selection of postoperative adjuvant chemotherapy based on the response of the primary tumor to preoperative chemotherapy. Cancer 49:1221–1230 Smeland S,Muller C, Alvegard TA,Wiklund T,Wiebe T, Bjork O, et al., 2003, Scandinavian sarcoma group osteosarcoma study SSG VIII: prognostic factors for outcome and the role of replacement salvage chemotherapy for poor histological responders. Eur J Cancer 39:488–494 Winkler K, Beron G, Kotz R, Salzer-KuntschikM, Beck J, BeckW, et al., 1984, Neoadjuvant chemotherapy for osteogenic sarcoma: results of a cooperative German/Austrian study. J Clin Oncol 2: 617–624 JuergensH, Kosloff C, Nirenberg A, MehtaBM, HuvosAG, Rosen G, 1981, Prognostic factors in the response of primary osteogenic sarcoma to preoperative chemotherapy, high-dose methotrexate with citrovorum factor). Natl Cancer Inst Monogr 221–6 Guillon MAM, Mary PMJ, Brugiere L, Marec-Berard P, Pacquement HD, Schmitt C, et al., 2011, Clinical characteristics and prognosis of osteosarcoma in young children: a retrospective series of 15 cases. BMC Cancer 11:407., DOI 10.1186/1471-2407- 11-407 Saeter G, Elomaa I,Wahlqvist Y, Alvegard TA,Wiebe T, Monge O, et al., 1997, Prognostic factors in bone sarcomas. Acta Orthop Scand Suppl 273:156–160 Janeway KA, Barkauskas DA, Krailo MD, Meyers PA, Schwartz CL, Ebb DH, et al., 2012, Outcome for adolescent and young adult patients with osteosarcoma: a report from the Children’s oncology group. Cancer 118:4597–4605., DOI 10.1002/cncr.27414 Bacci G, Ferrari S, Sangiorgi L, Picci P, Casadei R, Orlandi M, et al., 1994, Prognostic significance of serum lactate dehydrogenase in patients with osteosarcoma of the extremities. J Chemother 6:204– 210 Bacci G, Longhi A, Ferrari S, Lari S, Manfrini M, Donati D, et al., 2002, Prognostic significance of serum alkaline phosphatase in osteosarcoma of the extremity treated with neoadjuvant chemotherapy: recent experience at Rizzoli institute. Oncol Rep 9:171–175 Chen J, Sun M-X, Hua Y-Q, Cai Z-D, 2014, Prognostic significance of serum lactate dehydrogenase level in osteosarcoma: a meta- analysis. J Cancer Res Clin Oncol 0–5., DOI 10.1007/s00432-014- 1644-0 Trieb K, Kotz R, 2001, Proteins expressed in osteosarcoma and serum levels as prognostic factors. Int J Biochem Cell Biol 33: 11–17., DOI 10.1016/S1357-2725(00)00066-2 Wunder JS, Gokgoz N, Parkes R, Bull SB, Eskandarian S, Davis AM, et al., 2005, TP53 mutations and outcome in osteosarcoma: a prospective, multicenter study. J Clin Oncol 23:1483–1490., DOI 10. 1200/JCO.2005.04.074 Navalkele P, Jones SM, Jones JK, Salazar JD, Toy PC, Iyer RV, et al., 2013, Osteosarcoma tumor thrombus: a case report with a review of the literature. Tex Heart Inst J 40:75–78 Alvegard TA, Berg NO, Baldetorp B, Ferno M, Killander D, Ranstam J, et al., 1990, Cellular DNA content and prognosis of high-grade soft tissue sarcoma: the Scandinavian sarcoma group experience. J Clin Oncol 8:538–547 Lack EE, Steinberg SM, White DE, Kinsella T, Glatstein E, Chang AE, et al., 1989, Extremity soft tissue sarcomas: analysis of prognostic variables in 300 cases and evaluation of tumor necrosis as a factor in stratifying higher-grade sarcomas. J Surg Oncol 41:263–273 Gustafson P, Rydholm A, Willem H, Baldetorp B, Ferno M, Akerman M, 1993, Liposarcoma: a population-based epidemiologic and prognostic study of features of 43 patients, including tumor DNA content. Int J Cancer 55:541–546., DOI 10. 1002/ijc.2910550404 Halling AC, Wollan PC, Pritchard DJ, Vlasak R, Nascimento AG, 1996, Epithelioid sarcoma: a clinicopathologic review of 55 cases. Mayo Clin Proc 71:636–642., DOI 10.4065/71.7.636 Gustafson P, Willen H, Baldetorp B, Ferno M, Akerman M, Rydholm A, 1992, Soft tissue leiomyosarcoma: a populationbased epidemiologic and prognostic study of 48 patients, including cellular DNA content. Cancer 70:114–119., DOI 10.1002/1097- 0142(19920701)70:1<114::AID-CNCR2820700119>3.0.CO;2-U Carneiro A, Bendahl PO, Engellau J, Domanski HA, Fletcher CD, Rissler P, et al., 2011, A prognostic model for soft tissue sarcoma of the extremities and trunk wall based on size, vascular invasion, necrosis, and growth pattern. Cancer 117:1279–1287., DOI 10. 1002/cncr.25621 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 847 EP 852 DI 10.1007/s12253-016-0074-5 PG 6 ER PT J AU Zhu, L Liu, J Shenglin, M AF Zhu, Lucheng Liu, Jihong Shenglin, Ma TI Fluoropyrimidine-Based Chemotherapy as First-Line Treatment for Advanced Gastric Cancer: a Bayesian Network Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Advanced gastric cancer; Fluoropyrimidine; S-1; Capecitabine; Network meta-analysis ID Advanced gastric cancer; Fluoropyrimidine; S-1; Capecitabine; Network meta-analysis AB Fluoropyrimidine-based regimens are the most common treatments in advanced gastric cancer. We used a Bayesian network meta-analysis to identify the optimal fluoropyrimidine-based chemotherapy by comparing their relative efficacy and safety. We systematically searched databases and extracted data from randomized controlled trials, which compared fluoropyrimidine-based regimens as firstline treatment in AGC. The main outcomes were overall survival (OS), progression-free survival (PFS), overall response rate (ORR), and grade 3 or 4 adverse events (AEs). A total of 12 RCTs of 4026 patients were included in our network metaanalysis. Pooled analysis showed S-1 and capecitabine had a significant OS benefit over 5-Fu, with hazard ratios of 0.90 (95%CI = 0.81–0.99) and 0.88 (95%CI = 0.80–0.96), respectively. The result also exhibited a trend that S-1 and capecitabine prolonged PFS in contrast to 5-Fu, with hazard ratios of 0.84 (95%CI = 0.66–1.02) and 0.84 (95%CI = 0.65–1.03), respectively. Additionally, all the three fluoropyrimidinebased regimens were similar in terms of ORR and grade 3 or 4 AEs. Compared with regimens based on 5-Fu, regimens based on S-1 or capecitabine demonstrated a significant OS improvement without compromise of AEs as first-line treatment in AGC in Asian population. S-1 and capecitabine can be interchangeable according their different emphasis on AEs. C1 [Zhu, Lucheng] Hangzhou First People’s Hospital, Department of Medical Oncology, No.261, Huansha Road, Shangcheng District, 310006 Hangzhou, China. [Liu, Jihong] Hangzhou First People’s Hospital, Department of Medical Oncology, No.261, Huansha Road, Shangcheng District, 310006 Hangzhou, China. [Shenglin, Ma] Hangzhou First People’s Hospital, Department of Medical Oncology, No.261, Huansha Road, Shangcheng District, 310006 Hangzhou, China. RP Shenglin, M (reprint author), Hangzhou First People’s Hospital, Department of Medical Oncology, 310006 Hangzhou, China. EM mashenglin@outlook.com CR Jemal A, Siegel R, Xu J,Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60(5):277–300., DOI 10.3322/caac.20073 Rivera F, Vega-Villegas ME, Lopez-Brea MF, 2007, Chemotherapy of advanced gastric cancer. Cancer Treat Rev 33(4):315–324., DOI 10.1016/j.ctrv.2007.01.004 Murad AM, Santiago FF, Petroianu A, Rocha PR, Rodrigues MA, Rausch M, 1993, Modified therapy with 5-fluorouracil, doxorubicin, and methotrexate in advanced gastric cancer. Cancer 72(1):37–41 Pyrhonen S, Kuitunen T, Nyandoto P, Kouri M, 1995, Randomised comparison of fluorouracil, epidoxorubicin and methotrexate, FEMTX, plus supportive care with supportive care alone in patients with non-resectable gastric cancer. Br J Cancer 71(3):587–591 Glimelius B, Ekstrom K, Hoffman K, GrafW, Sjoden PO, Haglund U, Svensson C, Enander LK, Linne T, Sellstrom H, Heuman R, 1997, Randomized comparison between chemotherapy plus best supportive care with best supportive care in advanced gastric cancer. Ann Oncol 8(2):163–168 Van Cutsem E, Moiseyenko VM, Tjulandin S, Majlis A, Constenla M, Boni C, Rodrigues A, Fodor M, Chao Y, Voznyi E, Risse ML, Ajani JA, 2006, Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 study group. J Clin Oncol 24(31):4991–4997., DOI 10.1200/jco.2006.06. 8429 Cunningham D, Starling N, Rao S, Iveson T, NicolsonM, Coxon F, Middleton G, Daniel F, Oates J, Norman AR, 2008, Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med 358(1):36–46., DOI 10.1056/NEJMoa073149 Ajani JA, Rodriguez W, Bodoky G, Moiseyenko V, Lichinitser M, Gorbunova V, Vynnychenko I, Garin A, Lang I, Falcon S, 2010, Multicenter phase III comparison of cisplatin/S-1 with cisplatin/ infusional fluorouracil in advanced gastric or gastroesophageal adenocarcinoma study: the FLAGS trial. J Clin Oncol 28(9):1547– 1553., DOI 10.1200/jco.2009.25.4706 Boku N, Yamamoto S, Fukuda H, Shirao K, Doi T, Sawaki A, Koizumi W, Saito H, Yamaguchi K, Takiuchi H, Nasu J, Ohtsu A, 2009, Fluorouracil versus combination of irinotecan plus cisplatin versus S-1 in metastatic gastric cancer: a randomised phase 3 study. Lancet Oncol 10(11):1063–1069., DOI 10.1016/s1470-2045(09, 70259-1 Cervantes A, Roda D, Tarazona N, Rosello S, Perez-Fidalgo JA, 2013, Current questions for the treatment of advanced gastric cancer. Cancer Treat Rev 39(1):60–67., DOI 10.1016/j.ctrv.2012.09.007 Pieters A, Laurent S, Dero I, Van Damme N, Peeters M, 2008, The role of oral fluoropyrimidines in the treatment of advanced gastric cancer. Acta Gastroenterol Belg 71(4):361–366 Jin M, Lu H, Li J, Shen L, Chen Z, Shi Y, Song S, Qin S, Liu J, Ouyang X, 2008, Randomized 3-armed phase III study of S-1 monotherapy versus S-1/CDDP, SP, versus 5-FU/CDDP, FP, in patients, pts, with advanced gastric cancer, AGC): SC-101 study. J Clin Oncol 26(Suppl 15):4533 Kang YK, Kang WK, Shin DB, Chen J, Xiong J, Wang J, Lichinitser M, Guan Z, Khasanov R, Zheng L, Philco-Salas M, Suarez T, Santamaria J, Forster G, McCloud PI, 2009, Capecitabine/cisplatin versus 5-fluorouracil/cisplatin as first-line therapy in patients with advanced gastric cancer: a randomised phase III noninferiority trial. Ann Oncol 20(4):666–673., DOI 10. 1093/annonc/mdn717 Lee JL, Kang YK, Kang HJ, Lee KH, Zang DY, Ryoo BY, Kim JG, Park SR, Kang WK, Shin DB, Ryu MH, Chang HM, Kim TW, Baek JH, Min YJ, 2008, A randomised multicentre phase II trial of capecitabine vs S-1 as first-line treatment in elderly patients with metastatic or recurrent unresectable gastric cancer. Br J Cancer 99(4):584–590., DOI 10.1038/sj.bjc.6604536 Kim GM, Jeung HC, Rha SY, Kim HS, Jung I, Nam BH, Lee KH, Chung HC, 2012, A randomized phase II trial of S-1-oxaliplatin versus capecitabine-oxaliplatin in advanced gastric cancer. Eur J Cancer 48(4):518–526., DOI 10.1016/j.ejca.2011.12.017 HeMM,Wu WJ,Wang F,Wang ZQ, Zhang DS, Luo HY, QiuMZ, Wang FH, Ren C, Zeng ZL, Xu RH, 2013, S-1-based chemotherapy versus capecitabine-based chemotherapy as first-line treatment for advanced gastric carcinoma: a meta-analysis. PLoS One 8(12): e82798., DOI 10.1371/journal.pone.0082798 Caldwell DM, Ades AE, Higgins JP, 2005, Simultaneous comparison of multiple treatments: combining direct and indirect evidence. BMJ 331(7521):897–900., DOI 10.1136/bmj.331.7521.897 Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR, 2007, Practical methods for incorporating summary time-to-event data into meta-analysis. Trials 8:16., DOI 10.1186/1745-6215-8-16 Lu G, Ades AE, 2004, Combination of direct and indirect evidence in mixed treatment comparisons. Stat Med 23(20):3105–3124., DOI 10.1002/sim.1875 Fryback DG, Stout NK, Rosenberg MA, 2001, An elementary introduction to Bayesian computing using WinBUGS. Int J Technol Assess Health Care 17(1):98–113 Woods BS, Hawkins N, ScottDA(2010, Networkmeta-analysis on the log-hazard scale, combining count and hazard ratio statistics accounting for multi-arm trials: a tutorial. BMC Med Res Methodol 10:54., DOI 10.1186/1471-2288-10-54 Spiegelhalter DJ, Best NG, Carlin BP, Van Der Linde A, 2002, Bayesian measures of model complexity and fit. J Roy Stat Soc B 64(4):583–639., DOI 10.1111/1467-9868.00353 Brooks SP, Gelman A, 1998, General methods for monitoring convergence of iterative simulations. J Comput Graph Stat 7(4):434–455 Higgins J, Jackson D, Barrett J, Lu G, Ades A, White I, 2012, Consistency and inconsistency in network meta-analysis: concepts and models for multi-arm studies. Res Synth Methods 3(2):98–110 Dias S,Welton N, Caldwell D, Ades A, 2010, Checking consistency inmixed treatment comparison meta-analysis. Stat Med 29(7–8): 932–944 Xu RH, Sun GP, Lu HS, Peng LY, Xu JM, Zhong MZ, Zhang HL, Yu SY, LiW, Hu XH,Wang JJ, ChengY, Zhou JT, Guo ZQ, Guan Z, 2013, A phase III study of S-1 plus cisplatin versus fluorouracil plus cisplatin in patients with advanced gastric or gastroesophageal junction adenocarcinoma. J Clin Oncol 31:suppl; abstr 4025 Huang D, Ba Y, Xiong J, Xu N, Yan Z, Zhuang Z, Yu Z, Wan H, Zhang Y, Deng T, Zheng R, Guo Z, Hu C, Wang M, Yu Z, Yao Y, Meng J, 2013, A multicentre randomised trial comparing weekly paclitaxel + S-1 with weekly paclitaxel +5-fluorouracil for patients with advanced gastric cancer. Eur J Cancer 49(14):2995–3002., DOI 10.1016/j.ejca.2013.05.021 Ocvirk J, Rebersek M, Skof E, Hlebanja Z, Boc M, 2012, Randomized prospective phase II study to compare the combination chemotherapy regimen epirubicin, cisplatin, and 5-fluorouracil with epirubicin, cisplatin, and capecitabine in patients with advanced or metastatic gastric cancer. Am J Clin Oncol 35(3):237– 241., DOI 10.1097/COC.0b013e31820dc0b0 Nishikawa K, Morita S, Matsui T, Kobayashi M, Takeuchi Y, Takahashi I, Sato S, Miyashita Y, Tsuburaya A, Sakamoto J, Kakeji Y, Baba H, 2012, A randomized phase-II trial comparing sequential and concurrent paclitaxel with oral or parenteral fluorinated pyrimidines for advanced or metastatic gastric cancer. Gastric Cancer 15(4):363–369., DOI 10.1007/s10120-011-0124-3 Sanofi, 2011, Docetaxel and Oxaliplatin in Gastric Cancer. ClinicalTrialsgov:NCT00382720 Cocconi G, DeLisi V, Di Blasio B, 1982, Randomized comparison of 5-FU alone or combined with mitomycin and cytarabine, MFC, in the treatment of advanced gastric cancer. Cancer Treat Rep 66(6): 1263–1266 Kim NK, Park YS, Heo DS, Suh C, Kim SY, Park KC, Kang YK, Shin DB, Kim HT, Kim HJ, et al., 1993, A phase III randomized study of 5-fluorouracil and cisplatin versus 5-fluorouracil, doxorubicin, and mitomycin C versus 5-fluorouracil alone in the treatment of advanced gastric cancer. Cancer 71(12):3813–3818 Webb A, Cunningham D, Scarffe JH, Harper P, Norman A, Joffe JK, Hughes M, Mansi J, Findlay M, Hill A, Oates J, Nicolson M, Hickish T, O'Brien M, Iveson T, Watson M, Underhill C, Wardley A, Meehan M, 1997, Randomized trial comparing epirubicin, cisplatin, and fluorouracil versus fluorouracil, doxorubicin, and methotrexate in advanced esophagogastric cancer. J Clin Oncol 15(1): 261–267 Ross P, Nicolson M, CunninghamD,Valle J, SeymourM, Harper P, Price T, Anderson H, Iveson T, Hickish T, Lofts F, Norman A, 2002, Prospective randomized trial comparing mitomycin, cisplatin, and protracted venous-infusion fluorouracil, PVI 5-FU, with epirubicin, cisplatin, and PVI 5-FU in advanced esophagogastric cancer. J Clin Oncol 20(8):1996–2004 Ohtsu A, Shimada Y, Shirao K, Boku N, Hyodo I, Saito H, Yamamichi N, Miyata Y, Ikeda N, Yamamoto S, Fukuda H, Yoshida S, 2003, Randomized phase III trial of fluorouracil alone versus fluorouracil plus cisplatin versus uracil and tegafur plus mitomycin in patients with unresectable, advanced gastric cancer: the Japan clinical oncology group study, JCOG9205). J Clin Oncol 21(1):54–59 Okines AF, Norman AR, McCloud P, Kang YK, Cunningham D, 2009, Meta-analysis of the REAL-2 and ML17032 trials: evaluating capecitabine-based combination chemotherapy and infused 5- fluorouracil-based combination chemotherapy for the treatment of advanced oesophago-gastric cancer. Ann Oncol 20(9):1529–1534., DOI 10.1093/annonc/mdp047 Li DH, Pan ZK, Ye F, An HX, Wu JX, 2014, S-1-based versus 5- FU-based chemotherapy as first-line treatment in advanced gastric cancer: a meta-analysis of randomized controlled trials. Tumour Biol 35(8):8201–8208., DOI 10.1007/s13277-014-2099-2 Wagner AD, Unverzagt S, Grothe W, Kleber G, Grothey A, Haerting J, Fleig WE, 2010, Chemotherapy for advanced gastric cancer. Cochrane Database Syst Rev, 3):Cd004064., DOI 10.1002/ 14651858.CD004064.pub3 Ye JX, Liu AQ, Ge LY, Zhou SZ, Liang ZG, 2014, Effectiveness and safety profile of S-1-based chemotherapy compared with capecitabine-based chemotherapy for advanced gastric and colorectal cancer: a meta-analysis. Exp Ther Med 7(5):1271–1278., DOI 10. 3892/etm.2014.1576 Yang J, Zhou Y,Min K, Yao Q, Xu CN, 2014, S-1-based vs non-S- 1-based chemotherapy in advanced gastric cancer: a meta-analysis. World J Gastroenterol 20(33):11886–11893., DOI 10.3748/wjg.v20. i33.11886 Cipriani A, Furukawa TA, Salanti G, Geddes JR, Higgins JP, Churchill R, Watanabe N, Nakagawa A, Omori IM, McGuire H, Tansella M, Barbui C, 2009, Comparative efficacy and acceptability of 12 new-generation antidepressants: a multiple-treatments meta- analysis. Lancet 373(9665):746–758., DOI 10.1016/s0140- 6736(09)60046-5 Ajani JA, Faust J, Ikeda K,Yao JC, Anbe H, Carr KL, Houghton M, Urrea P, 2005, Phase I pharmacokinetic study of S-1 plus cisplatin in patients with advanced gastric carcinoma. J Clin Oncol 23(28): 6957–6965., DOI 10.1200/jco.2005.01.917 Daigo S, Takahashi Y, Fujieda M, Ariyoshi N, Yamazaki H, Koizumi W, Tanabe S, Saigenji K, Nagayama S, Ikeda K, Nishioka Y, Kamataki T, 2002, A novel mutant allele of the CYP2A6 gene, CYP2A6*11 , found in a cancer patient who showed poor metabolic phenotype towards tegafur. Pharmacogenetics 12(4):299–306 Yoshida R, Nakajima M, Nishimura K, Tokudome S, Kwon JT, Yokoi T, 2003, Effects of polymorphism in promoter region of human CYP2A6 gene, CYP2A6*9, on expression level of messenger ribonucleic acid and enzymatic activity in vivo and in vitro. Clin Pharmacol Ther 74(1):69–76., DOI 10.1016/ s0009-9236(03)00090-0 El Sayed YM, SadeeW, 1982, Metabolic activation of ftorafur [R, S-1-(tetrahydro-2-furanyl)-5-fluorouracil]: the microsomal oxidative pathway. Biochem Pharmacol 31(18):3006–3008 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 853 EP 861 DI 10.1007/s12253-016-0078-1 PG 9 ER PT J AU Geels, PY Pijnenborg, MAJ Gordon, BMB Fogel, M Altevogt, P Masadah, R Bulten, J van Kempen, CL Massuger, FAGL AF Geels, P Yvette Pijnenborg, M A Johanna Gordon, B M Bart Fogel, Mina Altevogt, Peter Masadah, Rina Bulten, Johan van Kempen, C Leon Massuger, F A G Leon TI L1CAM Expression is Related to Non-Endometrioid Histology, and Prognostic for Poor Outcome in Endometrioid Endometrial Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial carcinoma; Immunohistochemistry; L1CAM; Histopathological diagnosis; Prognostic value; Non-endometrioid ID Endometrial carcinoma; Immunohistochemistry; L1CAM; Histopathological diagnosis; Prognostic value; Non-endometrioid AB The majority of endometrial carcinomas are classified as Type I endometrioid endometrial carcinomas (EECs) and have a good prognosis. Type II non-endometrioid endometrial carcinomas (NEECs) have a significant worse outcome. Yet, 20 % of the EECs are associated with an unexplained poor outcome. The aim of this study was to determine if L1CAM expression, a recently reported biomarker for aggressive tumor behavior in endometrial carcinoma, was associated with clinicopathological features of EECs. A total of 103 patients diagnosed as EEC at the Radboud University Medical Centre, based on the pathology report were selected. L1CAM status of these tumors was determined, and histologic slides were reviewed by two expert pathologists. L1CAM-positivity was observed in 17 % (18/103). Review of the diagnostic slides revealed that 11 out of these 18 L1CAM-positive tumors (61 %) contained a serous- or mixed carcinoma component that was not initially mentioned in the pathology report. L1CAM-expression was associated with advanced age, poor tumor grade, and lymphovascular space invasion. A worse five year progression free survival rate was observed for patients with L1CAM-positive tumors (55.6 % for the L1CAM-positive group, compared to 83.3 % for the L1CAM-negative group P = 0.01). L1CAM expression carries prognostic value for histologically classified EEC and supports the identification of tumors with a NEEC component. C1 [Geels, P Yvette] Radboud University Nijmegen, Medical Centre, Department of Obstetrics and GynaecologyNijmegen, The Netherlands. [Pijnenborg, M A Johanna] Elisabeth-TweeSteden Hospital, Department of Obstetrics and Gynaecology, 5000 Tilburg, LA, The Netherlands. [Gordon, B M Bart] Radboud University Nijmegen, Medical Centre, Department of Obstetrics and GynaecologyNijmegen, The Netherlands. [Fogel, Mina] Kaplan Medical Centre, Department of PathologyRehovot, Israel. [Altevogt, Peter] German Cancer Research Centre, Tumour Immunology Program, D015, 69120 Heidelberg, Germany. [Masadah, Rina] Hasanuddin University Hospital, Department of PathologyMakassar, Indonesia. [Bulten, Johan] Radboud University Nijmegen, Medical Centre, Department of PathologyNijmegen, The Netherlands. [van Kempen, C Leon] Radboud University Nijmegen, Medical Centre, Department of PathologyNijmegen, The Netherlands. [Massuger, F A G Leon] Radboud University Nijmegen, Medical Centre, Department of Obstetrics and GynaecologyNijmegen, The Netherlands. RP Pijnenborg, MAJ (reprint author), Elisabeth-TweeSteden Hospital, Department of Obstetrics and Gynaecology, 5000 Tilburg, The Netherlands. EM H.Pijnenborg@planet.nl CR Parkin DM, Bray F, Ferlay J, et al., 2005, Global cancer statistics, 2002. CA Cancer J Clin 55(2):74–108 Prat J, Gallardo A, Cuatrecasas M, et al., 2007, Endometrial carcinoma: pathology and genetics. Pathology 39(1):72–87 Engelsen IB, Akslen LA, Salvesen HB, 2009, Biologic markers in endometrial cancer treatment. APMIS 117(10):693–707 Geels YP, Pijnenborg JM, van den Berg-van Erp SH, et al., 2012, Endometrioid endometrial carcinoma with atrophic endometrium and poor prognosis. Obstet Gynecol 120(5):1124–1131 Huszar M, Pfeifer M, Schirmer U, et al., 2010, Up-regulation of L1CAM is linked to loss of hormone receptors and E-cadherin in aggressive subtypes of endometrial carcinomas. J Pathol 220(5): 551–561 Zeimet AG, Reimer D, Huszar M, et al., 2013, L1CAM in earlystage type I endometrial cancer: results of a large multicenter evaluation. J Natl Cancer Inst 105(15):1142–1150 Brummendorf T, Kenwrick S, Rathjen FG, 1998, Neural cell recognition molecule L1: from cell biology to human hereditary brain malformations. Curr Opin Neurobiol 8(1):87–97 Bosse T, Nout RA, Stelloo E, Dreef E, Nijman HW, Jurgenliemk- Schulz IM, Jobsen JJ, Creutzberg CL, Smit VT, 2014, L1 cell adhesion molecule is a strong predictor for distant recurrence and overall survival in early stage endometrial cancer: pooled PORTEC trial results. Eur J Cancer 50(15):2602–2610 FogelM, Gutwein P,Mechtersheimer S, et al., 2003, L1 expression as a predictor of progression and survival in patients with uterine and ovarian carcinomas. Lancet 362(9387):869–875 Trovik J,Wik E,Werner HM, Krakstad C, Helland H, Vandenput I, Njolstad TS, Stefansson IM, Marcickiewicz J, Tingulstad S, 2013, Staff AC; MoMaTEC study group, Amant F, Akslen LA, Salvesen HB. Hormone receptor loss in endometrial carcinoma curettage predicts lymph node metastasis and poor outcome in prospective multicentre trial. Eur J Cancer 49(16):3431–3441 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 863 EP 868 DI 10.1007/s12253-016-0047-8 PG 6 ER PT J AU Roncati, L Barbolini, G Gatti, MA Pusiol, T Piscioli, F Maiorana, A AF Roncati, Luca Barbolini, Giuseppe Gatti, Morena Antonietta Pusiol, Teresa Piscioli, Francesco Maiorana, Antonio TI The Uncontrolled Sialylation is Related to Chemoresistant Metastatic Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Invasive ductal carcinoma; Sialic acid; Sialomucins; ST3GAL5 sialyltransferase; Sialidase (neuraminidase); Alcian Blue; DDD-Fast Blue B; Mercury Orange; Elemental microanalysis; Sulphur (S) ID Breast cancer; Invasive ductal carcinoma; Sialic acid; Sialomucins; ST3GAL5 sialyltransferase; Sialidase (neuraminidase); Alcian Blue; DDD-Fast Blue B; Mercury Orange; Elemental microanalysis; Sulphur (S) AB Among the scientific communities, there is a convergence of results supporting a direct relationship between dysregulated sialylation and poor prognosis in many human cancers. For this reason, we have retrospectively investigated 169 cases of invasive ductal carcinoma of the breast, coming from female patients aged between 31 and 76 years old. The whole series was subdivided into two prognostic groups: the first group consisted of 138 patients, who showed a post-treatment survival time more than 5 years, while the second group was made up by 31 patients, died within 5 years despite of chemotherapy. All the surgical specimens were fixed in 10%neutral buffered formalin, paraffin embedded and, then, submitted to routinely haematoxylin/eosin staining and to a further histochemical (Alcian Blue, DDD-Fast Blue B, Mercury Orange), immunohistochemical (ST3GAL5 sialyltransferase, Ki67, c-erbB2, ER, PR) and chemico-elemental characterization. In the 31 cases of breast cancer belonging to the second group, an overexpression of sialomucins and sialyltransferases has been detected. Our results lead us to support that in aggressive chemoresistant breast cancers, the altered expression of sialic acid, due to an uncontrolled sialylation, creates an excessive negative charge on cell membranes, which stimulates repulsion between neoplastic cells and their subsequent access into the blood stream. This event implies an earlymetastatization and a rapid disease progression with fatal outcome. The early application of Alcian Blue stain on diagnostic biopsies of breast cancer is able to cheaply reveal the sialomucin accumulations, providing for the disease course. C1 [Roncati, Luca] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, I-41124 Modena, MO, Italy. [Barbolini, Giuseppe] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, I-41124 Modena, MO, Italy. [Gatti, Morena Antonietta] National Research Council, Institute of Science and Technology for CeramicsFaenza, RA, Italy. [Pusiol, Teresa] Provincial Health Care Services, Santa Maria del Carmine Hospital, Institute of PathologyRovereto, TN, Italy. [Piscioli, Francesco] Provincial Health Care Services, Santa Maria del Carmine Hospital, Institute of PathologyRovereto, TN, Italy. [Maiorana, Antonio] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, I-41124 Modena, MO, Italy. RP Roncati, L (reprint author), University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, I-41124 Modena, Italy. EM emailmedical@gmail.com CR Schauer R, 2000, Achievements and challenges of sialic acid research. Glycoconj J 17:485–499 Cazet A, Julien S, Bobowski M, Krzewinski-Recchi MA, Harduin- Lepers A, Groux-Degroote S, et al., 2010, Consequences of the expression of sialylated antigens in breast cancer. Carbohydr Res 345:1377–1383 Christiansen MN, Chik J, Lee L, Anugraham M, Abrahams JL, Packer NH, 2014, Cell surface protein glycosylation in cancer. Proteomics 14:525–546 Miyagi T, Takahashi K, Hata K, Shiozaki K, Yamaguchi K, 2012, Sialidase significance for cancer progression. Glycoconj J 29:567– 577 Gatti AM,Montanari S, 2005, Risk assessment of microparticles and nanoparticles and human health. In: Nalwa HS, ed, Handbook of nanostructured biomaterials and their applications in nanobiotechnology. American Scientific Publishers, Portland, pp. 347–369 Elston CW, Ellis IO, 2002, Pathological prognostic factors in breast cancer. The value of histological grade in breast cancer: experience from a large study with long-term follow-up Histopathology 41:154– 161 Tian Y, Esteva FJ, Song J, Zhang H, 2012, Altered expression of sialylated glycoproteins in breast cancer using hydrazide chemistry and mass spectrometry. Mol Cell Proteomics 11:M111.011403 Kyselova Z, Mechref Y, Kang P, Goetz JA, Dobrolecki LE, Sledge GW, et al., 2008, Breast cancer diagnosis and prognosis through quantitative measurements of serum glycan profiles. Clin Chem 54: 1166–1175 DwekMV, Lacey HA, Leathem AJ, 1998, Breast cancer progression is associated with a reduction in the diversity of sialylated and neutral oligosaccharides. Clin Chim Acta 271:191–202 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 869 EP 873 DI 10.1007/s12253-016-0057-6 PG 5 ER PT J AU Jo, SY Kim, SM Yoo, JN Lee, HS AF Jo, Sol Yun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutation of MED25, a Transcription Regulator, and its Mutational Heterogeneity in Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Makinen N, MehineM, Tolvanen J, Kaasinen E, Li Y, Lehtonen HJ, Gentile M, Yan J, Enge M, Taipale M,Aavikko M, Katainen R, Virolainen E, Bohling T, Koski TA, Launonen V, Sjoberg J, Taipale J, Vahteristo P, Aaltonen LA, 2011, MED12, the mediator complex subunit 12 gene, Is mutated at high frequency in uterine leiomyomas. Science 334: 252–255. Lim WK, Ong CK, Tan J, Thike AA, Ng CC, Rajasegaran V, Myint SS, Nagarajan S, Nasir ND, McPherson JR, Cutcutache I, Poore G, Tay ST, Ooi WS, Tan VK, Hartman M, Ong KW, Tan BK, Rozen SG, Tan PH, Tan P, Teh BT, 2014, Exome sequencing identifies highly recurrent MED12 somatic mutations in breast fibroadenoma. Nat Genet 46:877–880 Poss ZC, Ebmeier CC, Taatjes DJ, 2013, The Mediator complex and transcription regulation. Crit Rev Biochem Mol Biol 48:575–608 Lee HK, Park UH, Kim EJ, Um SJ, 2007, MED25 is distinct from TRAP220/MED1 in cooperating with CBP for retinoid receptor activation. EMBO J 26:3545–3557 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Turunen M, Spaeth JM, Keskitalo S, Park MJ, Kivioja T, Clark AD, Makinen N, Gao F, Palin K, Nurkkala H, Vaharautio A, Aavikko M, Kampjarvi K, Vahteristo P, Kim CA, Aaltonen LA, Varjosalo M, Taipale J, Boyer TG, 2014, Uterine leiomyomalinked MED12 mutations disrupt mediator-associated CDK activity. Cell Rep 7:654–660 CalinGA, Gafa R, TibilettiMG, Herlea V, BecheanuG, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression in microsatellite instability high, MSI-H, colon cancers correlates with clinico-pathological parameters: A study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89:230–235 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 875 EP 876 DI 10.1007/s12253-016-0092-3 PG 2 ER PT J AU Choi, JE Kim, SM Yoo, JN Lee, HS AF Choi, Ji Eun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutation of ASPM Gene in Colorectal Cancers with Regional Heterogeneity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, Xie M, Zhang Q, McMichael JF, Wyczalkowski MA, Leiserson MD, Miller CA, Welch JS, Walter MJ, Wendl MC, Ley TJ, Wilson RK, Raphael BJ, Ding L, 2013, Mutational landscape and significance across 12 major cancer types. Nature 502:333–339 Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, Meyerson M, Gabriel SB, Lander ES, Getz G, 2014, Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505:495–501 Gonzalez-Perez A, Perez-Llamas C, Deu-Pons J, Tamborero D, Schroeder MP, Jene-Sanz A, Santos A, Lopez-Bigas N, 2013, IntOGen-mutations identifies cancer drivers across tumor types. Nat Methods 10:1081–1082 Fish JL, Kosodo Y, Enard W, Paabo S, Huttner WB, 2006, Aspm specifically maintains symmetric proliferative divisions of neuroepithelial cells. Proc Natl Acad Sci U S A 103:10438–10443 Wang WY, Hsu CC, Wang TY, Li CR, Hou YC, Chu JM, Lee CT, Liu MS, JJ S, Jian KY, Huang SS, Jiang SS, Shan YS, Lin PW, Shen YY, Lee MT, Chan TS, Chang CC, Chen CH, Chang IS, Lee YL, Chen LT, Tsai KK, 2013, A gene expression signature of epithelial tubulogenesis and a role for ASPM in pancreatic tumor progression. Gastroenterology 145:1110–1120 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Kim MS, JE O, Kim YR, Park SW, Kang MR, Kim SS, Ahn CH, Yoo NJ, Lee SH, 2010, Somatic mutations and losses of expression of microRNA regulation-related genes AGO2 and TNRC6A in gastric and colorectal cancers. J Pathol 221:139–146 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Iwakawa R, Kohno T, Totoki Y, Shibata T, Tsuchihara K, Mimaki S, Tsuta K, Narita Y, Nishikawa R, Noguchi M, Harris CC, Robles AI, Yamaguchi R, Imoto S, Miyano S, Totsuka H, Yoshida T, Yokota J, 2015, Expression and clinical significance of genes frequently mutated in small cell lung cancers defined bywhole exome/ RNA sequencing. Carcinogenesis 36:616–621 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2016 VL 22 IS 4 BP 877 EP 879 DI 10.1007/s12253-016-0108-z PG 3 ER PT J AU Behbahani, DG Ghahhari, MN Javidi, AM Molan, FA Feizi, N Babashah, S AF Behbahani, Dehbashi Golnoush Ghahhari, Mohammadi Nastaran Javidi, Amin Mohammad Molan, Farzi Asghar Feizi, Neda Babashah, Sadegh TI MicroRNA-Mediated Post-Transcriptional Regulation of Epithelial to Mesenchymal Transition in Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Cancer; Epithelial to mesenchymal transition; Post-transcriptional regulation; microRNAs ID Cancer; Epithelial to mesenchymal transition; Post-transcriptional regulation; microRNAs AB Epithelial to mesenchymal transition (EMT) program participates in tissue repair, embryogenesis and numerous pathological conditions, particularly cancer progression and tumor metastasis. A highly complex and strongly controlled post-transcriptionally regulated network of microRNAs (miRNAs) regulates the EMT process. miRNAs are critical parts of the post-transcriptional regulation of gene expression. A set of miRNAs target multiple components of major signaling pathways and downstream effectors of EMT. miRNAs associated with this process are involved in controlling tumor progression and invasiveness either as oncogenes or as tumor suppressors. Since several miRNAs directly affect EMT-related master regulators, they have been discovered to have the potential to be used as biomarkers or targets in EMT-based pathological conditions such as cancer. Therefore, comprehensive understanding of miRNA-EMT correlation with tumor metastatic spread may provide improvements to diagnostic tools or therapeutics for cancer. This review summarizes our current knowledge about some of these important miRNAs and focuses on their specific roles in regulation of the EMT process in cancer. C1 [Behbahani, Dehbashi Golnoush] Tarbiat Modares University, Faculty of Biosciences, Department of Molecular GeneticsTehran, Iran. [Ghahhari, Mohammadi Nastaran] Pasteur Institute of Iran, Department of BiochemistryTehran, Iran. [Javidi, Amin Mohammad] Tarbiat Modares University, Faculty of Biosciences, Department of Molecular GeneticsTehran, Iran. [Molan, Farzi Asghar] Tarbiat Modares University, Faculty of Biosciences, Department of Molecular GeneticsTehran, Iran. [Feizi, Neda] Pasteur Institute of Iran, Laboratory of Influenza ResearchTehran, Iran. [Babashah, Sadegh] Tarbiat Modares University, Faculty of Biosciences, Department of Molecular GeneticsTehran, Iran. RP Babashah, S (reprint author), Tarbiat Modares University, Faculty of Biosciences, Department of Molecular Genetics, Tehran, Iran. EM s.babashah@modares.ac;sadegh.babashah@gmail.com CR De Craene B, Berx G, 2013, Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer 13(2): 97–110 Nieto MA, 2013, Epithelial plasticity: a common theme in embryonic and cancer cells. Science 342(6159):1234850 Peinado H, Olmeda D, Cano A, 2007, Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 7(6):415–428 Thiery JP, Acloque H, Huang RY, Nieto MA, 2009, Epithelialmesenchymal transitions in development and disease. Cell 139(5): 871–890 Thiery JP, 2003, Epithelial–mesenchymal transitions in development and pathologies. Curr Opin Cell Biol 15(6):740–746 Ocana OH, Corcoles R, Fabra A, Moreno-Bueno G, Acloque H, Vega S, Barrallo-Gimeno A, Cano A, NietoMA, 2012)Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell 22(6):709–724 Lamouille S, Subramanyam D, Blelloch R, Derynck R, 2013, Regulation of epithelial–mesenchymal and mesenchymal-epithelial transitions by microRNAs. Curr Opin Cell Biol 25(2): 200–207 Thiery JP, 2002, Epithelial–mesenchymal transitions in tumour progression. Nat Rev Cancer 2(6):442–454 Kalluri R, Weinberg RA, 2009, The basics of epithelialmesenchymal transition. J Clin Invest 119(6):1420 Gavert N, Ben-Ze’ev A, 2008, Epithelial–mesenchymal transition and the invasive potential of tumors. Trends Mol Med 14(5):199– 209 Voutsadakis IA, 2016, Epithelial-mesenchymal transition, EMT, and regulation of EMT factors by steroid nuclear receptors in breast cancer: a review and in Silico investigation. J Clin Med 5(1)., DOI 10.3390/jcm5010011 Nieto MA, 2002, The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol 3(3):155–166., DOI 10.1038 /nrm757 Grau Y, Carteret C, Simpson P, 1984)Mutations and chromosomal rearrangements affecting the expression of snail, a Gene involved in embryonic patterning in Drosophila melanogaster. Genetics 108(2):347–360 Peinado H, Ballestar E, Esteller M, Cano A, 2004, Snail mediates E-cadherin repression by the recruitment of the Sin3A/histone deacetylase 1, HDAC1)/HDAC2 complex. Mol Cell Biol 24(1): 306–319 Wang Y, Shi J, Chai K, Ying X, Zhou BP, 2013, The role of snail in EMT and tumorigenesis. Curr Cancer Drug Targets 13(9):963– 972 Hemavathy K, Ashraf SI, Ip YT, 2000, Snail/slug family of repressors: slowly going into the fast lane of development and cancer. Gene 257(1):1–12 Moody SE, Perez D, Pan TC, Sarkisian CJ, Portocarrero CP, Sterner CJ, Notorfrancesco KL, Cardiff RD, Chodosh LA, 2005, The transcriptional repressor snail promotes mammary tumor recurrence. Cancer Cell 8(3):197–209., DOI 10.1016/j. ccr.2005.07.009 Chen WJ, Wang H, Tang Y, Liu CL, Li HL, Li WT, 2010, Multidrug resistance in breast cancer cells during epithelialmesenchymal transition is modulated by breast cancer resistant protein. Chin J Cancer 29(2):151–157 Medici D, Hay ED, Olsen BR, 2008, Snail and slug promote epithelial-mesenchymal transition through beta-catenin-T-cell factor-4-dependent expression of transforming growth factor-beta3. Mol Biol Cell 19(11):4875–4887., DOI 10.1091/mbc.E08-05- 0506 Liu Y-N, Abou-KheirW, Yin JJ, Fang L, Hynes P, Casey O, Hu D, Wan Y, Seng V, Sheppard-Tillman H, 2012, Critical and reciprocal regulation of KLF4 and SLUG in transforming growth factor β-initiated prostate cancer epithelial-mesenchymal transition. Mol Cell Biol 32(5):941–953 Vandewalle C, Van Roy F, Berx G, 2009, The role of the ZEB family of transcription factors in development and disease. Cell Mol Life Sci 66(5):773–787., DOI 10.1007/s00018-008-8465-8 Sanchez-Tillo E, Liu Y, de Barrios O, Siles L, Fanlo L, Cuatrecasas M, Darling DS, Dean DC, Castells A, Postigo A, 2012, EMT-activating transcription factors in cancer: beyond EMT and tumor invasiveness. Cell Mol Life Sci 69(20):3429– 3456., DOI 10.1007/s00018-012-1122-2 Watanabe O, Imamura H, Shimizu T, Kinoshita J, Okabe T, Hirano A, Yoshimatsu K, Konno S, Aiba M, Ogawa K, 2004, Expression of twist and wnt in human breast cancer. Anticancer Res 24(6):3851–3856 Martin TA, Goyal A, Watkins G, Jiang WG, 2005, Expression of the transcription factors snail, slug, and twist and their clinical significance in human breast cancer. Ann Surg Oncol 12(6):488– 496., DOI 10.1245/ASO.2005.04.010 Kwok WK, Ling MT, Lee TW, Lau TC, Zhou C, Zhang X, Chua CW, Chan KW, Chan FL, Glackin C, Wong YC, Wang X, 2005, Up-regulation of TWIST in prostate cancer and its implication as a therapeutic target. Cancer Res 65(12):5153–5162., DOI 10.1158 /0008-5472.CAN-04-3785 Rosivatz E, Becker I, Specht K, Fricke E, Luber B, Busch R, Hofler H, Becker KF, 2002, Differential expression of the epithelial-mesenchymal transition regulators snail, SIP1, and twist in gastric cancer. Am J Pathol 161(5):1881–1891., DOI 10.1016 /S0002-9440(10)64464-1 Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, Savagner P, Gitelman I, Richardson A, Weinberg RA, 2004, Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117(7):927–939., DOI 10.1016 /j.cell.2004.06.006 Babashah S, Soleimani M, 2011, The oncogenic and tumour suppressive roles of microRNAs in cancer and apoptosis. Eur J Cancer 47(8):1127–1137 Baranwal S, Alahari SK, 2010, miRNA control of tumor cell invasion and metastasis. Int J Cancer 126(6):1283–1290 Liu X, Fortin K, Mourelatos Z, 2008, MicroRNAs: biogenesis and molecular functions. Brain Pathol 18(1):113–121 Bartel DP, 2009, MicroRNAs: target recognition and regulatory functions. Cell 136(2):215–233 Babashah S, 2014, MicroRNAs: key regulators of oncogenesis. Springer International Publishing, Switzerland Pencheva N, Tavazoie SF, 2013, Control of metastatic progression by microRNA regulatory networks. Nat Cell Biol 15(6):546– 554 Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M, 2004, Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A 101(9):2999–3004 Huang T, Alvarez A, Hu B, Cheng S-Y, 2013, Noncoding RNAs in cancer and cancer stem cells. Chinese journal of cancer 32(11): 582 Esquela-Kerscher A, Slack FJ, 2006, Oncomirs—microRNAs with a role in cancer. Nat Rev Cancer 6(4):259–269 Sevignani C, Calin GA, Siracusa LD, Croce CM, 2006, Mammalian microRNAs: a small world for fine-tuning gene expression. Mamm Genome 17(3):189–202 Szafranska A, Davison T, John J, Cannon T, Sipos B, Maghnouj A, Labourier E, Hahn S, 2007, MicroRNA expression alterations are linked to tumorigenesis and non-neoplastic processes in pancreatic ductal adenocarcinoma. Oncogene 26(30):4442–4452 Bandyopadhyay S, Mitra R, Maulik U, Zhang MQ, 2010, Development of the human cancer microRNA network. Silence 1(1):6 Zhong X, Coukos G, Zhang L, 2012, miRNAs in human cancer. In: Next-GenerationMicroRNA Expression Profiling Technology Springer, pp 295–306 Zhang B, Pan X, Cobb GP, Anderson TA, 2007, microRNAs as oncogenes and tumor suppressors. Dev Biol 302(1):1–12 Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X, 2008, Characterization ofmicroRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 18(10):997–1006 Diaz-Lopez A, Moreno-Bueno G, Cano A, 2014, Role of microRNA in epithelial to mesenchymal transition and metastasis and clinical perspectives. Cancer Manag Res 6:205 Bullock MD, Sayan AE, Packham GK, Mirnezami AH, 2012, MicroRNAs: critical regulators of epithelial to mesenchymal, EMT, and mesenchymal to epithelial transition, MET, in cancer progression. Biol Cell 104(1):3–12 Ghahhari NM, Babashah S, 2015, Interplay between microRNAs and WNT/beta-catenin signalling pathway regulates epithelialmesenchymal transition in cancer. Eur J Cancer 51(12):1638– 1649., DOI 10.1016/j.ejca.2015.04.021 Ma L, Young J, Prabhala H, Pan E, Mestdagh P, Muth D, Teruya- Feldstein J, Reinhardt F, Onder TT, Valastyan S, 2010, miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nat Cell Biol 12(3):247–256 Brabletz S, Brabletz T, 2010, The ZEB/miR-200 feedback loop— a motor of cellular plasticity in development and cancer? EMBO Rep 11(9):670–677 Hill L, Browne G, Tulchinsky E, 2013, ZEB/miR-200 feedback loop: At the crossroads of signal transduction in cancer. Int J Cancer 132(4):745–754 Liu Y, Yin J, Abou-Kheir W, Hynes P, Casey O, Fang L, Yi M, Stephens R, Seng V, Sheppard-Tillman H, 2013)MiR-1 and miR- 200 inhibit EMT via Slug-dependent and tumorigenesis via Slugindependent mechanisms. Oncogene 32(3):296–306 Gregory PA, Bracken CP, Bert AG, Goodall GJ, 2008, MicroRNAs as regulators of epithelial-mesenchymal transition. Cell Cycle 7(20):3112–3117 Siemens H, Jackstadt R, Hunten S, Kaller M, Menssen A, Gotz U, Hermeking H, 2011, miR-34 and SNAIL form a double-negative feedback loop to regulate epithelial-mesenchymal transitions. Cell Cycle 10(24):4256–4271 Hudson RS, Yi M, Esposito D,Watkins SK, Hurwitz AA, Yfantis HG, Lee DH, Borin JF, Naslund MJ, Alexander RB, 2011, MicroRNA-1 is a candidate tumor suppressor and prognostic marker in human prostate cancer. Nucleic acids research: gkr1222 Datta J, Kutay H, Nasser MW, Nuovo GJ, Wang B, Majumder S, Liu C-G, Volinia S, Croce CM, Schmittgen TD, 2008, Methylation mediated silencing of MicroRNA-1 gene and its role in hepatocellular carcinogenesis. Cancer Res 68(13):5049–5058 Nohata N, Sone Y, Hanazawa T, FuseM, Kikkawa N, Yoshino H, Chiyomaru T, Kawakami K, Enokida H, Nakagawa M, 2011, miR-1 as a tumor suppressive microRNA targeting TAGLN2 in head and neck squamous cell carcinoma. Oncotarget 2, 1–2):29 Suzuki H, Takatsuka S, Akashi H, Yamamoto E, Nojima M, Maruyama R, Kai M, H-o Y, Sasaki Y, Tokino T, 2011, Genome-wide profiling of chromatin signatures reveals epigenetic regulation of MicroRNA genes in colorectal cancer. Cancer Res 71(17):5646–5658 Gumbiner BM, 2005, Regulation of cadherin-mediated adhesion in morphogenesis. Nat Rev Mol Cell Biol 6(8):622–634 Chen D, Sun Y,WeiY, Zhang P, Rezaeian AH, Teruya-Feldstein J, Gupta S, Liang H, Lin H-K, Hung M-C, 2012, LIFR is a breast cancermetastasis suppressor upstream of the Hippo-YAP pathway and a prognostic marker. Nat Med 18(10):1511–1517 Mott JL, Kobayashi S, Bronk SF, Gores GJ, 2007, Mir-29 regulates mcl-1 protein expression and apoptosis. Oncogene 26(42): 6133–6140 Garzon R, Heaphy CE, Havelange V, FabbriM, Volinia S, Tsao T, Zanesi N, Kornblau SM, Marcucci G, Calin GA, 2009, MicroRNA 29b functions in acute myeloid leukemia. Blood 114(26):5331–5341 Fang JH, Zhou HC, Zeng C, Yang J, Liu Y, Huang X, Zhang JP, Guan XY, Zhuang SM, 2011, MicroRNA-29b suppresses tumor angiogenesis, invasion, and metastasis by regulating matrix metalloproteinase 2 expression. Hepatology 54(5):1729–1740 Zhang Y-K,Wang H, Leng Y, Li Z-L, Yang Y-F, Xiao F-J, Li Q-F, Chen X-Q, Wang L-S, 2011, Overexpression of microRNA-29b induces apoptosis of multiple myeloma cells through down regulating mcl-1. Biochem Biophys Res Commun 414(1):233–239 Chou J, Lin JH, Brenot A, J-w K, Provot S, Werb Z, 2013, GATA3 suppresses metastasis and modulates the tumour microenvironment by regulating microRNA-29b expression. Nat Cell Biol 15(2):201–213 Ru P, Steele R, Newhall P, Phillips NJ, Toth K, Ray RB, 2012, miRNA-29b suppresses prostate cancer metastasis by regulating epithelial–mesenchymal transition signaling. Mol Cancer Ther 11(5):1166–1173 Huang Z, Huang D, Ni S, Peng Z, ShengW, Du X, 2010, Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. Int J Cancer 127(1):118–126 Wu Q, Lu Z, Li H, Lu J, Guo L, Ge Q, 2011, Next-generation sequencing of microRNAs for breast cancer detection. Biomed Res Int 2011 Gebeshuber CA, Zatloukal K, Martinez J, 2009, miR-29a suppresses tristetraprolin, which is a regulator of epithelial polarity and metastasis. EMBO Rep 10(4):400–405 Baffa R, Fassan M, Volinia S, O’Hara B, Liu CG, Palazzo JP, Gardiman M, Rugge M, Gomella LG, Croce CM, 2009, MicroRNA expression profiling of human metastatic cancers identifies cancer gene targets. J Pathol 219(2):214–221 Kumarswamy R, Mudduluru G, Ceppi P, Muppala S, Kozlowski M, Niklinski J, Papotti M, Allgayer H, 2012, MicroRNA-30a inhibits epithelial-to-mesenchymal transition by targeting Snai1 and is downregulated in non-small cell lung cancer. Int J Cancer 130(9):2044–2053 Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M, Stephens RM, Okamoto A, Yokota J, Tanaka T, 2006, Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell 9(3):189–198 Bandres E, Cubedo E, Agirre X,Malumbres R, Zarate R, Ramirez N, Abajo A, Navarro A, Moreno I, MonzoM(2006, Identification by Real-time PCR of 13 mature microRNAs differentially expressed in colorectal cancer and non-tumoral tissues. Mol Cancer 5(1):29 Budhu A, Jia HL, Forgues M, Liu CG, Goldstein D, Lam A, Zanetti KA, Ye QH, Qin LX, Croce CM, 2008, Identification of metastasis-related microRNAs in hepatocellular carcinoma. Hepatology 47(3):897–907 Porkka KP, PfeifferMJ,Waltering KK, Vessella RL, Tammela TL, Visakorpi T, 2007, MicroRNA expression profiling in prostate cancer. Cancer Res 67(13):6130–6135 Yan L-X, Huang X-F, Shao Q, Huang M-Y, Deng L, Wu Q-L, Zeng Y-X, Shao J-Y, 2008)MicroRNAmiR-21 overexpression in human breast cancer is associated with advanced clinical stage, lymph node metastasis and patient poor prognosis. RNA 14(11): 2348–2360 Kao C, Martiniez A, Shi X, Yang J, Evans C, Dobi A, Devere White R, Kung H, 2014, miR-30 as a tumor suppressor connects EGF/Src signal to ERG and EMT. Oncogene 33(19):2495–2503 Yao J, Liang L, Huang S, Ding J, Tan N, Zhao Y, Yan M, Ge C, Zhang Z, Chen T, 2010, MicroRNA-30d promotes tumor invasion and metastasis by targeting Galphai2 in hepatocellular carcinoma. Hepatology 51(3):846–856 Braun J, Hoang-Vu C, Dralle H, Huttelmaier S, 2010, Downregulation of microRNAs directs the EMT and invasive potential of anaplastic thyroid carcinomas. Oncogene 29(29): 4237–4244 Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, Patrawala L, Yan H, Jeter C, Honorio S, 2011, The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med 17(2):211–215 Kim NH, Kim HS, Li X-Y, Lee I, Choi H-S, Kang SE, Cha SY, Ryu JK, Yoon D, Fearon ER, 2011, A p53/miRNA-34 axis regulates Snail1-dependent cancer cell epithelial–mesenchymal transition. J Cell Biol 195(3):417–433 Martello G, Rosato A, Ferrari F, Manfrin A, Cordenonsi M, Dupont S, Enzo E, Guzzardo V, Rondina M, Spruce T, 2010, A MicroRNA targeting dicer for metastasis control. Cell 141(7): 1195–1207 Ma L, Teruya-Feldstein J, Weinberg RA, 2007, Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature 449(7163):682–688 Georges SA, Biery MC, S-y K, Schelter JM, Guo J, Chang AN, Jackson AL, Carleton MO, Linsley PS, Cleary MA, 2008, Coordinated regulation of cell cycle transcripts by p53-Inducible microRNAs, miR-192 and miR-215. Cancer Res 68(24):10105– 10112 Pichiorri F, Suh S-S, Rocci A, De Luca L, Taccioli C, Santhanam R, Zhou W, Benson DM, Hofmainster C, Alder H, 2010, Downregulation of p53-inducible microRNAs 192, 194, and 215 impairs the p53/MDM2 autoregulatory loop in multiple myeloma development. Cancer Cell 18(4):367–381 Braun CJ, Zhang X, Savelyeva I,Wolff S, Moll UM, Schepeler T, Orntoft TF, Andersen CL, DobbelsteinM, 2008, p53-Responsive micrornas 192 and 215 are capable of inducing cell cycle arrest. Cancer Res 68(24):10094–10104 Zhang Q, He X, Ma L, Li N, Yang J, Cheng Y, Cui H, 2011, Expression and significance of microRNAs in the p53 pathway in ovarian cancer cells and serous ovarian cancer tissues. Zhonghua zhong liu za zhi [Chinese journal of oncology] 33(12):885–890 Lewis BC, Klimstra DS, Socci ND, Xu S, Koutcher JA, Varmus HE, 2005, The absence of p53 promotes metastasis in a novel somatic mouse model for hepatocellular carcinoma. Mol Cell Biol 25(4):1228–1237 Ridgeway A,McMenamin J, Leder P, 2006, P53 levels determine outcome during β-catenin tumor initiation and metastasis in the mammary gland and male germ cells. Oncogene 25(25):3518– 3527 Chen Y-W, Klimstra DS, Mongeau ME, Tatem JL, Boyartchuk V, Lewis BC, 2007, Loss of p53 and Ink4a/Arf cooperate in a cell autonomous fashion to induce metastasis of hepatocellular carcinoma cells. Cancer Res 67(16):7589–7596 Hansen JE, Fischer LK, Chan G, Chang SS, Baldwin SW, Aragon RJ, Carter JJ, Lilly M, Nishimura RN, Weisbart RH, 2007, Antibody-mediated p53 protein therapy prevents liver metastasis in vivo. Cancer Res 67(4):1769–1774 Yang J, Weinberg RA, 2008, Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell 14(6):818–829 Kim T, Veronese A, Pichiorri F, Lee TJ, Jeon Y-J, Volinia S, Pineau P, Marchio A, Palatini J, Suh S-S, 2011, p53 regulates epithelial–mesenchymal transition through microRNAs targeting ZEB1 and ZEB2. J Exp Med 208(5):875–883 Park S-M, Gaur AB, Lengyel E, Peter ME, 2008, The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev 22(7):894–907 Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ, 2008, The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol 10(5):593–601 Diaz-Martin J, Diaz-Lopez A, Moreno-Bueno G, Castilla M, Rosa-Rosa JM, Cano A, Palacios J, 2014, A core microRNA signature associated with inducers of the epithelial-tomesenchymal transition. J Pathol 232(3):319–329 Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, Brabletz T, 2008, A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep 9(6):582–589 Korpal M, Ell BJ, Buffa FM, Ibrahim T, Blanco MA, Celia- Terrassa T, Mercatali L, Khan Z, Goodarzi H, Hua Y, 2011, Direct targeting of Sec23a by miR-200 s influences cancer cell secretome and promotes metastatic colonization. Nat Med 17(9): 1101–1108 Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A, 2008, Circulating microRNAs as stable bloodbased markers for cancer detection. Proc Natl Acad Sci 105(30): 10513–10518 Korpal M, Lee ES, Hu G, Kang Y, 2008, The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. J Biol Chem 283(22):14910–14914 Neves R, Scheel C, Weinhold S, Honisch E, Iwaniuk KM, Trompeter H-I, Niederacher D, Wernet P, Santourlidis S, Uhrberg M, 2010, Role of DNA methylation in miR-200c/141 cluster silencing in invasive breast cancer cells. BMC research notes 3(1):219 Castilla MA, Diaz-Martin J, Sarrio D, Romero-Perez L, Lopez- GarciaMA,Vieites B, Biscuola M, Ramiro-Fuentes S, IsackeCM, Palacios J, 2012, MicroRNA-200 family modulation in distinct breast cancer phenotypes. PLoS One 7(10):e47709 Cheng H, Zhang L, Cogdell DE, Zheng H, Schetter AJ, Nykter M, Harris CC, Chen K, Hamilton SR, Zhang W, 2011, Circulating plasma MiR-141 is a novel biomarker for metastatic colon cancer and predicts poor prognosis. PLoS One 6(3):e17745 Kimura S, Naganuma S, Susuki D, Hirono Y, Yamaguchi A, Fujieda S, Sano K, Itoh H, 2010, Expression of microRNAs in squamous cell carcinoma of human head and neck and the esophagus: miR-205 and miR-21 are specific markers for HNSCC and ESCC. Oncol Rep 23(6):1625–1633 Tran N, McLean T, Zhang X, Zhao CJ, Thomson JM, O’Brien C, Rose B, 2007, MicroRNA expression profiles in head and neck cancer cell lines. Biochem Biophys Res Commun 358(1):12–17 Wu H, Zhu S, Mo Y-Y, 2009, Suppression of cell growth and invasion by miR-205 in breast cancer. Cell Res 19(4):439–448 Gandellini P, Folini M, Longoni N, Pennati M, Binda M, Colecchia M, Salvioni R, Supino R, Moretti R, Limonta P, 2009, miR-205 exerts tumor-suppressive functions in human prostate through down-regulation of protein kinase Cε. Cancer Res 69(6):2287–2295 Matsushima K, Isomoto H, Yamaguchi N, Inoue N, Machida H, Nakayama T, Hayashi T, Kunizaki M, Hidaka S, Nagayasu T, 2011, MiRNA-205 modulates cellular invasion and migration via regulating zinc finger E-box binding homeobox 2 expression in esophageal squamous cell carcinoma cells. BioMed Central Limited Saini S,Majid S, Yamamura S, Tabatabai L, Suh SO, Shahryari V, Chen Y, Deng G, Tanaka Y, Dahiya R, 2011, Regulatory role of miR-203 in prostate cancer progression and metastasis. Clin Cancer Res 17(16):5287–5298 Zhang Z, Zhang B, Li W, Fu L, Fu L, Zhu Z, Dong J-T, 2011, Epigenetic silencing of miR-203 upregulates SNAI2 and contributes to the invasiveness of malignant breast cancer cells. Genes & cancer 2(8):782–791 Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, Rougvie AE, Horvitz HR, Ruvkun G, 2000, The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 403(6772):901–906., DOI 10.1038/35002607 Esquela-Kerscher A, Johnson SM, Bai L, Saito K, Partridge J, Reinert KL, Slack FJ, 2005, Post-embryonic expression of C. elegans microRNAs belonging to the lin-4 and let-7 families in the hypodermis and the reproductive system. Dev Dyn 234(4): 868–877., DOI 10.1002/dvdy.20572 Lee H, Han S, Kwon CS, Lee D, 2016, Biogenesis and regulation of the let-7 miRNAs and their functional implications. Protein Cell 7(2):100–113., DOI 10.1007/s13238-015-0212-y Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, Labourier E, Reinert KL, Brown D, Slack FJ, 2005, RAS is regulated by the let-7 microRNA family. Cell 120(5):635–647., DOI 10.1016/j.cell.2005.01.014 Bhat-Nakshatri P,Wang G, Collins NR, Thomson MJ, Geistlinger TR, Carroll JS, Brown M, Hammond S, Srour EF, Liu Y, Nakshatri H, 2009, Estradiol-regulated microRNAs control estradiol response in breast cancer cells. Nucleic Acids Res 37(14): 4850–4861., DOI 10.1093/nar/gkp500 Yu F, Yao H, Zhu P, Zhang X, Pan Q, Gong C, Huang Y, Hu X, Su F, Lieberman J, 2007, let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell 131(6):1109–1123 Romero-Perez L, Castilla MA, Lopez-Garcia MA, Diaz-Martin J, Biscuola M, Ramiro-Fuentes S, Oliva E, Matias-Guiu X, Prat J, Cano A, 2013, Molecular events in endometrial carcinosarcomas and the role of high mobility group AT-hook 2 in endometrial carcinogenesis. Hum Pathol 44(2):244–254 Yun J, Frankenberger CA, Kuo WL, Boelens MC, Eves EM, Cheng N, Liang H, Li WH, Ishwaran H, Minn AJ, 2011, Signalling pathway for RKIP and let-7 regulates and predicts metastatic breast cancer. EMBO J 30(21):4500–4514 Heo I, Joo C, Cho J, Ha M, Han J, Kim VN, 2008, Lin28mediates the terminal uridylation of let-7 precursor MicroRNA. Mol Cell 32(2):276–284 King CE, Cuatrecasas M, Castells A, Sepulveda AR, Lee J-S, Rustgi AK, 2011, LIN28B promotes colon cancer progression and metastasis. Cancer Res 71(12):4260–4268 Miyazawa J, Mitoro A, Kawashiri S, Chada KK, Imai K, 2004, Expression of mesenchyme-specific gene HMGA2 in squamous cell carcinomas of the oral cavity. Cancer Res 64(6):2024–2029 Motoyama K, Inoue H, Nakamura Y, Uetake H, Sugihara K,Mori M, 2008, Clinical significance of high mobility group A2 in human gastric cancer and its relationship to let-7 microRNA family. Clin Cancer Res 14(8):2334–2340 Di Cello F, Hillion J, Hristov A, Wood LJ, Mukherjee M, Schuldenfrei A, Kowalski J, Bhattacharya R, Ashfaq R, Resar LM(2008)HMGA2 participates in transformation in human lung cancer. Mol Cancer Res 6(5):743–750 Wang Y-C, Chen Y-L, Yuan R-H, Pan H-W, YangW-C, Hsu H-C, Jeng Y-M, 2010, Lin-28B expression promotes transformation and invasion in human hepatocellular carcinoma. Carcinogenesis 31(9):1516–1522 Helland A, Anglesio MS, George J, Cowin PA, Johnstone CN, House CM, Sheppard KE, Etemadmoghadam D, Melnyk N, Rustgi AK, 2011, Deregulation of MYCN, LIN28B and LET7 in a molecular subtype of aggressive high-grade serous ovarian cancers. PLoS One 6(4):e18064 Fu Z, KitagawaY, Shen R, Shah R, Mehra R, Rhodes D,Keller PJ, Mizokami A, Dunn R, Chinnaiyan AM, 2006, Metastasis suppressor gene Raf kinase inhibitor protein, RKIP, is a novel prognostic marker in prostate cancer. Prostate 66(3):248–256 Schuierer MM, Bataille F, Hagan S, Kolch W, Bosserhoff A-K, 2004, Reduction in Raf kinase inhibitor protein expression is associated with increased Ras-extracellular signal-regulated kinase signaling in melanoma cell lines. Cancer Res 64(15):5186– 5192 Hagan S, Al-Mulla F, Mallon E, Oien K, Ferrier R, Gusterson B, Garcia JJC, Kolch W, 2005, Reduction of Raf-1 kinase inhibitor protein expression correlates with breast cancer metastasis. Clin Cancer Res 11(20):7392–7397 Akaishi J, OndaM, Asaka S, Okamoto J, Miyamoto S, Nagahama M, Ito K, Kawanami O, Shimizu K, 2006, Growth-suppressive function of phosphatidylethanolamine-binding protein in anaplastic thyroid cancer. Anticancer Res 26(6B):4437–4442 Lu M-H, Huang C-C, Pan M-R, Chen H-H, Hung W-C, 2012, Prospero homeobox 1 promotes epithelial–mesenchymal transition in colon cancer cells by inhibiting E-cadherin via miR-9. Clin Cancer Res 18(23):6416–6425 Hu M, Xia M, Chen X, Lin Z, Xu Y, Ma Y, Su L, 2010, MicroRNA-141 regulates Smad interacting protein 1, SIP1, and inhibits migration and invasion of colorectal cancer cells. Dig Dis Sci 55(8):2365–2372 Q-j L, Zhou L, Yang F,Wang G-x, Zheng H,Wang D-s, He Y, K-f D, 2012, MicroRNA-10b promotes migration and invasion through CADM1 in human hepatocellular carcinoma cells. Tumor Biol 33(5):1455–1465 Tian Y, Luo A, Cai Y, Su Q, Ding F, Chen H, Liu Z, 2010, MicroRNA-10b promotes migration and invasion through KLF4 in human esophageal cancer cell lines. J Biol Chem 285(11): 7986–7994 Weiss FU, Marques IJ, Woltering JM, Vlecken DH, Aghdassi A, Partecke LI, Heidecke CD, Lerch MM, Bagowski CP, 2009, Retinoic acid receptor antagonists inhibit miR-10a expression and block metastatic behavior of pancreatic cancer. Gastroenterology 137(6):2136–2145. e2137 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 1 EP 12 DI 10.1007/s12253-016-0101-6 PG 12 ER PT J AU Tsai, ShH Liu, ChA Huang, KH Lan, YT Chen, MH Chao, Y Lo, SSh Li, FYA Wu, ChW Chiou, ShH Yang, MH Shyr, YM Fang, WL AF Tsai, Sheng-Han Liu, Chien-An Huang, Kuo-Hung Lan, Yuan-Tzu Chen, Ming-Huang Chao, Yee Lo, Su-Shun Li, Fen-Yau Anna Wu, Chew-Wun Chiou, Shih-Hwa Yang, Muh-Hwa Shyr, Yi-Ming Fang, Wen-Liang TI Advances in Laparoscopic and Robotic Gastrectomy for Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Laparoscopic; Robotic; Lymph node dissection; Gastric cancer; Elderly ID Laparoscopic; Robotic; Lymph node dissection; Gastric cancer; Elderly AB Robot-assisted gastrectomy has been reported to be a safe alternative to both conventional laparoscopy and the open approach for treating early gastric carcinoma. Currently, there are a limited number of published reports on this technique in the literature. We assessed the current status of robotic and laparoscopic surgery in the treatment of gastric cancer and compared the operative outcomes, learning curves, and oncological outcome of the two approaches. Robotic gastrectomy offers benefits that include increased ease of performing D2 lymph node dissection and reduced blood loss compared with laparoscopic gastrectomy. However, the operative time is longer, and robotic gastrectomy is more costly for the patients. Regarding to the operative and oncological outcomes, there appears to be no significant differences between laparoscopic and robotic gastrectomies after the surgeon overcomes the associated learning curves. Sharing the available knowledge regarding laparoscopic and robotic gastrectomies could shorten these learning curves. For elder patients,minimally invasive surgery that decreases the postoperative recovery time should be considered the preferred treatment. Prospective randomized studies are required to compare the surgical and oncological outcomes among laparoscopic, robotic, and open surgeries for both early and advanced gastric cancer. C1 [Tsai, Sheng-Han] Cheng Hsin General Hospital, Department of UrologyTaipei, Taiwan, Republic of China. [Liu, Chien-An] Taipei Veterans General Hospital, Department of RadiologyTaipei, Taiwan, Republic of China. [Huang, Kuo-Hung] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd, Beitou District, 11217 Taipei, Taiwan, Republic of China. [Lan, Yuan-Tzu] National Yang-Ming University, School of MedicineTaipei, Taiwan, Republic of China. [Chen, Ming-Huang] National Yang-Ming University, School of MedicineTaipei, Taiwan, Republic of China. [Chao, Yee] National Yang-Ming University, School of MedicineTaipei, Taiwan, Republic of China. [Lo, Su-Shun] National Yang-Ming University, School of MedicineTaipei, Taiwan, Republic of China. [Li, Fen-Yau Anna] National Yang-Ming University, School of MedicineTaipei, Taiwan, Republic of China. [Wu, Chew-Wun] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd, Beitou District, 11217 Taipei, Taiwan, Republic of China. [Chiou, Shih-Hwa] National Yang-Ming University, School of Medicine, Institute of Clinical MedicineTaipei, Taiwan, Republic of China. [Yang, Muh-Hwa] National Yang-Ming University, School of Medicine, Institute of Clinical MedicineTaipei, Taiwan, Republic of China. [Shyr, Yi-Ming] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd, Beitou District, 11217 Taipei, Taiwan, Republic of China. [Fang, Wen-Liang] Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, No. 201, Sec. 2, Shipai Rd, Beitou District, 11217 Taipei, Taiwan, Republic of China. RP Fang, WL (reprint author), Taipei Veterans General Hospital, Department of Surgery, Division of General Surgery, 11217 Taipei, Taiwan, Republic of China. EM s821094@hotmail.com CR Giulianotti PC, Coratti A, Angelini M, Sbrana F, Cecconi S, Balestracci T, Caravaglios G, 2003, Robotics in general surgery: personal experience in a large community hospital. Arch Surg 138: 777–784 Giulianotti PC, Angelini M, Coratti A et al, 2002,, 2002, Technique de la gastrectomie subtotale robotique. J Coelio- Chirurgie 43:55–60 Hashizume M, 2003, Sugimachi K, 2003, robot-assisted gastric surgery. Surg Clin North Am 83:1429–1444 Song J, Oh SJ, Kang WH, Hyung WJ, Choi SH, Noh SH, 2009, Robot-assisted gastrectomy with lymph node dissection for gastric cancer: lessons learned from an initial 100 consecutive procedures. Ann Surg 249:927–932 Woo Y, Hyung WJ, Pak KH, Inaba K, Obama K, Choi SH, Noh SH, 2011, Robotic gastrectomy as an oncologically sound alternative to laparoscopic resections for the treatment of early-stage gastric cancers. Arch Surg 146:1086–1092 Song J, Kang WH, Oh SJ, Hyung WJ, Choi SH, Noh SH, 2009, Role of robotic gastrectomy using da Vinci system compared with laparoscopic gastrectomy: initial experience of 20 consecutive cases. Surg Endosc 23:1204–1211 Heemskerk J, van Gemert WG, de Vries J, Greve J, Bouvy ND, 2007, Learning curve of robot-assisted laparoscopic surgery compared with conventional laparoscopic surgery: an experimental study evaluating skill acquisition of robot-assisted laparoscopic tasks compared with conventional laparoscopic tasks in inexperienced users. Surg Laparosc Endosc Percutan Tech 17:171–174 Park SS, KimMC, ParkMS, HyungWJ, 2012, Rapid adaptation of robotic gastrectomy for gastric cancer by experienced laparoscopic surgeons. Surg Endosc 26:60–67 Kim HI, Park MS, Song KJ, Woo Y, Hyung WJ, 2014, Rapid and safe learning of robotic gastrectomy for gastric cancer: multidimensional analysis in a comparison with laparoscopic gastrectomy. Eur J Surg Oncol 40:1346–1354 Huang KH, Lan YT, FangWL, Chen JH, Lo SS, Li AF, Chiou SH, Wu CW, Shyr YM, 2014, Comparison of the operative outcomes and learning curves between laparoscopic and robotic gastrectomy for gastric cancer. PLoS One 9:e111499 Huscher CG, Mingoli A, Sgarzini G, Sansonetti A, Di Paola M, Recher A, Ponzano C, 2005, Laparoscopic versus open subtotal gastrectomy for distal gastric cancer: five year results of a prospective randomized trial. Ann Surg 241:232–237 Hur H, Jeon HM, Kim W, 2008, Laparoscopy-assisted distal gastrectomy with D2 lymphadenectomy for T2b advanced gastric cancers: three years experience. J Surg Oncol 98:515–519 Lee SW, Nomura E, Bouras G, Tokuhara T, Tsunemi S, Tanigawa N, 2012, Long-term oncologic outcomes from laparoscopic gastrectomy for gastric cancer: a single-center experience of 601 consecutive resections. J Am Coll Surg 211:33–40 Hwang SI, Kim HO, Yoo CH, Shin JH, Son BH, 2009, Laparoscopic-assisted distal gastrectomy versus open distal gastrectomy for advanced gastric cancer. Surg Endosc 23:1252–1258 Lee J, Kim W, 2009, Long-term outcomes after laparoscopic assisted gastrectomy for advanced gastric cancer: analysis of consecutive 106 experiences. J Surg Oncol 100:693–698 Son T, Kwon IG, Hyung WJ, 2014)Minimally invasive surgery for gastric cancer treatment: current status and future perspectives. Gut Liver 8:229–236 Wu CW, Hsiung CA, Lo SS, Hsieh MC, Chen JH, Li AF, Lui WY, Whang-Peng J, 2006, Nodal dissection for patients with gastric cancer: a randomised controlled trial. Lancet Oncol 7:309–315 Jin SH, Kim DY, Kim H, Jeong IH, Kim MW, Cho YK, Han SU, 2007, Multidimensional learning curve in laparoscopy-assisted gastrectomy for early gastric cancer. Surg Endosc 21:28–33 Memon MA, Butler N, Memon B, 2010, The issue of lymphadenectomy during laparoscopic gastrectomy for gastric carcinoma. World J Gastrointest Oncol 2:65–67 D'Annibale A, Pende V, Pernazza G, Monsellato I, Mazzocchi P, Lucandri G, Morpurgo E, Contardo T, Sovernigo G, 2011, Full robotic gastrectomy with extended, D2, lymphadenectomy for gastric cancer: surgical technique and preliminary results. J Surg Res 166:113–120 Kim MC, Heo GU, Jung GJ, 2010, Robotic gastrectomy for gastric cancer: surgical techniques and clinical merits. Surg Endosc 24: 610–615 The Japanese Gastric Cancer Association, 2011, Japanese classification of gastric carcinoma: 3rd English edition. Gastric Cancer 14: 101–112 Kim H, Lee SK, Kim YM, Lee EH, Lim SJ, Kim SH, Yang J, Lim JS, Hyung WJ, 2015, Fluorescent iodized emulsion for pre- and intraoperative sentinel lymph node imaging: validation in a preclinical model. Radiology 275:196–204 Lee J, Kim YM, Woo Y, Obama K, Noh SH, Hyung WJ, 2015, Robotic distal subtotal gastrectomy with D2 lymphadenectomy for gastric cancer patients with high bodymass index: comparison with conventional laparoscopic distal subtotal gastrectomy with D2 lymphadenectomy. Surg Endosc 29:3251–3260 Lee HJ, Kim HH, Kim MC, Ryu SY, Kim W, Song KY, Cho GS, Han SU, Hyung WJ, Ryu SW; Korean Laparoscopic Gastrointestinal Surgery Study Group, 2009, The impact of a high body mass index on laparoscopy-assisted gastrectomy for gastric cancer. Surg Endosc 23:2473–2479 Lee JH, Paik YH, Lee JS, Ryu KW, Kim CG, Park SR, Kim YW, Kook MC, Nam BH, Bae JM, 2007, Abdominal shape of gastric cancer patients influences short-term surgical outcomes. Ann Surg Oncol 14:1288–1294 Noshiro H, Shimizu S, Nagai E, Ohuchida K, Tanaka M, 2003, Laparoscopy-assisted distal gastrectomy for early gastric cancer: is it beneficial for patients of heavier weight? Ann Surg 238: 680–685 Yasuda K, Inomata M, Shiraishi N, Izumi K, Ishikawa K, Kitano S, 2004, Laparoscopy assisted distal gastrectomy for early gastric cancer in obese and nonobese patients. Surg Endosc 18:1253–1256 Kim KH, KimMC, JungGJ, KimHH, 2006, The impact of obesity on LADG for early gastric cancer. Gastric Cancer 9:303–307 Park DJ, Lee HJ, Kim HH, Yang HK, Lee KU, Choe KJ, 2005, Predictors of operative morbidity and mortality in gastric cancer surgery. Br J Surg 92:1099–1102 Huang KH, Lan YT, FangWL, Chen JH, Lo SS, Hsieh MC, Li AF, Chiou SH,Wu CW(2012, Initial experience of robotic gastrectomy and comparison with open and laparoscopic gastrectomy for gastric cancer. J Gastrointest Surg 16:1303–1310 Hyun MH, Lee CH, Kim HJ, Tong Y, Park SS, 2013, Systematic review and meta-analysis of robotic surgery compared with conventional laparoscopic and open resections for gastric carcinoma. Br J Surg 100:1566–1578 Xiong B, Ma L, Zhang C, 2012, Robotic versus laparoscopic gastrectomy for gastric cancer: a meta-analysis of short outcomes. Surg Oncol 21:274–280 PatritiA, CeccarelliG, Bellochi R, Bartoli A, Spaziani A, Di Zitti L, Casciola L, 2008, Robot-assisted laparoscopic total and partial gastric resection with D2 lymph node dissection for adenocarcinoma. Surg Endosc 22:2753–2760 Pugliese R, Maggioni D, Sansonna F, Costanzi A, Ferrari GC, Di Lernia S, Magistro C, De Martini P, Pugliese F, 2010, Subtotal gastrectomy with D2 dissection by minimally invasive surgery for distal adenocarcinoma of the stomach: results and 5-year survival. Surg Endosc 24:2594–2602 Yu PW, Tang B, Zeng DZ, Zhao YL, Shi Y, Hao YX, Qian F, 2012, Robotic-assisted radical gastrectomy using da Vinci robotic system: a report of 41 cases. Zhonghua Wei Chang Wai Ke Za Zhi 15:121–124 Zhou JF, Yan S, Bo T, Hao Y, Zeng DZ, Zhao YL, Qian F, Yu PW, 2014, Robotic gastrectomy versus laparoscopic gastrectomy for gastric cancer: comparison of surgical performance and short-term outcomes. Surg Endosc 28:1779–1787 Kwon IG, Cho I, Guner A, Kim HI, Noh SH, Hyung WJ, 2015, Minimally invasive surgery as a treatment option for gastric cancer in the elderly: comparison with open surgery for patients 80 years and older. Surg Endosc 29:2321–2330 Lo SS,Wu CW, Chen JH, Li AF, HsiehMC, Shen KH, Lin HJ, Lui WY, 2007, Surgical results of early gastric cancer and proposing a treatment strategy. Ann Surg Oncol 14:340–347 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 13 EP 17 DI 10.1007/s12253-016-0131-0 PG 5 ER PT J AU Hosseinpour, S Mashhadiabbas, F Ahsaie, GM AF Hosseinpour, Sepanta Mashhadiabbas, Fatemeh Ahsaie, Ghazizadeh Mitra TI Diagnostic Biomarkers in Oral Verrucous Carcinoma: A Systematic Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Biomarker; Verrucous carcinoma; Oral carcinoma ID Biomarker; Verrucous carcinoma; Oral carcinoma AB Oral verrucous carcinoma (OVC), a low-grade variant of oral squamous cell carcinoma (OSCC), is most frequently seen in the oral cavity. No clear etiology has been found for this lesion, but human papilloma virus, chewing betel nuts, and ultraviolet radiation are suggested as probable causes. Differential diagnosis of OVC is challenging for oral pathologists. The aim of this study was to review the molecular-based approaches for differential diagnosis of OVC. An electronic search was conducted in Medline and Scopus from January 2004 to July 2015 limited to English language publications. Published papers on verrucous carcinoma (VC) were found according to the inclusion and exclusion criteria and analyzed qualitatively. Data extraction were performed according to PRISMA statement. A total of 423 articles were reviewed; out of which, 26 articles completely fulfilled the inclusion criteria. Most of the included studies investigated proliferative and apoptotic biomarkers such as p53 and Ki67. No definite conclusion was drawn for cytoskeletal biomarkers due to variability of factors and lack of significant expression. However, it seems that cytokeratin10 (CK 10) can be useful for differentiation of OVC and benign squamous lesions. Among cell surface and extracellular matrix biomarkers tissue biomarkers, matrix metalloproteinase (MMP)-2, −9, CD31 and CD68 seem to be useful for differentiation of OVC and OSCC and glucose transporter-1 (GLUT-1) can help in differentiation of OVC from oral epithelial dysplasia. Differences among OVC, OSCC and normal epithelium in expression profiles of the investigated biomarkers help in their differential diagnosis; although, clinicohistopathological similarities among verrucous hyperplasia, noninvasive OVC and invasive well-differentiated OSCC make the diagnosis difficult. Further studies are required to better differentiate these oral lesions. C1 [Hosseinpour, Sepanta] Shahid Beheshti University of Medical Sciences, School of Dentistry, Students’ Research OfficeTehran, Iran. [Mashhadiabbas, Fatemeh] Shahid Beheshti University of Medical Sciences, Dental School, Department of Oral and Maxillofacial Pathology, Daneshjou Boulevard, Evin, 19839 Tehran, Iran. [Ahsaie, Ghazizadeh Mitra] Shahid Beheshti University of Medical Sciences, School of Dentistry, Students’ Research OfficeTehran, Iran. RP Mashhadiabbas, F (reprint author), Shahid Beheshti University of Medical Sciences, Dental School, Department of Oral and Maxillofacial Pathology, 19839 Tehran, Iran. EM Fmashhadiabbas@yahoo.com CR Stransky N, Egloff AM, Tward AD, Kostic AD, Cibulskis K, Sivachenko A, Kryukov GV, Lawrence MS, Sougnez C, McKenna A, 2011, The mutational landscape of head and neck squamous cell carcinoma. Science 333(6046):1157–1160 Ackerman LV, 1948, Verrucous carcinoma of the oral cavity. Surgery 23(4):670–678 Eversole L, 2000, Papillary lesions of the oral cavity: relationship to human papillomaviruses. J Calif Dent Assoc 28(12):922–927 Sharan RN, Mehrotra R, Choudhury Y, Asotra K, 2012, Association of betel nut with carcinogenesis: revisit with a clinical perspective. PLoS One 7(8):e42759 Johnson N, 2001, Tobacco use and oral cancer: a global perspective. J Dent Educ 65(4):328–339 Schmitt J, Seidler A, Diepgen T, Bauer A, 2011, Occupational ultraviolet light exposure increases the risk for the development of cutaneous squamous cell carcinoma: a systematic review and metaanalysis. Br J Dermatol 164(2):291–307 Jacobson S, Shear M, 1972, Verrucous carcinoma of the mouth. J Oral Pathol Med 1(2):66–75 Garcia NG, Oliveira DT, Hanemann JAC, PereiraAAC, 2012, Oral verrucous carcinoma mimicking a chronic candidiasis: a case report. Case reports in oncological medicine 2012 Koch BB, Trask DK, Hoffman HT, Karnell LH, Robinson RA, Zhen W, Menck HR, 2001, National survey of head and neck verrucous carcinoma. Cancer 92(1):110–120 Yeh C-J, 2003, Treatment of verrucous hyperplasia and verrucous carcinoma by shave excision and simple cryosurgery. Int J Oral Maxillofac Surg 32(3):280–283 Schrader M, Laberke H, Jahnke K, 1987, Lymphatic metastases of verrucous carcinoma, Ackerman tumor). HNO 35(1):27–30 Jiang L, Wang S, Chen X, 2001, Immunohistochemical and ultrastructural study of basement membrane in oral verrucous carcinoma. Zhonghua kou qiang yi xue za zhi = Zhonghua kouqiang yixue zazhi=. Chinese J Stomatol 36(4):308 Barnes L, 2005, Pathology and genetics of head and neck tumours, 9. IARC, Shear M, Pindborg J, 1980, Verrucous hyperplasia of the oral mucosa. Cancer 46(8):1855–1862 Neville BW, 2009, Oral and maxillofacial pathology. Elsevier Brasil Kang C-J, Chang JT-C, Chen T-M, Chen I-H, Liao C-T, 2003, Surgical treatment of oral verrucous carcinoma. choice 1:2 McClure D, Gullane P, Slinger R, Wysocki G, 1984, Verrucous carcinoma–changing concepts in management. J Otolaryngol 13(1):7–12 Warshaw EM, Templeton S, Washington C, 2000, Verrucous carcinoma occurring in a lesion of oral lichen planus. Cutis New York 65(4):219–222 Rinker MH, Fenske NA, Scalf LA, Glass LF, 2001, Histologic variants of squamous cell carcinoma of the skin. Cancer Control 8(4):354–363 Lubbe J, Kormann A, Adams V, Hassam S, Gratz K, Panizzon R, Burg G, 1996, HPV-11-and HPV-16-associated oral verrucous carcinoma. Dermatology 192(3):217–221 Cooper JS, Pajak TF, Forastiere AA, Jacobs J, Campbell BH, Saxman SB, Kish JA, Kim HE, Cmelak AJ, Rotman M, 2004, Postoperative concurrent radiotherapy and chemotherapy for high-risk squamous-cell carcinoma of the head and neck. N Engl J Med 350(19):1937–1944 Thompson L, 2006, World Health Organization classification of tumours: pathology and genetics of head and neck tumours. Ear Nose Throat J 85(2):74–74 Gale N, Zidar N, 2006, Benign and potentially malignant lesions of the squamous epithelium and squamous cell carcinoma. Springer Moher D, Liberati A, Tetzlaff J, Altman DG, 2009, Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med 151(4):264–269 Angadi CV, Angadi VP, 2015, GLUT-1 immunoexpression in oral epithelial dysplasia, oral squamous cell carcinoma, and verrucous carcinoma. J Oral Sci 57(2):115–122 Wang Y-H, Tian X, Liu O-S, Fang X-D, Quan H-Z, Xie S, Gao S, Tang Z-G, 2014, Gene profiling analysis for patients with oral verrucous carcinoma and oral squamous cell carcinoma. Int J Clin Exp Med 7(7):1845 El-Rouby DH, 2010, Association of macrophages with angiogenesis in oral verrucous and squamous cell carcinomas. J Oral Pathol Med 39(7):559–564 Gao H, Ho J, Lee H, Yu C, 2009, The presence ofMerkel cells and CD10-and CD34-positive stromal cells compared in benign and malignant oral tumors. Oral Dis 15(4):259–264 Paral KM, Taxy JB, LingenMW(2014, CD34 and α smooth muscle actin distinguish verrucous hyperplasia from verrucous carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol 117(4):477–482 Laxmidevi LB, Angadi PV, Pillai RK, Chandreshekar C, 2010, Aberrant. BETA-catenin expression in the histologic differentiation of oral squamous cell carcinoma and verrucous carcinoma: an immunohistochemical study. J Oral Sci 52(4):633–640 Oliveira M, Silveira E, Godoy G, Amorim R, Costa A, Queiroz L, 2005, Immunohistochemical evaluation of intermediate filament proteins in squamous papilloma and oral verrucous carcinoma. Oral Dis 11(5):288–292 MohtashamN, Babakoohi S, ShivaA, Shadman A,Kamyab-Hesari K, Shakeri M, Sharifi-SistaniN(2013, Immunohistochemical study of p53, Ki-67, MMP-2 and MMP-9 expression at invasive front of squamous cell and verrucous carcinoma in oral cavity. Pathol, Res Pract 209(2):110 Zargaran M, Eshghyar N, Vaziri PB, Mortazavi H, 2011, Immunohistochemical evaluation of type IV collagen and laminin-332 γ2 chain expression in well-differentiated oral squamous cell carcinoma and oral verrucous carcinoma: a new recommended cut-off. J Oral Pathol Med 40(2):167–173 Arduino PG, Carrozzo M, Pagano M, Broccoletti R, Scully C, Gandolfo S, 2010, Immunohistochemical expression of basement membrane proteins of verrucous carcinoma of the oralmucosa. Clin Oral Investig 14(3):297–302 Ray JG, Mukherjee S, Pattanayak S, Chaudhuri K, 2011, Oral verrucous carcinoma–a misnomer? Immunohistochemistry based comparative study of two cases. BMJ Case Rep 2011., DOI 10.1136/bcr.11.2010.3479 Nishikawa T, Hara R, Ito Y, Masuno K, Tominaga K, Wato M, Watanabe Y, Mori M, Tanaka A, 2005, Aberrant expression patterns of histone H3 mRNA, p53, cyclin D1, and cyclin B1 in oral neoplastic epithelial lesions. Acta Histochem Cytochem 38(1):53–59 Tran TN, Liu Y, Takagi M, Yamaguchi A, Fujii H, 2005, Frequent promoter hypermethylation of RASSF1A and p16INK4a and infrequent allelic loss other than 9p21 in betel-associated oral carcinoma in a Vietnamese non-smoking/non-drinking female population. J Oral Pathol Med 34(3):150–156 Adegboyega PA, Boromound N, Freeman DH, 2005, Diagnostic utility of cell cycle and apoptosis regulatory proteins in verrucous squamous carcinoma. Appl ImmunohistochemMolMorphol 13(2): 171–177 Lin HP, Wang YP, Chiang CP, 2011, Expression of p53, MDM2, p21, heat shock protein 70, and HPV 16/18 E6 proteins in oral verrucous carcinoma and oral verrucous hyperplasia. Head Neck 33(3):334–340 Pentenero M, Donadini A, Di Nallo E, Maffei M, Marino R, Familiari U, Broccoletti R, Castagnola P, Gandolfo S, Giaretti W, 2011, Distinctive chromosomal instability patterns in oral verrucous and squamous cell carcinomas detected by highresolution DNA flow cytometry. Cancer 117(22):5052–5057 Wang Y, Tang Z, Zhao S, Xie X, Yao Z, Liu C, Qu B, Chen Y, Xiao Z, Li G, 2013, Proteomic analysis of human oral verrucous carcinoma. Afr J Biotechnol 10(60):13004–13013 Quan H, Tang Z, Zhao L, Wang Y, Liu O, Yao Z, Zuo J, 2012, Expression of αB-crystallin and its potential anti-apoptotic role in oral verrucous carcinoma. Oncol Lett 3(2):330 de Spindula-Filho JV, da Cruz AD, Oton-Leite AF, Batista AC, Leles CR, Alencar RCG, Saddi VA, Mendonca EF, 2011, Oral squamous cell carcinoma versus oral verrucous carcinoma: an approach to cellular proliferation and negative relation to human papillomavirus, HPV). Tumor Biol 32(2):409–416 Terada T, 2011, Verrucous carcinoma of the oral cavity: a histopathologic study of 10 Japanese cases. J Maxillofac Oral Surg 10(2):148–151 Zargaran M, Eshghyar N, Baghaei F, Moghimbeigi A, 2012, Assessment of cellular proliferation in oral verrucous carcinoma and well-differentiated oral squamous cell carcinoma using Ki67: a non-reliable factor for differential diagnosis? Asian Pac J Can Prev: APJCP 13(11):5811 Terada T, 2012, Squamous cell carcinoma arising within verrucous carcinoma of the oral cavity: a case report. Int J Clin Exp Pathol 5(4):363 Odar K, Bostjancic E, Gale N, Glavac D, Zidar N, 2012, Differential expression of microRNAs miR-21, miR-31, miR-203, miR-125a-5p and miR-125b and proteins PTEN and p63 in verrucous carcinoma of the head and neck. Histopathology 61(2):257 Patil GB, Hallikeri KS, Balappanavar AY, Hongal SG, Sanjaya PR, Sagari SG, 2013, Cyclin B1 overexpression in conventional oral squamous cell carcinoma and verrucous carcinoma-A correlation with clinicopathological features. Med Oral, Patol Oral Cir Bucal 18(4):e585 Mallick S, Breta M, Gupta SD, Dinda AK, Mohanty BK, Singh MK, 2015, Angiogenesis, proliferative activity and dna ploidy in oral verrucous carcinoma: a comparative study including verrucous hyperplasia and squamous cell carcinoma. Pathol Oncol Res 21(4): 1249–1257 Samman M, Wood HM, Conway C, Stead L, Daly C, Chalkley R, Berri S, Senguven B, Ross L, Egan P, 2015, A novel genomic signature reclassifies an oral cancer subtype. Int J Cancer 137(10): 2364–2373 Klieb HBE, Raphael SJ, 2007, Comparative study of the expression of p53, Ki67, E-cadherin andMMP-1 in verrucous hyperplasia and verrucous carcinoma of the oral cavity. Head and neck pathology 1(2):118–122 Poh CF, Zhang L, Lam WL, Zhang X, An D, Chau C, Priddy R, Epstein J, Rosin MP, 2001, A high frequency of allelic loss in oral verrucous lesions may explain malignant risk. Lab Investig 81(4): 629–634 Ho P-S, Chen P-L, Warnakulasuriya S, Shieh T-Y, Chen Y-K, Huang I-Y, 2009)Malignant transformation of oral potentially malignant disorders in males: a retrospective cohort study. BMC Cancer 9(1):260 Barnes L, Eveson J, Reichart P, Sidransky D, 2005, Oral cavity and oropharynx. World Health Organization Classification of tumors Pathology and genetics of Head and Neck tumors Lyon: International Agency for Research on Cancer Press:164–208 Niparko J, RubinsteinM, McClatchey K, 1988, Invasive squamous cell carcinoma within verrucous carcinoma. J Otolaryngol 17(1): 38–40 Rajendran R, Varghese I, Sugathan C, 1988, Ackerman’s tumour, verrucous carcinoma, of the oral cavity: a clinico-epidemiologic study of 426 cases. Aust Dent J 33(4):295–298 Jyothirmayi R, Sankaranarayanan R, Varghese C, Jacob R, Nair MK, 1997, Radiotherapy in the treatment of verrucous carcinoma of the oral cavity. Oral Oncol 33(2):124–128 Spiro RH, 1998, Verrucous carcinoma, then and now. Am J Surg 176(5):393–397 Yoshimura Y, Mishima K, Obara S, Nariai Y, Yoshimura H, Mikami T, 2001, Treatment modalities for oral verrucous carcinomas and their outcomes: contribution of radiotherapy and chemotherapy. Int J Clin Oncol 6(4):192–200 Sturgeon C, Hammond E, Chang S, Soletormos G, Hayes D, 2006, NACB: practice guidelines and recommendations for use of tumor markers in the clinic: quality requirements [section 2] 2008. Draft Guidelines Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL, 1987, Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235(4785):177–182 Hayes DF, Bast RC, Desch CE, Fritsche H, Kemeny NE, Jessup JM, Locker GY, Macdonald JS, Mennel RG, Norton L, 1996, Tumor marker utility grading system: a framework to evaluate clinical utility of tumor markers. J Natl Cancer Inst 88(20):1456–1466 Kurman RJ, Norris HJ, 1976, Endodermal sinus tumor of the ovary. A clinical and pathologic analysis of 71 cases. Cancer 38(6):2404– 2419 Cuperlovic-Culf M, Belacel N, Ouellette RJ, 2005, Determination of tumour marker genes from gene expression data. Drug Discov Today 10(6):429–437 Copper D, Schermer A, Sun T, 1985, Classification of human epithelia and their neoplasms using monoclonal antibodies to keratin: strategies, applications and limitations. Lab Investig 52:243–256 Gown A, Vogel A, 1984, Monoclonal antibodies to human intermediate filament proteins. II. Distribution of filament proteins in normal human tissues. Am J Pathol 114(2):309 Moll R, 1998, Cytokeratins as markers of differentiation in the diagnosis of epithelial tumors. Subcell Biochem 31:205–262 Logan CY, Nusse R, 2004, The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol 20:781–810 Queimado L, Obeso D, HatfieldMD, Yang Y, Thompson DM, Reis AM, 2008, Dysregulation of Wnt pathway components in human salivary gland tumors. Archives Otolaryngol–Head Neck Surg 134(1):94–101 Yu Z,Weinberger PM, Provost E, Haffty BG, Sasaki C, Joe J, Camp R, Rimm D, Psyrri A, 2005, β-catenin functions mainly as an adhesion molecule in patients with squamous cell cancer of the head and neck. Clin Cancer Res 11(7):2471–2477 Wilson DF, De-Jun J, Pierce AM, Wiebkin OW, 1999, Oral cancer: role of the basementmembrane in invasion. Aust Dent J 44(2):93–97 Souza LFP, Souza VF, Silva LDG, Santos JN, Reis SRA, 2007, Expression of basement membrane laminin in oral squamous cell carcinomas. Rev Bras Otorrinolaringol 73(6):768–774 Pinto GA, Vassallo J, Andrade LALDA, Magna LA, 1998, Immunohistochemical study of basement membrane collagen IV in uterine cervix carcinoma. Sao Paulo Medical Journal 116(6): 1846–1851 Carvalho KC, Cunha IW, Rocha RM, Ayala FR, Cajaiba MM, Begnami MD, Vilela RS, Paiva GR, Andrade RG, Soares FA, 2011, GLUT1 expression in malignant tumors and its use as an immunodiagnostic marker. Clinics 66(6):965–972 Barth PJ, Zu Schweinsberg TS, Ramaswamy A, Moll R, 2004, CD34+ fibrocytes, α-smooth muscle antigen-positive myofibroblasts, and CD117 expression in the stroma of invasive squamous cell carcinomas of the oral cavity, pharynx, and larynx. Virchows Arch 444(3):231–234 Ramaswamy A, Moll R, Barth PJ, 2003, CD34+ fibrocytes in tubular carcinomas and radial scars of the breast. Virchows Arch 443(4):536–540 Erdag G, Qureshi HS, Patterson JW, Wick MR, 2008, CD34- positive dendritic cells disappear from scars but are increased in pericicatricial tissue. J Cutan Pathol 35(8):752–756 Barth PJ, Moll R, Ramaswamy A, 2005, Stromal remodeling and SPARC, secreted protein acid rich in cysteine, expression in invasive ductal carcinomas of the breast. Virchows Arch 446(5):532– 536 Betz P, Nerlich A, Wilske J, Tubel J, Wiest I, Penning R, Eisenmengen W, 1992, Time-dependent pericellular expression of collagen type IV, laminin, and heparan sulfate proteoglycan in myofibroblasts. Int J Legal Med 105(3):169–172 Cotran RS, Kumar V, Collins T, Robbins SL, 1999, Robbins pathologic basis of disease.62–63, 82, 206–209, 302–303, 328–331 MohtashamN, Babakoohi S, ShivaA, Shadman A,Kamyab-Hesari K, Shakeri M-T, Sharifi-Sistani N, 2013, Immunohistochemical study of p53, Ki-67, MMP-2 and MMP-9 expression at invasive front of squamous cell and verrucous carcinoma in oral cavity. Pathol Res Prac 209(2):110–114 Cardon-Cardo C, 1995, Mutations of cell cycle regulators: biological and clinical implications for human neoplasia. Am J Pathol 147:545–560 Nozoe T, Korenaga D, Kabashima A, Ohga T, Saeki H, Sugimachi K, 2002, Significance of cyclin B1 expression as an independent prognostic indicator of patients with squamous cell carcinoma of the esophagus. Clin Cancer Res 8(3):817–822 Saito T, Nakajima T, Mogi K, 1999, Immunohistochemical analysis of cell cycle-associated proteins p16, pRb, p53, p27 andKi-67 in oral cancer and precancer with special reference to verrucous carcinomas. J Oral Pathol Med 28(5):226–232 Todd R, Hinds P, Munger K, Rustgi A, Opitz O, Suliman Y, Wong D, 2002, Cell cycle dysregulation in oral cancer. Crit Rev Oral Biol Med 13(1):51–61 Evan GI,VousdenKH(2001, Proliferation, cell cycle and apoptosis in cancer. Nature 411(6835):342–348 Macluskey M, Chandrachud LM, Pazouki S, Green M, Chisholm DM, Ogden GR, Schor SL, Schor AM, 2000, Apoptosis, proliferation, and angiogenesis in oral tissues. Possible relevance to tumour progression. J Pathol 191(4):368–375 Rousseau A, Lim M, Lin Z, Jordan R, 2001, Frequent cyclin D1 gene amplification and protein overexpression in oral epithelial dysplasias. Oral Oncol 37(3):268–275 Yuan Y, Zeng Z-Y, Liu X-H, Gong D-J, Tao J, Cheng H-Z, Huang S-D, 2011)MicroRNA-203 inhibits cell proliferation by repressing ΔNp63 expression in human esophageal squamous cell carcinoma. BMC Cancer 11(1):57 Saini S, Majid S, Yamamura S, Tabatabai L, Suh SO, Shahryari V, Chen Y, Deng G, Tanaka Y, Dahiya R, 2011, Regulatory role of miR-203 in prostate cancer progression and metastasis. Clin Cancer Res 17(16):5287–5298 Furuta M, Kozaki K-I, Tanaka S, Arii S, Imoto I, Inazawa J, 2009, miR-124 and miR-203 are epigenetically silenced tumorsuppressive microRNAs in hepatocellular carcinoma. Carcinogenesis 31:766–776 Bueno MJ, de Castro IP, de Cedron MG, Santos J, Calin GA, Cigudosa JC, Croce CM, Fernandez-Piqueras J, Malumbres M, 2008, Genetic and epigenetic silencing of microRNA-203 enhances ABL1 and BCR-ABL1 oncogene expression. Cancer Cell 13(6):496–506 Lena A, Shalom-Feuerstein R, di Val Cervo PR, Aberdam D, Knight R,Melino G, Candi E, 2008, miR-203 represses’ stemness’ by repressing Np63. Cell Death Differ 15(7):1187 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 19 EP 32 DI 10.1007/s12253-016-0150-x PG 14 ER PT J AU Reis, H Bertram, S Pott, L Canbay, A Gallinat, A Baba, AH AF Reis, Henning Bertram, Stefanie Pott, Leona Canbay, Ali Gallinat, Anja Baba, Andreas Hideo TI Markers of Hippo-Pathway Activity in Tumor Forming Liver Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hippo-pathway; HCC; Hepatocellular adenoma; FNH; Cirrhosis; Immunohistochemistry ID Hippo-pathway; HCC; Hepatocellular adenoma; FNH; Cirrhosis; Immunohistochemistry AB Hepatocellular Carcinoma (HCC) is a lethal cancer worldwide. Recently, the hippo signaling pathway has been implicated in tumorigenesis of HCC and other malignant tumors. Aim of the study was therefore to evaluate the hippo signaling pathway activity and its clinico-pathological associations and crosstalk in different tumor forming hepatocellular lesions (HCC, hepatocellular adenoma (HCA), focal nodular hyperplasia (FNH) and cirrhosis). A tissue micro array (TMA) from paired human tumorous and non-tumorous (NT) tissue samples of HCC (n = 92), HCA (n = 25), FNH (n = 28) and cirrhosis (n = 28; no NT) was constructed. The hippo-pathway related proteins of MST1/2, (nuclear(n)/cytoplasmic(c)) YAP and (phospho(p)) TAZ and interactors as Glypican3, RASSF1a, pAKT, pERK and pP70S6K were evaluated by immunohistochemistry (IHC). Proliferation was assessed by Ki67-IHC and apoptosis by TUNEL-technique. MST1/2- and nYAP-immunoreactivity was associated with lymph node status (p = 0.048, p = 0.001), higher grading (p = 0.012, p = 0.24) and unfavorable relapse-free survival (p = 0.004, p = 0.003). MST1/2, c/nYAP and pTAZ were significantly different between HCC/NT (p < 0.001, p = 0.029, p < 0.001, p < 0.001) and mono−/polyclonal hepatocellular lesions (HCC/HCA vs. FNH/cirrhosis; all p ≤ 0.001). Phospho-TAZ-negativity and nYAP-positivity were almost exclusively and MST1/2 exclusively detected in HCC. MST1/2 correlated with pP70S6K (p = 0.002), pERK (p = 0.042), RASSF1a-IRS (p = 0.002) and GPC3 (p < 0.001) and nYAP with GPC3 (p = 0.025), higher Ki67-indices (p = 0.016) and lower apoptosis rate (p = 0.078). MST1/2 and nYAP are unfavorable prognostic markers associated with an aggressive tumor-phenotype in HCC. Positive nYAP- and negative pTAZ-immunostaining were strong indicators of a monoclonal hepatocellular lesion. The unexpected findings for MST1/2 remain to be elucidated. C1 [Reis, Henning] University of Duisburg-Essen, University Hospital of Essen, Institute of Pathology, Hufelandstrasse 55, 45147 Essen, Germany. [Bertram, Stefanie] University of Duisburg-Essen, University Hospital of Essen, Institute of Pathology, Hufelandstrasse 55, 45147 Essen, Germany. [Pott, Leona] University of Duisburg-Essen, University Hospital of Essen, Institute of Pathology, Hufelandstrasse 55, 45147 Essen, Germany. [Canbay, Ali] University of Duisburg-Essen, University Hospital of Essen, Department of Gastroenterology and Hepatology, Hufelandstrasse 55, 45147 Essen, Germany. [Gallinat, Anja] University of Duisburg-Essen, University Hospital of Essen, Department of General, Visceral and Transplantation Surgery, Hufelandstrasse 55, 45147 Essen, Germany. [Baba, Andreas Hideo] University of Duisburg-Essen, University Hospital of Essen, Institute of Pathology, Hufelandstrasse 55, 45147 Essen, Germany. RP Baba, AH (reprint author), University of Duisburg-Essen, University Hospital of Essen, Institute of Pathology, 45147 Essen, Germany. EM Hideo.Baba@uk-essen.de CR Theise ND, 2014, Liver cancer. In: Stewart BW, Wild CP, eds, World cancer report 2014. International Agency for Research on Cancer, Lyon, pp. 403–412 Forner A, Llovet JM, Bruix J, 2012, Hepatocellular carcinoma. Lancet 379(9822):1245–1255., DOI 10.1016/S0140- 6736(11)61347-0 Harvey KF, Pfleger CM, Hariharan IK, 2003, The drosophila Mst ortholog, hippo, restricts growth and cell proliferation and promotes apoptosis. Cell 114(4):457–467 Barron DA, Kagey JD, 2014, The role of the hippo pathway in human disease and tumorigenesis. Clin Transl Med 3:25., DOI 10. 1186/2001-1326-3-25 Jie L, Fan W, Weiqi D, Yingqun Z, Ling X, Miao S, Ping C, Chuanyong G, 2013, The hippo-yes association protein pathway in liver cancer. Gastroenterol Res Pract 2013:187070., DOI 10.1155/ 2013/187070 Hall CA,Wang R, Miao J, Oliva E, Shen X,Wheeler T, Hilsenbeck SG, Orsulic S, Goode S, 2010, Hippo pathway effector yap is an ovarian cancer oncogene. Cancer Res 70(21):8517–8525., DOI 10. 1158/0008-5472.CAN-10-1242 Zhang L, Yang S, Chen X, Stauffer S, Yu F, Lele SM, Fu K, Datta K, Palermo N, Chen Y, Dong J, 2015, The hippo pathway effector, YAP, regulates motility, invasion and castration-resistant growth of prostate cancer cells.Mol Cell Biol 35(8):1350–1362., DOI 10.1128/ MCB.00102-15 Shao DD, Xue W, Krall EB, Bhutkar A, Piccioni F, Wang X, Schinzel AC, Sood S, Rosenbluh J, Kim JW, Zwang Y, Roberts TM, Root DE, Jacks T, Hahn WC, 2014, KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 158(1):171–184., DOI 10.1016/j.cell.2014.06.004 Wang Y, Dong Q, Zhang Q, Li Z, Wang E, Qiu X, 2010, Overexpression of yes-associated protein contributes to progression and poor prognosis of non-small-cell lung cancer. Cancer Sci 101(5):1279–1285., DOI 10.1111/j.1349-7006.2010.01511.x Hong L, Cai Y, Jiang M, Zhou D, Chen L, 2015, The hippo signaling pathway in liver regeneration and tumorigenesis. Acta Biochim Biophys Sin Shanghai 47(1):46–52., DOI 10.1093/abbs/gmu106 Yimlamai D, Christodoulou C, Galli GG, Yanger K, Pepe-Mooney B, Gurung B, Shrestha K, Cahan P, Stanger BZ, Camargo FD, 2014, Hippo pathway activity influences liver cell fate. Cell 157(6):1324–1338., DOI 10.1016/j.cell.2014.03.060 Zhou D, Conrad C, Xia F, Park JS, Payer B, Yin Y, Lauwers GY, Thasler W, Lee JT, Avruch J, Bardeesy N, 2009, Mst1 and Mst2 maintain hepatocyte quiescence and suppress hepatocellular carcinoma development through inactivation of the Yap1 oncogene. Cancer Cell 16(5):425–438., DOI 10.1016/j.ccr.2009.09.026 Xu MZ, Yao TJ, Lee NP, Ng IO, Chan YT, Zender L, Lowe SW, Poon RT, Luk JM, 2009, Yes-associated protein is an independent prognostic marker in hepatocellular carcinoma. Cancer 115(19): 4576–4585., DOI 10.1002/cncr.24495 Han SX, Bai E, Jin GH, He CC, Guo XJ, Wang LJ, Li M, Ying X, Zhu Q, 2014, Expression and clinical significance of YAP, TAZ, and AREG in hepatocellular carcinoma. J Immunol Res 2014: 261365., DOI 10.1155/2014/261365 Chen Q, Zhang N, Gray RS, Li H, Ewald AJ, Zahnow CA, Pan D, 2014, A temporal requirement for hippo signaling in mammary gland differentiation, growth, and tumorigenesis. Genes Dev 28(5):432–437., DOI 10.1101/gad.233676.113 Bioulac-Sage P, Balabaud C, Wanless I, 2010, Focal nodular hyperplasia and hepatocellular adenoma. In: Bosman FT, Carneiro F, Hruban RH, Theise ND, eds, WHO classification of Tumours of the digestive system. International Agency for Research on Cancer, Lyon, pp. 198–204 Theise ND, Curado MP, Franceschi S, Hytiroglou P, KudoM, Park YN, Sakamoto M, Torbeson M, Wee A, 2010, Hepatocellular carcinoma. In: Bosman FT, Carneiro F, Hruban RH, Theise ND, eds, WHO classification of Tumours of the digestive system. International Agency for Research on Cancer, Lyon, pp. 205–216 Sobin LH, Gospodarowicz MK,Wittekind C, 2009, TNM classification of malignant Tumours, 7th edn. Wiley-Blackwell, Hoboken Reis H, Putter C, Megger DA, Bracht T, Weber F, Hoffmann AC, Bertram S, Wohlschlager J, Hagemann S, Eisenacher M, Scherag A, Schlaak JF, Canbay A, Meyer HE, Sitek B, Baba HA, 2015, A structured proteomic approach identifies 14-3-3Sigma as a novel and reliable protein biomarker in panel based differential diagnostics of liver tumors. Biochim Biophys Acta 1854(6):641–650., DOI 10.1016/j.bbapap.2014.10.024 Schmitz KJ, Wohlschlaeger J, Lang H, Sotiropoulos GC, Malago M, Steveling K, Reis H, Cicinnati VR, Schmid KW, Baba HA, 2008, Activation of the ERK and AKTsignalling pathway predicts poor prognosis in hepatocellular carcinoma and ERK activation in cancer tissue is associated with hepatitis C virus infection. J Hepatol 48(1):83–90., DOI 10.1016/j.jhep.2007.08.018 Schmitz KJ, Wohlschlaeger J, Alakus H, Bohr J, Stauder MA, Worm K, Winde G, Schmid KW, Baba HA, 2007, Activation of extracellular regulated kinases, ERK1/2, but not AKT predicts poor prognosis in colorectal carcinoma and is associated with k-ras mutations. Virchows Arch 450(2):151–159., DOI 10.1007/s00428-006- 0342-y Li H, Wolfe A, Septer S, Edwards G, Zhong X, Abdulkarim AB, Ranganathan S, Apte U, 2012, Deregulation of hippo kinase signalling in human hepatic malignancies. Liver Int 32(1):38–47., DOI 10.1111/j.1478-3231.2011.02646.x Zheng Y, Wang W, Liu B, Deng H, Uster E, Pan D, 2015, Identification of Happyhour/MAP4K as alternative Hpo/Mst-like kinases in the hippo kinase Cascade. Dev Cell 34(6):642–655., DOI 10.1016/j.devcel.2015.08.014 Nehme NT, Pachlopnik Schmid J, Debeurme F, Andre-Schmutz I, Lim A, Nitschke P, Rieux-Laucat F, Lutz P, Picard C, Mahlaoui N, Fischer A, de Saint BG, 2012, MST1 mutations in autosomal recessive primary immunodeficiency characterized by defective naive T-cell survival. Blood 119(15):3458–3468., DOI 10.1182/blood- 2011-09-378364 Rawat SJ, Chernoff J, 2015, Regulation of mammalian Ste20, Mst, kinases. Trends Biochem Sci 40(3):149–156., DOI 10.1016/j.tibs. 2015.01.001 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 33 EP 39 DI 10.1007/s12253-016-0079-0 PG 7 ER PT J AU Simon, H Fischer, T Almasi, A Fischer, E AF Simon, Higin Fischer, Tamas Almasi, Attila Fischer, Emil TI Effects of Mesalazine on Morphological and Functional Changes in the Indomethacin-Induced Inflammatory Bowel Disease (Rat Model of Crohn’s Disease) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Inflammatory bowel disease; Crohn’s disease; Indomethacin; Mesalazine; p-Nitrophenol ID Inflammatory bowel disease; Crohn’s disease; Indomethacin; Mesalazine; p-Nitrophenol AB Morphological and functional changes have been investigated in the rat model of Crohn’s disease. The inflammatory bowel disease was induced by indomethacin (1 × 10 mg/kg s.c. for 3 days). Morphological alterations were evaluated by macroscopic scoring system and on the base of histological changes in the small intestine. Functional activities were studied by determination of the intestinal and hepatic elimination of p-Nitrophenol (PNP) and its metabolites (PNP-glucuronide: PNP-G and PNP-sulfate: PNP-S) during the luminal perfusion of PNP. It was found that the indomethacin induced severe macroscopic changes (hyperaemia, petechia, bleeding, erosions, ulcerations) and significant histological alterations in the small intestine of rats which were definitely inhibited by mesalazine (1000 mg/kg by gastric tube for 3 days). Disappearance of PNP from the luminal perfusion solution was diminished by indomethacin which was corrected by administration of mesalazine. Significant depression was found in the luminal appearance of PNP metabolites by giving of indomethacin and these alterations could not be compensated by mesalazine. Hepatic elimination of PNP (biliary excretion of PNP and its metabolites) was decreased definitely by indomethacin which was – at least partly - compensated by mesalazine. The findings of the present study suggest that the indomethacin-induced inflammation in the small intestine represents a useful rat model of Crohn’s disease. Morphological and functional alterations caused by indomethacin can be compensated by mesalazine. C1 [Simon, Higin] Pecsi Tudomanyegyetem, Gyogyszereszeti Intezet, 12 Szigeti utPecs, Hungary. [Fischer, Tamas] Pecsi Tudomanyegyetem, Gyogyszereszeti Intezet, 12 Szigeti utPecs, Hungary. [Almasi, Attila] Pecsi Tudomanyegyetem, Gyogyszereszeti Intezet, 2 Rokus utcaPecs, Hungary. [Fischer, Emil] Pecsi Tudomanyegyetem, Gyogyszereszeti Intezet, 12 Szigeti utPecs, Hungary. RP Fischer, E (reprint author), Pecsi Tudomanyegyetem, Gyogyszereszeti Intezet, Pecs, Hungary. EM emil.fischer@aok.pte.hu CR Stewart TH, Hetenyi C, Roswell H, Orizaga M, 1980, Ulcerative enterocolitis in dogs induced by drugs. J Pathol 131:363–378 Kent TH, Cardelli RM, Stamler FW, 1969, Small intestinal ulcers and intestinal flora in rats given indomethacin. Am J Pathol 54: 237–239 Colpaert S, Liu Z, DeGreef B, Rutgeerts P, Ceuppens JL,Geboes K, 2001, Effects of anti-tumour necrosis factor, interleukin-10 and antibiotic therapy in the indometacin-induced bowel inflammation rat model. Aliment Pharmacol Ther 15:1827–1836 Cooper RA, 1977, Abnormalities of cell-membrane fluidity in the pathogenesis of disease. N Engl J Med 297:371–377 Satoh H, Guth PH, GrossmanMI, 1983, Role of bacteria in gastric ulceration produced by indomethacin in the rat: cytoprotective action of antibiotics. Gastroenterology 84:483–489 Gullikson GW, Sender M, Bass P, 1982, Laxative-like effects of nonsteroidal anti-inflammatory drugs on intestinal fluid movement and membrane integrity. J Pharmacol Exp Ther 220:236–242 Matsumoto T, Iida M, Kuroki F, Hizawa K, Koga H, Fujishima M, 1994, Effects of diet on experimentally induced intestinal ulcers in rats: morphology and tissue leukotrienes. Gut 35:1058–1063 Yamada T, HoshinoM, Hayakawa T, Kamiya Y, Ohhara H,Mizuno K, Yamada H, Nakazawa T, Inagaki T, Uchida A, Miyaji M, Takeuchi T, 1996, Bile secretion in rats with indomethacininduced intestinal inflammation. Am J Phys 270:G804–G812 Reuter BK, Davies NM, Wallace JL, 1997, Nonsteroidal antiinflammatory drug enteropathy in rats: role of permeability, bacteria, and enterohepatic circulation. Gastroenterology 112:109–117 Anthony A, Pounder RE, Dhillon AP, Wakefield AJ, 2000, Similarities between ileal Crohn's disease and indomethacin experimental jejunal ulcers in the rat. Aliment Pharmacol Ther 14:241– 245 Ford J, Martin SW, Houston JB, 1995, Assessment of intestinal permeability changes induced by nonsteroidal anti-inflammatory drugs in the rat. Toxicol Metals 34:9–16 Eadsforth CV, Coveney PC, 1984)Measurement of phenol in urine using a high-performance liquid chromatographic method. Analyst 109:175–176 Kothare PA, Zimmerman CL, 2002, Intestinal metabolism: the role of enzyme localization in phenol metabolite kinetics. Drug Metab Dispos 30:586–594 Kuhn UD, Rost M, Muller D, 2001, Para-nitrophenol glucuronidation and sulfation in rat and human liver slices. Exp Toxicol Pathol 53:81–87 Almasi A, Fischer E, Perjesi P, 2006, A simple and rapid ion-pair HPLC method for simultaneous quantitation of 4-nitrophenol and its glucuronide and sulfate conjugates. J BiochemBiophysMethods 69:43–50 Almasi A, Fischer E, Perjesi P, 2011, HPLC quantification of 4- nitrophenol and its conjugated metabolites from bile. Sci Pharm 79: 837–847 Almasi A, Bojcsev S, Fischer T, Simon H, Perjesi P, Fischer E, 2013, Metabolic enzyme activities and drug excretion in the small intestine and in the liver in the rat. Acta Physiol Hung 100:478–488 Yamada T, Deitch E, Specian RD, Perry MA, Sartor RB, Grisham MB, 1993, Mechanisms of acute and chronic intestinal inflammation induced by indomethacin. Inflammation 17:641–662 Turek JJ, Schoenlein IA, 1993, Indomethacin-induced gastrointestinal ulcers in rats: effects of dietary fatty acids and endotoxin. Prostaglandins Leukot Essent Fat Acids 48:229–232 Nygard G, Anthony A, Piasecki C, Trevethick MA, Hudson M, Dhillon AP, Pounder RE, Wakefield AJ, 1994, Acute indomethacin-induced jejunal injury in the rat: early morphological and biochemical changes. Gastroenterology 106:567–575 Peeters M, Geypens B, Claus D, Nevens H, Ghoos Y, Verbeke G, Baert F, Vermeire S, Vlietinck R, Rutgeerts P, 1997, Clustering of increased small intestinal permeability in families with Crohn's disease. Gastroenterology 102:1546–1550 Bjarnason I, Smethurst P, Macpherson A, Walker F, McElnay JC, Passmore AP, Menzies IS, 1992, Glucose and citrate reduce the permeability changes caused by indomethacin in humans. Gastroenterology 102:1546–1550 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 41 EP 46 DI 10.1007/s12253-016-0069-2 PG 6 ER PT J AU Zhao, Chb Shi, L Pu, Hh Zhang, Qy AF Zhao, Chun-bo Shi, Lei Pu, Hai-hong Zhang, Qing-yuan TI The Promoting Effect of Radiation on Glucose Metabolism in Breast Cancer Cells under the Treatment of Cobalt Chloride SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Radiation; hypoxia; Glucosemetabolism; Hypoxia inducible factors-1α; Cancer cell ID Radiation; hypoxia; Glucosemetabolism; Hypoxia inducible factors-1α; Cancer cell AB We aimed to investigate the influence of radiation on hypoxia-treated breast cancers cells and its underlying mechanism. We mimicked the hypoxic response in MCF- 7 cells by the treatment of CoCl2. Meanwhile, hypoxic MCF-7 cells induced by CoCl2 or untreated MCF-7 cells were treated with or without radiation, and then treated with or without hypoxia inducible factors-1α (HIF-1α) inhibitor. Subsequently, glucose update and lactate release rate were determined by commercial kits, as well as the expressions of HIF-1α and the glucose metabolic pathway related genes, including fructose biphoshatase 1 (FBP1), glucose transporter 1 (GLUT1), lactate dehydrogenase A (LDHA), hexokinase 2 (HK2), and isocitrate dehydrogenase 2 (IDH20) were detected by western blotting and/or RT-PCR. The results showed that glucose uptake rate and lactate release rate were increased in cells under hypoxia and/or radiation condition compared with untreated cells (p < 0.05), while the addition of HIF-1α inhibitor decreased these rates in hypoxia + radiation treated cells (p < 0.05). In addition, compared with untreated cells, the mRNA and protein levels of HIF-1α were significantly increased under hypoxia and radiation condition (p < 0.05), while which decreased after the addition of HIF-1α inhibitor (p < 0.05). Similar content changing trends (all p < 0.05) were observed in FBP1, IDH2, GLUT1, and LDHA but not HK2. In conclusion, the combination of radiation and hypoxia could promote the glucose metabolism. Furthermore, HIF-1α might inhibit the promoting effect of radiation on glycolysis in hypoxic MCF-7 cells by regulating the glucose metabolic pathway. C1 [Zhao, Chun-bo] the Third Affiliated Hospital of Harbin Medical University, Department of Radiation Oncology, 150086 Harbin, Heilongjiang Province, China. [Shi, Lei] the Fourth Affiliated Hospital of Harbin Medical University, Department of Radiation OncologyHarbin, Heilongjiang Province, China. [Pu, Hai-hong] the Third Affiliated Hospital of Harbin Medical University, Department of Internal Medicine, 150086 Harbin, Heilongjiang Province, China. [Zhang, Qing-yuan] the Third Affiliated Hospital of Harbin Medical University, Department of Internal Medicine, 150086 Harbin, Heilongjiang Province, China. CR Siegel R,Ma J, Zou Z, Jemal A, 2014, Cancer statistics. CA Cancer J Clin 64(1):9–29 Noguchi S, Masuda N, Iwata H, Mukai H, Horiguchi J, Puttawibul P, Srimuninnimit V, Tokuda Y, Kuroi K, Iwase H, 2014, Efficacy of everolimus with exemestane versus exemestane alone in Asian patients with HER2-negative, hormone-receptor-positive breast cancer in BOLERO-2. Breast Cancer 21(6):703–714 Lundgren K, Nordenskjold B, Landberg G, 2009, Hypoxia, snail and incomplete epithelial–mesenchymal transition in breast cancer. Br J Cancer 101(10):1769–1781 Vaupel P, Mayer A, Hockel M, 2004, Tumor hypoxia and malignant progression. Methods Enzymol 381:335–354 Mees G, Dierckx R, Vangestel C, Van deWiele C, 2009, Molecular imaging of hypoxia with radiolabelled agents. Eur J Nucl Med Mol Imaging 36(10):1674–1686 Zeng W, Liu P, Pan W, Singh SR, Wei Y, 2015, Hypoxia and hypoxia inducible factors in tumor metabolism. Cancer Lett 356(2):263–267 Gatenby RA, Gillies RJ, 2004, Why do cancers have high aerobic glycolysis? Nat Rev Cancer 4(11):891–899 Dasu A, Toma-Dasu I, KarlssonM(2003, Theoretical simulation of tumour oxygenation and results from acute and chronic hypoxia. Phys Med Biol 48(17):2829 Denko NC, Fontana LA, Hudson KM, Sutphin PD, Raychaudhuri S, Altman R, Giaccia AJ, 2003, Investigating hypoxic tumor physiology through gene expression patterns. Oncogene 22(37):5907– 5914 Brahimi-Horn MC, Chiche J, Pouyssegur J, 2007, Hypoxia signalling controls metabolic demand. Curr Opin Cell Biol 19(2):223–229 J-w K, Tchernyshyov I, Semenza GL, Dang CV, 2006, HIF-1- mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab 3(3): 177–185 Semenza GL, 2011, Oxygen sensing, homeostasis, and disease. New England J Med 365(6):537–547 Germain S, Monnot C, Muller L, Eichmann A, 2010, Hypoxiadriven angiogenesis: role of tip cells and extracellular matrix scaffolding. Curr Opin Hematol 17(3):245–251 Baker L, Boult J,Walker-Samuel S, Chung Y, Jamin Y, Ashcroft M, Robinson S, 2012, The HIF-pathway inhibitor NSC-134754 induces metabolic changes and anti-tumour activity while maintaining vascular function. Br J Cancer 106(10):1638–1647 Cairns RA, Papandreou I, Sutphin PD, Denko NC, 2007, Metabolic targeting of hypoxia and HIF1 in solid tumors can enhance cytotoxic chemotherapy. Proc Natl Acad Sci 104(22):9445– 9450 Hu Y, Liu J, Huang H, 2013, Recent agents targeting HIF-1α for cancer therapy. J Cell Biochem 114(3):498–509 Lu H, Li X, Luo Z, Liu J, Fan Z, 2013, Cetuximab reverses the Warburg effect by inhibiting HIF-1–regulated LDH-A.Mol Cancer Ther 12(10):2187–2199 Lagadec C, Dekmezian C, Bauche L, Pajonk F, 2012, Oxygen levels do not determine radiation survival of breast cancer stem cells. PLoS One 7(3):29 Chandel N, Maltepe E, Goldwasser E, Mathieu C, Simon M, Schumacker P, 1998, Mitochondrial reactive oxygen species trigger hypoxia-induced transcription. Proc Natl Acad Sci 95(20): 11715–11720 Jiang B-H, Zheng JZ, Leung SW, Roe R, Semenza GL, 1997, Transactivation and inhibitory domains of hypoxia-inducible factor 1α modulation of transcriptional activity by oxygen tension. J Biol Chem 272(31):19253–19260 An WG, Kanekal M, Simon MC, Maltepe E, Blagosklonny MV, Neckers LM, 1998, Stabilization of wild-type p53 by hypoxiainducible factor 1α. Nature 392(6674):405–408 Wang G, Hazra TK, Mitra S, Lee H-M, Englander EW, 2000, Mitochondrial DNA damage and a hypoxic response are induced by CoCl2 in rat neuronal PC12 cells. Nucleic Acids Symp Ser 28(10):2135–2140 Lagadec C, Dekmezian C, Bauche L, Pajonk F, 2012, Oxygen levels do not determine radiation survival of breast cancer stem cells. PLoS One 7(3):e34545 Hoogsteen I, Marres H, Van Der Kogel A, Kaanders J, 2007, The hypoxic tumour microenvironment, patient selection and hypoxiamodifying treatments. Clin Oncol 19(6):385–396 Horsman MR, Wouters BG, Joiner MC, Overgaard J, 2009, The oxygen effect and fractionated radiotherapy. Basic clinical radiobiology London: Edward Arnold: 207–209 Gillies RJ, Gatenby RA, 2007, Adaptive landscapes and emergent phenotypes: why do cancers have high glycolysis? J Bioenerg Biomembr 39(3):251–257 Ganapathy V, Thangaraju M, Prasad PD, 2009, Nutrient transporters in cancer: relevance to Warburg hypothesis and beyond. Pharmacol Ther 121(1):29–40 Harrison L, Blackwell K, 2004, Hypoxia and anemia: factors in decreased sensitivity to radiation therapy and chemotherapy? The Oncologist 9(Supplement 5):31–40 Sattler UG, Mueller-KlieserW, 2009, The anti-oxidant capacity of tumour glycolysis. Int J Radiat Biol 85(11):963–971 DongC,Yuan T, Wu Y,WangY, Fan TW, Miriyala S, LinY,Yao J, Shi J, Kang T, 2013, Loss of FBP1 by snail-mediated repression provides metabolic advantages in basal-like breast cancer. Cancer Cell 23(3):316–331 Jiang P, DuW,Wang X, Mancuso A, Gao X,WuM, YangX, 2011, p53 regulates biosynthesis through direct inactivation of glucose-6- phosphate dehydrogenase. Nat Cell Biol 13(3):310–316 Behrooz A, Ismail-Beigi F, 1997, Dual control of glut1 glucose transporter gene expression by hypoxia and by inhibition of oxidative phosphorylation. J Biol Chem 272(9):5555–5562 Iyer NV, Kotch LE, Agani F, Leung SW, Laughner E,Wenger RH, Gassmann M, Gearhart JD, LawlerAM, Aimee YY, 1998, Cellular and developmental control ofO2 homeostasis by hypoxia-inducible factor 1α. Genes Dev 12(2):149–162 Ward PS, Thompson CB, 2012, Signaling in control of cell growth and metabolism. Cold Spring Harb Perspect Biol 4(7):a006783 Fantin VR, St-Pierre J, Leder P, 2006, Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell 9(6):425–434 Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ, 2009, IDH1 and IDH2 mutations in gliomas. New England J Med 360(8): 765–773 Drabovich AP, Pavlou MP, Dimitromanolakis A, Diamandis EP, 2012, Quantitative analysis of energy metabolic pathways in MCF-7 breast cancer cells by selected reaction monitoring assay. Mol Cell Proteomics 11(8):422–434 Lee J-W, Bae S-H, Jeong J-W, Kim S-H, Kim K-W, 2004, Hypoxia-inducible factor, HIF-1, α: its protein stability and biological functions. Exp Mol Med 36(1):1–12 Wang B,Wood IS, Trayhurn P, 2007, Dysregulation of the expression and secretion of inflammation-related adipokines by hypoxia in human adipocytes. Pflugers Arch 455(3):479–492 Ellinghaus P, Heisler I, Unterschemmann K, Haerter M, Beck H, Greschat S, Ehrmann A, Summer H, Flamme I, Oehme F, 2013, BAY 87-2243, a highly potent and selective inhibitor of hypoxiainduced gene activation has antitumor activities by inhibition of mitochondrial complex I. Cancer Med 2(5):611–624 Tennant DA, Duran RV, Gottlieb E, 2010, Targeting metabolic transformation for cancer therapy. Nat Rev Cancer 10(4): 267–277 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 47 EP 53 DI 10.1007/s12253-016-0076-3 PG 7 ER PT J AU Amato, M Perrone, G Righi, D Pellegrini, C Rabitti, C Di Matteo, F Crucitti, P Caputo, D Coppola, R Tonini, G Santini, D Onetti Muda, A AF Amato, Michelina Perrone, Giuseppe Righi, Daniela Pellegrini, Claudio Rabitti, Carla Di Matteo, Francesco Crucitti, Pierfilippo Caputo, Damiano Coppola, Roberto Tonini, Giuseppe Santini, Daniele Onetti Muda, Andrea TI HER2 Status in Gastric Cancer: Comparison between Primary and Distant Metastatic Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HER2; Gastric cancer; FISH; Immunoistochemistry; Metastatic disease ID HER2; Gastric cancer; FISH; Immunoistochemistry; Metastatic disease AB HER2 (human epidermal growth factor receptor-2) assessment in histological samples of gastric cancer is essential to determine which patients might benefit from trastuzumab therapy. HER2 is often evaluated in primary tumor even if trastuzumab therapy is used to treat metastatic disease. However, the exact relationship in terms of HER2 status between primary and metastatic tumors has not been fully clarified. We aimed to evaluate the HER2 status concordance between primary gastric cancer and corresponding distant metastasis. HER2 status was evaluated by IHC (immunohistochemistry) and/or FISH ( fluorescence in situ hybridization) in 41 patients in primary gastric cancer and in paired metastasis. HER2 was assessed according scoring criteria applied in clinical approach. HER2 positivity was found in 14,6 % primary tumors and in 24,4%corresponding metastasis. HER2 concordance rate between primary and metastasis was 80,5%(K-value = 0,388). Eight/41 (19,5 %)cases resulted discordant: 6 patients with metastatic HER2 positive lesions were found HER2 negative in primary cancers while 2 patient HER2 positive in primary lesion showed a negative conversion in metastasis. Our results showed a good concordance in terms of HER2 status between primary and metastatic lesions, as well as in biopsy and surgical removed specimens. However, the higher rate of HER2 positive status found in metastatic lesions underlined the importance of HER2 assessment in all samples obtained from different sites of gastric cancer disease. C1 [Amato, Michelina] University of Rome, Campus Bio-Medico, Department of Pathology, Via Alvaro del Portillo, 200Rome, Italy. [Perrone, Giuseppe] University of Rome, Campus Bio-Medico, Department of Pathology, Via Alvaro del Portillo, 200Rome, Italy. [Righi, Daniela] University of Rome, Campus Bio-Medico, Department of Pathology, Via Alvaro del Portillo, 200Rome, Italy. [Pellegrini, Claudio] University of Rome, Campus Bio-Medico, Department of Pathology, Via Alvaro del Portillo, 200Rome, Italy. [Rabitti, Carla] University of Rome, Campus Bio-Medico, Department of Pathology, Via Alvaro del Portillo, 200Rome, Italy. [Di Matteo, Francesco] University of Rome, Campus Bio-Medico, Endoscopic Unit, Via Alvaro del Portillo, 200Rome, Italy. [Crucitti, Pierfilippo] University of Rome, Campus Bio-Medico, Department of Surgery, Via Alvaro del Portillo, 200Rome, Italy. [Caputo, Damiano] University of Rome, Campus Bio-Medico, Department of Surgery, Via Alvaro del Portillo, 200Rome, Italy. [Coppola, Roberto] University of Rome, Campus Bio-Medico, Department of Surgery, Via Alvaro del Portillo, 200Rome, Italy. [Tonini, Giuseppe] University of Rome, Campus Bio-Medico, Oncology Unit, Via Alvaro del Portillo, 200Rome, Italy. [Santini, Daniele] University of Rome, Campus Bio-Medico, Oncology Unit, Via Alvaro del Portillo, 200Rome, Italy. [Onetti Muda, Andrea] University of Rome, Campus Bio-Medico, Department of Pathology, Via Alvaro del Portillo, 200Rome, Italy. RP Perrone, G (reprint author), University of Rome, Campus Bio-Medico, Department of Pathology, Rome, Italy. EM g.perrone@unicampus.it CR Hultman B, Mahteme H, Sundbom M, et al., 2014, Benchmarking of gastric cancer sensitivity to anti-cancer drugs ex vivo as a basis for drug selection in systemic and intraperitoneal therapy. J Exp Clin Cancer Res 21:110., DOI 10.1186/s13046-014-0110-9 Eckstein N, Roper L, Haas B, et al., 2014, Clinical pharmacology of tyrosine kinase inhibitors becoming generic drugs: the regulatory perspective. J Exp Clin Cancer Res 7:15 Bang YJ, Van Cutsem E, Feyereislova A, et al., 2010, ToGA trial investigators. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer, ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376:687–697 Ajani JA, Bentrem DJ, Besh S, et al., 2013, Gastric cancer, version 2.2013: featured updates to the NCCN guidelines. J Natl Compr Cancer Netw 11:531–546 Takehana T, Kunitomo K, Kono K, et al., 2002, Status of c-erbB-2 in gastric adenocarcinoma: a comparative study of immunohistochemistry, fluorescence in situ hybridization and enzymelinkedimmuno- sorbent assay. Int J Cancer 98:833–837 Tanner M, Hollmen M, Junttila TT, et al., 2005, Amplification of HER-2 in gastric carcinoma: association with topoisomerase IIalpha gene amplification, intestinal type, poor prognosis and sensitivity to trastuzumab. Ann Oncol 16:273–278 Gravalos C, Jimeno A, 2008, HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol 19:1523–1529 Hofmann M, Stoss O, Shi D, Buttner R, et al., 2008, Assessment of a HER2 scoring system for gastric cancer: results from a validation study. Histopathology 52:797–805 Marx AH, Tharun L, Muth J, et al., 2009, HER-2 amplification is highly homogenous in gastric cancer. Hum Pathol 40:769–777 Perrone G, Amato M, Callea M, et al., 2012, HER2 amplification status in gastric and gastro-oesophageal junction cancer in routine clinical practice: which sample should be used? Histopathology 61: 134–135 Sobin LH, Gospodarowicz MK, Wittekind C, 2009, TNM classification of malignant tumours, 7th edn. Weinheim, Germany, Wiley, pp. 70–73 Lauren P, 1965, The two histological main types of gastric carcinoma: diffuse and so-calles intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand 64:31–49 Bosman, FT, 2010, Gastric carcinoma. In: WHO classification of tumours of the digestive system IARC. 4th edn. Lyon pp 45–58 Landis JR, Koch GG, 1977, The measurement of observer agreement for categorical data. Biometrics 33:159–174 Niehans GA, Singleton TP, Dykoski D, et al., 1993, Stability of HER-2/neu expression over time and at multiple metastatic sites. J Natl Cancer Inst 85:1230–1235 Vincent-Salomon A, Jouve M, Genin P, 2002, HER2 status in patients with breast carcinoma is not modified selectively by preoperative chemotherapy and is stable during the metastatic process. Cancer 94:2169–2173 Pagni F, Zannella S, Ronchi S, et al., 2013, HER2 status of gastric carcinoma and corresponding lymph node metastasis. Pathol Oncol Res 19:103–109 Kochi M, Fujii M, Masuda S, 2013, Differing deregulation of HER2 in primary gastric cancer and synchronous related metastatic lymph nodes. Diagn Pathol 8:191 Gumusay O, Benekli M, Ekinci O, et al., 2015, Discordances in HER2 status between primary gastric cancer and corresponding metastatic sites. Jpn J Clin Oncol 45:416–421 Kim MA, Lee HJ, Yang HK, et al., 2011, Heterogeneous amplification of ERBB2 in primary lesions is responsible for the discordant ERBB2 status of primary and metastatic lesions in gastric carcinoma. Histopathology 59:822–831 Wei Q, Xu J, Shen L, 2014, HER2 expression in primary gastric cancers and paired synchronous lymph node and liver metastases. A possible road to target HER2 with radionuclides. Tumour Biol 35:6319–6326 Saito T, Nakanishi H, Mochizuki Y, 2015, Preferential HER2 expression in liver metastases and EGFR expression in peritoneal metastases in patients with advanced gastric cancer. Gastric Cancer 18:711–719 Ruschoff J, Hanna W, Bilous M, 2012, HER2 testing in gastric cancer: a practical approach. Mod Pathol 25:637–650 Cho EY, Park K, Do I, et al., 2013, Heterogeneity of ERBB2 in gastric carcinomas: a study of tissue microarray and matched primary and metastatic carcinomas. Mod Pathol 26:677–684 Yarden Y, Sliwkowski MX, 2001, Untangling the ErbB signalling network. Nat Rev Mol Cell Biol 2:127–123 Yu DH, HungMC, 1991, Expression of activated rat neu oncogene is sufficient toinduce experimental metastasis in 3 T3 cells. Oncogene 6:1991–1996 Guy CT, Webster MA, Schaller M, et al., 1992, Expression of the neu protooncogene in the mammary epithelium of transgenic mice induces metastatic disease. Proc Natl Acad Sci U S A 89: 10578–10582 Muller J, O’Connor R, Stulle K, et al., 1988, Characterization of human stomach cancer cell lines. Verh Dtsch Ges Pathol 72: 214–217 Pirrelli M, Caruso ML, Di Maggio M, et al., 2013, Are biopsy specimens predictive of HER2 status in gastric cancer patients? Dig Dis Sci 58:397–404 Wang T, Hsieh ET, Henry P, et al., 2014, Matched biopsy and resection specimens of gastric and gastroesophageal adenocarcinoma show high concordance in HER2 status. Hum Pathol 45:970–975 Yoshida H, Yamamoto N, Taniguchi H, et al., 2014, Comparison of HER2 status between surgically resected specimens and matched biopsy specimens of gastric intestinal-type adenocarcinoma. Virchows Arch 465:145–154 Yang J, Luo H, Li Y, 2012, Intratumoral heterogeneity determines discordant results of diagnostic tests for human epidermal growth factor receptor, HER, 2 in gastric cancer specimens. Cell Biochem Biophys 62:221–228 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 55 EP 61 DI 10.1007/s12253-016-0082-5 PG 7 ER PT J AU Lopez, F Sampedro, T Llorente, LJ Hermsen, M Alvarez-Marcos, C AF Lopez, Fernando Sampedro, Teresa Llorente, L Jose Hermsen, Mario Alvarez-Marcos, Cesar TI Alterations of p14ARF, p15INK4b, and p16INK4a Genes in Primary Laryngeal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DNA methylation; Larynx squamous cell carcinoma; Head and neck squamous cell carcinoma; MS-MLPA; CDKN2B; CDKN2A ID DNA methylation; Larynx squamous cell carcinoma; Head and neck squamous cell carcinoma; MS-MLPA; CDKN2B; CDKN2A AB The 9p21 gene cluster, harboring growth suppressive genes p14ARF, p15INK4b, and p16INK4a, is one of the major aberration hotspots in head and neck cancers.We try to elucidate specific aberrations affecting this region, throughout methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) assay. Methylation of the gene was investigated by MS-MLPA in a well-characterized series of 27 laryngeal squamous cell carcinomas and 20 samples of healthy mucosa. Aberrant promoter hypermethylation was confirmed using and methylation-specific. All samples studied except 3 (11 %) presented losses at 9p21 segment. The most common finding was the small deletion (exon 1α) of the p16INK4a locus (44 %). Deletion of the 9p21 gene cluster was identified in 5 cases (18 %). We only found methylation in 8 samples (30 %) for p15 IK4b-exon 1. Promoter methylation of p14 ARF, p15 IK4b and p16INK4a was not detected in any tumor sample. Methylation-specific polymerase chain reaction confirmed the results. Our data indicate that there may be a subgroup of patients in which epigenetic regulation of 9p21 segment might have little relevance. Nevertheless, MS-MLPA could not be suitable for the study of methylation at this region and further research is required. C1 [Lopez, Fernando] Department of Otorhinolaryngology and Instituto Universitario de Oncologia del Principado de Asturias, Avenida de Roma, 33011 Oviedo, Asturias, Spain. [Sampedro, Teresa] Hospital San Agustin, Department of Medical OncologyAviles, Asturias, Spain. [Llorente, L Jose] Department of Otorhinolaryngology and Instituto Universitario de Oncologia del Principado de Asturias, Avenida de Roma, 33011 Oviedo, Asturias, Spain. [Hermsen, Mario] Department of Otorhinolaryngology and Instituto Universitario de Oncologia del Principado de Asturias, Avenida de Roma, 33011 Oviedo, Asturias, Spain. [Alvarez-Marcos, Cesar] Department of Otorhinolaryngology and Instituto Universitario de Oncologia del Principado de Asturias, Avenida de Roma, 33011 Oviedo, Asturias, Spain. RP Lopez, F (reprint author), Department of Otorhinolaryngology and Instituto Universitario de Oncologia del Principado de Asturias, 33011 Oviedo, Spain. EM flopez_1981@yahoo.es CR WorshamMJ, Chen KM, Tiwari N, Pals G, Schouten JP, Sethi S, et al., 2006, Fine-mapping loss of gene architecture at the CDKN2B, p15INK4b), CDKN2A, p14ARF, p16INK4a), andMTAP genes in head and neck squamous cell carcinoma. Arch Otolaryngol Head Neck Surg 132:409–415 Marcos CA, Alonso-GuervosM, Prado NR, Gimeno TS, Iglesias FD, Hermsen M, et al., 2011, Genetic model of transformation and neoplastic progression in laryngeal epithelium. Head Neck 33:216–224 Laytragoon-Lewin N, Chen F, Castro J, Elmberger G, Rutqvist LE, Lewin F, et al., 2010, DNA content and methylation of p16, DAPK and RASSF1A gene in tumour and distant, normal mucosal tissue of head and neck squamous cell carcinoma patients. Anticancer Res 30:4643–4648 Xing EP, Nie Y, Song Y, Yang GY, Cai YC,Wang LD, et al., 1999, Mechanisms of inactivation of p14ARF, p15INK4b, and p16INK4a genes in human esophageal squamous cell carcinoma. Clin Cancer Res 5:2704–2713 Demokan S, Dalay N, 2011, Role of DNA methylation in head and neck cancer. Clin Epigenetics 2:123–150 Chen K, Sawhney R, KhanM, BenningerMS, Hou Z, Sethi S, et al., 2007, Methylation of multiple genes as diagnostic and therapeutic markers in primary head and neck squamous cell carcinoma. Arch Otolaryngol Head Neck Surg 133:1131–1138 Lopez F, Sampedro T, Llorente JL, Dominguez F, Hermsen M, Suarez C, et al., 2014, Utility of MS-MLPA in DNA methylation profiling in primary laryngeal squamous cell carcinoma. Oral Oncol 50:291–297 Lopez F, Llorente JL, Garcia-Inclan C, Alonso-Guervos M, Cuesta- Albalad MP, Fresno MF, et al., 2011, Genomic profiling of sinonasal squamous cell carcinoma. Head Neck 33:145–153 Schouten JP,McElgunn CJ,WaaijerR, ZwijnenburgD,Diepvens F, Pals G, 2002, Relative quantification of 40 nucleic acid sequences by multiplex ligation dependent probe amplification. Nucl Acids Res 30:e57 Nygren AO, Ameziane N, Duarte HM, Vijzelaar RN, Waisfisz Q, Hess CJ, et al., 2005, Methylation-specific MLPA, MS-MLPA): simultaneous detection of CpG methylation and copy number changes of up to 40 sequences. Nucl Acids Res 33:e128 Furonaka O, Takeshima Y, Awaya H, Ishida H, Kohno N, Inai K, 2004, Aberrant methylation of p 14(ARF), p 15(INK4b, and p 16(INK4a, Genes and location of the primary site in pulmonary squamous cell carcinoma. Pathol Int 54:549–555 Geradts J,Wilson PA, 1996, High frequency of aberrant p16(INK4A, expression in human breast cancer. Am J Pathol 149:15–20 Cabanillas R, Rodrigo JP, Ferlito A, Rinaldo A, FresnoMF, Aguilar C, et al., 2007, Is there an epidemiological link between human papillomavirus DNA and basaloid squamous cell carcinoma of the pharynx? Oral Oncol 43:327–332 Danahey DG, Tobin EJ, Schuller DE, Bier-Laning CM, Weghorst CM, Lang JC, 1999, p16 Mutation frequency and clinical correlation in head and neck cancer. Acta Otolaryngol 119:285–288 Shintani S, Nakahara Y, Mihara M, Ueyama Y, Matsumura T, 2001, Inactivation of the p14(ARF), p15(INK4B, And p16(INK4A, genes is a frequent event in human oral squamous cell carcinomas. Oral Oncol 37:498–504 Stephen JK, Chen KM, Shah V, Havard S, Kapke A, Lu M, et al., 2010, DNA hypermethylation markers of poor outcome in laryngeal cancer. Clin Epigenetics 1:61–69 Wong TS,ManMWL, LamAKY,Wei WI, KwongYL,Yuen APW, 2003, The study of p16 and p15 genemethylation in head and neck squamous cell carcinoma and their quantitative evaluation in plasma by real-time PCR. Eur J Cancer 39:1881–1887 Pierini S, Jordanov SH, Mitkova AV, Chalakov IJ, Melnicharov MB, Kunev KV, et al., 2014, Promoter hypermethylation of CDKN2A, MGMT, MLH1 and DAPK genes in laryngeal squamous cell carcinoma and their associations with clinical profiles of the patients. Head Neck 36:1103–1108 Li X, Gao L, Li H, Gao J, Yang Y, Zhou F, et al., 2013, Human papillomavirus infection and laryngeal cancer risk: a systematic review and meta-analysis. J Infect Dis 207:479–488 Rodrigo JP, Hermsen MA, Fresno MF, Brakenhoff RH, Garcia- Velasco F, Snijders PJ, et al., 2015, Prevalence of human papillomavirus in laryngeal and hypopharyngeal squamous cell carcinomas in northern Spain. Cancer Epidemiol 39:37–41 Ohta S, Uemura H,Matsui Y, Ishiguro H, Fujinami K, Kondo K, et al., 2009, Alterations of p16 and p14ARF genes and their 9p21 locus in oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 107:81–91 Ignatov A, Bischoff J, Schwarzenau C, Krebs T, Kuester D, Herrmann K, et al., 2008, p16 alterations increase the metastatic potential of endometrial carcinoma. Gynecol Oncol 111:365–371 Kobayashi N, Toyooka S, Yanai H, Soh J, Fujimoto N, Yamamoto H, et al., 2008, Frequent p16 inactivation by homozygous deletion or methylation is associated with a poor prognosis in Japanese patients with pleural mesothelioma. Lung Cancer 62:120–125 Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB, 1996, Methylation-specific PCR, a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S A 93:9821–9826 Wojdacz TK, Dobrovic A, 2007, Methylation-sensitive high resolution melting, MS-HRM): a new approach for sensitive and highthroughput assessment of methylation. Nucleic Acids Res 35(6):e41 Pavicic W, Perkio E, Kaur S, Peltomaki P, 2011, Altered methylation at microRNA-associated CpG islands in hereditary and sporadic carcinomas: a methylation-specific multiplex ligation dependent probe amplification, MS-MLPA)-based approach. Mol Med 17: 726–735 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 63 EP 71 DI 10.1007/s12253-016-0083-4 PG 9 ER PT J AU Yang, X Liu, Y Mani, H Olson, J Clawson, G Caruso, C Bruggeman, R Varlotto, MJ Zander, SD Rassaei, N AF Yang, Xuebin Liu, Yongjun Mani, Haresh Olson, Jeffrey Clawson, Gary Caruso, Carla Bruggeman, Richard Varlotto, M John Zander, S Dani Rassaei, Negar TI Biologic Evaluation of Diabetes and Local Recurrence in Non-Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; Epithelial-to-mesenchymal transition; Diabetes ID Non-small cell lung cancer; Epithelial-to-mesenchymal transition; Diabetes AB A recent multicenter study led by our institution demonstrated that local recurrence of non-small cell lung cancer (NSCLC) was significantly more frequent in patients with diabetes, raising the possibility of different tumor biology in diabetics. Epithelial-to-mesenchymal transition (EMT) plays a key role in local tumor recurrence and metastasis. In the present study, we investigated differences of tumor microenvironment between patients with and without diabetes by examining expression of EMT markers. Seventy-nine NSCLC patients were selected from the cohort of our early multicenter study. These patients were classified into 4 groups: 39 with adenocarcinoma with (n = 19) and without (n = 20) diabetes, and 40 with squamous cell carcinoma with (n = 20) andwithout (n = 20) diabetes. Immunohistochemical expression of eight EMT markers was analyzed, including transforming growth factor-beta (TGF-β), epidermal growth factor receptor (EGFR), insulin-like growth factor 1 receptor (IGF-1R), vimentin, E-cadherin, N-cadherin, HtrA1, and beta-catenin. Five markers (E-cadherin, HtrA1, TGF-β, IGF-1R and vimentin) demonstrated significantly higher expression in diabetics than in non-diabetics in both histology types. Ncadherin had higher expression in diabetics, though the difference did not reach statistical significance. EGFR showed a higher expression in diabetics in squamous cell carcinoma only. Beta-catenin was the only marker with no difference in expression between diabetics versus non-diabetics. Our findings suggest that diabetes is associated with enhanced EMT in NSCLC, which may contribute to growth and invasiveness of NSCLC. C1 [Yang, Xuebin] Penn State Hershey Medical center, Department of Pathology and Laboratory Medicine, 500 University Drive, 17033-0850 Hershey, PA, USA. [Liu, Yongjun] Penn State Hershey Medical center, Department of Pathology and Laboratory Medicine, 500 University Drive, 17033-0850 Hershey, PA, USA. [Mani, Haresh] Penn State Hershey Medical center, Department of Pathology and Laboratory Medicine, 500 University Drive, 17033-0850 Hershey, PA, USA. [Olson, Jeffrey] Penn State College of MedicineHershey, PA, USA. [Clawson, Gary] Penn State Hershey Medical center, Department of Pathology and Laboratory Medicine, 500 University Drive, 17033-0850 Hershey, PA, USA. [Caruso, Carla] Penn State Hershey Medical center, Department of Pathology and Laboratory Medicine, 500 University Drive, 17033-0850 Hershey, PA, USA. [Bruggeman, Richard] Penn State Hershey Medical center, Department of Pathology and Laboratory Medicine, 500 University Drive, 17033-0850 Hershey, PA, USA. [Varlotto, M John] University of Massachusetts, Medical School, Department of Radiation OncologyWorcester, MA, USA. [Zander, S Dani] Penn State Hershey Medical center, Department of Pathology and Laboratory Medicine, 500 University Drive, 17033-0850 Hershey, PA, USA. [Rassaei, Negar] Penn State Hershey Medical center, Department of Pathology and Laboratory Medicine, 500 University Drive, 17033-0850 Hershey, PA, USA. RP Rassaei, N (reprint author), Penn State Hershey Medical center, Department of Pathology and Laboratory Medicine, 17033-0850 Hershey, USA. EM nrassaei@hmc.psu.edu CR El-Sherif A, Fernando HC, Santos R, Pettiford B, Luketich JD, Close JM, et al., 2007, Margin and local recurrence after sublobar resection of non-small cell lung cancer. Ann Surg Oncol 14(8): 2400–2405 Martini N, Bains MS, Burt ME, Zakowski MF, McCormack P, Rusch VW, et al., 1995, Incidence of local recurrence and second primary tumors in resected stage I lung cancer. J Thorac Cardiovasc Surg 109(1):120–129 Varlotto JM, Recht A, Flickinger JC, Medford-Davis LN, Dyer AM, Decamp MM, 2009, Factors associated with local and distant recurrence and survival in patients with resected nonsmall cell lung cancer. Cancer 115(5):1059–1069 Varlotto J, Medford-Davis LN, Recht A, Flickinger J, Schaefer E, Shelkey J, et al., 2012, Confirmation of the role of diabetes in the local recurrence of surgically resected non-small cell lung cancer. Lung Cancer 75(3):381–390 Polyak K, Weinberg RA, 2009, Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer 9(4):265–273 Kalluri R, Weinberg RA, 2009, The basics of epithelialmesenchymal transition. J Clin Invest 119(6):1420–1428 Hills CE, Squires PE, 2010, TGF-beta1-induced epithelial-to-mesenchymal transition and therapeutic intervention in diabetic nephropathy. Am J Nephrol 31(1):68–74 Chien J, Staub J, SI H, Erickson-Johnson MR, Couch FJ, Smith DI, et al., 2004, A candidate tumor suppressor HtrA1 is downregulated in ovarian cancer. Oncogene 23(8):1636–1644 Baldi A, De LA, Morini M, Battista T, Felsani A, Baldi F, et al., 2002, The HtrA1 serine protease is down-regulated during human melanoma progression and represses growth of metastatic melanoma cells. Oncogene 21(43):6684–6688 Boyd JA, Hubbs JL, Kim DW, Hollis D, Marks LB, Kelsey CR, 2010, Timing of local and distant failure in resected lung cancer: implications for reported rates of local failure. J Thorac Oncol 5(2): 211–214 Brundage MD, Davies D, Mackillop WJ, 2002, Prognostic factors in non-small cell lung cancer: a decade of progress. Chest 122(3): 1037–1057 Pollack JR, 2007, A perspective on DNA microarrays in pathology research and practice. Am J Pathol 171(2):375–385 Neal JW, Gainor JF, Shaw AT, 2015, Developing biomarkerspecific end points in lung cancer clinical trials. Nat Rev Clin Oncol 12(3):135–146 Mantovani A, Allavena P, Sica A, Balkwill F, 2008, Cancer-related inflammation. Nature 454(7203):436–444 Vigneri P, Frasca F, Sciacca L, Pandini G, Vigneri R, 2009, Diabetes and cancer. Endocr Relat Cancer 16(4):1103–1123 Eliasz S, Liang S, Chen Y, De Marco MA, Machek O, Skucha S, et al., 2010, Notch-1 stimulates survival of lung adenocarcinoma cells during hypoxia by activating the IGF-1R pathway. Oncogene 29(17):2488–2498 Hills CE, Siamantouras E, Smith SW, Cockwell P, Liu KK, Squires PE, 2012, TGFbeta modulates cell-to-cell communication in early epithelial-to-mesenchymal transition. Diabetologia 55(3):812–824 Nerlich AG, Hagedorn HG, Boheim M, Schleicher ED, 1998, Patients with diabetes-induced microangiopathy show a reduced frequency of carcinomas. In Vivo 12(6):667–670 Fuxe J, Vincent T, Garcia d HA, 2010, Transcriptional crosstalk between TGF-beta and stem cell pathways in tumor cell invasion: role of EMT promoting Smad complexes. Cell Cycle 9(12):2363– 2374 Wendt MK, Allington TM, Schiemann WP, 2009, Mechanisms of the epithelial-mesenchymal transition by TGF-beta. Future Oncol 5(8):1145–1168 Micalizzi DS, Farabaugh SM, Ford HL, 2010, Epithelialmesenchymal transition in cancer: parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia 15(2):117–134 Pallen MJ, Wren BW, 1997, The HtrA family of serine proteases. Mol Microbiol 26(2):209–221 Wang N, Eckert KA, Zomorrodi AR, Xin P, Pan W, Shearer DA et al., 2012, Down-regulation of HtrA1 activates the epithelialmesenchymal transition and ATM DNA damage response pathways. PLoS One 7(6):e39446. Schmalhofer O, Brabletz S, Brabletz T, 2009, E-cadherin, betacatenin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev 28(1–2):151–166 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 73 EP 77 DI 10.1007/s12253-016-0086-1 PG 5 ER PT J AU Thakral, G Wey, A Rahman, M Fang, R Lum, Ch AF Thakral, Gaurav Wey, Andrew Rahman, Mobeen Fang, Rui Lum, Christopher TI Agreement of Different Methods for Tissue Based Detection of HER2 Signal in Invasive Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Fish; Altman bland; HER2 Recptor; Breast neoplasms/diagnosis; Breast neoplasms/treatment ID Fish; Altman bland; HER2 Recptor; Breast neoplasms/diagnosis; Breast neoplasms/treatment AB Breast cancer is the second leading cause of cancer mortality amongst American women. The HER2 gene encodes a cell surface receptor that affects cell proliferation and has been recognized as a diagnostic factor in treatment selection for invasive breast cancer. Examine accuracy in HER2 detection between manual count, computer assisted, and automated tiling algorithm. 42 randomly selected invasive breast cancer specimens were enumerated by fluorescence in situ hybridization (FISH)for HER2 and CEP17 markers using the Vysis HER2 assay (AbbotLaboratory, North Chicago, IL). Specimens were tested using three methods: Manual, computer assisted nuclei selection (Tissue FISH MetaSystems, Newton, MA), and automated enumeration (MetaSystems, Newton, MA). The greatest bias and widest agreement limits for HER2 and CEP17 were seen in Automatic versus Manual, the gold standard. HER2 values greater than 6 possessed the greatest bias and widest agreement limits. CEP17 comparison showed similar bias and agreement limits for each comparison. Kappa values indicated good agreement for all methods although Tissue FISH and Manual possessed better agreement. Higher agreement at lower HER2 & CEP17 count maybe due to fewer chromosomal aberrations, in which selection of field of views has less variation between methods. Alternatively, increased background signals seen in polyploidy may be responsible for the variations in signal count. Manual and Tissue FISH demonstrated good agreement amongst by both Altman Bland and Cohen’s Kappa. While the automatic method has good agreement at lower HER2, the sharp increase in variability at higher HER2 counts illustrates a limitation of the automatic method. C1 [Thakral, Gaurav] University of Hawaii and Kuakini Medical CenterHonolulu, HI, USA. [Wey, Andrew] University of Hawaii and Kuakini Medical CenterHonolulu, HI, USA. [Rahman, Mobeen] University of Hawaii and Kuakini Medical CenterHonolulu, HI, USA. [Fang, Rui] University of Hawaii and Kuakini Medical CenterHonolulu, HI, USA. [Lum, Christopher] University of Hawaii and Kuakini Medical CenterHonolulu, HI, USA. RP Thakral, G (reprint author), University of Hawaii and Kuakini Medical Center, Honolulu, USA. EM gthakral@hawaii.edu CR Humphrey LL, Helfand M, Chan BK, Woolf SH, 2002, Breast cancer screening: a summary of the evidence for the U.S. Preventive Services Task Force. Ann Intern Med 137(5 Part 1): 347–360 Heim S, Mitelman F., 2011, Cancer cytogenetics: chromosomal and molecular genetic abberations of tumor cells: John Wiley & Sons Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, et al., 2013, Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol Off J Am Soc Clin Oncol 31(31):3997–4013., DOI 10.1200/JCO.2013.50.9984 Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, et al., 2005, Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med 353(16): 1659–1672 Hanna WM, Ruschoff J, Bilous M, Coudry RA, Dowsett M, Osamura RY, et al., 2014, HER2 in situ hybridization in breast cancer: clinical implications of polysomy 17 and genetic heterogeneity. Mod Pathol: an official journal of the United States and Canadian Academy of Pathology, Inc. 27(1):4–18., DOI 10.1038 /modpathol.2013.103 Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL, 1987, Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235(4785):177–182 Hicks DG, Tubbs RR., 2005, Assessment of the HER2 status in breast cancer by fluorescence in situ hybridization: a technical review with interpretive guidelines. Hum Pathol 36(3):250–261., DOI 10.1016/j.humpath.2004.11.010. Furrer D, Jacob S, Caron C, Sanschagrin F, Provencher L, Diorio C., 2013, Validation of a new classifier for the automated analysis of the human epidermal growth factor receptor 2, HER2, gene amplification in breast cancer specimens. Diagn Pathol 8:17., DOI 10.1186 /1746-1596-8-17 Myles P, Cui J. I., 2007, Using the bland–Altman method to measure agreement with repeated measures. Br J Anaesth 99(3):309–311. Bland JM, Altman D., 1986, Statistical methods for assessing agreement between two methods of clinical measurement. Lancet 327(8476):307–310. Viera AJ, Garrett JM., 2005, Understanding interobserver agreement: the kappa statistic. Fam Med 37(5):360–363. Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, et al., 2014, Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. Arch Pathol Lab Med 138(2):241–256., DOI 10.5858/arpa.2013-0953-SA NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 79 EP 84 DI 10.1007/s12253-016-0091-4 PG 6 ER PT J AU Forghanifard, MM Khales, AS Farshchian, M Rad, A Homayouni-Tabrizi, M Abbaszadegan, RM AF Forghanifard, Mahdi Mohammad Khales, Ardalan Sima Farshchian, Moein Rad, Abolfazl Homayouni-Tabrizi, Masoud Abbaszadegan, Reza Mohammad TI Negative Regulatory Role of TWIST1 on SNAIL Gene Expression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Epithelial-mesenchymal transition (EMT); TWIST1; SNAIL; ESCC cell line ID Epithelial-mesenchymal transition (EMT); TWIST1; SNAIL; ESCC cell line AB Epithelial-mesenchymal transition (EMT) is crucial for specific morphogenetic movements during embryonic development as well as pathological processes of tumor cell invasion and metastasis. TWIST and SNAIL play vital roles in both developmental and pathological EMT. Our aim in this study was to investigate the functional correlation between TWIST1 and SNAIL in human ESCC cell line (KYSE-30). The packaging cell line GP293Twas cotransfected with either control retroviral pruf-IRES-GFP plasmid or pruf-IRES-GFPhTWIST1 and pGP plasmid. The KYSE-30 ESCC cells were transduced with produced viral particles and examined with inverted fluorescence microscope. DNA was extracted from transduced KYSE-30 cells and analyzed for copy number of integrated retroviral sequences in the target cell genome. The concentration of retroviral particles was determined by Realtime PCR. After RNA extraction and cDNA synthesis, the mRNA expression of TWIST1 and SNAIL was assessed by comparative real-time PCR amplification. Ectopic expression of TWIST1 in KYSE-30, dramatically reduces SNAIL expression. Retroviral transduction enforced TWIST1 overexpression in GFP-hTWIST1 nearly 9 folds in comparison with GFP control cells, and interestingly, this TWIST1 enforced expression caused a − 7 fold decrease of SNAIL mRNA expression in GFP-hTWIST1 compared to GFP control cells. Inverse correlation of TWIST1 and SNAIL mRNA levels may introduce novel molecular gene expression pathway controlling EMT process during ESCC aggressiveness and tumorigenesis. Consequently, these data extend the spectrum of biological activities of TWIST1 and propose that therapeutic repression of TWIST1 may be an effective strategy to inhibit cancer cell invasion and metastasis. C1 [Forghanifard, Mahdi Mohammad] Sabzevar University of Medical Sciences, Cellular and Molecular Research CenterSabzevar, Iran. [Khales, Ardalan Sima] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, Bu-Ali square, 9196773117 Mashhad, Iran. [Farshchian, Moein] ACECR-Khorasan Razavi Branch, Molecular Medicine Research DepartmentMashhad, Iran. [Rad, Abolfazl] Sabzevar University of Medical Sciences, Cellular and Molecular Research CenterSabzevar, Iran. [Homayouni-Tabrizi, Masoud] Islamic Azad University, Mashhad Branch, Department of Biochemistry and BiophysicsMashhad, Iran. [Abbaszadegan, Reza Mohammad] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, Bu-Ali square, 9196773117 Mashhad, Iran. RP Abbaszadegan, RM (reprint author), Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, 9196773117 Mashhad, Iran. EM abbaszadeganmr@mums.ac.ir CR Gavert N, Ben-Ze'ev A, 2008, Epithelial-mesenchymal transition and the invasive potential of tumors. Trends MolMed 14(5):199–209 Tsuji T, Ibaragi S, Hu GF, 2009, Epithelial-mesenchymal transition and cell cooperativity in metastasis. Cancer Res 69(18):7135–7139 Batlle E et al., 2000, The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol 2(2):84–89 Cano A et al., 2000, The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol 2(2):76–83 Hajra KM, Chen DY, Fearon ER, 2002, The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Res 62(6): 1613–1618 Eger A, A.K., Sonderegger S, et al., 2005, DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene 24:2375–2385 Comijn J, Berx G, A. A e, Vermassen P, 2001, The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell 7:1267–1278 Yang J et al., 2004, Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117(7):927–939 Sosic D, EN O, 2003, A new twist on twist–modulation of theNFkappa B pathway. Cell Cycle 2:76–78 Maestro R et al., 1999, Twist is a potential oncogene that inhibits apoptosis. Genes Dev 13:2207–2217 Connerney J et al., 2008, Twist1 homodimers enhance FGF responsiveness of the cranial sutures and promote suture closure. Dev Biol 318(2):323–334 Firulli AB, Conway SJ, 2008, Phosphoregulation of Twist1 provides a mechanism of cell fate control. Curr Med Chem 15(25): 2641–2647 Connerney J et al., 2006, Twist1 dimer selection regulates cranial suture patterning and fusion. Dev Dyn 235(5):1345–1357 Hong J et al., 2011, Phosphorylation of serine 68 of Twist1 by MAPKs stabilizes Twist1 protein and promotes breast cancer cell invasiveness. Cancer Res 71(11):3980–3990 Castanon I et al., 2001, Dimerization partners determine the activity of the twist bHLH protein during drosophila mesoderm development. Development 128(16):3145–3159 de Herreros AG et al., 2010, Snail family regulation and epithelial mesenchymal transitions in breast cancer progression. J Mammary Gland Biol Neoplasia 15(2):135–147 Peinado H, Olmeda D, CanoA, 2007, Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 7(6):415–428 Yuen HF et al., 2007, Upregulation of twist in oesophageal squamous cell carcinoma is associated with neoplastic transformation and distant metastasis. J Clin Pathol 60(5):510–514 Forghanifard MM et al., 2012, Expression analysis elucidates the roles of MAML1 and Twist1 in esophageal squamous cell carcinoma aggressiveness and metastasis. Ann Surg Oncol 19(3):743–749 Mauhin V et al., 1993, Definition of the DNA-binding site repertoire for the drosophila transcription factor SNAIL. Nucleic Acids Res 21(17):3951–3957 Peinado H et al., 2004, Snail mediates E-cadherin repression by the recruitment of the Sin3A/histone deacetylase 1, HDAC1)/HDAC2 complex. Mol Cell Biol 24(1):306–319 Barbera MJ et al., 2004, Regulation of snail transcription during epithelial to mesenchymal transition of tumor cells. Oncogene 23(44):7345–7354 Peinado H, Quintanilla M, Cano A, 2003, Transforming growth factor beta-1 induces snail transcription factor in epithelial cell lines: mechanisms for epithelial mesenchymal transitions. J Biol Chem 278(23):21113–21123 Peiro S et al., 2006, Snail1 transcriptional repressor binds to its own promoter and controls its expression. Nucleic Acids Res 34(7): 2077–2084 Ardalan Khales S et al., 2015, SALL4 as a new biomarker for early colorectal cancer. J Cancer Res Clin Oncol 141(2):229– 235 Liang CC, Park AY, Guan JL, 2007, In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc 2(2):329–333 Giampieri S et al., 2009, Localized and reversible TGF beta signalling switches breast cancer cells from cohesive to single cell motility. Nat Cell Biol 11(11):1287–1296 Tran DD et al., 2011, Temporal and spatial cooperation of Snail1 and Twist1 during epithelial-mesenchymal transition predicts for human breast cancer recurrence.Mol Cancer Res 9(12):1644–1657 Chambers AF, Groom AC, MacDonald IC, 2002, Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer 2(8): 563–572 Stoletov K et al., 2010, Visualizing extravasation dynamics of metastatic tumor cells. J Cell Sci 123(Pt 13):2332–2341 De Craene B, van Roy F, Berx G, 2005, Unraveling signalling cascades for the snail family of transcription factors. Cell Signal 17(5):535–547 Blanco MJ et al., 2002, Correlation of snail expression with histological grade and lymph node status in breast carcinomas. Oncogene 21(20):3241–3246 Giroldi LA et al., 1997, Role of E boxes in the repression of Ecadherin expression. Biochem Biophys Res Commun 241(2):453– 458 De Craene B et al., 2005, The transcription factor snail induces tumor cell invasion through modulation of the epithelial cell differentiation program. Cancer Res 65:6237–6244 DiDonato JA,Mercurio F,MK, 2012, NF-κB and the link between inflammation and cancer. Immunol Rev 246:379–400 Julien S et al., 2007, Activation of NF-kappaB by Akt upregulates snail expression and induces epithelium mesenchyme transition. Oncogene 26(53):7445–7456 Castanon I, Baylies MK, 2002, A twist in fate: evolutionary comparison of twist structure and function. Gene 287(1–2):11–22 Sosic D et al., 2003, Twist regulates cytokine gene expression through a negative feedback loop that represses NF-kappaB activity. Cell 112(2):169–180 Lander R et al., 2013, Interactions between twist and other core epithelial–mesenchymal transition factors are controlled by GSK3- mediated phosphorylation. Nat Commun 4:1542 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 85 EP 90 DI 10.1007/s12253-016-0093-2 PG 6 ER PT J AU Kumari, S Tewari, Sh Husain, N Agarwal, A Pandey, A Singhal, A Lohani, M AF Kumari, Swati Tewari, Shikha Husain, Nuzhat Agarwal, Akash Pandey, Anshuman Singhal, Ashish Lohani, Mohtashim TI Quantification of Circulating Free DNA as a Diagnostic Marker in Gall Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Circulating free DNA; Gall bladder cancer; Real time PCR (qPCR); DNA quantification ID Circulating free DNA; Gall bladder cancer; Real time PCR (qPCR); DNA quantification AB Gall bladder Carcinoma (GBC) is the fifth most common cancer of the digestive tract and frequently diagnosed in late stage of disease. Estimation of circulating free DNA (cfDNA) in serum has been applied as a "liquid biopsy" in several deep seated malignancies. Its value in diagnosis of gall bladder carcinoma has not been studied. The present study was designed to assess the role of cfDNA in the diagnosis of GBC and correlate levels with the TNM stage. Serum was collected from 34 patients with GBC and 39 age and sex matched controls including 22 cholecystitis and 17 healthy individuals. Serum cfDNA levels were measured through quantitative polymerase chain reaction (qPCR) by amplification of β-globin gene. Performance of the assay was calculated through the receiver operating characteristic (ROC) curve. The cfDNA level was significantly lower in healthy controls and cholecystitis (89.32 ± 59.76 ng/ml, 174.21 ± 99.93 ng/ml) compared to GBC (1245.91 ± 892.46 ng/ml, p = <0.001). The cfDNA level was significantly associated with TNM stage, lymph node involvement and jaundice (0.002, 0.027, and 0.041, respectively). Area under curve of ROC analysis for cancer group versus healthy and cholecystitis group was 1.00 and 0.983 with sensitivity of 100 %, 88.24 % and specificity of 100 % respectively. Quantitative analysis of cfDNA may distinguish cholecystitis and gall bladder carcinoma and may serve as new diagnostic, noninvasive marker adjunct to imaging for the diagnosis of GBC. C1 [Kumari, Swati] Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Pathology, 226010 Lucknow, India. [Tewari, Shikha] Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Pathology, 226010 Lucknow, India. [Husain, Nuzhat] Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Pathology, 226010 Lucknow, India. [Agarwal, Akash] Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Surgical Oncology, 226010 Lucknow, India. [Pandey, Anshuman] Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Gastrosurgery, 226010 Lucknow, India. [Singhal, Ashish] Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Surgical Oncology, 226010 Lucknow, India. [Lohani, Mohtashim] Integral University, Department of Biosciences, 226026 Lucknow, India. RP Husain, N (reprint author), Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Pathology, 226010 Lucknow, India. EM drnuzhathusain@hotmail.com CR Mishra S, Chaturvedi A, Mishra NC, Sharma ID, 2013, Carcinoma of the gallbladder. The Lancet Oncology 4:167–176 Hamdani NS, Qadri SK, Aggarwalla R, Bhartia VK, Chaudhuri S, Debakshi S, Baig SJ, Pal NK, 2013, Clinicopathological Study of Gall Bladder Carcinoma with Special Reference to Gallstones: Our 8-year Experience from Eastern India. Asian Pacific J Cancer Prev 13(11):5613–5617 Goldin RD, Roa JC, 2009, Gallbladder cancer: a morphological and molecular update. Histopathology 55:218–229 Kapoor VK, Michael AJ Mc, 2003, Gallbladder cancer: An ‘Indian’ disease. Natl Med J India 4:16. Peng HH, Zhang YD, Gong LS, Liu WD, Zhang Y, 2013, Increased expression of microRNA-335 predicts a favorable prognosis in primary gallbladder carcinoma. Onco Targets Ther 6: 1625–1630. Liu TY, Tan ZJ, Jiang L, JF G, XSW, Cao Y, LiML, KJW, Liu YB, 2013, Curcumin induces apoptosis in gallbladder carcinoma cell line GBC-SD cells. Cancer Cell Int 13:6 Stroun M, Maurice P, Vasioukhin V, Lyautey J, Lederry C, Lefort F, 2000, The origin and mechanism of circulating DNA. Ann N Y Acad Sci 906:161–168 Ellinger J, Bastian PJ, Haan KI, Heukamp LC, Buettner R, Fimmers R, Mueller SC, Von Ruecker A, 2008, Noncancerous PTGS2 DNA fragments of apoptotic origin in sera of prostate cancer patients qualify as diagnostic and prognostic indicators. Int J Cancer 122(1):138–143 Sozzi G, Conte D, Mariani L, 2001, Analysis of circulating tumor DNA in plasma at diagnosis and during follow-up of lung cancer patients. Cancer Res 61:4675–4678 Umetani N, Kim J, Hiramatsu S, 2006, Increased integrity of free circulating DNA in sera of patients with colorectal or periampullary cancer: direct quantitative PCR for ALU repeats. Clin Chem 52: 1062–1069. Trejo-Becerril C, Perez-Cardenas E, Trevino-Cuevas H, 2003, Circulating nucleosomes and response to chemotherapy: an in vitro, in vivo and clinical study on cervical cancer patients. Int J Cancer 104:663–668 Chang HW, Lee SM, Goodman SN, 2002, Assessment of plasma DNA levels, allelic imbalance, and CA 125 as diagnostic tests for cancer. J Natl. Cancer Inst 94:1697–1703 Gal S, Fidler C, Lo YMD, Taylor M, Han C, Moore J, Harris AL, Wainscoat JS, 2004, Quantitation of circulating DNA in the serum of breast cancer patients by real-time PCR. Br J Cancer 90:1211–1215 Ellinger J,Wittkamp V, Albers P, 2009, Cell-free circulating DNA: diagnostic value in patients with testicular germ cell cancer. J Urol 181:363–371 Ellinger J, Bastian PJ, Ellinger N, 2008, Apoptotic DNA fragments in serum of patients with muscle invasive bladder cancer: a prognostic entity. Cancer Lett 264:274–280 Altimari A, Grigioni AD, Benedettini E, 2008, Diagnostic role of circulating free plasma DNA detection in patients with localized prostate cancer. Am. J Clin Pathol 129:756–762 Sai S, Ichikawa D, Tomita H, Ikoma D, Tani N, Ikoma H, Kikuchi S, Fujiwara H, Ueda Y, Otsuji E, 2007, Quantification of Plasma Cell-free DNA in Patients with Gastric Cancer. Anticancer Res 27: 2747–2752 Chen K, Zhang H, Zhang L, SQ J, Qi J, Huang DF, Li F, Wei Q, Jing Zhang J, 2013, Value of circulating cell-free DNA in diagnosis of hepatocellular carcinoma. World J Gastroenterol 19(20):3143–3149 Lo YMD, Rainer TH, Chan LYS, Hjelm NM, Cocks RA, 2000, Plasma DNA as a Prognostic Marker in Trauma Patients. Clin Chem 46(3):319–323 Ghosh M, Sakhuja P, Singh S, Agarwal AK, 2013, p53 and beta catenin expression in gallbladder tissues and correlation with tumor progression in gallbladder cancer. Saudi J Gastroenterol 19:34–39 He CZ, Zhang KH, Li Q, Liu XH, Hong Y, Lv NH, 2013, Combined use of AFP, CEA, CA125 and CAl9–9 improves the sensitivity for the diagnosis of gastric cancer. BMC Gastroenterol 13:87. Zhang D, Yu M, Xu T, Xiong B, 2013, Predictive value of serum CEA, CA19–9 and CA125 in diagnosis of colorectal liver metastasis in Chinese population. Hepato-Gastroenterology 60:1297–1301 Nath A, Kumar S, SinghM, Nadeem T, Murti K, Gupta AK, Das P, Kumar A, 2015, Combined assessment of lipid peroxidation, Cea and Ca19.9 biomarkers in advanced stage gall bladder cancer. Int J Recent Sci Res 6::3136–3314 Leon SA, Shapiro B, Sklaroff DM, Yaros MJ, 1977, Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 37:646–650 Umetani N, Hiramatsu S, Hoon DSB, 2006, Higher amount of free circulating DNA in serum than in plasma is not mainly caused by contaminated extraneous DNA during separation. Annals NYAcad Sci 1075:299–307 Gormally E, Caboux E, Vineis P, Hainaut P, 2007, Circulating free DNA in plasma or serum as biomarker of carcinogenesis: Practical aspects and biological significance. Mutat Res 635:105–117 Pinzani P, Salvianti F, Pazzagli M, Orlando C, 2010, Circulating nucleic acids in cancer and pregnancy. Methods 50:302–307 Swarup V, Rajeswari MR, 2007, Circulating, cell-free, nucleic acids – a promising, Non-invasive tool for early detection of several human diseases. FEBS Lett 581:795–799 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 91 EP 97 DI 10.1007/s12253-016-0087-0 PG 7 ER PT J AU Groeger, S Howaldt, HP Raifer, H Gattenloehner, S Chakraborty, T Meyle, J AF Groeger, Sabine Howaldt, Hans-Peter Raifer, Hartmann Gattenloehner, Stefan Chakraborty, Trinad Meyle, Joerg TI Oral Squamous Carcinoma Cells Express B7-H1 and B7-DC Receptors in Vivo SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral squamous cell carcinomas; B7-H1; B7-Dc; In vivo; Immune evasion ID Oral squamous cell carcinomas; B7-H1; B7-Dc; In vivo; Immune evasion AB B7-H1 and B7-DC ligands are members of the B7 family with important regulatory functions in cell-mediated immune response. Both receptors are ligands of the programmed death receptor PD-1. B7-H1 expression has been detected in the majority of human carcinomas in vivo. B7- H1 mediated signals are able to negatively regulate activated T cell functions and survival, and enable tumor cells to overcome host response. The aim of this study was to investigate the expression of B7-H1 and B7-DC proteins in oral squamous cell carcinomas (OSCC) in vivo. Tissues from 15 samples were cryo-sected and following histological routine staining (HE), incubated with antibodies against human B7-H1 and B7-DC. Immuno-staining of pan-cytokeratin was performed to ascertain the epithelial origin of the tissue and CK 19 to demonstrate the proliferating stage. Confocal laser scanning microscopy confirmed the presence of both B7-H1 and B7- DC in all 15 OSCC. The B7-H1 and B7-DC staining was located in areas of the tissue that were identified as cancerous lesions in the previously stained HE sections before. Staining with Pan-CK and CK19 provided evidence for the epithelial origin and the proliferating stage of the tissue. The in vivo expression of the B7-H1 and B7-DC receptors in oral squamous cell carcinomas suggest that general mechanisms for immune evasion of tumors are also found in OSCC. C1 [Groeger, Sabine] Justus-Liebig-University, Department of Periodontology, Schlangenzahl 14, 35392 Giessen, Germany. [Howaldt, Hans-Peter] Justus-Liebig-University, Oral and Maxillofacial SurgeryGiessen, Germany. [Raifer, Hartmann] Phillips University, Institute for Medical Microbiology and HygieneMarburg, Germany. [Gattenloehner, Stefan] Justus-Liebig-University, Institute for PathologyGiessen, Germany. [Chakraborty, Trinad] Justus-Liebig-University, Institute for Medical MicrobiologyGiessen, Germany. [Meyle, Joerg] Justus-Liebig-University, Department of Periodontology, Schlangenzahl 14, 35392 Giessen, Germany. RP Groeger, S (reprint author), Justus-Liebig-University, Department of Periodontology, 35392 Giessen, Germany. EM Sabine.E.Groeger@dentist.med.uni-giessen.de CR Dong H, Zhu G, Tamada K, Chen L, 1999, B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin- 10 secretion. Nat Med 5(12):1365–1369 Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, Bowman MR, Carreno BM, Collins M, Wood CR, Honjo T, 2000, Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 192(7): 1027–1034 Chen Y, Zhang J, Guo G, Ruan Z, Jiang M, Wu S, Guo S, Fei L, Tang Y, Yang C, Jia Z,Wu Y, 2009, Induced B7-H1 expression on human renal tubular epithelial cells by the sublytic terminal complement complex C5b-9. Mol Immunol 46(3):375–383 LaGier AJ, Pober JS, 2006, Immune accessory functions of human endothelial cells are modulated by overexpression of B7-H1, PDL1. Hum Immunol 67(8):568–578 Yamazaki T, Akiba H, Iwai H, Matsuda H, AokiM, Tanno Y, Shin T, Tsuchiya H, Pardoll DM, Okumura K, Azuma M, Yagita H, 2002, Expression of programmed death 1 ligands by murine T cells and APC. J Immunol 169(10):5538–5545 IshidaY,AgataY, ShibaharaK, Honjo T, 1992, Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. Embo J 11(11):3887–3895 Wang S, Chen L, 2004, T lymphocyte co-signaling pathways of the B7-CD28 family. Cell Mol Immunol 1(1):37–42 Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L, 2002, Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 8(8):793–800 Subudhi SK, Zhou P, Yerian LM, Chin RK, Lo JC, Anders RA, Sun Y, Chen L,Wang Y, Alegre ML, Fu YX, 2004, Local expression of B7-H1 promotes organ-specific autoimmunity and transplant rejection. J Clin Invest 113(5):694–700 Gyrd-Hansen M, Meier P, 2010, IAPs: from caspase inhibitors to modulators of NF-kappaB, inflammation and cancer. Nature reviews. Cancer 10(8):561–574 Uppaluri R, Dunn GP, Lewis JS Jr, 2008, Focus on TILs: prognostic significance of tumor infiltrating lymphocytes in head and neck cancers. Cancer Immunity 8:16 Okazaki T, Honjo T, 2006, The PD-1-PD-L pathway in immunological tolerance. Trends Immunol 27(4):195–201 Tsushima F, Yao S, Shin T, Flies A, Flies S, Xu H, Tamada K, Pardoll DM, Chen L, 2007, Interaction between B7-H1 and PD-1 determines initiation and reversal of T-cell anergy. Blood 110(1): 180–185 Azuma T, Yao S, Zhu G, Flies AS, Flies SJ, Chen L, 2008, B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells. Blood 111(7): 3635–3643 Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G, 2013, Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res 73(12):3591–3603 Wang W, Lau R, Yu D, Zhu W, Korman A, Weber J, 2009, PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi, regulatory T cells. Int Immunol 21(9): 1065–1077 Zandberg DP, Strome SE, 2014, The role of the PD-L1:PD-1 pathway in squamous cell carcinoma of the head and neck. Oral Oncol 50(7):627–632 Ritprajak P, Azuma M, 2015, Intrinsic and extrinsic control of expression of the immunoregulatory molecule PD-L1 in epithelial cells and squamous cell carcinoma. Oral Oncol 51(3):221–228 Strome SE, Dong H, Tamura H, Voss SG, Flies DB, Tamada K, Salomao D, Cheville J, Hirano F, Lin W, Kasperbauer JL, Ballman KV, Chen L, 2003, B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. Cancer Res 63(19): 6501–6505 Tsushima F, Tanaka K, Otsuki N, Youngnak P, Iwai H, Omura K, Azuma M, 2006, Predominant expression of B7-H1 and its immunoregulatory roles in oral squamous cell carcinoma. Oral Oncol 42(3):268–274 Ohigashi Y, Sho M, Yamada Y, Tsurui Y, Hamada K, Ikeda N, Mizuno T, Yoriki R, Kashizuka H, Yane K, Tsushima F, Otsuki N, Yagita H, Azuma M, Nakajima Y, 2005, Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clinical cancer research : an official journal of theAmerican Association for Cancer Research 11(8):2947–2953 Vakkila J, Lotze MT, 2004, Inflammation and necrosis promote tumour growth. Nature reviews. Immunology 4(8):641–648 Ghebeh H, Mohammed S, Al-Omair A, Qattan A, Lehe C, Al- Qudaihi G, Elkum N, Alshabanah M, Bin Amer S, Tulbah A, Ajarim D, Al-Tweigeri T, Dermime S, 2006, The B7-H1, PD-L1, T lymphocyte-inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: correlation with important high-risk prognostic factors. Neoplasia 8(3):190–198 Thompson RH, Kwon ED, 2006, Significance of B7-H1 overexpression in kidney cancer. Clin Genitourin Cancer 5(3):206–211 Wintterle S, Schreiner B, Mitsdoerffer M, Schneider D, Chen L, Meyermann R, Weller M, Wiendl H, 2003, Expression of the B7- related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. Cancer Res 63(21):7462–7467 CPW, Jiang JT, Tan M, Zhu YB, Ji M, KF X, Zhao JM, Zhang GB, Zhang XG, 2006, Relationship between co-stimulatory molecule B7-H3 expression and gastric carcinoma histology and prognosis. World J Gastroenterol 12(3):457–459 Gupta PC, Murti PR, Bhonsle RB, 1996, Epidemiology of cancer by tobacco products and the significance of TSNA. Crit Rev Toxicol 26(2):183–198 Moreno-Lopez LA, Esparza-Gomez GC, Gonzalez-Navarro A, Cerero-Lapiedra R, Gonzalez-Hernandez MJ, Dominguez-Rojas V, 2000, Risk of oral cancer associated with tobacco smoking, alcohol consumption and oral hygiene: a case-control study in Madrid, Spain. Oral Oncol 36(2):170–174 Reichart PA, 2000, Oral mucosal lesions in a representative crosssectional study of aging Germans. Community Dent Oral Epidemiol 28(5):390–398 Zheng TZ, Boyle P, HF H, Duan J, Jian PJ, Ma DQ, Shui LP, Niu SR, Scully C, MacMahon B, 1990, Dentition, oral hygiene, and risk of oral cancer: a case-control study in Beijing, People's Republic of China. Cancer Causes Control 1(3):235–241 Youngnak-Piboonratanakit P, Tsushima F, Otsuki N, Igarashi H, Machida U, Iwai H, Takahashi Y, Omura K, Yokozeki H, Azuma M, 2004, The expression of B7-H1 on keratinocytes in chronic inflammatory mucocutaneous disease and its regulatory role. Immunology letters 94(3):215–222 Okudaira K, Hokari R, Tsuzuki Y, Okada Y, Komoto S, Watanabe C, Kurihara C, Kawaguchi A, Nagao S, Azuma M, Yagita H, Miura S, 2009, Blockade of B7-H1 or B7-DC induces an anti-tumor effect in a mouse pancreatic cancer model. Int J Oncol 35(4):741–749 Das S, Suarez G, Beswick EJ, Sierra JC, Graham DY, Reyes VE, 2006, Expression of B7-H1 on gastric epithelial cells: its potential role in regulating T cells during helicobacter pylori infection. J Immunol 176(5):3000–3009 Tezal M, Grossi SG, Genco RJ, 2005, Is periodontitis associated with oral neoplasms? J Periodontol 76(3):406–410 Tezal M, Sullivan MA, Reid ME, Marshall JR, Hyland A, Loree T, Lillis C, Hauck L, Wactawski-Wende J, Scannapieco FA, 2007, Chronic periodontitis and the risk of tongue cancer. Arch Otolaryngol Head Neck Surg 133(5):450–454 (UICC, IUaC, 2011, The International Union Against Cancer classification for malignant tumours. 7th edition) Algarra I, Collado A, Garrido F, 1997, Altered MHC class I antigens in tumors. Int J Clin Lab Res 27(2):95–102 Johnsen AK, France J, Nagy N, Askew D, Abdul-Karim FW, Gerson SL, Sy MS, Harding CV, 2001, Systemic deficits in transporter for antigen presentation, TAP)-1 or proteasome subunit LMP2 have little or no effect on tumor incidence. International journal of cancer Journal international du cancer 91(3):366–372 Zheng P, Sarma S, Guo Y, Liu Y, 1999, Twomechanisms for tumor evasion of preexisting cytotoxic T-cell responses: lessons from recurrent tumors. Cancer Res 59(14):3461–3467 de Vries TJ, Trancikova D, Ruiter DJ, van Muijen GN, 1998, High expression of immunotherapy candidate proteins gp100, MART-1, tyrosinase and TRP-1 in uveal melanoma. Br J Cancer 78(9):1156– 1161 Hofbauer GF, Kamarashev J, Geertsen R, Boni R, Dummer R, 1998, Melan A/MART-1 immunoreactivity in formalin-fixed paraffin- embedded primary and metastatic melanoma: frequency and distribution. Melanoma Res 8(4):337–343 DavidsonWF, Giese T, Fredrickson TN, 1998, Spontaneous development of plasmacytoid tumors in mice with defective Fas-Fas ligand interactions. J Exp Med 187(11):1825–1838 IrmlerM, ThomeM, HahneM, Schneider P, Hofmann K, SteinerV, Bodmer JL, SchroterM, Burns K, Mattmann C, Rimoldi D, French LE, Tschopp J, 1997, Inhibition of death receptor signals by cellular FLIP. Nature 388(6638):190–195 Takeda K, Smyth MJ, Cretney E, Hayakawa Y, Yamaguchi N, Yagita H, Okumura K, 2001, Involvement of tumor necrosis factor-related apoptosis-inducing ligand in NK cell-mediated and IFN-gamma-dependent suppression of subcutaneous tumor growth. Cell Immunol 214(2):194–200 TaylorMW, Feng GS, 1991, Relationship between interferon-gamma, indoleamine 2,3-dioxygenase, and tryptophan catabolism. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 5(11):2516–2522 Schwartz RH, 1990, A cell culture model for T lymphocyte clonal anergy. Sci, New York, NY 248(4961):1349–1356 Oyama T, Ran S, Ishida T, Nadaf S, Kerr L, Carbone DP, Gabrilovich DI, 1998, Vascular endothelial growth factor affects dendritic cell maturation through the inhibition of nuclear factorkappa B activation in hemopoietic progenitor cells. J Immunol 160(3):1224–1232 Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM, 2001, Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol 166(9):5398–5406 Kusmartsev SA, Li Y, Chen SH, 2000, Gr-1+ myeloid cells derived from tumor-bearing mice inhibit primary T cell activation induced through CD3/CD28 costimulation. J Immunol 165(2):779–785 Sutmuller RP, van Duivenvoorde LM, van Elsas A, Schumacher TN, Wildenberg ME, Allison JP, Toes RE, Offringa R, Melief CJ, 2001, Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+, regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J Exp Med 194(6):823–832 Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T, 2001, PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad SciUSA98(24):13866– 13871 Eppihimer MJ, Gunn J, Freeman GJ, Greenfield EA, Chernova T, Erickson J, Leonard JP, 2002, Expression and regulation of the PDL1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation 9(2):133–145 Mazanet MM, Hughes CC, 2002, B7-H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis. J Immunol 169(7):3581–3588 Petroff MG, Chen L, Phillips TA, Hunt JS, 2002, B7 family molecules: novel immunomodulators at the maternal-fetal interface. Placenta 23(Suppl A):S95–101 Wiendl H, Mitsdoerffer M, Schneider D, Chen L, Lochmuller H, Melms A, Weller M, 2003, Human muscle cells express a B7- related molecule, B7-H1, with strong negative immune regulatory potential: a novel mechanism of counterbalancing the immune attack in idiopathic inflammatory myopathies. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 17(13):1892–1894 Nakanishi J,Wada Y, Matsumoto K, Azuma M, Kikuchi K, Ueda S, 2007, Overexpression of B7-H1, PD-L1, significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer immunology, immunotherapy : CII 56(8):1173– 1182 Thompson RH, Dong H, Kwon ED, 2007, Implications of B7-H1 expression in clear cell carcinoma of the kidney for prognostication and therapy. Clinical cancer research : an official journal of the American Association for Cancer Research 13(2 Pt 2):709 s–715 s Thompson RH, Webster WS, Cheville JC, Lohse CM, Dong H, Leibovich BC, Kuntz SM, Sengupta S, Kwon ED, Blute ML, 2005, B7-H1 glycoprotein blockade: a novel strategy to enhance immunotherapy in patients with renal cell carcinoma. Urology 66(5 Suppl):10–14 Wang L, Ma Q, Chen X, Guo K, Li J, Zhang M, 2010, Clinical significance of B7-H1 and B7–1 expressions in pancreatic carcinoma. World J Surg 34(5):1059–1065 Konishi J, Yamazaki K, Azuma M, Kinoshita I, Dosaka-Akita H, NishimuraM(2004, B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression. Clinical cancer research : an official journal of the American Association for Cancer Research 10(15): 5094–5100 Seiwert TYBB, Weiss J, 2014, A phase Ib study of MK-3475 in patients with human papillomavirus, HPV)-associated and non- HPV–associated head and neck, H/N, cancer. J Clin Oncol 32(suppl 5S):abstr 6011 Groeger S, Domann E, Gonzales JR, Chakraborty T, Meyle J, 2011, B7-H1 and B7-DC receptors of oral squamous carcinoma cells are upregulated by Porphyromonas gingivalis. Immunobiology 216(12):1302–1310 Itoh M, Takahashi T, Sakaguchi N, Kuniyasu Y, Shimizu J, Otsuka F, Sakaguchi S, 1999, Thymus and autoimmunity: production of CD25 + CD4+ naturally anergic and suppressive T cells as a key function of the thymus in maintaining immunologic self-tolerance. J Immunol 162(9):5317–5326 Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M, 1995, Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains, CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155(3):1151–1164 Takahashi T, Kuniyasu Y, Toda M, Sakaguchi N, Itoh M, Iwata M, Shimizu J, Sakaguchi S, 1998, Immunologic self-tolerance maintained by CD25 + CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease by breaking their anergic/ suppressive state. Int Immunol 10(12):1969–1980 Kronenberg M, Rudensky A, 2005, Regulation of immunity by self-reactive T cells. Nature 435(7042):598–604 Sakaguchi S,Yamaguchi T, Nomura T, OnoM(2008, Regulatory T cells and immune tolerance. Cell 133(5):775–787 Tang Q, Bluestone JA, 2008, The Foxp3+ regulatory T cell: a jack of all trades, master of regulation. Nat Immunol 9(3):239–244 AusielloCM, Palma C, Maleci A, Spagnoli GC,Amici C, Antonelli G, Casciani CU, Cassone A, 1991, Cell mediated cytotoxicity and cytokine production in peripheral blood mononuclear cells of glioma patients. Eur J Cancer 27(5):646–650 Roussel E, Gingras MC, Grimm EA, Bruner JM, Moser RP, 1996, Predominance of a type 2 intratumoural immune response in fresh tumour-infiltrating lymphocytes from human gliomas. Clin Exp Immunol 105(2):344–352 Zou JP, Morford LA, Chougnet C, Dix AR, Brooks AG, Torres N, Shuman JD, Coligan JE, Brooks WH, Roszman TL, Shearer GM, 1999, Human glioma-induced immunosuppression involves soluble factor(s, that alters monocyte cytokine profile and surface markers. J Immunol 162(8):4882– 4892 Gasparoto TH, de Souza Malaspina TS, Benevides L, de Melo EJ, Jr., Costa MR, Damante JH, Ikoma MR, Garlet GP, Cavassani KA, da Silva JS, Campanelli AP, 2010, Patients with oral squamous cell carcinoma are characterized by increased frequency of suppressive regulatory T cells in the blood and tumor microenvironment. Cancer immunology, immunotherapy : CII 59(6):819–828 Schwarz, S, Butz, M, Morsczeck, C, Reichert, TE, Driemel, O, 2008, Increased number of CD25 FoxP3 regulatory T cells in oral squamous cell carcinomas detected by chromogenic immunohistochemical double staining. Journal of oral pathology & medicine : official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology 37(8): 485–489 Al-Qahtani, D, Anil, S, Rajendran, R, 2011, Tumour infiltrating CD25+ FoxP3+ regulatory T cells, Tregs, relate to tumour grade and stromal inflammation in oral squamous cell carcinoma. Journal of oral pathology & medicine: official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology 40(8):636–642 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 99 EP 110 DI 10.1007/s12253-016-0100-7 PG 12 ER PT J AU Shimizu, A Kaira, K Yasuda, M Asao, T Ishikawa, O AF Shimizu, Akira Kaira, Kyoichi Yasuda, Masahito Asao, Takayuki Ishikawa, Osamu TI Clinical and Pathological Significance of ER Stress Marker (BiP/GRP78 and PERK) Expression in Malignant Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ER stress; BiP/GRP78; PERK; Prognosis; Malignant melanoma; Skin cancer; Immunohistochemistry ID ER stress; BiP/GRP78; PERK; Prognosis; Malignant melanoma; Skin cancer; Immunohistochemistry AB Glucose-regulated protein of 78 kD (GRP78) also referred to as immunoglobulin heavy chain binding protein (BiP/GRP78) plays an important role in the endoplasmic reticulum (ER) stress. The level of BiP/GRP78 is highly elevated in various human cancers. The purpose of this study is to examine the prognostic significance of BiP/GRP78 expression in patients with malignant melanoma. A total of 133 malignant melanoma patients were analyzed, and tumor specimens were stained by immunohistochemistry for BiP/ GRP78, PKR-like endoplasmic reticulum kinase (PERK), Ki-67, p53 and microvessel density (MVD) determined by CD34. BiP/GRP78 and PERK were highly expressed in 40 % (53/133) and 78 % (104/133), respectively. BiP/ GRP78 disclosed a significant relationship with PERK expression, thickness, T factor, N factor, disease staging, cell proliferation (Ki-67) and MVD (CD34). By multivariate analysis, the high expression of BiP/GRP78 was identified as an independent prognostic factor for predicting poor survival against malignant melanoma. The increased BiP/GRP78 expression was clarified as an independent prognostic marker for predicting worse outcome. ER stress marker, BiP/GRP78 could be a powerful molecular target for the treatment of malignant melanoma. C1 [Shimizu, Akira] Gunma University, Graduate School of Medicine, Department of Dermatology, 3-39-22 Showa-machi, Maebashi, 371-8511 Gunma, Japan. [Kaira, Kyoichi] Gunma University, Graduate School of Medicine, Department of Oncology Clinical Development, Showa-machi, Maebashi, 371-8511 Gunma, Japan. [Yasuda, Masahito] Gunma University, Graduate School of Medicine, Department of Dermatology, 3-39-22 Showa-machi, Maebashi, 371-8511 Gunma, Japan. [Asao, Takayuki] Gunma University, Graduate School of Medicine, Department of Oncology Clinical Development, Showa-machi, Maebashi, 371-8511 Gunma, Japan. [Ishikawa, Osamu] Gunma University, Graduate School of Medicine, Department of Dermatology, 3-39-22 Showa-machi, Maebashi, 371-8511 Gunma, Japan. RP Shimizu, A (reprint author), Gunma University, Graduate School of Medicine, Department of Dermatology, 371-8511 Gunma, Japan. EM shimizuakira@gunma-u.ac.jp CR Hawryluk EB, Tsao H, 2014, Melanoma clinical features and genomic insights. Cold Spring Harb Perspect Med 4:a015388 PayetteMJ, KatzMIII, Grant-Kels JM, 2009, Melanoma prognostic factors found in the dermatopathology report. Clin Dermatol 27: 53–74 Hendershot LM, 2004, The ER function BiP is a master regulator of ER function. Mt Sinai J Med 71:289–297 Gazit G, Lu J, Lee AS, 1999, De-regulation of GRP stress protein expression in human breast cancer cell lines. Breast Cancer Res Treat 54:135–146 Li J, Lee AS, 2006, Stress induction of GRP78/BiP and its role in cancer. Curr Mol Med 6:45–54 Zhuang L, Scolyer RA, Lee CS,McCarthy SW, CooperWA, Zhang XD, et al., 2009, Expression of glucose-regulated stress protein GRP78 is related to progression of melanoma. Histopathology 54: 462–470 Papalas JA, Vollmer RT, Gonzalez-Gronow M, Pizzo SV, Burchette J, Youens KE, et al., 2010, Patterns of GRP78 and MTJ1 expression in primary cutaneous malignant melanoma. Mod Pathol 23: 134–143 Dong D, Stapleton C, Luo B, Xiong S, YeW, Zhang Y, et al., 2011, A critical role for GRP78/BiP in the tumor microenviroment for neovascularization during tumor growth and metastasis. Cancer Res 71:2848–2857 Croft A, Tay KH, Boyd SC, Guo ST, Jiang CC, Lai F, et al., 2014, Oncogenic activation of MEK/ERK primes melanoma cells for adaptation to endoplasmic reticulum stress. J Invest Dermatol 134:488–497 Vandewynckel YP, Laukens D, Bogaerts E, Paridaens A, Van den Bussche A, Verhelst X, et al., 2014, Modulation of the unfolded protein response impedes tumor cell adaptation to proteotoxic stress: a PERK for hepatocellular carcinoma therapy. Hepatol Int 9:93–104 Huang TT, Chen JY, Tseng CE, YC S, Ho HC, Lee MS, et al., 2010, Decreased GRP78 protein expression is a potential prognostic marker of oral squamous cell carcinoma in Taiwan. J Formos Med Assoc 109:326–337 Shimizu A, Kaira K, Kato M, Yasuda M, Takahashi A, Tominaga H, et al., 2015, Prognostic significance of L-type amino acid transporter 1, LAT1, expression in cutaneousmelanoma. Melanoma Res 25:399–405 Kaira K, EndoM, Abe M, Nakagawa K, Ohde Y, Okumura T, et al., 2010, Biologic correlation of 2-[18F]-fluoro-2-deoxy-D-glucose uptake on positron emission tomography in thymic epithelial tumors. J Clin Oncol 28:3746–3753 Ramsay RG, Ciznadija D, Mantamadiotis T, Anderson R, Pearson R, 2005, Expression of stress response protein glucose regulated protein-78 mediated by c-Myb. Int J Biochem Cell Biol 37:1254– 1268 Lee HY, Jung JH, Cho HM, Kim SH, Lee KM, Kim HJ, et al., 2015, GRP78 protein expression as prognostic values in neoadjuvant chemoradiotherapy and laparoscopic surgery for locally advanced rectal cancer. Cancer Res Treat 47:804–812 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 111 EP 116 DI 10.1007/s12253-016-0099-9 PG 6 ER PT J AU Romanowicz, H Pyziak, Jablonski, F Brys, M Forma, E Smolarz, B AF Romanowicz, Hanna Pyziak, Lukasz Jablonski, Filip Brys, Magdalena Forma, Ewa Smolarz, Beata TI Analysis of DNA Repair Genes Polymorphisms in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; DNA repair; Genes; Polymorphism ID Breast cancer; DNA repair; Genes; Polymorphism AB Genetic polymorphisms in the DNA repair genes may be associated with increased cancer risk. The purpose of this study was to evaluate the association of the DNA repair genes polymorphisms with the risk of breast cancer development. The study included 200 breast cancer patients and 200 healthy controls. The following polymorphisms were studied: C/G (Ser326Cys, rs1052133) of the hOGG1, A/C (IVS5 + 33, rs3212961) of the ERCC1, A/C (Lys939Gln, rs2228001) of the XPC, C/T (Thr241Met, rs861539) of the XRCC3, G/T (Leu787Leu, rs1800392) of the WRN and G/T (Ser307Ser, rs1056503) of the XRCC4 gene. Presented study showed statistically significant increase in the breast cancer development risk of the G/G hOGG1 genotype (OR 8.13; 95 % CI, 4.37– 15.14; p < 0.001) and for the G hOGG1 allele (OR 5.11; 95% CI, 3.69–7.06; p < 0.001), as well as for the C/C ERCC1 genotype (OR 10.61; 95 % CI, 5.72–19.69; p < 0.001) and the C ERCC1 allele (OR 4.66; 95% CI, 3.43–6.34; p < 0.001) in patients with breast cancer in comparison with healthy control group. We also observed positive association of the C/C XPC genotype (OR 3.80; 95 % CI, 2.27–6.38; p < 0.001) as well as the C XPC allele occurrence with an increased breast cancer development risk (OR 2.65; 95 % CI, 1.98–3.55; p < 0.001). Furthermore, we found an association of the G/T WRN gene polymorphism with increased risk of carcinoma. The hOGG1, ERCC1, XPC and WRN genes polymorphisms may be related to development of breast cancer. C1 [Romanowicz, Hanna] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. [Pyziak, Lukasz] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. [Jablonski, Filip] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. [Brys, Magdalena] Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, Pomorska 141/143, 90-237 Lodz, Poland. [Forma, Ewa] Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, Pomorska 141/143, 90-237 Lodz, Poland. [Smolarz, Beata] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. RP Romanowicz, H (reprint author), Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, 93-338 Lodz, Poland. EM hanna-romanowicz@wp.pl CR Yeo B, Turner N, Jones A, 2014, An update on the medical management of breast cancer. BMJ 348:g3608 Helleday T, Petermann E, Lundin C, Hodgson B, Sharma R, 2008, DNA repair pathways as targets for cancer therapy. Nat Rev Cancer 8:193–204 Matta J, Echenique M, Negron E, Morales L, VargasW, Gaetan FS, Lizardi ER, Torres A, Rosado JO, Bolanos G, Cruz JG, Laboy J, Barnes R, Medina SS, Romero A, Martinez R, Dutil J, Suarez E, Alvarez-Garriga C, Bayona M, 2012, The association of DNA repair with breast cancer risk in women. A comparative observational study. BMC Cancer 12:490 Melis JP, van Steeg H, Luijten M, 2013, Oxidative DNA damage and nucleotide excision repair. Antioxid Redox Signal 18:2409– 2419 Larrea AA, Lujan SA, Kunkel TA, 2010, DNA mismatch repair. Cell 141:730 Brandsma I, Gent DC, 2012, Pathway choice in DNA double strand break repair: observations of a balancing act. Genome Integr 3:9 Truong LN, Li Y, Shi LZ, Hwang PY, He J, Wang H, Razavian N, Berns MW, Wu X, 2013, Microhomology-mediated end joining and homologous recombination share the initial end resection step to repair DNA double-strand breaks in mammalian cells. Proc Natl Acad Sci U S A 110:7720–7725 Huang C-S, Chen J-J, Yang S-Y, Cheng C-W, Chern H-D, Chang K-J,Wu P-E, Shen C-Y, 2004, Breast Cancer Risk Associated with Genotypic Polymorphism of Oxidative DNA Damage Repair Genes - A Multigenic Study of Base Excision Repair and Transcription-Coupled Repair in Cancer Susceptibility. Journal of Genetics and Molecular Biology 15:116–136 Butkiewicz D, Rusin M, Enewold L, Shields PG, Chorazy M, Harris CC, 2001, Genetic polymorphisms in DNA repair genes and risk of lung cancer. Carcinogenesis 22:593–597 Lu AL, Li X, Gu Y, Wright PM, Chang DY, 2001, Repair of oxidative DNA damage: mechanisms and functions. Cell Biochem Biophys 35:141–170 Park J, Chen L, Tockman MS, Elahi A, Lazarus P, 2004, The human 8-oxoguanine DNA N-glycosylase 1, hOGG1, DNA repair enzyme and its association with lung cancer risk. Pharmacogenetics 14:103–109 Sinha RP, Hader DP, 2002)UV-induced DNAdamage and repair: a review. Photochem Photobiol Sci 1:225–236 Yang J, Liu X, Niu P, Zou Y, Gong Z, Yuan J, Wu T, 2007, Dynamic changes of XPA, XPC, XPF, XPG and ERCC1 protein expression and their correlations with levels of DNA damage in human bronchial epithelia cells exposed to benzo[a]pyrene. Toxicol Lett 174:10–17 Pierce AJ, Johnson RD, Thompson LH, Jasin M, 1999, XRCC3 promotes homology-directed repair of DNA damage inmammalian cells. Genes Dev 13:2633–2638 Brosh RM Jr, 2013, DNA helicases involved in DNA repair and their roles in cancer. Nat Rev Cancer 13:542–558 JunopMS,Modesti M, Guarne A, Ghirlando R, Gellert M, YangW, 2000, Crystal structure of the Xrcc4 DNA repair protein and implications for end joining. EMBO J 19:5962–5970 Sterpone S, Mastellone V, Padua L, Novelli F, Patrono C, Cornetta T, Giammarino D, Donato V, Testa A, Cozzi R, 2010, Singlenucleotide polymorphisms in BER and HRR genes, XRCC1 haplotypes and breast cancer risk in Caucasian women. J Cancer Res Clin Oncol 136:631–636 Romanowicz-Makowska H, Smolarz B, Makowski M, Polac I, Pertynski T, 2008, Ser326Cys polymorphism in DNA repair genes hOGG1 in breast cancer women. Pol J Pathol 59:201–204 Synowiec E, Stefanska J, Morawiec Z, Blasiak J, Wozniak K, 2008, Association between DNA damage, DNA repair genes variability and clinical characteristics in breast cancer patients. Mutat Res 648:65–72 Sangrajrang S, Schmezer P, Burkholder I, Waas P, Boffetta P, Brennan P, Bartsch H, Wiangnon S, Popanda O, 2008, Polymorphisms in three base excision repair genes and breast cancer risk in Thai women. Breast Cancer Res Treat 111:279–288 Cai Q, Shu XO, Wen W, Courtney R, Dai Q, Gao YT, Zheng W, 2006, Functional Ser326Cys polymorphism in the hOGG1 gene is not associated with breast cancer risk. Cancer Epidemiol Biomark Prev 15:403–404 Zhang Y, Newcomb PA, Egan KM, Titus-Ernstoff L, Chanock S, Welch R, Brinton LA, Lissowska J, Bardin-Mikolajczak A, Peplonska B, Szeszenia-Dabrowska N, Zatonski W, Garcia-Closas M, 2006, Genetic polymorphisms in base-excision repair pathway genes and risk of breast cancer. Cancer Epidemiol Biomark Prev 15: 353–358 Loizidou MA, Michael T, Neuhausen SL, Newbold RF, Marcou Y, Kakouri E, Daniel M, Papadopoulos P, Malas S, Hadjisavvas A, Kyriacou K, 2009, DNA-repair genetic polymorphisms and risk of breast cancer in Cyprus. Breast Cancer Res Treat 115:623–627 Roberts MR, Shields PG, Ambrosone CB, Nie J, Marian C, Krishnan SS, Goerlitz DS, Modali R, Seddon M, Lehman T, Amend KL, Trevisan M, Edge SB, Freudenheim JL, 2011, Single-nucleotide polymorphisms in DNA repair genes and association with breast cancer risk in the web study. Carcinogenesis 32: 1223–1230 Dhillon VS, Yeoh E, Fenech M, 2011, DNA repair gene polymorphisms and prostate cancer risk in South Australia-results of a pilot study. Urol Oncol 29:641–646 Agalliu I, Kwon EM, Salinas CA, Koopmeiners JS, Ostrander EA, Stanford JL, 2010, Genetic variation in DNA repair genes and prostate cancer risk: results from a population-based study. Cancer Causes Control 21:289–300 Chen L, Elahi A, Pow-Sang J, Lazarus P, Park J, 2003, Association between polymorphism of human oxoguanine glycosylase 1 and risk of prostate cancer. J Urol 170:2471–2474 Xu J, Zheng SL, Turner A, Isaacs SD, Wiley KE, Hawkins GA, Chang BL, Bleecker ER,Walsh PC, Meyers DA, IsaacsWB, 2002, Associations between hOGG1 sequence variants and prostate cancer susceptibility. Cancer Res 62:2253–2257 Li D, Suzuki H, Liu B, Morris J, Liu J, Okazaki T, Li Y, Chang P, Abbruzzese JL, 2009, DNA repair gene polymorphisms and risk of pancreatic cancer. Clin Cancer Res 15:740–746 McWilliams RR, Bamlet WR, Cunningham JM, Goode EL, de Andrade M, Boardman LA, Petersen GM, 2008, Polymorphisms in DNA repair genes, smoking, and pancreatic adenocarcinoma risk. Cancer Res 68:4928–4935 Gangwar R, Ahirwar D, Mandhani A, Mittal RD, 2009, Do DNA repair genes OGG1, XRCC3 and XRCC7 have an impact on susceptibility to bladder cancer in the north Indian population? Mutat Res 680:56–63 Arizono K, Osada Y, Kuroda Y, 2008, DNA repair gene hOGG1 codon 326 and XRCC1 codon 399 polymorphisms and bladder cancer risk in a Japanese population. Jpn J Clin Oncol 38:186–191 Karahalil B, Kocabas NA, Ozcelik T, 2006, DNA repair gene polymorphisms and bladder cancer susceptibility in a Turkish population. Anticancer Res 26:4955–4958 Srivastava A, Srivastava K, Pandey SN, Choudhuri G, Mittal B, 2009, Single-nucleotide polymorphisms of DNA repair genes OGG1 and XRCC1: association with gallbladder cancer in north Indian population. Ann Surg Oncol 16:1695–1703 Engin AB, Karahalil B, Engin A, Karakaya AE, 2011, DNA repair enzyme polymorphisms and oxidative stress in a Turkish population with gastric carcinoma. Mol Biol Rep 38:5379–5386 Palli D, Polidoro S, D’Errico M, Saieva C, Guarrera S, Calcagnile AS, Sera F, Allione A, Gemma S, Zanna I, Filomena A, Testai E, Caini S, Moretti R, Gomez-Miguel MJ, Nesi G, Luzzi I, Ottini L, Masala G, Matullo G, Dogliotti E, 2010, Polymorphic DNA repair and metabolic genes: a multigenic study on gastric cancer. Mutagenesis 25:569–575 Poplawski T, Arabski M, Kozirowska D, Blasinska-Morawiec M, Morawiec Z, Morawiec-Bajda A, Klupinska G, Jeziorski A, Chojnacki J, Blasiak J, 2006, DNA damage and repair in gastric cancer–a correlation with the hOGG1 and RAD51 genes polymorphisms. Mutat Res 601:83–91 Brevik A, Joshi AD, Corral R, Onland-Moret NC, Siegmund KD, Le Marchand L, Baron JA, Martinez ME, Haile RW, Ahnen DJ, Sandler RS, Lance P, Stern MC, 2010, Polymorphisms in base excision repair genes as colorectal cancer risk factors and modifiers of the effect of diets high in red meat. Cancer Epidemiol Biomark Prev 19:3167–3173 Stern MC, Conti DV, Siegmund KD, Corral R, Yuan JM, Koh WP, Yu MC, 2007, DNA repair single-nucleotide polymorphisms in colorectal cancer and their role asmodifiers of the effect of cigarette smoking and alcohol in the Singapore Chinese health study. Cancer Epidemiol Biomark Prev 16:2363–2372 Upadhyay R, Malik MA, Zargar SA, Mittal B, 2010, OGG1 Ser326Cys polymorphism and susceptibility to esophageal cancer in low and high at-risk populations of northern India. J Gastrointest Cancer 41:110–115 Xing DY, Tan W, Song N, Lin DX, 2001, Ser326Cys polymorphism in hOGG1 gene and risk of esophageal cancer in a Chinese population. Int J Cancer 95:140–143 Lan Q, Mumford JL, Shen M, Demarini DM, Bonner MR, He X, Yeager M, Welch R, Chanock S, Tian L, Chapman RS, Zheng T, Keohavong P, Caporaso N, Rothman N, 2004, Oxidative damagerelated genes AKR1C3 and OGG1 modulate risks for lung cancer due to exposure to PAH-rich coal combustion emissions. Carcinogenesis 25:2177–2181 Qian B, Zhang H, Zhang L, Zhou X, Yu H, Chen K, 2011, Association of genetic polymorphisms in DNA repair pathway genes with non-small cell lung cancer risk. Lung Cancer 73:138– 146 De Ruyck K, Szaumkessel M, De Rudder I, Dehoorne A, Vral A, Claes K, Velghe A, Van Meerbeeck J, Thierens H, 2007, Polymorphisms in base-excision repair and nucleotide-excision repair genes in relation to lung cancer risk. Mutat Res 631:101–110 Cho EY, Hildesheim A, Chen CJ, Hsu MM, Chen IH, Mittl BF, Levine PH, Liu MY, Chen JY, Brinton LA, Cheng YJ, Yang CS, 2003, Nasopharyngeal carcinoma and genetic polymorphisms of DNA repair enzymes XRCC1 and hOGG1. Cancer Epidemiol Biomark Prev 12:1100–1104 Elahi A, Zheng Z, Park J, Eyring K, McCaffrey T, Lazarus P, 2002, The human OGG1 DNA repair enzyme and its association with orolaryngeal cancer risk. Carcinogenesis 23:1229–1234 Krupa R, Sobczuk A, Poplawski T, Wozniak K, Blasiak J, 2011, DNA damage and repair in endometrial cancer in correlation with the hOGG1 and RAD51 genes polymorphism. Mol Biol Rep 38: 1163–1170 Zhao H, Qin C, Yan F, Wu B, Cao Q, Wang M, Zhang Z, Yin C, 2011, hOGG1 Ser326Cys polymorphism and renal cell carcinoma risk in a Chinese population. DNA Cell Biol 30:317–321 Li Q, Huang L, Rong L, Xue Y, Lu Q, Rui Y, Li J, Tong N, Wang M, Zhang Z, Fang Y, 2011, hOGG1 Ser326Cys polymorphism and risk of childhood acute lymphoblastic leukemia in a Chinese population. Cancer Sci 102:1123–1127 Stanczyk M, Sliwinski T, Cuchra M, Zubowska M, Bielecka- Kowalska A, Kowalski M, Szemraj J, Mlynarski W, Majsterek I, 2011, The association of polymorphisms in DNA base excision repair genes XRCC1, OGG1 and MUTYH with the risk of childhood acute lymphoblastic leukemia. Mol Biol Rep 38:445–451 Sakamoto T, Higaki Y, Hara M, Ichiba M, Horita M, Mizuta T, Eguchi Y, Yasutake T, Ozaki I, Yamamoto K, Onohara S, Kawazoe S, Shigematsu H, Koizumi S, Tanaka K, 2006, hOGG1 Ser326Cys polymorphism and risk of hepatocellular carcinoma among Japanese. J Epidemiol 16:233–239 Dianzani I, Gibello L, Biava A, GiordanoM, Bertolotti M, Betti M, Ferrante D, Guarrera S, Betta GP, Mirabelli D,Matullo G, Magnani C, 2006, Polymorphisms in DNA repair genes as risk factors for asbestos-related malignant mesothelioma in a general population study. Mutat Res 599:124–134 Wei B, Zhou Y, Xu Z, Xi B, Cheng H, Ruan J, ZhuM, Hu Q,Wang Q,Wang Z, Yan Z, Jin K, Zhou D, Xuan F, Huang X, Shao J, Lu P, 2011, The effect of hOGG1 Ser326Cys polymorphism on cancer risk: evidence from a meta-analysis. PLoS One 6:e27545 Gu D, Wang M, Zhang Z, Chen J, 2010, Lack of association between the hOGG1 Ser326Cys polymorphism and breast cancer risk: evidence from 11 case–control studies. Breast Cancer Res Treat 2010; 527–531. Guo XG, Wang Q2, Xia Y3, Zheng L, 2015, The C8092A polymorphism in the ERCC1 gene and breast carcinoma risk: A metaanalysis of case-control studies. Int J Clin Exp Med 8: 3691–3699. Jorgensen TJ, Visvanathan K, Ruczinski I, Thuita L, Hoffman S, Helzlsouer KJ, 2007, Breast cancer risk is not associated with polymorphic forms of xeroderma pigmentosum genes in a cohort of women from Washington County, Maryland. Breast Cancer Res Treat 101:65–71 Wang Z, Xu Y, Tang J,Ma H, Qin J, Lu C,Wang X, Hu Z,Wang X, Shen H, 2009, A polymorphism in Werner syndrome gene is associated with breast cancer susceptibility in Chinese women. Breast Cancer Res Treat 118:169–175 Sangrajrang S, Schmezer P, Burkholder I, Boffetta P, Brennan P, Woelfelschneider A, Bartsch H,Wiangnon S, Cheisilpa A, Popanda O, 2007, The XRCC3 Thr241Met polymorphism and breast cancer risk: a case-control study in a Thai population. Biomarkers 12:523– 532 Chiu CF, Wang HC, Wang CH, Wang CL, Lin CC, Shen CY, Chiang SY, Bau DT, 2008, A new single nucleotide polymorphism in XRCC4 gene is associated with breast cancer susceptibility in Taiwanese patients. Anticancer Res 28:267–270 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 117 EP 123 DI 10.1007/s12253-016-0110-5 PG 7 ER PT J AU Oh, RH An, HCh Yoo, JN Lee, HS AF Oh, Rim Hye An, Hyeok Chang Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutations in the Mononucleotide Repeats of TAF1 and TAF1L Genes in Gastric and Colorectal Cancers with Regional Heterogeneity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TAF1; TAF1L; Basal transcription component; Mutation; Cancers; Microsatellite instability ID TAF1; TAF1L; Basal transcription component; Mutation; Cancers; Microsatellite instability AB Initiation of transcription by RNA polymerase II requires TATA-box-binding protein (TBP)-associated factors (TAFs). TAF1 is a major scaffold by which TBP and TAFs interact in the basal transcription factor. TAF1L is a TAF1 homologue with 95 % amino acid identity with TAF1. TAF1 is involved in apoptosis induction and cell cycle regulation, but roles of TAF1 and TAF1L in tumorigenesis remain unknown. The aim of this study was to explore whether TAF1 and TAF1L genes were mutated in gastric (GC) and colorectal cancers (CRC). In a public database, we found that TAF1 and TAF1L genes had mononucleotide repeats in the coding sequences that might be mutation targets in the cancers with microsatellite instability (MSI). We analyzed the mutations in 79 GC and 124 CRC by single-strand conformation polymorphism analysis and DNA sequencing. In the present study, we found TAF1 frameshift mutations (3.8 % of CRC with MSI-H) and TAF1L frameshift mutations (2.9 % of GC and 3.8 % of CRC with MSI-H). These mutations were not found in stable MSI/low MSI (MSS/MSI-L) (0/90). In addition, we analyzed intratumoral heterogeneity (ITH) of TAF1 and TAF1L frameshift mutations in 16 CRC and found that two and one CRC harbored regional ITH of TAF1 and TAF1L frameshift mutations, respectively. Our data indicate that TAF1 and TAF1L genes harbored not only somatic mutations but also mutational ITH, which together might play a role in tumorigenesis of GC and CRC with MSI-H. Our results also suggest that ultra-regional mutation analysis is required for a comprehensive evaluation of mutation status in these tumors. C1 [Oh, Rim Hye] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [An, Hyeok Chang] The Catholic University of Korea, College of Medicine, Department of General Surgery, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Wassarman DA, Sauer F, 2001, TAF(II)250: a transcription toolbox. J Cell Sci 114:2895–2902 Lee DH, Gershenzon N, Gupta M, Ioshikhes IP, Reinberg D, Lewis BA, 2005, Functional characterization of core promoter elements: the downstream core element is recognized by TAF1. Mol Cell Biol 25:9674–9686 Juven-Gershon T, Kadonaga JT, 2010, Regulation of gene expression via the core promoter and the basal transcriptional machinery. Dev Biol 339:225–229 Hilton TL, Li Y, Dunphy EL, Wang EH, 2005, TAF1 histone acetyltransferase activity in Sp1 activation of the cyclin D1 promoter. Mol Cell Biol 25:4321–4332 Tavassoli P, Wafa LA, Cheng H, Zoubeidi A, Fazli L, Gleave M, Snoek R, Rennie PS, 2010, TAF1 differentially enhances androgen receptor transcriptional activity via its N-terminal kinase and ubiquitin-activating and -conjugating domains. Mol Endocrinol 24:696–708 Makino S, Kaji R, Ando S, Tomizawa M, Yasuno K, Goto S, Matsumoto S, Tabuena MD, Maranon E, Dantes M, Lee LV, Ogasawara K, Tooyama I, Akatsu H, Nishimura M, Tamiya G, 2007, Reduced neuron-specific expression of the TAF1 gene is associatedwith X-linked dystonia-parkinsonism. Am J Hum Genet 80:393–406 Kimura J, Nguyen ST, Liu H, Taira N,Miki Y, Yoshida K, 2008, A functional genome-wide RNAi screen identifies TAF1 as a regulator for apoptosis in response to genotoxic stress. Nucleic Acids Res 36:5250–5259 Zhao S, Choi M, Overton JD, Bellone S, Roque DM, Cocco E, Guzzo F, EnglishDP,Varughese J, Gasparrini S, Bortolomai I, Buza N, Hui P, Abu-Khalaf M, Ravaggi A, Bignotti E, Bandiera E, Romani C, Todeschini P, Tassi R, Zanotti L, Carrara L, Pecorelli S, Silasi DA, Ratner E, Azodi M, Schwartz PE, Rutherford TJ, Stiegler AL, Mane S, Boggon TJ, Schlessinger J, Lifton RP, SantinAD, 2013, Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci U S A 110:2916–2921 Wang PJ, Page DC, 2002, Functional substitution for TAF(II)250 by a retroposed homolog that is expressed in human spermatogenesis. Hum Mol Genet 11:2341–2346 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8: 305–311 Geiersbach KB, SamowitzWS, 2011)Microsatellite instability and colorectal cancer. Arch Pathol Lab Med 135:1269–1277 Ogino S, Goel A, 2008, Molecular classification and correlates in colorectal cancer. J Mol Diagn 10:13–27 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Calin GA, Gafa R, Tibiletti MG, Herlea V, Becheanu G, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression inmicrosatellite instability high, MSI-H, colon cancers correlates with clinico-pathological parameters: a study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89:230–235 Almendro V, Cheng YK, Randles A, Itzkovitz S, Marusyk A, Ametller E, Gonzalez-Farre X, Munoz M, Russnes HG, Helland A, Rye IH, Borresen-Dale AL, Maruyama R, van Oudenaarden A, DowsettM, Jones RL, Reis-Filho J, Gascon P, GonenM, Michor F, Polyak K, 2014, Inference of tumor evolution during chemotherapy by computational modeling and in situ analysis of genetic and phenotypic cellular diversity. Cell Rep 6:514–527 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 125 EP 130 DI 10.1007/s12253-016-0107-0 PG 6 ER PT J AU Sun, Y Zhu, L Chang, X Chen, J Lang, J AF Sun, Yin Zhu, Lan Chang, Xiaoyan Chen, Jie Lang, Jinghe TI Clinicopathological Features and Treatment Analysis of Rare Aggressive Angiomyxoma of the Female Pelvis and Perineum – a Retrospective Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Angiomyxoma; Diagnosis; Treatment; Prognosis; Genitalia; female; Pelvis ID Angiomyxoma; Diagnosis; Treatment; Prognosis; Genitalia; female; Pelvis AB The study was to evaluate the clinicopathological features of aggressive angiomyxoma (AAM) of the female pelvis and perineum and its treatments. This was a retrospective study of female patients with AAM admitted to our hospital. Clinical and pathological data were analyzed, as well as the postsurgical follow-up. Median age at initial presentation was 41 years. Thirteen patients had lesions involving adjacent organs. Eighteen patients underwent complete tumor resection, while one patient underwent partial tumor resection. The tumors were soft in texture, pink in color, and had mucus on the surface. A microscopic examination revealed that the tumors were non-encapsulated, with spindle cells and stellate cells of almost identical size loosely distributed in the myxoid stroma, and vessels of different sizes and wall thicknesses. Immunohistochemistry indicated that AAMs were strongly positive for CD34 and smooth muscle actin, moderately positive for desmin, estrogen receptors and progesterone receptor, and mostly negative for S-100. After a median follow-up of 24 months, the recurrence rate was 33.3 %. Four recurrences were in patients with positive initial margins. AAM is a slow growing, locally invasive, benign tumor. Complete resection could lead to lower recurrence rate compared with incomplete resection. Follow-up is necessary for recurrent cases with repeated surgeries. The overall prognosis could be favorable. C1 [Sun, Yin] Peking Union Medical College Hospital, Department of Obstetrics and Gynecology, 100000 Beijing, China. [Zhu, Lan] Peking Union Medical College Hospital, Department of Obstetrics and Gynecology, 100000 Beijing, China. [Chang, Xiaoyan] Peking Union Medical College Hospital, Department of Pathology, 100000 Beijing, China. [Chen, Jie] Peking Union Medical College Hospital, Department of Pathology, 100000 Beijing, China. [Lang, Jinghe] Peking Union Medical College Hospital, Department of Obstetrics and Gynecology, 100000 Beijing, China. RP Zhu, L (reprint author), Peking Union Medical College Hospital, Department of Obstetrics and Gynecology, 100000 Beijing, China. EM lanzhusci@126.com CR Steeper TA, Rosai J, 1983, Aggressive angiomyxoma of the female pelvis and perineum. Report of nine cases of a distinctive type of gynecologic soft-tissue neoplasm. Am J Surg Pathol 7(5):463–475 Chan YM, Hon E, Ngai SW, Ng TY, Wong LC, 2000, Aggressive angiomyxoma in females: is radical resection the only option? Acta Obstet Gynecol Scand 79(3):216–220 Fetsch JF, LaskinWB, Lefkowitz M, KindblomLG,Meis-Kindblom JM, 1996, Aggressive angiomyxoma: a clinicopathologic study of 29 female patients. Cancer 78(1):79–90., DOI 10.1002/(SICI)1097- 0142(19960701)78:1<79::AID-CNCR13>3.0.CO;2-4 Piura B, Shaco-Levy R, 2005, Pedunculated aggressive angiomyxoma arising from the vaginal suburethral area: case report and review of literature. Eur J Gynaecol Oncol 26(5):568–571 Chihara Y, Fujimoto K, Takada S, Hirayama A, ChoM, Yoshida K, Ozono S, Hirao Y, 2003, Aggressive angiomyxoma in the scrotum expressing androgen and progesterone receptors. Int J Urol 10(12): 672–675 Baruah S, Latthe P, Bhatti NR, Ghataura SS, 2004, Aggressive angiomyxoma of the vulva. Hosp Med 65(4):248–249 Chen L, Schink JC, Panares BN, Barbuto D, Lagasse LD, 1998, Resection of a giant aggressive angiomyxoma in the Philippines. Gynecol Oncol 70(3):435–439 Jeyadevan NN, Sohaib SA, Thomas JM, Jeyarajah A, Shepherd JH, Fisher C, 2003, Imaging features of aggressive angiomyxoma. Clin Radiol 58(2):157–162 Brunelle S, Bertucci F, Chetaille B, Lelong B, Piana G, Sarran A, 2013, Aggressive angiomyxoma with diffusion-weighted magnetic resonance imaging and dynamic contrast enhancement: a case report and review of the literature. Case Rep Oncol 6(2):373–381., DOI 10.1159/000353487 Smith HO, Worrell RV, Smith AY, Dorin MH, Rosenberg RD, Bartow SA, 1991, Aggressive angiomyxoma of the female pelvis and perineum: review of the literature. Gynecol Oncol 42(1):79–85 Corson SL, Rosato EL, Rosato FE, Eschelman DJ, 2002, Aggressive angiomyxoma of the pelvis: case report and review. Int J Fertil Womens Med 47(6):248–252 Outwater EK, Marchetto BE, Wagner BJ, Siegelman ES, 1999, Aggressive angiomyxoma: findings on CT and MR imaging. AJR Am J Roentgenol 172(2):435–438., DOI 10.2214/ajr.172.2.9930798 Stewart ST,McCarthy SM(2004, Case 77: aggressive angiomyxoma. Radiology 233(3):697–700., DOI 10.1148/radiol.2333021670 McCluggageWG, 2009, Recent developments in vulvovaginal pathology. Histopathology 54(2):156–173., DOI 10.1111/j.1365- 2559.2008.03098.x McCluggage WG, Patterson A, Maxwell P, 2000, Aggressive angiomyxoma of pelvic parts exhibits oestrogen and progesterone receptor positivity. J Clin Pathol 53(8):603–605 Gungor T, Zengeroglu S, Kaleli A, Kuzey GM, 2004, Aggressive angiomyxoma of the vulva and vagina. A common problem: misdiagnosis. Eur J Obstet Gynecol Reprod Biol 112(1):114–116 Siassi RM, Papadopoulos T, Matzel KE, 1999, Metastasizing aggressive angiomyxoma. N Engl J Med 341(23):1772., DOI 10.1056 /NEJM199912023412316 Dierickx I, Deraedt K, Poppe W, Verguts J, 2008, Aggressive angiomyxoma of the vulva: a case report and review of literature. Arch Gynecol Obstet 277(6):483–487., DOI 10.1007/s00404-008- 0561-3 Amezcua CA, Begley SJ, Mata N, Felix JC, Ballard CA, 2005, Aggressive angiomyxoma of the female genital tract: a clinicopathologic and immunohistochemical study of 12 cases. Int J Gynecol Cancer 15(1):140–145., DOI 10.1111/j.1048-891x.2005.15015.x Htwe M, Deppisch LM, Saint-Julien JS, 1995, Hormone-dependent, aggressive angiomyxoma of the vulva. Obstet Gynecol 86(4 Pt 2):697–699 Han-Geurts IJ, van Geel AN, van Doorn L,Md B, Eggermont AM, Verhoef C, 2006, Aggressive angiomyxoma: multimodality treatments can avoid mutilating surgery. Eur J Surg Oncol 32(10):1217– 1221., DOI 10.1016/j.ejso.2006.06.008 Simo M, Zapata C, Esquius J, Domingo J, 1992, Aggressive angiomyxoma of the female pelvis and perineum. Report of two cases and review of the literature. Br J Obstet Gynaecol 99(11): 925–927 McCluggage WG, Jamieson T, Dobbs SP, Grey A, 2006, Aggressive angiomyxoma of the vulva: dramatic response to gonadotropin-releasing hormone agonist therapy. Gynecol Oncol 100(3):623–625., DOI 10.1016/j.ygyno.2005.09.033 Steiner E, Schadmand-Fischer S, Schunk K, Bezzi I, Weikel W, Pilch H, Knapstein PG, 2001, Perineal excision of a large angiomyxoma in a young woman following magnetic resonance and angiographic imaging. Gynecol Oncol 82(3):568–570., DOI 10.1006/gyno.2001.6296 Magtibay PM, Salmon Z, Keeney GL, Podratz KC, 2006, Aggressive angiomyxoma of the female pelvis and perineum: a case series. Int J Gynecol Cancer 16(1):396–401., DOI 10.1111 /j.1525-1438.2006.00225.x Rhomberg W, Jasarevic Z, Alton R, Kompatscher P, Beer G, Breitfellner G, 2000, Aggressive angiomyxoma: irradiation for recurrent disease. Strahlenther Onkol 176(7):324–326 Fine BA, Munoz AK, Litz CE, Gershenson DM, 2001, Primary medical management of recurrent aggressive angiomyxoma of the vulva with a gonadotropin-releasing hormone agonist. Gynecol Oncol 81(1):120–122., DOI 10.1006/gyno.2000.6119 Brooks SE, Balidimos I, Reuter KL, Khan A, 1998, Virtual consult–aggressive angiomyxoma of the vulva: impact of GnRH agonists. Medscape Womens Health 3(3):4 Shinohara N, Nonomura K, Ishikawa S, Seki H, Koyanagi T, 2004, Medical management of recurrent aggressive angiomyxoma with gonadotropin-releasing hormone agonist. Int J Urol 11(6):432–435., DOI 10.1111/j.1442-2042.2004.00816.x Behranwala KA, Thomas JM, 2003, 'Aggressive' angiomyxoma: a distinct clinical entity. Eur J Surg Oncol 29(7):559–563 Rawlinson NJ,WestWW, NelsonM, Bridge JA, 2008, Aggressive angiomyxoma with t(12;21, and HMGA2 rearrangement: report of a case and review of the literature. Cancer Genet Cytogenet 181(2): 119–124., DOI 10.1016/j.cancergencyto.2007.11.008 Blandamura S, Cruz J, Faure Vergara L, Machado Puerto I, Ninfo V, 2003, Aggressive angiomyxoma: a second case of metastasis with patient's death. Hum Pathol 34(10):1072–1074 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 131 EP 137 DI 10.1007/s12253-016-0109-y PG 7 ER PT J AU Kullmann, T Gauthier, H Serrate, C Pouessel, D le Maignan, Ch Misset, JL Culine, S AF Kullmann, Tamas Gauthier, Helene Serrate, Camille Pouessel, Damien le Maignan, Christine Misset, Jean-Louis Culine, Stephane TI To Treat or Not to Treat Metastatic Cancer Patients with Poor Performance Status: a Prospective Experience SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Metastatic cancer; Performance status; Cytotoxic chemotherapy; Supportive care; Therapeutic decision; Survival ID Metastatic cancer; Performance status; Cytotoxic chemotherapy; Supportive care; Therapeutic decision; Survival AB Administration of cytotoxic chemotherapy for patients with metastatic cancer and poor performance status is a daily clinical challenge. Guidelines only help to select a therapeutic regimen but do not offer a clear response whether or not the patients should be treated.We performed a prospective analysis in 139 metastatic patients with performance status > 1 according to the Eastern Cooperative Oncology Group scale. A decision was considered correct if patients treated with a medical anticancer treatment lived over 3 months or alternatively patients not treated had a survival under 3 months. The predominant tumor type was non-small cell lung cancer. Patients were chemotherapy naive in 87 cases (63 %). A new line of medical anticancer treatment was started in 107 cases (77 %). The median survival of the study population was 11 weeks (range, 1–53). 84 patients (60 %) died within 3 months while 55 patients (40 %) lived more than 3 months after decision. Treatment decisions were considered as appropriate in 81 cases (58 %). No patient was considered as undertreated. The analysis by pathology allowed to identify pathologies where decisions were correct in the majority of the cases (renal, urothelial and small cell lung cancers), pathologies where appropriate and inappropriate decisions were balanced (prostate, ovarian and breast cancers) and pathologies where decisions for treatment were excessive (non-small cell lung cancer and unknown primary). This prospective study was conducted as part of the evaluation of professional practices in our department. Administration of a medical anticancer treatment validated with patients with good performance status may be harmful for patients with poor performance status. The findings resulted in recommendations for daily practice in order to help physicians, especially for the "don’t go" decisions. Until the identification of new prognostic factors for survival and/or the development of therapies making sensitive currently chemoresistant diseases, the initiation of a medical anticancer treatment outside standard situations should result from a consensual decision team or the inclusion in a clinical trial. C1 [Kullmann, Tamas] Hopital Saint Louis, Department of Medical OncologyParis, France. [Gauthier, Helene] Hopital Saint Louis, Department of Medical OncologyParis, France. [Serrate, Camille] Hopital Saint Louis, Department of Medical OncologyParis, France. [Pouessel, Damien] Hopital Saint Louis, Department of Medical OncologyParis, France. [le Maignan, Christine] Hopital Saint Louis, Department of Medical OncologyParis, France. [Misset, Jean-Louis] Hopital Saint Louis, Department of Medical OncologyParis, France. [Culine, Stephane] Hopital Saint Louis, Department of Medical OncologyParis, France. RP Kullmann, T (reprint author), Hopital Saint Louis, Department of Medical Oncology, Paris, France. EM kullmanndoki@hotmail.com CR Oken MM, Creech RH, Tormey DC et al, 1982, Toxicity and response criteria of the Eastern Cooperative Oncology group. Am J Clin Oncol 5:649–655 Liu S, Wang D, Chen B et al, 2011, The safety and efficacy of EGFR TKIs monotherapy versus single-agent chemotherapy using third-generation cytotoxics as the first-line treatment for patients with advanced non-small cell lung cancer and poor performance status. Lung Cancer 73:203–210 Arkenau HT, Barriuso J, IlmosD et al, 2009, Prospective validation of a prognostic score to improve patient selection for oncology phase I trials. J Clin Oncol 27:2692–2696 Braga S, 2011)Why do our patients get chemotherapy until the end of life? Ann Oncol 22:2345–2348 Arkenau HT, Olmos D, Ang JE et al, 2008, Clinical outcome and prognostic factors for patients treated within the context of a phase I study: the Royal Marsden Hospital experience. Br J Cancer 98: 1029–1033 Barbot AC, Mussault P, Ingrand P, Tourani JM, 2008, Assessing 2- months clinical prognosis in hospitalized patients with advanced solid tumors. J Clin Oncol 26:2538–2542 Maltoni M, Scarpi E, Pittureri C et al, 2012, Prospective comparison of prognostic scores in palliative care cancer populations. Oncologist 17:446–454 Massard C, Borget I, Le Deley MC et al, 2012, Prognostic value of circulating VEGFR2+ bone marrow-derived progenitor cells in patients with advanced cancer. Eur J Cancer 48:1354–1362 Yun YH, Kwak M, Park SM et al, 2007, Chemotherapy use and associated factors among cancer patients near the end of life. Ann Oncol 72:164–171 Braga S, Miranda A, Fonseca R et al, 2007, The aggressiveness of cancer care in the last three months of life: a retrospective single center analysis. Psychooncology 16:863–868 Harrington SE, Smith TJ, 2008, The role of chemotherapy at the end of life: "When is enough, enough?". JAMA 299:2667–2678 Temel JS,Greer JA,MuzikanskyAet al, 2010, Early palliative care for patients with metastatic non-small cell lung cancer. N Engl J Med 363:733–742 Greer JA, Pirl WF, Jackson VA et al, 2012, Effect of early palliative care on chemotherapy use and end-of-life care in patients with metastatic non-small-cell lung cancer. J Clin Oncol 30:394–400 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 139 EP 144 DI 10.1007/s12253-016-0111-4 PG 6 ER PT J AU Choi, JE Kim, SM Song, YS Yoo, JN Lee, HS AF Choi, Ji Eun Kim, Sung Min Song, Yong Sang Yoo, Jin Nam Lee, Hyung Sug TI Intratumoral Heterogeneity of Frameshift Mutations in MECOM Gene is Frequent in Colorectal Cancers with High Microsatellite Instability SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MECOM; Frameshift mutation; Colon cancer; Microsatellite instability; Intratumoral heterogeneity ID MECOM; Frameshift mutation; Colon cancer; Microsatellite instability; Intratumoral heterogeneity AB MECOM gene, also known as EVI, encodes a transcriptional regulator involved in hematopoiesis, apoptosis, development and proliferation. In blood system, MECOM is considered an oncogene, but in solid tumors it has both oncogenic and tumor suppressor activities. Low frequent somatic mutations of MECOM have been detected in many cancers including colorectal cancers (CRC), but the mutation status with respect to the microsatellite instability (MSI) has not been studied. There is an A7 mononucleotide repeat in MECOM coding sequences that could be a mutation target in the cancers with MSI. We analyzed the A7 of MECOM in 79 CRCs with high MSI (MSI-H) and 65 microsatellite stable/ low MSI (MSS/MSI-L) CRCs by single-strand conformation polymorphism analysis and DNA sequencing. Overall, we found MECOM frameshift mutations in 6 (7.6 %) CRCs with MSI-H, but not in MSS/MSI-L cancers (0/65) (p<0.025).We also analyzed intratumoral heterogeneity (ITH) of the MECOM frameshift mutation in 16 CRCs and found that four CRCs (25.0 %) harbored regional ITH of the frameshift mutations. Our data indicate that MECOM gene harbors both somatic frameshift mutations and mutational ITH, which together may be features of CRC with MSI-H. C1 [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Song, Yong Sang] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of PathologySeoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Weiser R, 2007, The oncogene and developmental regulator EVI1: expression, biochemical properties, and biological functions. Gene 396:346–357 Hoyt PR, Bartholomew C, Davis AJ, Yutzey K, Gamer LW, Potter SS, Ihle JN, Mucenski ML, 1997, The Evi1 proto-oncogene is required at midgestation for neural, heart, and paraxial mesenchyme development. Mech Dev 65:55–70 Kilbey A, Stephens V, Bartholomew C, 1999, Loss of cell cycle control by deregulation of cyclin-dependent kinase 2 kinase activity in evi-1 transformed fibroblasts. Cell Growth Differ 10:601–610 Alliston T, Ko TC, Cao Y, Liang YY, Feng XH, Chang C, Derynck R, 2005, Repression of bone morphogenetic protein and activininducible transcription by evi-1. J Biol Chem 280:24227–24237 Kurokawa M, Mitani K, Irie K, Matsuyama T, Takahashi T, Chiba S, Yazaki Y, Matsumoto K, Hirai H, 1998, The oncoprotein evi-1 represses TGF-beta signalling by inhibiting Smad3. Nature 394: 92–96 Kurokawa M, Mitani K, Yamagata T, Takahashi T, Izutsu K, Ogawa S, Moriguchi T, Nishida E, Yazaki Y, Hirai H, 2000, The evi-1 oncoprotein inhibits c-Jun N-terminal kinase and prevents stress-induced cell death. EMBO J 19:2958–2968 Liu Y, Chen L, Ko TC, Fields AP, Thompson EA, 2006, Evi1 is a survival factor which conveys resistance to both TGFbeta- and taxol-mediated cell death via PI3K/AKT. Oncogene 25:3565–3675 Buonamici S, Li D, Chi Y, Zhao R, Wang X, Brace L, Ni H, Saunthararajah Y, Nucifora G, 2004, EVI1 induces myelodysplastic syndrome in mice. J Clin Invest 114:713–719 Morishita K, Parganas E, William CL, Whittaker MH, Drabkin H, Oval J, Taetle R, Valentine MB, Ihle JN, 1992, Activation of EVI1 gene expression in human acute myelogenous leukemias by translocations spanning 300-400 kilobases on chromosome band 3q26. Proc Natl Acad Sci U S A 89:3937–3941 Nucifora G, Rowley JD, 1995, AML1 and the 8;21 and 3;21 translocations in acute and chronic myeloid leukemia. Blood 86:1–14 Brooks DJ,Woodward S, Thompson FH, Dos Santos B, Russell M, Yang JM, Guan XY, Trent J, Alberts DS, Taetle R, 1996, Expression of the zinc finger gene EVI-1 in ovarian and other cancers. Br J Cancer 74:1518–1525 Nanjundan M, Nakayama Y, KW C, Lahad J, Liu J, K L, WL K, Smith-McCune K, Fishman D, JW G, GB M, 2007, Amplification of MDS1/EVI1 and EVI1, located in the 3q26.2 amplicon, is associated with favorable patient prognosis in ovarian cancer. Cancer Res 67:3074–3084 Lee JJ, Sholl LM, Lindeman NI, Granter SR, Laga AC, Shivdasani P, Chin G, Luke JJ, Ott PA, Hodi FS, MC JM, Lin JY, Werchniak AE, Haynes HA, Bailey N, Liu R, Murphy GF, Lian CG, 2015, Targeted next-generation sequencing reveals high frequency of mutations in epigenetic regulators across treatment-naive patient melanomas. Clin Epigenetics 7:59 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 McGranahan N, Swanton C, 2015, Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 27:15–26 Kim TM, Jung SH, Baek IP, Lee SH, Choi YJ, Lee JY, Chung YJ, Lee SH, 2014, Regional biases in mutation screening due to intratumoural heterogeneity of prostate cancer. J Pathol 233:425– 435 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8: 305–311 Oh HR, An CH, Yoo NJ, Lee SH, 2015, Frameshift mutations of MUC15 gene in gastric and its regional heterogeneity in gastric and colorectal cancers. Pathol Oncol Res 21:713–718 Choi MR, An CH, Yoo NJ, Lee SH, 2015, Laminin gene LAMB4 is somatically mutated and expressionally altered in gastric and colorectal cancers. APMIS 123:65–71 Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, Xie M, Zhang Q, McMichael JF, Wyczalkowski MA, Leiserson MD, Miller CA, Welch JS, Walter MJ, Wendl MC, Ley TJ, Wilson RK, Raphael BJ, Ding L, 2013, Mutational landscape and significance across 12 major cancer types. Nature 502:333–339 Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, Meyerson M, Gabriel SB, Lander ES, Getz G, 2014, Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505:495–501 Gonzalez-Perez A, Perez-Llamas C, Deu-Pons J, Tamborero D, Schroeder MP, Jene-Sanz A, Santos A, Lopez-Bigas N, 2013, IntOGen-mutations identifies cancer drivers across tumor types. Nat Methods 10:1081–1082 Calin GA, Gafa R, Tibiletti MG, Herlea V, Becheanu G, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression inmicrosatellite instability high, MSI-H, colon cancers correlates with clinico-pathological parameters: a study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89:230–235 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 145 EP 149 DI 10.1007/s12253-016-0112-3 PG 5 ER PT J AU Moldvay, J Fabian, K Jackel, M Nemeth, Zs Bogos, K Furak, J Tiszlavicz, L Fillinger, J Dome, B Schaff, Zs AF Moldvay, Judit Fabian, Katalin Jackel, Marta Nemeth, Zsuzsanna Bogos, Krisztina Furak, Jozsef Tiszlavicz, Laszlo Fillinger, Janos Dome, Balazs Schaff, Zsuzsa TI Claudin-1 Protein Expression Is a Good Prognostic Factor in Non-Small Cell Lung Cancer, but only in Squamous Cell Carcinoma Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Claudin; Immunohistochemistry; Survival ID Lung cancer; Claudin; Immunohistochemistry; Survival AB The aim of the study was to investigate the correlation between claudin (CLDN) protein expression and clinicopathological parameters as well as survival in histological subtypes of non-small cell lung cancer. Archived surgical resection specimens of 137 pathologic stage I primary bronchial cancers including 49 adenocarcinomas of non-lepidic variants (ADC), 46 adenocarcinomas of lepidic variants (L-ADC), and 42 squamous cell carcinomas (SCC) were examined. Immunohistochemistry (IHC) using antibodies against CLDN1,-2,-3,-4,-7 proteins as well as semiquantitative estimation (IHC scores 0–5) were performed. Claudin IHC scores of L-ADC differed significantly from ADC (CLDN1: p = 0.009, CLDN2: p = 0.005, CLDN3: p = 0.004, CLDN4: p = 0.001, CLDN7: p < 0.001, respectively) and SCC (CLDN1: p < 0.001, CLDN3: p < 0.001, CLDN7: p < 0.001, respectively). Highly significant CLDN3-CLDN4 parallel expression could be demonstrated in ADC and L-ADC (p < 0.001 in both), which was not observed in SCC (p = 0.131). ADC and SCC showed no correlation with smoking, whereas in case of L-ADC heavier smoking correlated with higher CLDN3 expression (p = 0.020). Regarding claudin expression and survival, in SCC significant correlation could be demonstrated between CLDN1 IHC positivity and better survival (p = 0.038). In NSCLC as a whole, high CLDN2 expression proved to be a better prognostic factor when compared with cases where CLDN2 IHC score was 0–1 vs. 2–5 (p = 0.009), however, when analyzed separately, none of the histological subgroups showed correlation between CLDN2 expression and overall survival. The claudin expression pattern was significantly different not only between the SCC–ADC and SCC–L-ADC but also between the L-ADC and ADC histological subgroups, which strongly underlines that L-ADC represents a distinct entitywithin the ADC group. CLDN1 overexpression is a good prognostic factor in NSCLC, but only in the SCC subgroup. C1 [Moldvay, Judit] National Koranyi Institute of Pulmonology, Piheno u. 1, H-1122 Budapest, Hungary. [Fabian, Katalin] Semmelweis University, Department of Pulmonology, Diosarok u. 1/c, H-1125 Budapest, Hungary. [Jackel, Marta] Military Hospital, Department of Pathology, Robert Karoly krt. 44, H-1134 Budapest, Hungary. [Nemeth, Zsuzsanna] Queen’s University, Centre for Cancer Research and Cell Biology, 97 Lisburn Road, BT9 7AE Belfast, Ireland. [Bogos, Krisztina] National Koranyi Institute of Pulmonology, Piheno u. 1, H-1122 Budapest, Hungary. [Furak, Jozsef] University of Szeged, Department of Surgery, Szokefalvi-Nagy u. 6, H-6720 Szeged, Hungary. [Tiszlavicz, Laszlo] University of Szeged, Department of Pathology, Allomas u. 2, H-6720 Szeged, Hungary. [Fillinger, Janos] National Institute of Oncology, Department of Diagnostic Pathology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Dome, Balazs] National Koranyi Institute of Pulmonology, Piheno u. 1, H-1122 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. RP Moldvay, J (reprint author), National Koranyi Institute of Pulmonology, H-1122 Budapest, Hungary. EM drmoldvay@hotmail.com CR Yoshizawa A, Sumiyoshi S, Sonobe M, et al., 2013, Validation of the IASLC/ATS/ERS lung adenocarcinoma classification for prognosis and association with EGFR and KRAS gene mutations: analysis of 440 Japanese patients. J Thorac Oncol 8(1):52–61 Travis WD, Brambilla E, Riely GJ, 2013, New pathologic classification of lung cancer: relevance for clinical practice and clinical trials. J Clin Oncol 31(8):992–1001 Sawada N, Murata M, Kikuchi K, et al., 2003, Tight junctions and human diseases. Med Electron Microsc 36:147–156 Chao YC, Pan SH, Yang SC, et al., 2009, Claudin-1 is a metastasis suppressor and correlates with clinical outcome in lung adenocarcinoma. Am J Respir Crit Care Med 179(2):123–133 Epub 2008 Sep 11 Merikallio H, Kaarteenaho R, Paakko P, et al., 2011, Impact of smoking on the expression of claudins in lung carcinoma. Eur J Cancer 47(4):620–630 Zhang Z, Wang A, Sun B, et al., 2013, Expression of CLDN1 and CLDN10 in lung adenocarcinoma in situ and invasive lepidic predominant adenocarcinoma. J Cardiothorac Surg 8(1) Zhang ZF1, Pei BX, Wang AL, et al. Expressions of CLDN1 and insulin-like growth factor 2 are associated with poor prognosis in stage N2 non-small cell lung cancer. Chin Med J. 2013; 126(19):3668–74. Yamamoto T, Oshima T, Yoshihara K, et al., 2010, Reduced expression of claudin-7 is associated with poor outcome in non-small cell lung cancer. Oncol Lett 1(3):501–505 Travis WD, Brambilla E, Noguchi M, et al., 2011, International association for the study of lung cancer/american thoracic society/ european respiratory society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol 6:244–285 Moldvay J, Jackel M, Paska C, Soltesz I, Schaff Z, Kiss A, 2007, Distinct claudin expression profile in histologic subtypes of lung cancer. Lung Cancer 57(2):159–167 Epub 2007 Apr 6 Kominsky SL, Argani P, Korz D, et al., 2003, Loss of the tight junction protein claudin-7 correlates with histological grade in both ductal carcinoma in situ and invasive ductal carcinoma of the breast. Oncogene 22:2021–2033 Eftang LL, Esbensen Y, Tannaes TM, et al., 2013, Up-regulation of CLDN1 in gastric cancer is correlated with reduced survival. BMC Cancer., DOI 10.1186/1471-2407-13-586 Szasz AM, Tokes AM, Micsinai M, et al., 2011, Prognostic significance of claudin expression changes in breast cancer with regional lymph node metastasis. Clin Exp Metastasis 28(1):55–63 Epub 2010 Oct 21 Kolokytha P, Yiannou P, Keramopoulos D, et al., 2014, Claudin-3 and claudin-4: distinct prognostic significance in triple-negative and luminal breast cancer. Appl Immunohistochem Mol Morphol 22(2):125–131 Fritzsche FR, Oelrich B, Johannsen M, et al., 2008, Claudin-1 protein expression is a prognostic marker of patient survival in renal cell carcinomas. Clin Cancer Res 14(21):7035–7042 Resnick MB, Konkin T, Routhier J, et al., 2005, Claudin-1 is a strong prognostic indicator in stage II colonic cancer: a tissue microarray study. Mod Pathol 18:511–518 Jung H, Jun KH, Jung JH, et al., 2011, The expression of claudin-1, claudin-2, claudin-3, and claudin-4 in gastric cancer tissue. J Surg Res 167(2):e185–e191., DOI 10.1016/j.jss.2010.02.010 Hsueh C, Chang YS, Tseng NM, et al., 2010, Expression pattern and prognostic significance of claudins 1, 4, and 7 in nasopharyngeal carcinoma. Hum Pathol 41(7):944–950 Epub 2010 Mar 23 Melchers LJ, Bruine de Bruin L, Schnell U, et al., 2013, Lack of claudin-7 is a strong predictor of regional recurrence in oral and oropharyngeal squamous cell carcinoma. Oral Oncol 49(10):998– 1005 Kim CJ, Lee JW, Choi JJ, et al., 2011, High claudin-7 expression is associated with a poor response to platinum-based chemotherapy in epithelial ovarian carcinoma. Eur J Cancer Apr. 47(6):918–925., DOI 10.1016/j.ejca.2010.11.007 Epub 2010 Dec 4 Szasz AM, Nyirady P, Majoros A, et al., 2010, Beta-catenin expression and claudin expression pattern as prognostic factors of prostatic cancer progression. BJU Int 105(5):716–722 Epub 2009 Oct 10 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 151 EP 156 DI 10.1007/s12253-016-0115-0 PG 6 ER PT J AU Rekhi, B Verma, V Gulia, A Jambhekar, AN Desai, S Juvekar, LSh Bajpai, J Puri, A AF Rekhi, Bharat Verma, Vivek Gulia, Ashish Jambhekar, A Nirmala Desai, Subhash Juvekar, L Shashikant Bajpai, Jyoti Puri, Ajay TI Clinicopathological Features of a Series of 27 Cases of Post-Denosumab Treated Giant Cell Tumors of Bones: A Single Institutional Experience at a Tertiary Cancer Referral Centre, India SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Giant cell tumor of bone; Denosumab; Targeted therapy; Bone tumor ID Giant cell tumor of bone; Denosumab; Targeted therapy; Bone tumor AB Giant cell tumor of bone (GCTB) is mostly a benign tumor, but associated with recurrences and metastasis. Lately, denosumab is being utilized in the treatment of certain GCTBs. Twenty-seven tumors, analyzed in the present study, occurred in 16 males and 11 females (M: F = 1.45:1), in the age-range of 16 to 47 years (mean = 29.5, median = 29). Most tumors were identified in the tibia(6) and femur(6), followed by the humerus(3), radius(3), pelvis(3), fibula(3), sacrum(1), metacarpal(1) and metatarsal(1) bones. There were 18(66.6 %) primary and 9(33.3 %) recurrent tumors. Exact tumor size (19 cases) varied from 3.7 to 15 cm (mean = 7.8, median = 6.4). Eight of the 19 tumors (42.1 %) had size more than or equal to 8 cm. On histopathologic examination of postdenosumab treated specimens, more than half cases (15)(55.5 %) revealed complete absence of osteoclast-like giant cells (OCLGs) and 12 cases revealed residual OCLGCs. In addition, there was replacement by fibro-osseous tissue, including reactive woven bone or osteoid in most cases, followed by variable amount of spindle cells, hyalinisation, fibrosis and chronic inflammatory cells, including lymphocytes, macrophages and plasma cells. Post-treatment follow-up (25 cases, 92.5 %), over 7–27 months duration (median = 18), revealed 20 cases continuously disease–free. Five patients developed recurrences at 9, 12, 13, 14 and 18 months, respectively. Out of these, who underwent repeat surgical intervention, 4 patients are alive with no evidence of disease and a single patient, planned for a second surgery, is alive-with-disease. Denosumab was mostly offered to patients with large sized, borderline salvageable tumors, in order to decrease the morbidity of index surgical procedure, that led to disappearance of OCLGCs in most cases. Post-denosumab treated GCT cases appear as low grade osteosarcomas on histopathologic examination, but lack the clinical behaviour of an osteosarcoma, therefore may be considered as pseudo malignant bony lesions. C1 [Rekhi, Bharat] Tata Memorial Hospital, Department of Surgical Pathology, 8th Floor, Annex Building, Dr E.B. Road, Parel, 400012 Mumbai, India. [Verma, Vivek] Tata Memorial Hospital, Department of Surgical Oncology (Bone and Soft Tissues), Parel, 400012 Mumbai, India. [Gulia, Ashish] Tata Memorial Hospital, Department of Surgical Oncology (Bone and Soft Tissues), Parel, 400012 Mumbai, India. [Jambhekar, A Nirmala] Tata Memorial Hospital, Department of Surgical Pathology, 8th Floor, Annex Building, Dr E.B. Road, Parel, 400012 Mumbai, India. [Desai, Subhash] Tata Memorial Hospital, Department of Radiodiagnosis, Parel, 400012 Mumbai, India. [Juvekar, L Shashikant] Tata Memorial Hospital, Department of Radiodiagnosis, Parel, 400012 Mumbai, India. [Bajpai, Jyoti] Tata Memorial Hospital, Department of Medical Oncology, Parel, 400012 Mumbai, India. [Puri, Ajay] Tata Memorial Hospital, Department of Surgical Oncology (Bone and Soft Tissues), Parel, 400012 Mumbai, India. RP Rekhi, B (reprint author), Tata Memorial Hospital, Department of Surgical Pathology, 400012 Mumbai, India. EM rekhi.bharat@gmail.com CR Athanasou NA, Bansal M, Forsyth R, Reid RP, Sapi Z, 2013, Giant cell tumor of bone. In: Fletcher CD, Bridge JA, Hogendoorn PCW, Mertens F, eds, World Health Organization, WHO, classification of tumors of soft tissue and bone, 4th edn. IARC Press, Lyon pp 321–324 Beebe–Dimmer JL, Cetin K, Fryzek JP, Schuetze SM, Schwartz K, 2009, The epidemiology ofmalignant giant cell tumors of bone: an analysis of data fromthe surveillance, epidemiology and end results program, 1975–2004). Rare Tumors 1:e52 SzendroiM(2004, Giant-cell tumour of bone. J Bone Joint Surg Br 86:5–12 Gupta R, Seethalakshmi V, Jambhekar NA, Prabhudesai S, Merchant N, Puri A, Agarwal M, 2008, Clinicopathologic profile of 470 giant cell tumors of bone from a cancer hospital in western India. Ann Diagn Pathol 12:239–248 Yasuda H, Shima N, Nakagawa N, Yamaguchi K, Kinosaki M, Mochizuki S, Tomoyasu A, Yano K, Goto M, Murakami A, Tsuda E, Morinaga T, Higashio K, Udagawa N, Takahashi N, Suda T, 1998, Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc Natl Acad Sci U S A 95:3597–3602 Hsu H, Lacey DL, Dunstan CR, Solovyev I, Colombero A, Timms E, Tan HL, Elliott G, Kelley MJ, Sarosi I,Wang L, Xia XZ, Elliott R, Chiu L, Black T, Scully S, Capparelli C, Morony S, Shimamoto G, Bass MB, Boyle WJ, 1999, Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proc NatlAcad Sci US A 96:3540–3545 Huang L, Xu JK,Wood DJ, Zheng MH, 2000, Gene expression of osteoprotegerin ligand, osteoprotegerin, and receptor activator of NF-B in Giant cell tumor of bone possible involvement in tumor cell-induced osteoclast-like cell formation. Am J Pathol 156:761– 767 Morgan T, Atkins GJ, Trivett MK, Johnson SA, Kansara M, Schlicht SL, Slavin JL, Simmons P, Dickinson I, Powell G, Choong PF, Holloway AJ, Thomas DM(2005, Molecular profiling of giant cell tumor of bone and the osteoclastic localization of ligand for receptor activator of nuclear factor kappa B. Am J Pathol 167:117–128 Won KY, Kalil RK, Kim YW, Park YK, 2011, RANK signalling in bone lesions with osteoclast-like giant cells. Pathology 43:318–321 Balke M, Hardes J, 2010, Denosumab: a breakthrough in treatment of giant cell tumour of bone? Lancet Oncol 11:218–219 Lipton A, Jacobs I, 2011, Denosumab: benefits of RANK ligand inhibition in cancer patients. Curr Opin Support Palliat Care 5:258– 264 Thomas D, Henshaw R, Skubitz K, Staddon A, Blay JY, Roudier M, Smith J, Ye Z, Sohn W, Dansey R, Jun S, 2010, Denosumab in patients with giant-cell tumour of bone: an open-label, phase 2 study. Lancet Oncol 11:275–280 Chawla S, Henshaw R, Seeger L, Choy E, Blay JY, Ferrari S, Kroep J, Grimer R, Reichardt P, Rutkowski P, Schuetze S, Skubitz K, Staddon A, Thomas D, Qian Y, Jacobs I, 2013, Safety and efficacy of denosumab for adults and skeletally mature adolescents with giant cell tumour of bone: interim analysis of an open-label, parallel- group, phase 2 study. Lancet Oncol 14:901–908 Branstetter DG, Nelson SD, Manivel JC, Blay JY, Chawla S, Thomas DM, Jun S, Jacobs I, 2012, Denosumab induces tumor reduction and bone formation in patients with giant-cell tumor of bone. Clin Cancer Res 18:4415–4424 Wojcik J, Rosenberg AE, Bredella MA, Choy E, Hornicek FJ, Nielsen GP, Deshpande V, 2016, Denosumab-treated giant cell tumor of bone exhibits morphologic overlap with malignant giant cell tumor of bone. Am J Surg Pathol 40:72–80 Girolami I, Mancini I, Simoni A, Baldi GG, Simi L, Campanacci D, Beltrami G, Scoccianti G, D'Arienzo A, Capanna R, Franchi A, 2015, Denosumab treated giant cell tumour of bone: a morphological, immunohistochemical and molecular analysis of a series. J Clin Pathol., DOI 10.1136 /jclinpath-2015-203248 Desai S, Rekhi B, Jambhekar NA, 2011, Musculoskeletal system. Bone. In: Desai SS, Bal M, Rekhi B, Jambhekar NA, eds). Grossing of surgical oncology specimens: a practical guide towards complete pathology reporting. Tata Memorial Hospital pp. 70–79 Stadler N, Fingernagel T, Hofstaetter SG, Trieb K, 2015, A recurrent giant cell tumor of bone treated with denosumab. Clin Pract 5: 697., DOI 10.4081/cp.2015.697. eCollection Demirsoy U, Karadogan M, Selek O, Anik Y, Aksu G, Muezzinoglu B, Corapcioglu F, 2014, Golden bullet-denosumab: early rapid response of metastatic giant cell tumor of the bone. J Pediatr Hematol Oncol 36:156–158 Blay J, Chawla SP, Broto JM, Choy E, Dominkus M, Engellau J, Grimer R, Henshaw RM, Palmerini E, Reichardt P, Rutkowski P, Skubitz KM, Thomas DM, Zhao Y, Qian Y, Jacobs IA, 2011, Denosumab safety and efficacy in giant cell tumor of bone, gctb): interim results from a phase ii study [abstract 10034] J Clin Oncol 29. [Available online at: http://www.asco.org/ASCOv2 /Meetings/Abstracts?&vmview=abst_detail_view&confID=102 &abstractID=82649; cited August 16, 2013] Aponte-Tinao LA, Piuzzi NS, Roitman P, Farfalli GL, 2015, A high-grade sarcoma arising in a patient with recurrent benign giant cell tumor of the proximal tibia while receiving treatment with denosumab. Clinl Orthopaed Related Res 473:3050–3055 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 157 EP 164 DI 10.1007/s12253-016-0123-0 PG 8 ER PT J AU Matrai, Z Andrikovics, H Szilvasi, A Bors, A Kozma, A Adam, E Halm, G Karaszi, Tordai, A Masszi, T AF Matrai, Zoltan Andrikovics, Hajnalka Szilvasi, Aniko Bors, Andras Kozma, Andras Adam, Emma Halm, Gabriella Karaszi, Eva Tordai, Attila Masszi, Tamas TI Lipoprotein Lipase as a Prognostic Marker in Chronic Lymphocytic Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lipoprotein lipase; CLL; Prognosis; IgVH mutation; 13q deletion; Survival ID Lipoprotein lipase; CLL; Prognosis; IgVH mutation; 13q deletion; Survival AB The marked clinical heterogeneity of CLL makes early prognosis assessment important. Lipoprotein lipase (LPL) has been shown to confer adverse prognosis in CLL, recent data indicating it might also contribute to CLL cell survival and metabolism.We determined LPL mRNA expression in unselected peripheral blood of 84 CLL patients by RT PCR. Results were correlated with other prognostic markers and outcome. 30/84 (40 %) of cases were LPL positive based on the cutoff established by ROC analysis. In LPL positive patients significantly shorter median survival (136 vs 258 months, p < 0.0001) and time to first treatment intervals (36 vs 144 months, p < 0.002) were documented. LPL values correlated with male gender, higher stages, more treatment requirement, CD38 positivity and unmutated IgVH genes. Among cases with 13q deletion, LPL positivity identified a subcohort with poor outcome (median survival 108 months vs NR, p < 0.0001). In multivariate analysis, cytogenetic aberrations and LPL had significant impact on survival. Our results confirm that LPL is a strong predictor of outcome in CLL, able to improve prognostic accuracy in good risk cytogenetic subgroups. The relationship between its prognostic and functional role in CLL needs to be explored further. C1 [Matrai, Zoltan] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Andrikovics, Hajnalka] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Szilvasi, Aniko] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Bors, Andras] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Kozma, Andras] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Adam, Emma] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Halm, Gabriella] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Karaszi, Eva] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Tordai, Attila] National Institute of Haematology, Blood Transfusion and ImmunologyBudapest, Hungary. [Masszi, Tamas] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. RP Matrai, Z (reprint author), Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Budapest, Hungary. EM matraiz1@gmail.com CR Hamblin TJ, Davis Z, Gardiner A, Oscier DG, Stevenson FK, 1999, Unmutated Ig VH genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood 94(6):1848–1854 Damle RN, Wasil T, Fais F et al, 1999, IgV Gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood 94:1840–1847 Chiorazzi N, 2012, Implications of new prognostic markers in chronic lymphocytic leukemia. Hematology 2012:76–87 Krober A, Seiler T, Benner A, Bullinger L, Bruckle E, Lichter P, Dohner H, Stilgenbauer S, 2002, VH mutation status, CD38 expression level, genomic aberrations, and survival in chronic lymphocytic leukemia. Blood 100:1410–1416 Rosenwald A, Alizadeh AA, Widhopf G et al, 2001, Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med 194(11):1639–1647 Klein U, Tu Y, StolovitzkyGA, Mattioli M, Cattoretti G, Husson H, Freedman A, Inghirami G, Cro L, Baldini L, Neri A, Califano A, Dalla-Favera R, 2001, Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J Exp Med 194(11):1625–1638 Oppezzo P, Vasconcelos Y, Settegrana C et al, 2005, The LPL/ ADAM29 expression ratio is a novel prognosis indicator in chronic lymphocytic leukemia. Blood 106(2):650–657 Heintel D, Kienle D, Shehata M et al, 2005, High expression of lipoprotein lipase in poor risk B-cell chronic lymphocytic leukemia. Leukemia 19(7):1216–1223 Nuckel H, Huttmann A, Klein-Hitpass L, Schroers R, Fuhrer A, Sellmann L, Duhrsen U, Durig J, 2006, Lipoprotein lipase expression is a novel prognostic factor in B-cell chronic lymphocytic leukemia. Leuk Lymphoma 47(6):1053–1061 van’t Veer MB, Brooijmans AM, Langerak AW et al, 2006, The predictive value of lipoprotein lipase for survival in chronic lymphocytic leukemia. Haematologica 91(1):56–63 Van Bockstaele F, Pede V, Janssens A, Callewaert F, Offner F, Verhasselt B, Philippe J, 2007, Lipoprotein lipase mRNA expression in whole blood is a prognostic marker in B cell chronic lymphocytic leukemia. Clin Chem 53(2):204–212 Nikitin EA, Malakho SG, Biderman BV et al, 2007, Expression level of lipoprotein lipase and dystrophin genes predict survival in B-cell chronic lymphocytic leukemia. Leuk Lymphoma 48(5):912–922 Xu W, Li JY, Shen QD, Wu YJ, Yu H, Fan L, 2009, Expression level of lipoprotein lipase in Chinese patients with chronic lymphocytic leukemia and its correlationwith other prognostic factors. Int J Lab Hematol 31(5):552–559 Kaderi MA, KanduriM, Buhl AM et al, 2011, LPL is the strongest prognostic factor in a comparative analysis of RNA-based markers in early chronic lymphocytic leukemia. Haematologica 96(8): 1153–1160 Kristensen L, Kristensen T, Abildgaard N et al, 2015, LPL gene expression is associated with poor prognosis in CLL and closely related to NOTCH1 mutations. Eur J Haematol ., DOI 10.1111 /ejh.1270017 Dec Mead JR, Irvine SA, Ramji DP, 2002, Lipoprotein lipase: structure, function, regulation, and role in disease. J Mol Med 80(12):753–769 Kuemmerle NB, Rysman E, Lombardo PS et al, 2011, Lipoprotein lipase links dietary fat to solid tumor cell proliferation. Mol Cancer Ther 10(3):427–436 Dashnamoorthy R, Abermil N, Behesti A et al, 2014, The lipid addiction of diffuse large B-cell lymphoma and potential treatment strategies with novel fatty acid synthase, FASN, small molecule inhibitors. Blood 124:4490 Zaidi N, Lupien L, Kuemmerle NB, Kinlaw WB, Swinnen JV, Smans K, 2013, Lipogenesis and lipolysis: the pathways exploited by the cancer cells to acquire fatty acids. Prog Lipid Res 52(4):585–589 Rozovski U, Grgurevic S, Bueso-Ramos C et al, 2015, Aberrant LPL expression, driven by STAT3, mediates free fatty acid metabolism in CLL cells. Mol Cancer Res 13:944–953 Rozovski U, Hazan-Halevy I, Barzilai M, Keating MJ, Estrov Z, 2015, Metabolism pathways in chronic lymphocytic leukemia. Leuk Lymphoma 8:1–8 van Dongen JJ, Langerak AW, BruggemannM, Evans PA, Hummel M, Lavender FL et al, 2003, Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T- c e l l r e c e p t o r gene re c o m b i n a t i o n s i n s u s p e ct lymphoproliferations: report of the BIOMED-2 concerted action BMH4-CT98-3936. Leukemia 17(12):2257–2317 Matthews C, Catherwood M, Morris TC, Alexander HD, 2004, Routine analysis of IgVH mutational status in CLL patients using BIOMED-2 standardized primers and protocols. Leuk Lymphoma 45(9):1899–1904 Metz CE, 1978, Basic principles of ROC analysis. Semin Nucl Med 8:283–298 Matrai Z, Sherrington PD, Pettitt AR, Zuzel M, Cawley JC, 2003, CD38 expression in CLL is a marker of cell cycling activity. Blood 102(11):671a Damle RN, Temburni S, Calissano C, Yancopoulos S, Banapour T, Sison C, Allen SL, Rai KR, 2007, Chiorazzi N. CD38 expression labels an activated subset within chronic lymphocytic leukemia clones enriched in proliferating B cells. Blood 110:3352–3359 Zubiaur M, Fernandez O, Ferrero E, Salmeron J, Malissen B, Malavasi F, Sancho J, 2002, CD38 is associated with lipid rafts and upon receptor stimulation leads to Akt/protein kinase B and Erk activation in the absence of the CD3- immune receptor tyrosinebased activation motifs. J Biol Chem 277:13–22 Dohner H, Stilgenbauer S, Benner A, Leupolt E, Krober A, Bullinger L et al, 2003, Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med 343:1910–1916 Strefford JC, Parker H, Parker A, Robinson H, Chaplin T, Chen X et al, 2009, 13q deletion size predicts disease progression and response to treatment in patients with chronic lymphocytic leukaemia. Blood 114:671 Pettitt AR, Emmens CJ, Lin K, Harris RJ, 2005, Chronic lymphocytic leukaemia patients with a biallelic deletion of 13q14 have a more favourable profile of prognostic factors compared with cases that have a monoallelic 13q14 deletion. Blood 106:5019 Hernandez JA, Rodriguez AE, Gonzalez M et al, 2009, A high number of losses in 13q14 chromosome band is associated with a worse outcome and biological differences in patients wi t h B-cell chronic lymphoid leukemia. Haematologica 94:364–371 Ambrosio MR, Piccaluga PP, Ponzoni M, Rocca BJ, Malagnino V, OnoratiMet al, 2012, The alteration of lipid metabolism in Burkitt lymphoma identifies a novel marker: adipophilin. PLoS One 7(8): e44315., DOI 10.1371/journal.pone.0044315 Porpaczy E, Tauber S, Bilban M et al, 2013, Lipoprotein lipase in chronic lymphocytic leukaemia - strong biomarker with lack of functional significance. Leuk Res 37(6):631–636 Mansouri M, Sevov M, Fahlgren E, Tobin G, Jondal M, Osorio L, Roos G, Olivecrona G, Rosenquist R, 2010, Lipoprotein lipase is differentially expressed in prognostic subsets of chronic lymphocytic leukemia but displays invariably low catalytical activity. Leuk Res 34(3):301–306 Pallasch CP, Schwamb J, Konigs S, Schulz A, Debey S, Kofler D et al, 2008, Targeting lipid metabolism by the lipoprotein lipase inhibitor orlistat results in apoptosis of B-cell chronic lymphocytic leukemia cells. Leukemia 22:585–592 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 165 EP 171 DI 10.1007/s12253-016-0132-z PG 7 ER PT J AU Aziz, F Gao, W Yan, Q AF Aziz, Faisal Gao, Wei Yan, Qiu TI Fucosyltransferase-4 and Oligosaccharide Lewis Y Antigen as potentially Correlative Biomarkers of Helicobacter pylori CagA Associated Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Correlative biomarkers; Fucosyltransferase IV; Gastritis; Gastric ulcer; Gastric cancer; H. pylori CagA; Lewis Y; P-EGFR ID Correlative biomarkers; Fucosyltransferase IV; Gastritis; Gastric ulcer; Gastric cancer; H. pylori CagA; Lewis Y; P-EGFR AB H. pylori cytotoxin associated antigen A (CagA) plays a significant role in the progression of gastric cancer but their effect on fucosylation to develop gastric cancer is unknown. Fucosyltransferase IV (FUT4) is the key enzyme for synthesis of LewisY (LeY) carried by glycoproteins and glycolipids on the cell membrane. Herein, we compare the expression of CagA, p-EGFR, FUT4 and LeY in gastritis (n = 128, 176), gastric ulcer (n = 174, 213), and gastric cancer (n = 323, 261) tissue and serum samples, respectively by IHC and ELISA. Moreover, we investigated the potential correlation of CagA with FUT4 and LeY overexpression through EGFR activation. IHC and ELISA results showed higher positive cases of H. pylori CagA (83, 86 %), p-EGFR (81, 72 %), FUT4 (91, 97 %) and LeY (93, 92 %) in gastric cancer, compared to gastritis and gastric ulcer, H. pylori CagA (58, 67 & 59, 73 %), p-EGFR (52, 63 & 35, 47 %), FUT4 (68, 78 & 67, 82 %) and LeY (62,76 & 65, 85 %), respectively. We found a significant high expression (H-Value) of CagA (1.79, 1.66), p-EGFR (1.53, 1.58), FUT4 (2.14, 1.66) and LeY (1.69, 1.61) in gastric cancer tissues and serum, respectively as compared to chronic gastritis and gastric ulcers, CagA (0.64,1.14), p- EGFR (0.856, 0.678), FUT4 (0.949,1.197) and LeY (0.68,1.008) (P < 0.0001), respectively. Furthermore, H. pylori CagA showed significant correlation with p-EGFR (R–0.62, −0.74), FUT4 (R–0.81, −0.76) and LeY (R–0.82, −0.70) in gastric tissues and serum (P < 0.0001). H. pylori CagA plays key role in the development of gastric cancer with overexpression of FUT4/LeY, serve as potentially correlative biomarkers of H. pylori CagA associated gastric cancer. C1 [Aziz, Faisal] Dalian Medical University, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Department of Biochemistry and Molecular BiologyDalian, China. [Gao, Wei] Dalian Medical University, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Department of Biochemistry and Molecular BiologyDalian, China. [Yan, Qiu] Dalian Medical University, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Department of Biochemistry and Molecular BiologyDalian, China. RP Yan, Q (reprint author), Dalian Medical University, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Department of Biochemistry and Molecular Biology, Dalian, China. EM yanqiu63@126.com CR Aziz F, Sherwani SK, Akhtar SS, Kazmi SU, 2014a, Development of an in-house enzyme-linked immunosorbent assay based on surface whole cell antigen for diagnosis of helicobacter pylori infection in patients with gastroduodenal ulcer disease. World J Microbiol Biotechnol 30:305–315 Zhu Y, Zhong X, Zheng S, Du Q, Xu W, 2005, Transformed immortalized gastric epithelial cells by virulence factor cagA of helicobacter pylori through Erk mitogen-activated protein kinase pathway. Oncogene 24:3886–3895 Aziz F, Taj Y, Kazmi SU, 2013b, In-house enzyme-linked immunosorbent assay based on helicobacter pylori sonicate whole cell antigen for diagnosis of helicobacter pylori infection in Karachi. Pakistan Int J Microbiol Adv Immunol 1(4):24–31 Aziz F, Chen X, Yang X, Yan Q, 2014b, Prevalence and correlation with clinical diseases of helicobacter pylori cagA and vacA genotype among gastric patients from Northeast China. Biomed Res Int 2014:142980 Yamaoka Y, 2008, Increasing evidence of the role of helicobacter pylori SabA in the pathogenesis of gastroduodenal disease. J Infect Dev Ctries 1:174–181 HatakeyamaM, Higashi H, 2005, Helicobacter pylori CagA: a new paradigm for bacterial carcinogenesis. Cancer Sci 96:835–843 Asahi M, Azuma T, Ito S, Ito Y, Suto H, Nagai Y et al, 2000, Helicobacter pylori CagA protein can be tyrosine phosphorylated in gastric epithelial cells. J Exp Med 191:593–602 Odenbreit S, Puls J, Sedlmaier B, Gerland E, Fischer W, Haas R, 2000, Translocation of helicobacter pylori CagA into gastric epithelial cells by type IV secretion. Science 287:1497–1500 Menaker RJ, Ceponis PJ, Jones NL, 2004, Helicobacter pylori induces apoptosis of macrophages in association with alterations in the mitochondrial pathway. Infect Immun 72:2889–2898 De la Cruz-Herrara CF, Flores-Luna L, Guterrez-Xicotencatl L, Chihu-Amparan L, Sanchez-Aleman MA, Lazcano-Ponce E, Torres J, Ayala G, 2013, IgG2 response and low IgG titer specific to helicobacter pylori CagA as serological markers for gastric cancer. J Med Microbiol 62:591–598 Aziz F,Wang X, Liu J, Yan Q, 2016, Ginsenoside Rg3 induces FUT4- mediated apoptosis in H. Pylori CagA-treated gastric cancer cells by regulating SP1 and HSF1 expressions. Toxicol in Vitro 31:158–166 Moran AP, Gupta A, Joshi L, 2011, Sweet-talk: role of host glycosylation in bacterial pathogenesis of the gastrointestinal tract. Gut 60:1412–1425 Reis CA, Osorio H, Silva L, Gomes C,David L, 2010, Alteration in glycosylation as biomarkers for cancer detection. J Clin Pathol 63: 322–329 Delmotte P, Degroote S, Lafitte JJ, LamblinG, Perini JM, Roussel P, 2002, Tumor necrosis factor alpha increases the expression of glycosyltransferases and sulfotransferases responsible for the biosynthesis of sialylated and/or sulfated Lewis x epitopes in the human bronchial mucosa. J Biol Chem 277:424–431 Miyoshi E, Moriwaki K, Nakagawa T, 2008, Biological function of fucosylation in cancer biology. J Biochem 14:725–729 Moriwaki K, Miyoshi E, 2010, Fucosylation and gastrointestinal cancer. World J Hepatol 2:151–161 Yang X, Zhang Z, Jia S, Liu Y, Wang X, Yan Q, 2007, Overexpression of fucosyltransferase IV in A431 cell line increase cell proliferation. Int J Biochem Cell Biol 39:1722–1730 Sheu BS, Sheu SM, Yang HB, Huang AH,Wu JJ, 2003)Host gastric Lewis expression determines the bacterial density of helicobacter pylori in babA2 genopositive infection. Gut 52:927–932 Wang XQ, Zhu ZM, Fenderson BA, Zeng GQ, Cao YJ, Jiang GT, 1998, Effects of monoclonal antibody directed to LeYon implantation in the mouse. Mol Hum Reprod 4:295–300 Dettke M, Palfi G, Pursch E, Fischer MB, Loibner H, 2001, Increased expression of the blood group-related Lewis Y antigen on synovial fluid granulocytes of patients with arthritic joint diseases. Rheumatology, Oxford, 40:1033–1037 Madjd Z, Parsons T, Watson NF, Spendlove I, Ellis I, Durrant LG, 2006, High expression of Lewis y/b antigens is associated with decreased survival in lymph node negative breast carcinomas. Breast Cancer Res 7:80–87 Aziz F, Yang X, Wang X, Yan Q, 2014c, Anti-LeY antibody enhances therapeutic efficacy of celecoxib against gastric cancer by downregulation of MAPKs/COX-2 signaling pathway: correlation with clinical study. J Cancer Res Clin Oncol 141(7):1221–1235 Aziz F, Qiu Y, 2014, The role of anti-LeY antibody in the downregulation ofMAPKs/COX-2 pathway in gastric cancer. Curr Drug Targets 15:469–476 Lin Y, Ueda J, Kikuchi S, Totsuka Y, Wei WQ, Qiao YL, Inoue M, 2011, Comparative epidemiology of gastric cancer between Japan and China. World J Gastroenterol 17:4421–4428 Bjorkholm B, Falk P, Engstrand L, Nyren O, 2003, Helicobacter pylori: resurrection of the cancer link. J Intern Med 253:102–119 Figueroa G, Troncoso M, Toledo MS, Faundez G, Acuna R, 2002, Prevalence of serum antibodies to helicobacter pylori VacA and CagA and gastric diseases in Chile. J Med Microbiol 51:300–304 Khan A, Farooqui A, Raza Y, Rasheed F, Manzoor H, Akhtar SS, Quraishy MS, Rubino S, Kazmi SU, Paglietti B, 2013, Prevalence, diversity and diseases association of helicobacter pylori in dyspeptic patients from Pakistan. J Infect Dev Ctries 7:220–228 Liu JJ, Lin B, Hao YY, Li FF, Liu DW, Qi Y, Zhu LC, Zhang SL, Iwamori M, 2010, Lewis(y, antigen stimulates the growth of ovarian cancer cells via regulation of the epidermal growth factor receptor pathways. Oncol Rep 23:833–841 Keates S, Sougioultzis S, Keates AC, Zhao D, Peek RM Jr, Shaw LM, Kelly CP, 2001, cag + Helicobacter pylori induce transactivation of the epidermal growth factor receptor in AGS gastric epithelial cells. J Biol Chem 276:48127–48134 Zhang Z, Sun P, Liu J, Fu L, Yan J, Liu Y, Yu L, Wang X, Yan Q, 2008, Suppression of FUT1/FUt4 expression by SiRNA inhibits tumor growth. Biochim Biophys Acta 1783:287–296 Ilver D, Arnqvist A, Ogren J, Frick IM, Kersulyte D, Incecik ET, Berg DE, Covacci A, Engstrand L, Boren T, 1998, Helicobacter pylori adhesin binding fucosylated histo-blood group antigens revealed by retagging. Science 279:373–377 Kudo T, Ikehara Y, Togayachi A, Morozumi K, Watanabe M, Nakamura M et al, 1998, Up-regulation of a set of glycosyltransferases genes in human colorectal cancer. Lab Investig 78:797–711 Saito H, Hiraiwa N, Sawada-Kasugai M, Akamatsu S, Tachikawa T, Kasai Y et al, 1997, Altered mRNA expression of specific molecular species of fucosyl- and sialyl-transferases in human colorectal cancer tissues. Int J Cancer 71:556–564 Taniguchi A, Suga R, Matsumoto K, 2002, Expression and transcriptional regulation of the human alpha1,3-fucosyltransferase 4, FUT4, gene in myeloid and colon adenocarcinoma cell lines. Biochem Biophys Res Commun 273(1):370–376 Martin-Satue M, Marrugat R, Cancelas JA, Blanco J, 1998, Enhanced expression of α1,3-fucosyltransferase genes correlates with E-selectin-mediated adhesion and metastastic potential of human lung adnocarcinoma cells. Cancer Res 58:1544–1550 Yan X, Lin Y, Liu S, 2015, Faisal Aziz, Qiu Yan, fucosyltransferase IV, FUT4, as an effective biomarker for the diagnosis of breast cancer. Biomed Pharmacother 70:299–304 Dohi T, Abe K, Ohshiba S, 1989, Immunohistochemical study of carbohydrate antigen expression in gastric carcinoma. Gastroenterol Jpn 24:239–245 Sakamoto S.,Watanabe T., Tokumaru T., Takagi H., Nakazato H. and Lloyd K.O, 1989, Expression of Lewisa, Lewisb, Lewisx, Lewisy, siayl-Lewisa, and sialyl-Lewisx blood group antigens in human gastric carcinoma and in normal gastric tissue. Cancer Res 49:745–752. Kobayashi K, Sakamoto J, Kito T, Yamamura Y, Koshikawa T, Fujita M, Watanabe T, Nakazato H, 1993, Lewis blood grouprelated antigen expression in normal gastric epithelium, intestinal metaplasia, gastric adenoma, and gastric carcinoma. Am J Gastroenterol 88:919–924 Escrevente E, Machado C, Brito CA, Reis A, Stoeck S, Runz A, Marme P, Altevogt JC, 2006, Different expression levels of alpha3/ 4 fucosyltransferases and Lewis determinants in ovarian carcinoma tissues and cell lines. Int J Oncol 29:557–566 Wirth HP, Yang M, Karita M, Blaser MJ, 1990, Expression of human cell surface glycoconjugates Lewis x and Lewis y by helicobacter pylori isolates is related to CagA status. Infect Immun 64:4598–4605 Aziz F, Yang X,Wang X, Qiu Y, 2013a, Helicobacter pylori infection leads to gastric cancer with the coexpression of Lewis Y and CA724. Glycoconj J 30:411 Aziz F, Yang X, Wen Q, Qiu Y, 2015, A method for establishing human primary gastric epithelial cell culture from fresh surgical gastric tissues. Mol Med Rep 12(2):2939–2944 Gomes C, Almeida A, Ferreira JA, Silva L, Santos-Sousa H, Pintode- Sousa J, Santos LL, Amado F, Schwientek T, Levery SB, Mandel U, Clausen H, David L, Reis CA, Osorio H, 2013, Glycoproteomics analysis of serum from patients with gastric precancerous lesions. J Proteome Res 12:1454–1466 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 173 EP 179 DI 10.1007/s12253-016-0122-1 PG 7 ER PT J AU Ide, M Koba, Sh Sueoka-Aragane, N Sato, A Nagano, Y Inoue, T Misago, N Narisawa, Y Kimura, Sh Sueoka, E AF Ide, Masaru Koba, Shinichi Sueoka-Aragane, Naoko Sato, Akemi Nagano, Yuri Inoue, Takuya Misago, Noriyuki Narisawa, Yutaka Kimura, Shinya Sueoka, Eisaburo TI Mutation Profile of B-Raf Gene Analyzed by fully Automated System and Clinical Features in Japanese Melanoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE B-raf; Melanoma; Molecular targeted therapy; Tyrosine kinase inhibitors ID B-raf; Melanoma; Molecular targeted therapy; Tyrosine kinase inhibitors AB BRAF gene mutations have been observed in 30–50 % of malignant melanoma patients. Recent development of therapeutic intervention using BRAF inhibitors requires an accurate and rapid detection system for BRAF mutations. In addition, the clinical characteristics of the melanoma associated with BRAF mutations in Japanese patients have not been investigated on a large scale evaluation. We recently established quenching probe system (QP) for detection of an activating BRAF mutation, V600E and evaluated 113 melanoma samples diagnosed in Saga University Hospital from 1982 to 2011. The QP system includes fully automated genotyping, based on analysis of the probe DNA melting curve, which binds the target mutated site using a fluorescent guanine quenched probe. BRAF mutations were detected in 54 of 115 (47 %) including 51 of V600E and 3 of V600 K in Japanese melanoma cases. Among clinical subtypes of melanoma, nodular melanoma showed high frequency (12 of 15; 80 %) of mutation followed by superficial spreading melanoma (13 of 26; 50 %). The QP system is a simple and sensitive method to determine BRAF V600E mutation, and will be useful tool for patient-oriented therapy with BRAF inhibitors. C1 [Ide, Masaru] Saga University, Faculty of Medicine, Department of Internal MedicineSaga, Japan. [Koba, Shinichi] Saga University, Faculty of Medicine, Department of DermatologySaga, Japan. [Sueoka-Aragane, Naoko] Saga University, Faculty of Medicine, Department of Internal MedicineSaga, Japan. [Sato, Akemi] Saga University, Faculty of Medicine, Department of Internal MedicineSaga, Japan. [Nagano, Yuri] Saga University, Faculty of Medicine, Department of Internal MedicineSaga, Japan. [Inoue, Takuya] Saga University, Faculty of Medicine, Department of DermatologySaga, Japan. [Misago, Noriyuki] Saga University, Faculty of Medicine, Department of DermatologySaga, Japan. [Narisawa, Yutaka] Saga University, Faculty of Medicine, Department of DermatologySaga, Japan. [Kimura, Shinya] Saga University, Faculty of Medicine, Department of Internal MedicineSaga, Japan. [Sueoka, Eisaburo] Saga University, Faculty of Medicine, Department of Clinical Laboratory Medicine, 849-8501 Saga, Japan. RP Sueoka, E (reprint author), Saga University, Faculty of Medicine, Department of Clinical Laboratory Medicine, 849-8501 Saga, Japan. EM sueokae@cc.saga-u.ac.jp CR Balch CM, Gershenwald JE, Soong SJ, Thompson JF, Atkins MB, Byrd DR, Buzaid AC, Cochran AJ, Coit DG, Ding S, Eggermont AM, Flaherty KT, Gimotty PA, Kirkwood JM, McMasters KM, Mihm MC Jr, Morton DL, Ross MI, Sober AJ, Sondak VK, 2009, Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 27:6199–6206 Katanoda K, Hori M, Matsuda T, Shibata A, Nishino Y, Hattori M, Soda M, Ioka A, Sobue T, Nishimoto H, 2015 , An updated report on the trends in cancer incidence and mortality in Japan, 1958- 2013. Jpn J Clin Oncol 45(4):390–401. Chapman PB, Einhorn LH, Meyer ML, Saxman S, Destro AN, Panageas KS, Begg CB, Agarwala SS, Schuchter LM, Ernstoff MS, Houghton AN, Kirkwood JM, 1999, Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol 17:2745–2751 MiddletonMR, Grob JJ, Aaronson N, Fierlbeck G, TilgenW, Seiter S, Gore M, Aamdal S, Cebon J, Coates A, Dreno B, Henz M, Schadendorf D, Kapp A, Weiss J, Fraass U, Statkevich P, Muller M, Thatcher N, 2000, Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma. J Clin Oncol 18:158–166 Avril MF, Aamdal S, Grob JJ, Hauschild A, Mohr P, Bonerandi JJ, WeichenthalM, Neuber K, Bieber T, Gilde K, Guillem Porta V, Fra J, Bonneterre J, Saiag P, Kamanabrou D, Pehamberger H, Sufliarsky J, Gonzalez Larriba JL, Scherrer A, Menu Y, 2004, Fotemustine compared with dacarbazine in patients with disseminated malignant melanoma: a phase III study. J Clin Oncol 22(6): 1118–1125 Bedikian AY, Millward M, Pehamberger H, Conry R, Gore M, Trefzer U, Pavlick AC, DeConti R, Hersh EM, Hersey P, Kirkwood JM, Haluska FG, 2006, Oblimersen melanoma study group. Bcl-2 antisense, oblimersen sodium, plus dacarbazine in patients with advanced melanoma: the oblimersen melanoma study group. J Clin Oncol 24(29):4738–4745 Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI,Wolchok JD,Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ, 2010, Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363(8):711– 723 Robert C, Thomas L, Bondarenko I, O’Day S, Weber J, Garbe C, Lebbe C, Baurain JF, Testori A, Grob JJ, Davidson N, Richards J, Maio M, Hauschild A, Miller WH Jr, Gascon P, Lotem M, Harmankaya K, Ibrahim R, Francis S, Chen TT, Humphrey R, Hoos A,Wolchok JD, 2011, Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 364(26):2517– 2526 Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, Hogg D, Lorigan P, Lebbe C, Jouary T, Schadendorf D, Ribas A, O’Day SJ, Sosman JA, Kirkwood JM, Eggermont AM, Dreno B, Nolop K, Li J, Nelson B, Hou J, Lee RJ, Flaherty KT, McArthur GA, BRIM-3 Study Group, 2011, Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 364(26):2507–2516 Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA, 2002, Mutations of the BRAF gene in human cancer. Nature 417(6892):949–954 Curtin JA, Fridlyand J, Kageshita T, Patel HN, Busam KJ, Kutzner H, Cho KH, Aiba S, Brocker EB, LeBoit PE, Pinkel D, Bastian BC, 2005, Distinct sets of genetic alterations in melanoma. N Engl J Med 353(20):2135–2147 Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, Hamid O, Schuchter L, Cebon J, Ibrahim N, Kudchadkar R, Burris HA 3rd, Falchook G, Algazi A, Lewis K, Long GV, Puzanov I, Lebowitz P, Singh A, Little S, Sun P, Allred A, Ouellet D, Kim KB, Patel K, Weber J, 2012, Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med 367(18):1694–1703 Klein O, Clements A, Menzies AM, O’Toole S, Kefford RF, Long GV, 2013, BRAF inhibitor activity in V600R metastatic melanoma. Eur J Cancer 49(7):1797–1798 Ashida A, Uhara H, Kiniwa Y, Oguchi M, Murata H, Goto Y, Uchiyama A, Ogawa E, Hayashi K, Koga H, Okuyama R, 2012, Assessment of BRAF and KIT mutations in Japanese melanoma patients. J Dermatol Sci 66(3):240–242 Edmunds SC, Cree IA, Di Nicolantonio F, Hungerford JL, Hurren JS, Kelsell DP, 2003, Absence of BRAF gene mutations in uveal melanomas in contrast to cutaneous melanomas. Br J Cancer 88(9): 1403–1405 Tanaka R, Kuroda J, StevensonW, Ashihara E, Ishikawa T, Taki T, Kobayashi Y, Kamitsuji Y, Kawata E, Takeuchi M, Murotani Y, Yokota A, Hirai M, Majima S, Taniwaki M, Maekawa T, Kimura S, 2008, Fully automated and super-rapid system for the detection of JAK2V617F mutation. Leuk Res 32(9):1462–1467 Tiacci E, Trifonov V, Schiavoni G, Holmes A, Kern W, Martelli MP, Pucciarini A, Bigerna B, Pacini R, Wells VA, Sportoletti P, Pettirossi V, Mannucci R, Elliott O, Liso A, Ambrosetti A, Pulsoni A, Forconi F, Trentin L, Semenzato G, Inghirami G, Capponi M, Di Raimondo F, Patti C, Arcaini L, Musto P, Pileri S, Haferlach C, Schnittger S, Pizzolo G, Foa R, Farinelli L, Haferlach T, Pasqualucci L, Rabadan R, Falini B, 2011, BRAF mutations in hairy-cell leukemia. N Engl J Med 364(24):2305–2315 Pollock PM, Harper UL, Hansen KS, Yudt LM, Stark M, Robbins CM, Moses TY, Hostetter G, Wagner U, Kakareka J, Salem G, Pohida T, Heenan P, Duray P, Kallioniemi O, Hayward NK, Trent JM,Meltzer PS, 2003, High frequency of BRAF mutations in nevi. Nat Genet 33(1):19–20 Yuen ST, Davies H, Chan TL, Ho JW, Bignell GR, Cox C, Stephens P, Edkins S, TsuiWW, Chan AS, Futreal PA, StrattonMR,Wooster R, Leung SY, 2002, Similarity of the phenotypic patterns associated with BRAF and KRAS mutations in colorectal neoplasia. Cancer Res 62(22):6451–6455 Brose MS, Volpe P, Feldman M, Kumar M, Rishi I, Gerrero R, Einhorn E, Herlyn M, Minna J, Nicholson A, Roth JA, Albelda SM, Davies H, Cox C, Brignell G, Stephens P, Futreal PA, Wooster R, Stratton MR,Weber BL, 2002, BRAF and RAS mutations in human lung cancer and melanoma. Cancer Res 62(23): 6997–7000 Weber A, Langhanki L, Sommerer F, Markwarth A, Wittekind C, Tannapfel A, 2003, Mutations of the BRAF gene in squamous cell carcinoma of the head and neck. Oncogene 22(30):4757–4759 Lee JH, Choi JW, Kim YS, 2011, Frequencies of BRAF and NRAS mutations are different in histological types and sites of origin of cutaneousmelanoma: ameta-analysis. Br J Dermato 164(4):776–784 Ugurel S, Thirumaran RK, Bloethner S, Gast A, Sucker A,Mueller- Berghaus J, Rittgen W, Hemminki K, Becker JC, Kumar R, Schadendorf D, 2007, B-RAF and N-RAS mutations are preserved during short time in vitro propagation and differentially impact prognosis. PLoS One 2(2):e236 Long GV, Menzies AM, Nagrial AM, Haydu LE, Hamilton AL, Mann GJ, Hughes TM, Thompson JF, Scolyer RA, Kefford RF, 2011, Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol 29(10):1239–1246 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 181 EP 188 DI 10.1007/s12253-016-0121-2 PG 8 ER PT J AU Park, YJ Yoon, G AF Park, Y Ji Yoon, GhilSuk TI Overexpression of Aquaporin-1 is a Prognostic Factor for Biochemical Recurrence in Prostate Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Aquaporin; Prostate adenocarcinoma; Biochemical recurrence; Gleason score ID Aquaporin; Prostate adenocarcinoma; Biochemical recurrence; Gleason score AB Aquaporins (AQP) are transmembrane proteins that provide channels for water and solutes, and some are involved in tumor progression and invasion. We evaluated the expression of AQP-1, AQP-3, and AQP-5 and their clinicopathological significance in prostate adenocarcinomas (PCA). Prostatectomy specimens (n = 99) were retrieved from the surgical pathology archives and clinicopathological data were obtained from the medical database at Kyungpook National University Hospital. Immunohistochemical staining for AQP- 1, AQP-3, and AQP-5 was performed on tissue microarrays comprising paired malignant and benign prostatic tissues. Seventeen PCA cases (17.2 %) showed AQP-1 overexpression, specifically 7 tumors (9.7 %) with lower Gleason scores (GS) and 10 tumors (37.0 %) with higher GS, with statistical significance (P = 0.001). AQP-1 overexpression was significantly associated with higher GS (P = 0.001), higher pathologic T (pT) stages (P = 0.024), and biochemical recurrence (BR) (P = 0.002). The difference in AQP-3 and AQP-5 expression between neoplastic and non-neoplastic tissues was not established and there were no correlations with clinicopathological parameters. AQP-1 overexpression was evident in tumors with higher GS, it was less evident in tumors with lower GS, and it was associated with BR and a higher pT stage. AQP-1 overexpression is associated with prostate cancer progression. C1 [Park, Y Ji] Catholic University of Daegu, School of Medicine, Department of Pathology, Duryugongwon-ro 17-gil, Nam-gu, 42472 Daegu, South Korea. [Yoon, GhilSuk] Kyungpook National University School of Medicine, Kyungpook National University Medical Center, Department of PathologyDaegu, South Korea. RP Park, YJ (reprint author), Catholic University of Daegu, School of Medicine, Department of Pathology, 42472 Daegu, South Korea. EM pathpjy@naver.com CR Ferlay J, Autier P, Boniol M, Heanue M, Colombet M, Boyle P, 2007, Estimates of the cancer incidence and mortality in Europe in 2006. Ann Oncol 18:581–592 Perner S, Mosquera JM, Demichelis F, HoferMD, Paris PL, Simko J, Collins C, Bismar TA, Chinnaiyan AM, De Marzo AM, Rubin MA, 2007, TMPRSS2-ERG fusion prostate cancer: an early molecular event associated with invasion. Am J Surg Pathol 31:882–888 Sales AD, Lobo CH, Carvalho AA, Moura AA, Rodrigues AP, 2013, Structure, function, and localization of aquaporins: their possible implications on gamete cryopreservation. Genet Mol Res 12:6718–6732 Harris HW Jr, Zeidel ML, 1993, Water channels. Curr Opin Nephrol Hypertens 2:699–707 Yoshida T, Hojo S, Sekine S, Sawada S, Okumura T, Nagata T, Shimada Y, Tsukada K, 2013, Expression of aquaporin-1 is a poor prognostic factor for stage II and III colon cancer. Mol Clin Oncol 1:953–958 Moon C, Soria JC, Jang SJ, Lee J, Obaidul Hoque M, Sibony M, Trink B, Chang YS, Sidransky D, Mao L, 2003, Involvement of aquaporins in colorectal carcinogenesis. Oncogene 22:6699–6703 Otto W, Rubenwolf PC, Burger M, Fritsche HM, Rossler W, May M, Hartmann A, Hofstadter F, Wieland WF, Denzinger S, 2012, Loss of aquaporin 3 protein expression constitutes an independent prognostic factor for progression-free survival: an immunohistochemical study on stage pT1 urothelial bladder cancer. BMC Cancer 12:459–467 Machida Y, Ueda Y, Shimasaki M, Sato K, Sagawa M, Katsuda S, Sakuma T, 2011, Relationship of aquaporin 1, 3, and 5 expression in lung cancer cells to cellular differentiation, invasive growth, and metastasis potential. Hum Pathol 42(5):669–678 Verkman AS, Hara-Chikuma M, Papadopoulos MC, 2008, Aquaporins–new players in cancer biology. J Mol Med, Berl, 86(5):523–529 Saadoun S, Papadopoulos MC, Hara-Chikuma M, Verkman AS, 2005, Impairment of angiogenesis and cell migration by targeted aquaporin-1 gene disruption. Nature 434:786–792 Woo J, Lee J, Chae YK, Kim MS, Baek JH, Park JC, Park MJ, Smith IM, Trink B, Ratovitski E, Lee T, Park B, Jang SJ, Soria JC, Califano JA, Sidransky D, Moon C, 2008, Overexpression of AQP5, a putative oncogene, promotes cell growth and transformation. Cancer Lett 264:54–62 Monzani E, Bazzotti R, Perego C, La Porta CA, 2009, AQP1 is not only a water channel: it contributes to cell migration through Lin7/beta-catenin. PLoS One 4(7):e6167 Aishima S, Kuroda Y, Nishihara Y, Taguchi K, Iguchi T, Taketomi A, Maehara Y, Tsuneyoshi M, 2007, Down-regulation of aquaporin-1 in intrahepatic cholangiocarcinoma is related to tumor progression and mucin expression. Hum Pathol 38(12):1819–1825 Yang JH, Shi YF, Chen XD, Qi WJ, 2006, The influence of aquaporin-1 and microvessel density on ovarian carcinogenesis and ascites formation. Int J Gynecol Cancer 16(Suppl 1):400–405 Shi Z, Zhang T, Luo L, Zhao H, Cheng J, Xiang J, Zhao C, 2012, Aquaporins in human breast cancer: identification and involvement in carcinogenesis of breast cancer. J Surg Oncol 106(3):267–272 Hwang I, Jung SI, Hwang EC, Song SH, Lee HS, Kim SO, Kang TW, Kwon D, Park K, 2012, Expression and localization of aquaporins in benign prostate hyperplasia and prostate cancer. Chonnam Med J 48(3):174–178 Wang J, Tanji N, Sasaki T, Kikugawa T, Song X, Yokoyama M, 2008, Androgens upregulate aquaporin 9 expression in the prostate. Int J Urol 15(10):936–941 Pan XY, Guo H, Han J, Hao F, An Y, Xu Y, Xiaokaiti Y, Pan Y, Li XJ, 2012, Ginsenoside Rg3 attenuates cell migration via inhibition of aquaporin 1 expression in PC-3M prostate cancer cells. Eur J Pharmacol 683:27–34 Wang J, Tanji N, Kikugawa T, Shudou M, Song X, Yokoyama M, 2007, Expression of aquaporin 3 in the human prostate. Int J Urol 14:1088–1092 Wang J, Cai Y, Ren C, Ittmann M, 2006, Expression of variant TMPRSS2/ERG fusionmessenger RNAs is associated with aggressive prostate cancer. Cancer Res 66(17):8347–8351 Pirker R, Pereira JR, Pawel J, Krzakowski M, Ramlau R, Park K, Marinis F, Eberhardt W, Paz-Ares L, Storkel S, Schumacher KM, Heydebreck A, Celik I, O’Byrne KJ, 2012, EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: analysis of data from the phase 3 FLEX study. Lancet Oncol 13(1):33–42 Jemal A, Siegel R, Xu J,Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60(5):277–300 Bartholow TL, Becich MJ, Chandran UR, Parwani AV, 2011, Immunohistochemical analysis of ezrin-radixin-moesin-binding phosphoprotein 50 in prostatic adenocarcinoma. BMC Urol 11:12–20 Abascal F, Irisarri I, Zardoya R, 2014, Diversity and evolution of membrane intrinsic proteins. Biochim Biophys Acta 1840(5):1468– 1481 Tan M, Shao C, Bishop JA, Feng Z, Trock BJ,Westra WH, Ha PK, 2014, Aquaporin-1 promoter hypermethylation is associated with improved prognosis in salivary gland adenoid cystic carcinoma. Otolaryngol Head Neck Surg 150(5):801–807 King LS, Agre P, 1996, Pathophysiology of the aquaporin water channels. Annu Rev Physiol 58:619–648 Nico B, Ribatti D, 2010, Aquaporins in tumor growth and angiogenesis. Cancer Lett 294(2):135–138 Hu J, Verkman AS, 2006, Increased migration and metastatic potential of tumor cells expressing aquaporin water channels. FASEB J 20(11):1892–1894 Papadopoulos MC, Saadoun S, Verkman AS, 2008, Aquaporins and cell migration. Pflugers Arch 456(4):693–700 Shao C, Sun W, Tan M, Glazer CA, Bhan S, Zhong X, Fakhry C, Sharma R, Westra WH, Hoque MO, Moskaluk CA, Sidransky D, Califano JA, Ha PK, 2011, Integrated, genome-wide screening for hypomethylated oncogenes in salivary gland adenoid cystic carcinoma. Clin Cancer Res 17(13):4320–4330 Clapp C, Martinez de la Escalera G, 2006, Aquaporin-1: a novel promoter of tumor angiogenesis. Trends Endocrinol Metab 17(1):1–2 Huang Y, Murakami T, Sano F, Kondo K, Nakaigawa N, Kishida T, Kubota Y, Nagashima Y, Yao M, 2009, Expression of aquaporin 1 in primary renal tumors: a prognostic indicator for clear-cell renal cell carcinoma. Eur Urol 56(4):690–698 Otterbach F, Callies R, Adamzik M, Kimmig R, Siffert W, Schmid KW, Bankfalvi A, 2010, Aquaporin 1, AQP1, expression is a novel characteristic feature of a particularly aggressive subgroup of basallike breast carcinomas. Breast Cancer Res Treat 120(1):67–76 Ribatti D, Ranieri G, Annese T, Nico B, 2014, Aquaporins in cancer. Biochim Biophys Acta 1840(5):1550–1553 Liu J, Zhang WY, Ding DG, 2015, Expression of aquaporin 1 in bladder uroepithelial cell carcinoma and its relevance to recurrence. Asian Pac J Cancer Prev 16(9):3973–3976 Stigliano C, Aryal S, de Tullio DM, Nicchia G, Pascazio G, Svelto M, Decuzzi P, 2013, siRNA-chitosan complexes in poly(lactic-coglycolic acid, nanoparticles for the silencing of aquaporin-1 in cancer cells. Mol Pharm 10(8):3186–3194 Bin K, Shi-Peng Z, 2011, Acetazolamide inhibits aquaporin-1 expression and colon cancer xenograft tumor growth. Hepatogastroenterology 58:1502–1506 Chae YK, Woo J, Kim MJ, Kang SK, Kim MS, Lee J, Lee SK, Gong G, Kim YH, Soria JC, Jang SJ, Sidransky D,Moon C, 2008, Expression of aquaporin 5, AQP5, promotes tumor invasion in human non small cell lung cancer. PLoS One 3(5):e2162 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 189 EP 196 DI 10.1007/s12253-016-0145-7 PG 8 ER PT J AU Shrestha, B Bajracharya, D Byatnal, AA Kamath, A Radhakrishnan, R AF Shrestha, Bijayatha Bajracharya, Dipshikha Byatnal, Amit Aditi Kamath, Asha Radhakrishnan, Raghu TI May High MMP-2 and TIMP-2 Expressions Increase or Decrease the Aggressivity of Oral Cancer? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral squamous cell carcinoma; Invasion; Matrix metalloproteinases; Metastasis; MMP-2; TIMP-2 ID Oral squamous cell carcinoma; Invasion; Matrix metalloproteinases; Metastasis; MMP-2; TIMP-2 AB Matrix metalloproteinases-2 (MMP-2) and the tissue inhibitor of metalloproteinase-2 (TIMP-2), may presumably have an important role on the invasion and metastatic spread of malignancies attributed to an uncontrolled degradation of the extracellular matrix (ECM). A retrospective chart analysis was carried out to study the expression of MMP-2 and TIMP-2 on the archival samples of oral squamous cell carcinoma (OSCC) (n = 30) and normal mucosa (n = 10) by immunohistochemistry and compared with the clinicopathologic parameters of cases. Both MMP-2 and TIMP-2 expressions showed a positive correlation with the grades, stages and metastatic capacities of tumors (Spearman’s correlation, p < 0.05). Concomitant increase in the expression of TIMP- 2 and MMP-2 suggested that the rate of MMP-2/TIMP-2 expression is a better marker for characterization of MMP-2 concentration. High expression and/or activity of MMP-2 were linked with poorer survival in OSCC cases, while TIMPs have been shown to apparently act as either growthstimulating or suppressor factors for tumors. It was also revealed that MMP-2 and TIMP-2 were secreted by both tumor cells and stromal cells. A new concept, supposing the dynamic, anticancer partnership between the residual genome stabilizer machinery of tumor cells and defensive cells adjacent to tumors, may illuminate the controversial results. In conclusion, the stronger the infiltrative and metastatic capacity of cancers, the higher is the rate of MMP-2/TIMP-2 expression helping the arrival of humoral and cellular anticancer forces. C1 [Shrestha, Bijayatha] Chitwan Medical College, Department of Oral PathologyBharatpur, Chitwan, Nepal. [Bajracharya, Dipshikha] Hospital and Research Center, Kantipur Dental College, Department of Oral PathologyKathmandu, Nepal. [Byatnal, Amit Aditi] Oral Pathology and MicrobiologyHubli, Karnataka, India. [Kamath, Asha] Manipal University, Kasturba Medical College, Department of Community MedicineManipal, India. [Radhakrishnan, Raghu] Manipal University, Manipal College of Dental Sciences, Department of Oral Pathology and Microbiology, 576104 Manipal, India. RP Radhakrishnan, R (reprint author), Manipal University, Manipal College of Dental Sciences, Department of Oral Pathology and Microbiology, 576104 Manipal, India. EM raghu.radhakrishnan@gmail.com CR Nagase H,Woessner JF Jr, 1999, Matrix metalloproteinases. J Biol Chem 274:21491–21494 de Vicente JC, Fresno MF, Villalain L et al, 2005, Expression and clinical significance of matrix metalloproteinase-2 and matrix metalloproteinase-9 in oral squamous cell carcinoma. Oral Oncol 41:283–293 Ruokolainen H, Paakko P, Turpeenniemi-Hujanen T, 2006, Tissue and circulating immunoreactive protein forMMP-2 and TIMP-2 in head and neck squamous cell carcinoma–tissue immunoreactivity predicts aggressive clinical course. Mod Pathol 19(208):217 Lambert E, Dasse E, Haye B et al, 2004, TIMPs as multifacial proteins. Crit Rev Oncol Hematol 49:187–198 GaiottoMA, Focchi J, Ribalta JL et al, 2004, Comparative study of MMP-2, matrix metalloproteinase 2, immune expression in normal uterine cervix, intraepithelial neoplasias, and squamous cells cervical carcinoma. Am J Obstet Gynecol 190:1278–1282 Nagase H, Visse R, Murphy G, 2006, Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res 69:562–573 Ra HJ, Parks WC, 2007, Control of matrix metalloproteinase catalytic activity. Matrix Biol 26:587–596 Gorogh T, Beier UH, Baumken J et al, 2006, Metalloproteinases and their inhibitors: influence on tumor invasiveness and metastasis formation in head and neck squamous cell carcinomas. Head Neck 28:31–39 Bhuvarahamurthy V, Kristiansen GO, Johannsen M et al, 2006, In situ gene expression and localization of metalloproteinases MMP1, MMP2, MMP3, MMP9, and their inhibitors TIMP1 and TIMP2 in human renal cell carcinoma. Oncol Rep 15:1379–1384 Shima I, Sasaguri Y, Kusukawa J et al, 1992, Production of matrix metalloproteinase-2 and metalloproteinase-3 related to malignant behavior of esophageal carcinoma. A clinicopathologic study. Cancer 70:2747–2753 Bindhu OS, Ramadas K, Sebastian P et al, 2006, High expression levels of nuclear factor kappa B and gelatinases in the tumorigenesis of oral squamous cell carcinoma. Head Neck 28:916–925 Kusukawa J, Sasaguri Y, Shima I et al, 1993, Expression of matrix metalloproteinase-2 related to lymph node metastasis of oral squamous cell carcinoma. A clinicopathologic study. Am J Clin Pathol 99:18–23 Imanishi Y, Fujii M, Tokumaru Yet al, 2000, Clinical significance of expression of membrane type 1 matrix metalloproteinase and matrix metalloproteinase-2 in human head and neck squamous cell carcinoma. Hum Pathol 31:895–904 Shimada T, Nakamura H, Yamashita K et al, 2000, Enhanced production and activation of progelatinase a mediated by membranetype 1 matrix metalloproteinase in human oral squamous cell carcinomas: implications for lymph node metastasis. Clin Exp Metastasis 18:179–188 O-Charoenrat P, Rhys-Evans PH, Eccles SA, 2001, Expression of matrix metalloproteinases and their inhibitors correlates with invasion and metastasis in squamous cell carcinoma of the head and neck. Arch Otolaryngol Head Neck Surg 127:813–820 Hu X, Li D, Zhang W et al, 2004, Matrix metalloproteinase-9 expression correlates with prognosis and involved in ovarian cancer cell invasion. Arch Gynecol Obstet 286:1537–1543 Gao ZB, Duan YQ, Zhang L, Chen DW, Ding PT, 2005, Expression of matrix metalloproteinase 2 and its tissue inhibitor in oral squamous cell carcinoma. Int J Mol Med 16:599–603 Basset P, Bellocq JP,Wolf C et al, 1990, A novel metalloproteinase gene specifically expressed in stromal cells of breast carcinomas. Nature 348:699–704 Bisson C, Blacher S, PoletteMet al, 2003, Restricted expression of membrane type 1-matrix metalloproteinase by myofibroblasts adjacent to human breast cancer cells. Int J Cancer 105:7–13 Pyke C, Ralfkiaer E, Huhtala P et al, 1992, Localization of messenger RNA for Mr 72,000 and 92,000 type IV collagenases in human skin cancers by in situ hybridization. Cancer Res 52: 1336–1341 Charous SJ, Stricklin GP, Nanney LB et al, 1997, Expression of matrix metalloproteinases and tissue inhibitor of metalloproteinases in head and neck squamous cell carcinoma. Ann Otol Rhinol Laryngol 106:271–278 Ondruschka C, Buhtz P, Motsch C et al, 2002, Prognostic value of MMP-2, −9 and TIMP-1,-2 immunoreactive protein at the invasive front in advanced head and neck squamous cell carcinomas. Pathol Res Pract 198:509–515 Franchi A, Santucci M, Masini E et al, 2002, Expression of matrix metalloproteinase 1, matrix metalloproteinase 2, and matrix metalloproteinase 9 in carcinoma of the head and neck. Cancer 95:1902– 1910 Monteagudo C, Merino MJ, San-Juan J et al, 1990, Immunohistochemical distribution of type IV collagenase in normal, benign, and malignant breast tissue. Am J Pathol 136:585– 592 Hong SD, Hong SP, Lee JI, Lim CY, 2000, Expression of matrix metalloproteinase-2 and -9 in oral squamous cell carcinomas with regard to the metastatic potential. Oral Oncol 36:207–213 Bramhall SR, Neoptolemos JP, Stamp GWet al, 1997, Imbalance of expression of matrix metalloproteinases, MMPs, and tissue inhibitors of the matrix metalloproteinases, TIMPs, in human pancreatic carcinoma. J Pathol 182:347–355 Foda HD, Zucker S, 2001, Matrix metalloproteinases in cancer invasion, metastasis and angiogenesis. Drug Discov Today 6:478– 482 Egeblad M, Werb Z, 2002, New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2:161–174 Kawamata H, Nakashiro K, Uchida D et al, 1997, Possible contribution of active MMP2 to lymph-node metastasis and secreted cathepsin L to bone invasion of newly established human oralsquamous- cancer cell lines. Int J Cancer 70:120–127 Polette M, Nawrocki-Raby B, Gilles C et al, 2004, Tumour invasion and matrix metalloproteinases. Crit Rev Oncol Hematol 49: 179–186 Overall CM, Wrana JL, Sodek J, 1989, Independent regulation of collagenase, 72-kDa progelatinase, and metalloendoproteinase inhibitor expression in human fibroblasts by transforming growth factor-beta. J Biol Chem 264:1860–1869 Munshi HG, Wu YI, Mukhopadhyay S et al, 2004, Differential regulation of membrane type 1-matrix metalloproteinase activity by ERK 1/2- and p38 MAPK-modulated tissue inhibitor of metalloproteinases 2 expression controls transforming growth factorbeta1- induced pericellularcollagenolysis. J Biol Chem 279: 39042–39050 Kato K, Hara A, Kuno T et al, 2005, Matrix metalloproteinases 2 and 9 in oral squamous cell carcinomas: manifestation and localization of their activity. J Cancer Res Clin Oncol 131:340–346 Visscher DW, Hoyhtya M, Ottosen SK et al, 1994, Enhanced expression of tissue inhibitor of metalloproteinase-2, TIMP-2, in the stroma of breast carcinomas correlates with tumor recurrence. Int J Cancer 59:339–344 Pickett KL, Harber GJ, DeCarlo AA et al, 1999, 92K-GL, MMP-9, and 72 K-GL, MMP-2, are produced in vivo by human oral squamous cell carcinomas and can enhance FIB-CL, MMP-1, activity in vitro. J Dent Res 78:1354–1361 Bjorklund M, Koivunen E, 2005, Gelatinase-mediated migration and invasion of cancer cells. Biochim Biophys Acta 1755:37–69 Danilewicz M, Sikorska B, Wagrowska-Danilewicz M, 2003, Prognostic significance of the immunoexpression of matrix metalloproteinase MMP2 and its inhibitor TIMP2 in laryngeal cancer. Med Sci Monit 9:MT427 LiuWW, Zeng ZY,Wu QL et al, 2005, Overexpression ofMMP-2 in laryngeal squamous cell carcinoma: a potential indicator for poor prognosis. Otolaryngol Head Neck Surg 132:395–400 Yoshizaki T, Maruyama Y, Sato H et al, 2001, Expression of tissue inhibitor of matrix metalloproteinase-2 correlates with activation of matrix metalloproteinase-2 and predicts poor prognosis in tongue squamous cell carcinoma. Int J Cancer 95:44–50 Verstappen J, Von den Hoff JW, 2006, Tissue inhibitors of metalloproteinases, TIMPs): their biological functions and involvement in oral disease. J Dent Res 85:1074–1084 Visscher DW, Hoyhtya M, Ottosen SK et al, 1994, Enhanced expression of tissue inhibitor of metalloproteinase-2, TIMP-2, in the stroma of breast carcinomas correlates with tumor recurrence. Int J Cancer 59:339–344 Katayama A, Bandoh N, Kishibe K et al, 2004, Expressions of matrix metalloproteinases in early-stage oral squamous cell carcinoma as predictive indicators for tumor metastases and prognosis. Clin Cancer Res 10:634–640 Kikuchi R, Noguchi T, Takeno S et al, 2000, Immunohistochemical detection of membrane-type-1-matrix metalloproteinase in colorectal carcinoma. Br J Cancer 83:215–218 Hayakawa T, Yamashita K, Tanzawa K et al, 1992, Growthpromoting activity of tissue inhibitor of metalloproteinases-1, TIMP-1, for a wide range of cells. A possible new growth factor in serum. FEBS Lett 298:29–32 Ikenaka Y, Yoshiji H, Kuriyama S et al, 2003, Tissue inhibitor of metalloproteinases-1, TIMP-1, inhibits tumor growth and angiogenesis in the TIMP-1 transgenic mouse model. Int J Cancer 105: 340–346 Visse R, Nagase H, 2003, Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ Res 92:827–839 Hernandez-Barrantes S, Bernardo M, Toth M et al, 2002, Regulation of membrane type-matrix metalloproteinases. Semin Cancer Biol 12:131–138 Brew K, Dinakarpandian D, Nagase H, 2000, Tissue inhibitors of metalloproteinases: evolution, structure and function. Biochim Biophys Acta 1477:267–283 Kugler A, Hemmerlein B, Thelen P et al, 1998, Expression of metalloproteinase 2 and 9 and their inhibitors in renal cell carcinoma. J Urol 160:1914–1918 Kallakury BV, Karikehalli S, Haholu A et al, 2001, Increased expression of matrix metalloproteinases 2 and 9 and tissue inhibitors of metalloproteinases 1 and 2 correlate with poor prognostic variables in renal cell carcinoma. Clin Cancer Res 7:3113–3119 Di Nezza LA, Misajon A, Zhang J et al, 2002, Presence of active gelatinases in endometrial carcinoma and correlation of matrix metalloproteinase expression with increasing tumor grade and invasion. Cancer 94:1466–1475 Baker EA, Leaper DJ, Hayter JP et al, 2006, The matrix metalloproteinase system in oral squamous cell carcinoma. Br J Oral Maxillofac Surg 44:482–486 Suba Z, 2015, DNA-stabilization by the upregulation of estrogen signaling in BRCA gene mutation carriers. Drug Des Devel Ther 9: 2663–2675 Schiewer MJ, Knudsen KE, 2016, Linking DNA damage and hormone signaling pathways in cancer. Trends Endocrinol Metab 27: 216–225 Suba Z, 2016, Causal therapy of breast cancer irrelevant of age, tumor stage and ER-status. Recent Pat Anticancer Drug Discov 11: 254–266 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 197 EP 206 DI 10.1007/s12253-016-0149-3 PG 10 ER PT J AU Gaal, Zs Olah, Rejto, L Balint, LB Csernoch, L AF Gaal, Zsuzsanna Olah, Eva Rejto, Laszlo Balint, Laszlo Balint Csernoch, Laszlo TI Expression Levels of Warburg-Effect Related microRNAs Correlate with each Other and that of Histone Deacetylase Enzymes in Adult Hematological Malignancies with Emphasis on Acute Myeloid Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute myeloid leukemia; Warburg-effect; microRNA; oncomiR; Anti-oncomiR; Histone deacetylase ID Acute myeloid leukemia; Warburg-effect; microRNA; oncomiR; Anti-oncomiR; Histone deacetylase AB Disruption of epigenetic regulation and characteristic metabolic alterations (known as the Warburg-effect) are well-known hallmarks of cancer. In our study we investigated the expression levels of microRNAs and histone deacetylase enzymes via RT-qPCR in bone marrow specimens of adult patients suffering from hematological malignancies (total cohort n = 40), especially acute myeloid leukemia (n = 27). The levels of the three examined Warburg-effect related microRNAs (miR-378*, miR-23b, miR-26a) positively correlated with each other and the oncogenic miR-155 and miR- 125b, while negatively with the level of the tumorsuppressor miR-124. Significant relationships have been confirmed between the levels of SIRT6, HDAC4 and the microRNAs listed above. In NPM1-mutated AML (n = 6), the level of miR-125b was significantly lower than in the group of AML patients not carrying this mutation (n = 13) (p < 0.05). In M5 FAB type of AML (n = 5), the level of miR-124 was significantly higher compared to the M2 group (n = 7) (p < 0.05). In two cases of FAB M5 AML, the levels of SIRT6 and miR-26a increased during the first 4 weeks of treatment. In the total cohort, white blood cell count at the time of the diagnosis significantly correlated with the levels of HDAC4, SIRT6, miR-124 and miR- 26a. Our results suggest that Warburg-effect related microRNAs may have important role in the pathogenesis of leukemia, and the potential oncogenic property of HDAC4 and SIRT6 cannot be excluded in hematological malignancies. Elevated level of miR-125b can contribute to adverse prognosis of AML without NPM1 mutation. The prevailment of the tumorsuppressor property of miR-124 may depend on the accompanying genetic alterations. C1 [Gaal, Zsuzsanna] University of Debrecen, Faculty of Medicine, Department of Physiology, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. [Olah, Eva] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. [Rejto, Laszlo] University of Debrecen, Faculty of Medicine, Department of Internal Medicine, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. [Balint, Laszlo Balint] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular Biology, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. [Csernoch, Laszlo] University of Debrecen, Faculty of Medicine, Department of Physiology, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. RP Csernoch, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Physiology, H-4012 Debrecen, Hungary. EM csl@edu.unideb.hu CR Hanahan D, Weinberg RA, 2000, The hallmarks of cancer. Cell 100(1):57–70 Brait M, Sidransky D. Cancer Epigenetics: above and beyond. Toxicol Mech Methods 2011;21(4):275–288. Jin LH, Wei C, 2014, Role of microRNAs in the Warburg effect and mitochondrial metabolism in cancer. Asian Pac J Cancer Prev 15(17):7015–7019 Locasale JW, Cantley LC, 2011, Metabolic flux and the regulation of mammalian cell growth. Cell Metab 14(4):443–451 Zhao S, Xu W, Jiang Wet al, 2010, Regulation of cellular metabolism by protein lysine acetylation. Science 327(5968):1000–1004 Ward PS, Thompson CB, 2012, Metabolic reprogramming: a cancer hallmark evenWarburg did not anticipate. Cancer Cell 21(3):297–308 Havelange V, Garzon R, 2010, MicroRNAs: emerging key regulators of hematopoiesis. Am J Hematol 85(12):935–942 Spizzo R, Nicoloso MS, Croce CM, Calin GA, 2009, SnapShot: microRNAs in cancer. Cell 137(3):586–586 Marcucci G, Mrozek K, Radmacher MD et al, 2011, The prognostic and functional role of microRNAs in acute myeloid leukemia. Blood 117(4):1121–1129 Schotte D, De Menezes RX, Akbari Moqadam F et al, 2011, MicroRNA characterize genetic diversity and drug resistance in pediatric acute lymphoblastic leukemia. Haematologica 96(5):703–711 Agirre X, Vilas-Zornoza A, Jimenez-Velasco A et al, 2009, Epigenetic silencing of the tumor suppressor microRNA hsa-miR- 124a regulates CDK6 expression and confers a poor prognosis in acute lymphoblastic leukemia. Cancer Res 69(10):4443–4453 Khalaj M, Tavakkoli M, Stranahan AW, Park CY, 2014, Pathogenic microRNA's in myeloid malignancies. Front Genet 5: 361., DOI 10.3389/fgene.2014.00361 Eichner LJ, Perry MC, Dufour CR et al, 2010, miR-378(∗, mediates metabolic shift in breast cancer cells via the PGC-1β/ERRγ transcriptional pathway. Cell Metab 12(4):352–361 Gao P, Tchernyshyov I, Chang TC et al, 2009, C-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature 458(7239):762–765 Chen B, Liu Y, Jin X et al, 2014, MicroRNA-26a regulates glucose metabolism by direct targeting PDHX in colorectal cancer cells. BMC Cancer 14:443., DOI 10.1186/1471-2407-14-443 Sebastian C, Zwaans BM, Silberman DM et al, 2012, The histone deacetylase SIRT6 is a tumor suppressor that controls cancer metabolism. Cell 151(6):1185–1199 Zhong L, D'Urso A, Toiber D et al, 2010, The histone deacetylase Sirt6 regulates glucose homeostasis via Hif1alpha. Cell 140(2): 280–293 Zhong L, Mostoslavsky R, 2010, SIRT6: a master epigenetic gatekeeper of glucose metabolism. Transcription 1(1):17–21 Shan C, Elf S, Ji Q et al, 2014, Lysine acetylation activates 6- phosphogluconate dehydrogenase to promote tumor growth. Mol Cell 55(4):552–565 Czimmerer Z, Hulvely J, Simandi Z et al, 2013, Aversatile method to design stem-loop primer-based quantitative PCR assays for detecting small regulatory RNA molecules. PLoS One 8(1):e55168., DOI 10.1371/journal.pone.0055168 Zhu YD, Wang L, Sun C et al, 2012, Distinctive microRNA signature is associated with the diagnosis and prognosis of acute leukemia. Med Oncol 29(4):2323–2331 Jiang W, Min J, Sui X et al, 2015, MicroRNA-26a-5p and microRNA-23b-3p up-regulate peroxiredoxin III in acute myeloid leukemia. Leuk Lymphoma 56(2):460–471 Jiang S, Zhang LF, Zhang HWet al, 2012, A novel miR-155/miR- 143 cascade controls glycolysis by regulating hexokinase 2 in breast cancer cells. EMBO J 31(8):1985–1998 Chen L, Shi Y, Liu S et al, 2014, PKM2: the thread linking energy metabolism reprogramming with epigenetics in cancer. Int J Mol Sci 15(7):11435–11445 Busch S, Auth E, Scholl F et al, 2015, 5-lipoxygenase is a direct target of miR-19a-3p and miR-125b-5p. J Immunol 194(4):1646– 1653 Etchegaray JP, Chavez L, Huang Y et al, 2015, The histone deacetylase SIRT6 controls embryonic stem cell fate via TETmediated production of 5-hydroxymethylcytosine. Nat Cell Biol 17(5):545–557 Nakajima H, Kunimoto H, 2014, TET2 as an epigenetic master regulator for normal and malignant hematopoiesis. Cancer Sci 105(9):1093–1099 Cheng J, Guo S, Chen S et al, 2013, An extensive network of TET2-targeting microRNAs regulates malignant hematopoiesis. Cell Rep 5(2):471–481 Wang JC, Kafeel MI, Avezbakiyev B et al, 2011, Histone deacetylase in chronic lymphocytic leukemia. Oncology 81(5–6): 325–329 Zhou J, Bi C, ChngWJ et al, 2011, PRL-3, a metastasis associated tyrosine phosphatase, is involved in FLT3-ITD signaling and implicated in anti-AML therapy. PLoS One 6(5):e19798 Hirsch P, Qassa G, Marzac C et al, 2015, Acute myeloid leukemia in patients older than 75: prognostic impact of FLT3-ITD and NPM1 mutations. Leuk Lymphoma 56(1):147–150 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 207 EP 216 DI 10.1007/s12253-016-0151-9 PG 10 ER PT J AU Liu, P Zu, X Liu, L AF Liu, Peihua Zu, Xiongbing Liu, Longfei TI Genetic Screening in Pheochromocytoma/Paraganglioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Liu, Peihua] Central South University, Xiangya hospital, Department of Urology, 410008 Changsha, Hunan, China. [Zu, Xiongbing] Central South University, Xiangya hospital, Department of Urology, 410008 Changsha, Hunan, China. [Liu, Longfei] Central South University, Xiangya hospital, Department of Urology, 410008 Changsha, Hunan, China. RP Liu, L (reprint author), Central South University, Xiangya hospital, Department of Urology, 410008 Changsha, China. EM longfei_liu@163.com CR Patocs A, Lendvai NK, Butz H, Liko I, Sapi Z, Szucs N et al, 2016, Novel SDHB and TMEM127 mutations in patients with pheochromocytoma/Paraganglioma syndrome. Pathol Oncol Res 22(4):673–679 Kolackov K, Tupikowski K, Bednarek-Tupikowska G, 2012, Genetic aspects of pheochromocytoma. Adv Clin Exp Med 21(6): 821–829official organ Wroclaw Medical University Jochmanova I, Zhuang Z, Pacak K, 2015, Pheochromocytoma: gasping for air. Horm Cancer 6(5–6):191–205 Santos P, Pimenta T, Taveira-Gomes A, 2014, Hereditary Pheochromocytoma. Int J Surg Pathol 22(5):393–400 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 217 EP 217 DI 10.1007/s12253-016-0117-y PG 1 ER PT J AU Jo, SY Kim, SM Yoo, JN Lee, HS AF Jo, Sol Yun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI USP9X, a Putative Tumor Suppressor Gene, Exhibits Frameshift Mutations in Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Schwickart M, Huang X, Lill JR, Liu J, Ferrando R, French DM, Maecker H, O’Rourke K, Bazan F, Eastham-Anderson J, Yue P, Dornan D, Huang DC, Dixit VM, 2010, Deubiquitinase USP9X stabilizesMCL1 and promotes tumour cell survival. Nature 463:103–107 Kushwaha D, O’Leary C, Cron KR, Deraska P, Zhu K, D’Andrea AD, Kozono D, 2015, USP9X inhibition promotes radiationinduced apoptosis in non-small cell lung cancer cells expressing mid-to-high MCL1. Cancer Biol Ther 16:392–401 Perez-Mancera PA, Rust AG, van der Weyden L, Kristiansen G, Li A, Sarver AL, Silverstein KA, Grutzmann R, Aust D, Rummele P, Knosel T, Herd C, Stemple DL, Kettleborough R, Brosnan JA, Li A, Morgan R, Knight S, Yu J, Stegeman S, Collier LS, ten Hoeve JJ, de Ridder J, Klein AP, Goggins M, Hruban RH, Chang DK, Biankin AV, Grimmond SM, Initiative APCG, Wessels LF, Wood SA, Iacobuzio-Donahue CA, Pilarsky C, Largaespada DA, Adams DJ, Tuveson DA, 2012, The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma. Nature 486:266–270 Oosterkamp HM, Hijmans EM, Brummelkamp TR, Canisius S, Wessels LF, Zwart W, Bernards R, 2014, USP9X downregulation renders breast cancer cells resistant to tamoxifen. Cancer Res 74: 3810–3820 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of themicrosatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8:305–311 Oh HR, An CH, Yoo NJ, Lee SH, 2015, Frameshift mutations of MUC15 gene in gastric and its regional heterogeneity in gastric and colorectal cancers. Pathol Oncol Res 21:713–718 Harris DR, Mims A, Bunz F, 2012, Genetic disruption of USP9X sensitizes colorectal cancer cells to 5-fluorouracil. Cancer Biol Ther 13:1319–1324 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 219 EP 220 DI 10.1007/s12253-016-0140-z PG 2 ER PT J AU Jo, SY Song, YS Kim, SM Yoo, JN Lee, HS AF Jo, Sol Yun Song, Yong Sang Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutations of SMG7 Essential for Nonsense-Mediated mRNA Decay in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Song, Yong Sang] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of PathologySeoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR KaramR,Wengrod J, Gardner LB,WilkinsonMF, 2013, Regulation of nonsense-mediated mRNA decay: implications for physiology and disease. Biochim Biophys Acta 1829:624–633 Perrin-Vidoz L, Sinilnikova OM, Stoppa-Lyonnet D, Lenoir GM, Mazoyer S, 2002, The nonsense-mediated mRNA decay pathway triggers degradation of most BRCA1 mRNAs bearing premature termination codons. Hum Mol Genet 11:2805–2814 Karam R, Carvalho J, Bruno I, Graziadio C, Senz J, Huntsman D, Carneiro F, Seruca R, Wilkinson MF, Oliveira C, 2008, The NMD mRNA surveillance pathway downregulates aberrant E-cadherin transcripts in gastric cancer cells and in CDH1 mutation carriers. Oncogene 27:4255–4260 Snow BE, Erdmann N, Cruickshank J, Goldman H, Gill RM, Robinson MO, Harrington L, 2003, Functional conservation of the telomerase protein Est1p in humans. Curr Biol 13:698–704 Liu C, KaramR, Zhou R, Su F, Ji Y, Li G, XuG, Lu L,Wang C, Song M, Zhu J,Wang Y, Zhao Y, FooWC, Zuo M, Valasek MA, Javle M, Wilkinson MF, Lu Y, 2014, The UPF1 RNA surveillance gene is commonly mutated in pancreatic adenosquamous carcinoma. Nat Med 20:596–598 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Oh HR, An CH, Yoo NJ, Lee SH, 2015, Frameshift mutations of TAF1C gene, a core component for transcription by RNA polymerase I, and its regional heterogeneity in gastric and colorectal cancers. Pathology 47:101–104 Oh HR, An CH, Yoo NJ, Lee SH, 2015, Frameshift mutations of MUC15 gene in gastric and its regional heterogeneity in gastric and colorectal cancers. Pathol Oncol Res 21:713–718 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 221 EP 222 DI 10.1007/s12253-016-0141-y PG 2 ER PT J AU Choi, SK Kim, T Yoo, HK AF Choi, Seung-Kwon Kim, Taegu Yoo, Han Koo TI Spermatocytic Seminoma with Brain Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Choi, Seung-Kwon] Kyung Hee University, School of Medicine, Department of UrologySeoul, South Korea. [Kim, Taegu] Chung Dam Urologic ClinicNamyangju, South Korea. [Yoo, Han Koo] Kyung Hee University, School of Medicine, Department of Urology, #149 Sangil-Dong, Gangdong-Gu, 134-727 Seoul, South Korea. RP Yoo, HK (reprint author), Kyung Hee University, School of Medicine, Department of Urology, 134-727 Seoul, South Korea. EM malta1@naver.com CR Carriere P, Baade P, Fritschi L, 2007, Population based incidence and age distribution of spermatocytic seminoma. J Urol 178(1):125– 128., DOI 10.1016/j.juro.2007.03.024 Chung PW, Bayley AJ, Sweet J, Jewett MA, Tew-George B, Gospodarowicz MK, Warde PR, 2004, Spermatocytic seminoma: a review. Eur Urol 45(4):495–498., DOI 10.1016/j.eururo.2003.11.005 Reuter VE, 2005, Origins and molecular biology of testicular germ cell tumors. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 18 Suppl 2:S51– 60., DOI 10.1038/modpathol.3800309 Matoska J, Ondrus D, Hornak M, 1988, Metastatic spermatocytic seminoma. A case report with light microscopic, ultrastructural, and immunohistochemical findings. Cancer 62(6):1197–1201 Wetherell D, Lawrentschuk N, Gyomber D, 2013, Spermatocytic seminoma with sarcoma: an indication for adjuvant chemotherapy in localized disease. Korean journal of urology 54(12):884–887., DOI 10.4111/kju.2013.54.12.884 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2017 VL 23 IS 1 BP 223 EP 224 DI 10.1007/s12253-016-0142-x PG 2 ER PT J AU Sabour, L Sabour, M Ghorbian, S AF Sabour, Leila Sabour, Maryam Ghorbian, Saeid TI Clinical Applications of Next-Generation Sequencing in Cancer Diagnosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE NGS; Cancer; Next-generation sequencing; Diagnosis; Clinical practice ID NGS; Cancer; Next-generation sequencing; Diagnosis; Clinical practice AB With the advancement and improvement of new sequencing technology, next-generation sequencing (NGS) has been applied increasingly in cancer genomics research fields. More recently, NGS has been adopted in clinical oncology to advance personalized treatment of cancer. NGS is utilized to novel diagnostic and rare cancer mutations, detection of translocations, inversions, insertions and deletions, detection of copy number variants, detect familial cancer mutation carriers, provide the molecular rationale for appropriate targeted, therapeutic and prognostic. NGS holds many advantages, such as the ability to fully sequence all types of mutations for a large number of genes (hundreds to thousands) and the sensitivity, speed in a single test at a relatively low cost compared to be other sequencing modalities. Here we described the technology, methods and applications that can be immediately considered and some of the challenges that lie ahead. C1 [Sabour, Leila] Islamic Azad University, Ahar Branch, Department of Molecular BiologyAhar, Iran. [Sabour, Maryam] Islamic Azad University, Ahar Branch, Department of Molecular BiologyAhar, Iran. [Ghorbian, Saeid] Islamic Azad University, Ahar Branch, Department of Molecular BiologyAhar, Iran. RP Ghorbian, S (reprint author), Islamic Azad University, Ahar Branch, Department of Molecular Biology, Ahar, Iran. EM ghorbian20@yahoo.com CR Jones S, Zhang X, Parsons DW, Lin JC-H, Leary RJ, Angenendt P, Mankoo P, Carter H,KamiyamaH, JimenoA(2008, Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321(5897):1801–1806 Siegel R, Ward E, Brawley O, Jemal A, 2011, Cancer statistics, 2011. CA Cancer J Clin 61(4):212–236 Cahill DP, Kinzler KW, Vogelstein B, Lengauer C, 1999, Genetic instability and darwinian selection in tumours. Trends Genet 15(12):M57–M60 Subramanian J, Simon R, 2010, Gene expression–based prognostic signatures in lung cancer: ready for clinical use? J Natl Cancer Inst 102(7):464–474 Ioannidis JP, Panagiotou OA, 2011, Comparison of effect sizes associated with biomarkers reported in highly cited individual articles and in subsequent meta-analyses. JAMA 305(21):2200–2210 Kallioniemi A, Kallioniemi O-P, Sudar D, Rutovitz D, Gray JW, Waldman F, Pinkel D, 1992, Comparative genomic hybridization for molecular cytogenetic analysis of solid tumors. Science 258(5083):818–821 Armengol G, Capella G, Farre L, Peinado MA, Miro R, CaballinMR, 2001, Genetic evolution in the metastatic progression of human pancreatic cancer studied by CGH. Lab Investig 81(12):1703–1707 Jiang JK, Chen YJ, Lin CH, Yu I, Lin JK, 2005, Genetic changes and clonality relationship between primary colorectal cancers and their pulmonary metastases—an analysis by comparative genomic hybridization. Genes Chromosom Cancer 43(1):25–36 Nishizaki T, DeVries S, Chew K, Goodson WH, Ljung B-M, Thor A, Waldman FM, 1997, Genetic alterations in primary breast cancers and their metastases: direct comparison using modified comparative genomic hybridization. Genes Chromosom Cancer 19(4): 267–272 Petersen S, Aninat-Meyer M, Schluns K, Gellert K, Dietel M, Petersen I, 2000, Chromosomal alterations in the clonal evolution to the metastatic stage ofquamous cell carcinomas of the lung. Br J Cancer 82(1):65 Schmidt-Kittler O, Ragg T, Daskalakis A, Granzow M, Ahr A, Blankenstein TJ, Kaufmann M, Diebold J, Arnholdt H, Muller P, 2003, From latent disseminated cells to overt metastasis: genetic analysis of systemic breast cancer progression. Proc Natl Acad Sci 100(13):7737–7742 Wagle N, Berger MF, Davis MJ, Blumenstiel B, DeFelice M, Pochanard P, Ducar M, Van Hummelen P, MacConaill LE, Hahn WC, 2012, High-throughput detection of actionable genomic alterations in clinical tumor samples by targeted, massively parallel sequencing. Cancer discovery 2(1):82–93 Leary RJ, Kinde I, Diehl F, Schmidt K, Clouser C, Duncan C, Antipova A, Lee C, McKernan K, Francisco M, 2010, Development of personalized tumor biomarkers using massively parallel sequencing. Sci Transl Med 2(20):20ra14–20ra14 McBride DJ, Orpana AK, Sotiriou C, Joensuu H, Stephens PJ, Mudie LJ, Hamalainen E, Stebbings LA, Andersson LC, Flanagan AM, 2010, Use of cancer-specific genomic rearrangements to quantify disease burden in plasma from patients with solid tumors. Genes Chromosom Cancer 49(11):1062–1069 Mardis ER, 2011, A decade/'s perspective on DNA sequencing technology. Nature 470(7333):198–203 Meyerson M, Gabriel S, Getz G, 2010, Advances in understanding cancer genomes through second-generation sequencing. Nat Rev Genet 11(10):685–696 Ku, C.-S., Wu, M., Cooper, D.N., Naidoo, N., Pawitan, Y., Pang, B., Iacopetta, B., Soong, R., 2012. Technological advances in DNA sequence enrichment and sequencing for germline genetic diagnosis. Meldrum C, Doyle MA, Tothill RW, 2011, Next-generation sequencing for cancer diagnostics: a practical perspective. The Clinical Biochemist Reviews 32(4):177 Cronin M, Ross JS, 2011, Comprehensive next-generation cancer genome sequencing in the era of targeted therapy and personalized oncology. Biomark Med 5(3):293–305 Rizzo JM, Buck MJ, 2012, Key principles and clinical applications of "next-generation" DNA sequencing. Cancer Prev Res 5(7):887–900 Desai AN, Jere A, 2012, Next-generation sequencing: ready for the clinics? Clin Genet 81(6):503–510 Ross JS, Cronin M, 2011, Whole cancer genome sequencing by next-generation methods. Am J Clin Pathol 136(4):527–539 Schadt EE, Turner S, Kasarskis A, 2010, A window into thirdgeneration sequencing. Hum Mol Genet 19(R2):R227–R240 Mardis ER, 2010, The $1000 genome, the $100,000 analysis. Genome Med 2(11):84 Sanger F, Nicklen S, Coulson AR, 1977, DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci 74(12):5463–5467 Mardis ER, 2013, Next-generation sequencing platforms. Annu Rev Anal Chem 6:287–303 Ulahannan D, Kovac M, Mulholland P, Cazier J, Tomlinson I, 2013, Technical and implementation issues in using nextgeneration sequencing of cancers in clinical practice. Br J Cancer 109(4):827–835 Campbell PJ, Stephens PJ, Pleasance ED, O’Meara S, Li H, Santarius T, Stebbings LA, Leroy C, Edkins S, Hardy C, 2008, Identification of somatically acquired rearrangements in cancer using genome-wide massively parallel paired-end sequencing. Nat Genet 40(6):722–729 Chiang DY, Getz G, Jaffe DB, O’Kelly MJ, Zhao X, Carter SL, Russ C, Nusbaum C, Meyerson M, Lander ES, 2009, Highresolution mapping of copy-number alterations with massively parallel sequencing. Nat Methods 6(1):99–103 Mortazavi A,Williams BA,McCue K, Schaeffer L,Wold B, 2008, Mapping and quantifying mammalian transcriptomes by RNA-seq. Nat Methods 5(7):621–628 Thomas RK, Nickerson E, Simons JF, Janne PA, Tengs T, Yuza Y, Garraway LA, LaFramboise T, Lee JC, Shah K, 2006, Sensitive mutation detection in heterogeneous cancer specimens by massively parallel picoliter reactor sequencing. Nat Med 12(7):852–855 Shah SP, Morin RD, Khattra J, Prentice L, Pugh T, Burleigh A, Delaney A, Gelmon K, Guliany R, Senz J, 2009, Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature 461(7265):809–813 Campbell PJ, Yachida S, Mudie LJ, Stephens PJ, Pleasance ED, Stebbings LA, Morsberger LA, Latimer C, McLaren S, Lin M-L, 2010, The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467(7319):1109–1113 Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, Kamiyama M, Hruban RH, Eshleman JR, Nowak MA, 2010, Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467(7319):1114–1117 Ding L, Ellis MJ, Li S, Larson DE, Chen K,Wallis JW, Harris CC, McLellan MD, Fulton RS, Fulton LL, 2010, Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature 464(7291):999–1005 Robbins CM, Tembe WA, Baker A, Sinari S, Moses TY, Beckstrom-Sternberg S, Beckstrom-Sternberg J, Barrett M, Long J, Chinnaiyan A, 2011, Copy number and targeted mutational analysis reveals novel somatic events in metastatic prostate tumors. Genome Res 21(1):47–55 Morey M, Fernandez-Marmiesse A, Castineiras D, Fraga JM, Couce ML, Cocho JA, 2013, A glimpse into past, present, and future DNA sequencing. Mol Genet Metab 110(1):3–24 Foquet M, Samiee KT, Kong X, Chauduri BP, Lundquist PM, Turner SW, Freudenthal J, Roitman DB, 2008, Improved fabrication of zero-mode waveguides for single-molecule detection. J Appl Phys 103(3):034301 Eid J, Fehr A, Gray J, Luong K, Lyle J, Otto G, Peluso P, Rank D, Baybayan P, Bettman B, 2009, Real-time DNA sequencing from single polymerase molecules. Science 323(5910):133–138 Berger MF, Lawrence MS, Demichelis F, Drier Y, Cibulskis K, Sivachenko AY, Sboner A, Esgueva R, Pflueger D, Sougnez C, 2011, The genomic complexity of primary human prostate cancer. Nature 470(7333):214–220 Shah SP, Kobel M, Senz J, Morin RD, Clarke BA, Wiegand KC, Leung G, Zayed A, Mehl E, Kalloger SE, 2009, Mutation of FOXL2 in granulosa-cell tumors of the ovary. N Engl J Med 360(26):2719–2729 Jones SJ, Laskin J, Li YY, Griffith OL, An J, BilenkyM, Butterfield YS, Cezard T, Chuah E, Corbett R, 2010, Evolution of an adenocarcinoma in response to selection by targeted kinase inhibitors. Genome Biol 11(8):R82 Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, Senz J, McConechy MK, Anglesio MS, Kalloger SE, 2010, ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med 363(16):1532–1543 Ley TJ, Mardis ER, Ding L, Fulton B, McLellan MD, Chen K, Dooling D, Dunford-Shore BH, McGrath S, Hickenbotham M, 2008, DNA sequencing of a cytogenetically normal acute myeloid leukaemia genome. Nature 456(7218):66–72 Mardis ER, 2010, Cancer genomics identifies determinants of tumor biology. Genome Biol 11(5):211 Mardis ER, Wilson RK, 2009, Cancer genome sequencing: a review. Hum Mol Genet 18(R2):R163–R168 Morin RD, Johnson NA, Severson TM,Mungall AJ, An J, Goya R, Paul JE, Boyle M, Woolcock BW, Kuchenbauer F, 2010, Somatic mutations altering EZH2, Tyr641, in follicular and diffuse large Bcell lymphomas of germinal-center origin. Nat Genet 42(2):181– 185 Hudson TJ, AndersonW, Aretz A, Barker AD, Bell C, Bernabe RR, Bhan M, Calvo F, Eerola I, Gerhard DS, 2010, International network of cancer genome projects. Nature 464(7291):993–998 McLendon R, Friedman A, Bigner D, Van Meir EG, Brat DJ, Mastrogianakis GM, Olson JJ, Mikkelsen T, Lehman N, Aldape K, 2008, Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455(7216):1061– 1068 Shoubridge C, Tarpey PS, Abidi F, Ramsden SL, Rujirabanjerd S, Murphy JA, Boyle J, Shaw M, Gardner A, Proos A, 2010, Mutations in the guanine nucleotide exchange factor gene IQSEC2 cause nonsyndromic intellectual disability. Nat Genet 42(6):486–488 Bonnefond, A., Durand, E., Sand, O., De Graeve, F., Gallina, S., Busiah, K., Lobbens, S., Simon, A., Bellanne-Chantelot, C., Letourneau, L., 2010. Molecular diagnosis of neonatal diabetes mellitus using next-generation sequencing of the whole exome. Bolze A, Byun M, McDonald D, Morgan NV, Abhyankar A, Premkumar L, Puel A, Bacon CM, Rieux-Laucat F, Pang K, 2010, Whole-exome-sequencing-based discovery of human FADD deficiency. Am J Hum Genet 87(6):873–881 Esteller M, 2008, Epigenetics in cancer. N Engl J Med 358(11): 1148–1159 Garber JE, Offit K, 2005, Hereditary cancer predisposition syndromes. J Clin Oncol 23(2):276–292 Walsh T, Lee MK, Casadei S, Thornton AM, Stray SM, Pennil C, Nord AS, Mandell JB, Swisher EM, King M-C, 2010, Detection of inherited mutations for breast and ovarian cancer using genomic capture and massively parallel sequencing. Proc Natl Acad Sci 107(28):12629–12633 De Lellis L, Mammarella S, Curia MC, Veschi S, Mokini Z, Bassi C, Sala P, Battista P, Mariani-Costantini R, Radice P, 2011, Analysis of gene copy number variations using a method based on lab-on-a-chip technology. Tumori 98(1):126–136 Walsh T, Casadei S, Coats KH, Swisher E, Stray SM, Higgins J, Roach KC, Mandell J, Lee MK, Ciernikova S, 2006, Spectrum of mutations in BRCA1, BRCA2, CHEK2, and TP53 in families at high risk of breast cancer. JAMA 295(12):1379–1388 Ozcelik H, Shi X, ChangMC, Tram E, Vlasschaert M, Di Nicola N, Kiselova A, Yee D, Goldman A, DowarM(2012, Long-range PCR and next-generation sequencing of BRCA1 and BRCA2 in breast cancer. The Journal of Molecular Diagnostics 14(5):467–475 Hernan I, Borras E, de Sousa Dias M, Gamundi MJ, Mane B, Llort G, Agundez JA, Blanca M, Carballo M, 2012, Detection of genomic variations in BRCA1 and BRCA2 genes by long-range PCR and next-generation sequencing. The Journal of Molecular Diagnostics 14(3):286–293 De Leeneer K, Hellemans J, De Schrijver J, Baetens M, Poppe B, Van Criekinge W, De Paepe A, Coucke P, Claes K, 2011, Massive parallel amplicon sequencing of the breast cancer genes BRCA1 and BRCA2: opportunities, challenges, and limitations. HumMutat 32(3):335–344 Lee W, Jiang Z, Liu J, Haverty PM, Guan Y, Stinson J, Yue P, Zhang Y, Pant KP, Bhatt D, 2010, The mutation spectrum revealed by paired genome sequences from a lung cancer patient. Nature 465(7297):473–477 Pleasance ED, Stephens PJ, O’Meara S, McBride DJ, Meynert A, Jones D, Lin M-L, Beare D, Lau KW, Greenman C, 2010, A smallcell lung cancer genome with complex signatures of tobacco exposure. Nature 463(7278):184–190 Pleasance ED, Cheetham RK, Stephens PJ, McBride DJ, Humphray SJ, Greenman CD, Varela I, Lin M-L, Ordonez GR, Bignell GR, 2010, A comprehensive catalogue of somatic mutations from a human cancer genome. Nature 463(7278):191–196 Quail MA, Kozarewa I, Smith F, Scally A, Stephens PJ, Durbin R, Swerdlow H, Turner DJ, 2008, A large genome center’s improvements to the Illumina sequencing system. NatMethods 5(12):1005– 1010 Branton D, Deamer DW, Marziali A, Bayley H, Benner SA, Butler T, Di Ventra M, Garaj S, Hibbs A, Huang X, 2008, The potential and challenges of nanopore sequencing. Nat Biotechnol 26(10): 1146–1153 Iafrate AJ, Feuk L, Rivera MN, Listewnik ML, Donahoe PK, Qi Y, Scherer SW, Lee C, 2004, Detection of large-scale variation in the human genome. Nat Genet 36(9):949–951 Sebat J, Lakshmi B, Troge J, Alexander J, Young J, Lundin P, Maner S, Massa H, Walker M, Chi M, Navin N, Lucito R, Healy J, Hicks J, Ye K, Reiner A, Gilliam TC, Trask B, Patterson N, Zetterberg A,Wigler M, 2004, Large-scale copy number polymorphism in the human genome. Science 305(5683):525–528 Stankiewicz P, Lupski JR, 2010, Structural variation in the human genome and its role in disease. Annu Rev Med 61:437–455 Zhang F, Gu W, Hurles ME, Lupski JR, 2009, Copy number variation in human health, disease, and evolution. Annu RevGenomics Hum Genet 10:451–481 Korbel JO, Urban AE, Affourtit JP, Godwin B, Grubert F, Simons JF, Kim PM, Palejev D, Carriero NJ, Du L, Taillon BE, Chen Z, Tanzer A, Saunders AC, Chi J, Yang F, Carter NP, Hurles ME, Weissman SM, Harkins TT, Gerstein MB, Egholm M, Snyder M, 2007, Paired-end mapping reveals extensive structural variation in the human genome. Science 318(5849):420–426 Stephens PJ, McBride DJ, Lin ML, Varela I, Pleasance ED, Simpson JT, Stebbings LA, Leroy C, Edkins S, Mudie LJ, Greenman CD, Jia M, Latimer C, Teague JW, Lau KW, Burton J, Quail MA, Swerdlow H, Churcher C, Natrajan R, Sieuwerts AM, Martens JW, Silver DP, Langerod A, Russnes HE, Foekens JA, Reis-Filho JS, van ‘t Veer L, Richardson AL, Borresen-Dale AL, Campbell PJ, Futreal PA, Stratton MR, 2009, Complex landscapes of somatic rearrangement in human breast cancer genomes. Nature 462(7276):1005–1010 Abyzov A, Urban AE, Snyder M, GersteinM(2011, CNVnator: an approach to discover, genotype, and characterize typical and atypical CNVs from family and population genome sequencing. Genome Res 21(6):974–984 Ivakhno S, Royce T, Cox AJ, Evers DJ, Cheetham RK, Tavare S, 2010, CNAseg–a novel framework for identification of copy number changes in cancer from second-generation sequencing data. Bioinformatics 26(24):3051–3058 Kim TM, Luquette LJ, Xi R, Park PJ, 2010, rSW-seq: algorithm for detection of copy number alterations in deep sequencing data.BMC Bioinformatics 11:432 Klambauer G, Schwarzbauer K, Mayr A, Clevert DA, Mitterecker A, Bodenhofer U, Hochreiter S, 2012, cn.MOPS: mixture of Poissons for discovering copy number variations in nextgeneration sequencing data with a low false discovery rate. Nucleic Acids Res 40(9):e69 Magi A, Benelli M, Yoon S, Roviello F, Torricelli F, 2011, Detecting common copy number variants in high-throughput sequencing data by using joint SLM algorithm. Nucleic Acids Res 39(10):e65 Medvedev P, Stanciu M, BrudnoM(2009, Computationalmethods for discovering structural variation with next-generation sequencing. Nat Methods 6(11 Suppl):S13–S20 Miller CA, Hampton O, Coarfa C, Milosavljevic A, 2011, ReadDepth: a parallel R package for detecting copy number alterations from short sequencing reads. PLoS One 6(1):e16327 Waszak SM, Hasin Y, Zichner T, Olender T, Keydar I, Khen M, Stutz AM, Schlattl A, Lancet D, Korbel JO, 2010, Systematic inference of copy-number genotypes from personal genome sequencing data reveals extensive olfactory receptor gene content diversity. PLoS Comput Biol 6(11):e1000988 Xie C, Tammi MT, 2009, CNV-seq, a new method to detect copy number variation using high-throughput sequencing. BMC Bioinformatics 10:80 Yoon S, Xuan Z, Makarov V, Ye K, Sebat J, 2009, Sensitive and accurate detection of copy number variants using read depth of coverage. Genome Res 19(9):1586–1592 Holt C, Losic B, Pai D, Zhao Z, Trinh Q, Syam S, Arshadi N, Jang GH, Ali J, Beck T, McPherson J, Muthuswamy LB, 2014, Wave CNV: allele-specific copy number alterations in primary tumors and xenograft models from next-generation sequencing. Bioinformatics 30(6):768–774 de Sanjose S, Leone M, Berez V, Izquierdo A, Font R, Brunet JM, Louat T, Vilardell L, Borras J, Viladiu P, Bosch FX, Lenoir GM, Sinilnikova OM, 2003, Prevalence of BRCA1 and BRCA2 germline mutations in young breast cancer patients: a populationbased study. Int J Cancer 106(4):588–593 Moller P, Hagen AI, Apold J, Maehle L, Clark N, Fiane B, Lovslett K, Hovig E, Vabo A, 2007, Genetic epidemiology of BRCA mutations–family history detects less than 50 % of the mutation carriers. Eur J Cancer 43(11):1713–1717 Risch HA, McLaughlin JR, Cole DE, Rosen B, Bradley L, Fan I, Tang J, Li S, Zhang S, Shaw PA, Narod SA, 2006, Population BRCA1 and BRCA2 mutation frequencies and cancer penetrances: a kin-cohort study in Ontario, Canada. J Natl Cancer Inst 98(23): 1694–1706 Farmer H,McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, Santarosa M, Dillon KJ, Hickson I, Knights C, Martin NM, Jackson SP, Smith GC, Ashworth A, 2005, Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434(7035):917–921 Morgan JE, Carr IM, Sheridan E, Chu CE, Hayward B, Camm N, Lindsay HA, Mattocks CJ, Markham AF, Bonthron DT, Taylor GR, 2010, Genetic diagnosis of familial breast cancer using clonal sequencing. Hum Mutat 31(4):484–491 Schroeder C, Stutzmann F, Weber BH, Riess O, Bonin M, 2010, High-throughput resequencing in the diagnosis of BRCA1/2 mutations using oligonucleotide resequencing microarrays. Breast Cancer Res Treat 122(1):287–297 Summerer D, Wu H, Haase B, Cheng Y, Schracke N, Stahler CF, CheeMS, Stahler PF, BeierM(2009, Microarray-basedmulticycleenrichment of genomic subsets for targeted next-generation sequencing. Genome Res 19(9):1616–1621 Walsh T, Lee MK, Casadei S, Thornton AM, Stray SM, Pennil C, Nord AS, Mandell JB, Swisher EM, King MC, 2010, Detection of inherited mutations for breast and ovarian cancer using genomic capture and massively parallel sequencing. Proc Natl Acad Sci U S A 107(28):12629–12633 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 225 EP 234 DI 10.1007/s12253-016-0124-z PG 10 ER PT J AU Nobre, CGC de Araujo, MGJ de Medeiros Fernandes, AATh Cobucci, NOR Lanza, CFD Andrade, SV Fernandes, VJ AF Nobre, Cristina Guimaraes Camila de Araujo, Maria Galvao Joselio de Medeiros Fernandes, Allyrio Araujo Thales Cobucci, Ney Oliveira Ricardo Lanza, Carlos Ferreira Daniel Andrade, Sousa Vania Fernandes, Verissimo Jose TI Macrophage Migration Inhibitory Factor (MIF): Biological Activities and Relation with Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Macrophage migration inhibitory factor; Neoplasms; Tumorigenesis; Inflammation ID Macrophage migration inhibitory factor; Neoplasms; Tumorigenesis; Inflammation AB Macrophage migration inhibitory factor (MIF) emerged in recent years as an important inflammation mediator, playing a prominent role in the pathogenesis of various types of malignant neoplasm. MIF is a glycoprotein that presents a wide spectrum of biological activities and exerts a complex interaction with various cellular signaling pathways, causing imbalance of homeostasis. Experimental and clinical studies show that high levels of MIF are found in almost all types of human cancers and are implicated in seemingly all stages of development of the tumors. The production of MIF is triggered through an autocrine signal emitted by tumor cells, and stimulates the production of cytokines, chemokines, and growth as well as angiogenic factors that lead to growth of the tumor, increasing its aggressiveness and metastatic potential. MIF is produced by virtually all types of human body cells, in response to stress caused by different factors, leading to pathological conditions such as chronic inflammation and immunomodulation with suppression of immune surveillance and of immune response against tumors, angiogenesis, and carcinogenesis. In this review, we present recent advances on the biological activity of MIF, the signaling pathways with which it is involved and their role in tumorigenesis. C1 [Nobre, Cristina Guimaraes Camila] Federal University of Rio Grande do Norte, Department of Microbiology and Parasitology, 59072-970 Natal, RN, Brazil. [de Araujo, Maria Galvao Joselio] Federal University of Rio Grande do Norte, Department of Microbiology and Parasitology, 59072-970 Natal, RN, Brazil. [de Medeiros Fernandes, Allyrio Araujo Thales] University of Rio Grande do Norte State, Department of Biomedical Sciences, 59607-360 Mossoro, RN, Brazil. [Cobucci, Ney Oliveira Ricardo] Federal University of Rio Grande do Norte, Department of Microbiology and Parasitology, 59072-970 Natal, RN, Brazil. [Lanza, Carlos Ferreira Daniel] Federal University of Rio Grande do Norte, CEP, Department of Biochemisty, 59072-970 Natal, RN, Brazil. [Andrade, Sousa Vania] Federal University of Rio Grande do Norte, Department of Microbiology and Parasitology, 59072-970 Natal, RN, Brazil. [Fernandes, Verissimo Jose] Federal University of Rio Grande do Norte, Department of Microbiology and Parasitology, 59072-970 Natal, RN, Brazil. RP de Medeiros Fernandes, AATh (reprint author), University of Rio Grande do Norte State, Department of Biomedical Sciences, 59607-360 Mossoro, Brazil. EM thalesallyrio@yahoo.com.br CR Bloom BR, Bennett B, 1966, Mechanism of a reaction in vitro associated with delayed-type hypersensitivity. Science 153(731): 80–82 Takahashi N, Nishihira J, Sato Yet al, 1998, Involvement of macrophage migration inhibitory factor, MIF, in the mechanism of tumor cell growth. Mol Med 4(11):707–714 Onodera S, Nishihira J, Koyama Yet al, 2004, Macrophage migration inhibitory factor up-regulates the expression of interleukin-8 messenger RNA in synovial fibroblasts of rheumatoid arthritis patients: common transcriptional regulatory mechanism between interleukin-8 and interleukin-1beta. Arthritis Rheum 50(5):1437– 1447 Lerch JK, Puga DA, Bloom O, Popovich PG, 2014, Glucocorticoids and macrophage migration inhibitory factor, MIF, are neuroendocrine modulators of inflammation and neuropathic pain after spinal cord injury. Semin Immunol 26(5):409–414 Bernhagen J, Calandra T, Bucala R, 1998, Regulation of the immune response by macrophage migration inhibitory factor: biological and structural features. J Mol Med 76(3–4):151–161 Benigni F, Atsumi T, Calandra T et al, 2000, The proinflammatory mediator macrophage migration inhibitory factor induces glucose catabolism in muscle. J Clin Invest 106(10):1291–1300 Lolis E, Bucala R, 2003, Macrophage migration inhibitory factor. Expert Opin Ther Targets 7:153–164 Shi X, Leng L, Wang T et al, 2006, CD44 is the signaling component of the macrophage migration inhibitory factor-CD74 receptor complex. Immunity 25(4):595–606 Bifulco C, McDaniel K, Leng L, Bucala R, 2008, Tumor growthpromoting properties of macrophage migration inhibitory factor. Curr Pharm Des 14(36):3790–3801 Tillmann S, Bernhagen J, Noels H, 2013, Arrest functions of the MIF ligand/ receptor axes in atherogenesis. Front Immunol 4:115., DOI 10.3389/fimmu.2013.00115 Lue H, Thiele M, Franz J et al, 2007, Macrophage migration inhibitory factor, MIF, promotes cell survival by activation of the Akt pathway and role for CSN5/JAB1 in the control of autocrine MIF activity. Oncogene 26(35):5046–5059 Binsky I, Haran M, Starlets D et al, 2007, IL-8 secreted in a macrophage migration-inhibitory factor- and CD74-dependent manner regulates B cell chronic lymphocytic leukemia survival. Proc Natl Acad Sci U S A 104(33):13408–13413 Hoi AY, Iskander MN, Morand EF, 2007, Macrophage migration inhibitory factor: a therapeutic target across inflammatory diseases. Inflamm Allergy Drug Targets 6(3):183–190 Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA, 2013, Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer 13(11):759–771 Bernhagen J, Calandra T, Mitchell RA et al, 1993, MIF is a pituitary-derived cytokine that potentiates lethal endotoxaemia. Nature 365(6448):756–759 Hagemann T, Robinson SC, Thompson RG, Charles K, Kulbe H, Balkwill FR, 2007, Ovarian cancer cell-derived migration inhibitory factor enhances tumor growth, progression, and angiogenesis. Mol Cancer Ther 6(7):1993–2002 Wu S-P, Leng L, Feng Z et al, 2006, Macrophage migration inhibitory factor promoter polymorphisms and the clinical expression of scleroderma. Arthritis Rheum 54(11):3661–3669 Dambacher J, Staudinger T, Seiderer J et al, 2007, Macrophage migration inhibitory factor, MIF, −173G/C promoter polymorphism influences upper gastrointestinal tract involvement and disease activity in patients with Crohn’s disease. Inflamm Bowel Dis 13(1):71–82 Tong X, Zheng B, Tong Q et al, 2015, The MIF -173G/C gene polymorphism increase gastrointestinal cancer and hematological malignancy risk : evidence from a meta-analysis and FPRP test. Int J Clin Exp Med 8(9):15949–15957 Meyer-Siegler KL, Vera PL et al, 2007, Macrophage migration inhibitory factor, MIF, gene polymorphisms are associated with increased prostate cancer incidence. Genes Immun 8(8):646–652 Arisawa T, Tahara T, Shibata T et al, 2008, Functional promoter polymorphisms of the macrophage migration inhibitory factor gene in gastric carcinogenesis. Oncol Rep 19(1):223–228 Grieb G, Merk M, Berhagen J, Bucala R, 2010, Macrophage migration inhibitory factor, MIF): a promising biomarker. Drug News Perspect 23(4):257–264 Babu SN, Chetal G, Kumar S, 2012, Macrophage migration inhibitory factor: a potential marker for cancer diagnosis and therapy. Asian Pac J Cancer Prev 13(5):1737–1744 Krockenberger M, Engel JB, Kolb J et al, 2010, Macrophage migration inhibitory factor expression in cervical cancer. J Cancer Res Clin Oncol 136(5):651–657 Richard V, Kindt N, Saussez S, 2015, Macrophage migration inhibitory factor involvement in breast cancer, review). Int J Oncol 47(5):1627–1633 Fingerle-Rowson GR, Bucala R, 2001, Neuroendocrine properties of macrophage migration inhibitory factor, MIF ). Immunol Cell Biol 79(4):368–375 Conroy H, Mawhinney L, Donnelly SC, 2010, Inflammation and cancer:macrophage migration inhibitory factor, MIF)–the potential missing link. QJM 103(11):831–836 Rendon BE, Willer SS, Zundel W, Mitchell RA, 2009, Mechanisms of macrophage migration inhibitory factor, MIF)-dependent tumor microenvironmental adaptation. Exp Mol Pathol 86(3):180–185 Rosado JDD, Rodriguez-Sosa M, 2011, Macrophage migration inhibitory factor, MIF): a key player in protozoan infections. Int J Biol Sci 7(9):1239–1256 Lehmann LE, Weber SU, Fuchs D et al, 2008, Oxidoreductase macrophagemigration inhibitory factor is simultaneously increased in leukocyte subsets of patients with severe sepsis. Biofactors 33(4): 281–291 Simpson KD, Templeton DJ, Cross JV, 2012, Macrophage migration inhibitory factor promotes tumor growth and metastasis by inducing myeloid-derived suppressor cells in the tumor microenvironment. J Immunol 189(12):5533–5540 Yaddanapudi K, Rendon BE, Lamont G et al, 2016, MIF is necessary for late-stage melanoma patient MDSC immune suppression and differentiation. Cancer Immunol Res 4(2):101–112 Bucala R, 1996, MIF rediscovered : cytokine, pituitary hormone, and glucocorticoid-induced regulator of the immune response. FASEB J 10(14):1607–1613 Fingerle-Rowson G, Koch P, Bikoff R et al, 2003, Regulation of macrophage migration inhibitory factor expression by glucocorticoids in vivo. Am J Pathol 162(1):47–56 Mawhinney L, Armstrong ME, O′ Reilly C, et al, 2015, Macrophage migration inhibitory factor, MIF, enzymatic activity and lung cancer. Mol Med 20:729–735. Gregory JL, Leech MT, David JR, Yang YH, Dacumos A, Hickey MJ, 2004, Reduced leukocyte-endothelial cell interactions in the inflamed microcirculation of macrophage migration inhibitory factor-deficient mice. Arthritis Rheum 50(9):3023–3034 Bernhagen J, Krohn R, Lue H et al, 2007, MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med 13(5):587–596 Nishino T, Bernhagen J, Shiiki H, Calandra T, Dohi K, Bucala R, 1995, Localization of macrophage migration inhibitory factor, MIF, to secretory granules within the corticotrophic and thyrotrophic cells of the pituitary gland. Mol Med 1(7):781–788 Bando H,Matsumoto G, Bando M et al, 2002, Expression of macrophage migration inhibitory factor in human breast cancer: association with nodal spread. Jpn J Cancer Res 93(4):389–396 Lue H, Kleemann R, Calandra Tet al, 2002)Macrophagemigration inhibitory factor, MIF): mechanisms of action and role in disease. Microbes Infect 4(4):449–460 Suzuki M, Murata K, Tanaka E, Nishihira J, Sakai M, 1994, Crystallization and a preliminar X-ray diffraction study of macrophage migration inhibitory factor from human lymphocytes. J Mol Biol 235(3):1141–1143 Rosengren E, Bucala R, Aman P et al, 1996, The immunoregulatory mediator macrophage migration inhibitory factor, MIF, catalyzes a tautomerization reaction. Mol Med 2(1):143–149 Rosengren E, Aman P, Thelin S et al, 1997, The macrophage migration inhibitory factor MIF is a phenylpyruvate tautomerase. FEBS Lett 417(1):85–88 Kleemann R, Kapurniotu A, Frank RWet al, 1998, Disulfide analysis reveals a role for macrophage migration inhibitory factor, MIF, as thiol-protein oxidoreductase. J Mol Biol 280(1):85–102 Matsunaga J, Sinha D, Solano F, Santis C, Wistow G, Hearing V et al, 1999, Macrophage migration inhibitory factor, MIF)–its role in catecholamine metabolism. Cell Mol Biol, Noisy-le-grand, 45(7):1035–1040 El-turk F, Cascella M, Ouertatani-sakouhi H et al, 2008, The conformational flexibility of the carboxy terminal residues 105 − 114 is a keymodulator of the catalytic activity and stability of macrophage migration inhibitory factor. Biochemistry 47(40):10740–10756 Cooke G, Armstrong ME, Donnelly SC, 2009)Macrophage migration inhibitory factor, MIF), enzymatic activity and the inflammatory response. Biofactors 35(2):165–168 Vousden KH, Lane DP, 2007, P53 in health and disease. Nat Rev Mol Cell Biol 8(4):275–283 Jung H, Seong HA, Ha H, 2008, Critical role of cysteine residue 81 of macrophage migration inhibitory factor, MIF, in MIF-induced inhibition of p53 activity. J Biol Chem 283(29):20383–20396 HanelW,MollUM(2012, Links between mutant p53 and genomic instability. J Cell Biochem 113(2):433–439 Krockenberger M, Dombrowski Y, Weidler C et al, 2008, Macrophage migration inhibitory factor contributes to the immune escape of ovarian cancer by down-regulating NKG2D. J Immunol 180(11):7338–7348 Nakaya K, Nabata Y, Ichiyanagi T, An WW, 2012, Stimulation of dendritic cell maturation and induction of apoptosis in leukemia cells by a heat-stable extract from azuki bean, Vigna angularis), a promising immunopotentiating food and dietary supplement for cancer prevention. Asian Pac J Cancer Prev 13(2):607–611 Youn JI, Nagaraj S, Collazo M, Gabrilovich DI, 2008, Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol 181(8):5791–5802 Yaddanapudi K, Putty K, Rendon BE, et al, 2013, Control of tumor-associated macrophage alternative activation by macrophage migration inhibitory factor. J Immunol190(6):2984–2993. Guo C, Buranych A, Sarkar D, Fisher PB, Wang XY, 2013, The role of tumor-associated macrophages in tumor vascularization. Vasc Cell 5:20., DOI 10.1186/2045-824X-5-20 Shen YC, Thompson DL, Kuah MK et al, 2012, The cytokine macrophage migration inhibitory factor, MIF, acts as a neurotrophin in the developing inner ear of the zebrafish, Danio rerio. Dev Biol 363(1):84–94 Zhang X, Chen L, Wang Y et al, 2013, Macrophage migration inhibitory factor promotes proliferation and neuronal differentiation of neural stem/precursor cells through Wnt/β-catenin signal pathway. Int J Biol Sci 9(10):1108–1120 Massirer KB, Carromeu C, Griesi-Oliveira K, Muotri AR, 2011, Maintenance and differentiation of neural stem cells. Wiley Interdiscip Rev Syst Biol Med 3(1):107–114 Fernandes JV, Fernandes TAAM, Azevedo JCV et al, 2015, Link between chronic inflammation and human papillomavirus-induced carcinogenesis, review). Oncol Lett 9(3):1015–1026 Landskron G, De La Fuente M, Thuwajit P et al, 2014, Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res 2014:149185., DOI 10.1155/2014/149185 Fernandes JV, Cobucci RNO, Jatoba CAN, Fernandes TA, Azevedo JW, Araujo JM, 2015, The role of the mediators of inflammation in cancer development. Pathol Oncol Res 21(3):527– 534 Santos LL, Morand EF, 2006, The role of macrophage migration inhibitory factor in the inflammatory immune response and rheumatoid arthritis. Wien Med Wochenschr 156(1–2):11–18 Verjans E, Noetzel E, Bektas N et al, 2009, Dual role of macrophage migration inhibitory factor, MIF, in human breast cancer. BMC Cancer 9:230., DOI 10.1186/1471-2407-9-230 Gordon-Weeks AN, Lim SY, Yuzhalin AE, Jones K, Muschel R, 2015, Macrophage migration inhibitory factor : a key cytokine and therapeutic target in colon cancer. Cytokine Growth Factor Rev 26(4):451–461 Ren Y, Law S, Huang X, Lee PY, Bacher M, Srivastava G,Wong J, 2005, Macrophage migration inhibitory factor stimulates angiogenic factor expression and correlates with differentiation and lymph node status in patients with esophageal squamous cell carcinoma. Ann Surg 242(1):55–63 Cheng RJ, Deng WG, Niu CB, Li YY, Fu Y, 2011, Expression of macrophage migration inhibitory factor and CD74 in cervical squamous cell carcinoma. Int J Gynecol Cancer 21(6):1004–1012 TangW, Gou X, Liu Q, 2011, Expression ofmacrophagemigration inhibition factor, MIF, in serumof patients with prostate cancer and its clinical significance. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 27(1):97–98 Ren Y, Tsui HT, Poon RT et al, 2003, Macrophage migration inhibitory fator – roles in regulating tumor cell migration and expression. Of angiogenic factors in hepatocellular carcinoma. Int J Cancer 107(1):22–29 White ES, Flaherty KR, Carskadon S et al, 2003, Macrophage migration inhibitory factor and CXC chemokine expression in non-small cell lung cancer: role in angiogenesis and prognosis. Clin Cancer Res 9(2):853–860 Ren Y, Chan HM, Fan J et al, 2006, Inhibition of tumor growth and metastasis in vitro and in vivo by targeting macrophage migration inhibitory factor in human neuroblastoma. Oncogene 25(25):3501– 3508 Guo YS, Dai YP, Li Wet al, 2011, Expression and significance of macrophage migration inhibitory factor in bladder urothelial cell carcinoma. Zhonghua Zhong Liu Za Zhi 33(1):28–31 Bondza PK, Metz CN, Akoum A, 2008, Macrophage migration inhibitory factor up-regulates alpha(v)beta(3, integrin and vascular endothelial growth factor expression in endometrial adenocarcinoma cell line Ishikawa. J Reprod Immunol 77(2):142–151 Gabitass RF, Annels NE, Stocken DD, Pandha HA,MiddletonGW, 2011, Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunol Immunother 60(10):1419–1430 Zea AH, Rodriguez PC, Atkins MB et al, 2005, Arginase producing suppressor myeloid cells in renal cell carcinoma patients: a mechanism of tumor evasion. Cancer Res 65(8):3044–3048 Cohen NA, Stewart ML, Gavathiotis E et al, 2012, A competitive stapled peptide screen identifies a selective small molecule that overcomes MCL-1-dependent leukemia cell survival. Chem Biol 19(9):1175–1186 Gore Y, Starlets D, Maharshak N et al, 2008, Macrophage migration inhibitory factor induces B cell survival by activation of a CD74-CD44 receptor complex. J Biol Chem 283(5):2784–2792 Mitchell RA, 2004, Mechanisms and effectors of MIF-dependent promotion of tumourigenesis. Cell Signal 16(1):13–19 Tarnowski M, Grymula K, Liu R et al, 2011, Macrophage migration inhibitory factor is secreted by rhabdomyosarcoma cells, modulates tumor metastasis by binding to CXCR4 and CXCR7 receptors and inhibits recruitment of cancer-associated fibroblasts.Mol. Cancer Res 8(10):1328–1343 Oda S, Oda T, Nishi K et al, 2008, Macrophage migration inhibitory factor activates hypoxia-inducible factor in a p53-dependent manner. PLoS One 3(5):e2215., DOI 10.1371/journal.pone.0002215 Ichihara E, Kiura K, Tanimoto M, 2011, Targeting angiogenesis in cancer therapy. Acta Med Okayama 65(6):353–362 Veillat V, Carli C, Metz CN, Al-Abed Y, Naccache PH, Akoum A, 2010, Macrophage migration inhibitory factor elicits an angiogenic phenotype in human ectopic endometrial cells and triggers the production of major angiogenic factors via CD44, CD74, and MAPK signaling pathways. J Clin Endocrinol Metab 95(12): e403–e412., DOI 10.1210/jc.2010-0417 Funamizu N, Hu C, Lacy C et al, 2013, Macrophage migration inhibitory factor induces epithelial to mesenchymal transition, enhances tumor aggressiveness and predicts clinical outcome in resected pancreatic ductal adenocarcinoma. Int J Cancer 132(4): 785–794 Meyer-Siegler KL, Iczkowski KA, Vera PL, 2005, Further evidence for increased macrophage migration inhibitory factor expression in prostate cancer.BMC Cancer 5:73., DOI 10.1186/1471-2407- 5-73 McClelland M, Zhao L, Carskadon S, Arenberg D, 2009, Expression of CD74, the receptor for macrophage migration inhibitory factor, in non-small cell lung cancer. Am J Pathol 174(2):638– 646 Richard V, Kindt N, Decaestecker C et al, 2014, Involvement of macrophage migration inhibitory factor and its receptor, CD74, in human breast cancer. Oncol Rep 32(2):523–529 Dietlin TA, Hofman FM, Lund BT, Gilmore W, Stohlman SA, van der Veen RC, 2007, Mycobacteria-induced Gr-1+ subsets from distinct myeloid lineages have opposite effects on T cell expansion. J Leukoc Biol 81(5):1205–1212 Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S, 2007, Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res 67(9):4507–4513 Movahedi K, Guilliams M, Van Den Bossche J et al, 2008, Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell suppressive activity. Blood 111(8):4233–4244 Gasser S, Orsulic S, Brown EJ, Raulet DH, 2005, The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436(7054):1186–1190 Mittelbronn M, Platten M, Zeiner P et al, 2011, Macrophage migration inhibitory factor, MIF, expression in human malignant gliomas contributes to immune escape and tumour progression. Acta Neuropathol 122(3):353–365 Pende D, Rivera P, Marcenaro S et al, 2002, Major histocompatibility complex class I-related chain a and UL16-binding protein expression on tumor cell lines of different histotypes: analysis of tumor susceptibility to NKG2D-dependent natural killer cell cytotoxicity. Cancer Res 62(21):6178–6186 Tanese K,Hashimoto Y, Berkova Z et al, 2015, Cell surface CD74– MIF interactions drivemelanoma survival in response to interferon- γ. J Invest Dermatol 135(11):2901., DOI 10.1038/jid.2015.259 Wu S, Lian J, Tao H, Shang H, Zhang L, 2011, Correlation of macrophage migration inhibitory factor gene polymorphism with the risk of early-stage cervical cancer and lymphatic metastasis. Oncol Lett 2(6):1261–1267 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 235 EP 244 DI 10.1007/s12253-016-0138-6 PG 10 ER PT J AU Virag, J Haberler, Ch Baksa, G Piurko, V Hegedus, Z Reiniger, L Balint, K Chocholous, M Kiss, A Lotz, G Glasz, T Schaff, Zs Garami, M Hegedus, B AF Virag, Jozsef Haberler, Christine Baksa, Gabor Piurko, Violetta Hegedus, Zita Reiniger, Lilla Balint, Katalin Chocholous, Monika Kiss, Andras Lotz, Gabor Glasz, Tibor Schaff, Zsuzsa Garami, Miklos Hegedus, Balazs TI Region Specific Differences of Claudin-5 Expression in Pediatric Intracranial Ependymomas: Potential Prognostic Role in Supratentorial Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ependymoma; Claudin-5; Tight junction; Supratentorial; Choroid plexus epithelium; Prognosis ID Ependymoma; Claudin-5; Tight junction; Supratentorial; Choroid plexus epithelium; Prognosis AB Ependymomas are common pediatric brain tumors that originate from the ependyma and characterized by poor prognosis due to frequent recurrence. However, the current WHO grading system fails to accurately predict outcome. In a retrospective study, we analyzed 54 intracranial pediatric ependymomas and found a significantly higher overall survival in supratentorial cases when compared to infratentorial tumors. Next we performed region-specific immunohistochemical analysis of the ependyma in neonatal and adult ependyma from the central canal of spinal cord to the choroid plexus of lateral ventricles for components of cell-cell junctions including cadherins, claudins and occludin. We found robust claudin-5 expression in the choroid plexus epithelia but not in other compartments of the ependyma. Ultrastructural studies demonstrated distinct regional differences in cell-cell junction organization. Surprisingly, we found that 9 out of 20 supratentorial but not infratentorial ependymomas expressed high levels of the brain endothelial tight junction component claudin-5 in tumor cells. Importantly, we observed an increased overall survival in claudin-5 expressing supratentorial ependymoma. Our data indicates that claudin-5 expressing ependymomas may follow a distinct course of disease. The assessment of claudin-5 expression in ependymoma has the potential to become a useful prognostic marker in this pediatric malignancy. C1 [Virag, Jozsef] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Haberler, Christine] Medical University of Vienna, Department of NeuropathologyVienna, Austria. [Baksa, Gabor] Semmelweis University, 1st Department of AnatomyBudapest, Hungary. [Piurko, Violetta] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Hegedus, Zita] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Reiniger, Lilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Balint, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Chocholous, Monika] Medical University of Vienna, Department of PediatricsVienna, Austria. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Glasz, Tibor] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Garami, Miklos] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Hegedus, Balazs] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. RP Hegedus, B (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM balazs.hegedus@meduniwien.ac.at CR Bergeron C, Philip T, 2004, Childhood cancer. Epidemiologic, diagnostic, and therapeutic aspects. La Revue du praticien 54(13): 1489–1495 Maksoud YA, Hahn YS, Engelhard HH, 2002, Intracranial ependymoma. Neurosurg Focus 13(3):e4 Gelabert Gonzalez M, Garcia Allut A, Fernandez Villa JM, Gonzalez Garcia J, Martinez Rumbo R, 2001, Intracranial ependymomas. Rev Neurol 33(10):980–986 Yang CP, Hung IJ, Jaing TH, Shih LY, ChangWH, 2005, Cancer in infants: a reviewof 82 cases. Pediatr HematolOncol 22(6):463–481 Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P, 2007, The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114(2):97–109., DOI 10.1007/s00401-007-0243-4 Kilday JP, Rahman R, Dyer S, Ridley L, Lowe J, Coyle B, Grundy R, 2009, Pediatric ependymoma: biological perspectives. Mol Cancer Res 7(6):765–786., DOI 10.1158/1541-7786.MCR-08-0584 Godfraind C, 2009, Classification and controversies in pathology of ependymomas. Childs Nerv Syst 25(10):1185–1193., DOI 10.1007/s00381-008-0804-4 Preusser M, Heinzl H, Gelpi E, Hoftberger R, Fischer I, Pipp I, Milenkovic I, Wohrer A, Popovici F, Wolfsberger S, Hainfellner JA, 2008, Ki67 index in intracranial ependymoma: a promising histopathological candidate biomarker. Histopathology 53(1):39– 47., DOI 10.1111/j.1365-2559.2008.03065.x Milde T, Hielscher T, Witt H, Kool M, Mack SC, Deubzer HE, Oehme I, Lodrini M, Benner A, Taylor MD, von Deimling A, Kulozik AE, Pfister SM, Witt O, Korshunov A, 2012, Nestin expression identifies ependymoma patients with poor outcome. Brain Pathol., DOI 10.1111/j.1750-3639.2012.00600.x Ridley L, Rahman R, Brundler MA, Ellison D, Lowe J, Robson K, Prebble E, Luckett I, Gilbertson RJ, Parkes S, Rand V, Coyle B, Grundy RG, 2008, Multifactorial analysis of predictors of outcome in pediatric intracranial ependymoma. Neuro-Oncology 10(5):675– 689., DOI 10.1215/15228517-2008-036 Figarella-Branger D, Civatte M, Bouvier-Labit C, Gouvernet J, Gambarelli D, Gentet JC, Lena G, Choux M, Pellissier JF, 2000, Prognostic factors in intracranial ependymomas in children. J Neurosurg 93(4):605–613., DOI 10.3171/jns.2000.93.4.0605 Jaing TH, Wang HS, Tsay PK, Tseng CK, Jung SM, Lin KL, Lui TN, 2004, Multivariate analysis of clinical prognostic factors in children with intracranial ependymomas. J Neuro-Oncol 68(3):255–261 Kuncova K, Janda A, Kasal P, Zamecnik J, 2009, Immunohistochemical prognostic markers in intracranial ependymomas: systematic review and meta-analysis. Pathol Oncol Res 15(4):605–614., DOI 10.1007/s12253-009-9160-2 Carter M, Nicholson J, Ross F, Crolla J, Allibone R, Balaji V, Perry R, Walker D, Gilbertson R, Ellison DW, 2002, Genetic abnormalities detected in ependymomas by comparative genomic hybridisation. Br J Cancer 86(6):929–939., DOI 10.1038/sj.bjc.6600180 Korshunov A,Witt H, Hielscher T, Benner A, Remke M, Ryzhova M,Milde T, Bender S,Wittmann A, Schottler A, Kulozik AE,Witt O, von Deimling A, Lichter P, Pfister S, 2010, Molecular staging of intracranial ependymoma in children and adults. J Clin Oncol 28(19):3182–3190., DOI 10.1200/JCO.2009.27.3359 Mendrzyk F, Korshunov A, Benner A, Toedt G, Pfister S, Radlwimmer B, Lichter P, 2006, Identification of gains on 1q and epidermal growth factor receptor overexpression as independent prognostic markers in intracranial ependymoma. Clin Cancer Res 12, 7 Pt 1):2070–2079., DOI 10.1158/1078-0432.CCR-05-2363 Monoranu CM, Huang B, Zangen IL, Rutkowski S, Vince GH, Gerber NU, Puppe B, Roggendorf W, 2008, Correlation between 6q25.3 deletion status and survival in pediatric intracranial ependymomas. Cancer Genet Cytogenet 182(1):18–26., DOI 10.1016/j.cancergencyto.2007.12.008 Puget S, Grill J, Valent A, Bieche I, Dantas-Barbosa C, Kauffmann A, Dessen P, Lacroix L, Geoerger B, Job B, Dirven C, Varlet P, Peyre M, Dirks PB, Sainte-Rose C, Vassal G, 2009, Candidate genes on chromosome 9q33–34 involved in the progression of childhood ependymomas. J Clin Oncol 27(11):1884–1892., DOI 10.1200/JCO.2007.15.4195 Witt H,Mack SC, Ryzhova M, Bender S, Sill M, Isserlin R, Benner A, Hielscher T,Milde T, Remke M, Jones DT, Northcott PA, Garzia L, Bertrand KC, Wittmann A, Yao Y, Roberts SS, Massimi L, Van Meter T,WeissWA, Gupta N, GrajkowskaW, Lach B, Cho YJ, von Deimling A, Kulozik AE, Witt O, Bader GD, Hawkins CE, Tabori U, Guha A, Rutka JT, Lichter P, Korshunov A, Taylor MD, Pfister SM, 2011, Delineation of two clinically and molecularly distinct subgroups of posterior fossa ependymoma. Cancer Cell 20(2):143– 157., DOI 10.1016/j.ccr.2011.07.007 Raghunathan A, Wani K, Armstrong TS, Vera-Bolanos E, Fouladi M, Gilbertson R, Gajjar A, Goldman S, Lehman NL, Metellus P, Mikkelsen T, Necesito-Reyes MJ, Omuro A, Packer RJ, Partap S, Pollack IF, Prados MD, Robins HI, Soffietti R, Wu J, Miller CR, Gilbert MR, Aldape KD, Collaborative Ependymoma Research N, 2013, Histological predictors of outcome in ependymoma are dependent on anatomic site within the central nervous system. Brain Pathol 23(5):584–594., DOI 10.1111/bpa.12050 Phi JH, Wang KC, Park SH, Kim IH, Kim IO, Park KD, Ahn HS, Lee JY, Son YJ, Kim SK, 2012, Pediatric infratentorial ependymoma: prognostic significance of anaplastic histology. J Neuro-Oncol 106(3):619–626., DOI 10.1007/s11060-011-0699-x Maret D, Gruzglin E, Sadr MS, Siu V, Shan W, Koch AW, Seidah NG, Del Maestro RF, Colman DR, 2010, Surface expression of precursor N-cadherin promotes tumor cell invasion. Neoplasia 12(12):1066–1080 Lewis-Tuffin LJ, Rodriguez F, Giannini C, Scheithauer B, Necela BM, Sarkaria JN, Anastasiadis PZ, 2010, Misregulated E-cadherin expression associated with an aggressive brain tumor phenotype. PLoS One 5(10):e13665., DOI 10.1371/journal.pone.0013665 Krause G,Winkler L,Mueller SL, Haseloff RF, Piontek J, Blasig IE, 2008, Structure and function of claudins. Biochim Biophys Acta 1778(3):631–645., DOI 10.1016/j.bbamem.2007.10.018 Liebner S, Fischmann A, Rascher G, Duffner F, Grote EH, Kalbacher H, Wolburg H, 2000, Claudin-1 and claudin-5 expression and tight junction morphology are altered in blood vessels of human glioblastoma multiforme. Acta Neuropathol 100(3): 323–331 Takei H, Bhattacharjee MB, Rivera A, Dancer Y, Powell SZ, 2007, New immunohistochemical markers in the evaluation of central nervous system tumors: a review of 7 selected adult and pediatric brain tumors. Arch Pathol Lab Med 131(2):234–241 Wolburg H, Wolburg-Buchholz K, Kraus J, Rascher-Eggstein G, Liebner S, Hamm S, Duffner F, Grote EH, Risau W, Engelhardt B, 2003, Localization of claudin-3 in tight junctions of the bloodbrain barrier is selectively lost during experimental autoimmune encephalomyelitis and human glioblastoma multiforme. Acta Neuropathol 105(6):586–592 Szmydynger-Chodobska J, Pascale CL, Pfeffer AN, Coulter C, Chodobski A, 2007, Expression of junctional proteins in choroid plexus epithelial cell lines: a comparative study. Cerebrospinal Fluid Res 4:11 Schulzke JD, FrommM(2009, Tight junctions: molecular structure meets function. Annals of the New York Academy of Sciences 1165:1–6 Fanning AS, Mitic LL, Anderson JM, 1999, Transmembrane proteins in the tight junction barrier. J Am Soc Nephrol 10(6):1337–1345 Forster C, 2008, Tight junctions and the modulation of barrier function in disease. Histochem Cell Biol 130(1):55–70 Gonzalez-Mariscal L, Betanzos A, Nava P, Jaramillo BE, 2003, Tight junction proteins. Prog Biophys Mol Biol 81(1):1–44 Nordfors K, Haapasalo J, Sallinen PK, Haapasalo H, Soini Y, 2013, Expression of claudins relates to tumour aggressivity, location and recurrence in ependymomas. Histol Histopathol 28(9): 1137–1146 Masseguin C,Mani-Ponset L, Herbute S, Tixier-Vidal A, Gabrion J, 2001, Persistence of tight junctions and changes in apical structures and protein expression in choroid plexus epithelium of rats after short-term head-down tilt. J Neurocytol 30(5):365–377 Mathew TC, 2008, Regional analysis of the ependyma of the third ventricle of rat by light and electron microscopy. Anat Histol Embryol 37(1):9–18 Wolburg H, Wolburg-Buchholz K, Liebner S, Engelhardt B, 2001, Claudin-1, claudin-2 and claudin-11 are present in tight junctions of choroid plexus epithelium of the mouse. Neurosci Lett 307(2):77–80 Dahiya S, Lee d Y, Gutmann DH, 2011, Comparative characterization of the human and mouse third ventricle germinal zones. J Neuropathol Exp Neurol 70(7):622–633., DOI 10.1097/NEN.0b013 e31822200aa Vigh B, Manzano e Silva MJ, Frank CL, Vincze C, Czirok SJ, Szabo A, Lukats A, Szel A, 2004, The system of cerebrospinal fluid-contacting neurons. Its supposed role in the nonsynaptic signal transmission of the brain. Histol Histopathol 19(2):607–628 Vigh B, Vigh-Teichmann I, 1998, Actual problems of the cerebrospinal fluid-contacting neurons. Microsc Res Tech 41(1):57–83 Patonai A, Erdelyi-Belle B, Korompay A, Somoracz A, Straub BK, Schirmacher P, Kovalszky I, Lotz G, Kiss A, Schaff Z, 2011, Claudins and tricellulin in fibrolamellar hepatocellular carcinoma. Virchows Arch 458(6):679–688., DOI 10.1007/s00428-011-1077-y Schnitzer J, FrankeWW, SchachnerM(1981, Immunocytochemical demonstration of vimentin in astrocytes and ependymal cells of developing and adultmouse nervous system. J Cell Biol 90(2):435–447 Palm T, Figarella-Branger D, Chapon F, Lacroix C, Gray F, Scaravilli F, Ellison DW, Salmon I, Vikkula M, Godfraind C, 2009, Expression profiling of ependymomas unravels localization and tumor grade-specific tumorigenesis. Cancer 115(17):3955–3968 Taylor MD, Poppleton H, Fuller C, Su X, Liu Y, Jensen P, Magdaleno S, Dalton J, Calabrese C, Board J, Macdonald T, Rutka J, Guha A, Gajjar A, Curran T, Gilbertson RJ, 2005, Radial glia cells are candidate stem cells of ependymoma. Cancer Cell 8(4):323–335., DOI 10.1016/j.ccr.2005.09.001 Rogers HA, Kilday JP, Mayne C, Ward J, Adamowicz-Brice M, Schwalbe EC, Clifford SC, Coyle B, Grundy RG, 2012, Supratentorial and spinal pediatric ependymomas display a hypermethylated phenotype which includes the loss of tumor suppressor genes involved in the control of cell growth and death. Acta Neuropathol 123(5):711–725., DOI 10.1007/s00401-011-0904-1 Perez-Ramirez M, Hernandez-Jimenez AJ, Guerrero-Guerrero A, Benadon-Darszon E, Perezpena-Diazconti M, Siordia-Reyes AG, Garcia-Mendez A, de Leon FC-P, Salamanca-Gomez FA, Garcia- Hernandez N, 2016, Genomics and epigenetics: A study of ependymomas in pediatric patients. Clin Neurol Neurosurg 144: 53–58., DOI 10.1016/j.clineuro.2016.02.041 Rajeshwari M, Sharma MC, Kakkar A, Nambirajan A, Suri V, Sarkar C, Singh M, Saran RK, Gupta RK, 2016, Evaluation of chromosome 1q gain in intracranial ependymomas. J Neuro- Oncol 127(2):271–278., DOI 10.1007/s11060-015-2047-z Lippoldt A, Liebner S, Andbjer B, Kalbacher H,Wolburg H, Haller H, Fuxe K, 2000, Organization of choroid plexus epithelial and endothelial cell tight junctions and regulation of claudin-1, −2 and −5 expression by protein kinase C. Neuroreport 11(7):1427–1431 Lee d Y, Gianino SM, Gutmann DH, 2012, Innate neural stem cell heterogeneity determines the patterning of glioma formation in children. Cancer Cell 22(1):131–138., DOI 10.1016/j.ccr.2012.05.036 Sharma MK, Mansur DB, Reifenberger G, Perry A, Leonard JR, Aldape KD, Albin MG, Emnett RJ, Loeser S, Watson MA, Nagarajan R, Gutmann DH, 2007, Distinct genetic signatures among pilocytic astrocytomas relate to their brain region origin. Cancer Res 67(3):890–900., DOI 10.1158/0008-5472.CAN-06-0973 Gibson P, Tong Y, Robinson G, ThompsonMC, Currle DS, Eden C, Kranenburg TA, Hogg T, Poppleton H, Martin J, Finkelstein D, Pounds S, Weiss A, Patay Z, Scoggins M, Ogg R, Pei Y, Yang ZJ, Brun S, Lee Y, Zindy F, Lindsey JC, Taketo MM, Boop FA, Sanford RA, Gajjar A, Clifford SC, Roussel MF, McKinnon PJ, Gutmann DH, Ellison DW, Wechsler-Reya R, Gilbertson RJ, 2010, Subtypes of medulloblastoma have distinct developmental origins. Nature 468(7327):1095–1099., DOI 10.1038/nature09587 EllisonDW, KocakM, Figarella-Branger D, Felice G, Catherine G, Pietsch T, Frappaz D, Massimino M, Grill J, Boyett JM, Grundy RG, 2011, Histopathological grading of pediatric ependymoma: reproducibility and clinical relevance in European trial cohorts. J Negat Results Biomed 10(7)., DOI 10.1186/1477-5751-10-7 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 245 EP 252 DI 10.1007/s12253-016-0084-3 PG 8 ER PT J AU Rusz, O Pal, M Szilagyi, Rovo, L Varga, Z Tomisa, B Fabian, G Kovacs, L Nagy, O Mozes, P Reisz, Z Tiszlavicz, L Deak, P Kahan, Zs AF Rusz, Orsolya Pal, Margit Szilagyi, Eva Rovo, Laszlo Varga, Zoltan Tomisa, Bernadett Fabian, Gabriella Kovacs, Levente Nagy, Olga Mozes, Petra Reisz, Zita Tiszlavicz, Laszlo Deak, Peter Kahan, Zsuzsanna TI The Expression of Checkpoint and DNA Repair Genes in Head and Neck Cancer as Possible Predictive Factors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Head and neck cancer; Chemosensitivity; Radiosensitivity; DNA damage response; Gene expression ID Head and neck cancer; Chemosensitivity; Radiosensitivity; DNA damage response; Gene expression AB DNA damage response failure may influence the efficacy of DNA-damaging treatments. We determined the expression of 16 genes involved in distinct DNA damage response pathways, in association with the response to standard therapy. Twenty patients with locoregionally advanced, squamous cell head and neck carcinoma were enrolled. The treatment included induction chemotherapy (iChT) with docetaxel, cisplatin and 5-fluorouracil followed by concomitant chemoradiotherapy (ChRT) or radiotherapy (RT) alone. The volumetric metabolic therapeutic response was determined by [18F]FDG-PET/CT. In the tumor and matched normal tissues collected before treatment, the gene expressions were examined via the quantitative real-time polymerase chain reaction (qRT-PCR). The down-regulation of TP53 was apparently associated with a poor response to iChT, its up-regulation with complete regression in 2 cases. 7 cases with down-regulated REV1 expression showed complete regression after ChRT/RT, while 1 case with REV1 overexpression was resistant to RT. The overexpression of WRN was an independent predictor of tumor relapse. Our results suggest that an altered expression of REV1 predicts sensitivity to RT, while WRN overexpression is an unfavorable prognostic factor. C1 [Rusz, Orsolya] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Pal, Margit] University of Szeged, Department of Medical Chemistry, Temesvari krt. 62, H-6726 Szeged, Hungary. [Szilagyi, Eva] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Rovo, Laszlo] Szegedi Tudomanyegyetem, Ful-Orr-Gegeszeti Klinika, Tisza Lajos krt. 111, H-6725 Szeged, Hungary. [Varga, Zoltan] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Tomisa, Bernadett] University of Szeged, Department of Medical Chemistry, Temesvari krt. 62, H-6726 Szeged, Hungary. [Fabian, Gabriella] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Kovacs, Levente] University of Szeged, Department of Medical Chemistry, Temesvari krt. 62, H-6726 Szeged, Hungary. [Nagy, Olga] University of Szeged, Department of Medical Chemistry, Temesvari krt. 62, H-6726 Szeged, Hungary. [Mozes, Petra] Technische Universitat Munchen, Klinikum rechts der Isar, Department of Radiation Oncology, Ismaninger Straße 22, 81675 Munich, Germany. [Reisz, Zita] University of Szeged, Department of Pathology, Allomas utca 2, H-6720 Szeged, Hungary. [Tiszlavicz, Laszlo] University of Szeged, Department of Pathology, Allomas utca 2, H-6720 Szeged, Hungary. [Deak, Peter] University of Szeged, Department of Medical Chemistry, Temesvari krt. 62, H-6726 Szeged, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. RP Rusz, O (reprint author), University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. EM rusz.orsolya@med.u-szeged.hu CR Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, et al., 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403., DOI 10.1016/j. ejca.2012.12.027 PosnerMR, Hershock DM, Blajman CR, et al., 2007, Cisplatin and fluorouracil alone or with docetaxel in head and neck cancer. N Engl J Med 357:1705–1715., DOI 10.1056/NEJMoa070956 Vermorken JB, Remenar E, van Herpen C, et al., 2007, Cisplatin, fluorouracil, and docetaxel in unresectable head and neck cancer. N Engl J Med 357:1695–1704., DOI 10.1056/NEJMoa071028 Hitt R, Grau JJ, Lopez-Pousa A, et al., 2014, A randomized phase III trial comparing induction chemotherapy followed by chemoradiotherapy versus chemoradiotherapy alone as treatment of unresectable head and neck cancer. Ann Oncol 25:216–225., DOI 10.1093/annonc/mdt461 Helleday T, Petermann E, Lundin C, Hodgson B, Sharma RA, 2008, DNA repair pathways as targets for cancer therapy. Nat Rev Cancer 8:193–204., DOI 10.1038/nrc2342 Curtin NJ, 2012, DNA repair dysregulation from cancer driver to therapeutic target. Nat Rev Cancer 12:801–817., DOI 10.1038/nrc3399 Juhasz S, Balogh D, Hajdu I, et al., 2012, Characterization of human Spartan/C1orf124, an ubiquitin-PCNA interacting regulator of DNA damage tolerance. Nucleic Acids Res 40:10795–10808., DOI 10.1093/nar/gks850 Lange SS, Takata K, Wood RD, 2011, DNA polymerases and cancer. Nat Rev Cancer 11:96–110., DOI 10.1038/nrc2998 Kitano K, 2014, Structural mechanisms of human RecQ helicases WRN and BLM. Front Genet 5:366., DOI 10.3389/fgene.2014.00366 Parrilla-Castellar ER, Arlander SJH, Karnitz L, 2004, Dial 9–1-1 for DNA damage: The Rad9-Hus1-Rad1, 9–1–1, clamp complex. DNA Repair, Amst, 3:1009–1014., DOI 10.1016/j. dnarep.2004.03.032 Niida H, Nakanishi M, 2006, DNA damage checkpoints in mammals. Mutagenesis 21:3–9., DOI 10.1093/mutage/gei063 McGrogan BT, Gilmartin B, Carney DN, McCann A, 2008, Taxanes, microtubules and chemoresistant breast cancer. Biochim Biophys Acta 1785:96–132., DOI 10.1016/j.bbcan.2007.10.004 Miller AB, Hoogstraten B, StaquetM,Winkler A, 1981, Reporting results of cancer treatment. Cancer 47:207–214 Wiggenraad R, Mast M, van Santvoort J, Hoogendoorn M, Struikmans H, 2005, ConPas: a 3-D conformal parotid gland-sparing irradiation technique for bilateral neck treatment as an alternative to IMRT. Strahlenther Onkol 181:673–682., DOI 10.1007/s00066-005-1413-8 Nikolenyi A, Uhercsak G, Csenki M, et al., 2012, Tumour topoisomerase II alpha protein expression and outcome after adjuvant dose-dense anthracycline-based chemotherapy. Pathol Oncol Res 18:61–68., DOI 10.1007/s12253-011-9417-4 Rozen S, Skaletsky H, 2000, Primer3 on the WWW for general users and for biologist programmers. Methods Mol Biol 132:365–386 Lallemant B, Evrard A, Combescure C, et al., 2009, Reference gene selection for head and neck squamous cell carcinoma gene expression studies. BMC Mol Biol 10:78., DOI 10.1186/1471- 2199-10-78 Sharma S, Canman CE, 2012, REV1 and DNA polymerase zeta in DNA interstrand crosslink repair. Environ Mol Mutagen 53:725– 740., DOI 10.1002/em.21736 Lin X, Okuda T, Trang J, Howell SB, 2006, Human REV1 modulates the cytotoxicity and mutagenicity of cisplatin in human ovarian carcinoma cells. Mol Pharmacol 69:1748–1754., DOI 10.1124 /mol.105.020446 Xie K, Doles J, Hemann MT, Walker GC, 2010, Error-prone translesion synthesis mediates acquired chemoresistance. Proc Natl Acad Sci U S A 107:20792–20797., DOI 10.1073 /pnas.1011412107 Okada T, Sonoda E, Yoshimura M, et al., 2005, Multiple roles of vertebrate REV genes in DNA repair and recombination. Mol Cell Biol 25:6103–6111., DOI 10.1128/MCB.25.14.6103-6111.2005 Moeller BJ, Yordy JS, Williams MD, et al., 2011, DNA repair biomarker profiling of head and neck cancer: Ku80 expression predicts locoregional failure and death following radiotherapy. Clin Cancer Res 17:2035–2043., DOI 10.1158/1078- 0432.CCR-10-2641 Lee SY, Park HR, ChoNH, et al., 2013, Identifying genes related to radiation resistance in oral squamous cell carcinoma cell lines. Int J Oral Maxillofac Surg 42:169–176., DOI 10.1016/j.ijom.2012.10.022 Horiuchi C, Taguchi T, Yoshida T, et al., 2008, Early assessment of clinical response to concurrent chemoradiotherapy in head and neck carcinoma using fluoro-2-deoxy-d-glucose positron emission tomography. Auris Nasus Larynx 35:103–108., DOI 10.1016/j. anl.2007.05.003 Won HS, Lee YS, Jeon EK, et al., 2014, Clinical outcome of induction chemotherapy in locally advanced head and neck squamous cell carcinoma. Anticancer Res 34:5709–5714 Argiris A, 2005, Induction chemotherapy for head and neck cancer: will history repeat itself? J Natl Compr Cancer Netw 3:393–403 Bhatia KSS, King AD, Yu K-H, et al., 2010, Does primary tumour volumetry performed early in the course of definitive concomitant chemoradiotherapy for head and neck squamous cell carcinoma improve prediction of primary site outcome? Br J Radiol 83:964– 970., DOI 10.1259/bjr/27631720 Hoebers FJP, Pameijer FA, de Bois J, et al., 2008, Prognostic value of primary tumor volume after concurrent chemoradiation with daily low-dose cisplatin for advanced-stage head and neck carcinoma. Head Neck 30:1216–1223., DOI 10.1002/hed.20865 Gudkov AV, Komarova EA, 2003, The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer 3:117– 129., DOI 10.1038/nrc992 Shiga H, Heath EI, Rasmussen et al, 1999, Prognostic value of p53, glutathione S-transferase pi, and thymidylate synthase for neoadjuvant cisplatin-based chemotherapy in head and neck cancer. Clin Cancer Res 5:4097–4104 Falette N, Paperin MP, Treilleux I, et al., 1998, Prognostic value of P53 gene mutations in a large series of node-negative breast cancer patients. Cancer Res 58:1451–1455 Cabelguenne A, Blons H, deWaziers I, et al., 2000, p53 alterations predict tumor response to neoadjuvant chemotherapy in head and neck squamous cell carcinoma: a prospective series. J Clin Oncol 18:1465–1473 Santana P, Pena LA, Haimovitz-Friedman A, et al., 1996, Acid sphingomyelinase-deficient human lymphoblasts and mice are defective in radiation-induced apoptosis. Cell 86: 189–199 Chmura SJ, Mauceri HJ, Advani S, et al., 1997, Decreasing the apoptotic threshold of tumor cells through protein kinase C inhibition and sphingomyelinase activation increases tumor killing by ionizing radiation. Cancer Res 57:4340–4347 Hendry JH,West CM(1997, Apoptosis and mitotic cell death: their relative contributions to normal-tissue and tumour radiation response. Int J Radiat Biol 71:709–719 Jung M, Notario V, Dritschilo A, 1992, Mutations in the p53 gene in radiation-sensitive and -resistant human squamous carcinoma cells. Cancer Res 52:6390–6393 DiBiase SJ, Guan J, Curran WJ, Iliakis G, 1999, Repair of DNA double-strand breaks and radiosensitivity to killing in an isogenic group of p53 mutant cell lines. Int J Radiat Oncol Biol Phys 45: 743–751 Bunz F, Hwang PM, Torrance C, et al., 1999, Disruption of p53 in human cancer cells alters the responses to therapeutic agents. J Clin Invest 104:263–269., DOI 10.1172/JCI6863 Levine AJ, 2009, The common mechanisms of transformation by the small DNA tumor viruses: the inactivation of tumor suppressor gene products: p53. Virology 384:285–293., DOI 10.1016/j. virol.2008.09.034 Chau NG, Rabinowits G, Haddad RI, 2014, Human papillomavirusassociated oropharynx cancer, HPV-OPC): treatment options. Curr Treat Options in Oncol 15:595–610., DOI 10.1007/s11864-014-0309-1 Arai A, Chano T, Futami K, et al., 2011, RECQL1 and WRN proteins are potential therapeutic targets in head and neck squamous cell carcinoma. Cancer Res 71:4598–4607., DOI 10.1158/0008- 5472.CAN-11-0320 Wang L, Xie L, Wang J, Shen J, Liu B, 2013, Correlation between the methylation of SULF2 and WRN promoter and the irinotecan chemosensitivity in gastric cancer. BMC Gastroenterol 13:173., DOI 10.1186/1471-230X-13-173 Agrelo R, Cheng W-H, Setien F, et al., 2006, Epigenetic inactivation of the premature aging Werner syndrome gene in human cancer. Proc Natl Acad Sci U S A 103:8822–8827., DOI 10.1073/pnas.0600645103 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 253 EP 264 DI 10.1007/s12253-016-0088-z PG 12 ER PT J AU Chen, J Hu, L Wang, J Zhang, F Chen, J Xu, G Wang, Y Pan, Q AF Chen, Jian Hu, Lijuan Wang, Junjun Zhang, Fan Chen, Jie Xu, Gang Wang, Yumin Pan, Qinshi TI Low Expression LncRNA TUBA4B is a Poor Predictor of Prognosis and Regulates Cell Proliferation in Non-Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; lncRNAs; TUBA4B; Prognosis ID Non-small cell lung cancer; lncRNAs; TUBA4B; Prognosis AB We aimed to unveil the clinical roles and biological function of lncRNA TUBA4B in on-small cell lung cancer (NSCLC). The relative expression level of TUBA4B was estimated by qPCR in 114 pairs of NSCLC and NT samples and the relation of TUBA4B to clinical data of NSCLC patients was analyzed. We found TUBA4B was lower expressed in NSCLC and five cell lines. The lower expression of TUBA4B was remarkably correlated with advanced TNM stage and lymph node metastasis and served as a predictor for overall survival of NSCLC. After overexpression of TUBA4B, cell proliferation ability of A549 and NCI-H1299 remarkably decreased. Our study ascertained low expression TUBA4B in NSCLC tissue, cell lines and is a poor predictor for prognosis and can regulate cell proliferation in NSCLC. C1 [Chen, Jian] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang province, China. [Hu, Lijuan] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang province, China. [Wang, Junjun] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang province, China. [Zhang, Fan] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang province, China. [Chen, Jie] The First Affiliated Hospital of Wenzhou Medical University, Department of Intensive Care Unit, 325000 Wenzhou, Zhejiang province, China. [Xu, Gang] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang province, China. [Wang, Yumin] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang province, China. [Pan, Qinshi] The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, Zhejiang province, China. RP Wang, Y (reprint author), The First Affiliated Hospital of Wenzhou Medical University, Department of Laboratory Medicine, 325000 Wenzhou, China. EM wym0577@163.com CR Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, Feuer EJ, Thun MJ. Cancer statistics, 2005. CA: a cancer journal for clinicians. 2005;55:10–30. Gridelli C, Rossi A, Maione P, 2003, Treatment of non-small-cell lung cancer: state of the art and development of new biologic agents. Oncogene 22:6629–6638 Stewart DJ, 2010, Tumor and host factors that may limit efficacy of chemotherapy in non-small cell and small cell lung cancer. Crit Rev Oncol Hematol 75:173–234 Chen CH, Lai JM, Chou TY, Chen CY, Su LJ, Lee YC, Cheng TS, Hong YR, Chou CK,Whang-Peng J,Wu YC, Huang CY. VEGFA upregulates FLJ10540 and modulates migration and invasion of lung cancer via PI3K/AKT pathway. PLoS One 2009;4:e5052. Ogawa E, Takenaka K, Katakura H, Adachi M, Otake Y, Toda Y, Kotani H, Manabe T, Wada H, Tanaka F, 2008, Perimembrane aurora-a expression is a significant prognostic factor in correlation with proliferative activity in non-small-cell lung cancer, NSCLC). Ann Surg Oncol 15:547–554 Rachet B, Woods LM, Mitry E, Riga M, Cooper N, Quinn MJ, Steward J, Brenner H, Esteve J, Sullivan R, Coleman MP. Cancer survival in England and Wales at the end of the twentieth century. Br J Cancer 2008;99 Suppl 1:S2–10. Fu X, Ravindranath L, Tran N, Petrovics G, Srivastava S, 2006, Regulation of apoptosis by a prostate-specific and prostate cancerassociated noncoding gene, PCGEM1. DNA Cell Biol 25:135–141 Gupta RA, Shah N,Wang KC, Kim J, Horlings HM,Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL,Wang Y, Brzoska P, Kong B, Li R, West RB, van de Vijver MJ, Sukumar S, Chang HY, 2010, Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464:1071–1076 Zhang H, Chen Z, Wang X, Huang Z, He Z, Chen Y. Long noncoding RNA: a new player in cancer. J Hematol Oncol 2013;6:37. Hauptman N, Glavac D, 2013, Long non-coding RNA in cancer. Int J Mol Sci 14:4655–4669 ChenG,Wang Z,Wang D, Qiu C, Liu M, Chen X, Zhang Q,Yan G, Cui Q, 2013, LncRNADisease: a database for long-non-coding RNA-associated diseases. Nucleic Acids Res 41:D983–D986 Gibb EA, Brown CJ, Lam WL, 2011, The functional role of long non-coding RNA in human carcinomas. Mol Cancer 10:–38 Ji P, Diederichs S, Wang W, Boing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, Thomas M, Berdel WE, Serve H, Muller-Tidow C, 2003, MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene 22:8031–8041 Thai P, Statt S, Chen CH, Liang E, Campbell C, Wu R, 2013, Characterization of a novel long noncoding RNA, SCAL1, induced by cigarette smoke and elevated in lung cancer cell lines. Am J Respir Cell Mol Biol 49:204–211 YangY, LiH,Hou S, Hu B, Liu J,Wang J, 2013, The noncodingRNA expression profile and the effect of lncRNA AK126698 on cisplatin resistance in non-small-cell lung cancer cell. PLoS One 8:–e65309 Han L, Kong R, Yin DD, Zhang EB, TP X, DeW, Shu YQ, 2013, Low expression of long noncoding RNA GAS6-AS1 predicts a poor prognosis in patients with NSCLC. Med Oncol 30:–694 Ren S, Peng Z, Mao JH,YuY,Yin C,Gao X, Cui Z, Zhang J,Yi K, Xu W, Chen C, Wang F, Guo X, Lu J, Yang J, Wei M, Tian Z, Guan Y, TangL,XuC,WangL,GaoX, TianW,Wang J, YangH,Wang J, Sun Y, 2012, RNA-seq analysis of prostate cancer in the Chinese population identifies recurrent gene fusions, cancer-associated long noncoding RNAs and aberrant alternative splicings. Cell Res 22:806–821 DrobnikW, Liebisch G, Biederer C, Tr mbach B, Rogler G, Muller P, Schmitz G, 1999, Growth and cell cycle abnormalities of fibroblasts from Tangier disease patients. Arterioscler ThrombVasc Biol 19:28–38 Wang KC, Chang HY, 2011, Molecular mechanisms of long noncoding RNAs. Mol Cell 43:904–914 Khachane AN,HarrisonPM. Miningmammalian transcript data for functional long non-coding RNAs. PLoS One 2010;5:e10316. Loh YH, Wu Q, Chew JL, Vega VB, Zhang W, Chen X, Bourque G, George J, Leong B, Liu J,Wong KY, Sung KW, Lee CW, Zhao XD, Chiu KP, Lipovich L, Kuznetsov VA, Robson P, Stanton LW, Wei CL, Ruan Y, Lim B, Ng HH, 2006, The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nat Genet 38:431–440 Mattick JS, Gagen MJ, 2001, The evolution of controlled multitasked gene networks: the role of introns and other noncoding RNAs in the development of complex organisms. Mol Biol Evol 18:1611–1630 Mattick JS, 2010, Linc-ing long noncoding RNAs and enhancer function. Dev Cell 19:485–486 Orom UA, Derrien T, Beringer M, Gumireddy K, Gardini A, Bussotti G, Lai F, Zytnicki M, Notredame C, Huang Q, Guigo R, Shiekhattar R, 2010, Long noncoding RNAs with enhancer-like function in human cells. Cell 143:46–58 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 265 EP 270 DI 10.1007/s12253-016-0089-y PG 6 ER PT J AU Mahmoudian, AR Abbaszadegan, RM Gholamin, M AF Mahmoudian, Alsadat Reihaneh Abbaszadegan, Reza Mohammad Gholamin, Mehran TI Applying Subtractive Hybridization Technique to Enrich and Amplify Tumor-Specific Transcripts of Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Subtractive hybridization; ESCC; Enrichment; Tumor-specific transcripts; SMART ID Subtractive hybridization; ESCC; Enrichment; Tumor-specific transcripts; SMART AB Subtractive hybridization (SH) as an efficient and powerful approach can be applied to isolate differentially expressed transcripts as well as detect of involved mRNAs in various cellular processes, particularly diseases and malignancies. This procedure leads to the enrichment of specific low copy transcripts of tumor cells. Having developed a new approach for SH to isolate tumor specific transcripts, we facilitated discovery of uniquely expressed genes in esophageal squamous cell carcinoma (ESCC). Total RNA was extracted from the fresh tumoral and their adjacent normal tissues, and purified using the Switch Mechanism At the 5ʹ end of Reverse Transcript (SMART) method. Following cDNA synthesis of normal mRNAs using magnetic beads, it was hybridized with tumor mRNAs. To enhance efficiency of subtraction, hybridization was repeated three rounds. Finally, amplification of subtracted tumor-specific transcripts was carried out using in vitro transcription. The subtracted tumoral mRNAs was analyzed quantitatively using real-time PCR for both tumorspecific and housekeeping genes. The subtracted mRNA was confirmed as tumor-specific mRNA pool using RT-PCR and quantitative real-time PCR assessment. The elevated level of tumor-specific transcripts such as MAGE-A4 and CD44 as well as declined copy number of housekeeping genes such as GAPDH, β actin and β2-microglobulin, were confirmed in subtracted tumoral mRNA. The presence of tumor genes was confirmed after the SH procedure. The designed SH method in combination with SMART technique can isolate and amplify high quality tumor-specific transcripts even from small amount of tumor tissues. Removal of common transcripts from the extracted tumoral mRNAs using SH, leads to the enrichment of tumor-specific transcripts. The isolated transcripts are of interest because of their probable roles in ESCC progression and development. In addition, these tumor-specific mRNAs can be applied for future vaccine cancer studies. C1 [Mahmoudian, Alsadat Reihaneh] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, Bu-Ali Sq, Khorasan Razavi, 9196773117 Mashhad, Iran. [Abbaszadegan, Reza Mohammad] Mashhad University of Medical Sciences, Medical School, Medical Genetics Research CenterMashhad, Iran. [Gholamin, Mehran] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, Bu-Ali Sq, Khorasan Razavi, 9196773117 Mashhad, Iran. RP Gholamin, M (reprint author), Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human Genetics, 9196773117 Mashhad, Iran. EM GholaminM@mums.ac.ir CR Moghbeli M, Moghbeli F, Forghanifard MM, Garayali A, Abbaszadegan MR, 2013, Cancer stem cell markers in esophageal cancer. Am J Cancer Sci 2(1):37–50 Taleb S, Abbaszadegan MR, Moghbeli M, Roudbari NH, Forghanifard MM, 2014, HES1 as an independent prognostic marker in esophageal squamous cell carcinoma. J Gastrointest Cancer 45(4):466–471 Gholamin M, Moaven O, Memar B, Farshchian M, Naseh H, Malekzadeh R, et al., 2009, Overexpression and interactions of interleukin-10, transforming growth factor, and vascular endothelial growth factor in esophageal squamous cell carcinoma. World J Surg 33(7):1439–1445 GholaminM, Moaven O, Farshchian M,Mahmoudi M, Sankian M, Memar B, et al., 2010, Induction of cytotoxic T lymphocytes primed with tumor RNA-loaded dendritic cells in esophageal squamous cell carcinoma: preliminary step for DC vaccine design. BMC Cancer 10(1):261 Kahkhaie KR, Moaven O, Abbaszadegan MR, Montazer M, Gholamin M, 2014, Specific MUC1 splice variants are correlated with tumor progression in esophageal cancer. World J Surg 38(8): 2052–2057 Napier KJ, Scheerer M, Misra S, 2014, Esophageal cancer: A Review of epidemiology, pathogenesis, staging workup and treatment modalities. World J Gastrointest Oncol 6(5):112 Kurtz DM, Gambhir SS, 2014, Tracking cellular and immune therapies in cancer. Emerging Applications of Molecular Imaging to Oncology, 124:257-296 ForghanifardMM, GholaminM, Farshchian M,Moaven O,Memar B, Forghani MN, et al., 2011, Cancer-testis gene expression profiling in esophageal squamous cell carcinoma: identification of specific tumor marker and potential targets for immunotherapy. Cancer Biol Ther 12(3):191–197 Bringmann A, Held SAE, Heine A, Brossart P, 2010, RNA vaccines in cancer treatment. BioMed Res Int 2010 Moingeon P, 2001, Cancer vaccines. Vaccine 19(11):1305–1326 Mitchell DA, Nair SK, 2000, RNA-transfected dendritic cells in cancer immunotherapy. J Clin Investig 106(9):1065 Forghanifard MM, Amani J, Gheybi E, Abbaszadegan MR, 2012, In silico analysis of chimeric polytope of cancer/testis antigens for dendritic cell-based immune-gene therapy applications. Gene Ther Mol Biol 14:87–96 Silva JM, Videira M, Gaspar R, Preat V, Florindo HF, 2013, Immune system targeting by biodegradable nanoparticles for cancer vaccines. J Control Release 168(2):179–199 Matera L, 2010, The choice of the antigen in the dendritic cellbased vaccine therapy for prostate cancer. Cancer Treat Rev 36(2):131–141 Cronwright G, Le Blanc K, Gotherstrom C, Darcy P, Ehnman M, Brodin B, 2005, Cancer/testis antigen expression in human mesenchymal stem cells: down-regulation of SSX impairs cell migration and matrix metalloproteinase 2 expression. Cancer Res 65(6): 2207–2215 Gholamin M, Moaven O, Farshchian M, Rajabi-Mashhadi MT, Mahmoudi M, Sankian M, et al., 2010, Highly efficient transfection of dendritic cells derived from esophageal squamous cell carcinoma patient: optimization with green fluorescent protein and validation with tumor RNA as a tool for immuno-genetherapy. Iran J Biotechnol 8(2):121–126 Garg NK, Dwivedi P, Prabha P, Tyagi RK, 2013, RNA pulsed dendritic cells: an approach for cancer immunotherapy. Vaccine 31(8):1141–1156 Sagerstrm CG, Sun BI, Sive HL, 1997, Subtractive cloning: past, present, and future. Annu Rev Biochem 66(1):751–783 Wu N,Matand K,Williams S, 2009, A novel mRNA level subtraction method for quick identification of target-orientated uniquely expressed genes between peanut immature pod and leaf. Biological Procedures Online 12(1):44–55 Cho T-J, Park S-S, 1998, A simulation of subtractive hybridization. Nucleic Acids Res 26(6):1440–1448 Lin T-Y, Ying, S-Y, 2003, Subtractive hybridization for the identification of differentially expressed genes using uraci-DNA glycosylase and mung-bean nuclease. Generation of cDNA Libraries. Springer, p 239-251 Wang Z, Jones, MGK, 2003, cDNA generation on paramagnetic beads. Generation of cDNA Libraries. Springer, p 25-31 Barooei R, Mahmoudian RA, Abbaszadegan MR, Mansouri A, Gholamin M, 2015, Evaluation of thymic stromal lymphopoietin, TSLP, and its correlation with lymphatic metastasis in human gastric cancer. Med Oncol 32(8):1–8 Moghbeli M, Forghanifard MM, Aarabi A, Mansourian A, Abbaszadegan MR, 2014, Clinicopathological sex-related relevance of Musashi1 mRNA expression in esophageal squamous cell carcinoma patients. Pathol Oncol Res 20(2):427–433 Alsadat Mahmoudian R, Abbaszadegan MR, Mansouri A, Gholamin M, 2015, Amplification of Tumor Transcripts from Limited Quantity of Esophageal Squamous Cell Carcinoma Tissue Samples. Am J Cancer Sci 4(1):54–62 Mansouri A, Foroughmand AM, Abbaszadegan MR, Memar B, Mahmoudian RA, Gholamin M, 2015, Expression analysis of CD44 isoforms S and V3, in patients with esophageal squamous cell carcinoma. Iran J Basic Med Sci 18(4):380 Forghanifard MM, Gholamin M, Moaven O, Farshchian M, Ghahraman M, Aledavood A, et al., 2014, Neoantigen in esophageal squamous cell carcinoma for dendritic cell-based cancer vaccine development. Med Oncol 31(10):1–7 Nair SK, Morse M, Boczkowski D, Cumming RI, Vasovic L, Gilboa E, et al., 2002, Induction of tumor-specific cytotoxic T lymphocytes in cancer patients by autologous tumor RNAtransfected dendritic cells. Ann Surg 235(4):540 Hossein Naseh M, Bahram Memar M, Bagheri R, MontazerMD M, AbbaszadeganPhDMR, 2013, A Cancer-array approach elucidates the immune escape mechanism and defects in the DNA repair system in esophageal squamous cell carcinoma. Arch Iran Med 16(8): 463 Amatschek S, Koenig U, Auer H, Steinlein P, Pacher M, Gruenfelder A, et al., 2004, Tissue-wide expression profiling using cDNA subtraction and microarrays to identify tumor-specific genes. Cancer Res 64(3):844–856 Even-Desrumeaux K, Baty D, Chames P, 2011, State of the art in tumor antigen and biomarker discovery. Cancer 3(2):2554–2596 Srinivasan R, Wolchok JD, 2004, Tumor antigens for cancer immunotherapy: therapeutic potential of xenogeneic DNAvaccines. J Transl Med 2(1):12 Heiser A, Dahm P, Yancey DR, Maurice MA, Boczkowski D, Nair SK, et al., 2000, Human dendritic cells transfected with RNA encoding prostate-specific antigen stimulate prostate-specific CTL responses in vitro. J Immunol 164(10):5508–5514 Muller MR, Grunebach F, Nencioni A, Brossart P, 2003, Transfection of dendritic cells with RNA induces CD4-and CD8- mediated Tcell immunity against breast carcinomas and reveals the immunodominance of presented T cell epitopes. J Immunol 170(12):5892–5896 Gilboa E, Vieweg J, 2004, Cancer immunotherapy with mRNA transfected dendritic cells. Immunol Rev 199(1):251–263 Caspi RR, 2008, Immunotherapy of autoimmunity and cancer: the penalty for success. Nat Rev Immunol 8(12):970–976 Yang F, Yang X-F, 2005, New concepts in tumor antigens: their significance in future immunotherapies for tumors. Cell Mol Immunol 2(5):331–341 Ghafouri-Fard S, Modarressi M-H, 2009, Cancer-testis antigens: potential targets for cancer immunotherapy. Arch Iran Med 12(4): 395–404 Amos SM, Duong CPM, Westwood JA, Ritchie DS, Junghans RP, Darcy PK, et al., 2011, Autoimmunity associated with immunotherapy of cancer. Blood 118(3):499–509 Kaufman HL, Wolchok JD, 2006, Is tumor immunity the same thing as autoimmunity? Implications for cancer immunotherapy. J Clin Oncol 24(15):2230–2232 Blumberg B, Belmonte, JCI., 1999, Subtractive hybridization and construction of cDNA libraries. Molecular Embryology. Springer, p 555–574. Mishra RN, Ramesha A, Kaul T, Nair S, Sopory SK, Reddy MK, 2005, A modified cDNA subtraction to identify differentially expressed genes from plants with universal application to other eukaryotes. Anal Biochem 345(1):149–157 D’Urso OF, D’Urso PI, Storelli C, Marsigliante S, 2009, Solid phase subtractive cloning in differentially expressed genes identification. Mol Biol Rep 37(3):1435–1438 Byers RJ, Hoyland JA, Dixon J, Freemont AJ, 2000, Subtractive hybridization genetic takeaways and the search for meaning. Int J Exp Pathol 81(6):391–404 D’Urso OF, D’Urso PI, Storelli C, Marsigliante S, 2010, Solid phase subtractive cloning in differentially expressed genes identification. Mol Biol Rep 37(3):1435–1438 Smith CL, Bouchard J, Surdi G, Nguyen G, Pimpis K, Tolstoi LG, 2000, Comparative genomics: differential display and subtractive hybridization. Encycl Anal Chem 6:4893–4901 Seth D, Gorrell MD, McGuinness PH, Leo MA, Lieber CS, McCaughan GW, et al., 2003, SMART amplification maintains representation of relative gene expression: quantitative validation by real time PCR and application to studies of alcoholic liver disease in primates. J Biochem Biophys Methods 55(1):53–66 Hsu F-M, Cheng JC-H, Chang Y-L, Lee J-M, Koong AC, Chuang EY, 2015, Circulating mRNA Profiling in Esophageal Squamous Cell Carcinoma Identifies FAM84B As A Biomarker In Predicting Pathological Response to Neoadjuvant Chemoradiation. Sci Report 5 Wang J, Zhang K-Y, Liu S-M, Sen S, 2014, Tumor-associated circulating microRNAs as biomarkers of cancer. Molecules 19(2): 1912–1938 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 271 EP 279 DI 10.1007/s12253-016-0090-5 PG 9 ER PT J AU Gobbo, DA Fiori, S Ercoli, G Bernardo, DA Parafioriti, A Fabris, S Iurlo, A Neri, A Bosari, S Gianelli, U AF Gobbo, Del Alessandro Fiori, Stefano Ercoli, Giulia Bernardo, Di Andrea Parafioriti, Antonina Fabris, Sonia Iurlo, Alessandra Neri, Antonino Bosari, Silvano Gianelli, Umberto TI Primary Soft Tissue Lymphomas: Description of Seven Cases and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lymphoma; Soft tissue tumors; FISH; C-myc oncogene ID Lymphoma; Soft tissue tumors; FISH; C-myc oncogene AB The present study describes a series of primary soft tissue lymphomas, including immunohistochemical characterization by tissue microarray and cytogenetic profiling. Formalin-fixed, paraffin-embedded tissue samples were collected from patients who underwent soft tissue biopsy. Cases were selected according to the definition of primary soft tissue lymphoma as a lymphoid malignancy arising in soft tissues without evidence of other nodal or extranodal localization for a period of at least 6 months. Our series comprised seven patients with a mean age of 72 years. There were three diffuse large B-cell lymphomas (DLBCLs); one B-cell lymphoma, unclassifiable, with features intermediate between DLBCL and Burkitt lymphoma; one DLBCL derived from follicular lymphoma; one ALK-negative anaplastic large cell lymphoma; and one follicular lymphoma. Immunohistochemical and molecular profiles were consistent with the histological diagnoses. The present study contributes to our knowledge about uncommon presentation of lymphoid neoplasms and confirms previously published clinical-pathological data. We present, for the first time, the complete immunohistochemical profile and molecular cytogenetic studies of these lymphoid neoplasms. A rare case of a primary soft tissue ALK-negative anaplastic large cell lymphoma is described in detail. C1 [Gobbo, Del Alessandro] Ospedale Maggiore Policlinico, Fondazione IRCCS Ca’ Granda, Division of Pathology, via F. Sforza, 35, 20122 Milan, Italy. [Fiori, Stefano] Ospedale Maggiore Policlinico, Fondazione IRCCS Ca’ Granda, Division of Pathology, via F. Sforza, 35, 20122 Milan, Italy. [Ercoli, Giulia] Ospedale Maggiore Policlinico, Fondazione IRCCS Ca’ Granda, Division of Pathology, via F. Sforza, 35, 20122 Milan, Italy. [Bernardo, Di Andrea] Istituto Ortopedico G. Pini, Division of Pathology, p.zza Cardinale A. Ferrari 1, 20122 Milan, Italy. [Parafioriti, Antonina] Istituto Ortopedico G. Pini, Division of Pathology, p.zza Cardinale A. Ferrari 1, 20122 Milan, Italy. [Fabris, Sonia] Ospedale Maggiore Policlinico, Fondazione IRCCS Ca’ Granda, Hematology Unit, via F. Sforza, 35, 20122 Milan, Italy. [Iurlo, Alessandra] Ospedale Maggiore Policlinico, Fondazione IRCCS Ca’ Granda, Hematology Unit, via F. Sforza, 35, 20122 Milan, Italy. [Neri, Antonino] Ospedale Maggiore Policlinico, Fondazione IRCCS Ca’ Granda, Hematology Unit, via F. Sforza, 35, 20122 Milan, Italy. [Bosari, Silvano] Ospedale Maggiore Policlinico, Fondazione IRCCS Ca’ Granda, Division of Pathology, via F. Sforza, 35, 20122 Milan, Italy. [Gianelli, Umberto] Ospedale Maggiore Policlinico, Fondazione IRCCS Ca’ Granda, Division of Pathology, via F. Sforza, 35, 20122 Milan, Italy. RP Gianelli, U (reprint author), Ospedale Maggiore Policlinico, Fondazione IRCCS Ca’ Granda, Division of Pathology, 20122 Milan, Italy. EM umberto.gianelli@unimi.it CR Derenzini E, Casadei B, Pellegrini C, Argnani L, Pileri S, Zinzani PL, 2013, Non-hodgkin lymphomas presenting as soft tissue masses: a single center experience and meta-analysis of the published series. Clin Lymphoma Myeloma Leuk 13:258–265 Kransdorf MJ, 1995, Is it necessary to routinely use gadolinium in theMR imaging evaluation of soft-tissue tumors of the extremities? AJR Am J Roentgenol 165:1545 Yang J, Zhang F, Fang H, et al., 2010, Clinicopathologic features of primary lymphoma in soft tissue. Leuk Lymphoma 51: 2039–2046 Travis WD, Banks PM, Reiman HM, 1987, Primary extranodal soft tissue lymphoma of the extremities. Am J Surg Pathol 11: 359–366 Salamao DR, Nascimento AG, Lloyd RV, et al., 1996, Lymphoma in soft tissue: a clinicopathologic study of 19 cases. Hum Pathol 27: 253–257 O’Neill JK, Devaraj V, Silver DA, et al., 2007, Extranodal lymphomas presenting as soft tissue sarcomas to a sarcoma service over a two-year period. J Plast Reconstr Aesthet Surg 60:646– 654 Damron TA, Le MH, Rooney MT, et al., 1999, Lymphoma presenting as a soft tissue mass. A soft tissue sarcoma simulator. Clin Orthop Relat Res 360:221–230 Hudacko R, Rapkiewicz A, Berman RS, et al., 2011, ALKnegative anaplastic large cell lymphoma mimicking a soft tissue sarcoma. J Cytol 28:230–233 Knowles B, Serpell JW, 2003, Extra-nodal lymphoma presenting as a mimic of soft-tissue sarcoma. ANZ J Surg 73:26–30 Suresh S, Saifuddin A, O’Donnell P, 2008, Lymphoma presenting as a musculoskeletal soft tissue mass:MRI findings in 24 cases. Eur Radiol 18:2628–2634 Chun CW, Jee WH, Park HJ, et al., 2010, MRI features of skeletal muscle lymphoma. AJR Am J Roentgenol 195:1355– 1360 Hans CP,Weisenburger DD, Greiner TC, et al., 2004, Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood 103:275– 282 Lanham GR, Weiss SW, Enzinger FM, 1989, Malignant lymphoma. A study of 75 cases presenting in soft tissue. Am J Surg Pathol 13:1–10 Chun CW, Jee WH, Park HJ, Kim YJ, Park JM, Lee SH, Park SH, 2010, MRI features of skeletal muscle lymphoma. AJR Am J Roentgenol 195:1355–1360 Carroll G, BreidahlW, Robbins P, 2013, Musculoskeletal lymphoma: MRI of bone or soft tissue presentations. J Med Imaging Radiat Oncol 57:663–673 Pant V, Jambhekar NA, Madur B, Shet TM, Agarwal M, Puri A, Gujral S, Banavali M, Arora B, 2007, Anaplastic large cell lymphoma, ALCL, presenting as primary bone and soft tissue sarcoma–a study of 12 cases. Indian J Pathol Microbiol 50:303– 307 Driss M, Abbes I, Mrad K, Sassi S, Oubich F, Barsaoui S, Romdhane KB, 2009, Primary CD30/ALK-1 positive anaplastic large cell lymphoma of the skeletal muscle in a child. Pathologica 101:97–100 Ishii E, Honda K, Nakagawa A, et al., 2000, Primary CD30/ki- 1 positive anaplastic large cell lymphoma of skeletal muscle with der; 17t(1;17)(q11;p11). Cancer Genet Cytogenet 122:116– 120 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 281 EP 286 DI 10.1007/s12253-016-0096-z PG 6 ER PT J AU Fullar, A Firneisz, G Regos, E Dudas, J Szarvas, T Baghy, K Ramadori, G Kovalszky, I AF Fullar, Alexandra Firneisz, Gabor Regos, Eszter Dudas, Jozsef Szarvas, Tibor Baghy, Kornelia Ramadori, Giuliano Kovalszky, Ilona TI Response of Hepatic Stellate Cells to TGFB1 Differs from the Response of Myofibroblasts. Decorin Protects against the Action of Growth Factor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatic fibrosis; HSC; Myofibroblast; Decorin; TGF-β signaling ID Hepatic fibrosis; HSC; Myofibroblast; Decorin; TGF-β signaling AB Regardless to the exact nature of damage, hepatic stellate cells (HSCs) and other non-parenchymal liver cells transform to activated myofibroblasts, synthesizing the accumulating extracellular matrix (ECM) proteins, and transforming growth factor-β1 (TGF-β1) plays a crucial role in this process. Later it was discovered that decorin, member of the small leucin rich proteoglycan family is able to inhibit this action of TGF-β1. The aim of our present study was to clarify whether HSCs and activated myofibroblasts of portal region exert identical or different response to TGF-β1 exposure, and the inhibitory action of decorin against the growth factor is a generalized phenomenon on myofibroblast of different origin? To this end we measured mRNA expression and production of major collagen components (collagen type I, III and IV) of the liver after stimulation and co-stimulation with TGF-β1 and decorin in primary cell cultures of HSCs and myofibroblasts (MFs). Production of matrix proteins, decorin and members of the TGF-β1 signaling pathways were assessed on Western blots. Messenger RNA expression of collagens and TIEG was quantified by real-time RT-PCR. HSCs and MFs responded differently to TGF-β1 exposure. In contrast to HSCs in which TGF-β1 stimulated the synthesis of collagen type I, type III, and type IV, only the increase of collagen type IV was detected in portal MFs. However, in a combined treatment, decorin seemed to interfere with TGF-β1 and its stimulatory effect was abolished. The different mode of TGF-β1 action is mirrored by the different activation of signaling pathways in activated HSCs and portal fibroblasts. In HSCs the activation of pSMAD2 whereas in myofibroblasts the activation of MAPK pathway was detected. The inhibitory effect of decorin was neither related to the Smad-dependent nor to the Smadindependent signaling pathways. C1 [Fullar, Alexandra] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Firneisz, Gabor] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Regos, Eszter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Dudas, Jozsef] Medical University Innsbruck, Department of OtorhinolaryngologyInnsbruck, Austria. [Szarvas, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Baghy, Kornelia] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Ramadori, Giuliano] George August University, Department of Gastroenterology and EndocrinologyGottingen, Germany. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. RP Kovalszky, I (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM koval@korb1.sote.hu CR Blomhoff R, Wake K, 1991, Perisinusoidal stellate cells of the liver: important roles in retinol metabolism and fibrosis. FASEB J: Off Publ Fed Am Soc Exp Biol 5(3):271–277 Ramadori G, 1991, The stellate cell, Ito-cell, fat-storing cell, lipocyte, perisinusoidal cell, of the liver. New insights into pathophysiology of an intriguing cell. Virchows Archiv B, Cell Pathol Incl Mol Pathol 61(3):147–158 Neubauer K, Saile B, Ramadori G, 2001, Liver fibrosis and altered matrix synthesis. Can J Gastroenterol = J Can de Gastroenterol 15(3):187–193 Friedman SL, 1990, Cellular sources of collagen and regulation of collagen production in liver. Semin Liver Dis 10(1):20–29., DOI 10.1055/s-2008-1040454 Bhunchet E,Wake K, 1992, Role of mesenchymal cell populations in porcine serum-induced rat liver fibrosis. Hepatology 16(6): 1452–1473 Bataller R, Brenner DA, 2005, Liver fibrosis. J Clin Invest 115(2): 209–218., DOI 10.1172/JCI24282 Arthur MJ, 2000, Fibrogenesis II. Metalloproteinases and their inhibitors in liver fibrosis. Am J Physiol Gastrointest Liver Physiol 279(2):G245–G249 Nakatsukasa H, Nagy P, Evarts RP, Hsia CC, Marsden E, Thorgeirsson SS, 1990, Cellular distribution of transforming growth factor-beta 1 and procollagen types I, III, and IV transcripts in carbon tetrachloride-induced rat liver fibrosis. J Clin Invest 85(6):1833–1843., DOI 10.1172/JCI114643 Castilla A, Prieto J, Fausto N, 1991, Transforming growth factors beta 1 and alpha in chronic liver disease. Effects of interferon alfa therapy. N Engl J Med 324(14):933–940., DOI 10.1056/NEJM199104043241401 Border WA, Noble NA, 1994, Transforming growth factor beta in tissue fibrosis. N Engl J Med 331(19):1286–1292., DOI 10.1056 /NEJM199411103311907 Saile B, Matthes N, Knittel T, Ramadori G, 1999, Transforming growth factor beta and tumor necrosis factor alpha inhibit both apoptosis and proliferation of activated rat hepatic stellate cells. Hepatology 30(1):196–202., DOI 10.1002/hep.510300144 Saile B, Matthes N, El Armouche H, Neubauer K, Ramadori G, 2001, The bcl, NFkappaB and p53/p21WAF1 systems are involved in spontaneous apoptosis and in the anti-apoptotic effect of TGF-beta or TNF-alpha on activated hepatic stellate cells. Eur J Cell Biol 80(8):554–561 Guyot C, Lepreux S, Combe C, Doudnikoff E, Bioulac-Sage P, Balabaud C, Desmouliere A, 2006, Hepatic fibrosis and cirrhosis: the, myo)fibroblastic cell subpopulations involved. Int J Biochem Cell Biol 38(2):135–151., DOI 10.1016/j.biocel.2005.08.021 Forbes SJ, Russo FP, Rey V, Burra P, RuggeM,Wright NA, Alison MR, 2004, A significant proportion of myofibroblasts are of bone marrow origin in human liver fibrosis. Gastroenterology 126(4):955–963 Russo FP, Alison MR, Bigger BW, Amofah E, Florou A, Amin F, Bou-Gharios G, Jeffery R, Iredale JP, Forbes SJ, 2006, The bone marrow functionally contributes to liver fibrosis. Gastroenterology 130(6):1807–1821., DOI 10.1053/j.gastro.2006.01.036 Eyden B, 2008, The myofibroblast: phenotypic characterization as a prerequisite to understanding its functions in translational medicine. J Cell Mol Med 12(1):22–37., DOI 10.1111/j.1582-4934.2007.00213.x Yamaguchi Y, Mann DM, Ruoslahti E, 1990, Negative regulation of transforming growth factor-beta by the proteoglycan decorin. Nature 346(6281):281–284., DOI 10.1038/346281a0 Border WA, Noble NA, Yamamoto T, Harper JR, Yamaguchi Y, Pierschbacher MD, Ruoslahti E, 1992, Natural inhibitor of transforming growth factor-beta protects against scarring in experimental kidney disease. Nature 360(6402):361–364., DOI 10.1038 /360361a0 Dudas J, Kovalszky I, Gallai M, Nagy JO, Schaff Z, Knittel T, Mehde M, Neubauer K, Szalay F, Ramadori G, 2001, Expression of decorin, transforming growth factor-beta 1, tissue inhibitor metalloproteinase 1 and 2, and type IV collagenases in chronic hepatitis. Am J Clin Pathol 115(5):725–735., DOI 10.1309/J8CD-E9C8- X4NG-GTVG Jarmay K, GallaiM, Karacsony G, Ozsvar Z, Schaff Z, Lonovics J, Kovalszky I, 2000, Decorin and actin expression and distribution in patients with chronic hepatitis C following interferon-alfa-2b treatment. J Hepatol 32(6):993–1002 Takeuchi Y, Kodama Y, Matsumoto T, 1994, Bone matrix decorin binds transforming growth factor-beta and enhances its bioactivity. J Biol Chem 269(51):32634–32638 Bi Y, Stuelten CH, Kilts T,Wadhwa S, Iozzo RV, Robey PG, Chen XD, Young MF, 2005, Extracellular matrix proteoglycans control the fate of bonemarrow stromal cells. J Biol Chem 280(34):30481– 30489., DOI 10.1074/jbc.M500573200 Cui X, Shimizu I, Lu G, Itonaga M, Inoue H, Shono M, Tamaki K, Fukuno H, Ueno H, Ito S, 2003, Inhibitory effect of a soluble transforming growth factor beta type II receptor on the activation of rat hepatic stellate cells in primary culture. J Hepatol 39(5):731–737 Shi YF, Zhang Q, Cheung PY, Shi L, Fong CC, Zhang Y, Tzang CH, Chan BP, FongWF, Chun J, Kung HF, YangM(2006, Effects of rhDecorin on TGF-beta1 induced human hepatic stellate cells LX-2 activation. Biochim Biophys Acta 1760(11):1587–1595., DOI 10.1016/j.bbagen.2006.09.012 Baghy K, Iozzo RV, Kovalszky I, 2012, Decorin-TGFbeta axis in hepatic fibrosis and cirrhosis. J Histochem Cytochem 60(4):262– 268., DOI 10.1369/0022155412438104 Baghy K, Dezso K, Laszlo V, Fullar A, Peterfia B, Paku S, Nagy P, Schaff Z, Iozzo RV, Kovalszky I, 2011, Ablation of the decorin gene enhances experimental hepatic fibrosis and impairs hepatic healing in mice. Lab Investig 91(3):439–451., DOI 10.1038 /labinvest.2010.172 de Leeuw AM,McCarthy SP, Geerts A, Knook DL, 1984, Purified rat liver fat-storing cells in culture divide and contain collagen. Hepatology 4(3):392–403 Knittel T, Kobold D, Saile B, Grundmann A, Neubauer K, Piscaglia F, Ramadori G, 1999, Rat liver myofibroblasts and hepatic stellate cells: different cell populations of the fibroblast lineage with fibrogenic potential. Gastroenterology 117(5):1205–1221 Bradford MM, 1976, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72:248–254 Sahin MB, Schwartz RE, Buckley SM, Heremans Y, Chase L, Hu WS, Verfaillie CM, 2008, Isolation and characterization of a novel population of progenitor cells from unmanipulated rat liver. Liver Transplant: Off Publ Am Assoc Study Liver Dis Int Liver Transplant Soc 14(3):333–345., DOI 10.1002/lt.21380 Sanderson N, Factor V, Nagy P, Kopp J, Kondaiah P, Wakefield L, Roberts AB, Sporn MB, Thorgeirsson SS, 1995, Hepatic expression of mature transforming growth factor beta 1 in transgenic mice results in multiple tissue lesions. Proc Natl Acad Sci U S A 92(7):2572–2576 Dudas J, Mansuroglu T, Batusic D, Saile B, Ramadori G, 2007, Thy-1 is an in vivo and in vitro marker of liver myofibroblasts. Cell Tissue Res 329(3):503–514., DOI 10.1007/s00441-007-0437-z Dezso K, Jelnes P, Laszlo V, Baghy K, Bodor C, Paku S, Tygstrup N, Bisgaard HC, Nagy P, 2007, Thy-1 is expressed in hepatic myofibroblasts and not oval cells in stem cell-mediated liver regeneration. Am J Pathol 171(5):1529–1537., DOI 10.2353 /ajpath.2007.070273 Weiss TS, Lichtenauer M, Kirchner S, Stock P, Aurich H, Christ B, Brockhoff G, Kunz-Schughart LA, Jauch KW, Schlitt HJ, Thasler WE, 2008, Hepatic progenitor cells from adult human livers for cell transplantation. Gut 57(8):1129–1138., DOI 10.1136/gut.2007.143321 Ueberham E, Low R, Ueberham U, Schonig K, Bujard H, Gebhardt R, 2003, Conditional tetracycline-regulated expression of TGF-beta1 in liver of transgenic mice leads to reversible intermediary fibrosis. Hepatology 37(5):1067–1078., DOI 10.1053/jhep.2003.50196 Schonherr E, Sunderkotter C, Iozzo RV, Schaefer L, 2005)Decorin, a novel player in the insulin-like growth factor system. J Biol Chem 280(16):15767–15772., DOI 10.1074/jbc.M500451200 Goldoni S, Iozzo RV, 2008, Tumor microenvironment: modulation by decorin and related molecules harboring leucine-rich tandem motifs. Int J Cancer Journal Int du Cancer 123(11):2473–2479., DOI 10.1002/ijc.23930 Goldoni S, Humphries A, Nystrom A, Sattar S, Owens RT, McQuillan DJ, Ireton K, Iozzo RV, 2009, Decorin is a novel antagonistic ligand of the met receptor. J Cell Biol 185(4):743–754., DOI 10.1083/jcb.200901129 Baghy K, Horvath Z, Regos E, Kiss K, Schaff Z, Iozzo RV, Kovalszky I, 2013, Decorin interferes with platelet-derived growth factor receptor signaling in experimental hepatocarcinogenesis. FEBS J 280(10):2150–2164., DOI 10.1111/febs.12215 Yu MC, Chen CH, Liang X, Wang L, Gandhi CR, Fung JJ, Lu L, Qian S, 2004, Inhibition of T-cell responses by hepatic stellate cells via B7-H1-mediated T-cell apoptosis in mice. Hepatology 40(6): 1312–1321., DOI 10.1002/hep.20488 Chen CH, Kuo LM, Chang Y,WuW, Goldbach C, Ross MA, Stolz DB, Chen L, Fung JJ, Lu L, Qian S, 2006, In vivo immune modulatory activity of hepatic stellate cells in mice. Hepatology 44(5): 1171–1181., DOI 10.1002/hep.21379 Merline R, Moreth K, Beckmann J, Nastase MV, Zeng-Brouwers J, Tralhao JG, Lemarchand P, Pfeilschifter J, Schaefer RM, Iozzo RV, Schaefer L, 2011, Signaling by the matrix proteoglycan decorin controls inflammation and cancer through PDCD4 and MicroRNA-21. Sci Signal 4(199):ra75., DOI 10.1126/scisignal.2001868 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 287 EP 294 DI 10.1007/s12253-016-0095-0 PG 8 ER PT J AU Varga, N Mozes, J Keegan, H White, Ch Kelly, Ly Pilkington, L Benczik, M Schaff, Zs Sobel, G Koiss, R Babarczi, E Nyiri, M Kovacs, L Sebe, A Kaltenecker, B Geresi, A Kocsis, A O’Leary, J Martin, MC Jeney, Cs AF Varga, Norbert Mozes, Johanna Keegan, Helen White, Christine Kelly, Lynne Pilkington, Loretto Benczik, Marta Schaff, Zsuzsa Sobel, Gabor Koiss, Robert Babarczi, Edit Nyiri, Miklos Kovacs, Laura Sebe, Attila Kaltenecker, Borbala Geresi, Adrienn Kocsis, Adrienn O’Leary, John Martin, M Cara Jeney, Csaba TI The Value of a Novel Panel of Cervical Cancer Biomarkers for Triage of HPV Positive Patients and for Detecting Disease Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human papillomavirus; Cervical neoplasia; Case-control study; Cellular biomarker; biomarker; cervical cancer; carcinogenesis ID Human papillomavirus; Cervical neoplasia; Case-control study; Cellular biomarker; biomarker; cervical cancer; carcinogenesis AB In the era of primary vaccination against HPV and at the beginning of the low prevalence of cervical lesions, introduction of screening methods that can distinguish between lowand high-grade lesions is necessary in order to maintain the positive predictive value of screening. This case-control study included 562 women who attended cervical screening or were referred for colposcopy and 140 disease free controls, confirmed by histology and/or cytology. The cases were stratified by age. Using routine exfoliated liquid based cytological samples RT-PCR measurements of biomarker genes, high-risk HPV testing and liquid based cytology were performed and used to evaluate different testing protocols including sets of genes/tests with different test cut-offs for the diagnostic panels. Three new panels of cellular biomarkers for improved triage of hrHPV positive women (diagnostic panel) and for prognostic assessment of CIN lesions were proposed. The diagnostic panel (PIK3AP1, TP63 and DSG3) has the potential to distinguish cytologically normal hrHPV+ women from hrHPV+ women with CIN2+. The prognostic gene panels (KRT78, MUC5AC, BPIFB1 and CXCL13, TP63, DSG3) have the ability to differentiate hrHPV+ CIN1 and carcinoma cases. The diagnostic triage panel showed good likelihood ratios for all age groups. The panel showed age-unrelated performance and even better diagnostic value under age 30, a unique feature among the established cervical triage tests. The prognostic gene-panels demonstrated good discriminatory power and oncogenic, antioncogenic grouping of genes. The study highlights the potential for the gene expression panels to be used for diagnostic triage and lesion prognostics in cervical cancer screening. C1 [Varga, Norbert] CellCall Ltd, Roppentyu utca 48, 1134 Budapest, Hungary. [Mozes, Johanna] CellCall Ltd, Roppentyu utca 48, 1134 Budapest, Hungary. [Keegan, Helen] Trinity College Dublin, School of Medicine, Department of HistopathologyDublin, Ireland. [White, Christine] Trinity College Dublin, School of Medicine, Department of HistopathologyDublin, Ireland. [Kelly, Lynne] Trinity College Dublin, School of Medicine, Department of HistopathologyDublin, Ireland. [Pilkington, Loretto] Trinity College Dublin, School of Medicine, Department of HistopathologyDublin, Ireland. [Benczik, Marta] CellCall Ltd, Roppentyu utca 48, 1134 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, 1091 Budapest, Hungary. [Sobel, Gabor] Semmelweis University, 2nd Department of Obstetrics and Gynecology, Ulloi ut 78/a, 1082 Budapest, Hungary. [Koiss, Robert] Szent Istvan Korhaz, Nogyogyaszati Onkologiai Osztaly, Nagyvarad ter 1, 1087 Budapest, Hungary. [Babarczi, Edit] Szent Istvan Korhaz, Nogyogyaszati Onkologiai Osztaly, Nagyvarad ter 1, 1087 Budapest, Hungary. [Nyiri, Miklos] CellCall Ltd, Roppentyu utca 48, 1134 Budapest, Hungary. [Kovacs, Laura] CellCall Ltd, Roppentyu utca 48, 1134 Budapest, Hungary. [Sebe, Attila] Semmelweis University, Institute of Morphology and Physiology, Nagyvarad ter 4, 1089 Budapest, Hungary. [Kaltenecker, Borbala] GenoID Molecular Diagnostic Laboratory, Szanatorium u. 19, 1121 Budapest, Hungary. [Geresi, Adrienn] CellCall Ltd, Roppentyu utca 48, 1134 Budapest, Hungary. [Kocsis, Adrienn] CellCall Ltd, Roppentyu utca 48, 1134 Budapest, Hungary. [O’Leary, John] Trinity College Dublin, School of Medicine, Department of HistopathologyDublin, Ireland. [Martin, M Cara] Trinity College Dublin, School of Medicine, Department of HistopathologyDublin, Ireland. [Jeney, Csaba] Semmelweis University, Department of Medical Microbiology, Nagyvarad ter 4, 1089 Budapest, Hungary. RP Jeney, Cs (reprint author), Semmelweis University, Department of Medical Microbiology, 1089 Budapest, Hungary. EM jencsa@med.semmelweis-univ.hu CR Forouzanfar MH, Foreman KJ, Delossantos AM, et al., 2011, Breast and cervical cancer in 187 countries between 1980 and 2010: a systematic analysis. Lancet 378:1461–1484., DOI 10.1016/S0140-6736(11)61351-2 Vaccarella S, Lortet-Tieulent J, Plummer M, et al., 2013, Worldwide trends in cervical cancer incidence: impact of screening against changes in disease risk factors. Eur J Cancer 49:3262–3273., DOI 10.1016/j.ejca.2013.04.024 Elfstrom KM, Herweijer E, Sundstrom K, Arnheim-Dahlstrom L, 2014, Current cervical cancer prevention strategies including cervical screening and prophylactic human papillomavirus vaccination: a review. Curr Opin Oncol 26:120–129., DOI 10.1097/CCO.0000000000000034 Martin CM, O’Leary JJ, 2011, Histology of cervical intraepithelial neoplasia and the role of biomarkers. Best Pract Res Clin Obstet Gynaecol 25:605–615., DOI 10.1016/j.bpobgyn.2011.04.005 Horn L-C, Reichert A, Oster A, et al., 2008, Immunostaining for p16INK4a used as a conjunctive tool improves interobserver agreement of the histologic diagnosis of cervical intraepithelial neoplasia. Am J Surg Pathol 32:502–512., DOI 10.1097 /PAS.0b013e31815ac420 Belinsky SA, Nikula KJ, Palmisano WA, et al., 1998, Aberrant methylation of p16(INK4a, is an early event in lung cancer and a potential biomarker for early diagnosis. Proc Natl Acad Sci U S A 95:11891–11896 Schutte M, Hruban RH, Geradts J, et al., 1997, Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res 57:3126–3130 Henken FE, Wilting SM, Overmeer RM, et al., 2007, Sequential gene promoter methylation during HPV-induced cervical carcinogenesis. Br J Cancer 97:1457–1464., DOI 10.1038/sj.bjc.6604055 Louvanto K, Franco EL, Ramanakumar AV, et al., 2015, Methylation of viral and host genes and severity of cervical lesions associated with human papillomavirus type 16. Int J Cancer 136: E638–E645., DOI 10.1002/ijc.29196 Arbyn M, Ronco G, Cuzick J, et al., 2009, How to evaluate emerging technologies in cervical cancer screening? Int J Cancer 125: 2489–2496., DOI 10.1002/ijc.24774 Jeney C, Takacs T, Sebe A, Schaff Z, 2007, Detection and typing of 46 genital human papillomaviruses by the L1F/L1R primer system based multiplex PCR and hybridization. J Virol Methods 140:32– 42., DOI 10.1016/j.jviromet.2006.10.013 Holmes TG, Donkin A, Witten IH, 1994, Weka: A machine learning workbench. Proc Second Aust. New Zeal. Conf. Intell. Inf, Syst 357–361., DOI 10.1109/ANZIIS.1994.396988 Wentzensen N, Schwartz L, Zuna RE, et al., 2012, Performance of p16/Ki-67 immunostaining to detect cervical cancer precursors in a colposcopy referral population. Clin Cancer Res 18:4154–4162., DOI 10.1158/1078-0432.CCR-12-0270 Bergholz J, Xiao Z-X, 2012, Role of p63 in Development, Tumorigenesis and Cancer Progression. Cancer Microenviron 5: 311–322., DOI 10.1007/s12307-012-0116-9 Gius D, FunkMC, Chuang EY, et al., 2007, Profiling microdissected epithelium and stroma to model genomic signatures for cervical carcinogenesis accommodating for covariates. Cancer Res 67: 7113–7123., DOI 10.1158/0008-5472.CAN-07-0260 Dennis G, Sherman BT, Hosack DA, et al., 2003, DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol 4:P3 Patel V, Martin D, Malhotra R, et al., 2013, DSG3 as a biomarker for the ultrasensitive detection of occult lymph node metastasis in oral cancer using nanostructured immunoarrays. Oral Oncol 49:93– 101., DOI 10.1016/j.oraloncology.2012.08.001 Williamson L, Raess NA, Caldelari R, et al., 2006, Pemphigus vulgaris identifies plakoglobin as key suppressor of c-Myc in the skin. EMBO J 25:3298–3309., DOI 10.1038/sj.emboj.7601224 de Bruin A, Caldelari R, Williamson L, et al., 2007, Plakoglobindependent disruption of the desmosomal plaque in pemphigus vulgaris. Exp Dermatol 16:468–475., DOI 10.1111/j.1600- 0625.2007.00557.x Chen Y, Mistry DS, Sen GL, 2014, Highly rapid and efficient conversion of human fibroblasts to keratinocyte-like cells. J Invest Dermatol 134:335–344., DOI 10.1038/jid.2013.327 Yugawa T, Narisawa-Saito M, Yoshimatsu Y, et al., 2010, DeltaNp63alpha repression of the Notch1 gene supports the proliferative capacity of normal human keratinocytes and cervical cancer cells. Cancer Res 70:4034–4044., DOI 10.1158/0008-5472.CAN- 09-4063 Teissier S, Ben Khalifa Y, Mori M, et al., 2007, A new E6/P63 pathway, together with a strong E7/E2F mitotic pathway,modulates the transcriptome in cervical cancer cells. J Virol 81:9368–9376., DOI 10.1128/JVI.00427-07 Lin Z, Liu M, Li Z, et al., 2006, DeltaNp63 protein expression in uterine cervical and endometrial cancers. J Cancer Res Clin Oncol 132:811–816., DOI 10.1007/s00432-006-0130-8 Pimenta EM, Barnes BJ, 2014, Role of Tertiary Lymphoid Structures, TLS, in Anti-Tumor Immunity: Potential Tumor- Induced Cytokines/Chemokines that Regulate TLS Formation in Epithelial-Derived Cancers. Cancers, Basel, 6:969–997., DOI 10.3390/cancers6020969 Sambandam Y, Sundaram K, Liu A, et al., 2012, CXCL13 activation of c-Myc induces RANK ligand expression in stromal/preosteoblast cells in the oral squamous cell carcinoma tumor–bone microenvironment. Oncogene., DOI 10.1038 /onc.2012.24 Airoldi I, Cocco C, Morandi F, et al., 2008, CXCR5 may be involved in the attraction of human metastatic neuroblastoma cells to the bone marrow. Cancer Immunol Immunother 57:541–548., DOI 10.1007/s00262-007-0392-2 Singh S, Singh R, Sharma PK, et al., 2009, Serum CXCL13 positively correlates with prostatic disease, prostate-specific antigen and mediates prostate cancer cell invasion, integrin clustering and cell adhesion. Cancer Lett 283:29–35., DOI 10.1016/j.canlet.2009.03.022 Feng Q, Wei H, Morihara J, et al., 2012, Th2 type inflammation promotes the gradual progression of HPV-infected cervical cells to cervical carcinoma. Gynecol Oncol 127:412–419., DOI 10.1016/j. ygyno.2012.07.098 Guerrero-Preston R, Michailidi C, Marchionni L, et al., 2014, Key tumor suppressor genes inactivated by "greater promoter" methylation and somatic mutations in head and neck cancer. Epigenetics 9: 1031–1046., DOI 10.4161/epi.29025 Politopoulos I, Gibson J, Tapper W, et al., 2011, Composite likelihood-based meta-analysis of breast cancer association studies. J Hum Genet 56:377–382., DOI 10.1038/jhg.2011.23 Koutros S, Schumacher FR, Hayes RB, et al., 2010, Pooled analysis of phosphatidylinositol 3-kinase pathway variants and risk of prostate cancer. Cancer Res 70:2389–2396., DOI 10.1158/0008- 5472.CAN-09-3575 Oshima G, Wennerberg J, Yamatodani T, et al., 2012, Autocrine epidermal growth factor receptor ligand production and cetuximab response in head and neck squamous cell carcinoma cell lines. J Cancer Res Clin Oncol 138:491–499., DOI 10.1007 /s00432-011-1127-5 Hobbs RP, Lessard JC, Coulombe PA, 2012, Keratin intermediate filament proteins - novel regulators of inflammation and immunity in skin. J Cell Sci 125:5257–5258., DOI 10.1242/jcs.122929 FessingMY,Mardaryev AN, GdulaMR, et al., 2011, p63 regulates Satb1 to control tissue-specific chromatin remodeling during development of the epidermis. J Cell Biol 194:825–839., DOI 10.1083/jcb.201101148 Leung KP, D’Arpa P, Seth AK, et al., 2014, Dermal wound transcriptomic responses to Infection with Pseudomonas aeruginosa versus Klebsiella pneumoniae in a rabbit ear wound model. BMC Clin Pathol 14:20., DOI 10.1186/1472-6890-14-20 Chakraborty S, Bonthu N, Swanson BJ, Batra SK, 2011, Role of mucins in the skin during benign and malignant conditions. Cancer Lett 301:127–141., DOI 10.1016/j.canlet.2010.11.004 Yang Y, Liao Q, Wei F, et al., 2013, LPLUNC1 inhibits nasopharyngeal carcinoma cell growth via down-regulation of the MAP kinase and cyclin D1/E2F pathways. PLoS One 8:e62869., DOI 10.1371/journal.pone.0062869 Nevins JR, 1992, E2F: a link between the Rb tumor suppressor protein and viral oncoproteins. Science 258:424–429 Lambert APF, Anschau F, Schmitt VM, 2006, p16INK4A expression in cervical premalignant and malignant lesions. Exp Mol Pathol 80:192–196., DOI 10.1016/j.yexmp.2005.08.005 Hopman EH, Kenemans P, Helmerhorst TJ, 1998, Positive predictive rate of colposcopic examination of the cervix uteri: an overview of literature. Obstet Gynecol Surv 53:97–106., DOI 10.1097 /00006254-199802000-00021 (2012, ACOG Practice Bulletin Number 131: Screening for cervical cancer. Obstet Gynecol 120:1222–38., DOI 10.1097/AOG.0b013 e318277c92a NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 295 EP 305 DI 10.1007/s12253-016-0094-1 PG 11 ER PT J AU He, J Tan, W Tang, X Ma, J AF He, Jinfeng Tan, Wei Tang, Xuelian Ma, Jingping TI Variations in EGFR ctDNA Correlates to the Clinical Efficacy of Afatinib in Non Small Cell Lung Cancer with Acquired Resistance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non small cell lung cancer (NSCLC); EGFR mutations; Acquired resistance; Afatinib; ctDNA ID Non small cell lung cancer (NSCLC); EGFR mutations; Acquired resistance; Afatinib; ctDNA AB Monitoring of non small cell lung cancer (NSCLC) patients on afatinib after acquired resistance to 1st generation tyrosine kinase inhibitors is important. Circulating tumor DNA (ctDNA) offers an attractive means other than conventional tissue biopsy to characterize real time dynamic changes of the disease. In our study, we aim to ascertain the clinical value for ctDNA monitoring of NSCLC patientswith acquired resistance for afatinib treatment. 200 patients positive for the activating epithermal growth factor receptor (EGFR) mutations were recruited for the study. Baseline molecular profiling for L858R, Exon 19 deletion and T790M were performed. Thereafter, serial blood samples were taken and patients were assessed by overall survival (OS) to determine the usefulness of ctDNA monitoring. At baseline, matched tumor biopsy and ctDNA analysis had a concordance agreement of 93.5% for L858R and exon 19 deletion. We also determined that a large proportion of patients had the drug resistance mutation T790Mprior to starting afatinib and these patients were linked to a worse survival outcome. For patients that registered a drop in ctDNA levels after afatinib was administered, we observed that their survival outcome was more favorable (hazard ratio 1.56, (95% CI 1.04 to 2.43). ctDNA levels were mostly elevated after the 3rd sampling cycle. Our results suggest that ctDNA can be used to predict the clinical benefits of afatinib treatment. Pre and post blood sampling aids to identify patient groups that may benefit most from the treatment and ctDNA is relatively sensitive to address the dynamic changes of the disease at the molecular level. C1 [He, Jinfeng] Yangtze University, The Second Clinical Medical College, Jingzhou Central Hospital, Department of Respiratory Medicine, Renmin Road 1, 434020 Jingzhou, China. [Tan, Wei] Yangtze University, The Second Clinical Medical College, Jingzhou Central Hospital, Haemodialysis CentreJingzhou, China. [Tang, Xuelian] Yangtze University, The Second Clinical Medical College, Jingzhou Central Hospital, Department of Respiratory Medicine, Renmin Road 1, 434020 Jingzhou, China. [Ma, Jingping] Yangtze University, The Second Clinical Medical College, Jingzhou Central Hospital, Department of Respiratory Medicine, Renmin Road 1, 434020 Jingzhou, China. RP Ma, J (reprint author), Yangtze University, The Second Clinical Medical College, Jingzhou Central Hospital, Department of Respiratory Medicine, 434020 Jingzhou, China. EM mjpjzhospital@hotmail.com CR Gazdar AF, 2009, Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene 28(Suppl 1):S24–S31., DOI 10.1038/onc.2009.198 Shigematsu H, Gazdar AF, 2006, Somatic mutations of epidermal growth factor receptor signaling pathway in lung cancers. Int J Cancer 118(2):257–262., DOI 10.1002/ijc.21496 Shi Y, Au JS, Thongprasert S, Srinivasan S, Tsai CM, Khoa MT, Heeroma K, Itoh Y, Cornelio G, Yang PC, 2014, A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology, PIONEER). Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer 9, 2):154-162., DOI 10.1097/JTO.0000000000000033 Kris MG, Natale RB, Herbst RS, Lynch TJ Jr, Prager D, Belani CP, Schiller JH, Kelly K, Spiridonidis H, Sandler A, Albain KS, Cella D, Wolf MK, Averbuch SD, Ochs JJ, Kay AC, 2003, Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial. JAMA 290(16):2149–2158., DOI 10.1001 /jama.290.16.2149 Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, Thongprasert S, Campos D, Maoleekoonpiroj S, Smylie M, Martins R, van Kooten M, Dediu M, Findlay B, Tu D, Johnston D, Bezjak A, Clark G, Santabarbara P, Seymour L, National Cancer Institute of Canada Clinical Trials G, 2005, Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 353(2):123–132., DOI 10.1056/NEJMoa050753 Barker AJ, Gibson KH, GrundyW, Godfrey AA, Barlow JJ, Healy MP, Woodburn JR, Ashton SE, Curry BJ, Scarlett L, Henthorn L, Richards L, 2001, Studies leading to the identification of ZD1839, IRESSA): an orally active, selective epidermal growth factor receptor tyrosine kinase inhibitor targeted to the treatment of cancer. Bioorg Med Chem Lett 11(14):1911–1914 Wakeling AE, Guy SP,Woodburn JR, Ashton SE, Curry BJ, Barker AJ, Gibson KH, 2002, ZD1839, Iressa): an orally active inhibitor of epidermal growth factor signaling with potential for cancer therapy. Cancer Res 62(20):5749–5754 Fukuoka M, Yano S, Giaccone G, Tamura T, Nakagawa K, Douillard JY, Nishiwaki Y, Vansteenkiste J, Kudoh S, Rischin D, Eek R, Horai T, Noda K, Takata I, Smit E, Averbuch S, Macleod A, Feyereislova A, Dong RP, Baselga J, 2003, Multiinstitutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer, The IDEAL 1 Trial, [corrected]. J Clin Oncol Off J Am Soc Clin Oncol 21(12):2237–2246., DOI 10.1200/JCO.2003.10.038 Perez-Soler R, Chachoua A, Hammond LA, Rowinsky EK, Huberman M, Karp D, Rigas J, Clark GM, Santabarbara P, Bonomi P, 2004, Determinants of tumor response and survival with erlotinib in patients with non–small-cell lung cancer. J Clin Oncol Off J Am Soc Clin Oncol 22, 16):3238-3247., DOI 10.1200 /JCO.2004.11.057 Kosaka T, Yatabe Y, Endoh H, Yoshida K, Hida T, Tsuboi M, Tada H, Kuwano H, Mitsudomi T, 2006, Analysis of Epidermal Growth Factor Receptor Gene Mutation in Patients with Non–Small Cell Lung Cancer and Acquired Resistance to Gefitinib. Clin Cancer Res 12(19):5764–5769 Balak MN, Gong Y, Riely GJ, Somwar R, Li AR, Zakowski MF, Chiang A, Yang G, Ouerfelli O, Kris MG, Ladanyi M, Miller VA, Pao W, 2006, Novel D761Y and Common Secondary T790M Mutations in Epidermal Growth Factor Receptor–Mutant Lung Adenocarcinomas with Acquired Resistance to Kinase Inhibitors. Clin Cancer Res 12(21):6494–6501 Li D, Ambrogio L, Shimamura T, Kubo S, Takahashi M, Chirieac LR, Padera RF, Shapiro GI, Baum A, Himmelsbach F, Rettig WJ, Meyerson M, Solca F, Greulich H,Wong, K K, 2008, BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models. Oncogene 27, 34):4702-4711., DOI http://www. nature.com/onc/journal/v27/n34/suppinfo/onc2008109s1.html Solca F, Dahl G, Zoephel A, Bader G, Sanderson M, Klein C, Kraemer O, Himmelsbach F, Haaksma E, Adolf GR, 2012, Target Binding Properties and Cellular Activity of Afatinib, BIBW 2992), an Irreversible ErbB Family Blocker. J Pharmacol Exp Ther 343(2):342–350 Kwak EL, Sordella R, Bell DW, Godin-Heymann N, Okimoto RA, Brannigan BW, Harris PL, Driscoll DR, Fidias P, Lynch TJ, Rabindran SK, McGinnis JP, Wissner A, Sharma SV, Isselbacher KJ, Settleman J, Haber DA, 2005, Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proc Natl Acad Sci U S A 102(21):7665–7670 Katakami N,Atagi S, Goto K,Hida T, Horai T, Inoue A, IchinoseY, Koboyashi K, Takeda K, Kiura K, Nishio K, Seki Y, Ebisawa R, Shahidi M, Yamamoto N, 2013, LUX-Lung 4: a phase ii trial of afatinib in patients with advanced non–small-cell lung cancer who progressed during prior treatment with erlotinib, gefitinib, or both. J Clin Oncol 31(27):3335–3341 Taniguchi K, Uchida J, Nishino K, Kumagai T, Okuyama T, Okami J, Higashiyama M, Kodama K, Imamura F, Kato K, 2011, Quantitative Detection of EGFR Mutations in Circulating Tumor DNA Derived from Lung Adenocarcinomas. Clin Cancer Res 17(24):7808–7815., DOI 10.1158/1078-0432.ccr-11-1712 Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, Thornton K, Agrawal N, Sokoll L, Szabo SA, Kinzler KW, Vogelstein B, Diaz Jr LA, 2008, Circulating mutant DNA to assess tumor dynamics. Nature Medicine 14, 9):985-990. http://www. nature.com/nm/journal/v14/n9/suppinfo/nm.1789_S1.html Jackman D, Pao W, Riely GJ, Engelman JA, Kris MG, Janne PA, Lynch T, Johnson BE, Miller VA, 2010, Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol Off J Am Soc Clin Oncol 28(2):357–360., DOI 10.1200/JCO.2009.24.7049 Oxnard GR, Paweletz CP, Kuang Y, Mach SL, O'Connell A, Messineo MM, Luke JJ, Butaney M, Kirschmeier P, Jackman DM, Janne PA, 2014, Noninvasive Detection of Response and Resistance in EGFR-Mutant Lung Cancer Using Quantitative Next-Generation Genotyping of Cell-Free Plasma DNA. Clin Cancer Res 20(6):1698–1705 Watanabe M, Kawaguchi T, S-I I, Ando M, Tamiya A, Kubo A, Saka H, Takeo S, Adachi H, Tagawa T, Kakegawa S, Yamashita M, Kataoka K, Ichinose Y, Takeuchi Y, Sakamoto K, Matsumura A, Koh Y, 2015, Ultra-Sensitive Detection of the Pretreatment EGFR T790M Mutation in Non–Small Cell Lung Cancer Patients with an EGFR-Activating Mutation Using Droplet Digital PCR. Clin Cancer Res 21(15):3552–3560 Kidess E, Heirich K,Wiggin M, Vysotskaia V, Visser BC, Marziali A,Wiedenmann B, Norton JA, Lee M, Jeffrey SS, 2015, Mutation profiling of tumor DNA from plasma and tumor tissue of colorectal cancer patients with a novel, high-sensitivity multiplexed mutation detection platform. Oncotarget 6(4):2549 Szpechcinski A, Chorostowska-Wynimko J, Struniawski R, Kupis W, Rudzinski P, Langfort R, Puscinska E, Bielen P, Sliwinski P, Orlowski T, 2015, Cell-free DNA levels in plasma of patients with non-small-cell lung cancer and inflammatory lung disease. Br J Cancer 113(3):476–483., DOI 10.1038/bjc.2015.225 Zheng D, Ye X, Zhang MZ, Sun Y, Wang JY, Ni J, Zhang HP, Zhang L, Luo J, Zhang J, Tang L, Su B, Chen G, Zhu G, Gu Y, Xu JF, 2016, Plasma EGFR T790M ctDNA status is associated with clinical outcome in advanced NSCLC patients with acquired EGFR-TKI resistance. Sci Report 6:20913., DOI 10.1038/srep20913 Bai H, Mao L, Wang HS, Zhao J, Yang L, An TT, Wang X, Duan CJ,Wu NM, Guo ZQ, Liu YX, Liu HN,Wang YY,Wang J, 2009, Epidermal growth factor receptor mutations in plasma dna samples predict tumor response in chinese patients with stages iiib to iv non– small-cell lung cancer. J Clin Oncol 27(16):2653–2659 Kimura H, Suminoe M, Kasahara K, Sone T, Araya T, Tamori S, Koizumi F, Nishio K, Miyamoto K, Fujimura M, Nakao S, 2007, Evaluation of epidermal growth factor receptor mutation status in serum DNA as a predictor of response to gefitinib, IRESSA). Br J Cancer 97(6):778–784., DOI 10.1038/sj.bjc.6603949 Thress KS, Brant R, Carr TH, Dearden S, Jenkins S, Brown H, Hammett T, Cantarini M, Barrett JC, 2015, EGFR mutation detection in ctDNA from NSCLC patient plasma: A cross-platform comparison of leading technologies to support the clinical development of AZD9291. Lung Cancer 90(3): 509–515., DOI 10.1016/j.lungcan.2015.10.004 Yong E, 2014, Cancer biomarkers: Written in blood. Nature 511(7511):524–526., DOI 10.1038/511524a De Greve J, Moran T, Graas M-P, Galdermans D, Vuylsteke P, Canon J-L, Schallier D, Decoster L, Teugels E, Massey D, Chand VK, Vansteenkiste J, 2015, Phase II study of afatinib, an irreversible ErbB family blocker, in demographically and genotypically defined lung adenocarcinoma. Lung Cancer 88(1):63–69., DOI 10.1016/j.lungcan.2015.01.013 Bar J, Botser D, Navon R, Peled N, Biran H, Gal-Yam EN, Ben- Arieh S, Raskin S, Onn A A Single-arm phase II study of afatinib treatment for advanced EGFR-mutant non-small cell lung cancer, mNSCLC, patients, pts, with EGFR-tyrosine kinase inhibitor, TKI, resistance. In: ASCO Annual Meeting Proceedings, 2015. vol 15_suppl. p e19031 Lee JY, Sun JM, Lim SH, Kim HS, Yoo KH, Jung KS, Song HN, Ku BM, Koh J, Bae YH, Lee SH, Ahn JS, Park K, Ahn MJ, 2016, A phase Ib/II study of afatinib in combination with nimotuzumab in non-small cell lung cancer patients with acquired resistance to gefitinib or erlotinib. Clin Cancer Res 22(9):2139–2145., DOI 10.1158/1078-0432. CCR-15-1653 Zhang S, Zheng X, Huang H, Wu K, Wang B, Chen X, Ma S, 2015, Afatinib increases sensitivity to radiation in nonsmall cell lung cancer cells with acquired EGFR T790M mutation. Oncotarget 6(8):5832 Maheswaran S, Sequist LV, Nagrath S, Ulkus L, Brannigan B, Collura CV, Inserra E, Diederichs S, Iafrate AJ, Bell DW, Digumarthy S, Muzikansky A, Irimia D, Settleman J, Tompkins RG, Lynch TJ, Toner M, Haber DA, 2008, Detection of mutations in EGFR in circulating lung-cancer cells. N Engl J Med 359(4): 366–377., DOI 10.1056/NEJMoa0800668 Kim ES, Herbst RS,Wistuba II, Lee JJ, Blumenschein GR Jr, Tsao A, Stewart DJ, HicksME, Erasmus J Jr, Gupta S, Alden CM, Liu S, Tang X, Khuri FR, Tran HT, Johnson BE, Heymach JV, Mao L, Fossella F, Kies MS, Papadimitrakopoulou V, Davis SE, Lippman SM, HongWK, 2011, The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov 1(1):44–53., DOI 10.1158/2159-8274. CD-10-0010 Jekunen AP, 2015, Role of rebiopsy in relapsed non-small cell lung cancer for directing oncology treatments. J Oncol 2015:11., DOI 10.1155/2015/809835 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 307 EP 315 DI 10.1007/s12253-016-0097-y PG 9 ER PT J AU Lutkowska, A Roszak, A Jagodzinski, PP AF Lutkowska, Anna Roszak, Andrzej Jagodzinski, P Pawel TI 17β-hydroxysteroid dehydrogenase type Gene 1937 A > G Polymorphism as a Risk Factor for Cervical Cancer Progression in the Polish Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical carcinoma; HSD17B1; Polymorphisms ID Cervical carcinoma; HSD17B1; Polymorphisms AB The role of 17β-estradiol (E2) in the development of cervical tumor (CT) has been demonstrated. 17β Hydroxysteroid dehydrogenase type 1 (HSD17B1) converts estrone (E1) into E2.We aimed to study the distribution of the HSD17B1937 A > G (rs605059) single nucleotide polymorphism (SNP) in women (n = 383) with CT and controls (n = 401) from the Polish population. The p-trend value evaluated for HSD17B1 rs605059 was 0.0233 for all patients. The A/A vs G/G genotype significantly contributed to all patients with CT, and the Odds Ratio (OR) was 1.570 (95 % CI = 1.053–2.343; p = 0.0266). Stratification of the patients based on tumor stage and histological grade indicated the contribution of HSD17B1937 A > G to stages III and IV. The p-value was 0.0010. The OR for the A/Avs G/G genotype was 2.992 (95 % CI = 1.627–5.502, p = 0.0003), the OR for the A/G vs G/G genotype was 2.545 (95 % CI = 1.410–4.593, p = 0.0015) and the OR for the A/A and A/G vs G/G genotype was 2.724 (95 % CI = 1.546–4.799, p = 0.0004). Moreover, we observed a contribution of the rs605059 SNP to histological grade G3 status. The p-value was 0.0042. The OR for the A/A vs G/G genotype was 5.632 (95 % CI = 1.644–19.290, p = 0.0026), the OR for the A/G vs G/G genotype was 4.213 (95%CI = 1.244–14.265, p = 0.0113) and the OR for the A/A and A/G vs G/G genotype was 4.780 (95 % CI = 1.456– 15.687, p = 0.0033). Our study indicated that the HSD17B1937 A > G transition is a risk factor for CT, especially for stages III and IV and histological grade G3. C1 [Lutkowska, Anna] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. [Roszak, Andrzej] Greater Poland Cancer Center, Department of Radiotherapy and Gynecological OncologyPoznan, Poland. [Jagodzinski, P Pawel] Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 6 Swiecickiego St, 60-781 Poznan, Poland. RP Jagodzinski, PP (reprint author), Poznan University of Medical Sciences, Department of Biochemistry and Molecular Biology, 60-781 Poznan, Poland. EM pjagodzi@am.poznan.pl CR Forouzanfar MH, Foreman KJ, Delossantos AM, Lozano R, Lopez AD, Murray CJ, Naghavi M, 2011, Breast and cervical cancer in 187 countries between 1980 and 2010: a systematic analysis. Lancet 378:1461–1484 Walboomers JM, JacobsMV,ManosMM, Bosch FX, Kummer JA, Shah KV, Snijders PJ, Peto J, Meijer CJ, Munoz N, 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189:12–19 Nguyen HP, Ramirez-Fort MK, Rady PL, 2014, The biology of human papillomaviruses. Curr Probl Dermatol 45:19–32 Moody CA, Laimins LA, 2010, Human papillomavirus oncoproteins: pathways to transformation. Nat Rev Cancer 10: 550–560 Kasap B, Yetimalar H, Keklik A, Yildiz A, Cukurova K, Soylu F, 2011, Prevalence and risk factors for human papillomavirus DNA in cervical cytology. Eur J Obstet Gynecol Reprod Biol 159:168–171 International Collaboration of Epidemiological Studies of Cervical Cancer, Appleby P, Beral V, de Berrington Gonzalez A, Colin D, Franceschi S, Goodhill A, Green J, Peto J, Plummer M, Sweetland S, 2007, Cervical cancer and hormonal contraceptives: collaborative reanalysis of individual data for 16,573 women with cervical cancer and 35,509 women without cervical cancer from 24 epidemiological studies. Lancet 370:1609–1621 Castellsague X, Munoz N, 2003, Chapter 3: cofactors in human papillomavirus carcinogenesis—role of parity, oral contraceptives, and tobacco smoking. J Natl Cancer Inst Monogr:20–28 Almonte M, Albero G, Molano M, Carcamo C, Garcia PJ, Perez G, 2008, Risk factors for human papillomavirus exposure and co factors for cervical cancer in Latin America and the Caribbean. Vaccine 26:L16–L36 Deligeoroglou E, Giannouli A, Athanasopoulos N, Karountzos V, Vatopoulou A, Dimopoulos K, Creatsas G, 2013, HPV infection: Immunological aspects and their utility in future therapy. Infect Dis Obstet Gynecol 2013:540850 Ault KA, 2006, Epidemiology and natural history of human papillomavirus infections in the female genital tract. Infect Dis Obstet Gynecol 2006:40470 De Azambuja K, Barman P, Toyama J, Elashoff D, Lawson GW, Williams LK, Chua K, Lee D, Kehoe JJ, Brodkorb A, Schwiebert R, Kitchen S, Bhimani A,Wiley DJ, 2014, Validation of an HPV16- mediated carcinogenesis mouse model. In Vivo 28:761–767 Shai A, Brake T, Somoza C, Lambert PF, 2007, The human papillomavirus E6 oncogene dysregulates the cell cycle and contributes to cervical carcinogenesis through two independent activities. Cancer Res 67:1626–1635 Elson DA, Riley RR, Lacey A, Thordarson G, Talamantes FJ, Arbeit JM, 2000, Sensitivity of the cervical transformation zone to estrogen-induced squamous carcinogenesis. Cancer Res 60: 1267–1275 Cortes-Malagon EM, Bonilla-Delgado J, Diaz-Chavez J, Hidalgo- Miranda A, Romero-Cordoba S, Uren A, Celik H, McCormick M, Munguia-Moreno JA, Ibarra-Sierra E, Escobar-Herrera J, Lambert PF, Mendoza-Villanueva D, Bermudez-Cruz RM, Gariglio P, 2013, Gene expression profile regulated by the HPV16 E7 oncoprotein and estradiol in cervical tissue. Virology 447:155–165 Mitrani-Rosenbaum S, Tsvieli R, Tur-Kaspa R, 1989, Oestrogen stimulates differential transcription of human papillomavirus type 16 in SiHa cervical carcinoma cells. J Gen Virol 70:2227–2232 Correa I, Cerbon MA, Salazar AM, Solano JD, Garcia-Carranca A, Quintero A, 2002, Differential p53 protein expression level in human cancer-derived cell lines after estradiol treatment. Arch Med Res 33:455–459 Ruutu M,Wahlroos N, Syrjanen K, Johansson B, Syrjanen S, 2006, Effects of 17beta-estradiol and progesterone on transcription of human papillomavirus 16 E6/E7 oncogenes in CaSki and SiHa cell lines. Int J Gynecol Cancer 16:1261–1268 Wang Q, Li X, Wang L, Feng YH, Zeng R, Gorodeski G, 2004, Antiapoptotic effects of estrogen in normal and cancer human cervical epithelial cells. Endocrinology 145:5568–5579 Huang Y, Li J, Xiang L, Han D, Shen X, Wu X, 2012, 17β- Oestradiol activates proteolysis and increases invasion through phosphatidylinositol 3-kinase pathway in human cervical cancer cells. Eur J Obstet Gynecol Reprod Biol 165:307–312 Arbeit JM, Howley PM, Hanahan D, 1996, Chronic estrogeninduced cervical and vaginal squamous carcinogenesis in human papillomavirus type 16 transgenic mice. Proc Natl Acad Sci U S A 93:2930–2935 Brake T, Lambert PF, 2005, Estrogen contributes to the onset, persistence, and malignant progression of cervical cancer in a human papillomavirus-transgenic mouse model. ProcNatl Acad Sci U S A 102:2490–2495 Simpson ER, 2003, Sources of estrogen and their importance. J Steroid Biochem Mol Biol 86:225–230 Simpson E, Rubin G, Clyne C, Robertson K,O’Donnell L,Davis S, Jones M, 1999, Local estrogenbiosynthesis in males and females. Endocr Relat Cancer 6:131–137 Luu-The V, Labrie F, 2010, The intracrine sex steroid biosynthesis pathways. Prog Brain Res 181:177–192 Lukacik P, Kavanagh KL, Oppermann U(2006, Structure and function of human 17-hydroxysteroid dehydrogenases. Mol Cell Endocrinol 248:61–71 Vihko P, Isomaa V, Ghosh D, 2001, Structure and function of 17beta-hydroxysteroid dehydrogenase type 1 and type 2. Mol Cell Endocrinol 171:71–76 Sawetawan C, Milewich L,Word RA, Carr BR, RaineyWE, 1994, Compartmentalization of type I 17β-hydroxysteroid oxidoreductase in the human ovary. Mol Cell Endocrinol 99:161–168 Fournet-Dulguerov N, MacLusky NJ, Leranth CZ, Todd R, Mendelson CR, Simpson ER, N a f t o l i n F, 1987, Immunohistochemical localization of aromatase cytochrome P-450 and estradiol dehydrogenase in the syncytiotrophoblast of the human placenta. J Clin Endocrinol Metab 65:757–764 Feigelson HS, McKean-Cowdin R, Coetzee GA, Stram DO, Kolonel LN, Henderson BE, 2001, Building a multigenic model of breast cancer susceptibility: CYP17 and HSD17B1 are two important candidates. Cancer Res 61:785–789 AH W, Seow A, Arakawa K, Van Den Berg D, Lee HP, MC Y, 2003, HSD17B1 and CYP17 polymorphisms and breast cancer risk among Chinese women in Singapore. Int J Cancer 104:450–457 Yao L, Cao LH, Qiu LX, Yu L, 2010, The association between HSD17B1 Ser312Gly polymorphism and breast cancer risk: a meta-analysis including 31,053 subjects. Breast Cancer Res Treat 123:577–580 Kato I, Cichon M, Yee CL, Land S, Korczak JF, 2009, African American-preponderant single nucleotide polymorphisms, SNPs, and risk of breast cancer. Cancer Epidemiol 33:24–30 Obazee O, Justenhoven C, Winter S, Chang-Claude J, Rudolph A, Seibold P, Flesch-Janys D, Hannelius U, Li J, Humphreys K, Hall P, Giles G, Severi G, Baglietto L, Southey M, Rabstein S, Harth V, Lotz A, Pesch B, Bruning T, Baisch C, Ko YD, Hamann U, Brauch H, 2013, Confirmation of the reduction of hormone replacement therapy-related breast cancer risk for carriers of the HSD17B1_937_G variant. Breast Cancer Res Treat 138:543–548 Iwasaki M, Hamada GS, Nishimoto IN, Netto MM, Motola J Jr, Laginha FM, Kasuga Y, Yokoyama S, Onuma H, Nishimura H, Kusama R, Kobayashi M, Ishihara J, Yamamoto S, Hanaoka T, Tsugane S, 2010, Dietary isoflavone intake, polymorphisms in the CYP17, CYP19, 17beta-HSD1, and SHBG genes, and risk of breast cancer in case-control studies in Japanese, Japanese Brazilians, and non-Japanese Brazilians. Nutr Cancer 62:466–475 Luu-The V, 2001, Analysis and characteristics of multiple types of human 17beta-hydroxysteroid dehydrogenase. J Steroid Biochem Mol Biol 76:143–151 Miettinen M, Mustonen M, Poutanen M, Isomaa V, Wickman M, Soderqvist G, Vihko R, Vihko P, 1999, 17Beta-hydroxysteroid dehydrogenases in normal human mammary epithelial cells and breast tissue. Breast Cancer Res Treat 57:175–182 Vihko P, Harkonen P, Soronen P, Torn S, Herrala A, Kurkela R, Pulkka A, Oduwole O, Isomaa V, 2004, 17 beta-hydroxysteroid dehydrogenases–their role in pathophysiology. Mol Cell Endocrinol 215:83–88 Tomaszewska A, Roszak A, Pawlik P, Sajdak S, Jagodzinski PP, 2015, Increased 17ß-hydroxysteroid dehydrogenase type 1 levels in primary cervical cancer. Biomed Pharmacother 72:179–183 Jaakkola S, Pukkala E, Lyytinen HK, Ylikorkala O, 2012, Postmenopausal estradiol–progestagen therapy and risk for uterine cervical cancer. Int J Cancer 131:E537–E543 Cong RJ, Huang ZY, Cong L,Ye Y,Wang Z, Zha L, Cao LP,XWS, Yan J, Li YB, 2012, Polymorphisms in genes HSD17B1 and HSD17B2 and uterine leiomyoma risk in Chinese women. Arch Gynecol Obstet 286:701–705 Setiawan VW, Hankinson SE, Colditz GA, Hunter DJ, De Vivo I, 2004, HSD17B1 gene polymorphisms and risk of endometrial and breast cancer. Cancer Epidemiol Biomark Prev 13:213–219 Rinaldi S, Plummer M, Biessy C, Castellsague X, Overvad K, Kruger Kjaer S, Tjonneland A, Clavel-Chapelon F, Chabbert- Buffet N, Mesrine S, Lukanova A, Kaaks R, Weikert C, Boeing H, Trichopoulou A, Lagiou P, Trichopoulos D, Palli D, Agnoli C, Tumino R, Vineis P, Panico S, Bueno-de-Mesquita B, van Kranen HJ, Peeters PH, Bakken K, Lund E, Gram IT, Rodriguez L, Bosch FX, Sanchez MJ, Dorronsoro M, Navarro C, Gurrea AB, Kjellberg L, Dillner J, Manjer J, Butt S, Khaw KT, Wareham N, Allen NE, Travis R, Romieu I, Ferrari P, Riboli E, Franceschi S, 2011, Endogenous sex steroids and risk of cervical carcinoma: results from the EPIC study. Cancer Epidemiol Biomark Prev 20:2532–2540 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 317 EP 322 DI 10.1007/s12253-016-0103-4 PG 6 ER PT J AU Tan, Ch Qian, X Ge, Y Yang, B Wang, F Guan, Z Cai, J AF Tan, Cheng Qian, Xia Ge, Yangyang Yang, Baixia Wang, Feng Guan, Zhifeng Cai, Jing TI Oroxylin a could be a Promising Radiosensitizer for Esophageal Squamous Cell Carcinoma by Inducing G2/M Arrest and Activating Apoptosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE OroxylinA; Radiosensitization; ESCC; Apoptosis; G2/M arrest ID OroxylinA; Radiosensitization; ESCC; Apoptosis; G2/M arrest AB To evaluate the radiosensitization of Oroxylin A on esophageal carcinoma cell as well as the optimal scheduling of Oroxylin A and radiotherapy (RT). Cell proliferation was estimated by aCCK8 assay.Radiosensitizationwas evaluated by a clonogenic survival assay. The progressions of Cell apoptosis and Cell cycle were investigated by flow cytometry. Expressions of survivin and cell cycle regulators were evaluated by Western blot analysis. A dose-dependent cell survival reduction was found in response to radiation with or without Oroxylin A. The apoptosis rates were remarkably dosedependent higher in combination groups than in either Oroxylin A or radiation alone group. Besides, Oroxylin A could obviously radiosensitize ESCC cells by arresting tumor cells in G2/M phase and regulating cyclin B1 and Cdc 2 protein expression. Oroxylin A could be a promising radiosensitizer for esophageal squamous cell carcinoma by inducing G2/M phase blocking and activating cell apoptosis. C1 [Tan, Cheng] Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Department of Radiation Oncology, 226321 Nantong, China. [Qian, Xia] Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Department of Radiation Oncology, 226321 Nantong, China. [Ge, Yangyang] Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Department of Radiation Oncology, 226321 Nantong, China. [Yang, Baixia] Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Department of Radiation Oncology, 226321 Nantong, China. [Wang, Feng] Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Department of Radiation Oncology, 226321 Nantong, China. [Guan, Zhifeng] Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Department of Radiation Oncology, 226321 Nantong, China. [Cai, Jing] Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Department of Radiation Oncology, 226321 Nantong, China. RP Cai, J (reprint author), Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Department of Radiation Oncology, 226321 Nantong, China. EM cj7227@sina.com CR Zali H, Ahmadi G, Bakhshandeh R, Rezaei-Tavirani M, 2011, Proteomic analysis of gene expression during human esophagus cancer. J Paramedical Sci 2:37–44 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90 Thompson SK, Ruszkiewicz AR, Jamieson GG, Esterman A, Watson DI, Wijnhoven BP, et al., 2008, Improving the accuracy of TNM staging in esophageal cancer: a pathological review of resected specimens. Ann Surg Oncol 15(12):3447– 3458 ZhuH,Wang Q, Hu C, ZhangW, Quan L, LiuM, et al., 2011, High expression of survivin predicts poor prognosis in esophageal squamous cell carcinoma following radiotherapy. Tumour Biol 32: 1147–1153 Li J, Li ZN, Bao QL, Ge LP, Li XQ, Chen P, 2012, Evaluation of pleural fluid survivin and XIAP for the diagnosis of malignant pleural effusion. Tumour Biol 33:1803–1810 Salvesen GS, Duckett CSIAP, 2002, Proteins: blocking the road to death’s door. Nat Rev Mol Cell Biol 3:401–410 Wei L, Zhou Y, Qiao C, Ni T, Li Z, You Q, et al., 2015, Oroxylin a inhibits glycolysis-dependent proliferation of human breast cancer via promoting SIRT3-mediated SOD2 transcription and HIF1alpha destabilization. Cell Death Dis 6:e1714 Dinda B, SilSarma I, Dinda M, Rudrapaul P, 2015, Oroxylum indicum, L., Kurz, an important Asian traditional medicine: from traditional uses to scientific data for its commercial exploitation. J Ethnopharmacol 161:255–278 Minsky BD, Pajak TF, Ginsberg RJ, et al., 2002, INT 0123, Radiation Therapy Oncology Group 94–05, phase III trial of combinedmodality therapy for esophageal cancer: high-dose versus standarddoseradiation therapy. J Clin Oncol 20:1167–1174 Zou M, Hu C, You Q, Zhang A, Wang X, Guo Q, Oroxylin A, 2015, Induces autophagy in human malignant glioma cells via the mTOR-STAT3-notch signaling pathway. Mol Carcinog 54(11):1363–1375 Wei L, Dai Q, Zhou Y, ZouM, Li Z, Lu N, et al., 2013, Oroxylin a sensitizes non-small cell lung cancer cells to anoikis via glucosedeprivation- like mechanisms: c-Src and hexokinase II. Biochim Biophys Acta 1830(6):3835–3845 Cho HJ, YJ O, Han SH, Chung HJ, Kim CH, Lee NS, et al., 2013, Cdk1 protein-mediated phosphorylation of receptor-associated protein 80, RAP80, serine 677 modulates DNA damage-induced G2/ M checkpoint and cell survival. J Biol Chem 288(6):3768–3776 Hengartner MO, 2000, The biochemistry of apoptosis. Nature 407(6805):770–776 Pawlik TM, Keyomarsi K, 2004, Role of cell cycle in mediating sensitivity to radiotherapy. Int J Radiat Oncol Biol Phys 59(4):928–942 Saez-Ayala M, Cabezas-Herrera J, Rodriguez-Lopez JN, 2015, Targeting the epigenetic machinery of cancer cells. Oncogene 34(2):135–143 Herscher LL, 1999, Taxanes as radiosensitizers for head and neck cancer. Curr Opin Oncol 11(3):183–186 Zhai X, Yang Y, Wan J, Zhu R, Inhibition WY, 2013, Of LDH-A by oxamate induces G2/M arrest, apoptosis and increases radiosensitivity in nasopharyngeal carcinoma cells. Oncol Rep 30(6):2983– 2991 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 323 EP 328 DI 10.1007/s12253-016-0106-1 PG 6 ER PT J AU Zhang, Z He, Q Fu, S Zheng, Z AF Zhang, Zhe He, Qinsi Fu, Si Zheng, Zhi TI Estrogen Receptors in Regulating Cell Proliferation of Esophageal Squamous Cell Carcinoma: Involvement of Intracellular Ca2+ Signaling SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Estrogen receptor; Esophageal squamous cell carcinoma; Gender difference; Cell proliferation; Ca2+ signaling ID Estrogen receptor; Esophageal squamous cell carcinoma; Gender difference; Cell proliferation; Ca2+ signaling AB Esophageal cancer is a deadly disease in the esophagus with a poor prognosis. Over 90 % of esophageal cancer is esophageal squamous cell carcinoma (ESCC) and its pathogenic mechanisms remain unclear. Epidemiology study found a strong gender difference with a sex ratio of 8–9:1 in favor of males, but the molecular mechanisms for so striking gender difference are poorly understood so far. In the present study, we demonstrated the expression of estrogen receptors in human ESCC cells. 17β-E2 but not 17α-E2 was found to dosedependently suppress the cell proliferation of human ESCC cells, which was attenuated by estrogen receptor antagonist ICI1 82,780. Furthermore, 17β -E2 but not 17α-E2 10 nM markedly induced both intracellular Ca2+ release and extracellular Ca2+ entry into ESCC cells, which was again attenuated by estrogen receptor antagonist ICI182,780. Taken together, our data clearly demonstrate that estrogen exerts anti-proliferative action on human ESCC cells likely through estrogen receptor-Ca2+ signaling pathway, which may provide a reasonable explanation on the striking male predominance of ESCC. C1 [Zhang, Zhe] China-Japan Friendship Hospital, Department of Chinese Medical Gastrointestinal, 2 Yinghua Dongjie, 100029 Beijing, Beijing, China. [He, Qinsi] Nanchang University, 330029 Nanchang, Jiangxi, China. [Fu, Si] China-Japan Friendship Hospital, Department of Chinese Medical Gastrointestinal, 2 Yinghua Dongjie, 100029 Beijing, Beijing, China. [Zheng, Zhi] Nanchang University, 330029 Nanchang, Jiangxi, China. RP Zheng, Z (reprint author), Nanchang University, 330029 Nanchang, China. EM zheng_sheva@hotmail.com CR Kazerounian S, Pitari GM, Shah FJ, Frick GS, Madesh M, Ruiz- Stewart I, Schulz S, Hajnoczky G, Waldman SA, 2005, Proliferative signaling by store-operated calcium channels opposes colon cancer cell cytostasis induced by bacterial enterotoxins. J Pharmacol Exp Ther 314:1013–1022 Blot WJ, McLaughlin JK, 1999, The changing epidemiology of esophageal cancer. Semin Oncol 26:2–8 Wang Z, Tang L, Sun G, TangY, XieY,Wang S, Hu X, GaoW, Cox SB,Wang JS, 2006, Etiological study of esophageal squamous cell carcinoma in an endemic region: a population-based case control study in Huaian, China. BMC Cancer 6:287 Hogan AM, Collins D, Baird AW,Winter DC, 2009, Estrogen and its role in gastrointestinal health and disease. Int J Color Dis 24: 1367–1375 Wang QM, Qi YJ, Jiang Q, Ma YF,Wang LD, 2011, Relevance of serum estradiol and estrogen receptor beta expression from a highincidence area for esophageal squamous cell carcinoma in China. Med Oncol 28:188–193 Yang H, Sukocheva OA, Hussey DJ, Watson DI, 2012, Estrogen, male dominance and esophageal adenocarcinoma: is there a link? World J Gastroenterol 18:393–400 Prossnitz ER, Barton M, 2011, The G-protein-coupled estrogen receptor GPER in health and disease. Nat Rev Endocrinol 7:715– 726 Litle VR, Rice TW, 2011, The esophagus: do sex and gender matter? Semin Thorac Cardiovasc Surg 23:131–136 Liao WX, Magness RR, Chen DB, 2005, Expression of estrogen receptors-alpha and -beta in the pregnant ovine uterine artery endothelial cells in vivo and in vitro. Biol Reprod 72:530–537 Jiang HP, Teng RY, Wang Q, Zhang X,Wang HH, Cao J, Teng LS, 2008, Estrogen receptor alpha variant ERalpha46 mediates growth inhibition and apoptosis of human HT-29 colon adenocarcinoma cells in the presence of 17beta-oestradiol. Chin Med J 121:1025– 1031 Notarnicola M, Messa C, Orlando A, Bifulco M, Laezza C, Gazzerro P, Caruso MG, 2008, Estrogenic induction of cannabinoid CB1 receptor in human colon cancer cell lines. Scand J Gastroenterol 43:66–72 Renoir JM, Marsaud V, Lazennec G, 2013, Estrogen receptor signaling as a target for novel breast cancer therapeutics. Biochem Pharmacol 85:449–465 Boros S, Bindels RJ, Hoenderop JG, 2009, Active Ca(2+, reabsorption in the connecting tubule. Pflugers Arch 458:99–109 Peters AA, Simpson PT, Bassett JJ, Lee JM, Da Silva L, Reid LE, Song S, Parat MO, Lakhani SR, Kenny PA, Roberts-Thomson SJ, Monteith GR, 2012, Calcium channel TRPV6 as a potential therapeutic target in estrogen receptor-negative breast cancer. Mol Cancer Ther 11:2158–2168 Kahl CR, Means AR, 2003, Regulation of cell cycle progression by calcium/calmodulin-dependent pathways. Endocr Rev 24:719–736 Freedman ND, Lacey JV Jr, Hollenbeck AR, Leitzmann MF, Schatzkin A, Abnet CC, 2010, The association of menstrual and reproductive factors with upper gastrointestinal tract cancers in the NIH-AARP cohort. Cancer 116:1572–1581 Bodelon C, Anderson GL, Rossing MA, Chlebowski RT, Ochs- Balcom HM, Vaughan TL, 2011, Hormonal factors and risks of esophageal squamous cell carcinoma and adenocarcinoma in postmenopausal women. Cancer Prev Res, Phila, 4:840–850 Zuguchi M, Miki Y, Onodera Y, Fujishima F, Takeyama D, Okamoto H, Miyata G, Sato A, Satomi S, Sasano H, 2012, Estrogen receptor alpha and beta in esophageal squamous cell carcinoma. Cancer Sci 103:1348–1355 Levin ER, 2002, Cellular functions of plasma membrane estrogen receptors. Steroids 67:471–475 Levin ER, 1999, Cellular functions of the plasma membrane estrogen receptor. Trends Endocrinol Metab 10:374–377 Wang JY,Wang J,GolovinaVA, Li L, Platoshyn O,Yuan JX, 2000, Role of K(+, channel expression in polyamine-dependent intestinal epithelial cell migration. Am J Phys Cell Physiol 278:C303–C314 Pandiella A, Magni M, Lovisolo D, Meldolesi J, 1989, The effect of epidermal growth factor on membrane potential. Rapid hyperpolarization followed by persistent fluctuations. J Biol Chem 264: 12914–12921 Hazelton B,Mitchell B, Tupper J, 1979, Calcium, magnesium, and growth control in the WI-38 human fibroblast cell. J Cell Biol 83: 487–498 Pigozzi D, Ducret T, Tajeddine N, Gala JL, Tombal B, Gailly P, 2006, Calcium store contents control the expression of TRPC1, TRPC3 and TRPV6 proteins in LNCaP prostate cancer cell line. Cell Calcium 39:401–415 Sergeev IN, Vitamin D, 2012, Cellular Ca2+ signaling in breast cancer. Anticancer Res 32:299–302 Monteith GR, Davis FM, Roberts-Thomson SJ, 2012, Calcium channels and pumps in cancer: changes and consequences. J Biol Chem 287:31666–31673 Monteith GR, McAndrew D, Faddy HM, Roberts-Thomson SJ, 2007, Calcium and cancer: targeting Ca2+ transport. Nat Rev Cancer 7:519–530 Sarkar SN, Huang RQ, Logan SM, Yi KD, Dillon GH, Simpkins JW, 2008, Estrogens directly potentiate neuronal L-type Ca2+ channels. Proc Natl Acad Sci U S A 105:15148–15153 LL Y, Zhang JH, He YP, Huang P, Yue LM, 2009, Fast action of estrogen on intracellular calcium in dormant mouse blastocyst and its possible mechanism. Fertil Steril 91:611–615 Guo Z, Krucken J, Benten WP, Wunderlich F, 2002, Estradiolinduced nongenomic calcium signaling regulates genotropic signaling in macrophages. J Biol Chem 277:7044–7050 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 329 EP 334 DI 10.1007/s12253-016-0105-2 PG 6 ER PT J AU Ljujic, M Mijatovic, S Bulatovic, ZM Mojic, M Maksimovic-Ivanic, D Radojkovic, D Topic, A AF Ljujic, Mila Mijatovic, Sanja Bulatovic, Z Mirna Mojic, Marija Maksimovic-Ivanic, Danijela Radojkovic, Dragica Topic, Aleksandra TI Alpha-1-Antitrypsin Antagonizes Cisplatin-Induced Cytotoxicity in Prostate Cancer (PC3) and Melanoma Cancer (A375) Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Alpha-1-antitrypsin; Cisplatin; Prostate cancer; Melanoma cancer ID Alpha-1-antitrypsin; Cisplatin; Prostate cancer; Melanoma cancer AB Increased circulating alpha-1-antitrypsin (AAT) correlates with cancer stage/aggressiveness, but its role in cancer biology is unclear.We revealed antagonistic effect of AAT to cisplatin-induced cytotoxicity in prostate (PC3) and melanoma (A375) cancer cell lines. Moreover, AAT abrogated cytotoxicity of MEK inhibitor U0126 in PC3 cell line. Weaker antagonistic effect of AAT on cytotoxicity of PI3/Akt and NFkB inhibitors was also observed. In addition, cisplatin increased AAT gene expression in transfected PC3 cells. However, AAT derived from transfected PC3 cells did not antagonize cisplatin-induced cytotoxicity. In conclusion, these results suggest possible association between high circulating AAT and cisplatin resistance. C1 [Ljujic, Mila] University of Belgrade, Institute of Molecular Genetics and Genetic Engineering, Vojvode Stepe 444a, 11221 Belgrade, Serbia. [Mijatovic, Sanja] University of Belgrade, Institute for Biological Research, Bulevar despota Stefana 142, 11060 Belgrade, Serbia. [Bulatovic, Z Mirna] University of Belgrade, Institute for Biological Research, Bulevar despota Stefana 142, 11060 Belgrade, Serbia. [Mojic, Marija] University of Belgrade, Institute for Biological Research, Bulevar despota Stefana 142, 11060 Belgrade, Serbia. [Maksimovic-Ivanic, Danijela] University of Belgrade, Institute for Biological Research, Bulevar despota Stefana 142, 11060 Belgrade, Serbia. [Radojkovic, Dragica] University of Belgrade, Institute of Molecular Genetics and Genetic Engineering, Vojvode Stepe 444a, 11221 Belgrade, Serbia. [Topic, Aleksandra] Belgrade University, Faculty of Pharmacy, Institute of Medical Biochemistry, Vojvode Stepe 450, 11221 Belgrade, Serbia. RP Topic, A (reprint author), Belgrade University, Faculty of Pharmacy, Institute of Medical Biochemistry, 11221 Belgrade, Serbia. EM aleksandra.topic1@gmail.com CR Crystal RG, 1989, The alpha 1-antitrypsin gene and its deficiency states. Trends Genet 5:411–417 Blank CA, Brantly M, 1994, Clinical features and molecular characteristics of alpha 1-antitrypsin deficiency. Ann Allergy 72:105– 120 Mason DY, Cramer EM, Masse JM, Crystal R, Bassot JM, Breton- Gorius J, 1991, Alpha 1-antitrypsin is present within the primary granules of human polymorphonuclear leukocytes. Am J Pathol 139:623–628 Molmenti EP, Perlmutter DH, Rubin DC, 1993, Cell-specific expression of alpha 1-antitrypsin in human intestinal epithelium. J Clin Invest 92:2022–2034 Perlmutter DH, Daniels JD, Auerbach HS, De Schryver- Kecskemeti K, Winter HS, Alpers DH, 1989, The alpha 1- antitrypsin gene is expressed in a human intestinal epithelial cell line. J Biol Chem 264:9485–9490 Sallenave JM, Tremblay GM, Gauldie J, Richards CD, 1997, Oncostatin M, but not interleukin-6 or leukemia inhibitory factor, stimulates expression of alpha1-proteinase inhibitor in A549 human alveolar epithelial cells. J Interf Cytokine Res 17:337–346 Cichy J, Potempa J, Travis J, 1997, Biosynthesis of alpha1- proteinase inhibitor by human lung-derived epithelial cells. J Biol Chem 272:8250–8255 Ikari Y, Mulvihill E, Schwartz SM, 2001, Alpha 1-Proteinase inhibitor, alpha 1-antichymotrypsin, and alpha 2-macroglobulin are the antiapoptotic factors of vascular smooth muscle cells. J Biol Chem 276:11798–11803 Daemen MA, Heemskerk VH, van’t Veer C, Denecker G, TG W, Vandenabeele P,WA B, 2000, Functional protection by acute phase proteins alpha(1)-acid glycoprotein and alpha(1)-antitrypsin against ischemia/reperfusion injury by preventing apoptosis and inflammation. Circulation 102:1420–1426 Petrache I, Fijalkowska I, Zhen L, Medler TR, Brown E, Cruz P, Choe KH, Taraseviciene-Stewart L, Scerbavicius R, Shapiro L, Zhang B, Song S, Hicklin D, Voelkel NF, Flotte T, Tuder RM, 2006, A novel antiapoptotic role for alpha1-antitrypsin in the prevention of pulmonary emphysema.AmJ Respir Crit CareMed 173: 1222–1228 Greene CM, Miller SD, Carroll TP, Oglesby IK, Ahmed F, O’Mahony M, Taggart CC, McElvaney NG, O’Neill SJ, 2010, Anti-apoptotic effects of Z alpha1-antitrypsin in human bronchial epithelial cells. Eur Respir J 35:1155–1163 Petrache I, Fijalkowska I, Medler TR, Skirball J, Cruz P, Zhen L, Petrache HI, Flotte TR, Tuder RM, 2006, Alpha-1 antitrypsin inhibits caspase-3 activity, preventing lung endothelial cell apoptosis. Am J Pathol 169:1155–1166 Lockett AD, Van Demark M, Gu Y, Schweitzer KS, Sigua N, Kamocki K, Fijalkowska I, Garrison J, Fisher AJ, Serban K, Wise RA, Flotte TR, Mueller C, RG P Jr, Petrache HI, Tuder RM, Petrache I, 2012, Effect of cigarette smoke exposure and structural modifications on the α-1 antitrypsin interaction with caspases. Mol Med 18:445–454 HigashiyamaM, Doi O, Kodama K, Yokouchi H, Tateishi R, 1992, An evaluation of the prognostic significance of alpha-1-antitrypsin expression in adenocarcinomas of the lung: an immunohistochemical analysis. Br J Cancer 65:300–302 Kwon CH, Park HJ, Lee JR, Kim HK, Jeon TY, Jo HJ, Kim DH, Kim GH, Park DY, 2014, Serpin peptidase inhibitor clade a member 1 is a biomarker of poor prognosis in gastric cancer. Br J Cancer 111:1993–2002 Chang YH, Lee SH, Liao IC, Huang SH, Cheng HC, Liao PC, 2012, Secretomic analysis identifies alpha-1 antitrypsin, A1AT, as a required protein in cancer cell migration, invasion, and pericellular fibronectin assembly for facilitating lung colonization of lung adenocarcinoma cells. Mol Cell Proteomics 11:1320–1339 Kataoka H, Uchino H, Iwamura T, Seiki M, Nabeshima K, Koono M, 1999, Enhanced tumor growth and invasiveness in vivo by a carboxyl-terminal fragment of alpha1-proteinase inhibitor generated by matrix metalloproteinases: a possible modulatory role in natural killer cytotoxicity. Am J Pathol 154:457–468 Zelvyte I, Lindgren S, Janciauskiene S, 2003, Multiple effects of alpha1-antitrypsin on breast carcinoma MDA-MB 468 cell growth and invasiveness. Eur J Cancer Prev 12:117–124 ZhaoW, Yang Z, Liu X, Tian Q, Lv Y, Liang Y, Li C, Gao X, Chen L, 2013, Identification of α1-antitrypsin as a potential prognostic biomarker for advanced nonsmall cell lung cancer treated with epidermal growth factor receptor tyrosine kinase inhibitors by proteomic analysis. J Int Med Res 41:573–583 Topic A, Ljujic M, Petrovic-Stanojevic N, Dopudja-Pantic V, Radojkovic D, 2011, Phenotypes and serum level of alpha-1- antitrypsin in lung cancer. J BUON 16:672–676 Varela AS, Lopez Saez JJ, 1995, Utility of plasmatic levels of alpha-1-antiprotease, A1AP, as a cancer marker. Cancer Lett 89: 15–21 Perez-Holanda S, Blanco I, MenendezM, Rodrigo L, 2014, Serum concentration of alpha-1 antitrypsin is significantly higher in colorectal cancer patients than in healthy controls. BMC Cancer 14:355 Mijatovic S, Maksimovic-Ivanic D, Radovic J, Popadic D, Momcilovic M, Harhaji L, Miljkovic D, Trajkovic V, 2004, Aloe-emodin prevents cytokine-induced tumor cell death: the inhibition of auto-toxic nitric oxide release as a potential mechanism. Cell Mol Life Sci 61:1805–1815 McCubrey JA, Steelman LS, Abrams SL, Misaghian N, Chappell WH, Basecke J, Nicoletti F, Libra M, Ligresti G, Stivala F, Maksimovic-Ivanic D, Mijatovic S, Montalto G, Cervello M, Laidler P, Bonati A, Evangelisti C, Cocco L, Martelli AM, 2012, Targeting the cancer initiating cell: the ultimate target for cancer therapy. Curr Pharm Des 18:1784–1795 EMBL-EBI Expression Atlas, available on: http://www.ebi.ac. uk/gxa/experiments. Kubisch R, Meissner L, Krebs S, Blum H, Gunther M, Roidl A, Wagner EA, 2013, Comprehensive gene expression analysis of resistance formation upon metronomic cyclophosphamide therapy. Transl Oncol 6:1–9 El-Akawi ZJ, Abu-Awad AM, Sharara AM, Khader Y, 2010, The importance of alpha-1 antitrypsin, alpha1-AT, and neopterin serum levels in the evaluation of non-small cell lung and prostate cancer patients. Neuroendocrinol Lett 31:113–116 Zelvyte I, Wallmark A, Piitulainen E, Westin U, Janciauskiene S, 2004, Increased plasma levels of serine proteinase inhibitors in lung cancer patients. Anticancer Res 24:241–247 Li Y, Krowka MJ, Qi Y, Katzmann JA, Song Y, Mandrekar SJ, Yang P, 2011, Alpha1-antitrypsin deficiency carriers, serum alpha 1-antitrypsin concentration, and non-small cell lung cancer survival. J Thorac Oncol 6:291–295 Normandin K, Peant B, Le Page C, de Ladurantaye M, Ouellet V, Tonin PN, Provencher DM, Mes-Masson AM, 2010, Protease inhibitor SERPINA1 expression in epithelial ovarian cancer. Clin Exp Metastasis 27:55–69 Zelvyte I, Ohlsson B, Axelson J, Janciauskiene S, 2003, Diverse responses between human pancreatic cancer cell lines to native alpha 1-antitrypsin and its C-terminal fragment. Anticancer Res 23(3B):2267–2273 Zelvyte I, Sjogren HO, Janciauskiene S, 2002, Effects of native and cleaved forms of alpha1-antitrypsin on ME 1477 tumor cell functional activity. Cancer Detect Prev 26:256–265 Boulikas T, Vougiouka M, 2003, Cisplatin and platinum drugs at the molecular level. Oncol Rep 10:1663–1682 Fuertes MA, Castilla J, Alonso C, Perez JM, 2003, Cisplatin biochemical mechanism of action: from cytotoxicity to induction of cell death through interconnections between apoptotic and necrotic pathways. Curr Med Chem 10:257–266 Ljujic M, Mijatovic S, Bulatovic MZ, Mojic M, Maksimovic- Ivanic D, Radojkovic D, TopicA, 2016, Alpha-1 antitrypsin affects U0126-induced cytotoxicity in colon cancer cell line, HCT116. Mol Biol 50:153–156 Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD, Levine B, 2005, Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 122:927–939 Marquez RT, Xu L, 2012, Bcl-2:Beclin 1 complex: multiple, mechanisms regulating autophagy/apoptosis toggle switch. Am J Cancer Res 2:214–221 Wei Y, Pattingre S, Sinha S, Bassik M, Levine B, 2008, JNK1- mediated phosphorylation of Bcl-2 regulates starvation-induced autophagy. Mol Cell 30:678–688 Chekhun VF, Kovtonyuk OV, Todor IN, Kulik GI, 2005, Total proteolytic activity and levels of the main proteinase inhibitors in blood plasma of mice bearing Lewis lung carcinoma upon development of resistance to cisplatin. Exp Oncol 27:286–289 Dabbagh K, Laurent GJ, Shock A, Leoni P, Papakrivopoulou J, Chambers RC, 2001, Alpha-1-antitrypsin stimulates fibroblast proliferation and procollagen production and activates classical MAP kinase signalling pathways. J Cell Physiol 186:73–81 Gonias SL, Swaim MW, Massey MF, Pizzo SV, 1988, cisdichlorodiammineplatinum, II, as a selective modifier of the oxidation-sensitive reactive-center methionine in alpha 1- antitrypsin. J Biol Chem 263:393–397 Kloth JN, Gorter A, Fleuren GJ, Oosting J, Uljee S, ter Haar N, Dreef EJ, Kenter GG, Jordanova ES, 2008, Elevated expression of SerpinA1 and SerpinA3 in HLA-positive cervical carcinoma. J Pathol 215:222–230 Kwon CH, Park HJ, Choi JH, Lee JR, Kim HK, Jo HJ, Kim HS, Oh N, Song GA, do Y P, 2015, Snail and serpinA1 promote tumor progression and predict prognosis in colorectal cancer. Oncotarget 6:20312–20326 JY W, Cheng CC, Wang JY, DC W, Hsieh JS, Lee SC, Wang WM, 2014, Discovery of tumor markers for gastric cancer by proteomics. PLoS One 9:e84158 Goodarzi MT, Turner GA, 1995, Decreased branching, increased fucosylation and changed sialylation of alpha-1-proteinase inhibitor in breast and ovarian cancer. Clin Chim Acta 236:161–171 Comunale MA, Rodemich-Betesh L, Hafner J,Wang M, Norton P, Di Bisceglie AM, Block T, Mehta A, 2010, Linkage specific fucosylation of alpha-1-antitrypsin in liver cirrhosis and cancer patients: implications for a biomarker of hepatocellular carcinoma. PLoS One 5:e12419 Wang Y, Kuramitsu Y, Yoshino S, Takashima M, Zhang X, Ueno T, Suzuki N, Oka M, Nakamura K, 2011, Screening for serological biomarkers of pancreatic cancer by two-dimensional electrophoresis and liquid chromatography-tandem mass spectrometry. Oncol Rep 26:287–292 Grivennikov SI, Greten FR, Karin M, 2010, Immunity, inflammation, and cancer. Cell 140:883–899 Huang W, Ding X, Li B, Fan M, Zhou T, Sun H, Yi Y, Zhang J, 2013, Serum biomarkers analyzed by LC-MS/MS as predictors for short outcome of non-small cell lung cancer patients treated with chemoradiotherapy. Neoplasma 60:11–18 Wozniak B, Mila-Kierzenkowska C, WoYniak A, Drewa G, Soponska M, Drewa T, Krzyzynska-Malinowska E, Makarewicz R, Kowalski T, Szmytkowska K, 2008, The effect of combined therapy on activity of cathepsin D and alpha-1-antitrypsin in the blood serum of women with cervical cancer. Eur J Gynaecol Oncol 29:617–619 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 335 EP 343 DI 10.1007/s12253-016-0104-3 PG 9 ER PT J AU Zepeda-Nuno, SJ Guerrero-Velazquez, C Del Toro-Arreola, S Vega-Magana, N Angeles-Sanchez, J Haramati, J Pereira-Suarez, LA Bueno-Topete, RM AF Zepeda-Nuno, Sergio Jose Guerrero-Velazquez, Celia Del Toro-Arreola, Susana Vega-Magana, Natali Angeles-Sanchez, Julian Haramati, Jesse Pereira-Suarez, L Ana Bueno-Topete, R Miriam TI Expression of ADAM10, Fas, FasL and Soluble FasL in Patients with Oral Squamous Cell Carcinoma (OSCC) and their Association with Clinical-Pathological Parameters SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral cancer; ADAM10; sFasL; Fas; FasL ID Oral cancer; ADAM10; sFasL; Fas; FasL AB ADAM10 has been implicated in the progression of various solid tumors. ADAM10 regulates the cleavage of the FasL ectodomain from the plasma membrane of different cell types, generating the soluble FasL fragment (sFasL). Currently, there are few studies in oral squamous cell carcinoma (OSCC) that correlate levels of ADAM10 and FasL in the tumor microenvironment with clinical parameters of the disease. To determine the expression of ADAM10, Fas, FasL and sFasL in patients with OSCC and its association with TNM stage. Twenty-five patients with OSCC and 25 healthy controls were included. Biopsies of tumor tissue from patients with OSCC and buccal mucosa in controls were obtained. ADAM10, Fas, and FasL were analyzed by Western blotting. sFasL was quantified by ELISA. ADAM10 and Fas decreased significantly in OSCC compared with controls. Relatedly, within the OSCC group, Fas and ADAM10 decreased in accordance with tumor disease stage; in stages I/II, as well as in tumors of smaller diameter (T1-T2),ADAM10 showed higher levels when compared to patients with T3-T4 tumors and in stage III-IV. FasL in the tumor microenvironment and serum FasL showed no significant differences between both groups. Levels of complete FasL and cleaved FasL were positively correlated in controls; this correlation is preserved in patients with tumors in early stages (I-II), but is lost in later stage (IIIIV). The dysregulation of ADAM10, Fas and FasL could be useful indicators of the progression and severity of OSCC. C1 [Zepeda-Nuno, Sergio Jose] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Departamento de Microbiologia y Patologia, Laboratorio de PatologiaGuadalajara, Jalisco, Mexico. [Guerrero-Velazquez, Celia] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Instituto de Investigacion en Odontologia, Departamento de Clinicas Odontologicas IntegralesGuadalajara, Jalisco, Mexico. [Del Toro-Arreola, Susana] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Instituto de Investigacion en Enfermedades Cronico Degenerativas, Departamento de Biologia Molecular y Genomica, Sierra Mojada # 950, Colonia Independencia, 44340 Guadalajara, Jalisco, Mexico. [Vega-Magana, Natali] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Instituto de Investigacion en Enfermedades Cronico Degenerativas, Departamento de Biologia Molecular y Genomica, Sierra Mojada # 950, Colonia Independencia, 44340 Guadalajara, Jalisco, Mexico. [Angeles-Sanchez, Julian] Instituto Jalisciense de Cancerologia, Clinica de Tumores de Cabeza y CuelloGuadalajara, Jalisco, Mexico. [Haramati, Jesse] Centro Universitario de Ciencias Biologicas y Agropecuarias, Universidad de Guadalajara, Departamento de Biologia Celular y Molecular, Laboratorio de InmunologiaGuadalajara, Jalisco, Mexico. [Pereira-Suarez, L Ana] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Departamento de Fisiologia, Laboratorio de InmunologiaGuadalajara, Jalisco, Mexico. [Bueno-Topete, R Miriam] Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Instituto de Investigacion en Enfermedades Cronico Degenerativas, Departamento de Biologia Molecular y Genomica, Sierra Mojada # 950, Colonia Independencia, 44340 Guadalajara, Jalisco, Mexico. RP Bueno-Topete, RM (reprint author), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Instituto de Investigacion en Enfermedades Cronico Degenerativas, Departamento de Biologia Molecular y Genomica, 44340 Guadalajara, Mexico. EM ruthmyriamtop@hotmail.com CR International agency of research on cancer. Cancer mondial globocan, 2012, Database: summary tables by cancer. http://globocan.iarc.fr. Accessed 12 February 2016 Fong D, Spizzo G, Gostner JM, Gastl G, Moser P, Krammel C, Gerhard S, Rasse M, Laimer K, 2008, TROP2: a novel prognostic marker in squamous cell carcinoma of the oral cavity. Mod Pathol 21:186–191 Edwards DR, Handsley MM, Pennington CJ, 2008, The ADAM metalloproteinases. Mol Asp Med 29:258–289 Tousseyn T, Thathiah A, Jorissen E, Raemaekers T, Konietzko U, Reiss K, Maes E, Snellinx A, Serneels L, Nyabi O, Annaert W, Saftig P, Hartmann D, De Strooper B, 2009, ADAM10, the ratelimiting protease of regulated intramembrane proteolysis of notch and other proteins, is processed by ADAMS-9, ADAMS-15, and the gamma-secretase. J Biol Chem 284:11738–11747 Mochizuki S, Okada Y, 2007, ADAMs in cancer cell proliferation and progression. Cancer Sci 98:621–628 Jones A, Lambert D, Speight P, Whawell S, 2013, ADAM 10 is over expressed in oral squamous cell carcinoma and contributes to invasive behavior through a functional association with avb6 integrin. FEBS Lett 587:3529–3534 Ko SY, Lin SC, Wong YK, Liu CJ, Chang KW, Liu TY, 2007, Increase of disintergin metalloprotease 10, ADAM10, expression in oral squamous cell carcinoma. Cancer Lett 245:33–43 Pruessmeyer J, Ludwig A, 2009, The good, the bad and the ugly substrates for ADAM10 and ADAM17 in brain pathology, inflammation and cancer. Semin Cell Dev Biol 20:164–174 Strasser A, Jost PJ, Nagata S, 2009, The many roles of FAS receptor signaling in the immune system. Immunity 30:180–192 Schulte M, Reiss K, Lettau M, Maretzky T, Ludwig A, Hartmann D, de Strooper B, Janssen O, Saftig P, 2007, ADAM10 regulates FasL cell surface expression and modulates FasL-induced cytotoxicity and activation-induced cell death. Cell Death Differ 14:1040– 1049 Kirkin V, Cahuzac N, Guardiola-Serrano F, Huault S, Luckerath K, Friedmann E, Novac N, Wels WS, Martoglio B, Hueber AO, Zornig M, 2007, The Fas ligand intracellular domain is released by ADAM10 and SPPL2a cleavage in T-cells. Cell Death Differ 14: 1678–1687 Peter ME, Hadji A, Murmann AE, Brockway S, Putzbach W, Pattanayak A, Ceppi P, 2015, The role of CD95 and CD95 ligand in cancer. Cell Death Differ 22:549–559 Pietras K, Ostman A, 2010, Hallmarks of cancer: interactions with the tumor stromal. Exp Cell Res 316:1324–1331 GaidaMM, Haag N, Gunther F, Tschaharganeh DF, Schirmacher P, Friess H, Giese NA, Schmidt J, Wente MN, 2010, Expression of a disintegrin and metalloprotease 10 in pancreatic carcinoma. Int J Mol Med 26:281–288 You B, Shan Y, Shi S, Li X, You Y, 2015, Effects of ADAM10 upregulation on progression, migration, and prognosis of nasopharyngeal carcinoma. Cancer Sci 106:1506–1514 Guo J, He L, Yuan P,Wang P, Lu Y, Tong F,Wang Y, Yin Y, Tian J, Sun J, 2012, ADAM10 overexpression in human non-small cell lung cancer correlates with cell migration and invasion through the activation of the Notch1 signaling pathway. Oncol Rep 28: 1709–1718 Shaker M, Yokoyama Y, Mori S, Tsujimoto M, Kawaguchi N, Kiyono T, Nakano T, Matsuura N, 2011, Aberrant expression of disintegrin-metalloprotease proteins in the formation and progression of uterine cervical cancer. Pathobiology 78:149–161 Saftig P, Reiss K, 2011, The "a disintegrin and metalloproteases" ADAM10 and ADAM17: novel drug targets with therapeutic potential? Eur J Cell Biol 90:527–535 Weber S, Niessen MT, Prox J, Lullmann-Rauch R, Schmitz A, Schwanbeck R, Blobel CP, Jorissen E, de Strooper B, Niessen CM, Saftig P, 2011, The disintegrin/metalloproteinase Adam10 is essential for epidermal integrity and Notch-mediated signaling. Development 138:495–505 Ebsen H, Schroder A, Kabelitz D, Janssen O, 2013, Differential surface expression of ADAM10 and ADAM17 on human T lymphocytes and tumor cells. PLoS One 8:e76853 Pabois A, Devalliere J, Quillard T, Coulon F, Gerard N, Laboisse C, Toquet C, Charreau B, 2014, The disintegrin and metalloproteinase ADAM10 mediates a canonical notch-dependent regulation of IL-6 through Dll4 in human endothelial cells. Biochem Pharmacol 91: 510–521 Zhuang J, Wei Q, Lin Z, Zhou C, 2015, Effects of ADAM10 deletion on notch-1 signaling pathway and neuronal maintenance in adult mouse brain. Gene 555:150–158 Scholz F, Schulte A, Adamski F, Hundhausen C,Mittag J, Schwarz A, Kruse ML, Proksch E, Ludwig A, 2007, Constitutive expression and regulated release of the transmembrane chemokine CXCL16 in human and murine skin. J Invest Dermatol 127:1444–1455 Abel S, Hundhausen C, Mentlein R, Schulte A, Berkhout TA, Broadway N, Hartmann D, Sedlacek R, Dietrich S, Muetze B, Schuster B, Kallen KJ, Saftig P, Rose-John S, Ludwig A, 2004, The transmembrane CXC-chemokine ligand 16 is induced by IFNgamma and TNF-alpha and shed by the activity of the disintegrinlike metalloproteinase ADAM10. J Immunol 172:6362–6372 Gregory MS, Hackett CG, Abernathy EF, Lee KS, Saff RR, Hohlbaum AM, Moody KS, Hobson MW, Jones A, Kolovou P, Karray S, Giani A, John SW, Chen DF, Marshak-Rothstein A, Ksander BR, 2011, Opposing roles for membrane bound and soluble Fas ligand in glaucoma-associated retinal ganglion cell death. PLoS One 6:e17659 Wisniewski P, Ellert-Miklaszewska A, Kwiatkowska A, Kaminska B, 2010, Non-apoptotic Fas signaling regulates invasiveness of glioma cells and modulates MMP-2 activity via NFkappaBTIMP- 2 pathway. Cell Signal 22:212–220 Hoffmann TK, Dworacki G, Tsukihiro T,Meidenbauer N, Gooding W, Johnson JT, Whiteside TL, 2002, Spontaneous apoptosis of circulating T lymphocytes in patients with head and neck cancer and its clinical importance. Clin Cancer Res 8:2553–2562 Jablonska E, Kiersnowska-Rogowska B, Rogowski F, Parfienczyk A, PuzewskaW, Bukin M, 2005, Soluble form of TRAIL, Fas and FasL in the serum of patients with B-CLL. Rocz Akad Med Bialymst 50:204–207 Askenasy N, Yolcu ES, Yaniv I, Shirwan H, 2005, Induction of tolerance using Fas ligand: a double-edged immunomodulator. Blood 105:1396–1404 Mogi M, Fukuo K, Yang J, Suhara T, Ogihara T, 2001, Hypoxia stimulates release of the soluble form of fas ligand that inhibits endothelial cell apoptosis. Lab Investig 81:177–184 Fang L, Sun L, FF H, Chen QE, 2013, Effects of FasL expression in oral squamous cell cancer. Asian Pac J Cancer Prev 14:281–285 Das SN, Khare P, Singh MK, Sharma SC, 2011, Fas receptor, CD95, & Fas ligand, CD178, expression in patients with tobacco-related intraoral squamous cell carcinoma. Indian J Med Res 134:54–60 Lee SH, Jang JJ, Lee JY, Kim SY, Park WS, Shin MS, Dong SM, Na EY, Kim KM, Kim CS, Kim SH, Yoo NJ, 1998, Fas ligand is expressed in normal skin and in some cutaneous malignancies. Br J Dermatol 139:186–191 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 345 EP 353 DI 10.1007/s12253-016-0102-5 PG 9 ER PT J AU Moazeni-Roodi, A Allameh, A Harirchi, I Motiee-Langroudi, M Garajei, A AF Moazeni-Roodi, Abdolkarim Allameh, Abdolamir Harirchi, Iraj Motiee-Langroudi, Maziar Garajei, Ata TI Studies on the Contribution of Cox-2 Expression in the Progression of Oral Squamous Cell Carcinoma and H-Ras Activation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE OSCC; Oncogene; H-ras; Cox-2; Smoking; Iran ID OSCC; Oncogene; H-ras; Cox-2; Smoking; Iran AB The aim of this study was to investigate the relationship between the H-ras and Cox-2 gene expression in tumors from Iranian Oral Squamous Cell Carcinoma (OSCC) patients. Fresh tumor biopsies removed from oral cavity were collected from 67 new cases. Total RNA was extracted from biopsies and processed for quantification of H-ras and Cox-2 specific RNA expression using real-time PCR (QPCR). In addition, 59 gingival biopsies from apparently normal individuals were processed for QPCR assays. The results showed that Cox-2 expression at mRNA levels was at minimal levels in normal gingival biopsies. However, there was a surge in Cox-2 expression in tumor tissues (11.5 fold, p < 0.0001). Cox-2 expression was elevated depending on the tumor grade and there was a 1.7 fold increase (p = 0.003) in tumors diagnosed as MD/PD compared to that pathologically diagnosed as WD. This inflammatory marker was increased more significantly in smoker patients compared to non-smoker matching group. The H-ras expression at mRNA levels was significantly higher in OSCC samples compared to normal gingival (3 fold; p = 0.044). This expression was significantly higher in tumors diagnosed as MD/PD compared to WD(1.59 fold, p = 0.033). In conclusion, we found a correlation between H-ras expression and Cox-2 induction in OSCC tissue, suggesting that together these genes are contributing to cancer progression. Cox-2 is an early event in cancers of mucosal epithelial cells and a surge in Cox-2 expression in OSCC could be partly due to proinflammatory factors such as smoking. C1 [Moazeni-Roodi, Abdolkarim] Tarbiat Modares University, Faculty of Medical Sciences, Department of Clinical BiochemistryTehran, I.R., Iran. [Allameh, Abdolamir] Tarbiat Modares University, Faculty of Medical Sciences, Department of Clinical BiochemistryTehran, I.R., Iran. [Harirchi, Iraj] Tehran University of Medical Sciences, Cancer Institute of IranTehran, I.R., Iran. [Motiee-Langroudi, Maziar] Tehran University of Medical Sciences, Cancer Institute of IranTehran, I.R., Iran. [Garajei, Ata] Tehran University of Medical Sciences, Department of Head and Neck Surgical Oncology and Reconstructive Surgery, School of Medicine, Department of Oral and Maxillofacial Surgery, School of DentistryTehran, Iran. RP Allameh, A (reprint author), Tarbiat Modares University, Faculty of Medical Sciences, Department of Clinical Biochemistry, Tehran, Iran. EM allameha@modares.ac.ir CR Li TJ, Cui J, 2013, COX-2, MMP-7 expression in oral lichen planus and oral squamous cell carcinoma. Asian Pac J Trop Med 6(8):640–643., DOI 10.1016/S1995-7645(13)60110-8 Li D, Hao SH, Sun Y, Hu CM, Ma ZH, Wang ZM, Liu J, Liu HB, Ye M, Zhang YF, Yang DS, Shi G, 2015, Functional polymorphisms in COX-2 gene are correlated with the risk of oral cancer. Biomed Res Int 2015:580652., DOI 10.1155/2015/580652 Warnakulasuriya S, 2010, Living with oral cancer: epidemiology with particular reference to prevalence and life-style changes that influence survival. Oral Oncol 46(6):407–410., DOI 10.1016/j. oraloncology.2010.02.015 Yu KT, Ge C, Xu XF, Zou JC, Zou X, Zhen S, 2014, CYP1A1 polymorphism interactions with smoking status in oral cancer risk: evidence from epidemiological studies. Tumour Biol 35(11): 11183–11191., DOI 10.1007/s13277-014-2422-y Sand L,WallstromM, Hirsch JM(2014, Smokeless tobacco, viruses and oral cancer. Oral Health Dent Manag 13(2):372–378 Vlachopoulos C, Aznaouridis K, Bratsas A, Ioakeimidis N, Dima I, Xaplanteris P, Stefanadis C, Tousoulis D, 2015, Arterial stiffening and systemic endothelial activation induced by smoking: the role of COX-1 and COX-2. Int J Cardiol 189:293–298., DOI 10.1016/j. ijcard.2015.04.029 Ricciotti E, FitzGerald GA, 2011, Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol 31(5):986–1000., DOI 10.1161 /ATVBAHA.110.207449 Hamzic N, Blomqvist A, Nilsberth C, 2013, Immune-induced expression of lipocalin-2 in brain endothelial cells: relationship with interleukin-6, cyclooxygenase-2 and the febrile response. J Neuroendocrinol 25(3):271–280., DOI 10.1111/jne.12000 Pandey M, Prakash O, Santhi WS, Soumithran CS, Pillai RM, 2008, Overexpression of COX-2 gene in oral cancer is independent of stage of disease and degree of differentiation. Int J Oral Maxillofac Surg 37(4):379–383., DOI 10.1016/j.ijom.2008.01.004 He XP, Shao Y, Li XL, Xu W, Chen GS, Sun HH, Xu HC, Xu X, Tang D, Zheng XF, Xue YP, Huang GC, Sun WH, 2012, Downregulation of miR-101 in gastric cancer correlates with cyclooxygenase-2 overexpression and tumor growth. FEBS J 279(22):4201–4212., DOI 10.1111/febs.12013 Wu QB, Sun GP, 2015, Expression of COX-2 and HER-2 in colorectal cancer and their correlation. World J Gastroenterol 21(20): 6206–6214., DOI 10.3748/wjg.v21.i20.6206 Allameh A, Rasmi Y, Nasseri-Moghaddam S, TavangarSM, Sharifi R, Sadreddini M, 2009, Immunohistochemical analysis of selected molecular markers in esophagus precancerous, adenocarcinoma and squamous cell carcinoma in Iranian subjects. Cancer Epidemiol 33(1):79–84., DOI 10.1016/j.canep.2009.05.002 Biramijamal F, Allameh A, Mirbod P, Groene HJ, Koomagi R, Hollstein M, 2001, Unusual profile and high prevalence of p53 mutations in esophageal squamous cell carcinomas from northern Iran. Cancer Res 61(7):3119–3123 Liu B, Qu L, Yan S, 2015, Cyclooxygenase-2 promotes tumor growth and suppresses tumor immunity. Cancer Cell Int 15., DOI 10.1186/s12935-015-0260-7 Taketomi A, Shirabe K, Muto J, Yoshiya S, Motomura T, Mano Y, Ikegami T, Yoshizumi T, Sugio K, Maehara Y, 2013, A rare point mutation in the ras oncogene in hepatocellular carcinoma. Surg Today 43(3):289–292., DOI 10.1007/s00595-012-0462-8 Wang Y, Arlt VM, Roufosse CA, McKim KL, Myers MB, Phillips DH, Parsons BL, 2012, ACB-PCRmeasurement of H-ras codon 61 CAA–>CTA mutation provides an early indication of aristolochic acid I carcinogenic effect in tumor target tissues. Environ Mol Mutagen 53(7):495–504., DOI 10.1002/em.21710 Sen B, Peng S, Saigal B, Williams MD, Johnson FM, 2011, Distinct interactions between c-Src and c-met in mediating resistance to c-Src inhibition in head and neck cancer. Clin Cancer Res 17(3):514–524., DOI 10.1158/1078-0432.CCR-10-1617 Murugan AK,Munirajan AK, Tsuchida N, 2012, Ras oncogenes in oral cancer: the past 20 years. Oral Oncol 48(5):383–392., DOI 10.1016/j.oraloncology.2011.12.006 Chang F, Steelman LS, Shelton JG, Lee JT, Navolanic PM, Blalock WL, Franklin R, McCubrey JA, 2003, Regulation of cell cycle progression and apoptosis by the ras/Raf/MEK/ERK pathway, review. Int J Oncol 22(3):469–480 Vairaktaris E, Papakosta V, Derka S, Vassiliou S, Nkenke E, Spyridonidou S, Vylliotis A, Lazaris A, Kokkori A, Moulavassili P, Loukeri S, Perrea D, Donta I, Yapijakis C, Patsouris E, 2008, Hras and c-fos exhibit similar expression patterns during most stages of oral oncogenesis. In Vivo 22(5):621–628 Sathyan KM, Nalinakumari KR, Kannan S, 2007, H-ras mutation modulates the expression ofmajor cell cycle regulatory proteins and disease prognosis in oral carcinoma. Mod Pathol 20(11):1141– 1148., DOI 10.1038/modpathol.3800948 Wong CE, Yu JS, Quigley DA, To MD, Jen KY, Huang PY, Del Rosario R, Balmain A, 2013, Inflammation and Hras signaling control epithelial-mesenchymal transition during skin tumor progression. Genes Dev 27(6):670–682., DOI 10.1101/gad.210427.112 Lee KW, Kim MS, Kang NJ, Kim DH, Surh YJ, Lee HJ, Moon A, 2006, H-ras selectively up-regulates MMP-9 and COX-2 through activation of ERK1/2 and NF-kappaB: an implication for invasive phenotype in rat liver epithelial cells. Int J Cancer 119(8):1767– 1775., DOI 10.1002/ijc.22056 Lingen MW, Kalmar JR, Karrison T, Speight PM, 2008, Critical evaluation of diagnostic aids for the detection of oral cancer. Oral Oncol 44(1):10–22., DOI 10.1016/j.oraloncology.2007.06.011 Nguyen T, Tang Z, Younes M, Alsarraj A, Ramsey D, Fitzgerald S, Kramer JR, El-Serag HB, 2015, Esophageal COX-2 expression is increased in Barrett’s esophagus, obesity, and smoking. Dig Dis Sci 60(1):65–73., DOI 10.1007/s10620-014-3333-x Fujii R, Imanishi Y, Shibata K, Sakai N, Sakamoto K, Shigetomi S, Habu N, Otsuka K, Sato Y, Watanabe Y, Ozawa H, Tomita T, Kameyama K, Fujii M, Ogawa K, 2014, Restoration of Ecadherin expression by selective cox-2 inhibition and the clinical relevance of the epithelial-to-mesenchymal transition in head and neck squamous cell carcinoma. J Exp Clin Cancer Res 33., DOI 10.1186/1756-9966-33-40 Liu ES, Shin VY, Ye YN, Luo JC, WK W, Cho CH, 2005, Cyclooxygenase-2 in cancer cells and macrophages induces colon cancer cell growth by cigarette smoke extract. Eur J Pharmacol 518(1):47–55., DOI 10.1016/j.ejphar.2005.05.018 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 355 EP 360 DI 10.1007/s12253-016-0114-1 PG 6 ER PT J AU Liu, P Jiang, W Zhou, Sh Gao, J Zhang, H AF Liu, Pengfei Jiang, Wenhua Zhou, Shiyong Gao, Jun Zhang, Huilai TI Combined Analysis of ChIP Sequencing and Gene Expression Dataset in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Chromatin Immunoprecipitation combined with high-throughput sequencing (ChIP-seq); Human protein reference database (HPRD); Protein-protein interaction (PPI) ID Breast cancer; Chromatin Immunoprecipitation combined with high-throughput sequencing (ChIP-seq); Human protein reference database (HPRD); Protein-protein interaction (PPI) AB Breast cancer is a common malignancy in women and contribute largely to the cancer related death. The purpose of this study is to confirm the roles of GATA3 and identify potential biomarkers of breast cancer. Chromatin Immunoprecipitation combined with high-throughput sequencing (ChIP-Seq) (GSM1642515) and gene expression profiles (GSE24249) were downloaded from the Gene Expression Omnibus (GEO) database. Bowtie2 and MACS2 were used for the mapping and peak calling of the ChIP-Seq data respectively. ChIPseeker, a R bioconductor package was adopted for the annotation of the enriched peaks. For the gene expression profiles, we used affy and limma package to do normalization and differential expression analysis. The genes with fold change >2 and adjusted P-Value <0.05 were screened out. Besides, BETA (Binding and Expression Target Analysis) was used to do the combined analysis of ChIP-Seq and gene expression profiles. The Database for Annotation, Visualization and Integrated Discovery (DAVID) was used for the functional enrichment analysis of overlapping genes between the target genes and differential expression genes (DEGs). What’s more, the protein-protein interaction (PPI) network of the overlapping genes was obtained through the Human Protein Reference Database (HPRD). A total of 46,487 peaks were identified for GATA3 and out of which, 3256 ones were found to located at −3000 ~ 0 bp from the transcription start sites (TSS) of their nearby gene. A total of 236 down- and 343 up-regulated genes were screened out in GATA3 overexpression breast cancer samples compared with those in control. The combined analysis of ChIP-Seq and gene expression dataset showed GATA3 act as a repressor in breast cancer. Besides, 68 overlaps were obtained between the DEGs and genes included in peaks located at −3000 ~ 0 bp from TSS. Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways related to cancer progression and gene regulation were found to be enriched in those overlaps. In the PPI network, NDRG1, JUP and etc. were found to directly interact with large number of genes, which might indicate their important roles in the progression of breast cancer. C1 [Liu, Pengfei] National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Department of Lymphoma, Sino-US Center of Lymphoma and Leukemia, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, 300060 Tianjin, China. [Jiang, Wenhua] The Second Hospital of Tianjin Medical University, Department of Radiotherapy, 300211 Tianjin, China. [Zhou, Shiyong] National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Department of Lymphoma, Sino-US Center of Lymphoma and Leukemia, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, 300060 Tianjin, China. [Gao, Jun] Hengshui Harrison International Peace Hospital, Department of Oncology, 053000 Hebei, China. [Zhang, Huilai] National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Department of Lymphoma, Sino-US Center of Lymphoma and Leukemia, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, 300060 Tianjin, China. RP Zhang, H (reprint author), National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Department of Lymphoma, Sino-US Center of Lymphoma and Leukemia, 300060 Tianjin, China. EM snowlpf@163.com CR GradisharWJ,Anderson BO, Balassanian R, Blair SL, Burstein HJ, Cyr A, Elias AD, Farrar WB, Forero A, Giordano SH, Goetz M, Goldstein LJ, Hudis CA, Isakoff SJ, Marcom PK, Mayer IA, McCormick B, Moran M, Patel SA, Pierce LJ, Reed EC, Salerno KE, Schwartzberg LS, Smith KL, SmithML, Soliman H, Somlo G, Telli M, Ward JH, Shead DA, Kumar R, 2016, Invasive breast cancer version 1.2016, NCCN clinical practice guidelines in oncology. Journal of the National Comprehensive Cancer Network. JNCCN 14(3):324–354 Pusztai L, Ladanyi A, Szekely B, Dank M, 2016, Immunotherapy opportunities in breast cancer. Magyar onkologia 60(1):34–40 Liu D, 2016, Tumor necrosis factor-alpha, a regulator and therapeutic agent on breast cancer. Curr Pharm Biotechnol 17(6): 486–494 Maximiano S, Magalhaes P, Guerreiro MP, Morgado M, 2016, Trastuzumab in the Treatment of Breast Cancer. BioDrugs: clinical immunotherapeutics, biopharmaceuticals and gene therapy, DOI 10.1007/s40259–016–0162-9 Kober KM, Dunn L,Mastick J, Cooper B, Langford D,Melisko M, Venook A, ChenLM,Wright F, HammerM, Schmidt BL, Levine J, Miaskowski C, Aouizerat BE, 2016, Gene Expression Profiling of Evening Fatigue in Women Undergoing Chemotherapy for Breast Cancer. Biological research for nursing, DOI 10.1177/1099800416629209 Cao ZG, Qin XB, Liu FF, Zhou LL, 2015, Tryptophan-induced pathogenesis of breast cancer. Afr Health Sci 15(3):982–985., DOI 10.4314/ahs.v15i3.36 Park E, 2016, Data on cell cycle in breast cancer cell line, MDAMB- 231 with ferulic acid treatment. Data in brief 7:107–110., DOI 10.1016/j.dib.2016.02.001 Asch-Kendrick R, Cimino-Mathews A, 2016, The role of GATA3 in breast carcinomas: a review. Hum Pathol 48:37–47., DOI 10.1016 /j.humpath.2015.09.035 SiW, HuangW, ZhengY, YangY, Liu X, Shan L, Zhou X,Wang Y, Su D, Gao J, Yan R, Han X, Li W, He L, Shi L, Xuan C, Liang J, Sun L, Wang Y, Shang Y, 2015, Dysfunction of the Reciprocal Feedback Loop between GATA3- and ZEB2-Nucleated Repression Programs Contributes to Breast Cancer Metastasis. Cancer Cell 27(6):822–836., DOI 10.1016/j.ccell.2015.04.011 McCleskey BC, Penedo TL, Zhang K, Hameed O, Siegal GP,Wei S, 2015, GATA3 expression in advanced breast cancer: prognostic value and organ-specific relapse. Am J Clin Pathol 144(5):756– 763., DOI 10.1309/AJCP5MMR1FJVVTPK Dieci MV, Smutna V, Scott V, Yin G, Xu R, Vielh P, Mathieu MC, Vicier C, Laporte M, Drusch F, Guarneri V, Conte P, Delaloge S, Lacroix L, Fromigue O, Andre F, Lefebvre C, 2016)Whole exome sequencing of rare aggressive breast cancer histologies. Breast Cancer Res Treat 156(1):21–32., DOI 10.1007/s10549–016-3718-y Chu IM, Michalowski AM, Hoenerhoff M, Szauter KM, Luger D, SatoM, Flanders K, Oshima A, Csiszar K, Green JE, 2012, GATA3 inhibits lysyl oxidase-mediated metastases of human basal triplenegative breast cancer cells. Oncogene 31(16):2017–2027., DOI 10.1038/onc.2011.382 Gautier L, Cope L, Bolstad BM, Irizarry RA, 2004, Affy–analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 20(3):307–315., DOI 10.1093/bioinformatics/btg405 Langmead B, 2010, Aligning short sequencing reads with Bowtie. Current protocols in bioinformatics / editoral board, Andreas D Baxevanis [et al] Chapter 11:Unit 11 17., DOI 10.1002 /0471250953.bi1107s32 Feng J, Liu T, Qin B, Zhang Y, Liu XS, 2012, Identifying ChIP-seq enrichment usingMACS. Nat Protoc 7(9):1728–1740., DOI 10.1038 /nprot.2012.101 Yu G, Wang LG, He QY, 2015, ChIPseeker: an R/bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31(14):2382–2383., DOI 10.1093 /bioinformatics/btv145 Wang S, Sun H, Ma J, Zang C, Wang C, Wang J, Tang Q, Meyer CA, Zhang Y, Liu XS, 2013, Target analysis by integration of transcriptome and ChIP-seq data with BETA. Nat Protoc 8(12): 2502–2515., DOI 10.1038/nprot.2013.150 Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA, 2003, DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol 4(5):P3 Keshava Prasad TS, Goel R, Kandasamy K, Keerthikumar S, Kumar S, Mathivanan S, Telikicherla D, Raju R, Shafreen B, Venugopal A, Balakrishnan L, Marimuthu A, Banerjee S, Somanathan DS, Sebastian A, Rani S, Ray S, Harrys Kishore CJ, Kanth S, Ahmed M, Kashyap MK, Mohmood R, Ramachandra YL, Krishna V, Rahiman BA, Mohan S, Ranganathan P, Ramabadran S, Chaerkady R, Pandey A, 2009, Human Protein Reference Database–2009 update. Nucleic Acids Res 37(Database issue): D767–D772., DOI 10.1093/nar/gkn892 Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T, 2003, Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13(11):2498–2504., DOI 10.1101/gr.1239303 Lin Y, Zhao J, Hu X, Wang L, Liang L, Chen W, 2016, Transcription factor CCAAT/enhancer binding protein alpha upregulates microRNA let-7a-1 in lung cancer cells by direct binding. Cancer Cell Int 16:17., DOI 10.1186/s12935–016–0294-5 Li C, Deng L, Zhi Q,Meng Q, Qian A, Sang H, Li X, Xia J, 2016, MicroRNA-183 Functions As an Oncogene by Regulating PDCD4 in Gastric Cancer. Anti Cancer Agents Med Chem 16(4):447–455 Mitxelena J, Apraiz A, Vallejo-Rodriguez J, Malumbres M, Zubiaga AM, 2016, E2F7 regulates transcription and maturation of multiple microRNAs to restrain cell proliferation. Nucleic Acids Res., DOI 10.1093/nar/gkw146 Takaku M, Grimm SA,Wade PA, 2015, GATA3 in Breast Cancer: Tumor Suppressor or Oncogene? Gene Expr 16(4):163–168., DOI 10.3727/105221615X14399878166113 Tkocz D, Crawford NT, Buckley NE, Berry FB, Kennedy RD, Gorski JJ, Harkin DP, Mullan PB, 2012, BRCA1 and GATA3 corepress FOXC1 to inhibit the pathogenesis of basal- like breast cancers. Oncogene 31(32):3667–3678., DOI 10.1038 /onc.2011.531 Xiong Y, Castro E, Yagi R, Zhu J, Lesourne R, Love PE, Feigenbaum L, Bosselut R, 2013, Thpok-independent repression of Runx3 by Gata3 during CD4+ T-cell differentiation in the thymus. Eur J Immunol 43(4):918–928., DOI 10.1002/eji.201242944 Liu W, Yue F, Zheng M, Merlot A, Bae DH, Huang M, Lane D, Jansson P, Lui GY, Richardson V, Sahni S, Kalinowski D, Kovacevic Z, Richardson DR, 2015, The proto-oncogene c-Src and its downstream signaling pathways are inhibited by the metastasis suppressor, NDRG1. Oncotarget 6(11): 8851–8874., DOI 10.18632/oncotarget.3316 Salis O, Bedir A, Kilinc V, Alacam H, Gulten S, Okuyucu A, 2014, The anticancer effects of desferrioxamine on human breast adenocarcinoma and hepatocellular carcinoma cells. Cancer biomarkers: section A of Disease markers 14(6):419–426., DOI 10.3233/CBM-140422 Song JY, Lee JK, Lee NW, Jung HH, Kim SH, Lee KW, 2008, Microarray analysis of normal cervix, carcinoma in situ, and invasive cervical cancer: identification of candidate genes in pathogenesis of invasion in cervical cancer. International journal of gynecological cancer: official journal of the international gynecological cancer. Society 18(5):1051–1059., DOI 10.1111 /j.1525-1438.2007.01164.x Wang Y, Zheng T, 2014, Screening of hub genes and pathways in colorectal cancer with microarray technology. Pathol Oncol Res: POR 20(3):611–618., DOI 10.1007/s12253–013–9739-5 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 361 EP 368 DI 10.1007/s12253-016-0116-z PG 8 ER PT J AU Peng, G Liao, Y Shen, Ch AF Peng, Gang Liao, Yiwei Shen, Chenfu TI miRNA-429 Inhibits Astrocytoma Proliferation and Invasion by Targeting BMI1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma; miRNA-429; BMI1; Proliferation; Invasion ID Glioblastoma; miRNA-429; BMI1; Proliferation; Invasion AB Glioblastoma multiforme (GBM), the most common primary brain cancer in adults, is usually the most lethal type of brain tumor. MicroRNAs (miRNAs) are a class of small, non-coding RNA molecules that deeply involves with the regulation of gene expression and cellular processes, including proliferation, apoptosis, migration and invasion. The objective of the study is to investigate the effect of miRNA- 429 on human glioblastoma tissues and cell lines. miRNA- 429 expressions in human glioblastoma, normal brain tissue samples, and human malignant glioma cell lines (U87, U251 and LN229) were compared using reverse transcriptionquantitative PCR and western blot methods. U251 cell lines were transfected with miRNA-429 mimics, and then the effects of miRNA-429 on cell proliferation and invasion were investigated by CCK8 and Transwell invasion assay, respectively. It was found that miRNA-429 expression was significantly reduced in the examined Glioblastoma samples and human glioma cell lines. Overexpression of miRNA-429 inhibited Glioblastoma cell proliferation and invasion. Additionally, the present study also showed that BMI1 was a functional target of miRNA-429. Overexpression of BMI1 undermined the inhibition effect of miRNA-429 in glioblastoma and U251 cell lines. The current study demonstrated that miRNA-429, as a tumor suppressor gene, was capable of negatively regulating the expression of BMI1 in U251 cells. C1 [Peng, Gang] Central South University, Xiangya Hospital, Department of Neurosurgery, 87 XiangYa Road, 410008 Changsha, Hunan Province, China. [Liao, Yiwei] Central South University, Xiangya Hospital, Department of Neurosurgery, 87 XiangYa Road, 410008 Changsha, Hunan Province, China. [Shen, Chenfu] Central South University, Xiangya Hospital, Department of Neurosurgery, 87 XiangYa Road, 410008 Changsha, Hunan Province, China. RP Shen, Ch (reprint author), Central South University, Xiangya Hospital, Department of Neurosurgery, 410008 Changsha, China. EM shenchenfu@yahoo.com CR Wen PY, Kesari S, 2008, Malignant gliomas in adults. N Engl J Med 359(5):492–507., DOI 10.1056/NEJMra0708126 Johnson DR, O’Neill BP, 2012, Glioblastoma survival in the United States before and during the temozolomide era. J Neuro- Oncol 107(2):359–364., DOI 10.1007/s11060-011-0749-4 Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al., 2005, MicroRNA expression profiles classify human cancers. Nature 435(7043):834–838., DOI 10.1038/nature03702 Croce CM, 2009, Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet 10(10):704–714., DOI 10.1038/nrg2634 Cho WC, 2010, MicroRNAs in cancer - from research to therapy. Biochim Biophys Acta 1805(2):209–217., DOI 10.1016/j. bbcan.2009.11.003 Peng G, Yuan X, Yuan J, Liu Q, Dai M, Shen C, et al., 2015 Nov, miR-25 promotes glioblastoma cell proliferation and invasion by directly targeting NEFL. Mol Cell Biochem 409(1–2):103–111., DOI 10.1007/s11010-015-2516-x Epub 2015 Jul 26 Chan JA, Krichevsky AM, Kosik KS, 2005, MicroRNA-21 Is an Antiapoptotic Factor in Human Glioblastoma Cells. Cancer Res 65: 6029., DOI 10.1158/0008–5472.CAN-05-0137 Coon SW, Savera AT, Zarbo RJ, Benninger MS, Chase GA, Rybicki BA, et al., 2004, Prognostic implications of loss of heterozygosity at 8p21 and 9p21 in head and neck squamous cell carcinoma. Int J Cancer 111(2):206–212., DOI 10.1002/ijc.20254 Tang J, Li L, HuangW, Sui C,YangY, Lin X, et al., 2015, MiRNA- 429 increases the metastatic capability of HCC via regulating classic Wnt pathway rather than epithelial-mesenchymal transition. Cancer Lett 364(1):33–43., DOI 10.1016/j.canlet.2015.04.023 Yoneyama K, Ishibashi O, Kawase R, Kurose K, Takeshita T, 2015, miR-200a, miR-200b and miRNA-429 are onco-miRs that target the PTEN gene in endometrioid endometrial carcinoma. Anticancer Res 35(3):1401–1410 Zhang M, Dong BB, Lu M, Zheng MJ, Chen H, Ding JZ, et al.. miRNA-429 functions as a tumor suppressor by targeting FSCN1 in gastric cancer cells. Onco Targets Ther 2016;9:1123–1133., DOI 10.2147/OTT.S91879. eCollection 2016. PMID: 27042104 Lei W, Liu YE, Zheng Y, Qu L, 2015, MiRNA-429 inhibits oral squamous cell carcinoma growth by targeting ZEB1. Med Sci Monit 21:383–389., DOI 10.12659/MSM.893412 Zang W, Wang Y, Du Y, Xuan X, Wang T, Li M, et al., 2014, Differential expression profiling of microRNAs and their potential involvement in esophageal squamous cell carcinoma. Tumour Biol 35(4):3295–3304., DOI 10.1007/s13277-013-1432-5 Iorio MV, Croce CM, 2012, MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med 4(3):143–159., DOI 10.1002/emmm.201100209 Sana J, Hajduch M, Michalek J, Vyzula R, Slaby O, 2011, MicroRNAs and glioblastoma: roles in core signalling pathways and potential clinical implications. J Cell Mol Med 15(8):1636– 1644., DOI 10.1111/j.1582-4934.2011.01317.x Macoska JA, Trybus TM, Benson PD, Sakr WA, Grignon DJ, Wojno KD, et al., 1995, Evidence for three tumor suppressor gene loci on chromosome 8p in human prostate cancer. Cancer Res 55(22):5390–5395 Shenouda SK, Alahari SK, 2009, MicroRNA function in cancer: oncogene or a tumor suppressor? Cancer Metastasis Rev 28(3–4): 369–378., DOI 10.1007/s10555-009-9188-5 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116(2):281–297 Amaral JD, Xavier JM, Steer CJ, Rodrigues CM, 2010, Targeting the p53 pathway of apoptosis. Curr Pharm Des 16:2493–2503 Garg M, 2012, MicroRNAs, stem cells and cancer stem cells. World J Stem Cells 4:62–70 Qiu M, Liang Z, Chen L, Tan G, Wang K, Liu L, et al., 2015, MicroRNA-429 suppresses cell proliferation, epithelialmesenchymal transition, and metastasis by direct targeting of BMI1 and E2F3 in renal cell carcinoma. Urol Oncol. 33(7): 332.e9–332.18., DOI 10.1016/ j.urolonc. 2015. 03.016 Cristobal I, Rincon R, Manso R, Carames C, Aguilera O, Madoz- Gurpide J, et al., 2014, Deregulation of miR-200b, miR-200c and miRNA-429 indicates its potential relevant role in patients with colorectal cancer liver metastasis. J Surg Oncol 110(4):484–485., DOI 10.1002/jso.23661 Ye ZB, Ma G, Zhao YH, Xiao Y, Zhan Y, Jing C, et al., 2015, miRNA-429 inhibits migration and invasion of breast cancer cells in vitro. Int J Oncol 46(2):531–538., DOI 10.3892/ijo.2014.2759 Tian X,Wei Z,Wang J, Liu P, Qin Y, ZhongM(2015, MicroRNA- 429 inhibits the migration and invasion of colon cancer cells by targeting PAK6/cofilin signaling. Oncol Rep 34(2):707–714., DOI 10.3892/or.2015.4039 Zhu P, Zhang J, Zhu J, Shi J, Zhu Q, Gao Y, 2015, MiRNA-429 induces gastric carcinoma cell apoptosis through Bcl-2. Cell Physiol Biochem 37(4):1572–1580., DOI 10.1159/000 438524 Wang Y, Li M, ZangW, Ma Y,Wang N, Li P, et al., 2013, MiRNA- 429 up-regulation induces apoptosis and suppresses invasion by targeting Bcl-2 and SP-1 in esophageal carcinoma. Cell Oncol, Dordr, 36(5):385–394., DOI 10.1007/s13402-013-0144-6 Liu X, Liu Y, Wu S, Shi X, Li L, Zhao J, et al., 2014, Tumorsuppressing effects of miRNA-429 on human osteosarcoma. Cell Biochem Biophys 70(1):215–224., DOI 10.1007/s12013-014-9885-8 Ouyang Y, Gao P, Zhu B, Chen X, Lin F, Wang X, et al., 2015, Downregulation of microRNA-429 inhibits cell proliferation by targeting p27Kip1 in human prostate cancer cells. Mol Med Rep 11(2):1435–1441., DOI 10.3892/mmr.2014.2782 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 369 EP 376 DI 10.1007/s12253-016-0113-2 PG 8 ER PT J AU Madaras, B Horvath, Zs Graf, L Galffy, G Tamasi, L Ostoros, Gy Dome, B Morocz, Bartfai, Z Prohaszka, Z Kocsis, J AF Madaras, Balazs Horvath, Zsolt Graf, Laszlo Galffy, Gabriella Tamasi, Lilla Ostoros, Gyula Dome, Balazs Morocz, Eva Bartfai, Zoltan Prohaszka, Zoltan Kocsis, Judit TI Serum Heat Shock Protein 70, as a Potential Biomarker for Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Small cell lung cancer; Hsp70; Biomarker; Prognostic marker; Target ID Small cell lung cancer; Hsp70; Biomarker; Prognostic marker; Target AB The 70 kDa heat shock protein (Hsp70) is a highly conservative molecular chaperone, that has important role in cell integrity. Recently considerable amount of data are accumulating on the potential role of Hsp70 in carcinogenesis and tumor progression. Most papers are focusing on intracellular or membrane bound protein, however very limited data exist on serum Hsp70, that can also induce innate and adaptive immune response. Previously we have published data on the correlation between coloretal cancer progression and serum Hsp70 concentration. The objective of this study was to compare the serum Hsp70 level in patients with small cell lung cancer (SCLC n = 70) and age matched healthy controlls (n = 121) and correlate Hsp70 level with other known serum biomarkers (LDH and NSE) of the disease.We found that the serum level of Hsp70 was significantly higher in SCLC patients compared to control subjects (mean value 6.91 vs 2.47 ng/ml, p = 0.001). The highest Hsp70 concentration was measured in stage IV advanced SCLC (Stage IV versus Stage I-III disease: 9.91 vs 4.38 ng/ml, p = 0.003). The serum Hsp70 level correlated with serum LDH (r = 0.426, p < 0,001) and NSE level (r = 0.455, p < 0,001). We found that high serum Hsp70 level predicted unfavorable survival, risk of death within 1 year was more than 3 times higher in patients with high baseline Hsp70 level (HR:3.509, CI: 1.066–11.562; p = 0.039). Our observations indicate that serum Hsp70 could be a valuable diagnostic and prognostic marker in small cell lung cancer. C1 [Madaras, Balazs] National Institute of OncologyBudapest, Hungary. [Horvath, Zsolt] University of Debrecen, Department of OncologyDebrecen, Hungary. [Graf, Laszlo] Semmelweis University, 3rd Department of Internal MedicineBudapest, Hungary. [Galffy, Gabriella] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Tamasi, Lilla] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Ostoros, Gyula] National Koranyi Institute of Pulmonology, Department of Tumor BiologyBudapest, Hungary. [Dome, Balazs] National Koranyi Institute of Pulmonology, Department of Tumor BiologyBudapest, Hungary. [Morocz, Eva] County Hospital of PulmonologyTorokbalint, Hungary. [Bartfai, Zoltan] Soproni Erzsebet Oktato Korhaz es Rehabilitacios IntezetSopron, Hungary. [Prohaszka, Zoltan] Semmelweis University, 3rd Department of Internal MedicineBudapest, Hungary. [Kocsis, Judit] University of Debrecen, Department of OncologyDebrecen, Hungary. RP Kocsis, J (reprint author), University of Debrecen, Department of Oncology, Debrecen, Hungary. EM kocsis.judit@med.unideb.hu;kocsisjucidr@gmail.com CR Srivastava PK, Menoret A, Basu S, Binder RJ, McQuade KL, 1998, Heat shock proteins come of age: primitive functions acquire new roles in an adaptive world. Immunity 8(6):657–665 Rerole AL, Jego G, Garrido C, 2011, Hsp70: anti-apoptotic and tumorigenic protein. Methods Mol Biol 787:205–230., DOI 10.1007 /978-1-61779-295-3_16 Nylandsted J, Rohde M, Brand K, Bastholm L, Elling F, JaattelaM, 2000, Selective depletion of heat shock protein 70, Hsp70, activates a tumor-specific death program that is independent of caspases and bypasses Bcl-2. Proc Natl Acad Sci U S A 97(14): 7871–7876 Srivastava P, 2002, Roles of heat-shock proteins in innate and adaptive immunity. Nature reviews. Immunology 2(3):185–194., DOI 10.1038/nri749 Nicchitta CV, 2003, Re-evaluating the role of heat-shock proteinpeptide interactions in tumour immunity. Nature reviews. Immunology 3(5):427–432., DOI 10.1038/nri1089 Mosser DD, Morimoto RI, 2004, Molecular chaperones and the stress of oncogenesis. Oncogene 23(16):2907–2918., DOI 10.1038 /sj.onc.1207529 Whitesell L, Lindquist S, 2009, Inhibiting the transcription factor HSF1 as an anticancer strategy. Expert Opin Ther Targets 13(4): 469–478., DOI 10.1517/14728220902832697 Powers MV, Workman P, 2007, Inhibitors of the heat shock response: biology and pharmacology. FEBS Lett 581(19):3758– 3769., DOI 10.1016/j.febslet.2007.05.040 Calderwood SK, Khaleque MA, Sawyer DB, Ciocca DR, 2006, Heat shock proteins in cancer: chaperones of tumorigenesis. Trends Biochem Sci 31(3):164–172., DOI 10.1016/j. tibs.2006.01.006 Buzzard KA, Giaccia AJ, Killender M, Anderson RL, 1998, Heat shock protein 72modulates pathways of stress-induced apoptosis. J Biol Chem 273(27):17147–17153 Garrido CGS, Ravagnan L, Kroemer G, 2001, Heat shock proteins: endogenous modulators of apoptotic cell death. Biochem Biophys Res Commun 286(3):433–442 Ciocca DR, Arrigo AP, Calderwood SK, 2013, Heat shock proteins and heat shock factor 1 in carcinogenesis and tumor development: an update. Arch Toxicol 87(1):19–48., DOI 10.1007/s00204-012-0918-z Ciocca DR, Clark GM, Tandon AK, Fuqua SA, Welch WJ, McGuire WL, 1993, Heat shock protein hsp70 in patients with axillary lymph node-negative breast cancer: prognostic implications. J Natl Cancer Inst 85(7):570–574 Thanner F, Sutterlin MW, Kapp M, Rieger L, Kristen P, Dietl J, Gassel AM, Muller T, 2003, Heat-shock protein 70 as a prognostic marker in node-negative breast cancer. Anticancer Res 23(2 A): 1057–1062 Liu FF, Miller N, Levin W, Zanke B, Cooper B, Henry M, Sherar MD, Pintilie M, Hunt JW, Hill RP, 1996, The potential role of HSP70 as an indicator of response to radiation and hyperthermia treatments for recurrent breast cancer. Int J Hyperth 12(2):197–208 discussion 209–110 Vargas-Roig LM, Gago FE, Tello O, Aznar JC, Ciocca DR, 1998, Heat shock protein expression and drug resistance in breast cancer patients treated with induction chemotherapy. Int J Cancer 79(5): 468–475 Nanbu K, Konishi I,Mandai M, Kuroda H, Hamid AA, Komatsu T, Mori T, 1998, Prognostic significance of heat shock proteins HSP70 and HSP90 in endometrial carcinomas. Cancer Detect Prev 22(6):549–555 Piura B, Rabinovich A, Yavelsky V,Wolfson M, 2002, Heat shock proteins and malignancies of the female genital tract. Harefuah 141(11):969–972 Syrigos KN, Harrington KJ, Karayiannakis AJ, Sekara E, Chatziyianni E, Syrigou EI,Waxman J, 2003, Clinical significance of heat shock protein-70 expression in bladder cancer. Urology 61(3):677–680 Kumar S, Stokes J, 3rd, Singh UP, Scissum Gunn K, Acharya A, Manne U, MishraM(2016, Targeting Hsp70: a possible therapy for cancer. Cancer Let t 374(1):156–166., DOI 10.1016/j. canlet.2016.01.056 Multhoff G, Pockley AG, Schmid TE, Schilling D, 2015, The role of heat shock protein 70, Hsp70, in radiation-induced immunomodulation. Cancer Lett 368(2):179–184., DOI 10.1016/j. canlet.2015.02.013 Specht HM, Ahrens N, Blankenstein C, Duell T, Fietkau R, Gaipl US, Gunther C, Gunther S, Habl G, Hautmann H, Hautmann M, Huber RM, Molls M, Offner R, Rodel C, Rodel F, Schutz M, Combs SE, Multhoff G, 2015, Heat shock Protein 70, Hsp70, Peptide Activated Natural Killer, NK, Cells for the Treatment of Patients with Non-Small Cell Lung Cancer, NSCLC, after Radiochemotherapy, RCTx, - From Preclinical Studies to a Clinical Phase II Trial. Front Immunol 6:162., DOI 10.3389 /fimmu.2015.00162 Kocsis J, Madaras B, Toth EK, Fust G, Prohaszka Z, 2010, Serum level of soluble 70-kD heat shock protein is associated with high mortality in patients with colorectal cancer without distant metastasis. Cell Stress Chaperones 15(2):143–151., DOI 10.1007/s12192- 009-0128-7 Yeh CH, Tseng R, Zhang Z, Cortes J, O'Brien S, Giles F, Hannah A, Estrov Z, Keating M, Kantarjian H, Albitar M, 2009, Circulating heat shock protein 70 and progression in patients with chronic myeloid leukemia. Leuk Res 33(2):212–217., DOI 10.1016/j. leukres.2008.07.012 Gehrmann M, Specht HM, Bayer C, Brandstetter M, Chizzali B, Duma M, Breuninger S, Hube K, Lehnerer S, van Phi V, Sage E, Schmid TE, Sedelmayr M, Schilling D, Sievert W, Stangl S, Multhoff G, 2014, Hsp70–a biomarker for tumor detection and monitoring of outcome of radiation therapy in patients with squamous cell carcinoma of the head and neck. Radiat Oncol 9:131., DOI 10.1186/1748-717X-9-131 Kocsis J, Meszaros T, Madaras B, Toth EK, Kamondi S, Gal P, Varga L, Prohaszka Z, Fust G, 2011, High levels of acute phase proteins and soluble 70 kDa heat shock proteins are independent and additive risk factors for mortality in colorectal cancer. Cell Stress Chaperones 16(1):49–55., DOI 10.1007 /s12192-010-0220-z Dutta SK, Girotra M, Singla M, Dutta A, Otis Stephen F, Nair PP, Merchant NB, 2012, Serum HSP70: a novel biomarker for early detection of pancreatic cancer. Pancreas 41(4):530–534., DOI 10.1097/MPA.0b013e3182374ace Sato Y, Harada K, Sasaki M, Yasaka T, Nakanuma Y, 2012, Heat shock proteins 27 and 70 are potential biliary markers for the detection of cholangiocarcinoma. Am J Pathol 180(1):123–130., DOI 10.1016/j.ajpath.2011.09.010 Zimmermann M, Nickl S, Lambers C, Hacker S, Mitterbauer A, Hoetzenecker K, Rozsas A, Ostoros G, Laszlo V, Hofbauer H, Renyi-Vamos F, Klepetko W, Dome B, Ankersmit HJ, 2012, Discrimination of clinical stages in non-small cell lung cancer patients by serum HSP27 and HSP70: a multi-institutional case-control study. Clinica Chimica Acta; Int J Clin Chem 413(13–14): 1115–1120., DOI 10.1016/j.cca.2012.03.008 Qiao Y, Liu B, Li Z, 2008, Activation of NK cells by extracellular heat shock protein 70 through induction of NKG2D ligands on dendritic cells. Cancer Immun 8:12 Varano Della Vergiliana JF, Lansley SM, Porcel JM, Bielsa S, Brown JS, Creaney J, Temple SE, Waterer GW, Lee YC, 2013, Bacterial infection elicits heat shock protein 72 release from pleural mesothelial cells. PLoS One 8(5):e63873., DOI 10.1371/journal. pone.0063873 Brueckl WM, Herbst L, Lechler A, Fuchs F, Schoeberl A, Zirlik S, Klein P, Brunner TB, Papadopoulos T, Hohenberger W, Hahn EG, Wiest GH, 2006, Predictive and prognostic factors in small cell lung carcinoma, SCLC)–analysis from routine clinical practice. Anticancer Res 26(6C):4825–4832 Byhardt RW, Hartz A, Libnoch JA, Hansen R, Cox JD, 1986, Prognostic influence of TNM staging and LDH levels in small cell carcinoma of the lung, SCCL. Int J Radiat Oncol Biol Phys 12(5): 771–777 Sagman U, Feld R, Evans WK, Warr D, Shepherd FA, Payne D, Pringle J, Yeoh J, DeBoer G, Malkin A, 1991, The prognostic significance of pretreatment serum lactate dehydrogenase in patients with small-cell lung cancer. J Clin Oncol 9(6):954–961 Ganz PA, Ma PY, Wang HJ, Elashoff RM, 1987, Evaluation of three biochemical markers for serially monitoring the therapy of small-cell lung cancer. J Clin Oncol 5(3):472–479 Bremnes RM, Sundstrom S, Aasebo U, Kaasa S, Hatlevoll R, Aamdal S, Group NLCS, 2003, The value of prognostic factors in small cell lung cancer: results from a randomised multicenter study with minimum 5 year follow-up. Lung Cancer 39(3):303–313 Ando S, SuzukiM, Yamamoto N, Iida T, Kimura H, 2004, The prognostic value of both neuron-specific enolase, NSE, and Cyfra21-1 in small cell lung cancer. Anticancer Res 24(3b):1941–1946 Molina R, Auge JM, Filella X, Vinolas N, Alicarte J, Domingo JM, Ballesta AM, 2005, Pro-gastrin-releasing peptide, proGRP, in patients with benign and malignant diseases: comparison with CEA, SCC, CYFRA 21-1 and NSE in patients with lung cancer. Anticancer Res 25(3 A):1773–1778 Kocsis J, Meszaros T, Madaras B, Toth EK, Kamondi S, Gal P, Varga L, Prohaszka Z, Fust G, 2011, High levels of acute phase proteins and soluble 70 kDa heat shock proteins are independent and additive risk factors for mortality in colorectal cancer. Cell Stress Chaperones 16(1):49–55., DOI 10.1007 /s12192-010-0220-z Rozenberg P, Kocsis J, Saar M, Prohaszka Z, Fust G, Fishelson Z, 2013, Elevated levels of mitochondrial mortalin and cytosolic HSP70 in blood as risk factors in patients with colorectal cancer. Int J Cancer J Int du Cancer 133(2):514–518., DOI 10.1002/ijc.28029 Calderwood SK, Ciocca DR, 2008, Heat shock proteins: stress proteins with Janus-like properties in cancer. Int J Hyperth 24(1): 31–39., DOI 10.1080/02656730701858305 ShermanM, Multhoff G, 2007, Heat shock proteins in cancer. Ann N YAcad Sci 1113:192–201., DOI 10.1196/annals.1391.030 Radons J, Multhoff G, 2005, Immunostimulatory functions of membrane-bound and exported heat shock protein 70. Exerc Immunol Rev 11:17–33 Multhoff G, Mizzen L, Winchester CC, Milner CM, Wenk S, Eissner G, Kampinga HH, Laumbacher B, Johnson J, 1999, Heat shock protein 70, Hsp70, stimulates proliferation and cytolytic activity of natural killer cells. Exp Hematol 27(11): 1627–1636 Jagadish N, Parashar D, Gupta N, Agarwal S, Suri V, Kumar R, Suri V, Sadasukhi TC, Gupta A, Ansari AS, Lohiya NK, Suri A, 2016, Heat shock protein 70–2, HSP70–2, is a novel therapeutic target for colorectal cancer and is associated with tumor growth. BMC Cancer 16(1):561., DOI 10.1186/s12885-016-2592-7 Wen W, Liu W, Shao Y, Chen L, 2014, VER-155008, a small molecule inhibitor of HSP70 with potent anti-cancer activity on lung cancer cell lines. Exp Biol Med, Maywood, 239(5):638– 645., DOI 10.1177/1535370214527899 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 377 EP 383 DI 10.1007/s12253-016-0118-x PG 7 ER PT J AU Zidi, S Stayoussef, M Alsaleh, LB Gazouani, E Mezlini, A Ebrahim, HB Yacoubi-Loueslati, B Almawi, YW AF Zidi, Sabrina Stayoussef, Mouna Alsaleh, L Bano Gazouani, Ezzedine Mezlini, Amel Ebrahim, H Bashayer Yacoubi-Loueslati, Besma Almawi, Y Wassim TI Relationships between Common and Novel Interleukin-6 Gene Polymorphisms and Risk of Cervical Cancer: a Case-Control Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Interleukin-6; Cervical cancer; FIGO stages; Polymorphisms; Haplotypes; Tunisians ID Interleukin-6; Cervical cancer; FIGO stages; Polymorphisms; Haplotypes; Tunisians AB We investigated the association between six common and novel interleukin-6 (IL-6) polymorphisms with the risk of cervical cancer (CC) among Tunisians. Study subjects comprised 112 CC cases and 164 control women. Genotyping of IL-6 rs2069845, rs2069840, rs1474348, rs1800795, rs1800797, rs2069827 variants was done by real-time PCR, with defined clusters. The allelic and genotypic distributions of the tested IL-6 SNPs were comparable between CC patients and control women. Stratification according to FIGO staging revealed that rs1800795 homozygous major allele genotype (P = 0.033; OR =0.49(0.25–0.95)) and major allele (P = 0.037; OR = 0.57 (0.33–0.97)) were protective of CC. Moreover, carriage of rs1474348 major allele was also protective of CC (P = 0.014; OR = 0.53(0.32– 0.88)), while higher rs1474348 minor allele frequency was seen in CC patients with early FIGO stage (P = 0.044; OR = 0.39 (0.15–1.00)), thus implicating rs1474348 in CC evolution and progression of angiogenesis. Haploview analysis demonstrated high linkage disequilibrium (LD) between rs2069845, rs2069840, rs1474348 and rs1800795, and 6-locus haplotype analysis identified GACCCA haplotype to be positively associated with increased CC, while GAGGGG haplotype was negatively associated with CC, thus suggesting a protective role for this haplotype in CC. Furthermore, there was a significant association between the incidence of CC and the use hormonal contraception (P = 0.047; OR = 1.97 (0.94–4.13)) and smoking (P < 0.001; OR = 7.12 (2.97–17.04)). The IL-6 variants rs1800795 and rs1474348, and haplotypes GACCCA and GAGGGG, along with use of hormonal contraceptives and smoking, are major risk factors of CC susceptibility and evolution among Tunisian women. C1 [Zidi, Sabrina] El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 1092 Tunis, Tunisia. [Stayoussef, Mouna] El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 1092 Tunis, Tunisia. [Alsaleh, L Bano] Arabian Gulf University, Department of Medical BiochemistryManama, Bahrain. [Gazouani, Ezzedine] Military Hospital of Tunis, Laboratory of ImmunologyTunis, Tunisia. [Mezlini, Amel] Salah Azeiz Oncology InstituteTunis, Tunisia. [Ebrahim, H Bashayer] Arabian Gulf University, Department of Medical BiochemistryManama, Bahrain. [Yacoubi-Loueslati, Besma] El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 1092 Tunis, Tunisia. [Almawi, Y Wassim] Arabian Gulf University, Department of Medical BiochemistryManama, Bahrain. RP Zidi, S (reprint author), El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 1092 Tunis, Tunisia. EM ZIDISABRINA86@gmail.com CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127:2893–2917 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA: Cancer Journal for Clinicians 61:69–90 Bedoya AM, Jaramillo R, Baena A, Castano J, Olaya N, Zea AH et al, 2012, Location and density of immune cells in precursor lesions and cervical cancer. Cancer Microenviron., DOI 10.1007/s12307-012-0097-8 Villa LL, 2006, Biology of genital human papillomaviruses. International Journal of Gynecology and Obstetrics 94:3–7 Rajeevan MS, Swan DC, Nisenbaum R, Lee DR, Vernon SD, Ruffin MT et al, 2005, Epidemiologic and viral factors associated with cervical neoplasia in HPV-16-positive women. Int J Cancer 115:114–120 Ribeiro KCB, 2008, Cancer do colo do Utero: Epidemiologia do cancer do colo do utero – fatores demograficos e fatores de Risco. Tecmedd 18–23 Barton BE, Jackson JV, 1993, Protective role of interleukin 6 in the lipopolysaccharide-galactosamine septic shock model. Infect Immun 61:1496 Eustace D, Han X, Gooding R, Rowbottom A, Riches P, Heyderman E, 1993, Interleukin-6, IL-6, functions as an autocrine growth factor in cervical carcinomas in vitro. Gynecol Oncol 50:15–19 Tartour E, Gey A, Sastre-Garau X, Pannetier C, Mosseri V, Kourilsky P et al, 1994, Analysis of interleukin 6 gene expression in cervical neoplasia using a quantitative polymerase chain reaction assay: evidence for enhanced interleukin 6 gene expression in invasive carcinoma. Cancer Res 54:6243–6248 Castrilli G, Tatone D, Diodoro MG, Rosini S, Piantelli M, Musiani P, 1997, Interleukin 1 alpha and interleukin 6 promote the in vitro growth of both normal and neoplastic human cervical epithelial cells. Br J Cancer 75:855–859 Abdelwahab SI, Abdul AB, Zain ZN, Hadi AH, 2012, Zerumbone inhibits interleukin-6 and induces apoptosis and cell cycle arrest in ovarian and cervical cancer cells. Int Immunopharmacol 12:594–602 Zidi S, Stayoussef M, Gazouani E, Mezlini A,Yacoubi-Loueslati B, Almawi WY, 2015, Relationship of common vascular endothelial growth factor polymorphisms and haplotypes with the risk of cervical cancer in Tunisians. Cytokine 74(1):108–112 Cohen T, Nahari D, Cerem LW, Neufeld G, Levi BZ, 1996, Interleukin 6 induces the expression of vascular endothelial growth factor. J Biol Chem 271:736–741 Wei L-H, Kuo M-L, Chen C-A, Cheng W-F, Cheng S-P, Hsieh F-J et al, 2001, Interleukin-6 in cervical cancer: the relationship with vascular endothelial growth factor. Gynecol Oncol 82:49–56 Castro FA, Haimila K, Sareneva I, SchmittM, Lorenzo J, Kunkel N et al, 2009, Association of HLA-DRB1, interleukin-6 and cyclin D1 polymorphisms with cervical cancer in the Swedish population– a candidate gene approach. Int J Cancer 125:1851–1858 Gangwar R,Mittal B, Mittal RD, 2009, Association of interleukin- 6 -174G/C promoter polymorphism with risk of cervical cancer. Int J Biol Markers 24:11–16 Nogueira de Souza NC, Brenna SM, Campos F, Syrjanen KJ, Baracat EC, Silva ID, 2006, Interleukin-6 polymorphisms and the risk of cervical cancer. Int J Gynecol Cancer 16:1278–1282 Shi T-Y, ZhuM-L, He J, WangM-Y, Li Q-X, Zhou X-Yet al, 2013, Polymorphisms of the interleukin 6 gene contribute to cervical cancer susceptibility in eastern Chinese women. Hum Genet 132:301–312 Srivani R, Nagarajan B, 2003, A prognostic insight on in vivo expression of interleukin-6 in uterine cervical cancer. Int J Gynecol Cancer 13:331–339 Porto CRC, de Oliveira Kleine JPF, Longatto AF, da Silva IDCG, 2014, Polymorphism of interleukin-6 is not associated with the presence or absence of high HPV E6/E7. Anticancer Res 34: 3501–3504 Grimm C, Watrowski R, Baumuhlner K, Natter C, Tong D, Wolf A et al, 2011, Genetic variations of interleukin-1 and −6 genes and risk of cervical intraepithelial neoplasia. Gynecol Oncol 121: 537–541 Hammes LS, Tekmal RR, Naud P, Edelweiss MI, Kirma N, Valente PT e al. Macrophages, inflammation and risk of cervical intraepithelial neoplasia, CIN, progression—clinicopathological correlation. Gynecol Oncol 2007; 105: 157–165. Fishman D, Faulds G, Jeffery R, Mohamed-Ali V, Yudkin JS, Humphries S et al, 1998, The effect of novel polymorphisms in the interleukin-6, IL-6, gene on IL-6 transcription and plasma IL-6 levels, and an association with systemic-onset juvenile chronic arthritis. J Clin Investig 102(7):1369 Xu B, Niu X-B,Wang Z-D, ChengW, Tong N,Mi Y-Yet al, 2011, IL-6− 174G > C polymorphism and cancer risk: a meta-analysis involving 29,377 cases and 37,739 controls. Mol Biol Rep 38(4): 2589–2596 Zhang H, Xu Y, Li L, Liu R, Ma B, 2012, The interleukin-6 -174G/ C polymorphism and prostate cancer risk: a systematic review and meta-analysis. Urol Int 88:447–453 Upadhyay R, Jain M, Kumar S, Ghoshal UC, Mittal B, 2008, Association of interleukin-6, −174G > C, promoter polymorphism with risk of squamous cell esophageal cancer and tumor location: an exploratory study. Clincal Immunology 128:199–204 Slattery ML, Curtin K, Sweeney C, Wolff RK, Baumgartner RN, Baumgartner KB et al, 2008, Modifying effects of IL-6 polymorphisms on body size-associated breast cancer risk. Obesity 16:339–347 Vairaktaris E, Yiannopoulos A, Vylliotis A, Yapijakis C, Derka S, Vassiliou S et al, 2006, Strong association of interleukin-6 - 174 G > C promoter polymorphism with increased risk of oral cancer. Int J Biol Markers 21:246–250 Grimm C, Six L, Tomovski C, Baumuhlner K, Natter C, Tong D et al, 2005, A common interleukin-6 promoter polymorphism in patients with vulvar cancer. J Soc Gynecol Investig 12:617–620 Beral PV, de Gonzalez AB, Colin D, Franceschi S, Goodhill A, Green J, Peto J et al, 2007, Cervical cancer and hormonal contraceptives: collaborative reanalysis of individual data for 16,573 women with cervical cancer and 35,509 women without cervical cancer from 24 epidemiological studies. Lancet 370(9599):1609–1621 Hea Young O, 2016, Mi Kyung Kim, Sang-Soo Seo, Jae-Kwan Lee. Association of combined tobacco smoking and oral contraceptive use with cervical intraepithelial neoplasia 2 or 3 in Korean women. Journal of Epidemiology 26(1):22–29 Nicol AF, Fernandes ATG, Grinsztejn B, Russomano F, Silva JRL, Tristao A et al, 2005, Distribution of immune cell subsets and cytokine-producing cells in the uterine cervix of human papillomavirus, HPV)-infected women: influence of HIV-1 coinfection. Diagn Mol Pathol 14(1):39–47 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 385 EP 392 DI 10.1007/s12253-016-0127-9 PG 8 ER PT J AU Ersen, A Pekmezci, M Folpe, LA Tihan, T AF Ersen, Ayca Pekmezci, Melike Folpe, L Andrew Tihan, Tarik TI Comparision of New Diagnostic Tools for Malignant Peripheral Nerve Sheath Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Malignant peripheral nerve sheath tumor; SOX10; SOX2; p75NTR; H3K27 ID Malignant peripheral nerve sheath tumor; SOX10; SOX2; p75NTR; H3K27 C1 [Ersen, Ayca] Dokuz Eylul University, Faculty of Medicine, Department of PathologyIzmir, Turkey. [Pekmezci, Melike] University of California San Francisco, Department of Pathology, Division of Neuropathology, 94143 San Francisco, CA, USA. [Folpe, L Andrew] Mayo Clinic, Anatomic Pathology DivisionRochester, MN, USA. [Tihan, Tarik] University of California San Francisco, Department of Pathology, Division of Neuropathology, 94143 San Francisco, CA, USA. RP Ersen, A (reprint author), Dokuz Eylul University, Faculty of Medicine, Department of Pathology, Izmir, Turkey. EM ayca.ersen@deu.edu.tr CR Louis DN, Ohgaki H, Wiestler OD, 2016, Cavenee WKe. World Health Organization histological classification of Tumours of the central nervous system. Lyon, France, International Agency for Research on Cancer Rodriguez FJ, Folpe AL, Giannini C, Perry A, 2012, Pathology of peripheral nerve sheath tumors: diagnostic overview and update on selected diagnostic problems. Acta Neuropathol 123:295–319 Thway K, Hamarneh W, Miah AB, Fisher C, 2015, Malignant peripheral nerve sheath tumor with rhabdomyosarcomatous and glandular elements: rare epithelial differentiation in a triton tumor. Int J Surg Pathol 23:377–383 Louis DN, Ohgaki H, Wiestler OD, Cavenee WKe, 2016, World Health Organization histological classification of tumours of the central nervous system, Revised 4th Edition). In: Bosman FT, Jaffe ES, Lakhani SR, Ohgaki H, eds). International Agency for Research on Cancer, Lyon, France Kang Y, Pekmezci M, Folpe AL et al, 2014, Diagnostic utility of SOX10 to distinguish malignant peripheral nerve sheath tumor from synovial sarcoma, including intraneural synovial sarcoma. Mod Pathol 27:55–61 Cruz J, Reis-Filho JS, Lopes JM, 2004, Malignant peripheral nerve sheath tumour-like primary cutaneous malignant melanoma. J Clin Pathol 57:218–220 Lisa AC, Mark RW, 2006, Immunohistology of soft tissue and osseous neoplasms. In: Dabbs DJ, ed). Diagnostic Immunohistochemistry, 2nd edition. Elsevier Karamchandani JR, Nielsen TO, van de Rijn M, West RB, 2012, Sox10 and S100 in the diagnosis of soft-tissue neoplasms. Appl Immunohistochem Mol Morphol 20:445–450 Pekmezci M, Reuss DE, 2014, Hirbe AC et al. Morphologic and immunohistochemical features of malignant peripheral nerve sheath tumors and cellular schwannomas, Mod Pathol Cleven AH, Sannaa GA, Briaire-de Bruijn I et al, 2016, Loss of H3K27 tri-methylation is a diagnostic marker for malignant peripheral nerve sheath tumors and an indicator for an inferior survival. Mod Pathol 29:582–590 Schaefer IM, Fletcher CD, Hornick JL, 2016, Loss of H3K27 trimethylation distinguishes malignant peripheral nerve sheath tumors from histologic mimics. Mod Pathol 29:4–13 Bonetti B, Panzeri L, Carner M et al, 1997, Human neoplastic Schwann cells: changes in the expression of neurotrophins and their low-affinity receptor p 75. Neuropathol Appl Neurobiol 23:380–386 Folpe AL, Schmidt RA, Chapman D, Gown AM, 1998, Poorly differentiated synovial sarcoma: immunohistochemical distinction from primitive neuroectodermal tumors and high-grade malignant peripheral nerve sheath tumors. Am J Surg Pathol 22:673–682 Fanburg-Smith JC, Miettinen M, 2001, Low-affinity nerve growth factor receptor, p 75, in dermatofibrosarcoma protuberans and other nonneural tumors: a study of 1,150 tumors and fetal and adult normal tissues. Hum Pathol 32:976–983 Woodhoo A, Sommer L, 2008, Development of the Schwann cell lineage: from the neural crest to the myelinated nerve. Glia 56: 1481–1490 Nonaka M, Kohmura E, Hirata M, Hayakawa T, 1998, Metastatic meningeal hemangiopericytoma of thoracic spine. Clin Neurol Neurosurg 100:228–230 Lazova R, Tantcheva-Poor I, Sigal AC, 2010, P75 nerve growth factor receptor staining is superior to S100 in identifying spindle cell and desmoplasticmelanoma. J AmAcad Dermatol 63:852–858 Shivane A, Parkinson DB, Ammoun S, Hanemann CO, 2013, Expression of c-Jun and sox-2 in human schwannomas and traumatic neuromas. Histopathology 62:651–656 Rohrich M, Koelsche C, Schrimpf D et al, 2016, Methylationbased classification of benign and malignant peripheral nerve sheath tumors. Acta Neuropathol 131:877–887 Prieto-Granada CN, Wiesner T, Messina JL et al, 2016, Loss of H3K27me3 expression is a highly sensitivemarker for sporadic and radiation-induced MPNST. Am J Surg Pathol 40:479–489 Miller SJ, Rangwala F, Williams J et al, 2006, Large-scale molecular comparison of human schwann cells to malignant peripheral nerve sheath tumor cell lines and tissues. Cancer Res 66:2584–2591 Miettinen M, McCue PA, Sarlomo-Rikala M et al, 2015, Sox10–a marker for not only schwannian and melanocytic neoplasms but also myoepithelial cell tumors of soft tissue: a systematic analysis of 5134 tumors. Am J Surg Pathol 39:826–835 Rad E, Tee AR, 2016, Neurofibromatosis type 1: fundamental insights into cell signalling and cancer. Semin Cell Dev Biol 52: 39–46 Lee W, Teckie S, Wiesner T et al, 2014, PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors. Nat Genet 46:1227–1232 Stasik CJ, Tawfik O, 2006, Malignant peripheral nerve sheath tumor with rhabdomyosarcomatous differentiation, malignant triton tumor). Arch Pathol Lab Med 130:1878–1881 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 393 EP 398 DI 10.1007/s12253-016-0125-y PG 6 ER PT J AU Huang, KCh Evans, A Donnelly, B Bismar, AT AF Huang, Kuo-Cheng Evans, Andrew Donnelly, Bryan Bismar, A Tarek TI SPINK1 Overexpression in Localized Prostate Cancer: a Rare Event Inversely Associated with ERG Expression and Exclusive of Homozygous PTEN Deletion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ERG; PTEN deletion; Homozygous; Hemizygous; SPINK1; Prostate cancer; Progression; Biomarkers ID ERG; PTEN deletion; Homozygous; Hemizygous; SPINK1; Prostate cancer; Progression; Biomarkers AB SPINK1 is proposed as potential prognostic marker in prostate cancer (PCA). However, its relation to PTEN and ERG in localized PCA remains unclear. The study population consisted of two independent cohorts of men treated by radical prostatectomy for localized PCA (discovery n = 218 and validation n = 129). Patterns of association between SPINK1 and each of ERG and PTEN were evaluated by immunohistochemistry and fluorescence in situ hybridization. Associations between SPINK1 expression and various pathologic parameters and clinical outcome were also investigated. SPINK1 was expressed in 15.3 % and 10.9 % of cases in the discovery and validation cohort, respectively. SPINK expression was observed in 5.56 % of high-grade prostatic intraepithelial neoplasia and 1.1 % of adjacent morphologically benign prostatic glands. SPINK1 and ERG expression were almost exclusive, with only 1.0 % of the cases co-expressing both in the same core sample. SPINK1 interfocal and within-core heterogeneity was noted in 29.2 % and 64.6%of cases, respectively. SPINK1 expression was not significantly associated with PTEN deletion in the two cohorts (p = 0.871 for discovery cohort and p = 0.293 for validation cohort). While SPINK1 expression did occur with hemizygous PTEN deletion, there was a complete absence of SPINK1 expression in PCA showing homozygous PTEN deletion, which was confirmed in the validation cohort (p = 0.02). Despite SPINK1’s association with higher Gleason score (>7) (p = 0.02), it was not associated with other pathological parameters or biochemical recurrence postradical prostatectomy. We documented absolute exclusivity between SPINK1 overexpression and homozygous PTEN deletion in localized PCA. SPINK1 and ERG expressions are exclusive events in PCA. SPINK1 is not of added prognostic value in localized PCA. C1 [Huang, Kuo-Cheng] University of Calgary and Calgary Laboratory Services, Department of Pathology and Laboratory Medicine, 7007, 14sth st sw, T2V 1P9 Calgary, AB, Canada. [Evans, Andrew] University Health Network and University of Toronto, Department of PathologyToronto, ON, Canada. [Donnelly, Bryan] University of Calgary, Department of UrologyCalgary, AB, Canada. [Bismar, A Tarek] University of Calgary and Calgary Laboratory Services, Department of Pathology and Laboratory Medicine, 7007, 14sth st sw, T2V 1P9 Calgary, AB, Canada. RP Bismar, AT (reprint author), University of Calgary and Calgary Laboratory Services, Department of Pathology and Laboratory Medicine, T2V 1P9 Calgary, Canada. EM tarek.bismar@cls.ab.ca CR Horii A, Kobayashi T, Tomita N, Yamamoto T, Fukushige S, Murotsu T et al, 1987, Primary structure of human pancreatic secretory trypsin inhibitor, PSTI, gene. Biochem Biophys Res Commun 149(2):635–641 Stenman UH, 2002, Tumor-associated trypsin inhibitor. Clin Chem 48(8):1206–1209 Flavin R, Pettersson A, Hendrickson WK, Fiorentino M, Finn S, Kunz L et al, 2014, SPINK1 protein expression and prostate cancer progression. Clin Cancer Res 20(18):4904–4911., DOI 10.1158 /1078-0432.CCR-13-1341 Tomlins SA, Rhodes DR, Yu J, Varambally S, Mehra R, Perner S et al, 2008, The role of SPINK1 in ETS rearrangement-negative prostate cancers. Cancer Cell 13(6):519–528., DOI 10.1016/j. ccr.2008.04.016 Bhalla R, Kunju LP, Tomlins SA, Christopherson K, Cortez C, Carskadon S et al, 2013, Novel dual-color immunohistochemical methods for detecting ERG-PTEN and ERG-SPINK1 status in prostate carcinoma. Mod Pathol 26(6):835–848., DOI 10.1038 /modpathol.2012.234 Leinonen KA, Saramaki OR, Furusato B, Kimura T, Takahashi H, Egawa S et al., 2013). Loss of PTEN is associated with aggressive behavior in ERG-positive prostate cancer. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology, 22(12):2333–44., DOI 10.1158 /1055-9965.EPI-13-0333-T. Jhavar S, Brewer D, Edwards S, Kote-Jarai Z, Attard G, Clark J et al, 2009, Integration of ERG gene mapping and gene-expression profiling identifies distinct categories of human prostate cancer. BJU Int 103(9):1256–1269., DOI 10.1111/j.1464-410X.2008.08200.x Lippolis G, Edsjo A, Stenman UH, Bjartell A, 2013, A highdensity tissue microarray from patients with clinically localized prostate cancer reveals ERG and TATI exclusivity in tumor cells. Prostate Cancer Prostatic Dis 16(2):145–150., DOI 10.1038 /pcan.2013.7 Wang C, Wang L, Su B, Lu N, Song J, Yang X et al, 2014, Serine protease inhibitor Kazal type 1 promotes epithelial-mesenchymal transition through EGFR signaling pathway in prostate cancer. Prostate 74(7):689–701., DOI 10.1002/pros.22787 Leinonen KA, Tolonen TT, Bracken H, Stenman UH, Tammela TL, Saramaki OR et al, 2010, Association of SPINK1 expression and TMPRSS2:ERG fusion with prognosis in endocrine-treated prostate cancer. Clin Cancer Res 16(10):2845–2851., DOI 10.1158 /1078-0432.CCR-09-2505 Grupp K, Diebel F, Sirma H, Simon R, BreitmeyerK, Steurer S et al, 2013, SPINK1 expression is tightly linked to 6q15- and 5q21- deleted ERG-fusion negative prostate cancers but unrelated to PSA recurrence. Prostate 73(15):1690–1698., DOI 10.1002 /pros.22707 Terry S, Nicolaiew N, Basset V, Semprez F, Soyeux P,Maille P et al, 2015, Clinical value of ERG, TFF3, and SPINK1 for molecular subtyping of prostate cancer. Cancer 121(9):1422–1430., DOI 10.1002/cncr.29233 Brooks JD, Wei W, Hawley S, Auman H, Newcomb L, Boyer H et al, 2015, Evaluation of ERG and SPINK1 by immunohistochemical staining and Clinicopathological outcomes in a multiinstitutional radical prostatectomy cohort of 1067 patients. PLoS One 10(7):e0132343., DOI 10.1371/journal.pone.0132343 Schrecengost R, Knudsen KE, 2013, Molecular pathogenesis and progression of prostate cancer. Semin Oncol 40(3):244–258., DOI 10.1053/j.seminoncol.2013.04.001 Bismar TA, Yoshimoto M, Duan Q, Liu S, Sircar K, Squire JA, 2012, Interactions and relationships of PTEN, ERG, SPINK1 and AR in castration-resistant prostate cancer. Histopathology 60(4):645–652., DOI 10.1111/j.1365-2559.2011.04116.x Yoshimoto M, Cunha IW, Coudry RA, Fonseca FP, Torres CH, Soares FA et al, 2007, FISH analysis of 107 prostate cancers shows that PTEN genomic deletion is associated with poor clinical outcome. Br J Cancer 97(5):678–685 Epstein JI, Allsbrook WC, Jr., Amin MB, Egevad LL, Committee IG., 2005). The 2005 international society of urological pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol, 29(9):1228–1242. Huang KC, Alshalalfa M, Hegazy SA, Dolph M, Donnelly B, Bismar TA, 2014, The prognostic significance of combined ERG and androgen receptor expression in patients with prostate cancer managed by androgen deprivation therapy. Cancer Biol Ther 15(9): 1120–1128., DOI 10.4161/cbt.29689 Yoshimoto M, Ding K, Sweet JM, Ludkovski O, Trottier G, Song KS et al, 2013, PTEN losses exhibit heterogeneity in multifocal prostatic adenocarcinoma and are associated with higher Gleason grade. Mod Pathol: an official J US Can Acad Pathol, Inc 26(3): 435–447., DOI 10.1038/modpathol.2012.162 Teng LH, Wang C, Begin LR, Dolph M, Yilmaz A, Trpkov K et al, 2013, ERG protein expression and gene rearrangements are present at lower rates in metastatic and locally advanced castrationresistant prostate cancer compared to localized disease. Urology 82(2):394–399., DOI 10.1016/j.urology.2013.03.029 BraunM, Goltz D, Shaikhibrahim Z, VogelW, Bohm D, Scheble V et al, 2012, ERG protein expression and genomic rearrangement status in primary and metastatic prostate cancer–a comparative study of two monoclonal antibodies. Prostate Cancer Prostatic Dis 15(2):165–169., DOI 10.1038/pcan.2011.67 Smith SC, Tomlins SA, 2014, Prostate cancer SubtyPINg biomarKers and outcome: is clarity emERGing? Clin Cancer Res 20(18):4733–4736., DOI 10.1158/1078-0432.CCR-14-0818 Smith SC, Palanisamy N, Zuhlke KA, Johnson AM, Siddiqui J, Chinnaiyan AM et al, 2014, HOXB13 G84E-related familial prostate cancers: a clinical, histologic, and molecular survey. Am J Surg Pathol 38(5):615–626., DOI 10.1097/PAS.0000000000000090 Gumuskaya B,Gurel B, Fedor H, Tan HL,Weier CA, Hicks JL et al, 2013, Assessing the order of critical alterations in prostate cancer development and progression by IHC: further evidence that PTEN loss occurs subsequent to ERG gene fusion. Prostate Cancer Prostatic Dis 16(2):209–215., DOI 10.1038/pcan.2013.8 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 399 EP 407 DI 10.1007/s12253-016-0119-9 PG 9 ER PT J AU Arik, D Can, C Dundar, E Kabukcuoglu, S Pasaoglu, AF Arik, Deniz Can, Cavit Dundar, Emine Kabukcuoglu, Sare Pasaoglu, Ozgul TI Prognostic Significance of CD24 in Clear Cell Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Clear cell renal cell carcinoma; Cancer stem cell marker; CD24; Prognosis ID Clear cell renal cell carcinoma; Cancer stem cell marker; CD24; Prognosis AB The role of cancer stem cells in the initiation and progression of cancer has become a well-studied area of emerging research, and stem cells with different surface markers have been identified in various types of cancer. CD24 is a membrane protein that acts as the ligand for Pselectin and has been defined as a stem cell marker of colonic cancer. The immunohistochemical expression of CD24 is associated with worse patient outcomes in small cell lung cancer, hepatocellular carcinoma, breast cancer, and colon cancer. In this study, we used immunohistochemistry to determine CD24 expression in clear cell, papillary and chromophobe renal cell carcinoma and investigated its relationship with other clinicopathological parameters and prognosis. A total of 108 cases of clear cell, 12 papillary and 13 choromophobe renal cell carcinoma were examined. Clinicopathological features including age, gender, vascular invasion, tumor necrosis, and T stage were recorded. Clinical stage and overall survival and disease-free survival times were recorded. The immunohistochemical expression of CD24 was classified as low or high based on the percentage and intensity of positive staining. CD24 expression was associated with both tumor grade and recurrence rates. The survival analysis revealed that patients with high CD24 expression exhibited significantly lower overall and disease-free survival. Increased expression of CD24 is related to the prognosis of clear cell renal cell carcinoma. This is the first study identifying a strong association between CD24 expression levels and survival. Thus, CD24 expression may aid in predicting prognosis in clear cell renal cell carcinoma. C1 [Arik, Deniz] Eskisehir Osmangazi University, Faculty of Medicine, Department of Pathology, Meselik Kampusu, 26480 Eskisehir, Turkey. [Can, Cavit] Eskisehir Osmangazi University, Faculty of Medicine, Department of UrologyEskisehir, Turkey. [Dundar, Emine] Eskisehir Osmangazi University, Faculty of Medicine, Department of Pathology, Meselik Kampusu, 26480 Eskisehir, Turkey. [Kabukcuoglu, Sare] Eskisehir Osmangazi University, Faculty of Medicine, Department of Pathology, Meselik Kampusu, 26480 Eskisehir, Turkey. [Pasaoglu, Ozgul] Eskisehir Osmangazi University, Faculty of Medicine, Department of Pathology, Meselik Kampusu, 26480 Eskisehir, Turkey. RP Arik, D (reprint author), Eskisehir Osmangazi University, Faculty of Medicine, Department of Pathology, 26480 Eskisehir, Turkey. EM denarik@hotmail.com CR Znaor A, Lortet-Tieulent J, Laversanne M, Jemal A, Bray F, 2015, International variations and trends in renal cell carcinoma incidence and mortality. Eur Urol 67:519–530 Sun C, Song H, Zhang H, Hou C, Zhai T, Huang L, Zhang L, 2012, CD133 expression in renal cell carcinoma, RCC, is correlated with nuclear hypoxia-inducing factor 1α, HIF-1α). J Cancer Res Clin Oncol 138:1619–1624 Axelson H, Johansson ME, 2013, Renal stem cells and their implications for kidney cancer. Semin Cancer Biol 23:56–61 Carrasco E, Alvarez PJ, Prados J, Melguizo C, Rama AR, Aranega A, Rodriguez-Serrano F, 2014, Cancer stem cells and their implication in breast cancer. Eur J Clin Investig 44:678–687 Guler G, Balci S, Costinean S, Ussakli CH, Irkkan C, Suren D, Sari E, Altundag K, OzisikY, Jones S, Bacher J, Shapiro CL, Huebner K, 2012, Stem cell-related markers in primary breast cancers and associated metastatic lesions. Mod Pathol 25:949–955 Bane A, Viloria-Petit A, Pinnaduwage D, Mulligan AM, O’Malley FP, Andrulis IL, 2013, Clinical-pathologic significance of cancer stem cell marker expression in familial breast cancers. Breast Cancer Res Treat 140:195–205 Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF, 2003, Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100:3983–3988 Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB, 2003, Identification of a cancer stem cell in human brain tumors. Cancer Res 63:5821–5828 Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, Jacquemier J, Viens P, Kleer CG, Liu S, Schott A, Hayes D, BirnbaumD,WichaMS, Dontu G, 2007, ALDH1 is amarker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1:555–567 Chen M, Geng J, 2006, P-selectinmediates adhesion of leukocytes, platelets, and cancer cells in inflammation, thrombosis, and cancer growth and metastasis. Arch Immunol Ther Exp 54:75–84 Baumann P, Cremers N, Kroese F, Orend G, Chiquet-Ehrismann R, Uede T, Yagita H, Sleeman JP, 2005, CD24 expression causes the acquisition of multiple cellular properties associated with tumor growth and metastasis. Cancer Res 65:10783–10793 Suzuki T, Kiyokawa N, Taguchi T, Sekino T, Katagiri YU, Fujimoto J, 2001, CD24 induces apoptosis in human B cells via the glycolipid-enriched membrane domains/rafts-mediated signaling system. J Immunol 166:5567–5577 Kristiansen G, Sammar M, Altevogt P, 2004, Tumour biological aspects of CD24, a mucin-like adhesion molecule. J Mol Histol 35: 255–262 Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres- Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE, 1994, A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367:645–648 Yu Z, Pestell TG, Lisanti MP, Pestell RG, 2012, Cancer stem cells. Int J Biochem Cell Biol 44:2144–2151 Sammar M, Aigner S, Hubbe M, Schirrmacher V, Schachner M, Vestweber D, Altevogt P, 1994, Heat-stable antigen, CD24, as ligand for mouse P-selectin. Int Immunol 6:1027–1036 Gracz AD, Fuller MK,Wang F, Li L, Stelzner M, Dunn JC, Martin MG, Magness ST, 2013, Brief report: CD24 and CD44 mark human intestinal epithelial cell populations with characteristics of active and facultative stem cells. Stem Cells 31:2024–2030 DengW, Gu L, Li X, Zheng J, Zhang Y, Duan B, Cui J, Dong J, Du J, 2016, CD24 associates with EGFR and supports EGF/EGFR signaling via RhoA in gastric cancer cells. J Transl Med 14:32 Kristiansen G, Winzer KJ, Mayordomo E, Bellach J, Schluns K, Denkert C, Dahl E, Pilarsky C, Altevogt P, Guski H, Dietel M, 2003, CD24 expression is a new prognostic marker in breast cancer. Clin Cancer Res 9:4906–4913 Bretz NP, Salnikov AV, Perne C, Keller S, Wang X, Mierke CT, Fogel M, Erbe-Hofmann N, Schlange T, Moldenhauer G, Altevogt P, 2012, CD24 controls Src/STAT3 activity in human tumors. Cell Mol Life Sci 69:3863–3879 Droz D, Zachar D, Charbit L, Gogusev J, Chretein Y, Iris L, 1990, Expression of the human nephron differentiation molecules in renal cell carcinomas. Am J Pathol 137:895–905 Lee HJ, Kim DI, Kwak C, Ku JH, Moon KC, 2008, Expression of CD24 in clear cell renal cell carcinoma and its prognostic significance. Urol 72:603–607 Wu JX, Zhao YY, Wu X, An HX, 2014, Clinicopathological and prognostic significance of CD24 overexpression in patients with gastric cancer: a meta-analysis. PLoS One 9:e114746 Senner V, Sturm A, Baur I, Schrell UH, Distel L, PaulusW, 1999, CD24 promotes invasion of glioma cells in vivo. J Neuropathol Exp Neurol 58:795–802 Friederichs J, Zeller Y, Hafezi-Moghadam A, Grone HJ, Ley K, Altevogt P, 2000, The CD24/P-selectin binding pathway initiates lung arrest of human A125 adenocarcinoma cells. Cancer Res 60: 6714–6722 Tanaka T, Terai Y, Kogata Y, Ashihara K, Maeda K, Fujiwara S, Yoo S, Tanaka Y, Tsunetoh S, Sasaki H, Kanemura M, Tanabe A, Ohmichi M, 2015, CD24 expression as a marker for predicting clinical outcome and invasive activity in uterine cervical cancer. Oncol Rep 34:2282–2288 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 409 EP 416 DI 10.1007/s12253-016-0128-8 PG 8 ER PT J AU Docs, O Hegyi, K Mokanszky, A Monusne, A Beke, L Andras, Cs Bedekovics, J Mehes, G AF Docs, Otto Hegyi, Katalin Mokanszky, Attila Monusne, Aniko Beke, Livia Andras, Csilla Bedekovics, Judit Mehes, Gabor TI Mutant KRAS Status Is Associated with Increased KRAS Copy Number Imbalance: a Potential Mechanism of Molecular Heterogeneity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Amplification; CEP12; Colorectal cancer; Heterogeneity; KRAS ID Amplification; CEP12; Colorectal cancer; Heterogeneity; KRAS AB Mutation rates determined by allele-specific PCR can be highly different in KRAS exon 2 mutant colorectal carcinoma (CRC) samples suggesting intratumoural heterogeneity. To address the effect of KRAS gene copy number on the relative mutant allele frequency the KRAS locus was individually quantified following FISH analysis in 36 cases. We observed, that mutant KRAS status was associated with an elevated KRAS locus number (2.36 ± 0.42 vs 2.63 ± 0.75; p = 0.037) reflecting an increased aneuploidy status but no true amplification of the locus. In parallel, KRAS locus copy numbers showed significant intercellular variability (1–16 copies/cell nucleus) within individual tumours also indicating to a dynamic intratumoural oscillation of the mutant allele copy number. In conclusion, aneusomy is a common feature of KRAS mutant CRC and KRAS copy number variations may have an impact on the relative mutant allele frequency detected by allele specific PCR/sequencing), potentially leading to subclonal diversity and influencing tumour behaviour. C1 [Docs, Otto] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Hegyi, Katalin] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Mokanszky, Attila] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Monusne, Aniko] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Beke, Livia] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Andras, Csilla] University of Debrecen, Department of Oncology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Bedekovics, Judit] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. RP Mehes, G (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, H-4032 Debrecen, Hungary. EM gabor.mehes@med.unideb.hu CR Neumann J, Zeindl-Eberhart E, Kirchner T, Jung A, 2009, Frequency and type of KRAS mutations in routine diagnostic analysis of metastatic colorectal cancer. Pathol Res Pract 205(12):858–862 Lievre A, Bachet JB, Le Corre D et al, 2006, KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 66(8):3992–3995 van Krieken JH, Jung A, Kirchner T et al, 2008, KRAS mutation testing for predicting response to anti-EGFR therapy for colorectal carcinoma: proposal for an European quality assurance program. Virchows Arch 453(5):417–431 Amado RG, Wolf M, Peeters M et al, 2008, Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 26(10):1626–1634 Karapetis CS, Khambata-Ford S, Jonker DJ et al, 2008, K-ras mutations and benefit fromcetuximab in advanced colorectal cancer. N Engl J Med 359(17):1757–1765 Yu S, Xiao X, Lu J, Qian X, Liu B, Feng J, 2013, Colorectal Cancer Patients with Low Abundance of KRAS Mutation May Benefit from EGFR Antibody Therapy. PLoS One 8(7) Docs O, Fazakas F, Horvath NL et al, 2015, Changes of KRAS exon 2 codon 12/13 mutation status in recurrent colorectal cancer. Pathol Oncol Res 21(2):399–404 Bakhoum SF, Compton DA, 2012, Chromosomal instability and cancer: a complex relationship with therapeutic potential. J Clin Invest 122(4):1138–1143 Orum H, Nielsen PE, Egholm M, Berg RH, Buchardt O, Stanley C, 1993, Single base pair mutation analysis by BNA directed PCR clamping. Nucleic Acids Res 21(23):5332–5336 Cappuzzo F, Hirsch FR, Rossi E et al, 2005, Epidermal growth factor receptor Gene and Protein and Gefitinib sensitivity in non– small-cell lung cancer. J Natl Cancer Inst 97(9):643–655 Fehrmann RS, Karjalainen JM, Krajewska M et al, 2015, Gene expression analysis identifies global gene dosage sensitivity in cancer. Nat Genet 47(2):115–125 Moroni M, Veronese S, Benvenuti S et al, 2005, Gene copy number for epidermal growth factor receptor, EGFR, and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. Lancet Oncol 6(5):279–286 Seth R, Crook S, Ibrahem S, Fadhil W, Jackson D, Ilyas M, 2009, Concomitant mutations and splice variants in KRAS and BRAF demonstrate complex perturbation of the ras/Raf signalling pathway in advanced colorectal cancer. Gut 58(9): 1234–1241 Valtorta E, Misale S, Sartore-Bianchi A et al, 2013, KRAS gene amplification in colorectal cancer and impact on response to EGFR-targeted therapy. Int J Cancer 133(5): 1259–1265 Herrera LA, Prada D, Andonegui MA, Gonzalez AD, 2008, The epigenetic origin of aneuploidy. Curr Genomics 9(1):43–50 Hegyi K, Mehes G, 2012, Mitotic failures in cancer: aurora B kinase and its potential role in the development of aneuploidy. Pathol Oncol Res. 18(4):761–769 Soh J, Okumura N, Lockwood WW et al Oncogene mutations, copy number gains and mutant allele specific imbalance, MASI, frequently occur together in tumour cells. PLoS One s4(10):e7464 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 417 EP 423 DI 10.1007/s12253-016-0126-x PG 7 ER PT J AU Shinagawa, K Yanamoto, S Naruse, T Kawakita, A Morishita, K Sakamoto, Y Rokutanda, S Umeda, M AF Shinagawa, Kenichi Yanamoto, Souichi Naruse, Tomofumi Kawakita, Akiko Morishita, Kota Sakamoto, Yuki Rokutanda, Satoshi Umeda, Masahiro TI Clinical Roles of Interleukin-6 and STAT3 in Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Il-6; STAT3; Oral squamous cell carcinoma; Lymphangiogenesis ID Il-6; STAT3; Oral squamous cell carcinoma; Lymphangiogenesis AB The effect inflammation has on cancer prognosis is marked by the presence of cytokines and chemokines. Interleukin-6 (IL-6) is one a multifunctional cytokine that regulates inflammatory responses.We investigated the roles of IL- 6 and STAT3 and examined the relationship between IL-6 signaling and clinicopathological factors in patients with oral squamous cell carcinoma (OSCC). We retrospectively examined 116 patientswho underwent radical surgery for OSCC. IL- 6 and STAT3 expression were detected by immunohistochemistry. IL-6 and STAT3 positivity were detected by IHC, at 78.4 and 80.2 %, respectively. IL-6 expression was significantly associated with pattern of invasion (P = 0.004), vascular invasion (P = 0.003), and pathological nodal status (P = 0.019). Multivariate logistic regression analysis revealed that IL-6 expression was significantly associated with vascular invasion (P = 0.044). Meanwhile, there was no significant association between STAT3 expression and clinicopathological factors and no significant relationship between IL-6 and STAT3 expression. IL-6 expression was significantly associated with 5-year disease-free survival. These results suggest that IL-6 is involved in lymphangiogenesis and recurrence in OSCC. C1 [Shinagawa, Kenichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yanamoto, Souichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Naruse, Tomofumi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Kawakita, Akiko] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Morishita, Kota] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Sakamoto, Yuki] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Rokutanda, Satoshi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Umeda, Masahiro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. RP Yanamoto, S (reprint author), Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 852-8588 Nagasaki, Japan. EM syana@nagasaki-u.ac.jp CR Fitzmaurice C, Dicker D, Pain A et al, 2015, The global burden of cancer 2013. JAMA Oncol 1:505–527 Rogers SN, Brown JS,Woolgar JA et al, 2009, Survival following primary surgery for oral cancer. Oral Oncol 45:201–211 Turcotte S, Rosenberg SA, 2011, Immunotherapy for metastatic solid cancers. Adv Surg 45:341–360 Bell RB, Leidner R, Feng Z et al, 2015, Developing an immunotherapy strategy for the effective treatment of oral, head and neck squamous cell carcinoma. J Oral Maxillofac Surg 73:S107–S115 Chen HH, Chen IH, Liao CT et al, 2011, Preoperative circulating C-reactive protein levels predict pathological aggressiveness in oral squamous cell carcinoma: a retrospective clinical study. Clin Otolaryngol 36:147–153 Huang SF, Wei FC, Liao CT et al, 2012, Risk stratification in oral cavity squamous cell carcinoma by preoperative CRP and SCC antigen levels. Ann Surg Oncol 19:3856–3864 Fang HY, Huang XY, Chien HT et al, 2013, Refining the role of preoperative C-reactive protein by neutrophil/lymphocyte ratio in oral cavity squamous cell carcinoma. Laryngoscope 123:2690–2699 Culig Z, 2013, Interleukin-6 as a therapy target in oral squamous carcinoma. Expert Opin Ther Targets 17:53–59 Jinno T, Kawano S, Maruse Yet al, 2015, Increased expression of interleukin-6 predicts poor response to chemoradiotherapy and unfavorable prognosis in oral squamous cell carcinoma. Oncol Rep 33:2161–2168 Sato J, Ohuchi M, Wada M et al, 2015, Differences in sequential posttreatment salivary IL-6 levels between patients with and patients without locoregional recurrences of oral squamous cell carcinoma: part III of a cohort study. Oral Surg Oral Med Oral Pathol Oral Radiol 120:751–760 Coussens LM, Werb Z, 2002, Inflammation and cancer. Nature 420:860–867 Yu H, Kortylewski M, Pardoll D, 2007, Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment. Nat Rev Immunol 7:41–51 Heikkila K, Ebrahim S, Lawlor DA, 2007, A systematic review of the association between circulating concentrations of C reactive protein and cancer. J Epidemiol Community Health 61:824–833 Allin KH, Bojesen SE, Nordestgaard BG, 2009, Baseline Creactive protein is associated with incident cancer and survival in patients with cancer. J Clin Oncol 27:2217–2224 Yu H, Pardoll D, Jove R, 2009, STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer 9:798–809 Kruse AL, Luebbers HT, Gratz K et al, 2010, C-reactive protein levels: a prognostic marker for patients with head and neck cancer? Head Neck Oncol 2:21 Neurath MF, Finotto S, 2011, IL-6 signaling in autoimmunity, chronic inflammation and inflammation-associated cancer. Cytokine Growth Factor Rev 22:83–89 Paramanathan A, Saxena A, Morris DL, 2014, A systematic review and meta-analysis on the impact of pre-operative neutrophil lymphocyte ratio on long term outcomes after curative intent resection of solid tumours. Surg Oncol 23:31–39 Perisanidis C, Kornek G, Poschl PWet al, 2013, High neutrophilto- lymphocyte ratio is an independent marker of poor diseasespecific survival in patients with oral cancer. Med Oncol 30:334 de Oliveira MV, Fraga CA, Gomez RS et al, 2009, Immunohistochemical expression of interleukin-4, −6, −8, and −12 in inflammatory cells in surrounding invasive front of oral squamous cell carcinoma. Head Neck 31:1439–1446 Guo Y, Xu F, Lu T et al, 2012, Interleukin-6 signaling pathway in targeted therapy for cancer. Cancer Treat Rev 38:904–910 Tawara K, Oxford JT, Jorcyk CL et al, 2011, Clinical significance of interleukin, IL)-6 in cancer metastasis to bone: potential of anti- IL-6 therapies. Cancer Manag Res 3:177–189 Yoshimura A, 2006, Signal transduction of inflammatory cytokines and tumor development. Cancer Sci 97:439–447 Mali SB, 2015, Review of STAT3, signal transducers and activators of transcription, in head and neck cancer. Oral Oncol 51:565–569 Sobin LH, Gospodarowicz MK, Wittekind C, 2009, International Union against Cancer, UICC, TNM classification of malignant tumors. 7th ed. Wiley-Blackwell, Oxford Bryne M, Koppang HS, Lilleng R et al, 1992, Malignancy grading of the deep invasive margins of oral squamous cell carcinomas has high prognostic value. J Pathol 166:375–381 Shinriki S, Jono H, Ueda M et al, 2011, Interleukin-6 signaling regulates vascular endothelial growth factor-C synthesis and lymphangiogenesis in human oral squamous cell carcinoma. J Pathol 225:142–150 Yadav A, Kumar B, Datta J et al, 2011, IL-6 promotes head and neck tumor metastasis by inducing epithelial-mesenchymal transition via the LAK-STAT3-SNAIL signaling pathway. Mol Cancer Res 9:1658–1667 Otrock ZK, Makarem JA, Shamseddine AI, 2007, Vascular endothelial growth factor family of ligants and receptors: review. Blood Cells Mol Dis 38:258–268 Omoto I, Matsumoto M, Okumura H et al, 2014, Expression of vascular endothelial growth factor-C and vascular endothelial growth factor receptor-3 in esophageal squamous cell carcinoma. Oncol Lett 7:1027–1032 Arinaga M, Noguchi T, Takeno S et al, 2003, Clinical significance of vascular endothelial growth factor C and vascular endothelial growth factor receptor 3 in patients with nonsmall cell lung carcinoma. Cancer 97:457–464 Wakisaka N, Hirota K, Kondo S et al, 2012, Induction of lymphangiogenesis through vascular endothelial growth factor-C/ vascular endothelial growth factor receptor 3 axis and its correlation with lymph node metastasis in nasopharyngeal carcinoma. Oral Oncol 48:703–708 Duffy SA, Taylor JMG, Terrell JE et al, 2008, Interleukin-6 predicts recurrence and survival among head and neck cancer patients. Cancer 113:750–757 Jablonska J, Leschner S, Westphal K et al, 2010, Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. J Clin Invest 120:1151–1164 Li MX, Liu XM, Zhang XF et al, 2014, Prognostic role of neutrophil-to-lymphocyte ratio in colorectal cancer: a systematic review and meta-analysis. Int J Cancer 134:2403–2413 Shen L, Zhang H, Liang L et al, 2014, Baseline neutrophillymphocyte ratio, ≥2.8, as a prognostic factor for patients with locally advanced rectal cancer undergoing neoadjuvant chemoradiation. Radiat Oncol 9:295 Kim IY, You SH, Kim YW, 2014, Neutrophil-lymphocyte ratio predicts pathologic tumor response and survival after preoperative chemoradiation for rectal cancer. BMC Surg 14:94 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 425 EP 431 DI 10.1007/s12253-016-0134-x PG 7 ER PT J AU Ikegawa, Sh Doki, N Kaito, S Kurosawa, Sh Sakaguchi, M Harada, K Yamamoto, K Hino, Y Shingai, N Senoo, Y Watanabe, D Hagino, T Yoshioka, K Watakabe, K Igarashi, A Najima, Y Kobayashi, T Kakihana, K Sakamaki, H Ohashi, K AF Ikegawa, Shuntaro Doki, Noriko Kaito, Satoshi Kurosawa, Shuhei Sakaguchi, Masahiro Harada, Kaito Yamamoto, Keita Hino, Yutaro Shingai, Naoki Senoo, Yasushi Watanabe, Daisuke Hagino, Takeshi Yoshioka, Kosuke Watakabe, Kyoko Igarashi, Aiko Najima, Yuho Kobayashi, Takeshi Kakihana, Kazuhiko Sakamaki, Hisashi Ohashi, Kazuteru TI Central Nervous System Involvement at the Time of Allogeneic Hematopoietic Stem Cell Transplantation Is Associated with a Poor Outcome in Patients with Acute Myeloid Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Central nervous system involvement; Allogeneic hematopoietic stem cell transplantation; Acute myeloid leukemia ID Central nervous system involvement; Allogeneic hematopoietic stem cell transplantation; Acute myeloid leukemia AB Recent reports suggested that central nervous system (CNS) involvement (CNS+) in patients with acute myeloid leukemia (AML) before allogeneic hematopoietic stem cell transplantation (allo-HSCT) is not an independent predictor of survival after allo-HSCT. However, these studies did not analyze minimal residual disease in the CNS at the time of allo-HSCT. We evaluated the effect of residual CNS+ on the transplant outcomes of 214 AML patients in a single institution. Twenty-one (10%) patients were diagnosed with CNS+ prior to allo-HSCT. Of these, 13 patients had CNS disease at the time of allo-HSCT. The patients in CNS+ AML remission at the time of allo-HSCT had better overall survival (OS) than the patients who were not in remission (2-year OS: 55% vs. 7.7%, p = 0.0001). In multivariate analyses, CNS+ at the time of allo- HSCT (hazard ratio (HR), 1.9; 95% confidence interval (CI), 1.05–3.59; p = 0.04), age over 50 years at the time of allo-HSCT, and non-complete remission disease status in bone marrow at the time of allo-HSCT were independent adverse factors for OS. However, a prior history of CNS+ before allo-HSCT did not independently affect OS (HR, 1.27; 95% CI 0.53–2.07; p = 0.6). Early diagnosis and eradication of CNS+ at the time of allo-HSCT may be necessary to improve the outcome for patients with CNS+ AML. C1 [Ikegawa, Shuntaro] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Doki, Noriko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kaito, Satoshi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kurosawa, Shuhei] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Sakaguchi, Masahiro] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Harada, Kaito] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Yamamoto, Keita] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Hino, Yutaro] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Shingai, Naoki] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Senoo, Yasushi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Watanabe, Daisuke] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Hagino, Takeshi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Yoshioka, Kosuke] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Watakabe, Kyoko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Igarashi, Aiko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Najima, Yuho] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kobayashi, Takeshi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Kakihana, Kazuhiko] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Sakamaki, Hisashi] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. [Ohashi, Kazuteru] Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677 Tokyo, Japan. RP Doki, N (reprint author), Komagome Hospital, Tokyo Metropolitan Cancer and Infectious Disease Center, Hematology Division, 113-8677 Tokyo, Japan. EM n-doki@cick.jp CR Bommer M, von Harsdorf S, Dohner H, Bunjes D, Ringhoffer M, 2010, Neoplastic meningitis in patients with acute myeloid leukemia scheduled for allogeneic hematopoietic stem cell transplantation. Haematologica 11:1969–1972 Sanders KE, Ha CS, Cortes-Franco JE, Koller CA, Kantarjian HM, Cox JD, 2004, The role of craniospinal irradiation in adults with a central nervous system recurrence of leukemia. Cancer 100:2176– 2180 Mayadev JS,Douglas JG, Storer BE, Appelbaum FR, Storb R, 2011, Impact of cranial irradiation added to intrathecal conditioning in hematopoietic cell transplantation in adult acute myeloid leukemia with central nervous system involvement. Int J Radiat Oncol Biol Phys 80:193–198 Aoki J, Ishiyama K, Taniguchi S et al, 2014, Outcome of allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia patients with central nervous system involvement. Biol Blood Marrow Transplant 20:2029–2033 Bar M, Tong W, Othus M, Loeb KR, Estey EH, 2015, Central nervous system involvement in acute myeloid leukemia patients undergoing hematopoietic cell transplantation. Biol Blood Marrow Tranplant 21:546–551 Pui CH, Thiel E, 2009, Central nervous system disease in hematologic malignancies: historical perspective and practical applications. Semin Oncol 36:S2–S16 Ruutu T, Corradini P, Gratwohl A et al, 2005, Use of intrathecal prophylaxis in allogeneic haematopoietic stem cell transplantation for malignant blood diseases: a survey of the European Group for Blood and Marrow Transplantation EBMT. Bone Marrow Transplant 35:121–124 Oshima K, Kanda Y, Yamashita T et al, 2008, Central nervous system relapse of leukemia after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 14:1100–1107 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 433 EP 437 DI 10.1007/s12253-016-0162-6 PG 5 ER PT J AU Zhang, YY Wang, QM Niu, HL Liu, X Zhang, QL AF Zhang, Yao-Yao Wang, Qi-Ming Niu, Hui-Lin Liu, Xia Zhang, Qing-Ling TI The General Expression Analysis of WTX Gene in Normal and Cancer Tissues SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Wilms’ tumor suppressorX chromosome (WTX); Tumor suppressor; Immunohistochemical staining; In situ hybridization ID Wilms’ tumor suppressorX chromosome (WTX); Tumor suppressor; Immunohistochemical staining; In situ hybridization AB WTX (Wilms’ tumor suppressor X chromosome) is a novel putative tumor suppressor gene in Wilms’ tumor of kidney, its expression and function in other human cancers had not been explored. This study detected the expression of WTX in 459 cases of 15 organs of cancers and adjacent normal tissues by using immunohistochemical staining (IHC), and validated them by in situ hybridization (ISH) and quantitative real-time reverse transcription PCR (qRT-PCR). IHC and ISH data showed that WTX protein was generally expressed in normal tissues, but reduced expression in corresponding cancers. This study demonstrated that WTX downregulation is a common phenomenon in human cancers, WTX might be a general tumor-suppressor gene and biological marker of multiple cancer tissues. Apart from kidney, stomach is another target tissue of WTX gene. The germline and somatic mutations of WTX were screened in 12 gastric cancer patients and identified in one cases (8.3%). Mutation in the WTX gene might be one of the reasons of WTX loss in gastric cancer patients. C1 [Zhang, Yao-Yao] Nanfang Hospital, Department of Pathology, 510515 Guangzhou, China. [Wang, Qi-Ming] Nanfang Hospital, Department of Pathology, 510515 Guangzhou, China. [Niu, Hui-Lin] Nanfang Hospital, Department of Pathology, 510515 Guangzhou, China. [Liu, Xia] Nanfang Hospital, Department of Pathology, 510515 Guangzhou, China. [Zhang, Qing-Ling] Nanfang Hospital, Department of Pathology, 510515 Guangzhou, China. RP Zhang, QL (reprint author), Nanfang Hospital, Department of Pathology, 510515 Guangzhou, China. EM zqllc8@126.com CR Rivera MN, Kim WJ, Wells J, Driscoll DR, Brannigan BW, Han M, Kim JC, FeinbergAP, GeraldWL,Vargas SO, Chin L, Iafrate AJ, Bell DW, Haber DA, 2007, An X chromosome gene, WTX, is commonly inactivated in Wilms tumor. Science 315(5812):642–645 Alexandrescu, S., S. Akhavanfard, M.H. Harris, and S.O. Vargas, Clinical, pathologic, and genetic features of Wilms tumors with WTX gene mutation. Pediatr Dev Pathol, 2016. X-ing Out Tumor Suppression. Science, 2007. 315: p. 569 h. Bagchi S, 2007, New tumour suppressor linked toWilms’ tumour. Lancet Oncol 8(2):102 KimMK, Min DJ, Rabin M, Licht JD, 2011, Functional characterization ofWilms tumor-suppressorWTX and tumor-associated mutants. Oncogene 30(7):832–842 Jenkins ZA, van Kogelenberg M, Morgan T, Jeffs A, Fukuzawa R, Pearl E, Thaller C, Hing AV, Porteous ME, Garcia-Minaur S, Bohring A, Lacombe D, Stewart F, Fiskerstrand T, Bindoff L, Berland S, Ades LC, Tchan M, David A, Wilson LC, Hennekam RC, Donnai D, Mansour S, Cormier-Daire V, Robertson SP, 2009, Germline mutations in WTX cause a sclerosing skeletal dysplasia but do not predispose to tumorigenesis. Nat Genet 41(1):95–100 Fujita A, Ochi N, Fujimaki H, Muramatsu H, Takahashi Y, Natsume J, Kojima S, Nakashima M, Tsurusaki Y, Saitsu H, Matsumoto N, Miyake N, 2014, A novel WTX mutation in a female patient with osteopathia striata with cranial sclerosis and hepatoblastoma. Am J Med Genet A 164A(4):998–1002 Comai G, Boutet A, Neirijnck Y, Schedl A, 2010, Expression patterns of theWtx/Amer gene family during mouse embryonic development. Dev Dyn 239(6):1867–1878 He L, Ding Y, Zhang Q, Che X, He Y, Shen H,Wang H, Li Z, Zhao L, Geng J, Deng Y, Yang L, Li J, Cai J, Qiu L,Wen K, Xu X, Jiang S, 2006, Expression of elevated levels of pro-inflammatory cytokines in SARS-CoV-infected ACE2+ cells in SARS patients: relation to the acute lung injury and pathogenesis of SARS. J Pathol 210(3):288–297 Qingling Z, Lina Y, Li L, ShuangW, Yufang Y, Yi D, Divakaran J, Xin L, Yanqing D, 2011, LMP1 antagonizes WNT/beta-catenin signalling through inhibition of WTX and promotes nasopharyngeal dysplasia but not tumourigenesis in LMP1(B95-8, transgenic mice. J Pathol 223(5):574–583 MajorMB, Camp ND, Berndt JD, Yi X, Goldenberg SJ, Hubbert C, Biechele TL, Gingras AC, Zheng N, Maccoss MJ, Angers S,Moon RT, 2007, Wilms tumor suppressor WTX negatively regulates WNT/beta-catenin signaling. Science 316(5827):1043–1046 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), method. Methods 25(4):402–408 Armat M, Ramezani F, Molavi O, Sabzichi M, Samadi N, 2016, Six family of homeobox genes and related mechanisms in tumorigenesis protocols. Tumori:0 Kress E, Skah S, Sirakov M, Nadjar J, Gadot N, Scoazec JY, Samarut J, Plateroti M, 2010, Cooperation between the thyroid hormone receptor TRalpha1 and theWNT pathway in the induction of intestinal tumorigenesis. Gastroenterology 138(5):1863–1874 Rivlin N, Brosh R, Oren M, Rotter V, 2011, Mutations in the p53 tumor suppressor Gene: importantmilestones at the various steps of tumorigenesis. Genes Cancer 2(4):466–474 Holloway KR, Sinha VC, Bu W, Toneff M, Dong J, Peng Y, Li Y, 2016, Targeting oncogenes into a defined subset ofmammary cells demonstrates that the initiating oncogenic mutation defines the resulting tumor phenotype. Int J Biol Sci 12(4):381–388 ZhouQM, LiW, Zhang X, Chen YB, Chen XC, Guan YX, Ding Y, Wen XZ, Xia Q, Zhou Q, Peng RQ, Hou JH, Zhu XF, Zeng YX, Zhang XS, 2012, The mutation profiles of common oncogenes involved in melanoma in southern China. J Invest Dermatol 132(7):1935–1937 Evangelisti C, de Biase D, Kurelac I, Ceccarelli C, Prokisch H, Meitinger T, Caria P, Vanni R, Romeo G, Tallini G, Gasparre G, Bonora E, 2015, A mutation screening of oncogenes, tumor suppressor gene TP53 and nuclear encoded mitochondrial complex I genes in oncocytic thyroid tumors. BMC Cancer 15:157 Perotti D, Gamba B, SardellaM, Spreafico F, Terenziani M, Collini P, Pession A, Nantron M, Fossati-Bellani F, Radice P, 2008, Functional inactivation of the WTX gene is not a frequent event in Wilms’ tumors. Oncogene 27(33):4625–4632 Fukuzawa R, Holman SK, Chow CW, Savarirayan R, Reeve AE, Robertson SP, 2010)WTX mutations can occur both early and late in the pathogenesis ofWilms tumour. J Med Genet 47(11):791–794 Scheel SK, Porzner M, Pfeiffer S, Ormanns S, Kirchner T, Jung A, 2010, Mutations in the WTX-gene are found in some high-grade microsatellite instable, MSI-H, colorectal cancers. BMC Cancer 10:413 Chung NG, Kim MS, Chung YJ, Yoo NJ, Lee SH, 2008, Tumor suppressor WTX gene mutation is rare in acute leukemias. Leuk Lymphoma 49(8):1616–1617 Zilberman D, Coleman-Derr D, Ballinger T, Henikoff S, 2008, Histone H2A.Z and DNA methylation are mutually antagonistic chromatin marks. Nature 456(7218):125–129 Liu X, Wang Q, Niu H, Yang X, Sun J, Zhang Q, Ding Y, 2013, Promotermethylation ofWilms’ tumor gene on the X- chromosome in gastric cancer. Nan Fang Yi Ke Da Xue Xue Bao 33(3):318–321 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 439 EP 446 DI 10.1007/s12253-016-0168-0 PG 8 ER PT J AU Woyen, VTA Laczko, G Hoyer, S Hegyi, L AF Woyen, Vidar Tind Arne Laczko, Gergely Hoyer, Soren Hegyi, Laszlo TI The Missing Link in the Diagnostic Pathway of Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostatic carcinoma; Adenocarcinoma; Small cell carcinoma; Immunohistochemistry ID Prostatic carcinoma; Adenocarcinoma; Small cell carcinoma; Immunohistochemistry AB Prostate cancer is one of the most common cancers in the Western world. It is among the leading causes of cancer related death. While its incidence and survival increased significantly during the last few decades in Denmark, the mortality rate did not change for patients younger than 80 year old. Development of new techniques, such as multiparametric MRI, helps to increase the accuracy of diagnosis. However, a missing link in the diagnostic pathway may result in mistreatment if an acinar adenocarcinoma of prostate is transformed into a neuroendocrine phenotype such as small cell carcinoma. C1 [Woyen, Vidar Tind Arne] Regionshospitalet Viborg, Patologisk Institut, Pakhusvej 1, 8800 Viborg, Denmark. [Laczko, Gergely] Regionshospitalet Viborg, Urologisk AfdelingViborg, Denmark. [Hoyer, Soren] Aarhus Universitetshospital, Patologisk InstitutAarhus, Denmark. [Hegyi, Laszlo] Regionshospitalet Viborg, Patologisk Institut, Pakhusvej 1, 8800 Viborg, Denmark. RP Hegyi, L (reprint author), Regionshospitalet Viborg, Patologisk Institut, 8800 Viborg, Denmark. EM lhegyi@doctors.org.uk CR Cookson MS, Roth BJ, Dahm P, et al., 2015, Castration-Resistant Prostate Cancer. AUA Guideline. In AUA Guidelines https://www. auanet.org/education/guidelines/castration-resistant-prostatecancer. cfm. Accessed 27 June 2016 Mottet N, Bellmunt J, Briers E, et al., 2016, Prostate Cancer. https://uroweb.org/guideline/prostate-cancer/. Accessed 27 June 2016 Palmgren JS, Karavadia SS, Wakefield MR, 2007, Unusual and underappreciated: small cell carcinoma of the prostate. Semin Oncol 34:22–29 Spiess PE, Pettaway CA, Vakar-Lopez F et al, 2007, Treatment outcomes of small cell carcinoma of the prostate: a single-center study. Cancer 110:1729–1737 Li Z, Sun Y, Chen X et al, 2015, P 53 mutation directs AURKA overexpression via miR-25 and FBXW7 in prostatic small cell neuroendocrine carcinoma. Mol Cancer Res 13:584–591 Terry S, Beltran H, 2014, The many faces of neuroendocrine differentiation in prostate cancer progression. Front Oncol 4:60 Burchardt T, Burchardt M, ChenMWet al, 1999, Transdifferentiation of prostate cancer cells to a neuroendocrine cell phenotype in vitro and in vivo. J Urol 162:1800–1805 Santoni M, Conti A, Burattini L et al, 2014, Neuroendocrine differentiation in prostate cancer: novel morphological insights and future therapeutic perspectives. Biochim Biophys Acta 1846:630– 637 Vlachostergios PJ, Papandreou CN, 2015, Targeting neuroendocrine prostate cancer: molecular and clinical perspectives. Front Oncol 5:6 Small EJ, Huang J, Youngren J, et al., 2015, Characterization of neuroendocrine prostate cancer, NEPC, in patients with metastatic castration resistant prostate cancer, mCRPC, resistant to abiraterone, Abi, or enzalutamide, Enz): Preliminary results from the SU2C/PCF/AACR West Coast Prostate Cancer Dream Team, WCDT). 2015 ASCO Annual Meeting. http://meetinglibrary. asco.org/content/152182-156. Accessed 27 June 2016 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 447 EP 450 DI 10.1007/s12253-016-0183-1 PG 4 ER PT J AU Lee, HJ Kim, SS Kim, SM Yoo, JN Lee, HS AF Lee, Hwa Ju Kim, Soo Sung Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI WRN, the Werner Syndrome Gene, Exhibits Frameshift Mutations in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Lee, Hwa Ju] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Soo Sung] The Catholic University of Korea, College of Medicine, Department of Internal MedicineSeoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Futami K, IshikawaY, Goto M, Furuichi Y, SugimotoM(2008, Role of Werner syndrome gene product helicase in carcinogenesis and in resistance to genotoxins by cancer cells. Cancer Sci 99:843–848 Goto M, Miller RW, Ishikawa Y, Sugano H, 1996, Excess of rare cancers in Werner syndrome, adult progeria,, 1996, J. Cancer Epidemiol Biomark Prev 5:239–246 Shimamoto A, Sugimoto M, Furuichi Y, 2004, Molecular biology of Werner syndrome 2004. Int J Clin Oncol 9:288–298 Yamagata K, Kato J, Shimamoto A, Goto M, Furuichi Y, Ikeda H, 1998, Bloom's and Werner's syndrome genes suppress hyperrecombination in yeast sgs1 mutant: implication for genomic instability in human diseases. Proc Natl Acad Sci U S A 95: 8733–8738 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Choi MR, Gwak M, Yoo NJ, Lee SH, 2015, Regional bias of Intratumoral genetic heterogeneity of apoptosis-related genes BAX, APAF1, and FLASH in Colon cancers with high microsatellite instability. Dig Dis Sci 60:1674–1679 Kim TM, Jung SH, An CH, Lee SH, Baek IP, Kim MS, Park SW, Rhee JW, Lee SH, Chung YJ, 2015, Subclonal genomic architectures of primary and metastatic colorectal cancer based on Intratumoral genetic heterogeneity. Clin Cancer Res 21:4461–4472 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 451 EP 452 DI 10.1007/s12253-016-0173-3 PG 2 ER PT J AU Jo, SY Kim, SS Kim, SM Yoo, JN Lee, HS AF Jo, Sol Yun Kim, Soo Sung Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Frameshift Mutation of FXR1 Encoding a RNA-Binding Protein in Gastric and Colorectal Cancers with Microsatellite Instability SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE FXR1; Frameshift mutation; Tumor suppressor gene; Intrarumoral heterogeneity ID FXR1; Frameshift mutation; Tumor suppressor gene; Intrarumoral heterogeneity C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Soo Sung] The Catholic University of Korea, College of Medicine, Department of Internal MedicineSeoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Raheja R, Gandhi R, 2016, FXR1: linking cellular quiescence, immune genes and cancer. Cell Cycle 15:2695–2696 Vasudevan S, Steitz JA, 2007, AU-rich-element-mediated upregulation of translation by FXR1 and Argonaute 2. Cell 128:1105–1118 Qian J, HassaneinM, HoeksemaMD, Harris BK, Zou Y, Chen H, Lu P, Eisenberg R, Wang J, Espinosa A, Ji X, Harris FT, Rahman SM, Massion PP, 2015, The RNA binding protein FXR1 is a new driver in the 3q26-29 amplicon and predicts poor prognosis in human cancers. Proc Natl Acad Sci U S A 112:3469–3474 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Oh HR, An CH, Yoo NJ, Lee SH, 2015, Frameshift mutations of MUC15 gene in gastric and its regional heterogeneity in gastric and colorectal cancers. Pathol Oncol Res 21:713–718 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 453 EP 454 DI 10.1007/s12253-016-0177-z PG 2 ER PT J AU Robert, L Labat-Robert, J AF Robert, Ladislas Labat-Robert, Jacqueline TI Platelets, Micro-Particles and Elastase. A Review with Extrapolation to the Mechanism of Generation and Bio-Pathology of Platelet Fragments SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Robert, Ladislas] Hopital Hotel Dieu, Laboratoire de Recherche OphtalmologiqueParis, France. [Labat-Robert, Jacqueline] Hopital Hotel Dieu, Laboratoire de Recherche OphtalmologiqueParis, France. RP Robert, L (reprint author), Hopital Hotel Dieu, Laboratoire de Recherche Ophtalmologique, Paris, France. EM lrobert5@orange.fr CR Schiro A, Wilkinson RL, Weston R, Smyth V, Serracino-Inglott F, Alexander MY, 2015, Elevated levels of endothelial-derived microparticles, and serum CXCL9 and SCGF-β are associated with unstable asymptomatic carotid plaques. www.nature.Scientific Reports DOI:10.1038/srep16658 Dovizio M, Alberti S, Sacco A, Guillem-Llobat P, Schiavone S, Maier TJ, Steinhilber D, Patrignani P, 2015, Novel insights in the regulation of cyclooxygenase expression by platelet-cancer cell cross-talk. Biochem Soc Trans 43:707–714 Robert B, Legrand Y, Pignaud G, Caen J, Robert L, 1969, Activite elastolytique associee aux plaquettes sanguines. Pathol Biol 17: 615–622 Robert B, Szigeti M, Robert L, Legrand Y, Pignaud G, Caen J, 1970, Release of elastolytic activity from blood platelets. Nature 227:1248–1124 Legrand Y, Robert B, Szigeti M, Pignaud G, Caen J, Robert L, 1970, Etudes sur une protease elastinolytique des plaquettes sanguines humaines. Atherosclerosis 12:451–465 Robert B, Robert L, Legrand Y, Pignaud G, Caen J, 1971, Elastolytic protease in blood platelets. Ser Haemat 4:175–185 Robert B, Legrand Y, Soria C, Caen J, Robert L, 1972, Etude sur les proteases des plaquettes sanguines humaines: purification d’une elastase. C R Acad Sci 274:1749–1752 Robert B, Derouette J-C, Robert L, 1974, Mise en evidence d’une protease a activite elastolytique dans les extraits d’aortes humaines et animales. CR Acad Sci 278:3251–3254 Legrand Y, Robert B, 1975, Purification and characterization of human blood platelet elastase. In: Peeters H, ed, Protides of the biological fluids. Pergamon Press, Oxford, New York 22:419–424 Legrand Y, Pignaud G, Caen J, Robert B, Robert L, 1975, Separation of human blood platelet elastase and proelastase by affinity chromatography. Biochem Biophys Res Comm 63:224–231 Legrand Y, Caen J, Robert L, Wautier JL, 1977, Platelet elastase and leucocyte elastase are two different entities. Thrombosis Haemostasis 37:580–582 Robert L, Derouette J-C, Ordinas A, HornebeckW(1978, Plateletselastin interaction. In: Biochimie des tissus conjonctifs normaux et pathologiques. 6th Coll. Fed. Eur. Connective Tissue Clubs. CNRS, French National Research Center, Paris, France, n°287. I:155–156 Hornebeck W, Starkey PM, Gordon JL, Legrand Y, Pignaud G, Robert L, Caen J, Ehrlich HP, Barret A, 1980, Elastase-like enzyme of platelets. Letter to the editor. Thromb Haemost 42:1681–1683 Hornebeck W, Pignaud G, Legrand Y, Robert L, 1984, Differentiation of the elastase-type protease of platelets from other elastases. Clin Physiol Biochem 2:166–175 Hornebeck W, Legrand Y, 1980, Possible implication of two elastinolytic proteases isolated from tissue aorta and blood platelets in atherosclerosis. In: Robert AM, Robert L, eds, Frontiers in matrix biology. Karger, Basel 8:199–212 Hornebeck W, Legrand Y, 1989, Human platelet-derived elastase, EC 3.4.21). In: Robert L, HornebeckW(eds, Elastin and Elastases, vol 2. CRC Press, Boca Raton, pp 39–48 Nachman RL, Rafii S, 2008, Platelets, Petechiae, and preservation of the Vascular Wall. New England J Med 359:1261–1270 Bieth JG, 1989, Human neutrophil elastase. In: Robert L, Hornebeck W, eds, Elastin and elastases, Elastases, vol II. CRC Press, Boca Raton, pp 23–31 James HL, Wachtfogel PL, Zimmerman M, Colman RW, Chen AB, 1985)Aunique elastase in human platelets. JClin Invest 76:2330–2337 Hirsh J, Brain EA, 1983, Hemostasis & Thrombosis. A conceptual approach, Second Edition, Churchill Livingstone, Edinburgh, UK Robert L, Labat-Robert J, Hornebeck W, 1986, Aging and atherosclerosis. In: Gotto AM, Paoletti R, eds, Atheroclerosis rev, vol 14. Raven Press, New York Jacotot B, Coordinator, Clementy J, Emmerich J, Fruchart J-C, Gosse P, Guillo P, Robert L, eds,, 1993, Atherosclerose. Sandoz, Rueil-Malmaison Parks W, Mecham RP, eds,, 1998, Matrix Metalloproteinases. Academic Press, London, New York Hayflick L, Moorhead PS, 1961, The serial cultivation of human diploid cell strains. Exp Cell Res 37:585–621 Archilla-Marcos M, Robert L, 1993, Control of the biosynthesis and excretion of the elastase-type protease of human skin fibroblasts by the elastin receptor. Clin Physiol Biochem 10:86–91 Bernard E, HornebeckW, Robert L, 1994, Effect of hyaluronan on the elastase-type activity of human skin fibroblasts. Cell Biol Internat 18:967–971 Northrop JH, 1939, Crystalline Enzymes. Oxford University Press, London, UK Balo J, 1963, Connective tissue changes in atherosclerosis. In: Hall DA, ed, International review of connective tissue research, vol I. Academic press, New York, London, pp 241–306 Robert AM, Robert L, eds,, 1980, Biology and pathology of elastic tissues frontiers of matrix biology vol. 8, Karger, Basel Robert L, 2010, The Saga of κ-elastin or the promotion of elastin degradation products from "garbage" to receptor agonists and pharmacologically active principles. Conn Tissue Res 51:8–13 Robert L, Hornebeck W, eds,, 1989, Elastases. In: Elastin and Elastases, vol. II. CR C Press, Boca Raton, USA Chadwick DJ, Goode JA, eds,, 1995, Ciba foundation symposium 192. TheMolecular Biology and Pathology of Elastic Tissues. John Wiley & Sons, Chichester Robert AM, Grosgogeat Y, Reverdy V, Robert B, Robert L, 1971, Lesions arterielles produites chez le lapin par immunisation avec l’elastine et les glycoproteins de structure de l’aorte. Atherosclerosis 13:427–449 Fulop T, Wei SM, Robert L, Jacob M-P, 1990, Determination of elastin peptides in normal and atherosclerotic human sera by ELISA. Clin Physiol Biochem 8:273–282 Bizbiz L, Alperovitch A, Robert L and the EVA group, 1997, Aging of the vascular wall. Serum concentration of elastin peptides and elastase inhibitors in relation with cardiovascular risk factor The EVA study Atherosclerosis 131:73–78 Robert L, Robert AM, Jacotot B, 1998, Elastin-elastaseatherosclerosis revisited. Atherosclerosis 140:281–295 Kinlough-Rathbone RL, Mustard JF, 1987, Platelet interactions with components of the arterial wall. In: Olsson AG, ed, Atherosclerosis. Churchill Livingstone, Biology and Clinical Science Jolles G, Legrand YJ, Nurden A, eds,, 1986, Biology and Pathology of Platelet-Vessel Wall Interactions. Proceedings of the Rhone-Poulenc Sante – INSERM Conference, 30.09. to 02.10.1985. Academic Press, New York Robert L, Labat-Robert J, 1995, Extracellular matrix. In: Molecular basis of aging. chapter 15, CRC Press, Boca Raton, Florida, USA, pp 459–492 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 455 EP 458 DI 10.1007/s12253-017-0199-1 PG 4 ER PT J AU Kireeva, G Bespalov, GV Belyaeva, AO Senchik, K Stukov, NA Gafton, G Guseynov, K Belyaev, A AF Kireeva, S. Galina Bespalov, Grigorievich Vladimir Belyaeva, Alexandrovna Olesya Senchik, Y. Konstantin Stukov, Nikolayevich Aleksandr Gafton, I. Georgiy Guseynov, D. Konstantin Belyaev, M. Alexey TI Normothermic and Hyperthermic Intraperitoneal Chemoperfusions with Cisplatin to Treat Advanced Ovarian Cancer in Experimental Settings SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Kireeva, S. Galina] Russian Ministry of Health, N. N. Petrov Research Institute of Oncology, Pesochny, Leningradskaya str., 68, 197758 Saint-Petersburg, Russian Federation. [Bespalov, Grigorievich Vladimir] Russian Ministry of Health, N. N. Petrov Research Institute of Oncology, Pesochny, Leningradskaya str., 68, 197758 Saint-Petersburg, Russian Federation. [Belyaeva, Alexandrovna Olesya] Russian Ministry of Health, N. N. Petrov Research Institute of Oncology, Pesochny, Leningradskaya str., 68, 197758 Saint-Petersburg, Russian Federation. [Senchik, Y. Konstantin] Russian Ministry of Health, N. N. Petrov Research Institute of Oncology, Pesochny, Leningradskaya str., 68, 197758 Saint-Petersburg, Russian Federation. [Stukov, Nikolayevich Aleksandr] Russian Ministry of Health, N. N. Petrov Research Institute of Oncology, Pesochny, Leningradskaya str., 68, 197758 Saint-Petersburg, Russian Federation. [Gafton, I. Georgiy] Russian Ministry of Health, N. N. Petrov Research Institute of Oncology, Pesochny, Leningradskaya str., 68, 197758 Saint-Petersburg, Russian Federation. [Guseynov, D. Konstantin] Russian Ministry of Health, N. N. Petrov Research Institute of Oncology, Pesochny, Leningradskaya str., 68, 197758 Saint-Petersburg, Russian Federation. [Belyaev, M. Alexey] Russian Ministry of Health, N. N. Petrov Research Institute of Oncology, Pesochny, Leningradskaya str., 68, 197758 Saint-Petersburg, Russian Federation. RP Kireeva, G (reprint author), Russian Ministry of Health, N. N. Petrov Research Institute of Oncology, 197758 Saint-Petersburg, Russian Federation. EM galinakireyeva@mail.ru CR Sugarbaker PH, 2005, Technical handbook for the integration of cytoreductive surgery and perioperative intraperitoneal chemotherapy into the surgical management of gastrointestinal and gynecologic malignancy, 4th edn. The Ludann, Michigan Gremonprez F,WillaertW, CeelenW(2014, Intraperitoneal chemotherapy, IPC, for peritoneal carcinomatosis: review of animal models. J Surg Oncol 109:110–116., DOI 10.1002/jso.23464 Sugarbaker PH, 2003, Peritonectomy procedures. Surg Oncol Clin N Am12:703–727 Zeamari S, Floot B, Van der Vange N, Stewart FA, 2003, Pharmacokinetics and pharmacodynamics of cisplatin after intraoperative hyperthermic intraperitoneal chemoperfusion, HIPEC). Anticancer Res 23:1643–1648 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 459 EP 460 DI 10.1007/s12253-017-0202-x PG 2 ER PT J AU Liu, P Zu, X Liu, L AF Liu, Peihua Zu, Xiongbing Liu, Longfei TI Erratum to: Genetic Screening in Pheochromocytoma/ Paraganglioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Erratum to: Pathol. Oncol. Res. (2017) 23:217-217 DOI 10.1007/s12253-016-0117-y The original article contained an error. The corrected Acknowledgements section is shown below. Acknowledgements: This study was supported by the National Natural Science Foundation of China (No.81400773) and Natural Science Foundation of China Hunan Province (No.14JJ6002). C1 [Liu, Peihua] Central South University, Xiangya hospital, Department of Urology, 410008 Changsha, Hunan, China. [Zu, Xiongbing] Central South University, Xiangya hospital, Department of Urology, 410008 Changsha, Hunan, China. [Liu, Longfei] Central South University, Xiangya hospital, Department of Urology, 410008 Changsha, Hunan, China. RP Liu, L (reprint author), Central South University, Xiangya hospital, Department of Urology, 410008 Changsha, China. EM longfei_liu@163.com NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2017 VL 23 IS 2 BP 461 EP 461 DI 10.1007/s12253-017-0210-x PG 1 ER PT J AU Ding, AV Zhu, Z Mantz, AA Xiao, H Wakefield, RM Bai, Q Fang, Y AF Ding, A Vivi Zhu, Ziwen Mantz, A Alyse Xiao, Huaping Wakefield, R Mark Bai, Qian Fang, Yujiang TI The Role of IL-37 in Non-Cancerous Diseases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE IL-37; Non-cancerous diseases ID IL-37; Non-cancerous diseases AB IL-37 is a newly discovered cytokine belonging to IL-1 family consisting of 11 members, which have similar β- barrel structures and associate with Ig-like receptors. Extensive studies have been done with IL-37 since its discovery. These studies suggest that IL-37 does not only play a role in tumorigenesis, but also has anti-inflammatory properties in immune responses through the down regulation of proinflammatory molecules. We have previously reviewed the role of IL-37 in cancer. Here, we will focus on the role of IL-37 in non-cancerous Diseases. Such a study might be helpful to design new strategies to treat IL-37 associated diseases. C1 [Ding, A Vivi] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. [Zhu, Ziwen] University of Missouri, School of Medicine, Department of Surgery, 65212 Columbia, MO, USA. [Mantz, A Alyse] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. [Xiao, Huaping] The Affiliated Hospital of Xiangnan UniversityChenzhou, Hunan, China. [Wakefield, R Mark] University of Missouri, School of Medicine, Department of Surgery, 65212 Columbia, MO, USA. [Bai, Qian] University of Missouri, School of Medicine, Department of Surgery, 65212 Columbia, MO, USA. [Fang, Yujiang] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. RP Fang, Y (reprint author), Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, USA. EM yujiang.fang@dmu.edu CR Dinarello CA, 2013, Overview of the interleukin-1 family of ligands and receptors. In: Seminars in immunology, vol 6. Elsevier, pp 389–393 Quirk S,Agrawal DK(2014, Immunobiology of IL-37: mechanism of action and clinical perspectives. Expert Rev Clin Immunol 10(12):1703–1709 Boraschi D, Lucchesi D, Hainzl S, Leitner M, Maier E, Mangelberger D, Oostingh GJ, Pfaller T, Pixner C, Posselt G, 2011, IL-37: a new anti-inflammatory cytokine of the IL-1 family. Eur Cytokine Netw 22(3):127–147 Dinarello C, Arend W, Sims J, Smith D, Blumberg H, O'Neill L, Goldbach-Mansky R, Pizarro T, Hoffman H, Bufler P, 2010, IL-1 family nomenclature. Nat Immunol 11(11):973–973 Van de Veerdonk FL, Netea MG, 2013, New insights in the immunobiology of IL-1 family members. Front Immunol 4(167): 1–11 Nold MF, Nold-Petry CA, Zepp JA, Palmer BE, Bufler P, Dinarello CA, 2010, IL-37 is a fundamental inhibitor of innate immunity. Nat Immunol 11(11):1014–1022 Busfield S, Comrack C, Yu G, Chickering T, Smutko J, Zhou H, Leiby K, Holmgren L, Gearing D, Pan Y, 2000, Identification and gene organization of three novel members of the IL-1 family on human chromosome 2. Genomics 66(2):213–216 Kumar S, McDonnell PC, Lehr R, Tierney L, Tzimas MN, Griswold DE, Capper EA, Tal-Singer R, Wells GI, Doyle ML, 2000, Identification and initial characterization of four novel members of the interleukin-1 family. J Biol Chem 275(14):10308– 10314 Pan G, Risser P, Mao W, Baldwin DT, Zhong AW, Filvaroff E, Yansura D, Lewis L, Eigenbrot C, Henzel WJ, 2001, IL-1H, an interleukin 1-related protein that binds IL-18 receptor/IL-1Rrp. Cytokine 13(1):1–7 Smith DE, Renshaw BR, Ketchem RR, Kubin M, Garka KE, Sims JE, 2000, Four newmembers expand the interleukin-1 superfamily. J Biol Chem 275(2):1169–1175 Taylor SL, Renshaw BR, Garka KE, Smith DE, Sims JE, 2002, Genomic organization of the interleukin-1 locus. Genomics 79(5): 726–733 Kumar S, Hanning CR, Brigham-Burke MR, Rieman DJ, Lehr R, Khandekar S, Kirkpatrick RB, Scott GF, Lee JC, Lynch FJ, 2002, Interleukin-1F7B, IL-1H4/IL-1F7, is processed by caspase-1 and mature IL-1F7B binds to the IL-18 receptor but does not induce IFN-γ production. Cytokine 18(2):61–71 Luo Y, Cai X, Liu S, Wang S, Nold-Petry CA, Nold MF, Bufler P, Norris DA, Dinarello CA, Fujita M, 2013, 171: IL-37 suppresses contact hypersensitivity by inducing tolerogenic dendritic cells. Cytokine 63(3):283 GaoW, Kumar S, Lotze MT, Hanning C, Robbins PD, Gambotto A, 2003, Innate immunity mediated by the cytokine IL-1 homologue 4, IL-1H4/IL-1F7, induces IL-12-dependent adaptive and profound antitumor immunity. J Immunol 170(1):107–113 Ding VA, Zhu Z, Xiao H, Wakefield MR, Bai Q, Fang Y, 2016, The role of IL-37 in cancer. Med Oncol 33(7):1–7 Bufler P, Gamboni-Robertson F, Tania A, Soo-Hyun K, Dinarello CA, 2004, Interleukin-1 homologues IL-1F7b and IL-18 contain functional mRNA instability elements within the coding region responsive to lipopolysaccharide. Biochem J 381(2):503–510 Bulau A-M, Nold MF, Li S, Nold-Petry CA, Fink M, Mansell A, Schwerd T, Hong J, Rubartelli A, Dinarello CA, 2014, Role of caspase-1 in nuclear translocation of IL-37, release of the cytokine, and IL-37 inhibition of innate immune responses. Proc Natl Acad Sci 111(7):2650–2655 Martinon F, Tschopp J, 2007, Inflammatory caspases and inflammasomes: master switches of inflammation. Cell Death Differ 14(1):10–22 Bufler P, Azam T, Gamboni-Robertson F, Reznikov LL, Kumar S, Dinarello CA, Kim S-H, 2002, A complex of the IL-1 homologue IL-1F7b and IL-18-binding protein reduces IL-18 activity. Proc Natl Acad Sci 99(21):13723–13728 Dinarello CA, 1998, Interleukin-1, interleukin-1 receptors and interleukin-1 receptor antagonist. Int Rev Immunol 16(5–6):457–499 Novick D, Kim S-H, Fantuzzi G, Reznikov LL, Dinarello CA, Rubinstein M, 1999, Interleukin-18 binding protein: a novel modulator of the Th1 cytokine response. Immunity 10(1):127–136 Akira S, 2000, The role of IL-18 in innate immunity. Curr Opin Immunol 12(1):59–63 Wald D, Qin J, Zhao Z, Qian Y, Naramura M, Tian L, Towne J, Sims JE, Stark GR, Li X, 2003, SIGIRR, a negative regulator of toll-like receptor–interleukin 1 receptor signaling. Nat Immunol 4(9):920–927 Xiao H, Gulen MF, Qin J, Yao J, Bulek K, Kish D, Altuntas CZ, Wald D, Ma C, Zhou H, 2007, The toll–interleukin-1 receptor member SIGIRR regulates colonic epithelial homeostasis, inflammation, and tumorigenesis. Immunity 26(4):461–475 Magne D, Palmer G, Barton JL, Mezin F, Talabot-Ayer D, Bas S, Duffy T, Noger M, Guerne P-A, Nicklin MJ, 2006, The new IL-1 family member IL-1F8 stimulates production of inflammatory mediators by synovial fibroblasts and articular chondrocytes. Arthritis Res Ther 8(3):R80 Nold-Petry CA, Lo CY, Rudloff I, Elgass KD, Li S, Gantier MP, Lotz-Havla AS, Gersting SW, Cho SX, Lao JC, 2015, IL-37 requires the receptors IL-18R [alpha] and IL-1R8, SIGIRR, to carry out its multifaceted anti-inflammatory program upon innate signal transduction. Nat Immunol 16(4):354–365 Li S, Neff CP, Barber K, Hong J, Luo Y, Azam T, Palmer BE, Fujita M, Garlanda C, Mantovani A, 2015, Extracellular forms of IL-37 inhibit innate inflammation in vitro and in vivo but require the IL-1 family decoy receptor IL-1R8. Proc Natl Acad Sci 112(8):2497–2502 Sharma S, Kulk N, Nold MF, Graf R, Kim S-H, Reinhardt D, Dinarello CA, Bufler P, 2008, The IL-1 family member 7b translocates to the nucleus and down-regulates proinflammatory cytokines. J Immunol 180(8):5477–5482 Bulau A-M, Gruschka A, Schwaiger R, Dinarello CA, Bufler P, 2013, 30: anti-IL-37 antibody abrogates the protection in IL-37 transgenic mice to LPS-induced septic shock. Cytokine 63(3):250 Grimsby S, Jaensson H, Dubrovska A, Lomnytska M, Hellman U, Souchelnytskyi S, 2004, Proteomics-based identification of proteins interacting with Smad3: SREBP-2 forms a complex with Smad3 and inhibits its transcriptional activity. FEBS Lett 577(1): 93–100 Bouma G, Strober W, 2003, The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol 3(7):521–533 McNamee EN, Masterson JC, Jedlicka P, McManus M, Grenz A, Collins CB, NoldMF, Nold-Petry C, Bufler P, Dinarello CA, 2011, Interleukin 37 expression protects mice fromcolitis. ProcNatl Acad Sci 108(40):16711–16716 Mayer L, 2010, Evolving paradigms in the pathogenesis of IBD. J Gastroenterol 45(1):9–16 Imaeda H, Takahashi K, Fujimoto T, Kasumi E, Ban H, Bamba S, Sonoda H, Shimizu T, Fujiyama Y, Andoh A, 2013, Epithelial expression of interleukin-37b in inflammatory bowel disease. Clin Exp Immunol 172(3):410–416 Crispin JC, Liossis S-NC, Kis-Toth K, Lieberman LA, Kyttaris VC, Juang Y-T, Tsokos GC, 2010, Pathogenesis of human systemic lupus erythematosus: recent advances. Trends Mol Med 16(2):47–57 Jacob N, Stohl W, 2011, Cytokine disturbances in systemic lupus erythematosus. Arthritis Res Ther 13(4):228 Su D-L, Lu Z-M, Shen M-N, Li X, Sun, L-Y, 2012, Roles of proand anti-inflammatory cytokines in the pathogenesis of SLE. Biomed Res Int Bezalel S, Asher I, Elbirt D, Sthoeger ZM, 2012, Novel biological treatments for systemic lupus erythematosus: current and future modalities. Isr Med Assoc J 14(8):508–514 Song L, Qiu F, Fan Y, Ding F, Liu H, Shu Q, Liu W, Li X, 2013, Glucocorticoid regulates interleukin-37 in systemic lupus erythematosus. J Clin Immunol 33(1):111–117 Ye L, Ji L,Wen Z, ZhouY,HuD, LiY,Yu T, Chen B, Zhang J, Ding L, 2014, IL-37 inhibits the production of inflammatory cytokines in peripheral blood mononuclear cells of patients with systemic lupus erythematosus: its correlationwith disease activity. J Transl Med 12:69 Li C, Zhao P, Sun X, Che Y, Jiang, Y, 2013, Elevated levels of cerebrospinal fluid and plasma interleukin-37 in patients with Guillain-Barre syndrome. Mediat Inflamm Xia T, Zheng, X-f, Qian, B-h, Fang, H,Wang, J-j, Zhang, L-l, Pang, Y-f, Zhang, J, Wei, X-q, Xia, Z-f, 2015, plasma interleukin-37 is elevated in patients with rheumatoid arthritis: its correlation with disease activity and Th1/Th2/Th17-related cytokines. Dis Markers Yang L, Zhang J, Tao J, Lu T, 2015, Elevated serum levels of interleukin-37 are associated with inflammatory cytokines and disease activity in rheumatoid arthritis. APMIS 123(12):1025–1031 Ho T, Griffin J, 1999, Guillain-Barre syndrome. Curr Opin Neurol 12(4):389–394 Brennan FM, McInnes IB, 2008, Evidence that cytokines play a role in rheumatoid arthritis. J Clin Invest 118(11):3537–3545 Feldmann M, Maini R, 1999, The role of cytokines in the pathogenesis of rheumatoid arthritis. Rheumatology, Oxford, England, 38:3–7 Ye L, Huang Z, 2015, IL-37 restrains autoimmune diseases. Oncotarget 6(26):21775 Moschen AR, Molnar C, Enrich B, Geiger S, Ebenbichler CF, Tilg H, 2011, Adipose and liver expression of interleukin, IL)-1 family members in morbid obesity and effects of weight loss. Mol Med 17(7):840 Ballak DB, Van Diepen JA, Moschen AR, Jansen HJ, Hijmans A, Groenhof G-J, Leenders F, Bufler P, Boekschoten MV, Muller M Kersten S, Li S, et al., 2014, IL-37 protects against obesity-induced inflammation and insulin resistance. Nature communications 5: 4711 Bulau A-M, Fink M, Maucksch C, Kappler R, Mayr D,Wagner K, Bufler P, 2011, In vivo expression of interleukin-37 reduces local and systemic inflammation in concanavalin A-induced hepatitis. Sci World J 11:2480–2490 Sakai N, Van Sweringen HL, Belizaire RM, Quillin RC, Schuster R, Blanchard J, Burns JM, Tevar AD, Edwards MJ, Lentsch AB, 2012, Interleukin-37 reduces liver inflammatory injury via effects on hepatocytes and non-parenchymal cells. J Gastroenterol Hepatol 27(10):1609–1616 Colletti L, Remick D, Burtch G, Kunkel S, Strieter R, Campbell D Jr, 1990, Role of tumor necrosis factor-alpha in the pathophysiologic alterations after hepatic ischemia/reperfusion injury in the rat. J Clin Investig 85(6):1936 Husted T, Blanchard J, Schuster R, Shen H, Lentsch A, 2006, Potential role for IL-23 in hepatic ischemia/reperfusion injury. Inflamm Res 55(5):177–178 Lentsch AB, Yoshidome H, Cheadle WG, Miller FN, Edwards MJ, 1998, Chemokine involvement in hepatic ischemia/reperfusion injury in mice: roles for macrophage inflammatory protein-2 and KC. Hepatology 27(4):1172–1177 Lentsch AB, Yoshidome H, Kato A, Warner RL, Cheadle WG, Ward PA, Edwards MJ, 1999, Requirement for interleukin-12 in the pathogenesis of warm hepatic ischemia/reperfusion injury in mice. Hepatology 30(6):1448–1453 Suzuki S, Toledo-Pereyra LH, 1994, Interleukin 1 and tumor necrosis factor production as the initial stimulants of liver ischemia and reperfusion injury. J Surg Res 57(2):253–258 Wanner GA, Ertel W, Muller P, Hofer Y, Leiderer R, Menger MD, Messmer K, 1996, Liver ischemia and reperfusion induces a systemic inflammatory response through Kupffer cell activation. Shock 5(1):34–40 Jaeschke H, 2006, Mechanisms of liver injury. II Mechanisms of neutrophil-induced liver cell injury during hepatic ischemiareperfusion and other acute inflammatory conditions. Am J Physiol Gastrointest Liver Physiol 290(6):G1083–G1088 Jaeschke H, FarhoodA, 1991, Neutrophil and Kupffer cell-induced oxidant stress and ischemia-reperfusion injury in rat liver. Am J Physiol Gastrointest Liver Physiol 260(3):G355–G362 Li C, Ji H, Cai Y, Ayana DA, Lv P, Liu M, Jiang Y, 2013, Serum interleukin-37 concentrations and HBeAg seroconversion in chronic HBV patients during telbivudine treatment. J Interf Cytokine Res 33(10):612–618 Wu B-w, Q-t Z, Meng K, Q-w J, 2013, The potential role of IL-37 in atherosclerosis. Die Pharmazie-An Int J Pharm Sci 68(11):857–860 Ji Q, Zeng Q, Huang Y, Shi Y, Lin Y, Lu Z, Meng K,Wu B, Yu K, Chai, M, 2014, Elevated plasma IL-37, IL-18, and IL-18BP concentrations in patients with acute coronary syndrome. Mediat Inflamm Wu B, Meng K, Ji Q, Cheng M, Yu K, Zhao X, Tony H, Liu Y, Zhou Y, Chang C, 2014, Interleukin-37 ameliorates myocardial ischaemia/reperfusion injury in mice. Clin Exp Immunol 176(3): 438–451 Hojen JF, Rasmussen TA, Andersen KLD, Winckelmann AA, Laursen RR, Gunst JD, Moller HJ, Fujita M, Ostergaard L, Sogaard OS, 2015, Interleukin-37 expression is increased in chronic HIV-1-infected individuals and is associated with inflammation and the size of the total viral reservoir. Mol Med 21(1):337 Coll-Miro M, Francos-Quijorna I, Santos-Nogueira E, Torres-Espin A, Bufler P, Dinarello CA, Lopez-Vales R, 2016, Beneficial effects of IL-37 after spinal cord injury in mice. Proceedings of the National Academy of Sciences:201523212 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 463 EP 470 DI 10.1007/s12253-016-0137-7 PG 8 ER PT J AU Lee, HJ Yoo, JN Kim, SM Lee, HS AF Lee, Hwa Joo Yoo, Jin Nam Kim, Sung Min Lee, Hyung Sug TI Histone Demethylase Gene PHF2 Is Mutated in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PHF2; Tumor suppressor gene; Frameshift mutation; Cancer; Microsatellite instability; Intratumoral heterogeneity ID PHF2; Tumor suppressor gene; Frameshift mutation; Cancer; Microsatellite instability; Intratumoral heterogeneity AB Alterations of genes involved in histone modification are common in cancers. A histone demethylase-encoding gene PHF2 is considered a putative tumor suppressor gene (TSG). PHF2 is essential for p53-mediated TSG functions such as chemotherapy-mediated cancer cell killing. However, inactivating mutations of PHF2 that could inactivate its functions are not reported in cancers. In a genome database, we observed that the PHF2 gene possessed mononucleotide repeats, which could be mutated in cancers with high microsatellite instability (MSI-H). For this, we analyzed 124 colorectal cancers (CRCs) and 79 gastric (GCs) cancers for the mutations and their intratumoral heterogeneity (ITH). Twenty-two of 79 CRCs (27.8 %) and 7 of 34 GCs (20.6 %) harboring MSI-H exhibited frameshift mutations. However, we found no such mutations in microsatellite stable/low MSI (MSS/MSI-L) cancers. Also, we studied ITH for the detected frameshift mutations in 16 cases of CRCs and detected ITH in two (12.5 %) cases. Our data reveal that TSG gene PHF2 harbors mutational ITH as well as the frameshift mutations in CRC and GC with MSI-H. Based on this, it is suggested that frameshift mutations of PHF2 may play a role in tumorigenesis through its TSG inactivation in CRC and GC. C1 [Lee, Hwa Joo] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Jenuwein T, Allis CD, 2011, Translating the histone code. Science 293:1074–1080 Albert M, Helin K, 2010, Histone methyltransferases in cancer. Semin Cell Dev Biol 21:209–220 Bannister AJ, Schneider R, Kouzarides T, 2002, Histone methylation: dynamic or static? Cell 109:801–806 Hess JL, 2004, MLL: a histone methyltransferase disrupted in leukemia. Trends Mol Med 10:500–507 Morin RD, Johnson NA, Severson TM, Mungall AJ, An J, Goya R, Paul JE, Boyle M, Woolcock BW, Kuchenbauer F, Yap D, Humphries RK, Griffith OL, Shah S, Zhu H, Kimbara M, Shashkin P, Charlot JF, Tcherpakov M, Corbett R, Tam A, Varhol R, Smailus D, Moksa M, Zhao Y, Delaney A, Qian H, Birol I, Schein J, Moore R, Holt R, Horsman DE, Connors JM, Jones S, Aparicio S, Hirst M, Gascoyne RD, Marra MA, 2010, Somatic mutations altering EZH2, Tyr641, in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat Genet 42:181– 185 van Haaften G, Dalgliesh GL, Davies H, Chen L, Bignell G, Greenman C, Edkins S, Hardy C, O'Meara S, Teague J, Butler A, Hinton J, Latimer C, Andrews J, Barthorpe S, Beare D, Buck G, Campbell PJ, Cole J, Forbes S, Jia M, Jones D, Kok CY, Leroy C, LinML, McBrideDJ, Maddison M, Maquire S,McLayK, Menzies A, Mironenko T, Mulderrig L, Mudie L, Pleasance E, Shepherd R, Smith R, Stebbings L, Stephens P, Tang G, Tarpey PS, Turner R, Turrell K, Varian J,West S,Widaa S,Wray P, Collins VP, Ichimura K, Law S,Wong J, Yuen ST, Leung SY, Tonon G, DePinho RA, Tai YT, Anderson KC, Kahnoski RJ, Massie A, Khoo SK, Teh BT, Stratton MR, Futreal PA, 2009, Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet 41: 521–523 GL D, Furge K, Greenman C, Chen L, Bignell G, Butler A, Davies H, Edkins S, Hardy C, Latimer C, Teague J, Andrews J, Barthorpe S, Beare D, Buck G, PJ C, Forbes S, Jia M, Jones D, Knott H, Kok CY, Lau KW, Leroy C, Lin ML, McBride DJ, Maddison M, Maguire S, McLay K, Menzies A, Mironenko T, Mulderrig L, Mudie L, O'Meara S, Pleasance E, Rajasingham A, Shepherd R, Smith R, Stebbings L, Stephens P, Tang G, Tarpey PS, Turrell K, Dykema KJ, Khoo SK, Petillo D, Wondergem B, Anema J, Kahnoski RJ, Teh BT, Stratton MR, Futreal PA, 2010, Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463:360–363 Hasenpusch-Theil K, Chadwick BP, Theil T, Heath SK, Wilkinson DG, Frischauf AM, 1999, PHF2, a novel PHD finger gene located on human chromosome 9q22. Mamm Genome 10:294–298 Sinha S, Singh RK, Alam N, Roy A, Roychoudhury S, Panda CK, 2008, Alterations in candidate genes PHF2, FANCC, PTCH1 and XPA at chromosomal 9q22.3 region: pathological significance in early- and late-onset breast carcinoma. Mol Cancer 7:84 Ghosh A, Ghosh S, Maiti GP, Mukherjee S, Mukherjee N, Chakraborty J, Roy A, Roychoudhury S, Panda CK, 2012, Association of FANCC and PTCH1 with the development of early dysplastic lesions of the head and neck. Ann Surg Oncol 19(Suppl 3):S528–S538 Lee KH, Park JW, Sung HS, Choi YJ, Kim WH, Lee HS, Chung HJ, Shin HW, Cho CH, Kim TY, Li SH, Youn HD, Kim SJ, Chun YS, 2015, PHF2 histone demethylase acts as a tumor suppressor in association with p53 in cancer. Oncogene 34:2897–2909 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 McGranahan N, Swanton C, 2015, Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 27:15–26 Kim TM, Jung SH, Baek IP, Lee SH, Choi YJ, Lee JY, Chung YJ, Lee SH, 2014, Regional biases in mutation screening due to intratumoural heterogeneity of prostate cancer. J Pathol 233:425– 435 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8: 305–311 KimMS, Yoo NJ, Lee SH, 2015, Preferential occurrence of RHOA mutation in gastric and colorectal cancers. Pathology 47:598–599 Jo YS, Kim MS, Yoo NJ, Lee SH, 2016, NSD1 encoding a histone methyltransferase exhibits frameshift mutations in colorectal cancers. Pathology 48:284–286 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 Wiesner GL, Daley D, Lewis S, Ticknor C, Platzer P, Lutterbaugh J, MacMillen M, Baliner B, Willis J, Elston RC, Markowitz SD, 2003, A subset of familial colorectal neoplasia kindreds linked to chromosome 9q22.2-31.2. Proc Natl Acad Sci U S A 100:12961– 12965 Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, Meyerson M, Gabriel SB, Lander ES, Getz G, 2014, Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505:495–501 Calin GA, Gafa R, Tibiletti MG, Herlea V, Becheanu G, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression inmicrosatellite instability high, MSI-H, colon cancers correlates with clinico-pathological parameters: a study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89:230–235 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 471 EP 476 DI 10.1007/s12253-016-0130-1 PG 6 ER PT J AU Chang-Lee, NSh Hsu, HH Shibu, AM Ho, TJ Tsai, ChH Chen, MCh Tu, ChCh Viswanadha, PV Kuo, WW Huang, ChY AF Chang-Lee, Nu Shu Hsu, Hsi-Hsien Shibu, Asokan Marthandam Ho, Tsung-Jung Tsai, Chih-Hao Chen, Ming-Cheng Tu, Chuan-Chou Viswanadha, Padma Vijaya Kuo, Wei-Wen Huang, Chih-Yang TI E2/ERβ Inhibits PPARα to Regulate Cell-Proliferation and Enhance Apoptosis in Hep3B-Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Estrogen; Estrogen receptor; peroxisome proliferator-activated receptor; Hepatocellular carcinoma ID Estrogen; Estrogen receptor; peroxisome proliferator-activated receptor; Hepatocellular carcinoma AB Peroxisome proliferator-activated receptor-α (PPARα) is a member of the nuclear receptor superfamily involved in hepatocarcinogenesis in rodents. In previous studies on liver tumor tissues, PPARα mRNA expression was found to be significantly higher and overexpression of ERα inhibited the PPARα expression, cell-proliferation and also induced apoptosis in Hep3B cell. However, the role of ERβ is not known yet. Therefore, the aim of this study is to define the role of ERβ on PPARα in Hep3B cells. The effect of PPARα signaling cascade were monitored by inducing Hep3B cells by fenofibrate. Further the cells were transfected with pCMV-ERβ and the consequences of ERβ-overexpression on the PPARα induced changes such as enhanced cell-proliferation and suppressed apoptosis were determined using western blot analysis and TUNEL assay. The EMSA was used to identify whether ERβ modulates PPARα expression by binding to PPARα promoter region to repress PPARα promoter activity. In addition, the direct interaction between ERβ and PPARα proteins was verified by co-immunoprecipitation assay. Our results show that the overexpressed ERβ not only attenuated the effects of fenofibrate to induce the levels of apoptosis protein such as Cyt.c, Caspase 9 and Caspase 3 but also inhibited the levels of survival protein such Bcl-xL, p-Bad, cyclin A and cyclin E. All these effects of E2/ERβ resulted in the enhancement of mitochondria dependent apoptotic pathway and the attenuation of cell proliferation. Moreover, the overexpressed ERβ reduced the mRNA and protein levels of PPARα and its downstream Acyl-CoA oxidase (ACO). EMSA results show that ERβ directly binds to PPRE and inhibit PPARα gene expression and according to immunoprecipitation assay ERβ also binds strongly with PPARα. The E2/ERβ further inhibited the fenofibrate-induced nuclear translocation of PPARα. Taken together, ERβ might directly downregulate PPARα gene expression and inhibit the nuclear translocation to suppress the proliferation and induce the apoptosis of Hep3B cells. C1 [Chang-Lee, Nu Shu] Asia University, Department of Healthcare Administration, 413 Taichung, Taiwan, Republic of China. [Hsu, Hsi-Hsien] Mackay Memorial Hospital, Division of Colorectal Surgery, 104 Taipei, Taiwan, Republic of China. [Shibu, Asokan Marthandam] China Medical University, Graduate Institute of Basic Medical Science, 404 Taichung, Taiwan, Republic of China. [Ho, Tsung-Jung] China Medical University, Graduate Institute of Chinese Medicine, 404 Taichung, Taiwan, Republic of China. [Tsai, Chih-Hao] China Medical University, Graduate Institute of Basic Medical Science, 404 Taichung, Taiwan, Republic of China. [Chen, Ming-Cheng] Taichung Veterans General Hospital, Division of Colorectal Surgery, 407 Taichung, Taiwan, Republic of China. [Tu, Chuan-Chou] Taichung Armed Force General Hospital, Department of Internal Medicine, Division of Chest Medicine, 411 Taichung, Taiwan, Republic of China. [Viswanadha, Padma Vijaya] Bharathiar University, Department of Biotechnology, 641046 Coimbatore, India. [Kuo, Wei-Wen] China Medical University, Department of Biological Science and Technology, 402 Taichung, Taiwan, Republic of China. [Huang, Chih-Yang] China Medical University, Graduate Institute of Basic Medical Science, 404 Taichung, Taiwan, Republic of China. RP Huang, ChY (reprint author), China Medical University, Graduate Institute of Basic Medical Science, 404 Taichung, Taiwan, Republic of China. EM cyhuang@mail.cmu.edu.tw CR Bruix J, Boix L, SalaM, Llovet JM, 2004, Focus on hepatocellular carcinoma. Cancer Cell 5(3):215–219 Reddy JK, Krishnakantha TP, 1975, Hepatic peroxisome proliferation: induction by two novel compounds structurally unrelated to clofibrate. Science 190(4216):787–789 Yeldandi AV, Rao MS, Reddy JK, 2000, Hydrogen peroxide generation in peroxisome proliferator-induced oncogenesis. Mutat Res 448(2):159–177 Wu CY, Huang HM, Cho DY, 2015, An acute bleeding metastatic spinal tumor from HCC causes an acute onset of cauda equina syndrome. Biomedicine 5(3):18., DOI 10.7603/s40681-015-0018-5 Misra P, Reddy JK, 2014, Peroxisome proliferator-activated receptoralpha activation and excess energy burning in hepatocarcinogenesis. Biochimie 98:63–74., DOI 10.1016/j.biochi.2013.11.011 Reddy JK, Azarnoff DL, Hignite CE, 1980, Hypolipidaemic hepatic peroxisome proliferators form a novel class of chemical carcinogens. Nature 283(5745):397–398 Akriviadis EA, Llovet JM, Efremidis SC, Shouval D, Canelo R, Ringe B, Meyers WC, 1998, Hepatocellular carcinoma. Br J Surg 85(10):1319–1331., DOI 10.1046/j.1365-2168.1998.00865.x Lin CY, Strom A, Li Kong S, Kietz S, Thomsen JS, Tee JB, Vega VB, Miller LD, Smeds J, Bergh J, Gustafsson JA, Liu ET, 2007, Inhibitory effects of estrogen receptor beta on specific hormoneresponsive gene expression and association with disease outcome in primary breast cancer. Breast Cancer Res BCR 9(2):R25., DOI 10.1186/bcr1667 Lazennec G, 2006, Estrogen receptor beta, a possible tumor suppressor involved in ovarian carcinogenesis. Cancer Lett 231(2): 151–157., DOI 10.1016/j.canlet.2005.01.021 Treeck O, Pfeiler G, Horn F, Federhofer B, Houlihan H, Vollmer A, Ortmann O, 2007, Novel estrogen receptor beta transcript variants identified in human breast cancer cells affect cell growth and apoptosis of COS-1 cells. Mol Cell Endocrinol 264(1–2):50–60., DOI 10.1016/j.mce.2006.10.003 Behrens D, Gill JH, Fichtner I, 2007, Loss of tumourigenicity of stably ERbeta-transfected MCF-7 breast cancer cells. Mol Cell Endocrinol 274(1–2):19–29., DOI 10.1016/j.mce.2007.05.012 Paruthiyil S, Parmar H, Kerekatte V, Cunha GR, Firestone GL, Leitman DC, 2004, Estrogen receptor beta inhibits human breast cancer cell proliferation and tumor formation by causing a G2 cell cycle arrest. Cancer Res 64(1):423–428 Strom A, Hartman J, Foster JS, Kietz S, Wimalasena J, Gustafsson JA, 2004, Estrogen receptor beta inhibits 17beta-estradiol-stimulated proliferation of the breast cancer cell line T47D. Proc Natl Acad Sci U S A 101(6):1566–1571., DOI 10.1073/pnas.0308319100 Carruba G, 2007, Estrogen and prostate cancer: an eclipsed truth in an androgen-dominated scenario. J Cell Biochem 102(4):899–911., DOI 10.1002/jcb.21529 StettnerM, Kaulfuss S, Burfeind P, Schweyer S, Strauss A, Ringert RH, Thelen P, 2007, The relevance of estrogen receptor-beta expression to the antiproliferative effects observed with histone deacetylase inhibitors and phytoestrogens in prostate cancer treatment. Mol Cancer Ther 6(10):2626–2633., DOI 10.1158/1535-7163. MCT-07-0197 Cheng J, Lee EJ, Madison LD, Lazennec G, 2004, Expression of estrogen receptor beta in prostate carcinoma cells inhibits invasion and proliferation and triggers apoptosis. FEBS Lett 566(1–3):169– 172., DOI 10.1016/j.febslet.2004.04.025 Chen GG, Zeng Q, Tse GM, 2008, Estrogen and its receptors in cancer. Med Res Rev 28(6):954–974., DOI 10.1002/med.20131 Jeong S, Yoon M, 2007, Inhibition of the actions of peroxisome proliferator-activated receptor alpha on obesity by estrogen. Obesity 15(6):1430–1440., DOI 10.1038/oby.2007.171 Huang EJ,Wu CC, Huang HP, Liu JY, Lin CS, Chang YZ, Lin JA, Lin JG, Chen LM, Lee SD, KuoWW, Huang CY, 2006, Apoptotic and anti-proliferative effects of 17beta-estradiol and 17betaestradiol- like compounds in the Hep3B cell line. Mol Cell Biochem 290(1–2):1–7., DOI 10.1007/s11010-005-9000-y Marx N, Duez H, Fruchart JC, Staels B, 2004, Peroxisome proliferator-activated receptors and atherogenesis: regulators of gene expression in vascular cells. Circ Res 94(9):1168–1178., DOI 10.1161/01.RES.0000127122.22685.0A Lee CH, Olson P, Evans RM, 2003, Minireview: lipid metabolism, metabolic diseases, and peroxisome proliferator-activated receptors. Endocrinology 144(6):2201–2207., DOI 10.1210/en.2003-0288 Lefebvre P, Chinetti G, Fruchart JC, Staels B, 2006, Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J Clin Invest 116(3):571–580., DOI 10.1172/JCI27989 Venalainen T, Molnar F, Oostenbrink C, Carlberg C, Perakyla M, 2010, Molecular mechanism of allosteric communication in the human PPARalpha-RXRalpha heterodimer. Proteins 78(4):873– 887., DOI 10.1002/prot.22613 Ringseis R, Gutgesell A, Dathe C, Brandsch C, Eder K, 2007, Feeding oxidized fat during pregnancy up-regulates expression of PPARalpha-responsive genes in the liver of rat fetuses. Lipids Health Dis 6:6., DOI 10.1186/1476-511X-6-6 Peters JM, Cheung C, Gonzalez FJ, 2005, Peroxisome proliferatoractivated receptor-alpha and liver cancer: where do we stand? J Mol Med 83(10):774–785., DOI 10.1007/s00109-005-0678-9 Nishimura J, Dewa Y, Okamura T, Jin M, Saegusa Y, Kawai M, Umemura T, Shibutani M, Mitsumori K, 2008, Role of Nrf2 and oxidative stress on fenofibrate-induced hepatocarcinogenesis in rats. Toxicological Sciences: an Official Journal of the Society of Toxicology 106(2):339–349., DOI 10.1093/toxsci/kfn174 Bisteau X, Caldez MJ, Kaldis P, 2014, The complex relationship between liver cancer and the cell cycle: a story of multiple regulations. Cancer 6(1):79–111., DOI 10.3390/cancers6010079 Slingerland J, PaganoM(2000, Regulation of the cdk inhibitor p27 and its deregulation in cancer. J Cell Physiol 183(1):10–17., DOI 10.1002/(SICI)1097-4652(200004)183:1<10::AID-JCP2>3.0. CO;2-I Yue X, Zhang Z, Liang X, Gao L, Zhang X, Zhao D, Liu X,Ma H, Guo M, Spear BT, Gong Y, Ma C, 2012, Zinc fingers and homeoboxes 2 inhibits hepatocellular carcinoma cell proliferation and represses expression of cyclins a and E. Gastroenterology 142(7): 1559–1570 ., DOI 10.1053/j.gastro.2012.02.049e1552 Youssef J, Badr M, 2011, Peroxisome proliferator-activated receptors and cancer: challenges and opportunities. Br J Pharmacol 164(1):68–82., DOI 10.1111/j.1476-5381.2011.01383.x Hasmall SC, James NH, Macdonald N, Gonzalez FJ, Peters JM, Roberts RA, 2000, Suppression of mouse hepatocyte apoptosis by peroxisome proliferators: role of PPARalpha and TNFalpha. Mutat Res 448(2):193–200 Gonzalez FJ, Peters JM, Cattley RC, 1998, Mechanism of action of the nongenotoxic peroxisome proliferators: role of the peroxisome proliferator-activator receptor alpha. J Natl Cancer Inst 90(22): 1702–1709 Nishimura J, Dewa Y, MugurumaM, Kuroiwa Y, Yasuno H, Shima T, Jin M, Takahashi M, Umemura T, Mitsumori K, 2007, Effect of fenofibrate on oxidative DNA damage and on gene expression related to cell proliferation and apoptosis in rats. Toxicological Sciences: an Official Journal of the Society of Toxicology 97(1): 44–54., DOI 10.1093/toxsci/kfm011 Shah YM, Morimura K, Yang Q, Tanabe T, Takagi M, Gonzalez FJ, 2007, Peroxisome proliferator-activated receptor alpha regulates a microRNA-mediated signaling cascade responsible for hepatocellular proliferation. Mol Cell Biol 27(12):4238–4247., DOI 10.1128/MCB.00317-07 Fidaleo M, 2009, Human health risk assessment for peroxisome proliferators: more than 30 years of research. Experimental and Toxicologic Pathology: Official Journal of the Gesellschaft fur Toxikologische Pathologie 61(3):215–221., DOI 10.1016/j. etp.2008.09.002 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 477 EP 485 DI 10.1007/s12253-016-0136-8 PG 9 ER PT J AU Sahin, N Akatli, NA Celik, RM Ulutas, H Samdanci, TE Colak, C AF Sahin, Nurhan Akatli, Nur Ayse Celik, Reha Muhammet Ulutas, Hakki Samdanci, Turkmen Emine Colak, Cemil TI The Role of CD90 in the Differential Diagnosis of Pleural Malignant Mesothelioma, Pulmonary Carcinoma and Comparison with Calretinin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mesothelioma; CD 90; Pulmonary adenocarcinoma; Squamous cell carcinoma ID Mesothelioma; CD 90; Pulmonary adenocarcinoma; Squamous cell carcinoma AB Pleural Malignant Mesothelioma (MM) is a fatal disease that has been associated with asbestos exposure. Differential diagnosis between the pleural infiltration of pulmonary carcinomas and MM is rather difficult particularly for epitheloid type mesothelioma.We aimed to investigate the utility of CD90, a cancer stem cell marker, in the differential diagnosis of MM and lung carcinoma, its prognostic significance and compare its value with that of Calretinin. Ninety pathology specimens including MM (n:30), pulmonary adenocarcinoma (n:30) and pulmonary squamous cell carcinoma (n:30) were used in this study. Immunohistochemical comparision of CD 90 and Calretinin was made in all groups. Calretinin was positive in 20 cases with MM (64.5 %), and was negative in 10 (32.3 %). CD 90 was positive in 25 of these cases (80 %) and negative in 5 (16 %). On the other hand pulmonary adenocarcinomas and squamous cell carcinomas showed positivity with CD90, 63,6 % and 73 %, respectively. We think that CD 90 has no place in the differential diagnosis between mesothelioma and pulmonary carcinoma because of the low specificity in spite of the high sensitivity. C1 [Sahin, Nurhan] Inonu University, Turgut Ozal Medical Center, Department of Pathology, 44280 Malatya, Turkey. [Akatli, Nur Ayse] Inonu University, Turgut Ozal Medical Center, Department of Pathology, 44280 Malatya, Turkey. [Celik, Reha Muhammet] Inonu University, Turgut Ozal Medical Center, Department of Thoracic SurgeryMalatya, Turkey. [Ulutas, Hakki] Inonu University, Turgut Ozal Medical Center, Department of Thoracic SurgeryMalatya, Turkey. [Samdanci, Turkmen Emine] Inonu University, Turgut Ozal Medical Center, Department of Pathology, 44280 Malatya, Turkey. [Colak, Cemil] Inonu University, Medical Faculty, Department of BiostatisticMalatya, Turkey. RP Sahin, N (reprint author), Inonu University, Turgut Ozal Medical Center, Department of Pathology, 44280 Malatya, Turkey. EM sahin.nurhan@gmail.com CR Gueugnon F, Leclercq S, Blanquart C et al, 2011, Identification of novel markers for the diagnosis ofmalignant pleural mesothelioma. Am J Pathol 178:1033–1042 Panel V, VybergM,Weinreich UM et al, 2015, The established and future biomarkers of malignant pleural mesothelioma. Cancer Treat Rev 41:486–495 Ascoli V, 2015, Pathologic diagnosis of malignant mesothelioma: chronological prospect and advent of recommendations and guidelines. Ann Ist Super Sanita 51(1):52–59 Ucer O, Dagli AF, Kilicarslan A et al, 2013, Value of glut-1 and Koc markers in the differential diagnosis of reactive mesothelial hyperplasia, malignant mesothelioma, and pulmonary adenocarcinoma. Turkish J Pathol 29:94–100 Travis WD, Brambilla E, Muller-Hermelink HK, Harris CC, eds,, 2004, World Health Organization classification of Tumours. Pathology and genetics of tumors of the lung, pleura, thymus and heart. IARC Press: Lyonn Kawamura K, Hiroshima K, Suzuki T et al, 2013, CD 90 is a diagnostic marker to differentiate between malignant pleural mesothelioma and lung carcinoma with immunohistochemistry. Am J Clin Pathol 140:544–549 Amatya VJ, Takeshima Y, Kohno H et al, 2009, Caveolin-1 is a novel immunohistochemical marker to differentiate epithelioid mesothelioma from lung adenocarcinoma. Histopathology 55:10–19 Cassoni P, Daniele L,Maldi E et al, 2009, Caveolin-1 expression in lung carcinoma varies according to tumor histotype and is acquired de novo in brain metastases. Histopathology 55:20–27 Zieglee A, Cerciello F, Bigosch C et al, 2012, Proteomic surface some analysis of mesothelioma. Lung Cancer 75:189–196 Yilmaz G, Akyol G, Cakir A et al, 2014, Investigation of diagnostic utility and expression profiles of stem cellmarkers, CD 133 and CD 90, in hepatocellular carcinoma, small cell dysplasia, and cirrhosis. Pathol Res Pract 210:419–425 Hermann PC, Bhaskar S, Cioffi Met al, 2010, Cancer stem cells in solid tumors. Semin Cancer Biol 20:77–84 Ji J, 2012, Wang Xw. Clinical implications of cancer stem cell biology in hepatocellular carcinoma. Semin Oncol 39:461–472 Deniz H, Kibar Y, GuldurME et al, 2009, Is D2-40 a useful marker for distinguishing malignant mesothelioma from pulmonary adenocarcinoma and benign mesothelial proliferations? Pathol Res Pract 205:749–752 Chaouche-Mazouni S, Scherpereel A, Zaamoum R et al, 2015, Claudin 3, 4, and 15 expressions in solid tumors of lung adenocarcinoma versus malignant pleural mesothelioma. Ann Diagn Pathol 19:193–197 Yaziji H, Battifora H, Barry TS et al, 2006, Evaluation of 12 antibodies for distinguishing epithelioid mesothelioma from adenocarcinoma: identification of a three-antibody immunohistochemical panel with maximal sensitivity and specificity. Mod Pathol 19:514–523 Ordonez NG, 2006, The diagnostic utility of immunohistochemistry in distinguishing between epithelioid mesothelioma and squamous carcinoma of the lung: a comparative study. Mod Pathol 19: 417–428 Yang ZF, Ho DW, Ng MN et al, 2008, Significance of CD 90+ cancer stem cells in human liver cancer. Cancer Cell 13: 153–166 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 487 EP 491 DI 10.1007/s12253-016-0135-9 PG 5 ER PT J AU Gaal, Zs Olah, Rejto, L Erdodi, F Csernoch, L AF Gaal, Zsuzsanna Olah, Eva Rejto, Laszlo Erdodi, Ferenc Csernoch, Laszlo TI Strong Correlation between the Expression Levels of HDAC4 and SIRT6 in Hematological Malignancies of the Adults SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute myeloid leukemia; Epigenetics; Histone deacetylation; HDAC4; SIRT6 ID Acute myeloid leukemia; Epigenetics; Histone deacetylation; HDAC4; SIRT6 AB Histone deacetylase enzymes, confirmed to have important role in the pathogenesis of leukemia, are promising targets of epigenetic treatment. However, in acute myeloid leukemia, our knowledge on their expression levels is limited, and controversial data have been published about their potential oncogenic or tumorsuppressor properties in solid tumors. In our study, the expression levels of HDAC4 and SIRT6 were evaluated via Western blot analysis in 45 bone marrow samples (2 uninfiltrated and 43 concerned by different kinds of hematological malignancies), including 32 specimens obtained from patients with newly diagnosed AML. Significantly higher HDAC4 level was detected in case of FLT3-ITD mutation compared to the group of patients without carrying this mutation (p < 0.05). Compared to the non-infiltrated samples, the expression level of HDAC4 in AML M5 patients has been proved to be significantly higher (p < 0.05). Decreasing expression levels of both HDAC4 and SIRT6 were observed during the induction treatment of FAB M5 type AML. Strong correlation has been proved between the expression levels of HDAC4 and SIRT6 (r = 0.722 in full cohort and r = 0.794 in AML), that confirms the recently suggested cooperation between NAD+-independent and NAD+-dependent HDAC enzymes in leukemia. C1 [Gaal, Zsuzsanna] University of Debrecen, Faculty of Medicine, Department of Physiology, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. [Olah, Eva] Medical and Health Science Center, University of Debrecen, Department of Pediatrics, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. [Rejto, Laszlo] University of Debrecen, Faculty of Medicine, Department of Internal Medicine, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. [Erdodi, Ferenc] University of Debrecen, Faculty of Medicine, Department of Medical Chemistry, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. [Csernoch, Laszlo] University of Debrecen, Faculty of Medicine, Department of Physiology, Nagyerdei krt. 98, H-4012 Debrecen, Hungary. RP Rejto, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Internal Medicine, H-4012 Debrecen, Hungary. EM csl@edu.unideb.hu CR Haberland M, Montgomery RL, Olson EN, 2009, The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet 10:32–42 Linggi BE, Brandt SJ, Sun ZW, Hiebert SW(2005, Translating the histone code into leukemia. J Cell Biochem 96(5):938–950 Delcuve GP, Khan DH, Davie JR, 2012, Roles of histone deacetylases in epigenetic regulation: emerging paradigms from studies with inhibitors. Clin Epigenetics 4(1):5 de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB, 2003, Histone deacetylases, HDACs): characterization of the classical HDAC family. BiochemJ 370(Pt3):737–749 Marks PA, 2010, Histone deacetylase inhibitors: a chemical genetics approach to understanding cellular functions. Biochim Biophys Acta 1799(10–12):717–725 Yang XJ, Seto E, 2008, The Rpd3/Hda1 family of lysine deacetylases: from bacteria and yeast to mice and men. Nat Rev Mol Cell Biol 9(3):206–218 Verdin E, Dequiedt F, Kasler HG, 2003, Class II histone deacetylases: versatile regulators. Trends Genet 19(5):286–293 Barneda-Zahonero B, Parra M, 2012, Histone deacetylases and cancer. Mol Oncol 6(6):579–589 Bosch-Presegue L, Vaquero A, 2011, The dual role of sirtuins in cancer. Genes Cancer 2(6):648–662 Saunders LR, Verdin E, 2007, Sirtuins: critical regulators at the crossroads between cancer and aging. Oncogene 26(37):5489– 5504 Liu H, Hu Q, D'Ercole AJ, Ye P, 2009, Histone deacetylase regulates 11. Oligodendrocyte-specific gene expression and cell development in OL-1 oligodendroglia cells. Glia 57(1):1–12 Villagra A, Cheng F, Wang HW, Suarez I, Glozak M, Maurin M et al, 2009, The histone deacetylase HDAC11 regulates the expression of interleukin 10 and immune tolerance. Nat Immunol 10(1): 92–100 Smith KT, Workman JL, 2009, Histone deacetylase inhibitors: anticancer compounds. Int J Biochem Cell Biol 41(1):21–25 Bantscheff M, Hopf C, Savitski MM, Dittmann A, Grandi P, Michon AM, 2011, Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes. Nat Biotechnol 29(3):255–265 Khan O, La Thangue NB, 2012, HDAC inhibitors in cancer biology: emergingmechanisms and clinical applications. Immunol Cell Biol 90(1):85–94 Witt O, Deubzer HE,Milde T, Oehme I, 2009, HDAC family: what are the cancer relevant targets? Cancer Lett 277(1):8–21 Tabe Y, Jin L, Contractor R, Gold D, Ruvolo P, Radke S et al, 2007, Novel role of HDAC inhibitors in AML1/ETO AML cells: activation of apoptosis and phagocytosis through induction of annexin A1. Cell Death Differ 14(8):1443–1456 Holmlund T, Lindberg MJ, Grander D,Wallberg AE, 2013, GCN5 acetylates and regulates the stability of the oncoprotein E2A-PBX1 in acute lymphoblastic leukemia. Leukemia 27(3):578–585 Di Croce L, 2005, Chromatin modifying activity of leukemia associated fusion proteins. Hum Mol Genet 14(Review Issue 1):R77– R84 Hackanson B, Rimmele L, Benkißer M, Abdelkarim M, Fliegauf M, Jung M et al, 2012, HDAC6 as a target for antileukemic drugs in acute myeloid leukemia. Leuk Res 36(8):1055–1062 Novotny-Diermayr V, Hart S, Goh KC, Cheong A, Ong LC, Hentze H et al, 2012, The oral HDAC inhibitor pracinostat, SB939, is efficacious and synergistic with the JAK2 inhibitor pacritinib(SB1518, in preclinical models of AML. Blood Cancer J 2(5):e69 Zhou L, Ruvolo VR, McQueen T, Chen W, Samudio IJ, Conneely O et al, 2013)HDAC inhibition by SNDX-275, Entinostat, restores expression of silenced leukemia-associated transcription factors Nur77 and Nor1 and of key pro-apoptotic proteins in AML. Leukemia 27(6):1358–1368 Tran HT, Kim HN, Lee IK, Nguyen-Pham TN, Ahn JS, Kim YK et al, 2013, Improved therapeutic effect against leukemia by a combination of the histone methyltransferase inhibitor chaetocin and the histone deacetylase inhibitor trichostatin a. J KoreanMed Sci 28(2): 237–246 Cea M, Soncini D, Fruscione F, Raffaghello L, Garuti A, Emionite L et al, 2011, Synergistic interactions between HDAC and Sirtuin inhibitors in human leukemia cells. PLoS ONE 6(7):e22739 Van Damme M, Crompot E, Meuleman N, Mineur P, Bron D, Lagneaux L et al, 2012, HDAC isoenzyme expression is deregulated in chronic lymphocytic leukemia B-cells and has a complex prognostic significance. Epigenetics 7(12):1403–1412 Yu SL, Lee DC, Son JW, Park CG, Lee HY, Kang J, 2013, Histone deacetylase 4 mediates SMAD family member 4 deacetylation and induces 5-fluorouracil resistance in breast cancer cells. Oncol Rep 30(3):1293–1300 Wilson AJ, Byun DS, Nasser S,Murray LB, Ayyanar K, Arango D et al, 2008, HDAC4 promotes growth of colon cancer cells via repression of p21. Mol Biol Cell 19(10):4062–4075 Zhu L, Yang J, Zhao L, Yu X, Wang L, Wang F et al, 2015, Expression of hMOF, but not HDAC4, is responsible for the global histone H4K16 acetylation in gastric carcinoma. Int J Oncol 46(6): 2535–2545 Niegisch G, Knievel J, Koch A, Hader C, Fischer U, Albers P et al, 2013, Changes in histone deacetylase, HDAC, expression patterns and activity of HDAC inhibitors in urothelial cancers. Urol Oncol 31(8):1770–1779 Gruhn B, Naumann T, Gruner D, Walther M, Wittig S, Becker S et al, 2013, The expression of histone deacetylase 4 is associated with prednisone poor-response in childhood acute lymphoblastic leukemia. Leuk Res 37(10):1200–1207 Sebastian C, Zwaans BM, Silberman DM, Gymrek M, Goren A, Zhong L et al, 2012, The histone deacetylase SIRT6 is a tumor suppressor that controls cancer metabolism. Cell 151(6):1185–1199 KhongkowM, Olmos Y, Gong C, Gomes AR, Monteiro LJ, Yague E et al, 2013, SIRT6 modulates paclitaxel and epirubicin resistance and survival in breast cancer. Carcinogenesis 34(7):1476–1486 Chen X, Hao B, Liu Y, Dai D, Han G, Li Yet al, 2014, The histone deacetylase SIRT6 suppresses the expression of the RNA-binding protein PCBP2 in glioma. Biochem Biophys Res Commun 446(1): 364–369 Fukuda T,Wada-Hiraike O, Oda K, TanikawaM, Makii C, Inaba K et al, 2015, Putative tumor suppression function of SIRT6 in endometrial cancer. FEBS Lett 589(17):2274–2281 Zhang ZG, Qin CY, 2014, Sirt6 suppresses hepatocellular carcinoma cell growth via inhibiting the extracellular signal-regulated kinase signaling pathway. Mol Med Rep 9(3):882–888 Zhang J, Yin XJ, Xu CJ, Ning YX, Chen M, Zhang H et al, 2015, The histone deacetylase SIRT6 inhibits ovarian cancer cell proliferation via down-regulation of notch 3 expression. Eur Rev Med Pharmacol Sci 19(5):818–824 Wang JC, Kafeel MI, Avezbakiyev B, Chen C, Sun Y, Rathnasabapathy C et al, 2011, Histone deacetylase in chronic lymphocytic leukemia. Oncology 81(5–6):325–329 Zhou J, Bi C, ChngWJ, Cheong LL, Liu SC, Mahara S et al, 2011, PRL-3, a metastasis associated tyrosine phosphatase, is involved in FLT3-ITD signaling and implicated in anti-AML therapy. PLoS ONE 6(5):e19798 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 493 EP 504 DI 10.1007/s12253-016-0139-5 PG 12 ER PT J AU Koszo, R Santha, D Budi, L Erfan, J Gyorfy, K Horvath, Zs Kocsis, J Landherr, L Hitre, E Mahr, K Pajkos, G Papai, Zs Kahan, Zs AF Koszo, Renata Santha, Dora Budi, Laszlo Erfan, Jozsef Gyorfy, Karoly Horvath, Zsolt Kocsis, Judit Landherr, Laszlo Hitre, Erika Mahr, Karoly Pajkos, Gabor Papai, Zsuzsanna Kahan, Zsuzsanna TI Capecitabine in Combination with Docetaxel in First Line in HER2-Negative Metastatic Breast Cancer: an Observational Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Capecitabine; Docetaxel; HER2-negative metastatic breast cancer; Toxicity ID Capecitabine; Docetaxel; HER2-negative metastatic breast cancer; Toxicity AB Due to the limited experience with capecitabine plus docetaxel (XT) combination in the first-line treatment of metastatic breast cancer in Hungary, the main objective of the study was to analyze the effectiveness and tolerability of XT therapy. A prospective, open-label, non-randomized, single- arm, multicenter, observational study was designed. All female patients were eligible whose metastatic breast cancer could be treated with the XT protocol according to the summary of product characteristics of the drugs. The median progression free survival was 9.9 ± 3.0 months. Time to treatment failure was 4.6 ± 5.1 months on average. The overall response rate was 28.9 %, the clinical benefit rate was 73.3 %. The treatment was discontinued in 35.6 % of patients due to disease progression and in 20.0%due to adverse events (AE). 33 patients with a total of 73 AEs have been reported, and 13 of them had serious adverse events (SAE). The efficacy and the safety profile of XT chemotherapy proven in the study are consistent with the results demonstrated in randomized trials. First-line XT chemotherapy effectively improves the PFS in metastatic breast cancer. C1 [Koszo, Renata] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Santha, Dora] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Budi, Laszlo] County Hospital of Borsod-Abauj-Zemplen, Szentpeteri kapu 72-76, 3526 Miskolc, Hungary. [Erfan, Jozsef] Josa Andras County Hospital, Szent Istvan u. 68, 4400 Nyiregyhaza, Hungary. [Gyorfy, Karoly] Kaposi Mor Teaching Hospital, Tallian Gy. u. 20-32, 7400 Kaposvar, Hungary. [Horvath, Zsolt] University of Debrecen, Faculty of Medicine, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Kocsis, Judit] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Landherr, Laszlo] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Uzsoki u. 29-41, 1145 Budapest, Hungary. [Hitre, Erika] National Institute of Oncology, Rath Gyorgy u. 7-9, 1126 Budapest, Hungary. [Mahr, Karoly] County Hospital of Zala, Zrinyi M. u. 1, 8900 Zalaegerszeg, Hungary. [Pajkos, Gabor] Bacs-Kiskun County Hospital, Nyiri u. 38, 6000 Kecskemet, Hungary. [Papai, Zsuzsanna] Honved Korhaz, Robert Karoly korut 44, 1134 Budapest, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. RP Kahan, Zs (reprint author), University of Szeged, Department of Oncotherapy, 6720 Szeged, Hungary. EM kahan.zsuzsanna@med.u-szeged.hu CR Rich TA, Shepard RC,Mosley ST, 2004, Four decades of continuing innovation with fluorouracil: current and future approaches to fluorouracil chemoradiation therapy. J Clin Oncol 22:2214–2232 Lockshin A, Danenberg PV, 1981, Biochemical factors affecting the tightness of 5-fluorodeoxyuridylate binding to human thymidylate synthetase. Biochem Pharmacol 30:247–257 Milano G, Etienne MC, Renee N, Thyss A, SchneiderM, Ramaioli A, Demard F, 1994, Relationship between fluorouracil systemic exposure and tumor response and patient survival. J Clin Oncol 12:1291–1295 Miwa M, Ura M, Nishida M, Sawada N, Ishikawa T, Mori K, Shimma N, Umeda I, Ishitsuka H, 1998, Design of a novel oral fluoropyrimidine carbamate, capecitabine, which generates 5-fluorouracil selectively in tumours by enzymes concentrated in human liver and cancer tissue. Eur J Cancer 34:1274–1281 Budman DR, Meropol NJ, Reigner B, Creaven PJ, Lichtman SM, Berghorn E, Behr J, Gordon RJ, Osterwalder B, Griffin T, 1998, Preliminary studies of a novel oral fluoropyrimidine carbamate: capecitabine. J Clin Oncol 16:1795–1802 Sawada N, Ishikawa T, Fukase Y, Nishida M, Yoshikubo T, Ishitsuka H, 1998, Induction of thymidine phosphorylase activity and enhancement of capecitabine efficacy by taxol/taxotere in human cancer xenografts. Clin Cancer Res 4:1013–1019 Liang X, Yan Y, Wang L, Song G, DI L, Jiang H, Wang C, Li H, 2015, First-line chemotherapy with docetaxel plus capecitabine followed by capecitabine or hormone maintenance therapy for the treatment ofmetastatic breast cancer patients Oncol Lett 9: 987–993 Pronk LC, Vasey P, Sparreboom A, Reigner B, Planting AS, Gordon RJ, Osterwalder B, Verweij J, Twelves C, 2000, A phase I and pharmacokinetic study of the combination of capecitabine and docetaxel in patients with advanced solid tumours. Br J Cancer 83: 22–29 O’Shaughnessy J, Miles D, Vukelja S, Moiseyenko V, Ayoub JP, Cervantes G, Fumoleau P, Jones S, LuiWY,Mauriac L, Twelves C, Van Hazel G, Verma S, Leonard R, 2002, Superior survival with capecitabine plus docetaxel combination therapy in anthracyclinepretreated patients with advanced breast cancer: phase III trial results. J Clin Oncol 20:2812–2823 Beslija S, Obralic N, Basic H, Tatarevic A, Naila M, Banjin M, Cardzic A, Sosevic A, Pasic A, Ceric T, Salkic B, 2006, Randomized trial of sequence vs. combination of capecitabine, X, and docetaxel, T): XT vs. T followed byX after progression as firstline therapy for patients, pts)with metastatic breast cancer, MBC). J Clin Oncol 24(Suppl 18):571 Chan S, Romieu G, Huober J, Tubiana-Hulin M, Schneeweiss A, Lluch A, Llombart A, du Bois A, Kreienberg R, Mayordomo JI, Anton A, Harrison M, Jones A, Carrasco E, Thareau Vaury A, Frimodt-Moller B, Fumoleau B, 2009, Phase III study of gemcitabine plus docetaxel compared with capecitabine plus docetaxel for anthracycline-pretreated patients with metastatic breast cancer. J Clin Oncol 27:1753–1760 Buzdar AU, Xu B, Digumarti R, Goedhals L, Hu X, Semiglazov V, Cheporov S, Gotovkin E, Hoersch S, Rittweger K, Miles DW, O’Shaughnessy J, Tjulandin S, NO16853 trial group, 2012, Randomized phase II non-inferiority study, NO16853, of two different doses of capecitabine in combination with docetaxel for locally advanced/metastatic breast cancer. Ann Oncol 23:589–597 Vici P, Giotta F, Di Lauro L, SergiD, Vizza E,Mariani L, Latorre A, Pizzuti L, D’Amico C, Giannarelli D, Colucci G, 2011, A multicenter phase II randomized trial of docetaxel/gemcitabine versus docetaxel/capecitabine as first-line treatment for advanced breast cancer: a Gruppo Oncologico Italia Meridionale study. Oncology 81:230–236 Harvey V, Mouridsen H, Semiglazov V, Jakobsen E, Voznyi E, Robinson BA, Groult V, Murawsky M, Cold S, 2006, Phase III trial comparing three doses of docetaxel for second-line treatment of advanced breast cancer. J Clin Oncol 24:4963–4970 Soto C, Torrecillas L, Reyes S, Ramirez M, Perez L, Cervantes G, Gonzalez F, Tellez E, Cortes P, Benitez H, 2006, Capecitabine, X, and taxanes in patients, pts, with anthracycline-pretreated metastatic breast cancer, MBC): sequential vs. combined therapy results from a MOSG randomized phase III trial. J Clin Oncol 24, Suppl 18): 570 Mavroudis D, Papakotoulas P, Ardavanis A, Syrigos K, Kakolyris S, Ziras N, Kouroussis C, Malamos N, Polyzos A, Christophyllakis C, Kentepozidis N, Georgoulias V, Breast Cancer Investigators of the Hellenic Oncology Research Group, 2010, Randomized phase III trial comparing docetaxel plus epirubicin versus docetaxel plus capecitabine as firstline treatment in women with advanced breast cancer. Ann Oncol 21:48–54 Michalaki V, Gennatas S, Gennatas K, 2009, Low-dose capecitabine plus docetaxel as first-line therapy for metastatic breast cancer: phase II results. Anti-Cancer Drugs 20:204–207 Seidman AD, Brufsky A, Ansari RH, Hart LL, Stein RS, Schwartzberg LS, Stewart JF, Russell CA, Chen SC, Fein LE, De La CruzVargas JA, KimSB, Cavalheiro J, Zhao L, Gill JF, Obasaju CK, Orlando M, Tai DF, 2011, Phase III trial of gemcitabine plus docetaxel versus capecitabine plus docetaxel with planned crossover to the alternate single agent in metastatic breast cancer. Ann Oncol 22:1094–1101 Bachelot T, Bajard A, Ray-Coquard I, Provencal J, Coeffic D, Agostini C, Boisseau M, Kaphan R, Dramais D, Oprea C, Ferri- Dessens RM, Guastalla JP, Perol D, 2011, Final results of ERASME-4: a randomized trial of first-line docetaxel plus either capecitabine or epirubicin for metastatic breast cancer. Oncology 80:262–268 Yu J, Lj DI, Song G, Che L, Hf J, Zhu YI, Liang X, Jia J, Zhang J, Yang H, Wang XI, Xn Z, Ren J, 2011, Randomized clinical casecontrol trial for the comparison of docetaxel plus thiotepa versus docetaxel plus capecitabine in patients with metastatic breast cancer. Beijing Da Xue Xue Bao 43:151–156 Gradishar WJ, Meza LA, Amin B, Samid D, Hill T, Chen YM, Lower EE, Marcom PK, 2004, Capecitabine plus paclitaxel as front-line combination therapy for metastatic breast cancer: a multicenter phase II study. J Clin Oncol 22:2321–2327 Blum JL, Dees EC, Chacko A, Doane L, Ethirajan S, Hopkins J, McMahon R, Merten S, Negron A, Neubauer M, Ilegbodu D, Boehm KA, Asmar L, O’Shaughnessy JA, 2006, Phase II trial of capecitabine and weekly paclitaxel as first-line therapy for metastatic breast cancer. J Clin Oncol 24:4384–4390 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 505 EP 511 DI 10.1007/s12253-016-0129-7 PG 7 ER PT J AU Semjen, D Farkas, A Kalman, E Kaszas, B Kovacs, Pusztai, Cs Szuhai, K Tornoczky, T AF Semjen, David Farkas, Andras Kalman, Endre Kaszas, Balint Kovacs, Arpad Pusztai, Csaba Szuhai, Karoly Tornoczky, Tamas TI More Cases of Benign Testicular Teratomas are Detected in Adults than in Children. A Clinicopathological Study of 543 Testicular Germ Cell Tumor Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Benign teratoma; Mature teratoma; Germ cell tumor; Testis; Postpubertal; Testicular neoplasms ID Benign teratoma; Mature teratoma; Germ cell tumor; Testis; Postpubertal; Testicular neoplasms AB Benign testicular teratomas are always thought to be pediatric neoplasms and previously all the teratoid tumors in the adult testis regarded as malignant. Recently, three publications reported benign testicular teratomas in adulthood and the latest WHO classification refers them as "prepubertal type of teratomas" which rarely appear in adulthood. These neoplasms behave benign and seemingly analogous independently whether they appear in pre- or postpubertal patients. The aim of our study was to investigate the frequency of benign testicular teratomas both in children and adults. 593 cases of testicular neoplasms were found in a period of 17 years ranging from1998 to 2014 in the archive of our department (Department of Pathology, Medical Center, Pecs University). 543 cases diagnosed as germ cell tumor which have all been further evaluated in conjunction with the clinical data available. Of all germ cell tumor cases 14 (2.5 %) were pure teratomas. Ten out of 14 were the WHO-defined "conventional" teratoma, 4 of the 14 were the "benign or the so called prepubertal type" from which three occurred in adult patients. Only one of the 14 occurred in childhood, indicating that benign prepubertal type teratomas –which are regarded generally as childhood tumors- are more frequently detected in adults than in children. Benign adult testicular teratomas comprised 21 % of all pure teratoma cases in our series. Practicioners in the field have to be aware of its existence also in adulthood to avoid overtreatment and not to expose their patients to unnecessary chemotherapy, retroperitoneal lymphadenectomy (RLA) and the potential complications of these interventions. C1 [Semjen, David] University of Pecs, Department of Pathology, Rakoczi u 2, H-7624 Pecs, Hungary. [Farkas, Andras] University of Pecs, Department of Pediatrics, Jozsef Attila u. 7, H-7623 Pecs, Hungary. [Kalman, Endre] University of Pecs, Department of Pathology, Rakoczi u 2, H-7624 Pecs, Hungary. [Kaszas, Balint] University of Pecs, Department of Pathology, Rakoczi u 2, H-7624 Pecs, Hungary. [Kovacs, Arpad] Pecs University, Faculty of Health Sciences, Department of Diagnostic, Voromarty u. 4, H-7621 Pecs, Hungary. [Pusztai, Csaba] University of Pecs, Department of Urology, Munkacsy M. u. 2, H-7621 Pecs, Hungary. [Szuhai, Karoly] Leiden University, Medical Center, Department of Molecular Cell Biology, Einthovenweg 20, 2300RC Leiden, The Netherlands. [Tornoczky, Tamas] University of Pecs, Department of Pathology, Rakoczi u 2, H-7624 Pecs, Hungary. RP Semjen, D (reprint author), University of Pecs, Department of Pathology, H-7624 Pecs, Hungary. EM semjen.david@pte.hu CR Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM, 2016, The 2016 WHO classification of tumours of the urinary system and male genital organs-part a: renal, penile, and testicular tumours. Eur Urol 70(1):93–105 Srigley JR, Mackay B, Toth P, Ayala A, 1988, The ultrastructure and histogenesis of male germ neoplasia with emphasis on seminoma with early carcinomatous features. Ultrastruct Pathol 12(1):67–86 Zhang C, Berney DM, Hirsch MS, Cheng L, Ulbright TM, 2013, Evidence supporting the existence of benign teratomas of the postpubertal testis a clinical, histopathologic, and molecular genetic analysis of 25 cases. Am J Surg Pathol 37(6):827–835 Semjen D, Kalman E, Tornoczky T, Szuhai K, 2014, Further evidence of the existence of benign teratomas of the postpubertal testis. Am J Surg Pathol 38(4):580–581 Oosterhuis JW, Stoop JA, Rijlaarsdam MA, Biermann K, Smit VT, Hersmus R, Looijenga LH, 2015, Pediatric germ cell tumors presenting beyond childhood? Andrology 3(1):70–77., DOI 10.1111/andr.305 Oliver RT, Mason MD, Mead GM, von der Maase H, Rustin GJ, Joffe JK et al, 2005, Radiotherapy versus single-dose carboplatin in adjuvant treatment of stage I seminoma: a randomised trial. Lancet 366:293–300 Classen J, Schmidberger H, Meisner C, Souchon R, Sautter-Bihl M-L, Sauer R et al, 2003, Radiotherapy for stages IIA/B testicular seminoma: final report of a prospective multicenter clinical trial. J Clin Oncol 21:1101–1106 Carver BS, Shayegan B, Eggener S, Stasi J, Motzer RJ, Bosl GJ et al, 2007, Incidence of metastatic non seminomatous germ cell tumor outside the boundaries of a modified post chemotherapy retroperitoneal lymph node dissection. J Clin Oncol 25:4365–4369 Fischer CG,WaechterW, Kraus S, Fuentecilla Perez E,WeidnerW et al, 1998, Urologic tumors in the Federal Republic of Germany: data on 56,013 cases from hospital cancer registries. Cancer 82(4): 775–783 Jacobsen G, Barlebo H, Olsen J, Schultz HP, Starklint H, Sogaard H et al, 1984, Testicular germ cell tumours in Denmark 1976-1980. Pathology of 1058 consecutive cases. Krag Acta Radiol Oncol 23(4):239–247 D.E. Cooper, J.O. L'Esperance, M.S. Christman, B.K. Auge, 2008, Testis Cancer: a 20-year Epidemiological Review of the Experience at a Regional Military Medical Facility Adult Urology J Urol, 180(2):577–581; discussion 581-2., DOI 10.1016/j.juro.2008.04.032 Oosterhuis JW, Looijenga LH, 2005, Testicular germ-cell tumours in a broader perspective. Nat Rev Cancer 5(3):210–222 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 513 EP 517 DI 10.1007/s12253-016-0120-3 PG 5 ER PT J AU Jin, JY Jeong, WJ Paik, HJ Ahn, SH AF Jin, Ju Young Jeong, Woo-Jin Paik, Ho Jin Ahn, Soon-Hyun TI Role of Frozen Biopsy in Glottic Premalignant Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glottis premalignant lesion; Glottis cancer; Laser excision; Cordectomy; Frozen biopsy ID Glottis premalignant lesion; Glottis cancer; Laser excision; Cordectomy; Frozen biopsy AB Frozen biopsies are frequently used for decision making during surgery. This study aimed to evaluate the efficacy of frozen biopsy for guiding decision making before laser excision of glottic premalignant lesions. One hundred patients with 119 laser excisions were included in this study and reviewed retrospectively. After frozen biopsy, type I or II cordectomy was performed and the frozen result and final pathology of the excisional specimen were compared. The positive predictive value of frozen biopsy when the diagnosis is benign or malignant was relatively high (80.8 and 88.9 %, respectively) but the positive predictive value of a dysplasia or carcinoma in situ result was quite low (18.2 and 16.7 %). Under-diagnosis was frequent for dysplasia or carcinoma in situ (69.7 and 83.3 %). In particular, for lesions with suspicious features, lesions with dysplasia or carcinoma in situ had a much higher rate of under-diagnosis (81.8 and 100 %). Frozen biopsy was not reliable because the overall coincidence rate between final pathology and frozen biopsy was 63 %. Although a frozen biopsy result of a benign or malignant result was reliable, a dysplasia or carcinoma in situ result on frozen biopsy had a high risk of being an under-diagnosis. C1 [Jin, Ju Young] Seoul National University, College of Medicine, Bundang Hospital, Department of Otorhinolaryngology-Head and Neck Surgery, 173-82 Gumiro, Bundang-gu, 13620 Gyeonggi, South Korea. [Jeong, Woo-Jin] Seoul National University, College of Medicine, Bundang Hospital, Department of Otorhinolaryngology-Head and Neck Surgery, 173-82 Gumiro, Bundang-gu, 13620 Gyeonggi, South Korea. [Paik, Ho Jin] Seoul National University, College of Medicine, Bundang Hospital, Department of Pathology, 173-82 Gumiro, Bundang-gu, 13620 Gyeonggi, South Korea. [Ahn, Soon-Hyun] Seoul National University, College of Medicine, Bundang Hospital, Department of Otorhinolaryngology-Head and Neck Surgery, 173-82 Gumiro, Bundang-gu, 13620 Gyeonggi, South Korea. RP Ahn, SH (reprint author), Seoul National University, College of Medicine, Bundang Hospital, Department of Otorhinolaryngology-Head and Neck Surgery, 13620 Gyeonggi, South Korea. EM ahnsh30@snu.ac.kr CR Fleskens SA, van der Laak JA, Slootweg PJ, Takes RP, 2010, Management of laryngeal premalignant lesions in the Netherlands. Laryngoscope 120(7):1326–1335., DOI 10.1002/lary.20888 Dispenza F, De Stefano A, Marchese D, Martines F, Dispenza C, 2012, Management of laryngeal precancerous lesions. Auris Nasus Larynx 39(3):280–283., DOI 10.1016/j.anl.2011.08.002 Remacle M, Eckel HE, Antonelli A, Brasnu D, Chevalier D, Friedrich G, Olofsson J, Rudert HH, Thumfart W, de Vincentiis M, Wustrow TP, 2000, Endoscopic cordectomy. A proposal for a classification by the Working Committee. Eur Laryngol Soc Eur Arch Oto-Rhino-Laryngol : Off J Eur Fed Oto-Rhino-Laryngol Soc 257(4):227–231 Tomifuji M, Araki K, Niwa K, Miyagawa Y,Mizokami D, Kitagawa Y, Yamashita T, Matsunobu T, Shiotani A, 2013, Comparison of voice quality after laser cordectomy with that after radiotherapy or chemoradiotherapy for early glottic carcinoma. ORL; J Oto-Rhino- Laryngol Relat Spec 75(1):18–26., DOI 10.1159/000346934 Weller MD, Nankivell PC, McConkey C, Paleri V, Mehanna HM, 2010, The risk and interval to malignancy of patients with laryngeal dysplasia; a systematic review of case series and meta-analysis. Clin Otolaryngol : Off J ENT-UK; Off J Netherlands Soc Oto-Rhino- Laryngol Cervico-Fac Surg 35(5):364–372., DOI 10.1111/j.1749- 4486.2010.02181.x Cosway B, Paleri V, 2015, Laryngeal dysplasia: an evidence-based flowchart to guide management and follow up. J Laryngol Otol 129(6):598–599., DOI 10.1017/S0022215115000833 Cikojevic D, Gluncic I, Pesutic-Pisac V, 2008, Comparison of contact endoscopy and frozen section histopathology in the intraoperative diagnosis of laryngeal pathology. J Laryngol Otol 122(8): 836–839., DOI 10.1017/S0022215107000539 Mehanna H, Paleri V, Robson A, Wight R, Helliwell T, 2010, Consensus statement by otorhinolaryngologists and pathologists on the diagnosis and management of laryngeal dysplasia. Clin Otolaryngol : Off J ENT-UK; Off J Netherlands Soc Oto-Rhino- Laryngol Cervico-Fac Surg 35(3):170–176., DOI 10.1111/j.1749- 4486.2010.02119.x NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 519 EP 523 DI 10.1007/s12253-016-0143-9 PG 5 ER PT J AU Alanazi, M Pathan, AKA Shaik, PJ Alhadheq, A Khan, Z Khan, W Al Naeem, A Parine, RN AF Alanazi, Mohammed Pathan, Ali Khan Akbar Shaik, P Jilani Alhadheq, Abdullah Khan, Zahid Khan, Wajahatullah Al Naeem, Abdulrahman Parine, Reddy Narasimha TI The hOGG1 Ser326Cys Gene Polymorphism and Breast Cancer Risk in Saudi Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE OGG1; Saudi population; Breast cancer; OGG1 Ser326Cys ID OGG1; Saudi population; Breast cancer; OGG1 Ser326Cys AB The purpose of this study was to test the association between human 8-oxoguanine glycosylase 1 (hOGG1) gene polymorphisms and susceptibility to breast cancer in Saudi population. We have also aimed to screen the hOGG1 Ser326Cys polymorphism effect on structural and functional properties of the hOGG1 protein using in silico tools. We have analyzed four SNPs of hOGG1 gene among Saudi breast cancer patients along with healthy controls. Genotypes were screened using TaqMan SNP genotype analysis method. Experimental data was analyzed using Chi-square, t test and logistic regression analysis using SPSS software (v.16). In silco analysis was conducted using discovery studio and HOPE program. Genotypic analysis showed that hOGG1 rs1052133 (Ser326Cys) is significantly associated with breast cancer samples in Saudi population, however rs293795 (T >C), rs2072668 (C>G) and rs2075747 (G >A) did not show any association with breast cancer. The hOGG1 SNP rs1052133 (Ser326Cys) minor allele Tshowed a significant association with breast cancer samples (OR = 1.78, χ2 = 7.86, p = 0.02024). In silico structural analysis was carried out to compare the wild type (Ser326) and mutant (Cys326) protein structures. The structural prediction studies revealed that Ser326Cys variant may destabilize the protein structure and it may disturb the hOGG1 function. Taken together this is the first In silico study report to confirm Ser326Cys variant effect on structural and functional properties of hOGG1 gene and Ser326Cys role in breast cancer susceptibility in Saudi population. C1 [Alanazi, Mohammed] King Saud University, College of Science, Genome Research Chair, 11451 Riyadh, Saudi Arabia. [Pathan, Ali Khan Akbar] King Saud University, College of Science, Genome Research Chair, 11451 Riyadh, Saudi Arabia. [Shaik, P Jilani] King Saud University, College of Science, Genome Research Chair, 11451 Riyadh, Saudi Arabia. [Alhadheq, Abdullah] King Saud University, College of Science, Genome Research Chair, 11451 Riyadh, Saudi Arabia. [Khan, Zahid] King Saud University, College of Science, Genome Research Chair, 11451 Riyadh, Saudi Arabia. [Khan, Wajahatullah] King Saud bin Abdulaziz University for Health Sciences, College of Science and Health Professions, Basic Sciences Department, 11426 Riyadh, Saudi Arabia. [Al Naeem, Abdulrahman] King Fahad Medical City, Department of Women’s ImagingRiyadh, Saudi Arabia. [Parine, Reddy Narasimha] King Saud University, College of Science, Genome Research Chair, 11451 Riyadh, Saudi Arabia. RP Parine, RN (reprint author), King Saud University, College of Science, Genome Research Chair, 11451 Riyadh, Saudi Arabia. EM reddyparine@gmail.com CR Choi M, Scholl UI, Ji W, Liu T, Tikhonova IR, Zumbo P, Nayir A, Bakkaloglu A, Ozen S, Sanjad S, Nelson-Williams C, Farhi A, Mane S, Lifton RP, 2009, Genetic diagnosis by whole exome capture and massively parallel DNA sequencing. Proc Natl Acad Sci U S A 106(45):19096–19101., DOI 10.1073/pnas.0910672106 Castrilli G, Fabiano A, La Torre G, Marigo L, Piantelli C, Perfetti G, Ranelletti FO, Piantelli M, 2002, Expression of hMSH2 and hMLH1 proteins of the human DNA mismatch repair system in salivary gland tumors. J Oral Pathol Med: Off Publ Int Assoc Oral Pathol Am Acad Oral Pathol 31(4):234–238 Maynard S, Schurman SH, Harboe C, de Souza-Pinto NC, BohrVA, 2009, Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis 30(1):2–10., DOI 10.1093/carcin/bgn250 AlanaziM, Pathan AA, Abduljaleel Z, Shaik JP, Alabdulkarim HA, Semlali A, Bazzi MD, Parine NR, 2013, Association between PARP-1 V762A polymorphism and breast cancer susceptibility in Saudi population. PLoS One 8(12), e85541., DOI 10.1371/journal. pone.0085541 AlMutairi F, Ali Khan Pathan A, Alanazi M, Shalaby M, Alabdulkarim HA, Alamri A, Al Naeem A, Elrobh M, Shaik JP, Khan W, Khan Z, Reddy Parine N, 2015, Association of DNA repair gene APE1 Asp148Glu polymorphism with breast cancer risk. Dis Markers 2015:869512., DOI 10.1155/2015/869512 Al Mutairi FM, Alanazi M, Shalaby M, Alabdulkarim HA, Pathan AA, Parine NR, 2013, Association of XRCC1 gene polymorphisms with breast cancer susceptibility in Saudi patients. Asian Pac J Cancer Prev: APJCP 14(6):3809–3813 Dianov GL, Hubscher U, 2013, Mammalian base excision repair: the forgotten archangel. Nucleic Acids Res 41(6):3483–3490., DOI 10.1093/nar/gkt076 Kohno T, Shinmura K, Tosaka M, Tani M, Kim SR, Sugimura H, Nohmi T, Kasai H, Yokota J, 1998, Genetic polymorphisms and alternative splicing of the hOGG1 gene, that is involved in the repair of 8-hydroxyguanine in damaged DNA. Oncogene 16(25): 3219–3225., DOI 10.1038/sj.onc.1201872 Alanazi M, Pathan AA, Ajaj SA, Khan W, Shaik JP, Al Tassan N, Parine NR, 2013, DNA repair genes XRCC1, XRCC3, XPD, and OGG1 polymorphisms among the central region population of Saudi Arabia. Biol Res 46(2):161–167., DOI 10.4067/S0716- 97602013000200007 Minowa O, Arai T, Hirano M,Monden Y, Nakai S, FukudaM, Itoh M, Takano H, Hippou Y, Aburatani H, Masumura K, Nohmi T, Nishimura S, Noda T, 2000, Mmh/Ogg1 gene inactivation results in accumulation of 8-hydroxyguanine in mice. Proc Natl Acad Sci U S A 97(8):4156–4161., DOI 10.1073/pnas.050404497 Friedberg E, Walker G, Siede W, Wood R, Schultz R, 2006, In: Ellenberger T, ed, DNA repair and mutagenesis. ASM Press, Washington, DC Parker AR, O’Meally RN, Sahin F, Su GH, Racke FK, Nelson WG, DeWeese TL, Eshleman JR, 2003, Defective human MutY phosphorylation exists in colorectal cancer cell lines with wild-type MutY alleles. J Biol Chem 278(48):47937– 47945., DOI 10.1074/jbc.M306598200 Srivastava K, Srivastava A, Mittal B, 2010, Polymorphisms in ERCC2, MSH2, and OGG1 DNA repair genes and gallbladder cancer risk in a population of Northern India. Cancer 116(13): 3160–3169., DOI 10.1002/cncr.25063 Thameem F, Puppala S, Lehman DM, SternMP, Blangero J, Abboud HE, Duggirala R, Habib SL, 2010, The Ser, 326, Cys polymorphism of 8-oxoguanine glycosylase 1, OGG1, is associated with type 2 diabetes in Mexican Americans. Hum Hered 70(2):97–101 Zhang Y, 2008, I-TASSER server for protein 3D structure prediction. BMC Bioinforma 9:40., DOI 10.1186/1471-2105-9-40 Sameer AS, Nissar S, Abdullah S, Chowdri NA, Siddiqi MA, 2012, DNA repair gene 8-oxoguanine DNA glycosylase Ser326Cys polymorphism and colorectal cancer risk in a Kashmiri population. DNA Cell Biol 31(4):541–546., DOI 10.1089/dna.2011.1349 Yamane A, Kohno T, Ito K, Sunaga N, Aoki K, Yoshimura K, Murakami H, Nojima Y, Yokota J, 2004, Differential ability of polymorphic OGG1 proteins to suppress mutagenesis induced by 8-hydroxyguanine in human cell in vivo. Carcinogenesis 25(9): 1689–1694., DOI 10.1093/carcin/bgh166 Weiss JM, Goode EL, Ladiges WC, Ulrich CM, 2005, Polymorphic variation in hOGG1 and risk of cancer: a review of the functional and epidemiologic literature. Mol Carcinog 42(3): 127–141., DOI 10.1002/mc.20067 Elahi A, Zheng Z, Park J, Eyring K, McCaffrey T, Lazarus P, 2002, The human OGG1 DNA repair enzyme and its association with orolaryngeal cancer risk. Carcinogenesis 23(7):1229–1234 Kohno T, Kunitoh H, Toyama K, Yamamoto S, Kuchiba A, Saito D, Yanagitani N, Ishihara S, Saito R, Yokota J, 2006, Association of the OGG1-Ser326Cys polymorphism with lung adenocarcinoma risk. Cancer Sci 97(8):724–728., DOI 10.1111/j.1349-7006.2006.00240.x Kim KY, Han W, Noh DY, Kang D, Kwack K, 2013, Impact of genetic polymorphisms in base excision repair genes on the risk of breast cancer in a Korean population. Gene 532(2):192–196., DOI 10.1016/j.gene.2013.09.069 Lupo PJ, Lee LJ, Okcu MF, Bondy ML, Scheurer ME, 2012, An exploratory case-only analysis of gene-hazardous air pollutant interactions and the risk of childhood medulloblastoma. Pediatr Blood Cancer 59(4):605–610., DOI 10.1002/pbc.24105 Wiederstein M, Sippl MJ, 2007, ProSA-web: interactive web service for the recognition of errors in three-dimensional structures of proteins. Nucleic acids research 35, Web Server issue):W407-410., DOI 10.1093/nar/gkm290 Lovell SC, Davis IW, Adrendall WB, de Bakker PIW, Word JM, Prisant MG, Richardson JS, Richardson DC, 2003, Structure validation by C alpha geometry: phi, psi and C beta deviation. Proteins- Struct Funct Genet 50(3):437–450., DOI 10.1002/prot.10286 Sali A, Potterton L, Yuan F, Vanvlijmen H, Karplus M, 1995, Evaluation of comparative protein modeling by modeler. Proteins- Struct Funct Genet 23(3):318–326., DOI 10.1002/prot.340230306 Capriotti E, Fariselli P, Casadio R, 2005, I-Mutant2.0: predicting stability changes upon mutation from the protein sequence or structure. Nucleic Acids Res 33:W306–W310., DOI 10.1093/nar/gki375 Venselaar H, te Beek TAH, Kuipers RKP, Hekkelman ML, Vriend G, 2010, Protein structure analysis of mutations causing inheritable diseases. An e-Science approach with life scientist friendly interfaces. BMC Bioinform 11:548., DOI 10.1186/1471-2105-11-548 Guo SW, Thompson EA, 1992, Performing the exact test of hardyweinberg proportion for multiple alleles. Biometrics 48(2):361– 372., DOI 10.2307/2532296 Nohmi T, Kim SR, Yamada M, 2005, Modulation of oxidative mutagenesis and carcinogenesis by polymorphic forms of human DNA repair enzymes. Mutn Res-Fundam Mol Mech Mutagen 591(1–2):60–73., DOI 10.1016/j.mrfmmm.2005.03.033 Wei B, Zhou Y, Xu Z, Xi B, Cheng H, Ruan J, ZhuM, Hu Q,Wang Q,Wang Z, Yan Z, Jin K, Zhou D, Xuan F, Huang X, Shao J, Lu P, 2011, The effect of hOGG1 Ser326Cys polymorphism on cancer risk: evidence from a meta-analysis. PLoS One 6(11), e27545., DOI 10.1371/journal.pone.0027545 Canbay E, Cakmakoglu B, Zeybek U, Sozen S, Cacina C, Gulluoglu M, Balik E, Bulut T, Yamaner S, Bugra D, 2011, Association of APE1 and hOGG1 polymorphisms with colorectal cancer risk in a Turkish population. Curr Med Res Opin 27(7): 1295–1302., DOI 10.1185/03007995.2011.573544 Rodrigues P, de Marco G, Furriol J, Mansego ML, Pineda-Alonso M, Gonzalez-Neira A, Martin-Escudero JC, Benitez J, Lluch A, Chaves FJ, Eroles P, 2014, Oxidative stress in susceptibility to breast cancer: study in Spanish population. BMC Cancer 14:861., DOI 10.1186/1471-2407-14-861 Sangrajrang S, Schmezer P, Burkholder I, Waas P, Boffetta P, Brennan P, Bartsch H, Wiangnon S, Popanda O, 2008, Polymorphisms in three base excision repair genes and breast cancer risk in Thai women. Breast Cancer Res Treat 111(2):279–288., DOI 10.1007/s10549-007-9773-7 Synowiec E, Stefanska J, Morawiec Z, Blasiak J, Wozniak K, 2008, Association between DNA damage, DNA repair genes variability and clinical characteristics in breast cancer patients. Mutat Res 648(1–2): 65–72 Rossner P Jr, TerryMB, GammonMD, Zhang FF, Teitelbaum SL, Eng SM, Sagiv SK, Gaudet MM, Neugut AI, Santella RM, 2006, OGG1 polymorphisms and breast cancer risk. Cancer Epidemiol Biomarkers Prev: Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol 15(4):811–815., DOI 10.1158/1055-9965.EPI-05-0659 Peng Q, Lu Y, Lao X, Chen Z, Li R, Sui J, Qin X, Li S, 2014, Association between OGG1 Ser326Cys and APEX1 Asp148Glu polymorphisms and breast cancer risk: a meta-analysis. Diagn Pathol 9:108., DOI 10.1186/1746-1596-9-108 Luo H, Li Z, Qing Y, Zhang SH, Peng Y, Li Q, Wang D, 2014, Single nucleotide polymorphisms of DNA base-excision repair genes, APE1, OGG1 and XRCC1, associated with breast cancer risk in a Chinese population. Asian Pac J Cancer Prev: APJCP 15(3):1133–1140 Yuan WG, Xu LD, Feng YX, Yang Y, Chen WY, Wang JW, Pang D, Li DJ, 2010, The hOGG1 Ser326Cys polymorphism and breast cancer risk: a meta-analysis. Breast Cancer Res Treat 122(3):835– 842., DOI 10.1007/s10549-009-0722-5 Hill JW, EvansMK, 2006, Dimerization and opposite base-dependent catalytic impairment of polymorphic S326C OGG1 glycosylase. Nucleic Acids Res 34(5):1620–1632., DOI 10.1093/nar/gkl060 Simonelli V, Camerini S, Mazzei F, Van Loon B, Allione A, D’Errico M, Barone F, Minoprio A, Ricceri F, Guarrera S, Russo A, Dalhus B, Crescenzi M, Hubscher U, Bjoras M, Matullo G, Dogliotti E, 2013, Genotype-phenotype analysis of S326C OGG1 polymorphism: a risk factor for oxidative pathologies. Free Radic Biol Med 63:401–409., DOI 10.1016/j.freeradbiomed.2013.05.031 Sidorenko VS, Grollman AP, Jaruga P, Dizdaroglu M, Zharkov DO, 2009, Substrate specificity and excision kinetics of natural polymorphic variants and phosphomimetic mutants of human 8- oxoguanine-DNA glycosylase. Febs J 276(18):5149–5162., DOI 10.1111/j.1742-4658.2009.07212.x NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 525 EP 535 DI 10.1007/s12253-016-0146-6 PG 11 ER PT J AU Thanmalagan, RR Naorem, DL Venkatesan, A AF Thanmalagan, Rajeswary Raja Naorem, Devi Leimarembi Venkatesan, Amouda TI Expression Data Analysis for the Identification of Potential Biomarker of Pregnancy Associated Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Network analysis; PTM; Pregnancy-associated breast cancer; Association rule mining ID Network analysis; PTM; Pregnancy-associated breast cancer; Association rule mining AB Breast cancer affects every 1 of 3000 pregnant women or in the first post-partum year is referred as Pregnancy Associated Breast Cancer (PABC) in mid 30s. Even-though rare disease, classified under hormone receptor negative status which metastasis quickly to other parts by extra cellular matrix degradation. Hence it is important to find an optimal treatment option for a PABC patient. Also additional care should be taken to choose the drug; in order to avoid fetal malformation and post-partum stage side-effects. The adaptation of target based therapy in the clinical practice may help to substitute the mastectomy treatment. Recent studies suggested that certain altered Post Translational Modifications (PTMs) may be an indicative of breast cancer progression; an attempt is made to consider the over represented PTM as a parameter for gene selection. The public dataset of PABC from GEO were examined to select Differentially Expressed Genes (DEG). The corresponding PTMs for DEG were collected and association between them was found using data mining technique. Usually clustering algorithm has been applied for the study of gene expression with drawback of clustering of gene products based on specified features. But association rule mining method overcome this shortcoming and determines the useful and in depth relationships. From the association, genes were selected to study the interactions and pathways. These studies emphasis that the genes KLF12, FEN1 MUC1 and SP110, can be chosen as target, which control cancer development, without any harm to pregnancy as well as fetal developmental process. C1 [Thanmalagan, Rajeswary Raja] Pondicherry University, Centre for Bioinformatics, 605014 Puducherry, India. [Naorem, Devi Leimarembi] Pondicherry University, Centre for Bioinformatics, 605014 Puducherry, India. [Venkatesan, Amouda] Pondicherry University, Centre for Bioinformatics, 605014 Puducherry, India. RP Venkatesan, A (reprint author), Pondicherry University, Centre for Bioinformatics, 605014 Puducherry, India. EM amouda@bicpu.edu.in CR Keyser EA, Staat BC, Fausett MB, Shields AD, 2012, Pregnancyassociated breast cancer. Rev Obstet Gynecol 5(2):94–99 Jin H, Zangar RC, 2009, Protein modifications as potential biomarkers in breast cancer. Biomark Insights 4:191–200 Wang Y-C, Peterson SE, Loring JF, 2014, Protein post-translational modifications and regulation of pluripotency in human stem cells. Cell Res 24(2):143–160., DOI 10.1038/cr.2013.151 Becquet C, Blachon S, Jeudy B, Boulicaut J-F, GandrillonO, 2002, Strong-association-rule mining for large-scale gene-expression data analysis: a case study on human SAGE data. Genome Biol 3(12, research0067.0061-research0067.0016 Creighton C, Hanash S, 2003, Mining gene expression databases for association rules. Bioinformatics, Oxford, England, 19(1):79–86 Seeja KR, Alam MA, Jain SK, 2009, An association rule mining Approach for co-regulated Signature genes identification in cancer. J Circ Sys Comp 18(08):1409–1423., DOI 10.1142 /S0218126609005757 Pugazhendi D, 2013, Apriori algorithm on marine fisheries biological data. Int J Comp Sci & Eng Technol 4(12):1409–1411 Harvell DM, Kim J, O'Brien J, Tan AC, Borges VF, Schedin P, Jacobsen BM, Horwitz KB, 2013, Genomic signatures of pregnancy-associated breast cancer epithelia and stroma and their regulation by estrogens and progesterone. Horm & cancer 4(3): 140–153., DOI 10.1007/s12672-013-0136-z The UniProt C, 2012, Reorganizing the protein space at the universal protein resource, UniProt). Nucleic Acids Res 40(D1):D71–D75 Agrawal R, Srikant R, 1994, Fast algorithms for mining association rules in large databases. Paper presented at the proceedings of the 20th international conference on very large data bases Kanehisa M, Goto S, Kawashima S, Nakaya A, 2002, The KEGG databases at GenomeNet. Nucleic Acids Res 30(1):42–46 Zuberi K, Franz M, Rodriguez H, Montojo J, Lopes CT, Bader GD, Morris Q, 2013, GeneMANIA prediction server 2013 update. Nucleic Acids Res 41(W1):W115–W122 Huang DW, Sherman BT, Tan Q, Kir J, Liu D, Bryant D, Guo Y, Stephens R, Baseler MW, Lane HC, Lempicki RA, 2007, DAVID bioinformatics resources: expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Res 35(Web Server issue):W169–W175., DOI 10.1093 /nar/gkm415 Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T, 2003, Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13(11):2498–2504., DOI 10.1101/gr.1239303 Freeman LC, 1978, Centrality in social networks conceptual clarification. Soc Networks 1(3):215–239., DOI 10.1016/0378- 8733(78)90021-7 Valente TW, Coronges K, Lakon C, Costenbader E, 2008, How correlated are network centrality measures? Connections, Toronto, Ont, 28(1):16–26 Azuaje FJ, 2014, Selecting biologically informative genes in coexpression networks with a centrality score. Biol Direct 9:12., DOI 10.1186/1745-6150-9-12 Chen HM, Tanaka N, Mitani Y, Oda E, Nozawa H, Chen JZ, Yanai H, Negishi H, Choi MK, Iwasaki T, Yamamoto H, Taniguchi T, Takaoka A, 2009, Critical role for constitutive type I interferon signaling in the prevention of cellular transformation. Cancer Sci 100(3):449–456., DOI 10.1111/j.1349-7006.2008.01051.x Golubovskaya VM, Kweh FA, Cance WG, 2009, Focal adhesion kinase and cancer. Histol Histopathol 24(4):503–510 McCready J, Arendt LM, Glover E, Iyer V, Briendel JL, Lyle SR, Naber SP, Jay DG, Kuperwasser C, 2014, Pregnancy-associated breast cancers are driven by differences in adipose stromal cells present during lactation. Breast Cancer Res : BCR 16(1):R2., DOI 10.1186/bcr3594 Russo J, Moral R, Balogh GA, Mailo D, Russo IH, 2005, The protective role of pregnancy in breast cancer. Breast Cancer Res : BCR 7(3):131–142., DOI 10.1186/bcr1029 Britt K, Ashworth A, Smalley M, 2007, Pregnancy and the risk of breast cancer. Endocr Relat Cancer 14(4):907–933., DOI 10.1677 /erc-07-0137 Anders CK, Johnson R, Litton J, Phillips M, Bleyer A, 2009, Breast cancer before age 40 Years. Semin Oncol 36(3):237–249., DOI 10.1053/j.seminoncol.2009.03.001 Gabriel CA, Domchek SM, 2010, Breast cancer in young women. Breast Cancer Res : BCR 12(5):212–212., DOI 10.1186/bcr2647 Pawson T, Scott JD, 2005, Protein phosphorylation in signaling – 50 years and counting. Trends Biochem Sci 30(6):286–290., DOI 10.1016/j.tibs.2005.04.013 Knutson TP, Daniel AR, Fan D, Silverstein KA, Covington KR, Fuqua SA, Lange CA, 2012, Phosphorylated and sumoylationdeficient progesterone receptors drive proliferative gene signatures during breast cancer progression. Breast Cancer Res : BCR 14(3): R95., DOI 10.1186/bcr3211 Daniel AR, Faivre EJ, Lange CA, 2007, Phosphorylationdependent antagonism of sumoylation derepresses progesterone receptor action in breast cancer cells. Mol endocrinol, Baltimore, Md, 21(12):2890–2906., DOI 10.1210/me.2007-0248 McIlwain DR, Berger T, Mak TW(2013, Caspase functions in cell death and disease. Cold Spring Harb Perspect Biol 5(4):a008656., DOI 10.1101/cshperspect.a008656 Elmore S, 2007, Apoptosis: a review of programmed cell death. Toxicol Pathol 35(4):495–516., DOI 10.1080 /01926230701320337 Nakagawa H, Wakabayashi-Nakao K, Tamura A, Toyoda Y, Koshiba S, Ishikawa T, 2009, Disruption ofN-linked glycosylation enhances ubiquitin-mediated proteasomal degradation of the human ATP-binding cassette transporter ABCG2. FEBS J 276(24): 7237–7252., DOI 10.1111/j.1742-4658.2009.07423.x Nakanishi T, Ross DD, 2012, Breast cancer resistance protein, BCRP/ABCG2): its role in multidrug resistance and regulation of its gene expression. Chinese J Cancer 31(2):73–99., DOI 10.5732 /cjc.011.10320 Nakagawa H, Tamura A, Wakabayashi K, Hoshijima K, Komada M, Yoshida T, Kometani S, Matsubara T, Mikuriya K, Ishikawa T, 2008, Ubiquitin-mediated proteasomal degradation of nonsynonymous SNP variants of human ABC transporter ABCG2. Biochem J 411(3):623–631., DOI 10.1042/bj20071229 Zhang MH, Man HT, Zhao XD, Dong NI, Ma SL, 2014, Estrogen receptor-positive breast cancermolecular signatures and therapeutic potentials, review). Biomed Rep 2(1):41–52., DOI 10.3892 /br.2013.187 Schraenen A, de Faudeur G, Thorrez L, Lemaire K, Van Wichelen G, Granvik M, Van Lommel L, in’t Veld P, Schuit F, 2010, mRNA expression analysis of cell cycle genes in islets of pregnant mice. Diabetologia 53(12):2579–2588., DOI 10.1007 /s00125-010-1912-8 Zhang H, Zhu X, Chen J, Jiang Y, Zhang Q, Kong C, Xing J, Ding L, Diao Z, Zhen X, Sun H, Yan G, 2015, Kruppel-like factor 12 is a novel negative regulator of forkhead box O1 expression: a potential role in impaired decidualization. Reprod Biol Endocrinol : RB&E 13:80., DOI 10.1186/s12958-015-0079-z Chester N, Kuo F, Kozak C, O’Hara CD, Leder P, 1998, Stagespecific apoptosis, developmental delay, and embryonic lethality in mice homozygous for a targeted disruption in the murine Bloom’s syndrome gene. Genes Dev 12(21):3382–3393 LiW, Cornell R, 2007, Redundant activities of Tfap2a and Tfap2c are required for neural crest induction and development of other non-neural ectoderm derivatives in zebrafish embryos. Dev Biol 304(1):338–354., DOI 10.1016/j.ydbio.2006.12.042 Piccinni SA, Bolcato-Bellemin A-L, Klein A, Yang VW, Kedinger M, Simon-Assmann P, Lefebvre O, 2004, Kruppel-like factors regulate the Lama1 Gene encoding the laminin α1 chain. J Biol Chem 279(10):9103–9114., DOI 10.1074/jbc.M305804200 Spencer TE, Bazer FW, 2004, Uterine and placental factors regulating conceptus growth in domestic animals. J Anim Sci 82(ESuppl): E4–13 Iwase T, TanakaM, SuzukiM, Naito Y, Sugimura H, Kino I, 1993, Identification of protein-tyrosine kinase genes preferentially expressed in embryo stomach and gastric cancer. Biochem Biophys Res Commun 194(2):698–705. doi :10.1006 /bbrc.1993.1878 Hayata T, Blitz IL, Iwata N, Cho KWY, 2009, Identification of embryonic pancreatic genes using Xenopus DNA microarrays. Dev Dyn 238(6):1455–1466., DOI 10.1002/dvdy.21868 Sewduth RN, Jaspard-Vinassa B, Peghaire C, Guillabert A, Franzl N, Larrieu-Lahargue F, Moreau C, Fruttiger M, Dufourcq P, Couffinhal T, Duplaa C, 2014, The ubiquitin ligase PDZRN3 is required for vascular morphogenesis throughWnt/planar cell polarity signalling. Nat Commun 5:4832., DOI 10.1038/ncomms5832 De Hertogh R, Ekka E, Vanderheyden I, Glorieux B, 1986, Estrogen and progestogen receptors in the implantation sites and interembryonic segments of rat uterus endometrium andmyometrium. Endocrinology 119(2):680–684., DOI 10.1210/endo-119-2-680 Larsen E, Gran C, Saether BE, Seeberg E, Klungland A, 2003, Proliferation failure and gamma radiation sensitivity of Fen1 null mutant mice at the blastocyst stage. Mol Cell Biol 23(15):5346– 5353., DOI 10.1128/MCB.23.15.5346-5353.2003 Wang T, Ong P, Roscioli T, Cliffe ST, Church JA, 2012, Hepatic veno-occlusive disease with immunodeficiency, VODI): first reported case in the U.S. and identification of a unique mutation in Sp110. Clin Immunol, Orlando, Fla, 145(2):102–107., DOI 10.1016 /j.clim.2012.07.016 Horm TM, Schroeder JA, 2013, MUC1 and metastatic cancer: expression, function and therapeutic targeting. Cell Adhes Migr 7(2):187–198., DOI 10.4161/cam.23131 Alam M, Rajabi H, Ahmad R, Jin C, Kufe D, 2014, Targeting the MUC1-C oncoprotein inhibits self-renewal capacity of breast cancer cells. Oncotarget 5(9):2622–2634., DOI 10.18632 /oncotarget.1848 Uhlen M, Bjorling E, Agaton C, Szigyarto CA, Amini B, Andersen E, Andersson AC, Angelidou P, Asplund A, Asplund C, Berglund L, Bergstrom K, Brumer H, Cerjan D, Ekstrom M, Elobeid A, Eriksson C, Fagerberg L, Falk R, Fall J, Forsberg M, Bjorklund MG, Gumbel K, Halimi A, Hallin I, Hamsten C, Hansson M, Hedhammar M, Hercules G, Kampf C, Larsson K, Lindskog M, Lodewyckx W, Lund J, Lundeberg J, Magnusson K, Malm E, Nilsson P, Odling J, Oksvold P, Olsson I, Oster E, Ottosson J, Paavilainen L, Persson A, Rimini R, Rockberg J, Runeson M, Sivertsson A, Skollermo A, Steen J, Stenvall M, Sterky F, Stromberg S, Sundberg M, Tegel H, Tourle S, Wahlund E, Walden A, Wan J, Wernerus H, Westberg J, Wester K, Wrethagen U, Xu LL, Hober S, Ponten F, 2005, A human protein atlas for normal and cancer tissues based on antibody proteomics. Mol Cell Proteomics : MCP 4(12):1920–1932., DOI 10.1074/mcp.M500279-MCP200 Uhlen M, Oksvold P, Fagerberg L, Lundberg E, Jonasson K, Forsberg M, Zwahlen M, Kampf C, Wester K, Hober S, Wernerus H, Bjorling L, Ponten F, 2010, Towards a knowledge-based human protein atlas. Nat Biotechnol 28(12):1248–1250., DOI 10.1038 /nbt1210-1248 Ponten F, Jirstrom K, Uhlen M, 2008, The human protein atlas–a tool for pathology. J Pathol 216(4):387–393., DOI 10.1002/path.2440 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 537 EP 544 DI 10.1007/s12253-016-0133-y PG 8 ER PT J AU Salzman, R Starek, I Kucerova, L Skalova, A AF Salzman, Richard Starek, Ivo Kucerova, Ladislava Skalova, Alena TI Differing Lymphatic Vessels Density in Salivary Adenoid Cystic Carcinoma and Pleomorphic Adenoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adenoid cystic carcinoma; Lymphangiogenesis; Lymphatic vessel; Pleomorphic adenoma; Salivary gland ID Adenoid cystic carcinoma; Lymphangiogenesis; Lymphatic vessel; Pleomorphic adenoma; Salivary gland AB Benign and malignant tumours are known to express various factors inducing lymphangiogenesis. Despite their different biological behaviour, salivary pleomorphic adenomas (PA) and adenoid cystic carcinomas (SACC) show similar lymphatic network. Authors compare density of lymphatic network in these tumours. The retrospective study included 20 SACC and 20 PA from salivary tumours. Lymphatic vessel density (LVD) was identified using D2-40 antibody and counted. In SACC, intratumoral, respectively peritumoral, lymphatic vessels were identified in 100 %, respectively 93.8 %, of cases. The intratumoral and peritumoral LVD did not significantly differ from each other. However, they both were higher than normal parenchyma density. In PA, intratumoral LVD, with a single exception, revealed values of 0 and 1. The intratumoral was found to be lower than peritumoral density. The LVD in healthy gland, similar to peritumoral one, was significantly higher than intratumoral values. Direct comparison showed intratumoral and peritumoral LVD in PA to be lower than in SACC. This study comparing LVD in PA and SACC revealed higher values in SACC, outnumbering those in healthy salivary parenchyma and PA. It suggests the capability of this biologically aggressive neoplasm to induce lymphangiogenesis. C1 [Salzman, Richard] Palacky University Olomouc, Faculty of Medicine and Dentistry, University Hospital Olomouc, Department of Otorhinolaryngology, I. P. Pavlova 6, 775 20 Olomouc, Czech Republic. [Starek, Ivo] Palacky University Olomouc, Faculty of Medicine and Dentistry, University Hospital Olomouc, Department of Otorhinolaryngology, I. P. Pavlova 6, 775 20 Olomouc, Czech Republic. [Kucerova, Ladislava] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular Pathology, I. P. Pavlova 6, 775 20 Olomouc, Czech Republic. [Skalova, Alena] Charles University, Faculty of Medicine in Plzen, Department of Pathology, Alej svobody 80, 304 60 Pilsen, Czech Republic. RP Salzman, R (reprint author), Palacky University Olomouc, Faculty of Medicine and Dentistry, University Hospital Olomouc, Department of Otorhinolaryngology, 775 20 Olomouc, Czech Republic. EM richard.salzman@fnol.cz CR Stacker SA, Achen MG, Jussila L, Baldwin ME, Alitalo K, 2002, Metastasis: lymphangiogenesis and cancer metastasis. Nat Rev Cancer 2(8):573–583 Lee SK, Kwon MS, Lee YS, Choi SH, Kim SY et al, 2012, Prognostic value of expression of molecular markers in adenoid cystic cancer of the salivary gland compares with lymph node metastasis: a retrospective study. World J Surg Oncol 10:266–280 Tanigawa N, Kanazawa T, Satomura K, Hikasa Y, Hashida M et al, 1981, Experimental study on lymphatic vascular changes in the development of cancer. Lymphology 14:149–154 Cao Y, 2005, Opinion: emerging mechanisms of tumour lymphangiogenesis and lymphatic metastasis. Nat Rev Cancer 5: 735–743 Stacker SA, Caesar C, Baldwin ME, Thornton GE, Williams RA et al, 2001, VEGF-Dpromotes themetastatic spread of tumors cells via lymphatics. Nat Med 7:186–191 Soares AB, de Araujo VC, Juliano PB, Altemani A, 2009, Angiogenic and lymphangiogenic microvessel density in recurrent pleomorphic adenoma. J Oral Pathol Med 38(8):623–629 Stennert E, Wittenkindt C, Klussmann JP, Arnold G, Guntinas- Lichius O, 2004, Recurrent pleomorphic adenoma of the parotid gland: a prospective histopathological and immunohistochemical study. Laryngoscope 114:1538–1543 Salzman R, Starek I, Kucerova L, Skalova A, Hoza J, 2014, Neither expression of VEGF-C/D nor lymph vessel density supports lymphatic invasion as the mechanism responsible for local spread of recurrent salivary pleomorphic adenoma. Virchows Arch 464:29–34 Fujita G, Sato S, Kishino M, Iwai SI, Nanazawa M et al, 2011, Lymphatic vessels and related factors in adenoid cystic carcinoma of the salivary gland. Mod Pathol 24(7):885–891 Spiro RH, 1997, Distant metastasis in adenoid cystic carcinoma of salivary origin. Am J Surg 174(5):495–498 Armstrong JG, Harrison LB, Thaler HT, Friedlander‐Klar H, Fass DE et al, 1992, The indications for elective treatment of the neck in cancer of the major salivary glands. Cancer 69:615–619 Woolger JA, Triantafullou A, 2011, Contemporary salivary clinical pathology. Facts and dilemmas. In: Bradley PJ, Guntinas-Lichius O, eds, Salivary gland disorders and diseases: diagnosis and management. Thieme Verlag, Stuttgart, p 2011 Szanto PA, Luna M, Tortoledo ME, White RA, 1984, Histologic grading of adenoid cystic carcinomas of the salivary glands. Cancer 54:1062–1069 Teymoortash A, Schrader C, Shimoda H, Kato S,Werner JA, 2007, Evidence of lymphangiogenesis in Warthin’s tumor of the parotid gland. Oral Oncol 43:614–618 Soares AB, Ponchio L, Juliano PB, de Araujo VC, Altemani A, 2007, Lymphatic vascular density and lymphangiogenesis during tumor progression of carcinoma ex pleomorphic adenoma. J Clin Pathol 60(9):995–1000 Williams CS, Leek RD, Robson AM, Banerji S, Prevo R, 2003, Absence of lymphangiogenesis and intratumoural lymph vessels in human metastatic breast cancer. J Pathol 200:195–206 Schoppmann SF, Horvat R, Birner P, 2002, Lymphatic vessels and lymphangiogenesis in female cancer: mechanisms, clinical impact and possible implications for anti-lymphangiogenic therapies, Review). Oncol Rep 9:455–460 Schmid K, Birner P, Gravenhorst V, End A, Geleff S, 2005, Prognostic value of lymphatic and blood vessels invasion in neuroendocrine tumors of the lung. Am J Surg Pathol 29:324–328 Kyzas PA, Geleff S, Batistatou A, Agnantis NJ, Stefanou D, 2005, Evidence for lymphangiogenesis and its prognostic implications in head and neck squamous cell carcinoma. J Pathol 206:170–177 Mello MF, Costa AF, Freitas LL, Soares AB, de Araujo VC, 2000, Lymphatic vessel density and expressions of lymphangiogenic growth factors in salivary carcinomas. Neoplasma 58(4):331–336 Carmeliet P, Jain RK, 2000, Angiogenesis in cancer and other diseases. Nature 407:249–257 Padera TP, Kadambi A, di Tomaso E, Carreira CM, Brown EB et al, 2002, Lymphatic metastasis in the absence of functional intratumoral lymphatics. Science 296:1883–1886 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 545 EP 550 DI 10.1007/s12253-016-0147-5 PG 6 ER PT J AU Enkner, F Pichlhofer, B Zaharie, TA Krunic, M Holper, MT Janik, S Moser, B Schlangen, K Neudert, B Walter, K Migschitz, B Mullauer, L AF Enkner, Franz Pichlhofer, Bettina Zaharie, Teodor Alexandru Krunic, Milica Holper, Maria Tina Janik, Stefan Moser, Bernhard Schlangen, Karin Neudert, Barbara Walter, Karin Migschitz, Brigitte Mullauer, Leonhard TI Molecular Profiling of Thymoma and Thymic Carcinoma: Genetic Differences and Potential Novel Therapeutic Targets SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thymoma; Thymic carcinoma; Mutation; miRNA; Immunohistochemistry ID Thymoma; Thymic carcinoma; Mutation; miRNA; Immunohistochemistry AB Thymoma and thymic carcinoma are thymic epithelial tumors (TETs). We performed a molecular profiling to investigate the pathogenesis of TETs and identify novel targets for therapy. We analyzed 37 thymomas (18 type A, 19 type B3) and 35 thymic carcinomas. The sequencing of 50 genes detected nonsynonymous mutations in 16 carcinomas affecting ALK, ATM, CDKN2A, ERBB4, FGFR3, KIT, NRAS and TP53. Only two B3 thymomas had a mutation in noncoding regions of the SMARCB1 and STK11 gene respectively. Three type A thymomas harbored a nonsynonymous HRAS mutation. Fluorescence in situ hybridization detected in 38 % of carcinomas a CDKN2A, in 32 % a TP53 and in 8 % an ATM gene deletion, whereas only one B3 thymoma exhibited a CDKNA deletion, and none of the type A thymomas showed a gene loss. Sequencing of the total miRNA pool of 5 type A thymomas and 5 thymic carcinomas identified the C19MC miRNA cluster as highly expressed in type A thymomas, but completely silenced in thymic carcinomas. Furthermore, the miRNA cluster C14MC was downregulated in thymic carcinomas. Among non-clustered miRNAs, the upregulation of miR-21, miR-9- 3 and miR-375 and the downregulation of miR-34b, miR-34c, miR-130a and miR-195 in thymic carcinomas were most significant. The expression of ALK, HER2, HER3, MET, phospho-mTOR, p16INK4A, PDGFRA, PDGFRB, PD-L1, PTEN and ROS1 was investigated by immunohistochemistry. PDGFRA was increased in thymic carcinomas and PD-L1 in B3 thymomas and thymic carcinomas. In summary, our results reveal genetic differences between thymomas and thymic carcinomas and suggest potential novel targets for therapy. C1 [Enkner, Franz] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1090 Vienna, Austria. [Pichlhofer, Bettina] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1090 Vienna, Austria. [Zaharie, Teodor Alexandru] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1090 Vienna, Austria. [Krunic, Milica] Medical University Vienna, Center for Medical Statistics, Informatics, and Intelligent SystemsVienna, Austria. [Holper, Maria Tina] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1090 Vienna, Austria. [Janik, Stefan] Medical University of Vienna, Department of Thoracic Surgery, Wahringer Gurtel 18-20, 1090 Vienna, Austria. [Moser, Bernhard] Medical University of Vienna, Department of Thoracic Surgery, Wahringer Gurtel 18-20, 1090 Vienna, Austria. [Schlangen, Karin] Medical University Vienna, Center for Medical Statistics, Informatics, and Intelligent Systems, Wahringer Gurtel 18-20, 1090 Vienna, Austria. [Neudert, Barbara] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1090 Vienna, Austria. [Walter, Karin] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1090 Vienna, Austria. [Migschitz, Brigitte] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1090 Vienna, Austria. [Mullauer, Leonhard] Medical University of Vienna, Department of Pathology, Wahringer Gurtel 18-20, 1090 Vienna, Austria. RP Mullauer, L (reprint author), Medical University of Vienna, Department of Pathology, 1090 Vienna, Austria. EM leonhard.muellauer@meduniwien.ac.at CR TravisWD, Brambilla E, Burke AP, Marx A, Nicholson AG, Eds.,, 2015, WHO classification of tumours of the lung, pleura, thymus and heart. IARC Press, Lyon Kelly RJ, Petrini I, Rajan A, Wang Y, Giaccone G, 2011, Thymic malignancies: from clinical management to targeted therapies. J Clin Oncol: Off J Am Soc Clin Oncol 29(36):4820–4827., DOI 10.1200/Jco.2011.36.0487 Schirosi L, Nannini N, Nicoli D, Cavazza A, Valli R, Buti S, Garagnani L, Sartori G, Calabrese F, Marchetti A, Buttitta F, Felicioni L, Migaldi M, Rea F, Di Chiara F, Mengoli M, Rossi G, 2012, Activating c-KIT mutations in a subset of thymic carcinoma and response to different c-KIT inhibitors. Ann Oncol: Off J Eur Soc Med Oncol / ESMO 23(9):2409–2414.doi:10.1093 /Annonc/Mdr626 Yoh K, Nishiwaki Y, Ishii G, Goto K, Kubota K, Ohmatsu H, Niho S, Nagai K, Saijo N, 2008, Mutational status of EGFR and KIT in thymoma and thymic carcinoma. Lung Cancer 62(3):316–320 Petrini I, Meltzer PS, Kim IK, Lucchi M, Park KS, Fontanini G, Gao J, Zucali PA, Calabrese F, Favaretto A, Rea F, Rodriguez- Canales J, Walker Rl, Pineda M, Zhu YJ, Lau C, Killian KJ, Bilke S, Voeller D, Dakshanamurthy S, Wang Y, Giaccone G, 2014, A specific missense mutation in GTF2I occurs at high frequency in thymic epithelial tumors. Nat Genet 46, 8):844–849., DOI 10.1038/Ng.3016. http://Www.Nature.Com/Ng/Journal/V46 /N8/Abs/Ng.3016.Html-Supplementary-Information Wang Y, Thomas A, Lau C, Rajan A, Zhu Y, Killian JK, Petrini I, Pham T, Morrow B, Zhong X, Meltzer PS, Giaccone G, 2014, Mutations of epigenetic regulatory genes are common in thymic carcinomas. Sci Rep 4:7336., DOI 10.1038/Srep07336 Moreira AL, Won HH, McMillan R, Huang J, Riely GJ, Ladanyi M, Berger MF, 2015, Massively parallel sequencing identifies recurrent mutations in TP53 in thymic carcinoma associated with poor prognosis. J Thorac Oncol: Off Publ Int Assoc Study Lung Cancer 10(2):373–380., DOI 10.1097/Jto.0000000000000397 Shitara M, Okuda K, Suzuki A, Tatematsu T, Hikosaka Y, Moriyama S, Sasaki H, Fujii Y, Yano M, 2014, Genetic profiling of thymic carcinoma using targeted next-generation sequencing. Lung Cancer 86(2):174–179., DOI 10.1016/J.Lungcan.2014.08.020 Robinson MD, McCarthy DJ, Smyth GK, 2010, edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26(1):139–140., DOI 10.1093 /Bioinformatics/Btp616 Nonaka D, Rosai J, 2012, Is there a spectrum of cytologic atypia in type A thymomas analogous to that seen in type B thymomas? A pilot study of 13 cases. Am J Surg Pathol 36(6):889–894., DOI 10.1097/Pas.0b013e31824fff50 Musgrove EA, Caldon CE, Barraclough J, Stone A, Sutherland RL, 2011, Cyclin D as a therapeutic target in cancer. Nat Rev Cancer 11(8):558–572., DOI 10.1038/Nrc3090 Petrini I, Meltzer PS, Zucali PA, Luo J, Lee C, Santoro A, Lee HS, Killian KJ,WangY, TsokosM, RoncalliM, Steinberg SM,WangY, Giaccone G, 2012, Copy number aberrations of BCL2 and CDKN2A/B identified by array-CGH in thymic epithelial tumors. Cell Death Dis 3:E351. http://Www.Nature.Com/Cddis/Journal/V3 /N7/Suppinfo/Cddis201292s1.Html Yamamoto H, Oda Y, 2015, Gastrointestinal stromal tumor: recent advances in pathology and genetics. Pathol Int 65(1):9–18., DOI 10.1111/Pin.12230 Strobel P, Hartmann M, Jakob A, Mikesch K, Brink I, Dirnhofer S, Marx A, 2004, Thymic carcinoma with overexpression of mutated KIT and the response to imatinib. N Engl J Med 350(25):2625– 2626., DOI 10.1056/Nejm200406173502523 Bisagni G, Rossi G, Cavazza A, Sartori G, Gardini G, Boni C, 2009, Long lasting response to the multikinase inhibitor bay 43- 9006, Sorafenib, in a heavily pretreated metastatic thymic carcinoma. J Thorac Oncol: Off Publ Int Assoc Study Lung Cancer 4(6): 773–775., DOI 10.1097/Jto.0b013e3181a52e25 Dienstmann R, Rodon J, Prat A, Perez-Garcia J, Adamo B, Felip E, Cortes J, Iafrate AJ, Nuciforo P, Tabernero J, 2014, Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors. Ann Oncol: Off J Eur Soc Med Oncol / ESMO 25(3): 552–563., DOI 10.1093/Annonc/Mdt419 Liu X, Zhang W, Geng D, He J, Zhao Y, Yu L, 2014, Clinical significance of fibroblast growth factor receptor-3 mutations in bladder cancer: a systematic review and meta-analysis. Genet Mol Res: Gmr 13(1):1109–1120., DOI 10.4238/2014.February.20.12 Ho HK, Yeo AH, Kang TS, Chua BT, 2014, Current strategies for inhibiting FGFR activities in clinical applications: opportunities, challenges and toxicological considerations. Drug Discov Today 19(1):51–62., DOI 10.1016/J.Drudis.2013.07.021 Hallberg B, Palmer RH, 2013, Mechanistic insight into ALK receptor tyrosine kinase in human cancer biology. Nat Rev Cancer 13(10):685–700., DOI 10.1038/Nrc3580 Epstein LF, Chen H, Emkey R, Whittington DA, 2012, The R1275Q neuroblastomamutant and certain ATP-competitive inhibitors stabilize alternative activation loop conformations of anaplastic lymphoma kinase. J Biol Chem 287(44):37447–37457., DOI 10.1074/Jbc.M112.391425 Modjtahedi H, Cho BC, Michel MC, Solca F, 2014, A comprehensive review of the preclinical efficacy profile of the ErbB family blocker afatinib in cancer. Naunyn Schmiedeberg’s Arch Pharmacol 387(6):505–521., DOI 10.1007/S00210-014-0967-3 Prickett TD, Agrawal NS, Wei X, Yates KE, Lin JC, Wunderlich JR, Cronin JC, Cruz P, Rosenberg SA, Samuels Y, 2009, Analysis of the tyrosine kinome in melanoma reveals recurrent mutations in ERBB4. Nat Genet 41(10):1127–1132., DOI 10.1038/Ng.438 Michels J, Vitale I, Saparbaev M, Castedo M, Kroemer G, 2014, Predictive biomarkers for cancer therapy with PARP inhibitors. Oncogene 33(30):3894–3907., DOI 10.1038/Onc.2013.352 Williamson CT, Muzik H, Turhan AG, Zamo A, O’Connor MJ, Bebb DG, Lees-Miller SP, 2010, ATMdeficiency sensitizes mantle cell lymphoma cells to poly(ADP-ribose, polymerase-1 inhibitors. Mol Cancer Ther 9(2):347–357., DOI 10.1158/1535-7163.Mct-09- 0872 Weston VJ, Oldreive CE, Skowronska A, Oscier DG, Pratt G, Dyer MJ, Smith G, Powell JE, Rudzki Z,Kearns P, Moss PA, TaylorAM, Stankovic T, 2010, The PARP inhibitor olaparib induces significant killing of ATM-deficient lymphoid tumor cells in vitro and in vivo. Blood 116(22):4578–4587., DOI 10.1182/Blood-2010-01-265769 Stephen AG, Esposito D, Bagni RR, McCormick F, 2014, Dragging ras back in the ring. Cancer Cell 25(3):272–281., DOI 10.1016/J.Ccr.2014.02.017 Singh H, Longo DL, Chabner BA, 2015, Improving prospects for targeting RAS. J Clin Oncol: Off J Am Soc Clin Oncol 33(31): 3650–3659., DOI 10.1200/Jco.2015.62.1052 Noguer-Dance M, Abu-Amero S, Al-Khtib M, Lefevre A, Coullin P, Moore GE, Cavaille J, 2010, The primate-specific microRNA gene cluster, C19MC, is imprinted in the placenta. Hum Mol Genet 19(18):3566–3582., DOI 10.1093/Hmg/Ddq272 Morales-Prieto DM, Ospina-Prieto S, Chaiwangyen W, Schoenleben M, Markert UR, 2013, Pregnancy-associated miRNA-clusters. J Reprod Immunol 97(1):51–61., DOI 10.1016/J. Jri.2012.11.001 Kleinman CL, Gerges N, Papillon-Cavanagh S, Sin-Chan P, Pramatarova A, Quang DAK, Adoue V, Busche S, Caron M, Djambazian H, Bemmo A, Fontebasso AM, Spence T, Schwartzentruber J, Albrecht S, Hauser P, Garami M, Klekner A, Bognar L, Montes JL, Staffa A, Montpetit A, Berube P, Zakrzewska M, Zakrzewski K, Liberski PP, Dong Z, Siegel PM, Duchaine T, Perotti C, Fleming A, Faury D, Remke M, Gallo M, Dirks P, Taylor MD, Sladek R, Pastinen T, Chan JA, Huang A, Majewski J, Jabado N, 2014, Fusion of TTYH1 with the C19MC microRNA cluster drives expression of a brain-specific DNMT3B isoformin the embryonal brain tumor ETMR. Nat Genet 46, 1):39– 4 4 . d o i : 1 0 . 1 0 3 8 / N g . 2 8 4 9 . h t t p ://Www. N a t u r e . Com/Ng/Journal/V46/N1/Abs/Ng.2849.Html-Supplementary- Information Korshunov A, Remke M, Gessi M, Ryzhova M, Hielscher T, Witt H, Tobias V, Buccoliero AM, Sardi I, Gardiman MP, Bonnin J, Scheithauer B, Kulozik AE, Witt O, Mork S, Von Deimling A, Wiestler OD, Giangaspero F, Rosenblum M, Pietsch T, Lichter P, Pfister SM, 2010, Focal genomic amplification at 19q13.42 comprises a powerful diagnostic marker for embryonal tumors with ependymoblastic rosettes. Acta Neuropathol 120(2):253–260., DOI 10.1007/S00401-010-0688-8 Cairo S, Wang Y, De Reynies A, Duroure K, Dahan J, Redon MJ, Fabre M, McClelland M, Wang XW, Croce CM, Buendia MA, 2010, Stem cell-like micro-RNA signature driven by Myc in aggressive liver cancer. Proc Natl Acad Sci U S A 107(47):20471– 20476., DOI 10.1073/Pnas.1009009107 Flor I, Bullerdiek J, 2012, The dark side of a success story: microRNAs of the C19MC cluster in human tumours. J Pathol 227(3):270–274., DOI 10.1002/Path.4014 Rippe V, Dittberner L, Lorenz VN, Drieschner N, Nimzyk R, Sendt W, Junker K, Belge G, Bullerdiek J, 2010, The two stem cell microRNA gene clusters C19MC and miR-371-3 are activated by specific chromosomal rearrangements in a subgroup of thyroid adenomas. PLoS One 5(3), E9485., DOI 10.1371/Journal. Pone.0009485 Vaira V, Elli F, Forno I, Guarnieri V, Verdelli C, Ferrero S, Scillitani A, Vicentini L, Cetani F,Mantovani G, Spada A, Bosari S, Corbetta S, 2012, The microRNA cluster C19MC is deregulated in parathyroid tumours. J Mol Endocrinol 49(2):115–124., DOI 10.1530/Jme-11-0189 RadovichM, Solzak JP, Hancock BA, Conces ML, Atale R, Porter RR, Zhu J, Glasscock J, Kesler KA, Badve SS, Schneider BP, Loehrer PJ, 2016, A large microRNA cluster on chromosome 19 is a transcriptional hallmark ofWHO type A and AB thymomas. Br J Cancer 114(4):477–484., DOI 10.1038/Bjc.2015.425 Ucar O, Tykocinski LO, Dooley J, Liston A, Kyewski B, 2013, An evolutionarily conserved mutual interdependence between Aire and microRNAs in promiscuous gene expression. Eur J Immunol 43(7): 1769–1778., DOI 10.1002/Eji.201343343 Haller F, Von Heydebreck A, Zhang JD, Gunawan B, Langer C, Ramadori G, Wiemann S, Sahin O, 2010, Localization-and mutation- dependent microRNA, miRNA, expression signatures in gastrointestinal stromal tumours, GISTs), with a cluster of coexpressed miRNAs located at 14q32.31. J Pathol 220(1):71–86., DOI 10.1002/Path.2610 Lavon I, Zrihan D, Granit A, Einstein O, Fainstein N, Cohen M, Cohen M, Zelikovitch B, Shoshan Y, Spektor S, Reubinoff BE, Felig Y, Gerlitz O, Ben-Hur T, Smith Y, Siegal T, 2010, Gliomas display a microRNA expression profile reminiscent of neural precursor cells. Neuro-Oncology 12(5):422–433., DOI 10.1093 /Neuonc/Nop061 Cortez MA, Ivan C, Valdecanas D, Wang X, Peltier HJ, Ye Y, Araujo L, Carbone DP, Shilo K, Giri DK, Kelnar K, Martin D, Komaki R, Gomez DR, Krishnan S, Calin GA, Bader AG, Welsh JW, 2016, PDL1 Regulation by p53 via miR-34. J Natl Cancer Inst 108(1)., DOI 10.1093/Jnci/Djv303 Li B, Huang P, Qiu J, Liao Y, Hong J, Yuan Y, 2014, MicroRNA- 130a is down-regulated in hepatocellular carcinoma and associates with poor prognosis. Med Oncol 31(10):230., DOI 10.1007/S12032- 014-0230-2 Jia LF, Wei SB, Gong K, Gan YH, Yu GY, 2013, Prognostic implications of micoRNA miR-195 expression in human tongue squamous cell carcinoma. PLoS One 8(2), E56634., DOI 10.1371 /Journal.Pone.0056634 Liu L, Chen L, Xu Y, Li R, Du X, 2010, MicroRNA-195 promotes apoptosis and suppresses tumorigenicity of human colorectal cancer cells. Biochem Biophys Res Commun 400(2):236–240., DOI 10.1016/J.Bbrc.2010.08.046 Krichevsky AM, Gabriely G, 2009, miR-21: a small multi-faceted RNA. J Cell Mol Med 13(1):39–53., DOI 10.1111/J.1582- 4934.2008.00556.X Shen L, Wan Z, Ma Y, Wu L, Liu F, Zang H, Xin S, 2015, The clinical utility of microRNA-21 as novel biomarker for diagnosing human cancers. Tumour Biol: J Int Soc Oncodevelop Biol Med 36(3):1993–2005., DOI 10.1007/S13277-014-2806-Z Heller G, Weinzierl M, Noll C, Babinsky V, Ziegler B, Altenberger C, Minichsdorfer C, Lang G, Dome B, End- Pfutzenreuter A, Arns BM, Grin Y, Klepetko W, Zielinski C, Zochbauer-Muller S, 2012, Genome-wide miRNA expression profiling identifies miR-9-3 and miR-193a as targets for DNA methylation in non-small cell lung cancers. Clin Cancer Res: Off J Am Assoc Cancer Res 18(6):1619–1629., DOI 10.1158/1078-0432.Ccr-11-2450 Poy MN, Eliasson L, Krutzfeldt J, Kuwajima S, Ma X,MacDonald PE, Pfeffer S, Tuschl T, Rajewsky N, Rorsman P, Stoffel M, 2004, A pancreatic islet-specific microRNA regulates insulin secretion. Nature 432(7014):226–230., DOI 10.1038/Nature03076 Yan JW, Lin JS, He XX, 2014, The emerging role of miR-375 in cancer. Int J Cancer 135, 5):1011–1018., DOI 10.1002/ijc.28563 Weissferdt A, Lin H, Woods D, Tang X, Fujimoto J, Wistuba II, Moran CA, 2012, HER family receptor and ligand status in thymic carcinoma. Lung Cancer 77(3):515–521., DOI 10.1016/J. Lungcan.2012.05.108 Pan CC, Chen PCH, Wang LS, Lee JY, Chiang H, 2003, Expression of apoptosis-related markers and HER-2/neu in thymic epithelial tumours. Histopathology 43(2):165–172 Zhang Y, Du Z, ZhangM, 2016, Biomarker development in METtargeted therapy. Oncotarget 7(24):37370–37389, DOI 10.18632 /Oncotarget.8276 Awad MM, Oxnard GR, Jackman DM, Savukoski DO, Hall D, Shivdasani P, Heng JC, Dahlberg SE, Janne PA, Verma S, Christensen J, Hammerman PS, Sholl LM, 2016, MET exon 14 mutations in non-small-cell lung cancer are associated with advanced age and stage-dependent MET genomic amplification and c-MET overexpression. J Clin Oncol: Off J Am Soc Clin Oncol 34(7):721–730., DOI 10.1200/Jco.2015.63.4600 Xu K, Liu P, Wei W, 2014, mTOR signaling in tumorigenesis. Biochimica Et Biophysica Acta 1846(2):638–654., DOI 10.1016/J. Bbcan.2014.10.007 Cimpean AM, Ceausu R, Encica S, Gaje PN, Ribatti D, Raica M, 2011, Platelet-derived growth factor and platelet-derived growth factor receptor-alpha expression in the normal human thymus and thymoma. Int J Exp Pathol 92(5):340–344., DOI 10.1111/J.1365- 2613.2011.00777.X Meister M, Kahl P, Muley T, Morresi-Hauf A, Sebening C, Kern MA, Breinig M, Schnabel P, Dienemann H, Schirmacher P, Rieker RJ, 2009, Expression and mutational status of PDGFR in thymic tumours. Anticancer Res 29(10):4057–4061 Strobel P, Bargou R, Wolff A, Spitzer D, Manegold C, Dimitrakopoulou-Strauss A, Strauss L, Sauer C, Mayer F, Hohenberger P, Marx A, 2010, Sunitinib in metastatic thymic carcinomas: laboratory findings and initial clinical experience. Br J Cancer 103(2):196–200., DOI 10.1038/Sj.Bjc.6605740 Pagano M, Sierra NM, Panebianco M, Rossi G, Gnoni R, Bisagni G, Boni C, 2014, Sorafenib efficacy in thymic carcinomas seems not to require c-KIT or PDGFR-alpha mutations. Anticancer Res 34(9):5105–5110 Witkiewicz AK, Knudsen KE, Dicker AP, Knudsen ES, 2011, The meaning of p16(Ink4a, expression in tumors: functional significance, clinical associations and future developments. Cell Cycle 10(15):2497–2503 Zou W, Wolchok JD, Chen L, 2016, PD-L1, B7-H1, and PD-1 pathway blockade for cancer therapy: mechanisms, response biomarkers, and combinations. Sci Transl Med 8(328):328rv324., DOI 10.1126/Scitranslmed.Aad7118 Topalian SL, Taube JM, Anders RA, Pardoll DM, 2016, Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 16(5):275–287., DOI 10.1038 /Nrc.2016.36 Chen L, Han X, 2015, Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest 125(9):3384–3391., DOI 10.1172/Jci80011 Padda SK, Riess JW, Schwartz EJ, Tian L, Kohrt HE, Neal JW, West RB, Wakelee HA, 2015, Diffuse high intensity PD-L1 staining in thymic epithelial tumors. J Thorac Oncol: Off Publ Int Assoc Study Lung Ca n c e r 10(3):500–508. d o i :10.1097 /Jto.0000000000000429 Katsuya Y, Fujita Y, Horinouchi H, Ohe Y, Watanabe S, Tsuta K, 2015, Immunohistochemical status of PD-L1 in thymoma and thymic carcinoma. Lung Cancer 88(2):154–159., DOI 10.1016/J. Lungcan.2015.03.003 Patel SP, Kurzrock R, 2015, PD-L1 expression as a predictive biomarker in cancer immunotherapy.Mol Cancer Ther 14(4):847–856., DOI 10.1158/1535-7163.Mct-14-0983 Carracedo A, Alimonti A, Pandolfi PP, 2011, PTEN level in tumor suppression: how much is too little? Cancer Res 71(3):629–633., DOI 10.1158/0008-5472.Can-10-2488 Roh MR, Gupta S, Park KH, Chung KY, Lauss M, Flaherty KT, Jonsson G, Rha SY, Tsao H, 2016, Promoter methylation of PTEN is a significant prognostic factor in melanoma survival. J Invest Dermatol 136(5):1002–1011., DOI 10.1016/J.Jid.2016.01.024 Ye M, Zhang X, Li N, Zhang Y, Jing P, Chang N, Wu J, Ren X, Zhang J, 2016, ALK and ROS1 as targeted therapy paradigms and clinical implications to overcome crizotinib resistance. Oncotarget 7(11):12289–12304., DOI 10.18632/Oncotarget.6935 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 551 EP 564 DI 10.1007/s12253-016-0144-8 PG 14 ER PT J AU Zidi, S Stayoussef, M Alsaleh, LB Gazouani, E Mezlini, A Ebrahim, HB Yacoubi-Loueslati, B Almawi, YW AF Zidi, Sabrina Stayoussef, Mouna Alsaleh, L Bano Gazouani, Ezzedine Mezlini, Amel Ebrahim, H Bashayer Yacoubi-Loueslati, Besma Almawi, Y Wassim TI Effect of Follicle Stimulating Hormone Receptor Gene Polymorphisms in Cervical Cancer Risk SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE FSHR; Cervical cancer; FIGO stages; Polymorphisms; Haplotypes; Tunisians ID FSHR; Cervical cancer; FIGO stages; Polymorphisms; Haplotypes; Tunisians AB For the first time in the word, we investigated the association between five FSHR polymorphisms with the risk of cervical cancer among Tunisians. Study subjects comprised 112 Cervical Cancer (CC) patients and 164 control women. Genotyping of FSHR rs6166, rs1007541, rs11692782, rs2055571 and rs1394205 variants was done by realtime PCR, with defined clusters. The allelic distributions of the tested FSHR SNPs were comparable between CC patients and control women. In contrast, the heterozygous genotype of rs1007541 was associated with 1.8-fold increased risk of CC. Stratification according to FIGO staging revealed that the minor allele of rs1007541 was more frequent among advanced tumor stage patients, with 11-fold increased risk of CC [P < 0.0001; OR (95 % CI) = 11.32 (7.46–17.18)]. However, no significant allelic association was revealed in the rest of analyzed FSHR SNPs. Haploview analysis showed high Linkage disequilibrium (LD) between rs2055571 and rs1394205. Haplotype analysis revealed a lack of association between cases and controls. However, analysis of CC patient subgroups demonstrated enrichment of GGTAG haplotype in early tumor stage [P = 0.025; OR (95 % CI) = 0.07 (0.01– 0.70)]. The FSHR variants and haplotypes may be a genetic markers for CC susceptibility and evolution among Tunisian women. C1 [Zidi, Sabrina] El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 1092 Tunis, Tunisia. [Stayoussef, Mouna] El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 1092 Tunis, Tunisia. [Alsaleh, L Bano] Arabian Gulf University, Department of Medical BiochemistryManama, Bahrain. [Gazouani, Ezzedine] Military Hospital of Tunis, Laboratory of ImmunologyTunis, Tunisia. [Mezlini, Amel] Salah Azeiz Oncology InstituteTunis, Tunisia. [Ebrahim, H Bashayer] Arabian Gulf University, Department of Medical BiochemistryManama, Bahrain. [Yacoubi-Loueslati, Besma] El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 1092 Tunis, Tunisia. [Almawi, Y Wassim] Arabian Gulf University, Department of Medical BiochemistryManama, Bahrain. RP Zidi, S (reprint author), El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 1092 Tunis, Tunisia. EM ZIDISABRINA86@gmail.com CR Conway GS, Conway E, Walker C, HoEppner W, Gromoll J, Simoni M, 1999, Mutation screening and isoform prevalence of the follicle stimulating hormone receptor gene in women with premature ovarian failure, resistant ovary syndrome and polycystic ovary syndrome. Clin Endocrinol 51:97–99 Gromoll J, Pekel E, Nieschlag E, 1996a, The structure and organization of the human follicle-stimulating hormone receptor, FSHR, gene. Genomics 35:308–311 Chappel SC, 1995, Heterogeneity of follicle stimulating hormone: control and physiological function. Hum Reprod Update 1(5):479– 487 SimoniM GJ, Nieschlag E, 1997, The follicle-stimulating hormone receptor: biochemistry, molecular biology, physiology, and pathophysiology. Endocr Rev 18(6):739–773 Hsueh AJW, Bicsak TA, Jia XC, Dahl KD, Fauser BC, Galway AB, Czekala N, Pavlou SN, Papkoff H, Keene J et al, 1989, Granulosa cells as hormone targets: the role of biologically active folliclestimulating hormone in reproduction. Recent Prog Horm Res 45:209–277 Richards JS, 1980, Maturation of ovarian follicles: actions and interactions of pituitary and ovarian hormones on follicular cell differentiation. Physiol Rev 60:51–89 Salesse R, Remy JJ, Levin JM, Jallal B, Garnier J, 1991, Towards understanding the glycoprotein hormone receptors. Biochimie 73: 109–120 Gromoll J, SimoniM, Nieschlag E, 1996b, An activating mutation of the folliclestimulating hormone receptor autonomously sustains spermatogenesis in a hypophysectomized man. J Clin Endocrinol Metab 81:1367–1370 Yuni A, Gromoll J,Wunsch A, Asatiani K, ZitzmannM, Nieschlag E, Simoni M, 2005, Follicle-stimulating hormone receptor gene haplotype distribution in normozoospermic and azoospermic men. Journal of Andrology 26(4):494–499 Simoni M, Nieschlag E, Gromoll J, 2002, Isoforms and single nucleotide polymorphisms of the FSH receptor gene: implications for human reproduction. Hum Reprod Update 8(5):413–421 Pengo M, Ferlin A, Arredi B, Ganz F, Selice R, Garolla A, Foresta C, 2006, FSH receptor gene polymorphisms in fertile and infertile Italian men. Reprod BioMed Online 13(6):795–800 Aittomaki K, Lucena JL, Pakarinen P, Sistonen P, Tapanainen J, Gromoll J et al, 1995, Mutation in the follicle-stimulating hormone receptor gene causes hereditary hypergonadotropic ovarian failure. Cell 82:959–968 Dierich A, Sairam MR, Monaco L, Fimia GM, Gansmuller A, LeMeur M et al, 1998, Impairing follicle-stimulating hormone, FSH, signaling in vivo: targeted disruption of the FSH receptor leads to aberrant gametogenesis and hormonal imbalance. Proc Natl Acad Sci U S A 95:13612–13617 Jiang M, Aittomaki K, Nilsson C, Pakarinen P, Iitia A, Torresani T et al, 1998, The frequency of an inactivating point mutation, 566C3T, of the human follicle-stimulating hormone receptor gene in four populations using allele-specific hybridization and timeresolved fluorometry. J Clin Endocrinol Metab 83:4338–4343 Tapanainen JS, Aittomaki K, Min J, Vaskivuo T, Huhtaniemi IT, 1997, Men homozygous for an inactivating mutation of the follicle-stimulating hormone, FSH, receptor gene present variable suppression of spermatogenesis and fertility. Nat Genet 15:205–206 Fuller PJ, Verity K, Shen Y, Mamers P, Jobling T, Burger HG, 1998, No evidence of a role for mutations or polymorphisms of the folliclestimulating hormone receptor in ovarian granulosa cell tumors. J Clin Endocrinol Metab 83(1):274–279 Yang CQ, Chan KY, Ngan HY, Khoo US, Chiu PM, Chan QK et al, 2006, Single nucleotide polymorphisms of follicle-stimulating hormone receptor are associated with ovarian cancer susceptibility. Carcinogenesis 27(7):1502–1506 Ludwig AH, Murawska M, Panek G, Timorek A, Kupryjanczyk J, 2009, Androgen, progesterone, and FSH receptor polymorphisms in ovarian cancer risk and outcome. Endocr Relat Cancer 16(3): 1005–1016 Xue Q, Ma L, Yang S, Zhao J, Chen S, Xie Y,Wang J, Li T, Yu H, Peng Q, Deng Y, Li S, Qin A, 2014, The Asn680Ser polymorphism of the follicle stimulating hormone receptor gene and ovarian cancer risk: a meta-analysis. J Assist Reprod Genet 31(6):683–688 Tang H, Yan Y, Wang T, Zhang T, Shi W, Fan R, Yao Y, Zhai S, 2015, Effect of follicle-stimulating hormone receptor Asn680Ser polymorphism on the outcomes of controlled ovarian hyperstimulation: an updated meta- analysis of 16 cohort studies. J Assist Reprod Genet 32(12):1801–1810 JemalA, Bray F,MMC, Ferlay J,Ward E, Forman D, 2011)Global cancer statistics. CA Cancer J Clin 61:69–90 Lorincz AT, Reid R, Jenson AB, Greenberg MD, Lancaster W, Kurman RJ, 1992, Human papillomavirus infection of the cervix: relative risk associations of 15 common anogenital types. Obstet Gynecol 79:328–337 Schiffman MH, Brinton LA, 1995, The epidemiology of cervical carcinogenesis. Cancer 76:1888–1901 Pinion SB, Kennedy JH, Miller RW, MacLean AB, 1991, Oncogene expression in cervical intraepithelial neoplasia and invasive cancer of cervix. Lancet 337:819–820 Tartour E, Fossiez F, Joyeux I, Galinha A, Gey A, Claret E, Sastre- Garau X, Couturier J, Mosseri V, Vives V, Banchereau J, Fridman WH,Wijdenes J, Lebecque S, Sautes-Fridman C, 1999, Interleukin 17, a T-cell-derived cytokine, promotes tumorigenicity of human cervical tumors in nude mice. Cancer Res 59:3698–3704 Siraj A, Desestret V, Antoine M, Fromont G, Huerre M, Sanson M, Camparo P, Pichon C, Planeix F, Gonin J, Radu A,Ghinea N, 2013, Expression of follicle-stimulating hormone receptor by the vascular endothelium in tumor metastases. BMC Cancer 13(1):1 Nawrocka J, Starczewski A, 2007, Effects of metformin treatment in women with polycystic ovary syndrome depends on insulin resistance. Genetic polymorphisms of FSHR, CYP17, CYP1A1, CAPN10, INSR, SERPINE1 genes in adolescent girls with polycystic ovary syndrome. Gynecol Endocrinol 23:231–237 Wu X, Sallinen K, Zhou S, Su Y, Pollanen P, Erkkola R, 2000, Androgen excess contributes to altered growth hormone/insulinlike growth factor-1 axis in nonobese women with polycystic ovary syndrome. Fertil Steril 73:730–734 Greb RR, Behre HM, Simoni M, 2005, Pharmacogenetics in ovarian stimulation – current concepts and future options. Reprod BioMed Online 11:589–600 Castro F, Ruiz R, Montoro L, Perez-Hernandez D, Sanchez-Casas Padilla E, Real LM, Ruiz A, 2003, Role of follicle-stimulating hormone receptor Ser680Asn polymorphism in the efficacy of follicle-stimulating hormone. Fertil Steril 80:571–576 Perez Mayorga M, Gromoll J, Behre HM, Gassner C, Nieschlag E, SimoniM(2000, Ovarian response to follicle-stimulating hormone, FSH, stimulation depends on the FSH receptor genotype. J Clin Endocrinol Metab 85:3365–3369 Ferlin A, Pengo M, Selice R, Salmaso L, Garolla A, Foresta C, 2008, Analysis of single nucleotide polymorphisms of FSH receptor gene suggests association with testicular cancer susceptibility. Endocrine-Related Cancer 15:429–437 Almawi WY, Hubail B, Arekat DZ, Al-Farsi SM, Al-Kindi SK, Arekat MR, Mahmood N, Madan S, 2015, Leutinizing hormone/ choriogonadotropin receptor and follicle stimulating hormone receptor gene variants in polycystic ovary syndrome. Journal of assisted reproduction and genetics 32(4):607–614 Tugba Unsal, Ece Konac, Ediz Yesilkaya, Akin Yilmaz, Aysun Bideci, Hacer Ilke Onen, Peyami Cinaz, Adnan Menevse, 2009, Journal of assisted reproduction and genetics 26(4):205–216. Martin Heubner, Kathrin Riemann, Friedrich Otterbach, Rainer Kimmig, Sabine Kasimir-Bauer, Winfried Siffert, Pauline Wimberger, 2009, The haplotype of two FSHR polymorphisms in ovarian cancer-A potential role of ethnology in risk modification Gynecologic Oncology 112:486–489. Ahda Y, Gromoll J, Wunsch A et al, 2005, Follicle-stimulating hormone receptor gene haplotype distribution in normozoospermic and azoospermic men. J Androl 26(4):494–499 Castro FJ,Mor’on L, Montoro et al, 2004, Human controlled ovarian hyperstimulation outcome i s a polygenic t r a i t . Pharmacogenetics 14(5):285–293 Sudo S, Kudo M,Wada S, Sato O, Hsueh AJW, Fujimoto S, 2002, Genetic and functional analyses of polymorphisms in the human FSH receptor gene. Mol Hum Reprod 8(10):893–899 Ma L, Chen Y, Mei S, Liu C, Ma X, Li Y, Jiang Y, Ha L, Xu X, 2015, Single nucleotide polymorphisms in premature ovarian failure associated genes in a chinese hui population. Molecular Medicine Reports 12:2529–2538 Loutradis D, Patsoula E, Minas V, Giorgos A, Koussidis AA, Michalas S, Makrigiannakis A, 2006, FSH receptor gene polymorphisms have a role for different ovarian response to stimulation in patients entering IVF/ICSI-ET programs. J Assist Reprod Genet 23(4):177–184 Nakayama T, Kuroi N, Sano M, Tabara Y, Katsuya T, Ogihara T, Makita Y, Hata A, Yamada M, Takahashi N, Hirawa N, Umemura S, Miki T, Soma M, 2006, Mutation of the follicle-stimulating hormone receptor Gene 5-Untranslated region associated with female. Hypertension 48(3):512–518 Hea Young O, Kim MK, Seo S-S, Lee J-K, 2016, Association of combined tobacco smoking and oral contraceptive use with cervical intraepithelial neoplasia 2 or 3 in Korean women. Journal of Epidemiology 26(1):22–29 Zidi S, Stayoussef M, Alsaleh BL, Gazouani E,Mezlini A, Ebrahim BH, Yacoubi-Loueslati B, Almawi WY, 2016, Relationships between common and novel interleukin-6 Gene polymorphisms and risk of cervical cancer: a case-control study. Pathol Oncol Res., DOI 10.1007/s12253-016-0127-9 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 565 EP 572 DI 10.1007/s12253-016-0152-8 PG 8 ER PT J AU Zhang, Y Zhou, J Sun, M Sun, G Cao, Y Zhang, H Tian, R Zhou, L Duan, L Chen, X Lun, L AF Zhang, Yunyuan Zhou, Jun Sun, Meiling Sun, Guirong Cao, Yongxian Zhang, Haiping Tian, Runhua Zhou, Lan Duan, Liang Chen, Xian Lun, Limin TI Prognostic Value of microRNA-9 in Various Cancers: a Meta-analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE miR-9; Cancer; Prognosis; Meta-analysis ID miR-9; Cancer; Prognosis; Meta-analysis AB Recently, there are more and more evidences from studies have revealed the association between microRNA-9 (miR-9) expression and outcome in multiple cancers, but inconsistent results have also been reported. It is necessary to rationalize a meta analysis of all available data to clarify the prognostic role of miR-9. Eligible studies were selected through multiple search strategies and the quality was assessed by MOOSE. Data was extracted from studies according to the key statistics index. All analyses were performed using STATA software. Twenty studies were selected in the meta-analysis to evaluate the prognostic role of miR-9 in multiple tumors. MiR-9 expression level was an independent prognostic biomarker for OS in tumor patients using multivariate and univariate analyses. High expression levels of miR-9 was demonstrated to associated with poor overall survival (OS) (HR = 2.23, 95 % CI: 1.56–3.17, P < 0.05) and recurrence free survival/progress free survival (RFS/PFS) (HR = 2.08, 95 % CI: 1.33–3.27, P < 0.05). Subgroup analysis showed that residence region (China and Japan), sample size, cancer type (solid or leukemia), follow-up months and analysis method (qPCR) did not alter the predictive value of miR-9 on OS in various cancers. Furthermore, no significant associations were detected for miR-9 expression and lymph node metastasis or distant metastasis. The present results suggest that promoted miR-9 expression is associated with poor OS in patients with general cancers. C1 [Zhang, Yunyuan] The Affiliated Hospital of Qingdao University, Department of Clinical Laboratory, 266003 Qingdao, China. [Zhou, Jun] The Affiliated Hospital of Qingdao University, Department of Clinical Laboratory, 266003 Qingdao, China. [Sun, Meiling] The Affiliated Hospital of Qingdao University, Department of Clinical Laboratory, 266003 Qingdao, China. [Sun, Guirong] The Affiliated Hospital of Qingdao University, Department of Clinical Laboratory, 266003 Qingdao, China. [Cao, Yongxian] The Affiliated Hospital of Qingdao University, Department of Clinical Laboratory, 266003 Qingdao, China. [Zhang, Haiping] The Affiliated Hospital of Qingdao University, Department of Clinical Laboratory, 266003 Qingdao, China. [Tian, Runhua] The Affiliated Hospital of Qingdao University, Department of Clinical Laboratory, 266003 Qingdao, China. [Zhou, Lan] Chongqing Medical University, College of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, 400016 Chongqing, China. [Duan, Liang] the Second Hospital Affiliated to Chongqing Medical University, Department of Laboratory Medicine, 400010 Chongqing, China. [Chen, Xian] The Affiliated Hospital of Qingdao University, Department of Clinical Laboratory, 266003 Qingdao, China. [Lun, Limin] The Affiliated Hospital of Qingdao University, Department of Clinical Laboratory, 266003 Qingdao, China. RP Chen, X (reprint author), The Affiliated Hospital of Qingdao University, Department of Clinical Laboratory, 266003 Qingdao, China. EM cxkakicoco2014@163.com CR Fendler A, Jung K, 2013, MicroRNAs as new diagnostic and prognostic biomarkers in urological tumors. Crit RevOncog 18(4):289–302 Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66(1):7–30., DOI 10.3322/caac.21332 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116(2):281–97 Lee RC, Feinbaum RL, Ambros V, 1993, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75(5):843–54 Povey S, Lovering R, Bruford E, Wright M, Lush M, Wain H, 2001, The HUGO Gene Nomenclature Committee, HGNC). Hum Genet 109(6):678–80., DOI 10.1007/s00439-001-0615-0 Stroup DF, Berlin JA,Morton SC, Olkin I,Williamson GD, Rennie D, Moher D, Becker BJ, Sipe TA, Thacker SB, 2000, Metaanalysis of observational studies in epidemiology: a proposal for reporting. Meta-analysis Of Observational Studies in Epidemiology, MOOSE, group. JAMA 283(15):2008–12 Parmar MK, Torri V, Stewart L, 1998, Extracting summary statistics to perform meta-analyses of the published literature for survival endpoints. Stat Med 17(24):2815–34., DOI 10.1002/(SICI)1097- 0258(19981230)17:24<2815::AID-SIM110>3.0.CO;2-8 Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR, 2007, Practical methods for incorporating summary time-to-event data into meta-analysis. Trials 8:16., DOI 10.1186/1745-6215-8-16 Hu Y, Correa AM, Hoque A, Guan B, Ye F, Huang J, Swisher SG, Wu TT, Ajani JA, Xu XC, 2011, Prognostic significance of differentially expressed miRNAs in esophageal cancer. Int J Cancer 128(1):132–43., DOI 10.1002/ijc.25330 Li X, Lu Y, Chen Y, Lu W, Xie X, 2013, MicroRNA profile of paclitaxel-resistant serous ovarian carcinoma based on formalinfixed paraffin-embedded samples. BMC Cancer 13:216., DOI 10.1186/1471-2407-13-216 Pignot G, Cizeron-Clairac G, Vacher S, Susini A, Tozlu S, Vieillefond A, Zerbib M, Lidereau R, Debre B, Amsellem-Ouazana D, Bieche I, 2013, microRNA expression profile in a large series of bladder tumors: identification of a 3-miRNA signature associated with aggressiveness of muscle-invasive bladder cancer. Int J Cancer 132(11):2479–91., DOI 10.1002/ijc.27949 Wu Z, Wang L, Li G, Liu H, Fan F, Li Z, Li Y, Gao G, 2013, Increased expression of microRNA-9 predicts an unfavorable prognosis in human glioma. Mol Cell Biochem 384(1–2):263–8., DOI 10.1007/s11010-013-1805-5 Xu SH, Yang YL, Han SM, Wu ZH, 2014, MicroRNA-9 expression is a prognostic biomarker in patients with osteosarcoma.World J Surg Oncol 12:195., DOI 10.1186/1477-7819-12-195 Xu T, Liu X, Han L, Shen H, Liu L, Shu Y, 2014, Up-regulation of miR-9 expression as a poor prognostic biomarker in patients with non-small cell lung cancer. Clin Transl Oncol 16(5):469–75., DOI 10.1007/s12094-013-1106-1 Wu S, Jia S, Xu P, 2014, MicroRNA-9 as a novel prognostic biomarker in human laryngeal squamous cell carcinoma. Int J Clin Exp Med 7(12):5523–8 SongY, Li J, ZhuY,Dai Y, ZengT, LiuL,WangH,QinY, ZengM, Guan XY, Li Y, 2014, MicroRNA-9 promotes tumormetastasis via repressing E-cadherin in esophageal squamous cell carcinoma. Oncotarget 5(22):11669–80., DOI 10.18632/oncotarget.2581 Fei D, Li Y, Zhao D, Zhao K, Dai L, Gao Z, 2014, Serum miR-9 as a prognostic biomarker in patients with osteosarcoma. J Int Med Res 42(4):932–7., DOI 10.1177/0300060514534643 Cai L, Cai X, 2014, Up-regulation of miR-9 expression predicate advanced clinicopathological features and poor prognosis in patients with hepatocellular carcinoma. Diagn Pathol 9:1000., DOI 10.1186/s13000-014-0228-2 FariaAM, Sbiera S, Ribeiro TC, Soares IC, MarianiBM, Freire DS, de Sousa GR, Lerario AM, Ronchi CL, Deutschbein T,Wakamatsu A, Alves VA, Zerbini MC, Mendonca BB, Fragoso MC, Latronico AC, Fassnacht M, Almeida MQ, 2015, Expression of LIN28 and its regulatory microRNAs in adult adrenocortical cancer. Clin Endocrinol, Oxf, 82(4):481–8., DOI 10.1111/cen.12607 Zhang J, Chong CC, Chen GG, Lai PB, 2015, A seven-microRNA expression signature predicts survival in hepatocellular carcinoma. PLoS One 10(6), e0128628., DOI 10.1371/journal.pone.0128628 Gwak JM, Kim HJ, Kim EJ, Chung YR, Yun S, Seo AN, Lee HJ, Park SY, 2014, MicroRNA-9 is associated with epithelialmesenchymal transition, breast cancer stem cell phenotype, and tumor progression in breast cancer. Breast Cancer Res Treat 147(1):39–49., DOI 10.1007/s10549-014-3069-5 Sun J, Fang K, Shen H, Qian Y, 2015, MicroRNA-9 is a ponderable index for the prognosis of human hepatocellular carcinoma. Int J Clin Exp Med 8(10):17748–56 Lu J, XuX, Liu X, Peng Y, Zhang B,Wang L, Luo H, Peng X, Li G, TianW, HeM, Li X, 2014, Predictive value of miR-9 as a potential biomarker for nasopharyngeal carcinoma metastasis. Br J Cancer 110(2):392–8., DOI 10.1038/bjc.2013.751 White RA, Neiman JM, Reddi A, Han G, Birlea S,Mitra D, Dionne L, Fernandez P, Murao K, Bian L, Keysar SB, Goldstein NB, Song N, Bornstein S, Han Z, Lu X,Wisell J, Li F, Song J, Lu SL, Jimeno A, Roop DR, Wang XJ, 2013, Epithelial stem cell mutations that promote squamous cell carcinoma metastasis. J Clin Invest 123(10):4390–404., DOI 10.1172/JCI65856 Sugita F, Maki K, Nakamura Y, Sasaki K, Mitani K, 2014, Overexpression of MIR9 indicates poor prognosis in acute lymphoblastic leukemia. Leuk Lymphoma 55(1):78–86., DOI 10.3109/10428194.2013.790023 Maki K, Yamagata T, Sugita F, Nakamura Y, Sasaki K, Mitani K, 2012, Aberrant expression of MIR9 indicates poor prognosis in acute myeloid leukaemia. Br J Haematol 158(2):283–5., DOI 10.1111/j.1365-2141.2012.09118.x Feng JB, Zhang GY,Mai GL,Yang SH, Chen BS, 2014)miR-9 gene expression in esophageal cancer tissues and onocancerous tissues surrounding esophageal cancer. J Hainan Med Univ, 04): 458–461+ 464., DOI 10.13210/j.cnki.jhmu.20131228.023(in Chinese) Long QQ, Tao ZJ, Han JB, Zhao L, Yi YX, 2014, Hsa-miR-9 expression and its clinical significance in colorectal cancer tissues. Chin J Gerontol 34(2):307–8., DOI 10.3969/j.issn.1005- 9202.2014.02.009, in Chinese) Lewis BP, Burge CB, Bartel DP, 2005, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120(1):15–20., DOI 10.1016/j. cell.2004.12.035 Kim DJ, Linnstaedt S, Palma J, Park JC, Ntrivalas E, Kwak-Kim JY, Gilman-Sachs A, Beaman K, Hastings ML, Martin JN, Duelli DM, 2012, Plasma components affect accuracy of circulating cancer-related microRNA quantitation. J Mol Diagn 14(1):71–80., DOI 10.1016/j.jmoldx.2011.09.002 Redova M, Sana J, Slaby O, 2013, Circulating miRNAs as new blood-based biomarkers for solid cancers. Future Oncol 9(3):387– 402., DOI 10.2217/fon.12.192 Ma L, Young J, Prabhala H, Pan E, Mestdagh P, Muth D, Teruya- Feldstein J, Reinhardt F, Onder TT, Valastyan S, Westermann F, Speleman F, Vandesompele J, Weinberg RA, 2010, miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nat Cell Biol 12(3):247–56., DOI 10.1038/ncb2024 Senyuk V, Zhang Y, Liu Y, Ming M, Premanand K, Zhou L, Chen P, Chen J, Rowley JD, Nucifora G, Qian Z, 2013, Critical role of miR-9 in myelopoiesis and EVI1-induced leukemogenesis. Proc Natl Acad Sci U S A 110(14):5594–9., DOI 10.1073/pnas.1302645110 Wan HY, Guo LM, Liu T, Liu M, Li X, Tang H, 2010, Regulation of the transcription factor NF-kappaB1 by microRNA-9 in human gastric adenocarcinoma. Mol Cancer 9:16., DOI 10.1186/1476-4598- 9-16 Yu T, LiuK,WuY, Fan J, Chen J, Li C, Yang Q,Wang Z, 2014, MicroRNA-9 inhibits the proliferation of oral squamous cell carcinoma cells by suppressing expression of CXCR4 via the Wnt/beta-catenin signaling pathway. Oncogene 33(42):5017– 27., DOI 10.1038/onc.2013.448 Wang H, Zhang W, Zuo Y, Ding M, Ke C, Yan R, Zhan H, Liu J, Wang J, 2015, miR-9 promotes cell proliferation and inhibits apoptosis by targeting LASS2 in bladder cancer. Tumour Biol 36(12): 9631–40., DOI 10.1007/s13277-015-3713-7 Tian C, You MJ, Yu Y, Zhu L, Zheng G, Zhang Y, 2016, MicroRNA-9 promotes proliferation of leukemia cells in adult CD34-positive acute myeloid leukemia with normal karyotype by downregulation of Hes1. Tumour Biol 37(6):7461–71., DOI 10.1007/s13277-015-4581-x Zhu SW, Li JP, Ma XL, Ma JX, Yang Y, Chen Y, Liu W, 2015, miR-9 modulates osteosarcoma cell growth by targeting the GCIP tumor suppressor. Asian Pac J Cancer Prev 16(11):4509–13 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 573 EP 582 DI 10.1007/s12253-016-0148-4 PG 10 ER PT J AU Molnar, E Schultz, M Schmidt-Schultz, HTy Marcsik, A Buczko, K Zadori, P Biro, G Bernert, Zs Baumhoer, D Hajdu, T AF Molnar, Erika Schultz, Michael Schmidt-Schultz, H Tyede Marcsik, Antonia Buczko, Krisztina Zadori, Peter Biro, Gergely Bernert, Zsolt Baumhoer, Daniel Hajdu, Tamas TI Rare Case of an Ancient Craniofacial Osteosarcoma with Probable Surgical Intervention SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Craniofacial osteosarcoma; Paleopathology; Incomplete trephination ID Craniofacial osteosarcoma; Paleopathology; Incomplete trephination AB Osteosarcoma is the most common primary malignant bone tumor both today and in antiquity. Nevertheless, it is a comparatively rare tumor. This paper describes a case of a highly aggressive craniofacial lesion from the 11th–12th centuries AD, most likely representing osteosarcoma. During the paleopathological study, macroscopic, endoscopic, radiological, scanning-electron and light microscopic investigations were performed. The skull of the approximately 40–50 yearold female revealed several pathological findings. The most impressive macroscopic feature was an extensively spiculated periosteal reaction ("sunburst" pattern) in combination with a massive bone destruction most likely derived from a highly aggressive tumor originating in the ethmoidal area of the medial wall of the orbit. The central parts of the lesion showed excessive new and most probably neoplastic bone formation indicating an underlying high-grade osteosarcoma. The light microscopic examination revealed three different levels of bony structures representing different qualities of bone tissues. Besides the mass lesion, signs of a healed multiple incomplete trephination of the left parietal bone was observed. This case represents a unique example in which the concomitance of a tumor and an incomplete trephination could be observed from the skeletal remains of an ancient individual. The case opens new considerations as to whether surgical interventions, such as incomplete trephination, might have been used already in the Middle Ages as a therapeutic approach. C1 [Molnar, Erika] University of Szeged, Department of Anatomy, Kozep fasor 52, H-6726 Szeged, Hungary. [Schultz, Michael] University Medical School Gottingen, Department of Anatomy, Kreuzbergring 36, D-37075 Gottingen, Germany. [Schmidt-Schultz, H Tyede] University Medical School Gottingen, Department of Anatomy, Kreuzbergring 36, D-37075 Gottingen, Germany. [Marcsik, Antonia] University of Szeged, Department of Anatomy, Kozep fasor 52, H-6726 Szeged, Hungary. [Buczko, Krisztina] Hungarian Natural History Museum, Department of Botany, Konyves Kalman korut 40, H-1097 Budapest, Hungary. [Zadori, Peter] Kaposi Mor Teaching Hospital, Guba Sandor utca 40, H-7400 Kaposvar, Hungary. [Biro, Gergely] Kaposi Mor Teaching Hospital, Guba Sandor utca 40, H-7400 Kaposvar, Hungary. [Bernert, Zsolt] Hungarian Natural History Museum, Department of Anthropology, Ludovika ter 2, H-1083 Budapest, Hungary. [Baumhoer, Daniel] University Hospital Basel, Institute of Pathology, Bone Tumor Reference Center, Schonbeinstrasse 40, D-4031 Basel, Switzerland. [Hajdu, Tamas] Hungarian Natural History Museum, Department of Anthropology, Ludovika ter 2, H-1083 Budapest, Hungary. RP Molnar, E (reprint author), University of Szeged, Department of Anatomy, H-6726 Szeged, Hungary. EM balinte@bio.u-szeged.hu CR David AR, Zimmerman MR, 2010, Cancer: an old disease, a new disease or something in between? Nat Rev Cancer 10:728–733 Capasso LL, 2005, Antiquity of cancer. Int J Cancer 113:2–13 Thillaud PL, 2006, Paleopathology of cancers. Bull Cancer 93: 767–773 SchultzM, Parzinger H, Posdnjakov DV, Chikisheva TA, Schmidt- Schultz TH, 2007, Oldest known case of metastasizing prostate carcinoma diagnosed in the skeleton of a 2,700-year-old Scythian king from Arzhan, Siberia, Russia). Int J Cancer 121:2591–2595 Ortner DJ, 2011)What skeletons tell us. The story of human paleopathology. Virchows Arch 459(3):247–254., DOI 10.1007/s00428- 011-1122-x Halperin EC, 2004, Paleo-oncology: the role of ancient remains in the study of cancer. Perspect Biol Med 47:1–14 Rosenberg AE, Cleton-Jansen A-M, de Pinieux G, Deyrup AT, Hauben E, Squire J, 2013, Conventional osteosarcoma. In: Fletcher CDM, Bridge JA, Hogendoorn PCW, Mertens F, eds, WHO classification of Tumours of Tumours of soft tissue and bone. IARC Press, Lyon, pp. 282–288 Gadwal SR, Gannon FH, Fanburg-Smith JC, Becoskie EM, Thompson LD, 2001, Primary osteosarcoma of the head and neck in pediatric patients: a clinicopathologic study of 22 cases with a review of the literature. Cancer 91:598–605 Smith RB, Apostolakis LW, Karnell LH, Koch BB, Robinson RA, Zhen W, Menck HR, Hoffman HT, 2003, National Cancer Data Base report on osteosarcoma of the head and neck. Cancer 98: 1670–1680 Beutel A, Tanzer A, 1963, Rontgendiagnostik der Orbitae, der Augen und der Tranenwege. In: Olsson O, Strnad F, Vieten H, Zuppinger A, eds, Handbuch der Medizinischen Radiologie, Bd VII/2. Rontgendiagnostik des Schadels II, 1st edn. Springer, Berlin-Gottingen-Heidelberg, pp 673–818 Ortner DJ, 2003, Tumors and tumor-like lesions of bone. In: Ortner DJ, ed, Identification of pathological conditions in human skeletal remains, 2nd edn. Academic Press, New York, pp. 503–544 Farkas LG, Jozsa L, Paja L, Molnar J, 2007, Bone forming tumors on skeletons from a medieval Hungarian cemetery, Batmonostor). Paleopathol Newsl 140:14–22 Jozsa LG, Fothi E, 2003, Juxtacortical osteosarcoma on tibia and fibula from a medieval cemetery of Budapest. J Paleopath 15:23–31 Bona A, Papai Z, Maasz G, Gabor A, Toth GA, Jambor E, Schmidt J, Cs T, Cs F, Mark L, 2014, Mass spectrometric identification of ancient proteins as potential molecular biomarkers for a 2000-yearold osteogenic sarcoma. PLoS One 9(7):e103862., DOI 10.1371 /journal.pone.0103862 Russeva V, 2012, Religion, Magic or Medicine? New finds of trepanned skulls from Southeastern Bulgaria, 11th–13th c. Archaeologia Bulgarica XVI:77–95 Mariani-Costantini R, Catalano P, di Gennaro F, di Tota G, Angeletti LR, 2000, New light on cranial surgery in ancient Rome. Lancet 355:5–7 Jordanov J, Br D, Sp N, 1988, Symbolic trephination of skulls from the middle ages, IXth-Xth century, in Bulgaria. Acta Neurochir 92: 15–18 Zs B, Evinger S, Fothi E, 2006, New symbolic trephination cases from Hungary. Annls hist-nat Mus nat hung 98:177–183 Bereczki Z, Molnar E, Marcsik A, Palfi G, 2015, Rare types of trephination from Hungary shed new light on possible crosscultural connections in the Carpathian Basin. Int J Osteoarchaeol 25(3):322–333 Allodiatoris I, 1937, Adatok az Arpadkori alfoldi magyarsag anthropologiajahoz, data to the anthropology of the lowland Hungarians from the Arpadian-age). University of Pazmany Peter, Dissertation Zadori P, Bajzik G, Biro G, Lelovics Zs, Balassa T, Bernert Zs, Evinger S, Hajdu T, Marcsik A, Molnar E, Osz B, Palfi Gy, Wolff K, Repa I, 2016, Koponyacsont-laesiok komputertomografias vizsgalata es paleoradiologiai aspektusai, Computed tomographic examination of cranial lesions, a paleoradiological approach, Ideggyogy Sz 69:, in press) Schultz M, 2001, Paleohistopathology of bone: a new approach to the study of ancient diseases. Yearb Phys Anthropol 44:106–147 Schultz M, 2003, Light microscopic analysis in skeletal paleopathology. In: Ortner DJ, ed, Identification of pathological conditions in human skeletal remains, 2nd edn. Academic Press, New York, pp. 73–108 Schultz M, 2011, Light microscopic analysis of macerated pathologically changed bone. In: Crowder C, Stout S, eds, Bone histology. An anthropological perspective. CRC Press, Taylor & Francis Group, Boca Raton, pp. 253–296 Molnar M, Janovics R, Major I, Molnar M, Janovics R, Major I, Orsovszki J, Gonczi R, Veres M, Leonard AG, Castle SM, Lange TE, Wacker L, Hajdas I, Jull AJT, 2013, Status report of the new AMS 14C sample preparation lab of the Hertelendi Laboratory of environmental studies, Debrecen, Hungary. In: Jull AJT, Hatte C, eds). Proceedings of the 21st International Radiocarbon Conference. Radiocarbon 55:665–76 Weidenreich F, 1930, Das Knochengewebe. In: v. Mollendorff W, ed, Handbuch der Mikroskopischen Anatomie des Menschen, Bd. II, Die Gewebe, Zweiter Teil, Stutzgewebe, Knochengewebe, Skeletsystem. Verlag von Julius Springer, Berlin, pp. 391–520 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 583 EP 587 DI 10.1007/s12253-016-0153-7 PG 5 ER PT J AU Patai, V Bartak, KB Peterfia, B Micsik, T Horvath, R Sumanszki, Cs Peter, Z Patai, Valcz, G Kalmar, A Toth, K Krenacs, T Tulassay, Zs Molnar, B AF Patai, V Arpad Bartak, Kinga Barbara Peterfia, Balint Micsik, Tamas Horvath, Reka Sumanszki, Csaba Peter, Zoltan Patai, Arpad Valcz, Gabor Kalmar, Alexandra Toth, Kinga Krenacs, Tibor Tulassay, Zsolt Molnar, Bela TI Comprehensive DNA Methylation and Mutation Analyses Reveal a Methylation Signature in Colorectal Sessile Serrated Adenomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Biomarker; DNA methylation; Mutation; Serrated polyp; Sessile serrated adenoma; Traditional serrated adenoma ID Biomarker; DNA methylation; Mutation; Serrated polyp; Sessile serrated adenoma; Traditional serrated adenoma AB Colorectal sessile serrated adenomas (SSA) are hypothesized to be precursor lesions of an alternative, serrated pathway of colorectal cancer, abundant in genes with aberrant promoter DNA hypermethylation. In our present pilot study, we explored DNA methylation profiles and examined selected gene mutations in SSA. Biopsy samples from patients undergoing screening colonoscopy were obtained during endoscopic examination. After DNA isolation and quality analysis, SSAs (n = 4) and healthy controls (n = 5) were chosen for further analysis. DNA methylation status of 96 candidate genes was screened by q(RT)PCR using Methyl-Profiler PCR array system. Amplicons for 12 gene mutations were sequenced by GS Junior Instrument using ligated and barcoded adaptors. Analysis of DNA methylation revealed 9 hypermethylated genes in both normal and SSA samples. 12 genes (CALCA, DKK2, GALR2, OPCML, PCDH10, SFRP1, SFRP2, SLIT3, SST, TAC1, VIM, WIF1) were hypermethylated in all SSAs and 2 additional genes (BNC1 and PDLIM4) were hypermethylated in 3 out of 4 SSAs, but in none of the normal samples. 2 SSAs exhibited BRAF mutation and synchronous MLH1 hypermethylation and were microsatellite instable by immunohistochemical analysis. Our combined mutation and DNA methylation analysis revealed that there is a common DNA methylation signature present in pre-neoplastic SSAs. This study advocates for the use of DNA methylation as a potential biomarker for the detection of SSA; however, further investigation is needed to better characterize the molecular background of these newly recognized colorectal lesions. C1 [Patai, V Arpad] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Bartak, Kinga Barbara] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Peterfia, Balint] Hungarian Academy of Sciences, Molecular Medicine Research Group, Roosevelt ter 9, 1051 Budapest, Hungary. [Micsik, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Horvath, Reka] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Sumanszki, Csaba] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Peter, Zoltan] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Patai, Arpad] Markusovszky Hospital, Department of Medicine and Gastroenterology, Markusovszky Lajos utca 5, 9700 Szombathely, Hungary. [Valcz, Gabor] Hungarian Academy of Sciences, Molecular Medicine Research Group, Roosevelt ter 9, 1051 Budapest, Hungary. [Kalmar, Alexandra] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Toth, Kinga] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. RP Patai, V (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM arpad.patai@gmail.com CR East JE, Vieth M, Rex DK, 2015, Serrated lesions in colorectal cancer screening: detection, resection, pathology and surveillance. Gut 64:991–1000 Patai AV, Molnar B, Tulassay Z, Sipos F, 2013, The serrated pathway: the alternative route to colorectal cancer. World J Gastroenterol 19:607–615 Fearon ER, Vogelstein B, 1990, A genetic model for colorectal tumorigenesis. Cell 61:759–767 Patai AV, Molnar B, Kalmar A, Scholler A, Toth K, Tulassay Z, 2012, Role of DNA methylation in colorectal carcinogenesis. Dig Dis 30:310–315 Toth K, Sipos F, Kalmar A, Patai AV, Wichmann B, Stoehr R, Golcher H, Schellerer V, Tulassay Z, Molnar B, 2012, Detection of methylated SEPT9 in plasma is a reliable screening method for both left- and right-sided colon cancers. PLoS One 7:e46000 Toth K, Wasserkort R, Sipos F, Kalmar A, Wichmann B, Leiszter K, Valcz G, Juhasz M, Miheller P, Patai AV, Tulassay Z, Molnar B, 2014, Detection of methylated septin 9 in tissue and plasma of colorectal patients with neoplasia and the relationship to the amount of circulating cell-free DNA. PLoS One 9:e115415 Burgess NG, Tutticci NJ, Pellise M, Bourke MJ, 2014, Sessile serrated adenomas/polyps with cytologic dysplasia: a triple threat for interval cancer. Gastrointest Endosc 80:307–310 Erichsen R, Baron JA, Hamilton-Dutoit SJ, Snover DC, Torlakovic EE, Pedersen L, Froslev T, Vyberg M, Hamilton SR, Sorensen HT, 2016, Increased risk of colorectal cancer development among patients with serrated polyps. Gastroenterology 150:895–902.e5 Regula J, Rupinski M, Kraszewska E, Polkowski M, Pachlewski J, Orlowska J, Nowacki MP, Butruk E, 2006, Colonoscopy in colorectal-cancer screening for detection of advanced neoplasia. N Engl J Med 355:1863–1872 Rothwell PM,Wilson M, Elwin CE, Norrving B, Algra A,Warlow CP, Meade TW, 2010, Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials. Lancet 376:1741–1750 Heigh RI, Yab TC, Taylor WR, Hussain FT, Smyrk TC, Mahoney DW, Domanico MJ, Berger BM, Lidgard GP, Ahlquist DA, 2014, Detection of colorectal serrated polyps by stool DNA testing: comparison with fecal immunochemical testing for occult blood, FIT). PLoS One 9:e85659 Park SJ, Rashid A, Lee JH, Kim SG, Hamilton SR, Wu TT, 2003, Frequent CpG island methylation in serrated adenomas of the colorectum. Am J Pathol 162:815–822 Wynter CV, Walsh MD, Higuchi T, Leggett BA, Young J, Jass JR, 2004, Methylation patterns define two types of hyperplastic polyp associated with colorectal cancer. Gut 53:573–580 O'Brien MJ, Yang S, Mack C, Xu H, Huang CS, Mulcahy E, Amorosino M, Farraye FA, 2006, Comparison of microsatellite instability, CpG island methylation phenotype, BRAF and KRAS status in serrated polyps and traditional adenomas indicates separate pathways to distinct colorectal carcinoma end points. Am J Surg Pathol 30:1491–1501 Kim YH, Kakar S, Cun L, Deng G, Kim YS, 2008, Distinct CpG island methylation profiles and BRAF mutation status in serrated and adenomatous colorectal polyps. Int J Cancer 123:2587–2593 Subramaniam MM, Chan JY, Soong R, Ito K, Yeoh KG, Wong R, Guenther T, Will O, Chen CL, Kumarasinghe MP, Ito Y, Salto- Tellez M, 2009, RUNX3 inactivation in colorectal polyps arising through different pathways of colonic carcinogenesis. Am J Gastroenterol 104:426–436 Kim KM, Lee EJ, Ha S, Kang SY, Jang KT, Park CK, Kim JY, Kim YH, Chang DK, Odze RD, 2011, Molecular features of colorectal hyperplastic polyps and sessile serrated adenoma/polyps from Korea. Am J Surg Pathol 35:1274–1286 Kaji E, Uraoka T, Kato J, Hiraoka S, Suzuki H, Akita M, Saito S, Tanaka T, Ohara N, Yamamoto K, 2012, Externalization of sawtooth architecture in small serrated polyps implies the presence of methylation of IGFBP7. Dig Dis Sci 57:1261–1270 Fu X, Li L, Peng Y, 2012, Wnt signalling pathway in the serrated neoplastic pathway of the colorectum: possible roles and epigenetic regulatory mechanisms. J Clin Pathol 65:675–679 Beggs AD, Jones A, Shepherd N, Arnaout A, Finlayson C, Abulafi AM, Morton DG, Matthews GM, Hodgson SV, Tomlinson IP, 2013, Loss of expression and promoter methylation of SLIT2 are associated with sessile serrated adenoma formation. PLoS Genet 9: e1003488 Muto Y, Maeda T, Suzuki K, Kato T, Watanabe F, Kamiyama H, Saito M, Koizumi K, Miyaki Y, Konishi F, Alonso S, Perucho M, Rikiyama T, 2014, DNA methylation alterations of AXIN2 in serrated adenomas and colon carcinomas with microsatellite instability. BMC Cancer 14:466 Dhir M, Yachida S, Van Neste L, Glockner SC, Jeschke J, Pappou EP, Montgomery EA, Herman JG, Baylin SB, Iacobuzio-Donahue C, Ahuja N, 2011, Sessile serrated adenomas and classical adenomas: an epigenetic perspective on premalignant neoplastic lesions of the gastrointestinal tract. Int J Cancer 129:1889–1898 Gaiser T, Meinhardt S, Hirsch D, Killian JK, Gaedcke J, Jo P, Ponsa I, Miro R, Ruschoff J, Seitz G, Hu Y, Camps J, Ried T, 2013, Molecular patterns in the evolution of serrated lesion of the colorectum. Int J Cancer 132:1800–1810 Patai AV, Valcz G, Hollosi P, Kalmar A, Peterfia B, Patai A, Wichmann B, Spisak S, Bartak BK, Leiszter K, Toth K, Sipos F, Kovalszky I, Peter Z, Miheller P, Tulassay Z, Molnar B, 2015, Comprehensive DNA methylation analysis reveals a common ten-gene methylation signature in colorectal adenomas and carcinomas. PLoS One 10:e0133836 Holemon H, Korshunova Y, Ordway JM, Bedell JA, Citek RW, Lakey N, Leon J, Finney M, McPherson JD, Jeddeloh JA, 2007, MethylScreen: DNA methylation density monitoring using quantitative PCR. BioTechniques 43:683–693 Peterfia B, Kalmar A, Patai AV, Bodor A, Micsik T, Hollosi P, Egedi K, Wichmann B, Tulassay Z, Molnar B, 2016, Construction of a multiplex mutation hot spot PCR panel: the first step towards colorectal cancer genotyping on the GS Junior platform. J Cancer., DOI 10.7150/jca.16037 Valcz G, Patai AV, Kalmar A, Peterfia B, Furi I, Wichmann B, Muzes G, Sipos F, Krenacs T, Mihaly E, Spisak S, Molnar B, Tulassay Z, 2014, Myofibroblast-derived SFRP1 as potential inhibitor of colorectal carcinoma field effect. PLoS One 9:e106143 Lee EJ, Chun SM, Kim MJ, Jang SJ, Kim do S, Lee DH, Youk EG, 2016, Reappraisal of hMLH1 promoter methylation and protein expression status in the serrated neoplasia pathway. Histopathology 69:198–210 Hazewinkel Y, de Wijkerslooth TR, Stoop EM, Bossuyt PM, Biermann K, van de Vijver MJ, Fockens P, van Leerdam ME, Kuipers EJ, Dekker E, 2014, Prevalence of serrated polyps and association with synchronous advanced neoplasia in screening colonoscopy. Endoscopy 46:219–224 Sipos F,Muzes G, Patai AV, Furi I, Peterfia B, Hollosi P, Molnar B, Tulassay Z, 2013, Genome-wide screening for understanding the role of DNA methylation in colorectal cancer. Epigenomics 5:569– 581 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 589 EP 594 DI 10.1007/s12253-016-0154-6 PG 6 ER PT J AU Arzt, L Halbwedl, I Gogg-Kamerer, M Popper, HH AF Arzt, Lisa Halbwedl, Iris Gogg-Kamerer, Margit Popper, H Helmut TI Signal Transducer and Activator of Transcription 1 (STAT1) Knock-down Induces Apoptosis in Malignant Pleural Mesothelioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Malignant pleural mesothelioma; STAT signaling; miRNA; Apoptosis ID Malignant pleural mesothelioma; STAT signaling; miRNA; Apoptosis AB Malignant pleural mesothelioma (MPM) is the most common primary tumor of the pleura. Its incidence is still increasing in Europe and the prognosis remains poor. We investigated the oncogenic function of signal transducer and activator of transcription 1 (STAT1) in MPM in more detail. A miRNA profiling was performed on 52 MPM tissue samples. Upregulated miRNAs (targeting SOCS1/3) were knockeddown using miRNA inhibitors. mRNA expression levels of STAT1/3, SOCS1/3 were detected in MPM cell lines. STAT1 has been knocked-down using siRNA and qPCR was used to detect mRNA expression levels of all JAK/STAT family members and genes that regulate them. An immunohistochemical staining was performed to detect the expression of caspases. STAT1 was upregulated and STAT3 was downregulated, SOCS1/3 protein was not detected but it was possible to detect SOCS1/3mRNA in MPM cell lines. The upregulated miRNAs were successfully knocked-down, however the expected effect on SOCS1 expression was not detected. STAT1 knock-down had different effects on STAT3/5 expression. Caspase 3a and 8 expression was found to be increased after STAT1 knock-down. The physiologic regulation of STAT1 via SOCS1 is completely lost in MPM and it does not seem that the miRNAs identified by now, do inhibit the expression of SOCS1. MPM cell lines compensate STAT1 knock-down by increasing the expression of STAT3 or STAT5a, two genes which are generally considered to be oncogenes. And much more important, STAT1 knock-down induces apoptosis in MPM cell lines and STAT1 might therefore be a target for therapeutic intervention. C1 [Arzt, Lisa] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. [Halbwedl, Iris] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. [Gogg-Kamerer, Margit] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. [Popper, H Helmut] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. RP Arzt, L (reprint author), Medical University of Graz, Department of Pathology, A-8036 Graz, Austria. EM lisa.arzt@medunigraz.at CR Vogelzang NJ, Porta C, Mutti L, 2005, New agents in the management of advanced mesothelioma. Semin Oncol 32(3):336–350 Rawlings JS, Rosler KM, Harrison DA, 2004, The JAK/STAT signaling pathway. J Cell Sci 117(Pt 8):1281–1283., DOI 10.1242 /jcs.00963 Buard A, Vivo C, Monnet I, Boutin C, Pilatte Y, Jaurand MC, 1998, Human malignant mesothelioma cell growth: activation of janus kinase 2 and signal transducer and activator of transcription 1alpha for inhibition by interferon-gamma. Cancer Res 58(4):840–847 Kothmaier H, Quehenberger F, Halbwedl I, Morbini P, Demirag F, Zeren H, Comin CE, Murer B, Cagle PT, Attanoos R, Gibbs AR, Galateau-Salle F, Popper HH, 2008, EGFR and PDGFR differentially promote growth in malignant epithelioid mesothelioma of short and long term survivors. Thorax 63(4):345–351., DOI 10.1136/thx.2007.085241 Arzt L, Kothmaier H, Quehenberger F, Halbwedl I, Popper HH, 2011, STAT signalling in malignant mesothelioma: is there a regulatory effect of microRNAs? Magazine of European. Med Oncol 4(1):38–42 CaloV,MigliavaccaM, BazanV,MacalusoM, Buscemi M,Gebbia N, Russo A, 2003, STAT proteins: from normal control of cellular events to tumorigenesis. J Cell Physiol 197(2):157–168., DOI 10.1002/jcp.10364 Reich NC, Liu L, 2006, Tracking STAT nuclear traffic. Nat Rev Immunol 6(8):602–612., DOI 10.1038/nri1885 Sadzak I, Schiff M, Gattermeier I, Glinitzer R, Sauer I, Saalmuller A, Yang E, Schaljo B, Kovarik P, 2008, Recruitment of Stat1 to chromatin is required for interferon-induced serine phosphorylation of Stat1 transactivation domain. Proc Natl Acad Sci U S A 105(26): 8944–8949., DOI 10.1073/pnas.0801794105 Stephanou A, Latchman DS, 2005, Opposing actions of STAT-1 and STAT-3. Growth Factors 23(3):177–182., DOI 10.1080 /08977190500178745 Espert L, Dusanter-Fourt I, Chelbi-Alix MK, 2005, Negative regulation of the JAK/STAT: pathway implication in tumorigenesis. Bull Cancer 92(10):845–857 Wormald S, Hilton DJ, 2004, Inhibitors of cytokine signal transduction. J Biol Chem 279(2):821–824., DOI 10.1074/jbc. R300030200 Arzt L, Kothmaier H, Halbwedl I, Quehenberger F, Popper HH, 2014, Signal transducer and activator of transcription 1, STAT1, acts like an oncogene inmalignant pleuralmesothelioma. Virchows Arch 465(1):79–88., DOI 10.1007/s00428-014-1584-8 Meola N, Gennarino VA, Banfi S, 2009, microRNAs and genetic diseases. PathoGenetics 2(1):7., DOI 10.1186/1755-8417-2-7 Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, van der Meer AJ, Patick AK, Chen A, Zhou Y, Persson R, King BD, Kauppinen S, Levin AA, Hodges MR, 2013, Treatment of HCV infection by targeting microRNA. N Engl J Med 368(18):1685–1694., DOI 10.1056/NEJMoa1209026 Clinicaltrials.gov, 2013, A Multicenter Phase I Study of MRX34. http://clinicaltrials.gov/show/NCT01829971 Ueno T, Toyooka S, Fukazawa T, Kubo T, Soh J, Asano H, Muraoka T, Tanaka N, Maki Y, Shien K, Furukawa M, Sakaguchi M, Yamamoto H, Tsukuda K, Miyoshi S, 2014, Preclinical evaluation of microRNA-34b/c delivery for malignant pleural mesothelioma. Acta Med Okayama 68(1):23–26 Arzt L, Kothmaier H, Quehenberger F, Halbwedl I, Wagner K, Maierhofer T, Popper HH, 2011, Evaluation of formalin-free tissue fixation for RNA and microRNA studies. Exp Mol Pathol 91(2): 490–495., DOI 10.1016/j.yexmp.2011.05.007 Pass HI, Stevens EJ, Oie H, Tsokos MG, Abati AD, Fetsch PA, Mew DJ, Pogrebniak HW, MatthewsWJ, 1995, Characteristics of nine newly derived mesothelioma cell lines. Ann Thorac Surg 59(4):835–844 Reale FR, Griffin TW, Compton JM, Graham S, Townes PL, Bogden A, 1987, Characterization of a human malignant mesothelioma cell line, H-MESO-1): a biphasic solid and ascitic tumor model. Cancer Res 47(12):3199–3205 Shukla A, Hillegass JM, MacPherson MB, Beuschel SL, Vacek PM, Pass HI, Carbone M, Testa JR, Mossman BT, 2010, Blocking of ERK1 and ERK2 sensitizes human mesothelioma cells to doxorubicin. Mol Cancer 9:314., DOI 10.1186/1476-4598-9-314 Lesinski GB, Zimmerer JM, Kreiner M, Trefry J, Bill MA, Young GS, Becknell B, CarsonWE 3rd, 2010, Modulation of SOCS protein expression influences the interferon responsiveness of human melanoma cells. BMC Cancer 10:142., DOI 10.1186/1471-2407-10-142 Madonna S, Scarponi C, De Pita O, Albanesi C, 2008, Suppressor of cytokine signaling 1 inhibits IFN-gamma inflammatory signaling in human keratinocytes by sustaining ERK1/2 activation. FASEB J 22(9):3287–3297., DOI 10.1096/fj.08-106831 Klampfer L, 2006, Signal transducers and activators of transcription, STATs): novel targets of chemopreventive and chemotherapeutic drugs. Curr Cancer Drug Targets 6(2):107–121 Iwahori K, Serada S, Fujimoto M, Ripley B, Nomura S, Mizuguchi H, Shimada K, Takahashi T, Kawase I, Kishimoto T, Naka T, 2013, SOCS-1 gene delivery cooperates with cisplatin plus pemetrexed to exhibit preclinical antitumor activity against malignant pleural mesothelioma. Int J Cancer 132(2):459–471., DOI 10.1002/ijc.27611 Zhang B, Pan X, Cobb GP, Anderson TA, 2007, microRNAs as oncogenes and tumor suppressors. Dev Biol 302(1):1–12., DOI 10.1016/j.ydbio.2006.08.028 Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, Prueitt RL, Yanaihara N, Lanza G, Scarpa A, Vecchione A, Negrini M, Harris CC, CroceCM, 2006, A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 103(7): 2257–2261., DOI 10.1073/pnas.0510565103 Busacca S, Germano S, De Cecco L, Rinaldi M, Comoglio F, Favero F, Murer B, Mutti L, Pierotti M, Gaudino G, 2010, MicroRNA signature of malignant mesothelioma with potential diagnostic and prognostic implications. Am J Respir Cell Mol Biol 42(3):312–319., DOI 10.1165/rcmb.2009-0060OC Reid G, Pel ME, Kirschner MB, Cheng YY, Mugridge N, Weiss J, Williams M, Wright C, Edelman JJ, Vallely MP, McCaughan BC, Klebe S, Brahmbhatt H, MacDiarmid JA, van Zandwijk N, 2013, Restoring expression of miR-16: a novel approach to therapy for malignant pleural mesothelioma. Ann Oncol: Off J Eur Soc Med Oncol/ESMO 24(12):3128–3135., DOI 10.1093 /annonc/mdt412 Truini A, Coco S, Alama A, Genova C, Sini C, Dal Bello MG, Barletta G, Rijavec E, Burrafato G, Boccardo F, Grossi F, 2014, Role of microRNAs in malignant mesothelioma. Cell Mol life Sci: CMLS., DOI 10.1007/s00018-014-1584-5 Qing Y, Stark GR, 2004, Alternative activation of STAT1 and STAT3 in response to interferon-gamma. J Biol Chem 279(40): 41679–41685., DOI 10.1074/jbc.M406413200 NCBI F2R coagulation factor II, thrombin, receptor http://www. ncbi.nlmnih.gov/gene/2149 NCBI JUN proto-oncogene. http://www.ncbi.nlmnih. gov/gene/3725 NCBI CDKN1A cyclin-dependent kinase inhibitor 1A, p21, Cip1). http://www.ncbi.nlmnih.gov/gene/1026 Romanov VS, Pospelov VA, Pospelova TV, 2012, Cyclindependent kinase inhibitor p21(Waf1): contemporary view on its role in senescence and oncogenesis. Biochemistry. Biokhimiia 77(6):575–584., DOI 10.1134/S000629791206003X el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM, Lin D,Mercer WE, Kinzler KW, Vogelstein B, 1993)WAF1, a potential mediator of p53 tumor suppression. Cell 75(4):817–825 Lohr K, Moritz C, Contente A, Dobbelstein M, 2003, p21/ CDKN1A mediates negative regulation of transcription by p53. J Biol Chem 278(35):32507–32516., DOI 10.1074/jbc.M212517200 WangY, Blandino G, GivolD(1999, Induced p21waf expression in H1299 cell line promotes cell senescence and protects against cytotoxic effect of radiation and doxorubicin. Oncogene 18(16): 2643–2649., DOI 10.1038/sj.onc.1202632 Adams JM, Cory S, 2002, Apoptosomes: engines for caspase activation. Curr Opin Cell Biol 14(6):715–720 Earnshaw WC, Martins LM, Kaufmann SH, 1999, Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annu Rev Biochem 68:383–424., DOI 10.1146/annurev. biochem.68.1.383 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 595 EP 605 DI 10.1007/s12253-016-0157-3 PG 11 ER PT J AU Szczyrek, M Mlak, R Krawczyk, P Wojas-Krawczyk, K Powrozek, T Szudy-Szczyrek, A Zwolak, A Daniluk, J Milanowski, J AF Szczyrek, Michal Mlak, Radoslaw Krawczyk, Pawel Wojas-Krawczyk, Kamila Powrozek, Tomasz Szudy-Szczyrek, Aneta Zwolak, Agnieszka Daniluk, Jadwiga Milanowski, Janusz TI Polymorphisms of Genes Encoding Multidrug Resistance Proteins as a Predictive Factor for Second-Line Docetaxel Therapy in Advanced Non-small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NSCLC; Docetaxel; TUBB3; ABCC2; MDR1 ID NSCLC; Docetaxel; TUBB3; ABCC2; MDR1 AB Multidrug resistance (MDR) remains a substantial problem in chemotherapy. The purpose of the study was to investigate potential factors, including MDR genes polymorphisms, that could be used in qualification for second-line docetaxel therapy in non-small cell lung cancer (NSCLC) patients after failure of platinum based chemotherapy. Study group comprised of 58 Caucasian subjects. Evaluation of Single Nucleotide Polymorphisms (SNPs) of ABCC2/MRP2 and ABCB1/MDR1 genes was performed using the High Resolution Melting (HRM) technique. TUBB3 gene expression was evaluated on RNA isolated from tumor tissue. Results with p value of <0.05 were considered significant. Factors associated with reduced risk of disease progression included good performance status (PS), long period between diagnosis and docetaxel treatment, and smoking for <10 packyears. Disease control occurred more often in patients with G/G genotype of the ABCC2/MRP2 gene. Median overall survival was 4.25 months. Factors such as: good PS, disease control after docetaxel, long period from diagnosis to docetaxel, lack of significant weight loss, and third-line treatment were associated with prolongation of patients survival. Overall survival probability was significantly lower in patients with significant weight loss, poor PS, lack of disease control after docetaxel, and without third-line treatment. Factors that characterized the highest risk of survival shortening were: inability to apply third-line treatment, lack of best response to first-line therapy, poor PS, and C/G or G/G genotypes of ABCC2/MRP2 gene. We concluded that assessed factors had mainly prognostic and not predictive value. Finding reliable molecular predictors for second line docetaxel therapy requires further clinical trials. C1 [Szczyrek, Michal] Medical University of Lublin, Department of Pneumonology, Oncology and AllergologyLublin, Poland. [Mlak, Radoslaw] Medical University of Lublin, Department of Human PhysiologyLublin, Poland. [Krawczyk, Pawel] Medical University of Lublin, Department of Pneumonology, Oncology and AllergologyLublin, Poland. [Wojas-Krawczyk, Kamila] Medical University of Lublin, Department of Pneumonology, Oncology and AllergologyLublin, Poland. [Powrozek, Tomasz] Medical University of Lublin, Department of Pneumonology, Oncology and AllergologyLublin, Poland. [Szudy-Szczyrek, Aneta] Medical University of Lublin, Chair and Department of Haematooncology and Bone Marrow TransplantationLublin, Poland. [Zwolak, Agnieszka] Medical University of Lublin, Chair of Internal Medicine and Department of Internal Medicine in NursingLublin, Poland. [Daniluk, Jadwiga] Medical University of Lublin, Chair of Internal Medicine and Department of Internal Medicine in NursingLublin, Poland. [Milanowski, Janusz] Medical University of Lublin, Department of Pneumonology, Oncology and AllergologyLublin, Poland. RP Szczyrek, M (reprint author), Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Lublin, Poland. EM mszczyrek@yahoo.co.uk CR Shepherd FA, Dancey J, Ramlau R, Mattson K, Gralla R,O’Rourke Met al, 2000, Prospective randomized trial of docetaxel versus best supportive care in patients with non–small-cell lung cancer previously treated with patinum-based chemotherapy. J Clin Oncol 18: 2095–2103 Fossella FV, De Vore R, Kerr RN, Crawford J, Natale RR, Dunphy F et al, 2000, Randomized phase III trial of docetaxel versus vinorelbine or ifosfamide in patients with advanced non-small-cell lung cancer previously treated with platinum-containing chemotherapy regimens. The TAX 320 non-small cell lung cancer study group. J Clin Oncol 18(12):2354–2362 Stinchcombe TE, Socinski MA, 2008, Consideration for second line therapy of non-small cell lung cancer. Oncologist 13(supl 1): 28–36 Hanna N, Shepherd FA, Fossella FV, Pereira JR, DeMarinis F, von Pawel J et al, 2004, Randomized phase III trial of pemetrexed versus docetaxel in patients with non-small-cell lung cancer previously treated with chemotherapy. J Clin Oncol 1:1589–1597 Weiss GJ, Rosell R, Fossella F, Perry M, Stahel R, Barata F et al, 2007, The impact of induction chemotherapy on the outcome of second-line therapy with pemetrexed or docetaxel in patients with advanced non-small-cell lung cancer. Ann Oncol 18(3):453–460 Tibaldi C, Bernardini I, Chella A, Russo F, Vasile E, Malventi M et al, 2006, Second-line chemotherapy with a modified schedule of docetaxel in elderly patients with advanced-stage non-small-cell lung cancer. Clin Lung Cancer 7(6):401–405 Macedo-Perez EO, Morales-Oyarvide V, Mendoza-Garcia VO, Dorantes-Gallareta Y, Flores-Estrada D, Arrieta O, 2014, Long progression-free survival with first-line paclitaxel plus platinum is associated with improved response and progression-free survival with second-line docetaxel in advanced non-small-cell lung cancer. Cancer Chemother Pharmacol 74(4):681–690 Seve P, Dumontet CI, 2008, Class III beta-tubulin a predictive factor in patients receiving tubulin-binding agents? Lancet Oncol 9(2):168–175 Zhang HL, Ruan L, Zheng LM, Whyte D, Tzeng CM, Zhou XW, 2012, Association between class III β-tubulin expression and response to paclitaxel/vinorebine-based chemotherapy for non-small cell lung cancer: a meta-analysis. Lung Cancer 77(1):9–15 Kaira K, Takahashi T, Murakami H, Shukuya T, Kenmotsu H, Ono A et al, 2013)The role of βIII-tubulin in non-small cell lung cancer patients treated by taxane-based chemotherapy. Int J Clin Oncol 18(3):371–379 Haenisch S, Zimmermann U, Dazert E, Wruck CJ, Dazert P, Siegmund W, Kroemer HK, Warzok RW, Cascorbi I, 2007, Influence of polymorphisms of ABCB1 and ABCC2 on mRNA and protein expression in normal and cancerous kidney cortex. Pharmacogenomics J 7(1):56–65 Meier Y, Pauli-Magnus C, Zanger UM, Klein K, Schaeffeler E, Nussler AK, Nussler N, Eichelbaum M, Meier PJ, Stieger B, 2006, Interindividual variability of canalicular ATP-binding cassette, ABC)–transporter expression in human liver. Hepatology 44: 62–74 Isla D, Sarries C, Rosell R, Alonso G, Domine M, Taron M et al, 2004, Single nucleotide polymorphisms and outcome in docetaxel-cisplatin-treated advanced non-small-cell lung cancer. Ann Oncol 15(8):1194–1203 Kiyotani K, Mushiroda T, Kubo M, Zembutsu H, Sugiyama Y, Nakamura Y, 2008, Association of genetic polymorphisms in SLCO1B3 and ABCC2 with docetaxel-induced leukopenia. Cancer Sci 99(5):967–972 Campa D, Muller P, Edler L, Knoefel L, Barale R, Heussel CP, Thomas M, Canzian F, Risch AA, 2012, Comprehensive study of polymorphisms in ABCB1, ABCC2 and ABCG2 and lung cancer chemotherapy response and prognosis. Int J Cancer 131:2920– 2928 Lewis LD, Miller AA, Owzar K, Bies RR,Markova S, Jiang C et al, 2013, The relationship of polymorphisms in ABCC2 and SLCO1B3 with docetaxel pharmacokinetics and neutropenia: CALGB 60805. Pharmacogenet Genomics 23(1):29–33 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 607 EP 614 DI 10.1007/s12253-016-0156-4 PG 8 ER PT J AU Hu, W Li, X Li, Q Tan, Y Xu, B Xie, Q Deng, X Lu, B Jiang, J Wu, Ch AF Hu, Wenwei Li, Xiaodong Li, Qing Tan, Yan Xu, Bin Xie, Quanqin Deng, Xu Lu, Binfeng Jiang, Jingting Wu, Changping TI Interleukin-33 Expression does not Correlate with Survival of Gastric Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Interleukin-33; Gastric cancer; Clinical characteristics; Survival ID Interleukin-33; Gastric cancer; Clinical characteristics; Survival AB The aim of the study was to investigate IL-33 expression in gastric cancer (GC) and its association with the clinical characteristics and the prognosis. IL- 33 protein in tumor and corresponding adjacent tissues were detected by immunohistochemistry in 179 GC patients and clinical features plus prognostic value were analyzed via Pearson’s chi-square test and Kaplan– Meier test in Cox proportional hazards model, respectively. IL-33 protein levels were significantly lower in tumor tissues than adjacent tissues (29.05% vs. 78.77%, χ2 = 89.05, P < 0.001). The positive rate of IL-33 in the ulcerative type group was the lowest among all groups (P < 0.05). IL-33 levels were correlated with age (P = 0.025) and invasion depth (P = 0.030) while not significantly associated with the overall survival of GC patients. IL-33 expression is associated with age and invasive depth of GC patients but not an independent risk factor of prognosis. C1 [Hu, Wenwei] The Third Affiliated Hospital of Soochow University, Department of OncologyChangzhou, Jiangsu Province, China. [Li, Xiaodong] The Third Affiliated Hospital of Soochow University, Department of OncologyChangzhou, Jiangsu Province, China. [Li, Qing] The Third Affiliated Hospital of Soochow University, Department of PathologyChangzhou, Jiangsu Province, China. [Tan, Yan] The Third Affiliated Hospital of Soochow University, Department of PathologyChangzhou, Jiangsu Province, China. [Xu, Bin] The Third Affiliated Hospital of Soochow University, Department of Tumor Biological TreatmentChangzhou, Jiangsu Province, China. [Xie, Quanqin] The Third Affiliated Hospital of Soochow University, Department of Tumor Biological TreatmentChangzhou, Jiangsu Province, China. [Deng, Xu] The Third Affiliated Hospital of Soochow University, Department of Tumor Biological TreatmentChangzhou, Jiangsu Province, China. [Lu, Binfeng] University of Pittsburgh, School of Medicine, Department of ImmunologyPittsburgh, PA, USA. [Jiang, Jingting] The Third Affiliated Hospital of Soochow University, Department of Tumor Biological TreatmentChangzhou, Jiangsu Province, China. [Wu, Changping] The Third Affiliated Hospital of Soochow University, Department of OncologyChangzhou, Jiangsu Province, China. RP Wu, Ch (reprint author), The Third Affiliated Hospital of Soochow University, Department of Oncology, Changzhou, China. EM wcpjjt@163.com CR ChenW, Zheng R, Zeng H, Zhang S, He J, 2015, Annual report on status of cancer in China, 2011. Chinese journal of cancer research = Chung-kuo yen cheng yen chiu 27(1):2–12., DOI 10.3978/j. issn.1000-9604.2015.01.06 Dinarello CA, 1994, The biological properties of interleukin-1. Eur Cytokine Netw 5(6):517–531 Klemenz R, Hoffmann S, Werenskiold AK, 1989, Serum- and oncoprotein-mediated induction of a gene with sequence similarity to the gene encoding carcinoembryonic antigen. Proc Natl Acad Sci U S A 86(15):5708–5712 Bergers G, Reikerstorfer A, Braselmann S, Graninger P, Busslinger M, 1994, Alternative promoter usage of the Fos-responsive gene fit-1 generates mRNA isoforms coding for either secreted or membrane-bound proteins related to the IL-1 receptor. EMBO J 13(5):1176–1188 VerriWA Jr, Guerrero AT, Fukada SY, Valerio DA, Cunha TM, Xu D, Ferreira SH, LiewFY, Cunha FQ, 2008, IL-33 mediates antigeninduced cutaneous and articular hypernociception in mice. Proc Natl Acad Sci U S A 105(7):2723–2728., DOI 10.1073 /pnas.0712116105 Gao X,Wang X, Yang Q, Zhao X,WenW, Li G, Lu J, QinW, Qi Y, Xie F, Jiang J,Wu C, Zhang X, Chen X, Turnquist H, Zhu Y, Lu B, 2015, Tumoral expression of IL-33 inhibits tumor growth and modifies the tumor microenvironment through CD8+ T and NK cells. J Immunol 194(1):438–445., DOI 10.4049/jimmunol.1401344 Sun P, Ben Q, Tu S, Dong W, Qi X, Wu Y, 2011, Serum interleukin-33 levels in patients with gastric cancer. Dig Dis Sci 56(12):3596–3601., DOI 10.1007/s10620-011-1760-5 XX Y, Hu Z, Shen X, Dong LY, Zhou WZ, WH H, 2015, IL-33 promotes gastric cancer cell invasion and migration via ST2-ERK1/ 2 pathway. Dig Dis Sci 60(5):1265–1272., DOI 10.1007/s10620- 014-3463-1 Bie Q, Zhang P, Su Z, Zheng D, Ying X, Wu Y, Yang H, Chen D, Wang S, Xu H, 2014, Polarization of ILC2s in peripheral blood might contribute to immunosuppressive microenvironment in patients with gastric cancer. J Immunol Res 2014:923135., DOI 10.1155/2014/923135 Moussion C, Ortega N, Girard JP, 2008, The IL-1-like cytokine IL- 33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: a novel 'alarmin'? PLoS One 3(10):e3331., DOI 10.1371/journal.pone.0003331 Bergis D, Kassis V, Ranglack A, Koeberle V, Piiper A, Kronenberger B, Zeuzem S, Waidmann O, Radeke HH, 2013, High serum levels of the interleukin-33 receptor soluble ST2 as a negative prognostic factor in hepatocellular carcinoma. Transl Oncol 6(3):311–318 Jovanovic IP, Pejnovic NN, Radosavljevic GD, Pantic JM, Milovanovic MZ, Arsenijevic NN, Lukic ML, 2014, Interleukin- 33/ST2 axis promotes breast cancer growth and metastases by facilitating intratumoral accumulation of immunosuppressive and innate lymphoid cells. International Journal of Cancer Journal International Du Cancer 134(7):1669–1682., DOI 10.1002/ijc.28481 Brunner SM, Rubner C, Kesselring R, MartinM, Griesshammer E, Ruemmele P, Stempfl T, Teufel A, Schlitt HJ, Fichtner-Feigl S, 2015, Tumor-infiltrating, interleukin-33-producing effectormemory CD8(+, T cells in resected hepatocellular carcinoma prolong patient survival. Hepatology 61(6):1957–1967., DOI 10.1002 /hep.27728 Deng K, Wang H, Shan T, Chen Y, Zhou H, Zhao Q, Xia J, 2016, Tristetraprolin inhibits gastric cancer progression through suppression of IL-33. Sci Rep 6:24505., DOI 10.1038/srep24505 Bergis D, Kassis V, Radeke HH, 2016, High plasma sST2 levels in gastric cancer and their association with metastatic disease. Cancer Biomarkers : Section a of Disease Markers 16(1):117–125., DOI 10.3233/CBM-150547 Ye XL, Zhao YR, Weng GB, Chen YC, Wei XN, Shao JP, Ji H, 2015, IL-33-induced JNK pathway activation confers gastric cancer chemotherapy resistance. Oncol Rep 33(6):2746–2752., DOI 10.3892/or.2015.3898 Kim DY, Joo JK, Ryu SY, Park YK, Kim YJ, Kim SK, 2005, Clinicopathologic characteristics of gastric carcinoma in elderly patients: a comparison with young patients. World J Gastroenterol 11(1):22–26 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 615 EP 619 DI 10.1007/s12253-016-0167-1 PG 5 ER PT J AU Soleimani, Z Kheirkhah, D Sharif, RM Sharif, A Karimian, M Aftabi, Y AF Soleimani, Zahra Kheirkhah, Davood Sharif, Reza Mohammad Sharif, Alireza Karimian, Mohammad Aftabi, Younes TI Association of CCND1 Gene c.870G>A Polymorphism with Breast Cancer Risk: A Case-Control Study and a Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; CCND1 gene; Genetic polymorphism; c.870G>A; Meta-analysis ID Breast cancer; CCND1 gene; Genetic polymorphism; c.870G>A; Meta-analysis AB Cyclin D1 (CCND1) plays an essential role in regulating the progress of the cell cycle from G1 to S phase. There is a common c.870G>A polymorphism in the CCND1 gene. The aim of this study was to investigate the association of CCND1 gene c.870G>A polymorphism with breast cancer risk in a case-control study, which followed by a meta-analysis and an in silico analysis. Three hundred and thirty-five subjects composed of 174 women with breast cancer and 161 healthy controls were included in the case-control study. CCND1 gene c.870G>A genotyping was performed by PCR-RFLP. Meta-analysis was done for 14 studies composed of 7281 cases and 6820 controls. Some bioinformatics tools were applied to investigate the effects of c.870G>A on the mRNA splicing and structure. Our data obtained from casecontrol study revealed that GA genotype (OR: 1.89, 95%CI: 1.12–3.17, p = 0.017), AA genotype (OR: 1.95, 95%CI: 1.08– 3.53, p = 0.027), and A allele (OR: 1.44, 95%CI: 1.06–1.95, p = 0.019) were significantly associated with breast cancer risk. The results of meta-analysis showed a significant association between CCND1 c.870G>A polymorphism and breast cancer risk, especially in Caucasian population. In silico analysis revealed that c.870G>A transition affect CCND1 mRNA splicing and secondary structure. C1 [Soleimani, Zahra] Kashan University of Medical Sciences, Infectious Diseases Research CenterKashan, Iran. [Kheirkhah, Davood] Kashan University of Medical Sciences, Trauma Research CenterKashan, Iran. [Sharif, Reza Mohammad] Kashan University of Medical Sciences, Autoimmune Diseases Research CenterKashan, Iran. [Sharif, Alireza] Kashan University of Medical Sciences, Infectious Diseases Research CenterKashan, Iran. [Karimian, Mohammad] Kashan University of Medical Sciences, Gametogenesis Research CenterKashan, Iran. [Aftabi, Younes] Tabriz University of Medical Sciences, Drug Applied Research CenterTabriz, Iran. RP Kheirkhah, D (reprint author), Kashan University of Medical Sciences, Trauma Research Center, Kashan, Iran. EM kheirkhahdavood@gmail.com CR Makki J, 2015, Diversity of breast carcinoma: histological subtypes and clinical relevance. Clin Med Insights Pathol 8:23–31 Fabris VT, 2014, From chromosomal abnormalities to the identification of target genes in mouse models of breast cancer. Cancer Genet 207:233–246 Lee MM, Lin SS, 2000, Dietary fat and breast cancer. Annu Rev Nutr 20:221–248 Strumylaitė L, Mechonosina K, Tamasauskas S, 2010, Environmental factors and breast cancer. Medicina, Kaunas, 46: 867–873 Theodoropoulos GE, Michalopoulos NV, Pantou MP, Kontogianni P, Gazouli M, Karantanos T, Lymperi M, Zografos G, 2012, Caspase 9 promoter polymorphisms confer increased susceptibility to breast cancer. Cancer Genet 205:508–512 Musgrove EA, Caldon CE, Barraclough J, Stone A, Sutherland RL, 2011, Cyclin D as a therapeutic target in cancer. Nat Rev Cancer 11:558–572 Knudsen KE, Diehl JA, Haiman CA, Knudsen ES, 2006, Cyclin D1: polymorphism, aberrant splicing and cancer risk. Oncogene 25: 1620–1628 Petre-Draviam CE,Williams EB, Burd CJ, Gladden A, Moghadam H, Meller J, Diehl JA, Knudsen KE, 2005, A central domain of cyclin D1 mediates nuclear receptor corepressor activity. Oncogene 24:431–444 Kim KH, Roberts CW, 2014, Mechanisms by which SMARCB1 loss drives rhabdoid tumor growth. Cancer Genet 207:365–372 Betticher DC, Thatcher N, Altermatt HJ, Hoban P, Ryder WD, Heighway J, 1995, Alternate splicing produces a novel cyclin D1 transcript. Oncogene 11:1005–1011 Wu Y, Fu H, Zhang H, Huang H, ChenM, Zhang L, Yang H, Qin D, 2014, Cyclin D1, CCND1, G870A polymorphisms and cervical cancer susceptibility: a meta-analysis based on ten case–control studies. Tumour Biol 35:6913–6918 Grieu F,Malaney S,Ward R, Joseph D, Iacopetta B, 2003, Lack of association between CCND1 G870A polymorphism and the risk of breast and colorectal cancers. Anticancer Res 23:4257–4259 Krippl P, Langsenlehner U, Renner W, Yazdani-Biuki B, Wolf G, Wascher TC, Paulweber B, Weitzer W, Leithner A, Samonigg H, 2003, The 870G>A polymorphism of the cyclin D1 gene is not associated with breast cancer. Breast Cancer Res Treat 82:165–168 Forsti A, Angelini S, Festa F, Sanyal S, Zhang Z, Grzybowska E, Pamula J, Pekala W, Zientek H, Hemminki K, Kumar R, 2004, Single nucleotide polymorphisms in breast cancer. Oncol Rep 11: 917–922 Ceschi M, Sun CL, Van Den Berg D, Koh WP, MC Y, Probst- Hensch N, 2005, The effect of cyclin D1, CCND1, G870Apolymorphism on breast cancer risk is modified by oxidative stress among Chinese women in Singapore. Carcinogenesis 26:1457– 1464 Shu XO, Moore DB, Cai Q, Cheng J,WenW, Pierce L, Cai H, Gao Y, Zheng W, 2005, Association of cyclin D1 genotype with breast cancer risk and survival. Cancer Epidemiol Biomark Prev 14:91–97 Onay UV, Aaltonen K, Briollais L, Knight JA, Pabalan N, Kilpivaara O, Andrulis I, Blomqvist C, Nevanlinna H, Ozcelik H, 2008, Combined effect of CCND1 and COMT polymorphisms and increased breast cancer risk. BMC Cancer 8:6 Yu CP, Yu JC, Sun CA, Tzao C, Ho JY, Yen AM, 2008, Tumor susceptibility and prognosis of breast cancer associated with the G870A polymorphism of CCND1. Breast Cancer Res Treat 107: 95–102 Naidu R, Yip CH, Taib NA, 2008, Polymorphisms of HER2 Ile655Val and cyclin D1, CCND1, G870A are not associated with breast cancer risk but polymorphic allele of HER2 is associated with nodal metastases. Neoplasma 55:87–95 Justenhoven C, Pierl CB, Haas S, Fischer HP, Hamann U, Baisch C, Harth V, Spickenheuer A, Rabstein S, Vollmert C, Illig T, 2009, Polymorphic loci of E2F2, CCND1 and CCND3 are associated with HER2 status of breast tumors. Int J Cancer 124:2077–2081 Yaylim-Eraltan I, Ergen A, Gormus U, Arikan S, Kucucuk S, Sahin O, YIGIT N, Yildiz Y, Isbir T, 2009, Breast cancer and cyclin D1 gene polymorphism in Turkish women. In Vivo 23:767–772 Jeon S, Choi JY, Lee KM, Park SK, Yoo KY, Noh DY, Ahn SH, Kang D, 2010, Combined genetic effect of CDK7 and ESR1 polymorphisms on breast cancer. Breast Cancer Res Treat 121:737–742 Canbay E, Eraltan IY, Cercel A, Isbir T, Gazioglu E, Aydogan F, Cacina C, Cengiz A, Ferahman M, Zengin E, Unal H, 2010, CCND1 and CDKN1B polymorphisms and risk of breast cancer. Anticancer Res 30:3093–3098 Wasson MK, Chauhan PS, Singh LC, Katara D, Dev Sharma J, Zomawia E, Kataki A, Kapur S, Saxena S, 2014, Association of DNA repair and cell cycle gene variations with breast cancer risk in northeast Indian population: amultiple interaction analysis. Tumour Biol 35:5885–5894 Higgins JP, Thompson SG, Deeks JJ, Altman DG, 2003, Measuring inconsistency in meta-analyses. BMJ 327:557–560 Mantel N, Haenszel W, 1959, Statistical aspects of the analysisof data from retrospective studies of disease. J Natl Cancer Inst 22: 719–748 DerSimonian R, Laird N, 1986, Meta-analysis in clinical trials. Control Clin Trials 7:177–188 Begg CB, Mazumdar M, 1994, Operating characteristics of a rank correlation test for publication bias. Biometrics 50:1088–1101 Egger M, Davey Smith G, Schneider M, Minder C, 1997, Bias in meta-analysis detected by a simple, graphical test. BMJ 315:629– 634 Wang M, Marin A, 2006, Characterization and prediction of alternative splice sites. Gene 366:219–227 Hebsgaard SM, Korning PG, Tolstrup N, Engelbrecht J, Rouze P, Brunak S, 1996, Splice site prediction in Arabidopsis thaliana DNA by combining local and global sequence information. Nucleic Acids Res 24:3439–3452 Sabarinathan R, Tafer H, Seemann SE, Hofacker IL, Stadler PF, Gorodkin J, 2013, The RNAsnp web server: predicting SNP effects on local RNA secondary structure. Nucleic Acids Res 41:W475– W479 Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, KuhnM(2015, STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43:447–452 Ormandy CJ,Musgrove EA,Hui R,Daly RJ, 2003, Sutherland RL. Cyclin D1, EMS1 and 11q13 amplification in breast cancer. Breast Cancer Res Treat 78:323–335 Gillett C, Fantl V, Smith R, Fisher C, Bartek J, Dickson C, Barnes D, Peters G, 1994, Amplification and overexpression of cyclin D1 in breast cancer detected by immunohistochemical staining. Cancer Res 54:1812–1817 Baldin V, Lukas J, MarcoteMJ, Pagano M,Draetta G, 1993, Cyclin D1 is a nuclear protein required for cell cycle progression in G1. Genes Dev 7:812–821 Prall OW, Rogan EM, Musgrove EA, Watts CK, Sutherland RL, 1998, C-Myc or cyclin D1 mimics estrogen effects on cyclin ECdk2 activation and cell cycle reentry. Mol Cell Biol 18:4499–4508 Buckley MF, Sweeney KJ, Hamilton JA, Sini RL, Manning DL, Nicholson RI, DeFazio A,Watts CK, Musgrove EA, Sutherland RL, 1993, Expression and amplification of cyclin genes in human breast cancer. Oncogene 8:2127–2133 Lu C, Dong J, Ma H, Jin G, Hu Z, Peng Y, Guo X,Wang X, Shen H, 2008, CCND1 G870A polymorphism contributes to breast cancer susceptibility: a meta-analysis. Breast Cancer Res Treat 116:571– 575 Sawa H, Ohshima TA, Ukita H,Murakami H, Chiba Y, Kamada H, Hara M, Saito I, 1998, Alternatively spliced forms of cyclin D1 modulate entry into the cell cycle in an inverse manner. Oncogene 16:1701–1702 Weinstein IB, Begemann M, Zhou P, Han EK, Sgambato A, Doki Y, Arber N, Ciaparrone M, Yamamoto H, 1997, Disorders in cell circuitry associated with multistage cacinogenesis: exploitable targets for cancer prevention and therapy. Clin Cancer Res 3:2696– 2702 Zhang J, Li Y,Wang R,Wen D, SarbiaM, Kuang G,Wu M,Wei L, HeM, Zhang L,Wang S, 2003, Association of cyclin D1, G870A, polymorphism with susceptibility to esophageal and gastric cardiac carcinoma in a northern Chinese population. Int J Cancer 105:281– 284 Howe D, Lynas C, 2001, The cyclin D1 alternative transcripts [a] and [b] are expressed in normal and malignant lymphocytes and their relative levels are influenced by the polymorphism at codon 241. Haematologica 86:563–569 Jamali S, Karimian M, Nikzad H, Aftabi Y, 2016, The c. − 190 C>A transversion in promoter region of protamine1 gene as a genetic risk factor for idiopathic oligozoospermia. Mol Biol Rep 43: 795–802 Nikzad H, KarimianM, Sareban K, Khoshsokhan M, Hosseinzadeh Colagar A, 2015, MTHFR-Ala222Val and male infertility: a study in Iranian men, an updated meta-analysis and an in silico-analysis. Reprod BioMed Online 31:668–680 Karimian M, Nikzad H, Tameh AA, Taherian A, Darvishi FZ, Haghighatnia MJ, 2015, SPO11-C631T gene polymorphism: association with male infertility and an in silico-analysis. J Family Reprod Health 9:155–163 Karimian M, Hosseinzadeh Colagar A, 2016)Methionine synthase A2756G transition might be a risk factor for male infertility: evidences from seven case-control studies. Mol Cell Endocrinol 425: 1–10 Aftabi Y, Colagar AH, Mehrnejad F, 2016, An in silico approach to investigate the source of the controversial interpretations about the phenotypic results of the human AhR-gene G1661A polymorphism. J Theor Biol 21(393):1–5 Shen LX, Basilion JP, Stanton VP Jr, 1999, Single-nucleotide polymorphisms can cause different structural folds of mRNA. Proc Natl Acad Sci U S A 96:7871–7876 Wang DG, Fan JB, Siao CJ, Berno A, Young P, Sapolsky R, Ghandour G, Perkins N, Winchester E, Spencer J, Kruglyak L, 1998, Large-scale identification, mapping, and genotyping of single nucleotide polymorphisms in the human genome. Science 280: 1077–1082 Pabalan N, Bapat B, Sung L, Jarjanazi H, Francisco-Pabalan O, Ozcelik H, 2008, Cyclin D1 Pro241Pro, CCND1-G870A, polymorphism is associated with increased cancer risk in human populations: a meta-analysis. Cancer Epidemiol Biomark Prev 17:2773– 2781 Lu C, Dong J, Ma H, JinG,Hu Z, PengY,Guo X, Wang X, Shen H, 2009, CCND1 G870A polymorphism contributes to breast cancer susceptibility: ameta-analysis. Breast Cancer Res Treat 116:571–575 Cui J, Shen L, Wang Y, 2012, Specific CCND1 G870A alleles associated with breast cancer susceptibility: a meta-analysis of 5, 528 cases and 5,353 controls.Asian Pac J Cancer Prev 13:5023–5025 Dai X, Li T, Bai Z, Yang Y, Liu X, Zhan J, Shi B, 2015, Breast cancer intrinsic subtype classification, clinical use and future trends. Am J Cancer Res 5(10):2929 Perou CM, 2011, Molecular stratification of triple-negative breast cancers. Oncologist 16(Supplement 1):61–70 Tobin NP, Sims AH, Lundgren KL, Lehn S, Landberg G, 2011, Cyclin D1, Id1 and EMT in breast cancer. BMC Cancer 11(1):417 Arnold A, Papanikolaou A, 2005, Cyclin D1 in breast cancer pathogenesis. J Clin Oncol 23(18):4215–4224 Grillo M, Bott MJ, Khandke N, McGinnis JP, Miranda M, Meyyappan M, Rosfjord EC, Rabindran SK, 2006, Validation of cyclin D1/CDK4 as an anticancer drug target in MCF-7 breast cancer cells: effect of regulated overexpression of cyclin D1 and siRNA-mediated inhibition of endogenous cyclin D1 and CDK4 expression. Breast Cancer Res Treat 95(2): 185–194 Alao JP, 2007, The regulation of cyclin D1 degradation: roles in cancer development and the potential for therapeutic invention. Mol Cancer 6(1):1 Zang Y, Zhao S, Doll MA, Hein DW, 2004, The T341C, Ile114Thr, polymorphism of N-acetyltransferase 2 yields slow acetylator phenotype by enhanced protein degradation. Pharmacogenet Genomics 14(11):717–723 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 621 EP 631 DI 10.1007/s12253-016-0165-3 PG 11 ER PT J AU Butz, H Nemeth, K Czenke, D Liko, I Czirjak, S Zivkovic, V Baghy, K Korbonits, M Kovalszky, I Igaz, P Racz, K Patocs, A AF Butz, Henriett Nemeth, Kinga Czenke, Dora Liko, Istvan Czirjak, Sandor Zivkovic, Vladimir Baghy, Kornelia Korbonits, Marta Kovalszky, Ilona Igaz, Peter Racz, Karoly Patocs, Attila TI Systematic Investigation of Expression of G2/M Transition Genes Reveals CDC25 Alteration in Nonfunctioning Pituitary Adenomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pituitary adenoma; Cell cycle; G2/M transition; CDC25A; miRNA ID Pituitary adenoma; Cell cycle; G2/M transition; CDC25A; miRNA AB Dysregulation of G1/S checkpoint of cell cycle has been reported in pituitary adenomas. In addition, our previous finding showing that deregulation of Wee1 kinase by microRNAs together with other studies demonstrating alteration of G2/Mtransition in nonfunctioning pituitary adenomas (NFPAs) suggest that G2/M transition may also be important in pituitary tumorigenesis. To systematically study the expression of members of the G2/M transition in NFPAs and to investigate potential microRNA (miRNA) involvement. Totally, 80 NFPA and 14 normal pituitary (NP) tissues were examined. Expression of 46 genes encoding members of the G2/M transition was profiled on 34 NFPA and 10 NP samples on TaqMan Low Density Array. Expression of CDC25A and two miRNAs targeting CDC25Awere validated by individual quantitative real time PCR using TaqMan assays. Protein expression of CDC25A, CDC25C, CDK1 and phospho-CDK1 (Tyr-15) was investigated on tissue microarray and immunohistochemistry. Several genes’ expression alteration were observed in NFPA compared to normal tissues by transcription profiling. On protein level CDC25A and both the total and the phospho-CDK1 were overexpressed in adenoma tissues. CDC25A correlated with nuclear localized CDK1 (nCDK1) and with tumor size and nCDK1 with Ki-67 index. Comparing primary vs. recurrent adenomas we found that Ki-67 proliferation index was higher and phospho-CDK1 (inactive form) was downregulated in recurrent tumors compared to primary adenomas. Investigating the potential causes behind CDC25A overexpression we could not find copy number variation at the coding region nor expression alteration of CDC25A regulating transcription factors however CDC25A targeting miRNAs were downregulated in NFPA and negatively correlated with CDC25A expression. Our results suggest that among alterations of G2/M transition of the cell cycle, overexpression of the CDK1 and CDC25A may have a role in the pathogenesis of the NFPA and that CDC25A is potentially regulated by miRNAs. C1 [Butz, Henriett] Hungarian Academy of Sciences, Molecular Medicine Research Group, 46 Szentkiralyi str, H-1088 Budapest, Hungary. [Nemeth, Kinga] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Czenke, Dora] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Liko, Istvan] Hungarian Academy of SciencesBudapest, Hungary. [Czirjak, Sandor] National Institute of NeurosurgeryBudapest, Hungary. [Zivkovic, Vladimir] University of Belgrade, School of Medicine, Institute of Forensic MedicineBelgrade, Serbia. [Baghy, Kornelia] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Korbonits, Marta] Queen Mary University of London, Barts and the London School of Medicine, Department of EndocrinologyLondon, UK. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Igaz, Peter] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Racz, Karoly] Hungarian Academy of Sciences, Molecular Medicine Research Group, 46 Szentkiralyi str, H-1088 Budapest, Hungary. [Patocs, Attila] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. RP Butz, H (reprint author), Hungarian Academy of Sciences, Molecular Medicine Research Group, H-1088 Budapest, Hungary. EM butz.henriett@med.semmelweis-univ.hu CR Dolecek TA, Propp JM, Stroup NE, Kruchko C, 2012, CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005-2009. Neuro Oncol Suppl 5: v1–49 McDowell BD, Wallace RB, Carnahan RM, Chrischilles EA, Lynch CF, Schlechte JA, 2011, Demographic differences in incidence for pituitary adenoma. Pituitary 14:23–30 Daly AF, Tichomirowa MA, Beckers A, 2009, The epidemiology and genetics of pituitary adenomas. Best Pract Res Clin Endocrinol Metab 23:543–554 Ezzat S, Asa SL, CouldwellWT, Barr CE, Dodge WE, Vance ML, McCutcheon IE, 2004, The prevalence of pituitary adenomas: a systematic review. Cancer 101:613–619 Tjornstrand A, Gunnarsson K, Evert M, Holmberg E, Ragnarsson O, Rosen T, Filipsson Nystrom H, 2014, The incidence rate of pituitary adenomas in western Sweden for the period 2001-2011. Eur J Endocrinol 171:519–526 Beckers A, 2010, Higher prevalence of clinically relevant pituitary adenomas confirmed. Clin Endocrinol, Oxf, 72:290–291 Fernandez A, Karavitaki N,Wass JA, 2010, Prevalence of pituitary adenomas: a community-based, cross-sectional study in Banbury, Oxfordshire, UK). Clin Endocrinol, Oxf, 72:377–382 Heaney AP, 2005, Pituitary tumour pathogenesis. Br Med Bull 75– 76:81–97 Musat M, Morris DG, Korbonits M, Grossman AB, 2010, Cyclins and their related proteins in pituitary tumourigenesis. Mol Cell Endocrinol 326:25–29 Ogino A, Yoshino A, Katayama Y, Watanabe T, Ota T, Komine C, Yokoyama T, Fukushima T, 2005, The p15(INK4b)/p16(INK4a)/ RB1 pathway is frequently deregulated in human pituitary adenomas. J Neuropathol Exp Neurol 64:398–403 Yoshino A, Katayama Y, Ogino A, Watanabe T, Yachi K, Ohta T, Komine C, Yokoyama T, Fukushima T, 2007, Promoter hypermethylation profile of cell cycle regulator genes in pituitary adenomas. J Neurooncol 83:153–162 Zhao J, Dahle D, Zhou Y, Zhang X, Klibanski A, 2005, Hypermethylation of the promoter region is associatedwith the loss of MEG3 gene expression in human pituitary tumors. J Clin Endocrinol Metab 90:2179–2186 Gejman R, Batista DL, Zhong Y, Zhou Y, Zhang X, Swearingen B, Stratakis CA, Hedley-Whyte ET, Klibanski A, 2008, Selective loss ofMEG3 expression and intergenic differentiallymethylated region hypermethylation in the MEG3/DLK1 locus in human clinically nonfunctioning pituitary adenomas. J Clin Endocrinol Metab 93: 4119–4125 Simpson DJ, Frost SJ, Bicknell JE, Broome JC, McNicol AM, Clayton RN, Farrell WE, 2001, Aberrant expression of G, 1)/S regulators is a frequent event in sporadic pituitary adenomas. Carcinogenesis 22:1149–1154 Jordan S, Lidhar K, Korbonits M, Lowe DG, Grossman AB, 2000, Cyclin D and cyclin E expression in normal and adenomatous pituitary. Eur J Endocrinol 143:R1–R6 Turner HE, Nagy Z, Sullivan N, Esiri MM, Wass JA, 2000, Expression analysis of cyclins in pituitary adenomas and the normal pituitary gland. Clin Endocrinol, Oxf, 53:337–344 Butz H, Liko I, Czirjak S, Igaz P, Khan MM, Zivkovic V, Balint K, Korbonits M, Racz K, Patocs A, 2010, Down-regulation of Wee1 kinase by a specific subset of microRNA in human sporadic pituitary adenomas. J Clin Endocrinol Metab 95:E181–E191 Backert S, Gelos M, Kobalz U, Hanski ML, Bohm C, Mann B, Lovin N, Gratchev A, Mansmann U, Moyer MP, Riecken EO, Hanski C, 1999, Differential gene expression in colon carcinoma cells and tissues detected with a cDNA array. Int J Cancer 82:868– 874 Yoshida T, Tanaka S, Mogi A, Shitara Y, Kuwano H, 2004, The clinical significance of Cyclin B1 and Wee1 expression in nonsmall- cell lung cancer. Ann Oncol 15:252–256 Zhan X, Desiderio DM, 2010, Signaling pathway networks mined from human pituitary adenoma proteomics data. BMC Med Genomics 3:13 Yu R, Ren SG, Melmed S, 2002, Proteasome inhibitors induce apoptosis in growth hormone- and prolactin-secreting rat pituitary tumor cells. J Endocrinol 174:379–386 Yang JH, Hsia TC, Kuo HM, Chao PD, Chou CC,Wei YH, Chung JG, 2006, Inhibition of lung cancer cell growth by quercetin glucuronides via G2/M arrest and induction of apoptosis. Drug Metab Dispos 34:296–304 Al-Shraim M, Asa SL, 2006, The 2004World Health Organization classification of pituitary tumors: what is new? Acta Neuropathol 111:1–7 Butz H, Liko I, Czirjak S, Igaz P, Korbonits M, Racz K, Patocs A, 2011, MicroRNA profile indicates downregulation of the TGFbeta pathway in sporadic non-functioning pituitary adenomas. Pituitary 14:112–124 Kobayashi I, Oka H, Naritaka H, Sato Y, Fujii K, Kameya T, 2002, Expression of Pit-1 and growth hormone-releasing hormone receptor mRNA in human pituitary adenomas: difference among functioning, silent, and other nonfunctioning adenomas. Endocr Pathol 13:83–98 Asa SL, Bamberger AM, Cao B, Wong M, Parker KL, Ezzat S, 1996, The transcription activator steroidogenic factor-1 is preferentially expressed in the human pituitary gonadotroph. J Clin Endocrinol Metab 81:2165–2170 Daniely M, Aviram A, Adams EF, Buchfelder M, Barkai G, Fahlbusch R, Goldman B, Friedman E, 1998, Comparative genomic hybridization analysis of nonfunctioning pituitary tumors. J Clin Endocrinol Metab 83:1801–1805 Metzger AK, Mohapatra G, Minn YA, Bollen AW, Lamborn K, Waldman FM, Wilson CB, Feuerstein BG, 1999, Multiple genetic aberrations including evidence of chromosome 11q13 rearrangement detected in pituitary adenomas by comparative genomic hybridization. J Neurosurg 90:306–314 Harada K, Nishizaki T, Ozaki S, Kubota H, Harada K, Okamura T, Ito H, Sasaki K, 1999, Cytogenetic alterations in pituitary adenomas detected by comparative genomic hybridization. Cancer Genet Cytogenet 112:38–41 Trautmann K, Thakker RV, Ellison DW, Ibrahim A, Lees PD, Harding B, Fischer C, Popp S, Bartram CR, Jauch A, 2001, Chromosomal aberrations in sporadic pituitary tumors. Int J Cancer 91:809–814 Szymas J, Schluens K, Liebert W, Petersen I, 2002, Genomic instability in pituitary adenomas. Pituitary 5:211–219 Michaelis KA, Knox AJ, Xu M, Kiseljak-Vassiliades K, Edwards MG, Geraci M, Kleinschmidt-DeMasters BK, Lillehei KO, Wierman ME, 2011, Identification of growth arrest and DNAdamage- inducible gene beta, GADD45beta, as a novel tumor suppressor in pituitary gonadotrope tumors. Endocrinology 152:3603– 3613 Elston MS, Gill AJ, Conaglen JV, Clarkson A, Shaw JM, Law AJ, Cook RJ, Little NS, Clifton-Bligh RJ, Robinson BG, McDonald KL, 2008, Wnt pathway inhibitors are strongly down-regulated in pituitary tumors. Endocrinology 149:1235–1242 Evans CO, Young AN, Brown MR, Brat DJ, Parks JS, Neish AS, Oyesiku NM, 2001, Novel patterns of gene expression in pituitary adenomas identified by complementary deoxyribonucleic acid microarrays and quantitative reverse transcription-polymerase chain reaction. J Clin Endocrinol Metab 86:3097–3107 Morris DG, Musat M, Czirjak S, Hanzely Z, Lillington DM, Korbonits M, Grossman AB, 2005, Differential gene expression in pituitary adenomas by oligonucleotide array analysis. Eur J Endocrinol 153:143–151 Moreno CS, Evans CO, Zhan X, Okor M, Desiderio DM, Oyesiku NM, 2005, Novel molecular signaling and classification of human clinically nonfunctional pituitary adenomas identified by gene expression profiling and proteomic analyses. Cancer Res 65:10214– 10222 Zhan X, Desiderio DM, 2010, Signaling pathway networks mined from human pituitary adenoma proteomics data. BMC Med Genomics. 3:13 Boutros R, Lobjois V, Ducommun B, 2007, CDC25 phosphatases in cancer cells: key players? Good targets? Nat Rev Cancer 7:495– 507 Huang JC, Babak T, Corson TW, Chua G, Khan S, Gallie BL, Hughes TR, Blencowe BJ, Frey BJ, Morris QD, 2007, Using expression profiling data to identify human microRNA targets. Nat Methods. 4(12):1045–1049 Shi L, Zhang J, Pan T, Zhou J, GongW, Liu N, Fu Z, You Y, 2010, MiR-125b is critical for the suppression of human U251 glioma stem cell proliferation. Brain Res 1312:120–126 Lee SO, Masyuk T, Splinter P, Banales JM, Masyuk A, Stroope A, Larusso N, 2008, MicroRNA15a modulates expression of the cellcycle regulator Cdc25A and affects hepatic cystogenesis in a rat model of polycystic kidney disease. J Clin Invest 118:3714–3724 de Oliveira PE, Zhang L, Wang Z, Lazo JS, 2009, Hypoxiamediated regulation of Cdc25A phosphatase by p21 and miR-21. Cell Cycle 8:3157–3164 Sarkar S, Dey BK, Dutta A, 2010, MiR-322/424 and -503 are induced during muscle differentiation and promote cell cycle quiescence and differentiation by down-regulation of Cdc25A. Mol Biol Cell 21:2138–2149 Yang X, Feng M, Jiang X, Wu Z, Li Z, Aau M, Yu Q, 2009, miR- 449a and miR-449b are direct transcriptional targets of E2F1 and negatively regulate pRb-E2F1 activity through a feedback loop by targeting CDK6 and CDC25A. Genes Dev 23:2388–2393 Galaktionov K, Beach D, 1991, Specific activation of cdc25 tyrosine phosphatases by B-type cyclins: evidence for multiple roles of mitotic cyclins. Cell 67:1181–1194 Sadhu K, Reed SI, Richardson H, Russell P, 1990, Human homolog of fission yeast cdc25 mitotic inducer is predominantly expressed in G2. Proc Natl Acad Sci U S A 87:5139–5143 Fernandez-Vidal A, Mazars A,Manenti S, 2008, CDC25A: a rebel within the CDC25 phosphatases family? Anticancer Agents Med Chem 8:825–831 Mailand N, Podtelejnikov AV, Groth A,Mann M, Bartek J, Lukas J, 2002, Regulation of G, 2)/M events by Cdc25A through phosphorylation-dependent modulation of its stability. EMBO J 21:5911–5920 Li M, Yin S, Yuan J, Wei L, Ai JS, Hou Y, Chen DY, Sun QY, 2008, Cdc25A promotes G2/M transition in oocytes. Cell Cycle 7:1301–1302 Melixetian M, Klein DK, Sorensen CS, Helin K, 2009, NEK11 regulates CDC25A degradation and the IR-induced G2/M checkpoint. Nat Cell Biol 11:1247–1253 Kristjansdottir K, Rudolph J, 2004, Cdc25 phosphatases and cancer. Chem Biol 11:1043–1051 Wierinckx A, Auger C, Devauchelle P, Reynaud A, Chevallier P, Jan M, Perrin G, Fevre-Montange M, Rey C, Figarella-Branger D, Raverot G, Belin MF, Lachuer J, Trouillas J, 2007, A diagnostic marker set for invasion, proliferation, and aggressiveness of prolactin pituitary tumors. Endocr Relat Cancer 14:887–900 Raverot G, Wierinckx A, Dantony E, Auger C, Chapas G, Villeneuve L, Brue T, Figarella-Branger D, Roy P, Jouanneau E, Jan M, Lachuer J, Trouillas J, 2010, Prognostic factors in prolactin pituitary tumors: clinical, histological, and molecular data from a series of 94 patients with a long postoperative follow-up. J Clin Endocrinol Metab 95:1708–1716 Mraz M, Dolezalova D, Plevova K, Stano Kozubik K, Mayerova V, Cerna K, Musilova K, Tichy B, Pavlova S, Borsky M, Verner J, Doubek M, Brychtova Y, Trbusek M, Hampl A, Mayer J, Pospisilova S, 2012, MicroRNA-650 expression is influenced by immunoglobulin gene rearrangement and affects the biology of chronic lymphocytic leukemia. Blood 119:2110–2113 Chien WW, Domenech C, Catallo R, Kaddar T, Magaud JP, Salles G, Ffrench M, 2011, Cyclin-dependent kinase 1 expression is inhibited by p16(INK4a, at the post-transcriptional level through the microRNA pathway. Oncogene 30:1880–1891 Takeshita F, Patrawala L, Osaki M, Takahashi RU, Yamamoto Y, Kosaka N, Kawamata M, Kelnar K, Bader AG, Brown D, Ochiya T, 2010, Systemic delivery of synthetic microRNA-16 inhibits the growth of metastatic prostate tumors via downregulation of multiple cell-cycle genes. Mol Ther 18:181–187 Li XH, Wang EL, Zhou HM, Yoshimoto K, Qian ZR, 2014, MicroRNAs in human pituitary adenomas. Int J Endocrinol 2014: 435171 Sivapragasam M, Rotondo F, Lloyd RV, Scheithauer BW, Cusimano M, Syro LV, KovacsK, 2011, MicroRNAs in the human pituitary. Endocr Pathol 22:134–143 Liang S, Chen L,HuangH, Zhi D(2013, The experimental study of miRNA in pituitary adenomas. Turk Neurosurg 23:721–727 Bottoni A, Zatelli MC, Ferracin M, Tagliati F, Piccin D, Vignali C, Calin GA, Negrini M, Croce CM, Degli Uberti EC, 2007, Identification of differentially expressed microRNAs by microarray: a possible role for microRNA genes in pituitary adenomas. J Cell Physiol 210:370–377 D'Angelo D, Palmieri D, Mussnich P, Roche M, Wierinckx A, Raverot G, Fedele M, Croce CM, Trouillas J, Fusco A, 2012, Altered microRNA expression profile in human pituitary GH adenomas: down-regulation of miRNA targeting HMGA1, HMGA2, and E2F1. J Clin Endocrinol Metab 97:E1128–E1138 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 633 EP 641 DI 10.1007/s12253-016-0163-5 PG 9 ER PT J AU Niedworok, Ch Tschirdewahn, S Reis, H Lehmann, N Szucs, M Nyirady, P Romics, I Rubben, H Szarvas, T AF Niedworok, Christian Tschirdewahn, Stephan Reis, Henning Lehmann, Nils Szucs, Miklos Nyirady, Peter Romics, Imre Rubben, Herbert Szarvas, Tibor TI Serum Chromogranin A as a Complementary Marker for the Prediction of Prostate Cancer-Specific Survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chromogranin A; CGA; Prostate cancer; Prognosis; KRYPTOR ID Chromogranin A; CGA; Prostate cancer; Prognosis; KRYPTOR AB Better prognostication of clinically localized prostate cancer (PCA) is urgently needed. Former studies using different study end-points provided controversial results regarding the prognostic value of serum chromogranin A (CGA) in clinically localized PCA. However, serum CGA was not tested for correlation with the most significant study end-point of long-term disease-specific survival (DSS). CGA and matrix metalloproteinase-7 (MMP7) levels were measured by the BRAHMS KRYPTOR in two independent patient groups with 127 serum and 110 plasma samples. CGA and MMP7 concentrations were correlated with clinicopathological and survival data. In addition, we tested the combinations of CGA with PSA and with a currently identified prognostic factor, MMP7, for their prognostic value. CGA concentrations were significantly elevated in advanced compared to clinically localized cases both in serum and plasma samples (45 vs. 23 ng/ml, p < 0.001 and; 41 vs. 22 ng/ml; p = 0.002 respectively). In accordance, high CGA levels were correlated with poor DSS. In clinically localized cases, CGA levels alone were not prognostic, but its dichotomized combinations with PSA or MMP7 were independently associated with DSS (HR: 4.88, 95% CI: 1.35–17.71, p = 0.016, HR: 7.46, 1.65–33.63, p = 0.009, respectively). Elevated serum CGA levels in progressed PCA and its prognostic value suggest a potential for CGA in disease monitoring. Our results revealed no independent prognostic value for CGA as a single serum marker in clinically localized cases. However, when combining with PSA or MMP7, CGA may improve both marker’s performance in distinguishing between clinically significant and indolent PCAs. C1 [Niedworok, Christian] University of Duisburg-Essen, Department of UrologyEssen, Germany. [Tschirdewahn, Stephan] University of Duisburg-Essen, Department of UrologyEssen, Germany. [Reis, Henning] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Lehmann, Nils] University of Duisburg-Essen, University Hospital of Essen, Institute of Medical Informatics, Biometry and EpidemiologyEssen, Germany. [Szucs, Miklos] Semmelweis University, Department of Urology, Ulloi ut 78/b, 1082 Budapest, Hungary. [Nyirady, Peter] Semmelweis University, Department of Urology, Ulloi ut 78/b, 1082 Budapest, Hungary. [Romics, Imre] Semmelweis University, Department of Urology, Ulloi ut 78/b, 1082 Budapest, Hungary. [Rubben, Herbert] University of Duisburg-Essen, Department of UrologyEssen, Germany. [Szarvas, Tibor] University of Duisburg-Essen, Department of UrologyEssen, Germany. RP Szarvas, T (reprint author), University of Duisburg-Essen, Department of Urology, Essen, Germany. EM sztibusz@gmail.com CR Jemal A, Siegel R, Xu J,Ward E, 2011, Cancer statistics, 2010. CA Cancer J Clin 61:133–134 Schroder FH, Hugosson J, Roobol MJ, Tammela TL, Zappa M, Nelen V, Kwiatkowski M, Lujan M, Maattanen L, Lilja H, Denis LJ, Recker F, Paez A, Bangma CH, Carlsson S, Puliti D, Villers A, Rebillard X, Hakama M, Stenman UH, Kujala P, Taari K, Aus G, Huber A, van der Kwast TH, van Schaik RH, de Koning HJ, Moss SM, Auvinen A, 2014, ERSPC investigators. Screening and prostate cancer mortality: results of the European randomised study of screening for prostate cancer, ERSPC, at 13 years of follow-up. Lancet 384:2027–2035 Nesland JM, Holm R, Johannessen JV, Gould VE, 1988, Neuroendocrine differentiation in breast lesions. Pathol Res Pract 183:214–221 Sun Y, Niu J, Huang J, 2009, Neuroendocrine differentiation in prostate cancer. Am J Transl Res 1:148–162 O’Connor DT, Deftos LJ, 1986, Secretion of chromogranin a by peptide-producing endocrine neoplasms. N Engl J Med 314:1145–1151 Campana D, Nori F, Piscitelli L, Morselli-Labate AM, Pezzilli R, Corinaldesi R, Tomassetti P, 2007, Chromogranin a: is it a useful marker of neuroendocrine tumors? J Clin Oncol 25:1967–1973 Khan MO, Ather MH, 2011, Chromogranin A–serum marker for prostate cancer. J Pak Med Assoc 61:108–111 Szarvas T, Becker M, Vom Dorp F, Meschede J, Scherag A, Bankfalvi A, Reis H, Schmid KW, Romics I, Rubben H, Ergun S, 2011, Elevated serum matrix metalloproteinase 7 levels predict poor prognosis after radical prostatectomy. Int J Cancer 128:1486–1492 Sciarra A, Voria G, Monti S, Mazzone L, Mariotti G, Pozza M, D’Eramo G, Silverio FD, 2004, Clinical understaging in patients with prostate adenocarcinoma submitted to radical prostatectomy: predictive value of serum chromogranin a. Prostate 58:421–428 Alessandro S, Vincenzo G, Maria AG, Stefano S, Alessandro G, Salvatore M, Vincenzo T, Franco DS, 2007, Chromogranin a and biochemical progression-free survival in prostate adenocarcinomas submitted to radical prostatectomy. Endocr Relat Cancer 14:625–632 De Nunzio C, Albisinni S, Presicce F, Lombardo R, Cancrini F, Tubaro A, 2014, Serum levels of chromogranin a are not predictive of high-grade, poorly differentiated prostate cancer: results from an Italian biopsy cohort. Urol Oncol 32:80–84 Niedworok C, Vom Dorp F, Tschirdewahn S, Rubben H, Reis H, Szucs M, Szarvas T, 2016, Validation of the diagnostic and prognostic relevance of serumMMP7 levels in renal cell cancer by using a novel automated fluorescent immunoassay method. Int Urol Nephrol 48:355–361 Krabbe JG,Monaghan PJ, Russell J, de Rijke YB, 2016, Analytical evaluation of a second generation assay for chromogranin a; a dualsite study. Clin Chem Lab Med 54:e139–e142 Mathis G, 1993, Rare earth cryptates and homogeneous fluoroimmunoassays with human sera. Clin Chem 39:1953–1959 Sobin LHG, Gospodarowicz MK, Wittekind C, 2009, TNM classification of malignant Tumours. 7th ed. Wiley-Blackwell, Hoboken Berruti A, Mosca A, Tucci M, Terrone C, Torta M, Tarabuzzi R, Russo L, Cracco C, Bollito E, Scarpa RM, Angeli A, Dogliotti L, 2005, Independent prognostic role of circulating chromogranin a in prostate cancer patients with hormone-refractory disease. Endocr Relat Cancer 12:109–117 Burgio SL, Conteduca V, Menna C, Carretta E, Rossi L, Bianchi E, Kopf B, Fabbri F, Amadori D, De Giorgi U, 2014, Chromogranin a predicts outcome in prostate cancer patients treated with abiraterone. Endocr Relat Cancer 21:487–493 Conteduca V, Burgio SL, Menna C, Carretta E, Rossi L, Bianchi E, Masini C, Amadori D, De Giorgi U, 2014, Chromogranin a is a potential prognostic marker in prostate cancer patients treated with enzalutamide. Prostate 74:1691–1696 Harrell FE Jr, 2001, Regression modelling strategies with applications to linear models, logistic regression, and survival analysis. Springer, New York NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 643 EP 650 DI 10.1007/s12253-016-0171-5 PG 8 ER PT J AU Peng, W Wang, Z Fan, H AF Peng, Wei Wang, Zhuo Fan, Hong TI LncRNA NEAT1 Impacts Cell Proliferation and Apoptosis of Colorectal Cancer via Regulation of Akt Signaling SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Long noncoding RNA; NEAT1; Biomarker ID Colorectal cancer; Long noncoding RNA; NEAT1; Biomarker AB Long noncoding RNA (lncRNA) have been reported to modulate oncogenesis and be used to be target for tumor. The role of lncRNA NEAT1 (nuclear paraspeckle assembly transcript 1, Gene ID: 283131) in colorectal cancer (CRC) keeps unknown. This work was to investigate the pattern of lncRNA NEAT1 (NEAT1) expression in CRC and its functional value and biological significance. NEAT1 expression was analyzed in 56 cancer tissues and cell lines in CRC cases. Results showed that NEAT1 was significantly overexpressed in CRC cells and tissues. Clinicpathologic detection verified that high NEAT1 expression associated with bulk in CRC. The serum contents of NEAT1 were observably elevated comparing with healthy cases (P < 0.05). The levels of NEAT1 were elevated in distinguishing CRC from normal (ROCAUC = 0.9471; P < 0.01). Moreover, Kaplan–Meier analysis found that NEAT1 elevation led to adverse survival (P < 0.05). Further experiments illustrated that of NEAT1 knockdown signally inhibited growth and facilitated apoptosis. Importantly, we confirmed that Akt signaling pathway was inactivated after loss of NEAT1 in CRC. Taken together, this work support the first evidence that NEAT1 can be used to be a promising biomarker and target for novel treatment for human CRC. C1 [Peng, Wei] Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Department of Medical Oncology, No. 42 Baiziting Road, 210009 Nanjing, China. [Wang, Zhuo] Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Laboratory of Cancer Research, No. 42 Baiziting Road, 210009 Nanjing, China. [Fan, Hong] First People’s Hospital of Yunnan Province, Department of Gastroenterology, No. 175 Jinbi Road, 650032 Kunming, China. RP Peng, W (reprint author), Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Department of Medical Oncology, 210009 Nanjing, China. EM wpeng01@hotmail.com CR Weitz J, KochM, Debus J, Hohler T, Galle PR, BuchlerMW(2005, Colorectal cancer. Lancet 365(9454):153–165 Kjeldsen BJ, Kronborg O, Fenger C, Jorgensen OD, 1997, A prospective randomized study of follow-up after radical surgery for colorectal cancer. Br J Surg 84(5):666–669 Kinzler KW, Vogelstein B, 1996, Lessons from hereditary colorectal cancer. Cell 87(2):159–170 Walther A, Johnstone E, Swanton C,Midgley R, Tomlinson I, Kerr D, 2009, Genetic prognostic and predictive markers in colorectal cancer. Nat Rev Cancer 9(7):489–499 Wilusz JE, Sunwoo H, Spector DL, 2009, Long noncoding RNAs: functional surprises from the RNA world. Genes Dev 23:1494– 1504 Ponting CP, Oliver PL, Reik W, 2009, Evolution and functions of long noncoding RNAs. Cell 136:629–641 PengW,Wu G, Fan H,Wu J, Feng J, 2015, Long noncoding RNA SPRY4-IT1 predicts poor patient prognosis and promotes tumorigenesis in gastric cancer. Tumour biology: the journal of the international society for. Oncodevelopmental Biology and Medicine 36: 6751–6758 PengW, Fan H, 2015, Long non-coding RNA PANDAR correlates with poor prognosis and promotes tumorigenesis in hepatocellular carcinoma. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 72:113–118 PengW, Feng J, 2016, Long noncoding RNA LUNAR1 associates with cell proliferation and predicts a poor prognosis in diffuse large B-cell lymphoma. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 77:65–71 West JA, Davis CP, Sunwoo H, Simon MD, Sadreyev RI,Wang PI et al, 2014, The long noncoding RNAs NEAT1 and MALAT1 bind active chromatin sites. Mol Cell 55:791–802 Li Z, Liu YH, Diao HY, Ma J, Yao YL, 2016, Long noncoding RNA NEAT1 promotes glioma pathogenesis by regulating miR- 449b-5p/c-met axis. Tumor Biol 37:673–683 Zeng CW, Xu Y, Xu L, Yu XB, Cheng JJ, Yang LJ et al, 2014, Inhibition of long non-coding RNA NEAT1 impairs myeloid differentiation in acute promyelocytic leukemia cells. BMC Cancer 14:693 Wang P, Wu T, Zhou H, Jin Q, He G, Yu H et al, 2016, Long noncoding RNA NEAT1 promotes laryngeal squamous cell cancer through regulating miR-107/CDK6 pathway. Journal of experimental & clinical cancer research: CR 35:22 Peng W, Zhang J, Liu J, 2014, URG11 predicts poor prognosis of pancreatic cancer by enhancing epithelial-mesenchymal transitiondriven invasion.Medical oncology, Northwood, London, England, 31:64 Peng W, Wu J, Feng J, 2016, Long noncoding RNA HULC predicts poor clinical outcome and represents pro-oncogenic activity in diffuse large B-cell lymphoma. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 79:188–193 Li X, Zhang Y, Cao S, Chen X, Lu Y, Jin H et al, 2009, Reduction of TIP30 correlates with poor prognosis of gastric cancer patients and its restoration drastically inhibits tumor growth and metastasis. Int J Cancer 124:713–721 Vivanco I, Sawyers CL, 2002, The phosphatidylinositol 3-kinase- AKT pathway in human cancer. Nat Rev Cancer 2:489–501 Adams JM, Cory S, 2007, The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene 26:1324–1337 Chao DT, Korsmeyer SJ, 1998, BCL-2 FAMILY: regulators of cell death. Annu Rev Immunol 16:395–419 Adams JM, Cory S, 2001, Life-or-death decisions by the Bcl-2 protein family. Trends Biochem Sci 26:61–66 Han L, Zhang EB, Yin DD, Kong R, Xu TP, ChenWMet al, 2015, Low expression of long noncoding RNA PANDAR predicts a poor prognosis of non-small cell lung cancer and affects cell apoptosis by regulating Bcl-2. Cell Death Dis 6:e1665 Peng W, Wu JZ, Feng JF, 2016, Long noncoding RNA HULC predicts poor clinical outcome and represents pro-oncogenic activity in diffuse large B-cell lymphoma. Biomedicine & Pharmacotherapy 79:188–193 Altomare DA, Testa JR, 2005, Perturbations of the AKT signaling pathway in human cancer. Oncogene 24:7455–7464 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 651 EP 656 DI 10.1007/s12253-016-0172-4 PG 6 ER PT J AU Zhu, Y Liu, M Yun, X Wang, D Bai, Y Zhang, G Ji, B Jing, Ch AF Zhu, Yusen Liu, Min Yun, Xiaojing Wang, Dongmei Bai, Yuhuan Zhang, Guizhi Ji, Bei Jing, Changchun TI Meta-Analysis for the Therapeutic Effect of Neoadjuvant Therapy in Resectable Esophageal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal cancer; Neoadjuvant chemoradiotherapy; Neoadjuvant chemotherapy; Neoadjuvant radiotherapy; Meta-analysis ID Esophageal cancer; Neoadjuvant chemoradiotherapy; Neoadjuvant chemotherapy; Neoadjuvant radiotherapy; Meta-analysis AB We aimed to review the therapeutic effects of neoadjuvant chemoradiotherapy (NCRT), chemotherapy (NCT), and radiotherapy (NRT) on patients with resectable Esophageal cancer (EsC) by comparison with surgery alone (SA). PubMed, EMBASE and Cochrane were searched for eligible studies published up to March 2015. Cochrane reviews were used for quality assessment. Eight primary outcomes were analyzed. Risk ratios (RRs)/ hazard ratios (HRs) and corresponding 95% confidence intervals (95% CIs) were calculated using the random- or fixed- effects model. Heterogeneity was assessed using the Chi-square-based Q statistic and the I2test. Publication bias was examined by the Begg’s funnel plot. Totally 24 articles including 4718 EsC cases were eligible for this meta-analysis. The quality of the literatures was relatively high. Significant difference was found in five-year survival rate (RR = 1.45, 95% CI: 1.17–1.79, P < 0.01) between patients treated with NCT and SA, while the eight enrolled primary outcomes were all statistically different between NCRT and SA, and significant difference was identified in three-year survival between NCRT and NCT (RR = 1.35, 95% CI: 1.14–1.60, P < 0.01). No obvious publication bias was observed. NCRT and NCT provide an obvious benefit for EsC treatment over SA, and NCRT possesses a clear advantage compared with NCT. C1 [Zhu, Yusen] The Second People’s Hospital of Liaocheng, Department of Gastroenterology, 252600 Linqing, Shandong, China. [Liu, Min] The Second People’s Hospital of Liaocheng, Department of Emergency, 252600 Linqing, Shandong, China. [Yun, Xiaojing] The Second People’s Hospital of Liaocheng, Department of Gastroenterology, 252600 Linqing, Shandong, China. [Wang, Dongmei] The Second People’s Hospital of Liaocheng, Department of Gastroenterology, 252600 Linqing, Shandong, China. [Bai, Yuhuan] The Second People’s Hospital of Liaocheng, Department of Gastroenterology, 252600 Linqing, Shandong, China. [Zhang, Guizhi] The Second People’s Hospital of Liaocheng, Department of Gastroenterology, 252600 Linqing, Shandong, China. [Ji, Bei] The Second People’s Hospital of Liaocheng, Department of Gastroenterology, 252600 Linqing, Shandong, China. [Jing, Changchun] The Second People’s Hospital of Liaocheng, Department of Gastroenterology, 252600 Linqing, Shandong, China. RP Liu, M (reprint author), The Second People’s Hospital of Liaocheng, Department of Emergency, 252600 Linqing, China. EM minliu01@126.com CR Pennathur A, Gibson MK, Jobe BA, Luketich JD, 2013, Oesophageal carcinoma. Lancet 381:400–412 Vallbohmer D, Brabender J, Grimminger P, Schroder W, Holscher AH, 2011, Predicting response to neoadjuvant therapy in esophageal cancer. Expert Rev Anticancer Ther 11:1449–1455 Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics, 2009. CA Cancer J Clin 59:225–249 Shahbaz SarwarCM, Luketich JD, Landreneau RJ, AbbasG, 2010, Esophageal cancer: an update. Int J Surg 8:417–422 Krasna MJ, 2010, Multimodality therapy for esophageal cancer. Oncology, Williston Park, 24:1134–1138 D'journo XB, Thomas PA, 2014, Current management of esophageal cancer. J Thorac Dis 6(Suppl 2):S253–S264 Baba Y, Watanabe M, Yoshida N, Baba H, 2014, Neoadjuvant treatment for esophageal squamous cell carcinoma. World J Gastrointest Oncol 6:121–128 Shah RD, Cassano AD, Neifeld JP, 2014, Neoadjuvant therapy for esophageal cancer. World J Gastrointest Oncol 6:403–406 Pandey D, Pandey R, Garg PK, 2015, Neoadjuvant therapy and lymphadenectomy in esophageal cancer: both are essential tomaximize survival benefit. Ann Surg., DOI 10.1097/SLA.0000000000001160 Berry MF, 2014, Esophageal cancer: staging system and guidelines for staging and treatment. J Thorac Dis 6(Suppl 3):S289–S297 Gebski V, Burmeister B, Smithers BM, Foo K, Zalcberg J, Simes J, 2007, Survival benefits from neoadjuvant chemoradiotherapy or chemotherapy in oesophageal carcinoma: a meta-analysis. Lancet Oncol 8:226–234 Sjoquist KM, Burmeister BH, Smithers BM et al, 2011, Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: an updated meta-analysis. Lancet Oncol 12:681–692 Higgins JP, Green S, 2008, Cochrane handbook for systematic reviews of interventions, vol 5. Wiley Online Library, Chichester Higgins J, Green S, 2011, Cochrane handbook for systematic reviews of interventions, vol 5.1.0.Wiley Online Library, Chichester Allum WH, Stenning SP, Bancewicz J, Clark PI, Langley RE, 2009, Long-term results of a randomized trial of surgery with or without preoperative chemotherapy in esophageal cancer. J Clin Oncol 27:5062–5067 Ancona E, Ruol A, Santi S et al, 2001, Only pathologic complete response to neoadjuvant chemotherapy improves significantly the long term survival of patients with resectable esophageal squamous cell carcinoma. Cancer 91:2165–2174 Boonstra JJ, Kok TC, Wijnhoven BP et al, 2011, Chemotherapy followed by surgery versus surgery alone in patients with resectable oesophageal squamous cell carcinoma: long-term results of a randomized controlled trial. BMC Cancer 11:181 Bosset J-F, Gignoux M, Triboulet J-P et al, 1997, Chemoradiotherapy followed by surgery compared with surgery alone in squamous-cell cancer of the esophagus. N Engl J Med 337:161–167 Burmeister BH, Smithers BM, Gebski Vet al, 2005, Surgery alone versus chemoradiotherapy followed by surgery for resectable cancer of the oesophagus: a randomised controlled phase III trial. Lancet Oncol 6:659–668 Burmeister BH, Thomas JM, Burmeister EA et al, 2011, Is concurrent radiation therapy required in patients receiving preoperative chemotherapy for adenocarcinoma of the oesophagus? A randomised phase II trial. Eur J Cancer 47:354–360 Cao XF, He XT, Ji L, Xiao J, Lv J, 2009, Effects of neoadjuvant radiochemotherapy on pathological staging and prognosis for locally advanced esophageal squamous cell carcinoma. Dis Esophagus 22:477–481 Fujiwara Y, Yoshikawa R, Kamikonya N et al, 2013, Neoadjuvant chemoradiotherapy followed by esophagectomy vs. surgery alone in the treatment of resectable esophageal squamous cell carcinoma. Mol Clin Oncol 1:773–779 Kelsen DP, Ginsberg R, Pajak TF et al, 1998, Chemotherapy followed by surgery compared with surgery alone for localized esophageal cancer. N Engl J Med 339:1979–1984 Law S, Fok M, Chow S, Chu K-M, Wong J, 1997, Preoperative chemotherapy versus surgical therapy alone for squamous cell carcinoma of the esophagus: a prospective randomized trial. J Thorac Cardiovasc Surg 114:210–217 Lee J-L, Park S, Kim S-B et al, 2004, A single institutional phase III trial of preoperative chemotherapy with hyperfractionation radiotherapy plus surgery versus surgery alone for resectable esophageal squamous cell carcinoma. Ann Oncol 15:947–954 Lv J, Cao X-F, Zhu B, Ji L, Tao L, Wang D-D, 2010, Long-term efficacy of perioperative chemoradiotherapy on esophageal squamous cell carcinoma. World J Gastroenterol 16:1649 Maipang T, Vasinanukorn P, Petpichetchian C et al, 1994, Induction chemotherapy in the treatment of patients with carcinoma of the esophagus. J Surg Oncol 56:191–197 Mariette C, Dahan L, Mornex F et al, 2014, Surgery alone versus chemoradiotherapy followed by surgery for stage I and II esophageal cancer: final analysis of randomized controlled phase III trial FFCD 9901. J Clin Oncol 32:2416–2422 Natsugoe S, Okumura H, Matsumoto M et al, 2006, Randomized controlled study on preoperative chemoradiotherapy followed by surgery versus surgery alone for esophageal squamous cell cancer in a single institution. Dis Esophagus 19:468–472 Nygaard K, Hagen S, Hansen HS et al, 1992, Pre-operative radiotherapy prolongs survival in operable esophageal carcinoma: a randomized, multicenter study of pre-operative radiotherapy and chemotherapy. The second Scandinavian trial in esophageal cancer. World J Surg 16:1104–1109 Prise EL, Etienne PL, Meunier B et al, 1994, A randomized study of chemotherapy, radiation therapy, and surgery versus surgery for localized squamous cell carcinoma of the esophagus. Cancer 73: 1779–1784 Schlag PM, 1992, Randomized trial of preoperative chemotherapy for squamous cell cancer of the esophagus. Arch Surg 127:1446–1450 Stahl M, Walz MK, Stuschke M et al, 2009, Phase III comparison of preoperative chemotherapy compared with chemoradiotherapy in patients with locally advanced adenocarcinoma of the esophagogastric junction. J Clin Oncol 27:851–856 Tepper J, Krasna MJ, Niedzwiecki D et al, 2008, Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 26:1086–1092 Urba SG, Orringer MB, Turrisi A, Iannettoni M, Forastiere A, StrawdermanM(2001, Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol 19:305–313 Van Hagen P, Hulshof M, Van Lanschot J et al, 2012, Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366:2074–2084 Walsh TN, Noonan N, Hollywood D, Kelly A, Keeling N, Hennessy TP, 1996, A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 335:462–467 Ychou M, Boige V, Pignon J-P et al, 2011, Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol 29:1715–1721 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 657 EP 663 DI 10.1007/s12253-016-0164-4 PG 7 ER PT J AU Stefanko, E Rybka, J Jazwiec, B Haus, O Stapor, S Kuliczkowski, K Wrobel, T AF Stefanko, Ewa Rybka, Justyna Jazwiec, Bozena Haus, Olga Stapor, Sylwia Kuliczkowski, Kazimierz Wrobel, Tomasz TI Significance of OCT1 Expression in Acute Myeloid Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute myeloid leukemia; OCT1; Resistance to chemotherapy ID Acute myeloid leukemia; OCT1; Resistance to chemotherapy AB Organic cation transporter 1 (OCT1) is one of the membrane proteins in the large solute carrier (SLC) family. It participates in the transport of organic cations, i.e. nutrients, neurotransmitters, metabolites or drugs in an electrogenic manner and translocate various cationic cytostatics. Knowledge concerning the expression of drug transporters in tumor cells may help to develop cytotoxic agents that are targeted to specific tumors. OCT1 expression and its relationship to the proliferation of cancer cells, development of metastases and resistance to chemotherapy has been observed in solid tumors. There is no data concerning the significance of OCT1 expression in the clinical course and treatment results in acute myeloid leukemia (AML). The objective of the study was firstly to evaluate OCT1 mRNA expression in patients with newly diagnosed de novo AML, and secondly to compare the obtained results to the healthy control group as well as analyze them according to leukemia subtypes, CD34 expression, cytogenetic and molecular factors and treatment results. 101 patients with AML, excluding the subtype classified as M3 by French-American-British (FAB) criteria, were analyzed. The control group consisted of 26 healthy individuals. The evaluated material was bone marrow (BM). Real-time quantitative polymerase chain reaction (RQ-PCR) was used in the study as a method of evaluating OCT1 mRNA expression. The study showed a statistically significant lower expression of OCT1 mRNA in patients with AML in comparison to the control group. The level of OCT1 mRNA expression was lowest for CD34+ leukemia. No significant correlation between OCT1 mRNA expression and cytogenetic and molecular factors was observed. A significant influence of OCT1 mRNA expression on the clinical outcome of the disease was observed: patients with lower expression had higher chances of achieving complete remission (CR) and longer overall survival (OS). C1 [Stefanko, Ewa] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow TransplantationWroclaw, Poland. [Rybka, Justyna] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow TransplantationWroclaw, Poland. [Jazwiec, Bozena] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow TransplantationWroclaw, Poland. [Haus, Olga] Collegium Medicum Nicolaus Copernicus University, Department of Clinical GeneticsBydgoszcz, Poland. [Stapor, Sylwia] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow TransplantationWroclaw, Poland. [Kuliczkowski, Kazimierz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow TransplantationWroclaw, Poland. [Wrobel, Tomasz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow TransplantationWroclaw, Poland. RP Stefanko, E (reprint author), Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw, Poland. EM ewastefanko@gmail.com CR Christ O, Feuring-Buske M, Hiddemann W et al, 2007, Pathobiology of acute myeloid leukemia. Med Clin 102:290–295 Redmond KM, Wilson TR, Johnston PG et al, 2008, Resistance mechanisms to cancer chemotherapy. Front Biosci 13:5138–5154 Koepsell H, Lips K, Volk C, 2007, Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications. Pharm Res 7:1227–1251 Grundemann D, Gorboulev V, Gambaryan S et al, 1994, Drug excretion mediated by a new prototype of polyspecific transporter. Nature 372:549–552 Hayer-ZillgenM, BrussM, Bonisch H, 2002, Expression and pharmacological profile of the human organic cation transporters hOCT1, hOCT2, and hOCT3. Br J Pharmacol 136:829–836 Engler JR, Zannettino AC, Bailey CG et al, 2011, OCT-1 function varies with cell lineage but is not influenced by BCR-ABL. Haematologica 96:213–220 Komazawa H, Yamaguchi H, HidakaKet al, 2013, Renal uptake of substrates for organic anion transporters Oat1 and Oat3 and organic cation transporters Oct1 and Oct2 is altered in rats with adenineinduced chronic renal failure. J Pharm Sci 3:1086–1094 Nies AT, Koepsell H, Winter S et al, 2009, Expression of organic cation transporters OCT1, SLC22A1, and OCT3, SLC22A3, is affected by genetic factors and cholestasis in human liver. Hepatology 4:1227–1240 Huang Y, Sadee W, 2006, Membrane transporters and channels in chemoresistance and sensitivity of tumor cells. Cancer Lett 239: 168–182 Zhang S, Lovejoy KS, Shima JE, 2006, Organic cation transporters are determinants of oxaliplatin cytotoxicity. Cancer Res 66:8847–8857 Arimany-Nardi C,Montraveta A, Lee-Verges E et al, 2015, Human organic cation transporter 1, hOCT1, as a mediator of bendamustine uptake and cytotoxicity in chronic lymphocytic leukemia, CLL, cells. Pharmacogenomics J 4:363–371 Daley GQ, Van Etten RA, Baltimore D, 1990, Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome. Science 247:824–830 O’Hare T, Corbin AS, Druker BJ, 2006, Targeted CML therapy: controlling drug resistance, seeking cure. Curr Opin Genet Dev 16: 92–99 Apperley JF, 2007, Mechanisms of resistance to imatinib in chronic myeloid leukemia. Lancet Oncol 8:1018–1029 Wang L, Giannoudis A, Lane S et al, 2008, Expression of the uptake drug transporter hOCT1 is an important clinical determinant of the response to imatinib in chronic myeloid leukemia. Clin Pharmacol Ther 83:258–264 Jonker JW, Schinkel AH, 2004, Pharmacological and physiological functions of the polyspecific organic cation transporters: OCT1, 2 and 3, SLC22A1-3). J Pharmacol Exp Ther 308:2–9 Bennett JM, Catovsky D, Daniel MTet al, 1985, Proposals for the classification of the acute leukemias. A report of the French- American-British, FAB, co-operative group. Ann Intern Med 103: 620–625 Swerdlow SH, Campo E, Harris NL et al, 2008, WHO classification of Tumours of Haematopoietic and lymphoid tissues, 4th edn. International Agency for Research on Cancer, Lyon, France Dohner H, Estey EH, Amadori S et al, 2010, Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European leukemia net. Blood 115:453–474 Fey MF, Greil R, Jost LM, 2005, ESMO guidelines task force. ESMO minimum clinical recommendations for the diagnosis, treatment and follow-up of acute myeloblastic leukemia, AML, in adult patients. Ann Oncol 1:48–49 Sorror ML, Maris MB, Storb R et al, 2005, Hematopoietic cell transplantation, HCT)- specific comorbidity index: a new tool for risk assessment before allogeneic HCT. Blood 106:2912–2919 Yates J, Glidewell O, Wiernik P et al, 1982, Cytosine arabinoside with daunorubicin or adriamycin for therapy of acute myelocytic leukemia: a CALGB study. Blood 60:454–462 Wrzesien-Kus A, Robak T, Jamroziak K et al, 2002, The treatment of acute myeloid leukemia with mitoxantrone, etoposide and lowdose cytarabine in elderly patients - a report of polish acute leukemia group, PALG, phase II study. Neoplasma 49:405–411 Kabayashi D, Aizawa S, Maeda Tet al, 2004, Expression of organic cation transporterOCTN1 in hematopoietic cells during erythroid differentiation. Exp Hematol 32:1156–1162 Karlic H, Lohninger A, Laschan C et al, 2003, Downregulation of carnitine acyltransferases and organic cation transporter OCTN2 in mononuclear cells in healthy elderly and patients with myelodysplastic syndromes. J Mol Med 81:435–442 Bazeos A, Marin D, Gerrard G et al, 2009, BCR-ABL1 oncogene downregulates the expression of OCT1 in CML. ASH Annu Meet Abstr 114:3248 Bazeos A, Marin D, Reid AG et al, 2010, hOCT1 transcript levels and single nucleotide polymorphisms as predictive factors for response to imatinib in chronic myeloid leukemia. Leukemia 24: 1243–1245 Engler JR, Frede A, Saunders VA et al, 2010, Chronic myeloid leukemia CD34+ cells have reduced uptake of imatinib due to low OCT-1 activity. Leukemia 24:765–770 Razga F, Racil Z, Polakova KMet al, 2011, The predictive value of human organic cation transporter 1 and ABCB1 expression levels in different cell populations of patients with de novo chronic myelogenous leukemia. Int J Hematol 94:303–306 White DL, Dang P, Engler J et al, 2010, Functional activity of the OCT-1 protein is predictive of long-term outcome in patients with chronic-phase chronic myeloid leukemia treated with imatinib. J Clin Oncol 28:2761–2767 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 665 EP 671 DI 10.1007/s12253-016-0161-7 PG 7 ER PT J AU Khan, AS Zeng, Z Shia, J Paty, BP AF Khan, A Sajid Zeng, Zhaoshi Shia, Jinru Paty, B Philip TI EGFR Gene Amplification and KRAS Mutation Predict Response to Combination Targeted Therapy in Metastatic Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Cetuximab; Bevacizumab; Metastasis; EGFR; KRAS ID Colorectal cancer; Cetuximab; Bevacizumab; Metastasis; EGFR; KRAS AB Genetic variability in KRAS and EGFR predicts response to cetuximab in irinotecan refractory colorectal cancer. Whether these markers or others remain predictive in combination biologic therapies including bevacizumab is unknown. We identified predictive biomarkers from patients with irinotecan refractory metastatic colorectal cancer treated with cetuximab plus bevacizumab. Patients who received cetuximab plus bevacizumab for irinotecan refractory colorectal cancer in either of two Phase II trials conducted were identified. Tumor tissue was available for 33 patients. Genomic DNA was extracted and used for mutational analysis of KRAS, BRAF, and p53 genes. Fluorescence in situ hybridization was performed to assess EGFR copy number. The status of single genes and various combinations were tested for association with response. Seven of 33 patients responded to treatment. KRAS mutations were found in 14/33 cases, and 0 responded to treatment (p = 0.01). EGFR gene amplification was seen in 3/33 of tumors and in every case was associated with response to treatment (p < 0.001). TP53 and BRAF mutations were found in 18/33 and 0/33 tumors, respectively, and there were no associations with response to either gene. EGFR gene amplification and KRAS mutations are predictive markers for patients receiving combination biologic therapy of cetuximab plus bevacizumab for metastatic colorectal cancer. One marker or the other is present in the tumor of half of all patients allowing treatment response to be predicted with a high degree of certainty. The role for molecular markers in combination biologic therapy seems promising. C1 [Khan, A Sajid] Yale University School of Medicine, Department of Surgery, Section of Surgical Oncology, 310 Cedar Street, FMB 130, 06520 New Haven, CT, USA. [Zeng, Zhaoshi] Memorial Sloan-Kettering Cancer Center, Colorectal Service, Department of SurgeryNew York, NY, USA. [Shia, Jinru] Memorial Sloan-Kettering Cancer Center, Colorectal Service, Department of PathologyNew York, NY, USA. [Paty, B Philip] Memorial Sloan-Kettering Cancer Center, Colorectal Service, Department of SurgeryNew York, NY, USA. RP Khan, AS (reprint author), Yale University School of Medicine, Department of Surgery, Section of Surgical Oncology, 06520 New Haven, USA. EM sajid.khan@yale.edu CR Siegel RL, Miller KD, Jemal A, 2015, Cancer statistics, 2015. CA Cancer J Clin 65:5–29 Macdonald JS, 1999, Adjuvant therapy of colon cancer. CA Cancer J Clin 49:202–219 Meyerhardt JA, Mayer RJ, 2005, Systemic therapy for colorectal cancer. N Engl J Med 352:476–487 Ruzzo A, Graziano F, Canestrari E, Magnani E, 2010, Molecular predictors of efficacy to anti-EGFR agents in colorectal cancer patients. Curr Ca Drug Targets 10:68–79 Hicklin DJ, Ellis LM, 2005, Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol 23:1011–1027 Cunningham D, Humblet Y, Siena S et al, 2004, Cetuximab Monotherapy and cetuximab plus irinotecan in irinotecanrefractory metastatic colorectal cancer. N Engl J Med 351:337–345 Saltz LB, Lenz HJ, Kindler HL et al, 2007, Randomized phase II trial of cetuximab, bevacizumab, and irinotecan compared with cetuximab and bevacizumab alone in irinotecan-refractory colorectal cancer: the BOND-2 study. J Clin Oncol 25(29):4557–4561 Jonker DJ, O'Callaghan CJ, Karapetis CS et al, 2007, Cetuximab for the treatment of colorectal cancer. N Engl J Med 357:2040–2048 Tabernero J, Van Cutsem E, Diaz-Rubio E et al, 2007, Phase II trial of cetuximab in combination with fluorouracil, leucovorin, and Oxaliplatin in the first-line treatment ofmetastatic colorectal cancer. J Clin Oncol 25:5225–5232 MoroniM, Veronese S, Benvenuti S et al, 2005, Gene copy number for epidermal growth factor receptor, EGFR, and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. The Lancet Oncology 6:279–286 Lievre A, Bachet J-B, Le Corre D et al, 2006, KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 66:3992–3995 Di Fiore F, Blanchard F, Charbonnier F et al, 2007, Clinical relevance of KRAS mutation detection in metastatic colorectal cancer treated by cetuximab plus chemotherapy. Br J Cancer 96:1166–1169 Khambata-Ford S, Garrett CR,Meropol NJ et al, 2007, Expression of Epiregulin and Amphiregulin and K-ras mutation status predict disease control in metastatic colorectal cancer patients treated with cetuximab. J Clin Oncol 25:3230–3237 Sartore-Bianchi A, Moroni M, Veronese S et al, 2007, Epidermal growth factor receptor Gene copy number and clinical outcome of metastatic colorectal cancer treated with panitumumab. J Clin Oncol 25(22):3238–3245 Chung KY, Shia J, Kemeny NE et al, 2005, Cetuximab shows activity in colorectal cancer patients with tumors that do not express the epidermal growth factor receptor by immunohistochemistry. J Clin Oncol 23:1803–1810 Shia J, Klimstra DS, Li AR et al, 2005, Epidermal growth factor receptor expression and gene amplification in colorectal carcinoma: an immunohistochemical and chromogenic in situ hybridization study. Mod Pathol 18:1350–1356 Cerea G, Ricotta R, Schiavetto I et al, 2006, Cetuximab for treatment of metastatic colorectal cancer. Ann Oncol 17:vii66–vii67 Khanna M, Park P, Zirvi M et al, 1999, Multiplex PCR/LDR for detection of K-ras mutations in primary colon tumors. Oncogene 18:27–38 Sorlie T, Johnsen H, Vu P, Lind GE, Lothe R, Borresen-Dale AL, 2005, Mutation screening of the TP53 gene by temporal temperature gradient gel electrophoresis. Methods Mol Biol 291:207–216 Guldberg P, Nedergaard T, Nielsen HJ, Olsen AC, Ahrenkiel V, Zeuthen J, 1997, Single-step DGGE-based mutation scanning of the p53 gene: application to genetic diagnosis of colorectal cancer. Hum Mutat 9:348–355 Khan SA, Idrees K, Forslund A et al, 2008, Genetic variants in germline TP53 and MDM2 SNP309 are not associated with early onset colorectal cancer. J Surg Oncol 97(7):621–625 Spindler KL, Lindebjerg J, Nielsen JN et al, 2006, Epidermal growth factor receptor analyses in colorectal cancer: a comparison of methods. Int J Oncol 29(5):1159–1165 Nash GM, GimbelM, Shia J et al, 2010, KRAS mutation correlates with accelerated metastatic progression in patients with colorectal liver metastases. Ann Surg Oncol 17(2):572–578 Sinicrope FA, Mahoney MR, Yoon HH et al, 2015, Analysis of molecular markers by anatomic tumor site in stage III colon carcinomas from adjuvant chemotherapy trial NCCTGN0147, alliance). Clin Cancer Res 21(23):5294–5304 Kato J, Futamura M, Kanematsu M et al, 2016, Combination therapy with zoledronic acid and cetuximab effectively suppresses growth of colorectal cancer cells regardless or KRAS status. Int J Cancer 138(6):1516–1527 Dienstmann R, Salazer R, Tabernero J, 2015, Personalizing colon cancer adjuvant therapy: selecting optimal treatments for individual patients. J Clin Oncol 33(16):1787–1796 Gerlinger M, Rowan AJ, Horswell S et al, 2012, Intratumoral heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366(10):883–892 Benvenuti S, Sartore-Bianchi A, Di Nicolantonio F et al, 2007, Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti-epidermal growth factor receptor antibody therapies. Cancer Res 67:2643– 2648 Siena S, Sartre-Bianchi A, Di Nicolantonio F, Balfour J, Bardelli A., 2009, Biomarkers predicting clinical outcome of epidermal growth factor receptor-targeted therapy in metastatic colorectal cancer. J Natl Cancer Inst 101(19):1308–24 Pao W, Wang TY, Riely GJ et al, 2005, KRAS mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib. PLoS Med 2:e17 Lenz H-J, Van Cutsem E, Khambata-Ford S et al, 2006, Multicenter phase II and translational study of cetuximab in metastatic colorectal carcinoma refractory to irinotecan, Oxaliplatin, and Fluoropyrimidines. J Clin Oncol 24:4914– 4921 Chapman SJ, McKavanagh D, Burge ME et al, 2016, Effectiveness of bevacizumab and cetuximab in metastatic colorectal cancer across selected public hospitals in Queensland. Asia Pac J Clin Oncol., DOI 10.1111/ajco.12518 Larsen FO, Pfeiffer P, Nielsen D et al, 2011, Bevacizumab in combination with cetuximab and irinotecan after failure of cetuximab and irinotecan in patients with metastatic colorectal cancer. Acta Oncol 50(4):574–577 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 673 EP 677 DI 10.1007/s12253-016-0166-2 PG 5 ER PT J AU Sitarek, P Skala, E Toma, M Wielanek, M Szemraj, J Skorski, T Bialas, JA Sakowicz, T Kowalczyk, T Radek, M Wysokinska, H Sliwinski, T AF Sitarek, Przemyslaw Skala, Ewa Toma, Monika Wielanek, Marzena Szemraj, Janusz Skorski, Tomasz Bialas, J Adam Sakowicz, Tomasz Kowalczyk, Tomasz Radek, Maciej Wysokinska, Halina Sliwinski, Tomasz TI Transformed Root Extract of Leonurus sibiricus Induces Apoptosis through Intrinsic and Extrinsic Pathways in Various Grades of Human Glioma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Apoptosis; Cell cycle; Gene expression; Transformed roots of Leonurus sibiricus; Mitochondrial membrane potential; Reactive oxygen species ID Apoptosis; Cell cycle; Gene expression; Transformed roots of Leonurus sibiricus; Mitochondrial membrane potential; Reactive oxygen species AB This study determines the influence of transformed root (TR) extract of Leonurus sibiricus L. on various grades (IIII) of human glioma cells derived from patients. This plant occurs in southern Asia and Siberia and is widely used as a medicinal plant with various biological activities. Chromatographic profile of TR extract have revealed the presence of various polyphenolic compounds (4-hydroxybenzoic acid, gentisic acid, vanilic acid, 1,3-dicaffeoylquinic acid, α- resorcylic acid). We found TR root extract to have antiproliferative activity on glioma cells after 24 h of treatment. TR root extract induces apoptosis on various grades (I-III) of human glioma cells by the generation of reactive oxygen species (ROS) along with concurrent loss of mitochondrial membrane potential, enhanced S and G2/M phases of the cell cycle, and altered mRNA levels of Bax, Bcl-2, p53, Cas-3, Cas-8 and Cas- 9 factors involved in apoptosis. This work for the first time demonstrate that TR extract from L. sibiricus root has the potential to activate apoptosis in grade I-III human glioma cells through the intrinsic and extrinsic pathways. C1 [Sitarek, Przemyslaw] Medical University of Lodz, Department of Biology and Pharmaceutical Botany, Muszynskiego Street 1, 90-151 Lodz, Poland. [Skala, Ewa] Medical University of Lodz, Department of Biology and Pharmaceutical Botany, Muszynskiego Street 1, 90-151 Lodz, Poland. [Toma, Monika] University of Lodz, Department of Molecular GeneticsLodz, Poland. [Wielanek, Marzena] University of Lodz, Faculty of Biology and Environmental Protection, Department of Plant Physiology and BiochemistryLodz, Poland. [Szemraj, Janusz] Medical University of Lodz, Department of Medical BiotechnologyLodz, Poland. [Skorski, Tomasz] Temple University, School of Medicine, Department of Microbiology and Immunology, and Fels Institute for Cancer ResearchPhiladelphia, PA, USA. [Bialas, J Adam] Medical University of Lodz, 1st Chair of Internal Medicine, Department of Pneumology and AllergyLodz, Poland. [Sakowicz, Tomasz] The University of Lodz, Department of Genetics and Plant Molecular Biology and BiotechnologyLodz, Poland. [Kowalczyk, Tomasz] The University of Lodz, Department of Genetics and Plant Molecular Biology and BiotechnologyLodz, Poland. [Radek, Maciej] Medical University of Lodz, University Hospital WAM-CSW, Department of Neurosurgery, Surgery of Spine and Peripheral NervesLodz, Poland. [Wysokinska, Halina] Medical University of Lodz, Department of Biology and Pharmaceutical Botany, Muszynskiego Street 1, 90-151 Lodz, Poland. [Sliwinski, Tomasz] University of Lodz, Department of Molecular GeneticsLodz, Poland. RP Sitarek, P (reprint author), Medical University of Lodz, Department of Biology and Pharmaceutical Botany, 90-151 Lodz, Poland. EM przemyslaw.sitarek@umed.lodz.pl CR Wen PY, Kesari S, 2008, Malignant gliomas in adults. N Engl J Med 359(5):492–507 Stupp R,MasonWP, van den BentMJ,Weller M, Fisher B, Taphoorn MJ et al, 2005, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352(10):987–996 Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC et al, 2009, Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10(5):459–466 Cheng L,Wu Q, Guryanova OA, Huang Z, Huang Q, Rich JN et al, 2011, Elevated invasive potential of glioblastoma stem cells. Biochem Biophys Res Commun 406(4):643–648 Kortmann RD, Jeremic B, Weller M, Plasswilm L, Bamberg M, 2003, Radiochemotherapy of malignant glioma in adults. Clinical experiences. Strahlentherapie und Onkologie 179(4):219– 232 Atmaca H, Bozkurt E, 2016, Apoptotic and anti-angiogenic effects of Salvia triloba extract in prostate cancer cell lines. Tumor Biol 37(3):3639–3364 Peng X, Zhuang DD, Guo QS, 2015, Induction of S phase arrest and apoptosis by ethyl acetate extract from Tetrastigma hemsleyanum in human hepatoma HepG2 cells. Tumor Biol 36(4):2541–2550 Skala E, Sitarek P, Toma M, Szemraj J, Radek M, Nieborowska- Skorska M, Skorski T, Wysokinska H, Sliwinski T, 2016, Inhibition of human glioma cell proliferation by altered Bax/Bcl- 2-p53 expression and apoptosis induction by Rhaponticum Carthamoides extracts from transformed and normal roots. J Pharm Pharmacol 68(11):1454–1464., DOI 10.1111/jphp.12619 Shang X, Pan H, Wang X, He H, Li M, 2014, Leonurus japonicus Houtt.: ethnopharmacology, phytochemistry and pharmacology of an important traditional Chinese medicine. J Ethnopharmacol 152(1):14–32., DOI 10.1016/j.jep.2013.12.052 Chinwala MG, Gao M, Dai J, Shao J, 2003, In vitro anticancer activities of Leonurus Heterophyllus sweet, Chinese motherwort herb). J Altern Complement 9(4):511–518 Sitarek P, Skala E, Toma M, Wielanek M, Szemraj J, Kolasa M, Nieborowska-Skorska M, Skorski T, Wysokinska H, Sliwinski T, 2016a, A preliminary study of apoptosis induction in glioma cells via alteration of the Bax/Bcl-2-p53 axis by transformed and nontransformed root extracts of Leonurus sibiricus L. Tumor Biol 1–12 Schenk RU, Hildebrandt AC, 1972, Medium and techniques for induction of growth of monocotyledonous and dicotyledonous plant cell cultures. Can J Bot 50:199–204 Skala E, Kicel A, Olszewska MA, Kiss AK,Wysokinska H, 2015, Establishment of hairy root cultures of Rhaponticum carthamoides, Willd., Iljin for the production of biomass and caffeic acid derivatives. Biomed Res Int 181098:1–11 Sitarek P, Skala E, Wysokinska H, Wielanek M, Szemraj J, Toma M, Sliwinski T, 2016b, The effect of Leonurus sibiricus plant extracts on stimulating repair and protective activity against oxidative DNA damage in CHO cells and content of phenolic compounds. Oxidative Med Cell Longev 5738193:1–11., DOI 10.1155 /2016/5738193 Kleihues P, Louis DN, ScheithauerBW, Rorke LB, Reifenberger G, Burger PC, CaveneeWK(2002, TheWHO classification of tumors of the nervous system. J Neuropathol Exp Neurol 61:215–225 discussion 226–9 Mosmann T, 1983, Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65:55–63 RogalskaA,Koceva-ChylaA, Jozwiak Z, 2008, Aclarubicin-induced ROS generation and collapse of mitochondrial membrane potential in human cancer cell lines. Chem Biol Interact 176(1):58–70 Qu Y, Zhang J,Wu S, Li B, Liu S, Cheng J, 2012, SIRT1 promotes proliferation and inhibits apoptosis of human malignant glioma cell lines. Neurosci Lett 525(2):168–172 Agrawal M, 2015, Natural polyphenols based new therapeutic avenues for advanced biomedical applications. Drug Metab Rev 47(4):420–430., DOI 10.3109/03602532.2015.1102933 Orfali G, Duarte AC, Bonadio V, Martinez NP, de Araujo ME, Priviero FB, Carvalho PO, Priolli DG, 2016, Review of anticancer mechanisms of isoquercitin. World J Clin Oncol 7(2):189–199., DOI 10.5306/wjco.v7.i2.189 Zhou XJ, Liu M, Yan JJ, Cao Y, Liu P, 2012, Antidepressant-like effect of the extracted of Kai Xin san, a traditional Chinese herbal prescription, is explained by modulation of the central monoaminergic neurotransmitter system in mouse. J Ethnopharmacol 139(2): 422–428 HengartnerMO, 2002, The biochemistry of apoptosis. Nature 407: 770–776 de Kok TM, van Breda SG, Manson MM, 2008, Mechanisms of combined action of different chemopreventive dietary compounds: a review. Eur J Nutr 47(2):51–59 Mertens-Talcott SU, Talcott ST, Percival SS, 2003, Low concentrations of quercetin and ellagic acid synergistically influence proliferation, cytotoxicity and apoptosis in MOLT-4 human leukemia cells. J Nutr 133:2669–2674 Shimizu M, Deguchi A, Lim JT, Moriwaki H, Kopelovich L, Weinstein IB, 2005,, −)-Epi-gallocatechin gallate and polyphenon E inhibit growth and activation of the epidermal growth factor receptor and human epidermal growth factor receptor-2 signaling pathways in human colon cancer cells. Clin Cancer Res 11:2735– 2746 Dong XC, Bin Q, Zhi-Qiang G, Ying JY, 2012, Comparison of burst of reactive oxygen species and activation of caspase-3 in apoptosis of K562 and HL-60 cells induced by docetaxel. Cancer Lett 214:103–113 Ying-Tang H, Yi-Hsuan H, Tzhy-Chyuan H, Bonnie SP, Yeuk CL, Min-Hsiung P, 2006, Apoptosis-inducing active components from Corbicula fluminea through activation of caspase-2 and production of reactive oxygen species in human leukemia HL-60 cells. Food Chem Toxicol 44:1261–1272 Nunes C, Barbosa MRL, Laranjinha J, 2011, Nitric oxide and DOPAC-induced cell death: from GSH. Depletion to mitochondrial energy crisis. Mol Cell Neurosci 48:94–103 Sipos L, Szegedi Z, Fedorcsak I, Afra D, Szende B, 1998, Apoptosis and p53 expression in human gliomas. Pathol Oncol Res 4(4):267–270 Kug CK, Jin SK, Jung KS, In Gyu K, 2007, Enhance induction of mitochondrial damage and apoptosis in leukemia HL-60 cells by the Ganoderma lucidum and Duchesnea chysantha extracts. Cancer Lett 246:210–217 Muhammad K, Chuan D, Azhar R, Fei Y, Ting L, Hongwen G, Rong G, Lili Z, Kun Z, Xuedong F, Tonghui M, 2012, Isoalantolactone induces reactive oxygen species mediated apoptosis in pancreatic carcinoma PANC-1 cells. Int J Biol Sci 8(4):533– 547 Stowe DF, Camara AK, 2009, Mitochondrial reactive oxygen species production in excitable cells: modulators of mitochondrial and cell function. Antioxid Redox Signal 11:1373–1414 Kagan VE, Tyurin VA, Jiang J et al, 2005, Cytochrome c acts as a cardiolipin oxygenase required for release of proapoptotic factors. Nat Chem Biol 1(4):223–232 Juan ME, Wenzel U, Daniel H, Planas JM, 2008, Resveratrol induces apoptosis through ROS-dependent mitochondria pathway in HT-29 human colorectal carcinoma cells. J Agric Food Chem 56(12):4813–4818 Garcia A, Morales P, Arranz N, Delgado ME, Rafter J, Haza AI, 2009, Antiapoptotic effects of dietary antioxidants towards Nnitrosopiperidine and N-nitrosodibutylamine-induced apoptosis in HL-60 and HepG2 cells. J Appl Toxicol 29(5):403–413 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 679 EP 687 DI 10.1007/s12253-016-0170-6 PG 9 ER PT J AU Kuthi, L Jenei, A Hajdu, A Nemeth, I Varga, Z Bajory, Z Pajor, L Ivanyi, B AF Kuthi, Levente Jenei, Alex Hajdu, Adrienn Nemeth, Istvan Varga, Zoltan Bajory, Zoltan Pajor, Laszlo Ivanyi, Bela TI Prognostic Factors for Renal Cell Carcinoma Subtypes Diagnosed According to the 2016 WHO Renal Tumor Classification: a Study Involving 928 Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Renal cell carcinoma; ISUP grading; Microscopic tumor necrosis; Survival rates; Prognostic factors ID Renal cell carcinoma; ISUP grading; Microscopic tumor necrosis; Survival rates; Prognostic factors AB The morphotype and grade of renal cell carcinoma (RCC) in 928 nephrectomies were reclassified according to the 2016 WHO classification in order to analyze the distribution and outcomes of RCC subtypes in Hungary, to assess whether microscopic tumor necrosis is an independent prognostic factor in clear cell RCC, and to study whether a twotiered grading (low/high) for clear cell and papillary RCC provides similar prognostic information to that of the fourtiered ISUP grading system. 83.4% of the cohort were clear cell, 6.9% papillary, 4.5% chromophobe, 2.3% unclassified, 1.1% Xp11 translocation, 1.1% clear cell papillary, 0.3% collecting duct and 0.1% mucinous tubular and spindle cell RCCs. RCC occurred in 16 patients with end-stage kidney disease and none of them displayed features of acquired cystic kidney disease-associated RCC. The 5-year survival rates were as follows: chromophobe 100%, clear cell papillary 100%, clear cell low-grade 96%, papillary type 1 92%, clear cell high-grade 63%, papillary type 2 65%, unclassified 46%, Xp11 translocation 20%, and collecting duct 0%. The 5-year survival rates in low-grade and high-grade papillary RCC were 95% and 59%, respectively. In clear cell RCC, only the grade, the stage and the positive surgical margin proved to be independent prognostic factors statistically. Overall, papillary RCC occurred relatively infrequently; microscopic tumor necrosis in clear cell RCC did not predict the outcome independently of the tumor grading; and the assignment of clear cell and papillary RCCs into low-grade or high-grade tumors was in terms of survival no worse than the ISUP grading. C1 [Kuthi, Levente] University of Szeged, Department of Pathology, Allomas Street 1, H-6725 Szeged, Hungary. [Jenei, Alex] University of Szeged, Department of Pathology, Allomas Street 1, H-6725 Szeged, Hungary. [Hajdu, Adrienn] University of Szeged, Department of Pathology, Allomas Street 1, H-6725 Szeged, Hungary. [Nemeth, Istvan] University of Szeged, Department of Dermatology and Allergology, Koranyi Alley 6, H-6720 Szeged, Hungary. [Varga, Zoltan] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Bajory, Zoltan] University of Szeged, Department of Urology, Kalvaria Lane 57, H-6725 Szeged, Hungary. [Pajor, Laszlo] University of Szeged, Department of Urology, Kalvaria Lane 57, H-6725 Szeged, Hungary. [Ivanyi, Bela] University of Szeged, Department of Pathology, Allomas Street 1, H-6725 Szeged, Hungary. RP Kuthi, L (reprint author), University of Szeged, Department of Pathology, H-6725 Szeged, Hungary. EM kuthi.levente@med.u-szeged.hu CR Moch H, Humphrey PA, Ulbright TM, Reuter VE, 2016, WHO classification Tumours of the urinary system and male genital organs. International Agency for Research on Cancer, Lyon Srigley JR, Delahunt B, Eble JN et al, 2013, The International Society of Urological Pathology, ISUP, Vancouver classification of renal neoplasia. Am J Surg Pathol 37:1469–1489 Delahunt B, Cheville JC, Martignoni G et al, 2013, The International Society of Urological Pathology, ISUP, grading system for renal cell carcinoma and other prognostic parameters. Am J Surg Pathol 37:1490–1504 Reuter VE, Argani P, Zhou M, Delahunt B, Members of the ISUP Immunohistochemistry in Diagnostic Urologic Pathology Group, 2014, Best practices recommendations in the application of immunohistochemistry in the kidney tumors: report from the International Society of Urologic Pathology consensus conference. Am J Surg Pathol 38:e35–e49 Steffens S, Janssen M, Ross FC et al, 2014, The Fuhrman grading system has no prognostic value in patients with nonsarcomatoid chromophobe renal cell carcinoma. Hum Pathol 45:2411–2416 Delahunt B, McKenney JK, Lohse CM, Leibovich BC, Thompson RH, Boorjian SA, Cheville JC, 2013, A novel grading system for clear cell renal cell carcinoma incorporating tumor necrosis. Am J Surg Pathol 37:311–322 Tan PH, Cheng L, Rioux-Leclercq N et al, 2013, Renal tumors: diagnostic and prognostic biomarkers. Am J Surg Pathol 37:1518– 1531 Genega EM, Ghebremichael M, Najarian R et al, 2010, Carbonic anhydrase IX expression in renal neoplasms: correlation with tumor type and grade. Am J Clin Pathol 134:873–879 Gupta R, Billis A, Shah RB et al, 2012, Carcinoma of the collecting ducts of Bellini and renal medullary carcinoma: clinicopathologic analysis of 52 cases of rare aggressive subtypes of renal cell carcinoma with a focus on their interrelationship. Am J Surg Pathol 36: 1265–1278 Williamson SR, Cheng L, 2016, Clear cell renal cell tumors: not all that is "clear" is cancer. Urol Oncol 34:292.e17–292.e22 Sobin LH, Gospodarowicz MK, Wittekind C, 2009, TNM classification of malignant tumors, 7th edn. Wiley-Blackwell, Oxford Tarabeia J, Kaluski DN, BarchanaM, Dichtiar R, GreenMS, 2010, Renal cell cancer in Israel: sex and ethnic differences in incidence and mortality, 1980-2004. Cancer Epidemiol 34:226–231 Lipworth L, Morgans AK, Edwards TL et al, 2016, Renal cell cancer histologic subtype distribution differs by race and sex. BJU Int 117:260–265 Pichler M, Hutterer GC, Chromecki TF, Jesche J, Kampel-Kettner K, Pummer K, Zigeuner R, 2012, Renal cell carcinoma stage migration in a single European Centre over 25 years: effects on 5- and 10-year metastasis-free survival. Int Urol Nephrol 44:997–1004 Steffens S, Junker K, Roos FC et al, 2014, Small renal cell carcinomas–how dangerous are they really? Results of a large multicenter study. Eur J Cancer 50:739–745 Novara G, Ficarra V, Antonelli A et al, 2010, Validation of the 2009 TNMversion in a large multi-institutional cohort of patients treated for renal cell carcinoma: are further improvements needed? Eur Urol 58:588–595 Moch H, Gasser T, Amin MB, Torhorst J, Sauter G, Mihatsch MJ, 2000, Prognostic utility of the recently recommended histologic classification and revised TNM staging system of renal cell carcinoma: a Swiss experience with 588 tumors. Cancer 89:604–614 Rabjerg M, Mikkelsen MN, Walter S, Marcussen N, 2014, Incidental renal neoplasms: is there a need for routine screening? A Danish single-center epidemiological study. APMIS 122:708– 714 Zhou H, Zheng S, Truong LD, Ro JY, Ayala AG, Shen SS, 2014, Clear cell papillary renal cell carcinoma is the fourth most common histologic type of renal cell carcinoma in 290 consecutive nephrectomies for renal cell carcinoma. Hum Pathol 45:59–64 Dhakal HP, McKenney JK, Khor LY, Reynolds JP, Magi-Galluzzi C, Przybycin CG, 2016, Renal neoplasms with overlapping features of clear cell renal cell carcinoma and clear cell papillary renal cell carcinoma: a clinicopathologic study of 37 cases from a single institution. Am J Surg Pathol 40:141–154 Tickoo SK, dePeralta-Venturina MN, Harik LR et al, 2006, Spectrum of epithelial neoplasms in end-stage renal disease: an experience from 66 tumor-bearing kidneys with emphasis on histologic patterns distinct from those in sporadic adult renal neoplasia. Am J Surg Pathol 30:141–153 Vegso G, Toronyi E, Hajdu M et al, 2011, Renal cell carcinoma of the native kidney: a frequent tumor after kidney transplantation with favorable prognosis in case of early diagnosis. Transplant Proc 43: 1261–1264 Breda A, Luccarelli G, Rodriguez-Faba O et al, 2015, Clinical and pathological outcomes of renal cell carcinoma, RCC, in native kidneys of patients with end-stage renal disease: a long-term comparative retrospective study with RCC diagnosed in the general population. World J Urol 33:1–7 Song C, Hong SH, Chung JS, Byun SS, Kwak C, Jeong CW, Seo SI, Jeon HG, Seo IY, 2016, Renal cell carcinoma in end-stage disease: multi-institutional comparative analysis of survival. Int J Urol 23:465–471 Patard JJ, Leray E, Rioux-Leclercq N, Cindolo L et al, 2005, Prognostic value of histologic subtypes in renal cell carcinoma: a multicenter experience. J Clin Oncol 23:2763–2771 Leibovich BC, Lohse CM, Crispen PL, Boorjian SA, Thompson RH, BluteML, Cheville JC, 2010, Histological subtype is an independent predictor of outcome for patients with renal cell carcinoma. J Urol 183:1309–1315 Capitanio U, Cloutier V, Zini L et al, 2009, A critical assessment of the prognostic value of clear cell, papillary and chromophobe histological subtypes in renal cell carcinoma: a population-based study. BJUI 103:1496–1500 Volpe A, Novara G, Antonelli A et al, 2012, Chromophobe renal cell carcinoma, RCC): oncological outcomes and prognostic factors in a large multicentre series. BJU Int 110:76–83 Frees S, Kamal MM, Knoechlein L et al, 2016, Differences in overall and cancer-specific survival of patients presenting with chromophobe versus clear cell renal cell carcinoma: a propensity score matched ana l y s i s . Ur o l ogy. d o i :10. 1016/ j . urology.2016.05.048 Amin MB, Paner GP, Alvarado-Cabrero I, Young AN, Stricker HJ, Lyles RH, Moch H, 2008, Chromophobe renal cell carcinoma: histomorphologic characteristics and evaluation of conventional pathologic prognostic parameters in 145 cases. Am J Surg Pathol 32:1822–1834 Diolombi ML, Cheng L, Argani P, Epstein JI, 2015, Do clear cell papillary renal cell carcinomas have malignant potential? Am J Surg Pathol 39:1621–1634 Cornejo KM, Dong F, Zhou AG et al, 2015, Papillary renal cell carcinoma: correlation of tumor grade and histologic characteristics with clinical outcome. Hum Pathol 46:1411–1417 Warrick JI, Tsodikov A, Kunju LP et al, 2012, Papillary renal cell carcinoma revisited: a comprehensive histomorphologic study with outcome correlations. Hum Pathol 45:1139–1146 PichlerM, Hutterer GC, Chromecki TF, Pummer K, Mannweiler S, Zigeuner R, 2013, Presence and extent of histological tumour necrosis is an adverse prognostic factor in papillary type 1 but not in papillary type 2 renal cell carcinoma. Histopathology 62:219–228 Frank I, Blute ML, Cheville JC, Lohse CM, Weaver AL, Zincke H, 2002, An outcome prediction model for patients with clear cell renal cell carcinoma treated with radical nephrectomy based on tumor stage, size, grade and necrosis: the SSIGN score. J Urol 168:2395–2400 Lam JS, Shvarts O, Said JW et al, 2005, Clinicopathologic and molecular correlations of necrosis in the primary tumor of patients with renal cell carcinoma. Cancer 103:2517–2525 The Cancer Genom Atlas Network, 2016, Comprehensive molecular characterization of papillary renal-cell carcinoma. N Engl J Med 374:135–145 Becker A, Hickmann D, Hansen J et al, 2016, Critical analysis of a simplified Fuhrman grading scheme for prediction of cancer specific mortality in patients with clear cell renal cell carcinoma–impact on prognosis. Eur J Surg Oncol 42:419–425 Pierorazio PM, JohnsonMH, Patel HD et al, 2016, Management of Renal Masses and Localized Renal Cancer: systematic review and meta-analysis. J Urol., DOI 10.1016/j.juro.2016.04.081 Chamie K, Donin NM, Klopfer P et al, 2016, Adjuvant weekly girentuximab following nephrectomy for high-risk renal cell carcinoma: the ARISER randomized clinical trial. JAMA Oncol., DOI 10.1001/jamaoncol.2016.4419 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 689 EP 698 DI 10.1007/s12253-016-0179-x PG 10 ER PT J AU Toth, K Patai, V Kalmar, A Bartak, KB Nagy, BZs Galamb, O Wichmann, B Tulassay, Zs Molnar, B AF Toth, Kinga Patai, V Arpad Kalmar, Alexandra Bartak, Kinga Barbara Nagy, Brigitta Zsofia Galamb, Orsolya Wichmann, Barnabas Tulassay, Zsolt Molnar, Bela TI Circadian Rhythm of Methylated Septin 9, Cell-Free DNA Amount and Tumor Markers in Colorectal Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SEPT9; Colorectal cancer; Tumormarkers; Circadian rhythm; Cell-free DNA ID SEPT9; Colorectal cancer; Tumormarkers; Circadian rhythm; Cell-free DNA AB To determine the level of cell-free DNA (cfDNA), Septin 9 (SEPT9) and tumor markers (CEA, AFP, CA19–9, TPA, CA72–4). Plasma samples were collected four times a day (06:00, 12:00, 18:00, 24:00) from 9 patients with CRC (5 stage I-II, 4 stage III-IV), from one with colorectal adenoma and from one healthy control. CfDNA was isolated, quantified and bisulfite-converted. CfDNA and methylated SEPT9 were determined by RT-PCR. Plasma levels of conventional tumor markers were also measured. The lowest cfDNA concentrations were observed at 24:00 and 18:00 in stage I-III patients. In stage IV samples low cfDNA level (mean 48.2 ng/ml) were observed at several time points (6:00, 12:00, 18:00). The highest cfDNA levels were measured at 6:00 and 12:00 in CRC I-III stages and at 24:00 in stage IV samples (78.65 ng/ ml). Higher in-day differences were found in stage II (43– 48%) than in stage I samples (22%). Interestingly, the highest SEPT9 methylation level was found at 24:00 in most CRC cases, in contrast to the cfDNA levels. At 24:00, all cancer and adenoma cases were positive for SEPT9 methylation. At other time points (6:00, 12:00, 18:00) only 77.7% of CRC samples showed SEPT9 positivity. Stage I samples were SEPT9 positive only at 24:00. CEA and CA19–9 levels displayed correlation with the amount of cfDNA in case of late stage cases. Daytime activity can influence SEPT9 positivity in cases with low concentration of cfDNA. Thus, it may improve screening sensitivity by collecting samples earlier in the morning. C1 [Toth, Kinga] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Patai, V Arpad] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Kalmar, Alexandra] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Bartak, Kinga Barbara] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Nagy, Brigitta Zsofia] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Galamb, Orsolya] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. [Wichmann, Barnabas] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, 1088 Budapest, Hungary. RP Toth, K (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM drtothkinga@yahoo.com CR Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A, 2013, Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 10:472–484., DOI 10.1038/nrclinonc.2013.110 Mandel P, Metais P, 1948, Nucleic acids in human blood plasma. C R Acad Sci Paris 15:241–243 Leon SA, Shapiro B, Sklaroff DM, Yaros MJ, 1977, Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 15:646–650 Stroun M, Anker P, Maurice P, Lyautey J, Lederrey C, BeljanskiM, 1989, Neoplastic characteristics of the DNA found in the plasma of cancer patients. Oncology 15:318–322., DOI 10.1159/000226740 Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R, 2001, DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 15:1659–1665 Brancaccio P, Lippi G, Maffulli N, 2010, Biochemical markers of muscular damage. Clin Chem Lab Med 48:757–767., DOI 10.1515/CCLM.2010.179 Chevion S, Moran DS, Heled Y, Shani Y, Regev G, Abbou B, Berenshtein E, Stadtman ER, Epstein Y, 2003, Plasma antioxidant status and cell injury after severe physical exercise. Proc Natl Acad Sci U S A 100:5119–5123., DOI 10.1073/pnas.0831097100 Fatouros IG, Destouni A, Margonis K, Jamurtas AZ, Vrettou C, Kouretas D, Mastorakos G, Mitrakou A, Taxildaris K, Kanavakis E, Papassotiriou I, 2006, Cell-free plasma DNA as a novel marker of aseptic inflammation severity related to exercise overtraining. Clin Chem 52:1820–1824., DOI 10.1373/clinchem.2006.070417 Brinkmann V, Zychlinsky A, 2007, Beneficial suicide: why neutrophils die to make NETs. Nat Rev Microbiol 5:577–582., DOI 10.1038/nrmicro1710 Tjoa ML, Cindrova-Davies T, Spasic-Boskovic O, Bianchi DW, Burton GJ, 2006, Trophoblastic oxidative stress and the release of cell-free feto-placental DNA. Am J Pathol 169:400–404., DOI 10.2353/ajpath.2006.060161 Maron JL, Bianchi DW, 2007, Prenatal diagnosis using cellfree nucleic acids in maternal body fluids: a decade of progress. Am J Med Genet C Semin Med Genet 145C:5–17., DOI 10.1002 /ajmg.c.30115 Leung TN, Zhang J, Lau TK, Chan LYS, Lo D, 2001, Increased maternal plasma fetal DNA concentrations in women who eventually develop preeclampsia. Clin Chem 47:137–139 VeldersM, Treff G, Machus K, Bosnyak E, Steinacker J, Schumann U, 2013, Exercise is a potent stimulus for enhancing circulating DNase activity. Clin Biochem 47:471–474., DOI 10.1016/j. clinbiochem.2013.12.017 Stroun M, Lyautey J, Lederrey C, Olson-Sand A, Anker P, 2001, About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin Chim Acta 15:139–142., DOI 10.1016/S0009-8981(01)00665-9 Kettner NM, Katchy CA, Fu L, 2014, Circadian gene variants in cancer. Ann Med 46:208–220., DOI 10.3109/07853890.2014.914808 Lee JH, Sancar A, 2011, Regulation of apoptosis by the circadian clock through NF-kappa B signaling. Proc Natl Acad Sci U S A 108:12036–12041., DOI 10.1073/pnas.1108125108 Haus E, Smolensky MH, 1999, Biologic rhythms in the immune sys-tem. Chronobiol Int 16:581–622 Born J, Lange T, Hansen K, Molle M, Fehm HL, 1997, Effects of sleep and circadian rhythm on human circulating immune cells. J Immunol 158:4454–4464 Kawate T, Abo T, Hinuma S, Kumagai K, 1981, Studies of the bioperio-dicity of the immune response. II. Co-variations of murine T and B cells and a role of corticosteroid. J Immunol 126:1364– 1367 Braude S, Beck A, 2013, Complete blood counts with differential: more accurate reference ranges based oncircadian leukocyte trafficking. J Clin Pathol 66:909–910., DOI 10.1136/jclinpath-2013- 201776 Vitaterna MH, King DP, Chang AM, Kornhauser JM, Lowrey PL, McDonald JD, Dove WF, Pinto LH, Turek FW, Takahashi JS, 1994, Muta-genesis andmapping of a mouse gene, clock, essential for circadian behavior. Science 264:719–725 Lee C, Etchegaray JP, Cagampang FR, Loudon AS, Reppert SM, 2001, Posttranslational mechanisms regulate the mammalian circadian clock. Cell 107:855–867 Panda S, Antoch MP, Miller BH, Su AI, Schook AB, Straume M, Schultz PG, Kay SA, Takahashi JS, Hogenesch JB, 2002, Coordinated transcription of key pathways in the mouse by the circadian clock. Cell 109:307–320., DOI 10.1016/S0092-8674, 02)00722-5 Wood PA, Du-Quiton J, You S, Hrushesky WJ, 2006, Circadian clock coordinates cancer cell cycle progression, thymidylate synthase, and 5-fluorouracil therapeutic index. Mol Cancer Ther 5: 2023–2033., DOI 10.1158/1535-7163.MCT-06-0177 Filipski E, King VM, Li X, Granda TG, Mormont MC, Liu X, Claustrat B, Hastings MH, Levi F, 2002, Host circadian clock as a control point in tumor progression. J Natl Cancer Inst 94:690– 697., DOI 10.1093/jnci/94.9.690 Schernhammer ES, Laden F, Speizer FE, Willett WC, Hunter DJ, Kawachi I, Colditz GA, 2001, Rotating night shifts and risk of breast cancer in women participating in the nurses’ health study. J Natl Cancer Inst 93:1563–1568., DOI 10.1093/jnci/93.20.1563 Cash E, Sephton SE, Chagpar AB, Spiegel D, Rebholz WN, Zimmaro LA, Tillie JM, Dhabhar FS, 2015, Circadian disruption and biomarkers of tumor progression in breast cancer patients awaiting surgery. Brain Behav Immun 48:102–114., DOI 10.1016/j. bbi.2015.02.017 Focan C, Focan-Henrard D, Collette J, Mechkouri M, Levi F, Hrushesky W, Touitou Y, Franchimont P, 1986, Cancerassociated alteration of circadian rhythms in carcinoembryonic antigen, CEA, and alpha-fetoprotein, AFP, in humans. Anticancer Res 6:1137–1144 Frederiksen CB, Lomholt AF, Lottenburger T, Davis GJ, Dowell BL, Blankenstein MA, Christensen IJ, Brunner N, Nielsen HJ, 2008, Assessment of the biological variation of plasma tissue inhibitor of metalloproteinases-1. Int J Biol Markers 23:42–47 Grutzmann R, Molnar B, Pilarsky C, Habermann JK, Schlag PM, Saeger HD, Miehlke S, Stolz T, Model F, Roblick UJ, Bruch HP, Koch R, Liebenberg V, Devos T, Song X, Day RH, Sledziewski AZ, Lofton-Day C, 2008, Sensitive detection of colorectal cancer in peripheral blood by septin 9 DNA methylation assay. PLoS One 3:e3759., DOI 10.1371/journal.pone.0003759 deVos T, Tetzner R, Model F, Weiss G, Schuster M, Distler J, Steiger KV, Grutzmann R, Pilarsky C, Habermann JK, Fleshner PR, Oubre BM, Day R, Sledziewski AZ, Lofton-Day C, 2009, Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem 55:1337–1346., DOI 10.1373 /clinchem.2008.115808 Tanzer M, Balluff B, Distler J, Hale K, Leodolter A, Rocken C, Molnar B, Schmid R, Lofton-Day C, Schuster T, Ebert MP, 2010, Performance of epigenetic markers SEPT9 and ALX4 in plasma for detection of colorectal precancerous lesions. PLoS One 5:e9061., DOI 10.1371/journal.pone.0009061 Johnson DA, Barclay RL, Mergener K, Weiss G, Konig T, Beck J, Potter NT, 2014, Plasma Septin9 versus fecal immunochemical testing for colorectal cancer screening: a prospective multicenter study. PLoS One 9:e98238., DOI 10.1371/journal.pone.0098238 Potter NT, Hurban P, White MN, Whitlock KD, Lofton-Day CE, Tetzner R, Koenig T, Quigley NB, Weiss G, 2014, Validation of a real-time PCR-based qualitative assay for the detection of methylated SEPT9 DNA in human plasma. Clin Chem 60:1183–1191., DOI 10.1373/clinchem.2013.221044 Jin P, Kang Q, Wang X, Yang L, Yu Y, Li N, He YQ, Han X, Hang J, Zhang J, Song L, Han Y, Sheng JQ, 2014, Performance of a second generation methylated SEPT9 test in detecting colorectal neoplasm. J Gastroenterol Hepatol 30:830–833., DOI 10.1111/jgh.12855 Toth K, Sipos F, Kalmar A, Patai AV, Wichmann B, Stoehr R, Golcher H, Schellerer V, Tulassay Z, Molnar B, 2012, Detection of methylated SEPT9 in plasma is a reliable screening method for both left- and right-sided colon cancers. PLoS One 7:e46000., DOI 10.1371/journal.pone.0046000 Church TR, Wandell M, Lofton-Day C, Mongin SJ, Burger M, Payne SR, Castanos-Velez E, Blumenstein BA, Rosch T, Osborn N, Snover D, Day RW, Ransohoff DF, PRESEPT Clinical Study Steering Committee, Investigators and Study Team, 2014, Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut 63:317–325., DOI 10.1136/gutjnl-2012-304149 Toth K, Wasserkort R, Sipos F, Kalmar A, Wichmann B, Leiszter K, Valcz G, Juhasz M, Miheller P, Patai AV, Tulassay Z, Molnar B, 2014, Detection of methylated septin 9 in tissue and plasma of colorectal patients with neoplasia and the relationship to the amount of circulating cell-free DNA. PLoS One 9:e115415., DOI 10.1371 /journal.pone.0115415 VeldersM, Treff G, Machus K, Bosnyak E, Steinacker J, Schumann U, 2014, Exercise is a potent stimulus for enhancing circulating DNase activity. Clin Biochem 47:471–474., DOI 10.1016/j. clinbiochem.2013.12.017 Karantanos T, Theodoropoulos G, Pektasides D, Gazouli M, 2014, Clock genes: their role in colorectal cancer. World J Gastroenterol 20:1986–1992., DOI 10.3748/wjg.v20.i8.1986 Mazzoccoli G, Panza A, Valvano MR, Palumbo O, Carella M, Pazienza V, Biscaglia G, Tavano F, Di Sebastiano P, Andriulli A, Piepoli A, 2011, Clock gene expression levels and relationship with clinical and pathological features in colorectal cancer patients. Chronobiol Int 28:841–851., DOI 10.3109/07420528.2011.615182 Warren JD, Xiong W, Bunker AM, Vaughn CP, Furtado LV, Roberts WL, Fang JC, Samowitz WS, Heichman KA, 2011, Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer. BMC Med 9:133., DOI 10.1186/1741-7015-9-133 Ali T, Choe J, Awab A,Wagener TL, Orr WC, 2013, Sleep, immunity and inflammation in gastrointestinal disorders. World J Gastroenterol 19:9231–9239., DOI 10.3748/wjg.v19.i48.9231 Zubelewicz-Szkodzinska B, Muc-Wierzgon M, Wierzgon J, Brodziak A, 2001, Dynamics of circadian fluctuations in serum concentration of cortisol and TNF-alpha soluble receptors in gastrointestinal cancer patients. Oncol Rep 8:207–212., DOI 10.3892/or.8.1.207 Rich T, Innominato PF, Boerner J, Mormont MC, Iacobelli S, Baron B, Jasmin C, Levi F, 2005, Elevated serum cytokines correlated with altered behavior, serum cortisol rhythm, and dampened 24-hour rest-activity patterns in patients with metastatic colorectal cancer. Clin Cancer Res 11:1757–1764., DOI 10.1158/1078-0432.CCR-04-2000 Halberg F, Haus E, Lakatua DJ, Antinozzi R, Cornelissen G, 1995, Cancer marker assessment: case report on salivary and urinary CEA. In Vivo 9:311–314 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2017 VL 23 IS 3 BP 699 EP 706 DI 10.1007/s12253-016-0174-2 PG 8 ER PT J AU Masood, N Basharat, Z Khan, T Yasmin, A AF Masood, Nosheen Basharat, Zarrin Khan, Tabeer Yasmin, Azra TI Entangling Relation of Micro RNA-let7, miRNA-200 and miRNA-125 with Various Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE microRNAs; Cancer; miRNA-let7; miRNA-200; miRNA-125 ID microRNAs; Cancer; miRNA-let7; miRNA-200; miRNA-125 AB Involvement of micro RNAs (miRNA) is currently the focus for cancer studies as they effect the post transcriptional expression of different genes. Let-7 family is among the firstly discovered miRNAs that play important role in cell proliferation and dysregulation leading to cell based diseases including cancer. Another family, miRNA-200 prevents transformation of cell to malignant form and tumor formation by interacting with epidermal mesenchymal transition (EMT). Similarly miRNA-125 controls apoptosis and proliferation by affecting multiple genes involved in transcription, immunological defense, resistance against viral and bacterial infections that ultimately leads to cell proliferation, metastasis and finally cancer. All of these micro RNAs are known to be either upregulated or downregulated in various cancers. Current review is focused to elaborate the role of these three families of micro RNAs on different genes that ultimately cause cancer. In conclusion we can say that the miRNAs discussed here are mostly downregulated in various cancers with some exceptions when upregulation of miRNA-125 may be attributed to cancer formation. C1 [Masood, Nosheen] Fatima Jinnah Women University, The MallRawalpindi, Pakistan. [Basharat, Zarrin] Fatima Jinnah Women University, The MallRawalpindi, Pakistan. [Khan, Tabeer] Fatima Jinnah Women University, The MallRawalpindi, Pakistan. [Yasmin, Azra] Fatima Jinnah Women University, The MallRawalpindi, Pakistan. RP Masood, N (reprint author), Fatima Jinnah Women University, The Mall, Rawalpindi, Pakistan. EM nosheenmasood@hotmail.com CR Lee RC, Feinbaum RL, Ambros V, 1993, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75(5):843–854 Calin GA, Croce CM, 2006, MicroRNA signatures in human cancers. Nat Rev Cancer 6(11):857–866., DOI 10.1038/nrc1997 Iliopoulos D, Hirsch HA, Struhl K, 2009, An epigenetic switch involving NF-kappaB, Lin28, let-7 MicroRNA, and IL6 links inflammation to cell transformation. Cell 139(4):693–706., DOI 10.1016/j.cell.2009.10.014 Lee YS, Dutta A, 2007, The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. Genes Dev 21(9):1025–1030., DOI 10.1101/gad.1540407 Lee H, Han S, Kwon CS, Lee D, 2016, Biogenesis and regulation of the let-7 miRNAs and their functional implications. Protein & cell 7(2):100–113., DOI 10.1007/s13238-015-0212-y KumarMS, Erkeland SJ, Pester RE, ChenCY, EbertMS, Sharp PA, Jacks T, 2008, Suppression of non-small cell lung tumor development by the let-7 microRNA family. Proc Natl Acad Sci U S A 105(10):3903–3908., DOI 10.1073/pnas.0712321105 Brueckner B, Stresemann C, Kuner R, Mund C, Musch T, Meister M, Sultmann H, Lyko F, 2007, The human let-7a-3 locus contains an epigenetically regulated microRNA gene with oncogenic function. Cancer Res 67(4):1419–1423., DOI 10.1158/0008-5472.CAN- 06-4074 Shell S, Park SM, Radjabi AR, Schickel R, Kistner EO, Jewell DA, Feig C, Lengyel E, Peter ME, 2007, Let-7 expression defines two differentiation stages of cancer. Proc Natl Acad Sci U S A 104(27): 11400–11405., DOI 10.1073/pnas.0704372104 Barh D, Malhotra R, Ravi B, Sindhurani P, 2010)MicroRNA let-7: an emerging next-generation cancer therapeutic. Curr Oncol 17(1): 70–80 Yu F, Yao H, Zhu P, Zhang X, Pan Q, Gong C, Huang Y, Hu X, Su F, Lieberman J, Song E, 2007, Let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell 131(6):1109–1123., DOI 10.1016/j.cell.2007.10.054 Ariazi EA, Brailoiu E, Yerrum S, Shupp HA, Slifker MJ, Cunliffe HE, Black MA, Donato AL, Arterburn JB, Oprea TI, Prossnitz ER, Dun NJ, Jordan VC, 2010, The G protein-coupled receptor GPR30 inhibits proliferation of estrogen receptor-positive breast cancer cells. Cancer Res 70(3):1184–1194., DOI 10.1158/0008-5472. CAN-09-3068 Piskounova E, Polytarchou C, Thornton JE, LaPierre RJ, Pothoulakis C, Hagan JP, Iliopoulos D, Gregory RI, 2011, Lin28A and Lin28B inhibit let-7 microRNA biogenesis by distinct mechanisms. Cell 147(5):1066–1079. doi :10.1016/ j . cell.2011.10.039 Yun J, Frankenberger CA, KuoWL, BoelensMC, EvesEM, Cheng N, Liang H, Li WH, Ishwaran H, Minn AJ, Rosner MR, 2011, Signalling pathway for RKIP and let-7 regulates and predicts metastatic breast cancer. EMBO J 30(21):4500–4514., DOI 10.1038 /emboj.2011.312 Dangi-Garimella S, Yun J, Eves EM, Newman M, Erkeland SJ, Hammond SM, Minn AJ, Rosner MR, 2009, Raf kinase inhibitory protein suppresses a metastasis signalling cascade involving LIN28 and let-7. EMBO J 28(4):347–358., DOI 10.1038/emboj.2008.294 Uhlmann S, Zhang JD, Schwager A, Mannsperger H, Riazalhosseini Y, Burmester S, Ward A, Korf U, Wiemann S, Sahin O, 2010, miR-200bc/429 cluster targets PLCgamma1 and differentially regulates proliferation and EGF-driven invasion than miR-200a/141 in breast cancer. Oncogene 29(30):4297–4306., DOI 10.1038/onc.2010.201 ZhuW,Xu H, Zhu D, Zhi H,Wang T,Wang J, Jiang B, ShuY, Liu P, 2012, miR-200bc/429 cluster modulates multidrug resistance of human cancer cell lines by targeting BCL2 and XIAP. Cancer Chemother Pharmacol 69(3):723–731., DOI 10.1007/s00280-011- 1752-3 Wang G, Chan ES, Kwan BC, Li PK, Yip SK, Szeto CC, Ng CF, 2012, Expression of microRNAs in the urine of patients with bladder cancer. Clinical genitourinary cancer 10(2):106–113., DOI 10.1016/j.clgc.2012.01.001 Wiklund ED, Bramsen JB, Hulf T, Dyrskjot L, Ramanathan R, Hansen TB, Villadsen SB, Gao S, Ostenfeld MS, Borre M, Peter ME, Orntoft TF, Kjems J, Clark SJ, 2011, Coordinated epigenetic repression of the miR-200 family and miR-205 in invasive bladder cancer. Int J Cancer 128(6):1327–1334., DOI 10.1002/ijc.25461 Baffa R, Fassan M, Volinia S, O'Hara B, Liu CG, Palazzo JP, Gardiman M, Rugge M, Gomella LG, Croce CM, Rosenberg A, 2009, MicroRNA expression profiling of human metastatic cancers identifies cancer gene targets. J Pathol 219(2):214–221., DOI 10.1002/path.2586 Kohler CU, Bryk O, Meier S, Lang K, Rozynek P, Bruning T, Kafferlein HU, 2013, Analyses in human urothelial cells identify methylation of miR-152, miR-200b and miR-10a genes as candidate bladder cancer biomarkers. Biochem Biophys Res Commun 438(1):48–53., DOI 10.1016/j.bbrc.2013.07.021 Radisky DC, 2011, miR-200c at the nexus of epithelialmesenchymal transition, resistance to apoptosis, and the breast cancer stem cell phenotype. Breast cancer research : BCR 13(3): 110., DOI 10.1186/bcr2885 Teng Y, Mei Y, Hawthorn L, Cowell JK, 2014, WASF3 regulates miR-200 inactivation by ZEB1 through suppression of KISS1 leading to increased invasiveness in breast cancer cells. Oncogene 33(2):203–211., DOI 10.1038/onc.2012.565 Li X, Roslan S, Johnstone CN, Wright JA, Bracken CP, Anderson M, Bert AG, Selth LA, Anderson RL, Goodall GJ, Gregory PA, Khew-Goodall Y, 2014)MiR-200 can repress breast cancer metastasis through ZEB1-independent but moesin-dependent pathways. Oncogene 33(31):4077–4088., DOI 10.1038/onc.2013.370 Madhavan D, Zucknick M, Wallwiener M, Cuk K, Modugno C, Scharpff M, Schott S, Heil J, Turchinovich A, Yang R, Benner A, Riethdorf S, Trumpp A, Sohn C, Pantel K, Schneeweiss A, Burwinkel B, 2012, Circulating miRNAs as surrogate markers for circulating tumor cells and prognostic markers in metastatic breast cancer. Clinical cancer research : an official journal of the American Association for Cancer Research 18(21):5972–5982., DOI 10.1158 /1078-0432.CCR-12-1407 Chen Y, Sun Y, Chen L, Xu X, Zhang X, Wang B, Min L, Liu W, 2013, miRNA-200c increases the sensitivity of breast cancer cells to doxorubicin through the suppression of E-cadherin-mediated PTEN/Akt signaling. Mol Med Rep 7(5):1579–1584., DOI 10.3892 /mmr.2013.1403 Hu X, Schwarz JK, Lewis JS Jr, Huettner PC, Rader JS, Deasy JO, Grigsby PW,Wang X, 2010, AmicroRNA expression signature for cervical cancer prognosis. Cancer Res 70(4):1441–1448., DOI 10.1158/0008-5472.CAN-09-3289 Pedroza-Torres A, Lopez-Urrutia E, Garcia-Castillo V, Jacobo- Herrera N, Herrera LA, Peralta-Zaragoza O, Lopez-Camarillo C, De Leon DC, Fernandez-Retana J, Cerna-Cortes JF, Perez- Plasencia C, 2014, MicroRNAs in cervical cancer: evidences for a miRNA profile deregulated by HPV and its impact on radio-res i s tance. Molecules 19(5):6263–6281. doi :10.3390 /molecules19056263 Torres A, Torres K, Pesci A, Ceccaroni M, Paszkowski T, Cassandrini P, Zamboni G, Maciejewski R, 2013, Diagnostic and prognostic significance of miRNA signatures in tissues and plasma of endometrioid endometrial carcinoma patients. Int J Cancer 132(7):1633–1645., DOI 10.1002/ijc.27840 Diaz-Martin J, Diaz-Lopez A, Moreno-Bueno G, Castilla MA, Rosa-Rosa JM, Cano A, Palacios J, 2014, A core microRNA signature associated with inducers of the epithelial-to-mesenchymal transition. J Pathol 232(3):319–329., DOI 10.1002/path.4289 Toiyama Y, Hur K, Tanaka K, Inoue Y, Kusunoki M, Boland CR, Goel A, 2014, Serum miR-200c is a novel prognostic and metastasis-predictive biomarker in patients with colorectal cancer. Ann Surg 259(4):735–743., DOI 10.1097/SLA.0b013e3182a6909d Tu HF, Lin SC, Chang KW, 2013, MicroRNA aberrances in head and neck cancer: pathogenetic and clinical significance. Current opinion in otolaryngology & head and neck surgery 21(2):104– 111., DOI 10.1097/MOO.0b013e32835e1d6e Tamagawa S, Beder LB, Hotomi M, Gunduz M, Yata K, Grenman R, Yamanaka N, 2014, Role of miR-200c/miR-141 in the regulation of epithelial-mesenchymal transition and migration in head and neck squamous cell carcinoma. Int J Mol Med 33(4):879–886., DOI 10.3892/ijmm.2014.1625 Tellez CS, JuriDE,Do K, Bernauer AM, Thomas CL, Damiani LA, Tessema M, Leng S, Belinsky SA, 2011, EMT and stem cell-like properties associated with miR-205 and miR-200 epigenetic silencing are early manifestations during carcinogen-induced transformation of human lung epithelial cells. Cancer Res 71(8):3087–3097., DOI 10.1158/0008-5472.CAN-10-3035 Kundu ST, Byers LA, Peng DH, Roybal JD, Diao L,Wang J, Tong P, Creighton CJ, Gibbons DL, 2016, The miR-200 family and the miR-183 ~ 96 ~ 182 cluster target Foxf2 to inhibit invasion and metastasis in lung cancers. Oncogene 35(2):173–186., DOI 10.1038 /onc.2015.71 Feng B,Wang R, Chen LB, 2012, Review of miR-200b and cancer chemosensitivity. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 66(6):397–402. doi :10.1016/j. biopha.2012.06.002 Suryawanshi S, Vlad AM, Lin HM, Mantia-Smaldone G, Laskey R, Lee M, Lin Y, Donnellan N, Klein-Patel M, Lee T, Mansuria S, Elishaev E, Budiu R, Edwards RP, Huang X, 2013, Plasma microRNAs as novel biomarkers for endometriosis and endometriosis-associated ovarian cancer. Clinical cancer research : an official journal of the American Association for Cancer Research 19(5):1213–1224., DOI 10.1158/1078-0432.CCR-12-2726 Jabbari N, Reavis AN, McDonald JF, 2014, Sequence variation among members of the miR-200 microRNA family is correlated with variation in the ability to induce hallmarks of mesenchymalepithelial transition in ovarian cancer cells. Journal of ovarian research 7:12., DOI 10.1186/1757-2215-7-12 Watahiki A,Wang Y, Morris J, Dennis K, O'DwyerHM, Gleave M, Gout PW, Wang Y, 2011, MicroRNAs associated with metastatic prostate cancer. PLoS One 6(9):e24950., DOI 10.1371/journal. pone.0024950 Chen X, Wang X, Ruan A, Han W, Zhao Y, Lu X, Xiao P, Shi H, Wang R, Chen L, Chen S, Du Q, Yang H, ZhangX, 2014, miR-141 is a key regulator of renal cell carcinoma proliferation and metastasis by controlling EphA2 expression. Clinical cancer research : an official journal of the American Association for Cancer Research 20(10):2617–2630., DOI 10.1158/1078-0432.CCR-13-3224 Rodriguez A, Griffiths-Jones S, Ashurst JL, Bradley A, 2004, Identification of mammalian microRNA host genes and transcription units. Genome Res 14(10A):1902–1910., DOI 10.1101 /gr.2722704 Jiang L, Huang Q, Chang J, Wang E, Qiu X, 2011, MicroRNA HSA-miR-125a-5p induces apoptosis by activating p53 in lung cancer cells. Exp Lung Res 37(7):387–398., DOI 10.3109 /01902148.2010.492068 Yanokura M, Banno K, Iida M, Irie H, Umene K, Masuda K, Kobayashi Y, Tominaga E, Aoki D, 2015, MicroRNAS in endometrial cancer: recent advances and potential clinical applications. EXCLI J 14:190–198., DOI 10.17179/excli2014-590 Shang C, Lu YM, Meng LR, 2012, MicroRNA-125b down-regulation mediates endometrial cancer invasion by targeting ERBB2. Medical science monitor : international medical journal of experimental and clinical research 18(4):BR149–BR155 Nishida N, Yokobori T, Mimori K, Sudo T, Tanaka F, Shibata K, Ishii H, Doki Y, Kuwano H, Mori M, 2011, MicroRNA miR-125b is a prognostic marker in human colorectal cancer. Int J Oncol 38(5):1437–1443., DOI 10.3892/ijo.2011.969 Li W, Duan R, Kooy F, Sherman SL, Zhou W, Jin P, 2009, Germline mutation of microRNA-125a is associated with breast cancer. J Med Genet 46(5):358–360. doi :10. 1136 /jmg.2008.063123 Mitra S, Mukherjee N, Das S, Das P, Panda CK, Chakrabarti J, 2014, Anomalous altered expressions of downstream genetargets in TP53-miRNA pathways in head and neck cancer. Scientific reports 4:6280., DOI 10.1038/srep06280 Nakanishi H, Taccioli C, Palatini J, Fernandez-Cymering C, Cui R, Kim T, Volinia S, Croce CM, 2014, Loss of miR-125b-1 contributes to head and neck cancer development by dysregulating TACSTD2 and MAPK pathway. Oncogene 33(6):702–712., DOI 10.1038/onc.2013.13 Sun YM, Lin KY, Chen YQ, 2013, Diverse functions of miR-125 family in different cell contexts. J Hematol Oncol 6:6., DOI 10.1186 /1756-8722-6-6 Amir S, Ma AH, Shi XB, Xue L, Kung HJ, Devere White RW, 2013, Oncomir miR-125b suppresses p14(ARF, to modulate p53-dependent and p53-independent apoptosis in prostate cancer. PLoS One 8(4):e61064., DOI 10.1371/journal.pone.0061064 Jiang L, Huang Q, Zhang S, Zhang Q, Chang J, Qiu X, Wang E, 2010, Hsa-miR-125a-3p and hsa-miR-125a-5p are downregulated in non-small cell lung cancer and have inverse effects on invasion and migration of lung cancer cells. BMC Cancer 10:318., DOI 10.1186/1471-2407-10-318 Hsieh TH, Hsu CY, Tsai CF, Long CY, Wu CH, Wu DC, Lee JN, Chang WC, Tsai EM, 2015, HDAC inhibitors target HDAC5, upregulate microRNA-125a-5p, and induce apoptosis in breast cancer cells. Molecular therapy : the journal of the American Society of Gene Therapy 23(4):656–666., DOI 10.1038/mt.2014.247 Gonzalez-VallinasM, Breuhahn K, 2016, MicroRNAs are key regulators of hepatocellular carcinoma, HCC, cell dissemination-what we learned from microRNA-494. Hepatobiliary surgery and nutrition 5(4):372–376., DOI 10.21037/hbsn.2016.05.07 Guo X, Wu Y, Hartley RS, 2009, MicroRNA-125a represses cell growth by targeting HuR in breast cancer. RNA Biol 6(5):575–583 Wang S, Huang J, Lyu H, Lee CK, Tan J, Wang J, Liu B, 2013, Functional cooperation of miR-125a, miR-125b, and miR-205 in entinostat-induced downregulation of erbB2/erbB3 and apoptosis in breast cancer cells. Cell Death Dis 4:e556., DOI 10.1038 /cddis.2013.79 Huang L, Luo J, Cai Q, Pan Q, Zeng H, Guo Z, DongW, Huang J, Lin T, 2011)MicroRNA-125b suppresses the development of bladder cancer by targeting E2F3. Int J Cancer 128(8):1758–1769., DOI 10.1002/ijc.25509 Ichimi T, Enokida H, Okuno Y, Kunimoto R, Chiyomaru T, Kawamoto K, Kawahara K, Toki K, Kawakami K, Nishiyama K, Tsujimoto G, Nakagawa M, Seki N, 2009, Identification of novel microRNA targets based on microRNA signatures in bladder cancer. Int J Cancer 125(2):345–352., DOI 10.1002/ijc.24390 Banzhaf-Strathmann J, Edbauer D, 2014, Good guy or bad guy: the opposing roles of microRNA 125b in cancer. Cell communication and signaling : CCS 12:30., DOI 10.1186/1478-811X-12-30 Wang H, 2016, PredictingMicroRNA biomarkers for cancer using phylogenetic tree and microarray analysis. Int J Mol Sci 17(5)., DOI 10.3390/ijms17050773 Osanto S, Qin Y, Buermans HP, Berkers J, Lerut E, Goeman JJ, van Poppel H, 2012, Genome-wide microRNA expression analysis of clear cell renal cell carcinoma by next generation deep sequencing. PLoS One 7(6):e38298., DOI 10.1371/journal.pone.0038298 Merritt WM, Lin YG, Han LY, Kamat AA, Spannuth WA, Schmandt R, Urbauer D, Pennacchio LA, Cheng JF, Nick AM, Deavers MT, Mourad-Zeidan A, Wang H, Mueller P, Lenburg ME, Gray JW, Mok S, Birrer MJ, Lopez-Berestein G, Coleman RL, Bar-Eli M, Sood AK, 2008, Dicer, Drosha, and outcomes in patients with ovarian cancer. N Engl J Med 359(25):2641–2650., DOI 10.1056/NEJMoa0803785 Takebe T, EnomuraM, Yoshizawa E, Kimura M, Koike H, Ueno Y, Matsuzaki T, Yamazaki T, Toyohara T, Osafune K, Nakauchi H, Yoshikawa HY, Taniguchi H, 2015, Vascularized and complex organ buds from diverse tissues via mesenchymal cell-driven condensation. Cell Stem Cell 16(5):556–565., DOI 10.1016/j. stem.2015.03.004 Feng X, Wang Z, Fillmore R, Xi Y, 2014, MiR-200, a new star miRNA in human cancer. Cancer Lett 344(2):166–173., DOI 10.1016/j.canlet.2013.11.004 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 707 EP 715 DI 10.1007/s12253-016-0184-0 PG 9 ER PT J AU Dundr, P Nemejcova, K Laco, J Skalova, H Bauerova, L Matej, R Fischerova, D AF Dundr, Pavel Nemejcova, Kristyna Laco, Jan Skalova, Helena Bauerova, Lenka Matej, Radoslav Fischerova, Daniela TI Anastomosing Hemangioma of the Ovary: A Clinicopathological Study of Six Cases with Stromal Luteinization SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Anastomosing hemagioma; Capillary hemangioma; Ovary; Stromal luteinization; Urogenital tract ID Anastomosing hemagioma; Capillary hemangioma; Ovary; Stromal luteinization; Urogenital tract AB We report six cases of anastomosing hemangioma of the ovary. All lesions were unilateral and arose in 43 to 81 year old females. In all but one patient, the tumor was asymptomatic and represented incidental finding. The exception was a tumor associated with massive ascites and elevated CA 125. The tumors were, on cut section, spongy and dark violet in color. The size of tumors ranged from 0.5 to 3.5 cm. All lesions showed the same histological features and consisted of capillary sized anastomosing vessels with sinusoid-like pattern intermingled with sporadic medium sized vessels. Interestingly, in all cases there were areas of luteinized cells at the tumor periphery, which ranged from rare small nests to multiple and commonly confluent areas. In one tumor, components of mature adipose tissue were present. Immunohistochemically, all tumors were CD31 and CD34 positive. Other markers examined were negative, including; estrogen receptor, progesterone receptor, androgen receptor, and D2–40. Proliferative activity (Ki-67 index) was very low in all cases. Anastomosing hemangioma is a rare entity, only 8 lesions occurring in ovary has been described from its initial description in 2009.We report six additional cases with their clinicopathological correlation. C1 [Dundr, Pavel] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Nemejcova, Kristyna] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Laco, Jan] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, The Fingerland Department of PathologyHradec Kralove, Czech Republic. [Skalova, Helena] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Bauerova, Lenka] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Matej, Radoslav] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Fischerova, Daniela] Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Obstetrics and GynecologyPrague, Czech Republic. RP Dundr, P (reprint author), Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, 12800 Prague, Czech Republic. EM pdundr@seznam.cz CR Montgomery E, Epstein JI, 2009, Anastomosing hemangioma of the genitourinary tract: a lesion mimicking angiosarcoma. Am J Surg Pathol 33(9):1364–1369., DOI 10.1097/PAS.0b013e3181ad30a7 Kryvenko ON, Gupta NS, Meier FA, Lee MW, Epstein JI, 2011, Anastomosing hemangioma of the genitourinary system: eight cases in the kidney and ovary with immunohistochemical and ultrastructural analysis. Am J Clin Pathol 136(3):450–457., DOI 10.1309/AJCPJPW34QCQYTMT Omiyale AO, 2015, Anastomosing hemangioma of the kidney: a literature review of a rare morphological variant of hemangioma. Ann Transl Med 3(11):151., DOI 10.3978/j.issn.2305- 5839.2015.06.16 Tao LL, Dai Y, Yin W, Chen J, 2014, A case report of a renal anastomosing hemangioma and a literature review: an unusual variant histologically mimicking angiosarcoma. Diagn Pathol 9:159., DOI 10.1186/s13000-014-0159-y John I, Folpe AL, 2016, Anastomosing hemangiomas arising in unusual locations: a Clinicopathologic study of 17 soft tissue cases showing a predilection for the Paraspinal region. Am J Surg Pathol., DOI 10.1097/PAS.0000000000000627 Lin J, Bigge J, Ulbright TM,Montgomery E, 2013, Anastomosing hemangioma of the liver and gastrointestinal tract: an unusual variant histologically mimicking angiosarcoma. Am J Surg Pathol 37(11):1761–1765., DOI 10.1097/PAS.0b013e3182967e6c O'Neill AC, Craig JW, Silverman SG, Alencar RO, 2016, Anastomosing hemangiomas: locations of occurrence, imaging features, and diagnosis with percutaneous biopsy. Abdom Radiol, NY)., DOI 10.1007/s00261-016-0690-2 Metodiev D, Ivanova V, Omainikova B, Boshnakova T, 2015, Ovarian anastomosing hemangioma with stromal luteinization: a case report. Akush Ginekol, Sofiia, 54(9):58–61 Akbulut M, Bir F, Colakoglu N, Soysal ME, Duzcan SE, 2008, Ovarian hemangioma occurring synchronously with serous papillary carcinoma of the ovary and benign endometrial polyp. Ann Saudi Med 28(2):128–131 Anand MS, Shetty S, Mysorekar VV, Kumar RV, 2012, Ovarian hemangioma with stromal luteinization and HCG-producing mononucleate andmultinucleate cells of uncertain histogenesis: a rare coexistence with therapeutic dilemma. Indian J Pathol Microbiol 55(4):509–512., DOI 10.4103/0377-4929.107793 Comunoglu C, Atasoy L, Baykal C, 2010, Ovarian hemangioma occurring synchronously with contralateral mature cystic teratoma in an 81-year-old patient. Ups J Med Sci 115(4):297–299., DOI 10.3109/03009734.2010.502602 Correa-Rivas MS, Colon-Gonzalez G, Lugo-Vicente H, 2003, Cavernous hemangioma presenting as a right adnexal mass in a child. P R Health Sci J 22(3):311–313 Erdemoglu E, Kamaci M, Ozen S, Sahin HG, Kolusari A, 2006, Ovarian hemangioma with elevated CA125 and ascites mimicking ovarian cancer. Eur J Gynaecol Oncol 27(2):195–196 Gehrig PA, Fowler WC Jr, Lininger RA, 2000, Ovarian capillary hemangioma presenting as an adnexal mass with massive ascites and elevated CA-125. Gynecol Oncol 76(1):130–132., DOI 10.1006 /gyno.1999.5648 Grant JW, Millward-Sadler GH, 1986, Haemangioma of the ovary with associated endometrial hyperplasia. Case report Br J Obstet Gynaecol 93(11):1166–1168 Gucer F, Ozyilmaz F, Balkanli-Kaplan P, Mulayim N, Aydin O, 2004, Ovarian hemangioma presenting with hyperandrogenism and endometrial cancer: a case report. Gynecol Oncol 94(3):821– 824., DOI 10.1016/j.ygyno.2004.06.021 Gunes HA, Egilmez R,DulgerM(1990, Ovarian haemangioma. Br J Clin Pract 44(12):734–735 Gupta R, Singh S, Nigam S, Khurana N, 2006, Benign vascular tumors of female genital tract. Int J Gynecol Cancer 16(3):1195– 1200., DOI 10.1111/j.1525-1438.2006.00523.x Jurkovic I, Dudrikova K, Boor A, 1999, Ovarian hemangioma. Cesk Patol 35(4):133–135 Kaneta Y, Nishino R, Asaoka K, Toyoshima K, Ito K, Kitai H, 2003, Ovarian hemangioma presenting as pseudo-Meigs' syndrome with elevated CA125. J Obstet Gynaecol Res 29(3):132–135 Kim SS, Han SE, Lee NK, Choi KU, Joo JK, Suh DS, Kim HY, Kim KH, 2015, Ovarian cavernous hemangioma presenting as a large growing mass in a postmenopausal woman: a case report and review of the literature. J Menopausal Med 21(3):155–159., DOI 10.6118/jmm.2015.21.3.155 Koh LW, Sun YL, Koh PH, Chiu HY, Chen SY, Huang MH, 2007, Ovarian capillary hemangioma presenting as pseudo-Meigs' syndrome: a case report. J Minim Invasive Gynecol 14(3):367–369., DOI 10.1016/j.jmig.2006.10.030 Loverro G, Cormio G, Perlino E, Vicino M, Cazzolla A, Selvaggi L, 1998, Transforming growth factor-beta1 in hemangioma of the ovary. Gynecol Obstet Investig 46(3):210–213 McBurney RP, Trumbull M, 1955, Hemangioma of the ovary with ascites. Miss Doct 32(10):271–274 Miliaras D, Papaemmanouil S, Blatzas G, 2001, Ovarian capillary hemangioma and stromal luteinization: a case study with hormonal receptor evaluation. Eur J Gynaecol Oncol 22(5):369–371 Mirilas P, Georgiou G, Zevgolis G, 1999, Ovarian cavernous hemangioma in an 8-year-old girl. Eur J Pediatr Surg 9(2):116–118., DOI 10.1055/s-2008-1072225 Mitra B, Sengupta S, Rai A,Mehta J, Quader AR, Roy S, Borges A, 2013, Ovarian haemangioma: A rare case report. Int J Surg Case Rep 4(11):981–984., DOI 10.1016/j.ijscr.2013.07.029 M'Pemba Loufoua-Lemay AB, Peko JF, Mbongo JA, Mokoko JC, Nzingoula S, 2003, Ovarian torsion revealing an ovarian cavernous hemangioma in a child. Arch Pediatr 10(11):986–988 Ozana M, Formanek P, Sula F, Silhan J, 1994, Ovarian hemangioma with torsion. Ceska Gynekol 59(1):18–20 Pethe VV, Chitale SV, Godbole RN, Bidaye SV, 1991, Hemangioma of the ovary–a case report and review of literature. Indian J Pathol Microbiol 34(4):290–292 Rivasi F, Philippe E, Walter P, de Marco L, Ludwig L, 1996, Ovarian angioma. Report of 3 asymptomatic cases. Ann Pathol 16(6):439–441 Savargaonkar PR, Wells S, Graham I, Buckley CH, 1994, Ovarian haemangiomas and stromal luteinization. Histopathology 25(2): 185–188 Schoolmeester JK, Greipp PT, Keeney GL, Soslow RA, 2015, Ovarian hemangiomas do not harbor EWSR1 rearrangements: Clinicopathologic characterization of 10 cases. Int J Gynecol Pathol 34(5):437–444., DOI 10.1097/PGP.0000000000000171 Tran TA, Pernicone P, 2012, Anastomosing hemangioma with fatty changes of the genitourinary tract: a lesion mimicking angiomyolipoma. Cent European J Urol 65(1):40–42., DOI 10.5173 /ceju.2012.01.art13 Uppal S, Heller DS, Majmudar B, 2004, Ovarian hemangioma– report of three cases and review of the literature. Arch Gynecol Obstet 270(1):1–5., DOI 10.1007/s00404-003-0584-8 Yamawaki T, Hirai Y, Takeshima N, Hasumi K, 1996, Ovarian hemangioma associated with concomitant stromal luteinization and ascites. Gynecol Oncol 61(3):438–441., DOI 10.1006 /gyno.1996.0170 Ziari K, Alizadeh K, 2016, Ovarian hemangioma: a rare case report and review of the literature. Iran J Pathol 11(1):61–65 Akbulut M, Zekioglu O, Terek MC, Ozdemir N, 2009, Florid vascular proliferation in mature cystic teratoma of the ovary: case report and review of the literature. Tumori 95(1):104–107 Baker PM, Rosai J,YoungRH(2002, Ovarian teratomas with florid benign vascular proliferation: a distinctive finding associated with the neural component of teratomas that may be confused with a vascular neoplasm. Int J Gynecol Pathol 21(1):16–21 Itoh H, Wada T, Michikata K, Sato Y, Seguchi T, Akiyama Y, Kataoka H, 2004, Ovarian teratoma showing a predominant hemangiomatous element with stromal luteinization: report of a case and review of the literature. Pathol Int 54(4):279–283., DOI 10.1111/j.1440-1827.2004.01621.x Pongsuvareeyakul T, Settakorn J, Khorana J, Khunamornpong S, 2015, Ovarian teratoma with predominant hemangiomatous component in early childhood. J Pediatr Adolesc Gynecol 28(1): e1–e3., DOI 10.1016/j.jpag.2014.02.013 Illueca C, Machado I, Garcia A, Covisa A, Morales J, Cruz J, Traves V, Almenar S, 2011, Uncommon vascular tumor of the ovary. Primary ovarian epithelioid hemangioendothelioma or vascular sarcomatous transformation in ovarian germ cell tumor? Arch Gynecol Obstet 284(6):1589–1591., DOI 10.1007 /s00404-011-2036-1 Prus D, Rosenberg AE, Blumenfeld A, Udassin R, Ne'eman Z, Young RH, Ariel I, 1997, Infantile hemangioendothelioma of the ovary: a monodermal teratoma or a neoplasm of ovarian somatic cells? Am J Surg Pathol 21(10):1231–1235 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 717 EP 722 DI 10.1007/s12253-016-0186-y PG 6 ER PT J AU Plones, T Fischer, M Hohne, K Sato, H Muller-Quernheim, J Zissel, G AF Plones, Till Fischer, Mitja Hohne, Kerstin Sato, Hiromi Muller-Quernheim, Joachim Zissel, Gernot TI Turning back the Wheel: Inducing Mesenchymal to Epithelial Transition via Wilms Tumor 1 Knockdown in Human Mesothelioma Cell Lines to Influence Proliferation, Invasiveness, and Chemotaxis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Wilms tumor; Pleural; Asbestos; Pleuropneumonectomy; Decortication; Met; Stemcells; EPP; P/D; Epigenetics; EMT; Epithelial to mesenchymal transition; Cancer; Chemoresistance; Cisplatin; Drug ID Wilms tumor; Pleural; Asbestos; Pleuropneumonectomy; Decortication; Met; Stemcells; EPP; P/D; Epigenetics; EMT; Epithelial to mesenchymal transition; Cancer; Chemoresistance; Cisplatin; Drug AB Malignant pleural mesothelioma (MPM) is a highly aggressive tumor that arises from the surface of the pleura and is associated with a history of asbestos exposure. The tumor is characterized by a strong local invasiveness and a poor response to any single modality therapy. Therefore clinical outcome of patients with MPM is poor and median survival time of untreated patients with MPM is 7 months from initial diagnosis. The Wilms Tumor Protein 1 (WT1) is a transcription factor which is highly expressed by MPM and is involved in cellular development and survival. We evaluated the role of WT1 in two human MPM cell lines (MSTO and H2052) expressing high levels of WT1. We performed a knockdown of WT1 using siRNA. Knockdown of WT1 was confirmed by Westernblotting. After knockdown of WT1 we investigated the effect on proliferation, chemoresistance, chemotaxis and migration. We could demonstrate that knockdown of WT1 suppresses chemoresistance in both cell lines compared with control (scrambled siRNA). Additionally, WT1 knockdown reduces proliferation, chemotaxis and invasiveness of mesothelioma cell lines. WT1 reduces malignancy of malignant mesothelioma cell lines and might be a new molecular target in mesothelioma therapy. Further investigations are needed to discover the mechanisms of chemoresistance depending on WT1. C1 [Plones, Till] University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Fischer, Mitja] Medical Center University of Freiburg, Center for Medicine, Department of PneumologyFreiburg, Germany. [Hohne, Kerstin] Medical Center University of Freiburg, Center for Medicine, Department of PneumologyFreiburg, Germany. [Sato, Hiromi] Chiba University, Graduate School of Pharmaceutical Sciences, Department of Clinical Pharmacology&Pharmacometrics, 1-8-1 Inohana, Chuou-ku, 260-8675 Chiba, Japan. [Muller-Quernheim, Joachim] Medical Center University of Freiburg, Center for Medicine, Department of PneumologyFreiburg, Germany. [Zissel, Gernot] Medical Center University of Freiburg, Center for Medicine, Department of PneumologyFreiburg, Germany. RP Plones, T (reprint author), University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic Surgery, Essen, Germany. EM Till_ploenes@gmx.de CR Merritt N, Blewett CJ, Miller JD, Bennett WF, Young JE, Urschel JD, 2001, Survival after conservative, palliative, management of pleural malignant mesothelioma. J Surg Oncol 78:171–174 Ploenes T, Osei-Agyemang T, Nestle U, Waller CF, Passlick B, 2012, Malignant pleural mesothelioma. Dtsch Med Wochenschr 137:481–486 Ceresoli GL, Locati LD, Ferreri AJ, Cozzarini C, Passoni P, Melloni G, Zannini P, Bolognesi A, Villa E, 2001, Therapeutic outcome according to histologic subtype in 121 patients with malignant pleural mesothelioma. Lung Cancer 34:279–287 Casarsa C, Bassani N, Ambrogi F, Zabucchi G, Boracchi P, Biganzoli E, Coradini D, 2011, Epithelial-to-mesenchymal transition, cell polarity and stemness-associated features in malignant pleural mesothelioma. Cancer Lett 302:136–143 Peinado H, Olmeda D, CanoA, 2007, Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 7:415–428 Kushitani K, Takeshima Y, Amatya VJ, Furonaka O, Sakatani A, Inai K, 2007, Immunohistochemical marker panels for distinguishing between epithelioid mesothelioma and lung adenocarcinoma. Pathol Int 57:190–199 von Gise A, Zhou B, Honor LB, Ma Q, Petryk A, Pu WT, 2011, WT1 regulates epicardial epithelial to mesenchymal transition through beta-catenin and retinoic acid signaling pathways. Dev Biol 356:421–431 Davies JA, Ladomery M, Hohenstein P, Michael L, Shafe A, Spraggon L, Hastie N, 2004, Development of an siRNA-based method for repressing specific genes in renal organ culture and its use to show that theWt1 tumour suppressor is required for nephron differentiation. Hum Mol Genet 13:235–246 Chau YY, Hastie ND, 2012, The role of Wt1 in regulating mesenchyme in cancer, development, and tissue homeostasis. Trends Genet 28:515–524 Hohenstein P, Hastie ND, 2006, The many facets of the Wilms' tumour gene, WT1. Hum Mol Genet 15(Spec No 2):R196–201 Koesters R, Linnebacher M, Coy JF, Germann A, Schwitalle Y, Findeisen P, von Knebel Doeberitz M, 2004, WT1 is a tumorassociated antigen in colon cancer that can be recognized by in vitro stimulated cytotoxic T cells. Int J Cancer 109:385–392 Loeb DM, Evron E, Patel CB, Sharma PM, Niranjan B, Buluwela L, Weitzman SA, Korz D, Sukumar S, 2001, Wilms' tumor suppressor gene, WT1, is expressed in primary breast tumors despite tumor-specific promoter methylation. Cancer Res 61:921–925 Oji Y, Miyoshi S, Maeda H, Hayashi S, Tamaki H, Nakatsuka S, Yao M, Takahashi E, Nakano Y, Hirabayashi H, Shintani Y, Oka Y, Tsuboi A, Hosen N, Asada M, Fujioka T, Murakami M, Kanato K, Motomura M, Kim EH, Kawakami M, Ikegame K, Ogawa H, Aozasa K, Kawase I, Sugiyama H, 2002, Overexpression of the Wilms' tumor geneWT1 in de novo lung cancers. Int J Cancer 100: 297–303 Ploenes T, Osei-Agyemang T, Krohn A, Waller CF, Duncker-Rohr V, Elze M, Passlick B, 2013, Changes in lung function after surgery for mesothelioma. Asian Cardiovasc Thorac Ann 21:48–55 Ploenes T, Scholtes B, Krohn A, Burger M, Passlick B, Muller- Quernheim J, Zissel G, 2013, CC-chemokine ligand 18 induces epithelial to mesenchymal transition in lung cancer A549 cells and elevates the invasive potential. PLoS One 8:e53068 Miller-Hodges E, Hohenstein P, 2012)WT1 in disease: shifting the epithelial-mesenchymal balance. J Pathol 226:229–240 Sugiyama H, 2001, Wilms’ tumor Gene WT1: its oncogenic function and clinical application. Int J Hematol 73:177–187 Zamora-Avila DE, Franco-Molina MA, Trejo-Avila LM, Rodriguez-Padilla C, Resendez-Perez D, Zapata-Benavides P, 2007, RNAi silencing of the WT1 gene inhibits cell proliferation and induces apoptosis in the B16F10 murine melanoma cell line. Melanoma Res 17:341–348 Dohi S, Ohno S, Ohno Y, Soma G, Kyo S, Inoue M, 2009, Correlation between WT1 expression and cell proliferation in endometrial cancer. Anticancer Res 29:4887–4891 Rong Y, Cheng L, Ning H, Zou J, Zhang Y, Xu F, Liu L, Chang Z, Fu XY, 2006)Wilms' tumor 1 and signal transducers and activators of transcription 3 synergistically promote cell proliferation: a possible mechanism in sporadic Wilms' tumor. Cancer Res 66: 8049–8057 Hewitt SM, Hamada S, McDonnell TJ, Rauscher FJ 3rd, Saunders GF, 1995, Regulation of the proto-oncogenes bcl-2 and c-myc by theWilms' tumor suppressor geneWT1. Cancer Res 55:5386–5389 Englert C, Maheswaran S, Garvin AJ, Kreidberg J, Haber DA, 1997, Induction of p21 by the Wilms' tumor suppressor gene WT1. Cancer Res 57:1429–1434 Mayo MW, Wang CY, Drouin SS, Madrid LV, Marshall AF, Reed JC, Weissman BE, Baldwin AS, 1999, WT1 modulates apoptosis by transcriptionally upregulating the bcl-2 proto-oncogene. EMBO J 18:3990–4003 Inoue K, Sugiyama H, Ogawa H, Nakagawa M, Yamagami T, Miwa H, Kita K, Hiraoka A, Masaoka T, Nasu K et al, 1994, WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia. Blood 84:3071– 3079 Bergmann L, Miething C, Maurer U, Brieger J, Karakas T, Weidmann E, Hoelzer D, 1997, High levels of Wilms' tumor gene, wt1, mRNA in acute myeloid leukemias are associated with a worse long-term outcome. Blood 90:1217–1225 ChenMY, Clark AJ, Chan DC,Ware JL, Holt SE, Chidambaram A, Fillmore HL, Broaddus WC, 2011, Wilms' tumor 1 silencing decreases the viability and chemoresistance of glioblastoma cells in vitro: a potential role for IGF-1R de-repression. J Neuro-Oncol 103:87–102 Zapata-Benavides P, Manilla-Munoz E, Zamora-Avila DE, Saavedra-Alonso S, Franco-Molina MA, Trejo-Avila LM, Davalos-Aranda G, Rodriguez-Padilla C, 2012, WT1 silencing by RNAi synergizes with chemotherapeutic agents and induces chemosensitization to doxorubicin and cisplatin in B16F10 murine melanoma cells. Oncol Lett 3:751–755 BarbolinaMV, Adley BP, Shea LD, Stack MS, 2008)Wilms tumor gene protein 1 is associated with ovarian cancer metastasis and modulates cell invasion. Cancer 112:1632–1641 Jomgeow T, Oji Y, Tsuji N, Ikeda Y, Ito K, Tsuda A, Nakazawa T, Tatsumi N, Sakaguchi N, Takashima S, Shirakata T, Nishida S, Hosen N, Kawakami M, Tsuboi A, Oka Y, Itoh K, Sugiyama H, 2006)Wilms' tumor gene WT1 17AA(−)/KTS(−, isoform induces morphological changes and promotes cell migration and invasion in vitro. Cancer Sci 97:259–270 Zhang TF, Yu SQ, Guan LS, Wang ZY, 2003, Inhibition of breast cancer cell growth by the Wilms' tumor suppressor WT1 is associated with a destabilization of beta-catenin. Anticancer Res 23: 3575–3584 Maiti S, Alam R, Amos CI, Huff V, 2000, Frequent association of beta-catenin and WT1 mutations in Wilms tumors. Cancer Res 60: 6288–6292 Kirschner KM, Wagner N, Wagner KD, Wellmann S, Scholz H, 2006, The Wilms tumor suppressor Wt1 promotes cell adhesion through transcriptional activation of the alpha4integrin gene. J Biol Chem 281:31930–31939 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 723 EP 730 DI 10.1007/s12253-016-0181-3 PG 8 ER PT J AU Li, N Tan, Q Jing, W Luo, P Tu, J AF Li, Nandi Tan, Qian Jing, Wei Luo, Ping Tu, Jiancheng TI Long Non-Coding RNA SPRY4-IT1 Can Predict Unfavorable Prognosis and Lymph Node Metastasis: a Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE LncRNA; SPRY4-IT1; Lymph node metastasis; Prognosis ID LncRNA; SPRY4-IT1; Lymph node metastasis; Prognosis AB Emerging evidences suggested that long noncoding RNAs (lncRNAs) play an interesting role in the tumor development and progression in various types of cancer. The aim of this study was to analyse the potential prognostic value for cancer patients . We systematically searched the reports through PubMed, Web of Science, Medline, CNKI, and the Cochrane Library from inception to March, 2016, and carefully identified according to eligibility criteria. This quantitative meta-analysis collected all relevant articles to investigated the association of SPRY4-IT1 expression status with overall survival (OS) and lymph node metastasis (LNM). A total of 765 patients with cancer from 8 studies were included in the final analysis. The hazard ratio (HR) of OS and the odds ratios (OR) of LNM were calculated to assess the association . The meta-analysis results showed high SPRY4-IT1 expression could predict unfavorable OS in various cancers (pooled HR: 2.18, 95% CI: 1.45–3.27, p = 0.001). Moreover, we found high SPRY4-IT1 expression was related to LNM (pooled OR = 3.86, 95%CI:1.31–11.35, P = 0.01). LncRNA SPRY4-IT1 can serve as a new molecular marker for cancer metastasis and prognosis. C1 [Li, Nandi] Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory MedicineWuhan, China. [Tan, Qian] Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory MedicineWuhan, China. [Jing, Wei] Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory MedicineWuhan, China. [Luo, Ping] Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory MedicineWuhan, China. [Tu, Jiancheng] Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory MedicineWuhan, China. RP Tu, J (reprint author), Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory Medicine, Wuhan, China. EM jianchengtu@whu.edu.cn CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in globocan 2012. Int J Cancer 136:E359–E386 Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66:7–30 Knauss JL, Sun T, 2013, Regulatory mechanisms of long noncoding rnas in vertebrate central nervous system development and function. Neuroscience 235:200–214 Tano K, Akimitsu N, 2012, Long non-coding rnas in cancer progression. Front Genet 3:219 Li G, Zhang H, Wan X, Yang X, Zhu C, Wang A, He L, Miao R, Chen S, Zhao H, 2014, Long noncoding rna plays a key role in metastasis and prognosis of hepatocellular carcinoma. Biomed Res Int 2014:780521 YaoY, Li J,Wang L, 2014, Large intervening non-coding rna hotair is an indicator of poor prognosis and a therapeutic target in human cancers. Int J Mol Sci 15:18985–18999 Cai B, Wu Z, Liao K, Zhang S, 2014, Long noncoding rna hotair can serve as a common molecular marker for lymph node metastasis: a meta-analysis. Tumour Biol 35:8445–8450 Cheetham SW, Gruhl F, Mattick JS, Dinger ME, 2013, Long noncoding rnas and the genetics of cancer. Br J Cancer 108:2419–2425 Khaitan D, Dinger ME, Mazar J, Crawford J, Smith MA, Mattick JS, Perera RJ, 2011, The melanoma-upregulated long noncoding rna spry4-it1 modulates apoptosis and invasion. Cancer Res 71: 3852–3862 Xie HW, Wu QQ, Zhu B, Chen FJ, Ji L, Li SQ, Wang CM, Tong YS, Tuo L, Wu M, Liu ZH, Lv J, Shi WH, Cao XF, 2014, Long noncoding rna spry4-it1 is upregulated in esophageal squamous cell carcinoma and associated with poor prognosis. Tumour Biol 35: 7743–7754 Shi Y, Li J, Liu Y, Ding J, Fan Y, Tian Y, Wang L, Lian Y, Wang K, Shu Y, 2015, The long noncoding rna spry4-it1 increases the proliferation of human breast cancer cells by upregulating znf703 expression. Mol Cancer 14:51 Zhang HM, Yang FQ, Yan Y, Che JP, Zheng JH, 2014, High expression of long non-coding rna spry4-it1 predicts poor prognosis of clear cell renal cell carcinoma. Int J Clin Exp Pathol 7:5801–5809 Liu T, Shen SK, Xiong JG, Xu Y, ZhangHQ, Liu HJ, Lu ZG, 2016, Clinical significance of long noncoding rna spry4-it1 in melanoma patients. FEBS Open Bio 6:147–154 Zhao XL, Zhao ZH, Xu WC, Hou JQ, Du XY, 2015, Increased expression of spry4-it1 predicts poor prognosis and promotes tumor growth and metastasis in bladder cancer. Int J Clin Exp Pathol 8: 1954–1960 Zhou Y,Wang DL, Pang Q, 2016, Long noncoding rna spry4-it1 is a prognostic factor for poor overall survival and has an oncogenic role in glioma. Eur Rev Med Pharmacol Sci 20:3035–3039 Cui F,Wu D, He X,WangW, Xi J, Wang M, 2016, Long noncoding rna spry4-it1 promotes esophageal squamous cell carcinoma cell proliferation, invasion, and epithelial-mesenchymal transition. Tumor Biology, 2016, 37(8):1-6. Peng W, Wu G, Fan H, Wu J, Feng J, 2015, Long noncoding rna spry4-it1 predicts poor patient prognosis and promotes tumorigenesis in gastric cancer. Tumour Biol 36:6751–6758 Chen MW, Li JF, Zhuang CL, Liu YC, Chen ZC, He AB, Zhang QX, Huang WR, Cai ZM, 2016, Long noncoding rna spry4-it1 inhibited the progression of bladder cancer. Acta Universitatis Medicinalis Anhui 51:978-983,984. Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR, 2007, Practical methods for incorporating summary time-to-event data into meta-analysis. Trials 8:16 Xie HW, Chen FJ, Zhu B, Cao G, Jin L, Zhou GZ, Lv Z, Cao XF, 2013, Long noncoding rna spry4-it1 expression in esophageal squamous cell carcinoma and its effects on cell growth. Chinese Journal of Clinical Oncology 40(17):1011-1015., DOI 10.3969/j. issn.1000-8179.20130707 Qiu MT, Hu JW, Yin R, Xu L, 2013, Long noncoding rna: an emerging paradigm of cancer research. Tumour Biol 34:613–620 Guttman M, Rinn JL, 2012, Modular regulatory principles of large non-coding rnas. Nature 482:339–346 Bartonicek N,Maag JL, DingerME, 2016, Long noncoding rnas in cancer: mechanisms of action and technological advancements. Mol Cancer 15:43 Brunner AL, Beck AH, Edris B, Sweeney RT, Zhu SX, Li R, Montgomery K, Varma S, Gilks T, Guo X, Foley JW, Witten DM, Giacomini CP, Flynn RA, Pollack JR, Tibshirani R, Chang HY, van de Rijn M, West RB, 2012, Transcriptional profiling of long non-coding rnas and novel transcribed regions across a diverse panel of archived human cancers. Genome Biol 13:R75 Lai MC, Yang Z, Zhou L, Zhu QQ, Xie HY, Zhang F, Wu LM, Chen LM, Zheng SS, 2012, Long non-coding rna malat-1 overexpression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med Oncol 29:1810–1816 Li J, Wen W, Zhao S, Wang J, Chen J, Wang Y, Zhang Q, 2015, Prognostic role of hotair in four estrogen-dependent malignant tumors: a meta-analysis. Onco Targets Ther 8:1471–1482 Zhang J, Zhang B, Wang T, Wang H, 2015, Lncrna malat1 overexpression is an unfavorable prognostic factor in human cancer: evidence from a meta-analysis. Int J Clin Exp Med 8:5499–5505 Zhao W, Mazar J, Lee B, Sawada J, Li JL, Shelley J, Govindarajan S, Towler D,Mattick JS, Komatsu M, Dinger ME, Perera RJ, 2016, The long noncoding rna sprightly regulates cell proliferation in primary human melanocytes. J Invest Dermatol 136:819–828 Mazar J, Zhao W, Khalil AM, Lee B, Shelley J, Govindarajan SS, Yamamoto F, Ratnam M, Aftab MN, Collins S, Finck BN, Han X, Mattick JS, Dinger ME, Perera RJ, 2014, The functional characterization of long noncoding rna spry4-it1 in human melanoma cells. Oncotarget 5:8959–8969 Liu H, Lv Z, Guo E, 2015, Knockdown of long noncoding rna spry4-it1 suppresses glioma cell proliferation, metastasis and epithelial-mesenchymal transition. Int J Clin Exp Pathol 8: 9140–9146 Ru N, Liang J, Zhang F,WuW,Wang F, Liu X, Du Y, 2016, Spry4 intronic transcript 1 promotes epithelial-mesenchymal transition through association with snail1 in osteosarcoma. DNA Cell Biol 35:290–295 Zhang CY, Li RK, Qi Y, Li XN, Yang Y, Liu DL, Zhao J, Zhu DY, Wu K, Zhou XD, Zhao S, 2016, Upregulation of long noncoding rna spry4-it1 promotes metastasis of esophageal squamous cell carcinoma via induction of epithelial-mesenchymal transition. Cell Biol Toxicol 32(5):391–401., DOI 10.1007/s10565-016-9341-1 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 731 EP 736 DI 10.1007/s12253-016-0182-2 PG 6 ER PT J AU Hosoya, K Matsusaka, S Kashiwada, T Suzuki, K Ureshino, N Sato, A Miki, Y Kitera, K Hirai, M Hatake, K Kimura, Sh Sueoka-Aragane, N AF Hosoya, Kazuhisa Matsusaka, Satoshi Kashiwada, Tomomi Suzuki, Koichi Ureshino, Norio Sato, Akemi Miki, Yoshio Kitera, Kazuki Hirai, Mitsuharu Hatake, Kiyohiko Kimura, Shinya Sueoka-Aragane, Naoko TI Detection of KRAS Mutations in Plasma DNA Using a fully Automated Rapid Detection System in Colorectal Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Plasma DNA; KRAS mutation; Colorectal neoplasms; Molecular targeted therapy ID Plasma DNA; KRAS mutation; Colorectal neoplasms; Molecular targeted therapy AB KRAS mutations have been recognized as predictive markers of primary resistance to anti-EGFR-antibodies in colorectal cancer patients. In addition, newly detected KRAS mutations have been reported to be related with acquired resistance to chemotherapy containing anti-EGFR antibody. Considering this evidence, monitoring of KRAS mutations is indispensable for making treatment decisions, and the method should be non-invasive allowing repeated examinations. Recently, we established a novel automated sensitive detection system for KRAS mutations, named mutation-biased PCR quenching probe system (MBP-QP). The goal of our study was to investigate the potential for monitoring KRAS mutations during treatment with anti-EGFR antibodies. The detection limit of MBP-QP using a control plasmid containing KRAS mutations was 1–9 copies, and 0.05–0.3% mutant plasmid was detectable in a mixture of wild type and mutants. One-hundred twenty colorectal cancer patients were genotyped for KRAS mutations with MBP-QP as well as polymerase chain reaction reverse sequence-specific oligonucleotide (PCR-rSSO), which has already been applied to cancer tissue samples in the clinical setting. Concordance rates between plasma DNA and cancer tissues were 68% with MBP-QP and 66% with PCR-rSSO, indicating that these systems are equivalent in terms of detecting KRAS mutations with plasma DNA. KRAS mutations in plasma DNA were frequently observed in systemic metastatic cancer patients, and in three patients KRAS mutations appeared after chemotherapy containing anti-EGFR antibody. A prospective study is needed for clarifying whether KRAS mutations detected in plasma DNA are predictive markers of treatment efficacy with anti- EGFR antibody. C1 [Hosoya, Kazuhisa] Saga University, Faculty of Medicine, Department of Internal Medicine, 5-1-1 Nabeshima, 849-8501 Saga, Japan. [Matsusaka, Satoshi] Cancer Institute Hospital of Japanese Foundation for Cancer Research, Gastroenterological CenterTokyo, Japan. [Kashiwada, Tomomi] Saga University, Faculty of Medicine, Department of Internal Medicine, 5-1-1 Nabeshima, 849-8501 Saga, Japan. [Suzuki, Koichi] Jichi Medical University, Saitama Medical Center, Department of SurgerySaitama, Japan. [Ureshino, Norio] Saga Prefectural Hospital Koseikan, Department of Medical OncologySaga, Japan. [Sato, Akemi] Saga University, Faculty of Medicine, Department of Internal Medicine, 5-1-1 Nabeshima, 849-8501 Saga, Japan. [Miki, Yoshio] Japanese Foundation for Cancer Research, The Cancer Institute, Department of Genetic DiagnosisTokyo, Japan. [Kitera, Kazuki] ARKRAY Inc., Research and Development DivisionKyoto, Japan. [Hirai, Mitsuharu] ARKRAY Inc., Research and Development DivisionKyoto, Japan. [Hatake, Kiyohiko] Japanese Foundation for Cancer Research, Cancer Chemotherapy Center, Clinical ChemotherapyTokyo, Japan. [Kimura, Shinya] Saga University, Faculty of Medicine, Department of Internal Medicine, 5-1-1 Nabeshima, 849-8501 Saga, Japan. [Sueoka-Aragane, Naoko] Saga University, Faculty of Medicine, Department of Internal Medicine, 5-1-1 Nabeshima, 849-8501 Saga, Japan. RP Sueoka-Aragane, N (reprint author), Saga University, Faculty of Medicine, Department of Internal Medicine, 849-8501 Saga, Japan. EM sueokan@cc.saga-u.ac.jp CR Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, Tebbutt NC, Simes RJ, Chalchal H, Shapiro JD, Robitaille S, Price TJ, Shepherd L, Au HJ, Langer C, Moore MJ, Zalcberg JR, 2008, K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 359:1757–1765 Amado RG,Wolf M, Peeters M,Van Cutsem E, Siena S, Freeman DJ, Juan T, Sikorski R, Suggs S, Radinsky R, Patterson SD, Chang DD, 2008, Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 26:1626–1634 Malumbres M, Barbacid M, 2003, RAS oncogenes: the first 30 years. Nat Rev Cancer 3:459–465 Benvenuti S, Sartore-Bianchi A, Di Nicolantonio F, Zanon C, Moroni M, Veronese S, Siena S, Bardelli A, 2007, Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti-epidermal growth factor receptor antibody therapies. Cancer Res 67:2643–2648 Tol J, Punt CJ, 2010, Monoclonal antibodies in the treatment of metastatic colorectal cancer: a review. Clin Ther 32:437–453 De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, Kalogeras KT, Kotoula V, Papamichael D, Laurent- Puig P, Penault-Llorca F, Rougier P, Vincenzi B, Santini D, Tonini G, Cappuzzo F, FrattiniM,Molinari F, Saletti P, De Dosso S, Martini M, Bardelli A, Siena S, Sartore-Bianchi A, Tabernero J,Macarulla T, Di Fiore F, Gangloff AO, Ciardiello F, Pfeiffer P, Qvortrup C, Hansen TP, Van Cutsem E, Piessevaux H, Lambrechts D, Delorenzi M, Tejpar S, 2010, Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol 11:753–762 Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, Rivera F, Kocakova I, Ruff P, Blasinska-Morawiec M, Smakal M, Canon JL, Rother M, Williams R, Rong A, Wiezorek J, Sidhu R, Patterson SD, 2013, Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med 369:1023–1034 Bouchahda M, Karaboue A, Saffroy R, Innominato P, Gorden L, Guettier C, Adam R, Levi F, 2010, Acquired KRAS mutations during progression of colorectal cancer metastases: possible implications for therapy and prognosis. Cancer Chemother Pharmacol 66:605–609 Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, Bencardino K, Cercek A, Chen CT, Veronese S, Zanon C, Sartore-Bianchi A, Gambacorta M, Gallicchio M, Vakiani E, Boscaro V, Medico E, Weiser M, Siena S, Di Nicolantonio F, Solit D, Bardelli A, 2012, Emergence of KRAS mutations and acquired resistance to anti- EGFR therapy in colorectal cancer. Nature 486:532–536 Diaz LA Jr, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, Allen B, Bozic I, Reiter JG, Nowak MA, Kinzler KW, Oliner KS, Vogelstein B, 2012, The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 486:537– 540 Misale S, Nicolantonio F, Sartore-Bianchi A, Siena S, Bardelli A, 2014, Resistance to anti-EGFR therapy in colorectal cancer: from heterogeneity to convergent evolution. Cancer Discov 4:1269– 1280 Troiani T, Martinelli E, Napolitano S, Vitagliano D, Ciuffreda LP, Costantino S, Morgillo F, Capasso A, Sforza V, Nappi A, De Palma R, D'Aiuto E, Berrino L, Bianco R, Ciardiello F, 2013, Increased TGF-α as a mechanism of acquired resistance to the anti-EGFR inhibitor cetuximab through EGFR-MET interaction and activation of MET signaling in colon cancer cells. Clin Cancer Res 19:6751– 6765 Arena S, Bellosillo B, Siravegna G,Martinez A, Canadas I, Lazzari L, Ferruz N, Russo M, Misale S, Gonzalez I, IglesiasM, Gavilan E, Corti G, Hobor S, Crisafulli G, Salido M, Sanchez J, Dalmases A, Bellmunt J,De Fabritiis G, Rovira A, Di Nicolantonio F, Albanell J, Bardelli A, Montagut C, 2015, Emergence of multiple EGFR extracellular mutations during cetuximab treatment in colorectal cancer. Clin Cancer Res 21:2157–2166 Misale S, Bozic I, Tong J, Peraza-Penton A, Lallo A, Baldi F, Lin KH, Truini M, Trusolino L, Bertotti A, Di Nicolantonio F, Nowak MA, Zhang L,Wood KC, Bardelli A, 2015, Vertical suppression of the EGFR pathway prevents onset of resistance in colorectal cancers. Nat Commun 6:8305 Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, Varela I, Phillimore B, BegumS,McDonald NQ, Butler A, Jones D, Raine K, Latimer C, Santos CR, NohadaniM, Eklund AC, Spencer-Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA, Swanton C, 2012, Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366:883–892 Swanton C, 2012, Intratumor heterogeneity: evolution through space and time. Cancer Res 72:4875–4882 Nakamura T, Sueoka-Aragane N, Iwanaga K, Sato A, Komiya K, Abe T, Ureshino N, Hayashi S, Hosomi T, Hirai M, Sueoka E, Kimura S, 2011, A non-invasive system for monitoring resistance to EGFR tyrosine kinase inhibitors with plasma DNA. J Thorac Oncol 6:1639–1648 Nakamura T, Sueoka-Aragane N, Iwanaga K, Sato A, Komiya K, Kobayashi N, Hayashi S, Hosomi T, Hirai M, Sueoka E, Kimura S, 2012, Application of a highly sensitive detection system for epidermal growth factor receptor mutations in plasma DNA. J Thorac Oncol 7:1369–1381 Sueoka-Aragane N, Katakami N, Satouchi M, Yokota S, Aoe K, Iwanaga K, Otsuka K, Morita S, Kimura S, Negoro S, Hanshin- Saga Collaborative Cancer Study Group, 2015, Monitoring EGFR T790 M with plasma DNA from lung cancer patients in a prospective observational study. Cancer Sci 107:162–167 Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, Jia M, Shepherd R, Leung K, Menzies A, Teague JW, Campbell PJ, Stratton MR, Futreal PA, 2011, COSMIC: mining complete cancer genomes in the catalogue of somatic mutations in cancer. Nucleic Acids Res 39(Database issue):D945–D950 Tabernero J, Lenz HJ, Siena S, Sobrero A, Falcone A, Ychou M, Humblet Y, Bouche O, Mineur L, Barone C, Adenis A, Yoshino T, Goldberg RM, Sargent DJ,Wagner A, Laurent D, TeufelM, Jeffers M, Grothey A, Van Cutsem E, 2015, Analysis of circulating DNA and protein biomarkers to predict the clinical activity of regorafenib and assess prognosis in patients with metastatic colorectal cancer: a retrospective, exploratory analysis of the CORRECT trial. Lancet Oncol 16:937–948 Yoshino T, Muro K, Yamaguchi K, Nishina T, Denda T, Kudo T, OkamotoW, Taniguchi H, Akagi K, Kajiwara T, Hironaka S, Satoh T, 2015, Clinical validation of a multiplex kit for RAS mutations in colorectal cancer: results of the RASKET, RAS KEy testing, prospective, multicenter study. EBioMedicine 2:317–323 Thierry AR, Mouliere F, ElMessaoudi S, Mollevi C, Lopez-Crapez E, Rolet F, Gillet B, Gongora C, Dechelotte P, Robert B, Del Rio M, Lamy PJ, Bibeau F, Nouaille M, Loriot V, Jarrousse AS, Molina F, Mathonnet M, Pezet D, Ychou M, 2014, Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nat Med 20:430–435 Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, Antonarakis ES, Azad NS, Bardelli A, Brem H, Cameron JL, Lee CC, Fecher LA, Gallia GL, Gibbs P, Le D, Giuntoli RL, Goggins M, Hogarty MD, Holdhoff M, Hong SM, Jiao Y, Juhl HH, Kim JJ, Siravegna G, Laheru DA, Lauricella C, Lim M, Lipson EJ, Marie SK, Netto GJ, Oliner KS, Olivi A, Olsson L, Riggins GJ, Sartore-Bianchi A, Schmidt K, Shih lM, Oba-Shinjo SM, Siena S, Theodorescu D, Tie J, Harkins TT, Veronese S, Wang TL, Weingart JD, Wolfgang CL, Wood LD, Xing D, Hruban RH, Wu J, Allen PJ, Schmidt CM, Choti MA, Velculescu VE, Kinzler KW, Vogelstein B, Papadopoulos N, Diaz LA Jr, 2014, Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 6:224ra24 Lech G, Slotwinski R, Slodkowski M, Krasnodebski IW, 2016, Colorectal cancer tumour markers and biomarkers: recent therapeutic advances. World J Gastroenterol 22:1745–1755 Spindler KL, Pallisgaard N, Vogelius I, Jakobsen A, 2012, Quantitative cell-free DNA, KRAS, and BRAF mutations in plasma from patients with metastatic colorectal cancer during treatment with cetuximab and irinotecan. Clin Cancer Res 18:1177–1185 Sueoka-Aragane N, Sato A, Kobayashi N, Ide M, Yokoo M, Nagano Y, Sueoka E, Okada S, Kimura S, 2014, Correlation between plasma DNA and tumor status in an animal model. PLoS One 9:e111881 Schrag D,Weng S, Brooks G, Meyerhardt J, Venook A, 2016, The association between primary tumor location and survival in colorectal cancer. ASCO Annual Meeting NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 737 EP 744 DI 10.1007/s12253-016-0175-1 PG 8 ER PT J AU Shen, L Zhao, L Tang, J Wang, Z Bai, W Zhang, F Wang, Sh Li, W AF Shen, Li Zhao, Lizhi Tang, Jiquan Wang, Zhiwei Bai, Weisong Zhang, Feng Wang, Shouli Li, Weihua TI Key Genes in Stomach Adenocarcinoma Identified via Network Analysis of RNA-Seq Data SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Stomach adenocarcinoma; Gene expression data; Differentially expressed genes; Gene co-expression network; Functional enrichment analysis ID Stomach adenocarcinoma; Gene expression data; Differentially expressed genes; Gene co-expression network; Functional enrichment analysis AB RNA-seq data of stomach adenocarcinoma (STAD) were analyzed to identify critical genes in STAD. Meanwhile, relevant small molecule drugs, transcription factors (TFs) and microRNAs (miRNAs) were also investigated. Gene expression data of STAD were downloaded from The Cancer Genome Atlas (TCGA). Differential analysis was performed with package edgeR. Relationships with correlation coefficient > 0.6 were retained in the gene co-expression network. Functional enrichment analysis was performed for the genes in the network with DAVID and KOBASS 2.0. Modules were identified using Cytoscape. Relevant small molecules drugs, transcription factors (TFs) and microRNAs (miRNAs) were revealed by using CMAP and WebGestalt databases. A total of 520 DEGs were identified between 285 STAD samples and 33 normal controls, including 244 up-regulated and 276 downregulated genes. A gene co-expression network containing 53 DEGs and 338 edges was constructed, the genes of which were significantly enriched in focal adhesion, ECM-receptor interaction and vascular smooth muscle contraction pathways. Three modules were identified from the gene co-expression network and they were associated with skeletal system development, inflammatory response and positive regulation of cellular process, respectively. A total of 20 drugs, 9 TFs and 6 miRNAs were acquired that may regulate the DEGs. NFAT-COL1A1/ ANXA1, HSF2-FOS, SREBP-IL1RN and miR-26-COL5A2 regulation axes may be important mechanisms for STAD. C1 [Shen, Li] HanZhong Central Hospital, Department of Digestive Surgery, 723000 Hanzhong, Shaanxi, China. [Zhao, Lizhi] HanZhong Central Hospital, Department of Digestive Surgery, 723000 Hanzhong, Shaanxi, China. [Tang, Jiquan] HanZhong Central Hospital, Department of Digestive Surgery, 723000 Hanzhong, Shaanxi, China. [Wang, Zhiwei] HanZhong Central Hospital, Department of Digestive Surgery, 723000 Hanzhong, Shaanxi, China. [Bai, Weisong] HanZhong Central Hospital, Department of Digestive Surgery, 723000 Hanzhong, Shaanxi, China. [Zhang, Feng] HanZhong Central Hospital, Department of Digestive Surgery, 723000 Hanzhong, Shaanxi, China. [Wang, Shouli] HanZhong Central Hospital, Department of Digestive Surgery, 723000 Hanzhong, Shaanxi, China. [Li, Weihua] The People’s Hospital in Gansu Province, Center Lab, No, 204 west Donggang Road, 730000 Lanzhou, Gansu Province, China. RP Li, W (reprint author), The People’s Hospital in Gansu Province, Center Lab, 730000 Lanzhou, China. EM weiweihhh@aliyun.com CR Lindblad M, Rodriguez LAG, Lagergren J, 2005, Body mass, tobacco and alcohol and risk of esophageal, gastric cardia, and gastric non-cardia adenocarcinoma among men and women in a nested case-control study. Cancer Causes Control 16(3):285–294 Orditura M, Galizia G, Sforza V, Gambardella V, Fabozzi A, Laterza MM, Andreozzi F, Ventriglia J, Savastano B, Mabilia A et al, 2014, Treatment of gastric cancer. World J Gastroenterol 20(7):1635–1649 Ema A, Yamashita K, Ushiku H, Kojo K, Minatani N, Kikuchi M, Mieno H, Moriya H, Hosoda K, Katada N, 2014, Immunohistochemical analysis of RTKs expression identified HER3 as a prognostic indicator of gastric cancer. Cancer Sci 105(12):1591–1600 Wu X, Chen Y, Li G, Xia L, Gu R,Wen X,Ming X, Chen H, 2014, Her3 is associated with poor survival of gastric adenocarcinoma: Her3 promotes proliferation, survival and migration of human gastric cancer mediated by PI3K/AKT signaling pathway. Med Oncol 31(4):1–11 Zhang Y, Cai X, Chai N, Gu Y, Zhang S, Ding M, Cao H, Sha S, Yin J, LiM(2015, SIRT1 is reduced in gastric adenocarcinoma and acts as a potential tumor suppressor in gastric cancer. Gastrointest Tumors 2:109–123 Okada K, Fujiwara Y, Takahashi T, Nakamura Y, Takiguchi S, Nakajima K, Miyata H, Yamasaki M, Kurokawa Y, Mori M, 2013, Overexpression of forkhead box M1 transcription factor, FOXM1, is a potential prognostic marker and enhances chemoresistance for docetaxel in gastric cancer. Ann Surg Oncol 20(3):1035–1043 Jiang W, Zhou F, Li N, Li Q, Wang L, 2015, FOXM1-LDHA signaling promoted gastric cancer glycolytic phenotype and progression. Int J Clin Exp Pathol 8(6):6756–6763 Zhang M, Wang X, Li W, Cui Y, 2015, miR-107 and miR-25 simultaneously target LATS2 and regulate proliferation and invasion of gastric adenocarcinoma, GAC, cells. Biochem Biophys Res Commun 460(3):806–812 Sun J, Nishiyama T, Shimizu K, Kadota K, 2013, TCC: an R package for comparing tag count data with robust normalization strategies. BMC Bioinforma 14(1):1–14 Robinson MD, McCarthy DJ, Smyth GK, 2010, edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26(1):139–140 Benjamini Y, 2010, Discovering the false discovery rate. J R Stat Soc Ser B Stat Methodol 72(4):405–416 Dawson JA, Ye S, Kendziorski C, 2012, R/EBcoexpress: an empirical Bayesian framework for discovering differential co-expression. Bioinformatics 28(14):1939–1940 Smoot ME, Ono K, Ruscheinski J, Wang PL, Ideker T, 2011, Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics 27(3):431–432 Tanay A, Sharan R, Kupiec M, Shamir R, 2004, Revealing modularity and organization in the yeast molecular network by integrated analysis of highly heterogeneous genomewide data. Proc Natl Acad Sci U S A 101(9):2981–2986 Wu J, Mao X, Cai T, Luo J, Wei L, 2006, KOBAS server: a webbased platform for automated annotation and pathway identification. Nucleic Acids Res 34(Web Server issue):W720–W724 Rivera CG, Vakil R, Bader JS, 2010, NeMo: network module identification in Cytoscape. BMC Bioinforma 11(Suppl 1):S61 Maere S, Heymans K, Kuiper M, 2005, BiNGO: a Cytoscape plugin to assess overrepresentation of gene ontology categories in biological networks. Bioinformatics 21(16):3448–3449 Segal MR, Xiong H, Bengtsson H, Bourgon R, Gentleman R, 2012, Querying genomic databases: refining the connectivity map. Stat Appl Genet Mol Biol 11(2):1544–6115 Zhang B, Kirov S, Snoddy J, 2005, WebGestalt: an integrated system for exploring gene sets in various biological contexts. Nucleic Acids Res 33(suppl 2):W741–W748 Wang J, Duncan D, Shi Z, Zhang B, 2013, WEB-based GEne SeT AnaLysis toolkit, WebGestalt): update 2013. Nucleic Acids Res 41(Web Server issue):W77–W83 Wu Q, Li X, Yang H, Lu C, You J, Zhang Z, 2014, Extracellular matrix protein 1 is correlated to carcinogenesis and lymphatic metastasis of human gastric cancer. World J Surg Oncol 12(1): 132–132 Wala SJ, Karamchandani JR, Saleeb R, Evans A, Ding Q, Ibrahim R, Jewett M, Pasic M, Finelli A, Pace K, 2015, An integrated genomic analysis of papillary renal cell carcinoma type 1 uncovers the role of focal adhesion and extracellular matrix pathways. Mol Oncol 9(8):1667–1677 Shi Y, Wang J, Xin Z, Duan Z, Wang G, Li F, 2015, Transcription factors and microRNA-Co-regulated genes in gastric cancer invasion in ex vivo. PLoS One 10(4):1330–1339 Willis CM, Kluppel M, 2014, Chondroitin sulfate-E is a negative regulator of a pro-tumorigenic Wnt/beta-catenin-collagen 1 axis in breast cancer cells. PLoS One 9(8):e103966–e103966 Fischer H, Stenling R, Rubio C, Lindblom A, 2001, Colorectal carcinogenesis is associated with stromal expression of COL11A1 and COL5A2. Carcinogenesis 22(6):875–878 Hui-Ying X-Z, Chun-Min L, 2016, Inflammatory microenvironment contributes to epithelial-mesenchymal transition in gastric cancer. World J Gastroenterol 22(29):6619–6628 Deng Q, He B, Liu X, Yue J, Ying H, Pan Y, Sun H, Chen J,Wang F, Gao T, 2015, Prognostic value of pre-operative inflammatory response biomarkers in gastric cancer patients and the construction of a predictive model. J Transl Med 13(1):1–16 Raja UM, Gopal G, Shirley S, Ramakrishnan AS, Rajkumar T, 2017, Immunohistochemical expression and localization of cytokines/ chemokines/growth factors in gastric cancer. Cytokine 89: 82–90 Worst TS, Reiner V, Gabriel U,Weiß C, Erben P, Martini T, Bolenz C, 2014, IL1RN and KRT13 expression in bladder cancer: association with pathologic characteristics and smoking status. Ther Adv Urol 2013:184602–184602 Yu G, Wang J, Chen Y, Wang X, Pan J, Li Q, Xie K, 2008, Tissue microarray analysis reveals strong clinical evidence for a close association between loss of annexin A1 expression and nodal metastasis in gastric cancer. Clin Exp Metastasis 25(7):695–702 Zhou L, Zhang JS, Yu JC, Cui QC, ZhouWX, KangWM, Ma ZQ, 2010, Negative association of c-fos expression as a favorable prognostic indicator in gastric cancer. Arch Med Res 41(3):201–206 Oliveira-Ferrer L, Roßler K, Haustein V, Schroder C, Wicklein D, Maltseva D, Khaustova N, Samatov T, Tonevitsky A, Mahner S, 2014, C-FOS suppresses ovarian cancer progression by changing adhesion. Br J Cancer 110(3):753–763 MullerMR, Rao A, 2010, NFAT, immunity and cancer: a transcription factor comes of age. Nat Rev Immunol 10(9):645–656 Mancini M, Toker A, 2009, NFAT proteins: emerging roles in cancer progression. Nat Rev Cancer 9(11):810–820 Bjork JK, Akerfelt M, Joutsen J, PuustinenMC, Cheng F, Sistonen L, Nees M, 2015, Heat-shock factor 2 is a suppressor of prostate cancer invasion. Oncogene 35(14):1770–1784 Huang W-C, Li X, Liu J, Lin J, Chung LWK, 2011, Activation of androgen receptor, lipogenesis and oxidative stress converged by SREBP-1 is responsible for regulating growth and progression of prostate cancer cells. Mol Cancer Res 10(1):133–142 Qiu X, Zhu H, Liu S, Tao G, Jin J, Chu H,Wang M, Tong N, Gong W, Zhao Q, 2016, Expression and prognostic value of microRNA- 26a and microRNA-148a in gastric cancer. J Gastroenterol Hepatol., DOI 10.1111/jgh.13533 Giovanella BC, Hinz HR, Kozielski AJ, Stehlin JS, Silber R, Potmesil M, 1991, Complete growth inhibition of human cancer xenografts in nude mice by treatment with 20-(S)-camptothecin. Cancer Res 51(11):3052–3055 Mochizuki Y, Ohashi N, Kojima H, Ishigure K,Kinoshita T, Eguchi T, Fujitake S, Ito S, Fujiwara M, Kodera Y, 2013, CPT-11 as a second-line treatment for patients with advanced/metastatic gastric cancer who failed S-1, CCOG0702). Cancer Chemother Pharmacol 72(3):629–635 Lee MH, Yang JY, Cho Y, Park M, Woo HJ, Kim HW, Kwon HJ, Kim SH, Moon C, Kim TU, 2016)Menadione induces apoptosis in a gastric cancer cell line mediated by down-regulation of X-linked inhibitor of apoptosis. Int J Clin Exp Med 9(2):2437–2443 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 745 EP 752 DI 10.1007/s12253-016-0178-y PG 8 ER PT J AU Lakatos, G Petranyi, Szucs, A Nehez, L Harsanyi, L Hegyi, P Bodoky, Gy AF Lakatos, Gabor Petranyi, Agota Szucs, Attila Nehez, Laszlo Harsanyi, Laszlo Hegyi, Peter Bodoky, Gyorgy TI Efficacy and Safety of FOLFIRINOX in Locally Advanced Pancreatic Cancer. A Single Center Experience. SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic cancer; Locally advanced; FOLFIRINOX ID Pancreatic cancer; Locally advanced; FOLFIRINOX AB The management of locally advanced pancreatic cancer (LAPC) is a major challenge. Although new drugs are available for the treatment of metastatic disease, the optimal treatment of non-metastatic cases remains controversial. The role of neoadjuvant therapy is still a question of debate in this setting. The aim of the study was to prospectively collect and analyse data on efficacy and safety of a modified FOLFIRINOX regimen in LAPC patients treated in a single institution. Another major objective was to assess the capability of FOLFIRINOX to render primary non-resectable cancer to resectable. No bolus fluorouracil was given and a 20% dose reduction of oxaliplatin and irinotecan was applied. Primary GCSF prophylaxis was applied to prevent febrile neutropenia. Thirty-two patients (mean age 60.2 years, range: 40–77 years) have been enrolled into the study. All patients had ECOG performance status of 0 or 1. Best response to therapy was stable disease (SD) or partial regression (PR) in 18 (56.2%) and 6 (18.8%) cases. Two patients (6.3%) underwent surgical resection (100% R0). The most frequent grade 3/4 adverse events were nausea (18.8%), fatigue (12.5%) and diarrhea (12.5%). The incidence of severe neutropenia was 28.1%, with only one documented case of febrile neutropenia. The probability of disease progression was 25% and 50%after 75 and 160 days with 88.4% of possibility of disease progression after 500 days. OS probability was 92.1, 71.5% and 49.5% at 180-, 365 and 540 days. Our data does not support the capability of FOLFIRINOX to render primary non-resectable cancer to resectable. However, due to the high disease control rate observed, FOLFRINOX might be recommended as first line option for the palliative treatment of LAPC. Despite reduced chemotherapy doses significant toxicity has been seen. C1 [Lakatos, Gabor] Egyesitett Szent Istvan es Szent Laszlo Korhaz, Onkologiai Osztaly, 5-7 Albert Florian street, H-1097 Budapest, Hungary. [Petranyi, Agota] Egyesitett Szent Istvan es Szent Laszlo Korhaz, Onkologiai Osztaly, 5-7 Albert Florian street, H-1097 Budapest, Hungary. [Szucs, Attila] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Nehez, Laszlo] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Harsanyi, Laszlo] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Hegyi, Peter] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Bodoky, Gyorgy] Egyesitett Szent Istvan es Szent Laszlo Korhaz, Onkologiai Osztaly, 5-7 Albert Florian street, H-1097 Budapest, Hungary. RP Lakatos, G (reprint author), Egyesitett Szent Istvan es Szent Laszlo Korhaz, Onkologiai Osztaly, H-1097 Budapest, Hungary. EM lakagab@yahoo.com CR Ferlay J, Partensky C, Bray F, 2016, More deaths from pancreatic cancer than breast cancer in the EU by 2017. Acta Oncol 55(9–10): 1158–1160 Hariharan D, Saied A, Kocher HM, 2008, Analysis of mortality rates for pancreatic cancer across the world. HPB, Oxford, 10:58–62 Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2014, GLOBOCAN 2012 v1.1, cancer incidence and mortality worldwide: IARC CancerBase no. 11 [internet]. International Agency for Research on Cancer, Lyon Bodoky G, Lakatos G, 2014, Management of pancreatic cancer today. Klinikai Onkologia 1(1):23–29 Conroy T et al, 2011, FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 364(19):1817–1825 Suker M, Beumer BR, Sadot E, Marthey L, Faris JE, Mellon EA, El-Rayes BF, Wang-Gillam A, Lacy J, Hosein PJ, Moorcraft SY, Conroy T, Hohla F, Allen P, Taieb J, Hong TS, Shridhar R, Chau I, van Eijck CH, 2016, Groot Koerkamp B.FOLFIRINOX for locally advanced pancreatic cancer: a systematic review and patient-level meta-analysis. Lancet Oncol 17(6):801–810., DOI 10.1016/S1470- 2045(16)00172-8 Heinemann V, Haas M, Boeck S, 2013, Neoadjuvant treatment of borderline resectable and non-resectable pancreatic cancer. Ann Oncol 0:1–8 National Comprehensive Cancer Network, 2015). NCCN Guidelines Version 2. Pancreatic Adenocarcinoma. http://www. nccn.org Mahaseth H, Brutcher E, Kauh J, Hawk N, Kim S, Chen Z, Kooby DA, Maithel SK, Landry J, El-Rayes BF, 2013, Modified FOLFIRINOX regimen with improved safety and maintained efficacy in pancreatic adenocarcinoma. Pancreas 42(8):1311–1315., DOI 10.1097/MPA.0b013e31829e2006 Stein SM, James ES, Deng Y, Cong X, Kortmansky JS, Li J, Staugaard C, Indukala D, Boustani AM, Patel V, Cha CH, Salem RR, Chang B, Hochster HS, Lacy J, 2016, Final analysis of a phase II study of modified FOLFIRINOX in locally advanced and metastatic pancreatic cancer. Br J Cancer 114(7):737–743., DOI 10.1038 /bjc.2016.45 Marthey L, Sa-Cunha A, Blanc JF, Gauthier M, Cueff A, Francois E, Trouilloud I, Malka D, Bachet JB, Coriat R, Terrebonne E, De La Fouchardiere C, Manfredi S, Solub D, Lecaille C, Thirot Bidault A, Carbonnel F, Taieb J, 2015, FOLFIRINOX for locally advanced pancreatic adenocarcinoma: results of an AGEO multicenter prospective observational cohort. Ann Surg Oncol 22(1):295–301., DOI 10.1245/s10434-014-3898-9 Rombouts SJ,Walma MS, Vogel JA, van Rijssen LB,Wilmink JW, Mohammad NH, van Santvoort HC, Molenaar IQ, Besselink MG, 2016, Systematic review of resection rates and clinical outcomes after FOLFIRINOX-based treatment in patients with locally advanced pancreatic cancer. Ann Surg Oncol 23(13):4352–4360 Barton JG, Bois JP, Sarr MG, Wood CM, Qin R, Thomsen KM, Kendrick ML, Farnell MB, 2009, Predictive and prognostic value of CA19-9 in resected pancreatic adenocarcinoma. J Gastrointest Surg 13:2050–2058 Wang-Gillam A, Li CP, Bodoky G, Dean A, Shan YS, Jameson G, Macarulla T, Lee KH, Cunningham D, Blanc JF, Hubner RA, Chiu CF, Schwartsmann G, Siveke JT, Braiteh F, Moyo V, Belanger B, Dhindsa N, Bayever E, Von Hoff DD, Chen LT, 2016, NAPOLI-1 Study Group. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreaticcancer after previous gemcitabine-based therapy, NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet. Feb 6;387, 10018):545–57., DOI 10.1016/S0140-6736(15)00986-1. Epub 2015 Nov 29 Conroy T, Paillot B, Francois E et al, 2005, Irinotecan plus oxaliplatin and leucovorin-modulated fluorouracil in advanced pancreatic cancer—a Groupe Tumeurs digestives of the federation Nationale des Centres de Lutte Contre le cancer study. J Clin Oncol 23:1228–1236 Gunturu KS, Yao X, Cong X et al, 2013, FOLFIRINOX for locally advanced and metastatic pancreatic cancer: single institution retrospective review of efficacy and toxicity. Med Oncol 30:361 Hosein PJ, Macintyre J, Kawamura C et al, 2012, A retrospective study of neoadjuvant FOLFIRINOX in unresectable or borderlineresectable locally advanced pancreatic adenocarcinoma. BMC Cancer 12:199 Faris JE, Blaszkowsky LS, McDermott S, Guimaraes AR, Szymonifka J, Huynh MA, Ferrone CR, Wargo JA, Allen JN, Dias LE, Kwak EL, Lillemoe KD, Thayer SP, Murphy JE, Zhu AX, Sahani DV, Wo JY, Clark JW, Fernandez-del Castillo C, Ryan DP, Hong TS, 2013, FOLFIRINOX in locally advanced pancreatic cancer: the Massachusetts General Hospital cancer center experience. Oncologist 18(5):543–548., DOI 10.1634 /theoncologist.2012-0435 Peddi PF, Lubner S, McWilliams R et al, 2012, Multi-institutional experience with FOLFIRINOX in pancreatic adenocarcinoma. JOP 13:497–501 Rombouts SJ, Mungroop TH, Heilmann MN, van Laarhoven HW, Busch OR, Molenaar IQ, Besselink MG, Wilmink JW, 2016, FOLFIRINOX in locally advanced and metastatic pancreatic cancer: a single Centre cohort study. J Cancer 7(13):1861–1866 eCollection 2016 Blazer M,Wu C, Goldberg RM, Phillips G, Schmidt C,Muscarella P,Wuthrick E,Williams TM, Reardon J, Ellison EC, Bloomston M, Bekaii-Saab T, 2015, Neoadjuvant modified, m, FOLFIRINOX for locally advanced unresectable, LAPC, and borderline resectable, BRPC, adenocarcinoma of the pancreas. Ann Surg Oncol 22(4): 1153–1159., DOI 10.1245/s10434-014-4225-1 Boone BA, Steve J, Krasinskas AM, Zureikat AH, Lembersky BC, Gibson MK, Stoller RG, Zeh HJ, Bahary N, 2013, Outcomes with FOLFIRINOX for borderline resectable and locally unresectable pancreatic cancer. J Surg Oncol 108(4):236–241., DOI 10.1002 /jso.23392 Moorcraft SY, Khan K, Peckitt C,Watkins D, Rao S, Cunningham D, Chau I, 2014 Dec, FOLFIRINOX for locally advanced or metastatic pancreatic ductal adenocarcinoma: the RoyalMarsden experience. Clin Colorectal Cancer 13(4):232–238., DOI 10.1016/j. clcc.2014.09.005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 753 EP 759 DI 10.1007/s12253-016-0176-0 PG 7 ER PT J AU Li, Z Zhu, K Gong, X Vasilescu, S Sun, Y Hong, K Li, H Li, L Shan, Y AF Li, Zhandong Zhu, Ketong Gong, Xin Vasilescu, Steven Sun, Yu Hong, Kaiqing Li, Hao Li, Lin Shan, Yaming TI Inducing Polyclonal Eag1-Specific Antibodies by Vaccination with a Linear Epitope Immunogen and Its Relation to Breast Tumorigenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Eag1; Breast cancer; Polyclonal Eag1-specific antibodies; Immunohistochemical marker ID Eag1; Breast cancer; Polyclonal Eag1-specific antibodies; Immunohistochemical marker AB Ether a-go-go 1 (KCNH1, Kv10.1) (Eag1) is a voltage-gated potassium channel, which is commonly overexpressed in tested breast cancer patients. This occurrence makes it a potential molecular marker and a promising tool for breast cancer diagnosis and therapy. In order to explore protective or specific polyclonal antibodies for further research, potential linear epitopes from Eag1 were collected by sequence alignment. The sequence was synthesized and then coupled to the carrier protein keyhole limpet hemocyanin (KLH) for animal immunization. Polyclonal antibodies against Eag1 were produced and purified from the rabbit antisera. Enzyme linked immunosorbent assay (ELISA) and western blot were performed to characterize their specificities. Immunohistochemical staining was carried out on normal and cancerous breast tissue sections using the purified polyclonal Eag1-specific antibodies. The results indicate that the overexpression of Eag1 might be associated with an increased risk of progression to breas t cancer (Grade 1 tissue = 57.89%;Grade 2 tissue = 92.59%;Grade 3 tissue = 100%). These results also suggest that Eag1 gene is a putative growth-promoting gene that might be involved in breast tumorigenesis and development. Eag1 might further be represented as a potential target for some human diseases treatment. C1 [Li, Zhandong] Jilin Engineering Normal University, College of Biology and Food EngineeringChangchun, Jilin, China. [Zhu, Ketong] Jilin Engineering Normal University, College of Biology and Food EngineeringChangchun, Jilin, China. [Gong, Xin] Jilin University, School of Life Sciences, National Engineering Laboratory for AIDS VaccineChangchun, Jilin, China. [Vasilescu, Steven] University of Technology Sydney, School of Mathematical and Physical Sciences, Materials Analysis Unit, 2007 Ultimo, NSW, Australia. [Sun, Yu] Jilin Engineering Normal University, College of Biology and Food EngineeringChangchun, Jilin, China. [Hong, Kaiqing] Jilin Engineering Normal University, College of Biology and Food EngineeringChangchun, Jilin, China. [Li, Hao] Jilin Engineering Normal University, College of Biology and Food EngineeringChangchun, Jilin, China. [Li, Lin] Jilin University, China-Japan Union HospitalChangchun, Jilin, China. [Shan, Yaming] Jilin University, School of Life Sciences, National Engineering Laboratory for AIDS VaccineChangchun, Jilin, China. RP Li, H (reprint author), Jilin Engineering Normal University, College of Biology and Food Engineering, Changchun, China. EM muyelee@sina.com CR Li ZD, Wu Y, Bao YL, CL Y, Guan LL, Wang YZ, Meng XY, Li YX, 2009, Identification and characterization of human ARIP2 and its relation to breast cancer. Cytokine 46:251–259 StuhmerW, Alves F, Hartung F, Zientkowska M, Pardo LA, 2006, Potassium channels as tumour markers. FEBS Lett 580:2850–2852 Pardo LA, del Camino D, Sanchez A, Alves F, Bruggemann A, Beckh S, Stuhmer W, 1999, Oncogenic potential of EAG K(+, channels. EMBO J 18:5540–5547 Pardo LA, Stuhmer W, 2008, Eag1: an emerging oncological target. Cancer Res 68:1611–1613 Lin H, Li Z, Chen C, Luo X, Xiao J, Dong D, Lu Y, Yang B,Wang Z, 2011, Transcriptional and post-transcriptional mechanisms for oncogenic overexpression of ether a go-go K+ channel. PLoS One 6:e20362 Wadhwa S, Wadhwa P, Dinda AK, Gupta NP, 2009, Differential expression of potassium ion channels in human renal cell carcinoma. Int Urol Nephrol 41:251–257 Pardo LA, Bruggemann A, Camacho J, Stuhmer W, 1998, Cell cycle-related changes in the conducting properties of r-eag K+ channels. J Cell Biol 143:767–775 Weber C, Mello de Queiroz F, Downie BR, Suckow A, StuhmerW, Pardo LA, 2006, Silencing the activity and proliferative properties of the human EagI Potassium Channel by RNA interference. J Biol Chem 281:13030–13037 Gomez-Varela D, Zwick-Wallasch E, Knotgen H, Sanchez A, Hettmann T, Ossipov D, Weseloh R, Contreras-Jurado C, Rothe M, Stuhmer W, Pardo LA, 2007, Monoclonal antibody blockade of the human Eag1 potassium channel function exerts antitumor activity. Cancer Res 67:7343–7349 Hemmerlein B, Weseloh RM, Mello de Queiroz F, Knotgen H, Sanchez A, Rubio ME, Martin S, Schliephacke T, Jenke M, Heinz Joachim R, Stuhmer W, Pardo LA, 2006, Overexpression of Eag1 potassium channels in clinical tumours. Mol Cancer 5:41 Ludwig J, Weseloh R, Karschin C, Liu Q, Netzer R, Engeland B, Stansfeld C, Pongs O, 2000, Cloning and functional expression of rat eag2, a new member of the ether-a-go-go family of potassium channels and comparison of its distribution with that of eag1. Mol Cell Neurosci 16:59–70 Pardo LA, Contreras-Jurado C, Zientkowska M, Alves F, Stuhmer W, 2005, Role of voltage-gated potassium channels in cancer. J Membr Biol 205:115–124 Pardo LA, 2004, Voltage-gated potassium channels in cell proliferation. Physiology, Bethesda, 19:285–292 Conti M, 2004, Targeting K+ channels for cancer therapy. J Exp Ther Oncol 4:161–166 Ashcroft FM, 2006, From molecule to malady. Nature 440:440– 447 Patt S, Preussat K, Beetz C, Kraft R, Schrey M, Kalff R, Schonherr K, Heinemann SH, 2004, Expression of ether a go-go potassium channels in human gliomas. Neurosci Lett 368:249–253 Bijlenga P, Occhiodoro T, Liu JH, Bader CR, Bernheim L, Fischer- Lougheed J, 1998, An ether -a-go-go K+ current, Ih-eag, contributes to the hyperpolarization of human fusion-competent myoblasts. J Physiol 512(Pt 2):317–323 Meyer R, Heinemann SH, 1998, Characterization of an eag-like potassium channel in human neuroblastoma cells. J Physiol 508(Pt 1):49–56 Meyer R, Schonherr R, Gavrilova-RuchO,WohlrabW, Heinemann SH, 1999, Identification of ether a go-go and calcium-activated potassium channels in human melanoma cells. J Membr Biol 171: 107–115 Ouadid-Ahidouch H, Le Bourhis X, Roudbaraki M, Toillon RA, Delcourt P, Prevarskaya N, 2001, Changes in the K+ currentdensity of MCF-7 cells during progression through the cell cycle: possible involvement of a h-ether.A-gogo K+ channel. Receptors Channels 7:345–356 Gavrilova-Ruch O, Schonherr K, Gessner G, Schonherr R, Klapperstuck T, Wohlrab W, Heinemann SH, 2002, Effects of imipramine on ion channels and proliferation of IGR1 melanoma cells. J Membr Biol 188:137–149 Warmke JW, Ganetzky B, 1994, A family of potassium channel genes related to eag in drosophila and mammals. Proc Natl Acad Sci U S A 91:3438–3442 Farias LM, Ocana DB, Diaz L, Larrea F, Avila-Chavez E, Cadena A, Hinojosa LM, Lara G, Villanueva LA, Vargas C, Hernandez- Gallegos E, Camacho-Arroyo I, Duenas-Gonzalez A, Perez- Cardenas E, Pardo LA, Morales A, Taja-Chayeb L, Escamilla J, Sanchez-Pena C, Camacho J, 2004, Ether a go-go potassium channels as human cervical cancer markers. Cancer Res 64:6996–7001 Shinmura K, Igarashi H, Kato H, Koda K, Ogawa H, Takahashi S, Otsuki Y, Yoneda T, Kawanishi Y, Funai K, Takayama T, Ozono S, Sugimura H, 2015, BSND and ATP6V1G3: novel immunohistochemicalmarkers for Chromophobe renal cell carcinoma.Medicine, Baltimore, 94:e989 Wang RF, 1999, Human tumor antigens: implications for cancer vaccine development. J Mol Med, Berl, 77:640–655 Mitelman FJB, Mertens F, Mitelman Database of Chromosome Aberrations and Gene Fusions in Cancer. http://cgapncinihgov/ Chromosomes/Mitelman: Jenke M, Sanchez A, Monje F, StuhmerW,WeselohRM, Pardo LA, 2003, C-terminal domains implicated in the functional surface expression of potassium channels. EMBO J 22:395–403 Garcia-Ferreiro RE, Kerschensteiner D, Major F, Monje F, Stuhmer W, Pardo LA, 2004, Mechanism of block of hEag1 K+ channels by imipramine and astemizole. J Gen Physiol 124:301–317 Napp J, Monje F, Stuhmer W, Pardo LA, 2005, Glycosylation of Eag1, Kv10.1, potassium channels: intracellular trafficking and functional consequences. J Biol Chem 280:29506–29512 Natali PG, Nicotra MR, Bigotti A, Venturo I, Slamon DJ, Fendly BM, Ullrich A, 1990, Expression of the p185 encoded by HER2 oncogene in normal and transformed human tissues. Int J Cancer 45:457–461 Menard S, Tagliabue E, Campiglio M, Pupa SM, 2000, Role of HER2 gene overexpression in breast carcinoma. J Cell Physiol 182: 150–162 Bange J, Zwick E, Ullrich A, 2001, Molecular targets for breast cancer therapy and prevention. Nat Med 7:548–552 Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, Cameron D, Dowsett M, Barrios CH, Steger G, Huang CS, Andersson M, Inbar M, Lichinitser M, Lang I, Nitz U, Iwata H, Thomssen C, Lohrisch C, Suter TM, Ruschoff J, Suto T, Greatorex V,Ward C, Straehle C, McFadden E, Dolci MS, Gelber RD, 2005, Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med 353:1659–1672 Krieg PA, Melton DA, 1987, In vitro RNA synthesis with SP6 RNA polymerase. Methods Enzymol 155:397–415 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 761 EP 767 DI 10.1007/s12253-016-0158-2 PG 7 ER PT J AU Chen, J Wang, Z Lv, Q Du, Z Tan, Q Zhang, D Xiong, B Zeng, H Gou, J AF Chen, Jie Wang, Zu Lv, Qing Du, Zhenggui Tan, Qiuwen Zhang, Di Xiong, Bingjun Zeng, Helin Gou, Juxiang TI Comparison of Core Needle Biopsy and Excision Specimens for the Accurate Evaluation of Breast Cancer Molecular Markers: a Report of 1003 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Core needle biopsy; Hormone receptor; HER2; Ki-67; Molecular markers ID Breast cancer; Core needle biopsy; Hormone receptor; HER2; Ki-67; Molecular markers AB In this study, we compared the accuracy of marker evaluation in core needle biopsy (CNB) specimens versus excision specimens (ESs) from breast cancer patients. This retrospective study used data collected from the breast cancer database at the West China Hospital, China. Immunohistochemistry (IHC) results from CNB specimens and ESs were compared, using estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), and Ki-67 as markers. Molecular subtyping and endocrine therapy usage correlations based on CNB samples and ESs were evaluated. The results obtained from CNB samples and ESs exhibited substantial agreement for the detection of ER (κ = 0.522), PR(κ = 0.441), and HER2 (κ = 0.451), and also influenced endocrine therapy usage. Fair and poor correlations were observed for Ki-67 staining and molecular subtyping (κ = 0.195), respectively. This disagreement might be attributable to a combination of heterogeneity and large tumor size. This study indicates that the discordance rate in molecular marker staining between CNB specimens and ESs is significant enough that results obtained with CNB specimens should be used cautiously or verified using ESs. C1 [Chen, Jie] Sichuan University, West China Hospital, Department of Breast and Thyroid Surgery, 610041 Chengdu, China. [Wang, Zu] Sichuan University, West China Hospital, Tumor Molecular Laboratory, 610041 Chengdu, China. [Lv, Qing] Sichuan University, West China Hospital, Department of Breast and Thyroid Surgery, 610041 Chengdu, China. [Du, Zhenggui] Sichuan University, West China Hospital, Department of Breast and Thyroid Surgery, 610041 Chengdu, China. [Tan, Qiuwen] Sichuan University, West China Hospital, Department of Breast and Thyroid Surgery, 610041 Chengdu, China. [Zhang, Di] Sichuan University, West China Hospital, Department of Breast and Thyroid Surgery, 610041 Chengdu, China. [Xiong, Bingjun] Sichuan University, West China Hospital, Department of Breast and Thyroid Surgery, 610041 Chengdu, China. [Zeng, Helin] Sichuan University, West China Hospital, Department of Breast and Thyroid Surgery, 610041 Chengdu, China. [Gou, Juxiang] Sichuan University, West China Hospital, Department of Breast and Thyroid Surgery, 610041 Chengdu, China. CR NCCN Clinical Practice Guidelines in Oncology: Breast cancer, 2015, www.NCCN.com. Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D, 2000, Molecular portraits of human breast tumors. Nature 406:747–752 Goncalves AV, Thuler LC, Kestelman FP, Carmo PA, Lima CF, Cipolotti R, 2011, Underestimation of malignancy of core needle biopsy for nonpalpable breast lesions. Rev Bras Ginecol Obstet 33: 123–131 Dekker TJ, Smit VT, Hooijer GK, Van de Vijver MJ, Mesker WE, Tollenaar RA, Nortier JW, Kroep JR, 2013, Reliability of core needle biopsy for determining ER andHER2 status in breast cancer. Ann Oncol 24:931–937., DOI 10.1093/annonc/mds599 Chen X,YuanY, Gu Z, ShenK(2012)Accuracy of estrogen receptor, progesterone receptor, and HER2 status between core needle and open excision biopsy in breast cancer: a meta-analysis. Breast Cancer Res Treat 134:957–967., DOI 10.1007/s10549-012-1990-z Goldhirsch A, Wood WC, Coates A, Gelber RD, Thurlimann H, Senn HJ, Panel member, 2011, Subtypes—dealing with the diversity of breast cancer: highlights of the St Gallen international expert consensus on the primary therapy of early breast cancer 2011. Ann Oncol 22:1736–1747 Harris L, Fritsche H, Mennel R, Norton L, Ravdin P, Taube S, Somerfield MR, Hayes DF, Bast RC Jr, 2007, American Society of Clinical Oncology: American society of clinical oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J Clin Oncol 25:5287–5312 Hammond ME, Hayes DF, Wolff AC, Mangu PB, Temin S, 2010, American society of clinical oncology/college of American pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Oncol Pract 6:195–197., DOI 10.1200/JOP.777003 Usami S, Moriya T, Amari M, Suzuki A, Ishida T, Sasano H, Ohuchi N, 2007, Reliability of prognostic factors in breast carcinoma determined by core needle biopsy. Jpn J Clin Oncol 37:250– 255., DOI 10.1093/jjco/hym021 Ricci MD, Calvano Filho CM, Oliveira Filho HR, Filassi JR, Pinotti JA, Baracat EC, 2012, Analysis of the concordance rates between core needle biopsy and surgical excision in patients with breast cancer. Rev Assoc Med Bras 58:532– 536., DOI 10.1016/S0104-4230(12)70245-2 Petrau C, Clatot F, Cornic M, Berghian A, Veresezan L, Callonnec F, Baron M, Veyret C, Laberge S, Thery JC, Picquenot JM, 2015, Reliability of prognostic and predictive factors evaluated by needle Core biopsies of large breast invasive tumors. Am J Clin Pathol 144:555–562., DOI 10.1309/AJCP9KFVM2GZMNDV Litherland JC, Evans AJ, Wilson AR, Kollias J, Pinder SE, Elston CW, Ellis IO, Yeoman LJ, 1996, The impact of core-biopsy on preoperative diagnosis rate of screen detected breast cancers. Clin Radiol 51:562–565., DOI 10.1016/S0009-9260(96)80136-X Youk JH, Kim EK, Kim MJ, Oh KK, 2008, Sonographically guided 14-gauge core needle biopsy of breast masses: a review of 2,420 cases with long-term-follow-up. AJR Am J Roentgenol 190:202– 207., DOI 10.2214/AJR.07.2419 Collins LC, Connolly JL, Page DL, Goulart RA, Pisano ED, Fajardo LL, Berg WA, Caudry DJ, McNeil BJ, Schnitt SJ, 2004, Diagnostic agreement in the evaluation of image-guided breast core needle biopsies: results from a randomized clinical trial. Am J Surg Pathol 28:126–123., DOI 10.1097/00000478-200401000-00015 Crowe JP Jr, Rim A, Patrick RJ, Rybicki LA, Grundfest- Broniatowski SF, Kim JA, Lee KB, 2003, Does core needle breast biopsy accurately reflect breast pathology? Surgery 134:523–526 Crowe JP Jr, Patrick RJ, Rybicki LA, Grundfest SF, Kim JA, Lee KB, Rim A, 2003, Does ultrasound core breast biopsy predict histologic finding on excisional biopsy? Am J Surg 186:397–399 Li S, Yang X, Zhang Y, Fan L, Zhang F, Chen L, Zhou Y, Chen X, Jiang J, 2012, Assessment accuracy of core needle biopsy for hormone receptors in breast cancer: ameta-analysis. Breast Cancer Res Treat 135:325–334., DOI 10.1007/s10549-012-2063-z Sutela A, Vanninen R, Sudah M, Berg M, Kiviniemi V, Rummukainen J, Kataja V, Karja V, 2008, Surgical specimen can be replaced by core samples in assessment of ER, PR and HER-2 for invasive breast cancer. Acta Oncol 47(1):38–46., DOI 10.1080/02841860701441822 Seferina SC, Nap M, van den Berkmortel F, Wals J, Voogd AC, Tjan-Heijnen VC, 2013, Reliability of receptor assessment on core needle biopsy in breast cancer patients. Tumour Biol 34:987–994., DOI 10.1007/s13277-012-0635-5 Lorgis V, Algros MP, Villanueva C, Chaigneau L, Thierry- Vuillemin A, Nguyen T, Demarchi M, Bazan F, Sautiere JL, Maisonnette-Lescot Y, Ringenbach F, Bontemps P, Pivot X, 2011, Discordance in early breast cancer for tumour grade, estrogen receptor, progesterone receptors and human epidermal receptor-2 status between core needle biopsy and surgical excisional primary tumour. Breast 20:284–287., DOI 10.1016/j.breast.2010.12.007 Munch-Petersen HD, Rasmussen BB, Balslev E, 2014, Reliability of histological malignancy grade, ER and HER2 status on core needle biopsy vs surgical specimen in breast cancer. APMIS 122: 750–754., DOI 10.1111/apm.12213 Chen X, Zhu S, Fei X, Garfield DH, Wu J, Huang O, Li Y, Zhu L, He J, Chen W, Jin X, Shen K, 2015, Surgery time interval and molecular subtype may influence Ki67 change after core needle biopsy in breast cancer patients. BMC Cancer 15:822., DOI 10.1186/s12885-015-1853-1 Greer LT, Rosman M,MylanderWC, Hooke J, Kovatich A, Sawyer K, Buras RR, Shriver CD, Tafra L, 2013, Does breast tumor heterogeneity necessitate further immunohistochemical staining on surgical specimens? J Am Coll Surg 216:239–251., DOI 10.1016/j.jamcollsurg.2012.09.007 Ough M, Velasco J, Hieken TJ, 2011, A comparative analysis of core needle biopsy and final excision for breast cancer: histology and marker expression. Am J Surg 201:692– 694., DOI 10.1016/j.amjsurg.2010.02.015 Zhang Z, Yuan P, Guo H, Zhao L, Ying J, Wang M, Zhao H, Pan Q, Xu B, 2015, Assessment of hormone receptor and human epidermal growth factor receptor 2 status in breast carcinoma using thin-prep cytology fine needle aspiration cytology FISH experience from China. Medicine, Baltimore, 94:e981., DOI 10.1097/MD.0000000000000981 Chen X, Sun L, Mao Y, Zhu S, Wu J, Huang O, Li Y, Chen W, Wang J, Yuan Y, Fei X, Jin X, Shen K, 2013, Preoperative core needle biopsy is accurate in determining molecular subtypes in invasive breast cancer. BMC Cancer 13:390., DOI 10.1186/1471- 2407-13-390 Uy GB, Laudico AV, Carnate JM Jr, Lim FG, Fernandez AM, Rivera RR, Mapua CA, Love RR, 2010, Breast cancer hormone receptor assay results of core needle biopsy and modified radical mastectomy specimens from the same patients. Clin Breast Cancer 10:154–159., DOI 10.3816/CBC.2010.n.021 VandenBussche CJ, Cimino-Mathews A, Park BH, Emens LA, Tsangaris TN, Argani P, 2015, Reflex estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2 analysis of breast cancers in needle core biopsy specimens dramatically increases health care costs. Am J Surg Pathol 39:939–947., DOI 10.1097/PAS.0000000000000424 Douglas-Jones AG, Collett N, Morgan JM, Jasani B, 2001, Comparison of core estrogen receptor, ER, assay with excised tumour: intratumoral distribution of ER in breast carcinoma. J Clin Pathol 54:951–955 Tamaki K, Sasano H, Ishida T, Miyashita M, Takeda M, Amari M, Tamaki N, Ohuchi N, 2010, Comparison of core needle biopsy, CNB, and surgical specimens for accurate preoperative evaluation of ER, PgRand HER2 status of breast cancer patients. Cancer Sci 101:2074–2079., DOI 10.1111/j.1349-7006.2010.01630.x MelottiMK, BergWA, 2000, Core needle breast biopsy in patients undergoing anticoagulation therapy: preliminary results. AJR Am J Roentgenol 174:245–249., DOI 10.2214/ajr.174.1.1740245 Ozdemir A, Voyvoda NK, Gultekin S, Tuncbilek I, Dursun A, Yamac D, 2007, Can core biopsy be used instead of surgical biopsy in the diagnosis and prognostic factor analysis of breast carcinoma? Clin Breast Cancer 7:791–795., DOI 10.3816/CBC.2007.n.041 Hoda SA, Harigopal M, Harris GC, Pinder SE, Lee AH, Ellis IO, 2003, Expert opinion: reporting needle core biopsies of breast carcinomas. Histopathology 43:84–90 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 769 EP 775 DI 10.1007/s12253-017-0187-5 PG 7 ER PT J AU Kuronya, Zs Sinkovics, I Agoston, P Biro, K Bodrogi, I Bode, I Dank, M Gyergyay, F Vajdics, T Kolonics, Zs Nagyivanyi, K Ruzsa, Geczi, L AF Kuronya, Zsofia Sinkovics, Istvan Agoston, Peter Biro, Krisztina Bodrogi, Istvan Bode, Imre Dank, Magdolna Gyergyay, Fruzsina Vajdics, Timea Kolonics, Zsuzsanna Nagyivanyi, Krisztian Ruzsa, Agnes Geczi, Lajos TI A Retrospective Analysis of the First 41 mCRPC Patients with Bone Pain Treated with Radium-223 at the National Institute of Oncology in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bone pain; Metastatic castration-resistant prostate cancer (mCRPC); Radium-223 ID Bone pain; Metastatic castration-resistant prostate cancer (mCRPC); Radium-223 AB Radium-223 dichloride is an alpha-emitting radiopharmaceutical which significantly prolongs overall survival in patients with castration-resistant prostate cancer (CRPC) and symptomatic bone metastases. This was a retrospective analysis of the efficacy and safety of Radium-223 in the first 41 patients treated at a single center in Hungary. Radium-223 was given at a dose of 50 kBq/kg intravenously every 4 weeks for up to 6 cycles. Between 23rd July 2014 and 23rd February 2016, 41 patients were treated. Patient demographics, laboratory values, treatment outcomes and adverse events were collected from medical records. The mean age was 72.2 years (SD: 7.1). 24 patients received Radium-223 as first-line treatment (58%), 7 patients as second (17%), 3 as third (7.3%), 6 as (14.6%), and 1 as fifth-line therapy (2.4%). The mean number of cycles administered was 5.5 (SD: 1.1). The most common side effects were anemia (32% grade 1–3), nausea (28%, grade 1), diarrhea (4%, grade 2), thrombocytopenia (4%, grade 3). The mean baseline PSA level was 307.2 ng/ml (SD: 525.7), which increased to a mean value of 728.5 ng/ ml (SD: 1277) by the end of treatment. The baseline mean ALP of 521.1 U/L (SD: 728) decreased to 245.1 U/L (SD: 283.5). The majority of patients experienced a decrease (37%) or complete cessation (43%) of bone pain intensity. In our symptomatic prostate cancer patient population, Radium-223 proved to be efficient in terms of pain relief, with moderate side effects. No PSA response was detected, while alkaline phosphatase levels significantly decreased. C1 [Kuronya, Zsofia] National Institute of Oncology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Sinkovics, Istvan] National Institute of Oncology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Agoston, Peter] National Institute of Oncology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Biro, Krisztina] National Institute of Oncology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Bodrogi, Istvan] National Institute of Oncology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Bode, Imre] Jahn Ferenc South-Pest HospitalBudapest, Hungary. [Dank, Magdolna] Semmelweis University, 1st Department of Internal MedicineBudapest, Hungary. [Gyergyay, Fruzsina] National Institute of Oncology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Vajdics, Timea] National Institute of Oncology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Kolonics, Zsuzsanna] Kaposi Mor Teaching HospitalKaposvar, Hungary. [Nagyivanyi, Krisztian] National Institute of Oncology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Ruzsa, Agnes] Kaposi Mor Teaching HospitalKaposvar, Hungary. [Geczi, Lajos] National Institute of Oncology, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. RP Kuronya, Zs (reprint author), National Institute of Oncology, H-1122 Budapest, Hungary. EM kuronyaz@gmail.com CR Hotte SJ, Saad F, 2010, Current management of castrate-resistant prostate cancer. Curr Oncol 17(Suppl 2):S72–S79 Saad F, Chi KN, Finelli A, Hotte SJ, Izawa J, Kapoor A, Kassouf W, Loblaw A, North S, Rendon R, So A, Usmani N, Vigneault E, Fleshner NE, 2015, The 2015 CUA-CUOG guidelines for themanagement of castration-resistant prostate cancer, CRPC). Can Urol Assoc J 9(3–4):90–96., DOI 10.5489/cuaj.2526 Attard G, Parker C, Eeles RA, Schroder F, Tomlins SA, Tannock I, Drake CG, de Bono JS, 2016, Prostate cancer. Lancet 387(10013): 70–82 DePuy V, Anstrom KJ, Castel LD, Schulman KA, Weinfurt KP, Saad F, 2007, Effects of skeletal morbidities on longitudinal patient-reported outcomes and survival in patients with metastatic prostate cancer. Support Care Cancer 15(7):869–876 Bubendorf L, Schopfer A, Wagner U, Sauter G, Moch H, Willi N, Gasser TC, Mihatsch MJ, 2000, Metastatic patterns of prostate cancer: an autopsy study of 1,589 patients. Hum Pathol 31(5): 578–583 Pelger RC, Soerdjbalie-Maikoe V, Hamdy N, 2001, Strategies for management of prostate cancer-related bone pain. Drugs Aging 18(12):899–911 Sartor O, Coleman R, Nilsson S, Heinrich D, Helle SI, O'Sullivan JM, Fossa SD, Chodacki A, Wiechno P, Logue J, Widmark A, Johannessen DC, Hoskin P, James ND, Solberg A, Syndikus I, Vogelzang NJ, O'Bryan-Tear CG, Shan M, Bruland OS, Parker C, 2014, Effect of radium-223 dichloride on symptomatic skeletal events in patients with castrationresistant prostate cancer and bone metastases: results from a phase 3, double-blind, randomised trial. Lancet Oncol 15(7): 738–746., DOI 10.1016/S1470-2045(14)70183-4 Nilsson S, 2016, Radionuclide therapies in prostate cancer: integrating radium-223 in the treatment of patients with metastatic castration-resistant prostate cancer. Curr Oncol Rep 18(2):14., DOI 10.1007/s11912-015-0495-4 Shore N, 2015, Radium-223 dichloride for metastatic castrationresistant prostate cancer: the urologist's perspective. Urology 85(4): 717–724., DOI 10.1016/j.urology.2014.11.031 Kerr C, 2002,, 223)Ra targets skeletal metastases and spares normal tissue. Lancet Oncol 3(8):453 Bruland OS, Nilsson S, Fisher DR, Larsen RH, 2006, Highlinear energy transfer irradiation targeted to skeletal metastases by the alpha-emitter 223Ra: adjuvant or alternative to conventional modalities? Clin Cancer Res 12(20 Pt 2): 6250s–6257s Henriksen G, Breistol K, Bruland OS, Fodstad O, Larsen RH, 2002, Significant antitumor effect from bone-seeking, alphaparticle- emitting, 223)Ra demonstrated in an experimental skeletal metastases model. Cancer Res 62(11):3120–3215 Dorff TB, GrossME(2015, Radium 223: how can we optimize this new tool for metastatic castration-resistant prostate cancer? Am Soc Clin Oncol Educ Book:e270–e273., DOI 10.14694/EdBook_ AM.2015.35.e270 Nilsson S, Strang P, Aksnes AK, Franzen L, Olivier P, Pecking A, Staffurth J, Vasanthan S, Andersson C, Bruland OS, 2012, A randomized, dose-response, multicenter phase II study of radium-223 chloride for the palliation of painful bone metastases in patients with castration-resistant prostate cancer. Eur J Cancer 48(5):678– 686., DOI 10.1016/j.ejca.2011.12.023 Jadvar H, Quinn DI, 2013, Targeted α-particle therapy of bone metastases in prostate cancer. Clin Nucl Med 38(12):966–971., DOI 10.1097/RLU.0000000000000290 Brady D, Parker CC, O'Sullivan JM, 2013, Bone-targeting radiopharmaceuticals including radium-223. Cancer J 19(1):71–78., DOI 10.1097/PPO.0b013e318282479 Lien LM, Tvedt B, Heinrich D, 2015, Treatment of castrationresistant prostate cancer and bone metastases with radium-223 dichloride. Int J Urol Nurs 9(1):3–13 McGann S, Horton ER, 2015, Radium-223 dichloride: a novel treatment option for castration-resistant prostate cancer patients with symptomatic bone metastases. Ann Pharmacother 49(4): 469–476., DOI 10.1177/1060028014565444 Parker C, Nilsson S, Heinrich D, Helle SI, O'Sullivan JM, Fossa SD, Chodacki A, Wiechno P, Logue J, Seke M, Widmark A, Johannessen DC, Hoskin P, Bottomley D, James ND, Solberg A, Syndikus I, Kliment J,Wedel S, Boehmer S, Dall'OglioM, Franzen L, Coleman R, Vogelzang NJ, O'Bryan-Tear CG, Staudacher K, Garcia-Vargas J, Shan M, Bruland OS, Sartor O, Investigators ALSYMPCA, 2013, Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 369(3):213–223., DOI 10.1056/NEJMoa1213755 Bayer Pharma AG, 2015, Xofigo®, radium Ra 223 dichloride, solution for injection Summary of Product Characteristics, SmPC, http://www.accessdata.fda.gov/drugsatfda_docs/label/2013 /203971lbl.pdf Nilsson S, 2015, Radium-223 dichloride for the treatment of bone metastatic castration-resistant prostate cancer: an evaluation of its safety. Expert Opin Drug Saf 14(7):1127–2236., DOI 10.1517 /14740338.2015.1045874 Modi D, Hwang C, Mamdani H, Kim S, Gayar H, Vaishampayan U, Joyrich R, Heath EI, 2016, Radium-223 in heavily pretreated metastatic castrate-resistant prostate cancer. Clin Genitourin Cancer ., DOI 10.1016/j.clgc.2016.03.002pii: S1558-7673(16 )30056-8 Hoskin P, Sartor O, O'Sullivan JM, Johannessen DC, Helle SI, Logue J, Bottomley D, Nilsson S, Vogelzang NJ, Fang F, Wahba M, Aksnes AK, Parker C, 2014, Efficacy and safety of radium-223 dichloride in patients with castration-resistant prostate cancer and symptomatic bone metastases, with or without previous docetaxel use: a prespecified subgroup analysis from the randomised, doubleblind, phase 3 ALSYMPCA trial. Lancet Oncol 15(12):1397–1406., DOI 10.1016/S1470-2045(14)70474-7 Autio KA, Bennett AV, Jia X, Fruscione M, Beer TM, George DJ, CarducciMA, Logothetis CJ, Kane RC, Sit L, Rogak L, Morris MJ, Scher HI, Basch EM(2013, Prevalence of pain and analgesic use in men with metastatic prostate cancer using a patient-reported outcome measure. J Oncol Pract 9(5):223–229., DOI 10.1200 /JOP.2013.000876 Salvati M, Frati A, Russo N, Brogna C, Piccirilli M, D'Andrea G, Occhiogrosso G, Pichierri A, Caroli E, 2005, Brain metastasis from prostate cancer. Report of 13 cases and critical analysis of the literature. J Exp Clin Cancer Res 24(2):203–207 Gavrilovic IT, Posner JB, 2005, Brain metastases: epidemiology and pathophysiology. J Neuro-Oncol 75(1):5–14 Caffo O, Gernone A, Ortega C, Sava T, Carteni G, Facchini G, Re GL, Amadio P, Bortolus R, Pagliarulo V, Prati V, Veccia A, Galligioni E, 2012, Central nervous system metastases from castration-resistant prostate cancer in the docetaxel era. J Neuro- Oncol 107:191–196 Caffo O, Veccia A, Russo L, Galligioni E, 2012, Brain metastases from prostate cancer. Future Oncol 8(12):1585–1595 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 777 EP 783 DI 10.1007/s12253-017-0190-x PG 7 ER PT J AU Wang, Y AF Wang, Yan TI Transcriptional Regulatory Network Analysis for Gastric Cancer Based on mRNA Microarray SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Transcriptional regulatory network; Transcription factor ID Gastric cancer; Transcriptional regulatory network; Transcription factor AB We aimed to screen the differential expressed genes (DEGs) and transcriptional factors (TFs) related to gastric cancer. GSE19826 microarray data downloaded from Gene Expression Omnibus was used to identify the differentially expressed genes (DEGs) and PPI network of DEGs were constructed by the Retrieval of Interacting Genes database. Pathway enrichment analysis of DEGs were performed by Gene Set Enrichment Analysis. Then, the transcriptional regulatory network was constructed based on TRANSFAC database. Finally, regulatory impact factor (RIF) of TF was calculated. We identified 446 DEGs including 209 up- and 237 down-regulated genes. These DEGs were mainly significantly enriched in 5 pathways including ECM receptor interaction (p = 0.013899), spliceosome (p = 0.025591), bladder cancer (p = 0.026316), focal adhesion (p = 0.047809) and WNT signaling pathway (p = 0.048077). PPI network with 247 nodes and 913 edges were constructed and COL5A2 was the hub node. Transcriptional regulatory network with 6 differently expressed TFs, 58 non-differently expressed TFs, 44 DEGs and 735 non-DEGs was constructed. Finally, top 5 TFs including CRX, TFAP4, NKX2–1,MYB and RARG with higher ZRIF were screened. The identified DEGs such as COL5A2 and TOP2A, and TFs including EGR2, FOXM1, NKX2–1 and TFAP4 might be the critical genes and TFs for gastric cancer. C1 [Wang, Yan] China Medical University, Shengjing Hospital, Department of Gastroenterology, No. 36 Sanhao Road, 110004 Shenyang, China. RP Wang, Y (reprint author), China Medical University, Shengjing Hospital, Department of Gastroenterology, 110004 Shenyang, China. EM yan_wang741@163.com CR Ohtsu A, Shah MA, Van Cutsem E, Rha SY, Sawaki A, Park SR, Lim HY, Yamada Y, Wu J, Langer B, 2011, Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol 29(30):3968–3976 Vainio A, Auvinen A, 1996, Prevalence of symptoms among patients with advanced cancer: an international collaborative study. J Pain Symptom Manag 12(1):3–10 Wroblewski LE, Peek RM, Wilson KT, 2010, Helicobacter pylori and gastric cancer: factors that modulate disease risk. Clin Microbiol Rev 23(4):713–739 Kluijt I, Siemerink EJ, Ausems MG, van Os TA, de Jong D, Simoes-Correia J, van Krieken JH, Ligtenberg MJ, Figueiredo J, van Riel E, 2012, CDH1-related hereditary diffuse gastric cancer syndrome: clinical variations and implications for counseling. Int J Cancer 131(2):367–376 Sasako M, Sakuramoto S, Katai H, Kinoshita T, Furukawa H, Yamaguchi T, Nashimoto A, Fujii M, Nakajima T, Ohashi Y, 2011, Five-year outcomes of a randomized phase III trial comparing adjuvant chemotherapy with S-1 versus surgery alone in stage II or III gastric cancer. J Clin Oncol 29(33):4387–4393 Kim DH, Oh SJ, Oh CA, Choi MG, Noh JH, Sohn TS, Bae JM, Kim S, 2011, The relationships between perioperative CEA, CA 19-9, and CA 72-4 and recurrence in gastric cancer patients after curative radical gastrectomy. J Surg Oncol 104(6):585–591 Lai KW, Koh KX, Loh M, Tada K, Subramaniam MM, Lim XY, Vaithilingam A, Salto-Tellez M, Iacopetta B, Ito Y, 2010, MicroRNA-130b regulates the tumour suppressor RUNX3 in gastric cancer. Eur J Cancer 46(8):1456–1463 Fan X-y, Hu X-l, Han T-m, Wang N-n, Zhu Y-m, Hu W, Ma Z-h, Zhang C-j, Xu X, Ye Z-y, 2011, Association between RUNX3 promoter methylation and gastric cancer: a meta-analysis. BMC Gastroenterol 11(1):92 Chen Y,Wei X, Guo C, Jin H, Han Z, Han Y, Qiao T,Wu K, Fan D, 2011, Runx3 suppresses gastric cancer metastasis through inactivation of MMP9 by upregulation of TIMP-1. Int J Cancer 129(7): 1586–1598 Lin F, Liu Y, Lai C, Shan Y, Cheng H, Hsu P, Lee C, Lee Y, Wang H, Wang C, 2012, RUNX3-mediated transcriptional inhibition of Akt suppresses tumorigenesis of human gastric cancer cells. Oncogene 31(39):4302–4316 Ha Thi HT, Lim H-S, Kim J, Kim Y-M, Kim H-Y, Hong S, 2013, Transcriptional and post-translational regulation of Bim is essential for TGF-β and TNF-α-induced apoptosis of gastric cancer cell. Biochim Biophys Acta 1830(6):3584–3592 Emmanuel C, Huynh M, Matthews J, Kelly E, Zoellner H, 2013, TNF-α and TGF-β synergistically stimulate elongation of human endothelial cells without transdifferentiation to smooth muscle cell phenotype. Cytokine 61(1):38–40 Suganuma M, Watanabe T, Yamaguchi K, Takahashi A, Fujiki H, 2012, Human gastric cancer development with TNF-α-inducing protein secreted from Helicobacter pylori. Cancer Lett 322(2):133– 138 Guo S-L, Ye H, Teng Y, Wang Y-L, Yang G, Li X-B, Zhang C, Yang X, Yang Z-Z, Yang X, 2013, Akt-p53-miR-365-cyclin D1/ cdc25A axis contributes to gastric tumorigenesis induced by PTEN deficiency. Nat Commun 4:2544 Wang Q,Wen Y-G, Li D-P, Xia J, Zhou C-Z, Yan D-W, Tang H-M, Peng Z-H, 2012, Upregulated INHBA expression is associated with poor survival in gastric cancer. Med Oncol 29(1):77–83 Gautier L, Cope L, Bolstad BM, Irizarry RA, 2004, Affy—analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 20(3):307–315 Diboun I, Wernisch L, Orengo CA, Koltzenburg M, 2006, Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma. BMC Genomics 7(1):252 Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M, Roth A, Lin J, Minguez P, Bork P, von Mering C, 2013, STRING v9. 1: protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res 41(D1):D808–D815 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, 2005, Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102(43):15545–15550 Wingender E, 2008, The TRANSFAC project as an example of framework technology that supports the analysis of genomic regulation. Brief Bioinform 9(4):326–332 Forman D, SierraMS, 2014, Introduction: the current and projected global burden of gastric cancer. In: IARC Helicobacter pylori Working Group, ed, Helicobacter pylori eradication as a strategy for preventing gastric cancer [Internet]. International Agency for Research on Cancer, IARC Working Group Reports, No. 8), Lyon, p 5–15 [cited 2014 Apr 25]. Available from: http://www.iarc. fr/en/publications/pdfs-online/wrk/wrk8/index.php Postel-Vinay S, Veron AS, Tirode F, Pierron G, Reynaud S, Kovar H, Oberlin O, Lapouble E, Ballet S, Lucchesi C, 2012, Common variants near TARDBP and EGR2 are associated with susceptibility to Ewing sarcoma. Nat Genet 44(3):323–327 Feng D, Ye X, Zhu Z et al, 2015, Comparative transcriptome analysis between metastatic and non-metastatic gastric cancer reveals potential biomarkers. Mol Med Rep 11(1):386–392 Wu Q, Jin H, Yang Z, Luo G, Lu Y, Li K, Ren G, Su T, Pan Y, Feng B, 2010, MiR-150 promotes gastric cancer proliferation by negatively regulating the pro-apoptotic gene EGR2. Biochem Biophys Res Commun 392(3):340–345 Li X, Zhang Z, Yu M, Li L, Du G, Xiao W, Yang H, 2013, Involvement of miR-20a in promoting gastric cancer progression by targeting early growth response 2, EGR2). Int J Mol Sci 14(8): 16226–16239 Fu Z, Malureanu L, Huang J, Wang W, Li H, Van Deursen JM, Tindall DJ, Chen J, 2008, Plk1-dependent phosphorylation of FoxM1 regulates a transcriptional programme required for mitotic progression. Nat Cell Biol 10(9):1076–1082 Okada K, Fujiwara Y, Takahashi T, Nakamura Y, Takiguchi S, Nakajima K, Miyata H, Yamasaki M, Kurokawa Y, Mori M, 2013, Overexpression of forkhead box M1 transcription factor, FOXM1, is a potential prognostic marker and enhances chemoresistance for docetaxel in gastric cancer. Ann Surg Oncol 20(3):1035–1043 Li X, Yao R, Yue L, Qiu W, Qi W, Liu S, Yao Y, Liang J, 2014, FOXM1 mediates resistance to docetaxel in gastric cancer via upregulating stathmin. J Cell Mol Med 18(5):811–823 Qi W, Li X, Zhang Y, Yao R, Qiu W, Tang D, Liang J, 2014, Overexpression of her-2 upregulates FoxM1 in gastric cancer. Int J Mol Med 33(6):1531–1538 Li Z, Ying X, Chen H et al, 2014, MicroRNA-194 inhibits the epithelial-mesenchymal transition in gastric cancer cells by targeting FoxM1. Dig Dis Sci 59(9):2145–2152 Niimi T, Nagashima K,Ward JM, Minoo P, Zimonjic DB, Popescu NC, Kimura S, 2001, Claudin-18, a novel downstream target gene for the T/EBP/NKX2. 1 homeodomain transcription factor, encodes lung-and stomach-specific isoforms through alternative splicing. Mol Cell Biol 21(21):7380–7390 Sanada Y, Oue N, Mitani Y, Yoshida K, Nakayama H, Yasui W, 2006, Down-regulation of the claudin-18 gene, identified through serial analysis of gene expression data analysis, in gastric cancer with an intestinal phenotype. J Pathol 208(5):633–642 D’Annibale S, Kim J, Magliozzi R, Low TY, Mohammed S, Heck AJ, Guardavaccaro D, 2014, Proteasome-dependent degradation of transcription factor activating enhancer-binding protein 4, TFAP4, controls mitotic division. J Biol Chem 289(11):7730–7737 Xinghua L, Bo Z, Yan G, Lei W, Changyao W, Qi L, Lin Y, Kaixiong T, Guobin W, Jianying C, 2012, The overexpression of AP-4 as a prognostic indicator for gastric carcinoma. Med Oncol 29(2):871–877 Zhao Y, Zhou T, Li A, Yao H, He F,Wang L, Si J, 2009, A potential role of collagens expression in distinguishing between premalignant and malignant lesions in stomach. Anat Rec 292(5):692–700 Yin Y, Zhao Y, Li A-Q, Si J-M, 2009, Collagen: a possible prediction mark for gastric cancer. Med Hypotheses 72(2):163–165 Liang Z, Zeng X, Gao J, Wu S, Wang P, Shi X, Zhang J, Liu T, 2008, Analysis of EGFR, HER2, and TOP2A gene status and chromosomal polysomy in gastric adenocarcinoma from Chinese patients. BMC Cancer 8(1):363 Varis A,WolfM,Monni O, Vakkari M-L, Kokkola A, Moskaluk C, Frierson H, Powell SM, Knuutila S, Kallioniemi A, 2002, Targets of gene amplification and overexpression at 17q in gastric cancer. Cancer Res 62(9):2625–2629 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 785 EP 791 DI 10.1007/s12253-016-0159-1 PG 7 ER PT J AU Kalantari, E Asgari, M Nikpanah, S Salarieh, N Lari, HAM Madjd, Z AF Kalantari, Elham Asgari, Mojgan Nikpanah, Seyedehmoozhan Salarieh, Naghme Lari, Hossein Asadi Mohammad Madjd, Zahra TI Co-Expression of Putative Cancer Stem Cell Markers CD44 and CD133 in Prostate Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Tissue microarray; Immunohistochemistry; CD44; CD133 ID Prostate cancer; Tissue microarray; Immunohistochemistry; CD44; CD133 AB Cancer stem cells (CSCs) are the main players of prostate tumorigenesis thus; characterization of CSCs can pave the way for understanding the early detection, drug resistance, metastasis and relapse. The current study was conducted to evaluate the expression level and clinical significance of the potential CSC markers CD44 and CD133 in a series of prostate tissues. One hundred and forty eight prostate tissues composed of prostate cancer (PCa), high-grade prostatic intraepithelial neoplasia (HGPIN), and benign prostate hyperplasia (BPH) were immunostained for the putative CSC markers CD44 and CD133. Subsequently, the correlation between the expression of these markers and the clinicopathological variables was examined. A higher level of CD44 expression was observed in 42% of PCa, 57% of HGPIN, and 42% BPH tissues. In the case of CD133 expression PCa, HGPIN, and BPH samples demons t r a ted high immunoreactivity in 46%, 43%, and 42% of cells, respectively. Statistical analysis showed an inverse significant correlation between CD44 expression with Gleason score of PCa (P = 0.02), while no significant correlation was observed between CD133 expression and clinicopathological parameters. A significant reciprocal correlation was observed between the expression of two putative CSC markers CD44 and CD133 in PCa specimens while not indicating clinical significance. Further clinical investigation is required to consider these markers as targets of new therapeutic strategies for PCa. C1 [Kalantari, Elham] Iran University of Medical Sciences (IUMS), Oncopathology Research Center, Hemmat Street (Highway), Next to Milad Tower, 14496-14530 Tehran, Iran. [Asgari, Mojgan] Iran University of Medical Sciences (IUMS), Oncopathology Research Center, Hemmat Street (Highway), Next to Milad Tower, 14496-14530 Tehran, Iran. [Nikpanah, Seyedehmoozhan] Iran University of Medical Sciences (IUMS), Faculty of Medicine, Department of PathologyTehran, Iran. [Salarieh, Naghme] Iran University of Medical Sciences (IUMS), Faculty of Medicine, Department of PathologyTehran, Iran. [Lari, Hossein Asadi Mohammad] University of British Columbia (UBC), Faculty of Medicine, Department of Cellular, Anatomical and Physiological SciencesVancouver, BC, Canada. [Madjd, Zahra] Iran University of Medical Sciences (IUMS), Oncopathology Research Center, Hemmat Street (Highway), Next to Milad Tower, 14496-14530 Tehran, Iran. RP Asgari, M (reprint author), Iran University of Medical Sciences (IUMS), Oncopathology Research Center, 14496-14530 Tehran, Iran. EM mojgan_asgari@yahoo.com CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65:87–108 Jemal A, Siegel R, Xu J,Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60:277–300 Hurt EM, Kawasaki BT, Klarmann GJ, Thomas SB, Farrar WL, 2008, CD44+ CD24− prostate cells are early cancer progenitor/stem cells that provide a model for patients with poor prognosis. Br J Cancer 98:756–765 van den Hoogen C, van der Horst G, Cheung H, Buijs JT, Lippitt JM, Guzman-Ramirez N, Hamdy FC, Eaton CL, Thalmann GN, Cecchini MG, 2010, High aldehyde dehydrogenase activity identifies tumor-initiating and metastasis-initiating cells in human prostate cancer. Cancer Res 70:5163–5173 Dubrovska A, Elliott J, Salamone RJ, Kim S, Aimone LJ, Walker JR, Watson J, Sauveur-Michel M, Garcia-Echeverria C, Cho CY, 2010, Combination therapy targeting both tumor-initiating and differentiated cell populations in prostate carcinoma. Clin Cancer Res 16:5692–5702 Rangwala F, Omenetti A, Diehl AM, 2010, Cancer stem cells: repair gone awry? J Oncol 2011:465343 Reyes EE, Kunovac SK, Duggan R, Kregel S, Griend DJV, 2013, Growth kinetics of CD133-positive prostate cancer cells. Prostate 73:724–733 Ugolkov AV, Eisengart LJ, Luan C, Yang XJ, 2011, Expression analysis of putative stem cell markers in human benign and malignant prostate. Prostate 71:18–25 AM DM,WG N, AK M, DS C, 1998, Stem cell features of benign and malignant prostate epithelial cells. J Urol 160:2381–2392 Tirino V, Desiderio V, Paino F, De Rosa A, Papaccio F, La Noce M, Laino L, De Francesco F, Papaccio G, 2013, Cancer stem cells in solid tumors: an overview and new approaches for their isolation and characterization. FASEB J 27:13–24 Marhaba R, ZollerM(2004, CD44 in cancer progression: adhesion, migration and growth regulation. J Mol Histol 35:211–231 Patrawala L, Calhoun-Davis T, Schneider-Broussard R, Tang DG, 2007, Hierarchical organization of prostate cancer cells in xenograft tumors: the CD44+ α2β1+ cell population is enriched in tumor-initiating cells. Cancer Res 67:6796–6805 Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ, 2005, Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 65:10946–10951 Marker PC 2007 Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic andmetastatic progenitor cells: Patrawala L, Calhoun T, Schneider-Broussard R, Li H, Bhatia B, Tang S, Reilly JG, Chandra D, Zhou J, Claypool K, Coghlan L, Tang DG, Department of Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park- Research Division, Smithville, TX. In: Urologic Oncology: Seminars and Original Investigations,. vol 3. Elsevier, pp 277–278 Nagabhushan M, Pretlow TG, Guo Y-J, Amini SB, Pretlow TP, Sy M-S, 1996, Altered expression of CD44 in human prostate cancer during progression. Am J Clin Pathol 106:647–651 Kallakury BV, Yang F, Figge J, Smith KE, Kausik SJ, Tacy NJ, Fisher HA, Kaufman R, Figge H, Ross JS, 1996, Decreased levels of CD44 protein and mRNA in prostate carcinoma: correlation with tumor grade and ploidy. Cancer 78:1461–1469 Simon RA, di Sant’Agnese PA, Huang L-S, Xu H, Yao JL, Yang Q, Liang S, Liu J, Yu R, Cheng L, 2009, CD44 expression is a feature of prostatic small cell carcinoma and distinguishes it from its mimickers. Hum Pathol 40:252–258 Tirino V, Desiderio V, d’Aquino R, De Francesco F, Pirozzi G, Graziano A, Galderisi U, Cavaliere C, De Rosa A, Papaccio G, 2008, Detection and characterization of CD133+ cancer stem cells in human solid tumours. PLoS One 3:e3469 Tirino V, Camerlingo R, Franco R, Malanga D, La Rocca A, Viglietto G, Rocco G, Pirozzi G, 2009, The role of CD133 in the identification and characterisation of tumour-initiating cells in nonsmall- cell lung cancer. Eur J Cardiothorac Surg 36:446–453 Sabet MN, Rakhshan A, Erfani E, Madjd Z, 2014, Co-expression of putative cancer stem cell markers, CD133 and nestin, in skin tumors. Asian Pac J Cancer Prev 15:8161–8169 Mehrazma M, Madjd Z, Kalantari E, Panahi M, Hendi A, Shariftabrizi A, 2013, Expression of stem cell markers, CD133 and CD44, in pediatric solid tumors: a study using tissue microarray. Fetal Pediatr Pathol 32:192–204 Wognum AW, Eaves AC, Thomas TE, 2003, Identification and isolation of hematopoietic stem cells. Arch Med Res 34: 461–475 Richardson GD, Robson CN, Lang SH, Neal DE, Maitland NJ, Collins AT, 2004, CD133, a novel marker for human prostatic epithelial stem cells. J Cell Sci 117:3539–3545 Miki J, Furusato B, Li H, Gu Y, Takahashi H, Egawa S, Sesterhenn IA, McLeod DG, Srivastava S, Rhim JS, 2007, Identification of pPutative sStem cCell mMarkers, CD133 and CXCR4, in hTERT– Immortalized Primary Nonmalignant and Malignant Tumor- Derived Human Prostate Epithelial Cell Lines and in Prostate Cancer Specimens. Cancer Res 67:3153–3161 Epstein JI, Allsbrook WC Jr, Amin MB, Egevad LL, 2005, The 2005 International Society of Urological Pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol 29:1228–1242 Cheng L, Montironi R, Bostwick DG, Lopez-Beltran A, Berney DM, 2012, Staging of prostate cancer. Histopathology 60:87– 117., DOI 10.1111/j.1365-2559.2011.04025.x Madjd Z, Akbari ME, Zarnani AH, Khayamzadeh M, Kalantari E, Mojtabavi N, 2013, Expression of EMSY, a novel BRCA2-link protein, is associated with lymph node metastasis and increased tumor size in breast carcinomas. Asian Pac J Cancer Prev 15: 1783–1789 Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB, 2003, Identification of a cancer stem cell in human brain tumors. Cancer Res 63:5821–5828 Kalantari E, Saadi FH, Asgari M, Shariftabrizi A, Roudi R, Madjd Z, 2016, Increased expression of ALDH1A1 in prostate cancer is correlated with tumor aggressiveness: a tissue microarray study of Iranian patients. Appl ImmunohistochemMol Morphol AIMMOff Pu b l Soc Appl Immunohi s t o c h e m . doi :10. 1 0 97 /PAI.0000000000000343 Camp RL, Charette LA, Rimm DL, 2000, Validation of tissue microarray technology in breast carcinoma. Lab Investig 80: 1943–1949 Roudi R, Korourian A, Shariftabrizi A, Madjd Z, 2015, Differential expression of cancer stem cell markers ALDH1 and CD133 in various lung cancer subtypes. Cancer Invest 33:294–302 Madjd Z, Ramezani B, Molanae S, Asadi-Lari M, 2012, High expression of stem cell marker ALDH1 is associated with reduced BRCA1 in invasive breast carcinomas. Asian Pac J Cancer Prev 13: 2973–2978 Walker R, 2006, Quantification of immunohistochemistry—issues concerning methods, utility and semiquantitative assessment I. Histopathology 49:406–410 Nagel H, Laskawi R,Wahlers A, Hemmerlein B, 2004, Expression of matrix metalloproteinases MMP-2, MMP-9 and their tissue inhibitors TIMP-1,-2, and-3 in benign and malignant tumours of the salivary gland. Histopathology 44:222–231 Madjd Z, Pinder S, Paish C, Ellis I, Carmichael J, Durrant L, 2003, Loss of CD59 expression in breast tumours correlates with poor survival. J Pathol 200:633–639 Mohsenzadegan M, Madjd Z, Asgari M, Abolhasani M, Shekarabi M, Taeb J, Shariftabrizi A, 2013, Reduced expression of NGEP is associated with high-grade prostate cancers: a tissue microarray analysis. Cancer Immunol Immunother 62:1609–1618 Taeb J, Asgari M, Abolhasani M, FarajollahiMM, Madjd Z, 2014, Expression of prostate stem cell antigen, PSCA, in prostate cancer: a tissue microarray study of Iranian patients. Pathology-research and. Practice 210:18–23 Kerfoot C, Huang W, Rotenberg SA, 2004, Immunohistochemical analysis of advanced human breast carcinomas reveals downregulation of protein kinase Cα. J Histochem Cytochem 52:419–422 McCarty K Jr, Miller L, Cox E, Konrath J, McCarty K Sr, 1985, Estrogen receptor analyses. Correlation of biochemical and immunohistochemical methods using monoclonal antireceptor antibodies. Arch Pathol Lab Med 109:716–721 Sharma BK, Manglik V, Elias EG, 2010, Immuno-expression of human melanoma stem cell markers in tissues at different stages of the disease. J Surg Res 163:e11–e15 Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, Bronner-Fraser M, Kornblum HI, 2003, Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci 100:15178–15183 Chang B, Liu G, Xue F, Rosen DG, Xiao L, Wang X, Liu J, 2009, ALDH1 expression correlates with favorable prognosis in ovarian cancers. Mod Pathol 22:817–823 Nguyen LV, Vanner R, Dirks P, Eaves CJ, 2012, Cancer stem cells: an evolving concept. Nat Rev Cancer 12:133–143 Sullivan JP, Spinola M, Dodge M, Raso MG, Behrens C, Gao B, Schuster K, Shao C, Larsen JE, Sullivan LA, 2010, Aldehyde dehydrogenase activity selects for lung adenocarcinoma stem cells dependent on notch signaling. Cancer Res 70:9937–9948 Castellon EA, Valenzuela R, Lillo J, Castillo V, Contreras HR, Gallegos I, Mercado A, Huidobro C, 2012, Molecular signature of cancer stem cells isolated from prostate carcinoma and expression of stem markers in different Gleason grades and metastasis. Biol Res 45:297–305 Jaworska D, Krol W, Szliszka E, 2015, Prostate cancer stem cells: research advances. Int J Mol Sci 16:27433–27449 Patrawala L, Calhoun T, Schneider-Broussard R, Li H, Bhatia B, Tang S, Reilly J, Chandra D, Zhou J, Claypool K, 2006, Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene 25:1696–1708 Paradis V, Eschwege P, Loric S, Dumas F, Ba N, Benoit G, Jardin A, Bedossa P, 1998, De novo expression of CD44 in prostate carcinoma is correlated with systemic dissemination of prostate cancer. J Clin Pathol 51:798–802 De Marzo AM, Bradshaw C, Sauvageot J, Epstein JI, Miller GJ, 1998, CD44 and CD44v6 downregulation in clinical prostatic carcinoma: relation to Gleason grade and cytoarchitecture. Prostate 34: 162–168 Corbeil D, Roper K, Fargeas CA, Joester A, Huttner WB, 2001, Prominin: a story of cholesterol, plasma membrane protrusions and human pathology. Traffic 2:82–91 Wang C-C, De Marzo AM, Lotan TL, Epstein JI, 2015, Overlap of CD44 expression between prostatic small cell carcinoma and acinar adenocarcinoma. Hum Pathol 46:554–557 Trerotola M, Rathore S, Goel HL, Li J, Alberti S, Piantelli M, Adams D, Jiang Z, Languino LR, 2010, CD133, trop-2 and alpha2beta1 integrin surface receptors as markers of putative human prostate cancer stem cells. Am J Transl Res 2:135–144 Bussolati B, Bruno S, Grange C, Buttiglieri S, Deregibus MC, Cantino D, Camussi G, 2005, Isolation of renal progenitor cells from adult human kidney. Am J Pathol 166:545–555 Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, Conticello C, Ruco L, Peschle C, De Maria R, 2008, Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ 15:504–514 O’Brien CA, Pollett A, Gallinger S, Dick JE, 2007, A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445(7123):106–110 Ma S, Lee T, Zheng B, Chan K,Guan X(2008)CD133+ HCC cancer stem cells confer chemoresistance by preferential expression of the Akt/PKB survival pathway. Oncogene 27:1749–1758 Liu G, Yuan X, Zeng Z, Tunici P, Ng H, Abdulkadir IR, Lu L, Irvin D, Black KL, John SY, 2006, Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer 5:67 Vatansever H, Gumus B, Aydogdu O, Sivrikoz O, Turkoz-Uluer E, Kivanc M, Atesci Y, Bugdayci H, 2014, The role of stem/ progenitor cells and Wnt/β-catenin signaling pathway in the patients with prostate cancer. Minerva urologica e nefrologica = The Italian journal of urology and nephrology 66:249–255 Fargeas C, Huttner W, Corbeil D, 2007, Nomenclature of prominin-1, CD133, splice variants–an update. Tissue Antigens 69:602–606 Irollo E, Pirozzi G, 2013, CD133: to be or not to be, is this the real question. Am J Transl Res 5:563–581 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 793 EP 802 DI 10.1007/s12253-016-0169-z PG 10 ER PT J AU Di Michele, J Rotondo, F Kovacs, K Syro, VL Yousef, MG Cusimano, DM Di Ieva, A AF Di Michele, Joseph Rotondo, Fabio Kovacs, Kalman Syro, V Luis Yousef, M George Cusimano, D Michael Di Ieva, Antonio TI Vasculogenic Mimicry in Clinically Non-functioning Pituitary Adenomas: a Histologic Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Biomarkers; Pathology; Pituitary adenoma; Microvascularity; Vasculogenic mimicry ID Biomarkers; Pathology; Pituitary adenoma; Microvascularity; Vasculogenic mimicry AB The term "vasculogenic mimicry" (VM) refers to the phenomenon in which vascular-like channels, which are not lined by endothelial cells, are formed in tumors. Since its discovery in 1999, it has been observed in several tumor types and is proposed to provide blood perfusion to tumors in absence of coapted or neo-angiogenic blood vessels. Pituitary tumors are generally slow growing, benign adenomas which are less vascularized than the normal pituitary gland. To date, VM in pituitary adenomas has not been described. In this histological study, we assessed the presence of VM in a series of surgically resected clinically non-functioning pituitary adenomas (NFPAs) using CD34 and Periodic Acid-Schiff (PAS) double staining. To identify VM, slides were assessed for the presence of CD34- negative and PAS-positive channels indicating that they were not lined by endothelial cells. The histological staining pattern suggestive of VM was noted in 22/49 (44.9%) of the specimens studied. VM was observed in both recurring and non-recurring NFPAs. The incidence of VM present varied from case to case and within groups. There was no association between the presence of VM and gender, tumor size, Ki-67 index, recurrence or cavernous sinus invasion. VM was not noted in cases of nontumorous pituitaries. Our findings suggest the existence of a complementary perfusion system in pituitary adenomas, implying potential clinical implications with respect to response to therapy and clinical course. Further research is warranted to confirm the presence of VM in pituitary adenomas to elucidate its clinical relevance in patients diagnosed with a pituitary adenoma. C1 [Di Michele, Joseph] St. Michael’s Hospital, Department of SurgeryToronto, ON, Canada. [Rotondo, Fabio] St. Michael’s Hospital, Department of Laboratory Medicine, 30 Bond Street, M5B1W8 Toronto, ON, Canada. [Kovacs, Kalman] St. Michael’s Hospital, Department of Laboratory Medicine, 30 Bond Street, M5B1W8 Toronto, ON, Canada. [Syro, V Luis] Hospital Pablo Tobon Uribe and Clinica Medellin, Department of NeurosurgeryMedellin, Colombia. [Yousef, M George] St. Michael’s Hospital, Department of Laboratory Medicine, 30 Bond Street, M5B1W8 Toronto, ON, Canada. [Cusimano, D Michael] St. Michael’s Hospital, Department of SurgeryToronto, ON, Canada. [Di Ieva, Antonio] Macquarie University Hospital, Australian School of Advanced Medicine, Department of NeurosurgerySydney, Australia. RP Rotondo, F (reprint author), St. Michael’s Hospital, Department of Laboratory Medicine, M5B1W8 Toronto, Canada. EM rotondof@smh.ca CR Maniotis A, Folberg R, Hess A et al, 1999, Vascular Channel formation by humanmelanoma cells in vivo and in vitro:Vasculogenic mimicry. Am J Pathol 155:739–752 Shirakawa K, Wakasugi H, Heike Y et al, 2002, Vasculogenic mimicry and pseudo-comedo formation in breast cancer. Int J Cancer 99:821–828 Yue W, Chen Z, 2005, Does Vasculogenic mimicry exist in astrocytoma? J Histochem Cytochem 53:997–1002 Shaifer C, Huang J, Lin P, 2010, Glioblastoma cells incorporate into tumor vasculature and contribute to vascular radioresistance. Int J Cancer 127:2063–2075 Liu X, Zhang Q, Mu Y et al, 2011, Clinical significance of vasculogenic mimicry in human gliomas. J Neuro-Oncol 105: 173–179 Liu Z, Li Y, Zhao W, Ma Y, Yang X, 2011, Demonstration of vasculogenic mimicry in astrocytomas and effects of Endostar on U251 cells. Pathol Res Pract 207:645–651 Scully S, Francescone R, Faibi sh M e t a l, 2012, Transdifferentiation of glioblastoma stem-like cells intomural cells drives Vasculogenic mimicry in glioblastomas. J Neurosci 32: 12950–12960 Smith S, Tilly H, Ward J et al, 2012, CD105, Endoglin, exerts prognostic effects via its role in the microvascular niche of paediatric high grade glioma. Acta Neuropathol 124:99–110 Liu W, Meng M, Zhang B et al, 2015, Dehydroeffusol effectively inhibits human gastric cancer cell-mediated vasculogenic mimicry with low toxicity. Toxicol Appl Pharmacol 287:98–110 Wagenblast E, Soto M, Gutierrez-Angel S et al, 2015, A model of breast cancer heterogeneity reveals vascular mimicry as a driver of metastasis. Nature 520:358–362 Cao Z, Bao M, Miele L, Sarkar FH, Wang Z, Zhou Q, 2013, Tumour vasculogenic mimicry is associated with poor prognosis of human cancer patients: a systemic review and meta-analysis. Europ J Cancer 49:3914–3923 Carmeliet P, Jain RK, 2011, Molecular mechanisms and clinical applications of angiogenesis. Nature 473:298–307 El Hallani S, Boisselier B, Peglion F et al, 2010, A new alternative mechanism in glioblastoma vascularization: tubular vasculogenic mimicry. Brain 133:973–982 Soda Y, Myskiw C, Rommel A, Verma IM, 2013, Mechanisms of neovascularization and resistance to anti-angiogenic therapies in glioblastoma multiforme. J Mol Med, Berl, 91:439–448 Gartner LP, Hiatt JL, 1994, Color atlas of histology. Williams & Wilkins, Baltimore Asa SL, 1998, Tumors of the pituitary gland. In: Rosai J, ed, Atlas of tumor pathology, 3rd Series Fascicle, vol 22. Armed Forces Institute of Pathology, Washington, pp 1–214 Di Ieva A, Weckman A, Di Michele J et al, 2013, Microvascular morphometrics of the hypophysis and pituitary tumors: from bench to operating theatre. Microvasc Res 89:7–14 Rotondo F, Scheithauer BW, Kovacs K, Bell DC, 2009, Rab 3B immunoexpression in human pituitary adenomas. Appl Immunohistochem Mol Morphol 17:185–188 Kovacs K, Scheithauer BW, Horvath E, Lloyd RV, 1996, The World Health Organization classification of adenohypophysial neoplasms. A proposed five-tier scheme. Cancer 78:502–510 Asa SL, 2011, Tumors of the Pituitary Gland AFIP Fascicle 15/4, 1st edn. p 275. ISBN: 9781933477152 Folberg R, HendrixMJ, Maniotis AJ, 2000, Vasculogenic mimicry and tumor angiogenesis. Am J Pathol 156:361–381 Risau W, Lemmon V, 1988, Changes in the vascular extracellular matrix during embryonic vasculogenesis and angiogenesis. Dev Biol 125:441–450 Risau W, Flamme I, 1995, Vasculogenesis. Annu Rev Cell Dev Biol 11:73–91 Ferguson JE, Kelley RW, Patterson C, 2005, Mechanisms of endothelial differentiation in embryonic vasculogenesis. Arterioscler Thromb Vasc Biol 25:2246–2254 Folkman J, 1971, Tumor angiogenesis. Therapeutic implications. N Engl J Med 285:1182–1186 Pezzella F, Pastorino U, Tagliabue E et al, 1997, Non-small-cell lung carcinoma tumor growth without morphological evidence of neo-angiogenesis. Am J Pathol 151:1417–1423 Stessels F, Salgado G, van den Eynden I et al, 2004, Breast adenocarcinoma liver metastases, in contrast to colorectal cancer liver metastases, display a non-angiogenic growth pattern that preserves the stroma and lacks hypoxia. Br J Cancer 90:1429–1436 Donnem T, Hu J, Ferguson M et al, 2013, Vessel co-option in primary human tumors and metastases: an obstacle to effective anti-angiogenic treatment? Cancer Med 2:427–436 Leenders WP, Kusters B, de Waal RM, 2002, Vessel co-option: how tumors obtain blood supply in the absence of sprouting angiogenesis. Endothelium 9:83–87 Djonov V, Baum O, Burri PH, 2003, Vascular remodeling by intussusceptive angiogenesis. Cell Tiss Res 314:107–117 Gianni-Barrera R, Trani M, Reginato S, BanfiA, 2011, To sprout or to split? VEGF, notch and vascular morphogenesis. Biochem Soc Trans 39:1644–1648 Sun B, Zhang S, Zhang D et al, 2006, Vasculogenic mimicry is associated with high tumor grade, invasion and metastasis, and short survival in patients with hepatocellular carcinoma. Oncol Rep 16:693–698 Sun B, Zhang D, Zhang S, Zhang W, Guo H, Zhao X, 2007, Hypoxia influences vasculogenic mimicry channel formation and tumor invasion-related protein expression in melanoma. Cancer Lett 249:188–197 Sun B, Qie S, Zhang S et al, 2008, Role and mechanism of vasculogenic mimicry in gastrointestinal stromal tumors. Hum Pathol 39:444–451 Tang HS, Feng YJ, Yao LQ, 2009, Angiogensis, vasculogenesis and vasculogenic mimicry in ovarian cancer. Int J Gynecol Cancer 19:605–610 Liu R, Yang K, Meng C, Zhang Z, Xu Y, 2012, Vasculogenic mimicry is a marker of poor prognosis in prostate cancer. Cancer Biol Ther 13:527–533 Larson AR, Lee CW, Lezcano C et al, 2014, Melanoma spheroid formation involves laminin-associated vasculogenic mimicry. Am J Pathol 184:71–78 Yang JP, Liao YD, Mai DMet al, 2016, Tumor vasculogenic mimicry predicts poor prognosis in cancer patients: a meta-analysis. Angiogenesis 19:191–200 Ferreira JE, de Mello PA, de Magalhaes AV, Botelho CH, Naves LA, Nose V, Scmitt F, 2005, Non-functioning pituitary adenomas: clinical features and immunohistochemistry. Arg Neuropsiquiatr 63:1070–1078 Kohlberger PD, Obermair A, Sliutz G et al, 1996, Quantitative immunohistochemistry of factor VIII-related antigen in breast carcinoma: a comparison of computer-assisted image analysis with established counting methods. Am J Clin Pathol 105:705–710 Jugenburg M, Kovacs K, Stefaneanu L, Scheithauer BW, 1995, Vasculature in nontumorous hypophyses, pituitary adenomas, and carcinomas: a quantitative morphologic study. Endocr Pathol 6: 115–124 Turner HE, Nagy Z, Gatter KC, Esiri MM, Harris AL, Wass JAH, 2000, Angiogenesis in pituitary adenomas and the normal pituitary gland. J Clin Endocrinol Metab 85:1159–1162 Di Ieva A,Grizzi F, Ceva-GrimaldiGet al, 2007, Fractal dimension as a quantitator of the microvasculature of normal and adenomatous pituitary tissue. J Anat 211:673–680 Ruffini F, Graziani G, Levati L, Tentori L, D'Atri S, Lacal P, 2014, Cilengitide downmodulates invasiveness and vasculogenic mimicry of neuropilin 1 expressing melanoma cells through the inhibition of αvβ5 integrin. Int J Cancer 136:E545–E558 van der Schaft DW, Seftor RE, Seftor EA et al, 2004, Effects of angiogenesis inhibitors on vascular network formation by human endothelial and melanoma cells. J Natl Cancer Inst 96:1473–1477 Di Ieva A, Rotondo F, Syro LV, Cusimano MD, Kovacs K, 2014, Aggressive pituitary adenomas– diagnosis and emerging treatments. Nat Rev Endocrinol 10:423–475 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 803 EP 809 DI 10.1007/s12253-017-0196-4 PG 7 ER PT J AU Rath-Wolfson, L Bubis, G Shtrasburg, Sh Shvero, A Koren, R AF Rath-Wolfson, Lea Bubis, Golan Shtrasburg, Shmuel Shvero, Asaf Koren, Rumelia TI Seminal Tract Amyloidosis: Synchronous Amyloidosis of the Seminal Vesicles, Deferent Ducts and Ejaculatory Ducts SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Senile seminal vesicle amyloidosis (SSVA); Ejaculatory duct amyloidosis; Spermatic duct amyloidosis; Senile seminal tract amyloidosis (SSTA) ID Senile seminal vesicle amyloidosis (SSVA); Ejaculatory duct amyloidosis; Spermatic duct amyloidosis; Senile seminal tract amyloidosis (SSTA) AB Senile Seminal Vesicle Amyloidosis (SSVA) increases with age. Involvement of the whole seminal tract, i.e. the seminal vesicles, ejaculatory and deferent ducts was first reported by us in the International Symposium on Amyloidosis 1998. Since then we encountered four more cases of SSVA. In all these cases the ejaculatory and deferent ducts were also involved by amyloid. The amyloid was located mostly sub-epithelially, stained positively with Congo red, gave green birefringence under polarized light and was permanganate sensitive, slightly positive for lactoferrin immunostaining and negative for all known amyloid types. In recent years the amyloid was found to be derived from Semenogelin I, a major constituent of the seminal fluid which is present in the epithelial cells of the seminal vesicle and vas deference. This would explain the deposition of amyloid not only in the seminal vesicles but also in the deferent an ejaculatory ducts which transport the seminal fluid. In a review of the literature we found three more articles on SSVA in which the amyloid was not limited to the seminal vesicles alone. We propose to designate this type of amyloid as "Senile seminal Tract Amyloidosis" (SSTA) instead of "Senile Seminal Vesicle Amyloidosis (SSVA)". C1 [Rath-Wolfson, Lea] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. [Bubis, Golan] University of Nicosia, Medicine Program, St. George’s University of London, 93 Agiou Nikolau Street, Egnomi, 2408 Nicosia, Cyprus. [Shtrasburg, Shmuel] Sheba Medical Center, Heller Institute of Medical ResearchTel-Hashomer, Israel. [Shvero, Asaf] Sheba Medical Center, Department of UrologyTel-Hashomer, Israel. [Koren, Rumelia] A, Rabin Medical Center (Golda Campus), Department of PathologyPetach Tikvah, Israel. RP Bubis, G (reprint author), University of Nicosia, Medicine Program, St. George’s University of London, 2408 Nicosia, Cyprus. EM bubisgolan@gmail.com CR Pitkanen P, Westermark P, Cornwell GG 3rd, Murdoch W, 1983, Amyloid of the seminal vesicles. A distinctive and common localized form of senile amyloidosis. Am J Pathol 110:64–69 Fletcher MS, Herzberg Z, Pryor JP, 1981, The etiology and investigation of haemospermia. Br J Urol 53:669–671 Botash RJ, Poster RB, Abraham JL, Makhuli ZM, 1997, Senile seminal vesicle amyloidosis associated with hematospermia: demonstration by endorectal MRI. J Comput Assist Tomogr 21:748–749 Furuya S, Masumori N, Furuya R, Tsukamoto T, Isomura H, Tamakawa M, 2005, Characterization of localized seminal vesicle amyloidosis causing hemospermia: an analysis using immunohistochemistry and magnetic resonance imaging. J Urol 173:1273– 1277 Vandwalle J, Dugardin F, Petit T, Surga N, Paul A, Petit J, 2007, Haemospermia due to seminal vesicle amyloidosis. Treatment by laparoscopic vesiculectomy. A case report. Prog Urol 17:1382– 1384 Kono M, Kurokawa T, Takata M, Komatsu K, TsukaharaK, Kurose N, 2011, Localized amyloidosis of the seminal vesicle: a case report. Hinyokika Kiyo 57:99–101 Jun SY, Kim KR, Cho KS, Ro JY, 2003, Localized amyloidosis of seminal vesicle and vas deferens: report of two cases. J Korean Med Sci 18:447–451 Kaji Y, Sugimura K, Nagaoka S, Ishida T, 1992, Amyloid deposition in seminal vesicles mimicking tumor invasion from bladder cancer: MR findings. J Comput Assist Tomogr 16:989–991 Jager GJ, Ruijter ET, de la Rosette JJ, van de Kaa CA, 1997, Amyloidosis of the seminal vesicles simulating tumor invasion of prostatic carcinoma on endo-rectal MR images. Eur Radiol 7:552–554 Ramchandani P, Schnall MD, LiVolsi VA, Tomaszewski JE, Pollack HM, 1993, Senile amyloidosis of the seminal vesicles mimickingmetastatic spread of prostatic carcinoma onMRimages. AJR Am J Roentgenol 161:99–100 Koren R, Livneh A, Shtrasburg S, Goldwasser B, Linke R, Gal R, 1998, Organ-limited amyloidosis of the deferent ducts, seminal vesicle and ejaculatory ducts. In: Kyle RA, Gertz MA, eds, Amyloid and Amyloidosis. Parthenon, New York, pp 524–526 Koren R, Gur U, Lask D et al, 2010, Histopathological findings in the prostatic urethra evaluated by a new technique for processing radical prostatectomy specimens. Med Con 1:8–16 Wright JR, Calkins E, Humphrey RL, 1977, Potassium permanganate reaction in amyloidosis. A histologic method to assist in differentiating forms of this disease. Lab Investig 36:274–281 Shtrasburg S, Livneh A, Pras M, Weinstein T, Gal R, 1997, Detection of serum amyloid A-derived proteins in formalin-fixed paraffin-embedded tissues: reliability of the method and expansion of its spectrum. Am J Clin Pathol 108:289–294 Unger PD, Wang Q, Gordon RE, Stock R, Stone N, 1997, Localized amyloidosis of the seminal vesicle. Possible association with hormonally treated prostatic adenocarcinoma. Arch Pathol Lab Med 121:1265–1268 Erbersdobler A, Kollermann J, Graefen M, Rocken C, Schlomm T, 2009, Seminal vesicle amyloidosis does not provide any protection from invasion by prostate cancer. BJU Int 103:324–326 Argon A, Symþyr A, Sarsik B et al, 2012, Amyloidosis of seminal vesicles; incidence and pathologic characteristics. Turk Patoloji Derg 28:44–48 Kee KH, Lee MJ, Shen SS et al, 2008, Amyloidosis of seminal vesicles and ejaculatory ducts: a histologic analysis of 21 cases among 447 prostatectomy specimens. Ann Diagn Pathol 12:235–238 Yang Z, Laird A, Monaghan A, SeywrightM, Ahmad I, Leung HY, 2013, Incidental seminal vesicle amyloidosis observed in diagnostic prostate biopsies–are routine investigations for systemic amyloidosis warranted? Asian J Androl 15:149–151 Goldman H, 1963, Amyloidosis of seminal vesicles and vas deference primary and localized cases. Arch Pathol 75:94–98 Buckett WM, Luckas MJ, Gazvani MR, Aird IA, Lewis-Jones DI, 1997, Seminal plasma lactoferrin concentrations in normal and abnormal semen samples. J Androl 18:302–304 Tsutsumi Y, Serizawa A, Hori S, 1996, Localized amyloidosis of the seminal vesicle: identification of lactoferrin immunoreactivity in the amyloid material. Pathol Int 46:491–497 Suess K, Moch H, Epper R, Koller A, Durmuller U, Mihatsch J, 1998, Heterogeneity of seminal vesicle amyloid. Immunohistochemical detection of lactoferrin and amyloid of the prealbumin-transthyretin type. Pathologe 19:115–119 Cornwell GG 3rd, Westermark GT, Pitkanen P, Westermark P, 1992, Seminal vesicle amyloid: the first example of exocrine cell origin of an amyloid fibril precursor. J Pathol 167:297–303 Linke RP, Joswig R, Murphy CL et al, 2005, Senile seminal vesicle amyloid is derived from semenogelin I. J Lab Clin Med 145:187–193 Sharma N, Vishwanath S, Patel BK, 2016, Recombinant human semenogelin-1, Sg1, and Sg1, 1-159, form detergent stable amyloid like aggregates in vitro. Protein Pept Lett 23:87–96 Lundwall A, Olsson AY, Malm J, 2002, Semenogelin I and II, the predominant human seminal plasma proteins, are also expressed in non-genital tissues Mol. Hum Reprod 8:805–810 Sipe JD, Benson MD, Buxbaum JN, Ikeda S, Merlini G, Saraiva MJM, Westermark P, 2014, Nomenclature 2014: Amyloid fibril proteins and clinical classification of the amyloidosis. Amyloid 21:221–224 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 811 EP 814 DI 10.1007/s12253-017-0193-7 PG 4 ER PT J AU Fkih M’hamed, I Privat, M Trimeche, M Penault-Llorca, F Bignon, YJ Kenani, A AF Fkih M’hamed, Insaf Privat, Maud Trimeche, Mounir Penault-Llorca, Frederique Bignon, Yves-Jean Kenani, Abderraouf TI miR-10b, miR-26a, miR-146a And miR-153 Expression in Triple Negative Vs Non Triple Negative Breast Cancer: Potential Biomarkers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MicroRNAs; Human triple negative breast cancer; LNM; Biomarkers ID MicroRNAs; Human triple negative breast cancer; LNM; Biomarkers AB MicroRNAs (miRNAs) are small non-coding RNAs composed of 18–25 nucleotides that can posttranscriptionally regulate gene expression and have key regulatory roles in cancer, acting as both oncogenes and tumor suppressors. About 1000 genes in humans encode miRNAs, which account for approximately 3% of the human genome, and up to 30% of human protein coding genes may be regulated by miRNAs. The objective of this article is to evaluate the expression profile of four miRNAs previously implicated in triple negative breast cancer: miR-10b, miR-26a, miR-146a and miR-153, and to determine their possible interaction in triple negative and non triple negative breast cancer based on clinical outcome and the expression of BRCA1. 24 triplenegative and 13 non triple negative breast cancer cases, were studied by q-RT-PCR and immunohistochemistry to determine the expression of the four studied miRNAs and the BRCA1 protein, respectively. We observed that the BRCA1 protein was absent in 62.5% of the triple negative cases. Besides, the miR-146a and miR-26a were over expressed in triple negative breast cancer. These two miRNAs, miR-10b and miR-153 were significantly associated to lymph node metastases occurrence in triple negative breast carcinoma. All the analyzed microRNAs were not associated with the expression of BRCA1 in our conditions. Our work provides evidence that miR-146a, miR-26a, miR-10b and miR-153 could be defined as biomarkers in triple negative breast cancer to predict lymph node metastases (LNM). C1 [Fkih M’hamed, Insaf] Centre Jean Perrin, Departement of oncogenetics, 63011 Clermont-Ferrand, France. [Privat, Maud] Centre Jean Perrin, Departement of oncogenetics, 63011 Clermont-Ferrand, France. [Trimeche, Mounir] Farhat Hached Hospital, Department of Pathology, 4000 Sousse, Tunisia. [Penault-Llorca, Frederique] University of Auvergne, EA4677 ERTICAClermont-Ferrand, France. [Bignon, Yves-Jean] Centre Jean Perrin, Departement of oncogenetics, 63011 Clermont-Ferrand, France. [Kenani, Abderraouf] University of Monastir, Faculty of Medicine of Monastir, Laboratory of Biochemistry Research unit UR 12ES08 Cell Signaling and Disease, 5019 Monastir, Tunisia. RP Kenani, A (reprint author), University of Monastir, Faculty of Medicine of Monastir, Laboratory of Biochemistry Research unit UR 12ES08 Cell Signaling and Disease, 5019 Monastir, Tunisia. EM raouf.kenani@fmm.rnu.tn CR Kwan JYY, Psarianos P, Bruce JP, Yip KW, Liu F-F, 2016, The complexity of microRNAs in human cancer. J Radiat Res., DOI 10.1093/jrr/rrw009 Gyparaki M-T, Basdra EK, Papavassiliou AG, 2014, MicroRNAs as regulatory elements in triple negative breast cancer. Cancer Lett 354:1–4 van Schooneveld E, Wildiers H, Vergote I, Vermeulen PB, Dirix LY, Van Laere SJ, 2015, Dysregulation of microRNAs in breast cancer and their potential role as prognostic and predictive biomarkers in patient management. Breast Cancer Res 17:21 Arnedos M, Bihan C, Delaloge S, Andre F, 2012, Triple-negative breast cancer: are we making headway at least? Ther Adv Med Oncol 4:195–210 Yadav BS, Chanana P, Jhamb S, 2015, Biomarkers in triple negative breast cancer: a review. World J Clin Oncol 6:252–263 Andres R, Pajares I, Balmana J, Llort G, Ramon Y, Cajal T, Chirivella I et al, 2014, Association of BRCA1 germline mutations in young onset triple-negative breast cancer, TNBC). Clin Transl Oncol 16:280–284 Qiu J, Xue X, Hu C, Xu H, Kou D, Li R et al, 2016, Comparison of clinicopathological features and prognosis in triple-negative and non-triple negative breast cancer. J Cancer 7:167–173 Zaleska K, 2015, miRNA - therapeutic tool in breast cancer? Where are we now? Rep Pract Oncol Radiother 20:79–86 D’Ippolito E, Iorio MV, 2013, MicroRNAs and triple negative breast cancer. Int J Mol Sci 14:22202–22220 Popovska-Jankovic K, Noveski P, Chakalova L, Petrusevska G, Kubelka K, Plaseska-Karanfilska D, 2012, MicroRNAs in breast cancer -our initial results. Balkan J Med Genet 15:87–89 Fkih M’hamed I, Privat M, Ponelle F, Penault-Llorca F, Kenani A, Bignon Y-J, 2015, Identification of miR-10b, miR-26a, miR-146a and miR-153 as potential triple-negative breast cancer biomarkers. Cell Oncol, Dordr, 38:433–442 Kwiatkowski F, Girard M, Hacene K, Berlie J. 2000. [Sem: A suitable statistical software adaptated for research in oncology]. Bull Cancer.;87:715–21 Turner NC, Reis-Filho JS, Russell AM, Springall RJ, Ryder K, Steele D et al, 2007, BRCA1 dysfunction in sporadic basal-like breast cancer. Oncogene 26:2126–2132 Yamashita N, Tokunaga E, Kitao H, HitchinsM, Inoue Y, Tanaka K et al, 2015, Epigenetic inactivation of BRCA1 through promoter hypermethylation and its clinical importance in triple-negative breast cancer. Clin Breast Cancer 15:498–504 Garcia AI, Buisson M, Bertrand P, Rimokh R, Rouleau E, Lopez BS et al, 2011, Down-regulation of BRCA1 expression by miR- 146a and miR-146b-5p in triple negative sporadic breast cancers. EMBO Mol Med 3:279–290 Kumaraswamy E, Wendt KL, Augustine LA, Stecklein SR, Sibala EC, LiD et al, 2015, BRCA1 regulation of epidermal growth factor receptor, EGFR, expression in human breast cancer cells involves microRNA-146a and is critical for its tumor suppressor function. Oncogene 34:4333–4346 Iorio MV, Ferracin M, Liu C-G, Veronese A, Spizzo R, Sabbioni S et al, 2005, MicroRNA gene expression deregulation in human breast cancer. Cancer Res 65:7065–7070 Zhang L, Long X, 2015, Association of BRCA1 promoter methylation with sporadic breast cancers: evidence from 40 studies. Sci Rep 5:17869 Liu P, Tang H, Chen B, He Z, Deng M,Wu M et al, 2015, miR-26a suppresses tumour proliferation and metastasis by targeting metadherin in triple negative breast cancer. Cancer Lett 357:384–392 Stuckrath I, Rack B, JanniW, Jager B, Pantel K, Schwarzenbach H, 2015, Aberrant plasma levels of circulating miR-16, miR-107, miR-130a and miR-146a are associated with lymph node metastasis and receptor status of breast cancer patients. Oncotarget 6:13387– 13401 Rask L, Balslev E, Sokilde R, Hogdall E, Flyger H, Eriksen J, et al.. 2014. Differential expression of miR-139, miR-486 and miR-21 in breast cancer patients sub-classified according to lymph node status. Cell Oncol, Dordr); 37:215–27 ChenW, Cai F, Zhang B, Barekati Z, ZhongXY, 2013, The level of circulating miRNA-10b and miRNA-373 in detecting lymph node metastasis of breast cancer: potential biomarkers. Tumour Biol 34: 455–462 Nakata K, Ohuchida K, Mizumoto K, Kayashima T, Ikenaga N, Sakai H et al, 2011, MicroRNA-10b is overexpressed in pancreatic cancer, promotes its invasiveness, and correlates with a poor prognosis. Surgery 150:916–922 ChenW-J, Zhang E-N, Zhong Z-K, Jiang M-Z, Yang X-F, Zhou DM et al, 2015, MicroRNA-153 expression and prognosis in nonsmall cell lung cancer. Int J Clin Exp Pathol 8:8671–8675 Bao B, Ali S, Banerjee S, Wang Z, Logna F, Azmi AS et al, 2012, Curcumin analogue CDF inhibits pancreatic tumor growth by switching on suppressor microRNAs and attenuating EZH2 expression. Cancer Res 72:335–345 Visani M, de Biase D, Marucci G, Cerasoli S, Nigrisoli E, Bacchi Reggiani ML, et al.. 2014. Expression of 19 microRNAs in glioblastoma and comparison with other brain neoplasia of grades I-III. Mol Oncol. 8:417–30 Goncalves A, Sabatier R, Charafe-Jauffret E, Gilabert M, Provansal M, Tarpin C et al, 2013, Triple-negative breast cancer: histoclinical and molecular features, therapeutic management and perspectives. Bull Cancer 100:453–464 Cossu-Rocca P, Orru S, Muroni MR, Sanges F, Sotgiu G, Ena S et al, 2015, Analysis of PIK3CA mutations and activation pathways in triple negative breast cancer. PLoS One 10:e0141763 Wang J, Zhang C, Chen K, Tang H, Tang J, Song C et al, 2015, ERβ1 inversely correlates with PTEN/PI3K/AKT pathway and predicts a favorable prognosis in triple-negative breast cancer. Breast Cancer Res Treat 152:255–269 Park YH, Jung HH, Ahn JS, Im Y-H, 2013, Statin induces inhibition of triple negative breast cancer, TNBC, cells via PI3K pathway. Biochem Biophys Res Commun 439:275–279 De P, Sun Y, Carlson JH, Friedman LS, Leyland-Jones BR, Dey N, 2014, Doubling down on the PI3K-AKT-mTOR pathway enhances the antitumor efficacy of PARP inhibitor in triple negative breast cancer model beyond BRCA-ness. Neoplasia 16:43–72 Li J, Song Z, Wang Y, Yin Y, Liu Y, Yuan R et al, 2016, Overexpression of SphK1 enhances cell proliferation and invasion in triple-negative breast cancer via the PI3K/AKT signaling pathway. Tumour Biol., DOI 10.1007/s13277-016-4954-9 Phua YW, Nguyen A, Roden DL, Elsworth B, Deng N, Nikolic I et al, 2015, MicroRNA profiling of the pubertal mouse mammary gland identifies miR-184 as a candidate breast tumour suppressor gene. Breast Cancer Res 17:83 Wang F, Li L, Chen Z, ZhuM, Gu Y, 2016)MicroRNA-214 acts as a potential oncogene in breast cancer by targeting the PTEN-PI3K/ Akt signaling pathway. Int J Mol Med 37:1421–1428 Xu J-F, Zhang S-J, Zhao C, Qiu B-S, Gu H-F, Hong J-F et al, 2015, Altered microRNA expression profile in synovial fluid from patients with knee osteoarthritis with treatment of hyaluronic acid. Mol Diagn Ther 19:299–308 Chai Z-T, Zhu X-D, Ao J-Y, Wang W-Q, Gao D-M, Kong J, et al.. 2015. microRNA-26a suppresses recruitment of macrophages by down-regulating macrophage colony-stimulating factor expression through the PI3K/Akt pathway in hepatocellular carcinoma. J Hematol Oncol; 8:56 Song L, Duan P, Guo P, Li D, Li S, Xu Y et al, 2012, Downregulation of miR-223 and miR-153 mediates mechanical stretch-stimulated proliferation of venous smooth muscle cells via activation of the insulin-like growth factor-1 receptor. Arch Biochem Biophys 528:204–211 Chen Z-J,WeiW, Jiang G-M, Liu H,WeiW-D, Yang X et al, 2016, Activation of GPER suppresses epithelial mesenchymal transition of triple negative breast cancer cells via NF-κB signals. MolOncol., DOI 10.1016/j.molonc.2016.01.002 Bhaumik D, Scott GK, Schokrpur S, Patil CK, Campisi J, Benz CC, 2008, Expression of microRNA-146 suppresses NF-kappaB activity with reduction of metastatic potential in breast cancer cells. Oncogene 27:5643–5647 Tanic M, Zajac M, Gomez-Lopez G, Benitez J, Martinez-Delgado B, 2012, Integration of BRCA1-mediated miRNA and mRNA profiles reveals microRNA regulation of TRAF2 and NFκB pathway. Breast Cancer Res Treat 134:41–51 Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y et al, 2011, Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 121:2750–2767 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 815 EP 827 DI 10.1007/s12253-017-0188-4 PG 13 ER PT J AU Chen, J Zhang, Sh Lin, Y Yang, B Cao, J AF Chen, Jia Zhang, Shoude Lin, Yi Yang, Beibei Cao, Jiang TI Antitumor Efficacy of SLPI Promoter-Controlled Expression of Artificial microRNA Targeting EGFR in a Squamous Cell Carcinoma Cell Line SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR; Gene therapy; microRNA; Recombinant adenovirus; SLPI ID EGFR; Gene therapy; microRNA; Recombinant adenovirus; SLPI AB The purpose of this study was to develop a recombinant adenovirus with secretory leukoprotease inhibitor (SLPI) promoter-controlled expression for gene therapy of squamous cell carcinoma (SCC). An artificial microRNA targeting epidermal growth factor receptor (EGFR) was designed, and used to construct a replication-defective recombinant adenovirus with SLPI promoter-controlled expression. The silencing efficiency of this vector (Ad-SLPI-EGFRamiR) was detected in Hep-2 cells. Western blotting showed that the expression of 170 kD EGFR was significantly reduced in Hep- 2 cells 72 h after infection with Ad-SLPI-EGFRamiR. At a multiplicity of infection (MOI) of 200 pfu/cell, proliferation of Hep-2 cells was highly inhibited by Ad-SLPI-EGFRamiR (inhibition rate: ~70%). The apoptosis rate of Hep-2 cells at 72 h after infection with Ad-SLPI-EGFRamiR at a MOI 35 pfu/cell was 32.8%. The adenovirus constructed was able to specifically inhibit the growth of SCC cells in vitro. C1 [Chen, Jia] Zhejiang University, School of Medicine, The Second Affiliated Hospital, Department of Otorhinolaryngology, 88 Jiefang Road, 310009 Hangzhou, China. [Zhang, Shoude] Zhejiang University, School of Medicine, The Second Affiliated Hospital, Department of Otorhinolaryngology, 88 Jiefang Road, 310009 Hangzhou, China. [Lin, Yi] Zhejiang University, School of Medicine, The Second Affiliated Hospital, Department of Otorhinolaryngology, 88 Jiefang Road, 310009 Hangzhou, China. [Yang, Beibei] Zhejiang University, School of Medicine, The Second Affiliated Hospital, Department of Otorhinolaryngology, 88 Jiefang Road, 310009 Hangzhou, China. [Cao, Jiang] Zhejiang University, School of Medicine, The Second Affiliated Hospital, Clinical Research Center, 88 Jiefang Road, 310009 Hangzhou, China. RP Cao, J (reprint author), Zhejiang University, School of Medicine, The Second Affiliated Hospital, Clinical Research Center, 310009 Hangzhou, China. EM caoj@zju.edu.cn CR Alam M, Ratner D, 2001, Cutaneous squamous-cell carcinoma. N Engl J Med 344:975–983 Yan W, Wistuba II, Emmert-Buck MR, Erickson HS, 2011, Squamous cell carcinoma – similarities and differences among anatomical sites. Am J Cancer Res 1:275–300 Pfister DG, Ang KK, Brizel DM et al, 2011, Head and neck cancers. J Natl Compr Cancer Netw 9:596–650 Olsen KD, 2010, Reexamining the treatment of advanced laryngeal cancer. Head Neck 32:1–7 Zhang S, Chen J, Jiang H, MA H, Yang B, 2012, Anti-epidermal growth factor receptor therapy for advanced head and neck squamous cell carcinoma: a meta-analysis. Eur J Clin Pharmacol 68: 561–569 Bonner JA, Harari PM, Giralt J et al, 2006, Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 354:567–578 Vermorken JB, Mesia R, Rivera F et al, 2008, Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med 359:1116–1127 Moon C, Chae YK, Lee J, 2010, Targeting epidermal growth factor receptor in head and neck cancer: lessons learned from cetuximab. Exp Biol Med, Maywood, 235:907–920 Wheeler DL, Dunn EF, Harari PM, 2010, Understanding resistance to EGFR inhibitors-impact on future treatment strategies. Nat Rev Clin Oncol 7:493–507 Young NR, Liu J, Pierce C et al, 2013, Molecular phenotype predicts sensitivity of squamous cell carcinoma of the head and neck to epidermal growth factor receptor inhibition. Mol Oncol 7:359–368 Sharafinski ME, Ferris RL, Ferrone S, Grandis JR, 2010, Epidermal growth factor receptor targeted therapy of squamous cell carcinoma of the head and neck. Head Neck 32:1412–1421 Normanno N, De Luca A, Bianco C et al, 2006, Epidermal growth factor receptor, EGFR, signaling in cancer. Gene 366:2–16 Rapisarda A, Melillo G, 2012, Role of the VEGF/VEGFR axis in cancer biology and therapy. Adv Cancer Res 114:237–267 Herbst RS, 2004, Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Phys 59:21–26 Uribe P, Gonzalez S, 2011, Epidermal growth factor receptor, EGFR, and squamous cell carcinoma of the skin: molecular bases for EGFR-targeted therapy. Pathol Res Pract 207:337–342 Mitsudomi T, Yatabe Y, 2010, Epidermal growth factor receptor in relation to tumor development: EGFR gene and cancer. FEBS J 277:301–308 Rothenberg SM, Engelman JA, Le S, Riese DJ 2nd, Haber DA, Settleman J, 2008, Modeling oncogene addiction using RNA interference. Proc Natl Acad Sci U S A 105:12480–12484 Wang Z, Rao DD, Senzer N, Nemunaitis J, 2011, RNAinterference and cancer therapy. Pharm Res 28:2983–2995 Chang K, Elledge SJ, Hannon GJ, 2006, Lessons from nature: microRNA-based shRNA libraries. Nat Methods 3:707–714 Rein DT, Breidenbach M, Curiel DT, 2006, Current developments in adenovirus- based cancer gene therapy. Future Oncol 2:137–143 Dorer DE, Nettelbeck DM, 2009, Targeting cancer by transcriptional control in cancer gene therapy and viral oncolysis. Adv Drug Deliv Rev 61:554–571 Chen J, Yang B, Zhang S et al, 2012, Antitumor potential of SLPI promoter controlled recombinant caspase-3 expression in laryngeal carcinoma. Cancer Gene Ther 19:328–335 Hynes NE, Lane HA, 2005, ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer 5:341–354 Nukiwa T, Suzuki T, Fukuhara T, Kikuchi T, 2008, Secretory leukocyte peptidase inhibitor and lung cancer. Cancer Sci 99:849–855 Maemondo M, Saijo Y, Narumi K et al, 2004, Gene therapy with secretory leukoprotease inhibitor promoter-controlled replicationcompetent adenovirus for non-small cell lung cancer. Cancer Res 64:4611–4620 Rein DT, Breidenbach M, Kirby TO et al, 2005, A fiber-modified, secretory leukoprotease inhibitor promoter-based conditionally replicating adenovirus for treatment of ovarian cancer. Clin Cancer Res 11:1327–1335 Khare R, Chen CY, Weaver EA, Barry MA, 2011, Advances and future challenges in adenoviral vector pharmacology and targeting. Curr Gene Ther 11:241–258 Thaci B, Ulasov IV,Wainwright DA, LesniakMS, 2011, The challenge for gene therapy: innate immune response to adenoviruses. Oncotarget 2:113–121 Douglas JT, Rogers BE, RosenfeldME, Michael SI, FengM, Curiel DT, 1996, Targeted gene delivery by tropism-modified adenoviral vectors. Nat Biotechnol 14:1574–1578 Mizuguchi H, Hayakawa T, 2004, Targeted adenovirus vectors. Hum Gene Ther 15:1034–1044 Wu H, Han T, Belousova N et al, 2005, Identification of sites in adenovirus hexon for foreign peptide incorporation. J Virol 79: 3382–3390 Wickham TJ, Segal DM, Roelvink PWet al, 1996, Targeted adenovirus gene transfer to endothelial and smooth muscle cells by using bispecific antibodies. J Virol 70:6831–6838 Xia D, Henry LJ, Gerard RD, Deisenhofer J, 1994, Crystal structure of the receptor-binding domain of adenovirus type 5 fiber protein at 1.7 a resolution. Structure 2:1259–1270 Leemans CR, Braakhuis BJ, Brakenhoff RH, 2011, The molecular biology of head and neck cancer. Nat Rev Cancer 11:9–22 Widakowich C, de Castro G Jr, de Azambuja E, Dinh P, Awada A, 2007, Review: side effects of approved molecular targeted therapies in solid cancers. Oncologist 12:1443–1455 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 829 EP 835 DI 10.1007/s12253-016-0160-8 PG 7 ER PT J AU Karabon, L Tupikowski, K Tomkiewicz, A Partyka, A Pawlak-Adamska, E Wojciechowski, A Kolodziej, A Dembowski, J Zdrojowy, R Frydecka, I AF Karabon, Lidia Tupikowski, Krzysztof Tomkiewicz, Anna Partyka, Anna Pawlak-Adamska, Edyta Wojciechowski, Adam Kolodziej, Anna Dembowski, Janusz Zdrojowy, Romuald Frydecka, Irena TI Is the Genetic Background of Co-Stimulatory CD28/CTLA-4 Pathway the Risk Factor for Prostate Cancer? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CTLA-4; CD28; Gene polymorphisms; Prostate cancer ID CTLA-4; CD28; Gene polymorphisms; Prostate cancer AB The impairment of immunological surveillance caused by aberrant T cell activation can lead to an inadequate anti-tumor response. Therefore, deregulation in co-stimulatory pathway might be associated with cancer susceptibility. Here we undertook a prospective study to investigate whether genetic variations in gene encoding molecule CD28 and CTLA-4 playing pivotal role in regulating adoptive immune response can influence susceptibility to prostate cancer. Single nucleotide polymorphisms (SNPs) in CTLA-4 and CD28 genes were genotyped in 301 prostate cancer (PCa) patients and 301 controls. The distributions of the genotypes and haplotypes in the CTLA-4/CD28 SNPs were similar in both studied groups. However, the overrepresentation of carriers of CTLA- 4c.49A>G[A] allele and carriers of CTLA-4g.319C>T[T] allele in PCa as compared to controls was observed (p = 0.082 and p = 0.13, respectively). The risk of disease was higher (OR 1.78) for carriers of both susceptibility alleles as compared to carriers of protective genotypes (p = 0.03). The CTLA- 4c.49A>G and CTLA-4g.319C>T SNPs might be considered as low risk susceptibility locus for PCa. C1 [Karabon, Lidia] Wroclaw Medical University, Department of Urology and Oncological Urology, Borowska 213, 50-556 Wroclaw, Poland. [Tupikowski, Krzysztof] Wroclaw Medical University, Department of Urology and Oncological Urology, Borowska 213, 50-556 Wroclaw, Poland. [Tomkiewicz, Anna] Hirszfeld Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland. [Partyka, Anna] Hirszfeld Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland. [Pawlak-Adamska, Edyta] Hirszfeld Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland. [Wojciechowski, Adam] Wroclaw Medical University, Department of Urology and Oncological Urology, Borowska 213, 50-556 Wroclaw, Poland. [Kolodziej, Anna] Wroclaw Medical University, Department of Urology and Oncological Urology, Borowska 213, 50-556 Wroclaw, Poland. [Dembowski, Janusz] Wroclaw Medical University, Department of Urology and Oncological Urology, Borowska 213, 50-556 Wroclaw, Poland. [Zdrojowy, Romuald] Wroclaw Medical University, Department of Urology and Oncological Urology, Borowska 213, 50-556 Wroclaw, Poland. [Frydecka, Irena] Hirszfeld Institute of Immunology and Experimental Therapy, Department of Experimental Therapy, Weigla 12, 53-114 Wroclaw, Poland. RP Karabon, L (reprint author), Wroclaw Medical University, Department of Urology and Oncological Urology, 50-556 Wroclaw, Poland. EM lkarabon@iitd.pan.wroc.pl CR Chavan S, Bray F, Lortet-Tieulent J et al, 2014, International variations in bladder cancer incidence and mortality. Eur Urol 66:59– 73., DOI 10.1016/j.eururo.2013.10.001 Lichtenstein P, Holm NV, Verkasalo PK et al, 2000, Environmental and heritable factors in the causation of cancer–analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med 343: 78–85., DOI 10.1056/NEJM200007133430201 Heise M, Haus O, 2014, Hereditary prostate cancer. Postepy Hig Med Dosw, Online , 68:653–665., DOI 10.5604/17322693.1104682 Sissung TM, Price DK, Del RM et al, 2014, Genetic variation: effect on prostate cancer. Biochim Biophys Acta 1846:446–456., DOI 10.1016/j.bbcan.2014.08.007 Al Olama AA, Kote-Jarai Z, Berndt SI et al, 2014, A meta-analysis of 87,040 individuals identifies 23 new susceptibility loci for prostate cancer. Nat Genet 46:1103–1109 Eeles RA, Kote-Jarai Z, Al Olama AA et al, 2009, Identification of seven new prostate cancer susceptibility loci through a genome-wide association study. Nat Genet 41:1116–1121., DOI 10.1038/ng.450 Gudmundsson J, Sulem P, Rafnar Tet al, 2008, Common sequence variants on 2p15 and Xp11.22 confer susceptibility to prostate cancer. Nat Genet 40:281–283., DOI 10.1038/ng.89 Gudmundsson J, Sulem P, Gudbjartsson DF et al, 2012, A study based on whole-genome sequencing yields a rare variant at 8q24 associated with prostate cancer. Nat Genet 44:1326–1329., DOI 10.1038/ng.2437 Gudmundsson J, Sulem P, Gudbjartsson DF et al, 2009, Genomewide association and replication studies identify four variants associated with prostate cancer susceptibility. Nat Genet 41:1122–1126., DOI 10.1038/ng.448 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674., DOI 10.1016/j.cell.2011.02.013 Frauwirth KA, Thompson CB, 2002, Activation and inhibition of lymphocytes by costimulation. J Clin Invest 109:295–299., DOI 10.1172/JCI14941 Harper K, Balzano C, Rouvier E,Mattei MG, Luciani MF, Golstein P, 1991, CTLA-4 and CD28 activated lymphocyte molecules are closely related in both mouse and human as to sequence, message expression, gene structure, and chromosomal location. J Immunol 147:1037–1044 Schwartz JC, Zhang X, Fedorov AA, Nathenson SG, Almo SC, 2001, Structural basis for co-stimulation by the human CTLA-4/ B7-2 complex. Nature 410:604–608., DOI 10.1038/35069112 Marengere LE, Waterhouse P, Duncan GS et al, 1996, Regulation of T cell receptor signaling by tyrosine phosphatase SYP association with CTLA-4. Science 272:1170–1173 Parry RV, Chemnitz JM, Frauwirth KA et al, 2005, CTLA-4 and PD- 1 receptors inhibit T-cell activation by distinct mechanisms.Mol Cell Biol 25:9543–9553., DOI 10.1128/MCB.25.21.9543-9553.2005 Hurwitz AA, Kwon ED, van Elsas A, 2000, Costimulatory wars: the tumor menace. Curr Opin Immunol 12:589–596 Leach DR, Krummel MF, Allison JP, 1996, Enhancement of antitumor immunity by CTLA-4 blockade. Science 271:1734–1736 Mocellin S, NittiD, 2013, CTLA-4 blockade and the renaissance of cancer immunotherapy. Biochim Biophys Acta 1836:187–196., DOI 10.1016/j.bbcan.2013.05.003 Schneider H, Rudd CE, 2014, Diverse mechanisms regulate the surface expression of immunotherapeutic target ctla-4. Front Immunol 5:619., DOI 10.3389/fimmu.2014.00619 www.clinicaltrials.com, National Cancer Institute U, 2013, www. clinicaltrials.com; National Cancer Institute, USA Bertrand A, Kostine M, Barnetche T, Truchetet ME, Schaeverbeke T, 2015, Immune related adverse events associated with anti- CTLA-4 antibodies: systematic review and meta-analysis. BMC Med 13:211., DOI 10.1186/s12916-015-0455-8 Diener KR, Woods AE, Manavis J, Brown MP, Hayball JD, 2009, Transforming growth factor-beta-mediated signaling in T lymphocytes impacts on prostate-specific immunity and early prostate tumor progression. Lab Investig 89:142–151., DOI 10.1038 /labinvest.2008.123 May KF Jr, Gulley JL, Drake CG, Dranoff G, Kantoff PW, 2011, Prostate cancer immunotherapy. Clin Cancer Res 17(5233–5238): 1078–0432., DOI 10.1158/1078-0432.CCR-10-3402 Karabon L, Pawlak E, Tomkiewicz A et al, 2011, CTLA-4, CD28, and ICOS gene polymorphism associations with non-small-cell lung cancer. Hum Immunol 72:947–954., DOI 10.1016/j. humimm.2011.05.010 Shi YY, He L, 2005, SHEsis, a powerful software platform for analyses of linkage disequilibrium, haplotype construction, and genetic association at polymorphism loci. Cell Res 15:97–98 Svejgaard A, Ryder LP, 1994, HLA and disease associations: detecting the strongest association. Tissue Antigens 43:18–27 Ghaderi A, Yeganeh F, Kalantari T et al, 2004, Cytotoxic T lymphocyte antigen-4 gene in breast cancer. Breast Cancer Res Treat 86:1–7 Piras G, MonneM, Uras A et al, 2005, Genetic analysis of the 2q33 region containing CD28-CTLA4-ICOS genes: association with non-Hodgkin’s lymphoma. Br J Haematol 129:784–790 Su TH, Chang TY, Lee YJ et al, 2007, CTLA-4 gene and susceptibility to human papillomavirus-16-associated cervical squamous cell carcinoma in Taiwanese women. Carcinogenesis 28:1237–1240 Suwalska K, Pawlak E, Karabon L et al, 2008, Association studies of CTLA-4, CD28, and ICOS gene polymorphisms with B-cell chronic lymphocytic leukemia in the polish population. Hum Immunol 69:193–201 Wong YK, Chang KW, Cheng CY, Liu CJ, 2006, Association of CTLA-4 gene polymorphism with oral squamous cell carcinoma. J Oral Pathol Med 35:51–54 Pawlak E, Karabon L, Wlodarska-Polinska I et al, 2010, Influence of CTLA-4/CD28/ICOS gene polymorphisms on the susceptibility to cervical squamous cell carcinoma and stage of differentiation in the polish population. Hum Immunol 71:195–200 Hou HF, Jin X, Sun T, Li C, Jiang BF, Li QW, 2015, Cytotoxic T lymphocyte-associated antigen 4 gene polymorphisms and autoimmune thyroid diseases: an updated systematic review and cumulative meta-analysis. Int J Endocrinol 2015:747816., DOI 10.1155 /2015/747816 Brozzetti A, Marzotti S, Tortoioli C et al, 2010, Cytotoxic T lymphocyte antigen-4 Ala17 polymorphism is a genetic marker of autoimmune adrenal insufficiency: Italian association study and metaanalysis of European studies. Eur J Endocrinol 162:361–369., DOI 10.1530/EJE-09-0618 Wang J, Liu L, Ma J, Sun F, Zhao Z, Gu M, 2014, Common variants on cytotoxic T lymphocyte antigen-4 polymorphisms contributes to type 1 diabetes susceptibility: evidence based on 58 studies. PLoS One 9:e85982., DOI 10.1371/journal.pone.0085982 Lee YH, Bae SC, Choi SJ, Ji JD, Song GG, 2012, Association between the CTLA-4 + 49 A/G polymorphism and susceptibility to rheumatoid arthritis: a meta-analysis. Mol Biol Rep 39:5599– 5605., DOI 10.1007/s11033-011-1364-3 Chang WW, Zhang L, Yao YS, Su H, 2012, Association between CTLA-4 exon-1 + 49A/G polymorphism and systemic lupus erythematosus: an updated analysis. Mol Biol Rep 39:9159–9165., DOI 10.1007/s11033-012-1788-4 Chen SL, Deng F, Jiang F, Liu JJ, Meng W, 2012, Correlation between CTLA-4 gene polymorphism and systemic lupus erythematosus: a meta-analysis. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 28:1320–1323 Kouki T, Sawai Y, Gardine CA, FisfalenME, AlegreML, DeGroot LJ, 2000, CTLA-4 gene polymorphism at position 49 in exon 1 reduces the inhibitory function of CTLA-4 and contributes to the pathogenesis of graves’ disease. J Immunol 165:6606–6611 Anjos S, Nguyen A, Ounissi-Benkalha H, Tessier MC, Polychronakos C, 2002, A common autoimmunity predisposing signal peptide variant of the cytotoxic T-lymphocyte antigen 4 results in inefficient glycosylation of the susceptibility allele. J Biol Chem 277:46478–46486 Sun T, Zhou Y, YangMet al, 2008, Functional genetic variations in cytotoxic T-lymphocyte antigen 4 and susceptibility to multiple types of cancer. Cancer Res 68:7025–7034 Cozar JM, Romero JM, Aptsiauri N et al, 2007, High incidence of CTLA-4 AA, CT60, polymorphism in renal cell cancer. Hum Immunol 68:698–704 Cheng TY, Lin JT, Chen LT et al, 2006, Association of T-cell regulatory gene polymorphisms with susceptibility to gastric mucosaassociated lymphoid tissue lymphoma. J Clin Oncol 24:3483–3489 Karabon L, Pawlak-Adamska E, Tomkiewicz A et al., 2011, Variations in suppressor molecule CTLA-4 gene are related to susceptibility to multiple myeloma in a polish population. Pathol Oncol Res Solerio E, Tappero G, Iannace L et al, 2005, CTLA4 gene polymorphism in Italian patients with colorectal adenoma and cancer. Dig Liver Dis 37:170–175 Bouwhuis MG, Gast A, Figl A et al., 2009, Polymorphisms in the CD28/CTLA4/ICOS genes: role in malignant melanoma susceptibility and prognosis? Cancer Immunol Immunother WelshMM, Applebaum KM, Spencer SK, Perry AE, Karagas MR, NelsonHH(2009, CTLA4 variants,UV-induced tolerance, and risk of non-melanoma skin cancer. Cancer Res 69:6158–6163 Zhang X, Zhang L, Tian C, Yang L, Wang Z, 2014, Genetic variants and risk of cervical cancer: epidemiological evidence, metaanalysis and research review. BJOG 121:664–674., DOI 10.1111 /1471-0528.12638 Zhang Y, Zhang J, Deng Y, Tian C, Li X, Huang J, Fan H, 2011, Polymorphisms in the cytotoxic T-lymphocyte antigen 4 gene and cancer risk: a meta-analysis. Cancer 117:4312–4324., DOI 10.1002 /cncr.25979 Chistiakov DA, Savost’anov KV, Turakulov RI, Efremov IA, Demurov LM, 2006, Genetic analysis and functional evaluation of the C/T(−318, and a/G(−1661, polymorphisms of the CTLA-4 gene in patients affected with graves’ disease. Clin Immunol 118: 233–242., DOI 10.1016/j.clim.2005.09.017 Ligers A, Teleshova N, Masterman T, Huang WX, Hillert J, 2001, CTLA-4 gene expression is influenced by promoter and exon 1 polymorphisms. Genes Immun 2:145–152 Dilmec F, Ozgonul A, Uzunkoy A, Akkafa F, 2008, Investigation of CTLA-4 and CD28 gene polymorphisms in a group of Turkish patients with colorectal cancer. Int J Immunogenet 35:317–321 Ueda H, Howson JM, Esposito L et al, 2003, Association of the Tcell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature 423:506–511., DOI 10.1038/nature01621 Karabon L, Kosmaczewska A, BilinskaMet al, 2009, The CTLA- 4 gene polymorphisms are associated with CTLA-4 protein expression levels in multiple sclerosis patients and with susceptibility to disease. Immunology 128:e787–e796 Tupikowski K, Partyka A, Kolodziej A et al, 2015, CTLA-4 and CD28 genes’ polymorphisms and renal cell carcinoma susceptibility in the polish population - a prospective study. Tissue Antigens 86:353–361., DOI 10.1111/tan.12671 56 Zhao HY, Duan HX, Gu Y, 2014, Meta-analysis of the cytotoxic Tlymphocyte antigen 4 gene +6230G/A polymorphism and cancer risk. Clin Transl Oncol., DOI 10.1007/s12094-014-1159-9 57 Ploski R, Wozniak M, Pawlowski R et al, 2002, Homogeneity and distinctiveness of polish paternal lineages revealed by Y chromosome microsatellite haplotype analysis. Hum Genet 110:592–600 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 837 EP 843 DI 10.1007/s12253-016-0180-4 PG 7 ER PT J AU Roychowdhury, A Samadder, S Islam, SM Chaudhury, K Roy, A Banerjee, D Mandal, R Basu, SP Roychoudhury, S Panda, KCh AF Roychowdhury, Anirban Samadder, Sudip Islam, Saimul Md Chaudhury, Kalyansree Roy, Anup Banerjee, Dipanwita Mandal, Ranajit Basu, S Partha Roychoudhury, Susanta Panda, Kumar Chinmay TI Identification of Changes in the Human Papilloma Virus 16 (HPV16) Genome During Early Dissemination of Cervical Cancer Cells May Complement Histological Diagnosis of Lymph Node Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Uterine cervical carcinoma; Disseminated tumor cells (DTCs); Lymph node metastasis; Human papilloma virus (HPV) type 16; HPV16 integration; HPV16 methylation ID Uterine cervical carcinoma; Disseminated tumor cells (DTCs); Lymph node metastasis; Human papilloma virus (HPV) type 16; HPV16 integration; HPV16 methylation AB Cancer of the uterine cervix (CACX) is one of the most common carcinoma affecting women worldwide. During treatment, histologically lymph node (LN) metastasis and presence of HPV DNA in blood plasma act as a major prognostic factor. Despite the lack of apparent LN involvement, some early-invasive CACX patients have shown recurrences and poor survival. This is suggestive of undetected early dissemination of cancer cells characterized by presence of HPV DNA in histologically non-metastatic LNs which finally progresses into histologically visible metastasis. This present study investigated the status and origin of HPV genome during early dissemination by molecular analysis in primary tumor (PT), histologically non-metastatic pelvic lymph nodes (LNs) and blood plasma (BP) of same patient. First, CACX patients showing signs of early dissemination was identified by detection of HPV in PT (n = 22) and their corresponding histologically non-metastatic pelvic LNs (n = 45) and BP (n = 18) followed by typing of HPV16/18. This was followed by comparative analysis of the physical, copy number and methylation (enhancer/early/late) status of HPV16 genome present in LNs and BP with that of PT. Our study revealed for the first time that the HPV16 genome were frequently present in the integrated form though the copy number was low in both non-metastatic LNs and BP. However, the methylation pattern of PT was discordant with that of corresponding LNs and BP in majority of the cases. Critical assessment of HPV16 profiles established that the presence of hrHPV may be due to the early dissemination of PT cells having significant pathological implications. C1 [Roychowdhury, Anirban] Chittaranjan National Cancer Institute, Department of Oncogene RegulationKolkata, India. [Samadder, Sudip] Chittaranjan National Cancer Institute, Department of Oncogene RegulationKolkata, India. [Islam, Saimul Md] Chittaranjan National Cancer Institute, Department of Oncogene RegulationKolkata, India. [Chaudhury, Kalyansree] Burdwan Medical College, Burdwan, Department of Gynecology and ObstetricsBurdwan, West Bengal, India. [Roy, Anup] North Bengal Medical College and Hospital, Department of PathologySiliguri, West Bengal, India. [Banerjee, Dipanwita] Chittaranjan National Cancer Institute, Department of Gynecology OncologyKolkata, India. [Mandal, Ranajit] Chittaranjan National Cancer Institute, Department of Gynecology OncologyKolkata, India. [Basu, S Partha] World Health Organization (WHO), International Agency for Research on Cancer (IARC), India Screening Group (SCR), Early Detection and Prevention Section (EDP)Lyon, France. [Roychoudhury, Susanta] Saroj Gupta Cancer Centre & Research InstituteKolkata, India. [Panda, Kumar Chinmay] Chittaranjan National Cancer Institute, Department of Oncogene RegulationKolkata, India. RP Panda, KCh (reprint author), Chittaranjan National Cancer Institute, Department of Oncogene Regulation, Kolkata, India. EM ckpanda.cnci@gmail.com CR Jemal A et al, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Bosch FX, Munoz N, 2002, The viral etiology of cervical cancer. Virus Res 89(2):183–190 Walboomers JM et al, 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189(1):12–19 Woodman CB, Collins SI, Young LS, 2007, The natural history of cervical HPV infection: unresolved issues. Nat Rev Cancer 7(1): 11–22 Wong SY, Hynes RO, 2006, Lymphatic or hematogenous dissemination: how does a metastatic tumor cell decide? Cell Cycle 5(8): 812–817 NoventaMet al, 2014, Usefulness,methods and rationale of lymph nodes HPV-DNA investigation in estimating risk of early stage cervical cancer recurrence: a systematic literature review. Clin Exp Metastasis 31(7):853–867 LancasterWD et al, 1986, Human papillomavirus deoxyribonucleic acid in cervical carcinoma from primary and metastatic sites. Am J Obstet Gynecol 154(1):115–119 Park Y et al, 2011, Expected clinical applications of circulating tumor cells in breast cancer. World J Clin Oncol 2(8):303–310 Pornthanakasem W et al, 2001, Human papillomavirus DNA in plasma of patients with cervical cancer. BMC Cancer 1:2 Singh RK et al, 2006, Human papillomavirus prevalence in postradiotherapy uterine cervical carcinoma patients: correlation with recurrence of the disease. International Journal of Gynecological Cancer : Official Journal of the International Gynecological Cancer Society 16(3):1048–1054 Dutta S et al, 2016, Alteration of human papillomavirus type 16 genetic and epigenetic profiles in cervical cancer patients is indicative of poor disease prognosis: a cohort analysis. International Journal of Gynecological Cancer : Official Journal of the International Gynecological Cancer Society 26(4):750–757 Steenbergen RD et al, 2014, Clinical implications of, epi)genetic changes in HPV-induced cervical precancerous lesions. Nat Rev Cancer 14(6):395–405 Dutta S et al, 2012, Prevalence of human papillomavirus in women without cervical cancer : a population-based study in Eastern India. International Journal of Gynecological Pathology : Official Journal of the International Society of Gynecological Pathologists 31(2): 178–183 Dutta S et al, 2015, Physical and methylation status of human papillomavirus 16 in asymptomatic cervical infections changes with malignant transformation. J Clin Pathol 68(3):206–211 Mazumder Indra D et al, 2011, Genetic and epigenetic changes of HPV16 in cervical cancer differentially regulate E6/E7 expression and associate with disease progression. Gynecol Oncol 123(3):597–604 Chakrabarti S et al, 2001, Microsatellite instability in squamous cell carcinoma of head and neck from the Indian patient population. Int J Cancer 92(4):555–561 Sambrook J, Russell DW, Sambrook J, 2006, The condensed protocols from molecular cloning : a laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y Swan DC et al, 1999, Human papillomavirus, HPV, DNA copy number is dependent on grade of cervical disease and HPV type. J Clin Microbiol 37(4):1030–1034 Shukla S et al, 2014, Physical state & copy number of high risk human papillomavirus type 16 DNA in progression of cervical cancer. Indian J Med Res 139(4):531–543 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 845 EP 852 DI 10.1007/s12253-017-0189-3 PG 8 ER PT J AU Sumegi, K Duga, B Melegh, IB Banfai, Zs Kovesdi, E Maasz, A Melegh, B AF Sumegi, Katalin Duga, Balazs Melegh, I Bela Banfai, Zsolt Kovesdi, Erzsebet Maasz, Anita Melegh, Bela TI Marked Differences of Haplotype Tagging SNP Distribution, Linkage, and Haplotype Profile of APOA5 Gene in Roma Population Samples SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE APOA5; Triglyceride; Haplotype; Linkage disequilibrium; Roma ID APOA5; Triglyceride; Haplotype; Linkage disequilibrium; Roma AB Roma people are underprivileged, neglected population worldwide, with severe healthcare problems. They have significantly increased prevalence of cardiovascular morbidity, presumably related to their poor social status, alcohol consumption and smoking habits. Assuming that genetic background also plays a role in their susceptibility for cardiovascular diseases, we hypothesized that APOA5 gene polymorphisms, an important role-player in lipid metabolism and in the development of metabolic syndrome and cardio/ cerebrovascular events, may also be involved. We examined four APOA5 polymorphisms in 363 Roma and 404 Hungarian DNA samples. For rs662799, rs2266788, rs207560 and rs3135506 we found elevated plasma triglyceride levels in the risk allele carriers compared to non-carriers in both populations. At least a two-fold significant increase was detected in minor allele frequencies in Roma when compared to Hungarians, except the rs2266788 variant. Haplotype analysis revealed significant increase of APOA5*2, APOA5*4 in Roma, as opposed to the higher levels of APOA5*5 found in Hungarians. Different linkage disequilibrium was found between rs207560 and rs3135506 variants in Roma compared to Hungarians. The profound differences observed in almost all APOA5 polymorphisms in Roma require special attention, since these variants are known to associate with cardio/cerebrovascular susceptibility. C1 [Sumegi, Katalin] University of Pecs, Department of Medical Genetics and Child Development, Szigeti Street 12, H-7624 Pecs, Hungary. [Duga, Balazs] University of Pecs, Department of Medical Genetics and Child Development, Szigeti Street 12, H-7624 Pecs, Hungary. [Melegh, I Bela] University of Pecs, Department of Medical Genetics and Child Development, Szigeti Street 12, H-7624 Pecs, Hungary. [Banfai, Zsolt] University of Pecs, Department of Medical Genetics and Child Development, Szigeti Street 12, H-7624 Pecs, Hungary. [Kovesdi, Erzsebet] University of Pecs, Department of Medical Genetics and Child Development, Szigeti Street 12, H-7624 Pecs, Hungary. [Maasz, Anita] University of Pecs, Department of Medical Genetics and Child Development, Szigeti Street 12, H-7624 Pecs, Hungary. [Melegh, Bela] University of Pecs, Department of Medical Genetics and Child Development, Szigeti Street 12, H-7624 Pecs, Hungary. RP Melegh, B (reprint author), University of Pecs, Department of Medical Genetics and Child Development, H-7624 Pecs, Hungary. EM melegh.bela@pte.hu CR Krauss RM, 1998, Atherogenicity of triglyceride-rich lipoproteins. Am J Cardiol 81(4A):13B–17B Hodis HN, 1999, Triglyceride-rich lipoprotein remnant particles and risk of atherosclerosis. Circulation 99(22):2852–2854 Cullen P, 2000, Evidence that triglycerides are an independent coronary heart disease risk factor. Am J Cardiol 86(9):943–949 Haim M, Benderly M, Brunner D, Behar S, Graff E, Reicher-Reiss H, Goldbourt U, 1999, Elevated serumtriglyceride levels and longterm mortality in patients with coronary heart disease: the Bezafibrate infarction prevention, BIP, registry. Circulation 100(5):475–482 Pennacchio LA, Olivier M, Hubacek JA, Cohen JC, Cox DR, Fruchart JC, Krauss RM, Rubin EM, 2001, An apolipoprotein influencing triglycerides in humans and mice revealed by comparative sequencing. Science 294(5540):169–173., DOI 10.1126 /science.1064852 van der Vliet HN, Sammels MG, Leegwater AC, Levels JH, Reitsma PH, Boers W, Chamuleau RA, 2001, Apolipoprotein AV: a novel apolipoprotein associated with an early phase of liver regeneration. J Biol Chem 276(48):44512–44520., DOI 10.1074/jbc. M106888200 Sharma V, Ryan RO, Forte TM, 2012, Apolipoprotein A-V dependent modulation of plasma triacylglycerol: a puzzlement. Biochim Biophys Acta 1821(5):795–799., DOI 10.1016/j.bbalip.2011.12.002 Gin P, Yin L, Davies BS,Weinstein MM, Ryan RO, Bensadoun A, Fong LG, Young SG, Beigneux AP, 2008, The acidic domain of GPIHBP1 is important for the binding of lipoprotein lipase and chylomicrons. J Biol Chem 283(43):29554–29562., DOI 10.1074 /jbc.M802579200 Gin P, Beigneux AP, Davies B, Young MF, Ryan RO, Bensadoun A, Fong LG, Young SG, 2007, Normal binding of lipoprotein lipase, chylomicrons, and apo-AV to GPIHBP1 containing a G56R amino acid substitution. Biochim Biophys Acta 1771(12): 1464–1468., DOI 10.1016/j.bbalip.2007.10.005 Sun G, Bi N, Li G, Zhu X, Zeng W,Wu G, Xue H, Chen B, 2006, Identification of lipid binding and lipoprotein lipase activation domains of human apoAV. Chem Phys Lipids 143(1–2):22–28., DOI 10.1016/j.chemphyslip.2006.04.004 Lookene A, Beckstead JA, Nilsson S, Olivecrona G, Ryan RO, 2005, Apolipoprotein A-V-heparin interactions: implications for plasma lipoprotein metabolism. J Biol Chem 280(27):25383– 25387., DOI 10.1074/jbc.M501589200 Merkel M, Loeffler B, Kluger M, Fabig N, Geppert G, Pennacchio LA, Laatsch A, Heeren J, 2005, Apolipoprotein AV accelerates plasma hydrolysis of triglyceride-rich lipoproteins by interaction with proteoglycan-bound lipoprotein lipase. J Biol Chem 280(22): 21553–21560., DOI 10.1074/jbc.M411412200 Pennacchio LA, Olivier M, Hubacek JA, Krauss RM, Rubin EM, Cohen JC, 2002, Two independent apolipoprotein A5 haplotypes influence human plasma triglyceride levels. Hum Mol Genet 11(24):3031–3038 Pennacchio LA, Rubin EM, 2003, Apolipoprotein A5, a newly identified gene that affects plasma triglyceride levels in humans and mice. Arterioscler Thromb Vasc Biol 23(4):529–534., DOI 10.1161/01.ATV.0000054194.78240.45 Hubacek JA, Skodova Z, Adamkova V, Lanska V, Poledne R, 2004, The influence of APOAV polymorphisms, T-1131 > C and S19 > W, on plasma triglyceride levels and risk of myocardial infarction. Clin Genet 65(2):126–130 Kisfali P, Mohas M, Maasz A, Hadarits F, Marko L, Horvatovich K, Oroszlan T, Bagosi Z, Bujtor Z, Gasztonyi B, Wittmann I, Melegh B, 2008, Apolipoprotein A5 IVS3 + 476A allelic variant associates with increased trigliceride levels and confers risk for development of metabolic syndrome in Hungarians. Circ J 72(1):40–43 Maasz A, Kisfali P, Horvatovich K,MohasM, Marko L, CsongeiV, Farago B, Jaromi L, Magyari L, Safrany E, Sipeky C, Wittmann I, Melegh B, 2007, Apolipoprotein A5 T-1131C variant confers risk for metabolic syndrome. Pathol Oncol Res 13(3):243–247 Maasz A, Kisfali P, Jaromi L, Horvatovich K, Szolnoki Z, Csongei V, Safrany E, Sipeky C, Hadarits F, Melegh B, 2008, Apolipoprotein A5 gene IVS3 + G476A allelic variant confers susceptibility for development of ischemic stroke. Circ J 72(7):1065–1070 Maasz A, Kisfali P, Szolnoki Z, Hadarits F, Melegh B, 2008, Apolipoprotein A5 gene C56G variant confers risk for the development of large-vessel associated ischemic stroke. J Neurol 255(5): 649–654., DOI 10.1007/s00415-008-0768-z Zhang Z, Peng B, Gong RR, Gao LB, Du J, Fang DZ, Song YY, Li YH, Ou GJ, 2011, Apolipoprotein A5 polymorphisms and risk of coronary artery disease: a meta-analysis. Biosci Trends 5(4):165–172 Sarwar N, Sandhu MS, Ricketts SL, Butterworth AS, Di Angelantonio E, Boekholdt SM, Ouwehand W, Watkins H, Samani NJ, Saleheen D, Lawlor D, Reilly MP, Hingorani AD, Talmud PJ, Danesh J, 2010, Triglyceride-mediated pathways and coronary disease: collaborative analysis of 101 studies. Lancet 375(9726):1634–1639., DOI 10.1016/S0140-6736(10)60545-4 Kisfali P, Mohas M, Maasz A, Polgar N, Hadarits F, Marko L, Brasnyo P, Horvatovich K, Oroszlan T, Bagosi Z, Bujtor Z, Gasztonyi B, Rinfel J, Wittmann I, Melegh B, 2009, Haplotype analysis of the apolipoprotein A5 gene in patients with the metabolic syndrome. Nutr Metab Cardiovasc Dis 20(7):505–511., DOI 10.1016/j.numecd.2009.05.001 Kalaydjieva L, Gresham D, Calafell F, 2001, Genetic studies of the Roma, gypsies): a review. BMC Med Genet 2:5 Zeman CL, Depken DE, Senchina DS, 2003, Roma health issues: a review of the literature and discussion. Ethn Health 8(3):223–249., DOI 10.1080/1355785032000136434 Gresham D, Morar B, Underhill PA, Passarino G, Lin AA,Wise C, Angelicheva D, Calafell F, Oefner PJ, Shen P, Tournev I, de Pablo R, Kucinskas V, Perez-Lezaun A, Marushiakova E, Popov V, Kalaydjieva L, 2001, Origins and divergence of the Roma, gypsies).AmJ HumGenet 69(6):1314–1331., DOI 10.1086/324681 McKee M, 1997, The health of gypsies. BMJ 315(7117):1172–1173 Vozarova de Courten B, de Courten M, Hanson RL, Zahorakova A, Egyenes HP, Tataranni PA, Bennett PH, Vozar J, 2003, Higher prevalence of type 2 diabetes, metabolic syndrome and cardiovascular diseases in gypsies than in non-gypsies in Slovakia. Diabetes Res Clin Pract 62(2):95–103., DOI 10.1016/S0168822703001621 Dobranici M, Buzea A, Popescu R, 2012, The cardiovascular risk factors of the Roma, gypsies, people in Central-Eastern Europe: a review of the published literature. J Med Life 5(4):382–389 Barrett JC, Fry B, Maller J, Daly MJ, 2005, Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics 21(2): 263–265., DOI 10.1093/bioinformatics/bth457 Havasi V, Szolnoki Z, Talian G, Bene J, Komlosi K, Maasz A, Somogyvari F, Kondacs A, Szabo M, Fodor L, Bodor A, Melegh B, 2006, Apolipoprotein A5 gene promoter region T-1131C polymorphism associates with elevated circulating triglyceride levels and confers susceptibility for development of ischemic stroke. J Mol Neurosci 29(2):177–183., DOI 10.1385/JMN:29:2:177 Flecha A, 2013, Healthier lives for European minority groups: school and health care, lessons from the Roma. Int J Environ Res Public Health 10(8):3089–3111., DOI 10.3390/ijerph10083089 Bogdanovic D, Nikic D, Petrovic B, Kocic B, Jovanovic J, Nikolic M, Milosevic Z, 2007, Mortality of Roma population in Serbia, 2002-2005. Croat Med J 48(5):720–726 Masseria C, Mladovsky P, Hernandez-Quevedo C, 2010, The socio-economic determinants of the health status of Roma in comparison with non-Roma in Bulgaria, Hungary and Romania. Eur J Pub Health 20(5):549–554., DOI 10.1093/eurpub/ckq102 Alberty R, Albertyova D, Ahlers I, 2009, Distribution and correlations of non-high-density lipoprotein cholesterol in Roma and Caucasian children: the Slovak lipid community study. Coll Antropol 33(4):1015–1022 Dolinska S, Kudlackova M, Ginter E, 2007, The prevalence of female obesity in the world and in the Slovak gypsy women. Bratisl Lek Listy 108(4–5):207–211 Carrasco-Garrido P, Lopez de Andres A, Hernandez Barrera V, Jimenez-Trujillo I, Jimenez-Garcia R, 2011, Health status of Roma women in Spain. Eur J Pub Health 21(6):793–798., DOI 10.1093/eurpub/ckq153 Gualdi-Russo E, Zironi A, Dallari GV, Toselli S, 2009, Migration and health in Italy: a multiethnic adult sample. J Travel Med 16(2): 88–95., DOI 10.1111/j.1708-8305.2008.00280.x Krajcovicova-Kudlackova M, Blazicek P, Spustova V, Valachovicova M, Ginter E, 2004, Cardiovascular risk factors in young gypsy population. Bratisl Lek Listy 105(7–8):256–259 Sipeky C, Weber A, Szabo M, Melegh BI, Janicsek I, Tarlos G, Szabo I, Sumegi K, Melegh B, 2013, High prevalence of CYP2C19*2 allele in Roma samples: study on Roma and Hungarian population samples with review of the literature. Mol Biol Rep 40(8):4727–4735., DOI 10.1007/s11033-013-2569-4 Sipeky C, Csongei V, Jaromi L, Safrany E, Maasz A, Takacs I, Beres J, Fodor L, Szabo M, Melegh B, 2011, Genetic variability and haplotype profile of MDR1, ABCB1, in Roma and Hungarian population samples with a review of the literature. Drug Metab Pharmacokinet 26(2):206–215 Sipeky C, Lakner L, Szabo M, Takacs I, Tamasi V, Polgar N, Falus A, Melegh B, 2009, Interethnic differences of CYP2C9 alleles in healthy Hungarian and Roma population samples: relationship to worldwide allelic frequencies. Blood Cells Mol Dis 43(3):239–242., DOI 10.1016/j.bcmd.2009.05.005 Sipeky C, Csongei V, Jaromi L, Safrany E, Polgar N, Lakner L, Szabo M, Takacs I, Melegh B, 2009, Vitamin K epoxide reductase complex 1, VKORC1, haplotypes in healthy Hungarian and Roma population samples. Pharmacogenomics 10(6):1025–1032., DOI 10.2217/pgs.09.46 Moorjani P, Patterson N, Loh PR, Lipson M, Kisfali P, Melegh BI, Bonin M, Kadasi L, Riess O, Berger B, Reich D, Melegh B, 2013, Reconstructing Roma history from genome-wide data. PLoS One 8(3):e58633., DOI 10.1371/journal.pone.0058633 Mendizabal I, Lao O, Marigorta UM, Wollstein A, Gusmao L, Ferak V, Ioana M, Jordanova A, Kaneva R, Kouvatsi A, Kucinskas V, Makukh H, Metspalu A, Netea MG, de Pablo R, Pamjav H, Radojkovic D, Rolleston SJ, Sertic J, Macek M Jr, Comas D, Kayser M, 2012, Reconstructing the population history of European Romani from genome-wide data. Curr Biol 22(24): 2342–2349., DOI 10.1016/j.cub.2012.10.039 Evans D, Seedorf U, Beil FU, 2005, Polymorphisms in the apolipoprotein A5, APOA5, gene and type III hyperlipidemia. Clin Genet 68(4):369–372., DOI 10.1111/j.1399-0004.2005.00510.x Dallongeville J, Cottel D, Wagner A, Ducimetiere P, Ruidavets JB, Arveiler D, Bingham A, Ferrieres J, Amouyel P, Meirhaeghe A, 2008, The APOA5 Trp19 allele is associated with metabolic syndrome via its association with plasma triglycerides. BMC Med Genet 9:84., DOI 10.1186/1471-2350-9-84 Melegh BI, Duga B, Sumegi K, Kisfali P, Maasz A, Komlosi K, Hadzsiev K, Komoly S, Kosztolanyi G, Melegh B, 2012, Mutations of the apolipoprotein A5 gene with inherited hypertriglyceridaemia: review of the current literature. Curr Med Chem 19(36):6163–6170 Yin RX, Li YY, Lai CQ, 2011, Apolipoprotein A1/C3/A5 haplotypes and serum lipid levels. Lipids Health Dis 10:140., DOI 10.1186 /1476-511X-10-140 Furuya TK, Chen ES, Ota VK, Mazzotti DR, Ramos LR, Cendoroglo MS, Araujo LQ, Burbano RR, Smith MA, 2013, Association of APOA1 and APOA5 polymorphisms and haplotypes with lipid parameters in a Brazilian elderly cohort. Genet Mol Res 12(3):3495–3499., DOI 10.4238/2013.February.28.7 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 853 EP 861 DI 10.1007/s12253-017-0197-3 PG 9 ER PT J AU Papp, G Mihaly, D Sapi, Z AF Papp, Gergo Mihaly, Dora Sapi, Zoltan TI Unusual Signal Patterns of Break-apart FISH Probes Used in the Diagnosis of Soft Tissue Sarcomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Atypical FISH pattern; Break-apart probe; Soft tissue sarcoma ID Atypical FISH pattern; Break-apart probe; Soft tissue sarcoma AB Break-apart FISH probes are the most popular and reliable type of FISH probes used to confirm certain pathological diagnoses. The interpretation is usually easy, however, in some instances it is not so unequivocal. Our aim was to reveal and elucidate the problems occurring in the process of evaluation of the break-apart probe results. Altogether 301 soft tissue sarcomas with confirmed molecular tests using breakapart probes were assessed to reveal the frequency and type of unusual signal pattern. Among 89 synovial sarcoma (SS18) 11%, 12 alveolar rhabdomyosarcoma (FOXO1) 50%, 53 myxoid liposarcoma (DDIT3) 7.5%, 6 low grade fibromyxoid sarcoma (FUS) 67%, 93 Ewing sarcoma (EWSR1) 3%, 12 clear cell sarcoma (EWSR1) 8%, 5 desmoplastic small round cell tumor (EWSR1) 0%, 9 extraskeletal myxoid chondrosarcoma (EWSR1) 0%, 2 myoepithelial carcinoma (EWSR1) 50%, 14 dermatofibrosarcoma protuberans (COL1A1) 86% and 6 nodular fasciitis (USP6) 17% atypical break-apart signals were detected. Despite the unusual signal pattern type, the fusion genes were detected using either metaphase FISH, interphase FISH with translocation/TriCheck probe or RT-PCR methods. Although the interpretation problems in the process to evaluate the break-apart probe results is well known from sporadic case reports, a systemic overview to detect their frequency has not been performed so far. In our work we highlighted the relative frequency of this problem and pinpointed those signal-patterns which, despite their unusual appearance, can still confirm the diagnosis. C1 [Papp, Gergo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Mihaly, Dora] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. RP Sapi, Z (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM sapi.zoltan.dr@gmail.com CR Bridge JA, 2014, The role of cytogenetics and molecular diagnostics in the diagnosis of soft-tissue tumors. Modern pathology: an official journal of the United States and Canadian Academy of Pathology, Inc 27(Suppl 1):S80–S97. doi :10.1038 /modpathol.2013.179 Bridge JA, Cushman-Vokoun AM, 2011)Molecular diagnostics of soft tissue tumors. Arch Pathol Lab Med 135(5):588–601., DOI 10.1043/2010-0594-rair.1 Fletcher CD, Organization WH, Cancer IAfRo, 2013, WHO classification of tumours of soft tissue and bone. IARC press Ventura RA, Martin-Subero JI, Jones M, McParland J, Gesk S, Mason DY, Siebert R, 2006, FISH analysis for the detection of lymphoma-associated chromosomal abnormalities in routine paraffin-embedded tissue. The Journal of molecular diagnostics: JMD 8(2):141–151., DOI 10.2353/jmoldx.2006.050083 Romeo S, Dei Tos AP, 2010, Soft tissue tumors associated with EWSR1 translocation. Virchows Archiv: an international journal of pathology 456(2):219–234., DOI 10.1007/s00428-009-0854-3 Motoi T, Kumagai A, Tsuji K, Imamura T, Fukusato T, 2010, Diagnostic utility of dual-color break-apart chromogenic in situ hybridization for the detection of rearranged SS18 in formalinfixed, paraffin-embedded synovial sarcoma. Hum Pathol 41(10): 1397–1404., DOI 10.1016/j.humpath.2010.02.009 Argani P, Aulmann S, Karanjawala Z, Fraser RB, Ladanyi M, RodriguezMM(2009, Melanotic Xp11 translocation renal cancers: a distinctive neoplasm with overlapping features of PEComa, carcinoma, and melanoma. Am J Surg Pathol 33(4):609–619., DOI 10.1097/PAS.0b013e31818fbdff AmaryMF, Berisha F, Bernardi Fdel C, Herbert A, JamesM, Reis- Filho JS, Fisher C, Nicholson AG, Tirabosco R, Diss TC, Flanagan AM, 2007, Detection of SS18-SSX fusion transcripts in formalinfixed paraffin-embedded neoplasms: analysis of conventional RTPCR, qRT-PCR and dual color FISH as diagnostic tools for synovial sarcoma. Modern pathology: an official journal of the United States and Canadian Academy of Pathology, Inc 20(4):482–496., DOI 10.1038/modpathol.3800761 Matsumura T, Yamaguchi T, Seki K, Shimoda T, Wada T, Yamashita T, Hasegawa T, 2008, Advantage of FISH analysis using FKHR probes for an adjunct to diagnosis of rhabdomyosarcomas. Virchows Archiv: an international journal of pathology 452(3):251–258., DOI 10.1007/s00428-007-0554-9 Liu J, Guzman MA, Pezanowski D, Patel D, Hauptman J, Keisling M, Hou SJ, Papenhausen PR, Pascasio JM, Punnett HH, Halligan GE, de Chadarevian JP, 2011, FOXO1-FGFR1 fusion and amplification in a solid variant of alveolar rhabdomyosarcoma. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 24(10):1327–1335., DOI 10.1038 /modpathol.2011.98 Willmore-Payne C, Holden J, Turner KC, Proia A, Layfield LJ, 2008, Translocations and amplifications of chromosome 12 in liposarcoma demonstrated by the LSI CHOP breakapart rearrangement probe. Arch Pathol Lab Med 132(6):952–957., DOI 10.1043 /1543-2165(2008)132[952:taaoci]2.0.co;2 Smith SH, Weiss SW, Jankowski SA, Coccia MA, Meltzer PS, 1992, SAS amplification in soft tissue sarcomas. Cancer Res 52(13):3746–3749 Berner JM, Forus A, Elkahloun A, Meltzer PS, Fodstad O, Myklebost O, 1996, Separate amplified regions encompassing CDK4 and MDM2 in human sarcomas. Genes Chromosom Cancer 17(4):254–259., DOI 10.1002/(sici)1098-2264(199612)17 :4<254::aid-gcc7>3.0.co;2-2 Dei Tos AP, Doglioni C, Piccinin S, Sciot R, Furlanetto A, Boiocchi M, Dal Cin P, Maestro R, Fletcher CD, Tallini G, 2000, Coordinated expression and amplification of the MDM2, CDK4, and HMGI-C genes in atypical lipomatous tumours. J Pathol 190(5):531–536., DOI 10.1002/(sici)1096-9896(200004)190:5<531 ::aid-path579>3.0.co;2-w Bartuma H, Moller E, Collin A, Domanski HA, Von Steyern FV, Mandahl N, Mertens F, 2010, Fusion of the FUS and CREB3L2 genes in a supernumerary ring chromosome in low-grade fibromyxoid sarcoma. Cancer Genet Cytogenet 199(2):143–146., DOI 10.1016/j.cancergencyto.2010.02.011 Chien YC, Karolyi K, Kovacs I, 2016, Paravertebral low-grade fibromyxoid sarcoma with supernumerary ring chromosome: case report and literature review. Ann Clin Lab Sci 46(1):90–96 Jinawath N, Morsberger L, Norris-Kirby A, Williams LM, Yonescu R, Argani P, Griffin CA, Murphy KM, 2010, Complex rearrangement of chromosomes 1, 7, 21, 22 in Ewing sarcoma. Cancer Genet Cytogenet 201(1):42–47., DOI 10.1016/j.cancergencyto.2010.04.021 Szuhai K, IJszenga M, Tanke HJ, Taminiau AH, de Schepper A, van Duinen SG, Rosenberg C, Hogendoorn PC, 2007, Detection and molecular cytogenetic characterization of a novel ring chromosome in a histological variant of Ewing sarcoma. Cancer Genet Cytogenet 172(1):12–22., DOI 10.1016/j.cancergencyto.2006.07.007 Sirvent N, Maire G, Pedeutour F, 2003, Genetics of dermatofibrosarcoma protuberans family of tumors: from ring chromosomes to tyrosine kinase inhibitor treatment. Genes Chromosom Cancer 37(1):1–19., DOI 10.1002/gcc.10202 Chen J,Ye X, LiY,Wei C, Zheng Q, Zhong P,Wu S, Luo Y, Liao Z, Ye H, 2014, Chromosomal translocation involving USP6 gene in nodular fasciitis. Zhonghua bing li xue za zhi = Chinese journal of pathology 43(8):533–536 Geiersbach K, Rector LS, Sederberg M, Hooker A, Randall RL, Schiffman JD, South ST, 2011, Unknown partner for USP6 and unusual SS18 rearrangement detected by fluorescence in situ hybridization in a solid aneurysmal bone cyst. Cancer genetics 204(4): 195–202., DOI 10.1016/j.cancergen.2011.01.004 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 863 EP 871 DI 10.1007/s12253-017-0200-z PG 9 ER PT J AU Raza, Y Khan, A Khan, IA Khan, S Akhtar, ShS Mubarak, M Ahmed, A Kazmi, USh AF Raza, Yasir Khan, Adnan Khan, Iqbal Asif Khan, Saeed Akhtar, Shakeel Syed Mubarak, Muhammad Ahmed, Ayaz Kazmi, Urooj Shahana TI Combination of Interleukin 1 Polymorphism and Helicobacter pylori Infection: an Increased Risk of Gastric Cancer in Pakistani Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Helicobacter pylori; Gastritis; Gastric cancer; Cytokines; Polymorphism ID Helicobacter pylori; Gastritis; Gastric cancer; Cytokines; Polymorphism AB Helicobacter pylori is one of the major risk factors involved in the development ofgastritis and gastric cancer (GC). H. pylori infection leads to increased production of pro-inflammatory cytokines by the host. Carriage of specific polymorphisms in cytokine genesmay be associatedwith host susceptibility to the development of GC. We investigated the role of host genetic factors including polymorphisms of IL-1B and IL-1RN in correlation with gastritis and GC in H. pylori infected Pakistani population. A total of 230 gastritis cases and 100 GC cases were genotyped for IL 1B-511 and IL- 1RN penta-allelic variable number of tandem repeats (VNTRs). A combination of IL-1B-511*T and IL-1RN*2 alleles (OR 19.064; 95% CI 2.319–156.7; p = 0.001) in H. pylori infected individuals had markedly increased risk of GC development. In Pakistani population, an increased risk of GC development is associated with the carriage of IL-1B- 511*T and IL-1RN*2 alleles. Synergistic effect of H. pylori infection and IL-1B-511*T/IL-1RN*2 genotypes was also observed in association with significantly higher risk of developing GC. Further prospective and large scale studies are needed to establish the clinical impact of these findings. C1 [Raza, Yasir] Deewan Farooq Medical Complex, Sindh Institute of Urology and Transplantation, Stem Cell Research Laboratory, Chand Bibi RoadKarachi, Pakistan. [Khan, Adnan] University of Karachi, Department of Microbiology, Immunology and Infectious Diseases Research LaboratoryKarachi, Pakistan. [Khan, Iqbal Asif] Dow University of Health and Sciences, Department of Molecular PathologyKarachi, Pakistan. [Khan, Saeed] Dow University of Health and Sciences, Department of Molecular PathologyKarachi, Pakistan. [Akhtar, Shakeel Syed] Civil Hospital Karachi, Department of Surgery and MedicineKarachi, Pakistan. [Mubarak, Muhammad] Sindh Institute of Urology and Transplantation, Department of HistopathologyKarachi, Pakistan. [Ahmed, Ayaz] Dr. Panjwani Center For Molecular Medicine And Drug ResearchKarachi, Pakistan. [Kazmi, Urooj Shahana] University of Karachi, Department of Microbiology, Immunology and Infectious Diseases Research LaboratoryKarachi, Pakistan. RP Raza, Y (reprint author), Deewan Farooq Medical Complex, Sindh Institute of Urology and Transplantation, Stem Cell Research Laboratory, Karachi, Pakistan. EM yaaasirz@hotmail.com CR Parsonnet J, 1995, The incidence of helicobacter pylori infection. Aliment Pharmacol Ther 9(Suppl 2):45–51 Uemura N, Okamoto S, Yamamoto S, Matsumura N, Yamaguchi S et al, 2001, Helicobacter pylori infection and the development of gastric cancer. N Engl J Med 345(11):784–789 Goh KL, Chan WK, Shiota S, Yamaoka Y, 2011, Epidemiology of Helicobacter pylori infection and public health implications. Helicobacter 16(Suppl 1):1–9., DOI 10.1111/j.1523-5378.2011.00874.x Mohri Y, Tanaka K, Ohi M, Yokoe T, Miki C et al, 2010, Prognostic significance of host- and tumor-related factors in patients with gastric cancer. World J Surg 34(2):285–290., DOI 10.1007/s00268-009-0302-1 El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH et al, 2000, Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature 404(6776):398–402 Stoicov C, Saffari R, Cai X, Hasyagar C, Houghton J, 2004, Molecular biology of gastric cancer: helicobacter infection and gastric adenocarcinoma: bacterial and host factors responsible for altered growth signaling. Gene 341:1–17 Ha DM, Forman D, Goh KL, Fock KM, Mitchell HM. Ethnic Differences in Dietary Intake and the Association between Dietary Intake and Gastric Cancer Risk in Chinese Subjects Resident in Malaysia. In: Van Toi V, Khoa TQD, eds. The Third International Conference on the Development of Biomedical Engineering in Vietnam: BME2010, 11a€“14 January, 2010, Ho Chi Minh City, Vietnam. Berlin, Heidelberg: Springer Berlin Heidelberg. pp. 255–258. Peleteiro B, Lunet N, Wen X, Afonso LP, Mendes N et al, 2012, Association between environmental factors and CDX2 expression in gastric cancer patients. Eur J Cancer Prev 21(5):423–431., DOI 10.1097/CEJ0b013e32834ef1aa Shin CM, Kim N, Lee HS, Lee DH, Kim JS et al, 2011, Intrafamilial aggregation of gastric cancer: a comprehensive approach including environmental factors, Helicobacter pylori virulence, and genetic susceptibility. Eur J Gastroenterol Hepatol 23(5): 411–417., DOI 10.1097/MEG0b013e328343b7f5 Peek RM Jr, Blaser MJ, 2002, Helicobacter pylori and gastrointestinal tract adenocarcinomas. Nat Rev Cancer 2(1):28–37 Takashima M, Furuta T, Hanai H, Sugimura H, Kaneko E, 2001, Effects of helicobacter pylori infection on gastric acid secretion and serum gastrin levels in Mongolian gerbils. Gut 48(6):765–773 Santtila S, Savinainen K, HurmeM(1998, Presence of the IL-1RA allele 2, IL1RN*2, is associated with enhanced IL-1beta production in vitro. Scand J Immunol 47(3):195–198 Furuta T, El-Omar EM, Xiao F, Shirai N, TakashimaMet al, 2002, Interleukin 1beta polymorphisms increase risk of hypochlorhydria and atrophic gastritis and reduce risk of duodenal ulcer recurrence in Japan. Gastroenterology 123(1):92–105 Dinarello CA, 1996, Biologic basis for interleukin-1 in disease. Blood 87(6):2095–2147 Hwang IR, Kodama T, Kikuchi S, Sakai K, Peterson LE et al, 2002, Effect of interleukin 1 polymorphisms on gastric mucosal interleukin 1beta production in helicobacter pylori infection. Gastroenterology 123(6):1793–1803 Kato S, Onda M, Yamada S, Matsuda N, Tokunaga A et al, 2001, Association of the interleukin-1 beta genetic polymorphism and gastric cancer risk in Japanese. J Gastroenterol 36(10):696–699 Rocha GA, Guerra JB, Rocha AM, Saraiva IE, da Silva DA et al, 2005, IL1RN polymorphic gene and cagA-positive status independently increase the risk of noncardia gastric carcinoma. Int J Cancer 115(5):678–683 El-Omar EM, 2001, The importance of interleukin 1I2 inHelicobacter pylori associated disease. Gut 48:743–747 Tarlow JK, Blakemore AI, Lennard A, Solari R, Hughes HN et al, 1993, Polymorphism in human IL-1 receptor antagonist gene intron 2 is caused by variable numbers of an 86-bp tandem repeat. Hum Genet 91(4):403–404 Wang P, Xia HH, Zhang JY, Dai LP, Xu XQ et al, 2007, Association of interleukin-1 gene polymorphisms with gastric cancer: a meta-analysis. Int J Cancer 120(3):552–562 Chen A, Li CN, Hsu PI, Lai KH, Tseng HH et al, 2004, Risks of interleukin-1 genetic polymorphisms and helicobacter pylori infection in the development of gastric cancer. Aliment Pharmacol Ther 20(2):203–211 El-Omar EM, Rabkin CS, Gammon MD, Vaughan TL, Risch HA et al, 2003, Increased risk of noncardia gastric cancer associated with proinflammatory cytokine gene polymorphisms. Gastroenterology 124(5):1193–1201 Garza-Gonzalez E, Bosques-Padilla FJ, El-Omar E, Hold G, Tijerina-Menchaca R et al, 2005, Role of the polymorphic IL-1B, IL-1RN and TNF-A genes in distal gastric cancer in Mexico. Int J Cancer 114(2):237–241 Machado JC, Pharoah P, Sousa S, Carvalho R, Oliveira C et al, 2001, Interleukin 1B and interleukin 1RN polymorphisms are associated with increased risk of gastric carcinoma. Gastroenterology 121(4):823–829 Machado JC, Figueiredo C, Canedo P, Pharoah P, Carvalho R et al, 2003, A proinflammatory genetic profile increases the risk for chronic atrophic gastritis and gastric carcinoma. Gastroenterology 125(2):364–371 Xia HH, Lam SK, Huang XR, Wong WM, Leung SY et al, 2004, Helicobacter pylori infection is associated with increased expression of macrophage migratory inhibitory factor–by epithelial cells, T cells, and macrophages–in gastric mucosa. J Infect Dis 190(2): 293–302 Dixon MF, Genta RM, Yardley JH, Correa P, 1996, Classification and grading of gastritis. The updated Sydney system. International workshop on the histopathology of gastritis, Houston 1994. Am J Surg Pathol 20(10):1161–1181 Raza Y, Khan A, Farooqui A, Mubarak M, Facista A et al, 2014, Oxidative DNA damage as a potential early biomarker of Helicobacter pylori associated carcinogenesis. Pathol Oncol Res 20(4):839–846., DOI 10.1007/s12253-014-9762-1 Kuipers EJ, Lundell L, Klinkenberg-Knol EC, Havu N, Festen HP et al, 1996, Atrophic gastritis and helicobacter pylori infection in patients with reflux esophagitis treated with omeprazole or fundoplication. N Engl J Med 334(16):1018–1022 Noach LA, Bosma NB, Jansen J, Hoek FJ, Van Deventer SJH et al, 1994, Mucosal tumor necrosis factor-or, interleukin-1/3, and interleukin-8 production in patients with helicobacter pylori infection. Scand J Gastroenterol 29:425–429 Jung HC, Kim JM, Song IS, Kim CY, 1997, Helicobacter pylori induces an array of pro-inflammatory cytokines in human gastric epithelial cells: quantification ofmRNA for interleukin-8, −1 alpha/ beta, granulocyte-macrophage colony-stimulating factor, monocyte chemoattractant protein-1 and tumour necrosis factor-alpha. J Gastroenterol Hepatol 12: 473-480. Vamvakopoulos J, Green C, Metcalfe S, 2002, Genetic control of IL-1β bioactivity through differential regulation of the IL-1 receptor antagonist. Eur J Immunol 32:2988–2996 Witkin SS, Gerber S, Ledger WJ, 2002, Influence of interleukin-1 receptor antagonist gene polymorphism on disease. Clin Infect Dis 34(2):204–209 Bidwell J, Keen L, Gallagher G, Kimberly R, Huizinga T et al, 1999, Cytokine gene polymorphism in human disease: on-line databases. Genes Immun 1(1):3–19 Figueiredo C, Machado JC, Pharoah P, Seruca R, Sousa S et al, 2002, Helicobacter pylori and interleukin 1 genotyping: an opportunity to identify high-risk individuals for gastric carcinoma. J Natl Cancer Inst 94(22):1680–1687 Ruzzo A, Graziano F, Pizzagalli F, Santini D, Battistelli V et al, 2005, Interleukin 1B gene, IL-1B, and interleukin 1 receptor antagonist gene, IL-1RN, polymorphisms in helicobacter pylorinegative gastric cancer of intestinal and diffuse histotype. Ann Oncol 16(6):887–892 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 873 EP 880 DI 10.1007/s12253-017-0191-9 PG 8 ER PT J AU Liu, K Wan, J Bei, Y Chen, X Lu, M AF Liu, Kaitai Wan, Juefeng Bei, Yanping Chen, Xue Lu, Miaozhen TI Prognostic Impact of Different Histological Types on Gastric Adenocarcinoma: a Surveillance, Epidemiology, and End Results Database Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric carcinoma; Mucinous adenocarcinoma; Signet ring cell carcinoma; Survival; SEER ID Gastric carcinoma; Mucinous adenocarcinoma; Signet ring cell carcinoma; Survival; SEER AB The clinicopathological characteristics and prognosis of gastric mucinous adenocarcinoma (MAC) and signet ring cell carcinoma (SRC) are still controversial.We designed our study to evaluate the clinicopathologic features and prognosis of MAC, SRC and ordinary gastric adenocarcinoma (OGAC) by analyzing the Surveillance, Epidemiology, and End Results (SEER)-registered database. The 5-year overall survival (OS) of patients with SRC was significantly lower than that of patients with MAC (P = 0.001) and OGAC (P < 0.001), and there was no significant difference in 5- year OS between MAC and OGAC (P = 0.804). Furthermore, there were no significant differences of 5-years OS among these three groups at stage I, II and III (all P > 0.05) and no significant difference between MAC and OGAC at stage IV (P = 0.110). Patients in SRC group had significantly worse survival than those in MAC and OGAC at stage IV (both P = 0.008), with 5-year OS of 3.3%, 5.8%, and 5.8%, respectively. However, the histological type was not found to be an independent prognostic factor of gastric cancer according to the multivariate analysis with Cox regression. C1 [Liu, Kaitai] Ningbo Medical Center, Lihuili Hospital, Department of Radiation Oncology, No. 57, Xing’Ning Road, 315041 Ningbo, China. [Wan, Juefeng] Fudan University, Shanghai Cancer Center, Department of Radiation OncologyShanghai, China. [Bei, Yanping] Ningbo Medical Center, Lihuili Hospital, Department of Radiation Oncology, No. 57, Xing’Ning Road, 315041 Ningbo, China. [Chen, Xue] Ningbo Medical Center, Lihuili Hospital, Department of Radiation Oncology, No. 57, Xing’Ning Road, 315041 Ningbo, China. [Lu, Miaozhen] Ningbo Medical Center, Lihuili Hospital, Department of Radiation Oncology, No. 57, Xing’Ning Road, 315041 Ningbo, China. RP Lu, M (reprint author), Ningbo Medical Center, Lihuili Hospital, Department of Radiation Oncology, 315041 Ningbo, China. EM lmz2005@yeah.net CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87–108 Parkin DM, Bray FI, Devesa SS, 2001, Cancer burden in the year 2000. The global picture. Eur J Cancer 37(8):S4–66 Bosman FT, Carneiro F, Hruban RH, Theise ND, 2010, WHO classification of Tumours of the digestive system, fourth edition. Int Agency Res Cancer 3:1089 Lauren P, 1965, The two main histological types of gastric carcinoma: diffuse and so called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand 64:31–49 Ribeiro MM, Sarmento JA, Simos SMA, Bastos J, 1981, Prognostic significance of Lauren and Ming classifications and other pathologic parameters in gastric carcinoma. Cancer 47:780– 784 Ming SC, 1977, Gastric carcinoma: a pathological classification. Cancer 39(6):2475–2485 Sugano H, Nakamura K, Kato Y, 1982, Pathological studies of human gastric cancers. Acta Pathol Jpn 32:329–347 Kawamura H, Kondo Y, Osawa S, Nisida Y, Okada K, Isizu H, Uebayasi T, Takahasi M, Hata T, 2001, A clinicopathologic study of mucinous adenocarcinoma of the stomach. Gastric Cancer 4:83–86 Wu CY, Yeh HZ, Shih RT, Chen GH, 1998, A clinicopathologic study of mucinous gastric carcinoma including multivariate analysis. Cancer 83:1312–1318 Hoerr SO, Hazard JB, Bailey D, 1966, Prognosis in carcinoma of the stomach in relation to the microscopic type. Surg Gynecol Obstet 122:485–494 Adachi Y, Yasuda K, Inomata M, Shiraishi N, Kitano S, Sugimachi K, 2001, Clinicopathologic study of early-stage mucinous gastric carcinoma. Cancer 91:698–703 Isobe T, Hashimoto K, Kizaki J, Matono S,Murakami N, Kinugasa T, Aoyagi K, Akagi Y, 2015, Characteristics and prognosis of mucinous gastric carcinoma. Mol Clin Oncol 3(1):44–50 Yin C, Li D, Sun Z, Zhang T, Xu Y, Wang Z, Xu H, 2012, Clinicopathologic features and prognosis analysis ofmucinous gastric carcinoma. Med Oncol 29(2):864–870 Otsuji E, Yamaguchi Y, Sawai K, Takahashi T, 1998, Characterization of signet ring cell carcinoma of the stomach. J Surg Oncol 67:216–220 Hyung WJ, Noh SH, Lee JH, Huh JJ, Lah KH, Choi SH, Min JS, 2002, Early gastric carcinoma with signet ring cell histology. Cancer 94:78–83 Jiang CG, Wang ZN, Sun Z, Liu FN, Yu M, Xu HM, 2011, Clinicopathologic characteristics and prognosis of signet ring cell carcinoma of the stomach: results from a Chinese monoinstitutional study. J Surg Oncol 103(7):700–703 Maehara Y, Sakaguchi Y, Moriguchi S, Orita H, Korenaga D, Kohnoe S, Sugimachi K, 1992, Signet ring cell carcinoma of the stomach. Cancer 69:1645–1650 Gronnier C, Messager M, Robb WB, Thiebot T, Louis D, Luc G, Piessen G, Mariette C, 2013, Is the negative prognostic impact of signet ring cell histology maintained in early gastric adenocarcinoma. Surgery 154(5):1093–1099 Chiu CT, Kuo CJ, Yeh TS, Hsu JT, Liu KH, Yeh CN, Hwang TL, Jan YY, Lin CJ, 2011, Early signet ring cell gastric cancer. Dig Dis Sci 56:1749–1756 Zhang M, Zhu G, Zhang H, Gao H, Xue Y, 2010, Clinicopathologic features of gastric carcinoma with signet ring cell histology. J Gastrointest Surg 14(4):601–606 Piessen G, Messager M, Leteurtre E, Jean-Pierre T, Mariette C, 2009, Signet ring cell histology is an independent predictor of poor prognosis in gastric adenocarcinoma regardless of tumoral clinical presentation. Ann Surg 250(6):878–887 Yang XF, Yang L, Mao XY, Wu DY, Zhang SM, Xin Y, 2004, Pathobiological behavior and molecular mechanism of signet ring cell carcinoma and mucinous adenocarcinoma of the stomach: a comparative study. World J Gastroenterol 10(5):750–754 Kunisaki C, Akiyama H, Nomura M, Matsuda G, Otsuka Y, Ono HA, Shimada H, 2006, Clinicopathologic characteristics and surgical outcomes of mucinous gastric carcinoma. Ann Surg Oncol 13: 836–842 Zhang M, Zhu GY, Zhang HF, Gao HY, Han XF, Xue YW, 2010, Clinicopathologic characteristics and prognosis ofmucinous gastric carcinoma. J Surg Oncol 102:64–67 Adachi Y, Mori M, Kido A, Shimono R, Maehara Y, Sugimachi K, 1992, A clinicopathologic study of mucinous gastric carcinoma. Cancer 69:866–871 Antonioli DA, Goldman H, 1982, Changes in the location and type of gastric carcinoma. Cancer 50:775–781 Theuer CP, Nastansk F, Brewster WR, Butler JA, Anton-Culver H, 1999, Signet ring cell histology is associated with unique clinical features but does not affect gastric cancer survival. Am Surg 65: 915–921 Kim JP, Kim SC, Yang HK, 1994, Prognostic significance of signet ring cell carcinoma of the stomach. Surg Oncol 50:775–781 Ryu SY, Kim HG, Lee JH, Kim DY, 2014, Prognosis of early mucinous gastric carcinoma. Ann Surg Treat Res 87(1):5–8 Matsuyama S, Ohkura Y, Eguchi H, Kobayashi Y, Akagi K, Uchida K, Nakachi K, Gustafsson JA, Hayashi S, 2002, Estrogen receptor beta is expressed in human stomach adenocarcinoma. J Cancer Res Clin Oncol 128(6):319–324 Kitaoka H, 1983, Sex hormone dependency and endocrine therapy in diffuse carcinoma of the stomach.Gan To Kagaku Ryoho 10(12): 2453–2460 Kunisaki C, Shimada H, Nomura M, Matsuda G, Otsuka Y, Akiyama H, 2004, Therapeutic strategy for signet ring cell carcinoma of the stomach. Br J Surg 91(10):1319–1324 Ha TK, An JY, Youn HK, Noh JH, Sohn TS, Kim S, 2008, Indication for endoscopicmucosal resection in early signet ring cell gastric cancer. Ann Surg Oncol 15(2):508–513 Lee JH, Choi IJ, Kook MC, Nam BH, Kim YW, Ryu KW, 2010, Risk factors for lymph node metastasis in patients with early gastric cancer and signet ring cell histology. Br J Surg 97(5):732–736 Li C, Kim S, Lai JF, Hyung WJ, Choi WH, Choi SH, Noh SH, 2007, Advanced gastric carcinoma with signet ring cell histology. Oncology 72(1–2):64–68 Kwon KJ, Shim KN, Song EM, Choi JY, Kim SE, Jung HK, Jung SA, 2014, Clinicopathological characteristics and prognosis of signet ring cell carcinoma of the stomach. Gastric Cancer 17(1):43–53 Kim DY, Park YK, Joo JK, Ryu SY, Kim YJ, Kim SK, Lee JH, 2004, Clinicopathological characteristics of signet ring cell carcinoma of the stomach. ANZ J Surg 74(12):1060–1064 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 881 EP 887 DI 10.1007/s12253-017-0198-2 PG 7 ER PT J AU Farkas, T Muller, J Erdelyi, JD Csoka, M Kovacs, TG AF Farkas, Tamas Muller, Judit Erdelyi, J Daniel Csoka, Monika Kovacs, T Gabor TI Absolute Lymphocyte Count (ALC) after Induction Treatment Predicts Survival of Pediatric Patients with Acute Lymphoblastic Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ALC; ALL; Survival ID ALC; ALL; Survival AB Absolute Lymphocyte Count (ALC) has been recently established as a prognostic factor of survival in pediatric Acute Lymphoblastic Leukemia (ALL). A retrospective analysis of 132 patients treated according the BFM – ALLIC 2002 protocol was performed in a single institution. A possible association between ALC values and Overall Survival (OS) or Event-Free Survival (EFS) was evaluated at multiple time points during induction chemotherapy. ALC higher than 350 cells/μL measured on the 33th day of induction was associated with better Overall- and Event-Free Survival in both Kaplan-Meier (OS 88.6% vs. 40%; p < 0.001 / EFS 81.6% vs. 30%; p < 0.001) and Cox regression (OS HR 8.77 (3.31– 23.28); p < 0.001) and EFS HR 6.61 (2.79–15.63); p < 0.001) analyses. There was no association between survival and measured ALC values from earlier time points (day of diagnosis, days 8 and 15) of induction therapy. Patients with low ALC values tend to have higher risk (MR or HR groups) and a higher age at diagnosis (>10 years). With help of day 33 ALC values of 350 cells/μL cutoff it was possible to refine day 33 flow cytometry (FC) Minimal Residual Disease (MRD) results within the negative cohort: higher ALC values were significantly associated with better survival. ALC on day 33 (350 cells/μL) remained prognostic for OS and EFS in multivariate analysis after adjusting it for age, cytogenetics, immunophenotype and FC MRD of induction day 33. According to these findings ALC on day 33 of induction is a strong predictor of survival in pediatric ALL. C1 [Farkas, Tamas] Semmelweis UniversityBudapest, Hungary. [Muller, Judit] Semmelweis UniversityBudapest, Hungary. [Erdelyi, J Daniel] Semmelweis UniversityBudapest, Hungary. [Csoka, Monika] Semmelweis UniversityBudapest, Hungary. [Kovacs, T Gabor] Semmelweis UniversityBudapest, Hungary. RP Kovacs, TG (reprint author), Semmelweis University, Budapest, Hungary. EM kovi2@hotmail.com CR Hunger SP, Lu X, Devidas M, Camitta BM, Gaynon PS, Winick NJ, Reaman GH, CarrollWL, 2012, Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report fromthe children’s oncology group. J Clin Oncol 30:1663–1669 Pui CH, Campana D, Pei D, BowmanWP, Sandlund JT, Kaste SC, Ribeiro RC, Rubnitz JE, Raimondi SC, OnciuM, Coustan-Smith E, Kun LE, Jeha S, Cheng C, Howard SC, Simmons V, Bayles A, Metzger ML, Boyett JM, Leung W, Handgretinger R, Downing JR, Evans WE, Relling MV, 2009, Treating childhood acute lymphoblastic leukemia without cranial irradiation. N Engl J Med 360: 2730–2741 Pui CH, Yang JJ, Hunger SP, Pieters R, Schrappe M, Biondi A, Vora A, Baruchel A, Silverman LB, Schmiegelow K, Escherich G, Horibe K, Benoit YC, Izraeli S, Yeoh AE, Liang DC, Downing JR, Evans WE, Relling MV, Mullighan CG, 2015, Childhood acute lymphoblastic leukemia: progress through collaboration. J Clin Oncol 33:2938–2948 Basso G, Veltroni M, Valsecchi MG, Dworzak MN, Ratei R, Silvestri D, Benetello A, Buldini B, Maglia O, Masera G, Conter V, Arico M, Biondi A, Gaipa G, 2009, Risk of relapse of childhood acute lymphoblastic leukemia is predicted by flow cytometric measurement of residual disease on day 15 bone marrow. J Clin Oncol 27:5168–5174 Gaipa G, Cazzaniga G, Valsecchi MG, Panzer-Grumayer R, Buldini B, Silvestri D, Karawajew L, Maglia O, Ratei R, Benetello A, Sala S, Schumich A, Schrauder A, Villa T, Veltroni M, Ludwig WD, Conter V, Schrappe M, Biondi A, Dworzak MN, Basso G, 2012, Time point-dependent concordance of flow cytometry and real-time quantitative polymerase chain reaction for minimal residual disease detection in childhood acute lymphoblastic leukemia. Haematologica 97:1582–1593 Stary J, ZimmermannM, CampbellM, Castillo L, Dibar E, Donska S, Gonzalez A, Izraeli S, Janic D, Jazbec J, Konja J, Kaiserova E, Kowalczyk J, Kovacs G, Li CK, Magyarosy E, Popa A, Stark B, Jabali Y, Trka J, Hrusak O, RiehmH, Masera G, SchrappeM(2014, Intensive chemotherapy for childhood acute lymphoblastic leukemia: results of the randomized intercontinental trial ALL IC-BFM 2002. J Clin Oncol 32:174–184 Anoceto Martinez A, Gonzalez Otero A, Guerchicoff de Svarch E, Arencibia Nunez A, Jaime JC, Dorticos E, Sarduy S, Gonzalez L, 2012, Absolute lymphocyte count as a prognostic factor in children with acute lymphoblastic leukemia. An Pediatr 76:10e1–10e6 Behl D, Porrata LF, Markovic SN, Letendre L, Pruthi RK, Hook CC, Tefferi A, Elliot MA, Kaufmann SH, Mesa RA, Litzow MR, 2006, Absolute lymphocyte count recovery after induction chemotherapy predicts superior survival in acute myelogenous leukemia. Leukemia 20:29–34 Cheng Y, Luo Z, Yang S, Jia M, Zhao H, XuW, Tang Y, 2015, The ratio of absolute lymphocyte count at interim of therapy to absolute lymphocyte count at diagnosis predicts survival in childhood Blineage acute lymphoblastic leukemia. Leuk Res 39:144–150 De Angulo G, Hernandez M, Morales-Arias J, Herzog CE, Anderson P, Wolff J, Kleinerman ES, 2007, Early lymphocyte recovery as a prognostic indicator for high-risk Ewing sarcoma. J Pediatr Hematol Oncol 29:48–52 De Angulo G, Yuen C, Palla SL, Anderson PM, Zweidler-McKay PA, 2008, Absolute lymphocyte count is a novel prognostic indicator in ALL and AML: implications for risk stratification and future studies. Cancer 112:407–415 Hatzipantelis E, Pana ZD, Vlachou M, Papageorgiou T, Tragiannidis A, Athanassiadou F, 2014, Peripheral blood lymphocyte recovery and overall survival in pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer 61:181–183 Rabin KR, Gramatges MM, Borowitz MJ, Palla SL, Shi X, Margolin JF, Zweidler-McKay PA, 2012, Absolute lymphocyte counts refine minimal residual disease-based risk stratification in childhood acute lymphoblastic leukemia. Pediatr Blood Cancer 59: 468–474 Rubnitz JE, Campbell P, Zhou Y, Sandlund JT, Jeha S, Ribeiro RC, Inaba H, Bhojwani D, Relling MV, Howard SC, Campana D, Pui CH, 2013, Prognostic impact of absolute lymphocyte counts at the end of remission induction in childhood acute lymphoblastic leukemia. Cancer 119:2061–2066 Shen HQ, Feng JH, Tang YM, Song H, Yang SL, Shi SW, Xu WQ, 2013, Absolute lymphocyte count is associated with minimal residual disease level in childhood B-cell precursor acute lymphoblastic leukemia. Leuk Res 37:671–674 Siddiqui M, Ristow K, Markovic SN, Witzig TE, Habermann TM, Colgan JP, Inwards DJ, White WL, Ansell SM, Micallef IN, Johnston PB, Call TG, Porrata LF, 2006, Absolute lymphocyte count predicts overall survival in follicular lymphomas. Br J Haematol 134:596–601 Sun D, Elson P, Liedtke M, Medeiros BC, Earl M, Alizadeh A, Bates J, Sekeres MA, Coutre S, Kalaycio M, Sobecks R, Copelan E, Advani AS, 2012, Absolute lymphocyte count at day 28 independently predicts event-free and overall survival in adults with newly diagnosed acute lymphoblastic leukemia. Am J Hematol 87:957–960 Gupta A, Kapoor G, Jain S, Bajpai R, 2015, Absolute lymphocyte count recovery independently predicts outcome in childhood acute lymphoblastic leukemia: experience from a tertiary care cancer Center of a Developing Country. J Pediatr Hematol Oncol 37: e143–e149 Ishaqi MK, Afzal S, Dupuis A, Doyle J, Gassas A, 2008, Early lymphocyte recovery post-allogeneic hematopoietic stem cell transplantation is associated with significant graft-versus-leukemia effect without increase in graft-versus-host disease in pediatric acute lymphoblastic leukemia. Bone Marrow Transplant 41:245–252 Porrata LF, Inwards DJ, Micallef IN, Ansell SM, Geyer SM, Markovic SN, 2002, Early lymphocyte recovery post-autologous haematopoietic stem cell transplantation is associated with better survival in Hodgkin’s disease. Br J Haematol 117:629–633 Porrata LF, Ingle JN, Litzow MR, Geyer S, Markovic SN, 2001, Prolonged survival associated with early lymphocyte recovery after autologous hematopoietic stem cell transplantation for patients with metastatic breast cancer. Bone Marrow Transplant 28:865–871 Tedeschi SK, Jagasia M, Engelhardt BG, Domm J, Kassim AA, Chinratanalab W, Greenhut SL, Goodman S, Greer JP, Schuening F, Frangoul H, Savani BN, 2011, Early lymphocyte reconstitution is associated with improved transplant outcome after cord blood transplantation. Cytotherapy 13:78–82 Benjamin JE, Gill S, Negrin RS, 2010, Biology and clinical effects of natural killer cells in allogeneic transplantation. Curr Opin Oncol 22:130–137 Norell H, Moretta A, Silva-Santos B, Moretta L, 2013, At the bench: preclinical rationale for exploitingNK cells and gammadelta T lymphocytes for the treatment of high-risk leukemias. J Leukoc Biol 94:1123–1139 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 889 EP 897 DI 10.1007/s12253-017-0192-8 PG 9 ER PT J AU Ma, G Li, Q Dai, W Yang, X Sang, A AF Ma, Gang Li, Qianjun Dai, Weijie Yang, Xiaozhong Sang, Aiyu TI Prognostic Implications of miR-302a/b/c/d in Human Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; miR-302a/b/c/d; Clinicopathological feature; Disease-free survival; Overall survival ID Gastric cancer; miR-302a/b/c/d; Clinicopathological feature; Disease-free survival; Overall survival AB Background: The microRNA (miR)-302 family consisting four members, miR-302a, miR-302b, miR-302c and miR-302d, plays an important role in diverse biological processes, and regulates many pathological changes, including cancer. However, the involvement of the miR-302 family into human gastric cancer (GC) remains unclear. The aim of this study was to investigate the expression patterns of miR- 302a/b/c/d and determine their clinical significance in GC. Materials and methods: Expression levels of miR-302a/b/c/d in 160 pairs of human GC and matched normal mucosa tissues were detected by quantitative real-time polymerase chain reaction. Then, the associations of miR-302a/b/c/d expression with various clinicopathological characteristics and patients’ prognosis were statistically evaluated. Results: The expression levels of miR-302a, miR-302b and miR-302c in GC tissues were all significantly lower than those in matched normal mucosa (all P < 0.001), but miR-302d expression had no significant differences between cancer and normal groups. Additionally, GC patients with low miR-302a, miR-302b and miR-302c expression more frequently had positive lymph node metastasis (all P < 0.05), advanced TNM stage (all P < 0.05) and great depth of invasion (all P < 0.05). More importantly, low miR-302a, miR-302b and miR-302c expression in GC tissues were significantly associated with shorter disease-free and overall survivals of GC patients (all P < 0.05). Further multivariate analysis identified miR-302a, miR-302b and miR-302c as independent prognostic markers for GC patients. Conclusions: GC patients with the decreased expression of miR-302a, miR-302b and miR-302c may had aggressive cancer progression and unfavorable prognosis. Further rigorous validation based on a large cohort of clinical cases should be performed. C1 [Ma, Gang] Nanjing Medical University, Huai’an First People’s Hospital, Department of Gastroenterology, 6 Beijing Road West, 223300 Huainan, Jiangsu, China. [Li, Qianjun] Nanjing Medical University, Huai’an First People’s Hospital, Department of Gastroenterology, 6 Beijing Road West, 223300 Huainan, Jiangsu, China. [Dai, Weijie] Nanjing Medical University, Huai’an First People’s Hospital, Department of Gastroenterology, 6 Beijing Road West, 223300 Huainan, Jiangsu, China. [Yang, Xiaozhong] Nanjing Medical University, Huai’an First People’s Hospital, Department of Gastroenterology, 6 Beijing Road West, 223300 Huainan, Jiangsu, China. [Sang, Aiyu] Lianshui Third People’s Hospital, Department of Internal Medicine, 12 Gaogouzhen 307 Road South, 223411 Lianshui, Jiangsu, China. RP Yang, X (reprint author), Nanjing Medical University, Huai’an First People’s Hospital, Department of Gastroenterology, 223300 Huainan, China. EM xzyangha@sina.com CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65:87–108 Colquhoun A, Arnold M, Ferlay J, Goodman KJ, Forman D, Soerjomataram I, 2015, Global patterns of cardia and non-cardia gastric cancer incidence in 2012. Gut 64(12):1881–1888 Hashimoto T, Arai K, Yamashita Y, Iwasaki Y, Hishima T, 2013, Characteristics of intramural metastasis in gastric cancer. Gastric Cancer 16:537–542 Kanda M, Kodera Y, 2015, Recent advances in the molecular diagnostics of gastric cancer. World J Gastroenterol 21:9838–9852 He Z, Li B, 2015, Recent progress in genetic and epigenetic profile of diffuse gastric cancer. Cancer Transl Med. 1:159532., DOI 10. 4103/2395-3977.159532 Yan X, Xu H, Yan Z, 2015, Functional perspective and implications of gene expression by noncoding RNAs. Cancer Transl Med 1:163805., DOI 10.4103/2395-3977.163805 Kuninty PR, Schnittert J, Storm G, Prakash J, 2016, MicroRNA targeting to modulate tumor microenvironment. Front Oncol 6:3 Arora A, Singh S, Bhatt AN, Pandey S, Sandhir R, Dwarakanath BS, 2015, Interplay between metabolism and oncogenic process: role of microRNAs. Transl Oncogenomics 7:11–27 Lin SL, Chang DC, Chang-Lin S, Lin CH,Wu DT, Chen DT, Ying SY, 2008, Mir-302 reprograms human skin cancer cells into a pluripotent ES-cell-like state. RNA 14:2115–2124 Lin SL, Chang DC, Ying SY, Leu D, Wu DT, 2010, MicroRNA miR-302 inhibits the tumorigenecity of human pluripotent stem cells by coordinate suppression of the CDK2 and CDK4/6 cell cycle pathways. Cancer Res 70:9473–9482 Zhao L,Wang Y, Jiang L, HeM, Bai X, Yu L,WeiM, 2016)MiR- 302a/b/c/d cooperatively sensitizes breast cancer cells to adriamycin via suppressing P-glycoprotein(P-gp, by targeting MAP/ERK kinase kinase 1, MEKK1). J Exp Clin Cancer Res 35:25 Guo T, YuW, Lv S, Zhang C, Tian Y, 2015)MiR-302a inhibits the tumorigenicity of ovarian cancer cells by suppression of SDC1. Int J Clin Exp Pathol 8:4869–4880 Wei ZJ, Tao ML, Zhang W, Han GD, Zhu ZC, Miao ZG, Li JY, Qiao ZB, 2015, Up-regulation of microRNA-302a inhibited the proliferation and invasion of colorectal cancer cells by regulation of the MAPK and PI3K/Akt signaling pathways. Int J Clin Exp Pathol 8:4481–4491 Cai D, He K, Chang S, Tong D, Huang C, 2015, MicroRNA-302b enhances the sensitivity of hepatocellular carcinoma cell lines to 5- FU via targeting mcl-1 and DPYD. Int J Mol Sci 16:23668–23682 Ge T, Yin M, Yang M, Liu T, Lou G, 2014, MicroRNA-302b suppresses human epithelial ovarian cancer cell growth by targeting RUNX1. Cell Physiol Biochem 34:2209–2220 Wang L, Yao J, Shi X, Hu L, Li Z, Song T, Huang C, 2013, MicroRNA-302b suppresses cell proliferation by targeting EGFR in human hepatocellular carcinoma SMMC-7721 cells. BMC Cancer 13:448 Chen L, Min L,Wang X, Zhao J, Chen H, Qin J, Chen W, Shen Z, Tang Z, Gan Q, Ruan Y, Sun Y, Qin X, Gu J, 2015, Loss ofRACK1 promotes metastasis of gastric cancer by inducing a miR-302c/IL8 signaling loop. Cancer Res 75:3832–3841 Khalili M, VaseiM, Khalili D, Alimoghaddam K, Sadeghizadeh M, Mowla SJ, 2015, Downregulation of the genes involved in reprogramming, SOX2, c-MYC, miR-302, miR-145, and P21, in gastric adenocarcinoma. J Gastrointest Cancer 46:251–258 Liu L, Lian J, Zhang H, Tian H, Liang M, Yin M, Sun F, 2013, MicroRNA-302a sensitizes testicular embryonal carcinoma cells to cisplatin-induced cell death. J Cell Physiol 228:2294–2304 Chen PH, Shih CM, Chang WC, Cheng CH, Lin CW, Ho KH, Su PC, Chen KC, 2014, MicroRNA-302b-inhibited E2F3 transcription factor is related to all trans retinoic acid-induced glioma cell apoptosis. J Neurochem 131:731–742 Tabrizi M, Khalili M, Vasei M, Nouraei N, Mansour Samaei N, Khavanin A, Khajehei M, Mowla SJ, 2015, Evaluating the miR- 302b and miR-145 expression in formalin-fixed paraffin-embedded samples of esophageal squamous cell carcinoma. Arch Iran Med 18:173–178 Cataldo A, Cheung DG, Balsari A, Tagliabue E, Coppola V, Iorio MV, Palmieri D, Croce CM, 2016, miR-302b enhances breast cancer cell sensitivity to cisplatin by regulating E2F1 and the cellular DNA damage response. Oncotarget 7:786–797 Wang Y,Wei Y, Tong H, Chen L, Fan Y, Ji Y, JiaW, Liu D,Wang G, 2015, MiR-302c-3p suppresses invasion and proliferation of glioma cells via down-regulating metadherin, MTDH, expression. Cancer Biol Ther 16:1308–1315 Zhu K, Pan Q, Jia LQ, Dai Z, Ke AW, Zeng HY, Tang ZY, Fan J, Zhou J, 2014, MiR-302c inhibits tumor growth of hepatocellular carcinoma by suppressing the endothelial-mesenchymal transition of endothelial cells. Sci Rep 4:5524 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2017 VL 23 IS 4 BP 899 EP 905 DI 10.1007/s12253-017-0282-7 PG 7 ER PT J AU Zalatnai, A Perjesi, E Galambos, E AF Zalatnai, Attila Perjesi, Eszter Galambos, Eszter TI Much More than Trousseau Syndrome. The Broad Spectrum of the Pancreatic Paraneoplastic Syndromes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Pancreatic cancer; Paraneoplastic syndromes; Trousseau syndrome; Hypercoagulability; Venous thromboembolism; Tissue factor; De novo diabetes mellitus ID Pancreatic cancer; Paraneoplastic syndromes; Trousseau syndrome; Hypercoagulability; Venous thromboembolism; Tissue factor; De novo diabetes mellitus AB When 150 years ago Armand Trousseau proposed that some thrombotic events might be the first sign of concealed visceral malignancies, these findings seemed to be just of anecdotal interest. Since then, however, we have learned that adenocarcinomas, including pancreatic cancers could be associated with a wide spectrum of paraneoplastic syndromes. They may precede the detection of the tumor, may occur simultaneously or may develop during its progression. Due to various hematologic, endocrine, cutaneous, articular, neuromuscular, renal or even psychiatric syndromes, their correct interpretation is intriguing, and because their early signs are not necessarily recognized first by oncologists, the paraneoplastic syndromes pose a diagnostic challenge. Unfortunately, we cannot generalize about their mechanisms, because the molecular backgrounds are far-reaching. In most of the cases, the pancreatic cancer cells release various factors into the bloodstreamtriggering the coagulation cascade. These patients frequently present with venous thromboembolism, and sometimes they are resistant to anticoagulation. The simultaneous thrombotic and bleeding evens do reflect the abnormal hemostasis. In other instances autoantibodies are formed against cutaneous, renal, neuromuscular or nervous tissues, but the mechanism of some syndromes remains unclear. Clinicians should be aware that pancreatic carcinoma may be associated with not just the Trousseau-syndrome. C1 [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Perjesi, Eszter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Galambos, Eszter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. RP Zalatnai, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM zalatnai@korb1.sote.hu CR Trousseau A, 1865, Phlegmasia alba dolens. In: The Sydenham Society, ed, Clinique Medicale de l’Hotel-Dieu de Paris, vol. 3, 2nd ed. Balliere, Paris, pp 654–712 Iodice S, Gandini S, Lohr M, Lowenfels AB, Maisonneuve P, 2008, Venous thromboembolic events and organ-specific occult cancers: a review andmeta-analysis. J Thromb Haemost 6:781–788 Walker AJ, Card TR,West J, Crooks C, Grainge MJ, 2013, Incidence of venous thromboembolism in patients with cancer. A cohort study using linked United Kingdom databases. Eur J Cancer 49:1404–1413 Petterson TM, Marks RS, Ashrani AA, Bailey KR, Heit JA, 2015, Risk of site-specific cancer in incident venous thromboembolism: a population-based study. Thromb Res 135:472–478 Fahrig C, Heidrich H, Penninger C, 1998, The incidence of occult malignant diseases in patients with deep venous thrombosis of the pelvis and lower limb. Int J Angiol 7:249–251 Sallah S,Wan JY,Nguyen NP, 2002, Venous thrombosis in patients with solid tumors: determination of frequency and characteristics. Thromb Haemost 87:575–579 Kakkar AK, Levine M, Pinedo HM, Wolff R, Wong J, 2003, Venous thrombosis in cancer patients: insights from the FRONTLINE survey. Oncologist 8:381–388 Hillen HFP, 2000, Thrombosis in cancer patients. Ann Oncol 11(Suppl. 3):273–276 Hisada Y, Geddings JE, Ay C, Mackman N, 2015, Venous thrombosis and cancer: from mouse models to clinical trials. J Thromb Haemost 13:1372–1382 Khorana AA, Fine RL, 2004, Pancreatic cancer and thromboembolic disease. Lancet Oncol 5:655–663 Oh SY, Kim JH, Lee KW, Bang SM, Hwang JH, Oh D, Lee JS, 2008, Venous thromboembolism in patients with pancreatic adenocarcinoma: lower incidence in Asian ethnicity. Thromb Res 122: 485–490 Blastik M, Plavecz E, Zalatnai A, 2011, Pancreatic carcinomas in a 60-year, institute-based autopsy material with special emphasis of the metastatic pattern. Pancreas 40:478–480 Ogren M, Bergqvist D, Wahlander K, Eriksson H, Sternby NH, 2006, Trousseau’s syndrome – what is the evidence? A population-based autopsy study. Thromb Haemost 95:541–545 Monreal M, Fernandez-Llamarez J, Perandreu J, Urritia A, Sahuquillo JC, 1997, Occult cancer in patients with venous thromboembolism: which patients, which cancers. Thromb Haemost 78: 1316–1318 Oster MW, 1976, Thrombophlebitis and cancer. A review Angiology 27:557–567 Thayalasekaran S, Liddicoat H, Wood E, 2009, Thrombophlebitis migrans in a man with pancreatic adenocarcinoma: a case report. Cases J 2:6610 Jameson GS, Ramanathan RK, Borad MJ, Downhour M, Korn R, Von Hoff D, 2009, Marantic endocarditis associated with pancreatic cancer: a case series. Case Rep Gastroenterol 3:67–71 Piovanelli B, Rovetta R, Bonadei I, Vizzardi E, D’Aloia A, Metra M, 2013, Nonbacterial thrombotic endocarditis in pancreatic cancer. Monaldi Arch Chest Dis 80:189–192 Pathanjali Sharma PV, Babu SC, Shah PM, Seirafi R, Clauss RH, 1985, Arterial thrombosis and embolism in malignancy. J Cardiovasc Surg 26:479–483 Rigdon EE, 2000, Trousseau’s syndrome and acute arterial thrombosis. Cardiovasc Surg 8:214–218 Schattner A, Klepfish A, Huszar M, Shani A, 2002, Two patients with arterial thromboembolism among 311 patients with adenocarcinoma of the pancreas. Am J Med Sci 324:335–338 Mast C, Ramanathan RK, Feinstein DI, Rosen P, 2014, Disseminated intravascular coagulation secondary to advanced pancreatic cancer treated successfully with combination chemotherapy. Oncology 87:266–269 Kus T, Kalender ME, Aktas G, Suner A, Camci C, 2015, Atypical presentation of disseminated intravascular coagulation with synchronous peripheral venous thromboembolism and arterial gangrene in a pancreatic cancer patient: a case report. JOP 16:195–197 Varki A, 2007, Trousseau’s syndrome: multiple definitions and multiple mechanisms. Blood 110:1723–1729 Shah MM, Saif MW, 2010, Pancreatic cancer and thrombosis. Highlights from the "2010 ASCO annual meeting", Chicago, IL, USA, June 4-8, 2010. J Pancreas 11:331–333 Elyamany G, Alzahrani AM, Bukhary E, 2014, Cancer-associated thrombosis: an overview. Clin Med Insights Oncol 8:129–137 Sun W, Ren H, Gao CT, Ma WD, Luo L, Liu Y, Jin P, Hao JH, 2015, Clinical and prognostic significance of coagulation assays in pancreatic cancer patients with absence of venous thromboembolism. Am J Clin Oncol 38(6):550–556 Suzuki K, Aiura K, Kitagou M, Hoshimoto S, Takahashi S, Ueda M, Kitajima M, 2004, Platelets counts closely correlate with the disease-free survival interval of pancreatic cancer patients. Hepato- Gastroenterology 51:847–853 Chadha AS, Kocak-Uzel E, Das P, Minsky BD, Delclos ME, Mahmood U, Guha S, Ahmad M, Varadhachary GR, Javle M, Katz MH, Fleming JB, Wolff RA, Crane CH, Krishnan S, 2015, Paraneoplastic thrombocytosis independently predicts poor prognosis in patients with locally advanced pancreatic cancer. Acta Oncol 22:1–8 Heinmoller E, Scropp T, Kisker O, Simon B, Seitz R, Weinel RJ, 1995, Tumor cell-induced platelet aggregation in vitro by human pancreatic cancer cell lines. Scand J Gastroenterol 30:1008–1016 Uematsu T, Tsuchie K, Ukai K, Kimoto E, Funakawa T, Mizuno R, 1996, Granulocyte-colony stimulating factor produced by pancreatic carcinoma. Int J Pancreatol 19:135–139 Wahrenbrock M, Borsig L, Le D, Varki N, Varki A, 2003, Selectinmucin interactions as a possible molecular explanation for the association of trousseau syndrome with mucinous adenocarcinoma. J Clin Invest 112:853–862 Haas SL, Jesnowski R, SteinterM, Hummel F, Ringel J, Burstein C, Nizze H, Liebe S, Lohr JM, 2006, Expression of tissue factor in pancreatic adenocarcinoma is associated with activation of coagulation. World J Gastroenterol 12:4843–4849 Khorana AA, Ahrendt SA, Ryan CK, Francis CW, Hruban RH, Hu YC, Hostetter G, Harvey J, Taubman MB, 2007, Tissue factor expression, angiogenesis, and thrombosis in pancreatic cancer. Clin Cancer Res 13:2870–2875 Geddings JE, Mackman N, 2013, Tumor-derived tissue-factorpositive microparticles and venous thrombosis in cancer patients. Blood 122:1873–1880 Wang JG, Geddings JE, Aleman MM, Cardenas JC, Chantrathammachart P, Williams JC, Kirchhofer D, Bogdanov VY, Bach RR, Rak J, Church FC, Wolberg AS, Pawlinski R, Key NS, Yeh JJ, Mackman N, 2012, Tumor-derived tissue factor activates coagulation and enhances thrombosis in a mouse xenograft model of human pancreatic cancer. Blood 119:5543–5552 Zwicker JI, Liebman HA, Neuberg D, Lacroix R, Bauer KA, Furie BC, Furie B, 2009, Tumor-derived tissue factor-bearing microparticles are associated with venous thromboembolic events in malignancy. Clin Cancer Res 15:6830–6840 Thaler J, Ay C, Macman N, Bertina RM, Kaider A, Marosi C, Key NS, Barcel DA, Scheithauer W, Kornek G, Zielinski C, Pabiger I, 2012, Microparticle-associated tissue factor activity, venous thromboembolism and mortality in pancreatic, gastric, colorectal and brain cancer patients. J Thromb Haemost 10:1363–1370 Delluc A, Rousseau A, Delluc C, Le Moigne E, Le Gal G, Mottier D, Van Dreden P, Lacut K, 2011, Venous thromboembolism in patients with pancreatic cancer: implication of circulating tissue factor. Blood Coagul Fibrinolysis 22:295–300 Blom JW, Osanto S, Rosendaal FR, 2006, High risk of venous thrombosis in patients with pancreatic cancer: a cohort study of 202 patients. Eur J Cancer 42:410–414 Lyman GH, Eckert L, Wang Y, Wang H, Cohen A, 2013, Venous thromboembolism risk in patients with cancer receiving chemotherapy: a real-world analysis. Oncologist 18:1321–1329 Swieboda-Sadlej A, Kraj L, Krawczyk J, Nita E, Dwilewicz- Trojaczek J, 2012, Thrombocytosis in patients with pancreatic cancer treated with gemcitabine – does it have clinical significance? Description of 6 cases. Contemp Oncol, Pozn, 16:353–355 Kvolik S, Jukic M, Matijevic M, Marjanovic K, Glavas-Obravac L, 2010, An overview of coagulation disorders in cancer patients. Surg Oncol 19:e33–e46 Sah RP, Nagpal SJS, Mukhopadhyay D, Chari ST, 2013, New insights into pancreatic cancer-induced paraneoplastic diabetes. Nat Rev Gastroenterol Hepatol 10:423–433 Pannala R, Basu A, Petersen GM, Chari ST, 2009, New-onset diabetes: a potential clue to the early diagnosis of pancreatic cancer. Lancet Oncol 10:88–95 Javeed N, Sagar G, Dutta SK, Smyrk TC, Lau JS, Bhattacharya S, Truty M, Petersen GM, Kaufman RJ, Chari ST, Mukhopadhyay D, 2015, Pancreatic cancer-derived exosomes cause paraneoplastic β-cell dysfunction. Clin Cancer Res 21(7):1722–1733 Permert J, Larsson J, Westermark GT, Herrington MK, Christmanson L, Pour PM, Westermark P, Adrian TE, 1994, Islet amyloid polypeptide in patients with pancreatic cancer and diabetes. N Engl J Med 330:313–318 Batabayal P, Hoorn SV, Christophi C, Nikfarjam A, 2014, Association of diabetes mellitus and pancreatic adenocarcinoma: a meta-analysis of 88 studies. Ann Surg Oncol 21:2453–2462 Sturrock ND, Selby C, Hosking DJ, 1997, Spontaneous hypoglycaemia in a noninsulin-dependent diabetes mellitus patient with disseminated pancreatic carcinoma. Diabet Med 14:324–326 Ma RCW, Lo RSK, Tai MHL, Chan JCN, Chow CC, Woo JLF, 2006, Recurrent hypoglycaemia in a patient with metastatic pancreatic carcinoma. PLoS Med 3:e331 Cavestro GM, Mantovani M, Coruzzi P, Nouvenne A, Marcucci F, Franze A, DiMario F, Okolicsanyi L, 2002, Hypercalcaemia due to ectopic secretion of parathyroid related protein from pancreatic carcinoma: a case report. Acta Biomed 73:37–40 Rasnake MS, Glanton C, Omstein D, Osswald M, Garrison M, 2001, Hypercalcemia mediated by parathyroid hormone-related protein as an early manifestation of pancreatic adenocarcinoma metastasis: a case report. Am J Clin Oncol 24:416–417 Kobayashi N, Higurashi T, Iida H, Mawatari H, Endo H, Nozaki Y, Tomimoto A, Yoneda K, Akiyama T, Fujita K, Takahashi H, Yoneda M, Inamori M, Abe Y, Kirikoshi H, Kubota K, Saito S, Ueno N, Nakajima A, Yamanaka S, Inayama Y, 2008, Adenosquamous carcinoma of the pancreas associated with humoral hypercalcemia of malignancy, HHM). J Hepato-Biliary-Pancreat Surg 15:531–535 Ulla JL, Garcia-Doval I, Posada C, Soto S, Alvarez C, Ledo L, Vazquez-Astray E, 2008, Plantar keratoderma as a presenting sign of pancreatic adenocarcinoma. J Clin Ultrasound 36:108–109 Chee C, 2009, Panniculitis in a patient presenting with a pancreatic tumour and polyarthritis: a case report. J Med Case Rep 3:7331 Meier SB, Reinhart WH, 2002, Nodulare pannikulitis - ein paraneoplastisches Syndrom bei Pankreaskarzinom. Praxis 91: 1169–1172 Cox NH, Ramsay B, Dobson C, Comaish JS, 1996, Woody hands in a patient with pancreatic carcinoma: a variant of cancerassociated fasciitis–panniculitis syndrome. Br J Dermatol 135: 995–998 Matz H, Milner Y, Frusic-Zlotkin M, 1997, Brenner S. Paraneoplastic pemphigus associated with pancreatic carcinoma Acta Derm Venerol 77:289–291 Durden FM, Variyam E, Chren MM, 1996, Fat necrosis with features of erythema nodosum in a patient with metastatic pancreatic carcinoma. Int J Dermatol 35:39–41 Moreland LW, ed,, 2004, Palmar fasciitis. In: rheumatology and immunology therapy. Springer-Verlag, Berlin Heidelberg, p 682 Haroon M, PhelanM(2008, A paraneoplastic case of palmar fasciitis and polyarthritis syndrome. Nat Clin Pract Rheumatol 4:274–277 O’Neal KD, Butnor KJ, Perkinson KR, Proia AD, 2003, Bilateral diffuse uveal melanocytic proliferation associated with pancreatic carcinoma: a case report and literature review of this paraneoplastic syndrome. Surv Ophthalmol 48:613–625 Kumar S, Sethi S, Irani F, Bode BY, 2009, Anticyclic citrullinated peptide antibody-positive paraneoplastic polyarthritis in a patient with metastatic pancreatic cancer. Am J Med Sci 338:511–512 Veitch D, Tsai T, Joshua F, 2013, Palmar fasciitis and polyarthritis syndrome in pancreatic carcinoma. J Clin Rheumatol 19:203–205 Syrios J, Kechagias G, Xynos ID, Gamaletsou MN, Papageorgiou A, Agrogiannis G, Tsavarin N, 2011, Pancreatic adenocarcinomaassociated polymyositis treated with corticosteroids along with specific treatment. BMC Gastroenterol 11:33 Amroun KL, De Mestier L, Deguelte-Lardiere S, Diebold MD, Bouche O, Kianmanesh R, 2012, Pancreas cancer-associated polym y o s i t i s o f t h e l e g s r e g r e s s i n g a f t e r c e p h a l i c duodenopancreatectomy: case report and review of the literature. JOP 13:674–676 Aggarwal R, Oddis CV, 2011, Paraneoplastic myalgias and myositis. Rheum Dis Clin N Am 37:607–621 Kida Y, Maeshima E, Furukawa K, Ichikawa T, Goda M, Ichinose M, 2007, A case of polymyositis with a significantly high level of KL-6 associated with pancreatic cancer. Mod Rheumatol 17:262– 264 Caras S, Laurie S, Cronk W, Tompkins W, Brashear R, McCallum RW, 1996, Case report: pancreatic cancer presenting with paraneoplastic gastroparesis. Am J Med Sci 312:34–36 Wray SH, Dalmau J, Chen A, King S, Leigh RJ, 2011, Paraneoplastic disorders of eye movements. Ann N Y Acad Sci 1233:279–284 Aggarwal A, Williams D, 1997, Opsoclonus as a paraneoplastic manifestation of pancreatic carcinoma. J Neurol Neurosurg Psychiatry 63:687–688 Whelan TV, Hirszel P, 1988, Minimal-change nephropathy associated with pancreatic carcinoma. Arch Intern Med 148:975–976 Mayr M, Schmid RM, 2010, Pancreatic cancer and depression: myth and truth. BMC Cancer 10:569 Cosci F, Fava GA, Sonino N, 2015)Mood and anxiety disorders as early manifestation of medical illness: a systematic review. Psychother Psychosom 84:22–29 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 1 EP 10 DI 10.1007/s12253-017-0206-6 PG 10 ER PT J AU Min, H Sun, X Yang, X Zhu, H Liu, J Wang, Y Chen, G Sun, X AF Min, Hua Sun, Xiangdong Yang, Xi Zhu, Hongcheng Liu, Jia Wang, Yuandong Chen, Guangzong Sun, Xinchen TI Exosomes Derived from Irradiated Esophageal Carcinoma-Infiltrating T Cells Promote Metastasis by Inducing the Epithelial–Mesenchymal Transition in Esophageal Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tcells; Radiotherapy; Exosome; Esophageal carcinoma; Epithelial–mesenchymal transition; Metastasis ID Tcells; Radiotherapy; Exosome; Esophageal carcinoma; Epithelial–mesenchymal transition; Metastasis AB Exosomes are nanovesicles derived from tumor and normal cells that are detectable in human biological fluids, such as plasma, and cell culture supernatants. The function of exosome secretion from "normal" cells is unclear. Although numerous studies have investigated exosomes derived from hematopoietic cells, little is known regarding exosomes from T cells, even though these cells play significant roles in innate and acquired immunity. A CCK-8 assay was used to examine the ability of exosomes to inhibit TE13 cell proliferation. In vitro invasion and wound healing assays were conducted to explore the effects of exosomes on TE13 cell migration and invasion. A Western blottinganalys is was performed to investigate the effects of exosomes on the expression of the EMT-related moleculesβ-catenin, NF-κB and snail. This study aimed to investigate the effects of exosomes from irradiated T cells on the human esophageal squamous cell carcinoma (ESCC) cell line TE13 and revealed that exosomes inhibit the proliferation but promote the metastasis of TE13 cells in a dose-and time-dependent manner. Furthermore, exosomes significantly increased the expression of β-catenin, NF-κB and snail in TE13 cells. The results of this study suggest an important role for T cell-derived exosomes in the progression of esophageal carcinoma: T cell-derived exosomes promote esophageal cancer metastasis, likely by promoting the EMT through the upregulation of β-catenin and the NF-κB/snail pathway. Moreover, this study supports the use of exosomes as a nearly perfect example of biomimetic nanovesicles that could be utilized in future therapeutic strategies against various diseases, including cancer. C1 [Min, Hua] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, Jiangsu Province, China. [Sun, Xiangdong] The 81st Hospital of PLA, Department of Radiotherapy, 210000 Nanjing, Jiangsu Province, China. [Yang, Xi] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, Jiangsu Province, China. [Zhu, Hongcheng] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, Jiangsu Province, China. [Liu, Jia] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, Jiangsu Province, China. [Wang, Yuandong] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, Jiangsu Province, China. [Chen, Guangzong] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, Jiangsu Province, China. [Sun, Xinchen] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, Jiangsu Province, China. RP Sun, X (reprint author), The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, China. EM sunxinchen2012@163.com CR Yu S,Yang CS, Li J,YouW, Chen J, CaoY, Dong Z, QiaoY, 2015, Cancer prevention research in China. Cancer Prev Res, Phila, 8: 662–674., DOI 10.1158/1940-6207.CAPR-14-0469 D’Journo XB, Thomas PA, 2014, Current management of esophageal cancer. J Thorac Dis 6(Suppl 2):S253–S264., DOI 10.3978/j. issn.2072-1439.2014.04.16 Polyak K, Weinberg RA, 2009, Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer 9:265–273., DOI 10.1038/nrc2620 Chen X, Zhang H, Zhu H, Yang X, Yang Y, Yang Y, Min H, Chen G, Liu J, Lu J, Cheng H, Sun X, 2016, Endostatin combined with radiotherapy suppresses vasculogenic mimicry formation through inhibition of epithelial-mesenchymal transition in esophageal cancer. Tumour Biol 37:4679–4688., DOI 10.1007/s13277-015-4284-3 Ehrlich P, 1909, Nederlandsch Tijdschrift voor Geneeskunde: Ueber den jetzigne Stand Der Karzinomforchung. Weekblad Jaargang Eerst helft 5:273 Dougan M, Dranoff G, 2009, Immune therapy for cancer. Annu Rev Immunol 2 7 : 8 3 –11 7 . d o i : 1 0 . 11 4 6 / a n n u r e v. immunol.021908.132544 Schreiber RD, Old LJ, Smyth MJ, 2011, Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331:1565–1570., DOI 10.1126/science.1203486 DemariaS NB, Devitt ML, Babb JS, Kawashima N, Liebes L, Formenti SC, 2004, Ionizing radiation inhibition of distant untreated tumors, abscopal effect, is immune mediated. Int J Radiat Oncol Biol Phys 58:862–870., DOI 10.1016/j.ijrobp.2003.09.012 Levy C, Chargari C,Marabellea P, Perfettini JL, Magne N, Deutsch E, 2016, Can immunostimulatory agents enhance the abscopal effect of radiotherapy? Eur J Cancer 62:36–45., DOI 10.1016/j. ejca.2016.03.067 Zhang H, Xie Y, LiW, Chibbar R, Xiong S, Xiang J, 2011, CD4, +, T cell-released exosomes inhibit CD8(+, cytotoxic T-lymphocyte responses and antitumor immunity. Cell Mol Immunol 8:23–30., DOI 10.1038/cmi.2010.59 Colombo M, Thery C, 2014, Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol 30:255–289., DOI 10.1146/annurev-cellbio- 101512-122326 Tang XJ, Sun XY, Huang KM, Zhang L, Yang ZS, Zou DD,Wang B, Warnock GL, Dai LJ, Luo J, 2015, Therapeutic potential of CAR-T cell-derived exosomes: a cell-free modality for targeted cancer therapy. Oncotarget 6:44179–44190., DOI 10.18632 /oncotarget.6175 Pitt JM, CharrierM, Viaud S,Andre F, Besse B, Chaput N, Zitvogel L, 2014, Dendritic cell-derived exosomes as immunotherapies in the fight against cancer. J Immunol 193:1006–1011., DOI 10.4049 /jimmunol.1400703 Naslund TI, Gehrmann U, Qazi KR, Karlsson MC, Gabrielsson S, 2013, Dendritic cell-derived exosomes need to activate both T and B cells to induce antitumor immunity. J Immunol 190:2712–2719., DOI 10.4049/jimmunol.1203082 Liu H, GaoW, Yuan J,Wu C, Yao K, Zhang L, Ma L, Zhu J, Zou Y, Ge J, 2016, Exosomes derived from dendritic cells improve cardiac function via activation of CD4(+, T lymphocytes after myocardial infarction. J Mol Cell Cardiol 91:123–133., DOI 10.1016/j. yjmcc.2015.12.028 Zitvogel R, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, Ricciardi-Castagnoli P, Raposo G, Amigorena S, 1998, Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med 4:594–600., DOI 10.1038 /nm0598-594 Munich S, Sobo-Vujanovic A, Buchser WJ, Beer-Stolz D, Vujanovic NL, 2012, Dendritic cell exosomes directly kill tumor cells and activate natural killer cells via TNF superfamily ligands. Oncoimmunology 1:1074–1083., DOI 10.4161/onci.20897 Arscott WT, Tandle AT, Zhao S, Shabason JE, Gordon IK, Schlaff CD, Zhang G, Tofilon PJ, Camphausen KA, 2013, Ionizing radiation and glioblastoma exosomes: implications in tumor biology and cell migration. Transl Oncol 6:638–IN6., DOI 10.1593/tlo.13640 Lehmann BD, Paine MS, BrooksAM,McCubrey JA, Renegar RH, Wang R, Terrian DM, 2008, Senescence-associated exosome release from human prostate cancer cells. Cancer Res 68:7864– 7871., DOI 10.1158/0008-5472.CAN-07-6538 Kalluri R, Weinberg RA, 2009, The basics of epithelialmesenchymal transition. J Clin Invest 119:1420–1428., DOI 10.1172/JCI39104 Qin Y, Zhao D, Zhou HG,Wang XH, ZhongWL, Chen S, GuWG, Wang W, Zhang CH, Liu YR, Liu HJ, Zhang Q, Guo YQ, Sun T, Yang C, 2016, Apigenin inhibits NF-κB and snail signaling, EMT and metastasis in human hepatocellular carcinoma. Oncotarget., DOI 10.18632/oncotarget.9404 Lamb R, Ablett MP, Spence K, Landberg G, Sims AH, Clarke RB, 2013, Wnt pathway activity in breast cancer sub-types and stemlike cells. PLoS One 8:e67811., DOI 10.1371/journal.pone.0067811 MAX, 2016, YANW, DAIZ, GAOX, MAYet al.Baicalein suppresses metastasis of breast cancer cells by inhibiting eMT via downregulation of saTB1 and Wnt/β-catenin pathway. Drug Des Dev Ther 10:1419–1441., DOI 10.2147/DDDT.S102541 Denzer K, Kleijmeer MJ, Heijnen HF, Stoorvogel W, Geuze HJ, 2000, Exosome: from internal vesicle of the multivesicular body to intercellular signaling device. J Cell Sci 113:3365–3374 Cai Z, Yang F, Yu L, Yu Z, Jiang L,Wang Q, Yang Y,Wang L, Cao X, Wang J, 2012, Activated T cell exosomes promote tumor invasion via Fas signaling pathway. J Immunol 188:5954–5961., DOI 10.4049/jimmunol.1103466 Liang B, Peng P, Chen S, Li L, Zhang M, Cao D, Yang J, Li H, Gui T, Li X, Shen K, 2013, Characterization and proteomic analysis of ovarian cancer-derived exosomes. J Proteome 80:171–182., DOI 10.1016/j.jprot.2012.12.029 Raimondo F, Morosi L, Chinello C, Magni F, Pitto M, 2011, Advances inmembranous vesicle and exosome proteomics improving biological understanding and biomarker discovery. Proteomics 11:709–720., DOI 10.1002/pmic.201000422 Yu S, Cao H, Shen B, Feng J, 2015, Tumor-derived exosomes in cancer progression and treatment failure. Oncotarget 6:37151– 37168., DOI 10.18632/oncotarget.6022 Thery C, Duban L, Segura E, Veron P, Lantz O, Amigorena S, 2002, Indirect activation of naive CD4+ T cells by dendritic cellderived exosomes. Nat Immunol 3:1156–1162., DOI 10.1038/ni854 Escudier B, Dorval T, Chaput N, Andre F, Caby MP, Novault S, Flament C, Leboulaire C, Borg C, Amigorena S, Boccaccio C, Bonnerot C, Dhellin O, Movassagh M, Piperno S, Robert C, Serra V, Valente N, Le Pecq JB, Spatz A, Lantz O, Tursz T, Angevin E, Zitvogel L, 2005, Vaccination of metastatic melanoma patients with autologous dendritic cell, DC, derived-exosomes: results of thefirst phase I clinical trial. J Transl Med 3:10 Morse MA, Garst J, Osada T, Khan S, Hobeika A, Clay TM, Valente N, Shreeniwas R, Sutton MA, Delcayre A, Hsu DH, Le Pecq JB, Lyerly HK, 2005, A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J Transl Med 3:1 Pitt JM, Andre F, Amigorena S, Soria JC, Eggermont A, Kroemer G, Zitvogel L, 2016, Dendritic cell-derived exosomes for cancer therapy. J Clin Invest 126:1224–1232., DOI 10.1172/JCI81137 Peters PJ, Borst J, Oorschot V, Fukuda M, Krahenbuhl O, Tschopp J, Slot JW, Geuze HJ, 1991, Cytotoxic T lymphocyte granules are secretory lysosomes, containing both perforin and granzymes. J Exp Med 173:1099–1109., DOI 10.1084/jem.173.5.1099 Zagury D, Bernard J, Thierness N, Feldman M, Berke G, 1975, Isolation and characterization of individual functionally reactive cytotoxic T lymphocytes: conjugation, killing and recycling at the single cell level. Eur J Immunol 5:818–822., DOI 10.1002 /eji.1830051205 Peters PJ, Geuze HJ, Van der Donk HA, Slot JW, Griffith JM, Stam NJ, Clevers HC, Borst J, 1989)Molecules relevant for T cell-target cell interaction are present in cytolytic granules of human T lymphocytes. Eur J Immunol 19:1469–1475., DOI 10.1002 /eji.1830190819 Young JD, Nathan CF, Podack ER, PalladinoMA, Cohn ZA, 1986, Functional channel formation associated with cytotoxic T-cell granules. Proc Natl Acad Sci U S A 83:150–154., DOI 10.1073 /pnas.83.1.150 Young JD, Cohn ZA, 1987, Cellular and humoral mechanisms of cytotoxicity: structural and functional analogies. Adv Immunol 41: 269–332., DOI 10.1016/S0065-2776(08)60033-4 Henkart PA, Millard PJ, Reynolds CW, Henkart MP, 1984, Cytolytic activity of purified cytoplasmic granules from cytotoxic rat large granular lymphocyte tumors. J Exp Med 160:75–93., DOI 10.1084/jem.160.1.75 Podack ER, Konigsberg PJ, 1984, Cytolytic T cell granules. Isolation, structural, biochemical, and functional characterization. J Exp Med 160:695–710 van Zijl F, Mall S, Machat G, Pirker C, Zeillinger R, Weinhaeusel A, Bilban M, Berger W, Mikulits W, 2011, A human model of epithelial to mesenchymal transition to monitor drug efficacy in hepatocellular carcinoma progression. Mol Cancer Ther 10:850– 860., DOI 10.1158/1535-7163.MCT-10-0917 Tiwari N, Tatari M, Christofori G, 2012, EMT as the ultimate survival mechanism of cancer cells. Semin Cancer Biol 22:194–207., DOI 10.1016/j.semcancer.2012.02.013 Kumar S, Das A, Sen S, 2014, Extracellular matrix density promotes EMT by weakening cell-cell adhesions. Mol BioSyst 10: 838–850., DOI 10.1039/c3mb70431a Sun T, Sun BC, Zhao XL, Zhao N, Dong XY, Che N, Yao Z, Ma YM, Gu Q, Zong WK, Liu ZY, 2011, Promotion of tumor cell metastasis and vasculogenic mimicry by way of transcription coactivation by Bcl-2 and Twist1: a study of hepatocellular carcinoma. Hepatology 54:1690–1706., DOI 10.1002/hep.24543 Baranwal S, Alahari SK, 2009, Molecular mechanisms controlling E-cadherin expression in breast cancer. Biochem Biophys Res Commun 384:6–11., DOI 10.1016/j.bbrc.2009.04.051 Sun T, Zhao N, Zhao XL, Gu Q, Zhang SW, Che N,Wang XH, Du J, Liu YX, Sun BC, 2010, Expression and functional significance of Twist1 in hepatocellular carcinoma: its role in vasculogenic mimicry. Hepatology 51:545–556., DOI 10.1002/hep.23311 Kemler R, 1993, From cadherins to catenins: cytoplasmic protein interactions and regulation of cell adhesion. Trends Genet 9:317– 321., DOI 10.1016/0168-9525(93)90250-L Bienz M, 2005, β-catenin: a pivot between cell adhesion and Wnt signalling. Curr Biol 15:R64–R67., DOI 10.1016/j.cub.2004.12.058 Yook JI, Li XY, Ota I, Hu C, Kim HS, Kim NH, Cha SY, Ryu JK, Choi YJ, Kim J, Fearon ER, Weiss SJ, 2006, A Wnt-Axin2- GSK3beta cascade regulates snail1 activity in breast cancer cells. Nat Cell Biol 8:1398–1406., DOI 10.1038/ncb1508 Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, LeBleu VS, Mittendorf EA, Weitz J, Rahbari N, Reissfelder C, Pilarsky C, Fraga MF, Piwnica-Worms D, Kalluri R, 2015, Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 523:177–182., DOI 10.1038/nature14581 Yang C, Robbins PD, 2011, The roles of tumor-derived exosomes in cancer pathogenesis. Clin Dev Immunol 2011:842849., DOI 10.1155/2011/842849 Thery C, Boussac M, Veron P, Ricciardi-Castagnoli P, Raposo G, Garin J, Amigorena S, 2001, Proteomic analysis of dendritic cellderived exosomes: a secreted subcellular compartment distinct from apoptotic vesicles. J Immunol 166:7309–7318., DOI 10.4049 /jimmunol.166.12.7309 Wubbolts R, Leckie RS, Veenhuizen PT, Schwarzmann G, Mobius W, Hoernschemeyer J, Slot JW, Geuze HJ, Stoorvogel W, 2003, Proteomic and biochemical analyses of human B cell-derived exosomes. Potential implications for their function and multivesicular body formation. J Biol Chem 278:10963–10972., DOI 10.1074/jbc.M207550200 Van Niel G, Raposo G, Candalh C, Boussac M, Hershberg R, Cerf- Bensussan N, Heyman M, 2001, Intestinal epithelial cells secrete exosome-like vesicles. Gastroenterol 121:337–349., DOI 10.1053 /gast.2001.26263 Bard MP, Hegmans JP, Hemmes A, Luider TM, Willemsen R, Severijnen LA, van Meerbeeck JP, Burgers SA, Hoogsteden HC, Lambrecht BN, 2004, Proteomic analysis of exosomes isolated from human malignant pleural effusions. Am J Respir Cell Mol Biol 31:114–121., DOI 10.1165/rcmb.2003-0238OC Pisitkun T, Shen RF, Knepper MA, 2004, Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci U S A 101:13368–13373., DOI 10.1073/pnas.0403453101 Mathivanan S, Simpson RJ, 2009, ExoCarta: a compendium of exosomal proteins and RNA. Proteomics 9:4997–5000., DOI 10.1002/pmic.200900351 Kalra H, Simpson RJ, Ji H, Aikawa E, Altevogt P, Askenase P, Bond VC, Borras FE, Breakefield X, Budnik V, Buzas E, Camussi G, Clayton A, Cocucci E, Falcon-Perez JM, Gabrielsson S, Gho YS, Gupta D, Harsha HC, Hendrix A, 2012, Vesiclepedia: a compendium for extracellular vesicles with continuous community annotation. PLoS Biol 10:e1001450., DOI 10.1371/journal. pbio.1001450 Bosque A, Dietz L, Gallego-Lleyda A, Sanclemente M, Iturralde M, Naval J, Alava MA, Martinez-Lostao L, Thierse H, Anel A, 2016, Comparative proteomics of exosomes secreted by tumoral Jurkat T cells and normal human T cell blasts unravels a potential tumorigenic role for valosin-containing protein. Oncotarget 7: 29287–29305., DOI 10.18632/oncotarget.8678 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 11 EP 18 DI 10.1007/s12253-016-0185-z PG 8 ER PT J AU Manimaran, A Manoharan, Sh AF Manimaran, Asokan Manoharan, Shanmugam TI Tumor Preventive Efficacy of Emodin in 7,12-Dimethylbenz[a] Anthracene-Induced Oral Carcinogenesis: a Histopathological and Biochemical Approach SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral cancer; Emodin; Lipid peroxidation; Antioxidants; Apoptosis; Detoxification agents ID Oral cancer; Emodin; Lipid peroxidation; Antioxidants; Apoptosis; Detoxification agents AB The aim of the present study is to focus the chemopreventive potential of Emodin during 7,12-dimethylbenz [a]anthracene (DMBA) induced hamster buccal pouch carcinogenesis. Tumors were developed in the buccal pouches of golden Syrian hamsters by painting with 0.5% DMBA thrice a week for 14 weeks. The status of lipid peroxidation, antioxidants and detoxification agents were utilized as biochemical endpoints and the expression pattern of apoptotic proteins was employed as molecular endpoints in addition to the histopathological studies, to substantiate the anticancer potential of Emodin. Hamsters treated with DMBA + Emodin revealed mild to moderate precancerous lesions such as hyperplasia and dysplasia whereas 100% tumor formation was noticed in hamsters treated with DMBA alone. Also, Emodin treatment modulated the status of lipid peroxidation, antioxidants, phase I and II detoxification agents and apoptotic proteins in favor of the inhibition/reversal/suppression of the oral tumorigenesis in DMBA treated hamsters. The present study thus concludes that the chemopreventive potential of Emodin relies on its pro-apoptotic and antioxidant efficacy during DMBA induced hamster buccal pouch carcinogenesis. C1 [Manimaran, Asokan] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608002 Tamil Nadu, India. [Manoharan, Shanmugam] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai Nagar, 608002 Tamil Nadu, India. RP Manoharan, Sh (reprint author), Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, 608002 Tamil Nadu, India. EM sakshiman@rediffmail.com CR Torre LA, Bray F, Siegel RL, Ferlay J, Tieulent JL, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87–108 Manoharan S, Karthikeyan S, Essa MM, Manimaran A, Selvasundaram R, 2016, An overview of oral carcinogenesis. Int J nutr Pharmacol Neurol Dis 6(2):51–62., DOI 10.4103/2231- 0738.179964 Petti S, Masood M, Scully C, 2013, The magnitude of tobacco smoking-betel quid chewing-alcohol drinking interaction effect on oral cancer in South-East Asia. A meta-analysis of observational studies. PLoS One 8(11):e78999., DOI 10.1371/journal. pone.0078999 Ram H, Sarkar J, Kumar H, Konwar R, Bhatt ML, Mohammad S, 2011, Oral cancer: risk factors and molecular pathogenesis. J Maxillofac Oral Surg 10(2):132–137., DOI 10.1007/s12663-011- 0195-z More Y, D'Cruz AK, 2013, Oral cancer: review of current management strategies. Natl Med J India 26(3):152–158 Sallam RM, 2015, Proteomics in cancer biomarkers discovery: challenges and applications. Dis Markers 2015(1):e321370., DOI 10.1155/2015/321370 Manoharan S, Kavitha K, Balakrishnan S, Rajalingam K, 2008, Clerodendron Inerme protects cellular integrity during 7,12- dimethylbenz[a]-anthracene induced hamster buccal pouch carcinogenesis. Afr J Trad Comp Alter Med 5(2):213–222 Rajasekaran D, Manoharan S, Prabhakar MM, Manimaran A, 2015, Enicostemma littorale prevents tumor formation in 7,12- dimethylbenz(a)anthracene-induced hamster buccal pouch carcinogenesis. Hum Exp Toxicol 34(9):911–921., DOI 10.1177 /0960327114562033 Baskaran N, Manoharan S, Karthikeyan S, Prabhakar MM, 2012, Chemopreventive potential of coumarin in 7, 12-dimethylbenz[a] anthracene induced hamster buccal pouch carcinogenesis. Asian Pac J Cancer Prev 13(10):5273–5279 Manoharan S, Palanimuthu D, Baskaran N, Silvan S, 2012, Modulating effect of lupeol on the expression pattern of apoptotic markers in 7, 12-dimethylbenz(a)anthracene induced oral carcinogenesis. Asian Pac J Cancer Prev 13(11):5753–5757 Wang H, Khor TO, Shu L, 2012, Plants against cancer: a review on natural phytochemicals in preventing and treating cancers and their druggability. Anti Cancer Agents Med Chem 12(10):1281–1305 Stanicova J, Verebova V, Strejckova A, 2016, Potential anticancer agent hypericin and its model compound emodin: interaction with DNA. Ceska Slov Farm 65(1):28–31 LinWF,Wang C, Ling CQ, 2015, Research progress in anti-tumor effect of emodin. Zhongguo Zhong Yao Za Zhi 40(20):3937–3940 Chen G, Zhang J, Zhang H, Xiao Y, Kao X, Liu Y, Liu Z, 2015, Anti-inflammatory effect of emodin on lipopolysaccharide-induced keratitis in Wistar rats. Int J Clin Exp Med 8(8):12382–12389 KimMS, Park MJ, KimSJ, Lee CH,Yoo H, Shin SH, Song ES, Lee SH, 2005, Emodin suppresses hyaluronic acid-induced MMP-9 secretion and invasion of glioma cells. Int J Oncol 27(3):839–846 Xue J, Ding W, Liu Y, 2010, Anti-diabetic effects of emodin involved in the activation of PPARgamma on high-fat diet-fed and low dose of streptozotocin-induced diabetic mice. Fitoterapia 81(3): 173–177., DOI 10.1016/j.fitote.2009.08.020 Bhadauria M, 2010, Dose-dependent hepatoprotective effect of emodin against acetaminophen-induced acute damage in rats. Exp Toxicol Pathol 62(6):627–635., DOI 10.1016/j.etp.2009.08.006 Zhao XY, Qiao GF, Li BX, Chai LM, Li Z, Lu YJ, Yang BF, 2009, Hypoglycaemic and hypolipidaemic effects of emodin and its effect on L-type calciumchannels in dyslipidaemic-diabetic rats. Clin Exp Pharmacol Physiol 36(1):29–34. doi :10.1111/j.1440- 1681.2008.05051.x Yagi K, 1987, Lipid peroxides and human diseases. Chem Phys Lipids 45(2–4):337–351 Ohkawa H, Ohishi N, Yagi K, 1979, Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 95(2): 351–358 Desai ID, 1984, Vitamin E analysis methods for animal tissues. Methods Enzymol 105(1):138–147 Palan PR, MikhailMS, BasuMS, Romney SL, 1991, Plasma levels of antioxidant beta-carotene and alpha-tocopherol in uterine cervix dysplasia and cancer. Nutr Cancer 15(1):13–20 Beutler E, Kelly BM, 1963, The effect of sodium nitrite on red cell GSH. Experientia 19(1):96–97 Omaye ST, Turnbull JD, Sauberlich HE, 1979, Selected methods for the determination of ascorbic acid in animal cells, tissues, and fluids. Methods Enzymol 62(1):3–11 Kakkar BD, Viswanathan PN, 1984, A modified spectrophotometric assay of superoxide dismutase. Indian J BiochemBiophys 21(2): 130–132 Sinha AK, 1972, Colorimetric assay of catalase. Anal Biochem 47(2):389–394 Rotruck JT, Pope AL, Ganther HE, 1973, Selenium: biochemical role as a component of glutathione peroxidase. Science 179(4073): 588–590 Omura T, Sato R, 1964, The carbon monoxide binding pigment of liver microsomes. J Biol Chem 239(1):2370–2378 Tietze F, 1969, Enzymic method for quantitative determination of nanogram amounts of total and oxidized glutathione: applications to mammalian blood and other tissues. Anal Biochem 27(3):502– 522 Habig WH, Pabst MJ, JakobyWBC, 1974, Glutathione-S-transferases: the first enzymatic step in mercapturic acid formation. J Biol Chem 249(22):7130–7139 CarlbergBM,Mannervik G, 1985, Glutathione reductase.Methods Enzymol 113(1):484–490 Ernster L, 1967, DT-Diaphorase. In: Pullman ME, ed, Estabrook RW. Methods Enzymol Academic Press, New York, pp 309–317 Bradford MM, 1976, A rapid and sensitive for the quantitation of microgram quantitites of protein utilizing the principle of proteindye binding. Anal Biochem 72(1):248–254 Manoharan S, Balakrishnan S, Menon VP, Alias LM, Reena AR, 2009, Chemopreventive efficacy of curcumin and piperine during 7,12-dimethylbenz[a]anthracene induced hamster buccal pouch carcinogenesis. Singap Med J 50(2):139–146 Karthikeyan S, Manoharan S, 2016, Cromolyn inhibits 7, 12- dimethylbenz(a, anthracene induced oral cancer through apoptotic induction and suppression of cell proliferation. Int J Pharm Bio Sci 7(1):35–42 Manoharan S, Rajasekaran D, Prabhakar MM, Karthikeyan S, Manimaran A, 2015, Modulating effect of Enicostemma littorale on the expression pattern of apoptotic, cell proliferative, inflammatory and angiogenic markers during 7, 12-dimethylbenz, a, anthracene induced hamster buccal pouch carcinogenesis. Toxicol Int 22(1):130–140., DOI 10.4103/0971-6580.172276 Palanimuthu D, Baskaran N, Silvan S, Rajasekaran D, Manoharan S, 2012, Lupeol, a bioactive triterpene, prevents tumor formation during 7,12-dimethylbenz(a)anthracene induced oral carcinogenesis. Pathol Oncol Res 18(4):1029–1037 Karthikeyan S, Srinivasan R, Wani SA, Manoharan S, 2013, Chemopreventive potential of chrysin in 7,12- dimethylbenz(a)anthracene-induced hamster buccal pouch carcinogenesis. Int J Nutr Pharmacol Neurol Dis 3(1):46–53 Valavanidis A, Vlachogianni T, Fiotakis K, Loridas S, 2013, Pulmonary oxidative stress, inflammation and cancer: Respirable particulate matter, fibrous dusts and ozone as major causes of lung carcinogenesis through reactive oxygen species mechanisms. Int J Environ Res Pub Health 10(9):3886–3907., DOI 10.3390 /ijerph10093886 Manoharan S, Wani SA, Vasudevan K, Manimaran A, Prabhakar MM, Karthikeyan S, Rajasekaran D, 2013, Saffron reduction of 7, 12-dimethylbenz[a]anthracene-induced hamster buccal pouch carcinogenesis. Asian Pac J Cancer Prev 14(2):951–957 Danaraddi S, Koneru A, Hunasgi S, Ramalu S, VanishreeM(2014, Natural ways to prevent and treat oral cancer. J Oral Res Rev 6(1): 34–39 Lobo V, Patil A, Phatak A, Chandra N, 2010, Free radicals, antioxidants and functional foods: impact on human health. Pharm Rev 4(8):118–126., DOI 10.4103/0973-7847.70902 Horn W, Maier H, Born AJ, 1988, Reduction of cytochrome c by oral mucosa of patients with oropharyngeal cancer. Klin Wochenschr 66(11):105–107 Tzifi F, Economopoulou C, Gourgiotis D, Ardavanis A, Papageorgiou S, Scorilas A, 2012, The role of Bcl-2 family of apoptosis regulator proteins in acute and chronic leukemias. Adv Hematol 2012(1):e524308., DOI 10.1155/2012/524308 Rivlin N, Brosh R, Oren M, Rotter V, 2011, Mutations in the p53 tumor suppressor gene: important milestones at the various steps of tumorigenesis. Genes Cancer 2(4):466–474., DOI 10.1177 /1947601911408889 Manoharan S, SindhuG, NirmalMR, Vetrichelvi V, Balakrishnan S, 2011, Protective effect of berberine on expression pattern of apoptotic, cell proliferative, inflammatory and angiogenic markers during 7,12-dimethylbenz(a)anthracene induced hamster buccal pouch carcinogenesis. Pak J Biol Sci 14(20):918–932 Prabhakar MM, Vasudevan K, Karthikeyan S, Baskaran N, Manoharan S, 2012, Anti-cell proliferative efficacy of ferulic acid against 7, 12-dimethylbenz(a)anthracene induced hamster buccal pouch carcinogenesis. Asian Pac J Cancer Prev 13(10):5207–5211 Bose P, Klimowicz AC, Kornaga E, Petrillo SK, Matthews TW, 2012, Bax expression measured by AQUAnalysis is an independent prognostic marker in oral squamous cell carcinoma. BMC Cancer 12(1):e332., DOI 10.1186/1471-2407-12-332 Kaur H, Gupta S, 2013, An analysis of the expression of Bcl-2, podoplanin and lymph angiogenesis in benign and malignant salivary gland tumours. J Clin Exp Pathol 3(1):e145., DOI 10.4172 /2161-0681.1000145 Arya V, Singh S, Daniel MJ, 2016, Clinicopathological correlation of Bcl-2 oncoprotein expression in oral precancer and cancer. Oral Biol Craniofac Res 6(1):18–23., DOI 10.1016/j.jobcr.2015.12.011 Rahmani A, Alzohairy M, Babiker AY, Rizvi MA, Elkarimahmad HG, 2012, Clinicopathological significance of PTEN and bcl2 expressions in oral squamous cell carcinoma. Int J Clin Exp Pathol 5(9):965–971 Watanabe J, Kushihata F, Honda K, Sugita A, Tateishi N, Mominoki K, Matsuda S, Kobayashi N, 2004, Prognostic significance of Bcl-xL in human hepatocellular carcinoma. Surgery 135(6):604–612 Ranger AM, Zha J, Harada H, Datta SR, Danial NN, Gilmore AP, Kutok JL, Le Beau MM, Greenberg ME, Korsmeyer SJ, 2003, Bad-deficient mice develop diffuse large B cell lymphoma. Proc Natl Acad Sci 100(16):9324–9329 Yancey D, Nelson KC, Baiz D, Hassan S, Flores A, 2013, BAD dephosphorylation and decreased expression ofMCL-1 induce rapid apoptosis in prostate cancer cells. PLoS One 8(9):e74561., DOI 10.1371/journal.pone.0074561 Jiang L, Luo M, Liu D, Chen B, Zhang W, 2013, BAD overexpression inhibits cell growth and induces apoptosis via mitochondrial-dependent pathway in non-small cell lung cancer. Cancer Cell Int 13(1):53., DOI 10.1186/1475-2867-13-53 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 19 EP 29 DI 10.1007/s12253-017-0205-7 PG 11 ER PT J AU Wang, N Chen, K Xu, J Yuan, F Li, H Deng, F Zhang, L AF Wang, NaNa Chen, KeYu Xu, Jia Yuan, Fang Li, HongYu Deng, FengMei Zhang, LuShun TI Association of CAA and TATC Insertion/Deletion Genetic Polymorphisms in RTN4 3′-UTR with Hepatocellular Carcinoma Risk SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; RTN4; Nogo; Polymorphisms ID Hepatocellular carcinoma; RTN4; Nogo; Polymorphisms AB Evidence from recent researchers suggested that RTN4 is a multifunctional gene, including tumor suppression, apoptosis, vascular remodeling, and inhibition of axonal regeneration. The CAA and TATC insertion/deletion polymorphisms (CAA/TATC polymorphisms) of RTN4 3″-untranslated regions (UTRs) have been linked to cervical squamous cell carcinoma (CSCC), uterine leiomyomas (UL) and non-small cell lung cancer (NSCLC). However, the association between these two polymorphisms sites with Hepatocellular Carcinoma (HCC) risk was not carry out before. A total of 284 HCC patients and 484 control subjects were recruited for this study. The RTN4 CAA/TATC insertion/deletion genotypes were determined using polymerase chain reaction (PCR) assay. The ID/ DD genotypes of CAA were significantly associated with an increased risk of HCC compared with the II genotype (ID vs. II: OR = 1.50, 95% CI: 1.10–2.04; DD vs. II: OR = 2.00, 95%CI: 1.15–3.46). Meanwhile, the frequency of D allele of CAA was significantly related with an increased risk of HCC compared with the I allele (D vs. I: OR = 1.39, 95% CI: 1.12–1.73). The ID genotypes of TATC was significantly associated with an increased risk of HCC compared with the DD genotype (ID vs. DD: OR = 1.70, 95% CI: 1.23–2.33). The present study provided evidence that RTN4 CAA/TATC polymorphisms were associated with HCC development in Chinese Han population. C1 [Wang, NaNa] Chengdu Medical College, School of Medical Laboratory Science, 610500 Chengdu, China. [Chen, KeYu] Chengdu Medical College, School of Biomedical Sciences, 610500 Chengdu, China. [Xu, Jia] Chengdu Medical College, School of Medical Laboratory Science, 610500 Chengdu, China. [Yuan, Fang] Sichuan University, West China School of Preclinical and Forensic Medicine, Department of Immunology, 610041 Chengdu, China. [Li, HongYu] Sichuan University, West China Hospital, Department of Pancreatic Surgery, 610041 Chengdu, China. [Deng, FengMei] Chengdu Medical College, Department of Neurobiology, Department of Pathology and Pathophysiology, 610500 Chengdu, China. [Zhang, LuShun] Chengdu Medical College, Department of Neurobiology, Department of Pathology and Pathophysiology, 610500 Chengdu, China. RP Zhang, L (reprint author), Chengdu Medical College, Department of Neurobiology, Department of Pathology and Pathophysiology, 610500 Chengdu, China. EM zhangls2012@163.com CR Hoyos S, Escobar J, Cardona D, Guzman C, Mena A, Osorio G, Perez C, Restrepo JC, Correa G, 2015, Factors associated with recurrence and survival in liver transplant patients with HCC - a single center retrospective study. Ann Hepatol Off J Mex Assoc 14(1):58–63 Hong Y, Long J, Li H, Chen S, Liu Q, Zhang B, He X,Wang Y, Li H, Li Y, 2015, An analysis of Immunoreactive signatures in early stage hepatocellular carcinoma. Ebiomedicine 39(6):438–446 Mu LN, Cao W, Zhang ZF, Cai L, Jiang QW, You NC, Goldstein BY, Wei GR, Chen CW, Lu QY, 2007, Methylenetetrahydrofolate reductase, MTHFR, C677T and A1298C polymorphisms and the risk of primary hepatocellular carcinoma, HCC, in a Chinese population. Cancer Causes Control 18(6):665–675 Wang ZC, Liu LZ, Liu XY, Hu JJ,Wu YN, Shi JY, Yang LX, Duan M,Wang XY, Zhou J, Fan J, Gao Q, 2015, Genetic polymorphisms of the multidrug resistance 1 gene MDR1 and the risk of hepatocellular carcinoma. Tumour Biol: J Int Soc Oncodevelopmental Biol Med 36(9):7007–7015., DOI 10.1007/s13277-015-3407-1 Toyoda H, Kumada T, Tada T, Kiriyama S, Tanikawa M, Hisanaga Y, Kanamori A, Kitabatake S, Ito T, 2015, Risk factors of hepatocellular carcinoma development in non-cirrhotic patients with sustained virologic response for chronic hepatitis C virus infection. J Gastroenterol Hepatol 30, 7):1183–1189., DOI 10.1111/jgh.12915 Thompson KJ, Humphries JR, Niemeyer DJ, Sindram D, McKillop IH, 2015, The effect of alcohol on Sirt1 expression and function in animal and human models of hepatocellular carcinoma, HCC). Adv Exp Med Biol 815:361–373., DOI 10.1007/978-3-319-09614-8_21 Kazuaki N, Michiie S, Susumu Y, Satoru T, Setsuo H, 2005, Akt phosphorylation is a risk factor for early disease recurrence and poor prognosis in hepatocellular carcinoma. Cancer 103(2):307–312 Herceg Z, Paliwal A, 2011, Epigenetic mechanisms in hepatocellular carcinoma: how environmental factors influence the epigenome. Mutat Res/Fundam Mol Mech Mutagen 727(3):55–61 Yang J, Yu L, Bi AD, Zhao SY, 2000, Assignment of the human reticulon 4 gene, RTN4, to chromosome 2p14– > 2p13 by radiation hybrid mapping. Cytogenet Cell Genet 88(1–2):101–102 Shi S, Zhou B,Wang Y, Chen Y, Zhang K,Wang K, Quan Y, Song Y, Rao L, Zhang L, 2012, Genetic variation in RTN4 3'-UTR and susceptibility to cervical squamous cell carcinoma. DNA Cell Biol 31(6):1088–1094 Zhang K, Bai P, Shi S, Zhou B, Wang Y, Song Y, Rao L, Zhang L, 2013, Association of genetic variations in RTN4 3'-UTR with risk of uterine leiomyomas. Pathol Oncol Res Por 19(3):475–479 De-Yi L, Xu-Hua M, Ying-Hui Z, Xiao-Long Y, Wei-Ping W, Ya- Biao Z, Juan-Juan X, Ping Z, Jian-GuoW, Neetika A, 2014, RTN4 3'-UTR insertion/deletion polymorphism and susceptibility to nonsmall cell lung cancer in Chinese Han population. Asian Pac J Cancer Prev Apjcp 15(13):5249–5252 Novak G, Kim D, Seeman P, Tallerico T, 2002, Schizophrenia and nogo: elevated mRNA in cortex, and high prevalence of a homozygous CAA insert. Brain Res Mol Brain Res 107(2):183–189 Zhou B, Rao L, Li Y, Gao L, Li C, Chen Y, Xue H, LiangW, Lv M, Song Y, 2009, The association between dilated cardiomyopathy and RTN4 3'UTR insertion/deletion polymorphisms. Clin Chimica Acta; Int J Clin Chem 400(1–2):21–24 Chen Y, Zhou B, Li H, Peng Y, Wang Y, Rao L, 2010, Analysis of RTN4 3'UTR insertion/deletion polymorphisms in ventricular septal defect in a Chinese Han population. DNA Cell Biol 30(5):323– 327 Oertle T, Huber CPH, Schwab ME, 2003, Genomic structure and functional characterisation of the promoters of human and mouse nogo/rtn4. J Mol Biol 325(2):299–323 Yates D, 2012, Neural development: nogo to synapse formation. Nat Rev Neurosci 13(4):221–221 Liu X, Wang Y, Yong Z, Wei Z, Xu X, Niinobe M, Yoshikawa K, Lu C, Cheng H, 2009, Nogo-a inhibits necdin-accelerated neurite outgrowth by retaining necdin in the cytoplasm. Mol Cell Neurosci 41(1):51–61 Chen YC, Dong-Dong LU, Cao XR, Zhang XR, 2005, RTN4-C Gene expression in He patocellular carcinoma and its influence on SMMC7721 cell growth and apoptosis. Acta Genet Sin 32(9):891– 897 Tashiro K, Satoh A, Utsumi T, Chung C, Iwakiri Y, 2013, Absence of nogo-B, reticulon 4B, facilitates hepatic stellate cell apoptosis and diminishes hepatic fibrosis in mice - the American journal of pathology. Am J Pathol 182(3):786–795 Xiao JunWU, Cheng LY, Rui CH, Xiang CJ, Huang CG, RongYZ, Wei PJ, Luo C, Han HG, Lou MQ, 2008, Nogo-B expression in human brain gliomas and its relationship with tumor pathological grades. Chin J Cancer Biother 15(6):566–569 Li Q, Qi B, Oka K, Shimakage M, Yoshioka N, Inoue H, Hakura A, Kodama K, Stanbridge EJ, YutsudoM(2001, Link of a new type of apoptosis-inducing gene ASY/nogo-B to human cancer. Oncogene 20(30):3929–3936., DOI 10.1038/sj.onc.1204536 Sarkey JP, Chu M,Mcshane M, Bovo E,MouYA, ZimaAV, Tombe PPD, Kartje GL, Martin JL, 2011, Nogo-a knockdown inhibits hypoxia/Reoxygenation-induced activation of mitochondrialdependent apoptosis in cardiomyocytes. Biophys J 100(3):1044– 1055 Watari A, Yutsudo M, 2003, Multi-functional gene ASY/nogo/ RTN-X/RTN4: apoptosis, tumor suppression, and inhibition of neuronal regeneration. Apoptosis 8(1):5–9 Mendell JT, Dietz HC, 2001, When the message goes awry: disease-producingmutations that influencemRNA content and performance. Cell 107(4):411–414 Kuersten S, Goodwin EB, 2003, The power of the 3' UTR: translational control and development. Nat Rev Genet 4(8):626–637., DOI 10.1038/nrg1125 Li H, Chen Y, Zhou B, Peng Y, BaiW, Rao L, 2012, RNT4 3'-UTR insertion/deletion polymorphisms are not associated with atrial septal defect in Chinese Han population: a brief communication. DNA Cell Biol 31(6):1121–1124., DOI 10.1089/dna.2011.1386 Conne B, Stutz A, Vassalli JD, 2000, The 3' untranslated region of messenger RNA: a molecular 'hotspot' for pathology? Nat Med 6(6):637–641., DOI 10.1038/76211 Novak G, Tallerico T, 2006, Nogo a, B and C expression in schizophrenia, depression and bipolar frontal cortex, and correlation of nogo expression with CAA/TATC polymorphism in 3'-UTR. Brain Res 1120(1):161–171 Shaoqing S, Bin Z, Kui Z, Lin Z, 2013, Association between two genetic variants of CD226 gene and cervical squamous cell carcinoma: a case–control study. Gene 519(1):159–163 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 31 EP 34 DI 10.1007/s12253-017-0204-8 PG 4 ER PT J AU Virga, J Bognar, L Hortobagyi, T Zahuczky, G Csosz, Kallo, G Toth, J Hutoczki, G Remenyi-Puskar, J Steiner, L Klekner, AF Virga, Jozsef Bognar, Laszlo Hortobagyi, Tibor Zahuczky, Gabor Csosz, Eva Kallo, Gergo Toth, Judit Hutoczki, Gabor Remenyi-Puskar, Judit Steiner, Laszlo Klekner, Almos TI Tumor Grade versus Expression of Invasion-Related Molecules in Astrocytoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gliomas; Tumor grade; Extracellular matrix; Invasion; Expression ID Gliomas; Tumor grade; Extracellular matrix; Invasion; Expression AB Peritumoral infiltration is characteristic of astrocytomas even in low-grade tumors. Tumor cells migrate to neighbouring tissue and cause recurrence. The extracellular matrix (ECM) plays a role in tumor invasion; expression levels of its components’ have been linked to tumor invasion. This study determines the mRNA and protein expression of 20 invasion-related ECM components by examining non-tumor brain; grade I-II-III astrocytoma and glioblastoma samples. Expression levels were measured by QRT-PCR and massspectroscopy. The connection between the expression pattern and tumor grade is statistically analyzed. During the analysis of data, key molecules (brevican, cadherin-12, fibronectin and integrin-β1) correlating the most with tumor grade were selected. While the mRNA level of brevican, ErbB2, fibronectin, integrin-β1 and versican discriminates low-grade from high-grade gliomas, of proteins RHAMM, integrin-α1 and MMP2 seems important. The expression pattern was found to be distinctive for tumor grade, as statistical classifiers are capable of identifying an unknown sample’s grade using them. Furthermore, normal brain and glioma expression patterns, along with low-grade astrocytoma and glioblastoma samples, differ the most. Determining the invasion-related molecules’ expression profile provides extra information regarding the tumor’s clinical behavior. Additionally, identifying molecules playing a key role in glioma invasion could uncover potential therapeutic targets in the future. C1 [Virga, Jozsef] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Bognar, Laszlo] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Hortobagyi, Tibor] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Zahuczky, Gabor] UD-GenoMed Medical Genomic Technologies Ltd, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Csosz, Eva] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular Biology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Kallo, Gergo] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular Biology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Toth, Judit] University of Debrecen, Department of Oncology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Hutoczki, Gabor] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Remenyi-Puskar, Judit] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Steiner, Laszlo] UD-GenoMed Medical Genomic Technologies Ltd, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Klekner, Almos] University of Debrecen, Clinical Center, Department of Neurosurgery, Nagyerdei krt. 98, 4032 Debrecen, Hungary. RP Klekner, (reprint author), University of Debrecen, Clinical Center, Department of Neurosurgery, 4032 Debrecen, Hungary. EM neurosurgery.debrecen@freemail.hu CR Goodenberger ML, Jenkins RB, 2012, Genetics of adult glioma. Cancer Genet 205:613–621., DOI 10.1016/j.cancergen.2012.10.009 Ostrom QT, Gittleman H, Liao P et al, 2014, CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2007–2011. Neuro-Oncology 16:iv1–iv63., DOI 10.1093/neuonc/nou223 Louis DN, Ohgaki H, Wiestler OD et al, 2007, The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114:97–109., DOI 10.1007/s00401-007-0243-4 Tysnes BB, Mahesparan R, 2001, Biological mechanisms of glioma invasion and potential therapeutic targets. J Neuro-Oncol 53: 129–147 Agnihotri S, Burrell KE, Wolf A et al, 2013, Glioblastoma, a brief review of history, molecular genetics, animal models and novel therapeutic strategies. Arch Immunol Ther Exp 61:25–41., DOI 10.1007/s00005-012-0203-0 Chintala SK, Rao JK, 1996, Invasion of human glioma: role of extracellular matrix proteins. Front Biosci 1:d324–d339 Gingras M, Roussel E, Bruner JM et al, 1995, Comparison of cell adhesion molecule expression between glioblastoma multiforme and autologous normal brain tissue. J Neuroimmunol 57:143– 153., DOI 10.1016/0165-5728(94)00178-Q Mahesparan R, Read T-A, Lund-Johansen M et al, 2003, Expression of extracellular matrix components in a highly infiltrative in vivo glioma model. Acta Neuropathol 105:49–57 Klekner A, Virga J, Toth J et al, 2013, The role of extracellular matrix components in the invasion of intracranial malignancies. Magy Onkol 57:222–231 Coniglio SJ, Segall JE, 2013, Review: molecular mechanism of microglia stimulated glioblastoma invasion. Matrix Biol 32:372– 380., DOI 10.1016/j.matbio.2013.07.008 Knott JCA, Mahesparan R, Garcia-Cabrera I et al, 1998, Stimulation of extracellular matrix components in the normal brain by invading glioma cells. Int J Cancer 75:864–872 Varga I, HutoczkiG, Szemcsak CDet al, 2012, Brevican, neurocan, tenascin-C and versican are mainly responsible for the invasiveness of low-grade astrocytoma. Pathol Oncol Res 18:413–420 Varga I, Hutoczki G, Petras M et al, 2010, Expression of invasionrelated extracellular matrix molecules in human glioblastoma versus intracerebral lung adenocarcinoma metastasis. Zentralbl Neurochir 71:173–180 Klekner A, Varga I, Bognar L et al, 2010, Extracellular matrix of cerebral tumors with different invasiveness. Ideggyogy Sz 63:38–43 Bellail AC, Hunter SB, Brat DJ et al, 2004, Microregional extracellular matrix heterogeneity in brain modulates glioma cell invasion. Int J Biochem Cell Biol 36:1046–1069., DOI 10.1016/j. biocel.2004.01.013 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), method. Methods 25:402–408 Bradford M, 1976, Rapid and sensitive method for quantification of microgram quantities of protein utilizing principle of proteindye- binding. Anal Biochem 72:248–254 James A, Jorgensen C, 2010, Basic design of MRM assays for peptide quantification. Methods Mol Biol 658:167–185 Iuga C, Seicean A, Iancu C et al, 2014, Proteomic identification of potential prognostic biomarkers in resectable pancreatic ductal adenocarcinoma. Proteomics 14:945–955 Qian W-J, Jacobs JM, Liu T et al, 2006, Advances and challenges in liquid chromatography-mass spectrometrybased proteomics profiling for clinical applications. Mol Cell Proteomics 5:1727–1744 Lange V, Picotti P, Domon B, Aebersold R, 2008, Selected reaction monitoring for quantitative proteomics: a tutorial. Mol Syst Biol 4:222 D’Abaco GM, Kaye AH, 2007, Integrins: molecular determinants of glioma invasion. J Clin Neurosci 14:1041–1048., DOI 10.1016/j. jocn.2007.06.019 Fukushima Y, Ohnishi T, Arita N et al, 1998, Integrin alpha3beta1-mediated interaction with laminin-5 stimulates adhesion, migration and invasion of malignant glioma cells. Int J Cancer 76:63–72., DOI 10.1002/(SICI)1097-0215, 19980330)76:1<63::AID-IJC11>3.0.CO;2-H Yeh W-L, Lu D-Y, Liou H-C, Fu W-M, 2012, A forward loop between glioma and microglia: glioma-derived extracellular matrix-activated microglia secrete IL-18 to enhance the migration of glioma cells. J Cell Physiol 227:558–568., DOI 10.1002/jcp.22746 Farber K, Synowitz M, ZahnG et al, 2008, An alpha5beta1 integrin inhibitor attenuates glioma growth.Mol Cell Neurosci 39:579–585., DOI 10.1016/j.mcn.2008.08.005 Ljubimova JY, Lakhter AJ, Loksh A et al, 2001, Overexpression of α4 chain-containing laminins in human glial tumors identified by gene microarray analysis. Cancer Res 61:5601–5610 Nagato S, Nakagawa K, Harada H et al, 2005, Downregulation of laminin alpha4 chain expression inhibits glioma invasion in vitro and in vivo. Int J Cancer 117:41–50., DOI 10.1002/ijc.21102 Tanaka K, Hiraiwa N, Hashimoto H et al, 2004, Tenascin-C regulates angiogenesis in tumor through the regulation of vascular endothelial growth factor expression. Int J Cancer 108:31–40., DOI 10.1002/ijc.11509 Herold-Mende C, MuellerMM, BonsantoMMet al, 2002, Clinical impact and functional aspects of tenascin-C expression during glioma progression. Int J Cancer 98:362–369., DOI 10.1002/ijc.10233 Rousseau C, Ruellan AL, Bernardeau K et al, 2011, Syndecan-1 antigen, a promising new target for triple-negative breast cancer immuno-PET and radioimmunotherapy. A preclinical study on MDA-MB-468 xenograft tumors. EJNMMI Res 1:20., DOI 10.1186/2191-219X-1-20 Xu Y, Yuan J, Zhang Z et al, 2012, Syndecan-1 expression in human glioma is correlated with advanced tumor progression and poor prognosis. Mol Biol Rep 39:8979–8985 Gary SC, Kelly GM, Hockfield S, 1998, BEHAB/brevican: a brain-specific lectican implicated in gliomas and glial cell motility. Curr Opin Neurobiol 8:576–581., DOI 10.1016/S0959-4388, 98)80083-4 Wegrowski Y, Maquart F-X, 2006, Chondroitin sulfate proteoglycans in tumor progression. Adv Pharmacol 53:297–321., DOI 10.1016/S1054-3589(05)53014-X Silver DJ, Silver J, 2014, Contributions of chondroitin sulfate proteoglycans to neurodevelopment, injury, and cancer. Curr Opin Neurobiol 27:171–178., DOI 10.1016/j.conb.2014.03.016 Egeblad M, Werb Z, 2002, New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2:161–174 Lampert K, Machein U, Machein MR et al, 1998, Expression of matrix metalloproteinases and their tissue inhibitors in human brain tumors. Am J Pathol 153:429–437., DOI 10.1016/S0002-9440(10)65586-1 Takeichi M, 1993, Cadherins in cancer: implications for invasion and metastasis. Curr Opin Cell Biol 5:806–811., DOI 10.1016/0955- 0674(93)90029-P Asano K, Kubo O, Tajika Y et al, 1997, Expression and role of cadherins in astrocytic tumors. Brain Tumor Pathol 14:27–33 Zhao J, Li P, Feng H et al, 2013, Cadherin-12 contributes to tumorigenicity in colorectal cancer by promoting migration, invasion, adhersion and angiogenesis. J Transl Med 11:288 Xu G-F, XieW-F, 2013, Effect of ERBB2 expression on invasiveness of glioma TJ905 cells. Asian Pac J Trop Med 6:964–967., DOI 10.1016/S1995-7645(13)60172-8 Gao L, Li F, Dong B et al, 2010, Inhibition of STAT3 and ErbB2 suppresses tumor growth, enhances radiosensitivity, and induces mitochondria-dependent apoptosis in glioma cells. Int J Radiat Oncol Biol Phys 77:1223–1231., DOI 10.1016/j.ijrobp.2009.12.036 Turley EA, Hossain MZ, Sorokan T et al, 1994, Astrocyte and microglial motility in vitro is functionally dependent on the hyaluronan receptor RHAMM. Glia 12:68–80., DOI 10.1002 /glia.440120109 Akiyama Y, Jung S, Salhia B et al, 2001, Hyaluronate receptors mediating glioma cell migration and proliferation. J Neuro-Oncol 53:115–127 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 35 EP 43 DI 10.1007/s12253-017-0194-6 PG 9 ER PT J AU Jin, M Long, ZW Yang, J Lin, X AF Jin, Mei Long, Zi-Wen Yang, Jing Lin, Xiang TI Correlations of IGF-1R and COX-2 Expressions with Ras and BRAF Genetic Mutations, Clinicopathological Features and Prognosis of Colorectal Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Insulin-like growth factor receptor 1; Cyclooxygenase 2; Ras mutation; BRAF mutation; Clinicopathological feature; Prognosis ID Insulin-like growth factor receptor 1; Cyclooxygenase 2; Ras mutation; BRAF mutation; Clinicopathological feature; Prognosis AB This case-control study aims to investigate the correlations of insulin-like growth factor receptor 1 (IGF-1R) and cyclooxygenase 2 (COX-2) expressions with Ras and BRAF genetic mutations, clinicopathological features and prognosis of colorectal cancer (CRC) patients. A total of 213 CRC patients (case group) and 200 healthy individuals (control group) were selected from our hospital. Ras (K-Ras/N-Ras) and BRAF genetic mutations were detected by direct sequencing. The positive expression rates of IGF-IR and COX-2 in CRC and normal tissues were detected using immunohistochemistry. RTqPCR and Western blotting were applied to detect the mRNA and protein expressions of IGF-IR and COX-2 in CRC tissues and normal tissues. Total mutation rate of N-Ras, BRAF and KRas in case group were 5.2%, 12.2% and 47.4%, respectively. The expressions of IGF-IR and COX-2 were higher in CRC tissues with Ras and BRAF mutations than in those without. CRC tissues with Ras mutation showed higher COX-2 expression than those with BRAF mutation. IGF-IR and COX-2 expressions were correlated to infiltration degree, lymphatic metastasis (in CRC tissues with and without Ras and BRAF mutations), and Dukes stages (only in CRC tissues with Ras and BRAF mutations). CRC patients with negative expressions of IGF-IR and COX-2 had significantly higher accumulative survival rate and longer mean survival duration than those with positive expressions of IGF-IR and COX-2. These findings indicate that IGF-1R and COX-2 expressions may be associated with Ras and BRAF genetic mutations, clinicopathological feature and prognosis of CRC patients. C1 [Jin, Mei] Shigatse People’s Hospital, Department of Surgery, 85700 Shigatse, Tibet, China. [Long, Zi-Wen] Fudan University, Shanghai Cancer Center, Department of Gastric Cancer Surgery, 200032 Shanghai, China. [Yang, Jing] Shigatse People’s Hospital, Department of Surgery, 85700 Shigatse, Tibet, China. [Lin, Xiang] Shigatse People’s Hospital, Department of Surgery, 85700 Shigatse, Tibet, China. RP Long, ZW (reprint author), Fudan University, Shanghai Cancer Center, Department of Gastric Cancer Surgery, 200032 Shanghai, China. EM rudolfstar@163.com CR Ahmed FE, Ahmed NC, Vos PW, Bonnerup C, Atkins JN, Casey M, Nuovo GJ, Naziri W, Wiley JE, Mota H, Allison RR, 2013, Diagnostic microRNA markers to screen for sporadic human colon cancer in stool: I. Proof of principle. Cancer Genomics Proteomics 10(3):93–113 Hari DM, Leung AM, Lee JH, Sim MS, Vuong B, Chiu CG, Bilchik AJ, 2013, AJCC cancer staging manual 7th edition criteria for colon cancer: do the complex modifications improve prognostic assessment? J Am Coll Surg 217(2):181–190., DOI 10.1016/j. jamcollsurg.2013.04.018 Labianca R, Nordlinger B, Beretta GD, Brouquet A, Cervantes A, Group EGW, 2010, Primary colon cancer: ESMO clinical practice guidelines for diagnosis, adjuvant treatment and follow-up. Ann Oncol 21(Suppl 5):v70–v77., DOI 10.1093/annonc/mdq168 Jiang R, Wang H, Deng L, Hou J, Shi R, Yao M, Gao Y, Yao A, Wang X, Yu L, Sun B, 2013, IL-22 is related to development of human colon cancer by activation of STAT3. BMC Cancer 13:59., DOI 10.1186/1471-2407-13-59 Johnson CM, Wei C, Ensor JE, Smolenski DJ, Amos CI, Levin B, Berry DA, 2013, Meta-analyses of colorectal cancer risk factors. Cancer Causes Control 24(6):1207–1222., DOI 10.1007/s10552-013- 0201-5 Vanamala J, Reddivari L, Radhakrishnan S, Tarver C, 2010, Resveratrol suppresses IGF-1 induced human colon cancer cell proliferation and elevates apoptosis via suppression of IGF-1R/ Wnt and activation of p 53 signaling pathways. BMC Cancer 10: 238., DOI 10.1186/1471-2407-10-238 Ding J, ZhangZM, Xia Y, Liao GQ, Pan Y, Liu S, ZhangY, Yan ZS, 2013, LSD1-mediated epigenetic modification contributes to proliferation and metastasis of colon cancer. Br J Cancer 109(4):994– 1003., DOI 10.1038/bjc.2013.364 Yamamoto T, Oshima T, Yoshihara K, Nishi T, Arai H, Inui K, Kaneko T, Nozawa A, Adachi H, Rino Y, Masuda M, Imada T, 2012, Clinical significance of immunohistochemical expression of insulin-like growth factor-1 receptor and matrix metalloproteinase-7 in resected non-small cell lung cancer. Exp Ther Med 3(5):797–802., DOI 10.3892/etm.2012.493 Shen K, Liang Q, Xu K, Cui D, Jiang L, Yin P, Lu Y, Li Q, Liu J, 2012, MiR-139 inhibits invasion and metastasis of colorectal cancer by targeting the type I insulin-like growth factor receptor. Biochem Pharmacol 84(3):320–330., DOI 10.1016/j.bcp.2012.04.017 Ma J, Sawai H, Matsuo Y, Ochi N, Yasuda A, Takahashi H, Wakasugi T, Funahashi H, Sato M, Takeyama H, 2010, IGF-1 mediates PTEN suppression and enhances cell invasion and proliferation via activation of the IGF-1/PI3K/Akt signaling pathway in pancreatic cancer cells. J Surg Res 160(1):90–101., DOI 10.1016/j. jss.2008.08.016 Wu J, Yu E, 2014, Insulin-like growth factor receptor-1, IGF-IR, as a target for prostate cancer therapy. CancerMetastasis Rev 33(2–3): 607–617., DOI 10.1007/s10555-013-9482-0 King H, Aleksic T, Haluska P, MacaulayVM(2014, Can we unlock the potential of IGF-1R inhibition in cancer therapy? Cancer Treat Rev 40(9):1096–1105., DOI 10.1016/j.ctrv.2014.07.004 Cheng J, Fan XM, 2013, Role of cyclooxygenase-2 in gastric cancer development and progression. World J Gastroenterol 19(42): 7361–7368., DOI 10.3748/wjg.v19.i42.7361 Akutsu Y, Hanari N, Yusup G, Komatsu-Akimoto A, Ikeda N,Mori M, Yoneyama Y, Endo S, Miyazawa Y,Matsubara H, 2011, COX2 expression predicts resistance to chemoradiotherapy in esophageal squamous cell carcinoma. Ann Surg Oncol 18(10):2946–2951., DOI 10.1245/s10434-011-1645-z Roelofs HM, Te Morsche RH, van Heumen BW, Nagengast FM, Peters WH, 2014, Over-expression of COX-2 mRNA in colorectal cancer. BMC Gastroenterol 14:1., DOI 10.1186/1471-230X-14-1 Lehingue Y, Urtizberea JA, 1986, Tuberculosis control in Malawi. Time distribution of cessation of treatment and proposals for reorientation of the program. Bull Soc Pathol Exot Filiales 79(2):259–265 Rahman M, Selvarajan K, Hasan MR, Chan AP, Jin C, Kim J, Chan SK, Le ND, Kim YB, Tai IT, 2012, Inhibition of COX-2 in colon cancer modulates tumor growth and MDR-1 expression to enhance tumor regression in therapy-refractory cancers in vivo. Neoplasia 14(7):624–633 Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, Juan T, Sikorski R, Suggs S, Radinsky R, Patterson SD, Chang DD, 2008, Wild-type kras is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 26(10): 1626–1634., DOI 10.1200/JCO.2007.14.7116 Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, HumbletY, BodokyG, CunninghamD, Jassem J, Rivera F,Kocakova I, Ruff P, Blasinska-Morawiec M, Smakal M, Canon JL, Rother M, Oliner KS, Wolf M, Gansert J, 2010, Randomized, phase iii trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin, folfox4, versus folfox4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the prime study. J Clin Oncol 28(31):4697–4705., DOI 10.1200/JCO.2009.27.4860 Peeters M, Price TJ, Cervantes A, Sobrero AF, Ducreux M, Hotko Y, Andre T, Chan E, Lordick F, Punt CJ, Strickland AH,Wilson G, Ciuleanu TE, Roman L, Van Cutsem E, Tzekova V, Collins S, Oliner KS, Rong A, Gansert J, 2010, Randomized phase iii study of panitumumab with fluorouracil, leucovorin, and irinotecan, folfiri, compared with folfiri alone as second-line treatment in patients with metastatic colorectal cancer. J Clin Oncol 28(31):4706– 4713., DOI 10.1200/JCO.2009.27.6055 Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, Rivera F, Kocakova I, Ruff P, Blasinska-Morawiec M, Smakal M, Canon JL, Rother M, Williams R, Rong A, Wiezorek J, Sidhu R, Patterson SD, 2013, Panitumumab-folfox4 treatment and ras mutations in colorectal cancer. N Engl J Med 369(11):1023–1034., DOI 10.1056/NEJMoa1305275 XingM, 2015, Braf mutation and thyroid cancer recurrence. J Clin Oncol 33(22):2482–2483., DOI 10.1200/JCO.2015.61.4016 Richman SD, SeymourMT, Chambers P, Elliott F, Daly CL, Meade AM, Taylor G, Barrett JH, Quirke P, 2009, Kras and braf mutations in advanced colorectal cancer are associated with poor prognosis but do not preclude benefit from oxaliplatin or irinotecan: results from the mrc focus trial. J Clin Oncol 27(35):5931–5937., DOI 10.1200/JCO.2009.22.4295 Mastroleo I, 2015, Post-trial obligations in the declaration of Helsinki 2013: classification, reconstruction and interpretation. Dev World Bioeth., DOI 10.1111/dewb.12099 Sarma DP, 1988, Dukes’ classification of rectal cancer. South Med J 81(3):407–408 Elzagheid A, Emaetig F, Alkikhia L, Buhmeida A, Syrjanen K, El- Faitori O, Latto M, Collan Y, Pyrhonen S, 2013, High cyclooxygenase-2 expression is associated with advanced stages in colorectal cancer. Anticancer Res 33(8):3137–3143 Huang F, Chang H, Greer A, Hillerman S, Reeves KA, HurlburtW, Cogswell J, Patel D, Qi Z, Fairchild C, Ryseck RP, Wong TW, Finckenstein FG, Jackson J, Carboni JM, 2015, Irs2 copy number gain, kras and braf mutation status as predictive biomarkers for response to the igf-1r/ir inhibitor bms-754807 in colorectal cancer cell lines. Mol Cancer Ther 14(2):620–630., DOI 10.1158/1535- 7163.MCT-14-0794-T Cunningham MP, Essapen S, Thomas H, Green M, Lovell DP, Topham C, Marks C, Modjtahedi H, 2006, Coexpression of the igf-ir, egfr and her-2 is common in colorectal cancer patients. Int J Oncol 28(2):329–335 KaladyMF, Dejulius KL, Sanchez JA, Jarrar A, Liu X, Manilich E, Skacel M, Church JM, 2012, Braf mutations in colorectal cancer are associated with distinct clinical characteristics and worse prognosis. Dis Colon rectum 55(2):128–133., DOI 10.1097/DCR.0b013 e31823c08b3 Stoeltzing O, Liu W, Fan F, Wagner C, Stengel K, Somcio RJ, Reinmuth N, Parikh AA, Hicklin DJ, Ellis LM, 2007, Regulation of cyclooxygenase-2, cox-2, expression in human pancreatic carcinoma cells by the insulin-like growth factor-i receptor, igf-ir, system. Cancer Lett 258(2):291–300., DOI 10.1016/j. canlet.2007.09.009 Wu CM, Li TM, Hsu SF, Su YC, Kao ST, Fong YC, Tang CH, 2011, IGF-I enhances alpha5beta1 integrin expression and cell motility in human chondrosarcoma cells. J Cell Physiol 226(12): 3270–3277., DOI 10.1002/jcp.22688 Liu JF, Fong YC, Chang CS, Huang CY, Chen HT, YangWH, Hsu CJ, Jeng LB, Chen CY, Tang CH, 2010, Cyclooxygenase-2 enhances alpha2beta1 integrin expression and cell migration via EP1 dependent signaling pathway in human chondrosarcoma cells. Mol Cancer 9:43., DOI 10.1186/1476-4598-9-43 Kim HS, Moon HG, HanW, Yom CK, KimWH, Kim JH, Noh DY, 2012, COX2 overexpression is a prognostic marker for stage III breast cancer. Breast Cancer Res Treat 132(1):51–59., DOI 10.1007 /s10549-011-1521-3 Thiel A, Mrena J, Ristimaki A, 2011, Cyclooxygenase-2 and gastric cancer. Cancer Metastasis Rev 30(3–4):387–395., DOI 10.1007 /s10555-011-9312-1 Lee Y C YCN and, Hsu T, 2013, Abstract 1146: Stk4 downregulation promotes tumor invasion/migration and is associated with poor prognosis in human colon cancer. Cancer Res 73, 8 Supplement): 1146–1146 Zare Mirzaei A, Abdorrazaghi F, Lotfi M, Kazemi Nejad Band Shayanfar N, 2015, Prognostic value of lymph node ratio in comparison to lymph node metastases in stage iii colon cancer. Iran J Pathol 10(2):127–135 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 45 EP 57 DI 10.1007/s12253-017-0195-5 PG 13 ER PT J AU Eliyatkin, N Aktas, S Diniz, G Ozgur, HH Ekin, YZ Kupelioglu, A AF Eliyatkin, Nuket Aktas, Safiye Diniz, Gulden Ozgur, Hakan Halil Ekin, Yildirim Zubeyde Kupelioglu, Ali TI Expression of Stromal Caveolin- 1 May Be a Predictor for Aggressive Behaviour of Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Caveolin 1; Predictive biomarker ID Breast cancer; Caveolin 1; Predictive biomarker AB Caveolin-1 (Cav-1) is well known as a principal scaffolding protein of caveolae which are specialized plasma membrane structures. The role of Cav-1 in tumorigenesis of breast cancers is relatively less studied. The aim of the present study is to describe the biological roles of Cav-1 in breast cancers considering its contrasting dual functions as an oncogene and as a tumor suppressor. This study included 71 females with breast cancer who had been histopathologically diagnosed in Private Gunes Pathology Laboratory between the years 2007, and 2012. The mean age is 52.48 ± 12.8 years. Patients were followed up for a mean period of 47.97 ± 20.48 months. We didn’t determine Cav-1 positive tumor cells. In 36 cases (50.7%), there were stromal expressions of Cav-1. In the statistical analysis, there was a statistically significant correlation between Cav-1 expression and ER (p = 0.033), metastasis (p = 0.005), lymphatic invasion (p = 0.000), nodal metastasis (p = 0,003), perinodal invasion (p = 0.003), metastasis (p = 0.005) and survival (p = 0.009). We found that Cav-1 expression is associated with tumor size, histological grade, lymph node involvement. Accordingly, we have suggested that Cav-1 may be a predictive biomarker for breast cancer. C1 [Eliyatkin, Nuket] Adnan Menderes University, Medical Faculty, Pathology DepartmentAydin, Turkey. [Aktas, Safiye] Dokuz Eylul University, Institute of OncologyIzmir, Turkey. [Diniz, Gulden] Tepecik Education and Research Hospital, Pathology Department, Kibris Sehitleri Cad 51/11 Alsancak, 35220 Izmir, Turkey. [Ozgur, Hakan Halil] Private Gunes Pathology LaboratoryIzmir, Turkey. [Ekin, Yildirim Zubeyde] Tepecik Education and Research Hospital, Pathology Department, Kibris Sehitleri Cad 51/11 Alsancak, 35220 Izmir, Turkey. [Kupelioglu, Ali] Private Gunes Pathology LaboratoryIzmir, Turkey. RP Diniz, G (reprint author), Tepecik Education and Research Hospital, Pathology Department, 35220 Izmir, Turkey. EM agdiniz@gmail.com CR Messa C, 2012, Unmasking epithelial-mesenchymal transition in a breast cancer primary culture: a study report.BMC Res Notes 5:343 Bravata V, Cammarata FP, Forte GI, Minafra L, 2013, "Omics" of HER2-positive breast cancer. OMICS 7(3):119–129 Minafra L, Bravata V, Forte GI, Cammarata FP, GilardiMC,Messa C, 2014, Gene-expression profiling of epithelial–mesenchymal transition in primary breast cancer cell culture. Anticancer Res 34: 2173–2184 BravataV, Stefano A, Cammarata FP,Minafra L, RussoG, Nicolosi S, Pulizzi S, Gelfi C, Gilardi MC, Messa C, 2013, Genotyping analysis and 18F-FDG uptake in breast cancer patients: a preliminary research. J Exp Clin Cancer Res 32:23 American Cancer Society: Cancer Facts and Figures, 2015, Atlanta, GA, American Cancer Society, 2015 Banin Hirata BK, Oda JM, Losi Guembarovski R, Ariza CB, de Oliveira CE, Watanabe MA, 2014, Molecular markers for breast cancer: prediction on tumor behaviour. Dis Markers 2014:513158 PucciM, Bravata V, 2015, Forte GIet al. Caveolin-1, breast cancer and ionizing radiation. Cancer Genomics Proteomics 12(3):143–152 Razani B, Woodman SE, Lisanti MP, 2002, Caveolae: from cell biology to animal physiology. Pharmacol Rev 4:431–467 Lisanti MP, Scherer P, Tang Z-L, Sargiacomo M, 1994, Caveolae, caveolin and caveolin-rich membrane domains: a signaling hypothesis. Trends Cell Biol 4:231–235 Kalluri R, ZeisbergM(2006, Fibroblasts in cancer. Nat Rev Cancer 6:392–401 Tlsty TD, Hein PW, 2011, Know thy neighbor: stromal cells can contribute oncogenic signals. Curr Opin Genet Dev 11:54–59 WitkiewiczA DA, Sotgia F et al, 2009, An absence of stromal caveolin-1 expression predicts early tumor recurrence and poor clinical outcome in human breast cancers. Am J Pathol 174: 2023–2034 Qian N, Ueno T, Kawaguchi-Sakita N et al, 2011, Prognostic significance of tumor/stromal caveolin-1 expression in breast cancer patients. Cancer Sci 102:1590–1596 Shan-Wei W, Kan-Lun X, Shu-Qin R, Li-Li Z, Li-Rong C, 2012, Overexpression of caveolin-1 in cancer-associated fibroblasts predicts good outcome in breast cancer. Breast Care, Basel, 7(6):477–483 Sagara Y, Mimori K, Yoshinaga K et al, 2004, Clinical significance of Caveolin-1, Caveolin-2 and HER-2/neu mRNA expression in human breast cancer. Br J Cancer 91:959–965 Yang G, Truong LD, Timme TL et al, 1998, Elevated expression of caveolin is associated with prostate and breast cancer. Clin Cancer Res 4:1873–1880 Hurlstone AF, Reid G, Reeves JR et al, 1999, Analysis of the Caveolin-1 gene at human chromosome 7q31.1 in primary tumors and tumor-derived cell lines. Oncogene 18:1881–1890 Mercier I, CasimiroMC,Wang C et al, 2008, Human breast cancerassociated fibroblasts, CAFs, show caveolin-1 downregulation and RB tumor suppressor functional inactivation: implications for the response to hormonal therapy. Cancer Biol Ther 7:1212–1225 Sloan EK, Ciocca DR, Pouliot N et al, 2009, Stromal cell expression of Caveolin-1 predicts outcome in breast cancer. Am J Pathol 174:2035–2043 Rao X, Evans J, Chae H et al, 2013, CpG island shore methylation regulates caveolin-1 expression in breast cancer. Oncogene 32(38): 4519–4528 Engelman JA, Lee RJ, Karnezis A et al, 1998, Reciprocal regulation of neu tyrosine kinase activity and caveolin-1 protein expression in vitro and in vivo. Implications for human breast cancer. J Biol Chem 273:20448–20455 Chung YC, Kuo JF, Wei WC et al, 2015, Caveolin-1 dependent endocytosis enhances the chemosensitivity of HER-2 positive breast cancer cells to trastuzumab emtansine, T-DM1). PLoS One 10(7):e0133072 Park SS, Kim JE, Kim YA, Kim YC, Kim SW, 2005, Caveolin-1 is down-regulated and inversely correlated with HER2 and EGFR expression status in invasive ductal carcinoma of the breast. Histopathology 7(6):625–630 Savage K, Lambros MB, Robertson D et al, 2007, Caveolin-1 is overexpressed and amplified in a subset of basal-like and metaplastic breast carcinomas: a morphologic, ultrastructural, immunohistochemical, and in situ hybridization analysis. Clin Cancer Res 13: 90–101 Huang CF, Yu GT, Wang WM et al, 2014, Prognostic and predictive values of SPP1, PAI and caveolin-1 in patients with oral squamous cell carcinoma. Int J Clin Exp Pathol 7(9):6032–6039 Chatterjee M, Ben-Josef E, Thomas DG et al, 2015, Caveolin-1 is associated with tumor progression and confers a multi-modality resistance phenotype in pancreatic cancer. Sci Rep 12(5):10867 Sayhan S, Diniz G, Karadeniz T, Ayaz D, Kahraman DS, Gokcu M, Yildirim HT, 2015, Expression of caveolin-1 in peritumoral stroma is associated histological grade in ovarian serous tumors. Ginekol Pol 6(6):424–428 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 59 EP 65 DI 10.1007/s12253-017-0212-8 PG 7 ER PT J AU Chen, P Wang, YP Mou, Dl Li, ZY Qu, QM Wang, HY Deng, Y Li, XF Wang, T Xu, XH Zhao, G AF Chen, Ping Wang, Ying-Peng Mou, Dan-lei Li, Zheng-Yi Qu, Qiu-Min Wang, Hong-Yan Deng, Yuan Li, Xiao-Feng Wang, Ting Xu, Xian-Hao Zhao, Gang TI Pathological Findings in Myasthenia Gravis Patients with Thymic Hyperplasia and Thymoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Myasthenia gravis; Thymic hyperplasia; Thymoma; Thymectomy ID Myasthenia gravis; Thymic hyperplasia; Thymoma; Thymectomy AB Thymectomy is routinely carried out in patients with myasthenia gravis (MG) and thymomas. However, there is still a dispute as to whether MG patients with thymic hyperplasia should undergo thymectomy. We aimed to investigate the pathological findings in the thymus in patients with co-existing MG and thymic hyperplasia or thymomas treated with thymectomy, as well as effects of immunosuppression. Thirty-three patients with MG were selected and grouped accordingly: patients with no thymic abnormalities, patients with thymic hyperplasia, and patients with thymomas. All patients were treated with methylprednisolone alongside immunosuppression. A separate cohort of 24 MG patients with thymic hyperplasia or thymomas and treated with thymectomy were selected. As controls, 5 patients with thymomas or thymic carcinoma without MG were selected. Expression of CD5, extracellular regulated protein kinases1/2 mitogen activated protein kinase (ERK1/2MAPKs) and CD95 ligand (FasL) in the thymus was examined. Methylprednisolone and immunosuppressive therapy are highly effective in MG patients with normal thymus tissue and MG patients with thymic hyperplasia compared to MG patients with thymomas alone. CD5 expression was highest in MG patients with thymic hyperplasia, correlating with expression of ERK1/2MAPKs. FasL expression was similar across all groups. Thymomas may be distinguished from thymic hyperplasia by expression of CD5 and ERK1/2MAPKs. Thymectomy is the preferred treatment for MG patients with thymomas but may not be necessary in MG patients with thymic hyperplasia who are treated with immunosuppressive therapy. C1 [Chen, Ping] Fourth Military Medical University, Xijing Hospital, Department of Neurology, No. 169, Changle West Road, 710032 Xi’an, China. [Wang, Ying-Peng] Beijing Aerospace General Hospital, Department of Neurology, 100076 Beijing, China. [Mou, Dan-lei] Capital Medical University, Beijing You’an Hospital, Department of Infectious disease, 100075 Beijing, China. [Li, Zheng-Yi] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Neurology, 710061 Xi’an, China. [Qu, Qiu-Min] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Neurology, 710061 Xi’an, China. [Wang, Hong-Yan] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Pathology, 710061 Xi’an, China. [Deng, Yuan] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Pathology, 710061 Xi’an, China. [Li, Xiao-Feng] Xi’an Jiao Tong University, First Affiliated Hospital of Medical College, Department of Pathology, 710061 Xi’an, China. [Wang, Ting] Fourth Military Medical University, Xijing Hospital, Department of Neurology, No. 169, Changle West Road, 710032 Xi’an, China. [Xu, Xian-Hao] Federal Ministry of Health, Beijing Hospital, Department of Neurology, No. 1, Dahua Road, 100730 Beijing, China. [Zhao, Gang] Fourth Military Medical University, Xijing Hospital, Department of Neurology, No. 169, Changle West Road, 710032 Xi’an, China. RP Zhao, G (reprint author), Fourth Military Medical University, Xijing Hospital, Department of Neurology, 710032 Xi’an, China. EM zhaogang@fmmu.edu.cn CR Vincent A, 2002, Unravelling the pathogenesis of myasthenia gravis. Nat Rev Immunol 2(10):797–804 Moser B, Bekos C, Zimprich F, Nickl S, Klepetko W, Ankersmit J, 2012, The receptor for advanced glycation end products and its ligands in patients with myasthenia gravis. Biochem Biophys Res Commun 420(1):96–101 Berrih-Aknin S, Le Panse R, 2014, Myasthenia gravis: a comprehensive review of immune dysregulation and etiological mechanisms. J Autoimmun 52:90–100 Gronseth GS, Barohn R, 2000, Practice parameter: thymectomy for autoimmune myasthenia gravis, an evidence-based review). Report from the quality standards subcommittee of the American academy of neurology. Neurology 55:7–15 Bak V, Spalek P, Petrovajova T, Vichova B, Oravsky M, Luha J, Schnorrer M, 2013, Thymic tumours associated with Myasthenia gravis: a long term observation study of operated patients. Bratisl Lek Listy 114(8):464–468 Cavalcante P, Le Panse R, Berrih-Aknin S, Maggi L, Antozzi C, Baggi F, Bernasconi P, Mantegazza R, 2011, The thymus in myasthenia gravis: site of "innate autoimmunity"? Muscle Nerve 44(4): 467–484 Geuder KI, Marx A,Witzemann V, Schalke B, Kirchner T, Muller- Hermelink HK, 1992, Genomic organization and lack of transcription of the nicotinic acetylcholine receptor subunit genes in myasthenia gravis-associated thymoma. Lab Investig 66(4):452–458 Sandri A, Cusumano G, Lococo F et al, 2014, Long-term results after treatment for recurrent thymoma: a multicenter analysis. J Thorac Oncol 9(12):1796–1804 De Meel RH, Lipka AF, Van Zwet EW, Niks EH, Verschuuren JJ, 2015, Prognostic factors for exacerbations and emergency treatments in myasthenia gravis. J Neuroimmunol 282:123–125 Safieddine N, Liu G, Cuningham K et al, 2014, Prognostic factors for cure, recurrence and long-term survival after surgical resection of thymoma. J Thorac Oncol 9(7):1018–1022 Uzawa A, Kawaguchi N, Kanai Tet al, 2015, Two-year outcome of thymectomy in non-thymomatous late-onset myasthenia gravis. J Neurol 262(4):1019–1023 Azzam HS, DeJarnette JB, Huang K et al, 2001, Fine tuning of TCR signaling by CD5. J Immunol 166(9):5464–5472 Tarakhovsky A, Kanner SB, Hombach J et al, 1995)A role for CD5 in TCR-mediated signal transduction and thymocyte selection. Science 269(5223):535–537 Ragheb S, Lisak RP, 1992, Effect of clinical status and treatment on the frequency of CD5+ B cells in patients with myasthenia gravis. Neurology 42(5):1076–1080 Dalloul A, 2009, CD5: a safeguard against autoimmunity and a shield for cancer cells. Autoimmun Rev 8(4):349–353 Ragheb S, Lisak RP, 1990, The frequency of CD5+ B lymphocytes in the peripheral blood of patients with myasthenia gravis. Neurology 40(7):1120–1124 Colombara M, Antonini V, Riviera AP et al, 2005, Constitutive activation of p38 and ERK1/2 MAPKs in epithelial cells of myasthenic thymus leads to IL-6 and RANTES overexpression: effects on survival and migration of peripheral T and B cells. J Immunol 175(10):7021–7028 Friedlein G, El Hage F, Vergnon I et al, 2007, Human CD5 protects circulating tumor antigen-specific CTL from tumor-mediated activation- induced cell death. J Immunol 178(11):6821–6827 Lundy SK, Fox DA, 2009, Reduced Fas ligand-expressing splenic CD5+ B lymphocytes in severe collagen-induced arthritis. Arthritis Res Ther 11(4):R128 Aruna BV, Ben-David H, SelaMet al, 2006, A dual altered peptide ligand down-regulates myasthenogenic T cell responses and reverses experimental autoimmune myasthenia gravis via upregulation of Fas–FasL-mediated apoptosis. Immunology 118(3): 413–424 Zhang J, Gao JX, Salojin K et al, 2000, Regulation of Fas ligand expression during activation-induced cell death in T cells by p38 mitogen-activated protein kinase and c-Jun NH2-terminal kinase. Exp Med 191:1017–1030 Krammer PH, 2000, CD95's deadly mission in the immune system. Nature 407:789–495 Davidson WF, Haudenschild C, Kwon J, Williams MS, 2002, T cell receptor ligation triggers novel nonapoptotic cell death pathways that are Fas-independent or Fas-dependent. J Immunol 169:6218–6230 Jaretzki A 3rd, Barohn RJ, Ernstoff RM, Kaminski HJ, Keesey JC, Penn AS, Sanders DB, 2000)Myasthenia gravis: recommendations for clinical research standards. J Ann Thorac Surg 70(1):327–334 Osserman KE,GenkinsG, 1961, Studies inmyasthenia gravis.N Y State J Med 61:2076–2085 Murai H, 2015, Myasthenia gravis: epidemiology and prognosis. Nihon Rinsho 73(Suppl 7):472–476 Utsugisawa K, Nagane Y, Suzuki S, Suzuki N, 2012, Directions for use of corticosteroids and calcineurin inhibitors against generalized myasthenia gravis: therapeutic strategies that can lead to early improvements and veer away fromhigh-dose oral corticosteroids. Clin Neuro l52(11):1047–1050 Kim JY, Park KD, Richman DP, 2011, Treatment of myasthenia gravis based on its immunopathogenesis. J Clin Neurol 7(4):173–183 Diaz A, Black E, Dunning J, 2014, Is thymectomy in nonthymomatous myasthenia gravis of any benefit? Interact Cardiovasc Thorac Surg 18(3):381–389 Le Panse R, Bismuth J, Cizeron-Clairac G et al, 2010, Thymic remodeling associated with hyperplasia in myasthenia gravis. Autoimmunity 43(5–6):401–412 Yi Q, Ahlberg R, Pirskanen R, Lefvert AK, 1992, Levels of CD5+ B lymphocytes do not differ between patients with myasthenia gravis and healthy individuals. Neurology 42(5):1081–1084 Aruna BV, Ben-David H, Sela M, Mozes E, 2006, A dual altered peptide ligand down-regulates myasthenogenic Tcell responses and reverses experimental autoimmune myasthenia gravis via upregulation of Fas-FasL-mediated apoptosis. Immunology 118(3): 413–424 Shin HJ, Katz RL, 1998, Thymic neoplasia as represented by fine needle aspiration biopsy of anterior mediastinal masses. A practical approach to the differential diagnosis. Acta Cyto l42(4):855–864 Li H, Wang DL, Liu XW, Geng ZJ, Xie CM, 2013, Computed tomography characterization of neuroendocrine tumors of the thymus can aid identification and treatment. Acta Radio l54(2):175–180 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 67 EP 74 DI 10.1007/s12253-017-0213-7 PG 8 ER PT J AU He, T Guo, J Song, H Zhu, H Di, X Min, H Wang, Y Chen, G Dai, W Ma, J Sun, X Ma, J AF He, Tianli Guo, Jiayou Song, Hongmei Zhu, Hongcheng Di, Xiaoke Min, Hua Wang, Yuandong Chen, Guangzong Dai, Wangshu Ma, Jianhua Sun, Xinchen Ma, Jianxin TI Nutlin-3, an Antagonist of MDM2, Enhances the Radiosensitivity of Esophageal Squamous Cancer with Wild-Type p53 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal squamous cancer; MDM2; Radiotherapy; Nutlin-3; p53 ID Esophageal squamous cancer; MDM2; Radiotherapy; Nutlin-3; p53 AB Murine double minute 2 (MDM2) negatively regulates the activity of the p53 protein and plays a vital role in cell cycle arrest, apoptosis, and senescence mediated by p53. Nutlin-3, an antagonist of MDM2, is frequently used in anticancer studies. In many human tumors, nutlin-3 stabilizes p53 status and enhances p53 expression in cells with wild-type p53. However, the effect of nutlin-3 combined with radiotherapy on esophageal squamous cancer (ESCC) has not been reported. In this study, we examined whether nutlin-3 increases the radiosensitivity of ESCC in vitro and in vivo. We chose two cell lines, ECA-109 (wild-type p53) and TE- 13 (p53 mutated), for the following experiments. Cell proliferation and clonogenic survival experiments showed that nutlin-3 inhibits the cell growth and colony formation of ECA-109 cells in a dose-dependent manner. Flow cytometry analysis showed that the apoptosis rate of ECA-109 cells cotreated with nutlin-3 and irradiation(IR) was significantly increased compared with cells treated with irradiation or nutlin- 3 alone.Western blotting detected the expression of apoptosisassociated proteins in ECA-109 cells in response to nutlin-3 and irradiation. These effects were not evident in TE-13 cells. Xenograft mouse models indicated that nutlin-3 suppresses tumor growth and promotes radiosensitivity in the ESCC cell line ECA-109 in vivo. We have demonstrated that cotreatment of nutlin-3 with irradiation can significantly inhibit the growth and improve the radiosensitivity of ESCC cells with wild-type p53. The study suggests that nutlin-3 may be a potent therapeutic agent in conjunction with radiotherapy in ESCC. C1 [He, Tianli] Lianyungang Hospital Affiliated to Bengbu Medical College, The Second People’s Hospital of Lian Yungang, Department of radiotherapy, 161 Xingfu Road, 222000 Lian Yungang, Jiangsu Province, China. [Guo, Jiayou] The Dongfang Hospital of Lian Yungang, Department of radiotherapy, 57 Zhonghua West Road, 222000 Lian Yungang, Jiangsu Province, China. [Song, Hongmei] Taian City Central Hospital, 29 Longtan Road, 271000 Taian, Shandong Province, China. [Zhu, Hongcheng] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, China. [Di, Xiaoke] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, China. [Min, Hua] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, China. [Wang, Yuandong] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, China. [Chen, Guangzong] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, China. [Dai, Wangshu] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, China. [Ma, Jianhua] Lianyungang Hospital Affiliated to Bengbu Medical College, The Second People’s Hospital of Lian Yungang, Department of radiotherapy, 161 Xingfu Road, 222000 Lian Yungang, Jiangsu Province, China. [Sun, Xinchen] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, China. [Ma, Jianxin] The Dongfang Hospital of Lian Yungang, Department of radiotherapy, 57 Zhonghua West Road, 222000 Lian Yungang, Jiangsu Province, China. RP Ma, J (reprint author), The Dongfang Hospital of Lian Yungang, Department of radiotherapy, 222000 Lian Yungang, China. EM mjx3416@163.com CR Yang X, Yang B, Cai J, Zhang C, Zhang Q, Xu L et al, 2013, Berberine enhances radiosensitivity of esophageal squamous cancer by targeting hif-1α in vitro and in vivo. Cancer Biol Ther 14(11):1068–1073 Shridhar R, Almhanna K, Meredith KL, BiagioliMC, ChuongMD, Cruz A, Hoffe SE, 2013, Radiation therapy and esophageal cancer. Cancer Control J Moffitt Cancer Center 20(2):97–110 Momand J, Zambetti GP, Olson DC, George D, Levine AJ, 1992, The mdm-2, oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 69(7):1237–1245 Cahilly-Snyder L, Yang-Feng T, Francke U, George DL, 1987, Molecular analysis and chromosomal mapping of amplified genes isolated from a transformed mouse 3t3 cell line. Somat Cell Mol Genet 13(3):235–244 Fakharzadeh SS, Trusko SP, George DL, 1991, Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line. EMBO J 10(6):1565–1569 Polager S, Ginsberg D, 2009, P53 and e2f: partners in life and death. Nat Rev Cancer 9(10):738–748 Momand J, Jung D,Wilczynski S, Niland J, 1998, The mdm2 gene amplification database. Nucleic Acids Res 26(15):3453–3459 Freedman DA, Wu L, Levine AJ, 1999, Functions of the mdm2 oncoprotein. Cell Mol Life Sci: CMLS 55(1):96–107 Zhang, Wang H, 2000, Mdm2 oncogene as a novel target for human cancer therapy. Curr Pharm Des 6(4):393–416 Wan Y,WuW, Yin Z, Guan P, Zhou B, 2011, Mdm2 snp309, genegene interaction, and tumor susceptibility: an updated meta-analysis. BMC Cancer 11(1):–9 Rayburn E, Zhang R, He J, Wang H, 2005, Mdm2 and human malignancies: expression, clinical pathology, prognostic markers, and implications for chemotherapy. Curr Cancer Drug Targets 5(1):27–41 Okamoto H, Fujishima F, Nakamura Y, Zuguchi M, Miyata G, Kamei T et al, 2013, Murine double minute 2 and its association with chemoradioresistance of esophageal squamous cell carcinoma. Anticancer Res 33(4):1463–1471 Jiangjiang K, Srivenugopal M, Wang, Ruiwen et al, 2013, The mdm2-p53 pathway revisited. J Biol Res 27(4):254–271 Yuhan Z, Haiyang, Wenwei, 2014, The regulation of mdm2 oncogene and its impact on human cancers. Acta Biochim Biophys Sin 46(3):180–189 Bouska A, Lushnikova T, Plaza S, Eischen CM, 2008, Mdm2 promotes genetic instability and transformation independent of p53. Mol Cell Biol 28(15):4862–4874 Villalonga-Planells R, Coll-Mulet L, Martinez-Soler F, Castano E, Acebes JJ, Gimenez-Bonafe P et al, 2011, Activation of p53 by nutlin-3a induces apoptosis and cellular senescence in human glioblastoma multiforme. PLoS One 6(4):e18588–e18588 FuW, Ma Q, Chen L, Li P, Zhang M, Ramamoorthy S et al, 2009, Mdm2 acts downstream of p53 as an e3 ligase to promote foxo ubiquitination and degradation. J Biol Chem284(21):13987–14000 Gu L, Zhu N, Zhang H, Durden DL, Feng Y, Zhou M, 2009, Regulation of xiap translation and induction by mdm2 following irradiation. Cancer Cell 15(5):363–375 Luo H, Yount C, Lang H, Yang A, Riemer EC, LyonsK et al, 2013, Activation of p53 with nutlin-3a radiosensitizes lung cancer cells via enhancing radiation-induced premature senescence. Lung Cancer 81(2):167–173 Arya AK, Elfert A, Devling T, Eccles RM, Aslam MA, Rubbi CP et al, 2010, Nutlin-3, the small-molecule inhibitor of mdm2, promotes senescence and radiosensitises laryngeal carcinoma cells harbouring wild-type p53. Br J Cancer 103(2):186–195 Lehmann B, Mccubrey J, Jefferson H, Paine M, Chappell W, Terrian D, 2007, A dominant role for p53-dependent cellular senescence in radiosensitization of human prostate cancer cells. Cell Cycle 6(5):595–605 Cao, C, Shinohara, ET, Subhawong TK, Geng L, Kim KW & Albert JM et al., 1972). Radiosensitization of lung cancer by nutlin, an inhibitor of murine double minute 2. International Broadcasting Convention, 4–8 September 1972 [at] Grosvenor House, Park Lane, London W.1. Institution of Electrical Engineers Supiot S, Hill RP, Bristow RG, 2008, Nutlin-3 radiosensitizes hypoxic prostate cancer cells independent of p53. Mol Cancer Ther 7(4):993–999 Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z, Kong N, Kammlott U, Lukacs C, Klein C, Fotouhi N, Liu EA, 2007, In-vivo activation of the p53 pathway by small-molecule antagonists of mdm2. Tanpakushitsu Kakusan Koso Protein Nucleic Acid Enzyme 52(13 Suppl):844–848 Vassilev LT, 2007, Mdm2 inhibitors for cancer therapy. TrendsMol Med 13(1):23–31 Logan IR, Mcneill HV, Cook S, Lu X, Lunec J, Robson CN, 2007, Analysis of the mdm2 antagonist nutlin-3 in human prostate cancer cells. Prostate 67(8):900–906 Vanderborght A, Valckx A, Dun JV, Grandperret T, Schepper SD, Vialard J et al, 2006, Effect of an hdm-2 antagonist peptide inhibitor on cell cycle progression in p53-deficient h1299 human lung carcinoma cells. Oncogene 25(50):6672–6677 Stuhmer T, ChatterjeeM, Hildebrandt M, Herrmann P, Gollasch H, Gerecke C et al, 2005, Nongenotoxic activation of the p53 pathway as a therapeutic strategy for multiple myeloma. Blood 106(10): 3609–3617 Kojima K, Konopleva M, Samudio IJ, Shikami M, Cabreira- Hansen M, McQueen T, Ruvolo V, Tsao T, Zeng Z, Vassilev LT, Andreeff M, 2005, Mdm2 antagonists induce p53-dependent apoptosis in aml: implications for leukemia therapy. Blood 106(9): 3150–3159 Coll-Mulet L, Iglesias-Serret D, Santidrian AF, Cosialls AM, De FM, Castano E et al, 2006, Mdm2 antagonists activate p53 and synergize with genotoxic drugs in b-cell chronic lymphocytic leukemia cells. Blood 107(10):4109–4114 Drakos E, Thomaides A, Medeiros LJ, Li J, Leventaki V, Konopleva M et al, 2007, Inhibition of p53-murine double minute 2 interaction by nutlin-3a stabilizes p53 and induces cell cycle arrest and apoptosis in hodgkin lymphoma. Clin Cancer Res Off J Am Assoc Cancer Res 13(11):3380–3387 Ye Z, Fang J, Dai S, Wang Y, Fu Z, FengWet al, 2015, Microrna- 34a induces a senescence-like change via the down-regulation of sirt1 and up-regulation of p53 protein in human esophageal squamous cancer cells with a wild-type p53 gene background. Cancer Lett 370(2):216–221 Barnas C, Martel-Planche G, Furukawa Y, HollsteinM, Montesano R, Hainaut P, 1997, Inactivation of the p53 protein in cell lines derived from human esophageal cancers. Int J Cancer 71(1):79–87 Costa NMD, Hautefeuille A, Cros MP, Melendez ME, Waters T, Swann P et al, 2012, Transcriptional regulation of thymine dna glycosylase, tdg, by the tumor suppressor protein p53. Cell Cycle 11(24):4570–4578 Zhang C, Yang X, Zhang Q, Guo Q, He J, Qin Q et al, 2014, Stat3 inhibitor nsc74859 radiosensitizes esophageal cancer via the downregulation of hif-1α. Tumour Biol J Int Soc Oncodevelopmental Biol Med 35(10):9793–9799 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 75 EP 81 DI 10.1007/s12253-017-0215-5 PG 7 ER PT J AU Kiraly, PA Kallay, K Gango, A Kellner, Egyed, M Szoke, A Kiss, R Valyi-Nagy, I Csomor, J Matolcsy, A Bodor, Cs AF Kiraly, Peter Attila Kallay, Krisztian Gango, Ambrus Kellner, Adam Egyed, Miklos Szoke, Anita Kiss, Richard Valyi-Nagy, Istvan Csomor, Judit Matolcsy, Andras Bodor, Csaba TI Familial Acute Myeloid Leukemia and Myelodysplasia in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Familial MDS/AML; Genetic predisposition; Germline mutation ID Familial MDS/AML; Genetic predisposition; Germline mutation AB Although genetic predisposition to haematological malignancies has long been known, genetic testing is not yet the part of the routine diagnostics. In the last ten years, next generation sequencing based studies identified novel germline mutations in the background of familial aggregation of certain haematologic disorders including myelodysplastic syndromes (MDS) and acute myeloid leukaemia (AML). This is supported by the fact that the myeloid neoplasms with genetic predisposition represent a new category in the revised 2016 World Health Organization classification. According to the new classification, these disorders are subdivided based on the clinical and genetic features, including myeloid neoplasms with germline predisposition alone, or with pre-existing platelet disorder, cytopaenias or other organ failures. The predisposing genetic factors include mutations in the RUNX1, CEBPA, GATA2, ANKRD26, ETV6, DDX41, TERC or TERT and SRP72 genes. The genes affected in these syndromes are important regulators of haemopoiesis and are frequently implicated in leukaemogenesis, providing deeper insight into the understanding of normal and malignant haemopoiesis. Despite the growing knowledge of germline predisposing events in the background of familial myeloid malignancies, the germline genetic component is still unknown in a subset of these pedigrees. Here, we present the first study of inherited myeloid malignancies in Hungary.We identified three families with apparent clustering of myeloid malignancies with nine affected individuals across these pedigrees. All tested individuals were negative for CEBPA, GATA2, RUNX1, ANKRD26, ETV6, DDX41, TERC or TERT and SRP72 mutations, suggesting the presence of so far unidentified predisposing mutations. C1 [Kiraly, Peter Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kallay, Krisztian] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Gango, Ambrus] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kellner, Adam] Kaposi Mor Teaching Hospital, Department of HematologyKaposvar, Hungary. [Egyed, Miklos] Kaposi Mor Teaching Hospital, Department of HematologyKaposvar, Hungary. [Szoke, Anita] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Kiss, Richard] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Valyi-Nagy, Istvan] Del-Pesti CentrumkorhazBudapest, Hungary. [Csomor, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Matolcsy, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Bodor, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Bodor, Cs (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM bodor.csaba1@med.semmelweis-univ.hu CR Porter CC, 2016, Germ line mutations associated with leukemias. Hematol Am Soc Hematol Educ Prog 2016(1):302–308 WestAH, Godley LA, Churpek JE, 2014, Familialmyelodysplastic syndrome/acute leukemia syndromes: a review and utility for translational investigations. Ann N YAcad Sci 1310:111–118 Hahn CN, Chong CE, Carmichael CL,Wilkins EJ, Brautigan PJ, Li XC et al, 2011, Heritable GATA2 mutations associated with familial myelodysplastic syndrome and acute myeloid leukemia. Nat Genet 43(10):1012–1017 KirwanM,Walne AJ, Plagnol V, Velangi M, Ho A, Hossain U et al, 2012, Exome sequencing identifies autosomal-dominant SRP72 mutations associated with familial aplasia and myelodysplasia. Am J Hum Genet 90(5):888–892 Pippucci T, Savoia A, Perrotta S, Pujol-Moix N, Noris P, Castegnaro G et al, 2011, Mutations in the 5′ UTR of ANKRD26, the ankirin repeat domain 26 gene, cause an autosomal-dominant form of inherited thrombocytopenia, THC2. Am J Hum Genet 88(1):115–120 Polprasert C, Schulze I, Sekeres MA, Makishima H, Przychodzen B, Hosono N et al, 2015, Inherited and somatic defects in DDX41 in myeloid neoplasms. Cancer Cell 27(5):658–670 Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM et al, 2016, The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127(20):2391–2405 Duployez N, Lejeune S, Renneville A, Preudhomme C, 2016, Myelodysplastic syndromes and acute leukemia with genetic predispositions: a new challenge for hematologists. Expert Rev Hematol 9(12):1189–1202 Churpek JE, 2016, Godley LA. How I diagnose and manage individuals at risk for inherited myeloid malignancies, Blood Pabst T, Mueller BU, Zhang P, Radomska HS, Narravula S, Schnittger S et al, 2001, Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha, C/EBPalpha), in acute myeloid leukemia. Nat Genet 27(3):263–270 Li R, Sobreira N, Witmer PD, Pratz KW, Braunstein EM, 2016, Two novel germline DDX41 mutations in a family with inherited myelodysplasia/acute myeloid leukemia. Haematologica 101(6): e228–e231 Cardoso SR, Ryan G, Walne AJ, Ellison A, Lowe R, Tummala H et al, 2016, Germline heterozygous DDX41 variants in a subset of familial myelodysplasia and acute myeloid leukemia. Leukemia 30(10):2083–2086 Smith ML, Cavenagh JD, Lister TA, Fitzgibbon J, 2004, Mutation of CEBPA in familial acute myeloid leukemia. N Engl J Med 351(23):2403–2407 Dickinson RE, Griffin H, Bigley V, Reynard LN, Hussain R, Haniffa M et al, 2011, Exome sequencing identifies GATA-2 mutation as the cause of dendritic cell, monocyte, B and NK lymphoid deficiency. Blood 118(10):2656–2658 Hsu AP, Sampaio EP, Khan J, Calvo KR, Lemieux JE, Patel SYet al, 2011, Mutations in GATA2 are associated with the autosomal dominant and sporadic monocytopenia and mycobacterial infection, MonoMAC, syndrome. Blood 118(10):2653–2655 Armanios M, 2009, Syndromes of telomere shortening. Annu Rev Genomics Hum Genet 10:45–61 SongWJ, Sullivan MG, Legare RD, Hutchings S, Tan X, Kufrin D et al, 1999, Haploinsufficiency of CBFA2 causes familial thrombocytopenia with propensity to develop acute myelogenous leukaemia. Nat Genet 23(2):166–175 ZhangMY, Churpek JE, Keel SB,Walsh T, LeeMK, Loeb KR et al, 2015, Germline ETV6 mutations in familial thrombocytopenia and hematologic malignancy. Nat Genet 47(2):180–185 Haslam K, Langabeer SE, Hayat A, Conneally E, Vandenberghe E, 2016, Targeted next-generation sequencing of familial platelet disorder with predisposition to acute myeloid leukaemia. Br J Haematol 175(1):161–163 Preudhomme C, Renneville A, Bourdon V, Philippe N, Roche- Lestienne C, Boissel N et al, 2009, High frequency of RUNX1 biallelic alteration in acute myeloid leukemia secondary to familial platelet disorder. Blood 113(22):5583–5587 Ding LW, Ikezoe T, TanKT, Mori M, MayakondaA, ChienW, et al., 2016)Mutational profiling of a MonoMAC syndrome family with GATA2 deficiency. Leukemia Godley LA, 2014, Inherited predisposition to acute myeloid leukemia. Semin Hematol 51(4):306–321 DiNardo CD, Bannon SA, Routbort M, Franklin A, Mork M, Armanios M et al, 2016, Evaluation of patients and families with concern for predispositions to hematologic malignancies within the hereditary hematologic malignancy clinic, HHMC). Clin Lymphoma, Myeloma Leuk 16(7):417–28.e2 Noetzli L, Lo RW, Lee-Sherick AB, Callaghan M, Noris P, Savoia A et al, 2015, Germline mutations in ETV6 are associated with thrombocytopenia, red cell macrocytosis and predisposition to lymphoblastic leukemia. Nat Genet 47(5):535–538 Vulliamy T, Marrone A, Dokal I, Mason PJ, 2002, Association between aplastic anaemia and mutations in telomerase RNA. Lancet 359(9324):2168–2170 Yamaguchi H, Calado RT, Ly H, Kajigaya S, Baerlocher GM, Chanock SJ et al, 2005, Mutations in TERT, the gene for telomerase reverse transcriptase, in aplastic anemia. N Engl J Med 352(14):1413–1424 Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Buchner T et al, 2017, Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 129(4):424–447 Churpek JE, Lorenz R, Nedumgottil S, Onel K, Olopade OI, Sorrell A et al, 2013, Proposal for the clinical detection and management of patients and their family members with familial myelodysplastic syndrome/acute leukemia predisposition syndromes. Leuk Lymphoma 54(1):28–35 Green CL, Tawana K, Hills RK, Bodor C, Fitzgibbon J, Inglott S et al, 2013, GATA2 mutations in sporadic and familial acute myeloid leukaemia patients with CEBPA mutations. Br J Haematol 161(5):701–705 Greif PA, Dufour A, Konstandin NP, Ksienzyk B, Zellmeier E, Tizazu B et al, 2012, GATA2 zinc finger 1 mutations associated with biallelic CEBPA mutations define a unique genetic entity of acute myeloid leukemia. Blood 120(2):395–403 Osato M, Yanagida M, Shigesada K, Ito Y, 2001, Point mutations of the RUNx1/AML1 gene in sporadic and familial myeloid leukemias. Int J Hematol 74(3):245–251 Preudhomme C, Sagot C, Boissel N, Cayuela JM, Tigaud I, de Botton S et al, 2002, Favorable prognostic significance of CEBPA mutations in patients with de novo acute myeloid leukemia: a study from the acute leukemia French association, ALFA). Blood 100(8):2717–2723 Galili NT, Trifonov, V., Ewalt, M., Mukherjee, S., Rabadan, R., Raza, A., 2012, Identification of Dido1 Mutation Associated with Familial Myelodysplastic Syndrome, MDS)/Acute Myeloid Leukemia, AML, Blood. 120:169 Yang F, Gong Q, Shi W, Zou Y, Shi J, Wei F et al, 2016, Aberrant DNA methylation of acute myeloid leukemia and colorectal cancer in a Chinese pedigree with a MLL3 germline mutation. Tumour Biol 37(9):12609–12618 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 83 EP 88 DI 10.1007/s12253-017-0216-4 PG 6 ER PT J AU Golan, Sh Gerber, G Margel, D Rath-Wolfson, L Ehrlich, Y Koren, R Lifshitz, D AF Golan, Shay Gerber, Glenn Margel, David Rath-Wolfson, Lea Ehrlich, Yaron Koren, Rumelia Lifshitz, David TI A Novel Technique to Improve the Processing of Minute Ureteroscopic Biopsies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Upper urinary tract urothelial carcinoma; Ureteroscopy; Biopsy; Pathology ID Upper urinary tract urothelial carcinoma; Ureteroscopy; Biopsy; Pathology AB To examine the ability of a new specimen handling technique to improve histopathological yield of ureteroscopic biopsies, performed in patients with suspected upper tract urothelial carcinoma (UTUC). In a bi-center retrospective study we compared the results of the new tissue handling technique (group 1) with the standard technique (group 2). In the new technique, to achieve maximal tissue preservation, the specimen is mounted on filter paper prior to embedding in paraffin. Multivariate analysis was performed to determine which factors are associated with optimal histological results. We further compared the biopsies with the final specimen in a subgroup of patients who underwent nephroureterectomy (NU). Of 55 ureteroscopic biopsies, 1 biopsy from group 1 (new technique) and 3 biopsies from group 2 (standard technique) were inadequate for pathological examination. 51 UTUC specimens were analyzed. Tumor grade and stage were determined in 85% and 63% of the patients in group 1 and in 83% and 25% of group 2 (p=0.85 and p=0.007). Orientation was preserved in 82% of group 1 and 42% of group 2 (p=0.003). On multivariate analysis biopsy technique and biopsy diameter were found to predict stage determination (p=0.01 and p=0.007) and tissue orientation (p=0.005 and p=0.04). Among patients who underwent NU, stage concordance between the biopsy and final pathology was observed in 56% and 27% of the patients in group 1 and 2, respectively. The new processing technique for small UTUC forceps biopsies decreases the rate of biopsies with insufficient material and improves biopsy interpretation. C1 [Golan, Shay] University of Chicago Medicine, Department of Surgery, 5841 S. Maryland Ave, 60637 Chicago, IL, USA. [Gerber, Glenn] University of Chicago Medicine, Department of Surgery, 5841 S. Maryland Ave, 60637 Chicago, IL, USA. [Margel, David] Hasharon Hospital, Rabin Medical Center, Petah-Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Department of UrologyTel Aviv, Israel. [Rath-Wolfson, Lea] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of PathologyTel Aviv, Israel. [Ehrlich, Yaron] Hasharon Hospital, Rabin Medical Center, Petah-Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Department of UrologyTel Aviv, Israel. [Koren, Rumelia] Rabin Medical Center, Petah Tikva and Sackler Faculty of Medicine, Tel Aviv University, Department of PathologyTel Aviv, Israel. [Lifshitz, David] Hasharon Hospital, Rabin Medical Center, Petah-Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Department of UrologyTel Aviv, Israel. RP Golan, Sh (reprint author), University of Chicago Medicine, Department of Surgery, 60637 Chicago, USA. EM shaygo1@gmail.com CR Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics, 2012. CA Cancer J Clin 62(1):10–29 Golan S, Nadu A, Lifshitz D, 2015, The role of diagnostic ureteroscopy in the era of computed tomography urography. BMC Urol 15:74 Roupret M et al, 2013, European guidelines on upper tract urothelial carcinomas: 2013 update. Eur Urol 63(6):1059–1071 Cutress ML et al, 2012, Long-term endoscopic management of upper tract urothelial carcinoma: 20-year single-centre experience. BJU Int 110(11):1608–1617 Gadzinski AJ et al, 2010, Long-term outcomes of nephroureterectomy versus endoscopic management for upper tract urothelial carcinoma. J Urol 183(6):2148–2153 Hoffman A et al, 2014, Oncologic results of nephron sparing endoscopic approach for upper tract low grade transitional cell carcinoma in comparison to nephroureterectomy - a case control study. BMC Urol 14:97 Guarnizo E et al, 2000, Ureteroscopic biopsy of upper tract urothelial carcinoma: improved diagnostic accuracy and histopathological considerations using a multi-biopsy approach. J Urol 163(1):52–55 GorinMA et al, 2011, Initial clinical experience with use of ureteral access sheaths in the diagnosis and treatment of upper tract urothelial carcinoma. Urology 78(3):523–527 Wason SE et al, 2012, Ureteroscopic biopsy of upper tract urothelial carcinoma using a novel ureteroscopic biopsy forceps. Can J Urol 19(6):6560–6565 Kleinmann N et al, 2013, Ureteroscopic biopsy of upper tract urothelial carcinoma: comparison of basket and forceps. J Endourol 27(12):1450–1454 Folberg RCJ, Pe'er J Proceedings of the eye pathology course. 1990. Hadassah University hospital, Jerusalem Greene GL, P.D., Fleming ID, AJCC Cancer Staging Manual. Genitourinary sites: renal pelvis and ureter: New York: Springer Verlag Eylert MF et al, 2013, Prognosis is deteriorating for upper tract urothelial cancer: data for England 1985-2010. BJU Int 112(2): E107–E113 Eylert MF, et al., Prognosis is deteriorating for upper tract urothelial cancer: Data for England 1985-2010. BJU Int 112(2): p. E107–13. Raman JD et al, 2011, Incidence and survival of patients with carcinoma of the ureter and renal pelvis in the USA, 1973-2005. BJU Int 107(7):1059–1064 Cutress ML et al, 2012, Ureteroscopic and percutaneous management of upper tract urothelial carcinoma, UTUC): systematic review. BJU Int 110(5):614–628 Grasso M et al, 2012, Ureteroscopic and extirpative treatment of upper urinary tract urothelial carcinoma: a 15-year comprehensive review of 160 consecutive patients. BJU Int 110(11):1618–1626 Cutress ML et al, 2013, Endoscopic versus laparoscopic management of noninvasive upper tract urothelial carcinoma: 20-year single center experience. J Urol 189(6):2054–2060 Simhan J et al, 2014, Nephron-sparing management vs radical nephroureterectomy for low- or moderate-grade, low-stage upper tract urothelial carcinoma. BJU Int 114(2):216–220 Tavora F et al, 2009, Small endoscopic biopsies of the ureter and renal pelvis: pathologic pitfalls. Am J Surg Pathol 33(10): 1540–1546 Vashistha V, Shabsigh A, Zynger DL, 2013, Utility and diagnostic accuracy of ureteroscopic biopsy in upper tract urothelial carcinoma. Arch Pathol Lab Med 137(3):400–407 Rojas CP et al, 2013, Low biopsy volume in ureteroscopy does not affect tumor biopsy grading in upper tract urothelial carcinoma. Urol Oncol 31(8):1696–1700 Kiriyama T, Hironaka H, Fukuda K, 1976, Six years of experience with retrograde biopsy of intraureteral carcinoma using the Dormia stone basket. J Urol 116(3):308–310 Al-Qahtani SM et al, 2014, Can we improve the biopsy quality of upper urinary tract urothelial tumors? Single-center preliminary results of a new biopsy forceps. Urol Int 93(1):34–37 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 89 EP 94 DI 10.1007/s12253-017-0219-1 PG 6 ER PT J AU Uson, LSP Filho, CD Bugano, DGD Geyer, CF de Nigro Corpa, VM Goncalves, DSP Simon, DS Kaliks, AR AF Uson, Luiz Serrano Pedro Filho, Callegaro Donato Bugano, Diniz Gomes Diogo Geyer, Correa Felipe de Nigro Corpa, Vinicius Marcus Goncalves, David Scatena Paulo Simon, Daniel Sergio Kaliks, Aliosha Rafael TI Incidental Findings in Reduction Mammoplasty Specimens in Patients with No Prior History of Breast Cancer. An Analysis of 783 Specimens SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Mammoplasty; Breast reduction surgery; Incidental findings ID Breast cancer; Mammoplasty; Breast reduction surgery; Incidental findings AB Breast reduction surgery is a common procedure and the rate of incidental findings in the removed specimens varies between 0% and 4.6%. There are no guidelines about pathological evaluation of breast reduction surgery. We reviewed all pathology reports of patients undergoing breast reduction surgery in a single tertiary institution in Brazil from January 2008 to August 2014. Exclusion criteria were a personal history of breast cancer, unclear reason for mastectomy and incomplete data on the pathology report. We considered "relevant findings" flat epithelial atypia, atypical hyperplasia, carcinomas in situ and invasive carcinoma. Of 1672 specimens from breast reduction surgery, 783 met inclusion criteria. Median patient age was 40 (8–77), 91% underwent bilateral mastectomy and 57% of the specimens weighted less than 200 g. In 55% of cases, 4 or more paraffin blocks were sampled. There were 40 (5.1%) relevant findings and the most common was atypical lobular hyperplasia (16–2%). There were 3 invasive carcinomas (0.38%). In multivariate analysis, the only variables associated with a higher odds of relevant pathological findings were patient age ≥ 40 (OR 4.73 CI95% 1.98–11.3 p < 0.001) and sampling of ≥4 paraffin blocks from each specimen (OR 6.69 95% CI 2.25–19.9 p < 0.001). The incidence of pre-malignant and malignant lesions in specimens from breast reduction surgery is around 5%, but this risk is significantly higher for patients older than 40 years-old. Sampling at least 4 paraffin blocks from each specimen significantly increases detection rates. C1 [Uson, Luiz Serrano Pedro] Hospital Israelita Albert Einstein, Oncology Department, 627/701 Av. Albert Einstein, 05651-901 Sao Paulo, CEP, Brazil. [Filho, Callegaro Donato] Hospital Israelita Albert Einstein, Oncology Department, 627/701 Av. Albert Einstein, 05651-901 Sao Paulo, CEP, Brazil. [Bugano, Diniz Gomes Diogo] Hospital Israelita Albert Einstein, Oncology Department, 627/701 Av. Albert Einstein, 05651-901 Sao Paulo, CEP, Brazil. [Geyer, Correa Felipe] Hospital Israelita Albert Einstein, Pathology DepartmentSao Paulo, Brazil. [de Nigro Corpa, Vinicius Marcus] Hospital Israelita Albert Einstein, Pathology DepartmentSao Paulo, Brazil. [Goncalves, David Scatena Paulo] Hospital Israelita Albert Einstein, Epidemiology DepartmentSao Paulo, Brazil. [Simon, Daniel Sergio] Hospital Israelita Albert Einstein, Oncology Department, 627/701 Av. Albert Einstein, 05651-901 Sao Paulo, CEP, Brazil. [Kaliks, Aliosha Rafael] Hospital Israelita Albert Einstein, Oncology Department, 627/701 Av. Albert Einstein, 05651-901 Sao Paulo, CEP, Brazil. RP Uson, LSP (reprint author), Hospital Israelita Albert Einstein, Oncology Department, 05651-901 Sao Paulo, Brazil. EM pedroluiz_uson@hotmail.com CR The American Society for Aesthetic Plastic Surgery, Cosmetic Surgery National Data Bank Statistics, 2014, http://www.surgery. org/sites/default/files/2014-Stats.pdf. Accessed 20 March 2015 Kerrigan CL, Collins DE, Striplin D et al, 2001, The health burden of breast hypertrophy. Plast Reconstr Surg 108(6):1591–1599 American Cancer Society, Cancer Facts and Figures, 2015, http:// www.cancer.org/research/cancerfactsstatistics/cancerfactsfigures2015/ index. Accessed 20 March 2015 Tang CL, Brown MH, Levine R et al, 1999, Breast cancer found at the time of breast reduction. Plast Reconstr Surg 103(6):1682–1686 Boice JD Jr, Persson I, Brinton LA et al, 2000, Breast cancer following breast reduction surgery in Sweden. Plast Reconstr Surg 106(4):755–762 Viana GA, Pitanguy I, Torres E, 2005, Histopathological findings in surgical specimens obtained from reduction mammaplasties. Breast 14(3):242–248 IshagMT, Bashinsky DY, Beliaeva IVet al, 2003, Pathologic findings in reduction mammaplasty specimens. Am J Clin Pathol 120(3):377–380 Jansen DA, Murphy M, Kind GM et al, 1998, Breast cancer in reduction mammoplasty: case reports and a survey of plastic surgeons. Plast Reconstr Surg 101(2):361–364 Horo AG, Acker O, Roussel E et al, 2011, Mammoplasty for symmetry in breast reconstruction and histologic assessment. Can J Surg 54(3):201 Ayhan S, Basterzi Y, Yavuzer R et al, 2002, Histologic profiles of breast reduction specimens. Aesthet Plast Surg 26(3):203–205 Kakagia D, Fragia K, Grekou A et al, 2005, Reduction mammaplasty specimens and occult breast carcinomas. Eur J Surg Oncol 31(1):19–21 Samdanci ET, Firat C, Cakir E et al, 2011, The incidence of non-proliferative and precancerous lesions of reduction mammoplasty: evaluation of 273 cases. Eur Rev Med Pharmacol Sci 15(10):1207–1211 Talghini S, 2013, Is macromastia a risk factor for breast cancer? A study on 198 patients. Pak J Biol Sci 16(21):1348–1352 Kyriopoulos E, Kakagia D, Zapandioti P et al, 2012, Pathologic findings in breast reduction specimens: detection of occult premalignant and cancerous lesions. Oncol Res Treat 35(10):583–586 Aytac B, Sahsine T, Erturk FYet al, 2013, Evaluation of incidence and histolopathological findings of breast lesions in reduction mammoplasty specimens: Uludag University experience. J Pak Med Assoc 63(7):878–881 Clark CJ, Whang S, Paige KT, 2009, Incidence of precancerous lesions in breast reduction tissue: a pathologic review of 562 consecutive patients. Plast Reconstr Surg 124(4):1033–1039 Ambaye AB,MacLennan SE, Goodwin AJ et al, 2009, Carcinoma and atypical hyperplasia in reductionmammaplasty: increased sampling leads to increased detection. A prospective study. Plast Reconstr Surg 124(5):1386–1392 Kececi Y, Tasli FA,Yagci A et al, 2014, Histopathologic findings in breast reduction specimens. J Plast Surg Hand Surg 48(2):122–125 Zeger SL, Liang KY, Albert PS, 1988, Models for longitudinal data: a generalized estimating equation approach. Biometrics 44(4):1049–1060 Simpson PT, Reis-Filho JS, Gale Tet al, 2005, Molecular evolution of breast cancer. J Pathol 205(2):248–254 Reis-Filho JS, Lakhani SR, 2003, The diagnosis and management of pre-invasive breast disease: genetic alterations in pre-invasive lesions. Breast Cancer Res Treat 5:313–319 Shackney SE, Silverman JF, 2003)Molecular evolutionary patterns in breast cancer. Adv Anat Pathol 10(5):278–290 Buerger H, Simon R, Schafer KL et al, 2000, Genetic relation of lobular carcinoma in situ, ductal carcinoma in situ, and associated invasive carcinoma of the breast. Mol Pathol 53(3):118–121 London SJ, Connolly JL, Schnitt SJ et al, 1992, A prospective study of benign breast disease and the risk of breast cancer. JAMA 267(7):941–944 DupontWD, Parl FF, Hartmann WHet al, 1993, Breast cancer risk associatedwith proliferative breast disease and atypical hyperplasia. Cancer 71:1258–1258 Dupont WD, Page DL, 1985, Risk factors for breast cancer in women with proliferative breast disease. New Engl J Med 312(3): 146–151 Page DL, Schuyler PA, Dupont WD et al, 2003, Atypical lobular hyperplasia as a unilateral predictor of breast cancer risk: a retrospective cohort study. Lancet 361(9352):125–129 Snyderman RK, Lizardo JG, 1960, Statistical study of malignancies found before, during, or after routine breast plastic operations. Plast Reconstr Surg 25(3):253–256 Freedman BC, Smith SM, Estabrook A et al, 2012, Incidence of occult carcinoma and high-risk lesions in mammaplasty specimens. Int J Breast Cancer 2012:145630 Karabela-Bouropoulou V, Liapi-Avgeri G, Iliopoulou E et al, 1994, Histological findings in breast tissue specimens from reduction mammoplasties. Pathol Res Pract 190(8):792–798 Dotto J, Kluk M, Geramizadeh B et al, 2008, Frequency of clinically occult intraepithelial and invasive neoplasia in reduction mammoplasty specimens: a study of 516 cases. Int J Surg Pathol 16(1):25–30 Desouki MM, Li Z, Hameed O et al, 2013, Incidental atypical proliferative lesions in reduction mammoplasty specimens: analysis of 2498 cases from 2 tertiary women's health centers. Hum Pathol 44(9):1877–1881 Slezak S, Bluebond-Langner R, 2011, Occult carcinoma in 866 reduction mammaplasties: preserving the choice of lumpectomy. Plast Reconstr Surg 127(2):525–530 Keleher AJ, Langstein HN, Ames FC et al, 2003, Breast cancer in reduction mammaplasty specimens: case reports and guidelines. Breast 9(2):120–125 Brinton LA, Persson I, Boice JD Jr et al, 2001, Breast cancer risk in relation to amount of tissue removed during breast reduction operations in Sweden. Cancer 91(3):478–483 Fryzek JP, YeW, Nyren O et al, 2006, A nationwide epidemiologic study of breast cancer incidence following breast reduction surgery in a large cohort of Swedish women. Breast Cancer Res Treat 97(2): 131–134 Goodwin JT, Decroff C, Dauway E et al, 2013, The management of incidental findings of reduction mammoplasty specimens. Can J Plast Surg 21(4):226–228 Cook IS, Fuller CE, 2004, Does histopathological examination of breast reduction specimens affect patient management and clinical follow up? J Clin Pathol 57(3):286–289 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 95 EP 99 DI 10.1007/s12253-017-0230-6 PG 5 ER PT J AU Zou, Yh Li, Xd Zhang, Qh Liu, Dz AF Zou, Yuan-hang Li, Xue-dong Zhang, Qi-hao Liu, De-zhong TI RACK1 Silencing Induces Cell Apoptosis and Inhibits Cell Proliferation in Hepatocellular Carcinoma MHCC97-H Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE RACK1; Hepatocellular carcinoma; Cell proliferation; Cell apoptosis; Gene silencing; MHCC97-H ID RACK1; Hepatocellular carcinoma; Cell proliferation; Cell apoptosis; Gene silencing; MHCC97-H AB This study aimed to explore the effects of RACK1 gene silencing on the apoptosis and proliferation of hepatocellular carcinoma (HCC) MHCC97-H cells. After transfecting MHCC97-H cells with siRNA, RACK1 gene silencing model was established. The cells were divided into blank group, siRNA group and empty plasmid group, respectively. The mRNA and protein expressions of RACK1, cyclin D1 and BAX were determined by qRT-PCR and Western blotting. CCK-8 assay, flow cytometry and FITC-Annexin V/PI staining were used to determine cell viability, cell cycle and cell apoptosis, respectively. The results of qRT-PCR and Western blotting suggested that when compared with the blank group and the empty plasmid group, the mRNA and protein expressions of RACK1 and Cyclin D1 decreased significantly while the mRNA and protein BAX expressions increased substantially in the siRNA group (all P < 0.05). The results of CCK-8 assay revealed that the siRNA group exhibited significantly lower cell viability when compared with the blank group and the empty plasmid group (both P < 0.05); and the cell viability in the siRNA group decreased gradually with the increase of time. The results of flow cytometry and FITC-Annexin V/PI staining indicated that when compared with the blank group and the empty plasmid group, the proportion of cells in S phase was markedly lower and the apoptosis rate was significantly higher in the siRNA group (both P < 0.05). Our study suggests that inhibition of RACK1 could suppress cell proliferation and induce apoptosis in HCC MHCC97-H cells. C1 [Zou, Yuan-hang] The First Affiliated Hospital of Shantou University Medical College, Department of Hepatobiliary Surgery, No. 57, Changping Road, 515041 Shantou, Guangdong Province, China. [Li, Xue-dong] The First Affiliated Hospital of Shantou University Medical College, Department of Orthopedics, No. 57, Changping Road, 515041 Shantou, Guangdong Province, China. [Zhang, Qi-hao] The First Affiliated Hospital of Shantou University Medical College, Department of Orthopedics, No. 57, Changping Road, 515041 Shantou, Guangdong Province, China. [Liu, De-zhong] The First Affiliated Hospital of Shantou University Medical College, Department of Emergency Surgery, No. 57, Changping Road, 515041 Shantou, Guangdong Province, China. RP Liu, Dz (reprint author), The First Affiliated Hospital of Shantou University Medical College, Department of Emergency Surgery, 515041 Shantou, China. EM cowboyzhong@163.com CR Fonseca AL, Cha CH, 2014, Hepatocellular carcinoma: a comprehensive overview of surgical therapy. J Surg Oncol 110(6):712–719 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87–108 Honda T, 2016, Links between human LINE-1 retrotransposons and hepatitis virus-related hepatocellular carcinoma. Front Chem 4:21 El-Serag HB, Rudolph KL, 2007, Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 132(7):2557– 2576 Schlesinger S, Aleksandrova K, Pischon T, Fedirko V, Jenab M, Trepo E, Boffetta P, Dahm CC, Overvad K, Tjonneland A, Halkjaer J, Fagherazzi G, Boutron-Ruault MC, Carbonnel F, Kaaks R, Lukanova A, Boeing H, Trichopoulou A, Bamia C, Lagiou P, Palli D, Grioni S, Panico S, Tumino R, Vineis P, Bueno-de-Mesquita HB, van den Berg S, Peeters PH, Braaten T,Weiderpass E, Quiros JR, Travier N, Sanchez MJ, Navarro C, Barricarte A, Dorronsoro M, Lindkvist B, Regner S, Werner M, Sund M, Khaw KT,Wareham N, Travis RC, Norat T,Wark PA, Riboli E, Nothlings U, 2013, Abdominal obesity, weight gain during adulthood and risk of liver and biliary tract cancer in a European cohort. Int J Cancer 132(3):645–657 Maluccio M, Covey A, 2012, Recent progress in understanding, diagnosing, and treating hepatocellular carcinoma. CA Cancer J Clin 62(6):394–399 Gluer AM, Cocco N, Laurence JM, Johnston ES, Hollands MJ, Pleass HC, Richardson AJ, Lam VW, 2012, Systematic review of actual 10-year survival following resection for hepatocellular carcinoma. HPB, Oxford, 14(5):285–290 Wu J, Meng J, Du Y, Huang Y, Jin Y, Zhang J,Wang B, Zhang Y, Sun M, Tang J, 2013, RACK1 promotes the proliferation, migration and invasion capacity of mouse hepatocellular carcinoma cell line in vitro probably by PI3K/Rac1 signaling pathway. Biomed Pharmacother 67(4):313–319 WangWD,Wen Z, JiW,Ma Y, 2015, RACK1 expression contributes to JNK activity, but JNK activity does not enhance RACK1 expression in hepatocellular carcinoma SMMC-7721 cells. Oncol Lett 9(6):2767–2770 He X, Wang J, Messing EM, Wu G, 2011, Regulation of receptor for activated C kinase 1 protein by the von Hippel-Lindau tumor suppressor in IGF-I-induced renal carcinoma cell invasiveness. Oncogene 30(5):535–547 Li JJ, Xie D, 2015, RACK1, a versatile hub in cancer. Oncogene 34(15):1890–1898 Doan AT, Huttenlocher A, 2007, RACK1 regulates Src activity and modulates paxillin dynamics during cell migration. Exp Cell Res 313(12):2667–2679 Lu F, Zhang C, Wu WJ, Wu YM, 2012, RACK1 downregulation suppresses migration and proliferation of neuroblastoma cell lines. Oncol Rep 27(5):646–1652 Li J, Guo Y, Feng X,Wang Z,Wang Y, Deng P, Zhang D,Wang R, Xie L, Xu X, ZhouY, Ji N, Hu J, Zhou M, Liao G, GengN, Jiang L, Wang Z, Chen Q, 2012, Receptor for activated C kinase 1, RACK1): a regulator for migration and invasion in oral squamous cell carcinoma cells. J Cancer Res Clin Oncol 138(4):563–571 Guo Y,WangW,Wang J, Feng J,Wang Q, Jin J, Lv M, Li X, Li Y, Ma Y, Shen B, Zhang J, 2013, Receptor for activated C kinase 1 promotes hepatocellular carcinoma growth by enhancing mitogenactivated protein kinase kinase 7 activity. Hepatology 57:140–151 McCahill A, Warwicker J, Bolger GB, Houslay MD, Yarwood SJ, 2002, The RACK1 scaffold protein: a dynamic cog in cell response mechanisms. Mol Pharmacol 62(6):1261–1273 Wang F, Osawa T, Tsuchida R, Yuasa Y, Shibuya M, 2011, Downregulation of receptor for activated C-kinase 1, RACK1, suppresses tumor growth by inhibiting tumor cell proliferation and tumor-associated angiogenesis. Cancer Sci 102(11):2007–2013 Rajput S, Kumar BN, Dey KK, Pal I, Parekh A, Mandal M, 2013, Molecular targeting of Akt by thymoquinone promotes G(1, arrest through translation inhibition of cyclin D1 and induces apoptosis in breast cancer cells. Life Sci 93(21):783–790 Peng R, Jiang B, Ma J, Ma Z,Wan X, Liu H, Chen Z, Cheng Q, Chen R, 2013, Forced downregulation of RACK1 inhibits glioma development by suppressing Src/Akt signaling activity. Oncol Rep 30(5): 2195–2202 Mamidipudi V, Miller LD, Mochly-Rosen D, Cartwright CA, 2007, Peptide modulators of Src activity in G1 regulate entry into S phase and proliferation of NIH 3T3 cells. Biochem Biophys Res Commun 352(2):423–430 Lopez-Bergami P, Huang C, Goydos JS, Yip D, Bar-Eli M, Herlyn M, Smalley KS, Mahale A, Eroshkin A, Aaronson S, Ronai Z, 2007, Rewired ERK-JNK signaling pathways in melanoma. Cancer Cell 11(5):447–460 (2015, Inhibition of Notch- and EGFR signaling reduces cell viability and angiogenesis in glioblastoma multiforme. Journal/Cancer Research 75: 1369–1369. Sutton P, Borgia JA, Bonomi P, Plate JM, 2013, Lyn, a Src family kinase, regulates activation of epidermal growth factor receptors in lung adenocarcinoma cells. Mol Cancer 12:76 Wilson JM, Kunnimalaiyaan S, Kunnimalaiyaan M, Gamblin TC, 2015, Inhibition of the AKT pathway in cholangiocarcinoma by MK2206 reduces cellular viability via induction of apoptosis. Cancer Cell Int 15(1):13 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 101 EP 107 DI 10.1007/s12253-017-0214-6 PG 7 ER PT J AU Zhang, J Zhao, B Chen, X Wang, Z Xu, H Huang, B AF Zhang, Jiale Zhao, Bochao Chen, Xiuxiu Wang, Zhenning Xu, Huimian Huang, Baojun TI Silence of Long Noncoding RNA NEAT1 Inhibits Malignant Biological Behaviors and Chemotherapy Resistance in Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Long noncoding RNA; NEAT1; Gastric cancer; Malignant behaviors; Chemotherapy resistance ID Long noncoding RNA; NEAT1; Gastric cancer; Malignant behaviors; Chemotherapy resistance AB Gastric cancer (GC) is the most common solid tumor in digestive system. Nuclear-enriched abundant transcript 1 (NEAT1) gene is a lncRNA, and reveal potential oncogene role in several malignant tumors. The aim of this study is to investigate the expression and clinical significance of Nuclear Paraspeckle Assembly Transcript 1 (NEAT1) gene and its influence to malignant biologic behaviors and chemotherapy resistance to adriamycin in GC. This study found NEAT1 was up-regulated in GC tissues and cells, especially in in GC adriamycin-resistant cells. NEAT1 silence in SGC7901 cells could inhibit proliferation and invasion ability, and promote cell apoptosis significantly. NEAT1 silence in adriamycin-resistant SGC7901/ADR cells also depressed the half maximal inhibitory concentration (IC50) for adriamycin, chemotherapy resistance to adriamycin was inhibited significantly. NEAT1 knockdown promoted apoptosis in SGC7901/ ADR cells induced by adriamycin. In summary, lncRNA NEAT1 is high-expressed in GC and functions as an oncogene to modulate apoptosis, invasion, proliferation and chemotherapy resistance of GC cells, which might be a novel potential therapeutic target for GC. C1 [Zhang, Jiale] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, No.155 Nanjing North Street, Heping District, 110001 Shenyang, Liaoning, China. [Zhao, Bochao] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, No.155 Nanjing North Street, Heping District, 110001 Shenyang, Liaoning, China. [Chen, Xiuxiu] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, No.155 Nanjing North Street, Heping District, 110001 Shenyang, Liaoning, China. [Wang, Zhenning] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, No.155 Nanjing North Street, Heping District, 110001 Shenyang, Liaoning, China. [Xu, Huimian] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, No.155 Nanjing North Street, Heping District, 110001 Shenyang, Liaoning, China. [Huang, Baojun] China Medical University, Affiliated First Hospital, Department of Surgical Oncology, No.155 Nanjing North Street, Heping District, 110001 Shenyang, Liaoning, China. RP Huang, B (reprint author), China Medical University, Affiliated First Hospital, Department of Surgical Oncology, 110001 Shenyang, China. EM huang_bj@163.com CR Sugano K, 2015, Screening of gastric cancer in Asia. Best Pract Res Clin Gastroenterol 29(6):895–905 Newton AD, Datta J, Loaiza-Bonilla A, Karakousis GC, Roses RE, 2015, Neoadjuvant therapy for gastric cancer: current evidence and future directions. J Gastrointest Oncol 6(5):534–543 Jiang L, Wang W, Li G, Sun C, Ren Z, Sheng H, Gao H, Wang C, Yu H, 2016, High TUG1 expression is associated with chemotherapy resistance and poor prognosis in esophageal squamous cell carcinoma. Cancer Chemother Pharmacol 78(2):333–339 Shang C, Guo Y, Hong Y, Xue YX, 2016, Long non-coding RNA TUSC7, a target of miR-23b, plays tumor-suppressing roles in human Gliomas. Front Cell Neurosci 10:235 Zhang J, Yao T,Wang Y, Yu J, Liu Y, Lin Z, 2016, Long noncoding RNA MEG3 is downregulated in cervical cancerand affects cell proliferation and apoptosis by regulating miR-21. Cancer Biol Ther 17(1):104–113 Chen J, Hu L, Wang J, Zhang F, Chen J, Xu G, Wang Y, Pan Q, 2017, Low expression LncRNA TUBA4B is a poor predictor of prognosis and regulates cell proliferation in non-small cell lung cancer. Pathol Oncol Res 23(2):265–270 Wang X, Li M, Wang Z, Han S, Tang X, Ge Y, Zhou L, Zhou C, Yuan Q, Yang M, 2015, Silencing of long noncoding RNA MALAT1 by miR-101 and miR-217 inhibits proliferation, migration and invasion of esophageal squamous cell carcinoma cells. J Biol Chem 290(7):3925–3935 XianGuo C, ZongYao H, Jun Z, Song F, GuangYue L, LiGang Z, KaiPing Z, YangYang Z, ChaoZhao L, 2016, Promoting progression and clinicopathological significance of NEAT1 over-expression in bladder cancer. Oncotarget., DOI 10.18632/oncotarget.10084 Sun C, Li S, Zhang F, Xi Y, Wang L, Bi Y, Li D, 2016, Long noncoding RNA NEAT1 promotes non-small cell lung cancer progression through regulation of miR-377-3p-E2F3 pathway. Oncotarget., DOI 10.18632/oncotarget.10108 Chen X, Kong J, Ma Z, Gao S, Feng X, 2015, Up regulation of the long non-coding RNA NEAT1 promotes esophageal squamous cell carcinoma cell progression and correlates with poor prognosis. Am J Cancer Res 5(9):2808–2815 Fu JW, Kong Y, Sun X, 2016, Long noncoding RNA NEAT1 is an unfavorable prognostic factor and regulates migration and invasion in gastric cancer. J Cancer Res Clin Oncol 142(7):1571–1579 Ma Y, Liu L, Yan F, Wei W, Deng J, Sun J, 2016, Enhanced expression of long non-coding RNA NEAT1 is associated with the progression of gastric adenocarcinomas. World J Surg Oncol 14(1):41 Shang C, Guo Y, Zhang J, Huang B, 2016, Silence of long noncoding RNA UCA1 inhibits malignant proliferation and chemotherapy resistance to adriamycin in gastric cancer. Cancer Chemother Pharmacol 77(5):1061–1067 Shang C, Hong Y, Guo Y, Liu YH, Xue YX, 2016, miR-128 regulates the apoptosis and proliferation of glioma cells by targeting RhoE. Oncol Lett 11(1):904–908 Wang P, Liu YH, Yao YL, Li Z, Li ZQ, Ma J, Xue YX, 2015, Long non-coding RNA CASC2 suppresses malignancy in human gliomas by miR-21. Cell Signal 27(2):275–282 Zhen L, Yun-Hui L, Hong-Yu D, Jun M, Yi-Long Y, 2016, Long noncoding RNA NEAT1 promotes glioma pathogenesis by regulating miR-449b-5p/c-met axis. Tumour Biol 37(1):673–683 Wang P,Wu T, Zhou H, Jin Q, He G, Yu H, Xuan L,Wang X, Tian L, Sun Y, Liu M, Qu L, 2016, Long noncoding RNA NEAT1 promotes laryngeal squamous cellcancer through regulating miR- 107/CDK6 pathway. J Exp Clin Cancer Res 35:22 Adriaens C, Standaert L, Barra J, Latil M, Verfaillie A, Kalev P, Boeckx B, Wijnhoven PW, Radaelli E, Vermi W, Leucci E, Lapouge G, Beck B, van den Oord J, Nakagawa S, Hirose T, Sablina AA, Lambrechts D, Aerts S, Blanpain C, Marine JC, 2016, p53 induces formation of NEAT1 lncRNA-containing paraspeckles that modulate replication stress response and chemosensitivity. Nat Med 22(8):861–868 Gong W, Zheng J, Liu X, Ma J, Liu Y, Xue Y, 2016, Knockdown of NEAT1 restrained the malignant progression of glioma stem cells by activating microRNA let-7e. Oncotarget., DOI 10.18632/oncotarget.11403 Jiang P, Wu X, Wang X, Huang W, Feng Q, 2016, NEAT1 upregulates EGCG-induced CTR1 to enhance cisplatin sensitivity in lung cancer cells. Oncotarget., DOI 10.18632/oncotarget.9712 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 109 EP 113 DI 10.1007/s12253-017-0233-3 PG 5 ER PT J AU Acs, B Madaras, L Kovacs, AK Micsik, T Tokes, AM Gyorffy, B Kulka, J Szasz, MA AF Acs, Balazs Madaras, Lilla Kovacs, Attila Kristof Micsik, Tamas Tokes, Anna-Maria Gyorffy, Balazs Kulka, Janina Szasz, Marcell Attila TI Reproducibility and Prognostic Potential of Ki-67 Proliferation Index when Comparing Digital-Image Analysis with Standard Semi-Quantitative Evaluation in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ki-67 proliferation index; Breast cancer; Digital image analysis; Intra-class correlation; Concordance correlation; Prognosis ID Ki-67 proliferation index; Breast cancer; Digital image analysis; Intra-class correlation; Concordance correlation; Prognosis AB In this study, the reproducibility of Ki-67 proliferation index (KIPI) was investigated by comparing the semiquantitative (SQ) results of three assessors with those of digital image-analysis (DIA) methods. The prognostic significance of the two approaches was also correlated with clinical outcome. Tissue microarrays of duplicate 2 mm cores were constructed from representative areas of formalin-fixed and paraffin-embedded tumor blocks of 347 breast cancer patients. SQ evaluation of Ki-67 (MIB1 clone) immunostained slides was performed independently by three pathologists. DIA was completed using a fully automated histological pattern and cell recognition module for KIPI detection (DIA-1) and an adjustable module (DIA-2) with the possibility of manual corrections. To compare SQ and DIA evaluations intra-class correlation (ICC) and concordance correlation coefficients (CCC) were determined. The three SQ evaluations demonstrated a remarkable ICC (0.853). Significant difference and poor concordance occurred between SQ-1 and SQ-2 as well as between SQ-1 and SQ-3 (p ≤ 0.001, CCC ≤ 0.827 for both comparisons). Thus, the reference KIPI value (SQ-RV) was generated from the mean values of SQ-2 and SQ-3. SQ-RV and DIA-2 results showed substantial concordance (CCC = 0.963, at p = 0.754), while SQ-RV and DIA-1 values differed (p ≤ 0.001) at only moderate concordance (CCC = 0.906). In multivariate analysis, lymph node status and SQ-2 assessment were significantly associated with clinical outcome (p ≤ 0.012 for both comparisons). Our results confirm that KIPI is a significant prognostic marker in breast cancer, which can be can be reliably reproduced by using an adjustable DIA-2 image analysis module. C1 [Acs, Balazs] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Madaras, Lilla] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Kovacs, Attila Kristof] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Micsik, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 26 Ulloi ut, 1085 Budapest, Hungary. [Tokes, Anna-Maria] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Gyorffy, Balazs] MTA TTK, Lendulet Cancer Biomarker Research Group, 2 Magyar tudosok korutja, 1117 Budapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. [Szasz, Marcell Attila] Semmelweis University, 2nd Department of Pathology, 93 Ulloi ut, 1091 Budapest, Hungary. RP Szasz, MA (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM cac@korb2.sote.hu CR Senkus E, Kyriakides S, Penault-Llorca F, Poortmans P, Thompson A, Zackrisson S, Cardoso F, 2013, Primary breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 24(Suppl 6):vi7–v23., DOI 10.1093/annonc/mdt284 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144(5):646–674., DOI 10.1016/j.cell.2011.02.013 Gerdes J, Schwab U, Lemke H, Stein H, 1983, Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer 31(1):13–20 Coates AS,Winer EP, Goldhirsch A, Gelber RD, GnantM, Piccart- Gebhart M, Thurlimann B, Senn HJ, 2015, Tailoring therapiesimproving the management of early breast cancer: St Gallen international expert Consensus on the Primary therapy of early breast cancer 2015. Ann Oncol 26(8):1533–1546., DOI 10.1093/annonc/ mdv221 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, Senn HJ, 2013, Personalizing the treatment of women with early breast cancer: highlights of the St Gallen international expert Consensus on the Primary therapy of early breast cancer 2013. Ann Oncol 24(9):2206–2223., DOI 10.1093/annonc/ mdt303 Harris L, Fritsche H, Mennel R, Norton L, Ravdin P, Taube S, Somerfield MR, Hayes DF, Bast RC Jr, American Society of Clinical Oncology, 2007, American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J Clin Oncol 25(33):5287–5312., DOI 10.1200/JCO. 2007.14.2364 Dowsett M, Nielsen TO, A’Hern R, Bartlett J, Coombes RC, Cuzick J, Ellis M, Henry NL, Hugh JC, Lively T, McShane L, Paik S, Penault-Llorca F, Prudkin L, Regan M, Salter J, Sotiriou C, Smith IE, Viale G, Zujewski JA, Hayes DF, 2011, Assessment of Ki67 in breast cancer: recommendations from the international Ki67 in breast cancer working group. J Natl Cancer Inst 103(22): 1656–1664., DOI 10.1093/jnci/djr393 Polley MY, Leung SC, Gao D, Mastropasqua MG, Zabaglo LA, Bartlett JM, McShane LM, Enos RA, Badve SS, Bane AL, Borgquist S, Fineberg S, Lin MG, Gown AM, Grabau D, Gutierrez C, Hugh JC, Moriya T, Ohi Y, Osborne CK, Penault- Llorca FM, Piper T, Porter PL, Sakatani T, Salgado R, Starczynski J, Laenkholm AV, Viale G, Dowsett M, Hayes DF, Nielsen TO, 2015, An international study to increase concordance in Ki67 scoring. Mod Pathol 28(6):778–786., DOI 10.1038/ modpathol.2015.38 Polley MY, Leung SC, McShane LM, Gao D, Hugh JC, Mastropasqua MG, Viale G, Zabaglo LA, Penault-Llorca F, Bartlett JM, Gown AM, Symmans WF, Piper T, Mehl E, Enos RA, Hayes DF, Dowsett M, Nielsen TO, 2013, An international Ki67 reproducibility study. J Natl Cancer Inst 105(24):1897–1906., DOI 10.1093/jnci/djt306 Luporsi E, Andre F, Spyratos F, Martin PM, Jacquemier J, Penault- Llorca F, Tubiana-Mathieu N, Sigal-Zafrani B, Arnould L, Gompel A, Egele C, Poulet B, Clough KB, Crouet H, Fourquet A, Lefranc JP, Mathelin C, Rouyer N, Serin D, Spielmann M, Haugh M, Chenard MP, Brain E, de Cremoux P, Bellocq JP, 2012, Ki-67: level of evidence and methodological considerations for its role in the clinical management of breast cancer: analytical and critical review. Breast Cancer Res Treat 132(3):895–915., DOI 10.1007/ s10549-011-1837-z Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA, 2010, Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol 11(2):174–183., DOI 10.1016/s1470-2045(09, 70262-1 Soenksen D, 2009, Digital pathology at the crossroads of major health care trends: corporate innovation as an engine for change. Arch Pathol Lab Med 133(4):555–559., DOI 10.1043/1543-2165- 133.4.555 Kayser K, Borkenfeld S, Kayser G, 2012, How to introduce virtual microscopy, VM, in routine diagnostic pathology: constraints, ideas, and solutions. Anal Cell Pathol, Amst, 35(1):3–10., DOI 10. 3233/acp-2011-0044 Kayser K, Gortler J, Borkenfeld S, Kayser G, 2011, How to measure diagnosis-associated information in virtual slides. Diagn Pathol 6(Suppl 1):S9., DOI 10.1186/1746-1596-6-s1-s9 Laurinavicius A, Plancoulaine B, Laurinaviciene A, Herlin P, Meskauskas R, Baltrusaityte I, Besusparis J, Dasevicius D, Elie N, Iqbal Y, Bor C, 2014, A methodology to ensure and improve accuracy of Ki67 labelling index estimation by automated digital image analysis in breast cancer tissue. Breast Cancer Res 16(2): R35., DOI 10.1186/bcr3639 Riber-Hansen R, Vainer B, Steiniche T, 2012, Digital image analysis: a review of reproducibility, stability and basic requirements for optimal results. APMIS 120(4):276–289., DOI 10.1111/j.1600-0463. 2011.02854.x Ruifrok AC, Johnston DA, 2001, Quantification of histochemical staining by color deconvolution. Anal Quant Cytol Histol 23(4): 291–299 Altman DG, 1990, Practical statistics for medical research. Chapman and Hall, London McBride GB, 2005, A proposal for strength-of-agreement criteria for Lin’s Concordance Correlation Coefficient. NIWA Client Report: HAM2005–062 Kontzoglou K, Palla V, Karaolanis G, Karaiskos I, Alexiou I, Pateras I, Konstantoudakis K, Stamatakos M, 2013, Correlation between Ki67 and breast cancer prognosis. Oncology 84(4):219– 225., DOI 10.1159/000346475 Mikami Y, Ueno T, Yoshimura K, Tsuda H, Kurosumi M, Masuda S, Horii R, Toi M, Sasano H, 2013, Interobserver concordance of Ki67 labeling index in breast cancer: Japan breast cancer research group Ki67 ring study. Cancer Sci 104(11):1539–1543., DOI 10. 1111/cas.12245 Varga Z, Cassoly E, Li Q, Oehlschlegel C, Tapia C, Lehr HA, Klingbiel D, Thurlimann B, Ruhstaller T, 2015, Standardization for Ki-67 assessment in moderately differentiated breast cancer. A retrospective analysis of the SAKK 28/12 study. PLoS One 10(4): e0123435., DOI 10.1371/journal.pone.0123435 Cserni G, Voros A, Liepniece-Karele I, Bianchi S, Vezzosi V, Grabau D, Sapino A, Castellano I, Regitnig P, Foschini MP, Zolota V, Varga Z, Figueiredo P, Decker T, Focke C, Kulka J, Kaya H, Reiner-Concin A, Amendoeira I, Callagy G, Caffrey E, Wesseling J, Wells C, 2014, Distribution pattern of the Ki67 labelling index in breast cancer and its implications for choosing cut-off values. Breast 23(3):259–263., DOI 10.1016/j.breast.2014. 02.003 Voros A, Csorgo E, Kovari B, Lazar P, Kelemen G, Rusz O, Nyari T, Cserni G, 2015, Different methods of pretreatment Ki-67 labeling index evaluation in core biopsies of breast cancer patients treated with neoadjuvant chemotherapy and their relation to response to therapy. Pathol Oncol Res 21(1):147–155., DOI 10.1007/s12253- 014-9800-z Maeda I, Abe K, Koizumi H, Nakajima C, Tajima S, Aoki H, Tsuchiya J, Tsuchiya S, Tsuchiya K, Shimo A, Tsugawa K, Ueno T, Tatsunami S, TakagiM, 2015, Comparison between Ki67 labeling index determined using image analysis software with virtual slide system and that determined visually in breast cancer. Breast Cancer., DOI 10.1007/s12282-015-0634-7 Klauschen F, Wienert S, Schmitt WD, Loibl S, Gerber B, Blohmer JU, Huober J, Rudiger T, Erbstosser E, Mehta K, Lederer B, Dietel M, Denkert C, von Minckwitz G, 2015, Standardized Ki67 diagnostics using automated scoring-clinical validation in the GeparTrio breast cancer study. Clin Cancer Res 21(16):3651–3657., DOI 10. 1158/1078-0432.ccr-14-1283 Denkert C, Budczies J, von Minckwitz G, Wienert S, Loibl S, Klauschen F, 2015, Strategies for developing Ki67 as a useful biomarker in breast cancer. Breast., DOI 10.1016/j.breast.2015.07. 017 Joshi S,Watkins J, Gazinska P, Brown JP, Gillett CE, Grigoriadis A, Pinder SE, 2015, Digital imaging in the immunohistochemical evaluation of the proliferation markers Ki67, MCM2 and Geminin, in early breast cancer, and their putative prognostic value. BMC Cancer 15:546., DOI 10.1186/s12885-015-1531-3 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 115 EP 127 DI 10.1007/s12253-017-0220-8 PG 13 ER PT J AU Rong, L Huang, W Tian, Sh Chi, X Zhao, P Liu, F AF Rong, Li Huang, Wei Tian, Shangkun Chi, Xiangbo Zhao, Pan Liu, Fengfeng TI COL1A2 is a Novel Biomarker to Improve Clinical Prediction in Human Gastric Cancer: Integrating Bioinformatics and Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gastric cancer; biomarker; bioinformatics; COL1A2 ID gastric cancer; biomarker; bioinformatics; COL1A2 AB Gastric cancer is the third most common cause of cancer-related death in worldwide. It is crucial to target the key genes controlling pathogenesis in the early stage of gastric cancer. This study describes an integrated bioinformatics to identify molecular biomarkers for gastric cancer in patients’ cancer tissues. We reports differently expression genes in large gastric cancer cohorts from Gene Expression Ominus (GEO). Our findings revealed that 433 genes were significantly different expressed in human gastric cancer. Differently expression gene profile in gastric cancer was further validated by bioinformatic analyses, co-expression network construction. Based on the co-expression network and top-ranked genes, we identified collagen type I alpha 2 (COL1A2) which encodes the pro-alpha2 chain of type I collagen whose triple helix comprises two alpha1 chains and one alpha2 chain, was the key gene in a 37-gene network that modulates cell motility by interacting with the cytoskeleton. Furthermore, the prognostic role of COL1A2 was determined by use of immunohistochemistry on human gastric cancer tissue. COL1A2 was highly expressed in human gastric cancer as compared with normal gastric tissues. Statistical analysis showed COL1A2 expression level was significantly associated with histological type and lymph node status. However, there were no correlations between COL1A2 expression and age, lymph node numbers, tumor size, or clinical stage. In conclusion, the novel bioinformatics used in this study has led to identification of improving diagnostic biomarkers for human gastric cancer and could benefit further analyses of the key alteration during its progression. C1 [Rong, Li] Chongqing Infectious Disease Medical Center, Department of gastroenterology, 1#Huangjuewan, Xiaolongkan, Shapingba District, 400030 Chongqing, China. [Huang, Wei] Southwest University of Political Science and Law, Health Center, 401120 Chongqing, China. [Tian, Shangkun] Chongqing Infectious Disease Medical Center, Department of gastroenterology, 1#Huangjuewan, Xiaolongkan, Shapingba District, 400030 Chongqing, China. [Chi, Xiangbo] Chongqing Infectious Disease Medical Center, Department of gastroenterology, 1#Huangjuewan, Xiaolongkan, Shapingba District, 400030 Chongqing, China. [Zhao, Pan] Chongqing Infectious Disease Medical Center, Department of gastroenterology, 1#Huangjuewan, Xiaolongkan, Shapingba District, 400030 Chongqing, China. [Liu, Fengfeng] Chongqing Infectious Disease Medical Center, Department of gastroenterology, 1#Huangjuewan, Xiaolongkan, Shapingba District, 400030 Chongqing, China. RP Rong, L (reprint author), Chongqing Infectious Disease Medical Center, Department of gastroenterology, 400030 Chongqing, China. EM lirongcq@126.com CR Orditura M, Galizia G, Sforza V, Gambardella V, Fabozzi A, Laterza MM, Andreozzi F, Ventriglia J, Savastano B, Mabilia A, Lieto E, Ciardiello F, De Vita F, 2014, Treatment of gastric cancer. World J Gastroenterol 20:1635–1649 Lee YY, Derakhshan MH, 2013, Environmental and lifestyle risk factors of gastric cancer. Arch Iran Med 16:358–365 Chandanos E, Lagergren J, 2008, Oestrogen and the enigmatic male predominance of gastric cancer. Eur J Cancer 44:2397–2403 Qin J, Liu M, Ding Q, Ji X, Hao Y, Wu X, Xiong J, 2014, The direct effect of estrogen on cell viability and apoptosis in human gastric cancer cells. Mol Cell Biochem 395:99–107 Gonzalez CA, Sala N, Rokkas T, 2013, Gastric cancer: epidemiologic aspects. Helicobacter 18(Suppl 1):34–38 HatakeyamaM, Higashi H, 2005, Helicobacter pylori CagA: a new paradigm for bacterial carcinogenesis. Cancer Sci 96:835–843 Thrumurthy SG, Chaudry MA, Hochhauser D, Mughal M, 2013, The diagnosis and management of gastric cancer. BMJ 347:f6367 Chan TH, Qamra A, Tan KT, Guo J, Yang H, Qi L, Lin JS, Ng VH, Song Y, Hong H et al: ADARMediated RNA Editing Predicts Progression and Prognosis of Gastric Cancer. Gastroenterology 2016; 151(4):637–650 e610 Kataoka K, Shiraishi Y, TakedaY, Sakata S,Matsumoto M,Nagano S, Maeda T, Nagata Y, Kitanaka A, Mizuno S, Tanaka H, Chiba K, Ito S, Watatani Y, Kakiuchi N, Suzuki H, Yoshizato T, Yoshida K, Sanada M, Itonaga H, Imaizumi Y, Totoki Y, Munakata W, Nakamura H, Hama N, Shide K, Kubuki Y, Hidaka T, Kameda T, Masuda K, Minato N, Kashiwase K, Izutsu K, Takaori-Kondo A, Miyazaki Y, Takahashi S, Shibata T, Kawamoto H, Akatsuka Y, Shimoda K, Takeuchi K, Seya T, Miyano S and Ogawa S. Aberrant PD-L1 expression through 3′-UTR disruption in multiple cancers. Nature 2016; 534: 402–406. Burki TK: Progression-free survival with regorafenib in gastric cancer. The Lancet Oncology 2016; 17(8):e323 Venturi S, Donati FM, Venturi A, Venturi M, 2000, Environmental iodine deficiency: a challenge to the evolution of terrestrial life? Thyroid 10:727–729 Jakszyn P, Gonzalez CA, 2006, Nitrosamine and related food intake and gastric and oesophageal cancer risk: a systematic review of the epidemiological evidence.World J Gastroenterol 12:4296–4303 Barker N, HuchM, Kujala P, van deWetering M, Snippert HJ, van Es JH, Sato T, Stange DE, Begthel H, van den BornM, Danenberg E, van den Brink S, Korving J, Abo A, Peters PJ, Wright N, Poulsom R, Clevers H, 2010, Lgr5(+ve, stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 6:25–36 Ohtsu A, Ajani JA, Bai YX, Bang YJ, Chung HC, Pan HM, Sahmoud T, Shen L, Yeh KH, Chin K, Muro K, Kim YH, Ferry D, Tebbutt NC, Al-Batran SE, Smith H, Costantini C, Rizvi S, Lebwohl D, Van Cutsem E, 2013, Everolimus for previously treated advanced gastric cancer: results of the randomized, doubleblind, phase III GRANITE-1 study. J Clin Oncol 31:3935–3943 Wang K, Yuen ST, Xu J, Lee SP, Yan HH, Shi ST, Siu HC, Deng S, Chu KM, Law S, Chan KH, Chan AS, Tsui WY, Ho SL, Chan AK, Man JL, Foglizzo V, Ng MK, Chan AS, Ching YP, Cheng GH, Xie T, Fernandez J, Li VS, Clevers H, Rejto PA, Mao M, Leung SY, 2014, Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet 46:573–582 Ramucirumab extends gastric cancer survival. Cancer Discov 2014; 4: OF3. Garay J, PiazueloMB,Majumdar S, Li L, Trillo-Tinoco J, Del Valle L, Schneider BG, Delgado AG, Wilson KT, Correa P et al: The homing receptor CD44 is involved in the progression of precancerous gastric lesions in patients infected with Helicobacter pylori and in development of mucous metaplasia in mice. Cancer letters 2016; 371(1):90–98 Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, Liu J, Yue YG, Wang J, Yu K, Ye XS, Do IG, Liu S, Gong L, Fu J, Jin JG, Choi MG, Sohn TS, Lee JH, Bae JM, Kim ST, Park SH, Sohn I, Jung SH, Tan P, Chen R, Hardwick J, Kang WK, Ayers M, Hongyue D, Reinhard C, Loboda A, Kim S, Aggarwal A, 2015, Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med 21:449–456 Schlaermann P, Toelle B, Berger H, Schmidt SC, Glanemann M, Ordemann J, Bartfeld S,Mollenkopf HJ,Meyer TF, 2016, A novel human gastric primary cell culture system for modelling helicobacter pylori infection in vitro. Gut 65:202–213 Tabata S, Ikeda R, Yamamoto M, Shimaoka S, Mukaida N, Takeda Y, Yamada K, Soga T, Furukawa T, Akiyama S, 2014, Thymidine phosphorylase activates NFκB and stimulates the expression of angiogenic and metastatic factors in human cancer cells. Oncotarget 5:10473–10485 Kim SY, Park C, Kim HJ, Park J, Hwang J, Kim JI, ChoiMG, Kim S, Kim KM, Kang MS, 2015, Deregulation of immune response genes in patients with Epstein-Barr virus-associated gastric cancer and outcomes. Gastroenterology 148:137–147 e139 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 129 EP 134 DI 10.1007/s12253-017-0223-5 PG 6 ER PT J AU Brzozowska, A Powrozek, T Homa-Mlak, I Mlak, R Ciesielka, M Golebiowski, P Malecka-Massalska, T AF Brzozowska, Anna Powrozek, Tomasz Homa-Mlak, Iwona Mlak, Radoslaw Ciesielka, Marzanna Golebiowski, Pawel Malecka-Massalska, Teresa TI Polymorphism of Promoter Region of TNFRSF1A Gene (−610 T > G) as a Novel Predictive Factor for Radiotherapy Induced Oral Mucositis in HNC Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral mucositis; Radiotherapy; Head and neck cancer; Polymorphism; TNFRSF1A ID Oral mucositis; Radiotherapy; Head and neck cancer; Polymorphism; TNFRSF1A AB Every year, about 650 thousand new cases of Head and Neck Cancer (HNC) are diagnosed globally. Apart from surgery, radiotherapy (RTH), chemotherapy (CHT) or its combination is used in the treatment of HNC. One of the most frequent complications and, at the same time, limitations of RTH is oral mucositis (OM). Proinflammatory cytokines (including TNF-α) play a key role in the development of OM. Genetic alterations, i.e. single nucleotide polymorphisms (SNPs) within genes encoding for receptors for TNF (ie. TNFRSF1A) may change their function. The aim of this study was to investigate relationship between a polymorphism of TNFRSF1A and occurrence and severity of acute reaction after RTH for HNC patients. Data from 58 HNC patients (stages I-IV) were analyzed. All of them were irradiated using IMRT technique with doses 50-70Gy. Oral mucositis (OM)was evaluated according to RTOG/EORTC guidelines. DNA from HNC patients were isolated from whole blood and genotypes were determined by sequencing method. Patients with TT or GT genotype demonstrated higher risk of manifestation of grade 3 OM in 5th week of RTH (p=0.041; OR=9.240; 95% CI: 1.101–77.581) compared to GG carriers. Similarly, high risk of grade 3 OM in patients with T allele presence was noted in 6th week (p=0.030; OR=10.50; 95%CI:1.257–87.690) and in 7th week (p=0.008; OR=5.625; 95% CI: 1.584–19.975) of treatment compared to patients with GG homozygote. Our results indicate an association between SNP of TNFRSF1A (rs4149570) gene and risk of more severe OM related to radiation therapy for HNC patients. C1 [Brzozowska, Anna] Medical University of Lublin, Department of Oncology, Jaczewskiego 7, 20-090 Lublin, Poland. [Powrozek, Tomasz] Medical University of Lublin, Department of Human Physiology, Radziwillowska 11, 20-080 Lublin, Poland. [Homa-Mlak, Iwona] Medical University of Lublin, Department of Human Physiology, Radziwillowska 11, 20-080 Lublin, Poland. [Mlak, Radoslaw] Medical University of Lublin, Department of Human Physiology, Radziwillowska 11, 20-080 Lublin, Poland. [Ciesielka, Marzanna] Medical University of Lublin, Department of Forensic Medicine, Jaczewskiego 8, 20-090 Lublin, Poland. [Golebiowski, Pawel] Medical University of Lublin, Department of Oncology, Jaczewskiego 7, 20-090 Lublin, Poland. [Malecka-Massalska, Teresa] Medical University of Lublin, Department of Human Physiology, Radziwillowska 11, 20-080 Lublin, Poland. RP Brzozowska, A (reprint author), Medical University of Lublin, Department of Oncology, 20-090 Lublin, Poland. EM annabrzo@poczta.onet.pl CR Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics 2016. CA Cancer J Clin 66(1):7–30 Trotti A, Bellm LA, Epstein JB, Frame D, Fuchs HJ, Gwede CK et al, 2003, Mucositis incidence, severity and associated outcomes in patients with head and neck cancer receiving radiotherapy with or without chemotherapy: a systematic literature review. Radiother Oncol 66:253–262 Knox JJ, Puodziunas AL, Feld R, 2000, Chemotherapy-induced oral mucositis: prevention and management. Drugs Aging 17(4): 257–267 Cox JD, Stetz J, Pajak TF, 1995, Toxicity criteria of the radiation therapy Oncology group, RTOG, and the European Organization for Research and Treatment of cancer, EORTC). Int J Radiat Oncol Biol Phys 31:1341–1346 Sonis ST, Elting LS, Keefe D, Peterson DE, Schubert M, Hauer-Jensen Met al, 2004, Perspectives on cancer therapy-induced mucosal injury: pathogenesis, measurement, epidemiology, and consequences for patients. Cancer 100:1995–2025., DOI 10.1002/cncr.20162 Platek ME, McCloskey SA, Cruz M, BurkeMS, ReidME,Wilding GE et al, 2013, Quantification of the effect of treatment duration on local-regional failure after definitive concurrent chemotherapy and intensity-modulated radiation therapy for squamous cell carcinoma of the head and neck. Head Neck 35(5):684–688 Vera-Llonch M, Oster G, Hagiwara M, Sonis S, 2006, Oral mucositis in patients undergoing radiation treatment for head and neck carcinoma. Cancer 106(2):329–336 Venkatesh GH, Manjunath VB, Mumbrekar KD, Negi H, Fernandes DJ, Sharan K et al, 2014, Polymorphisms in radioresponsive genes and its association with acute toxicity among head and neck cancer patients. PLoS One 9(3):e89079., DOI 10. 1371/journal.pone.0089079 Pratesi N, Mangoni M, Mancini I, Paiar F, Simi L, Livi L et al, 2011, Association between single nucleotide polymorphisms in the XRCC1 and RAD51 genes and clinical radiosensitivity in head and neck cancer. Radiother Oncol 99:356–361 Werbrouck J, De Ruyck K, Duprez F, Veldeman L, Claes K, Van Eijkeren M et al, 2009, Acute normal tissue reactions in head-andneck cancer patient streated with IMRT: influence of dose and association with genetic polymorphisms in DNA DSB repair genes. J Radiot Oncol Biol Phys 73:1187–1195 Sonis ST, 2002, The biologic role of nuclear factor-kB in disease and its potential involvement in mucosal injury associated with antineoplastic therapy. Crit Rev Oral Biol Med 13:300–309 Kalliolias GD, Ivashkiv LB, 2016, TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol 12(1):49–62 Sonis S, Haddad R, Posner M, Watkins B, Fey E, Morgan TVet al, 2007, Gene expression changes in peripheral blood cells provide insight into the biological mechanisms associated with regimenrelated toxicities in patients being treated for head and neck cancers. Oral Oncol 43:289–300 Akmansu M, Unsal D, Bora H, Elbeg S, 2005, Influence of locoregional radiation treatment on tumor necrosis factor-alpha and interleukin-6 in the serum of patients with head and neck cancer. Cytokine 31:41–45 Xanthinaki A, Nicolatou-Galitis O, Athanassiadou P, Gonidi M, Kouloulias V, Sotiropoulou-Lontou A et al, 2008, Apoptotic and inflammation markers in oral mucositis in head and neck cancer patients receiving radiotherapy: preliminary report. Support Care Cancer 16:1025–1033 Haddad R, Sonis S, Posner M, Wirth L, Costello R, Braschayko P et al, 2009, Randomized phase 2 study of concomitant Chemoradiotherapy using weekly carboplatin/paclitaxel with or without daily subcutaneous Amifostine in patients with locally advanced head and neck cancer. Cancer 115:4514–4523 Meirovitz A, Kuten M, Billan S, Abdah-Bortnyak R, Sharon A, Peretz T et al, 2010, Cytokines levels, severity of acute mucositis and the need of PEG tube installation during chemo-radiation for head and neck cancer - a prospective pilot study. Radiat Oncol 5:16 Keefe DM, SchubertMM, Elting LS, Sonis ST, Epstein JB, Raber- Durlacher JE et al, 2007, Updated clinical practice guidelines for the prevention and treatment of mucositis. Cancer 109:820–831 Sonis ST, Watkins B, Fey E, Yuschak M, Parenti D, 2005, Mechanism of action of benzydamine in the treatment of oral mucositis. J Clin Oncol 23:8040 Sonis ST, Peterson RL, Edwards LJ, Lucey CA,Wang L, Mason L et al, 2000, Defining mechanisms of action of interleukin-11 on the progression of radiation-induced oral mucositis in hamsters. Oral Oncol 36:373–381 Alsbeih G, Al-Harbi N, Al-Hadyan K, El-Sebaie M, Al-Rajhi N, 2010, Association between normal tissue complications after radiotherapy and polymorphic variations in TGFB1 and XRCC1 genes. Radiat Res 173:505–511 Song Y-Z, Han F-J, Liu M, Xia C-C, Shi W-Y, Dong L-H, 2015, Association between single nucleotide polymorphisms in XRCC3 and radiation-induced adverse effects on normal tissue: ameta-analysis. PLoS One 10(6):e0130388., DOI 10.1371/journal.pone.0130388 Kornguth DG, Garden AS, ZhengY, DahlstromKR,Wei Q, Sturgis EM, 2005, Gastrostomy in oropharyngeal cancer patients with ERCC4, XPF, germline variants. Int J Radiat Oncol Biol Phys 62:665–671 Yu J, Huang Y, Liu L,Wang J, Yin J, Huang L et al, 2016, Genetic polymorphisms of Wnt/β-catenin pathway genes are associated with the efficacy and toxicities of radiotherapy in patients with nasopharyngeal carcinoma. Oncotarget 19., DOI 10.18632/ oncotarget.12754 Naumann P, Habermehl D, Welzel T, Debus J, Combs SE, 2013, Outcome after neoadjuvant chemoradiation and correlation with nutritional status in patients with locally advanced pancreatic cancer. Strahlenther Onkol 189(9):745–752 Steel GG, 2002, Basic clinical radiobiology, 3rd edn. Hodder Arnold, London NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 135 EP 143 DI 10.1007/s12253-017-0227-1 PG 9 ER PT J AU Heydari, K Saidijam, M Sharifi, RM Dermani, KF Asl, SS Shabab, N Najafi, R AF Heydari, Korosh Saidijam, Massoud Sharifi, Reza Mohammad Dermani, Karimi Fatemeh Asl, Soleimani Sara Shabab, Nooshin Najafi, Rezvan TI The Effect of miR-200c Inhibition on Chemosensitivity (5- FluoroUracil) in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; miR-200c; 5-FU resistance; E-cadherin; PTEN ID Colorectal cancer; miR-200c; 5-FU resistance; E-cadherin; PTEN AB 5-Fluorouracil (5-FU) as a chemotherapeutic drug is used to treat colorectal cancer (CRC). However, 5-FU is associated with acquired CRC resistance, which decreases the therapeutic potential of 5-FU. Several studies indicated that miR- 200c is also involved in chemotherapeutic drug resistance, but the exact mechanism of miR-200c mediated chemoresistance has not yet been fully understood. In this study, we examined the effect of inhibition of miR-200c on the sensitivity of HCT- 116 cells to 5-FU. HCT-116 cells were transfected with LNA anti-miR-200c for 48 h. mRNA expression of miR-200c was investigated by qRT-PCR. The protein expression of phosphatase and tensin homolog (PTEN) and E-cadherin were evaluated by western blotting. Annexin V/ PI staining and caspase 3 activity were used to detect apoptosis. LNA-anti-miR-200c inhibited the miR-200c expression in the transfected cells compared with that in the control group. LNA-anti-miR-200c suppressed the expression of PTEN and E-cadherin independent of the presence of the chemotherapeutic drug 5-FU. LNA-anti-miR-200c reduced the 5-FU-induced apoptosis and caspase 3 activity. miR-200c, as a novel prognostic marker in CRC, can be a potential therapeutic approach to overcome chemoresistance during 5-FU chemotherapy. C1 [Heydari, Korosh] Hamadan University of Medical Sciences, Research Center for Molecular MedicineHamadan, Iran. [Saidijam, Massoud] Hamadan University of Medical Sciences, Research Center for Molecular MedicineHamadan, Iran. [Sharifi, Reza Mohammad] Isfahan University of Medical Sciences, School of Medicine, Department of Genetics and Molecular BiologyIsfahan, Iran. [Dermani, Karimi Fatemeh] Hamadan University of Medical Sciences, Research Center for Molecular MedicineHamadan, Iran. [Asl, Soleimani Sara] Hamadan University of Medical Sciences, Faculty of Medicine, Department of AnatomyHamadan, Iran. [Shabab, Nooshin] Hamadan University of Medical Sciences, Research Center for Molecular MedicineHamadan, Iran. [Najafi, Rezvan] Hamadan University of Medical Sciences, Research Center for Molecular MedicineHamadan, Iran. RP Najafi, R (reprint author), Hamadan University of Medical Sciences, Research Center for Molecular Medicine, Hamadan, Iran. EM re.najafi@umsha.ac.ir;najafi2535@gmail.com CR Deng J, Lei W, Fu J-C, Zhang L, Li J-H, Xiong J-P, 2014, Targeting miR-21 enhances the sensitivity of human colon cancer HT-29 cells to chemoradiotherapy in vitro. Biochem Biophys Res Commun 443(3):789–795 Draht MX, Riedl RR, Niessen H, Carvalho B, Meijer GA, Herman JG et al, 2012, Promoter CpG island methylation markers in colorectal cancer: the road ahead. Epigenomics 4(2):179–194 Davoodi H, Hashemi SR, Seow HF, 2013, 5-fluorouracil induce the expression of TLR4 on HCT116 colorectal cancer cell line expressing different variants of TLR4. Iran J Pharm Res:IJPR 12(2):453 Boyer J, McLean EG, Aroori S, Wilson P, McCulla A, Carey PD et al, 2004, Characterization of p53 wild-type and null isogenic colorectal cancer cell lines resistant to 5-fluorouracil, oxaliplatin, and irinotecan. Clin Cancer Res 10(6):2158–2167 Hsu H-C, Liu Y-S, Tseng K-C, Hsu C-L, Liang Y, Yang T-S et al, 2013, Overexpression of Lgr5 correlates with resistance to 5-FUbased chemotherapy in colorectal cancer. Int J Color Dis 28(11): 1535–1546 WangW, Cassidy J, O’Brien V, Ryan KM, Collie-Duguid E, 2004, Mechanistic and predictive profiling of 5-fluorouracil resistance in human cancer cells. Cancer Res 64(22):8167–8176 Leichman L, Lenz H-J, Leichman C, Groshen S, Danenberg K, Baranda J et al, 1995, Quantitation of intratumoral thymidylate synthase expression predicts for resistance to protracted infusion of 5-fluorouracil and weekly leucovorin in disseminated colorectal cancers: preliminary report from an ongoing trial. Eur J Cancer 31(7):1306–1310 Lau M, Klausen C, Leung P, 2011, E-cadherin inhibits tumor cell growth by suppressing PI3K/Akt signaling via β-catenin-Egr1- mediated PTEN expression. Oncogene 30(24):2753–2766 Bushati N, Cohen SM, 2007, microRNA functions. Annu Rev Cell Dev Biol 23:175–205 Li Z,Wang L, ZhangW, Fu Y, Zhao H, Hu Yet al, 2007, Restoring E-cadherin-mediated cell–cell adhesion increases PTEN protein level and stability in human breast carcinoma cells. Biochem Biophys Res Commun 363(1):165–170 SlabyO, Jancovicova J, Lakomy R, SvobodaM, Poprach A, Fabian P et al, 2010, Expression of miRNA-106b in conventional renal cell carcinoma is a potential marker for prediction of early metastasis after nephrectomy. J Exp Clin Cancer Res 29(1):1 Philippidou D, Schmitt M, Moser D, Margue C, Nazarov PV, Muller A et al, 2010, Signatures of microRNAs and selected microRNA target genes in human melanoma. Cancer Res 70(10): 4163–4173 Karakatsanis A, Papaconstantinou I, Gazouli M, Lyberopoulou A, Polymeneas G, Voros D, 2013, Expression of microRNAs, miR- 21, miR-31, miR-122, miR-145, miR-146a, miR-200c, miR-221, miR-222, andmiR-223 in patients with hepatocellular carcinoma or intrahepatic cholangiocarcinoma and its prognostic significance. Mol Carcinog 52(4):297–303 Cochrane DR, Spoelstra NS, Howe EN, Nordeen SK, Richer JK, 2009, MicroRNA-200c mitigates invasiveness and restores sensitivity to microtubule-targeting chemotherapeutic agents. Mol Cancer Ther 8(5):1055–1066 Ceppi P, Mudduluru G, Kumarswamy R, Rapa I, Scagliotti GV, Papotti M et al, 2010, Loss of miR-200c expression induces an aggressive, invasive, and chemoresistant phenotype in non–small cell lung cancer. Mol Cancer Res 8(9):1207–1216 Feng X, Wang Z, Fillmore R, Xi Y, 2014, MiR-200, a new star miRNA in human cancer. Cancer Lett 344(2):166–173 Chen J,WangW, Zhang Y, Hu T, Chen Y, 2014, The roles of miR- 200c in colon cancer and associated molecular mechanisms. Tumor Biol 35(7):6475–6483 Sharifi M, Salehi R, Gheisari Y, Kazemi M, 2014, Inhibition of microRNA miR-92a induces apoptosis and inhibits cell proliferation in human acute promyelocytic leukemia through modulation of p63 expression. Mol Biol Rep 41(5):2799–2808 Mirzamohammadi S, Aali E, Najafi R, Kamarul T, Mehrabani M, Aminzadeh A et al, 2015, Effect of 17β-estradiol on mediators involved in mesenchymal stromal cell trafficking in cell therapy of diabetes. Cytotherapy 17(1):46–57 Wiklund ED, Bramsen JB, Hulf T, Dyrskjot L, Ramanathan R, Hansen TB et al, 2011, Coordinated epigenetic repression of the miR-200 family and miR-205 in invasive bladder cancer. Int J Cancer 128(6):1327–1334 Davalos V, Moutinho C, Villanueva A, Boque R, Silva P, Carneiro F et al, 2012, Dynamic epigenetic regulation of the microRNA-200 family mediates epithelial and mesenchymal transitions in human tumorigenesis. Oncogene 31(16):2062–2074 Magee P, Shi L, Garofalo M, 2015, Role of microRNAs in chemoresistance. Ann Transl Med 3(21):332., DOI 10.3978/j.issn. 2305-5839.2015.11.32 Chen Y, Sun Y, Chen L, Xu X, Zhang X, Wang B et al, 2013, miRNA-200c increases the sensitivity of breast cancer cells to doxorubicin through the suppression of E-cadherin-mediated PTEN/Akt signaling. Mol Med Rep 7(5):1579–1584 Yin Y, Shen W, 2008, PTEN: a new guardian of the genome. Oncogene 27(41):5443–5453 Lu Y-X, Yuan L, Xue X-L, Zhou M, Liu Y, Zhang C et al, 2014, Regulation of colorectal carcinoma Stemness, growth, and metastasis by an miR-200c-Sox2–negative feedback loop mechanism. Clin Cancer Res 20(10):2631–2642 Liao C, Chen W, Fan X, Jiang X, Qiu L, Chen C et al, 2014, MicroRNA-200c inhibits apoptosis in pituitary adenoma cells by targeting the PTEN/Akt signaling pathway. Oncol Res 21(3):129– 136 Berlanga P, Munoz L, Piqueras M, Sirerol JA, Sanchez-Izquierdo MD, Hervas D et al, 2016, miR-200c and phospho-AKT as prognostic factors and mediators of osteosarcoma progression and lung metastasis. Mol Oncol Vivanco I, Sawyers CL, 2002, The phosphatidylinositol 3-kinase– AKT pathway in human cancer. Nat Rev Cancer 2(7):489–501 Oki E, Baba H, Tokunaga E, Nakamura T, Ueda N, Futatsugi M et al, 2005, Akt phosphorylation associates with LOH of PTEN and leads to chemoresistance for gastric cancer. Int J Cancer 117(3): 376–380 Hur K, Toiyama Y, Takahashi M, Balaguer F, Nagasaka T, Koike J et al, 2013)MicroRNA-200c modulates epithelial-to-mesenchymal transition, EMT, in human colorectal cancer metastasis. Gut 62(9): 1315–1326 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods 25(4):–402, 8 Boominathan L, 2010, The tumor suppressors p53, p63, and p73 are regulators of microRNA processing complex. PLoS One 5(5): e10615 Sun J, Ding W, Zhi J, Chen W, 2015, MiR-200 suppresses metastases of colorectal cancer through ZEB1. Tumor Biol 1–7 Karimi Dermani F, Saidijam M, Amini R, Mahdavinezhad A, Heydari K, Najafi R, 2016, Resveratrol inhibits proliferation, invasion, and epithelial–Mesenchymal transition by increasing miR- 200c expression in HCT-116 colorectal cancer cells. J Cell Biochem Kim SM, Kim JS, Kim J-H, Yun C-O, Kim EM, Kim HK et al, 2010, Acquired resistance to cetuximab is mediated by increased PTEN instability and leads cross-resistance to gefitinib in HCC827 NSCLC cells. Cancer Lett 296(2):150–159 Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Endoh H et al, 2004, Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res 64(11):3753–3756 Gotanda K, Hirota T, Matsumoto N, Ieiri I, 2013, MicroRNA-433 negatively regulates the expression of thymidylate synthase, TYMS, responsible for 5-fluorouracil sensitivity in HeLa cells. BMC Cancer 13(1):369 Hewish M, Lord CJ, Martin SA, Cunningham D, Ashworth A, 2010, Mismatch repair deficient colorectal cancer in the era of personalized treatment. Nat Rev Clin Oncol 7(4):197–208 Bronckaers A, Gago F, Balzarini J, Liekens S, 2009, The dual role of thymidine phosphorylase in cancer development and chemotherapy. Med Res Rev 29(6):903–953 Holzner S, Senfter D, Stadler S, Staribacher A, Nguyen CH, Gaggl A et al, 2016, Colorectal cancer cell-derived microRNA200 modulates the resistance of adjacent blood endothelial barriers in vitro. Oncol Rep 36(5):3065–3071 Chen J,WangW, Zhang Y, Chen Y, Hu T, 2014, Predicting distant metastasis and chemoresistance using plasma miRNAs.Med Oncol 31(1):1–7 Toden S, Okugawa Y, Jascur T, Wodarz D, Komarova NL, Buhrmann C et al, 2015, Curcumin mediates chemosensitization to 5-fluorouracil through miRNA-induced suppression of epithelial-to-mesenchymal transition in chemoresistant colorectal cancer. Carcinogenesis 36(3):355–367 Bonci D, Coppola V, Musumeci M, Addario A, Giuffrida R, Memeo L et al, 2008, The miR-15a–miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat Med 14(11):1271–1277 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 145 EP 151 DI 10.1007/s12253-017-0222-6 PG 7 ER PT J AU Murugan, P Rao, P Tamboli, P Czerniak, B Guo, CCh AF Murugan, Paari Rao, Priya Tamboli, Pheroze Czerniak, Bogdan Guo, C Charles TI Primary Ewing Sarcoma / Primitive Neuroectodermal Tumor of the Kidney: A Clinicopathologic Study of 23 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Kidney; Ewing sarcoma; Histology; Immunohistochemistry; Cytogenetics; Clinical outcome ID Kidney; Ewing sarcoma; Histology; Immunohistochemistry; Cytogenetics; Clinical outcome AB Primary Ewing sarcoma / primitive neuroec todermal tumor (ES) of the kidney is a rare neoplasm with limited clinicopathologic data. We report 23 such cases with no history of ES elsewhere in the body. The patients included 13 male and 10 female, aged 8–70 years (mean, 31 years). The average tumor size was 11.7 cm (range, 5–20 cm). Microscopic analysis showed predominantly lobular growth (n = 14), with focal papillary (n = 3), alveolar (n = 1), and hemangiopericytoma-like (n = 1) patterns. Several tumors (n = 11) exhibited robust mitotic activity (>10 mitoses/10 high-power fields). Necrosis (n = 13) and lymphovascular invasion (n = 14) were common. Homer Wright rosettes (n = 6) and perivascular pseudorosettes (n = 1) were also identified. The tumors invaded the renal sinus or perinephric fat (n = 11), renal vein (n = 13), and adrenal gland (n = 2). Molecular and fluorescence in situ hybridization analysis showed rearrangement of EWSR1 gene (10/10), associated with EWSR1-FLI1 gene fusion (7/10). All patients with follow-up information (n = 18) had metastasis, commonly in the lungs (n = 12) and bone (n = 6). Twelve patients died of disease in a mean of 21 months; 6 patients were alive at a mean of 49 months after diagnosis. Primary kidney ES usually present at an advanced stage with extrarenal spread and metastasis. Although renal ES share histologic, immunohistochemical, and molecular features with their bone and soft tissue counterparts, they appear to be more aggressive tumors with poorer clinical outcome. C1 [Murugan, Paari] University of Minnesota, Department of Laboratory Medicine and PathologyMinneapolis, MN, USA. [Rao, Priya] The University of Texas, MD Anderson Cancer CenterHouston, TX, USA. [Tamboli, Pheroze] The University of Texas, MD Anderson Cancer CenterHouston, TX, USA. [Czerniak, Bogdan] The University of Texas, MD Anderson Cancer CenterHouston, TX, USA. [Guo, C Charles] The University of Texas, MD Anderson Cancer CenterHouston, TX, USA. RP Murugan, P (reprint author), University of Minnesota, Department of Laboratory Medicine and Pathology, Minneapolis, USA. CR Ewing J, 1921, Diffuse endothelioma of bone. Proc NY Pathol Soc 21:17–24 Angerwall L, Enzinger FM, 2013, Extraskeletal neoplasms resembling Ewings sarcoma. Cancer 1975:240–251 De Alava E, Pardo J, 2001, Ewing tumor: tumor biology and clinical applications. Int J Surg Pathol 9(1):7–17 Mor Y, Nass D, Raviv G, Neumann Y, Nativ O, Goldwasser B, 1994, Malignant peripheral primitive neuroectodermal tumor, PNET, of the kidney. Med Pediatr Oncol 23:437–440 Rowe RG, Thomas DG, Schuetze SM, Hafez KS, Lawlor ER, Chugh R, 2013, Ewing sarcoma of the kidney: case series and literature review of an often overlooked entity in the diagnosis of primary renal tumors. Urology 81(2):347–353 Parham DM, Roloson GJ, Feely M, Green DM, Bridge JA, Beckwith JB, 2001, Primary malignant neuroepithelial tumors of the kidney: a clinicopathologic analysis of 146 adult and pediatric cases from the National Wilms tumor study group pathology Center. Am J Surg Pathol 25(2):133–146 Toomey EC, Schiffman JD, Lessnick SL, 2010, Recent advances in the molecular pathogenesis of Ewing's sarcoma. Oncogene 29(32): 4504–4516 Goldblum JR, Folpe AL, Weiss SW, 2014, Malignant soft tissue tumors of uncertain type. In: Goldblum JR, Folpe AL, Weiss SW, eds, Enzinger and Weiss Soft Tissue Tumors. Elsevier Inc, Philedelphia, p 1028 Bartholow T, Parwani A, 2012, Renal primitive neuroectodermal tumors. Arch Pathol Lab Med 136(6):686–690 Jimenez RE, Folpe AL, Lapham RL, Ro JY, O'Shea PA,Weiss SW et al, 2002, Primary Ewings sarcoma/primitive neuroectodermal tumor of the kidney: a clinicopathologic and immunohistochemical analysis of 11 cases. Am J Surg Pathol 26(3):320–327 Folpe AL, Hill CE, Parham DM, O'shea PA, Weiss SW, 2000, Immunohistochemical detection of FLI-1 protein expression: a study of 132 round cell tumors with emphasis on CD99-positive mimics of Ewings sarcoma/primitive neuroectodermal tumor. Am J Surg Pathol 24(12):1657–1662 Rodriguez-Galindo C, Spunt SL, Pappo AS, 2003, Treatment of Ewing sarcoma family of tumors: current status and outlook for the future. Med Pediatr Oncol 40(5):276–287 Rajani R, Gibbs CP, 2012, Treatment of bone tumors. Surg Pathol Clin 5(1):301–318 Bing Z, Zhang P, Tomaszewski JE, Maclennan GT, 2009, Primary Ewing sarcoma/primitive neuroectodermal tumor of the kidney. J Urol 181(3):1341–1342 Thyavihally YB, Tongaonkar HB, Gupta S, Kurkure PA, Amare P, Muckaden MA et al, 2008, Primitive neuroectodermal tumor of the kidney: a single institute series of 16 patients. Urology 71(2):292–296 Argani P, Ladanyi M, 2003, Recent advances in pediatric renal neoplasia. Adv Anat Pathol 10(5):243–260 Ladenstein R, Potschger U, Le deley MC, Whelan J, Paulussen M, Oberlin O et al, 2010, Primary disseminated multifocal Ewing sarcoma: results of the euro-EWING 99 trial. J Clin Oncol 28(20): 3284–3291 Luo W, Gangwal K, Sankar S, Boucher KM, Thomas D, Lessnick SL, 2009, GSTM4 is a microsatellite-containing EWS/FLI target involved in Ewings sarcoma oncogenesis and therapeutic resistance. Oncogene 28(46):4126–4132 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 153 EP 159 DI 10.1007/s12253-017-0228-0 PG 7 ER PT J AU Roncati, L Gatti, MA Barbolini, G Piscioli, F Pusiol, T Maiorana, A AF Roncati, Luca Gatti, Morena Antonietta Barbolini, Giuseppe Piscioli, Francesco Pusiol, Teresa Maiorana, Antonio TI In Vivo Uptake of Rare Earth Metals by Triple-Negative Breast Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Triple-negative breast cancer; Rare earth metals (REM); Europium (Eu); Dysprosium (Dy); Praseodymium (Pr); Elemental microanalysis ID Triple-negative breast cancer; Rare earth metals (REM); Europium (Eu); Dysprosium (Dy); Praseodymium (Pr); Elemental microanalysis AB Rare earth metals (REM) are a group of 17 chemical elements in the periodic table, namely scandium (Sc), yttrium (Y) and the lanthanides. In relation to atomic volume and geological behavior, the lanthanides are further subdivided into light, medium and heavy REM. They find many applications in the technological field; however, their impact on the human health is still conflicting and, for many aspects, unknown. During a research program carried on 113 cases of female breast cancer, immunohistochemically categorized in Her2-positive (29 cases), Her2-negative (57 cases) and triple negative (27 cases), aimed to evaluate the role of environmental particulate in carcinogenesis by elemental microanalysis, for the first time in literature we have detected a REM uptake, in detail europium (Eu), dysprosium (Dy) and praseodymium (Pr), inside the neoplastic cells belonging to a single triple negative breast cancer. Curiously, the woman affected by this form of malignancy had worked in the ceramic industry, a well-known source of REM, during her life, and she was the one and only patient of our series to be dedicated to this activity. The medical repercussions of our findings are here discussed: in fact, a REM detection in only 1 of 113 examined cases seems to exclude active roles in breast carcinogenesis and discloses new possibilities for therapeutic developments in triple negative breast cancer. C1 [Roncati, Luca] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical Pathology, I-41124 Modena, MO, Italy. [Gatti, Morena Antonietta] National Research Council, Institute of Science and Technology for CeramicsFaenza, RA, Italy. [Barbolini, Giuseppe] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical Pathology, I-41124 Modena, MO, Italy. [Piscioli, Francesco] Provincial Health Care Services, Santa Maria del Carmine Hospital, Institute of PathologyRovereto, TN, Italy. [Pusiol, Teresa] Provincial Health Care Services, Santa Maria del Carmine Hospital, Institute of PathologyRovereto, TN, Italy. [Maiorana, Antonio] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical Pathology, I-41124 Modena, MO, Italy. RP Roncati, L (reprint author), University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical Pathology, I-41124 Modena, Italy. EM emailmedical@gmail.com CR Connelly NG, Damhus T, Hartshorn RM, Hutton AT, 2005, Nomenclature of inorganic chemistry: IUPAC recommendations. RSC Publishing, Cambridge Gschneidner KA, Cappellen J, 1987, 1787–1987 two hundred years of rare earths. North-Holland, Rare Earth Information Center Haxel GB, Hedrick JB, Orris GJ, 2002, Rare earth elements - critical resources for high technology. U.S. Geological Survey. http://pubs.usgs.gov/fs/2002/fs087-02/fs087-02.pdf. Accessed 20 November 2002 Zhang J, Chen Q,Wang N, Zhang J, 2003, Levels and distribution of 15 rare earth elements in tumor and normal lung tissue from the patients with lung cancer. Wei Sheng Yan Jiu 32:423–426 Dai Y, Li J, Li J, Yu L, Dai G, Hu A et al, 2002, Effects of rare earth compounds on growth and apoptosis of leukemic cell lines. In Vitro Cell Dev Biol Anim 38:373–375 Zhuang G, Zhou Y, Lu H, Lu W, Zhou M, Wang Y et al, 1996, Concentration of rare earth elements, As, and Th in human brain and brain tumors, determined by neutron activation analysis. Biol Trace Elem Res 53:45–49 Roncati L, Barbolini G, Piacentini F, Piscioli F, Pusiol T, Maiorana A, 2016, Prognostic factors for breast cancer: an immunomorphological update. Pathol Oncol Res 22:449–452 Gatti AM, Montanari S, 2005, Risk assessment of microparticles and nanoparticles and human health. In: Nalwa HS, ed, Handbook of nanostructured biomaterials and their applications in nanobiotechnology. American Scientific Publishers, Portland, pp 347–369 Azimi G, Dhiman R, Kwon HM, Paxson AT, Varanasi KK, 2013, Hydrophobicity of rare-earth oxide ceramics. NatMater 12:315–320 Shibata N, Pennycook SJ, Gosnell TR, Painter GS, Shelton WA, Becher PF, 2004, Observation of rare-earth segregation in silicon nitride ceramics at subnanometre dimensions. Nature 428:730–733 Ji YJ, Xiao B, Wang ZH, Cui MZ, Lu YY, 2000, The suppression effect of light rare earth elements on proliferation of two cancer cell lines. Biomed Environ Sci 13:287–292 Chen GJ, Wang ZG, Qiao X, Xu JY, Tian JL, Yan SP, 2013, Synthesis, DNA binding, photo-induced DNA cleavage, cytotoxicity studies of a family of heavy rare earth complexes. J Inorg Biochem 127:39–45 Zhou J, Wang LF, Wang JY, Tang N, 2001, Synthesis, characterization, antioxidative and antitumor activities of solid quercetin rare earth, III, complexes. J Inorg Biochem 83:41–48 Haley TJ, Komesu N, Colvin G, Koste L, Upham HC, 1965, Pharmacology and toxicology of europium chloride. J Pharm Sci 54:643–645 Bruce DW, Hietbrink BE, Dubois KP, 1963, The acute mammalian toxicity of rare earth nitrates and oxides. Toxicol Appl Pharmacol 5: 750–759 Giri S, Karakoti A, Graham RP, Maguire JL, Reilly CM, Seal S et al, 2013, Nanoceria: a rare-earth nanoparticle as a novel antiangiogenic therapeutic agent in ovarian cancer. PLoS One 8: e54578 Roncati L, Barbolini G, Gatti AM, Pusiol T, Piscioli F, Maiorana A, 2016, The uncontrolled sialylation is related to chemoresistant metastatic breast cancer. Pathol Oncol Res 22:869–873 FoulkesWD, Smith IE, Reis-Filho JS, 2010, Triple-negative breast cancer. N Engl J Med 363:1938–1948 Hudis CA, Gianni L, 2011, Triple-negative breast cancer: an unmet medical need. Oncologist 16:1–11 Basu S, Chen W, Tchou J, Mavi A, Cermik T, Czerniecki B et al, 2008, Comparison of triple-negative and estrogen receptor-positive/ progesterone receptor-positive/HER2-negative breast carcinoma using quantitative fluorine-18 fluorodeoxyglucose/positron emission tomography imaging parameters: a potentially useful method for disease characterization. Cancer 112:995–1000 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 161 EP 165 DI 10.1007/s12253-017-0209-3 PG 5 ER PT J AU Cserni, G Zombori, T Andreu, X Bianchi, S Regitnig, P Amendoeira, I Balmativola, D Kovacs, A Cordoba, A Reiner, A Kulka, J Kaya, H Liepniece-Karele, I Quinn, C Kovari, B AF Cserni, Gabor Zombori, Tamas Andreu, Xavier Bianchi, Simonetta Regitnig, Peter Amendoeira, Isabel Balmativola, Davide Kovacs, Aniko Cordoba, Alicia Reiner, Angelika Kulka, Janina Kaya, Handan Liepniece-Karele, Inta Quinn, Cecily Kovari, Bence TI Is Regression after Neoadjuvant Chemotherapy for Locally Advanced Breast Cancer Different in Sentinel and Non-sentinel Nodes? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sentinel lymph node; Non-sentinel lymph node; Breast cancer; Neoadjuvant therapy; Selective regression ID Sentinel lymph node; Non-sentinel lymph node; Breast cancer; Neoadjuvant therapy; Selective regression AB Tumor draining sentinel lymph nodes (SLNs) are the sites of selective changes as compared to non-SLNs. They show features of tumor-reactive lymphadenopathy, including increased total number of functional blood vessels, but a relative immunosuppressed status has also been described in them. We explored the hypothesis of a selective regression or non-regression in SLNs versus non-SLNs in 142 patients with 110 estrogen receptor-positive and 32 estrogen receptornegative tumors undergoing both SLN biopsy and axillary lymph node dissection after neoadjuvant therapy by assessing the tumoral (metastatic) and regression statuses of SLNs and non-SLNs separately. Of the 89 cases with signs of nodal regression, 22 cases (25%) were in favor of a selective nonregression in SLNs, 18 cases (20%) were supportive of a selective and more pronounced regression in the SLNs and the remaining showed equal degrees of regression or nonregression in SLNs and non-SLNs. The results indicate that there is no obvious difference in the degree of regressive histological changes shown by SLNs and NSLNs. Therefore, this phenomenon may not be a major contributor to the higher false negative rate of SLN biopsy after neoadjuvant treatment. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, 6000 Kecskemet, Hungary. [Zombori, Tamas] University of Szeged, Department of Pathology, Allomas u 1, 6725 Szeged, Hungary. [Andreu, Xavier] University Autonoma Barcelona, Corporacio Sanitaria Parc Tauli, Department of Pathology, 08202 Sabadell, Spain. [Bianchi, Simonetta] AOU Careggi, Department of Surgery and Translational Medicine, Largo G. A. Brambilla 3, 50134 Florence, Italy. [Regitnig, Peter] Medical University of Graz, Department of Pathology, Auenbruggerplatz 25, A-8036 Graz, Austria. [Amendoeira, Isabel] Centro Hospitalar de Sao Joao e IPATIMUP, Laboratorio de Anatomia Patologica, 4440-563 Porto, Portugal. [Balmativola, Davide] IRCCS, Fondazione del Piemonte per l’Oncologia (FPO), Candiolo Cancer Institute, Str. Prov. 142, km 3.95, 10060 Candiolo, Italy. [Kovacs, Aniko] Sahlgrenska University Hospital, Department of Clinical Pathology and Genetics, Gula straket 8, 41345 Gothenburg, Sweden. [Cordoba, Alicia] Complejo Hospitalario de Navarra, Department of Pathology, Irunlarrea 3, 31008 Navarra, Spain. [Reiner, Angelika] Donauspital, Pathologisch-Bakteriologisches Institut, Langobardenstraße, 122 Vienna, Austria. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kaya, Handan] Marmara University, School of Medicine, Department of PathologyIstanbul, Turkey. [Liepniece-Karele, Inta] Riga East Clinical University Hospital, Pathology Centre, Hipokrata St 2, LV-1038 Riga, Latvia. [Quinn, Cecily] St. Vincent’s University Hospital, Elm ParkDublin, Ireland. [Kovari, Bence] University of Szeged, Department of Pathology, Allomas u 1, 6725 Szeged, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, 6000 Kecskemet, Hungary. EM cserni@freemail.hu CR Greenall MJ, 1995, Why I favour axillary node sampling in the management of breast cancer. Eur J Surg Oncol 21:2–5 Cserni G, 1999, Estimating the overlap between sentinel lymph nodes and axillary node samples in breast cancer. Pathol Oncol Res 5:129–133 Qian CN, Berghuis B, Tsarfaty G, Bruch M, Kort EJ, Ditlev J, Tsarfaty I, Hudson E, Jackson DG, Petillo D, Chen J, Resau JH, Teh BT, 2006, Preparing the "soil": the primary tumor induces vasculature reorganization in the sentinel lymph node before the arrival of metastatic cancer cells. Cancer Res 66:10365–10376 Schule JM, Bergkvist L, Hakansson L, Gustafsson B, Hakansson A, 2004, CD28 expression in sentinel node biopsies from breast cancer patients in comparison with CD3-zeta chain expression. J Transl Med 2:45., DOI 10.1186/1479-5876-2-45 Schule J, Bergkvist L, Hakansson L, Gustafsson B, Hakansson A, 2002, Down-regulation of the CD3-zeta chain in sentinel node biopsies from breast cancer patients. Breast Cancer Res Treat 74:33–40 Cochran AJ, Huang RR, Lee J, Ikatura E, Leong SP, Essner R, 2006, Tumour–induced immune modulation of sentinel lymph nodes. Nat Rev Immunol 6:659–670 Straver ME, Loo CE, Alderliesten T, Rutgers EJ, Vrancken Peeters MT, 2010, Marking the axilla with radioactive iodine seeds, MARI procedure, may reduce the need for axillary dissection after neoadjuvant chemotherapy for breast cancer. Br J Surg 97:1226–1231 Wells CA, Amendoeira I, Bellocq JP et al, 2012, S2: pathology update. Quality assurance guidelines for pathology. In: Perry N, Broeders M, de Wolf C et al, eds, European guidelines for quality assurance in breast cancer screening and diagnosis, 4th edn, supplements. European Commission, Office for Official Publications of the European Union, Luxembourg, pp 73–120 Mocellin S, Goldin E, Marchet A, Nitti D, 2016, Sentinel node biopsy performance after neoadjuvant chemotherapy in locally advanced breast cancer: a systematic review and meta-analysis. Int J Cancer 138:472–480 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 167 EP 170 DI 10.1007/s12253-017-0229-z PG 4 ER PT J AU Pavlov, I Hadjiev, E Alaikov, T Spassova, S Stoimenov, A Naumova, E Shivarov, V Ivanova, M AF Pavlov, Ivan Hadjiev, Evgueniy Alaikov, Tzvetan Spassova, Sylva Stoimenov, Angel Naumova, Elissaveta Shivarov, Velizar Ivanova, Milena TI Calreticulin Mutations in Bulgarian MPN Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MPNs; CALR; JAK2; MPL; Mutations ID MPNs; CALR; JAK2; MPL; Mutations AB Somatic mutations in JAK2, MPL and CALR are recurrently identified in most of the cases with Philadelphia chromosome negative myeloproliferative neoplasms (MPNs). We applied four molecular genetic methods for identification of CALR exon 9 mutations, including high resolution melt (HRM) analysis, Sanger sequencing, semiconductor target genes sequencing and whole exome sequencing. A total of 78 patients with myeloid malignancies were included in the study.We identified 14 CALR exon 9 mutated cases out of 78 studied patients with myeloid malignancies. All mutated patients were diagnosed with MPN being either PMF (n = 7) or ET (n = 7). Nine cases had type 1 mutations and 5 cases had type 2 mutations. CALR exon 9, MPL exon 10 and JAK2 p. V617F were mutually exclusive. There were no statistically significant differences in the hematological parameters between the cases with CALR and JAK2 or MPL mutations. Notably, all four techniques were fully concordant in the detection of CALR mutations. This is one of the few reports on the CALR mutations frequency in South-eastern populations. Our study shows that the frequency and patterns of these mutations is identical to those in the patients’ cohorts from Western countries. Besides we demonstrated the utility of four different methods for their detection. C1 [Pavlov, Ivan] Medical University, Alexandrovska University Hospital, Laboratory of Clinical Immunology, 1 Georgi Sofiisky Blvd, 1431 Sofia, Bulgaria. [Hadjiev, Evgueniy] Medical University, Alexandrovska University Hospital, Department of Clinical HematologySofia, Bulgaria. [Alaikov, Tzvetan] Sofiamed University Hospital, Department of Clinical HematologySofia, Bulgaria. [Spassova, Sylva] Sofiamed University Hospital, Department of Clinical HematologySofia, Bulgaria. [Stoimenov, Angel] Sofiamed University Hospital, Laboratory of Transfusion MedicineSofia, Bulgaria. [Naumova, Elissaveta] Medical University, Alexandrovska University Hospital, Laboratory of Clinical Immunology, 1 Georgi Sofiisky Blvd, 1431 Sofia, Bulgaria. [Shivarov, Velizar] Sofiamed University Hospital, Department of Clinical HematologySofia, Bulgaria. [Ivanova, Milena] Medical University, Alexandrovska University Hospital, Laboratory of Clinical Immunology, 1 Georgi Sofiisky Blvd, 1431 Sofia, Bulgaria. RP Ivanova, M (reprint author), Medical University, Alexandrovska University Hospital, Laboratory of Clinical Immunology, 1431 Sofia, Bulgaria. EM mivanova@intech.bg CR Nangalia J, Grinfeld J, Green AR, 2016, Pathogenesis of myeloproliferative disorders. Annu Rev Pathol 11:101–126., DOI 10.1146/annurev-pathol-012615-044454 Nangalia J, Massie CE, Baxter EJ, Nice FL, Gundem G, Wedge DC, Avezov E, Li J, Kollmann K, Kent DG, Aziz A, Godfrey AL, Hinton J, Martincorena I, Van Loo P, Jones AV, Guglielmelli P, Tarpey P, Harding HP, Fitzpatrick JD, Goudie CT, Ortmann CA, Loughran SJ, Raine K, Jones DR, Butler AP, Teague JW, O'Meara S, McLaren S, BianchiM, Silber Y, Dimitropoulou D, Bloxham D, Mudie L, Maddison M, Robinson B, Keohane C, Maclean C, Hill K, Orchard K, Tauro S, Du MQ, Greaves M, Bowen D, Huntly BJ, Harrison CN, Cross NC, Ron D, Vannucchi AM, Papaemmanuil E, Campbell PJ, Green AR, 2013, Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med 369(25):2391–2405., DOI 10.1056/NEJMoa1312542 Klampfl T, Gisslinger H, Harutyunyan AS, Nivarthi H, Rumi E, Milosevic JD, Them NC, Berg T, Gisslinger B, Pietra D, Chen D, Vladimer GI, Bagienski K, Milanesi C, Casetti IC, Sant'Antonio E, Ferretti V, Elena C, Schischlik F, Cleary C, Six M, Schalling M, Schonegger A, Bock C, Malcovati L, Pascutto C, Superti-Furga G, Cazzola M, Kralovics R, 2013, Somaticmutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 369(25):2379–2390., DOI 10.1056/NEJMoa1311347 IvanovaMI, Shivarov VS, Hadjiev EA, Naumova EJ, 2011, Novel multiplex bead-based assay with LNA-modified probes for detection of MPL exon 10 mutations. Leuk Res 35(8):1120–1123., DOI 10.1016/j.leukres.2011.04.012 Shivarov V, Ivanova M, Hadjiev E, Naumova E, 2011, Rapid quantification of JAK2 V617F allele burden using a bead-based liquid assay with locked nucleic acid-modified oligonucleotide probes. Leuk Lymphoma 52(10):2023–2026., DOI 10.3109/10428194.2011.584995 Jones AV, Ward D, Lyon M, Leung W, Callaway A, Chase A, Dent CL, White HE, Drexler HG, Nangalia J, Mattocks C, Cross NC, 2015, Evaluation of methods to detect CALR mutations in myeloproliferative neoplasms. Leuk Res 39(1): 82–87., DOI 10.1016/j.leukres.2014.11.019 Ivanova M, Shivarov V, Pavlov I, Lilakos K, Naumova E, 2016, Clinical evaluation of a novel nine-gene panel for ion torrent PGM sequencing ofmyeloidmalignancies.Mol Diagn Ther 20(1):27–32., DOI 10.1007/s40291-015-0172-1 Guenova M, Stoeva V, Dimitrova S, Gercheva L, Petrov Y, Todorieva D, Grklanov V, Tsvetkova G, Tumbeva D, Grudeva- Popova J, Petkova N, Raynov J, Sivcheva L, Dimitrova G, Zhukova M, Topalov Y, Dikov T, Balatzenko G, Mihaylov G, 2016, Baseline characteristics and risk factors for leukemia-free and overall survival in bulgarian patients with primary myelofibrosis ? A national cohort study Tefferi A, Wassie EA, Guglielmelli P, Gangat N, Belachew AA, Lasho TL, Finke C, Ketterling RP, Hanson CA, Pardanani A, Wolanskyj AP, Maffioli M, Casalone R, Pacilli A, Vannucchi AM, Passamonti F, 2014, Type 1 versus type 2 calreticulin mutations in essential thrombocythemia: a collaborative study of 1027 patients. Am J Hematol 89(8):E121–E124., DOI 10.1002/ajh.23743 Tefferi A, Guglielmelli P, Lasho TL, Rotunno G, Finke C, Mannarelli C, Belachew AA, Pancrazzi A, Wassie EA, Ketterling RP, Hanson CA, Pardanani A, Vannucchi AM, 2014, CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: an international study of 570 patients. Leukemia 28(7): 1494–1500., DOI 10.1038/leu.2014.57 Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW, 2016, The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127(20): 2391–2405., DOI 10.1182/blood-2016-03-643544 Lundberg P, Karow A, Nienhold R, Looser R, Hao-Shen H, Nissen I, Girsberger S, Lehmann T, Passweg J, Stern M, Beisel C, Kralovics R, Skoda RC, 2014, Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms. Blood 123(14):2220–2228., DOI 10.1182/blood-2013-11-537167 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 171 EP 174 DI 10.1007/s12253-017-0226-2 PG 4 ER PT J AU Choi, JE Kim, SM Song, YS Yoo, JN Lee, HS AF Choi, Ji Eun Kim, Sung Min Song, Yong Sang Yoo, Jin Nam Lee, Hyung Sug TI Low Frequent Mutation of ARHGAP35, a Candidate Tumor Suppressor Gene, in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE ARHGAP35; RHO inhibition; Mutation; Cancers; Microsatellite instability ID ARHGAP35; RHO inhibition; Mutation; Cancers; Microsatellite instability C1 [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Song, Yong Sang] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of PathologySeoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR LeClerc S, Palaniswami R, Xie BX, GovindanMV(1991, Molecular cloning and characterization of a factor that binds the human glucocorticoid receptor gene and represses its expression. J Biol Chem 266:17333–17340 Tikoo A, Czekay S, Viars C,White S, Heath JK, Arden K,Maruta H, 2000, p190-a, a human tumor suppressor gene, maps to the chromosomal region 19q13.3 that is reportedly deleted in some gliomas. Gene 257:23–31 Bartolome RA, Diaz-Martinez M, Colo GP, Arellano-Sanchez N, Torres-Ayuso P, Kleinovink JW, Merida I, Teixido J, 2014, A Blk– p190RhoGAP signaling module downstream of activated Gα13 functionally opposes CXCL12-stimulated RhoA activation and cell invasion. Cell Signal 26:2551–2561 Wang DZ, Nur-E-Kamal MS, Tikoo A, Montague W, Maruta H, 1997, The GTPase and rho GAP domains of p190, a tumor suppressor protein that binds the M(r, 120,000 Ras GAP, independently function as anti-Ras tumor suppressors. Cancer Res 57:2478–2484 Sun Q, Cibas ES, Huang H, Hodgson L, Overholtzer M, 2014, Induction of entosis by epithelial cadherin expression. Cell Res 24: 1288–1298 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 175 EP 176 DI 10.1007/s12253-017-0208-4 PG 2 ER PT J AU Robert, L AF Robert, Ladislas TI Tribute to Prof. George Klein SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Robert, Ladislas] University of Paris 5, Hotel Dieu HospitalParis, France. RP Robert, L (reprint author), University of Paris 5, Hotel Dieu Hospital, Paris, France. EM Lrobert5@orange.fr NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 177 EP 177 DI 10.1007/s12253-017-0207-5 PG 1 ER PT J AU Soike, MTh Maize, CJ Ralston, SJ Hayes, B Swick, J AF Soike, M Thomas Maize, C John Ralston, S Jonathan Hayes, Benjamin Swick, Julie TI Dual Immunohistochemical Detection of Mitoses in Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Soike, M Thomas] Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 171 Ashley Avenue, MSC 908, 29425 Charleston, SC, USA. [Maize, C John] Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 171 Ashley Avenue, MSC 908, 29425 Charleston, SC, USA. [Ralston, S Jonathan] Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 171 Ashley Avenue, MSC 908, 29425 Charleston, SC, USA. [Hayes, Benjamin] Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 171 Ashley Avenue, MSC 908, 29425 Charleston, SC, USA. [Swick, Julie] Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 171 Ashley Avenue, MSC 908, 29425 Charleston, SC, USA. RP Soike, MTh (reprint author), Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 29425 Charleston, USA. EM soiketm2@msha.com CR Blach C, Soong SJ, Gershenwal JE, 2001, Prognostic factors analsyis of 17,600 melanoma patients: validation of the AMerican joint Commision on cancer melanoma staging system. Am J Clin Oncol 19:3622–3634 Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, editors. AJCC cancer staging manual, 7th ed). New York, NY: Springer; 2010 Gerdes J, Schwab U, Lemke H, Stein H, 1983, Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer 31(1):13–20 Hendzel M, 1997, Mitosis-specific phosphorylation of histone H3 initiates primarily within pericentromeric heterochromatin during G2 and spreads in an ordered fashion coincident with mitotic chromosome condensation. Chromosoma 6:348–360 Kawakami Y, Eliyahu S, Delgado C, Robbins P, 1994, Cloning of the Gene coding for a shared human melanoma antigen recognized by autologous Tcells infiltrating into tumor. Proc Natl Acad Sci U S A 91(9):3515–3519 Ponti G, Pollio A, Cesinaro AM, Pellecani G, 2012, Value and prognostic significance of mitotic rate in a retrospective series of pT1 cutaneous malignant melanoma patients. Cancer Epidemiol 36:303–305 Kesmodel S, Karakouisis C, Botbyl J, Canter R, 2005)Mitotic rate as a predictor of sentinel lymph node positivity in patients with thin melanomas. Ann Surg Oncol 12:449–458 Shibata K, Ajiro K, 1993, Cell cycle-dependent suppressive effect of histone H1 on mitosis-specific H3 phosphorylation. J Biol Chem 25:18431–18434 Ikenberg K, Pfaltz M, Rakozy C, Kempf W, 2012, Immunohistochemical dual staining as an adjunct in assessment of mitotic activity in melanoma. J Cutan Pathol 324-330 Schimming TT, Grabellus F, Roner M, Pechlivanis S, 2012, pHH3 immunostaining improves Interobserver agreement of mitotic index in thin melanomas. Am J Dermatopathol 34:266–269 Thareja S, Zager J, Sadhwani D, 2014, Analysis of tumor mitotic rate in thin metastatic melanomas compared with thin melanomas without metastasis using both the hematoxylin and eosin and antiphosphohistone 3 IHC stain. Am J Dermpathol 64-67 Hale CS, 2013, Mitotic rate in melanoma prognostic value of immunostaining and computer-assisted image analysis. Am J Surg Pathol 37(6):882–889 Tatzlaff M, Curry J, Ivan D, 2013, Immunodetection of pshophoistone H3 as a surrogate ofmitotic figure count and clincial outcome in cutaneous melanoma. Mod Pathol 26:1153–1160 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 179 EP 183 DI 10.1007/s12253-017-0203-9 PG 5 ER PT J AU Choi, JE Kim, SM Yoo, JN Lee, HS AF Choi, Ji Eun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI TRIO Gene Encoding Trio Rho Guanine Nucleotide Exchange Factor Harbors Frameshift Mutations of in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE TRIO; Frameshift mutation; Cancer; Microsatellite instability ID TRIO; Frameshift mutation; Cancer; Microsatellite instability C1 [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Schmidt S, Debant A, 2014, Function and regulation of the Rho guanine nucleotide exchange factor Trio. Small GTPases 5:e29769 Lane J,Martin TA,Mansel RE, JiangWG(2008, The expression and prognostic value of the guanine nucleotide exchange factors, GEFs, Trio, Vav1 and TIAM-1 in human breast cancer. Int Semin Surg Oncol 5:23 Molenaar JJ, Koster J, Zwijnenburg DA, van Sluis P, Valentijn LJ, van der Ploeg I, Hamdi M, van Nes J, Westerman BA, van Arkel J, Ebus ME, Haneveld F, Lakeman A, Schild L, Molenaar P, Stroeken P, van Noesel MM, Ora I, Santo EE, Caron HN, Westerhout EM, Versteeg R, 2012, Sequencing of neuroblastoma identifies chromothripsis and defects in neuritogenesis genes. Nature 483: 589–593 Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat JP, Nickerson E, Auclair D, Li L, Place C, Dicara D, Ramos AH, Lawrence MS, Cibulskis K, Sivachenko A, Voet D, Saksena G, Stransky N, Onofrio RC, Winckler W, Ardlie K, Wagle N,Wargo J, Chong K, Morton DL, Stemke-Hale K, Chen G, Noble M, MeyersonM, Ladbury JE, DaviesMA, Gershenwald JE,Wagner SN, Hoon DS, Schadendorf D, Lander ES, Gabriel SB, Getz G, Garraway LA, Chin L, 2012, A landscape of driver mutations in melanoma. Cell 150:251–263 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Jo YS, Choi MR, Song SY, Kim MS, Yoo NJ, Lee SH, 2016, Frameshift mutations of HSPA4 and MED13 in gastric and colorectal cancers. Pathol Oncol Res 22:769–772 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 185 EP 187 DI 10.1007/s12253-017-0211-9 PG 3 ER PT J AU Terada, T AF Terada, Tadashi TI A Retrospective Case Control Study of Ductal Plate Malformation-like Features in Consecutive 200 Autopsies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Terada, Tadashi] Shizuoka City Shimizu Hospital, Department of Pathology, No. 4-27-1, Kita-Ando, Aoi-ku, 420-8527 Shizuoka, Japan. RP Terada, T (reprint author), Shizuoka City Shimizu Hospital, Department of Pathology, 420-8527 Shizuoka, Japan. EM piyo0111jp@yahoo.co.jp CR Jorgensen MJ, 1977, The ductal plate malformation. Acta Pathol Microbiol Scand Suppl 257:1–87 Summerfield JA, Nagafuchi Y, Sherlock S, Cadafalch J, Scheuer PJ, 1986, Hepatobiliary fibropolycystic diseases: a clinical and histological review of 51 patients. J Hepatol 2:141–156 Van Eyken P, Sciot R, Callea F, Van der Steen K, Moerman P, Desmet VJ, 1988, The development of the intrahepatic bile ducts in man: a keratin immunohistochemical study. Hepatology 8:1586– 1595 Desmet VJ, 1992, Congenital disease of intrahepatic bile ducts: variations on the theme "ductal plate malformation". Hepatology 16:1069–1083 Desmet VJ, 1985, Intrahepatic bile duct under the lens. J Hepatol 1: 545–559 Roskams T, Desmet V, 2008, Embryology of extra- and intrahepatic bile ducts, the ductal plate. Anat Rec 291:628–635 Shah KD, Gerber MA, 1989, Development of intrahepatic bile ducts in humans: immunohistochemical study using monoclonal cytokeratin antibodies. Arch Pathol Lab Med 113:1135–1138 Haruna Y, Saito K, Spaulding S, Nalesnik MA, Gerber MA, 1996, Identification of bipotential progenitor cells in human liver development. Hepatology 23:476–481 Shar KD, Gerber MA, 1990, Development of intrahepatic bile ducts in humans: possible role of laminin. Arch Pathol Lab Med 114:597–600 Terada T, Nakanuma Y, 1993, Development of human intrahepatic peribiliary glands: histological, keratin immunohistochemical and mucus histochemical analyses. Lab Investig 68:261–269 Terada T, Nakanuma Y, 1994, Profiles of expression of carbohydrate chain structures during human intrahepatic bile duct development and maturation: a lectin-histochemical and immunohistochemical study. Hepatology 20:388–397 Terada T, Kitamura Y, Nakanuma Y, 1997, Normal and abnormal development of the intrahepatic biliary system: a review. Tohoku J Exp Med 181:19–32 Terada T, Ashida K, Kitamura Y, Matsunaga Y, Takashima K, Kato M, Ohta T, 1998, Expression of E-cadherin, alpha-catenin and beta-catenin during human intrahepatic bile duct development. J Hepatol 28:263–269 Terada T, 2012, Differentiation of intrahepatic peribiliary glands and pancreatic acinar cells from the remodeling ductal plate in human fetuses. Hepatology 56:2004–2005 Terada T, Nakanuma Y, 1995, Expression of pancreatic enzymes, α-amylase, trypsinogen and lipase, during human liver development and maturation. Gastroenterology 108:1236–1245 Terada T, Nakanuma Y, 1995, Detection of apoptosis and expression of apoptosis-related proteins during human intrahepatic bile duct development. Am J Pathol 146:67–74 Terada T, Okada Y, Nakanuma Y, 1995, Expression of matrix proteinases during human intrahepatic bile duct development: a possible role in biliary cell migration. Am J Pathol 147:1207–1213 Terada T, Nakanuma Y, 1993, Development of human peribiliary capillary plexus: a lectin-histochemical and immunohistochemical study. Hepatology 18:529–536 Terada T, Nakanuma Y, 1994, Expression of tenascin, type IV collagen and laminin during human intrahepatic bile duct development and in intrahepatic cholangiocarcinoma. Histopathology 25: 143–150 Terada T, Kato M, Horie S, Endo K, Kitamura Y, 1998, Expression of pancreatic alpha-amylase protein and messenger RNA on hilar primitive bile ducts and hepatocytes during human fetal liver organogenesis: an immunohistochemical and in situ hybridization study. Liver 18:313–319 Terada T, Ukita Y, Ueyama J, Ohta T, 2000, Protein expression of double- stranded RNA-activated protein kinase, PKR, in intrahepatic bile ducts in normal adult livers, fetal livers, primary biliary cirrhosis, hepatolithiasis and intrahepatic cholangiocarcinoma. Liver 20:450–457 Terada T, Ohta T, Nakanuma Y, 1994, Expression of transforming growth factor-α and its receptor during human liver development and maturation. Virchows Arch 424:669–675 Terada T, 2013, Human fetal ductal plate revisited: II. MUC1, MUC5AC, and MUC6 are expressed in human fetal ductal plate and MUC1 is expressed also in remodeling ductal plate, remodeled ductal plate and mature bile ducts of human fetal livers. Int J Clin Exp Pathol 6:571–585 Terada T, 2013, Smooth muscles and stem cells of embryonic guts express KIT, PDGFRRA, CD34 and many other stem cell antigens: suggestion that GISTarise from smooth muscles and gut stem cells. Int J Clin Exp Pathol 6:1038–1045 Terada T, 2013, Ductal plate in hepatoblasts in human fetal livers: I. Ductal plate-like structures with cytokeratins 7 and 19 are occasionally seen within human fetal hepatoblasts. Int J Clin Exp Pathol 6: 889–896 Terada T, 2015, Ontogenic development of nerve fibers in human fetal livers: an immunohistochemical study using neural cell adhesion molecule, NCAM, and neuron-specific enolase, NSE). Histochem Cell Biol 143:421–429 Terada T, 2014, Development of extrahepatic bile duct excluding gall bladder in human fetuses: histological, histochemical, and immunohistochemical analysis. Microsc Res Tech 77:832–840 Terada T, 2014, Human fetal ductal plate revisited. I. Ductal plate expresses NCAM, KIT, MET, PDGFRA, and neuroendocrine antigens, NSE, chromogranin, synaptophysin, and CD56). Microsc Res Tech 77:814–824 Terada T, 2013, What are ductal plate and ductal plate malformations of human livers? Liver Int 33:1294 Terada T, Nakanuma Y, Ohta G, 1987, Glandular elements around the intrahepatic bile ducts inman: theirmorphology and distribution in normal livers. Liver 7:1–8 Terada T, Nakanuma Y, 1987, Solitary cystic dilation of the intrahepatic bile duct: morphology of two autopsy cases and a review of the literature. Am J Gastroenterol 82:1301–1305 Terada T, Nakanuma Y, 1988, Morphological examination of intrahepatic bile ducts in hepatolithiasis. Virchows Arch A 413: 167–176 Terada T, Nakanuma Y, Kakita A, 1990, Pathologic observations of intrahepatic peribiliary glands in 1,000 consecutive autopsy livers: heterotopic pancreas in the liver. Gastroenterology 98:1333–1337 Terada T, Nakanuma Y, 1990, Pathological observations of intrahepatic peribiliary glands in 1,000 consecutive autopsy livers: II. A possible source of cholangiocarcinoma. Hepatology 12:92–97 Terada T, Nakanuma Y, 1990, Pathological observations of intrahepatic peribiliary glands in 1,000 consecutive autopsy livers: III. Survey of necroinflammation and cystic dilatation. Hepatology 12:1229–1233 Terada T, Nakanuma Y, 1992, Pathologic observations of intrahepatic peribiliary glands in 1,000 consecutive autopsy livers: IV. Hyperplasia of intramural and extramural glands. Hum Pathol 23:483–490 Nakanuma Y, Sato Y, Ikeda H, Harada K, Kobayashi M, Sano K, Uehara T, Yamamoto M, Arrizumi S, Park YN, Choi JH, Yu E, 2012, Intrahepatic cholangiocarcinoma with predominant “ductal plate malformation” pattern: a new subtype. Am J Surg Pathol 36: 1629–1635 Theise ND, Nakashima O, Park YN, Nakanuma Y, 2010, Combined hepatocellular-cholangiocellular carcinoma. In: Bosman FT, Carneiro F, Hruban RH, Theise ND, eds, WHO classification of tumours of the digestive system. IARC press, Lyon, pp 225–227 Desmet VJ, 2011, Ductal plates in hepatic ductular reactions: hypothesis and implications. I. Types of ductular reaction reconsidered. Virchows Arch 458:251–259 Terada T, 2011, Hepatic nodular hamartoma containing liver cysts, ductal plate malformations and peribiliary glands. Hepatol Res 41: 93–98 Terada T, 2012, Projected focal nodular hyperplasia, FNH, of the liver with pronounced atypical ductular reaction resembling ductal plate and expressing KIT. Hepatol Res 42:721–726 Terada T, Moriki T, 2010, Monolobar ductal plate malformation disease of the liver. Pathol Int 60:407–412 Terada T,Moriki T, 2011)Monolobar hepatobiliary fibropolycystic disease. Pathol Oncol Res 17:159–165 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 189 EP 194 DI 10.1007/s12253-017-0201-y PG 6 ER PT J AU Warth, A AF Warth, Arne TI cR and pR: The Residual Tumor Classification Revisited SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Warth, Arne] University of Heidelberg, Department of Pathology, INF 224, D-69120 Heidelberg, Germany. RP Warth, A (reprint author), University of Heidelberg, Department of Pathology, D-69120 Heidelberg, Germany. EM arne.warth@med.uni-heidelberg.de CR American Joint Committee on Cancer Statging and End Results Reporting, 1978). Manual for Staging of Cancer, 1st edn., reprint, Lippincott, Philadelphia Wittekind C, Compton CC, Greene FL et al, 2002, TNM residual tumor classification revisited. Cancer 94:2511–2516 American Joint Committee on Cancer, 1983, Manual for staging of cancer, 2nd edn. Lippincott, Philadelphia American Joint Committee on Cancer, 1988, Manual for staging of cancer, 3rd edn. Lippincott, Philadelphia Wittekind C, Compton C, Quirke P et al, 2009, A uniform residual tumor, R, classification: integration of the R classification and the circumferential margin status. Cancer 115:3483–3488 Cancer IUA, 1987, TNM classification of malignant tumours, 4th edn. Springer, Berlin Cancer IUA, 1992, TNM classification of malignant tumours, 4th edn. 2nd revision. Springer, Berlin AJCC, 1992, American Joint Committee on cancer manual for staging of cancer, 4th edn. Lippincott, Philadelphia AJCC, 1997, American Joint Committee on cancer manual for staging of cancer, 5th edn. Lippincott, Philadelphia Hoffmann H, Junker K, Kugler C et al, 2016, Application and interpretation of the R classification for lung cancer : results of a survey of certified lung cancer centers. Pathologe 37:258–268 Puppa G, Bortolasi L, Colombari R et al, 2011, Residual tumor, R, classification in colorectal cancer: reduced, expanded, or not uniform? Arch Pathol Lab Med 135:288 author reply 289 Wittekind C, 2015, New aspects for staging and grading of tumor metastases. Pathologe 36(Suppl 2):153–157 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2018 VL 24 IS 1 BP 195 EP 196 DI 10.1007/s12253-017-0221-7 PG 2 ER PT J AU Zidi, S AF Zidi, Sabrina TI RETRACTED ARTICLE: Impact of Toll-Like Receptors 2/3/4/9, IL-1-α/β and TNF-α Polymorphisms in Cervical Cancer Susceptibility in Tunisia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article AB This article published in Pathology and Oncology Research, has been retracted at the request of Editor-in-Chief and the authors Hasibe Verdi, Yaprak Yilmaz-Yalcin, A. C. Yazici and Fatma- Belgin Atac. The reasons for the retraction of the article are: – There are substantial parts in the article that were previously published in the article: "Involvement of Toll-like receptors in cervical cancer susceptibility among Tunisian women" by Sabrina Zidi, Hasibe Verdi, Yaprak Yilmaz- Yalcin, A.C. Yazici, Ezzedine Gazouani, Amel Mezlini, Fatma-Belgin Atac, Besma Yacoubi-Loueslati, Bulletin du Cancer, 2014; 101: E31-E35, DOI: 10.1684/bdc.2014. 2037. Although the article published in Pathology and Oncology Research uses a larger control group the two papers have very similar content, addressing the same problem with the same methodology and the differences in the articles were considered too minor. – In addition, this article was submitted to Pathology and Oncology Research without the knowledge or consent of all the authors. C1 [Zidi, Sabrina] El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 1092 Tunis, Tunisia. RP Zidi, S (reprint author), El Manar University, Faculty of Sciences of Tunis, Laboratory of Micro-Organisms and Active Biomolecules, 1092 Tunis, Tunisia. EM ZIDISABRINA86@gmail.com NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 197 EP 197 DI 10.1007/s12253-014-9793-7 PG 1 ER PT J AU Schneider, T Rosta, A Losonczy, H Radvanyi, G Ujj, Gy Egyed, M Illes, Jakucs, J Szerafin, L Gasztonyi, Z Masszi, T Ivanyi, J Demeter, J Dombi, P Toth, A Borbenyi, Z AF Schneider, Tamas Rosta, Andras Losonczy, Hajna Radvanyi, Gaspar Ujj, Gyorgy Egyed, Miklos Illes, Arpad Jakucs, Janos Szerafin, Laszlo Gasztonyi, Zoltan Masszi, Tamas Ivanyi, Janos Demeter, Judit Dombi, Peter Toth, Antal Borbenyi, Zita TI Efficacy and Tolerability of a 2-Year Rituximab Maintenance Therapy in Patients with Advanced Follicular Lymphoma after Induction of Response with Rituximab-Containing First Line-Regimens (HUSOM Study) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Follicular lymphoma; Rituximab; Maintenance; Event-free survival; Drug safety ID Follicular lymphoma; Rituximab; Maintenance; Event-free survival; Drug safety AB Follicular lymphoma is a lymphoid malignancy commonly showing slow progression which makes the treatment of the disease challenging. Rituximab monotherapy and rituximab added to standard chemotherapy has been proven to increase survival among patients with advanced stage of the disease. However, the benefit of a rituximab maintenance therapy after induction was still unclear at the time of the initiation of this study. HUSOM was a phase III open-label, single-arm, multi-centre study aimed to assess the efficacy and the safety of the 12 cycles of rituximab (375 mg/m2 every 8 weeks) maintenance therapy in patients had already presented partial or complete response to R-CVP or R-CHOP. Efficacy endpoints such as event-free survival and overall survival were estimated. Adverse events were recorded during the entire course of the study. A total number of 124 patients were enrolled by 15 Hungarian study sites. Out of these, 86 patients received 12 cycles of rituximab and 69 patients completed the 3-year follow-up phase as well. The probabilities of the event free survival and progression at 4.3 years were estimated to be 70.3% and 74.4%, respectively. The overall and the disease free survival at 4 years were estimated to be 90.7%and 87.9%, respectively. A total number of 85 adverse events were reported during the study out of which 5 AEs were considered to be related to the administration of rituximab. Analyses of the efficacy variables have revealed comparable results to those reported by controlled clinical trials (EORTC 20981, PRIMA) conducted in parallel with the HUSOM study. C1 [Schneider, Tamas] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. [Rosta, Andras] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. [Losonczy, Hajna] University of Pecs, I. Department of Internal Medicine, Ifjusag ut 13, 7624 Pecs, Hungary. [Radvanyi, Gaspar] Borsod-Abauj-Zemplen County Hospital, 2nd Department of Internal Medicine, Csabai kapu 9-11, 3529 Miskolc, Hungary. [Ujj, Gyorgy] Jasz-Nagykun-Szolnok County Hetenyi Geza Hospital, I. Department of Internal Medicine, Toszegi ut 21, 5000 Szolnok, Hungary. [Egyed, Miklos] Kaposi Mor Hospital, Department of Internal Medicine and Hematology, Tallian Gyula u. 20-34, 7400 Kaposvar, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Jakucs, Janos] Bekes County Pandy Kalman Hospital, Department of Internal Medicine, Semmelweis u. 1, 5700 Gyula, Hungary. [Szerafin, Laszlo] Szabolcs-Szatmar-Bereg County Josa Andras Hospital, Department of Haematology, Szent Istvan u. 68, 4400 Nyiregyhaza, Hungary. [Gasztonyi, Zoltan] Petz Aladar County Hospital, II. Department of Internal Medicine and Hematology, Vasvari Pal u. 2-4, 9024 Gyor, Hungary. [Masszi, Tamas] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Nagyvarad ter 1, 1097 Budapest, Hungary. [Ivanyi, Janos] Vas County Markusovszky Hospital, Department of Hematology, Markusovszky L. u. 5, 9700 Szombathely, Hungary. [Demeter, Judit] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor u. 2/A, 1083 Budapest, Hungary. [Dombi, Peter] Szent Borbala Hospital, Department of Hematology, Dozsa Gyorgy ut 77, 2800 Tatabanya, Hungary. [Toth, Antal] Tolna County Balassa Janos Hospital, IV. Department of Internal Medicine, Beri Balogh Adam u. 5-7, 7100 Szekszard, Hungary. [Borbenyi, Zita] University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, Koranyi Fasor 6, 6720 Szeged, Hungary. RP Schneider, T (reprint author), National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, 1122 Budapest, Hungary. EM schneidertamas@freemail.hu CR Greenhalgh J, Bagust A, Boland A et al, 2013, Rituximab for the first-line maintenance treatment of follicular non-Hodgkin's lymphoma: a NICE single technology appraisal. PharmacoEconomics 31(5):403–413 Gallagher CJ, Gregory WM, Jones AE et al, 1986, Follicular lymphoma: prognostic factors for response and survival. J Clin Oncol 4(10):1470–1480 Piro LD, White CA, Grillo-Lopez AJ et al, 1990, Extended rituximab, anti-CD20 monoclonal antibody, therapy for relapsed or refractory low-grade or follicular non-Hodgkin’s lymphoma. Ann Oncol 10:655–661 Tedder TF, Engel P, 1994, CD20: a regulator of cell-cycle progression of B lymphocytes. Immunol Today 15:450–454 Reff ME, Carner K, Chambers KS, 1994, Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood 83:435–445 Maloney D, Smith B, Appelbaum F, 1996, The anti-tumor effect of monoclonal anti-CD20 antibody, MAB, therapy includes direct anti-proliferative activity and induction of apopthosis in CD20 positive non-Hodgkin’s lymphoma, NHL, cell lines. Blood 88:637a Maloney DG, Liles TM, Czerwinski DK et al, 1994, Phase I clinical trial using escalating single-dose infusion of chimeric anti- CD20 monoclonal antibody, IDEC-C2B8, in patients with recurrent B-cell lymphoma. Blood 84:2457–2466 McLaughlin P, Grillo-Lopez AJ, Link BK et al, 1998, Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J Clin Oncol 16:2825–2833 Czuczman MS, Grillo-Lopez AJ,White CA et al, 1999, Treatment of patients with low-grade B-cell lymphoma with the combination of chimeric anti-CD20 monoclonal antibody and CHOP chemotherapy. J Clin Oncol 17:268–276 Czuczman MS, Fallon A, Mohr A et al, 2001, Phase II study of rituximab plus fludarabine in patients, pts, with low-grade lymphoma, LGL): final report. ASH 2001 Abstr no.: 2518 Hiddemann W, Dreyling MH, Forstpointner R et al, 2003, Combined Immuno-chemotherapy, R-CHOP, significantly improves time to treatment failure in first line therapy of follicular lymphoma results of a prospective randomized trial of the German low grade lymphoma study group, GLSG). ASH Abstr no.: 352 Marcus R, Imrie K, Belch A et al, 2003, An international multicentre, randomized, open-label, phase III trial comparing rituximab added to CVP chemotherapy to CVP chemotherapy alone in untreated stage III/IV follicular non-Hodgkins lymphoma. ASH Abstr no.: 87 Dreyling MH, Forstpointner R, Repp R et al, 2003, Combined Immuno-chemotherapy, R-FCM, results in superior remission and survival rates in recurrent follicular and mantle cell lymphoma final results of a prospective randomized trial of the German low grade lymphoma study group, GLSG). ASH Abstr. No.: 351 McLaughlin P, Rodriguez MA, Hagemeister FB et al, 2003, Stage IV indolent lymphoma: a randomized study of concurrent vs. sequential use of FND chemotherapy, fludarabine, mitoxantrone, dexamethasone, and rituximab, R, monoclonal antibody therapy, with interferon maintenance. Proc am Soc Clin Oncol 22:564 Vitolo U, Boccomini C, Ladetto M et al, 2002, High clinical and molecular response rate in elderly patients with advanced stage follicular lymhoma treated at diagnosis with a brief chemoimmunotherapy FND + rituximab. ASH Abstr no.: 1392 Gregory SA, Venugopal P, Adler S et al, 2003, A phase II study of fludarabine phosphate and mitoxantrone followed by anti-CD20 monoclonal antibody in the treatment of patients with newly diagnosed, advanced low-grade non-Hodgkin′s lymphoma, LGNHL): interim results ASH Abstr no.:1499 Czuczman MS, Koryzna A, Mohr A et al, 2005, Rituximab in combination with fludarabine chemotherapy in low-grade or follicular lymphoma. J Clin Oncol 2005 23(4):694–704 Maloney DG, Grillo-Lopez AJ, Bodkin DJ et al, 1997, IDECC2B8: results of a phase I multiple-dose trial in patients with relapsed non-Hodgkin’s lymphoma. J Clin Oncol 15:3266–3274 Maloney DG, Grillo-Lopez AJ,White CA et al, 1997, IDEC-C2B8, rituximab, anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin’s lymphoma. Blood 90: 2188–2195 Davis TA, Grillo-Lopez AJ,White CA et al, 2000, Rituximab anti- CD20 monoclonal antibody therapy in non-Hodgkin’s lymphoma: safety and efficacy of re-treatment. J Clin Oncol 18:3135–3143 Hainsworth JD, Litchy S, Morrissey L et al, 2003, Rituximab as first-line and maintenance therapy for indolent non-Hodgkin′s lymphoma, NHL): long-term follow-up of a minnie pearl cancer research network phase II trial. ASH Abstr no.: 1496 Ghielmini M, Schmitz SFH, Cogliatti SB et al, 2004, Prolonged treatment with rituximab in patients with follicular lymphoma significantly increases event-free survival and response duration compared with the standard weekly x 4 schedule. Blood 103(12):4416– 4423 Hochster H, Weller E, Gascoyne RD et al, 2009, Maintenance rituximab after cyclophosphamide, vincristine, and prednisone prolongs progression-free survival in advanced indolent lymphoma: results of the randomized phase III ECOG1496 study. J Clin Oncol 2009 27(10):1607–1614 van Oers MH, Klasa R, Marcus RE et al, 2006, Rituximab maintenance improves clinical outcome of relapsed/resistant follicular non-Hodgkin lymphoma in patients both with and without rituximab during induction: results of a prospective randomized phase 3 intergroup trial. Blood 108(10):3295–3301 Salles GL, Seymour JF, Offner F et al, 2011, Rituximab maintenance for 2 years in patients with high tumour burden follicular lymphoma responding to rituximab plus chemotherapy, PRIMA): a phase 3, randomised controlled trial. Lancet 377(9759):42–51 van Oers MH, Van Glabbeke M, Giurgea L et al, 2010, Rituximab maintenance treatment of relapsed/resistant follicular non- Hodgkin's lymphoma: long-term outcome of the EORTC 20981 phase III randomized intergroup study. J Clin Oncol 28(17):2853– 2858 Salles GL, Seymour JF, Feugier P et al, 2013, Updated 6 year follow-up of the PRIMA study confirms the benefit of 2-year rituximab maintenance in follicular lymphoma patients responding to frontline Immunochemotherapy. Blood 122(21):509 Dreyling M, Ghielmini M, Rule S et al, 2016, ESMO guidelines committee. Newly diagnosed and relapsed follicular lymphoma: ESMO clinical practice guidelines for diagnosis, treatment and follow- up. Ann Oncol 27(suppl 5):v83–v90 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 199 EP 205 DI 10.1007/s12253-017-0234-2 PG 7 ER PT J AU Malik, ASh Khan, SM Dar, M Mahboob, UH Shah, AM Shafi, MSh Mudassar, S AF Malik, A Showkat Khan, S Mosin Dar, Majeed Mahboob, Ul Hussain Shah, A Mohammad Shafi, M Sheikh Mudassar, Syed TI Molecular Alterations and Expression Dynamics of LATS1 and LATS2 Genes in Non-Small-Cell Lung Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Survival; Hazard ratio; Non-small cell lung cancer; LATS1; LATS2 ID Lung cancer; Survival; Hazard ratio; Non-small cell lung cancer; LATS1; LATS2 AB Large tumor suppressor (LATS) is an important member of the Hippo pathway which can regulate organ size and cell proliferation. However, very little is known about the expression and clinical significance of LATS in lung cancer especially from this part of the world. We elucidated the frequency of LATS1 &LATS2 promoter hypermethylation (by methylation-specific PCR) and expression (by real-time PCR) in sixty nine (n = 69) Non-Small Cell Lung Cancer (NSCLC) patients and their corresponding normal lung tissue samples.We found promoter hypermethylation frequencies of LATS1 & LATS1to be 66.66% (46/69) and 71% (49/69) in NSCLC tissues. Decreased LATS1 & LATS2 mRNA expression was found in 55% and 66.66% of NSCLC patients. The LATS1 mRNA expression was significantly higher in normal lung tissues. Also, the mRNA levels of LATS1 and LATS2 NSCLC tissues with hypermethylation were significantly lower. Multivariable analysis confirmed that LATS1 under expression increased the hazard of death after adjusting for other clinicopathological factors. Importantly, the loss of LATS1 mRNA expression was associated with overall short survival. LATS1 is an independent prognostic factor and may play an important role in NSCLC progression and may serve as a novel therapeutic target of NSCLC. C1 [Malik, A Showkat] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, Soura, 190011 Srinagar, Kashmir, India. [Khan, S Mosin] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, Soura, 190011 Srinagar, Kashmir, India. [Dar, Majeed] Sher-I-Kashmir Institute of Medical Sciences, Department of Cardiovascular and Thoracic Surgery, Soura, 190011 Srinagar, Kashmir, India. [Mahboob, Ul Hussain] University of Kashmir, Department of Biotechnology, 190006 Srinagar, Kashmir, India. [Shah, A Mohammad] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, Soura, 190011 Srinagar, Kashmir, India. [Shafi, M Sheikh] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, Soura, 190011 Srinagar, Kashmir, India. [Mudassar, Syed] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, Soura, 190011 Srinagar, Kashmir, India. RP Mudassar, S (reprint author), Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Srinagar, India. EM syed.mudassar@skims.ac.in CR Travis WD, 2011, Pathology of lung cancer. Clin Chest Med 32: 669–692 World Health Organization, 2012, Cancer, http://www.who.int/ cancer/en/) Travis LB, Gospodarowicz M, Curtis RE et al, 2002, Lung cancer following chemotherapy and radiotherapy for Hodgkin’s disease. J Natl Cancer Inst 94:182–192 O'Mahony D, Kummar S, Gutierrez ME, 2005, Non-small-cell lung cancer vaccine therapy: a concise review. J Clin Oncol 23: 9022–9028 Nanda KA, 2001, Consolidated report of the population based cancer registries, incidence & distribution of cancer, 1990–1996. Indian Council of Medical research, National Cancer Registry Program, New Delhi Pandith AA, Siddiqi MA, 2012, Burden of cancers in the valley of Kashmir: 5Year epidemiological study reveals a different scenario. Tumor Biol 33:1629–1637 Zheng T, Wang J, Jiang H, Liu L, 2011, Hippo signaling in oval cells and hepato-carcinogenesis. Cancer Lett 302:91–99 Halder G, Johnson RL, 2011, Development 138:9–22 Xia H, Qi H, Li Y, Pei J, Barton J, Blackstad M, Xu T, Tao W, 2002, LATS1 tumor suppressor regulates G2/M transition and apoptosis. Oncogene 21:1233–1241 TaoW, Zhang S, Turenchalk GS et al, 1999, Human homologue of the Drosophila melanogaster LATS tumour suppressor modulates CDC2 activity. Nat Genet 21:177–181 Avruch J, Zhou D, Bardeesy N, 2012, YAP oncogene overexpression supercharges colon cancer proliferation. Cell Cycle 11:1090– 1096 Hisaoka M, Tanaka A, Hashimoto H, 2002, Molecular alterations of h-warts/LATS1 tumor suppressor in human soft tissue sarcoma. Lab Investig 82:1427–1435 Matallanas D, Romano D et al, 2007, RASSF1A elicits apoptosis through an MST2 pathway directing proapoptotic transcription by the p73 tumor suppressor protein. Mol Cell 27:962–975 McPherson JP, Tamblyn L, Elia A et al, 2004, LATS2/Kpm is required for embryonic development, proliferation control and genomic integrity. EMBO J 23:3677–3688 Yabuta N, Okada N, Ito A et al, 2007, Lats2 is an essential mitotic regulator required for the coordination of cell division. J Biol Chem 282:19259–19271 Ke H, Pei J, Ni Z, Xia H, Qi H,Woods T, Kelekar A, TaoW(2004, Putative tumor suppressor LATS2 induces apoptosis through downregulation of Bcl-2 and Bcl-x(L). Exp Cell Res 298:329–338 Jiang Z, Li X, Hu J, ZhouW, Jiang Y, Li G, Lu D, 2006, Promoter hypermethylation-mediated down-regulation of LATS1 and LATS2 in human astrocytoma. Neurosci Res 56:450–458 Jimenez-Velasco A, Roman-Gomez J et al, 2005, Down-regulation of the large tumor suppressor 2, LATS/KPM, gene is associated with poor prognosis in acute lymphoblastic leukemia. Leukemia 19:2347–2350 Sasaki H, Hikosaka Y, Kawano O, Yano M, Fujii Y, 2010, Hypermethylation of thelargetumor suppressor genes in Japanese lung cancer. Oncol Lett 1:303–307 Takahashi Y, Miyoshi Y, Takahata C, Irahara N, Taguchi T, Tamaki Y, Noguchi S, 2005, Down-regulation of LATS1 and LATS2 mRNA expression by promoter hypermethylation and its association with biologically aggressive phenotype in human breast cancers. Clin Cancer Res 11:1380–1385 Bao Y, Hata Y, Ikeda M, Withanage K, 2011, Mammalian hippo pathway: from development to cancer and beyond. J Biochem 149: 361–379 Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB, 1996, Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc. Natl Acad Sci USA 93:9821–9826 Steinmann K, Sandner A, Schagdarsurengin U, Dammann RH, 2009, Frequent promoter hypermethylation of tumor-related genes in head and neck squamous cell carcinoma. Oncol Rep 22:1519– 1526 Xu-Yng L, Xiu-Peng Z, Jun-Hua W, Xue-Shan Q, En-Hua W, 2014, Expression of LATS1 contribute to good prognosis and can negatively regulate YAP oncoprotein in non-small cell lung cancer. Tumor Biol 35:6435–6443 Tianhai J, Dan L,Wei S,Wensheng Y, HaiqiaoW, Xiuwu B, 2012, Decreased exoression of LATS1 is correlated with the progression and prognosis of glioma. J Exp Clin Cancer Res 31:67–75 Wierzbicki PM, Adrych K, Kartanowicz D et al, 2013, Underexpression of LATS1 TSG in colorectal cancer is associated with promoter hypermethylation. World J Gastroenterol 19:4363– 4373 Zhou G-X, Xiao-Yu L, Qi Z et al, 2013, Effects of the hippo signaling pathway in human gastric cancer. Asian Pac J Cancer Prev 14:5199–5205 Xia H, Qi H, LiY, Pei J, Barton J, Blackstad M, Xu T, TaoW(2002, LATS1 tumor suppressor regulates G2/M transition and apoptosis. Oncogene 21:1233–1241 Yang X, Li DM, Chen W, Xu T, 2001, Human homologue of Drosophila LATS, negatively regulate growth by inducing G(2)M arrest or apoptosis. Oncogene 20:6516–6523 Liang K, Zhou G, Zhang Q, Li J, Zhang C, 2014, Expresssion of hippo pathway in colorectal cancer. Saudi J Gastroenterol 20:188– 194 Luo SY, Sit KY, Sihoe AD, Suen WS et al, 2014, Abberant large tumor suppressor 2, LATS2, gene expression correlates with EGFR mutation and survival in lung adenocarcinomas. Lung Cancer 85: 282–292 Yu T, Bachman J, Lai ZC, 2013, Evidence for a tumor suppressor role forthe large tumor suppressor genes LATS1 and LATS2 in humancancer. Genetics 195:1193–1196 Ishizaki K, Fujimoto J, Kumimoto H et al, 2002, Frequent polymorphic changes but rare tumor specific mutations of the LATS2 gene on 13q11-12 in esophageal squamous cell carcinoma. Int J Oncol 21:1053–1057 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 207 EP 214 DI 10.1007/s12253-017-0225-3 PG 8 ER PT J AU Li, Sh Li, R Wang, H Li, L Li, H Li, Y AF Li, Shuang Li, Rui Wang, Heping Li, Lisha Li, Huiyu Li, Yulin TI The Key Genes of Chronic Pancreatitis which Bridge Chronic Pancreatitis and Pancreatic Cancer Can be Therapeutic Targets SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Transcriptome; Chronic pancreatitis; Pancreatic cancer; Network analysis; Key factors ID Transcriptome; Chronic pancreatitis; Pancreatic cancer; Network analysis; Key factors AB An important question in systems biology is what role the underlying molecular mechanisms play in disease progression. The relationship between chronic pancreatitis and pancreatic cancer needs further exploration in a system view. We constructed the disease network based on gene expression data and protein-protein interaction.We proposed an approach to discover the underlying core network and molecular factors in the progression of pancreatic diseases, which contain stages of chronic pancreatitis and pancreatic cancer. The chronic pancreatitis and pancreatic cancer core network and key factors were revealed and then verified by gene set enrichment analysis of pathways and diseases. The key factors provide the microenvironment for tumor initiation and the change of gene expression level of key factors bridge chronic pancreatitis and pancreatic cancer. Some new candidate genes need further verification by experiments. Transcriptome profiling-based network analysis reveals the importance of chronic pancreatitis genes and pathways in pancreatic cancer development on a system level by computational method and they can be therapeutic targets. C1 [Li, Shuang] Ministry of Education, Jilin University, College of Basic Medical Sciences, The Key Laboratory of PathobiologyChangchun, China. [Li, Rui] Coordination Center of China, National Computer Network Emergency Response Technical TeamBeijing, China. [Wang, Heping] Tongji Medical School, Tongji Hospital, Department of NeurosurgeryWuhan, China. [Li, Lisha] Ministry of Education, Jilin University, College of Basic Medical Sciences, The Key Laboratory of PathobiologyChangchun, China. [Li, Huiyu] Ministry of Education, Jilin University, College of Basic Medical Sciences, The Key Laboratory of PathobiologyChangchun, China. [Li, Yulin] Ministry of Education, Jilin University, College of Basic Medical Sciences, The Key Laboratory of PathobiologyChangchun, China. RP Li, L (reprint author), Ministry of Education, Jilin University, College of Basic Medical Sciences, The Key Laboratory of Pathobiology, Changchun, China. EM lilisha@jlu.edu.cn CR Tiffin N, Andrade-Navarro MA, Perez-Iratxeta C, 2009, Linking genes to diseases: it's all in the data. Genome Medicine 1(8):77., DOI 10.1186/gm77 Hu JX, Thomas CE, Brunak S, 2016, Network biology concepts in complex disease comorbidities. Nat Rev Genet 17(10):615–629., DOI 10.1038/nrg.2016.87 Ryan CJ, Cimermancic P, Szpiech ZA, Sali A, Hernandez RD, Krogan NJ, 2013, High-resolution network biology: connecting sequence with function. Nat Rev Genet 14(12):865–879., DOI 10.1038/nrg3574 Gentles AJ, Gallahan D, 2011, Systems biology: confronting the complexity of cancer. Cancer Res 71(18):5961–5964., DOI 10.1158 /0008-5472.CAN-11-1569 Wang E, Lenferink A, O'Connor-McCourt M, 2007, Cancer systems biology: exploring cancer-associated genes on cellular networks. Cell. Mol. Life Sci. : CMLS 64(14):1752–1762., DOI 10.1007/s00018-007-7054-6 Alberghina L, Gaglio D, Moresco RM, Gilardi MC, Messa C, Vanoni M, 2014, A systems biology road map for the discovery of drugs targeting cancer cell metabolism. Curr Pharm Des 20(15): 2648–2666 Hunter DJ, 2005, Gene-environment interactions in human diseases. Nat Rev Genet 6(4):287–298., DOI 10.1038/nrg1578 Hyduke DR, Palsson BO, 2010, Towards genome-scale signalling network reconstructions. Nat Rev Genet 11(4):297–307., DOI 10.1038/nrg2750 Valladares-Ayerbes M, Haz-Conde M, Blanco-Calvo M, 2015, Systems oncology: toward the clinical application of cancer systems biology. Future Oncol 11(4):553–555., DOI 10.2217 /fon.14.255 Henderson D, Ogilvie LA, Hoyle N, Keilholz U, Lange B, Lehrach H, 2014, Personalizedmedicine approaches for colon cancer driven by genomics and systems biology: OncoTrack. Biotechnol J 9(9): 1104–1114., DOI 10.1002/biot.201400109 Calvano SE, Xiao W, Richards DR, Felciano RM, Baker HV, Cho RJ, Chen RO, Brownstein BH, Cobb JP, Tschoeke SK, Miller- Graziano C, Moldawer LL, Mindrinos MN, Davis RW, Tompkins RG, Lowry SF, 2005, A network-based analysis of systemic inflammation in humans. Nature 437(7061):1032–1037., DOI 10.1038/nature03985 Coffelt SB, de Visser KE, 2014, Cancer: inflammation lights the way to metastasis. Nature 507(7490):48–49., DOI 10.1038 /nature13062 Mantovani A, Allavena P, Sica A, Balkwill F, 2008, Cancer-related inflammation. Nature 454(7203):436–444., DOI 10.1038/nature07205 Grivennikov SI, Greten FR, Karin M, 2010, Immunity, inflammation, and cancer. Cell 140(6):883–899., DOI 10.1016/j. cell.2010.01.025 Lowenfels AB, Maisonneuve P, Cavallini G, Ammann RW, Lankisch PG, Andersen JR, Dimagno EP, Andren-Sandberg A, Domellof L, 1993, Pancreatitis and the risk of pancreatic cancer. International pancreatitis study group. N Engl J Med 328(20): 1433–1437 Hirsch HA, Iliopoulos D, Joshi A, Zhang Y, Jaeger SA, Bulyk M, Tsichlis PN, Shirley Liu X, Struhl K, 2010, A transcriptional signature and common gene networks link cancer with lipid metabolism and diverse human diseases. Cancer Cell 17(4):348–361., DOI 10.1016/j.ccr.2010.01.022 Stoehr PJ, Omond RA, 1989, The EMBL network file server. Nucleic Acids Res 17(16):6763–6764 Strimmer K, 2008, Fdrtool: a versatile R package for estimating local and tail area-based false discovery rates. Bioinformatics 24(12):1461–1462., DOI 10.1093/bioinformatics/btn209 Xia J, Gill EE, Hancock RE, 2015, NetworkAnalyst for statistical, visual and network-based meta-analysis of gene expression data. Nat Protoc 10(6):823–844., DOI 10.1038/nprot.2015.052 Su G, Morris JH, Demchak B, Bader GD, 2014, Biological network exploration with cytoscape 3. Curr Protoc Bioinformatics / editoral board, Andreas D Baxevanis [et al] 47:8 13 11-18 13 24., DOI 10.1002/0471250953.bi0813s47 Davis AP, Murphy CG, Johnson R, Lay JM, Lennon-Hopkins K, Saraceni-Richards C, Sciaky D, King BL, Rosenstein MC,Wiegers TC, Mattingly CJ, 2013, The comparative Toxicogenomics database: update 2013. Nucleic Acids Res 41(Database issue):D1104– D1114., DOI 10.1093/nar/gks994 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144(5):646–674., DOI 10.1016/j.cell.2011.02.013 Steele CW, Kaur Gill NA, Jamieson NB, Carter CR, 2016, Targeting inflammation in pancreatic cancer: clinical translation. World Journal of Gastrointestinal Oncology 8(4):380–388., DOI 10.4251/wjgo.v8.i4.380 Zhang J, Wang P, Ouyang H, Yin J, Liu A, Ma C, Liu L, 2013, Targeting cancer-related inflammation: Chinese herbal medicine inhibits epithelial-to-mesenchymal transition in pancreatic cancer. PLoS One 8(7):e70334., DOI 10.1371/journal.pone.0070334 Yu M, Ting DT, Stott SL,Wittner BS, Ozsolak F, Paul S, Ciciliano JC, Smas ME, Winokur D, Gilman AJ, Ulman MJ, Xega K, Contino G, Alagesan B, Brannigan BW, Milos PM, Ryan DP, Sequist LV, Bardeesy N, Ramaswamy S, Toner M, Maheswaran S, Haber DA, 2012, RNA sequencing of pancreatic circulating tumour cells implicates WNT signalling in metastasis. Nature 487(7408):510–513., DOI 10.1038/nature11217 Liang C, Li Y, Luo J, Zhang Z, 2015, A novel motif-discovery algorithm to identify co-regulatory motifs in large transcription factor and microRNA co-regulatory networks in human. B i o i n f o r m a t i c s 3 1, 1 4 , : 2 3 4 8 – 2 3 5 5 . d o i :1 0 . 1 0 9 3 /bioinformatics/btv159 del Sol A, Balling R, Hood L, Galas D, 2010, Diseases as network perturbations. Curr Opin Biotechnol 21(4):566–571., DOI 10.1016/j. copbio.2010.07.010 Yang Z, Zheng R, Gao Y, Zhang Q, 2014, Gene expression profiles on predicting protein interaction network and exploring of new treatments for lung cancer. Mol Biol Rep 41(12):8203–8210., DOI 10.1007/s11033-014-3722-4 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 215 EP 222 DI 10.1007/s12253-017-0217-3 PG 8 ER PT J AU Liu, Y Wang, W Li, Y Sun, F Lin, J Li, L AF Liu, Yansong Wang, Wei Li, Yan Sun, Feifei Lin, Jiaxiang Li, Li TI CKS1BP7, a Pseudogene of CKS1B, is Co-Amplified with IGF1R in Breast Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; CKS1BP7; IGF1R; Quantitative multi-gene fluorescence in situ hybridization ID Breast cancer; CKS1BP7; IGF1R; Quantitative multi-gene fluorescence in situ hybridization AB Pseudogenes have been reported to exhibit functional roles. Amplification or overexpression of CDC28 protein kinase regulatory subunit 1B (CKS1B) was found in various human cancers. But it was known little about CKS1B pseudogene 7 (CKS1BP7), a pseudogene sharing considerable sequence identity with CKS1B. The aim of this study was to evaluate copy number alterations (CNAs) of CKS1BP7 and address its potential roles in breast cancer. We detected copy numbers of CKS1BP7 and insulin-like growth factor 1 receptor (IGF1R) using quantitative multigene fluorescence in situ hybridization (QM-FISH) technique, compared their status in both invasive carcinoma and ductal carcinoma in situ (DCIS) components within the same tumors, and investigated the associations of CNAs with tumor features and patients outcomes. Amplification of CKS1BP7 (dot-like pattern) was found in 28.8% of all cases, while amplified IGF1R (cluster pattern) was identified in 24.2%of all patients. The two events often co-existed (p = 0.01). Within the same tumors, identical CNAs of CKS1BP7 and IGF1R were found in DCIS and invasive carcinoma. Moreover, amplification of both genes was more frequent in aneuploidy tumors and the tumors with high ki67, but wasn’t associated with patients’ outcome. In summary, CKS1BP7 amplification is a frequent event in breast cancer and often co-occurs with amplified IGF1R, which provides evidence supporting the interactions between CKS1BP7 and IGF1R during mammary carcinogenesis. Our findings suggest that CKS1BP7 as well as IGF1R may serve as potential biomarkers for early detection and predict prognosis in breast cancer. C1 [Liu, Yansong] Shandong Cancer Hospital Affiliated to Shandong University, Department of Breast Surgery, 250117 Jinan, Shandong, China. [Wang, Wei] Shandong Provincial Western Hospital, Department of Obstetrics and Gynecology, 250022 Jinan, Shandong, China. [Li, Yan] Shandong Cancer Hospital and Institute Affiliated to Shandong University, Department of Medical Oncology, 250117 Jinan, Shandong, China. [Sun, Feifei] The Second Hospital of Shandong University, Department of Pathology, 44#Wenhuaxi Road, 250012 Jinan, Shandong, China. [Lin, Jiaxiang] The Second Hospital of Shandong University, Department of Pathology, 44#Wenhuaxi Road, 250012 Jinan, Shandong, China. [Li, Li] The Second Hospital of Shandong University, Department of Pathology, 44#Wenhuaxi Road, 250012 Jinan, Shandong, China. RP Li, L (reprint author), The Second Hospital of Shandong University, Department of Pathology, 250012 Jinan, China. EM lillie6636@sdu.edu.cn CR Negrini S, Gorgoulis VG, Halazonetis TD, 2010, Genomic instability–an evolving hallmark of cancer. Nat Rev Mol Cell Biol 11(3):220–228., DOI 10.1038/nrm2858 Sieber OM, Heinimann K, Tomlinson IP, 2003, Genomic instability–the engine of tumorigenesis? Nat Rev Cancer 3(9): 701–708., DOI 10.1038/nrc1170 Simpson PT, Reis-Filho JS, Gale T, Lakhani SR, 2005, Molecular evolution of breast cancer. J Pathol 205(2):248– 254., DOI 10.1002/path.1691 Zhou BP, Hung MC, 2003, Dysregulation of cellular signaling by HER2/neu in breast cancer. Semin Oncol 30(5 Suppl 16):38–48 Saito S, Morita K, Hirano T, 2009, High frequency of common DNA copy number abnormalities detected by bacterial artificial chromosome array comparative genomic hybridization in 24 breast cancer cell lines. Hum Cell 22(1):1–10., DOI 10.1111/j.1749-0774. 2008.00061.x Smid M, Hoes M, Sieuwerts AM, Sleijfer S, Zhang Y, Wang Y, Foekens JA, Martens JW, 2011, Patterns and incidence of chromosomal instability and their prognostic relevance in breast cancer subtypes. Breast Cancer Res Treat 128(1):23–30., DOI 10.1007/ s10549-010-1026-5 Gogas H, Kotoula V, Alexopoulou Z, Christodoulou C, Kostopoulos I, Bobos M, Raptou G, Charalambous E, Tsolaki E, Xanthakis I, Pentheroudakis G, Koutras A, Bafaloukos D, Papakostas P, Aravantinos G, Psyrri A, Petraki K, Kalogeras KT, Pectasides D, Fountzilas G, 2016, MYC copy gain, chromosomal instability and PI3K activation as potentialmarkers of unfavourable outcome in trastuzumab-treated patients with metastatic breast cancer. J Transl Med 14(1):136., DOI 10.1186/s12967-016-0883-z Ertych N, Stolz A, Valerius O, Braus GH, Bastians H, 2016, CHK2-BRCA1 tumor-suppressor axis restrains oncogenic aurora-a kinase to ensure proper mitotic microtubule assembly. Proc Natl Acad Sci U S A 113(7):1817–1822., DOI 10.1073/pnas.1525129113 Demetrick DJ, Zhang H, Beach DH, 1996, Chromosomal mapping of the human genes CKS1 to 8q21 and CKS2 to 9q22. Cytogenet Cell Genet 73(3):250–254 Wang XC, Tian J, Tian LL, Wu HL, Meng AM, Ma TH, Xiao J, Xiao XL, Li CH, 2009, Role of Cks1 amplification and overexpression in breast cancer. Biochem Biophys Res Commun 379(4): 1107–1113., DOI 10.1016/j.bbrc.2009.01.028 Khattar V, Thottassery JV, 2013, Cks1: structure, emerging roles and implications in multiple cancers. J Cancer Ther 4(8):1341– 1354., DOI 10.4236/jct.2013.48159 Zhan F, Colla S, Wu X, Chen B, Stewart JP, Kuehl WM, Barlogie B, Shaughnessy JD Jr, 2007, CKS1B, overexpressed in aggressive disease, regulates multiple myeloma growth and survival through SKP2- and p27Kip1-dependent and -independent mechanisms. Blood 109(11):4995–5001., DOI 10.1182/blood-2006-07-038703 Lu Y, Zi X, Pollak M, 2004, Molecular mechanisms underlying IGF-I-induced attenuation of the growth-inhibitory activity of trastuzumab, Herceptin, on SKBR3 breast cancer cells. Int J Cancer 108(3):334–341., DOI 10.1002/ijc.11445 Liu QD, Hirohashi Y, Du XL, Greene MI, Wang Q, 2010, Nek2 targets the mitotic checkpoint proteins Mad2 and Cdc20: a mechanism for aneuploidy in cancer. ExpMol Pathol 88(2):225–233., DOI 10.1016/j.yexmp.2009.12.004 Welch JD, Baran-Gale J, Perou CM, Sethupathy P, Prins JF, 2015, Pseudogenes transcribed in breast invasive carcinoma show subtype-specific expression and ceRNA potential. BMC Genomics 16:113., DOI 10.1186/s12864-015-1227-8 Lakhani SR, Ellis IO, Schnitt SJ, Schnitt SJ, Tan PH, van de Vijver MJ, 2012, WHO classification of tumours of the breast, 4th edition). IARC, Lyon, p 19 Blegen H, Einhorn N, Sjovall K, Roschke A, Ghadimi BM, McShane LM, Nilsson B, Shah K, Ried T, Auer G, 2000, Prognostic significance of cell cycle proteins and genomic instability in borderline, early and advanced stage ovarian carcinomas. Int J Gynecol Cancer 10(6):477–487 Hammond ME, Hayes DF, Wolff AC, Mangu PB, Temin S, 2010, American society of clinical oncology/college of american pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Oncol Pract 6(4):195–197., DOI 10.1200/JOP.777003 Li L,Mu K, Zhou G, Lan L, Auer G, Zetterberg A, 2008, Genomic instability and proliferative activity as risk factors for distant metastases in breast cancer. Br J Cancer 99(3):513–519., DOI 10.1038/sj. bjc.6604479 Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JM, Bilous M, Fitzgibbons P, Hanna W, Jenkins RB, Mangu PB, Paik S, Perez EA, Press MF, Spears PA, Vance GH, Viale G, Hayes DF, American Society of Clinical O, College of American P, 2014, Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. Arch Pathol Lab Med 138, 2):241–256., DOI 10.5858/arpa.2013-0953-SA Balakirev ES, Ayala FJ, 2003, Pseudogenes: are they "junk" or functional DNA? Annu Rev Genet 37:123–151., DOI 10.1146/ annurev.genet.37.040103.103949 Moleirinho A, Seixas S, Lopes AM, Bento C, PrataMJ, Amorim A, 2013, Evolutionary constraints in the beta-globin cluster: the signature of purifying selection at the delta-globin, HBD, locus and its role in developmental gene regulation. Genome Biol Evol 5(3): 559–571., DOI 10.1093/gbe/evt029 Hindley J, Phear G, Stein M, Beach D, 1987, Sucl+ encodes a predicted 13-kilodalton protein that is essential for cell viability and is directly involved in the division cycle of Schizosaccharomyces pombe. Mol Cell Biol 7(1):504–511 Hayles J, Aves S, Nurse P, 1986, suc1 is an essential gene involved in both the cell cycle and growth in fission yeast. EMBO J 5(12): 3373–3379 Spruck C, Strohmaier H,Watson M, Smith AP, Ryan A, Krek TW, Reed SI, 2001, A CDK-independent function of mammalian Cks1: targeting of SCF(Skp2, to the CDK inhibitor p27Kip1. Mol Cell 7(3):639–650 Ganoth D, Bornstein G, Ko TK, Larsen B, Tyers M, Pagano M, Hershko A, 2001, The cell-cycle regulatory protein Cks1 is required for SCF(Skp2)-mediated ubiquitinylation of p27. Nat Cell Biol 3(3):321–324., DOI 10.1038/35060126 Bayani J, Selvarajah S, Maire G, Vukovic B, Al-Romaih K, Zielenska M, Squire JA, 2007, Genomic mechanisms and measurement of structural and numerical instability in cancer cells. Semin Cancer Biol 17(1):5–18., DOI 10.1016/j.semcancer. 2006.10.006 Almeida A,Muleris M, Dutrillaux B,Malfoy B, 1994, The insulinlike growth factor I receptor gene is the target for the 15q26 amplicon in breast cancer. Genes Chromosomes Cancer 11(1):63– 65 Farabaugh SM, Boone DN, Lee AV, 2015, Role of IGF1R in breast cancer subtypes, Stemness, and lineage differentiation. Front Endocrinol, Lausanne, 6:59., DOI 10.3389/fendo.2015.00059 Law JH, Habibi G, Hu K, Masoudi H, Wang MY, Stratford AL, Park E, Gee JM, Finlay P, Jones HE, Nicholson RI, Carboni J, Gottardis M, Pollak M, Dunn SE, 2008, Phosphorylated insulinlike growth factor-i/insulin receptor is present in all breast cancer subtypes and is related to poor survival. Cancer Res 68(24):10238– 10246., DOI 10.1158/0008-5472.CAN-08-2755 SlotkyM, ShapiraM, Ben-Izhak O, Linn S, Futerman B, Tsalic M, Hershko DD, 2005, The expression of the ubiquitin ligase subunit Cks1 in human breast cancer. Breast Cancer Res 7(5):R737–R744., DOI 10.1186/bcr1278 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 223 EP 229 DI 10.1007/s12253-017-0224-4 PG 7 ER PT J AU Zelga, P Przybylowska-Sygut, K Zelga, M Dziki, A Majsterek, I AF Zelga, Piotr Przybylowska-Sygut, Karolina Zelga, Marta Dziki, Adam Majsterek, Ireneusz TI The 116G > A MSH6 and IVS1-1121C > T PMS2 Genes Polymorphisms Modulate the Risk of the Sporadic Colorectal Cancer Development in Polish Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; SNP; MSH6; PMS2; DNA MMR ID Colorectal cancer; SNP; MSH6; PMS2; DNA MMR AB Colorectal cancer (CRC) is one of the most common cancers worldwide. DNA mismatch repair (MMR) is an evolutionarily conserved process that corrects mismatches generated during DNA replication. MMR defects were found to be associated with hereditary non-polyposis colorectal cancer (HNPCC) and a subset of sporadic colon cancers. The inheritance of common variations in MMR genes may influences individual susceptibility to the development of colorectal cancer. The purpose of the study was to evaluate the association between gene polymorphisms Glu39Gly (c.116G > A) of MSH6 gene and IVS1-1121C > T of PMS2 gene and sporadic colorectal cancer risk, in a case-control study comprising 200 patients and 200 controls origination from polish population. DNA was isolated from peripheral blood lymphocytes of enrolled patients, and gene polymorphisms were analysed by restriction fragment length polymorphism-polymerase chain reaction (RFLP-PCR) for MSH6 and TaqMan for PMS2. G/A variant of Glu39Gly (c.116G > A) genotype was associated with an increased risk of colorectal cancer (OR 1,65 95% CI:1,01–2,69 p = 0.44). Presence of A allele was also significantly higher in patient with CRC (OR 1,57 95% CI: 1,04–2,38 p = 0.032). Prevalence of this genotype was also markedly higher in females and patients above 60 years in CRC group (OR 2.25 95% CI: 1.22–4.14 p = 0.0098 and OR 2.74 95% CI: 1.27–5.93 p = 0.0097 respectively). None of such correlations was observed for genotype variants of IVS1-1121C > T PMS2. In conclusion, our data suggests that MSH6 Glu39Gly polymorphism is associated with the risk of developing sporadic colorectal cancer in polish population. Linkage to the female gender, onset above 60 years old and further increase of risk when combined with wild-type allele of PMS2 IVS1-1121C > T polymorphism indicates defective mismatch repair system. C1 [Zelga, Piotr] Medical University of Lodz, Department of General and Colorectal Surgery, Plac Hallera 1, 90-647 Lodz, Poland. [Przybylowska-Sygut, Karolina] Medical University of Lodz, Department of Chemistry and Clinical BiochemistryLodz, Poland. [Zelga, Marta] Medical University of Lodz, Department of General and Colorectal Surgery, Plac Hallera 1, 90-647 Lodz, Poland. [Dziki, Adam] Medical University of Lodz, Department of General and Colorectal Surgery, Plac Hallera 1, 90-647 Lodz, Poland. [Majsterek, Ireneusz] Medical University of Lodz, Department of Chemistry and Clinical BiochemistryLodz, Poland. RP Zelga, P (reprint author), Medical University of Lodz, Department of General and Colorectal Surgery, 90-647 Lodz, Poland. EM piotr_zelga@op.pl CR Torre LA, Bray F, Siegel RL et al, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65:87–108., DOI 10.3322/caac.21262 Romaguera D, Vergnaud A, Peeters PH et al, 2012, Is concordance with world cancer research fund/American institute for cancer research guidelines for cancer prevention related to subsequent risk of cancer ? Results from the EPIC study. Am J Clin Nutr 96:150–163., DOI 10.3945/ajcn.111.031674 Jass JR, 2007, Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology 50:113–130., DOI 10.1111/j.1365-2559.2006. 02549.x Samadder NJ, Vierkant RA, Tillmans LS et al, 2013, Associations between colorectal cancer molecular markers and pathways with clinicopathologic features in older women. Gastroenterology 145: 348–356., DOI 10.1053/j.gastro.2013.05.001 Ionov Y, Peinado MA, Malkhosyan S et al, 1993, Ubiquitous somatic mutations in simple repeatedsequences reveal a new mechanism for colonic carcinogenesis. Nature 363(6429): 558–561., DOI 10.1038/363558a0 Pritchard CC, GradyWM(2011, Colorectal cancer molecular biology moves into clinical practice. Gut 60:116–129., DOI 10.1136/gut. 2009.206250 Mori Y, Selaru FM, Sato F et al, 2003, The impact of microsatellite instability on the molecular phenotype of colorectal tumors. Cancer Res 63:4577–4582 Mrkonjic M, Roslin NM, Greenwood CM et al, 2010, Specific variants in the MLH1 gene region may drive DNA methylation, loss of protein expression, and MSI-H colorectal cancer. PLoS One 5:1–10., DOI 10.1371/journal.pone.0013314 Valle L, 2014, Genetic predisposition to colorectal cancer: where we stand and future perspectives. World J Gastroenterol 20:9828– 9849., DOI 10.3748/wjg.v20.i29.9828 Picelli S, Zajac P, Zhou XL et al, 2010, Common variants in human CRC genes as low-risk alleles. Eur J Cancer 46:1041–1048., DOI 10.1016/j.ejca.2010.01.013 Tenesa A, Dunlop MG, 2009, New insights into the aetiology of colorectal cancer from genome-wide association studies. Nat Rev Genet 10:353–358., DOI 10.1038/nrg2574 COGENT Study., Houlston RS,Webb E, Broderick P, Pittman AM, Di Bernardo MC, Lubbe S, Chandler I, Vijayakrishnan J, Sullivan K, Penegar S; Colorectal Cancer Association Study Consortium., Carvajal-Carmona L, Howarth K, Jaeger E, Spain SL, Walther A, Barclay E, Martin L, GormanM, Domingo E, Teixeira AS; CoRGI Consortium., Kerr D, Cazier JB, Niittymaki I, Tuupanen S, Karhu A, Aaltonen LA, Tomlinson IP, Farrington SM, Tenesa A, Prendergast JG, Barnetson RA, Cetnarskyj R, Porteous ME, Pharoah PD, Koessler T, Hampe J, Buch S, Schafmayer C, Tepel J, Schreiber S, Volzke H, Chang-Claude J, Hoffmeister M, Brenner H, Zanke BW, Montpetit A, Hudson TJ, Gallinger S, Campbell H, Dunlop MG. Meta-analysis of genome-wide association data identifies four new susceptibility loci for colorectal cancer. Nat Genet 40:1426–1435., DOI 10.1038/ng.262 Wu Y, Berends MJ,Mensink RG et al, 1999, Association of hereditary nonpolyposis colorectal cancer-related tumors displaying low microsatellite instability with MSH6 germline mutations. Am J Hum Genet 65:1291–1298., DOI 10.1086/302612 Parc YR, Halling KC, Wang L et al, 2000, MSH6 alterations in patients withmicrosatellite instability-low colorectal cancer. Cancer Res 60:2225–2231 Peltomaki P, 2001, Deficient DNA mismatch repair: a common etiologic factor for colon cancer. Hum Mol Genet 10:735–740., DOI 10.1093/hmg/10.7.735 Shamekh R, Cives M, Mejia J et al, 2015, Higher Frequency of PMS2 loss in colorectal tumors in Colombian population. FASEB J 29:926.18 Gill S, Lindor NM, Burgart LJ et al, 2005, Isolated loss of PMS2 expression in colorectal cancers: Frequency, patient age, and familial aggregation. Clin Cancer Res 11:6466–6471., DOI 10.1158/1078- 0432.CCR-05-0661 Shia J, Tang LH, Vakiani E et al, 2009, Immunohistochemistry as first-line screening for detecting colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome: a 2-antibody panel may be as predictive as a 4-antibody panel.AmJ Surg Pathol. 2009 Nov;33(11):1639-45., DOI 10.1097/PAS.0b013e3181b15aa2. Erratum in: Am J Surg Pathol. 2010 Mar;34(3):432 Bae JM, Kim JH, Kang GH, 2016, Molecular subtypes of colorectal cancer and their clinicopathologic features, with an emphasis on the serrated neoplasia pathway. Arch Pathol Lab Med 140:406– 412., DOI 10.5858/arpa.2015-0310-RA Koessler T, Azzato EM, Perkins B et al, 2009, Common germline variation in mismatch repair genes and survival after a diagnosis of colorectal cancer. Int J Cancer 124:1887–1891., DOI 10.1002/ijc.24120 Morris EJA, Penegar S, Whiffin N et al, 2015, A retrospective observational study of the relationship between single nucleotide polymorphisms associated with the risk of developing colorectal cancer and survival. PLoS One 10:1–11., DOI 10.1371/journal.pone.0117816 Papadopoulos N, Nicolaides NC, Liu B et al, 1995, Mutations of GTBP in genetically unstable cells. Science 268:1915–1917 Kunkel TA, Erie DA, 2005, Dna mismatch repair*. Annu Rev Biochem 74:681–710., DOI 10.1146/annurev.biochem.74.082803. 133243 Berndt SI, Platz EA, Fallin MD et al, 2007, Mismatch repair polymorphisms and the risk of colorectal cancer. Int J Cancer 120:1548– 1554., DOI 10.1002/ijc.22510 Campbell PT, Curtin K, Ulrich CM, et al. NIH public access 2010;58:661–667., DOI 10.1136/gut.2007.144220.Mismatch NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 231 EP 235 DI 10.1007/s12253-017-0231-5 PG 5 ER PT J AU Rubovszky, G Budai, B Ganofszky, E Horvath, Zs Juhos, Madaras, B Nagy, T Szabo, E Pinter, T Toth, E Nagy, P Lang, I Hitre, E AF Rubovszky, Gabor Budai, Barna Ganofszky, Erna Horvath, Zsolt Juhos, Eva Madaras, Balazs Nagy, Tunde Szabo, Eszter Pinter, Tamas Toth, Erika Nagy, Peter Lang, Istvan Hitre, Erika TI Predictive Value of Early Skin Rash in Cetuximab-Based Therapy of Advanced Biliary Tract Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acneiform rash; Predictive marker; Cetuximab; Chemotherapy; Cholangiocarcinoma ID Acneiform rash; Predictive marker; Cetuximab; Chemotherapy; Cholangiocarcinoma AB Randomized trials in advanced biliary tract cancer (BTC) did not show benefit of cetuximab addition over chemotherapy. This is probably due to the lack of predictive biomarkers. The aim of this study was to explore possible predictive factors. Between 2009 and 2014, 57 patients were treated in 3-week cycles with cetuximab (250 mg/m2/week, loading dose: 400 mg/m2), gemcitabine (1000 mg/m2 on day 1 and 8), and capecitabine (1300 mg/m2/day on days 1–14). The objective response rate (ORR), progression-free (PFS) and overall survival (OS) and the adverse events (AEs) were evaluated. An exploratory analysis was performed to find possible predictive factors on clinicopathological characteristics, routine laboratory parameters and early AEs, which occurred within 2 months from the beginning of treatment. The ORR was 21%. The median PFS and OS were 34 (95% CI: 24–40) and 54 (43–67) weeks, respectively. The most frequent AEs were skin toxicities. In univariate analysis performance status, previous stent implantation, thrombocyte count at the start of therapy, early neutropenia and skin rash statistically significantly influenced the ORR, PFS and/or OS. In multivariate Cox regression analysis only normal thrombocyte count at treatment start and early acneiform rash were independent markers of longer survival. In patients showing early skin rash compared to the others the median PFS was 39 vs. 13 weeks and the median OS was 67 vs. 26 weeks, respectively. It is suggested that early skin rash can be used as a biomarker to select patients who would benefit from the treatment with cetuximab plus chemotherapy. C1 [Rubovszky, Gabor] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Budai, Barna] National Institute of Oncology, Department of BiochemistryBudapest, Hungary. [Ganofszky, Erna] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Horvath, Zsolt] University of Debrecen, Department of OncologyDebrecen, Hungary. [Juhos, Eva] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Madaras, Balazs] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Nagy, Tunde] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Szabo, Eszter] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Pinter, Tamas] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Toth, Erika] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Nagy, Peter] National Institute of Oncology, Department of BiochemistryBudapest, Hungary. [Lang, Istvan] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Hitre, Erika] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. RP Rubovszky, G (reprint author), National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, 1122 Budapest, Hungary. EM garub@oncol.hu CR Randi G, Malvezzi M, Levi F et al, 2009, Epidemiology of biliary tract cancers: an update. Ann Oncol 20:146–159., DOI 10.1093/ annonc/mdn533 Valle J, Wasan H, Palmer DH et al, 2010, Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med 362:1273–1281., DOI 10.1056/NEJMoa0908721 Noel MS, Hezel AF, 2013, New and emerging treatment options for biliary tract cancer. Onco Targets Ther 6:1545–1452., DOI 10. 2147/OTT.S32545 Sharma A, Chaudhary SP, Shukla NK et al, 2014, A randomized controlled trial comparing modified gemcitabine plus oxaliplatin, mGEMOX, to gemcitabine plus cisplatin in management of unresectable gall bladder cancer. J Clin Oncol 32(Suppl):TPS4152 Ulahannan SV, Rahma OE, Duffy AG et al, 2015, Identification of active chemotherapy regimens in advanced biliary tract carcinoma: a review of chemotherapy trials in the past two decades. Hepat Oncol 2:39–50., DOI 10.2217/hep.14.36 Lee J, Park SH, Chang HM et al, 2012, Gemcitabine and oxaliplatin with or without erlotinib in advanced biliary-tract cancer: a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 13:181–188., DOI 10.1016/S1470-2045(11)70301-1 Malka D, Cervera P, Foulon S et al, 2014, Gemcitabine and oxaliplatin with or without cetuximab in advanced biliary-tract cancer, BINGO): a randomised, open-label, non-comparative phase 2 trial. Lancet Oncol 15:819–828., DOI 10.1016/S1470-2045(14)70212-8 Chen JS, Hsu C, Chiang NJ et al, 2015, A KRAS mutation statusstratified randomized phase II trial of gemcitabine and oxaliplatin alone or in combination with cetuximab in advanced biliary tract cancer. Ann Oncol 26:943–949., DOI 10.1093/annonc/mdv035 Petrelli F, Borgonovo K, Barni S, 2013, The predictive role of skin rash with cetuximab and panitumumab in colorectal cancer patients: a systematic review and meta-analysis of published trials. Target Oncol 8:173–181., DOI 10.1007/s11523-013-0257-x Borbath I, Ceratti A, Verslype C et al, 2013, Combination of gemcitabine and cetuximab in patients with advanced cholangiocarcinoma: a phase II study of the Belgian Group of Digestive Oncology. Ann Oncol 24:2824–2829., DOI 10.1093/annonc/mdt337 Rubovszky G, Lang I, Ganofszky E et al, 2013, Cetuximab, gemcitabine and capecitabine in patients with inoperable biliary tract cancer: a phase 2 study. Eur J Cancer 49:3806–3812., DOI 10. 1016/j.ejca.2013.07.143 Marcano-Bonilla L, Mohamed EA, Mounajjed T, Roberts LR, 2016, Biliary tract cancers: epidemiology, molecular pathogenesis and genetic risk associations. Chin Clin Oncol 5:61., DOI 10.21037/ cco.2016.10.09 Wang RT, Zhang LQ, Mu YP et al, 2015, Prognostic significance of preoperative platelet count in patients with gallbladder cancer.World J Gastroenterol 21:5303–5310., DOI 10.3748/wjg.v21.i17.5303 Baranyai Z, Josa V, Toth A et al, 2016, Paraneoplastic thrombocytosis in gastrointestinal cancer. Platelets 27:269–275., DOI 10.3109/09537104.2016.1170112 Lee SY, Kim HS, Choi YJ et al, 2016, A prognostic index to identify patients with intrahepatic cholangiocarcinoma who could benefit from gemcitabine plus cisplatin. Am J Ther 23:e1449– e1455., DOI 10.1097/MJT.0000000000000112 Helleday T, 2016, PARP inhibitor receives FDAbreakthrough therapy designation in castration resistant prostate cancer: beyond germline BRCA mutations. Ann Oncol 27:755–757., DOI 10.1093/ annonc/mdw048 Cai J, Ma H, Huang F et al, 2013, Correlation of bevacizumabinduced hypertension and outcomes of metastatic colorectal cancer patients treated with bevacizumab: a systematic review and metaanalysis. World J Surg Oncol 11:306., DOI 10.1186/1477-7819-11-306 Nagyivanyi K, Budai B, Biro K et al, 2016, Synergistic survival: a new phenomenon connected to adverse events of first-line sunitinib treatment in advanced renal cell carcinoma. Clin Genitourin Cancer in press., DOI 10.1016/j.clgc.2015.11.016 Rini BI, Cohen DP, Lu DR et al, 2011, Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst 103:763–773., DOI 10.1093/jnci/djr128 Hurwitz HI, Douglas PS, Middleton JP et al, 2013, Analysis of early hypertension and clinical outcome with bevacizumab: results from seven phase III studies. Oncologist 18:273–280., DOI 10.1634/ theoncologist.2012-0339 Huober J, Cole BF, Rabaglio M et al, 2014, Symptoms of endocrine treatment and outcome in the BIG 1-98 study. Breast Cancer Res Treat 143:159–169., DOI 10.1007/s10549-013-2792-7 Moore MJ, Goldstein D, Hamm J et al, 2007, Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada clinical trials Group. J Clin Oncol 25:1960– 1966., DOI 10.1200/JCO.2006.07.9525 Van Cutsem E, Li CP, Nowara E et al, 2014, Dose escalation to rash for erlotinib plus gemcitabine for metastatic pancreatic cancer: the phase II RACHEL study. Br J Cancer 111:2067–2075., DOI 10.1038/ bjc.2014.494 Havrilesky LJ, Reiner M, Morrow PK et al, 2015, A review of relative dose intensity and survival in patients with metastatic solid tumors. Crit Rev Oncol Hematol 93:203–210., DOI 10.1016/j. critrevonc.2014.10.006 Citron ML, 2004, Dose density in adjuvant chemotherapy for breast cancer. Cancer Investig 22:555–568., DOI 10.1081/CNV- 200027134 Rudek MA, Connolly RM, Hoskins JM et al, 2013, Fixeddose capecitabine is feasible: results from a pharmacokinetic and pharmacogenetic study in metastatic breast cancer. Breast Cancer Res Treat 139:135–143., DOI 10.1007/s10549- 013-2516-z Lortholary A, Hardy-Bessard AC, de Rauglaudre G et al, 2012, A GINECO randomized phase II trial of two capecitabine and weekly paclitaxel schedules in metastatic breast cancer. Breast Cancer Res Treat 131:127–135., DOI 10.1007/s10549-011-1776-8 Chan A, McGregor S, Liang W, 2014, Utilisation of primary and secondary G-CSF prophylaxis enables maintenance of optimal dose delivery of standard adjuvant chemotherapy for early breast cancer: an analysis of 1655 patients. Breast 23:676–682., DOI 10.1016/j. breast.2014.07.004 Shimoyama S, 2010, Pharmacogenetics of irinotecan: an ethnicitybased prediction of irinotecan adverse events. World J Gastrointest Surg 2:14–21., DOI 10.4240/wjgs.v2.i1.14 Bruckner HW, Hirschfeld A, Schwartz M, 2016, Targeted therapy for resistant cholangiocharcinoma with bevacizumab or cetuximab added to failed cytotoxic drug cores. Anticancer Res 36:399–402 Valle JW, Wasan H, Lopes A et al, 2015, Cediranib or placebo in combination with cisplatin and gemcitabine chemotherapy for patients with advanced biliary tract cancer, ABC-03): a randomised phase 2 trial. Lancet Oncol 16:967–978., DOI 10.1016/S1470- 2045(15)00139-4 Mizusawa J, Morizane C, Okusaka T et al, 2016, Randomized phase III study of gemcitabine plus S-1 versus gemcitabine plus cisplatin in advanced biliary tract cancer: Japan clinical Oncology Group study, JCOG1113, FUGA-BT). Jpn J Clin Oncol 46:385– 388., DOI 10.1093/jjco/hyv213 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 237 EP 244 DI 10.1007/s12253-017-0238-y PG 8 ER PT J AU Abbaszadegan, RM Moghbeli, M AF Abbaszadegan, Reza Mohammad Moghbeli, Meysam TI Role of MAML1 and MEIS1 in Esophageal Squamous Cell Carcinoma Depth of Invasion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NOTCH signaling pathway; HOX; mRNA expression; Transcription factor; Self renewal ID NOTCH signaling pathway; HOX; mRNA expression; Transcription factor; Self renewal AB Homeobox (HOX) transcription factors and NOTCH signaling pathway are critical regulators of stem cell functions, cell fate in development and homeostasis of gastrointestinal tissues. In the present study, we analyzed cross talk between NOTCH pathway and HOX genes through assessment of probable correlation between MAML1 and MEIS1 as the main transcription factor of NOTCH pathway and enhancer of HOX transcriptional machinery, respectively in esophageal squamous cell carcinoma (ESCC) patients. Fifty one ESCC cases were enrolled to assess the levels of Meis1 and Maml1 mRNA expression using real-time polymerase chain reaction (PCR). Only 3 out of 51 (5.9%) cases had MEIS1/MAML1 under expression and 2/51 (3.9%) cases had MEIS1/MAML1over expression. Nine out of 51 samples (17.6%) have shown MEIS1 under expression and MAML1 over expression. There was a significant correlation between MAML1and MEIS1mRNA expressions (p ≤ 0.05). There were significant correlations between MEIS1 under/MAML1 over expressed cases and tumor location (p = 0.05), tumor depth of invasion (p = 0.011), and sex (p = 0.04). Our results showed that MEIS1 may have a negative role in regulation of MAML1expression during the ESCC progression. C1 [Abbaszadegan, Reza Mohammad] Mashhad University of Medical Sciences, Immunology Research Center, Avicenna Research Institute, Division of Human GeneticsMashhad, Iran. [Moghbeli, Meysam] North Khorasan University of Medical SciencesBojnurd, Iran. RP Moghbeli, M (reprint author), North Khorasan University of Medical Sciences, Bojnurd, Iran. EM m.moghbeli@nkums.ac.ir;Meysam_moghbeli@yahoo.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90., DOI 10. 3322/caac.20107 Moghbeli M, Abbaszadegan MR, Farshchian M, Montazer M, Raeisossadati R, Abdollahi A, Forghanifard MM, 2013, Association of PYGO2 and EGFR in esophageal squamous cell carcinoma. Med Oncol 30(2):516 Moghbeli M, Abbaszadegan MR, Golmakani E, Forghanifard MM, 2016, Correlation of Wnt and NOTCH pathways in esophageal squamous cell carcinoma. J Cell Commun Signal 10(2):129–135 Moghbeli M, Forghanifard MM, Sadrizadeh A, Mozaffari HM, Golmakani E, Abbaszadegan MR, 2015, Role of Msi1 and MAML1 in regulation of notch signaling pathway in patients with esophageal squamous cell carcinoma. J Gastrointest Cancer 46(4): 365–369 Moghbeli M, Moghbeli F, Forghanifard MM, Garayali A, Abbaszadegan MR, 2013, Cancer stem cell markers in esophageal cancer. American Journal of Cancer Science 2(1):37–50 Moghbeli M, Rad A, Farshchian M, Taghehchian N, Gholamin M, Abbaszadegan MR, 2016, Correlation betweenMeis1 and Msi1 in esophageal squamous cell carcinoma. J Gastrointest Cancer 47(3): 273–277 Moghbeli M, Sadrizadeh A, Forghanifard MM, Mozaffari HM, Golmakani E, Abbaszadegan MR, 2016, Role of Msi1 and PYGO2 in esophageal squamous cell carcinoma depth of invasion. J Cell Commun Signal 10(1):49–53 Taleb S, Abbaszadegan MR, Moghbeli M, Roudbari NH, Forghanifard MM, 2014, HES1 as an independent prognostic marker in esophageal squamous cell carcinoma. J Gastrointest Cancer 45(4):466–471 Artavanis-Tsakonas S, Rand MD, Lake RJ, 1999, Notch signaling: cell fate control and signal integration in development. Science 284(5415):770–776 Hsieh JJ, Zhou S, Chen L, Young DB, Hayward SD, 1999, CIR, a corepressor linking the DNA binding factor CBF1 to the histone deacetylase complex. Proc Natl Acad Sci U S A 96(1):23–28 Kao HY, Ordentlich P, Koyano-Nakagawa N, Tang Z, Downes M, Kintner CR, Evans RM, Kadesch T, 1998, A histone deacetylase corepressor complex regulates the notch signal transduction pathway. Genes Dev 12(15):2269–2277 Taniguchi Y, Furukawa T, Tun T, Han H, Honjo T, 1998, LIM protein KyoT2 negatively regulates transcription by association with the RBP-J DNA-binding protein. Mol Cell Biol 18(1):644– 654 Oswald F, Tauber B, Dobner T, Bourteele S, Kostezka U, Adler G, Liptay S, Schmid RM, 2001, p300 acts as a transcriptional coactivator for mammalian notch-1. Mol Cell Biol 21(22):7761–7774., DOI 10.1128/MCB.21.22.7761-7774.2001 Kurooka H, Honjo T, 2000, Functional interaction between the mouse notch1 intracellular region and histone acetyltransferases PCAF and GCN5. J Biol Chem 275(22):17211–17220., DOI 10. 1074/jbc.M000909200 Wu L, Sun T, Kobayashi K, Gao P, Griffin JD, 2002, Identification of a family ofmastermind-like transcriptional coactivators for mammalian notch receptors. Mol Cell Biol 22(21):7688–7700 Kitagawa M, Oyama T, Kawashima T, Yedvobnick B, Kumar A, Matsuno K, Harigaya K, 2001, A human protein with sequence similarity to drosophila mastermind coordinates the nuclear form of notch and a CSL protein to build a transcriptional activator complex on target promoters. Mol Cell Biol 21(13):4337–4346., DOI 10. 1128/MCB.21.13.4337-4346.2001 Lin SE, Oyama T, Nagase T, Harigaya K, Kitagawa M, 2002, Identification of new human mastermind proteins defines a family that consists of positive regulators for notch signaling. J Biol Chem 277(52):50612–50620., DOI 10.1074/jbc.M209529200 Wu L, Aster JC, Blacklow SC, Lake R, Artavanis-Tsakonas S, Griffin JD, 2000, MAML1, a human homologue of drosophila mastermind, is a transcriptional co-activator for NOTCH receptors. Nat Genet 26(4):484–489., DOI 10.1038/82644 Jeffries S, Robbins DJ, Capobianco AJ, 2002, Characterization of a high-molecular-weight notch complex in the nucleus of notch(ic)- transformed RKE cells and in a human T-cell leukemia cell line. Mol Cell Biol 22(11):3927–3941 Saint Just Ribeiro M, Hansson ML,Wallberg AE, 2007, A proline repeat domain in the notch co-activator MAML1 is important for the p300-mediated acetylation ofMAML1. Biochem J 404(2):289– 298., DOI 10.1042/BJ20061900 Owens BM, Hawley RG, 2002, HOX and non-HOX homeobox genes in leukemic hematopoiesis. Stem Cells 20(5):364–379 Sitwala KV, Dandekar MN, Hess JL, 2008, HOX proteins and leukemia. Int J Clin Exp Pathol 1(6):461–474 Cillo C, CantileM, FaiellaA, Boncinelli E, 2001)Homeobox genes in normal and malignant cells. J Cell Physiol 188(2):161–169 Goh SL, Looi Y, Shen H, Fang J, Bodner C, Houle M, Ng AC, Screaton RA, Featherstone M, 2009, Transcriptional activation by MEIS1A in response to protein kinase a signaling requires the transducers of regulated CREB family of CREB co-activators. J Biol Chem 284(28):18904–18912 Huang H, Rastegar M, Bodner C, Goh SL, Rambaldi I, Featherstone M, 2005, MEIS C termini harbor transcriptional activation domains that respond to cell signaling. J Biol Chem 280(11):10119–10127 Wang Z, IwasakiM, Ficara F, Lin C, Matheny C,Wong SH, Smith KS, Cleary ML, 2010, GSK-3 promotes conditional association of CREB and its coactivators with MEIS1 to facilitate HOX-mediated transcription and oncogenesis. Cancer Cell 17(6):597–608 Boucherat O, Chakir J, Jeannotte L, 2012, The loss of Hoxa5 function promotes notch-dependent goblet cell metaplasia in lung airways. Biol Open 1(7):677–691 Chariot A, Gielen J, Merville MP, Bours V, 1999, The homeodomain-containing proteins: an update on their interacting partners. Biochem Pharmacol 58(12):1851–1857 Esparza SD, Chang J, Shankar DB, Zhang B, Nelson SF, Sakamoto KM, 2008, CREB regulates Meis1 expression in normal and malignant hematopoietic cells. Leukemia 22(3):665–667., DOI 10. 1038/sj.leu.2404933 Ferretti E, Villaescusa JC, Di Rosa P, Fernandez-Diaz LC, Longobardi E, Mazzieri R, Miccio A, Micali N, Selleri L, Ferrari G, Blasi F, 2006, Hypomorphic mutation of the TALE gene Prep1, pKnox1, causes a major reduction of Pbx and Meis proteins and a pleiotropic embryonic phenotype. Mol Cell Biol 26(15):5650– 5662., DOI 10.1128/MCB.00313-06 Moghbeli M, Maleknejad M, Arabi A, Abbaszadegan MR, 2012, Mutational analysis of uroporphyrinogen III cosynthase gene in Iranian families with congenital erythropoietic porphyria. Mol Biol Rep 39(6):6731–6735 Forghanifard MM, Moghbeli M, Raeisossadati R, Tavassoli A, Mallak AJ, Boroumand-Noughabi S, Abbaszadegan MR, 2013, Role of SALL4 in the progression and metastasis of colorectal cancer. J Biomed Sci 20:6 Forghanifard MM, Moaven O, Farshchian M, Montazer M, Raeisossadati R, Abdollahi A, Moghbeli M, Nejadsattari T, Parivar K, Abbaszadegan MR, 2012, Expression analysis elucidates the roles ofMAML1 and Twist1 in esophageal squamous cell carcinoma aggressiveness and metastasis. Ann Surg Oncol 19(3): 743–749 Grier DG, Thompson A, Kwasniewska A, McGonigle GJ, Halliday HL, Lappin TR, 2005, The pathophysiology of HOX genes and their role in cancer. J Pathol 205(2):154–171., DOI 10.1002/path. 1710 Wermuth PJ, Buchberg AM, 2005, Meis1-mediated apoptosis is caspase dependent and can be suppressed by coexpression of HoxA9 in murine and human cell lines. Blood 105(3):1222–1230., DOI 10.1182/blood-2004-03-0802 Yamashita T, Tazawa S, Yawei Z, Katayama H, Kato Y, Nishiwaki K, Yokohama Y, Ishikawa M, 2006, Suppression of invasive characteristics by antisense introduction of overexpressed HOX genes in ovarian cancer cells. Int J Oncol 28(4):931–938 Argiropoulos B, Yung E, Humphries RK, 2007, Unraveling the crucial roles of Meis1 in leukemogenesis and normal hematopoiesis. Genes Dev 21(22):2845–2849., DOI 10.1101/gad.1619407 Calvo KR, Knoepfler PS, Sykes DB, Pasillas MP, Kamps MP, 2001, Meis1a suppresses differentiation by G-CSF and promotes proliferation by SCF: potential mechanisms of cooperativity with Hoxa9 in myeloid leukemia. Proc Natl Acad Sci U S A 98(23): 13120–13125., DOI 10.1073/pnas.231115398 Kumar AR, Li Q, Hudson WA, Chen W, Sam T, Yao Q, Lund EA, Wu B, Kowal BJ, Kersey JH, 2009, A role for MEIS1 in MLLfusion gene leukemia. Blood 113(8):1756–1758., DOI 10.1182/ blood-2008-06-163287 Deveraux QL, Roy N, Stennicke HR, Van Arsdale T, Zhou Q, Srinivasula SM, Alnemri ES, Salvesen GS, Reed JC, 1998, IAPs block apoptotic events induced by caspase-8 and cytochrome c by direct inhibition of distinct caspases. EMBO J 17(8):2215–2223., DOI 10.1093/emboj/17.8.2215 Lindahl T, Satoh MS, Poirier GG, Klungland A, 1995, Posttranslational modification of poly(ADP-ribose, polymerase induced by DNA strand breaks. Trends Biochem Sci 20(10):405–411 Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, Wang X, 1997, Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 91(4):479–489 Brinkmann U, Brinkmann E, GalloM, Pastan I, 1995, Cloning and characterization of a cellular apoptosis susceptibility gene, the human homologue to the yeast chromosome segregation gene CSE1. Proc Natl Acad Sci U S A 92(22):10427–10431 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 245 EP 250 DI 10.1007/s12253-017-0243-1 PG 6 ER PT J AU Jianwei, Z Qi, L Quanquan, X Tianen, W Qingwei, W AF Jianwei, Zhang Qi, Li Quanquan, Xu Tianen, Wang Qingwei, Wang TI TMPRSS4 Upregulates TWIST1 Expression through STAT3 Activation to Induce Prostate Cancer Cell Migration SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Transmembrane protease serine 4; Epithelial-mesenchymal transition; Migration; Signal transducer and activator of transcription 3; TWIST1 ID Prostate cancer; Transmembrane protease serine 4; Epithelial-mesenchymal transition; Migration; Signal transducer and activator of transcription 3; TWIST1 AB Transmembrane protease serine 4 (TMPRSS4), a type-II transmembrane serine protease, is involved in the development and progression of wide range of tumors. However, the biological role of TMPRSS4 in prostate cancer remains obscure. Here, we investigated the effect of TMPRSS4 on proliferation and migration in prostate cancer and potential mechanisms. Our findings demonstrated over-expression of TMPRSS4 promoted the PC3 prostate cancer cells migration, which could be reversed by TMPRSS4 silencing. TMPRSS4 induced TWIST1 expression and followed progression of EMT along with upregulation of N-cadherin and downregulation of E-cadherin via STAT3 phosphorylation. Silencing TWIST1 significantly attenuated TMPRSS4-induced PC3 migration. Moreover, knockdown of STAT3 effectively attenuated TMPRSS4- induced TWIST1 expression and TWIST1 promoter activity. Taken together, we demonstrated a mechanistic cascade of TMPRSS4 up-regulating STAT3 activation and subsequent TWIST1 expression, leading to prostate cancer migration. C1 [Jianwei, Zhang] The First Affiliated Hospital of Zhengzhou University, Department of Urology, 450052 Zhengzhou, Henan Province, China. [Qi, Li] The First Affiliated Hospital of Zhengzhou University, Department of Urology, 450052 Zhengzhou, Henan Province, China. [Quanquan, Xu] The First Affiliated Hospital of Zhengzhou University, Department of Urology, 450052 Zhengzhou, Henan Province, China. [Tianen, Wang] The First Affiliated Hospital of Zhengzhou University, Department of Urology, 450052 Zhengzhou, Henan Province, China. [Qingwei, Wang] The First Affiliated Hospital of Zhengzhou University, Department of Urology, 450052 Zhengzhou, Henan Province, China. RP Qingwei, W (reprint author), The First Affiliated Hospital of Zhengzhou University, Department of Urology, 450052 Zhengzhou, China. EM kyl361@126.com CR Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63(1):11–30 Kim S, Lee JW, 2014, Membrane proteins involved in epithelialmesenchymal transition and tumor invasion: studies on TMPRSS4 and TM4SF5. Genomics Inform 12(1):12–20 Chikaishi Y, Uramoto H, Koyanagi Y et al, 2016, TMPRSS4 expression as a marker of recurrence in patients with lung cancer. Anticancer Res 36(1):121–127 Choi SY, Shin HC, Kim SYet al, 2008, Role of TMPRSS4 during cancer progression. Drug News Perspect 21(8):417–423 de Aberasturi AL, Calvo A, 2015, TMPRSS4: an emerging potential therapeutic target in cancer. Br J Cancer 112(1):4–8 Wu XY, Zhang L, Zhang KM et al, 2014, Clinical implication of TMPRSS4 expression in human gallbladder cancer. Tumour Biol 35(6):5481–5486 Huang A, Zhou H, Zhao H et al, 2014, TMPRSS4 correlates with colorectal cancer pathological stage and regulates cell proliferation and self-renewal ability. Cancer Biol Ther 15(3):297–304 Larzabal L, Nguewa PA, Pio R et al, 2011, Overexpression of TMPRSS4 in non-small cell lung cancer is associated with poor prognosis in patients with squamous histology. Br J Cancer 105(10):1608–1614 Jung H, Lee KP, Park SJ et al, 2008, TMPRSS4 promotes invasion, migration and metastasis of human tumor cells by facilitating an epithelial-mesenchymal transition. Oncogene 27(18):2635–2647 KimS, KangHY, NamEH et al, 2010)TMPRSS4 induces invasion and epithelial-mesenchymal transition through upregulation of integrin alpha5 and its signaling pathways. Carcinogenesis 31(4): 597–606 Taylor BS, Schultz N, Hieronymus H et al, 2010, Integrative genomic profiling of human prostate cancer. Cancer Cell 18(1):11–22 Shi G, Yang X, Dai B et al, 2014, Clinical significance of TMPRSS4 in prostate cancer. Int J Clin Exp Pathol 7(11):8053– 8058 Yang J, Mani SA, Donaher JL et al, 2004, Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117(7):927–939 Zhu QQ, Ma C, Wang Q et al, 2016, The role of TWIST1 in epithelial-mesenchymal transition and cancers. Tumour Biol 37(1):185–197 KwokWK, LingMT, LeeTWet al, 2005, Up-regulation of TWIST in prostate cancer and its implication as a therapeutic target. Cancer Res 65(12):5153–5162 Lee JH, Kim JE, Kim BG et al, 2016, STAT3-induced WDR1 overexpression promotes breast cancer cell migration. Cell Signal 28(11):1753–1760 Lo HW, Hsu SC, Xia W et al, 2007, Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res 67(19):9066–9076 Zheng J, Mercado-Uribe I, Rosen DG et al, 2010, Induction of papillary carcinoma in human ovarian surface epithelial cells using combined genetic elements and peritoneal microenvironment. Cell Cycle 9(1):140–146 Kumano M, Miyake H, Muramaki M et al, 2009, Expression of urokinase-type plasminogen activator system in prostate cancer: correlation with clinicopathological outcomes in patients undergoing radical prostatectomy. Urol Oncol 27(2):180–186 Huang A, Zhou H, Zhao H et al, 2013, High expression level of TMPRSS4 predicts adverse outcomes of colorectal cancer patients. Med Oncol 30(4):712 Hamamoto J, Soejima K,NaokiKet al, 2015)Methylation-induced downregulation of TFPI-2 causes TMPRSS4 overexpression and contributes to oncogenesis in a subset of non-small-cell lung carcinoma. Cancer Sci 106(1):34–42 Shiota M, Zardan A, Takeuchi A et al, 2012, Clusterin mediates TGF-β-induced epithelial-mesenchymal transition and metastasis via Twist1 in prostate cancer cells. Cancer Res 72(20):5261–5272 Zhao D, Besser AH, Wander SA et al, 2015, Cytoplasmic p27 promotes epithelial-mesenchymal transition and tumor metastasis via STAT3-mediated Twist1 upregulation. Oncogene 34(43): 5447–5459 Lee Y, Ko D, Min HJ et al, 2016, TMPRSS4 induces invasion and proliferation of prostate cancer cells through induction of slug and cyclin D1. Oncotarget., DOI 10.18632/oncotarget.10382 Min HJ, Lee MK, Lee JW et al, 2014,, 2014, TMPRSS4 induces cancer cell invasion through pro-uPAprocessing. BiochemBiophys Res Commun 446(1):1–7 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 251 EP 257 DI 10.1007/s12253-017-0237-z PG 7 ER PT J AU Zombori, T Cserni, G AF Zombori, Tamas Cserni, Gabor TI Immunohistochemical Analysis of the Expression of Breast Markers in Basal-like Breast Carcinomas Defined as Triple Negative Cancers Expressing Keratin 5 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Basal-like; Triple negative breast cancer; GATA-3; Mammaglobin; GCDFP-15; NY-BR-1 ID Basal-like; Triple negative breast cancer; GATA-3; Mammaglobin; GCDFP-15; NY-BR-1 AB Estrogen and progesterone receptors are possible markers for suggesting a mammary origin of metastatic carcinoma, but are useless in cases of triple negative breast cancers (TNBC). Five other potential markers of breast origin were investigated on tissue microarrays in a series of TNBCs showing keratin 5 expression, consistent with a basal-like phenotype. GATA-3 staining was observed in 82 of 115 triple negative cases (71.3%) including 23 cases with >5% staining. Mammaglobin staining was detected in 30 cases (26.0%) including 12 with >5% staining. GCDFP-15 was seen in 23 cases (20.0%) including 9 with >5% staining. NY-BR-1 positivity was present in 7 cases (6.0%) including 3 patients with >5% staining. BCA-225 staining was observed in 74 cases (64.3%); however this latter marker lacks also specificity owing to the reported widespread staining in other malignancies. GATA-3, mammaglobin and GCDFP-15 coexpression was seen in one case (0.9%), whereas GATA-3 and mammaglobin or mammaglobin and GCDFP-15 coexpression was present in 2 and 2 cases (1.7%), respectively. Using at least 5% staining as cut-off, the expression of any of the last 4 markers was 34.7%. The expression of GATA-3, mammaglobin, GCDFP- 15 and NY-BR-1 is lower in TNBC-s than in breast carcinomas in general, and this may be even lower in basal-like carcinomas. Although these markers are not fully specific, by using them, a subset of basal-like TNBC-s can be identified as of mammary origin. However, a substantial proportion will not show any staining with any of these markers. C1 [Zombori, Tamas] University of Szeged, Department of Pathology, Allomas u. 1, 6725 Szeged, Hungary. [Cserni, Gabor] University of Szeged, Department of Pathology, Allomas u. 1, 6725 Szeged, Hungary. RP Zombori, T (reprint author), University of Szeged, Department of Pathology, 6725 Szeged, Hungary. EM zomtam@gmail.com CR DeSantis C, Ma J, Bryan L, Jemal A, 2013, Breast cancer statistics, 2013. CA Cancer J Clin 64:52–62., DOI 10.3322/caac.21203 MalvezziM, Carioli G, Bertuccio P, Rosso T, Boffetta P, Levi F, La Vecchia C, Negri E, 2016, European cancer mortality predictions for the year 2016 with focus on leukaemias. Ann Oncol 27:725– 731., DOI 10.1093/annonc/mdw022 Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D, 2000, Molecular portraits of human breast tumours. Nature 406:747–752 Nielsen TO, Perou CM(2015, CCR 20th anniversary commentary: the development of breast cancer molecular subtyping. Clin Cancer Res 21:1779–1781., DOI 10.1158/1078-0432.CCR-14-2552 Hernandez BY, Green MD, Cassel KD, Pobutsky AM, Vu V, Wilkens LR, 2010, Preview of Hawaii cancer facts and figures. Hawaii Med J 69:223–224 Huo L, Gong Y, Guo M, Gilcrease MZ, Wu Y, Zhang H, Zhang J, Resetkova E, Hunt KK, Deavers MT(2015, GATA-binding protein 3 enhances the utility of gross cystic disease fluid protein-15 and mammaglobin a in triple-negative breast cancer by immunohistochemistry. Histopathology 67:245–254., DOI 10.1111/his.12645 Deftereos G, Sanguino RamirezAM, Silverman JF, KrishnamurtiU, 2015, GATA3 immunohistochemistry expression in histologic subtypes of primary breast carcinoma and metastatic breast carcinoma cytology. Am J Surg Pathol 39:1282–1289., DOI 10.1097/ PAS.0000000000000505 Chou J, Provot S, Werb Z, 2010, GATA3 in development and cancer differentiation: cells GATA have it! J Cell Physiol 222:42– 49., DOI 10.1002/jcp.21943 Esheba GE, Longacre TA, Atkins KA, Higgins JP, 2009, Expression of the urothelial differentiation markers GATA3 and placental S100, S100P, in female genital tract transitional cell proliferations. Am J Surg Pathol 33:347–353 Liu H, Shi J, Wilkerson ML, Lin F, 2012, Immunohistochemical evaluation of GATA3 expression in tumors and normal tissues: a useful immunomarker for breast and urothelial carcinomas. Am J Clin Pathol 138:57–64., DOI 10.1309/AJCP5UAFMSA9ZQBZ Ordonez NG, 2013, Value of GATA3 immunostaining in tumor diagnosis: a review. Adv Anat Pathol 20:352–360., DOI 10.1097/ PAP.0b013e3182a28a68 Miettinen M, McCue PA, Sarlomo-Rikala M, Rys J, Czapiewski P, Wazny K, Langfort R,Waloszczyk P, BiernatW, Lasota J,Wang Z, 2014, GATA3: a multispecific but potentially useful marker in surgical pathology: a systematic analysis of 2500 epithelial and nonepithelial tumors. Am J Surg Pathol 38:13–22., DOI 10.1097/ PAS.0b013e3182a0218f Voduc D, Cheang M, Nielsen T, 2008, GATA-3 expression in breast cancer has a strong association with estrogen receptor but lacks independent prognostic value. Cancer Epidemiol Biomark Prev 17:365–373., DOI 10.1158/1055-9965.EPI-06-1090 Hoch RV, Thompson DA, Baker RJ, Weigel RJ, 1999, GATA-3 is expressed in association with estrogen receptor in breast cancer. Int J Cancer 84:122–128 Tominaga N, Naoi Y, Shimazu K, Nakayama T, Maruyama N, Shimomura A, Kim SJ, Tamaki Y, Noguchi S, 2012, Clinicopathological analysis of GATA3-positive breast cancers with special reference to response to neoadjuvant chemotherapy. Ann Oncol 23:3051–3057., DOI 10.1093/annonc/mds120 Albergaria A, Paredes J, Sousa B,Milanezi F, Carneiro V, Bastos J, Costa S, Vieira D, Lopes N, Lam EW, Lunet N, Schmitt F, 2009, Expression of FOXA1 and GATA-3 in breast cancer: the prognostic significance in hormone receptor-negative tumours. Breast Cancer Res 11:R40., DOI 10.1186/bcr2327 Parikh P, Palazzo JP, Rose LJ, Daskalakis C, Weigel RJ, 2005, GATA-3 expression as a predictor of hormone response in breast cancer. J Am Coll Surg 200:705–710 Mehra R, Varambally S, Ding L, Shen R, Sabel MS, Ghosh D, Chinnaiyan AM, Kleer CG, 2005, Identification of GATA3 as a breast cancer prognostic marker by global gene expression metaanalysis. Cancer Res 65:11259–11264 Cimino-Mathews A, Subhawong AP, Illei PB, Sharma R, Halushka MK, Vang R, Fetting JH, Park BH, Argani P, 2013, GATA3 expression in breast carcinoma: utility in triple-negative, sarcomatoid, and metastatic carcinomas. Hum Pathol 44:1341–1349., DOI 10. 1016/j.humpath.2012.11.003 Ciocca V, Daskalakis C, Ciocca RM, Ruiz-Orrico A, Palazzo JP, 2009, The significance of GATA3 expression in breast cancer: a 10-year follow-up study. Hum Pathol 40:489–495., DOI 10.1016/j. humpath.2008.09.010 Jacquemier J, Charafe-Jauffret E, Monville F, Esterni B, Extra JM, Houvenaeghel G, Xerri L, Bertucci F, Birnbaum D, 2009, Association of GATA3, P53, Ki67 status and vascular peritumoral invasion are strongly prognostic in luminal breast cancer. Breast Cancer Res 11:R23., DOI 10.1186/bcr2249 Demir H, Turna H, Can G, Ilvan S, 2010, Clinicopathologic and prognostic evaluation of invasive breast carcinoma molecular subtypes and GATA3 expression. J Buon 15:774–782 Watson MA, Fleming TP, 1996, Mammaglobin, a mammaryspecific member of the uteroglobin gene family, is overexpressed in human breast cancer. Cancer Res 56:860–865 Watson MA, Darrow C, Zimonjic DB, Popescu NC, Fleming TP, 1998, Structure and transcriptional regulation of the human mammaglobin gene, a breast cancer associated member of the uteroglobin gene family localized to chromosome 11q13. Oncogene 16:817–824 Han JH, Kang Y, Shin HC, Kim HS, Kang YM, Kim YB, Oh SY, 2003, Mammaglobin expression in lymph nodes is an important marker of metastatic breast carcinoma. Arch Pathol Lab Med 127: 1330–1334 Bhargava R, Beriwal S, Dabbs DJ, 2007, Mammaglobin vs GCDFP-15: an immunohistologic validation survey for sensitivity and specificity. Am J Clin Pathol 127:103–113 Zafrakas M, Petschke B, Donner A, Fritzsche F, Kristiansen G, Knuchel R, Dahl E, 2006, Expression analysis of mammaglobin a, SCGB2A2, and lipophilin B, SCGB1D2, in more than 300 human tumors and matching normal tissues reveals their coexpression in gynecologic malignancies. BMC Cancer 6:88 Sasaki E, Tsunoda N, Hatanaka Y, Mori N, Iwata H, Yatabe Y, 2007, Breast-specific expression of MGB1/mammaglobin: an examination of 480 tumors from various organs and clinicopathological analysis ofMGB1-positive breast cancers. Mod Pathol 20:208– 214 Wang Z, Spaulding B, Sienko A, Liang Y, Li H, Nielsen G, Yub Gong G, Ro JY, Jim Zhai Q, 2009, Mammaglobin, a valuable diagnostic marker for metastatic breast carcinoma. Int J Clin Exp Pathol 2:384–389 Onuma K, Dabbs DJ, Bhargava R, 2008, Mammaglobin expression in the female genital tract: immunohistochemical analysis in benign and neoplastic endocervix and endometrium. Int J Gynecol Pathol 27:418–425., DOI 10.1097/PGP.0b013e31815d05ec Al-Joudi FS, Kaid FA, Ishak I, Mohamed N, Osman K, Alias IZ, 2011, Expression of human mammaglobin and clinicopathologic correlations in breast cancer: the findings in Malaysia. Indian J Pathol Microbiol 54:284–289., DOI 10.4103/0377-4929.81596 Lewis GH, Subhawong AP, Nassar H, Vang R, Illei PB, Park BH, Argani P, 2011, Relationship between molecular subtype of invasive breast carcinoma and expression of gross cystic disease fluid protein 15 and mammaglobin. Am J Clin Pathol 135:587–591., DOI 10.1309/AJCPMFR6OA8ICHNH Fritzsche FR, Thomas A,Winzer KJ, Beyer B, Dankof A, Bellach J, Dahl E, Dietel M, Kristiansen G, 2007, Co-expression and prognostic value of gross cystic disease fluid protein 15 and mammaglobin in primary breast cancer. Histol Histopathol 22: 1221–1230 Haagensen DE Jr,Mazoujian G, HolderWDJr, Kister SJ,Wells SA Jr, 1977, Evaluation of a breast cyst fluid protein detectable in the plasma of breast carcinoma patients. Ann Surg 185:279–285 Wick MR, Lillemoe TJ, Copland GT, Swanson PE, Manivel JC, Kiang DT, 1989, Gross cystic disease fluid protein-15 as a marker for breast cancer: immunohistochemical analysis of 690 human neoplasms and comparison with alpha-lactalbumin. Hum Pathol 20:281–287 Park SY, Kim BH, Kim JH, Lee S, Kang GH, 2007, Panels of immunohistochemical markers help determine primary sites ofmetastatic adenocarcinoma. Arch Pathol Lab Med 131:1561–1567 Jager D, Stockert E, Gure AO, Scanlan MJ, Karbach J, Jager E, Knuth A, Old LJ, Chen YT, 2001, Identification of a tissue-specific putative transcription factor in breast tissue by serological screening of a breast cancer library. Cancer Res 61:2055–2061 Jager D, Filonenko V, Gout I, Frosina D, Eastlake-Wade S, Castelli S, Varga Z,Moch H, Chen YT, Busam KJ, Seil I, Old LJ, Nissan A, Frei C, Gure AO, Knuth A, Jungbluth AA, 2007, NY-BR-1 is a differentiation antigen of the mammary gland. Appl Immunohistochem Mol Morphol 15:77–83 Giger OT, Lacoste E, Honegger C, Padberg B, Moch H, Varga Z, 2007, Expression of the breast differentiation antigen NY-BR-1 in a phyllodes tumor of the vulva. Virchows Arch 450:471–474 Varga Z, Theurillat JP, Filonenko V, Sasse B, Odermatt B, Jungbluth AA, Chen YT, Old LJ, Knuth A, Jager D, Moch H, 2006, Preferential nuclear and cytoplasmic NY-BR-1 protein expression in primary breast cancer and lymph node metastases. Clin Cancer Res 12:2745–2751 Theurillat JP, Zurrer-Hardi U, Varga Z, Storz M, Probst-Hensch NM, Seifert B, Fehr MK, Fink D, Ferrone S, Pestalozzi B, Jungbluth AA, Chen YT, Jager D, Knuth A, Moch H, 2007, NYBR- 1 protein expression in breast carcinoma: a mammary gland differentiation antigen as target for cancer immunotherapy. Cancer Immunol Immunother 56:1723–1731 Seil I, Frei C, Sultmann H, Knauer SK, Engels K, Jager E, Zatloukal K, Pfreundschuh M, Knuth A, Tseng-Chen Y, Jungbluth AA, Stauber RH, Jager D, 2007, The differentiation antigen NY-BR-1 is a potential target for antibody-based therapies in breast cancer. Int J Cancer 120:2635–2642 Woodard AH, Yu J, Dabbs DJ, Beriwal S, Florea AV, Elishaev E, Davison JM, Krasinskas AM, Bhargava R, 2011, NY-BR-1 and PAX8 immunoreactivity in breast, gynecologic tract, and other CK7+ carcinomas: potential use for determining site of origin. Am J Cl i n Pa t h o l 1 3 6 : 4 2 8 – 4 3 5 . d o i : 1 0 . 1 3 0 9 / AJCPUFNMEZ3MK1BK Balafoutas D, zur Hausen A, Mayer S, Hirschfeld M, Jaeger M, Denschlag D, Gitsch G, Jungbluth A, Stickeler E, 2013, Cancer testis antigens and NY-BR-1 expression in primary breast cancer: prognostic and therapeutic implications. BMC Cancer 13:271., DOI 10.1186/1471-2407-13-271 Liu H, 2014, Application of immunohistochemistry in breast pathology: a review and update. Arch Pathol Lab Med 138:1629– 1642., DOI 10.5858/arpa.2014-0094-RA Mesa-Tejada R, Palakodety RB, Leon JA, Khatcherian AO, Greaton CJ, 1988, Immunocytochemical distribution of a breast carcinoma associated glycoprotein identified by monoclonal antibodies. Am J Pathol 130:305–314 Loy TS, Chapman RK, Diaz-Arias AA, Bulatao IS, Bickel JT, 1991, Distribution of BCA-225 in adenocarcinomas. An immunohistochemical study of 446 cases. Am J Clin Pathol 96:326–329 Abramson VG, Lehmann BD, Ballinger TJ, Pietenpol JA, 2015, Subtyping of triple-negative breast cancer: implications for therapy. Cancer 121:8–16., DOI 10.1002/cncr.28914 Vranic S, Schmitt F, Sapino A, Costa JL, Reddy S, Castro M, Gatalica Z, 2013, Apocrine carcinoma of the breast: a comprehensive review. Histol Histopathol 28:1393–1409., DOI 10.14670/HH- 28.1393 Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C, Cowan D, Dressler L, Akslen LA, Ragaz J, Gown AM, Gilks CB, van de Rijn M, Perou CM, 2004, Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res 10:5367–5374 Sasahara M, Matsui A, Ichimura Y, Hirakata Y, Murata Y, Marui E, 2014, Overexpression of androgen receptor and forkhead-box A1 protein in apocrine breast carcinoma. Anticancer Res 34:1261– 1267 Kovari B, Rusz O, Schally AV, Kahan Z, Cserni G, 2014, Differential immunostaining of various types of breast carcinomas for growth hormone-releasing hormone, GHRH, receptor - apocrine epithelium and carcinomas emerging as uniformly positive. APMIS 122:824–831., DOI 10.1111/apm.12224 Ordonez NG, Sahin AA, 2014, Diagnostic utility of immunohistochemistry in distinguishing between epithelioid pleural mesotheliomas and breast carcinomas: a comparative study. Hum Pathol 45: 1529–1540., DOI 10.1016/j.humpath.2014.03.006 Krings G, Nystrom M, Mehdi I, Vohra P, Chen YY, 2014, Diagnostic utility and sensitivities of GATA3 antibodies in triplenegative breast cancer. Hum Pathol 45:2225–2232., DOI 10.1016/j. humpath.2014.06.022 Braxton DR, Cohen C, Siddiqui MT, 2015, Utility of GATA3 immunohistochemistry for diagnosis of metastatic breast carcinoma in cytology specimens. Diagn Cytopathol 43:271–277., DOI 10.1002/ dc.23206 Lew M, Pang JC, Jing X, Fields KL, RohMH, 2015, Young investigator challenge: the utility of GATA3 immunohistochemistry in the evaluation of metastatic breast carcinomas in malignant effusions. Cancer Cytopathol 123:576–581., DOI 10.1002/cncy.21574 Rakhshani N, Daryakar A, 2014, Are mammaglobin and GCDFP- 15 sensitive markers for diagnosis of metastatic basal-like triple negative breast carcinomas? Turk Patoloji Derg 30:18–22., DOI 10. 5146/tjpath.2013.01202 Darb-Esfahani S, von Minckwitz G, Denkert C, Ataseven B, Hogel B, Mehta K, Kaltenecker G, Rudiger T, Pfitzner B, Kittel K, Fiedler B,BaumannK, MollR, Dietel M, EidtmannH, Thomssen C, Loibl S, 2014, Gross cystic disease fluid protein 15, GCDFP-15, expression in breast cancer subtypes. BMC Cancer 14:546., DOI 10.1186/ 1471-2407-14-546 Pala EE, Bayol U, Cumurcu S, Keskin E, 2012, Immunohistochemical characteristics of triple negative/basal-like breast cancer. Turk Patoloji Derg 28:238–244., DOI 10.5146/tjpath. 2012.01130 Clark BZ, Beriwal S, Dabbs DJ, Bhargava R, 2014, Semiquantitative GATA-3 immunoreactivity in breast, bladder, gynecologic tract, and other cytokeratin 7-positive carcinomas. Am J Clin Pathol 142:64–71., DOI 10.1309/AJCP8H2VBDSCIOBF Gloyeske NC1, Woodard AH, Elishaev E, Yu J, Clark BZ, Dabbs DJ, Bhargava R, 2015, Immunohistochemical profile of breast cancer with respect to estrogen receptor and HER2 status. Appl Immunohistochem Mol Morphol 23:202–208., DOI 10.1097/PAI. 0000000000000076 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 259 EP 267 DI 10.1007/s12253-017-0246-y PG 9 ER PT J AU Cui, G Li, C Xu, G Sun, Z Zhu, L Li, Z Zheng, W Li, J Yuan, A AF Cui, Guanglin Li, Can Xu, Gang Sun, Zhenglu Zhu, Li Li, Zhengfen Zheng, Wei Li, Junling Yuan, Aping TI Tumor-Associated Fibroblasts and Microvessels Contribute to the Expression of Immunosuppressive Factor Indoleamine 2, 3-Dioxygenase in Human Esophageal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Fibroblasts; Endothelial cells; Indoleamine 2; 3-dioxygenase; Immunosuppression; carcinoma; esophagus ID Fibroblasts; Endothelial cells; Indoleamine 2; 3-dioxygenase; Immunosuppression; carcinoma; esophagus AB Recent studies have provided considerable evidence to support the hypothesis that tumor stroma plays a crucial role in the induction of immune tolerance to human cancers. Here, we investigated the contribution of reactive stromal tumorassociated fibroblasts (TAFs) and microvessels to the immunosuppressive factor indoleamine 2,3-dioxygenase (IDO) expression in the ESCC microenvironment. The immunohistochemical (IHC) analyses demonstrated a significant increased densities of TAFs and microvessels in the ESCC stroma, double IHCs showed that these increased TAFs and microvessels were with a high proliferation activity. Further IHC examinations revealed that increased expression of IDO were frequently observed in the stromal cells with TAF morphology and microvessels. Double immunofluorescence examinations confirmed the colocalization of IDO positive cells with SMA-alpha positive TAFs and CD34 positive endothelial cells in the ESCC stroma. Our current findings strongly suggest that the activated stromal TAFs and endothelial cells of microvessels contribute to the expression of IDO and then the orchestration of immunosuppressive microenvironment. C1 [Cui, Guanglin] the Second Affiliated Hospital of Zhengzhou University, Research Group of Gastrointestinal DiseasesZhengzhou, Henan, China. [Li, Can] the Second Affiliated Hospital of Zhengzhou University, Research Group of Gastrointestinal DiseasesZhengzhou, Henan, China. [Xu, Gang] the Second Affiliated Hospital of Zhengzhou University, Research Group of Gastrointestinal DiseasesZhengzhou, Henan, China. [Sun, Zhenglu] the Second Affiliated Hospital of Zhengzhou University, Research Group of Gastrointestinal DiseasesZhengzhou, Henan, China. [Zhu, Li] the Second Affiliated Hospital of Zhengzhou University, Research Group of Gastrointestinal DiseasesZhengzhou, Henan, China. [Li, Zhengfen] the Second Affiliated Hospital of Zhengzhou University, Research Group of Gastrointestinal DiseasesZhengzhou, Henan, China. [Zheng, Wei] the Second Affiliated Hospital of Zhengzhou University, Research Group of Gastrointestinal DiseasesZhengzhou, Henan, China. [Li, Junling] the Second Affiliated Hospital of Zhengzhou University, Research Group of Gastrointestinal DiseasesZhengzhou, Henan, China. [Yuan, Aping] the Second Affiliated Hospital of Zhengzhou University, Research Group of Gastrointestinal DiseasesZhengzhou, Henan, China. RP Cui, G (reprint author), the Second Affiliated Hospital of Zhengzhou University, Research Group of Gastrointestinal Diseases, Zhengzhou, China. EM guanglin.cui@yahoo.com CR Zhang Y, 2013, Epidemiology of esophageal cancer. World J Gastroenterol 19(34):5598–5606., DOI 10.3748/wjg.v19.i34.5598 Li MX, Cheng SJ, 1984, Carcinogenesis of esophageal cancer in Linxian, China. Chin Med J 97(5):311–316 Hofmeister V, Schrama D, Becker JC, 2008, Anti-cancer therapies targeting the tumor stroma. Cancer Immunol Immunother 57(1):1–17 Croci DO, Zacarias Fluck MF, Rico MJ, Matar P, Rabinovich GA, Scharovsky OG, 2007, Dynamic cross-talk between tumor and immune cells in orchestrating the immunosuppressive network at the tumor microenvironment. Cancer Immunol Immunother 56(11):1687–1700 Kim R, Emi M, Tanabe K, Arihiro K, 2006, Tumor-driven evolution of immunosuppressive networks duringmalignant progression. Cancer Res 66(11):5527–5536 Ishio T, Goto S, Tahara K, Tone S, Kawano K, Kitano S, 2004, Immunoactivative role of indoleamine 2,3-dioxygenase in human hepatocellular carcinoma. J Gastroenterol Hepatol 19(3):319–326 Karanikas V, Zamanakou M, Kerenidi T, Dahabreh J, Hevas A, Nakou M et al, 2007, Indoleamine 2,3-dioxygenase, IDO, expression in lung cancer. Cancer Biol Ther 6(8):1258–1262 Munn DH, Mellor AL, 2007, Indoleamine 2,3-dioxygenase and tumor-induced tolerance. J Clin Invest 117(5):1147–1154 Godin-Ethier J, Hanafi LA, Piccirillo CA, Lapointe R, 2011, Indoleamine 2,3-dioxygenase expression in human cancers: clinical and immunologic perspectives. Clin Cancer Res 17(22):6985– 6991., DOI 10.1158/1078-0432.CCR-11-1331 Muller AJ, Prendergast GC, 2007, Indoleamine 2,3-dioxygenase in immune suppression and cancer. Curr Cancer Drug Targets 7(1): 31–40 Ino K, Yoshida N, Kajiyama H, Shibata K, Yamamoto E, Kidokoro K et al, 2006, Indoleamine 2,3-dioxygenase is a novel prognostic indicator for endometrial cancer. Br J Cancer 95(11):1555–1561 Chung DJ, Rossi M, Romano E, Ghith J, Yuan J, Munn DH et al, 2009, Indoleamine 2,3-dioxygenase-expressing mature human monocyte-derived dendritic cells expand potent autologous regulatory T cells. Blood 114:555–563 Lob S, Konigsrainer A, Rammensee HG, Opelz G, Terness P, 2009, Inhibitors of indoleamine-2,3-dioxygenase for cancer therapy: can we see the wood for the trees? Nat Rev Cancer 9(6):445–452 Liu J, Lu G, Tang F, Liu Y, Cui G, 2009, Localization of indoleamine 2,3-dioxygenase in human esophageal squamous cell carcinomas. Virchows Arch 455(5):441–448., DOI 10.1007/s00428- 009-0846-3 Jia Y,Wang H,Wang Y,Wang T,Wang M, MaMet al, 2015, Low expression of Bin1, along with high expression of IDO in tumor tissue and draining lymph nodes, are predictors of poor prognosis for esophageal squamous cell cancer patients. Int J Cancer 137(5): 1095–1106., DOI 10.1002/ijc.29481 Gao YF, Peng RQ, Li J, Ding Y, Zhang X,Wu XJ et al, 2009, The paradoxical patterns of expression of indoleamine 2,3-dioxygenase in colon cancer. J Transl Med 7:71., DOI 10.1186/1479-5876-7-71 Gostner JM, Becker K, Uberall F, Fuchs D, 2015, The potential of targeting indoleamine 2,3-dioxygenase for cancer treatment. Expert Opin Ther Targets 19(5):605–615., DOI 10.1517/14728222.2014. 995092 Brandacher G, Perathoner A, Ladurner R, Schneeberger S, Obrist P, Winkler C et al, 2006, Prognostic value of indoleamine 2,3- dioxygenase expression in colorectal cancer: effect on tumorinfiltrating T cells. Clin Cancer Res 12(4):1144–1151 Selvan SR, Dowling JP, Kelly WK, Lin J, 2016, Indoleamine 2,3- dioxygenase, IDO): biology and target in cancer immunotherapies. Curr Cancer Drug Targets 16(9):755–764 Jochems C, Fantini M, Fernando RI, Kwilas AR, Donahue RN, Lepone LM et al, 2016, The IDO1 selective inhibitor epacadostat enhances dendritic cell immunogenicity and lytic ability of tumor antigen-specific T cells. Oncotarget 7(25):37762–37772., DOI 10. 18632/oncotarget.9326 Zhang G, Liu WL, Zhang L, Wang JY, Kuang MH, Liu P et al, 2011, Involvement of indoleamine 2,3-dioxygenase in impairing tumor-infiltrating CD8 T-cell functions in esophageal squamous cell carcinoma. Clin Dev Immunol 2011:384726., DOI 10.1155/ 2011/384726 Pan K, Wang H, Chen MS, Zhang HK, Weng DS, Zhou J et al, 2008, Expression and prognosis role of indoleamine 2,3-dioxygenase in hepatocellular carcinoma. J Cancer Res Clin Oncol 134(11):1247–1253 Liu J, Li Z, Cui J, Xu G, Cui G, 2012, Cellular changes in the tumor microenvironment of human esophageal squamous cell carcinomas. Tumour Biol 33(2):495–505., DOI 10.1007/s13277-011-0281-3 Cheng Y, Wang K, Ma W, Zhang X, Song Y, Wang J et al, 2015, Cancer-associated fibroblasts are associated with poor prognosis in esophageal squamous cell carcinoma after surgery. Int J Clin Exp Med 8(2):1896–1903 Munn DH,Mellor AL, 2016, IDO in the tumor microenvironment: inflammation, counter-regulation, and tolerance. Trends Immunol 37(3):193–207., DOI 10.1016/j.it.2016.01.002 Yuan A, Liu J, Liu Y, Bjornsen T, Varro A, Cui G, 2008, Immunohistochemical examination of gastrin, gastrin precursors, and gastrin/CCK-2 receptor in human esophageal squamous cell carcinomas. Pathol Oncol Res. 14(4):449–455 Yuan A, Liu J, Liu Y, CuiG(2007, Chromogranin A-positive tumor cells in human esophageal squamous cell carcinomas. Pathol Oncol Res 13(4):321–325 Cui G, Yuan A, Vonen B, Florholmen J, 2009, Progressive cellular response in the lamina propria of the colorectal adenoma-carcinoma sequence. Histopathology 54(5):550–560 Cui G, Qi H,Gundersen MD,Yang H, Christiansen I, SorbyeSWet al, 2015, Dynamics of the IL-33/ST2 network in the progression of human colorectal adenoma to sporadic colorectal cancer. Cancer Immunol Immunother 64(2):181–190., DOI 10.1007/s00262-014- 1624-x Yuan A, Steigen SE, Goll R, Vonen B, Husbekk A, Cui G et al, 2008, Dendritic cell infiltration pattern along the colorectal adenoma-carcinoma sequence. APMIS 116(6):445–456 Cui G, Koh TJ, Chen D, Zhao CM, Takaishi S, Dockray GJ et al, 2004, Overexpression of glycine-extended gastrin inhibits parietal cell loss and atrophy in the mouse stomach. Cancer Res 64(22): 8160–8166., DOI 10.1158/0008-5472.CAN-04-0876 Pollard JW, 2004, Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer 4(1):71–78 MuellerMM, Fusenig NE, 2004, Friends or foes - bipolar effects of the tumour stroma in cancer. Nat Rev Cancer 4(11):839–849., DOI 10.1038/nrc1477 Popov A, Schultze JL, 2008, IDO-expressing regulatory dendritic cells in cancer and chronic infection. J Mol Med 86(2):145–160 Takikawa O, 2005, Biochemical and medical aspects of the indoleamine 2,3-dioxygenase-initiated L-tryptophan metabolism. Biochem Biophys Res Commun 338(1):12–19 Pelak MJ, Snietura M, Lange D, Nikiel B, Pecka KM, 2015, The prognostic significance of indoleamine-2,3-dioxygenase and the receptors for transforming growth factor beta and interferon gamma in metastatic lymph nodes in malignant melanoma. Pol J Pathol 66(4):376–382 Sucher R, Kurz K,Weiss G, Margreiter R, Fuchs D, Brandacher G, 2010, IDO-mediated tryptophan degradation in the pathogenesis of malignant tumor disease. Int J Tryptophan Res 3:113–120 Sakurai K, Enomoto K,Amano S, Kimura T, Sugito K, Kimizuka K et al, 2004, Study of indoleamine 2,3-dioxygenase expression in patients of esophageal squamous cell carcinoma. Gan To Kagaku Ryoho 31(11):1780–1782 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 269 EP 275 DI 10.1007/s12253-017-0244-0 PG 7 ER PT J AU El-Balat, A Schmeil, I Karn, Th Becker, S Sanger, N Holtrich, U Arsenic, R AF El-Balat, Ahmed Schmeil, Iryna Karn, Thomas Becker, Sven Sanger, Nicole Holtrich, Uwe Arsenic, Ruza TI TFF3 Expression as Stratification Marker in Borderline Epithelial Tumors of the Ovary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ovarian cancer; Borderline tumors; Histological subtypes; Prognosis ID Ovarian cancer; Borderline tumors; Histological subtypes; Prognosis AB Borderline tumors (BOT) of the ovary account for 10% to 20% of ovarian neoplasms. Like ovarian cancer, BOT encompass several different histological subtypes (serous, mucinous, endometrioid, clear cell, transitional cell and mixed) with serous (SBOT) and mucinous (MBOT) the most common. Current hypotheses suggest low-grade serous carcinoma may develop in a stepwise fashion from SBOT whereas the majority of high grade serous carcinomas develop rapidly presumably from inclusion cysts or ovarian surface epithelium. The pathogenesis of mucinous ovarian tumors is still puzzling. Molecular markers could help to better define relationships between such entities. Trefoil factor-3 (TFF3) is an estrogen-regulated gene associated with prognosis in different types of cancer. It has also been included in a recent marker panel predicting subtypes of ovarian carcinoma. We analyzed the expression of TFF3 by immunohistochemistry in a cohort of 137 BOT and its association with histopathological features. Overall expression rate of TFF3 was 21.9%. None of the BOT with serous and endometrioid histology displayed strong TFF3 expression. On the other hand, TFF3 was highly expressed in 61.4% of MBOT cases and 33.3% of BOT with mixed histology (P < 0.001) suggesting a potential function of the protein in that subtypes. Associations of TFF3 expression with FIGO stage and micropapillary pattern were significant in the overall cohort but confounded by their correlation with histological subtypes. The highly specific expression of TFF3 in MBOT may help to further clarify potential relationships of tumors with mucinous histology and warrants further studies. C1 [El-Balat, Ahmed] Goethe University Frankfurt, Department of Obstetrics and Gynecology, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany. [Schmeil, Iryna] Goethe University Frankfurt, Department of Obstetrics and Gynecology, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany. [Karn, Thomas] Goethe University Frankfurt, Department of Obstetrics and Gynecology, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany. [Becker, Sven] Goethe University Frankfurt, Department of Obstetrics and Gynecology, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany. [Sanger, Nicole] Goethe University Frankfurt, Department of Obstetrics and Gynecology, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany. [Holtrich, Uwe] Goethe University Frankfurt, Department of Obstetrics and Gynecology, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany. [Arsenic, Ruza] Charite University Hospital, Institute of Pathology, Chariteplatz 1, 10117 Berlin, Germany. RP El-Balat, A (reprint author), Goethe University Frankfurt, Department of Obstetrics and Gynecology, 60590 Frankfurt, Germany. EM ahmed.el-balat@kgu.de CR Shih IM, Kurman RJ, 2005, Molecular pathogenesis of ovarian borderline tumors: new insights and old challenges. Clin Cancer Res 11(20):7273–7279., DOI 10.1158/1078-0432.CCR-05-0755 Skirnisdottir I, Garmo H, Wilander E, Holmberg L, 2008, Borderline ovarian tumors in Sweden 1960-2005: trends in incidence and age at diagnosis compared to ovarian cancer. Int J Cancer 123(8):1897–1901., DOI 10.1002/ijc.23724 Morice P, Uzan C, Fauvet R, Gouy S, Duvillard P, Darai E, 2012, Borderline ovarian tumour: pathological diagnostic dilemma and risk factors for invasive or lethal recurrence. Lancet Oncol 13(3): e103–e115., DOI 10.1016/S1470-2045(11)70288-1 Hart WR, 2005, Borderline epithelial tumors of the ovary. Mod Pathol 18(Suppl 2):S33–S50., DOI 10.1038/modpathol.3800307 du Bois A, Ewald-Riegler N, de Gregorio N, Reuss A, Mahner S, Fotopoulou C, Kommoss F, Schmalfeldt B, Hilpert F, Fehm T, Burges A, Meier W, Hillemanns P, Hanker L, Hasenburg A, Strauss HG, Hellriegel M, Wimberger P, Keyver-Paik MD, Baumann K, Canzler U, Wollschlaeger K, Forner D, Pfisterer J, Schroder W, Munstedt K, Richter B, Kommoss S, Hauptmann S, Arbeitsgmeinschaft Gynakologische Onkologie Study G, 2013, Borderline tumours of the ovary: a cohort study of the Arbeitsgmeinschaft Gynakologische Onkologie, AGO, study group. Eur J Cancer 49(8):1905–1914., DOI 10.1016/j.ejca.2013.01.035 Shih KK, Zhou Q, Huh J, Morgan JC, Iasonos A, Aghajanian C, Chi DS, Barakat RR, Abu-Rustum NR, 2011, Risk factors for recurrence of ovarian borderline tumors. Gynecol Oncol 120(3): 480–484., DOI 10.1016/j.ygyno.2010.11.016 Lenhard MS, Mitterer S, Kumper C, Stieber P, Mayr D, Ditsch N, Friese K, Burges A, 2009, Long-term follow-up after ovarian borderline tumor: relapse and survival in a large patient cohort. Eur J Obstet Gynecol Reprod Biol 145(2):189–194., DOI 10.1016/j. ejogrb.2009.04.031 Tinelli R, Tinelli A, Tinelli FG, Cicinelli E, Malvasi A, 2006, Conservative surgery for borderline ovarian tumors: a review. Gynecol Oncol 100(1):185–191., DOI 10.1016/j.ygyno.2005.09.021 Kaern J, Trope CG, Kristensen GB, Abeler VM, Pettersen EO, 1993, DNA ploidy; the most important prognostic factor in patients with borderline tumors of the ovary. Int J Gynecol Cancer 3(6):349–358 Ayhan A, Guvendag Guven ES, Guven S, Kucukali T, 2005, Recurrence and prognostic factors in borderline ovarian tumors. Gynecol Oncol 98(3):439–445., DOI 10.1016/j.ygyno.2005.05.033 Singer G, Oldt R 3rd, Cohen Y, Wang BG, Sidransky D, Kurman RJ, Shih Ie M, 2003, Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst 95(6):484–486 Seidman JD, Kurman RJ, 1996, Subclassification of serous borderline tumors of the ovary into benign and malignant types. A clinicopathologic study of 65 advanced stage cases. Am J Surg Pathol 20(11):1331–1345 Chan WY, Cheung KK, Schorge JO, Huang LW, Welch WR, Bell DA, Berkowitz RS, Mok SC, 2000, Bcl-2 and p53 protein expression, apoptosis, and p53 mutation in human epithelial ovarian cancers. Am J Pathol 156(2):409–417., DOI 10.1016/S0002-9440(10, 64744-X Longacre TA, McKenney JK, Tazelaar HD, Kempson RL, Hendrickson MR, 2005, Ovarian serous tumors of low malignant potential, borderline tumors): outcome-based study of 276 patients with long-term, > or =5-year, follow-up. Am J Surg Pathol 29(6): 707–723 Ortiz BH, Ailawadi M, Colitti C, Muto MG, DeaversM, Silva EG, Berkowitz RS, Mok SC, Gershenson DM, 2001, Second primary or recurrence? Comparative patterns of p53 and K-ras mutations suggest that serous borderline ovarian tumors and subsequent serous carcinomas are unrelated tumors. Cancer Res 61(19):7264– 7267 Emerson RE, Wang M, Liu F, Lawrence WD, Abdul-Karim FW, Cheng L, 2007, Molecular genetic evidence of an independent origin of serous low malignant potential implants and lymph node inclusions. Int J Gynecol Pathol 26(4):387–394., DOI 10.1097/pgp. 0b013e3180336287 Vang R, Shih IeM, Kurman RJ, 2013, Fallopian tube precursors of ovarian low- and high-grade serous neoplasms. Histopathology 62(1):44–58., DOI 10.1111/his.12046 Ates Ozdemir D, Usubutun A, 2016, PAX2, PAX8 and CDX2 expression in metastatic mucinous, primary ovarian mucinous and Seromucinous tumors and review of the literature. Pathol Oncol Res 22(3):593–599., DOI 10.1007/s12253-016-0040-2 Mok SC, Bell DA, Knapp RC, Fishbaugh PM, Welch WR, Muto MG, Berkowitz RS, Tsao SW, 1993, Mutation of K-ras protooncogene in human ovarian epithelial tumors of borderline malignancy. Cancer Res 53(7):1489–1492 Heinzelmann-Schwarz VA, Gardiner-Garden M, Henshall SM, Scurry JP, Scolyer RA, Smith AN, Bali A, Vanden Bergh P, Baron-Hay S, Scott C, Fink D, Hacker NF, Sutherland RL, O'Brien PM, 2006, A distinct molecular profile associated with mucinous epithelial ovarian cancer. Br J Cancer 94(6):904–913., DOI 10.1038/sj.bjc.6603003 Lau WH, Pandey V, Kong X, Wang XN, Wu Z, Zhu T, Lobie PE, 2015, Trefoil factor-3, TFF3, stimulates de novo angiogenesis in mammary carcinoma both directly and indirectly via IL-8/CXCR2. PLoS One 10(11):e0141947., DOI 10.1371/journal.pone.0141947 Ahmed ARH, Griffiths AB, Tilby MT, Westley BR, May FEB, 2012, TFF3 is a normal breast epithelial protein and is associated with differentiated phenotype in early breast cancer but predisposes to invasion and metastasis in advanced disease.AmJ Pathol 180(3): 904–916., DOI 10.1016/j.ajpath.2011.11.022 Perry JK, Kannan N, Grandison PM, Mitchell MD, Lobie PE, 2008, Are trefoil factors oncogenic? Trends Endocrinol Metab 19(2):74–81., DOI 10.1016/j.tem.2007.10.003 Mhawech-Fauceglia P, Wang D, Samrao D, Liu S, DuPont NC, Pejovic T, 2013, Trefoil factor family 3, TFF3, expression and its interaction with estrogen receptor, ER, in endometrial adenocarcinoma. Gynecol Oncol 130(1):174–180., DOI 10.1016/j.ygyno.2013. 03.030 Emami S, Rodrigues S, Rodrigue CM, Le Floch N, Rivat C, Attoub S, Bruyneel E, Gespach C, 2004, Trefoil factor family, TFF, peptides and cancer progression. Peptides 25(5):885–898., DOI 10.1016/ j.peptides.2003.10.019 PandeyV,Wu ZS, ZhangM, Li R, Zhang J, Zhu T, Lobie PE, 2014, Trefoil factor 3 promotes metastatic seeding and predicts poor survival outcome of patients with mammary carcinoma. Breast Cancer Res 16(5):429., DOI 10.1186/s13058-014-0429-3 Jatoi A, Vierkant RA, Hawthorne KM, Block MS, Ramus SJ, Larson NB, Fridley BL, Goode EL, 2016, Clinical and emergent biomarkers and their relationship to the prognosis of ovarian cancer. Oncology 90(2):59–68., DOI 10.1159/000442710 Kalloger SE, Kobel M, Leung S, Mehl E, Gao D, Marcon KM, Chow C, Clarke BA, Huntsman DG, Gilks CB, 2011, Calculator for ovarian carcinoma subtype prediction. Mod Pathol 24(4):512– 521., DOI 10.1038/modpathol.2010.215 LM MS, Altman DG, SauerbreiW, Taube SE, Gion M, Clark GM, Statistics Subcommittee of the NCIEWGoCD, 2005, Reporting recommendations for tumor marker prognostic studies, REMARK). J Natl Cancer Inst 97(16):1180–1184., DOI 10.1093/ jnci/dji237 Kurman RJ, Carcangiu,M.L., Herrington, C.S., Young, R.H, 2014, WHO classification of tumours of female reproductive organs. 4th ed. Lyon: International Agency for Research on Cancer, vol 6. Kjellev S, 2009, The trefoil factor family - small peptides with multiple functionalities. Cell Mol Life Sci 66(8):1350–1369., DOI 10.1007/s00018-008-8646-5 Thim L, Woldike HF, Nielsen PF, Christensen M, Lynch-Devaney K, Podolsky DK, 1995, Characterization of human and rat intestinal trefoil factor produced in yeast. Biochemistry 34(14):4757– 4764 Walker G, MacLeod K, Williams AR, Cameron DA, Smyth JF, Langdon SP, 2007, Estrogen-regulated gene expression predicts response to endocrine therapy in patients with ovarian cancer. Gynecol Oncol 106(3):461–468., DOI 10.1016/j.ygyno.2007.05.009 Kirikoshi H, KatohM(2002, Expression of TFF1, TFF2 and TFF3 in gastric cancer. Int J Oncol 21(3):655–659 Nowak M, Merz C, von Maessenhausen A, Vogel W, Boehm D, Svensson M, Carlsson J, Andren O, Perner S, 2015, Role of trefoil factor-3 peptide, TFF3, in prostate cancer progression. Lab invest 95:248a-248a Kurman RJ, Shih Ie M, 2010, The origin and pathogenesis of epithelial ovarian cancer: a proposed unifying theory. Am J Surg Pathol 34(3):433–443., DOI 10.1097/PAS.0b013e3181cf3d79 Seidman JD, Khedmati F, 2008, Exploring the histogenesis of ovarian mucinous and transitional cell, Brenner, neoplasms and their relationship with Walthard cell nests: a study of 120 tumors. Arch Pathol Lab Med 132(11):1753–1760., DOI 10.1043/1543- 2165-132.11.1753 Lee KR, Scully RE, 2000, Mucinous tumors of the ovary: a clinicopathologic study of 196 borderline tumors, of intestinal type, and carcinomas, including an evaluation of 11 cases with 'pseudomyxoma peritonei'. Am J Surg Pathol 24(11):1447–1464 Morito K, Nakamura J, Kitajima Y, Kai K, Tanaka T, Kubo H, Miyake S, NoshiroH, 2015, The value of trefoil factor 3 expression in predicting the longterm outcome and early recurrence of colorectal cancer. Int J Oncol 46(2):563–568., DOI 10.3892/ijo.2014.2755 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 277 EP 282 DI 10.1007/s12253-017-0240-4 PG 6 ER PT J AU Awkar, N Amireh, S Rai, S Shaaban, H Guron, G Maroules, M AF Awkar, Nelly Amireh, Sawsan Rai, Srijana Shaaban, Hamid Guron, Gunwant Maroules, Michael TI Association between Level of Tumor Markers and Development of VTE in Patients with Pancreatic, Colorectal and Ovarian Ca: Retrospective Case- Control Study in Two Community Hospitals SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tumor markers; Venous; Thromboembolism; Colorectal; Pancreatic; Ovarian ID Tumor markers; Venous; Thromboembolism; Colorectal; Pancreatic; Ovarian AB The risk of venous thromboembolism (VTE) is increased in patients with cancer. However, the role of tumor markers as potential indicators of increased risk of VTE is still undetermined. In this retrospective observational case control study, levels of the tumor markers CEA, CA 19–9 and CA 125 in patients with colorectal, pancreatic, and ovarian cancer respectively, who were admitted to two community hospitals between January 2001 and December 2011, were compared between patients who were VTE positive and those who were VTE negative. The primary goal of this study was to determine whether VTE positive cancer patients had higher tumor marker levels compared to VTE negative cancer patients. In our study, 66.7% (48/72) of patients who were positive for VTE had elevated tumor markers while 65.3% (66/101) of patients who were negative for VTE had low (normal) tumor markers, indicating an association of high tumor marker levels with the diagnosis of VTE. This was statistically significant with an odds ratio of 3.77 and p-value of <0.0001 (95% CI of 1.99–7.14). When the VTE group was further divided into DVT and PE groups, 70.2% (40/57) of patients in the DVT positive group had high tumor markers with a p value of <0.0001 and an odds ratio of 3.99 (95% CI of 2.02 to 7.89) while 57.9% (11/19) of patients in pulmonary embolism positive group had high tumor markers; this was, however, not statistically significant (p-value of 0.35 and a CI of 0.59 to 4.10). In this retrospective study of 173 individuals with a diagnosis of either colorectal, pancreatic, or ovarian Cancer, higher tumor marker levels (CEA, CA 19–9, and CA 125 respectively) were associated with an increased risk of VTE, either DVT or PE. However, when further divided into either DVT or PE groups, the association remained statistically significant only for DVT but not for PE. C1 [Awkar, Nelly] St Joseph’s Regional Medical Center, Department of Medical OncologyPaterson, NJ, USA. [Amireh, Sawsan] St Joseph’s Regional Medical Center, Seton Hall University School of Health and Medical Sciences, St Michael’s Medical Center, 111 Central AvenueNewark, NJ, USA. [Rai, Srijana] St Joseph’s Regional Medical Center, Seton Hall University School of Health and Medical Sciences, St Michael’s Medical Center, 111 Central AvenueNewark, NJ, USA. [Shaaban, Hamid] St Joseph’s Regional Medical Center, Seton Hall University School of Health and Medical Sciences, St Michael’s Medical Center, 111 Central AvenueNewark, NJ, USA. [Guron, Gunwant] St Joseph’s Regional Medical Center, Seton Hall University School of Health and Medical Sciences, St Michael’s Medical Center, 111 Central AvenueNewark, NJ, USA. [Maroules, Michael] St Joseph’s Regional Medical Center, Department of Medical OncologyPaterson, NJ, USA. RP Shaaban, H (reprint author), St Joseph’s Regional Medical Center, Seton Hall University School of Health and Medical Sciences, St Michael’s Medical Center, Newark, USA. EM hamidshaaban@gmail.com CR Buller HR, Van Doormaal FF, Van Sluis GL, Kamphuisen PW, 2007, Cancer and thrombosis: frommolecular mechanisms to clinical presentations. J Thromb Haemost 5(s1):246–254 Lyman GH, 2011, Venous thromboembolism in the patient with cancer. Cancer 117(7):1334–1349 Chee CE, Ashrani AA, Marks RS, Petterson TM, Bailey KR, Melton LJ, Heit JA, 2014, Predictors of venous thromboembolism recurrence and bleeding among active cancer patients: a population-based cohort study. Blood 123(25):3972–3978 Khorana AA, 2007, The NCCN clinical practice guidelines on venous thromboembolic disease: strategies for improving VTE prophylaxis in hospitalized cancer patients. Oncologist 12(11):1361– 1370 Prandoni P, Falanga A, Piccioli A, 2005, Cancer and venous thromboembolism. Lancet Oncol 6(6):401–410 Khorana AA, Francis CW, Culakova E, Kuderer NM, Lyman GH, 2007, Frequency, risk factors, and trends for venous thromboembolism among hospitalized cancer patients. Cancer 110(10):2339– 2346 Pabinger I, Thaler J, Ay C, 2013, Biomarkers for prediction of venous thromboembolism in cancer. Blood 122(12):2011–2018 ZhangY, Yang Y, ChenW, Guo L, Liang L, Zhai Z,Wang C, 2014, Prevalence and associations of VTE in patients with newly diagnosed lung cancer. CHEST Journal 146(3):650–658 Saadeh FA, Norris L, O’Toole S, Gleeson N, 2013, Venous thromboembolism in ovarian cancer: incidence, risk factors and impact on survival. Eur J Obstet Gynecol Reprod Biol 170(1):214–218 Wu X, Xue X, Tang J, Cheng X, Tian W, Jiang R, Zang R, 2013, Evaluation of risk factors for venous thromboembolism in Chinese women with epithelial ovarian cancer. Int J Gynecol Cancer 23(1): 65–72 Khorana AA, Kuderer NM, Culakova E, Lyman GH, Francis CW, 2008, Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood 111(10):4902–4907 Ay C, Dunkler D, Marosi C, Chiriac AL, Vormittag R, Simanek R, Quehenberger P, Zielinski C, Pabinger I, 2010, Prediction of venous thromboembolism in cancer patients. Blood 116(24):5377– 5382 Khorana AA, RaoMV(2007, Approaches to risk-stratifying cancer patients for venous thromboembolism. Thromb Res 120:S41–S50 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 283 EP 287 DI 10.1007/s12253-017-0239-x PG 5 ER PT J AU Aumayr, K Fathi, O Traub-Weidinger, T Niederle, B Koperek, O AF Aumayr, Klaus Fathi, Osmen Traub-Weidinger, Tatjana Niederle, Bruno Koperek, Oskar TI Expression of Hypoxia-Associated Protein HIF-1α in Follicular Thyroid Cancer is Associated with Distant Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HIF-1α; Hypoxia-associated proteins; Tenascin; Follicular thyroid cancer; Desmoplastic stroma reaction; Metastasis ID HIF-1α; Hypoxia-associated proteins; Tenascin; Follicular thyroid cancer; Desmoplastic stroma reaction; Metastasis AB Follicular thyroid carcinomas (FTCs) are the second most common malignant neoplasia of the thyroid and in general its prognosis is quite favorable. However, the occurrence of metastases or non-responsiveness to radioiodine therapy worsens the prognosis considerably. We evaluated immunohistochemically the expression of hypoxiaassociated proteins by hypoxia-induced factor 1α (HIF-1α), the stroma-remodeling marker Tenascin C, as well as markers for the epithelial-mesenchymal transition (EMT), namely Ecadherin and slug in a series of 59 sporadic FTCs. In addition, various clinicopathologic parameters were assessed like TNM-staging, age, tumor size as well as tumor characteristics like desmoplasia, necrosis, and calcification. Overexpression of HIF-1α was seen in 29 of 59 tumors (49.2%) including 21 (35.6%) FTC with strong expression of tumor cell groups. HIF-1α correlated significantly with metastasis (p < 0.001; Mann-Whitney U test), degree of desmoplasia (p = 0.042, Kruskal-Wallis test), tenascin C expression (p = 0.042, Kruskal-Wallis test), calcification (p < 0.025, Kruskal–Wallis test), necrosis (p = 0.002), age (p = 0.011, Kruskal-Wallis test) and tumor stage UICC (p = 0.022, Kruskal-Wallis test). Furthermore, metastasis was associated with the degree of desmoplasia (p = 0.014; Fisher’s exact test), calcification (p = 0.008, Fisher’s exact test), necrosis (p = 0.042, Fisher’s exact test), tumor size (p = 0.015, Mann-Whitney U test), and age (p = 0.001, Mann-Whitney U test). In a Cox proportional hazards model, only metastasis remained as an independent risk factor for overall survival (hazard rate: 10.2 [95% CI, 02.19 to 47.26]; p = 0.003). Our data suggest that HIF-1α plays a critical role in the remodeling of the extracellular matrix as well as metastasizing process of follicular thyroid carcinoma and targeting hypoxia-associated and -regulated proteins may be considered as potential targets for personalized medicine. C1 [Aumayr, Klaus] Medical University of Vienna, Department of Pathology, Waehringer Guertel 18-20, 1090 Vienna, Austria. [Fathi, Osmen] Medical University of Vienna, Department of Pathology, Waehringer Guertel 18-20, 1090 Vienna, Austria. [Traub-Weidinger, Tatjana] Medical University of Vienna, Department of RadiologyVienna, Austria. [Niederle, Bruno] Medical University of Vienna, Department of SurgeryVienna, Austria. [Koperek, Oskar] Medical University of Vienna, Department of Pathology, Waehringer Guertel 18-20, 1090 Vienna, Austria. RP Koperek, O (reprint author), Medical University of Vienna, Department of Pathology, 1090 Vienna, Austria. EM oskar.koperek@meduniwien.ac.at CR Desgrosellier JS, Cheresh DA, 2010, Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer 10:9– 22., DOI 10.1038/nrc2748 Grivennikov SI, Greten FR, Karin M, 2010, Immunity, inflammation, and cancer. Cell 140:883–899., DOI 10.1016/j. cell.2010.01.025 Birner P, Schindl M, Obermair A, Plank C, Breitenecker G, Oberhuber G, 2000, Overexpression of hypoxia-inducible factor 1alpha is a marker for an unfavorable prognosis in early-stage invasive cervical cancer. Cancer Res 60:4693– 4696 Kitada T, Seki S, Sakaguchi H, Sawada T, Hirakawa K, Wakasa K, 2003, Clinicopathological significance of hypoxia-inducible factor- 1alpha expression in human pancreatic carcinoma. Histopathology 43:550–555 Maxwell PH, Dachs GU, Gleadle JM, Nicholls LG, Harris AL, Stratford IJ, Hankinson O, Pugh CW, Ratcliffe PJ, 1997, Hypoxia-inducible factor-1 modulates gene expression in solid tumors and influences both angiogenesis and tumor growth. ProcNatl Acad Sci USA 94:8104–8109 Pugh CW, Gleadle J, Maxwell PH, 2001, Hypoxia and oxidative stress in breast cancer. Hypoxia signalling pathways. Breast Cancer Res: BCR 3:313–317 Schindl M, Schoppmann SF, Samonigg H, Hausmaninger H, Kwasny W, Gnant M, Jakesz R, Kubista E, Birner P, Oberhuber G, Group ABaCCS, 2002, Overexpression of hypoxia-inducible factor 1alpha is associated with an unfavorable prognosis in lymph node-positive breast cancer. Clin Cancer Res 8:1831–1837 Zalatnai A, 2006, Molecular aspects of stromal-parenchymal interactions in malignant neoplasms. Curr Mol Med 6:685– 693 De Wever O, Mareel M, 2003, Role of tissue stroma in cancer cell invasion. J Pathol 200:429–447., DOI 10.1002/ path.1398 Yoshida T, Akatsuka T, Imanaka-Yoshida K, 2015, Tenascin-C and integrins in cancer. Cell Adhes Migr 9:96–104., DOI 10.1080/ 19336918.2015.1008332 Dong W, Qin G, Shen R, 2016, Rab11-FIP2 promotes the metastasis of gastric cancer cells. Int J Cancer 138:1680–1688., DOI 10. 1002/ijc.29899 Huang CH, Yang WH, Chang SY, Tai SK, Tzeng CH, Kao JY, Wu KJ, Yang MH, 2009, Regulation of membrane-type 4 matrix metalloproteinase by SLUG contributes to hypoxia-mediated metastasis. Neoplasia 11:1371–1382 Rankin EB, Giaccia AJ, 2016, Hypoxic control of metastasis. Science 352:175–180., DOI 10.1126/science.aaf4405 Serrano-Gomez SJ, Maziveyi M, Alahari SK, 2016, Regulation of epithelial-mesenchymal transition through epigenetic and posttranslational modifications. Mol Cancer 15:18 Thiery JP, 2002, Epithelial-mesenchymal transitions in tumour progression nature reviews. Cancer 2:442–454., DOI 10.1038/nrc822 Chow SM, Law SC, Mendenhall WM, Au SK, Yau S, Yuen KT, Law CC, Lau WH, 2002, Follicular thyroid carcinoma: prognostic factors and the role of radioiodine. Cancer 95:488–498., DOI 10. 1002/cncr.10683 Ito Y, Hirokawa M, Masuoka H, Yabuta T, Fukushima M, Kihara M, Higashiyama T, Takamura Y, Kobayashi K, Miya A, Miyauchi A, 2013, Distant metastasis at diagnosis and large tumor size are significant prognostic factors of widely invasive follicular thyroid carcinoma. Endocr J 60:829–833 Ito Y, Miyauchi A, Tomoda C, HirokawaM, Kobayashi K, Miya A, 2013, Prognostic significance of patient age in minimally and widely invasive follicular thyroid carcinoma: investigation of three age groups. Endocr J 61:265–271 DeLellis RA, 2006, Pathology and genetics of thyroid carcinoma J Surg Oncol 94:662–669., DOI 10.1002/jso.20700 Sobin L, Gospodarowicz M, Wittekind C, 2009, TNM classification of malignant tumours. Wiley-Blackwell, New York Ghossein RA, Hiltzik DH, Carlson DL, Patel S, Shaha A, Shah JP, Tuttle RM, Singh B, 2006, Prognostic factors of recurrence in encapsulated Hurthle cell carcinoma of the thyroid gland: a clinicopathologic study of 50 cases. Cancer 106:1669–1676., DOI 10.1002/ cncr.21825 Lang W, Choritz H, Hundeshagen H, 1986, Risk factors in follicular thyroid carcinomas. A retrospective follow-up study covering a 14-year period with emphasis on morphological findings. Am J Surg Pathol 10:246–255 Burrows N, Resch J, Cowen RL, vonWasielewski R, Hoang-Vu C, West CM, Williams KJ, Brabant G, 2010, Expression of hypoxiainducible factor 1 alpha in thyroid carcinomas. Endocr Relat Cancer 17:61–72., DOI 10.1677/ERC-08-0251 Ito Y, Miyauchi A, Tomoda C, HirokawaM, Kobayashi K, Miya A, 2014, Prognostic significance of patient age in minimally and widely invasive follicular thyroid carcinoma: investigation of three age groups. Endocr J 61:265–271 Koperek O, Akin E, Asari R, Niederle B, Neuhold N, 2013, Expression of hypoxia-inducible factor 1 alpha in papillary thyroid carcinoma is associated with desmoplastic stromal reaction and lymph node metastasis. Virchows Archiv : Int J Pathol 463:795– 802., DOI 10.1007/s00428-013-1484-3 Koperek O, Bergner O, Pichlhofer B, Oberndorfer F, Hainfellner JA, Kaserer K, Horvat R, Harris AL, Niederle B, Birner P, 2011, Expression of hypoxia-associated proteins in sporadic medullary thyroid cancer is associated with desmoplastic stroma reaction and lymph node metastasis and may indicate somatic mutations in the VHL gene. J Pathol 225:63–72 Semenza G, 2012, Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci 33:207–214 Birner P, Gatterbauer B, Oberhuber G, Schindl M, Rossler K, Prodinger A, Budka H, Hainfellner JA, 2001, Expression of hypoxia-inducible factor-1 alpha in oligodendrogliomas: its impact on prognosis and on neoangiogenesis. Cancer 92:165–171 Cleven AHG, van Engeland M, Wouters BG, de Bruine AP, 2007, Stromal expression of hypoxia regulated proteins is an adverse prognostic factor in colorectal carcinomas. Cell Oncol 29:229–240 Schoppmann SF, Fenzl A, Schindl M, Bachleitner-Hofmann T, Nagy K, Gnant M, Horvat R, Jakesz R, Birner P, 2006, Hypoxia inducible factor-1alpha correlates with VEGF-C expression and lymphangiogenesis in breast cancer. Breast Cancer Res Treat 99: 135–141 Bertout JA, Patel SA, Simon MC, 2008, The impact of O2 availability on human cancer. Nat Rev Cancer 8:967–975., DOI 10.1038/ nrc2540 Dery M, Michaud M, Richard D, 2005, Hypoxia-inducible factor 1: regulation by hypoxic and non-hypoxic activators. Int J Biochem Cell Biol 37:535–540 Erler JT, Giaccia AJ, 2006, Lysyl oxidase mediates hypoxic control of metastasis. Cancer Res 66:10238–10241., DOI 10.1158/0008- 5472.CAN-06-3197 HockelM, Vaupel P, 2001, Tumor hypoxia: definitions and current clinical, biologic, andmolecular aspects. J Natl Cancer Inst 93:266– 276 Sullivan R, Graham CH, 2007, Hypoxia-driven selection of the metastatic phenotype. Cancer Metastasis Rev 26:319–331., DOI 10. 1007/s10555-007-9062-2 Rapisarda A, Shoemaker RH, Melillo G, 2009, Antiangiogenic agents and HIF-1 inhibitors meet at the crossroads. Cell Cycle, Georgetown, Tex, 8:4040–4043 Burrows N, Babur M, Resch J, Ridsdale S, Mejin M, Rowling EJ, Brabant G,Williams KJ, 2011, GDC-0941 inhibits metastatic characteristics of thyroid carcinomas by targeting both the phosphoinositide-3 kinase, PI3K, and hypoxia-inducible factor- 1α, HIF-1α, pathways. J Clin Endocrinol Metabol 96:E1934– E1943., DOI 10.4061/2011/762905 Manalo D, Rowan A, Lavoie T, Natarajan L, Kelly B, Ye S, Garcia J, Semenza G, 2005, Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF-1. Blood 105:659–669 Chen S, Chen JZ, Zhang JQ, Chen HX, Yan ML, Huang L, Tian YF, Chen YL,Wang YD, 2016, Hypoxia induces TWIST-activated epithelial-mesenchymal transition and proliferation of pancreatic cancer cells in vitro and in nude mice. Cancer Lett 383:73–84., DOI 10.1016/j.canlet.2016.09.027 Lan L, Luo Y, Cui D, Shi BY, DengW, Huo LL, Chen HL, Zhang GY, Deng LL, 2013, Epithelial-mesenchymal transition triggers cancer stem cell generation in human thyroid cancer cells. Int J Oncol 43:113–120., DOI 10.3892/ijo.2013.1913 Giannoni E, Bianchini F, Calorini L, Chiarugi P, 2011, Cancer associated fibroblasts exploit reactive oxygen species through a proinflammatory signature leading to epithelial mesenchymal transition and stemness. Antioxid Redox Signal 14:2361–2371 Zhu GH, Huang C, Feng ZZ, Lv XH, Qiu ZJ, 2013, Hypoxia-induced snail expression through transcriptional regulation by HIF-1α in pancreatic cancer cells. Dig Dis Sci 58:3503–3515 Chiquet-Ehrismann R, Tucker RP, 2004, Connective tissues: signalling by tenascins. Int J Biochem Cell Biol 36:1085–1089., DOI 10.1016/j.biocel.2004.01.007 Elvidge GP, Glenny L, Appelhoff RJ, Ratcliffe PJ, Ragoussis J, Gleadle JM, 2006, Concordant regulation of Gene expression by hypoxia and 2-Oxoglutarate-dependent dioxygenase inhibition: the role of HIF-1, HIF-2, and other pathways. J Biol Chem 281:15215– 15226 Colpaert C, Vermeulen P, van Beest P, Goovaerts G, Weyler J, Van DamP, Dirix L, VanMarck E, 2001, Intratumoral hypoxia resulting in the presence of a fibrotic focus is an independent predictor of early distant relapse in lymph node-negative breast cancer patients. Histopathology 39:416–425 Couvelard A, O'Toole D, Leek R, Turley H, Sauvanet A, Degott C, Ruszniewski P, Belghiti J, Harris AL, Gatter K, Pezzella F, 2005, Expression of hypoxia-inducible factors is correlated with the presence of a fibrotic focus and angiogenesis in pancreatic ductal adenocarcinomas. Histopathology 46:668–676 Takacova M, Bullova P, Simko V, Skvarkova L, Poturnajova M, Feketeova L, Babal P, Kivela AJ, Kuopio T, Kopacek J, Pastorek J, Parkkila S, Pastorekova S, 2014, Expression pattern of carbonic anhydrase IX in medullary thyroid carcinoma supports a role for RET-mediated activation of the HIF pathway. Am J Pathol 184: 953–965, DOI 10.1016/j.ajpath.2014.01.002 Faam B, Ghaffari MA, Ghadiri A, Azizi F, 2015, Epigenetic modifications in human thyroid cancer. Biomed Reports 3:3–8., DOI 10. 3892/br.2014.375 Nikiforov YE, 2011, Molecular analysis of thyroid tumors. Mod Pathol : Off J U S Can Acad Pathol, Inc 24:S34., DOI 10.1038/ modpathol.2010.167 Ebos JML, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS, 2009, Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15: 232–239 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 289 EP 296 DI 10.1007/s12253-017-0232-4 PG 8 ER PT J AU Gaceb, H Cherbal, F Bakour, R Ould-Rouis, A Mahfouf, H AF Gaceb, Hadjer Cherbal, Farid Bakour, Rabah Ould-Rouis, Abdelhalim Mahfouf, Hassen TI Clinicopathological and Molecular Study of Triple-Negative Breast Cancer in Algerian Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Algerian women; Breast cancer; Triple negative; Clinicopathological characteristics; BRCA1 mutations; Genetic testing ID Algerian women; Breast cancer; Triple negative; Clinicopathological characteristics; BRCA1 mutations; Genetic testing AB Triple-negative breast cancer (TNBC) is associated with aggressive tumor behavior, poor prognosis and BRCA1 mutations. There are limited data regarding TNBC among Algerian women. In this study, we sought to determine clinical and tumor characteristics associated with TNBC. We also screened for the prevalence of BRCA1 mutations in unselected cohort of TNBC patients. Clinical and tumor characteristics data of 877 breast cancer patients diagnosed between 2011 and 2015, were collected from cancer registry of public hospital of Rouiba. Patients were divided in two groups: those with TNBC and those with other breast cancer subtypes. Differences between the two groups with regard to clinical and tumor characteristics were compared using Fisher’s exact test. BRCA1 mutations analysis was performed in unselected cohort of 103 women with TNBC, including all exons where a mutation was previously found in Algerian population (exons 2, 3, 5, 11). The median age at diagnosis for TNBC and non- TNBC patients was 47.4 years and 49.4 years, respectively. The proportion of TNBC was 19.95%. Our data showed significant differences in menopausal status, TNM stage, histological type, tumor histological grade, Ki67 expression and family history of breast cancer between TNBC and non- TNBC patients. Four distinct deleterious mutations in BRCA1 gene were detected in eight young TNBC patients. TNBC is associated with young age, poor histopathological characteristics and family history of breast cancer. BRCA1 mutations have been detected in young TNBC patients. TNBC phenotype should be added as criterion to screen for BRCA1 mutations in Algerian women. C1 [Gaceb, Hadjer] USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, El Alia, Bab Ezzouar, 16111 Algiers, Algeria. [Cherbal, Farid] USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, El Alia, Bab Ezzouar, 16111 Algiers, Algeria. [Bakour, Rabah] USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, El Alia, Bab Ezzouar, 16111 Algiers, Algeria. [Ould-Rouis, Abdelhalim] USTHB, Faculty of Biological Sciences, Laboratory of Dynamics and BiodiversityAlgiers, Algeria. [Mahfouf, Hassen] University of Algiers, School of Medicine, Public Hospital Academic Medical Oncology ServicesRouiba, Algeria. RP Cherbal, F (reprint author), USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, 16111 Algiers, Algeria. EM farid.cherbal@gmail.com CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008 GLOBOCAN. Int J Cancer 127:2893–2917 GLOBOCAN: Breast cancer incidence and mortality worldwide, 2012, International agency for research on cancer; 2012. http:// globocan.iarc.fr/old/FactSheets/cancers/breast-new.asp. Accessed 21 March 2016 Podo F, Buydens LMC, Degani H et al, 2010, Triple_negative breast cancer: present challenges and new perspectives. Mol Oncol 4:209–229 Carey L, Winer E, Viale G, Cameron D, Gianni L, 2010, Triple negative breast cancer: disease entity or title of convenience? Nat Rev Clin Oncol 7:683–692 Dent R, TrudeauM, Pritchard KI et al, 2007, Triple negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res 13:4429–4434 Carey LA, Perou CM, Livasy CA et al, 2006, Race, breast cancer subtypes, and survival in the Carolina breast cancer study. JAMA 295:2492–2502 Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V, 2007, Descriptive analysis of estrogen receptor, ER)-negative, progesterone receptor, PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population- based study from the California cancer. Registry 109:1721–1728 Huo D, Ikpatt F, KhramtsovAet al, 2009, Population differences in breast cancer: survey in indigenous African women reveals overrepresentation of triple negative breast cancer. J Clin Oncol 27: 4515–4521 Stark A, KleerCG,Martin I et al, 2010, African ancestry and higher prevalence of triple negative breast cancer: findings from an international study. Cancer 116:4926–4932 Amirikia KC, Mills P, Bush J, Newman LA, 2011, Higher population-based incidence rates of triple-negative breast cancer among young African-American women: implications for breast cancer screening recommendations. Cancer 117:2747–2753 Ly M, Antoine M, Dembele AK et al, 2012, High incidence of triple-negative tumors in sub-Saharan Africa: a prospective study of breast cancer characteristics and risk factors in Malian women seen in a Bamako university hospital. Oncology 83:257–263 Su Y, Zheng Y, Zheng W et al, 2011, Distinct distribution and prognostic significance of molecular subtypes of breast cancer in Chinese women: a population- based cohort study. BMC Cancer 11:292 Banegas MP, Tao L, Altekruse S et al, 2014, Heterogeneity of breast cancer sub-types and survival among Hispanic women with invasive breast cancer in California. Breast Cancer Res Treat 144: 625–634 Singh M, Ding Y, Zhang LY et al, 2014, Distinct breast cancer subtypes in women with early- onset disease across races. Am J Cancer Res 4:337–352 Fourati A, Boussen H, El MayMVet al, 2014, Descriptive analysis of molecular subtypes in Tunisian breast cancer. Asia Pac J Clin Oncol 10:e69–e74 Bosch A, Eroles P, Zaragoza R, Vina JR, Lluch A, 2010, Triple-negative breast cancer: molecular features, pathogenesis, treatment and current lines of research. Cancer Treat Rev 36:206–205 Wong ESY, Shekar S, Chan CHTet al Predictive factors for BRCA1 and BRCA2 genetic testing in an Asian clinic-based population. PLoS ONE 7:e0134408., DOI 10.1371/journal.pone.0134408 De Ruitjter TC, Veeck J, Hoon JPJ, Van EngelandM, Tjan-Heijnen VC, 2011, Characteristics of triple-negative breast cancer. J Cancer Res Clin Oncol 137:183–192 Robertson L, Hanson H, Seal S et al, 2012, BRCA1 testing should be offered to individuals with triple-negative breast cancer diagnosed below 50 years. Br J Cancer 106:1234–1238 Stevens KN, Vachon CM, Couch FJ, 2012, Genetic susceptibility to triple-negative breast cancer. Cancer Res 7:2025–2030 Pern F, Bogdanova N, Schurmann LM et al, 2012, Mutation analysis of BRCA1, BRCA2, PALB2 and BRD7 in a hospital based series of German patients with triple-negative breast cancer. PLoS One 7: e47993., DOI 10.1371/journal.pone.0047993 Fong PC, Boss DS, Yap TA et al, 2009, Inhibition of poly, ADPribose, polymerase in tumors from BRCA mutation carriers. N Engl J Med 361:123–134 Tutt A, Robson M, Garber JE et al, 2010, Oral poly(ADP-ribose, polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet 376:235–244 Cherbal F, Bakour R, Adane S, Boualga K, 2013, BRCA1 and BRCA2 germline mutation spectrum in hereditary breast/ovarian cancer families from Maghrebian countries. Breast Dis 34:1–8 Sobin LH, Wittekind CH, 2002, Breast tumors. In: Sobin LH, Wittekinds CH, eds, Classification of malignant tumors, 6th edn. Wiley-Liss, Geneva, pp 131–142 Pennacchia I, Carbone A, Di Cerbo A, Vecchio FM, Arena V, 2015, 2013 ASCO/CAP updated guidelines for human epidermal growth factor receptor 2 testing: impact on routine practice. Breast 24:285e6 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, Panel members, 2011, Strategies for sub- types – dealing with the diversity of breast cancer: highlights of the St Gallen international expert consensus on the primary therapy of early breast cancer 2011. Ann Oncol 22:1736–1747., DOI 10.1093/annonc/ mdr304 Coates AS,Winer EP, Goldhirsch A, Gelber RD, Gnant M, Piccart- Gebhart M, Thurlimann B, Senn HJ, Panel Members, 2015, Tailoring therapies-improving the management of early breast cancer: St Gallen international expert consensus on the primary therapy of early breast cancer 2015. Ann Oncol 26:1533–1546., DOI 10. 1093/annonc/mdv221 Cherbal F, Bakour R, Adane S, Boualga K, Benais-Pont G, Maillet P, 2010, BRCA1 and BRCA2 germline mutations screening in Algerian breast/ovarian cancer families. Dis Markers 28:377–384 Rais G, Raissouni S, Aitelhaj M et al, 2012, Triple-negative breast cancer in Moroccan women: clinicopathological and therapeutic study at the National Institute of oncology. BMC Womens Health 12:35 Minicozzi P, Bella F, Toss A et al, 2013, Relative and disease-free survival for breast cancer in relation to subtype: a population-based study. J Cancer Res Clin Oncol 139:1569–1577 Clarke CA, Keegan TH, Yang J et al, 2012, Age-specific incidence of breast cancer subtypes: understanding the black-white crossover. J Natl Cancer Inst 104:1094–1101 Rakha EA, El-Sayed ME, Green AR, 2007, Prognostic markers in triple-negative breast cancer. Cancer 109:25–32 Ann J, Kang S, Kwun K, 2008, Clinicopathologic characteristics of triple negative breast cancer in early stages. Eur J Cancer Supp 6: 183 Qiu J, Xue X, Hu C et al, 2016, Comparison of Clinicopathological features and prognosis in triple-negative and non-triple negative breast cancer. J Cancer 7:167–173 Li CY, Zhang S, Zhang XB, Wang P, Hou GF, Zhang J, 2013, Clinicopathological and prognostic characteristics of triplenegative breast cancer, TNBC, in Chinese patients: a retrospective study. Asian Pac J Cancer Prev 14:3779–3784 Collaborative group on Hormonal factors in Breast Cancer, 2012, Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet 360:187–195 Anothaisintawee T, Wiratkapun C, Lerdsitthichai P et al, 2013, Risk factors of breast cancer: a systematic revew and meta-analysis. Asia Pac J Public Health 25:368–4943 Islami F, Liu Y, Jemal A et al, 2015, Breastfeeding and breast cancer risk by receptor status- a systematic review and meta-analysis. Ann Oncol 26:2398–2407 Shinde SS, Forman MR, Kuerer HM et al, 2010, Higher parity and shorter breast feeding duration: association with triple-negative phenotype of breast cancer. Cancer 116:4933–4943 Jung SY, Jeong J, Shin SH et al, 2010, The invasive lobular carcinoma as a prototype luminal a breast cancer: a retrospective cohort study. BMC Cancer 10:664 Koo JS, JungWH, 2011, Clinicopathlogic and Immunohistochemical characteristics of triple negative invasive lobular carcinoma. Yonsei Med J 52(1):89–97 Lee E, McKean-Cowdin R, Ma H et al, 2011, Characteristics of triple negative breast cancer in patients with a BRCA1 mutation: results from a population-based study of young women. J Clin Oncol 29:4373–4380 Wong-Brown MW, Meldrum CJ, Carpenter JE et al, 2015, Prevalence of BRCA1 and BRCA2 germlinemutations in patients with triple-negative breast cancer. Breast Cancer Res Treat 50:71–80 Foulkes WD, Metcalfe K, Sun P et al, 2004, Estrogen receptor status in BRCA1-BRCA2-related breast cancer: the influence of age, grade, and histological type. Clin Cancer Res 10:2029–2034 Gonzalez-Angulo AM, Timms KM, Liu S et al, 2011, Incidence and outcome of BRCA mutations in unselected patients with triple receptor-negative breast cancer. Clin Cancer Res 17:1082–1089 Hartman AR, Kaldate RR, Sailer LM et al, 2012, Prevalence of BRCA mutations in an unselected population of triple-negative breast cancer. Cancer 118:2787–2795 Rummel S, Varner E, Shriver CD, Ellsworth RE, 2013, Evaluation of BRCA1 mutations in an unselected patient population with triplenegative breast cancer. Breast Cancer Res Treat 137:119–125 Couch FJ, Hart SN, Sharma P et al, 2014, Inherited mutations in 17 breast cancer susceptibility genes among a large triple negative breast cancer cohort unselected for family history of breast cancer. J Clin Oncol., DOI 10.1200/JCO.2014.57.1414 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 297 EP 308 DI 10.1007/s12253-017-0242-2 PG 12 ER PT J AU El-Gendi, S Abu-Sheasha, G AF El-Gendi, Saba Abu-Sheasha, Ghada TI Ki-67 and Cell Cycle Regulators p53, p63 and cyclinD1 as Prognostic Markers for Recurrence/ Progression of Bladder Urothelial Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ki-67; cell cycle regulators; Bladder carcinoma; CART; Survival ID Ki-67; cell cycle regulators; Bladder carcinoma; CART; Survival AB Deregulation of the cell cycle regulating genes is common in urothelial bladder carcinoma (UBC).We aimed to examine the prognostic significance of ki-67, p53, p63 and cyclinD1expression in UBC and to identify optimal cut-off points to help identifying patients at high risk of tumor recurrence. We evaluated the immunohistochemical expression of ki-67, p53, p63 and cyclinD1 in 100 UBCs. The conventional and the classification and regression trees-guided (CART-guided) methods were utilized to determine the independent predictors of tumor recurrence. The p53 and Ki-67 expression didn’t associate significantly with tumor recurrence.p63 and cyclinD1 exhibited significant hazard ratios. Using CART, no recurrence was observed when p63 was ≥87.5%. The recurrence incidence increased and the disease free survival (DFS) time shortened as the p63 decreased. CyclinD1 associated significantly with tumor recurrence only if p63 was <35%. Using the CART cut-off values¬, cases were categorized into three groups; (groups I: p63 ≥ 35%, II: p63 < 35% and cyclinD1 < 10% and III: p63 < 35% and cyclinD1 ≥ 10%). Group I patients revealed the least incidence of recurrence at the longest DFS. Group III had the worst prognosis followed by Group II. p63 represents a surrogant biomarker to predict UBC recurrence.CyclinD1 can be used only when p63 is <35%. CART proved helpful with data among which the number of cases with positive outcomes is too small relative to the number of studied predictors. Large cohort studies for ki-67 and p53 are recommended to be performed with standardized criteria as regards patients’ characteristics, cut-off values, and follow-up time. C1 [El-Gendi, Saba] University of Alexandria, Faculty of Medicine, Department of Pathology, 29 Fawzy Moaaz Street, Smouha, 21646 Alexandria, Egypt. [Abu-Sheasha, Ghada] University of Alexandria, Medical Research Institute, Department of Biomedical Informatics and Medical StatisticsAlexandria, Egypt. RP El-Gendi, S (reprint author), University of Alexandria, Faculty of Medicine, Department of Pathology, 21646 Alexandria, Egypt. EM sabaelgendi@yahoo.com CR Wang L, Feng C, Ding G, Ding Q, Zhou Z, Jiang H, Wu Z, 2014, Ki67 and TP53 expressions predict recurrence of non-muscleinvasive bladder cancer. Tumour Biol 35:2989–2995 Muir CS, 1992, Cancer incidence in five continents. Classification Lyon: IARC Sci Publ:25–30 Sgambato A1, Migaldi M, Faraglia B, De Aloysio G, Ferrari P, Ardito R, De Gaetani C, Capelli G, Cittadini A, Trentini GP. Cyclin D1 expression in papillary superficial bladder cancer:its association with other cell cycle-associatedproteins, cell proliferation and clinical outcome. Int J Cancer. 2002; 97:671–678. Fernandez-Gomez J1, Madero R, Solsona E, Unda M, Martinez- Pineiro L, Gonzalez M, Portillo J, Ojea A, Pertusa C, Rodriguez- Molina J, Camacho JE, Rabadan M, Astobieta A, Montesinos M, Isorna S, Muntanola P, Gimeno A, Blas M, Martinez-Pineiro JA, 2009, Predicting non muscle invasive bladder cancer recurrence and progression in patients treated with bacillus Calmette-Guerin: the CUETO scoring model. J Urol 182:2195–2203 Bassi P, Ferrante GD, Piazza N, Spinadin R, Carando R, Pappagallo G, Pagano F, 1999, Prognostic factors of outcome after radical cystectomy for bladder cancer: a retrospective study of a homogeneous patient cohort. J Urol 161:1494–1497 Millan-Rodriguez F, Chechile-Toniolo G, Salvador-Bayarri J, Palou J, Vicente-Rodriguez J, 2000, Multivariate analysis of the prognostic factors of primary superficial bladder cancer. J Urol 163: 73–78 Pruthi RS, Baldwin N, Bhalani V,Wallen EM, 2008, Conservative management of low risk superficial bladder tumors. J Urol 179:87– 90 Devere White RW, Stapp E, 1998, Predicting prognosis in patients with superficial bladder cancer. Oncology 12:1717–1723 Cina SJ, Epstein JI, Endrizzi JM, Harmon WJ, Seay TM, SchoenbergMP, 2001, Correlation of Cystoscopic impression with histologic diagnosis of biopsy specimens of the bladder. Hum Pathol 32:630–637 Di Como CJ, Urist MJ, Babayan I, Drobnjak M, Hedvat CV, Teruya-Feldstein J, Pohar K, Hoos A, Cordon-Cardo C, 2002, p63 expression profiles in human normal and tumor tissues. Clin Cancer Res 8:494–501 Park BJ, Lee SJ, Kim JI, Lee SJ, Lee CH, Chang SG, Park JH, Chi SG, 2000, Frequent alteration of p63 expression in human primary bladder carcinomas. Cancer Res 60:3370–3374 Koga F, Kawakami S, Fujii Y, Saito K, Ohtsuka Y, Iwai A, Ando N, Takizawa T, Kageyama Y, Kihara K, 2003, Impaired p63 expression associates with poor prognosis and uroplakin III expression in invasive urothelial carcinoma of the bladder. Clin Cancer Res 9: 5501–5507 Choi W, Shah JB, Tran M, Svatek R, Marquis L, Lee IL, Yu D, AdamL,Wen S, ShenY, Dinney C,McConkeyDJ, Siefker-Radtke A, 2012, P63 expression defines a lethal subset of muscle-invasive bladder cancers. PLoS One 7:e30206 Diehl JA, 2002, Cycling to cancer with cyclin D1. Cancer Biol Ther 1:226–231 Klein EA, Assoian RK, 2008, Transcriptional regulation of the cyclin D1 gene at a glance. J Cell Sci 121:3853–3857 Gillett C, Smith P, Gregory W, Richards M, Millis R, Peters G, Barnes D, 1996, Cyclin D1 and prognosis in human breast cancer. Int J Cancer 69:92–99 Jin M, Inoue S, Umemura T, Moriya J, Arakawa M, Nagashima K, Kato H, 2001, Cyclin D1, p16 and retinoblastoma gene product expression as a predictor for prognosis in non-small cell lung cancer at stages I and II. Lung Cancer 34:207–218 Pignataro L, Pruneri G, Carboni N, Capaccio P, Cesana BM, Neri A, Buffa R, 1998, Clinical relevance of cyclin D1 protein overexpression in laryngeal squamous cell carcinoma. J Clin Oncol 16:3069–3077 Yu Z, Weinberger PM, Haffty BG, Sasaki C, Zerillo C, Joe J, Kowalski D, Dziura J, Camp RL, Rimm DL, Psyrri A, 2005, Cyclin d1 is a valuable prognostic marker in oropharyngeal squamous cell carcinoma. Clin Cancer Res 11:1160–1166 Akervall J, KurnitDM, Adams M, Zhu S, Fisher SG, Bradford CR, Carey TE, 2004, Overexpression of cyclin D1 correlates with sensitivity to cisplatin in squamous cell carcinoma cell lines of the head and neck. Acta Otolaryngol 124:851–857 Takeuchi H, Ozawa S, Ando N, Kitagawa Y, Ueda M, Kitajima M, 2003, Cell-cycle regulators and the Ki-67 labeling index can predict the response to chemoradiotherapy and the survival of patients with locally advanced squamous cell carcinoma of the esophagus. Ann Surg Oncol 10:792–800 Lee CC, Yamamoto S, Morimura K, Wanibuchi H, Nishisaka N, Ikemoto S, Nakatani T,Wada S, Kishimoto T, Fukushima S, 1997, Significance of cyclin D1 overexpression in transitional cell carcinomas of the urinary bladder and its correlation with histopathologic features. Cancer 79:780–789 Yuan L, Gu X, Shao J,WangM, Zhu Q, Zhang Z, 2010, Cyclin D1 G870A polymorphism is associated with risk and clinicopathologic characteristics of bladder cancer. DNA Cell Biol 29:611–617 Comperat E, Camparo P, Haus R, Chartier-Kastler E, Bart S, Delcourt A, Houlgatte A, Francois R, Capron F, Vieillefond A, 2006, Immunohistochemical expression of p63, p53 and MIB1 in urinary bladder carcinoma. A tissue microarray study of 158 cases. Virchows Arch 448:319–324 Sjodahl G, Lovgren K, Lauss M, Patschan O, Gudjonsson S, Chebil G, Aine M, Eriksson P, Mansson W, Lindgren D, Ferno M, Liedberg F, Hoglund M, 2013, Toward a molecular pathologic classification of urothelial carcinoma. Am J Pathol 183:681–691 Altman DG, McShane LM, Sauerbrei W, Taube SE. Reporting Recommendations for Tumor Marker Prognostic Studies, REMARK): Explanation and Elaboration. 2012 PLoS Med 9(5): e1001216. Eble JN, Sauter G, Epstein JI, 2004, Sesterhenn IA, Eds): World Health Organization classification of Tumours: Pathology and genetics of Tumours of the urinary system and Male genital organs. IARC Press, Lyon, pp 90–109 Rosai EJ, ed,, 2004, Rosai and Ackerman's surgical Pathology, 9th Edn). Elsevier, China, pp 1317–1359 Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP, 1998, Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4:844–847 Stepan A, Simionescu C, Margaritescu C, Ciurea R, 2011, P16, cerbB2 and Ki67 immunoexpression in urothelial carcinomas of the bladder a. Romanian J Morphol Embryol 52:653–658 Koyuncuer A, 2013, Immunohistochemical expression of p63, p53 in urinary bladder carcinoma. Indian J Pathol Microbiol 56:10–15 Therneau T., Atkinson B., Ripley B. 2015. Rpart: recursive partitioning and regression trees, version R package version 4.1-10). Core Team R, 2013, R: a language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria Strobl C, Malley J, Tutz G, 2009, An introduction to recursive partitioning:rationale, application, and characteristics of classification and regressiontrees, bagging, and random forests. Psychol Methods 14:323–348 Breiman L, Friedman JH, Olshen RA, Stone CJ, 1984, Classification and RegressionTrees. Wadsworth International Group, Belmont Sylvester RJ, 2011, How well can you actually predict which nonmuscle- invasive bladder cancer patients will progress? Eur Urol 60: 431–433 discussion 433–434 Zhou X, Zhang G, Tian Y. p53 status correlates with the risk of recurrence in non-muscle invasive bladder cancers treated with Bacillus Calmette– Guerin: A Meta-Analysis. PLoS One. 2015 5; 10: e0119476. Zervas A, Tzonou A, Gakiopoulou H, DimopoulosMA, 1998, The prevalence of bcl-2, p53, and Ki-67 immunoreactivity in transitional cell bladder carcinomas and their clinicopathologic correlates. Hum Pathol 29:146–154 Bernardini S, Adessi GL, Billerey C, Chezy E, Carbillet JP, Bittard H, 1999, Immunohistochemical detection of p53 protein overexpression versus gene sequencing in urinary bladder carcinomas. J Urol 162:1496–1501 Fridman JS, Lowe SW, 2003, Control of apoptosis by p53. Oncogene 22:9030–9040 Wagner U, Suess K, Luginbuhl T, Schmid U, Ackermann D, Zellweger T, Maurer R, Alund G, Knonagel H, Rist M, Jordan P, Moch H, Mihatsch MJ, Gasser TC, Sauter G, 1999, Cyclin D1 overexpression lacks prognostic significance in superficial urinary bladder cancer. J Pathol 188:44–50 Suwa Y, Takano Y, Iki M, Takeda M, Asakura T, Noguchi S, Masuda M, 1998, Cyclin D1 protein overexpression is related to tumor differentiation, but not to tumor progression or proliferative activity, in the transitional cell carcinoma of the bladder. J Urol 160: 897–900 Santos L, Amaro T, Costa C, Pereira S, Bento MJ, Lopes P, Oliveira J, Criado B, Lopes C, 2003, Ki-67 index enhances the prognostic accuracy of the urothelial superficial bladder carcinoma risk group classification. Int J Cancer 105:267–272 Yan Y, Andriole GL, Humphrey PA, Kibel AS, 2002, Patterns of multiple recurrences of superficial, ta/T1, transitional cell carcinomas of bladder and effects of clinicopathologic andbiochemical factors. Cancer 95:1239–1246 Ding W, Gou Y, Sun C, Xia G, Wang H, Chen Z, Tan J, Xu K, Qiang D, 2014, Ki-67 is an independent indicator in non-muscle invasive bladder cancer, NMIBC); combination of EORTC risk scores and Ki-67 expression could improve the risk stratification of NMIBC. Urol Oncol. 32(42):e13–e19 Shariat SF, Kim J, Raptidis G, Ayala G, Lerner S, 2003, Association of p53 and p21 expression with clinical outcome in patients with carcinoma in situ of the urinary bladder. Urology 61: 1140–1145 Malats N, Bustos A, Nascimento CM, Fernandez F, Rivas M, Puente D, Kogevinas M, Real FX, 2005, P53 as a prognostic marker for bladder cancer: a meta-analysis and review. Lancet Oncol 6: 678–686 Urist MJ, Di Como CJ, Lu ML, Charytonowicz E, Verbel D, Crum CP, Ince TA, McKeon FD, Cordon-Cardo C, 2002, Loss of p63 expression is associated with tumor progression in bladder cancer. Am J Pathol 161:1199–1206 Moll U, 2003, The role of p63 and p73 in tumor formation and progression: coming of age towards clinical usefulness. Clin Cancer Res 9:5437–5441 Lopez-Beltran A, Luque RJ, Alvarez-Kindelan J, Quintero A, Merlo F, Carrasco JC, Requena MJ, Montironi R, 2004, Prognostic factors in stage T1 grade 3 bladder cancer survival: the role of G1-S modulators, p53, p21Waf1, p27kip1, cyclin D1, and cyclin D3, and proliferation index, ki67- MIB1). Eur Urol 45:606– 612 Shariat SF, Ashfaq R, Sagalowsky AI, Lotan Y, 2006, Correlation of cyclin D1 and E1 expression with bladder cancer presence, invasion, progression, and metastasis. Hum Pathol 37:1568–1576 Yurakh AO, Ramos D, Calabuig-Farinas S, Lopez-Guerrero JA, Rubio J, Solsona E, Romanenko AM, Vozianov AF, Pellin A, Llombart-Bosch A, 2006, Molecular and immunohistochemical analysis of the prognostic value of cell-cycle regulators in urothelial neoplasms of the bladder. Eur Urol 50:506–515 Bringuier PP, Tamimi Y, Schuuring E, Schalken J, 1996, Expression of cyclin D1 and EMS1 in bladder tumours; relationship with chromosome 11q13 amplification. Oncogene 12:1747– 1753 Fristrup N, Birkenkamp-Demtroder K, Reinert T, Sanchez-Carbayo M, Segersten U, Malmstrom PU, Palou J, Alvarez-Mugica M, Pan CC, Ulhoi BP, Borre M, Orntoft TF, Dyrskjot L, 2013, Multicenter validation of cyclin D1,MCM7, TRIM29, and UBE2C as prognostic protein markers in non muscle-invasive bladder cancer. Am J Pathol 182:339–349 Levidou G, Saetta AA, Karlou M, Thymara I, Pratsinis H, Pavlopoulos P, Isaiadis D, Diamantopoulou K, Patsouris E, Korkolopoulou P, 2010, D-type cyclins in superficial and muscle-invasive bladder urothelial carcinoma: correlation with clinicopathological data and prognostic significance. J Cancer Res Clin Oncol 136:1563–1571 Tut VM, Braithwaite KL, Angus B, Neal DE, Lunec J, Mellon JK, 2001, Cyclin D1 expression in transitional cell carcinoma of the bladder: correlation with p53, waf1, pRb and Ki67. Br J Cancer 84: 270–275 Kindelan J, Quintero A, Merlo F, RequenaMJ, Montironi R, 2004, Prognostic factors in survival of patients with stage ta and T1 bladder urothelial tumors: the role of G1-S modulators, p53, p21Waf1, p27Kip1, cyclin D1, and cyclin D3), proliferation index, and clinicopathologic parameters. Am J Clin Pathol 122:444–452 Olsson H, Hultman P, Monsef N, Rosell J, Jahnson S, 2012, Immunohistochemical evaluation of cell cycle regulators: impact on predicting prognosis in stage t1 urinary bladder carcinoma. ISRN Urol 2012:379081 Sun Z, Tao Y, Li S, Ferguson KK,Meeker JD, Park SK, Batterman SA, Mukherjee B, 2013, Statistical strategies for constructing health risk models with multiple pollutants and their interactions: possible choices and comparisons. Environ Health 12:85 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 309 EP 322 DI 10.1007/s12253-017-0250-2 PG 14 ER PT J AU Lee, H Cho, JH Park, WJ Jung, SJ Choi, IJ Lee, JH AF Lee, Hyunsu Cho, Ji-Hyoung Park, Won-Jin Jung, Soo-Jung Choi, In-Jang Lee, Jae-Ho TI Loss of the Association between Telomere Length and Mitochondrial DNA Copy Number Contribute to Colorectal Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Mitochondria DNA copy number; Serrated polyps; Telomere; Tubular adenomas ID Colorectal cancer; Mitochondria DNA copy number; Serrated polyps; Telomere; Tubular adenomas AB Positive association between telomere length and mitochondrial DNA (mtDNA) copy number were introduced in healthy and patients with psychiatric disorder. Based on frequent genetic changes of telomere and mitochondria in colorectal carcinomas (CRC), we studied their clinical characteristics and their association in colorectal carcinogenesis. DNA was extracted from 109 CRCs, 64 colorectal tubular adenomas (TAs), and 28 serrated polyps (SPs), and then, telomere length and mtDNA copy number were analyzed in these legions by using a real-time PCR assay. Telomere length and mtDNA copy number (mean ± S.D) in CRCs was 1.87 ± 1.52 and 1.61 ± 1.37, respectively. In TAs and SPs, relative mtDNA copy number was 0.92 ± 0.71 and 1.84 ± 1.06, respectively, shoing statistical difference (p = 0.017). However, telomere length was similar in these precancerous legions. Telomere length and mtDNA copy number did not show clinical and prognostic values in CRCs, however, positive correlation between telomere length and mitochondrial DNA copy number were found in CRC (r = 0.408, p < 0.001). However, this association was not shown in precancerous lesions (r = −0.031, p = 0.765). This result suggests that loss of coregulation between telomeres and mitochondrial function may induce the initiation or play a role as trigger factor of colorectal carcinogenesis. C1 [Lee, Hyunsu] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeoldaero, Dalseo-Gu, 2800 Daegu, South Korea. [Cho, Ji-Hyoung] Keimyung University, School of Medicine, Department of General SurgeryDaegu, South Korea. [Park, Won-Jin] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeoldaero, Dalseo-Gu, 2800 Daegu, South Korea. [Jung, Soo-Jung] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeoldaero, Dalseo-Gu, 2800 Daegu, South Korea. [Choi, In-Jang] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeoldaero, Dalseo-Gu, 2800 Daegu, South Korea. [Lee, Jae-Ho] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeoldaero, Dalseo-Gu, 2800 Daegu, South Korea. RP Lee, JH (reprint author), Keimyung University, School of Medicine, Department of Anatomy, 2800 Daegu, South Korea. EM anato82@dsmc.or.kr CR Boland CR, Thibodeau SN, Hamilton SR et al, 1998, A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58:5248–5257 Popat S, Hubner R, Houlston RS, 2005, Systematic review of microsatellite instability and colorectal cancer prognosis. J Clin Oncol 23:609–618 Soreide K, Janssen EA, Soiland H, Korner H, Baak JP, 2006, Microsatellite instability in colorectal cancer. Br J Surg 93:395–406 Jass JR, 1999, Serrated adenoma and colorectal cancer. J Pathol 187:499–502 Jass JR, Whitehall VL, Young J, Leggett BA, 2002, Emerging concepts in colorectal neoplasia. Gastroenterology 123:862–876 Harley CB, Futcher AB, Greider CW, 1990, Telomeres shorten during ageing of human fibroblasts. Nature 345:458–460 Lee HC, Li SH, Lin JC,Wu CC, Yeh DC,Wei YH, 2004, Somatic mutations in the D-loop and decrease in the copy number of mitochondrialDNAin human hepatocellular carcinoma. Mutat Res 547: 71–78 Blasco MA, 2005, Telomeres and human disease: ageing, cancer and beyond. Nat Rev Genet 6:611–622 Blackburn EH, Greider CW, Szostak JW, 2006, Telomeres and telomerase: the path from maize, Tetrahymena and yeast to human cancer and aging. Nat Med 12:1133–1138 Liao LM, Baccarelli A, Shu XO et al, 2011, Mitochondrial DNA copy number and risk of gastric cancer : a report from the shanghai Women's health study. Cancer Epidemiol Biomark Prev 20:1944– 1949 Cui H, Huang P, Wang Z et al, 2013, Association of decreased mitochondrial DNA content with the progression of colorectal cancer. BMC Cancer 13:110 Luu HN, Long J, Wen Wet al, 2016, Association between genetic risk score for telomere length and risk of breast cancer. Cancer Causes Control 27:1219–1228 Kim JH, Kim HK, Ko JH, Bang H, Lee DC, 2013, The relationship between leukocyte mitochondrial DNA copy number and telomere length in community-dwelling elderly women. PLoS One 8:e67227 Qiu C, Enquobahrie DA, Gelaye B, Hevner K, Williams MA, 2015, The association between leukocyte telomere length and mitochondrial DNA copy number in pregnant women: a pilot study. Clin Lab 61:363–369 Tyrka AR, Carpenter LL, Kao HT et al, 2015, Association of telomere length and mitochondrial DNA copy number in a community sample of healthy adults. Exp Gerontol 66:17–20 Lee JH, Hwang I, Kang YN, Choi IJ, Kim DK, 2015, Genetic characteristics of mitochondrial DNAwas associated with colorectal carcinogenesis and its prognosis. PLoS One 10:e0118612 Chen T, He J, Shen L et al, 2011, Themitochondrial DNA4,977-bp deletion and its implication in copy number alteration in colorectal cancer. BMC Med Genet 12:8 Baldus SE, Schaefer KL, Engers R, Hartleb D, Stoecklein NH, Gabbert HE, 2010, Prevalence and heterogeneity of KRAS, BRAF, and PIK3CA mutations in primary colorectal adenocarcinomas and their corresponding metastases. Clin Cancer Res 16: 790–799 Tyrka AR, Parade SH, Price LH et al, 2016, Alterations of mitochondrial DNA copy number and telomere length with early adversity and psychopathology. Biol Psychiatry 79:78–86 Bao D, Ba Y, Zhou F et al, 2016, Alterations of telomere length and mtDNA copy number are associated with overall survival in hepatocellular carcinoma patients treated with transarterial chemoembolization. Cancer Chemother Pharmacol 78:791–799 Wong KK, Maser RS, Bachoo RM et al, 2003, Telomere dysfunction and Atm deficiency compromises organ homeostasis and accelerates ageing. Nature 421:643–648 Sahin E, Colla S, LiesaMet al, 2011, Telomere dysfunction induces metabolic and mitochondrial compromise. Nature 470:359–365 Hu J, Hwang SS, Liesa M et al, 2012, Antitelomerase therapy provokes ALT and mitochondrial adaptive mechanisms in cancer. Cell 148:651–663 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 323 EP 328 DI 10.1007/s12253-017-0245-z PG 6 ER PT J AU Tahmasebi Birgani, M Hajjari, M Shahrisa, A Khoshnevisan, A Shoja, Z Motahari, P Farhangi, B AF Tahmasebi Birgani, Maryam Hajjari, Mohammadreza Shahrisa, Arman Khoshnevisan, Atefeh Shoja, Zahra Motahari, Paria Farhangi, Baharak TI Long Non-Coding RNA SNHG6 as a Potential Biomarker for Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Biomarker; Long noncoding RNA; SNHG6; Systematic analysis ID Hepatocellular carcinoma; Biomarker; Long noncoding RNA; SNHG6; Systematic analysis AB Long Non-coding RNAs (lncRNAs) refer to all non-protein coding transcripts longer than 200 nucleotides. Their critical roles in different biological pathways have been already well established. Altered expression of lncRNAs can be involved in the cancer initiation and/or progression. Since patients with hepatocellular carcinoma (HCC) are usually diagnosed in late stages, developing diagnostic methods seems to be essential. In this study, the expression levels of different lncRNAs were systematically analysed in different genomic and transcriptome datasets. The analyses showed that SNHG6 is among the lncRNAs with distinctive dysregulation of expression and copy number variation in HCC tumors compared with normal tissues. The results also suggest that the dysregulation of SNHG6 is highly cancer type specific. Through co-occurrence analyses, we found that SNHG6 and its related co-expressed genes on 8q are involved in the structural integrity of ribosome and translation. This comprehensive in silico analysis, provides a resource for investigating SNHG6 in hepatocellular carcinoma and lays the groundwork for design of next researches. C1 [Tahmasebi Birgani, Maryam] Ahvaz Jundishapur University of Medical Sciences, School of Medicine, Department of Medical GeneticsAhvaz, Iran. [Hajjari, Mohammadreza] Shahid Chamran University of Ahvaz, Faculty of Sciences, Department of GeneticsAhvaz, Iran. [Shahrisa, Arman] Tarbiat Modares University, Faculty of Biosciences, Department of Molecular GeneticsTehran, Iran. [Khoshnevisan, Atefeh] Shahid Chamran University of Ahvaz, Faculty of Sciences, Department of GeneticsAhvaz, Iran. [Shoja, Zahra] Ahvaz Jundishapur University of Medical Sciences, School of Medicine, Department of Medical GeneticsAhvaz, Iran. [Motahari, Paria] Iranian Research Organization Science & Technology, Department of BiotechnologyTehran, Iran. [Farhangi, Baharak] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. RP Tahmasebi Birgani, M (reprint author), Ahvaz Jundishapur University of Medical Sciences, School of Medicine, Department of Medical Genetics, Ahvaz, Iran. EM tahmasebi-ma@ajums.ac.ir CR Wang C-H, Wey K-C, Mo L-R, Chang K-K, Lin R-C, Kuo J-J, 2014, Current trends and recent advances in diagnosis, therapy, and prevention of hepatocellular carcinoma. Asian Pac J Cancer Prev 16:3595–3604 Mazzocca A, Tahmasebi Birgani M, Sabba C, Carloni V, 2014, Tetraspanin-enriched Microdomains and hepatocellular carcinoma progression. Cancer Lett 351(1):23–29 Su C-H, Lin Y, Cai L, 2013, Genetic factors, viral infection, other factors and liver cancer: an update on current progress. Asian Pac J Cancer Prev 14:4953–4960 de Oliveria Andrade LJ, D'Oliveira A, Melo RC, De Souza EC, Costa Silva CA, Parana R, 2009, Association between hepatitis C and hepatocellular carcinoma. J Glob Infect Dis 1:33–37 El-Serag HB, 2012, Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology 142(1264–1273):e1261 Zhu K, Dai Z, Zhou J, 2013, Biomarkers for hepatocellular carcinoma: progression in early diagnosis, prognosis, and personalized therapy. Biomarker research 1:10 Hajjari M, Khoshnevisan A, Shin YK, 2014, Molecular function and regulation of long non-coding RNAs: paradigms with potential roles in cancer. Tumor Biol 35:10645–10663 Yarmishyn AA, Kurochkin IV, 2015, Long noncoding RNAs: a potential novel class of cancer biomarkers. Front Genet 6:1–10 Ayers D, 2013, Long non-coding RNAs: novel emergent biomarkers for cancer diagnostics. Nature 1:31–35 Hajjari M, Behmanesh M, Sadeghizadeh M, Zeinoddini M, 2013, Up-regulation of HOTAIR long non-coding RNA in human gastric adenocarcinoma tissues. Med Oncol 30:1–4 Hajjari M, Khoshnevisan A, 2013, Potential long non-coding RNAs to be considered as biomarkers or therapeutic targets in gastric cancer. Front Genet 4:1–3 Hajjari M, Khoshnevisan A, Shin YK, 2013, Long non-coding RNAs in hematologic malignancies: road to translational research. Front Genet 4:1–2 Shibata C, Otsuka M, Kishikawa T, Ohno M, Yoshikawa T, Takata A, Koike K, 2015, Diagnostic and therapeutic application of noncoding RNAs for hepatocellular carcinoma.World J Hepatol 7:1–6 Fang T-T, Sun X-J, Chen J, Zhao Y, Sun R-X, Ren N, Liu B-B, 2014, Long non-coding RNAs are differentially expressed in hepatocellular carcinoma cell lines with differing metastatic potential. Asian Pac J Cancer Prev 15:10513–10524 Goossens N, Nakagawa S, Sun X and Hoshida Y(2015, Cancer biomarker discovery and validation. Transl Cancer Res 4(3):256– 269 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, Antipin Y, Reva B, Goldberg AP, Sander C, Schultz N, 2012, The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2:401–404 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, 2013, Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6:l1 Petryszak R, Keays M, Tang YA, Fonseca NA, Barrera E, Burdett T, Fullgrabe A, Fuentes AM-P, Jupp S, Koskinen S, 2015, Expression Atlas update—an integrated database of gene and protein expression in humans, animals and plants. Nucleic Acids Res 44(D1):D746–D752 Petryszak R, Burdett T, Fiorelli B, Fonseca NA, Gonzalez-Porta M, Hastings E, Huber W, Jupp S, Keays M, Kryvych N, McMurry J, Marioni JC, Malone J, Megy K, Rustici G, Tang AY, Taubert J, Williams E, Mannion O, Parkinson HE, Brazma A, 2014, Expression atlas update–a database of gene and transcript expression from microarray- and sequencing-based functional genomics experiments. Nucleic Acids Res 42:D926–D932 Kapushesky M, Adamusiak T, Burdett T, Culhane A, Farne A, Filippov A, Holloway E, Klebanov A, Kryvych N, Kurbatova N, Kurnosov P, Malone J, Melnichuk O, Petryszak R, Pultsin N, Rustici G, Tikhonov A, Travillian RS, Williams E, Zorin A, Parkinson H, Brazma A, 2012, Gene expression atlas update–a value-added database of microarray and sequencing-based functional genomics experiments. Nucleic Acids Res 40:D1077–D1081 Cline MS, Craft B, Swatloski T, Goldman M, Ma S, Haussler D, Zhu J, 2013, Exploring TCGA pan-cancer data at the UCSC cancer genomics browser. Sci Rep 3:2652 Lopez B, Cline M, Broom B, Margolin A, Omberg L,Weinstein J, Axton M, 2013, Thread 4: data discovery, transparency and visualization. Nat Genet 45., DOI 10.1038/ng.2789 Goldman M, Craft B, Swatloski T, Ellrott K, Cline M, Diekhans M, Ma S, Wilks C, Stuart J, Haussler D, Zhu J, 2013, The UCSC cancer genomics browser: update 2013. Nucleic Acids Res 41: D949–D954 Sanborn JZ, Benz SC, Craft B, Szeto C, Kober KM, Meyer L, Vaske CJ, Goldman M, Smith KE, Kuhn RM, Karolchik D, Kent WJ, Stuart JM, Haussler D, Zhu J, 2011, The UCSC cancer genomics browser: update 2011. Nucleic Acids Res 39:D951–D959 Vaske CJ, Benz SC, Sanborn JZ, Earl D, Szeto C, Zhu J, Haussler D, Stuart JM, 2010, Inference of patient-specific pathway activities from multi-dimensional cancer genomics data using PARADIGM. Bioinformatics 26:i237–i245 Zhu J, Sanborn JZ, Benz S, Szeto C, Hsu F, Kuhn RM, Karolchik D, Archie J, Lenburg ME, Esserman LJ, Kent WJ, Haussler D, Wang T, 2009, The UCSC cancer genomics browser. Nat Methods 6:239–240 Wurmbach E, Yb C, Khitrov G, ZhangW, Roayaie S, Schwartz M, Fiel I, Thung S, Mazzaferro V, Bruix J, 2007, Genome-wide molecular profiles of HCV-induced dysplasia and hepatocellular carcinoma. Hepatology 45:938–947 Cao C, Zhang T, Zhang D,Xie L, Zou X, Lei L,Wu D, Liu L, 2016, The long non-coding RNA, SNHG6–003, functions as a competing endogenous RNA to promote the progression of hepatocellular carcinoma. Oncogene 36:1112–1122 Kim JH, You KR, Kim IH, Cho BH, Kim CY, Kim DG, 2004, Over-expression of the ribosomal protein L36a gene is associated with cellular proliferation in hepatocellular carcinoma. Hepatology 39:129–138 Wong QW-L, Li J, Ng SR, Lim SG, Yang H, Vardy LA, 2014, RPL39L is an example of a recently evolved ribosomal protein paralog that shows highly specific tissue expression patterns and is upregulated in ESCs and HCC tumors. RNA Biol 11:33–41 Weber RG, Pietsch T, von Schweinitz D, Lichter P, 2000, Characterization of genomic alterations in hepatoblastomas: a role for gains on chromosomes 8q and 20 as predictors of poor outcome. Am J Pathol 157:571–578 Xiang J-F,YinQ-F, ChenT, ZhangY, ZhangX-O, Wu Z, Zhang S, Wang H-B, Ge J, Lu X, 2014, Human colorectal cancer-specific CCAT1-L lncRNA regulates long-range chromatin interactions at the MYC locus. Cell Res 24:513–531 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 329 EP 337 DI 10.1007/s12253-017-0241-3 PG 9 ER PT J AU Long, H Guo, X Qiao, Sh Huang, Q AF Long, Houyong Guo, Xingjun Qiao, Shen Huang, Qingxing TI Tumor LXR Expression is a Prognostic Marker for Patients with Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Liver X receptor; Hepatocellular carcinoma; Prognosis; Invasion ID Liver X receptor; Hepatocellular carcinoma; Prognosis; Invasion AB Liver X receptor (LXR) activation exerts an antitumor effect. However, whether the tumor LXR expression has prognostic significance in hepatocellular carcinoma (HCC) patient has not been addressed yet. Primary HCC and the adjacent non-tumor tissues were obtained from 169 patients who underwent routine curative surgical treatment. All patients were followed for prognosis analyses. Tumor LXR was detected by immunohistochemical analysis. In in vitro study, several HCC cell lines were cultured for cellular protein detection of LXR and other cytokines, including nuclear factor kappa (NFκB), Matrix metalloproteinases 2 and 9 (MMP- 2 and -9). Meanwhile, the invasion ability of cultured HCC cell lines was performed. We found that LXR expression status in tumor samples is associated with the clinical characteristics, such as tumor stage and metastasis, of HCC patients. Prognosis analysis shows that tumor LXR expression status is closely related to the post-operative outcome in HCC patients who underwent surgical treatment. Patients with low LXR expression have a significantly lower mean 5-year overall survival rate and mean overall survival period than those with high LXR level. Our in vitro data reveal that HCC cell lines had increased NF-κB, MMP2, MMP9 and invasive ability than normal cell line, which are suppressed by LXR activation via NFκB pathway. Our data suggest that LXR could be used as a biomarker for HCC prognosis. Further study is warranted to explore the molecular mechanism under which LXR regulates tumor behaves. C1 [Long, Houyong] Jining NO.1 People’s Hospital, Department of Hepatobiliary Surgery, 6 Health Street, 272011 Jining, Shandong Province, China. [Guo, Xingjun] Dongying People’s Hospital, Department of Hepatobiliary Surgery, 257091 Dongying, Shandong Province, China. [Qiao, Shen] Jining NO.1 People’s Hospital, Department of Hepatobiliary Surgery, 6 Health Street, 272011 Jining, Shandong Province, China. [Huang, Qingxing] Shanxi Medical University Affiliated Tumor Hospital, Shanxi Tumor Hospital, Department of Digestive Endoscopic and Minimally invasive Surgery, 3 Zhigongxiejie Road, 030013 Taiyuan, Shanxi Province, China. RP Huang, Q (reprint author), Shanxi Medical University Affiliated Tumor Hospital, Shanxi Tumor Hospital, Department of Digestive Endoscopic and Minimally invasive Surgery, 030013 Taiyuan, China. EM dr_huang1@163.com;CAOHUANGX@163.COM CR Kim JU, Shariff MI, Crossey MM, Gomez-Romero M, Holmes E et al, 2016, Hepatocellular carcinoma: review of disease and tumor biomarkers. World J Hepatol 8:471–484 Chen KW, Ou TM, Hsu CW, Horng CT, Lee CC et al, 2015, Current systemic treatment of hepatocellular carcinoma: a review of the literature. World J Hepatol 7:1412–1420 Karaman B, Battal B, Sari S, Verim S, 2014, Hepatocellular carcinoma review: current treatment, and evidence-based medicine. World J Gastroenterol 20:18059–18060 BaranowskiM(2008, Biological role of liver X receptors. J Physiol Pharmacol 59(Suppl 7):31–55 El Roz A, Bard JM, Huvelin JM, Nazih H, 2012, LXR agonists and ABCG1-dependent cholesterol efflux inMCF-7 breast cancer cells: relation to proliferation and apoptosis. Anticancer Res 32:3007– 3013 Lo Sasso G, Bovenga F, Murzilli S, Salvatore L, Di Tullio G, et al., 2013, Liver X receptors inhibit proliferation of human colorectal cancer cells and growth of intestinal tumors in mice. Gastroenterology 144: 1497-1507, 1507 e1491-1413. Mehrotra A, Kaul D, Joshi K, 2011, LXR-alpha selectively reprogrammes cancer cells to enter into apoptosis. Mol Cell Biochem 349:41–55 Chuu CP, Lin HP, 2010, Antiproliferative effect of LXR agonists T0901317 and 22(R)-hydroxycholesterol on multiple human cancer cell lines. Anticancer Res 30:3643–3648 Na TY, Shin YK, Roh KJ, Kang SA, Hong I et al, 2009, Liver X receptormediates hepatitis B virusX protein-induced lipogenesis in hepatitis B virus-associated hepatocellular carcinoma. Hepatology 49:1122–1131 Russo V, 2011, Metabolism, LXR/LXR ligands, and tumor immune escape. J Leukoc Biol 90:673–679 McFadden JW, Corl BA, 2010, Activation of liver X receptor, LXR, enhances de novo fatty acid synthesis in bovine mammary epithelial cells. J Dairy Sci 93:4651–4658 Zhao G, Zhang H, Huang Z, Lv L, Yan F, 2016, Cortactin and Exo70 mediated invasion of hepatoma carcinoma cells by MMP- 9 secretion. Mol Biol Rep 43:407–414 Lempinen M, Lyytinen I, Nordin A, Tervahartiala T, Makisalo H et al, 2013, Prognostic value of serum MMP- 8, −9 and TIMP-1 in patients with hepatocellular carcinoma. Ann Med 45:482–487 Sakamoto Y, Mafune K, Mori M, Shiraishi T, Imamura H et al, 2000, Overexpression of MMP-9 correlates with growth of small hepatocellular carcinoma. Int J Oncol 17:237–243 Giannelli G, Bergamini C, Marinosci F, Fransvea E, Quaranta M et al, 2002, Clinical role of MMP-2/TIMP-2 imbalance in hepatocellular carcinoma. Int J Cancer 97:425–431 Joseph SB, Bradley MN, Castrillo A, Bruhn KW, Mak PA et al, 2004, LXR-dependent gene expression is important for macrophage survival and the innate immune response. Cell 119:299–309 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 339 EP 344 DI 10.1007/s12253-017-0249-8 PG 6 ER PT J AU Karai, B Hevessy, Zs Szantho, E Csathy, L Ujfalusi, A Gyurina, K Szegedi, I Kappelmayer, J Kiss, Cs AF Karai, Bettina Hevessy, Zsuzsanna Szantho, Eszter Csathy, Laszlo Ujfalusi, Aniko Gyurina, Katalin Szegedi, Istvan Kappelmayer, Janos Kiss, Csongor TI Expression of Coagulation Factor XIII Subunit A Correlates with Outcome in Childhood Acute Lymphoblastic Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Precursor B-cell acute lymphoblastic leukemia; Immunophenotype; Factor XIII-A; 'B-other' ALL ID Precursor B-cell acute lymphoblastic leukemia; Immunophenotype; Factor XIII-A; 'B-other' ALL AB Previously we identified B-cell lineage leukemic lymphoblasts as a new expression site for subunit A of blood coagulation factor XIII (FXIII-A). On the basis of FXIII-A expression, various subgroups of B-cell precursor acute lymphoblastic leukemia (BCP-ALL) can be identified. Fifty-five children with BCP-ALL were included in the study. Bone marrow samples were obtained by aspiration and the presence of FXIII-A was detected by flow cytometry. G-banding and fluorescent in situ hybridization was performed according to standard procedures. The 10-year event-free survival (EFS) and overall survival (OS) rate of FXIII-A-positive and FXIII-A-negative patients showed significant differences (EFS: 84% vs. 61%, respectively; p = 0.031; OS: 89% vs. 61%; p = 0.008). Of all the parameters examined, there was correlation only between FXIII-A expression and ‘B-other’ genetic subgroup. Further multivariate Cox regression analysis of FXIII-subtype and genetic group or ‘B-other’ subgroup identified the FXIII-A negative characteristic as an independent factor associated with poor outcome in BCP-ALL. We found an excellent correlation between long-term survival and the FXIII-A-positive phenotype of BCP lymphoblasts at presentation. The results presented seem to be convincing enough to suggest a possible role for FXIII-A expression in the prognostic grouping of childhood BCP-ALL patients. C1 [Karai, Bettina] University of Debrecen, Faculty of Medicine, Department of Laboratory Medicine, Nagyerdei krt 98, 4032 Debrecen, Hungary. [Hevessy, Zsuzsanna] University of Debrecen, Faculty of Medicine, Department of Laboratory Medicine, Nagyerdei krt 98, 4032 Debrecen, Hungary. [Szantho, Eszter] University of Debrecen, Faculty of Medicine, Department of Laboratory Medicine, Nagyerdei krt 98, 4032 Debrecen, Hungary. [Csathy, Laszlo] University of Debrecen, Faculty of Medicine, Department of Laboratory Medicine, Nagyerdei krt 98, 4032 Debrecen, Hungary. [Ujfalusi, Aniko] University of Debrecen, Faculty of Medicine, Department of Laboratory Medicine, Nagyerdei krt 98, 4032 Debrecen, Hungary. [Gyurina, Katalin] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-Oncology, Nagyerdei krt 98, 4032 Debrecen, Hungary. [Szegedi, Istvan] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-Oncology, Nagyerdei krt 98, 4032 Debrecen, Hungary. [Kappelmayer, Janos] University of Debrecen, Faculty of Medicine, Department of Laboratory Medicine, Nagyerdei krt 98, 4032 Debrecen, Hungary. [Kiss, Csongor] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-Oncology, Nagyerdei krt 98, 4032 Debrecen, Hungary. RP Kiss, Cs (reprint author), University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-Oncology, 4032 Debrecen, Hungary. EM kisscs@med.unideb.hu CR Conter V, Arico M, Basso G, Biondi A, Barisone E, Messina C, Parasole R, De Rossi G, Locatelli F, Pession A, Santoro N, Micalizzi C, Citterio M, Rizzari C, Silvestri D, Rondelli R, Lo Nigro L, Ziino O, Testi AM, Masera G, Valsecchi MG, Associazione Italiana di Ematologia ed Oncologia P, 2010, Longterm results of the Italian Association of Pediatric Hematology and Oncology, AIEOP, studies 82, 87, 88, 91 and 95 for childhood acute lymphoblastic leukemia. Leukemia 24(2):255–264 Moricke A, Zimmermann M, Reiter A, Henze G, Schrauder A, Gadner H, Ludwig WD, Ritter J, Harbott J, Mann G, Klingebiel T, Zintl F, Niemeyer C, Kremens B, Niggli F, Niethammer D,Welte K, Stanulla M, Odenwald E, Riehm H, Schrappe M, 2010, Longterm results of five consecutive trials in childhood acute lymphoblastic leukemia performed by the ALL-BFM study group from 1981 to 2000. Leukemia 24(2):265–284 Stary J, ZimmermannM, CampbellM, Castillo L, Dibar E, Donska S, Gonzalez A, Izraeli S, Janic D, Jazbec J, Konja J, Kaiserova E, Kowalczyk J, Kovacs G, Li CK, Magyarosy E, Popa A, Stark B, Jabali Y, Trka J, Hrusak O, RiehmH, Masera G, SchrappeM(2014, Intensive chemotherapy for childhood acute lymphoblastic leukemia: results of the randomized intercontinental trial ALL IC-BFM 2002. J Clin Oncol 32(3):174–184 Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, Harris NL, Le Beau MM, Hellstrom-Lindberg E, Tefferi A, Bloomfield CD, 2009, The 2008 revision of the World Health Organization, WHO, classification ofmyeloid neoplasms and acute leukemia: rationale and important changes. Blood 114(5):937–951 Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW, 2016, The 2016 revision to theWorld Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127(20):2391–2405 Moorman AV, 2016, New and emerging prognostic and predictive genetic biomarkers in B-cell precursor acute lymphoblastic leukemia. Haematologica 101(4):407–416 Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan-Smith E, Dalton JD, Girtman K, Mathew S,Ma J, Pounds SB, Su X, Pui CH, Relling MV, Evans WE, Shurtleff SA, Downing JR, 2007, Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature 446(7137):758–764 Cave H, van der Werff ten Bosch J, Suciu S, Guidal C, Waterkeyn C, Otten J, Bakkus M, Thielemans K, Grandchamp B, Vilmer E, 1998, Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia. European Organization for Research and Treatment of Cancer–childhood leukemia cooperative group. N Engl J Med 339(9):591–598 Coustan-Smith E, Sancho J, Hancock ML, Boyett JM, Behm FG, Raimondi SC, Sandlund JT, Rivera GK, Rubnitz JE, Ribeiro RC, Pui CH, Campana D, 2000, Clinical importance of minimal residual disease in childhood acute lymphoblastic leukemia. Blood 96(8):2691–2696 Dworzak MN, Froschl G, Printz D, Mann G, Potschger U, Muhlegger N, Fritsch G, Gadner H, Austrian Berlin-Frankfurt- Munster Study G, 2002, Prognostic significance and modalities of flow cytometric minimal residual disease detection in childhood acute lymphoblastic leukemia. Blood 99(6):1952–1958 Basso G, Veltroni M, Valsecchi MG, Dworzak MN, Ratei R, Silvestri D, Benetello A, Buldini B, Maglia O, Masera G, Conter V, Arico M, Biondi A, Gaipa G, 2009, Risk of relapse of childhood acute lymphoblastic leukemia is predicted by flow cytometric measurement of residual disease on day 15 bone marrow. J Clin Oncol 27(31):5168–5174 Borowitz MJ, Devidas M, Hunger SP, Bowman WP, Carroll AJ, Carroll WL, Linda S, Martin PL, Pullen DJ, Viswanatha D, Willman CL, Winick N, Camitta BM, Children's Oncology G, 2008, Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: a Children's Oncology group study. Blood 111(12):5477–5485 Mejstrikova E, Fronkova E, Kalina T, Omelka M, Batinic D, Dubravcic K, Pospisilova K, Vaskova M, Luria D, Cheng SH, Ng M, LeungY, Kappelmayer J,Kiss F, Izraeli S, Stark B, Schrappe M, Trka J, Stary J, Hrusak O, 2010, Detection of residual B precursor lymphoblastic leukemia by uniform gating flow cytometry. Pediatr Blood Cancer 54(1):62–70 Dworzak MN, Gaipa G, Ratei R, Veltroni M, Schumich A, Maglia O, Karawajew L, Benetello A, Potschger U, Husak Z, Gadner H, Biondi A, Ludwig WD, Basso G, 2008, Standardization of flow cytometric minimal residual disease evaluation in acute lymphoblastic leukemia: Multicentric assessment is feasible. Cytometry B Clin Cytom 74(6):331–340 van Dongen JJ, van der Velden VH, Bruggemann M, Orfao A, 2015)Minimal residual disease diagnostics in acute lymphoblastic leukemia: need for sensitive, fast, and standardized technologies. Blood 125(26):3996–4009 Boldt DH, Kopecky KJ, Head D, Gehly G, Radich JP, Appelbaum FR, 1994, Expression of myeloid antigens by blast cells in acute lymphoblastic leukemia of adults. The southwest Oncology group experience. Leukemia 8(12):2118–2126 Invernizzi R, De Fazio P, Iannone AM, Zambelli LM, Rastaldi MP, Ippoliti G, Ascari E, 1992, Immunocytochemical detection of factor XIII A–subunit in acute leukemia. Leuk Res 16(8):829–836 Kappelmayer J, Simon A, Katona E, Szanto A, Nagy L, Kiss A, Kiss C, Muszbek L, 2005, Coagulation factor XIII-A. A flow cytometric intracellular marker in the classification of acute myeloid leukemias. Thromb Haemost 94(2):454–459 Kiss F, Simon A, Csathy L, Hevessy Z, Katona E, Kiss C, Kappelmayer J, 2008, A coagulation factor becomes useful in the study of acute leukemias: studies with blood coagulation factor XIII. Cytometry A 73(3):194–201 Simon A, Bagoly Z, Hevessy Z, Csathy L, Katona E, Vereb G, Ujfalusi A, Szerafin L, Muszbek L, Kappelmayer J, 2012, Expression of coagulation factor XIII subunit a in acute promyelocytic leukemia. Cytometry B Clin Cytom 82(4):209–216 Kiss F, Hevessy Z, Veszpremi A, Katona E, Kiss C, Vereb G, Muszbek L, Kappelmayer JN, 2006, Leukemic lymphoblasts, a novel expression site of coagulation factor XIII subunit a. Thromb Haemost 96(2):176–182 Katona EE, Ajzner E, Toth K, Karpati L, Muszbek L, 2001, Enzyme-linked immunosorbent assay for the determination of blood coagulation factor XIII A-subunit in plasma and in cell lysates. J Immunol Methods 258(1–2):127–135 Simons A, Shaffer LG, Hastings RJ, 2013, Cytogenetic nomenclature: changes in the ISCN 2013 compared to the 2009 edition. Cytogenet Genome Res 141(1):1–6 Muszbek L, Yee VC, Hevessy Z, 1999, Blood coagulation factor XIII: structure and function. Thromb Res 94(5):271–305 Muszbek L, Adany R, Mikkola H, 1996, Novel aspects of blood coagulation factor XIII. I. Structure, distribution, activation, and function. Crit Rev Clin Lab Sci 33(5):357–421 Inbal A, Lubetsky A, Krapp T, Castel D, Shaish A, Dickneitte G, Modis L, Muszbek L, Inbal A, 2005, Impaired wound healing in factor XIII deficient mice. Thromb Haemost 94(2):432–437 Richardson VR, Cordell P, Standeven KF, Carter AM, 2013, Substrates of factor XIII-A: roles in thrombosis and wound healing. Clin Sci, Lond, 124(3):123–137 Bagoly Z, Katona E, Muszbek L, 2012, Factor XIII and inflammatory cells. Thromb Res 129:S77–S81 Serrano K, Devine DV, 2002, Intracellular factor XIII crosslinks platelet cytoskeletal elements upon platelet activation. Thromb Haemost 88(2):315–320 Jayo A, Conde I, Lastres P, Jimenez-Yuste V, Gonzalez-Manchon C, 2009, New insights into the expression and role of platelet factor XIII-A. J Thromb Haemost 7(7):1184–1191 Kulkarni S, Jackson SP, 2004, Platelet factor XIII and calpain negatively regulate integrin alpha(IIb)beta(3, adhesive function and thrombus growth. J Biol Chem 279(29):30697–30706 Malara A, Gruppi C, Rebuzzini P, Visai L, Perotti C, Moratti R, Balduini C, Tira ME, Balduini A, 2011, Megakaryocyte-matrix interaction within bone marrow: newroles for fibronectin and factor XIII-A. Blood 117(8):2476–2483 Guerrouahen BS, Al-Hijji I, Tabrizi AR, 2011, Osteoblastic and vascular endothelial niches, their control on normal hematopoietic stem cells, and their consequences on the development of leukemia. Stem Cells Int., DOI 10.4061/2011/375857 Den Boer ML, van Slegtenhorst M, De Menezes RX, Cheok MH, Buijs-Gladdines JG, Peters ST, Van Zutven LJ, Beverloo HB, Van der Spek PJ, Escherich G, Horstmann MA, Janka-Schaub GE, Kamps WA, Evans WE, Pieters R, 2009, A subtype of childhood acute lymphoblastic leukaemia with poor treatment outcome: a genome-wide classification study. Lancet Oncol 10(2):125–134 Harvey RC, Mullighan CG, Wang X, Dobbin KK, Davidson GS, Bedrick EJ, Chen IM, Atlas SR, Kang H, Ar K, Wilson CS, Wharton W, Murphy M, Devidas M, Carroll AJ, Borowitz MJ, Bowman WP, Downing JR, Relling M, Yang J, Bhojwani D, Carroll WL, Camitta B, Reaman GH, Smith M, Hunger SP, Willman CL, 2010, Identification of novel cluster groups in pediatric high-risk B-precursor acute lymphoblastic leukemia with gene expression profiling: correlation with genome-wide DNA copy number alterations, clinical characteristics, and outcome. Blood 116(23):4874–4884 Clappier E, Grardel N, Bakkus M, Rapion J, De Moerloose B, Kastner P, Caye A, Vivent J, Costa V, Ferster A, Lutz P, Mazingue F, Millot F, Plantaz D, Plat G, Plouvier E, Poiree M, Sirvent N, Uyttebroeck A, Yakouben K, Girard S, Dastugue N, Suciu S, Benoit Y, Bertrand Y, Cave H, European Organisation for R, Treatment of Cancer CsLG, 2015, IKZF1 deletion is an independent prognostic marker in childhood B-cell precursor acute lymphoblastic leukemia, and distinguishes patients benefiting from pulses during maintenance therapy: results of the EORTC Children's leukemia group study 58951. Leukemia 29(11):2154–2161 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 345 EP 352 DI 10.1007/s12253-017-0236-0 PG 8 ER PT J AU Lawania, Sh Singh, N Behera, D Sharma, S AF Lawania, Shweta Singh, Navneet Behera, Digamber Sharma, Siddharth TI XPC Polymorphism and Risk for Lung Cancer in North Indian Patients Treated with Platinum Based Chemotherapy and Its Association with Clinical Outcomes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Xeroderma pigmentosum group C; PCR-RFLP; overall survival; Platinum based chemotherapy ID Lung cancer; Xeroderma pigmentosum group C; PCR-RFLP; overall survival; Platinum based chemotherapy AB Xeroderma pigmentosum complementation group C plays an important role in the human repair system. As reported in previous studies its polymorphism are associated with lung cancer susceptibility. The purpose of this study is to investigate the association of XPC gene with lung cancer susceptibility, overall response and clinical outcomes amongst North Indians. A hospital based study of 370 lung cancer cases and 370 healthy controls was conducted and genotypes were determined using PCR-RFLP assay. Results were assessed using logistic linear regression adjusted for age, sex and smoking status. Survival analysis was conducted using Kaplan-Meier survival analysis and Cox regression analysis. The treatment outcomes of 167 lung cancer patients treated with platinum based chemotherapy were evaluated.The mutant genotypic variant of XPC Lys939Gln has been associated with elevated risk of lung cancer(OR:2.30;95%CI:1.41-3.73;p=0.0007) whereas XPC Ala499Val showed a highly protective effect (OR:0.25;95%CI:0.10-0.63;p=0.003). The mutant genotype of XPC Lys939Gln presented a higher risk of developing lung cancer in heavy smokers (OR: 3.71; 95%CI:1.46-9.45; p=0.005). The survival analysis presented that heterozygous genotype showed least survival in comparison with mutant genotype in XPC Ala499Val genetic variant whereas no significant association was observed in XPC Lys939Gln. In conclusion, XPC Lys939Gln is associated with significant risk towards the lung cancer whereas on contrary XPC Ala499Val shows a protective effect. C1 [Lawania, Shweta] Thapar University, Department of Biotechnology, 147002 Punjab, India. [Singh, Navneet] Post Graduate Institute of Medical Education & Research (PGIMER), Sector 14, Department of Pulmonary MedicineChandigarh, India. [Behera, Digamber] Post Graduate Institute of Medical Education & Research (PGIMER), Sector 14, Department of Pulmonary MedicineChandigarh, India. [Sharma, Siddharth] Thapar University, Department of Biotechnology, 147002 Punjab, India. RP Sharma, S (reprint author), Thapar University, Department of Biotechnology, 147002 Punjab, India. EM siddharthsharma.phd@thapar.edu CR Lyon, 2003, IARC: tobacco smoke and involuntary smoking. IARC Malik PS, Raina V, 2015, Lung cancer: prevalent trends & emerging concepts. Indian JMed Res 141(1):5–7 Corrao MA, Guindon GE, Cokkinides V, Sharma N, 2000, Building the evidence base for global tobacco control. Bull World Health Organ 78:884–890 Parkin DM, Pisani P, Lopez AD, Masuyer E, 1994, At least one in seven cases of cancer is caused by smoking: global estimates for 1985. Int J Cancer 59:494–504 Shields PG, Harris CC, 2000, Cancer risk and low-penetrance susceptibility genes in gene–environment interactions. J Clin Oncol 18:2309–2315 Lindahl T, Wood RD, 1999, Quality control by DNA repair. Science 286:1897–1905 Sancar A, 1995, DNA repair in humans. Annu Rev Genet 29:69–105 de Laat WL, Jaspers NGJ, Hoeijmakers JHJ, 1993, Molecular mechanism of nucleotide excision repair. Genes Dev 13:768–785 Wei Q, Spitz MR, 1997, The role of DNA repair capacity in susceptibility to lung cancer: a review. Cancer Metastasis Rev 16:295–307 Matutani C, Araki M, Sugawasa K, van der Spek PJ, Yamada A, Uchida A, Maekawa T, Bootsma D, Hoeijmakers JHJ, Hanaoka F, 1997, Identification and characterizationof XPC binding domain ofh HR23B. Mol Cell Biol 17:6915–6923 Khan SG, Metter EJ, Tarone RE, Bohr VA, Grossman L, Hedayati M, Bale SJ, Emmert S, Kraemer KH, 2000, A new xeroderma pigmentosum group C poly, AT, insertion/deletion polymorphism. Carcinogenesis 21:1821–1825 Sanyal S, Festa F, Sakano S, Zhang Z, Steineck G, Norming U, Wijkstrom H, Larsson P, Kumar R, Hemminki K, 2004, Polymorphisms in DNA repair and metabolic genes in bladder cancer. Carcinogenesis 25:729–734 Forsti A, Angelini S, Festa F, Sanyal S, Zhang Z, Grzybowska E, Pamula J, Pekala W, Zientek H, Hemminki K, Kumar R, 2004, Single nucleotidepolymorphisms in breast cancer. Oncol Rep 11: 917–922 SodhiKK, Bahl C, Singh N, Behera D, Sharma S, 2015, Functional genetic variants in pre-miR-146a and 196a2 genes are associated with risk of lung cancer in North Indians. Future Oncol 11(15): 2159–2173 Sugasawa K, Ng JM, Masutani C, Iwai S, van der Spek PJ et al, 1998, Xeroderma pigmentosum group C protein complex is the initiator of global genome nucleotide excision repair. Mol Cell 2: 223–232 Raaschou-Nielsen O, Sorensen M, Overvad K, Tjonneland A, Vogel U, 2008, Polymorphisms in nucleotide excision repair genes, smoking and intake of fruit and vegetables in relation to lung cancer. Lung Cancer 59:171–179 Bak H, Autrup H, Thomsen BL, Tjonneland A, Overvad K et al, 2008, Bulky DNA adducts as risk indicator of lung cancer in a Danish case-cohort study. Int J Cancer 1189:1618–1622 Jin B, Dong Y, Zhang X, Wang H, Han B, 2014, Association of XPC polymorphisms and lung cancer risk. A meta-analysis. Plos one 9(4):e93937 Kusumoto R,Masutani C, Sugasawa K et al, 2007, Diversity of the damage recognition step in the global genomic nucleotide excision repair in vitro. Mutat Res 485:219–227 Yokoi M, Masutani C, Maekawa T, Sugasawa K,OhkumaY, Hanaoka F, 2000, The xeroderma pigmentosum group C protein complex XPCHR23B plays an important role in the recruitment of transcription factor IIH to damaged DNA. J Biol Chem 275:9870–9875 Sugasawa K, Shimizu Y, Iwai S, Hanaoka F, 2002, A molecular mechanism for DNA damage recognition by the xeroderma pigmentosum group C protein complex. DNA Repair 1:95–107 Shen M, Berndt SI, Rothman N, Demarini DM, Mumford JL et al, 2005, Polymorphisms in theDNAnucleotide excision repair genes and lung cancer risk in XuanWei, China. Int J Cancer 116:768–773 Vodicka P, Kumar R, Stetina R, Sanyal S, Soucek P, Haufroid V, Dusinska M, Kuricova M, Zamecnikova M, Musak L, Buchancova J, NorppaH et al, 2004, Genetic polymorphisms in DNA repair genes and possible links with DNA repair rates, chromosomal aberrations and single-strand breaks in DNA. Carcinogenesis 25:757–763 Deutsch-Wenzel RP, Brune H, Grimmer G, Dettbarn G, Misfeld J, 1983, Experimental studies in rat lungs on the carcinogenicity and dose - r e s ponse r e l a t i o n s h i p s of e i g ht f r e q u e n t l y occurringenvironmental polycyclic aromatic hydrocarbons. J Natl Cancer Inst 71:539–544 Hoffman, 1993, Cigarette smoking and adenocarcinoma of the lung: the relevance of nicotine-derived nitrosamines. J Smoking Relat Disord 4:165–190 Xing D, Tan W, Wei Q, Lin D, 2002, Polymorphisms of the DNA repair gene XPD and risk of lung cancer in a Chinese population. Lung Cancer 38:123–129 Yin J, Vogel U,Ma Y, Guo L,Wang H, Qi R, 2006, Polymorphism of the DNA repair gene ERCC2 Lys751Gln and risk of lung cancer in a northeastern Chinese population. Cancer Genet Cytogenet 169:27–32 Zhu Y, Lai M, Yang H, Lin J, Huang M, Grossman HB, Colin PD, Wu X, 2006, Genotypes, haplotypes and diplotypes of XPC and risk of bladder cancer. Carcinogenesis 28(3):698–703 Letkova L, Matakova T, Musak L, Sarlinova M, Krutakova M, Slovakova P, Kavcova E, Jakusova V, Janickova M, Drgova A, Berzinec P, Halasova E, 2013, DNA repair genes polymorphism and lung cancer risk with the emphasis to sex differences.Mol Biol Rep 40:5261–5273 QiaoY, SpitzMR,GuoZ,MohammadH, LawrenceG, KennethKH, Wei Q, 2002, Rapid assessment of repair of ultraviolet DNA damage with a modified host-cell reactivation assay using a luciferase reporter gene and correlation with polymorphisms of DNA repair genes in normal human lymphocytes. Mutat Res 509:165–174 Nelson HH, Kelsey KT, Mott LA, Karagas MR, 2002, The XRCC1 Arg399Gln polymorphism, sunburn, and non-melanoma skin cancer: evidence of gene-environment interaction. Cancer Res 62:152–155 Bowen AR, Hanks AN, Allen SM, Alexander A, Diedrich MJ, Grossman D, 2003, Apoptosis regulators and responses in human melanocytic and keratinocytic cells. J Invest Dermatol 120:48–55 Hu Z, Wang Y, Wang X, Liang G, Miao X, Xu Y, Tan W, Wei Q, Lin D, Shen H, 2005, DNA repair gene XPC genotypes/haplotypes and risk of lung cancer in a Chinese population. Int J Cancer 115: 478–483 Matakideu A, Eisen T, Bridle H, Houlsten S. R., 2006, Nucleotide excision repair polymorphism modulate overall lung cancer survival and responsiveness to platinum based chemotherapy agents. Jr of Clinical Oncology 24: 10004. Vicent G, Mar T, 2008, Influence of DNA damage and repair upon the risk of treatment related leukemia. Leuk Lymphoma 49:204–217 Gao R, Price DK, Sissung T, Reed E, Figg WD, 2008, Ethnic disparities in Americans of European descent versus Americans of African descent related to polymorphic ERCC1, ERCC2, XRCC1, and PARP1. Mol Cancer Ther 7:1246–1250 Shi C, Qian J, Ma M, Zhang Y, Han B, 2014, Notch 3 protein, not its gene polymorphism, is associated with the chemotherapy response and prognosis of advanced NSCLC patient. Cell Physiol Biochem 34:743–752 Wu F, Zhang J, Liu Y, Zheng Y, Hu N, 2013)HIF1alpha genetic variants and protein expressions determine the response to platinum based chemotherapy and clinical outcome in patients with advanced NSCLC. Cell Physiol Biochem 32: 1566-1576. Sullivan I, Salazar J, Majem M, Pallares C, Del Rio E, Paez D, Baiget M, Barnadas, 2014, A: pharmacogenetics of the DNA repair pathways in advanced non-small cell lung cancer patients treated with platinum-based chemotherapy. Cancer Lett 353: 160–166. Tiseo M, Bordi P, Bortesi B, Boni L, Boni C, Baldini E, Grossi F, Recchia F, Zanelli F, Fontanini G, Naldi N, Campanini N, Azzoni C, Bordi C, Ardizzoni A, 2013, ERCC1/BRCA1 expression and gene polymorphisms as prognostic and predictive factors in advanced NSCLC treated with or without cisplatin. Br J Cancer 108:1695–1703 Xu TP, Shen H, Liu LX, Shu YQ, 2013, Association of ERCC1- C118T and -C8092A polymorphisms with lung cancer risk and survival of advanced-stage non-small cell lung cancer patients receiving platinum-based chemotherapy: a pooled analysis based on 39 reports. Gene 526:265–274 Li XD, Han JC, Zhang YJ, Li HB, Wu XY, 2013, Common variations of DNA repair genes are associated with response to platinum-based chemotherapy in NSCLCs. Asian Pac J Cancer Prev 14:145–148 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 353 EP 366 DI 10.1007/s12253-017-0252-0 PG 14 ER PT J AU Yang, Jy Li, D Zhang, Y Guan, Bx Gao, P Zhou, Xch Zhou, Chj AF Yang, Jing-yan Li, Dong Zhang, Yuan Guan, Bing-xin Gao, Ping Zhou, Xing-chen Zhou, Cheng-jun TI The Expression of MCM7 is a Useful Biomarker in the Early Diagnostic of Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MCM7; Ki67; Intestinal metaplastic; Gastrointestinal epithelial neoplasia ID MCM7; Ki67; Intestinal metaplastic; Gastrointestinal epithelial neoplasia AB The aim of this study was to investigate the expression of minichromosome maintenance complex component 7 (MCM7) in gastric mucosal lesions, further to find its potential effect as a biomarker to distinguish intraepithelial neoplasia from gastric mucosal lesions. MCM7 and Ki67 were detected in 93 cases of gastric mucosal lesions by immunohistochemistry. MCM7 and Ki67 expression in GT were lowest compared with other groups (P<0.001), meanwhile there were significant differences compared with Group IM and other groups in MCM7 and Ki67 expression (P<0.001). MCM7 and Ki67 expression in GSC were highest (P<0.05). Groups of LGN, HGN and GIC had no significant differences in MCM7 expression (P>0.05), but there was significant difference compared with Group LGN and Group GIC in Ki67 expression (P<0.05). MCM7 expression elevated with tumor grade increasing and had positive correlation with Ki67 significantly (r=0.940, P<0.001). Furthermore, in some cases, some tumor cells were immunoreactive to MCM7 but negative to Ki67. So we concluded that MCM7 is helpful for us to make differential diagnosis in pathological grade, MCM7 combination of Ki67 may serve as more sensitive proliferation markers for evaluation of gastric carcinoma and precancerous lesions. C1 [Yang, Jing-yan] The Second Hospital of Shandong University, Department of Pathology, 247#, Bei Yuan Street, 250033 Jinan, Shandong, China. [Li, Dong] Shandong Provincial Hospital Affiliated to Shandong University, Department of Orthopedics, 324#, Jing 5 Road, 250021 Jinan, Shandong, China. [Zhang, Yuan] The Second Hospital of Shandong University, Center of Evidence-based Medicine, 247#, Bei Yuan Street, 250033 Jinan, Shandong, China. [Guan, Bing-xin] The Second Hospital of Shandong University, Department of Pathology, 247#, Bei Yuan Street, 250033 Jinan, Shandong, China. [Gao, Ping] Yantai Affiliated Hospital of Binzhou Medical University, 717#, Jinbu Road, Muping District, 264100 Yantai, Shandong, China. [Zhou, Xing-chen] The Second Hospital of Shandong University, Department of Pathology, 247#, Bei Yuan Street, 250033 Jinan, Shandong, China. [Zhou, Cheng-jun] The Second Hospital of Shandong University, Department of Pathology, 247#, Bei Yuan Street, 250033 Jinan, Shandong, China. RP Zhou, Chj (reprint author), The Second Hospital of Shandong University, Department of Pathology, 250033 Jinan, China. EM chengjunzhou@sdu.edu.cn CR Pisani P, Parkin DM, Bray F, Ferlay J, 1999, Estimates of the worldwide mortality from 25 cancers in 1990. Int J cancer 83(1): 18–29 Zou XN, Duan JJ, Huangfu XM, Chen WQ, Zhao P, 2010, Analysis of stomach cancer mortality in the national retrospective sampling survey of death causes in China, 2004–2005. Zhonghua Yu Fang Yi Xue Za Zhi 44(5):390–397 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108., DOI 10.3322/caac.21262 CrewKD, Neugut AI, 2006, Epidemiology of gastric cancer.World J Gastroenterol 12(3):354–362 Zhang H, Sun LL, Meng YL, Song GY, Hu JJ, Lu P, Ji B, 2011, Survival trends in gastric cancer patients of Northeast China.World J Gastroenterol 17(27):3257–3262., DOI 10.3748/wjg.v17.i27.3257 Schlemper R, Riddell R, Yea K, Borchard F, Cooper H, Dawsey S, Dixon M, Fenoglio-Preiser C, Flejou J, Geboes K, 2000, The Vienna classification of gastrointestinal epithelial neoplasia. Gut 47(2):251–255 Kamiya T,Morishita T, Asakura H,Miura S, Munakata Y, Tsuchiya M, 1982, Long-term follow-up study on gastric adenoma and its relation to gastric protruded carcinoma. Cancer 50(11):2496–2503 Cristallini EG, Ascani S, Bolis GB, 1992, Association between histologic type of polyp and carcinoma in the stomach. Gastrointest Endosc 38(4):481–484 Lee J-H, Abraham SC, Kim H-S, Nam J-H, Choi C, LeeM-C, Park C-S, Juhng S-W, Rashid A, Hamilton SR, 2002, Inverse relationship between APC gene mutation in gastric adenomas and development of adenocarcinoma. Am J Pathol 161(2):611–618 Tye BK, 1999, MCM proteins in DNA replication. Annu Rev Biochem 68:649–686., DOI 10.1146/annurev.biochem.68.1.649 KangW, Tong JH, Chan AW, Cheng AS, Yu J, To K, 2014)MCM7 serves as a prognostic marker in diffuse-type gastric adenocarcinoma and siRNA-mediated knockdown suppresses its oncogenic function. Oncol Rep 31(5):2071–2078., DOI 10.3892/or.2014.3094 Choy B, LaLonde A, Que J, Wu T, Zhou Z, 2016, MCM4 and MCM7, potential novel proliferation markers, significantly correlated with Ki-67, Bmi1, and cyclin E expression in esophageal adenocarcinoma, squamous cell carcinoma, and precancerous lesions. Hum Pathol 57:126–135., DOI 10.1016/j.humpath.2016.07.013 Romanowski P, Madine MA, 1997, Mechanisms restricting DNA replication to once per cell cycle: the role of Cdc6p and ORC. Trends Cell Biol 7(1):9–10., DOI 10.1016/s0962-8924(97)30077-4 Todorov IT, Werness BA, Wang HQ, Buddharaju LN, Todorova PD, Slocum HK, Brooks JS, Huberman JA, 1998, HsMCM2/ BM28: a novel proliferation marker for human tumors and normal tissues. Lab Invest 78(1):73–78 Ritzi M, Baack M, Musahl C, Romanowski P, Laskey RA, Knippers R, 1998, Human minichromosome maintenance proteins and human origin recognition complex 2 protein on chromatin. J Biol Chem 273(38):24543–24549 Williams GH, Romanowski P, Morris L, Madine M, Mills AD, Stoeber K, Marr J, Laskey RA, Coleman N, 1998, Improved cervical smear assessment using antibodies against proteins that regulate DNA replication. Proc Natl Acad Sci U S A 95(25):14932–14937 Stoeber K, Tlsty TD, Happerfield L, Thomas GA, Romanov S, Bobrow L, Williams ED, Williams GH, 2001, DNA replication licensing and human cell proliferation. J Cell Sci 114(Pt 11): 2027–2041 Luo J-H, 2011, Oncogenic activity of MCM7 transforming cluster. World J Clin Oncol 2(2):120–124 Wei Q, Li J, Liu T, Tong X, Ye X, 2013, Phosphorylation of minichromosome maintenance protein 7, MCM7, by cyclin/ cyclin-dependent kinase affects its function in cell cycle regulation. J Biol Chem 288(27):19715–19725 Tsai HY, Hsi BL, Hung IJ, Yang CP, Lin JN, Chen JC, Tsai SF, Huang SF, 2004, Correlation ofMYCN amplification with MCM7 protein expression in neuroblastomas: a chromogenic in situ hybridization study in paraffin sections. Hum Pathol 35(11):1397–1403., DOI 10.1016/j.humpath.2004.07.014 Honeycutt KA, Chen Z, Koster MI, Miers M, Nuchtern J, Hicks J, Roop DR, Shohet JM, 2006, Deregulated minichromosomal maintenance protein MCM7 contributes to oncogene driven tumorigenesis. Oncogene 25(29):4027–4032., DOI 10.1038/sj.onc.1209435 Ren B, Yu G, Tseng GC, Cieply K, Gavel T, Nelson J, Michalopoulos G, Yu YP, Luo JH, 2006, MCM7 amplification and overexpression are associated with prostate cancer progression. Oncogene 25(7):1090–1098., DOI 10.1038/sj.onc.1209134 Nishihara K, Shomori K, Fujioka S, Tokuyasu N, Inaba A, Osaki M, Ogawa T, Ito H, 2008)Minichromosome maintenance protein 7 in colorectal cancer: implication of prognostic significance. Int J Oncol 33(2):245–251 Ota T, Clayton AC, Minot DM, Shridhar V, Hartmann LC, Gilks CB, Chien JR, 2011, Minichromosome maintenance protein 7 as a potential prognostic factor for progression-free survival in highgrade serous carcinomas of the ovary. Modern Pathol 24(2):277– 287., DOI 10.1038/modpathol.2010.202 Lau KM, Chan QK, Pang JC, Li KK, Yeung WW, Chung NY, Lui PC, Tam YS, Li HM, Zhou L, Wang Y, Mao Y, Ng HK, 2010, Minichromosome maintenance proteins 2, 3 and 7 in medulloblastoma: overexpression and involvement in regulation of cell migration and invasion. Oncogene 29(40):5475–5489., DOI 10.1038/onc. 2010.287 Marnerides A, Vassilakopoulos TP, Boltetsou E, Levidou G, Angelopoulou MK, Thymara I, Kyrtsonis MC, Pappi V, Tsopra O, Panayiotidis P, Pangalis GA, Beris P, Patsouris E, Korkolopoulou P, 2011, Immunohistochemical expression and prognostic significance of CCND3, MCM2 and MCM7 in Hodgkin lymhoma. Anticancer Res 31(10):3585–3594 Toyokawa G, Masuda K, Daigo Y, Cho HS, Yoshimatsu M, Takawa M, Hayami S, Maejima K, Chino M, Field HI, Neal DE, Tsuchiya E, Ponder BA, Maehara Y, Nakamura Y, Hamamoto R, 2011, Minichromosome maintenance protein 7 is a potential therapeutic target in human cancer and a novel prognostic marker of non-small cell lung cancer. Mol Cancer 10:65., DOI 10.1186/1476- 4598-10-65 Zhou YM, Zhang XF, Cao L, Li B, Sui CJ, Li YM, Yin ZF, 2012, MCM7 expression predicts post-operative prognosis for hepatocellular carcinoma. Liver int 32(10):1505–1509., DOI 10.1111/j.1478- 3231.2012.02846.x Ambs S, Prueitt RL, Yi M, Hudson RS, Howe TM, Petrocca F, Wallace TA, Liu C-G, Volinia S, Calin GA, 2008, Genomic profiling of microRNA and messenger RNA reveals deregulated microRNA expression in prostate cancer. Cancer Res 68(15): 6162–6170 Petrocca F, Visone R, Onelli MR, Shah MH, Nicoloso MS, de Martino I, Iliopoulos D, Pilozzi E, Liu C-G, Negrini M, 2008, E2F1-regulated microRNAs impair TGFβ-dependent cell-cycle arrest and apoptosis in gastric cancer. Cancer Cell 13(3):272–286 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90., DOI 10. 3322/caac.20107 Fujioka S, Shomori K, Nishihara K, Yamaga K, Nosaka K, Araki K, Haruki T, Taniguchi Y, Nakamura H, Ito H, 2009, Expression of minichromosome maintenance 7, MCM7, in small lung adenocarcinomas, pT1): prognostic implication. Lung cancer 65(2):223– 229., DOI 10.1016/j.lungcan.2008.11.007 Ishino H, Hara Y, Takekoshi S, Teshima T, Teramoto A, Osamura RY, Tagawa M, 2011, Ki-67 and minichromosome maintenance-7, MCM7, expression in canine pituitary corticotroph adenomas. Domest Anim Endocrinol 41(4):207–213., DOI 10.1016/j. domaniend.2011.07.002 Mueller J, Werner M, Siewert JR, 2000, Malignant progression in Barrett's esophagus: pathology and molecular biology. Recent Results Canc Res 155:29–41 Forones NM, Carvalho AP, Giannotti-Filho O, Lourenco LG, Oshima CT, 2005, Cell proliferation and apoptosis in gastric cancer and intestinal metaplasia. Arq Gastroenterol 42(1):30–34., DOI 10. 1590/S0004-28032005000100008 Al-Moundhri MS, Nirmala V, Al-Hadabi I, Al-Mawaly K, Burney I, Al-Nabhani M, Thomas V, Ganguly SS, Grant C, 2005, The prognostic significance of p53, p27 kip1, p21 waf1, HER-2/neu, and Ki67 proteins expression in gastric cancer: a clinicopathological and immunohistochemical study of 121 Arab patients. J Surg Oncol 91(4):243–252., DOI 10.1002/jso.20324 Zheng Y,Wang L, Zhang JP, Yang JY, Zhao ZM, Zhang XY, 2010, Expression of p53, c-erbB-2 and Ki67 in intestinal metaplasia and gastric carcinoma. World J Gastroenterol 16(3):339–344 Docea AO, Mitrut P, Grigore D, Pirici D, Calina DC, Gofita E, 2012, Immunohistochemical expression of TGF beta, TGF-beta), TGF beta receptor 1, TGFBR1), and Ki67 in intestinal variant of gastric adenocarcinomas. Rom J Morphol Embryo 53(3 Suppl): 683–692 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 367 EP 372 DI 10.1007/s12253-017-0251-1 PG 6 ER PT J AU Vosmik, M Laco, J Sirak, I Dvorak, J Lochman, P Hodek, M Mala, P Rejchrt, S Repak, R Lesko, M Ferko, A Ryska, A Melichar, B Petera, J AF Vosmik, Milan Laco, Jan Sirak, Igor Dvorak, Josef Lochman, Petr Hodek, Miroslav Mala, Petra Rejchrt, Stanislav Repak, Rudolf Lesko, Michal Ferko, Alexander Ryska, Ales Melichar, Bohuslav Petera, Jiri TI Histopathologic Features are more Important Prognostic Factors than Primary Tumour Location in Gastro-oesophageal Adenocarcinoma Treated with Preoperative Chemoradiation and Surgery SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Gastro-oesophageal junction cancer; Distal oesophageal cancer; Preoperative chemoradiation; Prognostic factor ID Gastric cancer; Gastro-oesophageal junction cancer; Distal oesophageal cancer; Preoperative chemoradiation; Prognostic factor AB The aim of present study was to evaluate the impact of primary tumour location and other factors on the outcome of preoperative chemoradiation followed by surgery in adenocarcinomas of distal oesophagus, gastro-oesophageal junction and stomach. We retrospectively reviewed the institutional patient database. The therapeutic response was re-evaluated as a percentage of residual tumor cells in surgical resection specimens. Overall survival (OS) and disease-free survival (DFS) were assessed. The effect primary tumour location, clinical and pathological TNM stage, and histopathological factors (histological type, grade, angioinvasion, perineural invasion, tumour response) on treatment outcome were evaluated. A total of 108 patients underwent preoperative chemoradiation for adenocarcinoma of distal oesophagus, gastro-oesophageal junction or stomach. The median prescribed dose of radiation was 45 Gy. The concurrent chemotherapy consisted of 5- fluorouracil +/− cisplatin +/− taxanes. R0 resection was achieved in 80 patients (74%). The complete response was observed in 19%. The median follow-up was 50.8 months. Three-year and 5-year OS and DFS were 36.2% and 25.3%; and 28.1% and 23.7%, respectively. Pretreatment T-stage, pathological N-stage, radicality of resection, histological subtype, grade, angioinvasion and perineural invasion, were identified as statistical significant OS predictors in univariate analysis; pathological N-stage, radicality of resection and angioinvasion, in multivariate analysis. The primary tumor location did not influence the prognosis. The pathologic response to chemoradiation had borderline significance. In conclusion, no prognostic impact of primary tumour location, in contrast to other investigated factors, was evident in the present study. The most important predictors of prognosis were angioinvasion status and pN– stage. C1 [Vosmik, Milan] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and RadiotherapyHradec Kralove, Czech Republic. [Laco, Jan] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, The Fingerland Department of PathologyHradec Kralove, Czech Republic. [Sirak, Igor] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and RadiotherapyHradec Kralove, Czech Republic. [Dvorak, Josef] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and RadiotherapyHradec Kralove, Czech Republic. [Lochman, Petr] University of Defense, Faculty of Military Health SciencesHradec Kralove, Czech Republic. [Hodek, Miroslav] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and RadiotherapyHradec Kralove, Czech Republic. [Mala, Petra] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and RadiotherapyHradec Kralove, Czech Republic. [Rejchrt, Stanislav] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Second Department of Internal Medicine-GastroenterologyHradec Kralove, Czech Republic. [Repak, Rudolf] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Second Department of Internal Medicine-GastroenterologyHradec Kralove, Czech Republic. [Lesko, Michal] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of SurgeryHradec Kralove, Czech Republic. [Ferko, Alexander] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of SurgeryHradec Kralove, Czech Republic. [Ryska, Ales] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, The Fingerland Department of PathologyHradec Kralove, Czech Republic. [Melichar, Bohuslav] Palacky University, Faculty of Medicine and Dentistry, University Hospital Olomouc, Department of OncologyOlomouc, Czech Republic. [Petera, Jiri] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and RadiotherapyHradec Kralove, Czech Republic. RP Vosmik, M (reprint author), Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, Hradec Kralove, Czech Republic. EM milan.vosmik@fnhk.cz CR Urschel JD, Vasan H, 2003, A meta-analysis of randomized controlled trials that compared neoadjuvant chemoradiation and surgery to surgery alone for resectable esophageal cancer. Am J Surg 185:538–543., DOI 10.1016/S0002-9610(03)00066-7 Fiorica F, Di Bona D, Schepis F, Licata A, Shahied L, Venturi A, Falchi AM, Craxi A, Camma C, 2004, Preoperative chemoradiotherapy for oesophageal cancer: a systematic review and meta-analysis. Gut 53:925–930., DOI 10.1136/gut.2003.025080 Shapiro J, van Lanschot JJ, Hulshof MC, van Hagen P, van Berge Henegouwen MI, Wijnhoven BP, van Laarhoven HW, Nieuwenhuijzen GA, Hospers GA, Bonenkamp JJ, Cuesta MA, Blaisse RJ, Busch OR, Ten Kate FJ, Creemers GJ, Punt CJ, Plukker JT, Verheul HM, Bilgen EJ, van Dekken H, van der Sangen MJ, Rozema T, Biermann K, Beukema JC, Piet AH, van RijCM, Reinders JG, Tilanus HW, Steyerberg EW, van der Gaast A, 2015, Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer, CROSS): longterm results of a randomised controlled trial. Lancet Oncol 16: 1090–1098., DOI 10.1016/S1470-2045(15)00040-6 Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, Scarffe JH, Lofts FJ, Falk SJ, Iveson TJ, Smith DB, Langley RE, Verma M, Weeden S, Chua YJ, Trial Participants MAGIC, 2006, Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med 355:11–20., DOI 10.1056/NEJMoa055531 Ychou M, Boige V, Pignon JP, Conroy T, Bouche O, Lebreton G, Ducourtieux M, Bedenne L, Fabre JM, Saint-Aubert B, Geneve J, Lasser P, Rougier P, 2011, Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCDmulticenter phase III trial. J Clin Oncol 29:1715–1721., DOI 10.1200/JCO.2010.33.0597 Macdonald JS, Smalley SR, Benedetti J, Hundahl SA, Estes NC, Stemmermann GN, Haller DG, Ajani JA, Gunderson LL, Jessup JM, Martenson JA, 2001, Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med 345:725–730., DOI 10. 1056/NEJMoa010187 Ajani JA, Winter K, Okawara GS, Donohue JH, Pisters PW, Crane CH, Greskovich JF, Anne PR, Bradley JD, Willett C, Rich TA, 2006, Phase II trial of preoperative chemoradiation in patients with localized gastric adenocarcinoma, RTOG 9904): quality of combined modality therapy and pathologic response. J Clin Oncol 24: 3953–3958., DOI 10.1200/JCO.2006.06.4840 Leong T, Smithers BM, Michael M, Gebski V, Boussioutas A, Miller D, Simes J, Zalcberg J, Haustermans K, Lordick F, Schuhmacher C, Swallow C, Darling G, Wong R, 2015, TOPGEAR: a randomised phase III trial of perioperative ECF chemotherapy versus preoperative chemoradiation plus perioperative ECF chemotherapy for resectable gastric cancer, an international, intergroup trial of the AGITG/TROG/EORTC/NCIC CTG). BMC Cancer 15:532., DOI 10.1186/s12885-015-1529-x Petera J, Dvorak J, Melichar B, Kabelac K, Zoul Z, Voboril Z, Hazukova R, 2002, Chemoradiotherapy in neoadjuvant treatment of gastric cancer. Klinicka Onkologie 15:216–218 Dvorak J, Melichar B, Petera J,Kabela K, VosmikM, Vesly P, Sirak I, Zoul Z, Ryska A, Jandik P, 2009, Preoperative neoadjuvant chemoradiation for locally advanced gastric adenocarcinoma. Rep Prac Oncol Radiother 14:169–175 Sirak I, Petera J, Hatlova J, VosmikM, Melichar B, Dvorak J, Zoul Z, Tycova V, LeskoM, HajduchM(2008, Epidermal growth factor receptor as a predictor of tumor response to preoperative chemoradiation in locally advanced gastric carcinoma. Strahlenther Onkol 184:592–597., DOI 10.1007/s00066-008-1880-9 Sobin LH, Gospodarowicz MK,Wittekind C, 2009, TNM classification of malignant tumors, 7th edn. Wiley-Blackwell, Oxford Mandard AM, Dalibard F, Mandard JC,Marnay J, Henry-Amar M, Petiot JF, Roussel A, Jacob JH, Segol P, Samama G et al, 1994, Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Cancer 73:2680–2686., DOI 10.1002/1097-0142(19940601, 73:11<2680::AID-CNCR2820731105>3.0.CO;2-C OrdituraM, Galizia G, Di Martino N, Ancona E, Castoro C, Pacelli R, Morgillo F, Rossetti S, Gambardella V, Farella A, Laterza MM, Ruol A, Fabozzi A, Napolitano V, Iovino F, Lieto E, Fei L, Conzo G, Ciardiello F, De Vita F, 2014, Effect of preoperative chemoradiotherapy on outcome of patients with locally advanced esophagogastric junction adenocarcinoma-a pilot study. Curr Oncol 21:125–133., DOI 10.3747/co.21.1570 Rohatgi PR, Mansfield PF, Crane CH, Wu TT, Sunder PK, Ross WA, Morris JS, Pisters PW, Feig BW, Gunderson LL, Ajani JA, 2006, Surgical pathology stage by American joint commission on cancer criteria predicts patient survival after preoperative chemoradiation for localized gastric carcinoma. Cancer 107:1475– 1482., DOI 10.1002/cncr.22180 Stahl M, Walz MK, Stuschke M, Lehmann N, Meyer HJ, Riera- Knorrenschild J, Langer P, Engenhart-Cabillic R, Bitzer M, Konigsrainer A, Budach W, Wilke H, 2009, Phase III comparison of preoperative chemotherapy compared with chemoradiotherapy in patients with locally advanced adenocarcinoma of the esophagogastric junction. J Clin Oncol 27:851–856., DOI 10.1200/ JCO.2008.17.0506 Burmeister BH, Thomas JM, Burmeister EA, Walpole ET, Harvey JA, Thomson DB, Barbour AP, Gotley DC, Smithers BM, 2011, Is concurrent radiation therapy required in patients receiving preoperative chemotherapy for adenocarcinoma of the oesophagus? A randomised phase II trial. Eur J Cancer 47:354–360., DOI 10.1016/ j.ejca.2010.09.009 Lowy AM, Feig BW, Janjan N, Rich TA, Pisters PW, Ajani JA, Mansfield PF, 2001, A pilot study of preoperative chemoradiotherapy for resectable gastric cancer. Ann Surg Oncol 8:519–524., DOI 10.1245/aso.2001.8.6.519 Ajani JA,Mansfield PF, Janjan N, Morris J, Pisters PW, Lynch PM, Feig B, Myerson R, Nivers R, Cohen DS, Gunderson LL, 2004, Multi-institutional trial of preoperative chemoradiotherapy in patients with potentially resectable gastric carcinoma. J Clin Oncol 22:2774–2780., DOI 10.1200/JCO.2004.01.015 Wydmanski J, Suwinski R, Poltorak S, Maka B, Miszczyk L, Wolny E, Bielaczyc G, Zajusz A, 2007, The tolerance and efficacy of preoperative chemoradiotherapy followed by gastrectomy in operable gastric cancer, a phase II study. Radiother Oncol 82:132– 136., DOI 10.1016/j.radonc.2007.01.009 Reed VK, Krishnan S, Mansfield PF, Bhosale PR, Kim M, Das P, Janjan NA, Delclos ME, Lowy AM, Feig BW, Pisters PW, Ajani JA, Crane CH, 2008, Incidence, natural history, and patterns of locoregional recurrence in gastric cancer patients treated with preoperative chemoradiotherapy. Int J Radiat Oncol Biol Phys 71:741– 747., DOI 10.1016/j.ijrobp.2007.10.030 Pepek JM, Chino JP,Willett CG, Palta M, Blazer Iii DG, Tyler DS, Uronis HE, Czito BG, 2013, Preoperative chemoradiotherapy for locally advanced gastric cancer. Radiat Oncol 8:6., DOI 10.1186/ 1748-717X-8-6 Rostom Y, Zaghloul H, Khedr G, El-Shazly W, Abd-Allah D, 2013, Docetaxel-based preoperative chemoradiation in localized gastric cancer: impact of pathological complete response on patient outcome. J Gastrointest Cancer 44:162–169., DOI 10.1007/s12029-012-9449-3 Trip AK, Poppema BJ, van Berge Henegouwen MI, Siemerink E, Beukema JC, Verheij M, Plukker JT, Richel DJ, Hulshof MC, van Sandick JW, Cats A, Jansen EP, Hospers GA, 2014, Preoperative chemoradiotherapy in locally advanced gastric cancer, a phase I/II feasibility and efficacy study. Radiother Oncol 112:284–288., DOI 10.1016/j.radonc.2014.05.003 Diaz-Gonzalez JA, Rodriguez J, Hernandez-Lizoain JL, Ciervide R, Gaztanaga M, San Miguel I, Arbea L, Aristu JJ, Chopitea A, Martinez-Regueira F, Valenti V, Garcia-Foncillas J, Martinez-Monge R, Sola JJ, 2011, Patterns of response after preoperative treatment in gastric cancer. Int J Radiat Oncol Biol Phys 80:698– 704., DOI 10.1016/j.ijrobp.2010.02.054 Chakravarty T, Crane CH, Ajani JA, Mansfield PF, Briere TM, Beddar AS, Mok H, Reed VK, Krishnan S, Delclos ME, Das P, 2012, Intensity-modulated radiation therapy with concurrent chemotherapy as preoperative treatment for localized gastric adenocarcinoma. Int J Radiat Oncol Biol Phys 83:581–586., DOI 10.1016/j. ijrobp.2011.07.035 Walsh TN, Noonan N, Hollywood D, Kelly A, Keeling N, Hennessy TP, 1996, A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 335:462– 467., DOI 10.1056/NEJM199608153350702 Rivera F, Galan M, Tabernero J, Cervantes A, Vega-Villegas ME, Gallego J, Laquente B, Rodriguez E, Carrato A, Escudero P, Massuti B, Alonso-Orduna V, Cardenal A, Saenz A, Giralt J, Yuste AL, Anton A, Aranda E, Spanish Cooperative Group for Digestive Tumor Therapy, 2009, Phase II trial of preoperative irinotecan-cisplatin followed by concurrent irinotecan-cisplatin and radiotherapy for resectable locally advanced gastric and esophagogastric junction adenocarcinoma. Int J Radiat Oncol Biol Phys 75:1430–1436., DOI 10.1016/j.ijrobp.2008.12.087 Klautke G, Foitzik T, Ludwig K, Ketterer P, Klar E, Fietkau R, 2004, Neoadjuvant radiochemotherapy in locally advanced gastric carcinoma. Strahlenther Onkol 180:695–700., DOI 10.1007/s00066- 004-9194-z CheedellaNK, Suzuki A, Xiao L,HofstetterWL, MaruDM, Taketa T, Sudo K, Blum MA, Lin SH,Welch J, Lee JH, BhutaniMS, Rice DC, Vaporciyan AA, Swisher SG, Ajani JA, 2013, Association between clinical complete response and pathological complete response after preoperative chemoradiation in patients with gastroesophageal cancer: analysis in a large cohort. Ann Oncol 24: 1262–1266., DOI 10.1093/annonc/mds617 Lockhart AC, Reed CE, Decker PA, Meyers BF, Ferguson MK, Oeltjen AR, Putnam JB, Cassivi SD, Montero AJ, Schefter TE, American College of Surgeons Oncology Group, 2014, Phase II study of neoadjuvant therapy with docetaxel, cisplatin, panitumumab, and radiation therapy followed by surgery in patients with locally advanced adenocarcinoma of the distal esophagus, ACOSOG Z4051). Ann Oncol 25:1039–1044., DOI 10.1093/ annonc/mdu091 Jacome AA, Sankarankutty AK, dos Santos JS, 2015, Adjuvant therapy for gastric cancer: what have we learned since INT0116? World J Gastroenterol 21:3850–3859., DOI 10.3748/wjg.v21.i13.3850 Badgwell B, Blum M, Estrella J, Chiang YJ, Das P, Matamoros A, Fournier K, Mansfield P, Ajani J, 2015, Predictors of survival in patients with resectable gastric cancer treated with preoperative chemoradiation therapy and gastrectomy. J Am Coll Surg 221:83– 90., DOI 10.1016/j.jamcollsurg.2015.04.004 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 373 EP 383 DI 10.1007/s12253-017-0253-z PG 11 ER PT J AU Wang, M Gao, W Dehong, L Teng, L AF Wang, Miao Gao, Wei Dehong, Lu Teng, Lianghong TI MiR-1271 Inhibits Cell Growth in Prostate Cancer by Targeting ERG SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; miR-1271; ERG; miR-21 ID Prostate cancer; miR-1271; ERG; miR-21 AB ETS-related gene (ERG) is an oncogene that is commonly found in prostate cancer (PCa). Several miRNAs have been reported to be associated with PCa. This study was undertaken to identify miRNAs that act as a tumor suppressor by targeting ERG. We collected 70 PCa and paired adjacent non-tumor (Adjacent-N) tissues and analyzed ERG expression by immunohistochemistry(IHC). Expression of 6 miRNAs (miR-21,-34a,-96,-125b,-150 and miR-1271) was analyzed by qRT-PCR. Luciferase reporter assay was performed to examine miRNA binding to the 3′-UTR of target genes. The effects of ectopic expression of miRNA on cell growth and MAPK signaling pathway were investigated in in PC-3 and LNCaP cell lines. Among 70 PCa cases, 13 (18.6%) were ERG positive. No significant difference of miR-34a, 96, 125b, and 150 expression was found between PCa and Adjacent-N tissues. Significantly higher level of miR-21 and lower level of miR-1271 expression were found in cancer tissues. Furthermore, miR-1271 was down-regulated in ERG-positive PCa cases (p < 0.05). Based on luciferase reporter assay, we identified ERG gene as a direct target gene for miR-1271. Transfection of a miR-1271 mimics into PC-3 and LNCaP cells repressed the ERG expression and significantly suppressed cell growth. Lastly, ectopic expression of miR-1271 inhibits AKT1, p38gama and CREB kinase activity. Our results suggested that reduced expression of miR- 1271 may be involved in the ERG expression and that miR- 1271 could be a therapeutic target for ERG-positive prostate cancer patients. C1 [Wang, Miao] Capital Medical University, Basic Medical College, Department of Pathology, No.45 Changchun Street, Xicheng District, 100053 Beijing, China. [Gao, Wei] Capital Medical University, Xuanwu Hospital, Department of PathologyBeijing, China. [Dehong, Lu] Capital Medical University, Xuanwu Hospital, Department of PathologyBeijing, China. [Teng, Lianghong] Capital Medical University, Xuanwu Hospital, Department of PathologyBeijing, China. RP Teng, L (reprint author), Capital Medical University, Xuanwu Hospital, Department of Pathology, Beijing, China. EM tenglh2012@163.com CR Tomlins SA et al, 2005, Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310(5748): 644–648 Mosquera JM et al, 2009, Prevalence of TMPRSS2-ERG fusion prostate cancer among men undergoing prostate biopsy in the United States. Clin Cancer Res 15(14):4706–4711 Magi-Galluzzi C et al, 2011, TMPRSS2-ERG gene fusion prevalence and class are significantly different in prostate cancer of Caucasian, African-American and Japanese patients. Prostate 71(5):489–497 Deng JH et al, 2013, MiRNA targets of prostate cancer. Methods Mol Biol 936:357–369 Shi XB et al, 2011, miR-125b promotes growth of prostate cancer xenograft tumor through targeting pro-apoptotic genes. Prostate 71(5):538–549 Yu JJ et al, 2014, miR-96 promotes cell proliferation and clonogenicity by down-regulating of FOXO1 in prostate cancer cells. Med Oncol 31(4):910 Zhang HL et al, 2011, Serum miRNA-21: elevated levels in patients with metastatic hormone-refractory prostate cancer and potential predictive factor for the efficacy of docetaxel-based chemotherapy. Prostate 71(3):326–331 Kojima K et al, 2010, MiR-34a attenuates paclitaxel-resistance of hormone-refractory prostate cancer PC3 cells through direct and indirect mechanisms. Prostate 70(14):1501–1512 Folini M et al, 2010, miR-21: an oncomir on strike in prostate cancer. Mol Cancer 9:12 Jensen KP, Covault J, 2011, Human miR-1271 is a miR-96 paralog with distinct non-conserved brain expression pattern. Nucleic Acids re 39(2):701–711 Humphrey PA et al, 2016, WHO classification: Tumours of the urinary system and male genital organs. IARC Press 94(4):49–64 Wang M et al, 2008, miRNA analysis in B-cell chronic lymphocytic leukaemia: proliferation centres characterized by low miR- 150 and high BIC/miR-155 expression. J Pathol 215(1):13–20 Wang M et al, 2009, JNK is constitutively active in mantle cell lymphoma: cell cycle deregulation and polyploidy by JNK inhibitor SP600125. J Pathol 218(1):95–103 Suarez CD, Deng X, Hu CD, 2014, Targeting CREB inhibits radiation-induced neuroendocrine differentiation and increases radiation-induced cell death in prostate cancer cells. Am J Cancer Res 4(6):850–861 Edlind MP, Hsieh AC, 2014, PI3K-AKT-mTOR signaling in prostate cancer progression and androgen deprivation therapy resistance. Asian J Androl 16(3):378–386 Chen L et al, 2004, ERK1/2 and p38 pathways are required for P2Y receptor-mediated prostate cancer invasion. Cancer Lett 215(2): 239–247 Powell IJ et al, 2014, Considering race and the potential for ERG expression as a biomarker for prostate cancer. Perinat Med 11(4): 409–412 Fayyaz S, Farooqi AA, 2013, miRNA and TMPRSS2-ERG do not mind their own business in prostate cancer cells. Immunogenetics 65(5):315–332 Park K et al, 2010, Antibody-based detection of ERG rearrangement-positive prostate cancer. Neoplasia 12(7):590– 598 Suh JH et al, 2012, ERG immunohistochemistry and clinicopathologic characteristics in Korean prostate adenocarcinoma patients. Korean J Pathol 46(5):423–428 Li X et al, 2014, MicroRNA-21, miR-21, post-transcriptionally downregulates tumor suppressor PDCD4 and promotes cell transformation, proliferation, and metastasis in renal cell carcinoma. Cell Physiol Biochem 33(6):1631–1642 Zhang JG et al, 2010, MicroRNA-21, miR-21, represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer, NSCLC). Clin Chim Acta 411(11–12):846–852 Guan Yet al, 2016, Association of microRNA-21 expression with clinicopathological characteristics and the risk of progression in advanced prostate cancer patients receiving androgen deprivation therapy. Prostate 76(11):986–993 Sun YM, Lin KY, Chen YQ, 2013, Diverse functions of miR-125 family in different cell contexts. J Hematol Oncol 6:6 Ma Yet al, 2012)miR-150 as a potential biomarker associated with prognosis and therapeutic outcome in colorectal cancer. Gut 61(10): 1447–1453 Liu DZ et al, 2015, MIR-150 promotes prostate cancer stem cell development via suppressing p27Kip1. Eur rev med Pharmacol Sci 19(22):4344–4352 Zhong J et al, 2017, Inhibition of DIXDC1 by microRNA-1271 suppresses the proliferation and invasion of prostate cancer cells. Biochem Biophys Res Commun 484(4):794–800 Yang M et al, 2014, miR-1271 regulates cisplatin resistance of human gastric cancer cell lines by targeting IGF1R, IRS1, mTOR, and BCL2. Anti Cancer Agents Med Chem 14(6):884–891 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 385 EP 391 DI 10.1007/s12253-017-0254-y PG 7 ER PT J AU Franco, DL Marrelli, D Voglino, C Vindigni, C Ferrara, F Mare, DG Iudici, L Marini, M Roviello, F AF Franco, De Lorenzo Marrelli, Daniele Voglino, Costantino Vindigni, Carla Ferrara, Francesco Mare, Di Giulio Iudici, Livio Marini, Mario Roviello, Franco TI Prognostic Value of Perineural Invasion in Resected Gastric Cancer Patients According to Lauren Histotype SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Perineural invasion; Lauren histotype; Extended lymphadenectomy; Prognostic factors ID Gastric cancer; Perineural invasion; Lauren histotype; Extended lymphadenectomy; Prognostic factors AB The purpose of this study is to investigate perineural invasion (PNI) as a prognostic factor in gastric cancer patients. 455 patients submitted to extended (D2 or more) lymphadenectomy (median number of 39 retrieved lymph nodes, range: 15–140) between 1995 and 2012 were retrospectively studied. Patients were categorized in two groups according to the PNI status, and PNI positivity was assessed in presence of cancer cells in the perinerium or the neural fascicles using hematoxylin and eosin staining. Median follow-up for surviving patients was 80.3 months. Survival analysis was performed by univariate and multivariate analysis, using a Cox proportional hazards model. 162 patients (33.9%) had positive PNI; this was strongly associated with advanced stages of disease, residual tumor, lymphovascular invasion, Lauren diffuse-mixed histotype and tumor size. Five-year cancer-related survival was 65,7% and 20,6% in PNI negative vs. positive groups, respectively (p < 0.001). The prognostic impact of PNI at univariate analysis was particularly evident in patients submitted to R0 surgery, early as well as advanced stage, advanced nodal stage and T status. At multivariate analysis, PNI did not result statistically significant in the overall series, but emerged as an independent prognostic factor in the group of patients with Lauren intestinal histotype (p = 0.005, hazard ratio: 1.99, 95% confidence interval 1.24–3.19). PNI is related to advanced stage and poor long-term survival in gastric cancer, and may serve as an adjunctive prognostic factor in the intestinal histotype. C1 [Franco, De Lorenzo] University of Siena, Department of Medicine, Surgery and Neurosciences, Unit of Surgical Oncology, 53100 Siena, Italy. [Marrelli, Daniele] University of Siena, Department of Medicine, Surgery and Neurosciences, Unit of Surgical Oncology, 53100 Siena, Italy. [Voglino, Costantino] University of Siena, Department of Medicine, Surgery and Neurosciences, Unit of Surgical Oncology, 53100 Siena, Italy. [Vindigni, Carla] University of Siena, Department of Medicine, Surgery and Neurosciences, Unit of Pathology, 53100 Siena, Italy. [Ferrara, Francesco] University of Siena, Department of Medicine, Surgery and Neurosciences, Unit of Surgical Oncology, 53100 Siena, Italy. [Mare, Di Giulio] University of Siena, Department of Medicine, Surgery and Neurosciences, Unit of Surgical Oncology, 53100 Siena, Italy. [Iudici, Livio] University of Siena, Department of Medicine, Surgery and Neurosciences, Unit of Surgical Oncology, 53100 Siena, Italy. [Marini, Mario] Santa Maria alle Scotte Hospital, Department of Oncology, Unit of gastroenterology and endoscopy, 53100 Siena, Italy. [Roviello, Franco] University of Siena, Department of Medicine, Surgery and Neurosciences, Unit of Surgical Oncology, 53100 Siena, Italy. RP Marrelli, D (reprint author), University of Siena, Department of Medicine, Surgery and Neurosciences, Unit of Surgical Oncology, 53100 Siena, Italy. EM marrelli@unisi.it CR Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D, Bray F, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403., DOI 10.1016/j.ejca.2012.12.027 Marrelli D, Pedrazzani C, Morgagni P, de Manzoni G, Pacelli F, Coniglio A, Marchet A, Saragoni L, Giacopuzzi S, Roviello F, Italian Research Group for Gastric Cancer, 2011, Changing clinical and pathological features of gastric cancer over time. Br J Surg 98: 1273–1283., DOI 10.1002/bjs.7528 Japanese Gastric Cancer Association, 2016, Japanese gastric cancer treatment guidelines 2014, ver. 4). Gastric Cancer,, DOI 10. 1007/s10120-016-0622-4 De Manzoni G, Marrelli D, Baiocchi GL, Morgagni P, Saragoni L, Degiuli M, Donini A, Fumagalli U, Mazzei MA, Pacelli F, Tomezzoli A, Berselli M, Catalano F, Di Leo A, Framarini M, Giacopuzzi S, Graziosi L, Marchet A, Marini M, Milandri C, Mura G, Orsenigo E, Quagliuolo V, Rausei S, Ricci R, Rosa F, Roviello G, Sansonetti A, Sgroi G, Tiberio GA, Verlato G, Vindigni C, Rosati R, Roviello F, 2017, The Italian research Group for Gastric Cancer, GIRCG, guidelines for gastric cancer staging and treatment: 2015. Gastric Cancer., DOI 10.1007/s10120- 016-0615-3 Waddell T, Verheij M, Allum W, Cunningham D, Cervantes A, Arnold D, European Society for Medical Oncology, ESMO), European Society of Surgical Oncology, ESSO), European Society of Radiotherapy and Oncology, ESTRO,, 2014, Gastric cancer: ESMO-ESSO-ESTRO clinical practice guidelines for diagnosis, treatment and follow-up. Eur J Surg Oncol 40:584–591., DOI 10.1016/j.ejso.2013.09.020 MacDonald JS, Smalley SR, Benedetti J, Hundahl SA, Estes NC, Stemmermann GN, Haller DG, Ajani JA, Gunderson LL, Jessup JM, Martenson JA, 2001, Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J med 345:725–730., DOI 10. 1056/NEJMoa010187 Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, Scarffe JH, Lofts FJ, Falk SJ, Iveson TJ, Smith DB, Langley RE, Verma M, Weeden S, Chua YJ, Trial Participants MAGIC, 2006, Perioperative chemotherapy versus surgery alone for Resectable gastroesophageal cancer. N Engl J med 355:11–20., DOI 10.1056/NEJMoa055531 Verlato G, Marrelli D, Accordini S, Bencivenga M, Di Leo A, Marchet A, Petrioli R, Zoppini G, Muggeo M, Roviello F, de Manzoni G, 2015, Short-term and long-term risk factors in gastric cancer. World J Gastroenterol 21:6434–6443., DOI 10.3748/wjg. v21.i21.6434 Marrelli D, Morgagni P, de Manzoni G, Marchet A, Baiocchi GL, Giacopuzzi S, Coniglio A, Mocellin S, Saragoni L, Roviello F, Italian Research Group for Gastric Cancer, 2015, External validation of a score predictive of recurrence after radical surgery for noncardia gastric cancer: results of a follow-up study. J am Coll Surg 221:280–290., DOI 10.1016/j.jamcollsurg.2015.03.042 Clayman GL, Lee JJ, HolsingerFC ZX, Duvic M, El-Naggar AK, Prieto VG, Altamirano E, Tucker SL, Strom SS, Kripke ML, Lippman SM, 2005)Mortality risk from squamous cell skin cancer. J Clin Oncol 23:759–765., DOI 10.1200/JCO.2005.02.155 Kang M, Oh JJ, Lee S, Hong SK, Lee SE, Byun SS, 2016, Perineural invasion and Lymphovascular invasion are associated with increased risk of biochemical recurrence in patients undergoing radical prostatectomy. Ann Surg Oncol 23:2699–2706., DOI 10. 1245/s10434-016-5153-z Shimada K, Nara S, Esaki M, Sakamoto Y, Kosuge T, Hiraoka N, 2011, Intrapancreatic nerve invasion as a predictor for recurrence after pancreaticoduodenectomy in patients with invasive ductal carcinoma of the pancreas. Pancreas 40:464–468., DOI 10.1097/MPA. 0b013e31820b5d37 Marrelli D, Pedrazzani C, Neri A, Corso G, DeStefano A, Pinto E, Roviello F, 2007, Complications after extended, D2, and superextended, D3, lymphadenectomy for gastric cancer: analysis of potential risk factors. Ann Surg Oncol 14:25–33., DOI 10.1245/ s10434-006-9063-3 Roviello F, Pedrazzani C, Marrelli D, Di Leo A, Caruso S, Giacopuzzi S, Corso G, de Manzoni G, 2010, Super-extended, D3, lymphadenectomy in advanced gastric cancer. Eur J Surg Oncol 36:439–446., DOI 10.1016/j.ejso.2010.03.008 Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, 2010, Stomach. In: American joint committee on cancer staging manual, 7th edn. Springer, New York, pp 117–126 Marrelli D, Morgagni P, de Manzoni G, Coniglio A, Marchet A, Saragoni L, Tiberio G, Roviello F, Italian Research Group for Gastric Cancer, IRGGC,, 2012, Prognostic value of the 7th AJCC/UICC TNM classification of noncardia gastric cancer: analysis of a large series from specialized western centers. Ann Surg 255:486–491., DOI 10.1097/SLA.0b013e3182389b1a Liebig C, Ayala G,Wilks JA, Berger DH, Albo D, 2009, Perineural invasion in cancer: a review of the literature. Cancer 115:3379– 3391., DOI 10.1002/cncr.24396 Deng J,YouQ, GaoY,Yu Q, Zhao P, ZhengY, FangW, Xu N, Teng L, 2014, Prognostic value of perineural invasion in gastric cancer: a systematic review and meta-analysis. PLoS One 9:e88907., DOI 10. 1371/journal.pone.0088907 Fouquet T, Germain A, Brunaud L, Bresler L, Ayav A, 2014, Is perineural invasion more accurate than other factors to predict early recurrence after pancreatoduodenectomy for pancreatic head adenocarcinoma? World J Surg 38:2132–2137., DOI 10.1007/s00268- 014-2465-7 Saeter T, Bogaard M, Vlatkovic L, Waaler G, Servoll E, Nesland JM, Axcrona K, Axcrona U, 2016, The relationship between perineural invasion, tumor grade, reactive stroma and prostate cancerspecific mortality: a clinicopathologic study on a population-based cohort. Prostate 76:207–214., DOI 10.1002/pros.23112 Sertkaya Z, Ozturk MI, Koca O, Gunes M, Karaman MI, 2014, Predictive values for extracapsular extension in prostate cancer patients with PSA values below 10 ng/mL. Turk J Urol 40:130–133., DOI 10.5152/tud.2014.00086 Ozcan F, 2001, Correlation of perineural invasion on radical prostatectomy specimens with other pathologic prognostic factors and PSA failure. Eur Urol 40:308–312 Duraker N, Sisman S, Can G, 2003, The significance of perineural invasion as a prognostic factor in patients with gastric carcinoma. Surg Today 33:95–10 Tanaka A, Watanabe T, Okuno K, Yasutomi M, 1994, Perineural invasion as a predictor of recurrence of gastric cancer. Cancer 73: 550–555 Bilici A, Seker M, Ustaalioglu BB, Kefeli U, Yildirim E, Yavuzer D, Aydin FM, Salepci T, Oncel M, Gumus M, 2010, Prognostic significance of perineural invasion in patients with gastric cancer who underwent curative resection. Ann Surg Oncol 17:2037–2044., DOI 10.1245/s10434-010-1027-y Jiang N, Deng JY, Liu Y, Ke B, Liu HG, Liang H, 2014, Incorporation of perineural invasion of gastric carcinoma into the 7th edition tumor-node-metastasis staging system. Tumour Biol 35: 9429–9436., DOI 10.1007/s13277-014-2258-5 Selcukbiricik F, Tural D, Buyukunal E, Serdengecti S, 2012, Perineural invasion independent prognostic factors in patients with gastric cancer undergoing curative resection. Asian Pac J Cancer Prev 13:3149–3152 Zhou ZH, Xu GF, Zhang WJ, Zhao HB, Wu YY, 2014, Reevaluating significance of perineural invasion in gastric cancer based on double immunohistochemical staining. Arch Pathol lab med 138:229–234., DOI 10.5858/arpa.2012-0669-OA Tianhang L, Guoen F, Jianwei B, Liye M, 2008, The effect of perineural invasion on overall survival in patients with gastric carcinoma. J Gastrointest Surg 12:1263–1267., DOI 10.1007/s11605- 008-0529-4 Scartozzi M, Galizia E, Verdecchia L, Berardi R, Graziano F, Catalano V, Giordani P, Mari D, Silva RR, Marmorale C, Zingaretti C, Cascinu S, 2006, Lymphatic, blood vessel and perineural invasion identifies early-stage high-risk radically resected gastric cancer patients. Br J Cancer 95:445–449 KimDH, Kim SM, Hyun JK, ChoiMG, Noh JH, Sohn TS, Bae JM, Kim S, 2013, Changes in postoperative recurrence and prognostic risk factors for patients with gastric cancer who underwent curative gastric resection during different time periods. Ann Surg Oncol 20: 2317–2327., DOI 10.1245/s10434-012-2700-0 Marrelli D, Polom K, Pascale V, Vindigni C, Piagnerelli R, De Franco L, Ferrara F, Roviello G, Garosi L, Petrioli R, Roviello F, 2016, Strong prognostic value of microsatellite instability in intestinal type non-cardia gastric cancer. Ann Surg Oncol 23:943–950., DOI 10.1245/s10434-015-4931-3 Roviello F,Marrelli D, Vindigni C, De Stefano A, Spina D, Pinto E, 1999, P53 accumulation is a prognostic factor in intestinal-type gastric carcinoma but not in the diffuse type. Ann Surg Oncol 6: 739–745., DOI 10.1007/s10434-999-0739-3 Marrelli D, Polom K, de Manzoni G, Morgagni P, Baiocchi GL, Roviello F, 2015, Multimodal treatment of gastric cancer in the west: where are we going? World J Gastroenterol 21:7954–7969., DOI 10.3748/wjg.v21.i26.7954 Cristescu R, Lee J, NebozhynM, KimKM, Ting JC,Wong SS, Liu J, Yue YG, Wang J, Yu K, Ye XS, Do IG, Liu S, Gong L, Fu J, Jin JG, ChoiMG, Sohn TS, Lee JH, Bae JM, Kim ST, Park SH, Sohn I, Jung SH, Tan P, Chen R, Hardwick J, Kang WK, Ayers M, Hongyue D, Reinhard C, Loboda A, Kim S, Aggarwal A, 2015, Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat med 21:449–456., DOI 10.1038/ nm.3850 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 393 EP 400 DI 10.1007/s12253-017-0257-8 PG 8 ER PT J AU Gorka, E Fabo, D Gezsi, A Czirbesz, K Fedorcsak, I Liszkay, G AF Gorka, Eszter Fabo, Daniel Gezsi, Andras Czirbesz, Kata Fedorcsak, Imre Liszkay, Gabriella TI Dabrafenib Therapy in 30 Patients with Melanoma Metastatic to the Brain: a Single-centre Controlled Retrospective Study in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Dabrafenib; Melanoma; Brain metastases; Targeted therapy; BRAF ID Dabrafenib; Melanoma; Brain metastases; Targeted therapy; BRAF AB Dabrafenib is a potent BRAF inhibitor, which showed intracranial tumor activity. The purpose of our retrospective analysis was to evaluate the efficacy of dabrafenib for patients with melanoma brain metastasis (BM). We studied 30 BRAF mutant melanoma patients with BM, who received dabrafenib after local control of the brain between 2014 and 2017. Eastern Cooperative Oncology Group Performance Status (ECOG) was 0–2. The control arm consisted of 204 melanoma patients from our institutional melanoma database with BM and ECOG 0–2 treated with local therapies and/or chemotherapy, between 2003 and 2015. We found the intracranial disease control rate (DCR) was 83% including four (13%) complete remissions (CR), nine (30%) partial remissions (PR) and twelve (40%) stable diseases (SD) in contrast to five (17%) progressive diseases (PD).With a median follow-up of 14months, median progression-free survival (PFS) and overall survival (OS) were 5.5 months, and 8.8 months, respectively. If calculated from BM onset, the OS turned to be 11.8 months on the dabrafenib arm, while it was only 6.0 months in the control arm (HR = 0.45, p = 0.0014). Higher risk of progression was observed with increasing ECOG (HR =4.06, p = 0.00027) and if more than 2 extracranial organs were involved (HR = 3.4, p = 0.0077). Elevated lactate dehydrogenase (LDH) was non-significantly associated with worse clinical outcome. Remarkable intracranial activity of dabrafenib in real practice was confirmed by our analysis. C1 [Gorka, Eszter] National Institute of Oncology, Department of Dermatology, Rath Gyorgy str. 7-9, H-1122 Budapest, Hungary. [Fabo, Daniel] National Institute of Clinical Neurosciences, Amerikai str. 57, H-1145 Budapest, Hungary. [Gezsi, Andras] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad sq. 4, H-1084 Budapest, Hungary. [Czirbesz, Kata] National Institute of Oncology, Department of Dermatology, Rath Gyorgy str. 7-9, H-1122 Budapest, Hungary. [Fedorcsak, Imre] National Institute of Clinical Neurosciences, Amerikai str. 57, H-1145 Budapest, Hungary. [Liszkay, Gabriella] National Institute of Oncology, Department of Dermatology, Rath Gyorgy str. 7-9, H-1122 Budapest, Hungary. RP Gorka, E (reprint author), National Institute of Oncology, Department of Dermatology, H-1122 Budapest, Hungary. EM goreszter@yahoo.com CR Falchook GS, Long GV, Kurzrock R et al, 2012, Dabrafenib in patients with melanoma, untreated brain metastases, and other solid tumours: a phase 1 dose escalation trial. Lancet 379(9829):1893–1901 Long GV, Trefzer U, DaviesMAet al, 2012, Dabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain, BREAK-MB): a multicentre, open-label, phase 2 trial. Lancet Oncol 13(11):1087–1095 Dummer R, Goldinger SM, Turtschi CP et al, 2014, Vemurafenib in patients with BRAF(V600, mutation-positive melanoma with symptomatic brain metastases: final results of an open-label pilot study. Eur J Cancer 50(3):611–621 Colombino M, Capone M, Lissia A et al, 2012, Vemurafenib in patients with BRAF/NRAS mutation frequencies among primary tumours and metastases in patients with melanoma. J Clin Oncol 30(20):2522–2529 Jakob JA, Bassett RL Jr, Ng CS et al, 2012, NRAS mutation status is an independent prognostic factor in metastaticmelanoma. Cancer 118(16):4014–4023 Mittapalli RK, Vaidhyanathan S, Dudek AZ et al, 2013, Mechanisms limiting distribution of the threonine-protein kinase B-RaF(V600E, inhibitor dabrafenib to the brain: implications for the treatment of melanoma brain metastases. J Pharmacol Exp Ther 344(3):655–664 Davies MA, Liu P, McIntyre S et al, 2011, Prognostic factors for survival in melanoma patients with brain metastases. Cancer 117(8):1687–1696 Fife KM, Colman MH, Stevens GN et al, 2004, Determinants of outcome in melanoma patients with cerebral metastases. J Clin Oncol 22(7):1293–1300 Sampson JH, Carter JH Jr, Friedman AH et al, 1998, Demographics, prognosis, and therapy in 702 patients with brain metastases from malignant melanoma. J Neurosurg 88(1):11–20 Dzienis MR, Atkinson V., 2013, Response rate to vemurafenib in BRAF-positive melanoma brain metastases. J Clin Oncol 31, abstract 9081 Harding JJ, Catalanotti F, Munhoz RR et al, 2015, A retrospective evaluation of Vemurafenib as treatment for BRAF-mutant melanoma brain metastases. Oncologist 20(7):789–797 Azer MW, Menzies AM, Haydu LE et al, 2014, Patterns of response and progression in patients with BRAF-mutant melanoma metastatic to the brain who were treated with dabrafenib. Cancer 120(4):530–536 Cocorocchio E, Gandini S, Alfieri S et al, 2016, Dabrafenib in metastatic melanoma: a monocentric 'real life' experience. Ecancermedicalscience 10:624 Lau DK, Andrews MC, Turner N et al, 2014, A single-centre experience of patients with metastatic melanoma enrolled in a dabrafenib named patient programme. Mel res 24(2):144–149 Hauschild A, Grob JJ, Demidov LV et al, 2012, Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, openlabel, phase 3 randomised controlled trial. Lancet 380(9839): 358–365 Long GV, Stroyakovskiy D, Gogas H et al, 2014, Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J med 371(20):1877–1888 Schadendorf D, Amonkar MM, Stroyakovskiy D et al, 2015, Health-related quality of life impact in a randomised phase III study of the combination of dabrafenib and trametinib versus dabrafenib monotherapy in patients with BRAF V600 metastatic melanoma. Eur J Cancer 51(7):833–840 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 401 EP 406 DI 10.1007/s12253-017-0256-9 PG 6 ER PT J AU da Silva, NG Filoni, TL Salvadori, CM Favero Salvadori, MD AF da Silva, Nicioli Glenda Filoni, Toshio Leandro Salvadori, Cecilia Maria Favero Salvadori, Maria Daisy TI Gemcitabine/Cisplatin Treatment Induces Concomitant SERTAD1, CDKN2B and GADD45A Modulation and Cellular Changes in Bladder Cancer Cells Regardless of the Site of TP53 Mutation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; Cell morphology; Chemotherapy; Cisplatin; Gene expression; Gemcitabine ID Bladder cancer; Cell morphology; Chemotherapy; Cisplatin; Gene expression; Gemcitabine AB Simultaneous use of cisplatin (CIS) and gemcitabine (GEN) for treating bladder cancer has increased because of their complementary effects. However, the molecular mechanisms underlying the activities of these two antineoplastic drugs are not fully known. Here, molecular biology techniques and microscopy were used to investigate transcriptomic and morphological changes in low and highgrade urinary bladder transitional carcinoma cell lines [RT4 - wild type TP53; 5637 - two TP53 mutations, one in codon 72 (Arg-Pro) and other in codon 280 (Arg-Thr) and T24 - inframe deletion of tyrosine 126 in the TP53 allele] simultaneously treated with CIS/GEN. Gene expression profile was evaluated by PCR arrays; cell morphology by scanning and transmission electron microscopy, and apoptosis was analyzed using fluorescent dye. Results showed concomitantly upregulation of CDKN2B (G1/S transition), GADD45A (DNA repair and apoptosis) and SERTAD1 (regulation of transcription) gene, increased number of nuclear chamfers and apoptotic cells, and reduced number of microfilaments, organelles and in the size of the nucleus in 5637 and T24 cells after simultaneous treatment with CIS/GEN. In conclusion, independently of the TP53 mutation status and tumor grade, CIS/ GEN induced gene modulation accompanied by changes in cell morphologies, which confirm the antiproliferative activity of the treatment protocol. These findings help to understand the pathways modulated by these antineoplastic agents and may provide insights for anti-cancer chemotherapy. C1 [da Silva, Nicioli Glenda] UFOP – Federal University of Ouro Preto, School of PharmacyOuro Preto, MG, Brazil. [Filoni, Toshio Leandro] USP – University of Sao Paulo, Institute of PhysicsSao Paulo, SP, Brazil. [Salvadori, Cecilia Maria] USP – University of Sao Paulo, Institute of PhysicsSao Paulo, SP, Brazil. [Favero Salvadori, Maria Daisy] UNESP – State University of Sao Paulo, Medical SchoolBotucatu, SP, Brazil. RP da Silva, NG (reprint author), UFOP – Federal University of Ouro Preto, School of Pharmacy, Ouro Preto, Brazil. EM nicioli@ef.ufop.br CR von der Maase H, Sengelov L, Roberts JT, Ricci S, Dogliotti L, Oliver T et al, 2005, Long-term survival results of a randomized trial comparing gemcitabine plus cisplatin, with methotrexate, vinblastine, doxorubicin, plus cisplatin in patients with bladder cancer. J Clin Oncol 23:4602–4608., DOI 10.1200/JCO.2005.07.757 Yuh BE, Ruel N, Wilson TG, Vogelzang N, Pal SK, 2013, Pooled analysis of clinical outcomes with neoadjuvant cisplatin and gemcitabine chemotherapy for muscle invasive bladder cancer. J Urol 189:1682–1686., DOI 10.1016/j.juro.2012.10.120 Wang D, Lippard SJ, 2005, Cellular processing of platinum anticancer drugs. Nat rev Drug Discov 4:307–319., DOI 10.1038/nrd1691 Toschi L, Finocchiaro G, Bartolini S, Gioia V, Cappuzzo F, 2005, Role of gemcitabine in cancer therapy. Future Oncol 1:7–17., DOI 10.1517/14796694.1.1.7 Coppee J-Y, 2008, Do DNA microarrays have their future behind them? Microbes Infect 10:1067–1071., DOI 10.1016/j.micinf.2008. 07.003 Rentsch CA, Stenner F, Ruiz C, Bubendorf L, 2015, Three genes to predict response to chemotherapy for bladder cancer: individualised cancer care at the doorstep. Eur Urol 68:968–969., DOI 10.1016/j. eururo.2015.07.050 Da Silva GN, de Camargo EA, Salvadori DM, 2012, Toxicogenomic activity of gemcitabine in two TP53-mutated bladder cancer cell lines: special focus on cell cycle-related genes. Mol Biol rep 39(12):10373–10382., DOI 10.1007/s11033-012-1916-1 Savio AL, da Silva GN, Salvadori DM(2015, Inhibition of bladder cancer cell proliferation by allyl isothiocyanate, mustard essential oil). Mutat res 771:29–35., DOI 10.1016/j.mrfmmm.2014.11.004 Goncalves EM, Ventura CA, Yano T, Rodrigues Macedo ML, Genari SC, 2006, Morphological and growth alterations in Vero cells transformed by cisplatin. Cell Biol Int 30:485–494., DOI 10. 1016/j.cellbi.2005.12.007 Chen NG, Chen KT, Lu CC, Lan YH, Lai CH, Chung YT et al, 2010, Allyl isothiocyanate triggers G2/M phase arrest and apoptosis in human brain malignant glioma GBM 8401 cells through a mitochondria-dependent pathway. Oncol rep 24:449–455 Nishiyama H, Watanabe J, Ogawa O, 2008, p53 and chemosensitivity in bladder cancer. Int J Clin Oncol 13:282–286., DOI 10.1007/s10147-008-0815-x Da Silva GN, de Castro Marcondes JP, de Camargo EA, da Silva Passos Junior GA, Sakamoto-Hojo ET, Salvadori DM, 2010, Cell cycle arrest and apoptosis in TP53 subtypes of bladder carcinoma cell lines treated with cisplatin and gemcitabine. Exp Biol med, Maywood, 235:814–824., DOI 10.1258/ebm.2010.009322 Camargo EA, da Silva GN, Gobette CP, Marcondes JP, Salvadori DM, 2013, No relationship between the amount of DNA damage and the level of hMLH1 and RASSF1A gene expression in bladder cancer cells treated with cisplatin and gemcitabine. Asian Pac J Cancer Prev 4:5941–5948., DOI 10.7314/APJCP.2013.14.10.5941 Rovida AFS, Endo KM, POlli AD, Bulla LMC, Scudeler EL, Abreu JAS et al, 2015, Use of neem, Azadirachta indica a Juss, oil in the control of Musca domestica L.(Diptera:Muscidae, in poultry breeding farms. J World's Poult res 5:73–83 Liu Y, Kwiatkowski DJ, 2015, Combined CDKN1A/TP53 mutation in bladder cancer is a therapeutic target. Mol Cancer Ther 14: 174–182., DOI 10.1158/1535-7163.MCT-14-0622-T Vaezy S, Zderic V, 2009, Image-guided therapy systems. Artech House INC, Washington DC Rajcevic U, Petersen K, Knol JC, Loos M, Bougnaud S, Klychnikov O et al, 2009, iTRAQ-based proteomics profiling reveals increased metabolic activity and cellular cross-talk in angiogenic compared with invasive glioblastoma phenotype. Mol Cell Proteomics 8:2595–2512., DOI 10.1074/mcp.M900124-MCP200 Suresh S, 2007, Biomechanics and biophysics of cancer cells. Acta Biomater 3:413–438., DOI 10.1016/j.actbio.2007.04.002 Wang X, Simpson ER, Brown KA, 2015, P53: protection against tumor growth beyond effects on cell cycle and apoptosis. Cancer res 75:5001–5007., DOI 10.1158/0008-5472.CAN-15-0563 Wang X, Wong SC, Pan J, Tsao SW, Fung KH, Kwong DL et al, 1998, Evidence of cisplatin-induced senescent-like growth arrest in nasopharyngeal carcinoma cells. Cancer res 58:5019–5022 Lu TX, Young KH, Xu W, Li JY, 2016, TP53 dysfunction in diffuse large B-cell lymphoma. Crit rev Oncol Hematol 97:47–55., DOI 10.1016/j.critrevonc.2015.08.006 Fechner G, Perabo FG, Schmidt DH, Haase L, Ludwig E, Schueller H et al, 2003, Preclinical evaluation of a radiosensitizing effect of gemcitabine in p53 mutant and p53 wild type bladder cancer cells. Urology 61:468–473., DOI 10.1016/S0090-4295(02)02156-8 Missiaglia E, Donadelli M, Palmieri M, Crnogorac-Jurcevic T, Scarpa A, Lemoine NR, 2005, Growth delay of human pancreatic cancer cells bymethylase inhibitor 5-aza-2. Oncogene 24:199–211., DOI 10.1038/sj.onc.1208018 Chim CS, Kwong YL, 2006, Adverse prognostic impact of CDKN2B hyper-methylation in acute promyelocytic leukemia. Leuk Lymphoma 47:815–825., DOI 10.1080/10428190500513827 Gupta S, Takhar PP, Degenkolbe R, Koh CH, Zimmermann H, Yang CM et al, 2003, The human papillomavirus type 11 and 16 E6 proteins modulate the cell-cycle regulator and transcription cofactor TRIP-Br1. Virology 317:155–164., DOI 10.1016/j.virol.2003.08.008 Darwish H, Cho JM, Loignon M, Alaoui-Jamali MA, 2007, Overexpression of SERTAD3, a putative oncogene located within the 19q13 amplicon, induces E2F activity and promotes tumor growth. Oncogene 26:4319–4328., DOI 10.1038/sj.onc.1210195 L'Esperance S, Bachvarova M, Tetu B, Mes-Masson AM, Bachvarov D, 2008, Global gene expression analysis of early response to chemotherapy treatment in ovarian cancer spheroids. BMC Genomics 9:99., DOI 10.1186/1471-2164-9-99 da Silva GN, de Camargo EA, Savio AL, Salvadori DM, 2014, MRE11A and SKP2 genes are associated with the increased cytotoxicity induced by the synergistic effects of cisplatin and gemcitabine in bladder cancer cells. Mol Biol rep 41:4613–4621., DOI 10.1007/s11033-014-3332-1 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 407 EP 417 DI 10.1007/s12253-017-0255-x PG 11 ER PT J AU Zhu, FF Zheng, FY Wang, HO Zheng, JJ Zhang, Q AF Zhu, Fang-Fang Zheng, Fei-Yun Wang, Hai-Ou Zheng, Jing-Jie Zhang, Qian TI Downregulation of lncRNA TUBA4B is Associated with Poor Prognosis for Epithelial Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ovarian cancer; lncRNA; Overall survival; Proliferation ID Ovarian cancer; lncRNA; Overall survival; Proliferation AB A host of studies have revealed that long noncoding RNAs (lncRNAs) are critically involved in the development and progression of epithelial ovarian cancer. LncRNA TUBA4B is recently identified to be a critical mediator in nonsmall cell lung cancer. However, the clinical roles and biological functions of lncRNA TUBA4B in epithelial ovarian cancer have yet to be fully clarified. The present study was conducted to explore the expression of lncRNA TUBA4B in human epithelial ovarian cancer tissues and potential roles of lncRNA TUBA4B in ovarian cancer cells. The matched epithelial ovarian cancer specimens and adjacent normal tissues were employed to detect the expression of lncRNA TUBA4B. The prognostic value of lncRNA TUBA4B for tumor progression and survival rate was investigated. The effects of lncRNA TUBA4B on ovarian cancer cell proliferation and migration were also explored. The expression of lncRNA TUBA4B was significantly decreased in epithelial ovarian cancer tissue specimens. The low lncRNA TUBA4B level was closely related with pathological grade, FIGO stage and lymph node metastases, and serum CA125 level. Enforced expression of lncRNA TUBA4B obviously reduced the proliferation of SKOV3 cells, and attenuated the activation of ERK and Akt signaling pathways. Our data demonstrate for the first time that lower lncRNA TUBA4B may be a novel independent prognostic biomarker for overall survival of epithelial ovarian cancer. Overexpression of lncRNA TUBA4B inhibits the proliferation of ovarian cancer cells. LncRNA TUBA4B may be an important target for therapeutic intervention in ovarian cancer. C1 [Zhu, Fang-Fang] the First Affiliated Hospital of Wenzhou Medical University, Department of Gynaecology, 1 Fuxue Lane, Lucheng District, 325000 Wenzhou, Zhejiang, China. [Zheng, Fei-Yun] the First Affiliated Hospital of Wenzhou Medical University, Department of Gynaecology, 1 Fuxue Lane, Lucheng District, 325000 Wenzhou, Zhejiang, China. [Wang, Hai-Ou] the First Affiliated Hospital of Wenzhou Medical University, Department of Obstetrics and GynecologWenzhou, Zhejiang, China. [Zheng, Jing-Jie] the First Affiliated Hospital of Wenzhou Medical University, Department of Gynaecology, 1 Fuxue Lane, Lucheng District, 325000 Wenzhou, Zhejiang, China. [Zhang, Qian] the First Affiliated Hospital of Wenzhou Medical University, Department of Gynaecology, 1 Fuxue Lane, Lucheng District, 325000 Wenzhou, Zhejiang, China. RP Zhang, Q (reprint author), the First Affiliated Hospital of Wenzhou Medical University, Department of Gynaecology, 325000 Wenzhou, China. EM qianzhangwzzj@yeah.net CR Ross JS, Ali SM,Wang K, Palmer G, Yelensky R, Lipson D,Miller VA, Zajchowski D, Shawver LK, Stephens PJ, 2013, Comprehensive genomic profiling of epithelial ovarian cancer by next generation sequencing-based diagnostic assay reveals new routes to targeted therapies. Gynecol Oncol 130(3):554–559., DOI 10.1016/j.ygyno.2013.06.019 Chudecka-Glaz AM, 2015, ROMA, an algorithm for ovarian cancer. Clinica chimica acta; international journal of clinical chemistry 440:143–151., DOI 10.1016/j.cca.2014.11.015 Su Y, Wu H, Pavlosky A, Zou LL, Deng X, Zhang ZX, Jevnikar AM, 2016, Regulatory non-coding RNA: new instruments in the orchestration of cell death. Cell Death Dis 7(8):e2333., DOI 10.1038/ cddis.2016.210 Li F, Hu CP, 2015, Long non-coding RNA Urothelial carcinoma associated 1, UCA1): insight into its role in human diseases. Crit Rev Eukaryot Gene Expr 25(3):191–197 Chen J, Hu L, Wang J, Zhang F, Chen J, Xu G, Wang Y, Pan Q, 2017, Low expression LncRNA TUBA4B is a poor predictor of prognosis and regulates cell proliferation in non-small cell lung cancer. Pathology Oncology Research: POR 23(2):265-270.doi: 10.1007/s12253-016-0089-y Sun HJ, Liu TY, Zhang F, Xiong XQ, Wang JJ, Chen Q, Li YH, Kang YM, Zhou YB, Han Y, Gao XY, Zhu GQ, 2015, Salusin-beta contributes to vascular remodeling associated with hypertension via promoting vascular smooth muscle cell proliferation and vascular fibrosis. BiochimBiophys Acta 1852(9):1709–1718., DOI 10.1016/j. bbadis.2015.05.008 Sun HJ, Zhao MX, Ren XS, Liu TY, Chen Q, Li YH, Kang YM, Wang JJ, Zhu GQ, 2016, Salusin-beta promotes vascular smooth muscle cell migration and intimal hyperplasia after vascular injury via ROS/NFkappaB/MMP-9 pathway. Antioxid Redox Signal 24(18):1045–1057., DOI 10.1089/ars.2015.6475 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90., DOI 10. 3322/caac.20107 Zhang M, Du X, 2016, Noncoding RNAs in gastric cancer: research progress and prospects. World J Gastroenterol 22(29): 6610–6618., DOI 10.3748/wjg.v22.i29.6610 Yang Y, Jiang Y,Wan Y, Zhang L, Qiu J, Zhou S, ChengW(2016, UCA1 functions as a competing endogenous RNA to suppress epithelial ovarian cancer metastasis. Tumour Biology : the Journal of the International Society for Oncodevelopmental Biology and Medicine 37(8):10633–10641., DOI 10.1007/s13277-016-4917-1 Gao Y, Meng H, Liu S, Hu J, Zhang Y, Jiao T, Liu Y, Ou J,Wang D, Yao L, Liu S, Hui N, 2015, LncRNA-HOST2 regulates cell biological behaviors in epithelial ovarian cancer through a mechanism involving microRNA let-7b. Hum Mol Genet 24(3):841–852., DOI 10.1093/hmg/ddu502 Qiu JJ, Lin YY, Ye LC, Ding JX, FengWW, Jin HY, ZhangY, Li Q, Hua KQ, 2014)Overexpression of long non-codingRNA HOTAIR predicts poor patient prognosis and promotes tumor metastasis in epithelial ovarian cancer. Gynecol Oncol 134(1):121–128., DOI 10. 1016/j.ygyno.2014.03.556 Wang L, Wang J, Fang J, Zhou H, Liu X, Su SB, 2015, High glucose induces and activates toll-like receptor 4 in endothelial cells of diabetic retinopathy. Diabetol Metab Syndr 7:89., DOI 10.1186/ s13098-015-0086-4 Hu X, Sun S, 2015, RAD51 Gene 135G/C polymorphism and ovarian cancer risk: a meta-analysis. Int J Clin Exp Med 8(12): 22365–22370 Liu X, Li G, 2015, MicroRNA-133b inhibits proliferation and invasion of ovarian cancer cells through Akt and Erk1/2 inactivation by targeting epidermal growth factor receptor. Int J Clin Exp Pathol 8(9):10605–10614 Huang WL, Li Z, Lin TY, Wang SW, Wu FJ, Luo CW, 2016, Thyrostimulin-TSHR signaling promotes the proliferation of NIH: OVCAR-3 ovarian cancer cells via trans-regulation of the EGFR pathway. Sci Report 6:27471., DOI 10.1038/srep27471 Dong YL, Kabir SM, Lee ES, Son DS, 2013, CXCR2-driven ovarian cancer progression involves upregulation of proinflammatory chemokines by potentiating NF-kappaB activation via EGFRtransactivated Akt signaling. PLoS One 8(12):e83789., DOI 10. 1371/journal.pone.0083789 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 419 EP 425 DI 10.1007/s12253-017-0258-7 PG 7 ER PT J AU Karbyshev, SM Grigoryeva, SE Volkomorov, VV Kremmer, E Huber, A Mitrofanova, VI Kaigorodova, VE Zavyalova, VM Kzhyshkowska, GJ Cherdyntseva, VN Choynzonov, LE AF Karbyshev, S Mikhail Grigoryeva, S Evgeniya Volkomorov, V Viktor Kremmer, Elisabeth Huber, Alexander Mitrofanova, V Irina Kaigorodova, V Evgeniya Zavyalova, V Marina Kzhyshkowska, G Julia Cherdyntseva, V Nadezda Choynzonov, L Evgeny TI Development of Novel Monoclonal Antibodies for Evaluation of Transmembrane Prostate Androgen-Induced Protein 1 (TMEPAI) Expression Patterns in Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Monoclonal antibody; Biomarker; Immunohistochemistry; Gastric cancer; PMEPA1; TMEPAI ID Monoclonal antibody; Biomarker; Immunohistochemistry; Gastric cancer; PMEPA1; TMEPAI AB Transmembrane prostate androgen-induced protein 1 (TMEPAI) is a single-span membrane protein, functionally involved in transforming growth factor beta signaling pathway. The particular protein presented in cells in three isoforms, which differs in the length of the soluble N-terminal extracellular domain, making it challenging for the immunochemical recognition. By using quantitative real-time polymerase chain reaction, we identified significant upregulation of PMEPA1 gene expression in malignant tissues of patients with gastric adenocarcinoma. The main part of commercially available anti-TMEPAI antibodies are having polyclonal nature or not suitable for immunocytochemical localization of target protein in tissue specimens. Hence, we decide to generate a set of novel rat monoclonal antibodies (mAb) directed against conservative C-terminal cytoplasmic epitope. Immunoblotting analysis showed that monoclonal antibodies, 2E1, 6C6, and 10A7 were able to recognize specifically target protein in transiently transfected HEK293T and CHO-K1 cells. Especially established mAb, named 10A7, showed the excellent binding ability to target protein in immunohistochemistry. By using developed antibodies, we observed pronounced expression of TMEPAI in normal gastric epithelial cells while tumor cells from gastric adenomas, and adenocarcinoma samples were mostly negative for target protein expression. Also, we found that gastric epithelium cells lose the TMEPAI expression concurrently with severe dysplasia progression, which probably caused by a mechanism involving specific microRNA. C1 [Karbyshev, S Mikhail] Russian Academy of Sciences, Cancer Research Institute, Tomsk National Research Medical Center, Department of Molecular Oncology and Immunology, Kooperativny Str. 5, 634050 Tomsk, Russian Federation. [Grigoryeva, S Evgeniya] Russian Academy of Sciences, Cancer Research Institute, Tomsk National Research Medical Center, Department of Molecular Oncology and Immunology, Kooperativny Str. 5, 634050 Tomsk, Russian Federation. [Volkomorov, V Viktor] Russian Academy of Sciences, Cancer Research Institute, Tomsk National Research Medical Center, Department of Molecular Oncology and Immunology, Kooperativny Str. 5, 634050 Tomsk, Russian Federation. [Kremmer, Elisabeth] Helmholtz Zentrum Munchen, German Research Center for Environmental Health, Institute of Molecular Immunology, Marchionini str. 25, 81377 Munich, Germany. [Huber, Alexander] Helmholtz Zentrum Munchen, German Research Center for Environmental Health, Institute of Molecular Immunology, Marchionini str. 25, 81377 Munich, Germany. [Mitrofanova, V Irina] Russian Academy of Sciences, Cancer Research Institute, Tomsk National Research Medical Center, Department of Molecular Oncology and Immunology, Kooperativny Str. 5, 634050 Tomsk, Russian Federation. [Kaigorodova, V Evgeniya] Russian Academy of Sciences, Cancer Research Institute, Tomsk National Research Medical Center, Department of Pathological Anatomy and Cytology, Kooperativny Str. 5, 634050 Tomsk, Russian Federation. [Zavyalova, V Marina] Tomsk State University, Laboratory for Translational Cellular and Molecular Biomedicine, 36 Lenin Ave, 634050 Tomsk, Russian Federation. [Kzhyshkowska, G Julia] Tomsk State University, Laboratory for Translational Cellular and Molecular Biomedicine, 36 Lenin Ave, 634050 Tomsk, Russian Federation. [Cherdyntseva, V Nadezda] Russian Academy of Sciences, Cancer Research Institute, Tomsk National Research Medical Center, Department of Molecular Oncology and Immunology, Kooperativny Str. 5, 634050 Tomsk, Russian Federation. [Choynzonov, L Evgeny] Russian Academy of Sciences, Cancer Research Institute, Tomsk National Research Medical Center, Department of Molecular Oncology and Immunology, Kooperativny Str. 5, 634050 Tomsk, Russian Federation. RP Grigoryeva, SE (reprint author), Russian Academy of Sciences, Cancer Research Institute, Tomsk National Research Medical Center, Department of Molecular Oncology and Immunology, 634050 Tomsk, Russian Federation. EM grigorieva@oncology.tomsk.ru CR Stone L, 2015, Prostate cancer: TGF-beta signalling regulator PMEPA1 halts metastases tobone. Nat Rev Urol 12(7):362 Xu LL et al, 2000, A novel androgen-regulated gene, PMEPA1, located on chromosome20q13 exhibits high level expression in prostate. Genomics 66(3):257–263 Watanabe Y et al, 2010, TMEPAI, a transmembrane TGF-betainducible protein, sequestersSmad proteins from active participation in TGF-beta signaling. Mol Cell 37(1):123–134 Vo Nguyen TT et al, 2014, TMEPAI/PMEPA1 enhances tumorigenic activities in lungcancer cells. Cancer Sci 105(3):334–341 Azami S et al, 2015, Cooperative induction of transmembrane prostate androgen inducedprotein TMEPAI/PMEPA1 by transforming growth factor-beta and epidermal growth factorsignaling. Biochem Biophys Res Commun 456(2):580–585 Hu Yet al., 2013, TMEPAI regulates EMT in lung cancer cells by modulating the ROS and IRS-1 signaling pathways. Carcinogenesis 34(8):1764–1772 Ellenrieder V, 2008, TGFbeta regulated gene expression by Smads and Sp1/KLF-liketranscription factors in cancer. Anticancer Res 28(3A):1531–1539 Gratchev A et al, 2008, Activation of a TGF-beta-specificmultistep gene expression programin mature macrophages requires glucocorticoid-mediated surface expression of TGF-betareceptor II. J Immunol 180(10):6553–6565 Brunschwig EB et al, 2003, PMEPA1, a transforming growth factor-beta-induced marker ofterminal colonocyte differentiation whose expression is maintained in primary and metastaticcolon cancer. Cancer Res 63(7):1568–1575 Nakano N et al, 2014, C18 ORF1, a novel negative regulator of transforming growth factorbeta signaling. J Biol Chem 289(18): 12680–12692 Fournier PG et al, 2015, The TGF-beta signaling regulator PMEPA1 suppresses prostate cancer metastases to bone. Cancer Cell 27(6):809–821 Nagaraj NS, Datta PK, 2010, Targeting the transforming growth factor-beta signalingpathway in human cancer. Expert Opin Investig Drugs 19(1):77–91 Li H et al, 2015, Silencing of PMEPA1 accelerates the growth of prostate cancer cellsthrough AR, NEDD4 and PTEN. Oncotarget 6(17):15137–15149 Singha PK et al, 2014, TGF-beta induced TMEPAI/PMEPA1 inhibits canonical Smadsignaling through R-Smad sequestration and promotes non-canonical PI3K/Akt signaling byreducing PTEN in triple negative breast cancer. Genes Cancer 5(9–10):320–336 Saadi A et al, 2010, Stromal genes discriminate preinvasive from invasive disease, predictoutcome, and highlight inflammatory pathways in digestive cancers. Proc Natl Acad Sci U S A 107(5):2177– 2182 Hirokawa YS et al, 2007, High level expression of STAG1/ PMEPA1 in an androgenindependentprostate cancer PC3 subclone. Cell Mol Biol Lett 12(3):370–377 Sharad S et al, 2014, Methylation of the PMEPA1 gene, a negative regulator of the androgenreceptor in prostate cancer. Epigenetics 2014.9(6):918–927 Volkomorov V et al, 2013, Search for potential gastric cancer markers using miRNAdatabases and gene expression analysis. Exp Oncol 35(1):2–7 Grigoryeva ES et al, 2013, Expression of Cyclophilin a in gastric adenocarcinoma Patientsand its inverse association with local relapses and distant metastasis. Pathol Oncol Res 20(2):467–473 Rae FK et al, 2001, Characterization of a novel gene, STAG1/ PMEPA1, upregulated inrenal cell carcinoma and other solid tumors. Mol Carcinog 32(1):44–53 Rajkumar T et al, 2010, Identification and validation of genes involved in gastrictumorigenesis. Cancer Cell Int 10:45 Rottach A et al, 2008, Generation and characterization of a rat monoclonal antibody specificfor PCNA. Hybridoma, Larchmt, 27(2):91–98 Shi SR, Liu C, Taylor CR, 2007, Standardization of immunohistochemistry forformalin-fixed, paraffin-embedded tissue sections based on the antigen-retrieval technique: fromexperiments to hypothesis. J Histochem Cytochem 55(2):105–109 HuangMet al, 2014, Generation of a monoclonal antibody specific to a new candidate tumorsuppressor, cell adhesion molecule 2. Tumour Biol 35(8):7415–7422 Harvey KF et al, 2002, N4WBP5, a potential target for ubiquitination by the Nedd4 familyof proteins, is a novel Golgiassociated protein. J Biol Chem 277(11):9307–9317 Fock KM, 2014, Review article: the epidemiology and prevention of gastric cancer. Aliment Pharmacol Ther 40(3):250–260 Duraes C et al, 2014, Biomarkers for gastric cancer: prognostic, predictive or targets oftherapy? Virchows Arch 464(3):367–378 Hofler H, Becker KF, 2003, Molecular mechanisms of carcinogenesis in gastric cancer. Recent Results Cancer Res 162:65–72 Fu H et al, 2009, TGF-beta promotes invasion and metastasis of gastric cancer cells byincreasing fascin 1 expression via ERK and JNK signal pathways. Acta Biochim Biophys Sin Shanghai 41(8): 648–656 Shinto O et al, 2010, Phosphorylated smad 2 in advanced stage gastric carcinoma. BMC Cancer 10:652 Kubiczkova L et al, 2012, TGF-beta - an excellent servant but a bad master. J Transl Med 10:183 Liu R et al, 2011, PMEPA1 promotes androgen receptor-negative prostate cell proliferationthrough suppressing the Smad 3/4-c-Mycp 21 Cip1 signaling pathway. J Pathol 223(5):683–694 Bjorling E, Uhlen M, 2008, Antibodypedia, a portal for sharing antibody and antigenvalidation data. Mol Cell Proteomics 7(10): 2028–2037 Bordeaux J et al, 2010, Antibody validation. BioTechniques 48(3): 197–209 Mischak H et al., 2010, Recommendations for biomarker identification and qualification inclinical proteomics. Sci Transl Med 2(46):46ps42 Uhlen M et al, 2015, Proteomics. Tissue-based map of the human proteome. Science 347(6220):1260419 Huttenhain R et al, 2012, Reproducible quantification of cancerassociated proteins in bodyfluids using targeted proteomics. Sci Transl Med 4(142):142ra94 Flejou JF, 2005, Barrett’s oesophagus: frommetaplasia to dysplasia and cancer. Gut 54(Suppl 1):i6–12 Jansson MD, Lund AH, 2012, MicroRNA and cancer. Mol Oncol 6(6):590–610 Wu WK et al, 2010, MicroRNA dysregulation in gastric cancer: a new player enters the game. Oncogene 29(43):5761–5771 Zhou H et al, 2013, TGF-beta 1 alters microRNA profile in human gastric cancer cells. Chin J Cancer Res 25(1):102–111 Grillari J, Hackl M, Grillari-Voglauer R, 2010, miR-17-92 cluster: ups and downs in cancer and aging. Biogerontology 11(4):501–506 Li J, Yang S, Yan Wet al, 2015, MicroRNA-19 triggers epithelialmesenchymal transition of lung cancer cells accompanied by growth inhibition. Lab Investig 95(9):1056–1070 Cellura D, Pickard K, Quaratino S et al, 2015, miR-19-mediated inhibition of transglutaminase-2 leads to enhanced invasion and metastasis in colorectal cancer. Mol Cancer Res 13(7):1095–1105 Tsai MM, Wang CS, Tsai CY et al, 2014, MicroRNA-196a/-196b promote cell metastasis via negative regulation of radixin in human gastric cancer. Cancer Lett 351(2):222–231 Wu Q, Yang Z, An Yet al, 2014, MiR-19a/b modulate the metastasis of gastric cancer cells by targeting the tumour suppressor MXD1. Cell Death Dis 27(5):e1144 Shrestha S, Hsu SD, Huang Wet al, 2014, A systematic review of microRNA expression profiling studies in human gastric cancer. Cancer Med 3(4):878–888 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 427 EP 438 DI 10.1007/s12253-017-0247-x PG 12 ER PT J AU Shinmura, K Kato, H Kawanishi, Y Kamo, T Inoue, Y Yoshimura, K Sugiyama, K Misawa, K Hosokawa, S Mineta, H Sugimura, H AF Shinmura, Kazuya Kato, Hisami Kawanishi, Yuichi Kamo, Takaharu Inoue, Yusuke Yoshimura, Katsuhiro Sugiyama, Kenta Misawa, Kiyoshi Hosokawa, Seiji Mineta, Hiroyuki Sugimura, Haruhiko TI BSND is a Novel Immunohistochemical Marker for Oncocytic Salivary Gland Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BSND; Immunohistochemical marker; Oncocytic salivary gland tumor; Oncocytoma; Warthin’s tumor ID BSND; Immunohistochemical marker; Oncocytic salivary gland tumor; Oncocytoma; Warthin’s tumor AB BSND protein, which is involved in chloride transport, is expressed in normal kidney and the inner ear and is known as an immunohistochemical marker for chromophobe renal cell carcinoma (RCC) and renal oncocytoma; however, other organs and tumor types exhibiting BSND expression have not yet been reported. In this study, we investigated the expression of BSND using data from the Cancer Genome Atlas (TCGA) database and by performing immunohistochemical analyses. As a result, we found that BSND was also expressed in the striated duct cells of normal salivary glands. Next, BSND expression was examined immunohistochemically in 7 types of salivary gland tumors, and BSND positivity was found in Warthin’s tumor (25 out of 25 cases; 100%) and oncocytoma (4/4; 100%), both of which are usually classified as oncocytic tumors, whereas BSND negativity was observed for pleomorphic adenoma (0/11), adenoid cystic carcinoma (0/7), acinic cell carcinoma (0/6), mucoepidermoid carcinoma (0/6), and salivary duct carcinoma (0/5). Finally, the expression of BSND mRNA in 30 types of tumors other than chromophobe RCC and salivary gland tumors was examined using data from the TCGA database, but none of these tumors exhibited BSND expression. These results suggest that BSND is expressed only in normal salivary glands and oncocytic salivary gland tumors such as Warthin’s tumor and oncocytoma in addition to the two known organs and the two known renal tumor types mentioned above. The selective expression pattern of BSND suggests that BSND is an excellent novel immunohistochemical marker for oncocytic salivary gland tumors. C1 [Shinmura, Kazuya] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Japan. [Kato, Hisami] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Japan. [Kawanishi, Yuichi] Hamamatsu University School of Medicine, Preeminent Medical Photonics Education and Research CenterHamamatsu, Japan. [Kamo, Takaharu] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Japan. [Inoue, Yusuke] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Japan. [Yoshimura, Katsuhiro] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Japan. [Sugiyama, Kenta] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Japan. [Misawa, Kiyoshi] Hamamatsu University School of Medicine, Department of Otolaryngology / Head and Neck SurgeryHamamatsu, Japan. [Hosokawa, Seiji] Hamamatsu University School of Medicine, Department of Otolaryngology / Head and Neck SurgeryHamamatsu, Japan. [Mineta, Hiroyuki] Hamamatsu University School of Medicine, Department of Otolaryngology / Head and Neck SurgeryHamamatsu, Japan. [Sugimura, Haruhiko] Hamamatsu University School of Medicine, Department of Tumor Pathology, 1-20-1 Handayama, Higashi Ward, 431-3192 Hamamatsu, Japan. RP Shinmura, K (reprint author), Hamamatsu University School of Medicine, Department of Tumor Pathology, 431-3192 Hamamatsu, Japan. EM kzshinmu@hama-med.ac.jp CR Estevez R, Boettger T, Stein V et al, 2001, Barttin is a Cl- channel beta-subunit crucial for renal Cl- reabsorption and inner ear K+ secretion. Nature 414:558–561 Birkenhager R, Otto E, Schurmann MJ et al, 2001, Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure. Nat Genet 29:310–314 Shinmura K, Igarashi H, Kato H et al, 2015, BSNDand ATP6V1G3: novel Immunohistochemical markers for Chromophobe renal cell carcinoma. Medicine, Baltimore, 94:e989 Barnes L, Eveson JW, Reichart P, Sidransky D, 2005)World Health Organization classification of tumors: pathology and genetics of the head and neck Tumours. IARC Press, Lyon Nagao T, Sato E, Inoue R et al, 2012, Immunohistochemical analysis of salivary gland tumors: application for surgical pathology practice. Acta Histochem Cytochem 45:269–282 Ellis GL, Auclair PL, 2008, Tumors of the salivary glands, AFIP atlas of tumor pathology: series 4, book 9). American Registry of Pathology, Washington, DC Shintaku M, Honda T, 1997, Identification of oncocytic lesions of salivary glands by anti-mitochondrial immunohistochemistry. Histopathology 31:408–411 Foschini MP, Marucci G, Eusebi V, 2002, Low-grade mucoepidermoid carcinoma of salivary glands: characteristic immunohistochemical profile and evidence of striated duct differentiation. Virchows Arch 440:536–542 Akai A, Yamamura Y, Nonaka M et al, 2014, 99mTcO4− accumulation in scintigraphy and expression of Na+/I− symporter in salivary gland tumors. Auris Nasus Larynx 41:532–538 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 439 EP 444 DI 10.1007/s12253-017-0248-9 PG 6 ER PT J AU Masood, N Basharat, Z Khan, T Yasmin, A AF Masood, Nosheen Basharat, Zarrin Khan, Tabeer Yasmin, Azra TI Erratum to: Entangling Relation of Micro RNA-let7, miRNA-200 and miRNA-125 with Various Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Erratum to: Pathol. Oncol. Res. DOI 10.1007/s12253-016-0184-0 The original version of this article unfortunately contained an error in the author group section. The author group should have been Nosheen Masood, Zarrin Basharat, Tabeer Khan and Azra Yasmin instead of Nosheen Masood & Azra Yasmin. The corrected author group is shown above. C1 [Masood, Nosheen] Fatima Jinnah Women University, The MallRawalpindi, Pakistan. [Basharat, Zarrin] Fatima Jinnah Women University, The MallRawalpindi, Pakistan. [Khan, Tabeer] Fatima Jinnah Women University, The MallRawalpindi, Pakistan. [Yasmin, Azra] Fatima Jinnah Women University, The MallRawalpindi, Pakistan. RP Masood, N (reprint author), Fatima Jinnah Women University, The Mall, Rawalpindi, Pakistan. EM nosheenmasood@hotmail.com NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2018 VL 24 IS 2 BP 445 EP 445 DI 10.1007/s12253-016-0184-0 PG 1 ER PT J AU Massari, F Ciccarese, Ch Hes, O Michal, M Calio, A Fiorentino, M Giunchi, F D’Amuri, A Sanguedolce, F Sabbatini, R Guida, A Ardizzoni, A Porta, C Iacovelli, R Tortora, G Cima, L Ortega, C Lapini, A Martignoni, G Brunelli, M AF Massari, Francesco Ciccarese, Chiara Hes, Ondrej Michal, Michal Calio, Anna Fiorentino, Michelangelo Giunchi, Francesca D’Amuri, Alessandro Sanguedolce, Francesca Sabbatini, Roberto Guida, Annalisa Ardizzoni, Andrea Porta, Camillo Iacovelli, Roberto Tortora, Giampaolo Cima, Luca Ortega, Cinzia Lapini, Alberto Martignoni, Guido Brunelli, Matteo TI The Tumor Entity Denominated "clear cell-papillary renal cell carcinoma" According to the WHO 2016 new Classification, have the Clinical Characters of a Renal Cell Adenoma as does Harbor a Benign Outcome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Clear cell papillary RCC; Renal adenomatoid tumour (RAT); WHO 2016 classification of renal neoplasia; Renal adenoma; Benign ID Clear cell papillary RCC; Renal adenomatoid tumour (RAT); WHO 2016 classification of renal neoplasia; Renal adenoma; Benign AB The new WHO 2016 classification of renal neoplasia encounters the new entity called "clear cell papillary renal cell carcinoma" (ccpRCC). The ccpRCC has been long included as a subtype of clear cell RCC histotype and it actually ranges from 2 to 9% in different routinely available cohort of renal carcinomas. Of important note, ccpRCC does not show any recurrences or metastases or lymph-node invasion and the outcome is always good. We reviewed twenty-four publications with available follow-up for patients (no. 362) affected by clear cell papillary RCCs/renal adenomatoid tumours and notably ccpRCC harbors an indolent clinical behavior after a mean of 38 months (3,5 years) of follow-up. This paper reviews the histological, molecular and clinical features characterizing ccpRCC, with the goal of focusing the knowledge of the benign fashion of this new tumour entity, supporting the idea of a new renal cell adenoma recruited morphologically from ex conventional clear cell RCC tumours. C1 [Massari, Francesco] S.Orsola-Malpighi Hospital, Division of OncologyBologna, Italy. [Ciccarese, Chiara] AOUI, University of Verona, Division of OncologyVerona, Italy. [Hes, Ondrej] Charles University, Faculty of Medicine in Plzen, Department of PathologyPilsen, Czech Republic. [Michal, Michal] Charles University, Faculty of Medicine in Plzen, Department of PathologyPilsen, Czech Republic. [Calio, Anna] AOUI, University of Verona, Department of Diagnostics and Public HealthVerona, Italy. [Fiorentino, Michelangelo] S.Orsola-Malpighi Hospital, Department of PathologyBologna, Italy. [Giunchi, Francesca] S.Orsola-Malpighi Hospital, Department of PathologyBologna, Italy. [D’Amuri, Alessandro] Card. G. Panico Hospital, Anatomic PathologyLecce, Italy. [Sanguedolce, Francesca] University of Foggia, Department of PathologyFoggia, Italy. [Sabbatini, Roberto] University of Modena & Reggio Emilia, Department of Oncology, Hematology & Respiratory DiseasesModena, Italy. [Guida, Annalisa] University of Modena & Reggio Emilia, Department of Oncology, Hematology & Respiratory DiseasesModena, Italy. [Ardizzoni, Andrea] S.Orsola-Malpighi Hospital, Division of OncologyBologna, Italy. [Porta, Camillo] I.R.C.C.S. San Matteo University Hospital Foundation, Division of OncologyPavia, Italy. [Iacovelli, Roberto] AOUI, University of Verona, Division of OncologyVerona, Italy. [Tortora, Giampaolo] AOUI, University of Verona, Division of OncologyVerona, Italy. [Cima, Luca] AOUI, University of Verona, Department of Diagnostics and Public HealthVerona, Italy. [Ortega, Cinzia] Asl Cn2 Alba-Bra, Institute for Cancer Research and Treatment, Division of OncologyPiemonte, Italy. [Lapini, Alberto] University of Florence, Carreggi Hospital, Urology Clinic, Peschiera del GardaFlorence, Italy. [Martignoni, Guido] AOUI, University of Verona, Department of Diagnostics and Public HealthVerona, Italy. [Brunelli, Matteo] AOUI, University of Verona, Department of Diagnostics and Public HealthVerona, Italy. RP Brunelli, M (reprint author), AOUI, University of Verona, Department of Diagnostics and Public Health, Verona, Italy. EM matteo.brunelli@univr.it CR Moch H, Humphrey PA, Ulbright TM, Reuter VE, 2016, WHO classification of Tumours of the urinary system and male genital organs, IARC WHO classification of Tumours). IARC, Lyon Srigley JR, Delahunt B, Eble JN, Egevad L, Epstein JI, Grignon D, Hes O, Moch H, Montironi R, Tickoo SK, Zhou M, Argani P, 2013, The International Society of Urological Pathology, ISUP, Vancouver classification of renal Neoplasia. Am J Surg Pathol 37(10):1469–1489., DOI 10.1097/PAS.0b013e318299f2d1 Tickoo SK, dePeralta-Venturina MN, Harik LR, Worcester HD, Salama ME, Young AN, Moch H, Amin MB, 2006, Spectrum of epithelial neoplasms in end-stage renal disease: an experience from 66 tumor-bearing kidneys with emphasis on histologic patterns distinct from those in sporadic adult renal neoplasia. Am J Surg Pathol 30(2):141–153 Aron M, Chang E, Herrera L, Hes O, Hirsch MS, Comperat E, Camparo P, Rao P, Picken M, Michal M, Montironi R, Tamboli P, Monzon F, Amin MB, 2015, Clear cell-papillary renal cell carcinoma of the kidney not associated with end-stage renal disease: clinicopathologic correlation with expanded immunophenotypic and molecular characterization of a large cohort with emphasis on relationship with renal angiomyoadenomatous tumor. Am J Surg Pathol 39(7):873–888., DOI 10.1097/PAS.0000000000000446 Adam J, Couturier J, Molinie V, Vieillefond A, Sibony M, 2011, Clear-cell papillary renal cell carcinoma: 24 cases of a distinct lowgrade renal tumour and a comparative genomic hybridization array study of seven cases. Histopathology 58(7):1064–1071., DOI 10. 1111/j.1365-2559.2011.03857.x Gobbo S, Eble JN, Grignon DJ, Martignoni G, MacLennan GT, Shah RB, Zhang S, Brunelli M, Cheng L, 2008, Clear cell papillary renal cell carcinoma: a distinct histopathologic and molecular genetic entity. Am J Surg Pathol 32(8):1239–1245., DOI 10.1097/PAS. 0b013e318164bcbb Williamson SR, Zhang S, Eble JN, Grignon DJ, Martignoni G, Brunelli M, Wang M, Gobbo S, Baldridge LA, Cheng L, 2013, Clear cell papillary renal cell carcinoma-like tumors in patients with von Hippel-Lindau disease are unrelated to sporadic clear cell papillary renal cell carcinoma.AmJ Surg Pathol 37(8):1131–1139., DOI 10.1097/PAS.0b013e318282dab8 Aydin H, Chen L, Cheng L, Vaziri S, He H, Ganapathi R, Delahunt B,Magi-Galluzzi C, ZhouM(2010, Clear cell tubulopapillary renal cell carcinoma: a study of 36 distinctive low-grade epithelial tumors of the kidney. Am J Surg Pathol 34(11):1608–1621., DOI 10.1097/ PAS.0b013e3181f2ee0b Park JH, Lee C, Suh JH, Moon KC, 2012, Clear cell papillary renal cell carcinoma: a report of 15 cases including three cases of concurrent other-type renal cell carcinomas. Korean J of Pathol 46(6): 541–547., DOI 10.4132/Korean JPathol.2012.46.6.541 Williamson SR, Eble JN, Cheng L, Grignon DJ, 2013, Clear cell papillary renal cell carcinoma: differential diagnosis and extended immunohistochemical profile. Mod Pathol: Official J of the United States Canadian Acad Pathol Inc 26(5):697–708., DOI 10.1038/ modpathol.2012.204 Mai KT, Kohler DM, Belanger EC, Robertson SJ, Wang D, 2008, Sporadic clear cell renal cell carcinoma with diffuse cytokeratin 7 immunoreactivity. Pathology 40(5):481–486., DOI 10.1080/ 00313020802197962 Zhou H, Zheng S, Truong LD, Ro JY, Ayala AG, Shen SS, 2014, Clear cell papillary renal cell carcinoma is the fourth most common histologic type of renal cell carcinoma in 290 consecutive nephrectomies for renal cell carcinoma. Hum Pathol 45(1):59–64., DOI 10. 1016/j.humpath.2013.08.004 Gill S, Kauffman EC, Kandel S, George S, Schwaab T, Xu B, 2015, Incidence of clear cell papillary renal cell carcinoma in low-grade renal cel l carcinoma cases: a 12-year retrospective Clinicopathologic study from a single cancer center. Int J Surg Pathol., DOI 10.1177/1066896915613432 Alexiev BA, Thomas C, Zou YS, 2014, Clear cell papillary renal cell carcinoma with angiomyomatous stroma: a histological, immunohistochemical, and fluorescence in situ hybridization study. Virchows Archiv: Intern J Pathol 464(6):709–716., DOI 10.1007/ s00428-014-1581-y Bhatnagar R, Alexiev BA, 2012, Renal-cell carcinomas in endstage kidneys: a clinicopathological study with emphasis on clearcell papillary renal-cell carcinoma and acquired cystic kidney disease-associated carcinoma. Int J Surg Pathol 20(1):19–28., DOI 10.1177/1066896911414273 Alexiev BA, Drachenberg CB, 2014, Clear cell papillary renal cell carcinoma: incidence, morphological features, immunohistochemical profile, and biologic behavior: a single institution study. Pathol Res Pract 210(4):234–241., DOI 10.1016/j.prp.2013.12.009 Shi SS, Shen Q, Xia QY, Tu P, Shi QL, Zhou XJ, Rao Q, 2013, Clear cell papillary renal cell carcinoma: a clinicopathological study emphasizing ultrastructural features and cytogenetic heterogeneity. Int J Clin Exp Pathol 6(12):2936–2942 Behdad A, Monzon F, Hes O, Michal M, Comperat E, Camparo P, Rao P, Picken M, Montironi R, Annaiah C, Herrera L, Arora K, Westfall DE, Tamboli P, Amin M, 2011, Relationship between sporadic clear cell-papillary renal cell carcinoma, CP-RCC, and renal angiomyoadenomatous tumor, RAT, of the kidney: analysis by virtual karyotyping, fluorescent in situ analysis and immunohistochemistry, IHC). Modern pathology: an official journal of the United States and Canadian Academy of Pathology, Inc 24(Suppl):179A Rohan SM, Xiao Y, Liang Y, Dudas ME, Al-Ahmadie HA, Fine SW, Gopalan A, Reuter VE, Rosenblum MK, Russo P, Tickoo SK, 2011, Clear-cell papillary renal cell carcinoma: molecular and immunohistochemical analysis with emphasis on the von Hippel- Lindau gene and hypoxia-inducible factor pathway-related proteins. Mod Pathol: Official J of the United States Canadian Acad Pathol Inc 24(9):1207–1220., DOI 10.1038/modpathol.2011.80 Leroy X, Camparo P, Gnemmi V, Aubert S, Flamand V, Roupret M, Fantoni JC, Comperat E, 2014, Clear cell papillary renal cell carcinoma is an indolent and low-grade neoplasm with overexpression of cyclin-D1. Histopathology 64(7):1032–1036., DOI 10.1111/his. 12359 YanWX, CaoWR, Zhao J, ZhangW,Wang XL, Yuan Q, Dang SQ, 2015, Clear cell papillary renal cell carcinoma: a clinicopathologic analysis of 6 cases. Int J Clin Exp Pathol 8(5):4595–4599 Michal M, Hes O, Havlicek F, 2000, Benign renal angiomyoadenomatous tumor: a previously unreported renal tumor. Ann Diagn Pathol 4(5):311–315., DOI 10.1053/adpa.2000. 17890 Singh C, Kendi AT, Manivel JC, Pambuccian SE, 2012, Renal angiomyoadenomatous tumor. Ann Diagn Pathol 16(6):470–476., DOI 10.1016/j.anndiagpath.2012.01.006 Kuroda N, Michal M, Hes O, Taguchi T, Tominaga A, Mizobuchi K, Ohe C, Sakaida N, Uemura Y, Shuin T, Lee GH, 2009, Renal angiomyoadenomatous tumor: fluorescence in situ hybridization. Pathol Int 59(9):689–691., DOI 10.1111/j.1440-1827.2009.02429.x Michal M, Hes O, Nemcova J, Sima R, Kuroda N, Bulimbasic S, Franco M, Sakaida N, Danis D, Kazakov DV, Ohe C, Hora M, 2009, Renal angiomyoadenomatous tumor: morphologic, immunohistochemical, and molecular genetic study of a distinct entity. Virchows Archiv: Intern J Pathol 454(1):89–99., DOI 10.1007/ s00428-008-0697-3 Deml KF, Schildhaus HU, Comperat E, von Teichman A, Storz M, Schraml P, Bonventre JV, Fend F, Fleige B, Nerlich A, Gabbert HE, GaBler N, Grobholz R, Hailemariam S, Hinze R, Knuchel R, Lhermitte B, Nesi G, Rudiger T, Sauter G, Moch H, 2015, Clear cell papillary renal cell carcinoma and renal angiomyoadenomatous tumor: two variants of a morphologic, immunohistochemical, and genetic distinct entity of renal cell carcinoma. Am J Surg Pathol 39(7):889–901., DOI 10.1097/PAS.0000000000000456 Fisher KE, Yin-Goen Q, Alexis D, Sirintrapun JS, Harrison W, Benjamin Isett R, Rossi MR, Moreno CS, Young AN, Osunkoya AO, 2014, Gene expression profiling of clear cell papillary renal cell carcinoma: comparison with clear cell renal cell carcinoma and papillary renal cell carcinoma. Mod Pathol 27(2):222–230., DOI 10. 1038/modpathol.2013.140 Wolfe A, Dobin SM, Grossmann P, Michal M, Donner LR, 2011, Clonal trisomies 7,10 and 12, normal 3p and absence of VHL gene mutation in a clear cell tubulopapillary carcinoma of the kidney. Virchows Archiv: Intern J Pathol 459(4):457–463., DOI 10.1007/ s00428-011-1137-3 Rao P,Monzon F, Jonasch E, Matin SF, Tamboli P, 2014, Clear cell papillary renal cell carcinoma in patients with von Hippel-Lindau syndrome–clinicopathological features and comparative genomic analysis of 3 cases. Hum Pathol 45(9):1966–1972., DOI 10.1016/j. humpath.2014.06.004 Lawrie CH, Larrea E, Larrinaga G, Goicoechea I, Arestin M, Fernandez-Mercado M, Hes O, Caceres F, Manterola L, Lopez JI, 2014, Targeted next-generation sequencing and non-coding RNA expression analysis of clear cell papillary renal cell carcinoma suggests distinct pathological mechanisms from other renal tumour subtypes. J Pathol 232(1):32–42., DOI 10.1002/path.4296 Raspollini MR, Castiglione F, Cheng L, Montironi R, Lopez- Beltran A, 2016, Genetic mutations in accordance with a low malignant potential tumour are not demonstrated in clear cell papillary renal cell carcinoma. J Clin Pathol., DOI 10.1136/jclinpath-2015- 203565 Munari E, Marchionni L, Chitre A, Hayashi M, Martignoni G, Brunelli M, Gobbo S, Argani P, Allaf M, Hoque MO, Netto GJ, 2014, Clear cell papillary renal cell carcinoma: micro-RNA expression profiling and comparison with clear cell renal cell carcinoma and papillary renal cell carcinoma. Hum Pathol 45(6):1130– 1138., DOI 10.1016/j.humpath.2014.01.013 Dhakal HP, McKenney JK, Khor LY, Reynolds JP, Magi-Galluzzi C, Przybycin CG, 2016, Renal neoplasms with overlapping features of clear cell renal cell carcinoma and clear cell papillary renal cell carcinoma: a Clinicopathologic study of 37 cases from a single institution. Am J Surg Pathol 40(2):141–154., DOI 10.1097/PAS. 0000000000000583 Gobbo S, Eble JN,Maclennan GT, Grignon DJ, Shah RB, Zhang S, Martignoni G, Brunelli M, Cheng L, 2008, Renal cell carcinomas with papillary architecture and clear cell components: the utility of immunohistochemical and cytogenetical analyses in differential diagnosis. Am J Surg Pathol 32(12):1780–1786., DOI 10.1097/PAS. 0b013e31818649ed Kuhn E, De Anda J, Manoni S, Netto G, Rosai J, 2006, Renal cell carcinoma associated with prominent angioleiomyoma-like proliferation: report of 5 cases and review of the literature. Am J Surg Pathol 30(11):1372–1381., DOI 10.1097/01.pas.0000213277.45715.82 Shannon BA, Cohen RJ, Segal A, Baker EG, Murch AR, 2009, Clear cell renal cell carcinoma with smooth muscle stroma. Hum Pathol 40(3):425–429., DOI 10.1016/j.humpath.2008.05.021 Iczkowski KA, Shanks JH, Burdge AH, Cheng L, 2013, Renal cell carcinoma with clear cells, smooth muscle stroma, and negative for 3p deletion: a variant of renal angiomyoadenomatous tumour? A case report. Histopathology 62(3):522–524., DOI 10.1111/his.12040 Martignoni G, Brunelli M, Segala D, Gobbo S, Borze I, Atanesyan L, Savola S, Barzon L, Masi G, Tardanico R, Zhang S, Eble JN, Chilosi M, Bohling T, Cheng L, Delahunt B, Knuutila S, 2014, Renal cell carcinoma with smooth muscle stroma lacks chromosome 3p and VHL alterations. Mod Pathol: Official J of the United States Canadian Acad Pathol Inc 27(5):765–774., DOI 10. 1038/modpathol.2013.180 Canzonieri V, Volpe R, Gloghini A, Carbone A, Merlo A, 1993, Mixed renal tumor with carcinomatous and fibroleiomyomatous components, associated with angiomyolipoma in the same kidney. Pathol Res Pract 189(8):951–956; discussion 957-959., DOI 10. 1016/S0344-0338(11)81110-6 Hakimi AA, Tickoo SK, Jacobsen A, Sarungbam J, Sfakianos JP, Sato Y, Morikawa T, Kume H, Fukayama M, Homma Y, Chen YB, Sankin AI, Mano R, Coleman JA, Russo P, Ogawa S, Sander C, Hsieh JJ, Reuter VE, 2015, TCEB1-mutated renal cell carcinoma: a distinct genomic andmorphological subtype. Mod Pathol: Official J of the United States Canadian Acad Pathol Inc 28(6):845–853., DOI 10.1038/modpathol.2015.6 Diolombi ML, Cheng L, Argani P, Epstein JI, 2015, Do clear cell papillary renal cell carcinomas have malignant potential? Am J Surg Pathol 39(12):1621–1634. do i :10.1097/PAS. 0000000000000513 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 447 EP 456 DI 10.1007/s12253-017-0271-x PG 10 ER PT J AU Lopez-Cortes, A Cabrera-Andrade, A Ona-Cisneros, F Echeverria, C Rosales, F Ortiz, M Tejera, E Paz-y-Mino, C AF Lopez-Cortes, Andres Cabrera-Andrade, Alejandro Ona-Cisneros, Fabian Echeverria, Carolina Rosales, Felipe Ortiz, Malena Tejera, Eduardo Paz-y-Mino, Cesar TI Breast Cancer Risk Associated with Genotype Polymorphisms of the Aurora Kinase a Gene (AURKA): a Case-Control Study in a High Altitude Ecuadorian Mestizo Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; AURKA; Ecuadorianpopulation; Genotyping; High altitude; Mestizo ID Breast cancer; AURKA; Ecuadorianpopulation; Genotyping; High altitude; Mestizo AB Breast cancer (BC) is the leading cause of cancer related death among women in 2014. The AURKA gene that encodes the protein called Aurora kinase A plays an important role in the progression of the cell cycle, by controlling and promoting the entry into the phase of mitosis. The single nucleotide polymorphism AURKA T91A (rs2273535) (Phe21Ile) has been identified as functional alternator of this kinase, the Ile allele is associated with the occurrence of chromosome segregation errors and tumor progression. Therefore, it is essential to know how BC risk is associated with histopathological characteristics, immunohistochemical characteristics, and genotype polymorphism in a high altitude Ecuadorian mestizo population. In this retrospective casecontrol study 200 individuals were analyzed. DNA was extracted from 100 healthy and 100 affected women. Genotypes were determined by genomic sequencing. We found significant association between the AURKA T91A (rs2273535) (Phe21Ile) genotype and an increased risk of BC development: Phe/Ile (odds ratio [OR] = 2.6; 95% confidence interval [CI] = 1.4–4.9; P = 0.004), Ile/Ile (OR = 3.8; 95% CI = 1.6–9.0; P = 0.002), and Phe/Ile + Ile/Ile (OR = 2.9; 95% CI = 1.6–5.2; P = 0.001). Additionally, the rs2273535 variant was associated with the tumor grade SBR III (OR = 9.6; 95% CI = 1.0–91.9; P = 0.048) and the Ki-67 ≥ 20 (OR = 16.5; 95% CI = 2.7–101.3; P = 0.002). In brief, this study provides the first evidence where the Ile allele of the AURKA gene could act as potentially predictive biomarker of BC in the high altitude Ecuadorian mestizo population that lives at 2800 m above sea level (masl). C1 [Lopez-Cortes, Andres] Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, Avenue Mariscal Sucre, 170129 Quito, Ecuador. [Cabrera-Andrade, Alejandro] Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, Avenue Mariscal Sucre, 170129 Quito, Ecuador. [Ona-Cisneros, Fabian] Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, Avenue Mariscal Sucre, 170129 Quito, Ecuador. [Echeverria, Carolina] Ministerio de Salud Publica, Direccion Nacional de Inteligencia de la Salud, Avenue Republica de El Salvador, 170515 Quito, Ecuador. [Rosales, Felipe] Hospital Oncologico Solon Espinosa Ayala, Departamento de Patologia, Avenue Eloy Alfaro, 170138 Quito, Ecuador. [Ortiz, Malena] Ministerio de Salud Publica del Ecuador, Hospital General Docente de Calderon, Avenue Capitan Giovanni Calles, 170206 Quito, Ecuador. [Tejera, Eduardo] Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, Avenue Mariscal Sucre, 170129 Quito, Ecuador. [Paz-y-Mino, Cesar] Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, Avenue Mariscal Sucre, 170129 Quito, Ecuador. RP Lopez-Cortes, A (reprint author), Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, 170129 Quito, Ecuador. EM aalc84@gmail.com CR Cancer Research UK. Worldwide cancer statistics. In: Cancer statistics. 2014. www.cancerresearchuk.org/cancer info/cancerstats/ world. Accessed 7 Nov 2014 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90 World Health Organization. International Agency for Research on Cancer. GLOBOCAN 2012: Estimated cancer incidence, mortality and prevalence worldwide in 2012. 2012. http://globocan.iarc.fr/ Default.aspx. Accessed 7 Nov 2016 Kumar V, Abbas A, Aster J., 2012, Robbins basic pathology. 9th ed. Philadelphia: Saunders National Cancer Institute at the National Institutes of Health, 2014, Histopathologic classification of breast cancer. In: Breast cancer treatment. http://www.cancer.gov/cancertopics/pdq/treatment/ breast/healthprofessional/page2. Accessed 7 Nov 2014 Kumar R, Sharma A, Tiwari R, 2012, Application of microarray in breast cancer: an overview. J Pharm Bioallied Sci 4:21–26 Banerji S, Cibulskis K, Rangel-Escareno C, Brown K, Carter S, Frederick A, Lawrance M, Sivachenko A, Sougnez C, Zou L, Cortes M, Fernandez-Lopez J, Peng S, Ardlie K, Auclair D, Bautista-Pina V, Duke F, Francis J, Jung J, Maffuz-Aziz A, Onofrio R, Parkin M, Pho N, Quintanar-Jurado V, Ramos A, Rebollar-Vega R, Rodriguez-Cuevas S, Romero-Cordoba S, Schumacher S, Stransky N, Thompson K, Uribe-Figueroa L, Baselga J, Beroukhim R, Polyak K, Sgroi D, Richardson A, Jimenez-Sanchez G, Lander E, Gabriel S, Garraway L, Golub T, Melendez-Zajgla J, Toker A, Getz G, Hidalgo-Miranda A, Meyerson M, 2012, Sequence analysis of mutations and translocations across breast cancer subtypes. Nature 486:405–409 Eisenstein M, 2015, Startups use short-read data to expand longread sequencing market. Nat Biotechnol 33:433–435 Wong KM, Hudson TJ, McPherson JD, 2011, Unraveling the genetics of cancer: genome sequencing and beyond. Annu Rev Genomics Hum Genet 12:407–430 Meyerson M, Gabriel S, Getz G, 2010, Advances in understanding cancer genomes through second-generation sequencing. Nat Rev Genet 11:685–696 Eifert C, Powers RS, 2012, From cancer genomes to oncogenic drivers, tumour dependencies and therapeutic targets. Nat Rev Cancer 12:572–578 Lovly CM,McDonald NT, Chen H, Ortiz-Cuaran S, Heukamp LC, Yan Y, Florin A, Ozretic L, Lim D,Wang L, Chen Z, Chen X, Lu P, Paik PK, Shen R, Jin H, Buettner R, Ansen S, Perner S, Brockmann M, Bos M,Wolf J, GardiziM,Wright GM, Solomon B, Rusell PA, Rogers TM, Suehara Y, Red-Brewer M, Tieu R, de Stanchina E, Wang Q, Zhao Z, Johonson DH, Horn L, Wong KK, Thomas RK, Ladanyi M, Pao W, 2014, Rationale for co-targeting IGF-1R and ALK in ALK fusion-positive lung cancer. Nat Med 20:1027–1034 Xia J, Jia P, Hutchinson KE, Dahlman KB, Johnson D, Sosman J, Pao W, Zhao Z, 2014, A meta-analysis of somatic mutations from next generation sequencing of 241 melanomas: a road map for the study of genes with potential clinical relevance. Mol Cancer Ther 13:1918–1928 Jia P, Jin H, Meador CB, Xia J, Ohashi K, Liu L, Pirazzoli V, Dahmal KB, Politi K, Michor F, Zhao Z, Pao W, 2013, Nextgeneration sequencing of paired tyrosine kinase inhibitor-sensitive and –resistant EGFR mutant lung cancer cell lines identifies spectrum of DNAchanges associated with drug resistance. Genome Res 23:1434–1445 Dahlman KB, Xia J, Hutchinson K, Ng C, Hucks D, Jia P, Atefi M, Su Z, Branch S, Lyle PL, Hicks DJ, Bozon V, Glaspy JA, Rosen N, Solit DB, Netterville JL, Vnencak-Jones c, Sosman JA, Ribas A, Zhao Z, Pao W, 2012, BRAF(L597, mutations in melanoma are associated with sensitivity to MEK inhibitors. Cancer Discov 2: 791–797 Cheng F, Zhao J, Zhao Z, 2016, Advances in computational approaches for prioritizing driver mutations and significantly mutated genes in cancer genomes. Brief Bioinform 17:642–656 Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S,Diaz LA Jr, Kinzler KW(2013, Cancer genome landscapes. Science 339:1546– 1558 Khan J, Ezan F, Cremet J, Fautrel A, Gilot D, Lambert M, Benaud C, Troadec MB, Prigent C, 2011, Overexpression of active aurora- C kinase results in cell transformation and tumour formation. PLoS One 6:e26512 Lim SK, Gopalan G, 2007, Aurora-a kinase interacting protein 1, AURKAIP1, promotes aurora-a degradation through an alternative ubiquitin-independent pathway. Biochem J 403:119–127 Fu J, Bian M, Jiang Q, Zhang C, 2007, Roles of aurora kinases in mitosis and tumorigenesis. Mol Cancer Res 5:1–10 Sun T, Miao X, Wang J, Tan W, Zhou Y, Yu C, Lin D, 2004, Functional Phe31Ile polymorphism in aurora a and risk of breast carcinoma. Carcinogenesis 25:2225–2230 Xu J, Wu X, Zhou WH, Liu AW, Wu JB, Deng JY, Yue CF, Yang SB, Wang J, Yuan ZY, Liu Q, 2013, Aurora-a identifies early recurrence and poor prognosis and promises a potential therapeutic target in triple negative breast cancer. PLoS One 8:e56919 Mendiola M, Barriuso J, Marino-Enriquez A, Redondo A, Dominguez-Caceres A, Hernandez-Cortes G, Perez-Fernandez E, Sanchez-Navarro I, Vera JA, Suarez A, Espinosa E, Gonzalez- Baron M, Palacios J, Hardisson D, 2009, Aurora kinases as prognostic biomarkers in ovarian carcinoma. Hum Pathol 40:631–638 Dai ZJ, Kang HF,Wang XJ, Shao YP, Lin S, Zhao Y, Ren HT, Min WL, Wang M, Liu XX, 2014, Association between genetic polymorphisms in AURKA, rs2273535 and rs1047972, and breast cancer risk: a meta-analysis involving 37,221 subjects. Cancer Cell Int 14:91 Lopez-Cortes A, Echeverria C, Ona-Cisneros F, Sanchez ME, Herrera C, Cabrera-Andrade A, Rosales F, Ortiz M, Paz-y-Mino C, 2015, Breast cancer risk associated with gene expression and genotype polymorphisms of thefolate-metabolizing MTHFR gene: a case-control study in a high altitude Ecuadorian mestizo population. Tumor Biol 36:6451–6461 Rodriguez S, Gaunt TR, Day IN, 2009, Hardy-Weinberg equilibrium testing of biological ascertainment for Mendelian randomization studies. Am J Epidemiol 169:505–514 Engle LJ, Simpson CL, Landers JE, 2006, Using high-throughput SNP technologies to study cancer. Oncogene 25:1594–1601 Fejerman L, Ahmadiyeh N, Hu D, Huntsman S, Beckman KB, Caswell JL, Tsung K, John EM, Torres-Mejia G, Carvajal- Carmona L, Echeverry MM, Tuazon AM, Ramirez C, COLUMBUS Consortium, Gognoux CR, Eng C, Gonzalez- Burchard E, Henderson B, Le Marchard L, Kooperberg C, Hou L, Agalliu I, Kraft P, Lindstrom S, Perez-Stable EJ, Haiman CA, Ziv E, 2014, Genome-wide association study of breast cancer in Latinas identifies novel protective variants on 6q25. Nat Commun 5:5260 Ruan Y, Song AP, Wang H, Xie YT, Han JY, Sajdik C, Tian XX, Fang WG, 2011, Genetic polymorphisms in AURKA and BRCA1 are associated with breast cancer susceptibility in a Chinese Han population. J Pathol 225:535–543 Webb EL, Rudd MF, Houlston RS, 2006, Case-control, kin-cohort and meta-analyses provide no support for STK15 F31I as a low penetrance colorectal cancer allele. Br J Cancer 95:1047–1049 Miao X, Sun T,Wang Y, Zhang X, TanW, Lin D, 2004, Functional STK15 Phe31Ile polymorphism is associated with the occurrence and advanced disease status of esophageal squamous cell carcinoma. Cancer Res 64:2680–2683 Tang W, Qiu H, Ding H, Sun B, Wang L, Yin J, Gu H, 2013, Association between the STK15 F31I polymorphism and cancer susceptibility: a meta-analysis involving 43,626 subjects. PLoS One 8:e82790 Sun T, Miao X, Wang J, Tan W, Zhou Y, Yu C, Lin D, 2004, Functional Phe31Ile polymorphism in aurora a and risk of breast carcinoma. Carcinogenesis 25:2225–2230 Dai Q, Cai QY, Shu XO, Ewart-Toland A, Wen WQ, Balmain A, Gao YT, Zheng W, 2004, Synergistic effects of STK15 gene polymorphisms and endogenous estrogen exposure in the risk of breast cancer. Cancer Epidemiol Biomark Prev 13:2065–2070 Lo YL, Yu JC, Chen ST, Yang HC, Fann CS, Mau YC, Shen CY, 2005, Breast cancer risk associated with genotypic polymorphism of the mitosis-regulating gene aurora-STK15/BTAK. Int J Cancer 115:276–283 MARIE-GENICA Consortium on Genetic Susceptibility for Menopausal Hormone Therapy Related Breast Cancer Risk, 2010, Polymorphisms in the BRCA1 and ABCB1 genes modulate menopausal hormone therapy associated breast cancer risk in postmenopausal women. Breast Cancer Res Treat 120:727–736 Tchatchou S, Wirtenberger M, Hemminki K, Sutter C, Meindl A, Wappenschmidt B, Kiechle M, Bugert P, Schmutzler RK, Bartram CR, Burweinkel B, 2007, Aurora kinases a and B and familial breast cancer risk. Cancer Lett 247:266–272 Vidarsdottir L, Bodvarsdottir SK, Hilmarsdottir H, Tryggvadottir L, Eyfjord JE, 2007, Breast cancer risk associated with AURKA 91T –>a polymorphism in relation to BRCA mutations. Cancer Lett 250:206–212 Fletcher O, Johnson N, Palles C, dos Santos Silva I, McCormack V, Whittaker J, Ashworth A, Peto J, 2006, Inconsistent association between the STK15 F31I genetic polymorphism and breast cancer risk. J Natl Cancer Inst 98:1014–1018 Cox DG, Hankinson SE, Hunter DJ, 2006, Polymorphisms of the AURKA, STK15/aurora kinase, gene and breast cancer risk, United States). Cancer Causes Control 17:81–83 Egan KM, Newcomb PA, Ambrosone CB, Trentham-Dietz A, Titus-Ernstoff L, Hampton JM, Kimura MT, Nagase H, 2004, STK15 polymorphism and breast cancer risk in a populationbased study. Carcinogenesis 25:2149–2153 Royce ME, Xia W, Sahin AA, Katayama H, Johnston DA, Hortobagyi G, Hung MC, 2004, STK15/ aurora-a expression in primary breast tumors is correlated with nuclear grade but not with prognosis. Cancer 100:12–19 Loman N, Johannson O, Kristoffersson U, Olsson H, Borg A, 2001, Family history of breast and ovarian cancers and BRCA1 and BRCA2 mutations in a population-based series of early-onset breast cancer. J Natl Cancer Inst 93:1215–1223 Lindstrom L, Karlsson E, Wilking UM, Johansson U, Hartman J, Lidbrink EK, Hatschek T, Skoog L, Bergh J, 2012, Clinically used breast cancer markers such as estrogen receptor, progesterone receptor and human epidermal growth factor receptor 2 are unstable throughout tumor progression. J Clin Oncol 30:2601–2608 Colditz G, Rosner BA, Chen WY, Holmes MD, Hankinson SE, 2004, Risk factors for breast cancer according to estrogen and progesterone receptor status. J Natl Cancer Inst 96:218–228 Lopez-Cortes A, Jaramillo-Koupermann G, MunozMJ, Cabrera A, Echeverria C, Rosales F, Vivar N, Paz-y-Mino C, 2013, Genetic polymorphisms in MTHFR, C677T, A1298C), MTR, A2756G, andMTRR, A66G, genes associated with pathological characteristics of prostate cancer in the Ecuadorian population. Am J Med Sci 346:447–454 Paz-y-Mino C, Robles P, Salazar C, Leone PE, Garcia Cardenas JM, Naranjo M, Lopez-Cortes A, 2016, Positive association of the androgen receptor CAG repeat length polymorphism with the risk of prostate cancer. Mol Med Rep 14:1791–1798 Paz-y-Mino C, Lopez-Cortes A, Munoz MJ, Cabrera A, Castro B, Sanchez ME, 2010, Incidence of the L858R and G719S mutations of the epidermal growth factor receptor oncogene in an Ecuadorian population with lung cancer. Cancer Genet Cytogenet 196:201–203 Paz-y-Mino C,MunozMJ, Lopez-Cortes A, Cabrera A, Palacios A, Castro B, Paz-y-Mino N, Sanchez ME, 2010, Frequency of polymorphisms pro198leu in GPX-1 gene and ile58thr in MnSOD gene in the altitude Ecuadorian population with bladder cancer. Oncol Res 18:395–400 Paz-y-Mino C, Lopez-Cortes A, Munoz MJ, Castro B, Cabrera A, Sanchez ME, 2010, Relationship of an hRAD54 gene polymorphism, 2290 C/T, in an Ecuadorian population with chronic myelogenous leukemia. Genet Mol Biol 33:646–649 Quinones LA, Lavanderos MA, Cayun JP, Garcia-Martin E, Agundez JA, Caceres DD, Roco AM, Morales JE, Herrera L, Encina G, Isaza CA, Redal MA, Larovere L, Soria NW, Eslava- Schmalbach J, Castaneda-Hernandez G, Lopez-Cortes A, Magno LA, LopezM, Chiurillo MA, Rodeiro I, Castro de Guerra D, Teran E, Estevez-Carrizo F, Lares Assef I, 2014, Perception of the usefulness of drug/gene pairs and barriers for pharmacogenomics in Latin America. Curr Drug Metab 15:202–208 52 Lopez-Cortes A, Guerrero S, Redal M, Alvarado A, Quinones L, 2017, State of Art of Cancer Pharmacogenomics in Latin American Populations. Int J Mol Sci 18(6):639 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 457 EP 465 DI 10.1007/s12253-017-0267-6 PG 9 ER PT J AU Lopez-Cortes, A Cabrera-Andrade, A Ona-Cisneros, F Echeverria, C Rosales, F Ortiz, M Tejera, E Paz-y-Mino, C AF Lopez-Cortes, Andres Cabrera-Andrade, Alejandro Ona-Cisneros, Fabian Echeverria, Carolina Rosales, Felipe Ortiz, Malena Tejera, Eduardo Paz-y-Mino, Cesar TI Erratum to: Breast Cancer Risk Associated with Genotype Polymorphisms of the Aurora Kinase a Gene (AURKA): a Case-Control Study in a High Altitude Ecuadorian Mestizo Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Erratum to: Pathol. Oncol. Res. https://doi.org/10.1007/s12253-017-0275-6 Unfortunately, the name of the fourth author Carolina Echeverria and her corresponding affiliation 2 were missing in the published online paper. The corrected author group is shown above and the corrected affiliation is shown below. C1 [Lopez-Cortes, Andres] Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, Avenue Mariscal Sucre, 170129 Quito, Ecuador. [Cabrera-Andrade, Alejandro] Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, Avenue Mariscal Sucre, 170129 Quito, Ecuador. [Ona-Cisneros, Fabian] Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, Avenue Mariscal Sucre, 170129 Quito, Ecuador. [Echeverria, Carolina] Ministerio de Salud Publica, Direccion Nacional de Inteligencia de la Salud, Avenue Republica de El Salvador, 170515 Quito, Ecuador. [Rosales, Felipe] Hospital Oncologico Solon Espinosa Ayala, Departamento de Patologia, Avenue Eloy Alfaro, 170138 Quito, Ecuador. [Ortiz, Malena] Ministerio de Salud Publica del Ecuador, Hospital General Docente de Calderon, Avenue Capitan Giovanni Calles, 170206 Quito, Ecuador. [Tejera, Eduardo] Universidad de las Americas, Facultad de Ciencias de la Salud, Avenue de los Granados, 170125 Quito, Ecuador. [Paz-y-Mino, Cesar] Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, Avenue Mariscal Sucre, 170129 Quito, Ecuador. RP Lopez-Cortes, A (reprint author), Universidad Tecnologica Equinoccial, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigacion Genetica y Genomica, 170129 Quito, Ecuador. EM aalc84@gmail.com NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 467 EP 467 DI 10.1007/s12253-017-0299-y PG 1 ER PT J AU Helbig, G Koclega, A Wozniczka, K Kopera, M Kyrcz-Krzemien, S AF Helbig, Grzegorz Koclega, Anna Wozniczka, Krzysztof Kopera, Malgorzata Kyrcz-Krzemien, Slawomira TI Long-Term Outcome of Autologous Hematopoietic Stem Cell Transplantation (AHSCT) for Acute Myeloid Leukemia (AML)- Single Center Retrospective Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute myeloid leukemia; Autologous hematopoietic stem cell transplantation; Relapse; Outcome ID Acute myeloid leukemia; Autologous hematopoietic stem cell transplantation; Relapse; Outcome AB For patients with acute myeloid leukemia (AML) in complete remission without an acceptable HLA donor, the autologous hematopoietic stem cell transplantation (AHSCT) may remain a therapeutic option as remission consolidation, however its role is still a subject of continued debate. One hundred and twenty patients who underwent AHSCT for AML were included in this retrospective single center analysis. The procedure was performed over a 19 years period and transplanted patients were in first complete remission (CR1; n = 109) or in second CR (CR2; n = 11). The median age at transplant was 37 years (range 18–64). The source of stem cells was bone marrow (n = 61; 50.8%), peripheral blood (n = 36; 30%) and bone marrow with peripheral blood (n = 23; 19.2%). The median time from AML diagnosis to AHSCT was 0.8 year (range 0.3–4.4) and the median follow-up after AHSCT for surviving patients was 12.8 years (range 3.1–20.5). The median LFS was 1.1 year. The probability of LFS calculated at 5 years and 10 years after transplantation was 28% (95% CI, 22%–32%) and 21% (95% CI, 18%–24%), respectively. The last relapse occurred 14.8 years after AHSCT and among patients who survived >2 years, 28.4% (27/95) had leukemia recurrence. The median OS was 1.7 years. The probability of OS after 5 years and 10 years was 29% and 22%, respectively. There was a tendency for increased LFS for patients younger than 50 years at transplant if compared to older population. AHSCT for AML was safe with acceptable toxicity profile. Leukemia recurrence remained the leading cause of death. C1 [Helbig, Grzegorz] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street 25, 40-032 Katowice, Poland. [Koclega, Anna] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street 25, 40-032 Katowice, Poland. [Wozniczka, Krzysztof] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street 25, 40-032 Katowice, Poland. [Kopera, Malgorzata] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street 25, 40-032 Katowice, Poland. [Kyrcz-Krzemien, Slawomira] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street 25, 40-032 Katowice, Poland. RP Helbig, G (reprint author), Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, 40-032 Katowice, Poland. EM ghelbig@o2.pl CR Dohner H, Estey EH, Amadori S et al, 2010, Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood 115:453–474 Cassileth PA, Harrington DP, Hines JD et al, 1988, Maintenance chemotherapy prolongs remission duration in adult acute nonlymphocytic leukemia. J Clin Oncol 6:583–587 Schlenk RF, 2014, Post-remission therapy for acute myeloid leukemia. Haematologica 99:1663–1670 Breems DA, Lowenberg B, 2007, Acute myeloid leukemia and the position of autologous stem cell transplantation. SeminHematol 44: 259–266 4. Breems DA, Lowenberg B, 2007, Acute myeloid leukemia and the position of autologous stem cell transplantation. SeminHematol 44: 259–266 Czerw T, Labopin M, Gorin NC et al, 2016, Long-term follow-up of patients with acute myeloid leukemia surviving and free of disease recurrence for at least 2 years after autologous stem cell transplantation: a report from the acute leukemia working Party of the European Society for blood and marrow Transplnatation. Cancer 122:1880–1887 Zuckerman T, Beyar-Katz O, Rowe JM, 2016, Should autotranplantation in acute myeloid leukemia in first complete remission be revisited. Curr Opin Hematol 23:88–94 Collisson EA, Lashkari A, Malone R, Paquette R, Emmanouilides C, Territo MC, Schiller GJ, 2003, Long-term outcome of autologous transplantation of peripheral blood progenitor cells as postremission management of adult myelogenous leukemia in first complete remission. Leukemia 17:2183–2188 Mehta J, Powles R, Singhal S et al, 2000, Autologous bonemarrow transplantation for acute myeloid leukemia in first remission: identification of modifiable prognostic factors. Bone Marrow Transplant 1995; 16: 499–506 Bensinger WI, Deeg HJ, 2000, Blood or marrow? Lancet 355: 1199–1200 Bhatia S, Robison LL, Francisco L et al, 2005, Late mortality in survivors of autologous hematopoietic cell transplantation: report from the bone marrow transplant survivor study. Blood 105:4215– 4222 Vellenga E, van Putten W, Ossenkoppele GJ et al, 2011, Autologous peripheral blood stem cell transplantation for acute myeloid leukemia. Blood 118:6037–6042 Cornelissen JJ, Versluis J, Passweg JR et al, 2015, Comparative therapeutic value of post-remission approaches in patients with acute myeloid leukemia aged 40-60 years. Leukemia 29:1041–1050 Keating A, DaSilva G, Perez WS et al, 2013, Autologous blood cell transplantation versus HLA-identical sibling transplantation for acute myeloid leukemia in first complete remission: a registry study from the Center for International Blood and Marrow Transplantation Research. Haematologica 98:185–192 Phirrmann M, Ehninger G, Thiede C et al, 2012, Prediction of postremission survival in acute myeloid leukemia: a posthoc analysis of the AML96 trial. Lancet Oncol 13:207–214 Thomas X, Suciu S, Rio B et al, 2007, Autologous stem cell transplantation after complete remission and first consolidation in acute myeloid leukemia patients aged 61-70 years: results of the prospective EORTC-GIMEMA AML-13 study. Haematologica 92:389– 396 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 469 EP 475 DI 10.1007/s12253-017-0266-7 PG 7 ER PT J AU Lee, HJ Song, YS Kim, SM Yoo, JN Lee, HS AF Lee, Hwa Ju Song, Yong Sang Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Intratumoral Heterogeneity of Frameshift Mutations of GLI1 Encoding a Hedgehog Signaling Protein in Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE GLI1; Frameshift mutation; Colon cancer; Microsatellite instability; Intratumoral heterogeneity ID GLI1; Frameshift mutation; Colon cancer; Microsatellite instability; Intratumoral heterogeneity AB GLI1 is a transcription factor for hedgehog signaling that plays a crucial role in signaling pathways for controlling cell proliferation, alterations of which are known to contribute to tumorigenesis. Aim of this study was to explore whether GLI1 gene is mutated in gastric (GC) and colorectal cancers (CRC). In a public database, we found that GLI1 had a G7 mononucleotide repeat in the coding sequences that could be a mutation target in the cancers with microsatellite instability (MSI). In this study, we analyzed frameshift mutation of GLI1 in 79 GCs and 129 CRCs (high MSI (MSI-H) or microsatellite stable (MSS)) by single-strand conformation polymorphism analysis and DNA sequencing. We found 10 frameshift mutations in the repeat, nine for CRCs and one for GC. All of the mutations were detected in cancers with MSI-H and there was a statistical difference in the frameshift mutation frequencies between the cancers with MSI-H (10/113) and MSS (0/90). We also analyzed intratumoral heterogeneity (ITH) of the frameshift mutation in 16 CRCs and found that the mutations exhibited regional ITH in three of the CRCs (18.8%). Our data indicate GLI1 harbored not only frameshift mutation but also its mutational ITH, which together could be a feature of GC and CRC with MSI-H. C1 [Lee, Hwa Ju] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Song, Yong Sang] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of PathologySeoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Lim L, Beachy PA, 2004, The hedgehog response network: sensors, switches, and routers. Science 304:1755–1759 Ruiz i Altaba A, 1999, Gli proteins encode context-dependent positive and negative functions: implications for development and disease. Development 126:3205–3216 Villavicencio EH, Walterhouse DO, Iannaccone PM, 2000, The sonic hedgehog-patched-gli pathway in human development and disease. Am J Hum Genet 67:1047–1054 Jacob J, Briscoe J, 2003, Gli proteins and the control of spinal-cord patterning. EMBO Rep 4:761–765 Sanchez P, ClementV, Ruiz i AltabaA(2005, Therapeutic targeting of the hedgehog-GLI pathway in prostate cancer. Cancer Res 65: 2990–2992 Hui CC, Angers S, 2011, Gli proteins in development and disease. Annu Rev Cell Dev Biol 27:513–537 Rubin JB, Rowitch DH, 2002, Medulloblastoma: a problem of developmental biology. Cancer Cell 2:7–8 Kinzler KW, Bigner SH, Bigner DD, Trent JM, Law ML, O'Brien SJ, Wong AJ, Vogelstein B, 1987, Identification of an amplified, highly expressed gene in a human glioma. Science 236:70–73 Oniscu A, James RM, Morris RG, Bader S, Malcomson RD, Harrison DJ, 2004, Expression of sonic hedgehog pathway genes is altered in colonic neoplasia. J Pathol 203:909–917 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Calin GA, Gafa R, Tibiletti MG, Herlea V, Becheanu G, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression in microsatellite instability high, MSI-H, colon cancers correlates with clinicopathological parameters: a study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89: 230–235 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8: 305–311 Jo YS, Choi MR, Song SY, Kim MS, Yoo NJ, Lee SH, 2016, Frameshift mutations of HSPA4 and MED13 in gastric and colorectal cancers. Pathol Oncol Res 22:769–772 Jo YS, Kim MS, Yoo NJ, Lee SH, 2016, Absence of PRKD1 mutation, a salivary tumor-specific mutation, in solid tumors and Leukemias. Pathol Oncol Res 22:231–232 Choi EJ, Kim MS, Song SY, Yoo NJ, Lee SH, 2017, Intratumoral heterogeneity of frameshift mutations inMECOM Gene is frequent in colorectal cancers with high microsatellite instability. Pathol Oncol Res 23:145–149 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Lee SW,MoskowitzMA, Sims JR, 2007, Sonic hedgehog inversely regulates the expression of angiopoietin-1 and angiopoietin-2 in fibroblasts. Int J Mol Med 19:445–451 Adolphe C, Hetherington R, Ellis T,Wainwright B, 2006, Patched1 functions as a gatekeeper by promoting cell cycle progression. Cancer Res 66:2081–2088 Athar M, Li C, Tang X, Chi S, Zhang X, Kim AL, Tyring SK, Kopelovich L, Hebert J, Epstein EH Jr, Bickers DR, Xie J, 2004, Inhibition of smoothened signaling prevents ultraviolet B-induced basal cell carcinomas through regulation of Fas expression and apoptosis. Cancer Res 64:7545–7552 Choi YJ, Kim MS, An CH, Yoo NJ, Lee SH, 2014, Regional bias of intratumoral genetic heterogeneity of nucleotide repeats in colon cancers with microsatellite instability. Pathol Oncol Res 20:965–971 Choi YJ, Rhee JK, Hur SY, KimMS, Lee SH, Chung YJ, Kim TM, Lee SH, 2017, Intraindividual genomic heterogeneity of high-grade serous carcinoma of the ovary and clinical utility of ascitic cancer cells for mutation profiling. J Pathol 241:57–66 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 477 EP 481 DI 10.1007/s12253-017-0272-9 PG 5 ER PT J AU Zhao, D Zhang, Y Song, L AF Zhao, Danyi Zhang, Yang Song, Lei TI MiR-16-1 Targeted Silences Far Upstream Element Binding Protein 1 to Advance the Chemosensitivity to Adriamycin in Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; microRNA-16-1; Far upstream element binding protein 1; Chemosensitivity; Adriamycin ID Gastric cancer; microRNA-16-1; Far upstream element binding protein 1; Chemosensitivity; Adriamycin AB Chemotherapy can prevent metastasis and recurrence of gastric cancer (GC), and is a well supplement for operation. But, chemotherapy resistance has severely restricted the application of chemotherapy. This study aimed to investigate the regulatory roles and molecular mechanism of miR-16-1 to the chemosensitivity to adriamycin in GC. In this study, the expression of miR-16-1 and FUBP1 was downregulated and up-regulated respectively in adriamycinresistant GC tissues and cell lines, and represented a negative relationship between them. MiR-16-1 could silence FUBP1 directly and specifically, FUBP1 was a target gene of miR- 16-1. Silence of FUBP1 inhibited the half maximal inhibitory concentration (IC50) of SGC7901/AR cell line to adriamycin, chemosensitivity enhanced significantly. Moreover, FUBP1 silence in SGC7901/AR cell line also inhibited proliferation and invasion, and advanced cell apoptosis. To sum up, the expression of miR-16-1 was positively related with the chemosensitivity of GC to adriamycin, and miR-16-1 could targeted silence FUBP1 to advance the chemosensitivity to adriamycin in GC, which might be a novel potential therapeutic target for GC. C1 [Zhao, Danyi] Dalian Medical University, The Second Hospital, Department of Oncology, No. 467 Zhongshan Road, Shahekou District, 116027 Dalian, China. [Zhang, Yang] Dalian Medical University, The Second Hospital, Department of Oncology, No. 467 Zhongshan Road, Shahekou District, 116027 Dalian, China. [Song, Lei] Dalian Medical University, The Second Hospital, Department of Oncology, No. 467 Zhongshan Road, Shahekou District, 116027 Dalian, China. RP Song, L (reprint author), Dalian Medical University, The Second Hospital, Department of Oncology, 116027 Dalian, China. EM 23326942@qq.com CR Sugano K, 2015, Screening of gastric cancer in Asia. Best Pract Res Clin Gastroenterol 29(6):895–905 Newton AD, Datta J, Loaiza-Bonilla A, Karakousis GC, Roses RE, 2015, Neoadjuvant therapy for gastric cancer: current evidence and future directions. J Gastrointest Oncol 6(5):534–543 Piletic K, Kunej T, 2016, MicroRNA epigenetic signatures in human disease. Arch Toxicol 90(10):2405–2419 Mohammadi A, Mansoori B, Baradaran B, 2016, The role of microRNAs in colorectal cancer. Biomed Pharmacother 84:705– 713 Sekhon K, Bucay N, Majid S, Dahiya R, Saini S, 2016, MicroRNAs and epithelial-mesenchymal transition in prostate cancer. Oncotarget 7(41):67597–67611 Yang Y, Zhang P, Zhao Y, Yang J, Jiang G, Fan J, 2016, Decreased MicroRNA-26a expression causes cisplatin resistance in human non-small cell lung cancer. Cancer Biol Ther 17(5):515–525 Li J, Ju J, Ni B, Wang H, 2016, The emerging role of miR-506 in cancer. Oncotarget 7(38):62778–62788 Pekarsky Y, Croce CM, 2015, Role of miR-15/16 in CLL. Cell Death Differ 22(1):6–11 Wang W, Chen J, Dai J, Zhang B, Wang F, Sun Y, 2016, MicroRNA-16-1 inhibits tumor cell proliferation and induces apoptosis in A549 non-small cell lung carcinoma cells. Oncol Res 24(5):345–351 Sam S, Sam MR, Esmaeillou M, Safaralizadeh R, 2016, Effective targeting Survivin, caspase-3 and MicroRNA-16-1 expression by methyl-3-pentyl-6-methoxyprodigiosene triggers apoptosis in colorectal cancer stem-like cells. Pathol Oncol Res 22(4):715–723 Zhang C, Fang X, Li W, Shi Q, Wu L, Chen X, Huang Z, Wu P, Wang Z, Liao Z, 2014, Influence of recombinant lentiviral vector encoding miR-15a/16-1 in biological features of human nasopharyngeal carcinoma CNE-2Z cells. Cancer Biother Radiopharm 29(10):422–427 Li X, Ling N, Bai Y, DongW, Hui GZ, Liu D, Zhao J, Hu J, 2013, MiR-16-1 plays a role in reducing migration and invasion of glioma cells. Anat Rec, Hoboken, 296(3):427–432 Cai CK, Zhao GY, Tian LY, Liu L, Yan K, Ma YL, Ji ZW, Li XX, Han K, Gao J, Qiu XC, Fan QY, Yang TT, Ma BA, 2012, miR-15a and miR-16-1 downregulate CCND1 and induce apoptosis and cell cycle arrest in osteosarcoma. Oncol Rep 28(5):1764–1770 Wang T, Hou J, Li Z, Zheng Z, Wei J, Song D, Hu T, Wu Q, Yang JY, Cai JC, 2017, miR-15a-3p and miR-16-1-3p negatively regulate Twist1 to repress gastric cancer cell invasion and metastasis. Int J Biol Sci 13(1):122–134 Kang W, Tong JH, Lung RW, Dong Y, Zhao J, Liang Q, Zhang L, Pan Y, Yang W, Pang JC, Cheng AS, Yu J, To KF, 2015 Feb 22, Targeting of YAP1 by microRNA-15a and microRNA-16-1 exerts tumor suppressor function in gastricadenocarcinoma. Mol Cancer 14:52., DOI 10.1186/s12943-015-0323-3 Shang C, Guo Y, Zhang J, Huang B, 2016, Silence of long noncoding RNA UCA1 inhibits malignant proliferation and chemotherapy resistance to adriamycin in gastric cancer. Cancer Chemother Pharmacol 77(5):1061–1067 Lu C, Shan Z, Li C, Yang L, 2017, MiR-129 regulates cisplatinresistance in human gastric cancer cells by targeting P-gp. Biomed Pharmacother 86:450–456 CaoW,WeiW, Zhan Z, Xie Y, Xiao Q, 2016)MiR-1284modulates multidrug resistance of gastric cancer cells by targeting EIF4A1. Oncol Rep 5:2583–2591 Li H,Wang Z, Zhou X, Cheng Y, Xie Z,Manley JL, Feng Y, 2013, Far upstream element-binding protein 1 and RNA secondary structure both mediate second-step splicing repression. Proc Natl Acad Sci U S A 110:E2687–E2695 Olanich ME, Moss BL, Piwnica-Worms D, Townsend RR, Weber JD, 2011, Identification of FUSE-binding protein 1 as a regulatory mRNA-binding protein that represses nucleophosmin translation. Oncogene 30:77–86 Samarin J, LaketaV, Malz M, Roessler S, Stein I, Horwitz E, Singer S, Dimou E, Cigliano A, BissingerM, Falk CS, Chen X, Dooley S, Pikarsky E, Calvisi DF, Schultz C, Schirmacher P, Breuhahn K, 2016, PI3K/AKT/mTOR-dependent stabilization of oncogenic far-upstream element binding proteins in hepatocellular carcinoma cells. Hepatology 63(3):813–826 Sheng H, Ying L, Zheng L, Zhang D, Zhu C, Wu J, Feng J, Su D, 2015, Down expression of FBP1 is a negative prognostic factor for non-small-cell lung cancer. Cancer Investig 33(5):197–204 Liu ZH, Hu JL, Liang JZ, Zhou AJ, LiMZ, Yan SM, Zhang X, Gao S, Chen L, Zhong Q, Zeng MS, 2015, Far upstream elementbinding protein 1 is a prognostic biomarker and promotes nasopharyngeal carcinoma progression. Cell Death Dis 6:e1920 Hong Y, Shi Y, Shang C, Xue Y, Liu Y, 2016, Influence of far upstream element binding protein 1 gene on chemotherapy sensitivity in human U251 glioblastoma cells. Arch Med Sci 12(1):156– 162 Ding Z, Liu X, Liu Y, Zhang J, Huang X, Yang X, Yao L, Cui G, Wang D, 2015, Expression of far upstream element, FUSE, binding protein 1 in human glioma is correlated with c-Myc and cell proliferation. Mol Carcinog 54(5):405–415 Yang L, Zhu JY, Zhang JG, Bao BJ, Guan CQ, Yang XJ, Liu YH, Huang YJ, Ni RZ, Ji LL, 2016, Far upstream element-binding protein 1, FUBP1, is a potential c-Mycregulator in esophageal squamous cell carcinoma, ESCC, and its expression promotes ESCC progression. Tumour Biol 37(3):4115–4126 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 483 EP 488 DI 10.1007/s12253-017-0263-x PG 6 ER PT J AU da Silva Calixto, P Lopes, SO dos Santos Maia, M Herrero, STS Longui, AC Farias Melo, GC de Carvalho Filho, RI Soares, FL de Medeiros, CA Delatorre, P khayat, SA Burbano, RR Lima, ME AF da Silva Calixto, Poliane Lopes, Sergio Otavio dos Santos Maia, Mayara Herrero, Satomi Takeno Sylvia Longui, Alberto Carlos Farias Melo, Germoglio Cynthia de Carvalho Filho, Rodrigues Ivan Soares, Ferreira Leonardo de Medeiros, Correia Arnaldo Delatorre, Plinio khayat, Salim Andre Burbano, Rodriguez Rommel Lima, Moura Eleonidas TI Single-Nucleotide Polymorphisms of the MSH2 and MLH1 Genes, Potential Molecular Markers for Susceptibility to the Development of Basal Cell Carcinoma in the Brazilian Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Basal cell carcinoma; Mismatch repair; Single nucleotide polymorphism; DSASP; Genotyping; Molecular markers ID Basal cell carcinoma; Mismatch repair; Single nucleotide polymorphism; DSASP; Genotyping; Molecular markers AB Basal cell carcinoma - BCC is considered a multifactorial neoplasm involving genetic, epigenetic and environmental factors. Where UVB radiation is considered the main physical agent involved in BCC carcinogenesis. The Brazil and state of Paraiba are exposed to high levels of UVB rays. The mismatch repair - MMR is important DNA repair mechanisms to maintain replication fidelity. Therefore, single nucleotide polymorphisms (SNPs) in genes encoding proteins involved in MMR may be potential molecular markers of susceptibility to BCC. The objective of this study was to evaluate and describe for the first time the SNPs rs560246973, rs2303425 and rs565410865 and risk of developing BCC. The present study analyzed 100 samples of paraffinembedded tissue from patients with histopathological diagnosis of BCC and 100 control samples. The results were obtained by genotyping method, Dideoxy Unique Allele Specific – PCR (DSASP). The SNPs rs2303425 were not associated with Basal Cell Carcinoma. However, the SNPs rs560246973 and rs565410865 was shown to be associated with the development of BCC when compared to control samples (P < 0.0001). The SNPs rs565410865 was also statistical significance between the genotypes of and the age group (p = 0.0027) and tumor location (p = 0,0191). The result suggests that SNPs rs2303425 and rs565410865 are associated with susceptibility to the development of BCC in the Brazilian population and may be considered as potential molecular markers for BCC. C1 [da Silva Calixto, Poliane] Universidade Federal da Paraiba, Departamento de Biologia MolecularJoao Pessoa, PB, Brazil. [Lopes, Sergio Otavio] Clinica Dermatologica Santa Catarina, Departamento de DermatologiaJoao Pessoa, PB, Brazil. [dos Santos Maia, Mayara] Universidade Federal da Paraiba, Departamento de Biologia MolecularJoao Pessoa, PB, Brazil. [Herrero, Satomi Takeno Sylvia] Universidade Federal da Paraiba, Departamento de Biologia MolecularJoao Pessoa, PB, Brazil. [Longui, Alberto Carlos] Faculdade de Ciencias Medicas da Santa Casa de Sao Paulo, Programa de Pos-Graduacao em Ciencias da SaudeSao Paulo, SP, Brazil. [Farias Melo, Germoglio Cynthia] Universidade Federal da Paraiba, Departamento de Biologia MolecularJoao Pessoa, PB, Brazil. [de Carvalho Filho, Rodrigues Ivan] Laboratorio de Analises Medicas – UNILAB, Depatamento de PatologiaJoao Pessoa, PB, Brazil. [Soares, Ferreira Leonardo] Universidade Estadual da Paraiba, Departamento de FarmaciaCampina Grande, PB, Brazil. [de Medeiros, Correia Arnaldo] Universidade Federal da Paraiba, Departamento de MedicinaJoao Pessoa, PB, Brazil. [Delatorre, Plinio] Universidade Federal da Paraiba, Departamento de Biologia MolecularJoao Pessoa, PB, Brazil. [khayat, Salim Andre] Universidade Federal do Para, Instituto de Ciencias BiologicasBelem, PA, Brazil. [Burbano, Rodriguez Rommel] Universidade Federal do Para, Instituto de Ciencias BiologicasBelem, PA, Brazil. [Lima, Moura Eleonidas] Universidade Federal da Paraiba, Departamento de Biologia MolecularJoao Pessoa, PB, Brazil. RP Lima, ME (reprint author), Universidade Federal da Paraiba, Departamento de Biologia Molecular, Joao Pessoa, Brazil. EM eleonidas@pq.cnpq.br CR Nikolaou V, Stratigos AJ, Tsao H, 2012, Hereditary Nonmelanoma skin cancer. Semin Cutan Med Surg 31:204–210 DePry JL, Reed KB, Cook-Norris RH et al, 2011, Iatrogenic immunosuppression and cutaneous malignancy. Clin Dermatol 29: 602–613 Lichter MD, Karagas MR, Mott LA et al, 2000, Therapeutic ionizing radiation and the incidence of basal cell carcinoma and squamous cell carcinoma. Arch Dermatol 136:1007–1011 Noubissi FK, KimT, Kawahara TN et al, 2014, Role of CRD-BP in the growth of human basal cell carcinoma cells. J Investig Dermatol 134:1718–1724 Zink BS, 2014, Cancer de pele: a importancia do seu diagnostico, tratamento e prevencao Revista HUPE 13:76–83 Kim S, Misra A, 2007, SNP genotyping: technologies and biomedical applications. Annu Rev Biomed Eng 9:289–320 Dizdaroglu M, 2015, Oxidatively induced DNA damage and its repair in cancer. Mutat Res 763:212–245 Erie DA, Weninger KR, 2014, Single molecule studies of DNA mismatch repair. DNA repair 20:71–81 Mjelle R, Hegre SA, Aas PA et al, 2015, Cell cycle regulation of human DNA repair and chromatin remodeling genes. DNA repair 30:53–67 Reyes GX, Schmidt TT, Kolodner RD et al, 2015, New insights into the mechanism of DNA mismatch repair. Chromosoma., DOI 10.1007/s00412-015-0514-0 Smith CE, Mendillo ML, Bowen N et al, 2013, Dominant mutations in S. cerevisiae PMS1 identify the Mlh1-Pms1 endonuclease active site and an exonuclease 1-independent mismatch repair pathway. PLoS Genet., DOI 10.1371/journal.pgen.1003869 Stojic L, Brun R, Jiricny J, 2004, Mismatch repair and DNA damage signalling. DNA Repair 3:1091–1101 Han HJ, Maruyama M, Baba S et al, 1995, Genomic structure of human mismatch repair gene, hMLH1, and its mutation analysis in patients with hereditary non-polyposis colorectal cancer, HNPCC). Hum Mol Genet 4:237–242 Fukuhara S, Chang I, Mitsui Yet al, 2014, DNA mismatch repair gene MLH1 induces apoptosis in prostate cancer cells. Oncotarget 5:11297–11307 Fishel R, Lescoe MK, Rao MR et al, 1993, The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell 75:1027–1038 Kolodner RD, Hall NR, Lipford J et al, 1994, Structure of the human MSH2 locus and analysis of two Muir-Torre kindreds for msh2 mutations. Genomics 24:516–526 Xiao XQ, Gong WD, Wang SZ et al, 2012, Polymorphisms of mismatch repair gene hMLH1 and hMSH2 and risk of gastric cancer in a Chinese population. Oncol Lett 3:591–598 Sun MZ, Ju HX, Zhou ZWet al, 2014, Single nucleotide polymorphisms of DNA mismatch repair genes MSH2 and MLH1 confer susceptibility to esophageal cancer. Int J Clin Exp Med 7:2329– 2333 Smith TR, Levine EA, Freimanis RI et al, 2008, Polygenic model of DNA repair genetic polymorphisms in human breast cancer risk. Carcinogenesis., DOI 10.1093/carcin/bgn193 Mrkonjic M, Raptis S, Green RC et al, 2007)MSH2− 118T> C and MSH6− 159C> T promoter polymorphisms and the risk of colorectal cancer. Carcinogenesis 28:2575–2580 Slovakova P,Majerova L,Matakova Tet al, 2015, Mismatch repair Gene polymorphisms and association with lung cancer development. In: lung cancer and autoimmune disorders. Adv Exp Med Biol 833:15–22 Nishi R, Okuda Y, Watanabe E et al, 2005, Centrin 2 stimulates nucleotide excision repair by interacting with Xeroderma Pigmentosum group C protein. Mol Cell Biol., DOI 10.1128/MCB. 25.13.5664-5674 Lima EM, Lopes OS, Soares LF et al, 2015, Dideoxy single allelespecific PCR - DSASP new method to discrimination allelic. Braz Arch Biol Technol 58:414–420 Birch-Johansen F, Jensen A,Mortensen L et al, 2010, Trends in the incidence of nonmelanoma skin cancer in Denmark 1978–2007: rapid incidence increase among young Danish women. Int J Cancer., DOI 10.1002/ijc.25411 Smolarz B, Makowska M, Samulak D et al, 2015, Gly322Asp and Asn127Ser single nucleotide polymorphisms, SNPs, of hMSH2 mismatch repair gene and the risk of triple-negative breast cancer in polish women. Familial Cancer., DOI 10.1007/s10689-014-9746-z Srivastava K, Srivastava A, Mittal B, 2010, Polymorphisms in ERCC2, MSH2, and OGG1 DNA repair genes and gallbladder cancer risk in a population of northern India. Cancer 116:3160– 3169 Hsieh YC, Cho EC, Tu SH et al, 2017)MSH2 rs2303425 polymorphism is associated with early-onset breast cancer in Taiwan. Ann Surg Oncol., DOI 10.1245/s10434-016-5168-5 Dahlman-Wright K, Qiao Y, Jonsson P et al, 2012, Interplay between AP-1 and estrogen receptor α in regulating gene expression and proliferation networks in breast cancer cells. Carcinogenesis., DOI 10.1093/carcin/bgs223 Ru LeeW, Chen CC, Liu S et al, 2006, 17b-estradiol, E2, induces cdc25A Gene expression in breast cancer cells by genomic and non-genomic pathways. J Cell Biochem 99:209–220 Miyamoto T, Shiozawa T, Kashima H et al, 2006, Estrogen upregulates mismatch repair activity in normal and malignant endometrial glandular cells. Endocrinology 147:4863–4870 van der Klift HM, Jansen AM, van der Steenstraten N et al, 2015, Splicing analysis for exonic and intronic mismatch repair gene variants associated with lynch syndrome confirms high concordance between minigene assays and patient RNA analyses. Mol Genet Genomic Med., DOI 10.1002/mgg3.145 Wibom C, Sjostrom S, Henriksson R et al, 2012, DNA-repair gene variants are associated with glioblastoma survival. Acta Oncol., DOI 10.3109/0284186X.2011.616284 Ward AJ, Cooper TA, 2010, NIH public access. J Pathol 220:152– 163 Zahary MN, Kaur G, Abu Hassan MR et al, 2012, Germline mutation analysis of MLH1 and MSH2 in malaysian lynch syndrome patients. World J Gastroenterol., DOI 10.3748/wjg.v18.i8.814 Langeberg WJ, Kwon EM, Koopmeiners JS et al, 2010, Population-based study of the association of variants in mismatch repair genes with prostate cancer risk and outcomes. Cancer Epidemiol Biomark Prev., DOI 10.1158/1055-9965.EPI-09-0800 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 489 EP 496 DI 10.1007/s12253-017-0265-8 PG 8 ER PT J AU Stasikowska-Kanicka, O Wagrowska-Danilewicz, M Danilewicz, M AF Stasikowska-Kanicka, Olga Wagrowska-Danilewicz, Malgorzata Danilewicz, Marian TI Immunohistochemical Analysis of Foxp3+, CD4+, CD8+ Cell Infiltrates and PD-L1 in Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Treg; FoxP3; PD-L1; Oral cancer ID Treg; FoxP3; PD-L1; Oral cancer AB The immunoexpression of the PD-L1 and the number of immune infiltrating cells have been shown to be a significant prognostic factors in various human cancers. Immunohistochemical method was used to examine the immunoexpression of PD-L1 and number of Foxp3+, CD4+ , CD8+ cells in 78 cases of oral squamous cell carcinomas (OSCCs): with better prognosis - OSCCBP (n = 37), and with poorer prognosis - OSCCPP (n = 41), and 18 cases of normal mucosa as a control. The immunoexpression of PD-L1 and the mean number of Foxp3+ cells was significantly increased in OSCCPP group in comparison to OSCCBP and control groups. The mean number of CD4+ cells was significantly increased in OSCCPP group in comparison to OSCCBP and control groups. CD8+ cells were significantly more numerous in OSCCBP group in comparison to OSCCPP and control group. In both OSCCPP and OSCCBP groups there were positive significant correlations between number of Foxp3+ and CD4+ cells. We found positive correlations between the immunoexpression of PD-L1 and numbers of Foxp3+ cells, and negative correlation between the immunoexpression of PD-L1 and numbers of CD8+ cells in both OSCCPP and OSCCBP groups. We found also significant positive correlation between immunoexpression of PD-L1 and the number of CD4+ cells in OSCCPP group. In conclusion, our findings support the hypothesis of involvement of Tregs and PD-L1 in OSCC development and progression. C1 [Stasikowska-Kanicka, Olga] Medical University of Lodz, Department of Nephropathology, ul. Czechoslowacka 8/10, 92-216 Lodz, Poland. [Wagrowska-Danilewicz, Malgorzata] Medical University of Lodz, Department of Nephropathology, ul. Czechoslowacka 8/10, 92-216 Lodz, Poland. [Danilewicz, Marian] Medical University of Lodz, Department of PathologyLodz, Poland. RP Stasikowska-Kanicka, O (reprint author), Medical University of Lodz, Department of Nephropathology, 92-216 Lodz, Poland. EM olga.stasikowska@umed.lodz.pl CR Siegel R, Ma J, Zou Z et al, 2014, Cancer statistics 2014. CA Cancer J Clin 64:9–29 Byakodi R, Byakodi S, Hiremath S et al, 2012, Oral cancer in India: an epidemiologic and clinical review. J Community Health 37:316–319 Scully C, Bagan J, 2009, Oral squamous cell carcinoma overview. Oral Oncol 45:301–308 Facciabene A, Motz GT, Coukos G, 2012, T-regulatory cells: key players in tumor immune escape and angiogenesis. Cancer Res 72: 2162–2171 Wang RF, 2008, CD8+ regulatory T cells, their suppressive mechanisms, and regulation in cancer. Hum Immunol 69:811–814 Zhang S, Ke X, Zeng S, et al, 2015, Analysis of CD8+ Treg cells in patients with ovarian cancer: a possible mechanism for immune impairment. Cell Mol Immunol Sep;12(5):580–591 Chaudhry A, Rudra D, Treuting P et al, 2009, CD4 regulatory T cells control TH17 responses in a Stat3-dependent manner. Science 326(5955):98691 Linterman MA, Pierson W, Lee SK et al, 2011, Foxp3 follicular regulatory T cells control the germinal center response. Nat Med 17(8):975–982 Sakaguchi S, Miyara M, Costantino CM et al, 2010, FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol 10:490–500 Hori S, Nomura T, Sakaguchi S, 2003, Control of regulatory T cell development by the transcription factor Foxp3. Science 299:1057–1061 Von Boehmer H, 2005, Mechanisms of suppression by suppressor T cells. Nat Immunol 6(4):338–344 DiPaolo RJ, Glass DD, Bijwaard KE et al, 2005, CD4+CD25+ T cells prevent the development of organ-specific autoimmune disease by inhibiting the differentiation of autoreactive effector Tcells. J Immunol 175:7135–7142 Sakaguchi S, 2005, Naturally arising Foxp3-expressing CD25+ CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol 6:345–352 Oleinika K, Nibbs RJ, Graham GJ et al, 2013, Suppression, subversion and escape: the role of regulatory T cells in cancer progression. Clin Exp Immunol 171:36–45 Chen L, 2004, Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol 4(5):336–347 Park JJ, Omiya R, Matsumura Y et al, 2010, B7–H1/CD80 interaction is required for the induction and maintenance of peripheral Tcell tolerance. Blood 116(8):1291–1298 Sharpe AH, Wherry EJ, Ahmed R et al, 2007, The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol 8:239–245 Freeman GJ, Long AJ, Iwai Yet al, 2000, Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 192: 1027–1034 Hino R, Kabashima K,Kato Yet al, 2010, Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer 116(7):1757–1766 Shi SJ,Wang LJ, Wang GD et al, 2013, B7-H1 expression is associated with poor prognosis in colorectal carcinoma and regulates the proliferation and invasion of HCT116 colorectal cancer cells. PLoS One 8(10):e76012 Velcheti V, Schalper KA, Carvajal DE et al, 2014, Programmed death ligand-1 expression in non-small cell lung cancer. Lab Investig 94(1):107–116 Chen XL, Yuan SX, Chen C et al, 2009, Expression of B7-H1 protein in human pancreatic carcinoma tissues and its clinical significance. Chin J Cancer 28(12):1328–1332 Gao Q, Wang XY, Qiu SJ et al, 2009, Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin Cancer Res 15(3):971–979 Azuma T, Yao S, Zhu G et al, 2008, H1 is a ubiquitous antiapoptotic receptor on cancer cells. Blood 111(7):3635–3643 Teng MW, Ngiow SF, Ribas A et al, 2015, Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res 75:2139–2145 Barnes L, Everson JW, Reichart P, et al, 2005, World Health Organization classification of Tumours. Pathology and Genetics Head and Neck Tumours IARC Press Lyon 168–176 Drennan S, Stafford ND, Greenman J et al, 2013, Increased frequency and suppressive activity of CD127 Treg cells in the peripheral circulation of patients with head and neck squamous cell carcinoma are associated with advanced stage and nodal involvement. Immunology., DOI 10.1111/imm.12144 Schott AK, Pries R,Wollenberg B, 2010, Permanent up-regulation of regulatory T-lymphocytes in patients with head and neck cancer. Int J Mol Med 26:67–75 Liang YJ, Liu HC, Su YX et al, 2011, Foxp3 expressed by tongue squamous cell carcinoma cells correlates with clinicopathologic features and overall survival in tongue squamous cell carcinoma patients. Oral Oncol 47(7):566–570 Hinz S, Pagerols-Raluy L, Oberg HH et al, 2007, Foxp3 expression in pancreatic carcinoma cells as a novel mechanism of immune evasion in cancer. Cancer Res 67(17):8344–8350 Merlo A, Casalini P, Carcangiu ML et al, 2009, FOXP3 expression and overall survival in breast cancer. J Clin Oncol 27(11):1746–1752 Chen CX, Rowell WA, Thomas RM et al, 2006, Transcriptional regulation by Foxp3 is associated with direct promoter occupancy and modulation of histone acetylation. J Biol Chem 281:36828–36834 Hiraoka N, Onozato K, Kosuge T et al, 2006, Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clin Cancer Res 12:5423–5434 Kobayashi N, Hiraoka N, Yamagami Wet al, 2007, FOXP3+ regulatory T cells affect the development and progression of hepatocarcinogenesis. Clin Cancer Res 13:902–911 Bohling SD, Allison KH, 2008, Immunosuppressive regulatory T cells are associated with aggressive breast cancer phenotypes: a potential therapeutic target. Mod Pathol 21:1527–1532 Suzuki H, Chikazawa N, Tasaka Tet al, 2010, Intratumoral CD8(+, T/ FOXP3, +, cell ratio is a predictivemarker for survival in patientswith colorectal cancer. Cancer Immunol Immunother 59(5):653–661 MaMW, Medicherla RC, Qian M et al, 2012, Immune response in melanoma: an in-depth analysis of the primary tumor and corresponding sentinel lymph node. Mod Pathol 25:1000–1010 Kim M, Grimmig T, GrimmMet al, 2013, Expression of Foxp3 in colorectal cancer but not in Treg cells correlates with disease progression in patients with colorectal cancer. PLoS One 8(1):e53630 Salama P, Phillips M, Grieu F et al, 2009, Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol 27(2):186–192 Correale P, Rotundo MS, Del Vecchio MT et al, 2010, Regulatory, FoxP3+, T-cell tumor infiltration is a favorable prognostic factor in advanced colon cancer patients undergoing chemo or chemoimmunotherapy. J Immunother 33:435–441 Viglietta V, Baecher-Allan C,Weiner HL et al, 2004, Loss of functional suppression by CD4+CD25+ regulatory T cells in patients with multiple sclerosis. J Exp Med 199:971–979 WatanabeMAE, Oda JMM, AmaranteMK et al, 2010, Regulatory T cells and breast cancer: implications for immunopathogenesis. Cancer Metastasis Rev 29:569–579 Knutson KL, Dang Y, Lu H et al, 2006, IL-2 immunotoxin therapy modulates tumor-associated regulatory T cells and leads to lasting immune-mediated rejection of breast cancers in neu-transgenic mice. J Immunol 177:84–91 Zhu S, Lin J, Qiao G et al, 2015, Differential regulation and function of tumor-infiltrating T cells in different stages of breast cancer patients. Tumour Biol 36(10):7907–7913 Mellman I, Coukos G, Dranoff G, 2011, Cancer immunotherapy comes of age. Nature 480:480–489 Dhodapkar MV, Sznol M, Zhao B et al, 2014, Induction of antigen-specific immunity with a vaccine targeting NY-ESO- 1 to the dendritic cell receptor DEC-205. Sci Transl Med 6: 232–251 MiyaraM, Sakaguchi S, 2011, Human FoxP3(+)CD4(+, regulatory T cells: their knowns and unknowns. Immunol Cell Biol 89(3):346–351 Ohigashi Y, Sho M, Yamada Y et al, 2005, Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clin Cancer Res 11:2947–2953 Lyford-Pike S, Peng S, Young GD, et al, 2013, Evidence for a role of the PD-1:PD-L1 pathway in immune resistance of HPVassociated head and neck squamous cell carcinoma. Cancer Res 15;73(6):1733-1741 Hirano F, Kaneko K, Tamura H et al, 2005, Blockade of B7–H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res 65(3):1089–1096 Thompson ED, ZahurakM,Murphy A et al, 2016, Patterns of PD-L1 expression and CD8 T cell infiltration in gastric adenocarcinomas and associated immune stroma. Gut., DOI 10.1136/gutjnl-2015-310839 LinYM, SungWW, HsiehMJ, et al, 2015, High PD-L1 expression correlates with metastasis and poor prognosis in oral squamous cell carcinoma. PLoS One 12;10(11): e0142656., DOI 10.1371/journal. pone.0142656 Tokito T, Azuma K, Kawahara A et al, 2016, Predictive relevance of PD-L1 expression combined with CD8+ TIL density in stage III non-small cell lung cancer patients receiving concurrent chemoradiotherapy. Eur J Cancer 55:7–14 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 497 EP 505 DI 10.1007/s12253-017-0270-y PG 9 ER PT J AU Cheng, Y Ping, J Chen, J AF Cheng, Yang Ping, Jian Chen, Jianjie TI Identification of Potential Gene Network Associated with HCV-Related Hepatocellular Carcinoma Using Microarray Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatitis C virus; Hepatocelluar carcinoma; Kyoto encyclopedia of gene and genomes pathway; Weighted gene co-expression network analysis; Differentially expressed gene ID Hepatitis C virus; Hepatocelluar carcinoma; Kyoto encyclopedia of gene and genomes pathway; Weighted gene co-expression network analysis; Differentially expressed gene AB In order to identify potential specific gene networks of Hepatitis C virus (HCV) related hepatocellular carcinoma (HCC), weighted gene co-expression network analysis (WGCNA) was performed, which may provide an insight into the potential mechanism of the HCC development. HCV-related HCC and normal sample data were downloaded from GEO, T test of limma package was used to screen different expression genes (DEGs); KEGG pathway was used to analyze related biochemical pathways, and WGCNA was used to construct clustering trees and screen hub genes in the HCC-specific modules. A total of 1151 DEGs were authenticated between the HCC and normal liver tissue samples, including 433 upregulated and 718 downregulated genes. Among these genes, three specific modules of HCC were constructed, including Tan, Yellow and Cyan, but only Yellow module had a significant enrichment score in substance combination module with three hub genes: SLA2547, EFNA4 and MME. Although Tan and Cyan separately had four and three hub genes, but the bio-functions of them did not have significant enrichment scores (score < 2). SLA2547, EFNA4 and MME may play important roles in the substance combination of HCV-related HCC, so studying the function of this gene network may provide us a deeper understanding of HCV-related HCC. C1 [Cheng, Yang] Hospital for Infectious Diseases of Pudong New Area, Department of Liver Disease, No.46 Nong 3018, East Huaxia Road, 201299 Shanghai, China. [Ping, Jian] Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China. [Chen, Jianjie] Hospital for Infectious Diseases of Pudong New Area, Department of Liver Disease, No.46 Nong 3018, East Huaxia Road, 201299 Shanghai, China. RP Chen, J (reprint author), Hospital for Infectious Diseases of Pudong New Area, Department of Liver Disease, 201299 Shanghai, China. EM prchenjianjie@126.com CR Jin B, Wang W, Du G, Huang GZ, Han LT, Tang ZY et al, 2015, Identifying hub genes and dysregulated pathways in hepatocellular carcinoma. Eur Rev Med Pharmacol Sci 19:592–601 Luna J, Scheel TH, Danino T, Shaw K, Mele A, Fak J et al, 2015, Hepatitis C virus RNA functionally sequesters miR-122. Cell 160: 1099–1110 Castello G, Scala S, Palmieri G, Curley SA, Izzo F, 2010, HCV-related hepatocellular carcinoma: from chronic inflammation to cancer. Clin Immunol 134:237–250 Petrizzo A, Caruso FP, Tagliamonte M, Tornesello ML, Ceccarelli M, Costa V et al, 2016, Identification and validation of HCC-specific Gene transcriptional signature for tumor antigen discovery. Sci Rep 6:29258 Kakehashi A, Ishii N, Sugihara E, Min G, Saya H, Wanibuchi H, 2016, CD44 variant 9 is a potential biomarker of tumor initiating cells predicting survival outcome in hepatitis C virus-positive patients with resected hepatocellular carcinoma. Cancer Sci 107:609– 618 Boj S, Vanes J, Huch M, Li VW, Jose A, Hatzis P et al, 2012, Diabetes risk gene and Wnt effector Tcf7l2/TCF4 controls hepatic response to perinatal and adult metabolic demand. Cell 151:1595– 1607 Park SH, Rehermann B, 2014, Immune responses to HCV and other hepatitis viruses. Immunity 40:13–24 Syed GH, Amako Y, Siddiqui A, 2009, Hepatitis C virus hijacks host lipid metabolism. Trends Endocrinol Metab 21:33–40 Lambert MP, Paliwal A, Vaissiere T, Chemin I, Zoulim F, Tommasino M et al, 2011, Aberrant DNA methylation distinguishes hepatocellular carcinoma associated with HBV and HCV infection and alcohol intake. J Hepatol 54:705–715 Hodo Y, Honda M, Tanaka A, Nomura Y, Arai K, Yamashita Tet al, 2013, Association of interleukin-28B genotype and hepatocellular carcinoma recurrence in patients with chronic hepatitis C. Clin Cancer Res 19:1827–1837 Zhang J, Baddoo M,Han C, Strong MJ, Cvitanovic J, MorozK et al, 2016, Gene network analysis reveals a novel 22-gene signature of carbon metabolism in hepatocellular carcinoma. Oncotarget 7: 49232–49245 Xu Y, Cui J, Puett D, 2014, Understanding cancer invasion and metastasis. Springer, New York Ueda T, Honda M, Horimoto K, Aburatani S, Saito S, Yamashita T et al, 2013, Gene expression profiling of hepatitis B- and hepatitis C-related hepatocellular carcinoma using graphical Gaussian modeling ☆. Genomics 101:238–248 † SC, † PSS, Lee J, Park J, Shin EC, Choi C, 2015, Prolonged silencing of diacylglycerol acyltransferase-1 induces a dedifferentiated phenotype in human liver cells. J Cell Mol Med 20:38–47 Irizarry RA, Hobbs B, Collin F, Beazerbarclay YD, Antonellis KJ, Scherf U et al, 2003, Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4: 249–264 Diboun I, Wernisch L, Orengo CA, Koltzenburg M, 2006, Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma. BMC Genomics 7:1–14 Wixon J, Kell D, 2000, The Kyoto encyclopedia of genes and genomes–KEGG. Yeast 17:48–55 Huang DW, Sherman BT, Lempicki RA, 2009, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature Protocol 4:44–57 Langfelder P, Horvath S, 2008)WGCNA: an R package for weighted correlation network analysis. BMC Bioinf 9:1–13 Bushkofsky JR, Maguire M, Larsen MC, Fong YH, Jefcoate CR, 2016, Cyp1b1 affects external control of mouse hepatocytes, fatty acid homeostasis and signaling involving HNF4α and PPARα. Arch Biochem Biophys 597:30–47 Lamming DW, Demirkan G, Boylan JM, Mihaylova MM, Peng T, Ferreira J et al, 2014, Hepatic signaling by themechanistic target of rapamycin complex 2, mTORC2). Faseb Journal Official Publication of the Federation of American Societies for Experimental Biology 28:300–315 Archer KJ,MasVR,Maluf DG, Fisher RA(2010, High-throughput assessment of CpG site methylation for distinguishing between HCV-cirrhosis and HCV-associated hepatocellular carcinoma. Molecular Genetics & Genomics Mgg 283:341–349 Tong J, Xie J, Ren H, Liu J, Zhang W, Wei C et al, 2015, Comparison of glomerular Transcriptome profiles of adult-onset steroid sensitive focal segmental Glomerulosclerosis and minimal change disease. PLoS One 10:e0140453 Arvanitis DN, Davy A, 1900, Regulation and misregulation of Eph/ephrin expression. Cell Adhes Migr 6:131–137 Saintigny P, Peng S, Zhang L, Sen B,Wistuba II, Lippman SM et al, 2012, Global evaluation of Eph receptors and Ephrins in lung adenocarcinomas identifies EphA4 as an inhibitor of cell migration and invasion. Mol Cancer Ther 11:2021–2032 Abdou AG, Abd el-Wahed MM, Asaad NY, Samaka RM, Abdallaha R, 2010, Ephrin A4 expression in osteosarcoma, impact on prognosis, and patient outcome. Indian J Cancer 47:46–52 Hernaez R, 2011, Genetic factors associated with the presence and progression of nonalcoholic fatty liver disease: a narrative review. Gastroenterol Hepatol 35:32–41 Sanchez-Antolin G, Almohalla-Alvarez C, Bueno P, Almansa R, Iglesias V, Rico L et al, 2015, Evidence of active pro-fibrotic response in blood of patients with cirrhosis. PLoS One 10:e0137128 English DP, Santin AD, 2012, Claudins overexpression in ovarian cancer: potential targets for Clostridium perfringens enterotoxin, CPE, based diagnosis and therapy. Int J Mol Sci 14:10412–10437 Lin H, Liu W, Fang Z, Liang X, Li J, Bai Y et al, 2015, Overexpression of DHX32 contributes to the growth and metastasis of colorectal cancer. Sci Rep 5:9247 Kim KH, Lee MS, 2014, Autophagy - a key player in cellular and body metabolism. Nat Rev Endocrinol 10:322–337 Sun T, Yi H, Yang C, Kishnani PS, Sun B, 2016, Starch binding domain-containing protein 1 plays a dominant role in glycogen transport to lysosomes in liver. J Biol Chem 291:16479–16484 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 507 EP 514 DI 10.1007/s12253-017-0273-8 PG 8 ER PT J AU Ruiz-Sauri, A Garcia-Bustos, V Granero, E Cuesta, S Sales, AM Marcos, V LLombart-Bosch, A AF Ruiz-Sauri, Amparo Garcia-Bustos, Victor Granero, Eduardo Cuesta, Salome Sales, A Maria Marcos, Victor LLombart-Bosch, Antonio TI Distribution of Vascular Patterns in Different Subtypes of Renal Cell Carcinoma. A Morphometric Study in Two Distinct Types of Blood Vessels SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Clear cell renal cell carcinoma; Papillary renal cell carcinoma; Chromophobe renal cell carcinoma; Microvascular density; Microvascular area; Morphometry; CD31; CD34; Renal vascular patterns ID Clear cell renal cell carcinoma; Papillary renal cell carcinoma; Chromophobe renal cell carcinoma; Microvascular density; Microvascular area; Morphometry; CD31; CD34; Renal vascular patterns AB To analyze the presence of mature and immature vessels as a prognostic factor in patients with renal cell carcinoma and propose a classification of renal cancer tumor blood vessels according to morphometric parameters. Tissue samples were obtained from 121 renal cell carcinoma patients who underwent radical nephrectomy. Staining with CD31 and CD34 was used to differentiate between immature (CD31+) and mature (CD34+) blood vessels. We quantified the microvascular density, microvascular area and different morphometric parameters: maximum diameter, minimum diameter, major axis, minor axis, perimeter, radius ratio and roundness. We found that the microvascular density was higher in CD31+ than CD34+ vessels, but CD34+ vessels were larger than CD31+ vessels, as well as being strongly correlated with the ISUP tumor grade.We also identified four vascular patterns: pseudoacinar, fascicular, reticular and diffuse. Pseudoacinar and fascicular patterns were more frequent in clear cell renal cell carcinoma (37.62 and 35.64% respectively), followed by reticular pattern (21.78%), while in chromophobe tumors the reticular pattern predominated (90%). The isolated pattern was present in all papillary tumors (100%). In healthy renal tissue, the pseudoacinar and isolated patterns were differentially found in the renal cortex and medulla respectively. We defined four distinct vascular patterns significantly related with the ISUP tumor grade in renal cell carcinomas. Further studies in larger series are needed in order to validate these results. Analysis of both mature and immature vessels (CD34+ and CD31+) provides additional information when evaluating microvascular density. C1 [Ruiz-Sauri, Amparo] University of Valencia, Department of Pathology, Avda. Blasco Ibanez n° 15, 46010 Valencia, Spain. [Garcia-Bustos, Victor] University of Valencia, Department of Pathology, Avda. Blasco Ibanez n° 15, 46010 Valencia, Spain. [Granero, Eduardo] University of Valencia, Department of Pathology, Avda. Blasco Ibanez n° 15, 46010 Valencia, Spain. [Cuesta, Salome] University of Valencia, Department of Pathology, Avda. Blasco Ibanez n° 15, 46010 Valencia, Spain. [Sales, A Maria] University of Valencia, Department of PathologyValencia, Spain. [Marcos, Victor] University of Valencia, Department of Pathology, Avda. Blasco Ibanez n° 15, 46010 Valencia, Spain. [LLombart-Bosch, Antonio] University of Valencia, Department of Pathology, Avda. Blasco Ibanez n° 15, 46010 Valencia, Spain. RP Ruiz-Sauri, A (reprint author), University of Valencia, Department of Pathology, 46010 Valencia, Spain. EM Amparo.Ruiz-Sauri@uv.es CR Zhang B, Ji H, Yan D, Liu S, Shi B, 2014, Lack of association of microvessel density with prognosis of renal cell carcinoma: evidence from meta-analysis. Tumor Biol 35(3):2769–2776 Dome B, Hendrix MJC, Paku S, Tovari J, Timar J, 2007, Alternative vascularization mechanisms in cancer. Am J Pathol 170(1):1–15 Srigley JR, Delahunt B, 2009, Uncommon and recently described renal carcinomas. Mod Pathol 22(Suppl 2):S2–23 Murphy WM, Grignon DG, Perlman EJ, 2004, Tumors of the kidney, bladder, and related urinary structures. American Registry of Pathology, Washington Delahunt B, Eble JN, McCredie MR, Bethwaite PB, Stewart JH, Bilous AM, 2001, Morphologic typing of papillary renal cell carcinoma: comparison of growth kinetics and patient survival in 66 cases. Hum Pathol 32:590–595 Storkel S, Eble JN, Adlakha K et al, 1997, Classification of renal carcinoma. Cancer 80:987–989 Thoenes W, Storkel S, Rompelt HJ, Moll R, Baum HP, Werner S, 1988, Chromophobe cell renal carcinoma and its variants. A report in 32 cases. J. Pathol 155:277–287 Hlatky L, Hahnfeldt P, Folkman J, 2002, Clinical application of antiangiogenic therapy: microvessel density, what it does and doesn’t tell us. J Nat Cancer Inst 94:883–893 Grizzi F, Colombo P, Barbieri B et al, 2001, Microscopic analysis and significance of vascular architectural complexity in renal cell carcinoma. Clin Cancer Res 7:3305–3307 Weidner N, Semple JP, Welch WR, Folkman J, 1991, Tumor angiogenesis and metastasis: correlation in invasive breast carcinoma. N Engl JMed 324:1–8 Grizzi F, Ceva-Grimaldi G, Dioguardi N, 2001a, Fractal geometry: a useful tool for quantifying irregular lesions in human liver biopsy specimens. Ital J Anat Embryol 106:337–346 Kovacs G, Akhtar M, Beckwith BJ et al, 1997, The Heidelberg classification of renal cell tumours. J Pathol 80:992–993 Ljungberg B, Bensalah K, Canfield S, Dabestani S, Hofmann F, Hora M, et al, 20015). EAU Guidelines on Renal Cell Carcinoma 2014 Update. Eur Urol 67(5): 913–924 Delahunt B, Srigley JR,Montironi R, Egevad L, 2014)Advances in renal neoplasia: recommendations from the 2012 International Society of Urological Pathology Consensus Conference. Urology 83(5):969–974 Poblet E, Gonzalez-Palacios F, Jimenez FJ, 1996, Different immunoreactivity of endotelial markers in well and poorly differentiated areas of angiosarcomas. Virchows Arch 428:217–221 Yilmazer D, Han U, Onal B, 2007, A comparison of the vascular density of VEGF expression withmicrovascular density determined with CD34 and CD31 staining and conventional prognostic markers in renal cell carcinoma. Int Urol Nephrol 39:691–698 Meert AP, Paesmans M, Martin B, Delmotte P, Berghmans T, Verdebout JM, Lafitte JJ, Mascaux C, Sculier JP, 2002, The role of microvessel density on the survival of patients with lung cancer: a systematic review of the literature with meta-analysis. Br J Cancer 87:694–701 Uzzan B, Nicolas P, Cucherat M, Perret GY, 2004, Microvessel density as a prognostic factor in women with breast cancer: a systematic review of the literature and meta-analysis. Cancer Res 64: 2941–2955 Des Guetz G, Uzzan B, Nicolas P, Cucherat M, Morere JF, Benamouzig R, Breau JL, Perret GY, 2006, Microvessel density and VEGF expression are prognostic factors in colorectal cancer. Meta-analysis of the literature. Br J Cancer 94:1823–1832 Ruiz-Sauri A, Valencia-Villa G, Romanenko A, Perez J, Garcia R, Garcia H, Benavent J, Sancho-Tello M, Carda C, Llombart-Bosch A, 2016, Influence of exposure to chronic persistent low-dose ionizing radiation on the tumor biology of clear-cell renal-cell carcinoma. An Immunohistochemical and morphometric study of angiogenesis and vascular related factors. Pathol Oncol Res 22(4):807– 815 Joo HJ, Oh DK, Kim YS, Lee KB, Kim SJ, 2004, Increased expression of caveolin-1 and microvessel density correlates with metastasis and poor prognosis in clear cell renal cell carcinoma. BJU Int 93:291–296 Nativ O, Sabo E, Reiss A,Wald M, Madjar S, Moskovitz B, 1998, Clinical significance of tumor angiogenesis in patients with localized renal cell carcinoma. Urology 51:693–696 Yoshino S, Kato M, Okada K, 1995, Prognostic significance of microvessel count in low stage renal cell carcinoma. Int J Urol 2: 156–160 MacLennan GT, Bostwick DG, 1995, Microvessel density in renal cell carcinoma: lack of prognostic significance. Urology 46:27–30 Minardi D, Lucarini G, Mazzucchelli R, Milanese G, Natali D, Galosi AB, Montironi R, Biagini G, Muzzonigro G, 2005, Prognostic role of Fuhrman grade and vascular endothelial growth factor in pT1a clear cell carcinoma in partial nephrectomy specimens. J Urol 174:1208–1212 Anastassiou G, Duensing S, Steinhoff G, Zorn U, Grosse J, Dallmann I, Kirchner H, Ganser A, Atzpodien J, 1996, Platelet endothelial cell adhesionmolecule-1, PECAM-1): a potential prognostic marker involved in leukocyte infiltration of renal cell carcinoma. Oncology 53:127–132 Imao T, Egawa M, Takashima H, Koshida K, Namiki M, 2004, Inverse correlation ofmicrovessel density with metastasis and prognosis in renal cell carcinoma. Int J Urol 11:948–953 Rioux-Leclercq N, Epstein JI, Bansard JY, Turlin B, Patard JJ, Manunta A, Chan T, Ramee MP, Lobel B, Moulinoux JP, 2001, Clinical significance of cell proliferation, microvessel density, and CD44 adhesion molecule expression in renal cell carcinoma. Hum Pathol 32:1209–1215 Sabo E, Boltenko A, Sova Y, Stein A, Kleinhaus S, Resnick MB, 2001, Microscopic analysis and significance of vascular architectural complexity in renal cell carcinoma. Clin Cancer Res 7:533– 537 Schraml P, Struckmann K, Hatz F, Sonnet S, Kully C, Gasser T, Sauter G, Mihatsch MJ, Moch H, 2002, VHL mutations and their correlation with tumour cell proliferation, microvessel density, and patient prognosis in clear cell renal cell carcinoma. J Pathol 196: 186–193 Cheng S-H, Liu J-M, Liu Q-Y, Luo D-Y, Liao B-.H, Li H,Wang KJ, 2014). Prognostic role of microvessel density in patients with renal cell carcinoma: a meta-analysis. Int J Clin Exp Pathol 7(9): 5855–5863 Ferician O, Cimpean AM, Ceasu AM, Dema A, Raica M, Cumpanas A, 2016, Heterogeneous vascular patterns in renal cell carcinomas. Pol J Pathol 67(1):46–53 Joshi S, Singh AR, Durden DL, 2015, Pan-PI-3 kinase inhibitor SF 1126 shows antitumor and antiangiogenic activity in renal cell carcinoma. Cancer Chemother Pharmacol 75:595–608 Schirner M, Hoffmann J, Menrad A, Schneider MR, 1998, Antiangiogenic chemotherapeutic agents: characterization in comparison to their tumor growth inhibition in human renal cell carcinoma models. Clin Cancer Res 4:1331–1336 Travnicek I, Branzovsky H, Kalusova K, Hess O, Holubec L, Pele KB, Urge T, Hora M, 2015, Tissue biomarkers in predicting response to sunitinib treatment of metastatic renal cell carcinoma. Anticancer Res 35:5661–5666 Porta C, Giglione P, Liguigli W, Paglino C, 2015, Dovitinib, CHIR258, TKI258): structure, development and preclinical and clinical activity. Future Oncol 11:39–50 Okon K, Kawa R, 2008, Microvascular network in renal carcinomas. Quantitative and tissue microarray immunohistochemical study. Pol J Pathol 59:107–115 Tinini T, Rossi F, Claudio PP, 2003, Molecular basis of angiogenesis and cancer. Oncogene 22:6549–6556 Eberhard A, Kahlert S, Goede V, Hemmerlein B, Plate KH, Augustin HG, 2000, Heterogeneity of angiogenesis and blood vessel maturation in human tumors: implications for antiangiogenic tumor therapies. Cancer Res 60:1388–1393 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 515 EP 524 DI 10.1007/s12253-017-0262-y PG 10 ER PT J AU Rabassa, EM Pereyra, A Pereyra, L Segal-Eiras, A Abba, CM Croce, VM AF Rabassa, E Martin Pereyra, Adrian Pereyra, Liliana Segal-Eiras, Amada Abba, C Martin Croce, V Maria TI Lewis x Antigen is Associated to Head and Neck Squamous Cell Carcinoma Survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Antigenic expression; Head and neck squamous cell carcinoma; Risk and prognostic factors; Survival ID Antigenic expression; Head and neck squamous cell carcinoma; Risk and prognostic factors; Survival AB Head and neck squamous cell carcinoma (HNSCC) is an aggressive disease with poor prognosis without appropriate prognostic markers. Previous research shows that Lewis antigens have been involved in carcinoma dissemination and patients´ survival. Fucosyl and sialyltransferases are the enzymes implicated in the Lewis antigens synthesis. The purpose of this study was to evaluate the prognostic utility of Lewis antigens in HNSCC. We conducted a prospective research including histological samples from 79 patients with primary HNSCC. Lewis x and sialyl Lewis x expression were detected by immunohistochemistry; patient’s data, progression free, and overall survival were documented. A statistical correlation study of antigenic expression and patients´ histopathological variables was performed. Cox regression models with internal validation procedures were employed to analyze survival data. By immunohistochemistry, Lewis x was detected in 34/79 (43%) tumor samples, while sialyl Lewis x only in 11/79 (14%). Lewis x expression showed a positive correlation with tumor differentiation and a better overall survival for Lewis x + patients was detected. Moreover, multivariate Cox’s regression analysis showed that Lewis x is an independent predictor of better overall survival. The in silico analysis supported the presence of deregulated fucosyl (FUT4) and sialyltransferase (ST3GAL4) in the Lewis synthetic pathway related to patient survival. These results suggest that Lewis x expression is associated with a better outcome in patients with HNSCC. C1 [Rabassa, E Martin] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA)La Plata, Argentina. [Pereyra, Adrian] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA)La Plata, Argentina. [Pereyra, Liliana] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA)La Plata, Argentina. [Segal-Eiras, Amada] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA)La Plata, Argentina. [Abba, C Martin] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA)La Plata, Argentina. [Croce, V Maria] Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA)La Plata, Argentina. RP Croce, VM (reprint author), Facultad de Ciencias Medicas, UNLP, Centro de Investigaciones Inmunologicas Basicas y Aplicadas (CINIBA), La Plata, Argentina. EM crocevir@hotmail.com CR Ferlay J, Soerjomataram I, Ervik M, et al, 2013, Cancer incidence and mortality worldwide: IARC CancerBase no. 11. International Agency for Research on Cancer, Lyon. Accessed 15 June 2015 Hashibe M, Brennan P, Chuang S-C et al, 2009, Interaction between tobacco and alcohol use and the risk of head and neck cancer: pooled analysis in the International head and neck cancer epidemiology consortium. Cancer Epidemiol Biomark Prev 18:541–550 Herrero R, Castellsague X, Pawlita M et al, 2003, Human papillomavirus and oral cancer: the International Agency for Research on Cancer multicenter study. J Natl Cancer Inst 95:1772–1783 Tiwana MS,Wu J, Hay J,Wong F, CheungW, Olson RA, 2014, 25 year survival outcomes for squamous cell carcinomas of the head and neck: population-based outcomes from a Canadian province. Oral Oncol 50:651–656 Cerezo L, Millan I, Torre A, Aragon G, Otero J, 1992, Prognostic factors for survival and tumor control in cervical lymph node metastases from head and neck cancer: a multivariate study of 492 cases. Cancer 69:1224–1234 Mandel U, 1992, Carbohydrates in oral epithelia and secretions: variation with cellular differentiation. APMIS Suppl 27:119–129 Hakomori S, 1989, Aberrant glycosylation in tumors and tumorassociated carbohydrate antigens. Adv Cancer Res 52:257–331 Dabelsteen E, Gao S, 2005, ABO blood-group antigens in oral cancer. J Dent Res 84:21–28 Takada A, Ohmori K, Yoneda T et al, 1993, Contribution of carbohydrate antigens sialyl Lewis a and sialyl Lewis X to adhesion of human cancer cells to vascular endothelium. Cancer Res 53:354– 361 Remmers N, Anderson JM, Linde EMet al, 2013, Aberrant expression of mucin core proteins and o-linked glycans associated with progression of pancreatic cancer. Clin Cancer Res 19:1981–1993 Croce MV, Isla-Larrain MT, Rua CE, Rabassa ME, Gendler SJ, Segal-Eiras A, 2003, Patterns of MUC1 tissue expression defined by an anti-MUC1 cytoplasmic tail monoclonal antibody in breast cancer. J Histochem Cytochem 51:781–788 University of California Santa Cruz, Xena. http://xena.ucsc.edu/ 2015. Accessed 15 June 2015 Croce MV, Rabassa ME, Pereyra A, Segal-Eiras A, 2008, Differential expression of MUC1 and carbohydrate antigens in primary and secondary head and neck squamous cell carcinoma. Head Neck 30:647–657 Rabassa ME, Croce MV, Pereyra A, Segal-Eiras A, 2006, MUC1 expression and anti-MUC1 serum immune response in head and neck squamous cell carcinoma, HNSCC): a multivariate analysis. BMC Cancer 6:253 CroceMV, RabassaME, Pereyra A, Segal-EirasA, 2005, Influence of sialic acid removal on MUC1 antigenic reactivity in head and neck carcinoma. Pathol Oncol Res 11:74–81 Croce MV, Rabassa ME, Price MR, Segal-Eiras A, 2001, MUC1 mucin and carbohydrate associated antigens as tumor markers in head and neck squamous cell carcinoma. Pathol Oncol Res England 7:284–291 Farmer RW, Richtsmeier WJ, Scher RL, 1998, Identification of sialyl Lewis-x in squamous cell carcinoma of the head and neck. Head Neck 20:726–731 Kurahara SI, ShinoharaM, Ikebe T et al, 1999, Immunohistochemical study of sialyl lea and sialyl le(x, antigen in oral squamous cell carcinoma: the association of sialyl lea expression with the metastatic potential. Head Neck 21:330–337 Shah MH, Sainger RN, Telang SD, Pancholi GH, Shukla SN, Patel PS, 2009, E-cadherin truncation and sialyl Lewis-X overexpression in oral squamous cell carcinoma and oral precancerous conditions. Neoplasma 56:40–47 Gunawardena I, Arendse M, Jameson MB, Plank LD, Gregor RT, 2015, Prognostic molecular markers in head and neck squamous cell carcinoma in a New Zealand population: matrix metalloproteinase-2 and sialyl Lewis x antigen. ANZ J Surg 85: 843–848 Teng YT, Nadimi H, Toto PD, 1989, Immunohistochemical localization of Leu-M1 carbohydrate antigen in human oral squamous cell carcinoma. J Oral Pathol Med 18:502–505 Hakomori SI, 1992, Lex and related structures as adhesion molecules. Histochem J 24:771–776 Liu N, Jin C, Zhu ZM et al, 1999, Stage-specific expression of alpha1,2-fucosyltransferase and alpha1, 3-fucosyltransferase, FT, during mouse embryogenesis. Eur J Biochem 265:258–263 Croce MV, Isla-Larrain M, Rabassa ME, Demichelis S, Colussi AG, Crespo M et al, 2007, Lewis x is highly expressed in normal tissues: a comparative immunohistochemical study and literature revision. Pathol Oncol Res 13:130–138 Ball ED, Schwarz LM, Bloomfield CD, 1991, Expression of the CD15 antigen on normal and leukemic myeloid cells: effects of neuraminidase and variable detection with a panel of monoclonal antibodies. Mol Immunol 28(9):951–958 Koh YW, Lee HJ, Ahn J-H, Lee JW, Gong G, 2013, Expression of Lewis X is associated with poor prognosis in triple-negative breast cancer. Am J Clin Pathol 139:746–753 Derolf AR, Bjorklund E, Mazur J, Bjorkholm M, Porwit A, 2008, Expression patterns of CD33 and CD15 predict outcome in patients with acute myeloid leukemia. Leuk Lymphoma 49:1279–1291 Altman DG, McShane LM, Sauerbrei W, Taube SE, 2012, Reporting recommendations for tumor marker prognostic studies, REMARK): explanation and elaboration. PLoS Med 9(5): e1001216 Azuma Y, Ito M, Taniguchi A, Matsumoto K, 2004, Expression of cell surface Lewis X and Yantigens and FUT4 mRNA is increased in Jurkat cells undergoing apoptosis. Biochim Biophys Acta, Gen Subj 1672:157–163 Chen H, Yuan B, Zheng Z, Liu Z, Wang S, 2011, Lewis X Oligosaccharides-heparanase complex targeting to DCs enhance antitumor response in mice. Cell Immunol 269:144–148 Milde-Langosch K, Schutze D, Oliveira-Ferrer L et al, 2015, Relevance of βGal-βGalNAc-containing glycans and the enzymes involved in their synthesis for invasion and survival in breast cancer patients. Breast Cancer Res Treat 151:515–528 Jin Y, Hoglund M, Jin C et al, 1998, FISH characterization of head and neck carcinomas reveals that amplification of band 11q13 is associated with deletion of distal 11q. Genes Chromosom Cancer 22:312–320 Mondal N, Buffone A, Stolfa G et al, 2015, ST3Gal-4 is the primary sialyltransferase regulating the synthesis of E-, P-, and Lselectin ligands on human myeloid leukocytes. Blood 125:687–696 Ito H, Hiraiwa N, Sawada-Kasugai M et al, 1997, Altered mRNA expression of specific molecular species of fucosyl- and sialyltransferases in human colorectal cancer tissues. Int J Cancer 71: 556–564 Kudo T, Ikehara Y, Togayachi A et al, 1998, Up-regulation of a set of glycosyltransferase genes in human colorectal cancer. Lab Investig 78:797–811 Ma B, Simala-Grant JL, Taylor DE, 2006, Fucosylation in prokaryotes and eukaryotes. Glycobiology 16:158R–184R Cailleau-Thomas A, Coullin P, Candelier JJ et al, 2000, FUT4 and FUT9 genes are expressed early in human embryogenesis. Glycobiology 10:789–802 Taniguchi A, Hasegawa Y, Higai K, Matsumoto K, 2000, Transcriptional regulation of human beta-galactoside alpha2, 6- sialyltransferase, hST6Gal I, gene during differentiation of the HL-60 cell line. Glycobiology 10:623–628 Julien S, Ivetic A, Grigoriadis A et al, 2011, Selectin ligand sialyl- Lewis x antigen drives metastasis of hormone-dependent breast cancers. Cancer Res 71:7683–7693 Zhang Z, Sun P, Liu J et al, 2008, Suppression of FUT1/FUT4 expression by siRNA inhibits tumor growth. Biochim Biophys Acta, Mol Cell Res 1783:287–296 Liu Y-C, Yen H-Y, Chen C-Y et al, 2011, Sialylation and fucosylation of epidermal growth factor receptor suppress its dimerization and activation in lung cancer cells. Proc Natl Acad Sci U S A 108:11332–11337 Roseman S, 2001, Reflections on Glycobiology J Biol Chem 276: 41527–41542 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 525 EP 531 DI 10.1007/s12253-017-0269-4 PG 7 ER PT J AU Filho, RSP Junior, AS Begnami, DM Ferreira, dOF Nakagawa, TW Spencer, MSBR Bezerra, ST Boggiss, EP Lopes, A AF Filho, Roberto Stevanato Paulo Junior, Aguiar Samuel Begnami, Dirlei Maria Ferreira, de Oliveira Fabio Nakagawa, Toshihiko Wilson Spencer, Matheus Sobreira Batista Ranyell Bezerra, Santoro Tiago Boggiss, Edward Philip Lopes, Ademar TI Estrogen Receptor β as a Prognostic Marker of Tumor Progression in Colorectal Cancer with Familial Adenomatous Polyposis and Sporadic Polyps SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Survival; Estrogen receptor; Familial adenomatous polyposis; Hormonal receptors; Colorectal carcinogenesis ID Colorectal cancer; Survival; Estrogen receptor; Familial adenomatous polyposis; Hormonal receptors; Colorectal carcinogenesis AB The incidence of colorectal cancer (CRC) is lower in women than in men, and sex steroids can be considered contributing factors because oral contraception usage and estrogen replacement therapy are associated with decreased risk. Conversely, colorectal polyp development in familial adenomatous polyposis (FAP) begins during puberty. The objectives were to evaluate the relationship between the expression of these hormone receptors and adenoma-carcinoma progression, CRC stage and overall survival.We studied 120 A.C. Camargo Cancer Center patients diagnosed with either FAP-associated or spontaneous adenomatous polyps or CRC to determine the immunohistochemical expression levels of estrogen receptor (ER)-α, ER-β and the progesterone and androgen receptors (480 analyses). The ER-β expression levels differed between the groups: the group with FAP polyps had lower ER-β expression than that of the sporadic polyp group.With transformation of the sporadic polyps to cancer, there was a considerable decrease in ER-β expression (from 90% with strong expression to 80% with absent or weak expression) (p < 0.001). The ER-β expression was lower in T3/T4 tumors than in T1/T2 tumors (p = 0.015). The 5-year overall survival of CRC patients positively expressing ER-β exceeded that of patients without detectable expression levels (74.8% vs. 44.3%, respectively; p = 0.035). There was no significant expression of the androgen or progesterone receptor or ER-α among the groups. Differences in ER-β expression represent a potential mechanism through which estrogen might alter the susceptibility to colon cancer, thereby confirming the possibility of a protective role of estrogen against colorectal carcinogenesis. C1 [Filho, Roberto Stevanato Paulo] A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery Department, R. Professor Antonio Prudente, 211 Liberdade, 01509-010 Sao Paulo, SP, Brazil. [Junior, Aguiar Samuel] A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery Department, R. Professor Antonio Prudente, 211 Liberdade, 01509-010 Sao Paulo, SP, Brazil. [Begnami, Dirlei Maria] A. C. Camargo Cancer Hospital, Department of PathologySao Paulo, SP, Brazil. [Ferreira, de Oliveira Fabio] A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery Department, R. Professor Antonio Prudente, 211 Liberdade, 01509-010 Sao Paulo, SP, Brazil. [Nakagawa, Toshihiko Wilson] A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery Department, R. Professor Antonio Prudente, 211 Liberdade, 01509-010 Sao Paulo, SP, Brazil. [Spencer, Matheus Sobreira Batista Ranyell] A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery Department, R. Professor Antonio Prudente, 211 Liberdade, 01509-010 Sao Paulo, SP, Brazil. [Bezerra, Santoro Tiago] A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery Department, R. Professor Antonio Prudente, 211 Liberdade, 01509-010 Sao Paulo, SP, Brazil. [Boggiss, Edward Philip] A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery Department, R. Professor Antonio Prudente, 211 Liberdade, 01509-010 Sao Paulo, SP, Brazil. [Lopes, Ademar] A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery Department, R. Professor Antonio Prudente, 211 Liberdade, 01509-010 Sao Paulo, SP, Brazil. RP Filho, RSP (reprint author), A.C. Camargo Cancer Center, Colorectal Tumor Nucleus of the Pelvic Surgery Department, 01509-010 Sao Paulo, Brazil. EM paulo.stevanato@accamargo.org.br CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90 Gebert JF, Dupon C, Kadmon M, HahnM, Herfarth C, von Knebel DM, Schackert HK, 1999, Combined molecular and clinical approaches for the identification of families with familial adenomatous polyposis coli. Ann Surg 229:350–361., DOI 10.1097/ 00000658-199903000-00008 Goss KH, Groden J, 2000, Biology of the adenomatous polyposis coli tumor suppressor. J Clin Oncol 18:1967–1979., DOI 10.1200/ JCO.2000.18.9.1967 Wong N, Lasko D, Rabelo R, Pinsky L, Gordon PH, Foulkes W, 2001, Genetic counseling and interpretation of genetic tests in familial adenomatous polyposis and hereditary nonpolyposis colorectal cancer. Dis Colon rectum 44:271–279., DOI 10.1007/ BF02234304 Vasen HF, Moslein G, Alonso A, Aretz S, Bernstein I, Bertario L, Blanco I, Bulow S, Burn J, Capella G, Colas C, Engel C, Frayling I, Friedl W, Hes FJ, Hodgson S, Jarvinen H, Mecklin JP, Moller P, Myrhoi T, Nagengast FM, Parc Y, Phillips R, Clark SK, de Leon MP, Renkonen-Sinisalo L, Sampson JR, Stormorken A, Tejpar S, Thomas HJ, Wijnen J, 2008, Guidelines for the clinical management of familial adenomatous polyposis, FAP). Gut 57:704–713., DOI 10.1136/gut.2007.136127 NieuwenhuisMH,Vasen HF, 2007, Correlations between mutation site in APC and phenotype of familial adenomatous polyposis, FAP): a review of the literature. Crit Rev Oncol Hematol 61: 153–161., DOI 10.1016/j.critrevonc.2006.07.004 Kennelly R, Kavanagh DO, Hogan AM, Winter DC, 2008, Oestrogen and the colon: potential mechanisms for cancer prevention. Lancet Oncol 9:385–391., DOI 10.1016/S1470-2045(08, 70100-1 Giardiello FM, Hylind LM, Trimbath JD, Hamilton SR, Romans KE, Cruz-Correa M, Corretti MC, Offerhaus JA, Yang VW, 2005, Oral contraceptives and polyp regression in familial adenomatous polyposis. Gastroenterology 128:1077–1080 Chlebowski RT, Wactawski-Wende J, Ritenbaugh C, Hubbell FA, Ascensao J, Rodabough RJ, Rosenberg CA, Taylor VM, Harris R, Chen C, Adams-Campbell LL, White E, Investigators W's HI, 2004, Estrogen plus progestin and colorectal cancer in postmenopausal women. N Engl J Med 350:991–1004., DOI 10.1056/ NEJMoa032071 Simon MS, Chlebowski RT, Wactawski-Wende J, Johnson KC, Muskovitz A, Kato I, Young A, Hubbell FA, Prentice RL, 2012, Estrogen plus progestin and colorectal cancer incidence and mortality. J Clin Oncol 30:3983–3990., DOI 10.1200/JCO.2012.42.7732 Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA, Black H, Bonds D, Brunner R, Brzyski R, Caan B, Chlebowski R, Curb D, Gass M, Hays J, Heiss G, Hendrix S, Howard BV, Hsia J, Hubbell A, Jackson R, Johnson KC, Judd H, Kotchen JM, Kuller L, LaCroix AZ, Lane D, Langer RD, Lasser N, Lewis CE, Manson J, Margolis K, Ockene J, O’Sullivan MJ, Phillips L, Prentice RL, Ritenbaugh C, Robbins J, Rossouw JE, Sarto G, Stefanick ML, 2004, Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA 291:1701–1712., DOI 10.1001/jama. 291.14.1701 Rennert G, Rennert HS, Pinchev M, Lavie O, Gruber SB, 2009, Use of hormone replacement therapy and the risk of colorectal cancer. J Clin Oncol 27:4542–4547., DOI 10.1200/JCO.2009.22. 0764 Green J, Czanner G, ReevesG,Watson J,Wise L, RoddamA, Beral V, 2012, Menopausal hormone therapy and risk of gastrointestinal cancer: nested case-control study within a prospective cohort, and meta-analysis. Int J Cancer 130:2387–2396., DOI 10.1002/ijc.26236 Lin KJ, Cheung WY, Lai JY, Giovannucci EL, 2012, The effect of estrogen vs. combined estrogen-progestogen therapy on the risk of colorectal cancer. Int J Cancer 130:419–430., DOI 10.1002/ijc.26026 Grodstein F, Newcomb PA, Stampfer MJ, 1999, Postmenopausal hormone therapy and the risk of colorectal cancer: a review and meta-analysis. Am J Med 106:574–582., DOI 10.1016/S0002- 9343(99)00063-7 Kouzmenko AP, Takeyama K, Kawasaki Y, Akiyama T, Kato S, 2008, Ligand-dependent interaction between estrogen receptor alpha and adenomatous polyposis coli. Genes Cells 13:723–730., DOI 10.1111/j.1365-2443.2008.01200.x Dawson PM, Shousha S, Blair SD, Carter GD, Jones J, Alaghband- Zadeh J, Theodorou NA, 1990, Oestrogen receptors in colorectal carcinoma. J Clin Pathol 43:149–151., DOI 10.1136/jcp.43.2.149 Hendrickse CW, Jones CE, Donovan IA, Neoptolemos JP, Baker PR, 1993, Oestrogen and progesterone receptors in colorectal cancer and human colonic cancer cell lines. Br J Surg 80:636–640., DOI 10.1002/bjs.1800800531 Di Leo A, Messa C, Russo F,Misciagna G, Guerra V, Taveri R, Leo S, 1994, Prognostic value of cytosolic estrogen receptors in human colorectal carcinoma and surrounding mucosa. Preliminary results. Dig Dis Sci 39:2038–2042., DOI 10.1007/BF02088144 Meggouh F, Lointier P, Pezet D, Saez S, 1991, Status of sex steroid hormone receptors in large bowel cancer. Cancer 67:1964–1970., DOI 10.1002/1097-0142(19910401)67:7<1964::AIDCNCR2820670724> 3.0.CO;2-S Slattery ML, Sweeney C, Murtaugh M, Ma KN, Caan BJ, Potter JD, Wolff R, 2006, Associations between vitamin D, vitamin D receptor gene and the androgen receptor gene with colon and rectal cancer. Int J Cancer 118:3140–3146., DOI 10.1002/ijc.21791 Slattery ML, Sweeney C, Murtaugh M, Ma KN, Wolff RK, Potter JD, Caan BJ, Samowitz W, 2005, Associations between ERalpha, ERbeta, and AR genotypes and colon and rectal cancer. Cancer Epidemiol Biomark Prev 14:2936–2942., DOI 10.1158/1055-9965. EPI-05-0514 Mostafaie N, Kallay E, Sauerzapf E, Bonner E, Kriwanek S, Cross HS, Huber KR, KruglugerW, 2009, Correlated downregulation of estrogen receptor beta and the circadian clock gene Per1 in human colorectal cancer. Mol Carcinog 48:642–647., DOI 10.1002/mc. 20510 Ruschoff J, DietelM, BarettonG, Arbogast S,Walch A, Monges G, ChenardMP, Penault-Llorca F, Nagelmeier I, SchlakeW, Hofler H, Kreipe HH, 2010, HER2 diagnostics in gastric cancer-guideline validation and development of standardized immunohistochemical testing. Virchows Arch 457:299–307., DOI 10.1007/s00428-010- 0952-2 Foley EF, Jazaeri AA, Shupnik MA, Jazaeri O, Rice LW, 2000, Selective loss of estrogen receptor beta in malignant human colon. Cancer Res 60:245–248 Cho NL, Javid SH, Carothers AM, Redston M, Bertagnolli MM, 2007, Estrogen receptors α and β are inhibitory modifiers of Apcdependent tumorigenesis in the proximal colon of min/+ mice. Cancer Res 67:2366–2372., DOI 10.1158/0008-5472.CAN-06-3026 Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, 2009, American joint committee on cancer staging manual, 7th edn. Springer, New York JassamN, Bell SM, Speirs V, Quirke P, 2005, Loss of expression of oestrogen receptor beta in colon cancer and its association with dukes’ staging. Oncol Rep 14:17–21 Konstantinopoulos PA, Kominea A, Vandoros G, Sykiotis GP, Andricopoulos P, Varakis I, Sotiropoulou-Bonikou G, PapavassiliouAG(2003, Oestrogen receptor beta, ERbeta, is abundantly expressed in normal colonic mucosa, but declines in colon adenocarcinoma paralleling the tumour’s dedifferentiation. Eur J Cancer 39:1251–1258., DOI 10.1016/S0959-8049(03)00239-9 Jordan VC, 2007, New insights into the metabolism of tamoxifen and its role in the treatment and prevention of breast cancer. Steroids 72:829–842., DOI 10.1016/j.steroids.2007.07.009 Kato S, Sato T, Watanabe T, Takemasa S, Masuhiro Y, Ohtake F, Matsumoto T, 2005, Function of nuclear sex hormone receptors in gene regulation. Cancer Chemother Pharmacol 56:4–9., DOI 10. 1007/s00280-005-0102-8 Ohtake F, Takeyama K, Matsumoto T, Kitagawa H, Yamamoto Y, Nohara K, Tohyama C, Krust A, Mimura J, Chambon P, Yanagisawa J, Fujii-Kuriyama Y, Kato S, 2003, Modulation of oestrogen receptor signalling by association with the activated dioxin receptor. Nature 423:545–550., DOI 10.1038/nature01606 Metivier R, Reid G, Gannon F, 2006, Transcription in four dimensions: nuclear receptor-directed initiation of gene expression. EMBO Rep 7:161–167., DOI 10.1038/sj.embor.7400626 Bjornstrom L, Sjoberg M, 2005, Mechanisms of estrogen receptor signaling: convergence of genomic and nongenomic actions on target genes. Mol Endocrinol 19:833–842., DOI 10.1210/me.2004- 0486 Vasudevan N, Pfaff DW, 2007, Membrane-initiated actions of estrogens in neuroendocrinology: emerging principles. Endocr Rev 28:1–19., DOI 10.1210/er.2005-0021 Nanda K, Bastian LA, Hasselblad V, Simel DL, 1999, Hormone replacement therapy and the risk of colorectal cancer: a meta-analysis. Obstet Gynecol 93:880–888., DOI 10.1016/S0029-7844(98, 00424-4 Slattery ML, Potter JD, Curtin K, Edwards S,Ma KN, Anderson K, Schaffer D, SamowitzWS, 2001, Estrogens reduce and withdrawal of estrogens increase risk of microsatellite instability-positive colon cancer. Cancer Res 61:126–130 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 533 EP 540 DI 10.1007/s12253-017-0268-5 PG 8 ER PT J AU Sienkiewicz, B Urbaniak-Kujda, D Dybko, J Drys, A Hurkacz, M Wrobel, T Wiela-Hojenska, A AF Sienkiewicz, Beata Urbaniak-Kujda, Donata Dybko, Jaroslaw Drys, Andrzej Hurkacz, Magdalena Wrobel, Tomasz Wiela-Hojenska, Anna TI Influence of CYP2C19 Genotypes on the Occurrence of Adverse Drug Reactions of Voriconazole among Hematological Patients after Allo-HSCT SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adverse drug reactions; Voriconazole; CYP2C19; Genotyping; Hematology ID Adverse drug reactions; Voriconazole; CYP2C19; Genotyping; Hematology AB The aim of this study was to determine the influence of different CYP2C19 genotypes on selected liver function parameters, and ADR occurrence during VCZ prophylaxis in adult patients after allo-HSCT (allogeneic hematopoietic stem cell transplantation). CYP2C19 mutations were determined in a cohort of 30 adults using PCR-RFLP methods established by Sim et al. and Goldstein and Blaisdell. The patients’ protocol included biometrical and biochemical data, information on the underlying disease, chemotherapy, molds infections occurring during VCZ treatment, adverse drug reactions typical for the use of voriconazole, and probable drug - drug interactions. The observation and reporting of ADR took place from the −1 until the +20th day of VCZ therapy. For statistical analysis the χ2 test was used (p < 0.05). Among the examined patients 23 suffered from at least one side effect during VCZ therapy. Most frequent ADR were gastrointestinal disturbances (n = 15), nervous system (n = 11) and skin (n = 7) disorders. Patients with at least one loss of function allele (*2) were more likely to experience adverse drug reactions than those, with different genotypes. Due to the limited number of patients the result could not be proven with a statistical significance. Previous determination of CYP2C19 genotype may be a useful tool for prevention of adverse drug reactions during VCZ prophylaxis among patients after allo-HSCT. C1 [Sienkiewicz, Beata] Wroclaw Medical University, Faculty of Pharmacy, 211a Borowska St, 50-556 Wroclaw, Poland. [Urbaniak-Kujda, Donata] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, 4 Wybrzeze Pasteura St, 50-367 Wroclaw, Poland. [Dybko, Jaroslaw] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, 4 Wybrzeze Pasteura St, 50-367 Wroclaw, Poland. [Drys, Andrzej] Wroclaw Medical University, Department of Physical Chemistry, 211a Borowska St, 50-556 Wroclaw, Poland. [Hurkacz, Magdalena] Wroclaw Medical University, Faculty of Pharmacy, 211a Borowska St, 50-556 Wroclaw, Poland. [Wrobel, Tomasz] Wroclaw Medical University, Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, 4 Wybrzeze Pasteura St, 50-367 Wroclaw, Poland. [Wiela-Hojenska, Anna] Wroclaw Medical University, Faculty of Pharmacy, 211a Borowska St, 50-556 Wroclaw, Poland. RP Sienkiewicz, B (reprint author), Wroclaw Medical University, Faculty of Pharmacy, 50-556 Wroclaw, Poland. EM beata.sienkiewicz@gmail.com CR Fleming S, Yannakou CK, Haeusler GM, Clark J, Grigg A, Heath CH, Bajel A et al, 2014, Consensus guidelines for antifungal prophylaxis in haematological malignancy and haemopoietic stem cell transplantation, 2014. Intern Med J 44:1283–1297 Gautier-Veyret E, Fonrose X, Tonini J, Thiebaut-Bertrand A, Bartoli M, Quesada J-L, Bulabois C-E, Cahn J-Y, Stanke- Labesque F, 2015, Variability of voriconazole plasma concentrations after allogeneic hematopoietic stem cell transplantation: impact of cytochrome P450 polymorphisms and comedications on initial and subsequent trough levels. Antimicrob Agents Chemother 59:2305–2314 Groll AH, Castagnola E, Cesaro S, Dalle JH, Engelhard D, Hope W, Roilides E, Styczynski J, Warris A, Lehrnbecher T, 2014, Fourth European conference on infections in leukaemia, ECIL-4): guidelines for diagnosis, prevention, and treatment of invasive fungal diseases in paediatric patients with cancer or allogeneic haemopoietic stem-cell transplantation. Lancet Oncol 15:327–340 Summary of Product CharacteristicsVfend, 2012, http://www.ema. europa.eu/docs/en_GB/document_library/EPAR_-_Product_ Information/human/000387/WC500049756.pdf. Accessed 10 Nov 2016 Moriyama B, Kadri S, Henning SA, Danner RL, Walsh TJ, 2015, Therapeutic drugmonitoring and genotypic screening in the clinical use of voriconazole. Curr Fungal Infect Rep 9:74–87 Obeng AO, Egelund EF, Alsultan A, Peloquin CA, Johnson JA, 2014, CYP2C19 polymorphisms and therapeutic drug monitoring of voriconazole: are we ready for clinical implementation of pharmacogenomics? Pharmacotherapy 34:703–718 Trubiano JA, Crowe A,Worth LJ, Thursky KA, SlavinMA, 2014, Put ting CYP2C19 genotyping to the test: uti l i ty of pharmacogenomic evaluation in a voriconazole-treated haematology cohort. J Antimicrob Chemother 70:1161–1165 Chau MM, Kong DCM, Hal SJ, Urbancic K, Trubiano JA, Cassumbhoy M, Wilkes J, Cooper CM, Roberts JA, Marriott DJE, Worth LJ, 2014, Consensus guidelines for optimizing antifungal drug delivery and monitoring to avoid toxicity and improve outcomes in patients with haematologicalmalignancy. Intern Med J 44:1364–1388 Purkins L, Wood N, Ghahramani P, Love ER, Eve MD, Fielding A, 2003, Coadministration of voriconazole and phenytoin: pharmacokinetic interaction, safety, and toleration. B J Clin Pharmacol 56:37–44 Damle B, Varma MV, Wood N, 2011, Pharmacokinetics of voriconazole administered concomitantly with fluconazole and population-based simulation for sequential use. Antimicrob Agents Chemother 55:5172–5177 Summary of Product Characteristics Sandimmun, 2013, http:// www.ema.europa.eu/docs/en_GB/document_library/Referrals_ document/Sandimmun_Neoral_30/WC500144886.pdf. Accessed 15 Nov 2016 Andes D, Azie N, Yang H, Harrington R, Kelley C, Tan R-D,Wu EQ, Franks B, Kristy R, Lee E, Khandelwal N, Spalding J, 2016, Drugdrug interaction associated with mold-active triazoles among hospitalized patients. Antimicrob Agents Chemother 60:3398–3406 Lopez JL, Tayek JA, 2016, Voriconazole-induced hepatitis via simvastatin- and lansoprazole-mediated drug interactions: a case report and review of the literature. Drug Metab Dispos 44:124–126 Vadlapatla RK, Patel M, Paturi DK, Pal D, Mitra AK, 2014, Clinically relevant drug-drug interactions between antiretrovirals and antifungals. Expert Opin Drug Metab Toxicol 10(4):561–580 Sienkiewicz B, Urbaniak-Kujda D, Dybko J, Wrobel T, Wiela- Hojenska A, 2017, Influence of CYP2C19*2/*17 genotype on adverse drug reactions of voriconazole in patients after allo-HSCT- a four case report. J Cancer Res Clin Oncol 143:1103–1106 Goldstein JA, Blaisdell J, 1996, Genetic tests which identify the principal defects in CYP2C19 responsible for the polymorphism in mephenytoin metabolism. Methods Enzymol 272:210–218 SimSC, Risinger C, DahlML, Aklillu E, Christensen M, Bertilsson L, Ingelman-SundbergM(2006, A common novel CYP2C19 gene variant causes ultrarapid drug metabolism relevant for the drug response to proton pump inhibitors and antidepressants. Clin Pharmacol Ther 79:103–113 Moriyama B, Falade O, Leung J, Penzak SR, Jjingo C, Huang X, Henning SA, Wilson WH, Walsh TJ, 2011, Prolonged half-life of Voriconazole in CYP2C19 homozygous poor metabolizer recieving vincristine chemotherapy: avoiding a serious adverse drug interaction. Mycoses 54:1–5 Moriyama B, Jarosinski PF, Figg WD, Henning SA, Danner RA, Penzak SR, Wayne AS et al, 2013, Pharmacokinetics of intravenous voriconazole in obese patients: implications of CYP2C19 homozygous poor metabolizer genotype. Pharmacotherapy 33:19–22 Bruggemann RJM, Blijlevens NMA, Burger DM, Franke B, Troke PF, Donnelly JP, 2010, Pharmacokinetics and safety of 14 days intravenous voriconazole in allogeneic haematopoietic stem cell transplant recipients. J Antimicrob Chemother 65:107–113 Kim SH, Yim DS, Choi SM, Kwon JC, Han S, Lee DG, Park C, Kwon EY, Park SH, Choi JH, Yoo JH, 2011, Voriconazole-related severe adverse events: clinical application of therapeutic drug monitoring in Korean patients. Int J Infect Dis 15:753–758 Chu HY, Jain R, Xie H, Pottinger P, Fredricks DN, 2013, Voriconazole therapeutic drug monitoring: retrospective cohort study of the relationship to clinical outcomes and adverse events. BMC Infect Dis 13:105 Koselke E, Kraft S, Smith J, Nagel J, 2012, Evaluation of the effect of obesity on voriconazole serum concentrations. J Antimicrob Chemother 67:2957–2962 Pascual A, Calandra T, Bolay S, Buclin T, Bille JMO, 2008, Voriconazole therapeutic drug monitoring in patients with invasive mycoses improves efficacy and safety outcomes. Clin Infect Dis 46:201–211 DoltonMJ, McLachlan AJ, 2014, Voriconazole pharmacokinetics and exposure-response relationships: assessing the links between exposure, efficacy and toxicity. Int J Antimicrob Agents 44:183–193 Summary of Product Characteristics Methotrexat-EBEWE, 2013, http://leki.urpl.gov.pl/files/26_MethotrexatEbewe.pdf. Accessed 15 Nov 2016 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 541 EP 545 DI 10.1007/s12253-017-0264-9 PG 5 ER PT J AU Pyo, JS Kim, HJ AF Pyo, Jung-Soo Kim, Heon Joo TI Clinicopathological Significance of Micropapillary Pattern in Lung Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung adenocarcinoma; Micropapillary pattern; Clinicopathological characteristics; Meta-analysis ID Lung adenocarcinoma; Micropapillary pattern; Clinicopathological characteristics; Meta-analysis AB The aim of this study was to elucidate the clinicopathological characteristics of the micropapillary (MP) subtype and its correlation with survival in lung adenocarcinoma. We investigated the clinicopathological characteristics, including the incidence, sex, smoking history, tumor si z e , lymph node metastasis, lymphovascular invasion, distant metastasis, genetic alteration, and prognosis in lung adenocarcinoma with the MP pattern through a meta-analysis. From 48 eligible studies, 19,502 lung adenocarcinomas were included. The incidence rate of the MP pattern was 0.101 [95% confidence interval (CI) 0.075–0.136]. There was no significant difference between stage I and III tumors. Lung adenocarcinoma with the MP pattern showed higher rates of lymphatic invasion (0.526, 95% CI 0.403–0.645). MP pattern was found in 0.150 (95% CI 0.008–0.790) of lung adenocarcinoma with distant metastasis. In lung adenocarcinoma with the MP pattern, the estimated rates of ALK, EGFR, and KRAS mutations were 0.102 (95% CI 0.027–0.322), 0.620 (95% CI 0.444–0.769), and 0.118 (95% CI 0.027–0.393), respectively. The MP pattern of lung adenocarcinoma was significantly correlated with worse overall and disease-free survival rates (hazard ratio 1.704, 95% CI 1.216–2.387, and 2.082, 95% CI 1.541–2.813, respectively). Taken together, identification of the MP pattern in lung adenocarcinoma is useful for prediction of clinicopathological characteristics and prognosis of patients. C1 [Pyo, Jung-Soo] Eulji University, School of Medicine, Department of Pathology, 95 Dunsanseo-ro, Seo-gu, 35233 Daejeon, South Korea. [Kim, Heon Joo] Eulji University, School of Medicine, Department of Pathology, 95 Dunsanseo-ro, Seo-gu, 35233 Daejeon, South Korea. RP Kim, HJ (reprint author), Eulji University, School of Medicine, Department of Pathology, 35233 Daejeon, South Korea. EM kjh2000@eulji.ac.kr CR TravisWD, Brambilla E, Burke AP, Marx A, Nicholson AG, 2015, WHO classification of tumours of the lung, pleura, thymus and heart, 4th edn. International Agency for Research on Cancer, Lyon Travis WD, Brambilla E, Nicholson AG et al, 2015, The 2015 World Health Organization classification of lung tumors: impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol 10:1243–1260 Yap TA, Gerlinger M, Futreal PA, Pusztai L, Swanton C, 2012, Intratumor heterogeneity: seeing the wood for the trees. Sci Transl Med 4:127ps10 Yoshizawa A, Motoi N, Riely GJ, Sima CS, Gerald WL, Kris MG et al, 2011, Impact of proposed IASLC/ATS/ERS classification of lung adenocarcinoma: prognostic subgroups and implications for further revision of staging basedon analysis of 514 stage I cases. Mod Pathol 24:653–664 Russell PA,Wainer Z,WrightGM, DanielsM, Conron M,Williams RA, 2011, Does lung adenocarcinoma subtype predict patient survival?: aclinicopathologic study based on the new international Association for theStudy of lung cancer/American Thoracic Society/European RespiratorySociety international multidisciplinary lung adenocarcinoma classification. J Thorac Oncol 6:1496– 1504 Warth A, Muley T, Meister M et al, 2012, The novel histologic International Association for the Study of LungCancer/American T h o r a c i c S o c i e t y / E u r o p e an Res p i r a t o ry So c i e t y classificationsystem of lung adenocarcinoma is a stageindependent predictor of survival. J Clin Oncol 30:1438–1446 Cai YR, Dong YJ, Wu HB et al, 2016, Micropapillary: a component more likely to harbour heterogeneous EGFR mutations in lung adenocarcinomas. Sci Rep 6:23755 Amin MB, Tamboli P, Merchant SH et al, 2002, Micropapillary component in lung adenocarcinoma: a distinctive histologic feature with possible prognostic significance. Am J Surg Pathol 26:358– 364 Sanchez-Mora N, Presmanes MC, Monroy V et al, 2008, Micropapillary lung adenocarcinoma: a distinctive histologic subtype with prognostic significance. Case series. HumPathol 39:324– 330 Makimoto Y, Nabeshima K, Iwasaki H et al, 2005, Micropapillary pattern: a distinct pathological marker to subclassify tumours with a significantly poor prognosis within small peripheral lung adenocarcinoma, G polymorphism and the risk of breast cancer: a case-control study. Tumour Biol 35:12099–12102 Tao YP, Wang WL, Li SY, Zhang J, Shi QZ, Zhao F, Zhao BS, 2012, Associations between polymorphisms in IL-12A, IL-12B, IL-12Rβ1, IL-27 gene and serum levels of IL-12p40, IL-27p28 with esophageal cancer. J Cancer Res Clin Oncol 138:1891–1900 Shi J, Yuan M, Liu S, Duan X, Chen J, 2016, Correlation of IL-27 genetic polymorphisms with the risk and survival of cervical cancer in a Chinese Han population. Tumour Biol 37:6875–6879 Zhang Z, Zhou B, Wu Y, 2014, Prognostic value of IL-27 polymorphisms and susceptibility to epithelial ovarium cancer in a chinese population. Immunogenetics 66:85 Meadows AT, Chintagumpala M, Dunkel IJ, Friedman D, Stoner JA, Villablanca JG, 2009, Children’s oncology group trials for Retinoblastoma. In: Singh A, Damato B, Pe’er J, Murphree AL, Perry JD, eds, Essentials of ophthalmic oncology. SLACK Incorporated, Thorofare, pp 204–205 Pizzo PA, Poplack DG, Adamson PC, Blaney SM, Helman LJ, 2015, Principles and practice of pediatric oncology, 7th edn. Lippincott Williams & Wilkins, Philadelphia, pp 463–497 Amirghofran Z, Asiaee E, Kamazani FM, 2016, Soluble CD44 and CD44v6 and prognosis in children with B-cell acute lymphoblastic leukemia. Asia Pac J Clin Oncol 12:e375–e382 Valibeigi B, Amirghofran Z, Golmoghaddam H, Hajihosseini R, Kamazani FM, 2014, Fas gene variants in childhood acute lymphoblastic leukemia and association with prognosis. Pathol Oncol Res 20:367–374 Amirghofran Z, Daneshbod Y, Gholijani N, Esmaeilbeig M, 2011, The influence of Bcl-2 and myeloid antigen expression on response to therapy in childhood acute lymphoblastic leukemia. Arch Iran Med 14:170–174 Pu Y, Chen P, Zhou B, Zhang P, Wang Y, Song Y, Zhang L, 2015, Association between polymorphisms in IL27 gene and renal cell carcinoma. Biomarkers 20:202–205 Lyu S, Ye L,Wang O, Huang G, Yang F, Liu Y, Dong S, 2015, IL- 27 rs153109 polymorphism increases the risk of colorectal cancer in Chinese Han population. Onco Targets Ther 16:1493–1497 Yu X, Zhang Z, Zhou B, Zhang L, Chen P, Duan R, Li L, Zeng X, XiM(2016, Genetic polymorphisms of interleukin-27 is associated with endometrial cancer susceptibility in Chinese Han women. Int J Clin Exp Pathol 9:2718–2725 Chae SC, Li CS, Kim KM, Yang JY, Zhang Q, Lee YC, 2007, Identification of polymorphisms in human interleukin-27 and their association with asthma in a Korean population. J Hum Genet 52: 355–361 Wang Z,Wang L, Fan R, Zhou J, Zhong J, 2014, Association of IL- 27 gene three polymorphisms with Crohn’s disease susceptibility in a Chinese Han population. Int J Clin Exp Pathol 7:8952–8957 Tang YJ, Wang JL, Nong LG, Lan CG, Zha ZG, Liao PH, 2014, Associations of IL-27 polymorphisms and serum IL-27p28 levels with osteosarcoma risk. Medicine, Baltimore, 93:e56 Zhou B, Zhang P, Tang T, Liao H, Zhang K, Pu Y, Chen P, Song Y, Zhang L, 2015, Polymorphisms and plasma levels of IL-27: impact on genetic susceptibility and clinical outcome of bladder cancer. BMC Cancer 15:433 Zhao B, Meng LQ, Huang HN, Pan Y, Xu QQ, 2009, A novel functional polymorphism, 16974 A/C, in the interleukin-12-3′ untranslated region is associated with risk of glioma. DNA Cell Biol 28:335–341 Zhang J, Qian X, Ning H, Yang J, Xiong H, Liu J, 2010, Activation of IL-27 p28 gene transcription by interferon regulatory factor 8 in cooperation with interferon regulatory factor 1. J Biol Chem 9: 21269–21281 Naumnik W, Naumnik B, Niewiarowska K, Ossolin Ska M, Chyczewska E, 2012, Novel cytokines:IL-27, IL-29, IL-31: can they be useful in clinical practice at the time diagnosis of lung cancer. Exp Oncol 34:348–353 Lu D, Zhou X, Yao L, Liu C, Jin F, Wu Y, 2014, Clinical implications of the interleukin 27 serum level in breast cancer. J Investig Med 62:627–631 Horlad H, Ma C, Yano H, Pan C, Ohnishi K, Fujiwara Y, Endo S, Kikukawa Y, Okuno Y, Matsuoka M, Takeya M, Komohara Y, 2016, An IL-27/Stat3 axis induces expression of programmed cell death 1 ligands, PD-L1/2, on infiltrating macrophages in lymphoma. Cancer Sci 107:1696–1704 Zorzoli A, Di Carlo E, Cocco E, Ognio E, Ribatti D, Ferretti E, Dufour C, Locatelli F, Montagna D, Airoldi I, 2012, Interleukin-27 inhibits the growth of pediatric acute myeloid leukemia in NOD/ SCID/II2rg−/− mice. Clin Cancer Res 18:1630–1640 Canale S, Cocco C, Frasson C, Seganfreddo E, Di Carlo E, Ognio E, Sorrentino C, Ribatti D, Zorzoli A, Basso G, Dufour C, Airoldi I, 2011, Interleukin-27 inhibits pediatric B-acute lymphoblastic leukemia cell spreading in a preclinical model. Leukemia 25: 1815–1824 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 653 EP 662 DI 10.1007/s12253-017-0295-2 PG 10 ER PT J AU Li, X Li, B Li, B Guo, T Sun, Z Li, X Chen, L Chen, W Chen, P Mao, Y Zeng, Y AF Li, Xiaohan Li, Bo Li, Boan Guo, Tongsheng Sun, Zhiqiang Li, Xiaoxi Chen, Lin Chen, Weijiao Chen, Peng Mao, Yuanli Zeng, Yanjun TI ITIH4: Effective Serum Marker, Early Warning and Diagnosis, Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mass spectrum; Liver diseases; Liver cancer; Cirrhosis patients; Biomarker; AFP ID Mass spectrum; Liver diseases; Liver cancer; Cirrhosis patients; Biomarker; AFP AB Hepatocellular carcinoma (HCC) is a highly lethal malignant tumor evolved from cirrhosis. It is quite significant to seek accurate, easy markers for early warning and diagnosis of HCC. Through prospective cohort follow-up study and mass spectrometry, we discovered and verified a serum marker valuable for early warning and diagnosis. Follow-up observation was performed on cirrhosis patients. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) was adopted to detect the serums of patients, and the serum polypeptides with a potential value in early HCC warning and diagnosis were screened. Electrospray ionization quadrupole time-of-flight tandem mass spectrometry (ESI-Q-TOF-MS/MS) was exploited to identify these screened polypeptides. Moreover, the serum marker concentration was determined by ELISA to validate the clinical value of the serum marker. Among 109 cirrhosis patients followed up for two years, 29 patients (26.6%) finally progressed into HCC. MALDI-TOF MS shows that the concentration of a 3155.66Da polypeptide was significantly different between the patients that progressed into HCC and those not. Through MS/MS identification, it is confirmed that the polypeptide is inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4). The serum ITIH4 concentrations in two groups were measured with ELISA and compared with Alpha-fetoprotein (AFP). Results show that serum ITIH4 and AFP concentrations were negatively correlated (r=−0.263, p=0.0006), and the ITIH4 concentration had a significant intergroup difference (p=0.000). Receiver operating characteristic (ROC) curve indicates that its predictive value (area under the curve, AUC) is 0.667, superior to AFP. For the patients progressing into HCC, serumsamples were separately collected when they were recruited and diagnosed as cirrhosis. Measurement on these samples reveals that ITIH4 was declining during the progression of HCC (p=0.006). By virtue of mass spectrometry, we discovered and identified a biomarker valuable for early HCC warning and diagnosis. This marker overperforms the commonly used AFP, demonstrating a bright prospect. C1 [Li, Xiaohan] Medical University of PLA, Graduate Student TeamBeijing, China. [Li, Bo] Medical University of PLA, Graduate Student TeamBeijing, China. [Li, Boan] Medical University of PLA, Graduate Student TeamBeijing, China. [Guo, Tongsheng] Medical University of PLA, Graduate Student TeamBeijing, China. [Sun, Zhiqiang] Medical University of PLA, Graduate Student TeamBeijing, China. [Li, Xiaoxi] Medical University of PLA, Graduate Student TeamBeijing, China. [Chen, Lin] Medical University of PLA, Graduate Student TeamBeijing, China. [Chen, Weijiao] Medical University of PLA, Graduate Student TeamBeijing, China. [Chen, Peng] Medical University of PLA, Graduate Student TeamBeijing, China. [Mao, Yuanli] Medical University of PLA, Graduate Student TeamBeijing, China. [Zeng, Yanjun] Beijing University of Technology, 100 Pingleyuan, Chaoyang DistrictBeijing, China. RP Zeng, Y (reprint author), Beijing University of Technology, Beijing, China. EM yjzeng@bjut.edu.cn CR Ferlay J, Soerjomataram I, Ervik M, 2013, Globocan 2012: estimated cancer incidence, mortality and prevalence worldwide in 2012. IARC CancerBase. http://globocan.iarc.fr Liovet JM, Burroughs A, Bruix J, BCLC, Liver Unitet, Digestive Disease Institute, IDIBAPS et al, 2003, Hepatocellular carcinoma. Lancet 362:1907–1917 Zhang Z, ZhangY,WangY, Xu L, XuW(2015, Alpha-fetoprotein- L3 and Golgi protein 73 may serve as candidate biomarkers for diagnosing alpha-fetoprotein-negative hepatocellular carcinoma. Onco Targets Ther 31(9):123–129 Jordi B,Morris S, 2011, Management of hepatocellular carcinoma: an update. Hepatol 53:1020–1022 Guidelines for diagnosis and treatment of primary hepatocellular carcinoma, 2011, Chin Clin Oncol 16:929–946 Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63:11–30 Virginia HG, Fanny T, 2013, Management of small hepatocellular carcinoma in cirrhosis: focus on portal hypertension. World J Gastroenterol 19:1193–1199 Zhao Y,Wang M, Cui C, Zhang L, Liao F, Li H, 2015, Significance of combined tests of serum golgi glycoprotein 73 and other biomarkers in diagnosis of small primary hepatocellular carcinoma. Cancer Biomark. 15:677–683 Bao YX, Yang Y, Zhao HR, Mao R, Xiao L, Zhang YF, 2013, Clinical significance and diagnostic value of Golgi-protein 73 in patients with early-stage primary hepatocellular carcinoma. Zhonghua Zhong Liu Za Zhi. 35:505–508 Witjes CD, van Aalten SM, Steyerberg EW, Borsboom GJ, de Man RA, Verhoef C, Ijzermans JN, 2013, Recently introduced biomarkers for screening of hepatocellular carcinoma: a systematic review and meta-analysis. Hepatol Int. 7:59–64 Taneja S, Ahmad I, Sen S, Kumar S, Arora R, Gupta VK, Aggarwal R et al, 2011, Plasma pep tidome profiling of acute hepatitis E patients by MALDI-TOF/TOF. Proteome Sci 9:5 Wang J, Wang X, Lin S, Chen C, Wang C, Ma Q, Jiang B et al, 2013, Identification of kininogen-1 as a serum biomarker for the early detection of advanced colorectal adenoma and colorectal cancer. PLoS One 8:e70519 Huijbers A, Velstra B, Dekker TJ, Mesker WE, van der Burgt YE, Mertens BJ, Deelder AM et al, 2010, Proteomic serum biomarkers and their potential application in cancer screening programs. Int J Mol Sci 11:4175–4193 Cobo F, 2013, Application of maldi-tof mass spectrometry in clinical virology: a review. Open. Virol J 27:84–90 Qiu F, Gao YH, Jiang CG, Tian YP, Zhang XJ, 2010, Serum proteomic profile analysis for endometrial carcinoma detection with MALDI-TOF MS. Arch Med Sci 6:245–252 TTang Y, Kitisin K, Jogunoori W, Li C, Deng C, Mueller SC, Ressom HW et al, 2008, Progenitor/stem cells give rise to liver cancer due to aberrant TGF-beta and IL-6 signaling. Proc Natl Acad Sci U S A 105:2445–2450 Bhanumathy CD, Tang Y, Monga SPS, Katuri V, Cox JA, Mishra B, Mishra L et al, 2002, Itih-4, a serine protease inhibitor regulated in interleukin-6-dependent liver formation: role in liver development and regeneration. Dev Dyn 223:59–69 The guidelines of prevention and treatment for chronic hepatitis B, 2010 version,, 2011, Chinese journal of experimental and clinical infectious disease, Electronic Version, 5:79–100 Guideline of prevention and treatment of hepatitis C, 2004, Zhonghua Yu Fang Yi Xue Za Zhi 38:210–215 Daniele B, Bencivenga A, Megna AS, 2004, Alpha fetoprotein and ultrasonography screening for hepatocellular carcinoma. Gastroenterology 127:S108–S112 Yang J, Yang SY, Hu XY, Huo SF, Shang WL, Li ZF, Lei N et al, 2010, Serum peptidome profiling in patients with lung cancer. Anat Rec, Hoboken, 293:2027–2033 Li J, Jin H, Li L, Shang L, Zhao Y, Wei F et al, 2012, Detection of murine toxoplasmosis using magnetic bead-based serum peptide profiling by MALDI-TOF MS. Vector Borne Zoonotic Dis 12: 462–466 Yang J, Gao Y, Zhao LY, Liu LY, Qin YN, Wang XE, Liu Y et al, 2014, Identification of potential serum proteomic biomarkers for clear cell renal cell carcinoma. PLoS One 9:e111364 Orvisky E, Drake SK, Martin BM, Abdel-Hamid M, Ressom HW, Varghese RS et al, 2006, Enrichment of low molecular weight fraction of serum for ms analysis of peptid enrichment of low molecular weight fraction of serum for ms analysis of peptides associated with hepatocellular carcinoma. Proteomics 6:2895–2902 Schwegler EE, Cazares L, Steel LF, Adam BL, Johnson DA, Semmes OJ, Block TM et al, 2005, SELDI-TOF MS profiling of serum for detection of the progression of chronic hepatitis C to hepatocellular carcinoma. Hepatology 41:634–642 Tian L, Wang Y, Xu DB, Gao YH, Wen XY, Tian YP, 2014, The differential diagnostic model for serous peptidomics in HBV carriers established by MALDI-TOF-MS analysis. Clin Biochem 47: 56–62 Noguchi Y, KurokawaMS, Okuse C,Matsumoto N, Nagai K, Sato T, AritoMet al, 2013, Serum peptides, represented by complement 3f des-arginine, are useful for prediction of the response to pegylated interferon-α plus ribavirin in patients with chronic hepatitis C. Hepatol Res 43:743–756 Ivancic MM, Irving AA, Jonakin KG, Dove WF, Sussman MF, 2014, The concentrations of EGFR, LRG1, ITIH4, and F5 in serum correlate with the number of colonic adenomas in ApcPirc/+ rats. Cancer Prev Res, Phila, 7:1160–1169 Subbannayya Y, Mir SA, Renuse S, 2012, Identification of differentially expressed serum proteins in gastric adenocarcinoma. J Proteome 127:80–88 Yang J, Xiong X, Liu S, 2015, Identification of novel serum peptides biomarkers for female breast cancer patients inWestern China. Proteomics 12:doi: 10.1002/pmic.201500321 Gangadharan B, Antrobus R, Dwek RA, Zitzmann N, 2007, Novel serum biomarker candidates for liver fibrosis in hepatitis C patients. Clin Chem 53:1792–1799 Noh CK, Kim SS, Kim DK, Lee HY, Cho HJ, Yoon SY, Hyun SA et al, 2014, Inter-alpha-trypsin inhibitor heavy chain H4 as a diagnostic and prognostic indicator in patients with hepatitis B virusassociated hepatocellular carcinoma. Clin Biochem 47:1257–1261 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 663 EP 670 DI 10.1007/s12253-017-0285-4 PG 8 ER PT J AU Szabo, E Kosa, Cs Babarczi, E Sulyok, M Ujhelyi, E Banhegyi, D Valyi-Nagy, I AF Szabo, Eszter Kosa, Csaba Babarczi, Edit Sulyok, Mihaly Ujhelyi, Eszter Banhegyi, Denes Valyi-Nagy, Istvan TI Prevalence of Anal Human Papillomavirus Infection in Hungarian Men Who Have Sex with Men SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Anal cancer screening; HIV-infected MSM; HPV; HPV genotype; MSM; Risk factor ID Anal cancer screening; HIV-infected MSM; HPV; HPV genotype; MSM; Risk factor AB Anal cancer is one of the leading causes of death in non-AIDS defining cancers. Most of these cancers are associated with high risk HPV infection. So far, the prevalence and the significance of anal HPV-infection have not been studied in the Hungarian MSM population. The main objective of our study was to determine the prevalence and associated risk factors of HPV-infection in the Hungarian MSM community, particularly in HIV-infected MSM. Out of 109 examinations 92 samples (80 HIV-infected and 12 HIV-negative MSM) were evaluated for both cytological abnormalities and HPV genotyping PCR. Using a questionnaire all enrolled individuals were interviewed about their sexual behavior, socioeconomic factors, drug use and other known or suspected risk factors. In the HIV-infected cohort 97.5% of the examined individuals were positive for any HPV type. In this group we detected high risk (HR) HPV in 88.8%, low risk (LR) HPV in 75.0% and probably high risk (PHR) HPV in 47.5% and multiple HPV infectionwas absolutely common (82.5%). In the HIV-negative MSM group the incidence of HPV-infection was 58.3%. The respective rate of HR-HPV, LR-HPV and PHR-HPV genotypes were 33.3%, 58.4%, and 16.7%. In the HIV-negative group both HPV infection frequency and the prevalence of the pertinent genotypes were much lower. The Hungarian MSM population is severely infected with HPV and HR-HPV. High-risk sexual behaviors are strong predictors for acquiring HR-HPV co-infections. Our results underline the necessity of anal cancer screening and the introduction of the vaccination program in the high-risk population. C1 [Szabo, Eszter] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Molekularis Genetikai Laboratorium, Albert Florian str. 5-7, 1097 Budapest, Hungary. [Kosa, Csaba] St. Istvan and St. Laszlo Hospital, 3th Department of Infectology, Albert Florian str. 5-7, 1097 Budapest, Hungary. [Babarczi, Edit] Municipal Hospital, Del-Pest, Department of Pathology, Nagyvarad square 1, 1097 Budapest, Hungary. [Sulyok, Mihaly] Semmelweis University, School of PhD Studies, Ulloi str. 26, 1085 Budapest, Hungary. [Ujhelyi, Eszter] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Molekularis Genetikai Laboratorium, Albert Florian str. 5-7, 1097 Budapest, Hungary. [Banhegyi, Denes] St. Istvan and St. Laszlo Hospital, 3th Department of Infectology, Albert Florian str. 5-7, 1097 Budapest, Hungary. [Valyi-Nagy, Istvan] St. Istvan and St Laszlo Hospital, Hepatology Center, Nagyvarad square 1, 1097 Budapest, Hungary. RP Szabo, E (reprint author), Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Molekularis Genetikai Laboratorium, 1097 Budapest, Hungary. EM eszter.szabo321@gmail.com CR Joseph DA, Miller JW, Wu X, Chen VW, Morris CR, Goodman MT, Villalon-Gomez JM, Williams MA, Cress RD, 2008, Understanding the burden of human papillomavirus-associated anal cancers in the US. Cancer 113:2892–2900., DOI 10.1002/cncr.23744 Jemal A, Simard EP, Dorell C, Noone AM, Markowitz LE, Kohler B, Eheman C, Saraiya M, Bandi P, Saslow D, Cronin KA, Watson M, Schiffman M, Jane Henley S, Schymura MJ, Anderson RN, Yankey D, Edwards BK, 2013, Annual report to the nation on the status of cancer, 1975-2009, featuring the burden and trends in human papillomavirus, HPV)-associated cancers and HPV vaccination coverage levels. J Natl Cancer Inst 105:175–201., DOI 10. 1093/jnci/djs491 Daling JR, Weis NS, Klopfenstein LL, Cochran LE, Chow WH, Daifuku R, 1982, Correlates of homosexual behavior and the incidence of anal cancer. JAMA 247:1988–1990 Daling JR, Weiss NS, Hislop TG, Maden C, Coates RJ, Sherman KJ, Ashley RL, Beagrie M, Ryan JA, Corey L, 1987, Sexual practices, sexually transmitted diseases, and the incidence of anal cancer. N Engl J Med 317:973–977., DOI 10.1056/ NEJM198710153171601 Piketty C, Selinger-Leneman H, Grabar S, Duvivier C, Bonmarchand M, Abramowitz L, Costagliola D, Mary-Krause M, 2008, Marked increase in the incidence of invasive anal cancer among HIV-infected patients despite treatment with combination antiretroviral therapy. AIDS 22:1203–1211., DOI 10.1097/QAD. 0b013e3283023f78 Chin-Hong PV, Vittinghoff E, Cranston RD, Buchbinder S, Cohen D, Colfax G, Da Costa M, Darragh T, Hess E, Judson F, Koblin B, Madison M, Palefsky JM, 2004, Age-Specific prevalence of anal human papillomavirus infection in HIV-negative sexually active men who have sex with men: the EXPLORE study. J Infect Dis 190:2070–2076., DOI 10.1086/425906 Frisch M, Smith E, Grulich A, Johansen C, 2003, Cancer in a population-based cohort ofmen and women in registered homosexual partnerships. Am J Epidemiol 157:966–972., DOI 10.1093/aje/ kwg067 Shiels MS, Pfeiffer RM, Chaturvedi AK, Kreimer AR, Engels EA, 2012, Impact of the HIV epidemic on the incidence rates of anal cancer in the United States. J Natl Cancer Inst 104:1591–1598., DOI 10.1093/jnci/djs371 Palella FJ, Baker RK, Moorman AC, Chmiel JS,Wood KC, Brooks JT, Holmberg SD, 2006, Mortality in the highly active antiretroviral therapy era: changing causes of death and disease in the HIV outpatient study. J Acquir Immune Defic Syndr 43:27–34., DOI 10. 1097/01.qai.0000233310.90484.16 Shiels MS, Cole SR, Kirk GD, Poole C, 2009, A meta-analysis of the incidence of non-AIDS cancers in HIV-infected individuals. J Acquir Immune Defic Syndr 52:611–622., DOI 10.1158/1940-6207. PREV-08-A117 Patel J, Salit IE, BerryMJ, de Pokomandy a, Nathan M, Fishman F, Palefsky J, Tinmouth J, 2014, Environmental scan of anal cancer screening practices: worldwide survey results. Cancer Med, DOI 10. 1002/cam4.250 Daling JR, MadeleineMM, Johnson LG, Schwartz SM, Shera KA, Wurscher MA, Carter JJ, Porter PL, Galloway DA, McDougall JK, 2004, Human papillomavirus, smoking, and sexual practices in the etiology of anal cancer. Cancer 101:270–280., DOI 10.1002/cncr. 20365 Kreuter A, Potthoff A, Brockmeyer NH, Gambichler T, Swoboda J, Stucker M, Schmitt M, Pfister H, Wieland U, 2010, Anal carcinoma in human immunodeficiency virus-positive men: results of a prospective study from Germany. Br J Dermatol 162:1269–1277., DOI 10.1111/j.1365-2133.2010.09712.x Parkin DM(2006, The global health burden of infection-associated cancers in the year 2002. Int J Cancer 118:3030–3044., DOI 10.1002/ ijc.21731 Palefsky JM, Holly EA, Ralston ML, Arthur SP, Jay N, Berry JM, DaCosta MM, Botts R, Darragh TM, 1998, Anal squamous intraepithelial lesions in HIV-positive and HIV-negative homosexual and misexual men: prevalence and risk factors. JAIDS J Acquir Immune Defic Syndr 177:361–367 Melo VH, Guimaraes MD, Rocha GM, Araujo AC, Carmo RA, Grinsztejn B, Pilotto JH, Palefsky JM, 2014, Prevalence and risk factors associated with anal intraepithelial neoplasia among HIVpositive men in Brazil. J Low Genit Tract Dis 18:128–135., DOI 10. 1097/LGT.0b013e31829ee855 Berry JM, Jay N, Cranston RD, Darragh TM, Holly E a., Welton ML, Palefsky JM, 2004, Progression of anal high-grade squamous intraepithelial lesions to invasive anal cancer among HIV-infected men who have sex with men. Int J Cancer 134:1147–1155., DOI 10. 1002/ijc.28431 Castle PE, Porras C, Quint WG, Rodriguez AC, Schiffman M, Gravitt PE, Gonzalez P, Katki HA, Silva S, Freer E, Van Doorn LJ, Jimenez S, Herrero R, Hildesheim A, 2008, Comparison of two PCR-based human papillomavirus genotyping methods. J Clin Microbiol 46:3437–3445., DOI 10.1128/JCM.00620-08 Coutlee F, Rouleau D, Petignat P, Ghattas G, Kornegay JR, Schlag P, Boyle S, Hankins C, Vezina S, Cote P, Macleod J, Voyer H, Forest P, Walmsley S, Franco E, Conners J, Grimshaw R, Haase D, Johnston L, SchlechW, Yuzicappi-Fayant A, Landis S, Smaill F, Austin T, Hammerberg O, Ralph T, Falutz J, Ferenczy A, Klein M, Labrecque L, Lalonde R, Noel G, Perron C, Routy JP, Toma E, Touchie C, Victor G, Cote L, Senay H, Trottier S, Williams K, Piche A, Sandre R, Binder L, Keystone D, Phillips A, Rachlis A, Salit I, Wagner C, Braitstein P, Burdge D, Harris M, Money D, Montaner J, 2006, Enhanced detection and typing of human papillomavirus, HPV, DNA in anogenital samples with PGMYprimers and the linear array HPV genotyping test. J Clin Microbiol 44: 1998–2006., DOI 10.1128/JCM.00104-06 Munoz N, Bosch FX, de Sanjose S, Herrero R, Castellsague X, Shah K V, Snijders PJF, Meijer CJLM, 2003, Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med 348:518–527., DOI 10.1056/NEJMoa021641 Piketty C, Darragh TM, Da Costa M, Bruneval P, Heard I, Kazatchkine MD, Palefsky JM, 2003, High prevalence of anal human papillomavirus infection and anal cancer precursors among HIV-infected persons in the absence of anal ontercourse. Ann Intern Med 138:453–459 Chiao EY, Giordano TP, Palefsky JM, Tyring S, El Serag H, 2006, Screening HIV-infected individuals for anal cancer precursor lesions: a systematic review. Clin Infect Dis 43:223–233., DOI 10. 1086/505219 Ortoski RA, Kell CS, 2011, Anal cancer and screening guidelines for human papillomavirus in men. J Am Osteopath Assoc 111:S35– S43 Palefsky JM, Holly EA, Hogeboom CJ, Berry JM, Jay N, Darragh TM, 1997, Anal cytology as a screening tool for anal squamous intraepithelial lesions. JAIDS J Acquir Immune Defic Syndr 14: 415–422 Goldie SJ, Kuntz KM, Weinstein MC, Freedberg KA, Welton ML, Palefsky JM, 1999, The clinical effectiveness and cost-effectiveness of screening for anal squamous intraepithelial lesions in homosexual and bisexual HIV-positive men. JAMA 281:1822– 1829., DOI 10.1001/jama.281.19.1822 Toth B, Sapi Z, Banhegyi D, MarschalkoM, Karpati S, 2015, Anal cytology. Orv Hetil 156:24–27., DOI 10.1556/OH.2015.30080 LamJMC, Hoch JS, Tinmouth J, Sano M, Raboud J, Salit IE, 2011, Cost-effectiveness of screening for anal precancers in HIV-positive men. AIDS 25:635–642., DOI 10.1097/QAD.0b013e3283434594 Cranston RD, Darragh TM, Holly EA, Jay N, Berry JM, Da Costa M, Efird JT, Palefsky JM, 2004, Self-collected versus cliniciancollected anal cytology specimens to diagnose anal intraepithelial neoplasia in HIV-positive men. J Acquir Immune Defic Syndr 36: 915–20 Chin-Hong P V, Berry JM, Cheng S-C, Catania JA, Da Costa M, Darragh TM, Fishman F, Jay N, Pollack LM, Palefsky JM, 2008, Comparison of patient- and clinician-collected anal cytology samples to screen for human papillomavirus–associated anal intraepithelial Neoplasia in men who have sex with men. Ann Intern Med 149:300–306 Palefsky JM, Giuliano AR, Goldstone S, Moreira ED, Aranda C, Jessen H, Hillman R, Ferris D, Coutlee F, Stoler MH, Marshall JB, Radley D,Vuocolo S, HauptRM,GurisD, Garner EIO, 2011, HPV vaccine against anal HPV infection and anal intraepithelial Neoplasia. N Engl J Med 365:1576–1585., DOI 10.1056/ NEJMoa1010971 Wilkin T, Lee JY, Lensing SY, Stier EA, Goldstone SE, Berry JM, Jay N, Aboulafia D, Cohn DL, Einstein MH, Saah A, Mitsuyasu RT, Palefsky JM, 2010, Safety and immunogenicity of the quadrivalent human papillomavirus vaccine in HIV-1-infected men. J Infect Dis 202:1246–53., DOI 10.1086/656320 Giuliano AR, Palefsky JM, Goldstone S, Moreira ED, Penny ME, Aranda C, Vardas E, Moi H, Jessen H, Hillman R, Chang Y-H, Ferris D, Rouleau D, Bryan J, Marshall JB, Vuocolo S, Barr E, Radley D, Haupt RM, Guris D, 2011, Efficacy of Quadrivalent HPV vaccine against HPV infection and disease in males. N Engl J Med 364:401–411., DOI 10.1056/NEJMoa0909537 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 671 EP 677 DI 10.1007/s12253-017-0292-5 PG 7 ER PT J AU Dilmac, S Erin, N Necdet, D Tanriover, G AF Dilmac, Sayra Erin, Nuray Necdet, Demir Tanriover, Gamze TI Nephronectin is Decreased in Metastatic Breast Carcinoma and Related to Metastatic Organs SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Nephronectin; Breast cancer; Metastasis; 4TLM; 4THM ID Nephronectin; Breast cancer; Metastasis; 4TLM; 4THM AB Breast cancer causes death mostly due to distant metastasis. During metastasis, cancer cells create new conditions in which normal tissue structure can be disturbed. Nephronectin, which is the primary ligand for α8β1 integrin, plays an important role in kidney development. There are conflicting findings regarding its role in cancer progression and metastasis, especially in breast carcinoma. The aim of this study was to determine changes in nephronectin expression in primary tumor tissues and metastatic visceral organs, using metastatic and non-metastatic cell lines in a mouse model of breast cancer. In our study, 4T1-Liver Metastatic and 4T1- Heart Metastatic cells, originally derived from 4T1-murine breast carcinoma, and non-metastatic 67NR carcinoma cells were used. Cancer cells were injected orthotopically into the mammary gland of 8–10 week-old Balb-c mice. Primary tumors, lung, liver tissues were collected on 12th and 25th days after the tumor injection. Immunohistochemistry was used to determine expression of nephronectin in tissues. We also investigated the expression levels of the protein by using western blot technique. We found that lung and liver tissue of control animals (not-injected with tumor cells) expressed nephronectin which was lost in animals bearing metastatic tumor for 25 days. In accordance, nephronectin staining of lung and liver was preserved in animals injected with nonmetastatic 67NR tumors. These results demonstrate that loss of nephronectin may play an important role in formation metastatic milieu for cancer cells. This is the first study demonstrating that tumor-induced loss of nephronectin expression in visceral organs in which metastatic growth takes place. C1 [Dilmac, Sayra] Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, Campus, 07070 Antalya, Turkey. [Erin, Nuray] Akdeniz University School of Medicine, Department of Medical PharmacologyAntalya, Turkey. [Necdet, Demir] Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, Campus, 07070 Antalya, Turkey. [Tanriover, Gamze] Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, Campus, 07070 Antalya, Turkey. RP Tanriover, G (reprint author), Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, 07070 Antalya, Turkey. EM gamzetanriover@akdeniz.edu.tr CR Gaffan J, Dacre J, JonesA, 2006, Educating undergraduatemedical students about oncology: a literature review. J Clin Oncol 24(12): 1932–1939., DOI 10.1200/JCO.2005.02.6617 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90., DOI 10. 3322/caac.20107 Weigelt B, Peterse JL, van’t Veer LJ, 2005, Breast cancer metastasis: markers and models. Nat Rev Cancer 5(8):591–602., DOI 10. 1038/nrc1670 Scully OJ, Bay BH, Yip G, Yu Y, 2012, Breast cancer metastasis. Cancer Genomics Proteomics 9(5):311–320 Erin N, Kale S, Tanriover G, Koksoy S, Duymus O, Korcum AF, 2013, Differential characteristics of heart, liver, and brain metastatic subsets of murine breast carcinoma. Breast Cancer Res Treat 139(3):677–689., DOI 10.1007/s10549-013-2584-0 Willis AL, Sabeh F, Li XY, Weiss SJ, 2013, Extracellular matrix determinants and the regulation of cancer cell invasion stratagems. J Microsc 251(3):250–260., DOI 10.1111/jmi.12064 Lu P, Takai K, Weaver VM, Werb Z, 2011, Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb Perspect Biol 3(12)., DOI 10.1101/cshperspect.a005058 Friedl P,Wolf K, 2003, Tumour-cell invasion and migration: diversity and escapemechanisms. Nat Rev Cancer 3(5):362–374., DOI 10. 1038/nrc1075 Egeblad M, Rasch MG, Weaver VM, 2010, Dynamic interplay between the collagen scaffold and tumor evolution. Curr Opin Cell Biol 22(5):697–706., DOI 10.1016/j.ceb.2010.08.015 Cheung KJ, Ewald AJ, 2014, Illuminating breast cancer invasion: diverse roles for cell-cell interactions. Curr Opin Cell Biol 30:99– 111., DOI 10.1016/j.ceb.2014.07.003 Hubmacher D, Apte SS, 2013, The biology of the extracellular matrix: novel insights. Curr Opin Rheumatol 25(1):65–70., DOI 10. 1097/BOR.0b013e32835b137b Lu P, Weaver VM, Werb Z, 2012, The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol 196(4):395–406., DOI 10.1083/jcb.201102147 Egeblad M, Nakasone ES,Werb Z, 2010, Tumors as organs: complex tissues that interface with the entire organism. Dev Cell 18(6): 884–901., DOI 10.1016/j.devcel.2010.05.012 Wiseman BS, Werb Z, 2002, Stromal effects on mammary gland development and breast cancer. Science 296(5570):1046–1049., DOI 10.1126/science.1067431 Brandenberger R, Schmidt A, Linton J,Wang D, Backus C, Denda S, Muller U, Reichardt LF, 2001, Identification and characterization of a novel extracellular matrix protein nephronectin that is associated with integrin alpha8beta1 in the embryonic kidney. J Cell Biol 154(2):447–458 Morimura N, Tezuka Y, Watanabe N, Yasuda M, Miyatani S, Hozumi N, Tezuka Ki K, 2001, Molecular cloning of POEM: a novel adhesion molecule that interacts with alpha8beta1 integrin. J Biol Chem 276(45):42172–42181., DOI 10.1074/jbc.M103216200 Erin N, Wang N, Xin P, Bui V, Weisz J, Barkan GA, Zhao W, Shearer D, Clawson GA, 2009, Altered gene expression in breast cancer liver metastases. Int J Cancer 124(7):1503–1516., DOI 10. 1002/ijc.24131 Erin N, Boyer PJ, Bonneau RH, Clawson GA, Welch DR, 2004, Capsaicin-mediated denervation of sensory neurons promotes mammary tumor metastasis to lung and heart. Anticancer Res 24(2B):1003–1009 Erin N, ZhaoW, Bylander J, Chase G, Clawson G, 2006, Capsaicininduced inactivation of sensory neurons promotes a more aggressive gene expression phenotype in breast cancer cells. Breast Cancer Res Treat 99(3):351–364., DOI 10.1007/s10549-006-9219-7 Kuphal S, Wallner S, Bosserhoff AK, 2008, Loss of nephronectin promotes tumor progression in malignant melanoma. Cancer Sci 99(2):229–233., DOI 10.1111/j.1349-7006.2007.00678.x Eckhardt BL, Parker BS, van Laar RK, Restall CM, Natoli AL, Tavaria MD, Stanley KL, Sloan EK, Moseley JM, Anderson RL, 2005, Genomic analysis of a spontaneous model of breast cancer metastasis to bone reveals a role for the extracellular matrix. Mol Cancer Res 3(1):1–13 Erin N, Nizam E, Tanriover G, Koksoy S, 2015, Autocrine control of MIP-2 secretion from metastatic breast cancer cells is mediated by CXCR2: a mechanism for possible resistance to CXCR2 antagonists. Breast Cancer Res Treat 150(1):57–69., DOI 10.1007/s10549- 015-3297-3 Erin N, Podnos A, Tanriover G, Duymus O, Cote E, Khatri I, Gorczynski RM, 2014, Bidirectional effect of CD200 on breast cancer development and metastasis, with ultimate outcome determined by tumor aggressiveness and a cancer-induced inflammatory response. Oncogene., DOI 10.1038/onc.2014.317 Erin N, Ulusoy O, 2009, Differentiation of neuronal from nonneuronal substance P. Regul Pept 152(1–3):108–113., DOI 10.1016/ j.regpep.2008.10.006 Green JR, 2003, Antitumor effects of bisphosphonates. Cancer 97(3 Suppl):840–847., DOI 10.1002/cncr.11128 Cohen MB, Griebling TL, Ahaghotu CA, Rokhlin OW, Ross JS, 1997, Cellular adhesion molecules in urologic malignancies. Am J Clin Pathol 107(1):56–63 Takagi J, 2004, Structural basis for ligand recognition by RGD, Arg-Gly-asp)-dependent integrins. Biochem Soc Trans 32(Pt3): 403–406., DOI 10.1042/BST0320403 Kahai S, Lee SC, Lee DY, Yang J, Li M,Wang CH, Jiang Z, Zhang Y, Peng C, Yang BB, 2009, MicroRNA miR-378 regulates nephronectin expression modulating osteoblast differentiation by targeting GalNT-7. PLoS One 4(10):e7535., DOI 10.1371/journal. pone.0007535 Linton JM, Martin GR, Reichardt LF, 2007, The ECM protein nephronectin promotes kidney development via integrin alpha8beta1-mediated stimulation of Gdnf expression. Development 134(13):2501–2509., DOI 10.1242/dev.005033 Kahai S, Lee SC, Seth A, Yang BB, 2010, Nephronectin promotes osteoblast differentiation via the epidermal growth factor-like repeats. FEBS Lett 584(1):233–238., DOI 10.1016/j.febslet.2009.11. 077 Patra C, Ricciardi F, Engel FB, 2012, The functional properties of nephronectin: an adhesion molecule for cardiac tissue engineering. Biomaterials 33(17):4327–4335., DOI 10.1016/j.biomaterials.2012. 03.021 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144(5):646–674., DOI 10.1016/j.cell.2011.02.013 Inacio Pinto N, Carnier J, Oyama LM, Otoch JP, Alcantara PS, Tokeshi F, Nascimento CM, 2015, Cancer as a Proinflammatory environment: metastasis and cachexia. Mediat Inflamm 2015: 791060., DOI 10.1155/2015/791060 Erin N, Korcum AF, Tanriover G, Kale S, Demir N, Koksoy S, 2015, Activation of neuroimmune pathways increases therapeutic effects of radiotherapy on poorly differentiated breast carcinoma. Brain Behav Immun 48:174–185., DOI 10.1016/j.bbi.2015.02.024 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 679 EP 688 DI 10.1007/s12253-017-0289-0 PG 10 ER PT J AU Pereira, SS Costa, MM Guerreiro, GS Monteiro, PM Pignatelli, D AF Pereira, S Sofia Costa, M Madalena Guerreiro, G Susana Monteiro, P Mariana Pignatelli, Duarte TI Angiogenesis and Lymphangiogenesis in the Adrenocortical Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adrenocortical tumors; Adrenocortical carcinoma; Lymphangiogenesis; Angiogenesis ID Adrenocortical tumors; Adrenocortical carcinoma; Lymphangiogenesis; Angiogenesis AB Adrenocortical tumors (ACT) are common adrenal tumors. The majority of ACTs are non-functioning and benign, while adrenocortical carcinomas (ACC) are rare, usually very aggressive and often metastasized when first diagnosed. Our aim was to assess whether blood and lymph vessel density within ACTs correlate with the malignancy character or tumor functionality. For that, the microvascular distribution was evaluated by immunohistochemistry staining with D2– 40 antibody, for lymph vessels and CD-31 antibody, for blood vessels, in ACCs (n = 15), adenomas with Cushing syndrome (n = 9) and non-functioning adenomas (n = 10). The percentage of stained area was quantified by computerized morphometric analysis. D2–40 expression was significantly lower in ACC as compared to adenomas with Cushing syndrome (p < 0.01) and correlated positively with the expression of the steroidogenic acute regulatory protein (StAR) (R2 = 0.553, p < 0.001). CD31 expression was found to be significantly higher in ACC as compared to adenomas with Cushing syndrome (p < 0.05). Our results show that angiogenesis is increased in ACC, suggesting that this phenomenon may have an important role in ACT biological behavior, while lymph vascular density seems to be more closely related to the tumor functional status than malignancy. C1 [Pereira, S Sofia] Universidade do Porto, Instituto de Investigacao e Inovacao em Saude (I3S)Porto, Portugal. [Costa, M Madalena] University of Porto, Clinical and Experimental Endocrinology, Department of AnatomyPorto, Portugal. [Guerreiro, G Susana] Universidade do Porto, Instituto de Investigacao e Inovacao em Saude (I3S)Porto, Portugal. [Monteiro, P Mariana] University of Porto, Clinical and Experimental Endocrinology, Department of AnatomyPorto, Portugal. [Pignatelli, Duarte] Universidade do Porto, Instituto de Investigacao e Inovacao em Saude (I3S)Porto, Portugal. RP Pignatelli, D (reprint author), Universidade do Porto, Instituto de Investigacao e Inovacao em Saude (I3S), Porto, Portugal. EM dpignatelli@ipatimup.pt CR Allolio B, Hahner S, Weismann D, Fassnacht M, 2004, Management of adrenocortical carcinoma. Clin Endocrinol 60(3): 273–287 Paduch R, 2016, The role of lymphangiogenesis and angiogenesis in tumor metastasis. Cell Oncol, Dordr, 39(5):397–410., DOI 10. 1007/s13402-016-0281-9 Alitalo K, Tammela T, Petrova TV, 2005, Lymphangiogenesis in development and human disease. Nature 438(7070):946–953., DOI 10.1038/nature04480 Pusztaszeri MP, Seelentag W, Bosman FT, 2006, Immunohistochemical expression of endothelial markers CD31, CD34, von Willebrand factor, and Fli-1 in normal human tissues. J Histochem Cytochem 54(4):385–395., DOI 10.1369/jhc.4A6514. 2005 Browning L, Bailey D, Parker A, 2008, D2-40 is a sensitive and specific marker in differentiating primary adrenal cortical tumours from both metastatic clear cell renal cell carcinoma and phaeochromocytoma. J Clin Pathol 61(3):293–296., DOI 10.1136/ jcp.2007.049544 Bernini GP, Moretti A, Bonadio AG, Menicagli M, Viacava P, Naccarato AG, Iacconi P, Miccoli P, Salvetti A, 2002, Angiogenesis in human normal and pathologic adrenal cortex. J Clin Endocrinol Metab 87(11):4961–4965., DOI 10.1210/jc.2001- 011799 Zhu Y, Xu Y, Chen D, Zhang C, RuiW, Zhao J, Zhu Q,Wu Y, Shen Z, Wang W, Ning G, Wang X, 2014, Expression of STAT3 and IGF2 in adrenocortical carcinoma and its relationship with angiogenesis. Clin Transl Oncol 16(7):644–649., DOI 10.1007/s12094- 013-1130-1 Pereira SS, Morais T, CostaMM,Monteiro MP, Pignatelli D, 2013, The emerging role of the molecular marker p27 in the differential diagnosis of adrenocortical tumors. Endocr Connect 2(3):137–145., DOI 10.1530/EC-13-0025 Logie JJ, Ali S, Marshall KM, HeckMM,Walker BR, Hadoke PW, 2010, Glucocorticoid-mediated inhibition of angiogenic changes in human endothelial cells is not caused by reductions in cell proliferation or migration. PLoS One 5(12):e14476., DOI 10.1371/ journal.pone.0014476 De Fraipont F, El Atifi M, Gicquel C, Bertagna X, Chambaz E, Feige J, 2000, Expression of the angiogenesis markers vascular endothelial growth factor-a, Thrombospondin-1, and plateletderived endothelial cell growth factor in human sporadic adrenocortical tumors: correlation with genotypic alterations 1. J Clin Endocrinol Metab 85(12):4734–4741 Moreira A, Pereira SS, Machado CL, Morais T, CostaM, Monteiro MP, 2014, Obesity inhibits lymphangiogenesis in prostate tumors. Int J Clin Exp Pathol 7(1):348–352 Ozerdem U, Wojcik EM, Duan X, Ersahin C, Barkan GA, 2013, Prognostic utility of quantitative image analysis of microvascular density in prostate cancer. Pathol Int 63(5):277–282., DOI 10.1111/ pin.12056 Pereira F, Pereira SS,MesquitaM,Morais T, CostaMM, Quelhas P, Lopes C, Monteiro MP, Leite V, 2017, Lymph node metastases in papillary and medullary thyroid carcinoma are independent of Intratumoral lymphatic vessel density. European Thyroid Journal 6(2):57–64 Kyle LH, Meyer RJ, Canary JJ, 1957, Mechanism of adrenal atrophy in Cushing’s syndrome due to adrenal tumor. N Engl J Med 257(2):57–61., DOI 10.1056/NEJM195707112570203 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 689 EP 693 DI 10.1007/s12253-017-0259-6 PG 5 ER PT J AU Ieni, A Angelico, G Giuffre, G Tuccari, G AF Ieni, Antonio Angelico, Giuseppe Giuffre, Giuseppe Tuccari, Giovanni TI Discordance Rate of HER2 Status in Primary Gastric Cancer and Synchronous Lymph Node Metastases: Its Impact on Therapeutic Decision and Clinical Management SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Ieni, Antonio] Azienda Ospedaliera Universitaria, Department of Human PathologyMessina, Italy. [Angelico, Giuseppe] Universita degli Studi di Catania, Policlinico G. Rodolico, Dipartimento Anatomia, Patologia diagnostica, Medicina legale, Igiene e Sanita PubblicaCatania, Italy. [Giuffre, Giuseppe] Azienda Ospedaliera Universitaria, Department of Human PathologyMessina, Italy. [Tuccari, Giovanni] Azienda Ospedaliera Universitaria, Department of Human PathologyMessina, Italy. RP Ieni, A (reprint author), Azienda Ospedaliera Universitaria, Department of Human Pathology, Messina, Italy. EM aieni@unime.it CR Amato M, Perrone G, Righi D et al, 2017, HER2 status in gastric cancer: comparison between primary and distant metastatic disease. Pathol Oncol Res 23:55–61 Ieni A, Barresi V, Rigoli L et al, 2015, HER2 status in premalignant, early, and advanced neoplastic lesions of the stomach. Dis Markers 2015:234851 Grillo F, Fassan M, Sarocchi F et al, 2016, HER2 heterogeneity in gastric/gastroesophageal cancers: from benchside to practice. World J Gastroenterol 22:5879–5887 Ahn S, Ahn S,Van VranckenMet al, 2015, Ideal number of biopsy tumor fragments for predicting HER2 status in gastric carcinoma resection specimens. Oncotarget 6:38372–38380 Gullo I, Grillo F, Molinaro L et al, 2015, Minimum biopsy set for HER2 evaluation in gastric and gastro-esophageal junction cancer. Endosc Int Open 3:E165–E170 Park SR, Park YS, Ryu MH et al, 2016, Extra-gain of HER2- positive cases through HER2 reassessment in primary and metastatic sites in advanced gastric cancer with initially HER2-negative primary tumours: results of GASTric cancer HER2 reassessment study 1, GASTHER1). Eur J Cancer 53: 42–50 Pagni F, Zannella S, Ronchi S et al, 2013, HER2 status of gastric carcinoma and corresponding lymph node metastasis. Pathol Oncol Res 19:103–109 Kochi M, FujiiM, Masuda S, et al., 2013, Differing deregulation of HER2 in primary gastric cancer and synchronous related metastatic lymph nodes. Diagn Pathol 21;8:191 Ieni A, Barresi V, Caltabiano R et al, 2014, Discordance rate of HER2 status in primary breast carcinomas versus synchronous axillary lymph node metastases: a multicenter retrospective investigation. Onco Targets Ther 7:1267–1272 Ieni A, Barresi V, Caltabiano R et al, 2014, Discordance rate of HER2 status in primary gastric carcinomas and synchronous lymph node metastases: a multicenter retrospective analysis. Int J Mol Sci 15:22331–22341 Ieni A, Barresi V, Branca G et al, 2015, Changes in human epidermal growth factor receptor 2 status between primary breast/gastric carcinomas and synchronous metastatic lymph nodes: how can we explain them? J Cancer Metastasis Treat 1:21–26 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 695 EP 696 DI 10.1007/s12253-017-0276-5 PG 2 ER PT J AU Kim, H Ahn, D Sohn, HJ Kim, YH Lee, JH Lee, H AF Kim, Heejin Ahn, Dongbin Sohn, Ho Jin Kim, Yong-Hee Lee, Jae-Ho Lee, Hyunsu TI TERT Promoter Mutation and Telomere Length in Salivary Gland Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Kim, Heejin] Kyungpook National University, Department of Otolaryngology - Head and Neck SurgeryDaegu, South Korea. [Ahn, Dongbin] Kyungpook National University, Department of Otolaryngology - Head and Neck SurgeryDaegu, South Korea. [Sohn, Ho Jin] Kyungpook National University, Department of Otolaryngology - Head and Neck SurgeryDaegu, South Korea. [Kim, Yong-Hee] Kyungpook National University, School of Medicine, Department of MicrobiologyDaegu, South Korea. [Lee, Jae-Ho] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeol-daero, Dalseo-gu, 1095 Daegu, South Korea. [Lee, Hyunsu] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeol-daero, Dalseo-gu, 1095 Daegu, South Korea. RP Lee, H (reprint author), Keimyung University, School of Medicine, Department of Anatomy, 1095 Daegu, South Korea. EM neuroana@dsmc.or.kr CR Horn S, Figl A, Rachakonda PS, Fischer C, Sucker A, Gast A, Kadel S, Moll I, Nagore E, Hemminki K, Schadendorf D, Kumar R, 2013, TERT promoter mutations in familial and sporadic melanoma. Science 339(6122):959–961., DOI 10.1126/science.1230062 Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA, 2013, Highly recurrent TERT promoter mutations in human melanoma. Science 339(6122):957–959., DOI 10.1126/science.1229259 Qu Y, Shi L,Wang D, Zhang B, Yang Q, Ji M, Shi B, Hou P, 2014, Low frequency of TERT promoter mutations in a large cohort of gallbladder and gastric cancers. Int J Cancer 134(12):2993–2994., DOI 10.1002/ijc.28633 Lee H, Jin J-D, La B-M, Park W-J, Choi I-J, Lee J-H, 2016, TERT promoter mutation, telomere length, and TERT expression in gastric cancer. Int J Clin Exp Pathol 9(2):1758–1763 Liu T,Wang N, Cao J, Sofiadis A, Dinets A, Zedenius J, Larsson C, Xu D, 2014, The age- and shorter telomere-dependent TERT promoter mutation in follicular thyroid cell-derived carcinomas. Oncogene 33(42):4978–4984., DOI 10.1038/onc.2013.446 Borah S, Xi L, Zaug AJ, Powell NM, Dancik GM, Cohen SB, Costello JC, Theodorescu D, Cech TR, 2015, Cancer. TERT promoter mutations and telomerase reactivation in urothelial cancer. Science 347(6225):1006–1010., DOI 10.1126/science.1260200 Oh C-M,Won Y-J, Jung K-W, Kong H-J, Cho H, Lee J-K, Lee DH, Lee KH, Community of Population-Based Regional Cancer R, 2016, Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2013. Cancer Res Treat 48(2):436–450., DOI 10. 4143/crt.2016.089 Schache AG, Hall G, Woolgar JA, Nikolaidis G, Triantafyllou A, Lowe D, Risk JM, Shaw RJ, Liloglou T, 2010, Quantitative promoter methylation differentiates carcinoma ex pleomorphic adenoma from pleomorphic salivary adenoma. Br J Cancer 103(12):1846– 1851., DOI 10.1038/sj.bjc.6605953 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 697 EP 698 DI 10.1007/s12253-017-0275-6 PG 2 ER PT J AU Kim, H Ahn, D Sohn, HJ Kim, YH Lee, JH Lee, H AF Kim, Heejin Ahn, Dongbin Sohn, Ho Jin Kim, Yong-Hee Lee, Jae-Ho Lee, Hyunsu TI Erratum to: TERT Promoter Mutation and Telomere Length in Salivary Gland Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report C1 [Kim, Heejin] Kyungpook National University, Department of Otolaryngology - Head and Neck SurgeryDaegu, South Korea. [Ahn, Dongbin] Kyungpook National University, Department of Otolaryngology - Head and Neck SurgeryDaegu, South Korea. [Sohn, Ho Jin] Kyungpook National University, Department of Otolaryngology - Head and Neck SurgeryDaegu, South Korea. [Kim, Yong-Hee] Kyungpook National University, School of Medicine, Department of MicrobiologyDaegu, South Korea. [Lee, Jae-Ho] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeol-daero, Dalseo-gu, 1095 Daegu, South Korea. [Lee, Hyunsu] Keimyung University, School of Medicine, Department of Anatomy, Dalgubeol-daero, Dalseo-gu, 1095 Daegu, South Korea. RP Lee, H (reprint author), Keimyung University, School of Medicine, Department of Anatomy, 1095 Daegu, South Korea. EM neuroana@dsmc.or.kr NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2018 VL 24 IS 3 BP 699 EP 699 DI 10.1007/s12253-017-0294-3 PG 1 ER PT J AU Gregorio, CA Lacerda, M Figueiredo, P Simoes, S Dias, S Moreira, NJ AF Gregorio, C Ana Lacerda, Manuela Figueiredo, Paulo Simoes, Sergio Dias, Sergio Moreira, Nuno Joao TI Therapeutic Implications of the Molecular and Immune Landscape of Triple-Negative Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Breast cancer; Triple-negative breast cancer; Intrinsic subtype; Gene expression profiling; Targeted therapies; Immune checkpoint inhibitors ID Breast cancer; Triple-negative breast cancer; Intrinsic subtype; Gene expression profiling; Targeted therapies; Immune checkpoint inhibitors AB Treatment and management of breast cancer imposes a heavy burden on public health care, and incidence rates continue to increase. Breast cancer is the most common female neoplasia and primary cause of death among women worldwide. The recognition of breast cancer as a complex and heterogeneous disease, comprising different molecular entities, was a landmark in our understanding of this malignancy. Valuing the impact of the molecular characteristics on tumor behavior enabled a better assessment of a patient’s prognosis and increased the predictive power to therapeutic response and clinical outcome. Molecular heterogeneity is also prominent in the triple-negative breast cancer subtype, and is reflected by the distinct prognostic and patient’s sensitivity to treatment, being chemotherapy the only systemic treatment currently available. From a therapeutic perspective, gene expression profiling of triple-negative tumors has notably contributed to the exploration of new druggable targets and brought to light the need to align these patients to the various therapies according to their triple-negative subtype. Additionally, the higher amount of tumor infiltrating lymphocytes, and the prevalence of an increased expression of PD-1 receptor and its ligand, PD-L1, in triple-negative tumors, created a new treatment opportunity with immune checkpoint inhibitors. This manuscript addresses the current knowledge on the molecular and immune profiles of breast cancer, and its impact on the development of targeted therapies, with a particular emphasis on the triple-negative subtype. C1 [Gregorio, C Ana] University of Coimbra, Center for Neuroscience and Cell Biology – CNC.IBILI, Rua Larga, 3004-504 Coimbra, Portugal. [Lacerda, Manuela] University of Porto, Department of Pathology and Immunology of ICBASPorto, Portugal. [Figueiredo, Paulo] IPOFG-EPE - Portuguese Institute of Oncology Francisco GentilCoimbra, Portugal. [Simoes, Sergio] University of Coimbra, Polo das Ciencias da Saude, FFUC - Faculty of PharmacyCoimbra, Portugal. [Dias, Sergio] University of Lisbon, Faculty of Medicine, IMM – Institute of Molecular MedicineLisbon, Portugal. [Moreira, Nuno Joao] University of Coimbra, Center for Neuroscience and Cell Biology – CNC.IBILI, Rua Larga, 3004-504 Coimbra, Portugal. RP Moreira, NJ (reprint author), University of Coimbra, Center for Neuroscience and Cell Biology – CNC.IBILI, 3004-504 Coimbra, Portugal. EM jmoreira@ff.uc.pt CR Tavassoli FA, 1999, Pathology of the Breast. McGraw Hill Professional Carter CL, Allen C, Henson DE, 1989, Relation of tumor size, lymph node status, and survival in 24,740 breast cancer cases. Cancer 63(1):181–187. , DOI 10.1002/1097- 0142(19890101)63:1<181::AID-CNCR2820630129>3.0.CO;2-H Le Doussal V, Tubiana-HulinM, Friedman S, Hacene K, Spyratos F, Brunet M, 1989, Prognostic value of histologic grade nuclear components of Scarff-Bloom-Richardson, SBR). An improved score modification based on a multivariate analysis of 1262 invasive ductal breast carcinomas. Cancer 64(9):1914–1921. https:// doi.org/10.1002/1097-0142(19891101)64:9<1914::AIDCNCR2820640926> 3.0.CO;2-G Sobin LH, Gospodarowicz MK,Wittekind C, 2011, TNM classification of malignant tumours. Wiley D'Eredita G, Giardina C, Martellotta M, Natale T, Ferrarese F, 2001, Prognostic factors in breast cancer: the predictive value of the Nottingham Prognostic Index in patients with a long-term follow-up that were treated in a single institution. Eur J Cancer 37(5):591–596. , DOI 10.1016/S0959-8049(00)00435-4 Balslev I,Axelsson CK, Zedeler K, Rasmussen BB, Carstensen B, Mouridsen HT, 1994, The Nottingham Prognostic Index applied to 9,149 patients from the studies of the Danish Breast Cancer Cooperative Group, DBCG). Breast Cancer Res Treat 32(3): 281–290. , DOI 10.1007/BF00666005 Sinn HP, Kreipe H, 2013, A Brief Overview of the WHO Classification of Breast Tumors, 4th Edition, Focusing on Issues and Updates from the 3rd Edition. Breast Care, Basel, 8(2):149– 154. , DOI 10.1159/000350774 Elston CW, Ellis IO, Pinder SE, 1999, Pathological prognostic factors in breast cancer. Crit Rev Oncol Hematol 31(3):209–223. , DOI 10.1016/S1040-8428(99)00034-7 Mohsin SK, Weiss H, Havighurst T, Clark GM, Berardo M, Roanh le D, To TV, Qian Z, Love RR, Allred DC, 2004, Progesterone receptor by immunohistochemistry and clinical outcome in breast cancer: a validation study. Mod Pathol 17(12): 1545–1554. , DOI 10.1038/modpathol.3800229 Harvey JM, Clark GM, Osborne CK, Allred DC, 1999, Estrogen receptor status by immunohistochemistry is superior to the ligandbinding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol 17(5):1474–1481. https://doi. org/10.1200/JCO.1999.17.5.1474 Wolff AC, Hammond MEH, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JMS, Bilous M, Fitzgibbons P, Hanna W, Jenkins RB, Mangu PB, Paik S, Perez EA, Press MF, Spears PA, Vance GH, Viale G, Hayes DF, 2013, Recommendations for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Update. J Clin Oncol 31(31):3997. , DOI 10. 1200/Jco.2013.50.9984 Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D, 2000, Molecular portraits of human breast tumours. Nature 406(6797):747–752. , DOI 10.1038/35021093 Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, Thorsen T, Quist H,Matese JC, Brown PO, Botstein D, Lonning PE, Borresen-Dale AL, 2001, Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98(19):10869–10874. , DOI 10.1073/pnas. 191367098 Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H, Pesich R, Geisler S, 2003, Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci 100(14):8418–8423 Hu Z, Fan C, Oh DS, Marron JS, He X, Qaqish BF, Livasy C, Carey LA, Reynolds E, Dressler L, Nobel A, Parker J, Ewend MG, Sawyer LR, Wu J, Liu Y, Nanda R, Tretiakova M, Ruiz Orrico A, Dreher D, Palazzo JP, Perreard L, Nelson E, Mone M, Hansen H, Mullins M, Quackenbush JF, Ellis MJ, Olopade OI, Bernard PS, Perou CM, 2006, The molecular portraits of breast tumors are conserved across microarray platforms. BMC Genomics 7(1):96. , DOI 10.1186/1471-2164-7-96 Herschkowitz JI, Simin K,Weigman VJ, Mikaelian I, Usary J, Hu ZY, Rasmussen KE, Jones LP, Assefnia S, Chandrasekharan S, Backlund MG, Yin YZ, Khramtsov AI, Bastein R, Quackenbush J, Glazer RI, Brown PH, Green JE, Kopelovich L, Furth PA, Palazzo JP, Olopade OI, Bernard PS, Churchill GA, Van Dyke T, Perou CM, 2007, Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol 8(5):R76. , DOI 10.1186/gb- 2007-8-5-r76 Prat A, Parker JS, Karginova O, Fan C, Livasy C, Herschkowitz JI, He X, Perou CM, 2010, Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res 12(5):R68. , DOI 10.1186/bcr2635 Sabatier R, Finetti P, Guille A, Adelaide J, Chaffanet M, Viens P, Birnbaum D, Bertucci F, 2014, Claudin-low breast cancers: clinical, pathological, molecular and prognostic characterization. Mol Cancer 13(1):228. , DOI 10.1186/1476-4598-13-228 Goldhirsch A,WoodWC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, Members P, 2011, Strategies for subtypes-dealing with the diversity of breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol 22(8):1736–1747. , DOI 10.1093/annonc/mdr304 Cheang MC, Chia SK, Voduc D, Gao D, Leung S, Snider J, Watson M, Davies S, Bernard PS, Parker JS, Perou CM, Ellis MJ, Nielsen TO, 2009, Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 101(10):736–750. , DOI 10.1093/jnci/djp082 Inwald EC, Koller M, Klinkhammer-Schalke M, Zeman F, Hofstadter F, Gerstenhauer M, Brockhoff G, Ortmann O, 2015, 4-IHC classification of breast cancer subtypes in a large cohort of a clinical cancer registry: use in clinical routine for therapeutic decisions and its effect on survival. Breast Cancer Res Treat 153(3):647–658. , DOI 10.1007/s10549-015-3572-3 Prat A, CheangMCU, Martin M, Parker JS, Carrasco E, Caballero R, Tyldesley S, Gelmon K, Bernard PS, Nielsen TO, 2012, Prognostic significance of progesterone receptor–positive tumor cells within immunohistochemically defined luminal A breast cancer. Journal of Clinical Oncology:JCO. 2012.2043. 4134., DOI , DOI 10.1200/JCO.2012.43.4134 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, Senn H-J, Albain KS, Andre F, Bergh J, 2013, Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol 24(9):2206–2223. , DOI 10.1093/annonc/mdt303 Tischkowitz M, Brunet JS, Begin LR, Huntsman DG, Cheang MC, Akslen LA, Nielsen TO, Foulkes WD, 2007, Use of immunohistochemical markers can refine prognosis in triple negative breast cancer. BMC Cancer 7(1):134. , DOI 10.1186/ 1471-2407-7-134 Rao C, Shetty J, Prasad KH, 2013, Immunohistochemical profile and morphology in triple - negative breast cancers. Journal of clinical and diagnostic research : JCDR 7(7):1361–1365. https:// doi.org/10.7860/JCDR/2013/5823.3129 Prat A, Adamo B, Cheang MCU, Anders CK, Carey LA, Perou CM, 2013, Molecular characterization of basal-like and nonbasal- like triple-negative breast cancer. Oncologist 18(2):123–133 Pogoda K, Niwinska A, Murawska M, Olszewski W, Nowecki Z, 2014, The outcome of special histologic types of triple-negative breast cancer, TNBC). In: ASCO Annual Meeting Proceedings. vol 15_suppl. p 1122., DOI , DOI 10.1200/jco.2014.32.15_ suppl.1122 Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C, Cowan D, Dressler L, Akslen LA, Ragaz J, Gown AM, Gilks CB, van de Rijn M, Perou CM, 2004, Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res 10(16):5367–5374. , DOI 10.1158/1078-0432.CCR-04- 0220 Senkus E, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rutgers E, Zackrisson S, Cardoso F, Committee EG, 2015, Primary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 26(Suppl 5):v8– , DOI 10.1093/annonc/mdv298 30. Coates AS, Winer EP, Goldhirsch A, Gelber RD, Gnant M, Piccart-Gebhart M, Thurlimann B, Senn HJ, Panel M, 2015, Tailoring therapies–improving the management of early breast cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015. Ann Oncol 26(8):1533– 1546. , DOI 10.1093/annonc/mdv221 Neuman HB, Morrogh M, Gonen M, Van Zee KJ, Morrow M, King TA, 2010, Stage IV breast cancer in the era of targeted therapy: does surgery of the primary tumor matter? Cancer 116(5):1226–1233. , DOI 10.1002/cncr.24873 Rashaan ZM, Bastiaannet E, Portielje JEA, van de Water W, van der Velde S, Ernst MF, van de Velde CJH, Liefers GJ, 2012, Surgery in metastatic breast cancer: Patients with a favorable profile seem to have the most benefit from surgery. Ejso-Eur J Surg Onc 38(1):52–56. , DOI 10.1016/j.ejso.2011.10.004 Ly BH, Vlastos G, Rapiti E, Vinh-Hung V, Nguyen NP, 2010, Local-regional radiotherapy and surgery is associated with a significant survival advantage in metastatic breast cancer patients. Tumori 96(6):947–954 Morrow M, Goldstein L, 2006, Surgery of the primary tumor in metastatic breast cancer: closing the barn door after the horse has bolted? J Clin Oncol 24(18):2694–2696. , DOI 10.1200/ JCO.2006.05.9824 Cardoso F, Costa A, Norton L, Senkus E, Aapro M, Andre F, Barrios CH, Bergh J, Biganzoli L, Blackwell KL, 2014, ESOESMO2nd international consensus guidelines for advanced breast cancer, ABC2). Breast 23(5):489–502. , DOI 10.1016/j. breast.2014.08.009 Pritchard KI, 2003, Endocrine therapy of advanced disease: analysis and implications of the existing data. Clin Cancer Res 9(1 Pt 2):460S–467S Lumachi F, Luisetto G, Basso SM, Basso U, Brunello A, Camozzi V, 2011, Endocrine therapy of breast cancer. Curr Med Chem 18(4):513–522. , DOI 10.2174/092986711794480177 Olson E,Mullins DA, 2013)When standard therapy fails in breast cancer: current and future options for HER2-positive disease. J Clin Trials 3:1000129. , DOI 10.4172/2167-0870. 1000129 Singh JC, Jhaveri K, Esteva FJ, 2014, HER2-positive advanced breast cancer: optimizing patient outcomes and opportunities for drug development. Br J Cancer 111(10):1888–1898. https://doi. org/10.1038/bjc.2014.388 Twelves C, Jove M, Gombos A, Awada A, 2016, Cytotoxic chemotherapy: Still the mainstay of clinical practice for all subtypes metastatic breast cancer. Crit Rev Oncol Hematol 100:74–87. , DOI 10.1016/j.critrevonc.2016.01.021 Li BT,WongMH, Pavlakis N, 2014, Treatment and Prevention of Bone Metastases from Breast Cancer: A Comprehensive Review of Evidence for Clinical Practice. J Clin Med 3(1):1–24. https:// doi.org/10.3390/jcm3010001 Ellis MJ, Suman VJ, Hoog J, Lin L, Snider J, Prat A, Parker JS, Luo JQ, DeSchryver K, Allred DC, Esserman LJ, Unzeitig GW, Margenthaler J, Babiera GV, Marcom PK, Guenther JM, Watson MA, Leitch M, Hunt K, Olson JA, 2011, Randomized Phase II Neoadjuvant Comparison Between Letrozole, Anastrozole, and Exemestane for Postmenopausal Women With Estrogen Receptor-Rich Stage 2 to 3 Breast Cancer: Clinical and Biomarker Outcomes and Predictive Value of the Baseline PAM50-Based Intrinsic Subtype-ACOSOG Z1031. J Clin Oncol 29(17):2342–2349. , DOI 10.1200/Jco.2010.31.6950 Cataliotti L, Buzdar AU, Noguchi S, Bines J, Takatsuka Y, Petrakova K, Dube P, de Oliveira CT, 2006, Comparison of anastrozole versus tamoxifen as preoperative therapy in postmenopausal women with hormone receptor-positive breast cancer - The Pre-Operative "Arimidex" Compared to Tamoxilen, PROAC7, trial. Cancer 106(10):2095–2103. , DOI 10. 1002/cncr.21872 Dowsett M, Smith IE, Ebbs SR, Dixon JM, Skene A, A'Hern R, Salter J, Detre S, Hills M, Walsh G, Group IT, 2007, Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer. J Natl Cancer Inst 99(2):167– 170. , DOI 10.1093/jnci/djk020 Dunbier AK, Anderson H, Ghazoui Z, Salter J, Parker JS, Perou CM, Smith IE, Dowsett M, 2011, Association between breast cancer subtypes and response to neoadjuvant anastrozole. Steroids 76(8):736–740. , DOI 10.1016/j.steroids.2011. 02.025 Davidson NE, O'Neill AM, Vukov AM, Osborne CK, Martino S, White DR, Abeloff MD, 2005, Chemoendocrine therapy for premenopausal women with axillary lymph node-positive, steroid hormone receptor-positive breast cancer: results from INT 0101, E5188). J Clin Oncol 23(25):5973–5982. , DOI 10.1200/ JCO.2005.05.551 LHRH-agonists in Early Breast Cancer Overview group, 2007, Use of luteinising-hormone-releasing hormone agonists as adjuvant treatment in premenopausal patients with hormone-receptorpositive breast cancer: a meta-analysis of individual patient data from randomised adjuvant trials. Lancet 369(9574):1711–1723 Francis PA, Regan MM, Fleming GF, Lang I, Ciruelos E, Bellet M, Bonnefoi HR, Climent MA, Da Prada GA, Burstein HJ, 2015, Adjuvant ovarian suppression in premenopausal breast cancer. N Engl J Med 372(5):436–446. , DOI 10.1056/ NEJMoa1412379 McGuire WL, 1978, Steroid receptors in human breast cancer. Cancer Res 38(11 Pt 2):4289–4291 Paik S, Tang G, Shak S, Kim C, Baker J, Kim W, Cronin M, Baehner FL, Watson D, Bryant J, Costantino JP, Geyer CE Jr, Wickerham DL, Wolmark N, 2006, Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptorpositive breast cancer. J Clin Oncol 24(23):3726–3734. https:// doi.org/10.1200/JCO.2005.04.7985 Albain KS, BarlowWE, Shak S, Hortobagyi GN, Livingston RB, Yeh IT, Ravdin P, Bugarini R, Baehner FL, Davidson NE, Sledge GW, Winer EP, Hudis C, Ingle JN, Perez EA, Pritchard KI, Shepherd L, Gralow JR, Yoshizawa C, Allred DC, Osborne CK, Hayes DF, Breast Cancer Intergroup of North A, 2010, Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptorpositive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol 11(1):55–65. , DOI 10.1016/S1470-2045(09)70314-6 Dowsett M, Allred C, Knox J, Quinn E, Salter J,Wale C, Cuzick J, Houghton J, Williams N, Mallon E, Bishop H, Ellis I, Larsimont D, Sasano H, Carder P, Cussac AL, Knox F, Speirs V, Forbes J, Buzdar A, 2008, Relationship between quantitative estrogen and progesterone receptor expression and human epidermal growth factor receptor 2, HER-2, status with recurrence in the arimidex, tamoxifen, alone or in combination trial. J Clin Oncol 26(7):1059– 1065. , DOI 10.1200/Jco.2007.12.9437 Johnston S, Pippen J, Pivot X, Lichinitser M, Sadeghi S, Dieras V, Gomez HL, Romieu G, Manikhas A, Kennedy MJ, Press MF, Maltzman J, Florance A, O'Rourke L, Oliva C, Stein S, Pegram M, 2009, Lapatinib Combined With Letrozole Versus Letrozole and Placebo As First-Line Therapy for Postmenopausal Hormone Receptor-Positive Metastatic Breast Cancer. J Clin Oncol 27(33): 5538–5546. , DOI 10.1200/Jco.2009.23.3734 Kaufman B, Mackey JR, Clemens MR, Bapsy PP, Vaid A, Wardley A, Tjulandin S, Jahn M, Lehle M, Feyereislova A, Revil C, Jones A, 2009, Trastuzumab plus anastrozole versus anastrozole alone for the treatment of postmenopausal women with human epidermal growth factor receptor 2-positive, hormone receptor-positive metastatic breast cancer: results from the randomized phase III TAnDEM study. J Clin Oncol 27(33):5529– 5537. , DOI 10.1200/JCO.2008.20.6847 Koeberle D, Ruhstaller T, Jost L, Pagani O, Zaman K, von Moos R, Oehlschlegel C, Crowe S, Pilop C, Thuerlimann B, Swiss Group for Clinical Cancer R, 2011, Combination of trastuzumab and letrozole after resistance to sequential trastuzumab and aromatase inhibitor monotherapies in patients with estrogen receptorpositive, HER-2-positive advanced breast cancer: a proof-ofconcept trial, SAKK 23/03). Endocr Relat Cancer 18(2):257– 264. , DOI 10.1530/ERC-10-0317 Vogel CL, Cobleigh MA, Tripathy D, Gutheil JC, Harris LN, Fehrenbacher L, Slamon DJ, Murphy M, Novotny WF, Burchmore M, Shak S, Stewart SJ, Press M, 2002, Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J Clin Oncol 20(3):719–726. , DOI 10.1200/Jco.20.3.719 Gianni L, Dafni U, Gelber RD, Azambuja E, Muehlbauer S, Goldhirsch A, Untch M, Smith I, Baselga J, Jackisch C, Cameron D, Mano M, Pedrini JL, Veronesi A, Mendiola C, Pluzanska A, Semiglazov V, Vrdoljak E, Eckart MJ, Shen Z, Skiadopoulos G, Procter M, Pritchard KI, Piccart-Gebhart MJ, Bell R, Herceptin Adjuvant Trial Study T, 2011, Treatment with trastuzumab for 1 year after adjuvant chemotherapy in patients with HER2-positive early breast cancer: a 4-year follow-up of a randomised controlled trial. Lancet Oncol 12(3):236–244. https:// doi.org/10.1016/S1470-2045(11)70033-X Perez EA, Romond EH, Suman VJ, Jeong JH, Sledge G, Geyer CE,Martino S, Rastogi P, Gralow J, Swain SM,Winer EP, Colon- Otero G, Davidson NE, Mamounas E, Zujewski JA, Wolmark N, 2014, Trastuzumab Plus Adjuvant Chemotherapy for Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer: Planned Joint Analysis of Overall Survival From NSABP B-31 and NCCTG N9831. J Clin Oncol 32(33):3744–3752. https://doi. org/10.1200/Jco.2014.55.5730 Slamon D, EiermannW, Robert N, Pienkowski T,MartinM, Press M, Mackey J, Glaspy J, Chan A, Pawlicki M, Pinter T, Valero V, Liu MC, Sauter G, von Minckwitz G, Visco F, Bee V, Buyse M, Bendahmane B, Tabah-Fisch I, Lindsay MA, Riva A, Crown J, Breast Cancer International Research G, 2011, Adjuvant trastuzumab in HER2-positive breast cancer. N Engl J Med 365(14):1273–1283. , DOI 10.1056/NEJMoa0910383 Tan-Chiu E, Yothers G, Romond E, Geyer CE, Ewer M, Keefe D, Shannon RP, Swain SM, Brown A, Fehrenbacher L, Vogel VG, Seay TE, Rastogi P, Mamounas EP, Wolmark N, Bryant J, 2005, Assessment of cardiac dysfunction in a randomized trial comparing doxorubicin and cyclophosphamide followed by paclitaxel, with or without trastuzumab as adjuvant therapy in node-positive, human epidermal growth factor receptor 2-overexpressing breast cancer: NSABP B-31. J Clin Oncol 23(31):7811–7819. https:// doi.org/10.1200/Jco.2005.02.4091 Liedtke C, Mazouni C, Hess KR, Andre F, Tordai A, Mejia JA, Symmans WF, Gonzalez-Angulo AM, Hennessy B, Green M, Cristofanilli M, Hortobagyi GN, Pusztai L, 2008, Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J Clin Oncol 26(8):1275–1281. , DOI 10.1200/JCO.2007.14.4147 Rouzier R, Perou CM, SymmansWF, Ibrahim N, Cristofanilli M, Anderson K, Hess KR, Stec J, Ayers M,Wagner P,Morandi P, Fan C, Rabiul I, Ross JS, Hortobagyi GN, Pusztai L, 2005, Breast cancer molecular subtypes respond differently to preoperative chemotherapy. Clin Cancer Res 11(16):5678–5685. , DOI 10.1158/1078-0432.CCR-04-2421 Carey LA, Dees EC, Sawyer L, Gatti L, Moore DT, Collichio F, Ollila DW, Sartor CI, Graham ML, Perou CM, 2007, The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes. Clin Cancer Res 13(8):2329–2334. , DOI 10.1158/1078-0432.CCR-06-1109 Peshkin BN, Alabek ML, Isaacs C, 2010, BRCA1/2 mutations and triple negative breast cancers. Breast Dis 32(1-2):25–33. , DOI 10.3233/BD-2010-0306 Byrski T, Gronwald J, Huzarski T, Grzybowska E, Budryk M, Stawicka M, Mierzwa T, Szwiec M, Wisniowski R, Siolek M, Dent R, Lubinski J, Narod S, 2010, Pathologic complete response rates in young women with BRCA1-positive breast cancers after neoadjuvant chemotherapy. J Clin Oncol 28(3):375–379. https:// doi.org/10.1200/JCO.2008.20.7019 Gronwald J, Byrski T, Huzarski T, Dent R, Bielicka V, Zuziak D, Wisniowski R, Lubinski J, Narod S, 2009, Neoadjuvant therapy with cisplatin in BRCA1-positive breast cancer patients. In: ASCO Annual Meeting Proceedings. vol 15S. p 502., DOI https:// doi.org/10.1200/jco.2009.27.15s.502 Leone JP, Guardiola V, Venkatraman A, Pegram MD, Welsh C, Silva O, Larrieux R, Franchesci D, Gomez C, Hurley J, 2009, Neoadjuvant platinum-based chemotherapy, CT, for triplenegative locally advanced breast cancer, LABC): retrospective analysis of 125 patients. In: ASCO Annual Meeting Proceedings. vol 15S. p 625., DOI , DOI 10.1200/jco. 2009.27.15_suppl.625 Sirohi B, Arnedos M, Popat S, Ashley S, Nerurkar A, Walsh G, Johnston S, Smith IE, 2008, Platinum-based chemotherapy in triple-negative breast cancer. Ann Oncol 19(11):1847–1852. , DOI 10.1093/annonc/mdn395 Liu M, Mo QG, Wei CY, Qin QH, Huang Z, He J, 2013, Platinum-based chemotherapy in triple-negative breast cancer: A meta-analysis. Oncol Lett 5(3):983–991. , DOI 10.3892/ ol.2012.1093 Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, Pietenpol JA, 2011, Identification of human triplenegative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 121(7):2750–2767. https:// doi.org/10.1172/JCI45014 Bertucci F, Finetti P, Cervera N, Charafe-Jauffret E, Mamessier E, Adelaide J, Debono S, Houvenaeghel G, Maraninchi D, Viens P, Charpin C, Jacquemier J, Birnbaum D, 2006, Gene expression profiling shows medullary breast cancer is a subgroup of basal breast cancers. Cancer Res 66(9):4636–4644. , DOI 10. 1158/0008-5472.CAN-06-0031 Marginean F, Rakha EA, Ho BC, Ellis IO, Lee AH, 2010, Histological features of medullary carcinoma and prognosis in triple-negative basal-like carcinomas of the breast. Mod Pathol 23(10):1357–1363. , DOI 10.1038/modpathol.2010.123 Hennessy BT, Gonzalez-Angulo AM, Stemke-Hale K, Gilcrease MZ, Krishnamurthy S, Lee JS, Fridlyand J, Sahin A, Agarwal R, Joy C, Liu W, Stivers D, Baggerly K, Carey M, Lluch A, Monteagudo C, He X, Weigman V, Fan C, Palazzo J, Hortobagyi GN, Nolden LK, Wang NJ, Valero V, Gray JW, Perou CM, Mills GB, 2009, Characterization of a naturally occurring breast cancer subset enriched in epithelial-to-mesenchymal transition and stem cell characteristics. Cancer Res 69(10):4116– 4124. , DOI 10.1158/0008-5472.CAN-08-3441 Farmer P, Bonnefoi H, Becette V, Tubiana-Hulin M, Fumoleau P, Larsimont D, Macgrogan G, Bergh J, Cameron D, Goldstein D, Duss S, Nicoulaz AL, Brisken C, Fiche M, Delorenzi M, Iggo R, 2005, Identification of molecular apocrine breast tumours by microarray analysis. Oncogene 24(29):4660–4671. , DOI 10.1038/sj.onc.1208561 Masuda H, Baggerly KA, Wang Y, Zhang Y, Gonzalez-Angulo AM, Meric-Bernstam F, Valero V, Lehmann BD, Pietenpol JA, Hortobagyi GN, Symmans WF, Ueno NT, 2013, Differential Response to Neoadjuvant Chemotherapy Among 7 Triple- Negative Breast Cancer Molecular Subtypes. Clin Cancer Res 19(19):5533–5540. , DOI 10.1158/1078-0432.CCR-13- 0799 BursteinMD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SA, Savage MI, Osborne CK, Hilsenbeck SG, Chang JC, Mills GB, Lau CC, Brown PH, 2015, Comprehensive genomic analysis identifies novel subtypes and targets of triplenegative breast cancer. Clin Cancer Res 21(7):1688–1698. , DOI 10.1158/1078-0432.CCR-14-0432 Jezequel P, Loussouarn D, Guerin-Charbonnel C, Campion L, Vanier A, Gouraud W, Lasla H, Guette C, Valo I, Verriele V, Campone M, 2015, Gene-expression molecular subtyping of triple-negative breast cancer tumours: importance of immune response. Breast Cancer Res 17(1):43. , DOI 10.1186/ s13058-015-0550-y The Cancer Genome Atlas Network, TCGA,, 2012, Comprehensive molecular portraits of human breast tumours. Nature 490(7418):61–70 Abramson VG, Lehmann BD, Ballinger TJ, Pietenpol JA, 2015, Subtyping of triple-negative breast cancer: implications for therapy. Cancer 121(1):8–16. , DOI 10.1002/cncr.28914 Gonzalez-Angulo AM, Stemke-Hale K, Palla SL, Carey M, Agarwal R, Meric-Berstam F, Traina TA, Hudis C, Hortobagyi GN, Gerald WL, Mills GB, Hennessy BT, 2009, Androgen receptor levels and association with PIK3CA mutations and prognosis in breast cancer. Clin Cancer Res 15(7):2472–2478. https:// doi.org/10.1158/1078-0432.CCR-08-1763 Lehmann BD, Bauer JA, Schafer JM, Pendleton CS, Tang L, Johnson KC, Chen X, Balko JM, Gomez H, Arteaga CL, Mills GB, Sanders ME, Pietenpol JA, 2014, PIK3CA mutations in androgen receptor-positive triple negative breast cancer confer sensitivity to the combination of PI3K and androgen receptor inhibitors. Breast Cancer Res 16(4):406. , DOI 10.1186/ s13058-014-0406-x De Summa S, Pinto R, Sambiasi D, Petriella D, Paradiso V, Paradiso A, Tommasi S, 2013, BRCAness: a deeper insight into basal-like breast tumors. Ann Oncol 24(suppl 8):viii13–viii21 Kenemans P, Verstraeten RA, Verheijen RH, 2004, Oncogenic pathways in hereditary and sporadic breast cancer. Maturitas 49(1):34–43. , DOI 10.1016/j.maturitas.2004.06.005 Audeh MW, 2014, Novel treatment strategies in triple-negative breast cancer: specific role of poly(adenosine diphosphate-ribose, polymerase inhibition. Pharmgenomics Pers Med 7:307–316. , DOI 10.2147/PGPM.S39765 Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, Mortimer P, Swaisland H, Lau A, O'Connor MJ, Ashworth A, Carmichael J, Kaye SB, Schellens JH, de Bono JS, 2009, Inhibition of poly(ADP-ribose, polymerase in tumors from BRCA mutation carriers. N Engl J Med 361(2):123–134. https:// doi.org/10.1056/NEJMoa0900212 Gelmon KA, TischkowitzM, Mackay H, Swenerton K, Robidoux A, Tonkin K, Hirte H, Huntsman D, Clemons M, Gilks B, Yerushalmi R, Macpherson E, Carmichael J, Oza A, 2011, Olaparib in patients with recurrent high-grade serous or poorly differentiated ovarian carcinoma or triple-negative breast cancer: a phase 2, multicentre, open-label, non-randomised study. Lancet Oncol 12(9):852–861. , DOI 10.1016/S1470-2045(11, 70214-5 Tutt A, RobsonM, Garber JE, Domchek SM, AudehMW,Weitzel JN, Friedlander M, Arun B, Loman N, Schmutzler RK, Wardley A, Mitchell G, Earl H, Wickens M, Carmichael J, 2010, Oral poly(ADP-ribose, polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet 376(9737):235–244. https://doi. org/10.1016/S0140-6736(10)60892-6 Isakoff SJ, Overmoyer B, Tung NM, Gelman RS, Giranda VL, Bernhard KM, Habin KR, Ellisen LW,Winer EP, Goss PE, 2010, A phase II trial of the PARP inhibitor veliparib, ABT888, and temozolomide for metastatic breast cancer. In: ASCO Annual Meeting Proceedings. vol 15_suppl. p 1019., DOI , DOI 10.1200/jco.2010.28.15_suppl.1019 O'Shaughnessy J, Osborne C, Pippen JE, Yoffe M, Patt D, Rocha C, Koo IC, Sherman BM, Bradley C, 2011, Iniparib plus chemotherapy in metastatic triple-negative breast cancer. N Engl J Med 364(3):205–214. , DOI 10.1056/NEJMoa1011418 O'Shaughnessy J, Schwartzberg L, Danso MA, Miller KD, Rugo HS, Neubauer M, Robert N, Hellerstedt B, Saleh M, Richards P, Specht JM, Yardley DA, Carlson RW, Finn RS, Charpentier E, Garcia-Ribas I, Winer EP, 2014, Phase III study of iniparib plus gemcitabine and carboplatin versus gemcitabine and carboplatin in patients with metastatic triple-negative breast cancer. J Clin Oncol 32(34):3840–3847. , DOI 10.1200/JCO.2014.55. 2984 Dent RA, Lindeman GJ, Clemons M, Wildiers H, Chan A, McCarthy NJ, Singer CF, Lowe ES, Watkins CL, Carmichael J, 2013, Phase I trial of the oral PARP inhibitor olaparib in combination with paclitaxel for first- or second-line treatment of patients with metastatic triple-negative breast cancer. Breast Cancer Res 15(5):R88. , DOI 10.1186/bcr3484 Watkins J, Weekes D, Shah V, Gazinska P, Joshi S, Sidhu B, Gillett C, Pinder S, Vanoli F, Jasin M, Mayrhofer M, Isaksson A, Cheang MCU, Mirza H, Frankum J, Lord CJ, Ashworth A, Vinayak S, Ford JM, Telli ML, Grigoriadis A, Tutt ANJ, 2015, Genomic Complexity Profiling Reveals That HORMAD1 Overexpression Contributes to Homologous Recombination Deficiency in Triple-Negative Breast Cancers. Cancer Discovery 5(5):488–505. , DOI 10.1158/2159-8290.CD-14-1092 Mayer IA, Jovanovic B, Abramson VG, Mayer EL, Sanders ME, Bardia A, Dillon PM, Kuba MG, Carpenter JT, Chang JC, Lehmann BD, Meszoely IM, Grau A, Shyr Y, Arteaga CL, Chen X, Pietenpol JA, 2013, A randomized phase II neoadjuvant study of cisplatin, paclitaxel with or without everolimus, an mTOR inhibitor, in patients with stage II/III triple-negative breast cancer, TNBC). Cancer Res 73, 24 Supplement):PD1-6-PD1-6., DOI , DOI 10.1158/0008-5472.SABCS13-PD1-6 Gonzalez-Angulo AM, Green MC, Murray JL, Palla SL, Koenig KH, Brewster AM, Valero V, Ibrahim NK,Moulder SL, Litton JK, 2011, Open label, randomized clinical trial of standard neoadjuvant chemotherapy with paclitaxel followed by FEC, T-FEC, versus the combination of paclitaxel and RAD001 followed by FEC, TR-FEC, in women with triple receptor-negative breast cancer, TNBC). In: ASCO Annual Meeting Proceedings. vol 15_suppl. p 1016., DOI , DOI 10.1200/jco.2011.29.15_suppl.1016 Singh J, Novik Y, Stein S, VolmM,Meyers M, Smith J, Omene C, Speyer J, Schneider R, Jhaveri K, Formenti S, Kyriakou V, Joseph B, Goldberg JD, Li X, Adams S, Tiersten A, 2014, Phase 2 trial of everolimus and carboplatin combination in patients with triple negative metastatic breast cancer. Breast Cancer Res 16(2):R32. , DOI 10.1186/bcr3634 Edelman G, Bedell C, ShapiroG, Pandya SS, Kwak EL, Scheffold C, Nguyen LT, Laird A, Baselga J, Rodon J, 2010, Aphase I doseescalation study of XL147, SAR245408), a PI3K inhibitor administered orally to patients, pts, with advanced malignancies. J Clin Oncol 28(15):3004. , DOI 10.1200/jco.2010.28.15_ suppl.3004 Jimeno A, Herbst RS, Falchook GS, Messersmith WA, Hecker S, Peterson S, Hausman DF, Kurzrock R, Eckhardt SG, Hong DS, 2010, Final results from a phase I, dose-escalation study of PX- 866, an irreversible, pan-isoform inhibitor of PI3 kinase. In: ASCO Annual Meeting Proceedings. vol 15_suppl. p 3089., DOI , DOI 10.1200/jco.2010.28.15_suppl.3089 Patnaik A, Appleman LJ, Mountz JM, Ramanathan RK, Beeram M, Tolcher AW, Papadopoulos KP, Lotze MT, Petro DP, Laymon C, 2011, A first-in-human phase I study of intravenous PI3K inhibitor BAY 80-6946 in patients with advanced solid tumors: Results of dose-escalation phase. In: ASCO Annual Meeting Proceedings. vol 15_suppl. p 3035., DOI , DOI 10.1200/ jco.2011.29.15_suppl.3035 Von Hoff DD, LoRusso P, Demetri GD, Weiss GJ, Shapiro G, Ramanathan RK,Ware JA, Raja R, Jin J, LevyGG(2011)Aphase I dose-escalation study to evaluate GDC-0941, a pan-PI3K inhibitor, administered QD or BID in patients with advanced or metastatic solid tumors. In: ASCO annual meeting proceedings. vol 15_suppl. p 3052 Lin J, Sampath D, Nannini MA, Lee BB, Degtyarev M, Oeh J, Savage H, Guan ZY, Hong R, Kassees R, Lee LB, Risom T, Gross S, Liederer BM, Koeppen H, Skelton NJ, Wallin JJ, Belvin M, Punnoose E, Friedman LS, Lin K, 2013, Targeting Activated Akt with GDC-0068, a Novel Selective Akt Inhibitor That Is Efficacious in Multiple Tumor Models. Clin Cancer Res 19(7): 1760–1772. , DOI 10.1158/1078-0432.CCR-12-3072 OliveiraM, Saura C, Gonzalez-Martin A, Andersen JC, Fisher JG, Calvo I, Ciruelos E, Gil M, De La Pena L, Llobet-Canela M, 2015, FAIRLANE: A phase II randomized, double-blind, study of the Akt inhibitor ipatasertib, Ipat, GDC-0068, in combination with paclitaxel, Pac, as neoadjuvant treatment for early stage triple-negative breast cancer, TNBC). In: ASCO Annual Meeting Proceedings. vol 15_suppl. p TPS1112., DOI https://doi. org/10.1200/jco.2015.33.15_suppl.tps1112 KimS, TanAR, ImS-A,Villanueva R,ValeroV, Saura C, Oliveira M, Isakoff SJ, Singel SM, Dent RA, 2015, LOTUS: A randomized, phase II, multicenter, placebo-controlled study of ipatasertib, Ipat, GDC-0068), an inhibitor of Akt, in combination with paclitaxel, Pac, as front-line treatment for patients, pts, with metastatic triple-negative breast cancer, TNBC). In: ASCO Annual Meeting Proceedings. vol 15_suppl. p TPS1111., DOI , DOI 10. 1200/jco.2015.33.15_suppl.tps1111 Ibrahim YH, Garcia-Garcia C, Serra V, He L, Torres-Lockhart K, Prat A, Anton P, Cozar P, Guzman M, Grueso J, Rodriguez O, Calvo MT, Aura C, Diez O, Rubio IT, Perez J, Rodon J, Cortes J, Ellisen LW, Scaltriti M, Baselga J, 2012, PI3K Inhibition Impairs BRCA1/2 Expression and Sensitizes BRCA-Proficient Triple- Negative Breast Cancer to PARP Inhibition. Cancer Discovery 2(11):1036–1047. , DOI 10.1158/2159-8290.CD-11-0348 McNamara KM, Moore NL, Hickey TE, Sasano H, Tilley WD, 2014, Complexities of androgen receptor signalling in breast cancer. Endocr Relat Cancer 21(4):T161–T181. , DOI 10. 1530/ERC-14-0243 Park S, Koo J, Park HS, Kim JH, Choi SY, Lee JH, Park BW, Lee KS, 2010, Expression of androgen receptors in primary breast cancer. Ann Oncol 21(3):488–492. , DOI 10.1093/ annonc/mdp510 Traina TA, Miller K, Yardley DA, O'Shaughnessy J, Cortes J, Awada A, Kelly CM, Trudeau ME, Schmid P, Gianni L, 2015, Results from a phase 2 study of enzalutamide, ENZA), an androgen receptor, AR, inhibitor, in advanced AR+ triple-negative breast cancer, TNBC). In: ASCO Annual Meeting Proceedings. vol 15_suppl. p 1003., DOI , DOI 10.1200/jco.2015.33. 15_suppl.1003 Yamaoka M, Hara T, Hitaka T, Kaku T, Takeuchi T, Takahashi J, Asahi S, Miki H, Tasaka A, Kusaka M, 2012, Orteronel, TAK- 700), a novel non-steroidal 17,20-lyase inhibitor: effects on steroid synthesis in human and monkey adrenal cells and serum steroid levels in cynomolgus monkeys. J Steroid Biochem Mol Biol 129(3-5):115–128. , DOI 10.1016/j.jsbmb.2012.01.001 Gucalp A, Tolaney S, Isakoff SJ, Ingle JN, Liu MC, Carey LA, Blackwell K, Rugo H, Nabell L, Forero A, Stearns V, Doane AS, Danso M, Moynahan ME, Momen LF, Gonzalez JM, Akhtar A, Giri DD, Patil S, Feigin KN, Hudis CA, Traina TA, Translational Breast Cancer Research C, 2013, Phase II trial of bicalutamide in patients with androgen receptor-positive, estrogen receptornegative metastatic Breast Cancer. Clin Cancer Res 19(19): 5505–5512. , DOI 10.1158/1078-0432.CCR-12-3327 Hu X, Zhang J, Xu B, Jiang Z, Ragaz J, Tong Z, Zhang Q, Wang X, Feng J, Pang D, Fan M, Li J,Wang B,Wang Z, Zhang Q, Sun S, Liao C, 2014, Multicenter phase II study of apatinib, a novel VEGFR inhibitor in heavily pretreated patients with metastatic triple-negative breast cancer. Int J Cancer 135(8):1961–1969. , DOI 10.1002/ijc.28829 Baselga J, Gomez P, Greil R, Braga S, ClimentMA,Wardley AM, Kaufman B, Stemmer SM, Pego A, Chan A, Goeminne JC, Graas MP, Kennedy MJ, Ciruelos Gil EM, Schneeweiss A, Zubel A, Groos J, Melezinkova H, Awada A, 2013, Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer. J Clin Oncol 31(20): 2586–2592. , DOI 10.1200/JCO.2012.46.2408 Carey LA, Rugo HS, Marcom PK, Mayer EL, Esteva FJ, Ma CX, Liu MC, Storniolo AM, Rimawi MF, Forero-Torres A,Wolff AC, Hobday TJ, Ivanova A, ChiuWK, Ferraro M, Burrows E, Bernard PS, Hoadley KA, Perou CM, Winer EP, 2012, TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol 30(21):2615–2623. , DOI 10.1200/JCO.2010.34. 5579 Tolaney SM, Tan S, Guo H, Barry W, Van Allen E, Wagle N, Brock J, Larrabee K, Paweletz C, Ivanova E, Janne P, Overmoyer B, Wright JJ, Shapiro GI, Winer EP, Krop IE, 2015, Phase II study of tivantinib, ARQ 197, in patients with metastatic triple-negative breast cancer. Investig New Drugs 33(5):1108– 1114. , DOI 10.1007/s10637-015-0269-8 Hicklin DJ, EllisLM(2005, Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol 23(5):1011–1027. , DOI 10.1200/JCO.2005.06.081 Linderholm BK, Hellborg H, Johansson U, Elmberger G, Skoog L, Lehtio J, Lewensohn R, 2009, Significantly higher levels of vascular endothelial growth factor, VEGF, and shorter survival times for patients with primary operable triple-negative breast cancer. Ann Oncol 20(10):1639–1646. , DOI 10.1093/ annonc/mdp062 Robert NJ, Dieras V, Glaspy J, Brufsky A, Bondarenko I, Lipatov O, Perez E, Yardley D, Zhou X, Phan S, 2009, RIBBON-1: randomized, double-blind, placebo-controlled, phase III trial of chemotherapy with or without bevacizumab, B, for first-line treatment of HER2-negative locally recurrent or metastatic breast cancer, MBC). In: ASCO Annual Meeting Proceedings. vol 15S. p 1005., DOI , DOI 10.1200/jco.2009.27.15s.1005 Miles D, Chan A, Romieu G, Dirix LY, Cortes J, Pivot X, Tomczak P, Taran T, Harbeck N, Steger GG, 2008, Randomized, double-blind, placebo-controlled, phase III study of bevacizumab with docetaxel or docetaxel with placebo as first-line therapy for patients with locally recurrent or metastatic breast cancer, mBC): AVADO. In: ASCO Annual Meeting Proceedings. vol 15_suppl. p LBA1011., DOI , DOI 10. 1200/jco.2008.26.15_suppl.lba1011 Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, Shenkier T, Cella D, Davidson NE, 2007, Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med 357(26):2666–2676. , DOI 10.1056/ NEJMoa072113 Brufsky A, Valero V, Tiangco B, Dakhil SR, Brize A, Bousfoul N, Rugo HS, Yardley DA, 2011, Impact of bevacizumab, BEV, on efficacy of second-line chemotherapy, CT, for triple-negative breast cancer, TNBC): Analysis of RIBBON-2. In: ASCO Annual Meeting Proceedings. vol 15_suppl. p 1010., DOI https:// doi.org/10.1200/jco.2011.29.15_suppl.1010 Cameron D, Brown J, Dent R, Jackisch C, Mackey J, Pivot X, Steger GG, Suter TM, Toi M, Parmar M, Laeufle R, Im YH, Romieu G, Harvey V, Lipatov O, Pienkowski T, Cottu P, Chan A, Im SA, Hall PS, Bubuteishvili-Pacaud L, Henschel V, Deurloo RJ, Pallaud C, Bell R, 2013, Adjuvant bevacizumab-containing therapy in triple-negative breast cancer, BEATRICE): primary results of a randomised, phase 3 trial. LancetOncol 14(10):933–942. , DOI 10.1016/S1470-2045(13)70335-8 Carpenter D, Kesselheim AS, Joffe S, 2011, Reputation and precedent in the bevacizumab decision. N Engl J Med 365(2):e3. , DOI 10.1056/NEJMp1107201 Livasy CA, Karaca G, Nanda R, Tretiakova MS, Olopade OI, Moore DT, Perou CM, 2006, Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol 19(2):264–271. , DOI 10.1038/modpathol.3800528 De Luca A, D'Alessio A, Maiello MR, Gallo M, Chicchinelli N, Pergameno M, Piccirilli MS, Normanno N, 2015, Evaluation of the pharmacokinetics of ixabepilone for the treatment of breast cancer. Expert Opin Drug Metab Toxicol 11(7):1177–1185. , DOI 10.1517/17425255.2015.1057497 Tredan O, Campone M, Jassem J, Vyzula R, Coudert B, Pacilio C, Prausova J, Hardy-Bessard A-C, Arance A, Mukhopadhyay P, 2015, Ixabepilone alone or with cetuximab as first-line treatment for advanced/metastatic triple-negative breast cancer. Clin Breast Cancer 15(1):8–15. , DOI 10.1016/j.clbc.2014.07.007 Carey LA, O'Shaughnessy JA, Hoadley K, Khambata-Ford S, Horak CE, Xu LA, Awad M, Brickman D, Muller S, Donato J, Asmar L, Stiljeman I, Ebbert M, Bernard P, Perou CM, 2009, Potential Predictive Markers of Benefit from Cetuximab in Metastatic Breast Cancer: An Analysis of Two Randomized Phase 2 Trials. Cancer Res 69(24):596s–596s. , DOI 10. 1158/0008-5472.SABCS-09-2014 Balko JM, Giltnane JM, Wang K, Schwarz LJ, Young CD, Cook RS, Owens P, Sanders ME, Kuba MG, Sanchez V, Kurupi R, Moore PD, Pinto JA, Doimi FD, Gomez H, Horiuchi D, Goga A, Lehmann BD, Bauer JA, Pietenpol JA, Ross JS, Palmer GA, Yelensky R, Cronin M, Miller VA, Stephens PJ, Arteaga CL, 2014, Molecular profiling of the residual disease of triplenegative breast cancers after neoadjuvant chemotherapy identifies actionable therapeutic targets. Cancer Discov 4(2):232–245. , DOI 10.1158/2159-8290.CD-13-0286 Marotta LLC, Almendro V, Marusyk A, Shipitsin M, Schemme J, Walker SR, Bloushtain-Qimron N, Kim JJ, Choudhury SA, Maruyama R, Wu Z, Gonen M, Mulvey LA, Bessarabova MO, Huh SJ, Silver SJ, Kim SY, Park SY, Lee HE, Anderson KS, Richardson AL, Nikolskaya T, Nikolsky Y, Liu XS, Root DE, HahnWC, Frank DA, Polyak K, 2011, The JAK2/STAT3 signaling pathway is required for growth of CD44(+)CD24(-, stem celllike breast cancer cells in human tumors. J Clin Investig 121(7): 2723–2735. , DOI 10.1172/JCI44745 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144(5):646–674. , DOI 10.1016/j.cell. 2011.02.013 Pardoll DM, 2012, The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12(4):252–264. https://doi. org/10.1038/nrc3239 Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, Hassel JC, Rutkowski P, McNeil C, Kalinka-Warzocha E, Savage KJ, Hernberg MM, Lebbe C, Charles J, Mihalcioiu C, Chiarion- SileniV, Mauch C, Cognetti F, AranceA, Schmidt H, Schadendorf D, Gogas H, Lundgren-Eriksson L, Horak C, Sharkey B,Waxman IM, Atkinson V, Ascierto PA, 2015, Nivolumab in Previously Untreated Melanoma without BRAF Mutation. N Engl J Med 372(4):320–330. , DOI 10.1056/Nejmoa1412082 Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, Dawson N, O'Donnell PH, Balmanoukian A, Loriot Y, Srinivas S, RetzMM, Grivas P, Joseph RW, GalskyMD, FlemingMT, Petrylak DP, Perez-Gracia JL, Burris HA, Castellano D, Canil C, Bellmunt J, Bajorin D, Nickles D, Bourgon R, Frampton GM, Cui N, Mariathasan S, Abidoye O, Fine GD, Dreicer R, 2016, Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 387(10031):1909–1920. https:// doi.org/10.1016/S0140-6736(16)00561-4 Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ, 2010, Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363(8):711–723. , DOI 10.1056/ NEJMoa1003466 Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, Harrington K, Kasper S, Vokes EE, Even C, Worden F, Saba NF, Iglesias Docampo LC, Haddad R, Rordorf T, Kiyota N, Tahara M, Monga M, Lynch M, Geese WJ, Kopit J, Shaw JW, Gillison ML, 2016, Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med 375(19):1856– 1867. , DOI 10.1056/NEJMoa1602252 Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, Waterhouse D, Ready N, Gainor J, Aren Frontera O, Havel L, Steins M, Garassino MC, Aerts JG, Domine M, Paz-Ares L, Reck M, Baudelet C, Harbison CT, Lestini B, Spigel DR, 2015, Nivolumab versus Docetaxel in Advanced Squamous-Cell Non- Small-Cell Lung Cancer. N Engl J Med 373(2):123–135. https:// doi.org/10.1056/NEJMoa1504627 Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, Castellano D, Choueiri TK, Gurney H, Donskov F, Bono P, Wagstaff J, Gauler TC, Ueda T, Tomita Y, Schutz FA, Kollmannsberger C, Larkin J, Ravaud A, Simon JS, Xu LA, Waxman IM, Sharma P, CheckMate I, 2015, Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med 373(19):1803–1813. , DOI 10.1056/NEJMoa1510665 Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn L, Carcereny E, Ahn MJ, Felip E, Lee JS, HellmannMD, Hamid O, Goldman JW, Soria JC, Dolled-Filhart M, Rutledge RZ, Zhang J, Lunceford JK, Rangwala R, Lubiniecki GM, Roach C, Emancipator K, Gandhi L, Investigators K, 2015, Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 372(21):2018–2028. , DOI 10.1056/NEJMoa1501824 Brown SD, Warren RL, Gibb EA, Martin SD, Spinelli JJ, Nelson BH, Holt RA, 2014, Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res 24(5):743–750. , DOI 10.1101/gr.165985.113 Loi S, Sirtaine N, Piette F, Salgado R, Viale G, Van Eenoo F, Rouas G, Francis P, Crown JP, Hitre E, de Azambuja E, Quinaux E, Di Leo A, Michiels S, Piccart MJ, Sotiriou C, 2013, Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98. J Clin Oncol 31(7):860–867. , DOI 10.1200/JCO.2011. 41.0902 Wimberly H, Brown JR, Schalper K, Haack H, Silver MR, Nixon C, Bossuyt V, Pusztai L, Lannin DR, Rimm DL, 2015, PD-L1 Expression Correlates with Tumor-Infiltrating Lymphocytes and Response to Neoadjuvant Chemotherapy in Breast Cancer. Cancer Immunol Res 3(4):326–332. , DOI 10.1158/2326-6066. CIR-14-0133 Ali HR, Glont SE, Blows FM, Provenzano E, Dawson SJ, Liu B, Hiller L, Dunn J, Poole CJ, Bowden S, Earl HM, Pharoah PDP, Caldas C, 2015, PD-L1 protein expression in breast cancer is rare, enriched in basal-like tumours and associated with infiltrating lymphocytes. Ann Oncol 26(7):1488–1493. , DOI 10. 1093/annonc/mdv192 Sabatier R, Finetti P, Mamessier E, Adelaide J, Chaffanet M, Ali HR, Viens P, Caldas C, Birnbaum D, Bertucci F, 2015, Prognostic and predictive value of PDL1 expression in breast cancer. Oncotarget 6(7):5449–5464. 10.18632/oncotarget.3216 Vacchelli E, Aranda F, Eggermont A, Galon J, Sautes-Fridman C, Cremer I, Zitvogel L, Kroemer G, Galluzzi L, 2014, TrialWatch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology 3(1):e27878. , DOI 10.4161/onci. 27878 Ramakrishnan R, Gabrilovich DI, 2011, Mechanism of synergistic effect of chemotherapy and immunotherapy of cancer. Cancer Immunol, Immunother : CII 60(3):419–423. , DOI 10. 1007/s00262-010-0930-1 Drake CG, Lipson EJ, Brahmer JR, 2014, Breathing new life into immunotherapy: review of melanoma, lung and kidney cancer. Nat Rev Clin Oncol 11(1):24–37. , DOI 10.1038/ nrclinonc.2013.208 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 701 EP 716 DI 10.1007/s12253-017-0307-2 PG 16 ER PT J AU Chen, Y Mo, J Jia, X He, Y AF Chen, Ying Mo, Jun Jia, Xi He, Yang TI The B7 Family Member B7-H6: a New Bane of Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE B7-H6; NKp30; Tumor immunity ID B7-H6; NKp30; Tumor immunity AB B7-H6 is a ligand of NKp30, which is an activating receptor of natural killer (NK) cells. High expression of B7-H6 is found in certain types of tumor cells, such as lymphoma, leukemia and gastric carcinoma. The expression of B7-H6 can be induced by inflammatory stress in healthy cells. The expression of B7-H6 is significantly correlated with distant metastasis status and post-operative prognosis in cancer patients. The effectiveness of B7-H6modified antitumor immunotherapy strategies had been verified in tumor-bearingmice, which opened a newdoor to targeted therapy. In this review, we will focus on the recent development on the roles of B7-H6 in tumor immunity, as well as mechanisms involved in the regulation of B7-H6 expression. C1 [Chen, Ying] The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, 215006 Suzhou, China. [Mo, Jun] The First Affiliated Hospital of Soochow University, Department of Orthopedics, 215006 Suzhou, China. [Jia, Xi] The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, 215006 Suzhou, China. [He, Yang] The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, 215006 Suzhou, China. RP He, Y (reprint author), The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, 215006 Suzhou, China. EM heyang1963@163.com CR Pende D, Rivera P, Marcenaro S, Chang CC, Biassoni R, Conte R, Kubin M, Cosman D, Ferrone S, Moretta L, Moretta A, 2002, Major histocompatibility complex class I-related chain a and UL16-binding protein expression on tumor cell lines of different histotypes: analysis of tumor susceptibility to NKG2D-dependent natural killer cell cytotoxicity. Cancer Res 62(21):6178–6186 Pende D, Parolini S, Pessino A, Sivori S, Augugliaro R, Morelli L, Marcenaro E, Accame L, Malaspina A, Biassoni R, Bottino C, Moretta L, Moretta A, 1999, Identification and molecular characterization of NKp30, a novel triggering receptor involved in natural cytotoxicity mediated by human natural killer cells. J Exp Med 190(10):1505–1516 Castriconi R, Dondero A, Augugliaro R, Cantoni C, Carnemolla B, Sementa AR, Negri F, Conte R, Corrias MV, Moretta L,Moretta A, Bottino C, 2004, Identification of 4Ig-B7-H3 as a neuroblastomaassociated molecule that exerts a protective role from an NK cellmediated lysis. Proc Natl Acad Sci U S A 101(34):12640–12645. , DOI 10.1073/pnas.0405025101 Byrd A, Hoffmann SC, Jarahian M, Momburg F, Watzl C, 2007, Expression analysis of the ligands for the natural killer cell receptors NKp30 and NKp44. PLoS One 2(12):e1339. , DOI 10. 1371/journal.pone.0001339 Biron CA, Nguyen KB, Pien GC, Cousens LP, Salazar-Mather TP, 1999, Natural killer cells in antiviral defense: function and regulation by innate cytokines. Annu Rev Immunol 17:189–220. https:// doi.org/10.1146/annurev.immunol.17.1.189 Brandt CS, BaratinM, Yi EC, Kennedy J, Gao Z, Fox B, Haldeman B, Ostrander CD, Kaifu T, Chabannon C, Moretta A, West R, Xu W, Vivier E, Levin SD, 2009, The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans. J Exp Med 206(7):1495–1503. , DOI 10.1084/jem.20090681 Joyce MG, Tran P, Zhuravleva MA, Jaw J, Colonna M, Sun PD, 2011, Crystal structure of human natural cytotoxicity receptor NKp30 and identification of its ligand binding site. Proc Natl Acad Sci U S A 108(15):6223–6228. , DOI 10.1073/ pnas.1100622108 Pogge von Strandmann E, Simhadri VR, von Tresckow B, Sasse S, Reiners KS, Hansen HP, Rothe A, Boll B, Simhadri VL, Borchmann P, McKinnon PJ, Hallek M, Engert A, 2007, Human leukocyte antigen-B-associated transcript 3 is released from tumor cells and engages the NKp30 receptor on natural killer cells. Immunity 27(6):965–974. , DOI 10.1016/j.immuni.2007.10.010 Arnon TI, Achdout H, Levi O,Markel G, Saleh N, Katz G, Gazit R, Gonen-Gross T, Hanna J, Nahari E, Porgador A, Honigman A, Plachter B, Mevorach D, Wolf DG, Mandelboim O, 2005, Inhibition of the NKp30 activating receptor by pp65 of human cytomegalovirus. Nat Immunol 6(5):515–523. , DOI 10. 1038/ni1190 Simhadri VR, Reiners KS, Hansen HP, Topolar D, Simhadri VL, Nohroudi K, Kufer TA, Engert A, Pogge von Strandmann E, 2008, Dendritic cells release HLA-B-associated transcript-3 positive exosomes to regulate natural killer function. PLoS One 3(10): e3377. , DOI 10.1371/journal.pone.0003377 Mavoungou E, Held J, Mewono L, Kremsner PG, 2007, A Duffy binding-like domain is involved in the NKp30-mediated recognition of plasmodium falciparum-parasitized erythrocytes by natural killer cells. J Infect Dis 195(10):1521–1531. , DOI 10. 1086/515579 Hecht ML, Rosental B, Horlacher T, Hershkovitz O, De Paz JL, Noti C, Schauer S, Porgador A, Seeberger PH, 2009, Natural cytotoxicity receptors NKp30, NKp44 and NKp46 bind to different heparan sulfate/heparin sequences. J Proteome Res 8(2):712–720. , DOI 10.1021/pr800747c Li Y,Wang Q, Mariuzza RA, 2011, Structure of the human activating natural cytotoxicity receptor NKp30 bound to its tumor cell ligand B7-H6. J Exp Med 208(4):703–714. , DOI 10. 1084/jem.20102548 Schlecker E, Fiegler N, Arnold A, Altevogt P, Rose-John S, Moldenhauer G, Sucker A, Paschen A, von Strandmann EP, Textor S, Cerwenka A, 2014, Metalloprotease-mediated tumor cell shedding of B7-H6, the ligand of the natural killer cell-activating receptorNKp30. Cancer Res 74(13):3429–3440. , DOI 10. 1158/0008-5472.CAN-13-3017 SalimiM, Xue L, Jolin H, Hardman C, Cousins DJ, McKenzie AN, Ogg GS, 2016, Group 2 innate lymphoid cells express functional NKp30 receptor inducing type 2 cytokine production. J Immunol 196(1):45–54. , DOI 10.4049/jimmunol.1501102 Matta J, Baratin M, Chiche L, Forel JM, Cognet C, Thomas G, Farnarier C, Piperoglou C, Papazian L, Chaussabel D, Ugolini S, Vely F, Vivier E, 2013, Induction of B7-H6, a ligand for the natural killer cell-activating receptor NKp30, in inflammatory conditions. Blood 122(3):394–404. , DOI 10.1182/blood-2013-01- 481705 Chen XJ, Shen J, Zhang GB, Chen WC, 2014, B7-H6 protein expression has no prognostic significance in human gastric carcinoma. Pathology oncology research : POR 20(1):203–207. https:// doi.org/10.1007/s12253-013-9686-1 Zhang X, Zhang G, Qin Y, Bai R, Huang J, 2014, B7-H6 expression in non-small cell lung cancers. Int J Clin Exp Pathol 7(10): 6936–6942 Guo JG, Guo CC, He ZQ, Liu ZG,Wang Y, Mou YG, 2016, Clinical significance of B7-H6 protein expression in astrocytoma. OncoTargets and therapy 9:3291–3297. , DOI 10.2147/OTT.S103771 Pesce S, Tabellini G, Cantoni C, Patrizi O, Coltrini D, Rampinelli F, Matta J, Vivier E, Moretta A, Parolini S, Marcenaro E, 2015, B7- H6-mediated downregulation of NKp30 in NK cells contributes to ovarian carcinoma immune escape. Oncoimmunology 4(4): e1001224. , DOI 10.1080/2162402X.2014.1001224 Zhou Y, Xu Y, Chen L, Xu B,Wu C, Jiang J, 2015, B7-H6 expression correlates with cancer progression and patient's survival in human ovarian cancer. Int J Clin Exp Pathol 8(8):9428–9433 Xu Z, Shen J,WangMH, Yi T, Yu Y, Zhu Y, Chen B, Chen J, Li L, Li M, Zuo J, Jiang H, Zhou D, Luan J, Xiao Z, 2016, Comprehensive molecular profiling of the B7 family of immuneregulatory ligands in breast cancer. Oncoimmunology 5(8): e1207841. , DOI 10.1080/2162402X.2016.1207841 Wu F, Wang J, Ke X, 2016, Knockdown of B7-H6 inhibits tumor progression and enhances chemosensitivity in B-cell non-Hodgkin lymphoma. Int J Oncol 48(4):1561–1570. , DOI 10.3892/ ijo.2016.3393 Gallinari P, Di Marco S, Jones P, Pallaoro M, Steinkuhler C, 2007, HDACs, histone deacetylation and gene transcription: from molecular biology to cancer therapeutics. Cell Res 17(3):195–211. https:// doi.org/10.1038/sj.cr.7310149 Minucci S, Pelicci PG, 2006, Histone deacetylase inhibitors and the promise of epigenetic, and more, treatments for cancer. Nat Rev Cancer 6(1):38–51. , DOI 10.1038/nrc1779 Witt O, Deubzer HE,Milde T, Oehme I, 2009, HDAC family: what are the cancer relevant targets? Cancer Lett 277(1):8–21. https://doi. org/10.1016/j.canlet.2008.08.016 Strahl BD, Allis CD, 2000, The language of covalent histone modifications. Nature 403(6765):41–45. , DOI 10.1038/47412 Fiegler N, Textor S, Arnold A, Rolle A, Oehme I, Breuhahn K, Moldenhauer G, Witzens-Harig M, Cerwenka A, 2013, Downregulation of the activating NKp30 ligand B7-H6 by HDAC inhibitors impairs tumor cell recognition by NK cells. Blood 122(5):684–693. , DOI 10.1182/blood-2013-02- 482513 Wu E, Croucher PI,McKie N, 1997, Expression of members of the novel membrane linked metalloproteinase family ADAM in cells derived from a range of haematological malignancies. Biochem Biophys Res Commun 235(2):437–442. , DOI 10.1006/ bbrc.1997.6714 Ohta S, Harigai M, Tanaka M, Kawaguchi Y, Sugiura T, Takagi K, Fukasawa C, Hara M, Kamatani N, 2001, Tumor necrosis factoralpha, TNF-alpha, converting enzyme contributes to production of TNF-alpha in synovial tissues from patients with rheumatoid arthritis. J Rheumatol 28(8):1756–1763 Horiuchi K, Kimura T, Miyamoto T, Takaishi H, Okada Y, Toyama Y, Blobel CP, 2007, Cutting edge: TNF-alpha-converting enzyme, TACE/ADAM17, inactivation in mouse myeloid cells prevents lethality from endotoxin shock. J Immunol 179(5):2686–2689 Ding X, Yang LY, Huang GW, Wang W, WQ L, 2004, ADAM17 mRNA expression and pathological features of hepatocellular carcinoma. World J Gastroenterol 10(18):2735–2739 Cesaro A, Abakar-Mahamat A, Brest P, Lassalle S, Selva E, Filippi J, Hebuterne X, Hugot JP, Doglio A, Galland F, Naquet P, Vouret- Craviari V, Mograbi B, Hofman PM, 2009, Differential expression and regulation of ADAM17 and TIMP3 in acute inflamed intestinal epithelia. Am J Physiol Gastrointest Liver Physiol 296(6):G1332– G1343. , DOI 10.1152/ajpgi.90641.2008 Lendeckel U, Kohl J, Arndt M, Carl-McGrath S, Donat H, Rocken C, 2005, Increased expression of ADAM family members in human breast cancer and breast cancer cell lines. J Cancer Res Clin Oncol 131(1):41–48. , DOI 10.1007/s00432-004-0619-y McGowan PM, McKiernan E, Bolster F, Ryan BM, Hill AD, McDermott EW, Evoy D, O'Higgins N, Crown J, Duffy MJ, 2008, ADAM-17 predicts adverse outcome in patients with breast cancer. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO 19(6):1075–1081. https:// doi.org/10.1093/annonc/mdm609 Lee SB, Schramme A, Doberstein K, Dummer R, Abdel-Bakky MS, Keller S, Altevogt P, ST O, Reichrath J, Oxmann D, Pfeilschifter J, Mihic-Probst D, Gutwein P, 2010, ADAM10 is upregulated in melanoma metastasis compared with primary melanoma. Journal Invest Dermatol 130(3):763–773. , DOI 10. 1038/jid.2009.335 Saftig P, Reiss K, 2011, The "a Disintegrin and Metalloproteases" ADAM10 and ADAM17: novel drug targets with therapeutic potential? Eur J Cell Biol 90(6–7):527–535. , DOI 10.1016/j. ejcb.2010.11.005 Semeraro M, Rusakiewicz S, Zitvogel L, Kroemer G, 2015, Natural killer cell mediated immunosurveillance of pediatric neuroblastoma. Oncoimmunology 4(11):e1042202. https://doi. org/10.1080/2162402X.2015.1042202 Textor S, Bossler F, Henrich KO, Gartlgruber M, Pollmann J, Fiegler N, Arnold A, Westermann F, Waldburger N, Breuhahn K, Golfier S, Witzens-Harig M, Cerwenka A, 2016, The protooncogene Myc drives expression of the NK cell-activating NKp30 ligand B7-H6 in tumor cells. Oncoimmunology 5(7): e1116674. , DOI 10.1080/2162402X.2015.1116674 Semeraro M, Rusakiewicz S, Minard-Colin V, Delahaye NF, Enot D, Vely F, Marabelle A, Papoular B, Piperoglou C, Ponzoni M, Perri P, Tchirkov A, Matta J, Lapierre V, Shekarian T, Valsesia- Wittmann S, Commo F, Prada N, Poirier-Colame V, Bressac B, Cotteret S, Brugieres L, Farace F, Chaput N, Kroemer G, Valteau- Couanet D, Zitvogel L, 2015, Clinical impact of the NKp30/B7-H6 axis in high-risk neuroblastoma patients. Sci Transl Med 7(283): 283ra255. , DOI 10.1126/scitranslmed.aaa2327 Kellner C, Maurer T, Hallack D, Repp R, van deWinkel JG, Parren PW, Valerius T, Humpe A, Gramatzki M, Peipp M, 2012, Mimicking an induced self phenotype by coating lymphomas with the NKp30 ligand B7-H6 promotes NK cell cytotoxicity. J Immunol 189(10):5037–5046. , DOI 10.4049/jimmunol. 1201321 Zhang T, MR W, Sentman CL, 2012, An NKp30-based chimeric antigen receptor promotes T cell effector functions and antitumor efficacy in vivo. J Immunol 189(5):2290–2299. , DOI 10. 4049/jimmunol.1103495 MR W, Zhang T, Gacerez AT, Coupet TA, DeMars LR, Sentman CL, 2015, B7H6-specific Bispecific T cell engagers lead to tumor elimination and host antitumor immunity. J Immunol 194(11): 5305–5311. , DOI 10.4049/jimmunol.1402517 Xu X, Narni-Mancinelli E, Cantoni C, Li Y, Guia S, Gauthier L, Chen Q, Moretta A, Vely F, Eisenstein E, Rangarajan S, Vivier E, Mariuzza RA, 2016, Structural insights into the inhibitory mechanism of an antibody against B7-H6, a stress-induced cellular ligand for the natural killer cell receptor NKp30. J Mol Biol 428(22): 4457–4466. , DOI 10.1016/j.jmb.2016.09.011 Ferlazzo G, TsangML, Moretta L, Melioli G, Steinman RM, Munz C, 2002, Human dendritic cells activate resting natural killer, NK, cells and are recognized via the NKp30 receptor by activated NK cells. J Exp Med 195(3):343–351 Vitale M, Della Chiesa M, Carlomagno S, Pende D, Arico M, Moretta L, Moretta A, 2005, NK-dependent DC maturation is mediated by TNFalpha and IFNgamma released upon engagement of the NKp30 triggering receptor. Blood 106(2):566–571. https://doi. org/10.1182/blood-2004-10-4035 Delahaye NF, Rusakiewicz S, Martins I, Menard C, Roux S, Lyonnet L, Paul P, Sarabi M, Chaput N, Semeraro M, Minard- Colin V, Poirier-Colame V, Chaba K, Flament C, Baud V, Authier H, Kerdine-Romer S, Pallardy M, Cremer I, Peaudecerf L, Rocha B, Valteau-Couanet D, Gutierrez JC, Nunes JA, Commo F, Bonvalot S, Ibrahim N, Terrier P, Opolon P, Bottino C, Moretta A, Tavernier J, Rihet P, Coindre JM, Blay JY, Isambert N, Emile JF, Vivier E, Lecesne A, Kroemer G, Zitvogel L, 2011, Alternatively spliced NKp30 isoforms affect the prognosis of gastrointestinal stromal tumors. Nat Med 17(6):700–707. https://doi. org/10.1038/nm.2366 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 717 EP 721 DI 10.1007/s12253-017-0357-5 PG 5 ER PT J AU Ghafoor, Q Baijal, Sh Taniere, P O’Sullivan, B Evans, M Middleton, G AF Ghafoor, Qamar Baijal, Shobhit Taniere, Phillipe O’Sullivan, Brendan Evans, Matthew Middleton, Gary TI Epidermal Growth Factor Receptor (EGFR) Kinase Inhibitors and Non-Small Cell Lung Cancer (NSCLC) – Advances in Molecular Diagnostic Techniques to Facilitate Targeted Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Non-small cell lung cancer; NSCLC; Epidermal growth factor receptor tyrosine kinase inhibitors; EGFR TKIs; Molecular diagnostic techniques ID Non-small cell lung cancer; NSCLC; Epidermal growth factor receptor tyrosine kinase inhibitors; EGFR TKIs; Molecular diagnostic techniques AB A subset of patients with non-small cell lung cancer (NSCLC) respond well to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs), due to the presence of sensitising mutations in the gene encoding EGFR. Mutations associated with resistance to first generation EGFR-TKIs have also been identified, which lead to therapeutic failure and the requirement for new drugs. Three generations of EGFR-TKIs have been developed and either have been, or are being, evaluated as first and/or second line therapeutic agents. In this review, we consider the advances in molecular diagnostic techniques that are used, or are in development, to facilitate the targeted EGFR TKI therapy of patients with NSCLC. A literature search was conducted in May 2017 using PubMed, and spanning the period September 2005 (EU approval date of erlotinib) to May 2017. Search terms used were: EGFR TKI, NSCLC, clinical trial, erlotinib, gefitinib, afatinib, EGFR mutations, Exon 19 deletion, and Leu858Arg. The use of molecular data, in conjunction with other clinical and diagnostic information, will assist physicians to make the best therapeutic choice for each patient with advanced NSCLC. Personalized medicine and a rapidly developing therapy landscape will enable these patients to achieve optimal responses to EGFR TKIs. C1 [Ghafoor, Qamar] University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Department of Oncology, B15 2TH Birmingham, UK. [Baijal, Shobhit] University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Department of Oncology, B9 5SS Birmingham, UK. [Taniere, Phillipe] University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Department of Cellular Pathology, B15 2TH Birmingham, UK. [O’Sullivan, Brendan] University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Department of Cellular Pathology, B15 2TH Birmingham, UK. [Evans, Matthew] University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Department of Cellular Pathology, B15 2TH Birmingham, UK. [Middleton, Gary] University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Department of Oncology, B15 2TH Birmingham, UK. RP Ghafoor, Q (reprint author), University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Department of Oncology, B15 2TH Birmingham, UK. EM Qamar.Ghafoor@uhb.nhs.uk CR Dearden S, Stevens J, Wu YL, Blowers D, 2013, Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology, mutMap). Ann Oncol: official journal of the European Society for Medical Oncology / ESMO 24(9):2371– 2376. , DOI 10.1093/annonc/mdt205 Marchetti A, Martella C, Felicioni L, Barassi F, Salvatore S, Chella A, Camplese PP, Iarussi T, Mucilli F, Mezzetti A, Cuccurullo F, Sacco R, Buttitta F, 2005, EGFR mutations in non-small-cell lung cancer: analysis of a large series of cases and development of a rapid and sensitive method for diagnostic screening with potential implications on pharmacologic treatment. J Clin Oncol: official journal of the American Society of Clinical Oncology 23(4):857– 865. , DOI 10.1200/JCO.2005.08.043 Kuan FC, Kuo LT, Chen MC, Yang CT, Shi CS, Teng D, Lee KD, 2015, Overall survival benefits of first-line EGFR tyrosine kinase inhibitors in EGFR-mutated non-small-cell lung cancers: a systematic review and meta-analysis. Br J Cancer 113(10):1519–1528. , DOI 10.1038/bjc.2015.356 Cancer Genome Atlas Research N, 2012, Comprehensive genomic characterization of squamous cell lung cancers. Nature 489(7417): 519–525. , DOI 10.1038/nature11404 Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, Majem M, Lopez-Vivanco G, Isla D, Provencio M, Insa A, Massuti B, Gonzalez-Larriba JL, Paz-Ares L, Bover I, Garcia- Campelo R, Moreno MA, Catot S, Rolfo C, Reguart N, Palmero R, Sanchez JM, Bastus R, Mayo C, Bertran-Alamillo J, Molina MA, Sanchez JJ, Taron M, Spanish Lung Cancer G, 2009, Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med 361(10):958–967. , DOI 10.1056/ NEJMoa0904554 Midha A, Dearden S, McCormack R, 2015, EGFR mutation incidence in non-small-cell lung cancer of adenocarcinoma histology: a systematic review and global map by ethnicity, mutMapII). Am J Cancer Res 5(9):2892–2911 European Medicines Agency, 2005, 2014 version accessed), Summary of Product Characteristics - Tarceva, erlotinib). http://www.ema.europa.eu/ European Medicines Agency, 2009, 2014 version accessed), Summary of Product Characteristics - Iressa, gefitinib). http://www.ema.europa.eu/ European Medicines Agency, 2014, Summary of Product Characteristics - Giotrif, afatinib). http://www.ema.europa.eu/ Juan O, Yousaf N, Popat S, 2017, First-line Epidermal Growth Factor Receptor, EGFR, Kinase Inhibitors for EGFR Mutant Non-small Cell Lung Cancer: And the Winner is….. Clin Oncol, R Coll Radiol, 29(1):e1–e4. , DOI 10.1016/j.clon.2016. 09.007 Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, Sunpaweravong P, Han B, Margono B, Ichinose Y, Nishiwaki Y, Ohe Y, Yang JJ, Chewaskulyong B, Jiang H, Duffield EL, Watkins CL, Armour AA, Fukuoka M, 2009, Gefitinib or carboplatinpaclitaxel in pulmonary adenocarcinoma. N Engl J Med 361(10): 947–957. , DOI 10.1056/NEJMoa0810699 Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, Zhang S, Wang J, Zhou S, Ren S, Lu S, Zhang L, Hu C, Hu C, Luo Y, Chen L, Ye M, Huang J, Zhi X, Zhang Y, Xiu Q, Ma J, Zhang L, You C, 2011, Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer, OPTIMAL, CTONG-0802): a multicentre, openlabel, randomised, phase 3 study. Lancet Oncol 12(8):735–742. , DOI 10.1016/S1470-2045(11)70184-X Yang JC, Hirsh V, Schuler M, Yamamoto N, O'Byrne KJ, Mok TS, Zazulina V, Shahidi M, Lungershausen J, Massey D, Palmer M, Sequist LV, 2013, Symptom control and quality of life in LUX-lung 3: a phase III study of afatinib or cisplatin/ pemetrexed in patients with advanced lung adenocarcinoma with EGFR mutations. J Clin Oncol Off J Am Soc Clin Oncol 31(27):3342–3350. , DOI 10.1200/JCO.2012.46.1764 Passaro A, Gori B, de Marinis F, 2013, Afatinib as first-line treatment for patients with advanced non-small-cell lung cancer harboring EGFR mutations: focus on LUX-lung 3 and LUX-lung 6 phase III trials. J Thorac Dis 5(4):383–384. , DOI 10.3978/j.issn. 2072-1439.2013.07.32 Sequist LV, Yang JC, Yamamoto N, O'Byrne K, Hirsh V, Mok T, Geater SL, Orlov S, Tsai CM, Boyer M, SuWC, Bennouna J, Kato T, Gorbunova V, Lee KH, Shah R, Massey D, Zazulina V, Shahidi M, Schuler M, 2013, Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol Off J Am Soc Clin Oncol 31(27): 3327–3334. , DOI 10.1200/JCO.2012.44.2806 Yang J-H, Sequist LV, Schuler M, Mok T, Yamamoto N, O'Byrne K, Hirsh V, Geater SL, Zhou C, Massey D, Zazulina V, Wu Y-L, investigators on behalf oL-LaL-L Overall survival, OS, in patients, pts, with advanced non-small cell lung cancer, NSCLC, harboring common, Del19/L858R, Epidermal Growth Factor Receptor mutations, EGFRmut): pooled analysis of two large open-label phase III studies, LUX-Lung 3 [LL3] and LUX-Lung 6 [LL6], comparing afatinib with chemotherapy, CT).. In: 50th American Society of Clinical Oncology, ASCO, Annual Meeting, Chicago, Il, USA, 30 May–3 June 2014. p Abstract 21366 Geater SL, Xu CR, Zhou C, Hu CP, Feng J, Lu S, Huang Y, Li W, Hou M, Shi JH, Lee KY, Palmer M, Shi Y, Lungershausen J, Wu YL, 2015, Symptom and quality of life improvement in LUX-lung 6: an open-label phase III study of Afatinib versus cisplatin/ gemcitabine in Asian patients with EGFR mutation-positive advanced non-small-cell lung cancer. J Thorac Oncol: official publication of the InternationalAssociation for the Study of Lung Cancer 10(6):883–889. , DOI 10.1097/JTO.0000000000000517 Park K, Tan EH, O'ByrneK, Zhang L, Boyer M,Mok T, HirshV, Yang JC, Lee KH, Lu S, Shi Y, Kim SW, Laskin J, Kim DW, Arvis CD, Kolbeck K, Laurie SA, Tsai CM, Shahidi M, Kim M, Massey D, Zazulina V, Paz-Ares L, 2016, Afatinib versus gefitinib as first-line treatment of patients with EGFR mutationpositive non-small-cell lung cancer, LUX-lung 7): a phase 2B, open-label, randomised controlled trial. Lancet Oncol 17(5): 577–589. , DOI 10.1016/S1470-2045(16)30033-X Paz-Ares L, Tan EH, O'Byrne K, Zhang L, Hirsh V, Boyer M, Yang JC, Mok T, Lee KH, Lu S, Shi Y, Lee DH, Laskin J, Kim DW, Laurie SA, Kolbeck K, Fan J, Dodd N, Marten A, Park K, 2017, Afatinib versus gefitinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: overall survival data from the phase IIb LUX-lung 7 trial. Ann Oncol: official journal of the European Society for Medical Oncology / ESMO 28(2):270–277. , DOI 10.1093/annonc/mdw611 Zugazagoitia J, Diaz A, Jimenez E, Nunez JA, Iglesias L, Ponce- Aix S, Paz-Ares L, 2017, Second-line treatment of non-small cell lung cancer: focus on the clinical development of Dacomitinib. Front Med, Lausanne, 4:36. , DOI 10.3389/fmed.2017. 00036 Ramalingam SS, O'Byrne K, Boyer M, Mok T, Janne PA, Zhang H, Liang J, Taylor I, Sbar EI, Paz-Ares L, 2016, Dacomitinib versus erlotinib in patients with EGFR-mutated advanced nonsmall-cell lung cancer, NSCLC): pooled subset analyses from two randomized trials. Ann Oncol: official journal of the European Society for Medical Oncology / ESMO 27(3):423–429. , DOI 10.1093/annonc/mdv593 Ou SH, Soo RA, 2015, Dacomitinib in lung cancer: a "lost generation" EGFR tyrosine-kinase inhibitor from a bygone era? Drug Des Devel Ther 9:5641–5653. , DOI 10. 2147/DDDT.S52787 Mok T, Cheng Y, Zhou X, Lee K, Nakagawa K, Niho S, Tsuji R, Rosell R, Jaime J, Migliorino MR, Pluzanski A, Linke R, Sbar E, Wang T, Wu Y-L Dacomitinib versus gefitinib for the first-line treatment of advanced EGFR mutation positive non-small cell lung cancer, ARCHER 1050): A randomized, open-label phase III trial.. In: American Society of Clinical Oncology, ASCO, Annual Meeting, Chicago, USA, 2–6 June 2017 2017 Juan O, Popat S, 2017, Treatment choice in epidermal growth factor receptor mutation-positive non-small cell lung carcinoma: latest evidence and clinical implications. Ther AdvMed Oncol 9(3):201– 216. , DOI 10.1177/1758834016687262 Remon J, Menis J, Hasan B, Peric A, De Maio E, Novello S, Reck M, Berghmans T, Wasag B, Besse B, Dziadziuszko R, 2017, The APPLE trial: feasibility and activity of AZD9291, Osimertinib, treatment on positive PLasma T790M in EGFRmutant NSCLC patients. EORTC 1613. Clin Lung Cancer. , DOI 10.1016/j.cllc.2017.02.005 Yang JC, Ahn MJ, Kim DW, Ramalingam SS, Sequist LV, Su WC, Kim SW, Kim JH, Planchard D, Felip E, Blackhall F, Haggstrom D, Yoh K, Novello S, Gold K, Hirashima T, Lin CC, Mann H, Cantarini M, Ghiorghiu S, Janne PA, 2017, Osimertinib in pretreated T790M-positive advanced nonsmall- cell lung cancer: AURA study phase II extension component. J Clin Oncol Off J Am Soc Clin Oncol 35(12):1288–1296. , DOI 10.1200/JCO.2016.70.3223 European Medicines Agency, 2017, Tagrisso, osimertinib, Summary of Product Characteristics.. First published 02/2016. Updated 03/2017 edn., Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, Shepherd FA, He Y, Akamatsu H, Theelen WS, Lee CK, Sebastian M, Templeton A, Mann H, Marotti M, Ghiorghiu S, Papadimitrakopoulou VA, Investigators A, 2017, Osimertinib or platinum-Pemetrexed in EGFR T790M-positive lung cancer. N Engl J Med 376(7):629–640. , DOI 10.1056/NEJMoa1612674 Yang J, 2016, Osimertinib, AZD9291, in pre-treated pts with T790M-positive advanced NSCLC: updated phase 1, P1, and pooled phase 2, P2, results.. Paper presented at the European lung cancer conference, ELCC, 2016, Geneva. Switzerland, 14 April 2016 Ramalingam S, 2016, Osimertinib as first-line treatment for EGFR mutation-positive advanced NSCLC: updated efficacy and safety results from two phase I expansion cohorts.. Paper presented at the European lung cancer conference, ELCC, 2016, Geneva. Switzerland, 14 April 2016 Ferguson KM, 2008, Structure-based view of epidermal growth factor receptor regulation. Annu Rev Biophys 37:353–373. , DOI 10.1146/annurev.biophys.37.032807.125829 Sharma SV, Bell DW, Settleman J, Haber DA, 2007, Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer 7(3):169–181. , DOI 10.1038/nrc2088 Thress KS, Paweletz CP, Felip E, Cho BC, StetsonD, Dougherty B, Lai Z, Markovets A, Vivancos A, Kuang Y, Ercan D, Matthews SE, Cantarini M, Barrett JC, Janne PA, Oxnard GR, 2015, Acquired EGFR C797S mutation mediates resistance to AZD9291 in nonsmall cell lung cancer harboring EGFR T790M. Nat Med 21(6): 560–562. , DOI 10.1038/nm.3854 Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA(2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350(21):2129–2139. , DOI 10.1056/NEJMoa040938 Evans M, O’Sullivan B, Hughes F, Swift C, Mullis T, Dessi R et al, 2016, The sectrum of rare and complicated EGFR mutations in NSCLC: a series of 17, 047 tumours. NCRI Cancer Conferecne, Liverpool Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, Kris MG, Miller VA, Ladanyi M, Riely GJ, 2013, Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res 19(8):2240–2247. , DOI 10.1158/1078-0432. CCR-12-2246 Yasuda H, Park E, Yun CH, Sng NJ, Lucena-Araujo AR, Yeo WL, HubermanMS, CohenDW, Nakayama S, Ishioka K,Yamaguchi N, Hanna M, Oxnard GR, Lathan CS, Moran T, Sequist LV, Chaft JE, Riely GJ, Arcila ME, Soo RA, Meyerson M, Eck MJ, Kobayashi SS, Costa DB, 2013, Structural, biochemical, and clinical characterization of epidermal growth factor receptor, EGFR, exon 20 insertion mutations in lung cancer. Sci Transl Med 5(216): 216ra177. , DOI 10.1126/scitranslmed.3007205 Haber DA, Bell DW, Sordella R, Kwak EL, Godin-Heymann N, Sharma SV, Lynch TJ, Settleman J, 2005, Molecular targeted therapy of lung cancer: EGFR mutations and response to EGFR inhibitors. Cold Spring Harb Symp Quant Biol 70:419–426. , DOI 10.1101/sqb.2005.70.043 Girard N, Sima CS, Jackman DM, Sequist LV, Chen H, Yang JC, Ji H, Waltman B, Rosell R, Taron M, Zakowski MF, Ladanyi M, Riely G, Pao W, 2012, Nomogram to predict the presence of EGFR activating mutation in lung adenocarcinoma. Eur Respir J 39(2):366–372. , DOI 10.1183/09031936.00010111 Wang J, Wang B, Chu H, Yao Y, 2016, Intrinsic resistance to EGFR tyrosine kinase inhibitors in advanced non-small-cell lung cancer with activating EGFRmutations. OncoTargets Ther 9:3711– 3726. , DOI 10.2147/OTT.S106399 Kim EK, Kim KA, Lee CY, Shim HS, 2017, The frequency and clinical impact of HER2 alterations in lung adenocarcinoma. PLoS One 12(2):e0171280. , DOI 10.1371/journal.pone. 0171280 Presutti D, Santini S, Cardinali B, Papoff G, Lalli C, Samperna S, Fustaino V, Giannini G, Ruberti G, 2015, MET gene amplification and MET receptor activation are not sufficient to predict efficacy of combined MET and EGFR inhibitors in EGFR TKI-resistant NSCLC cells. PLoS One 10(11):e0143333. , DOI 10.1371/journal.pone.0143333 Schrock AB, Frampton GM, Suh J, Chalmers ZR, Rosenzweig M, Erlich RL, Halmos B, Goldman J, Forde P, Leuenberger K, Peled N, Kalemkerian GP, Ross JS, Stephens PJ, Miller VA, Ali SM, Ou SH, 2016, Characterization of 298 patients with lung cancer harboring MET exon 14 skipping alterations. J Thorac Oncol: official publication of the International Association for the Study of Lung Cancer 11(9):1493–1502. , DOI 10.1016/j.jtho.2016.06.004 Vendrell JA, Mau-Them FT, Beganton B, Godreuil S, Coopman P, Solassol J, 2017, Circulating cell free tumor DNA detection as a routine tool for lung cancer patient management. Int J Mol Sci 18(2). , DOI 10.3390/ijms18020264 Normanno N, Denis MG, Thress KS, Ratcliffe M, Reck M, 2017, Guide to detecting epidermal growth factor receptor, EGFR, mutations in ctDNA of patients with advanced nonsmall- cell lung cancer. Oncotarget 8(7):12501–12516. , DOI 10.18632/oncotarget.13915 Ellison G, Zhu G, Moulis A, Dearden S, Speake G, McCormack R, 2013, EGFR mutation testing in lung cancer: a review of available methods and their use for analysis of tumour tissue and cytology samples. J Clin Pathol 66(2): 79–89. , DOI 10.1136/jclinpath-2012-201194 Overman MJ, Modak J, Kopetz S, Murthy R, Yao JC, Hicks ME, Abbruzzese JL, Tam AL, 2013, Use of research biopsies in clinical trials: are risks and benefits adequately discussed? J Clin Oncol Off J Am Soc Clin Oncol 31(1):17– 22. , DOI 10.1200/JCO.2012.43.1718 Thress KS, Brant R, Carr TH, Dearden S, Jenkins S, Brown H, Hammett T, Cantarini M, Barrett JC, 2015, EGFR mutation detection in ctDNA from NSCLC patient plasma: a crossplatform comparison of leading technologies to support the clinical development of AZD9291. Lung Cancer 90(3):509–515. , DOI 10.1016/j.lungcan.2015.10.004 Yang JC, Wu YL, Chan V, Kurnianda J, Nakagawa K, Saijo N, Fukuoka M, McWalter G, McCormack R, Mok TS, 2014, Epidermal growth factor receptor mutation analysis in previously unanalyzed histology samples and cytology samples fromthe phase III Iressa Pan-ASia study, IPASS). Lung Cancer 83(2):174–181. , DOI 10.1016/j.lungcan.2013.11.021 Chuang JC, Salahudeen AA, Wakelee HA, 2016, Rociletinib, a third generation EGFR tyrosine kinase inhibitor: current data and future directions. Expert Opin Pharmacother 17(7):989–993. , DOI 10.1517/14656566.2016.1162786 Barlesi F, Mazieres J, Merlio JP, Debieuvre D, Mosser J, Lena H, Ouafik L, Besse B, Rouquette I, Westeel V, Escande F, Monnet I, Lemoine A, Veillon R, Blons H, Audigier-Valette C, Bringuier PP, Lamy R, Beau-Faller M, Pujol JL, Sabourin JC, Penault-Llorca F, Denis MG, Lantuejoul S, Morin F, Tran Q, Missy P, Langlais A, Milleron B, Cadranel J, Soria JC, Zalcman G, Biomarkers France c, 2016, Routine molecular profiling of patients with advanced non-small-cell lung cancer: results of a 1- year nationwide programme of the French cooperative thoracic intergroup, IFCT). Lancet 387(10026):1415–1426. , DOI 10.1016/S0140-6736(16)00004-0 Riely GJ, Pao W, Pham D, Li AR, Rizvi N, Venkatraman ES, Zakowski MF, Kris MG, Ladanyi M, Miller VA, 2006, Clinical course of patients with non-small cell lung cancer and epidermal growth factor receptor exon 19 and exon 21 mutations treated with gefitinib or erlotinib. Clin Cancer Res 12(3 Pt 1):839–844. , DOI 10.1158/1078-0432.CCR-05-1846 Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, Palmero R, Garcia-Gomez R, Pallares C, Sanchez JM, Porta R, Cobo M, Garrido P, Longo F, Moran T, Insa A, De Marinis F, Corre R, Bover I, Illiano A, Dansin E, de Castro J, Milella M, Reguart N, Altavilla G, Jimenez U, Provencio M, Moreno MA, Terrasa J, Munoz-Langa J, Valdivia J, Isla D, Domine M, Molinier O, Mazieres J, Baize N, Garcia-Campelo R, Robinet G, Rodriguez-Abreu D, Lopez-Vivanco G, Gebbia V, Ferrera-Delgado L, Bombaron P, Bernabe R, Bearz A, Artal A, Cortesi E, Rolfo C, Sanchez-Ronco M, Drozdowskyj A, Queralt C, de Aguirre I, Ramirez JL, Sanchez JJ, Molina MA, Taron M, Paz-Ares L, Spanish Lung Cancer Group in collaboration with Groupe Francais de P-C, Associazione Italiana Oncologia T, 2012, Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutationpositive non-small-cell lung cancer, EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 13(3):239– 246. , DOI 10.1016/S1470-2045(11)70393-X Sequist LV, Martins RG, Spigel D, Grunberg SM, Spira A, Janne PA, Joshi VA,McCollum D, Evans TL, Muzikansky A, Kuhlmann GL, Han M, Goldberg JS, Settleman J, Iafrate AJ, Engelman JA, Haber DA, Johnson BE, Lynch TJ, 2008, First-line gefitinib in patients with advanced non-small-cell lung cancer harboring somatic EGFR mutations. J Clin Oncol Off J Am Soc Clin Oncol 26(15): 2442–2449. , DOI 10.1200/JCO.2007.14.8494 Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, Seto T, Satouchi M, Tada H, Hirashima T, Asami K, Katakami N, Takada M, Yoshioka H, Shibata K, Kudoh S, Shimizu E, Saito H, Toyooka S, Nakagawa K, Fukuoka M, West Japan Oncology G, 2010, Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor, WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 11(2):121–128. , DOI 10.1016/ S1470-2045(09)70364-X Fukuoka M, Wu YL, Thongprasert S, Sunpaweravong P, Leong SS, Sriuranpong V, Chao TY, Nakagawa K, Chu DT, Saijo N, Duffield EL, Rukazenkov Y, Speake G, Jiang H, Armour AA, To KF, Yang JC, Mok TS, 2011, Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia, IPASS). J Clin Oncol: official journal of the American Society of Clinical Oncology 29(21):2866–2874. , DOI 10.1200/JCO.2010.33.4235 Yang JC, Wu YL, Schuler M, Sebastian M, Popat S, Yamamoto N, Zhou C, Hu CP, O'Byrne K, Feng J, Lu S, Huang Y, Geater SL, Lee KY, Tsai CM, Gorbunova V, Hirsh V, Bennouna J, Orlov S, MokT, BoyerM, SuWC, LeeKH, KatoT,MasseyD, ShahidiM, Zazulina V, Sequist LV, 2015, Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma, LUX-lung 3 and LUX-lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol 16(2): 141–151. , DOI 10.1016/S1470-2045(14)71173-8 Astrazeneca, 2017, Tagrisso significantly improves progressionfree survival in the Phase III FLAURA trial for lung cancer. https://www.astrazeneca.com/media-centre/press-releases/2017/ tagrisso-significantly-improves-progression-free-survival-in-thephase- iii-flaura-trial-for-lung-cancer-27072017.html. Accessed 27 July 2017 Yang JC, Sequist LV, Geater SL, Tsai CM, Mok TS, Schuler M, Yamamoto N, Yu CJ, Ou SH, Zhou C, Massey D, Zazulina V, Wu YL, 2015, Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: a combined post-hoc analysis of LUX-lung 2, LUX-lung 3, and LUX-lung 6. Lancet Oncol 16(7):830–838. , DOI 10.1016/S1470-2045(15)00026-1 Popat S, Hughes L, O’Brien MER, Ahmad T, Lewanski C, Dernedde U, Jankowski P, Mulatero C, Shah R, Hicks J, Geldart T, Cominos M, Gray G, Spicer J, Bell K, Ngai Y, Hackshaw A, 2016, Afatinib benefits patients with confirmed/ suspected EGFR mutant NSCLC, unsuitable for chemotherapy, TIMELYphase II trial). Paper presented at the WCLC, Vienna, Austria, December 4-7 2016 Park K, Tan E, Zhang L, Hirsh V, O’Byrne K, Boyer M, Yang J, Mok T, Peil B, Marten A, Paz-Ares L, 2016, Afatinib versus gefitinib as first-line treatment for EGFR mutation-positive NSCLC patients aged ≥75 years: subgroup analysis of lux-lung 7. Paper presented at the WCLC, Vienna, Austria, December 4-7 2016 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 723 EP 731 DI 10.1007/s12253-017-0377-1 PG 9 ER PT J AU Davidson, TK Zhu, Z Balabanov, D Zhao, L Wakefield, RM Bai, Q Fang, Y AF Davidson, T Kristoffer Zhu, Ziwen Balabanov, Dean Zhao, Lei Wakefield, R Mark Bai, Qian Fang, Yujiang TI Beyond Conventional Medicine - a Look at Blueberry, a Cancer-Fighting Superfruit SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Blueberry; Phytochemicals; Cancer; Apoptosis; Anthocyanidin ID Blueberry; Phytochemicals; Cancer; Apoptosis; Anthocyanidin AB Nearly 40% of men and women will be diagnosed with cancer during their lifetime. Thus, there is a rapidly growing need for novel therapies to combat this deadly disease. One such method is the consumption of blueberries. Long coveted for their powerful antioxidant properties, more recent studies have demonstrated that blueberries also exhibit inherent abilities to prevent carcinogenesis, inhibit the proliferation of neoplastic cells, and reduce the risks of recurrence in patients in remission. This review will focus on the specific activities of blueberry derivatives in cancer cells across many different forms of cancer. Ultimately, such research could be helpful in the development of new strategies to treat cancer. C1 [Davidson, T Kristoffer] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. [Zhu, Ziwen] University of Missouri, School of Medicine, Department of Surgery, 65212 Columbia, MO, USA. [Balabanov, Dean] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. [Zhao, Lei] The First Affiliated Hospital of Anhui Medical University, Department of Infectious DiseaseHefei, China. [Wakefield, R Mark] University of Missouri, School of Medicine, Department of Surgery, 65212 Columbia, MO, USA. [Bai, Qian] University of Missouri, School of Medicine, Department of Surgery, 65212 Columbia, MO, USA. [Fang, Yujiang] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. RP Fang, Y (reprint author), Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, USA. EM yujiang.fang@dmu.edu CR Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(1):7–30. , DOI 10.3322/caac.21387 Slattery ML, Lundgreen A, Torres-Mejia G, Wolff RK, Hines L, Baumgartner K, John EM, 2014, Diet and lifestyle factors modify immune/inflammation response genes to alter breast cancer risk and prognosis: the breast cancer health disparities study. Mutat Res Fundam Mol Mech Mutagen 770:19–28 Lee G-Y, Lee JJ, Lee S-M, 2015, Antioxidant and anticoagulant status were improved by personalized dietary intervention based on biochemical and clinical parameters in cancer patients. Nutr Cancer 67(7):1083–1092 Nandakumar V, Singh T, Katiyar SK, 2008, Multi-targeted prevention and therapy of cancer by proanthocyanidins. Cancer Lett 269(2):378–387 Kresty LA, Weh KM, Zeyzus-Johns B, Perez LN, Howell AB, 2015, Cranberry proanthocyanidins inhibit esophageal adenocarcinoma in vitro and in vivo through pleiotropic cell death induction and PI3K/AKT/mTOR inactivation. Oncotarget 6(32):33438 Jeyabalan J, Aqil F, Munagala R, Annamalai L, Vadhanam MV, Gupta RC, 2014, Chemopreventive and therapeutic activity of dietary blueberry against estrogen-mediated breast cancer. J Agric Food Chem 62(18):3963–3971 Chen C, Li Y, Xu Z, 2010, Chemical principles and bioactivities of blueberry. Yao xue xue bao=Acta Pharm Sin 45(4):422–429 Johnson SA, Arjmandi BH, 2013, Evidence for anti-cancer properties of blueberries: a mini-review. Anticancer Agents Med, Formerly CurrMed Chem: Anti-Cancer Agents, 13(8):1142–1148 Howlader N, Noone A, Krapcho M, Garshell J, Neyman N, Altekruse S, Kosary C, Yu M, Ruhl J, Tatalovich Z, 2013, SEER cancer statistics review, Natl Cancer Inst. Bethesda, 1975–2010) Adams LS, Phung S, Yee N, Seeram NP, Li L, Chen S, 2010, Blueberry phytochemicals inhibit growth and metastatic potential of MDA-MB-231 breast cancer cells through modulation of the phosphatidylinositol 3-kinase pathway. Cancer Res 70(9):3594– 3605 Montales MTE, Rahal OM, Kang J, Rogers T, Prior RL, Wu X, Simmen RC, 2012, Repression of mammosphere formation of human breast cancer cells by soy isoflavone genistein and blueberry polyphenolic acids suggests diet-mediated targeting of cancer stemlike/ progenitor cells. Carcinogenesis 33(3):652–660 Kanaya N, Adams L, Takasaki A, Chen S, 2014, Whole blueberry powder inhibits metastasis of triple negative breast cancer in a xenograft mouse model through modulation of inflammatory cytokines. Nutr Cancer 66(2):242–248 Ravoori S, Vadhanam MV, Aqil F, Gupta RC, 2012, Inhibition of estrogen-mediated mammary tumorigenesis by blueberry and black raspberry. J Agric Food Chem 60(22):5547–5555 Bruix J, ShermanM(2011)Management of hepatocellular carcinoma: an update. Hepatology 53(3):1020–1022 Yi W, Akoh CC, Fischer J, Krewer G, 2006, Effects of phenolic compounds in blueberries and muscadine grapes on HepG2 cell viability and apoptosis. Food Res Int 39(5):628–638 Bingul I, Basaran-Kucukgergin C, Tekkesin MS, Olgac V, Dogru- Abbasoglu S, Uysal M, 2013, Effect of blueberry pretreatment on diethylnitrosamine-induced oxidative stress and liver injury in rats. Environ Toxicol Pharmacol 36(2):529–538 Bingul I, Basaran-Kucukgergin C, Aydin AF, Soluk-Tekkesin M, Olgac V, Dogru-Abbasoglu S, UysalM(2015, Blueberry treatment attenuated cirrhotic and preneoplastic lesions and oxidative stress in the liver of diethylnitrosamine-treated rats. Int J Immunopathol Pharmacol 29(3):426–37 ZhanW, LiaoX, YuL, TianT, LiuX,Liu J, CaiL-J,XiaoX,XieR-J, Yang Q, 2016, Effects of blueberries on migration, invasion, proliferation, the cell cycle and apoptosis in hepatocellular carcinoma cells. Biomedical reports 5(5):579–584 Yi W, Fischer J, Krewer G, Akoh CC, 2005, Phenolic compounds from blueberries can inhibit colon cancer cell proliferation and induce apoptosis. J Agric Food Chem 53(18):7320–7329 Suh N, Paul S, Hao X, Simi B, Xiao H, Rimando AM, Reddy BS, 2007, Pterostilbene, an active constituent of blueberries, suppresses aberrant crypt foci formation in the azoxymethaneinduced colon carcinogenesis model in rats. Clin Cancer Res 13(1):350–355 Minker C, Duban L, Karas D, Jarvinen P, Lobstein A, Muller CD, 2015, Impact of procyanidins from different berries on caspase 8 activation in colon cancer. Oxidative Med Cell Longev 2015:1–13 Qi C, Li S, Jia Y, Wang L, 2014, [Blueberry anthocyanins induce G2/M cell cycle arrest and apoptosis of oral cancer KB cells through down-regulation methylation of p53]. Yi chuan= Hereditas/Zhongguo yi chuan xue hui bian ji 36, 6):566–573 Baba AB, Kowshik J, Krishnaraj J, Sophia J, Dixit M, Nagini S, 2016, Blueberry inhibits invasion and angiogenesis in 7, 12- dimethylbenz [a] anthracene, DMBA)-induced oral squamous cell carcinogenesis in hamsters via suppression of TGF-β and NF-κB signaling pathways. J Nutr Biochem 35:37–47 Schmidt BM, Erdman JW, Lila MA, 2006, Differential effects of blueberry proanthocyanidins on androgen sensitive and insensitive human prostate cancer cell lines. Cancer Lett 231(2):240–246 Matchett MD, MacKinnon SL, Sweeney MI, Gottschall-Pass KT, Hurta RA, 2006, Inhibition of matrix metalloproteinase activity in DU145 human prostate cancer cells by flavonoids from lowbush blueberry, Vaccinium Angustifolium): possible roles for protein kinase C and mitogen-activated protein-kinase-mediated events. J Nutr Biochem 17(2):117–125 Kausar H, Jeyabalan J, Aqil F, Chabba D, Sidana J, Singh IP, Gupta RC, 2012, Berry anthocyanidins synergistically suppress growth and invasive potential of human non-small-cell lung cancer cells. Cancer Lett 325(1):54–62 Diaconeasa Z, Leopold L, Rugina D, Ayvaz H, Socaciu C, 2015, Antiproliferative and antioxidant properties of anthocyanin rich extracts from blueberry and blackcurrant juice. Int J Mol Sci 16(2): 2352–2365 Bunea A, Rugina D, Sconta Z, Pop RM, Pintea A, Socaciu C, Tabaran F, Grootaert C, Struijs K, VanCamp J, 2013, Anthocyanin determination in blueberry extracts from various cultivars and their antiproliferative and apoptotic properties in B16- F10 metastatic murine melanoma cells. Phytochemistry 95:436– 444 Davidson KT, Zhu Z, Bai Q, Xiao H, Wakefield MR, Fang Y, 2017, Blueberry as a potential Radiosensitizer for treating cervical cancer. Pathol Oncol Res 22(4):655–60 Fang Y, Moore BJ, Bai Q, Cook KM, Herrick EJ, Nicholl MB, 2013, Hydrogen peroxide enhances radiation-induced apoptosis and inhibition of melanoma cell proliferation. Anticancer Res 33(5):1799–1807 Fang Y, Bradley MJ, Cook KM, Herrick EJ, Nicholl MB, 2013, A potential role for resveratrol as a radiation sensitizer for melanoma treatment. J Surg Res 183(2):645–653 Fang Y, DeMarco VG, Nicholl MB, 2012, Resveratrol enhances radiation sensitivity in prostate cancer by inhibiting cell proliferation and promoting cell senescence and apoptosis. Cancer Sci 103(6):1090–1098 Gordillo G, FangH, Khanna S, Harper J, Phillips G, Sen CK, 2009, Oral administration of blueberry inhibits angiogenic tumor growth and enhances survival of mice with endothelial cell neoplasm. Antioxid Redox Signal 11(1):47–58 Kai H, Akamatsu E, Torii E, Kodama H, Yukizaki C, Sakakibara Y, Suiko M, Morishita K, Kataoka H, Matsuno K, 2011, Inhibition of proliferation by agricultural plant extracts in seven human adult Tcell leukaemia, ATL)-related cell lines. J NatMed 65(3–4):651–655 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 733 EP 738 DI 10.1007/s12253-017-0376-2 PG 6 ER PT J AU Ren, X Zhang, H Cong, H Wang, X Ni, H Shen, X Ju, Sh AF Ren, Xiaojuan Zhang, Hui Cong, Hui Wang, Xudong Ni, Hongbing Shen, Xianjuan Ju, Shaoqing TI Diagnostic Model of Serum miR-193a-5p, HE4 and CA125 Improves the Diagnostic Efficacy of Epithelium Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Epithelium ovarian cancer; miRNA; miR-193a-5p; HE4; CA125 ID Epithelium ovarian cancer; miRNA; miR-193a-5p; HE4; CA125 AB Epithelium ovarian cancer (EOC) is currently the prevalent malignant cancer worldwide. However, there is a lack of efficient biomarkers for EOC screening. Accumulating evidence reveals that serum miRNA detectable in various types of cancer. Therefore, we explore the diagnostic value of combined detection of plasma miR-193a-5p, HE4 and CA125 for EOC. Serum samples were collected from 45 patients with primary EOC, 30 patients with benign ovarian tumor patients and 40 healthy controls. The expression of serum miR-193a-5p was detected by real-time quantitative PCR, and serum HE4 and CA125 were detected by chemiluminescent immunoassay. Moreover, a diagnostic model combining miR-193a-5p, HE4 and CA125 or alone in EOC patients was evaluated by ROC curve analysis. The relative expression quantity (RQ) of serum miR-193a-5p in EOC patients, benign ovarian tumor patients and healthy control groups were 0.419 (0.093, 2.215), 3.667 (1.633, 6.691) and 1.130 (1.000, 7.087), respectively. The RQ of serum miR-193a-5p in EOC patients was significantly lower than that in benign ovarian tumor patients and healthy controls (both P < 0.001), and there was no significant difference between benign ovarian tumor patients and healthy controls (both P > 0.05). There was no significant correlation between serum miR-193-5p, HE4 and CA125 levels (both P > 0.05). Additionally a risk model for miR-193a-5p, HE4 and CA125 was correlated with Grading and Lymph node metastasis (P = 0.016, P = 0.029). The area under the receiver operating characteristic curve of a risk model for distinguishing EOC patients from healthy individuals was 0.996, which higher than any single biomarker. Combined detection of miR-193-5p, HE4 and CA125 by logistic regression analysis could greatly improved the diagnostic ability of EOC and may prove to be a candidate biomarker, providing new directions for further investigation. C1 [Ren, Xiaojuan] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, #20 Xisi Road, 226001 Nantong, JS, China. [Zhang, Hui] Nantong Tumor Hospital, Laboratory Medicine Center, #48 Qingnian Road, 226001 Nantong, JS, China. [Cong, Hui] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, #20 Xisi Road, 226001 Nantong, JS, China. [Wang, Xudong] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, #20 Xisi Road, 226001 Nantong, JS, China. [Ni, Hongbing] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, #20 Xisi Road, 226001 Nantong, JS, China. [Shen, Xianjuan] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, #20 Xisi Road, 226001 Nantong, JS, China. [Ju, Shaoqing] Affiliated Hospital of Nantong University, Department of Laboratory Medicine, #20 Xisi Road, 226001 Nantong, JS, China. RP Shen, X (reprint author), Affiliated Hospital of Nantong University, Department of Laboratory Medicine, 226001 Nantong, China. EM juanxia819@163.com CR Siegel RL, Miller KD, Jemal A, 2015, Cancer statistics, 2015. CA Cancer J Clin 65(1):5–29 Meinhold-Heerlein I, Hauptmann S, 2014, The heterogeneity of ovarian cancer. Arch Gynecol Obstet 289(2):237–239 Anderson GL,McIntosh M,Wu L, BarnettM, Goodman G, Thorpe JD, Bergan L, Thornquist MD, Scholler N, Kim N, O'Briant K, Drescher C, Urban N, 2010, Assessing lead time of selected ovarian cancer biomarkers: a nested case-control study. J Natl Cancer Inst 102(1):26–38 Bartel DP, 2009, MicroRNAs: target recognition and regulatory functions. Cell 136(2):215–233 Shapira I, Oswald M, Lovecchio J, Khalili H, Menzin A, Whyte J, Dos Santos L, Liang S, Bhuiya T, Keogh M, Mason C, Sultan K, Budman D, Gregersen PK, Lee AT, 2014, Circulating biomarkers for detection of ovarian cancer and predicting cancer outcomes. Br J Cancer 110(4):976–983 Wu H, Xiao Z, Wang K, Liu W, Hao Q, 2013, MiR-145 is downregulated in human ovarian cancer and modulates cell growth and invasion by targeting p70S6K1 and MUC1. Biochem Biophys Res Commun 441(4):693–700 Petrillo M, Zannoni GF, Beltrame L, Martinelli E, DiFeo A, Paracchini L, Craparotta I, Mannarino L, Vizzielli G, Scambia G, D'IncalciM, Romualdi C,Marchini S, 2016, Identification of highgrade serous ovarian cancer miRNA species associated with survival and drug response in patients receiving neoadjuvant chemotherapy: a retrospective longitudinal analysis using matched tumor biopsies. Ann Oncol 27(4):625–634 Zhang H, Wang Q, Zhao Q, Di W, 2013, MiR-124 inhibits the migration and invasion of ovarian cancer cells by targeting SphK1. J Ovarian Res 6(1):84 Montagnana M, BenatiM, Danese E, 2017, Circulating biomarkers in epithelial ovarian cancer diagnosis: from present to future perspective. Ann Transl Med 5(13):276 Mahdian-Shakib A, Dorostkar R, Tat M, Hashemzadeh MS, Saidi N, 2016, Differential role of microRNAs in prognosis, diagnosis, and therapy of ovarian cancer. Biomed Pharmacother 84:592–600 Zhou J, Duan H, Xie Y, Ning Y, Zhang X, Hui N,Wang C, Zhang J, Zhou J, 2016, MiR-193a-5p targets the coding region of AP-2α mRNA and induces cisplatin resistance in bladder cancers. J Cancer 7(12):1740–1746 Lin CH, Tsai CH, Yeh CT, Liang JL, Hung WC, Lin FC, Chang WL, Li HY, Yao YC, Hsu TI, Lee YC, Wang YC, Sheu BS, Lai WW, CalkinsMJ, Hsiao M, Lu PJ, 2016)MiR-193a-5p/ERBB2 act as concurrent chemoradiation therapy response indicator of esophageal squamous cell carcinoma. Oncotarget 7(26):39680–39693 Yu T, Li J, Yan M, Liu L, Lin H, Zhao F, Sun L, Zhang Y, Cui Y, Zhang F, Li J, He X, Yao M, 2015, MicroRNA-193a-3p and -5p suppress the metastasis of human non-small-cell lung cancer by downregulating the ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Oncogene 34(4):413–423 Wang X, Cao L, Wang Y, Wang X, Liu N, You Y, 2012, Regulation of let-7 and its target oncogenes, review). Oncol Lett 3:955–960 Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ, 2008, The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol 10(5):593–601 Lou Y, Yang X, Wang F, Cui Z, Huang Y, 2010, MicroRNA-21 promotes the cell proliferation, invasion and migration abilities in ovarian epithelial carcinomas through inhibiting the expression of PTEN protein. Int J Mol Med 26:819–827 Nakano H, Yamada Y, Miyazawa T, Yoshida T, 2013, Gain-offunction microRNA screens identify miR-193a regulating proliferation and apoptosis in epithelial ovarian cancer cells. Int J Oncol 42: 1875–1882 Wang C, Ding M, Xia M, Chen S, Van Le A, Soto-Gil R, Shen Y, Wang N,Wang J, GuW,Wang X, Zhang Y, Zen K, Chen X, Zhang C, Zhang CY, 2015, A five-miRNA panel identified from a multicentric case-control study serves as a novel diagnostic tool for ethnically diverse non-small-cell lung cancer patients. EBioMedicine 2(10):1377–1385 Dong H, Wang C, Lu S, Yu C, Huang L, Feng W, Xu H, Chen X, Zen K, Yan Q, Liu W, Zhang C, Zhang CY, 2016, A panel of four decreased serum microRNAs as a novel biomarker for early Parkinson's disease. Biomarkers 21(2):129–137 Prinsloo A, Pool R, Van Niekerk C, 2017, Preliminary data on microRNA expression profiles in a group of south African patients diagnosed with chronic myeloid leukaemia. Mol Clin Oncol 7(3): 386–390 Duffy MJ, Bonfrer JM, Kulpa J, Rustin GJ, Soletormos G, Torre GC, Tuxen MK, Zwirner M, 2005, CA125 in ovarian cancer: European group on tumor markers guidelines for clinical use. Int J Gynecol Cancer 15(5):679–691 Cho HY, Park SH, Park YH, Kim HB, Kang JB, Hong SH, Kyung MS, 2015, Comparison of HE4, CA125, and risk of ovarian malignancy algorithm in the prediction of ovarian cancer in Korean women. J Korean Med Sci 30(12):1777–1783 Piovano E, Attamante L, Macchi C, Cavallero C, Romagnolo C, Maggino T, Landoni F, Gadducci A, Sartori E, Gion M, Zola P, 2014, The role of HE4 in ovarian cancer follow-up: a review. Int J Gynecol Cancer 24(8):1359–1365 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 739 EP 744 DI 10.1007/s12253-018-0392-x PG 6 ER PT J AU Laskar, J Bhattacharjee, K Sengupta, M Choudhury, Y AF Laskar, Jeny Bhattacharjee, Kasturi Sengupta, Mahuya Choudhury, Yashmin TI Anti-Diabetic Drugs: Cure or Risk Factors for Cancer? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Anti-cancer effect; Anti-diabetic drugs; Cancer risk ID Anti-cancer effect; Anti-diabetic drugs; Cancer risk AB Anti-diabetic drugs are an important group of therapeutics used worldwide. Different anti-diabetic drugs lower blood glucose level by differentmechanisms. In recent years, numerous investigations have been performed based on both comparative and cohort studies, in order to establish the relationship between anti-diabetic pharmacotherapy and cancer incidence as well as mortality due to cancer. Some anti-diabetic drugs have been found to exhibit anti-cancer activitywhile others might increase the risk for cancer. The underlying cause for this disparity is likely to be the varying mechanisms of action of these drugs in controlling blood glucose level. This review discusses the various carcinogenic and/or anti-cancer effects of commonly used anti-diabetic drugs. The information is vital in view of the fact that diabetes mellitus is a commonly occurring disease with a rising incidence rate. C1 [Laskar, Jeny] Assam University, Department of Biotechnology, 788011 Silchar, India. [Bhattacharjee, Kasturi] Assam University, Department of Biotechnology, 788011 Silchar, India. [Sengupta, Mahuya] Assam University, Department of Biotechnology, 788011 Silchar, India. [Choudhury, Yashmin] Assam University, Department of Biotechnology, 788011 Silchar, India. RP Choudhury, Y (reprint author), Assam University, Department of Biotechnology, 788011 Silchar, India. EM yashminchoudhury@gmail.com;yashminchoudhury@aus.ac.in CR Vigneri P, Frasca F, Sciacca L, Pandini G, Vigneri R, 2009, Diabetes mellitus and cancer. Endocr Relat Cancer 16:1103– 1123. , DOI 10.1677/ERC-09-0087 Forouhi NG, Wareham NJ, 2014, Epidemiology of diabetes. Medicine, Abingdon, 42(12):698–702., DOI 10.1016/j. mpmed.2014.09.007 Garcia-Jimenez C, Gutierrez-Salmeron M, Chocarro-Calvo A, Garcia-Martinez JM, Castano A, de la Vieja A, 2016, From obesity to diabetes and cancer: epidemiological links and role of therapies. Br J Cancer 114:716–722. , DOI 10.1038/bjc.2016. 37 Fitzmaurice C, Allen C, Barber RM et al, 2017, Global, regional, and National Cancer Incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 32 Cancer groups, 1990 to 2015: a systematic analysis for the global burden of disease study. JAMA Oncol 3:524–548. , DOI 10.1001/jamaoncol.2016.5688 Richardson LC, Pollack LA, 2005, Therapy insight: influence of type 2 diabetes on the development, treatment and outcomes of cancer. Nat Clin Pract Oncol 2:48–53. , DOI 10.1038/ ncponc0062 Song S, Wang B, Zhang X, Hao L, Hu X, Li Z, Sun S, 2015, Long-term diabetes mellitus is associated with an increased risk of pancreatic Cancer: a meta-analysis. PLoS One 10:e0134321. , DOI 10.1371/journal.pone.0134321 Xu B, Zhou X, Li X, Liu C, Yang C, 2017, Diabetes mellitus carries a risk of esophageal cancer: a meta-analysis. Medicine, Baltimore, 96:e7944. https:/ /doi .org/10.1097/MD. 0000000000007944 Dehal AN, Newton CC, Jacobs EJ, Patel AV, Gapstur SM, Campbell PT, 2012, Impact of diabetes mellitus and insulin use on survival after colorectal Cancer diagnosis: the Cancer prevention study-II nutrition cohort. J Clin Oncol 30:53–59. https://doi. org/10.1200/JCO.2011.38.0303 Nguyen QT, Sanders L, Michael AP, Anderson SR, Nguyen LD, Johnson ZA, 2012, Diabetes medications and cancer risk: review of the literature. Am Health Drug Benefits 5:221–229 Anand P, Kunnumakara AB, Sundaram C, Harikumar KB, Tharakan ST, Lai OS, Sung B, Aggarwal BB, 2008, Cancer is a preventable disease that requires major lifestyle changes. Pharm Res 25:2097–2116. , DOI 10.1007/s11095-008-9661-9 Kolb H, Martin S, 2017, Environmental/lifestyle factors in the pathogenesis and prevention of type 2 diabetes. BMC Med 15: 131. , DOI 10.1186/s12916-017-0901-x Hua F, Yu J-J, Hu Z-W, 2016, Diabetes mellitus and cancer, common threads andmissing links. Cancer Lett 374:54–61. https://doi. org/10.1016/j.canlet.2016.02.006 Roberts CK, Hevener AL, Barnard RJ, 2013, Metabolic syndrome and insulin resistance: underlying causes and modification by exercise training. Compr Physiol 3(1):1–58. , DOI 10. 1002/cphy.c110062 UzunluluM, Telci Caklili O, Oguz A, 2016, Association between metabolic syndrome and Cancer. Ann Nutr Metab 68:173–179. , DOI 10.1159/000443743 Arcidiacono B, Iiritano S, Nocera A, Possidente K, Nevolo MT, Ventura V, Foti D, Chiefari E, Brunetti A, 2012, Insulin resistance and Cancer risk: an overview of the Pathogenetic mechanisms. Exp Diabetes Res 2012:1–12. , DOI 10.1155/2012/ 789174 Giovannucci E, Harlan DM, ArcherMC, Bergenstal RM, Gapstur SM, Habel LA, Pollak M, Regensteiner JG, Yee D, 2010, Diabetes and Cancer: a consensus report. CA Cancer J Clin 60: 207–221. , DOI 10.3322/caac.20078 Eckel RH, Kahn SE, Ferrannini E, Goldfine AB, Nathan DM, Schwartz MW, Smith RJ, Smith SR, Endocrine Society., American Diabetes Association., European Association for the Study of Diabetes, 2011, Obesity and type 2 diabetes mellitus: what can be unified and what needs to be individualized? Diabetes Care 34:1424–1430. , DOI 10.2337/dc11-0447 De Pergola G, Silvestris F, 2013, Obesity as a major risk factor for Cancer. J Obes 2013:1–11. , DOI 10.1155/2013/291546 Vona-Davis L, Rose DP, 2007, Adipokines as endocrine, paracrine, and autocrine factors in breast cancer risk and progression. Endocr Relat Cancer 14:189–206. , DOI 10.1677/ERC- 06-0068 Vasconcelos-dos-Santos A, Loponte HFBR, Mantuano NR, Oliveira IA, de Paula IF, Teixeira LK, de-Freitas-Junior JCM, Gondim KC, Heise N, Mohana-Borges R, Morgado-Diaz JA, Dias WB, Todeschini AR, 2017, Hyperglycemia exacerbates colon cancer malignancy through hexosamine biosynthetic pathway. Oncogenesis 6:e306. , DOI 10.1038/oncsis.2017.2 Gonzalez N, Prieto I, del P-NL et al, 2017, 2017 update on the relationship between diabetes and colorectal cancer: epidemiology, potential molecular mechanisms and therapeutic implications. Oncotarget 8:18456–18485. , DOI 10.18632/oncotarget. 14472 Ye A, 2009, Historical review of anti-diabetic drugs’ discovery. Zhonghua Yi Shi Za Zhi Beijing China 1980 39:229–231 Patade G, Marita AR, 2014, Metformin: a journey from countryside to the bedside. J Obes Metab Res 1:127. , DOI 10. 4103/2347-9906.134435 Bailey C, Day C, 2004, Metformin: its botanical background. Pract Diabetes Int 21:115–117. , DOI 10.1002/pdi.606 Rojas LBA, GomesMB, 2013, Metformin: an old but still the best treatment for type 2 diabetes. Diabetol Metab Syndr 5:6. https:// doi.org/10.1186/1758-5996-5-6 Barnett D, Craig JG, Robinson DS, Rogers MP, 1977, Effect of clofibrate on glucose tolerance in maturity onset diabetes. Br J Clin Pharmacol 4:455–458 Quianzon CCL, Cheikh IE, 2012, History of current non-insulin medications for diabetes mellitus. J Community Hosp Intern Med Perspect 2:19081. , DOI 10.3402/jchimp.v2i3.19081 Hiatt WR, Kaul S, Smith RJ, 2013, The cardiovascular safety of diabetes drugs — insights from the rosiglitazone experience. N Engl J Med 369:1285–1287. , DOI 10.1056/ NEJMp1309610 Mehanna A, 2013, Anti-diabetic agents: past, present and future. Future Med Chem 5:411–430. , DOI 10.4155/fmc.13.13 Jambon S, Chaptal J, Vedel A, Schaap J, 1942, Accidents hypoglycemiques graves par un sulfamido-thiadiazol. Montp Med 21:441–445 Mulder H, Schopman W, van der Lely AJ, 1991, Extrapancreatic insulin effect of glibenclamide. Eur J Clin Pharmacol 40:379–381. , DOI 10.1007/BF00265847 Sola D, Rossi L, Schianca GPC, Maffioli P, Bigliocca M, Mella R, Corliano F, Fra GP, Bartoli E, Derosa G, 2015, State of the art paper sulfonylureas and their use in clinical practice. Arch Med Sci 4:840–848. , DOI 10.5114/aoms.2015.53304 Bayliss WM, Starling EH, 1902, The mechanism of pancreatic secretion. J Physiol 28:325–353 Eng J, Kleinman WA, Singh L, Singh G, Raufman JP, 1992, Isolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum venom. Further evidence for an exendin receptor on dispersed acini from guinea pig pancreas. J Biol Chem 267:7402–7405 Neumiller JJ, 2009, Differential chemistry, structure), mechanism of action, and pharmacology of GLP-1 receptor agonists and DPP- 4 inhibitors. J Am Pharm Assoc 49:S16–S29. , DOI 10. 1331/JAPhA.2009.09078 Gibbs JP, Fredrickson J, Barbee T, Correa I, Smith B, Lin SL, Gibbs MA, 2012, Quantitative model of the relationship between Dipeptidyl Peptidase-4, DPP-4, inhibition and response: metaanalysis of Alogliptin, Saxagliptin, Sitagliptin, and Vildagliptin efficacy results. J Clin Pharmacol 52:1494–1505. , DOI 10.1177/0091270011420153 Gomis R, Espadero R-M, Jones R, Woerle HJ, Dugi KA, 2011, Efficacy and safety of initial combination therapy with linagliptin and pioglitazone in patients with inadequately controlled type 2 diabetes: a randomized, double-blind, placebo-controlled study. Diabetes Obes Metab 13:653–661. , DOI 10.1111/j. 1463-1326.2011.01391.x Meienhofer J, Schnabel E, BremerH, Brinkhoff O, Zabel R, Sroka W, Klostermeyer H, Brandenburg D, Okuda T, Zahn H, 1963, Notizen: Synthese der Insulinketten und ihre Kombination zu insulinaktiven Praparaten. Z Fur Naturforschung B 18. https:// doi.org/10.1515/znb-1963-1223 Kung YT, Du YC, Huang WT et al, 1965, Total synthesis of crystalline bovine insulin. Sci Sin 14(11):1710-6. Katsoyannis PG, Tometsko A, Zalut C, 1966, Insulin peptides. XII. Human insulin generation by combination of synthetic a and B chains 1. J Am Chem Soc 88:166–167. , DOI 10.1021/ ja00953a033 Mathieu C, Gale EAM, 2007, Inhaled insulin: gone with the wind? Diabetologia 51:1–5. , DOI 10.1007/s00125-007- 0875-x Sciacca L, Moli RL, Vigneri R, 2012, Insulin analogs and Cancer. Front Endocrinol 3. , DOI 10.3389/fendo.2012.00021 Pollak MN, 2012, Investigating metformin for Cancer prevention and treatment: the end of the beginning. Cancer Discov 2:778– 790. , DOI 10.1158/2159-8290.CD-12-0263 Maruthur NM, Tseng E, Hutfless S, Wilson LM, Suarez-Cuervo C, Berger Z, Chu Y, Iyoha E, Segal JB, Bolen S, 2016, Diabetes medications as Monotherapy or metformin-based combination therapy for type 2 diabetes: a systematic review andmeta-analysis. Ann Intern Med 164:740–751. , DOI 10.7326/M15-2650 Strowig SM, Aviles-Santa ML, Raskin P, 2002, Comparison of insulin monotherapy and combination therapy with insulin and metformin or insulin and troglitazone in type 2 diabetes Diabetes Care 25:1691–1698 Viollet B, Guigas B, Garcia NS, Leclerc J, Foretz M, Andreelli F, 2012, Cellular and molecular mechanisms ofmetformin: an overview. Clin Sci 122:253–270. , DOI 10.1042/CS20110386 Evans JMM, 2005, Metformin and reduced risk of cancer in diabetic patients. BMJ 330:1304–1305. , DOI 10.1136/bmj. 38415.708634.F7 Soranna D, Scotti L, Zambon A, Bosetti C, Grassi G, Catapano A, la Vecchia C, Mancia G, Corrao G, 2012, Cancer risk associated with use of metformin and sulfonylurea in type 2 diabetes: a metaanalysis. Oncologist 17:813–822. , DOI 10.1634/ theoncologist.2011-0462 Tang Y-L, Zhu L-Y, Li Y, Yu J,Wang J, Zeng XX, Hu KX, Liu JY, Xu JX, 2017, Metformin use is associated with reduced incidence and improved survival of endometrial Cancer: a meta-analysis. Biomed Res Int 2017:1–9. , DOI 10.1155/2017/5905384 Fan C, Wang Y, Liu Z et al, 2015, Metformin exerts anticancer effects through the inhibition of the sonic hedgehog signaling pathway in breast cancer. Int J Mol Med 36:204–214. https://doi. org/10.3892/ijmm.2015.2217 Gou S, Cui P, Li X, Shi P, Liu T, Wang C, 2013, Low concentrations of metformin selectively inhibit CD133+ cell proliferation in pancreatic cancer and have anticancer action. PLoS One 8:e63969. , DOI 10.1371/journal.pone.0063969 Tseng C-H, 2014, Metformin reduces thyroid cancer risk in Taiwanese patients with type 2 diabetes. PLoS One 9:e109852. , DOI 10.1371/journal.pone.0109852 Kato K, Gong J, Iwama H, Kitanaka A, Tani J, Miyoshi H, Nomura K, Mimura S, Kobayashi M, Aritomo Y, Kobara H, Mori H, Himoto T, Okano K, Suzuki Y, Murao K, Masaki T, 2012, The anti-diabetic drug metformin inhibits gastric cancer cell proliferation in vitro and in vivo. Mol Cancer Ther 11:549– 560. , DOI 10.1158/1535-7163.MCT-11-0594 Tseng C-H, 2016, Metformin reduces gastric cancer risk in patients with type 2 diabetes mellitus. Aging 8:1636–1649. https:// doi.org/10.18632/aging.101019 Tseng C-H(1990,, 2014)metformin significantly reduces incident prostate cancer risk in Taiwanese men with type 2 diabetes mellitus. Eur J Cancer Oxf Engl 50:2831–2837. , DOI 10.1016/j.ejca.2014.08.007 Tseng C-H, 2017, Metformin is associated with a lower risk of colorectal cancer in Taiwanese patients with type 2 diabetes: a retrospective cohort analysis. Diabetes Metab 43:438–445. , DOI 10.1016/j.diabet.2017.03.004 Tseng C-H, 2014, Metformin may reduce bladder cancer risk in Taiwanese patients with type 2 diabetes. Acta Diabetol 51:295– 303. , DOI 10.1007/s00592-014-0562-6 Tseng C-H, 2016, Metformin may reduce oral cancer risk in patients with type 2 diabetes. Oncotarget 7:2000–2008. https://doi. org/10.18632/oncotarget.6626 Kasznicki J, Sliwinska A, Drzewoski J, 2014, Metformin in cancer prevention and therapy. Ann Transl Med 2:57. , DOI 10.3978/j.issn.2305-5839.2014.06.01 Tseng C-H, 2017)Metformin and lung cancer risk in patients with type 2 diabetes mellitus mellitus. Oncotarget 8:41132–41142. , DOI 10.18632/oncotarget.17066 Tseng C-H, 2017, Metformin and esophageal cancer risk in Taiwanese patients with type 2 diabetes mellitus. Oncotarget 8: 18802–18810. , DOI 10.18632/oncotarget.13390 Tseng C-H, 2016, Metformin use and cervical cancer risk in female patients with type 2 diabetes mellitus. Oncotarget 7:59548– 59555. , DOI 10.18632/oncotarget.10934 Tseng C-H, 2016, use of metformin and risk of kidney cancer in patients with type 2 diabetes. Eur J Cancer Oxf Engl 52:19–25. , DOI 10.1016/j.ejca.2015.09.027 Tseng C-H, 2015, Metformin reduces ovarian cancer risk in Taiwanese women with type 2 diabetes mellitus. mellitus Diabetes mellitus Metab Res Rev 31:619–626. , DOI 10.1002/dmrr.2649 Tseng C-H, 2014, Metformin may reduce breast cancer risk in Taiwanese women with type 2 diabetes. Breast Cancer Res Treat 145:785–790. , DOI 10.1007/s10549-014-2985-8 Neumann A, Weill A, Ricordeau P, Fagot JP, Alla F, Allemand H, 2012, Pioglitazone and risk of bladder cancer among diabetic patients in France: a population-based cohort study. Diabetologia 55:1953–1962. , DOI 10.1007/s00125-012-2538-9 Ferrara A, Lewis JD, Quesenberry CP, Peng T, Strom BL, van den Eeden SK, Ehrlich SF, Habel LA, 2011, Cohort study of pioglitazone and cancer incidence in patients with diabetes. Diabetes Care 34:923–929. , DOI 10.2337/dc10-1067 Tseng C-H, 2017, Rosiglitazone reduces breast cancer risk in Taiwanese female patients with type 2 diabetes mellitus. Oncotarget 8:3042–3048. , DOI 10.18632/oncotarget. 13824 Tseng C-H, 2015, Rosiglitazone may reduce non-melanoma skin cancer risk in Taiwanese. BMC Cancer 15:41. , DOI 10. 1186/s12885-015-1057-8 Chang C-H, Lin J-W, Wu L-C, Lai MS, Chuang LM, 2012, Oral insulin Secretagogues, insulin, and Cancer risk in type 2 diabetes mellitus. J Clin Endocrinol Metab 97:E1170–E1175. https://doi. org/10.1210/jc.2012-1162 Tuccori M,Wu JW, Yin H, Majdan A, Azoulay L, 2015, The use of glyburide compared with other sulfonylureas and the risk of Cancer in patients with type 2 diabetes. Diabetes Care 38:2083– 2089. , DOI 10.2337/dc15-1358 Qi C, Zhou Q, Li B, Yang Y, Cao L, Ye Y, Li J, Ding Y, Wang H, Wang J, He X, Zhang Q, Lan T, Lee KKH, LiW, Song X, Zhou J, Yang X, Wang L, 2014, Glipizide, an anti-diabetic drug, suppresses tumor growth and metastasis by inhibiting angiogenesis. Oncotarget 5:9966–9979. , DOI 10.18632/oncotarget. 2483 Elashoff M, Matveyenko AV, Gier B, Elashoff R, Butler PC, 2011, Pancreatitis, pancreatic, and thyroid Cancer with glucagon-like Peptide-1–based therapies. Gastroenterology 141: 150–156. , DOI 10.1053/j.gastro.2011.02.018 Tseng C-H, 2016, Sitagliptin and pancreatic cancer risk in patients with type 2 diabetes. Eur J Clin Investig 46:70–79. , DOI 10.1111/eci.12570 Tseng C-H, 2016, Sitagliptin use and thyroid cancer risk in patients with type 2 diabetes. Oncotarget 7:24871–24879. https:// doi.org/10.18632/oncotarget.8399 Raz I, Bhatt DL, Hirshberg B, Mosenzon O, Scirica BM, Umez- Eronini A, Im KA, Stahre C, Buskila A, Iqbal N, Greenberger N, Lerch MM, 2014, Incidence of pancreatitis and pancreatic cancer in a randomized controlled multicenter trial, SAVOR-TIMI 53, of the dipeptidyl peptidase-4 inhibitor saxagliptin. Diabetes Care 37: 2435–2441. , DOI 10.2337/dc13-2546 Tseng C-H, 2015, Prolonged use of human insulin increases breast cancer risk in Taiwanese women with type 2 diabetes. BMC Cancer 15:846. , DOI 10.1186/s12885-015-1876-7 Gu Y, Wang C, Zheng Y, Hou X, Mo Y, Yu W, Zhang L, Hu C, Nan H, Chen L, Li J, Liu Y, Huang Z, Han M, Bao Y, Zhong W, JiaW(2013, Cancer incidence and mortality in patients with type 2 diabetes treated with human insulin: a cohort study in shanghai. PLoS One 8:e53411. , DOI 10.1371/journal.pone. 0053411 Ferguson RD, Gallagher EJ, Scheinman EJ, Damouni R, LeRoith D, 2013, The epidemiology and molecular mechanisms linking obesity, diabetes, and cancer. Vitam Horm, 93):51–98 Cui W, Ji M-J, Chen H et al, 2016, Insulin analogs detemir, glargine and lispro enhance proliferation of human thyroid and gastric normal or cancer cell lines. Int J Clin Exp Med 9:1603– 1611 Aizen D, Sarfstein R, Bruchim I,Weinstein D, Laron Z,Werner H, 2015, Proliferative and signaling activities of insulin analogues in endometrial cancer cells. Mol Cell Endocrinol 406:27–39. https:// doi.org/10.1016/j.mce.2015.02.011 Hemkens LG, Grouven U, Bender R, Gunster C, Gutschmidt S, Selke GW, Sawicki PT, 2009, Risk of malignancies in patients with diabetes treated with human insulin or insulin analogues: a cohort study. Diabetologia 52:1732–1744. , DOI 10. 1007/s00125-009-1418-4 Lin H-W, Tseng C-H, 2014, A review on the relationship between SGLT2 inhibitors and Cancer. Int J Endocrinol 2014:719578– 719576. , DOI 10.1155/2014/719578 Ptaszynska A, Cohen SM, Messing EM, Reilly TP, Johnsson E, Johnsson K, 2015, Assessing bladder Cancer risk in type 2 diabetes clinical trials: the Dapagliflozin drug development program as a Bcase study^. Diabetes Ther Res Treat Educ Diabetes mellitus Relat Disord 6:357–375. , DOI 10.1007/s13300-015- 0128-9 Tseng Y-H, Tsan Y-T, Chan W-C, Sheu WHH, Chen PC, 2015, Use of an α-Glucosidase inhibitor and the risk of colorectal Cancer in patients with diabetes mellitus: a Nationwide, population-based cohort study. Diabetes Care 38:2068–2074. , DOI 10.2337/dc15-0563 Dowling RJO, Goodwin PJ, Stambolic V, 2011, Understanding the benefit of metformin use in cancer treatment. BMC Med 9:33. , DOI 10.1186/1741-7015-9-33 Kahn BB, Alquier T, Carling D, Hardie DG, 2005, AMPactivated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab 1:15–25. , DOI 10.1016/j.cmet.2004.12.003 Dowling RJO, Niraula S, Stambolic V, Goodwin PJ, 2012, Metformin in cancer: translational challenges. J Mol Endocrinol 48:R31–R43. , DOI 10.1530/JME-12-0007 Gwinn DM, Shackelford DB, Egan DF,MihaylovaMM, Mery A, Vasquez DS, Turk BE, Shaw RJ, 2008, AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell 30:214–226. , DOI 10.1016/j.molcel.2008.03.003 Howell JJ, Hellberg K, Turner M, Talbott G, Kolar MJ, Ross DS, Hoxhaj G, Saghatelian A, Shaw RJ, Manning BD, 2017, Metformin inhibits hepatic mTORC1 signaling via dose-dependent mechanisms involving AMPK and the TSC complex. Cell Metab 25:463–471. , DOI 10.1016/j.cmet.2016.12.009 Xie J, Wang X, Proud CG, 2016, mTOR inhibitors in cancer therapy. F1000Research 5:2078. , DOI 10.12688/ f1000research.9207.1 Ben Sahra I, Regazzetti C, Robert G, Laurent K, le Marchand- Brustel Y, Auberger P, Tanti JF, Giorgetti-Peraldi S, Bost F, 2011, Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res 71:4366–4372. , DOI 10.1158/0008-5472.CAN-10-1769 Memmott RM, Mercado JR, Maier CR, Kawabata S, Fox SD, Dennis PA, 2010, Metformin prevents tobacco carcinogen– induced lung tumorigenesis. Cancer Prev Res Phila, Phila, 3: 1066–1076. , DOI 10.1158/1940-6207.CAPR-10-0055 Hirsch HA, Iliopoulos D, Struhl K, 2013, Metformin inhibits the inflammatory response associated with cellular transformation and cancer stem cell growth. Proc Natl Acad Sci U S A 110:972–977. , DOI 10.1073/pnas.1221055110 Malik S, Upadhyaya PK, Miglani S, 2011, Thiazolidinediones: a Plethro of biological load. Int J PharmTech Res 3:62–75 Sauter M, Kastenmuller K, Belling F, Wornle M, Ladurner R, Mussack T, Sitter T, 2012, Activation of peroxisome proliferator-activated receptor-gamma by Glitazones reduces the expression and release of monocyte Chemoattractant Protein-1 in human Mesothelial cells. Mediat Inflamm 2012:1–6. https://doi. org/10.1155/2012/217696 Yanai H, Adachi H, 2017, The low-dose, 7.5 mg/day, pioglitazone therapy. J Clin Med Res 9:821–825. , DOI 10. 14740/jocmr3144w Davidson MA, Mattison DR, Azoulay L, Krewski D, 2018, Thiazolidinedione drugs in the treatment of type 2 diabetes mellitus: past, present and future. Crit Rev Toxicol 48(1):52– 108. , DOI 10.1080/10408444.2017.1351420 Chang C-H, Lin J-W,Wu L-C, LaiMS, Chuang LM, Arnold Chan K, 2012, Association of thiazolidinediones with liver cancer and colorectal cancer in type 2 diabetes mellitus. mellitus Hepatol Baltim Md 55:1462–1472. , DOI 10.1002/hep.25509 Mamtani R, Haynes K, Bilker WB, Vaughn DJ, Strom BL, Glanz K, Lewis JD, 2012, Association between longer therapy with Thiazolidinediones and risk of bladder Cancer: a cohort study. JNCI J Natl Cancer Inst 104:1411–1421. , DOI 10.1093/ jnci/djs328 Blanquicett C, Roman J, Hart CM, 2008, Thiazolidinediones as anti-cancer agents. Cancer Ther 6:25–34 Chou F-S, Wang P-S, Kulp S, Pinzone JJ, 2007, Effects of thiazolidinediones on differentiation, proliferation, and apoptosis. Mol Cancer Res MCR 5:523–530. , DOI 10.1158/1541- 7786.MCR-06-0278 Okumura T, 2010, Mechanisms by which thiazolidinediones induce anti-cancer effects in cancers in digestive organs. J Gastroenterol 45:1097–1102. , DOI 10.1007/s00535- 010-0310-9 Wu C-W, Farrell GC, Yu J, 2012, Functional role of peroxisomeproliferator- activated receptor γ in hepatocellular carcinoma: PPARγ in hepatocellular carcinoma. J Gastroenterol Hepatol 27: 1665–1669. , DOI 10.1111/j.1440-1746.2012.07213.x Johnson JA, Gale EAM, 2010, Diabetes, insulin use, and Cancer risk: are observational studies part of the solution-or part of the problem? Diabetes 59:1129–1131. , DOI 10.2337/db10- 0334 Platts J, 2010, Insulin therapy and cancer risk in diabetes mellitus. ClinMed 10:509–512. , DOI 10.7861/clinmedicine.10-5- 509 Mannucci E, 2012, Insulin therapy and Cancer in type 2 diabetes. ISRN Endocrinol 2012:1–12. , DOI 10.5402/2012/ 240634 Home P, 2013, Insulin therapy and Cancer. Diabetes Care 36: S240–S244. , DOI 10.2337/dcS13-2002 Morrione A, Valentinis B, Xu SQ, Yumet G, Louvi A, Efstratiadis A, Baserga R, 1997, Insulin-like growth factor II stimulates cell proliferation through the insulin receptor. Proc Natl Acad Sci U S A 94:3777–3782 Tokajuk A, Krzyzanowska-Grycel E, Tokajuk A, Grycel S, Sadowska A, Car H, 2015, Anti-diabetic drugs and risk of cancer. Pharmacol Rep 67:1240–1250. , DOI 10.1016/j.pharep. 2015.05.005 Tseng C-H, Lee K-Y, Tseng F-H, 2015, An updated review on Cancer risk associated with Incretin Mimetics and enhancers. J Environ Sci Health Part C 33:67–124. , DOI 10.1080/ 10590501.2015.1003496 Liu Z, Habener JF, 2008, Glucagon-like Peptide-1 activation of TCF7L2-dependent Wnt signaling enhances pancreatic Beta cell proliferation. J Biol Chem 283:8723–8735. , DOI 10. 1074/jbc.M706105200 Funch D, Gydesen H, Tornoe K, Major-Pedersen A, Chan KA, 2014, A prospective, claims-based assessment of the risk of pancreatitis and pancreatic cancer with liraglutide compared to other anti-diabetic drugs. Diabetes ObesMetab 16:273–275. https://doi. org/10.1111/dom.12230 Proks P, Reimann F, Green N et al, 2002, Sulfonylurea stimulation of insulin secretion. Diabetes 51:S368–S376. , DOI 10. 2337/diabetesmellitus.51.2007.S368 Tseng C-H, 2012, Thyroid cancer risk is not increased in diabetic patients. PLoS One 7:e53096. , DOI 10.1371/journal. pone.0053096 Monami M, Lamanna C, Balzi D, Marchionni N, Mannucci E, 2009, Sulphonylureas and cancer: a case–control study. Acta Diabetol 46:279–284. , DOI 10.1007/s00592-008-0083-2 Li D, Yeung SJ, Hassan MM et al, 2009, Anti-diabetic therapies affect risk of pancreatic Cancer. Gastroenterology 137:482–488. , DOI 10.1053/j.gastro.2009.04.013 Pasello G, Urso L, Conte P, Favaretto A, 2013, Effects of sulfonylureas on tumor growth: a review of the literature. The Oncologist 18:1118–1125. , DOI 10.1634/theoncologist. 2013-0177 Bischoff H, 1995, The mechanism of alpha-glucosidase inhibition in the management of diabetes. Clin Investig Med 18:303–311 Kumar V, Prakash O, Kumar S, Narwal S, 2011, α-glucosidase inhibitors from plants: a natural approach to treat diabetes. Pharmacogn Rev 5:19–29. , DOI 10.4103/0973-7847. 79096 Matsuura H, Asakawa C, Kurimoto M, Mizutani J, 2002, α- Glucosidase inhibitor from the seeds of balsam pear, Momordica charantia , and the fruit bodies of Grifola frondosa. Biosci Biotechnol Biochem 66:1576–1578. , DOI 10. 1271/bbb.66.1576 Lai S-W, Liao K-F, Chen P-C, Tsai PY, Hsieh DPH, Chen CC, 2012, Antidiabetes drugs correlate with decreased risk of lung Cancer: a population-based observation in Taiwan. Clin Lung Cancer 13:143–148. , DOI 10.1016/j.cllc.2011.10.002 Chao EC, Henry RR, 2010, SGLT2 inhibition—a novel strategy for diabetes treatment. Nat Rev Drug Discov 9:551–559. https:// doi.org/10.1038/nrd3180 Taylor SR, Harris KB, 2013, The clinical efficacy and safety of sodium glucose Cotransporter-2 inhibitors in adults with type 2 diabetes mellitus. Pharmacotherapy 33:984–999. , DOI 10.1002/phar.1303 Ferrannini E, Ramos SJ, Salsali A, Tang W, List JF, 2010, Dapagliflozin Monotherapy in type 2 diabetic patients with inadequate glycemic control by diet and exercise: a randomized, double- blind, placebo-controlled, phase 3 trial. Diabetes Care 33: 2217–2224. , DOI 10.2337/dc10-0612 LeRoith D, Scheinman E, Bitton-Worms K, 2011, The role for insulin and insulin-like growth factors in the increased risk of Cancer in diabetes. Rambam Maimonides Med J 2:e0043. , DOI 10.5041/RMMJ.10043 Bowker SL, Majumdar SR, Veugelers P, Johnson JA, 2006, Increased cancer-relatedmortality for patients with type 2 diabetes mellitus who use sulfonylureas or insulin. Diabetes Care 29:254– 258 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 745 EP 755 DI 10.1007/s12253-018-0402-z PG 11 ER PT J AU Puneet, SM Kazmi, RH Kumari, S Tiwari, S Khanna, KA Narayan, G AF Puneet, S M Kazmi, Raza Hasan Kumari, Soni Tiwari, Satendra Khanna, Kumar Ajay Narayan, Gopeshwar TI Epigenetic Mechanisms and Events in Gastric Cancer-Emerging Novel Biomarkers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE DNA methylation; Epigenetics; Gastric cancer; Long noncoding RNA; microRNAs; Promoter methylation ID DNA methylation; Epigenetics; Gastric cancer; Long noncoding RNA; microRNAs; Promoter methylation AB Gastric cancer is one of the most common malignancy worldwide. The various genetic and epigenetic events have been found to be associated with its carcinogenesis. The epigenetic is a heritable and transient/reversible change in the gene expression that is not accompanied by modification in the DNA sequence. This event is characterized by the alteration in the promoter CpG island of the gene or histone modification. These events are associated with silencing of critical tumor suppressor gene and activation of oncogenes leading to carcinogenesis. The DNA methylation is a chemical change in the DNA sequence that most commonly occurs at cytosine moiety of CpG dinucleotide and histone, primarily on N- terminal tail that ultimately effect the interaction of DNA with chromatin modifying protein. Hypermethylation of tumor suppressor genes and global hypomethylation of oncogenes are widely studied epigenetic modifications. There are large number of publish reports regarding epigenetic events involving gastric cancer. These changes are potentially useful in identifying markers for early diagnosis and management of this lethal malignancy. Also, role of specific miRNAs and long non coding RNAs in regulation of gene expression is gaining interest and is a matter of further investigation. In this review, we aimed to summarize major epigenetic events (DNA methylation) in gastric cancer along with alteration in miRNAs and long non coding RNAs which plays an important role in pathology of this poorly understood malignancy. C1 [Puneet, S M] Banaras Hindu University, Institute of Medical Science, Department of Surgery, 221005 Varanasi, India. [Kazmi, Raza Hasan] Banaras Hindu University, Department of Molecular and Human Genetics, Institute of Science, 221005 Varanasi, India. [Kumari, Soni] Banaras Hindu University, Department of Molecular and Human Genetics, Institute of Science, 221005 Varanasi, India. [Tiwari, Satendra] Banaras Hindu University, Institute of Medical Science, Department of Surgery, 221005 Varanasi, India. [Khanna, Kumar Ajay] Banaras Hindu University, Institute of Medical Science, Department of Surgery, 221005 Varanasi, India. [Narayan, Gopeshwar] Banaras Hindu University, Department of Molecular and Human Genetics, Institute of Science, 221005 Varanasi, India. RP Puneet, SM (reprint author), Banaras Hindu University, Institute of Medical Science, Department of Surgery, 221005 Varanasi, India. EM puneetimsbhu@gmail.com CR Ushijima T, SasakoM(2004, Focus on gastric cancer. Cancer Cell 5(2):121–125 Tahara E, 2004, Genetic pathways of two types of gastric cancer. IARC Sci Publ 157:327–349 Smith MG, Hold GL, Tahara E, El-Omar EM, 2006, Cellular and molecular aspects of gastric cancer.World J Gastroenterol 12(19): 2979–2990 Park WS, Oh RR, Park JY, Lee SH, Shin MS, Kim YS, Kim SY, Lee HK, Kim PJ, Oh ST, Yoo NJ, Lee JY, 1999, Frequent somatic mutations of the beta-catenin gene in intestinal type gastric cancer. Cancer Res 59(17):4257–4260 Lee TL, LeungWK, ChanMW, Ng EK, Tong JH, Lo KW, Chung SC, Sung JJ, To KF, 2002, Detection of gene promoter hypermethylation in the tumor and serum of patients with gastric carcinoma. Clin Cancer Res 8(6):1761–1766 Maesawa C, Tamura G, Suzuki Y, Ogasawara S, Sakata K, Kashiwaba M, Satodate R, 1995, The sequential accumulation of genetic alterations characteristic of the colorectal adenoma carcinoma sequence does not occur between gastric adenoma and adenocarcinoma. J Pathol 176(3):249–258 Becker KF, Atkinson MJ, Reich U, Becker I, Nekarda H, Siewert JR, Hofler H, 1994, E-cadherin gene mutations provide clues to diffuse type gastric carcinomas. Cancer Res 54(14):3845–3852 Li QL, Ito K, Sakakura C, Fukamachi H, Ki I, Chi XZ et al, 2002, Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell 109(1):113–124 Hirst M, Marra MA, 2009, Epigenetics and human disease. Int J of Biochem Cell Bio 41(1):136–146 Panani AD, 2008, Cytogenetic and molecular aspects of gastric cancer: clinical implications. Cancer Letter 266(2):99–115 Mulero-Navarro S, Esteller M, 2008, Epigenetic biomarkers for human cancer: the time is now. Crit Rev Oncol Hematol 68(1):1– 11 Khan FA, Shukla AN(2006, Pathogenesis and treatment of gastric carcinoma: Ban up-date with brief review^. J Cancer Res Ther 2(4):196–199 Nitti D, Mocellin S, Marchet A, Pilati P, Lise M, 2008, Recent advances in conventional and molecular prognostic factors for gastric carcinoma. Surg Oncol Clin N Am 17(3):467–483 Fan CY, 2004, Epigenetic alterations in head and neck cancer: prevalence, clinical significance, and implications. Curr Oncol Rep 6(2):152–161 Tischoff I, Wittekind C, Tannapfel A, 2006, Role of epigenetic alterations in cholangiocarcinoma. J Hepato-Biliary-Pancreat Surg 13(4):274–279 Robertson KD, 2005, DNA methylation and human disease. Nat Rev Genet 6(8):597–610 Bird AP, 1986, CpG-rich islands and the function of DNA methylation. Nature 321(6067):209–213 Nakajima T, Enomoto S, Ushijima T, 2008, DNA methylation: a marker for carcinogen exposure and cancer risk. Environ Health Prev Med 13(1):8–15 Chen CZ, 2005, Micro-RNAs as oncogenes and tumour suppressors. N Engl J Med 353(17):1768–1771 Calin GA, Croce CM, 2006, Micro-RNA signatures in human cancers. Nat Rev Cancer 6(11):857–866 Hinshelwood RA, Clark SJ, 2008, Breast cancer epigenetics: normal human mammary epithelial cells as a model system. J Mol Med 86(12):1315–1328 Jones PA, Baylin SB, 2002, The fundamental role of epigenetic events in cancer. Nat Rev Genet 3(6):415–428 Karpf AR, Jones DA, 2002, Reactivating the expression of methylation silenced genes in human cancer. Oncogene 21(35):5450– 5461 Lund AH, van Lohuizen M, 2004, Polycomb complexes and silencing mechanisms. Curr Opin Cell Biol 16(3):239–246 Sparmann A, van Lohuizen M, 2006, Polycomb silencers control cell fate, development and cancer. Nat Rev Cancer 6(11):846–856 An C, Choi IS, Yao JC,Worah S, Xie K, Mansfield PF, Ajani JA, Rashid A, Hamilton SR,Wu TT, 2005, Prognostic significance of CpG island methylator phenotype and microsatellite instability in gastric carcinoma. Clin Cancer Res 11(2 Pt 1):656–663 Song SH, Jong HS, Choi HH, Kang SH, RyuMH, Kim NK, Kim WH, Bang YJ, 2000, Methylation of specific CpG sites in the promoter region could significantly down-regulate p16, INK4a, expression in gastric adenocarcinoma. Int J Cancer 87(2):236–240 Guimaraes AC, Lima EM, Khayat AS, Girao Faria MH, Barem Rabenhorst SH, Pitombeira MV, Assumpcao PP, de Oliveira Bahia M, Lima de Lima PD, de Arruda Cardoso Smith M, Burbano RR, 2007, Interrelationships among chromosome aneuploidy, promoter hypermethylation, and protein expression of the CDKN2A gene in individuals from northern Brazil with gastric adenocarcinoma. Cancer Genet Cytogenet 179(1):45–51 Lee JH, Park SJ, Abraham SC, Seo JS, Nam JH, Choi C, Juhng SW, Rashid A, Hamilton SR,Wu TT, 2004, Frequent CpG island methylation in precursor lesions and early gastric adenocarcinomas. Oncogene 23(26):4646–4654 Fleisher AS, Esteller M,Wang S, Tamura G, Suzuki H, Yin J, Zou TT, Abraham JM, Kong D, Smolinski KN, Shi YQ, Rhyu MG, Powell SM, James SP,Wilson KT, Herman JG,Meltzer SJ, 1999, Hypermethylation of the hMLH1 gene promoter in human gastric cancers with microsatellite instability. Cancer Res 59(5):1090– 1095 Leung SY, Yuen ST, Chung LP, Chu KM, Chan AS, Ho JC, 1999, hMLH1 promoter methylation and lack of hMLH1 expression in sporadic gastric carcinomas with high-frequency microsatellite instability. Cancer Res 59(1):159–164 Jung HY, Jung KC, Shim YH, Ro JY, Kang GH, 2001, Methylation of the hMLH1 promoter in multiple gastric carcinomas with microsatellite instability. Pathol Int 51(6):445–451 Oue N, Shigeishi H, Kuniyasu H, Yokozaki H, Kuraoka K, Ito R, Yasui W, 2001, Promoter hypermethylation of MGMT is associated with protein loss in gastric carcinoma. Int J Cancer 93(6):805– 809 Zazula M, Ferreira AM, Czopek JP, Kolodziejczyk P, Sinczak- Kuta A, Klimkowska A, Wojcik P, Okon K, Bialas M, Kulig J, Stachura J, 2006, CDH1 gene promoter hypermethylation in gastric cancer: relationship to Goseki grading, microsatellite instability status, and EBV invasion. Diagn Mol Pathol 15(1):24–29 Graziano F, Arduini F, RuzzoA, Bearzi I, Humar B,More H, Silva R, Muretto P, Guilford P, Testa E, Mari D, Magnani M, Cascinu S, 2004, Prognostic analysis of E-cadherin gene promoter hypermethylation in patients with surgically resected, node-positive, diffuse gastric cancer. Clin Cancer Res 10(8):2784–2789 Liu WT, Jiao HL, Yang YL, Wang D, Zhang WM, 2007, Correlation of E-cadherin hypermethylation to tumorigenesis and development of gastric cancer. Ai Zheng 26(11):1199–1203 Terres AM, Pajares JM, O'Toole D, Ahern S, Kelleher D, 1998, Helicobacter pylori infection is associated with downregulation of E-cadherin, a molecule involved in epithelial cell adhesion and proliferation control. J Clin Pathol 51(5):410–412 Chan AO, Lam SK,Wong BC,WongWM, YuenMF, Yeung YH, Hui WM, Rashid A, Kwong YL, 2003, Promoter methylation of E-cadherin gene in gastric mucosa associated with helicobacter pylori infection and in gastric cancer. Gut 52(4):502–506 Chan AO, 2006, E-cadherin in gastric cancer. World J Gastroenterol 12(2):199–203 Chan AW, Chan MW, Lee TL, Ng EK, Leung WK, Lau JY, Tong JH, Chan FK, To KF, 2005, Promoter hypermethylation of death associated protein-kinase gene associated with advance stage gastric cancer. Oncol Rep 13(5):937–941 Byun DS, Lee MG, Chae KS, Ryu BG, Chi SG, 2001, Frequent epigenetic inactivation of RASSF1A by aberrant promoter hypermethylation in human gastric adenocarcinoma. Cancer Res 61(19):7034–7038 Balgkouranidou I, Matthaios D, Karayiannakis A, Bolanaki H, Michailidis P, Xenidis N, Amarantidis K, Chelis L, Trypsianis G, Chatzaki E, Lianidou ES, Kakolyris S, 2015, Prognostic role of APC and RASSF1A promoter methylation status in cell free circulating DNA of operable gastric cancer patients. Mutat Res 778:46–51 Dote H, Toyooka S, Tsukuda K, Yano M, Ota T, Murakami M, Naito M, Toyota M, Gazdar AF, Shimizu N, 2005, Aberrant promoter methylation in human DAB2 interactive protein, hDAB2IP, gene in gastrointestinal tumour. Br J Cancer 92(6): 1117–1125 Qu Y, Dang S, Hou P, 2013, Genemethylation in gastric cancer. Clin Chim Acta 424:53–65. , DOI 10.1016/j.cca.2013.05.002 Byun DS, Cho K, Ryu BK, Lee MG, Kang MJ, Kim HR, Chi SG, 2003, Hypermethylation of XIAP associated factor 1, a putative tumor suppressor gene from the 17p13.2 locus, in human gastric adenocarcinomas. Cancer Res 63(21):7068–7075 Jee CD, Lee HS, Bae SI, Yang HK, Lee YM, Rho MS, Kim WH, 2005, Loss of caspase-1 gene expression in human gastric carcinomas and cell lines. Int J Oncol 26(5):1265–1271 Jung Y, Park J, Bang YJ, Kim TY, 2008, Gene silencing of TSPYL5 mediated by aberrant promoter methylation in gastric cancers. Lab Investig 88(2):153–160 Lee JH, Byun DS, Lee MG, Ryu BK, Kang MJ, Chae KS, Lee KY, Kim HJ, Park H, Chi SG, 2008, Frequent epigenetic inactivation of hSRBC in gastric cancer and its implication in attenuated p53 response to stresses. Int J Cancer 122(7):1573–1584 Cheng YY, Yu J,Wong YP, Man EP, To KF, Jin VX, Li J, Tao Q, Sung JJ, Chan FK, Leung WK, 2007, Frequent epigenetic inactivation of secreted frizzled-related protein 2, SFRP2, by promoter methylation in human gastric cancer. Br J Cancer 97(7):895–901 Sato H, Suzuki H, Toyota M, Nojima M, Maruyama R, Sasaki S, Takagi H, Sogabe Y, Sasaki Y, Idogawa M, Sonoda T, Mori M, Imai K, Tokino T, Shinomura Y, 2007, Frequent epigenetic inactivation of DICKKOPF family genes in human gastrointestinal tumours. Carcinogenesis 28(12):2459–2466 Nojima M, Suzuki H, Toyota M, Watanabe Y, Maruyama R, Sasaki S, Sasaki Y, Mita H, Nishikawa N, Yamaguchi K, Hirata K, Itoh F, Tokino T, Mori M, Imai K, Shinomura Y, 2007, Frequent epigenetic inactivation of SFRP genes and constitutive activation of Wnt signaling in gastric cancer. Oncogene 26(32): 4699–4713 Ebert MP, Yu J, Hoffmann J, Rocco A, Rocken C, Kahmann S, Muller O, Korc M, Sung JJ, Malfertheiner P, 2003, Loss of betacatenin expression in metastatic gastric cancer. J Clin Oncol 21(9): 1708–1714 Yu J, Cheng YY, Tao Q, Cheung KF, Lam CN, Geng H, Tian LW, Wong YP, Tong JH, Ying JM, Jin H, To KF, Chan FK, Sung JJ, 2009, Methylation of protocadherin 10, a novel tumor suppressor, is associated with poor prognosis in patients with gastric cancer. Gastroenterology 136(2):640–651 Takada H, Imoto I, Tsuda H, Nakanishi Y, Sakakura C, Mitsufuji S, Hirohashi S, Inazawa J, 2006, Genomic loss and epigenetic silencing of very-low-density lipoprotein receptor involved in gastric carcinogenesis. Oncogene 25(49):6554–6562 ChoiMC, Jong HS, Kim TY, Song SH, Lee DS, Lee JW, Kim TY, Kim NK, Bang YJ, 2004, ACAP12/Gravin is inactivated by epigeneticmechanism in human gastric carcinoma and shows growth suppressor activity. Oncogene 23(42):7095–7103 Oshimo Y, Oue N, Mitani Y, Nakayama H, Kitadai Y, Yoshida K, Chayama K, Yasui W, 2004, Frequent epigenetic inactivation of RIZ1 by promoter hypermethylation in human gastric carcinoma. Int J Cancer 110(2):212–218 Yamamichi N, Inada K, Ichinose M, Yamamichi-Nishina M, Mizutani T, Watanabe H, Shiogama K, Fujishiro M, Okazaki T, Yahagi N, Haraguchi T, Fujita S, Tsutsumi Y, Omata M, Iba H, 2007, Frequent loss of Brm expression in gastric cancer correlates with histologic features and differentiation state. Cancer Res 67(22):10727–10735 Hamai Y, Oue N, Mitani Y, Nakayama H, Ito R, Matsusaki K, Yoshida K, Toge T, Yasui W, 2003, DNA hypermethylation and histone hypoacetylation of the HLTF gene are associated with reduced expression in gastric carcinoma. Cancer Sci 94(8):692–698 Kim TY, Lee HJ,Hwang KS, Lee M, KimJW, Bang YJ, KangGH, 2004, Methylation of RUNX3 in various types of human cancers and premalignant stages of gastric carcinoma. Lab Investig 84(4): 479–484 Hayashi K, Yokozaki H, Goodison S, Oue N, Suzuki T, Lotan R, Yasui W, Tahara E, 2001, Inactivation of retinoic acid receptor beta by promoter CpG hypermethylation in gastric cancer. Differentiation 68(1):13–21 Oue N, Matsumura S, Nakayama H, Kitadai Y, Taniyama K, Matsusaki K, Yasui W, 2003, Reduced expression of the TSP1 gene and its association with promoter hypermethylation in gastric carcinoma. Oncology 64(4):423–439 Kim TY, Jong HS, Song SH, dimtchev A, Jeong SJ, Lee JW, Kim TY, Kim NK, Jung M, Bang YJ, 2003, Transcriptional silencing of the DLC-1 tumor suppressor gene by epigenetic mechanism in gastric cancer cells. Oncogene 22(25):3943–3951 de MaatMF, van deVelde CJ,Umetani N, de Heer P, PutterH, van Hoesel AQ,Meijer GA, van Grieken NC, Kuppen PJ, Bilchik AJ, Tollenaar RA, Hoon DS, 2007, Epigenetic silencing of cyclooxygenase-2 affects clinical outcome in gastric cancer. J Clin Oncol 25(31):4887–4894 Poplawski T, tomaszewska K, Galicki M, Morawiec Z, Blasiak J, 2008, Promoter methylation of cancer related genes in gastric carcinoma. Exp Oncol 30(2):112–116 Kang GH, Lee S, Kim JS, Jung HY, 2003, Profile of aberrant CpG island methylation along multistep gastric carcinogenesis. Lab Investig 83(4):519–526 Oue N, Motoshita J, Yokozaki H, Hayashi K, Tahara E, Taniyama K, Matsusaki K, Yasui W, 2002, Distinct promoter hypermethylation of p16INK4a, CDH1, and RAR-beta in intestinal, diffuseadherent, and diffuse-scattered type gastric carcinomas. J Pathol 198(1):55–59 Sepulveda JL, Gutierrez-Pajares JL, Luna A, Yao Y, Tobias JW, Thomas S, Woo Y, Giorgi F, Komissarova EV, Califano A,Wang TC, Sepulveda AR, 2016, High-definition CpG methylation of novel genes in gastric carcinogenesis identified by nextgeneration sequencing. Mod Pathol 29(2):182–193 Honda T, Tamura G, Waki T, Kawata S, Terashima M, Nishizuka S, Motoyama T, 2004, Demethylation of MAGE promoters during gastric cancer progression. Br J Cancer 90(4):838–843 Yanagawa N, Tamura G, Honda T, Endoh M, Nishizuka S, Motoyama T, 2004, Demethylation of the synuclein gamma gene CpG island in primary gastric cancers and gastric cancer cell lines. Clin Cancer Res 10(7):2447–2451 Lima EM, Leal MF, Burbano RR, Khayat AS, Assumpcao PP, Bello MJ, Rey JA, Smith MA, Casartelli C, 2008, Methylation status of ANAPC1, CDKN2A and TP53 promoter genes in individuals with gastric cancer. Braz J Med Biol Res 41(6):539–543 Heard E, Rougeulle C, Arnaud D, Avner P, Allis CD, Spector DL, 2001, Methylation of histone H3 at Lys-9 is an early mark on the X chromosome during X inactivation. Cell 107(6):727–738 Mermoud JE, Popova B, Peters AH, Jenuwein T, Brockdorff N, 2002, Histone H3 lysine 9 methylation occurs rapidly at the onset of random X chromosome inactivation. Curr Biol 12(3):247–251 Nguyen CT,Weisenberger DJ, VelicescuM, Gonzales FA, Lin JC, Liang G, Jones PA, 2002, Histone H3-lysine 9 methylation is associated with aberrant gene silencing in cancer cells and is rapidly reversed by 5-aza-2′-deoxycytidine. Cancer Res 62(22): 6456–6461 Schotta G, Lachner M, Sarma K, Ebert A, Sengupta R, Reuter G, Reinberg D, Jenuwein T, 2004, A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin. Genes Dev 18(11):1251–1262 Park YS, Jin MY, Kim YJ, Yook JH, Kim BS, Jang SJ, 2008, The global histone modification pattern correlates with cancer recurrence and overall survival in gastric adenocarcinoma. Ann Surg Oncol 15(7):1968–1976 Kondo Y, Shen L, Issa JP, 2003, Critical role of histone methylation in tumour suppressor gene silencing in colorectal cancer. Mol Cell Biol 23(1):206–215 Watanabe Y, Toyota M, Kondo Y, Suzuki H, Imai T, Ohe-Toyota M, Maruyama R, Nojima M, Sasaki Y, Sekido Y, Hiratsuka H, Shinomura Y, Imai K, Itoh F, Tokino T, 2007, PRDM5 identified as a target of epigenetic silencing in colorectal and gastric cancer. Clin Cancer Res 13(16):4786–4794 Ono S, Oue N, Kuniyasu H, Suzuki T, Ito R, Matsusaki K, Ishikawa T, Tahara E, Yasui W, 2002, Acetylated histone H4 is reduced in human gastric adenomas and carcinomas. J Exp Clin Cancer Res 21(3):377–382 Mitani Y, Oue N, Hamai Y, Aung PP, Matsumura S, Nakayama H, Kamata N, Yasui W, 2005, Histone H3 acetylation is associated with reduced p21(WAF1/CIP1, expression by gastric carcinoma. J Pathol 205(1):65–73 Xia G, Schneider-Stock R, Diestel A, Habold C, Krueger S, Roessner A, Naumann M, Lendeckel U, 2008, Helicobacter pylori regulates p21(WAF1, by histone H4 acetylation. Biochem Biophys Res Commun 369(2):526–531 Esteller M, 2011, Non-coding RNAs in human disease. Nat Rev Genet 12(12):861–874 Tekcham DS, Tiwari PK, 2016, Non-coding RNAs as emerging molecular targets in gallbladder cancer. Gene 588(1):79–85 Kang C, Song JJ, Lee J, KimMY, 2014, Epigenetics: an emerging player in gastric cancer. World J Gastroenterol 20(21):6433–6447 Suzuki H, Maruyama R, Yamamoto E, KaiM, 2012, DNA methylation and microRNA dysregulation in cancer. Mol Oncol 6(6): 567–578 Yin H, Song P, Su R, Yang G, Dong L, Luo M,Wang B, Gong B, Liu C, Song W, Wang F, Ma Y, Zhang J, Wang W, Yu J, 2016, DNA methylation mediated down-regulating of MicroRNA-33b and its role in gastric cancer. Sci Rep 6:18824. , DOI 10. 1038/srep18824 Li CL, Nie H,Wang M, Su LP, Li JF, Yu YY, YanM, Qu QL, Zhu ZG, Liu BY, 2012, microRNA-155 is downregulated in gastric cancer cells and involved in cell metastasis. Oncol Rep 27(6): 1960–1966 Li P, Chen X, Su L, Li C, Zhi Q, Yu B, Sheng H,Wang J, Feng R, Cai Q, Li J, Yu Y, Yan M, Liu B, Zhu Z, 2013, Epigenetic silencing of miR-338-3p contributes to tumorigenicity in gastric cancer by targeting SSX2IP. PLoS One 8(6):e66782. , DOI 10. 1371/journal.pone.0066782 Tsai KW, Liao YL, Wu CW, Hu LY, Li SC, Chan WC, Ho MR, Lai CH, Kao HW, FangWL, Huang KH, LinWC, 2011, Aberrant hypermethylation of miR-9 genes in gastric cancer. Epigenetics 6(10):1189–1197 Chen Q, Chen X, ZhangM, Fan Q, Luo S, Cao X, 2011)miR-137 is frequently down-regulated in gastric cancer and is a negative regulator of Cdc42. Dig Dis Sci 56(7):2009–2016 Deng H, Guo Y, Song H, Xiao B, SunW, Liu Z, Yu X, Xia T, Cui L, Guo J, 2013, MicroRNA-195 and microRNA-378 mediate tumor growth suppression by epigenetical regulation in gastric cancer. Gene 518(2):351–359 Wei B, SongY, Zhang Y, HuM(2013, microRNA-449a functions as a tumor-suppressor in gastric adenocarcinoma by targeting Bcl- 2. Oncol Lett 6(6):1713–1718 Ding L, Xu Y, ZhangW, DengY, SiM, Du Y, Yao H, Liu X, Ke Y, Si J, Zhou T, 2010, MiR-375 frequently downregulated in gastric cancer inhibits cell proliferation by targeting JAK2. Cell Res 20(7):784–793 Niinuma T, Suzuki H, Nojima M, Nosho K, Yamamoto H, Takamaru H, Yamamoto E, Maruyama R, Nobuoka T, Miyazaki Y, Nishida T, Bamba T, Kanda T, Ajioka Y, Taguchi T, Okahara S, Takahashi H, Nishida Y, Hosokawa M, Hasegawa T, Tokino T, Hirata K, Imai K, Toyota M, Shinomura Y, 2012, Upregulation of miR-196a and HOTAIR drive malignant character in gastrointestinal stromal tumors. Cancer Res 72(5):1126–1136 Wu H, Huang M, Lu M, Zhu W, Shu Y, Cao P, Liu P, 2013, Regulation of microtubule-associated protein tau, MAPT, by miR-34c-5p determines the chemosensitivity of gastric cancer to paclitaxel. Cancer Chemother Pharmacol 71(5):1159–1171 ZhuW, Xu H, Zhu D, Zhi H,Wang T,Wang J, Jiang B, Shu Y, Liu P, 2012, miR-200bc/429 cluster modulates multidrug resistance of human cancer cell lines by targeting BCL2 and XIAP. Cancer Chemother Pharmacol 69(3):723–731 Eddy SR, 2001, Non–coding RNA genes and the modern RNA world. Nat Rev Genet 2(12):919–929 S D, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A et al, 2012, Landscape of transcription in human cells. Nature 489(7414):101–108 Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, Maeda N, Oyama R, Ravasi T, Lenhard B, Wells C, Kodzius R, Shimokawa K, Bajic VB, Brenner SE, Batalov S, Forrest AR, Zavolan M, Davis MJ, Wilming LG, Aidinis V, Allen JE, Ambesi-Impiombato A, Apweiler R, Aturaliya RN, Bailey TL, Bansal M, Baxter L, Beisel KW, Bersano T, Bono H, Chalk AM, Chiu KP, Choudhary V, Christoffels A, Clutterbuck DR, Crowe ML, Dalla E, Dalrymple BP, de Bono B, Della Gatta G, di Bernardo D, Down T, Engstrom P, Fagiolini M, Faulkner G, Fletcher CF, Fukushima T, Furuno M, Futaki S, Gariboldi M, Georgii-Hemming P, Gingeras TR, Gojobori T, Green RE, Gustincich S, Harbers M, Hayashi Y, Hensch TK, Hirokawa N, Hill D, Huminiecki L, Iacono M, Ikeo K, Iwama A, Ishikawa T, JaktM, Kanapin A, Katoh M, Kawasawa Y, Kelso J, Kitamura H, Kitano H, Kollias G, Krishnan SP, Kruger A, Kummerfeld SK, Kurochkin IV, Lareau LF, Lazarevic D, Lipovich L, Liu J, Liuni S, McWilliam S, Madan Babu M, Madera M, Marchionni L, Matsuda H, Matsuzawa S, Miki H, Mignone F, Miyake S, Morris K, Mottagui-Tabar S, Mulder N, Nakano N, Nakauchi H, Ng P, Nilsson R, Nishiguchi S, Nishikawa S, Nori F, Ohara O, Okazaki Y, Orlando V, Pang KC, Pavan WJ, Pavesi G, Pesole G, Petrovsky N, Piazza S, Reed J, Reid JF, Ring BZ, Ringwald M, Rost B, Ruan Y, Salzberg SL, Sandelin A, Schneider C, Schonbach C, Sekiguchi K, Semple CA, Seno S, Sessa L, Sheng Y, ShibataY, Shimada H, ShimadaK, Silva D, Sinclair B, Sperling S, Stupka E, Sugiura K, Sultana R, Takenaka Y, Taki K, Tammoja K, Tan SL, Tang S, Taylor MS, Tegner J, Teichmann SA, Ueda HR, van Nimwegen E, Verardo R,Wei CL, Yagi K, Yamanishi H, Zabarovsky E, Zhu S, Zimmer A, HideW, Bult C, GrimmondSM, Teasdale RD, Liu ET, Brusic V, Quackenbush J, Wahlestedt C, Mattick JS, Hume DA, Kai C, Sasaki D, Tomaru Y, Fukuda S, Kanamori-Katayama M, Suzuki M, Aoki J, Arakawa T, Iida J, Imamura K, Itoh M, Kato T, Kawaji H, Kawagashira N, Kawashima T, Kojima M, Kondo S, Konno H, Nakano K, Ninomiya N, Nishio T, Okada M, Plessy C, Shibata K, Shiraki T, Suzuki S, Tagami M, Waki K, Watahiki A, Okamura-Oho Y, Suzuki H, Kawai J, Hayashizaki Y, FANTOM Consortium, RIKEN Genome Exploration Research Group and Genome Science Group, Genome Network Project Core Group,, 2005, The transcriptional landscape of the mammalian genome. Science 309(5740):1559–1563 Yang Q, Zhang RW, Sui PC, He HT, Ding L, 2015, Dysregulation of non-coding RNAs in gastric cancer. World J Gastroenterol 21(39):10956–10981 Arita T, IchikawaD, Konishi H, Komatsu S, Shiozaki A, Shoda K, Kawaguchi T, Hirajima S, Nagata H, Kubota T, Fujiwara H, Okamoto K, Otsuji E, 2013, Circulating long non-coding RNAs in plasma of patients with gastric cancer. Anticancer Res 33(8): 3185–3193 Zhou X,Yin C, Dang Y,Ye F, ZhangG(2015, Identification of the long non-coding RNA H19 in plasma as a novel biomarker for diagnosis of gastric cancer. Sci Rep 5:11516. , DOI 10. 1038/srep11516 Endo H, Shiroki T, Nakagawa T, Yokoyama M, Tamai K, Yamanami H, Fujiya T, Sato I, Yamaguchi K, Tanaka N, Iijima K, Shimosegawa T, Sugamura K, Satoh K, 2013, Enhanced expression of long non-coding RNA HOTAIR is associated with the development of gastric cancer. PLoS One 8(10):e77070. https:// doi.org/10.1371/journal.pone.0077070.eCollection2013 Zhao Y, Guo Q, Chen J, Hu J,Wang S, Sun Y, 2014, Role of long non-coding RNA HULC in cell proliferation, apoptosis and tumor metastasis of gastric cancer: a clinical and in vitro investigation. Oncol Rep 31(1):358–364 Sun M, Xia R, Jin F, Xu T, Liu Z, De W, Liu X, 2014, Downregulated long noncoding RNA MEG3 is associated with poor prognosis and promotes cell proliferation in gastric cancer. Tumour Biol 35(2):1065–1073 Cao WJ, Wu HL, He BS, Zhang YS, Zhang ZY, 2013, Analysis of long non-coding RNA expression profiles in gastric cancer. World J Gastroenterol 19(23):3658–3664 Li CY, Liang GY, Yao WZ, Sui J, Shen X, Zhang YQ, Peng H, HongWW, Ye YC, Zhang ZY, ZhangWH, Yin LH, Pu YP, 2016, Integrated analysis of long non-coding RNA competing interactions reveals the potential role in progression of human gastric cancer. Int J Oncol 48(5):1965–1976 Rich S, Ganz R, Levy PS, 1983, Comparative actions of hydralazine, nifedipine and amrinone in primary pulmonary hypertension. Am J Cardiol 52(8):1104–1107 Strolin Benedetti M, Rumigny JF, Dostert P, 1984, Mechanisms of action and biochemical toxicology of valproic acid. Encephale 10(4):177–188 Windle J, Prystowsky EN, Miles WM, Heger JJ, 1987, Pharmacokinetic and electrophysiologic interactions of amiodarone and procainamide. Clin Pharmacol Ther 41(6):603–610 Issa JP, Gharibyan V, Cortes J, Jelinek J, Morris G, Verstovsek S, Talpaz M, Garcia-Manero G, Kantarjian HM, 2005, Phase II study of low-dose decitabine in patients with chronic myelogenous leukemia resistant to imatinib mesylate. J Clin Oncol 23(17):3948–3956 Kaminskas Kaminskas E, Farrell A, Abraham S, Baird A, Hsieh LS, Lee SL, Leighton JK, Patel H, Rahman A, Sridhara R, Wang YC, Pazdur R, 2005, Approval summary: azacitidine for treatment of myelodysplastic syndrome subtypes. Clin Cancer Res 11(10): 3604–3608 Zambrano P, Segura-Pacheco B, Perez-Cardenas E, Cetina L, Revilla-Vazquez A, Taja-Chayeb L, Chavez-Blanco A, Angeles E, Cabrera G, Sandoval K, Trejo-Becerril C, Chanona-Vilchis J, Duenas-Gonzalez A, 2005, A phase I study of hydralazine to demethylate and reactivate the expression of tumor suppressor genes. BMC Cancer 5:44 Zhang X, Yashiro M, Ren J, Hirakawa K, 2006, Histone deacetylase inhibitor, trichostatin A, increases the chemosensitivity of anticancer drugs in gastric cancer cell lines. Oncol Rep 16(3):563–568 Riester D, Hildmann C, Schwienhorst A, 2007, Histone deacetylase inhibitors–turning epigenic mechanisms of gene regulation into tools of therapeutic intervention in malignant and other diseases. Appl Microbio Biotechnol 75(3):499–514 Satoh A, Toyota M, Itoh F, Sasaki Y, Suzuki H, Ogi K, Kikuchi T, Mita H, Yamashita T, Kojima T, Kusano M, Fujita M, Hosokawa M, Endo T, Tokino T, Imai K, 2003, Epigenetic inactivation of CHFR and sensitivity to microtubule inhibitors in gastric cancer. Cancer Res 63(24):8606–8613 Koga Y, Kitajima Y, Miyoshi A, Sato K, Sato S, Miyazaki K, 2006, The significance of aberrant CHFR methylation for clinical response to microtubule inhibitors in gastric cancer. J Gastroenterol 41(2):133–139 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 757 EP 770 DI 10.1007/s12253-018-0410-z PG 14 ER PT J AU Paskal, W Paskal, MA Debski, T Gryziak, M Jaworowski, J AF Paskal, Wiktor Paskal, M Adriana Debski, Tomasz Gryziak, Maciej Jaworowski, Janusz TI Aspects of Modern Biobank Activity – Comprehensive Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Biobank; Personalized medicine; Biorepository; Biospecimen; Tissue Banking ID Biobank; Personalized medicine; Biorepository; Biospecimen; Tissue Banking AB Biobanks play an increasing role in contemporary research projects. These units meet all requirements to regard them as a one of the most innovative and up-to-date in the field of biomedical research. They enable conducting wide-scale research by the professional collection of biological specimens and correlated clinical data. Pathology units may be perceived roots of biobanking. The review aims at describing the concept of biobanks, their model of function and scientific potential. It comprises the division of biobanks, sample preservation methods and IT solutions as well as guidelines and recommendations for management of a vast number of biological samples and clinical data. Therefore, appropriate standard operating procedures and protocols are outlined. Constant individualization of diagnostic process and treatment procedures creates the niche for translational units. Thus, the role of biobanks in personalized medicine was also specified. The exceptionality of biobanks poses some new ethical-legal issues which have various solutions, in each legal system, amongst the world. Finally, distribution and activity of European biobanks are mentioned. C1 [Paskal, Wiktor] Medical University of Warsaw, The Department of Histology and Embryology, ul. Banacha 1B, 02-097 Warsaw, Poland. [Paskal, M Adriana] Medical University of Warsaw, The Department of Histology and Embryology, ul. Banacha 1B, 02-097 Warsaw, Poland. [Debski, Tomasz] Centre of Postgraduate Medical Education, Plastic Surgery DepartmentWarsaw, Poland. [Gryziak, Maciej] Medical University of Warsaw, The Department of Applied PharmacyWarsaw, Poland. [Jaworowski, Janusz] Medical University of Warsaw, The Department of Applied PharmacyWarsaw, Poland. RP Paskal, W (reprint author), Medical University of Warsaw, The Department of Histology and Embryology, 02-097 Warsaw, Poland. EM wiktor.paskal@wum.edu.pl CR Fransson MN, Rial-Sebbag E, Brochhausen M, Litton JE, 2015, Toward a common language for biobanking. Eur J Hum Genet 23. , DOI 10.1038/ejhg.2014.45 Kang B, Park J, Cho S, LeeM, Kim N,Min H et al, 2013, Current status, challenges, policies, and bioethics of biobanks. Genomics inform 11(4):211–217. , DOI 10.5808/GI.2013.11.4.211 Sak J, Pawlikowski J, Goniewicz M, Witt M, 2012, Population biobanking in selected European countries and proposed model for a Polish national DNA bank. J Appl Genet 53(2):159–165. , DOI 10.1007/s13353-012-0082-4 Riegman PH, Morente MM, Betsou F, de Blasio P, Geary P, Marble Arch International Working Group on Biobanking for Biomedical R, 2008, Biobanking for better healthcare. Mol Oncol 2(3):213–222. , DOI 10.1016/j.molonc.2008.07. 004 Goebell PJ, Morente MM, 2010, New concepts of biobanks– strategic chance for uro-oncology. Urol Oncol 28(4):449–457. , DOI 10.1016/j.urolonc.2010.03.012 Asslaber M, Zatloukal K, 2007, Biobanks: transnational, European and global networks. Brief Funct Genom Proteomics 6(3):193–201. , DOI 10.1093/bfgp/elm023 Swede H, Stone CL, Norwood AR, 2007, National populationbased biobanks for genetic research. Genet Med 9(3):141–149. , DOI 10.1097/GIM.0b013e3180330039 Leitsalu L, Haller T, Esko T, Tammesoo ML, Alavere H, Snieder H et al, 2014, Cohort Profile: Estonian Biobank of the Estonian Genome Center, University of Tartu. Int J Epidemiol. https://doi. org/10.1093/ije/dyt268 Zawati M, Borry P, Howard H, 2011, Closure of population biobanks and direct-to-consumer genetic testing companies. Hum Genet 130(3):425–432. , DOI 10.1007/s00439- 011-1019-4 Awadalla P, Boileau C, Payette Y, Idaghdour Y, Goulet JP, Knoppers B et al., 2013, Cohort profile of the CARTaGENE study: Quebec's population-based biobank for public health and personalized genomics. Int J Epidemiol 42, 5):1285–1299. https:// doi.org/10.1093/ije/dys160 Andersson K, Bray F, Arbyn M, Storm H, Zanetti R, Hallmans G et al, 2010, The interface of population-based cancer registries and biobanks in etiological and clinical research–current and future perspectives. Acta Oncol 49(8):1227–1234. , DOI 10. 3109/0284186X.2010.496792 Fannin M, Kent J, 2015, Origin stories from a regional placenta tissue collection. New Genet Soc 34(1):25–51. , DOI 10. 1080/14636778.2014.999153 Bonnelykke K, Sleiman P, Nielsen K, Kreiner-Moller E, Mercader JM, Belgrave D et al, 2014, A genome-wide association study identifies CDHR3 as a susceptibility locus for early childhood asthma with severe exacerbations. Nat Genet 46(1):51–55. , DOI 10.1038/ng.2830 Haukkala A, Kujala E, Alha P, Salomaa V, Koskinen S, Swan H et al, 2013, The return of unexpected research results in a biobank study and referral to health care for heritable long QT syndrome. Public Health Genomics 16(5):241–250. , DOI 10.1159/ 000354105 Wood AR, Esko T, Yang J, Vedantam S, Pers TH, Gustafsson S et al, 2014, Defining the role of common variation in the genomic and biological architecture of adult human height. Nat Genet 46(11):1173–1186. , DOI 10.1038/ng.3097 http://www. nature.com/ng/journal/v46/n11/abs/ng.3097.html# supplementary-information Shui IM, Lindstrom S, Kibel AS, Berndt SI, Campa D, Gerke T et al, 2014, Prostate Cancer, PCa, Risk Variants and Risk of Fatal PCa in the National Cancer Institute Breast and Prostate Cancer Cohort Consortium. Eur Urol 65(6):1069–1075. , DOI 10.1016/j.eururo.2013.12.058 Nieuwenhuijsen MJ, Smith R, Golfinopoulos S, Best N, Bennett J, Aggazzotti G et al., 2009, Health impacts of long-term exposure to disinfection by-products in drinking water in Europe: HIWATE. J Water Health 7, 2):185–18207. , DOI 10.2166/wh.2009. 073 Jeong CH, Wagner ED, Siebert VR, Anduri S, Richardson SD, Daiber EJ et al, 2012, Occurrence and toxicity of disinfection byproducts in European drinking waters in relation with the HIWATE epidemiology study. Environ Sci Technol 46(21): 12120–12128. , DOI 10.1021/es3024226 McKenzie F, Ferrari P, Freisling H, Chajes V, Rinaldi S, de Batlle J et al, 2014, Healthy lifestyle and risk of breast cancer among postmenopausal women in the European Prospective Investigation into Cancer and Nutrition cohort study. Int J Cancer. , DOI 10.1002/ijc.29315 Engeset D, Braaten T, Teucher B, Kuhn T, Bueno-de-Mesquita HB, Leenders M et al, 2014, Fish consumption and mortality in the European Prospective Investigation into Cancer and Nutrition cohort. Eur J Epidemiol. , DOI 10.1007/s10654-014- 9966-4 Ose J, Fortner RT, Schock H, Peeters PH, Onland-Moret NC, Bueno-de-Mesquita HB et al, 2014, Insulin-like growth factor I and risk of epithelial invasive ovarian cancer by tumour characteristics: results from the EPIC cohort. Br J Cancer. , DOI 10.1038/bjc.2014.566 Reza M, Cox D, Phillips L, Johnson D, Manoharan V, Grieves M et al, 2017, MRC centre neuromuscular biobank, newcastle and london): supporting and facilitating rare and neuromuscular disease research worldwide. Neuromuscul Disord. , DOI 10. 1016/j.nmd.2017.07.001 De Souza YG, Greenspan JS, 2013, Biobanking past, present and future: responsibilities and benefits. AIDS, London, England, 27(3):303–312. , DOI 10.1097/QAD.0b013e32835c1244 Check Hayden E, 2015, Proposed Ebola biobank would strengthen African science. Nature 524(7564):146–147. , DOI 10. 1038/524146a Hutchinson E, 2014, Towards individualized cancer therapy: Challenges and prospects. Mol Oncol 8(1):1–8. , DOI 10.1016/j.molonc.2013.12.008 Guerin JS,MurrayDW, McGrathMM, YuilleMA,McPartlin JM, Doran PP, 2010, Molecular medicine ireland guidelines for standardized biobanking. Biopreserv Biobank 8(1):3–63. https://doi. org/10.1089/bio.2010.8101 Mohamadkhani A, Poustchi H, 2015, Repository of Human Blood Derivative Biospecimens in Biobank: Technical Implications. Middle East J Dig Dis 7(2):61–68 Shabihkhani M, Lucey GM,Wei B,Mareninov S, Lou JJ, Vinters HVet al, 2014, The procurement, storage, and quality assurance of frozen blood and tissue biospecimens in pathology, biorepository, and biobank settings. Clin Biochem 47(4–5):258– 266. , DOI 10.1016/j.clinbiochem.2014.01.002 Duale N, Lipkin WI, Briese T, Aarem J, Ronningen KS, Aas KK et al, 2014, Long-term storage of blood RNA collected in RNA stabilizing Tempus tubes in a large biobank–evaluation of RNA quality and stability. BMC Res Notes 7:633. , DOI 10. 1186/1756-0500-7-633 King MJ, Garcon L, Hoyer JD, Iolascon A, Picard V, Stewart G et al, 2015, ICSH guidelines for the laboratory diagnosis of nonimmune hereditary red cell membrane disorders. Int J Lab Hematol 37(3):304–325. , DOI 10.1111/ijlh.12335 Maimuna Mendy EC, Lawlor RT, Wright J, Wild CP, 2017, Common minimum technical standards and protocols for biobanks dedicated to cancer research. Int Agency Res Cancer 32. Higdon LE, Lee K, Tang Q, Maltzman JS, 2016, Virtual Global Transplant Laboratory Standard Operating Procedures for Blood Collection, PBMC Isolation, and Storage. Transplantat Direct 2(9):e101. , DOI 10.1097/txd.0000000000000613 33. Qin J, Alt JR, Hunsley BA, Williams TL, Fernando MR, 2014, Stabilization of circulating tumor cells in blood using a collection device with a preservative reagent. Cancer Cell Int 14(1):23. , DOI 10.1186/1475-2867-14-23 34. Riethdorf S, Fritsche H, Muller V, Rau T, Schindlbeck C, Rack B et al, 2007, Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system. Clin Cancer Res 13(3):920–928. https:// doi.org/10.1158/1078-0432.ccr-06-1695 35. Oxnard GR, Paweletz CP, Sholl LM, 2017, Genomic Analysis of Plasma Cell-Free DNA in Patients With Cancer. JAMA Oncol 3(6):740–741. , DOI 10.1001/jamaoncol.2016.2835 36. Ignatiadis M, Dawson SJ, 2014, Circulating tumor cells and circulating tumor DNA for precision medicine: dream or reality? Ann Oncol 25(12):2304–2313. , DOI 10.1093/annonc/ mdu480 37. Devonshire AS, Whale AS, Gutteridge A, Jones G, Cowen S, Foy CA et al, 2014, Towards standardisation of cell-free DNA measurement in plasma: controls for extraction efficiency, fragment size bias and quantification. Anal Bioanal Chem 406(26):6499– 6512. , DOI 10.1007/s00216-014-7835-3 38. Wong D, Moturi S, Angkachatchai V,Mueller R, DeSantis G, van den Boom D et al, 2013, Optimizing blood collection, transport and storage conditions for cell free DNA increases access to prenatal testing. Clin Biochem 46(12):1099–1104. , DOI 10. 1016/j.clinbiochem.2013.04.023 39. Hidestrand M, Stokowski R, Song K, Oliphant A, Deavers J, Goetsch M et al, 2012, Influence of temperature during transportation on cell-free DNA analysis. Fetal Diagn Ther 31(2):122– 128. , DOI 10.1159/000335020 40. CallariM, Tiberio P, De Cecco L, Cavadini E, Dugo M, Ghimenti C et al, 2013, Feasibility of circulating miRNA microarray analysis from archival plasma samples. Anal Biochem 437(2):123– 125. , DOI 10.1016/j.ab.2013.03.002 41. Spornraft M, Kirchner B, Haase B, Benes V, Pfaffl MW, Riedmaier I, 2014, Optimization of extraction of circulating RNAs from plasma–enabling small RNA sequencing. PLoS One 9(9):e107259. , DOI 10.1371/journal.pone.0107259 42. Sourvinou IS, Markou A, Lianidou ES, 2013, Quantification of circulating miRNAs in plasma: effect of preanalytical and analytical parameters on their isolation and stability. J Mol Diagn: JMD 15(6):827–834. , DOI 10.1016/j.jmoldx.2013.07.005 43. Baranyai T, Herczeg K, Onodi Z, Voszka I, Modos K, Marton N et al, 2015, Isolation of Exosomes fromBlood Plasma: Qualitative and Quantitative Comparison of Ultracentrifugation and Size Exclusion Chromatography Methods. PLoS One 10(12): e0145686. , DOI 10.1371/journal.pone.0145686 44. Kim JW, Wieckowski E, Taylor DD, Reichert TE, Watkins S, Whiteside TL, 2005, Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes. Clin Cancer Res 11(3):1010–1020 45. BestMG, Sol N, Kooi I, Tannous J,Westerman BA, Rustenburg F et al, 2015, RNA-Seq of Tumor-Educated Platelets Enables Blood-Based Pan-Cancer, Multiclass, and Molecular Pathway Cancer Diagnostics. Cancer Cell 28(5):666–676. , DOI 10.1016/j.ccell.2015.09.018 46. Aibaidula A, Lu JF, Wu JS, Zou HJ, Chen H, Wang YQ et al, 2015, Establishment and maintenance of a standardized glioma tissue bank: Huashan experience. Cell Tissue Bank 16(2):271– 281. , DOI 10.1007/s10561-014-9459-4 47. Wang Z, Lebron JA, Wolf JJ, 2015, Reliable quantification of mRNA in archived formalin-fixed tissue with or without paraffin embedding. J Pharmacol Toxicol Methods 71:103–109. https:// doi.org/10.1016/j.vascn.2014.09.007 48. Schweiger MR, Kerick M, Timmermann B, Albrecht MW, Borodina T, Parkhomchuk D et al, 2009, Genome-wide massively parallel sequencing of formaldehyde fixed-paraffin embedded, FFPE, tumor tissues for copy-number- and mutation-analysis. PLoS One 4(5):e5548. , DOI 10.1371/journal.pone. 0005548 49. Hedegaard J, Thorsen K, Lund MK, Hein AM, Hamilton-Dutoit SJ, Vang S et al, 2014, Next-generation sequencing of RNA and DNA isolated from paired fresh-frozen and formalin-fixed paraffin- embedded samples of human cancer and normal tissue. PLoS One 9(5):e98187. , DOI 10.1371/journal.pone.0098187 50. Florell SR, Coffin CM, Holden JA, Zimmermann JW, Gerwels JW, Summers BK et al, 2001, Preservation of RNA for functional genomic studies: amultidisciplinary tumor bank protocol.Modern pathology : an official journal of the United States and Canadian Academy of Pathology. Inc 14(2):116–128. , DOI 10. 1038/modpathol.3880267 51. Langdon SP, 2004, Cancer Cell Culture. Methods in Molecular Medicine, vol 88. Humana Press. , DOI 10.1385/ 1592594069 52. Mitra A,Mishra L, Li S, 2013, Technologies for deriving primary tumor cells for use in personalized cancer therapy. Trends Biotechnol 31(6):347–354. , DOI 10.1016/j.tibtech. 2013.03.006 53. Giannini C, Sarkaria JN, Saito A, Uhm JH, Galanis E, Carlson BL et al, 2005, Patient tumor EGFR and PDGFRA gene amplifications retained in an invasive intracranial xenograft model of glioblastoma multiforme. Neuro-Oncology 7(2):164–176. https://doi. org/10.1215/s1152851704000821 54. Perskvist N, Norman I, Eklund C, Litton JE, Dillner J, 2013, The Swedish cervical cytology biobank: sample handling and storage process. Biopreserv Biobank 11(1):19–24. , DOI 10. 1089/bio.2012.0036 55. Elliott P, Peakman TC, 2008, The UK Biobank sample handling and storage protocol for the collection, processing and archiving of human blood and urine. Int J Epidemiol 37(2):234–244. https:// doi.org/10.1093/ije/dym276 56. Witon M, Strapagiel D, Glenska-Olender J, Chroscicka A, Ferdyn K, Skokowski J et al, 2017, Organization of BBMRI.pl: The Polish Biobanking Network. Biopreserv Biobank 15(3):264– 269. , DOI 10.1089/bio.2016.0091 57. Vested A, Ramlau-Hansen CH, Olsen SF, Bonde JP, Kristensen SL, Halldorsson TI et al, 2013, Associations of in utero exposure to perfluorinated alkyl acids with human semen quality and reproductive hormones in adult men. Environ Health Perspect 121(4): 453–458. , DOI 10.1289/ehp.1205118 58. Lee JE, Kim JH, Hong EJ, Yoo HS, Nam HY, Park O, 2012, National Biobank of Korea: Quality control Programs of Collected-human Biospecimens. Osong Public Health Res Perspect 3(3):185–189. , DOI 10.1016/j.phrp.2012.07.007 59. Mathay C, Hamot G, Henry E, Georges L, Bellora C, Lebrun L et al, 2015, Method optimization for fecal sample collection and fecal DNA extraction. Biopreserv Biobank 13(2):79–93. https:// doi.org/10.1089/bio.2014.0031 60. Podzimek S, Vondrackova L, Duskova J, Janatova T, Broukal Z, 2016, Salivary Markers for Periodontal and General Diseases. Dis Markers 2016:9179632. , DOI 10.1155/2016/ 9179632 61. Eglash A, 2010, ABM clinical protocol #8: human milk storage information for home use for full-term infants, original protocol March 2004; revision #1March 2010). Breastfeed Med 5(3):127– 130. , DOI 10.1089/bfm.2010.9988 62. 2012 best practices for repositories collection, storage, retrieval, and distribution of biological materials for research international society for biological and environmental repositories, 2012, Biopreserv Biobank 10(2):79–161. , DOI 10.1089/bio. 2012.1022 63. He K, 2011, Trace elements in nails as biomarkers in clinical research. Eur J Clin Investig 41(1):98–102. , DOI 10. 1111/j.1365-2362.2010.02373.x 64. Liu LY, He K, Hites RA, Salamova A, 2016, Hair and Nails as Noninvasive Biomarkers of Human Exposure to Brominated and Organophosphate Flame Retardants. Environ Sci Technol 50(6): 3065–3073. , DOI 10.1021/acs.est.5b05073 65. Willemse EA, Koel-Simmelink MJ, Durieux-Lu S, van der Flier WM, Teunissen CE, 2015, Standard biobanking conditions prevent evaporation of body fluid samples. Clin Chim Acta 442:141– 145. , DOI 10.1016/j.cca.2015.01.036 66. Kofanova OA, Mommaerts K, Betsou F, 2015, Tube Polypropylene: A Neglected Critical Parameter for Protein Adsorption During Biospecimen Storage. Biopreserv Biobank 13(4):296–298. , DOI 10.1089/bio.2014.0082 67. Malm J, Lindberg H, Erlinge D, Appelqvist R, Yakovleva M, Welinder C et al, 2015, Semi-automated biobank sample processing with a 384 high density sample tube robot used in cancer and cardiovascular studies. Clin Transl Med 4(1):67. , DOI 10.1186/s40169-015-0067-0 68. Nussbeck SY, Skrowny D, O'Donoghue S, Schulze TG, Helbing K, 2014, How to Design Biospecimen Identifiers and Integrate Relevant Functionalities into Your Biospecimen Management System. Biopreserv Biobank 12(3):199–205. , DOI 10. 1089/bio.2013.0085 69. Riondino S, 2015, Sample PReanalytical Code for labeling of biospecimens: an analysis of specimen labeling protocols. J Biorepository Sci Appl Med 3:15–21 70. Artene SA, Ciurea ME, Purcaru SO, Tache DE, Tataranu LG, Lupu M et al, 2013, Biobanking in a constantly developing medical world. TheScientificWorldJOURNAL 2013:343275. https:// doi.org/10.1155/2013/343275 71. Betsou F, Lehmann S, Ashton G, Barnes M, Benson EE, Coppola D et al, 2010, Standard preanalytical coding for biospecimens: defining the sample PREanalytical code. Cancer Epidemiol Biomark Prev 19(4):1004–1011. , DOI 10.1158/1055- 9965.EPI-09-1268 72. Moore HM, Kelly AB, Jewell SD, McShane LM, Clark DP, Greenspan R et al, 2011, Biospecimen reporting for improved study quality, BRISQ). Cancer Cytopathol 119(2):92–101. , DOI 10.1002/cncy.20147 73. Merino-Martinez R, Norlin L, van Enckevort D, Anton G, Schuffenhauer S, Silander K et al, 2016, Toward Global Biobank Integration by Implementation of the Minimum Information About BIobank Data Sharing, MIABIS 2.0 Core). Biopreserv Biobank 14(4):298–306. , DOI 10.1089/bio. 2015.0070 74. Zika E, Paci D, Braun A, Rijkers-Defrasne S, Deschenes M, Fortier I et al, 2011, A European survey on biobanks: trends and issues. Public Health Genomics 14(2):96–103. , DOI 10. 1159/000296278 75. Izzo M, Mortola F, Arnulfo G, Fato MM, Varesio L, 2014, A digital repository with an extensible data model for biobanking and genomic analysis management. BMC Genomics 15(Suppl 3):S3. , DOI 10.1186/1471-2164-15-s3-s3 76. Sariyar M, Schluender I, Smee C, Suhr S, 2015, Sharing and Reuse of Sensitive Data and Samples: Supporting Researchers in Identifying Ethical and Legal Requirements. Biopreserv Biobank 13(4):263–270. , DOI 10.1089/bio.2015.0014 77. Tebbakha R, 2013, Biobank–Short Message Service for Linking Patients and Samples. Telemed e-Health 19(9):717–721. https:// doi.org/10.1089/tmj.2012.0231 78. Ravid R, 2008, Standard Operating Procedures, ethical and legal regulations in BTB, Brain/Tissue/Bio, banking: what is still missing? Cell Tissue Bank 9(2):121–137. , DOI 10.1007/ s10561-007-9055-y 79. Steinsbekk KS, Kare Myskja B, Solberg B, 2013, Broad consent versus dynamic consent in biobank research: is passive participation an ethical problem? Eur J Hum Gen: EJHG 21(9):897–902. , DOI 10.1038/ejhg.2012.282 80. Colledge F, Persson K, Elger B, Shaw D, 2014, Sample and data sharing barriers in biobanking: consent, committees, and compromises. Ann Diagn Pathol 18(2):78–81. , DOI 10.1016/j. anndiagpath.2013.12.002 81. Wolf SM, Lawrenz FP, Nelson CA, Kahn JP, Cho MK, Clayton EWet al, 2008, Managing incidental findings in human subjects research: analysis and recommendations. J Law Med Ethics 36(2): 219–248, 211. , DOI 10.1111/j.1748-720X.2008.00266.x 82. Wolf SM, Crock BN, Van Ness B, Lawrenz F, Kahn JP, Beskow LM et al, 2012, Managing Incidental Findings and Research Results in Genomic Research Involving Biobanks & Archived Datasets. Genet Med 14(4):361–384. , DOI 10.1038/ gim.2012.23 83. Fleming J, Critchley C, OtlowskiM, Stewart C, Kerridge I, 2015, Attitudes of the general public towards the disclosure of individual research results and incidental findings from biobank genomic research in Australia. Intern Med J 45(12):1274–1279. https:// doi.org/10.1111/imj.12911 84. Giesbertz NA, Bredenoord AL, van Delden JJ, 2015, Consent procedures in pediatric biobanks. Eur J Hum Gen: EJHG 23, 9): 1129–1134. , DOI 10.1038/ejhg.2014.267 85. Rush A, Battisti R, Barton B, Catchpoole D, 2015, Opinions of Young Adults on Re-Consenting for Biobanking. J Pediatr 167(4): 925–930. , DOI 10.1016/j.jpeds.2015.07.005 86. Virani AH, Longstaff H, 2014, Ethical Considerations in Biobanks: How a Public Health Ethics Perspective Sheds New Light on Old Controversies. J Genet Couns. , DOI 10. 1007/s10897-014-9781-9 87. Cadigan RJ, Lassiter D, Haldeman K, Conlon I, Reavely E, Henderson GE, 2013, Neglected ethical issues in biobank management: Results from a U.S. study. Life Sci Soc Policy 9(1):1. , DOI 10.1186/2195-7819-9-1 88. Melas PA, Sjoholm LK, Forsner T, Edhborg M, Juth N, Forsell Y et al, 2010, Examining the public refusal to consent to DNA biobanking: empirical data from a Swedish population-based study. J Med Ethics 36(2):93–98. , DOI 10.1136/jme. 2009.032367 89. Pereira S, Gibbs RA, McGuire AL, 2014, Open Access Data Sharing in Genomic Research. Gene 5(3):739–747. https://doi. org/10.3390/genes5030739 90. Hood L, 2013, Systems biology and p4 medicine: past, present, and future. Rambam Maimonides Med J 4(2):e0012. https://doi. org/10.5041/RMMJ.10112 91. Kreiner T, Irion S, 2013, Whole-genome analysis, stem cell research, and the future of biobanks. Cell Stem Cell 12(5):513–516. , DOI 10.1016/j.stem.2013.04.024 92. Gelpi E, van der Zee J, Turon Estrada A, Van Broeckhoven C, Sanchez-Valle R, 2014, TARDBP mutation p.Ile383Val associated with semantic dementia and complex proteinopathy. Neuropathol Appl Neurobiol 40(2):225–230. , DOI 10. 1111/nan.12063 93. Koszarska M, Kucsma N, Kiss K, Varady G, Gera M, Antalffy G et al, 2014, Screening the Expression of ABCB6 in Erythrocytes Reveals an Unexpectedly High Frequency of Lan Mutations in Healthy Individuals. PLoS One 9(10):e111590. , DOI 10.1371/journal.pone.0111590 94. Jenkinson C, Elliott V, Menon U, Apostolidou S, Fourkala OE, Gentry-Maharaj A et al, 2014, Evaluation in pre-diagnosis samples discounts ICAM-1 and TIMP-1 as biomarkers for earlier diagnosis of pancreatic cancer. J Proteome 113c:400–402. https:// doi.org/10.1016/j.jprot.2014.10.001 95. Guo X, Long J, Zeng C,Michailidou K, GhoussainiM, Bolla MK et al, 2015, Fine-Scale Mapping of the 4q24 Locus Identifies Two Independent Loci Associated with Breast Cancer Risk. Cancer Epidemiol Biomark Prev 24(11):1680–1691. , DOI 10. 1158/1055-9965.epi-15-0363 96. Kvistad SS, Myhr KM, Holmoy T, Saltyte Benth J, Wergeland S, Beiske AG et al, 2015, Bodymass index influence interferon-beta treatment response inmultiple sclerosis. J Neuroimmunol 288:92– 97. , DOI 10.1016/j.jneuroim.2015.09.008 97. Teumer A, Tin A, Sorice R, Gorski M, Yeo NC, Chu AY et al, 2015, Genome-wide Association Studies Identify Genetic Loci Associated with Albuminuria in Diabetes. Diabetes. https://doi. org/10.2337/db15-1313 98. Westerlind H, Mellander MR, Bresso F, Munch A, Bonfiglio F, Assadi G et al, 2015, Dense genotyping of immune-related loci identifies HLA variants associated with increased risk of collagenous colitis. Gut. , DOI 10.1136/gutjnl-2015-309934 99. Wakuda K, Kenmotsu H, SerizawaM, Koh Y, Isaka M, Takahashi S et al, 2014, Molecular profiling of small cell lung cancer in a Japanese cohort. Lung Cancer, Amst, Netherlands, 84(2):139– 144. , DOI 10.1016/j.lungcan.2014.02.013 100. Allegra M, Giacchero D, Segalen C, Dumaz N, Butori C, Hofman V et al, 2014, A new KIT mutation, N505I, in acral melanoma confers constitutive signaling, favors tumorigenic properties, and is sensitive to imatinib. J Investig Dermatol 134(5):1473–1476. , DOI 10.1038/jid.2013.525 101. Esplin ED, Oei L, SnyderMP(2014, Personalized sequencing and the future of medicine: discovery, diagnosis and defeat of disease. Pharmacogenomics 15(14):1771–1790. , DOI 10.2217/ pgs.14.117 102. Lisanti MP, Tanowitz HB, 2012, Translational discoveries, personalized medicine, and living biobanks of the future.AmJ Pathol 180(4):1334–1336. , DOI 10.1016/j.ajpath.2012.02.003 103. van Ommen GJ, Tornwall O, Brechot C, Dagher G, Galli J, Hveem K et al, 2014, BBMRI-ERIC as a resource for pharmaceutical and life science industries: the development of biobank-based Expert Centres. Eur J Hum Gen: EJHG. , DOI 10. 1038/ejhg.2014.235 104. Womack C, Mager SR, 2014, Human biological sample biobanking to support tissue biomarkers in pharmaceutical research and development. Methods, San Diego, Calif, 70(1):3– 11. , DOI 10.1016/j.ymeth.2014.01.014 105. Lu YC, Chang JT, Huang YC, Huang CC, ChenWH, Lee LYet al, 2014, Combined determination of circulating miR-196a and miR-196b levels produces high sensitivity and specificity for early detection of oral cancer. Clin Biochem. , DOI 10.1016/j. clinbiochem.2014.11.020 106. Malaguarnera M, Uccello M, Bellanca S, La Rosa B, Vacante M, Cristaldi E et al, 2014, Elevated chromogranin A serum levels in ovarian carcinoma patients. Indian J Cancer 51(3):315–318. , DOI 10.4103/0019-509x.146776 107. Nagar S, Ahmed S, Peeples C, Urban N, Boura J, Thibodeau B et al, 2014, Evaluation of genetic biomarkers for distinguishing benign from malignant thyroid neoplasms. Am J Surg 207(4): 596–601. , DOI 10.1016/j.amjsurg.2013.06.012 108. Letellier E, Schmitz M, Baig K, Beaume N, Schwartz C, Frasquilho S et al, 2014, Identification of SOCS2 and SOCS6 as biomarkers in human colorectal cancer. Br J Cancer 111(4): 726–735. , DOI 10.1038/bjc.2014.377 109. Bielinski SJ, Olson JE, Pathak J, Weinshilboum RM, Wang L, Lyke KJ et al, 2014, Preemptive genotyping for personalized medicine: design of the right drug, right dose, right time-using genomic data to individualize treatment protocol. Mayo Clin Proc 89(1):25–33. , DOI 10.1016/j.mayocp.2013.10.021 110. Almoguera B, Vazquez L, Connolly JJ, Bradfield J, Sleiman P, Keating B et al, 2014, Imputation of TPMT defective alleles for the identification of patients with high-risk phenotypes. Front Genet 5:96. , DOI 10.3389/fgene.2014.00096 111. Kawai VK, Cunningham A, Vear SI, Van Driest SL, Oginni A, Xu H et al, 2014, Genotype and risk of major bleeding during warfarin treatment. Pharmacogenomics 15(16):1973–1983. https://doi. org/10.2217/pgs.14.153 112. Henderson GE, Cadigan RJ, Edwards TP, Conlon I, Nelson AG, Evans JP et al, 2013, Characterizing biobank organizations in the U.S.: results from a national survey. Genome Med 5(1):3. https:// doi.org/10.1186/gm407 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 771 EP 785 DI 10.1007/s12253-018-0418-4 PG 15 ER PT J AU Kastelan, S Gverovic Antunica, A Beketic Oreskovic, L Salopek Rabatic, J Kasun, B Bakija, I AF Kastelan, Snjezana Gverovic Antunica, Antonela Beketic Oreskovic, Lidija Salopek Rabatic, Jasminka Kasun, Boris Bakija, Ivana TI Conjunctival Melanoma - Epidemiological Trends and Features SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Conjunctival melanoma; Epidemiology; Clinical features; Diagnosis; Management; Prognosis ID Conjunctival melanoma; Epidemiology; Clinical features; Diagnosis; Management; Prognosis AB Conjunctival melanoma is a rare but sight and life threatening malignancy. It accounts for 2%–5% of all ocular tumours and 5%–7% of all ocular melanomas with an incidence of 0.2–0.8 per million in the Caucasian population with rare cases reported in the non-Caucasians. In recent decades the incidence of uveal melanoma has been relatively stable whilst conjunctival and cutaneous melanoma have shown increasing incidence which may be connected to the result of environmental exposure to ultraviolet light. The dissimilarity in incidence between light and dark pigmented individuals observed in conjunctival melanomas compared to uveal and cutaneous melanomas may be related to differences in their histological structures and genetic profile. Recent molecular biological studies support the fact that each type of melanoma undergoes its own molecular changes and has characteristic biological behaviour. Further studies are required for each type of melanoma in order to ascertain their individual etiology and pathogenesis and based on this knowledge develop relevant preventative and treatment procedures. C1 [Kastelan, Snjezana] University Hospital Dubrava, Department of OphthalmologyZagreb, Croatia. [Gverovic Antunica, Antonela] General Hospital Dubrovnik, Department of OphthalmologyDubrovnik, Croatia. [Beketic Oreskovic, Lidija] University Hospital for Tumors, Department of OncologyZagreb, Croatia. [Salopek Rabatic, Jasminka] University Hospital Dubrava, Department of OphthalmologyZagreb, Croatia. [Kasun, Boris] Special Hospital for Medical Rehabilitation Stubicke TopliceStubicke Toplice, Croatia. [Bakija, Ivana] Psychiatric Hospital Sveti IvanZagreb, Croatia. RP Kastelan, S (reprint author), University Hospital Dubrava, Department of Ophthalmology, Zagreb, Croatia. EM snjezanakastelan@yahoo.com CR Kao A, Afshar A, Bloomer M, Damato B, 2016, Management of primary acquired melanosis, nevus, and conjunctival melanoma. Cancer Control 23(2):117–125 PMID: 27218788 Vora GK, Demirci H, Marr B, Mruthyunjaya P, 2017, Advances in the management of conjunctival melanoma. Surv Ophthalmol 62(1):26–42. Jun 16. pii: S0039–6257(16)30058–3. https://doi. org/10.1016/j.survophthal.2016.06.001. PMID:27321895 Wong JR, Nanji AA, Galor A, Karp CL, 2014, Management of conjunctival malignant melanoma: a review and update. Expert Rev Ophthalmol 9(3):185–204. , DOI 10.1586/17469899. 2014.921119 Lim LA, Madigan MC, Conway RM, 2013, Conjunctival melanoma: a review of conceptual and treatment advances. Clin Ophthalmol 6:521–531. , DOI 10.2147/OPTH.S38415 Norregaard JC, Gerner N, Jensen OA, Prause JU, 1996, Malignant melanoma of the conjunctiva: occurrence and survival following surgery and radiotherapy in a Danish population. Graefes Arch Clin Exp Ophthalmol 234(9):569–572 Lommatzsch PK, Lommatzsch RE, Kirsch I, Fuhrmann P, 1990, Therapeutic outcome of patients suffering from malignant melanomas of the conjunctiva. Br J Ophthalmol 74(10):615–619 Seregard S, Kock E, 1992, Conjunctival malignant melanoma in Sweden 1969-91. Acta Ophthalmol 70(3):289–296 Isager P, Engholm G, Overgaard J, Storm H, 2006, Uveal and conjunctival malignant melanoma in Denmark 1943–1997: observed and relative survival of patients followed through 2002. Ophthalmic Epidemiol 13(2):85–96 Vajdic CM, Kricker A, Giblin M,McKenzie J, Aitken J, Giles GG, Armstrong BK, 2003, Incidence of ocular melanoma in Australia from 1990 to 1998. Int J Cancer 105(1):117–122 Triay E, Bergman L, Nilsson B, All-Ericsson C, Seregard S, 2009, Time trends in the incidence of conjunctival melanoma in Sweden. Br J Ophthalmol 93(11):1524–1528. , DOI 10.1136/bjo. 2009.157933 Epub 2009 Jul 23 Vasalaki M, Fabian ID, Reddy MA, Cohen VM, Sagoo MS, 2017, Ocular oncology: advances in retinoblastoma, uveal melanoma and conjunctival melanoma. Br Med Bull1 121(1):107–119. https://doi. org/10.1093/bmb/ldw053 Review. PMID: 28069617 Yu GP, Hu DN, McCormic S, Finger PT, 2003, Conjunctival melanoma: is it increasing in the United States? Am J Ophthalmol 135(6):800–806 Tuomaala S, Kivela T, 2003, Conjunctival melanoma: is it increasing in the United States? Am J Ophthalmol 136(6):1189–1190 Chang AE, Karnell LH,Menck HR, 1998, The national cancer data base report on cutaneous and noncutaneous melanoma: a summary of 84,836 cases from the past decade. The American college of surgeons commission on cancer and the american cancer society. Cancer. 83(8):1664–1678 Tuomaala S, Eskelin S, Tarkkanen A, Kivela T, 2002, Populationbased assessment of clinical characteristic predicting outcome of conjunctival melanoma in whites. Invest Ophthalmol Vis Sci 43(11):3399–3408 Inskip PD, Devesa SS, Fraumeni JF Jr, 2003, Trends in the incidence of ocular melanoma in the United States, 1974-1998. Cancer Causes Control 14(3):251–257 Missotten GS, Keijser S, De Keizer RJ, De Wolff-Rouendaal D, 2005, Conjunctival melanoma in the Netherlands: a nationwide study. Invest Ophthalmol Vis Sci 46(1):75–82 Isager P, Osterlind A, Engholm G, Heegaard S, Lindegaard J, Overgaard J, Storm HH, 2005, Uveal and conjunctival malignant melanoma in Denmark, 1943-97: incidence and validation study. Ophthalmic Epidemiol 12(4):223–232. , DOI 10.1080/ 09286580591000836 Hu DN, Yu G, McCormick SA, Finger PT, 2008, Population-based incidence of conjunctival melanoma in various races and ethnic groups and comparison with other melanomas. Am J Ophthalmol 145(3):418–423. , DOI 10.1016/j.ajo.2007.10.022 Epub 2008 Jan 11 Park SJ, Oh CM, Kim BW, Woo SJ, Cho H, Park KH, 2015, Nationwide incidence of ocular melanoma in South Korea by using the national cancer registry database, 1999-2011). Invest Ophthalmol Vis Sci 56(8):4719–4724. , DOI 10.1167/ iovs.15-16532 Larsen AC, 2016, Conjunctival malignant melanoma in Denmark: epidemiology, treatment and prognosis with special emphasis on tumorigenesis and genetic profile. Acta Ophthalmol 94 Thesis 1: 1–27. , DOI 10.1111/aos.13100. PMID:27192168 Brownstein S, 2004, Malignant melanoma of the conjunctiva. Cancer Control 11(5):310–316 Zembowicz A, Mandal RV, Choopong P, 2010, Melanocytic lesions of the conjunctiva. Arch Pathol Lab Med 134(12):1785– 1792. , DOI 10.1043/2009-0522-RAR.1. Taban M, Traboulsi EI, 2007, Malignant melanoma of the conjunctiva in children: a review of the international literature 1965-2006. J Pediatr Ophthalmol Strabismus 44(5):277–282 Shields CL, Shields JA, Gunduz K, Cater J,Mercado GV, Gross N, Lally B, 2000, Conjunctival melanoma: risk factors for recurrence, exenteration, metastasis, and death in 150 consecutive patients. Arch Ophthalmol 118(11):1497–1507 Seregard S, 1998, Conjunctival melanoma. Surv Ophthalmol 42(4):321–350 Kenawy N, Lake SL, Coupland SE, Damato BE, 2013, Conjunctival melanoma and melanocytic intra-epithelial neoplasia. Eye, Lond, 27(2):142–152. , DOI 10.1038/eye.2012.254 Epub 2012 Dec 7 Jovanovic P, Mihajlovic M, Djordjevic-Jocic J, Vlajkovic S, Cekic S, Stefanovic V, 2013, Ocular melanoma: an overview of the current status. Int J Clin Exp Pathol 15;6(7):1230–1244. Shields CL, Shields JA, 2009, Ocularmelanoma: relatively rare but requiring respect. Clin Dermatol 27(1):122–133. , DOI 10. 1016/j.clindermatol.2008.09.010 Jakobiec FA, Folberg R, Iwamoto T, 1989, Clinicopathologic characteristics of premalignant andmalignantmelanocytic lesions of the conjunctiva. Ophthalmology 96(2):147–166 Ophthalmic Oncology Task Force AJCC-UICC, 2009, Malignant melanoma of the conjunctiva. In: Edge SE, Byrd DR, Carducci MA, Compton CA, eds, AJCC cancer staging manual. 7th edn. NY: Springer, New York, pp 539–546 Paridaens AD, Minassian DC, McCartney AC, Hungerford JL, 1994, Prognostic factors in primary malignant melanoma of the conjunctiva: a clinicopathological study of 256 cases. Br J Ophthalmol 78(4):252–259 Tuomaala S, Toivonen P, Al-Jamal R, Kivela T, 2007, Prognostic significance of histopathology of primary conjunctival melanoma in Caucasians. Curr Eye Res 32(11):939–952 Grossniklaus HE, Margo CE, Solomon AR, 1999, Indeterminate melanocytic proliferations of the conjunctiva. Arch Ophthalmol 117:1131–1136 Henderson E, Margo CE, 2008, Iris melanoma. Arch Pathol Lab Med 132(2):268–272 Perez MT, Suster S, 1999, Balloon cell change in cellular blue nevus. Am J Dermatopathol 21(2):181–184 Calonje E, 2004, Non-melanocytic lesions mimicking melanocytic lesions. Pathology 36(5):387–395 PMID:15370107 Maly A, Epstein D, Meir K, Pe’er J, 2008, Histological criteria for grading of atypia in melanocytic conjunctival lesions. Pathology 40(7):676–681 Shields CL, Kaliki S, Al-Dahmash SA, Lally SE, Shields JA, 2012, American joint committee on cancer, AJCC, clinical classification predicts conjunctival melanoma outcomes. Ophthal Plast Reconstr Surg 28(5):313–323. , DOI 10.1097/IOP.0b013e3182611670 Kivela T, Kujala E, 2013, Prognostication in eye cancer: the latest tumor, node, metastasis classification and beyond. Eye, Lond, 27(2):243–252. , DOI 10.1038/eye.2012.256 Review. PMID: 23258307 Yousef YA, Finger PT, 2012, Predictive value of the seventh edition American joint committee on cancer staging system for conjunctival melanoma. Arch Ophthalmol 130(5):599–606. https://doi. org/10.1001/archophthalmol.2011.2566. Esmaeli B, Eicher S, Popp J, Delpassand E, Prieto VG, Gershenwald JE, 2001, Sentinel lymph node biopsy for conjunctival melanoma. Ophthal Plast Reconstr Surg 17(6):436–442 Sheng X, Li S, Chi Z, Si L, Cui C, Mao L, Lian B, Tang B,Wang X, Yan X, Kong Y, Dai J, Guo J, 2015, Prognostic factors for conjunctivalmelanoma: a study in ethnic Chinese patients. Br J Ophthalmol 99(7):990–996. , DOI 10.1136/bjophthalmol-2014- 305730 Epub 2015 Jan 16 Salcedo-Hernandez RA, Luna-Ortiz K, Lino-Silva LS, Herrera- Gomez A, Villavicencio-Valencia V, Tejeda-Rojas M, Carrillo JF, 2014, Conjunctival melanoma: survival analysis in twenty-two Mexican patients. Arq Bras Oftalmol 77(3):155–158 Larsen AC, Dahmcke CM, Dahl C, Siersma VD, Toft PB, Coupland SE, Prause JU, Guldberg P, Heegaard S, 2015, A retrospective review of conjunctival melanoma presentation, treatment, and outcome and an investigation of features associated with BRAF mutations. JAMA Ophthalmol 133(11):1295–1303. , DOI 10.1001/jamaophthalmol.2015.3200 Griewank KG, Westekemper H, Murali R, Mach M, Schilling B, Wiesner T, Schimming T, Livingstone E, Sucker A, Grabellus F, Metz C, Susskind D, Hillen U, SpeicherMR,Woodman SE, Steuhl KP, Schadendorf D, 2013, Conjunctival Melanomas Harbor BRAF and NRAS mutations and copy number changes similar to cutaneous and mucosal melanomas. Clin. Cancer Res 19(12):3143–3152. , DOI 10.1158/1078-0432.CCR-13-2368. Lake SL, Jmor F, Dopierala J, Taktak AF, Coupland SE, Damato BE, 2011, Multiplex ligation-dependent probe amplification of conjunctival melanoma reveals common BRAF V600E gene mutation and gene copy number changes. Invest Ophthalmol Vis Sci 52(8):5598–5604 Spendlove HE, Damato BE, Humphreys J, Barker KT, Hiscott PS, Houlston RS, 2004, BRAF mutations are detectable in conjunctival but not uveal melanomas. Melanoma Res 14(6):449–452 Wallander ML, Layfield LJ, Emerson LL, Mamalis N, Davis D, Tripp SR, Holden JA, 2011, KIT mutations in ocular melanoma: frequency and anatomic distribution. Mod Pathol 24(8):1031– 1035. , DOI 10.1038/modpathol.2011.57 Epub 2011 Apr 8 Sheng X, Li S, Chi Z, Si L, Cui C, Mao L, Lian B, Tang B,Wang X, Yan X, Kong Y, Dai J, Guo J, 2015, Prognostic factors for conjunctivalmelanoma: a study in ethnic Chinese patients. Br J Ophthalmol 99(7):990–996. , DOI 10.1136/bjophthalmol-2014- 305730 PMID:25595173 Patel M, Smyth E, Chapman PB, Wolchok JD, Schwartz GK, Abramson DH, Carvajal RD, 2011, Therapeutic implications of the emerging molecular biology of uveal melanoma. Clin Cancer Res 17:2087–2100. , DOI 10.1016/j.jaad.2013.12.019 Dogrusoz M, Jager MJ, 2017, Genetic prognostication in uveal melanoma. Acta Ophthalmol, Nov 4). , DOI 10.1111/aos.13580 Swaminathan SS, Field MG, Sant D, Wang G, Galor A, Dubovy SR, Harbour JW, Karp CL, 2017)Molecular characteristics of conjunctival melanoma using whole-exome sequencing. 135(12): 1434–1437. , DOI 10.1001/jamaophthalmol.2017.4837. Pandiani C, Beranger GE, Leclerc J, Ballotti R, Bertolotto C, 2017, Focus on cutaneous and uveal melanoma specificities. Genes Dev 31(8):724–743. , DOI 10.1101/gad.296962.117 Kastelan S, Gverovic Antunica A, Beketic-Oreskovic L, Bakija I, BogadiM(2017, Uveal melanoma: clinical features and diagnostic procedures. Libri Oncol 45(2-3):81-88 Juzeniene A, Micu E, Porojnicu AC, Moan J, 2012, Malignant melanomas on head/neck and foot: differences in time and latitudinal trends in Norway. J Eur Acad Dermatol Venereol 26(7):821–827. , DOI 10.1111/j. 1468-3083.2011.04162.x Epub 2011 Jun 28 Yu GP, Hu DN, McCormick SA, 2006, Latitude and incidence of ocular melanoma. Photochem Photobiol 82(6):1621–1626 Griewank KG,Murali R, Schilling B, Scholz S, Sucker A, Song M, Susskind D, Grabellus F, Zimmer L, Hillen U, Steuhl KP, Schadendorf D, Westekemper H, Zeschnigk M, 2013, TERT promoter mutations in ocular melanoma distinguish between conjunctival and uveal tumours. Br J Cancer 23;109(2):497–501. https:// doi.org/10.1038/bjc.2013.312. Rivolta C, Royer-Bertrand B, Rimoldi D, Schalenbourg A, Zografos L, Leyvraz S, Moulin A, 2016, UV light signature in conjunctival melanoma; not only skin should be protected from solar radiation. J Hum Genet 61(4):361–362. , DOI 10. 1038/jhg.2015.152 Epub 2015 Dec 10 Franco-Lie I, Iversen T, Robsahm TE, Abdelnoor M, 2011, Incidence trends of melanoma of the skin compared with other localisations, in the Norwegian population, 1956-2005. Ann Oncol 22(6):1443–1450. , DOI 10.1093/annonc/mdq598 Epub 2010 Dec 6 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 787 EP 796 DI 10.1007/s12253-018-0419-3 PG 10 ER PT J AU Ishiwata, T Matsuda, Y Yoshimura, H Sasaki, N Ishiwata, Sh Ishikawa, N Takubo, K Arai, T Junko, A AF Ishiwata, Toshiyuki Matsuda, Yoko Yoshimura, Hisashi Sasaki, Norihiko Ishiwata, Shunji Ishikawa, Naoshi Takubo, Kaiyo Arai, Tomio Junko, Aida TI Pancreatic cancer stem cells: features and detection methods SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Pancreatic cancer; Cancer stem cell/CSC; Sphere; Side population; Cancer stem cell marker ID Pancreatic cancer; Cancer stem cell/CSC; Sphere; Side population; Cancer stem cell marker AB Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a high incidence of distant metastasis and recurrence. Cancer stem cells (CSCs), which are pluripotent, self-renewable, and capable of forming tumors, contribute to PDAC initiation and metastasis and are responsible for resistance to chemotherapy and radiation. Three types of experimental methods are commonly used to identify CSCs: CSC-specific marker detection, a sphere-formation assay that reveals cell proliferation under non-adherent conditions, and detection of side-population (SP) cells that possess high intracellular-to-extracellular pump functions. Several CSC-specific markers have been reported in PDACs, including CD133, CD24, CD44, CXCR4, EpCAM, ABCG2, c-Met, ALDH-1, and nestin. There remains controversy regarding which markers are specific to PDAC CSCs and which are expressed alone or in combination in CSCs. Examining characteristics of isolated CSCs and discovering CSC-specific treatment options are important to improve the prognosis of PDAC cases. This review summarizes CSC-detection methods for PDAC, including CSC-marker detection, the sphere-formation assay, and detection of SP cells. C1 [Ishiwata, Toshiyuki] Tokyo Metropolitan Institute of Gerontology, Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, 35-2 Sakae-cho, Itabashi-ku, 173-0015 Tokyo, Japan. [Matsuda, Yoko] Tokyo Metropolitan Institute of Gerontology, Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, 173-0015 Tokyo, Japan. [Yoshimura, Hisashi] Nippon Veterinary and Life Science University, School of Veterinary Nursing and Technology, Department of Applied Science, 180-0022 Tokyo, Japan. [Sasaki, Norihiko] Tokyo Metropolitan Institute of Gerontology, Research Team for Geriatric Medicine (Vascular Medicine), 173-0015 Tokyo, Japan. [Ishiwata, Shunji] Kindai University, Faculty of Pharmacy, Division of Medical Pharmaceutics & Therapeutics, 3-4-1 Kowakae, Higashi-Osaka, 577-8502 Osaka, Japan. [Ishikawa, Naoshi] Tokyo Metropolitan Institute of Gerontology, Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, 35-2 Sakae-cho, Itabashi-ku, 173-0015 Tokyo, Japan. [Takubo, Kaiyo] Tokyo Metropolitan Institute of Gerontology, Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, 35-2 Sakae-cho, Itabashi-ku, 173-0015 Tokyo, Japan. [Arai, Tomio] Tokyo Metropolitan Institute of Gerontology, Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, 173-0015 Tokyo, Japan. [Junko, Aida] Tokyo Metropolitan Institute of Gerontology, Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, 35-2 Sakae-cho, Itabashi-ku, 173-0015 Tokyo, Japan. RP Ishiwata, T (reprint author), Tokyo Metropolitan Institute of Gerontology, Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, 173-0015 Tokyo, Japan. EM tishiwat@tmig.or.jp CR Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D, Bray F, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49(6):1374–1403. , DOI 10.1016/j. ejca.2012.12.027 Jemal A, Siegel R,Ward E,HaoY, Xu J,Murray T, ThunMJ, 2008, Cancer statistics, 2008. CA Cancer J Clin 58(2):71–96 Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66(1):7–30. , DOI 10.3322/caac.21332 Ahrendt SA, Pitt HA, 2002, Surgical management of pancreatic cancer. Oncology, Williston Park, 16(6):725–734 discussion 734, 736-728, 740, 743 Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM, 2014, Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 74(11):2913–2921. , DOI 10.1158/0008-5472.CAN-14-0155 Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA, 2006, Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 20(10):1218–1249. , DOI 10.1101/gad. 1415606 Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, Kamiyama M, Hruban RH, Eshleman JR, Nowak MA, Velculescu VE, Kinzler KW, Vogelstein B, Iacobuzio-Donahue CA, 2010, Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467(7319):1114–1117. , DOI 10.1038/nature09515 Lee CJ, Dosch J, SimeoneDM(2008, Pancreatic cancer stem cells. J Clin Oncol 26(17):2806–2812. , DOI 10.1200/JCO. 2008.16.6702 Hermann PC, Mueller MT, Heeschen C, 2009, Pancreatic cancer stem cells–insights and perspectives. Expert Opin Biol Ther 9(10): 1271–1278. , DOI 10.1517/14712590903246362 Ischenko I, Seeliger H, Kleespies A, Angele MK, Eichhorn ME, Jauch KW, Bruns CJ, 2009, Pancreatic cancer stem cells: new understanding of tumorigenesis, clinical implications. Langenbeck's Arch Surg 395(1):1–10. , DOI 10.1007/ s00423-009-0502-z Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weissman IL, Wahl GM, 2006, Cancer stem cells– perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res 66(19):9339–9344. , DOI 10.1158/0008-5472.CAN-06-3126 Hruban RH, Adsay NV, Albores-Saavedra J, Compton C, Garrett ES, Goodman SN, Kern SE, Klimstra DS, Kloppel G, Longnecker DS, Luttges J, Offerhaus GJ, 2001, Pancreatic intraepithelial neoplasia: a new nomenclature and classification system for pancreatic duct lesions. Am J Surg Pathol 25(5):579–586 Kure S, Matsuda Y, Hagio M, Ueda J, Naito Z, Ishiwata T, 2012, Expression of cancer stem cell markers in pancreatic intraepithelial neoplasias and pancreatic ductal adenocarcinomas. Int J Oncol 41(4):1314–1324. , DOI 10.3892/ijo.2012.1565 Guerra C, Schuhmacher AJ, Canamero M, Grippo PJ, Verdaguer L, Perez-Gallego L, Dubus P, Sandgren EP, Barbacid M, 2007, Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell 11(3):291–302. , DOI 10.1016/j.ccr.2007.01.012 Zhu L, Shi G, Schmidt CM, Hruban RH, Konieczny SF, 2007, Acinar cells contribute to the molecular heterogeneity of pancreatic intraepithelial neoplasia. Am J Pathol 171(1):263–273. https://doi. org/10.2353/ajpath.2007.061176 Hruban RH, Adsay NV, Albores-Saavedra J, Anver MR, Biankin AV, Boivin GP, Furth EE, Furukawa T, Klein A, Klimstra DS, Kloppel G, Lauwers GY, Longnecker DS, Luttges J, Maitra A, Offerhaus GJ, Perez-Gallego L, Redston M, Tuveson DA, 2006, Pathology of genetically engineered mouse models of pancreatic exocrine cancer: consensus report and recommendations. Cancer Res 66(1):95–106. , DOI 10.1158/0008-5472.CAN-05- 2168 Oshima Y, Suzuki A, Kawashimo K, Ishikawa M, Ohkohchi N, Taniguchi H, 2007, Isolation of mouse pancreatic ductal progenitor cells expressing CD133 and c-Met by flow cytometric cell sorting. Gastroenterology 132(2):720–732. , DOI 10.1053/j.gastro. 2006.11.027 Morrison SJ, Spradling AC, 2008, Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell 132(4):598–611. , DOI 10.1016/j.cell.2008.01.038 Hwang RF, Moore T, Arumugam T, Ramachandran V, Amos KD, Rivera A, Ji B, Evans DB, Logsdon CD, 2008, Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res 68(3):918–926. , DOI 10.1158/0008-5472.CAN-07- 5714 De Santis M, Di Gianantonio E, Cesari E, Ambrosini G, Straface G, Clementi M, 2009, First-trimester itraconazole exposure and pregnancy outcome: a prospective cohort study of women contacting teratology information services in Italy. Drug Saf 32(3):239–244 Ikenaga N, Ohuchida K,Mizumoto K, Yu J, Kayashima T, Hayashi A, Nakata K, Tanaka M, 2010, Characterization of CD24 expression in intraductal papillary mucinous neoplasms and ductal carcinoma of the pancreas. Hum Pathol 41(10):1466–1474. https://doi. org/10.1016/j.humpath.2010.04.004 Hong SP, Wen J, Bang S, Park S, Song SY, 2009, CD44-positive cells are responsible for gemcitabine resistance in pancreatic cancer cells. Int J Cancer 125(10):2323–2331. , DOI 10.1002/ijc. 24573 Marechal R, Demetter P, Nagy N, Berton A, Decaestecker C, Polus M, Closset J, Deviere J, Salmon I, Van Laethem JL, 2009, High expression of CXCR4 may predict poor survival in resected pancreatic adenocarcinoma. Br J Cancer 100(9):1444–1451. https:// doi.org/10.1038/sj.bjc.6605020 Deng S, Yang X, Lassus H, Liang S, Kaur S, Ye Q, Li C,Wang LP, Roby KF, Orsulic S, Connolly DC, Zhang Y, Montone K, Butzow R, Coukos G, Zhang L, 2010, Distinct expression levels and patterns of stem cell marker, aldehyde dehydrogenase isoform 1, ALDH1), in human epithelial cancers. PLoS One 5(4):e10277. , DOI 10.1371/journal.pone.0010277 Ishiwata T,MatsudaY, Naito Z, 2011, Nestin in gastrointestinal and other cancers: Effects on cells and tumor angiogenesis. World J Gastroenterol 17(4):409–418. , DOI 10.3748/wjg.v17.i4. 409 Kawamoto M, Ishiwata T, Cho K, Uchida E, Korc M, Naito Z, Tajiri T, 2009, Nestin expression correlates with nerve and retroperitoneal tissue invasion in pancreatic cancer. Hum Pathol 40(2): 189–198. , DOI 10.1016/j.humpath.2008.02.022 Matsuda Y, Naito Z, Kawahara K, Nakazawa N, Korc M, Ishiwata T, 2011, Nestin is a novel target for suppressing pancreatic cancer cell migration, invasion and metastasis. Cancer Biol Ther 11(5): 512–523 Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, Bruns CJ, Heeschen C, 2007, Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1(3):313–323. , DOI 10. 1016/j.stem.2007.06.002 Matsuda Y, Yoshimura H, Ueda J, Naito Z, Korc M, Ishiwata T, 2014, Nestin delineates pancreatic cancer stem cells in metastatic foci of NOD/Shi-scid IL2Rgamma(null,, NOG, mice. Am J Pathol 184(3):674–685. , DOI 10.1016/j.ajpath.2013.11.014 Olempska M, Eisenach PA, Ammerpohl O, Ungefroren H, Fandrich F, Kalthoff H, 2007, Detection of tumor stem cellmarkers in pancreatic carcinoma cell lines. Hepatobiliary Pancreat Dis Int 6(1):92–97 Huang P, Wang CY, Gou SM, Wu HS, Liu T, Xiong JX, 2008, Isolation and biological analysis of tumor stem cells from pancreatic adenocarcinoma. World J Gastroenterol 14(24):3903–3907 Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM, 2007, Identification of pancreatic cancer stem cells. Cancer Res 67(3):1030–1037. , DOI 10.1158/0008-5472.CAN-06-2030 Li C, Wu JJ, Hynes M, Dosch J, Sarkar B, Welling TH, Pasca di Magliano M, Simeone DM, 2011, c-Met is a marker of pancreatic cancer stem cells and therapeutic target. Gastroenterology 141(6): 2218–2227 e2215. , DOI 10.1053/j.gastro.2011.08.009 Sarkar FH, Li Y, Wang Z, Kong D, 2009, Pancreatic cancer stem cells and EMT in drug resistance and metastasis. Minerva Chir 64(5):489–500 Matsuda Y, Kure S, Ishiwata T, 2012, Nestin and other putative cancer stem cell markers in pancreatic cancer. Med Mol Morphol 45(2):59–65. , DOI 10.1007/s00795-012-0571-x Bednar F, Simeone DM, 2009, Pancreatic cancer stem cells and relevance to cancer treatments. J Cell Biochem 107(1):40–45. , DOI 10.1002/jcb.22093 Ishimoto T, Nagano O, Yae T, Tamada M, Motohara T, Oshima H, Oshima M, Ikeda T, Asaba R, Yagi H, Masuko T, Shimizu T, Ishikawa T, Kai K, Takahashi E, Imamura Y, Baba Y, Ohmura M, SuematsuM, Baba H, Saya H, 2011, CD44 variant regulates redox status in cancer cells by stabilizing the xCTsubunit of system xc(−, and thereby promotes tumor growth. Cancer Cell 19(3):387–400. , DOI 10.1016/j.ccr.2011.01.038 Shmelkov SV, St Clair R, Lyden D, Rafii S, 2005, AC133/CD133/ Prominin-1. Int J BiochemCell Biol 37(4):715–719. , DOI 10.1016/j.biocel.2004.08.010 Wright MH, Calcagno AM, Salcido CD, Carlson MD, Ambudkar SV, Varticovski L, 2008, Brca1 breast tumors contain distinct CD44+/CD24- and CD133+ cells with cancer stem cell characteristics. Breast Cancer Res 10(1):R10. , DOI 10.1186/ bcr1855 Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB, 2003, Identification of a cancer stem cell in human brain tumors. Cancer Res 63(18):5821–5828 Suetsugu A, Nagaki M, Aoki H, Motohashi T, Kunisada T, Moriwaki H, 2006, Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells. Biochem Biophys Res Commun 351(4):820–824. , DOI 10.1016/j. bbrc.2006.10.128 O'Brien CA, Pollett A, Gallinger S, Dick JE, 2007, A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445(7123):106–110. , DOI 10.1038/ nature05372 Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R, 2007, Identification and expansion of human colon-cancer-initiating cells. Nature 445(7123):111–115. , DOI 10.1038/nature05384 Shepherd CJ, Rizzo S, Ledaki I, Davies M, Brewer D, Attard G, de Bono J, Hudson DL, 2008, Expression profiling of CD133+ and CD133- epithelial cells from human prostate. Prostate 68(9):1007– 1024. , DOI 10.1002/pros.20765 Immervoll H, Hoem D, Sakariassen PO, Steffensen OJ, Molven A, 2008, Expression of the "stemcellmarker" CD133 in pancreas and pancreatic ductal adenocarcinomas. BMC Cancer 8:48. https://doi. org/10.1186/1471-2407-8-48 Maeda S, Shinchi H, Kurahara H, Mataki Y, Maemura K, Sato M, Natsugoe S, Aikou T, Takao S, 2008, CD133 expression is correlated with lymph node metastasis and vascular endothelial growth factor-C expression in pancreatic cancer. Br J Cancer 98(8):1389– 1397. , DOI 10.1038/sj.bjc.6604307 Taieb N, Maresca M, Guo XJ, Garmy N, Fantini J, Yahi N, 2009, The first extracellular domain of the tumour stem cell marker CD133 contains an antigenic ganglioside-binding motif. Cancer Lett 278(2):164–173. , DOI 10.1016/j.canlet.2009.01.013 Boivin D, Labbe D, Fontaine N, Lamy S, Beaulieu E, Gingras D, Beliveau R, 2009, The stem cell marker CD133, prominin-1, is phosphorylated on cytoplasmic tyrosine-828 and tyrosine-852 by Src and Fyn tyrosine kinases. Biochemistry 48(18):3998–4007. , DOI 10.1021/bi900159d Chen YS, Wu MJ, Huang CY, Lin SC, Chuang TH, Yu CC, Lo JF, 2011, CD133/Src axis mediates tumor initiating property and epithelial-mesenchymal transition of head and neck cancer. PLoS One 6(11):e28053. , DOI 10.1371/journal.pone.0028053 Hashimoto O, Shimizu K, Semba S, Chiba S, Ku Y, Yokozaki H, Hori Y, 2011, Hypoxia induces tumor aggressiveness and the expansion of CD133-positive cells in a hypoxia-inducible factor-1alpha-dependent manner in pancreatic cancer cells. Pathobiology 78(4):181–192. , DOI 10.1159/000325538 McCord AM, Jamal M, Shankavaram UT, Lang FF, Camphausen K, Tofilon PJ, 2009, Physiologic oxygen concentration enhances the stem-like properties of CD133+ human glioblastoma cells in vitro. Mol Cancer Res 7(4):489–497. , DOI 10.1158/ 1541-7786.MCR-08-0360 Lonardo E, Hermann PC, Heeschen C, 2010, Pancreatic cancer stem cells - update and future perspectives. Mol Oncol 4(5):431– 442. , DOI 10.1016/j.molonc.2010.06.002 Thomas RM, Kim J, Revelo-Penafiel MP, Angel R, Dawson DW, Lowy AM, 2008, The chemokine receptor CXCR4 is expressed in pancreatic intraepithelial neoplasia. Gut 57(11):1555–1560. https:// doi.org/10.1136/gut.2007.143941 Jimeno A, Feldmann G, Suarez-Gauthier A, Rasheed Z, Solomon A, Zou GM, Rubio-Viqueira B, Garcia-Garcia E, Lopez-Rios F, Matsui W, Maitra A, Hidalgo M, 2009, A direct pancreatic cancer xenograft model as a platform for cancer stem cell therapeutic development. Mol Cancer Ther 8(2):310–314. , DOI 10. 1158/1535-7163.MCT-08-0924 Rausch V, Liu L, Kallifatidis G, Baumann B, Mattern J, Gladkich J, Wirth T, Schemmer P, Buchler MW, Zoller M, Salnikov AV, Herr I, 2010, Synergistic activity of sorafenib and sulforaphane abolishes pancreatic cancer stem cell characteristics. Cancer Res 70(12): 5004–5013. , DOI 10.1158/0008-5472.CAN-10-0066 Kim MP, Fleming JB,Wang H, Abbruzzese JL, ChoiW, Kopetz S, McConkey DJ, Evans DB, Gallick GE, 2011, ALDH activity selectively defines an enhanced tumor-initiating cell population relative to CD133 expression in human pancreatic adenocarcinoma. PLoS One 6(6):e20636. , DOI 10.1371/journal.pone. 0020636 Rao CV, Mohammed A, 2015, New insights into pancreatic cancer stem cells. World J Stem Cells 7(3):547–555. , DOI 10. 4252/wjsc.v7.i3.547 Sureban SM, May R, Lightfoot SA, Hoskins AB, Lerner M, Brackett DJ, Postier RG, Ramanujam R, Mohammed A, Rao CV, Wyche JH, Anant S, Houchen CW, 2011, DCAMKL-1 regulates epithelial-mesenchymal transition in human pancreatic cells through a miR-200a-dependent mechanism. Cancer Res 71(6): 2328–2338. , DOI 10.1158/0008-5472.CAN-10-2738 Sureban SM, May R, Ramalingam S, Subramaniam D, Natarajan G, Anant S, HouchenCW(2009, Selective blockade ofDCAMKL- 1 results in tumor growth arrest by a Let-7a MicroRNA-dependent mechanism. Gastroenterology 137(2):649–659, 659 e641-642. , DOI 10.1053/j.gastro.2009.05.004 Jaks V, Barker N, Kasper M, van Es JH, Snippert HJ, Clevers H, Toftgard R, 2008, Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat Genet 40(11):1291–1299. , DOI 10.1038/ ng.239 Mizuno N, Yatabe Y, Hara K, Hijioka S, Imaoka H, Shimizu Y, Ko SB, Yamao K, 2013, Cytoplasmic expression of LGR5 in pancreatic adenocarcinoma. Front Physiol 4:269. , DOI 10.3389/ fphys.2013.00269 Dorado J, Lonardo E, Miranda-Lorenzo I, Heeschen C, 2011, Pancreatic cancer stem cells: new insights and perspectives. J Gastroenterol 46(8):966–973. , DOI 10.1007/s00535-011- 0422-x Wu BL, Xu LY, Du ZP, Liao LD, Zhang HF, Huang Q, FangGQ, Li EM(2011)MiRNA profile in esophageal squamous cell carcinoma: downregulation of miR-143 and miR-145. World J Gastroenterol 17(1):79–88. , DOI 10.3748/wjg.v17.i1.79 Gou S, Liu T, Wang C, Yin T, Li K, Yang M, Zhou J, 2007, Establishment of clonal colony-forming assay for propagation of pancreatic cancer cells with stem cell properties. Pancreas 34(4): 429–435. , DOI 10.1097/MPA.0b013e318033f9f4 Gaviraghi M, Tunici P, Valensin S, Rossi M, Giordano C,Magnoni L, Dandrea M, Montagna L, Ritelli R, Scarpa A, Bakker A, 2011, Pancreatic cancer spheres are more than just aggregates of stem marker-positive cells. Biosci Rep 31(1):45–55. , DOI 10. 1042/BSR20100018 Yin T, Wei H, Gou S, Shi P, Yang Z, Zhao G, Wang C, 2011, Cancer stem-like cells enriched in panc-1 spheres possess increased migration ability and resistance to gemcitabine. Int JMol Sci 12(3): 1595–1604. , DOI 10.3390/ijms12031595 Ishiwata T, Hasegawa F, Michishita M, Sasaki N, Ishikawa N, Takubo K, Arai T, Aida J, 2018, Electron microscopic analysis of different cell types in human pancreatic cancer spheres. Oncol Lett 15:2485–2490. , DOI 10.3892/ol.2017.7554 Matsuda Y, Ishiwata T, Yoshimura H, Yamashita S, Ushijima T, Arai T, 2016, Systemic Administration of Small Interfering RNA Targeting Human Nestin Inhibits Pancreatic Cancer Cell Proliferation and Metastasis. Pancreas 45(1):93–100. https://doi. org/10.1097/MPA.0000000000000427 Bhagwandin VJ, Shay JW(2009, Pancreatic cancer stem cells: fact or fiction? Biochim Biophys Acta 1792(4):248–259. https://doi. org/10.1016/j.bbadis.2009.02.007 Hirschmann-Jax C, Foster AE, Wulf GG, Nuchtern JG, Jax TW, Gobel U, Goodell MA, Brenner MK, 2004, A distinct "side population" of cells with high drug efflux capacity in human tumor cells. Proc Natl Acad Sci U S A 101(39):14228–14233. , DOI 10.1073/pnas.0400067101 Wang YH, Li F, Luo B,Wang XH, Sun HC, Liu S, Cui YQ, Xu XX, 2009, A side population of cells from a human pancreatic carcinoma cell line harbors cancer stem cell characteristics. Neoplasma 56(5):371–378 Patrawala L, Calhoun T, Schneider-Broussard R, Zhou J, Claypool K, Tang DG, 2005, Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+ and ABCG2- cancer cells are similarly tumorigenic. Cancer Res 65(14):6207–6219. https:// doi.org/10.1158/0008-5472.CAN-05-0592 Burkert J, Otto WR,Wright NA, 2008, Side populations of gastrointestinal cancers are not enriched in stem cells. J Pathol 214(5): 564–573. , DOI 10.1002/path.2307 Zhou J,Wang CY, Liu T,Wu B, Zhou F, Xiong JX,Wu HS, Tao J, Zhao G, Yang M, Gou SM, 2008, Persistence of side population cells with high drug efflux capacity in pancreatic cancer. World J Gastroenterol 14(6):925–930 Ishiwata T, Matsuda Y, Yamamoto T, Uchida E, Korc M, Naito Z, 2012, Enhanced expression of fibroblast growth factor receptor 2 IIIc promotes human pancreatic cancer cell proliferation. Am J Pathol 180(5):1928–1941. , DOI 10.1016/j.ajpath.2012. 01.020 Telford WG, 2013, Stem cell identification by DyeCycle Violet side population analysis. Methods Mol Biol 946:163–179. https:// doi.org/10.1007/978-1-62703-128-8_11 Gottesman MM, Pastan I, 1993, Biochemistry of multidrug resistance mediated by the multidrug transporter. Annu Rev Biochem 62:385–427. , DOI 10.1146/annurev.bi.62.070193.002125 Mueller MT, Hermann PC, Witthauer J, Rubio-Viqueira B, Leicht SF, Huber S, Ellwart JW, Mustafa M, Bartenstein P, D'Haese JG, Schoenberg MH, Berger F, Jauch KW, Hidalgo M, Heeschen C, 2009, Combined targeted treatment to eliminate tumorigenic cancer stem cells in human pancreatic cancer. Gastroenterology 137(3): 1102–1113. , DOI 10.1053/j.gastro.2009.05.053 Eng JW, Mace TA, Sharma R, Twum DYF, Peng P, Gibbs JF, Pitoniak R, Reed CB, Abrams SI, Repasky EA, Hylander BL, 2016, Pancreatic cancer stem cells in patient pancreatic xenografts are sensitive to drozitumab, an agonistic antibody against DR5. J Immunother Cancer 4:33. , DOI 10.1186/s40425-016- 0136-y Lonardo E, Cioffi M, Sancho P, Sanchez-Ripoll Y, Trabulo SM, Dorado J, Balic A, Hidalgo M, Heeschen C, 2013, Metformin targets the metabolic achilles heel of human pancreatic cancer stem cells. PLoS One 8(10):e76518. , DOI 10.1371/journal. pone.0076518 Sancho P, Burgos-Ramos E, Tavera A, Bou Kheir T, Jagust P, Schoenhals M, Barneda D, Sellers K, Campos-Olivas R, Grana O, Viera CR, Yuneva M, Sainz B Jr, Heeschen C, 2015, MYC/ PGC-1alpha Balance Determines the Metabolic Phenotype and Plasticity of Pancreatic Cancer Stem Cells. Cell Metab 22(4):590– 605. , DOI 10.1016/j.cmet.2015.08.015 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 797 EP 805 DI 10.1007/s12253-018-0420-x PG 9 ER PT J AU Guo, P Chen, W Li, H Li, M Li, L AF Guo, Pingping Chen, Wenqi Li, Huiyu Li, Meiying Li, Lisha TI The Histone Acetylation Modifications of Breast Cancer and their Therapeutic Implications SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Breast cancer; Histone acetylation; Histone deacetylation; Therapy ID Breast cancer; Histone acetylation; Histone deacetylation; Therapy AB The histone acetylation modifications (HAMs) influence a large number of cellular functions. They are mediated through histone acetyltransferase (HAT) and histone deacetylase (HDAC). Nowadays, people have realized that HAMs are crucial for development and prognosis of breast cancer. Investigations about abnormal HAMs in breast cancer focus on initiating molecular mechanisms in breast cancer development, identification of new biomarkers to predict breast cancer aggressiveness and the therapeutic potential. As HAMs are reversible, breast cancer may be treated by restoring HAMs to normal levels. Indeed, some HDAC inhibitors have been approved by the US Food and Drug Administration to treat certain cancers. Furthermore, HAT inhibitors, HAT activators and HDAC activators may also be used as drugs to treat breast cancer. C1 [Guo, Pingping] Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of Pathobiology, 130021 Changchun, China. [Chen, Wenqi] Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of Pathobiology, 130021 Changchun, China. [Li, Huiyu] Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of Pathobiology, 130021 Changchun, China. [Li, Meiying] Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of Pathobiology, 130021 Changchun, China. [Li, Lisha] Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of Pathobiology, 130021 Changchun, China. RP Li, L (reprint author), Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of Pathobiology, 130021 Changchun, China. EM 43973966@qq.com CR Santos GC Jr, da Silva AP, Feldman L, Ventura GM, Vassetzky Y, de Moura Gallo CV, 2015, Epigenetic modifications, chromatin distribution and TP53 transcription in a model of breast cancer progression. J Cell Biochem 116(4):533–541. , DOI 10. 1002/jcb.25003 Thomas S, Thurn KT, Raha P, Chen S, Munster PN, 2013, Efficacy of Histone Deacetylase and Estrogen Receptor Inhibition in Breast Cancer Cells Due to Concerted down Regulation of Akt. PloS one 8(7). 10.1371/journal.pone.0068973 Kochan DZ, Kovalchuk O, 2015, Circadian disruption and breast cancer: an epigenetic link? Oncotarget 6(19):16866–16882. https:// doi.org/10.18632/oncotarget.4343 Rizzolo P, Silvestri V, Tommasi S, Pinto R, Danza K, Falchetti M, Gulino M, Frati P, Ottini L, 2013, Male breast cancer: genetics, epigenetics, and ethical aspects. Ann Oncol: official journal of the European Society forMedical Oncology/ESMO 24 Suppl 8:viii75- viii82., DOI , DOI 10.1093/annonc/mdt316 Ye Q, Holowatyj A, Wu J, Liu H, Zhang LH, Suzuki T, Yang ZQ, 2015, Genetic alterations of KDM4 subfamily and therapeutic effect of novel demethylase inhibitor in breast cancer. In: American Journal of Cancer Research 5, 4):1519−+ Yokoyama Y, Matsumoto A, Hieda M, Shinchi Y, Ogihara E, Hamada M, Nishioka Y, Kimura H, Yoshidome K, Tsujimoto M, Matsuura N, 2014, Loss of histone H4K20 trimethylation predicts poor prognosis in breast cancer and is associated with invasive activity. Breast Cancer Research 16(3). , DOI 10.1186/ Bcr3681 Cava C, Bertoli G, Castiglioni I, 2015, Integrating genetics and epigenetics in breast cancer: biological insights, experimental, computational methods and therapeutic potential, BMC Systems Biology 9. , DOI 10.1186/s12918-015-0211-x Britten A, Rossier C, TarightN, Ezra P, Bourgier C, 2013, Genomic classifications and radiotherapy for breast cancer. Eur J Pharmacol 717(1-3):67-70. , DOI 10.1016/j.ejphar.2012.11.069 Nagasawa S, Sedukhina AS, Nakagawa Y, Maeda I, Kubota M, Ohnuma S, Tsugawa K, Ohta T, Roche-Molina M, Bernal JA, Narvaez AJ, Jeyasekharan AD, Sato K, 2015, LSD1 Overexpression Is Associated with Poor Prognosis in Basal-Like Breast Cancer, and Sensitivity to PARP Inhibition. PloS one 10(2). , DOI 10.1371/journal.pone.0118002 Connolly R, Stearns V, 2012, Epigenetics as a Therapeutic Target in Breast Cancer. J Mammary Gland Biol Neoplasia 17(3-4):191- 204. , DOI 10.1007/s10911-012-9263-3 Schneider A, Chatterjee S, Bousiges O, Selvi BR, Swaminathan A, Cassel R, Blanc F, Kundu TK, Boutillier AL, 2013, Acetyltransferases, HATs, as Targets for Neurological Therapeutics. Neurotherapeutics: the journal of the American Society for Experimental NeuroTherapeutics 10(4):568-588. , DOI 10.1007/s13311-013-0204-7 Khan SI, Aumsuwan P, Khan IA, Walker LA, Dasmahapatra AK, 2012, Epigenetic Events Associated with Breast Cancer and Their Prevention by Dietary Components Targeting the Epigenome. Chem Res Toxicol 25(1):61-73. , DOI 10.1021/tx200378c Karsli-Ceppioglu S, Dagdemir A, Judes G, Ngollo M, Penault- Llorca F, Pajon A, Bignon YJ, Bernard-Gallon D, 2014, Epigenetic mechanisms of breast cancer: an update of the current knowledge. Epigenomics 6(6):651-664. , DOI 10.2217/ Epi.14.59 Gajer JM, Furdas SD, Grunder A, Gothwal M, Heinicke U, Keller K, Colland F, Fulda S, Pahl H, Fichtner I, Sippl W, Jung M, 2015, Histone acetyltransferase inhibitors block neuroblastoma cell growth in vivo. Oncogenesis 4. , DOI 10.1038/oncsis. 2014.51 Jovanovic J, Ronneberg JA, Tost J, Kristensen V, 2010, The epigenetics of breast cancer. Mol Oncol 4(3):242-254. , DOI 10.1016/j.molonc.2010.04.002 Seo J, Min SK, Park HR, Kim DH, Kwon MJ, Kim LS, Ju YS, 2014, Expression of Histone Deacetylases HDAC1, HDAC2, HDAC3, and HDAC6 in Invasive Ductal Carcinomas of the Breast. J Breast Cancer 17(4):323-331. , DOI 10.4048/ jbc.2014.17.4.323 Muller BM, Jana L, Kasajima A, Lehmann A, Prinzler J, Budczies J,Winzer KJ, Dietel M,WeichertW, Denkert C, 2013, Differential expression of histone deacetylases HDAC1, 2 and 3 in human breast cancer - overexpression of HDAC2 and HDAC3 is associated with clinicopathological indicators of disease progression. BMC Cancer 13. , DOI 10.1186/1471-2407-13-215 Mawatari T, Ninomiya I, Inokuchi M,Harada S, HayashiH, Oyama K, Makino I, Nakagawara H, Miyashita T, Tajima H, Takamura H, Fushida S, Ohta T, 2015, Valproic acid inhibits proliferation of HER2-expressing breast cancer cells by inducing cell cycle arrest and apoptosis through Hsp70 acetylation. Int J Oncol 47(6):2073- 2081. , DOI 10.3892/ijo.2015.3213 Falahi F, van Kruchten M, Martinet N, Hospers GAP, Rots MG, 2014, Current and upcoming approaches to exploit the reversibility of epigeneticmutations in breast cancer. Breast Cancer Research 16(4). , DOI 10.1186/S13058-014-0412-Z Messier TL, Gordon JAR, Boyd JR, Tye CE, Browne G, Stein JL, Lian JB, Stein GS, 2016, Histone H3 lysine 4 acetylation and methylation dynamics define breast cancer subtypes. Oncotarget 7(5):5094-5109 Basse C, Arock M, 2015, The increasing roles of epigenetics in breast cancer: Implications for pathogenicity, biomarkers, prevention and treatment. Int J Cancer 137(12):2785-2794. , DOI 10.1002/ijc.29347 FermentoME, GandiniNA, LangCA, Perez JE,Maturi HV, Curino AC, Facchinetti MM, 2010, Intracellular distribution of p300 and its differential recruitment to aggresomes in breast cancer, Retracted article. See vol. 94, pg. 418, 2013). Exp Mol Pathol 88, 2):256-264., DOI , DOI 10.1016/j.yexmp.2010.01.007 Yang H, Pinello CE, Luo J, Li DW, Wang YF, Zhao LY, Jahn SC, Saldanha SA, Planck J, Geary KR, Ma HC, Law BK, Roush WR, Hodder P, Liao DQ, 2013, Small-Molecule Inhibitors of Acetyltransferase p300 Identified by High-Throughput Screening Are Potent Anticancer Agents. Mol Cancer Ther 12(5):610-620. , DOI 10.1158/1535-7163.MCT-12-0930 Liu ZP, Luo XG, Liu L, ZhaoWW, Guo S, Guo Y,Wang N, He HP, Liao XH, Ma WJ, Zhou H, Zhang TC, 2013, Histone acetyltransferase p300 promotes MKL1-mediated transactivation of catechol- O-methyltransferase gene. Acta BiochimBiophys Sin 45(12):1002- 1010. , DOI 10.1093/abbs/gmt108 Derr RS, van Hoesel AQ, Benard A, Goossens-Beumer IJ, Sajet A, Dekker-Ensink NG, de Kruijf EM, Bastiaannet E, VTHBM S, van de Velde CJH, Kuppen PJK, 2014, High nuclear expression levels of histone-modifying enzymes LSD1, HDAC2 and SIRT1 in tumor cells correlate with decreased survival and increased relapse in breast cancer patients. BMC Cancer 14. , DOI 10.1186/ 1471-2407-14-604 Hervouet E, Claude-Taupin A, Gauthier T, Perez V, Fraichard A, Adami P, Despouy G,Monnien F, AlgrosMP, Jouvenot M, Delage- Mourroux R, Boyer-Guittaut M, 2015, The autophagy GABARAPL1 gene is epigenetically regulated in breast cancer models. BMC Cancer 15. , DOI 10.1186/S12885-015- 1761-4 Ray A, Alalem M, Ray BK, 2013, Loss of Epigenetic Kruppel-like Factor 4 Histone Deacetylase, KLF-4-HDAC)-mediated Transcriptional Suppression Is Crucial in Increasing Vascular Endothelial Growth Factor, VEGF, Expression in Breast Cancer. J Biol Chem 288(38):27232-27242. , DOI 10.1074/jbc. M113.481184 Gong C, Qu SH, Lv XB, Liu BD, Tan WG, Nie Y, Su FX, Liu Q, Yao HR, Song EW, 2014, BRMS1L suppresses breast cancer metastasis by inducing epigenetic silence of FZD10. Nature communications 5. , DOI 10.1038/Ncomms6406 Cai FF, Kohler C, Zhang B, Wang MH, Chen WJ, Zhong XY, 2011, Epigenetic Therapy for Breast Cancer. Int J Mol Sci 12(7): 4465-4476. , DOI 10.3390/ijms12074465 Secci D, Carradori S, Bizzarri B, Bolasco A, Ballario P, Patramani Z, Fragapane P, Vernarecci S, Canzonetta C, Filetici P, 2014, Synthesis of a novel series of thiazole-based histone acetyltransferase inhibitors. Bioorg Med Chem 22(5):1680-1689. , DOI 10.1016/j.bmc.2014.01.022 Xu LX, Li ZH, Tao YF, Li RH, Fang F, Zhao H, Li G, Li YH,Wang J, Feng X, Pan J, 2014, Histone acetyltransferase inhibitor II induces apoptosis in glioma cell lines via the p53 signaling pathway. J Exp Clin Canc Res 33. , DOI 10.1186/S13046-014-0108-3 Oike T, Komachi M, Ogiwara H,Amornwichet N, SaitohY, Torikai K, Kubo N, Nakano T, Kohno T, 2014, C646, a selective small molecule inhibitor of histone acetyltransferase p300, radiosensitizes lung cancer cells by enhancing mitotic catastrophe. Radiother Oncol 111(2):222-227. , DOI 10.1016/j.radonc.2014.03. 015 Lee YH, Kwak J, Choi HK, Choi KC, Kim S, Lee J, Jun W, Park HJ, Yoon HG, 2012, EGCG suppresses prostate cancer cell growth modulating acetylation of androgen receptor by anti-histone acetyltransferase activity. Int J Mol Med 30(1):69-74. , DOI 10. 3892/ijmm.2012.966 Gao CX, Bourke E, Scobie M, Famme MA, Koolmeister T, Helleday T, Eriksson LA, Lowndes NF, Brown JAL, 2014, Rational design and validation of a Tip60 histone acetyltransferase inhibitor. Scientific reports 4. , DOI 10.1038/Srep05372 Ye X, Yuan L, Zhang L, Zhao J, Zhang CM, Deng HY, 2014, Garcinol, an acetyltransferase inhibitor, suppresses proliferation of breast cancer cell line MCF-7 promoted by 17beta-estradiol. Asian Pacific journal of cancer prevention : APJCP 15(12):5001-5007 Federico M, Bagella L, 2011, Histone Deacetylase Inhibitors in the Treatment of Hematological Malignancies and Solid Tumors. Journal of Biomedicine and Biotechnology. , DOI 10. 1155/2011/475641 Contreras-Leal E, Hernandez-Oliveras A, Flores-Peredo L, Zarain- Herzberg A, Santiago-Garcia J, 2015, Histone deacetylase inhibitors promote the expression of ATP2A3 gene in breast cancer cell lines. Mol Carcinog. , DOI 10.1002/mc.22402 Lavoie R, Bouchain G, Frechette S, Woo SH, Abou Khalil E, Leit S, Fournel M, Yan PT, Trachy-Bourget MC, Beaulieu C, Li ZM, Besterman J, Delorme D, 2001, Design and synthesis of a novel class of histone deacetylase inhibitors. Bioorg Med Chem Lett 11(21):2847-2850. , DOI 10.1016/S0960-894x(01)00552- 2 Zhang T, Chen YH, Li JJ, Yang FF, Wu HG, Dai FJ, Hu MC, Lu XL, Peng Y, Liu MY, Zhao YX, Yi ZF, 2014, Antitumor Action of a Novel Histone Deacetylase Inhibitor, YF479, in Breast Cancer. Neoplasia 16(8):665-677. , DOI 10.1016/j.neo.2014.07. 009 Chatterjee N, Wang WLW, Conklin T, Chittur S, Tenniswood M, 2013, Histone deacetylase inhibitors modulate miRNA and mRNA expression, block metaphase, and induce apoptosis in inflammatory breast cancer cells. Cancer Biology & Therapy 14(7): 658-671. , DOI 10.4161/cbt.25088 Li LP, Sun YX, Liu JQ, Wu XD, Chen LJ, Ma L, Wu PF, 2015, Histone deacetylase inhibitor sodium butyrate suppresses DNA double strand break repair induced by etoposide more effectively in MCF-7 cells than in HEK293 cells. BMC biochemistry 16. , DOI 10.1186/S12858-014-0030-5Z Lee J, Bartholomeusz C, Mansour O, Humphries J, Hortobagyi GN, Ordentlich P, Ueno NT, 2014, A class I histone deacetylase inhibitor, entinostat, enhances lapatinib efficacy in HER2- overexpressing breast cancer cells through FOXO3-mediated Bim1 expression. Breast Cancer Res Treat 146(2):259-272. , DOI 10.1007/s10549-014-3014-7 ChiuHW, Yeh YL,Wang YC, Huang WJ, Chen YA, Chiou YS, Ho SY, Lin P, Wang YJ, 2013, Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, enhances radiosensitivity and suppresses lung metastasis in breast cancer in vitro and in vivo. PLoS One 8(10):e76340. , DOI 10.1371/journal.pone.0076340 Cody JJ, Markert JM, Hurst DR, 2014, Histone deacetylase inhibitors improve the replication of oncolytic herpes simplex virus in breast cancer cells. PLoS One 9(3):e92919. , DOI 10.1371/ journal.pone.0092919 Raha P, Thomas S, Thurn KT, Park J, Munster PN, 2015, Combined histone deacetylase inhibition and tamoxifen induces apoptosis in tamoxifen-resistant breast cancer models, by reversing Bcl-2 overexpression. Breast cancer research: BCR 17:26. https:// doi.org/10.1186/s13058-015-0533-z Min A, Im SA, Kim DK, Song SH, Kim HJ, Lee KH, Kim TY, Han SW, Oh DY, Kim TY, O'Connor MJ, Bang YJ, 2015, Histone deacetylase inhibitor, suberoylanilide hydroxamic acid, SAHA), enhances anti-tumor effects of the poly, ADP-ribose, polymerase, PARP, inhibitor olaparib in triple-negative breast cancer cells. Breast Cancer Research 17. , DOI 10.1186/s13058-015- 0534-y SalvadorMA,Wicinski J, Cabaud O, Toiron Y, Finetti P, Josselin E, Lelievre H, Kraus-Berthier L, Depil S, Bertucci F, Collette Y, Birnbaum D, Charafe-Jauffret E, Ginestier C, 2013, The Histone Deacetylase Inhibitor Abexinostat Induces Cancer Stem Cells Differentiation in Breast Cancer with Low Xist Expression. Clin Cancer Res 19(23):6520-6531. , DOI 10.1158/1078-0432. CCR-13-0877 Kubo M, Kanaya N, Petrossian K, Ye JJ, Warden C, Liu Z, Nishimura R, Osako T, Okido M, Shimada K, Takahashi M, Chu PG, Yuan YC, Chen SA, 2013, Inhibition of the proliferation of acquired aromatase inhibitor-resistant breast cancer cells by histone deacetylase inhibitor LBH589, panobinostat). Breast Cancer Res Treat 137(1):93-107. , DOI 10.1007/s10549-012-2332-x Dastjerdi MN, Salahshoor MR, Mardani M, Hashemibeni B, Roshankhah S, 2013, The effect of CTB on P53 protein acetylation and consequence apoptosis on MCF-7 and MRC-5 cell lines. Advanced biomedical research 2:24. , DOI 10.4103/ 2277-9175.108005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 807 EP 813 DI 10.1007/s12253-018-0433-5 PG 7 ER PT J AU Kelly, EM Mohan, MH Baird, WA Ryan, JE Winter, CD AF Kelly, E Michael Mohan, M Helen Baird, W Alan Ryan, J Elizabeth Winter, C Des TI Orphan Nuclear Receptors in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Orphan nuclear receptors; Colorectal cancer; Carcinogenesis; Signaling pathways; Nuclear receptors ID Orphan nuclear receptors; Colorectal cancer; Carcinogenesis; Signaling pathways; Nuclear receptors AB Colorectal cancer is one of the most common cancers worldwide, with an overall increased incidence annually. Despite improvements in treatment and surveillance, almost 50% develop recurrent and/or distant disease. Unknown cellular processes are the fundamental cause for treatment failure and metastatic disease. The interplay of chronic inflammation and carcinogenesis is well established. Recent work has highlighted the role of nuclear receptors and co-regulators in the inflammation to carcinogenesis process. Orphan nuclear receptors have been shown to be involved in numerous cellular processes, including both at a transcriptional and a non-genomic level. There is a significant emphasis to identify ligands that will interact and modify these nuclear receptors, with the long-term aim of developing novel pharmaceutical therapies. The identification of orphan nuclear receptor ligands will also help increase our current understanding of their role in cellular signaling, by enabling manipulation of these receptors. This review aims to provide a brief overview of some key orphan nuclear receptors which may be involved in colorectal cancer. C1 [Kelly, E Michael] University College Dublin, UCD Veterinary Sciences CentreDublin, Ireland. [Mohan, M Helen] University College Dublin, UCD Veterinary Sciences CentreDublin, Ireland. [Baird, W Alan] University College Dublin, UCD Veterinary Sciences CentreDublin, Ireland. [Ryan, J Elizabeth] St Vincent’s University Hospital, Centre for Colorectal Disease, Elm ParkDublin, Ireland. [Winter, C Des] St Vincent’s University Hospital, Centre for Colorectal Disease, Elm ParkDublin, Ireland. RP Kelly, EM (reprint author), University College Dublin, UCD Veterinary Sciences Centre, Dublin, Ireland. EM Kellym11@tcd.ie CR National Cancer Intelligence Network Data Briefing, 2009. Colorectal Cancer Survival by Stage. [online] Available at http:// www.ncin.org.uk/publications/data_briefings/colorectal_cancer_ survival_by_stage.aspx Rizzo A, Pallone F, Monteleone G, Fantini MC, 2011, Intestinal inflammation and colorectal cancer: a double edged sword?World J Gastroenterol 17:3092–3100 Riggins RB, Mazzotta MM, Maniya OZ, Clarke R, 2010, Orphan nuclear receptors in breast cancer pathogenesis and therapeutic response. Endocr Relat Cancer 17(3):213–231 Han YF, Cao GW, 2012, Role of nuclear receptor NR4A2 in gastrointestinal inflammation and cancers. World J Gastroenterol 18(47):6865–6873 Balkwill F, Mantovani A, 2001, Inflammation and cancer: back to Virchow? Lancet 357:539–545 Lee JS, Kim KI, Baek SH, 2008, Nuclear receptors and coregulators in inflammation and cancer. Cancer Lett 267:189–196 Safe S, Kim K, Li X, Lee SO, 2011, NR4A orphan receptors and cancer. Nucl Recept Signal 9:1–14 Evans RM(2005, The nuclear receptor superfamily: a rosetta stone for physiology. Mol Endocrinol 19:1429–1438 Novac N, Heinzel T, 2004, Nuclear receptors: overview and classification. Curr Drug Targets Inflamm Allergy 3:335–346 Robinson-Rechavi M, Garcia HE, Laudet V, 2003, The nuclear receptor superfamily. J Cell Sci 116:585–586 Zhao Y, Bruemmer D, 2010, NR4A Orphan Nuclear Receptors: Transcriptional regulators of gene expression in metabolism and vascular biology. Arterioscler Thromb Vasc Biol 30(8):1535–1541 Benoit G, Malewicz M, Perlmann T, 2009, Digging deep into the pockets of orphan nuclear receptors: insights from structural studies. Trends Cell Biol Chem 284:23286–23292 Gallastegui N, Mackinnon JA, Fletterick RJ, Estebanez-Perpina E, 2015, Advances in our structural understanding of orphan nuclear receptors.Trends Biochem Sci 2015;40(1):25–35 Wu H, Lin Y, LIW, Sun Z, GaoW, Zhang H, Xie L, Jiang F, Qin B, Yan T, Chen L, Zhao Y, Cao X, Wu Y, Lin B, Zhou H, Wong AS, Zhang XK, Zeng JZ, 2010, Regulation of Nurr 77 expression by beta-catenin and itsmitogenic effect on colon cancer cells. FASEB J 25:192–205 Shi Y, 2007, Orphan nuclear receptors in drug discovery. Drug Discov Today 12:440–445 Giguere V, Yang N, Segui P, Evans RM, 1988, Identification of new class of steroid hormone receptor. Nature 331:91–94 Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM, 2006, Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell 126:789–799 Claessens F, Gewirth DT, 2004, DNA recognition by nuclear receptors. Essays Biochem 40:59–72 Freedman LP, Luisi BF, Korszun ZR, Basavappa R, Sigler PB, Yamamoto KR, 1988, The function and structure of the metal coordination sites within the glucocorticoid receptor DNA binding domain. Nature 334:543–546 Zelcer N, Tontonoz P, 2006, Liver X receptors as integrators of metabolic and inflammatory signaling. J Clin Invest 116:607–614 Blumberg B, Evans RM, 1998, Orphan nuclear receptors – new ligands and new possibilities. Genes Dev 12:3149–3155 Ordentlich P, 2003, Identification of the antineoplastic agent 6- mercaptopurine as an activator of the orphan nuclear hormone receptor Nurr 1. J Biol Chem 278:24791–24799 Enmark E, Gustafsson JA, 1996, Orphan nuclear receptors – the first eight years. Mol Endocrinol 10:1293–1307 Boute N et al, 2002, The use of resonance energy transfer in highthroughput screening. BRET versus FRET. Trend Pharmacol 23: 351–354 Xu HE et al, 2002, Structural basis for antagonist-mediated recruitment of nuclear co-repressors by PPARalpha. Nature 415:813–817 Mohan HM, Aherne CM, Rogers AC, Baird AW,Winter DC, Murphy EP, 2012, Molecular pathways: The role of NR4A Orphan Nuclear Receptors in Cancer. Clin Cancer Res 18:3223–3228 Hanahan D, Weinberg RA, 2000, Cell 100:57–70 Martinez-Gonzalez J, Badimon L, 2005, The NR4A subfamily of nuclear receptors: new early genes regulated by growth factors in vascular cells. Cardiovasc Res 65:609–618 MaxwellMA,Muscat GE, 2006, The NR4A subgroup: immediate early response genes with pleiotropic physiological roles. Nucl Recept Signal 4:e002 Collins D, Hogan AM,Winter DC, 2011, Microbial and viral pathogens in colorectal cancer. Lancet Oncol 12(5):504–512 Hogan AM, Kennelly R, Collins D et al, 2009, Oestrogen inhibits human colonic motility by a non-genomic cell membrane receptordependent mechanism. Br J Surg 96(7):817–822 McMorrow JP, Murphy EP, 2011, Inflammation: a role for NR4A orphan nuclear receptors? Biochem Soc Trans 39(2):688–693 Holla VR, Wu H, Shi Q, Menter DG, Dubois RN, 2011, Nuclear orphan receptor NR4A2; Modulates fatty acid oxidation pathways in colorectal cancer. J Biol Chem 286(34):30003–30009 Han Y, Cai H,Ma L, DingY, Tan X, Liu Y, Su T, Yu Y, ChangW, Zhang H, Fu C, Cao G Nuclear orphan receptor NR4A2 confers chemoresistance and predicts unfavorable prognosis of colorectal carcinoma patients who received post-operative chemotherapy. Eur. J. Cancer. 2013; Jun 25. pii: S0959–8049(13)00461–9 Li X, Tai HH, 2009, Activation of thromboxane A2 receptors induces orphan nuclear receptors Nurr 1 expression and stimulates cell proliferation in human lung cancer cells. Carcinogenesis 30: 1606–1613 Holla VR, Mann JR, Shi Q, Dubois RN, 2006, Prostaglandin E2 regulates the nuclear receptor NR4A2 in colorectal cancer. J Biol Chem 281(5):2676–2268 Palorini R, De Rasmo D, Gaviraghi M, Danna LS, Signorile A, Cirulli C, Chiaradonna F, Alberghina L, Papa S. Oncogenic K-ras expression is associated with derangement of the cAMP/PKA pathway and forskolin-reversible alterations of mitochondrial dynamics and respiration. Oncogene 2012; 32(3):352–362 Leone V, di Palma A, Ricchi P, Acquaviva F, Giannouli M, Di Prisco AM, Iuliano F, Acquaviva AM, 2007, PGE2 inhibits apoptosis in human adenocarcinoma Caco-2 cell line through Ras- PI3K association and cAMP-dependant kinase A activation. Am J Physiol Gastrointest Liver Physiol 293:673–681 Zagani R, Hamzaoui N, Cacheux W, de Reynies A, Terris B, Chaussade S, Romagnolo B, Perret C, Lamarque D, 2009, Cyclooxygenase-2 inhibitors down-regulate osteopontin and NR4A2 new therapeutic targets for colorectal cancers. Gastroenterology 137:1358–1366 Takamoto N, You LR, Moses K, Chiang C, ZimmerWE, Schwartz RJ, DeMayo FJ, Tsai MJ, Tsai SY, 2005, COUP-TFII is essential for radial and anteroposterior patterning of the stomach. Development 132:2179–2189 Hamers AA, van Dam L, Teixeira Duarte JM et al, 2015, Defiency of nuclear receptors Nurr77 aggravates mouse experimental colitis by increased NFκB activity in marcophages. PLoS One 4;10(8): e0133598. , DOI 10.1371/journal.pone.0133598 Liang R, Lin Y, Yuan CL, Liu ZH, Li YQ, Luo XL, Ye JZ, Ye HH, 2018, High expression of estrogen-related receptor α is significantly associated with poor prognosis in patients with colorectal cancer. Oncol Lett 15(4):5933–5939 Xiao L,Wang J, Li J, Chen X, Xu P, Sun S, He D, Cong Y, Zhai Y, 2015, RORα inhibits adipocyte-conditioned medium-induced colorectal cancer cell proliferation and migration and chick embryo chorioallantoic membrane angiopoiesis. Am J Physiol Cell Physiol. 308(5):C385–96 Berman AY, Manna S, Schwartz NS, Katz YE, Sun Y, Behrmann CA, Yu JJ, Plas DR, Alayev A, Holz MK, 2017, ERRα regulates the growth of triple-negative breast cancer cells via S6K1-dependent mechanism. Signal Transduct Target Ther 2:e17035 Shin D, Kim IS, Lee JM, Shin SY, Lee JH, Baek SH, Cho KH, 2014, The hidden switches underlying RORα-mediated circuits that critically regulate uncontrolled cell proliferation. J Mol Cell Biol 6(4):338–348 Wen Z, Pan T, Yang S, Liu J, Tao H, Zhao Y, Xu D, ShaoW,Wu J, Liu X, Wang Y, Mao J, Zhu Y, 2017, Up-regulated NRIP2 in colorectal cancer initiating cells modulates the Wnt pathway by targeting RORβ. Mol Cancer 16(1):20 Wang C, Zhou Y, Ruan R, Zheng M, Han W, Liao L, 2015, High expression of COUP-TF II cooperated with negative Smad4 expression predicts poor prognosis in patients with colorectal cancer. Int J Clin Exp Pathol 8(6):7112–7121 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 815 EP 819 DI 10.1007/s12253-018-0440-6 PG 5 ER PT J AU Mandl, J Banhegyi, G AF Mandl, Jozsef Banhegyi, Gabor TI The ER – Glycogen Particle – Phagophore Triangle: A Hub Connecting Glycogenolysis and Glycophagy? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Glycogen particle; Glycogenolysis; Endoplasmic reticulum; Glycophagy; Lysosome; Phagophore ID Glycogen particle; Glycogenolysis; Endoplasmic reticulum; Glycophagy; Lysosome; Phagophore AB Glycogen particle is an intracellular organelle, which serves as a carbohydrate reserve in various cells. The function of glycogen is not entirely known in several cell types. Glycogen can be mobilized for different purposes, which can be related to cellular metabolic needs, intracellular redox state, metabolic state of the whole organism depending on regulatory aspects and also on cell functions. Essentially there are two different ways of glycogen degradation localized in different cellular organelles: glycogenolysis or lysosomal breakdown by acid alpha-glucosidase. While glycogenolysis occurs in glycogen particles connected to endoplasmic reticulum membrane, glycogen particles can be also combined with phagophores forming autophagosomes. A subdomain of the endoplasmic reticulum membrane - omegasomes - are the sites for phagophore formation. Thus, three organelles, the endoplasmic reticulum, the phagophore and the glycogen particle forms a triangle in which glycogen degradation occurs. The physiological significance, molecular logic and regulation of the two different catabolic paths are summarized and discussed with special aspect on the role of glycogen particles in intracellular organelle homeostasis and on molecular pathology of the cell. Pathological aspects and some diseases connected to the two different degradation pathways of glycogen particles are also detailed. C1 [Mandl, Jozsef] Semmelweis University, Department of Medical Chemistry, Molecular Biology and PathobiochemistryBudapest, Hungary. [Banhegyi, Gabor] Semmelweis University, Department of Medical Chemistry, Molecular Biology and PathobiochemistryBudapest, Hungary. RP Mandl, J (reprint author), Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Budapest, Hungary. EM mandl.jozsef@med.semmelweis-univ.hu CR Adeva-Andany MM, Gonzalez-Lucan M, Donapetry-Garcia C, Fernandez-Fernandez C, Ameneiros-Rodriguez E, 2016, Glycogen metabolism in humans. Biochem Biophys Acta Clin 5: 85–100 Duran J, Guinovart JJ, 2015, Brain glycogen in health and disease. Mol Asp Med 46:70–77 Csordas G, Varnai P, Golenar T, Roy S, Purkins G, Schneider TG, Balla T, Hajnoczky G, 2010, Imaging interorganelle contacts and local calcium dynamics at the ER-mitochondrial interface. Mol Cell 39:121–132 de Brito OM, Scorrano L, 2010, An intimate liaison: spatial organization of the endoplasmic reticulum-mitochondria relationship. EMBO J 29:2715–2723 Banhegyi G, Mandl J, 2001, The hepatic glycogenoreticular system. POR 7:107–110 Stapleton D, Nelson C, Parsawar K,McClain D, Gilbert-Wilson R, Barker E, Rudd B, Brown K, Hendrix W, O'Donnell P, Parker G, 2010, Analysis of hepatic glycogen-associated proteins. Proteomics 10:2320–2329 Mandl J, Meszaros T, Banhegyi G, Csala M, 2013, Minireview: Endoplasmic reticulum stress: Control in protein, lipid, and signal homeostasis. Mol Endocrinol 27:384–393 Csala M, Kereszturi E, Mandl J, Banhegyi G, 2012, The endoplasmic reticulum as the extracellular space inside the cell: role in protein folding and glycosylation. Antioxid Redox Signal 16:1100– 1108 Fawcett DW, 1955, Observations on the cytology and electron microscopy of hepatic cells. J Natl Cancer Inst 15:1457–1503 Cardell RR Jr, 1977, Smooth endoplasmic reticulum in rat hepatocytes during glycogen deposition and depletion. Int Rev Cytol 48: 221–279 Banhegyi G, Garzo T, Antoni F, Mandl J, 1988, Glycogenolysis – and not gluconeogenesis – is the source of UDP-glucuronic acid for glucuronidation. Biochim Biophys Acta 967:429–435 Mandl J, Banhegyi G, Kalapos M, Garzo T, 1995, Increased oxidation and decreased conjugation of drugs in the liver caused by starvation., review, Chem. Biol. Interactions 96:87–101 Helmika W, Wever R, 1997, A new model for the membrane topology of glucose-6-phosphatase: the enzyme involved in von Gierke disease. FEBS Lett 409:317–319 Pan CJ, Lei KJ, Annabi B et al, 1998, Transmembrane topology of glucose-6-phosphatase. J Biol Chem 273:6144–6148 Burchell G, Coughtrie MW, 1989, UDP-glucuronosyltransferases. Pharmacol Ther 43:261–289 Clarke DJ, Burchell G, 1994, Conjugation-Deconjugation reactions in Drug Metabolism and Toxicity. In: Kauffman FC, ed, Handbook of Experimental Pharmacology, vol 112. Springer Verlag, Budapest, pp 3–43 Braun L, Garzo T, Mandl J, Banhegyi G, 1994, Ascorbic acid synthesis is stimulated by enhanced glycogenolysis in murine liver. FEBS Lett 352:4–6 Braun L, CsalaM, PoussuA, Garzo T,Mandl J, Banhegyi G, 1996, Glutathione depletion induces glycogenolysis dependent ascorbate synthesis in isolated murine hepatocytes. FEBS Lett 388:173–176 Banhegyi G, Braun L, Csala M, Puskas F, Mandl J, 1997, Ascorbate metabolism and its regulation in animals. Free Radic Biol Med 23:793–803 Puskas F, Braun L, CsalaM, Kardon T, Marcolongo P, Benedetti A, Mandl J, Banhegyi G, 1998, Gulonolactone oxidase activitydependent intravesicular glutathione oxidation in rat liver microsomes. FEBS Lett 430:293–296 Dzyakanchuk AA, Balazs Z, Nashev LG, Amrein KE, Odermatt A, 2009, 11beta-Hydroxysteroid dehydrogenase 1 reductase activity is dependent on a high ratio of NADPH/NADP(+, and is stimulated by extracellular glucose. Mol Cell Endocrinol 301:137–141 Kereszturi E, Kalman FS, Kardon T, Csala M, Banhegyi G, 2010, Decreased prereceptorial glucocorticoid activating capacity in starvation due to an oxidative shift of pyridine nucleotides in the endoplasmic reticulum. FEBS Lett 584:4703–4708 Stapleton D, Nelson C, Parsawar K, Flores-Opazo M, McClain D, Parker G, 2013, The 3T3-L1 adipocyte glycogen proteome. Proteome Sci 22:11 Jiang S, Heller B, Tagliabracci VS, Zhai L, Irimia JM, DePaoli- Roach AA, Wells CD, Skurat AV, Roach PJ, 2010, Starch binding domain-containing protein 1/genethonin 1 is a novel participant in glycogen metabolism. J Biol Chem 285:34960–34971 Jiang S, Wells CD, Roach PJ, 2011, Starch-binding domain-containing protein 1, Stbd1, and glycogen metabolism: Identification of the Atg8 family interacting motif, AIM, in Stbd1 required for interaction with GABARAPL1. Biochem Biophys Res Commun 413:420–425 Rong Y, McPhee CK, Deng S, Huang L, Chen L, Liu M, Tracy K, Baehrecke EH, Yu L, Lenardo MJ, 2011, Spinster is required for autophagic lysosome reformation and mTOR reactivation following starvation. Proc Natl Acad Sci 108:7826–7831 Kotoulas OB, Phillips MJ, 1971, Fine structural aspects of the mobilization of hepatic glycogen. I. Acceleration of glycogen breakdown. Am J Pathol 63:1–22 Schiaffino S, Hanzlikova V, 1972, Autophagic degradation of glycogen in skeletal muscles of the newborn rat. J Cell Biol 52:41–51 Kotoulas OB, Kalamidas SA, Kondomerkos DJ, 2006, Glycogen autophagy in glucose homeostasis. Pathol Res Pract 202:631–638 Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H, Yoshimori T, Ohsumi Y, Tokuhisa T,Mizushima N, 2004, The role of autophagy during the early neonatal starvation period. Nature 432:1032–1036 KomatsuM,Waguri S, Ueno T, Iwata J,Murata S, Tanida I, Ezaki J, Mizushima N, Ohsumi Y, Uchiyama Y, Kominami E, Tanaka K, Chiba T, 2005, Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol 169:425–434 Kalamidas SA, Kotoulas OB, 2000, Glycogen autophagy in newborn rat hepatocytes. Histol Histopathol 15:1011–1018 Kondomerkos DJ, Kalamidas SA, Kotoulas OB, Hann AC, 2005, Glycogen autophagy in the liver and heart of newborn rats. The effects of glucagon, adrenalin or rapamycin. Histol Histopathol 20:689–696 Mellor KM, Varma U, Stapleton DI, Delbridge LM, 2014, Cardiomyocyte glycophagy is regulated by insulin and exposure to high extracellular glucose. Am J Physiol Heart Circ Physiol 306: H1240–H1245 Ravikumar B, Stewart A, Kita H, Kato K, Duden R, Rubinsztein DC, 2003, Raised intracellular glucose concentrations reduce aggregation and cell death caused by mutant huntingtin exon 1 by decreasing mTOR phosphorylation and inducing autophagy. Hum Mol Genet 12:985–994 Axe EL, Walker SA, Manifava M, Chandra P, Roderick HL, Habermann A, Griffiths G, Ktistakis NT et al, 2008, Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J Cell Biol 182:685–701 Ahn HH, Oh Y, Lee H, LeeW, Chang JW, Pyo HK, Nah do H, Jung YK, 2015, Identification of glucose-6-phosphate transporter as a key regulator functioning at the autophagy initiation step. FEBS Lett 589:2100–2109 Chatelain F, Pegorier JP, Minassian C, Bruni N, Tarpin S, Girard J, Mithieux G, 1998, Development and regulation of glucose-6- phosphatase gene expression in rat liver, intestine, and kidney: in vivo and in vitro studies in cultured fetal hepatocytes. Diabetes 47: 882–889 Froissart R, Piraud M, Boudjemline AM, Vianey-Saban C, Petit F, Hubert-Buron A, Eberschweiler PT, Gajdos V, Labrune P, 2011, Glucose-6-phosphatase deficiency. Orphanet J Rare Dis 6:27 Cavanagh JB, 1999, Corpora-amylacea and the family of polyglucosan diseases. Brain Res Brain Res Rev 29:265–295 Mandl J, Meszaros K, Antoni F, Spolarics Z, Garzo T, 1982, Reversible inhibition of RNA synthesis and irreversible inhibition of protein synthesis by D-galactosamine in isolated mouse hepatocytes. Mol Cell Biochem 46:25–30 Farah BL, Landau DJ, Sinha RA, Brooks ED, Wu Y, Fung SY, Tanaka T, Hirayama M, Bay BH, Koeberl DD, Yen PM, 2016, Induction of autophagy improves hepatic lipid metabolism in glucose-6-phosphatase deficiency. J Hepatol 64:370–379 Jeon JY, Lee H, Park J, Lee M, Park SW, Kim JS, Lee M, Cho B, Kim K, Choi AM, Kim CK, Yun M, 2015, The regulation of glucose-6-phosphatase and phosphoenolpyruvate carboxykinase by autophagy in low-glycolytic hepatocellular carcinoma cells. Biochem Biophys Res Commun 463:440–446 Kaur J, Debnath J, 2015, Autophagy at the crossroads of catabolism and anabolism. Nat Rev Mol Cell Biol 16:461–472 Rousset M, Zweibaum A, Fogh J, 1981, Presence of glycogen and growth-related variations in 58 cultured human tumor cell lines of various tissue origins. Cancer Res 41:1165–1170 Guo JY, Chen HY, Mathew R, Fan J, Strohecker AM, Karsli- Uzunbas G, Kamphorst JJ, Chen G, Lemons JM, Karantza Vet al, 2011, Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev 25:460–470 Yang S, Wang X, Contino G, Liesa M, Sahin E, Ying H, Bause A, Li Y, Stommel JM, Dell'antonio G, Mautner J, Tonon G, Haigis M, Shirihai OS, Doglioni C, Bardeesy N, Kimmelman AC, 2011, Pancreatic cancers require autophagy for tumor growth. Genes Dev 25:717–729 Kapuy O, Vinod PK, Mandl J, Banhegyi G, 2013, A cellular stressdirected bistable switch controls the crosstalk between autophagy and apoptosis. Mol BioSyst 9:296–306 Das CK, Mandal M, Kogel D, 2018, Pro-survival autophagy and cancer cell resistance to therapy. CancerMetastasis Rev. https://doi. org/10.1007/s10555-018-9727-z Amaravadi R, Kimmelman AC, White E, 2016, Recent insights into the function of autophagy in cancer. Genes Dev 30:1913–1930 Lashinger LM, O’Flanagan CH, Dunlap SM, Rasmussen AJ, Sweeney S, Guo JY, Lodi A, Tiziani S, White E, Hursting SD, 2016, Starving cancer from the outside and inside: Separate and combined effects of calorie restriction and autophagy inhibition on Ras-driven tumors. Cancer Metab 4:18 Dikic I, 2017, Proteasomal and Autophagic Degradation Systems. Annu Rev Biochem 86:193–224 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 821 EP 826 DI 10.1007/s12253-018-0446-0 PG 6 ER PT J AU Choi, JE Lee, HJ Kim, SM Song, YS Yoo, JN Lee, HS AF Choi, Ji Eun Lee, Hwa Ju Kim, Sung Min Song, Yong Sang Yoo, Jin Nam Lee, Hyung Sug TI Intratumoral Heterogeneity of Somatic Mutations for NRIP1, DOK1, ULK1, ULK2, DLGAP3, PARD3 and PRKCI in Colon Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tumor suppressor gene; Frameshift mutation; Colon cancer; Microsatellite instability ID Tumor suppressor gene; Frameshift mutation; Colon cancer; Microsatellite instability AB Both NRIP1 and DOK1 genes are considered candidate tumor suppressor genes (TSGs). Also, cell polarity-related genes PARD3, PRKCI and DLGAP3, and autophagy-related genes ULK1 and ULK2 genes are considered to play crucial roles in tumorigenesis. The aim of our study was to find whether these genes were mutated in colorectal cancer (CRC). In a genome database, we observed that each of these genes harbored mononucleotide repeats in the coding sequences, which could be mutated in cancers with high microsatellite instability (MSIH). For this, we studied 124 CRCs for the frameshift mutations of these genes and their intratumoral heterogeneity (ITH). NRIP1, DOK1, PARD3, PRKCI, DLGAP3, ULK1 and ULK2 harbored 18 (22.8%), 2 (2.5%), 2 (2.5%), 2 (2.5%), 5 (6.3%), 2 (2.5%) and 2 (2.5%) of 79 CRCs with MSI-H, respectively. However, we found no such mutations in microsatellite stable (MSS) cancers in the nucleotide repeats. We also studied ITH for the frameshift mutations in 16 cases of CRCs and detected that the frameshift mutations of NRIP1, DOK1, PARD3, PRKCI, DLGAP3, ULK1 and ULK2 showed regional ITH in 5 (31.3%), 2 (12.5%), 0 (0%), 0 (0%), 1 (6.3%), 1 (6.3%) and 3 (18.8%) cases, respectively. Our data exhibit that candidate cancer-related genes NRIP1, DOK1, PARD3, PRKCI, DLGAP3, ULK1 and ULK2 harbor mutational ITH as well as the frameshift mutations in CRC with MSI-H. Also, the results suggest that frameshift mutations of these genes might play a role in tumorigenesis through their inactivation in CRC. C1 [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hwa Ju] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Song, Yong Sang] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of PathologySeoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Docquier A, Harmand PO, Fritsch S, Chanrion M, Darbon JM, Cavailles V, 2010, The transcriptional coregulator RIP140 represses E2F1 activity and discriminates breast cancer subtypes. Clin Cancer Res 16:2959–2970 Lapierre M, Bonnet S, Bascoul-Mollevi C, Ait-Arsa I, Jalaguier S, Del Rio M, Plateroti M, Roepman P, Ychou M, Pannequin J, Hollande F, Parker M, Cavailles V, 2014, RIP140 increases APC expression and controls intestinal homeostasis and tumorigenesis. J Clin Invest 124:1899–1913 Di Cristofano A, Carpino N, Dunant N, Friedland G, Kobayashi R, Strife A,Wisniewski D, Clarkson B, Pandolfi PP, Resh MD, 1998, Molecular cloning and characterization of p56dok-2 defines a new family of RasGAP-binding proteins. J Biol Chem 273:4827–4830 Berger AH, Niki M, Morotti A, Taylor BS, Socci ND, Viale A, Brennan C, Szoke J, Motoi N, Rothman PB, Teruya-Feldstein J, Gerald WL, Ladanyi M, Pandolfi PP, 2010, Identification of DOK genes as lung tumor suppressors. Nat Genet 42:216–223 Zen K, Yasui K, Gen Y, Dohi O,Wakabayashi N, Mitsufuji S, Itoh Y, Zen Y, Nakanuma Y, Taniwaki M, Okanoue T, Yoshikawa T, 2009, Defective expression of polarity protein PAR-3 gene, PARD3, in esophageal squamous cell carcinoma. Oncogene 28: 2910–2918 Suzuki A, Yamanaka T, Hirose T, Manabe N, Mizuno K, Shimizu M, Akimoto K, Izumi Y, Ohnishi T, Ohno S, 2001, Atypical protein kinase C is involved in the evolutionarily conserved par protein complex and plays a critical role in establishing epithelia-specific junctional structures. J Cell Biol 152:1183–1196 Mazzarella R, Ciccodicola A, Esposito T, Arcucci A, Migliaccio C, Jones C, Schlessinger D, D'Urso M, D'Esposito M, 1995, Human protein kinase C iota gene, PRKCI, is closely linked to the BTK gene in Xq21.3. Genomics 26:629–631 Ebnet K, Suzuki A, Horikoshi Y, Hirose T, Meyer Zu Brickwedde MK, Ohno S, Vestweber D, 2001, The cell polarity protein ASIP/ PAR-3 directly associates with junctional adhesion molecule, JAM). EMBO J 20:3738–3748 Welch JM,Wang D, Feng G, 2004, Differential mRNA expression and protein localization of the SAP90/PSD-95-associated proteins, SAPAPs, in the nervous system of the mouse. J Comp Neurol 472: 24–39 Jiang X, Overholtzer M, Thompson CB, 2015, Autophagy in cellular metabolism and cancer. J Clin Invest 125:47–54 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 McGranahan N, Swanton C, 2015, Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 27:15–26 Choi YJ, Rhee JK, Hur SY, KimMS, Lee SH, Chung YJ, Kim TM, Lee SH, 2017, Intraindividual genomic heterogeneity of high-grade serous carcinoma of the ovary and clinical utility of ascitic cancer cells for mutation profiling. J Pathol 241:57–66 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8: 305–311 Jung SH, Kim MS, Jung CK, Park HC, Choi HJ, Maeng L, Min KO, Kim J, Park TI, Shin OR, Kim TJ, Xu H, Lee KY, Kim TM, Song SY, Lee C, Chung YJ, Lee SH, 2016, Whole-exome sequencing identifies recurrent AKT1 mutations in sclerosing hemangioma of lung. Proc Natl Acad Sci U S A 113:10672–10677 Jo YS, Choi MR, Song SY, Kim MS, Yoo NJ, Lee SH, 2016, Frameshift mutations of HSPA4 and MED13 in gastric and colorectal cancers. Pathol Oncol Res 22:769–772 Choi EJ, Kim MS, Song SY, Yoo NJ, Lee SH, 2017, Intratumoral heterogeneity of frameshift mutations in MECOM gene is frequent in colorectal cancers with high microsatellite instability. Pathol Oncol Res 23:145–149 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Nature Reviews Cell 144:646–674 Calin GA, Gafa R, Tibiletti MG, Herlea V, Becheanu G, Cavazzini L, Barbanti-Brodano G, Nenci I, Negrini M, Lanza G, 2000, Genetic progression inmicrosatellite instability high, MSI-H, colon cancers correlates with clinico-pathological parameters: a study of the TGRbetaRII, BAX, hMSH3, hMSH6, IGFIIR and BLM genes. Int J Cancer 89:230–235 Choi YJ, Kim MS, An CH, Yoo NJ, Lee SH, 2014, Regional bias of intratumoral genetic heterogeneity of nucleotide repeats in colon cancers with microsatellite instability. Pathol Oncol Res 20:965–971 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 827 EP 832 DI 10.1007/s12253-017-0297-0 PG 6 ER PT J AU Horvath, J Szabo, A Tar, I Dezso, B Kiss, Cs Marton, I Scholtz, B AF Horvath, Jozsef Szabo, Adrien Tar, Ildiko Dezso, Balazs Kiss, Csongor Marton, Ildiko Scholtz, Beata TI Oral Health May Affect the Performance of mRNA-Based Saliva Biomarkers for Oral Squamous Cell Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE OSCC; Saliva mRNA; Biomarker; Saliva based diagnostics; qPCR ID OSCC; Saliva mRNA; Biomarker; Saliva based diagnostics; qPCR AB Oral squamous cell carcinoma (OSCC) has a dismal 50% five-year survival rate, emphasizing the need to develop reliable and sensitive tools for early diagnosis. In this study we evaluated the performance of 7 previously identified, potential mRNA biomarkers of OSCC in saliva samples of Hungarian patients. Expression of the putative OSCC biomarkers (DUSP1, OAZ1, H3F3A, IL1B, IL8, SAT and S100P), 2 biomarkers of inflammation (IL6 and TNFα) and 8 putative normalizing genes was quantified from each sample using real-time quantitative PCR. In contrast with previous studies, the expression pattern of the 7 mRNA biomarkers was similar between OSCC patients and age-matched control patients in the Hungarian patient population. On the other hand, 5 of the 7 mRNA biomarkers were present at significantly higher levels in saliva samples of OSCC patients when compared to young control patients. The best biomarker combination could distinguish only the OSCC vs. young control patients, but not the OSCC vs. age-matched control patients. In conclusion, the significant differences between our results and previous studies, and the clinical characteristics of the patients suggest that inflammatory processes in the oral cavity may affect the performance of the 7 putative salivary mRNA biomarkers. Lastly, since IL6 mRNA was quantifiable in the majority of OSCC cases, but only in a few control samples, salivary IL6 mRNA may be utilized as part of a biomarker combination to detect OSCC. C1 [Horvath, Jozsef] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. [Szabo, Adrien] University of Debrecen, Faculty of Dentistry, Department of Oral and Maxillofacial SurgeryDebrecen, Hungary. [Tar, Ildiko] University of Debrecen, Faculty of Dentistry, Department of PeriodontologyDebrecen, Hungary. [Dezso, Balazs] University of Debrecen, Faculty of Dentistry, Department of Oral PathologyDebrecen, Hungary. [Kiss, Csongor] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-OncologyDebrecen, Hungary. [Marton, Ildiko] University of Debrecen, Faculty of Dentistry, Department of Restorative DentistryDebrecen, Hungary. [Scholtz, Beata] University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular BiologyDebrecen, Hungary. RP Scholtz, B (reprint author), University of Debrecen, Faculty of Medicine, Department of Biochemistry and Molecular Biology, Debrecen, Hungary. EM scholtz@med.unideb.hu CR Guerra EN, Acevedo AC, Leite AF, Gozal D, Chardin H, De Luca CG, 2015, Diagnostic capability of salivary biomarkers in the assessment of head and neck cancer: a systematic review and metaanalysis. Oral Oncol 51(9):805–818. , DOI 10.1016/j. oraloncology.2015.06.010 Li Y, ZhouX, St JohnMA,Wong DT, 2004, RNA profiling of cellfree saliva using microarray technology. J Dent Res 83(3):199–203. , DOI 10.1177/154405910408300303 Li Y, St John MA, Zhou X, Kim Y, Sinha U, Jordan RC, Eisele D, Abemayor E, Elashoff D, Park NH, Wong DT, 2004, Salivary transcriptome diagnostics for oral cancer detection. Clin Cancer Res 10(24):8442–8450. , DOI 10.1158/1078-0432.CCR- -1167 4. Yakob M, Fuentes L, Wang MB, Abemayor E, Wong DT, 2014, Salivary biomarkers for detection of oral squamous cell carcinoma - current state and recent advances. Curr Oral Health Rep 1(2):133– 141. , DOI 10.1007/s40496-014-0014-y Brinkmann O, Kastratovic DA, Dimitrijevic MV, Konstantinovic VS, Jelovac DB, Antic J, Nesic VS, Markovic SZ, Martinovic ZR, Akin D, Spielmann N, Zhou H, Wong DT, 2011, Oral squamous cell carcinoma detection by salivary biomarkers in a Serbian population. Oral Oncol 47(1):51–55. , DOI 10.1016/j. oraloncology.2010.10.009 Elashoff D, Zhou H, Reiss J, Wang J, Xiao H, Henson B, Hu S, ArellanoM, Sinha U, Le A,Messadi D,WangM, Nabili V, Lingen M, Morris D, Randolph T, Feng Z, Akin D, Kastratovic DA, Chia D, Abemayor E, Wong DT, 2012, Prevalidation of salivary biomarkers for oral cancer detection. Cancer Epidemiol Biomark Prev 21(4):664–672. , DOI 10.1158/1055-9965.EPI-11-1093 Lallemant B, Evrard A, Combescure C, Chapuis H, Chambon G, Raynal C, Reynaud C, Sabra O, Joubert D, Hollande F, Lallemant JG, Lumbroso S, Brouillet J-P, 2009, Clinical relevance of nine transcriptional molecular markers for the diagnosis of head and neck squamous cell carcinoma in tissue and saliva rinse. BMC Cancer 9:370. , DOI 10.1186/1471-2407-9-370 ErvikM, Lam F, Ferlay J, Mery L, Soerjomataram I, Bray F, 2016, Cancer today. Lyon, France: International Agency for Research on Cancer. Cancer today. Available from: http://gco.iarc.fr/today, accessed [01/05/2017] Alevizos I, Mahadevappa M, Zhang X, Ohyama H, Kohno Y, Posner M, Gallagher GT, Varvares M, Cohen D, Kim D, Kent R, Donoff RB, Todd R, Yung CM, Warrington JA, Wong DT, 2001, Oral cancer in vivo gene expression profiling assisted by laser capture microdissection and microarray analysis. Oncogene 20(43): 6196–6204. , DOI 10.1038/sj.onc.1204685 Chen C, Mendez E, Houck J, Fan W, Lohavanichbutr P, Doody D, Yueh B, Futran ND, UptonM, Farwell DG, Schwartz SM, Zhao LP, 2008, Gene expression profiling identifies genes predictive of oral squamous cell carcinoma. Cancer Epidemiol Biomark Prev 17(8): 2152–2162. , DOI 10.1158/1055-9965.EPI-07-2893 Sajnani MR, Patel AK, Bhatt VD, Tripathi AK, Ahir VB, Shankar V, Shah S, Shah TM, Koringa PG, Jakhesara SJ, Joshi CG, 2012, Identification of novel transcripts deregulated in buccal cancer by RNA-seq. Gene 507(2):152–158 Jiang Q, Yu YC, Ding XJ, Luo Y, Ruan H, 2014, Bioinformatics analysis reveals significant genes and pathways to target for oral squamous cell carcinoma. Asian Pac J Cancer Prev 15(5):2273– 2278. , DOI 10.1016/j.gene.2012.07.036 Liu J, Duan Y, 2012, Saliva: A potential media for disease diagnostics and monitoring. Oral Oncol 48(7):569–577. , DOI 10.1016/j.oraloncology.2012.01.021 Otto S, Kasler M, 2005, Trends in cancer mortality and morbidity in Hungarian and international statistics. Characteristics and potential outcome of public health screening programmes. Magy Onkol 49(2):99–101 103–7 Suba Z, Mihalyi S, Takacs D, Gyulai-Gaal S, 2009, Oral cancer: morbus Hungaricus in the 21st century. Fogorv Sz 102(2):63–68 PMID:19514245 Hermann P, Gera I, Borbely J, Fejerdy P, Madlena M, 2009, Periodontal health of an adult population in Hungary: findings of a national survey. J Clin Periodontol 36(6):449–457. https://doi. org/10.1111/j.1600-051X.2009.01395.x Nemes JA, Nemes Z, Marton IJ, 2005, p21WAF1/CIP1 expression is a marker of poor prognosis in oral squamous cell carcinoma. J Oral Pathol Med 34(5):274–279. , DOI 10.1111/j.1600- 0714.2005.00310.x Nemes JA, Deli L, Nemes Z, Marton IJ, 2006, Expression of p16(INK4A), p53, and Rb proteins are independent from the presence of human papillomavirus genes in oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 102(3): 344–352. , DOI 10.1016/j.tripleo.2005.10.069 Lang NP, Corbet EF, 1995, Periodontal diagnosis in daily practice. Int Dent J 45(1):3–15 PMID:7607742 Beikler T, Flemmig TF, 2011, Oral biofilm-associated diseases: trends and implications for quality of life, systemic health and expenditures. Periodontol 2000 55(1):87–103. , DOI 10. 1111/j.1600-0757.2010.00360.x Pindborg JJ, Reichart PA, Smith CJ, Van der Waal I, 1997, Histological typing of cancer and precancer of the oral mucosa. WHO international histological classification of tumours, 2nd edn. Springer, Berlin Head and neck tumors: Lip and oral cavity, 2009, In: Sobin LH, Gospodarowitz MK, Wittekind CH, editors. TNM classification of malignant tumors. 7th ed. Wiley-Blackwell Mehdi Khanlou K, Van Bockstaele E, 2012, A critique of widely used normalization software tools and an alternative method to identify reliable reference genes in red clover, Trifolium Pratense L., Planta 236(5):1381–1393. , DOI 10.1007/s00425-012- 1682-2 Hu Z, Zimmermann BG, Zhou H, Wang J, Henson BS, Yu W, Elashoff D, Krupp G, Wong DT, 2008, Exon-level expression profiling: a comprehensive transcriptome analysis of oral fluids. Clin Chem 54(5):824–832. , DOI 10.1373/clinchem. 2007.096164 Cheng YS, Jordan L, Rees T, Chen HS, Oxford L, Brinkmann O, Wong D, 2014, Levels of potential oral cancer salivary mRNA biomarkers in oral cancer patients in remission and oral lichen planus patients. Clin Oral Investig 18(3):985–993. , DOI 10.1007/s00784-013-1041-0 Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, Speleman F, 2002, Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol 3(7):RESEARCH0034 PMCID: PMC126239 Andersen CL, Jensen JL, Orntoft TF, 2004, Normalization of realtime quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res 64(15):5245–5250. , DOI 10.1158/0008-5472.CAN-04- 0496 Cheng YL, Jordan L, Chen HS, Kang D, Oxford L, Plemons J, Parks H, Rees T, 2017, Chronic periodontitis can affect the levels of potential oral cancer salivary mRNA biomarkers. J Periodontal Res 52(3):428–437. , DOI 10.1111/jre.12407 Ganzetti G, Campanati A, Santarelli A, Sartini D, Molinelli E, Brisigotti V, Di Ruscio G, Bobyr I, Emanuelli M, Offidani A, 2016, Salivary interleukin-1β: oral inflammatory biomarker in patients with psoriasis. J Int Med Res 44(1 suppl):10–14. https:// doi.org/10.1177/0300060515598902 Nie S, Zhang H, Mayer KM, Oppenheim FG, Little FF, Greenberg J, Uluer AZ, Walt DR, 2015, Correlations of salivary biomarkers with clinical assessments in patients with cystic fibrosis. PLoS One 10(8):e0135237. , DOI 10.1371/journal.pone.0135237 Rathnayake N, Akerman S, Klinge B, Lundegren N, Jansson H, Tryselius Y, Sorsa T, Gustafsson A, 2013, Salivary biomarkers for detection of systemic diseases. PLoS One 8(4):e61356. https:// doi.org/10.1371/journal.pone.0061356 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 833 EP 842 DI 10.1007/s12253-017-0296-1 PG 10 ER PT J AU Zhu, L Zhang, Sh Xun, Y Jiang, Y Xia, B Chen, X Wang, L Jiang, H Shenglin, M AF Zhu, Lucheng Zhang, Shirong Xun, Yanping Jiang, Yanping Xia, Bing Chen, Xueqin Wang, Limin Jiang, Hong Shenglin, Ma TI Comparison of the Amplification Refractory Mutation System, Super Amplification Refractory Mutation System, and Droplet Digital PCR for T790 M Mutation Detection in Non-small Cell Lung Cancer after Failure of Tyrosine Kinase Inhibitor Treatment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE T790 M; ctDNA; ARMS; ddPCR; SuperARMS; TKI resistance ID T790 M; ctDNA; ARMS; ddPCR; SuperARMS; TKI resistance AB Plasma mutation detection has the advantages of non-invasiveness and accessibility. Here, we evaluated three methods, the amplification refractory mutation system (ARMS), second-generation ARMS (SuperARMS), and droplet digital PCR (ddPCR), to assess their concordance and feasibility for the detection of mutations in plasma samples. Non-small lung cancer patients with stage IIIB/IV that were resistant to epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) treatment were enrolled. Blood samples were collected within 14 days after TKI resistance. Each sample was simultaneously assessed by the three methods. In total, 169 patients were enrolled; 54.4% were female, 72.2% were diagnosed with stage IV disease; and 97.6%had adenocarcinoma. T790Mmutations were detected in 42 (24.8%) of the 169 samples using ARMS, one of which carried the T790 M alone, 22 that also encoded exon 19 deletions, and 19 with L858R mutations. For the SuperARMS assay, 59 (34.9%) samples exhibited the T790 M mutation, and 110 (65.1%) showed no detectable T790 M mutation. ddPCR showed that 61 (36.1%) samples contained the T790 M mutation, whereas 108 (63.9%) were not positive. T790Mabundance ranged from 0.04%to 38.2%. The median T790 M abundance was 0.15% for total samples and 2.98% for T790 M mutation samples. The overall concordance was 78.7% (133/169) among ARMS, SuperARMS, and ddPCR. Compared with patients with stage III disease, patients with stage IV disease exhibited a higher T790 M mutation detection rate (28.7% vs. 14.9% by ARMS; 37.7% vs. 27.7% by SuperARMS; and 41.8% vs. 21.3% by ddPCR). Liquid biopsy showed promise and has the advantages of noninvasiveness and accessibility. T790 M detection based on circulating tumor DNA showed high concordance. Compared with non-digital platforms, ddPCR showed higher sensitivity and provided both frequency and abundance information, which might be important for treatment decisions. C1 [Zhu, Lucheng] Hangzhou Cancer Hospital, Department of OncologyHangzhou, China. [Zhang, Shirong] Nanjing Medical University, Hangzhou First People’s Hospital, Hangzhou Translational Medicine Research CenterNanjing, China. [Xun, Yanping] Nanjing Medical University, Hangzhou First People’s Hospital, Hangzhou Translational Medicine Research CenterNanjing, China. [Jiang, Yanping] Nanjing Medical University, Hangzhou First People’s Hospital, Hangzhou Translational Medicine Research CenterNanjing, China. [Xia, Bing] Hangzhou Cancer Hospital, Department of OncologyHangzhou, China. [Chen, Xueqin] Hangzhou Cancer Hospital, Department of OncologyHangzhou, China. [Wang, Limin] Nanjing Medical University, Hangzhou First People’s Hospital, Department of RespiratoryNanjing, China. [Jiang, Hong] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Thoracic SurgeryNanjing, China. [Shenglin, Ma] Nanjing Medical University, Hangzhou First People’s Hospital, Department of OncologyNanjing, China. RP Shenglin, M (reprint author), Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, Nanjing, China. EM mashenglin@medmail.com.cn CR Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, Sunpaweravong P, Han B, Margono B, Ichinose Y, Nishiwaki Y, Ohe Y, Yang JJ, Chewaskulyong B, Jiang H, Duffield EL,Watkins CL, Armour AA, Fukuoka M, 2009, Gefitinib or carboplatinpaclitaxel in pulmonary adenocarcinoma. N Engl J Med 361(10): 947–957., DOI 10.1056/NEJMoa0810699 Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, Gemma A, Harada M, Yoshizawa H, Kinoshita I, Fujita Y, Okinaga S, Hirano H,Yoshimori K, Harada T, Ogura T, Ando M, Miyazawa H, Tanaka T, Saijo Y, Hagiwara K, Morita S, Nukiwa T, 2010, Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med 362(25):2380–2388., DOI 10. 1056/NEJMoa0909530 Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, Seto T, Satouchi M, Tada H, Hirashima T, Asami K, Katakami N, Takada M, Yoshioka H, Shibata K, Kudoh S, Shimizu E, Saito H, Toyooka S, Nakagawa K, Fukuoka M, 2010, Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor, WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 11(2):121–128., DOI 10. 1016/s1470-2045(09)70364-x Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, Palmero R, Garcia-Gomez R, Pallares C, Sanchez JM, Porta R, Cobo M, Garrido P, Longo F, Moran T, Insa A, DeMarinis F, Corre R, Bover I, Illiano A, Dansin E, de Castro J, Milella M, Reguart N, Altavilla G, Jimenez U, Provencio M, Moreno MA, Terrasa J, Munoz-Langa J, Valdivia J, Isla D, Domine M, Molinier O, Mazieres J, Baize N, Garcia-Campelo R, Robinet G, Rodriguez- Abreu D, Lopez-Vivanco G, Gebbia V, Ferrera-Delgado L, Bombaron P, Bernabe R, Bearz A, Artal A, Cortesi E, Rolfo C, Sanchez-Ronco M, Drozdowskyj A, Queralt C, de Aguirre I, Ramirez JL, Sanchez JJ, Molina MA, Taron M, Paz-Ares L, 2012, Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutationpositive non-small-cell lung cancer, EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 13(3):239– 246., DOI 10.1016/s1470-2045(11)70393-x Zhou C,Wu YL, ChenG, Feng J, LiuXQ,Wang C, Zhang S,Wang J, Zhou S, Ren S, Lu S, Zhang L, Hu C, Hu C, Luo Y, Chen L, Ye M, Huang J, Zhi X, Zhang Y, Xiu Q, Ma J, Zhang L, You C, 2011, Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer, OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 12(8):735–742., DOI 10. 1016/s1470-2045(11)70184-x Kobayashi S, Boggon TJ, Dayaram T, Janne PA, Kocher O, Meyerson M, Johnson BE, Eck MJ, Tenen DG, Halmos B, 2005, EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med 352(8):786–792., DOI 10.1056/ NEJMoa044238 Cheng X, Chen H, 2014, Tumor heterogeneity and resistance to EGFR-targeted therapy in advanced nonsmall cell lung cancer: challenges and perspectives. Onco Targets Ther 7:1689–1704., DOI 10.2147/ott.s66502 Tan CS, Gilligan D, Pacey S, 2015, Treatment approaches for EGFR-inhibitor-resistant patients with non-small-cell lung cancer. Lancet Oncol 16(9):e447–e459., DOI 10.1016/s1470-2045(15, 00246-6 Janne PA, Yang JC, Kim DW, Planchard D, Ohe Y, Ramalingam SS, AhnMJ, KimSW, SuWC, Horn L,HaggstromD, Felip E,Kim JH, Frewer P, CantariniM, Brown KH, Dickinson PA, Ghiorghiu S, Ranson M, 2015, AZD9291 in EGFR inhibitor-resistant nonsmall- cell lung cancer. N Engl J Med 372(18):1689–1699., DOI 10. 1056/NEJMoa1411817 ArcilaME, Oxnard GR, Nafa K, Riely GJ, Solomon SB, Zakowski MF, Kris MG, Pao W, Miller VA, Ladanyi M, 2011, Rebiopsy of lung cancer patients with acquired resistance to EGFR inhibitors and enhanced detection of the T790M mutation using a locked nucleic acid-based assay. Clin Cancer Res 17(5):1169–1180., DOI 10.1158/1078-0432.ccr-10-2277 Goto K, Satouchi M, Ishii G, Nishio K, Hagiwara K, Mitsudomi T, Whiteley J, Donald E, McCormack R, Todo T, 2012, An evaluation study of EGFR mutation tests utilized for non-small-cell lung cancer in the diagnostic setting. Ann Oncol 23(11):2914–2919., DOI 10. 1093/annonc/mds121 Angulo B, Conde E, Suarez-Gauthier A, Plaza C, Martinez R, Redondo P, Izquierdo E, Rubio-Viqueira B, Paz-Ares L, Hidalgo M, Lopez-Rios F, 2012, A comparison of EGFR mutation testing methods in lung carcinoma: direct sequencing, real-time PCR and immunohistochemistry. PLoS One 7(8):e43842., DOI 10.1371/ journal.pone.0043842 Douillard JY, Ostoros G, Cobo M, Ciuleanu T, Cole R, McWalter G, Walker J, Dearden S, Webster A, Milenkova T, McCormack R, 2014, Gefitinib treatment in EGFR mutated caucasian NSCLC: circulating-free tumor DNA as a surrogate for determination of EGFR status. J Thorac Oncol 9(9):1345–1353., DOI 10.1097/jto. 0000000000000263 Taniguchi K, Uchida J, Nishino K, Kumagai T, Okuyama T, Okami J, Higashiyama M, Kodama K, Imamura F, Kato K, 2011, Quantitative detection of EGFR mutations in circulating tumor DNA derived from lung adenocarcinomas. Clin Cancer Res 17(24):7808–7815., DOI 10.1158/1078-0432.ccr-11-1712 KimHR, Lee SY, HyunDS, LeeMK, LeeHK, ChoiCM, Yang SH, Kim YC, Lee YC, Kim SY, Jang SH, Lee JC, Lee KY, 2013, Detection of EGFR mutations in circulating free DNA by PNAmediated PCR clamping. J Exp Clin Cancer Res 32(1):50., DOI 10. 1186/1756-9966-32-50 Zhang S, Xia B, Jiang H, Wang L, Xu R, Shi Y, Zhang J, Xu M, Cram DS, Ma S, 2016, Comprehensive profiling and quantitation of oncogenic mutations in non small-cell lung carcinoma using single molecule amplification and re-sequencing technology. Oncotarget., DOI 10.18632/oncotarget.10464 Forshew T, Murtaza M, Parkinson C, Gale D, Tsui DW, Kaper F, Dawson SJ, Piskorz AM, Jimenez-LinanM, Bentley D, Hadfield J, May AP, Caldas C, Brenton JD, Rosenfeld N, 2012, Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 4(136):136ra168., DOI 10.1126/scitranslmed.3003726 Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T, Piskorz AM, Parkinson C, Chin SF, Kingsbury Z, Wong AS, Marass F, Humphray S, Hadfield J, Bentley D, Chin TM, Brenton JD, Caldas C, Rosenfeld N, 2013, Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497(7447):108–112., DOI 10.1038/nature12065 Oxnard GR, Paweletz CP, Kuang Y, Mach SL, O'Connell A, Messineo MM, Luke JJ, Butaney M, Kirschmeier P, Jackman DM, Janne PA, 2014, Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative nextgeneration genotyping of cell-free plasma DNA. Clin Cancer Res 20(6):1698–1705., DOI 10.1158/1078-0432.ccr-13-2482 Janku F, Angenendt P, Tsimberidou AM, Fu S, Naing A, Falchook GS, Hong DS, Holley VR, Cabrilo G, Wheler JJ, Piha-Paul SA, Zinner RG, Bedikian AY, Overman MJ, Kee BK, Kim KB, Kopetz ES, Luthra R, Diehl F, Meric-Bernstam F, Kurzrock R, 2015, Actionable mutations in plasma cell-free DNA in patients with advanced cancers referred for experimental targeted therapies. Oncotarget 6(14):12809–12821., DOI 10.18632/oncotarget.3373 Bria E, Pilotto S, Amato E, Fassan M, Novello S, Peretti U, Vavala T, Kinspergher S, Righi L, Santo A, Brunelli M, Corbo V, Giglioli E, Sperduti I, Milella M, Chilosi M, Scarpa A, Tortora G, 2015, Molecular heterogeneity assessment by next-generation sequencing and response to gefitinib of EGFR mutant advanced lung adenocarcinoma. Oncotarget 6(14): 12783–12795., DOI 10.18632/oncotarget.3727 Jackman D, Pao W, Riely GJ, Engelman JA, Kris MG, Janne PA, Lynch T, Johnson BE, Miller VA, 2010, Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol 28(2): 357–360., DOI 10.1200/jco.2009.24.7049 Liu X, Lu Y, Zhu G, Lei Y, Zheng L, Qin H, Tang C, Ellison G, McCormack R, Ji Q, 2013, The diagnostic accuracy of pleural effusion and plasma samples versus tumour tissue for detection of EGFR mutation in patients with advanced non-small cell lung cancer: comparison of methodologies. J Clin Pathol 66(12):1065– 1069., DOI 10.1136/jclinpath-2013-201728 Diaz LA Jr, Bardelli A, 2014, Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol 32(6):579–586., DOI 10.1200/jco. 2012.45.2011 Nakamura T, Sueoka-Aragane N, Iwanaga K, Sato A, Komiya K, Abe T, Ureshino N, Hayashi S, Hosomi T, Hirai M, Sueoka E, Kimura S, 2011, A noninvasive system for monitoring resistance to epidermal growth factor receptor tyrosine kinase inhibitors with plasma DNA. J Thorac Oncol 6(10):1639–1648., DOI 10.1097/JTO. 0b013e31822956e8 Sakai K, Horiike A, Irwin DL, Kudo K, Fujita Y, Tanimoto A, Sakatani T, Saito R, Kaburaki K, Yanagitani N, Ohyanagi F, Nishio M, Nishio K, 2013, Detection of epidermal growth factor receptor T790M mutation in plasma DNA from patients refractory to epidermal growth factor receptor tyrosine kinase inhibitor. Cancer Sci 104(9):1198–1204., DOI 10.1111/cas.12211 Zheng D, Ye X, Zhang MZ, Sun Y, Wang JY, Ni J, Zhang HP, Zhang L, Luo J, Zhang J, Tang L, Su B, Chen G, Zhu G, Gu Y, Xu JF, 2016, Plasma EGFR T790M ctDNA status is associated with clinical outcome in advanced NSCLC patients with acquired EGFR-TKI resistance. Sci Rep 6:20913., DOI 10.1038/srep20913 Wang W, Song Z, Zhang Y, 2017, A comparison of ddPCR and ARMS for detecting EGFRT790M status in ctDNA fromadvanced NSCLC patients with acquired EGFR-TKI resistance. Cancer Med 6(1):154–162., DOI 10.1002/cam4.978 Goto K, Ichinose Y, Ohe Y, Yamamoto N, Negoro S, Nishio K, Itoh Y, Jiang H, Duffield E, McCormack R, Saijo N, Mok T, FukuokaM, 2012, Epidermal growth factor receptor mutation status in circulating freeDNAin serum: fromIPASS, a phase III study of gefitinib or carboplatin/paclitaxel in non-small cell lung cancer. J Thorac Oncol 7(1):115–121., DOI 10.1097/JTO.0b013e3182307f98 Yatabe Y, Kerr KM, Utomo A, Rajadurai P, Tran VK, Du X, Chou TY, Enriquez ML, Lee GK, Iqbal J, Shuangshoti S, Chung JH, Hagiwara K, Liang Z, Normanno N, Park K, Toyooka S, Tsai CM, Waring P, Zhang L, McCormack R, Ratcliffe M, Itoh Y, Sugeno M, Mok T, 2015, EGFR mutation testing practices within the Asia Pacific region: results of a multicenter diagnostic survey. J Thorac Oncol 10(3):438–445., DOI 10.1097/jto.0000000000000422 Thatcher N, Chang A, Parikh P, Rodrigues Pereira J, Ciuleanu T, von Pawel J, Thongprasert S, Tan EH, Pemberton K, Archer V, Carroll K, 2005, Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study, Iressa survival evaluation in lung cancer). Lancet 366(9496):1527–1537., DOI 10.1016/s0140-6736(05)67625-8 Pao W, Ladanyi M, 2007, Epidermal growth factor receptor mutation testing in lung cancer: searching for the ideal method. Clin Cancer Res 13(17):4954–4955., DOI 10.1158/1078-0432. ccr-07-1387 Zhu CQ, da Cunha SG, Ding K, Sakurada A, Cutz JC, Liu N, Zhang T, Marrano P, Whitehead M, Squire JA, Kamel-Reid S, Seymour L, Shepherd FA, Tsao MS, 2008, Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada clinical trials group study BR.21. J Clin Oncol 26(26):4268–4275., DOI 10.1200/jco.2007.14.8924 Fukuoka M,Wu YL, Thongprasert S, Sunpaweravong P, Leong SS, Sriuranpong V, Chao TY, Nakagawa K, Chu DT, Saijo N, Duffield EL, Rukazenkov Y, Speake G, Jiang H, Armour AA, To KF, Yang JC, Mok TS, 2011, Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia, IPASS). J Clin Oncol 29(21):2866–2874., DOI 10.1200/jco.2010.33.4235 Luo J, Shen L, Zheng D, 2014, Diagnostic value of circulating free DNA for the detection of EGFR mutation status in NSCLC: a systematic review and meta-analysis. Sci Rep 4:6269., DOI 10. 1038/srep06269 Nie K, Jia Y, Zhang X, 2015, Cell-free circulating tumor DNA in plasma/serum of non-small cell lung cancer. Tumour Biol 36(1):7– 19., DOI 10.1007/s13277-014-2758-3 Mok T, Wu YL, Lee JS, Yu CJ, Sriuranpong V, Sandoval-Tan J, Ladrera G, Thongprasert S, Srimuninnimit V, Liao M, Zhu Y, Zhou C, Fuerte F, Margono B, Wen W, Tsai J, Truman M, Klughammer B, Shames DS, Wu L, 2015, Detection and dynamic changes of EGFR mutations from circulating tumor DNA as a predictor of survival outcomes in NSCLC patients treated with first-line intercalated Erlotinib and chemotherapy. Clin Cancer Res 21(14):3196– 3203., DOI 10.1158/1078-0432.ccr-14-2594 Douillard JY, Ostoros G, Cobo M, Ciuleanu T, McCormack R, Webster A, Milenkova T, 2014, First-line gefitinib in Caucasian EGFR mutation-positive NSCLC patients: a phase- IV, open-label, single-arm study. Br J Cancer 110(1):55–62., DOI 10.1038/bjc.2013.721 Ma M, Shi C, Qian J, Teng J, Zhong H, Han B, 2016, Comparison of plasma and tissue samples in epidermal growth factor receptor mutation by ARMS in advanced non-small cell lung cancer. Gene., DOI 10.1016/j.gene.2016.06.053 Wang Z, Chen R, Wang S, Zhong J, Wu M, Zhao J, Duan J, Zhuo M, An T, Wang Y, Bai H, Wang J, 2014, Quantification and dynamic monitoring of EGFR T790M in plasma cell-free DNA by digital PCR for prognosis of EGFR-TKI treatment in advanced NSCLC. PLoS One 9(11):e110780., DOI 10.1371/journal.pone. 0110780 Pinheiro LB, Coleman VA, Hindson CM, Herrmann J, Hindson BJ, Bhat S, Emslie KR, 2012, Evaluation of a droplet digital polymerase chain reaction format for DNA copy number quantification. Anal Chem 84(2):1003–1011., DOI 10.1021/ac202578x Huggett JF, Foy CA, Benes V, Emslie K, Garson JA, Haynes R, Hellemans J, KubistaM, Mueller RD, Nolan T, PfafflMW, Shipley GL, Vandesompele J, Wittwer CT, Bustin SA, 2013, The digital MIQE guidelines: minimum information for publication of quantitative digital PCR experiments. Clin Chem 59(6):892–902., DOI 10. 1373/clinchem.2013.206375 Huggett JF, Novak T, Garson JA, Green C,Morris-Jones SD,Miller RF, Zumla A, 2008, Differential susceptibility of PCR reactions to inhibitors: an important and unrecognised phenomenon. BMC Res Notes 1:70., DOI 10.1186/1756-0500-1-70 Hindson BJ, Ness KD, Masquelier DA, Belgrader P, Heredia NJ, Makarewicz AJ, Bright IJ, Lucero MY, Hiddessen AL, Legler TC, Kitano TK, Hodel MR, Petersen JF, Wyatt PW, Steenblock ER, Shah PH, Bousse LJ, Troup CB, Mellen JC, Wittmann DK, Erndt NG, Cauley TH, Koehler RT, So AP, Dube S, Rose KA, Montesclaros L, Wang S, Stumbo DP, Hodges SP, Romine S, Milanovich FP, White HE, Regan JF, Karlin-Neumann GA, Hindson CM, Saxonov S, Colston BW, 2011, High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal Chem 83(22):8604–8610., DOI 10.1021/ac202028g Zhao H,Wilkins K, Damon IK, Li Y, 2013, Specific qPCR assays for the detection of orf virus, pseudocowpox virus and bovine papular stomatitis virus. J Virol Methods 194(1–2):229–234., DOI 10. 1016/j.jviromet.2013.08.027 Brunetto GS, Massoud R, Leibovitch EC, Caruso B, Johnson K, Ohayon J, Fenton K, Cortese I, Jacobson S, 2014, Digital droplet PCR, ddPCR, for the precise quantification of human Tlymphotropic virus 1 proviral loads in peripheral blood and cerebrospinal fluid of HAM/TSP patients and identification of viral mutations. J Neuro-Oncol 20(4):341–351., DOI 10.1007/s13365- 014-0249-3 Racki N, Dreo T, Gutierrez-Aguirre I, Blejec A, RavnikarM(2014, Reverse transcriptase droplet digital PCR shows high resilience to PCR inhibitors from plant, soil and water samples. Plant Methods 10(1):42., DOI 10.1186/s13007-014-0042-6 Doi H, Takahara T,Minamoto T, Matsuhashi S, Uchii K, Yamanaka H, 2015, Droplet digital polymerase chain reaction, PCR, outperforms real-time PCR in the detection of environmental DNA from an invasive fish species. Environ Sci Technol 49(9):5601–5608., DOI 10.1021/acs.est.5b00253 Yang X, ZhuoM, Ye X, Bai H,Wang Z, Sun Y, Zhao J, An T, Duan J, Wu M, Wang J, 2016, Quantification of mutant alleles in circulating tumor DNA can predict survival in lung cancer. Oncotarget., DOI 10.18632/oncotarget.8021 Balak MN, Gong Y, Riely GJ, Somwar R, Li AR, Zakowski MF, Chiang A, Yang G, Ouerfelli O, Kris MG, Ladanyi M, Miller VA, Pao W, 2006, Novel D761Yand common secondary T790M mutations in epidermal growth factor receptor-mutant lung adenocarcinomas with acquired resistance to kinase inhibitors. Clin Cancer Res 12(21):6494–6501., DOI 10.1158/1078-0432.ccr-06-1570 Zhou Q, Zhang XC, Chen ZH, Yin XL, Yang JJ, Xu CR, Yan HH, Chen HJ, Su J, ZhongWZ, Yang XN, An SJ,Wang BC, Huang YS, Wang Z, Wu YL, 2011, Relative abundance of EGFR mutations predicts benefit from gefitinib treatment for advanced non-smallcell lung cancer. J Clin Oncol 29(24):3316–3321., DOI 10.1200/jco. 2010.33.3757 Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, Shepherd FA, He Y, Akamatsu H, Theelen WS, Lee CK, Sebastian M, Templeton A, Mann H, Marotti M, Ghiorghiu S, Papadimitrakopoulou VA, 2017, Osimertinib or platinum- Pemetrexed in EGFR T790M-positive lung cancer. N Engl J Med 376(7):629–640., DOI 10.1056/NEJMoa1612674 Oxnard GR, Thress KS, Alden RS, Lawrance R, Paweletz CP, Cantarini M, Yang JC, Barrett JC, Janne PA, 2016, Association between plasma genotyping and outcomes of treatment with Osimertinib, AZD9291, in advanced non-small-cell lung cancer. J Clin Oncol 34(28):3375–3382., DOI 10.1200/jco.2016.66.7162 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 843 EP 851 DI 10.1007/s12253-017-0286-3 PG 9 ER PT J AU Feng, L Ma, J Ji, H Liu, Y Hu, W AF Feng, Linsen Ma, Jianhua Ji, Haiming Liu, Yichun Hu, Weixing TI MiR-184 Retarded the Proliferation, Invasiveness and Migration of Glioblastoma Cells by Repressing Stanniocalcin-2 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma; miR-184; STC2; Proliferation; Migration; Invasion ID Glioblastoma; miR-184; STC2; Proliferation; Migration; Invasion AB To investigate the repression of miR-184 on Stanniocalcin-2 (STC2) and how this axis affects the propagation, invasiveness and migration ability of glioblastoma cells. RT-PCR was employed to determine the miR-184 and STC2 mRNA expression both in tissues and cells. Western blot was employed to determine the protein expression levels. The cells were transfected via lipofection. MTT, colony formation, invasion and scratch healing assays were conducted to study the propagation, invasiveness and migratory ability of glioblastoma cells, respectively. The dual luciferase reporter gene assay was conducted to determine whether miR-184 could directly bind to STC2 mRNA 3’UTR. MiR-184 was under-expressed whereas STC2 was over-expressed in glioblastoma tissues and cell line. The up-regulation of miR-184 significantly suppressed the propagation, migratory ability and invasion of glioblastoma cells, whereas the overexpression of STC2 restored this effect. MiR-184 was confirmed to directly target STC2. MiR-184 could retard the propagation, invasiveness and migratory ability of glioblastoma cells by suppressing STC2. C1 [Feng, Linsen] Taixing People’s Hospital, Department of Neurosurgery, 225400 Taizhou, Jiangsu, China. [Ma, Jianhua] Taixing People’s Hospital, Department of Neurosurgery, 225400 Taizhou, Jiangsu, China. [Ji, Haiming] Taixing People’s Hospital, Department of Neurosurgery, 225400 Taizhou, Jiangsu, China. [Liu, Yichun] Taixing People’s Hospital, Department of Neurosurgery, 225400 Taizhou, Jiangsu, China. [Hu, Weixing] The First Affiliated Hospital of Nanjing Medical University, Department of Neurosurgery, No. 300 Guangzhou Road, 210029 Nanjing, Jiangsu, China. RP Hu, W (reprint author), The First Affiliated Hospital of Nanjing Medical University, Department of Neurosurgery, 210029 Nanjing, China. EM xinglaijun@126.com CR Kouhkan F, Mobarra N, Soufi-Zomorrod M et al, 2016, MicroRNA-129-1 acts as tumour suppressor and induces cell cycle arrest of GBM cancer cells through targeting IGF2BP3 and MAPK1. J Med Genet 53:24–33 Zhen L, Li J, Zhang M, et al. MiR-10b decreases sensitivity of glioblastoma cells to radiation by targeting AKT. J Biol Res, Thessalon). 2016;23:14 ZhangW, Zhang J, Yan Wet al, 2013, Whole-genome microRNA expression profiling identifies a 5-microRNA signature as a prognostic biomarker in Chinese patients with primary glioblastoma multiforme. Cancer 119:814–824 Sizoo EM, Braam L, Postma TJ et al, 2010, Symptoms and problems in the end-of-life phase of high-grade glioma patients. Neuro- Oncology 12:1162–1166 Agnihotri S, Burrell KE, Wolf A et al, 2013, Glioblastoma, a brief review of history, molecular genetics, animal models and novel therapeutic strategies. Arch Immunol Ther Exp 61:25–41 Cheng Z, Wang HZ, Li X et al, 2015, MicroRNA-184 inhibits cell proliferation and invasion, and specifically targets TNFAIP2 in Glioma. J Exp Clin Cancer Res 34:27 Iorio MV, Croce CM, 2012, MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med 4:143–159 Sethi S, Ali S, Sethi S et al, 2014, MicroRNAs in personalized cancer therapy. Clin Genet 86:68–73 Yang BF, Lu YJ, Wang ZG, 2009, MicroRNAs and apoptosis: implications in the molecular therapy of human disease. Clin Exp Pharmacol Physiol 36:951–960 Zhou R, Zhou X, Yin Z et al, 2015, Tumor invasion and metastasis regulated by microRNA-184 and microRNA-574-5p in small-cell lung cancer. Oncotarget 6:44609–44622 Su Z, Chen D, Li Yet al, 2015, microRNA-184 functions as tumor suppressor in renal cell carcinoma. Exp Ther Med 9:961–966 Qin CZ, Lou XY, Lv QL et al, 2015, MicroRNA-184 acts as a potential diagnostic and prognostic marker in epithelial ovarian cancer and regulates cell proliferation, apoptosis and inflammation. Pharmazie 70:668–673 Feng R, Dong L, 2015, Inhibitory effect of miR-184 on the potential of proliferation and invasion in human glioma and breast cancer cells in vitro. Int J Clin Exp Pathol 8:9376–9382 Malzkorn B,Wolter M, Liesenberg F et al, 2010, Identification and functional characterization of microRNAs involved in the malignant progression of gliomas. Brain Pathol 20:539–550 Chang AC, Janosi J, Hulsbeek M et al, 1995, A novel human cDNA highly homologous to the fish hormone stanniocalcin. Mol Cell Endocrinol 112:241–247 Chang AC, Jeffrey KJ, Tokutake Yet al, 1998, Human stanniocalcin, STC): genomic structure, chromosomal localization, and the presence of CAG trinucleotide repeats. Genomics 47:393–398 Wagner GF, Dimattia GE, 2006, The stanniocalcin family of proteins. J Exp Zool A Comp Exp Biol 305:769–780 Ito D, Walker JR, Thompson CS et al, 2004, Characterization of stanniocalcin 2, a novel target of the mammalian unfolded protein response with cytoprotective properties. Mol Cell Biol 24:9456–9469 Gagliardi AD, Kuo EY, Raulic S et al, 2005, Human stanniocalcin- 2 exhibits potent growth-suppressive properties in transgenic mice independently of growth hormone and IGFs. Am J Physiol Endocrinol Metab 288:E92–105 Wang Y, Gao Y, Cheng H et al, 2015, Stanniocalcin 2 promotes cell proliferation and cisplatin resistance in cervical cancer. Biochem Biophys Res Commun 466:362–368 Na SS, Aldonza MB, Sung HJ et al, 1854, Stanniocalcin-2, STC2): a potential lung cancer biomarker promotes lung cancer metastasis and progression. Biochim Biophys Acta 2015:668–676 Hou J, Wang Z, Xu H et al, 2015, Stanniocalicin 2 suppresses breast cancer cell migration and invasion via the PKC/claudin-1- mediated signaling. PLoS One 10:e0122179 Guo GX, Li QY, Ma WL et al, 2015, MicroRNA-485-5p suppresses cell proliferation and invasion in hepatocellular carcinoma by targeting stanniocalcin 2. Int J Clin Exp Pathol 8:12292–12299 Lin S, Guo Q, Wen J et al, 2014, Survival analyses correlate stanniocalcin 2 overexpression to poor prognosis of nasopharyngeal carcinomas. J Exp Clin Cancer Res 33:26 Su J, Guo B, Zhang Tet al, 2015, Stanniocalcin-1, a new biomarker of glioma progression, is associated with prognosis of patients. Tumour Biol 36:6333–6339 Yamamoto K, Tajima Y, Hasegawa A et al, 2016, Contrasting effects of stanniocalcin-related polypeptides on macrophage foam cell formation and vascular smooth muscle cell migration. Peptides 82:120–127 Jepsen MR, Kloverpris S, Botkjaer JA et al, 2016, The proteolytic activity of pregnancy-associated plasma protein-a is potentially regulated by stanniocalcin-1 and -2 during human ovarian follicle development. Hum Reprod 31:866–874 Zhou H, Li YY, ZhangWQ et al, 2014, Expression of stanniocalcin-1 and stanniocalcin-2 in laryngeal squamous cell carcinoma and correlations with clinical and pathological parameters. PLoS One 9:e95466 Law AY, Wong CK, 2013, Stanniocalcin-1 and -2 promote angiogenic sprouting in HUVECs via VEGF/VEGFR2 and angiopoietin signaling pathways. Mol Cell Endocrinol 374:73–81 Yao YL, Ma J, Wang P et al, 2015, miR-101 acts as a tumor suppressor by targeting Kruppel-like factor 6 in glioblastoma stem cells. CNS Neurosci Ther 21:40–51 Yuan Q, Gao W, Liu B et al, 2014, Upregulation of miR-184 enhances the malignant biological behavior of human glioma cell line A172 by targeting FIH-1. Cell Physiol Biochem 34:1125–1136 Gao B, Gao K, Li L et al, 2014, miR-184 functions as an oncogenic regulator in hepatocellular carcinoma, HCC). Biomed Pharmacother 68:143–148 Zhen Y, Liu Z, Yang H et al, 2013, Tumor suppressor PDCD4 modulates miR-184-mediated direct suppression of C-MYC and BCL2 blocking cell growth and survival in nasopharyngeal carcinoma. Cell Death Dis 4:e872 Wagner GF, Jaworski E, 1994, Calcium regulates stanniocalcin mRNA levels in primary cultured rainbow trout corpuscles of stannius. Mol Cell Endocrinol 99:315–322 Shen XJ, Gu K, Shi JP et al, 2014, Increased expression of stanniocalcin 2 is associated with tumor progression after radiotherapy in patients with cervical carcinoma. Int J Clin Exp Pathol 7:8770–8776 Arigami T, Uenosono Y, Ishigami S et al, 2013, Clinical significance of stanniocalcin 2 expression as a predictor of tumor progression in gastric cancer. Oncol Rep 30:2838–2844 Wang H, Wu K, Sun Y et al, 2012, STC2 is upregulated in hepatocellular carcinoma and promotes cell proliferation and migration in vitro. BMB Rep 45:629–634 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 853 EP 860 DI 10.1007/s12253-017-0298-z PG 8 ER PT J AU Uehara, K Ikehara, F Shibuya, R Nakazato, I Oshiro, M Kiyuna, M Tanabe, Y Toyoda, Z Kurima, K Kina, Sh Hisaoka, M Kinjo, T AF Uehara, Karina Ikehara, Fukino Shibuya, Ryo Nakazato, Iwao Oshiro, Mariko Kiyuna, Masaya Tanabe, Yasuka Toyoda, Zensei Kurima, Kiyoto Kina, Shinichiro Hisaoka, Masanori Kinjo, Takao TI Molecular Signature of Tumors with Monoallelic 13q14 Deletion: a Case Series of Spindle Cell Lipoma and Genetically-Related Tumors Demonstrating a Link Between FOXO1 Status and p38 MAPK Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Monoallelic 13q14 deletion; RB 1; FOXO 1; Reactive oxygen species; p38 MAPK ID Monoallelic 13q14 deletion; RB 1; FOXO 1; Reactive oxygen species; p38 MAPK AB Spindle cell/pleomorphic lipomas (SCLs), cellul a r angiofibromas (CAFs) and mammary-type myofibroblastomas (MFBs) are rare benign mesenchymal tumors with monoallelic 13q14 deletion. They are predicted to have a common pathogenic mechanism due to shared similar histological and immunohistochemical features; however, pathological consequences of monoallelic 13q14 deletion remain unknown. We previously reported a CAF case with monoallelic 13q14 deletion in which the tumor expressed decreased levels of FOXO1 and RB1, both of which were encoded in 13q14, and increased reactive oxygen species (ROS) levels. We further demonstrated the activation of p38 mitogen-activated protein kinase (p38 MAPK) pathway induced by oxidative stress. We hypothesized that SCLs, CAFs and MFBs would share common molecular signatures involving FOXO1, ROS and p38 MAPK and that their expression patterns were different from those tumors without monoallelic 13q14 deletion such as solitary fibrous tumors (SFTs). We compared the expression levels of FOXO1, RB1, ROS markers and several signal transduction factors between SCLs and SFTs. SCLs expressed decreased levels of FOXO1 and RB1, whereas SFTs showed no change. Both tumor types exhibited increased markers of ROS; however, nuclear localization of phosphorylated p38 was significantly more frequent in SCLs than that in SFTs, suggesting p38 MAPK activation by oxidative stress. SFTs showed lower p38 MAPK activity and higher β- catenin expression, implying that oxidative stress was caused by increased cellular proliferation stress. Finally, CAFs and MFBs showed changes similar to those observed in SCLs. Overall, tumors with monoallelic 13q14 deletion showed shared molecular signatures that might be associated with pathogenesis. C1 [Uehara, Karina] University of the Ryukyus, School of Health Sciences, Department of Basic Laboratory Sciences, 207 Uehara, Nishihara, 903-0215 Okinawa, Japan. [Ikehara, Fukino] University of the Ryukyus, School of Health Sciences, Department of Basic Laboratory Sciences, 207 Uehara, Nishihara, 903-0215 Okinawa, Japan. [Shibuya, Ryo] University of Occupational and Environmental Health, School of Medicine, Department of Pathology and OncologyFukuoka, Japan. [Nakazato, Iwao] Okinawa Prefectural Nanbu Medical Center and Children’s Medical Center, Department of PathologyOkinawa, Japan. [Oshiro, Mariko] Meio University, Faculty of International Studies, Health Information Management Major, Management and Information Science DivisionOkinawa, Japan. [Kiyuna, Masaya] Tomishiro Chuo Hospital, Department of PathologyOkinawa, Japan. [Tanabe, Yasuka] University of the Ryukyus, School of Health Sciences, Department of Basic Laboratory Sciences, 207 Uehara, Nishihara, 903-0215 Okinawa, Japan. [Toyoda, Zensei] University of the Ryukyus, School of Health Sciences, Department of Basic Laboratory Sciences, 207 Uehara, Nishihara, 903-0215 Okinawa, Japan. [Kurima, Kiyoto] University of the Ryukyus, Graduate School of Medicine, Department of Regenerative MedicineOkinawa, Japan. [Kina, Shinichiro] University of the Ryukyus, Graduate School of Medicine, Department of Oral and Maxillofacial Functional RehabilitationOkinawa, Japan. [Hisaoka, Masanori] University of Occupational and Environmental Health, School of Medicine, Department of Pathology and OncologyFukuoka, Japan. [Kinjo, Takao] University of the Ryukyus, School of Health Sciences, Department of Basic Laboratory Sciences, 207 Uehara, Nishihara, 903-0215 Okinawa, Japan. RP Kinjo, T (reprint author), University of the Ryukyus, School of Health Sciences, Department of Basic Laboratory Sciences, 903-0215 Okinawa, Japan. EM kinjotko@med.u-ryukyu.ac.jp CR Flucke U, van Krieken JH, Mentzel T, 2011, Cellular angiofibroma: analysis of 25 cases emphasizing its relationship to spindle cell lipoma and mammary-type myofibroblastoma. Mod Pathol 24:82–89 Fritchie KJ, Carver P, Sun Y, Batiouchko G, Billings SD, Rubin BP, Tubbs RR, Goldblum JR, 2012, Solitary fibrous tumor: is there a molecular relationship with cellular angiofibroma, spindle cell lipoma, and mammary-type myofibroblastoma? Am J Clin Pathol 137:963–970 Hameed M, Clarke K, Amer HZ, Mahmet K, Aisner S, 2007, Cellular angiofibroma is genetically similar to spindle cell lipoma: a case report. Cancer Genet Cytogenet 177:131–134 Greer EL, Brunet A, 2005, FOXO transcription factors at the interface between longevity and tumor suppression. Oncogene 24:7410–7425 Huang H, Tindall DJ, 2007, Dynamic FoxO transcription factors. J Cell Sci 120:2479–2487 Maiese K, Chong ZZ, Shang YC, Hou J, 2009, A BFOXO^ in sight: targeting Foxo proteins from conception to cancer. Med Res Rev 29:395–418 Arakaki K, Chinen K, Kamiya M, Tanabe Y, Tawata N, Ikehara F, Uehara K, Shimabukuro H, Kinjo T, 2014, Evidence for an association between increased oxidative stress and derangement of FOXO1 signaling in tumorigenesis of a cellular angiofibroma with monoallelic 13q14: a case report. Int J Clin Exp Pathol 7:8972–8979 Sangoi AR, Dulai MS, Beck AH, Brat DJ, Vogel H, 2009, Distinguishing chordoid meningiomas from their histologic mimics: an immunohistochemical evaluation. Am J Surg Pathol 33:669–681 Chen BJ, Marino-Enriquez A, Fletcher CD, Hornick JL, 2012, Loss of retinoblastoma protein expression in spindle cell/ pleomorphic lipomas and cytogenetically related tumors: an immunohistochemical study with diagnostic implications. Am J Surg Pathol 36:1119–1128 Magro G, Righi A, Casorzo L, Antonietta T, Salvatorelli L, Kacerovska D, Kazakov D, Michal M, 2012, Mammary and vaginal myofibroblastomas are genetically related lesions: fluorescence in situ hybridization analysis shows deletion of 13q14 region. Hum Pathol 43:1887–1893 Adegbola AA, Cox GF, Bradshaw EM, Hafler DA, Gimelbrant A, Chess A, 2015, Monoallelic expression of the human FOXP2 speech gene. Proc Natl Acad Sci U S A 112:6848–6854 Wang X, Liu JZ, Hu JX, Wu H, Li YL, Chen HL, Bai H, Hai CX, 2011, ROS-activated p38 MAPK/ERK-Akt cascade plays a central role in palmitic acid-stimulated hepatocyte proliferation. Free Radic Biol Med 51:539–551 Torres M, Forman HJ, 2003, Redox signaling and the MAP kinase pathways. Biofactors 17:287–296 Beccafico S, Morozzi G, Marchetti MC, Riccardi C, Sidoni A, Donato R, Sorci G, 2015, Artesunate induces ROS- and p38 MAPK-mediated apoptosis and counteracts tumor growth in vivo in embryonal rhabdomyosarcoma cells. Carcinogenesis 36:1071–1083 Casini N, Forte IM, Mastrogiovanni G, Pentimalli F, Angelucci A, Festuccia C, Tomei V, Ceccherini E, Di Marzo D, Schenone S, Botta M, Giordano A, Indovina P, 2015, SRC family kinase, SFK, inhibition reduces rhabdomyosarcoma cell growth in vitro and in vivo and triggers p38 MAP kinase-mediated differentiation. Oncotarget 6:12421–12435 Matsuo T, Shimose S, Kubo T, Fujimori J, Yasunaga Y, Sugita T, Ochi M, 2012, Correlation between p38 mitogen-activated protein kinase and human telomerase reverse transcriptase in sarcomas. J Exp Clin Cancer Res 31:5 Weinberg F, Chandel NS, 2009, Reactive oxygen speciesdependent signaling regulates cancer. Cell Mol Life Sci 66: 3663–3673 Tong L, Chuang CC,Wu S, Zuo L, 2015, Reactive oxygen species in redox cancer therapy. Cancer Lett 367:18–25 TsunekiM, Nakamura Y, Kinjo T, Nakanishi R, Arakawa H, 2015, Mieap suppresses murine intestinal tumor via its mitochondrial quality control. Sci Rep 5:12472 Hoogeboom D, Essers MA, Polderman PE, Voets E, Smits LM, Burgering BM, 2008, Interaction of FOXO with betacatenin inhibits beta-catenin/T cell factor activity. J Biol Chem 283:9224–9230 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 861 EP 869 DI 10.1007/s12253-017-0303-6 PG 9 ER PT J AU Perez-Ruiz, E Rueda, A Perez, L Rivas-Ruiz, F Torres, E de Luque, V Alvarez, M Sevilla, I Redondo, M Padilla-Ruiz, M Alba, E Alonso, L AF Perez-Ruiz, Elisabeth Rueda, Antonio Perez, Lidia Rivas-Ruiz, Francisco Torres, Esperanza de Luque, Vanesa Alvarez, Martina Sevilla, Isabel Redondo, Maximino Padilla-Ruiz, Maria Alba, Emilio Alonso, Lorenzo TI Expression and Prognostic Value of Oestrogen Receptor Beta in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Prognostic factor; Estrogen receptor beta; Wild-type isoform ERβ1 ID Colorectal cancer; Prognostic factor; Estrogen receptor beta; Wild-type isoform ERβ1 AB Differences between men and women in the incidence and biological mechanisms of colorectal cancer (CRC) suggest that estrogens may play a role in the pathogenesis of this disease. The identification of the human estrogen receptor beta (ERβ) and its expression in the intestinal mucosa led to further studies that revealed that estrogens have a protective function against CRC mediated by the activation of ERβ. However, ERβ expression and its role in CRC is controversial. The purpose of this study was to determine the distribution and prognostic value of ERβ expression in the intestinal mucosa of patients diagnosed and surgically treated for CRC, and its association with other known prognostic factors. A total of 109 paraffin-embedded samples of the wild-type ERβ isoformwere analyzed by immunohistochemical nuclear staining in patients with colorectal adenocarcinoma. Clinical/pathological and survival data were collected. Immunohistochemical quantification was performed using the category scoring system, which has been validated for assessing estrogen receptor alfa. The wildtype ERβ isoform –also called ERβ1– was positive in 101 patients (92.7%) and negative in nine patients (7.3%). Univariate analysis revealed that the absence of expression of the ERβ1 gene was correlated with mucinous adenocarcinoma (p < 0.05). Also, a non-significant tendency was observed for ERβ expression to be down-regulated in advanced tumors.With a median follow-up of 47 months, the overall survival and progression-free survival were not found to be associated with ERβ1 expression (p = 0.2). Although thewild-type ERβ isoform was expressed in most study patients with colorectal cancer, it does not seem to have any prognostic value for the course of the disease. Further studies should be conducted to investigate whether the down-regulation of ERβ expression has any biological function in mucinous colorectal cancer. C1 [Perez-Ruiz, Elisabeth] Hospital Virgen de la Victoria, Division of Medical OncologyMalaga, Spain. [Rueda, Antonio] Hospital Costa del Sol, Division of Medical OncologyMarbella, Spain. [Perez, Lidia] Hospital Virgen de la Victoria, Patology DeparmentMalaga, Spain. [Rivas-Ruiz, Francisco] Red de Investigacion en Servicios de Salud (REDISSEC)Marbella, Spain. [Torres, Esperanza] Hospital Virgen de la Victoria, Division of Medical OncologyMalaga, Spain. [de Luque, Vanesa] Hospital Virgen de la Victoria, Patology DeparmentMalaga, Spain. [Alvarez, Martina] Hospital Virgen de la Victoria, Patology DeparmentMalaga, Spain. [Sevilla, Isabel] Hospital Virgen de la Victoria, Division of Medical OncologyMalaga, Spain. [Redondo, Maximino] Red de Investigacion en Servicios de Salud (REDISSEC)Marbella, Spain. [Padilla-Ruiz, Maria] Red de Investigacion en Servicios de Salud (REDISSEC)Marbella, Spain. [Alba, Emilio] Hospital Virgen de la Victoria, Division of Medical OncologyMalaga, Spain. [Alonso, Lorenzo] Hospital Virgen de la Victoria, Division of Medical OncologyMalaga, Spain. RP Perez-Ruiz, E (reprint author), Hospital Virgen de la Victoria, Division of Medical Oncology, Malaga, Spain. EM eliperu@gmail.com CR Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics. CA Cancer J Clin 63:11–30 Sanchez MJ, Payer T, de Angelis R, Larranaga R, Capocaccia R, Martinez C, CIBERESP working group, 2010, Cancer incidence and mortality in Spain: estimates and projections for the period 1981–2012. Ann Oncol 21:30–36 Howe HL, Wu X, Ries LA, Cokkinides V, Ahmed F, Jemal A et al, 2006, Annual report to the nation on the status of cancer, 1975– 2003, featuring cancer among U.S. Hispanic/Latino populations. Cancer 107:1711 Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA, 1996, Cloning of a novel receptor expressed in rat prostate and ovary. Proc Natl Acad Sci U S A 93:5925–5930 Mosselman S, Polman J, Dijkema R, 1996, ER beta: identification and characterization of a novel human estrogen receptor. FEBS Lett 392:49–53 Moore JT, Mckee DD, Slentz-Kesler K, Moore LB, Jones SA, Horne EL et al, 1998, Cloning and characterization of human estrogen receptor beta isoforms. Biochem Biophys Res Commun 247:75–78 Hasson RM, Briggs A, Carothers AM, Davids JS, Wang J, Javid SH et al, 2014, Estrogen receptor alfa or beta loss in the colon of min/+ mice promotes crypt expansion and impairs TGFbeta and HNF3beta signaling. Carinogenesis 35:96–102 Barone M, Tanzi S, Lofano K, Scavo MP, Pricci M, Demarinis L et al, 2010, Dietaryy-induced ERbeta upregulation counteracts intestinal neoplasia development in intact male ApcMin/+ mice. Carcinogenesis 31:269–274 Calle EE, Miracle-Mahill HL, Thun MJ, Heath CW Jr, 1995, Estrogen replacement therapy and risk of fatal colon cancer in a prospective cohort of postmenopausical women. J Nat Cancer Inst 87:517–523 Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML et al, 2002, Risk and benefits of estrogen plus progestin in healthy postmenopausal women: principal result from the Women’s health initiative randomized controlled trial. JAMA 288:321–333 Rudolph A, Toth C, Hoffmeister M, RothW, Herpel E, Schirmacher P et al, 2013, Colorectal cancer risk associated with hormone use varies by expression of estrogen receptor-beta. Cancer Res 73: 3306–3315 JassamN, Bell SM, Speirs V, Quirke P, 2005, Loss of expression of oestrogen receptor β in colon cancer and its association with dukes` staging. Oncol Rep 14:17–21 Fang YJ, Lu ZH, Wang F, Wu XJ, Li LR, Zhang LY et al, 2010, Prognostic impact of ERβ and MMP7 expression on overall survival in colon cancer. Tumor Biol 31:651–658 Reproducido con permiso del AJCC: Greene FL, Page PL, Fleming ID, et al. AJCC Cancer Staging Manual 6th ed. New York: Springer-Verlag, 2002 Grivas PD, Tzelepi V, Sotiropoulou-Bonikou G, Kefalopoulou Z, Papavassiliou AG, Kalofonos H, 2009, Expression of ERβ, ER alpha and co-regulator PELP1/MNAR in colorectal cancer: prognosis significance and clinopathologic correlations. Cell Oncol 31: 235–247 Elbanna HG, Ebrahim MA, Abbas AM, Zalata K, Hashim MA, 2012, Potential value of estrogen receptor beta expression in colorectal carcinoma: interaction with apoptotic index. J Gastrointest Cancer 43:56–62 Harvey JM, Clark GM, Osborne CK, Allred DC, 1999, Estrogen receptor status by immunohistochemistry is superior to the ligandbinding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol 17:1474–1481 Xie LQ, Yu JP, Luo HS, 2004, Expression of estrogen receptor β in human colorectal cancer. Word J Gastroenterol 10:214–217 Campbell-Thompson M, Lynch J, Bhardwaj B, 2001, Expression of estrogen receptor subtypes and Erβ isoforms in colon cancer. Cancer Res 61:632–640 Taggarshe D, Lobocki C, Silberberg B, McKendrick A, Mittal VK, 2012, Clinicopathological significance of the expression of estrogen receptor-beta and vascular endotelial growth factor-a in colorectal cancer. Am Surg 78:1376–1382 Rudolph A, Toth C, Hoffmeister M, Roth W, Herpel E, Jansen L et al, 2012, Expresion of oestrogen receptor β and prognosis of colorectal cancer. Br J Cancer 107:831–839 Rath-Wolfson L, Purim O, Ram E, Morgenstern S, Koren R, Brenner B. Expression of estrogen receptor β1 in colorectal cancer: correlation with clinicopathological variables. Oncology Reports 27:2017–2022 Konstantinopoulos PA, Kominea A, Vandoros G, Sykiotis GP, Andricopoulos P, Varakis I et al, 2003, Oestrogen receptor beta is abundantly expressed in normal colonic mucosa, but declines in colon adenocarcinoma paralleling the tumour’s dedifferentiation. Eur J Cancer 39:1251–1258 Castiglione F, Taddei A, DeglÍnnocenti R, Buccoliero AM, Bechi P, Garbini F et al, 2008, Expression of estrogen receptor β in colon cancer progression. Diagn Mol Pathol 17:231–236 Wong NA, Malcomson RD, Jodrell DI, Groome NP, Harrison DJ, 2005, Saunders PT.ERbeta isoform expression in colorectal carcinomas: an in vivo and in vitro study of clinicapathological and molecular correlates. J Pathol 207:53–60 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 871 EP 879 DI 10.1007/s12253-017-0301-8 PG 9 ER PT J AU Foley, MN Coll, J Lowery, A Hynes, OS Kerin, JM Sheehan, M Brodie, C Sweeney, K AF Foley, M Niamh Coll, M. Jill Lowery, J. Aoife Hynes, Oliver Sean Kerin, J Michael Sheehan, Margaret Brodie, Caroline Sweeney, J. Karl TI Re-Appraisal of Estrogen Receptor Negative/Progesterone Receptor Positive (ER−/PR+) Breast Cancer Phenotype: True Subtype or Technical Artefact? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pathology; Immunohistochemistry; Receptors; estrogen; Receptors; progesterone; Breast neoplasms ID Pathology; Immunohistochemistry; Receptors; estrogen; Receptors; progesterone; Breast neoplasms AB Expression of the ER and PR receptors is routinely quantified in breast cancer as a predictive marker of response to hormonal therapy. Accurate determination of ER and PR status is critical to the optimal selection of patients for targeted therapy. The existence of an ER−/PR+ subtype is controversial, with debate centred on whether this represents a true phenotype or a technical artefact on immunohistochemistry (IHC). The aim of this study was to investigate the true incidence and clinico-pathological features of ER−/PR+ breast cancers in a tertiary referral symptomatic breast unit. Clinico-pathological data were collected on invasive breast cancers diagnosed between 1995 and 2005. IHC for ER and PR receptors was repeated on all cases which were ER−/PR+, with the same paraffin block used for the initial diagnostic testing. Concordance between the diagnostic and repeat IHC was determined using validated testing. Complete data, including ER and PR status were available for 697 patients diagnosed during the study period. On diagnostic IHC, the immunophenotype of the breast tumours was: ER+/PR+ in 396 (57%), ER−/PR- in 157 (23%), ER+/PR- in 88 (12%) and ER−/PR+ in 56 (8.6%) patients. On repeat IHC of 48/56 ER−/PR+ tumours 45.8% were ER+/PR+, 6% were ER+/PR and 43.7% were ER−/PR- None of the cases were confirmed to be ER−/PR+. The ER−/PR+ phenotypic breast cancer is likely to be the result of technical artefact. Prompt reassessment of patients originally assigned to this subtype who represent with symptoms should be considered to ensure appropriate clinical management. C1 [Foley, M Niamh] University College Hospital Galway, Department of SurgeryGalway, Ireland. [Coll, M. Jill] University College Hospital Galway, Department of SurgeryGalway, Ireland. [Lowery, J. Aoife] University College Hospital Galway, Department of SurgeryGalway, Ireland. [Hynes, Oliver Sean] University College Hospital Galway, Department of SurgeryGalway, Ireland. [Kerin, J Michael] University College Hospital Galway, Department of SurgeryGalway, Ireland. [Sheehan, Margaret] University College Hospital Galway, Department of SurgeryGalway, Ireland. [Brodie, Caroline] University College Hospital Galway, Department of SurgeryGalway, Ireland. [Sweeney, J. Karl] University College Hospital Galway, Department of SurgeryGalway, Ireland. RP Foley, MN (reprint author), University College Hospital Galway, Department of Surgery, Galway, Ireland. EM foleynm@tcd.ie CR Anderson WF, Chatterjee N, Ershler WB et al, 2002, Estrogen receptor breast cancer phenotypes in the surveillance, epidemiology, and end results database. Breast Cancer Res Treat 76:27–36 Nadji M, Gomez-Fernandez C, Ganjei-Azar P et al, 2005, Immunohistochemistry of estrogen and progesterone receptors reconsidered: experience with 5,993 breast cancers. Am J Clin Pathol 123:21–27 Early Breast Cancer Trialists’ Collaborative Group, 2011, Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet 378(9793):771–784 Ng CH, Pathy NB, Taib NA, Mun KS, Rhodes A, Yip CH, 2012, The estrogen receptor negative-progesterone receptor positive breast carcinoma is a biological entity and not a technical artifact. Asian Pac J Cancer Prev 13(4):1111–1113 Maleki Z, Shariat S, Mokri M, Atri M, 2012, ER-negative /PRpositive breast carcinomas or technical artifacts in immunohistochemistry? Arch Iran Med 15(6):366–369 Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL et al, 2010, American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol 28(16):2784–2795 Rakha EA, El-Sayed ME, Green AR, Paish EC, Powe DG, Gee J et al, 2007, Biologic and clinical characteristics of breast cancer with single hormone receptor positive phenotype. J Clin Oncol 25(30):4772–4778 De Maeyer L, Van Limbergen E, De Nys K, Moerman P, Pochet N, HendrickxWet al, 2008, Does Estrogen receptor-negative/progesterone receptor-positive breast carcinoma exist? J Clin Oncol 26(2): 335–336 Colditz GA, Rosner BA, Chen WY, Holmes MD, Hankinson SE, 2004, Risk factors for breast cancer according to estrogen and progesterone receptor status. JNCI J Natl Cancer Inst 96(3):218–228 Yu KD, Di GH, Wu J, Lu JS, Shen KW, Lu GYet al, 2008, Breast cancer patients with estrogen receptor-negative/progesterone receptor-positive tumors: being younger and getting less benefit from adjuvant tamoxifen treatment. J Cancer Res Clin Oncol 134(12):1347–1354 Kiani J, Khan A, Khawar H, Shuaib F, Pervez S, 2006, ER-alpha negative and PR-positive breast cancer: lab error or clinical entity? Pathol Oncol Res 12(4):223–227 Shen T, Brandwein GenslerM, HameedO, Siegal GP,Wei S, 2015, Characterization of estrogen receptor–negative/progesterone receptor– positive breast cancer. Hum Pathol 46(11):1776–1784 Dunwald LK, Rossing MA, Li CI, 2007, Hormone receptor status, tumor characteristics, and prognosis: a prospective cohort of breast cancer patients. Breast Cancer Res 9(1):R6 Rhodes A, Jasani B, Balaton AJ, Barnes DM, Miller KD, 2000, Frequency of oestrogen and progesterone receptor positivity by immunohistochemical analysis in 7016 breast carcinomas: correlation with patient age, assay sensitivity, threshold value, and mammographic screening. J Clin Pathol 53:688–696 Gown AM(2008, Current issues in ER andHER2 testing by IHC in breast cancer. Mod Pathol 21(Suppl 2):S8–15 Schnitt SJ, 2006, Estrogen receptor testing of breast cancer in current clinical practice: What’s the question? J Clin Oncol 24(12): 1797–1799 Goldstein NS, Ferkowicz M, Odish E et al, 2003, Minimum formalin fixation time for consistent estrogen receptor immunohistochemical staining of invasive breast carcinoma. Am J Clin Pathol 120:86–92 Williams JH, Mepham BL, Wright DH, 1997, Tissue preparation for immunocytochemistry. J Clin Pathol 50:422–428 van Diest PJ, van Dam P, Henzen-Longmans SC et al, 1997, A scoring system for immunohistochemical staining: consensus report of the task force for basic research EORTC-GCCG. J Clin Pathol 50:801–804 Larsson LI, 1993, Tissue preparationmethods for light microscopic immunohistochemistry. Appl Immunohistochem 1:2–16 Rhodes A, Jasani B, Balaton AJ, Barnes DM, Anderson E, Bobrow LG et al, 2001, Study of interlaboratory reliability and reproducibility of estrogen andprogesterone receptor assays in Europe. Documentation of poor reliability and identification of insufficient microwave antigen retrieval time as a major contributory element of unreliable assays. Am J Clin Pathol 115(1):44–58 Harvey JM, Clark GM, Osborne CK, Allred DC, 1999, Estrogen Receptor Status by Immunohistochemistry Is Superior to the Ligand-Binding Assay for Predicting Response to Adjuvant Endocrine Therapy in Breast Cancer. J Clin Oncol 17(5):1474– 1481 Chan M, Chang MC, Gonzalez R, Lategan B, del Barco E, Vera- Badillo F et al, 2015, Outcomes of estrogen receptor negative and progesterone receptor positive breast cancer. PLoS One 10(7): e0132449 Hefti MM, Hu R, Knoblauch NW, Collins LC, Haibe-Kains B, Tamimi RM, Beck AH, 2013, Estrogen receptor negative/ progesterone receptor positive breast cancer is not a reproducible subtype. Breast Cancer Res 15(4):R68 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 881 EP 884 DI 10.1007/s12253-017-0304-5 PG 4 ER PT J AU Zhang, X Hu, L Du, M Wei, X Zhang, J Hui, Y Chen, Ch Li, G Hou, J AF Zhang, Xuefeng Hu, Linkun Du, Mingzhan Wei, Xuedong Zhang, Jun Hui, Yu Chen, Cheng Li, Gang Hou, Jianquan TI Eukaryotic Elongation Factor 2 (eEF2) is a Potential Biomarker of Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Eukaryotic elongation factor 2; Immunohistochemistry ID Prostate cancer; Eukaryotic elongation factor 2; Immunohistochemistry AB Eukaryotic elongation factor 2 (eEF2), a key regulator of protein synthesis, is involved in the progression of several types of cancer. This first study was to investigate the relationships between eEF2 protein and prostate cancer (PCa). Immunohistochemical staining was used to verify eEF2 protein in a set of 97 formalin-fixed, paraffin-embedded primary PCa tissues. Expression of eEF2 protein in positive cells was characterized by cytoplasmic staining. Correlations with clinicopathological factors were evaluated by Chi-square or Fisher’s exact probability tests. eEF2 protein was found in 74 out of 97 (76.29%) patients. eEF2-positive had higher PSA and Gleason score than negative in all patients. In addition, the positive expression of eEF2 protein was significantly associated with PSA and Gleason score (P = 0.007 and 0.002). However, no significant correlations occurred between expression of eEF2 protein and TNM stage (P = 0.292). In those eEF2 proteinpositive patients, we have found staining intensity of eEF2 protein was not only associated with PSA and Gleason score, but also associated with TNM stage (P = 0, 0.014 and 0.001, respectively). To conclude, our study indicates that expression of eEF2 protein is a potential biomarker for evaluating PCa. C1 [Zhang, Xuefeng] First Affiliated Hospital of Soochow University, Department of Urology, 899 Pinghai Road, 215031 Suzhou, China. [Hu, Linkun] First Affiliated Hospital of Soochow University, Department of Urology, 899 Pinghai Road, 215031 Suzhou, China. [Du, Mingzhan] First Affiliated Hospital of Soochow University, Department of Pathology, 899 Pinghai Road, 215031 Suzhou, China. [Wei, Xuedong] First Affiliated Hospital of Soochow University, Department of Urology, 899 Pinghai Road, 215031 Suzhou, China. [Zhang, Jun] First Affiliated Hospital of Soochow University, Department of Urology, 899 Pinghai Road, 215031 Suzhou, China. [Hui, Yu] First Affiliated Hospital of Soochow University, Department of Urology, 899 Pinghai Road, 215031 Suzhou, China. [Chen, Cheng] First Affiliated Hospital of Soochow University, Department of Urology, 899 Pinghai Road, 215031 Suzhou, China. [Li, Gang] First Affiliated Hospital of Soochow University, Department of Urology, 899 Pinghai Road, 215031 Suzhou, China. [Hou, Jianquan] First Affiliated Hospital of Soochow University, Department of Urology, 899 Pinghai Road, 215031 Suzhou, China. RP Hou, J (reprint author), First Affiliated Hospital of Soochow University, Department of Urology, 215031 Suzhou, China. EM xf192@163.com CR Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66:7–30 Zhu Y, Wang HK, Qu YY, Ye DW, 2015, Prostate cancer in East Asia: evolving trend over the last decade. Asian J Androl 17:48–57 Sundi D, Schaeffer EM(2016, Progress in prognosis and prediction for men with prostate cancer. Eur Urol 22:30761–30768 Grzmil M, Hemmings BA, 2012, Translation regulation as a therapeutic target in cancer. Cancer Res 72:3891–3900 Tash JS, Chakrasali R, Jakkaraj SR, Hughes J, Smith SK, Hornbaker K, Heckert LL, Ozturk SB, Hadden MK, Kinzy TG, Blagg BS, Georg GL, 2008, Gamendazole, an orally active indazole carboxylic acid male contraceptive agent, targets HSP90AB1(HSP90BETA, and EEF1A, eEF1A, and stimulates II 1a transcription in rat sertoli cells. Biol Reprod 78:1139–1152 Tomlison VA, Newbery HJ, Wray NR, Jackson J, Larionov A, Miller WR, Dixon JM, Abbott CM, 2005, Translation elongation factor eEF1A2 is a potential oncoprotein that is overexpressed in two-thirds of breast tumors. BMC Cancer 5:113–119 Nakamura J, Aoyagi S, Nanchi I, Nakatsuka S, Hirata E, Shibata S, Fukuda M, Yamamoto Y, Fukuda I, Tatsumi N, Ueda T, Fujiki F, Nomura M, Nishida S, Shirakata T, Hosen N, Tsuboi A, Oka Y, Nezu R, Mori M, Doki Y, Aozasa K, Sugiyama H, Oji Y, 2009, Overexpression of eukaryotic elongation factor eEF2 in gastrointestinal cancers and its involvement in G2/M progression in the cell cycle. Int J Oncol 34:1181–1189 Oji Y, Tatsumi N, Fukuda M, Nakatsuka S, Aoyagi S, Hirata E, Nanchi I, Fujiki F, Nakajima H, Yamamoto Y, Shibata S, Nakamru M, Hasegawa K, Takagi S, Fukada I, Hoshikawa T, Murakami Y, 2014, The translation elongation factor eEF2 is a novel tumorassociated antigen overexpressed in various types of cancers. Int J Oncol 44:1461–1469 Sun HG, Dong XJ, Lu T, Yang MF, Wang XM, 2014, Clinical value of eukaryotic elongation factor 2 in non-small cell lung cancer patients. Asian Pac J Cancer Prev 14:6533–6535 Tan HY, Wang N, Tsao SW, Zhang Z, Feng Y, 2014, Suppression of vascular endothelial growth factor via inactivation of eukaryotic elongation factor 2 by alkaloids in coptidis rhizome in hepatocellular carcinoma. Integr Cancer Ther 13:425–434 Rhoads RE, 1991, Protein synthesis, cell growth and oncogenesis. Curr Opin Cell Biol 3:1019–1024 Anand N, Murthy S, Amann G,Wernick M, Porter LA, Cukier IH, Collins C, Gray JW, Diebold J, Demetrick DJ, Lee JM, 2002, Protein elongation factor eEF1A2 is a putative oncogene in ovarian cancer. Nat Genet 31:301–305 Yoshizawa A, Fukuoka J, Shimizu S, Shimizu S, Shilo K, Franks TJ, Hewitt SM, Fujii T, Cordon-cardo C, Jen J, Travis WD, 2010, Overexpression of phosphor-eIF4E is associated with survival through AKT pathway in on-small cell lung cancer. Clin Cancer Res 16:240–248 Pinke DE, Kalloger SE, Francetic T, Huntsman DG, Lee JM, 2008, The prognostic significance of elongation factor eEF1A2 on ovarian cancer. Gynecol Oncol 108:561–568 Hagner PR, Schneider A, Gartenhaus RB, 2010, Targeting the translational machinery as a novel treatment strategy for hematologic malignancies. Blood 115:2127–2135 Chen CY, Fang HY, Chiou SH, Yi SE, Huang CY, Chiang SF, Chang HW, Lin TY, Chiang IP, Chow KC, 2011, Sumoylation of eukaryotic elongation factor 2 is vital for protein stability and anti-apoptotic activity in lung adenocarcinoma cells. Cancer Sci 102: 1582–1589 Bilanges B, Stokoe D, 2007, Mechanisms of translational deregulation in human tumors and therapeutic intervention strategies. Oncogene 26:5973–5970 Wang N, Feng YB, Tan HY, Cheung F, Hong M, Lao L, Nagamatsu T, 2015, Inhibition of eukaryotic elongation factor-2 confers to tumor suppression by a herbal formulation huanglian-jiedu decoction in human hepatocellular carcinoma. J Ethnopharmacol 164: 309–318 Nowroozi MR, Momeni SA, Ohadian Moghadam S, Ayati E, Mortazavi A, Arfae S, Jamshidian H, Taherimahmoudi M, Ayati M, 2016, Prostate-specific antigen density and Gleason score predict adverse pathologic features in patients with clinically localized prostate cancer. Nephrourol Mon 8:39984–39987 Kgatle MM, Kalla AA, Islam MM, Sathekge M, Moorad R, 2016, Prostate cancer: epigenetic alterations, risk factors, and therapy. Prostate Cancer 6:435–445 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 885 EP 890 DI 10.1007/s12253-017-0302-7 PG 6 ER PT J AU Kozak, D Glowacka-Mrotek, I Nowikiewicz, T Siedlecki, Z Hagner, W Sowa, M Zegarski, W AF Kozak, Dominika Glowacka-Mrotek, Iwona Nowikiewicz, Tomasz Siedlecki, Zygmunt Hagner, Wojciech Sowa, Magdalena Zegarski, Wojciech TI Analysis of Undesirable Sequelae of Sentinel Node Surgery in Breast Cancer Patients – a Prospective Cohort Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Sentinel lymph node; Undesirable sequelae ID Breast cancer; Sentinel lymph node; Undesirable sequelae AB Use of sentinel lymph node biopsy limits the frequency and severity of sequelae of surgical treatment. However, the procedure itself may not be completely free of complications. The goal of this work was to analyze prospectively the occurrence of undesirable sequelae in patients undergoing sentinel lymph node biopsy as an isolated intervention in the axillary fossa. This prospective observational study was conducted on a group of 104 women. Patients were examined on five occasions: one day before the procedure, one day after the procedure, one month, three months, and six months after the procedure. At every stage of the study they were assessed for tactile sensation, range of motion in the shoulder joint, upper limb circumference, sensation abnormalities, winged scapula sign, and pain severity according to Visual Analogue Scale (VAS). In the study group we observed statistically significant differences, such as limited mobility in the shoulder joint (p ≤ 0.01), gradual increase in limb circumference on the operated side (p < 0.01) and pain (p ≤ 0.01). Despite relatively low invasiveness of the procedure, sentinel lymph node biopsy is not entirely devoid of the risk of undesirable sequelae. C1 [Kozak, Dominika] Medical College of the Nicolaus Copernicus University in Torun, Clinic and Chair of Oncological SurgeryBydgoszcz, Poland. [Glowacka-Mrotek, Iwona] Collegium Medicum of the Nicolaus Copernicus University in Torun, Department of Rehabilitation, Maria Curie-Sklodowskiej Street 9, 85-094 Bydgoszcz, Poland. [Nowikiewicz, Tomasz] Medical College of the Nicolaus Copernicus University in Torun, Clinic and Chair of Oncological SurgeryBydgoszcz, Poland. [Siedlecki, Zygmunt] Collegium Medicum of the Nicolaus Copernicus University in Torun, Department of NeurosurgeryBydgoszcz, Poland. [Hagner, Wojciech] Collegium Medicum of the Nicolaus Copernicus University in Torun, Department of Rehabilitation, Maria Curie-Sklodowskiej Street 9, 85-094 Bydgoszcz, Poland. [Sowa, Magdalena] Medical College of the Nicolaus Copernicus University in Torun, Clinic and Chair of Oncological SurgeryBydgoszcz, Poland. [Zegarski, Wojciech] Medical College of the Nicolaus Copernicus University in Torun, Clinic and Chair of Oncological SurgeryBydgoszcz, Poland. RP Glowacka-Mrotek, I (reprint author), Collegium Medicum of the Nicolaus Copernicus University in Torun, Department of Rehabilitation, 85-094 Bydgoszcz, Poland. EM iwona.glowacka@cm.umk.pl CR Ivens D, Hoe AL, Podd TJ et al, 1992, Assessment of morbidity from complete axillarym dissection. Br J Cancer 66(1):136–138 McWayne J, Heiney SP, 2005, Psychologic and social sequelae of secondary lymphedema: a review. Cancer 104(3):457– 466. , DOI 10.1002/cncr.21195 Glowacka I, Nowikiewicz T, Siedlecki Z et al, 2016, The assessment of the magnitude of frontal plane postural changes in breast cancer patients after breast-conserving therapy or mastectomy - follow-up results 1 year after the surgical procedure. Pathol Oncol Res 22(1):203–208. , DOI 10.1007/s12253-015-9995-7 Albert US, Koller M, Kopp I et al, 2006, Early self-reported impairments in arm functioning of primary breast cancer patients predict late side effects of axillary lymph node dissection: results from a population-based cohort study. Breast Cancer Res Treat 100(3): 285–292. , DOI 10.1007/s10549-006-9247-3 Belmonte R, Garin O, Segura M et al, 2012, Quality-of-life impact of sentinel lymph node biopsy versus axillary lymph node dissection in breast cancer patients. Value Health 15: 907–915. , DOI 10.1016/j.jval.2012.06.003 Del Bianco P, Zavagno G, Burelli P et al, 2008, Morbidity comparison of sentinel node biosy versus conventional axillary lymph node dissectionu for breast cancer patients: results of the sentinella-Givom Italia randomised clinical trial. EJSO 34: 508–513. , DOI 10.1016/j.ejso.2007.05.017 Kootstra JJ, Hoekstra HJ, Rietman J et al, 2010, A longitudinal comparison of arm morbidity in stage I-II breast cancer patients treated with sentinel lymph node biopsy, sentinel lymph node biopsy followed by completion lymph node dissection, or axillary lymph node dissection. Ann Surg Oncol 17:2384–2394. , DOI 10.1245/s10434-010-0981-8 Land S, Kopec J, Julian T et al, 2010, Patient-reported outcomes in sentinel nodenegative adjuvant breast cancer patients receiving sentinel-node biopsy or axillary dissection: National Surgical Adjuvant Breast and bowel project phase III protocol B-32. J Clin Oncol 28:3929–3936. , DOI 10.1200/JCO.2010.28.2491 Schulze T, Mucke J, Markwardt J et al, 2006, Long-term morbidity of patients with early breast cancer after sentinel lymph node biopsy compared to axillary lymph node dissection. J Surg Oncol 29:109– 119. , DOI 10.1002/jso.20406 Mansel RE, Fallowfield L, Kissin M et al, 2006, Randomized Multicenter Trial of Sentinel Node Biopsy Versus Standard Axillary Treatment in Operable Breast Cancer: The ALMANAC Trial. Journal of the National Cancer Institute 98(9):599–609. , DOI 10.1093/jnci/djj158 Fleissig A, Fallowfield LJ, Langridge CI et al, 2006, Post-operative arm morbidity and quality of life. Results of the ALMANAC randomised trial comparing sentinel node biopsy with standard axillary treatment in the management of patients with early breast cancer. Breast Cancer Res Treat 95:279–293. , DOI 10. 1007/s10549-005-9025-7 Giuliano AE, Haigh PI, Brennan MB et al, 2000, Prospective observational study of sentinel lymphadenectomy without further axillary dissection in patients with sentinel node-negative breast cancer. J Clin Oncol 18:2553–2559. , DOI 10.1200/JCO.2000. 18.13.2553 Purushotham A, Uponi S, Klevesath M et al, 2005, Morbidity after sentinel lymph node biopsy in primary breast cancer: results from a randomized controlled trial. J Clin Oncol 23: 4312–4321. , DOI 10.1200/JCO.2005.03.228 Liu C, Guo Y, Shi J et al, 2009, Late morbidity associated with a tumour-negative sentinel lymph node biopsy in primary breast cancer patients: a systematic review. Eur J Cancer 45:1560–1568. , DOI 10.1016/j.ejca.2009.02.012 Favarao K, Mantse J, Barros A, 2010, Shoulder mobility after axillary sentinel lymph node biopsy for early infiltrating breast cancer treatment. European Journal of Gynaecologic Oncology 31:23–26 Hack T, Kwan W, Thomas-MacLean R et al, 2010, Predictors of arm morbidity following breast cancer surgery. Psycho-Oncology 19:1205–1212. , DOI 10.1002/pon.1685 Martin RC, EdwardsMJ,Wong SL et al, 2000, Practical guidelines for optimal gamma probe detection of sentinel lymph nodes in breast cancer: results of a multi-institutional study. For the University of Louisville Breast Cancer Study Group. Surgery 128(2):139–144 Berg JW, 1955, The significance of axillary node levels in the study of breast carcinoma. Cancer 8:776–778 Peintinger F, Reitsamer R, Stranzl H et al, 2003, Comparison of quality of life and arm complaints after axillary lymph node dissection vs sentinel lymph node biopsy in breast cancer patients. Br J Cancer 89:648–652. , DOI 10.1038/sj.bjc.6601150 DiSipio T, Rye S, Newman B et al, 2013, Incidence of unilateral arm lymphoedema after breast cancer: a systematic review and meta-analysis. The Lancet Oncology 14(6):500– 515. , DOI 10.1016/S1470-2045(13)70076-7 Bulley C, Coutts F, Blyth C et al, 2013, Prevalence and impacts of upper limb morbidity after treatment for breast cancer: a crosssectional study of lymphedema and function. Cancer and Oncology Research 1(2):30–39. , DOI 10.1016/j.ejon. 2013.10.006 Mc Laughlin S, Wright M, Morris K et al, 2008, Prevalence in lymphedema in women with breast cancer 5 years after sentinel lymph node biopsy or axillary dissection: objective measurements. J Clin Oncol 26(32):5213–5219. , DOI 10.1200/JCO. 2008.16.3725 Gӧker M, Devoogdt N, Van De Putte G et al, 2013, Systematic review of breast cancer related lymphoedema: making a balanced decision to perform an axillary clearance. Facts, Views Vis Obstet Gynaecol 5(2):106–115 Ozcinar B, Guler S, Kocaman N et al, 2012, Breast cancer related lymphedema in patients with different loco-regional treatments. Breast 21(3):361–365 Togawa K, Huiyan M, Sullivan-Halley J et al, 2014, Risk factors for self-reported arm lymphedema among female breast cancer survivors: a prospective cohort study. Breast Cancer Res 16:414–419. , DOI 10.1016/j.breast.2012.03.002 Mansel R, MacNeill F, Horgan K et al, 2013, Results of a national training programme in sentinel lymph node biopsy for breast cancer. Br J Surg 100:654–661. , DOI 10.1002/bjs.9058 Khavanin N, Gart M, Berry T et al, 2014, Sentinel lymph node biopsy versus axillary lymphadenectomy in patients treated with lumpectomy: an analysis of short-term outcomes. Ann Surg Oncol 21(1):74–80. , DOI 10.1245/s10434-013-32483 Verbelen H, Gebruers N, Eckhout F et al, 2014, Shoulder and arm morbidity in sentinel node-negative breast cancer patients: a systematic review. Breast Cancer Res Treat 144: 21–31. , DOI 10.1007/s10549-014-2846-5 Petrek J, Senie RT, Peters M, Rosen PP, 2001, Lymphedema in a cohort of breast carcinoma survivors 20 years after diagnosis. Cancer 92(6):1368–1377 Lucci A, McCall M, Beitsch M et al, 2007, Surgical complications associated with sentinel lymph node dissection, SLND, plus axillary lymph node dissection compared with SLNB alone in the American College of Surgeons oncology group trial Z0011. J Clin Oncol 25:3657–3663. , DOI 10.1200/JCO.2006.07.4062 Francis W, Abghari P, Du W et al, 2006, Improving surgical outcomes: standardizing the reporting of incidence and severity of acute lymphedema after sentinel lymph node biopsy and axillary lymph node dissection. Am J Surg 192:363–369. , DOI 10. 1016/j.amjsurg.2006.08.018 Kootstra JJ, Dijkstra PU, Rietman H et al, 2013, A longitudinal study of shoulder and arm morbidity in breast cancer survivors 7 years after sentinel lymph node biopsy or axillary lymph node dissection. Breast Cancer Res Treat 139:125–134. , DOI 10. 1007/s10549-013-2509-y NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 891 EP 897 DI 10.1007/s12253-017-0306-3 PG 7 ER PT J AU Yang, L Yang, Z Li, D Liu, Z Zou, Q Yuan, Y Xu, H AF Yang, Liangliang Yang, Zhulin Li, Daiqiang Liu, Ziru Zou, Qiong Yuan, Yuan Xu, Huilan TI Overexpression of FZD1 and CAIX are Associated with Invasion, Metastasis, and Poor-Prognosis of the Pancreatic Ductal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic ductal adenocarcinoma; Immunohistochemistry; FZD1; CAIX ID Pancreatic ductal adenocarcinoma; Immunohistochemistry; FZD1; CAIX AB Approximately 80% of patients with pancreatic ductal adenocarcinoma (PDAC) have metastatic disease with poor prognosis, but clinically available biomarkers have not yet been identified. This study was to investigate the clinical significance of FZD1 and CAIX in PDACs. FZD1 and CAIX protein expression was measured using EnVision immunohistochemistry. Positive FZD1 or CAIX expression was significantly higher in PDAC than that in precursor lesions (p < 0.01). Positive FZD1 or CAIX expression was significantly lower in cases with well-differentiated adenocarcinoma, no-metastasis of the lymph node, no-invasion of regional tissues, and TNM I/II stage disease than in cases with poorlydifferentiated adenocarcinoma, metastasis and invasion, and TNM stage III+ IV stage disease (p < 0.05 or p < 0.01). The expression of FZD1 positively correlated with CAIX in PDAC (P = 0.000). Univariate Kaplan-Meier analysis showed that FZD1 and/or CAIX expression (p < 0.001) was significantly associated with shorter overall survival (p < 0.05).Cox multivariate analysis showed that differentiation, tumor mass, lymph node metastasis, invasion, TNM stage, FZD1 and CAIX levels negatively correlated with overall survival. Positive FZD1 and CAIX expressions are poor prognostic factors in PDAC patients. FZD1 and CAIX might be important biological markers for the carcinogenesis, metastasis, invasion, and prognosis of PDAC. C1 [Yang, Liangliang] Central South University, Public Health College, 410078 Changsha, Hunan, China. [Yang, Zhulin] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary DiseasesChangsha, Hunan, China. [Li, Daiqiang] Central South University, Second Xiangya Hospital, Department of PathologyChangsha, Hunan, China. [Liu, Ziru] Central South University, Second Xiangya Hospital, Research Laboratory of Hepatobiliary DiseasesChangsha, Hunan, China. [Zou, Qiong] Central South University, Third Xiangya Hospital, Department of PathologyChangsha, Hunan, China. [Yuan, Yuan] Central South University, Third Xiangya Hospital, Department of PathologyChangsha, Hunan, China. [Xu, Huilan] Central South University, Public Health College, 410078 Changsha, Hunan, China. RP Xu, H (reprint author), Central South University, Public Health College, 410078 Changsha, China. EM xhl6363@126.com CR Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ, 2009, Cancer statistics, 2009. CA Cancer J Clin 59:225–249 Puleo F, Marechal R, Demetter P, Bali MA, Bali MA, Calomme A, Closset J, Bachet JB, Deviere J, Van Laethem JL, 2015, New challenges in perioperative management of pancreatic cancer. World J Gastroenterol 21:2281–2293 Von Hoff DD, Ervin T, Arena FP et al, 2013, Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 369:1691–1703 Kahlert UD, Mooney SM, Natsumeda M, Steiger HJ, Maciaczyk J, 2017, Targeting cancer stem-like cells in glioblastoma and colorectal cancer through metabolic pathways. Int J Cancer 140:10–22 EckertAW, Kappler M, Schubert J, Taubert H, 2012, Correlation of expression of hypoxia-related proteins with prognosis in oral squamous cell carcinoma patients. Oral Maxillofac Surg 16:189–196 McDonald PC, Winum JY, Supuran CT, Dedhar S, 2012, Recent developments in targeting carbonic anhydrase IX for cancer therapeutics. Oncotarget 3:84–97 Parks SK, Chiche J, Pouyssegur J, 2011, pH control mechanisms of tumor survival and growth. J Cell Physiol 226:299–308 Swietach P, Patiar S, Supuran CT, Harris AL, Vaughan-Jones RD, 2009, The role of carbonic anhydrase 9 in regulating extracellular and intracellular ph in three-dimensional tumor cell growths. J Biol Chem 284:20299–20310 Shin HJ, Rho SB, Jung DC, Han IO, Oh ES, Kim JY, 2011, Carbonic anhydrase IX, CA9, modulates tumor-associated cell migration and invasion. J Cell Sci 124:1077–1087 Pastorekova S, Parkkila S, Pastorek J, Supuran CT, 2004, Carbonic anhydrases:current state of the art, therapeutic applications and future prospects. J Enz Inhib Med Chem 19:199–229 Potter C, Harris AL, 2004, Hypoxia inducible carbonic anhydrase IX, a marker of tumour hypoxia, survival pathway and therapy target. Cell Cycle 3:164–167 Lehtonen J, Shen B,Vihinen M, Casini A, ScozzafavaA, Supuran CT, Parkkila AK, Saarnio J, Kivela AJ, Waheed A, Sly WS, Parkkila S, 2004, Characterization of CAXIII, a novel member of the carbonic anhydrase isozyme family. J Biol Chem 279:2719–2727 TureciO, Sahin U,Vollmar E, Siemer S,Gottert E, Seitz G, Parkkila AK, Shah GN, Grubb JH, Pfreundschuh M, SlyWS, 1998, Human carbonic anhydrase XII: cDNA cloning, expression, and chromosomal localization of a carbonic anhydrase gene that is overexpressed in some renal cell cancers. Proc Natl Acad Sci USA 95:7608–7613 Opavsky R, Pastorekova S, Zelnik V, Gibadulinova A, Stanbridge EJ, Zavada J, Kettmenn R, Pastorek J, 1996, Human MN/CA9 gene, a novel member of the carbonic anhydrase family: structure and exon to protein domain relationships. Genomics 33:480–487 Supuran CT, 2008, Carbonic anhydrases-an overview. Curr Pharm Des 14:603–614 Leppilampi M, Saarnio J, Karttunen TJ, Kivela J, Pastorekova S, Pastorek J, Waheed A, Sly WS, Parkkila S, 2003, Carbonic anhydrase isozymes IX and XII in gastric tumors. World J Gastroenterol 9:1398–1403 Span PN, Bussink J, Manders P, 2003, Carbonic anhydrase IX expression levels and prognosis in human breast cancer: association with treatment outcome. Br J Cancer 89:271–276 Liao SY, Darcy KM, Randall LM, Tian C, Monk BJ, Burger RA, Fruehauf JP, PetersWA, Stock RJ, Stanbridge EJ, 2010, Prognostic relevance of carbonic anhydrase-IX in high-risk, early-stage cervical cancer:a Gynecologic Oncology Group study. Gynecol Oncol 116:452–458 Yokoyama S, Higashi M, Kitamoto S et al, 2016, Aberrant methylation of MUC1 and MUC4 promoters are potential prognostic biomarkers for pancreatic ductal adenocarcinomas. Oncotarget 7: 42553–42565 YuM, Zhou Q, Zhou Y, Fu Z, Tan L,Ye X, Zeng B, GaoW, Zhou J, Liu Y, Li Z, Lin Y, Lin Q, Chen R, 2015, Metabolic phenotypes in pancreatic cancer. PLoS One 10:e0115153 Hoang BH, Kubo T, Healey JH, Sowers R, Mazza B, Yang R, Huvos AG, Meyers PA, Gorlick R, 2004, Expression of LDL receptor-related protein 5, LRP5, as a novel marker for disease progression in high-grade osteosarcoma. Int J Cancer 109:106–111 Salsano E, Paterra R, Figus M, Menghi F, Maderna E, Pollo B, Solero CL, Massimi L, Finocchiaro G, 2012, Expression profile of frizzled receptors in human medulloblastomas. J Neuro oncol 106:271–280 Udartseva OO, Andreeva ER, Buravkova LB, 2015, WNTassociated gene expression in human mesenchymal stromal cells under hypoxic stress. Dokl Biochem Biophys 465:354–357 Cui XP, Xing Y, Chen JM, Dong SW, Ying DJ, Yew DT, 2011, Wnt/beta-catenin is involved in the proliferation of hippocampal neural stem cells induced byhypoxia. Ir J Med Sci 180:387–393 Liu HL, Liu D, Ding GR, Liao PF, Zhang JW, 2015, Hypoxiainducible factor-1α and Wnt/β-catenin signaling pathways promote the invasion of hypoxic gastriccancer cells. Mol Med Rep 12:3365–3373 Su H, Jin X, Zhang X, Zhao L, Lin B, Li L, Fei Z, Shen L, Fang Y, Pan H, Xie C, 2015, FH535 increases the radiosensitivity and reverses epithelial-to-mesenchymal transition of radioresistant esophageal cancer cell line KYSE-150R. J Transl Med 13:104 Milovanovic T, Planutis K, Nguyen A, Marsh JL, Lin F, Hope C, Holcombe RF, 2004, Expression ofWnt genes and frizzled 1 and 2 receptors in normal breast epithelium and infiltrating breast carcinoma. Int J Oncol 25:1337–1342 Merle P, Kim M, Herrmann M, Gupte A, Lefrancois L, Califano S, Trepo C, Tanaka S, Vitvitski L, de la Monte S, Wands JR, 2005, Oncogenic role of the frizzled-7/beta-catenin pathway in hepatocellular carcinoma. J Hepatol 43:854–862 Ueno K, HiuraM, Suehiro Y, Hazama S, Hirata H, OkaM, Imai K, Dahiya R, Hinoda Y, 2008, Frizzled-7 as a potential therapeutic target in colorectal cancer. Neoplasia 10:697–705 Holcombe RF, Marsh JL, Waterman ML, Lin F, Milovanovic T, Truong T, 2002, Expression of Wnt ligands and frizzled receptors in colonic mucosa and in colon carcinoma. Mol Pathol 55:220–226 Liu H, Chen Y, Zhou F, Jie L, Pu L, Ju J, Li F, Dai Z,Wang X, Zhou S, 2014, Sox9 regulates hyperexpression of Wnt1 and Fzd1 in human osteosarcoma tissues and cells. Int J Clin Exp Pathol 7: 4795–4805 Svastova E, Zilka N, Zat’ovicova M, Gibadulinova A, Ciampor F, Pastorek J, Pastorekova S, 2003, Carbonic anhydrase IX reduces Ecadherin- mediated adhesion of MDCK cells via interaction with bcatenin. Exp Cell Res 290:332–345 Jain RK, 1999, Transport of molecules, particles, and cells in solid tumours. Annu Rev Biomed Eng 1:241–321 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 899 EP 906 DI 10.1007/s12253-017-0284-5 PG 8 ER PT J AU Tang, L Wang, D Gu, D AF Tang, Liang Wang, Dong Gu, Dongyun TI Knockdown of Sox2 Inhibits OS Cells Invasion and Migration via Modulating Wnt/β-Catenin Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; Sox2; Wnt/β-catenin ID Osteosarcoma; Sox2; Wnt/β-catenin AB Osteosarcoma (OS) was a prevalent malignant bone tumor which threatens people’s health worldwide.Wnt/β catenin signaling pathway had been proved significant in various cancers, indicating its possible function in OS as well. Sox2, a crucial member among SOX family could regulate cells biologically. How Sox2 modulated Wnt/β catenin signaling pathway in OS remained to be discussed. The study aimed to investigate the effects of Sox2 on the invasion and migration of OS cells and the related molecular mechanisms. Twenty-four human OS and adjacent tissue samples were involved in this study. Human OS cell lines MG63 and HOS were selected for further investigation. The liposome carrier si-Sox2 which could interfere with the expression of Sox2 gene was built to transfect MG63 and HOS cells). QRT-PCR assay and western blot were utilized to analyze the expression ofm RNA and proteins of Sox2. Transwell assay and wound healing assay were conducted to test the invasion and migration level of cells. The expression of GSK3, β-catenin, cyclin D1 and c-myc proteins were detected by western blot assay after transfection with si-Sox2. Compared with normal tissues and cells, the expression of Sox2 in OS tissues and cells was significantly higher. The mRNA and protein levels of Sox2 significantly decreased after transfection with si-Sox2. The invasion and migration of OS cells were down-regulated significantly through the inhibition of Sox2 by inactivating Wnt/β-catenin signaling pathway related proteins. Knockdown of Sox2 could inhibit invasion and migration of OS cells via modulating Wnt/β-catenin signaling pathway. C1 [Tang, Liang] Shanghai Jiao Tong University School of Medicine, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Key Laboratory of Orthopaedic Implants, 200011 Shanghai, China. [Wang, Dong] Shanghai Jiao Tong University School of Medicine, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Key Laboratory of Orthopaedic Implants, 200011 Shanghai, China. [Gu, Dongyun] Shanghai Jiao Tong University School of Medicine, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Key Laboratory of Orthopaedic Implants, 200011 Shanghai, China. RP Gu, D (reprint author), Shanghai Jiao Tong University School of Medicine, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Key Laboratory of Orthopaedic Implants, 200011 Shanghai, China. EM dongyungu@sjtu.edu.cn CR Niu G, Li B, Sun L, An C, 2015)MicroRNA-153 inhibits osteosarcoma cells proliferation and invasion by targeting TGF-beta2. PLoS One 10(3):e0119225. , DOI 10.1371/journal.pone.0119225 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108. , DOI 10.3322/caac.21262 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, XQ Y, He J, 2016, Cancer statistics in China, 2015. CA Cancer J Clin 66(2):115–132. , DOI 10.3322/caac.21338 Isakoff MS, Bielack SS, Meltzer P, Gorlick R, 2015, Osteosarcoma: current treatment and a collaborative pathway to success. J Clin Oncol Off J Am Soc Clin Oncol 33(27):3029– 3035. , DOI 10.1200/JCO.2014.59.4895 Zhang RX, Li Y, Tian DD, Liu Y, NianW, Zou X, Chen QZ, Zhou LY, Deng ZL, He BC, 2016, Ursolic acid inhibits proliferation and induces apoptosis by inactivatingWnt/beta-catenin signaling in human osteosarcoma cells. Int J Oncol 49(5):1973–1982. https://doi. org/10.3892/ijo.2016.3701 Nataraj V, Batra A, Rastogi S, Khan SA, Sharma MC, Vishnubhatla S, Bakhshi S, 2015, Developing a prognostic model for patients with localized osteosarcoma treated with uniform chemotherapy protocol without high dose methotrexate: a single-center experience of 237 patients. J Surg Oncol 112(6):662–668. , DOI 10.1002/jso.24045 Basu-Roy U, Seo E, Ramanathapuram L, Rapp TB, Perry JA, Orkin SH, Mansukhani A, Basilico C, 2012, Sox2 maintains self renewal of tumor-initiating cells in osteosarcomas. Oncogene 31(18):2270–2282. , DOI 10.1038/onc.2011.405 Ebb D, Meyers P, Grier H, Bernstein M, Gorlick R, Lipshultz SE, Krailo M, Devidas M, Barkauskas DA, Siegal GP, Ferguson WS, Letson GD, Marcus K, Goorin A, Beardsley P, Marina N, 2012, Phase II trial of trastuzumab in combination with cytotoxic chemotherapy for treatment of metastatic osteosarcoma with human epidermal growth factor receptor 2 overexpression: a report from the children's oncology group. J Clin Oncol Off J Am Soc Clin Oncol 30(20):2545–2551. , DOI 10.1200/JCO.2011.37.4546 Chou AJ, Kleinerman ES, KrailoMD, Chen Z, Betcher DL, Healey JH, Conrad EU 3rd, NiederML,WeinerMA,Wells RJ,Womer RB, Meyers PA, Children's Oncology G, 2009, Addition of muramyl tripeptide to chemotherapy for patients with newly diagnosed metastatic osteosarcoma: a report from the Children's Oncology Group. Cancer 115(22):5339–5348. , DOI 10.1002/cncr.24566 Endicott AA, Morimoto LM, Kline CN, Wiemels JL, Metayer C, Walsh KM, 2017, Perinatal factors associated with clinical presentation of osteosarcoma in children and adolescents. Pediatr Blood Cancer 64(6). , DOI 10.1002/pbc.26349 Alfranca A, Martinez-Cruzado L, Tornin J, Abarrategi A,Amaral T, de Alava E, Menendez P, Garcia-Castro J, Rodriguez R, 2015, Bone microenvironment signals in osteosarcoma development. Cell Mol Life Sci 72(16):3097–3113. , DOI 10.1007/ s00018-015-1918-y Zhu XB, Zhang ZC, Han GS, Han JZ, Qiu DP, 2017, Overexpression of miR214 promotes the progression of human osteosarcoma by regulating the Wnt/betacatenin signaling pathway. Mol Med Rep 15(4):1884–1892. , DOI 10.3892/ mmr.2017.6203 Zhan P, Zhang B, Xi GM, Wu Y, Liu HB, Liu YF, WJ X, Zhu QQ, Cai F, Zhou ZJ, Miu YY, Wang XX, Jin JJ, Li Q, Qian LP, Lv TF, Song Y, 2017, PRC1 contributes to tumorigenesis of lung adenocarcinoma in association with the Wnt/beta-catenin signaling pathway. Mol Cancer 16(1):108. , DOI 10. 1186/s12943-017-0682-z Peng L, Liu Z, Xiao J, Tu Y,Wan Z, Xiong H, Li Y, XiaoW(2017, MicroRNA-148a suppresses epithelial-mesenchymal transition and invasion of pancreatic cancer cells by targeting Wnt10b and inhibiting the Wnt/beta-catenin signaling pathway. Oncol Rep 38(1):301–308. , DOI 10.3892/or.2017.5705 Zhu B, Cheng D, Li S, Zhou S, Yang Q, 2016, High expression of XRCC6 promotes human osteosarcoma cell proliferation through the beta-catenin/Wnt signaling pathway and is associated with poor prognosis. Int J Mol Sci 17(7). , DOI 10.3390/ ijms17071188 Lin CH, Ji T, Chen CF, Hoang BH, 2014, Wnt signaling in osteosarcoma. Adv Exp Med Biol 804:33–45. , DOI 10.1007/ 978-3-319-04843-7_2 Cai Y, Mohseny AB, Karperien M, Hogendoorn PC, Zhou G, Cleton-Jansen AM, 2010, Inactive Wnt/beta-catenin pathway in conventional high-grade osteosarcoma. J Pathol 220(1):24–33. , DOI 10.1002/path.2628 Jin Z, Han YX, Han XR, 2013, Degraded iota-carrageenan can induce apoptosis in human osteosarcoma cells via the Wnt/betacatenin signaling pathway. Nutr Cancer 65(1):126–131. https:// doi.org/10.1080/01635581.2013.741753 Yang C, Hou C, Zhang H, Wang D, Ma Y, Zhang Y, Xu X, Bi Z, Geng S, 2013, miR-126 functions as a tumor suppressor in osteosarcoma by targeting Sox2. Int J Mol Sci 15(1):423–437. https:// doi.org/10.3390/ijms15010423 Moradi A, Ghasemi F, Anvari K, Hassanian SM, Simab SA, Ebrahimi S, Hesari A, Forghanifard MM, Boroushaki MT, ShahidSales S, Avan A, 2017, The cross-regulation between SOX15 and Wnt signaling pathway. J Cell Physiol 232(12): 3221–3225. , DOI 10.1002/jcp.25802 Hussenet T, duManoir S, 2010, SOX2 in squamous cell carcinoma: amplifying a pleiotropic oncogene along carcinogenesis. Cell Cycle 9(8):1480–1486. , DOI 10.4161/cc.9.8.11203 Ten Kate FJC, van Olphen SH, Bruno MJ, Wijnhoven BPL, van Lanschot JJB, Looijenga LHJ, Fitzgerald RC, Biermann K, 2017, Loss of SRY-box2, SOX2, expression and its impact on survival of patients with oesophageal adenocarcinoma. Br J Surg 104(10): 1327–1337. , DOI 10.1002/bjs.10553 He J, Shi J, Zhang K, Xue J, Li J, Yang J, Chen J,Wei J, Ren H, Liu X, 2017, Sox2 inhibits Wnt-beta-catenin signaling and metastatic potency of cisplatin-resistant lung adenocarcinoma cells. Mol Med Rep 15(4):1693–1701. , DOI 10.3892/mmr.2017.6170 Li J, Shen J, Wang K, Hornicek F, Duan Z, 2016, The roles of sox family genes in sarcoma. Curr Drug Targets 17(15):1761–1772 Liu H, Chen Y, Zhou F, Jie L, Pu L, Ju J, Li F, Dai Z,Wang X, Zhou S, 2014, Sox9 regulates hyperexpression of Wnt1 and Fzd1 in human osteosarcoma tissues and cells. Int J Clin Exp Pathol 7(8): 4795–4805 Bao Y, Chen B, Wu Q, Hu K, Xi X, Zhu W, Zhong X, Chen J, 2017, Overexpression of miR-664 is associated with enhanced osteosarcoma cell migration and invasion ability via targeting SOX7. Clin Exp Med 17(1):51–58. , DOI 10.1007/ s10238-015-0398-6 Mansukhani A, Ambrosetti D, Holmes G, Cornivelli L, Basilico C, 2005, Sox2 induction by FGF and FGFR2 activating mutations inhibits Wnt signaling and osteoblast differentiation. J Cell Biol 168(7):1065–1076. , DOI 10.1083/jcb.200409182 Zou Y, Huang Y, Yang J, Wu J, Luo C, 2017, miR-34a is downregulated in human osteosarcoma stem-like cells and promotes invasion, tumorigenic ability and self-renewal capacity.Mol Med Rep 15(4):1631–1637. , DOI 10.3892/mmr.2017.6187 Ren C, Ren T, Yang K, Wang S, Bao X, Zhang F, Guo W, 2016, Inhibition of SOX2 induces cell apoptosis and G1/S arrest in Ewing's sarcoma through the PI3K/Akt pathway. J Exp Clin Cancer Res 35:44. , DOI 10.1186/s13046-016-0321-3 Martins-Neves SR, Corver WE, Paiva-Oliveira DI, van den Akker BE, Briaire-de-Bruijn IH, Bovee JV, Gomes CM, Cleton-Jansen AM, 2016, Osteosarcoma stem cells have active Wnt/beta-catenin and Overexpress SOX2 and KLF4. J Cell Physiol 231(4):876–886. , DOI 10.1002/jcp.25179 Li C, Shi X, Zhou G, Liu X, Wu S, Zhao J, 2013, The canonical Wnt-beta-catenin pathway in development and chemotherapy of osteosarcoma. Front Biosci 18:1384–1391 Krishnan V, Bryant HU, Macdougald OA, 2006, Regulation of bone mass by Wnt signaling. J Clin Invest 116(5):1202–1209. , DOI 10.1172/JCI28551 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 907 EP 913 DI 10.1007/s12253-018-0400-1 PG 7 ER PT J AU Crowley, PM Prabhakaran, NV Gilligan, MO AF Crowley, P Maeve Prabhakaran, N Vinitha Gilligan, M Oonagh TI Incidence of Contrast-Induced Nephropathy in Patients with Multiple Myeloma Undergoing Contrast-Enhanced Procedures SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Multiple myeloma; Contrast induced nephropathy; Complications ID Multiple myeloma; Contrast induced nephropathy; Complications AB Multiple myeloma (MM) is a malignant disorder characterized by clonal proliferation of plasma cells. Renal impairment is a common complication. Contrast-induced nephropathy (CIN) is a form of acute renal failure that can occur in the setting of IV contrast administration. It is more commonly seen in patients with pre-existing renal impairment. Patients with MM commonly require contrast enhanced procedures. The literature regarding the safety of IV contrast in this cohort is lacking. A retrospective review was carried out in a university hospital to identify the incidence of CIN in patients with MM and to look for associated risk factors. 94 patients and 165 procedures were included in the analysis. 10% of procedures resulted in CIN. 59% (10/17) of creatinines had normalized within one month of the procedure. The only factor found to be significant for the development of CIN was the timing of the procedure (<18mths verses >18mths post diagnosis of MM; p = 0.045). CIN appears to occur at an increased rate in patients with MM. However this may be an over-estimation given the common occurrence of renal impairment in this cohort and the close temporal relationship which often exists between systemic illness and contrast-enhanced procedures. C1 [Crowley, P Maeve] Cork University Hospital, Department of Haematology, WiltonCork, Ireland. [Prabhakaran, N Vinitha] University College Cork, Department of MedicineCork, Ireland. [Gilligan, M Oonagh] Cork University Hospital, Department of Haematology, WiltonCork, Ireland. RP Crowley, PM (reprint author), Cork University Hospital, Department of Haematology, Cork, Ireland. EM maeve.crowley@gmail.com CR Kyle RA, Rajkumar SV, 2009, Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma. Leukemia 23(1):3–9 Society American Cancer, 2013, How many people get multiple myeloma? [updated 2/13/2013; cited 2013 12/3/2013]; Available from: http://www.cancer.org/cancer/multiplemyeloma/ overviewguide/multiple-myeloma-overview-key-statistics Ireland National Cancer Rregistry, 2013, Cancer factsheet:multiple myeloma. [updated 16/12/2013; cited 2014 13/1/2014]; Available from: http://www.ncri.ie/ Blade J, Fernandez-Llama P, Bosch F et al, 1998, Renal failure in multiplemyeloma: presenting features and predictors of outcome in 94 patients from a single institution. Arch Intern Med 158(17): 1889–1893 Heher EC, RennkeHG, Laubach JP et al, 2013, Kidney disease and multiple myeloma. Cli J Am Soc Nephrol: CJASN 8(11):2007– 2017 Berns AS, 1989, Nephrotoxicity of contrast media. Kidney Int 36(4):730–740 Stacul F, van der Molen AJ, Reimer P et al, 2011, Contrast induced nephropathy: updated ESUR Contrast Media Safety Committee guidelines. Eur Radiol 21(12):2527–2541 Geenen RW, Kingma HJ, van der Molen AJ, 2013, Contrastinduced nephropathy: pharmacology, pathophysiology and prevention. Insights Imaging 4(6):811–820 Cicin I, Erdogan B, Gulsen E et al., 2014, Incidence of contrast induced nephropathy in hospitalised patients with cancer. Eur Radiol 24(1):184–190 McDonald JS, McDonald RJ, Comin J et al, 2013, Frequency of acute kidney injury following intravenous contrast medium administration: a systematic review and meta-analysis. Radiology 267(1): 119–128 Holman RL, 1939, Complete anuria due to blockage of renal tubules by protein casts in a case of multiple myeloma. Arch Path 27: 748 Bartels ED, Brun GC, Gammeltoft A et al, 1954, Acute anuria following intravenous pyelography in a patient with myelomatosis. Acta Med Scand 150(4):297–302 Brodwall EK, Knutsen SB, Myhre JR, 1956, Acute renal failure following intravenous pyelography in cases of myelomatosis. Acta Med Scand 156(4):263–266 Killmann SA, Gjorup S, Thaysen JH, 1957, Fatal Acute Renal Failure Following Intravenous Pyelography in a Patient with Multiple Myeloma. Acta Med Scand 158(1):43–46 Perillie PE, Conn HO, 1958, Acute Renal Failure after Intravenous Pyelography in Plasma Cell Myeloma. Jama-J Am Med Assoc 167(18):2186–2189 Macalister CL, Addison NV, 1961, Renal aspects ofmyelomatosis. Br J Urol 33:141–148 Healy JK, 1963, Acute Oliguric Renal Failure Associated with Multiple Myeloma - Report of 3 Cases. Brit Med J 1(5338): 1126–1130 Gross M, Mcdonald H, Waterhou K, 1968, Anuria Following Urography with Meglumine Diatrizoate, Renografin, in Multiple Myeloma. Radiology 90(4):780–781 Shulman G, 1977, Bence-Jones Myelomatosis and Intravenous Pyelography. S Afr Med J 51(17):574–576 Addis T, 1948, Glomerular Nephritis: Diagnosis and Treatment. MacMillan, New York Brown M, Battle JD, 1964, Effect of Urography on Renal Function in Patients with Multiple Myeloma. Can Med Assoc J 91:786–790 Vix VA, 1966, Intravenous Pyelography in Multiple Myeloma - a Review of 52 Studies in 40 Patients. Radiology 87(5):896–902 Morgan C, Hammack WJ, 1966, Intravenous Urography in Multiple Myeloma. New Engl J Med 275(2):77–79 Lasser EC, Lang JH, Zawadzhi ZA, 1966, Contrast Media - Myeloma Protein Precipitates in Urography. JAMA 198(8):945– 947 Myers GH,Witten DM, 1971, Acute Renal Failure after Excretory Urography in Multiple Myeloma. Amer J Roentgenol Ra 113(3): 583–588 Defronzo RA, Humphrey RL,Wright JR et al, 1975, Acute Renal- Failure in Multiple-Myeloma. Medicine 54(3):209–223 Carvallo A, Rakowski TA, Argy WP et al, 1978, Acute Renal- Failure Following Drip Infusion Pyelography. Am J Med 65(1): 38–45 Cohen DJ, Sherman WH, Osserman EF et al, 1984, Acute-Renal- Failure in Patients withMultiple-Myeloma. Am J Med. 76(2):247– 256 Pahade JK, LeBedis CA, Raptopoulos VD et al, 2011, Incidence of contrast-induced nephropathy in patients with multiple myeloma undergoing contrast-enhanced CT. AJR Am J Roentgenol 196(5): 1094–1101 Mccarthy CS, Becker JA, 1992, Multiple-Myeloma and Contrast- Media. Radiology 183(2):519–521 Preda L, Agazzi A, Raimondi S et al, 2011, Effect on renal function of an iso-osmolar contrast agent in patients with monoclonal gammopathies. Eur Radiol 21(1):63–69 ACR_Committee_on_Drugs_and_Contrast_Media, 2013, ACR manual on contrast media. 9th:[33–42]. Available from: http:// www.acr.org/~/media/ACR/Documents/PDF/QualitySafety/ Resources/Contrast%20Manual/2013_Contrast_Media.pdf Lewington A, MacTier R, Hoefield R, Sutton A et al, 2013, Prevention of Contrast Induced Acute Kidney Injury, CI-AKI, in adult patients. Available from: http://www.rcr.ac.uk/docs/ radiology/pdf/2013_RA_BCIS_RCR.pdf NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 915 EP 919 DI 10.1007/s12253-017-0300-9 PG 5 ER PT J AU Lu, ChTh Moretti, K Beckmann, K Cohen, P O’Callaghan, M AF Lu, Chengxuan Thomas Moretti, Kim Beckmann, Kerri Cohen, Penelope O’Callaghan, Michael TI ISUP Group 4 – a Homogenous Group of Prostate Cancers? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Gleason score; Grade group 4; Biopsy; Prostate cancer specific mortality ID Prostate cancer; Gleason score; Grade group 4; Biopsy; Prostate cancer specific mortality AB The International Society of Urological Pathology (ISUP) and the World Health Organisation have adopted a five-tiered prognostic grade group for prostate cancer in 2014. Grade group 4 is comprised of Gleason patterns 4 + 4, 3 + 5 and 5 + 3. Recent articles have suggested heterogeneity in their prognostic outcomes.We aimed to determine whether there was a difference in mortality outcomes within the ISUP 4 grouping, as identified on needle biopsy. A total of 4080 men who were diagnosed with non-metastatic (N0 M0) prostate cancer on biopsy with Gleason scores of 7, 8 and 9 were included. Multi-variable Cox Regression and Fine and Grey competing risk analysis were used to determine the All-Cause Mortality (ACM) and the Prostate Cancer Specific Mortality (PCSM) as a function of Gleason Scores (Gleason 3 + 4, 4 + 3, 4 + 4, 3 + 5/5 + 3, 9). Gleason score 4 + 4 was utilized as the referent. The 60 months’ prostate cancer specific mortality with Gleason patterns 4 + 4 and 3 + 5/5 + 3 were 17% and 20%respectively (P < 0.01). Patients with 3 + 5/5 + 3 disease, had no statistically significant difference in the ACM (adjusted hazard ratio [aHR] 0.99, 95% confidence interval [Cl] 0.68–1.4, p = 0.99) and PCSM risk (aHR 0.77, 95% Cl 0.47–1.2, p = 0.31) when compare with the referent group of patients. Patients with Gleason patterns 4 + 3 and 9 had statistically significant difference in their PCSM risk (aHR 0.70, 95% CI 0.54–0.91, P < 0.001 and aHR 1.5, 95% Cl 1.2–1.9, P < 0.001) when compared to the referent group. Our analysis suggest that ISUP group 4 is homogenous in terms of the allcause mortality and the prostate cancer specific morality risk as differentiated by the presence of Gleason 5 score. C1 [Lu, Chengxuan Thomas] University of AdelaideAdelaide, Australia. [Moretti, Kim] Queen Elisabeth Hospital, Repatriation HospitalWoodville, Australia. [Beckmann, Kerri] South Australia Prostate Cancer Clinical Outcomes CollaborationAdelaide, Australia. [Cohen, Penelope] Royal Adelaide Hospital, SA PathologyAdelaide, Australia. [O’Callaghan, Michael] South Australia Prostate Cancer Clinical Outcomes CollaborationAdelaide, Australia. RP Lu, ChTh (reprint author), University of Adelaide, Adelaide, Australia. EM thomascxlu@gmail.com CR Epstein JI, Egevad L, Amin MB, Delahunt B, Srigley JR, Humphrey PA, 2016, The 2014 International Society of Urological Pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma: definition of grading patterns and proposal for a new grading system. Am J Surg Pathol 40(2):244– 252. , DOI 10.1097/pas.0000000000000530 Huynh MA, Chen MH, Wu J, Braccioforte MH, Moran BJ, D'Amico AV, 2015, Gleason score 3 + 5 or 5 + 3 versus 4 + 4 prostate cancer: the risk of death. Eur Urol. , DOI 10.1016/ j.eururo.2015.08.054 Rusthoven CG, Waxweiler TV, DeWitt PE, Flaig TW, Raben D, Kavanagh BD, 2015, Gleason stratifications prognostic for survival in men receiving definitive external beam radiation therapy for localized prostate cancer. Urol Oncol 33(2):71.e11–71.e79. , DOI 10.1016/j.urolonc.2014.07.010 Harding-Jackson N, Kryvenko ON, Whittington EE, Eastwood DC, Tjionas GA, Jorda M, Iczkowski KA, 2016, Outcome of Gleason 3 + 5 = 8 prostate cancer diagnosed on needle biopsy: prognostic comparison with Gleason 4 + 4 = 8. J Urol 196(4): 1076–1081. , DOI 10.1016/j.juro.2016.05.105 van den Bergh RC, van der Kwast TH, de Jong J, Zargar H, Ryan AJ, Costello AJ, Murphy DG, van der Poel HG, 2016, Validation of the novel International Society of Urological Pathology 2014 fivetier Gleason grade grouping: biochemical recurrence rates for 3+5 disease may be overestimated. BJU Int 118(4):502–505. https://doi. org/10.1111/bju.13478 Mahal BA, Muralidhar V, Chen YW, Choueiri TK, Hoffman KE, JC H, Sweeney CJ, JB Y, Feng FY, Trinh QD, Nguyen PL, 2015, Gleason score 5 + 3 = 8 prostate cancer: much more like Gleason score 9? BJU Int. , DOI 10.1111/bju.13239 JacksonW, Hamstra DA, Johnson S, Zhou J, Foster B, Foster C, Li D, Song Y, Palapattu GS, Kunju LP, Mehra R, Feng FY, 2013, Gleason pattern 5 is the strongest pathologic predictor of recurrence, metastasis, and prostate cancer-specific death in patients receiving salvage radiation therapy following radical prostatectomy. Cancer 119(18):3287–3294. , DOI 10.1002/cncr.28215 Sabolch A, Feng FY, Daignault-Newton S, Halverson S, Blas K, Phelps L, Olson KB, Sandler HM, Hamstra DA, 2011, Gleason pattern 5 is the greatest risk factor for clinical failure and death from prostate cancer after dose-escalated radiation therapy and hormonal ablation. Int J Radiat Oncol Biol Phys 81(4):e351–e360. https://doi. org/10.1016/j.ijrobp.2011.01.063 Esserman LJ, Thompson IM, Reid B, Nelson P, Ransohoff DF, Welch HG, Hwang S, Berry DA, Kinzler KW, Black WC, Bissell M, Parnes H, Srivastava S, 2014, Addressing overdiagnosis and overtreatment in cancer: a prescription for change. The Lancet Oncology 15(6):e234–e242. , DOI 10.1016/s1470- 2045(13)70598-9 Epstein JI, Zelefsky MJ, Sjoberg DD, Nelson JB, Egevad L, Magi- Galluzzi C, Vickers AJ, Parwani AV, Reuter VE, Fine SW, Eastham JA,Wiklund P, Han M, Reddy CA, Ciezki JP, Nyberg T, Klein EA, 2016, A contemporary prostate cancer grading system: a validated alternative to the Gleason score. Eur Urol 69(3):428–435. https:// doi.org/10.1016/j.eururo.2015.06.046 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 921 EP 925 DI 10.1007/s12253-017-0331-2 PG 5 ER PT J AU Kiss, N Krolopp, Lorincz, K Banvolgyi, A Szipocs, R Wikonkal, N AF Kiss, Norbert Krolopp, Adam Lorincz, Kende Banvolgyi, Andras Szipocs, Robert Wikonkal, Norbert TI Stain-free Histopathology of Basal Cell Carcinoma by Dual Vibration Resonance Frequency CARS Microscopy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Basal cell carcinoma; Nonlinear microscopy; Coherent anti-stokes Raman scattering; Pseudo HE images; Cancer detection ID Basal cell carcinoma; Nonlinear microscopy; Coherent anti-stokes Raman scattering; Pseudo HE images; Cancer detection AB Basal cell carcinoma (BCC) is the most common malignancy in Caucasians. Nonlinear microscopy has been previously utilized for the imaging of BCC, but the captured images do not correlate with H&E staining. Recently, Freudiger et al. introduced a novel method to visualize tissue morphology analogous to H&E staining, using coherent anti-Stokes Raman scattering (CARS) technique. In our present work, we introduce a novel algorithm to post-process images obtained from dual vibration resonance frequency (DVRF) CARS measurements to acquire high-quality pseudo H&E images of BCC samples. We adapted our CARS setup to utilize the distinct vibrational properties of CH3 (mainly in proteins) and CH2 bonds (primarily in lipids). In a narrowband setup, the central wavelength of the pump laser is set to 791 nm and 796 nm to obtain optimal excitation. Due to the partial overlap of the excitation spectra and the 5–10 nm FWHM spectral bandwidth of our lasers, we set the wavelengths to 790 nm (proteins) and 800 nm (lipids). Nonresonant background from water molecules also reduces the chemical selectivity which can be significantly improved if we subtract the DVRF images from each other. As a result, we acquired two images: one for "lipids" and one for "proteins" when we properly set a multiplication factor to minimize the non-specific background. By merging these images, we obtained high contrast H&E "stained" images of BBC’s. Nonlinear microscope systems upgraded for real time DVRF CARS measurements, providing pseudo H&E images can be suitable for in vivo assessment of BCC in the future. C1 [Kiss, Norbert] Wigner RCP, Institute for Solid State Physics and Optics, H-1525 Budapest, Hungary. [Krolopp, Adam] R&D Ultrafast Lasers Ltd, H-1539 Budapest, Hungary. [Lorincz, Kende] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Banvolgyi, Andras] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Szipocs, Robert] Wigner RCP, Institute for Solid State Physics and Optics, H-1525 Budapest, Hungary. [Wikonkal, Norbert] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. RP Szipocs, R (reprint author), Wigner RCP, Institute for Solid State Physics and Optics, H-1525 Budapest, Hungary. EM r.szipocs@szipocs.com CR Marzuka AG, Book SE, 2015, Basal cell carcinoma: pathogenesis, epidemiology, clinical features, diagnosis, histopathology, and management. Yale J Biol Med 88(2):167–179 Clark CM, Furniss M, Mackay-Wiggan JM, 2014, Basal cell carcinoma: an evidence-based treatment update. Am J Clin Dermatol 15(3):197–216. , DOI 10.1007/s40257-014-0070-z Seidenari S, Arginelli F, Bassoli S, Cautela J, Cesinaro AM, Guanti M, Guardoli D, Magnoni C, Manfredini M, Ponti G, Konig K, 2013, Diagnosis of BCC by multiphoton laser tomography. Skin Res Technol 19(1):e297–e304. , DOI 10.1111/j.1600-0846.2012.00643.x Vogler N, Meyer T, Akimov D, Latka I, Krafft C, Bendsoe N, Svanberg K, Dietzek B, Popp J, 2010)Multimodal imaging to study the morphochemistry of basal cell carcinoma. J Biophotonics 3(10– 11):728–736. , DOI 10.1002/jbio.201000071 Heuke S, Vogler N,Meyer T, Akimov D, Kluschke F, Rowert-Huber HJ, Lademann J, Dietzek B, Popp J, 2013, Detection and discrimination of non-melanoma skin cancer by multimodal imaging. Healthcare, Basel, Switzerland, 1(1):64–83. , DOI 10. 3390/healthcare1010064 Freudiger CW, Pfannl R, Orringer DA, Saar BG, Ji M, Zeng Q, Ottoboni L, Wei Y, Waeber C, Sims JR, De Jager PL, Sagher O, Philbert MA, Xu X, Kesari S, Xie XS, Young GS, 2012, Multicolored stain-free histopathology with coherent Raman imaging. Lab Investig 92(10):1492–1502. , DOI 10.1038/ labinvest.2012.109 Haluszka D, Lorincz K, Kiss N, Szipocs R, Kuroli E, Gyongyosi N, Wikonkal NM, 2016, Diet-induced obesity skin changes monitored by in vivo SHG and ex vivo CARS microscopy. Biomed Opt Express 7(11):4480–4489. , DOI 10.1364/boe.7.004480 Duarte AS, Schnedermann C, Kukura P, 2016, Wide-field detected fourier transform CARS microscopy. Sci Rep 6:37516. https://doi. org/10.1038/srep37516 Krolopp A, Csakanyi A, Haluszka D, Csati D, Vass L, Kolonics A, Wikonkal N, Szipocs R, 2016, Handheld nonlinear microscope system comprising a 2 MHz repetition rate, mode-locked Yb-fiber laser for in vivo biomedical imaging. Biomed Opt Express 7(9):3531– 3542. , DOI 10.1364/boe.7.003531 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 927 EP 930 DI 10.1007/s12253-017-0356-6 PG 4 ER PT J AU Laus, CA da Silva, DCGI Cordeiro, BF Lo Turco, GE de Souza Viana, L Carvalho, LA AF Laus, Carolina Ana da Silva, Dale Cotrim Guerreiro Ismael Cordeiro, Bertuccez Fernanda Lo Turco, Guimaraes Edson de Souza Viana, Luciano Carvalho, Lopes Andre TI Is Lipidomic the Answer to the Search of a Biomarker for Organ Preservation Protocol in Head and Neck Squamous Cell Carcinoma? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Head and neck cancer; Induction chemotherapy; Lipidomics; Mass spectrometry; Biomarker; Response to treatment ID Head and neck cancer; Induction chemotherapy; Lipidomics; Mass spectrometry; Biomarker; Response to treatment AB In the last decade organ preservation protocols based on chemoradiotherapy (CRT) has been showing the possibility of preserving function without jeopardizing survival for locally advanced head and neck squamous cell carcinoma (HNSCC). Still, only a percentage of the patients will benefit from this approach and, to date, no biomarkers are known to correctly predict these patients. More recently, modern mass spectrometry method has been used to determine metabolic profiles, and lipidomics, in particular, emerged as a new field of study in oncology and other diseases. This study aimed to analyze the lipid profile on saliva from patients undergoing to a prospective, single center, open-label, nonrandomized phase II trial for organ preservation on HNSCC. The lipid analysis was performed using matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF-MS). Multivariate statistical analyses based on principal component analysis and orthogonal partial least square-discriminant analysis were applied to MALDI-TOFMS data to visualize differences between the lipid profiles and identify potential biomarkers. The results assisted on distinguishing complete responders from non-responders to the treatment protocol. In conclusion, we demonstrated that a group of lipids is differentially abundant in saliva from HNSCC patients submitted to an organ preservation protocol, being able to differentiate responders from non-responders. These results suggest the potential use of lipid biomarkers to identify patients who may benefit from this treatment. Also, we showed that saliva testing might be routinely used in clinical practice, for being a non-invasive alternative to blood testing, besides inexpensive and easy to obtain. C1 [Laus, Carolina Ana] Barretos Cancer Hospital, Molecular Oncology Research Center, 1331, Antenor Duarte Villela St,, 14784-400 Barretos, SP, Brazil. [da Silva, Dale Cotrim Guerreiro Ismael] Universidade Federal de Sao Paulo, Urology DisciplineSao Paulo, SP, Brazil. [Cordeiro, Bertuccez Fernanda] Universidade Federal de Sao Paulo, Urology DisciplineSao Paulo, SP, Brazil. [Lo Turco, Guimaraes Edson] Universidade Federal de Sao Paulo, Urology DisciplineSao Paulo, SP, Brazil. [de Souza Viana, Luciano] Barretos Cancer Hospital, Pio XII, Clinical Oncology DepartmentBarretos, SP, Brazil. [Carvalho, Lopes Andre] Barretos Cancer Hospital, Molecular Oncology Research Center, 1331, Antenor Duarte Villela St,, 14784-400 Barretos, SP, Brazil. RP Laus, CA (reprint author), Barretos Cancer Hospital, Molecular Oncology Research Center, 14784-400 Barretos, Brazil. EM anacarolinalaus@gmail.com CR Argiris A, Karamouzis MV, Raben D, Ferris RL, 2008, Head and neck cancer. Lancet 371(9625):1695–1709 Bligh EG, Dyer WJ, 1959, A rapid method of total lipid extraction and purification. Can J Biochem Physiol 37(8):911–917 Denaro N, Russi EG, Adamo V, Merlano MC, 2014, State-of-theart and emerging treatment options in the management of head and neck cancer: news from 2013. Oncology 86(4):212–229 Haddad RI, Shin DM, 2008, Recent advances in head and neck cancer. N Engl J Med 359(11):1143–1154 Hannun YA, Obeid LM, 2008, Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol 9(2):139– 150 Hilvo M, Gade S, Hyotylainen T, Nekljudova V, Seppanen-Laakso T, Sysi-Aho M et al, 2014, Monounsaturated fatty acids in serum triacylglycerols are associated with response to neoadjuvant chemotherapy in breast cancer patients. Int J Cancer 134(7):1725–1733 Holsinger FC, Lin HY, Bassot V, Laccourreye O, 2009, Platinbased exclusive chemotherapy for selected patients with squamous cell carcinoma of the larynx and pharynx. Cancer 115(17):3909– 3918 Lefebvre JL, Chevalier D, Luboinski B, Kirkpatrick A, Collette L, Sahmoud T, 1996, Larynx preservation in pyriform sinus cancer: preliminary results of a European Organization for Research and Treatment of Cancer phase III trial. EORTC Head and Neck Cancer Cooperative Group. J Natl Cancer Inst 88(13):890–899 Li X, Yuan YJ, 2011, Lipidomic analysis of apoptotic hela cells induced by Paclitaxel. Omics: J Integr Biol 15(10):655–664 Li G, DaM, ZhangW,Wu H, Ye J, Chen J et al, 2016, Alteration of serum lipid profile and its prognostic value in head and neck squamous cell carcinoma. Journal of oral pathology& medicine: official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology 45(3):167–172 Marur S, D'Souza G, Westra WH, Forastiere AA, 2010, HPVassociated head and neck cancer: a virus-related cancer epidemic. Lancet Oncol 11(8):781–789 Pignon JP, le Maitre A,Maillard E, Bourhis J, GroupM-NC, 2009, Meta-analysis of chemotherapy in head and neck cancer, MACHNC): an update on 93 randomised trials and 17,346 patients. Radiotherapy and oncology: journal of the European Society for Therapeutic Radiology and Oncology 92(1):4–14 Saddoughi SA, Garrett-Mayer E, Chaudhary U, O'Brien PE, Afrin LB, Day TA et al, 2011, Results of a phase II trial of gemcitabine plus doxorubicin in patients with recurrent head and neck cancers: serum C(1)(8)-ceramide as a novel biomarker for monitoring response. Clinical cancer research: an official journal of the American Association for Cancer Research 17(18):6097–6105 de Souza Viana L, de Aguiar Silva FC, Andrade Dos Anjos Jacome A, Calheiros CampeloMaia D, Duarte deMattos M, Arthur Jacinto A et al, 2016, Efficacy and safety of a cisplatin and paclitaxel induction regimen followed by chemoradiotherapy for patients with locally advanced head and neck squamous cell carcinoma. Head Neck 38(1):E970–E980 Stegemann C, Drozdov I, Shalhoub J, Humphries J, Ladroue C, Didangelos A et al, 2011, Comparative lipidomics profiling of human atherosclerotic plaques. Circ Cardiovasc Genet 4(3):232–242 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108 Wang Q, Gao P, Wang X, Duan Y, 2014, The early diagnosis and monitoring of squamous cell carcinoma via saliva metabolomics. Sci Rep 4:6802 Wood PL, 2012, Lipidomics of Alzheimer's disease: current status. Alzheimers Res Ther 4(1):5 Zhao C, Mao J, Ai J, Shenwu M, Shi T, Zhang D et al, 2013, Integrated lipidomics and transcriptomic analysis of peripheral blood reveals significantly enriched pathways in type 2 diabetes mellitus. BMC Med Genet 6(1):S12 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 931 EP 935 DI 10.1007/s12253-017-0336-x PG 5 ER PT J AU Kostovska, MI Jakimovska, M Popovska-Jankovic, K Kubelka-Sabit, K Karagjozov, M Plaseska-Karanfilska, D AF Kostovska, Maleva Ivana Jakimovska, Milena Popovska-Jankovic, Katerina Kubelka-Sabit, Katerina Karagjozov, Mitko Plaseska-Karanfilska, Dijana TI TIMP3 Promoter Methylation Represents an Epigenetic Marker of BRCA1ness Breast Cancer Tumours SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TIMP3; BRCA 1ness; MS-MLPA; Breast cancer ID TIMP3; BRCA 1ness; MS-MLPA; Breast cancer AB Tumours presenting BRCAness profile behave more aggressively and are more invasive as a consequence of their complex genetic and epigenetic alterations, caused by impaired fidelity of the DNA repair processes. Methylation of promoter CpG islands represents an alternative mechanism to inactivate DNA repair and tumour suppressor genes. In our study, we analyzed the frequency of methylation changes of 24 tumour suppressor genes and explored their association with BRCAness profile. BRCA1ness profile and aberrant methylation were studied in 233 fresh frozen breast tumour tissues by Multiplex Ligation-dependent Probe Amplification (MLPA) and Methylation Specific (MS)-MLPA methods, respectively. Our analyses revealed that 12.4% of the breast cancer (BC) patients had tumours with a BRCA1ness profile. TIMP3 showed significantly higher (p = 5.8х10−5) methylation frequency in tumours with BRCA1ness, while methylation of APC, GSTP1 and RASSF1 promoters was negatively associated with BRCA1ness (р = 0.0017, р = 0.007 and р = 0.046, respectively). TIMP3 methylation was also associated with triple negative (TN) BC. Furthermore, TN tumours showing BRCA1ness showed stronger association with TIMP3 methylation (p = 0.0008) in comparison to TN tumours without BRCA1ness (p = 0.009). In conclusion, we confirmed that TIMP3 methylation is a marker for TN tumours and furthermore we showed for the first time that TIMP3 promoter methylation is an epigenetic marker of BRCA1ness tumours. C1 [Kostovska, Maleva Ivana] Macedonian Academy of Sciences and Arts, Research Centre for Genetic Engineering and Biotechnology, Krste Misirkov 2Skopje, Macedonia. [Jakimovska, Milena] Macedonian Academy of Sciences and Arts, Research Centre for Genetic Engineering and Biotechnology, Krste Misirkov 2Skopje, Macedonia. [Popovska-Jankovic, Katerina] Macedonian Academy of Sciences and Arts, Research Centre for Genetic Engineering and Biotechnology, Krste Misirkov 2Skopje, Macedonia. [Kubelka-Sabit, Katerina] Clinical Hospital Acibadem SistinaSkopje, Macedonia. [Karagjozov, Mitko] Clinical Hospital Acibadem SistinaSkopje, Macedonia. [Plaseska-Karanfilska, Dijana] Macedonian Academy of Sciences and Arts, Research Centre for Genetic Engineering and Biotechnology, Krste Misirkov 2Skopje, Macedonia. RP Plaseska-Karanfilska, D (reprint author), Macedonian Academy of Sciences and Arts, Research Centre for Genetic Engineering and Biotechnology, Skopje, Macedonia. EM dijana@manu.edu.mk CR Lord CJ, Ashworth A, 2016, BRCAness revisited. Nat Rev Cancer 16(2):110–120 Turner N, Tutt A, Ashworth A, 2004, Hallmarks of 'BRCAness' in sporadic cancers. Nat Rev Cancer 4(10):814–819 Jones PA, Baylin SB, 2007, The epigenomics of cancer. Cell 128(4):683–692 Lips EH, Laddach N, Savola SP, Vollebergh MA, Oonk AM, Imholz AL, Wessels LF, Wesseling J, Nederlof PM, Rodenhuis S, 2011, Quantitative copy number analysis by multiplex ligationdependent probe amplification, MLPA, of BRCA1-associated breast cancer regions identifies BRCAness. Breast Cancer Res 13(5):R107 Coffa J, van de Wiel MA, Diosdado B, Carvalho B, Schouten J, Meijer GA, 2008, MLPAnalyzer: data analysis tool for reliable automated normalization of MLPA fragment data. Cell Oncol 30(4):323–335 Nygren AO, Ameziane N, Duarte HM, Vijzelaar RN, Waisfisz Q, Hess CJ, Schouten JP, Errami A, 2005, Methylation-specific MLPA, MS-MLPA): simultaneous detection of CpG methylation and copy number changes of up to 40 sequences. Nucleic Acids Res 33(14):e128 Cruz-Munoz W, Khokha R, 2008, The role of tissue inhibitors of metalloproteinases in tumorigenesis and metastasis. Crit Rev Clin Lab Sci 45(3):291–338 Lui EL, Loo WT, Zhu L, Cheung MN, Chow LW, 2005, DNA hypermethylation of TIMP3 gene in invasive breast ductal carcinoma. Biomed Pharmacother 59(2):S363–S365 Bachman KE, Herman JG, Corn PG, Merlo A, Costello JF, Cavenee WK, Baylin SB, Graff JR, 1999, Methylation-associated silencing of the tissue inhibitor of metalloproteinase-3 gene suggest a suppressor role in kidney, brain, and other human cancers. Cancer Res 59(4):798–802 Mylona E, Magkou C, Giannopoulou I, Agrogiannis G, Markaki S, Keramopoulos A, Nakopoulou L, 2006, Expression of tissue inhibitor of matrix metalloproteinases, TIMP)-3 protein in invasive breast carcinoma: relation to tumor phenotype and clinical outcome. Breast Cancer Res 8(5):R57 Widschwendter M, Siegmund KD, Muller HM, Fiegl H, Marth C, Muller-Holzner E, Jones PA, Laird PW(2004)Association of breast cancer DNA methylation profiles with hormone receptor status and response to tamoxifen. Cancer Res 64(11):3807–3813 Murria R, Palanca S, de Juan I, Alenda C, Egoavil C, Segui FJ, Garcia-Casado Z, Juan MJ, Sanchez AB, Segura A, Santaballa A, Chirivella I, Llop M, Perez G, Barragan E, Salas D, Bolufer P, 2015, Immunohistochemical, genetic and epigenetic profiles of hereditary and triple negative breast cancers. Relevance in personalized medicine. Am J Cancer Res 5(7):2330–2343 Holm K, Hegardt C, Staaf J, Vallon-Christersson J, Jonsson G, Olsson H, Borg A, RingnerM(2010)Molecular subtypes of breast cancer are associated with characteristic DNAmethylation patterns. Breast Cancer Res 12(3):18 Sunami E, Shinozaki M, Sim MS, Nguyen SL, Vu AT, Giuliano AE, Hoon DS, 2008, Estrogen receptor and HER2/neu status affect epigenetic differences of tumor-related genes in primary breast tumors. Breast Cancer Res 10(3):16 Murria Estal R, Palanca Suela S, de Juan Jimenez I, Alenda Gonzalez C, Egoavil Rojas C, Garcia-Casado Z, Lopez Guerrero JA, Juan Fita MJ, Sanchez Heras AB, Segura Huerta A, Santaballa Bertran A, Chirivella Gonzalez I, Llop Garcia M, Perez Simo G, Barragan Gonzalez E, Bolufer Gilabert P, 2016, Relationship of immunohistochemistry, copy number aberrations and epigenetic disorders with BRCAness pattern in hereditary and sporadic breast cancer. Familial Cancer 15(2):193–200 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 937 EP 940 DI 10.1007/s12253-018-0398-4 PG 4 ER PT J AU Ponti, G Maccaferri, M Mandrioli, M Manfredini, M Micali, S Cotugno, M Bianchi, G Ozben, T Pellacani, G Del Prete, Ch Tomasi, A AF Ponti, Giovanni Maccaferri, Monia Mandrioli, Mauro Manfredini, Marco Micali, Salvatore Cotugno, Michele Bianchi, Giampaolo Ozben, Tomris Pellacani, Giovanni Del Prete, Chiara Tomasi, Aldo TI Seminal Cell-Free DNA Assessment as a Novel Prostate Cancer Biomarker SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Seminal plasma cfDNA; Spermal cfDNA; Prostate cancer; Urological biomarkers; Fluorimetry; Prostate cancer screening; Liquid biopsy ID Seminal plasma cfDNA; Spermal cfDNA; Prostate cancer; Urological biomarkers; Fluorimetry; Prostate cancer screening; Liquid biopsy AB Cell-free DNA (cfDNA) includes circulating DNA fragments, which can be obtained from different human biological samples. cfDNA originates either from apoptotic and/or necrotic cells or is actively secreted by cancer cells. As yet, a quantification and size distribution assessment of seminal plasma cfDNA from prostate cancer patients has never been assessed. To discover a novel, sensitive, non-invasive biomarker of prostate cancer, through the fluorometric quantification and the electrophoretic analysis of seminal cfDNA in prostate cancer patients compared to healthy individuals. The concentration of seminal plasma cfDNA in prostate cancer patients was 2243.67 ± 1758 ng/μl, compared to 57.7 ± 4.8 ng/μl in healthy individuals (p < 0.05). Electrophoresis sites distribution patterns were different; ladder fragmentation was associated with prostate cancer patients and apoptotic electrophoretic fragmentation with healthy individuals. Human seminal fluid can be a valuable source of cfDNA in the setting of liquid biopsy procedures for the identification of novel oncological biomarkers. Seminal plasma cfDNA in prostate cancer patients is significantly more concentrated than that of age-matched, healthy controls. Fluorometric measurement and electrophoretic assessment allow a reliable quantification and characterization of seminal plasma cfDNA, which can be used routinely in prostate cancer screening programs. C1 [Ponti, Giovanni] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical PathologyModena, Italy. [Maccaferri, Monia] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical PathologyModena, Italy. [Mandrioli, Mauro] University of Modena and Reggio Emilia, Department of Life SciencesModena, Italy. [Manfredini, Marco] University of Modena and Reggio Emilia, Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Dermatology UnitModena, Italy. [Micali, Salvatore] University of Modena and Reggio Emilia, Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Division of UrologyModena, Italy. [Cotugno, Michele] University of Modena and Reggio Emilia, Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Division of UrologyModena, Italy. [Bianchi, Giampaolo] University of Modena and Reggio Emilia, Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Division of UrologyModena, Italy. [Ozben, Tomris] Akdeniz University, Biochemistry DepartmentAntalya, Turkey. [Pellacani, Giovanni] University of Modena and Reggio Emilia, Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Dermatology UnitModena, Italy. [Del Prete, Chiara] University of Modena and Reggio Emilia, Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Division of UrologyModena, Italy. [Tomasi, Aldo] University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical PathologyModena, Italy. RP Ponti, G (reprint author), University of Modena and Reggio Emilia, Department of Diagnostic and Clinical Medicine and Public Health, Division of Clinical Pathology, Modena, Italy. EM giovanni.ponti@unimore.it CR Stroun M, Lyautey J, Lederrey C, Olson-Sand A, Anker P, 2001, About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin Chim Acta 313:139–142 Lu JL, Liang ZY, 2016, Circulating free DNA in the era of precision oncology: pre- and post-analytical concerns. Chronic Dis Transl Med 2:223–230 Li HG, Huang SY, Zhou H, Liao AH, Xiong CL, 2009, Quick recovery and characterization of cell-free DNA in seminal plasma of normozoospermia and azoospermia: implications for noninvasive genetic utilities. Asian J Androl 11:703–709 Ponti G, Maccaferri M, Manfredini M, Kaleci S, Mandrioli M, Pellacani G, Ozben T, Depenni R, Bianchi G, Pirola GM, Tomasi A, 2018, The value of fluorimetry, Qubit, and spectrophotometry, NanoDrop, in the quantification of cell-free DNA, cfDNA, in malignant melanoma and prostate cancer patients. Clin Chim Acta 479:14–19 ZagoskinMV, Davis RE, Mukha DV, 2017, Double stranded RNA in human seminal plasma. Front Genet 8(154) Costa F, Barbisan F, Assmann CE, Araujo NKF, de Oliveira AR, Signori JP, Rogalski F, Bonadiman B, Fernandes MS, da Cruz IBM, 2017, Seminal cell-free DNA levels measured by PicoGreen fluorochrome are associated with sperm fertility criteria. Zygote 25:111–119 Duffy MJ, 2014, PSA in screening for prostate cancer: more good than harm or more harm than good? Adv Clin Chem 66:1–23 G. Ponti M. Maccaferri, M. Manfredini, M. Cotugno, G. Pellacani, A. Conti, S. Micali,M. Mandrioli, A. Tomasi,, 2018, Seminal cellfree DNA molecular profile as a novel diagnostic and prognostic prostate cancer biomarkers. Med Hyph. In press J. Sambrook E.F. Fritsch, T. Maniatis,, 1989, Molecular Cloning. Cold Spring Harbor, New York: Cold Spring Harbor Laboratory Press Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R, 2001, DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 61:1659–1665 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 941 EP 945 DI 10.1007/s12253-018-0416-6 PG 5 ER PT J AU Danakas, MA Bsirini, C Miyamoto, H AF Danakas, M Alexandra Bsirini, Caroline Miyamoto, Hiroshi TI The Impact of Routine Frozen Section Assessment During Penectomy on Surgical Margin Status and Long-Term Oncologic Outcomes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Frozen section assessment; Penile cancer; Penectomy; Prognosis; Surgical margin ID Frozen section assessment; Penile cancer; Penectomy; Prognosis; Surgical margin AB No recent studies have focused on assessing the role of intraoperative frozen section assessment (FSA) in the status of surgical margins (SMs) relating to the outcomes of penectomy cases. In this study, we investigated the utility of routine FSA of the SMs in men undergoing penectomy. A retrospective review identified consecutive patients who underwent partial (n = 26) or total (n = 12) penectomy for penile squamous cell carcinoma at our institution from 2004 to 2015. FSA of the SMs was performed in 21 (80.8%) partial and 10 (83.3%) total penectomies. FSAs were reported as positive (n = 3, 9.7%), atypical (n = 3, 9.7%), and negative (n = 25, 80.6%). All of the positive or negative FSA diagnoses were confirmed accurate on the frozen section controls, whereas the 3 cases with atypical FSA had non-malignant, atypical, and carcinoma cells, respectively, on the controls. Final SMs were positive in 6 (15.8%) penectomies, including 4 (12.9%) FSA cases versus 2 (28.6%) non-FSA cases (P = 0.569). Furthermore, initial positive (1 of 3) and atypical (3 of 3) FSA cases achieved negative conversion by excision of additional tissue sent for FSA. Kaplan-Meier analysis revealed that performing FSA or its number/diagnosis was not significantly associated with disease progression. Thus, performing FSA during penectomy does not appear to have any significant impact on final SM status nor long-term oncologic outcomes. However, as seen in at least 4 cases, select patients may benefit from the routine FSA. C1 [Danakas, M Alexandra] University of Rochester Medical Center, Department of Pathology & Laboratory Medicine, 601 Elmwood Avenue, 14642 Rochester, NY, USA. [Bsirini, Caroline] University of Rochester Medical Center, Department of Pathology & Laboratory Medicine, 601 Elmwood Avenue, 14642 Rochester, NY, USA. [Miyamoto, Hiroshi] University of Rochester Medical Center, Department of Pathology & Laboratory Medicine, 601 Elmwood Avenue, 14642 Rochester, NY, USA. RP Miyamoto, H (reprint author), University of Rochester Medical Center, Department of Pathology & Laboratory Medicine, 14642 Rochester, USA. EM hiroshi_miyamoto@urmc.rochester.edu CR Baldur-Felskov B, Hannibal CG, Munk C, Kjaer SK, 2012, Increased incidence of penile cancer and high-grade penile intraepithelial neoplasia in Denmark 1978-2008: a nationwide population-based study. Cancer Causes Control 23:273–280 Hakenberg OW, Comperat EM, Minhas S, Necchi A, Protzel C, Watkin N, European Association of Urology, 2015, EAU guidelines on penile cancer: 2014 update. Eur Urol 67:142–150 Philippou P, Shabbir M,Malone P, Nigam R,Muneer A, Ralph DJ, Minhas S, 2012, Conservative surgery for squamous cell carcinoma of the penis: resection margins and long-term oncological control. J Urol 188:803–808 Kakiuchi Y, Choy B, Gordetsky J, Izumi K, Wu G, Rashid H, Joseph JV, Miyamoto H, 2013, Role of frozen section analysis of surgical margins during robot-assisted laparoscopic radical prostatectomy: a 2,608-case experience. Hum Pathol 44:1556– 1562 Venigalla S, Wu G, Miyamoto H, 2013, The impact of routine frozen section analysis during partial nephrectomy on surgical margin status and tumor recurrence: A clinicopathologic study of 433 cases. Clin Genitourin Cancer 11:527–536 Miyamoto H, 2017, Clinical benefits of frozen section assessment during urological surgery: does it contribute to improving surgical margin status and patient outcomes as previously thought? Int J Urol 24:25–31 Shen SS, Truong LD, Ro JY, Ayala AG, 2012, Use of frozen section in genitourinary pathology. Pathology 44:427–433 Algaba F, Arce Y, Lopez-Beltran A, Montironi R, Mikuz G, Bono AV, European Society of Uropathology, Uropathology Working Group, 2005, Intraoperative frozen section diagnosis in urological oncology. Eur Urol 47:129–136 Bissada NK, Yakout HH, Fahmy WE, Gayed MS, Touijer AK, Greene GF, Hanash KA, 2003, Multi-institutional long-term experience with conservative surgery for invasive penile carcinoma. J Urol 169:500–502 Minhas S, Kayes O, Hegarty P, Kumar P, Freeman A, Ralph D, 2005, What surgical resection margins are required to achieve oncological control in men with primary penile cancer? BJU Int 96:1040–1043 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 947 EP 950 DI 10.1007/s12253-018-0439-z PG 4 ER PT J AU Zhu, L Zhang, Sh Xun, Y Jiang, Y Xia, B Chen, X Wang, L Jiang, H Shenglin, M AF Zhu, Lucheng Zhang, Shirong Xun, Yanping Jiang, Yanping Xia, Bing Chen, Xueqin Wang, Limin Jiang, Hong Shenglin, Ma TI Correction to: Comparison of the Amplification Refractory Mutation System, Super Amplification Refractory Mutation System, and Droplet Digital PCR for T790 M Mutation Detection in Non-small Cell Lung Cancer after Failure of Tyrosine Kinase Inhibitor Treatment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Correction to: Pathology & Oncology Research https://doi.org/10.1007/s12253-017-0286-3 The original version of this article unfortunately contained an error in the author group and affiliation sections. The affiliation of first author (Lucheng Zhu) should be Department of Oncology, Hangzhou Cancer Hospital and Department of Oncology, Hangzhou First People’s Hospital, Nanjing Medical University. The corrected author group is shown above and the corrected affiliations are shown below. C1 [Zhu, Lucheng] Hangzhou Cancer Hospital, Department of OncologyHangzhou, China. [Zhang, Shirong] Nanjing Medical University, Hangzhou First People’s Hospital, Hangzhou Translational Medicine Research CenterNanjing, China. [Xun, Yanping] Nanjing Medical University, Hangzhou First People’s Hospital, Hangzhou Translational Medicine Research CenterNanjing, China. [Jiang, Yanping] Nanjing Medical University, Hangzhou First People’s Hospital, Hangzhou Translational Medicine Research CenterNanjing, China. [Xia, Bing] Hangzhou Cancer Hospital, Department of OncologyHangzhou, China. [Chen, Xueqin] Nanjing Medical University, Hangzhou First People’s Hospital, Department of OncologyNanjing, China. [Wang, Limin] Nanjing Medical University, Hangzhou First People’s Hospital, Department of RespiratoryNanjing, China. [Jiang, Hong] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Thoracic SurgeryNanjing, China. [Shenglin, Ma] Nanjing Medical University, Hangzhou First People’s Hospital, Department of OncologyNanjing, China. RP Shenglin, M (reprint author), Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, Nanjing, China. EM mashenglin@medmail.com.cn NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2018 VL 24 IS 4 BP 951 EP 951 DI 10.1007/s12253-018-0421-9 PG 1 ER PT J AU Chen, W Ye, L Wen, D Chen, F AF Chen, Weiqing Ye, Lijun Wen, Dengcheng Chen, Feihua TI MiR-490-5p Inhibits Hepatocellular Carcinoma Cell Proliferation, Migration and Invasion by Directly Regulating ROBO1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Hep3B; MiR-490-5p; ROBO1 ID Hepatocellular carcinoma; Hep3B; MiR-490-5p; ROBO1 AB Studies have investigated the effect of ROBO1. All the same, the relationship between miR-490-5p and ROBO1, and the underlying mechanism are still unclear. We aimed to study the effect of microRNA-490-5p (miR-490-5p) on hepatocellular carcinoma (HCC) cell proliferation, migration and invasion by directly regulating ROBO1. The expression of miR-490-5p and ROBO1 in HCC tissues and cells were tested by RT-qPCR, and the Hep3B cells were selected for subsequent experiments. We confirmed the relationship between miR-490-5p and ROBO1 by luciferase reporter system. The effects of miR-490-5p on cell proliferation, migration and invasion of Hep3B cells were assessed by MTT assay, colony formation assay, wound healing assay and transwell assay, respectively. Flow cytometry was employed to detect the influence of miR-490-5p on cell cycle and apoptosis of Hep3B cells. The expression of miR-490-5p was down-regulated, while ROBO1 was up-regulated in HCC tissues and cells than the controls. MiR-490-5p can target ROBO1. MiR-490-5p inhibited cell proliferation, migration and invasion, but promoted cell apoptosis of Hep3B cells by inhibiting ROBO1.We confirmed that miR-490-5p could directly suppress ROBO1, which might be a potential mechanism in inhibiting HCC cell proliferation, migration and invasion. C1 [Chen, Weiqing] The People’s Hospital of Lin’an City, Department of General Surgery, No 548 Yijing Street, Jincheng town, 311300 Lin’an, Zhejiang Province, China. [Ye, Lijun] The People’s Hospital of Lin’an City, Department of Gynecology and Obstetrics, No 548 Yijing Street, Jincheng town, 311300 Lin’an, Zhejiang Province, China. [Wen, Dengcheng] The People’s Hospital of Lin’an City, Department of General Surgery, No 548 Yijing Street, Jincheng town, 311300 Lin’an, Zhejiang Province, China. [Chen, Feihua] The People’s Hospital of Lin’an City, Department of Gynecology and Obstetrics, No 548 Yijing Street, Jincheng town, 311300 Lin’an, Zhejiang Province, China. RP Chen, F (reprint author), The People’s Hospital of Lin’an City, Department of Gynecology and Obstetrics, 311300 Lin’an, China. EM jxwang3188@163.com CR Chuang KH, Whitney-Miller CL, Chu CY, Zhou Z, Dokus MK, Schmit S, Barry CT, 2015, MicroRNA-494 is a master epigenetic regulator of multiple invasion-suppressor microRNAs by targeting ten eleven translocation 1 in invasive human hepatocellular carcinoma tumors. Hepatology, Baltimore, Md, 62(2):466–480. https:// doi.org/10.1002/hep.27816 Allen MD, Luong P, Hudson C, Leyton J, Delage B, Ghazaly E, Cutts R, Yuan M, Syed N, Lo Nigro C, Lattanzio L, Chmielewska- Kassassir M, Tomlinson I, Roylance R, Whitaker HC, Warren AY, Neal D, Frezza C, Beltran L, Jones LJ, Chelala C, Wu BW, Bomalaski JS, Jackson RC, Lu YJ, Crook T, Lemoine NR, Mather S, Foster J, Sosabowski J, Avril N, Li CF, Szlosarek PW, 2014, Prognostic and therapeutic impact of argininosuccinate synthetase 1 control in bladder cancer as monitored longitudinally by PET imaging. Cancer Res 74(3):896–907. , DOI 10.1158/ 0008-5472.CAN-13-1702 Chinn GA, Hirokawa KE, Chuang TM, Urbina C, Patel F, Fong J, Funatsu N, Monuki ES, 2015, Agenesis of the corpus callosum due to defective glial wedge formation in Lhx2 mutant mice. Cereb Cortex 25(9):2707–2718. , DOI 10.1093/cercor/bhu067 Kim SH, Lee KH, Kim DY, Kwak E, Kim S, Kim KT, 2015, Rhythmic control of mRNA stability modulates circadian amplitude of mouse Period3 mRNA. J Neurochem 132(6):642–656. , DOI 10.1111/jnc.13027 Li S, Xu X, Xu X, Hu Z,Wu J, Zhu Y, Chen H, Mao Y, Lin Y, Luo J, Zheng X, Xie L, 2013, MicroRNA-490-5p inhibits proliferation of bladder cancer by targeting c-Fos. Biochem Biophys Res Commun 441(4):976–981. , DOI 10.1016/j.bbrc.2013.11.006 Chen K, Zeng J, Tang K, Xiao H, Hu J, Huang C, YaoW, Yu G, Xiao W, Guan W, Guo X, Xu H, Ye Z, 2016, miR-490-5p suppresses tumour growth in renal cell carcinoma through targeting PIK3CA. Biol Cell 108(2):41–50. , DOI 10.1111/boc.201500033 Zhang LY, LiuM, Li X, Tang H, 2013, miR-490-3p modulates cell growth and epithelial to mesenchymal transition of hepatocellular carcinoma cells by targeting endoplasmic reticulum-Golgi intermediate compartment protein 3, ERGIC3). J Biol Chem 288(6):4035– 4047. , DOI 10.1074/jbc.M112.410506 Saini V, Loers G, Kaur G, Schachner M, Jakovcevski I, 2016, Impact of neural cell adhesion molecule deletion on regeneration after mouse spinal cord injury. Eur J Neurosci 44(1):1734–1746. , DOI 10.1111/ejn.13271 Parray A, Siddique HR, Kuriger JK, Mishra SK, Rhim JS, Nelson HH, Aburatani H, Konety BR, Koochekpour S, Saleem M, 2014, ROBO1, A tumor suppressor and critical molecular barrier for localized tumor cells to acquire invasive phenotype: study in African- American and Caucasian prostate cancer models. Int J Cancer 135(11):2493–2506. , DOI 10.1002/ijc.28919 LiuX, Cai J, SunY, GongR, Sun D, ZhongX, Jiang S, He X, Bao E, Yang L, Li Y, 2015, MicroRNA-29a inhibits cell migration and invasion via targeting roundabout homolog 1 in gastric cancer cells. Mol Med Rep 12(3):3944–3950. , DOI 10.3892/mmr.2015.3817 Ito H, Funahashi S, Yamauchi N, Shibahara J, Midorikawa Y, Kawai S, Kinoshita Y, Watanabe A, Hippo Y, Ohtomo T, Iwanari H, Nakajima A, Makuuchi M, Fukayama M, Hirata Y, Hamakubo T, Kodama T, Tsuchiya M, Aburatani H, 2006, Identification of ROBO1 as a novel hepatocellular carcinoma antigen and a potential therapeutic and diagnostic target. Clin Cancer Res 12(11 Pt 1): 3257–3264. , DOI 10.1158/1078-0432.CCR-05-2787 Ao JY, Chai ZT, Zhang YY, Zhu XD, Kong LQ, Zhang N, Ye BG, Cai H, Gao DM, Sun HC, 2015, Robo1 Promotes angiogenesis in hepatocellular carcinoma through the rho family of guanosine triphosphatases' signaling pathway. Tumour Biol 36(11):8413– 8424. , DOI 10.1007/s13277-015-3601-1 Sullivan WJ, Christofk HR, 2015, The metabolic milieu of metastases. Cell 160(3):363–364. , DOI 10.1016/j.cell.2015.01.023 Viswanathan SR, Powers JT, Einhorn W, Hoshida Y, Ng TL, Toffanin S, O'Sullivan M, Lu J, Phillips LA, Lockhart VL, Shah SP, Tanwar PS, Mermel CH, Beroukhim R, Azam M, Teixeira J, Meyerson M, Hughes TP, Llovet JM, Radich J, Mullighan CG, Golub TR, Sorensen PH, Daley GQ, 2009, Lin28 Promotes transformation and is associated with advanced human malignancies. Nat Genet 41(7):843–848. , DOI 10.1038/ng.392 Loo JM, Scherl A, Nguyen A, Man FY,Weinberg E, Zeng Z, Saltz L, Paty PB, Tavazoie SF, 2015, Extracellular metabolic energetics can promote cancer progression. Cell 160(3):393–406. https://doi. org/10.1016/j.cell.2014.12.018 Hatziapostolou M, Polytarchou C, Aggelidou E, Drakaki A, Poultsides GA, Jaeger SA, Ogata H, Karin M, Struhl K, Hadzopoulou-Cladaras M, Iliopoulos D, 2011, An HNF4alpha-miRNA inflammatory feedback circuit regulates hepatocellular oncogenesis. Cell 147(6):1233–1247. https:// doi.org/10.1016/j.cell.2011.10.043 Kota J, Chivukula RR, O'Donnell KA, Wentzel EA, Montgomery CL, Hwang HW, Chang TC, Vivekanandan P, Torbenson M, Clark KR, Mendell JR, Mendell JT, 2009, Therapeutic microRNA delivery suppresses tumorigenesis in a murine liver cancer model. Cell 137(6):1005–1017. , DOI 10.1016/j.cell.2009.04.021 Ji J, Shi J, Budhu A,Yu Z, Forgues M, Roessler S,Ambs S, ChenY, Meltzer PS, Croce CM, Qin LX, Man K, Lo CM, Lee J, Ng IO, Fan J, Tang ZY, Sun HC, Wang XW, 2009, MicroRNA expression, survival, and response to interferon in liver cancer. N Engl J Med 361(15):1437–1447. , DOI 10.1056/NEJMoa0901282 Nakatani F, Ferracin M, Manara MC, Ventura S, Del Monaco V, Ferrari S, Alberghini M, Grilli A, Knuutila S, Schaefer KL, Mattia G, Negrini M, Picci P, Serra M, Scotlandi K, 2012, miR-34a predicts survival of Ewing's sarcoma patients and directly influences cell chemo-sensitivity and malignancy. J Pathol 226(5):796–805. , DOI 10.1002/path.3007 Shen J, Xiao Z, Wu WK, Wang MH, To KF, Chen Y, Yang W, Li MS, Shin VY, Tong JH, Kang W, Zhang L, Li M, Wang L, Lu L, Chan RL,Wong SH,Yu J, ChanMT, Chan FK, Sung JJ, Cheng AS, Cho CH, 2015, Epigenetic silencing of miR-490-3p reactivates the chromatin remodeler SMARCD1 to promote helicobacter pyloriinduced gastric carcinogenesis. Cancer Res 75(4):754–765. https:// doi.org/10.1158/0008-5472.CAN-14-1301 Sun KX, Chen Y, Chen S, Liu BL, Feng MX, Zong ZH, Zhao Y, 2016, The correlation between microRNA490-3p and TGFalpha in endometrial carcinoma tumorigenesis and progression. Oncotarget 7(8):9236–9249. 10.18632/Oncotarget.7061 Wojcicka A, Swierniak M, Kornasiewicz O, Gierlikowski W, Maciag M, Kolanowska M, Kotlarek M, Gornicka B, Koperski L, Niewinski G, Krawczyk M, Jazdzewski K, 2014, Next generation sequencing reveals microRNA isoforms in liver cirrhosis and hepatocellular carcinoma. Int J Biochem Cell Biol 53:208–217. , DOI 10.1016/j.biocel.2014.05.020 Han Y, Chen J, Zhao X, Liang C,WangY, Sun L, Jiang Z, Zhang Z, Yang R, Chen J, Li Z, Tang A, Li X, Ye J, Guan Z, Gui Y, Cai Z, 2011)MicroRNA expression signatures of bladder cancer revealed by deep sequencing. PLoS One 6(3):e18286. , DOI 10. 1371/journal.pone.0018286 Xu S, Gu G, Ni Q, Li N, Yu K, Li X, Liu C, 2015, The expression ofAEG-1 and cyclin D1 in human bladder urothelial carcinoma and their clinicopathological significance. Int J Clin Exp Med 8(11): 21222–21228 Niemoeller OM, Niyazi M, Corradini S, Zehentmayr F, Li M, Lauber K, Belka C, 2011, MicroRNA expression profiles in human cancer cells after ionizing radiation. Radiat Oncol 6:29. https://doi. org/10.1186/1748-717X-6-29 Pearson GR, Orr T, Rabin H, Cicmanec J, Ablashi D, Armstrong G, 1973, Antibody patterns to herpesvirus saimiri-induced antigens in owl monkeys. J Natl Cancer Inst 51(6):1939–1943 Zhang Z, Kang Y, Zhang Z, Zhang H, Duan X, Liu J, Li X, LiaoW, 2012, Expression of microRNAs during chondrogenesis of human adipose-derived stem cells. Osteoarthr Cartil 20(12):1638–1646. , DOI 10.1016/j.joca.2012.08.024 Tie J, Pan Y, Zhao L,Wu K, Liu J, Sun S, Guo X,Wang B, Gang Y, Zhang Y, Li Q, Qiao T, Zhao Q, Nie Y, Fan D, 2010, MiR-218 inhibits invasion and metastasis of gastric cancer by targeting the Robo1 receptor. PLoS Genet 6(3):e1000879. , DOI 10. 1371/journal.pgen.1000879 TangW,Tang J,He J, ZhouZ,QinY,Qin J, LiB,XuX,GengQ, Jiang W, Wu W, Wang X, Xia Y, 2015, SLIT2/ROBO1-miR-218-1-RET/ PLAG1: a new disease pathway involved in Hirschsprung's disease. J Cell Mol Med 19(6):1197–1207. , DOI 10.1111/jcmm.12454 Gara RK, Kumari S, Ganju A, YallapuMM, Jaggi M, Chauhan SC, 2015, Slit/Robo pathway: a promising therapeutic target for cancer. Drug Discov Today 20(1):156–164. , DOI 10.1016/j. drudis.2014.09.008 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 1 EP 9 DI 10.1007/s12253-017-0305-4 PG 9 ER PT J AU Delsin, EL Roberto, MG Fedatto, P Engel, EE Scrideli, AC Tone, GL Brassesco, SM AF Delsin, Elis Lara Roberto, Molinari Gabriela Fedatto, F. Paola Engel, Eduard Edgard Scrideli, Alberto Carlos Tone, Gonzaga Luiz Brassesco, Sol Maria TI Downregulated Adhesion-Associated microRNAs as Prognostic Predictors in Childhood Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microRNAs; Adhesion; Osteosarcoma; Metastasis ID microRNAs; Adhesion; Osteosarcoma; Metastasis AB miRNAs have been identified as key regulators of almost all cellular processes, therefore, their dysregulation is involved with several diseases, including cancer. miRNAs specifically related to the metastastic cascade are called metastamiRs and can be involvedwith different steps of this process, including loss of adhesion. Osteosarcoma (OS) is the most common primary malignant pediatric bone tumor that often presents metastatic disease at diagnosis; therefore, a deeper study of adhesionassociated miRNAs could shed light on its pathophysiology. Online databases were used to select four miRNAs (miR-139; miR-181b; miR-584; miR-708) predicted or validated to target proteins related to adherent junctions and focal adhesion pathways, and their expression levels and possible associations with clinical features evaluated in primary OS samples. Our results showed downregulation of miR-139-5p and miR-708-5p in OS samples compared to non-neoplastic controls. Moreover, lower expression of miR-708-5p was associated with poor overall survival and higher expression of miR-181b-5p related to worst chemotherapy response (low HUVOS level). Based on these results, we selected miR-139-5p and miR-708-5p for further functional testing. Inducing the expression of miR-139-5p diminished the clonogenic capacity of the HOS cell line, while upregulation of miR-708-5p was related to a lower cellular adhesion. In summary, this work identified new signatures of microRNA dysregulation that may serve as useful prognostic markers in this aggressive pediatric bone tumor. C1 [Delsin, Elis Lara] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of GeneticsSao Paulo, Brazil. [Roberto, Molinari Gabriela] University of Sao Paulo, Ribeirao Preto School of Medicine, Regional Blood CenterSao Paulo, Brazil. [Fedatto, F. Paola] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of PediatricsSao Paulo, Brazil. [Engel, Eduard Edgard] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of Biomechanics, Medicine and Rehabilitation of the Locomotor SystemSao Paulo, Brazil. [Scrideli, Alberto Carlos] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of PediatricsSao Paulo, Brazil. [Tone, Gonzaga Luiz] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of PediatricsSao Paulo, Brazil. [Brassesco, Sol Maria] University of Sao Paulo, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, Department of BiologySao Paulo, Brazil. RP Brassesco, SM (reprint author), University of Sao Paulo, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, Department of Biology, Sao Paulo, Brazil. EM solbrassesco@usp.br CR Gee HE, Ivan C, Calin GA, Ivan M, 2014, HypoxamiRs and cancer: from biology to targeted therapy. Antioxid Redox Signal 21(8): 1220–1238. , DOI 10.1089/ars.2013.5639 Olivieri F, Rippo MR, Procopio AD, Fazioli F, 2013, Circulating inflamma-miRs in aging and age-related diseases. Front Genet 4: 121. , DOI 10.3389/fgene.2013.00121 Schickel R, Boyerinas B, Park SM, Peter ME, 2008, MicroRNAs: key players in the immune system, differentiation, tumorigenesis and cell death. Oncogene 27(45):5959–5974. , DOI 10. 1038/onc.2008.274 Tutar L, Tutar E, Ozgur A, Tutar Y, 2015, Therapeutic Targeting of microRNAs in Cancer: Future Perspectives. Drug Dev Res 76(7): 382–388. , DOI 10.1002/ddr.21273 Hurst DR, Edmonds MD,Welch DR, 2009)Metastamir: the field of metastasis-regulatory microRNA is spreading. Cancer Res 69(19): 7495–7498. , DOI 10.1158/0008-5472.CAN-09-2111 Sengupta S, den Boon JA, Chen IH, Newton MA, Stanhope SA, Cheng YJ, Chen CJ, Hildesheim A, Sugden B, Ahlquist P, 2008, MicroRNA 29c is down-regulated in nasopharyngeal carcinomas, up-regulating mRNAs encoding extracellular matrix proteins. Proc Natl Acad Sci U S A 105(15):5874–5878. , DOI 10.1073/ pnas.0801130105 Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, Gerald WL, Massague J, 2008, Endogenous human microRNAs that suppress breast cancer metastasis. Nature 451(7175):147–152. , DOI 10.1038/nature06487 Gregory PA, Bracken CP, Bert AG, Goodall GJ, 2008, MicroRNAs as regulators of epithelial-mesenchymal transition. Cell Cycle 7(20):3112–3118. , DOI 10.4161/cc.7.20.6851 Ma L, Teruya-Feldstein J, Weinberg RA, 2007, Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature 449(7163):682–688. , DOI 10.1038/nature06174 Wang Y, Li Z, Zhao X, Zuo X, Peng Z, 2016, miR-10b promotes invasion by targeting HOXD10 in colorectal cancer. Oncol Lett 12(1):488–494. , DOI 10.3892/ol.2016.4628 Zhu S,Wu H,Wu F, NieD, Sheng S,Mo YY(2008)MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res 18(3):350–359. , DOI 10.1038/cr.2008.24 Hurst DR, Edmonds MD, Scott GK, Benz CC, Vaidya KS, Welch DR, 2009, Breast cancer metastasis suppressor 1 up-regulates miR- 146, which suppresses breast cancer metastasis. Cancer Res 69(4): 1279–1283. , DOI 10.1158/0008-5472.CAN-08-3559 Massague J, Obenauf AC, 2016, Metastatic colonization by circulating tumour cells. Nature 529(7586):298–306. , DOI 10. 1038/nature17038 Talmadge JE, Fidler IJ, 2010, AACR centennial series: the biology of cancer metastasis: historical perspective. Cancer Res 70(14): 5649–5669. , DOI 10.1158/0008-5472.CAN-10-1040 Nicoloso MS, Spizzo R, Shimizu M, Rossi S, Calin GA, 2009, MicroRNAs–the micro steering wheel of tumour metastases. Nat Rev Cancer 9(4):293–302. , DOI 10.1038/nrc2619 Le XF, Merchant O, Bast RC, Calin GA, 2010, The Roles of MicroRNAs in the Cancer Invasion-Metastasis Cascade. Cancer Microenviron 3(1):137–147. , DOI 10.1007/s12307-010- 0037-4 le Sage C, Nagel R, Egan DA, Schrier M,Mesman E,Mangiola A, Anile C, Maira G, Mercatelli N, Ciafre SA, Farace MG, Agami R, 2007, Regulation of the p27(Kip1, tumor suppressor by miR-221 and miR-222 promotes cancer cell proliferation. EMBO J 26(15): 3699–3708. , DOI 10.1038/sj.emboj.7601790 Yu F, Yao H, Zhu P, Zhang X, Pan Q, Gong C, Huang Y, Hu X, Su F, Lieberman J, Song E, 2007, let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell 131(6):1109–1123. , DOI 10.1016/j.cell.2007.10.054 Gorlick R, Khanna C, 2010, Osteosarcoma. J Bone Miner Res 25(4):683–691. , DOI 10.1002/jbmr.77 Kushlinskii NE, Fridman MV, Braga EA, 2016, Molecular Mechanisms and microRNAs in Osteosarcoma Pathogenesis. Biochemistry, Mosc, 81(4):315–328. , DOI 10.1134/ S0006297916040027 Ram Kumar RM, Boro A, Fuchs B, 2016, Involvement and Clinical Aspects of MicroRNA in Osteosarcoma. Int J Mol Sci 17(6). , DOI 10.3390/ijms17060877 Geng S, Zhang X, Chen J, Liu X, Zhang H, Xu X, Ma Y, Li B, Zhang Y, Bi Z, Yang C, 2014, The tumor suppressor role of miR- 124 in osteosarcoma. PLoS One 9(6):e91566. , DOI 10. 1371/journal.pone.0091566 Zhao H, Li M, Li L, Yang X, Lan G, Zhang Y, 2013, MiR-133b is down-regulated in human osteosarcoma and inhibits osteosarcoma cells proliferation, migration and invasion, and promotes apoptosis. PLoS One 8(12):e83571. , DOI 10.1371/journal.pone. 0083571 Huang J, Gao K, Lin J, Wang Q, 2014, MicroRNA-100 inhibits osteosarcoma cell proliferation by targeting Cyr61. Tumour Biol 35(2):1095–1100. , DOI 10.1007/s13277-013-1146-8 Wu P, Liang J, Yu F, Zhou Z, Tang J, Li K, 2016, miR-145 promotes osteosarcoma growth by reducing expression of the transcription factor friend leukemia virus integration 1. Oncotarget., DOI 10.18632/oncotarget.9948 Li X, Yang H, Tian Q, Liu Y, Weng Y, 2014, Upregulation of microRNA-17-92 cluster associates with tumor progression and prognosis in osteosarcoma. Neoplasma 61(4):453–460. https://doi. org/10.4149/neo_2014_056 Huang G, Nishimoto K, Zhou Z, Hughes D, Kleinerman ES, 2012, miR-20a encoded by the miR-17-92 cluster increases the metastatic potential of osteosarcoma cells by regulating Fas expression. Cancer Res 72(4):908–916. , DOI 10.1158/0008-5472. CAN-11-1460 Ma C, Zhan C, Yuan H, Cui Y, Zhang Z, 2016, MicroRNA-603 functions as an oncogene by suppressing BRCC2 protein translation in osteosarcoma. Oncol Rep 35(6):3257–3264. , DOI 10.3892/or.2016.4718 Xu SH, Yang YL, Han SM, Wu ZH, 2014, MicroRNA-9 expression is a prognostic biomarker in patients with osteosarcoma.World J Surg Oncol 12:195. , DOI 10.1186/1477-7819-12-195 Maire G, Martin JW, Yoshimoto M, Chilton-MacNeill S, Zielenska M, Squire JA, 2011, Analysis of miRNA-gene expression-genomic profiles reveals complex mechanisms of microRNA deregulation in osteosarcoma. Cancer Genet 204(3):138–146. , DOI 10. 1016/j.cancergen.2010.12.012 Jones KB, Salah Z, Del Mare S, Galasso M, Gaudio E, Nuovo GJ, Lovat F, LeBlanc K, Palatini J, Randall RL, Volinia S, Stein GS, Croce CM, Lian JB, Aqeilan RI, 2012, miRNA signatures associate with pathogenesis and progression of osteosarcoma. Cancer Res 72(7):1865–1877. , DOI 10.1158/0008-5472.CAN-11-2663 Namlos HM, Meza-Zepeda LA, Baroy T, Ostensen IH, Kresse SH, Kuijjer ML, Serra M, Burger H, Cleton-Jansen AM, Myklebost O, 2012, Modulation of the osteosarcoma expression phenotype by microRNAs. PLoS One 7(10):e48086. , DOI 10.1371/ journal.pone.0048086 Lulla RR, Costa FF, Bischof JM, ChouPM, de F Bonaldo M, Vanin EF, Soares MB, 2011, Identification of Differentially Expressed MicroRNAs in Osteosarcoma. Sarcoma 2011:732690. https://doi. org/10.1155/2011/732690 Wang W, Zhou X, Wei M, 2015, MicroRNA-144 suppresses osteosarcoma growth and metastasis by targeting ROCK1 and ROCK2. Oncotarget 6(12):10297–10308. 10.18632/oncotarget.3305 Shen L, Wang P, Yang J, Li X, 2014, MicroRNA-217 regulates WASF3 expression and suppresses tumor growth and metastasis in osteosarcoma. PLoS One 9(10):e109138. , DOI 10. 1371/journal.pone.0109138 Han K, Chen X, Bian N, Ma B, Yang T, Cai C, Fan Q, Zhou Y, Zhao TB, 2015, MicroRNA profiling identifies MiR-195 suppresses osteosarcoma cell metastasis by targeting CCND1. Oncotarget 6, 11):8875-8889., DOI 10.18632/oncotarget.3560 PoosK, Smida J,Nathrath M, Maugg D, Baumhoer D,Korsching E, 2013, How microRNA and transcription factor co-regulatory networks affect osteosarcoma cell proliferation. PLoS Comput Biol 9(8):e1003210. , DOI 10.1371/journal.pcbi.1003210 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), Method. Methods 25(4):402–408. , DOI 10.1006/ meth.2001.1262 Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C, 2006, Clonogenic assay of cells in vitro. Nat Protoc 1(5):2315– 2319. , DOI 10.1038/nprot.2006.339 Schneider CA, Rasband WS, Eliceiri KW, 2012, NIH Image to ImageJ: 25 years of image analysis. Nat Methods 9(7):671–675 Zhang J, Yan YG, Wang C, Zhang SJ, Yu XH, Wang WJ, 2015, MicroRNAs in osteosarcoma. Clin Chim Acta 444:9–17. https:// doi.org/10.1016/j.cca.2015.01.025 Zhang HD, Jiang LH, Sun DW, Li J, Tang JH, 2015, MiR-139-5p: promising biomarker for cancer. Tumour Biol 36(3):1355–1365. , DOI 10.1007/s13277-015-3199-3 Chen X, Shi K, Wang Y, Song M, Zhou W, Tu H, Lin Z, 2015, Clinical value of integrated-signature miRNAs in colorectal cancer: miRNA expression profiling analysis and experimental validation. Oncotarget 6, 35):37544-37556., DOI 10.18632/oncotarget.6065 Liu X, Duan B, Dong Y, He C, Zhou H, Sheng H, Gao H, Zhang X, 2014)MicroRNA-139-3p indicates a poor prognosis of colon cancer. Int J Clin Exp Pathol 7(11):8046–8052 Yonemori M, Seki N, Yoshino H, Matsushita R, Miyamoto K, Nakagawa M, Enokida H, 2016, Dual tumor-suppressors miR- 139-5p and miR-139-3p targeting matrix metalloprotease 11 in bladder cancer. Cancer Sci 107(9):1233–1242. , DOI 10. 1111/cas.13002 Ratert N, Meyer HA, Jung M, Lioudmer P, Mollenkopf HJ,Wagner I, Miller K, Kilic E, Erbersdobler A, Weikert S, Jung K, 2013, miRNA profiling identifies candidate mirnas for bladder cancer diagnosis and clinical outcome. J Mol Diagn 15(5):695–705. , DOI 10.1016/j.jmoldx.2013.05.008 Wang Z, Ding Q, Li Y, Liu Q, Wu W, Wu L, Yu H, 2016, Reanalysis of microRNA expression profiles identifies novel biomarkers for hepatocellular carcinoma prognosis. Tumour Biol. , DOI 10.1007/s13277-016-5369-3 Haakensen VD, Nygaard V, Greger L, Aure MR, Fromm B, Bukholm IR, Luders T, Chin SF, Git A, Caldas C, Kristensen VN, Brazma A, Borresen-Dale AL, Hovig E, Helland A, 2016, Subtype-specificmicro-RNAexpression signatures in breast cancer progression. Int J Cancer 139(5):1117–1128. , DOI 10. 1002/ijc.30142 Rask L, Balslev E, Sokilde R, Hogdall E, Flyger H, Eriksen J, Litman T, 2014, Differential expression of miR-139, miR-486 and miR-21 in breast cancer patients sub-classified according to lymph node status. Cell Oncol, Dordr, 37(3):215–227. https://doi. org/10.1007/s13402-014-0176-6 Krishnan K, Steptoe AL, Martin HC, Pattabiraman DR, Nones K, Waddell N,Mariasegaram M, Simpson PT, Lakhani SR, Vlassov A, Grimmond SM, Cloonan N, 2013, miR-139-5p is a regulator of metastatic pathways in breast cancer. RNA 19(12):1767–1780. , DOI 10.1261/rna.042143.113 Qiu G, Lin Y, Zhang H, Wu D, 2015, miR-139-5p inhibits epithelial-mesenchymal transition, migration and invasion of hepatocellular carcinoma cells by targeting ZEB1 and ZEB2. Biochem Biophys Res Commun 463(3):315–321. , DOI 10.1016/j. bbrc.2015.05.062 Yue S,Wang L, Zhang H, Min Y, Lou Y, Sun H, Jiang Y, ZhangW, Liang A, Guo Y, Chen P, Lv G, Zong Q, Li Y, 2015, miR-139-5p suppresses cancer cell migration and invasion through targeting ZEB1 and ZEB2 in GBM. Tumour Biol 36(9):6741–6749. https:// doi.org/10.1007/s13277-015-3372-8 LiQ, Liang X,Wang Y, MengX, XuY, Cai S,Wang Z, Liu J, Cai G, 2016, miR-139-5p Inhibits the Epithelial-Mesenchymal Transition and Enhances the Chemotherapeutic Sensitivity of Colorectal Cancer Cells by Downregulating BCL2. Sci Rep 6:27157., DOI , DOI 10.1038/srep27157 Watanabe K, AmanoY, Ishikawa R, SunoharaM, Kage H, Ichinose J, Sano A, Nakajima J, Fukayama M, Yatomi Y, Nagase T, Ohishi N, Takai D, 2015, Histone methylation-mediated silencing of miR- 139 enhances invasion of non-small-cell lung cancer. Cancer Med 4(10):1573–1582. , DOI 10.1002/cam4.505 Liu Y, Uzair-Ur-Rehman GY, Liang H, Cheng R, Yang F, Hong Y, Zhao C, Liu M, Yu M, Zhou X, Yin K, Chen J, Zhang J, Zhang CY, Zhi F, Chen X, 2016, miR-181b functions as an oncomiR in colorectal cancer by targeting PDCD4. Protein Cell 7(10):722–734. , DOI 10.1007/s13238-016-0313-2 Huang S,Wang J, Li J, Luo Q, ZhaoM, Zheng L, Dong X, Chen C, Che Y, Liu P, Qi J, Huang C, 2016, Serum microRNA expression profile as a diagnostic panel for gastric cancer. Jpn J Clin Oncol 46(9):811–818. , DOI 10.1093/jjco/hyw085 ZhengY, Lv X,WangX,Wang B, ShaoX, HuangY, Shi L, Chen Z, Huang J, Huang P, 2016, MiR-181b promotes chemoresistance in breast cancer by regulating Bim expression. Oncol Rep 35(2):683– 690. , DOI 10.3892/or.2015.4417 Sochor M, Basova P, Pesta M, Dusilkova N, Bartos J, Burda P, Pospisil V, Stopka T, 2014, Oncogenic microRNAs: miR-155, miR-19a, miR-181b, and miR-24 enable monitoring of early breast cancer in serum. BMC Cancer 14:448. , DOI 10.1186/ 1471-2407-14-448 Liu J, ShiW,Wu C, Ju J, Jiang J, 2014, miR-181b as a key regulator of the oncogenic process and its clinical implications in cancer, Review). Biomed Rep 2(1):7–11. , DOI 10.3892/br.2013.199 Cai B, An Y, Lv N, Chen J, Tu M, Sun J, Wu P, Wei J, Jiang K, Miao Y, 2013, miRNA-181b increases the sensitivity of pancreatic ductal adenocarcinoma cells to gemcitabine in vitro and in nude mice by targeting BCL-2. Oncol Rep 29(5):1769–1776. https:// doi.org/10.3892/or.2013.2297 Wang J, Sai K, Chen FR, Chen ZP, 2013, miR-181b modulates glioma cell sensitivity to temozolomide by targeting MEK1. Cancer Chemother Pharmacol 72(1):147–158. , DOI 10.1007/ s00280-013-2180-3 Wang X, Chen X, Meng Q, Jing H, Lu H, Yang Y, Cai L, Zhao Y, 2015)MiR-181b regulates cisplatin chemosensitivity and metastasis by targeting TGFβR1/Smad signaling pathway in NSCLC. Sci Rep 5:17618. , DOI 10.1038/srep17618 Shao JL, Li ZZ, Wang L, Jiao GL, Zhou ZG, Sun GD, 2016, microRNA-181b promotes migration and invasion of osteosarcoma cells by targeting N-myc downstream regulated gene 2. Nan Fang Yi Ke Da Xue Xue Bao 36(3):321–326 Nakajima G, Hayashi K, Xi Y, Kudo K, Uchida K, Takasaki K, Yamamoto M, Ju J, 2006, Non-coding MicroRNAs hsa-let-7g and hsa-miR-181b are Associated with Chemoresponse to S-1 in Colon Cancer. Cancer Genomics Proteomics 3(5):317–324 Hu L, Ai J, Long H, Liu W, Wang X, Zuo Y, Li Y, Wu Q, Deng Y, 2016, Integrative microRNA and gene profiling data analysis reveals novel biomarkers and mechanisms for lung cancer. Oncotarget 7, 8):8441-8454., DOI 10.18632/oncotarget.7264 Li X, Li D, Zhuang Y, Shi Q, Wei W, Ju X, 2013, Overexpression of miR-708 and its targets in the childhood common precursor Bcell ALL. Pediatr Blood Cancer 60(12):2060–2067. , DOI 10.1002/pbc.24583 Li G, Yang F, Xu H, Yue Z, Fang X, Liu J, 2015)MicroRNA-708 is downregulated in hepatocellular carcinoma and suppresses tumor invasion andmigration. Biomed Pharmacother 73:154–159. https:// doi.org/10.1016/j.biopha.2015.05.010 Lei SL, Zhao H, Yao HL, Chen Y, Lei ZD, Liu KJ, Yang Q, 2014, Regulatory roles of microRNA-708 and microRNA-31 in proliferation, apoptosis and invasion of colorectal cancer cells. Oncol Lett 8(4):1768–1774. , DOI 10.3892/ol.2014.2328 Lin KT, YehYM, ChuangCM, Yang SY, Chang JW, Sun SP,Wang YS, Chao KC,Wang LH, 2015, Glucocorticoids mediate induction of microRNA-708 to suppress ovarian cancer metastasis through targeting Rap1B. Nat Commun 6:5917. , DOI 10.1038/ ncomms6917 Ryu S, McDonnell K, Choi H, Gao D, HahnM, Joshi N, Park SM, Catena R, Do Y, Brazin J, Vahdat LT, Silver RB, Mittal V, 2013, Suppression of miRNA-708 by polycomb group promotes metastases by calcium-induced cell migration. Cancer Cell 23(1):63–76. , DOI 10.1016/j.ccr.2012.11.019 Saini S, Yamamura S, Majid S, Shahryari V, Hirata H, Tanaka Y, Dahiya R, 2011)MicroRNA-708 induces apoptosis and suppresses tumorigenicity in renal cancer cells. Cancer Res 71(19):6208–6219. , DOI 10.1158/0008-5472.CAN-11-0073 YanW,Li R,LiuY, YangP,WangZ, ZhangC,BaoZ, ZhangW,You Y, Jiang T, 2014, MicroRNA expression patterns in the malignant progression of gliomas and a 5-microRNA signature for prognosis. Oncotarget 5(24):12908–12915. 10.18632/oncotarget.2679 Xue H, Guo X, Han X,Yan S, Zhang J, Xu S, Li T, Zhang P, Gao X, Liu Q, Li G, 2016, MicroRNA-584-3p, a novel tumor suppressor and prognostic marker, reduces the migration and invasion of human glioma cells by targeting hypoxia-induced ROCK1. Oncotarget 7(4):4785–4805. 10.18632/oncotarget.6735 Wang XP, Deng XL, Li LY, 2014, MicroRNA-584 functions as a tumor suppressor and targets PTTG1IP in glioma. Int J Clin Exp Pathol 7(12):8573–8582 Guled M, Lahti L, Lindholm PM, Salmenkivi K, Bagwan I, Nicholson AG, Knuutila S, 2009, CDKN2A, NF2, and JUN are dysregulated among other genes by miRNAs in malignant mesothelioma -A miRNA microarray analysis. Genes Chromosomes Cancer 48(7):615–623. , DOI 10.1002/gcc.20669 Gaedcke J, GradeM, Camps J, Sokilde R, Kaczkowski B, Schetter AJ, Difilippantonio MJ, Harris CC, Ghadimi BM, Moller S, Beissbarth T, Ried T, Litman T, 2012, The rectal cancer microRNAome–microRNA expression in rectal cancer and matched normal mucosa. Clin Cancer Res 18(18):4919–4930. , DOI 10.1158/1078-0432.CCR-12-0016 Ueno K, Hirata H, Shahryari V, Chen Y, Zaman MS, Singh K, Tabatabai ZL, Hinoda Y, Dahiya R, 2011, Tumour suppressor microRNA-584 directly targets oncogene Rock-1 and decreases invasion ability in human clear cell renal cell carcinoma. Br J Cancer 104(2):308–315. , DOI 10.1038/sj.bjc.6606028 Xiang J, Wu Y, Li DS, Wang ZY, Shen Q, Sun TQ, Guan Q, Wang YJ, 2015, miR-584 Suppresses Invasion and Cell Migration of Thyroid Carcinoma by Regulating the Target Oncogene ROCK1. Oncol Res Treat 38(9):436–440. , DOI 10.1159/000438967 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 11 EP 20 DI 10.1007/s12253-017-0316-1 PG 10 ER PT J AU Nagy, Garzuly, F Padanyi, G Szucs, I Feldmann, Murnyak, B Hortobagyi, T Kalman, B AF Nagy, Adam Garzuly, Ferenc Padanyi, Gergely Szucs, Ivan Feldmann, Adam Murnyak, Balazs Hortobagyi, Tibor Kalman, Bernadette TI Molecular Subgroups of Glioblastoma– an Assessment by Immunohistochemical Markers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma; Molecular subgroups; Translation; Clinical setting ID Glioblastoma; Molecular subgroups; Translation; Clinical setting AB Comprehensive molecular characterization of and novel therapeutic approaches to glioblastoma have been explored as a result of advancements in biotechnologies. In this study, we aimed to bring basic research discoveries closer to clinical practice and ultimately incorporate molecular classification into the routine histopathological evaluation of grade IV gliomas. Integrated results of genome-wide sequencing, transcriptomic and epigenomic analyses by The Cancer Genome Atlas Network defined the classic, proneural, neural and mesenchymal subtypes of this tumor. In a retrospective cohort, we analyzed selected subgroup-defining molecular markers in formalin-fixed paraffin-embedded surgical specimens by immunohistochemistry. Quantitative and qualitative scores of marker expression were tested in hierarchical cluster analyses to evaluate segregations of the molecular subgroups, which then were correlated with clinical parameters including patients’ age, gender and overall survival. Our study has confirmed the separation of molecular glioblastoma subgroups with clear trends regarding clinical correlations. Future analyses in a larger, prospective cohort using similar methods are expected to facilitate the development of a molecular diagnostic panel that may complement routine histological work up and support prognostication as well as treatment decisions in glioblastoma. C1 [Nagy, Adam] University of Pecs, Faculty of Medicine, Department of Public HealthPecs, Hungary. [Garzuly, Ferenc] Vas County Markusovszky Hospital, 9700 Szombathely, Hungary. [Padanyi, Gergely] Vas County Markusovszky Hospital, 9700 Szombathely, Hungary. [Szucs, Ivan] St Borbala Hospital, Faculty of Medicine, Institute of Behavioral SciencesTatabanya, Hungary. [Feldmann, Adam] University of PecsPecs, Hungary. [Murnyak, Balazs] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Hortobagyi, Tibor] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Kalman, Bernadette] University of Pecs, Faculty of Medicine, Department of Public HealthPecs, Hungary. RP Kalman, B (reprint author), University of Pecs, Faculty of Medicine, Department of Public Health, Pecs, Hungary. EM bernadett.kalman@etk.pte.hu;kalman.bernadett@markusovszky.hu CR Stupp R, Mason WP, Van Den Bent MJ, Weller M, Fisher B, Taphoorn MJ et al, 2005, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl JMed 352(10):987– 996 Walker MD, Green SB, Byar DP, Alexander E Jr, Batzdorf U, Brooks WH et al, 1980, Randomized comparisons of radiotherapy and nitrosoureas for the treatment ofmalignant glioma after surgery. N Engl J Med 303(23):1323–1329 Laperriere N, Zuraw L, Cairncross G, Cancer Care Ontario Practice Guidelines Initiative Neuro-Oncology Disease Site Group, 2002, Radiotherapy for newly diagnosed malignant glioma in adults: a systematic review. Radiother Oncol 64(3):259–273 4 . Chaurasia A, Park SH, Seo JW, Park CK, 2016, Immunohistochemical analysis of ATRX, IDH1 and p53 in Glioblastoma and their correlations with patient survival. J Korean Med Sci 31(8):1208–1214 5. Eidel O, Burth S, Neumann JO, Kieslich PJ, Sahm F, Jungk C et al, 2017, Tumor infiltration in enhancing and non-enhancing parts of Glioblastoma: a correlation with histopathology. PLoS One 12(1): e0169292 6. The Cancer Genome Atlas, TCGA, Research Network, 2008, Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455:1061–1068 7. Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD et al, 2010, Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17(1):98–110 8. Brennan CW,Verhaak RG, McKennaA, Campos B, Noushmehr H, Salama SR et al, 2013, The somatic genomic landscape of glioblastoma. Cell 155(2):462–477 9. AubryM, de TayracM, Etcheverry A, Clavreul A, Saikali S,Menei P et al, 2016, From the core to beyond the margin: a genomic picture of glioblastoma intratumor heterogeneity. Oncotarget 6(14):12094–12109 10. Noushmehr H, Weisenberger DJ, Diefes K, Phillips HS, Pujara K, Berman BP et al, 2010, Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17(5):510–522 11. Segerman A, Niklasson M, Haglund C, Bergstrom T, Jarvius M, Xie Y et al, 2016, Clonal variation in drug and radiation response among Glioma-initiating cells is linked to proneural-Mesenchymal transition. Cell Rep 17(11):2994–3009 12. He ZC, Ping YF, Xu SL, LinY,Yu SC, KungHF et al, 2015, Lower MGMTexpression predicts better prognosis in proneural-like glioblastoma. Int J Clin Exp Med 8(11):20287–20294 13. Myung JK, jin Cho H, Kim H, Park CK, Lee SH, Choi SH et al, 2014, Prognosis of glioblastoma with oligodendroglioma component is associated with the IDH1 mutation and MGMT methylation status. Transl Oncol 7(6):712–719 14. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella- Branger D, Cavenee WKet al., 2016, The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol 131(6):803–820 15. Le Mercier M, Hastir D, Lopez XM, De Neve N, Maris C, Trepant AL et al, 2012, A simplified approach for the molecular classification of glioblastomas. PLoS One 7(9):e45475 16. Lee KS, Choe G, Nam KH, Seo AN, Yun S, Kim KJ et al, 2013, Immunohistochemical classification of primary and secondary glioblastomas. Korean J Pathol 47(6):541–548 17. Conroy S, Kruyt FA, Joseph JV, Balasubramaniyan V, Bhat KP, Wagemakers M et al, 2014, Subclassification of newly diagnosed glioblastomas through an immunohistochemical approach. PLoS One 9(12):e115687 18. Esteve-Codina A, Arpi O, Martinez-Garcia M, Pineda E, Mallo M, Gut M et al, 2017, A comparison of RNA-Seq results from paired formalin-fixed paraffin-embedded and fresh-frozen Glioblastoma tissue samples. PLoS One 12(1):e0170632 19. Colman H, Zhang Z, Sulma EP, McDonald JM, Shooshtari NL, Rivera A et al, 2010, A multigene predictor of outcome in glioblastoma. Neuro-Oncology 12(1):49–57 20. Joseph NM, Phillips J, Dahiya S, Felicella MM, Tihan T, Brat DJ et al, 2013, Diagnostic implications of IDH1-R132H and OLIG2 expression patterns in rare and challenging glioblastoma variants. Mod Pathol 26(3):315–326 21. Chen L, Lin ZX, Li GS, Zhou CF, Chen YP, Wang XF et al, 2015, Classification of microvascular patterns via cluster analysis reveals their prognostic significance in glioblastoma. Hum Pathol 46(1): 120–128 22. Denicolai E, Tabouret E, Colin C, Metellus P, Nanni I, Boucard C et al, 2016, Molecular heterogeneity of glioblastomas: does location matter? Oncotarget 7(1):902–913 23. Cai J, Zhang C, Zhan W, Wang G, Yao K, Wang Z et al, 2016, ATRX, IDH1-R132HandKi-67 immunohistochemistry as a classification scheme for astrocytic tumors. Oncoscience 3(7–8): 258–265 24. Kalman B, Szep E, Garzuly F, 2013, Post DE. Epidermal growth factor receptor as a therapeutic target in glioblastoma. NeuroMolecular Med 15(2):420–434 25. Lindberg OR, McKinney A, Engler JR, Koshkakaryan G, Gong H, Robinson AE et al, 2016, GBM heterogeneity as a function of variable epidermal growth factor receptor variant III activity. Oncotarget 7(48):79101–79116 26. Vizcaino MA, Shah S, Eberhart CG, Rodriguez FJ, 2015, Clinicopathologic implications of NF1 gene alterations in diffuse gliomas. Hum Pathol 46(9):1323–1330 27. Xie B, Fan X, Lei Y, Chen R,Wang J, Fu C et al, 2016, A novel de novo microdeletion at 17q11. 2 adjacent to NF1 gene associated with developmental delay, short stature, microcephaly and dysmorphic features. Mol Cytogenet 9(1):31–41 28. Way GP, Allaway RJ, Bouley SJ, Fadul CE, Sanchez Y, Greene CS, 2017, A machine learning classifier trained on cancer transcriptomes detects NF1 inactivation signal in glioblastoma. BMC Genomics 18(1):127–128 29. Nagy A, Eder K, Selak MA, Kalman B, 2015, Mitochondrial energy metabolism and apoptosis regulation in glioblastoma. Brain Res 1595:127–142 30. Setty P, Hammes J, Rothamel T, Vladimirova V, Kramm CM, Pietsch T et al, 2010, A pyrosequencing-based assay for the rapid detection of IDH1 mutations in clinical samples. J Mol Diagn 12(6):750–756 31. Bardella C, Al-Dalahmah O, Krell D, Brazauskas P, Al-Qahtani K, Tomkova M et al, 2016, Expression of Idh1 R132H in the murine subventricular zone stem cell niche recapitulates features of early Gliomagenesis. Cancer Cell 30(4):578–594 32. Zhu H, Zhang Y, Chen J, Qiu J, Huang K, Wu M, Xia C, 2017, IDH1 R132H mutation enhances cell migration by activating AKTmTOR signaling pathway, but sensitizes cells to 5-FU treatment as NADPH and GSH are reduced. PLoS One 12(1):e0169038 33. Yoon KS, LeeMC, Kang SS, Kim JH, Jung S, Kim YJ et al, 2001, p53 mutation and epidermal growth factor receptor overexpression in glioblastoma. J Korean Med Sci 16(4):481–488 34. Liu Y, Wang F, Liu Y, Yao Y, Lv X, Dong B et al, 2016, RNF135, RING finger protein, promotes the proliferation of human glioblastoma cells in vivo and in vitro via the ERK pathway. Sci Rep 6: e20624 35. Stieber D, Golebiewska A, Evers L, Lenkiewicz E, Brons NH, Nicot N et al, 2014, Glioblastomas are composed of genetically divergent clones with distinct tumourigenic potential and variable stem cell-associated phenotypes. Acta Neuropathol 127(2): 203–219 36. Cancer Genome Atlas, TCGA, Research Network, 2015, Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 517(7536):576–582 37. Eckel-Passow JE, Lachanc DH,Molinaro AM,Walsh KM, Decker PA, Sicotte H et al, 2015, Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors. N Engl J Med 372(26): 2499–2508 38. Olar A, Aldape KD, 2014, Using the molecular classification of glioblastoma to inform personalized treatment. J Pathol 232(2): 165–177 39. Murnyak B, Csonka T, Klekner A, Hortobagyi T, 2013, Occurrence and molecular pathology of low grade gliomas. Alacsony gradusu glialis daganatok elofordulasa es molekularis patologiaja., in Hungarian, Ideggyogyaszati Szemle/Clinical. Neuroscience 66:305–311 40. Murnyak B, Csonka T, Hegyi K, Mehes G, Klekner A, Hortobagyi T, 2013, Occurrence and molecular pathology of high grade gliomas., Magas gradusu gliomak elofordulasa es molekularis patologiaja.,, in Hungarian, Ideggyogyaszati Szemle/Clinical. Neuroscience 66:312–321 41. McKeever PE, Dennis TR, Burgess AC,Meltzer PS, Marchuk DA, Trent JM, 1996, Chromosome breakpoint at 17q11. 2 and insertion of DNA from three different chromosomes in a glioblastoma with exceptional glial fibrillary acidic protein expression. Cancer Genet Cytogenet 87(1):41–47 42. Vandenbroucke I,Van Oostveldt P, Coene E,De PaepeA,Messiaen L, 2004, Neurofibromin is actively transported to the nucleus. FEBS Lett 560(1–3):98–102 43. Liu YC, Wang YZ, 2015, Role of yes-associated protein 1 in gliomas: pathologic and therapeutic aspects. Tumor Biol 36(4): 2223–2227 44. Kondo I, Shimizu N, 1983, Mapping of the human gene for epidermal growth factor receptor, EGFR, on the p13→ q22 region of chromosome 7. Cytogenet Genome Res 35(1):9–14 45. Lopez-Gines C, Gil-Benso R, Ferrer-Luna R, Benito R, Serna E, Gonzalez-Darder J et al, 2010, New pattern of EGFR amplification in glioblastoma and the relationship of gene copy number with gene expression profile. Mod Pathol 23(6):856–865 46. Dasari VR, Velpula KK, Alapati K, Gujrati M, Tsung AJ, 2012, Cord blood stem cells inhibit epidermal growth factor receptor translocation to mitochondria in glioblastoma. PLoS One 7(2):e31884 47. Csonka T, Murnyak B, Szepesi R, Kurucz A, Klekner A, Hortobagyi T, 2014, Poly(ADP-ribose, polymerase-1, PARP1, and p53 labelling index correlates with tumour grade in meningiomas. Folia Neuropathol 52:111–120 48. Csonka T, Murnyak B, Szepesi R, Bencze J, Bognar L, Klekner A, Hortobagyi T, 2016, Assessment of candidate immunohistochemical prognostic markers of meningioma recurrence. Folia Neuropathol 54:114–126 49. Murnyak B, Kouhsari MC, Hershkovitch R, Kalman B, Marko- Varga G, Klekner A, Hortobagyi T, 2017, PARP1 expression and its correlation with survival is tumour molecular subtype dependent in glioblastoma.Oncotarget 8(28):46348–46362. , DOI 10. 18632/oncotarget.18013 50. Murnyak B, Hortobagyi T, 2016, Immunohistochemical correlates of TP53 somatic mutations in cancer. Oncotarget 7:64910–64920 51. Masuda H, Miller C, Koeffler HP, Battifora H, Cline MJ, 1987, Rearrangement of the p53 gene in human osteogenic sarcomas. Proc Natl Acad Sci 84(21):7716–7719 52. England B, Huang T, KarsyM(2013, Current understanding of the role and targeting of tumor suppressor p53 in glioblastoma multiforme. Tumor Biol 34(4):2063–2074 53. Smardova J, Liskova K, Ravcukova B, Kubiczkova L, Sevcikova S, Michalek J et al, 2013, High frequency of temperature-sensitive mutants of p53 in Glioblastoma. Pathol Oncol Res 19(3):421–428 54. Kawasoe T, Takeshima H, Yamashit S, Mizuguch S, Fukushima T, Yokogami K et al, 2015, Detection of p53 mutations in proliferating vascular cells in glioblastoma multiforme. J Neurosurg 122(2): 317–323 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 21 EP 31 DI 10.1007/s12253-017-0311-6 PG 11 ER PT J AU Singh, AS Ghosh, KS AF Singh, Anil Seram Ghosh, Kumar Sankar TI Metabolic Phase I (CYPs) and Phase II (GSTs) Gene Polymorphisms and Their Interaction with Environmental Factors in Nasopharyngeal Cancer from the Ethnic Population of Northeast India SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Nasopharyngeal carcinoma (NPC); Polymorphisms in metabolizing genes; Gene-gene interaction; Tobacco smokers and chewers; Gene-environment interaction; Northeast India ID Nasopharyngeal carcinoma (NPC); Polymorphisms in metabolizing genes; Gene-gene interaction; Tobacco smokers and chewers; Gene-environment interaction; Northeast India AB Multiple genetic and environmental factors and their interaction are believed to contribute in the pathogenesis of Nasopharyngeal Cancer (NPC). We investigate the role of Metabolic Phase I (CYPs) and Phase II (GSTs) gene polymorphisms, gene-gene and gene-environmental interaction in modulating the susceptibility to NPC in Northeast India. To determine the association of metabolic gene polymorphisms and environmental habits, 123 cases and 189 controls blood/ swab samples were used for PCR and confirmed by Sanger sequencing. Analysis for GSTM1 and GSTT1 gene polymorphism was done by multiplex PCR. The T3801C in the 3′- flanking region of CYP1A1 gene was detected by PCR-RFLP method. The Logistic regression analysis was used to estimate odds ratios (OR) and 95% confidence intervals (95% CI). The GSTM1 null genotype alone (OR = 2.76) was significantly associated with NPC risk (P < 0.0001). The combinations of GSTM1 null and GSTT1 null genotypes also higher, 3.77 fold (P < 0.0001), risk of NPC, while GSTM1 null genotype along with CYP1A1 T3801C TC + CC genotype had 3.22 (P = 0.001) fold risk. The most remarkable risk was seen among individual carrying GSTM1 null, GSTT1 null genotypes and CYP1A1 T3801C TC + CC genotypes (OR = 5.71, P = 0.001). Further; analyses demonstrate an enhanced risk of NPC in smoked meat (OR = 5.56, P < 0.0001) and fermented fish consumers (OR = 5.73, P < 0.0001) carrying GSTM1 null genotype. An elevated risk of NPC was noted in smokers (OR = 12.67, P < 0.0001) and chewers (OR = 5.68, P < 0.0001) with GSTM1 null genotype. However, smokers had the highest risk of NPC among individuals carrying GSTT1 null genotype (OR = 4.46, P = 0.001) or CYP1A1 T3801C TC + CC genotype (OR = 7.13, P < 0.0001). The association of null genotypes and mutations of metabolic neutralizing genes along with the environmental habits (tobacco smokers and chewers, smoke meat, fermented fishes) can be used as a possible biomarker for early detection and preventive measure of NPC. C1 [Singh, Anil Seram] Assam University, Department of Biotechnology, Molecular Medicine Laboratory, 788011 Silchar, Assam, India. [Ghosh, Kumar Sankar] Assam University, Department of Biotechnology, Molecular Medicine Laboratory, 788011 Silchar, Assam, India. RP Ghosh, KS (reprint author), Assam University, Department of Biotechnology, Molecular Medicine Laboratory, 788011 Silchar, India. EM drsankarghosh@gmail.com CR Chang ET, Adami HO, 2006, The enigmatic epidemiology of nasopharyngeal carcinoma. Cancer Epidemiol Biomark Prev 15(10): 1765–1777. , DOI 10.1158/1055-9965.EPI-06-0353 Kataki AC, Simons MJ, Das AK, Sharma K, Mehra NK, 2011, Nasopharyngeal carcinoma in the Northeastern states of India. Chin J Cancer 30(2):106–113 Ghosh SK, Singh AS, Mondal R, Kapfo W, Khamo V, Singh YI, 2014, Dysfunction of mitochondria due to environmental carcinogens in nasopharyngeal carcinoma in the ethnic group of northeast Indian population. Tumour Biol J Int Soc Oncodev Biol Med. , DOI 10.1007/s13277-014-1897-x Tsao SW, Yip YL, TsangCM, Pang PS, LauVM, Zhang G, LoKW, 2014, Etiological factors of nasopharyngeal carcinoma. Oral Oncol 50(5):330–338. , DOI 10.1016/j.oraloncology. 2014.02.006 Jia WH, Luo XY, Feng BJ, Ruan HL, Bei JX, Liu WS, Qin HD, Feng QS, Chen LZ, Yao SY, Zeng YX, 2010, Traditional Cantonese diet and nasopharyngeal carcinoma risk: a large-scale case-control study in Guangdong, China. BMC Cancer 10:446. , DOI 10.1186/1471-2407-10-446 Belbaraka R, Lalya I, Boulaamane L, Tazi M, Benjaafar N, Errihani H, 2013, Dietary risk factors of undifferenced nasopharyngeal carcinoma: a case-control study. Tunis Med 91(6):406–409 Gallicchio L, Matanoski G, Tao XG, Chen L, Lam TK, Boyd K, Robinson KA, Balick L, Mickelson S, Caulfield LE, Herman JG, Guallar E, Alberg AJ, 2006, Adulthood consumption of preserved and nonpreserved vegetables and the risk of nasopharyngeal carcinoma: a systematic review. Int J Cancer 119(5):1125–1135. https:// doi.org/10.1002/ijc.21946 Hildesheim A, West S, DeVeyra E, De Guzman MF, Jurado A, Jones C, Imai J, Hinuma Y, 1992, Herbal medicine use, Epstein- Barr virus, and risk of nasopharyngeal carcinoma. Cancer Res 52(11):3048–3051 Chelleng PK, Narain K, Das HK, ChetiaM,Mahanta J, 2000, Risk factors for cancer nasopharynx: a case-control study from Nagaland, India. Natl Med J India 13(1):6–8 Xue WQ, Qin HD, Ruan HL, Shugart YY, Jia WH, 2013, Quantitative association of tobacco smoking with the risk of nasopharyngeal carcinoma: a comprehensive meta-analysis of studies conducted between 1979 and 2011. Am J Epidemiol 178(3):325– 338. , DOI 10.1093/aje/kws479 Fachiroh J, Sangrajrang S, Johansson M, Renard H, Gaborieau V, Chabrier A, Chindavijak S, Brennan P, McKay JD, 2012, Tobacco consumption and genetic susceptibility to nasopharyngeal carcinoma, NPC, in Thailand. Cancer Causes Control: CCC 23(12):1995– 2002. , DOI 10.1007/s10552-012-0077-9 Ruan HL, Xu FH, LiuWS, Feng QS, Chen LZ, Zeng YX, Jia WH, 2010, Alcohol and tea consumption in relation to the risk of nasopharyngeal carcinoma in Guangdong, China. Front Med China 4(4):448–456. , DOI 10.1007/s11684-010-0280-6 Chen L, Gallicchio L, Boyd-Lindsley K, Tao XG, Robinson KA, Lam TK, Herman JG, Caulfield LE, Guallar E, Alberg AJ, 2009, Alcohol consumption and the risk of nasopharyngeal carcinoma: a systematic review. Nutr Cancer 61(1):1–15. , DOI 10. 1080/01635580802372633 Bei JX, Li Y, Jia WH, Feng BJ, Zhou G, Chen LZ, Feng QS, Low HQ, ZhangH, He F, Tai ES, Kang T, Liu ET, Liu J, ZengYX(2010, A genome-wide association study of nasopharyngeal carcinoma identifies three new susceptibility loci. Nat Genet 42(7):599–603. , DOI 10.1038/ng.601 Diggs DL, Huderson AC, Harris KL, Myers JN, Banks LD, Rekhadevi PV, Niaz MS, Ramesh A, 2011, Polycyclic aromatic hydrocarbons and digestive tract cancers: a perspective. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 29(4):324–357. , DOI 10.1080/10590501.2011.629974 Lachenmeier DW, Przybylski MC, Rehm J, 2012, Comparative risk assessment of carcinogens in alcoholic beverages using the margin of exposure approach. Int J Cancer 131(6):E995–1003. , DOI 10.1002/ijc.27553 Mondal R, Ghosh SK, 2013, Accumulation of mutations over the complete mitochondrial genome in tobacco-related oral cancer from northeast India. Mitochondrial DNA 24(4):432–439. https://doi. org/10.3109/19401736.2012.760551 Xue W, Warshawsky D, 2005, Metabolic activation of polycyclic and heterocyclic aromatic hydrocarbons and DNA damage: a review. Toxicol Appl Pharmacol 206(1):73–93. , DOI 10. 1016/j.taap.2004.11.006 Shukla D, Dinesh Kale A, Hallikerimath S, Yerramalla V, Subbiah V, Mishra S, 2013, Association between GSTM1 and CYP1A1 polymorphisms and survival in oral cancer patients. Biomed Pap Med Fac Univ Palacky Olomouc Czechoslovakia 157(4):304–310. , DOI 10.5507/bp.2013.028 Rodriguez-Antona C, Ingelman-Sundberg M, 2006, Cytochrome P450 pharmacogenetics and cancer. Oncogene 25(11):1679–1691. , DOI 10.1038/sj.onc.1209377 Mota P,Moura DS, ValeMG, Coimbra H, Carvalho L, Regateiro F, 2010, CYP1A1 m1 and m2 polymorphisms: genetic susceptibility to lung cancer. Rev Port Pneumol 16(1):89–98 Sabitha K, Reddy MV, Jamil K, 2010, Smoking related risk involved in individuals carrying genetic variants of CYP1A1 gene in head and neck cancer. Cancer Epidemiol 34(5):587–592. , DOI 10.1016/j.canep.2010.05.002 Sergentanis TN, Economopoulos KP, 2010, Four polymorphisms in cytochrome P450 1A1, CYP1A1, gene and breast cancer risk: a meta-analysis. Breast Cancer Res Treat 122(2):459–469. https:// doi.org/10.1007/s10549-009-0694-5 Mondal R, Ghosh SK, Choudhury JH, Seram A, Sinha K, Hussain M, Laskar RS, Rabha B, Dey P, Ganguli S, Nathchoudhury M, Talukdar FR, Chaudhuri B, Dhar B, 2013, Mitochondrial DNA copy number and risk of oral cancer: a report from Northeast India. PLoS One 8(3):e57771. , DOI 10.1371/journal. pone.0057771 Nosheen M, Ishrat M, Malik FA, Baig RM, Kayani MA, 2010, Association ofGSTM1 andGSTT1 gene deletions with risk of head and neck cancer in Pakistan: a case control study. Asian Pac J Cancer Prev 11(4):881–885 Sharma A, Das BC, Sehgal A, Mehrotra R, Kar P, Sardana S, Phukan R, Mahanta J, Purkayastha J, Saxena S, Kapur S, Chatterjee I, Sharma JK, 2013, GSTM1 and GSTT1 polymorphism and susceptibility to esophageal cancer in high- and low-risk regions of India. Tumour Biol J Int Soc Oncodev Biol Med 34(5): 3249–3257. , DOI 10.1007/s13277-013-0897-6 Shukla RK, Tilak AR, Kumar C, Kant S, Kumar A, Mittal B, Bhattacharya S, 2013, Associations of CYP1A1, GSTM1 and GSTT1 polymorphisms with lung cancer susceptibility in a Northern Indian population. Asian Pac J Cancer Prev 14(5):3345– 3349 Ihsan R, Chauhan PS, Mishra AK, Yadav DS, Kaushal M, Sharma JD, Zomawia E, Verma Y, Kapur S, Saxena S, 2011, Multiple analytical approaches reveal distinct gene-environment interactions in smokers and non smokers in lung cancer. PLoS One 6(12): e29431. , DOI 10.1371/journal.pone.0029431 Ghosh SK, Mondal R, 2012, Quick diagnosis of female genital tuberculosis using multiplex fast polymerase chain reaction in Southern Assam, India. Int J Gynaecol Obstet Off Organ Int Fed Gynaecol Obstet 118(1):72–73. , DOI 10.1016/j.ijgo.2012. 02.006 Mondal R, Ghosh SK, Talukdar FR, Laskar RS, 2013, Association of mitochondrial D-loop mutations with GSTM1 and GSTT1 polymorphisms in oral carcinoma: a case control study from northeast India. Oral Oncol 49(4):345–353. , DOI 10.1016/j. oraloncology.2012.11.003 Kiruthiga PV, Kannan MR, Saraswathi C, Pandian SK, Devi KP, 2011, CYP1A1 gene polymorphisms: lack of association with breast cancer susceptibility in the southern region, Madurai, of India. Asian Pac J Cancer Prev 12(8):2133–2138 Choudhury JH, Choudhury B, Kundu S, Ghosh SK, 2014, Combined effect of tobacco and DNA repair genes polymorphisms of XRCC1 and XRCC2 influence high risk of head and neck squamous cell carcinoma in northeast Indian population. Med Oncol 31(8):67. , DOI 10.1007/s12032-014-0067-8 Choudhury JH, Ghosh SK, 2014, Gene-environment interaction and susceptibility in head and neck cancer patients and in their first-degree relatives: a study of Northeast Indian population. J Oral Pathol Med Off Publ Int Assoc Oral Pathol Am Acad Oral Pathol. , DOI 10.1111/jop.12249 McIlwain CC, Townsend DM, Tew KD, 2006, Glutathione Stransferase polymorphisms: cancer incidence and therapy. Oncogene 25(11):1639–1648. , DOI 10.1038/sj.onc. 1209373 Sarkar S, Nagabhushan M, Soman CS, Tricker AR, Bhide SV, 1989, Mutagenicity and carcinogenicity of smoked meat from Nagaland, a region of India prone to a high incidence of nasopharyngeal cancer. Carcinogenesis 10(4):733–736 Guo X, O'Brien SJ, Zeng Y, Nelson GW, Winkler CA, 2008, GSTM1 and GSTT1 gene deletions and the risk for nasopharyngeal carcinoma in Han Chinese. Cancer Epidemiol Biomark Prev 17(7): 1760–1763. , DOI 10.1158/1055-9965.EPI-08-0149 Jiang Y, Li N, Dong P, Zhang N, Sun Y, Han M, Wen J, Chen M, 2011, Polymorphisms in GSTM1, GSTTI and GSTP1 and nasopharyngeal cancer in the east of China: a case-control study. Asian Pac J Cancer Prev 12(11):3097–3100 Wei Y, Zhou T, Lin H, Sun M, Wang D, Li H, Li B, 2013, Significant associations between GSTM1/GSTT1 polymorphisms and nasopharyngeal cancer risk. Tumour Biol J Int Soc Oncodev Biol Med 34(2):887–894. , DOI 10.1007/s13277-012- 0623-9 Cheng YJ, Chien YC, HildesheimA, HsuMM, Chen IH, Chuang J, Chang J,Ma YD, Luo CT, HsuWL, Hsu HH, Huang H, Chang JF, Chen CJ, Yang CS, 2003, No association between genetic polymorphisms of CYP1A1, GSTM1, GSTT1, GSTP1, NAT2, and nasopharyngeal carcinoma in Taiwan. Cancer Epidemiol Biomark Prev 12(2):179–180 Anantharaman D, Chaubal PM, Kannan S, Bhisey RA, Mahimkar MB, 2007, Susceptibility to oral cancer by genetic polymorphisms at CYP1A1, GSTM1 and GSTT1 loci among Indians: tobacco exposure as a risk modulator. Carcinogenesis 28(7):1455–1462. , DOI 10.1093/carcin/bgm038 Soya SS, Vinod T, Reddy KS, Gopalakrishnan S, Adithan C, 2007, Genetic polymorphisms of glutathione-S-transferase genes, GSTM1, GSTT1 and GSTP1, and upper aerodigestive tract cancer risk among smokers, tobacco chewers and alcoholics in an Indian population. Eur J Cancer 43(18):2698–2706. , DOI 10. 1016/j.ejca.2007.07.006 Ghosh SK, Singh AS, Mondal R, Kapfo W, Khamo V, Singh YI, 2014, Dysfunction of mitochondria due to environmental carcinogens in nasopharyngeal carcinoma in the ethnic group of Northeast Indian population. Tumour Biol J Int Soc Oncodev BiolMed 35(7): 6715–6724. , DOI 10.1007/s13277-014-1897-x Sam SS, Thomas V, Reddy SK, Surianarayanan G, Chandrasekaran A, 2008, CYP1A1 polymorphisms and the risk of upper aerodigestive tract cancers in an Indian population. Head Neck 30(12):1566–1574. , DOI 10.1002/hed.20897 Chandirasekar R, Kumar BL, Sasikala K, Jayakumar R, Suresh K, Venkatesan R, Jacob R, Krishnapriya EK, Kavitha H, Ganesh GK, 2014, Assessment of genotoxic andmolecular mechanisms of cancer risk in smoking and smokeless tobacco users. Mutat Res Genet Toxicol Environ Mutagen 767C:21–27. , DOI 10.1016/j. mrgentox.2014.04.007 Bartsch H, Rojas M, Nair U, Nair J, Alexandrov K, 1999, Genetic cancer susceptibility and DNA adducts: studies in smokers, tobacco chewers, and coke oven workers. Cancer Detect Prev 23(6):445– 453 Sharma R, Ahuja M, Panda NK, Khullar M, 2011, Interactions among genetic variants in tobacco metabolizing genes and smoking are associated with head and neck cancer susceptibility in north Indians. DNA Cell Biol 30(8):611–616. , DOI 10.1089/ dna.2010.1184 Matthias C, BockmuhlU, JahnkeV, JonesPW,Hayes JD, Alldersea J, Gilford J, Bailey L, Bath J, Worrall SF, Hand P, Fryer AA, Strange RC, 1998, Polymorphism in cytochrome P450 CYP2D6, CYP1A1, CYP2E1 and glutathione S-transferase, GSTM1, GSTM3, GSTT1 and susceptibility to tobacco-related cancers: studies in upper aerodigestive tract cancers. Pharmacogenetics 8(2):91–100 Varela-Lema L, Taioli E, Ruano-Ravina A, Barros-Dios JM, Anantharaman D, Benhamou S, Boccia S, Bhisey RA, Cadoni G, Capoluongo E, Chen CJ, Foulkes W, Goloni-Bertollo EM, Hatagima A, Hayes RB, Katoh T, Koifman S, Lazarus P, Manni JJ, Mahimkar M, Morita S, Park J, Park KK, Pavarino Bertelli EC, de Souza Fonseca Ribeiro EM, Roy B, Spitz MR, Strange RC,Wei Q, Ragin CC, 2008)Meta-analysis and pooled analysis of GSTM1 and CYP1A1 polymorphisms and oral and pharyngeal cancers: a HuGE-GSEC review. Genet Med Off J Am CollMed Genet 10(6): 369–384. , DOI 10.1097/GIM.0b013e3181770196 Choudhury JH, Ghosh SK, 2015, Promoter hypermethylation profiling identifies subtypes of head and neck cancer with distinct viral, environmental, genetic and survival characteristics. PLoS One 10(6):e0129808. , DOI 10.1371/journal.pone.0129808 Laskar RS, Talukdar FR, Choudhury JH, Singh SA, Kundu S, Dhar B, Mondal R, Ghosh SK, 2015, Association of HPV with genetic and epigenetic alterations in colorectal adenocarcinoma from Indian population. Tumour Biol J Int Soc Oncodev Biol Med 36(6):4661– 4670. , DOI 10.1007/s13277-015-3114-y Pietrusinski M, Kepczynski L, Jedrzejczyk A, Borkowska E, Traczyk-Borszynska M, Constantinou M, Kauzewski B, BorowiecM(2016, Detection of bladder cancer in urine sediments by a hypermethylation panel of selected tumor suppressor genes. Cancer Biomark Sect A Dis Mark. , DOI 10.3233/CBM- 160673 JiangW, Cai R, Chen QQ, 2015, DNA methylation biomarkers for nasopharyngeal carcinoma: diagnostic and prognostic tools. Asian Pac J Cancer Prev 16(18):8059–8065 Lo KW, Chung GT, To KF, 2012, Deciphering the molecular genetic basis of NPC through molecular, cytogenetic, and epigenetic approaches. Semin Cancer Biol 22(2):79–86. , DOI 10. 1016/j.semcancer.2011.12.011 Dai W, Zheng H, Cheung AK, Lung ML, 2016, Genetic and epigenetic landscape of nasopharyngeal carcinoma. Chinese. Clin Oncol 5(2):16. 10.21037/cco.2016.03.06 Talukdar FR, Ghosh SK, Laskar RS, Kannan R, Choudhury B, Bhowmik A, 2015, Epigenetic pathogenesis of human papillomavirus in upper aerodigestive tract cancers. Mol Carcinog 54(11): 1387–1396. , DOI 10.1002/mc.22214 Lleras RA, Smith RV, Adrien LR, Schlecht NF, Burk RD, Harris TM, Childs G, Prystowsky MB, Belbin TJ, 2013, Unique DNA methylation loci distinguish anatomic site and HPV status in head and neck squamous cell carcinoma. Clin Cancer Res 19(19):5444– 5455. , DOI 10.1158/1078-0432.CCR-12-3280 Talukdar FR, Ghosh SK, Laskar RS, Mondal R, 2013, Epigenetic, genetic and environmental interactions in esophageal squamous cell carcinoma from northeast India. PLoS One 8(4):e60996. https://doi. org/10.1371/journal.pone.0060996 Zhou L, Jiang W, Ren C, Yin Z, Feng X, Liu W, Tao Q, Yao K, 2005, Frequent hypermethylation of RASSF1A and TSLC1, and high viral load of Epstein-Barr virus DNA in nasopharyngeal carcinoma and matched tumor-adjacent tissues. Neoplasia 7(9): 809–815 Tong JH, Tsang RK, LoKW,Woo JK, Kwong J, ChanMW, Chang AR, van Hasselt CA, Huang DP, To KF, 2002, Quantitative Epstein-Barr virus DNA analysis and detection of gene promoter hypermethylation in nasopharyngeal, NP, brushing samples from patients with NP carcinoma. Clin Cancer Res 8(8):2612–2619 Tsai CN, Tsai CL, Tse KP, Chang HY, Chang YS, 2002, The Epstein-Barr virus oncogene product, latent membrane protein 1, induces the downregulation of E-cadherin gene expression via activation of DNA methyltransferases. Proc Natl Acad Sci U S A 99(15):10084–10089. , DOI 10.1073/pnas.152059399 Skalska L, White RE, Franz M, Ruhmann M, Allday MJ, 2010, Epigenetic repression of p16(INK4A, by latent Epstein-Barr virus requires the interaction of EBNA3A and EBNA3C with CtBP. PLoS Pathog 6(6):e1000951. , DOI 10.1371/journal.ppat. 1000951 Kulkarni V, Saranath D, 2004, Concurrent hypermethylation of multiple regulatory genes in chewing tobacco associated oral squamous cell carcinomas and adjacent normal tissues. Oral Oncol 40(2):145–153 Hasegawa M, Nelson HH, Peters E, Ringstrom E, Posner M, Kelsey KT, 2002, Patterns of gene promoter methylation in squamous cell cancer of the head and neck. Oncogene 21(27):4231– 4236. , DOI 10.1038/sj.onc.1205528 Chang HW, Ling GS, Wei WI, Yuen AP, 2004, Smoking and drinking can induce p15 methylation in the upper aerodigestive tract of healthy individuals and patients with head and neck squamous cell carcinoma. Cancer 101(1):125–132. , DOI 10. 1002/cncr.20323 Ishida E, Nakamura M, Ikuta M, Shimada K, Matsuyoshi S, Kirita T, Konishi N, 2005, Promotor hypermethylation of p14ARF is a key alteration for progression of oral squamous cell carcinoma. Oral Oncol 41(6):614–622. , DOI 10.1016/j.oraloncology.2005. 02.003 Puri SK, Si L, Fan CY, Hanna E, 2005, Aberrant promoter hypermethylation of multiple genes in head and neck squamous cell carcinoma. Am J Otolaryngol 26(1):12–17 Brait M, Ford JG, Papaiahgari S, Garza MA, Lee JI, Loyo M, Maldonado L, Begum S, McCaffrey L, Howerton M, Sidransky D, Emerson MR, Ahmed S, Williams CD, Hoque MO, 2009, Association between lifestyle factors and CpG island methylation in a cancer-free population. Cancer Epidemiol Biomark Prev 18(11):2984–2991. , DOI 10.1158/1055-9965.EPI-08- 1245 Lin RK, Hsieh YS, Lin P, Hsu HS, Chen CY, Tang YA, Lee CF, Wang YC, 2010, The tobacco-specific carcinogen NNK induces DNA methyltransferase 1 accumulation and tumor suppressor gene hypermethylation in mice and lung cancer patients. J Clin Invest 120(2):521–532. , DOI 10.1172/JCI40706 Bonsch D, Lenz B, Fiszer R, Frieling H, Kornhuber J, Bleich S, 2006, Lowered DNA methyltransferase, DNMT-3b, mRNA expression is associated with genomic DNA hypermethylation in patients with chronic alcoholism. J Neural Transm, Vienna, 113(9): 1299–1304. , DOI 10.1007/s00702-005-0413-2 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 33 EP 44 DI 10.1007/s12253-017-0309-0 PG 12 ER PT J AU Czirbesz, K Gorka, E Balatoni, T Panczel, G Melegh, K Kovacs, P Gezsi, A Liszkay, G AF Czirbesz, Kata Gorka, Eszter Balatoni, Timea Panczel, Gitta Melegh, Krisztina Kovacs, Peter Gezsi, Andras Liszkay, Gabriella TI Efficacy of Vemurafenib Treatment in 43 Metastatic Melanoma Patients with BRAF Mutation. Single-Institute Retrospective Analysis, Early Real-Life Survival Data SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Vemurafenib; Melanoma; LDH level; Targeted therapy; Treatment; Survival ID Vemurafenib; Melanoma; LDH level; Targeted therapy; Treatment; Survival AB BRAF inhibitor vemurafenib achieved improved overall survival over chemotherapy and have been approved by the FDA and EMA for the treatment of BRAF-mutated metastatic melanoma. The aim of our retrospective analysis was to determine the efficacy and safety of vemurafenib therapy for BRAF mutated metastatic melanoma and subsequently to prove the clinical benefit for the studied 43 patients, based on real-life data. From November 2012 to October 2015 we have selected 43 BRAF mutated, metastatic melanoma patients, treated with vemurafenib. The median follow-up time was 15.9 months. We evaluated progression free survival (PFS), overall survival (OS) and toxicities. According to the AJCC staging system 70% of the patients had stage M1c metastasis, including 6 with stable brain metastasis. Objective responses were noted in 51.1%, the disease control rate was achieved in 79% of the patients. Complete responses were attained by 5 patients (11.6%). Median PFS was 6.48 (95% CI:4.8–15.0) months, median OS was 11.47 (95% CI:8.08-NA) months. We found significant association between LDH level and OS in univariate (p = 0.000613) and multivariate analysis (p = 0.0168). The most common adverse events (AEs) included follicular hyperkeratosis, rash, arthralgia and photosensitivity. Grade 3 AEs, such as cutaneous squamous-cell carcinoma, QTcB interval prolongation, rash, arthralgia were reported in 7 patients (17%).We had noGrade 4 side effects. Similar to the previously published data our analysis confirms the improved survival with vemurafenib treatment (11.47 months) in patients with BRAF V600 mutation. Vemurafenib therapy was well tolerated, the AE profile was almost consistent with the previously reported data of randomised clinical trials. C1 [Czirbesz, Kata] National Institute of Oncology, Department of Dermatology, Rath Gyorgy utca 7-9, H-1122 Budapest, Hungary. [Gorka, Eszter] National Institute of Oncology, Department of Dermatology, Rath Gyorgy utca 7-9, H-1122 Budapest, Hungary. [Balatoni, Timea] National Institute of Oncology, Department of Dermatology, Rath Gyorgy utca 7-9, H-1122 Budapest, Hungary. [Panczel, Gitta] National Institute of Oncology, Department of Dermatology, Rath Gyorgy utca 7-9, H-1122 Budapest, Hungary. [Melegh, Krisztina] National Institute of Oncology, Department of Dermatology, Rath Gyorgy utca 7-9, H-1122 Budapest, Hungary. [Kovacs, Peter] National Institute of Oncology, Department of Dermatology, Rath Gyorgy utca 7-9, H-1122 Budapest, Hungary. [Gezsi, Andras] Semmelweis University, Department of Genetics, Cell and Immunobiology, Nagyvarad ter 4, H-1089 Budapest, Hungary. [Liszkay, Gabriella] National Institute of Oncology, Department of Dermatology, Rath Gyorgy utca 7-9, H-1122 Budapest, Hungary. RP Czirbesz, K (reprint author), National Institute of Oncology, Department of Dermatology, H-1122 Budapest, Hungary. EM czirbikatu@gmail.com CR Siegel RL, Miller KD, Jemal A, 2015, Cancer statistics. CA Cancer J Clin 65:5–29. , DOI 10.3322/caac.21254 Tsao H, Atkins MB, Sober AJ, 2004, Management of cutaneous melanoma. N Engl J Med 351(10):998–1012 Hodi FS, O'Day SJ, McDermott DF, Weber RW et al, 2010, Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363(8):711–723 Erratum in: N Engl J Med. 363(13):1290 Chapman PB, Hauschild A, Robert C et al, 2011, Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 364:2507–2516 McArthur GA, Chapman PB, Robert C et al, 2014, Safety and efficacy of vemurafenib in BRAF, V600E, and BRAF, V600K, mutation-positive melanoma, BRIM-3): extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol 15(3):323–332 Eisenhauer EA, Therasse P, Bogaerts J et al, 2009, New response evaluation criteria in solid tumours: revised RECIST guideline, version 1.1). Eur J Cancer 45:228–247 Hauschild A et al, 2012, Dabrafenib in BRAF-mutated metastatuc melanoma:a multicentre, open-label, phase 3 randomized controlled trial. Lancet 9839:358–365 Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA et al, 2010, Inhibition of Mutated, Activated BRAF in Metastatic Melanoma. N Engl J Med 363:809–819 Sosman JA, Kevin MD et al, 2012, Survival in BRAF V600- Mutant Advanced Melanoma Treated with Vemurafenib. N Engl J Med 366:707–714 Asic K, 2016, Dominant mechanisms of primary resistance differ from dominat mechanisms of secondary resistance to target therapies. Crit Rev Oncol Hematol 97:178–196 Trunzer K, Pavlick AC, Sosman JA, Ribas A et al, 2013, Pharmacodynamic effects and mechanisms of resistance to vemurafenib in patients with metastatic melanoma. J Clin Oncol 31(14):1767–1774 Rinderknecht JD, Goldinger SM et al, 2013, RASopathic Skin Eruptions during Vemurafenib Therapy. PlosOne 8(10, https://doi. org/10.1371/annotation/d884b668-04f7-4ae6-96a3-a199df5b43be. 2013 March 14 Zimmer L, Livingstone E, Hillen U, Domkes S, Becker A et al, 2012, Panniculitis with arthralgia in patients with melanoma treated with selective BRAF inhibitors and its management. Arch Dermatol 148(3):357–361 Manzano Jose L, Layos L et al, 2016, Resistant mechanisms to BRAF inhibitors in melanoma. Ann Transl Med 4(12):237 Larkin J, Ascierto PA, Dreno B et al, 2014, Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med 371: 1867–1876 Ascierto PA, McArthur et al, 2016, Cobimetinib combined with vemurafenib in advanced BRAF V600 -mutant melanoma, coBRIM): updated efficacy results from a randomised, doubleblind, phase 3 trial. Lancet Oncol 17:1248–1260 Long G et al, 2015, Dabrafenib and trametinib versus dabrafenib and placebo for Val 600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised, controlled trial. Lancet 386: 444–451 Robert C, Karaszewska B, Schachter J et al, 2015, Improved overall survival in melanoma combined dabrafenib and trametinib. N Engl J Med 372:30–39 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 45 EP 50 DI 10.1007/s12253-017-0324-1 PG 6 ER PT J AU Go, H Kang, JM Kim, PJ Lee, JLy Park, YJ Park, JM Ro, YJ Cho, MY AF Go, Heounjeong Kang, Jung Mun Kim, Pil-Jong Lee, Jae-Lyun Park, Y Ji Park, Ja-Min Ro, Y Jae Cho, Mee Yong TI Development of Response Classifier for Vascular Endothelial Growth Factor Receptor (VEGFR)-Tyrosine Kinase Inhibitor (TKI) in Metastatic Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Metastatic renal cell carcinoma; Vascular endothelial growth factor signaling; Tyrosine kinase inhibitors; Response classifier; Machine learning ID Metastatic renal cell carcinoma; Vascular endothelial growth factor signaling; Tyrosine kinase inhibitors; Response classifier; Machine learning AB Vascular endothelial growth factor receptor (VEGFR)-targeted therapy improved the outcome ofmetastatic renal cell carcinoma (mRCC) patients. However, a prediction of the response to VEGFR-tyrosine kinase inhibitor (TKI) remains to be elucidated. We aimed to develop a classifier for VEGFR-TKI responsiveness in mRCC patients. Among 101 mRCC patients, ones with complete response, partial response, or ≥24 weeks stable disease in response to VEGFR-TKI treatment were defined as clinical benefit group, whereas patients with <24 weeks stable disease or progressive disease were classified as clinical non-benefit group. Clinicolaboratoryhistopathological data, 41 gene mutations, 20 protein expression levels and 1733 miRNA expression levels were compared between clinical benefit and non-benefit groups. The classifier was built using support vector machine (SVM). Seventy-three patients were clinical benefit group, and 28 patients were clinical non-benefit group. Significantly different features between the groups were as follows: age, time from diagnosis to TKI initiation, thrombocytosis, tumor size, pT stage, ISUP grade, sarcomatoid change, necrosis, lymph node metastasis and expression of pAKT, PD-L1, PD-L2, FGFR2, pS6, PDGFRβ, HIF-1α, IL-8, CA9 and miR-421 (all, P < 0.05). A classifier including necrosis, sarcomatoid component and HIF-1α was built with 0.87 accuracy using SVM. When the classifier was checked against all patients, the apparent accuracy was 0.875 (95% CI, 0.782–0.938). The classifier can be presented as a simple decision tree for clinical use. We developed a VEGFR-TKI response classifier based on comprehensive inclusion of clinicolaboratory-histopathological, immunohistochemical, mutation and miRNA features that may help to guide appropriate treatment in mRCC patients. C1 [Go, Heounjeong] University of Ulsan College of Medicine, Asan Medical Center, Department of Pathology, 88, Olympic-ro 43-gil, Songpa-gu, 05505 Seoul, South Korea. [Kang, Jung Mun] University of Ulsan College of Medicine, Asan Medical Center, Department of Pathology, 88, Olympic-ro 43-gil, Songpa-gu, 05505 Seoul, South Korea. [Kim, Pil-Jong] Seoul National University College of Dental Medicine, Biomedical Knowledge Engineering Laboratory, 101 Daehakno, Jongni-gu, 03080 Seoul, South Korea. [Lee, Jae-Lyun] University of Ulsan College of Medicine, Asan Medical Center, Department of Oncology, 88, Olympic-ro 43-gil, Songpa-gu, 05505 Seoul, South Korea. [Park, Y Ji] Catholic University of Daegu, School of Medicine, Department of Pathology, 33, Duryugongwon-ro 17-gil, Nam-gu, 42472 Daegu, South Korea. [Park, Ja-Min] University of Ulsan College of Medicine, Asan Medical Center, Department of Pathology, 88, Olympic-ro 43-gil, Songpa-gu, 05505 Seoul, South Korea. [Ro, Y Jae] Cornell University, The Methodist Hospital and Weill Medical College, Department of Pathology and Genomic Medicine, 6565 Fannin St, 77030 Houston, TX, USA. [Cho, Mee Yong] University of Ulsan College of Medicine, Asan Medical Center, Department of Pathology, 88, Olympic-ro 43-gil, Songpa-gu, 05505 Seoul, South Korea. RP Cho, MY (reprint author), University of Ulsan College of Medicine, Asan Medical Center, Department of Pathology, 05505 Seoul, South Korea. EM yongcho@amc.seoul.kr CR Srinivasan R, Ricketts CJ, Sourbier C, Linehan WM, 2015, New strategies in renal cell carcinoma: targeting the genetic and metabolic basis of disease. Clinical cancer research : an official journal of the American Association for Cancer Research 21(1):10–17. , DOI 10.1158/1078-0432.CCR-13-2993 Rini BI, 2007, Vascular endothelial growth factor-targeted therapy in renal cell carcinoma: current status and future directions. Clinical cancer research : an official journal of the American Association for Cancer Research 13(4):1098–1106. , DOI 10.1158/1078- 0432.ccr-06-1989 Escudier B, Eisen T, StadlerWM, Szczylik C, Oudard S, Siebels M, Negrier S, Chevreau C, Solska E, Desai AA, Rolland F, Demkow T, Hutson TE, GoreM, Freeman S, Schwartz B, ShanM, Simantov R, Bukowski RM, 2007, Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 356(2):125–134. , DOI 10.1056/ NEJMoa060655 Maroto P, Rini B, 2014, Molecular biomarkers in advanced renal cell carcinoma. Clinical cancer research : an official journal of the American Association for Cancer Research 20(8):2060–2071. , DOI 10.1158/1078-0432.CCR-13-1351 Park JY, Lee JL, Baek S, Eo SH, Go H, Ro JY, Cho YM, 2014, Sarcomatoid features, necrosis, and grade are prognostic factors in metastatic clear cell renal cell carcinoma with vascular endothelial growth factor-targeted therapy. Hum Pathol. , DOI 10. 1016/j.humpath.2014.02.019 Sun M, Shariat SF, Cheng C, Ficarra V, Murai M, Oudard S, Pantuck AJ, Zigeuner R, Karakiewicz PI, 2011, Prognostic factors and predictive models in renal cell carcinoma: a contemporary review. Eur Urol 60(4):644–661. , DOI 10.1016/j.eururo. 2011.06.041 Funakoshi T, Lee CH, Hsieh JJ, 2014, A systematic review of predictive and prognostic biomarkers for VEGF-targeted therapy in renal cell carcinoma. Cancer Treat Rev 40(4):533–547. https:// doi.org/10.1016/j.ctrv.2013.11.008 Gamez-Pozo A, Anton-Aparicio LM, Bayona C, Borrega P, Gallegos Sancho MI, Garcia-Dominguez R, de Portugal T, Ramos-Vazquez M, Perez-Carrion R, Bolos MV, Madero R, Sanchez-Navarro I, Fresno Vara JA, Espinosa Arranz E, 2012, MicroRNA expression profiling of peripheral blood samples predicts resistance to first-line sunitinib in advanced renal cell carcinoma patients. Neoplasia 14(12):1144–1152 Garcia-Donas J, Leandro-Garcia LJ, Gonzalez Del Alba A, Morente M, Alemany I, Esteban E, Arranz JA, Climent MA, Gallardo E, Castellano DE, Bellmunt J, Mellado B, Puente J, Moreno F, Font A, Hernando S, Robledo M, Rodriguez-Antona C, 2013, Prospective study assessing hypoxia-related proteins as markers for the outcome of treatment with sunitinib in advanced clear-cell renal cell carcinoma. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO 24(9): 2409–2414. , DOI 10.1093/annonc/mdt219 Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, MooneyM, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J, 2009, New response evaluation criteria in solid tumours: revised RECIST guideline, version 1.1). Eur J Cancer 45(2):228–247. https://doi. org/10.1016/j.ejca.2008.10.026 Di Leo A, Jerusalem G, Petruzelka L, Torres R, Bondarenko IN, Khasanov R, Verhoeven D, Pedrini JL, Smirnova I, LichinitserMR, Pendergrass K, Garnett S, Lindemann JP, Sapunar F, Martin M, 2010, Results of the CONFIRM phase III trial comparing fulvestrant 250 mg with fulvestrant 500 mg in postmenopausal women with estrogen receptor-positive advanced breast cancer. Journal of clinical oncology : official Journal of the American Society of Clinical Oncology DDDD, 30):4594–4600., DOI https:// doi.org/10.1200/jco.2010.28.8415 Moch H, Humphrey PA, Ulbright TM, Reuter VE, 2016, WHO Classification Tumours of the Urinary system and Male Genital Organs, 4th edn. IARC Press, Lyon Delahunt B, Cheville JC, Martignoni G, Humphrey PA, Magi- Galluzzi C, McKenney J, Egevad L, Algaba F, Moch H, Grignon DJ, Montironi R, Srigley JR, 2013, The International Society of Urological Pathology, ISUP, grading system for renal cell carcinoma and other prognostic parameters. Am J Surg Pathol 37(10): 1490–1504. , DOI 10.1097/PAS.0b013e318299f0fb Mekhail TM, Abou-Jawde RM, Boumerhi G, Malhi S, Wood L, Elson P, Bukowski R, 2005, Validation and extension of the Memorial Sloan-Kettering prognostic factors model for survival in patients with previously untreated metastatic renal cell carcinoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 23(4):832–841. , DOI 10. 1200/jco.2005.05.179 Golshayan AR, George S, Heng DY, Elson P, Wood LS, Mekhail TM, Garcia JA, Aydin H, Zhou M, Bukowski RM, Rini BI, 2009, Metastatic sarcomatoid renal cell carcinoma treated with vascular endothelial growth factor-targeted therapy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 27(2):235–241. , DOI 10.1200/jco.2008.18.0000 Molina AM, Tickoo SK, Ishill N, Trinos MJ, Schwartz LH, Patil S, Feldman DR, Reuter VE, Russo P, Motzer RJ, 2011, Sarcomatoidvariant renal cell carcinoma: treatment outcome and survival in advanced disease. Am J Clin Oncol 34(5):454–459. https://doi. org/10.1097/COC.0b013e3181f47aa4 Dornbusch J, Zacharis A, Meinhardt M, Erdmann K, Wolff I, Froehner M, Wirth MP, Zastrow S, Fuessel S, 2013, Analyses of potential predictive markers and survival data for a response to sunitinib in patients with metastatic renal cell carcinoma. PLoS One 8(9):e76386. , DOI 10.1371/journal.pone.0076386 Saez MI, Trigo Perez JM, Perez-Rivas LG, Perez-Villa L, Villatoro R,Montesa A, Cid JI, LeonM, de Luque V, Fernandez C, Quero C, Pajares B, Alba E, 2012, Hypoxia-inducible factor, HIF, 1α and 2α as predictive markers of outcome to VEGFR tyrosine kinase inhibitors, TKI, in renal cell carcinoma, RCC). J Clin Oncol 30(suppl; abstr 4630):309S Krishnan VG, Westhead DR, 2003, A comparative study of machine-learningmethods to predict the effects of single nucleotide polymorphisms on protein function. Bioinformatics, Oxford, England, 19(17):2199–2209 Cruz JA, Wishart DS, 2006, Applications of machine learning in cancer prediction and prognosis. Cancer Informat 2:59–77 Brugarolas J, 2014, Molecular genetics of clear-cell renal cell carcinoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 32(18):1968–1976. , DOI 10.1200/jco.2012.45.2003 Rini BI, Jaeger E, Weinberg V, Sein N, Chew K, Fong K, Simko J, Small EJ, Waldman FM, 2006, Clinical response to therapy targeted at vascular endothelial growth factor in metastatic renal cell carcinoma: impact of patient characteristics and Von Hippel- Lindau gene status. BJU Int 98(4):756–762. , DOI 10. 1111/j.1464-410X.2006.06376.x Sato Y, Yoshizato T, Shiraishi Y, Maekawa S, Okuno Y, Kamura T, Shimamura T, Sato-Otsubo A, Nagae G, Suzuki H, Nagata Y, Yoshida K, Kon A, Suzuki Y, Chiba K, Tanaka H, Niida A, Fujimoto A, Tsunoda T, Morikawa T, Maeda D, Kume H, Sugano S, Fukayama M, Aburatani H, Sanada M, Miyano S, Homma Y, Ogawa S, 2013, Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet 45(8):860–867. , DOI 10.1038/ ng.2699 Cowey CL, Rathmell WK, 2009, VHL gene mutations in renal cell carcinoma: role as a biomarker of disease outcome and drug efficacy. Curr Oncol Rep 11(2):94–101 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 51 EP 58 DI 10.1007/s12253-017-0323-2 PG 8 ER PT J AU Aaberg-Jessen, Ch Fogh, L Dahl Sorensen, M Halle, B Brunner, N Winther Kristensen, B AF Aaberg-Jessen, Charlotte Fogh, Louise Dahl Sorensen, Mia Halle, Bo Brunner, Nils Winther Kristensen, Bjarne TI Overexpression of TIMP-1 and Sensitivity to Topoisomerase Inhibitors in Glioblastoma Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma; TIMP-1; TOP inhibitors; Chemosensitivity ID Glioblastoma; TIMP-1; TOP inhibitors; Chemosensitivity AB The multifunctional protein - tissue inhibitor of metalloproteinases-1 (TIMP-1) - has been associated with a poor prognosis in several types of cancers including glioblastomas. In addition, TIMP-1 has been associated with decreased response to chemotherapy, and especially the efficacy of the family of topoisomerase (TOP) inhibitors has been related to TIMP-1. As a second line treatment of glioblastomas, the vascular endothelial growth factor (VEGF) antibody bevacizumab is administered in combination with the TOP1 inhibitor irinotecan and glioblastoma cell levels of TIMP-1 could therefore potentially influence the efficacy of such treatment. In the present study, we aimed to investigate whether a high TIMP-1 expression in glioblastoma cell lines would affect the sensitivity to TOP inhibitors, and whether TIMP-1 overexpressing cells would have alterered growth and invasion. We established TIMP-1 overexpressing subclones from two human glioblastoma cell lines. TIMP-1 overexpressing U87MG cells were significantly more resistant than low TIMP-1 expressing clones and parental cells when exposed to SN-38 (TOP1 inhibitor) or epirubicin (TOP2 inhibitor). No significant differences were observed for the TIMP-1 transfected A172 cells. Implantation of both U87MG and A172 spheroids into organotypic brain slice cultures revealed a reduced growth of TIMP-1 overexpressing U87MG spheroids, however, no significant differences in invasion were observed. The present study suggests that TIMP-1 overexpression reduces the effect of TOP inhibitors in glioblastoma. TIMP-1 also appeared to reduce spheroid growth, but did not influence invasion. Whether TIMP-1 plays a role in irinotecan resistance and has a predictive potential in glioblastoma patients remains to be elucidated. C1 [Aaberg-Jessen, Charlotte] Odense University Hospital, Department of Pathology, J.B. Winsloew vej 15, DK-5000 Odense, Denmark. [Fogh, Louise] University of Copenhagen, Faculty of Health and Medical Sciences, Institute for Drug Design and Pharmacology, Strandboulevarden 49, DK-2100 Copenhagen, Denmark. [Dahl Sorensen, Mia] Odense University Hospital, Department of Pathology, J.B. Winsloew vej 15, DK-5000 Odense, Denmark. [Halle, Bo] Odense University Hospital, Department of Pathology, J.B. Winsloew vej 15, DK-5000 Odense, Denmark. [Brunner, Nils] University of Copenhagen, Faculty of Health and Medical Sciences, Institute for Drug Design and Pharmacology, Strandboulevarden 49, DK-2100 Copenhagen, Denmark. [Winther Kristensen, Bjarne] Odense University Hospital, Department of Pathology, J.B. Winsloew vej 15, DK-5000 Odense, Denmark. RP Winther Kristensen, B (reprint author), Odense University Hospital, Department of Pathology, DK-5000 Odense, Denmark. EM bjarne.winther.kristensen@rsyd.dk CR Louis DNW, CaveneeWK(2007)WHO classification of tumors of the central nervous system, 4 edn. International Agency for Research on Cancer, IARC), Lyon Agnihotri S, Gajadhar AS, Ternamian C, Gorlia T, Diefes KL, Mischel PS, Kelly J, McGown G, Thorncroft M, Carlson BL, Sarkaria JN, Margison GP, Aldape K, Hawkins C, Hegi M, Guha A, 2012, Alkylpurine-DNA-N-glycosylase confers resistance to temozolomide in xenograft models of glioblastoma multiforme and is associated with poor survival in patients. J Clin Invest 122(1):253–266. , DOI 10.1172/JCI59334 Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN, 2006, Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444(7120):756–760. , DOI 10.1038/ nature05236 DeanM(2009, ABC transporters, drug resistance, and cancer stem cells. J Mammary Gland Biol Neoplasia 14(1):3–9. , DOI 10.1007/s10911-009-9109-9 Dean M, Fojo T, Bates S, 2005, Tumour stem cells and drug resistance. Nat Rev Cancer 5(4):275–284. , DOI 10.1038/ nrc1590 Esteller M, Garcia-Foncillas J, Andion E, Goodman SN, Hidalgo OF, Vanaclocha V, Baylin SB, Herman JG, 2000, Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl JMed 343(19):1350–1354. https://doi. org/10.1056/nejm200011093431901 Nakai E, Park K, Yawata T, Chihara T, Kumazawa A, Nakabayashi H, Shimizu K, 2009, Enhanced MDR1 expression and chemoresistance of cancer stem cells derived from glioblastoma. Cancer Investig 27(9):901–908. , DOI 10.3109/ 07357900801946679 Salmaggi A, Boiardi A, Gelati M, Russo A, Calatozzolo C, Ciusani E, Sciacca FL, Ottolina A, Parati EA, La Porta C, Alessandri G, Marras C, Croci D, De Rossi M, 2006, Glioblastoma-derived tumorospheres identify a population of tumor stem-like cells with angiogenic potential and enhanced multidrug resistance phenotype. Glia 54(8):850–860. , DOI 10.1002/glia.20414 Chirco R, Liu XW, Jung KK, Kim HR, 2006, Novel functions of TIMPs in cell signaling. Cancer Metastasis Rev 25(1):99–113. , DOI 10.1007/s10555-006-7893-x Egeblad M, Werb Z, 2002, New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2(3):161–174. , DOI 10.1038/nrc745 Gomez DE, Alonso DF, Yoshiji H, Thorgeirsson UP, 1997, Tissue inhibitors of metalloproteinases: structure, regulation and biological functions. Eur J Cell Biol 74(2):111–122 Jiang Y, Goldberg ID, Shi YE, 2002, Complex roles of tissue inhibitors of metalloproteinases in cancer. Oncogene 21(14):2245– 2252. , DOI 10.1038/sj.onc.1205291 Bloomston M, Shafii A, Zervos E, Rosemurgy AS, 2005, TIMP-1 antisense gene transfection attenuates the invasive potential of pancreatic cancer cells in vitro and inhibits tumor growth in vivo. Am J Surg 189(6):675–679. , DOI 10.1016/j.amjsurg.2005.03.008 Guedez L, McMarlin AJ, Kingma DW, Bennett TA, Stetler- Stevenson M, Stetler-Stevenson WG, 2001, Tissue inhibitor of metalloproteinase-1 alters the tumorigenicity of Burkitt's lymphoma via divergent effects on tumor growth and angiogenesis. Am J Pathol 158(4):1207–1215. , DOI 10.1016/s0002-9440(10, 64070-9 Jung KK, Liu XW, Chirco R, Fridman R, Kim HR, 2006, Identification of CD63 as a tissue inhibitor of metalloproteinase-1 interacting cell surface protein. EMBO J 25(17):3934–3942. , DOI 10.1038/sj.emboj.7601281 Lambert E, Dasse E, Haye B, Petitfrere E, 2004, TIMPs as multifacial proteins. Crit Rev Oncol Hematol 49(3):187–198. , DOI 10.1016/j.critrevonc.2003.09.008 Stetler-Stevenson WG, 2008, Tissue inhibitors of metalloproteinases in cell signaling: metalloproteinase-independent biological activities. Sci Signal 1(27):re6. , DOI 10.1126/scisignal. 127re6 Yoshiji H, Harris SR, Raso E, Gomez DE, Lindsay CK, Shibuya M, Sinha CC, Thorgeirsson UP, 1998, Mammary carcinoma cells over-expressing tissue inhibitor of metalloproteinases-1 show enhanced vascular endothelial growth factor expression. International journal of cancer Journal international du cancer 75(1):81–87 Liu XW, Bernardo MM, Fridman R, Kim HR, 2003, Tissue inhibitor of metalloproteinase-1 protects human breast epithelial cells against intrinsic apoptotic cell death via the focal adhesion kinase/ phosphatidylinositol 3-kinase andMAPK signaling pathway. J Biol Chem 278(41):40364–40372. , DOI 10.1074/jbc. M302999200 Birgisson H, Nielsen HJ, Christensen IJ, Glimelius B, Brunner N, 2010, Preoperative plasma TIMP-1 is an independent prognostic indicator in patients with primary colorectal cancer: a prospective validation study. Eur J Cancer 46(18):3323–3331. , DOI 10.1016/j.ejca.2010.06.009 Holten-Andersen MN, Stephens RW, Nielsen HJ, Murphy G, Christensen IJ, Stetler-Stevenson W, Brunner N, 2000, High preoperative plasma tissue inhibitor of metalloproteinase-1 levels are associated with short survival of patients with colorectal cancer. Clin Cancer Res : an official journal of the American Association for Cancer Research 6(11):4292–4299 Kuvaja P, Wurtz SO, Talvensaari-Mattila A, Brunner N, Paakko P, Turpeenniemi-Hujanen T, 2007, High serum TIMP-1 correlates with poor prognosis in breast carcinoma - a validation study. Cancer Biomark : section A of Disease markers 3(6):293–300 Pesta M, Kulda V, Kucera R, Pesek M, Vrzalova J, Liska V, Pecen L, Treska V, Safranek J, Prazakova M, Vycital O, Bruha J, Holubec L, Topolcan O, 2011, Prognostic significance of TIMP-1 in nonsmall cell lung cancer. Anticancer Res 31(11):4031–4038 Rauvala M, Puistola U, Turpeenniemi-Hujanen T, 2005, Gelatinases and their tissue inhibitors in ovarian tumors; TIMP-1 is a predictive as well as a prognostic factor. Gynecol Oncol 99(3): 656–663. , DOI 10.1016/j.ygyno.2005.07.009 Schrohl AS, Holten-Andersen MN, Peters HA, Look MP, Meijervan Gelder ME, Klijn JG, Brunner N, Foekens JA, 2004, Tumor tissue levels of tissue inhibitor of metalloproteinase-1 as a prognostic marker in primary breast cancer. Clin Cancer Res : an official journal of the American Association for Cancer Research 10(7): 2289–2298 Wang CS, Wu TL, Tsao KC, Sun CF, 2006, Serum TIMP-1 in gastric cancer patients: a potential prognostic biomarker. Ann Clin Lab Sci 36(1):23–30 Aaberg-Jessen C, Christensen K, Offenberg H, Bartels A, Dreehsen T, Hansen S, Schroder HD, Brunner N, KristensenBW(2009, Low expression of tissue inhibitor of metalloproteinases-1, TIMP-1, in glioblastoma predicts longer patient survival. J Neuro-Oncol 95(1): 117–128. , DOI 10.1007/s11060-009-9910-8 Ejlertsen B, Jensen MB, Nielsen KV, Balslev E, Rasmussen BB, Willemoe GL, Hertel PB, Knoop AS, Mouridsen HT, Brunner N, 2010, HER2, TOP2A, and TIMP-1 and responsiveness to adjuvant anthracycline-containing chemotherapy in high-risk breast cancer patients. J Clin Oncol : official journal of the American Society of Clinical Oncology 28(6):984–990. , DOI 10.1200/jco. 2009.24.1166 Frederiksen C, Qvortrup C, Christensen IJ, Glimelius B, Berglund A, Jensen BV, Nielsen SE, Keldsen N, Nielsen HJ, Brunner N, Pfeiffer P, 2011, Plasma TIMP-1 levels and treatment outcome in patients treated with XELOX for metastatic colorectal cancer. Ann Oncol : official journal of the European Society for Medical Oncology / ESMO 22(2):369–375. , DOI 10.1093/ annonc/mdq354 Klintman M, Ornbjerg Wurtz S, Christensen IJ, Braemer Hertel P, Ferno M, Malmberg M, Mouridsen H, Cold F, Schrohl AS, Foekens JA, Malmstrom P, Brunner N, 2010, Association between tumor tissue TIMP-1 levels and objective response to first-line chemotherapy in metastatic breast cancer. Breast Cancer Res Treat 121(2):365–371. , DOI 10.1007/s10549-009-0483-1 Schrohl AS, Meijer-van Gelder ME, Holten-Andersen MN, Christensen IJ, Look MP, Mouridsen HT, Brunner N, Foekens JA, 2006, Primary tumor levels of tissue inhibitor of metalloproteinases- 1 are predictive of resistance to chemotherapy in patients with metastatic breast cancer. Clin Cancer Res : an official journal of the American Association for Cancer Research 12(23):7054–7058. , DOI 10.1158/1078-0432.ccr-06-0950 Sorensen NM, Bystrom P, Christensen IJ, Berglund A, Nielsen HJ, Brunner N, Glimelius B, 2007, TIMP-1 is significantly associated with objective response and survival in metastatic colorectal cancer patients receiving combination of irinotecan, 5-fluorouracil, and folinic acid. Clin Cancer Res : an official journal of the American Association for Cancer Research 13(14):4117–4122. https://doi. org/10.1158/1078-0432.ccr-07-0186 Gruppe DNO, 2012, Retningslinier for behandling af intrakranielle gliomer hos voksne. http://www.dnog.dk/assets/files/ Retningslinier%20PDF/DNOG%20gliom%20retningslinje% 202012.pdf Champoux JJ, 2000, Structure-based analysis of the effects of camptothecin on the activities of human topoisomerase I. Ann N YAcad Sci 922:56–64 Hsiang YH, Liu LF, 1988, Identification ofmammalian DNAtopoisomerase I as an intracellular target of the anticancer drug camptothecin. Cancer Res 48(7):1722–1726 Deng X, Fogh L, Lademann U, Jensen V, Stenvang J, Yang H, Brunner N, Schrohl AS, 2013, TIMP-1 overexpression does not affect sensitivity to HER2-targeting drugs in the HER2-geneamplified SK-BR-3 human breast cancer cell line. Tumour Biol : the journal of the International Society for Oncodevelopmental Biology and Medicine 34(2):1161–1170. , DOI 10.1007/ s13277-013-0659-5 Holten-Andersen MN, Christensen IJ, Nielsen HJ, Lilja H, Murphy G, Jensen V, Brunner N, Piironen T, 2002, Measurement of the noncomplexed free fraction of tissue inhibitor of metalloproteinases 1 in plasma by immunoassay. Clin Chem 48(8):1305–1313 Holten-Andersen MN, Murphy G, Nielsen HJ, Pedersen AN, Christensen IJ, Hoyer-Hansen G, Brunner N, Stephens RW, 1999, Quantitation of TIMP-1 in plasma of healthy blood donors and patients with advanced cancer. Br J Cancer 80(3–4):495–503. , DOI 10.1038/sj.bjc.6690384 Aaberg-Jessen C, Norregaard A, Christensen K, Pedersen CB, Andersen C, Kristensen BW, 2013, Invasion of primary gliomaand cell line-derived spheroids implanted into corticostriatal slice cultures. Int J Clin Exp Pathol 6(4):546–560 Hekmat O, Munk S, Fogh L, Yadav R, Francavilla C, Horn H, Wurtz SO, Schrohl AS, Damsgaard B, Romer MU, Belling KC, Jensen NF, Gromova I, Bekker-Jensen DB,Moreira JM, Jensen LJ, Gupta R, Lademann U, Brunner N, Olsen JV, Stenvang J, 2013, TIMP-1 increases expression and phosphorylation of proteins associated with drug resistance in breast cancer cells. J Proteome Res. , DOI 10.1021/pr400457u Fu ZY, Lv JH, Ma CY, Yang DP,Wang T, 2011, Tissue inhibitor of metalloproteinase-1 decreased chemosensitivity of MDA-435 breast cancer cells to chemotherapeutic drugs through the PI3K/AKT/NF-small ka, Cyril l icB pathway. Biomed Pharmacother = Biomedecine & pharmacotherapie 65(3):163– 167. , DOI 10.1016/j.biopha.2011.02.004 Wang T, Lv JH, Zhang XF, Li CJ, Han X, Sun YJ, 2010, Tissue inhibitor of metalloproteinase-1 protects MCF-7 breast cancer cells from paclitaxel-induced apoptosis by decreasing the stability of cyclin B1. Int J Cancer: Journal international du cancer 126(2): 362–370. , DOI 10.1002/ijc.24753 Willemoe GL, Hertel PB, Bartels A, Jensen MB, Balslev E, Rasmussen BB, Mouridsen H, Ejlertsen B, Brunner N, 2009, Lack of TIMP-1 tumour cell immunoreactivity predicts effect of adjuvant anthracycline-based chemotherapy in patients, n=647, with primary breast cancer. A Danish breast cancer cooperative group study. Eur J Cancer 45(14):2528–2536. , DOI 10. 1016/j.ejca.2009.05.029 Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, Vescovi A, 2004, Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res 64(19):7011–7021. https://doi. org/10.1158/0008-5472.can-04-1364 Lee J, Kotliarova S, Kotliarov Y, Li A, Su Q, Donin NM, Pastorino S, Purow BW, Christopher N, Zhang W, Park JK, Fine HA, 2006, Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 9(5):391– 403. , DOI 10.1016/j.ccr.2006.03.030 Yu SC, Ping YF, Yi L, Zhou ZH, Chen JH, Yao XH, Gao L, Wang JM, Bian XW, 2008, Isolation and characterization of cancer stem cells from a human glioblastoma cell line U87. Cancer Lett 265(1): 124–134. , DOI 10.1016/j.canlet.2008.02.010 Nakano A, Tani E, Miyazaki K, Yamamoto Y, Furuyama J, 1995, Matrix metalloproteinases and tissue inhibitors of metalloproteinases in human gliomas. J Neurosurg 83(2):298–307. , DOI 10.3171/jns.1995.83.2.0298 Matsuzawa K, Fukuyama K, Hubbard SL, Dirks PB, Rutka JT, 1996, Transfection of an invasive human astrocytoma cell line with a TIMP-1 cDNA: modulation of astrocytoma invasive potential. J Neuropathol Exp Neurol 55(1):88–96 Bigelow RL, Williams BJ, Carroll JL, Daves LK, Cardelli JA, 2009, TIMP-1 overexpression promotes tumorigenesis of MDAMB- 231 breast cancer cells and alters expression of a subset of cancer promoting genes in vivo distinct from those observed in vitro. Breast Cancer Res Treat 117(1):31–44. , DOI 10. 1007/s10549-008-0170-7 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 59 EP 69 DI 10.1007/s12253-017-0312-5 PG 11 ER PT J AU Wang, Y Yan, Z Lu, Q Wang, Y Sun, X Zhang, Sh AF Wang, Yuandong Yan, Zhang Lu, Qian Wang, Yiming Sun, Xinchen Zhang, Shu TI NRG-1 Stimulates Serum DJ-1 Increase in Breast Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Serum DJ-1; HER3; NRG-1; Breast cancers; MCF-7 cells ID Serum DJ-1; HER3; NRG-1; Breast cancers; MCF-7 cells AB To explore the relationship between the expression of DJ-1/HER3 and tumor grade in breast cancer, and investigate the effect of HER3 on NRG-1-mediated serum DJ-1 level in vivo.We analyze the expression level of DJ-1 and HER3 in 68 patients with different grades of breast cancer by immunostaining the tissue microarray. Besides, we investigated the serum DJ-1 level by ELISA. We found that the detectable DJ-1 protein expression is decreased, and the HER3 expression is increased in tumor tissue with the progression of breast cancer. There is a significant rise of DJ-1 in serumin vivo with the stimulation of NRG-1. Meanwhile, we found that HER3 knockdown abolishes NRG-1-induced serum DJ-1 increase and HER3 overexpress improves NRG-1-induced serum DJ- 1 increase. This study provides a serum biomarker for breast cancer. The results showed that DJ-1 was associated with clinical stage of breast cancer, and NRG-1 increased the dissociation of HER3 and DJ-1, with promoting the level of DJ-1 in peripheral blood. It is suggested that the level of DJ-1 in peripheral blood may be conducive to assess the prognosis of patients with breast cancer and serum DJ-1 levels can serve as an indicator of therapeutic effectiveness for the development of HER3 targeting breast cancer antibody therapies. C1 [Wang, Yuandong] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, Jiangsu, China. [Yan, Zhang] Huazhong University of Science and Technology, Tongji Medical College, Union Hospital, Department of Anesthesiology, 430022 Wuhan, Hubei, China. [Lu, Qian] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, Jiangsu, China. [Wang, Yiming] University of New South Wales, School of Biotechnology and Biomolecular Sciences, Kensington, 2052 Sydney, NSW, Australia. [Sun, Xinchen] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, Jiangsu, China. [Zhang, Shu] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 300 Guangzhou Road, 210029 Nanjing, Jiangsu, China. RP Zhang, Sh (reprint author), The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210029 Nanjing, China. EM zhangshu@njmu.edu.cn CR Allard L, Burkhard PR, Lescuyer P, Burgess JA, Walter N, Hochstrasser DF, Sanchez JC, 2005, PARK7 and nucleoside diphosphate kinase A as plasma markers for the early diagnosis of stroke. Clin Chem 51:2043–2051. , DOI 10.1373/clinchem.2005.053942 Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, Krieger E, Dekker MC, Squitieri F, Ibanez P, Joosse M, van Dongen JW, Vanacore N, van Swieten JC, Brice A, Meco G, van Duijn CM, Oostra BA, Heutink P, 2003, Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 299:256–259. , DOI 10.1126/science.1077209 Brenton JD, Carey LA, Ahmed AA, Caldas C, 2005, Molecular classification and molecular forecasting of breast cancer: ready for clinical application? J Clin Oncol 23: 7350–7360. , DOI 10.1200/JCO.2005.03.3845 Choi BK, Cai X, Yuan B, Huang Z, Fan X, Deng H, Zhang N, An Z, 2012, HER3 intracellular domains play a crucial role in HER3/HER2 dimerization and activation of downstream signaling pathways. Protein Cell 3:781–789. , DOI 10.1007/s13238-012-2065-y Chung C, Lee S, Hwang S, Park E, 2013, Systematic review of exercise effects on health outcomes in women with breast cancer. Asian Nurs Res, Korean Soc Nurs Sci, 7:149–159. , DOI 10.1016/j.anr.2013.07.005 Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, Kosaka T, Holmes AJ, Rogers AM, Cappuzzo F, Mok T, Lee C, Johnson BE, Cantley LC, Janne PA, 2007, MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316:1039–1043. , DOI 10.1126/science.1141478 Honbou K, Suzuki NN, Horiuchi M, Niki T, Taira T, Ariga H, Inagaki F, 2003, The crystal structure of DJ-1, a protein related to male fertility and Parkinson's disease. J Biol Chem 278:31380– 31384. , DOI 10.1074/jbc.M305878200 Karagianni M, Kaitelidou D, Kalokairinou A, Mantas J, 2014, Breast cancer in social media: a literature review. Stud Health Technol Inform 202:321 Le Naour F, Misek DE, Krause MC, Deneux L, Giordano TJ, Scholl S, Hanash SM, 2001, Proteomics-based identification of RS/DJ-1 as a novel circulating tumor antigen in breast cancer. Clin Cancer Res 7:3328–3335 Mahar I, Tan S, Davoli MA, Dominguez-Lopez S, Qiang C, Rachalski A, Turecki G, Mechawar N, 2011, Subchronic peripheral neuregulin-1 increases ventral hippocampal neurogenesis and induces antidepressant-like effects. PLoS One 6:e26610. , DOI 10.1371/journal.pone.0026610 Maita C, Tsuji S, Yabe I, Hamada S, Ogata A, Maita H, Iguchi- Ariga SM, Sasaki H, Ariga H, 2008, Secretion of DJ-1 into the serum of patients with Parkinson's disease. Neurosci Lett 431: 86–89. , DOI 10.1016/j.neulet.2007.11.027 Meetze K, Vincent S, Tyler S, Mazsa EK, Delpero AR, Bottega S, McIntosh D, Nicoletti R, Winston WM, Weiler S, Feng B, Gyuris J,Weng Z, 2015, Neuregulin 1 expression is a predictive biomarker for response to AV-203, an ERBB3 inhibitory antibody, in human tumor models. Clin Cancer Res 21:1106–1114. , DOI 10.1158/1078-0432.CCR-14-2407 Mujoo K, Choi BK, Huang Z, Zhang N, An Z, 2014, Regulation of ERBB3/HER3 signaling in cancer. Oncotarget 5:10222–10236. , DOI 10.18632/oncotarget.2655 Nagakubo D, Taira T, Kitaura H, Ikeda M, Tamai K, Iguchi-Ariga SM, Ariga H, 1997, DJ-1, a novel oncogene which transforms mouse NIH3T3 cells in cooperation with ras. Biochem Biophys Res Commun 231:509–513. , DOI 10.1006/bbrc.1997.6132 Oda M, Makita M, Iwaya K, Akiyama F, Kohno N, Tsuchiya B, Iwase T, Matsubara O, 2012, High levels of DJ-1 protein in nipple fluid of patients with breast cancer. Cancer Sci 103:1172–1176. , DOI 10.1111/j.1349-7006.2012.02267.x Pardo M, Garcia A, Thomas B, Pineiro A, Akoulitchev A, Dwek RA, Zitzmann N, 2006, The characterization of the invasion phenotype of uveal melanoma tumour cells shows the presence of MUC18 and HMG-1 metastasis markers and leads to the identification of DJ-1 as a potential serum biomarker. Int J Cancer 119: 1014–1022. , DOI 10.1002/ijc.21942 Sergina NV, Rausch M, Wang D, Blair J, Hann B, Shokat KM, Moasser MM, 2007, Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature 445:437–441. , DOI 10.1038/nature05474 Shames DS, Carbon J,Walter K, Jubb AM, Kozlowski C, Januario T, Do A, Fu L, Xiao Y, Raja R, Jiang B, Malekafzali A, Stern H, Settleman J, Wilson TR, Hampton GM, Yauch RL, Pirzkall A, Amler LC, 2013, High heregulin expression is associated with activated HER3 and may define an actionable biomarker in patients with squamous cell carcinomas of the head and neck. PLoS One 8: e56765. , DOI 10.1371/journal.pone.0056765 Sobin LH, Compton CC, 2010, TNM seventh edition: what's new, what's changed: communication from the International Union Against Cancer and the American Joint Committee on Cancer. Cancer-Am Cancer Soc 116: 5336–5339. , DOI 10.1002/cncr.25537 Soltoff SP, Carraway KR, Prigent SA, Gullick WG, Cantley LC, 1994, ErbB3 is involved in activation of phosphatidylinositol 3- kinase by epidermal growth factor. Mol Cell Biol 14:3550–3558 Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H, Pesich R, Geisler S, Demeter J, Perou CM, Lonning PE, Brown PO, Borresen-Dale AL, Botstein D, 2003, Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A 100:8418–8423. , DOI 10.1073/pnas.0932692100 Sotiriou C, Pusztai L, 2009, Gene-expression signatures in breast cancer. N Engl J Med 360:790–800. , DOI 10. 1056/NEJMra0801289 Tsuboi Y, Munemoto H, Ishikawa S, Matsumoto K, Iguchi-Ariga SM, Ariga H, 2008, DJ-1, a causative gene product of a familial form of Parkinson's disease, is secreted through microdomains. FEBS Lett 582:2643–2649. , DOI 10.1016/j.febslet.2008.06.043 Tsuchiya B, Iwaya K, Kohno N, Kawate T, Akahoshi T, Matsubara O, Mukai K, 2012, Clinical significance of DJ-1 as a secretory molecule: retrospective study of DJ-1 expression at mRNA and protein levels in ductal carcinoma of the breast. Histopathology 61:69–77. , DOI 10.1111/j.1365-2559.2012.04202.x Zhang N, Chang Y, Rios A, An Z, 2016, HER3/ErbB3, an emerging cancer therapeutic target. Acta Biochim Biophys Sin Shanghai 48:39–48. , DOI 10.1093/abbs/gmv103 Zhang S, Mukherjee S, Fan X, Salameh A, Mujoo K, Huang Z, Li L, To'A SG, Zhang N, An Z, 2016, Novel association of DJ-1 with HER3 potentiates HER3 activation and signaling in cancer. Oncotarget 7:65758–65769. , DOI 10.18632/oncotarget.11613 Zhou W, Zhu M, Wilson MA, Petsko GA, Fink AL, 2006, The oxidation state of DJ-1 regulates its chaperone activity toward alpha-synuclein. J Mol Biol 356:1036–1048. , DOI 10.1016/j.jmb.2005.12.030 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 71 EP 79 DI 10.1007/s12253-017-0326-z PG 9 ER PT J AU Davidson, TK Zhu, Z Bai, Q Xiao, H Wakefield, RM Fang, Y AF Davidson, T Kristoffer Zhu, Ziwen Bai, Qian Xiao, Huaping Wakefield, R Mark Fang, Yujiang TI Blueberry as a Potential Radiosensitizer for Treating Cervical Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Blueberry extract; Radiation; Apoptosis; Cervical cancer ID Blueberry extract; Radiation; Apoptosis; Cervical cancer AB Cervical cancer (CC) is a leading cause of death in women worldwide. Radiation therapy (RT) for CC is an effective alternative, but its toxicity remains challenging. Blueberry is amongst the most commonly consumed berries in the United States.We previously showed that resveratrol, a compound in red grapes, can be used as a radiosensitizer for prostate cancer. In this study, we found that the percentage of colonies, PCNA expression level and the OD value of cells from the CC cell line SiHa were all decreased in RT/Blueberry Extract (BE) group when compared to those in the RT alone group. Furthermore, TUNEL+ cells and the relative caspase-3 activity in the CC cells were increased in the RT/BE group compared to those in the RT alone group. The antiproliferative effect of RT/BE on cancer cells correlated with downregulation of pro-proliferative molecules cyclin D and cyclin E. The pro-apoptotic effect of RT/BE correlated with upregulation of the pro-apoptotic molecule TRAIL. Thus, BE sensitizes SiHa cells to RT by inhibition of proliferation and promotion of apoptosis, suggesting that blueberry might be used as a potential radiosensitizer to treat CC. C1 [Davidson, T Kristoffer] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. [Zhu, Ziwen] University of Missouri, School of Medicine, Department of SurgeryColumbia, MO, USA. [Bai, Qian] University of Missouri, School of Medicine, Department of SurgeryColumbia, MO, USA. [Xiao, Huaping] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. [Wakefield, R Mark] University of Missouri, School of Medicine, Department of SurgeryColumbia, MO, USA. [Fang, Yujiang] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. RP Fang, Y (reprint author), Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, USA. EM yujiang.fang@dmu.edu CR Siegel RL, Miller KD, Jemal A, 2015, Cancer statistics, 2015. CA Cancer J Clin 65(1):5–29 Nandakumar V, Singh T, Katiyar SK, 2008, Multi-targeted prevention and therapy of cancer by proanthocyanidins. Cancer Lett 269(2):378–387 Diaconeasa Z, Leopold L, Rugina D, Ayvaz H, Socaciu C, 2015, Antiproliferative and antioxidant properties of anthocyanin rich extracts from blueberry and blackcurrant juice. Int J Mol Sci 16(2): 2352–2365 Faria A, Pestana D, Teixeira D, De Freitas V, Mateus N, Calhau C, 2010, Blueberry anthocyanins and pyruvic acid adducts: anticancer properties in breast cancer cell lines. Phytother Res 24(12): 1862–1869 Jeyabalan J, Aqil F, Munagala R, Annamalai L, Vadhanam MV, Gupta RC, 2014, Chemopreventive and therapeutic activity of dietary blueberry against estrogen-mediated breast cancer. J Agric Food Chem 62(18):3963–3971 Kanaya N, Adams L, Takasaki A, Chen S, 2014, Whole blueberry powder inhibits metastasis of triple negative breast cancer in a xenograft mouse model through modulation of inflammatory cytokines. Nutr Cancer 66(2):242–248 Kausar H, Jeyabalan J, Aqil F, Chabba D, Sidana J, Singh IP, Gupta RC, 2012, Berry anthocyanidins synergistically suppress growth and invasive potential of human non-small-cell lung cancer cells. Cancer Lett 325(1):54–62 Matchett MD, MacKinnon SL, Sweeney MI, Gottschall-Pass KT, Hurta RA, 2006, Inhibition of matrix metalloproteinase activity in DU145 human prostate cancer cells by flavonoids from lowbush blueberry, Vaccinium angustifolium): possible roles for protein kinase C and mitogen-activated protein-kinase-mediated events. J Nutr Biochem 17(2):117–125 Montales MTE, Rahal OM, Kang J, Rogers TJ, Prior RL, Wu X, Simmen RC, 2012, Repression of mammosphere formation of human breast cancer cells by soy isoflavone genistein and blueberry polyphenolic acids suggests diet-mediated targeting of cancer stemlike/ progenitor cells. Carcinogenesis 33(3):652–660 Schmidt BM, Erdman JW, Lila MA, 2006, Differential effects of blueberry proanthocyanidins on androgen sensitive and insensitive human prostate cancer cell lines. Cancer Lett 231(2):240–246 Suh N, Paul S, Hao X, Simi B, Xiao H, Rimando AM, Reddy BS, 2007, Pterostilbene, an active constituent of blueberries, suppresses aberrant crypt foci formation in the azoxymethaneinduced colon carcinogenesis model in rats. Clin Cancer Res 13(1):350–355 Wedge DE, Meepagala KM, Magee JB, Smith SH, Huang G, Larcom LL, 2001, Anticarcinogenic activity of strawberry, blueberry, and raspberry extracts to breast and cervical cancer cells. J Med Food 4(1):49–51 Yi W, Fischer J, Krewer G, Akoh CC, 2005, Phenolic compounds from blueberries can inhibit colon cancer cell proliferation and induce apoptosis. J Agric Food Chem 53(18):7320–7329 Liu RH, 2013, Health-promoting components of fruits and vegetables in the diet. Adv Nutr: An International Review Journal 4(3): 384S–392S Routray W, Orsat V, 2011, Blueberries and their anthocyanins: factors affecting biosynthesis and properties. Compr Rev Food Sci Food Saf 10(6):303–320 Aqil F, Munagala R, Kausar H, Jeyabalan J, Gupta R, 2013, Enhanced activity of chemotherapeutic drugs by blueberry anthocyanidins and withaferin A against human lung cancer cells. Cancer Res 73(8 Supplement):3678–3678 Fang Y, DeMarco VG, Nicholl MB, 2012, Resveratrol enhances radiation sensitivity in prostate cancer by inhibiting cell proliferation and promoting cell senescence and apoptosis. Cancer Sci 103(6):1090–1098 Bingul I, Basaran-Kucukgergin C, Tekkesin MS, Olgac V, Dogru- Abbasoglu S, Uysal M, 2013, Effect of blueberry pretreatment on diethylnitrosamine-induced oxidative stress and liver injury in rats. Environ Toxicol Pharmacol 36(2):529–538 Bunea A, Rugina D, Sconta Z, Pop RM, Pintea A, Socaciu C, Tabaran F, Grootaert C, Struijs K, VanCamp J, 2013, Anthocyanin determination in blueberry extracts from various cultivars and their antiproliferative and apoptotic properties in B16- F10 metastatic murine melanoma cells. Phytochemistry 95:436– 444 Gordillo G, FangH, Khanna S, Harper J, Phillips G, Sen CK, 2009, Oral administration of blueberry inhibits angiogenic tumor growth and enhances survival of mice with endothelial cell neoplasm. Antioxid Redox Signal 11(1):47–58 Kai H, Akamatsu E, Torii E, Kodama H, Yukizaki C, Sakakibara Y, Suiko M,Morishita K, Kataoka H, Matsuno K, 2011, Inhibition of proliferation by agricultural plant extracts in seven human adult Tcell leukaemia, ATL)-related cell lines. J Nat Med 65(3–4):651– 655 Minker C, Duban L, Karas D, Jarvinen P, Lobstein A, Muller CD, 2015, Impact of Procyanidins from Different Berries on Caspase 8 Activation in Colon Cancer. Oxidative Med Cell Longev 2015:13. , DOI 10.1155/2015/154164 Qi C, Li S, Jia Y, Wang L, 2014, Blueberry anthocyanins induce G2/M cell cycle arrest and apoptosis of oral cancer KB cells through down-regulation methylation of p53. Yi chuan= Hereditas/Zhongguo yi chuan xue hui bian ji 36(6): 566–573 Ravoori S, Vadhanam MV, Aqil F, Gupta RC, 2012, Inhibition of estrogen-mediated mammary tumorigenesis by blueberry and black raspberry. J Agric Food Chem 60(22):5547–5555 Lohachoompol V, Srzednicki G, Craske J, 2004, The change of total anthocyanins in blueberries and their antioxidant effect after drying and freezing. Biomed Res Int 2004(5):248–252 Fang Y, Sharp GC, Braley-Mullen H, 2008, Interleukin-10 promotes resolution of granulomatous experimental autoimmune thyroiditis. Am J Pathol 172(6):1591–1602 Fang Y, Yu S, Braley-Mullen H, 2012, TGF-β promotes proliferation of thyroid epithelial cells in IFN-γ−/− mice by downregulation of p21 and p27 via AKT pathway. Am J Pathol 180(2): 650–660 Fang Y, Bradley MJ, Cook KM, Herrick EJ, Nicholl MB, 2013, A potential role for resveratrol as a radiation sensitizer for melanoma treatment. J Surg Res 183(2):645–653 Fang Y, Herrick EJ, Nicholl MB, 2012, A Possible Role for Perforin and Granzyme B in Resveratrol-Enhanced Radiosensitivity of Prostate Cancer. J Androl 33(4):752–760 Fang Y, Moore BJ, Bai Q, Cook KM, Herrick EJ, Nicholl MB, 2013, Hydrogen peroxide enhances radiation-induced apoptosis and inhibition of melanoma cell proliferation. Anticancer Res 33(5):1799–1807 Zhu Z, Davidson KT, Brittingham A, Wakefield MR, Bai Q, Xiao H, Fang Y, 2016, Trichomonas vaginalis: a possible foe to prostate cancer. Med Oncol 33(10):115 Fang Y, Sharp GC, Yagita H, Braley-Mullen H, 2008, A critical role for TRAIL in resolution of granulomatous experimental autoimmune thyroiditis. J Pathol 216(4):505–513 Fang Y, Wei Y, DeMarco V, Chen K, Sharp GC, Braley-Mullen H, 2007, Murine FLIP transgene expressed on thyroid epithelial cells promotes resolution of granulomatous experimental autoimmune thyroiditis in DBA/1 mice. Am J Pathol 170(3):875–887 Haie-Meder C, Morice P, Castiglione M, Group EGW, 2009, Cervical cancer: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol 20(suppl_4):iv27–iv28 Dasari S, Tchounwou PB, 2014, Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol 740:364–378 Tippayamontri T, Kotb R, Paquette B, Sanche L, 2012, Synergism in concomitant chemoradiotherapy of cisplatin and oxaliplatin and their liposomal formulation in the human colorectal cancerHCT116 model. Anticancer Res 32(10):4395–4404 Leisching G, Loos B, Botha M, Engelbrecht A-M, 2015, Bcl-2 confers survival in cisplatin treated cervical cancer cells: circumventing cisplatin dose-dependent toxicity and resistance. J Transl Med 13(1):1 Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M, Kroemer G, 2012, Molecular mechanisms of cisplatin resistance. Oncogene 31(15):1869–1883 Huang H, Huang SY, Chen TT, Chen JC, Chiou CL, Huang TM, 2004, Cisplatin restores p53 function and enhances the radiosensitivity in HPV16 E6 containing SiHa cells. J Cell Biochem 91(4): 756–765 Liu Y, Xing H, Han X, Shi X, Liang F, Chen G, Ma D, 2005, The mechanism of cisplatin-induced apoptosis in HeLa cells. Chin J Clin Oncol 2(6):866–869 Bloom J, Cross FR, 2007, Multiple levels of cyclin specificity in cell-cycle control. Nat Rev Mol Cell Biol 8(2):149–160 Resnitzky D, Reed SI, 1995, Different roles for cyclins D1 and E in regulation of the G1-to-S transition. Mol Cell Biol 15(7):3463– 3469 Zhang D, Li X, Chen C, Li Y, Zhao L, Jing Y, Liu W, Wang X, Zhang Y, Xia H, 2012, Attenuation of p38-mediated miR-1/133 expression facilitates myoblast proliferation during the early stage of muscle regeneration. PLoS One 7(7):e41478 Kelman Z, 1997, PCNA: structure, functions and interactions. Oncogene 14(6):629–640 Nicholl MB, Ledgewood CL, Chen X, Bai Q, Qin C, Cook KM, Herrick EJ, Diaz-Arias A,Moore BJ, FangY, 2014, Il-35 promotes pancreas cancer growth through enhancement of proliferation and inhibition of apoptosis: Evidence for a role as an autocrine growth factor. Cytokine 70(2):126–133 Zhu Z, Zhang D, Lee H, Menon AA, Wu J, Hu K, Jin Y, 2017, Macrophage-derived apoptotic bodies promote the proliferation of the recipient cells via shuttling microRNA-221/222. J Leukoc Biol 101(6):1349–1359 LeBlanc H, Ashkenazi A, 2003, Apo2L/TRAIL and its death and decoy receptors. Cell Death Differ 10(1):66–75 Fang Y, Chen X, Bai Q, Qin C, Mohamud AO, Zhu Z, Ball TW, Ruth CM, Newcomer DR, Herrick EJ, 2015, IL-9 inhibits HTB-72 melanoma cell growth through upregulation of p21 and TRAIL. J Surg Oncol 111(8):969–974 Nicholl MB, Chen X, Qin C, Bai Q, Zhu Z, Davis MR, Fang Y, 2016, IL-32α has differential effects on proliferation and apoptosis of human melanoma cell lines. J Surg Oncol 113(4):364–369 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 81 EP 88 DI 10.1007/s12253-017-0319-y PG 8 ER PT J AU Sun, Ch Ding, Y Wang, Sh Hu, W AF Sun, Cheng Ding, Yangyang Wang, Shimin Hu, Wei TI Long Non-Coding RNA SPRY4-IT1 Can Predict Poor Prognosis in Digestive System Malignancies: a Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lncRNA; SPRY4-IT1; Prognosis; Metastasis; Meta-analysis ID lncRNA; SPRY4-IT1; Prognosis; Metastasis; Meta-analysis AB Recent studies have reported that long non-coding RNA SPRY4 intron transcript 1 (SPRY4-IT1) is abnormally expressed in malignant digestive tumors and associated with prognosis. But its clinical relevance is unclear. Here, we performed a meta-analysis aims to evaluate the prognostic value of SPRY4-IT1 in digestive system malignancies. We systematic search the PubMed, Web of Science, ScienceDirect and Wiley Online Library database to eligible studies. The pooled hazard ratios (HRs) with a 95% confidence interval (95% CI) were calculated to explored the association of lncRNA SPRY4-IT1 with prognosis. Five studies were eligible for analysis, a total of 518 patients were included. Meta-analysis indicated that overexpression of SPRY4-IT1 was associated with poor over survival (OS) (HR = 1.24, 95% CI:0.49–1.98; random-effects model). The clinicopathological parameters analysis further showed that increased expression level of SPRY4-IT1 was positively correlated with lymph node metastasis (HR = 1.45, 95% CI =0.88–2.02; fixed-effects model), TNM stage (HR = 1.24,95% CI = 0.78–1.70; fixed-effects model), and invasion depth (HR = 1.25,95% CI = 0.63–1.88; fixed-effects model). lncRNA SPRY4-IT1 may serve as a potential prognostic marker in malignant digestive tumors. C1 [Sun, Cheng] The Second Hospital of Anhui Medical University, Department of PharmacologyHefei, Anhui, China. [Ding, Yangyang] The Second Hospital of Anhui Medical University, Department of Hematology / Hematological LabHefei, Anhui, China. [Wang, Shimin] The Second Hospital of Anhui Medical University, Department of PharmacologyHefei, Anhui, China. [Hu, Wei] The Second Hospital of Anhui Medical University, Department of PharmacologyHefei, Anhui, China. RP Hu, W (reprint author), The Second Hospital of Anhui Medical University, Department of Pharmacology, Hefei, China. EM scadyy@163.com CR Garzon R, Volinia S, Papaioannou D et al, 2014, Expression and prognostic impact of lncRNAs in acute myeloid leukemia. Proc Natl Acad Sci U S A 111:18679–18684 Kim SH, Lee HW, Go SI, Lee SI, Lee GW, 2016, Clinical significance of the preoperative platelet count and platelet-to-lymphocyte ratio, PLT-PLR, in patients with surgically resected non-small cell lung cancer. Oncotarget 7:36198–36206 Chan JC, Chan DL, Diakos CI et al, 2016, The lymphocyte-tomonocyte ratio is a superior predictor of overall survival in comparison to established biomarkers of resectable colorectal cancer. Ann Surg 265(3):539–546 Han Li C, Chen Y, 2015, Small and long non-coding RNAs: novel targets in perspective cancer therapy. Curr Genomics 16:319–326 Zarate R, Boni V, Bandres E, Garcia-Foncillas J, 2012, MiRNAs and LincRNAs: could they be considered as biomarkers in colorectal cancer? Int J Mol Sci 13:840–865 Yang Q, Zhang RW, Sui PC, He HT, Ding L, 2015, Dysregulation of non-coding RNAs in gastric cancer. World J Gastroenterol 21: 10956–10981 Sugihara H, Ishimoto T, Miyake K et al, 2015, Noncoding RNA expression aberration is associated with cancer progression and is a potential biomarker in esophageal squamous cell carcinoma. Int J Mol Sci 16:27824–27834 Wong DT, 2015, Salivary extracellular noncoding RNA: emerging biomarkers for molecular diagnostics. Clin Ther 37:540–551 Cao D, Ding Q, Yu W, Gao M, Wang Y, 2016, Long noncoding RNA SPRY4-IT1 promotes malignant development of colorectal cancer by targeting epithelial-mesenchymal transition. Onco Targets Ther 9:5417–5425 Tan W, Song ZZ, Xu Q et al, 2017, Up-regulated expression of spry4-it1 predicts poor prognosis in colorectal cancer. Med Sci Monit 23:309–314 PengW,Wu G, Fan H,Wu J, Feng J, 2015, Long noncoding RNA SPRY4-IT1 predicts poor patient prognosis and promotes tumorigenesis in gastric cancer. Tumour Bio l36:6751–6758 Xie M, F-q N, Sun M et al, 2015, Decreased long noncoding RNA SPRY4-IT1 contributing to gastric cancer cell metastasis partly via affecting epithelial-mesenchymal transition. J Transl Med 13:250 Xie H-W, Wu Q-Q, Zhu B et al, 2014, Long noncoding RNA SPRY4-IT1 is upregulated in esophageal squamous cell carcinoma and associated with poor prognosis. Tumor Biol 35:7743–7754 Ferlay J, Soerjomataram I, Dikshit R et al, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:E359–E386 George J, Patel T, 2015, Noncoding RNA as therapeutic targets for hepatocellular carcinoma. Semin Liver Dis 35:63–74 Jin K, Luo G, Xiao Z et al, 2015, Noncoding RNAs as potential biomarkers to predict the outcome in pancreatic cancer. Drug Des Dev Ther 9:1247–1255 Khaitan D, Dinger ME, Mazar J et al, 2011, The melanomaupregulated long noncodingRNASPRY4-IT1 modulates apoptosis and invasion. Cancer Res 71:3852–3862 Sun M, Liu XH, Lu KH et al, 2014, EZH2-mediated epigenetic suppression of long noncoding RNA SPRY4-IT1 promotes NSCLC cell proliferation and metastasis by affecting the epithelial-mesenchymal transition. Cell Death Dis 5:e1298 Shi Y, Li J, Liu Y et al, 2015, The long noncoding RNA SPRY4- IT1 increases the proliferation of human breast cancer cells by upregulating ZNF703 expression. Mol Cancer 14:51 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 89 EP 95 DI 10.1007/s12253-017-0327-y PG 7 ER PT J AU Nagy, BZs Bartak, KB Kalmar, A Galamb, O Wichmann, B Dank, M Igaz, P Tulassay, Zs Molnar, B AF Nagy, Brigitta Zsofia Bartak, Kinga Barbara Kalmar, Alexandra Galamb, Orsolya Wichmann, Barnabas Dank, Magdolna Igaz, Peter Tulassay, Zsolt Molnar, Bela TI Comparison of Circulating miRNAs Expression Alterations in Matched Tissue and Plasma Samples During Colorectal Cancer Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microRNA; Plasma; Circulating microRNA; Colorectal cancer; Colorectal adenoma; Microarray; Real-time PCR; Tissue ID microRNA; Plasma; Circulating microRNA; Colorectal cancer; Colorectal adenoma; Microarray; Real-time PCR; Tissue AB MicroRNAs (miRNAs) have been found to play a critical role in colorectal adenoma-carcinoma sequence. MiRNA-specific high-throughput arrays became available to detect promising miRNA expression alterations even in biological fluids, such as plasma samples, where miRNAs are stable. The purpose of this study was to identify circulating miRNAs showing altered expression between normal colonic (N), tubular adenoma (ADT), tubulovillous adenoma (ADTV) and colorectal cancer (CRC) matched plasma and tissue samples. Sixteen peripheral plasma and matched tissue biopsy samples (N n = 4; ADT n = 4; ADTV n = 4; CRC n = 4) were selected, and total RNA including miRNA fraction was isolated. MiRNAs from plasma samples were extracted using QIAamp Circulating Nucleic Acid Kit (Qiagen). Matched tissue-plasma miRNA microarray experiments were conducted by GeneChip® miRNA 3.0 Array (Affymetrix). RT-qPCR (microRNA Ready-to-use PCR Human Panel I + II; Exiqon) was used for validation. Characteristic miRNA expression alterations were observed in comparison of AD and CRC groups (miR-149*, miR-3196, miR-4687) in plasma samples. In the N vs. CRC comparison, significant overexpression of miR-612, miR-1296, miR-933, miR-937 and miR-1207 was detected by RT-PCR (p < 0.05). Similar expression pattern of these miRNAs were observed using microarray in tissue pairs, as well. Although miRNAs were also found in circulatory system in a lower concentration compared to tissues, expression patterns slightly overlapped between tissue and plasma samples. Detected circulating miRNA alterations may originate not only from the primer tumor but from other cell types including immune cells. C1 [Nagy, Brigitta Zsofia] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Bartak, Kinga Barbara] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Kalmar, Alexandra] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Galamb, Orsolya] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Wichmann, Barnabas] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Dank, Magdolna] Semmelweis University, Department of OncologyBudapest, Hungary. [Igaz, Peter] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. RP Nagy, BZs (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM nagyzsofiab@gmail.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90. https://doi. org/10.3322/caac.20107 Patai AV, Valcz G, Hollosi P, Kalmar A, Peterfia B, Patai A, Wichmann B, Spisak S, Bartak BK, Leiszter K, Toth K, Sipos F, Kovalszky I, Peter Z, Miheller P, Tulassay Z, Molnar B, 2015, Comprehensive DNA methylation analysis reveals a common ten-gene methylation signature in colorectal adenomas and carcinomas. PLoSOne 10(8):e0133836. , DOI 10.1371/journal. pone.0133836 Yan L, Zhao W, Yu H, Wang Y, Liu Y, Xie C, 2016, A comprehensive meta-analysis of MicroRNAs for predicting colorectal cancer. Medicine 95(9):e2738. , DOI 10.1097/MD. 0000000000002738 Esquela-Kerscher A, Slack FJ, 2006, Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer 6(4):259–269. , DOI 10. 1038/nrc1840 Nagy ZB, Wichmann B, Kalmar A, Galamb O, Bartak BK, Spisak S, Tulassay Z, Molnar B, 2017, Colorectal adenoma and carcinoma specific miRNA profiles in biopsy and their expression in plasma specimens. Clin Epigenetics 9:22. , DOI 10.1186/s13148- 016-0305-3 Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O'Briant KC, Allen A, Lin DW, Urban N, Drescher CW, Knudsen BS, Stirewalt DL, Gentleman R, Vessella RL, Nelson PS, Martin DB, Tewari M, 2008, Circulating microRNAs as stable blood-based markers for cancer detection. P Natl Acad Sci USA 105(30): 10513–10518. , DOI 10.1073/pnas.0804549105 Yuan T, Huang X,Woodcock M, DuM, Dittmar R,Wang Y, Tsai S, KohliM, Boardman L, Patel T,Wang L, 2016, Plasma extracellular RNA profiles in healthy and cancer patients. Sci Rep 6:19413. , DOI 10.1038/srep19413 Tufekci KU, Oner MG, Genc S, Genc K, 2010, MicroRNAs and multiple sclerosis. Autoimmune Dis 2011:807426. , DOI 10.4061/2011/807426 Ge Q, Shen Y, Tian F, Lu J, Bai Y, Lu Z, 2015, Profiling circulating microRNAs in maternal serum and plasma. Mol Med Rep 12(3): 3323–3330. , DOI 10.3892/mmr.2015.3879 Leidinger P, Galata V, Backes C, Stahler C, Rheinheimer S, Huwer H, Meese E, Keller A, 2015, Longitudinal study on circulating miRNAs in patients after lung cancer resection. Oncotarget 6(18): 16674–16685. 10.18632/oncotarget.4322 Ma R, Jiang T, Kang X, 2012, Circulating microRNAs in cancer: origin, function and application. J Exp Clin Cancer Res 31:38. , DOI 10.1186/1756-9966-31-38 Pathak S, Meng WJ, Nandy SK, Ping J, Bisgin A, Helmfors L, Waldmann P, Sun XF, 2015, Radiation and SN38 treatments modulate the expression of microRNAs, cytokines and chemokines in colon cancer cells in a p53-directed manner. Oncotarget 6(42): 44758–44780. , DOI 10.18632/oncotarget.5815 Tambyah PA, Sepramaniam S, Mohamed Ali J, Chai SC, Swaminathan P, Armugam A, Jeyaseelan K, 2013, microRNAs in circulation are altered in response to influenza a virus infection in humans. PLoS One 8(10):e76811. , DOI 10.1371/ journal.pone.0076811 Zhou N, Zhou Y, Tang Y, Yu W, 2016, MiR-519 inhibits gastric cancer cell activity through regulation of HuR expression. Journal of Central South University Medical Sciences 41(1):19–23. https:// doi.org/10.11817/j.issn.1672-7347.2016.01.003 Sheng L, He P, Yang X, Zhou M, Feng Q, 2015, miR-612 negatively regulates colorectal cancer growth and metastasis by targeting AKT2. Cell Death Dis 6:e1808. , DOI 10.1038/ cddis.2015.184 Bhajun R, Guyon L, Pitaval A, Sulpice E, Combe S, Obeid P, Haguet V, Ghorbel I, Lajaunie C, Gidrol X, 2015, A statistically inferredmicroRNA network identifies breast cancer targetmiR-940 as an actin cytoskeleton regulator. Sci Rep 5:8336. , DOI 10.1038/Srep08336 Bobowicz M, Skrzypski M, Czapiewski P, Marczyk M, Maciejewska A, Jankowski M, Szulgo-Paczkowska A, Zegarski W, Pawlowski R, Polanska J, Biernat W, Jaskiewicz J, Jassem J, 2016, Prognostic value of 5-microRNA based signature in T2- T3N0 colon cancer. Clin Exp Metastasis 33(8):765–773. https:// doi.org/10.1007/s10585-016-9810-1 Cakmak HA, Coskunpinar E, Ikitimur B, Barman HA, Karadag B, Tiryakioglu NO, Kahraman K, Vural VA, 2015, The prognostic value of circulating microRNAs in heart failure: preliminary results from a genome-wide expression study. J Cardiovasc Med 16(6): 431–437. , DOI 10.2459/JCM.0000000000000233 Shan X, Wen W, Zhu D, Yan T, Cheng W, Huang Z, Zhang L, Zhang H, Wang T, Zhu W, Zhu Y, Zhu J, 2017, miR 1296-5p inhibits the migration and invasion of gastric cancer cells by repressing ERBB2 expression. PLoS One 12(1):e0170298. https:// doi.org/10.1371/journal.pone.0170298 Wei WJ, Wang YL, Li DS, Wang Y, Wang XF, Zhu YX, Pan XD, Wang ZY, Wu Y, Jin L, Wang JC, Ji QH, 2015, Association study of single nucleotide polymorphisms in mature microRNAs and the risk of thyroid tumor in a Chinese population. Endocrine 49(2): 436–444. , DOI 10.1007/s12020-014-0467-8 Dong G, Zhang RF, Xu JJ, Guo YF, 2015, Association between microRNA polymorphisms and papillary thyroid cancer susceptibility. Int J Clin Exp Pathol 8(10):13450–13457 Poliseno L, Haimovic A, Segura MF, Hanniford D, Christos PJ, Darvishian F, Wang J, Shapiro RL, Pavlick AC, Berman RS, Hernando E, Zavadil J, Osman I, 2012, Histology-specific microRNA alterations in melanoma. J Invest Dermatol 132(7): 1860–1868. , DOI 10.1038/jid.2011.451 Zhang L, Zeng D, Chen Y, Li N, Lv Y, Li Y, Xu X, Xu G, 2016, miR-937 contributes to the lung cancer cell proliferation by targeting INPP4B. Life Sci 155:110–115. , DOI 10.1016/ j.lfs.2016.05.014 Li Y, Liang M, Zhang Z, 2014, Regression analysis of combined gene expression regulation in acute myeloid leukemia. PLoS Comput Biol 10(10):e1003908. , DOI 10.1371/journal. pcbi.1003908 SlatteryML, Herrick JS, Mullany LE, Valeri N, Stevens J, Caan BJ, Samowitz W, Wolff RK, 2015, An evaluation and replication of miRNAs with disease stage and colorectal cancer-specific mortality. Int J Cancer 137(2):428–438. , DOI 10.1002/ijc.29384 Wang W, Sun J, Li F, Li R, Gu Y, Liu C, Yang P, Zhu M, Chen L, TianW, Zhou H, Mao Y, Zhang L, Jiang J,Wu C, Hua D, ChenW, Lu B, Ju J, Zhang X, 2012, A frequent somatic mutation in CD274 3′-UTR leads to protein over-expression in gastric cancer by disrupting miR-570 binding. Hum Mutat 33(3):480–484. https:// doi.org/10.1002/humu.22014 Barsotti AM, Beckerman R, Laptenko O, Huppi K, Caplen NJ, Prives C, 2012, p53-dependent induction of PVT1 and miR- 1204. J Biol Chem 287(4):2509–2519. , DOI 10.1074/ jbc.M111.322875 Ali SheikhMS, Xia K, Li F, Deng X, Salma U, Deng H,WeiWei L, Yang TL, Peng J, 2015, Circulating miR-765 and miR-149: potential noninvasive diagnostic biomarkers for geriatric coronary artery disease patients. Biomed Res Int 2015:740301. , DOI 10. 1155/2015/740301 Chen YX, Gelfond JAL, McManus LM, Shireman PK, 2009, Reproducibility of quantitative RT-PCR array in miRNA expression profiling and comparison with microarray analysis. BMC Genomics 10:407. , DOI 10.1186/1471-2164-10-407 Wang ZS, Zhong M, Bian YH, Mu YF, Qin SL, Yu MH, Qin J, 2016, MicroRNA-187 inhibits tumor growth and invasion by directly targeting CD276 in colorectal cancer. Oncotarget 7(28): 44266–44276. 10.18632/oncotarget.10023 Tsang WP, Ng EK, Ng SS, Jin H, Yu J, Sung JJ, Kwok TT, 2010, Oncofetal H19-derived miR-675 regulates tumor suppressor RB in human colorectal cancer. Carcinogenesis 31(3):350–358. https:// doi.org/10.1093/carcin/bgp181 Song Q, Song J,Wang Q,Ma Y, Sun N,Ma J, Chen Q, Xia G, Huo Y, Yang L, Li B, 2016, miR-548d-3p/TP53BP2 axis regulates the proliferation and apoptosis of breast cancer cells. Cancer Med 5(2): 315–324. , DOI 10.1002/cam4.567 Larrea E, Sole C, Manterola L, Goicoechea I, Armesto M, Arestin M, Caffarel MM, Araujo AM, Araiz M, Fernandez-Mercado M, Lawrie CH, 2016, New concepts in cancer biomarkers: circulating miRNAs in liquid biopsies. Int J Mol Sci 17(5):627. , DOI 10.3390/ijms17050627 Ji X, Takahashi R, Hiura Y, Hirokawa G, Fukushima Y, Iwai N, 2009, Plasma miR-208 as a biomarker of myocardial injury. Clin Chem 55(11):1944–1949. , DOI 10.1373/ clinchem.2009.125310 Igaz I, Igaz P, 2015, Why is microRNA action tissue specific? A putative defense mechanism against growth disorders, tumor development or progression mediated by circulating microRNA? Med Hypotheses 85(5):530–533. , DOI 10.1016/j.mehy. 2015.07.013 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 97 EP 105 DI 10.1007/s12253-017-0308-1 PG 9 ER PT J AU Ozgoz, A Ozturk, HK Yukselturk, A Samli, H Baskan, Z Icduygu, MF Bacaksiz, M AF Ozgoz, Asuman Ozturk, Hekimler Kuyas Yukselturk, Aysegul Samli, Hale Baskan, Zuhal Icduygu, Mutlu Fadime Bacaksiz, Mehmet TI Genetic Variations of DNA Repair Genes in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; DNA damage; DNA repair; Polymorphism ID Breast cancer; DNA damage; DNA repair; Polymorphism AB Genetic variations in DNA repair genes may affect DNA repair capacity therefore increase risk for cancer. In our study, we evaluted the relation between DNA repair gene polymorphisms XRCC1 rs1799782, rs25487, rs25489; XPC rs2228000, rs2228001; XPD rs1799793, rs13181; XRCC3 rs861539; RAD51B rs10483813, rs1314913 and breast cancer risk for 202 Turkish cases in total, in which 102 patients with breast cancer and 100 controls. Genotyping of the DNA samples was carried out by multiplex PCR andmatrix-assisted laser desorption/ionization mass spectrometry with time of flight measurement (MALDI-TOF) using Sequenom MassARRAY 4 analyzer. Genotype and allele distributions were calculated between the groups. Odds ratios (ORs) and 95% confidence intervals (CIs) were reported. rs25487 AA genotype and A allele was found to be increased in the control group (respectively, OR 0.16 95% CI 0.02–1.06, p = 0.058; OR 1.55, 95% CI 1.01–2.36, p = 0.043) and rs861539 T allele was found to be decreased in the patient group (OR 1.53, 95% CI 1.01–2.30, p = 0.049). No association with breast cancer was found for the remaining SNPs. Our findings suggest that XRCC1 rs25487 AA genotype and A allele, XRCC3 rs861539 Tallelemay have protective effects in breast cancer for Turkish population. C1 [Ozgoz, Asuman] Kastamonu University, Kastamonu School of Medicine, Department of Medical Genetics, Kuzeykent Mah, Orgeneral Atilla Ates Pasa Cd. No: 19 C, 37200 Kastamonu, Turkey. [Ozturk, Hekimler Kuyas] Suleyman Demirel University, School of Medicine, Department of Medical GeneticsIsparta, Turkey. [Yukselturk, Aysegul] Kastamonu University, Fazil Boyner Faculty of Health Sciences, Department of Nutrition and DieteticsKastamonu, Turkey. [Samli, Hale] Uludag University, School of Veterinary Medicine, Department of GeneticsBursa, Turkey. [Baskan, Zuhal] Acibadem University, School of Medicine, Department of Medical OncologyIstanbul, Turkey. [Icduygu, Mutlu Fadime] Giresun University, School of Medicine, Department of Medical GeneticsGiresun, Turkey. [Bacaksiz, Mehmet] Alanya Ciplakli Family Health CenterAlanya-Antalya, Turkey. RP Ozgoz, A (reprint author), Kastamonu University, Kastamonu School of Medicine, Department of Medical Genetics, 37200 Kastamonu, Turkey. EM biologistasu@gmail.com CR Deligezer U, Dalay N, 2004, Association of the XRCC1 gene polymorphisms with cancer risk in Turkish breast cancer patients. Exp Mol Med 36(6):572–575 Patrono C, Sterpone S, Testa A, Cozzi R, 2014, Polymorphisms in base excision repair genes: Breast cancer risk and individual radiosensitivity. World J Clin Oncol 5(5):874–882 Shadrina AS, Ermolenko NA, Boyarskikh UA, Sinkina TV, Lazarev AF, Petrova VD et al, 2016, Polymorphisms in DNA repair genes and breast cancer risk in Russian population: a casecontrol study. Clin Exp Med 16(1):21–28 AlMutairi F, Pathan AA, Alanazi M, Shalaby M, Alabdulkarim HA, Alamri A et al, 2015, Association of DNA repair gene APE1 Asp148Glu polymorphism with breast cancer risk. Dis Markers. , DOI 10.1155/2015/869512 Wood RD, Mitchell M, Lindahl T, 2005, Human DNA repair genes. Mutat Res 577:275–283 Oeth P, del Mistro G, Marnellos G, Shi T, van den Boom D, 2009, Qualitative and quantitative genotyping using single base primer extension coupled with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, MassARRAY). Methods Mol Biol 578:307–343 Goode EL, Ulrich CM, Potter JD, 2002, Polymorphisms in DNA repair genes and associations with cancer risk. Cancer Epidemiol Biomark Prev 11(12):1513–1530 Feng YZ, Liu YL, He XF, Wei W, Shen XL, Xie DL, 2014, Association between the XRCC1 Arg194Trp polymorphism and risk of cancer: evidence from 201 case-control studies. Tumour Biol 35(11):10677–10697 Al Mutairi FM, Alanazi M, Shalaby M, Alabdulkarim HA, Pathan AA, Parine NR, 2013, Association of XRCC1 gene polymorphisms with breast cancer susceptibility in Saudi patiens. Asian Pac J Cancer Prev 14(6):3809–3813 Przybylowska-Sygut K, Stanczyk M, Kusinska R, Kordek R, Majsterek I, 2013, Association of the Arg194Trp and the Arg399Gln polymorphisms of the XRCC1 gene with risk occurrence and the response to adjuvant therapy among Polish women with breast cancer. Clin Breast Cancer 13(1):61–68 Smith TR, Miller MS, Lohman K, Lange EM, Case LD, Mohrenweiser HW et al, 2003, Polymorphisms of XRCC1 and XRCC3 genes and susceptibility to breast cancer. Cancer Let 190(2):183–190 Moullan N, Cox DG, Angele S, Romestaing P, Gerard JP, Hall J, 2003, Polymorphisms in the DNA repair gene XRCC1, breast cancer risk, and response to radiotherapy. Cancer Epidemiol Biomark Prev 12(11 Pt 1):1168–1174 Saadat M, Kohan L, Omidvari S, 2008, Genetic polymorphisms of XRCC1, codon 399, and susceptibility to breast cancer in Iranian women, a case-control study. Breast Cancer Res Treat 111(3):549–553 Kim SU, Park SK, Yoo KY, Yoon KS, Choi JY, Seo JS et al, 2002, XRCC1 genetic polymorphism and breast cancer risk. Pharmacogenetics 12(4):335–338 Loizidou MA, Michael T, Neuhausen SL, Newbold RF, Marcou Y, Kakouri E et al, 2008, Genetic polymorphisms in the DNA repair genes XRCC1, XRCC2 and XRCC3 and risk of breast cancer in Cyprus. Breast Cancer Res Treat 112(3):575–579 Thyagarajan B, Anderson KE, Folsom AR, Jacobs DR Jr, Lynch CF, Bargaje A et al, 2006, No association between XRCC1 and XRCC3 gene polymorphisms and breast cancer risk: Iowa Women's HealthStudy. Cancer Detect Prev 30(4):313–321 Lu J, Zhao H, Li S, Tian Z, Zhu X,Wang H et al, 2015, Correlation of rs1799793 polymorphism in ERCC2 and the clinical response to platinum-based chemotherapy in patients with triple negative breast cancer. Int J Clin Exp Med 8(2):2934–2938 Brewster AM, Jorgensen TJ, Ruczinski I, Huang HY, Hoffman S, Thuita L et al, 2006, Polymorphisms of the DNA repair genes XPD, Lys751Gln, and XRCC1, Arg399Gln and Arg194Trp): relationship to breast cancer risk and familial predisposition to breast cancer. Breast Cancer Res Treat 95(1):73–80 Smolarz B, Makowska M, Samulak D, Michalska MM, Mojs E, WilczakMet al, 2014, Single nucleotide polymorphisms, SNPs, of ERCC2, hOGG1, and XRCC1 DNA repair genes and the risk of triple-negative breast cancer in Polish women. Tumour Biol 35(4): 3495–3502 Parshad R, Sanford KK(2001, Radiation-induced chromatid breaks and deficient DNA repair in cancer predisposition. Crit Rev Oncol Hematol 37(2):87–96 Breast Cancer Association Consortium, 2006, Commonly studied singlenucleotide polymorphisms and breast cancer: results from the Breast Cancer Association Consortium. J Natl Cancer Inst 98: 1382–1396 Ali AM, AbdulKareem H, Al Anazi M, Reddy Parine N, Shaik JP, Alamri A et al, 2016, Polymorphisms in DNA repair gene XRCC3 and susceptibility to breast cancer in Saudi females. Biomed Res Int , DOI 10.1155/2016/8721052 Kuschel B, Auranen A, McBride S, Novik KL, Antoniou A, Lipscombe JM et al, 2002, Variants in DNA double-strand break repair genes and breast cancer susceptibility. Hum Mol Genet 11(12):1399–1407 Su CH, ChangWS, Hu PS, Hsiao CL, Ji HX, Liao CH et al, 2015, Contribution of DNA double-strand break repair gene XRCC3 genotypes to triple negative breast cancer risk. Cancer Genomics Proteomics 12(6):359–367 Romanowicz-Makowska H, Smolarz B, Zadrozny M, Westfal B, Baszczynski J, Polac I et al, 2011, Single nucleotide polymorphisms in the homologous recombination repair genes and breast cancer risk in Polish women. Tohoku J Exp Med 224(3):201–208 Vral A,Willems P, Claes K, Poppe B, Perletti G, Thierens H, 2011, Combined effect of polymorphisms in Rad51 and Xrcc3 on breast cancer risk and chromosomal radiosensitivity. Mol Med Rep 4(5): 901–912 Al Zoubi MS, 2015, X-ray repair cross-complementing protein 1 and 3 polymorphisms and susceptibility of breast cancer in a Jordanian population. Saudi Med J 36(10):1163–1167 Vachon CM, Scott CG, Fasching PA, Hall P, Tamimi RM, Li J et al, 2012, Common breast cancer susceptibility variants in LSP1 and RAD51L1 are associated with mammographic density measures that predict breast cancer risk. Cancer Epidemiol Biomark Prev 21(7):1156–1166 Bhatti P, Doody MM, Rajaraman P, Alexander BH, Yeager M, Hutchinson A et al, 2010, Novel breast cancer risk alleles and interaction with ionizing radiation among U.S. radiologic technologists. Radiat Res 173(2):214–224 Li SX, Sjolund A, Harris L, Sweasy JB, 2010, DNA repair and personalized breast cancer therapy. Environ MolMutagen 51(8–9): 897–908 Zhou Q, Zou BW, Xu Y, Xue JX, Meng MB, Liu FJ et al, 2015, DNA repair gene polymorphisms and clinical outcome of patients with primary small cell carcinoma of the esophagus. Tumour Biol 36(3):1539–1548 Quintela-Fandino M, Hitt R, Medina PP, Gamarra S, Manso L, Cortes-Funes H et al, 2006, DNA-repair gene polymorphisms predict favorable clinical outcome among patients with advanced squamous cell carcinoma of the head and neck treated with cisplatinbased induction chemotherapy. J Clin Oncol 24(26):4333–4339 Kalikaki A, Kanaki M, Vassalou H, Souglakos J, Voutsina A, GeorgouliasVet al, 2009, DNArepair gene polymorphisms predict favorable clinical outcome in advanced non-small-cell lung cancer. Clin Lung Cancer 10(2):118–123 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 107 EP 114 DI 10.1007/s12253-017-0322-3 PG 8 ER PT J AU Li, J Liu, Y Xue, J Xu, M Zhang, J Liu, J Wang, W AF Li, Jianchao Liu, Yifei Xue, Jianhua Xu, Mingming Zhang, Jianguo Liu, Junhua Wang, Wenyi TI Kruppel-Like Factor 8 Overexpression Correlates with Poor Prognosis in Non-Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Kruppel-like factor 8; Non-small cell lungcancer; Pithelial-mesenchymal transition; Prognosis ID Kruppel-like factor 8; Non-small cell lungcancer; Pithelial-mesenchymal transition; Prognosis AB Kruppel-like factor 8 (KLF8) plays a key role in cancer progression. However, its expression pattern and relationship with clinicopathological characteristics in non-small cell lung cancer (NSCLC) has not been completely elucidated. The study aimed to investigate the expression of KLF8 and its correlation with clinical pathologic features in NSCLC and to explore the potential mechanism. The expression of KLF8 in NSCLC was detected by RT-PCR,Western blot and immunohistochemistry. The expression of Vimentin and E-cadherin, epithelial-mesenchymal transition (EMT) markers, were detected by immunohistochemistry in the NSCLC. The relationship between KLF8 expression and various clinicopathological features or EMT markers was investigated. The results showed m-RNA and protein of KLF8 were overexpressed in NSCLC and the percent of KLF8 positive cells was positively correlated with TNM stage, lymph node metastasis and poor overall survive. Moreover, high expression of KLF8 correlated with E-cadherin low expression, and Vimentin overexpression. Additionally, COX multivariate regression analysis suggested that TNM stage, KLF8, E-cadherin and Vimentin were independent prognostic indicators for overall survival of patients with NSCLC. The data demonstrate that KLF8 is overexpressed in NSCLC. KLF8 overexpression promotes the malignancy of NSCLC, which mechanism may be involved in EMT. KLF8 maybe serve as a potential therapeutic target for NSCLC. C1 [Li, Jianchao] the First People’s Hospital of Yancheng, Department of Thoracic surgery, 166 Yulongxi Road, 224006 Yancheng, Jiangsu, China. [Liu, Yifei] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, Jiangsu, China. [Xue, Jianhua] Affiliated Hospital of Nantong University, Department of Thoracic Surgery, 20 Xisi Road, 226001 Nantong, Jiangsu, China. [Xu, Mingming] Affiliated Hospital of Nantong University, Department of Thoracic Surgery, 20 Xisi Road, 226001 Nantong, Jiangsu, China. [Zhang, Jianguo] Affiliated Hospital of Nantong University, Pathology Medicine Center, 226001 Nantong, Jiangsu, China. [Liu, Junhua] Affiliated Hospital of Nantong University, Department of Thoracic Surgery, 20 Xisi Road, 226001 Nantong, Jiangsu, China. [Wang, Wenyi] the First People’s Hospital of Yancheng, Department of Thoracic surgery, 166 Yulongxi Road, 224006 Yancheng, Jiangsu, China. RP Wang, W (reprint author), the First People’s Hospital of Yancheng, Department of Thoracic surgery, 224006 Yancheng, China. EM 420002318@qq.com CR Jemal A et al, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics. CA Cancer J Clin 62(1):10–29 Gao W et al, 2011, Circulating microRNAs: possible prediction biomarkers for personalized therapy of non-small-cell lung carcinoma. Clin Lung Cancer 12(1):14–17 Smith CB, Kelley AS,Meier DE, 2010, Evidence for new standard of care in non-small cell lung cancer patients. Semin Thorac Cardiovasc Surg 22(3):193–194 Pearson R et al, 2008, Kruppel-like transcription factors: a functional family. Int J Biochem Cell Biol 40(10):1996–2001 Zhao J et al, 2003, Identification of transcription factor KLF8 as a downstream target of focal adhesion kinase in its regulation of cyclin D1 and cell cycle progression. Mol Cell 11(6):1503–1515 Wang X, Zhao J, 2007, KLF8 transcription factor participates in oncogenic transformation. Oncogene 26(3):456–461 Quadrini KJ, Bieker JJ, 2002, Kruppel-like zinc fingers bind to nuclear import proteins and are required for efficient nuclear localization of erythroid Kruppel-like factor. J Biol Chem 277(35): 32243–32252 van Vliet J, Turner J, Crossley M, 2000, Human Kruppel-like factor 8: a CACCC-box binding protein that associates with CtBP and represses transcription. Nucleic Acids Res 28(9):1955–1962 Wang X et al, 2007, Kruppel-like factor 8 induces epithelial to mesenchymal transition and epithelial cell invasion. Cancer Res 67(15):7184–7193 Wang X et al, 2008, Activation of KLF8 transcription by focal adhesion kinase in human ovarian epithelial and cancer cells. J Biol Chem 283(20):13934–13942 Wang X et al, 2011, KLF8 promotes human breast cancer cell invasion and metastasis by transcriptional activation of MMP9. Oncogene 30(16):1901–1911 Fu WJ et al, 2010, Small interference RNA targeting Kruppel-like factor 8 inhibits the renal carcinoma 786-0 cells growth in vitro and in vivo. J Cancer Res Clin Oncol 136(8):1255–1265 Yang T et al, 2012, Kruppel-like factor 8 is a newWnt/beta-catenin signaling target gene and regulator in hepatocellular carcinoma. PLoS One 7(6):e39668 Li JC et al, 2010, Up-regulation of Kruppel-like factor 8 promotes tumor invasion and indicates poor prognosis for hepatocellular carcinoma. Gastroenterology 139(6):2146–2157 e12 Liu L et al, 2012, Lentivirus-delivered Kruppel-like factor 8 small interfering RNA inhibits gastric cancer cell growth in vitro and in vivo. Tumour Biol 33(1):53–61 Wang WF et al, 2013, Kruppel-like factor 8 overexpression is correlated with angiogenesis and poor prognosis in gastric cancer. World J Gastroenterol 19(27):4309–4315 Thiery JP, 2002, Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2(6):442–454 IwatsukiMet al, 2010, Epithelial-mesenchymal transition in cancer development and its clinical significance. Cancer Sci 101(2):293–299 Thiery JP et al, 2009, Epithelial-mesenchymal transitions in development and disease. Cell 139(5):871–890 Goldstraw P, 2009, The 7th Edition of TNM in Lung Cancer: what now? J Thorac Oncol 4(6):671–673 Tischler V et al, 2011, L1CAM protein expression is associated with poor prognosis in non-small cell lung cancer. Mol Cancer 10:127 Tamura M et al, 2005, Prognostic significance of dysadherin expression in patients with non-small cell lung cancer. J Thorac Cardiovasc Surg 130(3):740–745 Maeda J et al, 2008, Proteomic analysis of stage I primary lung adenocarcinoma aimed at individualisation of postoperative therapy. Br J Cancer 98(3):596–603 Richardson F et al, 2012, The evaluation of E-Cadherin and vimentin as biomarkers of clinical outcomes among patients with non-small cell lung cancer treated with erlotinib as second- or thirdline therapy. Anticancer Res 32(2):537–552 Chikaishi Y, Uramoto H, Tanaka F, 2011, The EMT status in the primary tumor does not predict postoperative recurrence or diseasefree survival in lung adenocarcinoma. Anticancer Res 31(12): 4451–4456 Soltermann A et al, 2008, Prognostic significance of epithelialmesenchymal and mesenchymal-epithelial transition protein expression in non-small cell lung cancer. Clin Cancer Res 14(22): 7430–7437 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 115 EP 121 DI 10.1007/s12253-017-0321-4 PG 7 ER PT J AU Park, YJ Hong, D Park, YJ AF Park, Y Ji Hong, Daegy Park, Young Ji TI Association between Morphological Patterns of Myometrial Invasion and Cancer Stem Cell Markers in Endometrial Endometrioid Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer stem cell marker; Epithelial-to-mesenchymal transition; Endometrioid carcinoma; Myometrial invasion ID Cancer stem cell marker; Epithelial-to-mesenchymal transition; Endometrioid carcinoma; Myometrial invasion AB In endometrial endometrioid adenocarcinoma (EEC), the depth of myometrial invasion (MI) is an important parameter for determining whether additional treatment is warranted. The present study investigated the association between MI patterns, cancer stem cell (CSC) phenotypes, and their clinicopathological significance in EEC. A total of 73 cases of EEC with MI were examined in this study. Haematoxylin and eosin-stained tissue specimens were analysed for MI pattern, which was categorised as infiltrating; expansile; adenomyosis (AM)-like; or microcystic, elongated, and fragmented (MELF)-type. The expression of CSC markers such as cluster of differentiation (CD)44, CD133, and Nanog1, as well as oestrogen receptor (ER) and progesterone receptor (PR) was examined by immunohistochemistry. Clinicopathological features including age, DOI, MI pattern, LVI, lymph node (LN) metastasis, disease progression, and survival outcome were recorded. Most examined cases (45/73) were International Federation of Gynecology and Obstetrics (FIGO) stage I. MI showed infiltrating (49.3%), AM-like (26.3%), MELF (15.1%), and expansile (9.6%) patterns. Tumours with the infiltrating pattern were associated with high FIGO grade (P = 0.002), reduced ER and PR, and CD44 expression (P = 0.014, 0.026, and 0.030, respectively); those with a MELF pattern showed LN metastasis (P < 0.001), lymphovascular invasion (P = 0.011), and reduced ER, CD44, and CD133 expression (P = 0.036, 0.006, and 0.016, respectively). EEC with infiltrating/MELF patterns of MI is associated with worse prognosis. These results suggest that CSC expression profiles are an unfavourable indicator of EEC. C1 [Park, Y Ji] Catholic University of Daegu, School of Medicine, Department of PathologyDaegu, South Korea. [Hong, Daegy] Kyungpook National University School of Medicine, Kyungpook National University Medical Center, Gynecologic Cancer CenterDaegu, South Korea. [Park, Young Ji] Kyungpook National University School of Medicine, Kyungpook National University Medical Center, Department of Pathology, 807 Hoguk-ro, Buk-gu, 41404 Daegu, South Korea. RP Park, YJ (reprint author), Kyungpook National University School of Medicine, Kyungpook National University Medical Center, Department of Pathology, 41404 Daegu, South Korea. EM jyparkmd@knu.ac.kr CR Cole AJ, Quick CM, 2013, Patterns of myoinvasion in endometrial adenocarcinoma: recognition and implications. Adv Anat Pathol 20(3):141–147 Euscher E, Fox P, Bassett R, Al-Ghawi H, Ali-Fehmi R, Barbuto D, Malpica A, 2013, The pattern ofmyometrial invasion as a predictor of lymph node metastasis or extrauterine disease in low-grade endometrial carcinoma. Am J Surg Pathol 37(11):1728–1736 ClarkeMF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Wahl GM, 2006, Cancer stem cells–perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res 66(19):9339–9344 Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres- Cortes J, Dick JE, 1994, A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367(6464):645–648 Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF, 2003, Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100(7):3983–3988 Singh SK, Hawkins C, Clark ID, Squire JA, Bayani J, Hide T, Dirks PB, 2004, Identification of human brain tumour initiating cells. Nature 432(7015):396–401 Brendel C, Scharenberg C, DohseM, Robey RW, Bates SE, Shukla S, Neubauer A, 2007, Imatinib mesylate and nilotinib, AMN107, exhibit high-affinity interaction with ABCG2 on primitive hematopoietic stem cells. Leukemia 21(6):1267–1275 Fan X,MatsuiW, Khaki L, Stearns D, Chun J, LiYM, Eberhart CG, 2006, Notch pathway inhibition depletes stem-like cells and blocks engraftment in embryonal brain tumors. Cancer Res 66(15):7445–7452 Phillips TM, McBride WH, Pajonk F, 2006, The response of CD24(−/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancer Inst 98(24):1777–1785 Szotek PP, Pieretti-Vanmarcke R, Masiakos PT, Dinulescu DM, Connolly D, Foster R, Donahoe PK, 2006, Ovarian cancer side population defines cells with stem cell-like characteristics and Mullerian Inhibiting Substance responsiveness. Proc Natl Acad Sci U S A 103(30):11154–11159 Bissell MJ, Radisky D, 2001, Putting tumours in context. Nat Rev Cancer 1(1):46–54 Gupta GP, Massague J, 2006, Cancer metastasis: building a framework. Cell 127(4):679–695 Hu M, Polyak K, 2008, Microenvironmental regulation of cancer development. Curr Opin Genet Dev 18(1):27–34 Mirantes C, Espinosa I, Ferrer I, Eaton EN, Ayyanan A, Zhou AY, Weinberg RA, 2013, Epithelial-to-mesenchymal transition and stem cells in endometrial cancer. Hum Pathol 44(10):1973–1981 Hollier BG, Evans K, Mani SA The epithelial-to-mesenchymal transition and cancer stemcells: a coalition against cancer therapies. J Mammary Gland Biol Neoplasia 14(1):29–43 Hugo H, Ackland ML, Blick T, Lawrence MG, Clements JA, Williams ED, Thompson EW, 2007, Epithelial-mesenchymal and mesenchymal-epithelial transitions in carcinoma progression. J Cell Physiol 213(2):374–383 Singh A, Settleman J, 2010, EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 29(34):4741–4751 Mani SA, GuoW, LiaoMJ, Dolcet X, Prat J, Matias-Guiu X, 2008, The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133(4):704–715 Nakamura M, Kyo S, Zhang B, Zhang X, Mizumoto Y, Takakura M, Inoue M, 2010, Prognostic impact of CD133 expression as a tumor-initiating cell marker in endometrial cancer. Hum Pathol 41(11):1516–1529 Mizrak D, Brittan M, Alison M, 2008, CD133: molecule of the moment. J Pathol 214(1):3–9 Hammond ME, Hayes DE, Dowsett M, Allred DC, Hagerty KL, Badve S American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer, unabridged version). Arch Pathol Lab Med 134(7):48–72 Koyuncuoglu M, Okysy E, Saatli B, Olgan S, Akin M, Saygili U, 2012, Tumor budding and E-Cadherin expression in endometrial carcinoma: are they prognostic factors in endometrial cancer? Gynecol Oncol 125(1):208–213 Rutella S, Bonanno G, Procoli A, Mariotti A, Corallo M, Prisco MG, Ferrandina G, 2009, Cells with characteristics of cancer stem/ progenitor cells express the CD133 antigen in human endometrial tumors. Clin Cancer Res 15(13):4299–4311 Friel AM, Zhang L, Curley MD, Therrien VA, Sergent PA, Belden SE, Rueda BR, 2010, Epigenetic regulation of CD133 and tumorigenicity of CD133 positive and negative endometrial cancer cells. Reprod Biol Endocrinol 8:147–160 Zagorianakou N, IoachimE, Mitselou A, Kitsou E, Zagorianakou P, Stefanaki S, Agnantis NJ, 2003, Glycoprotein CD44 expression in normal, hyperplasic and neoplastic endometrium. An immunohistochemical study including correlations with p53, steroid receptor status and proliferative indices, PCNA, MIB1). Eur J Gynaecol Oncol 24(6):500–504 Murray SK, Young RH, Scully RE, 2003, Unusual epithelial and stromal changes in myoinvasive endometrioid adenocarcinoma: a study of their frequency, associated diagnostic problems, and prognostic significance. Int J Gynecol Pathol 22(4):324–333 Castilla MA,Moreno-Bruno G, Romero-Perez L, Van De Vijver K, Biscuola M, Lopez-Garcia MA, Palacios J, 2011, Micro-RNA signature of the epithelial-mesenchymal transition in endometrial carcinosarcoma. J Pathol 223(1):72–80 Wik E, Raeder MB, Krakstad C, Trovik J, Birkeland E, Hoivik EA, Salvesen HB, 2013, Lack of estrogen receptor-alpha is associated with epithelial-mesenchymal transition and PI3K alterations in endometrial carcinoma. Clin Cancer Res 19(5):1094–1105 Stewart CJ, Little L, 2009, Immunophenotypic features of MELF pattern invasion in endometrial adenocarcinoma: evidence for epithelial-mesenchymal transition. Histopathology 55(1):91–101 Hanekamp EE, Gielen SC, De Ruiter PE, Chadha-Ajwani S, Brinkmann AO, Blok LJ, 2005, Differences in invasive capacity of endometrial cancer cell lines expressing different progesterone receptor isotypes: possible involvement of cadherins. J Soc Gynecol Investig 12(4):278–284 Dai D, Wolf DM, Litman ES, White MJ, Leslie KK, 2002, Progesterone inhibits human endometrial cancer cell growth and invasiveness: down-regulation of cellular adhesion molecules through progesterone B receptors. Cancer Res 62(3):881–886 Hanekamp EE, Kuhne EC, Smid-Koopman E, Chadha-Ajwani S, Huikeshoven FJ, Burger CW, Blok LJ, 2002, Loss of progesterone receptor may lead to an invasive phenotype in human endometrial cancer. Eur J Cancer 38(Suppl 6):S71–SS2 Guarino M, Rubino B, Ballabio G, 2007, The role of epithelialmesenchymal transition in cancer pathology. Pathology 39(3):305– 318 Thiery JP, 2002, Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2(6):442–454 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 123 EP 130 DI 10.1007/s12253-017-0320-5 PG 8 ER PT J AU Nemeth, Zs Boer, K Borbely, K AF Nemeth, Zsuzsanna Boer, Katalin Borbely, Katalin TI Advantages of 18F FDG-PET/CT over Conventional Staging for Sarcoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE 18F–FDG pet/Ct; Standardized uptake value; Imaging modalities; Sarcoma ID 18F–FDG pet/Ct; Standardized uptake value; Imaging modalities; Sarcoma AB The effective management of patients with sarcomas requires accurate diagnosis and staging. Imaging, such as ultrasonography (US), computed tomography (CT), magnetic resonance imaging (MRI) are the most freqently used methods for the detection of the lesion location, size, morphology and structural changes to adjacent tissues; however, these modalities provide little information about tumour biology. MRI is a robust and useful modality in tumour staging of sarcomas, however metabolic-fluorodeoxyglucose positron emission tomography/ computer tomography (18F–FDG PET/CT) provides greater accuracy to overall staging in combination with MRI [1]. The advantages of 18F–FDG PET/CT method compared with CT and MRI is that it provides a whole body imaging, maps the viability of the tumour or themetabolic activity of the tissue. Additionally, PET detects the most agressive part of the tumour, demonstrates the biological behaviour of the tumour and therefore has a predictive value. Little data ara available on the role of 18F–FDG PET/CT in the management of sarcomas. The present manuscript aims to provide a review of the major indications of 18F–FDG PET/CT for diagnosis, staging, restaging and monitoring response to therapy and to compare its usefulness with the conventional imaging modalities in the management of patients with sarcomas. C1 [Nemeth, Zsuzsanna] Szent Margit Hospital, V. Department of Internal Medicine - OncologyBudapest, Hungary. [Boer, Katalin] Szent Margit Hospital, V. Department of Internal Medicine - OncologyBudapest, Hungary. [Borbely, Katalin] National Institute of Oncology, PET/CT Outpatient DepartmentBudapest, Hungary. RP Nemeth, Zs (reprint author), Szent Margit Hospital, V. Department of Internal Medicine - Oncology, Budapest, Hungary. EM zsnemethzsuzsanna@gmail.com CR Faizi NA, Thulkar S, Sharma R et al, 2012, Magnetic resonance imaging and positron emission tomography-computed tomography evaluation of soft-tissue sarcoma with surgical and histopathological corelation. Indian J Nucl Med 27(4):213–220 Demetri GD, Antonia S, Benjamin RS et al, 2010, Soft-tissue sarcoma. J Natl Compr Cancer Netw 8(6):630–674 Stiller CA, Trama A, Serraino D et al, 2013, Descriptive epidemiology of sarcomas in Europe: report from the RARECARE project. Eur J Cancer 49(3):684–695. , DOI 10.1016/j.ejca.2012.09.011 Guillou L, Coindre JM, Bonichon F et al, 1997, Comparative study of the National Cancer Institute and French Federation of Cancer Centers Sarcoma Group grading systems in a population of 410 adult patients with soft-tissue sarcoma. J Clin Oncol 15:350–362 von Mehren M, Randall LR, Benjamin RS, et al Soft Tissue Sarcoma. NCCN Clinical Practice Guidelines in Oncology, NCCN Guidelines). Version 1.2017 https://www.nccn.org/ professionals/physician_gls/pdf/sarcoma.pdf The European Society for Medical Oncology, ESMO, Sarcoma Network Working Group, 2014, Soft-tissue and visceral sarcomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 25(3):102–112. , DOI 10.1093/ annonc/mdu254 Borbely K, 1999, A pozitron emisszios tomografia helye a korszeru betegvezetesben. Orv Hetil 140:171–178 Borbely K, Szilagyi I, Kasler M, 2011, IV. PET/CT Multidiszciplinaris Nemzeti Konszenzus Konferencia Allasfoglalasa. Mag Onkol 55(2):117–127 Wang X, Jacobs M, Fayad L, 2011, Therapeutic response in musculosceletal soft-tissue sarcomas: evaluation by magnetic resonance imaging. NMR Biomed 24(6):750–763 Lin X, Davion S, Bertsch EC, Omar I, Nayar R, Laskin WB, 2016, FNCLCC grading of soft-tissue sarcomas on needle core biopsies using surrogate markers. Hum Pathol 56:147 PubMedView ArticleGoogle Scholar Schwarzbach MH, Dimitrakopoulou-Strauss A, Willeke F et al, 2000, 18-F fluorodeoxy-glucose positron emission tomography imaging in soft-tissue sarcomas. Ann Surg 231:380–386 Volker T, Denecke T, Steffen I et al, 2007, Positron emission tomography for staging of pediatric sarcoma patients: results of a prospective multicenter trial. J Clin Oncol 25(34):5435–5441 Aoki J,Watanabe H, Shinozaki et al, 2001, T FDG PETof primary benign and malignant bone tumours: standardized uptake value in 52 lesions. Radiology 219:774–777 Borbely K, Szilagyi I, Kasler M, 2011, IV. PET/CT Multidiszciplinaris Nemzeti Konszenzus Konferencia Allasfoglalasa, 2011, Magyar Onkologia 55, 2):117–127 Franzius C, Sciuk J, Daldrup-Link HE et al, 2000, FDG-PET for detection of pulmonary metastases from malignant primary bone tumours: comparison with bone scintigraphy. Eur J Nucl Med 27: 1305–1311 O’Sullivan GJ, Carty FL, Cronin CG, 2015, Imaging of bone metastasis: an update. World J Radiol 7(8):202–211 Liangjun R, Zhen Z, Zhifeng C et al, 2016, 18F-Labeled NaF PETCT in detection of bone metastases in patients with preoperative lung cancer medicine. Medicine 95(16):1–6 Sampath SC, Sampath SC, Mosci C et al, 2015, Detection of osseous metastasis by 18F NaF/18F F-FDG PET/CT versus CT alone. Clin Nucl Med 40(3):e173–e177 Schwab JH, Boland PJ, Antonescu C et al, 2007, Spinal metastases from myxoid liposarcoma warrant screening with magnetic resonance imaging. Cancer 110:1815–1821 Adler LP, Blair HF, Makley JT et al, 1991, Noninvasive grading of musculoskeletal tumours using PET. J Nucl Med 32(8):1508–1512 Ioannidis JP, Lau J, 2003, 18F-FDG PET for the diagnosis and grading of soft-tissue sarcoma: a meta-analysis. J Nucl Med 44: 717–724 Kwee TC, Cheng G, Lam MG, Basu S, Alavi A, 2013, SUVof 2.5 should not be embraced as a magic threshold for separating benign from malignant lesions. Eur J Nucl Med Mol Imaging 40(10): 1475–1477 Hawkins DS, Schuetze SM, Butrynski JE et al, 2005, [18F] Fluorodeoxyglucose positron emission tomography predicts outcome for Ewing sarcoma family of tumours. J Clin Oncol 23:8828–8834 Schuetze SM, Rubin BP, Vernon C et al, 2005, Use of positron emission tomography in localized extremity soft-tissue sarcoma treated with neoadjuvant chemotherapy. Cancer 103:339–348 Evilevitch V, Weber WA, Tap WD et al, 2008, Reduction of glucose metabolic activity is more accurate than change in size at predicting histo-pathologic response to neoadjuvant therapy in high-grade soft-tissue sarcomas. Clin Cancer Res 14:715–720 Machulla HJ, Blocher A, KuntzschMet al, 2000, Simplified labeling approach for synthesizing 3′-deoxy-3′-[18F] fluorothymidine, [18F] FLT). J. Radioanal. Nucl. Chem. 243(3):843–846 Tateishi U, Kawai A, Hirokazu C et al, 2011, PET/CT allows stratification of responders to neoadjuvant chemotherapy for high grade sarcoma: a prospective study. Clin Nucl Med 36(7):526–532 Enzinger FM, 1970, Epithelioid sarcoma: a sarcoma stimulating a granuloma or a carcinoma. Cancer 26:1029–1041 Sanghera B, Wong WL, Sonoda L et al, 2014, FLT PET-CT in evaluation of treatment response. Indian J Nucl Med Apr-Jun 29(2):65–73 Buck AK, Halter G, Schirrmeister H et al, 2003, Imaging proliferation in lung tumours with PET: 18F-FLT versus 18F-FDG. J Nucl Med 44:1426–1431 Buck AK, Bommer M, Stilgenbauer S et al, 2006, Molecular imaging of proliferation in malignant lymphoma. Cancer Res 66: 11055–11061 Herrmann K, Buck AK, Schuster T, Junger A, Wieder HA, Graf N et al, 2011, Predictive value of initial 18F-FLT uptake in patients with aggressive non-Hodgkin lymphoma receiving R-CHOP treatment. J Nucl Med 52(5):690–696 Yamamoto Y, Ono Y, Aga F et al, 2012, Correlation of 18 F-FLT uptake with tumour grade and Ki-67 immunohistochemistry in patients with newly diagnosed and recurrent gliomas. J Nucl Med 53(12):1911–1915 Contractor KB, Kenny LM, Stebbing J, Rosso L, Ahmad R, Jacob J et al, 2011, [18F]-3′deoxy-3′-fluorothymidine positron emission tomography and breast cancer response to docetaxel. Clin Cancer Res 17(24):7664–7672 Buck AK, Hermann K, Buschenfelde CM et al, 2008, Imaging bone and soft-tissue tumours with the proliferation marker [18F] fluorodeoxythymidine. Clin. Cancer Res. 14(10):2970–2977 BenzMR, Czernin J, Allen-AuerbachMS, 2012, 3′-deoxy-3′-[18F] fluorothymidine positron emission tomography for response assessment in soft-tissue sarcoma: a pilot study to correlate imaging findings with tissue thymidine kinase 1 and Ki-67 activity and histopathologic response HHS Author Manuscript Cancer 118, 12): 3135–3144 Been LB, Suurmeijer AJ, Elsinga PH, Jager PL, van Ginkel RJ, Hoekstra HJ, 2007, 18F-fluorodeoxythymidine PET for evaluating the response to hyperthermic isolated limb perfusion for locally advanced soft-tissue sarcomas. J Nucl Med 48:367–372 Jerabek PA, Patrick TB, Kilbourn MR, Dischino DD, Welch MJ, 1986, Synthesis and biodistribution of of 18F-labeled fluoronitroimidazoles: potential in vivo markers of hypoxic tissue. Int J Rad Appl Instrum Part A 37:599–605 Rajendran JG, Wilson DC, Conrad EU et al, 2003, [18F] FMISO and [18F] FDG PET imaging in soft-tissue sarcomas: correlation of hypoxia, metabolism and VEGF expression. Eur J Nucl Med Mol Imaging 30(5):695–704 Borbely K, Zs N, Kasler M, 2014, Clinical application 18F-FDG PET/CT in the treatment of sarcomas. Hung Oncol 58(1):24–31 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 131 EP 136 DI 10.1007/s12253-017-0325-0 PG 6 ER PT J AU CJ, P HV, E Vijayakurup, V Menon, RG Nair, S Gopala, S AF CJ, Padmakrishnan HV, Easwer Vijayakurup, Vinod Menon, R Girish Nair, Suresh Gopala, Srinivas TI High LC3/Beclin Expression Correlates with Poor Survival in Glioma: a Definitive Role for Autophagy as Evidenced by In Vitro Autophagic Flux SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; Autophagy; LC3; Autophagic flux; p62; Beclin 1 ID Glioma; Autophagy; LC3; Autophagic flux; p62; Beclin 1 AB Recent studies suggest the role of autophagy, an evolutionarily conserved catabolic process, in determining the response of gliomas to treatment either positively or negatively. The study attempts to characterize autophagy in low and highgrade glioma by investigating the autophagic flux and clinical significance of autophagy proteins (LC3 and beclin 1) in a group of glioma patients. We evaluated the expression of autophagic markers in resected specimens of low-grade glioma (LGG) and high-grade glioma (HGG) tissues, by immunohistochemistry and Western blotting. Our results show that expression of autophagy proteins were more prominent in HGG than in LGG. Increased level of autophagic proteins in HGG can be due to an increased rate of autophagy or can be because of blockage in the final degradation step of autophagy (defective autophagy). To distinguish these possibilities, the autophagic flux assaywhich helps to determine the rate of degradation/synthesis of autophagic proteins (LC3-II and p62) over a period of time by blocking the final degradation step of autophagy using bafilomycin A1 was used. The assessment of autophagic flux in ex vivo culture of primary glioma cells revealed for the first time increased turnover of autophagy in high grade compared to low grade-glioma. Though autophagic markers were reduced in LGG, functionally autophagy was non defective in both grades of glioma. We then investigated whether autophagy in gliomas is regulated by nutrient sensing pathways including mTOR and promote cell survival by providing an alternate energy source in response to metabolic stress. The results depicted that the role of autophagy during stress varies with tissue and has a negative correlation with mTOR substrate phosphorylation.We also evaluated the expression of LC3 and beclin 1 with progression free survival (PFS) using Kaplan-Meier survival analysis and have found that patients with low LC3/beclin 1 expression had better PFS than those with high expression of LC3/beclin 1 in their tumors. Together, we provide evidence that autophagy is non-defective in glioma and also show that high LC3/beclin 1 expression correlates with poor PFS in both LGG and HGG. C1 [CJ, Padmakrishnan] Sree Chitra Tirunal Institute for Medical Sciences and Technology, Department of Biochemistry, 695011 Thiruvananthapuram, Kerala, India. [HV, Easwer] Sree Chitra Tirunal Institute for Medical Sciences and Technology, Department of Neurosurgery, 695011 Thiruvananthapuram, Kerala, India. [Vijayakurup, Vinod] Sree Chitra Tirunal Institute for Medical Sciences and Technology, Department of Biochemistry, 695011 Thiruvananthapuram, Kerala, India. [Menon, R Girish] Sree Chitra Tirunal Institute for Medical Sciences and Technology, Department of Neurosurgery, 695011 Thiruvananthapuram, Kerala, India. [Nair, Suresh] Sree Chitra Tirunal Institute for Medical Sciences and Technology, Department of Neurosurgery, 695011 Thiruvananthapuram, Kerala, India. [Gopala, Srinivas] Sree Chitra Tirunal Institute for Medical Sciences and Technology, Department of Biochemistry, 695011 Thiruvananthapuram, Kerala, India. RP Gopala, S (reprint author), Sree Chitra Tirunal Institute for Medical Sciences and Technology, Department of Biochemistry, 695011 Thiruvananthapuram, India. EM srinivasg@sctimst.ac.in CR Schwartzbaum JA, Fisher JL, Aldape KD, Wrensch M, 2006, Epidemiology and molecular pathology of glioma. Nat Clin Pract Neurol 2(9):494–503 quiz 1 p following 16 Maher EA, Furnari FB, Bachoo RM, Rowitch DH, Louis DN, Cavenee WK et al, 2001, Malignant glioma: genetics and biology of a grave matter. Genes Dev 15(11):1311–1333 Louis DN, Holland EC, Cairncross JG, 2001, Glioma classification: a molecular reappraisal. Am J Pathol 159(3):779–786 Lefranc F, Kiss R, 2006, Autophagy, the Trojan horse to combat glioblastomas. Neurosurg Focus 20(4):E7 Lefranc F, Brotchi J, Kiss R, 2005, Possible future issues in the treatment of glioblastomas: special emphasis on cell migration and the resistance of migrating glioblastoma cells to apoptosis. J Clin Oncol Off J Am Soc Clin Oncol 23(10):2411–2422 Kanzawa T, Germano IM, Komata T, Ito H, Kondo Y, Kondo S, 2004, Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Cell Death Differ 11(4):448–457 Ito H, Daido S, Kanzawa T, Kondo S, Kondo Y, 2005, Radiationinduced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int J Oncol 26(5):1401–1410 Lomonaco SL, Finniss S, Xiang C, Decarvalho A, Umansky F, Kalkanis SN et al, 2009, The induction of autophagy by gammaradiation contributes to the radioresistance of glioma stem cells. Int J Cancer 125(3):717–722 Jo GH, Bogler O, Chwae YJ, Yoo H, Lee SH, Park JB, et al, 2014, Radiation-induced autophagy contributes to cell death and induces apoptosis partly in malignant glioma cells. Cancer Res Treat 47(2): 221-241 Ge P, Luo Y, Fu S, Ji X, Ling F, 2009, Autophagy: a strategy for malignant gliomas’ resistance to therapy. Med Hypotheses 73(1):45–47 Zhuang W, Qin Z, Liang Z, 2009, The role of autophagy in sensitizing malignant glioma cells to radiation therapy. Acta Biochim Biophys Sin 41(5):341–351 Cuervo AM, 2004, Autophagy: in sickness and in health. Trends Cell Biol 14(2):70–77 Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R et al, 2006, Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441(7095):885–889 Chen N, Debnath J, 2010, Autophagy and tumorigenesis. FEBS Lett 584(7):1427–1435 HippertMM,O'Toole PS, ThorburnA, 2006, Autophagy in cancer: good, bad, or both? Cancer Res 66(19):9349–9351 He C, Klionsky DJ, 2009, Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet 43:67–93 Annovazzi L, Mellai M, Caldera V, Valente G, Tessitore L, Schiffer D, 2009, mTOR, S6 and AKT expression in relation to proliferation and apoptosis/autophagy in glioma. Anticancer Res 29(8):3087–3094 Fan QW, Knight ZA,GoldenbergDD,YuW,Mostov KE, StokoeD et al, 2006, A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma. Cancer Cell 9(5):341–349 Chang YY, Juhasz G, Goraksha-Hicks P, Arsham AM, Mallin DR, Muller LK et al, 2009, Nutrient-dependent regulation of autophagy through the target of rapamycin pathway. Biochem Soc Trans 37(Pt 1):232–236 Kondo Y, Kanzawa T, Sawaya R, Kondo S, 2005, The role of autophagy in cancer development and response to therapy. Nat Rev Cancer 5(9):726–734 Eskelinen EL, 2005, Doctor Jekyll and Mister Hyde: autophagy can promote both cell survival and cell death. Cell Death Differ 12(Suppl 2):1468–1472 Koukourakis MI, Mitrakas AG, Giatromanolaki A, 2016, Therapeutic interactions of autophagy with radiation and temozolomide in glioblastoma: evidence and issues to resolve. Br J Cancer 114(5):485–496 Yan Y, Xu Z, Dai S, Qian L, Sun L, Gong Z, 2016, Targeting autophagy to sensitive glioma to temozolomide treatment. J Exp Clin Cancer Res 35:23 Lefranc F, Facchini V, Kiss R, 2007, Proautophagic drugs: a novel means to combat apoptosis-resistant cancers, with a special emphasis on glioblastomas. Oncologist 12(12):1395–1403 Ballman KV, Buckner JC, Brown PD, Giannini C, Flynn PJ, LaPlant BR et al, 2007, The relationship between six-month progression-free survival and 12-month overall survival end points for phase II trials in patients with glioblastoma multiforme. Neuro-Oncology 9(1):29–38 Tang JY, Hsi E,HuangYC, Hsu NC, Chu PY, Chai CY, 2013, High LC3 expression correlates with poor survival in patients with oral squamous cell carcinoma. Hum Pathol 44(11):2558–2562 Lee YJ, Hah YJ, Kang YN, Kang KJ, Hwang JS, Chung WJ et al, 2013, The autophagy-related marker LC3 can predict prognosis in human hepatocellular carcinoma. PLoS One 8(11):e81540 Liu JL, Chen FF, Lung J, Lo CH, Lee FH, Lu YC et al, 2014, Prognostic significance of p62/SQSTM1 subcellular localization and LC3B in oral squamous cell carcinoma. Br J Cancer 111(5):944–954 Wang XJ, Zhou SL, Fu XD, Zhang YY, Liang B, Shou JX et al, 2015, Clinical and prognostic significance of high-mobility group box-1 in human gliomas. Exp Ther Med 9(2):513–518 Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, Askew DS et al, 2008, Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy 4(2):151–175 Kanzawa T, Kondo Y, Ito H, Kondo S, Germano I, 2003, Induction of autophagic cell death in malignant glioma cells by arsenic trioxide. Cancer Res 63(9):2103–2108 Carew JS, Espitia CM, Esquivel JA 2nd, Mahalingam D, Kelly KR, Reddy G et al, 2011, Lucanthone is a novel inhibitor of autophagy that induces cathepsin D-mediated apoptosis. J Biol Chem 286(8):6602–6613 Klionsky DJ, 2005, The correct way to monitor autophagy in higher eukaryotes. Autophagy 1(2):65 Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K et al, 2012, Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 8(4):445–544 Rubinsztein DC, Cuervo AM, Ravikumar B, Sarkar S, Korolchuk V, Kaushik S et al, 2009, In search of an Bautophagomometer^. Autophagy 5(5):585–589 Sadasivan S,Waghray A, Larner SF, DunnWA Jr, Hayes RL,Wang KK, 2006, Amino acid starvation induced autophagic cell death in PC-12 cells: evidence for activation of caspase-3 but not calpain-1. Apoptosis: Int J Programmed Cell Death 11(9):1573–1582 Ni HM, Bockus A, Wozniak AL, Jones K, Weinman S, Yin XM et al, 2011, Dissecting the dynamic turnover of GFP-LC3 in the autolysosome. Autophagy 7(2):188–204 Kathryn J. Huber-Keener and Jin-Ming Yang, 2011). Impact of Metabolic and Therapeutic Stresses on Glioma Progression and Therapy In: Prof. Clark Chen, Ed.), Advances in the Biology, Imaging and Therapies for Glioblastoma, InTech, Croatia Kang C,Avery L, 2008, To be or not to be, the level of autophagy is the question: dual roles of autophagy in the survival response to starvation. Autophagy 4(1):82–84 Huang X, Bai HM, Chen L, Li B, Lu YC, 2010, Reduced expression of LC3B-II and Beclin 1 in glioblastoma multiforme indicates a down-regulated autophagic capacity that relates to the progression of astrocytic tumors. J Clin Neurosci: Off J Neurosurg Soc Australasia 17(12):1515–1519 Ge PF, Zhang JZ, Wang XF, Meng FK, Li WC, Luan YX et al, 2009, Inhibition of autophagy induced by proteasome inhibition increases cell death in human SHG-44 glioma cells. Acta Pharmacol Sin 30(7):1046–1052 Pirtoli L, Cevenini G, Tini P, Vannini M, Oliveri G, Marsili S et al, 2009, The prognostic role of Beclin 1 protein expression in highgrade gliomas. Autophagy 5(7):930–936 Giatromanolaki A, Sivridis E, Mitrakas A, Kalamida D, Zois CE, Haider S et al, 2014, Autophagy and lysosomal related protein expression patterns in human glioblastoma. Cancer Biol Ther 15(11):1468–1478 Klionsky DJ, Elazar Z, Seglen PO, Rubinsztein DC, 2008, Does bafilomycin A1 block the fusion of autophagosomes with lysosomes? Autophagy 4(7):849–850 Asanuma K, Tanida I, Shirato I, Ueno T, Takahara H, Nishitani T et al, 2003, MAP-LC3, a promising autophagosomal marker, is processed during the differentiation and recovery of podocytes from PAN nephrosis. FASEB J: Off Publ Fed Am Soc Exp Biol 17(9):1165–1167 Tanida I, Minematsu-Ikeguchi N, Ueno T, Kominami E, 2005, Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy 1(2):84–91 Hosokawa N, Hara T, Kaizuka T, Kishi C, Takamura A, Miura Y et al, 2009, Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell 20(7):1981–1991 Kamada Y, Yoshino K, Kondo C, Kawamata T, Oshiro N, Yonezawa K et al, 2010, Tor directly controls the Atg1 kinase complex to regulate autophagy. Mol Cell Biol 30(4):1049–1058 Hau AM, Greenwood JA, Lohr CV, Serrill JD, Proteau PJ, Ganley IG et al, 2013, Coibamide a induces mTORindependent autophagy and cell death in human glioblastoma cells. PLoS One 8(6):e65250 Rodriguez FJ, Raabe EH, 2014, mTOR: a new therapeutic target for pediatric low-grade glioma? CNS Oncol 3(2):89–91 Franz DN, Leonard J, Tudor C, Chuck G, Care M, Sethuraman G et al, 2006, Rapamycin causes regression of astrocytomas in tuberous sclerosis complex. Ann Neurol 59(3):490–498 McBride SM, Perez DA, Polley MY, Vandenberg SR, Smith JS, Zheng S et al, 2010, Activation of PI3K/mTOR pathway occurs in most adult low-grade gliomas and predicts patient survival. J Neuro-Oncol 97(1):33–40 Hao CL, Li Y, Yang HX, Luo RZ, Zhang Y, Zhang MF et al, 2014, High level of microtubule-associated protein light chain 3 predicts poor prognosis in resectable esophageal squamous cell carcinoma. Int J Clin Exp Pathol 7(7):4213–4221 Karpathiou G, Sivridis E, KoukourakisMI,Mikroulis D, Bouros D, Froudarakis ME et al, 2011, Light-chain 3A autophagic activity and prognostic significance in non-small cell lung carcinomas. Chest 140(1):127–134 Wan XB, Fan XJ, ChenMY, Xiang J, Huang PY, Guo L et al, 2010, Elevated Beclin 1 expression is correlated with HIF-1alpha in predicting poor prognosis of nasopharyngeal carcinoma. Autophagy 6(3):395–404 Fujii S, Mitsunaga S, Yamazaki M, Hasebe T, Ishii G, Kojima M et al, 2008, Autophagy is activated in pancreatic cancer cells and correlates with poor patient outcome. Cancer Sci 99(9):1813–1819 Han C, Sun B, Wang W, Cai W, Lou D, Sun Y et al, 2011, Overexpression of microtubule-associated protein-1 light chain 3 is associated with melanoma metastasis and vasculogenic mimicry. Tohoku J Exp Med 223(4):243–251 Guo JY, Karsli-Uzunbas G, Mathew R, Aisner SC, Kamphorst JJ, Strohecker AM et al, 2013, Autophagy suppresses progression of K-ras-induced lung tumors to oncocytomas and maintains lipid homeostasis. Genes Dev 27(13):1447–1461 Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, Hibshoosh H et al, 1999, Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 402(6762):672–676 Hu YL, DeLay M, Jahangiri A,Molinaro AM, Rose SD, Carbonell WS et al, 2012, Hypoxia-induced autophagy promotes tumor cell survival and adaptation to antiangiogenic treatment in glioblastoma. Cancer Res 72(7):1773–1783 Kimmelman AC, 2011, The dynamic nature of autophagy in cancer. Genes Dev 25(19):1999–2010 Knizhnik AV, Roos WP, Nikolova T, Quiros S, Tomaszowski KH, Christmann M et al, 2013, Survival and death strategies in glioma cells: autophagy, senescence and apoptosis triggered by a single type of temozolomide-induced DNA damage. PLoS One 8(1):e55665 Winardi D, Tsai HP, Chai CY, Chung CL, Loh JK, Chen YH et al, 2014, Correlation of altered expression of the autophagy marker LC3B with poor prognosis in astrocytoma. Biomed Res Int 2014:723176 Karagounis IV, Kalamida D, Mitrakas A, Pouliliou S, Liousia MV, Giatromanolaki A et al, 2016, Repression of the autophagic response sensitises lung cancer cells to radiation and chemotherapy. Br J Cancer 115(3):312–321 Koukourakis MI, Kalamida D, Mitrakas A, Pouliliou S, Kalamida S, Sivridis E et al, 2015, Intensified autophagy compromises the efficacy of radiotherapy against prostate cancer. Biochem Biophys Res Commun 461(2):268–274 Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M et al, 2013, Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis 4:e838 Fimia GM, Kroemer G, Piacentini M, 2013, Molecular mechanisms of selective autophagy. Cell Death Differ 20(1):1–2 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 137 EP 148 DI 10.1007/s12253-017-0310-7 PG 12 ER PT J AU Maraz, A Csejtei, A Kocsis, J Szucs, M Kahan, Zs Bodoky, Gy Dank, M Mangel, L Revesz, J Varga, Z Geczi, L AF Maraz, Aniko Csejtei, Andras Kocsis, Judit Szucs, Miklos Kahan, Zsuzsanna Bodoky, Gyorgy Dank, Magdolna Mangel, Laszlo Revesz, Janos Varga, Zoltan Geczi, Lajos TI Assessment of the Role of Everolimus Therapy in Patients with Renal Cell Carcinoma Based on Daily Routine and Recent Research Results SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Metastatic kidney cancer; mTOR inhibitor; Everolimus; Anemia; ECOG; RCC ID Metastatic kidney cancer; mTOR inhibitor; Everolimus; Anemia; ECOG; RCC AB Everolimus is indicated for adults with metastatic renal cell carcinoma (mRCC) after failure of vascular endothelial growth factor receptor-tyrosine kinase inhibitors (TKI). Currently, the therapeutic applicability of EVE has been changing. Multicenter evaluation of efficacy and safety of everolimus in daily routine and definition of patient characteristics with favorable outcome. Data of 165 patients from 9 oncology institutes in Hungary were analyzed retrospectively. Everolimus therapy was used after one TKI in 10 mg starting dose. Physical and laboratory examinations and imaging tests were performed monthly and every 3 months, respectively. Median progression-free survival (PFS) was 5.4 months.Median overall survival (OS) was 16.2 months. PFS and OS results were more favorable in patients with ECOG 0–1 (pPFS = 0.033, pOS = 0.008) and after >9 months of TKI therapy (pPFS = 0.019, pOS = 0.045). Survival was longer in nonanemic patients with ECOG 0–1 than in anemic patients with ECOG 2–3, 30.9 and 7.7 months, respectively (p = 0.029). Dose reduction and treatment delay was required in 6.2% and 8.9% of patients, respectively. Common adverse events were exanthema, edema, stomatitis, anemia, and abnormal kidney functions and glucose levels. Results of this study show that everolimus is safe and efficacious in a real-world setting. Everyday practice showed that nonanemic patients with good performance status receiving TKI therapy for >9 months are favorable candidates for this treatment. Despite the efficiency of novel, registered drugs, everolimus still plays an important role during and after second-line therapy for mRCC when availability of modern remedies is limited. C1 [Maraz, Aniko] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Csejtei, Andras] Vas County Markusovszky Hospital, Markusovszky str. 5Szombathely, Hungary. [Kocsis, Judit] University of Debrecen, Department of Oncology, Nagyerdei krt. 98Debrecen, Hungary. [Szucs, Miklos] Semmelweis University, Department of Urology, Ulloi str. 78/bBudapest, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Bodoky, Gyorgy] Egyesitett Szent Istvan es Szent Laszlo Korhaz, Onkologiai Osztaly, Albert Florian str. 5-7Budapest, Hungary. [Dank, Magdolna] Semmelweis University, 1st Department of Internal Medicine, Tomo str. 25-29Budapest, Hungary. [Mangel, Laszlo] University of Pecs, Department of Oncology, Edesanyak 17Pecs, Hungary. [Revesz, Janos] Borsod County Hospital and University Academic Hospital, Institute of Radiotherapy and Clinical Oncology, Szentpeteri Kapu 72-76Miskolc, Hungary. [Varga, Zoltan] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. [Geczi, Lajos] National Institute of Oncology, Department of Chemotherapy and Clinicopharmacology, Rath Gyorgy str. 7-9Budapest, Hungary. RP Maraz, A (reprint author), University of Szeged, Department of Oncotherapy, 6720 Szeged, Hungary. EM dr.aniko.maraz@gmail.com CR Afinitor EMEA Summary of Product Characteristics: http://www. ema.europa.eu/docs/hu_HU/document_library/EPAR_-_Product_ Information/human/001038/WC500022814.pdf Hay N, Sonenberg N, 2004, Upstream and downstream of mTOR. Genes Dev 18:1926–1945 Eisen HJ, Tuzcu M, Dorent R et al, 2003, Everolimus for the prevention of allograft rejection and vasculopathy in cardiactransplant patients. N Engl J Med 349:847–858 Motzer RJ, Escudier B, Oudard S et al, 2008, Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372:449–456 Yao JC, Shah MH, Ito T et al, 2011, Everolimus for Advanced Pancreatic Neuroendocrine Tumors. N Engl J Med 364:514–523 Yao JC, Fazio N, Singh S et al, 2016, Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract, RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet 387:968–977 Baselga J, Campone M, Piccart M et al, 2012, Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med 366:520–529 Motzer RJ, Escudier B, Oudard S et al, 2010, Phase 3 trial of everolimus for metastatic renal cell carcinoma: final results and analysis of prognostic factors. Cancer 116:4256–4265 Calvo E, Escudier B, Motzer RJ et al, 2011, Everolimus in metastatic renal cell carcinoma: subgroup analysis of patients with 1 or 2 previous vascular endothelial growth factor receptor-tyrosine kinase inhibitor therapies enrolled in the phase III RECORD-1 study. Eur J Cancer 48:333–339 NCCN Guideline. https://www.nccn.org/professionals/physician_ gls/pdf/kidney.pdf Escudier B, Porta C, Schmidinger M et al, 2014, Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 25:49–56 Ljungberg B, Bensalah K, Canfield S et al, 2015, EAU Guidelines on Renal Cell Carcinoma: 2014 Update. Eur Urol 67:913–924 Motzer RJ, Escudier B, McDermott DF, el al, 2015, Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med 373:1803–1813 Choueiri TK, Escudier B, Powles T et al, 2015, Cabozantinib versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med 373:1814–1823 Choueiri TK, Escudier B, Powles T et al, 2016, Cabozantinib versus everolimus in advanced renal cell carcinoma, METEOR): final results from a randomised, open-label, phase 3 trial. Lancet Oncol 17:917–927 Motzer RJ, Hutson TE, Glen H et al, 2015, Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomized, phase 2, open-label, multicenter trial. Lancet Oncol 16:1473–1482 Escudier B, Porta C, Schmidinger M et al, 2016, Renal Cell Carcinoma: ESMO Clinical Practice Guidelines. Ann Oncol 27: 58–68 Therasse P, Arbuck SG, Eisenhauer EA et al, 2000, New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst 92:205–214 Common Terminology Criteria for Adverse Events, version 3.0 issued by National Cancer Institute EORTC, 09. 08, 2006, http:// ctep.cancer.gov/protocolDevelopment/electronic_applications/ docs/ctcaev3.pdf ECOG performance status. http://ecog.dfci.harvard.edu/general/ perf_stat.html Rini BI, Flaherty K, 2008, Clinical effect and future considerations for molecularly-targeted therapy in renal cancer. Uro Oncol Semin Orig Investig 26:543–549 Grunwald V, Karakiewicz PI, Bavbek SE et al, 2012, An international expanded-access programme of everolimus: addressing safety and efficacy in patients with metastatic renal cell carcinoma who progress after initial vascular endothelial growth factor receptortyrosine kinase inhibitor therapy. Eur J Cancer 48:324–332 Calvo E, Ravaud A, Bellmunt J, 2013)What is the optimal therapy for patients with metastatic renal cell carcinoma who progress on an initial VEGFr-TKI? Cancer Treat. Rev. 39:366–374 Bergmann L, Kube U, Doehn C et al, 2015, Everolimus in metastatic renal cell carcinoma after failure of initial anti–VEGF therapy: final results of a noninterventional study. BMC Cancer 15:303 Motzer RJ, Bacik J, Schwartz LH et al, 2004, Prognostic factors for survival in previously treated patients with metastatic renal cell carcinoma. J Clin Oncol 22:454–463 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 149 EP 156 DI 10.1007/s12253-017-0317-0 PG 8 ER PT J AU Li, D Yang, Z Liu, Z Zou, Q Yuan, Y AF Li, Daiqiang Yang, Zhulin Liu, Ziru Zou, Qiong Yuan, Yuan TI DDR2 and IFITM1 Are Prognostic Markers in Gallbladder Squamous Cell/Adenosquamous Carcinomas and Adenocarcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gallbladder; Squamous cell carcinoma; Adenosquamous carcinomas; DDR2; IFITM1; Immunohistochemistry ID Gallbladder; Squamous cell carcinoma; Adenosquamous carcinomas; DDR2; IFITM1; Immunohistochemistry AB This study was conducted to investigate the expressions of DDR2 and IFITM1 and their clinical and pathological significances in the rare type squamous cell/adenosquamous carcinomas (SC/ASC) and ordinary adenocarcinomas (AC) of gallbladder cancers. DDR2 and IFITM1 expression was examined in 69 SC/ASCs and 146 ACs using EnVision immunohistochemistry. Results showed that the percentage of positive DDR2 and IFITM1 expression was significantly higher in SC/ASC patients with high TNM stage, lymph node metastasis, invasion, and no resection surgery compared to patients with low TNM stages, no lymph node metastasis, no invasion, and resection surgery (P < 0.05 or P < 0.01). The positive rate of DDR2 was significantly higher in SC/ASC patients with large tumor sizes than patients with small tumor sizes (p < 0.05). The percentage of positive DDR2 and IFITM1 expressions was significantly higher in AC patients with high TNM stages that didn’t receive resection surgery compared to patients with low TNM stages that did receive resection surgery (P < 0.05 or P < 0.01). The positive rate of IFITM1 was significantly higher in AC patients with lymph node metastasis and invasion than in patients without metastasis and invasion (p < 0.05). Positive DDR2 and IFITM1 expression was closely associated with a decreased overall survival in SC/ASC and AC patients (P < 0.05 or P < 0.01). AUC analysis showed that DDR2 and IFITM1 was sensitive and specific for the diagnosis of SC/ASC (AUC = 0.740 and AUC =0.733, respectively) and AC (AUC = 0.710 and AUC =0.741, respectively). In conclusion, positive DDR2 and IFITM1 expression is a marker for the clinical severity, poor prognosis, and diagnosis of gallbladder SC/ASC and AC. C1 [Li, Daiqiang] Central South University, Second Xiangya Hospital, Department of PathologyChangsha, Hunan, China. [Yang, Zhulin] Central South University, The Second Xiangya Hospital, Department of General Surgery, 410011 Changsha, Hunan, China. [Liu, Ziru] Central South University, The Second Xiangya Hospital, Department of General Surgery, 410011 Changsha, Hunan, China. [Zou, Qiong] Central South University, Third Xiangya Hospital, Department of PathologyChangsha, Hunan, China. [Yuan, Yuan] Central South University, Third Xiangya Hospital, Department of PathologyChangsha, Hunan, China. RP Yang, Z (reprint author), Central South University, The Second Xiangya Hospital, Department of General Surgery, 410011 Changsha, China. EM yangzhulin8@sina.com CR Jemal A, Siegel R,Ward E,HaoY, Xu J,Murray T, ThunMJ, 2008, Cancer statistics, 2008. CA Cancer J Clin 58:71–96 Jayaraman S, JarnaginWR, 2010)Management of gallbladder cancer. Gastroenterol Clin N Am 39:331–342 Ootani T, Shirai Y, Tsukada K, Muto T, 1992, Relationship between gallbladder carcinoma and the segmental type of adenomyomatosis of the gallbladder. Cancer 69:2647–2652 Roa JC, Tapia O, Cakir A, Basturk O, Dursun N, Akdemir D, Saka B, Losada H, Bagci P, Adsay NV, 2011, Squamous cell and adenosquamous carcinomas of the gallbladder: clinicopathological analysis of 34 cases identified in 606 carcinomas. Mod Pathol 24: 1069–1078 Reid KM, Ramos-Dela MA, Donohue JH, 2007, Diagnosis and surgical management of gallbladder cancer: a review. J Gastrointest Surg 11:671–681 Duffy A, Capanu M, Abou-Alfa GK, Huitzil D, Jarnagin W, Fong Y, D'AngelicaM, Dematteo RP, Blumgart LH, O'Reilly EM(2008, Gallbladder cancer, GBC): 10-year experience at Memorial Sloan Kettering Cancer Centre, MSKCC). J Surg Oncol 98:485–489 HawkinsWG, DeMatteo RP, Jarnagin WR, Ben-Porat L, Blumgart LH, Fong Y, 2004, Jaundice predicts advanced disease and early mortality in patients with gallbladder cancer. Ann Surg Oncol 11: 310–315 Cubertafond P, Gainant A, Cucchiaro G, 1994, Surgical treatment of 724 carcinomas of the gallbladder. Results of the French Surgical Association Survey. Ann Surg 219:275–280 Lee SE, Jang JY, Kim SW, Han HS, Kim HJ, Yun SS, Cho BH, Yu HC, Lee WJ, Yoon DS et al, 2014, Surgical strategy for T1 gallbladder cancer: a nationwide multicenter survey in South Korea. Ann Surg Oncol 21:3654–3660 Fairweather M, Balachandran VP, D'Angelica MI, 2016, Surgical management of biliary tract cancers. Chin Clin Oncol 5:63 Chijiiwa K, Nakano K, Ueda J, Noshiro H, Nagai E, Yamaguchi K, TanakaM(2001, Surgical treatment of patients with T2 gallbladder carcinoma invading the subserosal layer. J Am Coll Surg 192:600– 607 Horgan AM, Amir E,Walter T, Knox JJ, 2012, Adjuvant therapy in the treatment of biliary cancer: a systematic review and meta-analysis. J Clin Oncol 30:1934–1940 Sahu S, Sun W, 2017, Targeted therapy in biliary tract cancerscurrent limitations and potentials in the future. J Gastrointest Oncol 8:324–336 MichelG, Tonon T, Scornet D, Cock JM,Kloareg B, 2010, The cell wall polysaccharide metabolism of the brown alga Ectocarpus siliculosus. Insights into the evolution of extracellular matrix polysaccharides in Eukaryotes. New Phytol 188:82–97 Jablonska-Trypuc A, Matejczyk M, Rosochacki S, 2016, Matrix metalloproteinases, MMPs), the main extracellular matrix, ECM, enzymes in collagen degradation, as a target for anticancer drugs. J Enzyme Inhib Med Chem 31(sup1):177–183 Thakur V, Bedogni B, 2016, The membrane tethered matrix metalloproteinase MT1-MMP at the forefront of melanoma cell invasion and metastasis. Pharmacol Res 111:17–22 Vogel W, Gish GD, Alves F, Pawson T, 1997, The discoidin domain receptor tyrosine kinases are activated by collagen. Mol Cell 1:13–23 Sivakumar L, Agarwal G, 2010, The influence of discoidin domain receptor 2 on the persistence length of collagen type I fibers. Biomaterials 31:4802–4808 Herrera-Herrera ML, Quezada-Calvillo R, 2012, DDR2 plays a role in fibroblast migration independent of adhesion ligand and collagen activated DDR2 tyrosine kinase. Biochem Biophys Res Commun 429:39–44 Badiola I, Villace P, Basaldua I, Olaso E, 2011, Down regulation of discoidin domain receptor 2 in A375 human melanoma cells reduces its experimental liver metastasis ability. Oncol Rep 26:971– 978 Badiola I, Olaso E, Crende O, Friedman SL, Vidal-Vanaclocha F, 2012, Discoidin domain receptor 2 deficiency predisposes hepatic tissue to colon carcinoma metastasis. Gut 61:1465–1472 Ren T, Zhang J, Zhang J, Liu X, Yao L, 2013, Increased expression of discoidin domain receptor 2, DDR2): a novel independent prognostic marker of worse outcome in breast cancer patients. Med Oncol 30:397 Sasaki H, Shitara M, Yokota K, Okuda K, Hikosaka Y, Moriyama S, Yano M, Fujii Y, 2012, DDR2 polymorphisms and mRNA expression in lung cancers of Japanese patients. Oncol Lett 4:33–37 Park JW, Lee YS, Kim JS, Lee SK, Kim BH, Lee JA, Lee NO, Kim SH, Hong EK, 2015, Downregulation of discoidin domain receptor 2 decreases tumor growth of hepatocellularcarcinoma. J Cancer Res Clin Oncol 141:197319–197383 Wang YG, Xu L, Jia RR, Wu Q, Wang T, Wei J, Ma JL, Shi M, Li ZS, 2016, DDR2 induces gastric cancer cell activities via activating mTORC2 signaling and is associated with clinicopathological characteristics of gastric cancer. Dig Dis Sci 61:2272–2283 Azemikhah M, Ashtiani HA, Aghaei M, Rastegar H, 2015, Evaluation of discoidin domain receptor-2, DDR2, expression level in normal, benign, and malignant human prostate tissues. Res Pharm Sci 10:356–363 Kim D, Ko P, You E, Rhee S, 2014, The intracellular juxtamembrane domain of discoidin domain receptor 2, DDR2, is essential for receptor activation and DDR2-mediated cancer progression. Int J Cancer 135:2547–2557 Fan Y, Xu Z, Fan J, Huang L, Ye M, Shi K, Huang Z, Liu Y, He L, Huang J,Wang Y, Li Q, 2016, Prognostic significance of discoidin domain receptor 2, DDR2, expression in ovarian cancer. Am J Transl Res 8:2845–2850 Johnson MC, Sangrador-Vegas A, Smith TJ, Cairns MT, 2006, Cloning and characterization of two genes encoding rainbow trout homologues of the IFITM protein family. Vet Immunol Immunopathol 110:357–362 Ogony J, Choi HJ, Lui A, Cristofanilli M, Lewis-Wambi J, 2016, Interferon-induced transmembrane protein 1, IFITM1, overexpression enhances the aggressive phenotype of SUM149 inflammatory breast cancer cells in a signal transducer and activator of transcription 2, STAT2)-dependent manner. Breast Cancer Res 18:25 Lee J, Goh SH, Song N, Hwang JA, Nam S, Choi IJ, Shin A, Kim IH, Ju MH, Jeong JS, Lee YS, 2012, Overexpression of IFITM1 has clinicopathologic effects on gastric cancer and is regulated by an epigenetic mechanism. Am J Pathol 181:43–52 He J, Li J, FengW, Chen L, Yang K, 2015, Prognostic significance of INF-induced transmembrane protein 1 in colorectal cancer. Int J Clin Exp Pathol 8:16007–16013 Kim NH, Sung HY, Choi EN, Lyu D, Choi HJ, Ju W, Ahn JH, 2014, Aberrant DNA methylation in the IFITM1 promoter enhances the metastatic phenotype in an intraperitoneal xenograft model of human ovarian cancer. Oncol Rep 31:2139–2146 Yu F, Ng SS, Chow BK, Sze J, Lu G, PoonWS, Kung HF, LinMC, 2011, Knockdown of interferon-induced transmembrane protein 1, IFITM1, inhibits proliferation, migration, and invasion of glioma cells. J Neuro-Oncol 103:187–195 Kim JY, Kim H, Suk K, Lee WH(2010, Activation of CD147 with cyclophilin a induces the expression of IFITM1 through ERK and PI3K in THP-1 cells. Mediat Inflamm 2010:821940 Deraz EM, Kudo Y, Yoshida M, Obayashi M, Tsunematsu T, Tani H, Siriwardena SB, Keikhaee MR, Qi G, Iizuka S et al, 2011, MMP-10/stromelysin-2 promotes invasion of head and neck cancer. PLoS One 6:e25438 He JD, Luo HL, Li J, Feng WT, Chen LB, 2012, Influences of the interferon induced transmembrane protein 1 on the proliferation, invasion, and metastasis of the colorectal cancer SW480 cell lines. Chin Med J 125:517–522 Kondo M, Dono K, Sakon M, Shimizu J, Nagano H, Nakamori S, Umeshita K,Wakasa K, Monden M, 2002, Adenosquamous carcinoma of the gallbladder. Hepato-Gastroenterology 49:1230–1234 Muzio G, Maggiora M, Paiuzzi E, Oraldi M, Canuto RA, 2012, Aldehyde dehydrogenases and cell proliferation. Free Radic Biol Med 52:735–746 Lemmon MA, Schlessinger J, 2010, Cell signaling by receptor tyrosine kinases. Cell 141:1117–1134 Evans SS, Lee DB, Han T, Tomasi TB, Evans RL, 1990, Monoclonal antibody to the interferon-inducible protein leu-13 triggers aggregation and inhibits proliferation of leukemic B cells. Blood 76:2583–2593 Hatano H, Kudo Y, Ogawa I, Tsunematsu T, Kikuchi A, Abiko Y, Takata T, 2008, IFN-induced transmembrane protein 1 promotes invasion at early stage of head and neck cancer progression. Clin Cancer Res 14:6097–6105 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 157 EP 167 DI 10.1007/s12253-017-0314-3 PG 11 ER PT J AU Darvasi, O Szabo, MP Nemeth, K Szabo, K Spisak, S Liko, I Czirjak, S Racz, K Igaz, P Patocs, A Butz, H AF Darvasi, Otto Szabo, Marton Peter Nemeth, Kinga Szabo, Katalin Spisak, Sandor Liko, Istvan Czirjak, Sandor Racz, Karoly Igaz, Peter Patocs, Attila Butz, Henriett TI Limitations of high throughput methods for miRNA expression profiles in non-functioning pituitary adenomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE miRNA; miRNA profiling; Non-functioning pituitary adenoma ID miRNA; miRNA profiling; Non-functioning pituitary adenoma AB Microarray, RT-qPCR based arrays and nextgeneration- sequencing (NGS) are available high-throughput methods for miRNA profiling (miRNome). Analytical and biological performance of these methods were tested in identification of biologically relevant miRNAs in non-functioning pituitary adenomas (NFPA). miRNome of 4 normal pituitary (NP) and 8 NFPA samples was determined by these platforms and expression of 21 individual miRNAs was measured on 30 (20 NFPA and 10 NP) independent samples. Complex bioinformatics was used. 132 and 137 miRNAs were detected by all three platforms in NP and NFPA, respectively, of which 25 were differentially expressed (fold change > 2). The strongest correlation was observed between microarray and TaqManarray, while the data obtained by NGS were the most discordant despite of various bioinformatics settings. As a technical validation we measured the expression of 21 selected miRNAs by individual RT-qPCR and we were able to validate 35.1%, 76.2%and 71.4%of the miRNAs revealed by SOLiD, TLDA and microarray result, respectively. We performed biological validation using an extended number of samples (20 NFPAs and 8 NPs). Technical and biological validation showed high correlation (p < 0.001; R = 0.96). Pathway and network analysis revealed several common pathways but no pathway showed the same activation score. Using the 25 platform-independent miRNAs developmental pathways were the top functional categories relevant for NFPA genesis. The difference among high-throughput platforms is of great importance and selection of screening method can influence experimental results. Validation by another platform is essential in order to avoid or to minimalize the platform specific errors. C1 [Darvasi, Otto] Hungarian Academy of Sciences and Semmelweis University, Hereditary Endocrine Tumors Research GroupBudapest, Hungary. [Szabo, Marton Peter] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. [Nemeth, Kinga] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Szabo, Katalin] Semmelweis University, 2nd Department of Internal MedicineBudapest, Hungary. [Spisak, Sandor] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. [Liko, Istvan] Hungarian Academy of Sciences and Semmelweis University, Hereditary Endocrine Tumors Research GroupBudapest, Hungary. [Czirjak, Sandor] National Institute of NeurosurgeryBudapest, Hungary. [Racz, Karoly] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. [Igaz, Peter] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. [Patocs, Attila] Hungarian Academy of Sciences and Semmelweis University, Hereditary Endocrine Tumors Research GroupBudapest, Hungary. [Butz, Henriett] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. RP Butz, H (reprint author), Hungarian Academy of Sciences, Molecular Medicine Research Group, Budapest, Hungary. EM butz.henriett@med.semmelweis-univ.hu CR Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T, 2001, Identification of novel genes coding for small expressed RNAs. Science 294:853–858. , DOI 10.1126/science.1064921 Place RF, Li L-C, Pookot D et al, 2008, MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci U S A 105:1608–1613. , DOI 10.1073/pnas. 0707594105 Orom UA, Nielsen FC, Lund AH, 2008, MicroRNA-10a binds the 5’UTR of ribosomal proteinmRNAs and enhances their translation. Mol Cell 30:460–471. , DOI 10.1016/j.molcel.2008.05. 001 Zhang Z, Florez S, Gutierrez-Hartmann A et al, 2010, MicroRNAs regulate pituitary development, and microRNA 26b specifically targets lymphoid enhancer factor 1, Lef-1), which modulates pituitary transcription factor 1, Pit-1, expression. J Biol Chem 285:34718–34728. , DOI 10.1074/jbc.M110.126441 Chen K, Rajewsky N, 2006, Natural selection on human microRNA binding sites inferred from SNP data. Nat Genet 38: 1452–1456. , DOI 10.1038/ng1910 Lewis BP, Burge CB, Bartel DP, 2005, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120:15–20. , DOI 10. 1016/j.cell.2004.12.035 Dolecek TA, Propp JM, Stroup NE, Kruchko C, 2012, CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005-2009. Neuro-Oncol 14(Suppl 5):v1–49. , DOI 10.1093/neuonc/nos218 Dworakowska D, Grossman AB, 2009, The pathophysiology of pituitary adenomas. Best Pract Res Clin Endocrinol Metab 23: 525–541. , DOI 10.1016/j.beem.2009.05.004 Mayson SE, Snyder PJ, 2014, Silent, clinically nonfunctioning, pituitary adenomas. J Neuro-Oncol 117:429–436. , DOI 1007/s11060-014-1425-2 10. SivapragasamM, Rotondo F, Lloyd RVet al, 2011, MicroRNAs in the human pituitary. Endocr Pathol 22:134–143. , DOI 10. 1007/s12022-011-9167-6 Li X-H, Wang EL, Zhou H-M et al, 2014, MicroRNAs in Human Pituitary Adenomas. Int J Endocrinol 2014:435171. , DOI 10.1155/2014/435171 Pritchard CC, Cheng HH, Tewari M, 2012, MicroRNA profiling: approaches and considerations. Nat Rev Genet 13:358–369. https:// doi.org/10.1038/nrg3198 Mestdagh P, Hartmann N, Baeriswyl L et al, 2014, Evaluation of quantitative miRNA expression platforms in the microRNA quality control, miRQC, study. Nat Methods 11:809–815. , DOI 10.1038/nmeth.3014 Git A, Dvinge H, Salmon-Divon M et al, 2010, Systematic comparison of microarray profiling, real-time PCR, and next-generation sequencing technologies for measuring differential microRNA expression. RNA N Y N 16:991–1006. , DOI 10.1261/rna. 1947110 Butz H, Liko I, Czirjak S et al, 2010, Down-regulation of Wee1 kinase by a specific subset of microRNA in human sporadic pituitary adenomas. J Clin Endocrinol Metab 95:E181–E191. https:// doi.org/10.1210/jc.2010-0581 Butz H, Nemeth K, Czenke D et al, 2016, Systematic Investigation of Expression of G2/M Transition Genes Reveals CDC25 Alteration in Nonfunctioning Pituitary Adenomas. Pathol Oncol Res POR. , DOI 10.1007/s12253-016-0163-5 Thompson IR, Chand AN, King PJ et al, 2012, Expression of guanylyl cyclase-B, GC-B/NPR2, receptors in normal human fetal pituitaries and human pituitary adenomas implicates a role for Ctype natriuretic peptide. Endocr Relat Cancer 19:497–508. https:// doi.org/10.1530/ERC-12-0129 Trivellin G, Butz H, Delhove J et al, 2012, MicroRNA miR-107 is overexpressed in pituitary adenomas and inhibits the expression of aryl hydrocarbon receptor-interacting protein in vitro. Am J Physiol Endocrinol Metab 303:E708–E719. , DOI 10.1152/ ajpendo.00546.2011 Harmati M, Tarnai Z, Decsi G et al, 2016, Stressors alter intercellular communication and exosome profile of nasopharyngeal carcinoma cells. J Oral Pathol Med Off Publ Int Assoc Oral Pathol Am Acad Oral Pathol. , DOI 10.1111/jop.12486 Butz H, Liko I, Czirjak S et al, 2011, MicroRNA profile indicates downregulation of the TGFβ pathway in sporadic non-functioning pituitary adenomas. Pituitary 14:112–124. , DOI 10.1007/ s11102-010-0268-x Butz H, Szabo PM, Nofech-Mozes R et al, 2014, Integrative bioinformatics analysis reveals new prognostic biomarkers of clear cell renal cell carcinoma. Clin Chem 60:1314–1326. , DOI 10. 1373/clinchem.2014.225854 Butz H, Szabo PM, Khella HWZ et al, 2015, miRNA-target network reveals miR-124as a key miRNA contributing to clear cell renal cell carcinoma aggressive behaviour by targeting CAV1 and FLOT1. Oncotarget 6:12543–12557. 10.18632/oncotarget.3815 Wang B, Howel P, Bruheim S et al, 2011, Systematic evaluation of three microRNA profiling platforms: microarray, beads array, and quantitative real-time PCR array. PLoS One 6:e17167. https://doi. org/10.1371/journal.pone.0017167 Chevillet JR, Lee I, Briggs HA et al, 2014, Issues and prospects of microRNA-based biomarkers in blood and other body fluids. Mol Basel Switz 19:6080–6105. ht tps: / /doi.org/10.3390/ molecules19056080 Farr RJ, Januszewski AS, Joglekar MVet al, 2015, A comparative analysis of high-throughput platforms for validation of a circulating microRNA signature in diabetic retinopathy. Sci Rep 5:10375. , DOI 10.1038/srep10375 Bottoni A, Zatelli MC, Ferracin M et al, 2007, Identification of differentially expressed microRNAs by microarray: a possible role for microRNA genes in pituitary adenomas. J Cell Physiol 210: 370–377. , DOI 10.1002/jcp.20832 Liang S, Chen L,HuangH, Zhi D(2013, The experimental study of miRNA in pituitary adenomas. Turk Neurosurg 23:721–727. , DOI 10.5137/1019-5149.JTN.7425-12.1 Chambers TJG, Giles A, Brabant G, Davis JRE, 2013, Wnt signalling in pituitary development and tumorigenesis. Endocr Relat Cancer 20:R101–R111. , DOI 10.1530/ERC-13-0005 Elston MS, Gill AJ, Conaglen JV et al, 2008, Wnt pathway inhibitors are strongly down-regulated in pituitary tumors. Endocrinology 149:1235–1242. , DOI 10.1210/en.2007- 0542 Colli LM, Saggioro F, Serafini LN et al, 2013, Components of the canonical and non-canonical Wnt pathways are not mis-expressed in pituitary tumors. PLoS One 8:e62424. , DOI 10.1371/ journal.pone.0062424 Formosa R, Gruppetta M, Falzon S et al, 2012, Expression and clinical significance of Wnt players and survivin in pituitary tumours. Endocr Pathol 23:123–131. , DOI 10.1007/ s12022-012-9197-8 Lebrun J-J, 2009, Activin, TGF-beta and menin in pituitary tumorigenesis. Adv Exp Med Biol 668:69–78 Ruebel KH, Leontovich AA, Tanizaki Y et al, 2008, Effects of TGFbeta1 on gene expression in the HP75 human pituitary tumor cell line identified by gene expression profiling. Endocrine 33:62– 76. , DOI 10.1007/s12020-008-9060-3 Zhenye L, Chuzhong L, Youtu W et al, 2014, The expression of TGF-β1, Smad3, phospho-Smad3 and Smad7 is correlated with the development and invasion of nonfunctioning pituitary adenomas. J Transl Med 12:71. , DOI 10.1186/1479-5876-12-71 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 169 EP 182 DI 10.1007/s12253-017-0330-3 PG 14 ER PT J AU Jain, D Tikku, G Bhadana, P Dravid, Ch Grover, KR AF Jain, Dhruv Tikku, Gargi Bhadana, Pallavi Dravid, Chandrashekhar Grover, Kumar Rajesh TI The Impact of Peritumoral Retraction Clefting & Intratumoral Eosinophils on Overall Survival in Oral Squamous Carcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Carcinoma; Squamous cell; Oral cavity; Overall survival; Tumor-associated tissue eosinophilia; Retraction clefting ID Carcinoma; Squamous cell; Oral cavity; Overall survival; Tumor-associated tissue eosinophilia; Retraction clefting AB This retrospective study aimed to investigate the impact of peritumoral retraction clefts (RC) and tumorassociated tissue eosinophilia (TATE) as predictors of overall survival (OS) in oral squamous cell carcinoma (OSCC) patients. Their relationships with tumor-factors were also examined. Eighty-seven OSCC cases (pTNM: I + II/III + IV; 32/ 55), post-curative surgery, comprised the study cohort. Three observers independently estimated the percent RC semiquantitatively in the selected tumor sections. Additionally, stromal eosinophils were counted in ten consecutive highpower fields of intratumoral and peritumoral regions to evaluate the corresponding TATE. The percent RC ranged between 0% -90% (Mean ± SD: 16 ± 24%; Median: 5%). The stromal eosinophils were greater in peritumoral as compared to intratumoral region. The events of death and tumor recurrence were reached in 16 (18.4%) and 36 (41%) cases respectively. The 3-years OS was 69% [Median OS: 1880 days; Mean follow up: 471(Range; 36–1880) days]. Increased percent RC exhibited relationship with pathologic stage (pTNM III&IV), primary tumor (pT III&IV), tumor depth > 4 mmand categorical tumor recurrence. Additionally, peritumoral eosinophilic infiltrates increased with increasing tumor depths and muscle invasion. Kaplan-Meier curves revealed significantly reduced OS in OSCC cases exhibiting: increased percent RC (>2.5%), mild -moderate/absent intratumoral TATE (versus intense TATE) or categorical tumor recurrence. In subsequent multivariate tests, all the three variables retained significance. Additionally, intraclass correlation coefficient demonstrated acceptable internal consistency for the observers who estimated percent RC. In conclusion, RC and intratumoral TATE proved to be independent predictors of OS in our OSCC cohort. Additionally, increased percent RC pointed towards aggressive tumor behaviour. C1 [Jain, Dhruv] Delhi State Cancer Institute, Department of Oncopathology, Dilshad Garden, 110095 Delhi, India. [Tikku, Gargi] Delhi State Cancer Institute, Department of Oncopathology, Dilshad Garden, 110095 Delhi, India. [Bhadana, Pallavi] Delhi State Cancer Institute, Department of Oncopathology, Dilshad Garden, 110095 Delhi, India. [Dravid, Chandrashekhar] Delhi State Cancer Institute, Department of Surgical Oncology, 110095 Delhi, India. [Grover, Kumar Rajesh] Delhi State Cancer Institute, Department of Clinical Oncology, 110095 Delhi, India. RP Jain, D (reprint author), Delhi State Cancer Institute, Department of Oncopathology, 110095 Delhi, India. EM drdhruv_jain@yahoo.co.in CR McKenney JK, Gomez JA, Desai S, Lee MW, Amin MB, 2001, Morphologic expressions of urothelial carcinoma in situ: a detailed evaluation of its histologic patterns with emphasis on carcinoma in situ with microinvasion. Am J Surg Pathol 25:356–362 Irie J, Manucha V, Ioffe OB, Silverberg SG, 2007, Artefact as the pathologist’s friend: peritumoral retraction in in situ and infiltrating duct carcinoma of the breast. Int J Surg Pathol 15:53–59 Kossard S, Epstein EH Jr, Cerio R et al, 2006, Basal cell carcinoma. In: LeBoit PE, Burg G, Weedon D, Sarasain A, eds, World Health Organization Classification of Tumors. Pathology and Genetics of Skin Tumours, 3rd edn. IARC Press, Lyon, pp 13–19 Kruslin B, Tomas D, Rogatsch H, Novosel I, Cupic H, Belicza M, Kraus O, Mikuz G, 2003, Periacinar retraction clefting in the prostatic needle core biopsies: an important diagnostic criterion or a simple artifact? Virchows Arch 443:524–527 Bell KA, Smith Sehdev AE, Kurman RJ, 2001, Refined diagnostic criteria for implants associated with ovarian atypical proliferative serous tumors, borderline, and micropapillary serous carcinomas. Am J Surg Pathol 25:419–423 Acs G, Khakpour N, Kiluk J, Lee MC, Laronga C, 2015, The presence of Extensive Retraction Clefts in Invasive Breast Carcinomas correlates with lymphatic invasion and nodal metastasis and predicts poor outcome. A prospective validation study of 2742 consecutive cases. Am J Surg Pathol 39:325–337 Acs G, Paragh G, Rakosy Z, Laronga C, Zhang PJ, 2012, The extent of retraction clefts correlates with lymphatic vessel density and VEGF-C expression and predicts nodal metastasis and poor prognosis in early-stage breast carcinoma. Mod Pathol 25:163–177 Acs G, Dumoff KL, Solin LJ, Pasha T, Xu X, Zhang PJ, 2007, Extensive retraction artifact correlates with lymphatic invasion and nodal metastasis and predicts poor outcome in early stage breast carcinoma. Am J Surg Pathol 31:129–140 Tomas D, Spajic B, Milosevic M, Demirovic A, Marusic Z, Kruslin B, 2011, Extensive retraction artifact predicts biochemical recurrence -free survival in prostatic carcinoma. Histopathology 58:447–454 Bujas T, Pavic I, Lenicek T, Mijic A, Kruslin B, Tomas D, 2008, Peritumoral retraction clefting correlates with advanced stage squamous cell carcinoma of the esophagus. Pathol Oncol Res 14:443–447 Pereira MC, Oliveira DT, Kowalski LP, 2011, The role of eosinophils and eosinophil cationic protein in oral cancer: a review. Arch Oral Bio 56:353–358 Jain D, Tikku G, Bhadana P, Dravid C, Grover RK, 2017, A semiquantitativeWorld Health Organization grading scheme evaluating worst tumor differentiation predicts disease-free survival in oral squamous carcinoma patients. Ann Diagn Pathol 29:1–6 Nunnally JC, Bernstein IH, 1994, Psychometric theory. McGraw- Hill, New York Acs G, Paragh G, Chuang ST, Laronga C, Zhang PJ, 2009, The presence of micropapillary features and retraction artifact in core needle biopsy material predicts lymph node metastasis in breast carcinoma. Am J Surg Pathol 33:202–210 Rakosy Z, Paragh G, Acs G, 2010, A distinct gene expression profiling is associated with aggressive breast carcinomas showing prominent retraction artifact. Mod Pathol 23(suppl 1s):67A Gombos Z, Xu X, Chu CS, Zhang PJ, Acs G, 2005, Peritumoral lymphatic vessel density and vascular endothelial growth factor C expression in early stage squamous carcinoma of the uterine cervix. Clin Cancer Res 11:8364–8371 Kruslin B, Tomas D, Cviko A, Cupic H, Odak L, BeliczaM(2006, Periacinar clefting and p63 immunostaining in prostatic intraepithelial neoplasia and prostatic carcinoma. Pathol Oncol Res 12:205–209 Tomas D,Kruslin B, 2004, The potential value of, myo)fibroblastic stromal reaction in the diagnosis of prostatic adenocarcinoma. Prostate 61:324–331 Dorta RG, Landman G, Kowalski LP, Lauris JR, Latorre MR, Oliveira DT, 2002, Tumour-associated tissue eosinophilia as a prognostic factor in oral squamous cel l carcinomas. Histopathology 41:152–157 Goldsmith MM, Cresson DH, Askin FB, 1987, Part II. The prognostic significance of stromal eosinophilia in head and neck cancer. Otolaryngol Head Neck Surg 96:319–324 Goldsmith MM, Belchis DA, Cresson DH, Merritt WD, Askin FB, 1992, The importance of the eosinophil in head and neck cancer. Otolaryngol Head Neck Surg 106:27–33 Wong DT, Bowen SM, Elovic A, Gallagher GT, Weller PF, 1999, Eosinophil ablation and tumor development. Oral Oncol 35:496–501 Horiuchi K, Mishima K, Ohsawa M, Sugimura M, Aozasa K, 1993, Prognostic factors for well-differentiated squamous cell carcinoma in the oral cavity with emphasis on immunohistochemical evaluation. J Surg Oncol 53:92–96 Tadbir AA, Ashraf MJ, Sardari Y, 2009, Prognostic significance of stromal eosinophilic infiltration in oral squamous cell carcinoma. J Craniofac Surg 20:287–289 Oliveira DT, Tjioe KC, Assao A, Faustino SES, Carvalho AL, Landman G, Kowalski LP, 2009, Tissue eosinophilia and its association with tumoral invasion of oral cancer. Int J Surg Pathol 17: 244–249 Lundqvist L, Stenlund H, Laurell G, Nylander K, 2012, The importance of stromal inflammation in squamous cell carcinoma of the tongue. J Oral Pathol Med 41:379–383 Simson L, Ellyard JI, Dent LA, Matthaei KI, Rothenberg ME, Foster PS, Smyth MJ, Parish CR, 2007, Regulation of carcinogenesis by IL-5 and CCL11: a potential role for eosinophils in tumor immune surveillance. J Immunol 178:4222–4229 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 183 EP 189 DI 10.1007/s12253-017-0328-x PG 7 ER PT J AU Afsharpad, M Nowroozi, RM Mobasheri, BM Ayati, M Nekoohesh, L Saffari, M Zendehdel, K Modarressi, HM AF Afsharpad, Mandana Nowroozi, Reza Mohammad Mobasheri, Beigom Maryam Ayati, Mohsen Nekoohesh, Leila Saffari, Mojtaba Zendehdel, Kazem Modarressi, Hossein Mohammad TI Cancer-Testis Antigens as New Candidate Diagnostic Biomarkers for Transitional Cell Carcinoma of Bladder SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer testis antigen; Clinical markers; Transitional cell carcinoma; Urinary bladder neoplasms ID Cancer testis antigen; Clinical markers; Transitional cell carcinoma; Urinary bladder neoplasms AB To evaluate the diagnostic potential of 23 candidate genes, belonging to a category of tumor-specific antigens known as cancer-testis antigens (CTAs), in transitional cell carcinoma (TCC) patients. The expression of 16 known candidate CTAs and seven testis restricted/selective genes, predominantly expressed in the testis, was evaluated by reverse transcriptase-polymerase chain reaction (RT-PCR). Urinary exfoliated cells (UECs) and cancerous tissues of 73 TCC patients were used as cases, while 25 tumor-free adjacent bladder tissue specimens along with bladder tissue specimens and UECs of five non-TCC individuals were analyzed as controls. Among the known CTAs only MAGEA3, MAGEB4, TSGA10, PIWIL2, OIP5, and ODF4 were expressed specifically in TCC tissues and UEC samples. ACTL7A, AURKC, and CGB2 were testis-restricted/selective genes that indicated specific expression in cases in comparison to controls. MAGEA3, MAGEB4, and ODF4 mRNA was detectable in more than 50% of both TCC tissues, and UEC samples. Slight differences were detected in the mRNA expression pattern of candidate genes between the UEC samples and tumor tissues. Different panels formed by combinations of these genes can show up to 95.9% and 94.5% of positivity in TCC tissues and UEC samples, respectively, suggesting their diagnostic and surveillance potential. Meanwhile the RT-PCR assay of at least MAGEA3, MAGEB4, and ODF4 may be particularly useful for diagnostic and surveillance of TCC in the form of a multi-biomarker panel. C1 [Afsharpad, Mandana] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Nowroozi, Reza Mohammad] Tehran University of Medical Sciences, Uro-Oncology Research CenterTehran, Iran. [Mobasheri, Beigom Maryam] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Ayati, Mohsen] Tehran University of Medical Sciences, Uro-Oncology Research CenterTehran, Iran. [Nekoohesh, Leila] Tehran University of Medical Sciences, School of Advanced Technologies in Medicine, International Campus, Department of Medical BiotechnologyTehran, Iran. [Saffari, Mojtaba] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Zendehdel, Kazem] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Modarressi, Hossein Mohammad] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. RP Modarressi, HM (reprint author), Tehran University of Medical Sciences, Cancer Institute of Iran, Tehran, Iran. EM modaresi@tums.ac.ir CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108 Masoompour SM, Yarmohammadi H, Rezaianzadeh A, Lankarani KB, 2011, Cancer incidence in southern Iran, 1998–2002: results of population-based cancer registry. Cancer Epidemiol 35(5):e42–e47 Babjuk M, Burger M, Zigeuner R, Shariat SF, van Rhijn BWG, Comperat E, Sylvester RJ, Kaasinen E, Bohle A, Palou Redorta J, Roupret M, 2013, EAU guidelines on non–muscle-invasive urothelial carcinoma of the bladder: update 2013. Eur Urol 64(4): 639–653. , DOI 10.1016/j.eururo.2013.06.003 Lotan Y, Svatek RS, Malats N, 2008, Screening for bladder cancer: a perspective. World J Urol 26:13–18 Turco P, Houssami N, Bulgaresi P, Troni GM, Galanti L, Cariaggi MP, Cifarelli P, Crocetti E, Ciatto S, 2011, Is conventional urinary cytology still reliable for diagnosis of primary bladder carcinoma? Accuracy based on data linkage of a consecutive clinical series and cancer registry. Acta Cytol 55(2):193–196. , DOI 10.1159/ 000320861 Simpson AJ, Caballero OL, Jungbluth A, Chen YT, Old LJ, 2005, Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer 5(8):615–625. , DOI 10.1038/nrc1669 Caballero OL, Chen YT, 2009, Cancer/testis, CT, antigens: potential targets for immunotherapy. Cancer Sci 100(11):2014–2021. , DOI 10.1111/j.1349-7006.2009.01303.x8 Scanlan MJ, Gordon CM, Williamson B, Lee SY, Chen YT, Stockert E, Jungbluth A, Ritter G, Jager D, Jager E, 2002, Identification of cancer/testis genes by database mining and mRNA expression analysis. Int J Cancer 98(4):485–492 Scanlan MJ, Simpson AJ, Old LJ, 2004, The cancer/testis genes: review, standardization, and commentary. Cancer Immun 4:1 Sharma P, Shen Y, Wen S, Bajorin DF, Reuter VE, Old LJ, Jungbluth AA, 2006, Cancer-testis antigens: expression and correlation with survival in human urothelial carcinoma. Clin Cancer Res 12(18):5442–5447. , DOI 10.1158/1078–0432.ccr- 06-0527 Zamuner FT, Karia BT, de Oliveira CZ, Santos CR, Carvalho AL, Vettore AL, 2015, A comprehensive expression analysis of cancer testis antigens in head and neck squamous cell carcinoma revels MAGEA3/6 as a marker for recurrence. Mol Cancer Ther 14(3): 828–834. , DOI 10.1158/1535–7163.mct-14-0796 Bellati F, Napoletano C, Tarquini E, Palaia I, Landi R, Manci N, Spagnoli G, Rughetti A, Panici PB, Nuti M, 2007, Cancer testis antigen expression in primary and recurrent vulvar cancer: association with prognostic factors. Eur J Cancer 43(17):2621–2627. , DOI 10.1016/j.ejca.2007.08.031 Bar-Haim E, PazA, Machlenkin A, Hazzan D, Tirosh B, Carmon L, Brenner B, Vadai E, Mor O, Stein A, 2004)MAGE-A8 overexpression in transitional cell carcinoma of the bladder: identification of two tumour-associated antigen peptides. Br J Cancer 91(2):398– 407 Fradet Y, Picard V, Bergeron A, LaRue H, 2005, Cancer-testis antigen expression in bladder cancer. Prog Urol 15(6 Suppl 1): 1303–1313 Picard V, Bergeron A, LarueH, FradetY(2007)MAGE-A9mRNA and protein expression in bladder cancer. Int J Cancer 120(10): 2170–2177. , DOI 10.1002/ijc.22282 Bergeron A, Picard V, LaRue H, Harel F, Hovington H, Lacombe L, Fradet Y, 2009, High frequency of MAGE-A4 and MAGE-A9 expression in high-risk bladder cancer. Int J Cancer 125(6):1365– 1371 Dyrskjot L, Zieger K, Kissow Lildal T, Reinert T, Gruselle O, Coche T, Borre M, Orntoft TF, 2012, Expression of MAGE-A3, NY-ESO-1, LAGE-1 and PRAME in urothelial carcinoma. Br J Cancer 107(1):116–122. , DOI 10.1038/bjc.2012.215 Yin B, Liu G, Wang XS, Zhang H, Song YS, Wu B, 2012, Expression profile of cancer-testis genes in transitional cell carcinoma of the bladder. Urol Oncol 30(6):886–892. , DOI 10. 1016/j.urolonc.2010.08.017 Mengus C, Schultz-Thater E, Coulot J, Kastelan Z, Goluza E, Coric M, Spagnoli GC, Hudolin T, 2013, MAGE-A10 cancer/testis antigen is highly expressed in high-grade non-muscle-invasive bladder carcinomas. Int J Cancer 132(10):2459–2463. , DOI 10. 1002/ijc.27914 Kawai T, Enomoto Y, Morikawa T, Matsushita H, Kume H, Fukayama M, Yamaguchi H, Kakimi K, Homma Y, 2014, High expression of heat shock protein 105 predicts a favorable prognosis for patients with urinary bladder cancer treated with radical cystectomy. Mol Clin Oncol 2(1):38–42. , DOI 10.3892/ mco.2013.203 Hofmann O, Caballero OL, Stevenson BJ, Chen YT, Cohen T, Chua R, Maher CA, Panji S, Schaefer U, Kruger A, Lehvaslaiho M, Carninci P, Hayashizaki Y, Jongeneel CV, Simpson AJ, Old LJ, HideW(2008, Genome-wide analysis of cancer/testis gene expression. Proc Natl Acad Sci U S A 105(51):20422–20427. https://doi. org/10.1073/pnas.0810777105 Eble J, Sauter G, Epstein J, Sesterhenn I, 2004, World Health Organization classification of tumors. Tumors of the urinary system and genital organs. IARC Press, Lyon, Pathology and Genetics Patard JJ, Brasseur F, Gil-Diez S, Radvanyi F, Marchand M, Francois P, Abi-Aad A, Van Cangh P, Abbou CC, Chopin D et al, 1995, Expression of MAGE genes in transitional-cell carcinomas of the urinary bladder. Int J Cancer 64(1):60–64 Katayama H, Brinkley WR, Sen S, 2003, The Aurora kinases: role in cell transformation and tumorigenesis. Cancer Metastasis Rev 22(4):451–464 Wang J, Liu X, Dou Z, Chen L, Jiang H, Fu C, Fu G, Liu D, Zhang J, Zhu T, 2014, Mitotic regulator Mis18β interacts with and specifies the centromeric assembly of molecular chaperone holliday junction recognition protein, HJURP). J Biol Chem 289(12):8326–8336 Mansouri K, Mostafie A, Rezazadeh D, Shahlaei M, Modarressi MH, 2016, New function of TSGA10 gene in angiogenesis and tumor metastasis: a response to a challengeable paradox. Hum Mol Genet 25(2):233–244 Yang B, O'Herrin SM, Wu J, Reagan-Shaw S, Ma Y, Bhat KM, Gravekamp C, Setaluri V, Peters N, Hoffmann FM, 2007, MAGEA, mMage-b, and MAGE-C proteins form complexes with KAP1 and suppress p53-dependent apoptosis inMAGE-positive cell lines. Cancer Res 67(20):9954–9962 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 191 EP 199 DI 10.1007/s12253-017-0313-4 PG 9 ER PT J AU Zejun, D Kun, Y Dehong, L Xueling, Q AF Zejun, Duan Kun, Yao Dehong, Lu Xueling, Qi TI Proximal-Type Epithelioid Sarcoma in Skull Base: a Pathological Diagnosis Challenge with Other Intracranial Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Proximal type epithelioid sarcoma; Middle cranial fossa; Skull base; Atypical teratoid/rhabdoid tumour ID Proximal type epithelioid sarcoma; Middle cranial fossa; Skull base; Atypical teratoid/rhabdoid tumour AB Proximal-type ES (PES) is a rare and aggressive sarcoma originated from soft tissues with uncertain differentiation. It mainly affects middle-aged patients and often locates in proximal extremity and deep-seated tissues. Only one case of PES located in the skull base has been reported to date. Herein, we report two cases of PES occurred in the middle cranial fossa in two middle-aged Chinese women. Microscopically, the tumors were consisted of epithelial-like cells with or without rhabdoid cells. And frequent mitotic activity and coagulation necrosis were present. Immunohistochemically, tumor cells in the two cases were positive for vimentin, AE1/AE3, epithelial membrane antigen (EMA), CD34, and synaptophysin. A few number of tumor cells expressed CD56. They were completely negative for integrase interactor-1 (INI1). Besides, TP53 positive cells were observed (>50%) in the two cases. The MIB-1 proliferation index was high up to 50–70%. Fluorescence in situ hybridization showed the monoallelic deletions of INI1. Intracranial PES is needed to identify with other mimic tumors, especially rhabdoid meningioma, epithelioid MPNST and adult AT/RT. The prognosis of the two patients was very poor. They died respectively less than a month and half a month after surgery. Tumor grew rapidly and was easy to infiltrate into the surrounding tissues. It may suggest that the prognosis of PES occurred at the base of skull was worse than in other sites. C1 [Zejun, Duan] Capital Medical University, San Bo Brain Hospital, Department of PathologyBeijing, China. [Kun, Yao] Capital Medical University, San Bo Brain Hospital, Department of Pathology, Haidian DistrictBeijing, China. [Dehong, Lu] Capital Medical University, San Bo Brain Hospital, Department of Pathology, Haidian DistrictBeijing, China. [Xueling, Qi] Capital Medical University, San Bo Brain Hospital, Department of PathologyBeijing, China. RP Xueling, Q (reprint author), Capital Medical University, San Bo Brain Hospital, Department of Pathology, Beijing, China. EM xszqxl169@163.com CR Enzinger FM, 1970, Epithelioid sarcoma. A sarcoma simulating a granuloma or a carcinoma. Cancer 26:1029–1041 Chase DK, Enzinger FM, 1985, Epithelioid sarcoma. Diagnosis, prognostic indicators, and treatment. Am J Surg Pathol 9:241–263 Bos GD, Pritchard DJ, Reiman HM et al, 1988, Epithelioid sarcoma. An analysis of fifty-one cases. Bone and Joint Surg 70A:862– 870 Guillou L, Wadden C, Coindre JM et al, 1997, BProxim al-type^ epithelioid sarcoma, a distinctive aggressive neop lasm showing rhabdoid features. Clinicopathologic, immunohistochemical and ultrastructural study of a series. Am J Surg Pathol 21(2):130–146 Fletcher CDM, Unni KK, Mertens F, 2002)WHO classification of tumors: pathology and genetics of tumors of soft tissue and bone. Hernational Agency for Research on Cancer, IARC, Press, Lyon, pp 205–207 dos Santos LM, Nogueira L,Matsuo CYet al, 2013, Proximal-type epithelioid sarcoma - case report. An Bras Dermatol 88(3):444–447 Guzzetta AA, Montgomery EA, Lyu H et al, 2012, Epithelioid sarcoma: one institution’s experience with a rare sarcoma. J Surg Res 117:116–122 Poisson JL, Rubinas TC, 2010, Proximal-type epithelioid sarcoma. Labmedicine 41(8):467–470 Argenta PA, Thomas S, Chura JC, 2007, BProximal-type^ epithelioid sarcoma vs. malignant rhabdoid tumor of the vulva: a case report, review of the literature, and an argument for consolidation. Gynecol Oncol 107:130–135 Hasegawa T, Matsuno Y, Shimoda T, 2001, Proximal-type epithelioid sarcoma: a clinicopathologic study of 20 cases. Mod Pathol 14(7):655–663 Zevallos-Giampietri E-A, Barrionuevo C, 2005, Proximal-type epithelioid sarcoma: report of two cases in the perineum: differential diagnosis and review of soft tissue tumors with epithelioid and/or rhabdoid features. Immunohistochem Mol Morphol 13(3):221–230 Tholpady A, Lonergan CL,WickMR(2010, Proximal-type epithelioid sarcoma of the vulva: relationship to malignant extrarenal rhabdoid tumor. Int J Gynecol Pathol 29(6):600–604 Rekhi B, Gorad BD, Chinoy RF, 2007, Proximal-type epithelioid sarcoma- a rare, aggressive subtype of epithelioid sarcoma presenting as a recurrent perineal mass in a middle-aged male [J]. World J Surg Oncol 5:28 Thway K, Jones RL, Noujaim J et al, 2016, Epithelioid sarcoma: diagnostic features and genetics. Adv Anat Pathol 23:41–49 LianDWQ, Lee HY, Tan KK et al, 2010, Proximal type epithelioid sarcoma arising in the base of the skull: a diagnostic challenge [J]. Clin Pathol 63:472–474 Sav A, Rotondo F, Syro LV et al, 2015, Invasive, atypical and aggressive pituitary adenomas and carcinomas [J]. Endocrinol Metab Clin 44(1):99–104 Rosenblum MK, 2007, The 2007 WHO classification of nervous system tumors: newly recognized members of the mixed glioneuronal group [J]. Brain Pathol 17:147–149 Kreiger P, Judkins A, Russo P et al, 2009, Loss of INI1 expression defines a unique subset of pediatric undifferentiated soft tissue sarcomas [J]. Mod Pathol 22:142–150 Rakheja D,WilsonKS, Meehan J et al, 2005, BProx imal-type^ and classic epithelioid sarcom as represent a clinicopathologic continuum: case report [J]. Pediatr Dev Pathol 8(1):105–114 Sur M, Nayler SJ, 2001, Proximal epithelioid sarcoma amisnomer[J]. Histopathology 39:641–643 Kohashi K, Izumi T, Oda Y et al, 2009, Infrequent SMARCB1/ INI1 gene alteration in epithelioid sarcoma: a useful tool in distinguishing epithelioid sarcoma from malignant rhabdoid tumor [J]. Hum Pathol 40:349–355 Hornick J, Dal Cin P, Fletcher C, 2009, Loss of INI1 expression is characteristic of both conventional and proximal-type epithelioid sarcoma [J]. Am J Surg Pathol 33:542–550 Hornick JL, Paola DC, Fletcher CDM, 2008, Loss of INI1 expression is characteristic of both conventional and proximal-type epithelioid sarcoma [J]. Am J Surg Pathol 33(4):542–550 Modena P, Lualdi E, Facchinetti F et al, 2005, SMARCB1/INI1 tumor suppressor gene is frequently inactivated in epithelioid sarcomas [J]. Cancer Res 65:4012–4019 Hollmann TJ, Hornick JL, 2011, INI1-deficient tumors: diagnostic features and molecular genetics. Am J Surg Pathol 35(10):e47–e63 Sapi Z, Papp G, Szendroi M et al, 2016, Epigenetic regulation of SMARCB1 By miR-206, −381 and −671-5p is evident in a variety of SMARCB1 immunonegative soft tissue sarcomas, while miR- 765 appears specific for epithelioid sarcoma. A miRNA study of 223 soft tissue sarcomas. Genes Chromosomes Cancer 55(10):786– 802 Rosenblum MK, 2007, The 2007 WHO classification of nervous system tumors: newly recognized members of the mixed glioneuronal group [J]. Brain Pathol 17:163–172 Perry A, Fuller CE, Judkins AR et al, 2005, INI1 expression is retained in composite rhabdoid tumors, including rhabdoid meningiomas. Mod Pathol 18:951–958 Samaras V, Stamatelli A, Samaras E et al, 2009, Atypical teratoid/ rhabdoid tumor of the central nervous system in an 18-year-old patient [J]. Clin Neuropathol 28:1–10 Thway K, Fisher C, 2014, Malignant peripheral nerve sheath tumor: pathology and genetics. Ann Diagn Pathol 18:109–116 LaskinWB,Weiss SW, Bratthauer GL, 1991, Epithelioid variant of malignant peripheral nerve sheath tumor, Malignant Epithelioid Schwannoma). Am J Surg Pathol 15(12):1136–1145 Karkavelas G, Petrakis G, Beretouli E et al, 1999, Epithelioid malignant peripheral nerve sheath tumor: report of four cases. Apmis Acta Pathologica Microbiologica Et Immunogica scandinavica 107(4):401–403 Wolf PS, Flum DR, TanasMR et al, 2008, Epithelioid sarcoma: the University of Washington experience [J]. Am J Surg 96:407–412 Guzzetta AA, Montgomery EA, Lyu H et al, 2012, Epithelioid sarcoma: one institution’s experience with a rare sarcoma. J Surg Res 77:116–122 Gasparini P, Facchinetti F, Boeri M et al, 2011, Prognostic determinants in epithelioid sarcoma. Eur J Cancer 47:287–295 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 201 EP 208 DI 10.1007/s12253-017-0318-z PG 8 ER PT J AU Seyedin, NS Mitchell, LD Mott, LS Russo, K Tracy, RCh Snow, NA Parkhurst, RJ Smith, CM Buatti, MJ Watkins, MJ AF Seyedin, N Steven Mitchell, L Darrion Mott, L Sarah Russo, J. Kyle Tracy, R Chad Snow, N Anthony Parkhurst, R Jessica Smith, C Mark Buatti, M John Watkins, M John TI Is More Always Better? An Assessment of the Impact of Lymph Node Yield on Outcome for Clinically Localized Prostate Cancer with Low/Intermediate Risk Pathology (pT2-3a/pN0) Managed with Prostatectomy Alone SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Radical prostatectomy; Lymph node dissection; Localized prostate cancer; Biochemical relapse free survival ID Radical prostatectomy; Lymph node dissection; Localized prostate cancer; Biochemical relapse free survival AB The clinical impact of lymph node dissection extent remains undetermined in the contemporary setting, as reflected in care pattern variations. Despite some series demonstrating a direct relationship between number of lymph nodes identified and detection of nodal involvement, the correlation between lymph node yield and disease control or survival outcomes remains unclear. Patients with clinically localized prostate cancer, pre-RP PSA <30, and pT2-3a/N0 disease at RP were retrospectively identified from two databases for inclusion. Those who received pre- or post-RP radiotherapy or hormone therapy were excluded. Kaplan-Meier method was employed for survival probability estimation. Cox regression models were used to assess bRFS differences between subsets. From 2002 to 2010, 667 eligible patients were identified. The median age was 61 yrs. (range, 43–76), with median PSA 5.6 ng/dL (0.9–28.0). At RP, most patients had pT2c (64%) disease with Gleason Score (GS) ≤6 (43%) or 7 (48%); 218 (33%) patients had positive margins (M+). At median clinical and PSA follow-up of 96 and 87 months, respectively, 146 patients (22%) experienced PSA failure with an estimated bRFS of 81%/76% at 5/8 years. For patients who underwent LND, univariable analysis identified PSA (at diagnosis), higher GS (≥7, at biopsy or RP), intermediate/high risk stratification, M+ as adversely associated with bRFS (all p < 0.01). A higher number of LNs excised was not associated with improved bRFS for the entire cohort (HR = 0.97, p = 0.27), nor for any clinical risk stratum, biopsy GS, or RP GS subgroup. This study did not demonstrate an association between LN yield and bRFS in patients with clinically localized pT2- 3a/pN0 prostate cancer managed with RP alone, either in the entire population or with substratification by clinical risk stratum or GS. C1 [Seyedin, N Steven] University of Iowa, Carver School of Medicine, Department of Radiation Oncology, 200 Hawkins Drive, 52242 Iowa City, IA, USA. [Mitchell, L Darrion] The Ohio State University, Department of Radiation OncologyColumbus, OH, USA. [Mott, L Sarah] University of Iowa, Holden Comprehensive Cancer CenterIowa City, IA, USA. [Russo, J. Kyle] Bismarck Cancer CenterBismarck, ND, USA. [Tracy, R Chad] University of Iowa, Department of UrologyIowa City, IA, USA. [Snow, N Anthony] University of Iowa, Department of PathologyIowa City, IA, USA. [Parkhurst, R Jessica] University of Iowa, Carver School of Medicine, Department of Radiation Oncology, 200 Hawkins Drive, 52242 Iowa City, IA, USA. [Smith, C Mark] University of Iowa, Carver School of Medicine, Department of Radiation Oncology, 200 Hawkins Drive, 52242 Iowa City, IA, USA. [Buatti, M John] University of Iowa, Carver School of Medicine, Department of Radiation Oncology, 200 Hawkins Drive, 52242 Iowa City, IA, USA. [Watkins, M John] University of Iowa, Carver School of Medicine, Department of Radiation Oncology, 200 Hawkins Drive, 52242 Iowa City, IA, USA. RP Watkins, MJ (reprint author), University of Iowa, Carver School of Medicine, Department of Radiation Oncology, 52242 Iowa City, USA. EM john.m.watkins.md@hotmail.com CR Ferlay JSI, Ervik M, Dikshit R, et al. GLOBOCAN 2012 v1.0, Cancer incidence and mortality worldwide: IARC CancerBase No 11 [Internet] Lyon, France: International Agency for Research on Cancer; 2013. Available from: http://globocan.iarc.fr, accessed on 01/11/2017 Lu-Yao GL, Greenberg ER, 1994, Changes in prostate cancer incidence and treatment in USA. Lancet 343:251–254 Abdollah F, Suardi N, GallinaAet al, 2013, Extended pelvic lymph node dissection in prostate cancer: a 20-year audit in a single center. Ann Oncol 24:1459–1466 Briganti A, Chun FK, Salonia A et al, 2007, Critical assessment of ideal nodal yield at pelvic lymphadenectomy to accurately diagnose prostate cancer nodal metastasis in patients undergoing radical retropubic prostatectomy. Urology 69:147–151 Heidenreich A, Ohlmann CH, Polyakov S, 2007, Anatomical extent of pelvic lymphadenectomy in patients undergoing radical prostatectomy. Eur Urol 52:29–37 Bhatta-Dhar N, Reuther AM, Zippe C, Klein EA, 2004, No difference in six-year biochemical failure rates with or without pelvic lymph node dissection during radical prostatectomy in low-risk patients with localized prostate cancer. Urology 63:528–531 Allaf ME, Palapattu GS, Trock BJ, Carter HB, Walsh PC, 2004, Anatomical extent of lymph node dissection: impact on men with clinically localized prostate cancer. J Urol 172:1840–1844 Kluth LA, Xylinas E, Rieken M et al, 2014, Does increasing the nodal yield improve outcomes in contemporary patients without nodal metastasis undergoing radical prostatectomy? Urol Oncol 32:47.e1–47.e8 Epstein JI, Srigley J, Grignon D, Humphrey P, Association of Directors of A, Surgical P, 2008, Recommendations for the reporting of prostate carcinoma: Association of Directors of Anatomic and Surgical Pathology. Am J Clin Pathol 129:24–30 D’Amico AV, Whittington R, Malkowicz SB et al Pretreatment nomogram for prostate-specific antigen recurrence after radical prostatectomy or external-beam radiation therapy for clinically localized prostate cancer. J Clin Oncol 17:168–172 Masterson TA, Bianco FJ, Vickers AJ et al, 2006, The association between total and positive lymph node counts, and disease progression in clinically localized prostate cancer. J Urol 175:1320–1324 Joslyn SA, Konety BR, 2006, Impact of extent of lymphadenectomy on survival after radical prostatectomy for prostate cancer. Urology 68:121–125 Schreiber D, Rineer J, Sura S et al, 2011, Radical prostatectomy for cT3-4 disease: an evaluation of the pathological outcomes and patterns of care for adjuvant radiation in a national cohort. BJUInt 108: 360–365 Gandaglia G, Trinh QD, Hu JC et al, 2014, The impact of robotassisted radical prostatectomy on the use and extent of pelvic lymph node dissection in the "post-dissemination" period. Eur J Surg Oncol 40:1080–1086 Berglund RK, Sadetsky N, DuChane J, Carroll PR, Klein EA, 2007, Limited pelvic lymph node dissection at the time of radical prostatectomy does not affect 5-year failure rates for low, intermediate and high risk prostate cancer: results from CaPSURE. J Urol 177:526–529 Murphy AM, Berkman DS, Desai M, McKiernan JM, Badani KK, 2009, The number of negative pelvic lymph nodes removed does not affect the risk of biochemical failure after radical prostatectomy. BJU Int 105:176–179 Schiavina R, Manferrari F, Garofalo M et al, 2010, The extent of pelvic lymph node dissection correlates with the biochemical recurrence rate in patients with intermediate- and high-risk prostate cancer. BJU Int 108:1262–1268 Abdollah F, Sun M, Thuret R et al, 2012, Lymph node count threshold for optimal pelvic lymph node staging in prostate cancer. Int J Urol 19:645–651 Brawley OW, 2012, Trends in prostate cancer in the United States. J Nat Cancer Inst Monogr 2012:152–156 Briganti A, Blute ML, Eastham JH et al, 2009, Pelvic lymph node dissection in prostate cancer. Eur Urol 55:1251–1265 DiMarco DS, Zincke H, Sebo TJ et al, 2005, The extent of lymphadenectomy for pTXN0 prostate cancer does not affect prostate cancer outcome in the prostate specific antigen era. J Urol 173: 1121–1125 Joniau S, Van den Bergh L, Lerut E et al, 2013, Mapping of pelvic lymph node metastases in prostate cancer. Eur Urol 63:450–458 Pagliarulo V, Hawes D, Brands FH et al, 2006, Detection of occult lymph node metastases in locally advanced node-negative prostate cancer. J Clin Oncol 24:2735–2742 Maurer T, Gschwend JE, Rauscher I et al, 2016, Diagnostic efficacy of, 68)Gallium-PSMA positron emission tomography compared to conventional imaging for lymph node staging of 130 consecutive patients with intermediate to high risk prostate cancer. J Urol 195: 1436–1443 Mattei A, Fuechsel FG, Dhar NB et al, 2008, The template of the primary lymphatic landing sites of the prostate should be revisited: results of a multimodality mapping study. Eur Urol 53:118–125 Grivas N,Wit E, Pos F et al, 2017, Sentinel lymph node dissection to select clinically node-negative prostate cancer pateitns for pelvic radiation therapy: effect on biochemical and systemic progression. Int J Radiat Oncol Biol Phys 97:347–354 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 209 EP 215 DI 10.1007/s12253-017-0349-5 PG 7 ER PT J AU Felkai, L Banusz, R Kovalszky, I Sapi, Z Garami, M Papp, G Karaszi, K Varga, E Csoka, M AF Felkai, Luca Banusz, Rita Kovalszky, Ilona Sapi, Zoltan Garami, Miklos Papp, Gergo Karaszi, Katalin Varga, Edit Csoka, Monika TI The Presence of ALK Alterations and Clinical Relevance of Crizotinib Treatment in Pediatric Solid Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ALK; Crizotinib; Soft tissue sarcoma; Neuroblastoma; Inflammatory myofibroblastic tumor ID ALK; Crizotinib; Soft tissue sarcoma; Neuroblastoma; Inflammatory myofibroblastic tumor AB Soft tissue sarcomas (STS) and neuroblastomas (NBL), are childhood malignancies still associated with poor prognoses despite the overall improvement in childhood tumor survival of the past decades. Anaplastic lymphoma kinase (ALK) inhibition is promising new strategy to improve the outcome of these pediatric tumors. Eighteen histologic samples of pediatric STS and 19 NBL patients were analyzed for ALK abnormalities using fluorescent in situ hybridization (FISH) with break-apart probes and immunohistochemistry (IHC). ALK alterations were presented in 20 of the 37 sections. The presence of ALK alteration in NBL samples were detected using IHC in 84,2% of all cases compared to 21,1% FISH positivity. In STS cases the results were less different (IHC 16,7%vs FISH 22,2%). The difference can be explained by the different type of molecular alterations. FISH method detected translocation and amplification, but not the point mutation of ALK gene. IHC confirmed the diagnosis by detecting the expression of ALK protein.After ALK positivity was proven, the effectiveness and safety of the crizotinib therapy was examined in 4 patients (1 alveolar rhabdomyosarcoma (RMA), 1 embryonal rhabdomyosarcoma (RME), 1 inflammatory myofibroblastic tumor (IMT), 1 NBL). We observed continuous remission of the IMT patient, all other cases the inhibitor treatment was not curative.Our findings underline the importance of screening the ALK status parallel with both IHC and FISH. Crizotinib treatment had a long-term effect in ALK positive IMT patients, however itwas only temporary efficient in relapsed, progressive STS and NBL. C1 [Felkai, Luca] Semmelweis University, 2nd Department of Pediatrics, Tuzolto u. 7-9, H-1094 Budapest, Hungary. [Banusz, Rita] Semmelweis University, 2nd Department of Pediatrics, Tuzolto u. 7-9, H-1094 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Garami, Miklos] Semmelweis University, 2nd Department of Pediatrics, Tuzolto u. 7-9, H-1094 Budapest, Hungary. [Papp, Gergo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Karaszi, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Varga, Edit] Semmelweis University, 2nd Department of Pediatrics, Tuzolto u. 7-9, H-1094 Budapest, Hungary. [Csoka, Monika] Semmelweis University, 2nd Department of Pediatrics, Tuzolto u. 7-9, H-1094 Budapest, Hungary. RP Csoka, M (reprint author), Semmelweis University, 2nd Department of Pediatrics, H-1094 Budapest, Hungary. EM dr.csoka.monika@gmail.com CR MorrisSWet al, 1995, Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma. Science 267: 316–317 Kruczynski A, Delsol G, Laurent C, Brousset P, Lamant L, 2012, Anaplastic lymphoma kinase as a therapeutic target. Expert Opin Ther Targets 16:1127–1138 Mosse YP,Wood A, Maris JM, 2009, Inhibition of ALK signaling for cancer therapy. Clin Cancer Res 15:5609–5614 Palmer RH, Vernersson E, Grabbe C, Hallberg B, 2009, Anaplastic lymphoma kinase: signalling in development and disease. Biochem J 420:345–361 Hallberg B, Palmer RH, 2013, Mechanistic insight into ALK receptor tyrosine kinase in human cancer biology. Nat Rev Cancer 13: 685–700 Murakami Y, Mitsudomi T, Yatabe Y, Screening Method A, 2012, For the ALK fusion gene in NSCLC. Front Oncol 2:24 Selinger CI et al, 2013, Testing for ALK rearrangement in lung adenocarcinoma: a multicenter comparison of immunohistochemistry and fluorescent in situ hybridization. Mod Pathol 26:1545–1553 Li XQ, Hisaoka M, Shi DR, Zhu XZ, Hashimoto H, 2004, Expression of anaplastic lymphoma kinase in soft tissue tumors: an immunohistochemical and molecular study of 249 cases. Hum Pathol 35:711–721 Mino-Kenudson M et al, 2010, A novel, highly sensitive antibody allows for the routine detection of ALK-rearranged lung adenocarcinomas by standard immunohistochemistry. Clin Cancer Res 16:1561–1571 Minca EC et al, 2013, ALK status testing in non-small cell lung carcinoma: correlation between ultrasensitive IHC and FISH. J Mol Diagn 15:341–346 Murga-Zamalloa C, Lim MS, 2014, ALK-driven tumors and targeted therapy: focus on crizotinib. Pharmgenomics Pers Med 7: 87–94 Subramaniam MM, Piqueras M, Navarro S, Noguera R, 2009, Aberrant copy numbers ofALK gene is a frequent genetic alteration in neuroblastomas. Hum Pathol 40:1638–1642 Lowe EJ, Lim MS, 2013, Potential therapies for anaplastic lymphoma kinase-driven tumors in children: progress to date. Paediatr Drugs 15:163–169 Griffin CA et al, 1999, Recurrent involvement of 2p23 in inflammatory myofibroblastic tumors. Cancer Res 59:2776–2780 Christensen JG et al, 2007, Cytoreductive antitumor activity of PF- 2341066, a novel inhibitor of anaplastic lymphoma kinase and cmet, in experimental models of anaplastic large-cell lymphoma. Mol Cancer Ther 6:3314–3322 Mosse YP et al, 2013, Safety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: a Children's oncology group phase 1 consortium study. Lancet Oncol 14:472–480 De Brouwer S et al, 2010, Meta-analysis of neuroblastomas reveals a skewed ALK mutation spectrum in tumors with MYCN amplification. Clin Cancer Res 16:4353–4362 Passoni L et al, 2009, Mutation-independent anaplastic lymphoma kinase overexpression in poor prognosis neuroblastoma patients. Cancer Res 69:7338–7346 Peron M, Lovisa F, Poli E, Basso G, Bonvini P, 2015, Understanding the interplay between expression, mutation and activity of ALK receptor in Rhabdomyosarcoma cells for clinical application of small-molecule inhibitors. PLoS One 10:e0132330 Kiratli H, Uzun S, Varan A, Akyuz C, Orhan D, 2016, Management of anaplastic lymphoma kinase positive orbitoconjunctival inflammatory myofibroblastic tumor with crizotinib. J AAPOS 20:260–263 Gaudichon J et al, 2016, Complete and repeated response of a metastatic ALK-rearranged inflammatory Myofibroblastic tumor to Crizotinib in a teenage girl. J Pediatr Hematol Oncol 38:308–311 Lawrence B et al, 2000, TPM3-ALK and TPM4-ALK oncogenes in inflammatory myofibroblastic tumors.AmJ Pathol 157:377–384 Schonherr C et al, 2012, Anaplastic lymphoma kinase, ALK, regulates initiation of transcription of MYCN in neuroblastoma cells. Oncogene 31:5193–5200 Moore NF et al, 2014, Molecular rationale for the use of PI3K/AKT/mTOR pathway inhibitors in combination with crizotinib in ALK-mutated neuroblastoma. Oncotarget 5:8737–8749 Carpenter EL et al, 2012, Antibody targeting of anaplastic lymphoma kinase induces cytotoxicity of human neuroblastoma. Oncogene 31:4859–4867 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 217 EP 224 DI 10.1007/s12253-017-0332-1 PG 8 ER PT J AU Mitrakas, L Gravas, S Papandreou, Ch Koukoulis, G Karasavvidou, F Dimakopoulos, G Weingartner, K Karatzas, A Zachos, I Tzortzis, V AF Mitrakas, Lampros Gravas, Stavros Papandreou, Christos Koukoulis, Georgios Karasavvidou, Foteini Dimakopoulos, Georgios Weingartner, Karl Karatzas, Anastasios Zachos, Ioannis Tzortzis, Vasilios TI Primary High-Grade Non-Muscle-Invasive Bladder Cancer: High NFκB Expression in Tumor Specimens Distinguishes Patients Who are at Risk for Disease Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NFκΒ; High-grade; Non-muscle-invasive; Bladder cancer; Progression; BCG ID NFκΒ; High-grade; Non-muscle-invasive; Bladder cancer; Progression; BCG AB To investigate the potential prognostic role of NFκB expression in primary high-grade non-muscle-invasive bladder cancer. Patients with primary high-grade non-muscle-invasive bladder cancer who received induction and maintenance BCG therapy were retrospectively included. Recurrence and progression were histologically proven. Intensity and extent of immunochemistry were assessed. The final evaluation of the NFκB staining was done by combining intensity and extent as product and expressing it as low NFκΒ expression or high NFκB expression . Epidemiological, pathological, clinical parameters and NFκB expression were statistically analyzed for recurrence (REC), progression (PR), recurrence-free survival (RFS) and progression-free survival (PFS). NFκB is significantly associated with disease progression (p < 0,001 in univariate analysis and p = 0,001, Odds Ratio = 14,484, 95% Confidence Interval = 3187-65,821 in multivariate analysis), but not with recurrence. The median value of NFκB expression as product is significantly higher for the patients with progression in comparison to patients with recurrence only (p = 0,003) and patients without recurrence or progression (p = 0,001). Patients’ age is significantly associated (p = 0,001 in univariate analysis and p = 0,003, Odds Ratio = 1273, 95% Confidence Interval = 1086–1492 in multivariate analysis) with disease recurrence. High NFκB expression in primary high-grade non-muscle-invasive bladder cancer, treated with postoperative intravesical BCG immunotherapy, could represent an unfavorable prognostic factor. C1 [Mitrakas, Lampros] University Hospital of Larissa, Faculty of Medicine-School of Health Sciences-University of Thessaly, Department of UrologyLarissa, Greece. [Gravas, Stavros] University Hospital of Larissa, Faculty of Medicine-School of Health Sciences-University of Thessaly, Department of UrologyLarissa, Greece. [Papandreou, Christos] University Hospital of Larissa, Faculty of Medicine-School of Health Sciences-University of Thessaly, Department of OncologyLarissa, Greece. [Koukoulis, Georgios] University of Thessaly, Medical School, Department of PathologyLarissa, Greece. [Karasavvidou, Foteini] University of Thessaly, Medical School, Department of PathologyLarissa, Greece. [Dimakopoulos, Georgios] Science and Technology Park of Epirus, Medical StatisticsIoannina, Greece. [Weingartner, Karl] Akademisches Lehrkrankenhaus der Friedrich-Alexander-Universitat Erlangen-Nurnberg, Sozialstiftung Bamberg-Klinikum am Bruderwald, Klinik fur Urologie und Kinderurologie, Buger Straße 80, 96049 Bamberg, Germany. [Karatzas, Anastasios] University Hospital of Larissa, Faculty of Medicine-School of Health Sciences-University of Thessaly, Department of UrologyLarissa, Greece. [Zachos, Ioannis] University Hospital of Larissa, Faculty of Medicine-School of Health Sciences-University of Thessaly, Department of UrologyLarissa, Greece. [Tzortzis, Vasilios] University Hospital of Larissa, Faculty of Medicine-School of Health Sciences-University of Thessaly, Department of UrologyLarissa, Greece. RP Mitrakas, L (reprint author), University Hospital of Larissa, Faculty of Medicine-School of Health Sciences-University of Thessaly, Department of Urology, Larissa, Greece. EM lamprosmit@gmail.com CR BabjukM, BohleA, Burger M, Capoun O, CohenD, ComperatEM et al, 2016, EAU Guidelines on Non-Muscle-invasive Urothelial Carcinoma of the Bladder: Update 2016. Eur Urol. , DOI 10.1016/j.eururo.2016.05.041 Sylvester RJ, Brausi MA, KirkelsWJ, HoeltlW, Calais Da Silva F, Powell PH et al, 2010, Long-term efficacy results of EORTC Genito-Urinary Group randomized phase 3 study 30911 comparing intravesical instillations of epirubicin, bacillus Calmette-Guerin, and bacillus Calmette-Guerin plus isoniazid in patients with intermediate- and high-risk stage Ta T1 urothelial carcinoma of the bladder. Eur Urol 57(5):766–773 Soloway M, Khoury S, 2012, Bladder cancer. 2nd edition 2012: ICUD-EAU; 254, 261. EDITIONS 21, Paris, France Lee CT, Dunn RL, Ingold C, Montie JE,Wood DP Jr, 2007, Earlystage bladder cancer surveillance does not improve survival if highrisk patients are permitted to progress to muscle invasion. Urology 69(6):1068–1072 Herr HW, Sogani PC, 2001, Does early cystectomy improve the survival of patients with high risk superficial bladder tumors? J Urol 166(4):1296–1299 Zachos I, Konstantinopoulos PA, Vandoros GP, Karamouzis MV, Papatsoris AG, Podimatas T et al, 2009, Predictive value of telomerase reverse transcriptase expression in patients with high risk superficial bladder cancer treated with adjuvant BCG immunotherapy. J Cancer Res Clin Oncol 135(9):1169–1175 Saint F, Salomon L, Quintela R, Cicco A, Hoznek A, Abbou CC et al, 2003, Do prognostic parameters of remission versus relapse after bacillus Calmette-Guerin, BCG, immunotherapy exist? Analysis of a quarter century of literature. Eur Urol 43(4):351–360 Ghosh S, May M, Kopp EB, 1998, NFκB and Rel proteins: evolutionary conserved mediators of immune responses. Annu Rev Immunol 16:225–260 Levidou G, Saetta AA, Korkolopoulou P, Papanastasiou P, Gioti K, Pavlopoulos P et al, 2008, Clinical significance of nuclear factor, NF)-Κb levels in urothelial carcinoma of the urinary bladder. Virchows Arch 452(3):295–304 Kang S, Kim YB, Kim MH, Yoon KS, Kim JW, Park NH et al, 2005, Polymorphism in the nuclear factor kappa-B binding promoter region of cyclooxygenase-2 is associated with an increased risk of bladder cancer. J Can Let 217(1):11–16 Pahl HL, 1999, Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene 18(49):6853–6866 Lamm DL, Blumenstein BA, Crissman JD, Montie JE, Gottesman JE, Lowe BA et al, 2000, Maintenance bacillus Calmette-Guerin immunotherapy for recurrent TA, T1 and carcinoma in situ transitional cell carcinoma of the bladder: a randomized Southwest Oncology Group Study. J Urol 163(4):1124–1129 Shukla S, MacLennan GT, Fu P, Patel J, Marengo SR, Resnick MI et al, 2004, Nuclear factor-kappa B/p65, Rel A, is constitutively activated in human prostate adenocarcinoma and correlates with disease progression. Neoplasia 6(4):390–400 Vlachostergios PJ, Karasavvidou F, Kakkas G, Moutzouris G, Patrikidou A, Voutsadakis IA et al, 2012, Expression of neutral endopeptidase, endothelin-1 and nuclear factor kappa B in prostate cancer: interrelations and associations with prostate-specific antigen recurrence after radical prostatectomy. Prostate Cancer 2012: 452795 Zachos I, Tzortzis V, Mitrakas L, Samarinas M, Karatzas A, Gravas S et al, 2014, Tumor size and T stage correlate independently with recurrence and progression in high-risk non-muscle-invasive bladder cancer patients treated with adjuvant BCG. Tumour Biol 35(5): 4185–4189 Sylvester RJ, van der Meijden AP, Oosterlinck W, Witjes JA, Bouffioux C, Denis et al, 2006, Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials. Eur Urol 49(3):466–465; discussion 475–477 Nakshatri H, Bhat-Nakshatri P, Martin DA, Goulet RJ, Sledge GW, 1997, Constitutive activation of NF-B during progression of breast cancer to hormone-independent growth. Mol Cell Biol 17:3629– 3639 Lind DS, Hochwald SN, Malaty J, Rekkas S, Hebig P, Mishra G et al, 2001, Nuclear factor-kappa B is upregulated in colorectal cancer. Surgery 2:363–369 Visconti R, Cerutti J, Battista S, FedeleM, Trapasso F, Zeki K et al, 1997, Expression of the neoplastic phenotype by human thyroid carcinoma cell lines requires NF-κB p65 protein expression. Oncogene 15:1987–1994 Levidou G, Korkolopoulou P, Nikiteas N, Tzanakis N, Thymara I, Saetta AA et al, 2007, Expression of nuclear factor kappaB in human gastric carcinoma: relationship with IkappaBa and prognostic significance. Virchows Arch 450(5):519–527 SasakiN,Morisaki T, Hashizume K,Yao T, TsuneyoshiM, Noshiro H et al, 2001, Nuclear factor-kappaB p65, RelA, transcription factor is constitutively activated in human gastric carcinoma tissue. Clin Cancer Res 12:4136–4142 WangW, Abbruzzese JL, Evans DB, Larry L, Cleary KR, Chiao PJ, 1999, The nuclear factor-κB/RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells. Clin Cancer Res 5:119–127 Lessard L, Mes-Masson AM, Lamarre L,Wall L, Lattouf JB, Saad F, 2003, NF-κB nuclear localization and its prognostic significance in prostate cancer. BJU Int 91:417–420 Suh J, Payvandi F, Edelstein LC, Amenta PS, ZongWX, Celinas C et al, 2002, Mechanisms of constitutive NFkappaB activation in human prostate cancer cells. Prostate 52(3):183–200 Gasparian AV, Yao YJ, Kowalczyk D, Lyakh LA, Karseladze A, Slaga TJ et al, 2002, The role of IKK in constitutive activation of NF-kappaB transcription factor in prostate carcinoma cells. J Cell Sci 115:141–151 Xie DH, Tang XD, Xia SJ, Tan JM, Wang XH, Cai Y, 2002, Expression of NF-kappa B in human bladder cancer and its clinical significance. Ai Zheng 21(6):663–667 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 225 EP 231 DI 10.1007/s12253-017-0340-1 PG 7 ER PT J AU Seclaman, E Narita, D Anghel, A Cireap, N Ilina, R Sirbu, OI Marian, C AF Seclaman, Edward Narita, Diana Anghel, Andrei Cireap, Natalia Ilina, Razvan Sirbu, Ovidiu Ioan Marian, Catalin TI MicroRNA Expression in Laser Micro-dissected Breast Cancer Tissue Samples – a Pilot Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; MicroRNA; Laser capture microdissection (LCM) ID Breast cancer; MicroRNA; Laser capture microdissection (LCM) AB Breast cancer continues to represent a significant public health burden despite outstanding research advances regarding the molecular mechanisms of cancer biology, biomarkers for diagnostics and prognostic and therapeutic management of this disease. The studies of micro RNAs in breast cancer have underlined their potential as biomarkers and therapeutic targets; however most of these studies are still done on largely heterogeneous whole breast tissue samples. In this pilot study we have investigated the expression of four micro RNAs (miR-21, 145, 155, 92) known to be involved in breast cancer, in homogenous cell populations collected by laser capture microdissection from breast tissue section slides. Micro RNA expression was assessed by real time PCR, and associations with clinical and pathological characteristics were also explored. Our results have confirmed previous associations of miR-21 expression with poor prognosis characteristics of breast cancers such as high stage, large and highly proliferative tumors. No statistically significant associations were found with the other micro RNAs investigated, possibly due to the small sample size of our study. Our results also suggest that miR-484 could be a suitable endogenous control for data normalization in breast tissues, these results needing further confirmation by future studies. In summary, our pilot study showed the feasibility of detecting micro RNAs expression in homogenous laser captured microdissected invasive breast cancer samples, and confirmed some of the previously reported associations with poor prognostic characteristics of breast tumors. C1 [Seclaman, Edward] University of Medicine and Pharmacy, Biochemistry Department, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania. [Narita, Diana] University of Medicine and Pharmacy, Biochemistry Department, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania. [Anghel, Andrei] University of Medicine and Pharmacy, Biochemistry Department, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania. [Cireap, Natalia] University of Medicine and Pharmacy, Department of Surgical OncologyTimisoara, Romania. [Ilina, Razvan] University of Medicine and Pharmacy, Department of Surgical OncologyTimisoara, Romania. [Sirbu, Ovidiu Ioan] University of Medicine and Pharmacy, Biochemistry Department, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania. [Marian, Catalin] University of Medicine and Pharmacy, Biochemistry Department, Pta Eftimie Murgu Nr. 2, 300041 Timisoara, Romania. RP Marian, C (reprint author), University of Medicine and Pharmacy, Biochemistry Department, 300041 Timisoara, Romania. EM cmarian@umft.ro CR Al-Nakhle H, Burns PA, Cummings M, Hanby AM, Hughes TA, Satheesha S et al, 2010, Estrogen receptor {beta}1 expression is regulated by miR-92 in breast cancer. Cancer Res 70:4778–4784 Andersen CL, Jensen JL, Orntoft TF, 2004, Normalization of realtime quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res 64:5245–5250 Blenkiron C, Goldstein LD, Thorne NP, Spiteri I, Chin SF, Dunning MJ et al, 2007, MicroRNA expression profiling of human breast cancer identifies new markers of tumor subtype. Genome Bio 8: R214 Calin GA, Croce CM, 2006, MicroRNA signatures in human cancers. Nat Rev Cancer 6:857–866 Chen J,Wang X, 2014, MicroRNA-21 in breast cancer: diagnostic and prognostic potential. Clin Transl Oncol 16:225–233 Chen L, Li Y, Fu Y, Peng J, Mo MH, Stamatakos M et al, 2013, Role of deregulated microRNAs in breast cancer progression using FFPE tissue. PLoS One 8:e54213 Davoren PA, McNeill RE, Lowery AJ, Kerin MJ, Miller N, 2008, Identification of suitable endogenous control genes for microRNA gene expression analysis in human breast cancer. BMC Mol Biol 9:76 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H et al, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403 Fetica B, Balacescu O, Balacescu L, Rus M, Berindan-Neagoe I, 2014, An alternative and sensitive method based on LCM and QPCR for HER2 testing in breast cancer. Cancer Biomark 14:129–135 Foekens JA, Sieuwerts AM, Smid M, Look MP, de Weerd V, Boersma AW, et al, 2008, Four miRNAs associated with aggressiveness of lymph node-negative, estrogen receptor-positive human breast cancer. Proc Natl Acad Sci U S A 105:13021–13026 Hagrass HA, Sharaf S, Pasha HF, Tantawy EA, Mohamed RH, Kassem R, 2015, Circulating microRNAs - a new horizon in molecular diagnosis of breast cancer. Genes Cancer 6:281–287 Hannafon BN, Sebastiani P, De Las Morenas A, Lu J, Rosenberg CL, 2011, Expression of microRNA and their gene targets are dysregulated in preinvasive breast cancer. Breast Cancer Res 13: R24 Hasemeier B, Christgen M, Kreipe H, Lehmann U, 2008, Reliable microRNA profiling in routinely processed formalin-fixed paraffinembedded breast cancer specimens using fluorescence labelled bead technology. BMC Biotechnol 8:90 Hu Z, Dong J, Wang LE, Ma H, Liu J, Zhao Yet al, 2012, Serum microRNA profiling and breast cancer risk: the use of miR-484/191 as endogenous controls. Carcinogenesis 33:828–834 Huang Q, Gumireddy K, Schrier M, le Sage C, Nagel R, Nair S, et al., 2008, The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat Cell Biol 10:202–210 Hui AB, Shi W, Boutros PC, Miller N, Pintilie M, Fyles T et al, 2009, Robust global micro-RNA profiling with formalin-fixed paraffin-embedded breast cancer tissues. Lab Investig 89:597–606 Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S et al, 2005, MicroRNA gene expression deregulation in human breast cancer. Cancer Res 65:7065–7070 Iorio MV, Casalini P, Tagliabue E, Menard S, Croce CM, 2008, MicroRNA profiling as a tool to understand prognosis, therapy response and resistance in breast cancer. Eur J Cancer 44:2753– 2759 Khoshnaw SM, Powe DG, Ellis IO, Green AR, 2011, Detection and quantification of microRNAs in laser-microdissected formalinfixed paraffin-embedded breast cancer tissues. Methods Mol Biol 755:119–142 Klopfleisch R,Weiss AT, Gruber AD, 2011, Excavation of a buried treasure DNA, mRNA, miRNA and protein analysis in formalin fixed, paraffin embedded tissues. Histol Histopathol 26:797–810 Kodahl AR, LyngMB, Binder H, Cold S, Gravgaard K, Knoop AS et al, 2014, Novel circulating microRNA signature as a potential non-invasive multi-marker test in ER-positive early-stage breast cancer: a case control study. Mol Oncol 8:874–883 Kong W, Zhao JJ, He L, Cheng JQ, 2009, Strategies for profiling microRNA expression. J Cell Physiol 218:22–25 Liu H,McDowell TL, Hanson NE, Tang X, Fujimoto J, Rodriguez- Canales J, 2014, Laser capturemicrodissection for the investigative pathologist. Vet Pathol 51:257–269 Lou G, Ma N, Xu Y, Jiang L, Yang J, Wang C et al, 2015, Differential distribution of U6, RNU6-1, expression in human carcinoma tissues demonstrates the requirement for caution in the internal control gene selection for microRNA quantification. Int J Mol Med 36(5):1400–1408 Lu J, Getz G,Miska EA, Alvarez-Saavedra E, Lamb J, Peck D et al, 2005, MicroRNA expression profiles classify human cancers. Nature 435:834–838 Ma L, Teruya-Feldstein J, Weinberg RA, 2007, Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature 449:682–688 Mar-Aguilar F, Mendoza-Ramirez JA, Malagon-Santiago I, Espino-Silva PK, Santuario-Facio SK, Ruiz-Flores P et al, 2013, Serum circulating microRNA profiling for identification of potential breast cancer biomarkers. Dis Markers 34:163–169 Mattiske S, Suetani RJ, Neilsen PM, Callen DF, 2012, The oncogenic role of miR-155 in breast cancer. Cancer Epidemiol Biomark Prev 21:1236–1243 Min W, Wang B, Li J, Han J, Zhao Y, Su W et al, 2014, The expression and significance of five types of miRNAs in breast cancer. Med Sci Monit Basic Res 20:97–104 Murray GI, 2011, Laser Capture Microdissection: Methods and Protocols. Methods in molecular biology. Springer Narita D, Anghel A, Seclaman E, Ilina R, Cireap N, Ursoniu S, 2010, Molecular profiling of ADAM12 gene in breast cancers. Romanian J Morphol Embryol 51:669–676 Ng EK, Li R, Shin VY, Jin HC, Leung CP, Ma ES et al, 2013, Circulating microRNAs as specific biomarkers for breast cancer detection. PLoS One 8:e53141 Nilsson S, Moller C, Jirstrom K, Lee A, Busch S, Lamb R et al, 2012, Downregulation of miR-92a is associated with aggressive breast cancer features and increased tumour macrophage infiltration. PLoS One 7:e36051 Pan F, Mao H, Deng L, Li G, Geng P, 2014, Prognostic and clinicopathological significance of microRNA-21 overexpression in breast cancer: a meta-analysis. Int J Clin Exp Pathol 7:5622–5633 Pfaffl MW, Tichopad A, Prgomet C, Neuvians TP, 2004, Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper–Excel-based tool using pair-wise correlations. Biotechnol Lett 26:509–515 Rice J, Roberts H, Rai SN, Galandiuk S, 2015, Housekeeping genes for studies of plasma microRNA: A need for more precise standardization. Surgery 158(5):1345–1351 Schmittgen TD, Lee EJ, Jiang J, Sarkar A, Yang L, Elton TS et al, 2008, Real-time PCR quantification of precursor and mature microRNA. Methods 44:31–38 SiML, Zhu S,Wu H, Lu Z,Wu F,Mo YY(2007)miR-21-mediated tumor growth. Oncogene 26:2799–2803 Si H, Sun X, Chen Y, Cao Y, Chen S, Wang H et al, 2013, Circulating microRNA-92a and microRNA-21 as novel minimally invasive biomarkers for primary breast cancer. J Cancer Res Clin Oncol 139:223–229 Siebolts U, Varnholt H, Drebber U, Dienes HP, Wickenhauser C, Odenthal M, 2009, Tissues from routine pathology archives are suitable for microRNA analyses by quantitative PCR. J Clin Pathol 62:84–88 Silver N, Best S, Jiang J, Thein SL, 2006, Selection of housekeeping genes for gene expression studies in human reticulocytes using real-time PCR. BMC Mol Biol 7:33 Society AC. Cancer Facts & Figures 2015, 2015, Atlanta: American Cancer Society Stahel R, Bogaerts J, Ciardiello F, de Ruysscher D, Dubsky P, Ducreux M, et al, 2015, Optimising translational oncology in clinical practice: strategies to accelerate progress in drug development. Cancer Treat Rev 41:129–135 Stefani G, Slack FJ, 2008, Small non-coding RNAs in animal development. Nat Rev Mol Cell Biol 9:219–230 Sun EH, Zhou Q, Liu KS, Wei W, Wang CM, Liu XF et al, 2014, Screening miRNAs related to different subtypes of breast cancer with miRNAs microarray. Eur Rev Med Pharmacol Sci 18:2783– 2788 Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, et al, 2002, Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol 3:RESEARCH0034 Wang J,Wu J, 2012, Role of miR-155 in breast cancer. Front Biosci, Landmark Ed, 17:2350–2355 Wang F, Hou J, Jin W, Li J, Yue Y, Jin H et al, 2014, Increased circulating microRNA-155 as a potential biomarker for breast cancer screening: a meta-analysis. Molecules 19:6282–6293 Xi Y, Nakajima G, Gavin E, Morris CG, Kudo K, Hayashi K et al, 2007, Systematic analysis of microRNA expression of RNA extracted from fresh frozen and formalin-fixed paraffin-embedded samples. RNA 13:1668–1674 Yan X, Chen X, Liang H, Deng T, Chen W, Zhang S et al, 2014, miR-143 and miR-145 synergistically regulate ERBB3 to suppress cell proliferation and invasion in breast cancer. Mol Cancer 13:220 Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Giannakakis A et al, 2006, microRNAs exhibit high frequency genomic alterations in human cancer. Proc Natl Acad Sci U S A 103:9136–9141 Zou C, Xu Q, Mao F, Li D, Bian C, Liu LZ et al, 2012, MiR-145 inhibits tumor angiogenesis and growth by N-RAS and VEGF. Cell Cycle 11:2137–2145 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 233 EP 239 DI 10.1007/s12253-017-0343-y PG 7 ER PT J AU Nagyivanyi, K Budai, B Kuronya, Zs Gyergyay, F Biro, K Bodrogi, I Geczi, L AF Nagyivanyi, Krisztian Budai, Barna Kuronya, Zsofia Gyergyay, Fruzsina Biro, Krisztina Bodrogi, Istvan Geczi, Lajos TI Outcome of Restarted Sunitinib Treatment in Patients with Metastatic Renal Cell Carcinoma: a Retrospective Trial and Combined Case Reports from Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Drug holiday; Overall survival; Patient’s choice; Progression-free survival; Toxicity ID Drug holiday; Overall survival; Patient’s choice; Progression-free survival; Toxicity AB In practice it is still not clear whether a drug holiday in sunitinib (Su) treatment can be safety, without impairing the overall outcome of patients with metastatic renal cell carcinoma (mRCC). The aim was to retrospectively evaluate the outcome in patients who restarted Su after an interruption of ≥3 months and a combined analysis of case studies from literature. From 556 patients treated between January 2006 and March 2016 a group of 38 patients were selected whose treatment was interrupted for other reasons than disease progression. During interruption Su was restarted in case of RECIST-defined progression. The primary objective was the objective response (OR) and progression free survival (PFS) of baseline and restarted therapy. The secondary objective was the overall survival (OS) calculated from the start of baseline treatment. Multivariate survival analysis was also applied. The major causes of interruption were toxicity (39%) and patient’ choice (24%). Median duration of interruption was 7 (range 3–41) months. The OR of baseline and restarted treatment was 63% and 39%, respectively. After a median follow-up of 76 (95% CI 65–79) months the median PFS of baseline and restarted treatment was 21 (18–27) and 14 (10–18) months, respectively. The median OS was 61 (56–80) months. In multivariate analysis the lack of OR of restated treatment was an independent predictor of shorter PFS of restarted Su. According to our findings and also on combined case studies from literature restarted Su can be effective in selected cases of patients who progressed during treatment holiday. C1 [Nagyivanyi, Krisztian] National Institute of Oncology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Budai, Barna] National Institute of Oncology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Kuronya, Zsofia] National Institute of Oncology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Gyergyay, Fruzsina] National Institute of Oncology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Biro, Krisztina] National Institute of Oncology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Bodrogi, Istvan] National Institute of Oncology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. [Geczi, Lajos] National Institute of Oncology, Rath Gy. u. 7-9, 1122 Budapest, Hungary. RP Budai, B (reprint author), National Institute of Oncology, 1122 Budapest, Hungary. EM budai@oncol.hu CR Koo DH, Park I, Ahn JH et al, 2016, Long-term outcomes of tyrosine kinase inhibitor discontinuation in patients with metastatic renal cell carcinoma. Cancer Chemother Pharmacol 77:339–347. , DOI 10.1007/s00280-015-2942-1 Griffioen AW, Mans LA, de Graaf AM et al, 2012, Rapid angiogenesis onset after discontinuation of sunitinib treatment of renal cell carcinoma patients. Clin Cancer Res 18:3961–3971. https://doi. org/10.1158/1078-0432.CCR-12-0002 Albiges L, Oudard S, Negrier S et al, 2012, Complete remission with tyrosine kinase inhibitors in renal cell carcinoma. J Clin Oncol 30:482–487. , DOI 10.1200/JCO.2011.37.2516 Johannsen M, Staehler M, Ohlmann CH et al, 2011, Outcome of treatment discontinuation in patients with metastatic renal cell carcinoma and no evidence of disease following targeted therapy with or withoutmetastasectomy. Ann Oncol 22:657–663. , DOI 10.1093/annonc/mdq437 Nagyivanyi K, Budai B, Biro K et al, 2016, Synergistic survival: a new phenomenon connected to adverse events of first-line sunitinib treatment in advanced renal cell carcinoma. Clin Genitourin Cancer 14:314–322. , DOI 10.1016/j.clgc.2015.11.016 Ornstein MC, Wood LS, Elson P et al, 2017, A phase II study of intermittent sunitinib in previously untreated patients with metastatic renal cell carcinoma. J Clin Oncol 35:1764–1769. https://doi. org/10.1200/JCO.2016.71.1184 Johannsen M, Florcken A, Bex A et al, 2009, Can tyrosine kinase inhibitors be discontinued in patients with metastatic renal cell carcinoma and a complete response to treatment? A multicentre, retrospective analysis. Eur Urol 55:1430–1438. , DOI 10. 1016/j.eururo.2008.10.021 Miura Y, Fujii Y, Shimomura A et al, 2014, Temporal cessation of sunitinib treatment in patients with metastatic renal cell carcinoma: a retrospective study. Ann Oncol 25(Suppl.5):v46. , DOI 10.1093/annonc/mdu435.10 Demiselle J, Lheureux S, Clarisse B et al, 2011, Metastatic renal cancer: evolution of five complete response cases after the antiangiogenic discontinuation. Bull Cancer 98:626–632. https:// doi.org/10.1684/bdc.2011.1368 Bono P ESMO e-learning: renal cell cancer - clinical case study 1 and 2. http://oncologypro.esmo.org/content/download/22271/ 368712/file/Renal-Cell-Cancer-Bono.pdf Vaz MA, Pachon V, Grande E et al, 2011, Complete response to sunitinib in a patient with relapsed irresectable renal cell carcinoma. Anti-Cancer Drugs 22(8):817–821. , DOI 10.1097/CAD. 0b013e3283437ff9 Zhao B, Wood LS, James K, Rini BI, 2015, Is it safe to restart antivascular endothelial growth factor therapy in patients with renal cell carcinoma after cardiac ischemia? Case Rep Oncol Med 2015: 817578. , DOI 10.1155/2015/817578 Calvo OF, Vazquez DD, Lopez MR, Aparicio LM, 2010, Renal cell carcinoma: complete response.Anti-Cancer Drugs 21(Suppl 1): S17–S18. , DOI 10.1097/01.cad.0000361531.59299.5a Paule B, Brion N, 2010, Efficacy of sunitinib in patients with renal cell carcinoma with bone metastases. Anticancer Res 30:5165– 5168 Shablak A, O'Dwyer J, Hawkins R, Board R, 2011, Management of a new isolated metastasis during sunitinib treatment in renal cell carcinoma patients: a lesson from two cases. Urol Int 86:245–248. , DOI 10.1159/000321908 Gomez-Abuin G, Karam AA, Mezzadri NA, Bas CA, 2009, Acalculous cholecystitis in a patient with metastatic renal cell carcinoma treated with sunitinib. Clin Genitourin Cancer 7:62–63. , DOI 10.3816/CGC.2009.n.011 van der Veldt AA, van den Eertwegh AJ, Boven E, 2007, Adverse effects of the tyrosine-kinase inhibitor sunitinib, a new drug for the treatment of advanced renal-cell cancer. Ned Tijdschr Geneeskd 151:1142–1147 Szmit S, Zagrodzka M, KurzynaM et al, 2009, Sunitinib malate, a receptor tyrosine kinase inhibitor, is effective in the treatment of restrictive heart failure due to heart metastases from renal cell carcinoma. Cardiology 114:67–71. , DOI 10.1159/000213049 Kim H, Shoji S, Usui Yet al, 2013, A case of complete response of multiple lung metastases of renal cell carcinoma with the flexible administration of sunitinib. Hinyokika Kiyo 59:7–10 Fukui S, Toyoshima Y, Inoue T et al, 2016, Reversible posterior leukoencephalopathy syndrome developing after restart of sunitinib therapy for metastatic renal cell carcinoma. Case Rep Med 2016: 6852951. , DOI 10.1155/2016/6852951 Schmidinger M, Larkin J, Ravaud A, 2012, Experience with sunitinib in the treatment of metastatic renal cell carcinoma. Ther Adv Urol 4:253–265. , DOI 10.1177/1756287212454933 Schmidinger M, Bojic A, Vogl UM et al, 2009, Management of cardiac adverse events occurring with sunitinib treatment. Anticancer Res 29:1627–1629 Molina AM, Jia X, Feldman DR et al, 2013, Long-term response to sunitinib therapy for metastatic renal cell carcinoma. Clin Genitourin Cancer 11:297–302. , DOI 10.1016/j.clgc. 2013.04.001 Iacovelli R,Massari F, Albiges L et al, 2015, Evidence and clinical relevance of tumor flare in patients who discontinue tyrosine kinase inhibitors for treatment of metastatic renal cell carcinoma. Eur Urol 68:154–160. , DOI 10.1016/j.eururo.2014.10.034 Hammers HJ, Verheul HM, Salumbides B et al, 2010, Reversible epithelial to mesenchymal transition and acquired resistance to sunitinib in patients with renal cell carcinoma: evidence from a xenograft study.Mol Cancer Ther 9:1525–1535. , DOI 10.1158/ 1535-7163.MCT-09-1106 Collinson FJ, GregoryWM,McCabe C et al, 2012, The STAR trial protocol: a randomised multi-stage phase II/III study of Sunitinib comparing temporary cessation with allowing continuation, at the time of maximal radiological response, in the first-line treatment of locally advanced/metastatic renal cancer. BMC Cancer 12:598. , DOI 10.1186/1471-2407-12-598 Greef B, Eisen T, 2016, Medical treatment of renal cancer: new horizons. Br J Cancer 115:505–516. , DOI 10.1038/bjc. 2016.230 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 241 EP 247 DI 10.1007/s12253-017-0345-9 PG 7 ER PT J AU Kravchick, S Cherniavsky, E Verchovsky, G Peled, R AF Kravchick, Sergey Cherniavsky, Eugenia Verchovsky, Guy Peled, Ronit TI Multidetector Computed Tomographic Urography (MDCTU): Its Practical Role in Diagnosis of Upper Tract Urothelial Cancer in Patients 50 years and Older with Different Types of Hematuria SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Upper tract TCC; Computer tomography; Urography; Hematuria; Ureteroscopy ID Upper tract TCC; Computer tomography; Urography; Hematuria; Ureteroscopy AB MDCTU is a preferred method for the investigation of malignant lesions in the upper urinary tract. However, to decrease unnecessary radiation exposure the indications for the exam in different groups of patients should be assessed. In this study, we evaluated the role of MDCTU in patients older than 50 years who presented with different types of hematuria. In a retrospective manner, we assessed the radiologic reports of 173 patients ≥50 years who underwent MDCTU as a part of the evaluation for hematuria. To estimate the accuracy of MDCTU in the detection of upper urinary tract urothelial carcinoma (UUTUC) we compared MDCTU findings with the results of ureteroscopy.We also evaluated which factors can predict ureteroscopic confirmation of MDCTU based diagnosis. In this list we also included diabetes mellitus and anticoagulant medications. As a result, 140 (103 males and 37 females) patients met the inclusion criteria. Mean patients' age was 69.7±16.98. Smokers and passive smokers comprised 38.6% and 26.4% of our patients, while 37.8% of our patients suffered from DM and 45% took anticoagulant medications. MDCTU suspected urothelial carcinoma in 17% (n=24) of our patients: UUTUC in eight and bladder urothelial carcinoma (BUC) in 16patients. Ureteroscopy had diagnosed UUTUC (with/without concurrent urothelial carcinoma of the bladder) in 9 patients: 6 with suspicious lesions in MDCTU and 3 additional patients with CIS/small low grade TCC. MDCTU had a sensitivity of 66.7%, specificity - 98.5%, positive predictive value - 75% and negative predictive value - 97.7%. The logistic regression model revealed five strong predictors for UUTUC: positive/atypical cytology, recurrent hematuria, MDCTU signs, age and Warfarin treatment. Finally a source of hematuria was diagnosed in 57% of patients, while MDCTU individual accuracy reached 42%. We found that MDCTU can effectively identify patients in whom further endoscopy is unnecessary. Otherwise, elder patients with positive/atypical cytology and recurrent microscopic hematuria, who have MDCTU signs and take Warfarin, should undergo endoscopic evaluation. C1 [Kravchick, Sergey] Assaf Harofeh Medical Center, Department of UrologyZrefin, Israel. [Cherniavsky, Eugenia] Barzilay Medical Center, Department of RadiologyAshkelon, Israel. [Verchovsky, Guy] Assaf Harofeh Medical Center, Department of UrologyZrefin, Israel. [Peled, Ronit] Ben Gurion University, Epidemiology and StatisticsBeer-Sheva, Israel. RP Kravchick, S (reprint author), Assaf Harofeh Medical Center, Department of Urology, Zrefin, Israel. EM kravchick.sergey@gmail.com CR Munoz JJ, Ellison LM, 2000, Upper tract urothelial neoplasms: incidence and survival during the last 2 decades. J Urol 164(5): 1523–1525 Roupreˆt M, Babjuk M, Comperat E et al, 2013, European guidelines on upper tract urothelial carcinomas: 2013 update. Eur Urol 63:1059–1071 Babjuk M, Burger M, Zigeuner R et al, 2013, EAU guidelines on non–muscle-invasive urothelial carcinoma of the bladder: update 2013. Eur Urol 64:639–653 Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics, 2012. CA Cancer J Clin 62:10–29 Roupreˆt M, Babjuk M, Comperat E, Zigeuner R et al, 2015, European Association of Urology guidelines on upper urinary tract urothelial cell carcinoma: 2015 update. Eur Urol 68:868–879 Azemar MD, Comperat E, Richard F, Cussenot O, Roupret M, 2011, Bladder recurrence after surgery for upper urinary tract urothelial cell carcinoma: frequency, risk factors, and surveillance. Urol Oncol 29(2):130–136 Raman JD, Ng CK, Scherr DS et al, 2010, Impact of tumor location on prognosis for patients with upper tract urothelial carcinomamanaged by radical Nephroureterectony. Eur Urol 57(6):1072–1079 LiWM, Shen JT, Li CC et al, 2010, Oncologic outcomes following three different Aproaches to the distal ureter and bladder cuff in Nephroureterectomy for primary upper urinary tract urothelial carcinoma. Eur Urol 57(6):963–969 Margulis V, Shariat SF, Matin SF et al, 2009, Outcomes of radical nephroureterectomy: a series from the upper tract urothelial carcinoma collaboration. Cancer 115(6):1224–1233 Van Der Molen AJ, Cowan NC, Mueller-Lisse UG et al, 2008, CT urography: definition, indications and techniques. A guideline for clinical practice. Eur Radiol 18(1):4–17 Wang LJ, Wong YC, Chuang CK et al, 2009, Diagnostic accuracy of transitional cell carcinoma on multidetector computerized tomography urography in patients with gross hematuria. J Urol 181(2):524–531 Wang LJ,Wong YC, Huang CC et al, 2010)Multidetector computerized tomography urography ismore accurate than excretory urography for diagnosing transitional cell carcinoma of the upper tract in adults with hematuria. J Urol 183(1):48–55 Caoili EM, Cohan RH, Korobkin M et al, 2002, Urinary tract abnormalities: initial experience with multi-detector row CT urography. Radiology 222:353–360 Chlapoutakis K, Theocharopoulos N, YarmenitisS DJ, 2010, Performance of computed tomographic urography in diagnosis of upper urinary tract urothelial carcinoma, in patients presenting with hematuria: systematic review and meta-analysis. Eur J Radiol 73:334–338 Loo RK, Lieberman SF, Slezak JM et al, 2013, Stratifying risk of urinary tract malignant tumors in patients with asymptomatic microscopic hematuria. Mayo Clin Proc 88(2):129–138 O’Connor OJ, McSweeney SE, Maher MM, 2008, Imaging of hematuria. Radiol Clin N Am 46:113–132 Edwards TJ, Dickinson AJ, Natale S, Gosling J,McGrath JS, 2006, A prospective analysis of the diagnostic yield resulting from the attendance of 4020 patients at a protocol-driven haematuria clinic. BJU Int 97:301–305 Cauberg EC1, Nio CY, de la Rosette JM, Laguna MP, de ReijkeTM, 2011, Computed tomography-urography for upper urinary tract imaging: is it required for all patients who present with hematuria? J Endourol 25(11):1733–1740 Lisanti CJ1, Toffoli TJ, Stringer MT, DeWitt RM, Schwope RB, 2014, CTevaluation of the upper urinary tract in adults younger than 50 years with asymptomatic microscopic hematuria: is IV contrast enhancement needed? AJR Am J Roentgenol 203(3):615–619 Raman SP, Horton KM, Fishman EK, 2012, Transitional cell carcinoma of the upper urinary tract: optimizing image interpretation with 3D reconstructions. Abdom Imaging 37:1129–1140 Johnson PT, Horton KM, Fishman EK, 2010, Optimizing detectability of renal pathology with MDCT: protocols, pearls, and pitfalls. AJR 194:1001–1012 Woolcott CG1, Maskarinec G, Haiman CA, Henderson BE, Kolonel LN, 2011, Diabetes and urothelial cancer risk: the multiethnic cohort study. Cancer Epidemiol 35(6):551–554 Jung H, Gleason JM, Loo RK et al, 2011, Association of hematuria on microscopic urinalysis and risk of urinary tract cancer. J Urol 185(5):1698–1703 Munoz JJ, Ellison LM, 2000, Upper tract urothelial neoplasms: incidence and survival during the last 2 decades. J Urol 164: 1523–1525 van Osch FH, Jochems SH, van Schooten FJ, Bryan RT, Zeegers MP, 2016, Significant role of lifetime cigarette smoking in worsening bladder cancer and upper tract urothelial carcinoma prognosis: a meta-analysis. J Urol 195(4):872–879 Caoili EM, Cohan RH, Inampudi P et al, 2005, MDCT urography of upper tract urothelial neoplasms. AJR 184:1873–1881 Xu AD, Ng CS, Kamat A, Grossman HB, Dinney C, Sandler CM, 2010, Significance of upper urinary tract urothelial thickening and filling defect seen onMDCT urography in patients with a history of urothelial neoplasms. AJR 195:959–965 Browne RF, Meehan CP, Colville J, Power R, Torreggiani WC, 2005, Transitional cell carcinoma of the upper urinary tract: spectrum of imaging findings. Radiographics 25:1609–1627 Xu AD1, Ng CS, Kamat A, Grossman HB, Dinney C, Sandler CM, 2010, Significance of upper urinary tract urothelial thickening and filling defect seen onMDCT urography in patients with a history of urothelial neoplasms. AJR Am J Roentgenol 195(4):959–965 Khadra MH, Pickard RS, Charlton M et al, 2000, A prospective analysis of 1,930 patients with hematuria to evaluate current diagnostic practice. J Urol 163(2):524–527 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 249 EP 254 DI 10.1007/s12253-017-0333-0 PG 6 ER PT J AU Eide, N Faye, SR Hoifodt, KH Sandvik, L Qvale, AG Faber, R Jebsen, P Kvalheim, G Fodstad, AF Eide, Nils Faye, S Ragnar Hoifodt, K Hanne Sandvik, Leiv Qvale, A Geir Faber, Rowan Jebsen, Peter Kvalheim, Gunnar Fodstad, Oystein TI The Results of Stricter Inclusion Criteria in an Immunomagnetic Detection Study of Micrometastatic Cells in Bone Marrow of Uveal Melanoma Patients - Relevance for Dormancy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Micrometastasis in bone marrow; Uveal melanoma; Exclusion criteria; Immunomagnetic detection; Survival; Tumour dormancy ID Micrometastasis in bone marrow; Uveal melanoma; Exclusion criteria; Immunomagnetic detection; Survival; Tumour dormancy AB Approximately 50% of uveal melanoma patients develop metastases. We want to evaluate the effect of stricter criteria on our data from our previous study correlating survival and bone marrow (BM) micrometastasis results using our immunomagnetic separation (IMS) method. Mononuclear cell fractions (MNC) isolated from BM were examined for tumour cells and the patients were classified as BM positive (BM+) or BM negative (BM-). The study originally included 328 consecutive patients with uveal melanoma from 1997 to 2006. The cohort was limited to 217 patients when we introduced cyto- or histopathological verification of melanoma cells in the patient as a main new criterion for inclusion. Tumour cells were found in BM-samples in 38.7% (95% CI, 32–45) at enrolment. Until the latest workup 43.8% (95% CI, 38–50) of patients had developed melanoma metastases. After a minimum follow-up time of 8.5 years, 60.4% (95% CI, 54–66) of patients had died. The causes were: melanoma metastases 69.5%, another type of cancer 5.4% and non-cancerous causes 19.5%. Overall median survival was shorter for the BM- patients (11.3 years) (95% CI, 10–12) compared to the BM+ (16.5 years) (95% CI, 12–14), p = 0.04, log rank test. All-cause mortality and specific melanoma mortality estimated after 12 year follow-up showed a highly significant difference comparing BM- and BM+, p = 0.010 and p = 0,017, respectively. IMS yields a high fraction of BM+ samples due to micrometastasis at diagnosis and these cells appear to have a positive prognostic impact strengthening our previous report. The late recurrences support the concept of tumour dormancy. C1 [Eide, Nils] Oslo University Hospital HF, Department of Ophthalmology, Nydalen, 0424 Oslo, Norway. [Faye, S Ragnar] Oslo University Hospital HF, Department of DermatologyOslo, Norway. [Hoifodt, K Hanne] Oslo University Hospital HF, Department of Tumor BiologyOslo, Norway. [Sandvik, Leiv] Oslo University Hospital HF, Department of StatisticsOslo, Norway. [Qvale, A Geir] Oslo University Hospital HF, Department of Ophthalmology, Nydalen, 0424 Oslo, Norway. [Faber, Rowan] Oslo University Hospital HF, Department of Ophthalmology, Nydalen, 0424 Oslo, Norway. [Jebsen, Peter] Oslo University Hospital HF, Division of PathologyOslo, Norway. [Kvalheim, Gunnar] Oslo University Hospital HF, Department of OncologyOslo, Norway. [Fodstad, Oystein] Oslo University Hospital HF, Department of Tumor BiologyOslo, Norway. RP Eide, N (reprint author), Oslo University Hospital HF, Department of Ophthalmology, 0424 Oslo, Norway. EM uxeidn@ous-hf.no;nils.andreas.eide@gmail.com CR Bergman L, Seregard S, Nilsson B, Ringborg U, Lundell G, Ragnarsson-Olding B, 2002, Incidence of uveal melanoma in Sweden from 1960 to 1998. Invest Ophthalmol Vis Sci 43(8): 2579–2583 Jensen OA, 1982, Malignant melanomas of the human uvea: 25- year follow-up of cases in Denmark, 1943–1952. Acta Ophthalmol 60(2):161–182 Kujala E, Makitie T, Kivela T, 2003, Very long-term prognosis of patients with malignant uveal melanoma. Invest Ophthalmol Vis Sci 44(11):4651–4659 Singh AD, 2001, Uveal melanoma: implications of tumor doubling time. Ophthalmology 108(5):829–831 Eyles J, Puaux A-L,Wang X, Toh B, Prakash C, HongM, Tan TG, Zheng L, Ong LC, Jin Y, Kato M, Prevost-Blondel A, Chow P, Yang H, Abastado J-P, 2010, Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. J Clin Invest 120(6):2030–2039. , DOI 10. 1172/JCI42002 Eide N, Faye RS, HoifodtHK, Overgaard R, Jebsen P, Kvalheim G, Fodstad O, 2009, Immunomagnetic detection of micrometastatic cells in bone marrow in uveal melanoma patients. Acta Ophthalmol 87(8):830–836. , DOI 10.1111/j.1755-3768. 2008.01378.x Eide N, Faye RS, Hoifodt HK, Sandstad B, Qvale G, Faber R, Jebsen P, Kvalheim G, Fodstad O, 2015, Immunomagnetic detection of micrometastatic cells in bone marrow of uveal melanoma patients: a paradox. Acta Ophthalmol 93(1):59–66. , DOI 10.1111/aos.12462 Coupland SE, Sidiki S, Clark BJ, McClaren K, Kyle P, Lee WR, 1996, Metastatic choroidal melanoma to the contralateral orbit 40 years after enucleation. Arch Ophthalmol 114(6):751–756. https:// doi.org/10.1001/archopht.1996.01100130743022 Mooy CM, Luyten GP, de Jong PT, Luider TM, Stijnen T, van de Ham F, van Vroonhoven CC, Bosman FT, 1995, Immunohistochemical and prognostic analysis of apoptosis and proliferation in uveal melanoma. Am J Pathol 147, 4):1097–1104 Pantel K, Cote RJ, Fodstad O, 1999, Detection and clinical importance of micrometastatic disease. J Natl Cancer Inst 91(13):1113– 1124 Borthwick NJ, Thombs J, Polak M, Gabriel FG, Hungerford JL, Damato B, Rennie IG, Jager MJ, Cree IA, 2011, The biology of micrometastases from uveal melanoma. J Clin Pathol 64(8):666– 671. , DOI 10.1136/jcp.2010.087999 Bronkhorst IHG, Jager MJ, 2013, Inflammation in uveal melanoma. Eye 27(2):217–223. , DOI 10.1038/eye.2012.253 Ma J, Usui Y, TakeuchiM, Okunuki Y, Kezuka T, Zhang L,Mizota A, Goto H, 2010, Human uveal melanoma cells inhibit the immunostimulatory function of dendritic cells. Exp Eye Res 91(4):491–499. , DOI 10.1016/j.exer.2010.06.025 Suesskind D, Ulmer A, Schiebel U, Fierlbeck G, Spitzer B, Spitzer MS, Bartz-Schmidt KU, Grisanti S, 2011, Circulating melanoma cells in peripheral blood of patients with uveal melanoma before and after different therapies and association with prognostic parameters: a pilot study. Acta Ophthalmol 89(1):17–24. , DOI 10.1111/j.1755-3768.2009.01617.x Torres V, Triozzi P, Eng C, Tubbs R, Schoenfiled L, Crabb JW, Saunthararajah Y, Singh AD, 2011, Circulating tumor cells in uveal melanoma. Future Oncol 7(1):101–109. , DOI 10.2217/ fon.10.143 Edge SB, Compton CC, 2010, The American joint committee on cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol 17(6):1471–1474. , DOI 10.1245/s10434-010-0985-4 Khan S, Finger PT, Yu GP, Razzaq L, Jager MJ, de Keizer RJ, Sandkull P, Seregard S, Gologorsky D, Schefler AC, Murray TG, Kivela T, Giuliari GP, McGowan H, Simpson ER, Corriveau C, Coupland SE, Damato BE, 2012, Clinical and pathologic characteristics of biopsy-proven irismelanoma: amulticenter international study. Arch Ophthalmol 130(1):57–64. , DOI 10.1001/ archophthalmol.2011.286 Li Y, Madigan MC, Lai K, Conway RM, Billson FA, Crouch R, Allen BJ, 2003, Human uveal melanoma expresses NG2 immunoreactivity. Br J Ophthalmol 87(5):629–632 Char DH, Miller T, 1995, Accuracy of presumed uveal melanoma diagnosis before alternative therapy. Br J Ophthalmol 79(7):692– 696 Damato B, 2010, Does ocular treatment of uveal melanoma influence survival? Br J Cancer 103(3):285–290 Finger PT, Kurli M, Reddy S, Tena LB, Pavlick AC, 2005, Whole body PET/CT for initial staging of choroidal melanoma. Br J Ophthalmol 89(10):1270–1274. , DOI 10.1136/bjo.2005. 069823 Faye RS, Aamdal S, Hoifodt HK, Jacobsen E, Holstad L, Skovlund E, Fodstad O, 2004, Immunomagnetic detection and clinical significance of micrometastatic tumor cells in malignant melanoma patients. Clin Cancer Res 10(12 Pt 1):4134–4139. , DOI 10.1158/1078-0432.ccr-03-0408 Eide N, Hoifodt HK, Nesland JM, Faye RS, Qvale GA, Faber RT, Jebsen P,KvalheimG, Fodstad O(2013, Disseminated tumour cells in bonemarrow of patients with uveal melanoma. Acta Ophthalmol 91(4):343–348. , DOI 10.1111/j.1755-3768.2012.02449.x Ulmer A, Schmidt-Kittler O, Fischer J, Ellwanger U, Rassner G, Riethmuller G, Fierlbeck G, Klein CA, 2004, Immunomagnetic enrichment, genomic characterization, and prognostic impact of circulating melanoma cells. Clin Cancer Res 10(2):531–537 Tura A, Luke J,Merz H, Reinsberg M, Luke M, Jager MJ, Grisanti S, 2014, Identification of circulating melanoma cells in uveal melanoma patients by dual-marker immunoenrichment. Invest Ophthalmol Vis Sci 55(7):4395–4404. , DOI 10.1167/ iovs.14-14512 Freeman JB, Gray ES, Millward M, Pearce R, Ziman M, 2012, Evaluation of a multi-marker immunomagnetic enrichment assay for the quantification of circulating melanoma cells. J Transl Med 10:192. , DOI 10.1186/1479-5876-10-192 Cools-Lartigue JJ, McCauley CS, Marshall JC, Di Cesare S, Gregoire F, Antecka E, Logan P, Burnier MN, 2008, Immunomagnetic isolation and in vitro expansion of human uveal melanoma cell lines. Mol Vis 14:50–55 Callejo SA, Antecka E, Blanco PL, Edelstein C, Burnier MN Jr, 2006, Identification of circulating malignant cells and its correlation with prognostic factors and treatment in uveal melanoma. A prospective longitudinal study. Eye 21(6):752–759 Tura A, Merz H, ReinsbergM, LukeM, Jager MJ, Grisanti S, Luke J, 2016, Analysis of monosomy-3 in immunomagnetically isolated circulating melanoma cells in uveal melanoma patients. Pigment Cell Melanoma Res 29(5):583–589. , DOI 10.1111/pcmr. 12507 Demicheli R, Fornili M, Biganzoli E, 2014, Bimodal mortality dynamics for uveal melanoma: a cue for metastasis development traits? BMC Cancer 14:392–392. , DOI 10.1186/1471- 2407-14-392 Zimmerman LE, McLean IW, FosterWD, 1978, Does enucleation of the eye containing a malignant melanoma prevent or accelerate the dissemination of tumour cells. Br J Ophthalmol 62:420–425 Packard RBS, 1980, Pattern of mortality in choroidal malignant melanoma. Br J Ophthalmol 64:565–575 EideMB, LiestolK, Lingjaerde OC, HystadME,Kresse SH, Meza- Zepeda L, Myklebost O, Troen G, Aamot HV, Holte H, Smeland EB, Delabie J, 2010, Genomic alterations reveal potential for higher grade transformation in follicular lymphoma and confirm parallel evolution of tumor cell clones. Blood 116(9):1489–1497. , DOI 10.1182/blood-2010-03-272278 Marshall JC, Nantel A, Blanco P, Ash J, Cruess SR, Burnier MN Jr, 2007, Transcriptional profiling of human uveal melanoma from cell lines to intraocular tumors to metastasis. Clin Exp Metastasis 24(5):353–362. , DOI 10.1007/s10585-007-9072-z Mackie RM, Reid R, Junor B, 2003, Fatal melanoma transferred in a donated kidney 16 years after melanoma surgery. N Engl J Med 348:567–568 Kolandjian NA,Wei C, Patel SP, Richard JL, Dett T, Papadopoulos NE, Bedikian AY, 2013, Delayed systemic recurrence of uveal melanoma. Am J Clin Oncol 36(5):443–449. , DOI 10. 1097/COC.0b013e3182546a6b Burg MA, Grako KA, Stallcup WB, 1998, Expression of the NG2 proteoglycan enhances the growth and metastatic properties ofmelanoma cells. J Cell Physiol 177(2):299–312. , DOI 10. 1002/(sici)1097-4652(199811)177:2<299:aid-jcp12>3.0.co;2-5 Eide N, Garred O, Beiske K, Fodstad O, 2016, Bilateral uveal melanomas with different gene expression detected with 7 years interval. Acta Ophthalmol 94(1):99–102. , DOI 10.1111/ aos.12857 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 255 EP 262 DI 10.1007/s12253-017-0355-7 PG 8 ER PT J AU Schroeder, CA Xiao, H Zhu, Z Li, Q Bai, Q Wakefield, RM Mann, DJ Fang, Y AF Schroeder, C Andrew Xiao, Huaping Zhu, Ziwen Li, Qing Bai, Qian Wakefield, R Mark Mann, D Jeffrey Fang, Yujiang TI A Potential Role for Green Tea as a Radiation Sensitizer for Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Green tea; Prostate cancer; Radiation therapy ID Green tea; Prostate cancer; Radiation therapy AB Prostate cancer (PCa) is the most common noncutaneous cancer in the United States. There is currently a lack of safe and effective radiosensitizers that can enhance the effectiveness of radiation treatment (RT) for Pca. Clonogenic assay, PCNA staining, Quick Cell Proliferation assay, TUNEL staining and caspase-3 activity assay were used to assess proliferation and apoptosis in DU145 Pca cells. RTPCR/ IHC were used to investigate the mechanisms.We found that the percentage of colonies, PCNA staining intensity, and the optical density value of DU145 cells were decreased (RT/ GT vs. RT). TUNEL + cells and the relative caspase-3 activity were increased (RT/GT vs. RT). Compared to RT, the antiproliferative effect of RT/GT-correlated with increased expression of the anti-proliferative molecule p16. Compared to RT, the pro-apoptotic effect of RT/GT correlated with decreased expression of the anti-apoptotic molecule Bcl-2. GT enhances RT sensitivity of DU145 by inhibiting proliferation and promoting apoptosis. C1 [Schroeder, C Andrew] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. [Xiao, Huaping] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. [Zhu, Ziwen] University of Missouri, School of Medicine, Department of SurgeryColumbia, MO, USA. [Li, Qing] The Affiliated Hospital of Xiangnan UniversityChenzhou, Hunan, China. [Bai, Qian] University of Missouri, School of Medicine, Department of SurgeryColumbia, MO, USA. [Wakefield, R Mark] University of Missouri, School of Medicine, Department of SurgeryColumbia, MO, USA. [Mann, D Jeffrey] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. [Fang, Yujiang] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, IA, USA. RP Fang, Y (reprint author), Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, USA. CR Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(1):7–30. , DOI 10.3322/caac.21387 Bernard W, Christopher P, 2014, World cancer report 2014. International Agency for Research on Cancer. World Health Organization, Lyon, France Zhang S, Shan L, Li Q,Wang X, Li S, Zhang Y, Fu J, Liu X, Li H, ZhangW(2014, Systematic analysis of the multiple bioactivities of green tea through a network pharmacology approach. Evid Based Complement Alternat Med 2014:512081. , DOI 10.1155/ 2014/512081 Fang Y, DeMarco VG, Nicholl MB, 2012, Resveratrol enhances radiation sensitivity in prostate cancer by inhibiting cell proliferation and promoting cell senescence and apoptosis. Cancer Sci 103(6):1090–1098 Davalli P, Rizzi F, Caporali A, Pellacani D, Davoli S, Bettuzzi S, Brausi M, D’Arca D, 2012, Anticancer activity of green tea polyphenols in prostate gland. Oxidative Med Cell Longev 2012: 984219. , DOI 10.1155/2012/984219 Liang X, Gao J, Sun X, Zhu L, Jia Y, Gu Y, Han C, Zhang X, Hou S, 2013, [Tea polyphenols inhibits the proliferation of prostate cancer DU145 cells]. Zhonghua nan ke xue=. Natl J Androl 19(6):495–500 Kwak TW, Park SB, Kim H-J, Jeong Y-I, Kang DH, 2017, Anticancer activities of epigallocatechin-3-gallate against cholangiocarcinoma cells. OncoTargets Ther 10:137 Naponelli V, Ramazzina I, Lenzi C, Bettuzzi S, Rizzi F, 2017, Green tea Catechins for prostate cancer prevention: present achievements and future challenges. Antioxidants 6(2):26 Oya Y,Mondal A, Rawangkan A, Umsumarng S, Iida K,Watanabe T, Kanno M, Suzuki K, Li Z, Kagechika H, Shudo K, Fujiki H, Suganuma M, 2017, Down-regulation of histone deacetylase 4, −5 and −6 as a mechanism of synergistic enhancement of apoptosis in human lung cancer cells treated with the combination of a synthetic retinoid, Am80 and green tea catechin. J Nutr Biochem 42:7–16. , DOI 10.1016/j.jnutbio.2016.12.015 Tsai Y-J, Chen B-H, 2016, Preparation of catechin extracts and nanoemulsions from green tea leaf waste and their inhibition effect on prostate cancer cell PC-3. Int J Nanomedicine 11:1907 Henning SM, Aronson W, Niu Y, Conde F, Lee NH, Seeram NP, Lee R-P, Lu J, Harris DM, Moro A, 2006, Tea polyphenols and theaflavins are present in prostate tissue of humans and mice after green and black tea consumption. J Nutr 136(7):1839–1843 Fang Y, Bradley MJ, Cook KM, Herrick EJ, Nicholl MB, 2013, A potential role for resveratrol as a radiation sensitizer for melanoma treatment. J Surg Res 183(2):645–653 Fang Y, Braley-Mullen H, 2008, Cultured murine thyroid epithelial cells expressing transgenic Fas-associated death domain-like interleukin- 1β converting enzyme inhibitory protein are protected from Fas-mediated apoptosis. Endocrinology 149(7):3321–3329 Fang Y, Sharp GC, Braley-Mullen H, 2008, Interleukin-10 promotes resolution of granulomatous experimental autoimmune thyroiditis. Am J Pathol 172(6):1591–1602 Fang Y, Wei Y, DeMarco V, Chen K, Sharp GC, Braley-Mullen H, 2007, Murine FLIP transgene expressed on thyroid epithelial cells promotes resolution of granulomatous experimental autoimmune thyroiditis in DBA/1 mice. Am J Pathol 170(3):875–887 Fang Y, Moore BJ, Bai Q, Cook KM, Herrick EJ, Nicholl MB, 2013, Hydrogen peroxide enhances radiation-induced apoptosis and inhibition of melanoma cell proliferation. Anticancer Res 33(5):1799–1807 Fang Y, Herrick EJ, Nicholl MB, 2012, A possible role for perforin and Granzyme B in resveratrol-enhanced Radiosensitivity of prostate cancer. J Androl 33(4):752–760 Fang Y, Sharp GC, Yagita H, Braley-Mullen H, 2008, A critical role for TRAIL in resolution of granulomatous experimental autoimmune thyroiditis. J Pathol 216(4):505–513 Dulaney CR, Osula DO, Yang ES, Rais-Bahrami S, 2016, Prostate radiotherapy in the era of advanced imaging and precision medicine. Prostate. Cancer 2016:4897515. , DOI 10.1155/2016/ 4897515 Johnson D, Walker C, 1999, Cyclins and cell cycle checkpoints. Annu Rev Pharmacol Toxicol 39(1):295–312 Nobori T, 1994, Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Trends Genet 10(7):228 Helgadottir H, Hoiom V, Jonsson G, Tuominen R, Ingvar C, Borg A, Olsson H, Hansson J, 2014, High risk of tobacco-related cancers in CDKN2A mutation-positive melanoma families. J Med Genet 51(8):545–552 Caldas C, Hahn SA, da Costa LT, Redston MS, Schutte M, Seymour AB, Weinstein CL, Hruban RH, Yeo CJ, Kern SE, 1994, Frequent somatic mutations and homozygous deletions of the p16, MTS1, gene in pancreatic adenocarcinoma. Nat Genet 8(1):27–32 Griffith TS, Brunner T, Fletcher SM, Green DR, Ferguson TA, 1995, Fas ligand-induced apoptosis as a mechanism of immune privilege. Science 270(5239):1189–1192 Thompson CB, 1995, Apoptosis in the pathogenesis and treatment of disease. Science 267(5203):1456 Zhu Z, Zhang D, Lee H, Menon AA, Wu J, Hu K, Jin Y, 2017, Macrophage-derived apoptotic bodies promote the proliferation of the recipient cells via shuttling microRNA-221/222. J Leukoc Biol 101(6):1349–1359. , DOI 10.1189/jlb.3A1116-483R Zhang H, Xu Q, Krajewski S, Krajewska M, Xie Z, Fuess S, Kitada S, Pawlowski K, Godzik A, Reed JC, 2000, BAR: an apoptosis regulator at the intersection of caspases and Bcl-2 family proteins. Proc Natl Acad Sci 97(6):2597–2602 Yip K, Reed J, 2008, Bcl-2 family proteins and cancer. Oncogene 27(50):6398–6406 Khanna K,Wie T, Song Q, Burrows S, Moss D, Krajewski S, Reed J, Lavin M, 1996, Expression of p53, bcl-2, bax, bcl-x2 and c-myc in radiation-induced apoptosis in Burkitt's lymphoma cells. Cell Death Differ 3(3):315–322 Krajewska M, Krajewski S, Epstein JI, Shabaik A, Sauvageot J, Song K, Kitada S, Reed JC, 1996, Immunohistochemical analysis of bcl-2, bax, bcl-X, and mcl-1 expression in prostate cancers. Am J Pathol 148(5):1567 Tron V, Krajewski S, Klein-Parker H, Li G, Ho V, Reed J, 1995, Immunohistochemical analysis of Bcl-2 protein regulation in cutaneous melanoma. Am J Pathol 146(3):643 Tsujimoto Y, Cossman J, Jaffe E, Croce CM, 1985, Involvement of the bcl-2 gene in human follicular lymphoma. Science 228(4706): 1440–1443 Yip KW, Shi W, Pintilie M, Martin JD, Mocanu JD, Wong D, MacMillan C, Gullane P, O'Sullivan B, Bastianutto C, 2006, Prognostic significance of the Epstein-Barr virus, p53, Bcl-2, and survivin in nasopharyngeal cancer. Clin Cancer Res 12(19):5726–5732 Zhu Z, Davidson KT, Brittingham A, Wakefield MR, Bai Q, Xiao H, Fang Y, 2016, Trichomonas vaginalis: a possible foe to prostate cancer. Med Oncol 33(10):115 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 263 EP 268 DI 10.1007/s12253-017-0358-4 PG 6 ER PT J AU Martinez-Useros, J Li, W Georgiev-Hristov, T Fernandez-Acenero, JM Borrero-Palacios, A Perez, N Celdran, A Garcia-Foncillas, J AF Martinez-Useros, Javier Li, Weiyao Georgiev-Hristov, Tihomir Fernandez-Acenero, J Maria Borrero-Palacios, Aurea Perez, Nuria Celdran, Angel Garcia-Foncillas, Jesus TI Clinical Implications of NRAS Overexpression in Resectable Pancreatic Adenocarcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic ductal adenocarcinoma; PDAC; NRAS; KRAS; TGCA; Biomarker; Progression-free survival; Overall survival; Grade of differentiation ID Pancreatic ductal adenocarcinoma; PDAC; NRAS; KRAS; TGCA; Biomarker; Progression-free survival; Overall survival; Grade of differentiation AB Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal forms of cancer, and its incidence is rising worldwide. Although survival can be improved by surgical resection, when detected at an early stage, this type of cancer is usually asymptomatic, and disease becomes only apparent after metastasis. Adjuvant treatment does not improve survival, thus after surgery there is a lack of predictive and prognosis biomarkers to predict treatment response and survival. The mitogen-activated protein-kinase and phosphoinositide 3- kinase signalling pathways play a crucial role in cancer development and progression. Especially, activated RAS proteins promote cell proliferation through constitutive stimulation of the downstream effectors RAF-MEK-ERK and PI3K-AKT. Mutational status of NRAS is required in several types of cancer like colorectal or cutaneous melanoma. However, mutations in this gene are very scarce in PDAC patients, and NRAS determination is not usually performed in clinical practice for this kind of tumor. In this study, we analyse the association between NRAS protein expression and progression-free survival and overall survival of an homogenous cohort of pancreatic ductal adenocarcinoma patients from a single-centre. Interestingly, we found that patients with high expression not only showed longer progression-free survival than those patients with low expression (22 versus 9 months, respectively) (P = 0.013), but also longer overall survival (43 versus 19 months, respectively) (P = 0.020). These results confirm NRAS expression could be used to differentiate patients according to their prognosis. Proportional hazard model revealed NRAS expression together with grade of differentiation as pathological variables to predict patient’s outcome. C1 [Martinez-Useros, Javier] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology Division, Av. Reyes Catolicos 2, 28040 Madrid, Spain. [Li, Weiyao] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology Division, Av. Reyes Catolicos 2, 28040 Madrid, Spain. [Georgiev-Hristov, Tihomir] University Hospital Fundacion Jimenez Diaz, Health Research Institute FJD-UAM, Department of Surgery, Av. Reyes Catolicos 2, 28040 Madrid, Spain. [Fernandez-Acenero, J Maria] Hospital Clinico San Carlos, Department of Surgical Pathology, C/ Profesor Martin Lagos, 28040 Madrid, Spain. [Borrero-Palacios, Aurea] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology Division, Av. Reyes Catolicos 2, 28040 Madrid, Spain. [Perez, Nuria] University Hospital Fundacion Jimenez Diaz, Department of Pathology, Av. Reyes Catolicos 2, 28040 Madrid, Spain. [Celdran, Angel] University Hospital Fundacion Jimenez Diaz, Health Research Institute FJD-UAM, Department of Surgery, Av. Reyes Catolicos 2, 28040 Madrid, Spain. [Garcia-Foncillas, Jesus] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology Division, Av. Reyes Catolicos 2, 28040 Madrid, Spain. RP Garcia-Foncillas, J (reprint author), University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology Division, 28040 Madrid, Spain. EM jesus.garciafoncillas@oncohealth.eu CR Hariharan D, Saied A, Kocher HM, 2008, Analysis of mortality rates for pancreatic cancer across the world. HPB 10(1):58–62 Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66(1):7–30. , DOI 10.3322/caac.21332 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Egawa S, Takeda K, Fukuyama S,Motoi F, Sunamura M,Matsuno S, 2004, Clinicopathological aspects of small pancreatic cancer. Pancreas 28(3):235–240 Ariyama J, Suyama M, Satoh K, Sai J, 1998, Imaging of small pancreatic ductal adenocarcinoma. Pancreas 16(3):396–401 Kelsen DP, Portenoy R, Thaler H, Tao Y, BrennanM(1997, Pain as a predictor of outcome in patients with operable pancreatic carcinoma. Surgery 122(1):53–59 HidalgoM, 2010, Pancreatic cancer. N Engl J Med 362(17):1605– 1617. , DOI 10.1056/NEJMra0901557 Stathis A, Moore MJ, 2010, Advanced pancreatic carcinoma: current treatment and future challenges. Nat Rev Clin Oncol 7(3):163– 172. , DOI 10.1038/nrclinonc.2009.236 Fong ZV,Winter JM, 2012, Biomarkers in pancreatic cancer: diagnostic, prognostic, and predictive. Cancer J 18(6):530–538. https:// doi.org/10.1097/PPO.0b013e31827654ea Martinez-Useros J, Garcia-Foncillas J, 2016, The Role of BRCA2 Mutation Status as Diagnostic, Predictive, and Prognosis Biomarker for Pancreatic Cancer. Biomed Res Int 2016:1869304. , DOI 10.1155/2016/1869304 Maithel SK, Maloney S, Winston C, Gonen M, D'Angelica MI, Dematteo RP, Jarnagin WR, Brennan MF, Allen PJ, 2008, Preoperative CA 19-9 and the yield of staging laparoscopy in patients with radiographically resectable pancreatic adenocarcinoma. Ann Surg Oncol 15(12):3512–3520. , DOI 10.1245/ s10434-008-0134-5 Goonetilleke KS, Siriwardena AK, 2007, Systematic review of carbohydrate antigen, CA 19-9, as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol 33(3):266–270. , DOI 10.1016/j.ejso.2006.10.004 Bosman FTCF, Hruban RH, Theise ND, eds,, 2010, World Health Organization Classification of Tumours of the Digestive System. IARC Press, Lyon Kloppel GHR, Longnecker DS, Adler G, Kern SE, Partanen TJ, 2000, Ductal adenocarcinoma of the pancreas. World Health Organization Classification of Tumours. Pathology and Genetics of Tumours of the Digestive System. IARC Press, Lyon Kim JE, Lee KT, Lee JK, Paik SW, Rhee JC, Choi KW, 2004, Clinical usefulness of carbohydrate antigen 19-9 as a screening test for pancreatic cancer in an asymptomatic population. J Gastroenterol Hepatol 19(2):182–186 Kawai S, Suzuki K, Nishio K, Ishida Y, Okada R, Goto Y, Naito M, Wakai K, Ito Y, Hamajima N, 2008, Smoking and serum CA19-9 levels according to Lewis and secretor genotypes. Int J Cancer 123(12):2880–2884. , DOI 10.1002/ijc.23907 BurottoM, Chiou VL, Lee JM, Kohn EC, 2014, The MAPK pathway across different malignancies: a new perspective. Cancer 120(22):3446–3456. , DOI 10.1002/cncr.28864 Kolch W, 2005, Coordinating ERK/MAPK signalling through scaffolds and inhibitors. Nat Rev Mol Cell Biol 6(11):827–837. , DOI 10.1038/nrm1743 Colicelli J, 2004, Human RAS superfamily proteins and related GTPases. Sci STKE 2004(250):RE13. , DOI 10.1126/ stke.2502004re13 Malumbres M, Barbacid M, 2003, RAS oncogenes: the first 30 years. Nat Rev Cancer 3(6):459–465. , DOI 10.1038/ nrc1097 Roberts PJ, Der CJ, 2007, Targeting the Raf-MEK-ERK mitogenactivated protein kinase cascade for the treatment of cancer. Oncogene 26(22):3291–3310. , DOI 10.1038/sj.onc. 1210422 Colombino M, CaponeM, Lissia A, Cossu A, Rubino C, De Giorgi V, Massi D, Fonsatti E, Staibano S, Nappi O, Pagani E, Casula M, Manca A, Sini M, Franco R, Botti G, Caraco C, Mozzillo N, Ascierto PA, Palmieri G, 2012, BRAF/NRASmutation frequencies among primary tumors and metastases in patients with melanoma. J Clin Oncol 30(20):2522–2529. , DOI 10.1200/JCO.2011. 41.2452 Edlundh-Rose E, Egyhazi S, Omholt K, Mansson-Brahme E, Platz A, Hansson J, Lundeberg J, 2006, NRAS and BRAF mutations in melanoma tumours in relation to clinical characteristics: a study based on mutation screening by pyrosequencing. Melanoma Res 16(6):471–478. , DOI 10.1097/01.cmr.0000232300. 22032.86 Seo JS, Ju YS, Lee WC, Shin JY, Lee JK, Bleazard T, Lee J, Jung YJ, Kim JO, Yu SB, Kim J, Lee ER, Kang CH, Park IK, Rhee H, Lee SH, Kim JI, Kang JH, Kim YT, 2012, The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Res 22(11):2109–2119. , DOI 10.1101/gr. 145144.112 Bansal M, Gandhi M, Ferris RL, Nikiforova MN, Yip L, Carty SE, Nikiforov YE, 2013)Molecular and histopathologic characteristics of multifocal papillary thyroid carcinoma.AmJ Surg Pathol 37(10): 1586–1591. , DOI 10.1097/PAS.0b013e318292b780 Cancer_Genome_Atlas_Network, 2012, Comprehensive molecular characterization of human colon and rectal cancer. Nature 487(7407):330–337. , DOI 10.1038/nature11252 Lievre A, Bachet JB, Le Corre D, Boige V, Landi B, Emile JF, Cote JF, Tomasic G, Penna C, Ducreux M, Rougier P, Penault-Llorca F, Laurent-Puig P, 2006, KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 66(8): 3992–3995. , DOI 10.1158/0008-5472.CAN-06-0191 Smit VT, Boot AJ, Smits AM, Fleuren GJ, Cornelisse CJ, Bos JL, 1988, KRAS codon 12 mutations occur very frequently in pancreatic adenocarcinomas. Nucleic Acids Res 16(16):7773–7782 Kanda M, Matthaei H, Wu J, Hong SM, Yu J, Borges M, Hruban RH, Maitra A, Kinzler K, Vogelstein B, Goggins M, 2012, Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology 142(4):730–733 e739. , DOI 10.1053/j.gastro.2011.12.042 Mitsuhashi K, Nosho K, Sukawa Y, Matsunaga Y, Ito M, Kurihara H, Kanno S, Igarashi H, Naito T, Adachi Y, Tachibana M, Tanuma T, Maguchi H, Shinohara T, Hasegawa T, Imamura M, Kimura Y, Hirata K, Maruyama R, Suzuki H, Imai K, Yamamoto H, Shinomura Y, 2015, Association of Fusobacterium species in pancreatic cancer tissues with molecular features and prognosis. Oncotarget 6(9):7209–7220. 10.18632/oncotarget.3109 Hamidi H, Lu M, Chau K, Anderson L, Fejzo M, Ginther C, Linnartz R, Zubel A, Slamon DJ, Finn RS, 2014)KRASmutational subtype and copy number predict in vitro response of human pancreatic cancer cell lines to MEK inhibition. Br J Cancer 111(9): 1788–1801. , DOI 10.1038/bjc.2014.475 Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK,Wilson PJ, PatchAM,Wu J, Chang DK, Cowley MJ, Gardiner BB, Song S, Harliwong I, Idrisoglu S, Nourse C, Nourbakhsh E, Manning S, Wani S, Gongora M, Pajic M, Scarlett CJ, Gill AJ, Pinho AV, Rooman I, Anderson M, Holmes O, Leonard C, Taylor D, Wood S, Xu Q, Nones K, Fink JL, Christ A, Bruxner T, Cloonan N, Kolle G, Newell F, Pinese M, Mead RS, Humphris JL, Kaplan W, Jones MD, Colvin EK, Nagrial AM, Humphrey ES, Chou A, Chin VT, Chantrill LA, Mawson A, Samra JS, Kench JG, Lovell JA, Daly RJ, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Kakkar N, Zhao F, Wu YQ,WangM,Muzny DM, FisherWE, Brunicardi FC, Hodges SE, Reid JG, Drummond J, ChangK, Han Y, Lewis LR, Dinh H, Buhay CJ, Beck T, Timms L, SamM, Begley K, Brown A, Pai D, Panchal A, Buchner N, De Borja R, Denroche RE, Yung CK, Serra S, Onetto N, Mukhopadhyay D, Tsao MS, Shaw PA, Petersen GM, Gallinger S, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Schulick RD, Wolfgang CL, Morgan RA, Lawlor RT, Capelli P, Corbo V, Scardoni M, Tortora G, Tempero MA, Mann KM, Jenkins NA, Perez-Mancera PA, Adams DJ, Largaespada DA, Wessels LF, Rust AG, Stein LD, Tuveson DA, Copeland NG, Musgrove EA, Scarpa A, Eshleman JR, Hudson TJ, Sutherland RL, Wheeler DA, Pearson JV, McPherson JD, Gibbs RA, Grimmond SM, 2012, Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491(7424):399– 405. , DOI 10.1038/nature11547 Deschenes-Simard X, Gaumont-Leclerc MF, Bourdeau V, Lessard F, Moiseeva O, Forest V, Igelmann S, Mallette FA, Saba-El-Leil MK, Meloche S, Saad F, Mes-Masson AM, Ferbeyre G, 2013, Tumor suppressor activity of the ERK/MAPK pathway by promoting selective protein degradation. Genes Dev 27(8):900–915. , DOI 10.1101/gad.203984.112 Serrano M, Lin AW, ME MC, Beach D, Lowe SW, 1997, Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88(5):593–602 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, Cerami E, Sander C, Schultz N, 2013, Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6(269):pl1. , DOI 10.1126/scisignal.2004088 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, Antipin Y, Reva B, Goldberg AP, Sander C, Schultz N, 2012, The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2(5):401–404. , DOI 10. 1158/2159-8290.CD-12-0095 Li B, Ruotti V, StewartRM, Thomson JA, Dewey CN(2010, RNASeq gene expression estimation with read mapping uncertainty. Bioinformatics 26(4):493–500. , DOI 10.1093/ bioinformatics/btp692 Adsay NV, Basturk O, Bonnett M, Kilinc N, Andea AA, Feng J, Che M, Aulicino MR, Levi E, Cheng JD, 2005, A proposal for a new and more practical grading scheme for pancreatic ductal adenocarcinoma. Am J Surg Pathol 29(6):724–733 Wagner M, Redaelli C, Lietz M, Seiler CA, Friess H, BuchlerMW, 2004, Curative resection is the single most important factor determining outcome in patients with pancreatic adenocarcinoma. Br J Surg 91(5):586–594. , DOI 10.1002/bjs.4484 Cucchetti A, Ercolani G, Taffurelli G, Serenari M, Maroni L, Pezzilli R, Del Gaudio M, Ravaioli M, Cescon M, Pinna AD, 2016, A comprehensive analysis on expected years of life lost due to pancreatic cancer. Pancreatology 16(3):449–453 Ducreux M, Cuhna AS, Caramella C, Hollebecque A, Burtin P, Goere D, Seufferlein T, Haustermans K, Van Laethem JL, Conroy T,Arnold D(2015, Cancer of the pancreas: ESMOClinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 56: v56–v68 Tempero MA MM, Al-Hawary M, Behrman SW, Benson A1, Berlin JD, Cha C, Chiorean EG, Chung V, Cohen SJ, Czito B, Dillhoff M, Feng M, Ferrone CR, Hardacre J, Hawkins W, Herman J, Hoffman JP, Ko AH, Komanduri S, Koong A, Lowy AM, Ma WW, Moravek C, Mulvihill SJ, Nakakura EK, O’Reilly EM, Obando J, Reddy S, Thayer S,Weekes CD,Wolff RA,Wolpin BM, Burns J, Darlow S, 2016, Pancreatic Adenocarcinoma Version 1.2016. NCCN Clinical Practice Guidelines in Oncology, NCCN Guidelines®, National Comprehensive Cancer Network Abbruzzese JL, 2002, New applications of gemcitabine and future directions in the management of pancreatic cancer. Cancer 95(4 Suppl):941–945. , DOI 10.1002/cncr.10753 Li D, Xie K, Wolff R, Abbruzzese JL, 2004, Pancreatic cancer. Lancet 363(9414):1049–1057. , DOI 10.1016/S0140- 6736(04)15841-8 Winter JM, Yeo CJ, Brody JR, 2013, Diagnostic, prognostic, and predictive biomarkers in pancreatic cancer. J Surg Oncol 107(1): 15–22 Sakai W, Swisher EM, Karlan BY, Agarwal MK, Higgins J, Friedman C, Villegas E, Jacquemont C, Farrugia DJ, Couch FJ, Urban N, Taniguchi T, 2008, Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers. Nature 451(7182):1116–1120 Kakavand H, Walker E, Lum T, Wilmott JS, Selinger CI, Smith E, Saw RP, Yu B, Cooper WA, Long GV, O'Toole SA, Scolyer RA, 2016, BRAF(V600E, and NRAS(Q61L/Q61R, mutation analysis in metastatic melanoma using immunohistochemistry: a study of 754 cases highlighting potential pitfalls and guidelines for interpretation and reporting. Histopathology 69(4):680–686. https://doi. org/10.1111/his.12992 Martinez-Useros J, Georgiev-Hristov T, Fernandez-Acenero MJ, Borrero-Palacios A, Indacochea A, Guerrero S, Li W, Cebrian A, Gomez Del Pulgar T, Puime-Otin A, Del Puerto-Nevado L, Rodriguez-Remirez M, Perez N, Celdran A, Gebauer F, Garcia- Foncillas J, 2017, UNR/CDSE1 expression as prognosis biomarker in resectable pancreatic ductal adenocarcinoma patients: A proofof- concept. PLoS One 12(8):e0182044. , DOI 10.1371/ journal.pone.0182044 De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, Kalogeras KT, Kotoula V, Papamichael D, Laurent- Puig P, Penault-Llorca F, Rougier P, Vincenzi B, Santini D, Tonini G, Cappuzzo F, Frattini M, Molinari F, Saletti P, De Dosso S, Martini M, Bardelli A, Siena S, Sartore-Bianchi A, Tabernero J, Macarulla T, Di Fiore F, Gangloff AO, Ciardiello F, Pfeiffer P, Qvortrup C, Hansen TP, Van Cutsem E, Piessevaux H, Lambrechts D, Delorenzi M, Tejpar S, 2010, Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol 11(8):753–762. , DOI 10.1016/S1470-2045(10, 70130-3 Palomba G, Doneddu V, Cossu A, Paliogiannis P,Manca A, Casula M, Colombino M, Lanzillo A, Defraia E, Pazzola A, Sanna G, Putzu C, Ortu S, Scartozzi M, Ionta MT, Baldino G, Sarobba G, Capelli F, Sedda T, Virdis L, Barca M, Gramignano G, Budroni M, Tanda F, Palmieri G, 2016, Prognostic impact of KRAS, NRAS, BRAF, and PIK3CA mutations in primary colorectal carcinomas: a population-based study. J Transl Med 14(1):292. , DOI 10. 1186/s12967-016-1053-z Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat JP, Nickerson E, Auclair D, Li L, Place C, Dicara D, Ramos AH, Lawrence MS, Cibulskis K, Sivachenko A, Voet D, Saksena G, Stransky N, Onofrio RC,WincklerW, Ardlie K,Wagle N,Wargo J, Chong K,Morton DL, Stemke-Hale K, Chen G, Noble M, Meyerson M, Ladbury JE, Davies MA, Gershenwald JE, Wagner SN, Hoon DS, Schadendorf D, Lander ES, Gabriel SB, Getz G, Garraway LA, Chin L, 2012, A landscape of driver mutations in melanoma. Cell 150(2):251–263. , DOI 10.1016/j. cell.2012.06.024 Prior IA, Lewis PD, Mattos C, 2012, A comprehensive survey of Rasmutations in cancer. Cancer Res 72(10):2457–2467. https://doi. org/10.1158/0008-5472.CAN-11-2612 Fiore D, Donnarumma E, Roscigno G, Iaboni M, Russo V, Affinito A, Adamo A, De Martino F, Quintavalle C, Romano G, Greco A, Soini Y, Brunetti A, Croce CM, Condorelli G, 2016, miR-340 predicts glioblastoma survival and modulates key cancer hallmarks through down-regulation of NRAS. Oncotarget 7(15):19531– 19547. 10.18632/oncotarget.6968 Wang L, Shi ZM, Jiang CF, Liu X, Chen QD, Qian X, Li DM, Ge X, Wang XF, Liu LZ, You YP, Liu N, Jiang BH, 2014, MiR-143 acts as a tumor suppressor by targeting N-RAS and enhances temozolomide-induced apoptosis in glioma. Oncotarget 5(14): 5416–5427. 10.18632/oncotarget.2116 LidskyM, Antoun G, Speicher P, Adams B, Turley R, Augustine C, Tyler D, Ali-Osman F, 2014, Mitogen-activated protein kinase, MAPK, hyperactivation and enhanced NRAS expression drive acquired vemurafenib resistance in V600E BRAF melanoma cells. J Biol Chem 289(40):27714–27726. , DOI 10.1074/jbc. M113.532432 Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, Chen Z, Lee MK, Attar N, Sazegar H, Chodon T, Nelson SF, McArthur G, Sosman JA, Ribas A, Lo RS, 2010, Melanomas acquire resistance to B-RAF(V600E, inhibition by RTK or N-RAS upregulation. Nature 468(7326):973–977. , DOI 10.1038/nature09626 Birkeland E, Busch C, Berge EO, Geisler J, Jonsson G, Lillehaug JR, Knappskog S, Lonning PE, 2013, Low BRAF and NRAS expression levels are associated with clinical benefit from DTIC therapy and prognosis in metastatic melanoma. Clin Exp Metastasis 30(7):867–876. , DOI 10.1007/s10585-013- 9587-4 Eisfeld AK, Schwind S, Hoag KW, Walker CJ, Liyanarachchi S, Patel R, Huang X,Markowitz J, DuanW, OttersonGA, CarsonWE 3rd, Marcucci G, Bloomfield CD, de la Chapelle A, 2014, NRAS isoforms differentially affect downstream pathways, cell growth, and cell transformation. Proc Natl Acad Sci U S A 111(11):4179– 4184. , DOI 10.1073/pnas.1401727111 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 269 EP 278 DI 10.1007/s12253-017-0341-0 PG 10 ER PT J AU Laszlo, Z Farkas, R Schally, VA Pozsgai, E Papp, A Bognar, L Tornoczki, T Mangel, L Bellyei, Sz AF Laszlo, Zoltan Farkas, Robert Schally, V Andrew Pozsgai, Eva Papp, Andras Bognar, Laura Tornoczki, Tamas Mangel, Laszlo Bellyei, Szabolcs TI Possible Predictive Markers of Response to Therapy in Esophageal Squamous Cell Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Radiochemotherapy; AKT; Small heat shock protein; Predictiv factors; Esopageal cancer ID Radiochemotherapy; AKT; Small heat shock protein; Predictiv factors; Esopageal cancer AB The aim of the present study was to investigate the relationship between the intensity of biomarker expression and the response to radiochemotherapy in patients with advanced esophageal squamous cell cancer (ESCC). Ninety-two patients with locally advanced ESCC were examined retrospectively. Pre-treatment tumor samples were stained for proteins SOUL, Hsp 16.2, Growth Hormone-Releasing Hormone Receptor (GHRH-R) and p-Akt using immunhistochemistry methods. Kaplan-Meier curves were used to show the relationship between intensity of expression of biomarkers and clinical parameters and 3-year OS. A significant correlation was found between high intensity staining for Hsp 16.2, p-Akt and SOUL and poor response to NRCT. Application of a higher dose of radiation and higher dose of cisplatin resulted in better clinical and histopathological responses, respectively. Among the clinical parameters, the localization of the tumor in the upper-third of the esophagus and less than 10% weight loss were independent prognostic factors for increased 3- year OS. Hsp16.2, p-Akt and SOUL are predictors of negative response to NRCT, therefore these biomarkers may become promising targets for therapy. Furthermore, level of expression of p-Akt, weight loss and the localization of the tumor are significant factors in the prediction of OS in ESCC. C1 [Laszlo, Zoltan] University of Pecs, Department of Oncology, 7624 Pecs, Hungary. [Farkas, Robert] University of Pecs, Department of Oncology, 7624 Pecs, Hungary. [Schally, V Andrew] University of Pecs, Institute of Biochemistry and Medical ChemistryPecs, Hungary. [Pozsgai, Eva] University of Pecs, Institute of Biochemistry and Medical ChemistryPecs, Hungary. [Papp, Andras] University of Pecs, Institute of Biochemistry and Medical ChemistryPecs, Hungary. [Bognar, Laura] University of Pecs, Institute of Biochemistry and Medical ChemistryPecs, Hungary. [Tornoczki, Tamas] University of Pecs, Institute of Biochemistry and Medical ChemistryPecs, Hungary. [Mangel, Laszlo] University of Pecs, Department of Oncology, 7624 Pecs, Hungary. [Bellyei, Szabolcs] University of Pecs, Department of Oncology, 7624 Pecs, Hungary. RP Bellyei, Sz (reprint author), University of Pecs, Department of Oncology, 7624 Pecs, Hungary. EM bellyeisz@gmail.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386. https://doi. org/10.1002/ijc.29210 Keighley MR, 2003, Gastrointestinal cancers in Europe. Aliment Pharmacol Ther 18(Suppl 3):7–30 Mariette C, Piessen G, Triboulet JP, 2007, Therapeutic strategies in oesophageal carcinoma: role of surgery and other modalities. The Lancet Oncology 8(6):545–553. , DOI 10.1016/S1470- 2045(07)70172-9 Gebski V, Burmeister B, Smithers BM, Foo K, Zalcberg J, Simes J, Australasian Gastro-Intestinal Trials G, 2007, Survival benefits from neoadjuvant chemoradiotherapy or chemotherapy in oesophageal carcinoma: a meta-analysis. The Lancet Oncology 8(3):226– 234. , DOI 10.1016/S1470-2045(07)70039-6 Hanna A, Birla R, Iosif C, Boeriu M, Constantinoiu S, 2016, Benefits and Disadvantages of Neoadjuvant Radiochemotherapy, RCT, in the Multimodal Therapy of Squamous Esophageal Cancer, ESC). Chirurg 111(1):12–25 Nakajima M, Kato H, 2013, Treatment options for esophageal squamous cell carcinoma. Expert Opin Pharmacother 14(10): 1345–1354. , DOI 10.1517/14656566.2013.801454 Ordu AD, Nieder C, Geinitz H, Scherer V, Kup PG, Schuster T, Combs SE, Fakhrian K, 2014, Association between radiation dose and pathological complete response after preoperative radiochemotherapy in esophageal squamous cell cancer. Anticancer Res 34(12):7255–7261 Geh JI, Bond SJ, Bentzen SM, Glynne-Jones R, 2006, Systematic overview of preoperative, neoadjuvant, chemoradiotherapy trials in oesophageal cancer: evidence of a radiation and chemotherapy dose response. Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology 78(3):236–244. , DOI 10.1016/j.radonc.2006.01.009 Gillham CM, Reynolds J, Hollywood D, 2007, Predicting the response of localised oesophageal cancer to neo-adjuvant chemoradiation. World Journal of Surgical Oncology 5:97. , DOI 1186/1477-7819-5-97 10. Sarbia M, Ott N, Puhringer-Oppermann F, Brucher BL, 2007, The predictive value of molecular markers, p53, EGFR, ATM, CHK2, in multimodally treated squamous cell carcinoma of the oesophagus. Br J Cancer 97(10):1404–1408. , DOI 10.1038/sj.bjc. 6604037 Zhu Z, Yu W, Fu X, Sun M, Wei Q, Li D, Chen H, Xiang J, Li H, Zhang Y, ZhaoW, Zhao K, 2015, Phosphorylated AKT1 is associated with poor prognosis in esophageal squamous cell carcinoma. Journal of experimental & clinical cancer research : CR 34:95. , DOI 10.1186/s13046-015-0212-z Ui T, Morishima K, Saito S, Sakuma Y, Fujii H, Hosoya Y, Ishikawa S, Aburatani H, Fukayama M, Niki T, Yasuda Y, 2014, The HSP90 inhibitor 17-N-allylamino-17-demethoxy geldanamycin, 17-AAG, synergizes with cisplatin and induces apoptosis in cisplatin-resistant esophageal squamous cell carcinoma cell lines via the Akt/XIAP pathway. Oncol Rep 31(2):619–624. , DOI 10.3892/or.2013.2899 Jin Z, Yan W, Jin H, Ge C, Xu Y, 2016, Psoralidin inhibits proliferation and enhances apoptosis of human esophageal carcinoma cells via NF-kappaB and PI3K/Akt signaling pathways. Oncol Lett 12(2):971–976. , DOI 10.3892/ol.2016.4716 Wang X, Li X, Li C, He C, Ren B, Deng Q, GaoW,Wang B, 2016, Aurora-A modulates MMP-2 expression via AKT/NF-kappaB pathway in esophageal squamous cell carcinoma cells. Acta Biochim Biophys Sin 48(6):520–527. , DOI 10.1093/ abbs/gmw030 Xue L, Yang L, Jin ZA, Gao F, Kang JQ, Xu GH, Liu B, Li H, Wang XJ, Liu LJ, Wang BL, Liang SH, Ding J, 2014, Increased expression of HSP27 inhibits invasion and metastasis in human esophageal squamous cell carcinoma. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine 35(7):6999–7007. , DOI 10.1007/s13277-014- 1946-5 Chen JH, Chen LM, Xu LY, Wu MY, Shen ZY, 2006, Expression and significance of heat shock proteins in esophageal squamous cell carcinoma. Zhonghua zhong liu za zhi [Chinese journal of oncology] 28(10):758–761 Farkas R, Pozsgai E, Bellyei S, Cseke L, Szigeti A, Vereczkei A, Marton S, Mangel L, Horvath OP, Papp A, 2011, Correlation between tumor-associated proteins and response to neoadjuvant treatment in patients with advanced squamous-cell esophageal cancer. Anticancer Res 31(5):1769–1775 Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT, Christian MC, Gwyther SG, 2000, New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 92, 3):205–216 Mandard AM, Dalibard F, Mandard JC,Marnay J, Henry-Amar M, Petiot JF, Roussel A, Jacob JH, Segol P, Samama G et al, 1994, Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Cancer 73(11):2680–2686 Gerard JP, Conroy T, Bonnetain F, Bouche O, Chapet O, Closon-Dejardin MT, Untereiner M, Leduc B, Francois E, Maurel J, Seitz JF, Buecher B, Mackiewicz R, Ducreux M, Bedenne L, 2006, Preoperative radiotherapy with or without concurrent fluorouracil and leucovorin in T3-4 rectal cancers: results of FFCD 9203. J Clin Oncol 24(28):4620– 4625. , DOI 10.1200/JCO.2006.06.7629 Sauer R, Becker H, HohenbergerW, Rodel C,Wittekind C, Fietkau R, Martus P, Tschmelitsch J, Hager E, Hess CF, Karstens JH, Liersch T, Schmidberger H, Raab R, 2004, Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med 351(17):1731–1740. , DOI 10.1056/NEJMoa040694 Bellyei S, Szigeti A, Boronkai A, Pozsgai E, Gomori E,Melegh B, Janaky T, Bognar Z, Hocsak E, Sumegi B, Gallyas F, Jr., 2007, Inhibition of cell death by a novel 16.2 kD heat shock protein predominantly via Hsp90 mediated lipid rafts stabilization and Akt activation pathway. Apoptosis 12, 1):97–112., DOI https://doi. org/10.1007/s10495-006-0486-x Bellyei S, Szigeti A, Pozsgai E, Boronkai A, Gomori E, Hocsak E, Farkas R, Sumegi B, Gallyas F, Jr., 2007, Preventing apoptotic cell death by a novel small heat shock protein. Eur J Cell Biol 86, 3): 161–171., DOI , DOI 10.1016/j.ejcb.2006.12.004 Szigeti A, Bellyei S, Gasz B, Boronkai A, Hocsak E, Minik O, Bognar Z, Varbiro G, Sumegi B, Gallyas F, Jr., 2006, Induction of necrotic cell death and mitochondrial permeabilization by heme binding protein 2/SOUL. FEBS Lett 580, 27):6447–6454., DOI , DOI 10.1016/j.febslet.2006.10.067 Somji S, Sens MA, Lamm DL, Garrett SH, Sens DA, 2001, Metallothionein isoform 1 and 2 gene expression in the human bladder: evidence for upregulation of MT-1X mRNA in bladder cancer. Cancer Detect Prev 25(1):62–75 de Manzoni G, Pedrazzani C, Laterza E, Pasini F, Grandinetti A, Bernini M, Ruzzenente A, Zerman G, Tomezzoli A, Cordiano C, 2005, Induction chemoradiotherapy for squamous cell carcinoma of the thoracic esophagus: impact of increased dosage on long-term results. Ann Thorac Surg 80(4):1176–1183. , DOI 10. 1016/j.athoracsur.2005.02.048 Pasini F, de Manzoni G, Pedrazzani C, Grandinetti A, Durante E, Gabbani M, Tomezzoli A, Griso C, Guglielmi A, Pelosi G, Maluta S, Cetto GL, Cordiano C, 2005, High pathological response rate in locally advanced esophageal cancer after neoadjuvant combined modality therapy: dose finding of a weekly chemotherapy schedule with protracted venous infusion of 5-fluorouracil and dose escalation of cisplatin, docetaxel and concurrent radiotherapy. Annals of oncology : official journal of the European Society for Medical Oncology 16(7):1133–1139. , DOI 10.1093/annonc/ mdi207 Hanna A, Birla R, Iosif C, Boeriu M, Tomsa R, Puscasu A, Constantinoiu S, 2015, Evaluation of Neoadjuvant Radiochemotherapy Response, RCT, in Squamous Esophageal Cancer, ESC, and Implications in Therapeutic Conduct. Chirurg 110(3):214–223 Parcellier A, Schmitt E, Brunet M, Hammann A, Solary E, Garrido C, 2005, Small heat shock proteins HSP27 and alphaB-crystallin: cytoprotective and oncogenic functions. Antioxid Redox Signal 7(3–4):404–413. , DOI 10.1089/ars.2005.7.404 Xu Y, Chen Z, Zhang G, Xi Y, Sun R,Wang X,WangW, Chai F, Li X, 2016, HSP90B1 overexpression predicts poor prognosis in NSCLC patients. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine 37(10):14321–14328. , DOI 10.1007/s13277- 016-5304-7 Chang X, Zhao J, Tian F, Jiang Y, Lu J, Ma J, Zhang X, Jin G, Huang Y, Dong Z, Liu K, Dong Z, 2016, Aloe-emodin suppresses esophageal cancer cell TE1 proliferation by inhibiting AKT and ERK phosphorylation. Oncol Lett 12(3):2232–2238. https://doi. org/10.3892/ol.2016.4910 Sha K, Yeh S, Chang C, Nastiuk KL, Krolewski JJ, 2015, TNF signaling mediates an enzalutamide-induced metastatic phenotype of prostate cancer and microenvironment cell co-cultures. Oncotarget 6(28):25726–25740. 10.18632/oncotarget.4535 Li X, Wang S, Ren H, Ma J, Sun X, Li N, Liu C, Huang K, Xu M, Ming L, 2016, Molecular correlates and prognostic value of tmTNF-alpha expression in colorectal cancer of 5-Fluorouracil- Based Adjuvant Therapy. Cancer biology & therapy 17(6):684– 692. , DOI 10.1080/15384047.2016.1187551 Plonowski A, Schally AV, Letsch M, Krupa M, Hebert F, Busto R, Groot K, Varga JL, 2002, Inhibition of proliferation of PC-3 human prostate cancer by antagonists of growth hormone-releasing hormone: lack of correlation with the levels of serum IGF-I and expression of tumoral IGF-II and vascular endothelial growth factor. Prostate 52(3):173–182. , DOI 10.1002/pros.10105 Kahan Z, Arencibia JM, Csernus VJ, Groot K, Kineman RD, Robinson WR, Schally AV, 1999, Expression of growth hormone-releasing hormone, GHRH, messenger ribonucleic acid and the presence of biologically active GHRH in human breast, endometrial, and ovarian cancers. J Clin Endocrinol Metab 84(2): 582–589. , DOI 10.1210/jcem.84.2.5487 Garcia-Fernandez MO, Schally AV, Varga JL, Groot K, Busto R, 2003, The expression of growth hormone-releasing hormone, GHRH, and its receptor splice variants in human breast cancer lines; the evaluation of signaling mechanisms in the stimulation of cell proliferation. Breast Cancer Res Treat 77(1):15–26 Rekasi Z, Czompoly T, Schally AV, Halmos G, 2000, Isolation and sequencing of cDNAs for splice variants of growth hormonereleasing hormone receptors from human cancers. Proc Natl Acad Sci U S A 97(19):10561–10566. , DOI 10.1073/pnas. 180313297 Halmos G, Schally AV, Varga JL, Plonowski A, Rekasi Z, Czompoly T, 2000, Human renal cell carcinoma expresses distinct binding sites for growth hormone-releasing hormone. Proc Natl Acad Sci U S A 97(19):10555–10560. , DOI 10.1073/ pnas.180313097 Szereday Z, Schally AV, Szepeshazi K, Bajo AM, Hebert F, Halmos G, Nagy A, 2003, Effective treatment of H838 human non-small cell lung carcinoma with a targeted cytotoxic somatostatin analog, AN-238. Int J Oncol 22(5):1141–1146 Hohla F, Moder A, Mayrhauser U, Hauser-Kronberger C, Schally AV,Varga JL, ZarandiM, Buchholz S, Huber R, Aigner E, Ritter M, Datz C, 2008, Differential expression of GHRH receptor and its splice variant 1 in human normal and malignant mucosa of the oesophagus and colon. Int J Oncol 33(1):137–143 Wu CC, Chang CM, Hsu TW, Lee CH, Chen JH, Huang CY, Lee CC, 2016, The effect of individual and neighborhood socioeconomic status on esophageal cancer survival in working-age patients in Taiwan. Medicine 95(27):e4140. , DOI 10.1097/MD. 0000000000004140 Koppert LB, Lemmens VE, Coebergh JW, Steyerberg EW, Wijnhoven BP, Tilanus HW, Janssen-Heijnen ML, 2012, Impact of age and co-morbidity on surgical resection rate and survival in patients with oesophageal and gastric cancer. Br J Surg 99(12): 1693–1700. , DOI 10.1002/bjs.8952 Kandaz M, Ertekin MV, Bilici M, 2012, Retrospective analysis of patients with esophageal cancer treated with radiotherapy and/or chemoradiotherapy. Tumori 98(4):445–450. , DOI 10. 1700/1146.12638 Neuhof D, Neumayer F, Einbeck W, Haschemian K, Mai SK, Hochhaus A, Willeke F, Rudi J, Debus J, Wenz F, 2005, Retrospective evaluation of combined modality treatment and prognostic factors in patients with esophageal cancer. Acta Oncol 44(2): 168–173. , DOI 10.1080/02841860510029563 Di Fiore F, Lecleire S, Pop D, Rigal O, Hamidou H, Paillot B, Ducrotte P, Lerebours E,Michel P, 2007, Baseline nutritional status is predictive of response to treatment and survival in patients treated by definitive chemoradiotherapy for a locally advanced esophageal cancer. Am J Gastroenterol 102(11):2557–2563. , DOI 10. 1111/j.1572-0241.2007.01437.x ZemanovaM, Novak F, Vitek P, Pazdro A, SmejkalM, Pazdrova G, Petruzelka L, 2012, Outcomes of patients with oesophageal cancer treated with preoperative chemoradiotherapy, followed by tumor resection: influence of nutritional factors. Journal of BUON : official journal of the Balkan Union of Oncology 17(2):310–316 Papp A, Cseke L, Farkas R, Pavlovics G, Horvath G, Varga G, Szigeti A, Bellyei S, Marton S, Poto L, Kalmar K, Vereczkei A, Pozsgai E, Horvath OP, 2010, Chemo-radiotherapy in locally advanced squamous cell oesophageal cancer–are upper third tumours more responsive? Pathol Oncol Res 16(2):193–200. , DOI 10.1007/s12253-009-9206-5 Papp A, Cseke L, Pavlovics G, Farkas R, Varga G, Marton S, Poto L, Esik O, Horvath OP, 2007, The effect of preoperative chemoradiotherapy in the treatment of locally advanced squamous cell carcinoma in the upper- and middle-thirds of the esophagus. Magy Seb 60(3):123–129. , DOI 10.1556/MaSeb.60. 2007.3.1 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 279 EP 288 DI 10.1007/s12253-017-0342-z PG 10 ER PT J AU Beardo, P Cacho, TD Izquierdo, L Alcover-Garcia, BJ Alcaraz, A Extramiana, J Mallofre, C AF Beardo, Pastora Cacho, Truan David Izquierdo, Laura Alcover-Garcia, Bautista Joan Alcaraz, Antonio Extramiana, Javier Mallofre, Carmen TI Cancer-Specific Survival Stratification Derived from Tumor Expression of Tissue Inhibitor of Metalloproteinase-2 in Non-Metastatic Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Matrix metalloproteinases; Tissue inhibitors of metalloproteinases; Renal cellcarcinoma; Metastasis; Survival ID Matrix metalloproteinases; Tissue inhibitors of metalloproteinases; Renal cellcarcinoma; Metastasis; Survival AB Degradation of the extracellular matrix is a prerequisite for the processes of cancer cell invasion and metastasis. The purpose of our study was to assess the association of matrix metalloproteinases (MMP-1, MMP-2, MMP-3, MMP-9) and their inhibitors (TIMP-1 and TIMP-2) with renal cell carcinoma (RCC) progression and cancer-specific survival (CSS), using immunohistochemical analysis of 60 formalin- fixed, paraffin-embedded sections of tumor tissue and normal tissue near the tumor from surgical T1-3bN0 M0 RCC specimens. Significant overexpression of MMP-2 in tumor and normal tissue was correlated with advanced stages, tumor size, sarcomatous differentiation and clinical symptoms. Overall survival was 31.7% (55.2% M0, 9.7% M1) and CSS 56.7%(100% M0, 16.1% M1) with a follow-up of 76 (5–230) months. Fuhrman grade [HR 2.87 (95% CI: 1.28–6.45); p = 0.01], tumor size [HR 1.13 (95% CI: 1.03–1.26); p = 0.009] and low TIMP-2 expression [HR 0.35 (95% CI: 0.16–0.78); p = 0.01] were independent predictive factors of CSS and stratified the patients into three groups with different rates of 10-year CSS; [100%, 73.9% and 20.5% for the good, intermediate and poor prognosis group respectively (p = 0.000006)] . This study offers strong evidence that TIMP-2 expression in tumor tissue may play a crucial role in progression and poor prognosis in human localized and locally advanced RCC. C1 [Beardo, Pastora] Araba University Hospital, Department of Urology, c/ Jose Atxotegi s/n, 01009 Vitoria-Gasteiz, Araba, Spain. [Cacho, Truan David] Marques de Valdecilla University Hospital, Department of Urology, Av. Valdecilla 25, 39008 Santander, Cantabria, Spain. [Izquierdo, Laura] Clinic Hospital, Department of Urology, c/ Villarroel 170, 08036 Barcelona, Cataluna, Spain. [Alcover-Garcia, Bautista Joan] Clinic Hospital, Department of Urology, c/ Villarroel 170, 08036 Barcelona, Cataluna, Spain. [Alcaraz, Antonio] Clinic Hospital, Department of Urology, c/ Villarroel 170, 08036 Barcelona, Cataluna, Spain. [Extramiana, Javier] Araba University Hospital, Department of Urology, c/ Jose Atxotegi s/n, 01009 Vitoria-Gasteiz, Araba, Spain. [Mallofre, Carmen] Universitat Autonoma of Barcelona and Clinic Hospital, Department of Pathology, c/ Villarroel 170, 08036 Barcelona, Cataluna, Spain. RP Beardo, P (reprint author), Araba University Hospital, Department of Urology, 01009 Vitoria-Gasteiz, Spain. EM beardo.pastora@gmail.com CR Bourboulia D, Stetler-Stevenson WG, 2010, Matrix metalloproteinases, MMPs, and tissue inhibitors of metalloproteinases, TIMPs): positive and negative regulators in tumor cell adhesion. Semin Cancer Biol 20:161–168 Cho NH, Shim HS, Rha SY, Kang SH, Hong SH, Choi ID et al, 2003, Increased expression of matrix metalloproteinase 9 correlates with poor prognostic variables renal cell carcinoma. Eur Urol 44:560–566 Eble JN, Sauter G, Epstein JI, Sesterhenn IA, Eds,, 2004, World health organization classification of tumors. Pathology and Genetics of Tumors of the Urinary System and Male Genital Organs. IARC Press. Lyon, France, pp16–18 Edge SB, Compton CC, 2009, AJCC Cancer Staging Manual, 7th edn. Springer Verlag, New York, pp 547–560 Fuhrman SA, Lasky LC, Limas C, 1982, Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol 6:655–663 Hagemann T, Gunawan B, Schulz M, Fuzesi L, Binder C, 2001, mRNA expression of matrix metalloproteases and their inhibitors differs in subtypes of renal cell carcinomas. Eur J Urol 37:1839– 1846 Hemmerlein B, Johanns U, Halbfass J, Bottcher T, Heuser M, Radzun HJ et al, 2004, The balance between MMP-2/−9 and TIMP-1/2 is shifted towards MMP in renal cell carcinomas and can be further disturbed by hydrogen peroxide. Int J Oncol 24(5): 1069–1076 International Agency for Research on Cancer. GLOBOCAN 2012. Kidney - estimated incidence, all ages: both sexes. http://globocan.iarc.fr/old/summary_table_site-html.asp?selection=10210&title= Kidney&sex=0&type=0&window=1&africa=1&america= 2&asia=3&europe=4&oceania=5&build=6&sort=0&submit=% C2%A0Execute%C2%A0. Accessed 29 Apr 2016 Kallakury BVS, Karikehalli S, Haholu A, Sheehan CE, Azumi N, Ross JS, 2001, Increased expression of matrix metalloproteinases 2 and 9 and tissue inhibitors of metalloproteinases 1 and 2 correlate with poor prognostic variables in renal cell carcinoma. Clin Cancer Res 7:3113–3119 Kawata N, Nagane Y, Hirakata H, Ichinose T, Okada Y, Yamaguchi K et al, 2007, Significant relationship ofmatrix metalloproteinase 9 with nuclear grade and prognostic impact of tissue inhibitor of metalloproteinase 2 for incidental clear cell renal cell carcinoma. Urology 69(6):1049–1043 Klein T, Bischoff R, 2011, Physiology and pathophysiology of matrix metalloproteases. Amino Acids 41:271–290 Kugler A, Hemmerlein B, Thelen M, Kallerhof M, Radzun HJ, Ringert RH, 1998, Expression of metalloproteinases 2 and 9 and their inhibitors in renal cell carcinoma. J Urol 160:1914–1918 Lein M, Jung K, Laube C, Hubner T, Winkelmann B, Stephan C et al, 2000)Matrixmetalloproteinases and their inhibitors in plasma and tumor tissue of patients with renal cell carcinoma. Int J Cancer 85:801–804 Lin H, Pan JC, Zhang FM, Huang B, Chen X, Zhuang JT et al, 2015, Matrix metalloproteinase-9 is required for vasculogenic mimicry by clear renal carcinoma cells. Urol Oncol 33(4):168 e9-16 Liotta LA, 1986, Tumor invasion and Metastases-Role of Extracellular Matrix: Rhoads Memorial Award Lecture. Cancer Res 46(1):1–7 Lu H, Yang Z, Zhang H, Gan M, Zhou T, Wang S, 2013, The expression and clinical significance of matrix metalloproteinase 7 and tissue inhibitor of matrix metalloproteinases 2 in clear cell renal cell carcinoma. Exp Ther Med 5(3):890–896 Lu H, Cao X, Zhang H, Sun G, Fan G, Chen L et al, 2014, Imbalance between MMP-2, 9 and TIMP-1 promote the invasion and metastasis of renal cell carcinoma via SKP2 signaling pathways. Tumor Biol 35(10):9807–9813 Miyake H, Nishikawa M, Tei H, Furukawa J, Harada K, Fujisawa M, 2014, Significance of circulating matrix metalloproteinase-9 to tissue inhibitor of metalloproteinases-2 ratio as a predictor of disease progression in patients with metastatic renal cell carcinoma receiving sunitinib. Urol Oncol 32(5):548–548 Ohba K, Miyata Y, Koga S, Kanda S, Kanetake H, 2005, Expression of nm23-H1 product in sarcomatous cancer cells of renal cell carcinoma: correlation with tumor stage and expression of matrix metalloproteinase-2, matrix metalloproteinase-9, sialyl Lewis X, and c-erbB-2. Urology 65(5):1029–1034 Pozzi A, LeVine WF, Gardner HA, 2002, Low plasma levels of matrix metalloproteinase 9 permit increased tumor angiogenesis. Oncogene 21:272–281 Quiao ZK, Li YL, Lu HT,Wang KL, XuWH, 2013, Expression of tissue levels of matrix metalloproteinases and tissue inhibitors of metalloproteinases in renal cell carcinoma. World J Sur Oncol 3(11):1–6 Slaton JW, Inoue K, Perrotte P, El-NaggarAK, SwansonDA, Fidler IJ et al, 2001, Expression levels of genes that regulate metastasis and angiogenesis correlate with advanced pathological stage of renal cell carcinoma. Am J Pathol 158(2):735–743 Sun M, Shariat SF, Cheng C, Ficarra V, Murai M, Oudard S et al, 2011, Prognostic Factors and Predictive Models in Renal Cell Carcinoma: A Contemporary Review. Eur Urol 60:644–661 Takahashi M, Oka N, Narroda T, Nishitani MA, Kanda K, Kanayama HO et al, 2002, Prognostic significance of matrix metalloproteinases-2 activation ratio in renal cell carcinoma. Int J Urol 9(10):531–538 Walther MM, Kleiner DE, Lubensky IA, Pozzatti R, Nyguen T, Gnarra JR et al, 1997, Progelatinase A mRNA expression in cell lines derived from tumors in patients with metastatic renal cell carcinoma correlates inversely with survival. Urology 50:295–301 Yang SD, Sun RC, MuHJ, Xu ZQ, Zhou ZY, 2010, The expression and clinical significance of TGF-beta 1 and MMP2 in human renal clear cell carcinoma. Int J Surg Pathol 18(2):85–93 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 289 EP 299 DI 10.1007/s12253-017-0339-7 PG 11 ER PT J AU Awasthi, A Kumar, P Srikanth, VCh Sahi, Sh Puria, R AF Awasthi, Ankita Kumar, Pharvendra Srikanth, V Chittur Sahi, Shakti Puria, Rekha TI Invitro Evaluation of Torin2 and 2, 6-Dihydroxyacetophenone in Colorectal Cancer Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mTOR; Kinase inhibitor; Drug development; Docking; Natural Compound ID mTOR; Kinase inhibitor; Drug development; Docking; Natural Compound AB Colorectal cancer (CRC) is one of the most prevalent cancers diagnosed worldwide. Despite recent advances, resistance to cytotoxic and targeted therapy remains one of the greatest challenges in long-term management of colorectal cancer therapy. Recently established role of mTOR signaling in proliferation of CRC has incited for evaluation of mTOR kinase specific inhibitors in CRC therapy. Second generation mTOR kinase inhibitors including Torin2 has demonstrated efficient anticancer properties against variety of cancers and are in various stages of drug development. The time and financial constraints concomitant from discovery to development of efficient chemical inhibitors has redirected attention towards investigation of wide spread naturally occurring largely inexpensive compounds for their therapeutic potential. One such naturally occurring compound acetophenone derivative polyphenolic compound 2, 6-Dihydroxyacetophenone (DHAP) inhibits cell growth in different conditions.We investigated anticancer properties of both Torin2 and DHAP against colorectal cancer in HCT8 cell lines. Both Torin2 and DHAP inhibited growth of CRC cells at different concentrations by restricting multiple cellular functions e.g., cell cycle progression, cell migration and induced apoptosis. Treatment of HCT8 cells with natural compound DHAP resulted in reduced expression of mTOR pathway specific genes p70S6K1 and AKT1. In silico docking studies showed affinity of DHAP to mTOR kinase like Torin2. Taken together, our result vouches for role of Torin2 in CRC therapy and recommends DHAP an mTOR inhibitor, as a potential lead in the development of new therapeutic regimes against colorectal cancer. C1 [Awasthi, Ankita] Gautam Buddha University, School of Biotechnology, Greater NOIDA, 201312 Gautam Budh Nagar, India. [Kumar, Pharvendra] NCR Biotech Cluster, Regional Centre for Biotechnology, 3rd Milestone, Gurgaon-Faridabad Highway, Village BhankariFaridabad, Haryana, India. [Srikanth, V Chittur] NCR Biotech Cluster, Regional Centre for Biotechnology, 3rd Milestone, Gurgaon-Faridabad Highway, Village BhankariFaridabad, Haryana, India. [Sahi, Shakti] Gautam Buddha University, School of Biotechnology, Greater NOIDA, 201312 Gautam Budh Nagar, India. [Puria, Rekha] Gautam Buddha University, School of Biotechnology, Greater NOIDA, 201312 Gautam Budh Nagar, India. RP Puria, R (reprint author), Gautam Buddha University, School of Biotechnology, 201312 Gautam Budh Nagar, India. EM rpuria@gbu.ac.in CR Lee C, Murray DW, Barber RT et al, 1998, Siegel Et Al 1998.Pdf. Deep Res Part II Top Stud Oceanogr 45:2489–2501 Society AC, 2014, Colorectal Cancer Facts & Figures 2014-2016. Color Cancer Facts Fig :1–32. , DOI 10.1101/gad.1593107 Guertin DA, Sabatini DM, 2005, An expanding role for mTOR in cancer. Trends Mol Med 11:353–361. , DOI 10.1016/j. molmed.2005.06.007 Liu Q, Thoreen C,Wang J et al, 2009)MTORmediated anti-cancer drug discovery. Drug Discov Today Ther Strateg 6:47–55. https:// doi.org/10.1016/j.ddstr.2009.12.001 Benjamin D, Colombi M, Moroni C, Hall MN, 2011, Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov 10:868–880. , DOI 10.1038/nrd3531 Manning G,Whyte DB, Martinez R et al, 2002, The protein kinase complement of the human genome. Science 298:1912–1934. , DOI 10.1126/science.1075762 LoPiccolo J, Blumenthal GM, Bernstein WB, Dennis PA, 2008, Targeting the PI3K/Akt/mTOR pathway: Effective combinations and clinical considerations. Drug Resist Updat 11:32–50. https:// doi.org/10.1016/j.drup.2007.11.003 Efeyan A, Sabatini DM, 2010, MTOR and cancer: Many loops in one pathway. Curr Opin Cell Biol 22:169–176. , DOI 10. 1016/j.ceb.2009.10.007 Murugan AK, Alzahrani A, Xing M, 2013, Mutations in critical domains confer the human mTOR gene strong tumorigenicity. J Biol Chem 288:6511–6521. , DOI 10.1074/jbc.M112. 399485 Grabiner BC, Nardi V, Birsoy K et al, 2014, A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity. Cancer Discov 4:554–563. https:// doi.org/10.1158/2159-8290.CD-13-0929 Li W, Chang J, Wang S, et al, 2015, miRNA-99b-5p suppresses liver metastasis of colorectal cancer by down-regulating mTOR. 6 Chresta CM, Davies BR, Hickson I et al, 2010, AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res 70:288–298. , DOI 10. 1158/0008-5472.CAN-09-1751 Fujishita T, Aoki K, Lane HA et al, 2008, Inhibition of the mTORC1 pathway suppresses intestinal polyp formation and reduces mortality in ApcDelta716 mice. Proc Natl Acad Sci U S A 105:13544–13549. , DOI 10.1073/pnas.0800041105 Roulin D, Waselle L, Dormond-Meuwly A et al, 2011, Targeting renal cell carcinoma with NVP-BEZ235, a dual PI3K/mTOR inhibitor, in combination with sorafenib. Mol Cancer 10:90. https://doi. org/10.1186/1476-4598-10-90 Jiang BH, Liu LZ, 2008, Role of mTOR in anticancer drug resistance: Perspectives for improved drug treatment. Drug Resist Updat 11:63–76. , DOI 10.1016/j.drup.2008.03.001 Hammond WA, Swaika A, Mody K, 2015, Pharmacologic resistance in colorectal cancer: a review. Ther Adv Med Oncol. https:// doi.org/10.1177/1758834015614530 Liu Q, Xu C, Kirubakaran S, Zhang X, Hur W, Liu Y, et al, 2013, Characterization of Torin2, an ATP-competitive inhibitor ofmTOR, ATM, and ATR. Cancer Res 73:2574–2586 Wang C, Wang X, Su Z, et al, 2015, The novel mTOR inhibitor Torin-2 induces autophagy and downregulates the expression of UHRF1 to suppress hepatocarcinoma cell growth. 1708–1716. , DOI 10.3892/or.2015.4146 Simioni C, Cani A, Martelli AM, et al Activity of the novel mTOR inhibitor Torin-2 in B-precursor acute lymphoblastic leukemia and its therapeutic potential to prevent Akt reactivation. 5 Ahmed M, Hussain AR, Bavi P et al, 2014, High prevalence of mTOR complex activity can be targeted using Torin2 in papillary thyroid carcinoma. Carcinogenesis 35:1564–1572. , DOI 10.1093/carcin/bgu051 Udayakumar D, Pandita RK, Horikoshi N et al, 2016, Torin2 Suppresses Ionizing Radiation-Induced DNA Damage Repair. Radiat Res 538:527–538. , DOI 10.1667/RR14373.1 Martinez-Luis S, Perez-Vasquez A, Mata R, 2007, Natural products with calmodulin inhibitor properties. Phytochemistry 68:1882– 1903. , DOI 10.1016/j.phytochem.2007.02.025 Badr CE, Van Hoppe S, Dumbuya H et al, 2013, Targeting cancer cells with the natural compound obtusaquinone. J Natl Cancer Inst 105:643–653. , DOI 10.1093/jnci/djt037 Sigurdsson S, Ogmundsdottir HM, Hallgrimsson J, Gudbjarnason S, 2005, Antitumour activity of Angelica archangelica leaf extract. In: In Vivo, Brooklyn), vol 19, pp 191–194 Erdemgil FZ, Sanli S, Sanli N et al, 2007, Determination of pKa values of some hydroxylated benzoic acids in methanol-water binary mixtures by LC methodology and potentiometry. Talanta 72: 489–496. , DOI 10.1016/j.talanta.2006.11.007 Nakamura ES, Kurosaki F, Arisawa M, Mukainaka T, 2002, Cancer chemopreventive effects of constituents of Caesalpinia ferrea and related compounds. Cancer Lett 177:119–124 Pursh PC, 2014, Polyphenols with Anti-Proliferative Activities from Penthorum chinense Pursh.:11045–11055. , DOI 10. 3390/molecules190811045 Bracht K, Nicholls AM, Liu Y, Bodmer WF, 2010, 5-Fluorouracil response in a large panel of colorectal cancer cell lines is associated with mismatch repair deficiency. Brit J Cancer 103:340–346 Miller JM, 2003, GraphPad PRISM. Analysis 52:2–3. www. graphpad.com Sadowski SM, Boufraqech M, Zhang L et al, 2015, Torin2 targets dysregulated pathways in anaplastic thyroid cancer and inhibits tumor growth and metastasis. Oncotarget 6:18038–18049. https:// doi.org/10.18632/oncotarget.3833 Beck DE, 2009, Colorectal cancer. Rev Gastroenterol Mex 8:2–3. , DOI 10.1038/nrgastro.2011.208 Liang C-C, Park AY, Guan J-L, 2007, In vitro scratch assay: a convenient and inexpensivemethod for analysis of cellmigration in vitro. Nat Protoc 2:329–333. , DOI 10.1038/nprot.2007.30 Evan GI,VousdenKH(2001, Proliferation, cell cycle and apoptosis in cancer. Nature 411:342–348. , DOI 10.1038/35077213 Francipane MG, Lagasse E, 2013, mTOR pathway in colorectal cancer: an update. Oncotarget 5:49–66. , DOI 10.18632/ oncotarget.1548 Wang XW, Zhang YJ, 2014, Targeting mTOR network in colorectal cancer therapy.World J Gastroenterol 20:4178–4188. https://doi. org/10.3748/wjg.v20.i15.4178 Cai Z, Ke J, He X et al, 2014, Significance of mTOR signaling and its inhibitor against cancer stem-like cells in colorectal cancer. Ann Surg Oncol 21:179–188. , DOI 10.1245/s10434-013- 3146-8 PasstoorsWM, Beekman M, Deelen J et al, 2013, Gene expression analysis of mTOR pathway: Association with human longevity. Aging Cell 12:24–31. , DOI 10.1111/acel.12015 Riquelme I, Tapia O, Espinoza JA et al, 2016, The Gene Expression Status of the PI3K/AKT/mTOR Pathway in Gastric Cancer Tissues and Cell Lines. Pathol Oncol Res. , DOI 10.1007/s12253-016-0066-5 Wang N, Wu R, Cheng X et al, 2014, New insights into mTOR structure and regulation. Chinese Sci Bull 59:2927–2935. https:// doi.org/10.1007/s11434-014-0417-0 Friesner RA, Banks JL, Murphy RB et al, 2004, Glide: A New Approach for Rapid, Accurate Docking and Scoring. 1. Method and Assessment of Docking Accuracy. J Med Chem 47:1739– 1749. , DOI 10.1021/jm0306430 Friesner RA, Murphy RB, Repasky MP et al, 2006, Extra precision glide: Docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. J Med Chem 49:6177– 6196. , DOI 10.1021/jm051256o Link A, Balaguer F, Shen Yet al, 2013, Curcumin Modulates DNA Methylation in Colorectal Cancer Cells. PLoS One. , DOI 10.1371/journal.pone.0057709 ZhaoX, Song H, Zuo Z et al, 2013, Identification ofmiRNA-miRNA synergistic relationships in colorectal cancer. Int J BiolMacromol 55: 98–103. , DOI 10.1016/j.ijbiomac.2012.12.006 Blaser B, Waselle L, Dormond-Meuwly A et al, 2012, Antitumor activities of ATP-competitive inhibitors of mTOR in colon cancer cells. BMC Cancer 12:86. , DOI 10.1186/1471-2407-12-86 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 301 EP 309 DI 10.1007/s12253-017-0347-7 PG 9 ER PT J AU Ding, Y Sun, Ch Li, J Hu, L Li, M Liu, J Pu, L Xiong, Sh AF Ding, Yangyang Sun, Cheng Li, Jingrong Hu, Linhui Li, Manman Liu, Jun Pu, Lianfang Xiong, Shudao TI The Significance of Long Non-coding RNA HULC in Predicting Prognosis and Metastasis of Cancers: a Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lncRNA; HULC; Neoplasms; Prognosis; Metastasis; Meta-analysis ID lncRNA; HULC; Neoplasms; Prognosis; Metastasis; Meta-analysis AB Long non-coding RNAs (lncRNAs) have been demonstrated that they not only play important roles in tumorgenicity but also associate with cancer prognosis. Recently, highly up-regulated in liver cancer (HULC) is abnormally expressed in liver cancer and other cancers, and participated in cancers progression; however, it is unclear whether its expression is associated with prognosis. Here, we performed a meta-analysis and systematic review to evaluate the prognostic value and metastasis of HULC in various cancer patients. The meta-analysis was performed using a systematic search of PubMed,Web of Science, ScienceDirect and Wiley Online Library database to eligible studies. The pooled hazard ratios (HRs) with a 95% confidence interval (95% CI) were calculated to assess its prognosis and metastasis in human cancer. A total of 1134 patients from 11 studies were included. The results indicated that overexpression of HULC was associated with poor overall survival (OS) (HR = 1.89, 95% CI: 1.32–2.47). Furthermore, subgroup analysis showed that cancer type (digestive system cancer or non-digestive system cancers) and sample size (more or less than 100) significantly associated between HULC and OS. In addition, overexpression of HULC expression was significantly associated with metastasis in cancers (HR = 2.67, 95% CI: 0.94– 4.39). The meta-analysis indicated that lncRNA HULC could serve as a new molecular marker for cancer prognosis and metastasis. C1 [Ding, Yangyang] The Second Hospital of Anhui Medical University, Department of Hematology / Hematological LabHefei, Anhui, China. [Sun, Cheng] The Second Hospital of Anhui Medical University, Department of PharmacologyHefei, Anhui, China. [Li, Jingrong] The Second Hospital of Anhui Medical University, Department of EmergencyHefei, Anhui, China. [Hu, Linhui] The Second Hospital of Anhui Medical University, Department of Hematology / Hematological LabHefei, Anhui, China. [Li, Manman] The Second Hospital of Anhui Medical University, Department of Hematology / Hematological LabHefei, Anhui, China. [Liu, Jun] The Second Hospital of Anhui Medical University, Department of Hematology / Hematological LabHefei, Anhui, China. [Pu, Lianfang] The Third People’s Hospital of Bengbu, Department of HematologyBengbu, Anhui, China. [Xiong, Shudao] The Second Hospital of Anhui Medical University, Department of Hematology / Hematological LabHefei, Anhui, China. RP Xiong, Sh (reprint author), The Second Hospital of Anhui Medical University, Department of Hematology / Hematological Lab, Hefei, China. EM xshdao@ahmu.edu.cn CR Wu S, Powers S, Zhu W, Hannun YA, 2016, Substantial contribution of extrinsic risk factors to cancer development. Nature 529: 43–47 Garzon R, Volinia S, Papaioannou D, Nicolet D, Kohlschmidt J, Yan PS, Mrozek K, Bucci D, Carroll AJ, Baer MR, Wetzler M, Carter TH, Powell BL, Kolitz JE, Moore JO, Eisfeld AK, Blachly JS, Blum W, Caligiuri MA, Stone RM, Marcucci G, Croce CM, Byrd JC, Bloomfield CD, 2014, Expression and prognostic impact of lncRNAs in acute myeloid leukemia. Proc Natl Acad Sci U S A 111:18679–18684 Kim SH, Lee HW, Go SI, Lee SI, Lee GW, 2016, Clinical significance of the preoperative platelet count and platelet-to-lymphocyte ratio, PLT-PLR, in patients with surgically resected non-small cell lung cancer. Oncotarget 7:36198–36206 Chan JC, Chan DL, Diakos CI, Engel A, Pavlakis N, Gill A, Clarke SJ, 2016, The lymphocyte-to-monocyte ratio is a superior predictor of overall survival in comparison to established biomarkers of resectable colorectal cancer. Ann Surg 265:539–546 Han Li C, Chen Y, 2015, Small and long non-coding RNAs: novel targets in perspective cancer therapy. Curr Genomics 16:319–326 Ulitsky I, 2013, D.P. Bartel. lincRNAs: genomics, evolution, and mechanisms. Cell 154:26–46 Kapusta A, Feschotte C, 2014, Volatile evolution of long noncoding RNA repertoires: mechanisms and biological implications. Trends Genet 30:439–452 Ylipaa A, Kivinummi K, Kohvakka A, Annala M, Latonen L, Scaravilli M, Kartasalo K, Leppanen SP, Karakurt S, Seppala J, Yli-Harja O, Tammela TL, Zhang W, Visakorpi T, Nykter M, 2015, Transcriptome sequencing reveals PCAT5 as a novel ERG-regulated long noncoding RNA in prostate cancer. Cancer Res 75:4026–4031 Panzitt K, Tschernatsch MM, Guelly C, Moustafa T, Stradner M, Strohmaier HM, Buck CR, Denk H, Schroeder R, Trauner M, Zatloukal K, 2007, Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology 132:330–342 Sun X-H, Yang L-B, Geng X-L, Wang R, Zhang Z-C, 2015, Increased expression of IncRNA HULC indicates a poor prognosis and promotes cell metastasis in osteosarcoma. Int J Clin Exp Pathol 8:2994–3000 Wang YF, Zhang S, Li XQ,Wang Y, 2016, Expression of lncRNA HULC in cervical cancer and its correlation with tumor progression and patient survival. Eur Rev Med Pharmacol Sci 20:3987–3991 Yang XJ, Huang CQ, Peng CW, Hou JX, Liu JY, 2016, Long noncoding RNA HULC promotes colorectal carcinoma progression through epigenetically repressing NKD2 expression. Gene 592:172–178 Shi F, Xiao F, Ding P, Qin H, Huang R, Long Noncoding RNA, 2016, Highly up-regulated in liver cancer predicts unfavorable outcome and regulates metastasis by MMPs in triple-negative breast cancer. Arch Med Res 47:446–453 Peng W, Wu J, Feng J, 2016, Long noncoding RNA HULC predicts poor clinical outcome and represents pro-oncogenic activity in diffuse large B-cell lymphoma. Biomed Pharmacother 79:188–193 Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR, 2007, Practical methods for incorporating summary time-to-event data into meta-analysis. Trials 8:16 Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R, Jemal A, 2016, Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin 66:271–289 Fang K, Liu P, Dong S, Guo Y, Cui X, Zhu X, Li X, Jiang L, Liu T, Magnetofection YW, 2016, Based on superparamagnetic iron oxide nanoparticle-mediated low lncRNA HOTAIR expression decreases the proliferation and invasion of glioma stem cells. Int J Oncol 49:509–518 Gutschner T, Hammerle M, Eissmann M, Hsu J, Kim Y, Hung G, Revenko A, Arun G, Stentrup M, Gross M, Zornig M, MacLeod AR, Spector DL, Diederichs S, 2013, The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res 73:1180–1189 Du Y, Kong G, You X, Zhang S, Zhang T, Gao Y, Ye L, Zhang X, 2012, Elevation of highly up-regulated in liver cancer, HULC, by hepatitis B virus X protein promotes hepatoma cell proliferation via down-regulating p18. J Biol Chem 287:26302–26311 Zhang Y, Li Z, Zhang Y, Zhong Q, Chen Q, Zhang L, 2015, Molecular mechanism of HEIH and HULC in the proliferation and invasion of hepatoma cells. Int J Clin Exp Med 8:12956–12962 Ma Y, Huang D, Yang F, Tian M, Wang Y, Shen D, Wang Q, Chen Q, Zhang L, Long Noncoding RNA, 2016, Highly upregulated in liver cancer regulates the tumor necrosis factor-alpha-induced apoptosis in human vascular endothelial cells. DNA Cell Biol 35:296–300 Lu Z, Xiao Z, Liu F, Cui M, LiW, Yang Z, Li J, Ye L, Zhang X, 2016, Long non-coding RNA HULC promotes tumor angiogenesis in liver cancer by up-regulating sphingosine kinase 1, SPHK1). Oncotarget 7:241–254 Cui M, Xiao Z, Wang Y, Zheng M, Song T, Cai X, Sun B, Ye L, ZhangX, 2015, Long noncoding RNAHULC modulates abnormal lipid metabolism in hepatoma cells through an miR-9-mediated RXRA signaling pathway. Cancer Res 75:846–857 Maciel Uzan VR, Lengert AvH, Boldrini E, Penna V, Scapulatempo-Neto C, Scrideli CA, de Moraes Filho AP, Bezerra Cavalcante CE, de Oliveira CZ, Lopes LF, Vidal DO, 2016, High expression of HULC is associated with poor prognosis in osteosarcoma patients. PLoS One 11:e0156774 Li D, Liu X, Zhou J, Hu J, Zhang D, Liu J, Qiao Y, Zhan Q, LncRNA HULC, 2016, Modulates the phosphorylation of YB-1 through serving as a scaffold of ERK and YB-1 to enhance hepatocarcinogenesis. Hepatology 65:1612–1627 Yu X, Zheng H, Chan MT, Wu WK, 2016, HULC: an oncogenic long non-coding RNA in human cancer. J Cell Mol Med 21:410– 417 Yang Z, Lu Y, Xu Q, Tang B, Park CK, Chen X, 2015, HULC and H19 played different roles in overall and diseasefree survival from hepatocellular carcinoma after curative hepatectomy: a preliminary analysis from gene expression omnibus. Dis Markers 2015:191029 Li S-P, Xu H-X, Yu Y, He J-D,Wang Z, Xu Y-J,Wang C-Y, Zhang H-M, Zhang R-X, Zhang J-J, Yao Z, Shen Z-Y, LncRNA HULC, 2016, Enhances epithelial-mesenchymal transition to promote tumorigenesis and metastasis of hepatocellular carcinoma via the miR-200a-3p/ZEB1 signaling pathway. Oncotarget 7:42431–42446 Yan H, Tian R, ZhangM,Wu J, Ding M, He J, 2017, High expression of long noncoding RNA HULC is a poor predictor of prognosis and regulates cell proliferation in glioma. Onco Targets Ther 10: 113–120 Jin C, ShiW,Wang F, Shen X, Qi J, Cong H, Yuan J, Shi L, Zhu B, Luo X, Zhang Y, Long SJ, 2016, Non-coding RNA HULC as a novel serum biomarker for diagnosis and prognosis prediction of gastric cancer. Oncotarget 7:51763–51772 Peng W, Gao W, Feng J, 2014, Long noncoding RNA HULC is a novel biomarker of poor prognosis in patients with pancreatic cancer. Med Oncol 31:346 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 311 EP 318 DI 10.1007/s12253-017-0351-y PG 8 ER PT J AU Legras, A Roussel, H Mangiameli, G Arame, A Grand, B Pricopi, C Badia, A Gibault, L Badoual, C Fabre, E Laurent-Puig, P Blons, H Le Pimpec-Barthes, F AF Legras, Antoine Roussel, Helene Mangiameli, Giuseppe Arame, Alex Grand, Bertrand Pricopi, Ciprian Badia, Alain Gibault, Laure Badoual, Cecile Fabre, Elizabeth Laurent-Puig, Pierre Blons, Helene Le Pimpec-Barthes, Francoise TI Mutational Diversity of Lung Cancer and Associated Lymph Nodes. An Exploratory Prospective Study of 4 Resected cIIIA-N2 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung neoplasms; Lymph nodes; Genetics; Biomarkers; tumour; High-throughput nucleotide sequencing ID Lung neoplasms; Lymph nodes; Genetics; Biomarkers; tumour; High-throughput nucleotide sequencing AB Mutational heterogeneity could explain different metastatic patterns among IIIA-N2 lung cancer and influence prognosis. The identification of subclonal mutations using deep sequencing to evaluate the degree of molecular heterogeneity may improve IIIA-N2 classification. The aim of this prospective study was to assess mutational and immunohistochemical characteristics in primary tumours and involved lymph nodes (LN) in operated patients. Four patients operated for primary lung carcinoma and unisite N2 mediastinal involvement were consecutively selected. Samples (tumour and paired LN) were analysed for PD1, PD-L1 and CD8 immunostaining. Somatic mutation testing was performed by deep targeted next generation sequencing (NGS), with the AmpliSeq™ Colon and Lung Cancer Panel (LifeTechnology). A total of 9 primary lung cancer samples and 10 LN stations were analysed. For each cancer, we found 2 mutations, with allelic ratios from 3% to 72%. Mutational patterns were heterogeneous for 2 primary tumours. In 3 cases, mutations observed in the primary tumour were not found in LN metastases (ALK, FGFR3, MET). Inversely, in 1 case, a KRAS mutation was found in LN but not in the primary tumour. All primary tumours were found PD-L1 positive while CD8+ Tcells infiltrate varied. In the different examined LN samples, PD-L1 expression, CD8+ and PD1+ Tcells infiltrate were not similar to the primary tumour. This preliminary prospective study shows the diversity of intra-tumour and LN mutations using routinelyused targeted NGS, concerning both mutated gene and allelic ratio. Further studies are needed to evaluate its prognostic impact. C1 [Legras, Antoine] Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Thoracic Surgery and Lung Transplantation Department, 20 rue Leblanc, 75908 Paris, France. [Roussel, Helene] Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Pathology DepartmentParis, France. [Mangiameli, Giuseppe] Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Thoracic Surgery and Lung Transplantation Department, 20 rue Leblanc, 75908 Paris, France. [Arame, Alex] Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Thoracic Surgery and Lung Transplantation Department, 20 rue Leblanc, 75908 Paris, France. [Grand, Bertrand] Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Thoracic Surgery and Lung Transplantation Department, 20 rue Leblanc, 75908 Paris, France. [Pricopi, Ciprian] Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Thoracic Surgery and Lung Transplantation Department, 20 rue Leblanc, 75908 Paris, France. [Badia, Alain] Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Thoracic Surgery and Lung Transplantation Department, 20 rue Leblanc, 75908 Paris, France. [Gibault, Laure] Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Pathology DepartmentParis, France. [Badoual, Cecile] Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Pathology DepartmentParis, France. [Fabre, Elizabeth] Paris-Descartes University, Sorbonne Paris Cite University, Saints-Peres Research Center, INSERM UMR-S1147, CNRS SNC 5014Paris, France. [Laurent-Puig, Pierre] Paris-Descartes University, Sorbonne Paris Cite University, Saints-Peres Research Center, INSERM UMR-S1147, CNRS SNC 5014Paris, France. [Blons, Helene] Paris-Descartes University, Sorbonne Paris Cite University, Saints-Peres Research Center, INSERM UMR-S1147, CNRS SNC 5014Paris, France. [Le Pimpec-Barthes, Francoise] Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Thoracic Surgery and Lung Transplantation Department, 20 rue Leblanc, 75908 Paris, France. RP Le Pimpec-Barthes, F (reprint author), Assistance Publique Hopitaux de Paris, Georges Pompidou European Hospital, Thoracic Surgery and Lung Transplantation Department, 75908 Paris, France. EM francoise.lepimpec-barthes@aphp.fr CR Asamura H, Chansky K, Crowley J et al, 2015, The International Association for the Study of Lung Cancer lung cancer staging project: proposals for the revision of the N descriptors in the forthcoming 8th edition of the TNM classification for lung cancer. J Thorac Oncol 10:1675–1684 Decaluwe H, De Leyn P, Vansteenkiste J et al, 2009, Surgical multimodality treatment for baseline resectable stage IIIA-N2 non-small cell lung cancer. Degree of mediastinal lymph node involvement and impact on survival. Eur J Cardiothorac Surg 36: 433–439 Arame A, Mordant P, Riquet M, 2014, Pneumonectomy for stage IIIA NSCLC: a chance, not a calamity. Ann Thorac Surg 97:382 Riquet M, Mordant P, Pricopi C et al, 2014, A review of 250 tenyear survivors after pneumonectomy for non-small-cell lung cancer. Eur J Cardiothorac Surg 45:876–881 Legras A, Mordant P, Arame A et al, 2014, Long-term survival of patients with pN2 lung cancer according to the pattern of lymphatic spread. Ann Thorac Surg 97:1156–1162 Zhang J, Fujimoto J, Zhang J et al, 2014, Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science 346:256–259 De Bruin EC, McGranahan N, Mitter R et al, 2014, Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science 346:251–256 Um S-W, Joung J-G, Lee H et al, 2016, Molecular evolution patterns in metastatic lymph nodes reflect the differential treatment response of advanced primary lung cancer. Cancer Res 76:6568–6576 Pecuchet N, Legras A, Laurent-Puig P, BlonsH, 2016, Lung cancer molecular testing, what role for next generation sequencing and circulating tumor DNA. Ann Pathol 36:80–93 Garon EB, Rizvi NA, Hui R et al, 2015, Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 372:2018–2028 Mountain CF, Dresler CM, 1997, Regional lymph node classification for lung cancer staging. Chest 111:1718–1723 Riquet M, Bagan P, Le Pimpec Barthes F et al, 2007, Completely resected non-small cell lung cancer: reconsidering prognostic value and significance of N2 metastases. Ann Thorac Surg 84:1818–1824 Kozower BD, Larner JM, Detterbeck FC, Jones DR, 2013, Special treatment issues in non-small cell lung cancer: diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 143: e369S–e399S Robinson LA, Ruckdeschel JC,WagnerH et al, 2007, Treatment of non-small cell lung cancer-stage IIIA: ACCP evidence-based clinical practice guidelines, 2nd edition). Chest 132:243S–265S Cancer bronchique non a petites cellules - Referentiel national de RCP - Ref, 2017, RECOKBRNONPETCEL15 | Institut National Du Cancer. http://www.e-cancer.fr/Expertises-et-publications/ Catalogue-des-publications/Cancer-bronchique-non-a-petitescellules- Referentiel-national-de-RCP. Accessed 26 June 2017 Jamal-Hanjani M, Quezada SA, Larkin J, Swanton C, 2015, Translational implications of tumor heterogeneity. Clin Cancer Res 21:1258–1266 Hiley C, de Bruin EC, McGranahan N, Swanton C, 2014, Deciphering intratumor heterogeneity and temporal acquisition of driver events to refine precision medicine. Genome Biol 15:453 Baldia PH,Maurer A, Heide Tet al, 2016, Fibroblast growth factor receptor, FGFR, alterations in squamous differentiated bladder cancer: a putative therapeutic target for a small subgroup. Oncotarget 7: 71429–71439 Shirole NH, Pal D, Kastenhuber ER et al, 2016, TP53 exon-6 truncating mutations produce separation of function isoforms with pro-tumorigenic functions. elife 6:e25532 Benderra M-A, Aspeslagh S, Postel-Vinay S et al, 2016, Acquired EGFR mutation as the potential resistance driver to Crizotinib in a MET-mutated tumor. J Thorac Oncol 11:e21–e23 Waqar SN, Cottrell CE, Morgensztern D, 2015, MET mutation associated with responsiveness to crizotinib. J Thorac Oncol 10:e29–e31 Koh J, Go H, Keam B et al, 2015, Clinicopathologic analysis of programmed cell death-1 and programmed cell death-ligand 1 and 2 expressions in pulmonary adenocarcinoma: comparison with histology and driver oncogenic alteration status. Mod Pathol 28:1154–1166 D’Incecco A, Andreozzi M, Ludovini Vet al, 2015, PD-1 and PDL1 expression in molecularly selected non-small-cell lung cancer patients. Br J Cancer 112:95–102 Vieira T, Antoine M, Hamard C et al, 2016, Sarcomatoid lung carcinomas show high levels of programmed death ligand-1, PDL1, and strong immune-cell infiltration by TCD3 cells and macrophages. Lung Cancer Amst Neth 98:51–58 Uruga H, Bozkurtlar E, Huynh TG et al, 2016, Programmed cell death ligand, PD-L1, expression in stage II and III lung adenocarcinomas and nodal metastases. J Thorac Oncol 12:458–466 KimM-Y, Koh J, KimS et al, 2015, Clinicopathological analysis of PD-L1 and PD-L2 expression in pulmonary squamous cell carcinoma: comparison with tumor-infiltrating T cells and the status of oncogenic drivers. Lung Cancer Amst Neth 88:24–33 Taube JM, Klein A, Brahmer JR et al, 2014, Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 20: 5064–5074 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 319 EP 325 DI 10.1007/s12253-017-0352-x PG 7 ER PT J AU Yu, Yh Wei, Chy Qin, Qh Mo, Qg Huang, Z Lian, B AF Yu, Ying-hua Wei, Chang-yuan Qin, Qing-hong Mo, Qin-guo Huang, Zhen Lian, Bin TI Efficacy of Iodine-125 Seed Implantation in Locoregionally Recurrent and Unresectable Breast Cancer: a Retrospective Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Iodine-125 seed implantation; Brachytherapy; Locoregionally; Unresectable; Breast cancer ID Iodine-125 seed implantation; Brachytherapy; Locoregionally; Unresectable; Breast cancer AB The management of locoregionally recurrent and unresectable breast cancer is a therapeutic challenge. This retrospective study aimed to assess the efficacy of 125I seed implantation brachytherapy as a palliative management in locoregionally recurrent breast cancer. We analyzed 36 locoregionally recurrent and unresectable breast cancers in our hospital between 2012 and 2016. All patients were treated with CT-guided 125I seed permanent implantation. The dose distribution of 125I seeds was calculated using a computerized treatment planning system. Complete response, partial response, stable disease, and local tumor control rates were calculated. Long-term efficacy was assessed based on survival rates ranging from 1 to 4 years. The follow-up period ranged from 6 to 53 months. The median local control was 28 months (95% CI: 16.2–39.8 months). The percentage of patients who showed 6-month, 1-year, 2-year, and 3-year local control was 97.2%, 77.8%, 52.8%, and 33.3%, respectively. Median survival time for all patients was 48 months (95% CI: 40.9– 55.1 months); 1-year, 2-year, 3-year, and 4-year survival rates were 97.2%, 80.6%, 63.9%, and 46.5%, respectively. Pain relief response rate was 88.9%.No serious complications were detected during the follow-up period. The results of this study demonstrate that 125I seed implantation could be considered a feasible and promising minimally invasive therapy for locoregionally recurrent and unresectable breast carcinoma. C1 [Yu, Ying-hua] Affiliated Tumor Hospital of Guangxi Medical University, Departmant of Breast Surgery, NO. 71, He Di Lu, 530021 Nanning, Guangxi, China. [Wei, Chang-yuan] Affiliated Tumor Hospital of Guangxi Medical University, Departmant of Breast Surgery, NO. 71, He Di Lu, 530021 Nanning, Guangxi, China. [Qin, Qing-hong] Affiliated Tumor Hospital of Guangxi Medical University, Departmant of Breast Surgery, NO. 71, He Di Lu, 530021 Nanning, Guangxi, China. [Mo, Qin-guo] Affiliated Tumor Hospital of Guangxi Medical University, Departmant of Breast Surgery, NO. 71, He Di Lu, 530021 Nanning, Guangxi, China. [Huang, Zhen] Affiliated Tumor Hospital of Guangxi Medical University, Departmant of Breast Surgery, NO. 71, He Di Lu, 530021 Nanning, Guangxi, China. [Lian, Bin] Affiliated Tumor Hospital of Guangxi Medical University, Departmant of Breast Surgery, NO. 71, He Di Lu, 530021 Nanning, Guangxi, China. RP Wei, Chy (reprint author), Affiliated Tumor Hospital of Guangxi Medical University, Departmant of Breast Surgery, 530021 Nanning, China. EM weicy63@aliyun.com CR Khakpour N, Lucci A, 2005, Repeat breast-conserving surgery for in-breast local breast carcinoma recurrence: the potential role of partial breast irradiation. Women's Oncol Rev 5:75–76 Fisher B, Anderson S, Bryant J, Margolese RG, DeutschMet al, 2002, Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 347:1233–1241 Buchholz T, 2011, 3–29 Locoregional radiation therapy in patients with high-risk breast cancer receiving adjuvant chemotherapy: 20- year results of the British Columbia randomized trial. Breast Dis Year B Q 5:269–271 Cheng L, Swartz MD, Zhao H, Kapadia AS, Lai D et al, 2012, Hazard of recurrence among women after primary breast cancer treatment-a 10-year follow-up using data from SEER-Medicare. Cancer Epidemiol Biomarkers Prev 21:800–809 Siglin J, Champ CE, Vakhnenko Y, Anne PR, Simone NL, 2012, Radiation therapy for locally recurrent breast cancer. Int J Breast Cancer 2012:571946–571946 Rauschecker H, Clarke M, Gatzemeier W, Recht A, 2001, Systemic therapy for treating locoregional recurrence in women with breast cancer. Cochrane Database Syst Rev, 4):CD002195. , DOI 10.1002/14651858.CD002195 Li Y-f, Z-q L, Y-s Z, L-m D, C-y W et al, 2016, Implantation of radioactive 125 I seeds improves the prognosis of locally advanced pancreatic cancer patients: a retrospective study. J Huazhong Univ Sci Technol Med Sci 36:205–210 Niu L, Luo X, Zeng J, Kong X, Fang G et al, 2016, Cryoablation combined with Iodine-125 implantation in the treatment of cardiac metastasis from alveolar soft part sarcoma: a case report. Biomedicine Hub 1:4–4 Duan C, Zheng L, Jin M, Zhang D, ZhaoWet al, 2016, Combined chemotherapy and 125 I-particle implantation for treatment of children with head and neck soft tissue sarcomas improve the shortterm efficacy: Beijing Children's hospital experience over 2 years. Int J Clin Exp Med 9:11901–11906 Mahmood U, Blanchard P, Pugh TJ, Swanson D, GraberWet al, 2016, Prospective cohort of permanent seed implantation prostate brachytherapy for intermediate risk prostate cancer: analysis of patient satisfaction and interference with daily activities between 125-iodine, 103- palladium and 131-cesium. Brachytherapy 15:S201–S202 Wang JJ, Yuan HS, Li JN, Jiang YL, Tian SQ et al, 2010, CTguided radioactive seed implantation for recurrent rectal carcinoma after multiple therapy. Med Oncol 27:421–429 Cox CE, Furman B, Stowell N, Ebert M, Clark J et al, 2003, Radioactive seed localization breast biopsy and lumpectomy: can specimen radiographs be eliminated? Ann Surg Oncol 10:1039–1047 van Riet YE, Maaskant AJ, Creemers GJ, van Warmerdam LJ, Jansen FH et al, 2010, Identification of residual breast tumour localization after neo-adjuvant chemotherapy using a radioactive 125 iodine seed. Eur J Surg Oncol 36:164–169 Mullins JP, Grams MP, Herman MG, Brinkmann DH, Antolak JA, 2016, Treatment planning for metals using an extended CT number scale. J Appl Clin Med Phys 17(6):179–188 Alnaghy S, Loo K, Cutajar D, Jalayer M, Tenconi C et al, 2016, BrachyView: multiple seed position reconstruction and comparison with CT post-implant dosimetry. J Instrum 11:P05002 Miller A, Hoogstraten B, Staquet M, Winkler A, 1981, Reporting results of cancer treatment. Cancer 47:207–214 pain Coc, 1986, Descriptions of chronic pain syndromes and definitions of pain terms: prepared by the International Association for the Study of Pain, subcommittee on taxonomy. Pain Suppl 3:S1–226 Cardoso F, Harbeck N, Fallowfield L, Kyriakides S, Senkus E, et al., 2012, Locally recurrent or metastatic breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Annals of oncology 23: vii11-vii19 Dahlstrom KK, Andersson AP, Andersen M, Krag C, 1993, Wide local excision of recurrent breast cancer in the thoracic wall. Cancer 72:774–777 Bethke KP, 1996, Breast conservation: predictors and treatment of local recurrence. Wiley online Library pp. 332–338 Petrella F, Radice D, Borri A, Galetta D, Gasparri R, Casiraghi M et al, 2014, Chest wall resection and reconstruction for locally recurrent breast cancer: from technical aspects to biological assessment. Surg J R Coll Surg Edinb Irel 14(1):26 Salvadori B, Marubini E, Miceli R, Conti A, Cusumano F et al, 1999, Reoperation for locally recurrent breast cancer in patients previously treated with conservative surgery. Br J Surg 86:84–87 Li W, Guan J, Yang L, Zheng X, Yu Y et al, 2015, Iodine-125 brachytherapy improved overall survival of patients with inoperable stage III/IV non-small cell lung cancer versus the conventional radiotherapy. Med Oncol 32:1–8 Reveiz L, Rueda JR, Cardona AF, 2012, Palliative endobronchial brachytherapy for non-small cell lung cancer. The Cochrane Library Shi L, Li X, Pei H, Zhao J, Qiang W et al, 2016, Phase II study of computed tomography-guided 125 I-seed implantation plus chemotherapy for locally recurrent rectal cancer. Radiother Oncol 118:375–381 Brahimaj B, Lamba M, Breneman JC, Warnick RE, 2016, Iodine- 125 seed migration within brain parenchyma after brachytherapy for brain metastasis: case report. J Neurosurg 125(5):1167–1170 Welch H, Gorski D, Albertsen P, Andriole G, Crawford E et al, 2016, Trends in metastatic breast and prostate cancer. N Engl J Med 2016:594–596 Dahiya M, 2016, Brachytherapy: A review. J Crit Rev 3:6–10 Wierzbicka M, Bartochowska A, Strnad V, Strojan P, Mendenhall WM et al, 2016, The role of brachytherapy in the treatment of squamous cell carcinoma of the head and neck. Eur Arch Otorhinolaryngol 273:269–276 Rasmusson E, Gunnlaugsson A, Kjellen E, Nilsson P, Einarsdottir M,Wieslander E, Fransson P, Ahlgen G, Blom R, 2016, Low-dose rate brachytherapy with I-125 seeds has an excellent 5-year outcome with few side effects in patients with low-risk prostate cancer. Acta Oncol 55(8):1016–1021 Huo X,Wang H,Yang J, Li X,YanWet al, 2016, Effectiveness and safety of CT-guided 125 I seed brachytherapy for postoperative locoregional recurrence in patients with non–small cell lung cancer. Brachytherapy 15:370–380 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 327 EP 332 DI 10.1007/s12253-017-0361-9 PG 6 ER PT J AU Polom, K Das, K Marrelli, D Roviello, G Pascale, V Voglino, C Rho, H Tan, P Roviello, F AF Polom, Karol Das, Kakoli Marrelli, Daniele Roviello, Giandomenico Pascale, Valeria Voglino, Costantino Rho, Henry Tan, Patrick Roviello, Franco TI KRAS Mutation in Gastric Cancer and Prognostication Associated with Microsatellite Instability Status SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Stomach cancer; KRAS mutation; Mismatch repair deficiency; Molecular; Prognosis ID Stomach cancer; KRAS mutation; Mismatch repair deficiency; Molecular; Prognosis AB Microsatellite instability (MSI) is one of the subgroups based on the new molecular classification of gastric cancer (GC). In this study, we analyzed the role of KRAS status in MSI GC and the impact of MSI status on KRAS mutation. We performed analysis on 595 GC patients. Polymerase chain reaction (PCR) was used for the screening of KRAS mutation (exon 2) and 5 quasi-monomorphic mononucleotide repeats, namely, BAT-26, BAT-25, NR -24, NR-21, and NR-27 were used to determine the MSI status. The KRAS and MSI status were then compared with clinicopathologic data of the GC patients. MSI GC was found in 20.3% of all cases. KRAS mutation was seen in 24 patients; 18 were MSI (75%) and 6 were microsatellite stable (MSS) (25%). MSI GC patients with KRAS mutation were older and mostly female, but MSS presented more advanced Tand N stage of the disease, more cardia tumors, and adjuvant treatment. Five-year survival was 72.2% for KRAS mutation patients with MSI and 0% for MSS (p < 0.001). Although KRAS mutations in GC are linked with MSI in the majority of cases, KRAS mutations with MSS status presented with a poor prognosis and a worse outcome. In multivariate analysis, MSI was associated with better survival (p < 0.001) but KRAS was with worse survival (p = 0.304). Our study suggests that KRAS mutations are based on MSI status rather than different codon subtypes ofmutation, and such a division could be used to determine the GC patient outcome. C1 [Polom, Karol] University of Siena, General Surgery and Surgical Oncology Department, viale Bracci 16, 53-100 Siena, Italy. [Das, Kakoli] Duke-NUS Medical School, Cancer and Stem Cell Biology Programme, 8 College Road, 169857 Singapore, Singapore. [Marrelli, Daniele] University of Siena, General Surgery and Surgical Oncology Department, viale Bracci 16, 53-100 Siena, Italy. [Roviello, Giandomenico] San Donato Hospital, Department of Oncology, Medical Oncology Unit, Via Nenni 20, 52100 Arezzo, Italy. [Pascale, Valeria] University of Siena, General Surgery and Surgical Oncology Department, viale Bracci 16, 53-100 Siena, Italy. [Voglino, Costantino] University of Siena, General Surgery and Surgical Oncology Department, viale Bracci 16, 53-100 Siena, Italy. [Rho, Henry] Poznan University of Medical SciencesPoznan, Poland. [Tan, Patrick] Duke-NUS Medical School, Cancer and Stem Cell Biology Programme, 8 College Road, 169857 Singapore, Singapore. [Roviello, Franco] University of Siena, General Surgery and Surgical Oncology Department, viale Bracci 16, 53-100 Siena, Italy. RP Polom, K (reprint author), University of Siena, General Surgery and Surgical Oncology Department, 53-100 Siena, Italy. EM polom.karol@gmail.com CR Ferlay J, Soerjomataram I, Dikshit R et al, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E366 Marrelli D, Polom K, de Manzoni G, Morgagni P, Baiocchi GL, Roviello F., 2015, Multimodal treatment of non-cardia gastric cancer in West: where are we going? World J Gastroenterol 21(26): 7954–7969 Roviello G, Polom K, Petrioli R et al, 2016, Monoclonal antibodies-based treatment in gastric cancer: current status and future perspectives. Tumour Biol 37(1):127–140 Bang, YJ, Van Cutsem E, Feyereislova A et al., 2010, Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer, ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376(9742):687–697 Cancer Genome Atlas Research Network et al, 2014, Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513(7517):202–209 Cristescu R, Lee J, Nebozhyn M et al, 2015, Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med 21(5):449–456 Isa N, 2014, Evidence based radiation oncology with existing technology. Rep Pract Oncol Radiother 19(6):259–266 Polom W, Markuszewski M, Rho YS, Matuszewski M, 2014, Usage of invisible near infrared light, NIR, fluorescence with indocyanine green, ICG, and methylene blue, MB, in urological oncology. Part 1. Cent Europe J Urol 67(2):142–148 Subhash VV, Yeo MS, Tan WL, Yong WP, 2015, Strategies and Advancements in Harnessing the Immune System for Gastric Cancer Immunotherapy. J Immunol Res 2015:308574. https://doi. org/10.1155/2015/308574 Yamamoto H, Adachi Y, Taniguchi H et al, 2012, Interrelationship between microsatellite instability and microRNA in gastrointestinal cancer. World J Gastroenterol 18:2745–2755 Beghelli S, de Manzoni G, Barbi S et al, 2006, Microsatellite instability in gastric cancer is associated with better prognosis in only stage II cancers. Surgery 139:347–356 Corso G, Pedrazzani C, Marrelli D, Pascale V, Pinto E, Roviello F, 2009, Correlation of microsatellite instability at multiple loci with long-term survival in advanced gastric carcinoma. Arch Surg 144: 722–727 Marrelli D, Polom K, Pascale V et al, 2016, Strong Prognostic Value of Microsatellite Instability in Intestinal Type Non-cardia Gastric Cancer. Ann Surg Oncol 23(3):943–950 Polom K,Marano L, Marrelli D, De Luca R, Roviello G, Savelli V, Tan P, Roviello F, 2017, Meta-analysis of microsatellite instability on clinicopathological characteristics and overall survival in gastric cancer. Br J Surg. , DOI 10.1002/bjs.10663 Misale S, Yaeger R, Hobor S et al, 2012, Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 486(7404):532–536 Heindl S, Eggenstein E, Keller S et al, 2012, Relevance of MET activation and genetic alterations of KRAS and E-cadherin for cetuximab sensitivity of gastric cancer cell lines. J Cancer Res Clin Oncol 138(5):843–858 Shi M, Shi H, Ji J et al, 2014, Cetuximab inhibits gastric cancer growth in vivo, independent of KRAS status. Curr Cancer Drug Targets 14(2):217–224 Lee SH, Lee JW, Soung YH et al, 2003, BRAF and KRAS mutations in stomach cancer. Oncogene 22(44):6942–6945 Brennetot C, Duval A, Hamelin R et al, 2003, Frequent Ki-ras mutations in gastric tumors of the MSI phenotype.Gastroenterology 125(4): 1282 Oliveira C, Pinto M, Duval A et al, 2003, BRAF mutations characterize colon but not gastric cancer with mismatch repair deficiency. Oncogene 22(57):9192–9196 Oliveira C, Velho S, Moutinho C et al, 2007, KRAS and BRAF oncogenic mutations in MSS colorectal carcinoma progression. Oncogene 26:158–163 Boland CR, Thibodeau SN, Hamilton SR et al, 1998, A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 58(22):5248–5257 Janjigian YY, Werner D, Pauligk C et al, 2012, Prognosis of metastatic gastric and gastroesophageal junction cancer by HER2 status: a European and USA International collaborative analysis. Ann Oncol 23(10):2656–2662 Fuchs CS, Tomasek J, Yong CJ et al, 2014, Ramucirumab monotherapy for previously treated advanced gastric or gastrooesophageal junction adenocarcinoma, REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 383(9911):31–39 Gao J, Aksoy BA, Dogrusoz U et al, 2013, Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6(269):pl1 Cerami E, Gao J, Dogrusoz U et al, 2012, The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Canc Discov 2(5):401–404 Queiros P, Pinheiro H, Carvalho J et al, 2015, KRAS mutations in microsatellite instable gastric tumours: impact of targeted treatment and intratumoural heterogeneity. Virchows Arch 467(4):383–392 van Grieken NC, Aoyama T, Chambers PA, et al., 2013)KRAS and BRAF mutations are rare and related to DNA mismatch repair deficiency in gastric cancer from the East and theWest: results from a large international multicentre study. Br J Cancer 108(7):1495– 1501., DOI , DOI 10.1038/bjc.2013.109 Zhao W, Chan TL, Chu KM et al, 2004, Mutations of BRAF and KRAS in gastric cancer and their association with microsatellite instability. Int J Cancer 108(1):167–169 Velho S, Moutinho C, Cirnes L et al, 2008, BRAF, KRAS and PIK3CA mutations in colorectal serrated polyps and cancer: primary or secondary genetic events in colorectal carcinogenesis? BMC Cancer 8:255. , DOI 10.1186/1471-2407-8-255 Duggan BD, Felix JC, Muderspach LI et al, 1994, Microsatellite instability in sporadic endometrial carcinoma. J Natl Cancer Inst 86: 1216–1221 de Cuba EM, Snaebjornsson P, Heideman DA et al, 2016, Prognostic value of BRAF and KRAS mutation status in stage II and III microsatellite instable colon cancers. Int J Cancer 138(5): 1139–1145 Nout RA, Bosse T, Creutzberg CL et al, 2012, Improved risk assessment of endometrial cancer by combined analysis of MSI, PI3K-AKT, Wnt/β-catenin and P53 pathway activation. Gynecol Oncol 126(3):466–473. , DOI 10.1016/j.ygyno.2012.05. 012 Yokota T, Shibata N, Ura Tet al, 2010, Cycleave polymerase chain reaction method is practically applicable for V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog, KRAS)/V-raf murine sarcoma viral oncogene homolog B1, BRAF, genotyping in colorectal cancer. Transl Res 156:98–105 Warneke VS, Behrens HM, Haag J et al, 2013, Prognostic and putative predictive biomarkers of gastric cancer for personalized medicine. Diagn Mol Pathol 22(3):127–137 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 333 EP 340 DI 10.1007/s12253-017-0348-6 PG 8 ER PT J AU Ren, K Xin, G Xiao, M He, W Kang, J AF Ren, Ke Xin, Gou Xiao, Mingzhao He, Weiyang Kang, Jian TI Pim-2 Cooperates with Downstream Factor XIAP to Inhibit Apoptosis and Intensify Malignant Grade in Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pim-2; XIAP; Apoptosis; Malignant grade ID Pim-2; XIAP; Apoptosis; Malignant grade AB To find the exact downstream effector of Pim-2 pathway in prostate cancer cells, and to determine the means by which it affects prostate cancer. XIAP, Pim-2 and p-eIF4B expressions were detected in PCA and BPH tissues. Then the Pim-2 and XIAP expressions were manipulated using transfection or RNAi in prostatic cells. Finally, Pim-2/eIF4B/XIAP levels were examined in PCA tissues with different clinicopathologic features. XIAP was significantly higher in PCA than in BPH tissues. When Pim-2 was transfected into noncancerous prostate epithelial cells RWPE-1, Pim-2, p-eIF4B and XIAP were all significantly increased and the apoptosis rate was significantly decreased. When XIAP was transfected into RWPE-1 cells, XIAP was significantly increased with no impact on Pim-2, p-eIF4B and the apoptosis rate. When Pim-2 SiRNA was transfected into prostate cancer cells PC-3, Pim-2, p-eIF4B and XIAP were significantly decreased and the apoptosis rate was significantly increased. When XIAP SiRNA was transfected into PC-3 cells, XIAP was significantly decreased with no impact on Pim-2, p-eIF4B and apoptosis rate. Pim-2, p-eIF4B and XIAP were all significantly higher in PCA tissues with GS ≥8 than those with GS ≤7. XIAP is the downstream factor of Pim-2 pathway in prostate cancer cells. Pim-2 and XIAP cooperate in inhibiting the apoptosis of prostate cancer cells. The activation of Pim-2 pathway may predict higher GS in prostate cancer. C1 [Ren, Ke] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 1 Youyi Road, Yuzhong Distrct, 400016 Chongqing, China. [Xin, Gou] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 1 Youyi Road, Yuzhong Distrct, 400016 Chongqing, China. [Xiao, Mingzhao] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 1 Youyi Road, Yuzhong Distrct, 400016 Chongqing, China. [He, Weiyang] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 1 Youyi Road, Yuzhong Distrct, 400016 Chongqing, China. [Kang, Jian] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 1 Youyi Road, Yuzhong Distrct, 400016 Chongqing, China. RP Ren, K (reprint author), Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 400016 Chongqing, China. EM renke_rk@163.com CR White E, 2003, The Pims and outs of survival signaling: role for the Pim-2 protein kinase in the suppression of apoptosis by cytokines. Genes Dev 17(15):1813–1816. , DOI 10.1101/gad. 1123103 Ren K, Gou X, Xiao M,WangM, Liu C, Tang Z, HeW(2013, The over-expression of Pim-2 promote the tumorigenesis of prostatic carcinoma through phosphorylating eIF4B. Prostate 73(13):1462– 1469. , DOI 10.1002/pros.22693 Yang J, Wang J, Chen K, Guo G, Xi R, Rothman PB, Whitten D, Zhang L, Huang S, Chen JL, 2013, eIF4B phosphorylation by pim kinases plays a critical role in cellular transformation by Abl oncogenes. Cancer Res 73(15):4898–4908. , DOI 10.1158/ 0008-5472.CAN-12-4277 Shahbazian D, Parsyan A, Petroulakis E, Topisirovic I, Martineau Y, Gibbs BF, SvitkinY, SonenbergN(2010, Control of cell survival and proliferation by mammalian eukaryotic initiation factor 4B. Mol Cell Biol 30(6):1478–1485. , DOI 10.1128/MCB. 01218-09 Swanson GP, Basler JW, 2010, Prognostic factors for failure after prostatectomy. J Cancer 2:1–19 PMID: 21197260 Mand G-H, Meier P, 2010, IAPs: from caspase inhibitors to modulators of nf-kappab, inflammation and cancer. Nat Rev Cancer 10(8):561–574. , DOI 10.1038/nrc2889 Chen LS, Redkar S, Taverna P, Cortes JE, Gandhi V, 2011, Mechanisms of cytotoxicity to Pim kinase inhibitor, SGI-1776, in acute myeloid leukemia. Blood 118(3):693–702. , DOI 10. 1182/blood-2010-12-323022 Chen LS, Redkar S, Bearss D, Wierda WG, Gandhi V, 2009, Pim kinase inhibitor, SGI-1776, induces apoptosis in chronic lymphocytic leukemia cells. Blood 114(19):4150–4157. , DOI 10. 1182/blood-2009-03-212852 Ayala GE, Dai H, Ittmann M, Li R, Powell M, Frolov A, Wheeler TM, Thompson TC, Rowley D, 2004, Growth and survival mechanisms associated with perineural invasion in prostate cancer. Cancer Res 64(17):6082–6090. , DOI 10.1158/0008- 5472.CAN-04-0838 Kapelko-Slowik K, Urbaniak-Kujda D, Wolowiec D, Jazwiec B, Dybko J, Jakubaszko J, Slowik M, Kuliczkowski K, 2013, Expression of PIM-2 and NF-κB genes is increased in patients with acute myeloid leukemia, AML, and acute lymphoblastic leukemia, ALL, and is associated with complete remission rate and overall survival. Postepy Hig Med Dosw, Online, 67:553–559 PMID: 23752607 Asano J, Nakano A, Oda A, Amou H, Hiasa M, Takeuchi K, Miki H, Nakamura S, Harada T, Fujii S, Kagawa K, Endo I, Yata K, Sakai A, Ozaki S, Matsumoto T, Abe M, 2011, The serine/ threonine kinase Pim-2 is a novel anti-apoptotic mediator in myeloma cells. Leukemia 25(7):1182–1188. , DOI 10.1038/ leu.2011.60 Ren K, Zhang W, Shi Y, Gong J, 2010, Pim-2 activates API-5 to inhibit the apoptosis of hepatocellular carcinoma cells through NFkappaB pathway. Pathol Oncol Res 16(2):229–237. , DOI 10.1007/s12253-009-9215-4 Park YH, Seo SY, Lee E, JH K, Kim HH, Kwak C, 2013, Simvastatin induces apoptosis in castrate resistant prostate cancer cells by deregulating nuclear factor-κB pathway. J Urol 189(4):1547–1552. , DOI 10.1016/j.juro.2012. 10.030 Kim HR, Park CG, Jung JY, 2014, Acacetin, 5,7-dihydroxy-4′- methoxyflavone, exhibits in vitro and in vivo anticancer activity through the suppression of NF-κB/Akt signaling in prostate cancer cells. Int J Mol Med 33(2):317–324. , DOI 10.3892/ijmm. 2013.1571 Zhang P, Chen L, Song Y, Li X, Sun Y, Xiao Y, Xing Y, 2016, Tetraiodothyroacetic acid and transthyretin silencing inhibit pro-metastatic effect of L-thyroxin in anoikis-resistant prostate cancer cells through regulation of MAPK/ERK pathway. Exp Cell Res 347(2):350–359. , DOI 10.1016/j.yexcr.2016. 08.019 Berezovskaya O, Schimmer AD, Glinskii AB, Pinilla C, Hoffman RM, Reed JC, Glinsky GV, 2005, Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells. Cancer Res 65:2378–2386. , DOI 10.1158/0008-5472. CAN-04-2649 Nomura T, Yamasaki M, Nomura Y, Mimata H, 2005, Expression of the inhibitors of apoptosis proteins in cisplatin-resistant prostate cancer cells. Oncol Rep 14:993–997 PMID: 16142363 Tamm I, Kornblau SM, Segall H, Krajewski S,Welsh K, Kitada S, Scudiero DA, Tudor G, Qui YH, Monks A, Andreeff M, Reed JC, 2000, Expressionand prognostic signicance of IAP-family genes in human cancers and myeloid leukemias. Clin Cancer Res 6:1796– 1803 PMID: 10815900 Rodriguez-Berriguete G, Torrealba N, Ortega MA, Martinez- Onsurbe P, Olmedilla G, Paniagua R, Guil-Cid M, Fraile B, Royuela M, 2015, Prognostic value of inhibitors of apoptosis proteins, IAPs, and caspases in prostate cancer: caspase-3 forms and XIAP predict biochemical progression after radical prostatectomy. BMC Cancer 15:809. , DOI 10.1186/s12885-015-1839-z Seligson DB, Hongo F, Huerta-Yepez S,Mizutani Y,Miki T, Yu H, Horvath S, Chia D, Goodglick L, Bonavida B, 2007, Expression of x-linked inhibitor of apoptosis protein is a strong predictor of human prostate cancer recurrence. Clin Cancer Res 13(20):6056– 6063. , DOI 10.1158/1078-0432.CCR-07-0960 Hwang C, Oetjen KA, Kosoff D, Wojno KJ, Albertelli MA, Dunn RL, Robins DM, Cooney KA, Duckett CS, 2008, X-linked inhibitor of apoptosis deficiency in the tramp mouse prostate cancer model. Cell Death Differ 15(5):831–840. , DOI 10.1038/ cdd.2008.15 Ferreira CG, van der Valk P, Span SW, Ludwig I, Smit EF, Kruyt FA, Pinedo HM, van Tinteren H, Giaccone G, 2001, Expression of x-linked inhibitor of apoptosis as a novel prognostic marker in radically resected non-small cell lung cancer patients. Clin Cancer Res 7(8):2468–2474 PMID: 11489828 Aggarwal BB, Ichikawa H, 2005)Molecular targets and anticancer potential of indole-3 carbinol and its derivatives. Cell Cycle 4(9): 1201–1215. , DOI 10.4161/cc.4.9.1993 Kashkar H, 2010, X-linked inhibitor of apoptosis: a chemoresistance factor or a hollow promise. Clin Cancer Res 16(18):4496–4502. , DOI 10.1158/1078-0432.CCR-10- 1664 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 341 EP 348 DI 10.1007/s12253-017-0353-9 PG 8 ER PT J AU Dvorak, K Higgins, A Palting, J Cohen, M Brunhoeber, P AF Dvorak, Katerina Higgins, Amanda Palting, John Cohen, Michael Brunhoeber, Patrick TI Immunohistochemistry with Anti-BRAF V600E (VE1) Mouse Monoclonal Antibody is a Sensitive Method for Detection of the BRAF V600E Mutation in Colon Cancer: Evaluation of 120 Cases with and without KRAS Mutation and Literature Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BRAF V600E; KRAS; Colon cancer; DNA sequencing; Immunohistochemistry ID BRAF V600E; KRAS; Colon cancer; DNA sequencing; Immunohistochemistry AB The major aim of this study was to evaluate the performance of anti-BRAF V600E (VE1) antibody in colorectal tumors with and without KRAS mutation. KRAS and BRAF are two major oncogenic drivers of colorectal cancer (CRC) that have been frequently described as mutually exclusive, thus the BRAF V600E mutation is not expected to be present in the cases with KRAS mutation. In addition, a review of 25 studies comparing immunohistochemistry (IHC) using the anti-BRAF V600E (VE1) antibody with BRAF V600E molecular testing in 4041 patient samples was included. One-hundred and twenty cases with/without KRAS or BRAF mutations were acquired. The tissue were immunostained with anti-BRAF V600E (VE1) antibody with OptiView DAB IHC detection kit. The KRAS mutated cases with equivocal immunostaining were further evaluated by Sanger sequencing for BRAF V600E mutation. Thirty cases with BRAF V600E mutation showed unequivocal, diffuse, uniform, positive cytoplasmic staining and 30 cases with wild-type KRAS and BRAF showed negative staining with anti-BRAF V600E (VE1) antibody. Out of 60 cases with KRAS mutation, 56 cases (93.3%) were negative for BRAF V600E mutation by IHC. Four cases showed weak, equivocal, heterogeneous, cytoplasmic staining along with nuclear staining in 25–90% of tumor cells. These cases were confirmed to be negative for BRAF V600E mutation by Sanger sequencing. Overall, IHC with anti-BRAF V600E (VE1) antibody using recommended protocol with OptiView detection is optimal for detection of BRAF V600E mutation in CRC. Our data are consistent with previous reports indicating that KRAS and BRAF V600E mutation are mutually exclusive. C1 [Dvorak, Katerina] Roche Tissue Diagnostics, 1910 E. Innovation Park DriveTucson, AZ, USA. [Higgins, Amanda] Roche Tissue Diagnostics, 1910 E. Innovation Park DriveTucson, AZ, USA. [Palting, John] Roche Tissue Diagnostics, 1910 E. Innovation Park DriveTucson, AZ, USA. [Cohen, Michael] Roche Tissue Diagnostics, 1910 E. Innovation Park DriveTucson, AZ, USA. [Brunhoeber, Patrick] Roche Tissue Diagnostics, 1910 E. Innovation Park DriveTucson, AZ, USA. RP Dvorak, K (reprint author), Roche Tissue Diagnostics, Tucson, USA. EM katerina.dvorak@roche.com CR Adackapara CA, Sholl LM, Barletta JA et al, 2013, Immunohistochemistry using the BRAF V600E mutation-specific monoclonal antibody VE1 is not a useful surrogate for genotyping in colorectal adenocarcinoma. Histopathology 63:187–193 Affolter K, SamowitzW, Tripp S et al, 2013, BRAF V600E mutation detection by immunohistochemistry in colorectal carcinoma. Genes, chromosomes & cancer 52:748–752 Bledsoe JR, Kamionek M, Mino-Kenudson M, 2014, BRAF V600E immunohistochemistry is reliable in primary and metastatic colorectal carcinoma regardless of treatment status and shows high intratumoral homogeneity. Am J Surg Pathol 38:1418–1428 Boulagnon C, Dudez O, Beaudoux O et al, 2016, BRAFV600E G e n e M u t a t i o n i n C o l o n i c Ad e n o c a r c i n o m a s . Immunohistochemical Detection Using Tissue Microarray and Clinicopathologic Characteristics: An 86 Case Series. Appl Immunohistochem Mol Morphol 24:88–96 Capper D, Preusser M, Habel A et al, 2011, Assessment of BRAF V600E mutation status by immunohistochemistry with a mutationspecific monoclonal antibody. Acta Neuropathol 122:11–19 Capper D, Berghoff AS, Mager l e M e t a l, 2012, Immunohistochemical testing of BRAF V600E status in 1,120 tumor tissue samples of patients with brain metastases. Acta Neuropathol 123:223–233 Capper D, Voigt A, Bozukova G et al, 2013, BRAF V600Especific immunohistochemistry for the exclusion of Lynch syndrome in MSI-H colorectal cancer. International journal of cancer Journal international du cancer 133:1624–1630 Davies H, Bignell GR, Cox C et al, 2002, Mutations of the BRAF gene in human cancer. Nature 417:949–954 Day F, Muranyi A, Singh S et al, 2015, A mutant BRAF V600Especific immunohistochemical assay: correlation with molecular mutation status and clinical outcome in colorectal cancer. Target Oncol 10:99–109 Douillard JY, Oliner KS, Siena S et al, 2013, Panitumumab- FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med 369:1023–1034 Dvorak K, Aggeler B, Palting J et al, 2014, Immunohistochemistry with the anti-BRAF V600E, VE1, antibody: impact of preanalytical conditions and concordance with DNA sequencing in colorectal and papillary thyroid carcinoma. Pathology 46:509–517 Estrella JS, Tetzlaff MT, Bassett RL Jr et al, 2015, Assessment of BRAF V600E Status in Colorectal Carcinoma: Tissue-Specific Discordances between Immunohistochemistry and Sequencing. Mol Cancer Ther 14:2887–2895 Hang JF, Li AF, Chang SC et al, 2016, Immunohistochemical detection of the BRAF V600E mutant protein in colorectal cancers in Taiwan is highly concordant with the molecular test. Histopathology 69:54–62 Hartman DJ, Brand RE, Hu H et al, 2013, Lynch syndromeassociated colorectal carcinoma: frequent involvement of the left colon and rectum and late-onset presentation supports a universal screening approach. Hum Pathol 44:2518–2528 Hernowo BS, Ariyanni F, Suryanti S et al, 2014, Use of BRAF V600E as a molecular marker in aggressive colorectal cancer. Acta Med Indones 46:104–110 Ilie MI, Long-Mira E, Hofman Vet al, 2014, BRAFV600E mutation analysis by immunohistochemistry in patients with thoracic metastases from colorectal cancer. Pathology 46:311–315 Jin M, Hampel H, Zhou X et al, 2013, BRAF V600E mutation analysis simplifies the testing algorithm for Lynch syndrome. Am J Clin Pathol 140:177–183 Koinuma K, Shitoh K,Miyakura Yet al, 2004, Mutations of BRAF are associated with extensive hMLH1 promoter methylation in sporadic colorectal carcinomas. Int J Cancer 108:237–242 1 9 . Kuan SF, Na v i n a S, Cressman KL e t a l, 2014, Immunohistochemical detection of BRAF V600E mutant protein using the VE1 antibody in colorectal carcinoma is highly concordant with molecular testing but requires rigorous antibody optimization. Hum Pathol 45:464–472 20. Lasota J, Kowalik A,Wasag B et al, 2014, Detection of the BRAF V600E mutation in colon carcinoma: critical evaluation of the imunohistochemical approach. Am J Surg Pathol 38:1235–1241 21. Loes IM, Immervoll H, Angelsen JH et al, 2015, Performance comparison of three BRAF V600E detection methods in malignant melanoma and colorectal cancer specimens. Tumour Biol 36:1003– 1013 22. Marin C, Beauchet A, Capper D et al, 2013, Detection of BRAF p.V600E Mutations in Melanoma by Immunohistochemistry Has a Good Interobserver Reproducibility. Arch Pathol Lab Med 138:71– 75 23. Morkel M, Riemer P, Blaker H et al, 2015, Similar but different: distinct roles for KRAS and BRAF oncogenes in colorectal cancer development and therapy resistance. Oncotarget 6:20785–20800 24. Nolan S, Arnason T, Drucker A et al, 2014, The utility of BRAFV600E mutation-specific antibody for colon cancers with microsatellite instability. Appl Immunohistochem Mol Morphol 22:e8–e13 25. Oikonomou E, Koustas E, Goulielmaki M et al, 2014, BRAF vs RAS oncogenes: are mutations of the same pathway equal? Differential signalling and therapeutic implications. Oncotarget 5: 11752–11777 26. Pakneshan S, Salajegheh A, Smith RA et al, 2013, Clinicopathological relevance of BRAF mutations in human cancer. Pathology 45:346–356 27. Parsons MT, Buchanan DD, Thompson B et al, 2012, Correlation of tumour BRAF mutations and MLH1 methylation with germline mismatch repair, MMR, gene mutation status: a literature review assessing utility of tumour features for MMR variant classification. J Med Genet 49:151–157 28. Piton N, Borrini F, Bolognese A et al, 2015, KRAS and BRAF Mutation Detection: Is Immunohistochemistry a Possible Alternative to Molecular Biology in Colorectal Cancer? Gastroenterol Res Pract 2015:753903 29. Qiu T, Lu H, Guo L et al, 2015, Detection of BRAF mutation in Chinese tumor patients using a highly sensitive antibody immunohistochemistry assay. Sci Rep 5:9211 30. Rajagopalan H, Bardelli A, Lengauer C et al, 2002, Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature 418:934 31. Ren J, Li G, Ge J et al, 2012, Is K-ras gene mutation a prognostic factor for colorectal cancer: a systematic review and meta-analysis. Dis Colon rectum 55:913–923 32. Ritterhouse LL, Barletta JA, 2015, BRAF V600E mutationspecific antibody: A review. Semin Diagn Pathol 32:400–408 33. RossleM, SiggM, Ruschoff JH et al, 2013, Ultra-deep sequencing confirms immunohistochemistry as a highly sensitive and specific method for detecting BRAF V600E mutations in colorectal carcinoma. Virchows Arch 463:623–631 34. Roth RM, Hampel H, Arnold CA et al, 2015, A modified Lynch syndrome screening algorithm in colon cancer: BRAF immunohistochemistry is efficacious and cost beneficial. Am J Clin Pathol 143:336–343 35. Routhier CA, Mochel MC, Lynch K et al, 2013, Comparison of 2 monoclonal antibodies for immunohistochemical detection of BRAF V600E mutation in malignant melanoma, pulmonary carcinoma, gastrointestinal carcinoma, thyroid carcinoma, and gliomas. Hum Pathol 44:2563–2570 36. Sajanti S, Sirnio P, Vayrynen JP et al, 2014, VE1 immunohistochemistry accurately detects BRAF V600E mutations in colorectal carcinoma and can be utilized in the detection of poorly differentiated colorectal serrated adenocarcinoma. Virchows Arch 464:637– 643 37. Schafroth C, Galvan JA, Centeno I et al, 2015, VE1 immunohistochemistry predicts BRAF V600E mutation status and clinical outcome in colorectal cancer. Oncotarget 6:41453–41463 38. Seppala TT, Bohm JP, Friman M et al, 2015, Combination of microsatellite instability and BRAF mutation status for subtyping colorectal cancer. Br J Cancer 112:1966–1975 39. Sinicrope FA, Smyrk TC, Tougeron D et al, 2013, Mutationspecific antibody detects mutant BRAF protein expression in human colon carcinomas. Cancer 119(15):2765–2770 40. Taieb J, Le Malicot K, Shi Q et al, 2017, Prognostic value of BRAF and KRAS mutations in MSI andMSS stage III colon cancer. J Natl Cancer Inst 109. , DOI 10.1093/jnci/djw272 41. Thiel A, Heinonen M, Kantonen J et al, 2013, BRAF mutation in sporadic colorectal cancer and Lynch syndrome. Virchows Archiv : an international journal of pathology 463:613–621 42. Thiel A, Heinonen M, Kantonen J et al, 2013, BRAF mutation in sporadic colorectal cancer and Lynch syndrome. Virchows Arch 463:613–621 43. Toon CW, Walsh MD, Chou A et al, 2013, BRAFV600E immunohistochemistry facilitates universal screening of colorectal cancers for Lynch syndrome. Am J Surg Pathol 37:1592–1602 44. Toon CW, Chou A, DeSilva K et al, 2014, BRAFV600E immunohistochemistry in conjunction with mismatch repair status predicts survival in patients with colorectal cancer. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 27:644–650 45. Vakiani E, Yaeger R, Brooke S et al, 2015, Immunohistochemical detection of the BRAF V600E mutant protein in colorectal neoplasms. Appl Immunohistochem Mol Morphol 23:438–443 46. Yuan L, Chi Y, Chen W et al, 2015, Immunohistochemistry and microsatellite instability analysis inmolecular subtyping of colorectal carcinoma based on mismatch repair competency. Int J Clin Exp Med 8:20988–21000 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 349 EP 359 DI 10.1007/s12253-017-0344-x PG 11 ER PT J AU Dura, M Nemejcova, K Jaksa, R Bartu, M Kodet, O Ticha, I Michalkova, R Dundr, P AF Dura, Miroslav Nemejcova, Kristyna Jaksa, Radek Bartu, Michaela Kodet, Ondrej Ticha, Ivana Michalkova, Romana Dundr, Pavel TI Expression of Glut-1 in Malignant Melanoma and Melanocytic Nevi: an Immunohistochemical Study of 400 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glut-1; Malignant melanoma; Melanocytic nevus; Immunohistochemistry; Follow-up ID Glut-1; Malignant melanoma; Melanocytic nevus; Immunohistochemistry; Follow-up AB The glucose transporter-1 (Glut-1) is a cell membrane glycoprotein involved in glucose uptake. An increased expression of Glut-1 is an important cell adaptation mechanism against hypoxia. An upregulation of Glut- 1 can be found in several types of malignant tumors, which are able to reprogram their metabolism from oxidative phosphorylation to aerobic glycolysis (Warburg effect). However, the data regarding melanocytic lesions is equivocal. We performed comprehensive immunohistochemical analysis of the Glut-1 expression in 225 malignant melanomas (MM) and 175 benign nevi. Only the membranous expression of Glut-1 was regarded as positive. The expression of Glut-1 (the cut-off for positivity was determined as H-score 15) was found in 69/225 malignant melanomas. The number of positive cases and the H-score of Glut-1 increased where there was a higher Breslow thickness (p < 0.00001) when comparing pT1- pT4 MM groups. All benign nevi were classified as negative. In conclusion, the membranous expression of Glut-1 is a common feature of a malignant melanoma but this type of expression is very rare in benign melanocytic nevi. Our results suggest that the membranous expression of Glut-1 can be used as a surrogate marker in the assessing of the biological nature of benign and malignant melanocytic lesions. However, despite its high specificity, the sensitivity of this marker is relatively low. Moreover, due to the fact that the increased expression of Glut-1 correlates with a shorter survival period (10-year disease free survival, recurrence free survival and metastasis free survival and MFS), it can be used as a prognostically adverse factor. C1 [Dura, Miroslav] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Nemejcova, Kristyna] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Jaksa, Radek] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Bartu, Michaela] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Kodet, Ondrej] Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Dermatology and VenereologyPrague, Czech Republic. [Ticha, Ivana] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Michalkova, Romana] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Dundr, Pavel] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. RP Dundr, P (reprint author), Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, 12800 Prague, Czech Republic. EM pdundr@seznam.cz CR Thorens B, Mueckler M, 2010, Glucose transporters in the 21st century. Am J Physiol Endocrinol Metab298:E141–E145 Iwasaki K, Yabushita H, Ueno T, Wakatsuki A, 2015, Role of hypoxia-inducible factor-1alpha, carbonic anhydrase-IX, glucose transporter-1 and vascular endothelial growth factor associated with lymph node metastasis and recurrence in patients with locally advanced cervical cancer. Oncol Lett 10:1970–1978 Ma X, Hui Y, Lin L, Wu Y, Zhang X, Liu P, 2015, Clinical significance of COX-2, GLUT-1 and VEGF expressions in endometrial cancer tissues. Pak J Med Sci 31:280–284 Younes M, Lechago LV, Somoano JR, Mosharaf M, Lechago J, 1996, Wide expression of the human erythrocyte glucose transporter Glut1 in human cancers. Cancer Res 56:1164–1167 Nemejcova K, Rosmusova J, Bartu M, Dura M, Ticha I, Dundr P, 2017, Expression of Glut-1 in normal endometrium and endometrial lesions. Int J Surg Pathol 25:389–396 Voldstedlund M, Dabelsteen E, 1997, Expression of GLUT1 in stratified squamous epithelia and oral carcinoma from humans and rats. APMIS 105:537–545 Parente P, Coli A, Massi G, Mangoni A, Fabrizi MM, Bigotti G, 2008, Immunohistochemical expression of the glucose transporters Glut-1 and Glut-3 in human malignant melanomas and benign melanocytic lesions. J Exp Clin Cancer Res 27:34 Carvalho KC, Cunha IW, Rocha RM, Ayala FR, Cajaiba MM, Begnami MD et al, 2011, GLUT1 expression in malignant tumors and its use as an immunodiagnostic marker. Clinics 66:965–972 Wachsberger PR, Gressen EL, Bhala A, Bobyock SB, Storck C, Coss RA, al e, 2002, Variability in glucose transporter-1 levels and hexokinase activity in human melanoma. Melanoma Res 12:35–43 Hirsch FR, Varella-Garcia M, Bunn PA Jr, Di Maria MV, Veve R, Bremmes RMet al, 2003, Epidermal growth factor receptor in nonsmall- cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol 21: 3798–3807 Dawes SM, Tsai S, Gittleman H, Barnholtz-Sloan JS, Bordeaux JS, 2016, Racial disparities in melanoma survival. J Am Acad Dermatol 75:983–991 Kwong LN, Davies MA, 2014, Targeted therapy for melanoma: rational combinatorial approaches. Oncogene 33:1–9 Pavri SN, Clune J, Ariyan S, Narayan D, 2016, Malignant Melanoma: Beyond the Basics. Plast Reconstr Surg 138:330e-340e Gerami P, Li G, Pouryazdanparast P, Blondin B, Beilfuss B, Slenk C et al, 2012, A highly specific and discriminatory FISH assay for distinguishing between benign and malignant melanocytic neoplasms. Am J Surg Pathol 36:808–817 Wang L, Rao M, Fang Y, Hameed M, Viale A, Busam K et al, 2013, A genome-wide high-resolution array-CGH analysis of cutaneous melanoma and comparison of array-CGH to FISH in diagnostic evaluation. J Mol Diagn 15:581–591 Gerami P, Jewell SS, Morrison LE, Blondin B, Schulz J, Ruffalo T et al, 2009, Fluorescence in situ hybridization, FISH, as an ancillary diagnostic tool in the diagnosis ofmelanoma.AmJ Surg Pathol 33:1146–1156 Bedikian AY, Millward M, Pehamberger H, Conry R, Gore M, Trefzer U et al, 2006, Bcl-2 antisense, oblimersen sodium, plus dacarbazine in patients with advanced melanoma: the Oblimersen melanoma study group. J Clin Oncol 24:4738–4745 Dar AA, Majid S, Bezrookove V, Phan B, Ursu S, Nosrati M et al, 2016, BPTF transduces MITF-driven prosurvival signals in melanoma cells. Proc Natl Acad Sci U S A 113:6254–6258 Boni R, Doguoglu A, Burg G,Muller B, Dummer R, 1996, MIB-1 immunoreactivity correlates with metastatic dissemination in primary thick cutaneous melanoma. J AmAcad Dermatol 35:416–418 Gimotty PA, Van Belle P, Elder DE, Murry T, Montone KT, Xu X et al, 2005, Biologic and prognostic significance of dermal Ki67 expression, mitoses, and tumorigenicity in thin invasive cutaneous melanoma. J Clin Oncol 23:8048–8056 Ladstein RG, Bachmann IM, Straume O, Akslen LA, 2010, Ki-67 expression is superior to mitotic count and novel proliferation markers PHH3, MCM4 and mitosin as a prognostic factor in thick cutaneous melanoma. BMC Cancer 10:140 Mihic-Probst D, Ikenberg K, Tinguely M, Schraml P, Behnke S, Seifert B et al, 2012, Tumor cell plasticity and angiogenesis in human melanomas. PLoS One 7:e33571 Slominski A, Kim TK, Brozyna AA, Janjetovic Z, Brooks DL, Schwab LP et al, 2014, The role of melanogenesis in regulation of melanoma behavior: melanogenesis leads to stimulation of HIF- 1alpha expression and HIF-dependent attendant pathways. Arch Biochem Biophys 563:79–93 Ilmonen S, HernbergM, Pyrhonen S, Tarkkanen J,Asko-Seljavaara S, 2005, Ki-67, Bcl-2 and p53 expression in primary andmetastatic melanoma. Melanoma Res 15:375–381 Matin RN, Chikh A, Chong SL, Mesher D, Graf M, Sanza P, et al., 2013, p63 is an alternative p53 repressor in melanoma that confers chemoresistance and a poor prognosis. J Exp Med 210:581–603 Rieger E, Hofmann-Wellenhof R, Soyer HP, Kofler R, Cerroni L, Smolle J et al, 1993, Comparison of proliferative activity as assessed by proliferating cell nuclear antigen, PCNA, and Ki-67 monoclonal antibodies in melanocytic skin lesions. A quantitative immunohistochemical study. J Cutan Pathol 20:229–236 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Klement RJ, Kammerer U, 2011, Is there a role for carbohydrate restriction in the treatment and prevention of cancer? Nutr Metab, Lond, 8:75 Annibaldi A, Widmann C, 2010, Glucose metabolism in cancer cells. Curr Opin Clin Nutr Metab Care 13:466–470 Pelicano H, RH X, Du M, Feng L, Sasaki R, Carew JS et al, 2006, Mitochondrial respiration defects in cancer cells cause activation of Akt survival pathway through a redox-mediated mechanism. J Cell Biol 175:913–923 Robey RB, Hay N, 2009, Is Akt the "Warburg kinase"?-Aktenergy metabolism interactions and oncogenesis. Semin Cancer Biol 19:25–31 Dronca RS, Allred JB, Perez DG, Nevala WK, Lieser EA, Thompson M et al, 2014, Phase II study of temozolomide, TMZ, and everolimus, RAD001, therapy for metastatic melanoma: a north central cancer treatment group study, N0675. Am J Clin Oncol 37:369–376 Zhang D, Li J, Wang F, Hu J, Wang S, Sun Y, 2014, 2-Deoxy-Dglucose targeting of glucose metabolism in cancer cells as a potential therapy. Cancer Lett 355:176–183 Liu Y, Cao Y, Zhang W, Bergmeier S, Qian Y, Akbar H et al, 2012, A small-molecule inhibitor of glucose transporter 1 downregulates glycolysis, induces cell-cycle arrest, and inhibits cancer cell growth in vitro and in vivo. Mol Cancer Ther 11:1672–1682 Brown RS Wahl RL. Overexpression of Glut-1 glucose transporter in human breast cancer. An immunohistochemical study. Cancer 72:2979–2985 Haber RS, Rathan A, Weiser KR, Pritsker A, Itzkowitz SH, Bodian C et al, 1998, GLUT1 glucose transporter expression in colorectal carcinoma: a marker for poor prognosis. Cancer 83:34–40 Nagase Y, Takata K, Moriyama N, Aso Y, Murakami T, Hirano H, 1995, Immunohistochemical localization of glucose transporters in human renal cell carcinoma. J Urol 153:798–801 Ogawa J, Inoue H, Koide S, 1997, Glucose-transporter-type-I-gene amplification correlates with sialyl-Lewis-X synthesis and proliferation in lung cancer. Int J Cancer 74:189–192 Yamamoto T, Seino Y, Fukumoto H, Koh G, Yano H, Inagaki N et al, 1990, Over-expression of facilitative glucose transporter genes in human cancer. Biochem Biophys Res Commun 170:223–230 Baer SC, Casaubon L, Younes M, 1997, Expression of the human erythrocyte glucose transporter Glut1 in cutaneous neoplasia. J Am Acad Dermatol 37:575–577 Park SG, Lee JH, Lee WA, Han KM, 2012, Biologic correlation between glucose transporters, hexokinase-II, Ki-67 and FDG uptake in malignant melanoma. Nucl Med Biol 39: 1167–1172 Yamada K, Brink I, Bisse E, Epting T, Engelhardt R, 2005, Factors influencing [F-18] 2-fluoro-2-deoxy-D-glucose, F-18 FDG, uptake in melanoma cells: the role of proliferation rate, viability, glucose transporter expression and hexokinase activity. J Dermatol 32:316–334 Lee JH, Gulec SA, Kyshtoobayeva A, Sim MS, Morton DL, 2009, Biological factors, tumor growth kinetics, and survival after metastasectomy for pulmonary melanoma. Ann Surg Oncol 16:2834–2839 Eichhoff OM, Zipser MC, Xu M, Weeraratna AT, Mihic D, Dummer R et al, 2010, The immunohistochemistry of invasive and proliferative phenotype switching in melanoma: a case report. Melanoma Res 20:349–355 Lu H, Forbes RA, Verma A, 2002, Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg effect in carcinogenesis. J Biol Chem 277:23111–23115 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 361 EP 368 DI 10.1007/s12253-017-0363-7 PG 8 ER PT J AU Gao, LM Wang, F Zheng, Y Fu, ZZ Zheng, L Chen, LL AF Gao, Li-Ming Wang, Fang Zheng, Yue Fu, Zhan-Zhao Zheng, Lei Chen, Lan-Lan TI Roles of Fibroblast Activation Protein and Hepatocyte Growth Factor Expressions in Angiogenesis and Metastasis of Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Fibroblast activation protein; Hepatocyte growth factor; Angiogenesis; Metastasis; Microvessel density ID Gastric cancer; Fibroblast activation protein; Hepatocyte growth factor; Angiogenesis; Metastasis; Microvessel density AB This study aims to explore the roles of fibroblast activation protein (FAP) and hepatocyte growth factor (HGF) expressions in the angiogenesis and metastasis of gastric cancer (GC). From May 2012 to December 2015, 110 GC patients who received surgical treatment in the First Hospital of Qinhuangdao were selected. The HGF and FAP expressions in 110 cases of GC, 130 cases of normal gastric mucosa and 115 cases of gastric ulcer were detected by streptavidinperosidase (SP) method. Venous blood HGF level of GC patients was tested by enzyme-linked immunosorbent assay (ELISA). The micro-vessel number of the patients in the three groups were calculated and analyzed. In GC group, positive expression rates of FAP and HGF protein were 61.8% and 67.3% respectively, which were both higher than those in normal gastric mucosa and gastric ulcer groups. The microvessel numbers in patients of the normal gastric mucosa and gastric ulcer groups are far less than that in GC group. FAP, HGF and micro-vessel density (MVD) were significantly correlated with infiltration depth, tumor-node-metastasis (TNM) staging, lymph node metastasis (LNM) and distant metastasis. The results of ELISA showed that serum HGF level was related to tumor size, infiltration degree, TNM staging, LNM and distant metastasis. FAP and HGF expressions in GC were positively correlated with MVD, and the expressions of FAP and HGF in GC were in positive correlation. Our study provided evidence that high FAP and HGF expressions may be positively correlated with the angiogenesis and metastasis of GC. C1 [Gao, Li-Ming] First Hospital of Qinhuangdao, Department of Oncology, No. 258 Wenhua Road, Haigang District, 066000 Qinhuangdao, Hebei Province, China. [Wang, Fang] First Hospital of Qinhuangdao, Department of Oncology, No. 258 Wenhua Road, Haigang District, 066000 Qinhuangdao, Hebei Province, China. [Zheng, Yue] First Hospital of Qinhuangdao, Department of Gastroenterology, 066000 Qinhuangdao, Hebei Province, China. [Fu, Zhan-Zhao] First Hospital of Qinhuangdao, Department of Oncology, No. 258 Wenhua Road, Haigang District, 066000 Qinhuangdao, Hebei Province, China. [Zheng, Lei] First Hospital of Qinhuangdao, Department of Oncology, No. 258 Wenhua Road, Haigang District, 066000 Qinhuangdao, Hebei Province, China. [Chen, Lan-Lan] First Hospital of Qinhuangdao, Department of Oncology, No. 258 Wenhua Road, Haigang District, 066000 Qinhuangdao, Hebei Province, China. RP Fu, ZZ (reprint author), First Hospital of Qinhuangdao, Department of Oncology, 066000 Qinhuangdao, China. EM Tshgaoliming@163.com CR Shirahata A, Sakata M, Kitamura Y, Sakuraba K, Yokomizo K, Goto T, Mizukami H, Saito M, Ishibashi K, Kigawa G, Nemoto H, Hibi K, 2010, MACC 1 as a marker for peritoneal-disseminated gastric carcinoma. Anticancer Res 30:3441–3444 Jang BG, KimWH, 2011, Molecular pathology of gastric carcinoma. Pathobiology 78:302–310 Izuishi K, Mori H, 2016, Recent strategies for treating stage iv gastric cancer: roles of palliative gastrectomy, chemotherapy, and radiotherapy. J Gastrointestin Liver Dis 25:87–94 Songun I, Putter H, Kranenbarg EM, Sasako M, van de Velde CJ, 2010, Surgical treatment of gastric cancer: 15-year follow-up results of the randomised nationwide Dutch D1D2 trial. Lancet Oncol 11:439–449 Marrelli D, De Stefano A, de Manzoni G, Morgagni P, Di Leo A, Roviello F, 2005, Prediction of recurrence after radical surgery for gastric cancer: a scoring system obtained from a prospective multicenter study. Ann Surg 241:247–255 Marrelli D, Pedrazzani C, Neri A, Corso G, DeStefano A, Pinto E, Roviello F, 2007, Complications after extended, D2, and superextended, D3, lymphadenectomy for gastric cancer: analysis of potential risk factors. Ann Surg Oncol 14:25–33 Wang RF, Zhang LH, Shan LH, Sun WG, Chai CC, Wu HM, Ibla JC, Wang LF, Liu JR, 2013, Effects of the fibroblast activation protein on the invasion and migration of gastric cancer. Exp Mol Pathol 95:350–356 Lordick F, 2014, Targeting the HGF/MET pathway in gastric cancer. Lancet Oncol 15:914–916 Graziano F, Galluccio N, Lorenzini P, Ruzzo A, Canestrari E, D'Emidio S, Catalano V, Sisti V, Ligorio C, Andreoni F, Rulli E, Di Oto E, Fiorentini G, Zingaretti C, De Nictolis M, Cappuzzo F, Magnani M, 2011, Genetic activation of the MET pathway and prognosis of patients with high-risk, radically resected gastric cancer. J Clin Oncol 29:4789–4795 Abbas O, Richards JE, MahalingamM(2010, Fibroblast-activation protein: a single marker that confidently differentiates morpheaform/infiltrative basal cell carcinoma from desmoplastic trichoepithelioma. Mod Pathol 23:1535–1543 El Khoury J, Kurban M, Kibbi AG, Abbas O, 2014, Fibroblastactivation protein: valuable marker of cutaneous epithelial malignancy. Arch Dermatol Res 306:359–365 Tchou J, Zhang PJ, BiY, Satija C,Marjumdar R, Stephen TL, Lo A, Chen H, Mies C, June CH, Conejo-Garcia J, Pure E, 2013, Fibroblast activation protein expression by stromal cells and tumor-associated macrophages in human breast cancer. Hum Pathol 44:2549–2557 Wen Y,Wang CT, Ma TT, Li ZY, Zhou LN, Mu B, Leng F, Shi HS, Li YO,Wei YQ, 2010, Immunotherapy targeting fibroblast activation protein inhibits tumor growth and increases survival in a murine colon cancer model. Cancer Sci 101:2325–2332 Yanovitch T, Li YJ, Metlapally R, Abbott D, Viet KN, Young TL, 2009, Hepatocyte growth factor and myopia: genetic association analyses in a Caucasian population. Mol Vis 15:1028–1035 Cai L, Johnstone BH, Cook TG, Liang Z, Traktuev D, Cornetta K, Ingram DA, Rosen ED, March KL, 2007, Suppression of hepatocyte growth factor production impairs the ability of adipose-derived stem cells to promote ischemic tissue revascularization. Stem Cells 25:3234–3243 Nakagami H, Maeda K, Morishita R, Iguchi S, Nishikawa T, Takami Y, Kikuchi Y, Saito Y, Tamai K, Ogihara T, Kaneda Y, 2005, Novel autologous cell therapy in ischemic limb disease through growth factor secretion by cultured adipose tissue-derived stromal cells. Arterioscler Thromb Vasc Biol 25:2542–2547 Zhao L, Yasumoto K, Kawashima A, Nakagawa T, Takeuchi S, Yamada T, Matsumoto K, Yonekura K, Yoshie O, Yano S, 2013, Paracrine activation of MET promotes peritoneal carcinomatosis in scirrhous gastric cancer. Cancer Sci 104:1640–1646 Wang W, Xu DZ, Li YF, Guan YX, Sun XW, Chen YB, Kesari R, Huang CY, Li W, Zhan YQ, Zhou ZW, 2011, Tumor-ratiometastasis staging system as an alternative to the 7th edition UICC TNM system in gastric cancer after D2 resection–results of a single-institution study of 1343 Chinese patients. Ann Oncol 22: 2049–2056 Wroblewski LE, Peek RM Jr, Wilson KT, 2010, Helicobacter pylori and gastric cancer: factors that modulate disease risk. Clin Microbiol Rev 23:713–739 Lo PC, Chen J, Stefflova K, Warren MS, Navab R, Bandarchi B, Mullins S, Tsao M, Cheng JD, Zheng G, 2009, Photodynamic molecular beacon triggered by fibroblast activation protein on cancer-associated fibroblasts for diagnosis and treatment of epithelial cancers. J Med Chem 52:358–368 Inokuchi M, Otsuki S, FujimoriY, Sato Y, NakagawaM,KojimaK, 2015, Clinical significance of MET in gastric cancer. World J Gastrointest Oncol 7:317–327 Erbersdobler A, Isbarn H, Dix K, Steiner I, Schlomm T, Mirlacher M, Sauter G, Haese A, 2010, Prognostic value of microvessel density in prostate cancer: a tissue microarray study. World J Urol 28: 687–692 Du JR, Jiang Y, Zhang YM, Fu H, 2003, Vascular endothelial growth factor and microvascular density in esophageal and gastric carcinomas. World J Gastroenterol 9:1604–1606 Liu F, Qi L, Liu B, Liu J, ZhangH, Che D, Cao J, Shen J, Geng J, Bi Y, Ye L, Pan B, Yu Y, 2015, Fibroblast activation protein overexpression and clinical implications in solid tumors: a meta-analysis. PLoS One 10:e0116683 Cai F, Li Z, Wang C, Xian S, Xu G, Peng F, Wei Y, Lu Y, 2013, Short hairpin RNA targeting of fibroblast activation protein inhibits tumor growth and improves the tumor microenvironment in a mouse model. BMB Rep 46:252–257 Guo T, Yang J, Yao J, Zhang Y, Da M, Duan Y, 2013, Expression of MACC1 and c-Met in human gastric cancer and its clinical significance. Cancer Cell Int 13:121 Noguchi E, Saito N, Kobayashi M, Kameoka S, 2015, Clinical significance of hepatocyte growth factor/c-Met expression in the assessment of gastric cancer progression. Mol Med Rep 11:3423– 3431 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 369 EP 376 DI 10.1007/s12253-017-0359-3 PG 8 ER PT J AU Song, XF Wang, QH Huo, R AF Song, Xiao-Fei Wang, Qi-Hua Huo, Ran TI Effects of microRNA-708 on Epithelial-Mesenchymal Transition, Cell Proliferation and Apoptosis in Melanoma Cells by Targeting LEF1 through the Wnt Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MicroRNA-708; Epithelial-mesenchymal transition; Melanoma cells; Lymphoid enhancer-binding factor-1; Wnt signaling pathway ID MicroRNA-708; Epithelial-mesenchymal transition; Melanoma cells; Lymphoid enhancer-binding factor-1; Wnt signaling pathway AB This study was conducted in order to elucidate the role microRNA-708 (miR-708) plays between proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT) involving melanoma cells by targeting using LEF1 through the Wnt signaling pathway. Male Kunming mice were selected and subsequently divided into normal and model groups to take part in this study. Following cell line selection, the B16 cells with the highest miR-708 expression were selected and assigned into the control, blank, negative control (NC), miR-708 mimic, miR-708 inhibitor, siRNA-LEF1, and miR-708 inhibitor + siRNA-LEF1 groups. A Bioinformatics Web service and dual-luciferase reporter assay were conducted in order to determine the relationship between LEF1 and miR-708. The RT-qPCR method was performed in order to detect the miR-708 expression and mRNA expressions of LEF1, β-catenin, Wnt3a, N-cadherin, Bcl-2, Bax, Caspase3, Ecadherin, and western blotting was used in order to detect the protein expressions of these genes. MTT assay, scratch test, Transwell assay, and flow cytometry were all conducted in order to detect the cell proliferation, migration, invasion, and cycle/apoptosis, respectively. LEF1 was verified as the target gene of miR-708. In comparison with the normal group, the model group had reduced expressions of miR-708, Bax, Caspase3, and E-cadherin, while showing elevated expressions of LEF1, β-catenin, Bcl-2, Wnt3a, and N-cadherin. In comparison to the blank and control groups, the miR-708, mimic, and siRNA-LEF1 groups had elevated expressions of Bax, Caspase3, and Ecadherin, while also showing enhanced cell apoptosis. The miR-708, mimic, and siRNA-LEF1 groups also had decreased expressions of LEF1, β-catenin, Bcl-2, Wnt3a, and N-cadherin, and reduced optical density value 48 h and 72 h after transfection. Besides, these two groups showed declined cell migration and invasion, as well as lengthened G0/G1 phase (increased cell number) and shortened S phase (decreased cell number). Our findings demonstrated that an overexpressed miR-708 inhibits the proliferation, invasion, migration, and EMT, but also promotes the apoptosis of melanoma cells by targeting LEF1 through the suppression of the Wnt signaling pathway. C1 [Song, Xiao-Fei] Shandong Provincial Hospital Affiliated to Shandong University, Department of Aesthetic Plastic and Burn Surgery, No. 324, Jingwuweiqi Road, 250021 Jinan, China. [Wang, Qi-Hua] Linyi Dermatology Hospital, Department of Dermatology, 276003 Linyi, China. [Huo, Ran] Shandong Provincial Hospital Affiliated to Shandong University, Department of Aesthetic Plastic and Burn Surgery, No. 324, Jingwuweiqi Road, 250021 Jinan, China. RP Huo, R (reprint author), Shandong Provincial Hospital Affiliated to Shandong University, Department of Aesthetic Plastic and Burn Surgery, 250021 Jinan, China. EM huoran8003@163.com CR Busser B, Lupo J, Sancey L, Mouret S, Faure P, Plumas J, Chaperot L, Leccia MT, Coll JL, Hurbin A, Hainaut P, Charles J, 2017, Plasma circulating tumor DNA levels for the monitoring of melanoma patients: landscape of available technologies and clinical applications. Biomed Res Int 2017:5986129 Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, Carter SL, Stewart C, Mermel CH, Roberts SA, Kiezun A, Hammerman PS, McKenna A, Drier Y, Zou L, Ramos AH, Pugh TJ, Stransky N, Helman E, Kim J, Sougnez C, Ambrogio L, Nickerson E, Shefler E, Cortes ML, Auclair D, Saksena G,Voet D, Noble M, DiCara D, Lin P, Lichtenstein L, Heiman DI, Fennell T, Imielinski M, Hernandez B, Hodis E, Baca S, Dulak AM, Lohr J, Landau DA, CJ W, Melendez-Zajgla J, Hidalgo-Miranda A, Koren A, McCarroll SA, Mora J, Lee RS, Crompton B, Onofrio R, Parkin M, Winckler W, Ardlie K, Gabriel SB, Roberts CW, Biegel JA, Stegmaier K, Bass AJ, Garraway LA, Meyerson M, Golub TR, Gordenin DA, Sunyaev S, Lander ES, Getz G, 2013, Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499:214–218 Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67:7–30 Miller AJ, Mihm MC Jr, 2006, Melanoma. N Engl J Med 355:51– 65 Wu S, Singh RK, 2011, Resistance to chemotherapy and molecularly targeted therapies: rationale for combination therapy in malignant melanoma. Curr Mol Med 11:553–563 Melis C, Rogiers A, Bechter O, van den Oord JJ, 2017, Molecular genetic and immunotherapeutic targets in metastatic melanoma. Virchows Arch. , DOI 10.1007/s00428-017-2113-3 Ndoye AWeeraratna AT, 2016, Autophagy- an emerging target for melanoma therapy. F1000research 5:1888 Lelli D, Pedone C, Sahebkar A, 2017, Curcumin and treatment of melanoma: the potential role ofmicroRNAs. Biomed Pharmacother 88:832–834 Sun V, Zhou WB, Majid S, Kashani-Sabet M, Dar AA, 2014, MicroRNA-mediated regulation of melanoma. Br J Dermatol 171:234–241 Luo C,Weber CE, OsenW, Bosserhoff AK, Eichmuller SB, 2014, The role of microRNAs in melanoma. Eur J Cell Biol 93:11–22 Leibowitz-Amit R, Sidi Y, Avni D, 2012, Aberrations in the micro- RNA biogenesis machinery and the emerging roles of micro-RNAs in the pathogenesis of cutaneous malignant melanoma. Pigment Cell Melanoma Res 25:740–757 Li G, Yang F, Xu H, Yue Z, Fang X, Liu J, 2015)MicroRNA-708 is downregulated in hepatocellular carcinoma and suppresses tumor invasion and migration. Biomed Pharmacother 73:154–159 Saini S, Yamamura S, Majid S, Shahryari V, Hirata H, Tanaka Y, Dahiya R, 2011)MicroRNA-708 induces apoptosis and suppresses tumorigenicity in renal cancer cells. Cancer Res 71:6208–6219 Lin KT, YehYM, ChuangCM, Yang SY, Chang JW, Sun SP,Wang YS, Chao KC,Wang LH, 2015, Glucocorticoids mediate induction of microRNA-708 to suppress ovarian cancer metastasis through targeting Rap1B. Nat Commun 6:5917 Murakami T, Saitoh I, Sato M, Inada E, SodaM, Oda M, Domon H, Iwase Y, Sawami T,Matsueda K, Terao Y, Ohshima H, Noguchi H, Hayasaki H, 2017, Isolation and characterization of lymphoid enhancer factor-1-positive deciduous dental pulp stem-like cells after transfection with a piggyBac vector containing LEF1 promoterdriven selection markers. Arch Oral Biol 81:110–120 Long GV, Fung C,Menzies AM, Pupo GM, Carlino MS, Hyman J, Shahheydari H, Tembe V, Thompson JF, Saw RP, Howle J, Hayward NK, Johansson P, Scolyer RA, Kefford RF, Rizos H, 2014, Increased MAPK reactivation in early resistance to dabrafenib/trametinib combination therapy of BRAF-mutant metastatic melanoma. Nat Commun 5:5694 Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga- Kalabis M, Cipolla AK, Wubbenhorst B, Xu X, Gimotty PA, Kee D, Santiago-Walker AE, Letrero R, D'Andrea K, Pushparajan A, Hayden JE, Brown KD, Laquerre S, McArthur GA, Sosman JA, Nathanson KL, Herlyn M, 2010, Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell 18:683–695 O'Connell MP, Marchbank K, Webster MR, Valiga AA, Kaur A, Vultur A, Li L, Herlyn M, Villanueva J, Liu Q, Yin X, Widura S, Nelson J, Ruiz N, Camilli TC, Indig FE, Flaherty KT, Wargo JA, Frederick DT, Cooper ZA, Nair S, Amaravadi RK, Schuchter LM, Karakousis GC, Xu W, Xu X, Weeraratna AT, 2013, Hypoxia induces phenotypic plasticity and therapy resistance in melanoma via the tyrosine kinase receptors ROR1 and ROR2. Cancer Discov 3:1378–1393 Ayuk SM, Abrahamse H, Houreld NN, 2016, The role of photobiomodulation on gene expression of cell adhesionmolecules in diabetic wounded fibroblasts in vitro. J Photochem Photobiol B 161:368–374 Jang JS, Jeon HS, Sun Z, Aubry MC, Tang H, Park CH, Rakhshan F, SchultzDA, Kolbert CP, Lupu R, Park JY, Harris CC,Yang P, Jen J, 2012, Increased miR-708 expression in NSCLC and its association with poor survival in lung adenocarcinoma from never smokers. Clin Cancer Res 18:3658–3667 Ryu S, McDonnell K, Choi H, Gao D, HahnM, Joshi N, Park SM, Catena R, Do Y, Brazin J, Vahdat LT, Silver RB, Mittal V, 2013, Suppression of miRNA-708 by polycomb group promotes metastases by calcium-induced cell migration. Cancer Cell 23:63–76 Perez EC, Machado J Jr, Aliperti F, Freymuller E, Mariano M, Lopes JD, 2008, B-1 lymphocytes increase metastatic behavior of melanoma cells through the extracellular signal-regulated kinase pathway. Cancer Sci 99:920–928 Zhuang L, Lee CS, Scolyer RA,McCarthy SW, Palmer AA, Zhang XD, Thompson JF, Bron LP, Hersey P, 2005, Activation of the extracellular signal regulated kinase, ERK, pathway in human melanoma. J Clin Pathol 58:1163–1169 Hess AR, Postovit LM, Margaryan NV, Seftor EA, Schneider GB, Seftor RE, Nickoloff BJ, Hendrix MJ, 2005, Focal adhesion kinase promotes the aggressive melanoma phenotype. Cancer Res 65: 9851–9860 Guo P, Lan J, Ge J, Nie Q, Mao Q, Qiu Y, 2013, miR-708 acts as a tumor suppressor in human glioblastoma cells. Oncol Rep 30:870–876 Cho JH, Robinson JP, Arave RA, Burnett WJ, Kircher DA, Chen G, Davies MA, Grossmann AH, VanBrocklin MW, McMahon M, Holmen SL, 2015)AKT1 activation promotes development ofmelanoma metastases. Cell Rep 13:898–905 Vizkeleti L, Ecsedi S, Rakosy Z, Orosz A, Lazar V, Emri G, Koroknai V, Kiss T, Adany R, Balazs M, 2012, The role of CCND1 alterations during the progression of cutaneous malignant melanoma. Tumour Biol 33:2189–2199 De Donatis GM, Pape EL, Pierron A, Cheli Y, Hofman V, Hofman P, Allegra M, Zahaf K, Bahadoran P, Rocchi S, Bertolotto C, Ballotti R, Passeron T, 2016, NF-kB2 induces senescence bypass in melanoma via a direct transcriptional activation of EZH2. Oncogene 35:2735–2745 Redondo P, Lloret P, Idoate M, Inoges S, 2005, Expression and serum levels ofMMP-2 andMMP-9 during human melanoma progression. Clin Exp Dermatol 30:541–545 Wu L, Zhao JC, Kim J, Jin HJ, Wang CY, Yu J, 2013, ERG is a critical regulator of Wnt/LEF1 signaling in prostate cancer. Cancer Res 73:6068–6079 Liang J, Li Y, Daniels G, Sfanos K, De Marzo A,Wei J, Li X, ChenW, Wang J, Zhong X, Melamed J, Zhao J, Lee P, 2015, LEF1 targeting EMT in prostate cancer invasion is regulated by miR-34a. Mol Cancer Res 13:681–688 Cui XP, Xing Y, Chen JM, Dong SW, Ying DJ, Yew DT, 2011, Wnt/beta-catenin is involved in the proliferation of hippocampal neural stem cells induced by hypoxia. Ir J Med Sci 180:387–393 Castiglia D, Bernardini S, Alvino E, Pagani E, De Luca N, Falcinelli S, Pacchiarotti A, Bonmassar E, Zambruno G, D'Atri S, 2008, Concomitant activation of Wnt pathway and loss of mismatch repair function in human melanoma. Genes Chromosom Cancer 47:614–624 Sun L, Liu T, Zhang S, Guo K, Liu Y, 2017, Oct4 induces EMT through LEF1/beta-catenin dependent WNT signaling pathway in hepatocellular carcinoma. Oncol Lett 13:2599–2606 Spangler B, Vardimon L, Bosserhoff AK, Kuphal S, 2011, Posttranscriptional regulation controlled by E-cadherin is important for c-Jun activity in melanoma. Pigment Cell Melanoma Res 24:148– 164 Qi J, Chen N,Wang J, Siu CH, 2005, Transendothelial migration of melanoma cells involves N-cadherin-mediated adhesion and activation of the beta-catenin signaling pathway. Mol Biol Cell 16: 4386–4397 Chen H, Gao X, Sun Z,Wang Q, Zuo D, Pan L, Li K, Chen J, Chen G, Hu K, Li K, Shah AS, Huang T, Zeeshan BhattiM, Tong L, Jiao C, Liu J, Chen T, Yao L, Dang Y, Liu T, Li L, 2017, REGgamma accelerates melanoma formation by regulating Wnt/beta-catenin signaling pathway. Exp Dermatol 26:1118–1124 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 377 EP 389 DI 10.1007/s12253-017-0334-z PG 13 ER PT J AU Kakkassery, V Skosyrski, S Luth, A Kleuser, B van der Giet, vdM Tate, R Reinhard, J Faissner, A Joachim, S Kociok, N AF Kakkassery, Vinodh Skosyrski, Sergej Luth, Anja Kleuser, Burkhard van der Giet, van der Markus Tate, Rothwelle Reinhard, Jacqueline Faissner, Andreas Joachim, C. Stephanie Kociok, Nobert TI Etoposide Upregulates Survival Favoring Sphingosine-1-Phosphate in Etoposide-Resistant Retinoblastoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Retinoblastoma; Sphingosine-1-phosphate; Chemotherapy resistance ID Retinoblastoma; Sphingosine-1-phosphate; Chemotherapy resistance AB Improved knowledge of retinoblastoma chemotherapy resistance is needed to raise treatment efficiency. The objective of this study was to test whether etoposide alters glucosyl-ceramide, ceramide, sphingosine, and sphingosine-1-phosphate (sphingosine-1-P) levels in parental retinoblastoma cells (WERI Rb1) or their etoposide-resistant subclones (WERI EtoR). WERI Rb1 and WERI EtoR were incubated with 400 ng/ml etoposide for 24 h. Levels of glucosyl-ceramides, ceramides, sphingosine, sphingosine-1-P were detected by Q-TOF mass spectrometry. Statistical analysis was done by ANOVA followed by Tukey post-hoc test (p < 0.05). The mRNA expression of sphingolipid pathways enzymes in WERI Rb1, WERI EtoR and four human retinoblastoma tissue samples was analyzed by quantitative real-time PCR. Pathways enzymes mRNA expression confirmed similarities of human sphingolipid metabolism in both cell lines and tissue samples, but different relative expression. Significant up-regulation of sphingosine was seen in both cell lines (p < 0.001). Only sphingosine-1-P up-regulation was significantly increased in WERI EtoR (p < 0.01), but not in WERI Rb1 (p > 0.2). Both cell lines upregulate proapoptotic sphingosine after etoposide incubation, but only WERI EtoR produces additional survival favorable sphingosine- 1-P. These data may suggest a role of sphingosine- 1-P in retinoblastoma chemotherapy resistance, although this seems not to be the only resistance mechanism. C1 [Kakkassery, Vinodh] Charite Universitatsmedizin, Department of OphthalmologyBerlin, Germany. [Skosyrski, Sergej] Charite Universitatsmedizin, Department of OphthalmologyBerlin, Germany. [Luth, Anja] University Potsdam, Department of Nutrition SciencePotsdam, Germany. [Kleuser, Burkhard] University Potsdam, Department of Nutrition SciencePotsdam, Germany. [van der Giet, van der Markus] Charite Universitatsmedizin, Campus Benjamin Franklin, Department of NephrologyBerlin, Germany. [Tate, Rothwelle] University of Strathclyde, Strathclyde Institute of Pharmacy and Biomedical SciencesGlasgow, UK. [Reinhard, Jacqueline] Ruhr-University Bochum, Faculty of Biology and Biotechnology, Department of Cell Morphology and Molecular NeurobiologyBochum, Germany. [Faissner, Andreas] Ruhr-University Bochum, Faculty of Biology and Biotechnology, Department of Cell Morphology and Molecular NeurobiologyBochum, Germany. [Joachim, C. Stephanie] Ruhr-University Bochum, Department of OphthalmologyBochum, Germany. [Kociok, Nobert] Charite Universitatsmedizin, Department of OphthalmologyBerlin, Germany. RP Kakkassery, V (reprint author), Charite Universitatsmedizin, Department of Ophthalmology, Berlin, Germany. EM vinodh.kakkassery@gmail.com CR Sanders BM, Draper GJ, Kingston JE, 1988, Retinoblastoma in Great Britain 1969-80: incidence, treatment, and survival. Br J Ophthalmol 72(8):576–583 Abramson DH, Schefler AC, 2004, Update on retinoblastoma. Retina 24(6):828–848 Schuler AO, Bornfeld N, 2000, Current therapy aspects of intraocular tumors. Ophthalmologe 97(3):207–222 Shields CL, De Potter P, Himelstein BP, Shields JA, Meadows AT, Maris JM, 1996, Chemoreduction in the initial management of intraocular retinoblastoma. Arch Ophthalmol 114(11):1330–1338 Friedman DL, Himelstein B, Shields CL, Shields JA, Needle M, Miller D et al, 2000, Chemoreduction and local ophthalmic therapy for intraocular retinoblastoma. J Clin Oncol 18(1):12–17 Toma NM, Hungerford JL, Plowman PN, Kingston JE, Doughty D, 1995, External beam radiotherapy for retinoblastoma: II. Lens sparing technique. Br J Ophthalmol 79(2):112–117 Hungerford JL, Toma NM, Plowman PN, Doughty D, Kingston JE, 1997, Whole-eye versus lens-sparing megavoltage therapy for retinoblastoma. Front Radiat Ther Oncol 30:81–87 Abramson DH, Marr BP, Dunkel IJ, Brodie S, Zabor EC, Driscoll SJ et al, 2012, Intra-arterial chemotherapy for retinoblastoma in eyes with vitreous and/or subretinal seeding: 2-year results. Br J Ophthalmol 96(4):499–502 Shields CL, Manjandavida FP, Lally SE, Pieretti G, Arepalli SA, Caywood EH et al, 2014, Intra-arterial chemotherapy for retinoblastoma in 70 eyes: outcomes based on the international classification of retinoblastoma. Ophthalmology 121(7):1453–1460 Munier FL, Beck-Popovic M, Balmer A, Gaillard MC, Bovey E, Binaghi S, 2011, Occurrence of sectoral choroidal occlusive vasculopathy and retinal arteriolar embolization after superselective ophthalmic artery chemotherapy for advanced intraocular retinoblastoma. Retina 31(3):566–573 Hannun YA, Obeid LM, 2008, Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol 9(2):139–150 Li W, Yu CP, Xia JT, Zhang L, Weng GX, Zheng HQ et al, 2009, Sphingosine kinase 1 is associated with gastric cancer progression and poor survival of patients. Clin Cancer Res 15(4):1393–1399 French KJ, Schrecengost RS, Lee BD, Zhuang Y, Smith SN, Eberly JL et al, 2003, Discovery and evaluation of inhibitors of human sphingosine kinase. Cancer Res 63(18):5962–5969 Johnson KR, Johnson KY, Crellin HG, Ogretmen B, Boylan AM, Harley RA et al, 2005, Immunohistochemical distribution of sphingosine kinase 1 in normal and tumor lung tissue. J Histochem Cytochem 53(9):1159–1166 Li J, Guan HY, Gong LY, Song LB, Zhang N, Wu J et al, 2008, Clinical significance of sphingosine kinase-1 expression in human astrocytomas progression and overall patient survival. Clin Cancer Res 14(21):6996–7003 Van Brocklyn JR, Jackson CA, Pearl DK, Kotur MS, Snyder PJ, Prior TW, 2005, Sphingosine kinase-1 expression correlates with poor survival of patients with glioblastoma multiforme: roles of sphingosine kinase isoforms in growth of glioblastoma cell lines. J Neuropathol Exp Neurol 64(8):695–705 Kohno M, Momoi M, Oo ML, Paik JH, Lee YM, Venkataraman K et al, 2006, Intracellular role for sphingosine kinase 1 in intestinal adenoma cell proliferation. Mol Cell Biol 26(19):7211–7223 Kawamori T, Kaneshiro T, Okumura M, Maalouf S, Uflacker A, Bielawski J et al, 2009, Role for sphingosine kinase 1 in colon carcinogenesis. FASEB J 23(2):405–414 Bayerl MG, Bruggeman RD, Conroy EJ, Hengst JA, King TS, Jimenez M et al, 2008, Sphingosine kinase 1 protein and mRNA are overexpressed in non-Hodgkin lymphomas and are attractive targets for novel pharmacological interventions. Leuk Lymphoma 49(5):948–954 Ruckhaberle E, Rody A, Engels K, Gaetje R, von Minckwitz G, Schiffmann S et al, 2008, Microarray analysis of altered sphingolipid metabolism reveals prognostic significance of sphingosine kinase 1 in breast cancer. Breast Cancer Res Treat 112(1): 41–52 Erez-Roman R, Pienik R, Futerman AH, 2010, Increased ceramide synthase 2 and 6 mRNA levels in breast cancer tissues and correlation with sphingosine kinase expression. Biochem Biophys Res Commun 391(1):219–223 Grammatikos G, TeichgraberV, Carpinteiro A, Trarbach T,WellerM, Hengge UR et al, 2007, Overexpression of acid sphingomyelinase sensitizes glioma cells to chemotherapy. Antioxid Redox Signal 9(9): 1449–1456 Morita Y, Perez GI, Paris F, Miranda SR, Ehleiter D, Haimovitz- Friedman A et al, 2000, Oocyte apoptosis is suppressed by disruption of the acid sphingomyelinase gene or by sphingosine-1- phosphate therapy. Nat Med 6(10):1109–1114 Lacour S, Hammann A, Grazide S, Lagadic-Gossmann D, Athias A, Sergent O et al, 2004, Cisplatin-induced CD95 redistribution into membrane lipid rafts of HT29 human colon cancer cells. Cancer Res 64(10):3593–3598 Akao Y, Banno Y, Nakagawa Y, Hasegawa N, Kim TJ, Murate T et al, 2006, High expression of sphingosine kinase 1 and S1P receptors in chemotherapy-resistant prostate cancer PC3 cells and their camptothecin-induced up-regulation. Biochem Biophys Res Commun 342(4):1284–1290 Pchejetski D, Golzio M, Bonhoure E, Calvet C, Doumerc N, Garcia V et al, 2005, Sphingosine kinase-1 as a chemotherapy sensor in prostate adenocarcinoma cell and mouse models. Cancer Res 65(24):11667–11675 Guillermet-Guibert J, Davenne L, Pchejetski D, Saint-Laurent N, Brizuela L, Guilbeau-Frugier C et al, 2009, Targeting the sphingolipid metabolism to defeat pancreatic cancer cell resistance to the chemotherapeutic gemcitabine drug. Mol Cancer Ther 8(4):809–820 Baran Y, Salas A, Senkal CE, Gunduz U, Bielawski J, Obeid LM et al, 2007, Alterations of ceramide/sphingosine 1-phosphate rheostat involved in the regulation of resistance to imatinib-induced apoptosis in K562 human chronic myeloid leukemia cells. J Biol Chem 282(15):10922–10934 Stephan H, Boeloeni R, Eggert A, Bornfeld N, Schueler A, 2008, Photodynamic therapy in retinoblastoma: effects of verteporfin on retinoblastoma cell lines. Invest Ophthalmol Vis Sci 49(7):3158–3163 Mergler S, Cheng Y, Skosyrski S, Garreis F, Pietrzak P, Kociok N et al, 2012, Altered calcium regulation by thermosensitive transient receptor potential channels in etoposide-resistant WERI-Rb1 retinoblastoma cells. Exp Eye Res 94(1):157–173 Pewzner-Jung Y, Tavakoli Tabazavareh S, Grassme H, Becker KA, Japtok L, Steinmann J et al, 2014, Sphingoid long chain bases prevent lung infection by Pseudomonas aeruginosa. EMBO Mol Med 6(9):1205–1214 Japtok L, Schmitz EI, Fayyaz S, Kramer S, Hsu LJ, Kleuser B, 2015, Sphingosine 1-phosphate counteracts insulin signaling in pancreatic beta-cells via the sphingosine 1-phosphate receptor subtype 2. FASEB J 29(8):3357–3369 Gulbins E, Palmada M, Reichel M, Luth A, Bohmer C, Amato D et al, 2013, Acid sphingomyelinase-ceramide system mediates effects of antidepressant drugs. Nat Med 19(7):934–938 Ye J, CoulourisG, Zaretskaya I, Cutcutache I, Rozen S, Madden TL, 2012, Primer-BLAST: a tool to design target-specific primers for polymerase chain reaction. BMC Bioinformatics 13:134 Sobue S, Nemoto S, Murakami M, Ito H, Kimura A, Gao S et al, 2008, Implications of sphingosine kinase 1 expression level for the cellular sphingolipid rheostat: relevance as a marker for daunorubicin sensitivity of leukemia cells. Int J Hematol 87(3):266–275 Xia P, Gamble JR, Wang L, Pitson SM, Moretti PA, Wattenberg BW et al, 2000, An oncogenic role of sphingosine kinase. Curr Biol: CB 10(23):1527–1530 Sankala HM, Hait NC, Paugh SW, Shida D, Lepine S, Elmore LW et al, 2007, Involvement of sphingosine kinase 2 in p53- independent induction of p21 by the chemotherapeutic drug doxorubicin. Cancer Res 67(21):10466–10474 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 391 EP 399 DI 10.1007/s12253-017-0360-x PG 9 ER PT J AU Toy, H Etli, O Celik, EZ Alikanoglu, SA AF Toy, Hatice Etli, Ozlem Celik, Esin Zeliha Alikanoglu, Sezgin Arsenal TI Associations Between Nucleus Size, and Immunohistochemical Galectin-3, Cytokeratine-19 and Hbme-1 Markers in Thyroid Papillary Carcinoma: A Morphometric Analyze SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary thyroid carcinoma; Nucleus diameter; Morphometric analyze ID Papillary thyroid carcinoma; Nucleus diameter; Morphometric analyze AB This study aimed to evaluate the morphometric measurements in cases with papillary thyroid carcinoma, and determine a cut-off value to support diagnosis. Fifty cases with a diagnosis of papillary thyroid carcinoma (PTC) were included in the study with their Galectine-3, CK-19 and HBME-1 immunohistochemical staining results. Demographic and clinical data gathered from pathology reports, which included demographic information such as patients’ sex, age, macroscopic tumor size, number of tumor focuses; prognostic parameters such as lenfovascular invasion, perineural invasion, thyroid capsule invasion; and results of immunohistochemical CK- 19, Galectin-3 and HBME-1 staining. Longest nuclear diameters of 150 tumor cells and 150 normal thyrocytes of each case were manually measured in an image analysis software, and mean longest nuclear diameters (MLND-TC and MLND-NC), and also tumor cell/normal cell longest nuclear diameter ratio (TC/NC-LNDR) were calculated. MLND-TC was higher than MLND-NC. The cases with higher MLND-TC had increased risk of capsule invasion in case of a negative staining with Galectine-3, HBME-1, or CK-19. When TC/NC-LNDR was high, number of tumor focus tended to be multiple and lymphovascular invasion risk was also increased. Subtypes of PTC were not differed regarding staining patterns. And finally, increased TC/NC-LNDR was associated with increased risk of having poor prognostic factors. The results of this study suggest that MLND-NC, MLND-TC, and TC/NCLNDR are valuable and easy-to-use measures, which can assist routine histology practice. C1 [Toy, Hatice] Necmettin Erbakan University Meram, Medicine Faculty, Department of PathologyKonya, Turkey. [Etli, Ozlem] Pendik State Hospital, Department of PathologyIstanbul, Turkey. [Celik, Esin Zeliha] Selcuk University, Medicine Faculty, Department of PathologyKonya, Turkey. [Alikanoglu, Sezgin Arsenal] Antalya Training and Research Hospital, Department of PathologyAntalya, Turkey. RP Toy, H (reprint author), Necmettin Erbakan University Meram, Medicine Faculty, Department of Pathology, Konya, Turkey. EM haticetoy@hotmail.com CR Cheung CC et al, 2001, Immunohistochemical diagnosis of papillary thyroid carcinoma. Mod Pathol 14(4):338–342 Coli A et al, 2002, Galectin-3, a marker of well-differentiated thyroid carcinoma, is expressed in thyroid nodules with cytological atypia. Histopathology 40(1):80–87 Collan Y, 1986, Diagnostic morphometry: relevant background to decision-making in diagnostic histopathology. In: In Science of Form: Proceedings of the First International Symposium for Science on Form 1986, KTK Scientific Publishers, Tokyo, p. 533- 542 Isic Dencic T et al, 2013, Cytokeratin19 expression discriminates papillary thyroid carcinoma from other thyroid lesions and predicts its aggressive behavior. Med Oncol 30(1):362 Kefeli M et al, 2010, Morphometric analysis in cytologic evaluation of papillary thyroid carcinoma. Anal Quant Cytol Histol 32(4): 234–238 Kumar V, et al, 2014, Robbins and cotran pathologic basis of disease – 2014. Online: ElsevierHealth Sciences Kuzey G, 2007, Temel Patoloji, 1st edn. Gunes Kitabevi, Istanbul Lee TK et al, 1987, The significance of nuclear diameter in the biologic behavior of thyroid carcinomas: a retrospective study of 127 cases. Hum Pathol 18(12):1252–1256 Lee KJ et al, 2011, Analysis of the clinicopathologic features of papillary thyroid microcarcinoma based on 7-mm tumor size. World J Surg 35(2):318–323 Liu Z et al, 2014, Cytokeratin 19, thyroperoxidase, HBME-1 and galectin-3 in evaluation of aggressive behavior of papillary thyroid carcinoma. Int J Clin Exp Med 7(8):2304–2308 Mai KT et al, 2002, Reduced HBME-1 immunoreactivity of papillary thyroid carcinoma and papillary thyroid carcinoma-related neoplastic lesions with Hurthle cell and/or apocrine-like changes. Histopathology 40(2):133–142 de Matos PS et al, 2005, Usefulness of HBME-1, cytokeratin 19 and galectin-3 immunostaining in the diagnosis of thyroid malignancy. Histopathology 47(4):391–401 Mills SE, 2004, Sternberg’s diagnostic surgical pathology, vol 1, 4th edn. Lippincott Williams and Wilkins, Philadelphia Nagashima T, Suzuki M, Nakajima N, 1997, Cytologic morphometric approach for the prediction of lymph node involvement in papillary thyroid cancer. Anal Quant Cytol Histol 19(1):49–54 Saggiorato E et al, 2001, Galectin-3 as a presurgical immunocytodiagnostic marker of minimally invasive follicular thyroid carcinoma. J Clin Endocrinol Metab 86(11):5152– 5158 Salmon I et al, 1992, Comparison of morphonuclear features in normal, benign neoplastic thyroid tissue by digital cell image analysis. Anal Quant Cytol Histol 14(1):47–54 Schlumberger MJ, 2004, Papillary thyroid carcinoma. Orphanet Encyclopedia. https://www.orpha.net/data/patho/GB/uk-PTC.pdf. Accessed 16 Nov 2017 Seok JY, et al, 2015, Prognostic value of HBME - 1 in papillary thyroid carcinoma. In: 15th Congress of the Asian Association of Endocrine Surgeons. Seoul, Korea Sorensen FB, 1992, Quantitative analysis of nuclear size for objective malignancy grading: a review with emphasis on new, unbiased stereologic methods. Lab Investig 66(1):4–23 Wallander M et al, 2010, Follicular variant of papillary carcinoma: reproducibility of histologic diagnosis and utility of HBME-1 immunohistochemistry and BRAF mutational analysis as diagnostic adjuncts. Appl Immunohistochem Mol Morphol 18(3):231–235 Weber KB et al, 2004, The use of a combination of galectin-3 and thyroid peroxidase for the diagnosis and prognosis of thyroid cancer. Am J Clin Pathol 122(4):524–531 Wu G et al, 2013, Combined staining for immunohistochemical markers in the diagnosis of papillary thyroid carcinoma: improvement in the sensitivity or specificity? J Int Med Res 41(4):975–983 Zink D, Fischer AH, Nickerson JA, 2004, Nuclear structure in cancer cells. Nat Rev Cancer 4(9):677–687 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 401 EP 408 DI 10.1007/s12253-017-0337-9 PG 8 ER PT J AU Shibuya, I Takami, M Miyamoto, A Karakawa, A Dezawa, A Nakamura, Sh Kamijo, R AF Shibuya, Isao Takami, Masamichi Miyamoto, Arei Karakawa, Akiko Dezawa, Akira Nakamura, Shigeru Kamijo, Ryutaro TI In Vitro Study of the Effects of Denosumab on Giant Cell Tumor of Bone: Comparison with Zoledronic Acid SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Giant cell tumor of bone; Osteoclast; Bisphosphonate; Denosumab; Bone tumor ID Giant cell tumor of bone; Osteoclast; Bisphosphonate; Denosumab; Bone tumor AB Giant cell tumor of bone (GCTB) is a locally aggressive primary bone tumor that contains numerous osteoclasts formed from marrow-derived precursors through receptor activator of nuclear factor κ-B ligand (RANKL), an osteoclast differentiation factor expressed in neoplastic cells of GCTB. Denosumab, a fully human monoclonal antibody targeting RANKL, has recently been used for the treatment of GCTB, and superior treatment effects have been reported. The aimof this work was to elucidate the mechanismof action of denosumab, and the differences between denosumab and zoledronic acid at the level of GCTB cells.We isolated GCTB cells from 3 patients and separated them into osteoclasts, osteoclast precursors and proliferating spindle-shaped stromal cells (the true neoplastic component), and examined the action of denosumab on differentiation, survival and bone resorption activity of osteoclasts. Denosumab and zoledronic acid inhibited osteoclast differentiation from mononuclear cells containing osteoclast precursors. Zoledronic acid inhibited osteoclast survival, whereas an inhibitory effect of denosumab on osteoclast survival was not observed. The inhibitory effect on bone resorption by both agents was confirmed in culture on dentin slices. Furthermore, zoledronic acid showed dosedependent inhibition of cell growth of neoplastic cells whereas denosumab had no inhibitory effect on these cells. Denosumab has an inhibitory effect on osteoclast differentiation, but no inhibitory effects on survival of osteoclasts or growth of neoplastic cells in GCTBs. C1 [Shibuya, Isao] Showa University, School of Dentistry, Department of Biochemistry, 1-5-8 Hatanodai, Shinagawa-ku, 142-8555 Tokyo, Japan. [Takami, Masamichi] Showa University, School of Dentistry, Department of Pharmacology, 1-5-8 Hatanodai, Shinagawa-ku, 142-8555 Tokyo, Japan. [Miyamoto, Arei] Showa University, School of Dentistry, Department of Biochemistry, 1-5-8 Hatanodai, Shinagawa-ku, 142-8555 Tokyo, Japan. [Karakawa, Akiko] Showa University, School of Dentistry, Department of Pharmacology, 1-5-8 Hatanodai, Shinagawa-ku, 142-8555 Tokyo, Japan. [Dezawa, Akira] Teikyo University, Mizonokuchi Hospital, Department of Orthopaedic Surgery, 3-8-3 Mizonokuchi, Takatsu-ku, 213-8507 Kawasaki, Kanagawa, Japan. [Nakamura, Shigeru] Teikyo University, Mizonokuchi Hospital, Department of Orthopaedic Surgery, 3-8-3 Mizonokuchi, Takatsu-ku, 213-8507 Kawasaki, Kanagawa, Japan. [Kamijo, Ryutaro] Showa University, School of Dentistry, Department of Biochemistry, 1-5-8 Hatanodai, Shinagawa-ku, 142-8555 Tokyo, Japan. RP Shibuya, I (reprint author), Showa University, School of Dentistry, Department of Biochemistry, 142-8555 Tokyo, Japan. EM ishibu@med.teikyo-u.ac.jp CR Wulling M, Engels C, Jesse N et al, 2001, The nature of giant cell tumor of bone. J Cancer Res Clin Oncol 127:467–474 Stewart MJ, Richardson TR, 1952, Giant cell tumor of bone. J Bone Joint Surg, Am Vol, 34-A:272–286 Raskin KA, Schwab JH, Mankin HJ et al, 2013, Giant cell tumor of bone. J Am Acad Orthop Surg 21:118–126 Chambers TJ, Fuller K, McSheehy PM et al, 1985, The effects of calcium regulating hormones on bone resorption by isolated human osteoclastoma cells. J Pathol 145:297–305 Kanehisa J, Izumo T, Takeuchi M et al, 1991, In vitro bone resorption by isolated multinucleated giant cells from giant cell tumour of bone: light and electron microscopic study. Virchows Arch A Pathol Anat Histopathol 419:327–338 Drake FH, Dodds RA, James IE et al, 1996, Cathepsin K, but not cathepsins B, L, or S, is abundantly expressed in human osteoclasts. J Biol Chem 271:12511–12516 Huang L, Xu J, Wood DJ et al, 2000, Gene expression of osteoprotegerin ligand, osteoprotegerin, and receptor activator of NFkappaB in giant cell tumor of bone: possible involvement in tumor cell-induced osteoclast-like cell formation. Am J Pathol 156:761– 767 Joyner CJ, Quinn JM, Triffitt JT et al, 1992, Phenotypic characterisation of mononuclear and multinucleated cells of giant cell tumour of bone. Bone Miner 16:37–48 Lau YS, Sabokbar A, Gibbons CL et al, 2005, Phenotypic and molecular studies of giant-cell tumors of bone and soft tissue. Hum Pathol 36:945–954 Campanacci M, Baldini N, Boriani S et al, 1987, Giant-cell tumor of bone. J Bone Joint Surg Am 69:106–114 Arbeitsgemeinschaft K, Becker WT, Dohle J et al, 2008, Local recurrence of giant cell tumor of bone after intralesional treatment with and without adjuvant therapy. J Bone Joint Surg Am 90:1060– 1067 Mori Y, Tsuchiya H, Karita M et al, 2000, Malignant transformation of a giant cell tumor 25 years after initial treatment. ClinOrthop Relat Res 381:185–191 Rock MG, Sim FH, Unni KK et al, 1986, Secondary malignant giant-cell tumor of bone. Clinicopathological assessment of nineteen patients. J Bone Joint Surg Am 68:1073–1079 Nascimento AG, Huvos AG, Marcove RC, 1979, Primary malignant giant cell tumor of bone: a study of eight cases and review of the literature. Cancer 44:1393–1402 Bertoni F, Present D, Sudanese A et al, 1988, Giant-cell tumor of bone with pulmonary metastases. Six case reports and a review of the literature. Clin Orthop Relat Res 237:275–285 Kay RM, Eckardt JJ, Seeger LL et al, 1994, Pulmonary metastasis of benign giant cell tumor of bone. Six histologically confirmed cases, including one of spontaneous regression. Clin Orthop Relat Res 302:219–230 Nakashima T, Hayashi M, Takayanagi H, 2012, New insights into osteoclastogenic signaling mechanisms. Trends Endocrinol Metab 23:582–590 Suda T, Takahashi N, Udagawa N et al, 1999, Modulation of osteoclast differentiation and function by the newmembers of the tumor necrosis factor receptor and ligand families. Endocr Rev 20:345– 357 Blair HC, Athanasou NA, 2004, Recent advances in osteoclast biology and pathological bone resorption. Histol Histopathol 19: 189–199 Shiotani A, Takami M, Itoh K et al, 2002, Regulation of osteoclast differentiation and function by receptor activator of NFkB ligand and osteoprotegerin. Anat Rec 268:137–146 Blackley HR, Wunder JS, Davis AM et al, 1999, Treatment of giant-cell tumors of long bones with curettage and bone-grafting. J Bone Joint Surg Am 81:811–820 XCY, Xu M, Song RXet al, 2010, Long-term outcome of giant cell tumors of bone around the knee treated by en bloc resection of tumor and reconstruction with prosthesis. Orthop Surg 2:211–217 Puthoor DK, Puthezhath K, 2012, Management of giant cell tumor of bone: computerized tomography based selection strategy and approaching the lesion through the site of cortical break. Orthop Surg 4:76–82 Singh AS, Chawla NS, Chawla SP, 2015)Giant-cell tumor of bone: treatment options and role of denosumab. Biologics 9:69–74 Tse LF,Wong KC, Kumta SM et al, 2008, Bisphosphonates reduce local recurrence in extremity giant cell tumor of bone: a casecontrol study. Bone 42:68–73 Balke M, Campanacci L, Gebert C et al, 2010, Bisphosphonate treatment of aggressive primary, recurrent and metastatic Giant cell tumour of bone. BMC Cancer 10:462 Yang T, Zheng XF, Lin X et al, 2013, Postoperative irrigation with bisphosphonates may reduce the recurrence of giant cell tumor of bone. Med Hypotheses 81:851–852 Gouin F, Rochwerger AR, Di Marco A et al, 2014, Adjuvant treatment with zoledronic acid after extensive curettage for giant cell tumours of bone. Eur J Cancer 50:2425–2431 Bekker PJ,Holloway DL, Rasmussen AS et al, 2004, Asingle-dose placebo-controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res 19:1059–1066 Lacey DL, Boyle WJ, Simonet WS et al, 2012, Bench to bedside: elucidation of the OPG-RANK-RANKL pathway and the development of denosumab. Nat Rev Drug Discov 11:401–419 Thomas D, Henshaw R, Skubitz K et al, 2010, Denosumab in patients with giant-cell tumour of bone: an open-label, phase 2 study. Lancet Oncol 11:275–280 Lewiecki EM, 2010, Clinical use of denosumab for the treatment for postmenopausal osteoporosis. Curr Med Res Opin 26:2807– 2812 Morgan T, Atkins GJ, Trivett MK et al, 2005, Molecular profiling of giant cell tumor of bone and the osteoclastic localization of ligand for receptor activator of nuclear factor kappaB. Am J Pathol 167:117–128 Roux S, Amazit L,Meduri G et al, 2002, RANK, receptor activator of nuclear factor kappa B, and RANK ligand are expressed in giant cell tumors of bone. Am J Clin Pathol 117:210–216 Thomas DM, 2012, RANKL, denosumab, and giant cell tumor of bone. Curr Opin Oncol 24:397–403 Hakozaki M, Tajino T, Yamada H et al, 2014, Radiological and pathological characteristics of giant cell tumor of bone treated with denosumab. Diagn Pathol 9:111 Yamagishi T, Kawashima H, Ogose A et al, 2016, Disappearance of giant cells and presence of newly formed bone in the pulmonary metastasis of a sacral giant-cell tumor following denosumab treatment: a case report. Oncol Lett 11:243–246 van Beek E, Pieterman E, Cohen L et al, 1999, Farnesyl pyrophosphate synthase is the molecular target of nitrogen-containing bisphosphonates. Biochem Biophys Res Commun 264:108–111 Martin M, Bell R, Bourgeois H et al, 2012, Bone-related complications and quality of life in advanced breast cancer: results from a randomized phase III trial of denosumab versus zoledronic acid. Clin Cancer Res 18:4841–4849 Lipton A, Fizazi K, Stopeck AT et al, 2012, Superiority of denosumab to zoledronic acid for prevention of skeletal-related events: a combined analysis of 3 pivotal, randomised, phase 3 trials. Eur J Cancer 48:3082–3092 Scagliotti GV, Hirsh V, Siena S et al, 2012, Overall survival improvement in patients with lung cancer and bone metastases treated with denosumab versus zoledronic acid: subgroup analysis from a randomized phase 3 study. J Thorac Oncol 7:1823–1829 Branstetter DG, Nelson SD, Manivel JC et al, 2012, Denosumab induces tumor reduction and bone formation in patients with giantcell tumor of bone. Clin Cancer Res 18:4415–4424 Cheng YY, Huang L, Lee KM et al, 2004, Bisphosphonates induce apoptosis of stromal tumor cells in giant cell tumor of bone. Calcif Tissue Int 75:71–77 Nishisho T, Hanaoka N,Miyagi R et al, 2015, Local administration of zoledronic acid for giant cell tumor of bone. Orthopedics 38:e25– e30 Salerno M, Avnet S, Alberghini M et al, 2008, Histogenetic characterization of giant cell tumor of bone. Clin Orthop Relat Res 466: 2081–2091 Avnet S, Salerno M, Zini N et al, 2013, Sustained autocrine induction and impaired negative feedback of osteoclastogenesis in CD14(+, cells of giant cell tumor of bone. Am J Pathol 182: 1357–1366 Takami M, Takahashi N, Udagawa N et al, 2000, Intracellular calcium and protein kinase C mediate expression of receptor activator of nuclear factor-kappaB ligand and osteoprotegerin in osteoblasts. Endocrinology 141:4711–4719 Agarwal A, Larsen BT, Buadu LD et al, 2013, Denosumab chemotherapy for recurrent giant-cell tumor of bone: a case report of neoadjuvant use enabling complete surgical resection. Case Rep Oncol Med 2013:496351 Akaike K, Suehara Y, Takagi T et al, 2014, An eggshell-like mineralized recurrent lesion in the popliteal region after treatment of giant cell tumor of the bone with denosumab. Skelet Radiol 43: 1767–1772 Mattei TA, Ramos E, Rehman AA et al, 2014, Sustained long-term complete regression of a giant cell tumor of the spine after treatment with denosumab. Spine J 14:e15–e21 Atkins J, Kostakis P, Vincent C et al, 2006, RANK expression as a cell surface marker of human osteoclast precursors in peripheral blood, bone marrow, and giant cell tumors of bone. J Bone Miner Res 21:1339–1349 Liverani C, Mercatali L, Spadazzi C et al, 2014, CSF-1 blockade impairs breast cancer osteoclastogenic potential in co-culture systems. Bone 66:214–222 Cheng YY, Huang L, Kumta SM et al, 2003, Cytochemical and ultrastructural changes in the osteoclast-like giant cells of giant cell tumor of bone following bisphosphonate administration. Ultrastruct Pathol 27:385–391 Lau CP, Huang L, Wong KC et al, 2013, Comparison of the antitumor effects of denosumab and zoledronic acid on the neopastic cells of giant cell tumor of bone. Connect Tissue Res 54:439–449 Ueda T, Morioka H, Nishida Y et al, 2015, Objective tumor response to denosumab in patients with giant cell tumor of bone: a multicenter phase II trial. Ann Oncol 26:2149–2154 Akiyama T, Choong PF, Dass CR, 2010, RANK-fc inhibits malignancy via inhibiting ERK activation and evoking caspase-3- mediated anoikis in human osteosarcoma cells. Clin Exp Metastasis 27:207–215 James IE, Dodds RA, Olivera DL et al, 1996, Human osteoclastoma-derived stromal cells: correlation of the ability to form mineralized nodules in vitro with formation of bone in vivo. J BoneMiner Res 11:1453–1460 Murata A, Fujita T, Kawahara N et al, 2005, Osteoblast lineage properties in giant cell tumors of bone. J Orthop Sci 10:581–588 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 409 EP 419 DI 10.1007/s12253-017-0362-8 PG 11 ER PT J AU Bulibu, J Wang, W Tang, Y Li, N Saifuding, K AF Bulibu, Jilisihan Wang, Wei Tang, Yong Li, Na Saifuding, Keyoumu TI Association Between Polymorphisms in the Promoter Region of microRNA-34b/c and the Chemoradiotherapy Efficacy for Locally Advanced Esophageal Squamous Cell Carcinoma in Chinese Han Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MicroRNA-34b/c; Esophageal squamous cell carcinoma; Promoter region; Polymorphism; Chemoradiotherapy efficacy; Chinese Han population ID MicroRNA-34b/c; Esophageal squamous cell carcinoma; Promoter region; Polymorphism; Chemoradiotherapy efficacy; Chinese Han population AB The study aims to explore the association between polymorphisms in the promoter region of microRNA-34b/c (miR-34b/c) and the chemoradiotherapy efficacy for locally advanced esophageal squamous cell carcinoma (ESCC) in Chinese Han population. A total of 175 locally advanced ESCC cases and 186 healthy individuals were enrolled as the case and control groups. Denaturing high performance liquid chromatography (DHPLC) was applied to determine the genotypes of subjects. Subjects in the case group were classified into complete response (CR), partial response (PR), stable disease (SD) and progressive disease (PD). CR + PR were defined as the sensitive group, and SD + PD were defined as the resistance group. All patients were followed up for 3 ~ 36 months. Receiver operating characteristic (ROC) curve was used to evaluate the predictive value of rs4938723 in the promoter region of miR-34b/c in the chemoradiotherapy efficacy for patients with locally advanced ESCC. The distribution of genotype and allele of rs4938723 in the promoter region of miR-34b/c was significantly different between the case and control group (both P < 0.05), and CC genotype and C allele could decrease the risk of ESCC (CC genotype: OR = 0.57, 95% CI = 0.32 ~ 0.99, P = 0.045; C allele: OR = 0.72, 95% CI = 0.54 ~ 0.97, P = 0.032). MiR-34b/c rs4938723 was associated with ESCC TNM staging, differentiation degree, and lymph node metastasis (LNM) for ES CC patients (all P < 0.05). The chemoradiotherapy efficacy of patients with CC genotype was better than that of patients with (TT + TC) genotypes (P < 0.05). ROC curve results showed that the area under curve (AUC), sensitivity and specificity were 0.777, 85.1% and 71.3%, respectively. The average median progression free survival (PFS) of patients with (TT + TC) genotypes was significantly shorter than those patients with CC genotype (P < 0.05). Our study provides evidence that miR-34b/c rs4938723 is closely related with the chemoradiotherapy efficacy for locally advanced ESCC. C1 [Bulibu, Jilisihan] The Affiliated Tumor Hospital of Xinjiang Medical University, Department of Gastroenterology, No. 789, Suzhou East Street, 830000 Urumqi, Xinjiang Uygur Autonomous Region, China. [Wang, Wei] The Affiliated Tumor Hospital of Xinjiang Medical University, Department of Gastroenterology, No. 789, Suzhou East Street, 830000 Urumqi, Xinjiang Uygur Autonomous Region, China. [Tang, Yong] The Affiliated Tumor Hospital of Xinjiang Medical University, Department of Gastroenterology, No. 789, Suzhou East Street, 830000 Urumqi, Xinjiang Uygur Autonomous Region, China. [Li, Na] The Affiliated Tumor Hospital of Xinjiang Medical University, Department of Gastroenterology, No. 789, Suzhou East Street, 830000 Urumqi, Xinjiang Uygur Autonomous Region, China. [Saifuding, Keyoumu] The Affiliated Tumor Hospital of Xinjiang Medical University, Department of Gastroenterology, No. 789, Suzhou East Street, 830000 Urumqi, Xinjiang Uygur Autonomous Region, China. RP Saifuding, K (reprint author), The Affiliated Tumor Hospital of Xinjiang Medical University, Department of Gastroenterology, 830000 Urumqi, China. EM saifudingkeyoumu@126.com CR Chen J, Kwong DL, Cao T, Hu Q, Zhang L,Ming X, Chen J, Fu L, Guan X, 2015, Esophageal squamous cell carcinoma, ESCC): advance in genomics and molecular genetics. Dis Esophagus 28:84– 89 Wu C, Hu Z, He Z, Jia W, Wang F, Zhou Y, Liu Z, Zhan Q, Liu Y, Yu D, Zhai K, Chang J, Qiao Y, Jin G, Liu Z, Shen Y, Guo C, Fu J, Miao X, Tan W, Shen H, Ke Y, Zeng Y, Wu T, Lin D, 2011, Genome-wide association study identifies three new susceptibility loci for esophageal squamous-cell carcinoma in Chinese populations. Nat Genet 43:679–684 ZhangX, Li M, Meng X, Kong L, ZhangY,Wei G, Zhang X, Shi F, HuM, Zhang G, Yu J, 2014, Involved-field irradiation in definitive chemoradiotherapy for locally advanced esophageal squamous cell carcinoma. Radiat Oncol 9:64 Cho SH, Shim HJ, Lee SR, Ahn JS, Yang DH, Kim YK, Nam TK, Lee JJ, Kim HJ, Chung IJ, 2008, Concurrent chemoradiotherapy with S-1 and cisplatin in advanced esophageal cancer. Dis Esophagus 21:697–703 Li DJ, Li HW, He B, Wang GM, Cai HF, Duan SM, Liu JJ, Zhang YJ, Cui Z, Jiang H, 2016, Patterns of failure after involved field radiotherapy for locally advanced esophageal squamous cell carcinoma. J BUON 21:1268–1273 Hiyoshi Y, Kamohara H, Karashima R, Sato N, Imamura Y, Nagai Y, Yoshida N, Toyama E, Hayashi N,Watanabe M, Baba H, 2009, MicroRNA-21 regulates the proliferation and invasion in esophageal squamous cell carcinoma. Clin Cancer Res 15:1915–1922 Kano M, Seki N, Kikkawa N, Fujimura L, Hoshino I, Akutsu Y, Chiyomaru T, Enokida H, NakagawaM,Matsubara H, 2010, miR- 145, miR-133a and miR-133b: tumor-suppressive miRNAs target FSCN1 in esophageal squamous cell carcinoma. Int J Cancer 127: 2804–2814 Corney DC, Flesken-Nikitin A, Godwin AK,Wang W, Nikitin AY, 2007, MicroRNA-34b and MicroRNA-34c are targets of p53 and cooperate in control of cell proliferation and adhesion-independent growth. Cancer Res 67:8433–8438 Suzuki H, Yamamoto E, Nojima M, Kai M, Yamano HO, Yoshikawa K, Kimura T, Kudo T, Harada E, Sugai T, Takamaru H, Niinuma T, Maruyama R, Yamamoto H, Tokino T, Imai K, Toyota M, Shinomura Y, 2010, Methylation-associated silencing of microRNA-34b/c in gastric cancer and its involvement in an epigenetic field defect. Carcinogenesis 31:2066–2073 ToyotaM, Suzuki H, Sasaki Y, Maruyama R, Imai K, Shinomura Y, Tokino T, 2008, Epigenetic silencing of microRNA-34b/c and Bcell translocation gene 4 is associated with CpG island methylation in colorectal cancer. Cancer Res 68:4123–4132 Lin Z, Chen L, Song M, Shi KQ, Tang KF, 2014, Association between a polymorphism in miR-34b/c and susceptibility to cancer–a meta-analysis. Asian Pac J Cancer Prev 15:7251–7255 Wang X, Lu X, Fang Y, Chen H, Deng X, Peng C, Li H, Shen B, 2014, Association between miR34b/c polymorphism rs4938723 and cancer risk: a meta-analysis of 11 studies including 6169 cases and 6337 controls. Med Sci Monit 20:1977–1982 Rice TW, 2015, Esophageal cancer staging. Korean J Thorac Cardiovasc Surg 48:157–163 Sugimura K, Miyata H, Tanaka K, Hamano R, Takahashi T, Kurokawa Y, Yamasaki M, Nakajima K, Takiguchi S, Mori M, Doki Y, 2012, Let-7 expression is a significant determinant of response to chemotherapy through the regulation of IL-6/STAT3 pathway in esophageal squamous cell carcinoma. Clin Cancer Res 18:5144–5153 Guo Y, Chen Z, Zhang L, Zhou F, Shi S, Feng X, Li B,Meng X, Ma X, Luo M, Shao K, Li N, Qiu B, Mitchelson K, Cheng J, He J, 2008, Distinctive microRNA profiles relating to patient survival in esophageal squamous cell carcinoma. Cancer Res 68:26–33 Ji TX, Zhi C, Guo XG, Zhou Q,Wang GQ, Chen B,Ma FF, 2015, MiR-34b/c rs4938723 polymorphism significantly decreases the risk of digestive tract cancer: meta-analysis. Asian Pac J Cancer Prev 16:6099–6104 Xu Y, Liu L, Liu J, Zhang Y, Zhu J, Chen J, Liu S, Liu Z, Shi H, Shen H, Hu Z, 2011, A potentially functional polymorphism in the promoter region of miR-34b/c is associated with an increased risk for primary hepatocellular carcinoma. Int J Cancer 128:412–417 Wang Z, Wu J, Zhang G, Cao Y, Jiang C, Ding Y, 2013, Associations of miR-499 and miR-34b/c polymorphisms with susceptibility to hepatocellular carcinoma: an evidence-based evaluation. Gastroenterol Res Pract 2013:719202 Son MS, JangMJ, Jeon YJ, Kim WH, Kwon CI, Ko KH, Park PW, Hong SP, Rim KS, Kwon SW, Hwang SG, Kim NK, 2013, Promoter polymorphisms of pri-miR-34b/c are associated with hepatocellular carcinoma. Gene 524:156–160 Yin J,Wang X, Zheng L, Shi Y,Wang L, Shao A, TangW, Ding G, Liu C, Liu R, Chen S, Gu H, 2013, Hsa-miR-34b/c rs4938723 T>C and hsa-miR-423 rs6505162 C>A polymorphisms are associated with the risk of esophageal cancer in a Chinese population. PLoS One 8:e80570 Corsini LR, Bronte G, Terrasi M, Amodeo V, Fanale D, Fiorentino E, Cicero G, Bazan V, Russo A, 2012, The role of microRNAs in cancer: diagnostic and prognostic biomarkers and targets of therapies. Expert Opin Ther Targets 16(Suppl 2):S103–S109 Hermeking H, 2010, The miR-34 family in cancer and apoptosis. Cell Death Differ 17:193–199 Kumamoto K, Spillare EA, Fujita K, Horikawa I, Yamashita T, Appella E, Nagashima M, Takenoshita S, Yokota J, Harris CC, 2008, Nutlin-3a activates p53 to both down-regulate inhibitor of growth 2 and up-regulate mir-34a, mir-34b, and mir-34c expression, and induce senescence. Cancer Res 68:3193–3203 Cho CY, Huang JS, Shiah SG, Chung SY, Lay JD, Yang YY, Lai GM, Cheng AL, Chen LT, Chuang SE, 2016, Negative feedback regulation of AXL by miR-34a modulates apoptosis in lung cancer cells. RNA 22:303–315 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 421 EP 427 DI 10.1007/s12253-017-0366-4 PG 7 ER PT J AU Li, X Wang, X Xie, J Liang, B Wu, J AF Li, Xiaohui Wang, Xinjun Xie, Jingwei Liang, Bo Wu, Jianheng TI Suppression of Angiotensin-(1–7) on the Disruption of Blood-Brain Barrier in Rat of Brain Glioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ang-(1-7); Glioma; Blood-brain barrier; Claudin-5; ZO-1 ID Ang-(1-7); Glioma; Blood-brain barrier; Claudin-5; ZO-1 AB Glioblastoma multiforme (GBM) is the most primary brain tumor, specially characterized with the damage of blood-brain barrier (BBB). The Ang-(1–7) was proven to have an inhibitory effect on glioblastoma growth. However, its role on blood–brain barrier (BBB) and the underlying molecular mechanism remains unclear. In this study, Ang-(1–7) significantly relieved the damage of blood-brain barrier in rats with intracranial U87 gliomas as evaluated by magnetic resonance imaging (MRI). Furthermore, its treatment attenuated BBB permeability, tumor growth and edema formation. Similarly, Ang-(1–7) also decreased U87 glioma cells barrier permeability in vitro. Further analysis showed that Ang-(1–7) could effectively restore tight junction protein (claudin-5 and ZO-1) expression levels both in rats and U87 glioma cells by affecting the activation of JNK pathway. SP600125, an inhibitor of JNK, significantly enhanced the expression of Claudin-5 and ZO-1, and decreased the disruption of BBB and enhanced the efficiency of Ang-(1–7) in glioma rats. Taken together, this study demonstrated a protective role of Ang-(1–7) in glioma-induced blood-brain barrier damage by regulating tight junction protein expression. Accordingly, Ang-(1– 7) may become a promising therapeutic agent against glioma. C1 [Li, Xiaohui] The Fifth Affiliated Hospital of Zhengzhou University, Department of Neurosurgery, No.3 Kangfuqian Street, Erqi District, 450052 Zhengzhou, China. [Wang, Xinjun] The Fifth Affiliated Hospital of Zhengzhou University, Department of Neurosurgery, No.3 Kangfuqian Street, Erqi District, 450052 Zhengzhou, China. [Xie, Jingwei] The Fifth Affiliated Hospital of Zhengzhou University, Department of Neurosurgery, No.3 Kangfuqian Street, Erqi District, 450052 Zhengzhou, China. [Liang, Bo] The Fifth Affiliated Hospital of Zhengzhou University, Department of Neurosurgery, No.3 Kangfuqian Street, Erqi District, 450052 Zhengzhou, China. [Wu, Jianheng] The Fifth Affiliated Hospital of Zhengzhou University, Department of Neurosurgery, No.3 Kangfuqian Street, Erqi District, 450052 Zhengzhou, China. RP Wang, X (reprint author), The Fifth Affiliated Hospital of Zhengzhou University, Department of Neurosurgery, 450052 Zhengzhou, China. EM wangjiang066@sina.com CR Hawkins BT, Davis TP, 2005, The blood-brain barrier/ neurovascular unit in health and disease. Pharmacol Rev 57(2): 173–185 Yang Y, Rosenberg GA, 2011, Blood–brain barrier breakdown in acute and chronic cerebrovascular disease. Stroke 42(11):3323– 3328 Wang Z, Leng Y, Tsai L-K, Leeds P, Chuang DM, 2011, Valproic acid attenuates blood–brain barrier disruption in a rat model of transient focal cerebral ischemia: the roles of HDAC and MMP-9 inhibition. J Cereb Blood Flow Metab 31(1):52–57 Mendes B, Marques C, Carvalho I, Costa P, Martins S, Ferreira D, Sarmento B, 2015, Influence of glioma cells on a new co-culture in vitro blood-brain barrier model for characterization and validation of permeability. Int J Pharm 490(1–2):94–101 Barrier L, Fauconneau B, Noel A, Ingrand S, 2010, Ceramide and related-sphingolipid levels are not altered in disease-associated brain regions of APPSL, and APPSL/PS1M146L mouse models of alzheimer's disease: relationship with the lack of neurodegeneration. Int J Alzheimers Dis 2011:920958 Jiao H,Wang Z, Liu Y,Wang P, Xue Y, 2011, Specific role of tight junction proteins claudin-5, occludin, and ZO-1 of the blood–brain barrier in a focal cerebral ischemic insult. J Mol Neurosci 44(2): 130–139 Ferrario CM, Jessup J, Gallagher PE, Averill DB, Brosnihan KB, Ann Tallant E, Smith RD, Chappell MC, 2005, Effects of reninangiotensin system blockade on renal angiotensin-(1-7, forming enzymes and receptors. Kidney Int 68(5):2189–2196 Nishimura Y, Ito T, Saavedra JM, 2000, Angiotensin II AT1 blockade normalizes cerebrovascular autoregulation and reduces cerebral ischemia in spontaneously hypertensive rats. Stroke 31(10):2478– 2486 Fleegal-DeMotta MA, Doghu S, Banks WA, 2009, Angiotensin II modulates BBB permeability via activation of the AT1 receptor in brain endothelial cells. J Cereb Blood Flow Metab 29(3):640–647 Benter IF, Yousif MH, Cojocel C et al, 2007, Angiotensin-(1–7, prevents diabetes-induced cardiovascular dysfunction. Am J Phys Heart Circ Phys 292(1):H666–H672 Weidensteiner C, ReichardtW, Shami PJ et al, 2013, Effects of the nitric oxide donor JS-K on the blood-tumor barrier and on orthotopic U87 rat gliomas assessed by MRI. Nitric Oxide 30:17– 25 Patabendige A, Skinner RA, Abbott NJ, 2013, Establishment of a simplified< i> in vitro porcine blood–brain barrier model with high transendothelial electrical resistance. Brain Res 1521:1–15 Amasheh S, Schmidt T,MahnM, Florian P,Mankertz J, Tavalali S, Gitter AH, Schulzke JD, FrommM(2005, Contribution of claudin- 5 to barrier properties in tight junctions of epithelial cells. Cell Tissue Res 321(1):89–96 Alsadi R, Ye D, Boivin M et al, 2013, Interleukin-6 modulation of intestinal epithelial tight junction permeability is mediated by JNK pathway activation of claudin-2 gene. PLoS One 9(3):e85345 Huhndorf M, Moussavi A, Kramann N, Will O, Hattermann K, Stadelmann C, Jansen O, Boretius S, 2016, Alterations of the blood-brain barrier and regional perfusion in tumor development: MRI insights from a rat C6 glioma model. PLoS One 11(12): e0168174 Subashi E, Cordero FJ, Halvorson KG et al, 2016, Tumor location, but not H3.3K27M, significantly influences the blood–brain-barrier permeability in a genetic mouse model of pediatric high-grade glioma. J Neuro-Oncol 126(2):1–9 Stegmayr C, Oliveira D, Niemietz N et al, 2017, Influence of bevacizumab on blood-brain barrier permeability and O-(2-18Ffluoroethyl)- L-tyrosine uptake in rat gliomas. Eur J Nucl Med Mol Imaging 44(3):408–416 HuangY, Hoffman C, Rajappa P, Kim JH, HuW, Huse J, Tang Z, Li X, Weksler B, Bromberg J, Lyden DC, Greenfield JP, 2013, Oligodendrocyte progenitor cells promote neovascularization in glioma by disrupting the blood-brain barrier. Cancer Res 74(4): 1011–1021 Liu Y, Wang D, Wang H et al, 2014, The protective effect of HET0016 on brain edema and blood–brain barrier dysfunction after cerebral ischemia/reperfusion. Brain Res 1544:45–53 Zlokovic BV, 2008, The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron 57(2):178–201 Walter FR, Veszelka S, Pasztoi M, Peterfi ZA, Toth A, Rakhely G, Cervenak L, Abraham CS, Deli MA, 2015, Tesmilifene modifies brain endothelial functions and opens the blood–brain/blood–glioma barrier. J Neurochem 134(6):1040–1054 HuangY, Hoffman C, Rajappa P, Kim JH, HuW, Huse J, Tang Z, Li X, Weksler B, Bromberg J, Lyden DC, Greenfield JP, 2014, Oligodendrocyte progenitor cells promote neovascularization in glioma by disrupting the blood-brain barrier. Cancer Res 74(4): 1011–1021 Ostergaard L, Hochberg FH, Rabinov JD et al, 1999, Early changes measured by magnetic resonance imaging in cerebral blood flow, blood volume, and blood-brain barrier permeability following dexamethasone treatment in patients with brain tumors. J Neurosurg 90(2):300–305 Higashida T, Peng C, Li J, Dornbos D, Teng K, Li X, Kinni H, Guthikonda M, Ding Y, 2011, Hypoxia-inducible factor-1α contributes to brain edema after stroke by regulating aquaporins and glycerol distribution in brain. Curr Neurovasc Res 8(1):44–51 Doolittle ND, Miner ME, Hall WA, Siegal T, Hanson EJ, Osztie E, McAllister LD, Bubalo JS, Kraemer DF, Fortin D, Nixon R, Muldoon LL, Neuwelt EA, 2000, Safety and efficacy of a multicenter study using intraarterial chemotherapy in conjunction with osmotic opening of the blood-brain barrier for the treatment of patients with malignant brain tumors. Cancer 88(3):637–647 Nitta T, Hata M, Gotoh S, Seo Y, Sasaki H, Hashimoto N, Furuse M, Tsukita S, 2003, Size-selective loosening of the blood-brain barrier in claudin-5–deficient mice. J Cell Biol 161(3):653–660 Rodgers LS, Beam MT, Anderson JM, Fanning AS, 2013, Epithelial barrier assembly requires coordinated activity ofmultiple domains of the tight junction protein ZO-1. J Cell Sci 126(7):1565– 1575 Koto T, Takubo K, Ishida S, Shinoda H, Inoue M, Tsubota K, Okada Y, Ikeda E, 2007, Hypoxia disrupts the barrier function of neural blood vessels through changes in the expression of claudin-5 in endothelial cells. Am J Pathol 170(4):1389–1397 Chen D, Wei XT, Guan JH, Yuan JW, Peng YT, Song L, Liu YH, 2012, Inhibition of c-Jun N-terminal kinase prevents blood-brain barrier disruption and normalizes the expression of tight junction proteins clautin-5 and ZO-1 in a rat model of subarachnoid hemorrhage. Acta Neurochir 154(8):1469–1476 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 429 EP 435 DI 10.1007/s12253-018-0471-z PG 7 ER PT J AU Langabeer, ES Haslam, K AF Langabeer, E Stephen Haslam, Karl TI Neutrophilia and the JAK2 V617F Mutation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Langabeer, E Stephen] St. James’s Hospital, Central Pathology Laboratory, Cancer Molecular DiagnosticsDublin, Ireland. [Haslam, Karl] St. James’s Hospital, Central Pathology Laboratory, Cancer Molecular DiagnosticsDublin, Ireland. RP Langabeer, ES (reprint author), St. James’s Hospital, Central Pathology Laboratory, Cancer Molecular Diagnostics, Dublin, Ireland. EM slangabeer@stjames.ie CR Arber DA, Orazi A, Hasserjian R et al, 2016, The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127:2391–2405 Langabeer SE, 2012, Referral centre variation in requesting JAK2 V617F mutation analysis for the investigation of a myeloproliferative neoplasm. J Clin Pathol 65:1149–1150 Gong JZ, Cook JR, Greiner RC et al, 2013, Laboratory practice guidelines for detecting and reporting JAK2 and MPL mutations in myeloproliferative neoplasms: a report for the Association for Molecular Pathology. J Mol Diagn 15:733–744 Weir AB, Lewis JB Jr, Arteta-Bulos R, 2011, Chronic idiopathic neutrophilia: experience and recommendations. South Med J 104: 499–504 Ogunleye F, Ibrahim M, Allen E et al, 2016, BCR-ABL testing by polymerase chain reaction in patients with neutrophilia: the William Beaumont Hospital experience and the case for rational laboratory tests request. J Oncol Pract 12:e1001–e1005 Tefferi A, Noel P, Hanson CA, 2011, Uses and abuses of JAK2 and MPL mutation tests in myeloproliferative neoplasms. J Mol Diagn 13:461–466 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 437 EP 438 DI 10.1007/s12253-017-0315-2 PG 2 ER PT J AU Marton, I Posfai, Csomor, A Vecsei, L Borbenyi, Z Sas, K AF Marton, Imelda Posfai, Eva Csomor, Angela Vecsei, Laszlo Borbenyi, Zita Sas, Katalin TI Cerebrovascular Complications and Polycythaemia Vera SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Marton, Imelda] University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, Semmelweis 8Szeged, Hungary. [Posfai, Eva] University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, Semmelweis 8Szeged, Hungary. [Csomor, Angela] University of Szeged, Department of RadiologySzeged, Hungary. [Vecsei, Laszlo] University of Szeged, Albert Szent-Gyorgyi Clinical Centre, Medical Faculty, Department of NeurologySzeged, Hungary. [Borbenyi, Zita] University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, Semmelweis 8Szeged, Hungary. [Sas, Katalin] University of Szeged, Albert Szent-Gyorgyi Clinical Centre, Medical Faculty, Department of NeurologySzeged, Hungary. RP Marton, I (reprint author), University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, Szeged, Hungary. EM imeldamarton@gmail.com CR Falanga A, Marchetti M, 2012, Thrombotic disease in the myeloproliferative neoplasms. Hematology Am Soc Hematol Educ Program 2012:571–581. , DOI 10.1182/asheducation- 2012.1.571 2012/1/571 Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Vardiman JW, eds,, 2008)WHO classification of tumours of haematopoietic and lymphoid tissues. IARC, Lyon Teffe r i A, 2012, Polycythemia vera and essent i a l thrombocythemia: 2012 update on diagnosis, risk stratification, and management. Am J Hematol 87(3):285–293. , DOI 10.1002/ajh.23135 Zoraster RM, Rison RA, 2013, Acute embolic cerebral ischemia as an initial presentation of polycythemia vera: a case report. J Med Case Rep 7:131. , DOI 10.1186/1752-1947-7-131 Sanahuja J, Marti-Fabregas J, Marti-Vilalta JL, 2005, Carotid thrombus and cerebral infarction as the initial clinical manifestation of polycythemia vera. Neurologia 20(4):194–196 Arboix A, Besses C, 1997, Cerebrovascular disease as the initial clinical presentation of haematological disorders. Eur Neurol 37(4): 207–211 Abdel-Rahman I, Murphy C, 2015, 2015, Recurrent ischaemic stroke unveils polycythaemia vera. BMJ Case Rep. https://doi. org/10.1136/bcr-2014-207625 Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin N, Scott MA, Erber WN, Green AR, Cancer Genome P, 2005, Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 365(9464):1054–1061. , DOI 10.1016/S0140- 6736(05)71142-9 Teffe r i A, 2013, Polycythemia vera and essent i a l thrombocythemia: 2013 update on diagnosis, risk-stratification, and management. Am J Hematol 88(6):507–516. , DOI 1002/ajh.23417 10. Voko Z, Szeles G, Kardos L, Nemeth R, Adany R, 2008, The epidemiology of cerebrovascular diseases in Hungary after the millennium. LAM, Lege Artis Medicinae, 18(1):31–38 Adams BD, Baker R, Lopez JA, Spencer S, 2010, Myeloproliferative disorders and the hyperviscosity syndrome. Hematol Oncol Clin North Am 24(3):585–602. , DOI 10.1016/j.hoc.2010.03.004 Santisakultarm TP, Paduano CQ, Stokol T, Southard TL, Nishimura N, Skoda RC, Olbricht WL, Schafer AI, Silver RT, Schaffer CB, 2014, Stalled cerebral capillary blood flow in mouse models of essential thrombocythemia and polycythemia vera revealed by in vivo two-photon imaging. J Thromb Haemost. , DOI 10.1111/jth.1273 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2019 VL 25 IS 1 BP 439 EP 442 DI 10.1007/s12253-017-0329-9 PG 4 ER PT J AU Zhang, R Zhang, J Luo, W Luo, Z Shi, Sh AF Zhang, Rui Zhang, Jianwu Luo, Wei Luo, Zhuang Shi, Shaoqing TI WWP2 Is One Promising Novel Oncogene SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE WWP2; Cancer; Akt; Pathogenesis ID WWP2; Cancer; Akt; Pathogenesis AB WWP2 is an E3 ubiquitin ligase and plays an important role in regulation of many cellular biological activities through ubiquitination and degradation of its substrates. Recently accumulating evidences indicate that WWP2 plays a crucial part in the pathogenesis in different types of tumors. In this report, the role of this gene especially in tumorigenesis was reviewed. WWP2 is dysregulated in various of tumors, and it promotes carcinogenesis mainly through PTEN/Akt signaling pathway. WWP2 also participates in anti-cancer agents’ sensitivity, indicating WWP2 may be a novel target for cancer treatment.WWP2 is one promising novel oncogene. C1 [Zhang, Rui] The Seventh People’s Hospital of Chengdu, Department of Thoracic Surgery, 640021 Chengdu, Sichuan, China. [Zhang, Jianwu] North Sichuan Medical College, School of Pharmacy, 637100 Nanchong, Sichuan, China. [Luo, Wei] The People’s Hospital of Leshan, Department of Respiratory Medicine, 640000 Leshan, Sichuan, China. [Luo, Zhuang] The First Affiliated Hospital of Kunming Medical University, Department of Pumnary and Critical Care Medicine, 650032 Kunming, Yunnan, China. [Shi, Shaoqing] The First Affiliated Hospital of Kunming Medical University, Department of Pumnary and Critical Care Medicine, 650032 Kunming, Yunnan, China. RP Luo, Z (reprint author), The First Affiliated Hospital of Kunming Medical University, Department of Pumnary and Critical Care Medicine, 650032 Kunming, China. EM sky4511@126.com CR Li W, Bengtson MH, Ulbrich A, Matsuda A, Reddy VA, Orth A, Chanda SK, Batalov S, Joazeiro CA, 2008, Genome-wide and functional annotation of human E3 ubiquitin ligases identifies MULAN, a mitochondrial E3 that regulates the organelle's dynamics and signaling. PLoS One 3:e1487 Bernassola F, Karin M, Ciechanover A, Melino G, 2008, The HECT family of E3 ubiquitin ligases: multiple players in cancer development. Cancer Cell 14:10–21 Lu PJ, Zhou XZ, Shen M, Lu KP, 1999, Function ofWWdomains as phosphoserine- or phosphothreonine-binding modules. Science 283:1325–1328 Pirozzi G, McConnell SJ, Uveges AJ, Carter JM, Sparks AB, Kay BK, Fowlkes DM, 1997, Identification of novel human WW domain- containing proteins by cloning of ligand targets. J Biol Chem 272:14611–14616 Wood JD, Yuan J, Margolis RL, Colomer V, Duan K, Kushi J, Kaminsky Z, Kleiderlein JJ, Sharp AH, Ross CA, 1998, Atrophin-1, the DRPLA gene product, interacts with two families ofWWdomain-containing proteins.Mol CellNeurosci 11:149–160 Xu H,WangW, Li C, Yu H, Yang A,Wang B, Jin Y, 2009)WWP2 promotes degradation of transcription factor OCT4 in human embryonic stem cells. Cell Res 19:561–573 Soond SM, Chantry A, 2011, Selective targeting of activating and inhibitory Smads by distinct WWP2 ubiquitin ligase isoforms differentially modulates TGFbeta signalling and EMT. Oncogene 30: 2451–2462 Liao B, Jin Y, 2010, Wwp2 mediates Oct4 ubiquitination and its own auto-ubiquitination in a dosage-dependent manner. Cell Res 20:332–344 Xu HM, Liao B, Zhang QJ, Wang BB, Li H, Zhong XM, Sheng HZ, Zhao YX, Zhao YM, Jin Y, 2004, Wwp2, an E3 ubiquitin ligase that targets transcription factor Oct-4 for ubiquitination. J Biol Chem 279:23495–23503 McDonald FJ, Western AH, McNeil JD, Thomas BC, Olson DR, Snyder PM, 2002, Ubiquitin-protein ligase WWP2 binds to and downregulates the epithelial Na(+, channel. Am J Physiol Renal Physiol 283:F431–F436 Li H, Zhang Z, Wang B, Zhang J, Zhao Y, Jin Y, 2007, Wwp2- mediated ubiquitination of the RNA polymerase II large subunit in mouse embryonic pluripotent stem cells. Mol Cell Biol 27:5296– 5305 Luo X, Wang B, Tang F, Zhang J, Zhao Y, Li H, Jin Y, 2014, Wwp2 targets SRG3, a scaffold protein of the SWI/SNF-like BAF complex, for ubiquitination and degradation. Biochem Biophys Res Commun 443:1048–1053 Yang Y, Liao B, Wang S, Yan B, Jin Y, Shu HB, Wang YY, 2013, E3 ligase WWP2 negatively regulates TLR3-mediated innate immune response by targeting TRIF for ubiquitination and degradation. Proc Natl Acad Sci U S A 110:5115–5120 Lv Y, Zhang K, Gao H, 2014, Paip1, an effective stimulator of translation initiation, is targeted by WWP2 for ubiquitination and degradation. Mol Cell Biol 34:4513–4522 Chen A, Gao B, Zhang J, McEwen T, Ye SQ, Zhang D, Fang D, 2009, The HECT-type E3 ubiquitin ligaseAIP2 inhibits activationinduced T-cell death by catalyzing EGR2 ubiquitination. Mol Cell Biol 29:5348–5356 ChenW, Jiang X, Luo Z, 2014)WWP2: a multifunctional ubiquitin ligase gene. Pathol Oncol Res 20:799–803 Chantry A, 2011, WWP2 ubiquitin ligase and its isoforms: new biological insight and promising disease targets. Cell Cycle 10: 2437–2439 Choi BH, Che X, Chen C, Lu L, Dai W, 2015, WWP2 is required for normal cell cycle progression. Genes cancer 6:371–377 Clements AE, Bravo V, Koivisto C, Cohn DE, Leone G, 2015, WWP2 and its association with PTEN in endometrial cancer. Gynecol Oncol Rep 13:26–29 Fukumoto C, Nakashima D, Kasamatsu A, Unozawa M, Shida- Sakazume T, Higo M, Ogawara K, Yokoe H, Shiiba M, Tanzawa H, Uzawa K, 2014)WWP2 is overexpressed in human oral cancer, determining tumor size and poor prognosis in patients: downregulation of WWP2 inhibits the AKT signaling and tumor growth in mice. Oncoscience 1:807–820 Liang J, QiWF, Xie S,Wang WF, Zhang XL, Zhou XP, Hu JX, Yu RT, 2017, Expression of WW domain-containing protein 2 is correlated with pathological grade and recurrence of glioma. J Cancer Res Ther 13:1032–1037 Soond SM, Smith PG,Wahl L, Swingler TE, Clark IM, Hemmings AM, Chantry A, 2013, Novel WWP2 ubiquitin ligase isoforms as potential prognostic markers and molecular targets in cancer. Biochim Biophys Acta 1832:2127–2135 Xu SQ, Qin Y, Pan DB, Ye GX, Wu CJ, Wang S, Jiang JY, Fu J, Wang CJ, 2016, Inhibition ofWWP2 suppresses proliferation, and induces G1 cell cycle arrest and apoptosis in liver cancer cells. Mol Med Rep 13:2261–2266 Yang R, He Y, Chen S, Lu X, Huang C, Zhang G, 2016, Elevated expression ofWWP2 in human lung adenocarcinoma and its effect on migration and invasion. Biochem Biophys Res Commun 479: 146–151 Jung JG, Stoeck A, Guan B,Wu RC, Zhu H, Blackshaw S, Shih Ie M,Wang TL, 2014, Notch3 interactome analysis identifiedWWP2 as a negative regulator of Notch3 signaling in ovarian cancer. PLoS Genet 10:e1004751 Maddika S, Kavela S, Rani N, Palicharla VR, Pokorny JL, Sarkaria JN, Chen J, 2011, WWP2 is an E3 ubiquitin ligase for PTEN. Nat Cell Biol 13:728–733 Qin Y, Xu SQ, Pan DB, Ye GX, Wu CJ, Wang S, Wang CJ, Jiang JY, Fu J, 2016, Silencing ofWWP2 inhibits adhesion, invasion, and migration in liver cancer cells. Tumour Biol 37:6787–6799 Yu Z, Li T,Wang C,Deng S, Zhang B, HuoX,WangX, ZhongY, Ma X, 2016, Gamabufotalin triggers c-Myc degradation via induction of WWP2 in multiple myeloma cells. Oncotarget 7:15725–15737 Ding ZY, Huang YJ, Tang JD, Li G, Jiang PQ, Wu HT, 2016, Silencing of hypoxia-inducible factor-1alpha promotes thyroid cancer cell apoptosis and inhibits invasion by downregulatingWWP2, WWP9, VEGF and VEGFR2. Exp Ther Med 12:3735–3741 Wiesner S, Ogunjimi AA,Wang HR, Rotin D, Sicheri F,Wrana JL, Forman-Kay JD, 2007, Autoinhibition of the HECT-type ubiquitin ligase Smurf2 through its C2 domain. Cell 130:651–662 Liu J,Wan L, Yuan Z, Zhang J, Guo J, Malumbres M, ZouW,Wei W, 2016, Cdh1 inhibits WWP2-mediated ubiquitination of PTEN to suppress tumorigenesis in an APC-independent manner. Cell discov 2:15044 Mund T, Graeb M, Mieszczanek J, Gammons M, Pelham HR, Bienz M, 2015, Disinhibition of the HECT E3 ubiquitin ligase WWP2 by polymerized Dishevelled. Open Biol 5:150185 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 443 EP 446 DI 10.1007/s12253-018-0506-5 PG 4 ER PT J AU Shree, H Ramani, P Sherlin, H Sukumaran, G Jeyaraj, G Don, K Santhanam, A Ramasubramanian, A Sundar, R AF Shree, K. Hema Ramani, Pratibha Sherlin, Herald Sukumaran, Gheena Jeyaraj, Gifrrina Don, R. Kanchi Santhanam, Archana Ramasubramanian, Abilasha Sundar, R TI Saliva as a Diagnostic Tool in Oral Squamous Cell Carcinoma – a Systematic Review with Meta Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE MMP-9; Chemerin; Saliva; Markers ID MMP-9; Chemerin; Saliva; Markers AB Whole saliva is mainly composed of fluid produced by major and minor salivary glands. Major salivary glands including parotid, submandibular, and sublingual glands, are known to secrete fluid transported from serum as well as surrounding glandular tissues [1]. Beside the secretions from salivary glands, oral mucosa, periodontium, as well as oral microflora also contribute to the final content of whole saliva [1]. Whole saliva therefore represents a complex balance among local and systemic sources [2]. This allows for the application of saliva in the diagnosis not only for salivary gland disorders but also for oral diseases and systemic conditions [2]. The role of saliva as a diagnostic tool in detecting Oral Squamous Cell Carcinoma. Articles published in PUBMED, EMBASE, COCHRANE, GOOGLE, manual search and back references of the articles for last 5 years extracted 77 articles. Studies which considered saliva as a diagnostic tool were included. Statistical analysis with Receivers Operating Curve to establish sensitivity and specificity of the salivary biomarkers as a diagnostic tool to detect Oral Squamous Cell Carcinoma were included for meta analysis. The measure of effect with 95% confidence interval were meta analysed for 9 articles in which 308 healthy individuals compared with 340 patients with Oral Squamous Cell Carcinoma. Highly sensitive salivary biomarkers for detecting Oral Squamous Cell Carcinoma were MMP-9, Chemerin, Choline + Betaine + Pipecolinic Acid + I – Carnitine(confidence interval ranges from 0.83–1.0). The narrow confidence interval of 0.95 + (0.88–1.00) was seen for MMP-9 followed by 1.00 + (0.78–1.00) for chemerin. Highly specific biomarkers for Oral Squamous Cell Carcinoma were MMP-9 (specificity −100%,), Chemerin(specificity-100%), over expressed mi RNA 136 with specificity of 0.88(0.69–0.97), under expressed mi RNA 27B with specificity of 1.0(0.66–1.00). Saliva can be used as a diagnostic tool with highly sensitive and specific markers namely MMP-9, Chemerin for early detection of Oral Squamous Cell Carcinoma. C1 [Shree, K. Hema] Saveetha Dental College, 162, Masilamani Nagar, Seneerkuppam bypass Road, Poonamallee, 600077 Chennai, Tamil Nadu, India. [Ramani, Pratibha] Saveetha Dental College, 162, Masilamani Nagar, Seneerkuppam bypass Road, Poonamallee, 600077 Chennai, Tamil Nadu, India. [Sherlin, Herald] Saveetha Dental College, 162, Masilamani Nagar, Seneerkuppam bypass Road, Poonamallee, 600077 Chennai, Tamil Nadu, India. [Sukumaran, Gheena] Saveetha Dental College, 162, Masilamani Nagar, Seneerkuppam bypass Road, Poonamallee, 600077 Chennai, Tamil Nadu, India. [Jeyaraj, Gifrrina] Saveetha Dental College, 162, Masilamani Nagar, Seneerkuppam bypass Road, Poonamallee, 600077 Chennai, Tamil Nadu, India. [Don, R. Kanchi] Saveetha Dental College, 162, Masilamani Nagar, Seneerkuppam bypass Road, Poonamallee, 600077 Chennai, Tamil Nadu, India. [Santhanam, Archana] Saveetha Dental College, 162, Masilamani Nagar, Seneerkuppam bypass Road, Poonamallee, 600077 Chennai, Tamil Nadu, India. [Ramasubramanian, Abilasha] Saveetha Dental College, 162, Masilamani Nagar, Seneerkuppam bypass Road, Poonamallee, 600077 Chennai, Tamil Nadu, India. [Sundar, R] Jamals Grandeur, Kauveri Nagar Main Road, Velappanchavadi, 600077 Chennai, Tamil Nadu, India. RP Shree, H (reprint author), Saveetha Dental College, 600077 Chennai, India. EM hemashree9111990@gmail.com CR Caporossi L, Santoro A, Papaleo B, 2010 Sep, Saliva as an analytical matrix: state of the art and application for biomonitoring. Biomark Biochem Indic Expo Response Susceptibility Chem 15(6):475–487 Good DM, Thongboonkerd V, Novak J, Bascands J-L, Schanstra JP, Coon JJ, Dominiczak A, Mischak H, 2007 Dec, Body fluid proteomics for biomarker discovery: lessons from the past hold the key to success in the future. J Proteome Res 6(12):4549–4555 jpsr05101306.pdf [Internet]. [cited 2017 Dec 30]. Available from: http://www.jpsr.pharmainfo.in/Documents/Volumes/vol5issue10/ jpsr05101306.pdf Malamud D, Rodriguez-Chavez IR, 2011 Jan, Saliva as a Diagnostic Fluid. Dent Clin N Am 55(1):159–178 Radhika T, Jeddy N, Nithya S, Muthumeenakshi RM, 2016 Nov, Salivary biomarkers in oral squamous cell carcinoma – an insight. J Oral Biol Craniofacial Res 6(Suppl 1):S51–S54 Gualtero DF, Castillo AS, 2016 Apr 2, Biomarkers in saliva for the detection of oral squamous cell carcinoma and their potential use for early diagnosis: a systematic review. Acta Odontol Scand 74(3): 170–177 Lee LT, Wong YK, Hsiao HY, Wang YW, Chan MY, Chang KW, 2017 Nov 27, Evaluation of saliva and plasma cytokine biomarkers in patients with oral squamous cell carcinoma. Int J Oral Maxillofac Surg Ghallab NA, Shaker OG, 2017 Apr, Serum and salivary levels of chemerin and MMP-9 in oral squamous cell carcinoma and oral premalignant lesions. Clin Oral Investig 21(3):937–947 Malhotra R, Urs AB, Chakravarti A, Kumar S, Gupta VK, Mahajan B, 2016 Jul, Correlation of Cyfra 21-1 levels in saliva and serum with CK19 mRNA expression in oral squamous cell carcinoma. Tumour Biol J Int Soc Oncodevelopmental Biol Med 37(7):9263– 9271 Zahran F, Ghalwash D, Shaker O, Al-Johani K, Scully C, 2015 Sep, Salivary microRNAs in oral cancer. Oral Dis 21(6):739–747 Rajkumar K, Nandhini G, Ramya R, Rajashree P, Kumar AR, Anandan SN, 2014 Sep, Validation of the diagnostic utility of salivary interleukin 8 in the differentiation of potentially malignant oral lesions and oral squamous cell carcinoma in a region with high endemicity. Oral Surg Oral Med Oral Pathol Oral Radiol 118(3): 309–319 Wang Q, Gao P, Cheng F, Wang X, Duan Y, 2014 Feb, Measurement of salivary metabolite biomarkers for early monitoring of oral cancer with ultra performance liquid chromatographymass spectrometry. Talanta 119:299–305 Momen-Heravi F, Trachtenberg AJ, Kuo WP, Cheng YS, 2014 Jul, Genomewide study of salivary MicroRNAs for detection of Oral Cancer. J Dent Res 93(7 Suppl):86S–93S Rajkumar K, Ramya R, Nandhini G, Rajashree P, Ramesh Kumar A, Nirmala Anandan S, 2015 Jan, Salivary and serum level of CYFRA 21-1 in oral precancer and oral squamous cell carcinoma. Oral Dis 21(1):90–96 Wang Q, Gao P, Wang X, Duan Y, 2014 Jan 1, Investigation and identification of potential biomarkers in human saliva for the early diagnosis of oral squamous cell carcinoma. Clin Chim Acta Int J Clin Chem 427:79–85 Castagnola M, Scarano E, Passali GC, Messana I, Cabras T, Iavarone F, di Cintio G, Fiorita A, de Corso E, Paludetti G, 2017 Apr, Salivary biomarkers and proteomics: future diagnostic and clinical utilities. Acta Otorhinolaryngol Ital 37(2):94–101 Al-Tarawneh SK, Border MB, Dibble CF, Bencharit S, 2011 Jun, Defining salivary biomarkers using mass spectrometry-based proteomics: a systematic review. OMICS J Integr Biol 15(6):353–361 Wang N, Wang Q-J, Feng Y, Shang W, Cai M, 2013 Jul 19, Overexpression of chemerin was associated with tumor angiogenesis and poor clinical outcome in squamous cell carcinoma of the oral tongue. Clin Oral Investig 18 Vilen S-T, Salo T, Sorsa T, Nyberg P. Fluctuating Roles of Matrix Metalloproteinase-9 inOral Squamous Cell Carcinoma. SciWorld J [Internet]. 2013 Jan 8 [cited 2018 Jan 5];2013. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3556887/ Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011 Apr, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Manikandan M, Deva Magendhra Rao AK, Arunkumar G, ManickavasagamM, Rajkumar KS, Rajaraman R, et al. Oral squamous cell carcinoma: microRNA expression profiling and integrative analyses for elucidation of tumourigenesis mechanism. Mol Cancer [Internet]. 2016 Apr 7 [cited 2018 Jan 5];15. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4823852/ Londin E, Loher P, Telonis AG, Quann K, Clark P, Jing Y et al, 2015 Mar 10, Analysis of 13 cell types reveals evidence for the expression of numerous novel primate- and tissue-specific microRNAs. Proc Natl Acad Sci U S A 112(10):E1106–E1115 Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M, Croce CM, 2004 Mar 2, Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A 101(9):2999–3004 Wei J, Xie G, Zhou Z, Shi P, Qiu Y, Zheng X, Chen T, Su M, Zhao A, JiaW(2011 Nov 1, Salivarymetabolite signatures of oral cancer and leukoplakia. Int J Cancer 129(9):2207–2217 Tanaka T, Narazaki M, Kishimoto T. IL-6 in Inflammation, Immunity, and Disease. Cold Spring Harb Perspect Biol [Internet]. 2014 Oct [cited 2018 Jan 4];6(10). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4176007/ Baracos VE,MackenzieML, 2006 Jan, Investigations of branchedchain amino acids and their metabolites in animal models of cancer. J Nutr 136(1 Suppl):237S–242S Jinno T, Kawano S, Maruse Y, Matsubara R, Goto Y, Sakamoto T, Hashiguchi Y, Kaneko N, Tanaka H, Kitamura R, Toyoshima T, Jinno A, Moriyama M, Oobu K, Kiyoshima T, Nakamura S, 2015 May, Increased expression of interleukin-6 predicts poor response to chemoradiotherapy and unfavorable prognosis in oral squamous cell carcinoma. Oncol Rep 33(5):2161–2168 BickelM(1993 May, The role of interleukin-8 in inflammation and mechanisms of regulation. J Periodontol 64(5 Suppl):456–460 Interleukin 8 - an overview | ScienceDirect Topics [Internet]. [cited 2018 Jan 4]. Available from: https://www.sciencedirect.com/topics/ neuroscience/interleukin-8 Lopez-Castejon G, Brough D, 2011 Aug, Understanding the mechanism of IL-1β secretion. Cytokine Growth Factor Rev 22(4):189– 195 IL1B - Interleukin-1 beta precursor - Homo sapiens, Human, - IL1B gene & protein [Internet]. [cited 2018 Jan 4]. Available from: http://www.uniprot.org/uniprot/P01584 Nakano Y, Kobayashi W, Sugai S, Kimura H, Yagihashil S, 1999 Aug 1, Expression of tumor necrosis factor-α and Interleukin-6 in Oral squamous cell carcinoma. Jpn J Cancer Res 90(8):858–866 Niemann AM, Goeroegh T, Gottschlich S, Lippert BM,Werner JA, 1997 Aug, Cut-off value determination of CYFRA 21-1 for squamous cell carcinomas of the head and neck, SCCHN). Anticancer Res 17(4B):2859–2860 Hsu Y-P, Hsieh C-H, Chien H-T, Lai C-H, Tsao C-K, Liao C-T, et al. Serum markers of CYFRA 21-1 and C-reactive proteins in oral squamous cell carcinoma.World J Surg Oncol [Internet]. 2015 Aug 21 [cited 2018 Jan 5];13. Available from: https://www.ncbi. nlm.nih.gov/pmc/articles/PMC4546149/ NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 447 EP 453 DI 10.1007/s12253-019-00588-2 PG 7 ER PT J AU Tan, J Jin, X Wang, K AF Tan, Jiufeng Jin, Xuefei Wang, Kaichen TI Integrated Bioinformatics Analysis of Potential Biomarkers for Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Differentially expressed genes; Methylation; Network ID Prostate cancer; Differentially expressed genes; Methylation; Network AB The aim was to expound the pathogenesis of prostate cancer and to identify the potentially biomarkers for prostate cancer (PC). DNA methylation microarray data GSE38240 containing 8 prostate cancer metastases and 4 normal prostate samples as well as gene expression profile data GSE26910 containing 6 prostate primary tumors and 6 normal samples were used. Differentially expressed genes (DEGs) and differently methylated sites of PC were screened and the regulatory network was constructed with DEGs-related transcription factors (TFs). The obtained hub genes were subjected to proteinprotein interaction network analysis. Enrichment analysis of down-regulated DEGs were performed. Total 351 DEGs including 190 down-regulated and 161 upregulated genes and 3234 differently methylated sites were identified. In total 69 DEGs-related TFs were found. Regulatory network contained 1301 nodes and 2527 connection pairs and that FOXA1 (forkhead box A1), BZRAP1-AS1 (benzodiazapine receptor associated protein 1 antisense RNA 1) and KRT8 (keratin 8) were the top three nodes of it. The enriched GO terms were mainly biological activity of the blood and cells-related. Total 29 DEGs (such as AGTR1, angiotensin II receptor, type 1) and 57 none-DEGs involved in the PPI network. Biological functions in blood circulation and the involved AGTR1 may play important roles in PC by gene-methylation. Besides, BZRAP1-AS1 may be novel biomarker related with PC. C1 [Tan, Jiufeng] China-Japan Union Hospital of Jilin University, Department of urology, 126 Xiantai ST, 130033 Changchun, Jilin Province, China. [Jin, Xuefei] China-Japan Union Hospital of Jilin University, Department of urology, 126 Xiantai ST, 130033 Changchun, Jilin Province, China. [Wang, Kaichen] China-Japan Union Hospital of Jilin University, Department of urology, 126 Xiantai ST, 130033 Changchun, Jilin Province, China. RP Wang, K (reprint author), China-Japan Union Hospital of Jilin University, Department of urology, 130033 Changchun, China. EM fyxehei@163.com CR Aryee MJ, Liu W, Engelmann JC, Nuhn P, Gurel M, Haffner MC, Esopi D, Irizarry RA, Getzenberg RH, Nelson WG, Luo J, Xu J, Isaacs WB, Bova GS, Yegnasubramanian S, 2013, DNA methylation alterations exhibit intraindividual stability and interindividual heterogeneity in prostate cancer metastases. Sci TranslMed 5(169): 169ra110. , DOI 10.1126/scitranslmed.3005211 Bonci D, Coppola V, Musumeci M, Addario A, Giuffrida R, Memeo L, D'Urso L, Pagliuca A, Biffoni M, Labbaye C, 2008, The miR-15a–miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. NatMed 14(11):1271–1277 Currie D, McKnight A, Patterson C, Sadlier D, Maxwell A, 2010, Investigation of ACE, ACE2 and AGTR1 genes for association with nephropathy in Type 1 diabetes mellitus. Diabet Med 27(10): 1188–1194 Das PM, Singal R, 2004, DNA methylation and cancer. J Clin Oncol 22(22):4632–4642 Dedeurwaerder S, Defrance M, Calonne E, Denis H, Sotiriou C, Fuks F, 2011, Evaluation of the Infinium Methylation 450K technology. Epigenomics 3(6):771–784. , DOI 10.2217/epi.11. 105 Elbaz N, Bedecs K, Masson M, Sutren M, Strosberg AD, Nahmias C, 2000, Functional trans-inactivation of insulin receptor kinase by growth-inhibitory angiotensin II AT2 receptor. Mol Endocrinol 14(6):795–804 Gakhar G, YuLee G, Seandel M, Bander N, Nanus D, 2013, Prostate circulating tumor cells metastasize to bone via E-selectin expressed on endothelial cells. Can Res 73(8 Supplement):5130 Gautier L, Cope L, Bolstad BM, Irizarry RA, 2004, affy–analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 20(3):307–315. , DOI 10.1093/bioinformatics/btg405 Karolchik D, Barber GP, Casper J, Clawson H, Cline MS, Diekhans M, Dreszer TR, Fujita PA, Guruvadoo L, Haeussler M, Harte RA, Heitner S, Hinrichs AS, Learned K, Lee BT, Li CH, Raney BJ, Rhead B, Rosenbloom KR, Sloan CA, Speir ML, Zweig AS, Haussler D, Kuhn RM, Kent WJ, 2014, The UCSC Genome Browser database: 2014 update. Nucleic Acids Res 42(Database issue):D764–D770. , DOI 10.1093/nar/gkt1168 Kwabi-Addo B, Wang S, Furbert-Harris P, Yegnasubramanian S, Devaney J, 2013, Functional characterization of Basonuclin 1, BNC1): a novel tumor suppressor gene commonly downregulated in human prostate cancer. Can Res 73(8 Supplement):1974 Lin RJ, XiaoDW, Liao LD, Chen T, Xie ZF, HuangWZ,WangWS, Jiang TF, Wu BL, Li EM, 2012, MiR-142-3p as a potential prognostic biomarker for esophageal squamous cell carcinoma. J Surg Oncol 105(2):175–182 Lin Q, Tan HT, Lim HS, Chung MC, 2013, Sieving through the cancer secretome. Biochim Biophys Acta 1834(11):2360–2371. , DOI 10.1016/j.bbapap.2013.01.030 Logothetis CJ, Gallick GE, Maity SN, Kim J, Aparicio A, Efstathiou E, Lin S-H, 2013, Molecular classification of prostate cancer progression: foundation for marker-driven treatment of prostate cancer. Cancer Discov 3(8):849–861 Mitchell C, Zakar T, Sykes S, Pringle K, Lumbers E, 2010, 141. Methylation of genes of the renin angiotensin system, ras, in early human amnion. Reprod Fertil Dev 22(9):59–59 Modarresi F, Faghihi MA, Lopez-Toledano MA, Fatemi RP, Magistri M, Brothers SP, van der Brug MP, Wahlestedt C, 2012, Inhibition of natural antisense transcripts in vivo results in genespecific transcriptional upregulation. Nat Biotechnol 30(5):453– 459 Oh S, Shin S, Lightfoot SA, Janknecht R, 2013, 14-3-3 proteins modulate the ETS transcription factor ETV1 in prostate cancer. Can Res 73(16):5110–5119 Planche A, Bacac M, Provero P, Fusco C, Delorenzi M, Stehle JC, Stamenkovic I, 2011, Identification of prognostic molecular features in the reactive stroma of human breast and prostate cancer. PLoS One 6(5):e18640. , DOI 10.1371/journal.pone. 0018640 Prasad NB, Somervell H, Tufano RP, Dackiw AP, Marohn MR, Califano JA, Wang Y, Westra WH, Clark DP, Umbricht CB, 2008, Identification of genes differentially expressed in benign versus malignant thyroid tumors. Clin Cancer Res 14(11):3327– 3337 Rhodes DR, Ateeq B, Cao Q, Tomlins SA, Mehra R, Laxman B, Kalyana-Sundaram S, Lonigro RJ, Helgeson BE, Bhojani MS, 2009, AGTR1 overexpression defines a subset of breast cancer and confers sensitivity to losartan, an AGTR1 antagonist. Proc Natl Acad Sci U S A 106(25):10284–10289 Richiardi L, Fiano V, Grasso C, Zugna D, Delsedime L, Gillio-Tos A, Merletti F, 2013, Methylation of APC and GSTP1 in nonneoplastic tissue adjacent to prostate tumour and mortality from prostate cancer. PLoS One 8(7):e68162 Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T, 2003, Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13(11):2498–2504. , DOI 10.1101/gr. 1239303 Sherman BT, Huang d W, Tan Q, Guo Y, Bour S, Liu D, Stephens R, Baseler MW, Lane HC, Lempicki RA, 2007, DAVID Knowledgebase: a gene-centered database integrating heterogeneous gene annotation resources to facilitate high-throughput gene functional analysis. BMC Bioinf. 8:426. , DOI 10.1186/ 1471-2105-8-426 Skarn M, Baroy T, Stratford EW, Myklebost O, 2013, Epigenetic Regulation and Functional Characterization of MicroRNA-142 in Mesenchymal Cells. PLoS One 8(11):e79231 Szklarczyk D, Franceschini A, Kuhn M, Simonovic M, Roth A, Minguez P, Doerks T, Stark M, Muller J, Bork P, 2011, The STRING database in 2011: functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res 39(suppl 1):D561–D568 Tjensvoll K, Nordgard O, Smaaland R, 2014, Circulating tumor cells in pancreatic cancer patients:methods of detection and clinical implications. Int J Cancer 134(1):1–8. , DOI 10.1002/ijc. 28134 Verma A, Facchina SL, Hirsch DJ, Song S-Y, Dillahey LF, Williams JR, Snyder SH, 1998, Photodynamic tumor therapy: mitochondrial benzodiazepine receptors as a therapeutic target. Mol Med 4(1):40 ViolaMV, Fromowitz F, Oravez S, Deb S, Finkel G, Lundy J, Hand P, Thor A, Schlom J, 1986, Expression of ras oncogene p21 in prostate cancer. N Engl J Med 314(3):133–137 Voeller H, Sugars L, Pretlow T, Gelmann E, 1994, p53 oncogene mutations in human prostate cancer specimens. J Urol 151(2):492– 495 Wang F,Wang X-S, Yang G-H, Zhai P-F, Xiao Z, Xia L-Y, Chen LR, Wang Y, Wang X-Z, Bi L-X, 2012, miR-29a and miR-142-3p downregulation and diagnostic implication in human acute myeloid leukemia. Mol Biol Rep 39(3):2713–2722 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 455 EP 460 DI 10.1007/s12253-017-0346-8 PG 6 ER PT J AU Laas, E Ballester, M Cortez, A Graesslin, O Darai, E AF Laas, Enora Ballester, Marcos Cortez, Annie Graesslin, Olivier Darai, Emile TI Unsupervised Clustering of Immunohistochemical Markers to Define High-Risk Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial cancer; High-risk endometrial cancer; Immunohistochemistry; Unsupervised clustering; Principal component analysis ID Endometrial cancer; High-risk endometrial cancer; Immunohistochemistry; Unsupervised clustering; Principal component analysis AB Considerable heterogeneity exists in outcomes of early endometrial cancer (EC) according to the type but also the histological grading. Our goal was to describe the immunohistochemical profiles of type I EC according to grades and type II EC, to identify groups of interacting proteins using principal component analysis (PCA) and unsupervised clustering. We studied 13 immunohistochemical markers (steroid receptors, pro/anti-apoptotic proteins,metalloproteinases (MMP) and tissue inhibitor of metalloproteinase (TIMP), and CD44 isoforms known for their role in endometrial pathology. Co-expressed proteins associated with the type, grade and outcome of EC were determined by PCA and unsupervised clustering. PCA identified three functional groups of proteins from 43 tissue samples (38 type I and 5 type II EC): the first was characterized by p53 expression; the second by MMPs, bcl-2, PR B and CD44v6; and the third by ER alpha, PR A, TIMP-2 and CD44v3. Unsupervised clustering found two main clusters of proteins, with both type I grade 3 and type II EC exhibiting the same cluster profile. PCA and unsupervised clustering of immunohistochemical markers in EC contribute to a better comprehension and classification of the disease. C1 [Laas, Enora] AP-HP, Hopital Tenon, Service de Gynecologie-Obstetrique, 4 rue de la Chine, 75020 Paris, France. [Ballester, Marcos] AP-HP, Hopital Tenon, Service de Gynecologie-Obstetrique, 4 rue de la Chine, 75020 Paris, France. [Cortez, Annie] Universite Pierre et Marie Curie, Institut Universitaire de CancerologieParis, France. [Graesslin, Olivier] CHU de Reims, Hopital Alix de Champagne, Service de gynecologie-obstetrique, 45 rue Cognacq-Jay, 51100 Reims, France. [Darai, Emile] AP-HP, Hopital Tenon, Service de Gynecologie-Obstetrique, 4 rue de la Chine, 75020 Paris, France. RP Laas, E (reprint author), AP-HP, Hopital Tenon, Service de Gynecologie-Obstetrique, 75020 Paris, France. EM enora.laas@curie.fr CR Prat J, 2004, Prognostic parameters of endometrial carcinoma. Hum Pathol 35:649–662 Pansare V, Munkarah AR, Schimp Vet al, 2007, Increased expression of hypoxia-inducible factor 1alpha in type I and type II endometrial carcinomas. Mod Pathol 20:35–43. , DOI 10.1038/ modpathol.3800718 Llobet D, Pallares J, Yeramian A et al, 2009, Molecular pathology of endometrial carcinoma: practical aspects from the diagnostic and therapeutic viewpoints. J Clin Pathol 62:777–785. , DOI 10.1136/jcp.2008.056101 Lax SF, 2004, Molecular genetic pathways in various types of endometrial carcinoma: from a phenotypical to a molecular-based classification. Virchows Arch 444:213–223. , DOI 10. 1007/s00428-003-0947-3 Colombo N, Preti E, Landoni F et al, 2011, Endometrial cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow- up. Ann Oncol 22(Suppl 6):vi35–vi39. , DOI 10. 1093/annonc/mdr374 BallesterM, Dubernard G, Lecuru F et al, 2011, Detection rate and diagnostic accuracy of sentinel-node biopsy in early stage endometrial cancer: a prospective multicentre study, SENTI-ENDO). Lancet Oncol 12:469–476. , DOI 10.1016/S1470- 2045(11)70070-5 Nugent EK, Bishop EA, Mathews CA et al, 2012, Do uterine risk factors or lymph node metastasis more significantly affect recurrence in patients with endometrioid adenocarcinoma? Gynecol Oncol 125:94–98. , DOI 10.1016/j.ygyno.2011.11.049 Voss MA, Ganesan R, Ludeman L et al, 2012, Should grade 3 endometrioid endometrial carcinoma be considered a type 2 cancer-a clinical and pathological evaluation. Gynecol Oncol 124: 15–20. , DOI 10.1016/j.ygyno.2011.07.030 Alvarez T, Miller E, Duska L, Oliva E, 2012, Molecular profile of grade 3 endometrioid endometrial carcinoma: is it a type I or type II endometrial carcinoma? Am J Surg Pathol 36:753–761. https://doi. org/10.1097/PAS.0b013e318247b7bb Zannoni GF, Vellone VG, Arena V et al, 2010, Does high-grade endometrioid carcinoma, grade 3 FIGO, belong to type I or type II endometrial cancer? A clinical–pathological and immunohistochemical study. Virchows Arch 457:27–34. , DOI 10. 1007/s00428-010-0939-z Poncelet C,Walker F,Madelenat P et al, 2001, Expression of CD44 standard and isoforms V3 and V6 in uterine smooth muscle tumors: a possible diagnostic tool for the diagnosis of leiomyosarcoma. Hum Pathol 32:1190–1196 Laas E, Ballester M, Cortez A et al, 2014, Supervised clustering of immunohistochemical markers to distinguish atypical and nonatypical endometrial hyperplasia. Gynecol Endocrinol:1–4. https:// doi.org/10.3109/09513590.2014.989981 Laas E, Ballester M, Cortez A et al, 2014, Supervised clustering of immunohistochemical markers to distinguish atypical endometrial hyperplasia from grade 1 endometrial cancer. Gynecol Oncol. , DOI 10.1016/j.ygyno.2014.02.018 Zhang G, Wu L, Li B et al, 2013, Retrospective analysis of prognostic variables and clinical outcomes in surgically staged intermediate risk endometrial carcinoma. Eur J Obstet Gynecol Reprod Biol 169:309–316. , DOI 10.1016/j.ejogrb.2013.02.025 Fukuda K, Mori M, Uchiyama M et al, 1998, Prognostic significance of progesterone receptor immunohistochemistry in endometrial carcinoma. Gynecol Oncol 69:220–225. , DOI 10. 1006/gyno.1998.5023 Singh P, Smith CL, Cheetham G et al, 2008, Serous carcinoma of the uterus-determination of HER-2/neu status using immunohistochemistry, chromogenic in situ hybridization, and quantitative polymerase chain reaction techniques: its significance and clinical correlation. Int J Gynecol Cancer 18:1344–1351. , DOI 10.1111/j.1525-1438.2007.01181.x Giangrande PH,McDonnell DP, 1999, The a and B isoforms of the human progesterone receptor: two functionally different transcription factors encoded by a single gene. Recent Prog Horm Res 54: 291–313 discussion 313-314 Wik E, Raeder MB, Krakstad C et al, 2013, Lack of estrogen receptor-α is associated with epithelial-Mesenchymal transition and PI3K alterations in endometrial carcinoma. Clin Cancer Res. , DOI 10.1158/1078-0432.CCR-12-3039 Jongen V, Briet J, de Jong R et al, 2009, Expression of estrogen receptor-alpha and -beta and progesterone receptor-a and -B in a large cohort of patients with endometrioid endometrial cancer. Gynecol Oncol 112:537–542. , DOI 10.1016/j.ygyno.2008.10.032 Lapi Nska-Szumczyk S, Supernat A, Majewska H et al, 2014, HER2-positive endometrial cancer subtype carries poor prognosis. Clin Transl Sci. , DOI 10.1111/cts.12207 Hanekamp EE, Gielen SC, Smid-Koopman E et al, 2003, Consequences of loss of progesterone receptor expression in development of invasive endometrial cancer. Clin Cancer Res 9:4190–4199 Saegusa M, Okayasu I, 2000, Changes in expression of estrogen receptors alpha and beta in relation to progesterone receptor and pS2 status in normal and malignant endometrium. Jpn J Cancer Res 91:510–518 Sho T, Hachisuga T, Nguyen TTet al, 2014, Expression of estrogen receptor-α as a prognostic factor in patients with uterine serous carcinoma. Int J Gynecol Cancer 24:102–106. , DOI 10. 1097/IGC.0000000000000029 Soslow RA, Shen PU, Chung MH et al, 2000, Cyclin D1 expression in high-grade endometrial carcinomas–association with histologic subtype. Int J Gynecol Pathol 19:329–334 Vang R, Whi t a k e r BP, Farhood AI et al, 2001, Immunohistochemical analysis of clear cell carcinoma of the gynecologic tract. Int J Gynecol Pathol 20:252–259 Ansink AC, Cross PA, Scorer P et al, 1997, The hormonal receptor status of uterine carcinosarcomas, mixed mullerian tumours): an immunohistochemical study. J Clin Pathol 50:328–331 Demopoulos RI, Mesia AF, Mittal K, Vamvakas E, 1999, Immunohistochemical comparison of uterine papillary serous and papillary endometrioid carcinoma: clues to pathogenesis. Int J Gynecol Pathol 18:233–237 Reid-Nicholson M, Iyengar P, Hummer AJ et al, 2006, Immunophenotypic diversity of endometrial adenocarcinomas: implications for differential diagnosis. Mod Pathol 19:1091–1100. , DOI 10.1038/modpathol.3800620 Tashiro H, Isacson C, Levine R et al, 1997, p53 gene mutations are common in uterine serous carcinoma and occur early in their pathogenesis. Am J Pathol 150:177–185 Matias-Guiu X, Davidson B, 2014, Prognostic biomarkers in endometrial and ovarian carcinoma. Virchows Arch 464:315–331. , DOI 10.1007/s00428-013-1509-y Alkushi A, Lim P, Coldman A et al, 2004, Interpretation of p53 immunoreactivity in endometrial carcinoma: establishing a clinically relevant cut-off level. Int J Gynecol Pathol 23:129–137 Dupont J, Wang X, Marshall DS et al, 2004, Wilms tumor gene, WT1, and p53 expression in endometrial carcinomas: a study of 130 cases using a tissue microarray. Gynecol Oncol 94:449–455. , DOI 10.1016/j.ygyno.2004.05.014 Mitselou A, Ioachim E, Kitsou E et al, 2003, Immunohistochemical study of apoptosis-related Bcl-2 protein and its correlation with proliferation indices, Ki67, PCNA), tumor suppressor genes, p53, pRb), the oncogene c-erbB-2, sex steroid hormone receptors and other clinicopathological features, in normal, hyperplastic and neoplastic endometrium. In Vivo 17:469–477 ZhangM, Zhang P, Zhang C et al, 2009, Prognostic significance of Bcl-2 and Bax protein expression in the patients with oral squamous cell carcinoma. J Oral Pathol Med 38:307–313. , DOI 10. 1111/j.1600-0714.2008.00689.x Miturski R, Semczuk A, Tomaszewski J, Jakowicki J, 1998, Bcl-2 protein expression in endometrial carcinoma: the lack of correlation with p53. Cancer Lett 133:63–69 Zhang R, He Y, Zhang X et al, 2012, Estrogen receptor-regulated microRNAs contribute to the BCL2/BAX imbalance in endometrial adenocarcinoma and precancerous lesions. Cancer Lett 314:155– 165. , DOI 10.1016/j.canlet.2011.09.027 ZhengW, Cao P, ZhengMet al, 1996, p53 overexpression and bcl- 2 persistence in endometrial carcinoma: comparison of papillary serous and endometrioid subtypes. Gynecol Oncol 61:167–174. , DOI 10.1006/gyno.1996.0120 Moriyama T, Littell RD, Debernardo R et al, 2004, BAG-1 expression in normal and neoplastic endometrium. Gynecol Oncol 94: 289–295. , DOI 10.1016/j.ygyno.2004.04.026 Cancer Genome Atlas Research Network, Kandoth C, Schultz N et al, 2013, Integrated genomic characterization of endometrial carcinoma. Nature 497:67–73. , DOI 10.1038/nature12113 Hussein YR, Weigelt B, Levine DA et al, 2014, Clinicopathological analysis of endometrial carcinomas harboring somatic POLE exonuclease domain mutations. Mod Pathol. , DOI 10.1038/ modpathol.2014.143 Uzan C, Cortez A, Dufournet C et al, 2004, Eutopic endometrium and peritoneal, ovarian and bowel endometriotic tissues express a different profile of matrix metalloproteinases-2, −3 and −11, and of tissue inhibitor metalloproteinases-1 and -2. Virchows Arch 445: 603–609. , DOI 10.1007/s00428-004-1117-y Graesslin O, Cortez A, Uzan C et al, 2006, Endometrial tumor invasiveness is related to metalloproteinase 2 and tissue inhibitor of metalloproteinase 2 expressions. Int J Gynecol Cancer 16:1911– 1917. , DOI 10.1111/j.1525-1438.2006.00717.x Ferguson SE, Olshen AB, Viale A et al, 2004, Gene expression profiling of tamoxifen-associated uterine cancers: evidence for two molecular classes of endometrial carcinoma. Gynecol Oncol 92:719–725. , DOI 10.1016/j.ygyno.2003.10.038 Moser PL, Kieback DG, Hefler L et al, 1999, Immunohistochemical detection of matrix metalloproteinases, MMP, 1 and 2, and tissue inhibitor of metalloproteinase 2, TIMP 2, in stage IB cervical cancer. Anticancer Res 19:4391–4393 Wang F-Q, So J, Reierstad S, Fishman DA, 2005, Matrilysin, MMP-7, promotes invasion of ovarian cancer cells by activation of progelatinase. Int J Cancer 114:19–31. , DOI 10.1002/ijc.20697 KatsuraM, Furumoto H, NishimuraMet al, 1998, Overexpression of CD44 variants 6 and 7 in human endometrial cancer. Gynecol Oncol 71:185–189. , DOI 10.1006/gyno.1998.5169 Tempfer C, Haeusler G, Kaider A et al, 1998, The prognostic value of CD44 isoform expression in endometrial cancer. Br J Cancer 77:1137 Ayhan A, Tok EC, Bildirici I,AyhanA(2001, Overexpression of CD44 variant 6 in human endometrial cancer and its prognostic significance. Gynecol Oncol 80:355–358. , DOI 10.1006/gyno.2000.6014 Hoshimoto K, Yamauchi N, Takazawa Yet al, 2003, CD44 variant 6 in endometrioid carcinoma of the uterus: its expression in the adenocarcinoma component is an independent prognostic marker. Pathol Res Pract 199:71–77 Gun BD, Bahadir B, Bektas S et al, 2012, Clinicopathological significance of fascin and CD44v6 expression in endometrioid carcinoma. Diagn Pathol 7:15 Yeung KY, Ruzzo WL, 2001, Principal component analysis for clustering gene expression data. Bioinformatics 17:763–774 Wang Y, Ma X, Xi C et al, 2013, Correlation between estrogen receptor status and clinicopathologic parameters in endometrial cancer: a comparative study by immunohistochemistry using different scoring systems. Zhonghua Bing Li Xue Za Zhi 42:509–514 Qureshi A, Pervez S, 2010, Allred scoring for ER reporting and it’s impact in clearly distinguishing ER negative from ER positive breast cancers. J Pak Med Assoc 60:350–353 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 461 EP 469 DI 10.1007/s12253-017-0335-y PG 9 ER PT J AU Trabzonlu, L Muezzinoglu, B Corakci, A AF Trabzonlu, Levent Muezzinoglu, Bahar Corakci, Aydin TI BCL-2 and PAX2 Expressions in EIN which Had Been Previously Diagnosed as Non-Atypical Hyperplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial intraepithelial neoplasia; PAX2; BCL-2; Immunohistochemistry; Uterus; Endometrial hyperplasia ID Endometrial intraepithelial neoplasia; PAX2; BCL-2; Immunohistochemistry; Uterus; Endometrial hyperplasia AB The relationship between PAX2 and another anti-apoptotic gene, BCL-2, has been shown in a limited number of studies. The aims of this study are to investigate the value of PAX2 and BCL-2 expressions in lesions which have been defined as nonatypical hyperplasia in terms of detecting EIN and to evaluate the relations of these proteins in EIN. For this purpose, 108 cases of nonatypical endometrial hyperplasia diagnosed from 2006 to 2011 were re-evaluated. Immunohistochemical studies with PAX2 and BCL-2 were performed in 20 cases with EIN and 34 cases with benign hyperplasia. The mean BCL-2 immunohistochemistry scores of benign hyperplasia and EIN cases were 4.06 ± 1.04 and 4.63 ± 2.03, respectively. The mean BCL-2 score of EIN cases was significantly higher than benign hyperplasia (p = 0.021). The mean PAX2 scores of benign hyperplasia and EIN cases were 4.32 ± 1.07 and 2.19 ± 2.34, respectively. The mean PAX2 scores of EIN cases were significantly lower than benign hyperplasia (p = 0.001). BCL-2 expression was increased compared to normal endometrium in 66.7% of EIN cases, and PAX2 expression was decreased in 73.3%. Consistent with this, in 60% of cases, BCL-2 expression was increased compared to normal endometrium, while PAX2 expression was decreased. BCL-2 and PAX2 protein expression changes occur in early phases of endometrial tumorigenesis. These changes are often seen as a simultaneous increase in BCL-2 expression and decrease in PAX2 expression. C1 [Trabzonlu, Levent] Johns Hopkins School of Medicine, Department of Pathology, 600 N Wolfe Street, 21287 Baltimore, MD, USA. [Muezzinoglu, Bahar] Kocaeli University School of Medicine, Department of PathologyKocaeli, Turkey. [Corakci, Aydin] Kocaeli University School of Medicine, Department of Gynecology and ObstetricsKocaeli, Turkey. RP Trabzonlu, L (reprint author), Johns Hopkins School of Medicine, Department of Pathology, 21287 Baltimore, USA. EM leventtrabzonlu186@gmail.com CR Kurman RJ, Kaminski PF, Norris HJ, 1985, The behavior of endometrial hyperplasia. A long-term study of Buntreated^ hyperplasia in 170 patients. Cancer 56:403–412 Mutter GL, Baak JP, Crum CP et al, 2000, Endometrial precancer diagnosis by histopathology, clonal analysis, and computerized morphometry. J Pathol 190:462–469. , DOI 10.1002/(SICI, 1096-9896(200003)190:4<462::AID-PATH590>3.0.CO;2-D Mutter GL, Chaponot ML, Fletcher JA, 1995, A polymerase chain reaction assay for non-random X chromosome inactivation identifies monoclonal endometrial cancers and precancers. Am J Pathol 146:501–508 Baak JP, MutterGL, Robboy SJ et al, 2005, Themolecular genetics and morphometry-based endometrial intraepithelial neoplasia classification system predicts disease progression in endometrial hyperplasia more accurately than the 1994 World Health Organization classification system. Cancer 103:2304–2312. , DOI 10. 1002/cncr.21058 Hecht JL, Mutter GL, 2006, Molecular and pathologic aspects of endometrial carcinogenesis. J Clin Oncol 24:4783–4791. https:// doi.org/10.1200/JCO.2006.06.7173 Tsujimoto Y, Finger LR, Yunis J et al, 1984, Cloning of the chromosome breakpoint of neoplastic B cells with the t, 14;18, chromosome translocation. Science 226:1097–1099 Kapucuogl u N, Aktepe F, Kaya H e t al, 200 7 , Immunohistochemical expression of PTEN in normal, hyperplastic and malignant endometrium and its correlation with hormone receptors, bcl-2, bax, and apoptotic index. Pathol Res Pract 203: 153–162. , DOI 10.1016/j.prp.2007.01.003 Kokawa K, Shikone T, Otani T, Nakano R, 1999, Apoptosis and the expression of Bax and Bcl-2 in squamous cell carcinoma and adenocarcinoma of the uterine cervix. Cancer 85:1799–1809. , DOI 10.1002/(SICI)1097-0142(19990415)85:8<1799:: AID-CNCR21>3.0.CO;2-M Vaskivuo TE, Stenback F, Tapanainen JS, 2002, Apoptosis and apoptosis-related factors Bcl-2, Bax, tumor necrosis factor-α, and NF-κB in human endometrial hyperplasia and carcinoma. Cancer 95:1463–1471. , DOI 10.1002/cncr.10876 Stuart ET, Gruss P, 1996, PAX: developmental control genes in cell growth and differentiation. Cell Growth Differ 7:405–412 Stuart ET, Haffner R, Oren M, Gruss P, 1995, Loss of p53 function through PAX-mediated transcriptional repression. EMBO J 14: 5638 Allison KH, Upson K, Reed SD et al, 2012, PAX2 loss by immunohistochemistry occurs early and often in endometrial hyperplasia. Int J Gynecol Pathol 31:159–167. , DOI 10.1097/PGP. 0b013e318226b376 Quick CM, Laury AR, Monte NM, Mutter GL, 2012, Utility of PAX2 as a marker for diagnosis of endometrial intraepithelial neoplasia. Am J Clin Pathol 138:678–684. , DOI 10.1309/ AJCP8OMLT7KDWLMF Winyard PJ, Risdon RA, Sams VR et al, 1996, The PAX2 transcription factor is expressed in cystic and hyperproliferative dysplastic epithelia in human kidney malformations. J Clin Invest 98: 451–459. , DOI 10.1172/JCI118811 Zhang LP, Shi XY, Zhao CYet al, 2011, RNA interference of pax2 inhibits growth of transplanted human endometrial cancer cells in nude mice. Chin J Cancer 30:400–406 Mutter GL, 2002, Diagnosis of premalignant endometrial disease. J Clin Pathol 55:326–331 Zaino RJ, Carinelli S, Ellenson LH, et al, 2014, Tumours of the uterine corpus: epithelial tumours and precursors. In: Kurman RJ, CarcangiuM, Herrington C,Young R, eds)WHO Classif. Tumours female Reprod. Organs, 4th ed. WHO Press, Lyon, pp 125–126 Hecht JL, Ince T, Baak JP et al, 2005, Prediction of endometrial carcinoma by subjective endometrial intraepithelial neoplasia diagnosis. Mod Pathol 18:324–330. , DOI 10.1038/modpathol. 3800328 Popat VC, Vora DN, Gadhvi NS et al, 2010, Comparison of endometrial intraepithelial neoplasiawithWHO endometrial hyperplasia classification system. A comparative study of 150 cases. Histopathology 57:646–648. , DOI 10.1111/j.1365-2559. 2010.03672.x Allred DC, Harvey JM, Berardo M, Clark GM, 1998, Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11:155–168 Bissonnette RP, Echeverri F, Mahboubi A, Green DR, 1992, Apoptotic cell death induced by c-myc is inhibited by bcl-2. Nature 359:552–554. , DOI 10.1038/359552a0 Hemann MT, Lowe SW, 2006, The p53–Bcl-2 connection. Cell Death Differ 13:1256–1259. , DOI 10.1038/sj.cdd. 4401962 Bozkurt KK, Yalcin Y, Erdemoglu E et al, 2016, The role of immunohistochemical adrenomedullin and Bcl-2 expression in development of type-1 endometrial adenocarcinoma Adrenomedullin expression in endometrium. Pathol Res Pract 212:450–455. https:// doi.org/10.1016/j.prp.2016.02.021 Amalinei C, Cianga C, Balan R et al, 2011, Immunohistochemical analysis of steroid receptors, proliferation markers, apoptosis related molecules, and gelatinases in non-neoplastic and neoplastic endometrium. Ann Anat - Anat Anzeiger 193:43–55. , DOI 10.1016/j.aanat.2010.09.009 Wu H, Chen Y, Liang J et al, 2005, Hypomethylation-linked activation of PAX2 mediates tamoxifen-stimulated endometrial carcinogenesis. Nature 438:981–987. , DOI 10.1038/ nature04225 Strissel PL, Ellmann S, Loprich E et al, 2008, Early aberrant insulin-like growth factor signaling in the progression to endometrial carcinoma is augmented by tamoxifen. Int J Cancer 123:2871– 2879. , DOI 10.1002/ijc.23900 Kahraman K, Kiremitci S, Taskin S et al, 2012, Expression pattern of PAX2 in hyperplastic and malignant endometrium. Arch Gynecol Obstet 286:173–178. , DOI 10.1007/s00404- 012-2236-3 Joiner AK, Quick CM, Jeffus SK, 2015, Pax2 expression in simultaneously diagnosed WHO and EIN classification systems. Int J Gynecol Pathol 34:40–46. , DOI 10.1097/pgp. 0000000000000185 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 471 EP 476 DI 10.1007/s12253-017-0378-0 PG 6 ER PT J AU Szekerczes, T Galamb, Kocsis, A Benczik, M Takacs, T Martonos, A Jaray, B Kiss, A Jeney, Cs Nyiri, M Schaff, Zs Sobel, G AF Szekerczes, Timea Galamb, Adam Kocsis, Adrienn Benczik, Marta Takacs, Tibor Martonos, Attila Jaray, Balazs Kiss, Andras Jeney, Csaba Nyiri, Miklos Schaff, Zsuzsa Sobel, Gabor TI Dual-Stained Cervical Cytology and Histology with Claudin-1 and Ki67 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; Claudin-1/Ki67 immunochemistry; p16/Ki67 reaction; Claudins ID Cervical cancer; Claudin-1/Ki67 immunochemistry; p16/Ki67 reaction; Claudins AB Several biomarkers are in use to improve the sensitivity and specificity of cervical cancer screening. Previously, increased expression of tight junction protein claudin-1 (CLDN1) was detected in premalignant and malignant cervical lesions and applied for cytology screening. To improve the specificity, a double immunoreaction with CLDN1/Ki67 was developed in the recent study. Parallel p16/Ki67 (CINtec® PLUS) and CLDN1/Ki67 dual-stained cytology and histology were performed and compared. p16/ Ki67 immunoreaction showed positivity in 317 out of 1596 smears with negativity in 1072 and unacceptable reactions in 207 samples. CLDN1/Ki67 dual staining was positive in 200 of 1358 samples, negative in 962, whereas 196 smears could not be evaluated due to technical reasons. Considering the high-grade squamous intraepithelial lesion cytology as gold standard, sensitivity of CLDN1/Ki67 reaction was 76%, specificity was 85.67%, while for p16/Ki67 sensitivity was 74% and specificity was 81.38%. Comparison of CLDN1/Ki67 and p16/Ki67 dual stainings showed the results of the two tests not to be significantly different. Analysing histological slides from 63 cases, the results of the two tests agreed perfectly. As conclusion the sensitivity and specificity proved to be similar using p16/Ki67 and CLDN1/Ki67 double immunoreactions both on LBC samples and on histological slides. C1 [Szekerczes, Timea] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Galamb, Adam] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Kocsis, Adrienn] CellCall LtdBudapest, Hungary. [Benczik, Marta] CellCall LtdBudapest, Hungary. [Takacs, Tibor] CellCall LtdBudapest, Hungary. [Martonos, Attila] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. [Jaray, Balazs] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Jeney, Csaba] CellCall LtdBudapest, Hungary. [Nyiri, Miklos] CellCall LtdBudapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Sobel, Gabor] Semmelweis University, 2nd Department of Obstetrics and GynecologyBudapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM schaff.zsuzsa@med.semmelweis-univ.hu CR Hillemans P, Soergel P, Hertel H, Jentschke M, 2016, Epidemiology and early detection of cervical cancer. Oncol Res Treat 39:1–6 Sawaya GF, Smith-McCune K, 2016, Cervical cancer screening. Obstet Gynecol 127:459–467 Lees BF, Erickson BK, Huh WK(2016, Cervical cancer screening: evidence behind the guidelines.AmJ Obstet Gynecol 214:438–443 Ocque R, Austin RM, 2016, Follow-up of women with negative pap test results and abnormal clinical signs or symptoms. Am J Clin Pathol 145:560–567 Ikenberg H, Bergeron C, Schmidt D, Griesser H, Alameda F, Angeloni C, Bogers J, Dachez R, Denton K, Hariri J, Keller T, von Knebel Doeberitz M, Neumann HH, Puig-Tintore LM, Sideri M, Rehm S, Ridder R, PALMS Study Group, 2013, Screening for cervical cancer precursors with p16/Ki-67 dual-stained cytology: results of the PALMS study. J Natl Cancer Inst 105:1550–1557 Wentzensen N, Fetterman B, Castle PE, Schiffman M, Wood SN, Stiemerling E, Tokugawa D, Bodelon C, Roitras N, Lorey T, Kinney W, 2015, p16/Ki-67 dual stain cytology for detection of cervical precancer in HPV-positive women. J Natl Cancer Inst 107: 1–8 Cuzick J, Clavel C, Petry KU, Meijer CJ, Hoyer H, Ratnam S, Szarewski A, Birembaut P, Kulasingam S, Sasieni P, Iftner T, 2006, Overview of the European and north American studies on HPV testing in primary cervical cancer screening. Int J Cancer 119: 1095–1101 PlummerM, Schiffman M, Castle PE,Maucort-Boulch D,Wheeler CM, ALTS Group, 2007, A 2-year prospective study of human papillomavirus persistence among women with a cytological diagnosis of atypical squamous cells of undetermined significance or low-grade squamous intraepithelial lesion. J Infect Dis 195:1582– 1589 Benczik M, Galamb A, Koiss R, Kovacs A, Jaray B, Szekely T, Szekerczes T, Schaff Z, Sobel G, Jeney C, 2016, Claudin-1 as a biomarker of cervical cytology and histology. Pathol Oncol Res 22: 179–188 Bergeron C, Ikenberg H, SideriM, Denton K, Bogers J, Schmidt D, Alameda F, Keller T, Rehm S, Ridder R, PALMS Study Group, 2015, Prospective evaluation of p16/Ki-67 dual-stained cytology for managing women with abnormal Papanicolaou cytology: PALMS study results. Cancer Cytopathol 123:373–381 Kocsis A, Takacs T, Jeney C, Schaff Z, Koiss R, Jaray B, Sobel G, Pap K, Szekely I, Ferenci T, Lai HC, Nyiri M, Benczik M, 2017, Performance of a new HPVand biomarker assay in management of hrHPV positive women: Subanalysis of the ongoing multicenter TRACE Clinical Trial, n>6,000, to evaluate POU4F3 as a potential biomarker of cervical precancer and cancer. Int J Cancer 140:1119– 1133 Sobel G, Paska C, Szabo I, Kiss A, Kadar A, Schaff Z, 2005, Increased expression of claudins in cervical squamous intraepithelial neoplasia and invasive carcinoma. Hum Pathol 36: 162–169 Szabo I, Kiss A, Schaff Z, Sobel G, 2009, Claudins as diagnostic and prognostic markers in gynecological cancer. Histol Histopathol 24:1607–1615 Lee JW, Lee SJ, Seo J, Song SY, Ahn G, Park CS, Lee JH, KimBG, Bae DS, 2005, Increased expressions of claudin-1 and claudin-7 during the progression of cervical neoplasia. Gynecol Oncol 97:53– 59 Lal-Nag M, Morin PJ, 2009, The claudins. Genome Biol 10:235 Nayar R, Wilbur DC, 2015, The pap test and Bethesda 2014. Cancer Cytopathol 123:271–281 Solomon D, 2015, Foreword. In: Nayar F, Wilbur DC, ed, The Bethesda system for reporting cervical cytology. Definitions, criteria, and explanatory notes, 3rd ed. Springer International Publishing Switzerland, Cham, pp v-vii Wentzensen N, von Knebel Doeberitz M, 2007, Biomarkers in cervical cancer screening. Dis Markers 23:315–330 Lee S, Rose MS, Sahasrabuddhe VV, Zhao R, Duggan MA, 2017, Tissue-based immunohistochemical biomarker accuracy in the diagnosis of malignant glandular lesions of the uterine cervix: a systematic review of the literature and meta-analysis. Int J Gynecol Pathol 36:310–322 Polman NJ, UijterwaalMH,Witte BI, Berkhof J, van Kemenade FJ, Spruijt JW, van Baal WM, Graziosi PG, van Dijken DK, Verheijen RH, Helmerhorst TJ, Steenbergen RD, Heideman DA, Ridder R, Snijders PJ, Meijer CJ, 2017, Good performance of p16/ki-67 dualstained cytology for surveillance of women treated for high-grade CIN. Int J Cancer 140:423–430 Yu LL, Guo HQ, Lei XQ, Qin Y, Wu ZN, Kang LN, Zhang X, Qiao YL, Chen W, 2016, p16/Ki-67 co-expression associates high risk human papillomavirus persistence and cervical histopathology: a 3-year cohort study in China. Oncotarget 7:64810– 64819 Possati-Resende JC, Fregnani JHTG, Kerr LM, Mauad EC, Longatto-Filho A, Scapulatempo-Neto C, 2015, The accuracy of p16/Ki67 and HPV test in the detection of CIN2/3 in women diagnosed with ASC-US or LSIL. PLoS One 10:e0134445 Furuse M, Fujita K, Hiiragi T, Fujimoto K, Tsukita S, 1998, Claudin-1 and -2: novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol 141:1539–1550 Morin PJ, 2005, Claudin proteins in human cancer: promising new targets for diagnosis and therapy. Cancer Res 65:9603–9606 Soini Y, 2005, Expression of claudins 1, 2, 3, 4, 5 and 7 in various types of tumours. Histopathology 46:551–560 Cunniffe C, Brankin B, Lambkin H, Ryan F, 2014, The role of claudin-1 and claudin-7 in cervical tumorigenesis. Anticancer Res 34:2851–2857 Akimoto T, Takasawa A, Murata M, Kojima Y, Takasawa K, Nojima M, Aoyama T, Hiratsuka Y, Ono Y, Tanaka S, Osanai M, Hasegawa T, Saito T, Sawada N, 2016, Analysis of the expression and localization of tight junction transmembrane proteins, claudin- 1, −4, −7, occludin and JAM-A, in human cervical adenocarcinoma. Histol Histopathol 31:921–931 Gyorffy H, Holczbauer A, Nagy P, Szabo Z, Kupcsulik P, Paska C, Papp J, Schaff Z, Kiss A, 2005, Claudin expression in Barrett’s esophagus and adenocarcinoma. Virchows Arch 447:961–968 Tokes AM, Kulka J, Paku S, Szik A, Paska C, Novak PK, Szilak L, Kiss A, Bogi K, Schaff Z, 2005, Claudin-1, −3 and −4 proteins and mRNA expression in benign and malignant breast lesions: a research study. Breast Cancer Res 7:R296–R305 Lodi C, Szabo E, Holczbauer A, Batmunkh E, Szijarto A, Kupcsulik P, Kovalszky I, Paku S, Illyes G, Kiss A, Schaff Z, 2006, Claudin-4 differentiates biliary tract cancers from hepatocellular carcinomas. Mod Pathol 19:460–469 Szasz AM, Nyirady P, MajorosA, Szendroi A, SzucsM, Szekely E, Tokes AM, Romics I, Kulka J, 2010, Beta-catenin expression and claudin expression pattern as prognostic factors of prostatic cancer progression. BJU Int 105:716–722 English DP, Santin AD, 2013, Claudins overexpression in ovarian cancer: potential targets for Clostridium Perfringens enterotoxin, CPE, based diagnosis and therapy. Int J Mol Sci 14: 10412–10437 Vazquez-Ortiz G, Ciudad CJ, Pina P, Vazquez K, Hidalgo A, Latorre B, Garcia JA, Salamanca F, Peralta-Rodriguez R, Rangel A, Salcedo M, 2005, Gene identification by cDNA arrays in HPVpositive cervical cancer. Arch Med Res 36:448–458 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 477 EP 486 DI 10.1007/s12253-018-0384-x PG 10 ER PT J AU Kallay, K Zakarias, D Csordas, K Benyo, G Kassa, Cs Sinko, J Strehn, A Horvath, O Vasarhelyi, B Krivan, G AF Kallay, Krisztian Zakarias, David Csordas, Katalin Benyo, Gabor Kassa, Csaba Sinko, Janos Strehn, Anita Horvath, Orsolya Vasarhelyi, Barna Krivan, Gergely TI Antithymocyte Globuline Therapy and Bradycardia in Children SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Antithymocyte globulin; ATG; Bradycardia; Children ID Antithymocyte globulin; ATG; Bradycardia; Children AB In antithymocyte globulin (ATG) treated patients occasionally bradycardia has been noticed. Therefore, we retrospectively analyzed the occurrence of bradycardia in ATG-treated children. Usingmedical records between 2007 and 2012 we identified children undergoing a combined therapy with ATG and glucocorticoids (ATG group, n = 22). The incidence of bradycardia was compared to that registered in children treated with glucocorticoids alone (glucocorticoid alone group, n = 21). Heart rates (HR) were registered before and on days 0–3, 4–7 and 8–14 after the ATG or steroid administration. The rate of bradycardic episodes was higher during ATG therapy than in the steroid alone group, while severe bradycardia occurred only in the ATG group (97 versus 32, p = 0.0037, and 13 versus 0, p = 0.0029, respectively). There was an interaction between the time and treatment group on HR (p = 0.046). Heart rates in ATG and steroid alone groups differed significantly on day 0–3 and day 4–7 (p = 0.046, p = 0.006, respectively).Within the ATG group HR was lower on days 4–7 compared to the days before and the days 8–14 values (p < 0.001, 95% CI: 0.020–0.074). These findings indicate that transient asymptomatic bradycardia is probably more common with ATG therapy than previously reported. HR should be closely monitored during and after ATG therapy. C1 [Kallay, Krisztian] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Albert Florian street 5-7, H-1097 Budapest, Hungary. [Zakarias, David] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Albert Florian street 5-7, H-1097 Budapest, Hungary. [Csordas, Katalin] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Albert Florian street 5-7, H-1097 Budapest, Hungary. [Benyo, Gabor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Albert Florian street 5-7, H-1097 Budapest, Hungary. [Kassa, Csaba] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Albert Florian street 5-7, H-1097 Budapest, Hungary. [Sinko, Janos] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Albert Florian street 5-7, H-1097 Budapest, Hungary. [Strehn, Anita] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Albert Florian street 5-7, H-1097 Budapest, Hungary. [Horvath, Orsolya] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Albert Florian street 5-7, H-1097 Budapest, Hungary. [Vasarhelyi, Barna] Semmelweis University, Department of Laboratory MedicineBudapest, Hungary. [Krivan, Gergely] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Albert Florian street 5-7, H-1097 Budapest, Hungary. RP Kallay, K (reprint author), Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, H-1097 Budapest, Hungary. EM dr.kallay@gmail.com CR George B, Mathews V, Viswabandya A, Lakshmi KM, Srivastava A, ChandyM(2010, Allogeneic hematopoietic stem cell transplantation is superior to immunosuppressive therapy in Indian children with aplastic anemia–a single-center analysis of 100 patients. Pediatr Hematol Oncol 27(2):122–131 Karapinar DY, Karadas N, Ay Y, Akin M, Balkan C, Aydinok Y, Kavakli K, 2014, Rabbit antithymocyte globulin treatment in childhood acquired severe aplastic anemia. Pediatr Hematol Oncol 31(1):20–28 Jiang S, Wang Y, Shi W, Shao Y, Qiao X, Lin J, Kuang H, Xie X, 2009, The benefit of ATG in immunosuppressive therapy of children with moderate aplastic anemia. Pediatr Hematol Oncol 26(5): 313–320 Deyell RJ, Shereck EB, Milner RA, Schultz KR, 2011, Immunosuppressive therapy without hematopoietic growth factor exposure in pediatric acquired aplastic anemia. Pediatr Hematol Oncol 28(6):469–478 Scheinberg P, Townsley D, Dumitriu B, Scheinberg P,Weinstein B, Rios O,Wu CO,Young NS, 2014, Horse antithymocyte globulin as salvage therapy after rabbit antithymocyte globulin for severe aplastic anemia. Am J Hematol 89(5):467–469 Scheinberg P, Nunez O, Weinstein B, Scheinberg P, Biancotto A, 2011)Wu CO, et al. horse versus rabbit antithymocyte globulin in acquired aplastic anemia. N Engl J Med 365(5):430–438 Godown J, Deal AM, Riley K, Bailliard F, Blatt J, 2011)Worsening bradycardia following antithymocyte globulin treatment of severe aplastic anemia. J Pediatr Pharmacol Ther 16(3):218–221 Tvede N, Nielsen LP, Andersen V, 1986, Bradycardia after highdose intravenous methylprednisolone therapy. Scand J Rheumatol 15(3):302–304 Taylor MR, Gaco D, 2013, Symptomatic sinus bradycardia after a treatment course of high-dose oral prednisone. The Journal of Emergency Medicine 45(3):e55–e58 Al Shibli A, Al Attrach I, HamdanMA, 2012, Bradycardia following oral corticosteroid use: case report and literature review. Arab Journal of Nephrology and Transplantation 5(1):47–49 Akikusa JD, Feldman BM, Gross GJ, Silverman ED, Schneider R, 2007, Sinus bradycardia after intravenous pulse methylprednisolone. Pediatrics 119(3):e778–e782 Michaelson ME, 1972, MA;. Congenital complete heart block: an international study of the natural history. In: Clinics C, ed, Brest AE, MA. FA Davis, Philadelphia, p 85 Kugler J, 1990, Sinus node dysfunction. In: Gilette PG, Jr AG, eds, Pediatric arrhythmias: electrophysiology and pacing. WB Saunders, Philadelphia, p 250 Fox R, Lumb MR, Hawkins DF, 1990, Persistent fetal sinus bradycardia associated with maternal anti-Ro antibodies. Case report. Br J Obstet Gynaecol 97(12):1151–1153 Brucato A, Cimaz R, Catelli L, Meroni P, 2000, Anti-Roassociated sinus bradycardia in newborns. Circulation 102(11): E88–E89 Sacks JH, Samai C, Gomez K, Kanaan U, 2013, Maternal antibody-associated fetal second-degree heart block and atrial flutter: case report and review. Pediatr Cardiol 34(8):2040–2043 Rosenthal E, 1998, New insights into the pathogenesis of anti-Ro antibody associated congenital complete heart block. Lupus 7(3): 135–136 Rein AJ, Mevorach D, Perles Z, Gavri S, Nadjari M, Nir A et al, 2009, Early diagnosis and treatment of atrioventricular block in the fetus exposed to maternal anti-SSA/Ro-SSB/la antibodies: a prospective, observational, fetal kinetocardiogram-based study. Circulation 119(14):1867–1872 Kelly EN, Sananes R, Chiu-Man C, Silverman ED, Jaeggi E, 2014, Prenatal anti-Ro antibody exposure, congenital complete atrioventricular heart block, and high-dose steroid therapy: impact on neurocognitive outcome in school-age children. Arthritis Rheum 66(8):2290–2296 Iida M, Inamura N, Takeuchi M, 2006, Newborn infant with maternal anti-SSA antibody-induced complete heart block accompanying cardiomyopathy. Circulation Journal : Official Journal of the Japanese Circulation Society 70(1):147–149 Costedoat-Chalumeau N, Georgin-Lavialle S, Amoura Z, Piette JC, 2005, Anti-SSA/Ro and anti-SSB/la antibody-mediated congenital heart block. Lupus 14(9):660–664 Costedoat-Chalumeau N, Amoura Z, Villain E, Cohen L, Piette JC, 2005, Anti-SSA/Ro antibodies and the heart: more than complete congenital heart block? A review of electrocardiographic and myocardial abnormalities and of treatment options. Arthritis Research& Therapy 7(2):69–73 Mohacsi P, Martinelli M, Banz Y, Boesch C, 2012, The clinical relevance of antibody-mediated rejection: a new era of heart transplantation. European Journal of Cardio-Thoracic Surgery : Official Journal of the European Association for Cardio-Thoracic Surgery 42(6):1047–1049 Daly KP, Chandler SF, Almond CS, Singh TP, Mah H, Milford E, Matte GS, Bastardi HJ, Mayer JE, Fynn-Thompson F, Blume ED, 2013, Antibody depletion for the treatment of crossmatch-positive pediatric heart transplant recipients. Pediatr Transplant 17(7):661– 669 Chih S, Tinckam KJ, Ross HJA, 2013, Survey of current practice for antibody-mediated rejection in heart transplantation. Am J Transplant Off J Am Soc Transplant Am Soc Transplant Surg 13(4):1069–1074 van der Gugten A, Bierings M, Frenkel J, 2008, Glucocorticoidassociated bradycardia. J Pediatr Hematol Oncol 30(2):172–175 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 487 EP 492 DI 10.1007/s12253-018-0403-y PG 6 ER PT J AU Blancas, S Medina-Berlanga, R Ortiz-Garcia, L Loredo-Ramirez, A Santos, L AF Blancas, Sugela Medina-Berlanga, Rogelio Ortiz-Garcia, Liliana Loredo-Ramirez, Alfredo Santos, Leticia TI Protein Expression Analysis in Uterine Cervical Cancer for Potential Targets in Treatment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Uterine cervical cancer; SEL1L; Notch3; SOCS3; Immunohistochemistry; Tissue microarray ID Uterine cervical cancer; SEL1L; Notch3; SOCS3; Immunohistochemistry; Tissue microarray AB Specific markers in lesions of the human uterine cervix cancer (UCC) are still needed for prognostic, diagnostic and/or therapeutic purposes. In this study we evaluated key molecules at protein level between normal epithelium, cervical intraepithelial neoplasia (CIN1–3) and invasive cancer of a group of molecules previously reported at mRNA level. For that purpose, human formalin-fixed paraffin embedded tissue microarrays (TMAs) were constructed containing 205 Mexican tissue core specimens. Immunohistochemistry and quantitative analysis of histological staining was performed against twenty-two distinct proteins for each core and the processing platform ImageJ. In the progression of the disease we found key statistical differences for the proteins SEL1, Notch3 and SOCS3. High expressions of SEL1L, Notch3 and SOCS3 have potential value to increase the prognostic of UCC in combination with markers such as p16INK4a. This study identified key drivers in cervical carcinogenesis that should be evaluated for the development of UCC therapies. C1 [Blancas, Sugela] A.C. (IPICYT), Instituto Potosino de Investigacion Cientifica y Tecnologica, Division de Biologia MolecularSan Luis Potosi, Mexico. [Medina-Berlanga, Rogelio] A.C. (IPICYT), Instituto Potosino de Investigacion Cientifica y Tecnologica, Division de Biologia MolecularSan Luis Potosi, Mexico. [Ortiz-Garcia, Liliana] Universidad Politecnica de Penjamo, Facultad de Ingenieria en Biotecnologia, PenjamoGuanajuato, Mexico. [Loredo-Ramirez, Alfredo] Laboratorio de Patologia Quirurgica, Mariano Arista 743, Interior 208San Luis Potosi, Mexico. [Santos, Leticia] A.C. (IPICYT), Instituto Potosino de Investigacion Cientifica y Tecnologica, Division de Biologia MolecularSan Luis Potosi, Mexico. RP Santos, L (reprint author), A.C. (IPICYT), Instituto Potosino de Investigacion Cientifica y Tecnologica, Division de Biologia Molecular, San Luis Potosi, Mexico. EM lsantos@ipicyt.edu.mx CR Walboomers JM, JacobsMV,ManosMM, Bosch FX, Kummer JA, Shah KV, Snijders PJ, Peto J, Meijer CJ, Munoz N, 1999, Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189(1):12–19. , DOI 10.1002/(SICI, 1096-9896(199909)189:1<12::AID-PATH431>3.0.CO;2-F Malinowski DP, 2007)Multiple biomarkers inmolecular oncology. I. Molecular diagnostics applications in cervical cancer detection. Expert Rev Mol Diagn 7(2):117–131. , DOI 10.1586/ 14737159.7.2.117 Brown CA, Bogers J, Sahebali S, Depuydt CE, De Prins F, Malinowski DP, 2012, Role of protein biomarkers in the detection of high-grade disease in cervical cancer screening programs. J Oncol 2012:289315–289311. , DOI 10.1155/2012/289315 Santos L, Leon-Galvan MF, Marino-Marmolejo EN, Barba de la Rosa AP, De Leon RA, Gonzalez-Amaro R, Guevara- Gonzalez RG, 2011, Identification of differential expressed transcripts in cervical cancer of Mexican patients. Tumour Biol: J Int Soc Oncodev Biol Med 32(3):561–568. https:// doi.org/10.1007/s13277-010-0151-4 Palermo R, Checquolo S, Bellavia D, Talora C, Screpanti I, 2014, The molecular basis of notch signaling regulation: a complex simplicity. Curr Mol Med 14(1):34–44 Allenspach EJ, Maillard I, Aster JC, PearWS, 2002, Notch signaling in cancer. Cancer Biol Ther 1(5):466–476 Arafa M, Boniver J, Delvenne P, 2010, Progression model tissue microarray, TMA, for the study of uterine carcinomas. DisMarkers 28(5):267–272. , DOI 10.3233/DMA-2010-0709 Zhu H, Zhou X, Redfield S, Lewin J, Miele L, 2013, Elevated Jagged-1 and Notch-1 expression in high grade and metastatic prostate cancers. Am J Transl Res 5(3):368–378 Cornish TC, Halushka MK, 2009, Color deconvolution for the analysis of tissue microarrays. Anal Quant Cytol Histol/ Int Acad Cytol Am Soc Cytol 31(5):304–312 Shapiro SS, Wilk MB, 1965, An analysis of variance test for normality, complete samples). Biometrika 52:591-&. , DOI 10.2307/2333709 Keyes TK, Levy MS, 1997, Analysis of Levene's test under design imbalance. J Educ Behav Stat 22(2):227–236 Box GEP, Cox DR, 1964, An analysis of transformations. J Roy Stat Soc B 26(2):211–252 Cicchett Dv, Collins D, Latvis V, 1972, Program for Comparing Group Means - One-Way Anova and Tukey Multiple-Range Tests. Behav Sci 17, 4):399-& Keselman HJ, Murray R, 1974, Tukey tests for pair-wise contrasts following analysis of variance - is there a type 4 error. Psychol Bull 81(9):608–609. , DOI 10.1037/h0036938 Li JJ, 2011, A simple and flexible holm gatekeeping procedure. Biom J 53(5):797–809. , DOI 10.1002/bimj.201000040 Ikenberg H, Bergeron C, Schmidt D, Griesser H, Alameda F, Angeloni C, Bogers J, Dachez R, Denton K, Hariri J, Keller T, von Knebel Doeberitz M, Neumann HH, Puig-Tintore LM, Sideri M, Rehm S, Ridder R, Group PS, 2013, Screening for cervical cancer precursors with p16/Ki-67 dual-stained cytology: results of the PALMS study. J Natl Cancer Inst 105(20):1550–1557. https:// doi.org/10.1093/jnci/djt235 Wentzensen N, Fetterman B, Castle PE, Schiffman M, Wood SN, Stiemerling E, Tokugawa D, Bodelon C, Poitras N, Lorey T, Kinney W, 2015, p16/Ki-67 dual stain cytology for detection of cervical Precancer in HPV-positive women. J Natl Cancer Inst 107(12):djv257. , DOI 10.1093/jnci/djv257 Garner DM, 2016, RE: p16/Ki-67 dual stain cytology for detection of cervical Precancer in HPV-positive women. J Natl Cancer Inst 108(2):djv389. , DOI 10.1093/jnci/djv389 Benczik M, Galamb A, Koiss R, Kovacs A, Jaray B, Szekely T, Szekerczes T, Schaff Z, Sobel G, Jeney C, 2016, Claudin-1 as a biomarker of cervical cytology and histology. Pathol Oncol Res 22(1):179–188. , DOI 10.1007/s12253-015-9990-z Klaes R, Friedrich T, Spitkovsky D, Ridder R, Rudy W, Petry U, Dallenbach-Hellweg G, Schmidt D, von Knebel Doeberitz M, 2001, Overexpression of p16(INK4A, as a specific marker for dysplastic and neoplastic epithelial cells of the cervix uteri. Int J Cancer 92(2):276–284 Volgareva G, Zavalishina L, Andreeva Y, Frank G, Krutikova E, Golovina D, Bliev A, Spitkovsky D, Ermilova V, Kisseljov F, 2004, Protein p16 as a marker of dysplastic and neoplastic alterations in cervical epithelial cells. BMC Cancer 4(58). https://doi. org/10.1186/1471-2407-4-58 Harada Y, Ozaki K, Suzuki M, Fujiwara T, Takahashi E, Nakamura Y, Tanigami A, 1999, Complete cDNA sequence and genomic organization of a human pancreas-specific gene homologous to Caenorhabditis elegans sel-1. J Hum Genet 44(5):330–336. , DOI 10.1007/s100380050171 Li S, Francisco AB, Munroe RJ, Schimenti JC, Long Q, 2010, SEL1L deficiency impairs growth and differentiation of pancreatic epithelial cells. BMC Dev Biol 10:19. , DOI 10.1186/ 1471-213X-10-19 Cattaneo M, Lotti LV, Martino S, Alessio M, Conti A, Bachi A, Mariani-Costantini R, Biunno I, 2011, Secretion of novel SEL1L endogenous variants is promoted by ER stress/UPR via endosomes and shed vesicles in human cancer cells. PLoS One 6(2):e17206. , DOI 10.1371/journal.pone.0017206 Ashktorab H, Green W, Finzi G, Sessa F, Nouraie M, Lee EL, Morgano A, Moschetta A, Cattaneo M, Mariani-Costantini R, Brim H, Biunno I, 2012, SEL1L, an UPR response protein, a potential marker of colonic cell transformation. Dig Dis Sci 57(4): 905–912. , DOI 10.1007/s10620-011-2026-y Wang SS, Hsiao R, Limpar MM, Lomahan S, Tran TA, Maloney NJ, Ikegaki N, TangXX(2014, Destabilization ofMYC/MYCN by the mitochondrial inhibitors, metaiodobenzylguanidine, metformin and phenformin. Int J Mol Med 33(1):35–42. , DOI 10. 3892/ijmm.2013.1545 Duffy DJ, Krstic A, Schwarzl T, Higgins DG, Kolch W, 2014, GSK3 inhibitors regulate MYCN mRNA levels and reduce neuroblastoma cell viability throughmultiple mechanisms, including p53 and Wnt signaling. Mol Cancer Ther 13(2):454–467. https://doi. org/10.1158/1535-7163.MCT-13-0560-T Kang JH, Rychahou PG, Ishola TA, Qiao J, Evers BM, Chung DH, 2006, MYCN silencing induces differentiation and apoptosis in human neuroblastoma cells. Biochem Biophys Res Commun 351(1):192–197. , DOI 10.1016/j.bbrc.2006.10.020 Cui H,KongY, Xu M, ZhangH(2013, Notch3 functions as a tumor suppressor by controlling cellular senescence. Cancer Res 73(11): 3451–3459. , DOI 10.1158/0008-5472.CAN-12-3902 Jaskula-Sztul R, Eide J, Tesfazghi S, Dammalapati A, HarrisonAD, Yu XM, Scheinebeck C, Winston-McPherson G, Kupcho KR, Robers MB, Hundal AK, Tang W, Chen H, 2015, Tumorsuppressor role of Notch3 in medullary thyroid carcinoma revealed by genetic and pharmacological induction. Mol Cancer Ther 14(2): 499–512. , DOI 10.1158/1535-7163.MCT-14-0073 PelulloM, Quaranta R, Talora C, Checquolo S, Cialfi S, Felli MP, te Kronnie G, Borga C, Besharat ZM, Palermo R, Di Marcotullio L, Capobianco AJ, Gulino A, Screpanti I, Bellavia D, 2014, Notch3/ Jagged1 circuitry reinforces notch signaling and sustains T-ALL. Neoplasia 16(12):1007–1017. , DOI 10.1016/j.neo.2014. 10.004 Jung JG, Stoeck A, Guan B,Wu RC, Zhu H, Blackshaw S, Shih Ie M,Wang TL, 2014, Notch3 interactome analysis identifiedWWP2 as a negative regulator of Notch3 signaling in ovarian cancer. PLoS Genet 10(10):e1004751. , DOI 10.1371/journal.pgen. 1004751 Bianchi L, Canton C, Bini L, Orlandi R, Menard S, Armini A, CattaneoM, Pallini V, Bernardi LR, Biunno I, 2005, Protein profile changes in the human breast cancer cell line MCF-7 in response to SEL1L gene induction. Proteomics 5(9):2433–2442. https://doi. org/10.1002/pmic.200401283 Yeasmin S, Nakayama K, Rahman MT, Rahman M, Ishikawa M, Iida K, Otsuki Y, Kobayashi H, Nakayama S, Miyazaki K, 2010, Expression of nuclear Notch3 in cervical squamous cell carcinomas and its association with adverse clinical outcomes. Gynecol Oncol 117(3):409–416. , DOI 10.1016/j.ygyno.2010.03.004 Tripathi R, Rath G, Jawanjal P, Sharma S, Singhal P, Bhambhani S, Hussain S, Bharadwaj M, 2014, Clinical impact of de-regulated Notch-1 and Notch-3 in the development and progression of HPV-associated different histological subtypes of precancerous and cancerous lesions of human uterine cervix. PLoS One 9(6): e98642. , DOI 10.1371/journal.pone.0098642 Bellavia D, Campese AF, Alesse E, Vacca A, Felli MP, Balestri A, Stoppacciaro A, Tiveron C, Tatangelo L, Giovarelli M, Gaetano C, Ruco L, Hoffman ES, Hayday AC, Lendahl U, Frati L, Gulino A, Screpanti I, 2000, Constitutive activation of NF-kappaB and T-cell leukemia/lymphoma in Notch3 transgenic mice. EMBO J 19(13):3337–3348. https:// doi.org/10.1093/emboj/19.13.3337 Bellavia D, Campese AF, Checquolo S, Balestri A, Biondi A, Cazzaniga G, Lendahl U, Fehling HJ, Hayday AC, Frati L, von Boehmer H, Gulino A, Screpanti I, 2002, Combined expression of pTalpha and Notch3 in Tcell leukemia identifies the requirement of preTCR for leukemogenesis. Proc Natl Acad Sci U S A 99, 6): 3788–3793. , DOI 10.1073/pnas.062050599 ZhangY, Jia L, Lee SJ,WangMM(2007, Conserved signal peptide of Notch3 inhibits interaction with proteasome. Biochem Biophys Res Commun 355(1):245–251. , DOI 10.1016/j.bbrc. 2007.01.151 Palermo R, Checquolo S, Giovenco A, Grazioli P, Kumar V, Campese AF, Giorgi A, Napolitano M, Canettieri G, Ferrara G, Schinina ME, Maroder M, Frati L, Gulino A, Vacca A, Screpanti I, 2012, Acetylation controls Notch3 stability and function in T-cell leukemia. Oncogene 31(33):3807–3817. , DOI 10.1038/ onc.2011.533 Sasi W, Sharma AK, Mokbel K, 2014, The role of suppressors of cytokine signalling in human neoplasms. Mol Biol Int 2014(630797):1–24. , DOI 10.1155/2014/630797 Puhr M, Santer FR, Neuwirt H, Marcias G, Hobisch A, Culig Z, 2010, SOCS-3 antagonises the proliferative and migratory effects of fibroblast growth factor-2 in prostate cancer by inhibition of p44/ p42MAPKsignalling. Endocr Relat Cancer 17(2):525–538. https:// doi.org/10.1677/ERC-10-0007 Kim MH, Kim MS, Kim W, Kang MA, Cacalano NA, Kang SB, Shin YJ, Jeong JH, 2015, Suppressor of cytokine signaling, SOCS, genes are silenced by DNA hypermethylation and histone deacetylation and regulate response to radiotherapy in cervical cancer cells. PLoS One 10(4):e0123133. , DOI 10.1371/ journal.pone.0123133 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 493 EP 501 DI 10.1007/s12253-018-0401-0 PG 9 ER PT J AU Singh, L Saini, N Pushker, N Bakhshi, S Sen, S Nag, CT Kashyap, S AF Singh, Lata Saini, Neeru Pushker, Neelam Bakhshi, Sameer Sen, Seema Nag, C Tapas Kashyap, Seema TI Mutational Analysis of the Mitochondrial DNA Displacement-Loop Region in Human Retinoblastoma with Patient Outcome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Retinoblastoma; D-loop; Mitochondrial DNA; DNA sequencing; Electron microscopy ID Retinoblastoma; D-loop; Mitochondrial DNA; DNA sequencing; Electron microscopy AB Alteration in mitochondrial DNA plays an important role in the development and progression of cancer. The Displacement Loop (Dloop) region of mitochondrial DNA (mtDNA) is the regulatory region for its replication and transcription. Therefore, we aimed to characterize mutations in the D-loop region of mitochondrial DNA along with themorphological changes and analyzed their impact on survival in retinoblastoma patients. mtDNA D-loop region was amplified by Nested-Polymerase Chain Reaction (Nested-PCR) and mutations were analyzed in 60 tumor samples from retinoblastoma patients by DNA sequencing. Transmission electron microscopy was performed on 5 retinoblastoma specimens. Mutations were correlated with clinical, histopathological parameters and patient survival. D-loop mutations were found in total of 52/60 (86.6%) patients. The most common mutations were T to C and C to T followed by A to G. There were 5.81% mutations which were not previously reported in the MITOMAP database. A73G (83.33%) were the most frequent mutations found in our cases and it was statistically significant with poor tumor differentiation and age. In addition, this study was further analyzed formorphological changes in retinoblastoma that had disorganized, swollen and less numbers of mitochondria on electron microscopy. This is the first study showing high frequency of mtDNA mutation which might be due to abnormal morphology of mitochondria in retinoblastoma. Our results indicate that pathogenic mtDNA D-loop mutations may be involved in tumorigenesis of retinoblastoma tumor. C1 [Singh, Lata] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular PathologyNew Delhi, India. [Saini, Neeru] Institute of Genomics and Integrative Biology, Functional Genomics UnitNew Delhi, India. [Pushker, Neelam] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of OphthalmologyNew Delhi, India. [Bakhshi, Sameer] All India Institute of Medical Sciences, IRCH, Department of Medical OncologyNew Delhi, India. [Sen, Seema] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular PathologyNew Delhi, India. [Nag, C Tapas] All India Institute of Medical Sciences, Department of AnatomyNew Delhi, India. [Kashyap, Seema] All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular PathologyNew Delhi, India. RP Singh, L (reprint author), All India Institute of Medical Sciences, Dr. R. P. Centre for Ophthalmic Sciences, Department of Ocular Pathology, New Delhi, India. EM lata.aiims@gmail.com CR Larsson NG, Clayton DA, 1995, Molecular genetics aspects of human mitochondrial disorders. Annu Rev Genet 29:151–178 Lane N, 2006, Mitochondrial disease: powerhouse of disease. Nature 440:600–602 Falk MJ, 2010, Neurodevelopmental manifestations of mitochondrial disease. J Dev Behav Pediatr 31:610 Taanman JW, 1999, The mitochondrial genome: structure, transcription, translation and replication. Biochimi Biophys Acta, BBA)-Bioenergetics 9(1410):103–123 Anderson S, Bankier AT, Barrell BG et al, 1981, Sequence and organization of the human mitochondrial genome. Nature 290: 457–465 Vives-Bauza C, Gonzalo R, Manfredi G et al, 2006, Enhanced ROS production and antioxidant defenses in cybrids harbouring mutations in mtDNA. Neurosci Lett 391:136–141 Stoneking M, 2000, Hypervariable sites in the mtDNA control region are mutational hotspots. Am J Hum Genet 67:1029–1032 Van Oven M, Kayser M, 2009, Updated comprehensive phylogenetic tree of global human mitochondrial DNA variation. Hum Mutat 30:E386–E394 Mims MP, Hayes TG, Zheng S et al, 2006, Mitochondrial DNA G10398A polymorphism and invasive breast cancer in African- American women. Cancer Res 66:1880–1881 Janovjak H, Sapra KT, Kedrov A et al, 2008, From valleys to ridges: exploring the dynamic energy landscape of single membrane proteins. Chem Phys Chem 9:954–966 Arismendi-Morillo G, 2011, Electron microscopy morphology of the mitochondrial network in gliomas and their vascular microenvironment. Biochim Biophys Acta, BBA)-Bioenergetics 1807: 602–608 Lohmann DR, 1999, RB1 gene mutations in retinoblastoma. Hum Mutat 14:283–288 Wallace DC, Chalkia D, 2013, Mitochondrial DNA genetics and the heteroplasmy conundrum in evolution and disease. Cold Spring Harb Perspect Biol 5:a021220 Alonso A, Martin P, Albarran C et al, 1997, Detection of somatic mutations in the mitochondrial DNA control region of colorectal and gastric tumors by heteroduplex and single-strand conformation analysis. Electrophoresis 18:682–685 Jarrett SG, Lewin AS, Boulton ME, 2010, The importance of mitochondria in age-related and inherited eye disorders. Ophthalmic Res 44:179–190 Schrier SA, Falk MJ, 2011, Mitochondrial disorders and the eye. Curr Opin Ophthalmol 22:325–331 Wallace DC, Singh G, Lott MT et al, 1998, Mitochondrial DNA mutation associated with Leber's hereditary optic neuropathy. Science 242:1427 Barreau E, Brossas JY, Courtois Y, Treton JA, 1996, Accumulation of mitochondrial DNA deletions in human retina during aging. Invest Ophthalmol Vis Sci 37:384–391 Kowluru RA, 2005, Diabetic retinopathy: mitochondrial dysfunction and retinal capillary cell death. Antioxid Redox Signal 7:1581 Saraswathy S, Rao NA, 2009, Mitochondrial proteomics in experimental autoimmune uveitis oxidative stress. Invest Ophthalmol Vis Sci 50:5559–5566 Tezel G, 2006, Oxidative stress in glaucomatous neurodegeneration: mechanisms and consequences. Prog Retin Eye Res 25:490–513 DiMauro S, Garone C, 2010, Historical perspective on mitochondrial medicine. Dev Disabil Res Rev 16:106–113 Suzuki M, Toyooka S, Miyajima K et al, 2003, Alterations in the mitochondrial displacement loop in lung cancers. Clin Cancer Res 9:5636–5641 Habano W, Sugai T, Yoshida T et al, 1999, Mitochondrial gene mutation, but not large-scale deletion, is a feature of colorectal carcinomas with mitochondrial microsatellite instability. Int J Cancer 2683:625–629 Liu VW, Shi HH, Cheung AN et al, 2001, High incidence of somatic mitochondrial DNA mutations in human ovarian carcinomas. Cancer Res 61:5998–6001 Okochi O, Hibi K, Uemura T et al, 2002, Detection of mitochondrial DNA alterations in the serum of hepatocellular carcinoma patients. Clin Cancer Res 8:2875–2878 Rosson D, Keshgegian AA, 2004, Frequent mutations in the mitochondrial control region DNA in breast tissue. Cancer Lett 215:89–94 Tamura G, Nishizuka S, Maesawa C et al, 1999, Mutations in mitochondrial control region DNA in gastric tumours of Japanese patients. Eur J Cancer 35:316–319 Levin BC, Cheng H, Reeder DJ, 1999, A human mitochondrial DNA standard reference material for quality control in forensic identification, medical diagnosis, and mutation detection. Genomics 55:135–146 Miyazono F, Schneider PM, Metzger R et al, 2002, Mutations in the mitochondrial DNA D-Loop region occurs frequently in adenocarcinoma in Barrett’s esophagus. Oncogene 21:3780–3783 Hibi K, Nakayama H, Yamazaki T et al, 2001, Mitochondrial DNA alteration in esophageal cancer. Int J Cancer 92:319–321 Sharma H, Singh A, Sharma C et al, 2005, Mutations in the mitochondrial DNA D-loop region is frequent in cervical cancer. Cancer Cell Int 16:34 Chatterjee A, Mambo E, Sidransky D, 2006, Mitochondrial DNA mutations in human cancer. Oncogene 25:4663–4674 Sharawat SK, Bakhshi R, Vishnubhatla S, Bakhshi S, 2010, Mitochondrial D-loop variations in paediatric acute myeloid leukaemia: a potential prognostic marker. Br J Haematol 149:391–398 Richard SM, Bailliet G, Paez GL et al, 2000, Nuclear and mitochondrial genome instability in human breast cancer. Cancer Res 60:4231–4237 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 503 EP 512 DI 10.1007/s12253-018-0391-y PG 10 ER PT J AU Inno, A Di Noia, V Martini, M D’Argento, E Di Salvatore, M Arena, V Schinzari, G Orlandi, A Larocca, ML Cassano, A Barone, C AF Inno, Alessandro Di Noia, Vincenzo Martini, Maurizio D’Argento, Ettore Di Salvatore, Mariantonietta Arena, Vincenzo Schinzari, Giovanni Orlandi, Armando Larocca, Maria Luigi Cassano, Alessandra Barone, Carlo TI Erlotinib for Patients with EGFR Wild-Type Metastatic NSCLC: a Retrospective Biomarkers Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR wild-type; NSCLC; Erlotinib; K-RAS mutation; IGF1-R expression; C-MET expression ID EGFR wild-type; NSCLC; Erlotinib; K-RAS mutation; IGF1-R expression; C-MET expression AB Erlotinib is approved for the treatment of patients with EGFR mutation positive, metastatic NSCLC. It is also approved as second/ third line therapy for EGFR mutation negative patients, but in this setting the benefit of erlotinib is modest and there is no validated biomarker for selecting EGFR wild-type patients who may benefit the most from the treatment. We retrospectively assessed EGFR and K-RAS mutational status, and EGFR, c-MET and IGF1-R expression in tumor samples of 72 patients with metastatic NSCLC treated with erlotinib after at least one prior line of chemotherapy, from 2008 to 2012. We analyzed the association between biomarkers and outcome (RR, PFS, and OS). EGFR mutated patients achieved a better RR (56% vs 8%, p = .002), PFS (10 vs 3 months, HR 0.53, p = 0.48) and OS (20 vs 6 months, HR 0.55, p = .07), compared to EGFR wild-type patients. Among 63 EGFR wild-type patients, those with EGFR high-expression had a better outcome in terms of RR (40% vs 2%, p = .002), PFS (7.5 vs 2 months, HR 0.45, p = .007) and OS (30 vs 5 months, HR 0.34, p < .001) compared to patients with EGFR intermediate or low/negative-expression. IGF1-R expression, c-MET expression and K-RAS mutational status did not significantly affect the outcome; however, no patients with K-RAS mutation or c-MET high-expression achieved an objective response. In patients with metastatic, chemo-refractory EGFR wild-type NSCLC, EGFR high-expression may represent a positive predictor of activity for erlotinib, whereas K-RAS mutation and c-MET high-expression may predict lack of activity. These findings deserve further prospective evaluation. C1 [Inno, Alessandro] Ospedale Sacro Cuore Don Calabria, Cancer Care Center, Medical Oncology Unit, Via don A. Sempreboni 5, 37024 Negrar, Verona, Italy. [Di Noia, Vincenzo] Universita Cattolica del Sacro Cuore, Department of Medical OncologyRome, Italy. [Martini, Maurizio] Universita Cattolica del Sacro Cuore, Department of PathologyRome, Italy. [D’Argento, Ettore] Universita Cattolica del Sacro Cuore, Department of Medical OncologyRome, Italy. [Di Salvatore, Mariantonietta] Universita Cattolica del Sacro Cuore, Department of Medical OncologyRome, Italy. [Arena, Vincenzo] Universita Cattolica del Sacro Cuore, Department of PathologyRome, Italy. [Schinzari, Giovanni] Universita Cattolica del Sacro Cuore, Department of Medical OncologyRome, Italy. [Orlandi, Armando] Universita Cattolica del Sacro Cuore, Department of Medical OncologyRome, Italy. [Larocca, Maria Luigi] Universita Cattolica del Sacro Cuore, Department of PathologyRome, Italy. [Cassano, Alessandra] Universita Cattolica del Sacro Cuore, Department of Medical OncologyRome, Italy. [Barone, Carlo] Universita Cattolica del Sacro Cuore, Department of Medical OncologyRome, Italy. RP Inno, A (reprint author), Ospedale Sacro Cuore Don Calabria, Cancer Care Center, Medical Oncology Unit, 37024 Negrar, Italy. EM alessandro.inno@sacrocuore.it CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90. https://doi. org/10.3322/caac.20107 Minuti G, D'Incecco A, Cappuzzo F, 2013, Targeted therapy for NSCLC with driver mutations. Expert Opin Biol Ther 13:1401– 1412. , DOI 10.1517/14712598.2013.827657 Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A et al, 2016, Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung Cancer. N Engl J Med 375: 1823–1833. , DOI 10.1056/NEJMoa1606774 Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E et al, 2015, Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung Cancer. N Engl J Med 373:123–135. , DOI 10.1056/NEJMoa1504627 Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE et al, 2015, Nivolumab versus docetaxel in advanced Nonsquamous non-small-cell lung Cancer. N Engl J Med 373: 1627–1639. , DOI 10.1056/NEJMoa1507643 Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP et al, 2015, Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 372:2018–2028. , DOI 10.1056/ NEJMoa1501824 Vallee A, Sagan C, Le Loupp AG, Bach K, Dejoie T, Denis MG, 2013, Detection of EGFR gene mutations in non-small cell lung cancer: lessons from a single-institution routine analysis of 1,403 tumor samples. Int J Oncol 43:1045–1051. , DOI 10.3892/ ijo.2013.2056 Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H et al, 2010, Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med 362:2380–2388. , DOI 10.1056/NEJMoa0909530 Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E et al, 2012, Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutationpositive non-small-cell lung cancer, EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 13:239–246. , DOI 10.1016/S1470-2045(11)70393-X Sequist LV, Yang JC, Yamamoto N, O'Byrne K, Hirsh V, Mok T et al, 2013, Phase III study of Afatinib or cisplatin plus Pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 31:3327–3334. , DOI 10.1200/JCO. 2012.44.2806 Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, Thongprasert S et al, 2005, Erlotinib in previously treated nonsmall- cell lung cancer. N Engl J Med 353:123–132. https://doi. org/10.1056/NEJMoa050753 Reck M, van Zandwijk N, Gridelli C, Baliko Z, Rischin D, Allan S et al, 2010, Erlotinib in advanced non-small cell lung cancer: efficacy and safety findings of the global phase IV Tarceva lung Cancer survival treatment study. J Thorac Oncol 5:1616–1622 Rossi S, D'Argento E, BassoM, Strippoli A,DadduzioV, Cerchiaro E et al, 2015)Different EGFR genemutations in exon 18, 19 and 21 as prognostic and predictive markers in NSCLC: a single institution analysis. Mol Diagn Ther 20:55–63. , DOI 10.1007/ s40291-015-0176-x Natalicchio MI, Improta G, Zupa A, Cursio OE, Stampone E, Possidente L et al, 2014, Pyrosequencing evaluation of lowfrequency KRAS mutant alleles for EGF receptor therapy selection in metastatic colorectal carcinoma. Future Oncol 10:713–723. , DOI 10.2217/fon.13.233 Inno A, Di Salvatore M, Cenci T, Martini M, Orlandi A, Strippoli A et al, 2011, Is there a role for IGF1R and c-MET pathways in resistance to cetuximab in metastatic colorectal cancer? Clin Colorectal Cancer 10:325–332. , DOI 10.1016/j.clcc.2011.03.028 Gaber R, Watermann I, Kugler C, Reinmuth N, Huber RM, Schnabel PA et al, 2014, Correlation of EGFR expression, gene copy number and clinicopathological status in NSCLC. Diagn Pathol 0:165. , DOI 10.1186/s13000-014-0165-0 Hirsch FR, Varella-Garcia M, Bunn PA Jr, Di Maria MV, Veve R, Bremmes RMet al, 2003, Epidermal growth factor receptor in nonsmall- cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol 21: 3798–3807. , DOI 10.1200/JCO.2003.11.069 Coudert B, Ciuleanu T, Park K, Wu YL, Giaccone G, Brugger W et al, 2012, Survival benefit with erlotinib maintenance therapy in patients with advanced non-small-cell lung cancer, NSCLC, according to response to first-line chemotherapy. Ann Oncol 23: 388–394. , DOI 10.1093/annonc/mdr125 Garassino MC, Martelli O, Broggini M, Farina G, Veronese S, Rulli E et al, 2013, Erlotinib versus docetaxel as second-line treatment of patients with advanced non-small-cell lung cancer and wild-type EGFR tumours, TAILOR): a randomised controlled trial. Lancet Oncol. 14:981–988. , DOI 10.1016/S1470-2045(13)70310-3 Laurie SA, Goss GD, 2013, Role of epidermal growth factor receptor inhibitors in epidermal growth factor receptor wild-type nonsmall- cell lung cancer. J Clin Oncol 31:1061–1069. , DOI 10.1200/JCO.2012.43.4522 Zer A, Leighl NB, 2014, Second-line therapy in non-small-cell lung cancer: the DELTA between different genotypes widens. J Clin Oncol 32:1874–1881 Hirsch FR, Varella-Garcia M, Bunn PA Jr, Franklin WA, Dziadziuszko R, Thatcher N et al, 2006, Molecular predictors of outcome with gefitinib in a phase III placebo-controlled study in advanced non-small-cell lung cancer. J Clin Oncol 24:5034–5042 Parra HS, Cavina R, Latteri F, Zucali PA, Campagnoli E, Morenghi E et al, 2004, Analysis of epidermal growth factor receptor expression as a predictive factor for response to gefitinib, 'Iressa', ZD1839, in non-small-cell lung cancer. Br J Cancer 91:208–212. , DOI 10.1038/sj.bjc.6601923 Clark GM, Zborowski DM, Culbertson JL, Whitehead M, Savoie M, Seymour L et al, 2006, Clinical utility of epidermal growth factor receptor expression for selecting patients with advanced non-small cell lung cancer for treatment with erlotinib. J Thorac Oncol 1:837–846 Lohinai Z, Klikovits T,Moldvay J, Ostoros G, Raso E, Timar J et al, 2017, KRAS-mutation incidence and prognostic value are metastatic site-specific in lung adenocarcinoma: poor prognosis in patients with KRAS mutation and bone metastasis. Sci Rep 7:39721. , DOI 10.1038/srep39721 Cserepes M, Ostoros G, Lohinai Z, Raso E, Barbai T, Timar J et al, 2014, Subtype-specific KRAS mutations in advanced lung adenocarcinoma: a retrospective study of patients treated with platinumbased chemotherapy. Eur J Cancer 50:1819–1828. , DOI 10.1016/j.ejca.2014.04.001 Califano R, Landi L, Cappuzzo F, 2012, Prognostic and predictive value of K-RAS mutations in non-small cell lung cancer. Drugs 72(Suppl 1):28–36. , DOI 10.2165/1163012-S0- 000000000-00000 Rulli E, Marabese M, Torri V, Farina G, Veronese S, Bettini A et al, 2015, Value of KRAS as prognostic or predictive marker in NSCLC: results from the TAILOR trial. Ann Oncol 26:2079– 2084. , DOI 10.1093/annonc/mdv318 Zhu CQ, da Cunha Santos G, Ding K, Sakurada A, Cutz JC, Liu N et al, 2008, Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada clinical trials group study BR.21. J Clin Oncol 26:4268–4275. , DOI 10.1200/ JCO.2007.14.8924 Del Re M, Tiseo M, Bordi P, D'Incecco A, Camerini A, Petrini I et al, 2017, Contribution of KRAS mutations and c.2369C > T, p.T790M, EGFR to acquired resistance to EGFR-TKIs in EGFR mutant NSCLC: a study on circulating tumor DNA. Oncotarget 8: 13611–13619. , DOI 10.18632/oncotarget.6957 Dziadziuszko R, Wynes MW, Singh S, Asuncion BR, Ranger- Moore J, Konopa K et al, 2012, Correlation between MET gene copy number by silver in situ hybridization and protein expression by immunohistochemistry in non-small cell lung cancer. J Thorac Oncol 7:340–347. , DOI 10.1097/JTO.0b013e318240ca0d Passiglia F, Van Der Steen N, Raez L, Pauwels P, Gil-Bazo I, Santos E et al, 2014, The role of cMet in non-small cell lung cancer resistant to EGFR-inhibitors: did we really find the target? Curr Drug Targets 15:1284–1292 Schildhaus HU, Schultheis AM, Ruschoff J, Binot E, Merkelbach- Bruse S, Fassunke J et al, 2015, MET amplification status in therapy- naive adeno- and squamous cell carcinomas of the lung. Clin Cancer Res 21:907–915. , DOI 10.1158/1078-0432.CCR- 14-0450 Meert AP, Martin B, Verdebout JM, Noel S, Ninane V, Sculier JP, 2005, Is there a relationship between c-erbB-1 and c-erbB-2 amplification and protein overexpression in NSCLC? Lung Cancer 47: 325–336 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 513 EP 520 DI 10.1007/s12253-018-0404-x PG 8 ER PT J AU Zalatnai, A Galambos, E Perjesi, E AF Zalatnai, Attila Galambos, Eszter Perjesi, Eszter TI Importance of Immunohistochemical Detection of Somatostatin Receptors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Neuroendocrine tumors; Immunohistochemistry; Somatostatin receptors ID Neuroendocrine tumors; Immunohistochemistry; Somatostatin receptors AB The long-acting somatostatin analogs represent important weapons in treatment protocols of patients with neuroendocrine tumors. Because these peptides preferentially bind to the specific somatostatin receptors, the targeted therapy requires detection of them. As one of the national consulting centers, here we present the results of the immunohistochemically positive neuroendocrine neoplasms diagnosed between 2010 and 2014. Twenty-four paraffin-embedded cases (14 females 10 men, 21–79 years) from different localizations were found to express somatostatin-receptor type 2 (SSTR2). None of the patients has received previous hormonal therapy. The immune reactions have shown membranous, cytoplasmic or mixed patterns. There was no correlation between the expression and the chromogranin A levels, the grades or the hormonal activity/inactivity of the given neoplasms. Our results show that the immunohistochemical detection of SSTR2 is a quick, reliable and effective tool that provides useful information to the oncologists for the therapeutic decision. Because the incidence of the neuroendocrine tumors is still low, centralized pathological units are needed to perform such technique. C1 [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26,, H-1085 Budapest, Hungary. [Galambos, Eszter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26,, H-1085 Budapest, Hungary. [Perjesi, Eszter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26,, H-1085 Budapest, Hungary. RP Zalatnai, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM zalatnai@korb1.sote.hu CR Pollak MN, Schally AV, 1998, Mechanisms of antineoplastic action of somatostatin analogs. Proc Soc Exp BiolMed 217:143–152 Rai U, Thrimawithana TR, Valery C,Young SA, 2015, Therapeutic uses of somatostatin and its analogues: Current view and potential applications. Pharmacol Ther 152:98–110 Albertelli M,Nazzari E, Sciallero S, Grillo F,Morbelli S,De Cian F, Cittadini G, Ambrosetti E, Ciarmiello A, Ferone D, 2017, Antitumoral effects of somatostatin analogs: a lesson from the CLARINET study. J Endocrinol Investig May 26. , DOI 10.1007/s40618-017-0692-0 Chalabi M, Duluc C, Caron P, Vezzosi D, Guillermet-Guibert J, Pyronnet S, Bousquet C, 2014, Somatostatin analogs: does pharmacology impact antitumor efficacy? Trends EndocrinolMetab 25: 115–127 Pyronnet S, Bousquet C,Najib S, Azar R, Laklai H, Susini C, 2008, Antitumor effects of somatostatin. Mol Cell Endocrinol 286:230– 237 Pan MG, Florio T, Stork PJS, 1992, G protein activation of a hormone-stimulated protein-tyrosine phosphatase in human tumor cells. Science 256:1215–1217 Teijeiro R, Rios R, Costoya JA, Castro R, Bello JL, Devesa J, Arce VM, 2002, Activation of human somatostatin receptor 2 promotes apoptosis through a mechanism that is independent from induction of p53. Cell Physiol Biochem 12:31–38 Murray RD, Kim K, Ren SG, Chelly M, Umehara Y, Melmed S, 2004, Central and peripheral actions of somatostatin on the growth hormone–IGF-I axis. J Clin Invest 114:349–356 Bosman FT, Carneiro F, 2010, WHO Classification of Tumours, Pathology and Genetics of Tumours of the Digestive. IARC Press, System Narayanan S, Kunz PL, 2015, Role of somatostatin analogues in the treatment of neuroendocrine tumors. Natl Compr Canc Netw 13:109–117 Childs A, Vesely C, Ensell L, Lowe H, Luong TV, Caplin ME, Toumpanakis C, Thirlwell C, Hartley JA, Meyer T, 2016, Expression of somatostatin receptors 2 and 5 in circulating tumor cells from patients with neuroendocrine tumors. Br J Cancer 115: 1540–1547 Gonzalez B, Vargas G, Ramirez C, Asa S, Cheng S, Sandoval C, Mercado M, 2014, Cytoplasmic expression of SSTR2 and 5 by immunohistochemistry and by RT/PCR is not associated with the pharmacological response to octreotide. Endocrinol Nutr 61:523– 530 Sclafani F, Carnaghi C, Di Tommaso L, Rodari M, Destro A, Rimassa L, Giordano L, Chiti A, Roncalli M, Santoro A, 2011, Detection of somatostatin receptor subtypes 2 and 5 by somatostatin receptor scintigraphy and immunohistochemistry: clinical implications in the diagnostic and therapeutic management of gastroenteropancreatic neuroendocrine tumors. Tumori 97:620– 628 Diakatou E, Alexandraki KI, Tsolakis AV, Kontogeorgos G, Chatzellis E, Leonti A, Kaltsas GA, 2015, Somatostatin and dopamine receptor expression in neuroendocrine neoplasms: correlation of immunohistochemical findings with somatostatin receptor scintigraphy visual scores. Clin Endocrinol 83:420–428 Mehta S, de Reuver PR, Gill P, Andrici J, D’Urso L, Mittal A, Pavlakis N, Clarke S, Samra JS, Gill AJ, 2015, Somatostatin Receptor SSTR-2a Expression is a Stronger Predictor for Survival Than Ki-67 in Pancreatic Neuroendocrine Tumors. Medicine, Baltimore, 94:e1281 Wang Y, Wang W, Jin K, Fang C, Lin Y, Xue L, Feng S, Zhou Z, Shao C, Chen M, Yu X, Chen J, 2017, Somatostatin receptor expression indicates improved prognosis in gastroenteropancreatic neuroendocrine neoplasms, and octreotide long-acting release is effective and safe in Chinese patients with advanced gastroenteropancreatic neuroendocrine tumors. Oncol Lett 13: 1165–1174 Qian ZR, Li T, Ter-Minassian M, Yang J, Chan JA, Brais LK, Masugi Y, Thiaglingam A, Brooks N, Nishihara R, Bonnemarie M, Masuda A, Inamura K, Kim SA, Mima K, Sukawa Y, Dou R, Lin X, Christiani DC, Schmidlin F, Fuchs CS, Mahmood U, Ogino S, Kulke MH, 2016, Association Between Somatostatin Receptor Expression and Clinical Outcomes in Neuroendocrine Tumors. Pancreas 45:1386–1393 Brunner P, Jorg AC, Glatz K, Bubendorf L, Radojewski P, Umlauft M, Marincek N, Spanjol PM, Krause T, Dumont RA, Maecke HR, Muller-Brand J, Briel M, Schmitt A, Perren A, Walter MA, 2017, The pr o gn o s t i c a n d pr e d i c t i v e va l u e o f s s t r 2 - immunohistochemistry and sstr2-targeted imaging in neuroendocrine tumors. Eur J Nucl Med Mol Imaging 44:468–475 Liu Q, Cescato R, Dewi DA, Rivier J, Reubi JC, Schonbrunn A, 2005, Receptor signaling and endocytosis are differentially regulated by somatostatin analogs. Mol Pharmacol 68:90–101 Pisarek H, Pawlikowski M, Kunert-Radek J, Winczyk K, 2010, Does the response of GH-secreting pituitary adenomas to octreotide depend on the cellular localization of the somatostatin receptor subtypes SSTR2 and SSTR5? Endokrynol Pol 61:178–181 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 521 EP 525 DI 10.1007/s12253-018-0426-4 PG 5 ER PT J AU Wang, L Aireti, A Aihaiti, A Li, K AF Wang, Lei Aireti, Ailixiati Aihaiti, Aizezi Li, Kun TI Expression of microRNA-150 and its Target Gene IGF2BP1 in Human Osteosarcoma and their Clinical Implications SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; microRNA-150; Insulin-like growth factor 2 mRNA-binding protein 1; Clinicopathological feature; Overall survival; Disease-free survival ID Osteosarcoma; microRNA-150; Insulin-like growth factor 2 mRNA-binding protein 1; Clinicopathological feature; Overall survival; Disease-free survival AB Previous study revealed that microRNA (miR)-150 might function as a tumor suppressor in osteosarcoma partially by targeting Insulin-Like Growth Factor 2 mRNA-Binding Protein 1 (IGF2BP1). The aim of this study was to investigate the clinical significance of miR-150-IGF2BP1 axis in human osteosarcoma which remains unclear. At first, expression levels of miR-150, and IGF2BP1 mRNA and protein in 20 osteosarcoma and matched adjacent noncancerous tissues were respectively detected by quantitative real-time PCR and western blot analyses. Then, subcellular localization and expression pattern of IGF2BP1 protein in 100 osteosarcoma tissues were examined by immunohistochemistry. Associations of miR-150/IGF2BP1 expression with various clinicopathological features and patients’ prognosis were also statistically evaluated. As a result, miR-150 expression was significantly decreased, while IGF2BP1 mRNA and protein expressionwere dramatically increased in osteosarcoma tissues compared to matched adjacent noncancerous tissues (all P < 0.001). Immunostaining of IGF2BP1 protein was localized in cytoplasm of tumor cells in osteosarcoma tissues. Statistically, low miR-150 expression and/or high IGF2BP1 protein immunoreactive score were all significantly associated with high tumor grade, presence of metastasis and recurrence, as well as poor response to chemotherapy (all P < 0.05). Moreover, miR-150, IGF2BP1 and combined miR-150/IGF2BP1 expressions were all identified as independent prognostic factors for overall and disease-free survivals of osteosarcoma patients (all P < 0.05). In conclusion, our data suggest that miR-150 and its downstream target IGF2BP1 may be a crucial axis for the development, progression and patients’ prognosis of ostesarcoma. The newly identified miR-150/IGF2BP1 axismight be a novel potential therapeutic target for osteosarcoma treatment. C1 [Wang, Lei] People’s Hospital of Xinjiang Uygur Autonomous Region, Department of Orthopedics Center Spine Surgery, 830000 Urumqi, China. [Aireti, Ailixiati] People’s Hospital of Xinjiang Uygur Autonomous Region, Department of Orthopedics Center Spine Surgery, 830000 Urumqi, China. [Aihaiti, Aizezi] People’s Hospital of Xinjiang Uygur Autonomous Region, Department of Orthopedics Center Spine Surgery, 830000 Urumqi, China. [Li, Kun] People’s Hospital of Xinjiang Uygur Autonomous Region, Department of Orthopedics Center Spine Surgery, 830000 Urumqi, China. RP Wang, L (reprint author), People’s Hospital of Xinjiang Uygur Autonomous Region, Department of Orthopedics Center Spine Surgery, 830000 Urumqi, China. EM wanglei20170311@qq.com CR Fagioli F, Biasin E, Mereuta OM, Muraro M, Luksch R, Ferrari S, Aglietta M, Madon E, 2008, Poor prognosis osteosarcoma: new therapeutic approach. Bone Marrow Transplant 41(Suppl 2): S131–S134 Bieling P, Rehan N, Winkler P, Helmke K, Maas R, Fuchs N, Bielack S, Heise U, Jurgens H, Treuner J, Romanowski R, Exner U, Kotz R, Winkler K, 1996, Tumor size and prognosis in aggressively treated osteosarcoma. J Clin Oncol 14:848–858 Bielack SS, Hecker-Nolting S, Blattmann C, Kager L, 2016, Advances in the management of osteosarcoma. F1000Res 5:2767 AndersonME, 2016, Update on Survival in Osteosarcoma. Orthop Clin North Am 47:283–292 Piletic K, Kunej T, 2016, MicroRNA epigenetic signatures in human disease. Arch Toxicol 90:2405–2419 Rupaimoole R, Slack FJ, 2017, MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov 16:203–222 Shah MY, Ferrajoli A, Sood AK, Lopez-Berestein G, Calin GA, 2016, microRNA therapeutics in Cancer-an emerging concept. EBioMedicine 12:34–42 Makarova JA, ShkurnikovMU,Wicklein D, Lange T, Samatov TR, Turchinovich AA, Tonevitsky AG, 2016, Intracellular and extracellular microRNA: an update on localization and biological role. Prog Histochem Cytochem 51:33–49 Ram KumarRM, Boro A, Fuchs B, 2016, Involvement and clinical aspects of MicroRNA in osteosarcoma. Int J Mol Sci 17:E877 Kuninty PR, Schnittert J, Storm G, Prakash J, 2016, MicroRNA targeting to modulate tumor microenvironment. Front Oncol 6:3 Vasilatou D, Papageorgiou S, Pappa V, Papageorgiou E, Dervenoulas J, 2010, The role of microRNAs in normal and malignant hematopoiesis. Eur J Haematol 84:1–16 Wang F, Ren X, Zhang X, 2015, Role of microRNA-150 in solid tumors. Oncol Lett 10:11–16 Li X, Chen L, Wang W, Meng FB, Zhao RT, Chen Y, 2015, MicroRNA-150 inhibits cell invasion and migration and is downregulated in human osteosarcoma. Cytogenet Genome Res 146: 124–135 Zhan C, Li C, Zhang H, Tang H, Ji F, Qiao SC, XuWD,Wang ZW, 2016, MicroRNA-150 upregulation reduces osteosarcoma cell invasion and metastasis by downregulating Ezrin. Oncol Lett 12: 3457–3462 Wang L, Wang W, Li J, Chen S, Zhan R, 2016, MicroRNA-150 inhibits osteosarcoma cell proliferation by targeting RUNX2 gene. Zhong Nan Da Xue Xue Bao Yi Xue Ban 41:1285–1290 Qu Y, Pan S, Kang M, Dong R, Zhao J, 2016, MicroRNA-150 functions as a tumor suppressor in osteosarcoma by targeting IGF2BP1. Tumour Biol 37:5275–5284 Ning K, Zhang H,Wang Z, Li K, 2017, Prognostic implications of Kindlin proteins in human osteosarcoma. Onco Targets Ther 10: 657–665 Zhao J, Xu H, He M, Wu Y, 2014, Glucocorticoid receptor DNA binding factor 1 expression and osteosarcoma prognosis. Tumour Biol 35:12449–12458 Liao Y, Feng Y, Shen J, Gao Y, Cote G, Choy E, Harmon D, Mankin H, Hornicek F, Duan Z, 2016, Clinical and biological significance of PIM1 kinase in osteosarcoma. J Orthop Res 34: 1185–1194 Abd El-Rehim DM, Osman NA, 2015, Expression of a disintegrin and metalloprotease 8 and endostatin in human osteosarcoma: implication in tumor progression and prognosis. J Egypt Natl Canc Inst 27:1–9 Watanabe A, Tagawa H, Yamashita J, Teshima K, Nara M, Iwamoto K, Kume M, Kameoka Y, Takahashi N, Nakagawa T, Shimizu N, Sawada K, 2011, The role of microRNA-150 as a tumor suppressor in malignant lymphoma. Leukemia 25:1324– 1334 Sakr M, Takino T, Sabit H, Nakada M, Li Z, Sato H, 2016, miR- 150-5p and miR-133a suppress glioma cell proliferation and migration through targeting membrane-type-1 matrix metalloproteinase. Gene 587:155–162 WangX, Ren Y,Wang Z, Xiong X, Han S, PanW, Chen H, Zhou L, Zhou C, Yuan Q, Yang M, 2015, Down-regulation of 5S rRNA by miR-150 and miR-383 enhances c-Myc-rpL11 interaction and inhibits proliferation of esophageal squamous carcinoma cells. FEBS Lett 589(24 Pt B):3989–3997 Huang S, Chen Y,WuW, Ouyang N, Chen J, Li H, Liu X, Su F, Lin L, Yao Y, 2013, miR-150 promotes human breast cancer growth and malignant behavior by targeting the pro-apoptotic purinergic P2X7 receptor. PLoS One 8:e80707 Li H, Ouyang R,Wang Z, ZhouW, ChenH, JiangY, ZhangY, Li H, Liao M, Wang W, Ye M, Ding Z, Feng X, Liu J, Zhang B, 2016, MiR-150 promotes cellular metastasis in non-small cell lung cancer by targeting FOXO4. Sci Rep 6:39001 Wu Q, Jin H, Yang Z, Luo G, Lu Y, Li K, Ren G, Su T, Pan Y, Feng B, Xue Z,Wang X, Fan D, 2010)MiR-150 promotes gastric cancer proliferation by negatively regulating the pro-apoptotic gene EGR2. Biochem Biophys Res Commun 392:340–345 Wang S, Yin J, Li T, Yuan L, Wang D, He J, Du X, Lu J, 2015, Upregulated circulating miR-150 is associated with the risk of intrahepatic cholangiocarcinoma. Oncol Rep 33:819–825 Li T, Xie J, Shen C, Cheng D, Shi Y,Wu Z, Zhan Q, Deng X, Chen H, Shen B, Peng C, Li H, Zhu Z, 2014, miR-150-5p inhibits hepatoma cell migration and invasion by targetingMMP14. PLoS One 9:e115577 Yang K, He M, Cai Z, Ni C, Deng J, Ta N, Xu J, Zheng J, 2015, A decrease in miR-150 regulates the malignancy of pancreatic cancer by targeting c-Myb and MUC4. Pancreas 44:370–379 Aherne ST, Madden SF, Hughes DJ, Pardini B, Naccarati A, Levy M, Vodicka P, Neary P, Dowling P, Clynes M, 2015, Circulating miRNAs miR-34a and miR-150 associated with colorectal cancer progression. BMC Cancer 15:329 Li J, Hu L, Tian C, Lu F, Wu J, Liu L, 2015, microRNA-150 promotes cervical cancer cell growth and survival by targeting FOXO4. BMC Mol Biol 16:24 Wuerkenbieke D, Wang J, Li Y, Ma C, 2015, miRNA-150 downregulation promotes pertuzumab resistance in ovarian cancer cells via AKT activation. Arch Gynecol Obstet 292:1109–1116 Liu DZ, Zhang HY, Long XL, Zou SL, ZhangXY, HanGY, Cui ZG, 2015, MIR-150 promotes prostate cancer stem cell development via suppressing p27Kip1. Eur Rev Med Pharmacol Sci 19:4344– 4352 Yisraeli JK, 2005, VICKZ proteins: a multi-talented family of regulatory RNA-binding proteins. Biol Cell 97:87–96 Yaniv K, Yisraeli JK, 2002, The involvement of a conserved family of RNA binding proteins in embryonic development and carcinogenesis. Gene 287:49–54 Gutschner T, Hammerle M, Pazaitis N, Bley N, Fiskin E, Uckelmann H, Heim A, Groβ M, Hofmann N, Geffers R, Skawran B, Longerich T, Breuhahn K, Schirmacher P, Muhleck B, Huttelmaier S, Diederichs S, 2014, Insulin-like growth factor 2 mRNA-binding protein 1, IGF2BP1, is an important protumorigenic factor in hepatocellular carcinoma. Hepatology 59:1900–1911 Su Y, Xiong J, Hu J, Wei X, Zhang X, Rao L, 2016, MicroRNA- 140-5p targets insulin like growth factor 2 mRNA binding protein 1, IGF2BP1, to suppress cervical cancer growth and metastasis. Oncotarget 7:68397–68411 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 527 EP 533 DI 10.1007/s12253-018-0454-0 PG 7 ER PT J AU Szasz, R Altai, E Pal, K Dombi, P Ivanyi, J Jakucs, J Joni, N Illes, Tarkanyi, I Szerafin, L Nagy, Zs Farkas, P Nagy, Piukovics, K Ujj, Gy Schneider, T AF Szasz, Robert Altai, Elvira Pal, Katalin Dombi, Peter Ivanyi, Janos Jakucs, Janos Joni, Natalia Illes, Arpad Tarkanyi, Ilona Szerafin, Laszlo Nagy, Zsolt Farkas, Peter Nagy, Agnes Piukovics, Klara Ujj, Gyorgy Schneider, Tamas TI Effectiveness of the Combination of Rituximab and Standard Chemotherapeutic Regimens in Previously Untreated Patients with Chronic Lymphocytic Leukaemia in Real-Life: Results from a Noninterventional Study (CILI Study) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chronic lymphocytic leukemia; Rituximab; Overall response rate ID Chronic lymphocytic leukemia; Rituximab; Overall response rate AB Chronic lymphocytic leukemia (CLL) is one of the most common haematological malignancies exhibiting remarkable heterogeneity in clinical course. Rituximab added to standard chemotherapy has been proven to increase response rate and eventually survival among previously untreated CLL patients. CILI was an open-label, non-randomized, single arm, multicentric, observational study aimed to collect real-life effectiveness data for rituximab used according to the current label in combination with standard chemotherapy in previously untreated CLL patients. Overall response rates (ORR) in the entire study population as well as in various subgroups were estimated. Adverse events were recorded during the entire course of the study. A total number of 150 patients were enrolled by 15 Hungarian study sites. Out of these, 82 patients received 6 cycles of rituximab containing treatment. Overall response rates of 88.24% (CI95%: 81.6–93.12%) and 94.59% (CI95%: 86.73–98.51%) were recorded in the intent-to-treat (ITT) and per-protocol (PP) populations, respectively. In both study populations, somewhat higher ORR was observed in patients aged ≥65 years. Subgroups defined according to either chromosomal aberrations (presence of 11q and 17p deletions) showed apparently high ORRs, though these rates were most probably biased by low patient numbers. 144 adverse events were reported during the study, of which 15 AEs were considered to be related to the administration of rituximab. Analyses of the efficacy variables have revealed comparable results to those previously reported by controlled clinical trials. C1 [Szasz, Robert] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Altai, Elvira] Veszprem County Csolnoky Ferenc Hospital, Korhaz u. 1, 8200 Veszprem, Hungary. [Pal, Katalin] Fejer County Szent Gyorgy Hospital, Seregelyesi ut 3, 8000 Szekesfehervar, Hungary. [Dombi, Peter] Szent Borbala Hospital, Department of Hematology, Dozsa Gyorgy ut 77, 2800 Tatabanya, Hungary. [Ivanyi, Janos] Vas County Markusovszky Hospital, Department of Hematology, Markusovszky L. u. 5, 9700 Szombathely, Hungary. [Jakucs, Janos] Bekes County Pandy Kalman Hospital, Department of Internal Medicine, Semmelweis u. 1, 5700 Gyula, Hungary. [Joni, Natalia] Ferenc Markhot County Hospital, Szechenyi u. 27-29, 3300 Eger, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Tarkanyi, Ilona] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor u. 2/A, 1083 Budapest, Hungary. [Szerafin, Laszlo] Szabolcs-Szatmar-Bereg County Josa Andras Hospital, Department of Haematology, Szent Istvan u. 68, 4400 Nyiregyhaza, Hungary. [Nagy, Zsolt] Borsod-Abauj-Zemplen County Hospital, 2nd Department of Internal Medicine, Csabai kapu 9-11, 3529 Miskolc, Hungary. [Farkas, Peter] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4, 1125 Budapest, Hungary. [Nagy, Agnes] University of Pecs, I. Department of Internal Medicine, Ifjusag ut 13, 7624 Pecs, Hungary. [Piukovics, Klara] University of Szeged, 2nd Department of Internal Medicine and Cardiology Centre, Koranyi Fasor 6, 6720 Szeged, Hungary. [Ujj, Gyorgy] Jasz-Nagykun-Szolnok County Hetenyi Geza Hospital, I. Department of Internal Medicine, Toszegi ut 21, 5004 Szolnok, Hungary. [Schneider, Tamas] National Institute of Oncology, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary. RP Szasz, R (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, 4032 Debrecen, Hungary. EM szaszr@med.unideb.hu CR Yu EM, Kittai A, Tabbara IA, 2015, Chronic lymphocytic leukemia: current concepts. Anticancer Res 35(10):5149–5165 Matutes E, Polliack A, 2000, Morphological and immunophenotypic features of chronic lymphocytic leukemia. Rev Clin Exp Hematol 4(1): 22–47 Binet JL, Auquier A, Dighiero G et al, 1981, A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis. Cancer 48:198–206 Rai KR, Sawitsky A, Cronkite EP et al, 1975, Clinical staging of chronic lymphocytic leukemia. Blood 46:219–234 Baliakas P, Mattsson M, Stamatopoulos K et al, 2015, Prognostic indices in chronic lymphocytic leukaemia: where do we stand how do we proceed? J Intern Med 279(4):347–357 Rossi D, Terzi-di-Bergamo L, De Paoli L et al, 2015, Molecular prediction of durable remission after first-line fludarabinecyclophosphamide- rituximab in chronic lymphocytic leukemia. Blood 126(16):1921–1924 Eichhorst BF, Busch R, Hopfinger G et al, 2006, Fludarabine plus cyclophosphamide versus fludarabine alone in first-line therapy of younger patients with chronic lymphocytic leukemia. Blood 107: 885–891 Flinn IW, Neuberg DS, Grever MR et al, 2007, Phase III trial of fludarabine plus cyclophosphamide compared with fludarabine for patients with previously untreated chronic lymphocytic leukemia: US intergroup trial E2997. J Clin Oncol 25:793–798 Byrd JC, Peterson BL, MorrisonVAet al, 2003, Randomized phase 2 study of fludarabine with concurrent versus sequential treatment with rituximab in symptomatic, untreated patients with B-cell chronic lymphocytic leukemia: results from Cancer and leukemia group B 9712, CALGB 9712). Blood 101(1):6–14 Cramer P, Isfort S, Bahlo J et al, 2015, Outcome of advanced chronic lymphocytic leukemia following different first-line and relapse therapies: a meta-analysis of five prospective trials by the German CLL study group, GCLLSG). Haematologica 100(11):1451–1459 Foa R, Del Giudice I, Cuneo A et al, 2014, Chlorambucil plus rituximab with or without maintenance rituximab as first-line treatment for elderly chronic lymphocytic leukemia patients. Am J Hematol 89(5):480–486 Goede V, Fischer K, Busch R et al, 2014, Obinutuzumab plus Chlorambucil in patients with CLL and coexisting conditions. N Engl J Med 370:1101–1110 Tedder TF, Engel P, 1994, CD20: a regulator of cell-cycle progression of B lymphocytes. Immunol Today 15:450–454 Reff ME, Carner K, Chambers KS, 1994, Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood 83:435–445 Maloney D, Smith B, Appelbaum F, 1996, The anti-tumor effect of monoclonal anti-CD20 antibody, MAB, therapy includes direct anti-proliferative activity and induction of apoptosis in CD20 positive non-Hodgkin’s lymphoma, NHL, cell lines. Blood 637a:88 Maloney DG, Liles TM, Czerwinski DK et al, 1994, Phase I clinical trial using escalating single-dose infusion of chimeric anti- CD20 monoclonal antibody, IDEC-C2B8, in patients with recurrent B-cell lymphoma. Blood 84:2457–2466 Eichhorst B, Robak T, Montserrat E et al, 2015, Chronic lymphocytic leukaemia: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 26(Suppl 5):v78–v84 Thurmes P, Call T, Slager S et al, 2009, Comorbid conditions and survival in unselected, newly diagnosed patients with chronic lymphocytic leukemia. Leuk Lymphoma 49:49–56 Jain N, O’Brian S, 2015, Initial treatment of CLL: integrating biology and functional status. Blood 126:463–470 HallekM(2017, Chronic lymphocytic leukemia: 2017 update on diagnosis, risk stratification, and treatment. Am J Hematol 92(9):946–965 Schulz H, Klein SK, Rehwald U, 2002, Phase 2 study of a combined immunochemotherapy using rituximab and fludarabine in patients with chronic lymphocytic leukemia. Blood 100(9):3115–3120 Fischer K, Cramer P, Busch R et al, 2012, Bendamustine in combination with rituximab for previously untreated patients with chronic lymphocytic leukemia: a multicenter phase II trial of the German chronic lymphocytic leukemia study group. J Clin Oncol 30(26):3209–3216 Keating MJ, O'Brien S, Albitar M et al, 2005, Early results of a chemoimmunotherapy regimen of fludarabine, cyclophosphamide, and rituximab as initial therapy for chronic lymphocytic leukemia. J Clin Oncol 23(18):4079–4088 Hallek M, Fischer K, Fingerle-Rowson G et al, 2010, Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial. Lancet 376(9747):1164–1174 Foon KA, Boyiadzis M, Land SR et al, 2009, Chemoimmunotherapy with low-dose fludarabine and cyclophosphamide and high dose rituximab in previously untreated patients with chronic lymphocytic leukemia. J Clin Oncol 27(4):498–503 Dohner H, Stilgenbauer S, Benner A et al, 2000, Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med 343(26):1910–1916 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 535 EP 540 DI 10.1007/s12253-018-0474-9 PG 6 ER PT J AU Niu, Y Yang, X Chen, Y Zhang, L Jin, X Tang, Y Li, L Yu, L Guo, Y Wang, H AF Niu, Yuna Yang, Xue Chen, Yifei Zhang, Linbo Jin, Xinyue Tang, Youjing Li, Li Yu, Lu Guo, Yilin Wang, Hui TI BCL3 Expression Is a Potential Prognostic and Predictive Biomarker in Acute Myeloid Leukemia of FAB Subtype M2 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BCL3; Expression; Prognosis; Acute myeloid leukemia ID BCL3; Expression; Prognosis; Acute myeloid leukemia AB Although the implication of BCL3 has been disclosed in human chronic lymphocytic leukemia as well as other solid tumors, the diagnostic and prognostic of BCL3 expression in acute myeloid leukemia (AML) remains largely unclear. In this study, we isolated total RNA from bone marrow mononuclear cells collected from 101 de novo AML patients and 27 healthy donors. After reverse transcription, quantitative real-time PCR was performed to detect BCL3 expression level. BCL3 mRNA level was significantly down-regulated in BMMCs of AML patients compared with healthy controls (P = 0.0015). BCL3 was showed a higher level in AML patients with poor-risk karyotypes than that of in patients with favorable/intermediate-risk karyotypes (P = 0.014). ROC analysis demonstrated that BCL3 could effectively differentiate AML patients from normal controls. Among the French-American-British (FAB) subtypes, the frequency of low BCL3 expression in M2 subtypes is significantly higher than that of in the other subtypes M1/M4/M5/M6/M7 (P = 0.006), and mildly lower in myelomonocytic/monocytic subtypes M4/M5 (P = 0.064) than those in M1/M2/M6/M7 subtypes. Chromosome analysis revealed that BCL3low patients had a remarkably higher frequency of t (8;21) abnormality (P = 0.0047) and lower frequency of normal karyotype (P = 0.0059) than BCL3high patients. BCL3high patients showed a significantly higher frequency of FLT3-ITD mutation (P = 0.028) and lower frequency of C-Kit mutation (P = 0.0232) than BCL3low patients. Although there were no significant differences in complete remission and overall survival between BCL3low and BCL3high groups, patients with high BCL3 expression markedly shorter overall survival (OS, P = 0.049), relapse-free survival (RFS, P = 0.027) and disease-free survival (DFS, P = 0.042) in M2AML than low BCL3 expression patients. Additionally, in AMLs of M2 subtype, high BCL3 expression patients had markedly lower complete remission (CR) rate (P = 0.0317) after the second induction treatment than patientswith BCL3 low expression. Thus, these findings indicated that BCL3 appeared as a promising molecular biomarker of pediatric acute myeloid leukemia with unfavorable prognosis. C1 [Niu, Yuna] Xinxiang Medical University, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, 453003 Xinxiang, Henan, China. [Yang, Xue] Xinxiang Medical University, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, 453003 Xinxiang, Henan, China. [Chen, Yifei] Xinxiang Medical University, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, 453003 Xinxiang, Henan, China. [Zhang, Linbo] Xinxiang Medical University, the Third Affiliated Hospital, Department of Laboratory Medicine, 453003 Xinxiang, Henan, China. [Jin, Xinyue] Xinxiang Medical University, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, 453003 Xinxiang, Henan, China. [Tang, Youjing] Xinxiang Medical University, the First Affiliated Hospital, Laboratory of HematologyWeihui, Henan, China. [Li, Li] Xinxiang Medical University, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, 453003 Xinxiang, Henan, China. [Yu, Lu] Xinxiang Medical University, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, 453003 Xinxiang, Henan, China. [Guo, Yilin] Xinxiang Medical University, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, 453003 Xinxiang, Henan, China. [Wang, Hui] Xinxiang Medical University, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, 453003 Xinxiang, Henan, China. RP Wang, H (reprint author), Xinxiang Medical University, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, 453003 Xinxiang, China. EM huiwang65@yeah.net CR de Rooij JD, Zwaan CM, van den Heuvel-Eibrink M, 2015, Pediatric AML: From Biology to Clinical Management. Journal of clinical medicine 4(1):127–149. , DOI 10.3390/ jcm4010127 RascheM, Zimmermann M, Borschel L, Bourquin JP, Dworzak M, Klingebiel T, Lehrnbecher T, Creutzig U, Klusmann JH, Reinhardt D, 2018, Successes and challenges in the treatment of pediatric acute myeloid leukemia: a retrospective analysis of the AMLBFM trials from 1987 to 2012. Leukemia. , DOI 10.1038/ s41375-018-0071-7 Yu MG, Zheng HY, (2017), Acute Myeloid Leukemia: Advancements in Diagnosis and Treatment. Chinese medical journal 130(2):211–218. , DOI 10.4103/0366-6999.198004 Dombret H, Gardin C, 2016, An update of current treatments for adult acute myeloid leukemia. Blood 127(1):53–61. , DOI 10.1182/blood-2015-08-604,520 Ohno H, Doi S, Yabumoto K, Fukuhara S, McKeithan TW, 1993, Molecular characterization of the t(14;19)(q32;q13, translocation in chronic lymphocytic leukemia. Leukemia 7(12):2057–2063 Martin-Subero JI, Wlodarska I, Bastard C, Picquenot JM, Hoppner J, Giefing M, Klapper W, Siebert R, 2006, Chromosomal rearrangements involving the BCL3 locus are recurrent in classical Hodgkin and peripheral T cell lymphoma. Blood 108(1):401–402; author reply 402–403. , DOI 10.1182/blood-2005-09-3843 Mottok A, Steidl C, 2018, Biology of classical Hodgkin lymphoma: implications for prognosis and novel therapies. Blood 131(15): 1654–1665. , DOI 10.1182/blood-2017-09-772,632 Au WY, Horsman DE, Ohno H, Klasa RJ, Gascoyne RD, 2002, Bcl-3/IgH translocation, 14;19)(q32;q13, in non-Hodgkin’s lymphomas. Leukemia & lymphoma 43(4):813–816. , DOI 10.1080/10428190290016935 Wakefield A, Soukupova J, Montagne A, Ranger J, French R, Muller WJ, Clarkson RW, 2013, Bcl3 selectively promotes metastasis of ERBB2-driven mammary tumors. Cancer research 73(2): 745–755. , DOI 10.1158/0008-5472.CAN-12-1321 Chung GT, Lou WP, Chow C, To KF, Choy KW, Leung AW, Tong CY, Yuen JW, Ko CW, Yip TT, Busson P, Lo KW, 2013, Constitutive activation of distinct NF-kappaB signals in EBVassociated nasopharyngeal carcinoma. The Journal of pathology 231(3):311–322. , DOI 10.1002/path.4239 Zhao H, Wang W, Zhao Q, Hu G, Deng K, Liu Y, 2016, BCL3 exerts an oncogenic function by regulating STAT3 in human cervical cancer. OncoTargets and therapy 9:6619–6629. , DOI 10.2147/OTT.S118184 Dimitrakopoulos FI, Antonacopoulou AG, Kottorou A, Marousi S, Koukourikou I, Kalofonou M, Panagopoulos N, Scopa C, Dougenis D, Papadaki H, Papavassiliou AG, Kalofonos HP, 2015, Variant of BCL3 gene is strongly associated with five-year survival of non-small-cell lung cancer patients. Lung cancer 89(3): 311–319. , DOI 10.1016/j.lungcan.2015.06.006 Dai J, Lu Y, Wang J, Yang L, Han Y, Wang Y, Yan D, Ruan Q, Wang S, 2016, A four-gene signature predicts survival in clear-cell renal-cell carcinoma. Oncotarget 7, 50):82712–82,726. https://doi. org/10.18632/oncotarget.12631 Nguyen-Khac F, Chapiro E, Lesty C, Grelier A, Luquet I, Radford- Weiss I, Lefebvre C, Fert-Ferrer S, Callet-Bauchu E, Lippert E, Raggueneau V, Michaux L, Barin C, Collonge-Rame MA, Mugneret F, Eclache V, Taviaux S, Dastugue N, Richebourg S, Struski S, Talmant P, Baranger L, Gachard N, Gervais C, Quilichini B, Settegrana C, Maloum K, Davi F, Merle-Beral H, 2011, Specific chromosomal IG translocations have different prognoses in chronic lymphocytic leukemia. American journal of blood research 1(1):13–21 Schulten HJ, BangashM, KarimS, Dallol A,Hussein D, Merdad A, Al-Thoubaity FK, Al-Maghrabi J, Jamal A, Al-Ghamdi F, Choudhry H, Baeesa SS, Chaudhary AG, Al-Qahtani MH, 2017, Comprehensive molecular biomarker identification in breast cancer brain metastases. Journal of translational medicine 15(1):269. , DOI 10.1186/s12967-017-1370-x Nishida Y, Takeuchi K, Tsuda K, Ugai T, Sugihara H, Yamakura M, Takeuchi M, Matsue K, 2013, Acquisition of t(11;14, in a patient with chronic lymphocytic leukemia carrying both t(14;19)(q32;q13.1, and + 12. European journal of haematology 91(2):179–182. , DOI 10.1111/ejh.12119 Alpatov R, Carstens B, Harding K, Jarrett C, Balakhani S, Lincoln J, Brzeskiewicz P, Guo Y, Ohene-Mobley A, LeRoux J, McDaniel V, Meltesen L, Minka D, Patel M, Manavi C, Swisshelm K, 2015, Rare double-hit with two translocations involving IGH both, with BCL2 and BCL3, in a monoclonal B cell lymphoma/leukemia. Molecular cytogenetics 8:101. , DOI 10.1186/s13039- 015-0203-y Martin-Subero JI, Ibbotson R, Klapper W, Michaux L, Callet- Bauchu E, Berger F, Calasanz MJ, De Wolf-Peeters C, Dyer MJ, Felman P, Gardiner A, Gascoyne RD, Gesk S, Harder L, Horsman DE, Kneba M, Kuppers R, Majid A, Parry-Jones N, Ritgen M, Salido M, Sole F, Thiel G, Wacker HH, Oscier D, Wlodarska I, Siebert R, 2007, A comprehensive genetic and histopathologic analysis identifies two subgroups of B cell malignancies carrying a t(14;19)(q32;q13, or variant BCL3-translocation. Leukemia 21(7):1532–1544. , DOI 10.1038/sj.leu.2404695 NishikoriM, Maesako Y, Ueda C, KurataM, Uchiyama T, Ohno H, 2003, High-level expression of BCL3 differentiates t(2;5)(p23;q35)-positive anaplastic large cell lymphoma from Hodgkin disease. Blood 101(7):2789–2796. , DOI 10. 1182/blood-2002-08-2464 Slovak ML, Bedell V, Hsu YH, Estrine DB, Nowak NJ, Delioukina ML, Weiss LM, Smith DD, Forman SJ, 2011, Molecular karyotypes of Hodgkin and Reed-Sternberg cells at disease onset reveal distinct copy number alterations in chemosensitive versus refractory Hodgkin lymphoma. Clinical cancer research: an official journal of the American Association for Cancer Research 17(10):3443– 3454. , DOI 10.1158/1078-0432.CCR-10-1071 Chang TP, Vancurova I, 2014, Bcl3 regulates pro-survival and proinflammatory gene expression in cutaneous T cell lymphoma. Biochimica et biophysica acta 1843(11):2620–2630. https://doi. org/10.1016/j.bbamcr.2014.07.012 Ibrahim HA, Amen F, Reid AG, Naresh KN, 2011, BCL3 rearrangement, amplification and expression in diffuse large B cell lymphoma. European journal of haematology 87(6):480–485. , DOI 10.1111/j.1600-0609.2011.01684.x Saamarthy K, Bjorner S, Johansson M, Landberg G, Massoumi R, Jirstrom K, Masoumi KC, 2015, Early diagnostic value of Bcl-3 localization in colorectal cancer. BMC cancer 15:341. https://doi. org/10.1186/s12885-015-1342-6 Kreisel D, Sugimoto S, Tietjens J, Zhu J, Yamamoto S, Krupnick AS, Carmody RJ, Gelman AE, 2011, Bcl3 prevents acute inflammatory lung injury in mice by restraining emergency granulopoiesis. The Journal of clinical investigation 121(1):265– 276. , DOI 10.1172/JCI42596 Croker BA, Metcalf D, Robb L,WeiW,Mifsud S, DiRago L, Cluse LA, Sutherland KD, Hartley L, Williams E, Zhang JG, Hilton DJ, Nicola NA, Alexander WS, Roberts AW(2004, SOCS3 is a critical physiological negative regulator of G-CSF signaling and emergency granulopoiesis. Immunity 20(2):153–165 Strauss L, Sangaletti S, Consonni FM, Szebeni G, Morlacchi S, Totaro MG, Porta C, Anselmo A, Tartari S, Doni A, Zitelli F, Tripodo C, Colombo MP, Sica A, 2015, RORC1 Regulates Tumor-Promoting BEmergency^ Granulo-Monocytopoiesis. Cancer cell 28(2):253–269 , DOI 10.1016/j.ccell.2015.07. 006 Weyrich AS, Denis MM, Schwertz H, Tolley ND, Foulks J, Spencer E, Kraiss LW, Albertine KH, McIntyre TM, Zimmerman GA(2007, mTOR-dependent synthesis of Bcl-3 controls the retraction of fibrin clots by activated human platelets. Blood 109(5): 1975–1983. , DOI 10.1182/blood-2006-08-042192 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 541 EP 548 DI 10.1007/s12253-018-0476-7 PG 8 ER PT J AU Koeneman, MM Ovestad, TI Janssen, AME Ummelen, M Kruitwagen, FPMR Hopman, HA Kruse, JA AF Koeneman, M Margot Ovestad, T Irene Janssen, A M Emiel Ummelen, Monique Kruitwagen, F P M Roy Hopman, H Anton Kruse, J Arnold TI Gain of Chromosomal Region 3q26 as a Prognostic Biomarker for High-Grade Cervical Intraepithelial Neoplasia: Literature Overview and Pilot Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE hTERC; 3q26; Cervical intraepithelal neoplasia; Prognosis; Biomarker ID hTERC; 3q26; Cervical intraepithelal neoplasia; Prognosis; Biomarker AB Approximately 20–40% of high-grade Cervical Intraepithelial Neoplasia (CIN) regresses spontaneously, but the natural prognosis of an individual lesion is unpredictable. Gain of the chromosomal 3q region, which contains the human telomerase RNA gene on 3q26, is found in CIN lesions and cervical carcinoma and shows correlation with disease grade. The aim of this study is to assess whether 3q26 gain as a single genetic marker can predict the natural prognosis of highgrade CIN, by performing a review of the literature and pilot study. A literature review was conducted. Additionally, we performed a pilot study in 19 patients with histologically confirmed high-grade CIN lesions who were followed for a mean of 115 days, after which loop excision was performed. Fluorescent in situ hybridization analysis was performed on the initial diagnostic biopsies to determine gain of 3q26. Eight studies were included in the literature overview, with a total of 407 patients. Of these, only 22 patients had high-grade lesions. All studies found an association between 3q26 gain and disease prognosis. Positive predictive values (PPV) ranged from 50 to 93%, negative predictive values (NPV) ranged from 75 to 100%. Only five out of 155 patients (3.2%) without 3q26 gain showed disease persistence or progression. In our pilot study on 3q26 gain in high-grade CIN, the PPV of 3q26 gain for disease persistence was 67%, the NPV 100%. All four patients without 3q26 gain showed disease regression. In conclusion, the absence of 3q26 gain in diagnostic biopsies may be applied to identify high-grade CIN lesions with a high probability of disease regression. C1 [Koeneman, M Margot] Maastricht University, Medical Centre, Department of Obstetrics and GynecologyMaastricht, The Netherlands. [Ovestad, T Irene] Stavanger University Hospital, Department of PathologyStavanger, Norway. [Janssen, A M Emiel] Stavanger University Hospital, Department of PathologyStavanger, Norway. [Ummelen, Monique] Maastricht University, GROW - School for Oncology and Developmental BiologyMaastricht, The Netherlands. [Kruitwagen, F P M Roy] Maastricht University, Medical Centre, Department of Obstetrics and GynecologyMaastricht, The Netherlands. [Hopman, H Anton] Maastricht University, GROW - School for Oncology and Developmental BiologyMaastricht, The Netherlands. [Kruse, J Arnold] Maastricht University, Medical Centre, Department of Obstetrics and GynecologyMaastricht, The Netherlands. RP Koeneman, MM (reprint author), Maastricht University, Medical Centre, Department of Obstetrics and Gynecology, Maastricht, The Netherlands. EM margot.koeneman@mumc.nl CR zur Hausen H, 2000, Papillomaviruses causing cancer: evasion from host-cell control in early events in carcinogenesis. J Natl Cancer Inst 92(9):690–698 McCredie MR, Sharples KJ, Paul C, Baranyai J, Medley G, Jones RW et al, 2008, Natural history of cervical neoplasia and risk of invasive cancer in women with cervical intraepithelial neoplasia 3: a retrospective cohort study. Lancet Oncol 9(5):425–434 Trimble CL, Piantadosi S, Gravitt P, Ronnett B, Pizer E, Elko A et al, 2005, Spontaneous regression of high-grade cervical dysplasia: effects of human papillomavirus type andHLA phenotype. Clin Cancer Res 11(13):4717–4723 Ovestad IT, Gudlaugsson E, Skaland I, Malpica A, Munk AC, Janssen EA et al, 2011, The impact of epithelial biomarkers, local immune response and human papillomavirus genotype in the regression of cervical intraepithelial neoplasia grades 2-3. J Clin Pathol 64(4):303–307 Munk AC, Gudlaugsson E, Ovestad IT, Lovslett K, Fiane B, Hidle B et al, 2012, Interaction of epithelial biomarkers, local immune response and condom use in cervical intraepithelial neoplasia 2-3 regression. Gynecol Oncol 127(3):489–494 Grimm C, Polterauer S, Natter C, Rahhal J, Hefler L, Tempfer CB et al, 2012, Treatment of cervical intraepithelial neoplasia with topical imiquimod: a randomized controlled trial. Obstet Gynecol 120(1):152–159 Barken SS, Rebolj M, Andersen ES, Lynge E, 2012, Frequency of cervical intraepithelial neoplasia treatment in a well-screened population. Int J Cancer 130(10):2438–2444 Moody CA, Laimins LA, 2010, Human papillomavirus oncoproteins: pathways to transformation. Nat Rev Cancer 10(8):550–560 Stanley MA, 2012, Epithelial cell responses to infection with human papillomavirus. Clin Microbiol Rev 25(2):215–222 Litjens RJ, Hopman AH, van deVijver KK, Ramaekers FC, Kruitwagen RF, Kruse AJ, 2013, Molecular biomarkers in cervical cancer diagnosis: a critical appraisal. Expert Opin Med Diagn 7(4):365–377 Koeneman MM, Kruitwagen RF, Nijman HW, Slangen BF, Van Gorp T, Kruse AJ, 2015, Natural history of high-grade cervical intraepithelial neoplasia: a review of prognostic biomarkers. Expert Rev Mol Diagn 15(4):527–546 Thomas LK, Bermejo JL, Vinokurova S, Jensen K, Bierkens M, Steenbergen R et al, 2014, Chromosomal gains and losses in human papillomavirus-associated neoplasia of the lower genital tract - a systematic review and meta-analysis. Eur J Cancer 50(1):85–98 Seppo A, Jalali GR, Babkowski R, Symiakaki H, Rodolakis A, Tafas T et al, 2009, Gain of 3q26: a genetic marker in low-grade squamous intraepithelial lesions, LSIL, of the uterine cervix. Gynecol Oncol 114(1):80–83 Earley A, Lamont JL, Dahabreh IJ, Cowan J, Feldman S, Uhlig K, 2014, Fluorescence in situ hybridization testing for the diagnosis of high-grade cervical abnormalities: a systematic review. J Low Genit Tract Dis 18(3):218–227 HopmanAH, TheelenW, Hommelberg PP, KampsMA, Herrington CS, Morrison LE et al, 2006, Genomic integration of oncogenic HPV and gain of the human telomerase gene TERC at 3q26 are strongly associated events in the progression of uterine cervical dysplasia to invasive cancer. J Pathol 210(4):412–419 Wright TC, Compagno J, Romano P, Grazioli V, Verma Y, Kershnar E et al, 2015, Amplification of the 3q chromosomal region as a specific marker in cervical cancer. Am J Obstet Gynecol 213:51.e1-8 Hopman AH, van Hooren E, van de Kaa CA, Vooijs PG, Ramaekers FC, 1991, Detection of numerical chromosome aberrations using in situ hybridization in paraffin sections of routinely processed bladder cancers. Mod Pathol 4(4):503–513 Pahlplatz MM, deWilde PC, Poddighe P, van Dekken H, Vooijs GP, Hanselaar AG, 1995, A model for evaluation of in situ hybridization spot-count distributions in tissue sections. Cytometry 20(3):193–202 Heselmeyer-Haddad K, Sommerfeld K, White NM, Chaudhri N, Morrison LE, Palanisamy N et al, 2005, Genomic amplification of the human telomerase gene, TERC, in pap smears predicts the development of cervical cancer. Am J Pathol 166(4):1229–1238 Alameda F, Espinet B, Corzo C, Munoz R, Bellosillo B, Lloveras B et al, 2009, 3q26, hTERC, gain studied by fluorescence in situ hybridization as a persistence-progression indicator in low-grade squamous intraepithelial lesion cases. Hum Pathol 40(10):1474–1478 Jalali GR, Herzog TJ, Dziura B, Walat R, Kilpatrick MW, 2010, Amplification of the chromosome 3q26 region shows high negative predictive value for nonmalignant transformation of LSIL cytologic finding. Am J Obstet Gynecol 202:581.e1-5 Lan YL, Yu L, Jia CW, Wu YM, Wang SY, 2012, Gain of human telomerase RNA gene is associated with progression of cervical intraepithelial neoplasia grade I or II. Chin Med J 125(9):1599–1602 Rodolakis A, Biliatis I, Symiakaki H, Kershnar E, Kilpatrick MW, HaidopoulosDet al, 2012, Role of chromosome 3q26 gain in predicting progression of cervical dysplasia. Int J Gynecol Cancer 22(5):742–747 Obermann EC, Savic Prince S, Barascud A, Grilli B, Herzog M, Kaup D et al, 2013, Prediction of outcome in patients with lowgrade squamous intraepithelial lesions by fluorescence in situ hybridization analysis of human papillomavirus, TERC, and MYC. Cancer Cytopathol 121(8):423–431 Li L, Jiang W, Zeng SY, Li L, 2014, Prospective study of hTERC gene detection by fluorescence in situ hybridization, FISH, in cervical intraepithelial neoplasia 1 natural prognosis. Eur J Gynaecol Oncol 35(3):289–291 Ravaioli S, Tumedei MM, Amadori A, Puccetti M, Chiadini E, Bravaccini S, 2017, Role of telomerase in cervical lesions as prognostic marker: a comparison between immunohistochemistry and fluorescence in situ hybridization. J LowGenit Tract Dis 21(1):42–46 Olaharski AJ, Sotelo R, Solorza-Luna G, Gonsebatt ME, Guzman P, MoharAet al, 2006, Tetraploidy and chromosomal instability are early events during cervical carcinogenesis. Carcinogenesis 27(2):337–343 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 549 EP 557 DI 10.1007/s12253-018-0480-y PG 9 ER PT J AU Salem, MS Hamed, RA Fayez, GA Eldeen, NG AF Salem, M Sohair Hamed, R Ahmed Fayez, G Alaaeldin Eldeen, Nour Ghada TI Non-target Genes Regulate miRNAs-Mediated Migration Steering of Colorectal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MiRNAs; colorectal carcinoma; Non-target genes; MAP4K4; Migration ID MiRNAs; colorectal carcinoma; Non-target genes; MAP4K4; Migration AB MicroRNAs (miRNAs) trigger a two-layer regulatory network directly or through transcription factors and their co-regulators. Unlike miR-375, the role of miR-145 and miR-224 in inhibiting or driving cancer cell migration is controversial. This study is a step towards addressing the potential of miR-375, miR-145 and miR-224 expression modulation to inhibit colorectal carcinoma (CRC) cells migration in vitro through regulation of non-target genes VEGFA, TGFβ1, IGF1, CD105 and CD44. Transwell migration assay results revealed a significant subdue of migration ability of cells transfected with miR-375 and miR-145 mimics and miR-224 inhibitor. Real time PCR data showed that expression of VEGFA, TGFβ1, IGF1, CD105 and CD44 was downregulated as a consequence of exogenous re-expression of miR-375 and inhibition of miR-224. On the other hand, ectopic expression of miR-145 did not affect VEGFA, TGFβ1 and CD44 expression, while it elevated CD105 and suppressed IGF1 expression. MAP4K4, a predicted target of miR-145, was validated as a target that could play a role in miR-145-mediated regulation of migration. At mRNA level, no change was observed in expression of MAP4K4 in cells with restored expression of miR-145, while western blotting analysis revealed a 25% reduction of protein level. By applying luciferase reporter assay, a significant decrease in luciferase activity was observed, supporting that miR-145 directly target 3’ UTR of MAP4K4. The study highlighted the involvement of non-target genes VEGFA, TGFβ1, IGF1, CD105 and CD44 in mediating anti- and pro-migratory effect of miR-375 and miR-224, respectively, and validated MAP4K4 as a direct target of anti-migratory miR-145. C1 [Salem, M Sohair] National Research Centre, Molecular Genetics and Enzymology Department, 33 El-Buhouth St., Dokki, 12622 Giza, Egypt. [Hamed, R Ahmed] National Research Centre, Phytochemistry Department, 33 El-Buhouth St., Dokki, 12622 Giza, Egypt. [Fayez, G Alaaeldin] National Research Centre, Molecular Genetics and Enzymology Department, 33 El-Buhouth St., Dokki, 12622 Giza, Egypt. [Eldeen, Nour Ghada] National Research Centre, Molecular Genetics and Enzymology Department, 33 El-Buhouth St., Dokki, 12622 Giza, Egypt. RP Salem, MS (reprint author), National Research Centre, Molecular Genetics and Enzymology Department, 12622 Giza, Egypt. EM ssalem_nrc@hotmail.com CR Arner P, Kulyte A, 2015, MicroRNA regulatory networks in human adipose tissue and obesity. Nat Rev Endocrinol 11:276–288 Bassoy EY, Chiusolo V, Jacquemin G, Riccadonna C, Walker PR, Martinvalet D, 2016, Glioma Stemlike cells enhance the killing of glioma differentiated cells by cytotoxic lymphocytes. PLoS One 11: e0153433 Benetti A, Berenzi A, Gambarotti M, Garrafa E, Gelati M, Dessy E, Portolani N, Piardi T, Giulini SM, Caruso A, Invernici G, Parati EA, Nicosia R, Alessandri G, 2008, Transforming growth factor-beta1 and CD105 promote the migration of hepatocellular carcinomaderived endothelium. Cancer Res 68:8626–8634 Bradford MM, 1976, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72:248–254 Broderick JA, Zamore PD, 2011, MicroRNA therapeutics. Gene Ther 18:1104–1110 Chen Y, Gou X, Ke X, Cui H, Chen Z, 2012, Human tumor cells induce angiogenesis through positive feedback between CD147 and insulin-like growth factor-I. PLoS One 7:e40965 Collins CS, Hong J, Sapinoso L, Zhou Y, Liu Z, Micklash K, Schultz PG, Hampton GM, 2006, A small interfering RNA screen for modulators of tumor cell motility identifies MAP4K4 as a promigratory kinase. Proc Natl Acad Sci U S A 103:3775–3780 Cui F,Wang S, Lao I et al, 2016, miR-375 inhibits the invasion and metastasis of colorectal cancer via targeting SP1 and regulating EMT-associated genes. Oncol Rep 36:487–493 Deng B, Wang B, Fang J, Zhu X, Cao Z, Lin Q, Zhou L, Sun X, 2016, MiRNA-203 suppresses cell proliferation, migration and invasion in colorectal cancer via targeting of EIF5A2. Sci Rep 6: 28301 Dong R, Liu X, Zhang Q, Jiang Z, Li Y,Wei Y, Li Y, Yang Q, Liu J, Wei JJ, Shao C, Liu Z, Kong B, 2014, miR-145 inhibits tumor growth and metastasis by targetingmetadherin in high-grade serous ovarian carcinoma. Oncotarget 5:10816–10829 Emdad L, Das SK, Dasgupta S et al, 2013, AEG-1/MTDH/LYRIC: signaling pathways, downstream genes, interacting proteins, and regulation of tumor angiogenesis. Adv Cancer Res 120:75–111 Faltejskova P, Svoboda M, Srutova K, Mlcochova J, Besse A, Nekvindova J, Radova L, Fabian P, Slaba K, Kiss I, Vyzula R, Slaby O, 2012, Identification and functional screening of microRNAs highly deregulated in colorectal cancer. J Cell Mol Med 16:2655–2666 Feng Y, Zhu J, Ou C, Deng Z, Chen M, Huang W, Li L, 2014, MicroRNA-145 inhibits tumour growth and metastasis in colorectal cancer by targeting fascin-1. Br J Cancer 110:2300–2309 Fix LN, Shah M, Efferth T, Farwell MA, Zhang B, 2010, MicroRNA expression profile of MCF-7 human breast cancer cells and the effect of green tea polyphenon-60. Cancer Genomics Proteomics 7:261–277 Gao P, Xing AY, Zhou GY, Zhang TG, Zhang JP, Gao C, Li H, Shi DB, 2013, The molecular mechanism of microRNA-145 to suppress invasion-metastasis cascade in gastric cancer. Oncogene 32: 491–501 Gao Y, Feng B, Han S, Zhang K, Chen J, Li C, Wang R, Chen L, 2016, The roles of MicroRNA-141 in human cancers: from diagnosis to treatment. Cell Physiol Biochem 38:427–448 Gotte M, Mohr C, Koo CY et al, 2010, miR-145-dependent targeting of junctional adhesion molecule a and modulation of fascin expression are associated with reduced breast cancer cell motility and invasiveness. Oncogene 29:6569–6580 Hu Y, Leo C, Yu S, Huang BCB, Wang H, Shen M, Luo Y, Daniel-Issakani S, Payan DG, Xu X, 2004, Identification and functional characterization of a novel human misshapen/Nck interacting kinase-related kinase, hMINK beta. J Biol Chem 279:54387–54397 Ikushima H, Miyazono K, 2010, TGFbeta signalling: a complex web in cancer progression. Nat Rev Cancer 10:415–424 Jansson MD, Lund AH, 2012, MicroRNA and cancer. Mol Oncol 6:590–610 Ke TW, Hsu HL, Wu YH et al, 2014, MicroRNA-224 suppresses colorectal cancer cell migration by targeting Cdc42. Dis Markers 2014:617150 Lee NY, Ray B, How T, Blobe GC, 2008, Endoglin promotes transforming growth factor beta-mediated Smad 1/5/8 signaling and inhibits endothelial cell migration through its association with GIPC. J Biol Chem 283:32527–32533 Li Q, Ding C, Chen C, Zhang Z, Xiao H, Xie F, Lei L, Chen Y, Mao B, Jiang M, Li J, Wang D, Wang G, 2014, miR-224 promotion of cell migration and invasion by targeting Homeobox D 10 gene in human hepatocellular carcinoma. J Gastroenterol Hepatol 29:835– 842 Ling H, Pickard K, Ivan C, Isella C, Ikuo M, Mitter R, Spizzo R, Bullock MD, Braicu C, Pileczki V, Vincent K, Pichler M, Stiegelbauer V, Hoefler G, Almeida MI, Hsiao A, Zhang X, Primrose JN, Packham GK, Liu K, Bojja K, Gafa R, Xiao L, Rossi S, Song JH, Vannini I, Fanini F, Kopetz S, Zweidler- McKay P, Wang X, Ionescu C, Irimie A, Fabbri M, Lanza G, Hamilton SR, Berindan-Neagoe I, Medico E, Mirnezami AH, Calin GA, Nicoloso MS, 2016, The clinical and biological significance of MIR-224 expression in colorectal cancer metastasis. Gut 65:977–989 Liu F, Liu Y, Shen J, Zhang G, Han J, 2016, MicroRNA-224 inhibits proliferation and migration of breast cancer cells by downregulating fizzled 5 expression. Oncotarget 7:49130–49142 Loftus JC, Yang Z, Kloss J et al, 2013, A novel interaction between Pyk2 and MAP4K4 is integrated with glioma cell migration. J Signal Transduct 2013:956580 Michael MZ, Sm OC, Van Holst Pellekaan NG et al, 2003, Reduced accumulation of specific microRNAs in colorectal neoplasia. Mol Cancer Res 1:882–891 Neuzillet C, De Gramont A, Tijeras-Raballand A et al, 2014, Perspectives of TGF-beta inhibition in pancreatic and hepatocellular carcinomas. Oncotarget 5:78–94 Neuzillet C, Tijeras-Raballand A, Cohen R, Cros J, Faivre S, Raymond E, de Gramont A, 2015, Targeting the TGFbeta pathway for cancer therapy. Pharmacol Ther 147:22–31 Pillai RS, 2005, MicroRNA function: multiple mechanisms for a tiny RNA? RNA 11:1753–1761 Qin J,Wang F, Jiang H, Xu J, Jiang Y,Wang Z, 2015, MicroRNA- 145 suppresses cell migration and invasion by targeting paxillin in human colorectal cancer cells. Int J Clin Exp Pathol 8:1328–1340 Rupaimoole R, Slack FJ, 2017, MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov 16:203–222 Sakurai T, Kudo M, 2011, Signaling pathways governing tumor angiogenesis. Oncology 81(Suppl 1):24–29 Salem SM, Hamed AR,Mosaad RM, 2017, MTDH and MAP3K1 are direct targets of apoptosis-regulating miRNAs in colorectal carcinoma. Biomed Pharmacother 94:767–773 Shao Y, Chen H, Jiang X, Chen S, Li P, Ye M, Li Q, SunW, Guo J, 2014, Low expression of lncRNA-HMlincRNA717 in human gastric cancer and its clinical significances. Tumour Biol 35:9591– 9595 Speranza MC, Frattini V, Pisati F, Kapetis D, Porrati P, Eoli M, Pellegatta S, Finocchiaro G, 2012, NEDD9, a novel target of miR-145, increases the invasiveness of glioblastoma. Oncotarget 3:723–734 Stiegelbauer V, Vychytilova-Faltejskova P, Karbiener M, Pehserl AM, Reicher A, Resel M, Heitzer E, Ivan C, Bullock M, Ling H, Deutsch A, Wulf-Goldenberg A, Adiprasito JB, Stoeger H, Haybaeck J, Svoboda M, Stotz M, Hoefler G, Slaby O, Calin GA, Gerger A, Pichler M, 2017, miR-196b-5p regulates colorectal cancer cell migration and metastases through interaction with HOXB7 and GALNT5. Clin Cancer Res 23:5255–5266 Tu K, Yu H, Hua YJ, Li YY, Liu L, Xie L, Li YX, 2009, Combinatorial network of primary and secondary microRNAdriven regulatory mechanisms. Nucleic Acids Res 37:5969–5980 Vizio B, Biasi F, Scirelli T, Novarino A, Prati A, Ciuffreda L, Montrucchio G, Poli G, Bellone G, 2013, Pancreatic-carcinomacell- derived pro-angiogenic factors can induce endothelial-cell differentiation of a subset of circulating CD34+ progenitors. J Transl Med 11:314 Walsh LA, Damjanovski S, 2011, IGF-1 increases invasive potential of MCF 7 breast cancer cells and induces activation of latent TGF-beta1 resulting in epithelial to mesenchymal transition. Cell Commun Signal 9:10 Wang S, Liu H, Ren L, Pan Y, Zhang Y, 2008, Inhibiting colorectal carcinoma growth and metastasis by blocking the expression of VEGF using RNA interference. Neoplasia 10:399–407 Wang Y, Tang Q, Li M, Jiang S, Wang X, 2014, MicroRNA-375 inhibits colorectal cancer growth by targeting PIK3CA. Biochem Biophys Res Commun 444:199–204 Wang B, Shen ZL, Gao ZD, Zhao G,Wang CY, Yang Y, Zhang JZ, Yan YC, Shen C, Jiang KW, Ye YJ, Wang S, 2015, MiR-194, commonly repressed in colorectal cancer, suppresses tumor growth by regulating the MAP4K4/c-Jun/MDM2 signaling pathway. Cell Cycle 14:1046–1058 Wang B, Lv K, ChenWet al, 2016)miR-375 andmiR-205 regulate the invasion and migration of laryngeal squamous cell carcinoma synergistically via AKT-mediated EMT. Biomed Res Int 2016: 9652789 Xu L, Wen T, Liu Z, Xu F, Yang L, Liu J, Feng G, An G, 2016, MicroRNA-375 suppresses human colorectal cancer metastasis by targeting frizzled 8. Oncotarget 7:40644–40656 Yu Q, Stamenkovic I, 2000, Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis. Genes Dev 14:163–176 YuanW, Sui C, Liu Q, TangW, An H, Ma J, 2014, Up-regulation of microRNA-145 associates with lymph node metastasis in colorectal cancer. PLoS One 9:e102017 Zaravinos A, Soufla G, Bizakis J, Spandidos DA(2008, Expression analysis of VEGFA, FGF2, TGFbeta1, EGF and IGF1 in human nasal polyposis. Oncol Rep 19:385–391 Zemel R, Bachmetov L, Ad-El D et al, 2009, Expression of liverspecific markers in naive adipose-derived mesenchymal stem cells. Liver Int 29:1326–1337 Zhang GJ, ZhouH, Xiao HX, LiY, Zhou T, 2013, Up-regulation of miR-224 promotes cancer cell proliferation and invasion and predicts relapse of colorectal cancer. Cancer Cell Int 13:104 Zhang H, Pu J, Qi T, QiM, Yang C, Li S, Huang K, Zheng L, Tong Q, 2014, MicroRNA-145 inhibits the growth, invasion, metastasis and angiogenesis of neuroblastoma cells through targeting hypoxiainducible factor 2 alpha. Oncogene 33:387–397 Zhang GJ, Li JS, Zhou H, Xiao HX, Li Y, Zhou T, 2015, MicroRNA-106b promotes colorectal cancer cell migration and invasion by directly targeting DLC1. J Exp Clin Cancer Res 34:73 Zheng YB, Xiao K, Xiao GC, Tong SL, Ding Y, Wang QS, Li SB, Hao ZN, 2016, MicroRNA-103 promotes tumor growth and metastasis in colorectal cancer by directly targeting LATS2. Oncol Lett 12:2194–2200 Zou D-B, Wei X, Hu R-L, Yang XP, Zuo L, Zhang SM, Zhu HQ, Zhou Q, Gui SY, Wang Y, 2015, Melatonin inhibits the migration of colon cancer RKO cells by down-regulating myosin light chain kinase expression through cross-talk with p38 MAPK. Asian Pac J Cancer Prev 16:5835–5842 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 559 EP 566 DI 10.1007/s12253-018-0502-9 PG 8 ER PT J AU Hu, H Wang, T Pan, R Yang, Y Li, B Zhou, C Zhao, J Huang, Y Duan, Sh AF Hu, Haochang Wang, Tiangong Pan, Ranran Yang, Yong Li, Bin Zhou, Cong Zhao, Jun Huang, Yi Duan, Shiwei TI Hypermethylated Promoters of Secreted Frizzled-Related Protein Genes are Associated with Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SFRP1; SFRP2; DNA methylation; Colorectal cancer; Quantitative methylation specific PCR ID SFRP1; SFRP2; DNA methylation; Colorectal cancer; Quantitative methylation specific PCR AB Colorectal cancer (CRC) is one of the leading causes of death worldwide. Aberrant DNA methylation has been recognized as one of the most common molecular alterations in CRC. The goal of this study was to investigate the diagnostic value of SFRP1 and SFRP2 methylation for CRC. A total of 80 pairs of CRC patients were recruited to test the association of SFRP1 and SFRP2 promotor methylation with CRC. Methylation assay was performed using quantitative methylation-specific polymerase chain reaction (qMSP) method. In this study, we found the methylation levels of SFRP1 and SFRP2 in CRC tumor tissues were significantly higher than those in the adjacent non-tumor tissues (SFRP1: P = 2E-5; SFRP2: P = 0.014). Further bioinformatics analysis of TCGA data confirmed the association of the two genes with CRC (SFRP1: P = 7E-21; SFRP2: P = 5E-24). Luciferase reporter gene assay showed that the recombinant plasmids with SFRP1 and SFRP2 fragments could significantly enhance promoter activity (SFRP1: P = 0.002; SFRP2: P = 0.004). In addition, SFRP1 and SFRP2 methylation were inversely correlated with the mRNA expression displayed by TCGA data mining (SFRP1: r = −0.432, P = 4E-11; SFRP2: r = −0.478, P = 1E-13). GEO data analysis indicated that SFRP1 and SFRP2 expression were increased in three CRC cell lines (COLO320, HCT116 and HT29) after 5′-AZA-deoxycytidine treatment, suggesting that DNA methylation played an important role in regulating gene expression of the two genes. Our results confirmed that promoter methylation of SFRP1 and SFRP2 contributed to the risk of CRC. C1 [Hu, Haochang] Ningbo University, School of Medicine, Medical Genetics CenterNingbo, Zhejiang, China. [Wang, Tiangong] Ningbo University, School of Medicine, Medical Genetics CenterNingbo, Zhejiang, China. [Pan, Ranran] Ningbo University, School of Medicine, Medical Genetics CenterNingbo, Zhejiang, China. [Yang, Yong] Ningbo University, School of Medicine, Medical Genetics CenterNingbo, Zhejiang, China. [Li, Bin] Ningbo University, School of Medicine, Medical Genetics CenterNingbo, Zhejiang, China. [Zhou, Cong] Ningbo University, School of Medicine, Medical Genetics CenterNingbo, Zhejiang, China. [Zhao, Jun] Ningbo University, School of Medicine, Medical Genetics CenterNingbo, Zhejiang, China. [Huang, Yi] Ningbo University, School of Medicine, Ningbo First Hospital, Department of NeurosurgeryNingbo, Zhejiang, China. [Duan, Shiwei] Ningbo University, School of Medicine, Medical Genetics CenterNingbo, Zhejiang, China. RP Duan, Sh (reprint author), Ningbo University, School of Medicine, Medical Genetics Center, Ningbo, China. EM duanshiwei@nbu.edu.cn CR Huang J, Tan ZR, Yu J, Li H, Lv QL, Shao YY, Zhou HH, 2017, DNA hypermethylated status and gene expression of PAX1/SOX1 in patients with colorectal carcinoma. Onco Targets Ther 10:4739– 4751 Li J, Chen C, Bi X, Zhou C, Huang T, Ni C, Yang P, Chen S, Ye M, Duan S, 2017, DNA methylation of CMTM3, SSTR2, and MDFI genes in colorectal cancer. Gene 630:1–7 Hu H, Chen X,Wang C, Jiang Y, Li J, Ying X et al, 2017, The role of TFPI2 hypermethylation in the detection of gastric and colorectal cancer. Oncotarget 8:84054–84065 Dashwood RH, 1999, Early detection and prevention of colorectal cancer, review). Oncol Rep 6:277–281 Wu D, Chen X, Xu Y, Wang H, Yu G, Jiang L, Hong Q, Duan S, 2017, Prognostic value of MLH1 promoter methylation in male patients with esophageal squamous cell carcinoma. Oncol Lett 13: 2745–2750 Liu S, Chen X, Chen R, Wang J, Zhu G, Jiang J et al, 2017, Diagnostic role of Wnt pathway gene promoter methylation in non small cell lung cancer. Oncotarget 8:36354–36367 Ma D, Jiang C, Hu X, Liu H, Li Q, Li T et al, 2014, Methylation patterns of the IFN-gamma gene in cervical cancer tissues. Sci Rep 4:6331 Piras G,Monne M, Palmas AD, Calvisi A, Asproni R, Vacca F, Pilo L, Gabbas A, Latte G, 2014, Methylation analysis of the phosphates and tensin homologue on chromosome 10 gene, PTEN, in multiple myeloma. Clin Epigenetics 6:16 Cahill N, Rosenquist R, 2013, Uncovering the DNA methylome in chronic lymphocytic leukemia. Epigenetics 8:138–148 Chen C, Wang L, Liao Q, Huang Y, Ye H, Chen F et al, 2013, Hypermethylation of EDNRB promoter contributes to the risk of colorectal cancer. Diagn Pathol 8:199 Wang Z, Yuan X, Jiao N, Zhu H, Zhang Y, Tong J, 2012, CDH13 and FLBN3 gene methylation are associated with poor prognosis in colorectal cancer. Pathol Oncol Res 18:263–270 Oh TJ, Oh HI, Seo YY, Jeong D, Kim C, Kang HW, Han YD, Chung HC, Kim NK, An S, 2017, Feasibility of quantifying SDC2 methylation in stool DNA for early detection of colorectal cancer. Clin Epigenetics 9:126 Huang J, Zhang YL, Teng XM, Lin Y, Zheng DL, Yang PY, Han ZG, 2007, Down-regulation of SFRP1 as a putative tumor suppressor gene can contribute to human hepatocellular carcinoma. BMC Cancer 7:126 TakedaM, Nagasaka T, Dong-Sheng S, Nishie H,Oka T, Yamada E et al, 2011, Expansion of CpG methylation in the SFRP2 promoter region during colorectal tumorigenesis. Acta Med Okayama 65: 169–177 Park SK, Baek HL, Yu J, Kim JY, Yang HJ, Jung YS, Choi KY, Kim H, Kim HO, Jeong KU, Chun HK, Kim K, Park DI, 2017, Is methylation analysis of SFRP2, TFPI2, NDRG4, and BMP3 promoters suitable for colorectal cancer screening in the Korean population? Intest Res 15:495–501 Bartak BK, Kalmar A, Peterfia B, Patai AV, Galamb O, Valcz G et al, 2017, Colorectal adenoma and cancer detection based on altered methylation pattern of SFRP1, SFRP2, SDC2, and PRIMA1 in plasma samples. Epigenetics 12:751– 763 Babaei H, Mohammadi M, Salehi R, 2016, DNA methylation analysis of secreted frizzled-related protein 2 gene for the early detection of colorectal cancer in fecal DNA. Niger Med J 57:242–245 Zhang H, Zhu YQ, Wu YQ, Zhang P, Qi J, 2014, Detection of promoter hypermethylation of Wnt antagonist genes in fecal samples for diagnosis of early colorectal cancer. World J Gastroenterol 20:6329–6335 Huang Z, Li L, Wang J, 2007, Hypermethylation of SFRP2 as a potential marker for stool-based detection of colorectal cancer and precancerous lesions. Dig Dis Sci 52:2287–2291 Caldwell GM, Jones C, Gensberg K, Jan S, Hardy RG, Byrd P, Chughtai S, Wallis Y, Matthews GM, Morton DG, 2004, The Wnt antagonist sFRP1 in colorectal tumorigenesis. Cancer Res 64:883–888 Li B, Chen X, Jiang Y, Yang Y, Zhong J, Zhou C, Hu H, Duan S, 2017, CCL2 promoter hypomethylation is associated with gout risk in Chinese Han male population. Immunol Lett 190:15–19 Cannas V, Daino GL, Corona A, Esposito F, Tramontano E, 2015, A luciferase reporter gene assay to measure Ebola virus viral protein 35-associated inhibition of doublestranded RNA-stimulated, retinoic acid-inducible gene 1- mediated induction of interferon beta. J Infect Dis 212(Suppl 2):S277–S281 Yu J, Zhai X, Li X, Zhong C, Guo C, Yang F, Yuan Y, Zheng S, 2017, Identification of MST1 as a potential early detection biomarker for colorectal cancer through a proteomic approach. Sci Rep 7:14265 Yamashita K, Watanabe M, 2009, Clinical significance of tumor markers and an emerging perspective on colorectal cancer. Cancer Sci 100:195–199 Li X, Kong L, Liao S, Lu J, Ma L, Long X, 2017, The expression and significance of feces cyclooxygensae-2 mRNA in colorectal cancer and colorectal adenomas. Saudi J Gastroenterol 23:28–33 Wu Y, Gong L, Xu J, Mou Y, Xu X, Qian Z, 2017, The clinicopathological significance of HES1 promoter hypomethylation in patients with colorectal cancer. Onco Targets Ther 10:5827–5834 Zarkavelis G, Boussios S, Papadaki A, Katsanos KH, Christodoulou DK, Pentheroudakis G, 2017, Current and future biomarkers in colorectal cancer. Ann Gastroenterol 30:613–621 Sugai T, Yoshida M, Eizuka M, Uesugii N, Habano W, Otsuka K, Sasaki A, Yamamoto E, Matsumoto T, Suzuki H, 2017, Analysis of the DNA methylation level of cancer-related genes in colorectal cancer and the surrounding normal mucosa. Clin Epigenetics 9:55 Chen WD, Han ZJ, Skoletsky J, Olson J, Sah J, Myeroff L, Platzer P, Lu S, Dawson D, Willis J, Pretlow TP, Lutterbaugh J, Kasturi L, Willson JKV, Rao JS, Shuber A, Markowitz SD, 2005, Detection in fecal DNA of colon cancer-specific methylation of the nonexpressed vimentin gene. J Natl Cancer Inst 97:1124–1132 Huang S, Zhong X, Gao J, Song R, Wu H, Zi S et al, 2014, Coexpression of SFRP1 and WIF1 as a prognostic predictor of favorable outcomes in patients with colorectal carcinoma. Biomed Res Int 2014:256723 Veeck J, Niederacher D, An H, Klopocki E, Wiesmann F, Betz B, Galm O, Camara O, DurstM, Kristiansen G, Huszka C, Knuchel R, Dahl E, 2006, Aberrant methylation of the Wnt antagonist SFRP1 in breast cancer is associated with unfavourable prognosis. Oncogene 25:3479–3488 Ba S, Xuan Y, Long ZW, Chen HY, Zheng SS, 2017, MicroRNA-27a promotes the proliferation and invasiveness of Colon Cancer cells by targeting SFRP1 through the Wnt/beta-catenin signaling pathway. Cell Physiol Biochem 42: 1920–1933 Liu Y, Zhou Q, Zhou D, Huang C, Meng X, Li J, 2017, Secreted frizzled-related protein 2-mediated cancer events: Friend or foe? Pharmacol Rep 69:403–8 Worm OrntoftMB(2018, Reviewof blood-based colorectal Cancer screening: how far are circulating cell-free DNA methylation markers from clinical implementation? Clin Colorectal Cancer 17: e415–ee33 Harada T, Yamamoto E, Yamano HO, Nojima M, Maruyama R, Kumegawa K, Ashida M, Yoshikawa K, Kimura T, Harada E, Takagi R, Tanaka Y, Aoki H, Nishizono M, Nakaoka M, Tsuyada A, Niinuma T, Kai M, Shimoda K, Shinomura Y, Sugai T, Imai K, Suzuki H, 2014, Analysis of DNA methylation in bowel lavage fluid for detection of colorectal cancer. Cancer Prev Res 7:1002– 1010 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 567 EP 575 DI 10.1007/s12253-018-0505-6 PG 9 ER PT J AU Durak, GM Canda, T Yilmaz, B Seker, SN Kokkoz, ES Alicikus, AZ Akturk, N Gorken, BI Ellidokuz, H Sevinc, IA Saydam, S Sarioglu, S AF Durak, Guray Merih Canda, Tulay Yilmaz, Betul Seker, Sena Nazli Kokkoz, Eryigit Seda Alicikus, Arican Zumre Akturk, Nesrin Gorken, Bilkay Ilknur Ellidokuz, Hulya Sevinc, Ibrahim Ali Saydam, Serdar Sarioglu, Sulen TI Prognostic Importance of Tumor Deposits in the Ipsilateral Axillary Region of Breast Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Tumor deposits; Axillary dissection; Prognosis ID Breast cancer; Tumor deposits; Axillary dissection; Prognosis AB Tumor deposits (TD) are irregular discrete tumor masses in adipose tissue, discontinuous from the primary tumor, that are described in various cancers. The incidence and/or prognostic value of TD in breast carcinomas have not been studied so far. We reevaluated 145 breast cancer patients, diagnosed and treated between 2001 and 2006 at our institution for the presence and incidence of TD. Histologic type, grade, size of the primary tumor, estrogen receptor, progesterone receptor, human epidermal growth factor receptor-2 status of the tumor, and presence of peritumoral lymphovascular invasion were included in the data. TD were detected in 42 cases (29.0%). The mean age of the patients was 52.2 years (27–82). Most patients (79.3%) had either invasive carcinoma of no special type (NST) or invasive lobular carcinoma, and most tumors (86.9%) were either grade 2 or 3. After excluding TD from the number of metastatic lymph nodes, the pN status of 9 patients changed. Univariate analysis of 110 patients with follow-up information revealed that the new pN status (p = 0.036), presence of local recurrence (p = 0.016) and TD (p = 0.003) were significantly correlated with distant metastases. The median follow-up of the patients was 84 months (5–161), 10-year disease-free survival and overall survival were 67.2% and 73.7%, respectively. In multivariate analysis, presence of TD remained independently associated with distant metastasis (p = 0.002). The probability of distant metastasis was 3.3 times higher in patients with TD. These results emphasize that TD are present in breast cancer patients, and that their presence should warn the clinician in terms of possible distant metastasis. Therefore, presence of TD, the evaluation of which is neither time consuming nor require sophisticated methods, should be included in pathology reports. C1 [Durak, Guray Merih] Dokuz Eylul University, Faculty of Medicine, Department of Pathology, Inciralti, 35340 Izmir, Turkey. [Canda, Tulay] Dokuz Eylul University, Faculty of Medicine, Department of Pathology, Inciralti, 35340 Izmir, Turkey. [Yilmaz, Betul] Dokuz Eylul University, Faculty of Medicine, Department of Pathology, Inciralti, 35340 Izmir, Turkey. [Seker, Sena Nazli] Dokuz Eylul University, Faculty of Medicine, Department of Pathology, Inciralti, 35340 Izmir, Turkey. [Kokkoz, Eryigit Seda] Dokuz Eylul University, Faculty of Medicine, Department of Pathology, Inciralti, 35340 Izmir, Turkey. [Alicikus, Arican Zumre] Dokuz Eylul University, Faculty of Medicine, Department of Radiation OncologyIzmir, Turkey. [Akturk, Nesrin] Dokuz Eylul University, Faculty of Medicine, Department of Radiation OncologyIzmir, Turkey. [Gorken, Bilkay Ilknur] Dokuz Eylul University, Faculty of Medicine, Department of Radiation OncologyIzmir, Turkey. [Ellidokuz, Hulya] Dokuz Eylul University, Institute of OncologyIzmir, Turkey. [Sevinc, Ibrahim Ali] Dokuz Eylul University, Faculty of Medicine, Department of General SurgeryIzmir, Turkey. [Saydam, Serdar] Dokuz Eylul University, Faculty of Medicine, Department of General SurgeryIzmir, Turkey. [Sarioglu, Sulen] Dokuz Eylul University, Faculty of Medicine, Department of Pathology, Inciralti, 35340 Izmir, Turkey. RP Durak, GM (reprint author), Dokuz Eylul University, Faculty of Medicine, Department of Pathology, 35340 Izmir, Turkey. EM merihguray@yahoo.com CR Gabriel WB, Dukes C, Bussey HJR, 1935, Lymphatic spread in cancer of the rectum. Br J Surg 23:395–413 Puppa G, Ueno H, KayaharaM, Capelli P, CanzonieriV, Colombari R,Maisonneuve P, Pelosi G, 2009, Tumor deposits are encountered in advanced colorectal cancer and other adenocarcinomas: an expanded classification with implications for colorectal cancer staging system including a unifying concept of in-transit metastases. Mod Pathol 22:410–415 Sun Z, Wang ZN, Xu YY, Zhu GL, Huang BJ, Xu Y, Liu FN, Zhu Z, Xu HM, 2012, Prognostic significance of tumor deposits in gastric cancer patients who underwent radical surgery. Surgery 151:871–881 Ersen A, Unlu MS, Akman T, Sagol O, Oztop I, Atila K, Bora S, Ellidokuz H, Sarioglu S, 2014, Tumor deposits in gastric carcinomas. Pathol Res Pract 210:565–570 Sarioglu S, Akbulut N, Iplikci S, Aydin B, Dogan E, Unlu M, Ellidokuz H, Ada E, Akman F, Ikiz AO, 2016, Tumor deposits in head and neck carcinomas. Head Neck 38(Suppl 1):E256–E260 Goldstein NS, Turner JR, 2000, Pericolonic tumor deposits in patients with T3N+M0 colon adenocarcinomas: markers of reduced disease free survival and intraabdominal metastases and their implications for TNM classification. Cancer 88:2228–2238 Nagtegaal ID, Quirke P, 2007, Colorectal tumour deposits in the mesorectum and pericolon: a critical review. Histopathology 51: 141–149 Ratto C, Ricci R, Rossi C, Morelli U, Vecchio FM, Doglietto GB, 2002, Mesorectal microfoci adversely affect the prognosis of patients with rectal cancer. Dis Colon Rectum 45:733–742 ShimadaY, TakiiY(2010, Clinical impact ofmesorectal extranodal cancer tissue in rectal cancer: detailed pathological assessment using whole-mount sections. Dis Colon Rectum 53:771–778 Tong LL, Gao P,Wang ZN, Song YX, Xu YY, Sun Z, Xing CZ, Xu HM, 2012, Is the seventh edition of the UICC/AJCC TNM staging system reasonable for patients with tumor deposits in colorectal cancer? Ann Surg 255:208–213 Ueno H, Mochizuki H, 1997, Clinical significance of extrabowel skipped cancer infiltration in rectal cancer. Surg Today 27:617–622 Ueno H, Mochizuki H, Hashiguchi Y, Ishiguro M, Miyoshi M, KajiwaraY, Sato T, Shimazaki H,Hase K, 2007, Extramural cancer deposits without nodal structure in colorectal cancer: optimal categorization for prognostic staging. Am J Clin Pathol 127:287–294 Ueno H, Hashiguchi Y, Shimazaki H, Shinto E, Kajiwara Y, Nakanishi K, Kato K, Maekawa K, Nakamura T, Yamamoto J, Hase K, 2014, Peritumoral deposits as an adverse prognostic indicator of colorectal cancer. Am J Surg 207:70–77 Puppa G, Maisonneuve P, Sonzogni A, Masullo M, Capelli P, Chilosi M, Menestrina F, Viale G, Pelosi G, 2007, Pathological assessment of pericolonic tumor deposits in advanced colonic carcinoma: relevance to prognosis and tumor staging. Mod Pathol 20: 843–855 Hortobagyi GN, Connolly JL, D’Orsi CJ et al, 2017, Breast. In: AminMA(ed, AJCC Cancer stagingmanual, 8th edn. Springer, pp 589–628 Lakhani SR, Ellis IO, Schnitt SJ, Tan PH, van de Vijver MJ, eds, 2012, WHO classification of Tumours of the breast, 4th ed. Lyon, IARC Press Yamauchi C, Hasebe T, Iwasaki M, Imoto S, Wada N, Fukayama M, Ochiai A, 2007, Accurate assessment of lymph vessel tumor emboli in invasive ductal carcinoma of the breast according to tumor areas, and their prognostic significance. Hum Pathol 38:247–259 Marinho VF, Metze K, Sanches FS, Rocha GF, Gobbi H, 2008, Lymph vascular invasion in invasive mammary carcinomas identified by the endothelial lymphatic marker D2-40 is associated with other indicators of poor prognosis. BMC Cancer 8:64 Nagtegaal ID, Tot T, Jayne DG, McShane P, Nihlberg A, Marshall HC, Pahlman L, Brown JM, Guillou PJ, Quirke P, 2011, Lymph nodes, tumor deposits, and TNM: are we getting better? J Clin Oncol 29:2487–2492 Lee HS, Lee HE, Yang HK, Kim WH, 2013, Perigastric tumor deposits in primary gastric cancer: implications for patient prognosis and staging. Ann Surg Oncol 20:1604–1613 Belt EJ, van Stijn MF, Bril H, de Lange-de Klerk ES, Meijer GA, Meijer S, Stockmann HB, 2010, Lymph node negative colorectal cancers with isolated tumor deposits should be classified and treated as stage III. Ann Surg Oncol 17:3203–3211 Song YX, Gao P, Wang ZN, Liang JW, Sun Z, Wang MX, Dong YL,Wang XF, XuHM(2012, Can the tumor deposits be counted as metastatic lymph nodes in the UICC TNM staging system for colorectal cancer? PLoS One 7:e34087 Rock JB,Washington MK, Adsay NV, Greenson JK, Montgomery EA, Robert ME, Yantiss RK, Lehman AM, Frankel WL, 2014, Debating deposits: an interobserver variability study of lymph nodes and pericolonic tumor deposits in colonic adenocarcinoma. Arch Pathol Lab Med 138:636–642 Goldstein NS, Turner JR, 2000, Pericolonic tumour deposits in patients with T3N+M0 colon adenocarcinomas: markers of reduced disease free survival and intra-abdominal metastases and their implications for TNM classification. Cancer 88:2228–2238 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 577 EP 583 DI 10.1007/s12253-018-0515-4 PG 7 ER PT J AU Kotowski, U Kadletz, L Schneider, S Oberndorfer, F Erovic, MB Grasl, ChM Lill, C Heiduschka, G AF Kotowski, Ulana Kadletz, Lorenz Schneider, Sven Oberndorfer, Felicitas Erovic, M Boban Grasl, Ch Matthaeus Lill, Claudia Heiduschka, Gregor TI ELMO3 – a Negative Prognostic Marker in Minor Salivary Gland Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ELMO3; Molecular prognostic marker; Minor salivary gland cancer; Immunohistochemistry ID ELMO3; Molecular prognostic marker; Minor salivary gland cancer; Immunohistochemistry AB Engulfment and cell motility 3 protein (ELMO3) is a protein that is involved in cell migration and promotes the remodeling of the cytoskeleton. Moreover, it is described as a prognostic marker in several cancers. The aim of this study was to evaluate ELMO3 expression in patients with minor salivary gland carcinoma. The expression of ELMO3 was examined by immunohistochemistry. The intensity of staining was evaluated and data was correlated to clinical outcome. Forty-six patients with complete clinical data were included into statistical analysis. ELMO3 expression was observed in 85% of the cases. High staining intensity of ELMO3 correlated with a significantly worse disease free survival (p = .0495) and a higher recurrence rate (p = .0071). In conclusion, it is still difficult to predict the clinical outcome of patients with minor salivary gland carcinoma. Evaluation of ELMO3 might serve as a clinical prognostic marker in future. C1 [Kotowski, Ulana] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck Surgery, Waehringer Guertel 18-20, 1090 Vienna, Austria. [Kadletz, Lorenz] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck Surgery, Waehringer Guertel 18-20, 1090 Vienna, Austria. [Schneider, Sven] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck Surgery, Waehringer Guertel 18-20, 1090 Vienna, Austria. [Oberndorfer, Felicitas] Medical University of Vienna, Department of Pathology, Waehringer Guertel 18-20, 1090 Vienna, Austria. [Erovic, M Boban] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck Surgery, Waehringer Guertel 18-20, 1090 Vienna, Austria. [Grasl, Ch Matthaeus] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck Surgery, Waehringer Guertel 18-20, 1090 Vienna, Austria. [Lill, Claudia] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck Surgery, Waehringer Guertel 18-20, 1090 Vienna, Austria. [Heiduschka, Gregor] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck Surgery, Waehringer Guertel 18-20, 1090 Vienna, Austria. RP Lill, C (reprint author), Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck Surgery, 1090 Vienna, Austria. EM claudia.lill@meduniwien.ac.at CR Strick MJ, Kelly C, Soames JV, McLean NR, 2004, Malignant tumours of the minor salivary glands–a 20 year review. Br J Plast Surg 57:624–631. , DOI 10.1016/j.bjps.2004.04.017 Barnes L, World Health Organization, International Agency for Research on Cancer, 2005, Pathology and genetics of head and neck Tumours. IARC Jones AS, Beasley NJ, Houghton DJ, Helliwell TR, Husband DJ, 1998, Tumours of the minor salivary glands. Clin Otolaryngol Allied Sci 23:27–33 Lopes MA, Santos GC, Kowalski LP, 1998, Multivariate survival analysis of 128 cases of oral cavity minor salivary gland carcinomas. Head Neck 20:699–706 Vander Poorten VL, Balm AJ, Hilgers FJ et al, 2000, Stage as major long term outcome predictor in minor salivary gland carcinoma. Cancer 89:1195–1204 Erovic BM, Schopper C, Pammer J, Vormittag L, Maleki A, Brunner M, Heiduschka G, Grasl MC, Thurnher D, 2010, Multimodal treatment of patients with minor salivary gland cancer in the case of recurrent disease. Head Neck 32:1167–1172. https:// doi.org/10.1002/hed.21312 Carrillo JF,Maldonado F, Carrillo LC, Ramirez-OrtegaMC, Pizano JGG, Melo C, Chanona JG, Luna-Ortiz K, Ocana LFO, 2011, Prognostic factors in patients with minor salivary gland carcinoma of the oral cavity and oropharynx. Head Neck 33:1406–1412. , DOI 10.1002/hed.21641 Schneider S, Thurnher D, Seemann R, Brunner M, Kadletz L, GhanimB, Aumayr K, Heiduschka G, Lill C, 2016, The prognostic significance of β-catenin, cyclin D1 and PIN1 in minor salivary gland carcinoma: β-catenin predicts overall survival. Eur Arch Otorhinolaryngol 273:1283–1292. , DOI 10.1007/s00405- 015-3609-6 Goyette M-A, Cote J-F, 2014, NSCLC metastasis: going with ELMO3. Oncotarget 5:5850–5851. , DOI 10.18632/ oncotarget.2341 Fan W, Yang H, Xue H, Sun Y, Zhang J, 2015, ELMO3 is a novel biomarker for diagnosis and prognosis of non-small cell lung cancer. Int J Clin Exp Pathol 8:5503–5508 Kristensen LS, Soes S, Hansen LL, 2014, ELMO3: a direct driver of cancer metastasis? Cell Cycle 13:2483–2484. , DOI 10. 4161/15384101.2014.947228 Kadletz L, Heiduschka G, Wiebringhaus R, Gurnhofer E, Kotowski U, Haymerle G, Brunner M, Barry C, Kenner L, 2016, ELMO3 expression indicates a poor prognosis in head and neck squamous cell carcinoma - a short report. Cell Oncol, Dordr, 40:193–198. , DOI 10.1007/s13402-016- 0310-8 Haymerle G, Kadletz L, Wiebringhaus R, Golabi B, Mildner M, Thurnher D, Heiduschka G, 2017, ELMO3 predicts poor outcome in T1 laryngeal cancer. Clin Otolaryngol 42:1181–1186. https://doi. org/10.1111/coa.12845 Soes S, Daugaard IL, Sorensen BS, Carus A,MattheisenM, Alsner J, Overgaard J, Hager H, Hansen LL, Kristensen LS, 2014, Hypomethylation and increased expression of the putative oncogene ELMO3 are associated with lung cancer development and metastases formation. Oncoscience 1:367–374. , DOI 10. 18632/oncoscience.42 Coskun M, Boyd M, Olsen J, Troelsen JT, 2010, Control of intestinal promoter activity of the cellular migratory regulator gene ELMO3 by CDX2 and SP1. J Cell Biochem 109:1118–1128. , DOI 10.1002/jcb.22490 Kadletz L, Bigenzahn J, Thurnher D, Stanisz I, Erovic BM, Schneider S, Schmid R, Seemann R, Birner P, Heiduschka G, 2015, Evaluation of polo-like kinase 1 as a potential therapeutic target in Merkel cell carcinoma. Head Neck 38:E1918–E1925. , DOI 10.1002/hed.24349 Baddour HM Jr, Fedewa SA, Chen AY, 2016, Five- and 10-year cause-specific survival rates in carcinoma of the minor salivary gland. JAMA Otolaryngol Head Neck Surg 142:67–73. https:// doi.org/10.1001/jamaoto.2015.2805 Loh KS, Barker E, Bruch G, O'Sullivan B, Brown DH, Goldstein DP, Gilbert RW, Gullane PJ, Irish JC, 2009, Prognostic factors in malignancy of the minor salivary glands. Head Neck 31:58–63. , DOI 10.1002/hed.20924 Anderson JN, Beenken SW, Crowe R, Soong SJ, Peters G, Maddox WA, Urist MM, 1995, Prognostic factors in minor salivary gland cancer. Head Neck 17:480–486 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 585 EP 591 DI 10.1007/s12253-018-0475-8 PG 7 ER PT J AU Kumar, P Surya, V Urs, BA Augustine, J Mohanty, S Gupta, S AF Kumar, Priya Surya, Varun Urs, B Aadithya Augustine, Jeyaseelan Mohanty, Sujata Gupta, Sunita TI Sarcomas of the Oral and Maxillofacial Region: Analysis of 26 Cases with Emphasis on Diagnostic Challenges SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sarcoma; Oral and maxillofacial region; Molecular diagnostics ID Sarcoma; Oral and maxillofacial region; Molecular diagnostics AB Sarcomas of the Oral and Maxillofacial Region (SOMR) are rare lesions which pose diagnostic and management challenges.We analyzed 26 cases of SOMR with respect to clinical presentation, histopathological subtype, treatment modalities, recurrence, and treatment outcome. In our series, Osteosarcoma (OS) was the most common type of sarcoma (7 cases), followed by 5 cases of Ewing’s Sarcoma (ES), 3 cases each of Chondrosarcoma (CS) and Leiomyosarcoma (LMS), 2 cases each of Malignant Peripheral Nerve Sheath Tumor (MPNST), Pleomorphic Undifferentiated Sarcoma (PUS), Myeloid Sarcoma (MS)and Rhabdomyosarcoma (RMS). Surgery was the primary treatment modality in most cases and was combined with adjuvant chemo/ radiotherapy in few cases. 24 of the 26 cases were followed up for an average period of 40.67 months. Adverse disease outcomes like recurrence were seen in 2 cases whereas death due to the disease was reported in 7 cases. In view of the diagnostic challenges faced in SOMRs, it appears practical to stress on the underlying genetic aspects of the disease process rather than histological subtyping to improve disease outcome. C1 [Kumar, Priya] Maulana Azad Institute of Dental Sciences, Department of Oral Pathology, 7th floorNew Delhi, India. [Surya, Varun] Maulana Azad Institute of Dental Sciences, Department of Oral Pathology, 7th floorNew Delhi, India. [Urs, B Aadithya] Maulana Azad Institute of Dental Sciences, Department of Oral Pathology, 7th floorNew Delhi, India. [Augustine, Jeyaseelan] Maulana Azad Institute of Dental Sciences, Department of Oral Pathology, 7th floorNew Delhi, India. [Mohanty, Sujata] Maulana Azad Institute of Dental Sciences, Department of Oral and Maxillofacial SurgeryNew Delhi, India. [Gupta, Sunita] Maulana Azad Institute of Dental Sciences, Department of Oral Medicine & RadiologyNew Delhi, India. RP Kumar, P (reprint author), Maulana Azad Institute of Dental Sciences, Department of Oral Pathology, New Delhi, India. EM drpri_kumar@yahoo.com CR KrausDHSarcomas of the head and neck. Curr Oncol Rep 4:68–75 Greene F, Page D, Fleming I, Fritz A, Balch C, Haller D et al, eds,, 2002, American joint committee on Cancer: AJCC Cancer staging manual, 6th edn. Springer, New York, pp 77–87 Shellenberger T et al, 2009, Sarcomas of the head and neck region. Curr Oncol Rep 11:135–142 Tran LM, Mark R, Meier R, Calcaterra TC, Parker RG, 1992, Sarcomas of the head and neck . Prognostic factors and treatment strategies. Cancer 70:169–177 O'Neill JP, Bilsky MH, Kraus D, 2013, Head and neck sarcomas: epidemiology, pathology, and management. Neurosurg Clin N Am 24(1):67–78 Stavrakas M, Nixon I, Andi K, Oakley R, Jeannon JP, Lyons A, McGurk M, Urbano TG, Thavaraj S, Simo R, 2016, Head and neck sarcomas: clinical and histopathological presentation, treatment modalities, and outcomes. J Laryngol Otol 130(9):850–859 Siegel RL, Miller KD, Jemal A, 2015, Cancer statistics, 2015. CA Cancer J Clin 65(1):5–29 Peng KA, Grogan T, Wang MB, 2014, Head and neck sarcomas: analysis of the SEER database. Otolaryngol Head Neck Surg 151(4):627–633 Wanebo HJ, Koness RJ, Macfarlane JK, Eilber FR, Byers RM, Elias EG et al, 1992, Head and neck sarcoma: report of the head and neck sarcoma registry. Head Neck 14:1–7 Aljabab A, Nason R, Kazi R, Pathak K, 2011, Head and neck soft tissue sarcoma. Indian J Surg Oncol 2:286–290 Arya S, Chaukar D, Pai P, 2012, Imaging in oral cancers. Indian J Radiol Imaging 22:195–208 Chaudhary M, Chaudhary SD, 2012, Osteosarcoma of jaws. J Oral Maxillofac Pathol 16(2):233–238 Hong HR, Jin S, Koo HJ, Roh JL, Kim JS, Cho KJ, Choi SH, Nam SY, Kim SY, 2014, Clinical values of, 18, F-FDG PET/CT in oral cavity cancer with dental artifacts on CT or MRI. J Surg Oncol 110(6):696–701 Kumar R, Chauhan A, Vellimana AK, Chawla M, 2006, Role of PET/PET-CT in the management of sarcomas. Expert Rev Anticancer Ther 6(8):1241–1250 Wu JS, Hochman MG, 2009, Soft-tissue tumors and tumorlike lesions: a systematic imaging approach. Radiology 253(2):297–316 Rekhi B, Gorad BD, Kakade AC, Chinoy R, 2007, Scope of FNAC in the diagnosis of soft tissue tumors: a study from a tertiary cancer referral Centre in India. Cytojournal 31:4–20 Yoshida A, Ushiku T, Motoi T, Beppu Y, Fukayama M, Tsuda H, Shibata T, 2012, MDM2 and CDK4 immunohistochemical coexpression in high-grade osteosarcoma: correlation with a dedifferentiated subtype. Am J Surg Pathol 36(3):423–431 Tabareau-Delalande F, Collin C, Gomez-Brouchet A, Bouvier C, Decouvelaere AV, de Muret A, Pages JC, de Pinieux G, 2014, Chromosome 12 long arm rearrangement covering MDM2 and RASAL1 is associated with aggressive craniofacial juvenile ossifying fibroma and extracranial psammomatoid fibro-osseous lesions. Mod Pathol 28(1):48–56 Argon A, Doganavsargil B, UnalYildirim F, Sezak M, Midilli R, Oztop F, 2015, Osteosarcomas of jaw: experience of a single Centre. J Plast Surg Hand Surg 49(1):13–18 Yildiz FR, Avci A, Dereci O, Erol B, Celasun B, Gunhan O, 2014, Gnathic osteosarcomas, experience of four institutions from Turkey. Int J Clin Exp Pathol 7(6):2800–2808 Paparella ML, Olvi LG, Brandizzi D, Keszler A, Santini-Araujo E, Cabrini RL, 2013, Osteosarcoma of the jaw: an analysis of a series of 74 cases. Histopathology 63(4):551–557 Gomez-Brouchet A, Mourcin F, Gourraud PA, Bouvier C, De Pinieux G, Le Guelec S, Brousset P, Delisle MB, Schiff C, 2010, Galectin-1 is a powerful marker to distinguish chondroblastic osteosarcoma and conventional chondrosarcoma. Hum Pathol 41(9): 1220–1230 Unni KK, 2001, Cartilaginous lesions of bone. J Orthop Sci 6:457– 472 Lee RJ, Arshi A, Schwartz HC, Christensen RE, 2015, Characteristics and prognostic factors of osteosarcoma of the jaws: a retrospective cohort study. JAMA Otolaryngol Head Neck Surg 141(5):470–477 Okada K, Hasegawa T, Yokoyama R et al, 2003, Prognostic relevance of rosette-like features in osteosarcoma. J Clin Pathol 56: 831–834 Urs AB, Kumar P, Rawat G,Mohanty S, 2016, Ewing’s sarcoma of the jaws: an institutional study of four cases. Int J Pediatr Otorhinolaryngol Extra 13:33–39 Lucas DR, Bentley G, Dan ME, Tabaczka P, Poulik JM, Mott MP, 2001, Ewing sarcoma vs lymphoblastic lymphoma.A comparative immunohistochemical study. Am J Clin Pathol 115(1):11–17 Yu L, Li M, Lin R, Mu Y, Zhao J, 2014, Mesenchymal chondrosarcoma of the right buccal region: a case report and review of the literature. Oncol Lett 8(6):2557–2560. , DOI 10. 3892/ol.2014.2595 Wehrli BM, HuangW, De Crombrugghe B, Ayala AG, Czerniak B, 2003, Sox9, a master regulator of chondrogenesis, distinguishes mesenchymal chondrosarcoma from other small blue round cell tumors. Hum Pathol 34(3):263–269 McKay KM, Deavers MT, Prieto VG, 2012, Chondroid melanoma metastatic to lung and skin showing SOX-9 expression: a potential diagnostic pitfall. Int J Surg Pathol 20:169–172 Nakashima Y, Unni KK, Shives TC, Swee RG, Dahlin DC, 1986, Mesenchymal chondrosarcoma of bone and soft tissue. A review of 111 cases. Cancer 15;57(12):2444–2453 Vencio EF, Reeve CM, Unni KK, Nascimento AG, 1998, Mesenchymal chondrosarcoma of the jaw bones: clinicopathologic study of 19 cases. Cancer 15;82(12):2350–2355 Yan B, Li Y, Pan J, Xia H, Li LJ, 2010, Primary oral leiomyosarcoma: a retrospective clinical analysis of 20 cases. Oral Dis 16(2):198–203 Vilos GA, Rapidis AD, Lagogiannis GD, Apostolidis C, 2005, Leiomyosarcomas of the oral tissuses: clinicopathologic analysis of 50 cases. J Oral Maxillofac Surg 63:1461–1477 Nikitakis NG, Lopes MA, Bailey JS, Blanchaert RH Jr, Ord RA, Sauk JJ, 2002, Oral leiomyosarcoma: review of the literature and report of two cases with assessment of the prognostic and diagnostic significance of immunohistochemical and molecular markers. Oral Oncol 38(2):201–208 Fisher C, 2004)Myofibrosarcoma. Virchows Arch 445(3):215–223 Montgomery E, Fisher C, 2001, Myofibroblastic differentiation in malignant fibrous histiocytoma, pleomorphic myofibrosarcoma): a clinicopathological study. Histopathology 38(6):499–509 Perez EA, Kassira N, Cheung MC, Koniaris LG, Neville HL, Sola JE, 2011, Rhabdomyosarcoma in children: a SEER population based study. J Surg Res 170(2):e243–ee51 Chen E, Ricciotti R, Futran N, Oda D, 2017, Head and neck rhabdomyosarcoma: clinical and pathologic characterization of seven cases. Head Neck Pathol 11(3):321–326 Woodruff JM, Perino G, 1994, Non-germ-cell or teratomatous malignant tumors showing additional rhabdomyoblastic differentiation, with emphasis on the malignant triton tumor. Semin Diagn Pathol 11:69–81 Devesa P, Mitchell TE, Scott I, Moffat D, 2006, Malignant peripheral nerve sheath tumors of the head and neck: two cases and a review of the literature. Ear Nose Throat J 85:392–396 Cleven AH, Sannaa GA, Briaire-de Bruijn I, Ingram DR, van de Rijn M, Rubin BP, de Vries MW, Watson KL, Torres KE, Wang WL, van Duinen SG, Hogendoorn PC, Lazar AJ, Bovee JV, 2016, Loss of H3K27 tri-methylation is a diagnosticmarker for malignant peripheral nerve sheath tumors and an indicator for an inferior survival. Mod Pathol 29(6):582–590 Guo A, Liu A, Wei L, Song X, 2012, Malignant peripheral nerve sheath tumors: differentiation patterns and Immunohistochemical features - a mini-review and our new findings. J Cancer 3:303–309 Kumar P, Singh H, Khurana N, Urs AB, Augustine J, Tomar R, 2017, Diagnostic challenges with intraoral myeloid sarcoma: report of two cases& review of world literature. Exp Oncol 39:78–85 Demicco EG, 2013, Sarcoma diagnosis in the age of molecular pathology. Adv Anat Pathol 20(4):264–274 Marino-Enriquez A, Bovee JVMG, 2016)Molecular pathogenesis, diagnostic, prognostic and predictive molecular markers in sarcoma. Surgical pathology clinics 9(3):457–473 Antonescu CR, Owosho AA, Zhang L, Chen S, Deniz K, Huryn JM, Kao YC, Huang SC, Singer S, Tap W, Schaefer IM, Fletcher CD, 2017, Sarcomas with CIC-rearrangements are a distinct pathologic entity with aggressive outcome: a Clinicopathologic and molecular study of 115 cases. Am J Surg Pathol 41(7):941–949 Bovee JV, Hogendoorn PC, 2010, Molecular pathology of sarcomas: concepts and clinical implications. Virchows Arch 456(2): 193–199 Casali PG, Jost L, Sleijfer S, Verweij J, Blay JY, 2008, Soft tissue sarcomas: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol 19(Suppl 2):ii89–ii93 Jaetli V, Gupta S, 2011, Mesenchymal chondrosarcoma of maxilla: a rare case report. Med Oral Patol Oral Cir Bucal 1;16(4):e493– e496 Gupta SR, Saran RK, Sharma P, Urs AB, 2015, A rare case of Extraskeletal mesenchymal chondrosarcoma with dedifferentiation arising from the buccal space in a young male. J Maxillofac Oral Surg 14(Suppl 1):293–299 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 593 EP 601 DI 10.1007/s12253-018-0510-9 PG 9 ER PT J AU Okuyama, K Fukushima, H Naruse, T Yanamoto, S Tsuchihashi, H Umeda, M AF Okuyama, Kohei Fukushima, Hiromasa Naruse, Tomofumi Yanamoto, Souichi Tsuchihashi, Hiroki Umeda, Masahiro TI CD44 Variant 6 Expression and Tumor Budding in the Medullary Invasion Front of Mandibular Gingival Squamous Cell Carcinoma Are Predictive Factors for Cervical Lymph Node Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD44 variant 6; Tumor budding; Mandibular gingival squamous cell carcinoma; Medullary invasion; Cervical lymph node metastasis ID CD44 variant 6; Tumor budding; Mandibular gingival squamous cell carcinoma; Medullary invasion; Cervical lymph node metastasis AB Oral squamous cell carcinoma (OSCC) with invasion into the mandibular medullary space has been reported to be a predictive factor for cervical lymph node metastasis (CLNM). As CLNM has been associated with the stemness of cancer cells, we aimed to evaluate the relationship between clinical characteristics and immunohistochemical findings on the invasion front of the medullary invasive OSCC and CLNM. The medical records of 25 patients with the mandibular medullary invasive OSCC who were performed mandibulectomy and neck dissection in our department from 2010 to 2016 were examined. Serial sections were stained with antibodies against CD44 variant 6 (CD44v6) to examine cancer stemness and to evaluate the number of tumor buds in the medullary invasion front of the mandibular invasive OSCC. Categorical data were analyzed by Fisher’s exact test. The expression of CD44v6 and the number of tumor buds between the groups with and without pathological CLNM (CLNM+ and CLNM-, respectively) were analyzed using the Mann-Whitney U test. Of the 25 patients, 11 patients were CLNM+. Of the several measured variables, histologic differentiation of the mandibular invasive OSCC was a significant factor for CLNM+. CD44v6 expression and tumor bud formation in the medullary invasion front of the mandibular invasive OSCC were significantly higher in the CLNM+ group, suggests that both CD44v6 and tumor budding in the medullary invasion front are predictive factors for CLNM. C1 [Okuyama, Kohei] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1, Sakamoto, Nagasaki-shi, 852-8588 Nagasaki, Japan. [Fukushima, Hiromasa] Nagasaki University, School of DentistryNagasaki, Japan. [Naruse, Tomofumi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1, Sakamoto, Nagasaki-shi, 852-8588 Nagasaki, Japan. [Yanamoto, Souichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1, Sakamoto, Nagasaki-shi, 852-8588 Nagasaki, Japan. [Tsuchihashi, Hiroki] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1, Sakamoto, Nagasaki-shi, 852-8588 Nagasaki, Japan. [Umeda, Masahiro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1, Sakamoto, Nagasaki-shi, 852-8588 Nagasaki, Japan. RP Okuyama, K (reprint author), Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 852-8588 Nagasaki, Japan. EM okuyamak@nagasaki-u.ac.jp CR Rautava J, Luukkaa M, Heikinheimo K, Alin J, Grenman R, Happonen RP, 2007, Squamous cell carcinomas arising from different types of oral epithelia differ in their tumor and patient characteristics and survival. Oral Oncol 43:911–919 Shingaki S, Nomura T, Takada M, Kobayashi T, Suzuki I, Nakajima T, 2002, Squamous cell carcinomas of the mandibular alveolus: analysis of prognostic factors. Oncology 62:17–24 Fitzpatrick SG, Neuman AN, Cohen DM, Bhattacharyya I, 2012, The clinical and histologic presentation of gingival squamous cell carcinoma: a study of 519 cases. Oral Surg Oral Med Oral Pathol Oral Radiol 114:509–515 Soo KC, Spiro RH, King W, Harvey W, Strong EW, 1988, Squamous carcinoma of the gums. Am J Surg 156:281–285 Overholt SM, Eicher SA, Wolf P, Weber RS, 1996, Prognostic factors affecting outcome in lower gingival carcinoma. Laryngoscope 106:1335–1339 Seoane J, Varela-Centelles PI, Walsh TF, Lopez-Cedrun JL, Vazquez I, 2006, Gingival squamous cell carcinoma: diagnostic delay or rapid invasion? J Periodontol 77:1229–1233 Gomez D, Faucher A, Picot V, Siberchicot F, Renaud-Salis JL, Bussieres E, Pinsolle J, 2000, Outcome of squamous cell carcinoma of the gingiva: a follow-up study of 83 cases. J Craniomaxillofac Surg 28:331–335 Ebrahimi A, Murali R, Gao K, Elliott MS, Clark JR, 2011, The prognostic and staging implications of bone invasion in oral squamous cell carcinoma. Cancer 117:4460–4467 Lubek J, El-HakimM, Salama AR et al, 2011, Gingival carcinoma: retrospective analysis of 72 patients and indications for elective neck dissection. Br J Oral Maxillofac Surg 49:182–185 Niu LX, Feng ZE, Wang DC, Zhang JY, Sun ZP, Guo CB, 2017, Prognostic factors in mandibular gingival squamous cell carcinoma: a 10-year retrospective study. Int J Oral Maxillofac Surg 46:137– 143 Ogura I, Kurabayashi T, Amagasa T, Okada N, Sasaki T, 2002, Mandibular bone invasion by gingival carcinoma on dental CT images as an indicator of cervical lymph node metastasis. Dentomaxillofac Radiol 31:339–343 Platz H, Fries R, Hudec Met al, 1983, The prognostic relevance of various factors at the time of the first admission of the patient. retrospective DOSAC study on carcinoma of the oral cavity J Maxillofac Surg 11:3–12 Okura M, Yanamoto S, Umeda M, Otsuru M, Ota Y, Kurita H, Kamata T, Kirita T, Yamakawa N, Yamashita T, Ueda M, Komori T, Hasegawa T, Aikawa T, Japan Oral Oncology Group, 2016, Prognostic and staging implications of mandibular canal invasion in lower gingival squamous cell carcinoma. Cancer Med 5:3378– 3385 Matsushita Y, Yanamoto S, Yamada S, Mori H, Adachi M, Takahashi H, Naruse T, Ikeda H, Shiraishi T, Minamikawa T, Shibuya Y, Komori T, Asahina I, Umeda M, 2015, Correlation between degree of mandibular bone invasion and prognosis in carcinoma of the mandibular gingiva: soft tissue classification based on UICC classification. J Oral Maxillofac Surg Med Pathol 27: 631–636 National Comprehensive Cancer Network NCCN Guidelines for Treatment of Cancer by Site. https://www.nccn.org/professionals/physician_gls/default.aspx#site [Accessibility verified April 15, 2018] Orian-Rousseau V, 2010, CD44, a therapeutic target for metastasizing tumours. Eur J Cancer 46:1271–1277 Monteiro LS, Delgado ML, Ricardo S, do Amaral B, Salazar F, Pacheco JJ, Lopes CA, Bousbaa H, Warnakulasuryia S, 2016, Prognostic significance of CD44v6, p63, podoplanin and MMP-9 in oral squamous cell carcinomas. Oral Dis 22:303–312 Yanamoto S, Yamada S, Takahashi H et al, 2014, Expression of the cancer stem cell markers CD44v6 and ABCG2 in tongue cancer: effect of neoadjuvant chemotherapy on local recurrence. Int J Oncol 44:1153–1162 Wang C, Huang H,Wang A et al, 2011, Tumor budding correlates with poor prognosis and epithelial-mesenchymal transition in tongue squamous cell carcinoma. J Oral Pathol Med 40:545–551 Shimizu S, Miyazaki A, Sonoda T, Koike K, Ogi K, Kobayashi JI, Kaneko T, Igarashi T, Ueda M, Dehari H, Miyakawa A, Hasegawa T, Hiratsuka H, 2018, Tumor budding is an independent prognostic marker in early stage oral squamous cell carcinoma: with special reference to the mode of invasion and worst pattern of invasion. PLoS One 13:e0195451 Hong KO Oh KY1, shinWJ et al. tumor budding is associated with poor prognosis of oral squamous cell carcinoma and histologically represents an epithelial-mesenchymal transition process. Hum Pathol in Press Mitrovic B, Schaeffer DF, Riddell RH, Kirsch R, 2012, Tumor budding in colorectal carcinoma: time to take notice. Modern Pathol 25:1315–1325 Pinborg JJ, Reichart PA, Smith CJ et al, 1997, World Health Organization histological typing of cancer and precancer of the oral mucosa, 2nd edn, Springer Sloan P, Gale N, Hunter K et al, 2017, Malignant surface epithelial tumours: squamous cell carcinoma. In: el-Naggar AK, Chan JKC, Grandis JR, Takata T, Slootweg PJ, eds, WHO classification of tumours of the head and neck, 4th edn. IARC Press, Lyon Okuyama K,Michi Y,Mizutani M, YamashiroM, Kaida A, Harada K, 2016, Clinical study on mandibular fracture after marginal resection of the mandible. Oral Surg Oral Med Oral Pathol Oral Radiol 121:461–467 Fonseca I, Pereira T, Rosa-Santos J, Soares J, 2001, Expression of CD44 isoforms in squamous cell carcinoma of the border of the tongue: a correlation with histological grade, pattern of stromal invasion, and cell differentiation. J Surg Oncol 76:115–120 Masaki T, Goto A, SugiyamaM,Matsuoka H, Abe N, Sakamoto A, Atomi Y, 2001, Possible contribution of CD44 variant 6 and nuclear beta-catenin expression to the formation of budding tumor cells in patients with T1 colorectal carcinoma. Cancer 92:2539– 2546 Seki M, Sano T, Yokoo S, Oyama T, 2016, Histologic assessment of tumor budding in preoperative biopsies to predict nodal metastasis in squamous cell carcinoma of the tongue and floor of the mouth. Head Neck 38(Suppl 1):E1582–E1590 Nomura T, Shibahara T, Cui NH, Noma H, 2005, Patterns of mandibular invasion by gingival squamous cell carcinoma. J Oral Maxillofac Surg 63:1489–1493 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 603 EP 609 DI 10.1007/s12253-018-0529-y PG 7 ER PT J AU Alabiad, AM Harb, AO Taha, FH Shafaay, ShEB Gertallah, ML Salama, N AF Alabiad, Ali Mohamed Harb, A Ola Taha, F Heba Shafaay, Sh El Basant Gertallah, M Loay Salama, Nashaat TI Prognostic and Clinic-Pathological Significances of SCF and COX-2 Expression in Inflammatory and Malignant Prostatic Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SCF; COX-2; Prostatic adenocarcinoma; Immunohistochemistry; Prognosis ID SCF; COX-2; Prostatic adenocarcinoma; Immunohistochemistry; Prognosis AB The initiation of prostatic malignancy has been linked to chronic inflammation. Stem cell factor (SCF) is an inflammatory cytokine that is specific to the c-KIT receptor which is type III receptor tyrosine kinase (RTK). Cyclooxygenases (COXs) are themain enzymes which are responsible for prostaglandins production from arachidonic acid. COX2 is an enzyme which is produced under different pathological conditions. The aim of our study; is to investigate the clinicopathological and the prognostic significance of SCF and COX-2 expression in prostatic adenocarcinoma (PC), chronic prostatitis and nodular prostatic hyperplasia (NPH) in a trial to clarify the role of inflammation as a risk factor for prostatic carcinogenesis and cancer progression. SCF and COX-2 tissue protein expression were evaluated in 50 cases of PC, 20 cases of chronic prostatitis and 10 cases of NPH using immunohistochemistry, patients were followed up for 5 years. The relationship between their levels of expressions, clinicopathological, and prognostic criteria were studied. SCF expression in PC was positively correlated with advanced patient age (p = <0.001), high level of PSA (p = 0.010), higher Gleason score (p = 0.011). COX-2 expression in PC was positively correlated with advanced patient age (p = <0.001), high level of PSA (p = 0.016), advanced D’Amico risk group (p = 0.038). High levels of expression of both SCF& COX-2 are associated with higher incidence of tumor relapse, worse disease overall survival and free survival (p < 0.001). SCF and COX-2 are associated with PC progression and associated with poor prognosis in PC patients. C1 [Alabiad, Ali Mohamed] Zagazig University, Faculty of Medicine, Pathology DepartmentZagazig, Egypt. [Harb, A Ola] Zagazig University, Faculty of Medicine, Pathology DepartmentZagazig, Egypt. [Taha, F Heba] Zagazig University, Faculty of Medicine, Medical Oncology DepartmentZagazig, Egypt. [Shafaay, Sh El Basant] Zagazig University, Faculty of Medicine, Clinical Oncology and Nuclear Medicine DepartmentZagazig, Egypt. [Gertallah, M Loay] Zagazig University, Faculty of Medicine, General Surgery DepartmentZagazig, Egypt. [Salama, Nashaat] Zagazig University, Faculty of Medicine, Urology DepartmentZagazig, Egypt. RP Harb, AO (reprint author), Zagazig University, Faculty of Medicine, Pathology Department, Zagazig, Egypt. EM olaharb2015@gmail.com CR Xu F, Li M, Zhang C, Cui J, Liu J, Li J, Jiang H, 2017, Clinicopathological and prognostic significance of COX-2 Immunohistochemical expression in breast Cancer: a meta-analysis. Oncotarget 8(4):6003–6012. , DOI 10.18632/ oncotarget.13990 Yoo S, Se YC, You D, Kim CS, 2016, New drugs in prostate Cancer. Prostate International 4(2):37–42. , DOI 10.1016/ j.prnil.2016.05.001 Abeshouse A, Ahn J, Akbani R, Ally A, Amin S, Andry CD, Annala M et al, 2016, Prostate Cancer. N Engl J Med 58(1):1–6. , DOI 10.1017/CBO9781107415324.004. Lee KC, Cho IR, 2017, Chronic prostatitis/chronic pelvic pain syndrome in adolescents compared with that in young adults. Investigative and clinical urology 58(4):267–270 Cardoso HJ, Figueira MI, Socorro S, 2017, The stem cell factor, SCF)/c-KIT signaling in testis and prostate cancer. Journal of cell communication and signaling 11(4):297–307 Tayel SI, El-Hefnway SM, El Gayed EMA, Abdelaal GA, 2017, Association of stem cell factor gene expression with severity and atopic state in patients with bronchial asthma. Respir Res 18(1):21 Huang, Bo, Zhang Lei, Gui-mei Zhang, Dong Li, Chuanwang Song, Bo Li, Yanyan Liu, et al. 2014. BSCF-mediated mast cell infiltration and activation exacerbate the inflammation and immunosuppression in tumor microenvironment SCF-mediated mast cell infiltration and activation exacerbate the inflammation and immunosuppression in tumor microenvironment^. 112, 4): 1269–1279. , DOI 10.1182/blood-2008-03-147033 Bilani N, Bahmad H, Abou-Kheir W, 2017, Prostate cancer and aspirin use: synopsis of the proposed molecular mechanisms. Front Pharmacol 8:145 Calisma, Immunohistokimyasal, 2016, BImmunohistochemical Study of Cyclooxygenase-2 Expression in Prostate Carcinoma : It ’ S Relation with Apoptosis and Angiogenesis^. 144–147 Hsu SM, Raine L, Fanger HX, 1981, Use of avidin-biotinperoxidase complex, ABC, in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody, PAP, procedures. Journal of Histochemistry & Cytochemistry 29(4):577–580 Siddique HR, Parray A, Zhong W, Karnes RJ, Bergstralh EJ, Koochekpour S, Saleem M, 2013, Correction: BMI1, stem cell factor acting as novel serum-biomarker for Caucasian and African-American prostate Cancer. PLoS One 8(6) Wang X, Ren H, Zhao T, Chen J, Sun W, Sun Y, Lang M, 2014, Stem cell factor is a novel independent prognostic biomarker for hepatocellular carcinoma after curative resection. Carcinogenesis 35(10):2283–2290 Wang X, Dong J, Jia L, Zhao T, LangM, Li Z, Qin T, 2017, HIF-2- dependent expression of stem cell factor promotes metastasis in hepatocellular carcinoma. Cancer Lett 393:113–124 Bellone G, Smirne C, Carbone A, Buffolino A, Scirelli T, Prati A, Emanuelli G, 2006, KIT/stem cell factor expression in premalignant and malignant lesions of the colon mucosa in relationship to disease progression and outcomes. Int J Oncol 29(4):851–859 Gao C, Li S, Zhao T, Chen J, Ren H, Zhang H, Zhao X, 2015, SCF, regulated by HIF-1α, promotes pancreatic ductal adenocarcinoma cell progression. PLoS One 10(3):e0121338 Yasuda A, Sawai H, Takahashi H, Ochi N, Matsuo Y, Funahashi H, Sato M, Okada Y, Takeyama H, Manabe T, 2006, The stem cell factor/c-kit receptor pathway enhances proliferation and invasion of pancreatic Cancer cells. Mol Cancer 5:46. , DOI 10.1186/ 1476-4598-5-46. Esposito I, Kleeff J, Bischoff SC, Fischer L, Collecchi P, Iorio M, Bevilacqua G, BuchlerMW, Friess H, 2002, The stem cell factor-ckit system and mast cells in human pancreatic Cancer. Lab Investig 82(11):1481–1492. , DOI 10.1097/01.LAB.0000036875. 21209.F9 Ulivi P, Zoli W, Medri L, Amadori D, Saragoni L, Barbanti F, Silvestrini R, 2004, C-kit and SCF expression in normal and tumor breast tissue. Breast Cancer Res Treat 83(1):33–42 Eberle F, Leinberger FH, Saulich MF, Seeger W, Engenhart- Cabillic R, Hanze J, Hattar K, Dikomey E, Subtil FSB, 2017, In Cancer cell lines inhibition of SCF/c-kit pathway leads to Radiosensitization only when SCF is strongly over-expressed. Clinical and Translational Radiation Oncology 2:69–75. https:// doi.org/10.1016/j.ctro.2017.02.001 Krishnamachary B, Stasinopoulos I, Kakkad S, PenetMF, Jacob D, Wildes F, Bhujwalla ZM(2017, Breast cancer cell cyclooxygenase- 2 expression alters extracellular matrix structure and function and numbers of cancer associated fibroblasts. Oncotarget 8(11):17981 Wang W, Bergh A, Damber JE, 2005, Cyclooxygenase-2 expression correlates with local chronic inflammation and tumor neovascularization in human prostate cancer. Clin Cancer Res 11(9):3250– 3256 Hu Z, Yang Y, Zhao Y, Huang Y, 2017, The prognostic value of cyclooxygenase-2 expression in patients with esophageal cancer: evidence from a meta-analysis. OncoTargets and therapy 10: 2893–2901 Zha S, Sauvageot J, EwingCM, Faith DA, NelsonWG, IsaacsWB, Gage WR, Saria EA, Putzi MJ, De Marzo AM, 2001, Cyclooxygenase-2 is up-regulated in proliferative inflammatory atrophy of the prostate, but not in prostate carcinoma. Cancer Res 61(24):8617–8623 Malaysiana S, Siklooksigenase PT, Prostat A, Gred M, National Cancer Registry, 2015, High expression of Cyclooxygenase-2 in high grade human prostate adenocarcinoma. In: 44, 5): 727–33 Khor LY, BaeK, Pollack A, Elizabeth H Hammond M, Grignon DJ, Venkatesan VM, Rosenthal SA et al, 2007, COX-2 expression predicts prostate-Cancer outcome: analysis of data from the RTOG 92-02 trial. Lancet Oncol 8(10):912–920. , DOI 10.1016/S1470-2045(07)70280-2 Richardsen E, Uglehus RD, Due J, Busch C, Busund LT, 2010, COX-2 is overexpressed in primary prostate cancer with metastatic potential and may predict survival. A comparison study between COX-2, TGF-β, IL-10 and, Ki67. Cancer Epidemiol 34(3):316– 322 Xu F, Li M, Zhang C, Cui J, Liu J, Li J, Jiang H, 2017, Clinicopathological and prognostic significance of COX-2 immunohistochemical expression in breast cancer: a meta-analysis. Oncotarget 8(4):6003–6012 Mukherjee R, Edwards J, Underwood MA, Bartlett JM, 2005, The relationship between angiogenesis and cyclooxygenase-2 expression in prostate cancer. BJU Int 96(1):62–66 Ceylan Y, Lekili M, Muezzinoglu T, Nese N, Isisag A, 2016, Predictive value of cyclooxygenase-2 over-expression for identifying prostate cancer from benign prostatic hyperplasia in prostate biopsy specimens. Minerva urologica e nefrologica= The Italian journal of urology and nephrology 68(3):255–262 Khodier MM, Mohmoud SA, Gabal SM, Abou-Sriea MA, 2009, Cyclooxygenase-2, COX-2, expression in Egyptian cases of benign prostatic hyperplasia and prostatic adenocarcinoma. Acad J Cancer Res 2(1):33–39 Kim BH, Kim CI, Chang HS, Choe MS, Jung HR, Kim DY, Park CH, 2011, Cyclooxygenase-2 overexpression in chronic inflammation associated with benign prostatic hyperplasia: is it related to apoptosis and angiogenesis of prostate cancer? Korean journal of urology 52(4):253–259 Madaan S, Abel PD, Chaudhary KS, Hewitt R, Stott MA, Stamp GWH, Lalani EN, 2000, Cytoplasmic induction and overexpression of cyclooxygenase-2 in human prostate cancer: implications for prevention and treatment. BJU Int 86(6):736–741 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 611 EP 624 DI 10.1007/s12253-018-0534-1 PG 14 ER PT J AU Kiraly, G Hargitai, Z Kovacs, I Szeman-Nagy, G Juhasz, I Banfalvi, G AF Kiraly, Gabor Hargitai, Zoltan Kovacs, Ilona Szeman-Nagy, Gabor Juhasz, Istvan Banfalvi, Gaspar TI Metastatic Spread from Abdominal Tumor Cells to Parathymic Lymph Nodes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Carcinoma cell line; Murine metastasis model; Parathymic lymph node; Colloidal ink ID Carcinoma cell line; Murine metastasis model; Parathymic lymph node; Colloidal ink AB Metastatic studies on rats showed that after subrenal implantation of tumor cells under the capsule of the kidney or subhepatic implantation under Glisson’s capsule of the liver generated primary tumors in these organs. It was assumed that tumor cells that escaped through the disrupted peripheral blood vessels of primary tumors entered the peritoneal cavity, crossed the diaphragm, and appeared in the thoracal, primarily in the parathymic lymph nodes. This explanation did not answer the question whether distant lymph nodes were reached via the blood stream from the primary tumor or through the thoracal lymphatic vessels. In this work, we investigated the metastatic pathway in C3H/HeJ mice, after direct intraperitoneal administration of murine SCC VII cells bypassing the hematogenic spread of tumor cells. The direct pathway was also mimicked by intraperitoneal injection of Pelican Ink colloidal particles, which appeared in the parathymic lymph nodes, similarly to the tumor cells that caused metastasis in the parathymic lymph nodes and in the thymic tissue. The murine peritoneal-parathymic lymph node route indicates a general mechanism of tumor progression from the abdominal effusion. This pathway starts with the growth of abdominal tumors, continues as thoracal metastasis in parathymic lymph nodes and may proceed as mammary lymph node metastasis. C1 [Kiraly, Gabor] University of Debrecen, Faculty of Medicine, Department of Medical Microbiology, 1 Egyetem Square, 4010 Debrecen, Hungary. [Hargitai, Zoltan] Kenezy Teaching Hospital, Department of Pathology, 2-28 Bartok Street, 4031 Debrecen, Hungary. [Kovacs, Ilona] Kenezy Teaching Hospital, Department of Pathology, 2-28 Bartok Street, 4031 Debrecen, Hungary. [Szeman-Nagy, Gabor] University of Debrecen, Faculty of Medicine, Department of Medical Microbiology, 1 Egyetem Square, 4010 Debrecen, Hungary. [Juhasz, Istvan] University of Debrecen, Department of Surgery and Operative Techniques, 98 Nagyerdei korut, 4012 Debrecen, Hungary. [Banfalvi, Gaspar] University of Debrecen, Faculty of Medicine, Department of Medical Microbiology, 1 Egyetem Square, 4010 Debrecen, Hungary. RP Juhasz, I (reprint author), University of Debrecen, Department of Surgery and Operative Techniques, 4012 Debrecen, Hungary. EM ji@med.unideb.hu CR Trencsenyi G, Kertai P, Somogyi C, Nagy G, Dombradi Z, Gacsi M, Banfalvi G, 2007, Chemically induced carcinogenesis affecting chromatin structure in rat hepatocarcinoma cells. DNA Cell Biol 26:649–655 http://online.liebertpub.com/doi/abs/10.1089/dna. 2007.0587 Trencsenyi G, Kertai P, Bako F, Hunyadi J, Marian T, Hargitai Z, Pocsi I, Muranyi E, Hornyak L, Banfalvi G, 2009, Renal capsule- Parathymic lymph node complex: a new in vivo metastatic model in rats. Anticancer Res 29:2121–2126 http://ar.iiarjournals.org/ content/29/6/2121.long Cui ZY, Ahn JS, Lee JY, Kim WS, Lim HY, Jeon HJ, Suh SW, Kim JH, Kong WH, Kang JM, Nam DH, Park K, 2006, Mouse orthotopic lung cancer model induced by PC14PE6. Cancer Res Treat 38:234–239 https://www.ncbi.nlm.nih.gov/pmc/articles/ PMC2741646/ Paget S, 1989, The distribution of secondary growths in cancer of the breast. Lancet 133(3421):571–573 https://www.ncbi.nlm.nih. gov/pubmed/2673568 Hoffman RM, 1999, Orthotopic metastatic mouse models for anticancer drug discovery and evaluation: a bridge to the clinic. Investig New Drugs 17:343–359 https://www.ncbi.nlm.nih.gov/ pubmed?term=orthotopic%20metastatic%20mouse%20model% 20for%20anticancer%20drug%20discovery%20and% 20evaluation%20a%20bridge%20to%20the%20clinic.&cmd= correctspelling Rozsa D, Trencsenyi G, Kertai P, Marian T, Nagy G, Banfalvi G, 2009, Lymphatic spread ofmesenchymal renal tumor tometastatic parathymic lymph nodes in rat. Histol Histopathol 24:1367–1379 http://www.hh.um.es/Abstracts/Vol_24/24_11/24_11_1367.htm Marco AJ, Domingo M, Ruberte J, Carretero A, Briones V, Dominguez L, 1992, Lymphatic drainage of Listeria inonocytogenes and Indian ink inoculated in the peritoneal cavity of the mouse. Lab Anim 26:200–205 http://journals.sagepub.com/ doi/abs/10.1258/002367792780740549?url_ver=Z39.88-2003&rfr_id=ori%3Arid%3Acrossref.org&rfr_dat=cr_pub% 3Dpubmed& Banfalvi G, 2012a, Role of parathymic lymph nodes in metastatic tumor development. Cancer Metastasis Rev 31:89–97 https://link. springer.com/article/10.1007%2Fs10555-011-9331-y Steer HW, Lewis DA, 1983, Peritoneal cell responses to acute gastro-intestinal inflammation. J Pathol 140:237–253 http:// onlinelibrary.wiley.com/doi/10.1002/path.1711400306/full Jian J, Liu C, Gong Y, Su L, Zhang B, Wang Z, Wang D, Zhou Y, Xu F, Li P, Zheng Y, Song L, Zhou X, 2014, India ink incorporated multifunctional phase-transition Nanodroplets for photoacoustic/ ultrasound dual-modality imaging and photoacoustic effect based tumor therapy. Theranostics 4(10):1026–1038 http://www.thno.org/ v04p1026.htm Rafferty P, Egenolf D, Brosnan K, Makropoulos D, Jordan J, Meshaw K, Walker M, Volk A, Bugelski PJ, 2012, Immunotoxicologic effects of cyclosporine on tumor progression in models of squamous cell carcinoma and B-cell lymphoma in C3H mice. J Immunotoxicol 9:43–55 http://www.tandfonline. com/doi/full/10.3109/1547691X.2011.614646 Moore A, Sergeyev N, Bredow S, Weissleder R, 1998, A model system to quantitate tumor burden in locoregional lymph nodes during cancer spread. Invasion Metastasis 18:192–197 https:// www.ncbi.nlm.nih.gov/pubmed/10640905 Morris B, Courtice FC, 1977, Cells and immunoglobulins in lymph. Lymphology 10:62–69 https://www.ncbi.nlm.nih.gov/ pubmed/329011 Miller JF, 1963, Role of the thymus in immunity. Brit Med J 24 2(5355):459–464. https://www.ncbi.nlm.nih.gov/pmc/articles/ PMC1874012/ Bonney WM, Battenberg JD, 1967, Transthoracic thymectomy in rats. Transplantation 5(3):544–546. https://www.ncbi.nlm.nih.gov/ pubmed/?term=Bonney+WM%2C+Battenberg+JD.+ Transthoracic+thymectomy+in+rats Blau JN, Gaugas JM, 1968, Parathymic lymph nodes in rats and mice. Immunology 14:763–765 https://www.ncbi.nlm.nih.gov/ pubmed/?term=Blau+JN%2C+Gaugas+JM.+Parathymic+lymph+ nodes+in+rats+and+mice Tanegashima A, Yamashita A, Yamamoto H, Fukunaga T, 1999, Human parathymic lymph node: morphological and functional significance. Immunology 97(2):301–308 https://www.ncbi.nlm.nih. gov/pmc/articles/PMC2326821/ Severeanu G, 1909, Die Lymphgefaße der Thymus. Arch Anat Entw Gesch 93 Siegler R, 1669-1678, Rich MA, 1963, unilateral histogenesis of AKR thymic lymphoma. Cancer Res Volume 23:10(1, http:// cancerres.aacrjournals.org/content/23/10_Part_1/1669.long Workman P, Twentyman P, Balkwill F, 1988, United Kingdom coordinating committee on Cancer research, UKCCCR, guidelines for the welfare of animals in experimental neoplasia, second edition). Br J Cancer 77(1):1–10 https://ciepal-azur.unice.fr/ Oncology%20animal%20guides.pdf Kanazawa H, Rapacchietta D, Kallman RF, 1988, Scheduledependent therapeutic gain from the combination of fractionated irradiation and cis-diamminedichloroplatinum, II, in C3H/km mouse model systems. Cancer Res 48:3158–3164 http:// cancerres.aacrjournals.org/content/48/11/3158.short Dunn TB, 1954, Normal and pathologic anatomy of the reticular tissue in laboratory mice, with a classification and discussion of neoplasms. J Natl Cancer Inst 14(6):1281–1433 https://www.ncbi. nlm.nih.gov/pubmed/13233863 Glomset DA, 1938, The incidence of metastasis of malignant tumors to the adrenals. Am J Cancer 32:57–61 http://cancerres. aacrjournals.org/content/amjcancer/32/1/57.full.pdf Trencsenyi G, Marian T, Bako F, Emri M, Nagy G, Kertai P, Banfalvi G, 2014a, Metastatic hepatocarcinoma he/De tumor model in rat. J Cancer 5(7):548–558. , DOI 10.7150/jca.9315 Trencsenyi G, Nagy G, Kahlik B, Nemeth E, Kertai P, Kiss A, Banfalvi G, 2014b, Lymphoid metastasis of rat My2/De leukemia. Leuk Res 38:586–593. , DOI 10.1016/j.leukres.2014.02. 006 Pitt ML, Anderson AO, 1988, Direct transdiaphragmatic traffic of peritoneal macrophages to the lung. Adv Exp Med Biol 237:627– 632 https://link.springer.com/chapter/10.1007/978-1-4684-5535- 9_95 MacCallum WG, 1903, On the mechanism of absorption of granular materials from the peritoneum. Bull Johns Hopkins Hosp 14: 105–115 Olin T, Saldeen T, 1964, The lymphatic pathways from the peritoneal cavity: a lymphangiographic study in the rat. Cancer Res 24: 1700–1711 http://cancerres.aacrjournals.org/content/24/10/1700. long Banfalvi G, 2012b, Metastatic view of breast cancer. Cancer Metastasis Rev 31:815–822. , DOI 10.1007/s10555-012- 9392-6 Gray H, Pickering PT, Howden R, 1974, Gray’s anatomy. Philadelphia Courage Books Handley RS, Thackray AC, 1954, Invasion of the internal mammary glands in carcinoma of the breast. Br J Cancer 1:15–20 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 625 EP 633 DI 10.1007/s12253-018-0492-7 PG 9 ER PT J AU Ou, X Zhang, Gt Xu, Z Chen, Js Xie, Y Liu, JK Liu, XP AF Ou, Xi Zhang, Guang-tao Xu, Zhe Chen, Jing-sen Xie, Yong Liu, Ji-Kui Liu, Xiao-Ping TI Desumoylating Isopeptidase 2 (DESI2) Inhibits Proliferation and Promotes Apoptosis of Pancreatic Cancer Cells through Regulating PI3K/AKT/mTOR Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Desumoylating isopeptidase 2; Pancreatic cancer; Phosphatidylinositide 3-kinases; AKT; mTOR ID Desumoylating isopeptidase 2; Pancreatic cancer; Phosphatidylinositide 3-kinases; AKT; mTOR AB This study aimed to investigate the effects of desumoylating isopeptidase 2 (DESI2) on tumor cell proliferation, apoptosis and invasion of pancreatic cancer, and to assess the signaling pathway involved. Overexpression and silence of DESI2 were designed and the experiments were divided into 5 groups: a normal control group, an interference control group (shRNA-NC); an interference group (sh-DESI2); an overexpression control group (NC), an overexpression group (DESI2). Quantitative real time polymerase chain reaction (qRT-PCR) was used to screen the appropriate interference sequence. The silencing and overexpression of DESI2 were confirmed by qRT-PCR and western blotting. Cell cycling, apoptosis, invasion, and the expression of phosphatidylinositol-3-kinase (PI3K)-protein kinase B (AKT)-mammalian target of rapamycin (mTOR) pathway and caspase 3 were measured. Overexpression and silence of DESI2 were successfully designed in two pancreatic cancer cells, and the interference effect of sh-DESI2–3 showed the best silencing effects. The biological activities of DESI2 were detected in both ASPC-1 and PANCE-1 cells. Our results showed that cell proliferation was significantly increased in the sh-DESI2 group, while decreased in DESI2 group compared with the control group in both cell lines. In ASPC-1 cells, the events in G1 phase decreased and in S phase increased obviously in the sh-DESI2 group, compared with control group. An opposite result was found when DESI2 was overexpressed. In PANCE-1 cells, the events in G2 phase were higher in the sh-DESI2 group, while in the DESI2 group was significantly lower than that in control group. In ASPC-1 and PANCE-1 cells, sh-DESI2 group showed decreased apoptosis, increased cell invasion and increased expression of AKT, p-Akt, PI3K, p-PI3K, p-mTOR and mTOR and decreased caspase 3 expression compared with the control group, while overexpression of DESI2 leaded to increased apoptosis, decreased cell invasion and reduced expression of AKT, p-Akt, PI3K, p-PI3K, p-mTOR and mTOR and increased expression of caspase 3. DESI2 regulates the proliferation and apoptosis of pancreatic cancer cells through PI3K/AKT/mTOR signaling pathway. C1 [Ou, Xi] Peking University Shenzhen Hospital, Department of Hepatopancreatobiliary Surgery, Lian Hua Road 1120, 518036 Shenzhen, China. [Zhang, Guang-tao] Peking University Shenzhen Hospital, Department of Hepatopancreatobiliary Surgery, Lian Hua Road 1120, 518036 Shenzhen, China. [Xu, Zhe] The Third People’s Hospital of Shenzhen, The Third Ward of Liver Disease, 518112 Shenzhen, China. [Chen, Jing-sen] Shenzhen Maternity and Child Healthcare Hospital, Breast Department, 518028 Shenzhen, China. [Xie, Yong] Peking University Shenzhen Hospital, Department of Hepatopancreatobiliary Surgery, Lian Hua Road 1120, 518036 Shenzhen, China. [Liu, Ji-Kui] Peking University Shenzhen Hospital, Department of Hepatopancreatobiliary Surgery, Lian Hua Road 1120, 518036 Shenzhen, China. [Liu, Xiao-Ping] Peking University Shenzhen Hospital, Department of Hepatopancreatobiliary Surgery, Lian Hua Road 1120, 518036 Shenzhen, China. RP Liu, JK (reprint author), Peking University Shenzhen Hospital, Department of Hepatopancreatobiliary Surgery, 518036 Shenzhen, China. EM liu89201@sina.com CR Seipel AH, Wiklund F, Wiklund NP, Egevad L, 2013, Histopathological features of ductal adenocarcinoma of the prostate in 1,051 radical prostatectomy specimens. Virchows Arch 462(4): 429–436. , DOI 10.1007/s00428-013-1385-5 Lin C, Yan H, Yang J, Li L, Tang M, Zhao X, Nie C, Luo N,Wei Y, Yuan Z, 2017, Combination of DESI2 and IP10 gene therapy significantly improves therapeutic efficacy against murine carcinoma. Oncotarget 8(34):56281–56295. , DOI 10.18632/ oncotarget.17623 Shen CC, Cui XY, He Y, Kang YH, Yi C, Yang JL, Gou LT, 2015, High phosphorylation status of AKT/mTOR signal in DESI2-reduced pancreatic ductal adenocarcinoma. Pathol Oncol Res 21(2):267–272. , DOI 10.1007/s12253-014-9817-3 Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, Harris M, Reni M, Dowden S, Laheru D, Bahary N, Ramanathan RK, Tabernero J, Hidalgo M, Goldstein D, Van Cutsem E, Wei X, Iglesias J, Renschler MF, 2013, Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 369(18):1691– 1703. , DOI 10.1056/NEJMoa1304369 Janku F, Wheler JJ,Westin SN,Moulder SL, Naing A, Tsimberidou AM, Fu S, Falchook GS, Hong DS, Garrido-Laguna I, Luthra R, Lee JJ, Lu KH, Kurzrock R, 2012, PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations. J Clin Oncol Off J Am Soc Clin Oncol 30(8): 777–782. , DOI 10.1200/JCO.2011.36.1196 Zhu G, Liu Y,Wang Y, Bi X, BaudryM(2015, Different patterns of electrical activity lead to long-term potentiation by activating different intracellular pathways. J Neurosci 35(2):621–633. https://doi. org/10.1523/JNEUROSCI.2193-14.2015 Li J, Yang S, Zhu G, 2017, Postnatal calpain inhibition elicits cerebellar cell death and motor dysfunction. Oncotarget 8(50): 87997–88007. , DOI 10.18632/oncotarget.21324 Lastwika KJ,WilsonW3rd, Li QK, Norris J, Xu H, Ghazarian SR, Kitagawa H, Kawabata S, Taube JM, Yao S, Liu LN, Gills JJ, Dennis PA, 2016, Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung Cancer. Cancer Res 76(2):227–238. , DOI 10.1158/0008-5472. CAN-14-3362 Li J, Liang X, Yang X, 2012, Ursolic acid inhibits growth and induces apoptosis in gemcitabine-resistant human pancreatic cancer via the JNKand PI3K/Akt/NF-kappaB pathways. Oncol Rep 28(2): 501–510. , DOI 10.3892/or.2012.1827 Zhu G, Wang X, Wu S, Li Q, 2012, Involvement of activation of PI3K/Akt pathway in the protective effects of puerarin against MPP+−induced human neuroblastoma SH-SY5Y cell death. Neurochem Int 60(4):400–408. , DOI 10.1016/j.neuint. 2012.01.003 Strausberg RL, Feingold EA, Grouse LH, Derge JG, Klausner RD, Collins FS, Wagner L, Shenmen CM, Schuler GD, Altschul SF, Zeeberg B, Buetow KH, Schaefer CF, Bhat NK, Hopkins RF, Jordan H, Moore T, Max SI, Wang J, Hsieh F, Diatchenko L, Marusina K, Farmer AA, Rubin GM, Hong L, Stapleton M, Soares MB, Bonaldo MF, Casavant TL, Scheetz TE, Brownstein MJ, Usdin TB, Toshiyuki S, Carninci P, Prange C, Raha SS, Loquellano NA, Peters GJ, Abramson RD, Mullahy SJ, Bosak SA, McEwan PJ, McKernan KJ, Malek JA, Gunaratne PH, Richards S, Worley KC, Hale S, Garcia AM, Gay LJ, Hulyk SW, Villalon DK,MuznyDM, Sodergren EJ, Lu X, Gibbs RA, Fahey J, Helton E, Ketteman M, Madan A, Rodrigues S, Sanchez A, Whiting M, Madan A, Young AC, Shevchenko Y, Bouffard GG, Blakesley RW, Touchman JW, Green ED, Dickson MC, Rodriguez AC, Grimwood J, Schmutz J, Myers RM, Butterfield YS, Krzywinski MI, Skalska U, Smailus DE, Schnerch A, Schein JE, Jones SJ, Marra MA, Mammalian Gene Collection Program T, 2002, Generation and initial analysis of more than 15,000 fulllength human and mouse cDNA sequences. Proc Natl Acad Sci U S A 99(26):16899–16903. , DOI 10.1073/pnas.242603899 Yan F, Ruan XZ, Yang HS, Yao SH, Zhao XY, Gou LT, Ma FX, Yuan Z, Deng HX, Wei YQ, 2010, Identification, characterization, and effects of Xenopus laevis PNAS-4 gene on embryonic development. J Biomed Biotechnol 2010:134764–134769. https://doi. org/10.1155/2010/134764 Yan F, Gou L, Yang J, Chen L, Tong A, Tang M, Yuan Z, Yao S, Zhang P, Wei Y, 2009, A novel pro-apoptosis gene PNAS4 that induces apoptosis in A549 human lung adenocarcinoma cells and inhibits tumor growth in mice. Biochimie 91(4):502–507. https:// doi.org/10.1016/j.biochi.2008.12.002 Yuan Z, GuoW, Yang J, Li L,WangM, Lei Y,Wan Y, Zhao X, Luo N, Cheng P, Liu X, Nie C, Peng Y, Tong A,Wei Y, 2015, PNAS-4, an early DNA damage response gene, induces S phase arrest and apoptosis by activating checkpoint kinases in lung Cancer cells. J Biol Chem 290(24):14927–14944. , DOI 10.1074/jbc. M115.658419 Li L, Chen DB, Lin C, Cao K, Wan Y, Zhao XY, Nie CL, Yuan Z, Wei YQ, 2013, hPNAS-4 inhibits proliferation through S phase arrest and apoptosis: underlying action mechanism in ovarian cancer cells. Apoptosis 18(4):467–479. , DOI 10.1007/ s10495-012-0797-z Roy SK, Srivastava RK, Shankar S, 2010, Inhibition of PI3K/AKT and MAPK/ERK pathways causes activation of FOXO transcription factor, leading to cell cycle arrest and apoptosis in pancreatic cancer. J Mol Signal 5:10. , DOI 10.1186/1750-2187-5-10 Yuan Z, Yan F,Wang YS, Liu HY, Gou LT, Zhao XY, Lai ST, Deng HX, Li J, Ding ZY, Xiong SQ, Kan B, Mao YQ, Chen LJ,Wei YQ, Zhao X, 2009, PNAS-4, a novel pro-apoptotic gene, can potentiate antineoplastic effects of cisplatin. Cancer Chemother Pharmacol 65(1):13–25. , DOI 10.1007/s00280-009-0998-5 Qi X, Song X, Liu P, Yi T, Li S, Xie C, Zheng Y, Bai Y, Sun C,Wei Y, Zhao X, 2011, Antitumor effects of PLGA nanoparticles encapsulating the human PNAS-4 gene combined with cisplatin in ovarian cancer. Oncol Rep 26(3):703–710. , DOI 10.3892/or. 2011.1337 Hou S, Zhao Z, Yan F, Chen X, Deng H, ChenX, Wang Y, Wei Y, 2009, Genetic transfer of PNAS-4 induces apoptosis and enhances sensitivity to gemcitabine in lung cancer. Cell Biol Int 33(3):276– 282. , DOI 10.1016/j.cellbi.2008.11.014 Yuan Z, Liu H, Yan F,Wang Y, Gou L, Nie C, Ding Z, Lai S, Zhao Y, Zhao X, Li J, Deng H, Mao Y, Chen L, Wei Y, Zhao X, 2009, Improved therapeutic efficacy against murine carcinoma by combining honokiol with gene therapy of PNAS-4, a novel proapoptotic gene. Cancer Sci 100(9):1757–1766. , DOI 10. 1111/j.1349-7006.2009.01242.x Flanagan L, Meyer M, Fay J, Curry S, Bacon O, Duessmann H, John K, Boland KC, McNamara DA, Kay EW, Bantel H, Schulze- Bergkamen H, Prehn JH, 2016, Low levels of Caspase-3 predict favourable response to 5FU-based chemotherapy in advanced colorectal cancer: Caspase-3 inhibition as a therapeutic approach. Cell Death Dis 7:e2087. , DOI 10.1038/cddis.2016.7 Baharara J, Ramezani T, Divsalar A, Mousavi M, Seyedarabi A, 2016, Induction of apoptosis by green synthesized gold nanoparticles through activation of Caspase-3 and 9 in human cervical Cancer cells. Avicenna J Med Biotechnol 8(2):75–83 Dolka I, Krol M, Sapierzynski R, 2016, Evaluation of apoptosisassociated protein, Bcl-2, Bax, cleaved caspase-3 and p53, expression in canine mammary tumors: an immunohistochemical and prognostic study. Res Vet Sci 105:124–133. , DOI 10. 1016/j.rvsc.2016.02.004 Xu Z, Zhang F, Bai C, Yao C, Zhong H, Zou C, Chen X, 2017, Sophoridine induces apoptosis and S phase arrest via ROSdependent JNK and ERK activation in human pancreatic cancer cells. J Exp Clin Cancer Res 36(1):124. , DOI 10.1186/ s13046-017-0590-5 Cheng X,Wang B, Jin Z, Ma D, YangW, Zhao R, Jing X, Shen B, Peng C, Qiu W, 2016, Pseudomonas aeruginosa-mannosesensitive hemagglutinin inhibits pancreatic cancer cell proliferation and induces apoptosis via the EGFR pathway and caspase signaling. Oncotarget 7(47):77916–77925. , DOI 10.18632/ oncotarget.12844 Chiu TL, Su CC, 2017, Tanshinone IIA increases protein expression levels of PERK, ATF6, IRE1alpha, CHOP, caspase3 and caspase12 in pancreatic cancer BxPC3 cellderived xenograft tumors. Mol Med Rep 15(5):3259–3263. , DOI 10.3892/mmr. 2017.6359 Castel P, Toska E, Zumsteg ZS, Carmona FJ, Elkabets M, Bosch A, Scaltriti M, 2014, Rationale-based therapeutic combinations with PI3K inhibitors in cancer treatment. Mol Cell Oncol 1(3):e963447. , DOI 10.4161/23723548. 2014.963447 Wang MC, Jiao M, Wu T, Jing L, Cui J, Guo H, Tian T, Ruan ZP, Wei YC, Jiang LL, Sun HF, Huang LX, Nan KJ, Li CL, 2016, Polycomb complex protein BMI-1 promotes invasion and metastasis of pancreatic cancer stem cells by activating PI3K/AKT signaling, an ex vivo, in vitro, and in vivo study. Oncotarget 7(8):9586– 9599. , DOI 10.18632/oncotarget.7078 Shimizu T, Tolcher AW, Papadopoulos KP, Beeram M, Rasco DW, Smith LS, Gunn S, Smetzer L, Mays TA, Kaiser B, Wick MJ, Alvarez C, Cavazos A,Mangold GL, Patnaik A, 2012, The clinical effect of the dual-targeting strategy involving PI3K/AKT/mTOR and RAS/MEK/ERK pathways in patients with advanced cancer. Clin Cancer Res 18(8):2316–2325. , DOI 10.1158/1078- 0432.CCR-11-2381 Gao N, Flynn DC, Zhang Z, Zhong XS, Walker V, Liu KJ, Shi X, JiangBH(2004, G1 cell cycle progression and the expression ofG1 cyclins are regulated by PI3K/AKT/mTOR/p70S6K1 signaling in human ovarian cancer cells. Am J Physiol Cell Physiol 287(2): C281–C291. , DOI 10.1152/ajpcell.00422.2003 Liu CY, HuMH, Hsu CJ, Huang CT,Wang DS, TsaiWC, Chen YT, Lee CH, Chu PY, Hsu CC, Chen MH, Shiau CW, Tseng LM, Chen KF, 2016, Lapatinib inhibits CIP2A/PP2A/p-Akt signaling and induces apoptosis in triple negative breast cancer cells. Oncotarget 7(8):9135–9149. , DOI 10.18632/oncotarget.7035 Chen J, Shao R, Li F, Monteiro M, Liu JP, Xu ZP, Gu W, 2015, PI3K/Akt/mTOR pathway dual inhibitor BEZ235 suppresses the stemness of colon cancer stem cells. Clin Exp Pharmacol Physiol 42(12):1317–1326. , DOI 10.1111/1440-1681.12493 Eser S, Schnieke A, Schneider G, Saur D, 2014, Oncogenic KRAS signalling in pancreatic cancer. Br J Cancer 111(5):817–822. https:// doi.org/10.1038/bjc.2014.215 Cheng HB, Bo Y, Shen WX, Ren XG, Tan JN, Jia ZR, Xu CL, 2015, Longikaurin E induces apoptosis of pancreatic cancer cells via modulation of the p38 and PI3K/AKT pathways by ROS. Naunyn Schmiedeberg's Arch Pharmacol 388(6):623–634. https:// doi.org/10.1007/s00210-015-1107-4 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 635 EP 646 DI 10.1007/s12253-018-0487-4 PG 12 ER PT J AU Takacs, F Tolnai-Kriston, Cs Hernadfoi, M Szabo, O Szaloki, G Szepesi, Czeti, Matolcsy, A Barna, G AF Takacs, Ferenc Tolnai-Kriston, Csilla Hernadfoi, Mark Szabo, Orsolya Szaloki, Gabor Szepesi, Agota Czeti, Agnes Matolcsy, Andras Barna, Gabor TI The Effect of CD86 Expression on the Proliferation and the Survival of CLL Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CLL; CD86; Micro-environment; Proliferation marker ID CLL; CD86; Micro-environment; Proliferation marker AB Micro-environment plays important role in the pathogenesis of CLL by providing protective niche for CLL cells. Several molecules play important role in communication between CLL cells and immune cells like CD86.Some of the data suggest that CLL patients with high CD86 level need earlier treatments and cells with higher CD86 expression has higher proliferation rate but the role of CD86 in the survival and proliferation of CLL cells is unclear. We investigated the effect of CD86 expression to CLL cells in 50 peripheral blood and 15 lymph node biopsy samples from CLL patients. Our results showed that the expressions of CD86 increased significantly after 7 day culturing in medium, or in the presence of bone marrow stromal cells (BMSCs).We found positive correlation between CD86 and CD23 expression (p < 0.05), but no correlation with other markers. Furthermore, no correlation were found between the CD86 expression and the proliferation of CLL cells. Analysis of clinical data showed that cases with high CD86 expression had lower level of serum lymphocyte count (p < 0.04) at the time of the diagnosis. CD86 shows multiple appearances in the lymph nodes containing pseudofollicules, but no correlation was found between CD86 positivity, and Ki67 positivity. Our results suggest that the use of CD86 molecule as a proliferation marker for CLL is highly questionable. However, the CD86 molecule may interfere with the immune system of patients with CLL by activating and depleting immune functions. That can be the reason why CD86 positivity may mean worse prognosis. C1 [Takacs, Ferenc] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Tolnai-Kriston, Csilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Hernadfoi, Mark] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Szabo, Orsolya] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Szaloki, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Szepesi, Agota] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Czeti, Agnes] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Matolcsy, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Barna, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. RP Barna, G (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM barna.gabor@med.semmelweis-univ.hu CR Zhang S,Kipps TJ, 2014, The pathogenesis of chronic lymphocytic leukemia. Annu Rev Pathol 9:103–118 HallekMet al, 2008, Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood 111(12):5446–5456 Gorczyca W, 2010, Flow Cytometry in Neoplastic Hematology. Informa Healthcare London, London Burger JA et al, 2000, Blood-derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell-derived factor-1. Blood 96(8):2655–2663 Panayiotidis P et al, 1996, Human bone marrow stromal cells prevent apoptosis and support the survival of chronic lymphocytic leukaemia cells in vitro. Br J Haematol 92(1):97–103 Plander M et al, 2011, Chronic lymphocytic leukemia cells induce anti-apoptotic effects of bone marrow stroma. Ann Hematol 90(12): 1381–1390 Alhakeem SS et al, 2018, Chronic Lymphocytic Leukemia- Derived IL-10 Suppresses Antitumor Immunity. J Immunol 200(12):4180–4189 Joss A et al, 2000, IL-10 directly acts on T cells by specifically altering the CD28 co-stimulation pathway. Eur J Immunol 30(6): 1683–1690 Brzostek J, Gascoigne NR, Rybakin V, 2016, Cell Type-Specific Regulation of Immunological Synapse Dynamics by B7 Ligand Recognition. Front Immunol 7:24 Dai ZS et al, 2009, Defective expression and modulation of B7–2/ CD86 on B cells in B cell chronic lymphocytic leukemia. Int J Hematol 89(5):656–663 Huemer M et al, 2014, AID induces intraclonal diversity and genomic damage in CD86(+, chronic lymphocytic leukemia cells. Eur J Immunol 44(12):3747–3757 Swerdlow SH et al, 2016, The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 127(20): 2375–2390 Kriston C et al, 2018, In contrast to high CD49d, low CXCR4 expression indicates the dependency of chronic lymphocytic leukemia, CLL, cells on the microenvironment. Ann Hematol 97(11): 2145–2152 Kim SH, Lee CE, 2011, Counter-regulationmechanismof IL-4 and IFN-α signal transduction through cytosolic retention of the pYSTAT6: pY-STAT2:p48 complex. Eur J Immunol 41(2):461–472 Deszo EL et al, 2004, IL-4-dependent CD86 expression requires JAK/STAT6 activation and is negatively regulated by PKCdelta. Cell Signal 16(2):271–280 Hock BD, MacPherson SA, McKenzie JL, 2017, Idelalisib and caffeine reduce suppression of T cell responses mediated by activated chronic lymphocytic leukemia cells. PLoS One 12(3): e0172858 Gerdes J et al, 1983, Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer 31(1):13–20 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 647 EP 652 DI 10.1007/s12253-018-0512-7 PG 6 ER PT J AU Li, Z Zhang, F Li, Y Qi, X Guo, Y AF Li, Zhang Zhang, Fengxiang Li, Yanxin Qi, Xiangjun Guo, Yaming TI Triiodothyronine Promotes Cell Proliferation of Breast Cancer via Modulating miR-204/Amphiregulin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; T3; Cell viability; miR-204; AREG ID Breast cancer; T3; Cell viability; miR-204; AREG AB Breast cancer (BC) severely threatens women’s life, and Triiodothyronine (T3) shows a positive role on BC cell proliferation, while the potential mechanism underlying it is still unclear. T3 was used to stimulate BC cell lines MCF-7 and T47-D. Real-time PCR was performed to determine the expression of miRNAs, while western blot was used to measure protein expression of Amphiregulin (AREG), AKT and p-AKT. The interaction between miR-204 and AREG was determined using luciferase reporter assay. MTT was performed to detect cell viability. The expression of miR-204 was decreased, while AREG and p-AKT was increased in T3 stimulated BC cell lines. T3 stimulation promoted cell viability. miR-204 targets AREG to regulate its expression. T3 promoted expression of AREG and p-AKT, while miR-204 overexpression reversed the effect of T3, however, pcDNA AREG transfection abolished the effect of miR-204 mimic. T3 promoted cell viability of BC cells via modulating the AKT signaling pathway. The detailed mechanism was that the down-regulated miR-204 that induced by T3 stimulation promoted the expression of AREG, the up-regulated AREG activated AKT signaling pathway, while the activated AKT signaling promoted cell proliferation. C1 [Li, Zhang] Tongliao City Hospital, Department of Breast and Thyroid Surgery, No.668 Keerqin Rd., Keerqin District, 028000 Tongliao, Inner Mongolia, China. [Zhang, Fengxiang] Tongliao City Hospital, Department of Breast and Thyroid Surgery, No.668 Keerqin Rd., Keerqin District, 028000 Tongliao, Inner Mongolia, China. [Li, Yanxin] Tongliao City Hospital, Department of Breast and Thyroid Surgery, No.668 Keerqin Rd., Keerqin District, 028000 Tongliao, Inner Mongolia, China. [Qi, Xiangjun] Tongliao City Hospital, Department of Breast and Thyroid Surgery, No.668 Keerqin Rd., Keerqin District, 028000 Tongliao, Inner Mongolia, China. [Guo, Yaming] Tongliao City Hospital, Department of Breast and Thyroid Surgery, No.668 Keerqin Rd., Keerqin District, 028000 Tongliao, Inner Mongolia, China. RP Li, Z (reprint author), Tongliao City Hospital, Department of Breast and Thyroid Surgery, 028000 Tongliao, China. EM wupi5885@163.com CR Galton VA, Schneider MJ, Clark AS, St Germain DL, 2009, Life without thyroxine to 3,5,3′-triiodothyronine conversion: studies in mice devoid of the 5′-deiodinases. Endocrinology 150:2957–2963 Gereben B, Zavacki AM, Ribich S, Kim BW, Huang SA, SimonidesWS, Zeold A, Bianco AC, 2008, Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling. Endocr Rev 29:898–938 Saraiva PP, Figueiredo NB, Padovani CR, BrentaniMM, Nogueira CR, 2005, Profile of thyroid hormones in breast cancer patients. Braz J Med Biol Res 38:761–765 Dinda S, Sanchez A, Moudgil V, 2002, Estrogen-like effects of thyroid hormone on the regulation of tumor suppressor proteins, p53 and retinoblastoma, in breast cancer cells. Oncogene 21:761– 768 Conde I, Paniagua R, Zamora J, Blanquez MJ, Fraile B, Ruiz A, ArenasMI, 2006, Influence of thyroid hormone receptors on breast cancer cell proliferation. Ann Oncol 17:60–64 Hall LC, Salazar EP, Kane SR, Liu N, 2008, Effects of thyroid hormones on human breast cancer cell proliferation. J Steroid Biochem Mol Biol 109:57–66 Rohini M, Gokulnath M, Miranda PJ, Selvamurugan N, 2018, MiR-590-3p inhibits proliferation and promotes apoptosis by targeting activating transcription factor 3 in human breast Cancer cells. Biochimie 154:10–18 Yan L, YuMC, Gao GL, Liang HW, Zhou XY, Zhu ZT, Zhang CY, Wang YB, Chen X, 2018, MiR-125a-5p functions as a tumour suppressor in breast cancer by downregulating BAP1. J Cell Biochem 119:8773–8783 Huang Q, Gumireddy K, Schrier M, le Sage C, Nagel R, Nair S, Egan DA, Li A, Huang G, Klein-Szanto AJ, Gimotty PA, Katsaros D, Coukos G, Zhang L, Pure E, Agami R, 2008, The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat Cell Biol 10:202–210 Chen J, Wang BC, Tang JH, 2012, Clinical significance of microRNA-155 expression in human breast cancer. J Surg Oncol 106:260–266 Dinami R, Ercolani C, Petti E, Piazza S, Ciani Y, Sestito R, Sacconi A, Biagioni F, le Sage C, Agami R, Benetti R, Mottolese M, Schneider C, Blandino G, Schoeftner S, 2014, miR-155 drives telomere fragility in human breast cancer by targeting TRF1. Cancer Res 74:4145–4156 Mackiewicz M, Huppi K, Pitt JJ, Dorsey TH, Ambs S, Caplen NJ, 2011, Identification of the receptor tyrosine kinase AXL in breast cancer as a target for the humanmiR-34a microRNA. Breast Cancer Res Treat 130:663–679 Piovan C, Palmieri D, Di Leva G, Braccioli L, Casalini P, Nuovo G, Tortoreto M, Sasso M, Plantamura I, Triulzi T, Taccioli C, Tagliabue E, Iorio MV, Croce CM, 2012, Oncosuppressive role of p53-induced miR-205 in triple negative breast cancer. Mol Oncol 6:458–472 BaiWD,YeXM, ZhangMY, ZhuHY,Xi WJ, HuangX, Zhao J,Gu B, Zheng GX, Yang AG, Jia LT, 2014)MiR-200c suppresses TGFbeta signaling and counteracts trastuzumab resistance and metastasis by targeting ZNF217 and ZEB1 in breast cancer. Int J Cancer 135:1356–1368 Fu J, Xu X, Kang L, Zhou L,Wang S, Lu J, Cheng L, Fan Z, Yuan B, Tian P, Zheng X, Yu C, Ye Q, Lv Z, 2014, miR-30a suppresses breast cancer cell proliferation and migration by targeting Eya2. Biochem Biophys Res Commun 445:314–319 Li W, Jin X, Zhang Q, Zhang G, Deng X, Ma L, 2014, Decreased expression of miR-204 is associated with poor prognosis in patients with breast cancer. Int J Clin Exp Pathol 7:3287–3292 Shen SQ, Huang LS, Xiao XL, Zhu XF, Xiong DD, Cao XM, Wei KL, Chen G, Feng ZB, 2017, miR-204 regulates the biological behavior of breast cancer MCF-7 cells by directly targeting FOXA1. Oncol Rep 38:368–376 Willmarth NE, Ethier SP, 2008, Amphiregulin as a novel target for breast cancer therapy. J Mammary Gland Biol Neoplasia 13:171– 179 Figueiredo NB, Cestari SH, Conde SJ, Luvizotto RA, De SibioMT, Perone D, KatayamaML, Carraro DM, Brentani HP, BrentaniMM, Nogueira CR, 2014, Estrogen-responsive genes overlap with triiodothyronine-responsive genes in a breast carcinoma cell line. ScientificWorldJournal 2014:969404 Wang X, Masri S, Phung S, Chen S, 2008, The role of amphiregulin in exemestane-resistant breast cancer cells: evidence of an autocrine loop. Cancer Res 68:2259–2265 Kondapaka SB, Fridman R, Reddy KB, 1997, Epidermal growth factor and amphiregulin up-regulate matrix metalloproteinase-9, MMP-9, in human breast cancer cells. Int J Cancer 70:722–726 Sacconi A, Biagioni F, Canu V, Mori F, Di Benedetto A, Lorenzon L, Ercolani C, Di Agostino S, Cambria AM, Germoni S, Grasso G, Blandino R, Panebianco V, Ziparo V, Federici O, Muti P, Strano S, Carboni F, Mottolese M, Diodoro M, Pescarmona E, Garofalo A, Blandino G, 2012, miR-204 targets Bcl-2 expression and enhances responsiveness of gastric cancer. Cell Death Dis 3:e423 Xia Z, Liu F, Zhang J, Liu L, 2015, Decreased expression of MiRNA-204-5p contributes to glioma progression and promotes glioma cell growth, migration and invasion. PLoS One 10: e0132399 Wu ZY, Wang SM, Chen ZH, Huv SX, Huang K, Huang BJ, Du JL, Huang CM, Peng L, Jian ZX, Zhao G, 2015, MiR-204 regulates HMGA2 expression and inhibits cell proliferation in human thyroid cancer. Cancer Biomark 15:535– 542 Guo W, Zhang Y, Zhang Y, Shi Y, Xi J, Fan H, Xu S, 2015, Decreased expression of miR-204 in plasma is associated with a poor prognosis in patients with non-small cell lung cancer. Int J Mol Med 36:1720–1726 Wu L, Chen Z, Xing Y, 2018, MiR-506-3p inhibits cell proliferation, induces cell cycle arrest and apoptosis in retinoblastoma by directly targeting NEK6. Cell Biol Int. , DOI 10.1002/cbin. 11041 Zhou S, Li S, Zhang W, Tong H, Li S, Yan Y, 2018, MiR-139 promotes differentiation of bovine skeletal muscle-derived satellite cells by regulating DHFR gene expression. J Cell Physiol 234(1): 632–641 Frasor J, Stossi F, Danes JM, Komm B, Lyttle CR, Katzenellenbogen BS, 2004, Selective estrogen receptor modulators: discrimination of agonistic versus antagonistic activities by gene expression profiling in breast cancer cells. Cancer Res 64: 1522–1533 Ghazanchaei A, Mansoori B, Mohammadi A, Biglari A, Baradaran B, 2018, Restoration of miR-152 expression suppresses cell proliferation, survival, and migration through inhibition of AKT-ERK pathway in colorectal cancer. J Cell Physiol 234:769–776 Li X, Liu H, Wang J, Qin J, Bai Z, Chi B, Yan W, Chen X, 2018, Curcumol induces cell cycle arrest and apoptosis by inhibiting IGF- 1R/PI3K/Akt signaling pathway in human nasopharyngeal carcinoma CNE-2 cells. Phytother Res 32(11):2214–2225 KimJW, KimDK, Min A, LeeKH, NamHJ, KimJH, KimJS, Kim TY, Im SA, Park IA, 2016, Amphiregulin confers trastuzumab resistance via AKT and ERK activation in HER2-positive breast cancer. J Cancer Res Clin Oncol 142(1):157–165 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 653 EP 658 DI 10.1007/s12253-018-0525-2 PG 6 ER PT J AU Cornel, MCK Wouters, K Van de Vijver, KK van der Wurff, AMA van Engeland, M Kruitwagen, FPMR Pijnenborg, MAJ AF Cornel, M C Karlijn Wouters, Kim Van de Vijver, K Koen van der Wurff, A M Anneke van Engeland, Manon Kruitwagen, F P M Roy Pijnenborg, M A Johanna TI Gene Promoter Methylation in Endometrial Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Methylation; Endometrial hyperplasia; Endometrial cancer; P16; K-Ras; hMLH1 ID Methylation; Endometrial hyperplasia; Endometrial cancer; P16; K-Ras; hMLH1 AB Up to 60% of untreated atypical hyperplastic endometrium will develop into endometrial carcinoma (EC), and for those who underwent a hysterectomy a coexisting EC is found in up to 50%. Gene promoter methylation might be related to the EC development. The aim of this study is to determine changes in gene promoter profiles in normal endometrium, atypical hyperplasia (AH) and EC in relation to K-Ras mutations. A retrospective study was conducted in patients diagnosed with endometrial hyperplasia with and without subsequent EC. Promoter methylation of APC, hMLh1, O6-MGMT, P14, P16, RASSF1, RUNX3 was analysed on pre-operative biopsies, and correlated to the final histological diagnosis, and related to the presence of K-Ras mutations. In the study cohort (n=98), differences in promoter methylation were observed for hMLH1, O6-MGMT, and P16. Promoter methylation of hMLH1 and O6-MGMT gradually increased from histologically normal endometrium to AH to EC; 27.3, 36.4% and 38.0% for hMLH1 and 8.3%, 18.2% and 31.4% for O6-MGMT, respectively. P16 promoter methylation was significantly different in AH (7.7%) compared to EC (38%). K-Ras mutations were observed in 12.1% of AH, and in 19.6% of EC cases. No association of K-Ras mutation with promoter methylation of any of the tested genes was found. In conclusion, hMLH1 and O6-MGMT promoter methylation are frequently present in AH, and thus considered to be early events in the carcinogenesis of EC, whereas P16 promoter methylation was mainly present in EC, and not in precursor lesions supporting a late event in the carcinogenesis. C1 [Cornel, M C Karlijn] Maastricht University, GROW - School for Oncology and Developmental BiologyMaastricht, The Netherlands. [Wouters, Kim] Maastricht University Medical Center, Department of PathologyMaastricht, The Netherlands. [Van de Vijver, K Koen] Ghent University Hospital, Department of PathologyGhent, Belgium. [van der Wurff, A M Anneke] Department of Pathology, Elisabeth-TweeStedenTilburg, The Netherlands. [van Engeland, Manon] Maastricht University, GROW - School for Oncology and Developmental BiologyMaastricht, The Netherlands. [Kruitwagen, F P M Roy] Maastricht University, GROW - School for Oncology and Developmental BiologyMaastricht, The Netherlands. [Pijnenborg, M A Johanna] Radboud University Nijmegen, Medical Centre, Department of Obstetrics and GynaecologyNijmegen, The Netherlands. RP Cornel, MCK (reprint author), Maastricht University, GROW - School for Oncology and Developmental Biology, Maastricht, The Netherlands. EM karlijncornel@gmail.com CR Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63(1):11–30 Boll D, Karim-Kos HE, Verhoeven RH, Burger CW, Coebergh JW, van de Poll-Franse LV et al, 2013, Increased incidence and improved survival in endometrioid endometrial cancer diagnosed since 1989 in The Netherlands: a population based study. Eur J Obstet Gynecol Reprod Biol 166(2):209–214 Albertini AF, Devouassoux-Shisheboran M, Genestie C, 2012, Pathology of endometrioid carcinoma. Bull Cancer 99(1):7–12 Muraki Y, Banno K, Yanokura M, Kobayashi Y, Kawaguchi M, Nomura H et al, 2009, Epigenetic DNA hypermethylation: clinical applications in endometrial cancer, Review). Oncol Rep 22(5): 967–972 Emons G, Beckmann MW, Schmidt D, Mallmann P, 2015, New WHO Classification of Endometrial Hyperplasias. Geburtshilfe Frauenheilkd 75(2):135–136 Kurman RJ, Kaminski PF, Norris HJ, 1985, The behavior of endometrial hyperplasia.Along-term study of "untreated" hyperplasia in 170 patients. Cancer 56(2):403–412 Di Domenico M, Santoro A, Ricciardi C, IaccarinoM, Iaccarino S, FredaM et al, 2011, Epigenetic fingerprint in endometrial carcinogenesis: the hypothesis of a uterine field cancerization. Cancer Biol Ther 12(5):447–457 Eddib A, Allaf B, Lee J, Yeh J, 2012, Risk for advanced-stage endometrial cancer in surgical specimens from patients with complex endometrial hyperplasia with atypia. Gynecol Obstet Investig 73(1):38–42 Morice P, Leary A, Creutzberg C, Abu-Rustum N, Darai E, 2016, Endometrial cancer. Lancet, London, England, 387(10023):1094– 1108 Widra EA, Dunton CJ,McHugh M, Palazzo JP, 1995, Endometrial hyperplasia and the risk of carcinoma. Int J Gynecol Cancer: official journal of the International Gynecological Cancer Society 5(3): 233–235 Karamursel BS, Guven S, Tulunay G, Kucukali T, Ayhan A, 2005, Which surgical procedure for patients with atypical endometrial hyperplasia? Int J Gynecol Cancer: official journal of the International Gynecological Cancer Society 15(1):127–131 Trimble CL, Kauderer J, Zaino R, Silverberg S, Lim PC, Burke JJ 2nd et al, 2006, Concurrent endometrial carcinoma in women with a biopsy diagnosis of atypical endometrial hyperplasia: a Gynecologic Oncology Group study. Cancer 106(4):812–819 Daud S, Jalil SS, Griffin M, Ewies AA, 2011, Endometrial hyperplasia - the dilemma of management remains: a retrospective observational study of 280 women. Eur J Obstet Gynecol Reprod Biol 159(1):172–175 Giede KC, Yen TW, Chibbar R, Pierson RA, 2008, Significance of Concurrent Endometrial Cancer in Women With a Preoperative Diagnosis of Atypical Endometrial Hyperplasia. Journal of obstetrics and gynaecology Canada : JOGC =. Journal d'obstetrique et gynecologie du Canada : JOGC 30(10):896–901 Banno K, Yanokura M, Susumu N, Kawaguchi M, Hirao N, Hirasawa A et al, 2006, Relationship of the aberrant DNA hypermethylation of cancer-related genes with carcinogenesis of endometrial cancer. Oncol Rep 16(6):1189–1196 Buell-Gutbrod R, Cavallo A, Lee N, Montag A, Gwin K, 2015, Heart and Neural Crest Derivatives Expressed Transcript 2, HAND2): a novel biomarker for the identification of atypical hyperplasia and Type I endometrial carcinoma. Int J Gynecol Pathol: official journal of the International Society of Gynecological Pathologists. 34(1):65–73 Gallos ID, Krishan P, Shehmar M, Ganesan R, Gupta JK, 2013, LNG-IUS versus oral progestogen treatment for endometrial hyperplasia: a long-term comparative cohort study. Hum Reprod, Oxford, England, 28(11):2966–2971 Hubbs JL, Saig RM, Abaid LN, Bae-Jump VL, Gehrig PA, 2013, Systemic and local hormone therapy for endometrial hyperplasia and early adenocarcinoma. Obstet Gynaecol 121(6):1172–1180 Robbe EJ, van Kuijk SM, de Boed EM, Smits LJ, van der Wurff AA, Kruitwagen RF et al, 2012, Predicting the coexistence of an endometrial adenocarcinoma in the presence of atypical complex hyperplasia: immunohistochemical analysis of endometrial samples. Int J Gynecol Cancer: official journal of the International Gynecological Cancer Society 22(7):1264–1272 Sheng Y, Wang H, Liu D, Zhang C, Deng Y, Yang F et al, 2016, Methylation of tumor suppressor gene CDH13 and SHP1 promoters and their epigenetic regulation by the UHRF1/PRMT5 complex in endometrial carcinoma. Gynecol Oncol 140(1):145– 151 Yang YF, Liao YY, Peng NF, Li LQ, Xie SR, Wang RB, 2012, Prediction of coexistent carcinomas risks by subjective EIN diagnosis and comparison with WHO classification in endometrial hyperplasias. Pathol Res Pract 208(12):708–712 Arafa M, Kridelka F, Mathias V, Vanbellinghen JF, Renard I, Foidart JM et al, 2008, High frequency of RASSF1A and RARb2 gene promoter methylation in morphologically normal endometrium adjacent to endometrioid adenocarcinoma. Histopathology 53(5):525–532 Berg A, Hoivik EA, Mjos S, Holst F, Werner HM, Tangen IL et al, 2015, Molecular profiling of endometrial carcinoma precursor, primary and metastatic lesions suggests different targets for treatment in obese compared to non-obese patients. Oncotarget 6(2): 1327–1339 van der Putten LJM, van Hoof R, Tops BBJ, Snijders M, van den Berg-van Erp SH, van der Wurff AAM et al, 2017, Molecular profiles of benign and, pre)malignant endometrial lesions. Carcinogenesis 38(3):329–335 Bjaanaes MM, Fleischer T, Halvorsen AR, Daunay A, Busato F, Solberg S et al, 2016, Genome-wide DNA methylation analyses in lung adenocarcinomas: Association with EGFR, KRAS and TP53 mutation status, gene expression and prognosis. Mol Oncol 10(2): 330–343 Lagarda H, Catasus L, Arguelles R, Matias-Guiu X, Prat J, 2001, K-ras mutations in endometrial carcinomas with microsatellite instability. J Pathol 193(2):193–199 Zhou XC, Dowdy SC, Podratz KC, Jiang SW, 2007, Epigenetic considerations for endometrial cancer prevention, diagnosis and treatment. Gynecol Oncol 107(1):143–153 Ronnett BM, Kurman RJ, 2011, Blaustein's pathology of female genital tract. Springer-Verlag, New York Gallos ID, Alazzam M, Clark TJ, 2016, Management of endometrial hyperplasia. RCOG/BSGE Green-top Guideline No. 67. 67:2– https://www.rcog.org.uk/globalassets/documents/guidelines/ green-top-guidelines/gtg_67_endometrial_hyperplasia.pdf 30. Pecorelli S, 2009, Revised FIGO staging for carcinoma of the vulva, cervix, and endometrium. Int J Gynaecol Obstet: the official organ of the International Federation of Gynaecology and Obstetrics. 105(2):103–104 Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB, 1996, Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S A 93(18):9821–9826 van Engeland M, Weijenberg MP, Roemen GM, Brink M, de Bruine AP, Goldbohm RA et al, 2003, Effects of dietary folate and alcohol intake on promoter methylation in sporadic colorectal cancer: the Netherlands cohort study on diet and cancer. Cancer Res 63(12):3133–3137 Hu ZY, Tang LD, Zhou Q, Xiao L, Cao Y, 2015, Aberrant promoter hypermethylation of p16 gene in endometrial carcinoma. Tumour Biol: the journal of the International Society for Oncodevelopmental Biology and Medicine 36(3):1487–1491 GuidaM, Sanguedolce F, Bufo P, Di Spiezio SA, Bifulco G, Nappi C et al, 2009, Aberrant DNA hypermethylation of hMLH-1 and CDKN2A/p16 genes in benign, premalignant and malignant endometrial lesions. Eur J Gynaecol Oncol 30(3):267–270 Nebot-Bral L, Brandao D, Verlingue L, Rouleau E, Caron O, Despras E et al, 2017, Hypermutated tumours in the era of immunotherapy: The paradigm of personalised medicine. Eur J Cancer, Oxford, England : 1990, 84:290–303 Yamashita H, Nakayama K, Ishikawa M, Nakamura K, Ishibashi T, Sanuki K et al, 2018, Microsatellite instability is a biomarker for immune checkpoint inhibitors in endometrial cancer. Oncotarget 9(5):5652–5664 Jacinto FV, Esteller M, 2007, MGMT hypermethylation: a prognostic foe, a predictive friend. DNA Repair 6(8):1155–1160 Rimel BJ, Huettner P, Powell MA, Mutch DG, Goodfellow PJ, 2009, Absence of MGMT promoter methylation in endometrial cancer. Gynecol Oncol 112(1):224–228 Chao H, Sun J, Lu S, 2000, Methylation and expression of the p16 gene in endometrial carcinoma. Zhonghua Zhong Liu Za Zhi 22: 228–231 Salvesen HB, Das S, Akslen LA, 2000, Loss of nuclear p16 protein expression is not associated with promoter methylation but defines a subgroup of aggressive endometrial carcinomas with poor prognosis. Clin Cancer Res: an official journal of the American Association for Cancer Research 6(1):153–159 Yanokura M, Banno K, Susumu N, Kawaguchi M, Kuwabara Y, Tsukazaki K et al, 2006, Hypermethylation in the p16 promoter region in the carcinogenesis of endometrial cancer in Japanese patients. Anticancer Res 26(2a):851–856 Colombo N, Creutzberg C, Amant F, Bosse T, Gonzalez-Martin A, Ledermann J et al, 2016, ESMO-ESGO-ESTRO Consensus Conference on Endometrial Cancer: Diagnosis, Treatment and Follow-up. Int J Gynecol Cancer: official journal of the International Gynecological Cancer Society 26(1):2–30 Huerta S, 2008, Recent advances in the molecular diagnosis and prognosis of colorectal cancer. Expert Rev Mol Diagn 8(3):277– 288 Elnatan J, Goh HS, Smith DR, 1996, C-KI-RAS activation and the biological behaviour of proximal and distal colonic adenocarcinomas. Eur. J. Cancer, Oxford, England : 1990, 32a(3):491–497 Thoury A, Descatoire V, Kotelevets L, Kannengiesser C, Bertrand G, Theou-Anton N et al, 2014, Evidence for different expression profiles for c-Met, EGFR, PTEN and the mTOR pathway in low and high grade endometrial carcinomas in a cohort of consecutive women. Occurrence of PIK3CA and KRas mutations and microsatellite instability. Histol Histopathol 29(11):1455–1466 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 659 EP 667 DI 10.1007/s12253-018-0489-2 PG 9 ER PT J AU Lorincz, T Holczer, M Kapuy, O Szarka, A AF Lorincz, Tamas Holczer, Marianna Kapuy, Orsolya Szarka, Andras TI The Interrelationship of Pharmacologic Ascorbate Induced Cell Death and Ferroptosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pharmacologic ascorbate; Cell death; ROS; Lipid peroxidation; Ferroptosis ID Pharmacologic ascorbate; Cell death; ROS; Lipid peroxidation; Ferroptosis AB Pharmacologic ascorbate induced cell death and ferroptosis share common features such as iron dependency, production of ROS, lipid peroxidation, caspase independency and the possible involvement of autophagy. These observations lead us to hypothesize that ferroptosis may also be involved in cancer cell death due to pharmacologic ascorbate treatment. Thus cell death of HT-1080 cell line was induced by ferroptosis inducers and pharmacologic ascorbate then the mechanism of cell death was compared. The EC50 value of pharmacologic ascorbate on HT-1080 cell line was found to be 0.5 mM that is in the range of the most ascorbate sensitive cell lines. However either of the specific inhibitors of ferroptosis (ferrostatin-1 and liproxstatin-1) could not elevate the viability of pharmacologic ascorbate treated cells suggesting that ferroptosis was not involved in the pharmacologic ascorbate induced cell death. α-tocopherol that could effectively elevate the viability of erastin and RSL3 treated HT1080 cells failed to mitigate the cytotoxic effect of pharmacologic ascorbate further strengthened this assumption. Furthermore at lower concentrations (0.1–0.5 mM) ascorbate could avoid the effects of ferroptosis inducers. Our results indicate that pharmacologic ascorbate induced cytotoxicity and ferroptosis – albeit phenotypically they show similar traits – are governed by different mechanisms. C1 [Lorincz, Tamas] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, Szent Gellert ter 4, 1111 Budapest, Hungary. [Holczer, Marianna] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, 1444 Budapest, Hungary. [Kapuy, Orsolya] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, 1444 Budapest, Hungary. [Szarka, Andras] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, Szent Gellert ter 4, 1111 Budapest, Hungary. RP Szarka, A (reprint author), Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular Biology, 1111 Budapest, Hungary. EM szarka@mail.bme.hu CR Toth SZ, Lorincz T, Szarka A, 2017, Concentration does matter: The beneficial and potentially harmful effects of ascorbate in humans and plants. Antioxid Redox Signal 00:ars.2017.7125 Liou G-Y, Storz P, 2010, Reactive oxygen species in cancer. Free Radic Res 44:479–496 Rouleau L, Antony AN, Bisetto S, Newberg A, Doria C, Levine M, Monti DA, Hoek JB, 2016, Synergistic effects of ascorbate and sorafenib in hepatocellular carcinoma: new insights into ascorbate cytotoxicity. Free Radic Biol Med 95:308–322 Kehrer JP, 2000, The Haber-Weiss reaction and mechanisms of toxicity. Toxicology 149:43–50 Adachi T, Nonomura S, Horiba M, Hirayama T, Kamiya T, Nagasawa H, Hara H, 2016, Iron stimulates plasma-activated medium- induced A549 cell injury. Sci Rep 6:20928 Mandl J, Szarka A, Banhegyi G, 2009, Vitamin C: update on physiology and pharmacology. Br J Pharmacol 157:1097–1110 Chen Q, Espey MG, KrishnaMC,Mitchell JB, Corpe CP, Buettner GR, Shacter E, LevineM(2005, Pharmacologic ascorbic acid concentrations selectively kill cancer cells: action as a pro-drug to deliver hydrogen peroxide to tissues. Proc Natl Acad Sci U S A 102: 13604–13609 Chen Q, Espey MG, Sun AY, Pooput C, Kirk KL, Krishna MC, Khosh DB, Drisko J, Levine M, 2008, Pharmacologic doses of ascorbate act as a prooxidant and decrease growth of aggressive tumor xenografts in mice. Proc Natl Acad Sci 105:11105–11109 Parrow NL, Leshin JA, Levine M, 2013, Parenteral ascorbate as a cancer therapeutic: a reassessment based on pharmacokinetics. Antioxid Redox Signal 19:2141–2156 Schoenfeld JD, Sibenaller ZA, Mapuskar KA et al, 2017, O2 and H2O2 mediated disruption of Fe metabolism causes the differential susceptibility of NSCLC and GBMCancer cells to pharmacological ascorbate. Cancer Cell 31:1–14 Hong S-W, Jin D-H, Hahm E-S, Yim SH, Lim JS, Kim KI, Yang Y, Lee SS, Kang JS, Lee WJ, Lee WK, Lee MS, 2007, Ascorbate, vitamin C, induces cell death through the apoptosis-inducing factor in human breast cancer cells. Oncol Rep 18:811–815 Kang JS, Cho D,KimY-I,HahmE, YangY, KimD, Hur D, Park H, Bang S, Hwang YI, Lee WJ, 2003, l-Ascorbic acid, vitamin C, induces the apoptosis of B16 murine melanoma cells via a caspase-8?Independent pathway. Cancer Immunol Immunother 52: 693–698 Lin S-Y, Lai W-W, Chou C-C, Kuo HM, Li TM, Chung JG, Yang JH, 2006, Sodium ascorbate inhibits growth via the induction of cell cycle arrest and apoptosis in human malignant melanoma A375.S2 cells. Melanoma Res 16:509–519 Chen P, Yu J, Chalmers B, Drisko J, Yang J, Li B, Chen Q, 2012, Pharmacological ascorbate induces cytotoxicity in prostate cancer cells through ATP depletion and induction of autophagy. Anti- Cancer Drugs 23:437–444 Cullen JJ, 2010, Ascorbate induces autophagy in pancreatic cancer. Autophagy 6:421–422 Fukui M, Yamabe N, Choi H-J, Polireddy K, Chen Q, Zhu BT, 2015, Mechanism of ascorbate-induced cell death in human pancreatic cancer cells: role of Bcl-2, Beclin 1 and autophagy. Planta Med 81:838–846 Dolma S, Lessnick SL, Hahn WC, Stockwell BR, 2003, Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell 3:285–296 Yang WS, Stockwell BR, 2008, Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol 15: 234–245 Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, Morrison B III, Stockwell BR, 2012, Ferroptosis: an irondependent form of nonapoptotic cell death. Cell 149:1060–1072 Gao M, Monian P, Pan Q, Zhang W, Xiang J, Jiang X, 2016, Ferroptosis is an autophagic cell death process. Cell Res 26: 1021–1032 Sun X, Ou Z, Xie M et al, 2015, HSPB1 as a novel regulator of ferroptotic cancer cell death. Oncogene:1–9 YangWS, Sriramaratnam R,Welsch ME et al, 2014, Regulation of ferroptotic cancer cell death by GPX4. Cell 156:317–331 Louandre C, Ezzoukhry Z, Godin C, Barbare JC, Maziere JC, Chauffert B, Galmiche A, 2013, Iron-dependent cell death of hepatocellular carcinoma cells exposed to sorafenib. Int J Cancer 133: 1732–1742 Lachaier E, Louandre C, Godin C, Saidak Z, Baert M, Diouf M, Chauffert B, Galmiche A, 2014, Sorafenib induces ferroptosis in human cancer cell lines originating from different solid tumors. Anticancer Res 34:6417–6422 Lorincz T, Jemnitz K, Kardon T, Mandl J, Szarka A, 2015, Ferroptosis is involved in acetaminophen induced cell death. Pathol Oncol Res 21:1115–1121 Lorincz T, Szarka A, 2017, The determination of hepatic glutathione at tissue and subcellular level. J Pharmacol Toxicol Methods 88:32–39 Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, AichlerM,Walch A, Eggenhofer E, Basavarajappa D, Radmark O, Kobayashi S, Seibt T, Beck H, Neff F, Esposito I,Wanke R, Forster H,YefremovaO, Heinrichmeyer M, Bornkamm GW, Geissler EK, Thomas SB, Stockwell BR, O’Donnell VB, Kagan VE, Schick JA, Conrad M, 2014, Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol 16:1180–1191 Ma S, Henson ES, Chen Y, Gibson SB, 2016, Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis 7:e2307 Finley JW, Wheeler EL, Witt SC, 1981, Oxidation of glutathione by hydrogen peroxide and other oxidizing agents. J Agric Food Chem 29:404–407 Lopalco A, Dalwadi G, Niu S, Schowen RL, Douglas J, Stella VJ, 2016, Mechanism of decarboxylation of pyruvic acid in the presence of hydrogen peroxide. J Pharm Sci 105:705–713 Merkofer M, Kissner R, Hider RC, Brunk UT, Koppenol WH, 2006, Fenton chemistry and iron chelation under physiologically relevant conditions: electrochemistry and kinetics. Chem Res Toxicol 19:1263–1269 Degterev A, Hitomi J, Germscheid M, Ch'en IL, Korkina O, Teng X, Abbott D, Cuny GD, Yuan C, Wagner G, Hedrick SM, Gerber SA, Lugovskoy A, Yuan J, 2008, Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol 4:313–321 Bocsi J, Nagy K, Tyihak E, Trezl L, Szende B, 1998, Reduction of apoptosis of in vitro cultured lymphocytes of HIV-positive persons by N(G)-hydroxy-methylated-L-arginine and 1’-methylascorbigen. Acta Biol Hung 49:331–337 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 669 EP 679 DI 10.1007/s12253-018-0539-9 PG 11 ER PT J AU Pang, Y Liu, Z Liu, Sh AF Pang, Yanan Liu, Zhiyong Liu, Shanrong TI Identification of Key Potential Targets and Pathway for Arsenic Trioxide by Systemic Bioinformatics Analysis in Pancreatic Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Arsenic trioxide; Genes/proteins interaction; Bioinformatics analysis; Pancreatic cancer ID Arsenic trioxide; Genes/proteins interaction; Bioinformatics analysis; Pancreatic cancer AB Arsenic trioxide is an approved chemotheraputic agent for the treatment of acute promyelocytic leukemia (APL). Recently, numerous studies suggested that arsenic trioxide acts as anti-cancer roles in various human malignancies. However, the molecular mechanisms are not fully elucidated. In this study, we explored the critical targets of arsenic trioxide and their interaction network systematically by searching the publicly available published database like DrugBank (DB) and STRING. Seven direct protein targets (DPTs) and 111 DPT-associated genes were identified. The enrichment analysis of arsenic trioxide associated genes/ proteins revealed 10 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Among these pathways, phosphatidylinositol- 4,5-bisphosphate-3-kinase -Akt (PI3K-Akt) single pathway and pancreatic cancer pathway are highly correlated with arsenic trioxide and have 5 overlapped targets. Then we investigated the gene alternation of selected critical genes in pancreatic cancer studies using cBio portal. These results indicated that arsenic trioxide could act anti-tumor function through PI3K-Akt single pathway and identified critical genes might be therapeutic targets for pancreatic cancer. C1 [Pang, Yanan] Second Military Medical University, Changhai Hospital, Department of Laboratory Diagnostics, 200433 Shanghai, China. [Liu, Zhiyong] Second Military Medical University, Changhai Hospital, Department of Laboratory Diagnostics, 200433 Shanghai, China. [Liu, Shanrong] Second Military Medical University, Changhai Hospital, Department of Laboratory Diagnostics, 200433 Shanghai, China. RP Liu, Sh (reprint author), Second Military Medical University, Changhai Hospital, Department of Laboratory Diagnostics, 200433 Shanghai, China. EM liushanrong@hotmail.com CR Kulik-Kupka K, Koszowska A, Bronczyk-Puzon A et al, 2016, Arsenic - poison or medicine? Med Pr 67(1):89–96. https://doi. org/10.13075/mp.5893.00322 Wang ZG, Rivi R, Delva L, Konig A, Scheinberg DA, Gambacorti- Passerini C, Gabrilove JL, Warrell RP Jr, Pandolfi PP, 1998, Arsenic trioxide and melarsoprol induce programmed cell death in myeloid leukemia cell lines and function in a PML and PMLRARalpha independent manner. Blood 92(5):1497–1504 Sun RC, Board PG, Blackburn AC, 2011, Targeting metabolism with arsenic trioxide and dichloroacetate in breast cancer cells. Mol Cancer 10:142. , DOI 10.1186/1476-4598-10-142 Ma ZB, Xu HY, Jiang M, Yang YL, Liu LX, Li YH, 2014, Arsenic trioxide induces apoptosis of human gastrointestinal cancer cells. World J Gastroenterol 20(18):5505–5510. , DOI 10.3748/wjg.v20.i18.5505 Yang MH, Zang YS, Huang H, Chen K, Li B, Sun GY, Zhao XW, 2014, Arsenic trioxide exerts anti-lung cancer activity by inhibiting angiogenesis. Curr Cancer Drug Targets 14(6):557–566 Zheng L, Jiang H, Zhang ZWet al, 2016, Arsenic trioxide inhibits viability and induces apoptosis through reactivating the Wnt inhibitor secreted frizzled related protein-1 in prostate cancer cells. Onco Targets Ther 9:885–894. , DOI 10.2147/ott.s92129 Gao JK, Wang LX, Long B, Ye XT, Su JN, Yin XY, Zhou XX, Wang ZW(2015, Arsenic trioxide inhibits cell growth and invasion via Down- regulation of Skp2 in pancreatic Cancer cells. Asian Pac J Cancer Prev 16(9):3805–3810 Pihlak R, Valle JW, McNamara MG, 2017, Germline mutations in pancreatic cancer and potential new therapeutic options. Oncotarget. , DOI 10.18632/oncotarget.17291 Li X, Ding X, Adrian TE, 2003, Arsenic trioxide induces apoptosis in pancreatic cancer cells via changes in cell cycle, caspase activation, and GADD expression. Pancreas 27(2):174–179 Barabasi AL, Gulbahce N, Loscalzo J, 2011, Network medicine: a network-based approach to human disease. Nat Rev Genet 12(1): 56–68. , DOI 10.1038/nrg2918 Vidal M, Cusick ME, Barabasi AL, 2011, Interactome networks and human disease. Cell 144(6):986–998. , DOI 10.1016/ j.cell.2011.02.016 Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, Kuhn M, Bork P, Jensen LJ, von Mering C, 2015, STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43(Database issue):D447–D452. , DOI 10.1093/nar/gku1003 Ruan HH, Zhang Z,Wang SY, Nickels LM, Tian L, Qiao JJ, Zhu J, 2017, Tumor necrosis factor receptor-associated factor 6, TRAF6, mediates ubiquitination-dependent STAT3 activation upon Salmonella typhimurium infection. Infect Immun 85. https://doi. org/10.1128/iai.00081-17 Manne RK, Agrawal Y, Bargale A, Patel A, Paul D, Gupta NA, Rapole S, Seshadri V, Subramanyam D, Shetty P, SantraMK, 2017, A MicroRNA/ubiquitin ligase feedback loop regulates slugmediated invasion in breast Cancer. Neoplasia 19(6):483–495. , DOI 10.1016/j.neo.2017.02.013 Guo YY, Yi L, Zheng Yet al, 2017, Ubiquitin C-terminal hydrolase L1, UCH-L1, promotes hippocampus-dependent memory via its deubiquitinating effect on TrkB. J Neurosci 37:5978–5995. , DOI 10.1523/jneurosci.3148-16.2017 Das A, Qian J, Tsang WY, 2017, USP9X counteracts differential ubiquitination of NPHP5 by MARCH7 and BBS11 to regulate ciliogenesis. PLoS Genet 13(5):e1006791. , DOI 10.1371/ journal.pgen.1006791 Wang Y, Kuramitsu Y, Baron B, Kitagawa T, Tokuda K, Akada J, Maehara SI, Maehara Y, Nakamura K, 2017, PI3K inhibitor LY294002, as opposed to wortmannin, enhances AKT phosphorylation in gemcitabine-resistant pancreatic cancer cells. Int J Oncol 50(2):606–612. , DOI 10.3892/ijo.2016.3804 Ebrahimi S, Hosseini M, Shahidsales S, Maftouh M, Ferns GA, Ghayour-Mobarhan M, Hassanian SM, Avan A, 2017, Targeting the Akt/PI3K signaling pathway as a potential therapeutic strategy for the treatment of pancreatic Cancer. Curr Med Chem 24:1321– 1331. , DOI 10.2174/0929867324666170206142658 Jiao Y, Yonescu R, Offerhaus GJ et al, 2014, Whole-exome sequencing of pancreatic neoplasms with acinar differentiation. J Pathol 232(4):428–435. , DOI 10.1002/path.4310 Biankin AV,Waddell N, Kassahn KS et al, 2012, Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491(7424):399–405. , DOI 10.1038/nature11547 Cao Y, Gao Z, Li L, Jiang X, Shan A, Cai J, Peng Y, Li Y, Jiang X, Huang X,Wang J,Wei Q, Qin G, Zhao J, Jin X, Liu L, Li Y,Wang W,Wang J, Ning G, 2013)Whole exome sequencing of insulinoma reveals recurrent T372R mutations in YY1. Nat Commun 4:2810. , DOI 10.1038/ncomms3810 Jiao Y, Shi C, Edil BH, deWilde RF, Klimstra DS, Maitra A, Schulick RD, Tang LH, Wolfgang CL, Choti MA, Velculescu VE, Diaz LA, Vogelstein B, Kinzler KW, Hruban RH, Papadopoulos N, 2011, DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331(6021):1199– 1203. , DOI 10.1126/science.1200609 Witkiewicz AK, McMillan EA, Balaji U et al, 2015, Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun 6:6744. , DOI 10.1038/ncomms7744 Bailey P, Chang DK, Nones K et al, 2016, Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 531(7592):47– 52. , DOI 10.1038/nature16965 Scarpa A, Chang DK, Nones K et al, 2017, Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 543(7643):65– 71. , DOI 10.1038/nature21063 Yurgelun MB, Masciari S, Joshi VA, Mercado RC, Lindor NM, Gallinger S, Hopper JL, Jenkins MA, Buchanan DD, Newcomb PA, Potter JD, Haile RW, Kucherlapati R, Syngal S, Colon Cancer Family Registry, 2015, Germline TP53 mutations in patients with early-onset colorectal Cancer in the Colon Cancer family registry. JAMA Oncol 1(2):214– 221. , DOI 10.1001/jamaoncol.2015.0197 Leroy B, Anderson M, Soussi T, 2014, TP53 mutations in human cancer: database reassessment and prospects for the next decade. Hum Mutat 35(6):672–688. , DOI 10.1002/humu.22552 Hu L, Zhu YT, Qi C, Zhu YJ, 2009, Identification of Smyd4 as a potential tumor suppressor gene involved in breast cancer development. Cancer Res 69(9):4067–4072. , DOI 10.1158/0008- 5472.can-08-4097 Manokawinchoke J, Limjeerajarus N, Limjeerajarus C, Sastravaha P, Everts V, Pavasant P, 2015, Mechanical force-induced TGFB1 increases expression of SOST/POSTN by hPDL cells. J Dent Res 94(7):983–989. , DOI 10.1177/0022034515581372 Hull ML, Johan MZ, Hodge WL, Robertson SA, Ingman WV, 2012, Host-derived TGFB1 deficiency suppresses lesion development in a mouse model of endometriosis. Am J Pathol 180(3):880– 887. , DOI 10.1016/j.ajpath.2011.11.013 Shen SJ, Zhang YH, Gu XX, Jiang SJ, Xu LJ, 2017, Yangfei Kongliu formula, a compound Chinese herbal medicine, combined with cisplatin, inhibits growth of lung cancer cells through transforming growth factor-beta1 signaling pathway. J Integr Med 15(3):242–251. , DOI 10.1016/s2095-4964(17)60330-3 Anvarian Z, Nojima H, van Kappel EC, Madl T, Spit M, Viertler M, Jordens I, Low TY, van Scherpenzeel RC, Kuper I, Richter K, Heck AJR, Boelens R, Vincent JP, Rudiger SGD, Maurice MM, 2016, Axin cancer mutants form nanoaggregates to rewire the Wnt signaling network. Nat Struct Mol Biol 23(4):324–332. , DOI 10.1038/nsmb.3191 Wu R, Zhai Y, Fearon ER, Cho KR, 2001, Diverse mechanisms of beta-catenin deregulation in ovarian endometrioid adenocarcinomas. Cancer Res 61(22):8247–8255 Chen GQ, Zhu J, Shi XG, Ni JH, Zhong HJ, SiGY, JinXL, TangW, Li XS, XongSM,ShenZX, SunGL,Ma J, ZhangP,ZhangTD,GazinC, Naoe T, Chen SJ,Wang ZY, Chen Z, 1996, In vitro studies on cellular andmolecularmechanisms of arsenic trioxide, As2O3, in the treatment of acute promyelocytic leukemia: As2O3 induces NB4 cell apoptosis with downregulation of Bcl-2 expression and modulation of PMLRAR alpha/PML proteins. Blood 88(3):1052–1061 Noguera NI, Pelosi E, Angelini DF et al, 2017, High-dose ascorbate and arsenic trioxide selectively kill acute myeloid leukemia and acute promyelocytic leukemia blasts in vitro. Oncotarget 8(20):32550–32565. , DOI 10.18632/oncotarget.15925 He H, An R, Hou J, Fu W, 2017, Arsenic trioxide induced rhabdomyolysis, a rare but severe side effect, in an APL patient: a case report. Front Med 11(2):284–286. https://doi. org/10.1007/s11684-017-0514-y Zhang HN, Yang L, Ling JY, Czajkowsky DM, Wang JF, Zhang XW, Zhou YM, Ge F, Yang MK, Xiong Q, Guo SJ, le HY, Wu SF, Yan W, Liu B, Zhu H, Chen Z, Tao SC, 2015, Systematic identification of arsenic-binding proteins reveals that hexokinase-2 is inhibited by arsenic. Proc Natl Acad Sci U S A 112(49):15084– 15089. , DOI 10.1073/pnas.1521316112 Cekanova M, Fernando RI, Siriwardhana N, SukhthankarM, Parra C,Woraratphoka J,Malone C, Strom A, Baek SJ,Wade PA, Saxton AM, Donnell RM, Pestell RG, Dharmawardhane S, Wimalasena J, 2015, BCL-2 family protein, BAD is down-regulated in breast cancer and inhibits cell invasion. Exp Cell Res 331(1):1–10. , DOI 10.1016/j.yexcr.2014.11.016 Sastry KS, Al-Muftah MA, Li P et al, 2014, Targeting proapoptotic protein BAD inhibits survival and self-renewal of cancer stem cells. Cell Death Differ 21(12):1936–1949. , DOI 10.1038/cdd.2014.140 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 681 EP 690 DI 10.1007/s12253-018-0543-0 PG 10 ER PT J AU Li, N Tian, YW Xu, Y Meng, DD Gao, L Shen, Wj Liu, Zl Xu, ZQ AF Li, Ning Tian, Yong-Wei Xu, Yue Meng, Dan-Dan Gao, Ling Shen, Wen-jie Liu, Zong-lan Xu, Zhi-Qiao TI Combined Treatment with Autologous CIK Cells, Radiotherapy and Chemotherapy in Advanced Cervical Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Autologous CIK cell; Advanced cervical cancer; Radiochemotherapy ID Autologous CIK cell; Advanced cervical cancer; Radiochemotherapy AB To investigate the clinical efficacy of autologous cytokine induced killer (CIK) cells transfusion combined with radiochemotherapy in the treatment of advanced cervical cancer. A total of 89 hospitalized patients with advanced cervical cancer were admitted and divided into the treatment group (44 cases, autologous CIK cells transfusion combined with radiochemotherapy) and the control group (45 cases, radiochemotherapy) by a randomized non-blind method. Comparisons of therapeutic efficacies, immune functions, life qualities and survival rates were analyzed between the two groups. The short-term therapeutic efficacy of the treatment group was significantly higher than that of the control group. There was no significant difference in 1, 2 and 3 year survival rates between the two groups. Compared with pre-treatment, levels of CD3+, CD4+/CD8+ in peripheral blood were increased in the CIK group,whichwere reduced in the control group. In the CIK group,only the feeling was depressed on the 25th day post-treatment (T25) compared with the day before treatment (B1). However in the control group, the function of body, role, social and holistic health was obvious disordered on day T25 compared with day B1. On day T25, there were significant differences in function of body, social and holistic health between two groups. Autologous CIK cells transfusion combined with radiochemotherapy shows better short-term efficacy than radiochemotherapy alone in the treatment of advanced cervical cancer, which obviously improves immune function and life quality of patients with low side effects. C1 [Li, Ning] Kaifeng Central Hospital, The Center of Tumor Diagnosis and Treatment, No.85 Hedao Street, Longting District, 475001 Kaifeng, Henan, China. [Tian, Yong-Wei] Kaifeng Central Hospital, The Center of Tumor Diagnosis and Treatment, No.85 Hedao Street, Longting District, 475001 Kaifeng, Henan, China. [Xu, Yue] Kaifeng Central Hospital, The Center of Tumor Diagnosis and Treatment, No.85 Hedao Street, Longting District, 475001 Kaifeng, Henan, China. [Meng, Dan-Dan] Kaifeng Central Hospital, The Center of Tumor Diagnosis and Treatment, No.85 Hedao Street, Longting District, 475001 Kaifeng, Henan, China. [Gao, Ling] Kaifeng Central Hospital, The Center of Tumor Diagnosis and Treatment, No.85 Hedao Street, Longting District, 475001 Kaifeng, Henan, China. [Shen, Wen-jie] Kaifeng Central Hospital, The Department of Statistics, No.85 Hedao Street, Longting District, 475001 Kaifeng, Henan, China. [Liu, Zong-lan] Kaifeng Central Hospital, The Department of Statistics, No.85 Hedao Street, Longting District, 475001 Kaifeng, Henan, China. [Xu, Zhi-Qiao] Kaifeng Central Hospital, The Center of Tumor Diagnosis and Treatment, No.85 Hedao Street, Longting District, 475001 Kaifeng, Henan, China. RP Xu, ZQ (reprint author), Kaifeng Central Hospital, The Center of Tumor Diagnosis and Treatment, 475001 Kaifeng, China. EM liningkfyy@163.com CR Hu Y, Sun X, Mao C, et al, 2017, Upregulation of long noncoding RNATUG1 promotes cervical cancer cell proliferation and migration. Cancer Med 6(2):471–482 Artan, I.M., et al., The knowledge, attitude and behavioral-intent regarding cervical cancer and the human papillomavirus, HPV, vaccine: A cross-sectional study among female university students in Ajman, UAE Ferlay J et al, 2010, Cancer incidence and mortality worldwide: IARC CancerBase no.11. IARC 136(5):E359–E386 Vidal JP et al, 2015, Genetic diversity of HPV16 and HPV18 in Brazilian patients with invasive cervical cancer. J Med Virol 88(7): 1279–1287 Haque W, Verma V, Fakhreddine M, Hatch S, Butler EB, Teh BS, 2017, Addition of chemotherapy to definitive radiotherapy for IB1 and IIA1 cervical cancer: analysis of the National Cancer Data Base. Gynecol Oncol 144(1):28–33 Zheng Y, Hu B, Xie S, Chen X, Hu Y, Chen W, Li S, Hu B, 2017, Dendritic cells infected by ad-sh-SOCS1 enhance cytokine-induced killer, CIK, cell immunotherapeutic efficacy in cervical cancer models. Cytotherapy 19(5):617–628 Niam M, Linn YC, Fook Chong S, Lim TJ, Chu S, Choong A, Yong HX, Suck G, Chan M, Koh M, 2011, Clinical scale expansion of cytokine-induced killer cells is feasible from healthy donors and patients with acute and chronic myeloid leukemia at various stages of therapy. Exp Hematol 39(9):897–903.e1 Arber C, Abhyankar H, Heslop HE, Brenner MK, Liu H, Dotti G, Savoldo B, 2013, The immunogenicity of virus-derived 2A sequences in immunocompetent individuals. Gene Ther 20(9):958–962 Liu AM et al, 2011, Antitumor effects of cytokine- induced killer cells on cervical cancer HeLa cells in vitro and in vivo. Carcino, Terato & Muta 23(5):353–161 Kim HM, Lim J, Kang JS, Park SK, Lee K, Kim JY, Kim YJ, Hong JT, Kim Y, Han SB, 2009, Inhibition of human cervical carcinoma growth by cytokine-induced killer cells in nude mouse xenograft model. Int Immunopharmacol 9(3):375–380 Green JA, Kirwan JM, Tierney JF, Symonds P, Fresco L, Collingwood M,Williams CJ, 2001, Survival and recurrence after concomitant chemotherapy and radiotherapy for cancer of the uterine cervix: a systematic review and meta-analysis. Lancet 358(9284):781–786 Zagon IS,Mclaughlin PJ, 2012, Targeting opioidergic pathways as a novel biological treatment for advanced pancreatic cancer. Expert Rev Gastroent 6(2):133–135 Mesiano G, Todorovic M, Gammaitoni L, Leuci V, Giraudo Diego L, Carnevale-Schianca F, Fagioli F, Piacibello W, Aglietta M, Sangiolo D, 2012, Cytokine-induced killer, CIK, cells as feasible and effective adoptive immunotherapy for the treatment of solid tumors. Expert Opin Biol Ther 12(6):673–684 Jonathan Benjamin M, 2013, Early infusion of donorderived CIK cells as consolidative immunotherapy following non-Myeloablative allogeneic transplantation: safety and feasibility. Blood 122 Linn YC, Yong HX, NiamM, Lim TJ, Chu S, Choong A, Chuah C, Goh YT,HwangW, Loh Y, NgHJ, Suck G, ChanM, KohM(2012, A phase I/II clinical trial of autologous cytokine-induced killer cells as adjuvant immunotherapy for acute and chronic myeloid leukemia in clinical remission. Cytotherapy 14(7):851–859 Geng J, Zhang Q, Tong J, 2016, Effect of chemoradiation combined with DC-CIK cells biological therapy on the treatment of patients with middle-advanced non-small cell lung cancer. J Clin Med in Prac 20(9):48–50 Shi L et al, 2012, Adjuvant immunotherapy with CIK cells for stage III gastric Cancer. J Basic Clin Onco 25(3):62–65 Luo X et al, 2016, Curative effect of autologous DC-CIK cells on metastatic renal cell carcinoma. J Mod Oncol 15:2426–2429 Chen B et al, 2015, Effectiveness of immune therapy combined with chemotherapy on the immune function and recurrence rate of cervical cancer. Exp Ther Med 9(3):1063–1067 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 691 EP 696 DI 10.1007/s12253-018-0541-2 PG 6 ER PT J AU Li, D Jiang, Y Yang, W AF Li, Ding Jiang, Yunhui Yang, Wan TI Approach the Invasive Potential with Hurthle Cell Tumors of Thyroid SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hurthle cell tumors; Immunohistochemistry; P53; CyclinD1; Ki-67 ID Hurthle cell tumors; Immunohistochemistry; P53; CyclinD1; Ki-67 AB To observe the expression of P53, CyclinD1, Ki-67, Galectin-3, COX-2, Bcl-2 and approach their contribution on assessing the invasive potential for Hurthle cell tumors. Seventy-three cases of Hurthle cell tumor were collected for immunohistochemistry staining. The patients were followed up with 8 months to 5 years. Tumors were divided into four grades according to invasion and diameter:(1) extremely low risk (27 cases that less than 2 cm and without invasion), (2) low risk (18 cases that within 2–3.9 cm and without invasion), (3) moderate risk (21 cases that achieve 4 cm and without invasion), (4) high risk (7 cases that with invasion of capsule/vessel in spite of the diameter). Immunostaining presented that all 73 cases were positive with Galectin-3, COX-2 and Bcl-2. For each group, P53 positive were 29.6%, 55.6%, 90.5%, 100.0%; CyclinD1 stained with 7.4%,22.2%,52.4%,100.0% and Ki-67 were 0.0%,5.6%,9.5%,28.6%, respectively. The higher risk of tumor, the more cases that positive expressed P53 and CyclinD1. After following up within 49 patients, two of the recurring cases were positive with P53 and CyclinD1 and one of which was also highly expressed Ki-67. Detecting P53, CyclinD1 and Ki-67 might provide reference for invasive potential assessment with Hurthle cell tumors but not Galectin-3, COX-2 and Bcl-2. C1 [Li, Ding] the First People’s Hospital of Jingmen, Department of Pathology, 448000 Jingmen, Hubei Province, China. [Jiang, Yunhui] the Second People’s Hospital of Jingmen, Department of Pathology, 448000 Jingmen, Hubei Province, China. [Yang, Wan] the First People’s Hospital of Jingmen, Department of Pathology, 448000 Jingmen, Hubei Province, China. RP Li, D (reprint author), the First People’s Hospital of Jingmen, Department of Pathology, 448000 Jingmen, China. EM dl789521@126.com CR Hoos A, Stojadinovic A, Singh B, Dudas ME, Leung DHY, Shaha AR, Shah JP, Brennan MF, Cordon-Cardo C, Ghossein R, 2002, Clinical significance of molecular expressing profiles of Hurthle cell tumors of the thyroid gland analyzed via tissue microarrays [J]. Am J Pathol 160(1):175–183 Vogrin A, Besic H, Besic N, Music MM, 2016, Recurrence rate in regional lymph nodes in 737 patients with follicular or Hurthle cell neoplasms [J]. Radiol Oncol 50(3):269–273 Ahmadi S, Stang M, Jiang XS, Sosa JA, 2016, Hurthle cell carcinoma: current perspectives [J]. Onco Targets and Therapy 9:6873–6884 Ernaga Lorea A,Migueliz Bermejo I, Anda Apinaniz E et al, 2018, Comparison of clinical characteristics of patients with follicular thyroid carcinoma and Hurthle cell carcinoma [J]. Endocrinol Diabetes Nutr 65(3):136–142 Sippel RS, Elaraj DM, Khanafshar E, Zarnegar R, Kebebew E, Duh QY, Clark OH, 2008, Tumor size predicts maligant potential in Hurthle cell neoplasms of the thyroid [J]. World J Surg 32(5): 702–707 Sugino K, Ito K,Mimura T, Kameyama K, Iwasaki H, Ito K, 2001, Hurthle cell tumor of the thyroid: analysis of 188 cases [J].World J Surg 25(9):1160–1163 Ding L, Zou X, Yang Wet al, 2016, Analyze two cases of encapsulated papillary oncocytic neoplasms, EPONs, of the thyroid [J]. Int J Clin Exp Pathol 9(7):7493–7497 Ristevska N, Stojanoski S, Gjorceva DP, 2015, Appearance of Hurthle cell carcinoma soon after surgical extirpation of Hurthle cell adenoma and follicular adenoma of the thyroid gland [J]. Radiol Oncol 49(1):26–31 Chao TC, Lin JD, Chen MF, 2005, Surgical treatment of Hurthle cell tumors of the thyroid [J]. World J Surg 29(2):164–168 Besic N, Hocevar M, Zgajnar L et al, 2006, Aggressiveness of therapy and prognosis of patients with Hurthle cell papillary thyroid carcinoma [J]. Thyroid 16(1):67–72 Anila KR, Nayak N, George PS, Jayasree K, 2018, Cytomorphologic spectrum of Hurthle cell lesions of thyroid: a study of 54 cases [J]. Gulf J Oncolog 1(26):6–10 Krhin B, Goricar K, Gazic B, Dolzan V, Besic N, 2016, Functional polymorphisms in antioxidant genes in Hurthle cell thyroid neoplasm - an association of GPX1 polymorphism and recurrent Hurthle cell thyroid carcinoma [J]. Radiol Oncol 50(3):289–296 Zhang YW, Greenblatt DY, Repplinger D, Bargren A, Adler JT, Sippel RS, Chen H, 2008, Older age and larger tumor size predict malignancy in hurthle cell neoplasms of the thyroid [J]. Ann Surg Oncol 15(10):2842–2846 Ferreira-da-Silva A, Valacca C, Rios E, Populo H, Soares P, Sobrinho-Simoes M, Scorrano L, Maximo V, Campello S, 2015, Mitochondrial dynamics protein Drp1 is overexpressed in oncocytic thyroid tumors and regulates cancer cell migration [J]. PLoS One 10(3):e0122308 Petric R, Gazic B, Goricar K et al, 2016, Expression of miRNA and occurrence of distant metastases in patients with Hurthle cell carcinoma [J]. Int J Endocrinol 8945247:2016 Monaco M, Chiappetta G, Aiello C et al, 2014, CBX7 expression in Oncocytic thyroid neoplastic lesions, Hurthle cell adenomas and carcinomas, [J]. Eur Thyroid J 3(4):211–216 Gopal RK, Kubler K, Calvo SE, Polak P, Livitz D, Rosebrock D, Sadow PM, Campbell B, Donovan SE, Amin S, Gigliotti BJ, Grabarek Z, Hess JM, Stewart C, Braunstein LZ, Arndt PF, Mordecai S, Shih AR, Chaves F, Zhan T, Lubitz CC, Kim J, Iafrate AJ, Wirth L, Parangi S, Leshchiner I, Daniels GH, Mootha VK, Dias-Santagata D, Getz G, McFadden DG, 2018)Widespread chromosomal losses and mitochondrial DNA alterations as genetic drivers in Hurthle cell carcinoma [J]. Cancer Cell 34(2):242–255 Giaginis C, Alexandrou P, Delladetsima I, Karavokyros I, Danas E, Giagini A, Patsouris E, Theocharis S, 2016, Clinical significance of Hu-antigen receptor, HuR, and cyclooxygenase-2, COX-2, expression in human malignant and benign thyroid lesions [J]. Pathol Oncol Res 22(1):189–196 Savin S, Cvejic D, Isic T, Petrovic I, Paunovic I, Tatic S, HavelkaM, 2006, Thyroid peroxidase immunohistochemistry in differential diagnosis of thyroid tumors [J]. Endocr Pathol 17(1):53–60 Sumana BS, Shashidhar S, Shivarudrappa AS, 2015, Galectin-3 immunohistochemical expression in thyroid neoplasms [J]. J Clin Diagn Res 9(11):EC07–EC11 Saleh HA, Jin B, Barnwell J, Alzohaili O, 2010, Utility of immunohistochemical markers in differentiating benign from malignant follicular derived thyroid nodules [J]. Diagn Pathol 5:9 Sun L, Wei X, Liu X, Zhou D, Hu F, Zeng Y, Sun Y, Luo S, Zhang Y, Yi XP, 2016, Expression of prostaglandin E2 and EP receptors in human papillary thyroid carcinoma [J]. Tumour Biol 37(4):4689–4697 Liang M, Zhao J, 2018, Protein expressions of AIB1, p53 and Bcl- 2 in epithelial ovarian cancer and their correlations with the clinical pathological features and prognosis [J]. Eur Rev Med Pharmacol Sci 22(16):5134–5139 Ru Y, Chen XJ, Zhao ZW, Zhang PF, Feng SH, Gao Q, Gao SG, Feng XS, 2017, CyclinD1 and p57kip2 as biomarkers in differentiation, metastasis and prognosis of gastric cardia adenocarcinoma [J]. Oncotarget 8(43):73860–73870 Malmstrom PU, Hemdan T, Segersten U, 2017, Validation of the ezrin, CK20, and Ki-67 as potential predictive markers for BCG instillation therapy of non-muscle-invasive bladder cancer [J]. Urol Oncol 35(8):532.e1–532.e6 Mu N, Juhlin CC, Tani E, Sofiadis A, Reihner E, Zedenius J, Larsson C, Nilsson IL, 2018, High Ki-67 index in fine needle aspiration cytology of follicular thyroid tumors is associated with increased risk of carcinoma [J]. Endocrine 61(2):293–302 Erickson LA, Jin L, Goellner JR, Lohse C, Pankratz VS, Zukerberg LR, Thompson GB, van Heerden JA, Grant CS, Lloyd RV, 2000, Pathologic features, proliferative activity, and CyclinD1 expression in Hurthle cell neoplasms of the thyroid [J]. Mod Pathol 13(2):186–192 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 697 EP 701 DI 10.1007/s12253-018-0546-x PG 5 ER PT J AU Qin, L Luo, JZ Tang, XL Han, ChG AF Qin, Lu Luo, Jin-Zhu Tang, Xue-Lian Han, Chuan-Gang TI Identification of Long Noncoding RNA MIR22HG as a Novel Biomarker in Thyroid Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thyroid cancer; MIR22HG; Long non-coding RNA; Bioinformatics analysis; Biomarker ID Thyroid cancer; MIR22HG; Long non-coding RNA; Bioinformatics analysis; Biomarker AB Thyroid cancer (TC) is the one of the most common endocrine malignancy. However, currently there are no specific and sensitive biomarkers for predicting the prognosis for TC. In this study, we for the first time showed MIR22HG was down-regulated in thyroid cancer by analyzing public datasets, including TCGA, GSE29265, GSE33630, and GSE55091. Furthermore, we observed the lower expression levels of MIR22HG were significantly related to higher age, lymph node metastasis status, residual tumor status, N stage, Grade, and T stage in TC. We also observed higher MIR22HG expression was associated with longer overall and disease-free survival time in TC. In order to explore the potential mechanisms of MIR22HG regulating TC progression, 4 hub gene networks regulated by MIR22HG were constructed in the present study. Bioinformatics analysis showed MIR22HG was associated with apoptotic process, regulation of transcription, mRNA splicing, regulation of cell cycle, and Hippo signaling pathway in TC. These results suggested MIR22HG could serve as a novel biomarker for thyroid cancer. C1 [Qin, Lu] Yangtze University, The Second Clinical Medical College, Jingzhou Central Hospital, Department of Thyroid Vascular Surgery, 430200 Jingzhou, Hubei, China. [Luo, Jin-Zhu] Yangtze University, The Second Clinical Medical College, Jingzhou Central Hospital, Department of Clinical Laboratory, 430200 Jingzhou, Hubei, China. [Tang, Xue-Lian] Yangtze University, The Second Clinical Medical College, Jingzhou Central Hospital, Department of Respiratory Medicine, 430200 Jingzhou, Hubei, China. [Han, Chuan-Gang] The First People’s Hospital of Jiangxia District Wuhan City, Department of Anesthesiology, No. 1, Zhifang Culture Avenue, Jiangxia District, 430200 Wuhan, Hubei, China. RP Han, ChG (reprint author), The First People’s Hospital of Jiangxia District Wuhan City, Department of Anesthesiology, 430200 Wuhan, China. EM 34826551@qq.com CR Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(1):7–30 Siegel RL, Miller KD, Jemal A, 2015, Cancer statistics, 2015. CA Cancer J Clin 65(1):5–29 Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66(1):7–30 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J, 2016, Cancer statistics in China, 2015. CA Cancer J Clin 66(2):115–132 Wang Z, Yang B, Zhang M, GuoW,Wu Z,Wang Y, Jia L, Li S, Xie W, Yang D, Caesar-Johnson SJ, Demchok JA, Felau I, Kasapi M, Ferguson ML, Hutter CM, Sofia HJ, Tarnuzzer R,Wang Z, Yang L, Zenklusen JC, Zhang J(J), Chudamani S, Liu J, Lolla L, Naresh R, Pihl T, Sun Q, Wan Y, Wu Y, Cho J, DeFreitas T, Frazer S, Gehlenborg N, Getz G, Heiman DI, Kim J, Lawrence MS, Lin P, Meier S, Noble MS, Saksena G, Voet D, Zhang H, Bernard B, Chambwe N, Dhankani V, Knijnenburg T, Kramer R, Leinonen K, Liu Y, Miller M, Reynolds S, Shmulevich I, Thorsson V, Zhang W, Akbani R, Broom BM, Hegde AM, Ju Z, Kanchi RS, Korkut A, Li J, Liang H, Ling S, Liu W, Lu Y, Mills GB, Ng KS, Rao A, Ryan M, Wang J, Weinstein JN, Zhang J, Abeshouse A, Armenia J, Chakravarty D, Chatila WK, Bruijn I, Gao J, Gross BE, Heins ZJ, Kundra R, la K, Ladanyi M, Luna A, NissanMG, Ochoa A, Phillips SM, Reznik E, Sanchez-Vega F, Sander C, Schultz N, Sheridan R, Sumer SO, Sun Y, Taylor BS,Wang J, Zhang H, Anur P, Peto M, Spellman P, Benz C, Stuart JM,Wong CK, Yau C, Hayes DN, Parker JS, Wilkerson MD, Ally A, Balasundaram M, Bowlby R, Brooks D, Carlsen R, Chuah E, Dhalla N, Holt R, Jones SJM, Kasaian K, Lee D, Ma Y, Marra MA, Mayo M, Moore RA, Mungall AJ, Mungall K, Robertson AG, Sadeghi S, Schein JE, Sipahimalani P, Tam A, Thiessen N, Tse K, Wong T, Berger AC, Beroukhim R, CherniackAD, Cibulskis C,Gabriel SB, Gao GF, Ha G, Meyerson M, Schumacher SE, Shih J, Kucherlapati MH, Kucherlapati RS, Baylin S, Cope L, Danilova L, Bootwalla MS, Lai PH, Maglinte DT, van den Berg DJ, Weisenberger DJ, Auman JT, Balu S, Bodenheimer T, Fan C, Hoadley KA, Hoyle AP, Jefferys SR, Jones CD, Meng S, Mieczkowski PA, Mose LE, Perou AH, Perou CM, Roach J, Shi Y, Simons JV, Skelly T, Soloway MG, Tan D, Veluvolu U, Fan H, Hinoue T, Laird PW, Shen H, Zhou W, Bellair M, Chang K, Covington K, Creighton CJ, Dinh H, Doddapaneni HV, Donehower LA, Drummond J, Gibbs RA, Glenn R, Hale W, Han Y, Hu J, Korchina V, Lee S, Lewis L, Li W, Liu X, Morgan M, Morton D, Muzny D, Santibanez J, Sheth M, Shinbrot E, Wang L, Wang M, Wheeler DA, Xi L, Zhao F, Hess J, Appelbaum EL, Bailey M, Cordes MG, Ding L, Fronick CC, Fulton LA, Fulton RS, Kandoth C, Mardis ER, McLellan MD, Miller CA, Schmidt HK, Wilson RK, Crain D, Curley E, Gardner J, Lau K, Mallery D, Morris S, Paulauskis J, Penny R, Shelton C, Shelton T, Sherman M, Thompson E, Yena P, Bowen J, Gastier-Foster JM, Gerken M, Leraas KM, Lichtenberg TM, Ramirez NC, Wise L, Zmuda E, Corcoran N, Costello T, Hovens C, Carvalho AL, de Carvalho AC, Fregnani JH, Longatto-Filho A, Reis RM, Scapulatempo- Neto C, Silveira HCS, Vidal DO, Burnette A, Eschbacher J, Hermes B, Noss A, Singh R, Anderson ML, Castro PD, Ittmann M, Huntsman D, Kohl B, le X, Thorp R, Andry C, Duffy ER, Lyadov V, Paklina O, Setdikova G, Shabunin A, Tavobilov M, McPherson C, Warnick R, Berkowitz R, Cramer D, Feltmate C, Horowitz N, Kibel A, Muto M, Raut CP, Malykh A, Barnholtz- Sloan JS, Barrett W, Devine K, Fulop J, Ostrom QT, Shimmel K, Wolinsky Y, Sloan AE, de Rose A, Giuliante F, Goodman M, Karlan BY, Hagedorn CH, Eckman J, Harr J, Myers J, Tucker K, Zach LA, Deyarmin B, Hu H, Kvecher L, Larson C, Mural RJ, Somiari S, Vicha A, Zelinka T, Bennett J, Iacocca M, Rabeno B, Swanson P, Latour M, Lacombe L, Tetu B, Bergeron A, McGraw M, Staugaitis SM, Chabot J, Hibshoosh H, Sepulveda A, Su T, Wang T, Potapova O, Voronina O, Desjardins L, Mariani O, Roman-Roman S, Sastre X, Stern MH, Cheng F, Signoretti S, Berchuck A, Bigner D, Lipp E, Marks J, McCall S, McLendon R, Secord A, Sharp A, Behera M, Brat DJ, Chen A, Delman K, Force S, Khuri F, Magliocca K, Maithel S, Olson JJ, Owonikoko T, Pickens A, Ramalingam S, Shin DM, Sica G, van Meir EG, Zhang H, Eijckenboom W, Gillis A, Korpershoek E, Looijenga L, OosterhuisW, Stoop H, van Kessel KE, Zwarthoff EC, Calatozzolo C, Cuppini L, Cuzzubbo S, DiMeco F, Finocchiaro G, Mattei L, Perin A, Pollo B, Chen C, Houck J, Lohavanichbutr P, Hartmann A, Stoehr C, Stoehr R, Taubert H, Wach S, Wullich B, Kycler W, Murawa D, Wiznerowicz M, Chung K, Edenfield WJ, Martin J, Baudin E, Bubley G, Bueno R, de Rienzo A, Richards WG, Kalkanis S, Mikkelsen T, Noushmehr H, Scarpace L, Girard N, Aymerich M, Campo E, Gine E, Guillermo AL, van Bang N, Hanh PT, Phu BD, Tang Y, Colman H, Evason K, Dottino PR, Martignetti JA, Gabra H, Juhl H, Akeredolu T, Stepa S, Hoon D, Ahn K, Kang KJ, Beuschlein F, Breggia A, BirrerM, Bell D, Borad M, Bryce AH, Castle E, Chandan V, Cheville J, Copland JA, Farnell M, Flotte T, Giama N, Ho T, Kendrick M, Kocher JP, Kopp K, Moser C, Nagorney D, O’Brien D, O’Neill BP, Patel T, Petersen G, Que F, Rivera M, Roberts L, Smallridge R, Smyrk T, Stanton M, Thompson RH, Torbenson M, Yang JD, Zhang L, Brimo F, Ajani JA, Gonzalez AMA, Behrens C, Bondaruk J, Broaddus R, Czerniak B, Esmaeli B, Fujimoto J, Gershenwald J, Guo C, Lazar AJ, Logothetis C, Meric-Bernstam F, Moran C, Ramondetta L, Rice D, Sood A, Tamboli P, Thompson T, Troncoso P, Tsao A, Wistuba I, Carter C, Haydu L, Hersey P, Jakrot V, Kakavand H, Kefford R, Lee K, Long G, Mann G, Quinn M, Saw R, Scolyer R, Shannon K, Spillane A, Stretch J, Synott M, Thompson J, Wilmott J, al-Ahmadie H, Chan TA, Ghossein R, Gopalan A, Levine DA, Reuter V, Singer S, Singh B, Tien NV, Broudy T, Mirsaidi C, Nair P, Drwiega P, Miller J, Smith J, Zaren H, Park JW, Hung NP, Kebebew E, Linehan WM, Metwalli AR, Pacak K, Pinto PA, Schiffman M, Schmidt LS, Vocke CD,Wentzensen N,Worrell R, Yang H, Moncrieff M, Goparaju C, Melamed J, Pass H, Botnariuc N, Caraman I, Cernat M, Chemencedji I, Clipca A, Doruc S, Gorincioi G, Mura S, Pirtac M, Stancul I, Tcaciuc D, Albert M, Alexopoulou I, Arnaout A, Bartlett J, Engel J, Gilbert S, Parfitt J, Sekhon H, Thomas G, Rassl DM, Rintoul RC, Bifulco C, Tamakawa R, Urba W, Hayward N, Timmers H, Antenucci A, Facciolo F, Grazi G, Marino M, Merola R, de Krijger R, Gimenez-Roqueplo AP, Piche A, Chevalier S, McKercher G, Birsoy K, Barnett G, Brewer C, Farver C, Naska T, Pennell NA, Raymond D, Schilero C, Smolenski K, Williams F, Morrison C, Borgia JA, Liptay MJ, Pool M, Seder CW, Junker K, Omberg L, Dinkin M, Manikhas G, Alvaro D, Bragazzi MC, Cardinale V, Carpino G, Gaudio E, Chesla D, Cottingham S, Dubina M, Moiseenko F, Dhanasekaran R, Becker KF, Janssen KP, Slotta-Huspenina J, Abdel-Rahman MH, Aziz D, Bell S, Cebulla CM, Davis A, Duell R, Elder JB, Hilty J, Kumar B, Lang J, Lehman NL, Mandt R, Nguyen P, Pilarski R, Rai K, Schoenfield L, Senecal K, Wakely P, Hansen P, Lechan R, Powers J, Tischler A, Grizzle WE, Sexton KC, Kastl A, Henderson J, Porten S, Waldmann J, Fassnacht M, Asa SL, Schadendorf D, Couce M, Graefen M, Huland H, Sauter G, Schlomm T, Simon R, Tennstedt P, Olabode O, Nelson M, Bathe O, Carroll PR, Chan JM, Disaia P, Glenn P, Kelley RK, Landen CN, Phillips J, Prados M, Simko J, Smith-McCune K, VandenBerg S, Roggin K, Fehrenbach A, Kendler A, Sifri S, Steele R, Jimeno A, Carey F, Forgie I, MannelliM, CarneyM, Hernandez B, Campos B, Herold-Mende C, Jungk C, Unterberg A, von Deimling A, Bossler A, Galbraith J, Jacobus L, Knudson M, Knutson T, Ma D, Milhem M, Sigmund R, Godwin AK, Madan R, Rosenthal HG, Adebamowo C, Adebamowo SN, Boussioutas A, Beer D, Giordano T, Mes-Masson AM, Saad F, Bocklage T, Landrum L, Mannel R, Moore K, Moxley K, Postier R, Walker J, Zuna R, Feldman M, Valdivieso F, Dhir R, Luketich J, Pinero EMM, Quintero-Aguilo M, Carlotti CG Jr, Dos Santos JS, Kemp R, Sankarankuty A, Tirapelli D, Catto J, Agnew K, Swisher E, Creaney J, Robinson B, Shelley CS, Godwin EM, Kendall S, Shipman C, Bradford C, Carey T, Haddad A, Moyer J, Peterson L, Prince M, Rozek L, Wolf G, Bowman R, Fong KM, Yang I, Korst R, Rathmell WK, Fantacone-Campbell JL, Hooke JA, Kovatich AJ, Shriver CD, DiPersio J, Drake B, Govindan R, Heath S, Ley T, van Tine B, Westervelt P, Rubin MA, Lee JI, Aredes ND, Mariamidze A, 2018, lncRNA epigenetic landscape analysis identifies EPIC1 as an oncogenic lncRNA that interacts with MYC and promotes cell-cycle progression in Cancer. Cancer Cell 33(4):706–720 Shen Z, HaoW, Zhou C, Deng H, Ye D, Li Q, Lin L, Cao B, Guo J, 2018, Long non-coding RNA AC026166.2-001 inhibits cell proliferation and migration in laryngeal squamous cell carcinoma by regulating the miR-24-3p/p27 axis. Sci Rep 8(1):3375 Tosoian JJ, Patel HD, Mamawala M, Landis P, Wolf S, Elliott DJ, Epstein JI, Carter HB, Ross AE, Sokoll LJ, Pavlovich CP, 2017, Longitudinal assessment of urinary PCA3 for predicting prostate cancer grade reclassification in favorablerisk men during active surveillance. Prostate Cancer Prostatic Dis 20(3):339–342 Alshalalfa M, Verhaegh GW, Gibb EA, Santiago-Jimenez M, Erho N, Jordan J et al, 2017, Low PCA3 expression is a marker of poor differentiation in localized prostate tumors: exploratory analysis from 12,076 patients. Oncotarget 8(31):50804–50813 Chunhua L, Zhao H, Zhao H, Lu Y, Wu J, Gao Z, Li G, Zhang Y, Wang K, 2018, Clinical significance of peripheral blood PCA3 gene expression in early diagnosis of prostate Cancer. Transl Oncol 11(3):628–632 Nakanishi H, Groskopf J, Fritsche HA, Bhadkamkar V, Blase A, Kumar SV, Davis JW, Troncoso P, Rittenhouse H, Babaian RJ, 2008, PCA3 molecular urine assay correlates with prostate cancer tumor volume: implication in selecting candidates for active surveillance. J Urol 179(5):1804–1809 1809-1810 KimD, LeeWK, Jeong S, SeolMY, KimH, KimKS, Lee EJ, Lee J, Jo YS, 2016, Upregulation of long noncoding RNA LOC100507661 promotes tumor aggressiveness in thyroid cancer. Mol Cell Endocrinol 431:36–45 Barros-Filho MC, Marchi FA, Pinto CA, Rogatto SR, Kowalski LP, 2015, High diagnostic accuracy based on CLDN10, HMGA2, and LAMB3 transcripts in papillary thyroid carcinoma. J Clin Endocrinol Metab 100(6):E890–E899 Dom G, TarabichiM, Unger K, Thomas G, Oczko-Wojciechowska M, Bogdanova T, Jarzab B, Dumont JE, Detours V, Maenhaut C, 2012, A gene expression signature distinguishes normal tissues of sporadic and radiation-induced papillary thyroid carcinomas. Br J Cancer 107(6):994–1000 Hou S, Lin Q, Guan F, Lin C, 2018, LncRNA TNRC6C-AS1 regulates UNC5B in thyroid cancer to influence cell proliferation, migration, and invasion as a competing endogenous RNA of miR- 129-5p. J Cell Biochem 119(10):8304–8316 Liao T, Qu N, Shi RL, Guo K, Ma B, Cao YM et al, 2017, BRAFactivated LncRNA functions as a tumor suppressor in papillary thyroid cancer. Oncotarget 8(1):238–247 Qin Y, SunW, Zhang H, Zhang P,Wang Z, DongW,He L, Zhang T, Shao L, Zhang W, Wu C, 2018, LncRNA GAS8-AS1 inhibits cell proliferation through ATG5-mediated autophagy in papillary thyroid cancer. Endocrine 59(3):555–564 Tani H, Onuma Y, Ito Y, Torimura M, 2014, Long non-coding RNAs as surrogate indicators for chemical stress responses in human-induced pluripotent stem cells. PLoS One 9(8):e106282 Su W, Feng S, Chen X, Yang X, Mao R, Guo C et al, 2018, Silencing of long noncoding RNA MIR22HG triggers cell survival/death signaling via oncogenes YBX1, MET, and p21 in lung cancer. Cancer Res 78(12):3207–3219 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 703 EP 710 DI 10.1007/s12253-018-0521-6 PG 8 ER PT J AU Nasri, S Humara, B Anjomshoaa, A Moradi, N Gholipour, N Mashjoor, S Zhang, P AF Nasri, Soroush Humara, Bostjan Anjomshoaa, Ahmad Moradi, Nourodin Gholipour, Naghmeh Mashjoor, Sakineh Zhang, Peng TI Early Hereditary Diffuse Gastric Cancer (eHDGC) is Characterized by Subtle Genomic Instability and Active DNA Damage Response Soroush SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Genomic instability; Diffuse gastric cancer; Array CGH; DNA fragility ID Genomic instability; Diffuse gastric cancer; Array CGH; DNA fragility AB Diffuse gastric cancer (DGC) is one of the two primary types of stomach cancer. Carriers of germline mutations in the gene encoding E-cadherin are predisposed to DGC. The primary aim of the present study was to determine if genomic instability is an early event in DGC and how it may lead to disease progression. Chromosomal aberrations in early intramucosal hereditary diffuse gastric cancer (eHDGC) were assessed using array comparative genomic hybridization (array CGH). Notably, no aneuploidy or other large-scale chromosomal rearrangements were detected. Instead, all aberrations affected small regions (< 4.8 Mb) and were predominantly deletions. Analysis of DNA sequence patterns revealed that essentially all aberrations possessed the characteristics of common fragile sites. These results and the results of subsequent immunohistochemical examinations demonstrated that unlike advanced DGC, eHDGCs is characterized by low levels of genomic instability at fragile sites. Furthermore, they express an active DNA damage response, providing a molecular basis for the observed indolence of eHDGC. This finding is an important step to understanding the pathology underlying natural history of DGC and supports a revision of the current definition of eHDGC as a malignant disease. C1 [Nasri, Soroush] University of Otago, Department of Biochemistry, Cancer Genetics Laboratory, 9054 Dunedin, New Zealand. [Humara, Bostjan] University of Otago, Department of Biochemistry, Cancer Genetics Laboratory, 9054 Dunedin, New Zealand. [Anjomshoaa, Ahmad] University of Otago, Department of Biochemistry, Cancer Genetics Laboratory, 9054 Dunedin, New Zealand. [Moradi, Nourodin] Milad Center for Medical GeneticsSari, Iran. [Gholipour, Naghmeh] Milad Center for Medical GeneticsSari, Iran. [Mashjoor, Sakineh] Hormozgan University, Faculty of Marine Science and Technology, Department of Marine biologyBandar Abbas, Iran. [Zhang, Peng] Australian National University, ANU College of Medicine, Biology and Environment, Research School of Biology, Division of Biomedical Science and BiochemistryCanberra, Australia. RP Gholipour, N (reprint author), Milad Center for Medical Genetics, Sari, Iran. EM ngholipour@nigeb.ac.ir CR Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 CrewKD, 2006, Neugut AI. Epidemiology of gastric cancer.World J Gastroenterol 12:354–362 Lauren P, 1965, The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand 64: 31–49 Ekstrom AM, Hansson LE, Signorello LB, Lindgren A, Bergstrom R, Nyren O, 2000, Decreasing incidence of both major histologic subtypes of gastric adenocarcinoma-a population-based study in Sweden. Br J Cancer 83:391–396 Henson DE, Dittus C, Younes M, Nguyen H, Albores-Saavedra J, 2004, Differential trends in the intestinal and diffuse types of gastric carcinoma in the United States, 1973-2000: increase in the signet ring cell type. Arch Pathol Lab Med 128:765–770 Kattan MW, Karpeh MS, Mazumdar M, Brennan MF, 2003, Postoperative nomogram for disease-specific survival after an R0 resection for gastric carcinoma. J Clin Oncol 21:3647–3650 Stone J, Bevan S, Cunningham D, Hill A, Rahman N, Peto J, Marossy A, Houlston RS, 1999, Low frequency of germline Ecadherin mutations in familial and nonfamilial gastric cancer. Br J Cancer 79:1935–1937 Guilford P, Hopkins J, Harraway J, McLeod M, McLeod N, Harawira P, Taite H, Scoular R, Miller A, Reeve AE, 1998, Ecadherin germline mutations in familial gastric cancer. Nature 392:402–405 Becker KF, Atkinson MJ, Reich U, Becker I, Nekarda H, Siewert JR, Hofler H, 1994, E-cadherin gene mutations provide clues to diffuse type gastric carcinomas. Cancer Res 54:3845–3852 Tamura G, Yin J,Wang S, Fleisher AS, Zou T, Abraham JM, Kong D, Smolinski KN,Wilson KT, James SP, Silverberg SG, Nishizuka S, Terashima M, Motoyama T, Meltzer SJ, 2000, E-cadherin gene promoter hypermethylation in primary human gastric carcinomas. J Natl Cancer Inst 92:569–573 Machado JC, Oliveira C, Carvalho R, Soares P, Berx G, Caldas C, Seruca R, Carneiro F, Sobrinho-Simoes M, 2001, E-cadherin gene, CDH1, promoter methylation as the second hit in sporadic diffuse gastric carcinoma. Oncogene 20:1525–1528 Blair V, Martin I, Shaw D, Winship I, Kerr D, Arnold J, Harawira P, McLeod M, Parry S, Charlton A, Findlay M, Cox B, Humar B, More H, Guilford P, 2006, Hereditary diffuse gastric cancer: diagnosis and management. Clin Gastroenterol Hepatol 4:262–275 Humar B, Fukuzawa R, Blair V, Dunbier A, More H, Charlton A, , Kim WH, Reeve AE, Martin I, Guilford P, 2007, Destabilized adhesion in the gastric proliferative zone and c-Src kinase activation mark the development of early diffuse gastric cancer. Cancer Res 67: 2480–2489 Le Borgne R, Bellaiche Y, Schweisguth F, 2002, Drosophila Ecadherin regulates the orientation of asymmetric cell division in the sensory organ lineage. Curr Biol 12:95–104 Schluter MA, Pfarr CS, Pieczynski J, Whiteman EL, Hurd TW, Fan S, Liu CJ, Margolis B, 2009, Trafficking of Crumbs3 during cytokinesis is crucial for lumen formation. Mol Biol Cell 20(22):4652–4663 Stehbens SJ, Akhmanova A, Yap AS, 2009, Microtubules and cadherins: aneglected partnership. Front Biosci 14:3159–3167 Nasri S, Anjomshoaa A, Song S, Guilford P, McNoe L, Black M, Phillips V, Reeve A, Humar B, 2010, Oligonucleotide array outperforms SNP array on formalin-fixed paraffin-embedded clinical samples. Cancer Genet Cytogenet 198(1):1–6 Cleveland WS, 1979, Robust locally weighted regression and smoothing scatterplots. J Am Stat Assoc 74(368):829–836 van Dijk MC, Rombout PD, Boots-Sprenger SH, Straatman H, Bernsen MR, Ruiter DJ, Jeuken JW, 2005, Multiplex ligationdependent probe amplification for the detection of chromosomal gains and losses in formalin-fixed tissue. Diagn Mol Pathol 14(1):9–16 Sarai A, Mazur J, Nussinov R, Jernigan RL, 1989, Sequence dependence of DNA conformational flexibility. Biochem 28(19): 7842–7849 Mishmar D, Rahat A, Scherer SW, Nyakatura G, Hinzmann B, Kohwi Y, Mandel-Gutfroind Y, Lee JR, Drescher B, Sas DE, Margalit H, 1998, Molecular characterization of a common fragile site, FRA7H, on human chromosome 7 by the cloning of a simian virus 40 integration site. Proc Natl Acad Sci U S A 95:8141–8146 Humar B, Guilford P, 2009, Hereditary diffuse gastric cancer: a manifestation of lost cell polarity. Cancer Sci 100(7):1151–1157 Mimata A, Fukamachi H, Eishi Y, Yuasa Y, 2011, Loss of Ecadherin in mouse gastric epithelial cells induces signet ring-like cells, a possible precursor lesion of diffuse gastric cancer. Cancer Sci 102(5):942–950 Thoma CR, Toso A, Gutbrodt KL, Reggi SP, Frew IJ, Schraml P, Hergovich A, Moch H,Meraldi P, KrekW(2009, VHL loss causes spindle misorientation and chromosome instability. Nature Cell Biol 11(8):994–1001 Little SE, Vuononvirta R, Reis-Filho JS, Natrajan R, Iravani M, Fenwick K, Mackay A, Ashworth A, Pritchard-Jones K, Jones C, 2006, Array CGH using whole genome amplification of freshfrozen and formalin-fixed, paraffin-embedded tumor DNA. Genomics 87(2):298–306 Mc Sherry EA, Mc Goldrick A, Kay EW, Hopkins AM, Gallagher WM, Dervan PA, 2007, Formalin-fixed paraffin-embedded clinical tissues show spurious copy number changes in array-CGH profiles. Clin Genet 72(5):441–447 Talseth-Palmer BA, Bowden NA, Hill A, Meldrum C, Scott RJ, 2008, Whole genome amplification and its impact on CGH array profiles. BMC Res Notes 1(1):56 De Smith AJ, Tsalenko A, Sampas N, Scheffer A, Yamada NA, Tsang P, Ben-Dor A, Yakhini Z, Ellis RJ, Bruhn L, Laderman S, 2007, Array CGH analysis of copy number variation identifies 1284 new genes variant in healthy white males: implications for association studies of complex diseases. Hum Mol Genet 16(23): 2783–2794 Brunet A, Armengol L, Heine D, Rosell J, Garcia-Aragones M, Gabau E, Estivill X, Guitart M, 2009, BAC array CGH in patients with Velocardiofacial syndrome-like features reveals genomic aberrations on chromosome region 1q21. 1. BMCMed Genet 10(1):144 Ambros IM, Brunner B, Aigner G, Bedwell C, Beiske K, Benard J, Bown N, Combaret V, Couturier J, Defferrari R, Gross N, 2011, A multilocus technique for risk evaluation of patients with neuroblastoma. Clin Cancer Res 17(4):792–804 Shen Y, Wu BL, 2009, Microarray-based genomic DNA profiling technologies in clinical molecular diagnostics. Clin Chem 55(4): 659–669 Hills A, Ahn JW, Donaghue C, Thomas H, Mann K, Ogilvie CM, 2010, MLPA for confirmation of array CGH results and determination of inheritance. Mol Cytogen 3(1):19 Cook JG, 2009, Replication licensing and the DNA damage checkpoint. Front Biosci, Landmark edition, 14:5013 Bakkenist CJ, Kastan MB, 2003, DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature 421(6922):499–506 Abraham RT, 2001, Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev 15(17):2177–2196 Kumagai A, Dunphy WG, 2006, How cells activate ATR. Cell Cycle 5(12):1265–1268 CasperAM, Durkin SG, ArltMF, Glover TW(2004, Chromosomal instability at common fragile sites in Seckel syndrome. Am J Hum Genet 75(4):654–660 Durkin SG, Glover TW, 2007, Chromosome fragile sites. Annu Rev Genet 41:169–192 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144(5):646–674 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 711 EP 721 DI 10.1007/s12253-018-0547-9 PG 11 ER PT J AU Park, YJ Hong, GyD Chong, OG Park, YJ AF Park, Young Ji Hong, Gy Dae Chong, Oh Gun Park, Y Ji TI Tumor Budding is a Valuable Diagnostic Parameter in Prediction of Disease Progression of Endometrial Endometrioid Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial endometrioid carcinoma; Tumor budding; Prognosis; Lymphovascular invasion; Lymph node involvement ID Endometrial endometrioid carcinoma; Tumor budding; Prognosis; Lymphovascular invasion; Lymph node involvement AB Recently, tumor budding (TB) found at the invasive margin has been related to lymph node involvement (LNI), local recurrence, and poor prognosis in various cancers. We assessed the presence of TB in endometrial endometrioid carcinoma (EEC), and examined the immunohistochemical (IHC) profiles to define its clinicopathological significance. Ninety-six EECs were obtained from 2008 to 2013. During the follow-up, ten patients experienced disease progression; of these, three patients succumbed to the disease. All hematoxylin and eosin-stained slides were scrutinized for the presence of TB. IHC stainings for estrogen receptor (ER), progesterone receptor (PR), β-catenin, and E-cadherin were performed. All cases were grouped as FIGO grade (G) 1 (47.9%), G2 (29.2%), and G3 (22.9%). The distribution for depth of invasion (DOI) was 68.5% with a DOI of less than half and 31.5% with a DOI of more than half. Myometrial invasion was characterized as infiltrating pattern (52.1%), adenomyosis-like (20.8%), microcystic, elongated, and fragmented (17.7%), or expansile (9.4%). TB was identified in 63 cases (65.6%). Lymphovascular invasion (LVI) and LNI were identified in 47 and 37 cases, respectively. TB was associated with deep DOI (p = 0.001), higher FIGO grade (p = 0.006), LVI (p < 0.0001), and LNI (p < 0.0001). TB showed loss of ER (p < 0.0001) and PR (p < 0.0001), reduced E-cadherin (p < 0.0001) expression, and aberrant β-catenin expression (p = 0.042). In EECs, TB was associated with deep DOI, less-differentiated histology, frequent LVI, and LNI; furthermore, TB was closely related to epithelial-mesenchymal transition phenotype and downregulation of hormonal receptors. Therefore, TB might be a determinant histologic clue for prediction of disease progression in EECs. C1 [Park, Young Ji] Kyungpook National University, School of Medicine, Chilgok Hospital, Department of PathologyDaegu, South Korea. [Hong, Gy Dae] Kyungpook National University, School of Medicine, Chilgok Hospital, Department of Obstetrics and GynecologyDaegu, South Korea. [Chong, Oh Gun] Kyungpook National University, School of Medicine, Chilgok Hospital, Department of Obstetrics and GynecologyDaegu, South Korea. [Park, Y Ji] Kyungpook National University, School of Medicine, Chilgok Hospital, Department of PathologyDaegu, South Korea. RP Park, YJ (reprint author), Kyungpook National University, School of Medicine, Chilgok Hospital, Department of Pathology, Daegu, South Korea. EM pathpjy@naver.co.kr CR Cole AJ, Quick CM, 2013, Patterns of myoinvasion in endometrial adenocarcinoma: recognition and implications. Adv Anat Pathol 20(3):141–147 Bignotti E, Ravaggi A, Romani C, FalchettiM, Lonardi S, Facchetti F, Pecorelli S, Varughese J, Cocco E, Bellone S, Schwartz PE, Rutherford TJ, Santin AD, 2011, Trop-2 overexpression in poorly differentiated endometrial endometrioid carcinoma: implications for immunotherapy with hRS7, a humanized anti-trop-2 monoclonal antibody. Int J Gynecol Cancer 21(9):1613–1621 Euscher E, Fox P, Bassett R, al-Ghawi H, Ali-Fehmi R, Barbuto D, Djordjevic B, Frauenhoffer E, Kim I, Hong SR, Montiel D, Moschiano E, Roma A, Silva E, Malpica A, 2013, The pattern of myometrial invasion as a predictor of lymph node metastasis or extrauterine disease in low-grade endometrial carcinoma. Amer J Surg Pathol 37(11):1728–1736 Choi HJ, Park KJ, Shin JS, Roh MS, Kwon HC, Lee HS, 2007, Tumor budding as a prognosticmarker in stage-III rectal carcinoma. Int J Color Dis 22(8):863–868 Kanazawa H, Mitomi H, Nishiyama Y, Kishimoto I, Fukui N, Nakamura T, Watanabe M, 2008, Tumour budding at invasive margins and outcome in colorectal cancer. Color Dis 10(1):41–47 Roh MS, Lee JI, Choi PJ, 2004, Tumor budding as a useful prognostic marker in esophageal squamous cell carcinoma. Dis Esophagus 17(4):333–337 Sarioglu S, Acara C, Akman FC, Dag N, Ecevit C, Ikiz AO, Cetinayak OH, Ada E, 2010, Tumor budding as a prognostic marker in laryngeal carcinoma. Pathol Res Pract 206(2):88–92 Koyuncuoglu M, Okyay E, Saatli B, Olgan S, Akin M, Saygili U, 2012, Tumor budding and E-cadherin expression in endometrial carcinoma: are they prognostic factors in endometrial cancer? Gynecol Oncol 125(1):208–213 Wei J, Zhang W, Feng L, Gao W, 2017, Comparison of fertilitysparing treatments in patients with early endometrial cancer and atypical complex hyperplasia: a meta-analysis and systematic review. Medicine, Baltimore, 96(37):e8034 Hanekamp EE, Kuhne EC, Smid-Koopman E et al, 2002, Loss of progesterone receptor may lead to an invasive phenotype in human endometrial cancer. Eur J Cancer 38(Suppl 6):S71–S72 Hanekamp EE, Gielen SC, Smid-Koopman E, Kuhne LC, de Ruiter PE, Chadha-Ajwani S, Brinkmann AO, Grootegoed JA, Burger CW, Huikeshoven FJ, Blok LJ, 2003, Consequences of loss of progesterone receptor expression in development of invasive endometrial cancer. Clin Cancer Res 9(11):4190–4199 Huang B, Cai J, Xu X, Guo S, Wang Z, 2016, High-grade tumor budding stratifies early-stage cervical Cancer with recurrence risk. PLoS One 11(11):e0166311 Pirker R, Pereira JR, von Pawel J, Krzakowski M, Ramlau R, Park K, de Marinis F, Eberhardt WEE, Paz-Ares L, Storkel S, Schumacher KM, von Heydebreck A, Celik I, O'Byrne KJ, 2012, EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: analysis of data from the phase 3 FLEX study. Lancet Oncol 13(1):33–42 Neubauer NL, Lurain JR, 2011, The role of lymphadenectomy in surgical staging of endometrial cancer. Int J Surg Oncol 2011: 814649 Lefringhouse JR, Elder JW, Baldwin LA, Miller RW, DeSimone CP, van Nagell JR Jr, Samoyoa LM,West DS, Dressler EV, Liu M, Ueland FR, 2017, Prospective validation of an intraoperative algorithm to guide surgical staging in early endometrial cancer. Gynecol Oncol 145(1):50–54 Zhang C,Wang C, FengW, 2012, Clinicopathological risk factors for pelvic lymph node metastasis in clinical early-stage endometrioid endometrial adenocarcinoma. Int J Gynecol Cancer 22(8):1373–1377 Koskas M, Fournier M, Vanderstraeten A, Walker F, Timmerman D, Vergote I, Amant F, 2016, Evaluation of models to predict lymph node metastasis in endometrial cancer: a multicentre study. Eur J Cancer 61:52–60 Montserrat N, Mozos A, Llobet D, Dolcet X, Pons C, de Herreros AG, Matias-Guiu X, Prat J, 2012, Epithelial to mesenchymal transition in early stage endometrioid endometrial carcinoma. Hum Pathol 43(5):632–643 Stewart CJ, 2013, Epithelial-to-mesenchymal transition in endometrioid adenocarcinoma of the endometrium. Hum Pathol 44(9):1956–1957 Colas E, Pedrola N, Devis L, Ertekin T, Campoy I, Martinez E, Llaurado M, Rigau M, Olivan M, Garcia M, Cabrera S, Gil- Moreno A, Xercavins J, Castellvi J, Garcia A, Ramon y Cajal S, Moreno-Bueno G, Dolcet X, Alameda F, Palacios J, Prat J, Doll A, Matias-Guiu X, Abal M, Reventos J, 2012, The EMT signaling pathways in endometrial carcinoma. Clin Transl Oncol 14(10): 715–720 Stewart CJ, Little L, 2009, Immunophenotypic features of MELF pattern invasion in endometrial adenocarcinoma: evidence for epithelial-mesenchymal transition. Histopathology 55(1):91–101 van der Horst PH,Wang Y, Vandenput I, Kuhne LC, Ewing PC, van IJckenWFJ, van der Zee M, Amant F, Burger CW, Blok LJ, 2012, Progesterone inhibits epithelial-to-mesenchymal transition in endometrial cancer. PLoS One 7(1):e30840 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 723 EP 730 DI 10.1007/s12253-018-0554-x PG 8 ER PT J AU Zimpfer, A Schneider, B Blanck, O Riedel, K Zhivov, A Jonigk, D Erbersdobler, A Junemann, A Andratschke, N Hildebrandt, G Guthoff, FR Kakkassery, V AF Zimpfer, Annette Schneider, Bjoern Blanck, Oliver Riedel, Katrin Zhivov, Andrey Jonigk, Danny Erbersdobler, Andreas Junemann, Anselm Andratschke, Nicolaus Hildebrandt, Guido Guthoff, F Rudolf Kakkassery, Vinodh TI Pathologic Features of Tumor Activity and Stability in Uveal Melanoma Specimens after Fractionated CyberKnife Radiosurgery SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Uveal melanoma; CyberKnife; Stereotactic radiosurgery; Proliferation index; DNA image cytometry ID Uveal melanoma; CyberKnife; Stereotactic radiosurgery; Proliferation index; DNA image cytometry AB To evaluate uveal melanoma cell activity and pathologic features after stereotactic CyberKnife radiosurgery in specimens from five patients. Specimens from five patients treated by CyberKnife radiosurgery in three fractions were included in this study. Because of persistent retinal detachment in 3 patients, tumour endoresection was performed at four, seven and ten month after CyberKnife radiosurgery. At nine and twelve months after treatment, enucleation of the eye globe was performed in 2 patients because of secondary tumour bleeding and missing regression. After histomorphological analysis and determination of Ki67-proliferation index, DNA cytophotometry, fluorescence in-situ hybridization evaluation for chromosome 3 loss, GNA11and GNAQ mutation analysis were performed. Four of the five tumours included in this study showed variable radiation-induced morphologic changes in the form of enlargement of cells and nuclei, cytoplasmic vacuolisation and nuclear fragmentation. The DNA content of a large fraction of tumour cells was hypoploid. On the other hand, single strikingly hyperchromatic melanoma cells showed marked aneuploidy. The proliferation fraction in the three endoresected tumours was very low (<1%), but it was elevated in the enucleation cases. Monosomy 3 was detected in two of the endoresection cases, but none of the enucleation cases. None of the patients experienced a local tumour recurrence, but two of the patients developed liver metastasis. Many melanoma cells seemed to be vital within the first 6 months after CyberKnife radiosurgery, but obvious radiation-induced morphologic changes, including tumour necrosis, hypoploid DNA content plus low Ki-67 index could indicate sublethal cell damage. C1 [Zimpfer, Annette] University Medicine Rostock, Institute of Pathology, Strempelstr. 14, 18057 Rostock, Germany. [Schneider, Bjoern] University Medicine Rostock, Institute of Pathology, Strempelstr. 14, 18057 Rostock, Germany. [Blanck, Oliver] University Medical Center Schleswig-Holstein, Department for Radiation Oncology, Arnold-Heller-Str. 3, 24105 Kiel, Germany. [Riedel, Katrin] University Medicine Rostock, Department of Ophthalmology, Doberaner Str. 140, 18057 Rostock, Germany. [Zhivov, Andrey] OSG MVZ Betriebs GmbH, Ophtalmology, OMS, Anesthetics, Buger Str, 82, 96049 Bamberg, Germany. [Jonigk, Danny] Hannover Medical School, Institute of Pathology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany. [Erbersdobler, Andreas] University Medicine Rostock, Institute of Pathology, Strempelstr. 14, 18057 Rostock, Germany. [Junemann, Anselm] University Medicine Rostock, Department of Ophthalmology, Doberaner Str. 140, 18057 Rostock, Germany. [Andratschke, Nicolaus] University Hospital Zurich, Department of Radiation Oncology, Ramistr. 100, 8091 Zurich, Switzerland. [Hildebrandt, Guido] University Medical Center Rostock, Department of Radiotherapy, Sudring 75, 18059 Rostock, Germany. [Guthoff, F Rudolf] University Medicine Rostock, Department of Ophthalmology, Doberaner Str. 140, 18057 Rostock, Germany. [Kakkassery, Vinodh] University Medicine Rostock, Department of Ophthalmology, Doberaner Str. 140, 18057 Rostock, Germany. RP Zimpfer, A (reprint author), University Medicine Rostock, Institute of Pathology, 18057 Rostock, Germany. EM annette.zimpfer@med.uni-rostock.de CR Eagle CR, 2011, Eye pathology. An atlas and text. Lippincott Williams & Wilkins, Philadelphia, pp 177–206 Font RL, Croxatto JO, Rao NA, 2006, AFIP atlas of tumor pathology. Tumors of the eye and ocular adnexa. Armed Forces Institute of Pathology, Washington DC Scotto J, Fraumeni JF, Lee JA, 1976, Melanomas of the eye and other noncutaneous sites: epidemiologic aspects. J Natl Cancer Inst 56:489–491 Bergman L, Seregard S, Nilsson B, Lundell G, Ringborg U, Ragnarsson-Olding B, 2002, Incidence of uveal melanoma in Sweden from 1960 to 1998. Invest Ophthalmol Vis Sci 43:2579–2583 Singh AD, Topham A, 2003, Incidence of uveal melanoma in the United States: 1973–1997. Ophthalmology 110:956–961 Damato B, 2012, Progress in the management of patients with uveal melanoma. The 2012 Ashton lecture. Eye, Lond, 2012 Sep; 26(9):1157-72. , DOI 10.1038/eye.2012.126 American Brachytherapy Society - Ophthalmic Oncology Task Force, 2014, The American brachytherapy society consensus guidelines for plaque brachytherapy of uveal melanoma and retinoblastoma. Brachytherapy 13:1–14. , DOI 10.1016/j.brachy.2013.11.008 Damato B, 1997, Adjunctive plaque radiotherapy after local resection of uveal melanoma. Front Radiat Ther Oncol 30:123–132 Bechrakis NE, Bornfeld N, Zoller I, Foerster MH, 2002, Iodine 125 plaque brachytherapy versus transscleral tumor resection in the treatment of large uveal melanomas. Ophthalmology 109:1855–1861 Zorlu F, Selek U, Kiratli H, 2009, Initial results of fractionated CyberKnife radiosurgery for uveal melanoma. J Neuro-Oncol 94: 111–117. , DOI 10.1007/s11060-009-9811-x Wackernagel W, Holl E, Tarmann L, Mayer C, Avian A, Schneider M, Kapp KS, Langmann G, 2014, Local tumour control and gamma-knife radiosurgery of choroidal melanomas. Br J Ophthalmol 98:218–223. , DOI 10.1136/bjophthalmol- 2013-304031 Schirmer CM, Chan M, Mignano J, Duker J,MelhusCS,Williams LB, Wu JK, Yao KC, 2009, Dose de-escalation with gamma knife radiosurgery in the treatment of choroidal melanoma. Int J RadiatOncol Biol Phys 75:170–176. , DOI 10.1016/j.ijrobp.2008.10.077 Sarici AM, Pazarli H, 2013, Gamma-knife-based stereotactic radiosurgery for medium- and large-sized posterior uveal melanoma. Graefes Arch Clin Exp Ophthalmol 251:285–294. , DOI 10.1007/s00417-012-2144-z Damato B, Patel I, Campbell IR, Mayles HM, Errington RD, 2005, Local tumor control after, 106)Ru brachytherapy of choroidal melanoma. Int J Radiat Oncol Biol Phys 63:385–391 Russo A, Laguardia M, Damato B, 2012, Eccentric ruthenium plaque radiotherapy of posterior choroidal melanoma. Graefes Arch Clin Exp Ophthalmol 250(10):1533–1540. , DOI 10.1007/s00417-012-1962-3 Stoffelns BM, Kutzner J, Jochem T, 2002, Retrospective analysis of ruthenium-106 brachytherapy for small and medium-sized malignant melanoma of the posterior choroid. Klin Monatsbl Augenheilkd 219(4):216–220. , DOI 10.1055/s-2002-30654 Shields CL, Cater J, Shields JA, Chao A, Krema H, Materin M, Brady LW, 2002, Combined plaque radiotherapy and transpupillary thermotherapy for choroidal melanoma: tumor control and treatment complications in 270 consecutive patients. Arch Ophthalmol 120:933–940 Klaus H, Lommatzsch PK, Fuchs U, 1991, Histopathology studies in human malignant melanomas of the choroid after unsuccessful treatment with106Ru/106Rh ophthalmic applicators. Graefes Arch Clin Exp Ophthalmol 229:480–486 Heindl LM, Lotter M, Strnad V, Sauer R, Naumann GO, Knorr HL, 2007, High-dose 106Ruthenium plaque brachytherapy for posterior uveal melanoma. A clinico-pathologic study. Ophthalmologe 104:149–157. , DOI 10.1007/s00347-006-1451-3 no authors listed, 1998, Histopathologic characteristics of uveal melanomas in eyes enucleated from the collaborative ocular melanoma study. COMS report no. 6. Am J Ophthalmol 125:745–766 Avery RB, Diener-West M, Reynolds SM, Grossniklaus HE, Green WR, AlbertDM(2008, Histopathologic characteristics of choroidal melanoma in eyes enucleated after iodine 125 brachytherapy in the collaborative ocular melanoma study. Arch Ophthalmol 126:207– 212. , DOI 10.1001/archopthalmol.2007.50 Shields CL, Shields JA, Karlsson U,Menduke H, Brady LW(1990, Enucleation after plaque radiotherapy for posterior uveal melanoma. Histopathologic findings. Ophthalmology 97:1665–1670 Brierley JD, Gospodarowicz MK, Wittekind C, eds,, 2017, 8th edition of the Union for International Cancer Control / American Joint Committee on Cancer TNM classification. Wiley, Oxford McLean IW, Foster WD, Zimmerman LE, Gamel JW, 1983, Modifications of Callender’s classification of uveal melanoma at the armed forces Institute of Pathology. Am J Ophthalmol 96:502–209 Gouvea AF, Santos Silva AR, Speight PM, Hunter K, Carlos R, Vargas PA, de Almeida OP, Lopes MA, 2013, High incidence of DNA ploidy abnormalities and increased Mcm2 expression may predict malignant change in oral proliferative verrucous leukoplakia. Histopathology 62:551–562. , DOI 10.1111/his.12036 Schilling H, Sehu KW, Lee WR, 1997, A histologic study, including DNA quantification and Ki-67 labeling index, in uveal melanomas after brachytherapy with ruthenium plaques. Invest Ophthalmol Vis Sci 38:2081–2092 Van den Bosch T, van Beek JG, Vaarwater J, VerdijkRM, Naus NC, Paridaens D, de Klein A, Kilic E, 2012, Higher percentage of FISH-determined monosomy 3 and 8q amplification in uveal melanoma cells relate to poor patient prognosis. Invest Ophthalmol Vis Sci 53:2668–2674. , DOI 10.1167/iovs.11-8697 Schneider B, Riedel K, Zhivov A, Huehns M, Zettl H, Guthoff RF, Junemann A, Erbersdobler A, Zimpfer A, 2017, Frequent and Yet Unreported GNAQ and GNA11 Mutations are found in Uveal Melanomas. Pathol Oncol Res. , DOI 10.1007/s12253-017-0371-7 Rand RW, Khonsary A, Brown WJ, Winter J, Snow HD, 1987, Leksell stereotactic radiosurgery in the treatment of eye melanoma. Neurol Res 9:142–146 Kang DW, Lee SC, Park YG, Chang JH, 2012, Long-term results of gamma knife surgery for uveal melanomas. J Neurosurg 117: 108–114 Crawford JB, Char DH, 1987, Histopathology of uveal melanomas treated with charged particle radiation. Ophthalmol 94:639–643 Gragoudas ES, Egan KM, Saornil MA, Walsh SM, Albert DM, Seddon JM, 1993, The time course of irradiation changes in proton beam-treated uveal melanomas. Ophthalmology 100:1555–1559 Singh AD, Eagle RC Jr, Shields CL, Shields JA, 2003, Clinicopathologic reports, case reports, and small case series: enucleation following transpupillary thermotherapy of choroidal melanoma: clinicopathologic correlations. Arch Ophthalmol 121:397–400 Fernandes BF, Weisbrod D, Yucel YH, Follwell M, Krema H, Heydarian M, Xu W, Payne D, McGowan H, Simpson ER, Laperriere N, Sahgal A, 2011, Neovascular glaucoma after stereotactic radiotherapy for juxtapapillary choroidal melanoma: histopathologic and dosimetric findings. Int J Radiat Oncol Biol Phys 80:377–384. , DOI 10.1016/j.ijrobp.2010.04.073 Song WK, Yang WI, Byeon SH, Koh HJ, Kwon OW, Lee SC, 2010, Clinicopathologic report of uveal melanoma with persistent exudative retinal detachment after gamma knife radiosurgery. Ophthalmologica 224:16–21. , DOI 10.1159/000233231 Chappell MC, Char DH, Cole TB, Harbour JW, Mishra K, Weinberg VK, Phillips TL, 2012, Uveal melanoma: molecular pattern, clinical features, and radiation response. Am J Ophthalmol 154:227–232. , DOI 10.1016/j.ajo.2012.02.022 Prescher G, Bornfeld N, Hirche H, Horsthemke B, Jockel KH, Becher R, 1996, Prognostic implications of monosomy 3 in uveal melanoma. Lancet 347:1222–1225 Coupland SE, Lake SL, Zeschnigk M, Damato BE, 2013, Molecular pathology of uveal melanoma. Eye 27:230–242. , DOI 10.1038/eye.2012.255 Seregard S, Lundell G, Lax I, af Trampe E, Kock E, 1997, Tumor cell proliferation after failed ruthenium plaque radiotherapy for posterior uveal melanoma. Acta Ophthalmol Scand 75:148–154 Chiquet C, Grange JD, Ayzac L, Chauvel P, Patricot LM, Devouassoux-Shisheboran M, 2000, Effects of proton beam irradiation on uveal melanomas: a comparative study of Ki-67 expression in irradiated versus non-irradiated melanomas. Br J Ophthalmol 84:98–102 Pe'er J, Stefani FH, Seregard S, Kivela T, Lommatzsch P, Prause JU, Sobottka B, Damato B, Chowers I, 2001, Cell proliferation activity in posterior uveal melanoma after Ru-106 brachytherapy: an EORTC ocular oncology group study. Br J Ophthalmol 85:1208–1212 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 731 EP 740 DI 10.1007/s12253-018-00565-1 PG 10 ER PT J TI Relationship Between -2028 C/T SELP Gene Polymorphism, Concentration of Plasma P-Selectin and Risk of Malnutrition in Head and Neck Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Malnutrition; Cachexia; Head and neck cancer; SELP; P-selectin ID Malnutrition; Cachexia; Head and neck cancer; SELP; P-selectin AB Until today there is a lack of molecular factors, that could predict either cancer malnutrition or cachexia. Among potential mechanisms, that contribute to development of above syndromes, the systemic inflammatory response with overproduction of cytokines and adhesion molecules is the most likely. Recent papers suggested crucial role of P-selectin adhesion molecule in the initiation of leukocytes recruitment to the site of injury during inflammation, promotion of tumor aggressiveness and contribution to cancer cachexia. The aim of the study was to investigate SELP -2028 C/T polymorphism as a risk factor of malnutrition in 66 head and neck cancer (HNC) patients subjected to radiotherapy. Genotyping was conducted by real-time PCR method by means of TaqMan SNP Genotyping Assay. P-selectin Human ELISA Kit was used to determine P-selectin concentration in each extracted plasma samples. CC homozygous subjects had 4-fold higher risk score of being qualified as severely malnourished compared to other genotype carriers (p = 0.015). However, the TT homozygous patients were at lowest risk of severe weight loss >10% during the therapy period (OR = 0.20; p = 0.019).We also noted, that CC genotype carriers had significantly higher risk of early death incidence compared to CT or TT genotype (median survival time: 29 vs 34 months; HR = 3.02; p = 0.0085). Studied SELP -2028 C/T seems to be a novel attractive predictive factor of cancer malnutrition in HNC patients, perhaps in a future, patients carrying unfavorable CC genotype could be earlier scheduled for pharmaceutical intervention with parenterall nutrition, therefore they could be prevented from the development of severe malnutrition or even cachexia. CR O’Neill J, Shaha A, 2011, Nutrition management of patients with malignancies of the head and neck. Surg Clin North Am 91:631–639 Alshadwi A, Nadershah M, Carlson ER, Young LS, Burke PA, Daley BJ, 2013, Nutritional considerations for head and neck cancer patients: a review of the literature. J Oral Maxillofac Surg 71(11):1853–1860 Unsal D, Mentes B, Akmansu M, Uner A, Oguz M, Pak Y, 2006, Evaluation of nutritional status in cancer patients receiving radiotherapy: a prospective study. Am J Clin Oncol 29:183–188 GorencM, Kozjek NR, Strojana P, 2015, Malnutrition and cachexia in patients with head and neck cancer treated with, chemo)radiotherapy. Rep Pract Oncol Radiother 20(4):249–258 Dhanapal R, Saraswathi TR, Govind RN, 2011, Cancer cachexia. J Oral Maxillofac Pathol 15(3):257–260 Aoyagi T, Terracina KP, Raza A, Matsubara H, Takabe K, 2015, Cancer cachexia, mechanism and treatment. World J Gastrointest Oncol 7(4):17–29 Donohoe CL, Ryan AM, Reynolds JV, 2011, 601434, Cancer Cachexia: mechanisms and clinical implications. Gastroenterol Res Pract 2011:1–13 Imhof BA, Dunon D, 1995, Leukocyte migration and adhesion. Adv Immunol 58:345–416 Geng JG, Chen M, Chou KC, 2004, P-selectin cell adhesion molecule in inflammation, thrombosis, cancer growth and metastasis. Curr Med Chem 11(16):2153–2160 Reiner AP, Carlson CS, Thyagarajan B, Rieder MJ, Polak JF, Siscovick DS, Nickerson DA, Jacobs DR Jr, Gross MD, 2008, Soluble P-selectin, SELP polymorphisms, and atherosclerotic risk in European-American and African-African young adults: the coronary artery risk development in young adults, CARDIA, study. Send to Arterioscler Thromb Vasc Biol 28(8):1549–1555 Burkhardt J, Blume M, Petit-Teixeira E, Hugo Teixeira V, Steiner A, Quente E, Wolfram G, Scholz M, Pierlot C, Migliorini P, Bombardieri S, Balsa A, Westhovens R, Barrera P, Radstake TRDJ, Alves H, Bardin T, Prum B, Emmrich F, Cornelis F, Ahnert P, Kirsten H, 2014, Cellular adhesion gene SELP is associated with rheumatoid arthritis and displays differential allelic expression. PLoS One 9(8):e103872 Fearon KC, Glass DJ, Guttridge DC, 2012, Cancer Cachexia: mediators, signaling, and metabolic pathways. Cell Metab 16(2):153–166 Avan A, Avan A, Le Large TYet al, 2014, AKT1 and SELP polymorphisms predict the risk of developing Cachexia in pancreatic Cancer patients. PLoS ONE 9(9):e108057 Kaur R, Singh J, KaurM(2017, Structural and functional impact of SNPs in P-selectin gene: a comprehensive in silico analysis. Open Life Sciences 12(1):19–33 Volcik KA, Ballantyne CM, Coresh J, Folsom AR, Boerwinkle E, 2007, Specific P-selectin and P-selectin glycoprotein ligand–1 genotypes/haplotypes are associated with risk of incident CHD and ischemic stroke: the atherosclerosis risk in communities, ARIC, study. Atherosclerosis 195:e76–e82 JacobinVM, Deramchia K, Mornet S et al, 2011)MRI of inducible P-selectin expression in human activated platelets involved in the early stages of atherosclerosis. NMR Biomed 24:413–424 Miller MA, Kerry SM, Dong Y, Strazzullo P, Cappuccio FP, 2004, Association between the Thr715Pro Pselectin gene polymorphism and soluble P-selectin levels in a multiethnic population in South London. Thromb Haemost 92:1060–1065 Volcik KA, Ballantyne CM, Coresh J, Folsom AR, Wu KK, Boerwinkle E, 2006, P-selectin Thr715Pro polymorphism predicts P-selectin levels but not risk of incident coronary heart disease or ischemic stroke in a cohort of 14595 participants: the atherosclerosis risk in communities study. Atherosclerosis 186:74–79 Kou L, Yang N, Chen G et al, 2016, Association of SELP genetic polymorphisms and additional gene-smoking interaction on cardiovascular disease in Chinese Han population. Int J Clin Exp Pathol 9(9):9612–9618 Morris DL, Graham RR, Erwig LP, Gaffney PM, Moser KL, Behrens TW, Vyse TJ, Graham DSC, 2009, Variation in the upstream region of P-selectin, SELP, is a risk factor for SLE. Genes Immun 10:404–413 Tan BH, Fladvad T, Braun TP et al, 2012, P-selectin genotype is associated with the development of cancer cachexia. EMBO Mol Med 4(6):462–471 Johns N, Stretch C, Tan BHL, 2017, New genetic signatures associated with cancer cachexia as defined by low skeletal muscle index and weight loss. J Cachexia Sarcopenia Muscle 8(1):122–130 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 741 EP 749 DI 10.1007/s12253-018-00578-w PG 9 ER PT J AU Aydin, AH Toptas, T Bozkurt, S Aydin, A Erdogan, G Pestereli, E Simsek, T AF Aydin, Ayik Hulya Toptas, Tayfun Bozkurt, Selen Aydin, Armagan Erdogan, Gulgun Pestereli, Elif Simsek, Tayup TI Stanniocalcin-2 May Be a Potentially Valuable Prognostic Marker in Endometrial Cancer: a Preliminary Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Stanniocalcin-2; Endometrial cancer; Prognostic factor; Recurrence ID Stanniocalcin-2; Endometrial cancer; Prognostic factor; Recurrence AB In the current study, we primarily aimed to investigate stanniocalcin-2 (STC2) protein expression pattern in hysterectomy specimens from patientswith endometrioid type endometrial cancer (EC) using immunohistochemistry. Secondly, in order to clarify its prognostic impact, we examined relationships of the expression levels of STC2 with clinicopathologic features and outcome of patients. Histopathology slides of 49 patients were stained with the monoclonal mouse antibody targeting STC2 protein. The expression levels of STC2 were classified based on three-tiered semiquantitative scheme: negative expression, expression level of 0; low-expression, expression level of 1+; and high-expression, expression levels of 2+ and 3+. Recurrence-free survival (RFS) was used as the primary prognostic outcome. Immunohistochemical analysis revealed that 73.5% of tissue samples exhibited positive staining with STC2. The intensity of staining with STC2 was weak in 40.8%, moderate in 22.4%, and strong in 10.2%. Thirty-eight percent of samples showed negative expression; 18.4%, low-expression (1+); and 42.8%, high-expression (2 to 3+). High-expression of STC2 was significantly associated with grade 2–3 tumors (p = 0.026) and disease recurrence (p = 0.013). Multivariate analysis revealed that both the tumor grade and STC2 were independent predictors of disease recurrence. Kaplan-Meier analyses confirmed that patients with highexpression of STC2 had a significantly poorer RFS than those with negative or low STC2 expression (p = 0.037); although overall survival did not differ with respect to expression levels of STC2 (p = 0.148). In conclusion, high-expression of STC2 is a negative prognostic factor, associated with increased risk of recurrence in endometrioid EC. C1 [Aydin, Ayik Hulya] Akdeniz University, School of Medicine, Department of Gynecologic Oncology, Dumlupinar Boulevard, H-Block, K:1, Konyaalti, 07059 Antalya, Turkey. [Toptas, Tayfun] Antalya Training and Research Hospital, Department of Gynecologic OncologyAntalya, Turkey. [Bozkurt, Selen] Akdeniz University, School of Medicine, Department of Biostatistics and Medical InformaticsAntalya, Turkey. [Aydin, Armagan] Antalya Research and Training Hospital, Antalya, Department of Medical OncologyAntalya, Turkey. [Erdogan, Gulgun] Akdeniz University, School of Medicine, Department of Gyneco-pathologyAntalya, Turkey. [Pestereli, Elif] Akdeniz University, School of Medicine, Department of Gyneco-pathologyAntalya, Turkey. [Simsek, Tayup] Akdeniz University, School of Medicine, Department of Gynecologic Oncology, Dumlupinar Boulevard, H-Block, K:1, Konyaalti, 07059 Antalya, Turkey. RP Aydin, AH (reprint author), Akdeniz University, School of Medicine, Department of Gynecologic Oncology, 07059 Antalya, Turkey. EM hulya_ayik@hotmail.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:E359–E386. , DOI 10.1002/ijc.29210 Ueda SM, Kapp DS, Cheung MK, Shin JY, Osann K, Husain A, Teng NN, Berek JS, Chan JK, 2008, Trends in demographic and clinical characteristics in women diagnosed with corpus cancer and their potential impact on the increasing number of deaths. Am J Obstet Gynecol 198:218.e1–218.e6. , DOI 10.1016/j.ajog. 2007.08.075 Creasman WT, Odicino F, Maisonneuve P, Quinn MA, Beller U, Benedet JL, Heintz AP, Ngan HY, Pecorelli S, 2006, Carcinoma of the corpus uteri. FIGO 26th annual report on the results of treatment in gynecological Cancer. Int J Gynaecol Obstet 95(Suppl 1):S105– S143 Banno K, Kisu I, Yanokura M, Masuda K, Ueki A, Kobayashi Y, Susumu N, Aoki D, 2012, Epigenetics and genetics in endometrial cancer: new carcinogenic mechanisms and relationship with clinical practice. Epigenomics 4:147–162. , DOI 10.2217/epi. 12.13 Chu SJ, Zhang J, Zhang R, Lu WW, Zhu JS, 2015, Evolution and functions of stanniocalcins in cancer. Int J Immunopathol Pharmacol 28:14–20. , DOI 10.1177/0394632015572745 Takei Y, Yamamoto H, Sato T, Otani A, Kozai M, Masuda M, Taketani Y, Muto-Sato K, Lanske B, Takeda E, 2012, Stanniocalcin 2 is associated with ectopic calcification in α- klotho mutant mice and inhibits hyperphosphatemia-induced calcification in aortic vascular smooth muscle cells. Bone 50:998–1005. , DOI 10.1016/j.bone.2012.01.006 Ziello JE, Jovin IS, Huang Y, 2007, Hypoxia-inducible factor, HIF)-1 regulatory pathway and its potential for therapeutic intervention in malignancy and ischemia. Yale J Biol Med 80:51–60 Kim PH, Na SS, Lee B, Kim JH, Cho JY, 2015, Stanniocalcin 2 enhances mesenchymal stem cell survival by suppressing oxidative stress. BMB Rep 48:702–707 Zhong H, Hanrahan C, van der Poel H, Simons JW, 2001, Hypoxia-inducible factor 1alpha and 1beta proteins share common signaling pathways in human prostate cancer cells. Biochem Biophys Res Commun 284:352–356 Zagzag D, Zhong H, Scalzitti JM, Laughner E, Simons JW, Semenza GL, 2000, Expression of hypoxia-inducible factor 1alpha in brain tumors: association with angiogenesis, invasion, and progression. Cancer 88:2606–2618 Arigami T, Uenosono Y, Ishigami S, Yanagita S, Hagihara T, Haraguchi N, Matsushita D, Hirahara T, Okumura H, Uchikado Y, Nakajo A, Hokita S, Natsugoe S, 2013, Clinical significance of stanniocalcin 2 expression as a predictor of tumor progression in gastric cancer. Oncol Rep 30:2838–2844. , DOI 10. 3892/or.2013.2775 Zhang ZH, Wu YG, Qin CK, Rong ZH, Su ZX, Xian GZ, 2014, Stanniocalcin 2 expression predicts poor prognosis of hepatocellular carcinoma. Oncol Lett 8:2160–2164 Shen XJ, Gu K, Shi JP, Yao JQ,Wu JC, 2014, Increased expression of stanniocalcin 2 is associated with tumor progression after radiotherapy in patients with cervical carcinoma. Int J Clin Exp Pathol 7: 8770–8776 Zhou H, Li YY, Zhang WQ, Lin D, ZhangWM, DongWD, 2014, Expression of stanniocalcin-1 and stanniocalcin-2 in laryngeal squamous cell carcinoma and correlations with clinical and pathological parameters. PLoS One 9:e95466. , DOI 10.1371/ journal.pone.0095466 Chen B, Zeng X, He Y, Wang X, Liang Z, Liu J, Zhang P, Zhu H, Xu N, Liang S, 2016, STC2 promotes the epithelial-mesenchymal transition of colorectal cancer cells through AKT-ERK signaling pathways. Oncotarget 7:71400–71416. , DOI 10.18632/ oncotarget.12147 Dondeti VR, Wubbenhorst B, Lal P, Gordan JD, D'Andrea K, Attiyeh EF, Simon MC, Nathanson KL, 2012, Integrative genomic analyses of sporadic clear cell renal cell carcinoma define disease subtypes and potential new therapeutic targets. Cancer Res 72:112– 121. , DOI 10.1158/0008-5472.CAN-11-1698 Law AY, Wong CK, 2010, Stanniocalcin-2 promotes epithelialmesenchymal transition and invasiveness in hypoxic human ovarian cancer cells. Exp Cell Res 316:3425–3434. , DOI 10. 1016/j.yexcr.2010.06.026 Yang S, Ji Q, Chang B, Wang Y, Zhu Y, Li D, Huang C, Wang Y, Sun G, Zhang L, Guan Q, Xiang J, Wei W, Lu Z, Liao T, Meng J, Wang Z, Ma B, Zhou L, Wang Y, Yang G, 2017, STC2 promotes head and neck squamous cell carcinoma metastasis through modulating the PI3K/AKT/snail signaling. Oncotarget 8:5976–5991. , DOI 10.18632/oncotarget.13355 Terakawa N, Kanamori Y, Yoshida S, 2003, Loss of PTEN expression followed by Akt phosphorylation is a poor prognostic factor for patients with endometrial cancer. Endocr Relat Cancer 10:203– 208 Cully M, You H, Levine AJ, Mak TW, 2006, Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat Rev Cancer 6:184–192 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 751 EP 757 DI 10.1007/s12253-018-00576-y PG 7 ER PT J AU da Silva Filho, FA Vieira-de-Mello, SG Santos, BdP de Melo Rego, JBM Ribeiro-Silva, A Beltrao, ICE AF da Silva Filho, Felix Antonio Vieira-de-Mello, Souto Gabriela Santos, Barros dos Petra de Melo Rego, Jesus Barreto Moacyr Ribeiro-Silva, Alfredo Beltrao, Isidoro Carneiro Eduardo TI N-Acetylglucosaminyltransferase III (GnT-III) but not N-Acetylgalactosaminyltransferase-6 and 8 are Differentially Expressed in Invasive and In Situ Ductal Carcinoma of the Breast SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glycosiltranferases; Glycocode; Lectins; In situ ductal carcinoma; Invasive ductal carcinoma ID Glycosiltranferases; Glycocode; Lectins; In situ ductal carcinoma; Invasive ductal carcinoma AB Mammary carcinoma is the most common malignant tumor in women, and it is the leading cause of mortality. In tumor context, glycosylation promotes post translational modifications necessary for cell progression, emerging as a relevant tumor hallmarker. This study aimed to analyze the association between polypeptide Nacetylgalactosaminyltransferase- 6 (ppGalNAc-T6), −T8, N-acetylglucosaminyltransferase III (GnT-III) expression, Phaseolus vulgaris-leucoagglutinin (PHA-L), wheat germ agglutinin (WGA) and peanut agglutinin (PNA) staining with clinic-histopathological factors from patients with pure ductal carcinoma in situ (DCIS) and DCIS with invasive ductal carcinoma (DCIS-IDC) of breast. Formalin-fixed and paraffin-embedded samples (n = 109) were analyzed. In pure DCIS samples GnT-III was over-expressed in comedo lesions (p = 0.007). In DCIS-IDC, GnT-III expression was associated with high nuclear grade tumors (p = 0.039) while the presence of PHA-L and WGA were inversely related to HER-2 expression (p = 0.001; p = 0.036, respectively). These findings pointed to possible involvement of GnT-III, ppGalNAc-T8, L-PHA and WGA as probes in prognostic evaluation of DCIS. C1 [da Silva Filho, Felix Antonio] Federal University of Pernambuco, Immunomodulation and New Therapeutic Approaches Laboratory, 50670-901 Recife, Pernambuco, Brazil. [Vieira-de-Mello, Souto Gabriela] Federal University of Pernambuco, Immunomodulation and New Therapeutic Approaches Laboratory, 50670-901 Recife, Pernambuco, Brazil. [Santos, Barros dos Petra] Federal University of Pernambuco, Biomarkers in cancer Laboratory, 50670-901 Recife, Pernambuco, Brazil. [de Melo Rego, Jesus Barreto Moacyr] Federal University of Pernambuco, Immunomodulation and New Therapeutic Approaches Laboratory, 50670-901 Recife, Pernambuco, Brazil. [Ribeiro-Silva, Alfredo] University of Sao Paulo, Faculty of Medicine of Ribeirao Preto, Department of Pathology, 14049-900 Ribeirao Preto, Sao Paulo, Brazil. [Beltrao, Isidoro Carneiro Eduardo] Federal University of Pernambuco, Biomarkers in cancer Laboratory, 50670-901 Recife, Pernambuco, Brazil. RP de Melo Rego, JBM (reprint author), Federal University of Pernambuco, Immunomodulation and New Therapeutic Approaches Laboratory, 50670-901 Recife, Brazil. EM moacyr.rego@gmail.com CR Strous GJ, Dekker J, 1992, Mucin-type glycoproteins. Crit Rev Biochem Mol Biol 27:57–92. , DOI 10.3109/ 10409239209082559 Triantafyllou A, Fletcher D, Scott J, 2004, Glycosylations in demilunar and central acinar cells of the submandibular salivary gland of ferret investigated by lectin histochemistry. Arch Oral Biol 49:697–703. , DOI 10.1016/j.archoralbio.2004.04. 005 Yang X, Liu S, Yan Q, 2013, Role of fucosyltransferase IV in epithelial–mesenchymal transition in breast cancer cells. Cell Death Dis 4:735. , DOI 10.1038/cddis.2013.241 Ohtsubo K,Marth JD, 2006, Glycosylation in cellular mechanisms of health and disease. Cell 126:855–867. , DOI 10.1016/j. cell.2006.08.019 Kariya Y, Gu J, 2011, N-glycosylation of ß4 integrin controls the adhesion and motility of keratinocytes. PLoS One 6:27084. https:// doi.org/10.1371/journal.pone.0027084 Kim Y, Park J, Lee H, Lee SY, Kim SJ, 2012, TGF-β sensitivity is determined by N-linked glycosylation of the type II TGF-β receptor. Biochem J 445:403–411. , DOI 10.1042/BJ20111923 Xu Q, Isaji T, Lu Y, GuW, Kondo M, Fukuda T, Du Y, Gu J, 2012, Roles of N-Acetylglucosaminyltransferase III in epithelial-tomesenchymal transition induced by transforming growth factor β1, TGF-β1, in epithelial cell lines. J Biol Chem 287:16563– 16574. , DOI 10.1074/jbc.M111.262154 Guo H, Johnson H, Randolph M, Lee I, Pierce M, 2009, Knockdown of GnT-Va expression inhibits ligand-induced downregulation of the epidermal growth factor receptor and intracellular signaling by inhibiting receptor endocytosis. Glycobiology 19: 547–559. , DOI 10.1093/glycob/cwp023 Lowe JB, Marth JD, 2003, A genetic approach to mammalian glycan function. Annu Rev Biochem 72:673–691. , DOI 1146/annurev.biochem.72.121801.161809 10. Gozde K, Attila G, Ozlem K, 2009, Human Cancer protein-protein interaction network: a structural perspective. PLoS Comput Biol 5: 1000601. , DOI 10.1371/journal.pcbi.1000601 Li M, Song L, Qin X, 2010, Glycan changes: cancer metastasis and anti-cancer vaccines. J Biosci 35:665–673. , DOI 10.1007/ s12038-010-0073-8 Victorino VJ, Campos FC, Herrera AC, Colado Simao AN, Cecchini AL, Panis C, Cecchini R, 2013, Overexpression of HER-2/neu protein attenuates the oxidative systemic profile in women diagnosed with breast cancer. Tumor Biol 35:3025–3034. , DOI 10.1007/s13277-013-1391-x Butt S, Harlid S, Borgquist S, IvarssonM, Landberg G, Dillner J-M, 2012, Genetic predisposition, parity, age at first childbirth and risk for breast cancer. BMC Res Notes 5:414. , DOI 10.1186/ 1756-0500-5-414 Yang XR, Chang-Claude J, Goode EL, Couch FJ, Nevanlinna H, Milne RL, Gaudet M, Schmidt MK, Broeks A, Cox A, Fasching PA, Hein R, Spurdle AB, Blows F, Driver K, Flesch-Janys D, Heinz J, Sinn P, Vrieling A, Heikkinen T, Aittomaki K, Heikkila P, Blomqvist C, Lissowska J, Peplonska B, Chanock S, Figueroa J, Brinton L, Hall P, Czene K, Humphreys K, Darabi H, Liu J, van ‘t Veer LJ, van Leeuwen FE, Andrulis IL, Glendon G, Knight JA, Mulligan AM, O’Malley FP, Weerasooriya N, John EM, Beckmann MW, Hartmann A, Weihbrecht SB, Wachter DL, Jud SM, Loehberg CR, Baglietto L, English DR, Giles GG, McLean CA, Severi G, Lambrechts D, Vandorpe T,Weltens C, Paridaens R, Smeets A, Neven P, Wildiers H, Wang X, Olson JE, Cafourek V, Fredericksen Z, KoselM, Vachon C, Cramp HE, Connley D, Cross SS, Balasubramanian SP, Reed MWR, Dork T, Bremer M, Meyer A, Karstens JH, Ay A, Park-Simon TW, Hillemanns P, Arias Perez JI, Rodriguez PM, Zamora P, Benitez J, Ko YD, Fischer HP, Hamann U, Pesch B, Bruning T, Justenhoven C, Brauch H, Eccles DM, Tapper WJ, Gerty SM, Sawyer EJ, Tomlinson IP, Jones A, Kerin M, Miller N, McInerney N, Anton-Culver H, Ziogas A, Shen CY, Hsiung CN, Wu PE, Yang SL, Yu JC, Chen ST, Hsu GC, Haiman CA, Henderson BE, le Marchand L, Kolonel LN, Lindblom A,Margolin S, Jakubowska A, Lubinski J, Huzarski T, Byrski T, Gorski B, Gronwald J, Hooning MJ, Hollestelle A, van den Ouweland AMW, Jager A, KriegeM, Tilanus-LinthorstMMA, Collee M, Wang-Gohrke S, Pylkas K, Jukkola-Vuorinen A, Mononen K, Grip M, Hirvikoski P, Winqvist R, Mannermaa A, Kosma VM, Kauppinen J, Kataja V, Auvinen P, Soini Y, Sironen R, Bojesen SE, Dynnes Orsted D, Kaur-Knudsen D, Flyger H, Nordestgaard BG, Holland H, Chenevix-Trench G, Manoukian S, Barile M, Radice P, Hankinson SE, Hunter DJ, Tamimi R, Sangrajrang S, Brennan P, McKay J, Odefrey F, Gaborieau V, Devilee P, Huijts PEA, Tollenaar RAEM, Seynaeve C, Dite GS, Apicella C, Hopper JL, Hammet F, Tsimiklis H, Smith LD, Southey MC, Humphreys MK, Easton D, Pharoah P, Sherman ME, Garcia-Closas M, 2011, Associations of breast cancer risk factors with tumor subtypes: a pooled analysis from the breast cancer association consortium studies. J Natl Cancer Inst 103:250–263. , DOI 10.1093/jnci/djq526 Wei Z, Er-li G, Yi-li Z, Zhai Q, Zou ZY, Guo GL, Chen GR, Zheng HM, Huang GL, Zhang XH, 2012, Different distribution of breast ductal carcinoma in situ, ductal carcinoma in situ with microinvasion, and invasion breast cancer. World J Surg Oncol 10:262. , DOI 10.1186/1477-7819-10-262 Liu Y, Perez M, Schootman M, Aft RL, Gillanders WE, Ellis MJ, Jeffe DB, 2010, A longitudinal study of factors associated with perceived risk of recurrence in women with ductal carcinoma in situ and early stage invasive breast cancer. Breast Cancer Res Treat 124:835–844. , DOI 10.1007/s10549-010-0912-1 Beaman E, Brooks SA, 2014, The extended ppGalNAc-T family and their functional involvement in the metastatic cascade. Histol Histopathol 29:293–304. , DOI 10.14670/HH-29.293 Pinho SS, Oliveira P, Cabral J, Carvalho S, Huntsman D, Gartner F, Seruca R, Reis CA, Oliveira C, 2012, Loss and recovery of Mgat3 and GnT-III mediated E-cadherin N-glycosylation is a mechanism involved in epithelial-mesenchymal-epithelial transitions. PLoS One 7:33191. , DOI 10.1371/journal.pone.0033191 Andrade CG, Cabral Filho PE, Tenorio DP, Santos BS, Beltrao EI, Fontes A, Carvalho LB Jr, 2013, Evaluation of glycophenotype in breast cancer by quantum dot-lectin histochemistry. Int J Nanomedicine 8:4623. , DOI 10.2147/IJN.S51065 Cazet A, Julien S, Bobowski M, Burchell J, Delannoy P, 2010, Tumour-associated carbohydrate antigens in breast cancer. Breast Cancer Res 12:204. , DOI 10.1186/bcr2577 Ribeiro-Silva A, Moutinho MAH, Moura B, Vale FR, Zucoloto S, 2006, Expression of checkpoint kinase 2 in breast carcinomas: correlation with key regulators of tumor cell proliferation, angiogenesis, and survival. Histol Histopathol 21:373-82. , DOI 10.14670/HH-21.373 Santos PB, Zanetti JS, Silva AR, Beltrao EIC, 2012, Beta 1 integrin predicts survival in breast cancer: a clinicopathological and immunohistochemical study. Diagn Pathol 7:104. , DOI 10. 1186/1746-1596-7-104 Hammond ME, Hayes DF, Dowsett M, 2010, American society of clinical oncology/college of American pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol 28:2784–95. https://www.ncbi.nlm.nih.gov/pubmed/16437383 Wolff AC, Hammond ME, Schwartz JN, 2007, American society of clinical oncology/college of American pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 131:18–43. https:// doi.org/10.1043/15432165(2007)131[18:ASOCCO]2.0.CO;2 Oliveira-Costa JP, Zanetti JS, Oliveira LR, Soares FA, Ramalho LZ, Silva Ramalho F, Garcia SB, Ribeiro-Silva A, 2010, Significance of topoisomerase IIIβ expression in breast ductal carcinomas: strong associations with disease-specific survival and metastasis. Hum Pathol 41:1624–1630. , DOI 10.1016/j.humpath. 2010.01.027 Dosaka-Akita H, Miyoshi E, Suzuki O, Itoh T, Katoh H, Taniguchi N, 2004, Expression of N-acetylglucosaminyltransferase Vis associated with prognosis and histology in non-small cell lung cancers. Clin Cancer Res 10:1773–1779. , DOI 10.1158/1078- 0432.CCR-1047-3 Kitada S, Yamada S, Kuma A, Ouchi S, Tasaki T, Nabeshima A, Noguchi H,Wang KY, Shimajiri S, Nakano R, Izumi H, Kohno K, M a t s u m o t o T, Sa s a g u r i Y, 2 0 1 3 , Po l y p e p t i d e nacetylgalactosaminyl transferase 3 independently predicts highgrade tumours and poor prognosis in patients with renal cell carcinomas. Br J Cancer 23:472–481. , DOI 10.1038/bjc.2013. 331 Christiansen MN, Chik J, Lee L, Abrahams JL, Packer NH, 2014, Cell surface protein glycosylation in cancer. Proteomics 14:525– 546. , DOI 10.1002/pmic.201300387 Hu M, Yao J, Carroll DK, Weremowicz S, Chen H, Carrasco D, Richardson A, Violette S, Nikolskaya T, Nikolsky Y, Bauerlein EL, Hahn WC, Gelman RS, Allred C, Bissell MJ, Schnitt S, Polyak K, 2008, Regulation of in situ to invasive breast carcinoma transition. Cancer Cell 13:394–406. , DOI 10.1016/j.ccr.2008.03.007 Lange T, Samatov TR, Tonevitsky AG, Schumacher U, 2014, Importance of altered glycoprotein-bound N- and O-glycans for epithelial-to-mesenchymal transition and adhesion of cancer cells. Carbohydr Res 389:39–45. , DOI 10.1016/j.carres.2014. 01.010 Akama R, Sato Y, Kariya Y, Isaji T, Fukuda T, Lu L, Taniguchi N, Ozawa M, Gu J, 2008, N-acetylglucosaminyltransferase III expression is regulated by cell-cell adhesion via the E-cadherin-cateninactin complex. Proteomics 8:83221–83228. , DOI 10. 1002/pmic.200800038 Pinho SS, Reis CA, Paredes J, Magalhaes AM, Ferreira AC, Figueiredo J, Xiaogang W, Carneiro F, Gartner F, Seruca R, 2009, The role of N-acetylglucosaminyltransferase III and V in the post-transcriptional modifications of E-cadherin. Hum Mol Genet 18:2599–2608. , DOI 10.1093/hmg/ddp194 Pinho SS, Figueiredo J, Cabral J, Carvalho S, Dourado J, Magalhaes A, Gartner F, Mendonfa AM, Isaji T, Gu J, Carneiro F, Seruca R, Taniguchi N, Reis CA, 2013, E-cadherin and adherens-junctions stability in gastric carcinoma: functional implications of glycosyltransferases involving N-glycan branching biosynthesis, N-acetylglucosaminyltransferases III and V. Biochim Biophys Acta 1830:2690–2700. , DOI 10.1016/j.bbagen. 2012.10.021 Livasy CA, Karaca G, Nanda R, Tretiakova MS, Olopade OI, Moore DT, Perou CM, 2006, Phenotypic evaluation of the basallike subtype of invasive breast carcinoma.Mod Pathol 19:264–271. , DOI 10.1038/modpathol.3800528 Ozkan-Gurdal S, Cabioglu N, Ozcinar B, Muslumanoglu M, Ozmen V, Kecer M, Yavuz E, Igci A, 2014, Factors predicting microinvasion in ductal carcinoma in situ. Asian Pac J Cancer Prev 15:55–60 https://www.ncbi.nlm.nih.gov/pubmed/24528005 Shekhar MP, Tait L, Pauley RJ, Wu GS, Santner SJ, Nangia- Makker P, Shekhar V, Nassar H, Visscher DW, Heppner GH, Miller FR, 1991, Comedo-ductal carcinoma in situ: a paradoxical role for programmed cell death. Cancer Biol Cancer Res Treat 18: 11–17 https://www.ncbi.nlm.nih.gov/pubmed/18787417 Vieira-de-Mello GS, Silva Filho AF, dos Santos PB, Rego MJBM, Beltrao EIC, 2013, Lectin histochemistry reveals changes in carbohydrate expression on morphological types of breast ductal carcinoma in situ. J Cytol Histol 4:3. , DOI 10.4172/ 2157-7099.1000179 Duggal S, Robin J, Julian TB, 2013, Ductal carcinoma in situ: an overview. Expert Rev Anticancer Ther 13:955–962. , DOI 10.1586/14737140.2013.820557 Freire T, Berois N, Sonora C, Varangot M, Barrios E, Osinaga E, 2006, UDP-N-acetyl-D-galactosamine:polypeptide Nacetylgalactosaminyltransferase 6, ppGalNAc-T6, mRNA as a potential new marker for detection of bone marrow-disseminated breast cancer cells. Int J Cancer 119:1383–1388. , DOI 10.1002/ijc.21959 Raman J, Guan Y, Perrine CL, Gerken TA, Tabak LA, 2012, UDP-N-acetyl-α-D-galactosamine: polypeptide Nacetylgalactosaminyltransferases: completion of the family tree. Glycobiology 22:768–777. , DOI 10.1093/ glycob/cwr183 Wu C, Guo X,WangW,Wang Y, Shan Y, Zhang B, SongW,Ma S, Ge J, Deng H, Zhu M, 2010, N-Acetylgalactosaminyltransferase- 14 as a potential biomarker for breast cancer by immunohistochemistry. BMC Cancer 10:123. , DOI 10.1186/1471-2407-10- 123 Steven A, Leisz S, Massa C, Iezzi M, Lattanzio R, Lamolinara A, Bukur J,Muller A, Hiebl B,Holzhausen HJ, Seliger B, 2013)HER- 2/neu mediates oncogenic transformation via altered CREB expression and function. Mol Cancer Res 11:1462–1477. , DOI 10.1158/1541-7786.MCR-13-0125 Zhou BP, Liao Y, Xia W, Spohn B, Lee MH, Hung MC, 2001, Cytoplasmic localization of p21Cip1/WAF1 by Akt-induced phosphorylation in HER-2/neu overexpressing cells. Nat Cell Biol 3: 245–252. , DOI 10.1038/35060032 Aranda V, Haire T, Nolan ME, Calarco JP, Rosenberg AZ, Fawcett JP, Pawson T, Muthuswamy SK, 2006, Par6–aPKC uncouples ErbB2 induced disruption of polarized epithelial organization from proliferation control. Nat Cell Biol 8:1235–1245. , DOI 10. 1038/ncb1485 Handerson T, Camp R, Harigopal M, Rimm D, Pawelek J, 2005, β1, 6-branched oligosaccharides are increased in lymph node metastases and predict poor outcome in breast carcinoma. Clin Cancer Res 11:2969–2973. , DOI 10.1158/1078-0432 Li D, Li Y,Wu X, Li Q, Yu J, Gen J, Zhang XL, 2008, Knockdown of Mgat5 inhibits breast cancer cell growth with activation of CD4+ T cells and macrophages. J Immunol 180:3158–3165. https://doi. org/10.4049/jimmunol.180.5.3158 ChungM, Chang HR, Bland KI,Wanebo HJ, 1996, Younger women with breast carcinoma have a poorer prognosis than older women. Cancer 77:97–103. , DOI 10.1002/(SICI)1097- 0142(19960101)77:1<97::AID-CNCR16>3.0.CO;2-3 Shannona C, Smithb IE, 2003, Breast cancer in adolescents and young women. Eur J Cancer 39:2632–2642. , DOI 10. 1016/S0959-8049(03)00669-5 Rego MJBM, Vieira de Mello GS, da Silva Santos CA, Chammas R, Beltrao EIC, 2013, Implications on glycobiological aspects of tumor hypoxia in breast ductal carcinoma in situ. Med Mol Morphol 46:92–96. , DOI 10.1007/s00795-013-0013-4 Abbott KL, Aoki K, Lim JM, Porterfield M, Johnson R, O'Regan RM, Wells L, Tiemeyer M, Pierce M, 2008, Targeted glycoproteomic identification of biomarkers for human breast carcinoma. J Proteome Res 7:1470–1480. , DOI 10.1021/ pr700792g NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 759 EP 768 DI 10.1007/s12253-019-00593-5 PG 10 ER PT J AU Jin, J Guo, Q Xie, J Jin, D Zhu, Y AF Jin, Jianying Guo, Qunyi Xie, Jingjing Jin, Dan Zhu, Yanan TI Combination of MEK Inhibitor and the JAK2-STAT3 Pathway Inhibition for the Therapy of Colon Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MEK inhibitors; Colon cancer; JAK2; STAT3 ID MEK inhibitors; Colon cancer; JAK2; STAT3 AB The study aimed to investigate the reason of HCT116 cell resistance to MEK inhibitor, and the combination treatment effects of MEK inhibitor AZD6244 and JAK2/STAT3 inhibitor AG490 on colon cancer in vitro and in vivo, including cell viability, apoptosis, and explore the partial mechanisms focused on AZD6244 promoted the activation of JAK2-STAT3 pathways. In vitro, we examined the HCT116 cell viability by CCK8, cell apoptosis by flow cytometry;Western blot measured p-ERK, p-JAK2, p- STAT3 and STAT3 expression. In vivo, nude mice were subcutaneously injected by HCT116 cells. The tumor volume and weight were detected. HCT116 cell resistance to MEK inhibitor AZD6244, which inhibited the activation of ERK and promoted the activation of JAK2-STAT3 signaling. The combination treatment of AZD6244 and AG490 significantly inhibited cell viability and induced cell apoptosis, and completely inhibited the activation of ERK and JAK2-STAT3 signaling. Combination treatment of AZD6244 and AG490 had a stronger effect than that of AZD6244 as a monotherapy in vitro and in vivo. The treatment of AZD6244 on K-Ras mutations HCT116 cells promoted the activation of JAK2/STAT3 signaling. JAK2/STAT3 inhibitor AG490 synergistically increases effects of AZD6244 on colon cancer in vitro and in vivo. Collectively, these results provide a rationale for combining inhibitors of the JAK/STAT pathway and MEK inhibitors to reduce the potential impact of drug resistance. C1 [Jin, Jianying] Taizhou Hospital, Department of Blood Oncology, 318000 Taizhou, China. [Guo, Qunyi] Taizhou Hospital, Department of Blood Oncology, 318000 Taizhou, China. [Xie, Jingjing] Taizhou Hospital, Department of Blood Oncology, 318000 Taizhou, China. [Jin, Dan] Taizhou Hospital, Department of Blood Oncology, 318000 Taizhou, China. [Zhu, Yanan] Emergency Center of Taizhou Hospital, 318000 Taizhou, China. RP Zhu, Y (reprint author), Emergency Center of Taizhou Hospital, 318000 Taizhou, China. EM 13736248866@163.com CR Zhang W, Tong D, Liu F, Li DW, Li JJ et al, 2016, RPS7 inhibits colorectal cancer growth via decreasing HIF-1α-mediated glycolysis. Oncotarget 7:5800–5814 [PubMed: 26735579] Lin J, Webber EM, Senger CA, Holmes RS, Whitlock EP, 2011, Systematic review of pharmacogenetic testing for predicting clinical benefit to anti-EGFR therapy in metastatic colorectal cancer. Am J Cancer Res 1:650–662 [PubMed: 21779535] Grady WM, Pritchard CC, 2014, Molecular alterations and biomarkers in colorectal cancer. Toxicol Pathol 42:124–139 [PubMed: 24178577] Vaughn CP, ZoBell SD, Furtado LV, Baker CL, Samowitz WS, 2011, Frequency of KRAS, BRAF, and NRAS mutations in colorectal Cancer. Gene Chromosome Cancer 50:307–312 [PubMed: 21305640] Pollock CB, Shirasawa S, Sasazuki T, KolchW, Dhillon AS, 2005, Oncogenic K-RAS is required to maintain changes in cytoskeletal organization, adhesion, and motility in colon cancer cells. Cancer Res 65:1244–1250 [PubMed: 15735008] Van Der Hoeven D, Cho KJ, Ma X, Chigurupati S, Parton RG, Hancock JF, 2013, Fendiline inhibits K-Ras plasma membrane localization and blocks K-Ras signal transmission. Mol Cell Biol 33:237–251 [PubMed: 23129805] Surade S, Blundell TL, 2012, Structural biology and drug discovery of difficult targets: the limits of ligandability. Chem Biol 19:42– 50 [PubMed: 22284353] Chan DA, Giaccia AJ, 2011, Harnessing synthetic lethal interactions in anticancer drug discovery. Nat Rev Drug Discov 10:351– 364 [PubMed: 21532565] Corcoran RB, Contino G, Deshpande V, Tzatsos A, Conrad C, Benes CH, Levy DE, Settleman J, Engelman JA, Bardeesy N, 2011, STAT3 plays a critical role in KRAS-induced pancreatic tumorigenesis. Cancer Res 71:5020–5029 [PubMed: 21586612] Abrams SL, Ruvolo PP, Ruvolo VR, Ligresti G, Martelli AM, Cocco L, Ratti S, Tafuri A, Steelman LS, Candido S, Libra M, McCubrey J, 2017, Targeting signaling and apoptotic pathways involved in chemotherapeutic drug-resistance of hematopoietic cells. Oncotarget 8(44):76525–76557 [PubMed: 29100331] Kim DJ, Lee MH, Reddy K, Li Y, Lim DY, Xie H, Lee SY, Yeom YI, Bode AM, Dong Z, 2013, CInQ-03, a novel allosteric MEK inhibitor, suppresses cancer growth in vitro and in vivo. Carcinogenesis 34:1134–1143 [PubMed: 23354306] Jing J, Greshock J, Holbrook JD, Gilmartin A, Zhang X,McNeil E, Conway T, Moy C, Laquerre S, Bachman K, Wooster R, Degenhardt Y, 2012, Comprehensive predictive biomarker analysis forMEK inhibitor GSK1120212. Mol Cancer Ther 11:720–729 [PubMed: 22169769] Jessen WJ, Miller SJ, Jousma E, Wu JQ, Rizvi TA, Brundage ME, Eaves D, Widemann B, Kim MO, Dombi E, Sabo J, Hardiman Dudley A, Niwa-Kawakita M, Page GP, Giovannini M, Aronow BJ, Cripe TP, Ratner N, 2013, MEK inhibition exhibits efficacy in human and mouse neurofibromatosis tumors. J Clin Invest 123: 340–347 [PubMed: 23221341] Carlino MS, Todd JR, Gowrishankar K, Mijatov B, Pupo GM, Fung C, Snoyman S, Hersey P, Long GV, Kefford RF, Rizos H, 2014, Differential activity of MEK and ERK inhibitors in BRAF inhibitor resistant melanoma. Mol Oncol 8:544–554 [PubMed: 24476679] Chen HJ, Yang ZD, Ding CY, Chu LL, Zhang YY, Terry K, Liu H, Shen Q, Zhou J, 2013, Fragment-based drug design and identification of HJC0123, a novel orally bioavailable STAT3 inhibitor for cancer therapy. Eur J Med Chem 62: 498–507 [PubMed: 23416191] Yeh TC, Marsh V, Bernat BA, Ballard J, Colwell H, Evans RJ, Parry J, Smith D, Brandhuber BJ, Gross S, Marlow A, Hurley B, Lyssikatos J, Lee PA, Winkler JD, Koch K, Wallace E, 2007, Biological characterization of ARRY-142886, AZD6244), a potent highly selectivemitogen-actived protein kinase kinase 1/2 inhibitor. Clin Cancer Res 13:1576–1583 [PubMed: 17332304] Zhou BY, Der CJ, Cox AD, 2016, The role of wild type RAS isoforms in cancer. Semin Cell Dev Biol 58:60–69 [PubMed: 27422332] Haigis KM, Kendall KR, Wang YF, Cheung A, Haigis MC, Glickman JN, Niwa-Kawakita M, Sweet-Cordero A, Sebolt- Leopold J, Shannon KM, Settleman J, Giovannini M, Jacks T, 2008, Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat Genet 40:600–608 [PubMed: 18372904] Caunt CJ, Sale MJ, Smith PD, Cook SJ, 2015, MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road. Nat Rev Cancer 15:577–592 [PubMed: 26399658] Zhao Y, Adjei AA, 2014, The clinical development ofMEK inhibitors. Nat Rev Clin Oncol 11:385–400 [PubMed: 24840079] Yeh JJ, Routh ED, Rubinas T, Peacock J, Martin TD, Shen XJ, Sandler RS, Kim HJ, Keku TO, der CJ, 2009, KRAS/BRAF mutation status and ERK1/2 activation as biomarkers for MEK1/2 inhibitor therapy in colorectal cancer. Mol Cancer Ther 8:834– 843 [PubMed: 19372556] Nagaria TS, Shi C, Leduc C, Hoskin V, Sikdar S et al, 2017, Combined targeting of Raf and Mek synergistically inhibits tumorigenesis in triple negative breast cancer model systems. Oncotarget 8:80804–80819 [PubMed: 29113345] Hur EH, Goo BK, Moon J, Choi Y, Hwang JJ, Kim CS, Bae KS, Choi J, Cho SY, Yang SH, Seo J, Lee G, Lee JH, 2017, Induction of immunoglobulin transcription factor 2 and resistance to MEK inhibitor in melanoma cells. Oncotarget 8:41387–41400 [PubMed: 28574827] Ulivi P, Arienti C, Amadori D, Fabbri F, Carloni S, Tesei A, Vannini I, Silvestrini R, Zoli W, 2009, Role of RAF/MEK/ ERK pathway, p-STAT-3 and Mcl-1 in sorafenib activity in human pancreatic cancer cell lines. J Cell Physiol 220:214– 221 [PubMed: 19288493] Tian F, Yang X, Liu Y, Yuan X, Fan T, Zhang F, Zhao J, Lu J, Jiang Y, Dong Z, Yang Y, 2017, Constitutive activated STAT3 is an essential regulator and therapeutic target in esophageal squamous cell carcinoma. Oncotarget 8:88719– 88729 [PubMed: 29179470] Mohanty SK, Yagiz K, Pradhan D, Luthringer DJ, Amin MB, Alkan S, Cinar B, 2017, STAT3 and STAT5A are potential therapeutic targets in castration-resistant prostate cancer. Oncotarget 8: 85997–86010 PubMed: 29156772] Holderfield M, Deuker MM, McCormick F, McMahon M, 2014, Targeting RAF kinases for cancer therapy: BRAF-mutated melanoma and beyond. Nat Rev Cancer 14:455–467. [PubMed: 24957944] NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 769 EP 775 DI 10.1007/s12253-019-00592-6 PG 7 ER PT J AU Kar, S Majumder, DD AF Kar, Subrata Majumder, Dutta Dwijesh TI A Novel Approach of Mathematical Theory of Shape and Neuro-Fuzzy Based Diagnostic Analysis of Cervical Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; Pap smear images segmentation; Features extraction; Shape theory; Neuro-fuzzy classification system ID Cervical cancer; Pap smear images segmentation; Features extraction; Shape theory; Neuro-fuzzy classification system AB This study aims to detect the abnormal growth of tissue in cervix region for diagnosis of cervical cancer using Pap test of patients. The proposed methodology classifies cervical cancer for pattern recognition either benign or malignant stages using shape and neuro-fuzzy based diagnostic model. In this experiment, firstly the authors segment Pap smear images of cervical cells using fuzzy c-means clustering algorithm and shape theory to classify them according to the presence of abnormality of the cells. Secondly the features extraction process is performed in the part of nucleus and cytoplasm on the squamous and glandular cells and the authors used input variables such as cytoplasm area (CA), cytoplasm circularity (CC), nucleus area (NA), nucleus circularity (NC), nucleus-cytoplasm ratio (NCR), and maximum nucleus brightness (MNB) in fuzzy tools and used fuzzy rules to evaluate the cervical cancer risk status as an output variable. The proposed neuro-fuzzy network system was developed for early detection of cervical cancer. A neural network was trained with 15-Pap image datasets where Levenberg–Marquardt(LM) a feed-forward back-propagation algorithmwas used to get the status of the cervical cancer. Out of 15 samples database, 11 data set for training, 2 data set for validation and 2 data set for test were used in the ANN classification system. The presented fuzzy expert system(FES) successfully identified the presence of cervical cancer in the Pap smear images using the extracted features and the use of neuro-fuzzy system(NFS) for the identification of cervical cancer at the early stages and achieve a satisfactory performance with 100% accuracy. C1 [Kar, Subrata] Dumkal Institute of Engineering & Technology, Department of Mathematics, 742406 Murshidabad, West Bengal, India. [Majumder, Dutta Dwijesh] Indian Statistical Institute, Department of Electronics and Communication Sciences Unit, 700108 Kolkata, West Bengal, India. RP Kar, S (reprint author), Dumkal Institute of Engineering & Technology, Department of Mathematics, 742406 Murshidabad, India. EM subrata.bacchu@gmail.com CR Jantzen J, Dounias, G, 2006, Analysis of PAP smear image data. Proc. ofNature Inspired Smart Information System,NISIS 2006, pp 1–10 Majumder DD, Bhattacharya M, 1998a, A new shape based technique for classification and registration: application to multimodal medical images. Int J of Image Processing and Communication 4(3–4):45–70 Sreedevi MT, Usha BS, Sandya S, 2012, Pap smear image based detection of cervical cancer. Int J Comput Appl 45(20):0975–8887 Shah Sunny S et al, 2016, Current technologies and recent developments for screening of HPV-associated cervical and orpharyngeal cancers. Cancers 8(85):1–27 Banarjee DK, Parui SK, Majumder DD, 1994, A shape metric for 3-D objects. Indian Journal Pure Appl Math 25(1&2):95–111 Majumder DD, 2015, Physics, chemistry and biology of nanoscale bio-technology-need for a unified framework for nanoarchitectures, DNA structures and nano-computing. EKHOMANJARI: a magazine on Science and Culture, BKC College, Kolkata, pp 13–32 Parui SK,Majumder DD, 1982, A new definition of shape similarity. Pattern Recogn Lett 1:87–120 Plissiti ME, Nikou C, Charchanti A, 2010, Automated detection of cell nuclei in pap smear images using morphological reconstruction and clustering. IEEE Trans Inf Technol Biomed 15(2):233–241 Genctav A, Aksoy S, Onde S, 2012, Unsupervised segmentation and classification of cervical cell images. Pattern Recogn 45:4151– 4168 Leica Microsystems, http://www.leica-microsystems.com/ Mahanta LB, Nath CK, Karan S, Nath DC, Majumder DD, Sharma JD, 2011, Fuzzy mathematical and shape theoretic approach to cervical cell classification. Int J Comput Appl, 0975–8887), 36,, 6):1–5 Gallegos-Funes FJ, Gomez-Mayora ME, Lopez-Bonilla JL, Cruz- Santiago R, 2009, Rank M- type radial basis function, RMRBF, neural network for pap smear microscopic image classification. Apeiron 16(4):542–554 Rosidi B, Jalil N, Pista NM, Ismail LH, Mengko ETL, 2011, Classification of cervical cells based on labeled colour intensity distribution. International Journal of Biology and Biomedical Engineering 5(4):230–238 Talukdar J, Nath CK, Talukdar PH, 2013, Fuzzy clustering based image segmentation of pap smear images of cervical cancer cell using FCM algorithm. Int. Journal of Engineering and Innovative Technology(IJEIT, 3(1):460–462 Plissiti ME, Nikou C, Charchant A, 2011, Combining shape, texture and intensity features for cell nuclei extraction in pap smear images. Pattern Recogn Lett 32(6):838–853 Majumder DD, 1995, A study on a mathematical theory of shapes in relation to pattern recognition and computer vision. Indian Journal of Theoretical Physics 43(4):19–30 Kar S, Majumder DD, 2017, A mathematical theory of shape and neuro-fuzzy methodology- based diagnostic analysis: a comparative study on early detection and treatment planning of brain cancer. Int J Clin Oncol 22(4):667–681 Byriel J, 1999, Neuro-fuzzy classification of cells in Cervical smears, M.Sc Thesis, Technical University of Denmark, 1999 Majumder DD, Majumdar KK, 2004, Complexity analysis, uncertainty management, and fuzzy dynamical systems: a cybernetic approach. Kybernetes 33(7):1143–1184 Majumder DD, Bhattacharya M, 2000, Cybernetic approach to medical technology: application to cancer screening and other diagnostics. Kybernetes 29:871–895 Wen C, Ming-Yu L, Horng-Jyh H, Yen-Chem H, Chien-Hung C, Kun-His T, Chi-Hung H, 2009, Automatic segmentation of abnormal cell nuclei from microscopic image analysis for cervical cancer screening. 2009 IEEE 3rd International Conference on Nano/ Molecular Medicine and Engineering, tainan, 18–21 Oct 2009 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 777 EP 790 DI 10.1007/s12253-019-00582-8 PG 14 ER PT J AU Tan, L Wu, Y Ma, X Yan, Y Shao, Sh Liu, J Ma, H Liu, R Chai, L Ren, J AF Tan, Li Wu, Yinying Ma, Xiaowei Yan, Yanli Shao, Shuai Liu, Jiaxin Ma, Hailin Liu, Rui Chai, Linyan Ren, Juan TI A Comprehensive Meta-Analysis of Association between EGFR Mutation Status and Brain Metastases in NSCLC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Epidermal growth factor receptor; NSCLC; Brain metastases; Meta-analysis ID Epidermal growth factor receptor; NSCLC; Brain metastases; Meta-analysis AB Non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutation have different clinicopathological characteristics compared with EGFR wild type NSCLC. A growing number of studies focused on the relevance between EGFR mutation status and brain metastases (BM) in NSCLC, but it remains controversial. Therefore, this study performed a comprehensive meta-analysis to untangle this issue. Several electronic databases including Pubmed, Embase, Web of science and Cochrane database were thoroughly searched. The odds ratio (OR) with 95% confidence interval (95% CI) was pooled to evaluate the relevance. Meta-regression analysis and subgroup analysis were conducted according to the heterogeneity. A total of 26 studies were identified finally in this meta-analysis. The overall OR was 1.58 (95% CI: 1.36–1.84), which indicated that EGFR mutation had a positive association with BM of NSCLC. The subgroup analysis resulted from eleven studies with lung adenocarcinoma revealed a higher possibility of BM in NSCLC with EGFR mutation compared with EGFR wild (p < 0.05). There was no significant difference in the risk of BM between NSCLC EGFR exon 19 mutation and exon 21 point mutation (p = 0.23). This meta-analysis suggests that EGFR mutation can be a risk factor for BM in NSCLC. C1 [Tan, Li] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi, China. [Wu, Yinying] First Affiliated Hospital of Xi’an Jiaotong University, Department of Medical Oncology, 710061 Xi’an, Shaanxi, China. [Ma, Xiaowei] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi, China. [Yan, Yanli] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi, China. [Shao, Shuai] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi, China. [Liu, Jiaxin] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi, China. [Ma, Hailin] First Affliated Hospital, Xi’an Jiaotong University, Department of Oncology Radiotherapy, 710061 Xi’an, Shaanxi, China. [Liu, Rui] First Affliated Hospital, Xi’an Jiaotong University, Department of Oncology Radiotherapy, 710061 Xi’an, Shaanxi, China. [Chai, Linyan] First Affliated Hospital, Xi’an Jiaotong University, Department of Oncology Radiotherapy, 710061 Xi’an, Shaanxi, China. [Ren, Juan] First Affliated Hospital, Xi’an Jiaotong University, Department of Oncology Radiotherapy, 710061 Xi’an, Shaanxi, China. RP Ren, J (reprint author), First Affliated Hospital, Xi’an Jiaotong University, Department of Oncology Radiotherapy, 710061 Xi’an, China. EM 869491533@qq.com CR Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(1):7–30. , DOI 10.3322/caac.21387 Barnholtz-Sloan JS, Sloan AE, Davis FG, Vigneau FD, Lai P, Sawaya RE, 2004, Incidence proportions of brain metastases in patients diagnosed, 1973 to 2001, in the metropolitan Detroit Cancer surveillance system. J Clin Oncol 22(14):2865–2872. , DOI 10.1200/jco.2004.12.149 Mujoomdar A, Austin JHM, Malhotra R, Powell CA, Pearson GDN, ShiauMC, Raftopoulos H, 2007, Clinical predictors of metastatic disease to the brain from non–small cell lung carcinoma: primary tumor size, cell type, and lymph node Metastases1. Radiology 242(3):882–888 Han G, Bi J, Tan W,Wei X,Wang X, Ying X, Guo X, Zhou X, Hu D, ZhenW(2016, A retrospective analysis in patients with EGFRmutant lung adenocarcinoma: is EGFR mutation associated with a higher incidence of brain metastasis? Oncotarget 7(35):56998– 57010. , DOI 10.18632/oncotarget.10933 Langer CJ, Mehta MP, 2005, Current management of brain metastases, with a focus on systemic options. J Clin Oncol 23(25):6207– 6219. , DOI 10.1200/jco.2005.03.145 Welsh JW, Komaki R, Amini A, Munsell MF, Unger W, Allen PK, Chang JY,Wefel JS, McGovern SL, Garland LL, Chen SS,Holt J, Liao Z, Brown P, Sulman E, Heymach JV, Kim ES, Stea B, 2013, Phase II trial of erlotinib plus concurrent whole-brain radiation therapy for patients with brain metastases from non-small-cell lung cancer. J Clin Oncol 31(7):895–902. , DOI 10.1200/jco.2011.40.1174 Park SJ, Kim HT, Lee DH, KimKP, KimSW, Suh C, Lee JS, 2012, Efficacy of epidermal growth factor receptor tyrosine kinase inhibitors for brain metastasis in non-small cell lung cancer patients harboring either exon 19 or 21 mutation. Lung Cancer 77(3):556– 560. , DOI 10.1016/j.lungcan.2012.05.092 Yatabe Y, Takahashi T, Mitsudomi T, 2008, Epidermal growth factor receptor gene amplification is acquired in association with tumor progression of EGFR-mutated lung cancer. Cancer Res 68(7):2106–2111. , DOI 10.1158/0008-5472.can-07-5211 Lichtenberger BM, Tan PK, Niederleithner H, Ferrara N, Petzelbauer P, Sibilia M, 2010, Autocrine VEGF signaling synergizes with EGFR in tumor cells to promote epithelial cancer development. Cell 140(2):268–279. , DOI 10.1016/j.cell. 2009.12.046 Larsen AK, Ouaret D, El Ouadrani K, Petitprez A, 2011, Targeting EGFR and VEGF(R, pathway cross-talk in tumor survival and angiogenesis. Pharmacol Ther 131(1):80–90. , DOI 10. 1016/j.pharmthera.2011.03.012 Stanic K, Zwitter M, Hitij NT, Kern I, Sadikov A, Cufer T, 2014, Brain metastases in lung adenocarcinoma: impact of EGFR mutation status on incidence and survival. Radiol Oncol 48(2):173–183. , DOI 10.2478/raon-2014-0016 Hendriks LE, Smit EF, Vosse BA, Mellema WW, Heideman DA, Bootsma GP, Westenend M, Pitz C, de Vries GJ, Houben R, Grunberg K, Bendek M, Speel EJ, Dingemans AM, 2014, EGFR mutated non-small cell lung cancer patients: more prone to development of bone and brain metastases? Lung Cancer 84(1):86–91. , DOI 10.1016/j.lungcan.2014.01.006 BaekMY, Ahn HK, Park KR, Park HS, Kang SM, Park I, Kim YS, Hong J, Sym SJ, Park J, Lee JH, Shin DB, Cho EK, 2018, Epidermal growth factor receptor mutation and pattern of brain metastasis in patients with non-small cell lung cancer. Korean J Intern Med 33(1):168–175. , DOI 10.3904/kjim.2015.158 Wells GA, Shea BJ, O'Connell D, Peterson J, Welch V, Losos M, Tugwell P, 2014, The Newcastle–Ottawa scale, NOS, for assessing the quality of non-randomized studies in Meta-analysis. Appl Eng Agric 18, 6):pags. 727–734 Lee YJ, Park IK, ParkM-S, Choi HJ, Cho BC, Chung KY, Kim SK, Chang J, Moon JW, Kim H, Choi SH, Kim J-H, 2009, Activating mutations within the EGFR kinase domain: amolecular predictor of disease-free survival in resected pulmonary adenocarcinoma. J Cancer Res Clin Oncol 135(12):1647–1654. , DOI 10. 1007/s00432-009-0611-7 Doebele RC, Lu X, Sumey C, Maxson DA, Weickhardt AJ, Oton AB, Bunn PA Jr, Baron AE, Franklin WA, Aisner DL, Varella- Garcia M, Camidge DR, 2012, Oncogene status predicts patterns of metastatic spread in treatment-naive nonsmall cell lung cancer. Cancer 118(18):4502–4511. , DOI 10.1002/cncr.27409 Enomoto Y, Takada K, Hagiwara E, Kojima E, 2013, Distinct features of distant metastasis and lymph node stage in lung adenocarcinoma patients with epidermal growth factor receptor gene mutations. Respir Investig 51(3):153–157. , DOI 10.1016/j. resinv.2013.02.004 Akamatsu H, Kaira K, Murakami H, Serizawa M, Koh Y, Ono A, Shukuya T, Tsuya A, Nakamura Y, Kenmotsu H, Naito T, Takahashi T, Endo M, Harada H, Nakajima T, Yamamoto N, 2014, The impact of clinical outcomes according to EGFR mutation status in patients with locally advanced lung adenocarcinoma who recieved concurrent chemoradiotherapy. Am J Clin Oncol 37(2):144–147. , DOI 10.1097/COC.0b013e31826e04f9 Fujimoto D, Ueda H, Shimizu R, Kato R, Otoshi T, Kawamura T, Tamai K, Shibata Y, Matsumoto T, Nagata K, Otsuka K, Nakagawa A, Otsuka K, Katakami N, Tomii K, 2014, Features and prognostic impact of distant metastasis in patients with stage IV lung adenocarcinoma harboring EGFR mutations: importance of bone metastasis. Clin Exp Metastasis 31(5):543–551. , DOI 10.1007/ s10585-014-9648-3 Shin DY, Na II, Kim CH, Park S, Baek H, Yang SH, 2014, EGFR mutation and brain metastasis in pulmonary adenocarcinomas. J Thorac Oncol 9(2):195–199 Iuchi T, Shingyoji M, Itakura M, Yokoi S, Moriya Y, Tamura H, Yoshida Y, Ashinuma H, Kawasaki K, Hasegawa Y, Sakaida T, Iizasa T, 2015, Frequency of brain metastases in non-small-cell lung cancer, and their association with epidermal growth factor receptor mutations. Int J Clin Oncol 20(4):674–679. https://doi. org/10.1007/s10147-014-0760-9 Li B, Sun SZ, Yang M, Shi JL, Xu W, Wang XF, Song MM, Chen HM, 2015, The correlation between EGFR mutation status and the risk of brain metastasis in patients with lung adenocarcinoma. J Neuro-Oncol 124(1):79–85. , DOI 10.1007/s11060-015- 1776-3 Schuette W, Schirmacher P, Eberhardt WE, Fischer JR, von der Schulenburg JM, Mezger J, Schumann C, Serke M, Zaun S, Dietel M, Thomas M, 2015, EGFR mutation status and first-line treatment in patients with stage III/IV non-small cell lung cancer in Germany: an observational study. Cancer Epidemiol Biomark Prev 24(8):1254–1261. , DOI 10.1158/1055-9965.EPI-14-1149 Tanaka K, Hida T, Oya Y, Oguri T, Yoshida T, Shimizu J, Horio Y, Hata A, Kaji R, Fujita S, Sekido Y, Kodaira T, Kokubo M, Katakami N, Yatabe Y, 2015, EGFR mutation impact on definitive concurrent Chemoradiation therapy for inoperable stage III adenocarcinoma. J Thorac Oncol 10(12):1720–1725. , DOI 10. 1097/JTO.0000000000000675 Yagishita S, Horinouchi H, Katsui Taniyama T, Nakamichi S, Kitazono S, Mizugaki H, Kanda S, Fujiwara Y, Nokihara H, Yamamoto N, Sumi M, Shiraishi K, Kohno T, Furuta K, Tsuta K, Tamura T, 2015, Epidermal growth factor receptor mutation is associated with longer local control after definitive chemoradiotherapy in patients with stage III nonsquamous non-small-cell lung cancer. Int J Radiat Oncol Biol Phys 91(1):140–148. https://doi. org/10.1016/j.ijrobp.2014.08.344 Chen YM, Lai CH, Rau KM, Huang CH, Chang HC, Chao TY, Tseng CC, Fang WF, Chen YC, Chung YH, Wang YH, Su MC, Huang KT, Liu SF, ChenHC, ChangYC, Chang YP,Wang CC, Lin MC, 2016, Advanced non-small cell lung cancer patients at the extremes of age in the era of epidermal growth factor receptor tyrosine kinase inhibitors. Lung Cancer 98:99–105. https://doi. org/10.1016/j.lungcan.2016.05.020 Hsu F, Caluwe AD, Anderson D, Nichol A, Toriumi T, Ho C, 2016, EGFR mutation status on brain metastases from non-small cell lung cancer. Lung Cancer 96:101–107 Li BT, Lou E, Hsu M, Yu HA, Naidoo J, Zauderer MG, Sima C, Johnson ML, Daras M, DeAngelis LM, Fleisher M, Kris MG, Azzoli CG, 2016, Serum biomarkers associated with clinical outcomes fail to predict brain metastases in patients with stage IV nonsmall cell lung cancers. PLoS One 11(1):e0146063. , DOI 10.1371/journal.pone.0146063 Li H, Cao J, Zhang X, Song X,WangW, Jia S, Li Z, Jia H, Cao X, Zhou W, Lian J, Han S, Yang W, Xi Y, Lian S, Jing H, 2017, Correlation between status of epidermal growth factor receptor mutation and distant metastases of lung adenocarcinoma upon initial diagnosis based on 1063 patients in China. Clin Exp Metastasis 34(1):63–71. , DOI 10.1007/s10585-016-9822-x Luo YH, Wu CH, Huang CY, Wu CW, Wu WS, Lee YC, Whang- Peng J, Chen YM, 2017, Brain metastasis features and association with tumor epidermal growth factor receptor mutation in patients with adenocarcinoma of the lung. Asia Pac J Clin Oncol 13(5): e440–e448. , DOI 10.1111/ajco.12576 Renaud S, Seitlinger J, Falcoz PE, Schaeffer M, Voegeli AC, Legrain M, Beau-Faller M, Massard G, 2016, Specific KRAS amino acid substitutions and EGFR mutations predict site-specific recurrence and metastasis following non-small-cell lung cancer surgery. Br J Cancer 115(3):346–353. , DOI 10.1038/bjc.2016.182 Mizuno T, Yatabe Y, Kuroda H, Sakakura N, Sakao Y, 2016, Impact of the oncogenic status on the mode of recurrence in resected non-small cell lung cancer. Jpn J Clin Oncol 46(10):928– 934. , DOI 10.1093/jjco/hyw094 Bhatt VR, D’Souza SP, Smith LM, Cushmanvokoun AM, Noronha V, Verma V, Joshi A, Chougule A, Jambhekar N, Kessinger A, 2017, Epidermal growth factor receptor mutational status and brain metastases in non–small-cell lung Cancer. J Glob Oncol 3(3):208–217 GeM, Zhuang Y, Zhou X, Huang R, Liang X, Zhan Q, 2017, High probability and frequency of EGFR mutations in non-small cell lung cancer with brain metastases. J Neuro-Oncol 135(2):413– 418. , DOI 10.1007/s11060-017-2590-x Russo A, Franchina T, Ricciardi GR, Fanizza C, Scimone A, Chiofalo G, Giordano A, Adamo V, 2017, Influence of EGFR mutational status on metastatic behavior in non squamous non small cell lung cancer. Oncotarget 8(5):8717–8725 Wang BX,OuW,Mao XY, Liu Z, Wu HQ,Wang SY(2017, Impacts of EGFR mutation andEGFR-TKIs on incidence of brainmetastases in advanced non-squamous NSCLC. Clin Neurol Neurosurg 160:96– 100. , DOI 10.1016/j.clineuro.2017.06.022 Higgins JP, Thompson SG, Deeks JJ, Altman DG, 2003, Measuring inconsistency in meta-analyses. BMJ 327(7414):557– 560. , DOI 10.1136/bmj.327.7414.557 Hubbs JL, Boyd JA, Hollis D, Chino JP, SaynakM, Kelsey CR, 2010, Factors associated with the development of brain metastases: analysis of 975 patients with early stage nonsmall cell lung cancer. Cancer 116(21):5038–5046. , DOI 10.1002/cncr.25254 Sun DS, Hu LK, CaiY, LiXM, Ye L, Hou HY,WangCH, JiangYH, 2014, A systematic review of risk factors for brain metastases and value of prophylactic cranial irradiation in non-small cell lung cancer. Asian Pac J Cancer Prev : APJCP 15(3):1233–1239 Chen G, Wang Z, Liu XY, Liu FY, 2011, High-level CXCR4 expression correlates with brain-specific metastasis of non-small cell lung cancer. World J Surg 35(1):56–61. , DOI 10.1007/ s00268-010-0784-x Arora S, Ranade AR, Tran NL, Nasser S, Sridhar S, Korn RL, Ross JT, DhruvH, FossKM, Sibenaller Z, Ryken T, Gotway MB,Kim S,Weiss GJ, 2011, MicroRNA-328 is associated with, non-small, cell lung cancer, NSCLC, brain metastasis and mediates NSCLC migration. Int J Cancer 129(11):2621–2631. , DOI 10.1002/ijc.25939 Yoo JY, Yang SH, Lee JE, Cho DG, Kim HK, Kim SH, Kim IS, Hong JT, Sung JH, Son BC, 2012, E-cadherin as a predictive marker of brain metastasis in non-small-cell lung cancer, and its regulation by pioglitazone in a preclinical model. J Neuro-Oncol 109(2):219–227 Jorissen RN, Walker F, Pouliot N, Garrett TP, Ward CW, Burgess AW, 2003, Epidermal growth factor receptor: mechanisms of activation and signalling. Exp Cell Res 284(1):31–53 Dearden S, Stevens J, Wu YL, Blowers D, 2013, Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology, mutMap). Annals Oncol 24(9):2371– 2376 Heon S, Yeap BY, Britt GJ, Costa DB, Rabin MS, Jackman DM, Johnson BE, 2010, Development of central nervous system metastases in patients with advanced non-small cell lung cancer and somatic EGFR mutations treated with gefitinib or erlotinib. Clin Cancer Res 16(23):5873–5882. , DOI 10.1158/1078- 0432.ccr-10-1588 Sekine A, Kato T, Hagiwara E, Shinohara T, Komagata T, Iwasawa T, Satoh H, Tamura K, Kasamatsu T, Hayashihara K, Saito T, Takahashi H, Ogura T, 2012, Metastatic brain tumors from nonsmall cell lung cancer with EGFR mutations: distinguishing influence of exon 19 deletion on radiographic features. Lung Cancer 77(1):64–69. , DOI 10.1016/j.lungcan.2011.12.017 Breindel JL, Haskins JW, Cowell EP, Zhao M, Nguyen DX, Stern DF, 2013, EGF receptor activates MET throughMAPK to enhance non-small cell lung carcinoma invasion and brain metastasis. Cancer Res 73(16):5053–5065. , DOI 10.1158/0008- 5472.can-12-3775 Benedettini E, Sholl LM, Peyton M, Reilly J, Ware C, Davis L, Vena N, Bailey D, Yeap BY, Fiorentino M, Ligon AH, Pan BS, Richon V, Minna JD, Gazdar AF, Draetta G, Bosari S, Chirieac LR, Lutterbach B, Loda M, 2010, Met activation in non-small cell lung cancer is associated with de novo resistance to EGFR inhibitors and the development of brain metastasis. Am J Pathol 177(1): 415–423. , DOI 10.2353/ajpath.2010.090863 Nie F, Yang J, Wen S, An YL, Ding J, Ju SH, Zhao Z, Chen HJ, Peng XG, Wong ST, Zhao H, Teng GJ, 2012, Involvement of epidermal growth factor receptor overexpression in the promotion of breast cancer brain metastasis. Cancer 118(21):5198–5209. , DOI 10.1002/cncr.27553 Singh M, Garg N, Venugopal C, Hallett R, Tokar T, McFarlane N, Mahendram S, Bakhshinyan D, Manoranjan B, Vora P, Qazi M, Arpin CC, Page B, Haftchenary S, Rosa DA, Lai PS, Gomez- Biagi RF, Ali AM, Lewis A, Geletu M, Murty NK, Hassell JA, Jurisica I, Gunning PT, Singh SK, 2015, STAT3 pathway regulates lung-derived brain metastasis initiating cell capacity through miR- 21 activation. Oncotarget 6(29):27461–27477. , DOI 10. 18632/oncotarget.4742 Gao SP, Mark KG, Leslie K, Pao W, Motoi N, Gerald WL, Travis WD, Bornmann W, Veach D, Clarkson B, Bromberg JF, 2007, Mutations in the EGFR kinase domain mediate STAT3 activation via IL-6 production in human lung adenocarcinomas. J Clin Invest 117(12):3846–3856. , DOI 10.1172/jci31871 Pottgen C, EberhardtW, Grannass A, Korfee S, Stuben G, Teschler H, Stamatis G, Wagner H, Passlick B, Petersen V, Budach V, Wilhelm H, Wanke I, Hirche H, Wilke HJ, Stuschke M, 2007, Prophylactic cranial irradiation in operable stage IIIA non smallcell lung cancer treated with neoadjuvant chemoradiotherapy: results from a German multicenter randomized trial. J Clin Oncol 25(31):4987–4992. , DOI 10.1200/jco.2007.12.5468 Gore EM,BaeK,Wong SJ, SunA, Bonner JA, Schild SE,GasparLE, Bogart JA, Werner-Wasik M, Choy H, 2011, Phase III comparison of prophylactic cranial irradiation versus observation in patients with locally advanced non-small-cell lung cancer: primary analysis of radiation therapy oncology group study RTOG 0214. J Clin Oncol 29(3): 272–278. , DOI 10.1200/jco.2010.29.1609 Dimitropoulos C, Hillas G, Nikolakopoulou S, Kostara I, Sagris K, Vlastos F, Alchanatis M, 2011, Prophylactic cranial irradiation in non-small cell lung cancer patients: who might be the candidates? Cancer Manag Res:287–294 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 791 EP 799 DI 10.1007/s12253-019-00598-0 PG 9 ER PT J AU Sipka, S Brugos, B Czifra, G Griger, Z Balogh, N Tarr, T Papp, G Biro, T Zeher, M AF Sipka, Sandor Brugos, Boglarka Czifra, Gabriella Griger, Zoltan Balogh, Norbert Tarr, Tunde Papp, Gabor Biro, Tamas Zeher, Margit TI Season Dependent Changes in the Expression of Protein Kinase C Isoenzymes in a Female Patient with Systemic Lupus Erythematosus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE European white women; Peripheral blood mononuclear cells (PBMC); Protein kinase C (PKC); Season dependence; Systemic lupus erythematosus (SLE) ID European white women; Peripheral blood mononuclear cells (PBMC); Protein kinase C (PKC); Season dependence; Systemic lupus erythematosus (SLE) AB We aimed to answer the question whether the decreased expression of protein kinase C (PKC) isoenzymes in the peripheral blood mononuclear cells (PBMC) of patients with systemic lupus erythematosus (SLE) is inherited or not. For this reason we examined the expression of PKC isoenzymes in a European white girl with acute SLE and in her healthy mother and father simultaneously in summer and winter during one year using western blotting and densitometry. We found that in the father the expression of PKC isoenzymes did not differ from that of eight healthy controls included women and men. However, in the "SLE-free" mother and in the patient arrived in July with acute symptoms of lupus, the expression of PKC isoenzymes showed a season dependent undulation in parallel. Namely, in summer the expression values were significantly lower, in winter they were significantly higher than those in the controls. Thus, the decreased expression of PKC isoenzymes in the PBMC of SLE patient is not a disease specific marker; it appears also in her lupus free mother. This phenomenon may be due to a season dependent female genetic background. However, the low PKC levels in summer can still decrease further the low production of IL-2 in T cells of lupus patients augmenting the existing AP-1 defects. This is the first report on the season and female dependent inherited changing of PKC expression in a European white patient with SLE and her mother. Further studies are needed to confirm these findings in other populations. C1 [Sipka, Sandor] University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, Moricz Zs. str. 22, H-4032 Debrecen, Hungary. [Brugos, Boglarka] University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, Moricz Zs. str. 22, H-4032 Debrecen, Hungary. [Czifra, Gabriella] University of Debrecen, Faculty of Medicine, Department of Physiology, Nagyerdei blvd, 98, H-4032 Debrecen, Hungary. [Griger, Zoltan] University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, Moricz Zs. str. 22, H-4032 Debrecen, Hungary. [Balogh, Norbert] University of Debrecen, Faculty of Medicine, Department of Physiology, Nagyerdei blvd, 98, H-4032 Debrecen, Hungary. [Tarr, Tunde] University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, Moricz Zs. str. 22, H-4032 Debrecen, Hungary. [Papp, Gabor] University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, Moricz Zs. str. 22, H-4032 Debrecen, Hungary. [Biro, Tamas] University of Debrecen, Faculty of Medicine, Department of Physiology, Nagyerdei blvd, 98, H-4032 Debrecen, Hungary. [Zeher, Margit] University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, Moricz Zs. str. 22, H-4032 Debrecen, Hungary. RP Sipka, S (reprint author), University of Debrecen, Faculty of Medicine, Division of Clinical Immunology, H-4032 Debrecen, Hungary. EM sipka@iiibel.dote.hu CR Biro T, Griger Z, Kiss E, Papp H, Aleksza M, Kovacs I, Zeher M, Bodolay E, Csepany T, Szucs K, Gergely P, Kovacs L, Szegedi G, Sipka S, 2004, Abnormal cell specific expression of certain protein kinase C isoenzymes in peripheral mononuclear cells of patients with systemic lupus erythematosus: effect of corticosteroid application. Scand J Immunol 60:421–442 Webster JR Moenter SM, Woodfill CJ, Karsch FJ, 1991, Role of the thyroid gland in seasonal reproduction. II. Thyroxine allows a season-specific suppression of gonadotropin secretion in sheep. Endocrinology 129:176–183 Cedeno S, Cifarelli DF, Blasini AM, Placeres F, Alonso G, Rodriguez MA, 2003, Defective activity of ERK-1 and ERK-2 mitogen activated protein kinases in peripheral blood T lymphocytes from patients with systemic lupus erythematosus: potential role of altered coupling of Ras guanine nucleotide exchange factor hSos to adapter protein Grb2 in lupus T cells. Clin Immunol 106: 41–49 Gorelik G, Fang JY, Wu A, Sawalha AH, Richardson B, 2007, Impaired T cell protein kinase C delta activation decreases ERK pathway signalling in idiopathic and hydralazine induced lupus. J Immunol 179:5553–5563 Konya C, Paz Z, Tsokos GC, 2014, The role of T cells in systemic lupus erythematosus: an update. Curr Opin Rheumatol 26:493–501 Moulton VR, Tsokos GS, 2012, Why do women get lupus? Clin Immunol 144:53–56 McCuaig RD, Dunn J, Li J, Masch A, Knaute T, Schutkowski M, Zerweck J, Rao S, 2015, PKC-theta is a novel SC35 splicing factor regulator in response to Tcell activation. Front Immunol 6:562–576 Hughes GC, Choubey D, 2004, Modulation of autoimmune rheumatic diseases by oestrogen and progesterone. Nat Rev Rheumatol 10:740–751 Panchanathan R, Shen H, Bupp MG, Gould KA, Choubey D, 2009, Female and male sex hormones differentially regulate expressions of lfi202, an interferon-inducible lupus susceptibility gene within the Nba2 interval. J Immunol 183:7031–7038 Tesar V, Hruskova Z, 2015, Lupus nephritis: a different disease in European patients? Kidney Dis 1:110–118 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 801 EP 805 DI 10.1007/s12253-019-00591-7 PG 5 ER PT J AU Darre, T Aboubakari, AS N’Bortche, KB Bassowa, A Napo-Koura, G AF Darre, Tchin Aboubakari, Abdoul-Samadou N’Bortche, K Bingo Bassowa, Akila Napo-Koura, Gado TI Association of Schistosomiasis with Cervical Cancer in Togo: the Consequence of this Association SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE Cervical cancer; Schistosomiasis; Carcinoma; HPV; Togo ID Cervical cancer; Schistosomiasis; Carcinoma; HPV; Togo AB Objective of the study was to determine the association of cervical cancer association and schistosomiasis infection. We conducted a retrospective and descriptive study of cases of cervical cancer and identified cases associated with schistosomiasis. A total of 1027 cases of cervical cancer were collected, and 19 cases revealed an association with schistosomiasis. This association was mainly related to squamous carcinoma with 18/19 cases. All patients were from rural areas. Of the 19 cases of association, 17 cases showed signs of HPV infection. Our data show a high degree of HPV infection that causes cervical cancer and not schistosomiasis. C1 [Darre, Tchin] University Teaching Hospital of Lome, Department of PathologyLome, Togo. [Aboubakari, Abdoul-Samadou] The University Teaching Hospital of Kara, Department Obstetrics and GynecologyKara, Togo. [N’Bortche, K Bingo] The University Teaching Hospital of Lome, Department Obstetrics and GynecologyLome, Togo. [Bassowa, Akila] The University Teaching Hospital of Lome, Department Obstetrics and GynecologyLome, Togo. [Napo-Koura, Gado] University Teaching Hospital of Lome, Department of PathologyLome, Togo. RP Darre, T (reprint author), University Teaching Hospital of Lome, Department of Pathology, Lome, Togo. EM paolodarre@yahoo.fr CR Plummer M, de Martel C, Vignat J et al, 2016, Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob Health 4(9):e609–e616 Darre T, Kpatcha K, Tchaou M et al, 2015, Histological aspect of urinary schistosomiasis in Togo: results of a cohort of 192 cases. Bull Soc Pathol Exot 108(2):124–125 GayeAM, Doh K, ThiamI et al, 2016, Schistosomiasis and cancer: a fortuitous association or relationships cause and effect. Bull Cancer 103(9):806–807 Mandong BM, Ngbea JA, Raymond V, 2013, Role of parasites in cancer. Niger J Med 22(2):89–92 Riffenburgh RH, Olson PE, Johnstone PA, 1997, Association of schistosomiasis with cervical cancer: detecting bias in clinical studies. East Afr Med J 74(1):14–16 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 807 EP 808 DI 10.1007/s12253-017-0350-z PG 2 ER PT J AU Stoyanov, G Kitanova, M Dzhenkov, D Ghenev, P AF Stoyanov, George Kitanova, Martina Dzhenkov, Deyan Ghenev, Peter TI The Diagnostic Dilemma of Epithelial Marker Expression in Glioblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Stoyanov, George] Medical University - Varna Prof. Dr. Paraskev Stoyanov, Faculty of Medicine, Department of Anatomy and Cell BiologyVarna, Bulgaria. [Kitanova, Martina] Medical University - Varna Prof. Dr. Paraskev Stoyanov, Faculty of Medicine, Department of General and Clinical Pathology, Forensic Medicine and DeontologyVarna, Bulgaria. [Dzhenkov, Deyan] Medical University - Varna Prof. Dr. Paraskev Stoyanov, Faculty of Medicine, Department of General and Clinical Pathology, Forensic Medicine and DeontologyVarna, Bulgaria. [Ghenev, Peter] Medical University - Varna Prof. Dr. Paraskev Stoyanov, Faculty of Medicine, Department of General and Clinical Pathology, Forensic Medicine and DeontologyVarna, Bulgaria. RP Stoyanov, G (reprint author), Medical University - Varna Prof. Dr. Paraskev Stoyanov, Faculty of Medicine, Department of Anatomy and Cell Biology, Varna, Bulgaria. EM georgi.geesh@gmail.com CR Terada T, 2015, Expression of cytokeratins in glioblastoma multiforme. Pathol Oncol Res 21(3):817–819 Kriho VK, Yang HY,Moskal JR, Skalli O, 1997, Keratin expression in astrocytomas: an immunofluorescent and biochemical reassessment. Virchows Arch 431(2):139–147 Oh D, Prayson RA, 1999, Evaluation of epithelial and keratin markers in glioblastoma multiforme: an immunohistochemical study. Arch Pathol Lab Med 123(10):917–920 Stoyanov GS, Dzhenkov D, Ghenev P, 2017, Cytokeratin AE1/AE3 mimicry in glioblastoma. Script Sci Med 49(1):47-52 Stoyanov GS, Dzhenkov D, Ghenev P, 2017, The great imitator– EMA positive glioblastoma multiforme. Script Sci Med 49(1):21-25 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 809 EP 810 DI 10.1007/s12253-017-0354-8 PG 2 ER PT J AU Sabato, C Bastos-Rodrigues, L Chaves Moraes, D Friedman, E De Marco, L Resende, V AF Sabato, Cristina Bastos-Rodrigues, Luciana Chaves Moraes, Debora Friedman, Eitan De Marco, Luiz Resende, Vivian TI Genetic Analysis of Brazilian Patients with Gallbladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Sabato, Cristina] Universidade Federal de Minas Gerais, Department of Surgery, Av. Alfredo Balena 190, room 114, 30130-100 Belo Horizonte, Brazil. [Bastos-Rodrigues, Luciana] Universidade Federal de Minas Gerais, Department of NutritionBelo Horizonte, Brazil. [Chaves Moraes, Debora] Universidade Federal de Minas Gerais, Department of Surgery, Av. Alfredo Balena 190, room 114, 30130-100 Belo Horizonte, Brazil. [Friedman, Eitan] Chaim Sheba Medical Center, The Susanne Levy Gertner Oncogenetics UnitTel-Hashomer, Israel. [De Marco, Luiz] Universidade Federal de Minas Gerais, Department of Surgery, Av. Alfredo Balena 190, room 114, 30130-100 Belo Horizonte, Brazil. [Resende, Vivian] Universidade Federal de Minas Gerais, Department of Surgery, Av. Alfredo Balena 190, room 114, 30130-100 Belo Horizonte, Brazil. RP De Marco, L (reprint author), Universidade Federal de Minas Gerais, Department of Surgery, 30130-100 Belo Horizonte, Brazil. EM ldemarco@ufmg.br;luiz.demarco@gmail.com CR Bal MM, Ramadwar M, Deodhar K, Shrikhande S, 2015, Pathology of gallbladder carcinoma: current understanding and new perspectives. Pathol. Oncol. Res. 21:509–525 Jain K,Mohapatra T, Das P, MisraMC, Gupta SD, GhoshM, Kabra M, Bansal VK, Kumar S, Sreenivas V, Garg PK, 2014, Sequential occurrence of preneoplastic lesions and accumulation of loss of heterozygosity in patients with gallbladder stones suggest causal association with gallbladder cancer. Ann. Surg. 260:1073–1080 Espinoza JA, Bizama C, Garcia P, Ferreccio C, Javle M,Miquel JF, Koshiol J, Roa JC, 2016, The inflammatory inception of gallbladder cancer. Biochim. Biophys. Acta 1865:245–254 Nakamura H, Arai Y, Totoki Y, Shirota T, Elzawahry A, Kato M, Hama N, Hosoda F, Urushidate T, Ohashi S, Hiraoka N, Ojima H, Shimada K, Okusaka T, Kosuge T, Miyagawa S, Shibata T, 2015, Genomic spectra of biliary tract cancer. Nat. Genet. 47:1003–1010 Bastos-Rodrigues L, Pimenta JR, Pena SD, 2006, The genetic structure of human populations studied through short insertiondeletion polymorphisms. Ann. Hum. Genet. 70:658–665 Serra S, 2014, Precursor neoplastic lesions of the biliary tract. J. Clin. Pathol. 67:875–882 Nagahashi M, Ajioka Y, Lang I, Szentirmay Z, Kasler M, Nakadaira H, Yokoyama N, Watanabe G, Nishikura K, Wakai T, Shirai Y, Hatakeyama K, Yamamoto M, 2008, Genetic changes of p53, K-ras, and microsatellite instability in gallbladder carcinoma in high-incidence areas of Japan and Hungary. World J. Gastroenterol. 14:70–75 Saetta AA, 2006, K-ras, p53 mutations, and microsatellite instability, MSI, in gallbladder cancer. J. Surg. Oncol. 93:644–649 Hanada K, Tsuchida A, Iwao T, Eguchi N, Sasaki T, Morinaka K, Matsubara K, Kawasaki Y, Yamamoto S, Kajiyama G, 1999, Gene mutations of K-ras in gallbladder mucosae and gallbladder carcinoma with an anomalous junction of the pancreaticobiliary duct. Am. J. Gastroenterol. 94:1638–1642 Bakos RM, Besch R, Zoratto GG, Godinho JM, Mazzotti NG, Ruzicka T, Bakos L, Santos SE, Ashton-Prolla P, Berking C, Giugliani R, 2011, The CDKN2A p.A148T variant is associated with cutaneous melanoma in southern Brazil. Exp. Dermatol. 20: 890–893 Soufir N, Ribojad M, Magnaldo T, Thibaudeau O, Delestaing G, Daya-Grosjean L, Rivet J, Sarasin A, Basset-Seguin N, 2002, Germline and somatic mutations of the INK4a-ARF gene in a xeroderma pigmentosum group C patient. J Invest Dermatol 119: 1355–1360 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 811 EP 814 DI 10.1007/s12253-018-0407-7 PG 4 ER PT J AU Wiwanitkit, V AF Wiwanitkit, Viroj TI Early Skin Rash and Cetuximab-Based Therapy of Advanced Biliary Tract Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Wiwanitkit, Viroj] Hainan Medical UniversityHaikou, China. RP Wiwanitkit, V (reprint author), Hainan Medical University, Haikou, China. CR RubovszkyG,BudaiB,GanofszkyE,HorvathZ,JuhosE,Madaras B,NagyT,SzaboE,PinterT,TothE,NagyP,LangI,HitreE(2017 Apr 29)Predictive Value of Early Skin Rash in Cetuximab-Based Therapy of Advanced Biliary Tract Cancer. Pathol Oncol Res. , DOI 10.1007/s12253-017-0238-y [Epubaheadof print] Opneja A, Mahajan S, Kapoor S, Marur S, Yang SH, Manno R, 2015)Unusual Paraneoplastic Presentation of Cholangiocarcinoma.Case RepMed2015:806835 MacLean JD, Graeme-CookFM, 2002Apr 18)Case records of the Massachusetts General Hospital. Weekly clinicopathological exercises.Case12-2002.A50-year-oldmanwith eosinophilia andfluctuatinghepaticlesions.NEnglJMed346(16):1232–1239 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 815 EP 815 DI 10.1007/s12253-018-0424-6 PG 1 ER PT J AU Stein, KM Morris, KL Martin, GM AF Stein, K Matthew Morris, K Lindsay Martin, G Mike TI JAK Pseudokinase Domain Variants Highlight nRTK VUSs Identified with Next-Generation Sequencing in Solid Tumor Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Stein, K Matthew] West Cancer CenterMemphis, TN, USA. [Morris, K Lindsay] University of Tennessee, Health Science Center, College of MedicineMemphis, TN, USA. [Martin, G Mike] West Cancer CenterMemphis, TN, USA. RP Stein, KM (reprint author), West Cancer Center, Memphis, USA. EM mkstein@westclinic.com CR Lupardus PJ, Ultsch M, Wallweber H et al, 2014, Structure of the pseudokinase-kinase domains from protein kinase TYK2 reveals a mechanism for Janus kinase, JAK, autoinhibition. Proc Natl Acad Sci U S A 111:8025–8030 Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS, 2010, Sunyaev SR. NatMethods 7(4):248– 249 Cante-Barrett K, Uitdehaag JC, Meijerink JP, 2016, Structural modeling of JAK1 mutations in T-cell acute lymphoblastic leukemia reveals a second contact site between pseudokinase and kinase domains. Haematologica 101(5):e189–e191 Perez C, Gonzalez-Rincon J, Onaindia A et al, 2015, Mutated JAK kinases and deregulated STAT activity are potential therapeutic targets in cutaneous T-cell lymphoma. Haematologica 100(11):e450–e453 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 817 EP 818 DI 10.1007/s12253-018-0443-3 PG 2 ER PT J AU Ganesan, R Sivalingam, N AF Ganesan, Ramamoorthi Sivalingam, Nageswaran TI Transforming Growth Factor Beta 2 Inhibits Growth and Proliferation Potential of Smad4 and p53 Mutated Human Colon Adenocarcinoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Ganesan, Ramamoorthi] SRM Institute of Science and Technology, School of Bioengineering, Department of Biotechnology, 603203 Kattankulathur, Tamilnadu, India. [Sivalingam, Nageswaran] SRM Institute of Science and Technology, School of Bioengineering, Department of Biotechnology, 603203 Kattankulathur, Tamilnadu, India. RP Sivalingam, N (reprint author), SRM Institute of Science and Technology, School of Bioengineering, Department of Biotechnology, 603203 Kattankulathur, India. EM nageswaran.s@ktr.srmuniv.ac.in CR Siegel R, Ma J, Zou Z, Jemal A, 2014, Cancer statistics 2014. CA Cancer J Clin 64:9–29 Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics 2016. CA Cancer J Clin 66:7–30 Wang L, Xue M, Chung DC, 2016, c-Myc is regulated by HIF-2α in chronic hypoxia and influences sensitivity to 5-FU in colon cancer. Oncotarget 7:78910–78917 Principe DR, DeCant B, Staudacher J et al, 2015, Loss of TGFβ signaling promotes colon cancer progression and tumor-associated inflammation. Oncotarget 8:3826–3839 Meulmeester E, Dijke P, 2011, The dynamic roles of TGF-β in cancer. J Pathol 223:206–219 Connolly EC, Freimuth J,Akhurst RJ, 2012, Complexities of TGF- β targeted cancer therapy. Int J Biol Sci 8:964–978 Derynck R, Zhang YE, 2003, Smad-dependent and Smadindependent pathways in TGF-beta family signalling. Nature 425: 577–584 Ramamoorthi G, Sivalingam N, 2014, Molecular mechanism of TGF-β signaling pathway in colon carcinogenesis and status of curcumin as chemopreventive strategy. Tumour Biol 35:7295–7305 Lampropoulos P, Zizi-Sermpetzoglou A, Rizos S, Kostakis A, Nikiteas N, Papavassiliou AG, 2012, TGF-beta signalling in colon carcinogenesis. Cancer Lett 314:1–7 Zhang B, Halder SK, Kashikar ND et al, 2010, Antimetastatic role of Smad4 signaling in colorectal cancer. Gastroenterology 138: 969–980 Kardamakis D, Ravazoula P, Zolota V, Salakou S, Tepetes K, Kalogeropoulou C, 2004, The potential role of TGFβ1, TGFβ2 and TGFβ3 protein expression in colorectal carcinomas. Strahlenther Onkol 18:201–208 Gulubova M, Manolova I, Ananiev J, Julianov A, Yovchev Y, Peeva K, 2010, Role of TGF-β1, its receptor TGFβRII, and Smad proteins in the progression of colorectal cancer. Int J Color Dis 25:591–599 Han S, Bui NT, Ho MT, Kim YM, Cho M, Shin DB, 2016, Dexamethasone inhibits TGF-β1-induced cell migration by regulating the ERK and AKT pathways in human colon cancer cells via CYR61. Cancer Res Treat 48:1141–1153 Massague J, 2008, TGF beta in Cancer. Cell 134:215–230 Chen S, Zhu J, Zuo S et al, 2015, 1,25(OH)2D3 attenuates TGF- β1/β2-induced increased migration and invasion via inhibiting epithelial-mesenchymal transition in colon cancer cells. Biochem Biophys Res Commun 468:130–135 Watson JL, Hill R, Yaffe PB et al, 2010, Curcumin causes superoxide anion production and p53-independent apoptosis in human colon cancer cells. Cancer Lett 297:1–8 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 819 EP 821 DI 10.1007/s12253-018-0423-7 PG 3 ER PT J AU Jo, SY Kim, SS Kim, SM Yoo, JN Lee, HS AF Jo, Sol Yun Kim, Soo Sung Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Candidate Tumor Suppressor Gene EAF2 is Mutated in Colorectal and Gastric Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Soo Sung] The Catholic University of Korea, College of Medicine, Department of Internal MedicineSeoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Simone F, Luo RT, Polak PE, Kaberlein JJ, Thirman MJ, 2003, ELLassociated factor 2, EAF2), a functional homolog of EAF1 with alternative ELL binding properties. Blood 101:2355–2362 Xiao W, Zhang Q, Jiang F, Pins M, Kozlowski JM, Wang Z, 2003, Suppression of prostate tumor growth by U19, a novel testosterone-regulated apoptosis inducer. Cancer Res 63:4698– 4704 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Oh HR, An CH, Yoo NJ, Lee SH, 2015, Frameshift mutations of MUC15 gene in gastric and its regional heterogeneity in gastric and colorectal cancers. Pathol Oncol Res 21:713–718 Liu JX, Zhang D, Xie X, Ouyang G, Liu X, Sun Y, Xiao W, 2013, Eaf1 and Eaf2 negatively regulate canonical Wnt/β-catenin signaling. Development 140:1067–1078 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 823 EP 824 DI 10.1007/s12253-018-0461-1 PG 2 ER PT J AU Son, JH Choi, JE Yoo, JN Lee, HS AF Son, Ji Hyun Choi, Ji Eun Yoo, Jin Nam Lee, Hyung Sug TI Promoter Mutation Analysis of ALDOA Gene in Solid Tumors and Acute Leukemias SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Son, Ji Hyun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Chang YC, Yang YC, Tien CP, Yang CJ, Hsiao M, 2018, Roles of aldolase family genes in human cancers and diseases. Trends Endocrinol Metab 29:549–559 Ji S, Zhang B, Liu J, Qin Y, Liang C, Shi S, Jin K, Liang D, Xu W, Xu H, Wang W, Wu C, Liu L, Liu C, Xu J, Ni Q, Yu X, 2016, ALDOAfunctions as an oncogene in the highlymetastatic pancreatic cancer. Cancer Lett 374:127–135 Rheinbay E, Parasuraman P, Grimsby J, Tiao G, Engreitz JM, Kim J, Lawrence MS, Taylor-Weiner A, Rodriguez-Cuevas S, Rosenberg M, Hess J, Stewart C, Maruvka YE, Stojanov P, Cortes ML, Seepo S, Cibulskis C, Tracy A, Pugh TJ, Lee J, Zheng Z, Ellisen LW, Iafrate AJ, Boehm JS, Gabriel SB, Meyerson M, Golub TR, Baselga J, Hidalgo-Miranda A, Shioda T, Bernards A, Lander ES, Getz G, 2017, Recurrent and functional regulatory mutations in breast cancer. Nature 547:55–60 Heng J, Guo X, Wu W, Wang Y, Li G, Chen M, Peng L, Wang S, Dai L, Tang L, Wang J, 2017, Integrated analysis of promoter mutation, methylation and expression of AKT1 gene in Chinese breast cancer patients. PLoS One 12:e0174022 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2019 VL 25 IS 2 BP 825 EP 826 DI 10.1007/s12253-018-0530-5 PG 2 ER PT J AU Zhang, Xp Cheng, Qh Yang, Hj Qiao, Eq AF Zhang, Xi-ping Cheng, Qi-hui Yang, Hong-jian Qiao, En-qi TI Research Progresses in Cancer Stem Cells of Three Common Fertility-Related Female Malignancies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Cancer stem cells; Female; Malignant tumors; Breast Cancer; Ovarian Cancer; Cervical Cancer ID Cancer stem cells; Female; Malignant tumors; Breast Cancer; Ovarian Cancer; Cervical Cancer AB With abilities to renew themselves and lead to heterogeneity of tumors, cancer stem cells (CSCs) are similar to stem cells. As three leading causes of death that endanger women’s health, breast cancer, ovarian cancer and cervical cancer are characterized by high degree of malignancy, metastasis and recurrence. Associated with women’s fertility, these three malignancies are common and representative among females. These years, research findings have suggested that CSCs are closely connected with many cancers (including aforementioned three malignancies) and several processes of tumors such as their genesis and development. CSCs have become great concerns for current cancer treatment and interventions. This paper does not only summarize roles of CSCs in genesis, development, drug resistance, metastasis and recurrence of breast cancer, ovarian cancer and cervical cancer, but also proposes potential methods of treatment and intervention, in hope of inspiring readers and researchers. C1 [Zhang, Xi-ping] Zhejiang Cancer Hospital, Department of Breast Surgery, 310022 Hangzhou, Zhejiang Province, China. [Cheng, Qi-hui] Hangzhou First People’s Hospital,, Department of Obstetrics and Gynecology, 310006, Hangzhou, Zhejiang Province, China. [Yang, Hong-jian] Zhejiang Cancer Hospital, Department of Breast Surgery, 310022 Hangzhou, Zhejiang Province, China. [Qiao, En-qi] Zhejiang Cancer Hospital, Department of Breast Surgery, 310022 Hangzhou, Zhejiang Province, China. RP Zhang, Xp (reprint author), Zhejiang Cancer Hospital, Department of Breast Surgery, 310022 Hangzhou, China. EM zxp99688@sina.com CR Luo M, Clouthier SG, Deol Y, Liu S, Nagrath S, Azizi E,Wicha MS, 2015, Breast cancer stem cells: current advances and clinical implications. Methods Mol Biol 1293:1–49 ClarkeMF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weissman IL, 2006, Wahl GM. Cancer stem cells– perspectives on current status and future directions: AACR workshop on cancer stem cells. Cancer Res 66(19):9339–9344 Geng SQ, Alexandrou AT, Li JJ, 2014, Breast cancer stem cells: Multiple capacities in tumor metastasis. Cancer Lett 349(1):1–7 Malanchi I, Santamaria-Martinez A, Susanto E, Peng H, Lehr HA, Delaloye JF, Huelsken J, 2011, Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 481(7379):85–89 Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA, 2008, The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133(4):704–715 Yang F, Xu J, Tang L, Guan X, 2017, Breast cancer stem cell: the roles and therapeutic implications. CellMol Life Sci 74(6):951–966 Kwak JH, Lee NH, Lee HY, Hong IS, Nam JS, 2016, HIF2alpha/ EFEMP1 cascade mediates hypoxic effects on breast cancer stem cell hierarchy. Oncotarget 7(28):43518–43533 Li YW, Shen H, Frangou C, Yang N, Guo J, Xu B, Bshara W, Shepherd L, Zhu Q, Wang J, Hu Q, Liu S, Morrison CD, Sun P, Zhang J, 2015, Characterization of TAZ domains important for the induction of breast cancer stem cell properties and tumorigenesis. Cell Cycle 14(1):146–156 Miyoshi Y, Shien T, Ogiya A, Ishida N, Yamazaki K, Horii R, Horimoto Y, Masuda N, Yasojima H, Inao T, Osako T, Takahashi M, Tomioka N, Endo Y, Hosoda M, Doihara H, Miyoshi S, Yamashita H, Collaborative Study Group of Scientific Research of the Japanese Breast Cancer S, 2016, Differences in expression of the cancer stem cell marker aldehyde dehydrogenase 1 among estrogen receptor-positive/human epidermal growth factor receptor type 2-negative breast cancer cases with early, late, and no recurrence. Breast Cancer Res 18(1):73–84 Liu S, Cong Y, Wang D, Sun Y, Deng L, Liu Y, Martin-Trevino R, Shang L, McDermott SP, Landis MD, Hong S, Adams A, D'Angelo R, Ginestier C, Charafe-Jauffret E, Clouthier SG, BirnbaumD,Wong ST, Zhan M, Chang JC, Wicha MS, 2013, Breast cancer stem cells transition between epithelial and mesenchymal states reflective of their normal counterparts. Stem Cell Reports 2(1):78–91 Barnawi R, Al-Khaldi S, Majed Sleiman G, Sarkar A, Al-Dhfyan A, Al-Mohanna F, Ghebeh H, Al-AlwanM(2016, Fascin is critical for the maintenance of breast Cancer stem cell pool predominantly via the activation of the notch self-renewal pathway. Stem Cells 34(12):2799–2813 Wang YC, Tsai CF, Chuang HL, Chang YC, Chen HS, Lee JN, Tsai EM, 2016, Benzyl butyl phthalate promotes breast cancer stem cell expansion via SPHK1/S1P/S1PR3 signaling. Oncotarget 7(20): 29563–29576 Lawson DA, Bhakta NR, Kessenbrock K, Prummel KD, Yu Y, Takai K, Zhou A, Eyob H, Balakrishnan S, Wang CY, Yaswen P, Goga A, Werb Z, 2015, Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature 526(7571):131–135 Witt AE, Lee CW, Lee TI, Azzam DJ,Wang B, Caslini C, Petrocca F, Grosso J, Jones M, Cohick EB, Gropper AB, Wahlestedt C, Richardson AL, Shiekhattar R, Young RA, Ince TA, 2017, Identification of a cancer stem cell-specific function for the histone deacetylases, HDAC1 and HDAC7, in breast and ovarian cancer. Oncogene 36(12):1707–1720 Meng T, Liu J, Wen L, Yuan M, Cheng B, Hu Y, Zhu Y, Liu X, Yuan H, Hu F, 2016, Multi-cycle chemotherapy with the glycolipid-like polymeric micelles evade cancer stem cell enrichment in breast cancer therapy. Oncotarget 7(45):72978–72989 Kim SH, Kaschula CH, Priedigkeit N, Lee AV, Singh SV, 2016, Forkhead box Q1 is a novel target of breast Cancer stem cell inhibition by Diallyl Trisulfide. J Biol Chem 291(26):13495–13508 Lee KM, Lee M, Lee J, Kim SW, Moon HG, Noh DY, Han W, 2016, Enhanced anti-tumor activity and cytotoxic effect on cancer stem cell population of metformin-butyrate compared with metformin HCl in breast cancer. Oncotarget 7(25):38500–38512 Yeo SK, Guan JL, 2016, Hierarchical heterogeneity in mammary tumors and its regulation by autophagy. Autophagy 12(10): 1960–1961 Wang T, Fahrmann JF, Lee H, Li Y-J, Tripathi SC, Yue C, Zhang C, Lifshitz V, others, 2017, JAK/STAT3-Regulated Fatty Acid β- Oxidation Is Critical for Breast Cancer Stem Cell Self-Renewal and Chemoresistance. Cell Stem Cell 21(3):295–296 Yeo SK, French R, Spada F, Clarkson R, 2013, Opposing roles of Nfkb2 gene products p100 and p52 in the regulation of breast cancer stem cells. Stem Cell Rep 27;2(1):78–91 Yu D, Holm R, Goscinski MA, Trope CG, Nesland JM, Suo Z, 2016, Prognostic and clinicopathological significance of Cacna2d1 expression in epithelial ovarian cancers: a retrospective study. Am J Cancer Res 6(9):2088–2097 Yin X, Wang X, Shen B, Jing Y, Li Q, Cai MC, Gu Z, Yang Q, Zhang Z, Liu J, Li H, Di W, Zhuang G, 2016, AVEGF-dependent gene signature enriched in mesenchymal ovarian cancer predicts patient prognosis. Sci Rep 6:31079 ChenWC, Hsu HP, Li CY, Yang YJ, Hung YH, Cho CY,Wang CY, Weng TY, Lai MD, 2016, Cancer stem cell marker CD90 inhibits ovarian cancer formation via beta3 integrin. Int J Oncol 49(5): 1881–1889 Kim DK, Seo EJ, Choi EJ, Lee SI, Kwon YW, Jang IH, Kim SC, Kim KH, Suh DS, Seong-Jang K, Lee SC, Kim JH, 2016, Crucial role of HMGA1 in the self-renewal and drug resistance of ovarian cancer stem cells. Exp Mol Med 48:e255 Januchowski R, Swierczewska M, Sterzynska K, Wojtowicz K, Nowicki M, Zabel M, 2016, Increased expression of several collagen genes is associated with drug resistance in ovarian Cancer cell lines. J Cancer 7(10):1295–1310 Kang HG, Kim DH, Kim SJ, Cho Y, Jung J, Jang W, Chun KH, 2016, Galectin-3 supports stemness in ovarian cancer stemcells by activation of the Notch1 intracellular domain. Oncotarget 7(42): 68229–68241 Ridley AJ, 2013, RhoA, RhoB and RhoC have different roles in cancer cell migration. J Microsc 251(3):242–249 Sang XB, Sun KX,Wang LL, Chen S, Wu DD, Zong ZH, Zhao Y, 2016, Effects and mechanism of RhoC downregulation in NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 827 EP 835 DI 10.1007/s12253-018-0448-y PG 9 ER PT J AU Liu, C Jin, J Liang, D Gao, Z Zhang, Y Guo, T He, Y AF Liu, Congmin Jin, Jing Liang, Di Gao, Zhaoyu Zhang, Yachen Guo, Tiantian He, Yutong TI Long Noncoding RNA PVT1 as a Novel Predictor of Metastasis, Clinicopathological Characteristics and Prognosis in Human Cancers: a Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Long non-coding RNA; Plasmacytoma variant translocation 1; Metastasis; Clinical stage; Prognosis; Meta-analysis ID Long non-coding RNA; Plasmacytoma variant translocation 1; Metastasis; Clinical stage; Prognosis; Meta-analysis AB The present meta-analysis aimed to systematically evaluates the metastasis, clinical stage, and prognostic value regarding the expression levels of PVT1 in various cancers. Relevant literatures were searched in PubMed, Cochrane Library, Web of science, Embase databases, Chinese National Knowledge Infrastructure and Wanfang from inception up to 22 August 2017. After data were extracted, a meta-analysis was performed using Review Manager 5.3 and Stata 12.0 software. The meta-analysis showed that high expression of PVT1 could predict more lymph node metastasis (LNM) (Odds ratio, OR = 2.83, 95%confidence interval, CI: 1.76–4.54, P < 0.0001), distant metastasis (DM) (OR = 3.60, 95% CI: 1.08–12.03, P = 0.04), advanced clinical stage (OR = 4.37, 95% CI: 3.45–5.54, P < 0.00001) and poor overall survival (Hazard ratio, HR = 2.08, 95% CI: 1.82–2.37, P < 0.00001)in cancer. Subgroup analysis in different systems also showed the same results, including respiratory system, digestive system, urinary system and other systems, especially in respiratory system (LNM, OR = 4.57, 95% CI: 2.41–8.68, P < 0.00001; clinical stage, OR = 5.59, 95% CI: 3.59–8.71, P < 0.00001; OS, HR = 2.43, 95% CI: 1.98–2.99, P < 0.00001). These results suggest that PVT1 could serve as a novel biomarker for metastasis, clinical stage and poor prognosis in various tumors. C1 [Liu, Congmin] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 050000 Shijiazhuang, Hebei, China. [Jin, Jing] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 050000 Shijiazhuang, Hebei, China. [Liang, Di] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 050000 Shijiazhuang, Hebei, China. [Gao, Zhaoyu] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 050000 Shijiazhuang, Hebei, China. [Zhang, Yachen] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 050000 Shijiazhuang, Hebei, China. [Guo, Tiantian] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 050000 Shijiazhuang, Hebei, China. [He, Yutong] Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 050000 Shijiazhuang, Hebei, China. RP He, Y (reprint author), Fourth Hospital of Hebei Medical University, Department of Medical Oncology, 050000 Shijiazhuang, China. EM hytong69@yahoo.com CR GLOBOCAN, 2012, Estimated Cancer incidence, mortality and prevalence worldwide in 2012. http://globocan.iarc.fr/Pages/ burden_sel.aspx.[J] Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66:7–30 ChenW, Zheng R, Baade PD et al, 2016, Cancer statistics in China, 2015. CA Cancer J Clin 66:115–132 Yang B, Luo T, Zhang M et al, 2017, The novel long noncoding RNA RP11-357H14.17 acts as an oncogene by promoting cell proliferation and invasion in diffuse-type gastric cancer. Onco Targets Ther 10:2635–2643 Cui M, You L, Ren X et al, 2016, Long non-coding RNA PVT1 and cancer. Biochem Biophys Res Commun 471:10–14 Guo K,Yao J,Yu Q et al, 2017, The expression pattern of long noncoding RNA PVT1 in tumor tissues and in extracellular vesicles of colorectal cancer correlates with cancer progression. Tumour Biol 39:1010428317699122 Wu BQ, Jiang Y, Zhu F et al, 2017, Long noncoding RNA PVT1 promotes EMT and cell proliferation and migration through downregulating p21 in pancreatic Cancer cells. Technol Cancer Res Treat 1:1533034617700559 Guan Y, Kuo WL, Stilwell JL et al, 2007, Amplification of PVT1 contributes to the pathophysiology of ovarian and breast cancer. Clin Cancer Res 13:5745–5755 Huang T, Liu HW, Chen JQ et al, 2017, The long noncoding RNA PVT1 functions as a competing endogenous RNA by sponging miR-186 in gastric cancer. Biomed Pharmacother 88:302–308 Huang C, Liu S, Wang H et al, 2016, LncRNA PVT1 overexpression is a poor prognostic biomarker and regulates migration and invasion in small cell lung cancer. Am J Transl Res 8:5025–5034 Zhuang C, Li J, Liu Y et al, 2015, Tetracycline-inducible shRNA targeting long non-coding RNA PVT1 inhibits cell growth and induces apoptosis in bladder cancer cells. Oncotarget 6:41194– 41203 Zheng X, Hu H, Li S, 2016, High expression of lncRNA PVT1 promotes invasion by inducing epithelial-to-mesenchymal transition in esophageal cancer. Oncol Lett 12:2357–2362 Iden M, Fye S, Li K et al, 2016, The lncRNA PVT1 contributes to the cervical Cancer phenotype and associates with poor patient prognosis. PLoS One 11 Cui D, Yu CH, Liu Met al, 2016, Long non-coding RNA PVT1 as a novel biomarker for diagnosis and prognosis of non-small cell lung cancer. Tumour Biol 37:4127–4134 Wan L, Sun M, Liu GJ et al, 2016, Long noncoding RNA PVT1 promotes non-small cell lung Cancer cell proliferation through epigenetically regulating LATS2 expression. Mol Cancer Ther 15: 1082–1094 Yang YR, Zang SZ, Zhong CL et al, 2014, Increased expression of the lncRNA PVT1 promotes tumorigenesis in non-small cell lung cancer. Int J Clin Exp Pathol 7:6929–6935 Ding J, Li D, Gong M et al, 2014, Expression and clinical significance of the long non-coding RNA PVT1 in human gastric cancer. Onco Targets Ther 7:1625–1630 Huang C, Yu W, Wang Q et al, 2015, Increased expression of the lncRNA PVT1 is associated with poor prognosis in pancreatic cancer patients. Minerva Med 106:143–149 Kong R, Zhang EB, Yin DD et al, 2015, Long noncoding RNA PVT1 indicates a poor prognosis of gastric cancer and promotes cell proliferation through epigenetically regulating p15 and p16. Mol Cancer 14:015–0355 Ren XX, Xiao YB, Zhang L et al, 2016, Expression and clinical significance of PVT1 gene in blood of the patients with gastric cancer. Chin J Cancer Biother 23:4 Takahashi Y, Sawada G, Kurashige J et al, 2014, Amplification of PVT-1 is involved in poor prognosis via apoptosis inhibition in colorectal cancers. Br J Cancer 110:164–171 Wang F, Yuan JH,Wang SB et al, 2014, Oncofetal long noncoding RNA PVT1 promotes proliferation and stem cell-like property of hepatocellular carcinoma cells by stabilizing NOP2. Hepatology 60:1278–1290 Yuan CL, Li H, Zhu L et al, 2016, Aberrant expression of long noncoding RNA PVT1 and its diagnostic and prognostic significance in patients with gastric cancer. Neoplasma 63:442–449 Huang Y, Mu ZY, Lin HL et al, 2015, Expression and clinical significance of lncRNA PTV1 in renal carcinoma tissues. Shandong Med 55:3 Ding C, Yang Z, Lv Z et al, 2015, Long non-coding RNA PVT1 is associated with tumor progression and predicts recurrence in hepatocellular carcinoma patients. Oncol Lett 9:955–963 XuMD,Wang Y,WengWet al, 2017, A positive feedback loop of lncRNA-PVT1 and FOXM1 facilitates gastric Cancer growth and invasion. Clin Cancer Res 23:2071–2080 Martini P, Paracchini L, Caratti G et al, 2017, lncRNAs as novel indicators of Patients' prognosis in stage I epithelial ovarian Cancer: a retrospective and multicentric study. Clin Cancer Res 23:2356–2366 Zhao L, Kong H, Sun H et al, 2017, LncRNA-PVT1 promotes pancreatic cancer cells proliferation and migration through acting as a molecular sponge to regulate miR-448. J Cell Physiol 28:26072 Lan T, Yan X, Li Z et al, 2017, Long non-coding RNA PVT1 serves as a competing endogenous RNA for miR-186-5p to promote the tumorigenesis and metastasis of hepatocellular carcinoma. Tumour Biol 39:1010428317705338 Song J, Wu X, Liu F et al, 2017, Long non-coding RNA PVT1 promotes glycolysis and tumor progression by regulating miR-497/ HK2 axis in osteosarcoma. Biochem Biophys Res Commun 490: 217–224 Wu D, Li Y, Zhang H et al, 2017, Knockdown of Lncrna PVT1 enhances Radiosensitivity in non-small cell lung Cancer by sponging Mir-195. Cell Physiol Biochem 42:2453–2466 Gou X, Zhao X, Wang Z, 2017, Long noncoding RNA PVT1 promotes hepatocellular carcinoma progression through regulating miR-214. Cancer Biomark 31 CBM-170331 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 837 EP 847 DI 10.1007/s12253-018-0451-3 PG 11 ER PT J AU Ren, HY Shen, JX Mao, XM Zhang, XY Zhou, P Li, SY Zheng, ZW Shen, DY Meng, JR AF Ren, Hong-Yue Shen, Jin-Xing Mao, Xiao-Mei Zhang, Xiao-Yun Zhou, Pan Li, Si-Yang Zheng, Zhi-Wei Shen, Dong-Yan Meng, Jia-Rong TI Correlation Between Tumor Vasculogenic Mimicry and Poor Prognosis of Human Digestive Cancer Patients: A Systematic Review and Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Vasculogenic mimicry; digestive cancer; prognosis; meta-analysis ID Vasculogenic mimicry; digestive cancer; prognosis; meta-analysis AB Vasculogenic mimicry (VM) is a new pattern of blood supplement independent of endothelial vessels, which is related with tumor invasion, metastasis and prognosis. However, the role of VM in the prognosis of cancer patients is controversial. This study aimed to perform a meta-analysis of the published data to attempt to clarify the prognostic value of VM in the digestive cancer. Relevant studies were retrieved from the PubMed, Web of Science, Cochrane Library, Chinese National Knowledge Infrastructure and VIP databases published before March 29, 2018. Studies were included if they detected VM in the digestive cancer and analyzed the overall survival (OS) or disease-free survival (DFS) according to VM status. Two independent reviewers screened the studies, extracted data, and evaluated the quality of included studies with the Newcastle-Ottawa scale. Meta-analysis was performed using STATA 12.0 software. A total of 22 studies with 2411 patients were included in this meta-analysis. Meta-analysis showed that VM was related with the poor OS (HR = 2.30, 95% CI: 2.06–2.56, P < 0.001) and DFS (HR = 2.60, 95% CI: 2.07–3.27, P < 0.001) of patients with digestive cancer. Subgroup analysis showed VM was related with tumor differentiation, lymph node metastasis and TNM stage. Moreover, the present meta-analysis was reliable, and there was no obvious publication bias. This meta-analysis suggested that VM was a poor prognosis of digestive cancer patients. Further large and well-designed studies are required. C1 [Ren, Hong-Yue] The Affiliated Southeast Hospital of Xiamen University, Department of Pathology, 363000 Zhangzhou, Fujian Province, China. [Shen, Jin-Xing] The First Affiliated Hospital of Xiamen University, Biobank, No 55 Zhenhai Road, 361003 Xiamen, Fujian Province, China. [Mao, Xiao-Mei] The First Affiliated Hospital of Xiamen University, Biobank, No 55 Zhenhai Road, 361003 Xiamen, Fujian Province, China. [Zhang, Xiao-Yun] The First Affiliated Hospital of Xiamen University, Biobank, No 55 Zhenhai Road, 361003 Xiamen, Fujian Province, China. [Zhou, Pan] The First Affiliated Hospital of Xiamen University, Biobank, No 55 Zhenhai Road, 361003 Xiamen, Fujian Province, China. [Li, Si-Yang] The First Affiliated Hospital of Xiamen University, Biobank, No 55 Zhenhai Road, 361003 Xiamen, Fujian Province, China. [Zheng, Zhi-Wei] The First Affiliated Hospital of Xiamen University, Biobank, No 55 Zhenhai Road, 361003 Xiamen, Fujian Province, China. [Shen, Dong-Yan] The First Affiliated Hospital of Xiamen University, Biobank, No 55 Zhenhai Road, 361003 Xiamen, Fujian Province, China. [Meng, Jia-Rong] The Affiliated Southeast Hospital of Xiamen University, Department of Pathology, 363000 Zhangzhou, Fujian Province, China. RP Meng, JR (reprint author), The Affiliated Southeast Hospital of Xiamen University, Department of Pathology, 363000 Zhangzhou, China. EM mengjiarong175@163.com CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108 Jemal A, Siegel R, Xu J,Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60(5):277–300 Qiao L, Liang N, Zhang J, Xie J, Liu F, Xu D, Yu X, Tian Y, 2015, Advanced research on vasculogenic mimicry in cancer. J Cell Mol Med 19(2):315–326 Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe'er J, Trent JM, Meltzer PS, HendrixMJ, 1999, Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 155(3):739–752 Williamson SC, Metcalf RL, Trapani F, Mohan S, Antonello J, Abbott B, Leong HS, Chester CP, Simms N, Polanski R, Nonaka D, Priest L, Fusi A, Carlsson F, Carlsson A, HendrixMJ, Seftor RE, Seftor EA, Rothwell DG, Hughes A, Hicks J, Miller C, Kuhn P, Brady G, Simpson KL, Blackhall FH, Dive C, 2016, Vasculogenic mimicry in small cell lung cancer. Nat Commun 7:13322 Guo Q, Yuan Y, Jin Z, Xu T, Gao Y, Wei H, Li C, Hou W, Hua B, 2016, Association between Tumor Vasculogenic Mimicry and the Poor Prognosis of Gastric Cancer in China: An Updated Systematic Review and Meta-Analysis. Biomed Res Int 2016:2408645 Baeten CI, Hillen F, Pauwels P, de Bruine AP, Baeten CG, 2009, Prognostic role of vasculogenic mimicry in colorectal cancer. Dis Colon Rectum 52(12):2028–2035 Shen Y, Quan J, Wang M, Li S, Yang J, Lv M, Chen Z, Zhang L, Zhao X, Yang J, 2017, Tumor vasculogenic mimicry formation as an unfavorable prognostic indicator in patients with breast cancer. Oncotarget 8(34):56408–56416 Shirakawa K, Wakasugi H, Heike Y, Watanabe I, Yamada S, Saito K, Konishi F, 2002, Vasculogenic mimicry and pseudo-comedo formation in breast cancer. Int J Cancer 99(6):821–828 Yang JP, Liao YD, Mai DM, Xie P, Qiang YY, Zheng LS, Wang MY,Mei Y, Meng DF, Xu L, Cao L, Yang Q, Yang XX,WangWB, Peng LX, Huang BJ, Qian CN, 2016, Tumor vasculogenicmimicry predicts poor prognosis in cancer patients: a meta-analysis. Angiogenesis 19(2):191–200 Sun B, Zhang S, Zhao X, Zhang W, Hao X, 2004, Vasculogenic mimicry is associated with poor survival in patients with mesothelial sarcomas and alveolar rhabdomyosarcomas. Int J Oncol 25(6): 1609–1614 van Beurden A, Schmitz RF, van Dijk CM, Baeten CI, 2012, Periodic acid Schiff loops and blood lakes associated with metastasis in cutaneous melanoma. Melanoma Res 22(6):424–429 Knobloch K,Yoon U, Vogt PM(2011, Preferred reporting items for systematic reviews and meta-analyses, PRISMA, statement and publication bias. J Craniomaxillofac Surg 39(2):91–92 Moher D, Liberati A, Tetzlaff J, Altman DG, 2009, Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. J Clin Epidemiol 62(10):1006–1012 Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, McQuay HJ, 1996, Assessing the quality of reports of randomized clinical trials: is blinding necessary? Control Clin Trials 17(1):1–12 Chai DM, Bao ZQ, Hu JG, Ma L, Feng ZZ, Tao YS, 2013, Vasculogenic mimicry and aberrant expression of HIF-lalpha/Ecad are associated with worse prognosis of esophageal squamous cell carcinoma. J Huazhong Univ Sci Technolog Med Sci 33(3): 385–391 Guzman G, Cotler SJ, Lin AY, Maniotis AJ, Folberg R, 2007, A pilot study of vasculogenic mimicry immunohistochemical expression in hepatocellular carcinoma. Arch Pathol Lab Med 131(12): 1776–1781 Li M, Gu Y, Zhang Z, Zhang S, Zhang D, Saleem AF, Zhao X, Sun B, 2010, Vasculogenic mimicry: a new prognostic sign of gastric adenocarcinoma. Pathol Oncol Res 16(2):259–266 Li J, Ma XJ, Du XB, Feng G, Tan BX, 2014, The influence of vasculogenic mimicry expression onprognosis of patients with primary liver cancer. Pract J Clin Med 11(02):102–106 Li J, 2016, The role of Cyclooxygenase-2 in gastic cancer vasculogenic mimicry and its molecular mechanism. Chinese master’s dissertation of Tianjin medical university Liao SP, Gao Q, 2013, Expressions and clinical significance of vasculogenic mimicry and relatedprotein Mig-7 and MMP-2 in gastric carcinoma. J Cell Molecular Immunol 29(02):194–196 LiuWB, Xu GL, JiaWD, Li JS, Ma JL, Chen K,Wang ZH, Ge YS, RenWH, Yu JH,WangW,Wang XJ, 2011, Prognostic significance and mechanisms of patterned matrix vasculogenic mimicry in hepatocellular carcinoma. Med Oncol 28(Suppl 1):S228–S238 Lv J, Sun B, Sun H, ZhangY, Sun J, Zhao X, Gu Q, DongX, Che N, 2017, Significance of Vasculogenic Mimicry Formation in Gastric Carcinoma. Oncol Res Treat 40(1–2):35–41 Shao B, 2016, Correlation between the overexpression of LOXL2 and the metastasis and prognosic value in hepatocellar carcinoma. J Clin Exp Pathol 32(2):123–126 Song WQ, Yu L, Wu SW, Tao YS, 2013, Expression of vascular endothelial growth factor and its correlation with prognosis in esophageal squamous cell carcinoma. Chin J Gerontol 23:5855–5857 Song YY, Sun LD, Liu ML, Liu ZL, Chen F, Zhang YZ, Zheng Y, Zhang JP, 2014, STAT3, p-STAT3 and HIF-1alpha are associated with vasculogenic mimicry and impact on survival in gastric adenocarcinoma. Oncol Lett 8(1):431–437 Sun B, Zhang S, Zhang D, Du J, Guo H, Zhao X, ZhangW, Hao X, 2006, Vasculogenic mimicry is associated with high tumor grade, invasion and metastasis, and short survival in patients with hepatocellular carcinoma. Oncol Rep 16(4):693–698 SunW, Shen ZY, Zhang H, Fan YZ, ZhangWZ, Zhang JT, Lu XS, Ye C, 2012, Overexpression of HIF-1 alpha in primary gallbladder carcinoma and its relation to vasculogenic mimicry and unfavourable prognosis. Oncol Rep 27(6):1990–2002 Wang L, Lin L, Chen X, Sun L, Liao YL, Huang N, LiaoWJ, 2015, Metastasis-associated in colon cancer-1 promotes vasculogenic mimicry in gastric cancer by upregulating TWIST1/2. Oncotarget 6(13):11492–11506 Yang Z, 2011, Preliminary study on the mimicry of angiogenesis in gastric cancer.. Chinese master’s dissertation of university of south China Yang ZH, Sun BC, Li YL, Zhao XL, ZhaoXM, Gu Q, An JD,Dong XY, Liu F, Wang Y, 2015, ZEB2 promotes vasculogenic mimicry by TGF-beta 1 induced epithelial-to-mesenchymal transition in hepatocellular carcinoma. Exp Mol Pathol 98(3):352–359 Yang J, Zhu DM, Zhou XG, Yin N, Zhang Y, Zhang ZX, Li DC, Zhou J, 2017, HIF-2 alpha promotes the formation of vasculogenic mimicry in pancreatic cancer by regulating the binding of Twist1 to the VE-cadherin promoter. Oncotarget 8(29):47801–47815 Zhang JX, Zhang GX, Hu PP, Deng GD, Liu QQ, Qiao LL, Luo H, Zhang JD, 2017, Vasculogenic mimicry is associated with increased tumor-infiltrating neutrophil and poor outcome in esophageal squamous cell carcinoma. Onco Targets Ther 10:2923–2930 Zheng QH, Yang YH, Lin XF, 2012, A preliminary study of vasculogenic mimicry in human gallbladder carcinoma and its clinical significance. J Fujian Med Univ 46(4):248–250 Zhou L, Yu L, Feng ZZ, Gong XM, Cheng ZN, Yao N,Wang DN, Wu SW, 2015, Aberrant Expression of Markers of Cancer Stem Cells in Gastric Adenocarcinoma and their Relationship to VasculogenicMimicry. Asian Pac J Cancer Prev 16(10):4177–4183 Zhu B, Zhou L, Yu L, Wu S, Song W, Gong X, Wang D, 2017, Evaluation of the correlation of vasculogenic mimicry, ALDH1, KAI1 and microvessel density in the prediction of metastasis and prognosis in colorectal carcinoma. BMC Surg 17(1):47 Cassidy RJ, Liu Y, Patel K, Zhong J, Steuer CE, Kooby DA, Russell MC, Gillespie TW, Landry JC, 2017, Can we eliminate neoadjuvant chemoradiotherapy in favor of neoadjuvant multiagent chemotherapy for select stage II/III rectal adenocarcinomas: Analysis of the National Cancer Data base. Cancer 123(5):783–793 Delgado-Bellido D, Serrano-Saenz S, Fernandez-Cortes M, Oliver FJ, 2017, Vasculogenic mimicry signaling revisited: focus on nonvascular VE-cadherin. Mol Cancer 16(1):65 Li S, Meng W, Guan Z, Guo Y, Han X, 2016, The hypoxia-related signaling pathways of vasculogenic mimicry in tumor treatment. Biomed Pharmacother 80:127–135 SunW, Shen ZY, Zhang H, Fan YZ, ZhangWZ, Zhang JT, Lu XS, Ye C, 2012, Overexpression of HIF-1alpha in primary gallbladder carcinoma and its relation to vasculogenic mimicry and unfavourable prognosis. Oncol Rep 27(6):1990–2002 Du J, Sun B, Zhao X, Gu Q, Dong X,Mo J, Sun T,Wang J, Sun R, Liu Y, 2014, Hypoxia promotes vasculogenic mimicry formation by inducing epithelial-mesenchymal transition in ovarian carcinoma. Gynecol Oncol 133(3):575–583 Hendrix MJ, Seftor EA, Meltzer PS, Gardner LM, Hess AR, Kirschmann DA, Schatteman GC, Seftor RE, 2001, Expression and functional significance of VE-cadherin in aggressive human melanoma cells: role in vasculogenic mimicry. Proc Natl Acad Sci U S A 98(14):8018–8023 Wang H, Lin H, Pan J, Mo C, Zhang F, Huang B,Wang Z, Chen X, Zhuang J,Wang D, Qiu S, 2016, VasculogenicMimicry in Prostate Cancer: The Roles of EphA2 and PI3K. J Cancer 7(9):1114–1124 Hao X, Sun B, Zhang S, Zhao X, 2002, Microarray study of vasculogenic mimicry in bi-directional differentiation malignant tumor. Zhonghua Yi Xue Za Zhi 82(19):1298–1302 Stacker SA, Achen MG, 2013, The VEGF signaling pathway in cancer: the road ahead. Chin J Cancer 32(6):297–302 Sun B, Zhang D, Zhao N, Zhao X, 2017, Epithelial-to-endothelial transition and cancer stem cells: two cornerstones of vasculogenic mimicry in malignant tumors. Oncotarget 8(18):30502–30510 Liu T, Sun B, Zhao X, Li Y, Zhao X, Liu Y, Yao Z, Gu Q, Dong X, Shao B, Lin X, Liu F, An J, 2015, USP44+ Cancer Stem Cell Subclones Contribute to Breast Cancer Aggressiveness by Promoting Vasculogenic Mimicry. Mol Cancer Ther 14(9):2121–2131 Wu S, Yu L, Wang D, Zhou L, Cheng Z, Chai D, Ma L, Tao Y, 2012, Aberrant expression of CD133 in non-small cell lung cancer and its relationship to vasculogenic mimicry. BMC Cancer 12:535 Liu Y, Sun B, Liu T, Zhao X, Wang X, Li Y, Meng J, Gu Q, Liu F, Dong X, Liu P, Sun R, Zhao N, 2016, Function of AURKA protein kinase in the formation of vasculogenic mimicry in triple-negative breast cancer stem cells. OncoTargets and therapy 9:3473–3484 Wu HB, Yang S,Weng HY, Chen Q, Zhao XL, Fu WJ, Niu Q, Ping YF, Wang JM, Zhang X, Yao XH, Bian XW, 2017, Autophagyinduced KDR/VEGFR-2 activation promotes the formation of vasculogenic mimicry by glioma stem cells. Autophagy 13(9): 1528–1542 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 849 EP 858 DI 10.1007/s12253-018-0496-3 PG 10 ER PT J AU Al-Rugeebah, A Alanazi, M Parine, RN AF Al-Rugeebah, Arwa Alanazi, Mohammed Parine, Reddy Narasimha TI MEG3: an Oncogenic Long Non-coding RNA in Different Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE MEG3; lncRNA; p53; Cancer; Methylation; Angiogenesis ID MEG3; lncRNA; p53; Cancer; Methylation; Angiogenesis AB Long noncoding RNAs (lncRNAs) have recently considered as central regulators in diverse biological processes and emerged as vital players controlling tumorigenesis. Several lncRNAs can be classified into oncogenes and tumor suppressor genes depending on their function in cancer. A maternally expressed gene 3 (MEG3) gene transcripts a 1.6 kb lncRNA whose act as an antitumor component in different cancer cells, such as breast, liver, glioma, colorectal, cervical, gastric, lung, ovarian and osteosarcoma cancer cells. The present review highlights biological function of MEG3 to repress tumor through regulating the major tumor suppressor genes p53 and Rb, inhibiting angiogenesis-related factor, or controlling miRNAs. On the other hand, previous studies have also suggested that MEG3 mediates epithelial-mesenchymal transition (EMT). However, deregulation of MEG3 is associated with the development and progression of cancer, suggesting that MEG3 may function as a potential biomarker and therapeutic target for human cancers. C1 [Al-Rugeebah, Arwa] King Saud University, College of Science, Genome Research ChairRiyadh, Saudi Arabia. [Alanazi, Mohammed] King Saud University, College of Science, Genome Research ChairRiyadh, Saudi Arabia. [Parine, Reddy Narasimha] King Saud University, College of Science, Genome Research ChairRiyadh, Saudi Arabia. RP Parine, RN (reprint author), King Saud University, College of Science, Genome Research Chair, Riyadh, Saudi Arabia. EM reddyparine@gmail.com CR Prensner JR, Iyer MK, Balbin OA, Dhanasekaran SM, Cao Q, Brenner JC, Laxman B, Asangani IA, Grasso CS, Kominsky HD, Cao X, Jing X, Wang X, Siddiqui J, Wei JT, Robinson D, Iyer HK, Palanisamy N, Maher CA, Chinnaiyan AM, 2011, Transcriptome sequencing across a prostate cancer cohort identifies PCAT-1, an unannotated lincRNA implicated in disease progression. Nat Biotechnol 29(8):742–749. , DOI 10. 1038/nbt.1914 Ren S, Peng Z, Mao JH, Yu Y, Yin C, Gao X, Cui Z, Zhang J, Yi K, XuW, Chen C, Wang F, Guo X, Lu J, Yang J,Wei M, Tian Z, Guan Y, Tang L, Xu C,Wang L, Gao X, TianW,Wang J, Yang H, Wang J, Sun Y, 2012, RNA-seq analysis of prostate cancer in the Chinese population identifies recurrent gene fusions, cancerassociated long noncoding RNAs and aberrant alternative splicings. Cell Res 22(5):806–821. , DOI 10.1038/cr.2012.30 Tsai MC, Spitale RC, Chang HY, 2011, Long intergenic noncoding RNAs: new links in cancer progression. Cancer Res 71(1):3– 7. , DOI 10.1158/0008-5472.CAN-10-2483 HuarteM, Rinn JL, 2010, Large non-coding RNAs: missing links in cancer? Hum Mol Genet 19(R2):R152–R161. , DOI 10.1093/hmg/ddq353 Gutschner T, Diederichs S, 2012, The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol 9(6):703–719. https:// doi.org/10.4161/rna.20481 Brunner AL, Beck AH, Edris B, Sweeney RT, Zhu SX, Li R, Montgomery K, Varma S, Gilks T, Guo X, Foley JW, Witten DM, Giacomini CP, Flynn RA, Pollack JR, Tibshirani R, Chang HY, van de Rijn M, West RB, 2012, Transcriptional profiling of long non-coding RNAs and novel transcribed regions across a diverse panel of archived human cancers. Genome Biol 13(8): R75. , DOI 10.1186/gb-2012-13-8-r75 Martens-Uzunova ES, Bottcher R, Croce CM, Jenster G, Visakorpi T, Calin GA, 2014, Long noncoding RNA in prostate, bladder, and kidney cancer. Eur Urol 65(6):1140–1151. https:// doi.org/10.1016/j.eururo.2013.12.003 Schmitt AM, Chang HY, 2016, Long noncoding RNAs in Cancer pathways. Cancer Cell 29(4):452–463. , DOI 10.1016/j. ccell.2016.03.010 Zhou Y, Zhang X, Klibanski A, 2012, MEG3 noncoding RNA: a tumor suppressor. J Mol Endocrinol 48(3):R45–R53. https://doi. org/10.1530/JME-12-0008 Wylie AA, Murphy SK, Orton TC, Jirtle RL, 2000, Novel imprinted DLK1/GTL2 domain on human chromosome 14 contains motifs that mimic those implicated in IGF2/H19 regulation. Genome Res 10(11):1711–1718 Zhang X, Rice K,Wang Y, ChenW, Zhong Y, Nakayama Y, Zhou Y, Klibanski A, 2010)Maternally expressed gene 3, MEG3, noncoding ribonucleic acid: isoform structure, expression, and functions. Endocrinology 151(3):939–947. , DOI 10.1210/en. 2009-0657 Miyoshi N, Wagatsuma H, Wakana S, Shiroishi T, Nomura M, Aisaka K, Kohda T, SuraniMA, Kaneko-Ishino T, Ishino F, 2000, Identification of an imprinted gene, Meg3/Gtl2 and its human homologue MEG3, first mapped on mouse distal chromosome 12 and human chromosome 14q. Genes Cells 5(3):211–220 Zhang X, Zhou Y, Mehta KR, Danila DC, Scolavino S, Johnson SR, Klibanski A, 2003, A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor cells. J Clin Endocrinol Metab 88(11):5119–5126. , DOI 10.1210/jc.2003- 030222 Tan MC, Widagdo J, Chau YQ, Zhu T, Wong JJ, Cheung A, Anggono V, 2017, The activity-induced Long non-coding RNA Meg3 modulates AMPA receptor surface expression in primary cortical neurons. Front Cell Neurosci 11:124. , DOI 10. 3389/fncel.2017.00124 Murphy SK,Wylie AA, Coveler KJ, Cotter PD, Papenhausen PR, Sutton VR, Shaffer LG, Jirtle RL, 2003, Epigenetic detection of human chromosome 14 uniparental disomy. Hum Mutat 22(1): 92–97. , DOI 10.1002/humu.10237 Zhao J, Zhang X, Zhou Y, Ansell PJ, Klibanski A, 2006, Cyclic AMP stimulates MEG3 gene expression in cells through a cAMPresponse element, CRE, in the MEG3 proximal promoter region. Int J Biochem Cell Biol 38(10):1808–1820. , DOI 10. 1016/j.biocel.2006.05.004 da Rocha ST, Edwards CA, Ito M, Ogata T, Ferguson-Smith AC, 2008, Genomic imprinting at the mammalian Dlk1-Dio3 domain. Trends Genet 24(6):306–316. , DOI 10.1016/j.tig.2008. 03.011 Benetatos L, Vartholomatos G, Hatzimichael E, 2011, MEG3 imprinted gene contribution in tumorigenesis. Int J Cancer 129(4):773–779. , DOI 10.1002/ijc.26052 Wallace C, Smyth DJ, Maisuria-Armer M,Walker NM, Todd JA, Clayton DG, 2010, The imprinted DLK1-MEG3 gene region on chromosome 14q32.2 alters susceptibility to type 1 diabetes. Nat Genet 42(1):68–71. , DOI 10.1038/ng.493 Balik V, Srovnal J, Sulla I, Kalita O, Foltanova T, Vaverka M, Hrabalek L, Hajduch M, 2013, MEG3: a novel long noncoding potentially tumour-suppressing RNA in meningiomas. J Neuro- Oncol 112(1):1–8. , DOI 10.1007/s11060-012-1038-6 Zhou Y, Zhong Y, Wang Y, Zhang X, Batista DL, Gejman R, Ansell PJ, Zhao J, Weng C, Klibanski A, 2007, Activation of p53 by MEG3 non-coding RNA. J Biol Chem 282(34):24731– 24742. , DOI 10.1074/jbc.M702029200 Vogelstein B, Lane D, Levine AJ, 2000, Surfing the p53 network. Nature 408(6810):307–310. , DOI 10.1038/35042675 Vegran F, Rebucci M, Chevrier S, Cadouot M, Boidot R, Lizard- Nacol S, 2013, Only missense mutations affecting the DNA binding domain of p53 influence outcomes in patients with breast carcinoma. PLoS One 8(1):e55103. , DOI 10.1371/ journal.pone.0055103 Lane DP, 1992, Cancer. p53, guardian of the genome. Nature 358(6381):15–16. , DOI 10.1038/358015a0 Zilfou JT, Lowe SW, 2009, Tumor suppressive functions of p53. Cold Spring Harb Perspect Biol 1(5):a001883. , DOI 10. 1101/cshperspect.a001883 Manfredi JJ, 2010, The Mdm2-p53 relationship evolves: Mdm2 swings both ways as an oncogene and a tumor suppressor. Genes Dev 24(15):1580–1589. , DOI 10.1101/gad.1941710 Yee KS, Vousden KH(2005, Complicating the complexity of p53. Carcinogenesis 26(8):1317–1322. , DOI 10.1093/carcin/ bgi122 Riley T, Sontag E, Chen P, Levine A, 2008, Transcriptional control of human p53-regulated genes. Nat Rev Mol Cell Biol 9(5): 402–412. , DOI 10.1038/nrm2395 Green DR, Kroemer G, 2009, Cytoplasmic functions of the tumour suppressor p53. Nature 458(7242):1127–1130. https://doi. org/10.1038/nature07986 Haupt S, Haupt Y, 2017, P53 at the start of the 21st century: lessons from elephants. F1000Res 6:2041. , DOI 10. 12688/f1000research.12682.1 Hu D, Su C, JiangM, Shen Y, Shi A, Zhao F, Chen R, Shen Z, Bao J, Tang W, 2016, Fenofibrate inhibited pancreatic cancer cells proliferation via activation of p53 mediated by upregulation of LncRNA MEG3. Biochem Biophys Res Commun 471(2):290– 295. , DOI 10.1016/j.bbrc.2016.01.169 Liu G, Chen X, 2006, Regulation of the p53 transcriptional activity. J Cell Biochem 97(3):448–458. , DOI 10.1002/jcb. 20700 Meek DW, Anderson CW, 2009, Posttranslational modification of p53: cooperative integrators of function. Cold Spring Harb Perspect Biol 1(6):a000950. , DOI 10.1101/cshperspect. a000950 Kruse JP, Gu W, 2009, Modes of p53 regulation. Cell 137(4): 609–622. , DOI 10.1016/j.cell.2009.04.050 Lee JT, Gu W, 2010, The multiple levels of regulation by p53 ubiquitination. Cell Death Differ 17(1):86–92. , DOI 10. 1038/cdd.2009.77 Zhu J, Liu S, Ye F, Shen Y, Tie Y, Zhu J,Wei L, Jin Y, Fu H,Wu Y, Zheng X, 2015, Long noncoding RNA MEG3 interacts with p53 protein and regulates partial p53 target genes in hepatoma cells. PLoS One 10(10):e0139790. , DOI 10.1371/journal. pone.0139790 Banerjee S, Kumar BR, Kundu TK, 2004, General transcriptional coactivator PC4 activates p53 function. Mol Cell Biol 24(5): 2052–2062 Li N, Richard S, 2016, Sam68 functions as a transcriptional coactivator of the p53 tumor suppressor. Nucleic Acids Res 44(18): 8726–8741. , DOI 10.1093/nar/gkw582 Chene P, 2001, The role of tetramerization in p53 function. Oncogene 20(21):2611–2617. , DOI 10.1038/sj.onc. 1204373 Follis AV, Llambi F, Ou L, Baran K, Green DR, Kriwacki RW, 2014, The DNA-binding domain mediates both nuclear and cytosolic functions of p53. Nat Struct Mol Biol 21(6):535–543. , DOI 10.1038/nsmb.2829 Joerger AC, Fersht AR, 2007, Structure-function-rescue: the diverse nature of common p53 cancer mutants. Oncogene 26(15): 2226–2242. , DOI 10.1038/sj.onc.1210291 Petitjean A, Mathe E, Kato S, Ishioka C, Tavtigian SV, Hainaut P, Olivier M, 2007, Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat 28(6): 622–629. , DOI 10.1002/humu.20495 Levine AJ, 1997, p53, the cellular gatekeeper for growth and division. Cell 88(3):323–331 Huang SS, Huang JS, 2005, TGF-beta control of cell proliferation. J Cell Biochem 96(3):447–462. , DOI 10.1002/jcb. 20558 Corre J, Hebraud B, Bourin P, 2013, Concise review: growth differentiation factor 15 in pathology: a clinical role? Stem Cells Transl Med 2(12):946–952. , DOI 10.5966/sctm.2013- 0055 Wang P, Chen D, Ma H, Li Y, 2017, Long non-coding RNA MEG3 regulates proliferation and apoptosis in non-small cell lung cancer via the miR-205-5p/LRP1 pathway. RSC Adv 7(78): 49710–49719 Shi Y, Lv C, Shi L, Tu G, 2018, MEG3 inhibits proliferation and invasion and promotes apoptosis of human osteosarcoma cells. Oncol Lett 15(2):1917–1923. , DOI 10.3892/ol.2017. 7463 Lyu Y, Lou J, Yang Y, Feng J, Hao Y, Huang S, Yin L, Xu J, Huang D, Ma B, Zou D, Wang Y, Zhang Y, Zhang B, Chen P, Yu K, Lam EW,Wang X, Liu Q, Yan J, Jin B, 2017, Dysfunction of the WT1-MEG3 signaling promotes AML leukemogenesis via p53-dependent and -independent pathways. Leukemia 31(12): 2543–2551. , DOI 10.1038/leu.2017.116 Giacinti C, Giordano A, 2006, RB and cell cycle progression. Oncogene 25(38):5220–5227. , DOI 10.1038/sj.onc. 1209615 Zhang Z, Li M, Wang H, Agrawal S, Zhang R, 2003, Antisense therapy targeting MDM2 oncogene in prostate cancer: effects on proliferation, apoptosis, multiple gene expression, and chemotherapy. Proc Natl Acad Sci U S A 100(20):11636–11641. https://doi. org/10.1073/pnas.1934692100 Al-Khalaf HH, Lach B, Allam A, Hassounah M, Alkhani A, Elkum N, Alrokayan SA, Aboussekhra A, 2008, Expression of survivin and p16(INK4a)/Cdk6/pRB proteins and induction of apoptosis in response to radiation and cisplatin in meningioma cells. Brain Res 1188:25–34. , DOI 10.1016/j.brainres. 2007.10.074 He Y, Luo Y, Liang B, Ye L, Lu G, He W, 2017, Potential applications of MEG3 in cancer diagnosis and prognosis. Oncotarget 8(42):73282–73295. , DOI 10.18632/oncotarget.19931 Kumar MM, Goyal R, 2017, LncRNA as a therapeutic target for angiogenesis. Curr TopMed Chem 17(15):1750–1757. https://doi. org/10.2174/1568026617666161116144744 Li CY, Shan S, Huang Q, Braun RD, Lanzen J, Hu K, Lin P, Dewhirst MW, 2000, Initial stages of tumor cell-induced angiogenesis: evaluation via skin window chambers in rodent models. J Natl Cancer Inst 92(2):143–147 Johnson KE,Wilgus TA(2014, Vascular endothelial growth factor and angiogenesis in the regulation of cutaneous wound repair. Adv Wound Care, New Rochelle, 3(10):647–661. , DOI 10. 1089/wound.2013.0517 Lee SH, Lee S, Yang H, Song S, Kim K, Saunders TL, Yoon JK, Koh GY, Kim I, 2014, Notch pathway targets proangiogenic regulator Sox17 to restrict angiogenesis. Circ Res 115(2):215–226. , DOI 10.1161/CIRCRESAHA.115.303142 Zhan R, Xu K, Pan J, Xu Q, Xu S, Shen J, 2017, Long noncoding RNA MEG3 mediated angiogenesis after cerebral infarction through regulating p53/NOX4 axis. Biochem Biophys Res Commun 490(3):700–706. , DOI 10.1016/j.bbrc.2017. 06.104 Gordon FE, Nutt CL, Cheunsuchon P, Nakayama Y, Provencher KA, Rice KA, Zhou Y, Zhang X, Klibanski A, 2010, Increased expression of angiogenic genes in the brains of mouse meg3-null embryos. Endocrinology 151(6):2443–2452. , DOI 10. 1210/en.2009-1151 Su W, Xie W, Shang Q, Su B, 2015, The Long noncoding RNA MEG3 is downregulated and inversely associated with VEGF levels in osteoarthritis. Biomed Res Int 2015:356893. https://doi. org/10.1155/2015/356893 Zhang CY, Yu MS, Li X, Zhang Z, Han CR, Yan B, 2017, Overexpression of long non-coding RNA MEG3 suppresses breast cancer cell proliferation, invasion. and angiogenesis through AKT pathway Tumour Biol 39(6):1010428317701311. , DOI 10.1177/1010428317701311 Liu J, Li Q, Zhang KS, Hu B, Niu X, Zhou SM, Li SG, Luo YP, WangY, Deng ZF, 2017, Downregulation of the Long non-coding RNA Meg3 promotes angiogenesis after ischemic brain injury by activating notch signaling. Mol Neurobiol 54(10):8179–8190. , DOI 10.1007/s12035-016-0270-z Ruan W, Zhao F, Zhao S, Zhang L, Shi L, Pang T, 2018, Knockdown of long noncoding RNA MEG3 impairs VEGFstimulated endothelial sprouting angiogenesis via modulating VEGFR2 expression in human umbilical vein endothelial cells. Gene 649:32–39. , DOI 10.1016/j.gene.2018.01.072 He C, YangW, Yang J, Ding J, Li S,Wu H, Zhou F, Jiang Y, Teng L, Yang J, 2017, Long noncoding RNA MEG3 negatively regulates proliferation and angiogenesis in vascular endothelial cells. DNA Cell Biol 36(6):475–481. , DOI 10.1089/dna.2017. 3682 Sharma S, Kelly TK, Jones PA, 2010, Epigenetics in cancer. Carcinogenesis 31(1):27–36. , DOI 10.1093/carcin/ bgp220 Cheung HH, Lee TL, Rennert OM, ChanWY, 2009, DNA methylation of cancer genome. Birth Defects Res C Embryo Today 87(4):335–350. , DOI 10.1002/bdrc.20163 Deaton AM, Bird A, 2011, CpG islands and the regulation of transcription. Genes Dev 25(10):1010–1022. , DOI 10. 1101/gad.2037511 Delpu Y, Cordelier P, Cho WC, Torrisani J, 2013, DNA methylation and cancer diagnosis. Int J Mol Sci 14(7):15029–15058. , DOI 10.3390/ijms140715029 Gejman R, Batista DL, Zhong Y, Zhou Y, Zhang X, Swearingen B, Stratakis CA, Hedley-Whyte ET, Klibanski A, 2008, Selective loss of MEG3 expression and intergenic differentially methylated region hypermethylation in the MEG3/DLK1 locus in human clinically nonfunctioning pituitary adenomas. J Clin Endocrinol Metab 93(10):4119–4125. , DOI 10.1210/jc.2007-2633 Liu X, Gao Q, Li P, Zhao Q, Zhang J, Li J, Koseki H, Wong J, 2013, UHRF1 targets DNMT1 for DNA methylation through cooperative binding of hemi-methylated DNA and methylated H3K9. Nat Commun 4:1563 Lu KH, LiW, Liu XH, Sun M, ZhangML,WuWQ, Xie WP, Hou YY, 2013, Long non-coding RNA MEG3 inhibits NSCLC cells proliferation and induces apoptosis by affecting p53 expression. BMC Cancer 13:461. , DOI 10.1186/1471-2407-13-461 Zhuo H, Tang J, Lin Z, Jiang R, Zhang X, Ji J, Wang P, Sun B, 2016, The aberrant expression of MEG3 regulated by UHRF1 predicts the prognosis of hepatocellular carcinoma. Mol Carcinog 55(2):209–219. , DOI 10.1002/mc.22270 Li J, Bian EB, He XJ, Ma CC, Zong G,Wang HL, Zhao B, 2016, Epigenetic repression of long non-coding RNA MEG3 mediated by DNMT1 represses the p53 pathway in gliomas. Int J Oncol 48(2):723–733. , DOI 10.3892/ijo.2015.3285 Yu B, Lv X, Su L, Li J, Yu Y, Gu Q, Yan M, Zhu Z, Liu B, 2016, MiR-148a functions as a tumor suppressor by targeting CCK-BR via inactivating STAT3 and Akt in human gastric Cancer. PLoS One 11(8):e0158961. , DOI 10.1371/journal.pone. 0158961 Shamma A, Suzuki M, Hayashi N, Kobayashi M, Sasaki N, Nishiuchi T, Doki Y, Okamoto T, Kohno S, Muranaka H, Kitajima S, Yamamoto K, Takahashi C, 2013, ATM mediates pRB function to control DNMT1 protein stability and DNA methylation. Mol Cell Biol 33(16):3113–3124. , DOI 10.1128/ MCB.01597-12 Kruer TL, Dougherty SM, Reynolds L, Long E, de Silva T, Lockwood WW, Clem BF, 2016, Expression of the lncRNA maternally expressed gene 3, MEG3, contributes to the control of lung Cancer cell proliferation by the Rb pathway. PLoS One 11(11):e0166363. , DOI 10.1371/journal.pone.0166363 Li Y, Ren M, Zhao Y, Lu X, Wang M, Hu J, Lu G, He S, 2017, MicroRNA-26a inhibits proliferation and metastasis of human hepatocellular carcinoma by regulating DNMT3B-MEG3 axis. Oncol Rep 37(6):3527–3535. , DOI 10.3892/or.2017. 5579 Gao Y, Huang P, Zhang J, 2017, Hypermethylation of MEG3 promoter correlates with inactivation of MEG3 and poor prognosis in patients with retinoblastoma. J Transl Med 15(1):268. , DOI 10.1186/s12967-017-1372-8 Polyak K, 2011, Heterogeneity in breast cancer. J Clin Invest 121(10):3786–3788. , DOI 10.1172/JCI60534 Padua Alves C, Fonseca AS, Muys BR, de Barros ELBR, Burger MC, de Souza JE, Valente V, Zago MA, SilvaWA Jr, 2013, Brief report: the lincRNA Hotair is required for epithelial-tomesenchymal transition and stemness maintenance of cancer cell lines. Stem Cells 31(12):2827–2832. , DOI 10.1002/ stem.1547 Hou P, Zhao Y, Li Z, Yao R, Ma M, Gao Y, Zhao L, Zhang Y, Huang B, Lu J, 2014, LincRNA-ROR induces epithelial-tomesenchymal transition and contributes to breast cancer tumorigenesis and metastasis. Cell Death Dis 5:e1287. , DOI 10. 1038/cddis.2014.249 Li H, Zhu L, Xu L, Qin K, Liu C, Yu Y, Su D, Wu K, Sheng Y, 2017, Long noncoding RNA linc00617 exhibits oncogenic activity in breast cancer. Mol Carcinog 56(1):3–17. , DOI 10. 1002/mc.22338 Shahryari A, Jazi MS, Samaei NM, Mowla SJ, 2015, Long noncoding RNA SOX2OT: expression signature, splicing patterns, and emerging roles in pluripotency and tumorigenesis. Front Genet 6:196. , DOI 10.3389/fgene.2015.00196 Redis RS, Sieuwerts AM, LookMP, Tudoran O, Ivan C, Spizzo R, Zhang X, deWeerd V, Shimizu M, Ling H, Buiga R, Pop V, Irimie A, Fodde R, Bedrosian I, Martens JW, Foekens JA, Berindan- Neagoe I, Calin GA, 2013, CCAT2, a novel long non-coding RNA in breast cancer: expression study and clinical correlations. Oncotarget 4(10):1748–1762. , DOI 10.18632/ oncotarget.1292 Mourtada-Maarabouni M, Pickard MR, Hedge VL, Farzaneh F, Williams GT, 2009, GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene 28(2): 195–208. , DOI 10.1038/onc.2008.373 Zhao Z, Chen C, Liu Y, Wu C, 2014, 17beta-estradiol treatment inhibits breast cell proliferation, migration and invasion by decreasing MALAT-1 RNA level. Biochem Biophys Res Commun 445(2):388–393. , DOI 10.1016/j.bbrc.2014.02.006 Zhang JJ, Guo SH, Jia BQ, 2016, Down-regulation of long noncoding RNA MEG3 serves as an unfavorable risk factor for survival of patients with breast cancer. Eur Rev Med Pharmacol Sci 20(24):5143–5147 Sun L, LiY,Yang B, 2016, Downregulated long non-coding RNA MEG3 in breast cancer regulates proliferation, migration and invasion by depending on p53's transcriptional activity. Biochem Biophys Res Commun 478(1):323–329. , DOI 10.1016/ j.bbrc.2016.05.031 Zhang W, Shi S, Jiang J, Li X, Lu H, Ren F, 2017, LncRNA MEG3 inhibits cell epithelial-mesenchymal transition by sponging miR-421 targeting E-cadherin in breast cancer. Biomed Pharmacother 91:312–319. , DOI 10.1016/j.biopha. 2017.04.085 Mondal T, Subhash S, Vaid R, Enroth S, Uday S, Reinius B, Mitra S,Mohammed A, James AR, Hoberg E, Moustakas A, Gyllensten U, Jones SJ, Gustafsson CM, Sims AH, Westerlund F, Gorab E, Kanduri C, 2015, MEG3 long noncoding RNA regulates the TGF-beta pathway genes through formation of RNA-DNA triplex structures. Nat Commun 6:7743. , DOI 10.1038/ ncomms8743 Laursen L, 2014, A preventable cancer. Nature 516(7529):S2–S3. , DOI 10.1038/516S2a Li T, Zheng Q, An J, Wu M, Li H, Gui X, Pu H, Lu D, 2016, SET1A cooperates with CUDR to promote liver Cancer growth and hepatocyte-like stem cell malignant transformation epigenetically. Mol Ther 24(2):261–275. , DOI 10.1038/mt.2015. 208 Li H, An J, Wu M, Zheng Q, Gui X, Li T, Pu H, Lu D, 2015, LncRNA HOTAIR promotes human liver cancer stem cell malignant growth through downregulation of SETD2. Oncotarget 6(29):27847–27864. , DOI 10.18632/oncotarget.4443 Wang Y, He L, Du Y, Zhu P, Huang G, Luo J, Yan X, Ye B, Li C, Xia P, Zhang G, Tian Y, Chen R, Fan Z, 2015, The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling. Cell Stem Cell 16(4):413–425. , DOI 10.1016/j.stem.2015.03.003 Wang X, Sun W, Shen W, Xia M, Chen C, Xiang D, Ning B, Cui X, Li H, Li X, Ding J, Wang H, 2016, Long non-coding RNA DILC regulates liver cancer stem cells via IL-6/STAT3 axis. J Hepatol 64(6):1283–1294. , DOI 10.1016/j.jhep.2016. 01.019 He JH, Han ZP, Liu JM, Zhou JB, Zou MX, Lv YB, Li YG, Cao MR, 2017, Overexpression of Long non-coding RNA MEG3 inhibits proliferation of hepatocellular carcinoma Huh7 cells via negative modulation of miRNA-664. J Cell Biochem 118(11): 3713–3721. , DOI 10.1002/jcb.26018 Zheng Q, Lin Z, Xu J, Lu Y,Meng Q,Wang C, Yang Y, Xin X, Li X, Pu H, 2018, Long noncoding RNA MEG3 suppresses liver cancer cells growth through inhibiting β-catenin by activating PKM2 and inactivating PTEN. Cell Death Dis 9(3):253 He Y, Wu YT, Huang C, Meng XM, Ma TT, Wu BM, Xu FY, Zhang L, LvXW, Li J, 2014, Inhibitory effects of long noncoding RNA MEG3 on hepatic stellate cells activation and liver fibrogenesis. Biochim Biophys Acta 1842(11):2204–2215. , DOI 10.1016/j.bbadis.2014.08.015 Thakkar JP, Dolecek TA, Horbinski C, Ostrom QT, Lightner DD, Barnholtz-Sloan JS, Villano JL, 2014, Epidemiologic and molecular prognostic review of glioblastoma. Cancer Epidemiol Biomark Prev 23(10):1985–1996. , DOI 10.1158/1055- 9965.EPI-14-0275 Galasso M, Dama P, Previati M, Sandhu S, Palatini J, Coppola V, Warner S, Sana ME, Zanella R, Abujarour R, Desponts C, Teitell MA, Garzon R, Calin G, Croce CM,Volinia S, 2014)Alarge scale expression study associates uc.283-plus lncRNA with pluripotent stem cells and human glioma. Genome Med 6(10):76. https://doi. org/10.1186/s13073-014-0076-4 Ellis BC, Molloy PL, Graham LD, 2012, CRNDE: a Long noncoding RNA involved in CanceR, neurobiology, and DEvelopment. Front Genet 3:270. , DOI 10.3389/fgene. 2012.00270 Yao Y, Ma J, Xue Y, Wang P, Li Z, Liu J, Chen L, Xi Z, Teng H, Wang Z, Li Z, Liu Y, 2015, Knockdown of long non-coding RNA XIST exerts tumor-suppressive functions in human glioblastoma stem cells by up-regulating miR-152. Cancer Lett 359(1):75–86. , DOI 10.1016/j.canlet.2014.12.051 Wang P, Ren Z, Sun P, 2012, Overexpression of the long noncoding RNA MEG3 impairs in vitro glioma cell proliferation. J Cell Biochem 113(6):1868–1874. , DOI 10.1002/jcb. 24055 Shang S, Hua F, Hu ZW, 2017, The regulation of beta-catenin activity and function in cancer: therapeutic opportunities. Oncotarget 8(20):33972–33989. , DOI 10.18632/ oncotarget.15687 Gong X, Huang M, 2017, Long non-coding RNA MEG3 promotes the proliferation of glioma cells through targeting Wnt/ beta-catenin signal pathway. Cancer Gene Ther 24(9):381–385. , DOI 10.1038/cgt.2017.32 Qin N, Tong GF, Sun LW, Xu XL, 2017, Long noncoding RNA MEG3 suppresses glioma cell proliferation, migration, and invasion by acting as a competing endogenous RNA of miR-19a. Oncol Res 25(9):1471–1478. , DOI 10.3727/ 096504017X14886689179993 Zhang L, Liang X, Li Y, 2017, Long non-coding RNA MEG3 inhibits cell growth of gliomas by targeting miR-93 and inactivating PI3K/AKT pathway. Oncol Rep 38(4):2408–2416. , DOI 10.3892/or.2017.5871 Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F, 2017, Global patterns and trends in colorectal cancer incidence and mortality. Gut 66(4):683–691. , DOI 10. 1136/gutjnl-2015-310912 Zhang Z, Zhou C, ChangY, Zhang Z, Hu Y, Zhang F, Lu Y, Zheng L, Zhang W, Li X, Li X, 2016, Long non-coding RNA CASC11 interacts with hnRNP-K and activates theWNT/beta-catenin pathway to promote growth and metastasis in colorectal cancer. Cancer Lett 376(1):62–73. , DOI 10.1016/j.canlet.2016.03.022 Sun L, Jiang C, Xu C, Xue H, Zhou H, Gu L, Liu Y, Xu Q, 2017, Down-regulation of long non-coding RNA RP11-708H21.4 is associated with poor prognosis for colorectal cancer and promotes tumorigenesis through regulating AKT/mTOR pathway. Oncotarget 8(17):27929–27942. , DOI 10.18632/ oncotarget.15846 Han P, Li JW, Zhang BM, Lv JC, Li YM, Gu XY, Yu ZW, Jia YH, Bai XF, Li L, Liu YL, Cui BB, 2017, The lncRNA CRNDE promotes colorectal cancer cell proliferation and chemoresistance via miR-181a-5p-mediated regulation of Wnt/beta-catenin signaling. Mol Cancer 16(1):9. , DOI 10.1186/s12943-017- 0583-1 Menigatti M, Staiano T, Manser CN, Bauerfeind P, Komljenovic A, Robinson M, Jiricny J, Buffoli F, Marra G, 2013, Epigenetic silencing of monoallelically methylated miRNA loci in precancerous colorectal lesions. Oncogenesis 2:e56. , DOI 10.1038/ oncsis.2013.21 Liang WC, Fu WM, Wong CW, Wang Y, Wang WM, Hu GX, Zhang L, Xiao LJ, Wan DC, Zhang JF, Waye MM, 2015, The lncRNA H19 promotes epithelial to mesenchymal transition by functioning as miRNA sponges in colorectal cancer. Oncotarget 6(26):22513–22525. , DOI 10.18632/oncotarget.4154 Qi P, Xu MD, Ni SJ, Shen XH, Wei P, Huang D, Tan C, Sheng WQ, Zhou XY, Du X, 2015, Down-regulation of ncRAN, a long non-coding RNA, contributes to colorectal cancer cell migration and invasion and predicts poor overall survival for colorectal cancer patients. Mol Carcinog 54(9):742–750. , DOI 10. 1002/mc.22137 Liu Q, Huang J, Zhou N, Zhang Z, Zhang A, Lu Z,Wu F,Mo YY, 2013, LncRNA loc285194 is a p53-regulated tumor suppressor. Nucleic Acids Res 41(9):4976–4987. , DOI 10.1093/nar/ gkt182 Yin DD, Liu ZJ, Zhang E, Kong R, Zhang ZH, Guo RH, 2015, Decreased expression of long noncoding RNA MEG3 affects cell proliferation and predicts a poor prognosis in patients with colorectal cancer. Tumour Biol 36(6):4851–4859. , DOI 10. 1007/s13277-015-3139-2 ZhuY, ChenP, GaoY,TaN,ZhangY, Cai J,ZhaoY,Liu S, Zheng J, 2018, MEG3 activated by vitamin D inhibits colorectal Cancer cells proliferation and migration via regulating Clusterin. EBioMedicine 30:148–157. , DOI 10.1016/j.ebiom. 2018.03.032 Li L, Shang J, Zhang Y, Liu S, Peng Y, Zhou Z, Pan H, Wang X, Chen L, Zhao Q, 2017)MEG3 is a prognostic factor for CRC and promotes chemosensitivity by enhancing oxaliplatin-induced cell apoptosis. Oncol Rep 38(3):1383–1392. , DOI 10.3892/ or.2017.5828 Cao X, Zhuang S, Hu Y, Xi L, Deng L, Sheng H, Shen W, 2016, Associations between polymorphisms of long non-coding RNA MEG3 and risk of colorectal cancer in Chinese. Oncotarget 7(14):19054–19059. , DOI 10.18632/oncotarget.7764 Forouzanfar MH, Foreman KJ, Delossantos AM, Lozano R, Lopez AD, Murray CJ, Naghavi M, 2011, Breast and cervical cancer in 187 countries between 1980 and 2010: a systematic analysis. Lancet 378(9801):1461–1484. , DOI 10.1016/ S0140-6736(11)61351-2 Huang L, Liao LM, Liu AW,Wu JB, Cheng XL, Lin JX, ZhengM, 2014, Overexpression of long noncoding RNA HOTAIR predicts a poor prognosis in patients with cervical cancer. Arch Gynecol Obstet 290(4):717–723. , DOI 10.1007/s00404-014- 3236-2 Cao S, Liu W, Li F, Zhao W, Qin C, 2014, Decreased expression of lncRNA GAS5 predicts a poor prognosis in cervical cancer. Int J Clin Exp Pathol 7(10):6776–6783 Liao LM, Sun XY, Liu AW,Wu JB, Cheng XL, Lin JX, Zheng M, Huang L, 2014, Low expression of long noncoding XLOC_010588 indicates a poor prognosis and promotes proliferation through upregulation of c-Myc in cervical cancer. Gynecol Oncol 133(3):616–623. , DOI 10.1016/j.ygyno.2014.03. 555 Jiang S, Wang HL, Yang J, 2015, Low expression of long noncoding RNA LET inhibits carcinogenesis of cervical cancer. Int J Clin Exp Pathol 8(1):806–811 Yang M, Zhai X, Xia B, Wang Y, Lou G, 2015, Long noncoding RNA CCHE1 promotes cervical cancer cell proliferation via upregulating PCNA. Tumour Biol 36(10):7615–7622. https://doi. org/10.1007/s13277-015-3465-4 Qin R, Chen Z, Ding Y, Hao J, Hu J, Guo F, 2013, Long noncoding RNA MEG3 inhibits the proliferation of cervical carcinoma cells through the induction of cell cycle arrest and apoptosis. Neoplasma 60(5):486–492 Zhang J, Yao T,Wang Y, Yu J, Liu Y, Lin Z, 2016, Long noncoding RNA MEG3 is downregulated in cervical cancer and affects cell proliferation and apoptosis by regulating miR-21. Cancer Biol Ther 17(1):104–113. , DOI 10.1080/15384047.2015. 1108496 Zhang J, Lin Z, Gao Y, Yao T, 2017, Downregulation of long noncoding RNA MEG3 is associated with poor prognosis and promoter hypermethylation in cervical cancer. J Exp Clin Cancer Res 36(1):5. , DOI 10.1186/s13046-016-0472-2 Zhang J, Yao T, Lin Z, Gao Y, 2017, Aberrant methylation of MEG3 functions as a potential plasma-based biomarker for cervical Cancer. Sci Rep 7(1):6271. , DOI 10.1038/s41598- 017-06502-7 Wang X,Wang Z,Wang J,Wang Y, Liu L, Xu X, 2017, LncRNA MEG3 has anti-activity effects of cervical cancer. Biomed Pharmacother 94:636–643. , DOI 10.1016/j.biopha. 2017.07.056 Torre LA, Siegel RL, Ward EM, Jemal A, 2016, Global Cancer incidence and mortality rates and trends–An update. Cancer Epidemiol Biomark Prev 25(1):16–27. , DOI 10.1158/ 1055-9965.EPI-15-0578 Sun M, Nie F, Wang Y, Zhang Z, Hou J, He D, Xie M, Xu L, De W, Wang Z, Wang J, 2016, LncRNA HOXA11-AS promotes proliferation and invasion of gastric Cancer by scaffolding the chromatin modification factors PRC2, LSD1, and DNMT1. Cancer Res 76(21):6299–6310. , DOI 10.1158/0008- 5472.CAN-16-0356 Xu TP, Huang MD, Xia R, Liu XX, Sun M, Yin L, Chen WM, Han L, Zhang EB, Kong R, DeW, ShuYQ(2014, Decreased expression of the long non-coding RNA FENDRR is associated with poor prognosis in gastric cancer and FENDRR regulates gastric cancer cell metastasis by affecting fibronectin1 expression. J Hematol Oncol 7:63. , DOI 10.1186/s13045-014-0063-7 Sun TT, He J, Liang Q, Ren LL, Yan TT, Yu TC, Tang JY, Bao YJ, Hu Y, Lin Y, Sun D, Chen YX, Hong J, Chen H, ZouW, Fang JY, 2016, LncRNA GClnc1 promotes gastric carcinogenesis and may act as a modular scaffold of WDR5 and KAT2A complexes to specify the histone modification pattern. Cancer Discov 6(7): 784–801. , DOI 10.1158/2159-8290.CD-15-0921 Wu SW, Hao YP, Qiu JH, Zhang DB, Yu CG, LiWH, 2017, High expression of long non-coding RNA CCAT2 indicates poor prognosis of gastric cancer and promotes cell proliferation and invasion. Minerva Med 108(4):317–323. , DOI 10.23736/ S0026-4806.17.04703-6 Hu Y,Ma Z, He Y, LiuW, Su Y, Tang Z, 2017, LncRNA-SNHG1 contributes to gastric cancer cell proliferation by regulating DNMT1. Biochem Biophys Res Commun 491(4):926–931. , DOI 10.1016/j.bbrc.2017.07.137 Yan J, Guo X, Xia J, Shan T, Gu C, Liang Z, ZhaoW, Jin S, 2014, MiR-148a regulates MEG3 in gastric cancer by targeting DNA methyltransferase 1. Med Oncol 31(3):879. , DOI 10. 1007/s12032-014-0879-6 Wei GH,Wang X, 2017, lncRNAMEG3 inhibit proliferation and metastasis of gastric cancer via p53 signaling pathway. Eur Rev Med Pharmacol Sci 21(17):3850–3856 Sun M, Xia R, Jin F, Xu T, Liu Z, De W, Liu X, 2014, Downregulated long noncoding RNA MEG3 is associated with poor prognosis and promotes cell proliferation in gastric cancer. Tumour Biol 35(2):1065–1073. , DOI 10.1007/s13277- 013-1142-z PengW, Si S, Zhang Q, Li C, Zhao F,Wang F, Yu J, Ma R, 2015, Long non-coding RNA MEG3 functions as a competing endogenous RNA to regulate gastric cancer progression. J Exp Clin Cancer Res 34:79. , DOI 10.1186/s13046-015- 0197-7 Wong MCS, Lao XQ, Ho KF, Goggins WB, Tse SLA, 2017, Incidence and mortality of lung cancer: global trends and association with socioeconomic status. Sci Rep 7(1):14300. https://doi. org/10.1038/s41598-017-14513-7 Dela Cruz CS, Tanoue LT, Matthay RA, 2011, Lung cancer: epidemiology, etiology, and prevention. Clin Chest Med 32(4):605– 644. , DOI 10.1016/j.ccm.2011.09.001 Zhang L, Zhou XF, Pan GF, Zhao JP, 2014, Enhanced expression of long non-coding RNA ZXF1 promoted the invasion and metastasis in lung adenocarcinoma. Biomed Pharmacother 68(4): 401–407. , DOI 10.1016/j.biopha.2014.03.001 Whiteside EJ, Seim I, Pauli JP, O'Keeffe AJ, Thomas PB, Carter SL, Walpole CM, Fung JN, Josh P, Herington AC, Chopin LK, 2013, Identification of a long non-coding RNA gene, growth hormone secretagogue receptor opposite strand, which stimulates cell migration in non-small cell lung cancer cell lines. Int J Oncol 43(2):566–574. , DOI 10.3892/ijo.2013.1969 Nie FQ, Zhu Q, Xu TP, Zou YF, Xie M, Sun M, Xia R, Lu KH, 2014, Long non-coding RNA MVIH indicates a poor prognosis for non-small cell lung cancer and promotes cell proliferation and invasion. Tumour Biol 35(8):7587–7594. , DOI 10.1007/ s13277-014-2009-7 Liu Z, Sun M, Lu K, Liu J, Zhang M, Wu W, De W, Wang Z, Wang R, 2013, The long noncoding RNA HOTAIR contributes to cisplatin resistance of human lung adenocarcinoma cells via downregualtion of p21(WAF1/CIP1, expression. PLoS One 8(10):e77293. , DOI 10.1371/journal.pone.0077293 Qiu M, Xu Y, Yang X,Wang J, Hu J, Xu L, Yin R, 2014, CCAT2 is a lung adenocarcinoma-specific long non-coding RNA and promotes invasion of non-small cell lung cancer. Tumour Biol 35(6): 5375–5380. , DOI 10.1007/s13277-014-1700-z Sun M, Liu XH, Lu KH, Nie FQ, Xia R, Kong R, Yang JS, Xu TP, Liu YW, Zou YF, Lu BB, Yin R, Zhang EB, Xu L, De W, Wang ZX, 2014, EZH2-mediated epigenetic suppression of long noncoding RNA SPRY4-IT1 promotes NSCLC cell proliferation and metastasis by affecting the epithelial-mesenchymal transition. Cell Death Dis 5:e1298. , DOI 10.1038/cddis.2014.256 Song H, Li Y, Lee J, Schwartz AL, Bu G, 2009, Low-density lipoprotein receptor-related protein 1 promotes cancer cell migration and invasion by inducing the expression of matrix metalloproteinases 2 and 9. Cancer Res 69(3):879–886. , DOI 10. 1158/0008-5472.CAN-08-3379 HuangXY, ShiGM, Devbhandari RP, KeAW,WangY,WangXY, Wang Z, Shi YH, Xiao YS, Ding ZB, Dai Z, Xu Y, Jia WP, Tang ZY, Fan J, Zhou J, 2012, Low level of low-density lipoprotein receptor-related protein 1 predicts an unfavorable prognosis of hepatocellular carcinoma after curative resection. PLoS One 7(3):e32775. , DOI 10.1371/journal.pone.0032775 TerashimaM, Tange S, Ishimura A, Suzuki T, 2017)MEG3 Long noncoding RNA contributes to the epigenetic regulation of epithelial-mesenchymal transition in lung Cancer cell lines. J Biol Chem 292(1):82–99. , DOI 10.1074/jbc.M116. 750950 Xia Y, He Z, Liu B, Wang P, Chen Y, 2015, Downregulation of Meg3 enhances cisplatin resistance of lung cancer cells through activation of the WNT/beta-catenin signaling pathway. Mol Med Rep 12(3):4530–4537. , DOI 10.3892/mmr.2015.3897 Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63(1):11–30. , DOI 10.3322/caac.21166 Fu Y, Biglia N,Wang Z, Shen Y, Risch HA, Lu L, Canuto EM, Jia W, Katsaros D, Yu H, 2016, Long non-coding RNAs, ASAP1- IT1, FAM215A, and LINC00472, in epithelial ovarian cancer. Gynecol Oncol 143(3):642–649. , DOI 10.1016/j.ygyno. 2016.09.021 Chen Q, Liu X, Xu L, Wang Y, Wang S, Li Q, Huang Y, Liu T, 2016, Long non-coding RNA BACE1-AS is a novel target for anisomycin-mediated suppression of ovarian cancer stem cell proliferation and invasion. Oncol Rep 35(4):1916–1924. https://doi. org/10.3892/or.2016.4571 Wu DI, Wang T, Ren C, Liu L, Kong D, Jin X, Li X, Zhang G, 2016, Downregulation of BC200 in ovarian cancer contributes to cancer cell proliferation and chemoresistance to carboplatin. Oncol Lett 11(2):1189–1194. , DOI 10.3892/ol.2015.3983 Szafron LM, Balcerak A, Grzybowska EA, Pienkowska-Grela B, Podgorska A, Zub R, Olbryt M, Pamula-Pilat J, Lisowska KM, Grzybowska E, Rubel T, Dansonka-Mieszkowska A, Konopka B, Kulesza M, Lukasik M, Kupryjanczyk J, 2015, The putative oncogene, CRNDE, is a negative prognostic factor in ovarian cancer patients. Oncotarget 6(41):43897–43910. , DOI 10. 18632/oncotarget.6016 Hu X, Feng Y, Zhang D, Zhao SD, Hu Z, Greshock J, Zhang Y, Yang L, Zhong X, Wang LP, Jean S, Li C, Huang Q, Katsaros D, Montone KT, Tanyi JL, Lu Y, Boyd J, Nathanson KL, Li H,Mills GB, Zhang L, 2014, A functional genomic approach identifies FAL1 as an oncogenic long noncoding RNA that associates with BMI1 and represses p21 expression in cancer. Cancer Cell 26(3): 344–357. , DOI 10.1016/j.ccr.2014.07.009 Gao Y, Meng H, Liu S, Hu J, Zhang Y, Jiao T, Liu Y, Ou J,Wang D, Yao L, Liu S, Hui N, 2015, LncRNA-HOST2 regulates cell biological behaviors in epithelial ovarian cancer through a mechanism involving microRNA let-7b. Hum Mol Genet 24(3):841– 852. , DOI 10.1093/hmg/ddu502 Sheng X, Li J, Yang L, Chen Z, Zhao Q, Tan L, Zhou Y, Li J, 2014, Promoter hypermethylation influences the suppressive role of maternally expressed 3, a long non-coding RNA, in the development of epithelial ovarian cancer. Oncol Rep 32(1):277–285. , DOI 10.3892/or.2014.3208 Xiu YL, Sun KX, Chen X, Chen S, Zhao Y, Guo QG, Zong ZH, 2017, Upregulation of the lncRNA Meg3 induces autophagy to inhibit tumorigenesis and progression of epithelial ovarian carcinoma by regulating activity of ATG3. Oncotarget 8(19):31714– 31725. , DOI 10.18632/oncotarget.15955 Moore DD, Luu HH, 2014, Osteosarcoma. Cancer Treat Res 162: 65–92. , DOI 10.1007/978-3-319-07323-1_4 Wang B, Su Y, Yang Q, Lv D, ZhangW, Tang K,Wang H, Zhang R, Liu Y, 2015, Overexpression of Long non-coding RNA HOTAIR promotes tumor growth and metastasis in human osteosarcoma. Mol Cell 38(5):432–440. , DOI 10.14348/ molcells.2015.2327 LuoW, He H, XiaoW, Liu Q, Deng Z, Lu Y,WangQ, Zheng Q, Li Y, 2016, MALAT1 promotes osteosarcoma development by targeting TGFA via MIR376A. Oncotarget 7(34):54733–54743. , DOI 10.18632/oncotarget.10752 Chan LH, Wang W, Yeung W, Deng Y, Yuan P, Mak KK, 2014, Hedgehog signaling induces osteosarcoma development through Yap1 and H19 overexpression. Oncogene 33(40):4857–4866. , DOI 10.1038/onc.2013.433 Ruan W, Wang P, Feng S, Xue Y, Li Y, 2016, Long non-coding RNA small nucleolar RNA host gene 12, SNHG12, promotes cell proliferation and migration by upregulating angiomotin gene expression in human osteosarcoma cells. Tumour Biol 37(3):4065– 4073. , DOI 10.1007/s13277-015-4256-7 Yin Z, Ding H, He E, Chen J, LiM(2016, Overexpression of long non-codingRNAMFI2 promotes cell proliferation and suppresses apoptosis in human osteosarcoma. Oncol Rep 36(4):2033–2040. , DOI 10.3892/or.2016.5013 Tian ZZ, Guo XJ, ZhaoYM, FangY, 2015, Decreased expression of long non-coding RNA MEG3 acts as a potential predictor biomarker in progression and poor prognosis of osteosarcoma. Int J Clin Exp Pathol 8(11):15138–15142 Sahin Y, Altan Z, Arman K, Bozgeyik E, Koruk Ozer M, Arslan A, 2017, Inhibition of miR-664a interferes with the migration of osteosarcoma cells via modulation of MEG3. Biochem Biophys Res Commun 490(3):1100–1105. , DOI 10.1016/j.bbrc. 2017.06.174 Yu F, GengW, Dong P, Huang Z, Zheng J, 2018, LncRNA-MEG3 inhibits activation of hepatic stellate cells through SMO protein and miR-212. Cell Death Dis 9(10):1014. , DOI 10.1038/ s41419-018-1068-x Xue R, Li Y, Li X,Ma J, An C, Ma Z, 2018, miR-185 affected the EMT, cell viability and proliferation via DNMT1/MEG3 pathway in TGF-beta1-induced renal fibrosis. Cell Biol Int. , DOI 10.1002/cbin.11046 He H, Dai J, Zhuo R, Zhao J,Wang H, Sun F, Zhu Y, Xu D, 2018, Study on the mechanism behind lncRNA MEG3 affecting clear cell renal cell carcinoma by regulating miR-7/RASL11B signaling. J Cell Physiol 233(12):9503–9515. , DOI 10.1002/ jcp.26849 Long J, Pi X, 2018, lncRNA-MEG3 suppresses the proliferation and invasion of melanoma by regulating CYLD expression mediated by sponging miR-499-5p. Biomed Res Int 2018:2086564. , DOI 10.1155/2018/2086564 Li Z, Yang L, Liu X, Nie Z, Luo J, 2018, Long noncoding RNA MEG3 inhibits proliferation of chronic myeloid leukemia cells by sponging microRNA21. Biomed Pharmacother 104:181–192. , DOI 10.1016/j.biopha.2018.05.047 Xu J, Xu Y, 2017, The lncRNA MEG3 downregulation leads to osteoarthritis progression via miR-16/SMAD7 axis. Cell Biosci 7: 69. , DOI 10.1186/s13578-017-0195-x Li J, Zi Y, Wang W, Li Y, 2018, Long noncoding RNA MEG3 inhibits cell proliferation and metastasis in chronic myeloid leukemia via targeting miR-184. Oncol Res 26(2):297–305. https://doi. org/10.3727/096504017X14980882803151 Braconi C, Kogure T, Valeri N, Huang N, Nuovo G, Costinean S, Negrini M, Miotto E, Croce CM, Patel T, 2011, microRNA-29 can regulate expression of the long non-coding RNA geneMEG3 in hepatocellular cancer. Oncogene 30(47):4750–4756. https://doi. org/10.1038/onc.2011.193 GuoW, Dong Z, Liu S, Qiao Y, Kuang G, Guo Y, Shen S, Liang J, 2017, Promoter hypermethylation-mediated downregulation of miR-770 and its host gene MEG3, a long non-coding RNA, in the development of gastric cardia adenocarcinoma. Mol Carcinog 56(8):1924–1934. , DOI 10.1002/mc.22650 Wang Y, Kong D, 2018, Knockdown of lncRNA MEG3 inhibits viability, migration, and invasion and promotes apoptosis by sponging miR-127 in osteosarcoma cell. J Cell Biochem 119(1): 669–679. , DOI 10.1002/jcb.26230 Wang H, Li H, Zhang L, Yang D, 2018, Overexpression ofMEG3 sensitizes colorectal cancer cells to oxaliplatin through regulation of miR-141/PDCD4 axis. Biomed Pharmacother 106:1607–1615. , DOI 10.1016/j.biopha.2018.07.131 Wang P, Chen D, Ma H, Li Y, 2017, LncRNA MEG3 enhances cisplatin sensitivity in non-small cell lung cancer by regulating miR-21-5p/SOX7 axis. Onco Targets Ther 10:5137–5149. , DOI 10.2147/OTT.S146423 Liu Y, Yue P, Zhou T, Zhang F,Wang H, Chen X, 2018, LncRNA MEG3 enhances, 131)I sensitivity in thyroid carcinoma via sponging miR-182. Biomed Pharmacother 105:1232–1239. , DOI 10.1016/j.biopha.2018.06.087 Zhang J, Liu J, Xu X, Li L, 2017, Curcumin suppresses cisplatin resistance development partly via modulating extracellular vesicle-mediated transfer of MEG3 and miR-214 in ovarian cancer. Cancer Chemother Pharmacol 79(3):479–487. , DOI 10.1007/s00280-017-3238-4 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 859 EP 874 DI 10.1007/s12253-019-00614-3 PG 16 ER PT J AU Xu, N Wang, L Sun, P Xu, S Fu, Sh Sun, Z AF Xu, Ning Wang, Lili Sun, Ping Xu, Suyang Fu, Shiping Sun, Zhihua TI Low Arid1a Expression Correlates with Poor Prognosis and Promotes Cell Proliferation and Metastasis in Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ARID1A; Osteosarcoma; Tumour suppressor; Proliferation; Metastasis Prognosis ID ARID1A; Osteosarcoma; Tumour suppressor; Proliferation; Metastasis Prognosis AB AT-rich interactive domain-containing protein 1A (ARID1A) has been shown to function as a tumour suppressor in various malignancies. However, the biological role of ARID1A in osteosarcoma is not clear. The present study aimed to investigate the expression pattern, prognostic value and the biological role of ARID1A in human osteosarcoma. ARID1A expression in 53 osteosarcoma surgical specimens was examined by quantitative real-time polymerase chain reaction, and its clinical significance was analysed. The role of ARID1A in cell proliferation, apoptosis, and metastasis were examined. ARID1A mRNA expression were significantly down-regulated in osteosarcoma tumours from that in matched adjacent non-tumour tissues. ARID1A expression was significantly inversely correlated with tumour stage and distant metastasis, as well as poor overall survival in patients with osteosarcoma. Furthermore, ARID1A mRNA was downregulated in four human osteosarcoma cell lines MG-63, U2OS, HOS and Saos-2. Restoring of ARID1A expression in MG-63 and U2OS cells significantly inhibited cell proliferation and metastasis in vitro. Collectively, our data demonstrate that ARID1A may serve as a tumour suppressor in osteosarcoma progression, and represent a valuable prognostic marker and potential therapeutic target for osteosarcoma. C1 [Xu, Ning] Shanghai Eighth People’s Hospital, Departments of Orthopedics, 200235 Shanghai, China. [Wang, Lili] Bengbu Medical College, 233030 Bengbu, Anhui, China. [Sun, Ping] Shanghai Eighth People’s Hospital, Departments of Orthopedics, 200235 Shanghai, China. [Xu, Suyang] Shanghai Eighth People’s Hospital, Departments of Orthopedics, 200235 Shanghai, China. [Fu, Shiping] Shanghai Eighth People’s Hospital, Departments of Orthopedics, 200235 Shanghai, China. [Sun, Zhihua] Xiangyang Central Hospital, Departments of Oncology, No 136 Jingzhou street, 441021 Xiangyang, Hubei, China. RP Sun, Z (reprint author), Xiangyang Central Hospital, Departments of Oncology, 441021 Xiangyang, China. EM drzh_sun802@sina.com CR Aljubran AH, Griffin A, Pintilie M, Blackstein M, 2009, Osteosarcoma in adolescents and adults: survival analysis with and without lung metastases. Ann Oncol: Off J Eur Soc Med Oncol / ESMO 20(6):1136–1141. , DOI 10.1093/annonc/mdn731 Allo G, Bernardini MQ, Wu RC, Shih Ie M, Kalloger S, Pollett A, Gilks CB, Clarke BA, 2014, ARID1A loss correlates with mismatch repair deficiency and intact p53 expression in high-grade endometrial carcinomas. Mod Pathol: Off J US Can Acad Pathol, Inc 27(2):255–261. , DOI 10.1038/modpathol.2013.144 Bitler BG, Aird KM, Garipov A, Li H, Amatangelo M, Kossenkov AV, Schultz DC, Liu Q, Shih Ie M, Conejo-Garcia JR, Speicher DW, Zhang R, 2015, Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat Med 21(3): 231–238. , DOI 10.1038/nm.3799 Cho H, Kim JS, Chung H, Perry C, Lee H, Kim JH, 2013, Loss of ARID1A/BAF250a expression is linked to tumor progression and adverse prognosis in cervical cancer. Hum Pathol 44(7):1365– 1374. , DOI 10.1016/j.humpath.2012.11.007 Dhaini HR, Thomas DG, Giordano TJ, Johnson TD, Biermann JS, Leu K, Hollenberg PF, Baker LH, 2003, Cytochrome P450 CYP3A4/5 expression as a biomarker of outcome in osteosarcoma. J Clin Oncol Off J Am Soc Clin Oncol 21(13):2481–2485. https:// doi.org/10.1200/JCO.2003.06.015 Fadare O, Gwin K, Desouki MM, Crispens MA, Jones HW 3rd, Khabele D, Liang SX, ZhengW,Mohammed K, Hecht JL, Parkash V, 2013, The clinicopathologic significance of p53 and BAF-250a, ARID1A, expression in clear cell carcinoma of the endometrium. Mod Pathol: Off J US Can Acad Pathol, Inc 26(8):1101–1110. , DOI 10.1038/modpathol.2013.35 Gemoll T, Epping F, Heinrich L, Fritzsche B, Roblick UJ, Szymczak S, Hartwig S, Depping R, Bruch HP, Thorns C, Lehr S, Paech A, Habermann JK, 2015, Increased cathepsin D protein expression is a biomarker for osteosarcomas, pulmonarymetastases and other bone malignancies. Oncotarget 6(18):16517–16526. 10. 18632/oncotarget.4140 Guan B, Wang TL, Shih Ie M, 2011, ARID1A, a factor that promotes formation of SWI/SNF-mediated chromatin remodeling, is a tumor suppressor in gynecologic cancers. Cancer Res 71(21):6718– 6727. , DOI 10.1158/0008-5472.CAN-11-1562 He F, Li J, Xu J, Zhang S, Xu Y, Zhao W, Yin Z, Wang X, 2015, Decreased expression of ARID1A associates with poor prognosis and promotes metastases of hepatocellular carcinoma. J Exp Clin Cancer Res 34:47. , DOI 10.1186/s13046-015-0164-3 Jones S,WangTL,ShihIeM,MaoTL,NakayamaK,RodenR,GlasR, Slamon D, Diaz LA Jr, Vogelstein B, Kinzler KW, Velculescu VE, Papadopoulos N, 2010, Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330(6001): 228–231. , DOI 10.1126/science.1196333 Jones S, Li M, Parsons DW, Zhang X, Wesseling J, Kristel P, Schmidt MK, Markowitz S, Yan H, Bigner D, Hruban RH, Eshleman JR, Iacobuzio-Donahue CA, Goggins M, Maitra A, Malek SN, Powell S, Vogelstein B, Kinzler KW, Velculescu VE, Papadopoulos N(2012, Somaticmutations in the chromatin remodeling gene ARID1A occur in several tumor types. Hum Mutat 33(1):100–103. , DOI 10.1002/humu.21633 Kersting C, Gebert C, Agelopoulos K, Schmidt H, van Diest PJ, Juergens H, Winkelmann W, Kevric M, Gosheger G, Brandt B, Bielack S,BuergerH(2007, Epidermal growth factor receptor expression in high-grade osteosarcomas is associated with a good clinical outcome. Clin Cancer Res: Off J Am Assoc Cancer Res 13(10): 2998–3005. , DOI 10.1158/1078-0432.CCR-06-2432 KimMJ,GuMJ,ChangHK,YuE(2015)LossofARID1Aexpressionis associated with poor prognosis in small intestinal carcinoma. Histopathology 66(4):508–516. , DOI 10.1111/his.12566 LiY, NakkaM, KellyAJ, LauCC,KrailoM, BarkauskasDA,Hicks JM, Man TK, 2016, p27 Is a Candidate Prognostic Biomarker and Metastatic Promoter in Osteosarcoma. Cancer Res 76(13):4002– 4011. , DOI 10.1158/0008-5472.CAN-15-3189 McConechy MK, Ding J, Senz J, Yang W, Melnyk N, Tone AA, Prentice LM, Wiegand KC, McAlpine JN, Shah SP, Lee CH, Goodfellow PJ, Gilks CB, Huntsman DG, 2014, Ovarian and endometrial endometrioid carcinomas have distinct CTNNB1 and PTEN mutation profiles. Mod Pathol 27(1):128–134. https://doi. org/10.1038/modpathol.2013.107 Nagl NG Jr, Wang X, Patsialou A, Van Scoy M, Moran E, 2007, Distinct mammalian SWI/SNF chromatin remodeling complexes with opposing roles in cell-cycle control. EMBO J 26(3):752– 763. , DOI 10.1038/sj.emboj.7601541 Park JH, Lee C, Suh JH, Chae JY, Kim HW, Moon KC, 2015, Decreased ARID1A expression correlates with poor prognosis of clear cell renal cell carcinoma. Hum Pathol 46(3):454–460. https:// doi.org/10.1016/j.humpath.2014.12.002 Suryo Rahmanto Y, Jung JG, Wu RC, Kobayashi Y, Heaphy CM, Meeker AK, Wang TL, Shih Ie M, 2016, Inactivating ARID1A Tumor Suppressor Enhances TERT Transcription and Maintains Telomere Length in Cancer Cells. J Biol Chem 291(18):9690– 9699. , DOI 10.1074/jbc.M115.707612 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108. , DOI 10.3322/caac.21262 Wang DD, Chen YB, Pan K,WangW, Chen SP, Chen JG, Zhao JJ, Lv L, Pan QZ, Li YQ,Wang QJ, Huang LX, Ke ML, He J, Xia JC, 2012, Decreased expression of the ARID1A gene is associated with poor prognosis in primary gastric cancer. PLoS One 7(7): e40364. , DOI 10.1371/journal.pone.0040364 Wei XL,Wang DS, Xi SY,WuWJ, Chen DL, Zeng ZL,Wang RY, Huang YX, Jin Y, Wang F, Qiu MZ, Luo HY, Zhang DS, Xu RH, 2014, Clinicopathologic and prognostic relevance of ARID1A protein loss in colorectal cancer. World J Gastroenterol: WJG 20(48):18404–18412. , DOI 10.3748/wjg.v20.i48.18404 Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, Senz J, McConechy MK, Anglesio MS, Kalloger SE, Yang W, Heravi- Moussavi A, Giuliany R, Chow C, Fee J, Zayed A, Prentice L, Melnyk N, Turashvili G, Delaney AD, Madore J, Yip S, McPherson AW, Ha G, Bell L, Fereday S, Tam A, Galletta L, Tonin PN, Provencher D, Miller D, Jones SJ, Moore RA, Morin GB, Oloumi A, Boyd N, Aparicio SA, Shih Ie M, Mes-Masson AM, Bowtell DD, Hirst M, Gilks B, Marra MA, Huntsman DG, 2010)ARID1Amutations in endometriosis-associated ovarian carcinomas. N Engl J Med 363(16):1532–1543. , DOI 10. 1056/NEJMoa1008433 Wu JN, Roberts CW, 2013, ARID1A mutations in cancer: another epigenetic tumor suppressor? Cancer Discov 3(1):35–43. https:// doi.org/10.1158/2159-8290.CD-12-0361 Wu RC, Wang TL, Shih Ie M, 2014, The emerging roles of ARID1A in tumor suppression. Cancer Biol Ther 15(6):655–664. , DOI 10.4161/cbt.28411 WuRC,AyhanA,MaedaD,KimKR, ClarkeBA, ShawP,ChuiMH, Rosen B, Shih IeM,WangTL, 2014, Frequent somaticmutations of the telomerase reverse transcriptase promoter in ovarian clear cell carcinoma but not in other major types of gynaecologicalmalignancy. J Pathol 232(4):473–481. , DOI 10.1002/path.4315 Yamamoto S, Tsuda H, Takano M, Tamai S, Matsubara O, 2012, PIK3CA mutations and loss of ARID1A protein expression are early events in the development of cystic ovarian clear cell adenocarcinoma. Virchows Arch: Int J Pathol 460(1):77–87. https://doi. org/10.1007/s00428-011-1169-8 Yang SZ,Wang AQ, Du J,Wang JT, YuWW, Liu Q,Wu YF, Chen SG, 2016, Low expression of ARID1A correlates with poor prognosis in intrahepatic cholangiocarcinoma. World J Gastroenterol: WJG 22(25):5814–5821. , DOI 10.3748/wjg.v22.i25.5814 Yang L, Wei S, Zhao R, Wu Y, Qiu H, Xiong H, 2016, Loss of ARID1A expression predicts poor survival prognosis in gastric cancer: a systematic meta-analysis from 14 studies. Sci Rep 6: 28919. , DOI 10.1038/srep28919 Zang ZJ, Cutcutache I, Poon SL, Zhang SL, McPherson JR, Tao J, Rajasegaran V, Heng HL, Deng N, Gan A, Lim KH, Ong CK, Huang D, Chin SY, Tan IB, Ng CC, Yu W, Wu Y, Lee M, Wu J, Poh D, Wan WK, Rha SY, So J, Salto-Tellez M, Yeoh KG, Wong WK, Zhu YJ, Futreal PA, Pang B, Ruan Y, Hillmer AM, Bertrand D, Nagarajan N, Rozen S, Teh BT, Tan P, 2012, Exome sequencing of gastric adenocarcinoma identifies recurrent somatic mutations in cell adhesion and chromatin remodeling genes. Nat Genet 44(5): 570–574. , DOI 10.1038/ng.2246 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 875 EP 881 DI 10.1007/s12253-017-0338-8 PG 7 ER PT J AU Peng, W Wu, J Fan, H Lu, J Feng, J AF Peng, Wei Wu, Jianzhong Fan, Hong Lu, Jianwei Feng, Jifeng TI LncRNA EGOT Promotes Tumorigenesis Via Hedgehog Pathway in Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; EGOT; lncRNA; Tumorigenesis; Hedgehog ID Gastric cancer; EGOT; lncRNA; Tumorigenesis; Hedgehog AB Gastric cancer (GC) is one of the mostly terminal malignancies with poor prognosis. Long noncoding RNA EGOT (EGOT) acts as a crucial regulator in the breast cancer. However, the function of EGOT in GC remains unknown. This work was to explore the clinical value and biological significance of EGOT in GC. EGOT levels in GC tissue and cell were analyzed by qRT–PCR. After knockdown of EGOT, GC cell growth and cycle progression were detected. The expression of EGOT was observably elevated in GC.Upregulation of EGOT was related with lymphatic metastasis and TNM stage. In addition, knockdown of EGOT by siRNA could significantly inhibit GC cell proliferation and arrest cycle progression in G1 phase. Moreover, EGOT mediated cyclin D1 expression in GC cells which was regulated by Hedgehog pathway. Further, loss of EGOT downregulated Hedgehog signaling pathway in GC cells. EGOT functions as an oncogene in GC, and may be useful as a conceivable diagnostic and prognostic biomarker for GC tumorigenesis. C1 [Peng, Wei] Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Department of Medical Oncology, No. 42 Baiziting Road, 210009 Nanjing, China. [Wu, Jianzhong] Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Laboratory of Cancer Research, No. 42 Baiziting Road, 210009 Nanjing, China. [Fan, Hong] First People’s Hospital of Yunnan Province, Department of Gastroenterology, No. 175 Jinbi Road, 650032 Kunming, China. [Lu, Jianwei] Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Department of Medical Oncology, No. 42 Baiziting Road, 210009 Nanjing, China. [Feng, Jifeng] Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Department of Medical Oncology, No. 42 Baiziting Road, 210009 Nanjing, China. RP Feng, J (reprint author), Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Department of Medical Oncology, 210009 Nanjing, China. EM willgeek01@163.com CR Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66(1):7–30 Hartgrink HH, Jansen EPM, van Grieken NCT, van de Velde CJH, 2009, Gastric cancer. Lancet 374(9688):477–490 Leung WK, MS W, Kakugawa Yet al, 2008, Screening for gastric cancer in Asia: current evidence and practice. Lancet Oncol 9(3): 279–287 Modlin IM, Lye KD, KiddM(2003, A 5-decade analysis of 13,715 carcinoid tumors. Cancer 97(4):934–959 Ponting CP, Oliver PL, Reik W, 2009, Evolution and functions of long noncoding RNAs. Cell 136(4):629–641 Wapinski O, Chang HY, 2011, Long noncoding RNAs and human disease. Trends Cell Biol 21(6):354–361 Wagner LA, Christensen CJ, Dunn DM et al, 2007, EGO, a novel, noncoding RNA gene, regulates eosinophil granule protein transcript expression. Blood 109(12):5191–5198 SP X, Zhang JF, Sui SY et al, 2015, Downregulation of the long noncoding RNA EGOT correlates with malignant status and poor prognosis in breast cancer. Tumour Biol J Int Soc Oncodev Biol Med 36(12):9807–9812 Peng W, GQ W, Fan H, JZ W, Feng JF, 2015, Long noncoding RNA SPRY4-IT1 predicts poor patient prognosis and promotes tumorigenesis in gastric cancer. Tumor Biol 36(9):6751–6758 Peng W, Wu J, Feng J, 2017, LincRNA-p21 predicts favorable clinical outcome and impairs tumorigenesis in diffuse large B cell lymphoma patients treated with R-CHOP chemotherapy. Clin Exp Med 17(1):1–8. , DOI 10.1007/s10238-015-0396-8 TsaiMC, Spitale RC, Chang HY, 2011, Long intergenic noncoding RNAs: new links in cancer progression. Cancer Res 71(1):3–7 Prensner JR, Chinnaiyan AM(2011, The emergence of lncRNAs in cancer biology. Cancer Discov 1(5):391–407 Michailidou K, Hall P, Gonzalez-Neira A et al, 2013, Large-scale genotyping identifies 41 new loci associated with breast cancer risk. Nat Genet 45(4):353–361 61e1-2 Taipale J, Beachy PA, 2001, The Hedgehog and Wnt signaling pathways in cancer. Nature 411(6835):349–354 Berman DM, Karhadkar SS, Maitra A et al, 2003, Widespread requirement for Hedgehog ligand stimulation in growth of digestive tract tumours. Nature 425(6960):846–851 Varnat F, Duquet A, Malerba M et al, 2009, Human colon cancer epithelial cells harbour active HEDGEHOG-GLI signalling that is essential for tumour growth, recurrence, metastasis and stem cell survival and expansion. EMBO Mol Med 1(6–7):338–351 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 883 EP 887 DI 10.1007/s12253-017-0367-3 PG 5 ER PT J AU Xiao, GQ Barrett, MM Yang, Q Unger, DP AF Xiao, Guang-Qian Barrett, M Mary Yang, Qi Unger, D Pamela TI Clinicopathologic and Immunohistochemical Study of Combined Small Cell Carcinoma and Urothelial Carcinoma Molecular Subtype SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Urothelial carcinoma; Small cell carcinoma of urinary bladder; Molecular subtype; Immunohistochemistry ID Urothelial carcinoma; Small cell carcinoma of urinary bladder; Molecular subtype; Immunohistochemistry AB Muscle invasive bladder cancer, an aggressive disease with heterogeneous molecular profiles, has recently been subclassified into three major molecular subtypes -basal, luminal and "p53-like" urothelial carcinomas (UCas), which bear prognostic and therapeutic implication. Similar to breast cancer, basal and luminal subtype UCas are designated by basal (CK5/14) and luminal (CK20) markers. The "p53-like" subtype presents with wild-type p53 gene with upregulated p53 pathways and is implicated in chemoresistance. Urinary bladder is one of the most common primary sites of extrapulmonary small cell carcinoma (SmCC). Bladder SmCC frequently coexists with UCa; however, the relation of SmCC with specific UCa molecular subtypes has not been studied. The aim of this study is to investigate the clinicopathology and immunophenotypes of the combined SmCC and UCa molecular subtypes. A total of 22 combined SmCC and UCa cases were studied for the clinicopathology and immunohistochemical (IHC) profiles by luminal and basal cell markers as well as Her2/Neu and p53. Our results demonstrated that all the urinary bladder SmCCs were associated with high grade UCas. They were more commonly seen in older male patients with a smoking history and had a poor prognosis. Based on the reported molecular subtyping, the UCas could be immunohistochemically subclassified into luminal, basal, dual and null types, which showed different clinicopathologic and IHC features. Compared to non-SmCC associated UCa, the subtypes of UCa in the combined SmCCs and UCas were characterized by: 1) Although overall luminal type was still relatively more common in men, basal marker-expressing subtypes were significantly increased in incidence and were more common in women. 2) Her2/Neu overexpression was more commonly observed in luminal than basal cell marker-expressing UCas. 3) IHC overexpression of p53 was common in all the subtypes, with UCas and SmCCs sharing the same p53 expression pattern. Although limited by relatively a small number of cases, the results of this study will enhance our understanding of the combined SmCC and UCa entity and potentially lead to a future therapeutic management. C1 [Xiao, Guang-Qian] University of Southern California, Keck Medical Center, Department of Pathology, 1500 San Pablo Street, 90033 Los Angeles, CA, USA. [Barrett, M Mary] University of Rochester Medical Center, Department of Pathology & Laboratory MedicineRochester, NY, USA. [Yang, Qi] University of Rochester Medical Center, Department of Pathology & Laboratory MedicineRochester, NY, USA. [Unger, D Pamela] Lenox Hill Hospital-Northwell Health System, Department of PathologyNew York, NY, USA. RP Xiao, GQ (reprint author), University of Southern California, Keck Medical Center, Department of Pathology, 90033 Los Angeles, USA. CR Falke J, Witjes JA, 2011, Contemporary management of low-risk bladder cancer. Nat Rev Urol 8(1):42–49 Knowles MA, Hurst CD, 2015, Molecular biology of bladder cancer: new insights into pathogenesis and clinical diversity. Nat Rev Cancer 15(1):25–41.5 Shah JB, McConkey DJ, Dinney CP, 2011, New strategies in muscle-invasive bladder cancer: on the road to personalized medicine. Clin Cancer Res 17(9):2608–2612 Choi W, Porten S, Kim S, Willis D, Plimack ER, Hoffman-Censits J, Roth B, Cheng T, Tran M, Lee IL, Melquist J, Bondaruk J et al, 2014, Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 25(2):152–165 Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T et al, 2001, Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98(19):10869–10874 Volkmer JP, Sahoo D, Chin RK, Ho PL, Tang C, Kurtova AV, Willingham SB et al, 2012, Three differentiation states riskstratify bladder cancer into distinct subtypes. Proc Natl Acad Sci U S A 109(6):2078–2083 Riaz SP, Luchtenborg M, Coupland VH, Spicer J, Peake MD, Moller H, 2012, Trends in incidence of small cell lung cancer and all lung cancer. Lung Cancer 75:280–284 Cheng L, Pan CX, Yang XJ, Lopez-Beltran A, MacLennan GT, Lin H, Kuzel TM et al, 2004, Small cell carcinoma of the urinary bladder: a clinicopathologic analysis of 64 patients. Cancer 101: 957–962 Pant-Purohit M, Lopez-Beltran A, Montironi R, MacLennan GT, Cheng L, 2010, Small cell carcinoma of the urinary bladder. Histol Histopathol 25(2):217–221 Zhao X, Flynn EA, 2012, Small cell carcinoma of the urinary bladder: a rare, aggressive neuroendocrine malignancy. Arch Pathol Lab Med 136(11):1451–1459 WHO Classification of Tumours of the Lung, Pleura, Thymus and Heart, 2015, 4th edition. In: Travis, W.D., Brambilla, E., Burke, A.P., Marx, A., Nicholson, A. G. eds. WHO Classification of Tumours; IARC press WHO Classification of Tumours of the Urinary System and Male Genital Organs, 2016, 4th edition. In: Moch, H, Humphrey, PA, Ulbright, TM, Reuter, VE, eds. WHO Classification of Tumours; IARC press American Joint Committee on Cancer. AJCC Cancer Staging Manual. 7th ed. New York: Springer; 2010; p569–p578 ASCO–CAP HER2 Test Guideline Recommendations, CAP, 2013). h t t p : / /www.cap.org/apps/docs/committees/ immunohistochemistry/summary of recommendations.pdf Hou CP, Lin YH, Chen CL, Chang PL, Tsui KH, 2013, Clinical outcome of primary small cell carcinoma of the urinary bladder. Onco Targets Ther 6:1179–1185 Pradhan D, Green A, Fuhrer KA, Bastacky SI, Dhir R, Parwani A, Roy S, 2016, Utility of immunohistochemistry for distinguishing luminal and basal subtypes of urothelial carcinoma on formalin fixed paraffin embedded tissue. Mod Pathol 29(suppl. 2):256A TravisWD, 2012, Update on small cell carcinoma and its differentiation from squamous cell carcinoma and other non-small cell carcinomas. Mod Pathol 25(Suppl 1):S18–S30 Shepard HM, Lewis GD, Sarup JC, Fendly BM, Maneval D, Mordenti J, Figari I et al, 1991, Monoclonal antibody therapy of human cancer: taking the HER2 protooncogene to the clinic. J Clin Immunol 11:117–127 Kern JA, Schwartz DA, Nordberg JE, Weiner DB, Greene MI, Torney L, Robinson RA. p185neu expression in human lung adenocarcinomas predicts shortened survival. Cancer Res 1990;50: 5184–5187 Ochs AM, Wong L, Kakani V, Neerukonda S, Gorske J, Rao A, Riggs M, Ward H, Keville L, 2004, Expression of vascular endothelial growth factor and HER2/neu in stage II colon cancer and correlation with survival. Clin Colorectal Cancer 4:262–267 Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A et al, 2010, Trial investigators. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer, ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376:687–697 MJ G, Hong SM, Jung SJ, 2013, HER2 protein expression and HER2 gene amplification are infrequent in small intestinal carcinomas. Virchows Arch 462:603–607 Marin A, Arranz E, Sanchez A, Aunon P, Baron M, 2010, Role of anti-her-2 therapy in bladder carcinoma. J Cancer Res Clin Oncol 136:1915–1920 LaeM, Couturier J, Oudard S, Radvanyi F, Beuzeboc P, Vieillefond A, 2010, Assessing HER2 gene amplification as a potential target for therapy in invasive urothelial bladder cancer with a standardized methodology: results in 1005 patients. Ann Oncol 21:815–819 Coogan CL, Estrada CR, Kapur S, Bloom KJ, 2004, HER-2/neu protein overexpression and gene amplification in human transitional cell carcinoma of the bladder. Urology 63:786–790 Ching CB, Amin MB, Tubbs RR et al, 2011, HER2 gene amplification occurs frequently in the micropapillary variant of urothelial carcinoma: analysis by dual-color in situ hybridization. Mod Pathol 24(8):1111–1119 Sasaki Y, Sasaki T, Kawai T et al, 2014, HER2 protein overexpression and gene amplification in upper urinary tract urothelial carcinoma-an analysis of 171 patients. Intl J Clin Exp Pathol 7(2): 699–708 Soriano P, Navarro S, Gil M, Llombart-Bosch A, 2004, Small-cell carcinoma of the urinary bladder: a clinico-pathological study of ten cases. Virchows Arch 445(3):292–297 Wang X, Jones TD, Maclennan GT et al, 2005, P53 expression in small cell carcinoma of the urinary bladder: biological and prognostic implications. Anticancer Res 25(3B):2001–2004 Siefker-Radtke AO, Dinney CP, Abrahams NA, Moran C, Shen Y, Pisters LL, Grossman HB et al, 2004, Evidence supporting preoperative chemotherapy for small cell carcinoma of the bladder: a retrospective review of the M. D. Anderson cancer experience. J Urol 172(2):481–484 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 889 EP 895 DI 10.1007/s12253-017-0369-1 PG 7 ER PT J AU Chen, J Wang, H Li, Z AF Chen, Jun Wang, Hong Li, Zhiming TI Association between Polymorphisms of X-Ray Repair Cross Complementing Group 1 Gene and Pancreatic Cancer Risk: a Systematic Review with Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE XRCC1; Polymorphisms; Pancreatic cancer; Meta-analysis ID XRCC1; Polymorphisms; Pancreatic cancer; Meta-analysis AB Emerging evidences have shown that common genetic polymorphisms in X-ray repair cross complementing group 1 (XRCC1) gene may be associated with the development of pancreatic cancer, but individually published studies and previous meta-analyses revealed inconclusive results. The aim of our study was to investigate the association between polymorphisms in XRCC1 gene and pancreatic cancer risk. We conducted a search of PubMed, Embase, the Cochrane Library and Web of Science databases. Odds ratios (ORs) with 95% confidence intervals (CIs) were determined as measures of the strength of association between polymorphisms of XRCC1 and pancreatic cancer risk. Sensitivity analysis and publication bias were evaluated. All analyses were undertaken using the STATA 13.0. A total of 10 studies were included in this systematic review. Five common functional singlenucleotide polymorphisms (SNPs) in XRCC1 gene were found, including Arg399Gln G > A (rs25487), Arg194Trp C > T (rs1799782), Arg280His G > A (rs25489), c.1517G > C, c.1471G > A. Results from our stratified analysis based on Hardy-Weinberg equilibrium (HWE) showed that there was a robust significant association between Arg280His polymorphism and pancreatic cancer risk (allelic model, OR 0.743, 95% CI 0.576–0.958, P = 0.022; heterozygous model, OR 0.701, 95% CI 0.525–0.936, P = 0.016; dominant model, OR 0.710, 95% CI 0.537–0.939, P = 0.016). We also found a statistically significant association between c.1517G > C polymorphism and pancreatic cancer risk (Allelic model, OR 1.252, 95% CI 1.064–1.473, P = 0.007). No significant results were obtained for Arg399Gln, Arg194Trp and c.1471G > A polymorphisms. The present meta-analysis suggested that Arg280His and c.1517G > C polymorphisms in XRCC1 gene were associated with pancreatic cancer risk. C1 [Chen, Jun] Xinjiang Medical University, Affiliated Hospital of Traditional Chinese Medicine, Department of General Surgery, No. 116 the Yellow River Road, 830000 Urumqi, China. [Wang, Hong] Xinjiang Medical University, Affiliated Hospital of Traditional Chinese Medicine, Department of General Surgery, No. 116 the Yellow River Road, 830000 Urumqi, China. [Li, Zhiming] Shihezi Hospital of Traditional Chinese Medicine, Department of SurgeryShihezi, China. RP Chen, J (reprint author), Xinjiang Medical University, Affiliated Hospital of Traditional Chinese Medicine, Department of General Surgery, 830000 Urumqi, China. EM chen_jun@hainan.net CR Vaccaro V, Gelibter A, Bria E, Iapicca P, Cappello P, Di Modugno F et al, 2012, Molecular and genetic bases of pancreatic cancer. Curr Drug Targets 13(6):731–743 Krejs GJ, 2010, Pancreatic cancer: epidemiology and risk factors. Dig Dis 28(2):355–358. , DOI 10.1159/000319414 Ginsberg G, Angle K, Guyton K, Sonawane B, 2011, Polymorphism in the DNA repair enzyme XRCC1: utility of current database and implications for human health risk assessment. Mutat Res Fundam Mol Mech Mutagen 727(1–2):1–15 Wood RD, Mitchell M, Sgouros J, Lindahl T, 2001, Human DNA repair genes. Science 291(5507):1284–1289 Tudek B, 2007, Base excision repair modulation as a risk factor for human cancers. Mol Asp Med 28(3–4):258–275. Epub 2007/07/14. , DOI 10.1016/j.mam.2007.05.003 Vidal AE, Boiteux S, Hickson ID, Radicella JP, 2001, XRCC1 coordinates the initial and late stages of DNA abasic site repair through protein–protein interactions. EMBO J 20(22):6530–6539 Feng YZ, Liu YL, He XF, Wei W, Shen XL, Xie DL, 2014, Association between the XRCC1 Arg194Trp polymorphism and risk of cancer: evidence from 201 case-control studies. Tumour biology: the journal of the International Society for Oncodevelopmental Biology and Medicine 35(11):10677–10697. , DOI 10.1007/s13277-014-2326-x Li J, Li Z, Feng L, Guo W, Zhang S, 2013, Polymorphisms of DNA repair gene XRCC1 and hepatocellular carcinoma risk among east Asians: a meta-analysis. Tumour biology: the journal of the International Society for Oncodevelopmental Biology and Medicine 34(1):261–269. , DOI 10.1007/s13277-012-0546-5 Dai L, Duan F,Wang P, Song C,Wang K, Zhang J, 2012, XRCC1 gene polymorphisms and lung cancer susceptibility: a metaanalysis of 44 case-control studies. Mol Biol Rep 39(10):9535– 9547. , DOI 10.1007/s11033-012-1818-2 Moher D, Liberati A, Tetzlaff J, Altman DG, 2009, Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med 6(7):e1000097. , DOI 10. 1371/journal.pmed.1000097 Rohlfs RV, Weir BS, 2008, Distributions of hardy-Weinberg equilibrium test statistics. Genetics 180(3):1609–1616. , DOI 10.1534/genetics.108.088005 Huedo-Medina TB, Sanchez-Meca J, Marin-Martinez F, Botella J, 2006, Assessing heterogeneity in meta-analysis: Q statistic or I2 index? Psychol Methods 11(2):193–206. , DOI 10.1037/ 1082-989x.11.2.193 DerSimonian R, Laird N, 1986, Meta-analysis in clinical trials. Control Clin Trials 7(3):177–188. , DOI 10.1016/0197- 2456(86)90046-2 Mantel N, Haenszel W, 1959, Statistical aspects of the analysis of data from retrospective studies of disease. J Natl Cancer Inst 22(4): 719–748 Begg CB, Mazumdar M, 1994, Operating characteristics of a rank correlation test for publication bias. Biometrics 50(4):1088–1101 Egger M, Davey Smith G, Schneider M, Minder C, 1997, Bias in meta-analysis detected by a simple, graphical test. BMJ, Clinical research ed, 315(7109):629–634 Wang LJ,Wang HT,Wang XX, 2016, Association of XRCC1 gene polymorphisms and pancreatic cancer risk in a Chinese population. Genet Mol Res 15(2):gmr8080 Jiang H, Wu D, Ma D, Lin G, Liang J, Jin J, 2013, Association between X-ray repair cross-complementation group 1 rs25487 polymorphism and pancreatic cancer risk. Tumor Biol 34(6): 3417–3421 Shen WD, Chen HL, Liu PF, 2011, XRCC1 polymorphisms and pancreatic cancer: a meta-analysis. Chin J Cancer Res 23(3):165– 170. , DOI 10.1007/s11670-011-0165-5 Mcwilliams RR, Bamlet WR, Cunningham JM, Goode EL, Andrade MD, Boardman LA et al, 2008, Polymorphisms in DNA repair genes, smoking, and pancreatic adenocarcinoma risk. Cancer Res 68(12):4928–4935 Zhao ZM, Li CG, Hu MG, Gao YX, Liu R, 2014, Influence of the c.1517G>C genetic variant in the XRCC1 gene on pancreatic cancer susceptibility in a Chinese population. Genet Mol Res 13(2):4466–4472. , DOI 10.4238/ 2014.June.16.5 Hou BH, Jian ZX, Cui P, Li SJ, Tian RQ, Ou JR, 2016, Association and intragenic single-nucleotide polymorphism interactions of the XRCC1 polymorphisms for pancreatic cancer susceptibility. Pancreas 45(4):546–551. , DOI 10.1097/mpa. 0000000000000482 Chen H, Tang C, Liu M, Zhou B, Kuang Y, Yuan T et al, 2013, Association of XRCC1 gene single nucleotide polymorphisms and susceptibility to pancreatic cancer in Chinese. Tumor Biol 35(1): 27–32. , DOI 10.1007/s13277-013-1001-y NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 897 EP 904 DI 10.1007/s12253-017-0364-6 PG 8 ER PT J AU Tsai, HL Chen, YT Yeh, YS Huang, ChW Ma, ChJ Wang, JY AF Tsai, Hsiang-Lin Chen, Yi-Ting Yeh, Yung-Sung Huang, Ching-Wen Ma, Cheng-Jen Wang, Jaw-Yuan TI Apical Lymph Nodes in the Distant Metastases and Prognosis of Patients with Stage III Colorectal Cancer with Adequate Lymph Node Retrieval Following FOLFOX Adjuvant Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Apical lymph nodes; Distant metastases; Prognosis; Stage III colorectal cancer ID Apical lymph nodes; Distant metastases; Prognosis; Stage III colorectal cancer AB The aim of the study was to assess apical lymph nodes (APNs) for predicting distant metastases in patients with stage III colorectal cancer (CRC) curatively treated with FOLFOX adjuvant chemotherapy and adequate lymph node retrieval. We investigated the correlation between APN metastasis and clinical outcomes. This retrospective study examined 97 patients. All patients were followed until death, loss to follow-up, or May 2017. Clinicopathological variables, including the APN status, were assessed. Multivariate logistic regression model was used to identify the independent risk factors for APN and distant metastases, and Cox proportional regression model was used to evaluate the association between APN metastasis and oncologic outcomes. Multivariate analyses revealed the N2 stage as an independent predictor of APN metastasis [P = 0.036; odds ratio (OR): 3.016; 95% confidence interval (CI): 1.076– 8.499], while APN metastasis was an independent risk factor for distant metastases (P < 0.001; OR: 13.876; 95% CI: 3.815–50.475). Furthermore, APN metastasis was an independent risk factor for poorer disease-free survival (DFS) and overall survival (OS) (P < 0.001 and P = 0.005, respectively). The liver (31.6%) was the most common site of distant metastases in patients with APN metastases. APN metastasis is an important prognostic factor for node-positive CRC; it enhanced the distant metastases in patients with stage III CRC curatively treated with adequate lymph node retrieval following FOLFOX adjuvant chemotherapy. Therefore, for patients with stage III CRC involving APN metastasis, prospectively randomized trials are mandatory to investigate different therapeutic strategies in addition to conventional FOLFOX adjuvant chemotherapy. C1 [Tsai, Hsiang-Lin] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, No. 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. [Chen, Yi-Ting] Kaohsiung Medical University, College of Medicine, Department of PathologyKaohsiung, Taiwan, Republic of China. [Yeh, Yung-Sung] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, No. 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. [Huang, Ching-Wen] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, No. 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. [Ma, Cheng-Jen] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, No. 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. [Wang, Jaw-Yuan] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, No. 100, Tzyou 1st Road, 807 Kaohsiung, Taiwan, Republic of China. RP Wang, JY (reprint author), Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, 807 Kaohsiung, Taiwan, Republic of China. EM cy614112@ms14.hinet.net;jayuwa@cc.kmu.edu.tw CR Kawada H, Kurita N, Nakamura F, Kawamura J, Hasegawa S, Kotake K, Sugihara K, Fukuhara S, Sakai Y, 2014, Incorporation of apical lymph status into the seventh edition of the TNM classification improves prediction of prognosis in stage III colorectal cancer. Br J Surg 101:1143–1152 Chapuis PH, Dent OF, Fisher R, Newland RC, PheilsMT, Smyth E, Colquhoun K, 1985, A multivariate analysis of clinical and pathological variables in prognosis after resection of large bowel cancer. Br J Surg 72:698–702 Gabriel WB, Dukes C, Busset HJR, 1935, Lymphatic spread in cancer of the rectum. Br J Surg 23:395–413 Malassagne B, Valleur P, Serra J, Sarnacki S, Galian A, Hoang C, Hautefeuille P, 1993, Relationship of apical lymph node involvement to survival in resected colon carcinoma. Dis Colon Rectum 36:645–653 Cserni G, Vinh-Hung V, Burzykowski T, 2002, Is there aminimum number of lymph nodes that should be histologically assessed for a reliable nodal staging of T3N0M0 colorectal carcinomas? J Surg Oncol 81:63–69 Chafai N, Chan CLH, Bokey EL, Dent OF, Sinclair G, Chapuis PH, 2005, What factors influence survival in patients with unresected synchronous liver metastases after resection of colorectal cancer? Color Dis 7:176–181 Williams GT, Quirke P, Shepherd NA, 2007, 2nd edition Standards and datasets for reporting cancers: Dataset for colorectal cancer. The Royal College of Pathologists, London Kang J, Hur H, Min BS, Kim NK, Lee KY, 2011, Prognostic impact of inferior mesenteric artery lymph node metastasis in colorectal cancer. Ann Surg Oncol 18:704–710 Huh JW, Kim YJ, Kim HR, 2012, Distribution of lymph node metastases is an independent predictor of survival for sigmoid colon and rectal cancer. Ann Surg 255:70–78 Kobayashi H, Ueno H, Hashiguchi Y, Mochizuki H, 2006, Distribution of lymph node metastasis is a prognostic index in patients with stage III colon cancer. Surgery 139:516–522 Gundara JS, Gill AJ, Hugh TJ, Samra JS, 2013, Redefining the apical lymph node at right hemicolectomy. Eur J Surg Oncol 39: 662–665 Green FL, Stewart AK, Norton HJ, 2002, A new TNM staging strategy for node-positive, stage III, colon cancer: an analysis of 50,042 patients. Ann Surg 236:416–421 Ang CW, Tweedle M, Campbell F, 2011, Apical node metastasis independently predicts poor survival in Dukes’ C colorectal cancer. Color Dis 13:526–531 Sobin LH, Gospodarowicz MK,Wittekind C, 2009, TNM classification of malignant tumors, 7th edn.Wiley, New York, pp 100–105 Hamilton SR, Vogelstein B, Kudo S, Riboli E, Nakamura S, Hainaut P, Rubio CA, Sobin LH, Fogt F, Winawer SJ, Goldgar DE, Jass JR, 2000, Carcinoma of the colon and rectum. In: Hamilton SR, Aaltonen LA, eds, World Health Organization Classification of Tumors, Pathology and Genetics of Tumors of the Digestive System, Lyon, pp105–119 Sobin LH, Hermanek P, Hutter RV, 1988, TNM classification of malignant tumors. A comparison between the new, 1987, and the old edition. Cancer 61:2310–2314 Tang R,Wang JY, Chen JS, Chang-Chien CR, Tang S, Lin SE, You YT, Hsu KC, Ho YS, Fan HA, 1995, Survival impact of lymph node metastasis in TNM stage III carcinoma of the colon and rectum. J Am Coll Surg 180:705–712 Suzuki O, Sekishita Y, Shiono T, Ono K, Fujimori M, Kondo S, 2006, Number of lymph node metastases is better predictor of prognosis than level of lymph node metastasis in patients with node-positive colon cancer. J Am Coll Surg 202:732–736 Chen H, Wang Y, Liu H, Hu Y, Zhao L, Li G, Chi P, 2015, Factors influencing apical node metastasis in colorectal cancer patients treated with laparoscopic radical resection with D3 lymphadenectomy: results from two centers in China. Surg Today 45:569–575 Vaccaro CA, Bonadeo FA, BenatiML, QuintanaGM, Rubinstein F, Mullen E, Telenta M, Lastiri JM, 2004, Colorectal cancer staging: reappraisal of N/PN classification. Dis Colon Rectum 47:66–69 Kim JC, Lee KH, CS Y, Kim HC, Kim JR, Chang HM, Kim JH, Kim JS, Kim TW, 2004, The clinicopathological significance of inferior mesenteric lymph node metastasis in colorectal cancer. Eur J Surg Oncol 30:271–279 Gunderson LL, Jessup JM, Sargent DJ, Greene FL, Stewart A, 2010, Revised tumor and node categorization for rectal cancer based on surveillance, epidemiology, and end results and rectal pooled analysis outcomes. J Clin Oncol 28:256–263 Jessup J, Gunderson L, Greene F, 2011, 2010 Staging System for Colon and Rectal Carcinoma. Ann Surg Oncol 18:1513–1517 Yi JW, Lee TG, Lee HS, Heo SC, Jeong SY, Park KJ, Kang SB, 2012, Apical-node metastasis in sigmoid colon or rectal cancer: is it a factor that indicates a poor prognosis after high ligation? Int J Color Dis 27:81–87 Japanese Society for Cancer of the Colon and Rectum, 1998, General Rules for Clinical and Pathologic Studies on Cancer of the Colon, Rectum and Anus, 6th edn. Kanchara, Tokyo, pp 18–29 Liang JT, Huang KC, Lai HS, Lee PH, Sun CT, 2007, Oncologic results of laparoscopic D3 lymphadenectomy for male sigmoid and upper rectal cancer with clinically positive lymph nodes. Ann Surg Oncol 14:1980–1990 Duben J, Dudesek B, Hnatek L, Vazan P, Bakala J, Gatek J, 2010, Lymphatic mapping and biopsy of sentinel lymph nodes using combined methodology of in vivo application of patent blue and radionuclide and ex vivo detection of metastatic affection of lymph nodes in colorectal carcinoma. Rozhl Chir 89:695–701 ShiozawaM, AkikeM, Yamada R, Godai T, Yamamoto N, Saito H, Sugimasa Y, Takemiya S, Rino Y, Imada T, 2007, Clinicopathological features of skip metastasis in colorectal cancer. Hepato-Gastroenterology 54:81–84 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 905 EP 913 DI 10.1007/s12253-017-0381-5 PG 9 ER PT J AU Bartak, KB Kalmar, A Galamb, O Wichmann, B Nagy, BZs Tulassay, Zs Dank, M Igaz, P Molnar, B AF Bartak, Kinga Barbara Kalmar, Alexandra Galamb, Orsolya Wichmann, Barnabas Nagy, Brigitta Zsofia Tulassay, Zsolt Dank, Magdolna Igaz, Peter Molnar, Bela TI Blood Collection and Cell-Free DNA Isolation Methods Influence the Sensitivity of Liquid Biopsy Analysis for Colorectal Cancer Detection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Liquid biopsy; Plasma; cfDNA isolation; Blood collection; Colorectal cancer; DNA methylation ID Liquid biopsy; Plasma; cfDNA isolation; Blood collection; Colorectal cancer; DNA methylation AB During colorectal cancer (CRC) development tumor-derived cell-free DNA (cfDNA) can be released into the bloodstream. Many different cfDNA isolation methods and specific blood collection tubes preventing the release of genomic DNA and stabilizing cfDNA with preservative reagents became available. These factors may affect greatly on the further liquid biopsy analyses. Our aim was to test different blood collection tubes and cfDNA isolation methods to determine whether these factors influence the cfDNA amount and the promoter methylation of four previously described hypermethylated biomarkers. Three manual isolation methods (High Pure Viral Nucleic Acid Large Volume Kit; Epi proColon 2.0 Kit; Quick-cfDNA™ Serum & Plasma Kit) and automated sample preparation systems (InviGenius and InviGenius PLUS) were examined. Furthermore, K3EDTA Vacuette tubes and Streck Cell-Free DNA BCT® tubes were compared. After cfDNA isolation and bisulfite conversion of samples, the methylation level of SFRP1, SFRP2, SDC2, and PRIMA1 were defined with MethyLight assays.We have ascertained that there are differences between the cfDNA amounts depending on the isolation methods. Higher cfDNA yield was observed using InviGenius system than column-based manual isolation method; however, InviGenius PLUS has produced lower cfDNA amounts. No remarkable variance could be found between K3EDTA and Streck tubes; slightly higher cfDNA quantity was detected in 60% of plasma samples using Streck tubes. In point of methylation level and frequency, manual column-based isolation produced more consistent results. Automated cfDNA extraction systems are easy-to-use and high-throughput; however, further improvements in the isolation protocols might lead to the increase of the sensitivity of further methylation analysis. C1 [Bartak, Kinga Barbara] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, H-1088 Budapest, Hungary. [Kalmar, Alexandra] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, H-1088 Budapest, Hungary. [Galamb, Orsolya] Hungarian Academy of Sciences, Molecular Medicine Research Group, H-1088 Budapest, Hungary. [Wichmann, Barnabas] Hungarian Academy of Sciences, Molecular Medicine Research Group, H-1088 Budapest, Hungary. [Nagy, Brigitta Zsofia] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, H-1088 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, H-1088 Budapest, Hungary. [Dank, Magdolna] Semmelweis University, Department of Oncology, H-1083 Budapest, Hungary. [Igaz, Peter] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi str. 46, H-1088 Budapest, Hungary. [Molnar, Bela] Hungarian Academy of Sciences, Molecular Medicine Research Group, H-1088 Budapest, Hungary. RP Bartak, KB (reprint author), Semmelweis University, 2nd Department of Internal Medicine, H-1088 Budapest, Hungary. EM bartak.barbara.kinga@gmail.com CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127(12):2893–2917 Forde KA, 2006, Colonoscopic screening for colon cancer. Surg Endosc 2:S471–S474 Janz T, Lu K, Povlow MR, Urso B, 2016, A review of colorectal cancer detection modalities, stool DNA, and fecal immunochemistry testing in adults over the age of 50. Cureus 8(12):e931 Inadomi JM, 2017, Screening for colorectal neoplasia. N Engl J Med 376(2):149–156 Grady WM, Carethers JM, 2008, Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology 135(4):1079– 1099 Kim MS, Lee J, Sidransky D, 2010, DNA methylation markers in colorectal cancer. Cancer Metastasis Rev 29(1):181–206 Ehrlich M, 2002, DNA methylation in cancer: too much, but also too little. Oncogene 21(35):5400–5413 Kurdyukov S, Bullock M, 2016, DNA methylation analysis: choosing the right method. Biology, Basel, 5(1) Diaz LA Jr, Bardelli A, 2014, Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol 32:579–586 Lam K, Pan K, Linnekamp JF, Medema JP, Kandimalla R, 2016, DNA methylation based biomarkers in colorectal cancer: a systematic review. Biochim Biophys Acta 1866(1):106–120 Schwarzenbach H, Hoon DS, Pantel K, 2011, Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer 11(6): 426–437 van der Vaart M, Pretorius PJ, 2008, Circulating DNA. Its origin and fluctuation. Ann N YAcad Sci 1137:18–26 Thierry AR, Mouliere F, Gongora C, Ollier J, Robert B, Ychou M, Del RioM, Molina F, 2010, Origin and quantification of circulating DNA in mice with human colorectal cancer xenografts. Nucleic Acids Res 38(18):6159–6175 Jung K, Fleischhacker M, Rabien A, 2010, Cell-free DNA in the blood as a solid tumor biomarker–a critical appraisal of the literature. Clin Chim Acta 411(21–22):1611–1624 Malentacchi F, Pizzamiglio S, Verderio P, Pazzagli M, Orlando C, Ciniselli CM, Gunther K, Gelmini S, 2015, Influence of storage conditions and extraction methods on the quantity and quality of circulating cell-free DNA, ccfDNA): the SPIDIA-DNAplas external quality assessment experience. Clin Chem Lab Med 53(12): 1935–1942 Tamkovich SN, Cherepanova AV, Kolesnikova EV, Rykova EY, Pyshnyi DV, Vlassov VV, Laktionov PP, 2006, Circulating DNA and DNase activity in human blood. Ann N YAcad Sci 1075:191– 196 Patutina O, Mironova N, Ryabchikova E, Popova N, Nikolin V, Kaledin V, Vlassov V, Zenkova M, 2011, Inhibition of metastasis development by daily administration of ultralow doses of RNase A and DNase I. Biochimie 93(4):689–696 Patel PS, Patel BP, Rawal RM, Raval GN, Patel MM, Patel JB, Jha FP, Patel DD, 2000, Evaluation of serum alkaline DNase activity in treatment monitoring of head and neck cancer patients. Tumour Biol 21(2):82–89 Xue X, Teare MD, Holen I, Zhu YM, Woll PJ, 2009, Optimizing the yield and utility of circulating cell-free DNA from plasma and serum. Clin Chim Acta 404(2):100–104 Toth K, Bartak BK, Tulassay Z, Molnar B, 2016, Circulating cellfree nucleic acids as biomarkers in colorectal cancer screening and diagnosis. Expert Rev Mol Diagn 16(2):239–252 Bartak BK, Kalmar A, Peterfia B, Patai AV, Galamb O, Valcz G, Spisak S, Wichmann B, Nagy ZB, Toth K, Tulassay Z, Igaz P, Molnar B, 2017, Colorectal adenoma and cancer detection based on altered methylation pattern of SFRP1, SFRP2, SDC2, and PRIMA1 in plasma samples. Epigenetics. , DOI 10.1080/ 15592294.2017.1356957 Medina Diaz I, Nocon A, Mehnert DH, Fredebohm J, Diehl F, Holtrup F, 2016, Performance of Streck cfDNA blood collection tubes for liquid biopsy testing. PLoS One 11(11):e0166354 Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A, 2013, Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 10(8):472–484 Romero A, Acosta-Eyzaguirre D, Sanz J, Moreno F, Serrano G, Diaz-Rubio E, Caldes T, Garcia-Saenz JA, 2015, Identification of E545k mutation in plasma from a PIK3CA wild-type metastatic breast cancer patient by array-based digital polymerase chain reaction: circulating-free DNA a powerful tool for biomarker testing in advance disease. Transl Res 166(6):783–787 Henriksen SD, Madsen PH, Larsen AC, Johansen MB, Drewes AM, Pedersen IS, Krarup H, Thorlacius-Ussing O, 2016, Cellfree DNA promoter hypermethylation in plasma as a diagnostic marker for pancreatic adenocarcinoma. Clin Epigenetics 8:117 Schmidt B, Beyer J, Dietrich D, Bork I, Liebenberg V, FleischhackerM(2015, Quantification of cell-free mSHOX2 plasma DNA for therapy monitoring in advanced stage non-small cell, NSCLC, and small-cell lung cancer, SCLC, patients. PLoS One 10(2):e0118195 Fleischhacker M, Schmidt B, Weickmann S, Fersching DM, Leszinski GS, Siegele B, Stotzer OJ, Nagel D, Holdenrieder S, 2011, Methods for isolation of cell-free plasma DNA strongly effect DNAyield. Clin Chim Acta 412(23–24):2085–2088 Huang DJ, Zimmermann BG, HolzgreveW, Hahn S, 2005, Use of an automated method improves the yield and quality of cell-free fetal DNA extracted from maternal plasma. Clin Chem 51(12): 2419–2420 Kang Q, Henry NL, Paoletti C, Jiang H, Vats P, Chinnaiyan AM, Hayes DF, Merajver SD, Rae JM, Tewari M, 2016, Comparative analysis of circulating tumor DNA stability in K3EDTA, Streck, and CellSave blood collection tubes. Clin Biochem 49(18):1354– 1360 Norton SE, Lechner JM, Williams T, Fernando MR, 2013, A stabilizing reagent prevents cell-free DNA contamination by cellular DNA in plasma during blood sample storage and shipping as determined by digital PCR. Clin Biochem 46(15):1561–1565 Denis MG, Knol AC, Theoleyre S, Vallee A, Dreno B, 2015, Efficient detection of BRAF mutation in plasma of patients after long-term storage of blood in cell-free DNA blood collection tubes. Clin Chem 61(6):886–888 Toro PV, Erlanger B, Beaver JA, Cochran RL, VanDenBerg DA, Yakim E, Cravero K, Chu D, Zabransky DJ, Wong HY, Croessmann S, Parsons H, Hurley PJ, Lauring J, Park BH, 2015, Comparison of cell stabilizing blood collection tubes for circulating plasma tumor DNA. Clin Biochem 48(15):993–998 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 915 EP 923 DI 10.1007/s12253-018-0382-z PG 9 ER PT J AU Kumari, S Husain, N Agarwal, A Neyaz, A Gupta, S Chaturvedi, A Lohani, M Sonkar, AA AF Kumari, Swati Husain, Nuzhat Agarwal, Akash Neyaz, Azfar Gupta, Sameer Chaturvedi, Arun Lohani, Mohtashim Sonkar, Arun Abhinav TI Diagnostic Value of Circulating Free DNA Integrity and Global Methylation Status in Gall Bladder Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gall bladder carcinoma (GBC); Circulating free DNA (cfDNA); cfDNA integrity; Global DNA methylation; Liquid biopsy ID Gall bladder carcinoma (GBC); Circulating free DNA (cfDNA); cfDNA integrity; Global DNA methylation; Liquid biopsy AB The current study investigates the role of circulating free DNA (cfDNA) as a liquid biopsy in diagnosis gall bladder carcinoma (GBC) utilizing levels of long DNA fragments (ALU247) derived from tumor necrosis, short apoptotic fragments (ALU115) denoting total cfDNA and cfDNA integrity denoting ratio of ALU247 and ALU115. The global methylation status of cfDNA was also estimated with the hypothesis that these parameters provide a diagnostic distinction between cancer and non-cancer subjects, with higher or altered values favoring presence of malignancy. Study group included 60 cases of GBC and 36 controls including diseased controls (cholecystitis) and healthy subjects. Median levels of ALU115, ALU247 and cfDNA integrity were significantly different in GBC at 1790.88, 673.75, 0.4718 vs. controls at 840.73, 165.03, 0.1989 ng/ml respectively. Global DNA methylation was not significantly different between GBC at 0.679% and controls at 0.695%. The sensitivity and specificity of ALU 247 in discriminating GBC from controls was highest with a sensitivity, specificity and diagnostic accuracy of 80.0%, 86.1% and 82.2% respectively. Global DNA methylation showed lowest sensitivity of 55.0% and specificity of 50.0%. Clinicopathological parameters showing significant association with cfDNA integrity, on ROC curve analysis, showed significant diagnostic discrimination of the tumor stage, lymphovascular invasion, disease stage and grade histology. This is a first time analysis of ALU115, ALU247 and cfDNA integrity in the diagnosis of GBC and confirms that the combination of ALU247 and cfDNA integrity provides good sensitivity, specificity and diagnostic accuracy in discriminating GBC from controls as well correlates with aggressive disease parameters. C1 [Kumari, Swati] Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Pathology, 226010 Lucknow, U.P., India. [Husain, Nuzhat] Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Pathology, 226010 Lucknow, U.P., India. [Agarwal, Akash] Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Surgical Oncology, 226010 Lucknow, U.P., India. [Neyaz, Azfar] Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Pathology, 226010 Lucknow, U.P., India. [Gupta, Sameer] King George’s Medical University, Department of Surgical Oncology, 226003 Lucknow, U.P., India. [Chaturvedi, Arun] King George’s Medical University, Department of Surgical Oncology, 226003 Lucknow, U.P., India. [Lohani, Mohtashim] Integral University, Department of Biosciences, 226026 Lucknow, U.P., India. [Sonkar, Arun Abhinav] King George’s Medical University, Department of Surgery, 226003 Lucknow, U.P., India. RP Husain, N (reprint author), Dr. Ram Manohar Lohia Institute of Medical Sciences, Department of Pathology, 226010 Lucknow, India. EM drnuzhathusain@hotmail.com CR Goldin RD, Roa JC, 2009, Gallbladder cancer: a morphological and molecular update. Histopathology 55(2):218–229 Gourgiotis S, Kocher HM, Solaini L, Yarollahi A, Tsiambas E, Salemis NS, 2008, Gallbladder cancer. Am J Surg 196(2):252–264 Bazoua G, Hamza N, Lazim T, 2007, Do we need histology for a normal-looking gallbladder? J Hepato-Biliary-Pancreat Surg 14(6): 564–568 Goebel G, Zitt M, Muller HM, 2005, Circulating nucleic acids in plasma or serum, CNAPS, as prognostic and predictive markers in patients with solid neoplasias. Dis Markers 21(3):105–120 Ellinger J, Wittkamp V, Albers P, Perabo FG, Mueller SC, von Ruecker A, Bastian PJ, 2009, Cell-free circulating DNA: diagnostic value in patients with testicular germ cell cancer. J Urol 181(1): 363–371 Su YH, Wang M, Brenner DE, Norton PA, Block TM, 2008, Detection of mutated K-ras DNA in urine, plasma, and serum of patients with colorectal carcinoma or adenomatous polyps. Ann N YAcad Sci 1137:197–206 Kumari S, Tewari S, Husain N, Agarwal A, Pandey A, Singhal A, Lohani M, 2017, Quantification of circulating free DNA as a diagnostic marker in gall bladder cancer. Pathol Oncol Res 23(1):91–97 Umetani N, Giuliano AE, Hiramatsu SH, Amersi F, Nakagawa T, Martino S, Hoon DS, 2006, Prediction of breast tumor progression by integrity of free circulating DNA in serum. J Clin Oncol 24(26): 4270–4276 Umetani N, Kim J, Hiramatsu S, Reber HA, Hines OJ, Bilchik AJ, Hoon DS, 2006, Increased integrity of free circulating DNA in sera of patients with colorectal or periampullary cancer: direct quantitative PCR for ALU repeats. Clin Chem 52(6):1062–1069 Schwarzenbach H, Hoon DS, Pantel K, 2011, Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer 11(6): 426–437 Hwu HR, Roberts JW, Davidson EH, Britten RJ, 1986, Insertion and/or deletion of many repeated DNA sequences in human and higher ape evolution. Proc Natl Acad Sci U S A 83(11):3875–3879 Gu Z, Wang H, Nekrutenko A, Li WH, 2000, Densities, length proportions, and other distributional features of repetitive sequences in the human genome estimated from 430 megabases of genomic sequence. Gene 259:81–88 Giacona MB, Ruben GC, Iczkowski KA, Roos TB, Porter DM, SorensonGD(1998, Cell-freeDNAin human blood plasma: length measurements in patients with pancreatic cancer and healthy controls. Pancreas 17(1):89–97 Wyllie AH, 1980, Glucocorticoid-induced thymocyte apoptosis is associated with endogenous endonuclease activation. Nature 284(5756):555–556 Jin Z, El-Deiry WS, 2005, Overview of cell death signaling pathways. Cancer Biol Ther 4(2):139–163 Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R, 2001, DNA fragments in the blood plasma of cancer patients: quantitation and evidence for their origin from apoptotic and necrotic cells. Cancer Res 61(4):1659–1665 Merlo A, Herman JG, Mao L, Lee DJ, Gabrielson E, Burger PC, Baylin SB, Sidransky D, 1995, 5' CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/ CDKN2/MTS1 in human cancers. Nat Med 1(7):686–692 Herman JG, Umar A, Polyak K, Graff JR, Ahuja N, Issa JP, Markowitz S, Willson JK, Hamilton SR, Kinzler KW, Kane MF, Kolodner RD, Vogelstein B, Kunkel TA, Baylin SB, 1998, Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma. Proc Natl Acad Sci U S A 95(12):6870–6875 Muller HM, Widschwendter A, Fiegl H, Ivarsson L, Goebel G, Perkmann E, Marth C,WidschwendterM(2003, DNAmethylation in serum of breast cancer patients: an independent prognostic marker. Cancer Res 63(22):7641–7645 Mori T, O’Day SJ, Umetani N, Martinez RS, Kitago M, Koyanagi K, Kuo C, Takeshima TL, Milford R, Wang JH, Vu DV, Nguyen SL, Hoon D, 2005, Predictive utility of circulating methylated DNA in serum of melanoma patients receiving biochemotherapy. J Clin Oncol 23(36):9351–9358 Laird PW, Jaenisch R, 1996, The role of DNA methylation in cancer genetic and epigenetics. Annu Rev Genet 30:441–464 Dagan T, Sorek R, Sharon E, Ast G, Graur D, 2004, Alu Gene: a database of Alu elements incorporated within protein-coding genes. Nucleic Acids Res 32:D489–D492 Parpart-Li S, Bartlett B, Popoli M, Adleff V, Tucker L, Steinberg R, Georgiadis A, Phallen J, Brahmer JR, Azad NA, Browner I, Laheru DA, Velculescu VE, Sausen M, Diaz LA, 2016, The effect of preservative and temperature on the analysis of circulating tumor DNA. Clin Cancer Res. , DOI 10.1158/1078-0432.CCR- 16-1691 UtomoWK, Janmaat VT, Verhaar AP, Cros J, Levy P, Ruszniewski P, den Berg MS, Jenster G, Bruno MJ, Braat H, Fuhler GM, Peppelenbosch MP, 2016, DNA integrity as biomarker in pancreatic cyst fluid. Am J Cancer Res 6(8):1837–1841 Agostini M, Pucciarelli S, Enzo MV, Del Bianco P, Briarava M, Bedin C, Maretto I, Friso ML, Lonardi S, Mescoli C, Toppan P, Urso E, Nitti D, 2011, Circulating cell-free DNA: a promising marker of pathologic tumor response in rectal cancer patients receiving preoperative chemoradiotherapy. Ann Surg Oncol 18(9): 2461–2468 Stroun M, Maurice P, Vasioukhin V, Lyautey J, Lederrey C, Lefort F, Rossier A, Chen XQ, Anker P, 2000, The origin and mechanism of circulating DNA. Ann N YAcad Sci 906:161–168 Diaz LA Jr, Bardelli A, 2014, Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol 32(6):579–586 Kamat AA, Sood AK, Dang D, Gershenson DM, Simpson JL, Bischoff FZ, 2006, Quantification of total plasma cell-free DNA in ovarian cancer using real-time PCR. Ann N Y Acad Sci 1075: 230–234 Chiu TW, Young R, Chan LY, Burd A, Lo DY, 2006, Plasma cellfree DNA as an indicator of severity of injury in burn patients. Clin Chem Lab Med 44(1):13–17 Zeerleder S, Zwart B,WuilleminWA, Aarden LA, Groeneveld AB, Caliezi C, van Nieuwenhuijze AE, van Mierlo GJ, Eerenberg AJ, Lammle B, Hack CE, 2003, Elevated nucleosome levels in systemic inflammation and sepsis. Crit Care Med 31(7):1947–1951 Holdenrieder S, Eichhorn P, Beuers U, Samtleben W, Schoenermarck U, Zachoval R, Nagel D, Stieber P, 2006, Nucleosomal DNA fragments in autoimmune diseases. Ann N Y Acad Sci 1075:318–327 Ohira T, Sakai K, Matsubayashi J, Kajiwara N, Kakihana M, Hagiwara M, Hibi M, Yoshida K, Maeda J, Ohtani K, Nagao T, Nishio K, Ikeda N, 2016, Tumor volume determines the feasibility of cell-free DNA sequencing for mutation detection in non-small cell lung cancer. Cancer Sci 107(11):1660–1666 Agostini M, Enzo MV, Bedin C, Belardinelli V, Goldin E, Del Bianco P, Maschietto E, D'Angelo E, Izzi L, Saccani A, Zavagno G, Nitti D, 2012, Circulating cell-free DNA: a promising marker of regional lymphonode metastasis in breast cancer patients. Cancer Biomark 11(2-3):89–98 de Vos T, Molnar B, 2017, Screening for colorectal cancer based on the promoter methylation status of the septin 9 gene in plasma cell free DNA. J Clin Epigenet 3:1. , DOI 10.21767/2472- 1158.100040 Toth K, Galamb O, Spisak S, Wichmann B, Sipos F, Valcz G, Leiszter K, Molnar B, Tulassay Z, 2011, The influence of methylated septin 9 gene on RNA and protein level in colorectal cancer. Pathol Oncol Res 17(3):503–509 Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, Bencardino K, Cercek A, Chen CT, Veronese S, Zanon C, Sartore-Bianchi A, Gambacorta M, Gallicchio M, Vakiani E, Boscaro V, Medico E, Weiser M, Siena S, Di Nicolantonio F, Solit D, Bardelli A, 2012, Emergence of KRAS mutations and acquired resistance to anti- EGFR therapy in colorectal cancer. Nature 486(7404):532–536 Diaz LA,Williams RT,Wu J, Kinde I, Hecht JR, Berlin J, Allen B, Bozic I, Reiter JG, NowakMA, Kinzler KW, Oliner KS, Vogelstein B, 2012, The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 486(7404):537–540 Tang Q, Cheng J, Cao X, Surowy H, Burwinkel B, 2016, Bloodbased DNA methylation as biomarker for breast cancer: a systematic review. Clin Epigenetics 8:115 Wong IH, Lo YM, Zhang J, Liew CT, Ng MH, Wong N, Lai PB, Lau WY, Hjelm NM, Johnson PJ, 1999, Detection of aberrant p16 methylation in the plasma and serum of liver cancer patients. Cancer Res 59(1):71–73 Silva JM, Dominguez G, Villanueva MJ, Gonzalez R, Garcia JM, Corbacho C, Provencio M, Espana P, Bonilla F, 1999, Aberrant DNA methylation of the p16INK4a gene in plasma DNA of breast cancer patients. Br J Cancer 80(8):1262–1264 Goessl C, Krause H, Muller M, Heicappell R, Schrader M, Sachsinger J, Miller K, 2000, Fluorescent methylation-specific polymerase chain reaction forDNA-based detection of prostate cancer in bodily fluids. Cancer Res 60(21):5941–5945 Ehrlich M, 2002, DNA methylation in cancer: too much, but also too little. Oncogene 21(35):5400–5413 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 925 EP 936 DI 10.1007/s12253-017-0380-6 PG 12 ER PT J AU Wang, M An, Sh Wang, D Ji, H Geng, M Guo, X Wang, Z AF Wang, Ming An, Shuhong Wang, Diyi Ji, Haizhen Geng, Min Guo, Xingjing Wang, Zhaojin TI Quantitative Proteomics Identify the Possible Tumor Suppressive Role of Protease-Activated Receptor-4 in Esophageal Squamous Cell Carcinoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Proteomic; Protease-activated receptors-4; Apoptosis; Tumor suppressor; Esophageal squamous cell carcinoma ID Proteomic; Protease-activated receptors-4; Apoptosis; Tumor suppressor; Esophageal squamous cell carcinoma AB Exposure to carcinogens of tobacco smoke may result in methylation of protease-activated receptors-4 (PAR4) gene and further induces the loss of PAR4 expression, which is considered to be involved in carcinogenesis of esophageal squamous cell carcinoma (ESCC). Here we employed a TMT-based quantitative proteomic approach to identify PAR4-regulated changes of proteomic profiles in ESCC cells and to identify potentially therapeutic value. A total of 33 proteins were found significantly changed with 15 up-regulated and 18 down-regulated in PAR4-activating peptide (PAR4-AP) treated ESCC cells compared with controls. Bioinformatics analysis showed that key higher expressed proteins included those associated with apoptosis and tumor suppressor (e.g. CASP9), and lower expressed proteins included those associated with anti-apoptosis, autophagy and promoting cell proliferation (e.g. CHMP1B, PURA, PARG and HIST1H2AH).Western blot verified changes in five representative proteins including CASP9, CHMP1B, PURA, PARG and HIST1H2AH. Immunohistochemistry analysis showed that CHMP1B, PURA, PARG and HIST1H2AH expression in ESCC tissues were significantly higher than those in adjacent nontumorous tissues. Our findings will be helpful in further investigations into the functions and molecular mechanisms of PAR4 in ESCC. C1 [Wang, Ming] Taishan Medical University, Department of Human Anatomy, 2 Ying Sheng Dong Lu, 271000 Taian, China. [An, Shuhong] Taishan Medical University, Department of Human Anatomy, 2 Ying Sheng Dong Lu, 271000 Taian, China. [Wang, Diyi] Affiliated Hospital of Taishan Medical University, Department of Pathology, 271000 Taian, China. [Ji, Haizhen] Taishan Medical University, Department of Physiology, 271000 Taian, China. [Geng, Min] Taishan Medical University, Department of Human Anatomy, 2 Ying Sheng Dong Lu, 271000 Taian, China. [Guo, Xingjing] Taishan Medical University, Department of Physiology, 271000 Taian, China. [Wang, Zhaojin] Taishan Medical University, Department of Human Anatomy, 2 Ying Sheng Dong Lu, 271000 Taian, China. RP Wang, Z (reprint author), Taishan Medical University, Department of Human Anatomy, 271000 Taian, China. EM zjwang@tsmc.edu.cn CR Ramachandran R, Hollenberg MD, 2008, Proteinases and Signalling: pathophysiological and therapeutic implications via PARs and more. Br J Pharmacol 153:S263–S282 Elste AP, Petersen I, 2010, Expression of proteinase-activated receptor 1-4, PAR 1-4, in human cancer. J Mol Histol 41:89–99 Ohashi S, Miyamoto S, Kikuchi O, Goto T, Amanuma Y, Muto M, 2015, Recent advances from basic and clinical studies of esophageal squamous cell carcinoma. Gastroenterology 149:1700–1715 Steevens J, Schouten LJ, Goldbohm RA, van den Brandt PA, 2010, Alcohol consumption, cigarette smoking and risk of subtypes of oesophageal and gastric cancer: a prospective cohort study. Gut 59:39–48 Tanaka F, Yamamoto K, Suzuki S, Inoue H, Tsurumaru M, Kajiyama Y, Kato H, Igaki H, Furuta K, Fujita H, Tanaka T, Tanaka Y, Kawashima Y, Natsugoe S, Setoyama T, Tokudome S, Mimori K, Haraguchi N, Ishii H, MoriM(2010, Strong interaction between the effects of alcohol consumption and smoking on oesophageal squamous cell carcinoma among individuals with ADH1B and/or ALDH2 risk alleles. Gut 59:1457–1464 Fasanelli F, Baglietto L, Ponzi E, Guida F, Campanella G, Johansson M, Grankvist K, Johansson M, Assumma MB, Naccarati A, Chadeau-Hyam M, Ala U, Faltus C, Kaaks R, Risch A, de Stavola B, Hodge A, Giles GG, Southey MC, Relton CL, Haycock PC, Lund E, Polidoro S, Sandanger TM, Severi G, Vineis P, 2015, Hypomethylation of smoking-related genes is associated with future lung cancer in four prospective cohorts. Nat Commun 6: 10192 Lee S, Jiang P, Wang W, Feng W, Yu G, 2014, The decreased expression of protease-activated receptor 4 in esophageal squamous carcinoma. Neoplasma 61:546–552 Vergnolle N, 2000, Review article: proteinase-activated receptors - novel signals for gastrointestinal pathophysiology. Aliment Pharmacol Ther 14:257–266 Zhou C, Zhong Q, Rhodes LV, Townley I, Bratton MR, Zhang Q, Martin EC, Elliott S, Collins-Burow BM, Burow ME, Wang G, 2012, Proteomic analysis of acquired tamoxifen resistance in MCF-7 cells reveals expression signatures associated with enhanced migration. Breast Cancer Res 14:R45 Cox J, Mann M, 2008, MaxQuant enables high peptide identification rates, individualized p.P.B.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol 26:1367– 1372 Kim B, Srivastava SK, Kim SH, 2015, Caspase-9 as a therapeutic target for treating cancer. Expert Opin Ther Targets 19:113–127 Loeser H, Waldschmidt D, Kuetting F, Schallenberg S, Zander T, Bollschweiler E, Hoelscher A, Weckermann K, Plum P, Alakus H, Buettner R, Quaas A, 2017, Somatic BRCA1-associated protein 1, BAP1, loss is an early and rare event in esophageal adenocarcinoma. Mol Clin Oncol 7:225–228 Zhou J, Yang J, Fan X, Hu S, Zhou F, Dong J, Zhang S, Shang Y, Jiang X, Guo H, Chen N, Xiao X, Sheng J, Wu K, Nie Y, Fan D, 2016, Chaperone-mediated autophagy regulates proliferation by targeting RND3 in gastric cancer. Autophagy 12:515–528 Lin Z, Guo Z, Xu Y, Zhao X, 2014, Identification of a secondary promoter of CASP8 and its related transcription factor PURα. Int J Oncol 45:57–66 Shahmoradgoli M, Mannherz O, Engel F, Heck S, Kramer A, Seiffert M, Pscherer A, Lichter P, 2011, Antiapoptotic function of charged multivesicular body protein 5: a potentially relevant gene in acute myeloid leukemia. Int J Cancer 128:2865–2871 Wang H, Wang L, Erdjument-Bromage H, Vidal M, Tempst P, Jones RS, Zhang Y, 2004, Role of histone H2A ubiquitination in Polycomb silencing. Nature 431:873–878 Zhang Y, Yu G, Jiang P, Xiang Y, Li W, Lee W, Zhang Y, 2011, Decreased expression of protease-activated receptor 4 in human gastric cancer. Int J Biochem Cell Biol 43:1277–1283 Bhatnagar S, Gazin C, Chamberlain L, Ou J, Zhu X, Tushir JS, Virbasius CM, Lin L, Zhu LJ, Wajapeyee N, Green MR, 2014, TRIM37 is a new histone H2A ubiquitin ligase and breast cancer oncoprotein. Nature 516:116–120 Liu H, Johnson EM, 2002, Distinct proteins encoded by alternative transcripts of the PURG gene, located contrapodal to WRN on chromosome 8, determined by differential termination/ polyadenylation. Nucleic Acids Res 30:2417–2426 Spitzer C, Li F, Buono R, Roschzttardtz H, Chung T, Zhang M, Osteryoung KW, Vierstra RD, Otegui MS, 2015, The endosomal protein CHARGEDMULTIVESICULAR BODY PROTEIN1 regulates the autophagic turnover of plastids in Arabidopsis. Plant Cell 27:391–402 Hu B, Jiang D, Chen Y,Wei L, Zhang S, Zhao F, Ni R, Lu C,Wan C, 2015, High CHMP4B expression is associated with accelerated cell proliferation and resistance to doxorubicin in hepatocellular carcinoma. Tumour Biol 36:2569–2581 Michael JV, Wurtzel JGT, Mao GF, Rao AK, Kolpakov MA, Sabri A, Hoffman NE, Rajan S, Tomar D, Madesh M, Nieman MT, Yu J, Edelstein LC, Rowley JW, Weyrich AS, Goldfinger LE, 2017, Platelet microparticles infiltrating solid tumors transfer miRNAs that suppress tumor growth. Blood 130:567–580 Kolpakov MA, Rafiq K, Guo X, Hooshdaran B,Wang T, Vlasenko L, Bashkirova YV, Zhang X, Chen X, Iftikhar S, Libonati JR, Kunapuli SP, Sabri A, 2016, Protease-activated receptor 4 deficiency offers cardioprotection after acute ischemia reperfusion injury. J Mol Cell Cardiol 90:21–29 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 937 EP 943 DI 10.1007/s12253-018-0395-7 PG 7 ER PT J AU Semushina, GS Aronov, AD Moiseeva, VE AF Semushina, G Svetlana Aronov, A Dmitry Moiseeva, V Ekaterina TI Local Interleukin-2 Immunotherapy of Breast Cancer: Benefit and Risk in a Spontaneous Mouse Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Interleukin-2; 3S–paradigm; Spontaneous mouse model; BLRB ID Breast cancer; Interleukin-2; 3S–paradigm; Spontaneous mouse model; BLRB AB Earlier, naturally arising mammary cancer in BLRB female mice was shown to reproduce some key pathological characteristics of the familial set of human breast cancer. Then we advanced a novel 3S–paradigm of anticancer research that helped to develop selection criteria and to estimate benefit/risk of local interleukin-2 (IL-2) effects in this spontaneous mouse model. In this paper, the efficacy of single and triple local IL-2 doses is compared using properly selected murine BLRB females based on our previously published data. Only BLRB females bearing spontaneous mammary tumors without subclinical period were used. The tumor growth rate and recipient survival of single and triple IL-2 applications were compared with corresponding parameter values of untreated control. Tumor growth rate was decreased in both experimental groups versus control parameter values. Single IL-2 application resulted in a significant prolongation of the average survival time while triple application caused acute tumor rejection in some females decreasing the survival time of the rest of the recipients. As a result, proper treatment protocol in accurately selected females allowed increasing the complete response rate to 14% in spontaneous mouse model of breast cancer. In conclusion, our approaches may demonstrate the principle methodology developing preselection procedure for breast cancer patients for local IL-2 therapy application. C1 [Semushina, G Svetlana] Russian Academy of Sciences, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya str., 16/10, GSP-7, 117997 Moscow, Russian Federation. [Aronov, A Dmitry] Russian Academy of Sciences, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya str., 16/10, GSP-7, 117997 Moscow, Russian Federation. [Moiseeva, V Ekaterina] Russian Academy of Sciences, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya str., 16/10, GSP-7, 117997 Moscow, Russian Federation. RP Moiseeva, VE (reprint author), Russian Academy of Sciences, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russian Federation. EM evmoise@gmail.com CR Dhupkar P, Gordon N, 2017, Interleukin-2: old and new approaches to enhance immune-therapeutic efficacy. Adv Exp Med Biol 995:33–51. , DOI 10.1007/978-3-319-53156-4_2 Adler A, Stein JA, Kedar E, Naor D, Weiss DW, 1984, Intralesional injection of interleukin-2-expanded autologous lymphocytes in melanoma and breast cancer patients: a pilot study. J Biol Response Mod 3:491–500 Israel L, Cour V, Pihan I, Morere JF, Breau JL, Franks CR, Palmer P, Loriaux E, 1989, Some theoretical and practical limitations of interleukin-2. Ten cases of advanced breast cancer treated with continuous infusion of IL-2. Cancer Treat Rev 16(Suppl A):169–171 Li Y, Zhou L, Sun B, Li X, Duan K, Wu Y, Ouyang W, 2015, Interleukin-2 administration after modified radical mastectomy in breast cancer therapy increases peripheral regulatory T cells. Int J Clin Exp Med 8:7816–7822 Nicolini A, Carpi A, Ferrari P, Biava MP, Rossi G, 2016, Immunotherapy and hormone-therapy in metastatic breast cancer: a review and an update. Curr Drug Targets 17:1127–1139 Den Otter W, Jacobs JJ, Battermann JJ, Hordijk GJ, Krastev Z, Moiseeva EV, Stewart RJ, Ziekman PG, Koten JW, 2008, Local therapy of cancer with free IL-2. Cancer Immunol Immunother 57: 931–950. , DOI 10.1007/s00262-008-0455-z Li X, Lu P, Li B, Zhang W, Yang R, Chu Y, Luo K, 2017, Interleukin 2 and interleukin 10 function synergistically to promote CD8+ T cell cytotoxicity, which is suppressed by regulatory T cells in breast cancer. Int J Biochem Cell Biol 87:1–7. , DOI 10. 1016/j.biocel.2017.03.003 Moiseyeva E, Semushina S, Chaadaeva A, Kessler J, 2010, Theoretical and applied implications of dual interleukin-2 nature: single local application in a mouse breast cancer model. Int J Appl Fundam Res 4:18–23 Rojas G, Infante YC, Pupo A, Carmenate T, 2014, Fine epitope specificity of antibodies against interleukin-2 explains their paradoxical immunomodulatory effects. MAbs 6:273–285. https://doi. org/10.4161/mabs.27224 Paciotti GF, Tamarkin L, 1988, Interleukin-2 differentially affects the proliferation of a hormone-dependent and a hormoneindependent human breast cancer cell line in vitro and in vivo. Anticancer Res 8:1233–1239 Katano M, Matsuo T, Morisaki T, Naito K, Nagumo F, Kubota E, Nakamura M, Hisatsugu T, Tadano J, 1994, Increased proliferation of a human breast carcinoma cell line by recombinant interleukin-2. Cancer Immunol Immunother 39:161–166 Moiseeva EV, Rapoport EM, Bovin NV, Miroshnikov AI, Chaadaeva AV, Krasilshschikova MS, Bojenko VK, Bijleveld C, van Dijk JE, Den Otter W, 2005, Galectins as markers of aggressiveness of mouse mammary carcinoma: towards a lectin target therapy of human breast cancer. Breast Cancer Res Treat 91:227– 241. , DOI 10.1007/s10549-005-0289-8 Moiseeva E, 2009, Anti-breast cancer drug testing. Original approaches. Novel set of mouse models. Lambert Academic Publishing, Saarbrucken Dialla PO, Dabakuyo TS, Marilier S, Gentil J, Roignot P, Darut- Jouve A, Poillot ML, Quipourt V, Arveux P, 2012, Populationbased study of breast cancer in older women: prognostic factors of relative survival and predictors of treatment. BMC Cancer 12: 472. , DOI 10.1186/1471-2407-12-472 Vargo-Gogola T, Rosen JM, 2007, Modelling breast cancer: one size does not fit all. Nat Rev Cancer 7:659–672. , DOI 10. 1038/nrc2193 Moiseeva EV, Semushina SG, Skrabelinskaya EI, Sadovnikova ES, Pakhomova IA, Den Otter W, 2010, Effect of triple interleukin-2 application in spontaneous mouse model of breast cancer. Cytokines Inflamm 9:27–31 Moiseeva EV, Semushina SG, Chaadaeva AV, Sadovnikova ES, Kessler IuV, 2010, Criteria for the effectiveness of interleukin-2 immunotherapy in a spontaneous murine mammary tumor model. Vopr Onkol 56:443–449 Aronov DA, Semushina SG,Moiseeva EV, 2017, Prognostic value of immunological blood parameters in the mouse model of familial breast cancer. Mod Probl Sci Educ. , DOI 10.17513/spno. 26945 Rawal R, Bertelsen L, Olsen JH, 2006, Cancer incidence in firstdegree relatives of a population-based set of cases of early-onset breast cancer. Eur J Cancer 42:3034–3040. , DOI 10.1016/ j.ejca.2006.04.025 Moiseeva EV, Merkulova IB, Bijleveld C, Koten JW, Miroshnikov AI, Den OtterW(2003, Therapeutic effect of a single peritumoural dose of IL-2 on transplanted murine breast cancer. Cancer Immunol Immunother 52:487–496. , DOI 10.1007/s00262-003- 0385-8 Hisamatsu K, Toge T, Yanagawa E, Hamada Y, Seto Y, Yamada H, Toi M, Hattori T, 1986, Immune reactivities of lymphocytes from peripheral blood, regional lymph nodes and tumor-infiltrating lymphocytes in breast cancer. Gan To Kagaku Ryoho 13:2555–2561 Inokuma M, dela Rosa C, Schmitt C, Haaland P, Siebert J, Petry D, Tang M, Suni MA, Ghanekar SA, Gladding D, Dunne JF, Maino VC, Disis ML, Maecker HT, 2007, Functional T cell responses to tumor antigens in breast cancer patients have a distinct phenotype and cytokine signature. J Immunol 179:2627–2633. , DOI 10.4049/jimmunol.179.4.2627 Wanebo HJ, Pace R, Hargett S, Katz D, Sando J, 1986, Production of and response to interleukin-2 in peripheral blood lymphocytes of cancer patients. Cancer 57:656–662 Vaage J, 1987, Local and systemic effects during interleukin-2 therapy of mouse mammary tumors. Cancer Res 47:4296–4298 Ames IH, 2004, Chemoimmunotherapy of murine mammary adenocarcinomas. Anticancer Res 24:2249–2256 Lee JC, Kim DC, Gee MS, Saunders HM, Sehgal CM, Feldman MD, Ross SR, LeeWM(2002, Interleukin-12 inhibits angiogenesis and growth of transplanted but not in situ mouse mammary tumor virus-induced mammary carcinomas. Cancer Res 62:747–755 AtkinsMB, LotzeMT, Dutcher JP, Fisher RI,Weiss G, Margolin K, Abrams J, Sznol M, Parkinson D, Hawkins M, Paradise C, Kunkel L, Rosenberg SA, 1999, High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 17:2105–2116. , DOI 10.1200/JCO.1999.17.7.2105 Sokol DK, 2013, BFirst do no harm^ revisited. BMJ 347:f6426– f6426. , DOI 10.1136/bmj.f6426 Frei E 3rd, 1985, Curative cancer chemotherapy. Cancer Res 45: 6523–6537 Arrington AK, Goldstein L, Kruper L, Vito C, Yim J, Chen SL, 2014, Life expectancy after curative-intent treatment of breast cancer: impact on long-term follow-up care. Am Surg 80:604–609 Cherny NI, Sullivan R, Dafni U, Kerst JM, Sobrero A, Zielinski C, de Vries EGE, Piccart MJ, 2015, A standardised, generic, validated approach to stratify the magnitude of clinical benefit that can be anticipated from anti-cancer therapies: the European Society for Medical Oncology magnitude of clinical benefit scale, ESMOMCBS). Ann Oncol 26:1547–1573. , DOI 10.1093/ annonc/mdv249 Jiang X, Shapiro DJ, 2014, The immune system and inflammation in breast cancer. Mol Cell Endocrinol 382:673–682. , DOI 10.1016/j.mce.2013.06.003 Gala RR,Martino S,Wiejowski SA, Kresge CL, del Mar AlonsoM, 1989, Serum interleukin 2, IL-2, levels and prolactin bioactive/ radioimmunoassay ratios of women at risk for breast cancer. Eur J Cancer Clin Oncol 25:1603–1605 Erdei E, Kang H, Meisner A, White K, Pickett G, Baca C, Royce M, BerwickM(2010, Polymorphisms in cytokine genes and serum cytokine levels among new Mexican women with and without breast cancer. Cytokine 51:18–24. , DOI 10.1016/j.cyto. 2010.03.014 Ponce R, 2008, Adverse consequences of immunostimulation. J Immunotoxicol 5:33–41. h t t p s : / / d o i . o rg/ 1 0 .1080/ 15476910801897920 Shrikant P, Mescher MF, 2002, Opposing effects of IL-2 in tumor immunotherapy: promoting CD8 T cell growth and inducing apoptosis. J Immunol 169:1753–1759. , DOI 10.4049/ jimmunol.169.4.1753 Moiseeva EV, Semushina SG, Bojenko VK, 2011, Towards personalized paradigm of experimental anticancer research with B3S^ concept. EPMA J 2:114–115. , DOI 10.1007/s13167-011- 0119-5 Moiseeva EV, Aronov DA, Semushina SG, Bozhenko VK, 2014, Advantages of a new paradigm of experimental oncology: individual prognosis of immunotherapy outcome based on surrogate marker analysis. Mol Med 2:47–50 Characiejus D, Hodzic J, Jacobs JJL, 2010, BFirst do no harm^ and the importance of prediction in oncology. EPMA J 1:369–375 Croci S, Pedrazzi G, Passeri G, Delsignore R, Ortalli I, 2002, Red cell Hb oxidation of healthy subjects compared to breast cancer patients. Anticancer Res 22:2903–2906 Mishra S, Sharma DC, Sharma P, 2004, Studies of biochemical parameters in breast cancer with and without metastasis. Indian J Clin Biochem 19:71–75. , DOI 10.1007/BF02872394 Moiseeva EV, Semushina SG, Aronov DA, Murashev AN, Shishkin AM, Bojenko VK, 2012, Interleukin-2 immunotherapy against transplanted mammary cancer: survival time depends on initial hematological status. Vestn RNCRR 4:27 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 945 EP 951 DI 10.1007/s12253-018-0396-6 PG 7 ER PT J AU Mo, JS Park, YR Chae, SCh AF Mo, Ji-Su Park, Young-Ran Chae, Soo-Cheon TI MicroRNA 196B Regulates HOXA5, HOXB6 and GLTP Expression Levels in Colorectal Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MicroRNA; MIR196B; HOXA5; GLTP; Colorectal cancer ID MicroRNA; MIR196B; HOXA5; GLTP; Colorectal cancer AB MiRNAs are non-coding RNAs that play important roles in the pathogenesis of human diseases by regulating target gene expression in specific cells or tissues. Previously we identified colorectal cancer (CRC) associated MIR196B, which was specifically up-regulated in CRC cells and tissue. We also identified 18 putative MIR196B target genes by comparing the mRNAs down-regulated in MIR196B-overexpressed cells with MIR196B target genes predicted by public bioinformatics tools. In this study, we verified the association between MIR196B and three genes, HOXA5, HOXB6 and GLTP. HOXA5, HOXB6 and GLTP transcripts were directly down-regulated by MIR196B. The mRNA and proteins levels of HOXA5, HOXB6 and GLTP were also down-regulated in CRC cells by the up-regulated MIR196B. GLTP protein expression was decreased in CRC tissues compared to adjacent non-tumor tissues. These results suggest that HOXA5, HOXB6, and GLTP were direct target genes of MIR196B in CRC cells, and that the up-regulated MIR196B in CRC tissue regulates the expression levels of HOXA5, HOXB6, and GLTP during colorectal carcinogenesis. C1 [Mo, Ji-Su] Wonkwang University, School of Medicine, Department of Pathology, 54538 Iksan, Chonbuk, South Korea. [Park, Young-Ran] Wonkwang University, School of Medicine, Department of Pathology, 54538 Iksan, Chonbuk, South Korea. [Chae, Soo-Cheon] Wonkwang University, School of Medicine, Department of Pathology, 54538 Iksan, Chonbuk, South Korea. RP Chae, SCh (reprint author), Wonkwang University, School of Medicine, Department of Pathology, 54538 Iksan, South Korea. EM chaesc@wku.ac.kr CR Ambros V, 2004, The functions of animal microRNAs. Nature 431:350–355. , DOI 10.1038/nature02871 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116:281–297. , DOI 10.1016/S0092- 8674(04)00045-5 Garzon R, Fabbri M, Cimmino A, Calin GA, Croce CM, 2005, MicroRNA expression and function in cancer. Trends Mol Med 12:580–587. , DOI 10.1016/j.molmed.2006.10.006 Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR, 2005, MicroRNA expression profiles classify human cancers. Nature 435:834–838. , DOI 10.1038/nature03702 Brennecke J, Hipfner DR, Stark A, Russell RB, Cohen SM, 2003, Bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in drosophila. Cell 113:25–36. , DOI 10.1016/S0092- 8674(03)00231-9 Ma L, Teruya-Feldstein J, Weinberg RA, 2007, Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature 449:682–688. , DOI 10.1038/nature06174 Su H, Yang JR, Xu T, Huang J, Xu L, Yuan Y, Zhuang SM, 2009, MicroRNA-101, down-regulated in hepatocellular carcinoma, promotes apoptosis and suppresses tumorigenicity. Cancer Res 69: 1135–1142. , DOI 10.1158/0008-5472.CAN-08-2886 Mo JS, Alam KJ, Kang IH, Park WC, Seo GS, Choi SC, Kim HS, Moon HB, Yun KJ, Chae SC, 2015, MicroRNA 196B regulates FAS-mediated apoptosis in colorectal cancer cells. Oncotarget 6: 2843–2855. , DOI 10.18632/oncotarget.3066 Yekta S, Shih IH, Bartel DP, 2004, MicroRNA-directed cleavage of HOXB8 mRNA. Science 304:594–596. , DOI 10.1126/ science.1097434 Tanzer A, Amemiya CT, Kim CB, Stadler PF, 2005, Evolution of microRNAs located within Hox gene clusters. J Exp Zool B Mol Dev Evol 304:75–85. , DOI 10.1002/jez.b.21021 Mo JS, Alam KJ, Kim HS, Lee YM, Yun KJ, Chae SC, 2016, MicroRNA 429 regulates mucin gene expression and secretion in murine model of colitis. J Crohns Colitis 10:850–859. https://doi. org/10.1093/ecco-jcc/jjw033 Alam KJ, Mo JS, Han SH, Park WC, Kim HS, Yun KJ, Chae SC, 2017, MicroRNA 375 regulates proliferation and migration of colon cancer cells by suppressing the CTGF-EGFR signaling pathway. Int J Cancer 141:1614–1629. , DOI 10.1002/ijc.30861 Li Z, Huang H, Chen P, He M, Li Y, Arnovitz S, Jiang X, He C, Hyjek E, Zhang J, Zhang Z, Elkahloun A, Cao D, Shen C, Wunderlich M, Wang Y, Neilly MB, Jin J, Wei M, Lu J, Valk PJ, Delwel R, Lowenberg B, Le Beau MM, Vardiman J, Mulloy JC, Zeleznik-Le NJ, Liu PP, Zhang J, Chen J, 2012, miR-196b directly targets both HOXA9/MEIS1 oncogenes and FAS tumour suppressor in MLL-rearranged leukaemia. Nat Commun 3:688. https://doi. org/10.1038/ncomms1681 Liu Y, Zheng W, Song Y, Ma W, Yin H, 2013, Low expression of miR-196b enhances the expression of BCR-ABL1 and HOXA9 oncogenes in chronic myeloid leukemogenesis. PLoS One 8: e68442. , DOI 10.1371/journal.pone.0068442 How C, Hui AB, Alajez NM, Shi W, Boutros PC, Clarke BA, Yan R, Pintilie M, Fyles A, Hedley DW, Hill RP, Milosevic M, Liu FF, 2013, MicroRNA-196b regulates the Homeobox B7-vascular endothelial growth factor Axis in cervical cancer. PLoS One 8: e67846. , DOI 10.1371/journal.pone.0067846 Li Y, Zhang M, Chen H, Dong Z, Ganapathy V, Thangaraju M, Huang S, 2010, Ratio of miR-196s to HOXC8 mRNA correlates with breast cancer cell migration and metastasis. Cancer Res 70: 7894–7904. , DOI 10.1158/0008-5472.CAN-10-1675 Raman V, Martensen SA, Reisman D, Evron E, Odenwald WF, Jaffee E, Marks J, Sukumar S, 2000, Compromised HOXA5 function can limit p53 expression in human breast tumours. Nature 405: 974–978. , DOI 10.1038/35016125 Henderson GS, van Diest PJ, Burger H, Russo J, Raman V, 2006, Expression pattern of a homeotic gene:HOXA5, in normal breast and in breast tumors. Cell Oncol 28:305–313. , DOI 10. 1155/2006/974810 Gendronneau G, Lemieux M, Morneau M, Paradis J, Tetu B, Frenette N, Aubin J, Jeannotte L, 2010, Influence of Hoxa5 on p53 TumorigenicOutcome in mice. Am J Pathol 176:995–1005. , DOI 10.2353/ajpath.2010.090499 Mandeville I, Aubin J, LeBlanc M, Lalancette-Hebert M, Janelle MF, Tremblay GM, Jeannotte L, 2006, Impact of the loss of Hoxa5 function on lung alveogenesis. Am J Pathol 169:1312–1327. , DOI 10.2353/ajpath.2006.051333 Packer AI, Mailutha KG, Ambrozewicz LA,Wolgemuth DJ, 2000, Regulation of the Hoxa4 and Hoxa5 genes in the embryonic mouse lung by retinoic acid and TGFbeta1: implications for lung development and patterning. Dev Dyn 217:62–74. , DOI 10.1002/, SICI)1097-0177(200001)217:1<62::AID-DVDY6>3.0.CO;2-U Golpon HA, Geraci MW, Moore MD, Miller HL, Miller GJ, Tuder RM, Voelkel NF, 2001, HOX genes in human lung: altered expressionin primary pulmonary hypertension and emphysema. Am J Pathol 158(3):955–966. , DOI 10.1016/S0002- 9440(10)64042-4 Liu XH, Lu KH, Wang KM, Sun M, Zhang EB, Yang JS, Yin DD, Liu ZL, Zhou J, Liu ZJ, De W, Wang ZX, 2012, MicroRNA-196a promotes non-small cell lung cancer cell proliferation and invasion through targetingHOXA5.BMC Cancer 12:348. , DOI 10. 1186/1471-2407-12-348 Mattjus P, Pike HM, Molotkovsky JG, Brown RE, 2000, Charged membrane surfaces impede the protein mediated transfer of glycosphingolipids between phospholipid bilayers. Biochemistry 39:1067–1075. , DOI 10.1021/bi991810u Tuuf J, Mattjus P, 2007, Human glycolipid transfer protein–intracellular localization and effects on the sphingolipid synthesis. Biochim Biophys Acta 1771:1353–1363. , DOI 10.1016/ j.bbalip.2007.09.001 Gao Y, Chung T, Zou X, Pike HM, Brown RE, 2011, Human glycolipid transfer protein, GLTP, expressionmodulates cell shape. PLoS One 6:e19990. , DOI 10.1371/journal.pone.0019990 Zou X, Chung T, Lin X, Malakhova ML, Pike HM, Brown RE, 2008, Human glycolipid transfer protein, GLTP, genes: organization, transcriptional status and evolution. BMC Genomics 9:72. , DOI 10.1186/1471-2164-9-72 Zou X, Gao Y, Ruvolo VR, Gardner TL, Ruvolo PP, Brown RE, 2011, Human glycolipid transfer protein gene, GLTP, expression is regulated by Sp1 and Sp3. J Biol Chem 286:1301–1311. https:// doi.org/10.1074/jbc.M110.127837 Kjellberg MA, Backman AP, Ohvo-Rekila H, Mattjus P, 2014, Alternation in the glycolipid transfer protein expression causes changes in the cellular Lipidome. PLoS One 9:e97263. https://doi. org/10.1371/journal.pone.0097263 Mattjus P, 2009, Glycolipid transfer proteins and membrane interaction. Biochim Biophys Acta 1788:267–272. , DOI 10. 1016/j.bbamem.2008.10.003 Kojima N, Hakomori S, 1991, Cell adhesion, spreading, and motility of GM3- expressing cells based on glycolipid-glycolipid interaction. J Biol Chem 266:17552–17558 Boggs JM, Menikh A, Rangaraj G, 2000, Trans interaction between galactosyl ceramide and cerebroside sulphate across opposed bilayers. Biophys J 78:874–885. , DOI 10.1016/S0006- 3495(00)76645-8 Schnaar RL, 2004, Glycolipid-mediated cell-cell recognition in inflammation and nerve regeneration. Arch Biochem Biophys 426: 163–172. , DOI 10.1016/j.abb.2004.02.019 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 953 EP 959 DI 10.1007/s12253-018-0399-3 PG 7 ER PT J AU Azevedo, PA Silva, NS Reichert, A Lima, F Junior, E Rueff, J AF Azevedo, P Ana Silva, N Susana Reichert, Alice Lima, Fernando Junior, Esmeraldina Rueff, Jose TI The Role of Caspase Genes Polymorphisms in Genetic Susceptibility to Philadelphia-Negative Myeloproliferative Neoplasms in a Portuguese Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Philadelphia-negative myeloproliferative neoplasms; Genetic susceptibility; Caspase genes polymorphisms; Janus kinase 2 ID Philadelphia-negative myeloproliferative neoplasms; Genetic susceptibility; Caspase genes polymorphisms; Janus kinase 2 AB Our main aim was to evaluate the role of caspases’ genes SNPs in Philadelphia-chromosome negative chronic myeloproliferative neoplasms (PN-MPNs) susceptibility. A case-control study in 133 Caucasian Portuguese PN-MPNs patients and 281 matched controls was carried out, studying SNPs in apoptosis related caspases: rs1045485 and rs1035142 (CASP8), rs1052576, rs2308950, rs1132312 and rs1052571 (CASP9), rs2227309 and rs2227310 (CASP7) and rs13006529 (CASP10). After stratification by pathology diagnosis for essential thrombocythemia (ET), female gender or JAK2 positive, there is a significant increased risk for those carrying at least one variant allele for CASP9 (C653T) polymorphism (OR 2.300 CI 95% [1.180– 4.484], P = 0.014). However, when considered individually, none of the studied caspases polymorphisms was associated with PN-MPNs risk. Our results do not reveal a significant involvement of caspase genes polymorphisms on the individual susceptibility towards PN-MPNs as a whole. However, for essential thrombocythemia (ET), female gender or JAK2 positive, there is a significant increased risk to those carrying at least one variant allele for CASP9. Although larger studies are required to confirm these results and to provide conclusive evidence of association between these and other caspases variants and PN-MPNs susceptibility, these new data may contribute to a best knowledge of the pathophysiology of these disorders and, in the future, to a more rational and efficient choice of therapeutic strategies to be adopted in PN-MPNs treatment. C1 [Azevedo, P Ana] Nova Medical School / Faculdade de Ciencias Medicas, Universidade Nova de Lisboa, Centre for Toxicogenomics and Human Health (ToxOmics), Campo dos Martires da Patria, 130, 1169-056 Lisbon, Portugal. [Silva, N Susana] Nova Medical School / Faculdade de Ciencias Medicas, Universidade Nova de Lisboa, Centre for Toxicogenomics and Human Health (ToxOmics), Campo dos Martires da Patria, 130, 1169-056 Lisbon, Portugal. [Reichert, Alice] Centro Hospitalar de Lisboa Ocidental (CHLO), Hospital de S. Francisco Xavier, Department of Clinical Haematology, Estrada Forte do Alto do Duque, 1449-005 Lisbon, Portugal. [Lima, Fernando] Centro Hospitalar de Lisboa Ocidental (CHLO), Hospital de S. Francisco Xavier, Department of Clinical Haematology, Estrada Forte do Alto do Duque, 1449-005 Lisbon, Portugal. [Junior, Esmeraldina] Centro Hospitalar de Lisboa Ocidental (CHLO), Hospital de S. Francisco Xavier, Department of Clinical Pathology, Estrada Forte do Alto do Duque, 1449-005 Lisbon, Portugal. [Rueff, Jose] Nova Medical School / Faculdade de Ciencias Medicas, Universidade Nova de Lisboa, Centre for Toxicogenomics and Human Health (ToxOmics), Campo dos Martires da Patria, 130, 1169-056 Lisbon, Portugal. RP Silva, NS (reprint author), Nova Medical School / Faculdade de Ciencias Medicas, Universidade Nova de Lisboa, Centre for Toxicogenomics and Human Health (ToxOmics), 1169-056 Lisbon, Portugal. EM snsilva@nms.unl.pt;jose.rueff@nms.unl.pt CR Swerdlow CESH, Harris NL et al, 2008, WHO classification of Tumours of Haematopoieticand lymphoid tissues., 4 edition, October 27, 2008, ed. World Health Organization, Lyon Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW, 2016, The 2016 revision to theWorld Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127:2391–2405 James C, Ugo V, Le Couedic JP, Staerk J, Delhommeau F, Lacout C, Garcon L, Raslova H, Berger R, Bennaceur-Griscelli A, Villeval JL, Constantinescu SN, Casadevall N, Vainchenker W, 2005, A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 434:1144–1148 Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg JR, Tichelli A, Cazzola M, Skoda RC, 2005, A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 352: 1779–1790 Levine RL,Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, Boggon TJ, Wlodarska I, Clark JJ, Moore S, Adelsperger J, Koo S, Lee JC, Gabriel S, Mercher T, D'Andrea A, Frohling S, Dohner K, Marynen P, Vandenberghe P, Mesa RA, Tefferi A, Griffin JD, Eck MJ, Sellers WR, Meyerson M, Golub TR, Lee SJ, Gilliland DG, 2005, Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 7:387–397 Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin N, Scott MA, Erber WN, Green AR, Project CG, 2005, Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 365:1054–1061 Scott LM, Tong W, Levine RL, Scott MA, Beer PA, Stratton MR, Futreal PA, Erber WN, McMullin MF, Harrison CN, Warren AJ, Gilliland DG, Lodish HF, Green AR, 2007, JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med 356:459–468 Tognon R, Nunes NeS, Castro FA, 2013, Apoptosis deregulation in myeloproliferative neoplasms. Einstein, Sao Paulo, 11:540–544 Tefferi A, Pardanani A, 2015, Myeloproliferative neoplasms: a contemporary review. JAMA Oncol 1:97–105 Nangalia J, Massie CE, Baxter EJ, Nice FL, Gundem G, Wedge DC, Avezov E, Li J, Kollmann K, Kent DG, Aziz A, Godfrey AL, Hinton J, Martincorena I, Van Loo P, Jones AV, Guglielmelli P, Tarpey P, Harding HP, Fitzpatrick JD, Goudie CT, Ortmann CA, Loughran SJ, Raine K, Jones DR, Butler AP, Teague JW, O'Meara S, McLaren S, Bianchi M, Silber Y, Dimitropoulou D, Bloxham D, Mudie L, Maddison M, Robinson B, Keohane C, Maclean C, Hill K, Orchard K, Tauro S, DuMQ, Greaves M, Bowen D, Huntly BJ, Harrison CN, Cross NC, Ron D, Vannucchi AM, Papaemmanuil E, Campbell PJ, Green AR, 2013, Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med 369:2391–2405 Rueff J, Rodrigues AS, 2016, Cancer drug resistance: a brief overview from a genetic viewpoint. Methods Mol Biol 1395:1–18 Rice KL, Lin X, Wolniak K, Ebert BL, Berkofsky-Fessler W, Buzzai M, Sun Y, Xi C, Elkin P, Levine R, Golub T, Gilliland DG, Crispino JD, Licht JD, Zhang W, 2011, Analysis of genomic aberrations and gene expression profiling identifies novel lesions and pathways in myeloproliferative neoplasms. Blood Cancer J 1:e40 Valentino L, Pierre J, 2006, JAK/STAT signal transduction: regulators and implication in hematological malignancies. Biochem Pharmacol 71:713–721 Bolufer P, Barragan E, Collado M, Cervera J, Lopez JA, Sanz MA, 2006, Influence of genetic polymorphisms on the risk of developing leukemia and on disease progression. Leuk Res 30:1471–1491 Delhommeau F, Jeziorowska D, Marzac C, Casadevall N, 2010, Molecular aspects of myeloproliferative neoplasms. Int J Hematol 91:165–173 Beer PA, Delhommeau F, LeCouedic JP, Dawson MA, Chen E, Bareford D, Kusec R, McMullin MF, Harrison CN, Vannucchi AM, Vainchenker W, Green AR, 2010, Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. Blood 115:2891–2900 Kilpivaara O, Levine RL, 2008, JAK2 and MPL mutations in myeloproliferative neoplasms: discovery and science. Leukemia 22: 1813–1817 Bjorkholm M, Hultcrantz M, Derolf A, 2014, Leukemic transformation in myeloproliferative neoplasms: therapy-related or unrelated? Best Pract Res Clin Haematol 27:141–153 Levine RL, 2009, Mechanisms of mutations in myeloproliferative neoplasms. Best Pract Res Clin Haematol 22:489–494 Campregher PV, Santos FP, Perini GF, Hamerschlak N, 2012, Molecular biology of Philadelphia-negative myeloproliferative neoplasms. Rev Bras Hematol Hemoter 34:150–155 Goldar S, Khaniani MS, Derakhshan SM, Baradaran B, 2015, Molecular mechanisms of apoptosis and roles in cancer development and treatment. Asian Pac J Cancer Prev 16:2129–2144 Kiraz Y, Adan A, Kartal Yandim M, Baran Y, 2016, Major apoptotic mechanisms and genes involved in apoptosis. Tumour Biol 37:8471–8486 Zaman S, Wang R, Gandhi V, 2014, Targeting the apoptosis pathway in hematologic malignancies. Leuk Lymphoma 55:1980–1992 Green DR, Llambi F, 2015, Cell death signaling. Cold Spring Harb Perspect Biol 7 Nunes NS, Tognon R, Moura LG, Kashima S, Covas DT, Santana M, Souto EX, Zanichelli MA, Simoes BP, Souza AM, Castro FA, 2013, Differential expression of apoptomiRs in myeloproliferative neoplasms. Leuk Lymphoma 54:2047–2051 Tognon R, Gasparotto EP, Leroy JM, Oliveira GL, Neves RP, Carrara ReC, Kashima S, Covas DT, Santana M, Souto EX, Zanichelli MA, Velano CE, Simoes BP, Alberto FL, Miyashiro K, de Souza AM, Amarante-Mendes GP, de Castro FA, 2011, Differential expression of apoptosis-related genes from death receptor pathway in chronic myeloproliferative diseases. J Clin Pathol 64:75–82 Tognon R,Gasparotto EP, Neves RP, Nunes NS, Ferreira AF, Palma PV, Kashima S, Covas DT, Santana M, Souto EX, Zanichelli MA, Simoes BP, de Souza AM, Castro FA, 2012, Deregulation of apoptosis-related genes is associated with PRV1 overexpression and JAK2 V617F allele burden in essential thrombocythemia and myelofibrosis. J Hematol Oncol 5:2 Tognon R, Nunes NS, Ambrosio L, Souto EX, Perobelli L, Simoes BP, Souza MC, Chauffaille MeL, Attie de Castro F, 2016, Apoptosis- and cell cycle-related genes methylation profile in myeloproliferative neoplasms. Leuk Lymphoma 57:1201–1204 Olsson M, Zhivotovsky B, 2011, Caspases and cancer. Cell Death Differ 18:1441–1449 Mambet C, Matei L, Necula LG, Diaconu CC, 2016, A link between the driver mutations and dysregulated apoptosis in BCRABL1 negative myeloproliferative neoplasms. J Immunoassay Immunochem 37:331–345 Tefferi A, Vardiman JW, 2008, Classification and diagnosis of myeloproliferative neoplasms: the 2008 World Health Organization criteria and point-of-care diagnostic algorithms. Leukemia 22:14–22 Riedl SJ, Salvesen GS, 2007, The apoptosome: signalling platform of cell death. Nat Rev Mol Cell Biol 8:405–413 Cho SG, Choi EJ, 2002, Apoptotic signaling pathways: caspases and stress-activated protein kinases. J Biochem Mol Biol 35:24–27 Philchenkov A, ZavelevichM, Kroczak TJ, LosM(2004, Caspases and cancer: mechanisms of inactivation and new treatment modalities. Exp Oncol 26:82–97 Ng PW, Porter AG, Janicke RU, 1999, Molecular cloning and characterization of two novel pro-apoptotic isoforms of caspase- 10. J Biol Chem 274:10301–10308 Oliver L, Vallette FM, 2005, The role of caspases in cell death and differentiation. Drug Resist Updat 8:163–170 Petit E, Oliver L,Vallette FM(2009, Themitochondrial outer membrane protein import machinery: a new player in apoptosis? Front Biosci, Landmark Ed, 14:3563–3570 Green DR, Kroemer G, 2004, The pathophysiology of mitochondrial cell death. Science 305:626–629 Wang X, 2001, The expanding role of mitochondria in apoptosis. Genes Dev 15:2922–2933 Peter ME, Krammer PH, 2003, The CD95(APO-1/Fas, DISC and beyond. Cell Death Differ 10:26–35 Creagh EM, Conroy H, Martin SJ, 2003, Caspase-activation pathways in apoptosis and immunity. Immunol Rev 193:10–21 Ding HF, Lin YL, McGill G, Juo P, Zhu H, Blenis J, Yuan J, Fisher DE, 2000, Essential role for caspase-8 in transcription-independent apoptosis triggered by p53. J Biol Chem 275:38905–38911 Testa U, 2004, Apoptotic mechanisms in the control of erythropoiesis. Leukemia 18:1176–1199 Malherbe JA, Fuller KA, Mirzai B, Kavanagh S, So CC, Ip HW, Guo BB, Forsyth C, Howman R, Erber WN, 2016, Dysregulation of the intrinsic apoptotic pathway mediates megakaryocytic hyperplasia in myeloproliferative neoplasms. J Clin Pathol 69(11):1017-1024 Lan Q, Morton LM, Armstrong B, Hartge P, Menashe I, Zheng T, Purdue MP, Cerhan JR, Zhang Y, Grulich A, Cozen W, Yeager M, Holford TR, Vajdic CM, Davis S, Leaderer B, Kricker A, Schenk M, Zahm SH, Chatterjee N, Chanock SJ, Rothman N, Wang SS, 2009, Genetic variation in caspase genes and risk of non-Hodgkin lymphoma: a pooled analysis of 3 population-based case-control studies. Blood 114:264–267 Kelly JL, Novak AJ, Fredericksen ZS, Liebow M, Ansell SM, Dogan A, Wang AH, Witzig TE, Call TG, Kay NE, Habermann TM, Slager SL, Cerhan JR, 2010, Germline variation in apoptosis pathway genes and risk of non-Hodgkin's lymphoma. Cancer Epidemiol Biomark Prev 19:2847–2858 Park JY, Park JM, Jang JS, Choi JE, Kim KM, Cha SI, Kim CH, Kang YM, Lee WK, Kam S, Park RW, Kim IS, Lee JT, Jung TH, 2006, Caspase 9 promoter polymorphisms and risk of primary lung cancer. Hum Mol Genet 15:1963–1971 Lin J, Lu C, Stewart DJ, Gu J, Huang M, Chang DW, Lippman SM, Wu X, 2012, Systematic evaluation of apoptotic pathway gene polymorphisms and lung cancer risk. Carcinogenesis 33: 1699–1706 Theodoropoulos GE, Michalopoulos NV, Pantou MP, Kontogianni P, Gazouli M, Karantanos T, Lymperi M, Zografos GC, 2012, Caspase 9 promoter polymorphisms confer increased susceptibility to breast cancer. Cancer Genet 205:508–512 Liamarkopoulos E, Gazouli M, Aravantinos G, Tzanakis N, Theodoropoulos G, Rizos S, Nikiteas N, 2011, Caspase 8 and caspase 9 gene polymorphisms and susceptibility to gastric cancer. Gastric Cancer 14:317–321 Florena AM, Tripodo C, Di Bernardo A, Iannitto E, Guarnotta C, Porcasi R, Ingrao S, Abbadessa V, Franco V, 2009, Different immunophenotypical apoptotic profiles characterise megakaryocytes of essential thrombocythaemia and primary myelofibrosis. J Clin Pathol 62:331–338 Lindholm Sorensen A, Hasselbalch HC, 2015, Smoking and Philadelphia-negative chronic myeloproliferative neoplasms. Eur J Haematol 97(1):63-69 Hasselbalch HC, 2015, Smoking as a contributing factor for development of polycythemia vera and related neoplasms. Leuk Res 39: 1137–1145 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 961 EP 969 DI 10.1007/s12253-018-0411-y PG 9 ER PT J AU Urbanovska, I Megova, HM Dwight, Z Kalita, O Uvirova, M Simova, J Tuckova, L Buzrla, P Palecek, T Hajduch, M Dvorackova, J Drabek, J AF Urbanovska, Irena Megova, Houdova Magdalena Dwight, Zachary Kalita, Ondrej Uvirova, Magdalena Simova, Jarmila Tuckova, Lucie Buzrla, Petr Palecek, Tomas Hajduch, Marian Dvorackova, Jana Drabek, Jiri TI IDH Mutation Analysis in Glioma Patients by CADMA Compared with SNaPshot Assay and two Immunohistochemical Methods SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE IDH1; IDH2; CADMA; Glioma; Mutation testing ID IDH1; IDH2; CADMA; Glioma; Mutation testing AB Mutations in IDH1/2 genes are a marker of good prognosis for glioma patients, associated with low grade gliomas and secondary glioblastomas. Immunohistochemistry and Sanger sequencing are current standards for IDH1/2 genotyping while many other methods exist. The aim of this study was to validate Competitive amplification of differentially melting amplicons (CADMA) PCR for IDH genotyping by comparison with SNaPshot assay and two immunohistochemical methods. In our study, 87 glioma patients (46 from Olomouc and 41 from Ostrava) were analyzed. IDH1/2 mutations in native bioptical samples were analyzed at DNA level by CADMA and SNaPshot while IDH1 mutations in FFPE samples were analyzed at protein level by two IHC methods. CADMA PCR sensitivity for IDH1 was 96.4% and specificity 100% for 86 concluded samples. SNaPshot assay sensitivity was 92.9% and specificity of 100% for 85 concluded samples. IHC in the laboratory no. 2 reached sensitivity 85.7% and specificity 100% for 86 concluded samples. IHC in the laboratory no. 4 reached sensitivity of 96.4% and specificity of 79.7% in 74 concluded samples. Only one IDH2 mutation was found by SNaPshot while CADMA yielded false negative result. In conclusion, CADMA is a valid method for IDH1 p.(R132H) testing with higher sensitivity than SNaPshot assay. Also, molecular genetic methods of IDH1 testing from native samples were more robust than IHC from FFPE. C1 [Urbanovska, Irena] CGB Laboratory Inc.Ostrava, Czech Republic. [Megova, Houdova Magdalena] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Hnevotinska 5, 779 00 Olomouc, Czech Republic. [Dwight, Zachary] University of Utah, Department of PathologySalt Lake City, UT, USA. [Kalita, Ondrej] University Hospital Olomouc, Department of NeurosurgeryOlomouc, Czech Republic. [Uvirova, Magdalena] CGB Laboratory Inc.Ostrava, Czech Republic. [Simova, Jarmila] CGB Laboratory Inc.Ostrava, Czech Republic. [Tuckova, Lucie] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Department of Clinical and Molecular PathologyOlomouc, Czech Republic. [Buzrla, Petr] University of Ostrava, Faculty of Medicine and University Hospital, Institute of Pathology, Syllabova 19, 703 00 Ostrava, Czech Republic. [Palecek, Tomas] University Hospital Ostrava, Neurosurgery ClinicOstrava, Czech Republic. [Hajduch, Marian] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Hnevotinska 5, 779 00 Olomouc, Czech Republic. [Dvorackova, Jana] University of Ostrava, Faculty of Medicine, Department of Biomedical SciencesOstrava, Czech Republic. [Drabek, Jiri] Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Hnevotinska 5, 779 00 Olomouc, Czech Republic. RP Drabek, J (reprint author), Palacky University and University Hospital in Olomouc, Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, 779 00 Olomouc, Czech Republic. EM jiri_drabek@seznam.cz CR Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL, Olivi A, McLendon R, Rasheed BA, Keir S, Nikolskaya T, Nikolsky Y, Busam DA, Tekleab H, Diaz LA Jr, Hartigan J, Smith DR, Strausberg RL, Marie SK, Shinjo SM, Yan H, Riggins GJ, Bigner DD, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, KinzlerKW(2008, An integrated genomic analysis of human glioblastoma multiforme. Science 321:1807–1812 Hempel JM, Bidsas S, Schittenhelm J, Brendle C, Bender B, Wassmann H, Skardelly M, Tabatabai G, Vega SC, Ernemann U, Klose U, 2016, In vivo molecular profiling of human glioma using diffusion kurtosis imaging. J Neuro-Oncol 131:93–101 Nesvick CL, Zhang C, Edwards NA, Montgomery BK, Lee M, Yang C, Wang H, Zhu D, Heiss JD, Merrill MJ, Ray-Chaudhury A, Zhuang Z, 2016, ZEB1 expression is increased in IDH1-mutant lower-grade gliomas. J Neuro-Oncol 130:111–122 Yang Y, Mao Q,Wang X, Liu Y, Mao Y, Zhou Q, Luo J, 2016, An analysis of 170 glioma patients and systematic review to investigate the association between IDH-1 mutations and preoperative gliomarelated epilepsy. J Clin Neurosci 31:56–62 Rodriguez FJ, Vizcaino MA, Lin MT, 2016, Recent advances on the molecular pathology of glial neoplasms in children and adults. J Mol Diagn 18:620–634 Turkalp Z, Karamchandani J, Das S, 2014, IDH mutation in glioma: new insights and promises for the future. JAMA Neurol 71: 1319–1325 Kingsbury JM, Shamaprasad N, Billmyre RB, Heitman J, Cardenas ME, 2016, Cancer-associated isocitrate dehydrogenase mutations induce mitochondrial DNA instability. Hum Mol Genet 25:3524–3538 Masui K, CaveneeWK,Mischel PS, 2016, Cancer metabolism as a central driving force of glioma pathogenesis. Brain Tumor Pathol 33:161–168 Cairns RA, Mak TW, 2013, Oncogenic isocitrate dehydrogenase mutations: mechanisms, models, and clinical opportunities. Cancer Discov 3:730–741 Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel-Wahab O, Edwards CR, Khanin R, Figueroa ME, Melnick A, Wellen KE, O'Rourke DM, Berger SL, Chan TA, Levine RL, Mellinghoff IK, Thompson CB, 2012, IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 483:474–478 Johannessen TA, Mukherjee J, Viswanath P, Ohba S, Ronen SM, Bjerkvig R, Pieper RO, 2016, Rapid conversion of mutant IDH1 from driver to passenger in a model of human gliomagenesis. Mol Cancer Res 14:976–983 Capper D, Weissert S, Balss J, Habel A, Meyer J, Jager D, Ackermann U, Tessmer C, Korshunov A, Zentgraf H, Hartmann C, von Deimling A, 2010, Characterization of R132H mutationspecific IDH1 antibody binding in brain tumors. Brain Pathol 20: 245–254 Balss J, Meyer J, Mueller W, Korshunov A, Hartmann C, von Deimling A, 2008, Analysis of the IDH1 codon 132 mutation in brain tumors. Acta Neuropathol 116:597–602 Felsberg J, Wolter M, Seul H, Friedensdorf B, Goppert M, Sabel MC, Reifenberger G, 2010, Rapid and sensitive assessment of the IDH1 and IDH2 mutation status in cerebral gliomas based on DNA pyrosequencing. Acta Neuropathol 119:501–507 Setty P, Hammes J, Rothamel T, Vladimirova V, Kramm CM, Pietsch T, Waha A, 2010, A pyrosequencing-based assay for the rapid detection of IDH1 mutations in clinical samples. JMol Diagn 12:750–756 Horbinski C, Kelly L, Nikiforov YE, Durso MB, Nikiforova MN, 2010, Detection of IDH1 and IDH2 mutations by fluorescence melting curve analysis as a diagnostic tool for brain biopsies. J Mol Diagn 12:487–492 Aibaidula A, Zhao W, Wu JS, Chen H, Shi ZF, Zheng LL, Mao Y, Zhou LF, Sui GD, 2016, Microfluidics for rapid detection of isocitrate dehydrogenase 1 mutation for intraoperative application. J Neurosurg 124:1611–1618 Meyer J, Pusch S, Balss J, Capper D, Mueller W, Christians A, Hartmann C, von Deimling A, 2010, PCR- and restriction endonuclease-based detection of IDH1 mutations. Brain Pathol 20:298–300 Perizzolo M, Winkfein B, Hui S, Krulicki W, Chan JA, Demetrick DJ, 2012, IDH mutation detection in formalin-fixed paraffin-embedded gliomas using multiplex PCR and single-base extension. Brain Pathol 22:619–624 Boisselier B, Marie Y, Labussiere M, Ciccarino P, Desestret V, Wang X, Capelle L, Delattre JY, Sanson M, 2010, COLD PCR HRM: a highly sensitive detection method for IDH1 mutations. Hum Mut 31:1360–1365 Zacher A, Kaulich K, Stepanow S,Wolter M, Kohrer K, Felsberg J, Malzkorn B, Reifenberger G, 2016, Molecular diagnostics of gliomas using next generation sequencing of a glioma-tailored gene panel. Brain Pathol 27:146–159 Polivka J, Polivka J Jr, Rohan V, Pesta M, Repik T, Pitule P, Topolcan O, 2014, Isocitrate dehydrogenase-1 mutations as prognostic biomarker in glioblastoma multiforme patients in West Bohemia. Biomed Res Int 2014:735659 van den Bent MJ, Hartmann C, PreusserM, Strobel T, Dubbink HJ, Kros JM, von Deimling A, Boisselier B, Sanson M, Halling KC, Diefes KL, Aldape K, Giannini C, 2013, Interlaboratory comparison of IDH mutation detection. J Neuro-Oncol 112:173–178 Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P, 2007, The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114:97–109 Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella- Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW, 2016, The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol 131:803–820 Houdova Megova M, Drabek J, Dwight Z, Trojanec R, Koudelakova V, Vrbkova J, Kalita O, Mlcochova S, Rabcanova M, Hajduch M, 2017, Isocitrate dehydrogenase mutations are better prognostic marker than O6-methylguanine-DNA methyltransferase promoter methylation in glioblastomas - a retrospective, single-Centre molecular genetics study of gliomas. Klinicka onkologie 30:361–371 Agarwal S, Sharma MC, Jha P, Pathak P, Suri V, Sarkar C, Chosdol K, Suri A, Kale SS, Mahapatra AK, Jha P, 2013, Comparative study of IDH1 mutations in gliomas by immunohistochemistry and DNA sequencing. Neuro-Oncology 15:718–726 Ramos-Vara JA, Miller MA, 2014, When tissue antigens and antibodies get along: revisiting the technical aspects of immunohistochemistry–the red, brown, and blue technique. Vet Pathol 51:42–87 Preusser M, Wohrer A, Stary S, Hoftberger R, Streubel B, Hainfellner JA, 2011, Value and limitations of immunohistochemistry and gene sequencing for detection of the IDH1- R132H mutation in diffuse glioma biopsy specimens. J Neuropathol Exp Neurol 70:715–723 Loussouarn D, Le Loupp AG, Frenel JS, Leclair F, von Deimling A, Aumont M, Martin S, Campone M, Denis MG, 2012, Comparison of immunohistochemistry, DNA sequencing and allele-specific PCR for the detection of IDH1 mutations in gliomas. Int J Oncol 40:2058–2062 Gilbert MT, Haselkorn T, Bunce M, Sanchez JJ, Lucas SB, Jewell LD,Van Marck E,WorobeyM(2007, The isolation of nucleic acids from fixed, paraffin-embedded tissues-which methods are useful when? P ONE 2:e537 Huijsmans CJ, Damen J, van der Linden JC, Savelkoul PH, Hermans MH, 2010, Comparative analysis of four methods to extract DNA from paraffin-embedded tissues: effect on downstream molecular applications. BMC Res Notes 3:239 Takano S, Tian W, Matsuda M, Yamamoto T, Ishikawa E, Kaneko MK, Yamazaki K, Kato Y,Matsumura A, 2011, Detection of IDH1 mutation in human gliomas: comparison of immunohistochemistry and sequencing. Brain Tumor Pathol 28:115–123 Kaneko MK, Tsujimoto Y, Hozumi Y, Goto K, Kato Y, 2013, Novel monoclonal antibodies GMab-r1 and LMab-1 specifically recognize IDH1-R132G and IDH1-R132L mutations. Monoclon Antib Immunodiagn Immunother 32:224–228 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 971 EP 978 DI 10.1007/s12253-018-0413-9 PG 8 ER PT J AU Leyh-Bannurah, SR Trudel, D Latour, M Zaffuto, E Grosset, AA Tam, Ch Ouellet, V Graefen, M Budaus, L Aprikian, GA Lacombe, L Fleshner, EN Gleave, EM Mes-Masson, AM Saad, F Karakiewicz, IP AF Leyh-Bannurah, Sami-Ramzi Trudel, Dominique Latour, Mathieu Zaffuto, Emanuele Grosset, Andree-Anne Tam, Christine Ouellet, Veronique Graefen, Markus Budaus, Lars Aprikian, G Armen Lacombe, Louis Fleshner, E Neil Gleave, E Martin Mes-Masson, Anne-Marie Saad, Fred Karakiewicz, I Pierre TI A Multi-Institutional Validation of Gleason Score Derived from Tissue Microarray Cores SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Radical prostatectomy; Prostate cancer; Gleason grade agreement; Biochemical recurrence; Validation ID Radical prostatectomy; Prostate cancer; Gleason grade agreement; Biochemical recurrence; Validation AB To test the agreement between high-grade PCa at RP and TMA, and the ability of TMA to predict BCR. Validation of concordance between tissue microarray (TMA) and radical prostatectomy (RP) high-grade prostate cancer (PCa) is crucial because latter determines the treated natural history of PCa. We hypothesized that TMA Gleason score is in agreement with RP pathology and capable of accurately predicting biochemical recurrence (BCR). Data were provided from a multi-institutional Canadian sample of 1333 TMA and RP specimens with complete clinicopathological data. First, rate of agreement between TMA and high-grade Gleason at RP or biopsy and RP was tested. Second, ability of RP, TMA and biopsy to predict BCR was compared. Multivariable (MVA) Cox regression models were fitted and BCR rates were illustrated with Kaplan-Meier plots. Agreement between RP and TMA and between RP and biopsy was 72.6% (95% CI:69.7–75.5) and 60.4% (95% CI:57.2–63.6), respectively. In MVA predicting BCR, the accuracy for RP, TMA and biopsy was 0.73, 0.72 and 0.68, respectively. TMA added discriminatory ability among exclusively low-grade Gleason RP patients (p = 0.02), but did not improve BCR discrimination in exclusive high-grade PCa RP patients (p = 0.8). TMA Gleason grade accurately reflects presence of high-grade Gleason in RP specimen, accurately predicts BCR rates after RP and improves prediction of BCR in low-grade Gleason patients at RP. C1 [Leyh-Bannurah, Sami-Ramzi] University of Montreal Health Center, Cancer Prognostics and Health Outcomes UnitMontreal, Canada. [Trudel, Dominique] Universite de Montreal, Department of Pathology and Cellular BiologyMontreal, Canada. [Latour, Mathieu] Universite de Montreal, Department of Pathology and Cellular BiologyMontreal, Canada. [Zaffuto, Emanuele] University of Montreal Health Center, Cancer Prognostics and Health Outcomes UnitMontreal, Canada. [Grosset, Andree-Anne] l’Universite de Montreal, Institut du Cancer de Montreal and Centre de recherche du centre hospitalierMontreal, QC, Canada. [Tam, Christine] l’Universite de Montreal, Institut du Cancer de Montreal and Centre de recherche du centre hospitalierMontreal, QC, Canada. [Ouellet, Veronique] l’Universite de Montreal, Institut du Cancer de Montreal and Centre de recherche du centre hospitalierMontreal, QC, Canada. [Graefen, Markus] Martini-Clinic, Prostate Cancer Center Hamburg-EppendorfHamburg, Germany. [Budaus, Lars] Martini-Clinic, Prostate Cancer Center Hamburg-EppendorfHamburg, Germany. [Aprikian, G Armen] McGill University, Research Institute of McGill University Health Center and Department of Surgery (Urology)Montreal, QC, Canada. [Lacombe, Louis] CHU de QuebecQuebec, Quebec, Canada. [Fleshner, E Neil] University of Toronto, University Health Network, Department of SurgeryToronto, ON, Canada. [Gleave, E Martin] University of British Columbia, Vancouver Prostate Centre & Department of Urologic ScienceVancouver, BC, Canada. [Mes-Masson, Anne-Marie] Universite de Montreal, Department of MedecineMontreal, Canada. [Saad, Fred] Universite de Montreal, Department of SurgeryMontreal, Canada. [Karakiewicz, I Pierre] University of Montreal Health Center, Cancer Prognostics and Health Outcomes UnitMontreal, Canada. RP Leyh-Bannurah, SR (reprint author), University of Montreal Health Center, Cancer Prognostics and Health Outcomes Unit, Montreal, Canada. CR Santoni M, Scarpelli M, Mazzucchelli R, Lopez-Beltran A, Cheng L, Epstein JI, Cascinu S, Briganti A, Catto JW, Montorsi F, Montironi R, 2016, Current Histopathologic and molecular Characterisations of prostate Cancer: towards individualised prognosis and therapies. European Urology 69(2):186–190. https://doi. org/10.1016/j.eururo.2015.05.041 Wittschieber D, Kollermann J, Schlomm T, Sauter G, Erbersdobler A, 2010, Nuclear grading versus Gleason grading in small samples containing prostate cancer: a tissue microarray study. Pathology oncology research: POR 16(4):479–484. , DOI 10.1007/ s12253-010-9270-x Epstein JI, Zelefsky MJ, Sjoberg DD, Nelson JB, Egevad L, Magi- Galluzzi C, Vickers AJ, Parwani AV, Reuter VE, Fine SW, Eastham JA,Wiklund P, Han M, Reddy CA, Ciezki JP, Nyberg T, Klein EA, 2016, A contemporary prostate Cancer grading system: a validated alternative to the Gleason score. European Urology 69(3):428–435. , DOI 10.1016/j.eururo.2015.06.046 Loeb S, Folkvaljon Y, Robinson D, Lissbrant IF, Egevad L, Stattin P, 2015, Evaluation of the 2015 Gleason grade groups in a Nationwide population-based cohort. European Urology 69:1135– 1141. , DOI 10.1016/j.eururo.2015.11.036 Sauter G, Steurer S, Clauditz TS, Krech T, Wittmer C, Lutz F, Lennartz M, Janssen T, Hakimi N, Simon R, von Petersdorff- Campen M, Jacobsen F, von Loga K, Wilczak W, Minner S, Tsourlakis MC, Chirico V, Haese A, Heinzer H, Beyer B, Graefen M,Michl U, Salomon G, Steuber T, Budaus LH, Hekeler E, Malsy- Mink J, Kutzera S, Fraune C, Gobel C, Huland H, Schlomm T, 2016, Clinical utility of quantitative Gleason grading in prostate biopsies and prostatectomy specimens. European Urology 69(4): 592–598. , DOI 10.1016/j.eururo.2015.10.029 Timilshina N, Ouellet V, Alibhai SM, Mes-Masson AM, Delvoye N, Drachenberg D, Finelli A, Jammal MP, Karakiewicz P, Lapointe H, Lattouf JB, Lynch K, Paradis JB, Sitarik P, So A, Saad F, 2016, Analysis of active surveillance uptake for low-risk localized prostate cancer in Canada: a Canadian multi-institutional study.World Journal of Urology 35:595–603. , DOI 10.1007/s00345-016-1897-0 Meyer CP, Hansen J, Boehm K, Tilki D, Abdollah F, Trinh QD, Fisch M, Sauter G, Graefen M, Huland H, Chun FK, Ahyai SA, 2016, Tumor volume improves the long-term prediction of biochemical recurrence-free survival after radical prostatectomy for localized prostate cancer with positive surgical margins. World Journal of Urology 35:199–206. , DOI 10.1007/s00345- 016-1861-z Budaus L, Isbarn H, Eichelberg C, Lughezzani G, Sun M, Perrotte P, Chun FK, Salomon G, Steuber T, Kollermann J, Sauter G, Ahyai SA, Zacharias M, Fisch M, Schlomm T, Haese A, Heinzer H, Huland H, Montorsi F, Graefen M, Karakiewicz PI, 2010, Biochemical recurrence after radical prostatectomy: multiplicative interaction between surgical margin status and pathological stage. The Journal of Urology 184(4):1341–1346. , DOI 10. 1016/j.juro.2010.06.018 Isbarn H, Ahyai SA, Chun FK, Budaus L, Schlomm T, Salomon G, Zacharias M, Erbersdobler A, Kollermann J, Sauter G, Huland H, Graefen M, Steuber T, 2009, Prevalence of a tertiary Gleason grade and its impact on adverse histopathologic parameters in a contemporary radical prostatectomy series. European Urology 55(2):394– 401. , DOI 10.1016/j.eururo.2008.08.015 Suardi N, Porter CR, Reuther AM,Walz J, Kodama K, Gibbons RP, Correa R, Montorsi F, Graefen M, Huland H, Klein EA, Karakiewicz PI, 2008, Anomogram predicting long-termbiochemical recurrence after radical prostatectomy. Cancer 112(6):1254– 1263. , DOI 10.1002/cncr.23293 Briganti A, Karnes RJ, Joniau S, Boorjian SA, Cozzarini C, Gandaglia G, Hinkelbein W, Haustermans K, Tombal B, Shariat S, Sun M, Karakiewicz PI, Montorsi F, Van Poppel H, Wiegel T, 2014, Prediction of outcome following early salvage radiotherapy among patients with biochemical recurrence after radical prostatectomy. European Urology 66(3):479–486. , DOI 10.1016/j. eururo.2013.11.045 Lughezzani G, Briganti A, Karakiewicz PI, Kattan MW, Montorsi F, Shariat SF, Vickers AJ, 2010, Predictive and prognostic models in radical prostatectomy candidates: a critical analysis of the literature. European Urology 58(5):687–700. , DOI 10.1016/j. eururo.2010.07.034 Lughezzani G, Budaus L, Isbarn H, Sun M, Perrotte P, HaeseA, Chun FK, Schlomm T, Steuber T, Heinzer H, Huland H, Montorsi F, Graefen M, Karakiewicz PI, 2010, Head-to-head comparison of the three most commonly used preoperative models for prediction of biochemical recurrence after radical prostatectomy. European Urology 57(4):562–568. , DOI 10.1016/j.eururo.2009.12.003 Walz J, Chun FK, Klein EA, Reuther A, Saad F, Graefen M,Huland H, Karakiewicz PI, 2009, Nomogram predicting the probability of early recurrence after radical prostatectomy for prostate cancer. The Journal of Urology 181(2):601–607; discussion 607-608. https:// doi.org/10.1016/j.juro.2008.10.033 Epstein JI, Egevad L, Amin MB, Delahunt B, Srigley JR, Humphrey PA, Grading C, 2016, The 2014 International Society of Urological Pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma: definition of grading patterns and proposal for a new grading system. The American Journal of Surgical Pathology 40(2):244–252. , DOI 10.1097/PAS. 0000000000000530 Bubendorf L, Nocito A, Moch H, Sauter G, 2001, Tissue microarray, TMA, technology: miniaturized pathology archives for highthroughput in situ studies. The Journal of Pathology 195(1):72–79. , DOI 10.1002/path.893 Rubin MA, Dunn R, Strawderman M, Pienta KJ, 2002, Tissue microarray sampling strategy for prostate Cancer biomarker analysis. The American Journal of Surgical Pathology 26(3):312– 319. , DOI 10.1097/00000478-200203000-00004 Diallo JS, Aldejmah A,Mouhim AF, Fahmy MA, Koumakpayi IH, Sircar K, Begin LR, Mes-Masson AM, Saad F, 2008, Coassessment of cytoplasmic and nuclear androgen receptor location in prostate specimens: potential implications for prostate cancer development and prognosis. BJU International 101(10):1302– 1309. , DOI 10.1111/j.1464-410X.2008.07514.x Gannon PO, Lessard L, Stevens LM, Forest V, Begin LR, Minner S, Tennstedt P, Schlomm T, Mes-Masson AM, Saad F, 2013, Largescale independent validation of the nuclear factor-kappa B p65 prognostic biomarker in prostate cancer. European Journal of Cancer 49(10):2441–2448. , DOI 10.1016/j.ejca.2013.02.026 Koumakpayi IH, Le Page C, Mes-Masson AM, Saad F, 2010, Hierarchical clustering of immunohistochemical analysis of the activated ErbB/PI3K/Akt/NF-kappaB signalling pathway and prognostic significance in prostate cancer. British Journal of Cancer 102(7):1163–1173. , DOI 10.1038/sj.bjc.6605571 Ross AE, Johnson MH, Yousefi K, Davicioni E, Netto GJ, Marchionni L, Fedor HL, Glavaris S, Choeurng V, Buerki C, Erho N, Lam LL, Humphreys EB, Faraj S, Bezerra SM, Han M, Partin AW, Trock BJ, Schaeffer EM, 2016, Tissue-based genomics augments post-prostatectomy risk stratification in a natural history cohort of intermediate- and high-risk men. European Urology 69(1):157–165. , DOI 10.1016/j.eururo.2015.05.042 Ruela-de-Sousa RR, Hoekstra E, Hoogland AM, Souza Queiroz KC, Peppelenbosch MP, Stubbs AP, Pelizzaro-Rocha K, van Leenders GJ, Jenster G, Aoyama H, Ferreira CV, Fuhler GM, 2016, Low-molecular-weight protein tyrosine phosphatase predicts prostate Cancer outcome by increasing the metastatic potential. European Urology 69(4):710–719. , DOI 10.1016/j. eururo.2015.06.040 Belledant A, Hovington H, Garcia L, Caron P, Brisson H, Villeneuve L, Simonyan D, Tetu B, Fradet Y, Lacombe L, Guillemette C, Levesque E, 2016, The UGT2B28 sex-steroid inactivation pathway is a regulator of Steroidogenesis and modifies the risk of prostate Cancer progression. European Urology 69(4): 601–609. , DOI 10.1016/j.eururo.2015.06.054 Bostrom PJ, Bjartell AS, Catto JW, Eggener SE, Lilja H, Loeb S, Schalken J, Schlomm T, Cooperberg MR, 2015, Genomic predictors of outcome in prostate Cancer. European Urology 68:1033– 1044. , DOI 10.1016/j.eururo.2015.04.008 Cullen J, Rosner IL, Brand TC, Zhang N, Tsiatis AC, Moncur J, Ali A, Chen Y, Knezevic D, Maddala T, Lawrence HJ, Febbo PG, Srivastava S, Sesterhenn IA, McLeod DG, 2015, A biopsy-based 17-gene genomic prostate score predicts recurrence after radical prostatectomy and adverse surgical pathology in a racially diverse population of men with clinically low- and intermediate-risk prostate Cancer. European Urology 68(1):123–131. , DOI 10. 1016/j.eururo.2014.11.030 Eggener S, 2015, Prostate Cancer screening biomarkers: an emerging embarrassment of 'Riches'? European Urology 70:54–55. , DOI 10.1016/j.eururo.2015.09.002 Schlomm T, Weischenfeldt J, Korbel J, Sauter G, 2015, The aging prostate is never "normal": implications from the genomic characterization of multifocal prostate cancers. European Urology 68: 348–350. , DOI 10.1016/j.eururo.2015.04.012 Klein EA, Yousefi K, Haddad Z, Choeurng V, Buerki C, Stephenson AJ, Li J, Kattan MW, Magi-Galluzzi C, Davicioni E, 2015, A genomic classifier improves prediction of metastatic disease within 5 years after surgery in node-negative high-risk prostate cancer patients managed by radical prostatectomy without adjuvant therapy. European Urology 67(4):778–786. , DOI 10. 1016/j.eururo.2014.10.036 Tomlins SA, Day JR, Lonigro RJ, Hovelson DH, Siddiqui J, Kunju LP, Dunn RL, Meyer S, Hodge P, Groskopf J, Wei JT, Chinnaiyan AM, 2015, Urine TMPRSS2:ERG plus PCA3 for individualized prostate Cancer risk assessment. European Urology 70:45–53. , DOI 10.1016/j.eururo.2015.04.039 Mehra R, Udager AM, Ahearn TU, Cao X, Feng FY, Loda M, Petimar JS, Kantoff P, Mucci LA, Chinnaiyan AM, 2015, Overexpression of the long non-coding RNA SChLAP1 independently predicts lethal prostate Cancer. European Urology 70:549– 552. , DOI 10.1016/j.eururo.2015.12.003 Rubin MA, Girelli G, Demichelis F, 2016, Genomic correlates to the newly proposed grading prognostic groups for prostate Cancer. European Urology 69(4):557–560. , DOI 10.1016/j.eururo. 2015.10.040 Chun FK, Briganti A, GraefenM,Montorsi F, Porter C, Scattoni V, GallinaA,Walz J, Haese A, Steuber T, Erbersdobler A, SchlommT, Ahyai SA, Currlin E, Valiquette L, Heinzer H, Rigatti P, Huland H, Karakiewicz PI, 2007, Development and external validation of an extended 10-core biopsy nomogram. EuropeanUrology 52(2):436– 444. , DOI 10.1016/j.eururo.2006.08.039 SiddiquiMM, Rais-Bahrami S, Turkbey B, George AK, Rothwax J, Shakir N, Okoro C, Raskolnikov D, Parnes HL, Linehan WM, 2015, Comparison of MR/ultrasound fusion–guided biopsy with ultrasound-guided biopsy for the diagnosis of prostate cancer. JAMA 313(4):390–397 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 979 EP 986 DI 10.1007/s12253-018-0408-6 PG 8 ER PT J AU Yonekura, S Terauchi, F Hoshi, K Yamaguchi, T Kawai, Sh AF Yonekura, Satoru Terauchi, Fumihito Hoshi, Kenji Yamaguchi, Takehiko Kawai, Shigeo TI Androgen Receptor Predicts First and Multiple Recurrences in Non-Muscle Invasive Urothelial Carcinoma of the Bladder SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Androgen receptor; Recurrence; Bladder cancer; Immunohistochemistry ID Androgen receptor; Recurrence; Bladder cancer; Immunohistochemistry AB The aim of this study is to investigate the role of androgen receptor (AR) expression on clinicopathologic characteristics, first recurrence free survival (RFS), progression free survival (PFS) and multiple recurrences in non-muscle invasive bladder cancer (NMIBC). AR expression in 40 paraffin-embedded specimens of primarily diagnosed NMIBC after transurethral resection was examined by immunohistochemistry using a monoclonal AR antibody. Associations between AR expression and clinicopathologic features and prognosis were statistically assessed. Multivariate Cox proportional hazards model was applied for evaluating predictive factors on RFS and PFS. For multiple recurrences, we used the Andersen-Gill model. AR was positive in 20/40 (50%) cases. Twenty-three patients (57.5%) had no recurrence, 10 (25.0%) had one recurrence, and 7 (17.5%) experiencedmore than one recurrence. AR expression and clinicopathologic features were not significantly correlated (P >0.05). Univariate analyses showed that AR expression was significantly associated with RFS and PFS (P <0.05). Via multivariate analyses, positive AR expression was significantly associated with lower risk of first recurrence (hazard ratio (HR) = 0.265; 95% confidence interval (95% CI) = 0.084–0.829; P = 0.022).Multivariate analysis of PFS was not feasible in our cohort.Using the multivariate Andersen-Gillmodel, positive AR expression in the primary tumor was an independent factor predicting lower risk ofmultiple recurrences (HR = 0.387, 95% CI = 0.161–0.927, P = 0.033). Androgen receptor expression is associated with first and multiple recurrences in NMIBC. C1 [Yonekura, Satoru] Tochigi Medical Center Shimotsuga Hospital, Department of Pathology, 420-1 Ohira-machi Kawatsure, 329-4498 Tochigi, Tochigi, Japan. [Terauchi, Fumihito] Tochigi Medical Center Shimotsuga Hospital, Department of Urology, 420-1 Ohira-machi Kawatsure, 329-4498 Tochigi, Tochigi, Japan. [Hoshi, Kenji] Tochigi Medical Center Shimotsuga Hospital, Department of Pathology, 420-1 Ohira-machi Kawatsure, 329-4498 Tochigi, Tochigi, Japan. [Yamaguchi, Takehiko] Dokkyo Medical University, Koshigaya Hospital, Department of Pathology, 2-1-50 Minami-Koshigaya, 343-8555 Koshigaya, Saitama, Japan. [Kawai, Shigeo] Tochigi Medical Center Shimotsuga Hospital, Department of Pathology, 420-1 Ohira-machi Kawatsure, 329-4498 Tochigi, Tochigi, Japan. RP Yonekura, S (reprint author), Tochigi Medical Center Shimotsuga Hospital, Department of Pathology, 329-4498 Tochigi, Japan. EM yonechian@gmail.com CR Sylvester RJ et al, 2006, Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials. Eur Urol 49(3):466–477 Scosyrev E et al, 2009, Sex and racial differences in bladder cancer presentation and mortality in the US. Cancer 115(1):68– 74 Zaitsu M et al, 2015, Sex differences in bladder cancer pathology and survival: analysis of a population-based cancer registry. Cancer Med 4(3):363–370 Hartge P et al, 1990, Unexplained excess risk of bladder cancer in men. J Natl Cancer Inst 82(20):1636–1640 Rosenzweig BA et al, 1995, Location and concentration of estrogen, progesterone, and androgen receptors in the bladder and urethra of the rabbit. Neurourol Urodyn 14(1):87–96 Jing Y et al, 2014, Activated androgen receptor promotes bladder cancer metastasis via Slug mediated epithelial-mesenchymal transition. Cancer Lett 348(1–2):135–145 Miyamoto H et al, 2007, Promotion of bladder cancer development and progression by androgen receptor signals. J Natl Cancer Inst 99(7):558–568 Hsu JW et al, 2013, Decreased tumorigenesis and mortality from bladder cancer in mice lacking urothelial androgen receptor. Am J Pathol 182(5):1811–1820 Wu JT et al, 2010, Androgen receptor is a potential therapeutic target for bladder cancer. Urology 75(4):820–827 Lombard AP, Mudryj M(2015, The emerging role of the androgen receptor in bladder cancer. Endocr Relat Cancer 22(5):R265–R277 Boorjian S et al, 2004, Androgen receptor expression is inversely correlated with pathologic tumor stage in bladder cancer. Urology 64(2):383–388 Kauffman EC et al, 2011, Role of androgen receptor and associated lysine-demethylase coregulators, LSD1 and JMJD2A, in localized and advanced human bladder cancer. Mol Carcinog 50(12):931– 944 Mashhadi R et al, 2014, Role of steroid hormone receptors in formation and progression of bladder carcinoma: a case-control study. Urol J 11(6):1968–1973 Mir C et al, 2011, Loss of androgen receptor expression is not associated with pathological stage, grade, gender or outcome in bladder cancer: a large multi-institutional study. BJU Int 108(1): 24–30 Miyamoto H et al, 2012, Expression of androgen and oestrogen receptors and its prognostic significance in urothelial neoplasm of the urinary bladder. BJU Int 109(11):1716–1726 Tuygun C et al, 2011, Sex-specific hormone receptors in urothelial carcinomas of the human urinary bladder: a comparative analysis of clinicopathological features and survival outcomes according to receptor expression. Urol Oncol 29(1):43–51 Nam JK et al, 2014, Prognostic value of sex-hormone receptor expression in non-muscle-invasive bladder cancer. Yonsei Med J 55(5):1214–1221 Takashi M et al, 2002, Evaluation of multiple recurrence events in superficial bladder cancer patients treated with intravesical bacillus Calmette-Guerin therapy using the Andersen-Gill’s model. Int Urol Nephrol 34(3):329–334 Yan Y et al, 2002, Patterns of multiple recurrences of superficial, Ta/T1, transitional cell carcinoma of bladder and effects of clinicopathologic and biochemical factors. Cancer 95(6):1239–1246 Izumi K et al, 2014, Androgen deprivation therapy prevents bladder cancer recurrence. Oncotarget 5(24):12665–12674 FreedmanNDet al, 2011, Association between smoking and risk of bladder cancer among men and women. JAMA 306(7):737–745 Montironi R, Lopez-Beltran A, 2005, The 2004 WHO classification of bladder tumors: a summary and commentary. Int J Surg Pathol 13(2):143–153 Andersen PK, Gill RD, 1982, Cox’s regression model for counting processes: a large sample study. Ann Stat 10(4):1100–1120 Ide H, Inoue S, Miyamoto H, 2017, Histopathological and prognostic significance of the expression of sex hormone receptors in bladder cancer: a meta-analysis of immunohistochemical studies. PLoS One 12(3):e0174746 Li Y, Izumi K, Miyamoto H, 2012, The role of the androgen receptor in the development and progression of bladder cancer. Jpn J Clin Oncol 42(7):569–577 Lin Z et al, 2003, The expression of bcl-2 and bcl-6 protein in normal and malignant transitional epithelium. Urol Res 31(4): 272–275 Amirghofran Z, Monabati A, Gholijani N, 2004, Androgen receptor expression in relation to apoptosis and the expression of cell cycle related proteins in prostate cancer. Pathol Oncol Res 10(1): 37–41 Shiota M et al, 2015, Secondary bladder cancer after anticancer therapy for prostate cancer: reduced comorbidity after androgendeprivation therapy. Oncotarget 6(16):14710–14719 Morales EE et al, 2016, Finasteride reduces risk of bladder cancer in a large prospective screening study. Eur Urol 69(3):407–410 Izumi K et al, 2016, Expression of androgen receptor in nonmuscle- invasive bladder cancer predicts the preventive effect of androgen deprivation therapy on tumor recurrence. Oncotarget 7(12):14153-14160 Haapala K et al, 2001, Androgen receptor alterations in prostate cancer relapsed during a combined androgen blockade by orchiectomy and bicalutamide. Lab Investig 81(12):1647–1651 Thompson J et al, 2003, Androgen receptor mutations in highgrade prostate cancer before hormonal therapy. Lab Investig 83(12):1709–1713 Gonzalez-Zulueta M et al, 1993, Microsatellite instability in bladder cancer. Cancer Res 53(23):5620–5623 Teng XY et al, 2010, CAG repeats in the androgen receptor gene are shorter in patients with pulmonary, esophageal or bladder carcinoma and longer in women with uterine leiomyoma. Oncol Rep 23(3):811–818 Liu CH et al, 2008, Androgen receptor gene polymorphism may affect the risk of urothelial carcinoma. J Biomed Sci 15(2):261–269 Wu S et al, 2013, Somatic mutation of the androgen receptor gene is not associated with transitional cell carcinoma: a Bnegative^ study by whole-exome sequencing analysis. Eur Urol 64(6): 1018–1019 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 987 EP 994 DI 10.1007/s12253-018-0431-7 PG 8 ER PT J AU Koszo, R Varga, L Fodor, E Kahan, Zs Cserhati, A Hideghety, K Egyud, Zs Szabo, Cs Borzasi, E Szabo, D Mullner, K Varga, Z Maraz, A AF Koszo, Renata Varga, Linda Fodor, Emese Kahan, Zsuzsanna Cserhati, Adrienne Hideghety, Katalin Egyud, Zsofia Szabo, Csilla Borzasi, Emoke Szabo, Dorottya Mullner, Kitti Varga, Zoltan Maraz, Aniko TI Prone Positioning on a Belly Board Decreases Rectal and Bowel Doses in Pelvic Intensity-Modulated Radiation Therapy (IMRT) for Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; IMRT; Prone; Belly board; Small bowel; Rectum ID Prostate cancer; IMRT; Prone; Belly board; Small bowel; Rectum AB The presence of normal tissues in the irradiated volume limits dose escalation during pelvic radiotherapy (RT) for prostate cancer. Supine and prone positions on a belly board were compared by analyzing the exposure of organs at risk (OARs) using intensity modulated RT (IMRT). The prospective trial included 55 high risk, localized or locally advanced prostate cancer patients, receiving definitive image-guided RT. Computed tomography scanning for irradiation planning was carried out in both positions. Gross tumor volume, clinical and planning target volumes (PTV) and OARs were delineated, defining subprostatic and periprostatic rectal subsegments. At the height of the largest antero-posterior (AP) diameter of the prostate, rectal diameters and distance from the posterior prostate wall were measured. IMRT plans were generated. Normal tissue exposure and structure volumes were compared between supine and prone plans using paired t-test. In the volumes of the prostate, PTV, colon and small bowel, no significant differences were found. In prone position, all rectal volumes, diameters, and rectum–prostate distance were significantly higher, the irradiated colon and small bowel volume was lower in dose ranges of 20–40 Gy, and the exposure to all rectal segments was more favorable in 40–75 Gy dose ranges. No significant difference was found in the exposure of other OARs. Prone positioning on a belly board is an appropriate positioning method aiming rectum and bowel protection during pelvic IMRT of prostate cancer. The relative reduction in rectal exposure might be a consequence of the slight departure between the prostate and rectal wall. C1 [Koszo, Renata] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Varga, Linda] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Fodor, Emese] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Cserhati, Adrienne] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Hideghety, Katalin] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Egyud, Zsofia] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Szabo, Csilla] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Borzasi, Emoke] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Szabo, Dorottya] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Mullner, Kitti] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Varga, Zoltan] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. [Maraz, Aniko] University of Szeged, Department of Oncotherapy, Koranyi Alley 12, H-6720 Szeged, Hungary. RP Koszo, R (reprint author), University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. EM koszorenata@gmail.com CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: Globocan 2008. Int J Cancer 127:2893–2917 Juloori A, Shah C, Stephans K, Vassil A, Tendulkar R, 2016, Evolving paradigm of radiotherapy for high-risk prostate cancer: current consensus and continuing controversies. Prostate Cancer 2016:2420786 Lukka H,Warde P, Pickles T, Morton G, Brundage M, Souhami L, Canadian GU Radiation Oncologist Group, 2001, Controversies in prostate cancer radiotherapy: consensus development. Canadian GU Radiation Oncologist Group. Can J Urol 8:1314–1322 Andreyev HJ, 2007, Gastrointestinal problems after pelvic radiotherapy: the past, the present and the future. Clin Oncol, R Coll Radiol, 19:790–799 Stacey R, Green JT, 2014, Radiation-induced small bowel disease: latest developments and clinical guidance. Ther Adv Chronic Dis 5: 15–29 Theis V, Sripadam R, Ramani V, Lal S, 2010, Chronic radiation enteritis. Clin Oncol 22:70–83 Kennedy G, Heise C, 2005, Radiation colitis and proctitis. Clin Colon Rectal Surg 20:64–72 Olopade F, Norman A, Blake P, Dearnaley DP, Harrington KJ, Khoo Vet al, 2005, A modified inflammatory bowel disease questionnaire and the Vaizey incontinence questionnaire are simple ways to identify patients with significant gastrointestinal symptoms after pelvic radiotherapy. Br J Cancer 92:1663–1670 Wiesendanger-Wittmer E, Sijtsema N, Muijs C, Beukema JC, 2012, Systematic review of the role of a belly board device in radiotherapy delivery in patients with pelvic malignancies. Radiother Oncol 102:325–334 Zelefsky MJ, Cowen D, Fuks Z, ShikeM, Burman C, Jackson A et al, 1999, Long term tolerance of high dose three-dimensional conformal radiotherapy in patients with localized prostate carcinoma. Cancer 85:2460–2468 Sobin LH, Gospodarowicz MK, Wittekind C, 2009, TNM classification of malignant tumors, 7th edn. Wiley-Blackwell, London Mellinger GT, Gleason D, Bailar J 3rd, 1967, The histology and prognosis of prostatic cancer. J Urol 97:331–337 Lawton CAF, Michalski J, El-Naga I, BuyyounouskiMK, LeeWR, Menard C et al, 2009, RTOG GU radiation oncology specialists reach consensus on pelvic lymph node volumes for high-risk prostate cancer. Int J Radiat Oncol Biol Phys 74:383–387 Viswanathan AN, Yorke ED, Marks LB, Eifel PJ, Shipley WU, 2010, Radiation dose-volume effects of the urinary bladder. Int J Radiat Oncol Biol Phys 76(Suppl 3):S116–S122 Gami B, Harrington K, Blake P, Dearnaley D, Tait D, Davies J, Norman AR, Andreyev HJN, 2003, How patients manage gastrointestinal symptoms after pelvic radiotherapy. Aliment Pharmacol Therapeut 18:987–994 Andreyev H, 2007, Gastrointestinal symptoms after pelvic radiotherapy: a new understanding to improvemanagement of symptomatic patients. Lancet Oncol 8:1007–1017 Letschert JGJ, 1995, The prevention of radiation-induced small bowel complications. Eur J Cancer 31:1361–1365 Michalski JM, Gay H, Jackson A, Tucker SL, Deasy JO, 2010, Radiation dose-volume effects in radiation-induced rectal injury. Int J Radiat Oncol Biol Phys 76(Suppl 3):S123–S129 Baglan KL, Frazier RC, Yan D, Huang RR, Martinez AA, Robertson JM, 2002, The dose–volume relationship of acute small bowel toxicity from concurrent 5-FU-based chemotherapy and radiation therapy for rectal cancer. Int J Radiat Oncol Biol Phys 52: 176–183 Roeske JC, Bonta D, Mell LK, Lujan AE, Mundt AJ, 2003, A dosimetric analysis of acute gastrointestinal toxicity in women receiving intensity-modulated whole-pelvic radiation therapy. Radiother Oncol 69:201–207 Fiorino C, Valdagni R, Rancati T, Sanguineti G, 2009, Dosevolume effects for normal tissues in external radiotherapy: pelvis. Radiother Oncol 93:153–167 Peeters ST, Hoogeman MS, Heemsbergen WD, Hart AA, Koper PC, Lebesque JV, 2006, Rectal bleeding, fecal incontinence, and high stool frequency after conformal radiotherapy for prostate cancer: normal tissue complication probability modeling. Int J Radiat Oncol Biol Phys 66:11–19 SchaakeW, van der Schaaf A, van Dijk LV, Bongaerts AH, van den Bergh AC, Langendijk JA, 2016, Normal tissue complication probability, NTCP, models for late rectal bleeding, stool frequency and fecal incontinence after radiotherapy in prostate cancer patients. Radiother Oncol 119:381–387 Drean G, Acosta O, Ospina JD, Fargeas A, Lafond C, Correge G et al, 2016, Identification of a rectal subregion highly predictive of rectal bleeding in prostate cancer IMRT. Radiother Oncol 119:388– 397 van Lin EN, Hoffmann AL, van Kollenburg P, Leer JW, Visser AG, 2005, Rectal wall sparing effect of three different endorectal balloons in 3D conformal and IMRT prostate radiotherapy. Int J Radiat Oncol Biol Phys 63:565–576 Serrano N, Kalman NS, Anscher MS, 2017, Reducing rectal injury in men receiving prostate cancer radiation therapy: current perspectives. Cancer Manag Res 9:339–350 Zelefsky MJ, Happersett L, Leibel SA, Burman CM, Schwartz L, Dicker AP et al, 1997, The effect of treatment positioning on normal tissue dose in patients with prostate cancer treated with threedimensional conformal radiotherapy. Int J Radiat Oncol Biol Phys 37:13–19 McLaughlin PW, Wygoda A, Sahijdak W, Sandler HM, Marsh L, Roberson P et al, 1999, The effect of patient position and treatment technique in conformal treatment of prostate cancer. Int J Radiat Oncol Biol Phys 45:407–413 O’Neil L, Armstrong J, Buckney S, Assiri M, Cannon M, Holmberg O, 2008, A phase II trial for the optimisation of treatment position in the radiation therapy of prostate cancer. Radiother Oncol 88:61–66 Bajon T, Piotrowski T, Antczak A, Bak B,Blasiak B, Kazmierskaa J, 2011, Comparison of dose-volume histograms for supine and prone position in patients irradiated for prostate cancer – a preliminary study. Rep Pract Oncol Radiother 16:65–70 Bayley AJ, Catton CN, Haycocks T, Kelly V, Alasti H, Bristow R et al, 2003, A randomized trial of supine vs. prone positioning in patients undergoing escalated dose conformal radiotherapy for prostate cancer. Radiother Oncol 70:37–44 Kato T, Obata Y, Kadoya N, Fuwa N, 2009, A comparison of prone three-dimensional conformal radiotherapy with supine intensitymodulated radiotherapy for prostate cancer: which technique is more effective for rectal sparing? Br J Radiol 82:654–661 DrzymalaM, HawkinsMA, Henrys AJ, Bedford J, Norman A, Tait DM, 2009, The effect of treatment position, prone or supine, on dose–volume histograms for pelvic radiotherapy in patients with rectal cancer. Br J Radiol 82:321–327 Kim TH, Chie EK, Kim DY, Park SY, Cho KH, Jung KH et al, 2005, Comparison of the belly board device method and the distended bladder method for reducing irradiated small bowel volumes in preoperative radiotherapy of rectal cancer patients. Int J Radiat Oncol Biol Phys 62:769–775 Huh SJ, Park W, Ju SG, Lee JE, Han Y, 2004, Small-bowel displacement system for the sparing of small bowel in threedimensional conformal radiotherapy for cervical cancer. Clin Oncol 16:467–473 Martin J, Fitzpatrick K, Horan G,McCloy R, Buckney S, O'Neill L et al, 2005, Treatment with a belly-board device significantly reduces the volume of small bowel irradiated and results in low acute toxicity in adjuvant radiotherapy for gynecologic cancer: results of a prospective study. Radiother Oncol 74:267–274 Pinkawa M, Gagel B, Demirel C, Schmachtenberg A, Asadpour B, Eble MJ, 2003, Dose–volume histogram evaluation of prone and supine patient position in external beam radiotherapy for cervical and endometrial cancer. Radiother Oncol 69:99–105 Mundt A, Roeske J, Lujan A, 2002, Intensity-modulated radiation therapy in gynecologic malignancies. Med Dosimetry 27:131–136 Portelance L, Chao K, Grigsby P, Bennet H, Low D, 2001, Intensity-modulated radiation therapy, IMRT, reduces small bowel, rectum, and bladder doses in patients with cervical cancer receiving pelvic and Para-aortic irradiation. Int J Radiat Oncol Biol Phys 51:261–266 Stromberger C, Kom Y, Kawgan-Kagan M, Mensing T, Jahn U, Schneider A et al, 2010, Intensity-modulated radiotherapy in patients with cervical cancer. An intra-individual comparison of prone and supine positioning. Radiat Oncol 5:63–68 Beriwal S, Jain SK, Heron DE, de Andrade RS, Lin CJ, Kim H, 2007, Dosimetric and toxicity comparison between prone and supine position IMRT for endometrial cancer. Int J Radiat Oncol Biol Phys 67:485–489 Kim TH, Kim DY, Cho KH, Kim YH, Jung KH, Ahn JB et al, 2005, Comparative analysis of the effects of belly board and bladder distension in postoperative radiotherapy of rectal cancer patients. Strahlenther Onkol 181(9):601–605 Saynak M, Kucucuk S, Aslay I, 2008, Abdominal pillow for the sparing of small bowel in four-field conventional pelvic radiotherapy. Eur J Gynaecol Oncol 29:643–648 Fu YT, Lam JC, Tze JM, 1995, Measurement of irradiated small bowel volume in pelvic irradiation and the effect of a belly board. Clin Oncol, R Coll Radiol, 7:188–192 Robertson JM, Lockman D, Yan D, Wallace M, 2008, The dose-volume relationship of small bowel irradiation and acute grade 3 diarrhea during chemoradiotherapy for rectal cancer. Int J Radiat Oncol Biol Phys 70:413–418 Gunnlaugsson A, Kjellen E, Nilsson P, Bendahl PO, Willner J, Johnsson A, 2007, Dose–volume relationships between enteritis and irradiated bowel volumes during 5-fluorouracil and oxaliplatin based chemoradiotherapy in locally advanced rectal cancer. Acta Oncol 46:937–944 Gill SK, Reddy K, Campbell N, Chen C, Pearson D, 2015, Determination of optimal PTV margin for patients receiving CBCT-guided prostate IMRT: comparative analysis based on CBCT dose calculation with four different margins. J Appl Clin Med Phys 16:252–262 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 995 EP 1002 DI 10.1007/s12253-018-0436-2 PG 8 ER PT J AU Chen, YT Tsai, HP Wu, ChCh Chen, ChY Chai, ChY Kwan, AL AF Chen, Yi-Ting Tsai, Hung-Pei Wu, Chun-Chieh Chen, Chiao-Yun Chai, Chee-Yin Kwan, Aij-Lie TI High-level Sp1 is Associated with Proliferation, Invasion, and Poor Prognosis in Astrocytoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Astrocytoma; Specificity protein 1; WHO grade; Immunohistochemistry staining; Apoptosis ID Astrocytoma; Specificity protein 1; WHO grade; Immunohistochemistry staining; Apoptosis AB Astrocytoma is the most common and the most lethal primary brain tumor in adults. Grade IV glioblastoma is usually refractory to currently available surgical, chemotherapeutic, and radiotherapeutic treatments. The Specificity protein 1 (Sp1) transcription factor is known to regulate tumorigenesis in many cancers. The aim of this study was to investigate the clinicopathologic role of Sp1 protein in the carcinogenesis of astrocytoma. This study analyzed 98 astrocytoma cases treated at Kaohsiung Medical University Hospital during 2002–2012. Clinicopathologic parameters associated with Sp1 were analyzed by chi-square test, Kaplan-Meier analysis, and Cox regression analyses. In vitro proliferation, invasion, and migration were compared between nonsiRNA groups and Sp1 siRNA groups. In glioblastoma cells treated with Sp1 siRNA, Western blot was also used to detect expressions of Sp1, Ki-67, VEGF, cyclin D1, E-cadherin, cleaved caspase-3 and Bax proteins. Expression of Sp1 was significantly associated with WHO grade (p = 0.005) and with overall survival time (p < 0.001). Multivariate analysis further revealed that prognosis of astrocytoma was significantly associated with Sp1 expression (p = 0.036) and IDH-1 expression (p < 0.001). In vitro silencing of Sp1 downregulated Sp1, Ki-67, and cyclin D1 but upregulated E-cadherin, Bax, and cleaved caspase-3. These data suggest that Sp1 is a potential prognostic marker and therapeutic target in astrocytoma. C1 [Chen, Yi-Ting] Kaohsiung Medical University, College of Medicine, Department of PathologyKaohsiung, Taiwan, Republic of China. [Tsai, Hung-Pei] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Department of NeurosurgeryKaohsiung, Taiwan, Republic of China. [Wu, Chun-Chieh] Kaohsiung Medical University, College of Medicine, Department of PathologyKaohsiung, Taiwan, Republic of China. [Chen, Chiao-Yun] Kaohsiung Medical University, College of Medicine, Faculty of Medicine, Department of RadiologyKaohsiung, Taiwan, Republic of China. [Chai, Chee-Yin] Kaohsiung Medical University, College of Medicine, Department of PathologyKaohsiung, Taiwan, Republic of China. [Kwan, Aij-Lie] Kaohsiung Medical University, College of Medicine, Department of PathologyKaohsiung, Taiwan, Republic of China. RP Chai, ChY (reprint author), Kaohsiung Medical University, College of Medicine, Department of Pathology, Kaohsiung, Taiwan, Republic of China. EM cychai@kmu.edu.tw CR Louis DN, HO ODW, CaveneeWK, 2016, WHO Classification of Tumors of the Central Nervous System, 4th edn. WHO, Geneva Coin F, Marinoni JC, Rodolfo C, Fribourg S, Pedrini AM, Egly JM, 1998, Mutations in the XPD helicase gene result in XP and TTD phenotypes, preventing interaction between XPD and the p44 subunit of TFIIH. Nat Genet 20(2):184–188 Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL, Burkhard C, Schuler D, Probst-Hensch NM, Maiorka PC, Baeza N, Pisani P, Yonekawa Y, Yasargil MG, Lutolf UM, Kleihues P, 2004, Genetic pathways to glioblastoma: a population-based study. Cancer Res 64(19):6892–6899 Dynan WS, Tjian R, 1983, The promoter-specific transcription factor Sp1 binds to upstream sequences in the SV40 early promoter. Cell 35(1):79–87 Kadonaga JT, Courey AJ, Ladika J, Tjian R, 1988, Distinct regions of Sp1 modulate DNA binding and transcriptional activation. Science 242(4885):1566–1570 Karlseder J, Rotheneder H, Wintersberger E, 1996, Interaction of Sp1 with the growth- and cell cycle-regulated transcription factor E2F. Mol Cell Biol 16(4):1659–1667 Safe S, 2015, MicroRNA-Specificity Protein, Sp, Transcription Factor Interactions and Significance in Carcinogenesis. Curr Pharmacol Rep 1(2):73–78 Safe S, Abdelrahim M, 2005, Sp transcription factor family and its role in cancer. Eur J Cancer 41(16):2438–2448 Chiefari E, Brunetti A, Arturi F, Bidart JM, Russo D, Schlumberger M, Filetti S, 2002, Increased expression of AP2 and Sp1 transcription factors in human thyroid tumors: a role in NIS expression regulation? BMC Cancer 2:35 Kumar AP, Butler AP, 1999, Enhanced Sp1 DNA-binding activity in murine keratinocyte cell lines and epidermal tumors. Cancer Lett 137(2):159–165 Shi Q, Le X, Abbruzzese JL, Peng Z, Qian CN, Tang H, Xiong Q, Wang B, Li XC, Xie K, 2001, Constitutive Sp1 activity is essential for differential constitutive expression of vascular endothelial growth factor in human pancreatic adenocarcinoma. Cancer Res 61(10):4143–4154 Wang J,KangM, Qin YT,Wei ZX, Xiao JJ,Wang RS, 2015, Sp1 is over-expressed in nasopharyngeal cancer and is a poor prognostic indicator for patients receiving radiotherapy. Int J Clin Exp Pathol 8(6):6936–6943 Yao JC, Wang L, WeiD,GongW,HassanM,Wu TT, Mansfield P, Ajani J, Xie K, 2004, Association between expression of transcription factor Sp1 and increased vascular endothelial growth factor expression, advanced stage, and poor survival in patients with resected gastric cancer. Clin Cancer Res 10(12 Pt 1):4109–4117 Zannetti A, DelVecchio S, CarrieroMV, Fonti R, Franco P, Botti G, D'Aiuto G, Stoppelli MP, Salvatore M, 2000, Coordinate upregulation of Sp1 DNA-binding activity and urokinase receptor expression in breast carcinoma. Cancer Res 60(6):1546–1551 Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P, 2007, The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114(2):97–109 Guan H, Cai J, Zhang N, Wu J, Yuan J, Li J, Li M, 2012, Sp1 is upregulated in human glioma, promotes MMP-2-mediated cell invasion and predicts poor clinical outcome. Int J Cancer 130(3):593–601 Combs SE, Rieken S, Wick W, Abdollahi A, von Deimling A, Debus J, Hartmann C, 2011, Prognostic significance of IDH-1 and MGMT in patients with glioblastoma: one step forward, and one step back? Radiat Oncol 6:115 Cohen AL, Holmen SL, Colman H, 2013, IDH1 and IDH2 mutations in gliomas. Curr Neurol Neurosci Rep 13(5):345 Sipayya V, Sharma I , Sharma KC, Singh A, 2012, Immunohistochemical expression of IDH1 in gliomas: a tissue microarray-based approach. J Cancer Res Ther 8(4):598–601 Ogura R, Tsukamoto Y, Natsumeda M, Isogawa M, Aoki H, Kobayashi T, Yoshida S, Okamoto K, Takahashi H, Fujii Y, Kakita A, 2015, Immunohistochemical profiles of IDH1, MGMT and P53: practical significance for prognostication of patients with diffuse gliomas. Neuropathology 35(4):324–335 Baur F, Nau K, Sadic D, Allweiss L, Elsasser HP, Gillemans N, de Wit T, Kruger I, Vollmer M, Philipsen S, Suske G, 2010, Specificity protein 2, Sp2, is essential for mouse development and autonomous proliferation of mouse embryonic fibroblasts. PLoS One 5(3):e9587 Marin M, Karis A, Visser P, Grosveld F, Philipsen S, 1997, Transcription factor Sp1 is essential for early embryonic development but dispensable for cell growth and differentiation. Cell 89(4): 619–628 Nguyen-Tran VT, Kubalak SW, Minamisawa S, Fiset C, Wollert KC, Brown AB, Ruiz-Lozano P, Barrere-Lemaire S, Kondo R, Norman LW, Gourdie RG, Rahme MM, Feld GK, Clark RB, Giles WR, Chien KR, 2000, A novel genetic pathway for sudden cardiac death via defects in the transition between ventricular and conduction system cell lineages. Cell 102(5):671–682 LeeWS,Kwon J,YunDH, LeeYN,WooEY, Park MJ, Lee JS, Han YH, Bae IH, 2014, Specificity protein 1 expression contributes to Bcl-w-induced aggressiveness in glioblastoma multiforme. Mol Cells 37(1):17–23 Black AR, Black JD, Azizkhan-Clifford J, 2001, Sp1 and kruppellike factor family of transcription factors in cell growth regulation and cancer. J Cell Physiol 188(2):143–160 Bouwman P, Philipsen S, 2002, Regulation of the activity of Sp1- related transcription factors. Mol Cell Endocrinol 195(1–2):27–38 Nicolas M, Noe V, Jensen KB, Ciudad CJ, 2001, Cloning and characterization of the 5'-flanking region of the human transcription factor Sp1 gene. J Biol Chem 276(25):22126–22132 Johnson-Pais T, Degnin C, Thayer MJ, 2001, pRB induces Sp1 activity by relieving inhibition mediated by MDM2. Proc Natl Acad Sci U S A 98(5):2211–2216 Wierstra I, 2008, Sp1: emerging roles–beyond constitutive activation of TATA-less housekeeping genes. Biochem Biophys Res Commun 372(1):1–13 Dong Q, Cai N, Tao T, Zhang R, YanW, Li R, Zhang J, Luo H, Shi Y, LuanW, Zhang Y, You Y,Wang Y, Liu N, 2014, An axis involving SNAI1, microRNA-128 and SP1 modulates glioma progression. PLoS One 9(6):e98651 Zhang R, Luo H, Wang S, Chen W, Chen Z, Wang HW, Chen Y, Yang J, Zhang X, Wu W, Zhang SY, Shen S, Dong Q, Zhang Y, Jiang T, Lu D, Zhao S, You Y, Liu N, Wang H, 2014, MicroRNA- 377 inhibited proliferation and invasion of human glioblastoma cells by directly targeting specificity protein 1. Neuro-Oncology 16(11):1510–1522 Luo J, Wang X, Xia Z, Yang L, Ding Z, Chen S, Lai B, Zhang N, 2015, Transcriptional factor specificity protein 1, SP1, promotes the proliferation of glioma cells by up-regulating midkine, MDK). Mol Biol Cell 26(3):430–439 Atadja P, Wong H, Veillete C, Riabowol K, 1995, Overexpression of cyclin D1 blocks proliferation of normal diploid fibroblasts. Exp Cell Res 217(2):205–216 Butt S, Borgquist S, Anagnostaki L, Landberg G, Manjer J, 2014, Breastfeeding in relation to risk of different breast cancer characteristics. BMC Res Notes 7:216 Huang M, Tang SN, Upadhyay G, Marsh JL, Jackman CP, Srivastava RK, Shankar S, 2014, Rottlerin suppresses growth of human pancreatic tumors in nude mice, and pancreatic cancer cells isolated from Kras(G12D, mice. Cancer Lett 353(1):32–40 Monga SP, 2014, Role and regulation of beta-catenin signaling during physiological liver growth. Gene Expr 16(2):51–62 Okumura N, Nakano S, Kay EP, Numata R, Ota A, Sowa Y, Sakai T, Ueno M, Kinoshita S, Koizumi N, 2014, Involvement of cyclin D and p27 in cell proliferation mediated by ROCK inhibitors Y- 27632 and Y-39983 during corneal endothelium wound healing. Invest Ophthalmol Vis Sci 55(1):318–329 Castro-GameroAM, Borges KS,Moreno DA, Suazo VK, Fujinami MM, de Paula Gomes Queiroz R, de Oliveira HF, Carlotti CG Jr, Scrideli CA, Tone LG, 2013, Tetra-O-methyl nordihydroguaiaretic acid, an inhibitor of Sp1-mediated survivin transcription, induces apoptosis and acts synergistically with chemo-radiotherapy in glioblastoma cells. Investig New Drugs 31(4):858–870 Jiang NY, Woda BA, Banner BF, Whalen GF, Dresser KA, Lu D, 2008, Sp1, a new biomarker that identifies a subset of aggressive pancreatic ductal adenocarcinoma. Cancer Epidemiol Biomark Prev 17(7):1648–1652 Wang XB, Peng WQ, Yi ZJ, Zhu SL, Gan QH, 2007, Expression and prognostic value of transcriptional factor sp1 in breast cancer. Ai Zheng 26(9):996–1000 Lesser IM, Rubin RT, Rifkin A, Swinson RP, Ballenger JC, Burrows GD, Dupont RL, Noyes R, Pecknold JC, 1989, Secondary depression in panic disorder and agoraphobia. II. Dimensions of depressive symptomatology and their response to treatment. J Affect Disord 16(1):49–58 Qin H, Sun Y, Benveniste EN, 1999, The transcription factors Sp1, Sp3, and AP-2 are required for constitutive matrix metalloproteinase-2 gene expression in astroglioma cells. J Biol Chem 274(41):29130–29137 Cho JJ, Chae JI, Yoon G, Kim KH, Cho JH, Cho SS, Cho YS, Shim JH, 2014, Licochalcone A, a natural chalconoid isolated from Glycyrrhiza inflata root, induces apoptosis via Sp1 and Sp1 regulatory proteins in oral squamous cell carcinoma. Int J Oncol 45(2):667–674 Duan H, Heckman CA, Boxer LM, 2005, Histone deacetylase inhibitors down-regulate bcl-2 expression and induce apoptosis in t(14;18, lymphomas. Mol Cell Biol 25(5):1608–1619 Lee TJ, Jung EM, Lee JT, Kim S, Park JW, Choi KS, Kwon TK, 2006, Mithramycin A sensitizes cancer cells to TRAIL-mediated apoptosis by down-regulation of XIAP gene promoter through Sp1 sites. Mol Cancer Ther 5(11):2737–2746 Asanuma K, Tsuji N, Endoh T, Yagihashi A, Watanabe N, 2004, Survivin enhances Fas ligand expression via up-regulation of specificity protein 1-mediated gene transcription in colon cancer cells. J Immunol 172(6):3922–3929 Ulrich E, Kauffmann-Zeh A, Hueber AO,Williamson J, Chittenden T, Ma A, Evan G, 1997, Gene structure, cDNA sequence, and expression of murine Bak, a proapoptotic Bcl-2 family member. Genomics 44(2):195–200 Kim YH, Park JW, Lee JY, Kwon TK, 2004, Sodium butyrate sensitizes TRAIL-mediated apoptosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon cancer cells. Carcinogenesis 25(10):1813–1820 Yoshida T,Maeda A, TaniN, Sakai T, 2001, Promoter structure and transcription initiation sites of the human death receptor 5/TRAILR2 gene. FEBS Lett 507(3):381–385 Ganapathy M, Ghosh R, Jianping X, Zhang X, Bedolla R, Schoolfield J, Yeh IT, Troyer DA, Olumi AF, Kumar AP, 2009, Involvement of FLIP in 2-methoxyestradiol-induced tumor regression in transgenic adenocarcinoma of mouse prostate model. Clin Cancer Res 15(5):1601–1611 French LE, Tschopp J, 2002, Defective death receptor signaling as a cause of tumor immune escape. Semin Cancer Biol 12(1):51–55 Maksimovic-Ivanic D, Stosic-Grujicic S, Nicoletti F, Mijatovic S, 2012, Resistance to TRAIL and how to surmount it. Immunol Res 52(1–2):157–168 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1003 EP 1013 DI 10.1007/s12253-018-0422-8 PG 11 ER PT J AU Kiss, N Haluszka, D Lorincz, K Gyongyosi, N Bozsanyi, Sz Banvolgyi, A Szipocs, R Wikonkal, N AF Kiss, Norbert Haluszka, Dora Lorincz, Kende Gyongyosi, Nora Bozsanyi, Szabolcs Banvolgyi, Andras Szipocs, Robert Wikonkal, Norbert TI Quantitative Analysis on Ex Vivo Nonlinear Microscopy Images of Basal Cell Carcinoma Samples in Comparison to Healthy Skin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Basal cell carcinoma; Nonlinear microscopy; Second-harmonic generation; Collagen structure; Quantitative analysis ID Basal cell carcinoma; Nonlinear microscopy; Second-harmonic generation; Collagen structure; Quantitative analysis AB Basal cell carcinoma (BCC) is the most frequent malignant neoplasm in the Caucasian population. There are several therapeutic options for BCC, but surgical excision is considered gold standard treatment. As BCCs often have poorly defined borders, the clinical assessment of the tumor margins can be challenging. Therefore, there is an increasing demand for efficient in vivo imaging techniques for the evaluation of tumor borders prior to and during surgeries. In the near future, nonlinear microscopy techniques might meet this demand.We measured the two-photon excitation fluorescence (TPEF) signal of nicotinamide adenine dinucleotide hydride (NADH) and elastin and second harmonic generation (SHG) signal of collagen on 10 ex vivo healthy control and BCC skin samples and compared the images by different quantitative image analysis methods. These included integrated optical density (IOD) measurements on TPEF and SHG images and application of fast Fourier transform (FFT), CT-FIRE and CurveAlign algorithms on SHG images to evaluate the collagen structure. In the BCC samples, we found significantly lower IOD of both the TPEF and SHG signals and higher collagen orientation index utilizing FFT. CT-FIRE algorithm revealed increased collagen fiber length and decreased fiber angle while CurveAlign detected higher fiber alignment of collagen fibers in BCC. These results are in line with previous findings which describe pronounced changes in the collagen structure of BCC. In the future, these novel image analysis methods could be integrated in handheld nonlinear microscope systems, for sensitive and specific identification of BCC. C1 [Kiss, Norbert] Wigner RCP, Institute for Solid State Physics and Optics, H-1525 Budapest, Hungary. [Haluszka, Dora] Wigner RCP, Institute for Solid State Physics and Optics, H-1525 Budapest, Hungary. [Lorincz, Kende] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Gyongyosi, Nora] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Bozsanyi, Szabolcs] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Banvolgyi, Andras] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Szipocs, Robert] Wigner RCP, Institute for Solid State Physics and Optics, H-1525 Budapest, Hungary. [Wikonkal, Norbert] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. RP Szipocs, R (reprint author), Wigner RCP, Institute for Solid State Physics and Optics, H-1525 Budapest, Hungary. EM r.szipocs@szipocs.com CR Goldenberg G, Karagiannis T, Palmer JB, Lotya J, O'Neill C, Kisa R, Herrera V, Siegel DM, 2016, Incidence and prevalence of basal cell carcinoma, BCC, and locally advanced BCC, LABCC, in a large commercially insured population in the United States: a retrospective cohort study. J Am Acad Dermatol 75(5):957–966 e952. , DOI 10.1016/j.jaad.2016.06.020 Lomas A, Leonardi-Bee J, Bath-Hextall F, 2012, A systematic review of worldwide incidence of nonmelanoma skin cancer. Br J Dermatol 166(5):1069–1080 Zanetti R, Rosso S, Martinez C, Nieto A, Miranda A, Mercier M, Loria D, Osterlind A, Greinert R, Navarro C, 2006, Comparison of risk patterns in carcinoma and melanoma of the skin in men: a multi-centre case–case–control study. Br J Cancer 94:743–751 Crowson A, 2006, Basal cell carcinoma: biology, morphology and clinical implications. Mod Pathol 19:S127–S147 Gandhi SA, Kampp J, 2015, Skin cancer epidemiology, detection, and management. Med Clin North Am 99(6):1323–1335 Seidenari S, Arginelli F, Bassoli S, Cautela J, Cesinaro AM, Guanti M, Guardoli D, Magnoni C, Manfredini M, Ponti G, 2013, Diagnosis of BCC by multiphoton laser tomography. Skin Res Technol 19(1) Cicchi R, Kapsokalyvas D, Pavone FS, 2014, Clinical nonlinear laser imaging of human skin: a review. Biomed Res Int 2014: 903589. , DOI 10.1155/2014/903589 Tong L, Cheng J-X, 2011, Label-free imaging through nonlinear optical signals. Mater Today 14(6):264–273. , DOI 10. 1016/S1369-7021(11)70141-9 BaluM, Zachary CB, HarrisRM, Krasieva TB, Konig K, Tromberg BJ, Kelly KM, 2015, In vivo multiphoton microscopy of basal cell carcinoma. JAMA Dermatol 151(10):1068–1074. , DOI 1001/jamadermatol.2015.0453 10. Seidenari S, Arginelli F, Bassoli S, Cautela J, Cesinaro AM, Guanti M, Guardoli D, Magnoni C, Manfredini M, Ponti G, Konig K, 2013, Diagnosis of BCC by multiphoton laser tomography. Skin Res Technol 19(1):e297–e304. , DOI 10.1111/j.1600- 0846.2012.00643.x Kiss N, Krolopp A, Lorincz K, Banvolgyi A, Szipocs R,Wikonkal N, 2017, Stain-free histopathology of basal cell carcinoma by dual vibration resonance frequency CARS microscopy. Pathol Oncol Res , DOI 10.1007/s12253-017-0356-6 Wu S, Li H, Yang H, Zhang X, Li Z, Xu S, 2011, Quantitative analysis on collagen morphology in aging skin based on multiphoton microscopy. J Biomed Opt 16(4):040502. , DOI 10. 1117/1.3565439 Bredfeldt JS, Liu Y, Pehlke CA, Conklin MW, Szulczewski JM, Inman DR, Keely PJ, Nowak RD, Mackie TR, Eliceiri KW, 2014, Computational segmentation of collagen fibers from secondharmonic generation images of breast cancer. J Biomed Opt 19(1) Drifka CR, Loeffler AG, Mathewson K, Mehta G, Keikhosravi A, Liu Y, Lemancik S, Ricke WA, Weber SM, Kao WJ, 2016, Comparison of picrosirius red staining with second harmonic generation imaging for the quantification of clinically relevant collagen fiber features in histopathology samples. J Histochem Cytochem 64(9):519–529 Li J, Bower AJ, Vainstein V, Gluzman-Poltorak Z, Chaney EJ, Marjanovic M, Basile LA, Boppart SA, 2015, Effect of recombinant interleukin-12 on murine skin regeneration and cell dynamics using in vivo multimodal microscopy. Biomed Opt Express 6(11): 4277–4287. , DOI 10.1364/boe.6.004277 Haluszka D, Lorincz K, Kiss N, Szipocs R, Kuroli E, Gyongyosi N, Wikonkal NM(2016, Diet-induced obesity skin changesmonitored by in vivo SHG and ex vivo CARS microscopy. Biomed Opt Express 7(11):4480–4489. , DOI 10.1364/BOE.7.004480 Lorincz K, Haluszka D, Kiss N, Gyongyosi N, Banvolgyi A, Szipocs R,Wikonkal NM, 2017, Voluntary exercise improves murine dermal connective tissue status in high-fat diet-induced obesity. Arch Dermatol Res 309(3):209–215. , DOI 10.1007/ s00403-017-1715-6 Cicchi R, Kapsokalyvas D, De Giorgi V, Maio V, Van Wiechen A, Massi D, Lotti T, Pavone FS, 2010, Scoring of collagen organization in healthy and diseased human dermis by multiphoton microscopy. J Biophotonics 3(1-2):34–43 James W, Cooley P, Peter DW, 1967, Historical notes on the fast Fourier transform. Proc IEEE 55(10):1675 Ayres C, Bowlin GL, Henderson SC, Taylor L, Shultz J, Alexander J, Telemeco TA, Simpson DG, 2006, Modulation of anisotropy in electrospun tissue-engineering scaffolds: analysis of fiber alignment by the fast Fourier transform. Biomaterials 27(32):5524–5534 Lu K, Chen J, Zhuo S, Zheng L, Jiang X, Zhu X, Zhao J, 2009, Multiphoton laser scanning microscopy of localized scleroderma. Skin Res Technol 15(4):489–495 Walsh AJ, Cook RS, Lee JH, Arteaga CL, Skala MC, 2015, Collagen density and alignment in responsive and resistant trastuzumab-treated breast cancer xenografts. J Biomed Opt 20(2) Gilje O, O'Leary PA, Baldes EJ, 1953, Capillary microscopic examination in skin diseases. AMA Arch Derm Syphilol 68(2):136– 147 Goldman L, 1958, A simple portable skin microscope for surface microscopy. AMA Arch Derm 78(2):246–247. , DOI 10. 1001/archderm.1958.01560080106017 Argenziano G, Soyer HP, Chimenti S, Talamini R, Corona R, Sera F, Binder M, Cerroni L, De Rosa G, Ferrara G, 2003, Dermoscopy of pigmented skin lesions: results of a consensus meeting via the internet. J Am Acad Dermatol 48, 5):679–693 Braun RP, Rabinovitz HS, Oliviero M, KopfAW, Saurat J-H, 2005, Dermoscopy of pigmented skin lesions. J Am Acad Dermatol 52(1):109–121. , DOI 10.1016/j.jaad.2001.11.001 Anagnostopoulos CN, Vergados DD, Anagnostopoulos I, Mintzias P, 2014, Total dermoscopy score calculation using quantitative measurements in digital dermoscopy. Conf Proc IEEE Eng Med Biol Soc 2014:6744–6747. , DOI 10. 1109/embc.2014.6945176 Alexander H, Miller DL, 1979, Determining skin thickness with pulsed ultra sound. J Invest Dermatol 72(1):17–19 Lockwood G, Turnball D, Christopher D, Foster FS, 1996, Beyond 30MHz [applications of high-frequency ultrasound imaging]. IEEE Eng Med Biol Mag 15(6):60–71 Piotrzkowska-Wroblewska H, Litniewski J, Szymanska E, Nowicki A, 2015, Quantitative sonography of basal cell carcinoma. Ultrasound Med Biol 41(3):748–759. , DOI 10.1016/j. ultrasmedbio.2014.11.016 Fercher A, Roth E, 1986, Ophthalmic laser interferometry. In: Optical instrumentation for biomedical laser applications. International Society for Optics and Photonics, pp 48–52 Gambichler T, Orlikov A, Vasa R, Moussa G, Hoffmann K, Stucker M, Altmeyer P, Bechara FG, 2007, In vivo optical coherence tomography of basal cell carcinoma. J Dermatol Sci 45(3):167–173. , DOI 10.1016/j.jdermsci.2006.11.012 Marvdashti T, Duan L, Aasi SZ, Tang JY, Ellerbee Bowden AK, 2016, Classification of basal cell carcinoma in human skin using machine learning and quantitative features captured by polarization sensitive optical coherence tomography. Biomed Opt Express 7(9): 3721–3735 Calzavara-Pinton P, Longo C, Venturini M, Sala R, Pellacani G, 2008, Reflectance confocal microscopy for in vivo skin imaging. Photochem Photobiol 84(6):1421–1430. , DOI 10.1111/j. 1751-1097.2008.00443.x Gambichler T, Jaedicke V, Terras S, 2011, Optical coherence tomography in dermatology: technical and clinical aspects. Arch Dermatol Res 303(7) Kadouch DJ, Schram ME, Leeflang MM, Limpens J, Spuls PI, de Rie MA, 2015, In vivo confocal microscopy of basal cell carcinoma: a systematic review of diagnostic accuracy. J Eur Acad Dermatol Venereol 29(10):1890–1897. , DOI 10. 1111/jdv.13224 Lin SJ, Jee SH, Kuo CJ, Wu RJ, Lin WC, Chen JS, Liao YH, Hsu CJ, Tsai TF, Chen YF, Dong CY, 2006, Discrimination of basal cell carcinoma from normal dermal stroma by quantitative multiphoton imaging. Opt Lett 31(18):2756–2758 Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG, Keely PJ, 2006, Collagen reorganization at the tumor-stromal interface facilitates local invasion. BMC Med 4(1):38. , DOI 10.1186/1741-7015-4-38 ShortMA, Lui H,McLean D, Zeng H, Alajlan A, Chen XK, 2006, Changes in nuclei and peritumoral collagen within nodular basal cell carcinomas via confocal micro-Raman spectroscopy. J Biomed Opt 11(3):34004. , DOI 10.1117/1.2209549 Krolopp A, Csakanyi A, Haluszka D, Csati D, Vass L, Kolonics A, Wikonkal N, Szipocs R, 2016, Handheld nonlinear microscope system comprising a 2 MHz repetition rate, mode-locked Yb-fiber laser for in vivo biomedical imaging. Biomed Opt Express 7(9): 3531–3542. , DOI 10.1364/boe.7.003531 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1015 EP 1021 DI 10.1007/s12253-018-0445-1 PG 7 ER PT J AU Decsi, G Soki, J Pap, B Dobra, G Harmati, M Kormondi, S Pankotai, T Braunitzer, G Minarovits, J Sonkodi, I Urban, E Nemeth, BI Nagy, K Buzas, K AF Decsi, Gabor Soki, Jozsef Pap, Bernadett Dobra, Gabriella Harmati, Maria Kormondi, Sandor Pankotai, Tibor Braunitzer, Gabor Minarovits, Janos Sonkodi, Istvan Urban, Edit Nemeth, Balazs Istvan Nagy, Katalin Buzas, Krisztina TI Chicken or the Egg: Microbial Alterations in Biopsy Samples of Patients with Oral Potentially Malignant Disorders SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE OPMD; Oral microbiome; Metagenome sequencing; Lichen; Leukoplakia ID OPMD; Oral microbiome; Metagenome sequencing; Lichen; Leukoplakia AB Oral carcinogenesis often leads to the alteration of the microbiota at the site of the tumor, but data are scarce regarding the microbial communities of oral potentially malignant disorders (OPMDs). Punch biopsies were taken from healthy and nonhealthy mucosa of OPMD patients to analyze the microbiome using metagenome sequencing. In healthy oral mucosa biopsies the bacterial phyla Firmicutes, Fusobacteria, Proteobacteria, Actinobacteria and Bacteroidetes were detected by Ion Torrent sequencing. The same phyla as well as the phyla Fibrobacteres and Spirochaetes were present in the OPMD biopsies. On the species level, there were 10 bacterial species unique to the healthy tissue and 35 species unique to the OPMD lesions whereas eight species were detected in both samples. We observed that the relative abundance of Streptococcus mitis decreased in the OPMD lesions compared to the uninvolved tissue. In contrast, the relative abundance of Fusobacterium nucleatum, implicated in carcinogenesis, was elevated in OPMD.We detected markedly increased bacterial diversity in the OPMD lesions compared to the healthy oral mucosa. The ratio of S. mitis and F. nucleatum are characteristically altered in the OPMD lesions compared to the healthy mucosa. C1 [Decsi, Gabor] University of Szeged, Faculty of Dentistry, Department of Oral Surgery, Tisza Lajos krt. 64, H-6720 Szeged, Hungary. [Soki, Jozsef] University of Szeged, Albert Szent-Gyorgyi Clinical Centre, Institute of Clinical Microbiology, Semmelweis u. 6, H-6725 Szeged, Hungary. [Pap, Bernadett] Hungarian Academy of Sciences, Biological Research Center, Temesvari krt. 62, H-6726 Szeged, Hungary. [Dobra, Gabriella] Hungarian Academy of Sciences, Biological Research Center, Temesvari krt. 62, H-6726 Szeged, Hungary. [Harmati, Maria] Hungarian Academy of Sciences, Biological Research Center, Temesvari krt. 62, H-6726 Szeged, Hungary. [Kormondi, Sandor] University of Szeged, Albert Szent-Gyorgyi Clinical Centre, Department of Traumatology, Semmelweis u. 6, H-6725 Szeged, Hungary. [Pankotai, Tibor] University of Szeged, Department of Medical Chemistry, Kozep fasor 52, H-6726 Szeged, Hungary. [Braunitzer, Gabor] dicomLAB Ltd, Pulz u. 46/b, H-6724 Szeged, Hungary. [Minarovits, Janos] University of Szeged, Faculty of Dentistry, Department of Oral Biology and Experimental Dental Research, Tisza Lajos krt. 64, H-6720 Szeged, Hungary. [Sonkodi, Istvan] University of Szeged, Faculty of Dentistry, Department of Oral Surgery, Tisza Lajos krt. 64, H-6720 Szeged, Hungary. [Urban, Edit] University of Szeged, Albert Szent-Gyorgyi Clinical Centre, Institute of Clinical Microbiology, Semmelweis u. 6, H-6725 Szeged, Hungary. [Nemeth, Balazs Istvan] University of Szeged, Department of Dermatology and Allergology, H-6720 Szeged, Hungary. [Nagy, Katalin] University of Szeged, Faculty of Dentistry, Department of Oral Surgery, Tisza Lajos krt. 64, H-6720 Szeged, Hungary. [Buzas, Krisztina] Hungarian Academy of Sciences, Biological Research Center, Temesvari krt. 62, H-6726 Szeged, Hungary. RP Buzas, K (reprint author), Hungarian Academy of Sciences, Biological Research Center, H-6726 Szeged, Hungary. EM kr.buzas@gmail.com;buzas.krisztina@brc.mta.hu CR Morse DE, Psoter WJ, Cleveland D et al, 2007, Smoking and drinking in relation to oral cancer and oral epithelial dysplasia. Cancer Causes Control 18(9):919–929 Jepsen SA, Closmann JJ, 2008, The insidious nature and presentation of oral squamous cell carcinoma in the low-risk population. Gen Dent 56(1):78–82 Llewellyn CD, Linklater K, Bell J et al, 2003, Squamous cell carcinoma of the oral cavity in patients aged 45 years and under: a descriptive analysis of 116 cases diagnosed in the South East of England from 1990 to 1997. Oral Oncol 39(2):106–114 Nagy KN, Sonkodi I, Szoke I et al, 1998, Themicroflora associated with human oral carcinomas. Oral Oncol 34(4):304–308 Sarode SC, Sarode GS, Tupkari JV, 2012, Oral potentially malignant disorders: precising the definition. Oral Oncol 48(9):759–760 van der Waal I, 2009, Potentially malignant disorders of the oral and oropharyngeal mucosa; terminology, classification and present concepts of management. Oral Oncol 45(4–5):317–323 Sonkodi I, 2009, Oral and Maxilofacial medicine. Semmelweis Publisher, Budapest Silverman S Jr, Gorsky M, 1997, Proliferative verrucous leukoplakia. A follow-up study of 54 cases. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 84(2):154–157 Feher E, Gall T, Murvai M et al, 2009, Investigation of the occurrence of torque tenovirus in malignant and potentially malignant disorders associated with human papillomavirus. J Med Virol 81(11):1975–1981 Feher E, Kardos G, Gall T et al, 2011, Comparison of diversity of torque teno virus 1 in different mucosal tissues and disorders. Acta Microbiol Immunol Hung 58(4):319–337 Kis A, Feher E, Gall T et al, 2009, Epstein-Barr virus prevalence in oral squamous cell cancer and in potentially malignant oral disorders in an eastern Hungarian population. Eur J Oral Sci 117(5):536–540 Wu L, Feng J, Shi L et al, 2013, Candidal infection in oral leukoplakia: a clinicopathologic study of 396 patients from eastern China. Ann Diagn Pathol 17(1):37–40 Hebbar PB, Pai A, D S., 2013, Mycological and histological associations of Candida in oral mucosal lesions. J Oral Sci 55(2):157–160 Dilhari A, Weerasekera MM, Siriwardhana A et al, 2016, Candida infection in oral leukoplakia: an unperceived public health problem. Acta Odontol Scand 74(7):565–569 Gainza-Cirauqui ML, Nieminen MT, Novak Frazer L et al, 2013, Production of carcinogenic acetaldehyde by Candida albicans from patients with potentially malignant oral mucosal disorders. J Oral Pathol Med. 42(3):243–249 Bakri MM, Cannon RD, Holmes AR et al, 2014, Detection of Candida albicans ADH1 and ADH2 mRNAs in human archival oral biopsy samples. J Oral Pathol Med 43(9):704–710 Kazanowska-Dygdala M, Dus I, Radwan-Oczko M, 2016, The presence of helicobacter pylori in oral cavities of patientsn with leukoplakia and oral lichenplanus. J Appl Oral Sci 24(1):18–23 Mizuki H, Kawamura T, Nagasawa D, 2015, In situ immunohistochemical detection of intracellular mycoplasma salivarium in the epithelial cells of oral leukoplakia. J Oral PatholMed. 44(2):134–144 Meisel P, Holtfreter B, Biffar R et al, 2012, Association of periodontitis with the risk of oral leukoplakia. Oral Oncol 48(9):859–863 Hu X, Zhang Q, Hua H et al, 2016, Changes in the salivary microbiota of oral leukoplakia and oral cancer. Oral Oncol 56:e6–e8 Hernandez BY, Zhu X, Goodman MT et al, 2017, Betel nut chewing, oral premalignant lesions, and the oral microbiome. PLoS One 12(2):e0172196 Srinivas K, Aravinda K, Ratnakar P et al, 2011, Oral lichen planus – Review on etiopathogenesis. Natl J Maxillofac Surg 2(1):15–16 Payeras MR, Cherubini K, Figueiredo MA et al, 2013, Oral lichen planus: focus on etiopathogenesis. Arch Oral Biol 58(9):1057– 1069 Georgakopoulou EA, Achtari MD, Achtaris M et al, 2012, Oral lichen planus as a preneoplastic inflammatory model. J Biomed Biotechnol 2012:759626 Roopashree MR, Gondhalekar RV, Shashikanth MC et al, 2010, Pathogenesis of oral lichen planus–a review. J Oral Pathol Med. 39(10):729–734 Gorouhi F, Davari P, Fazel N, 2014, Cutaneous and mucosal lichen planus: a comprehensive review of clinical subtypes, risk factors, diagnosis, and prognosis. ScientificWorld Journal 2014:742826 Backman K, Jontell M, 2007, Microbial-associated oral lichenoid reactions. Oral Dis 13(4):402–406 Taghavi Zenouz A, Mehdipour M, Jafari Heydarlou M et al, 2010, Relationship between Lichen Planus and Helicobacter pylori Infection. J Dent Res Dent Clin Dent Prospects 4(1):17–20 Attia EA, Abdel Fattah NS, Abdella HM, 2010, Upper gastrointestinal findings and detection of helicobacter pylori in patients with oral lichen planus. Clin Exp Dermatol 35(4):355–360 Pourshahidi S, Fakhri F, Ebrahimi H et al, 2012, Lack of association between helicobacter pylori infection and oral lichen planus. Asian Pac J Cancer Prev 13(5):1745–1747 Izol B, Karabulut AA, Biyikoglu I et al, 2010, Investigation of upper gastrointestinal tract involvement and H. Pylori presence in lichen planus: a case-controlled study with endoscopic and histopathological findings. Int J Dermatol 49(10):1121–1126 Hulimavu SR, Mohanty L, Tondikulam NV et al, 2014, No evidence for Helicobacter pylori in oral lichen planus. J Oral Pathol Med. 43(8):576–578 Boorghani M, Gholizadeh N, Taghavi Zenouz A et al, 2010, Oral lichen planus: clinical features, etiology, treatment and management; a review of literature. J Dent Res Dent Clin Dent Prospects. 4(1):3–9 Ertugrul AS, Arslan U, Dursun R et al, 2013, Periodontopathogen profile of healthy and oral lichen planus patients with gingivitis or periodontitis. Int J Oral Sci 5(2):92–97 El-Naggar AK, Chan JKC, Takata T, Grandis JR, Slootweg PJ, 2017, Tumours of the oral cavity andmobile tongue In: The fourth edition of the head and neckWorld Health Organization blue book: editors' perspectives. Hum Pathol 66:10–12 Sharma PK, Capalash N, Kaur J, 2007, An improved method for single step purification of metagenomic DNA. Mol Biotechnol 36(1):61–63 Periasamy S, Chalmers NI, Du-Thumm L et al, 2009, Fusobacterium nucleatum ATCC 10953 requires Actinomyces naeslundii ATCC 43146 for growth on saliva in a three-species community that includes Streptococcus oralis 34. Appl Environ Microbiol 75(10):3250–3257 Rubinstein MR, Wang X, Liu W et al, 2013, Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating Ecadherin/ β-catenin signaling via its FadA adhesion. Cell Host Microbe 14(2):195–206 Schmidt BL, Kuczynski J, Bhattacharya A et al, 2014, Changes in abundance of oral microbiota associated with oral cancer. PLoS One 9(6):e98741 Al-Hebshi NN, Nasher AT,MaryoudMYet al, 2017, Inflammatory bacteriome featuring fusobacterium nucleatum and Pseudomonas aeruginosa identified in association with oral squamous cell carcinoma. Sci Rep 7(1):1834 Arduino PG, Romano F, Sasia D et al, 2017, Subgingival microbiota in white patients with Desquamative gingivitis: a cross-sectional study. J Periodontol 88(7):643–650 Kreth J,Merritt J, Qi F, 2009, Bacterial and host interactions of oral streptococci. DNA Cell Biol 28(8):397–403 Van Hoogmoed CG, Geertsema-Doornbusch GI, Teughels Wet al, 2008, Reduction of periodontal pathogens adhesion by antagonistic strains. Oral Microbial Immunol 23(1):43–48 Standar K, Kreikemeyer B, Redanz S et al, 2010, Setup of an in vitro test system for basic studies on biofilm behavior of mixedspecies cultures with dental and periodontal pathogens. PLoS One 5(10):e13135 Otsuki K, Imai N, 2017, Effects of lactoferrin in 6 patients with refractory bacterial vaginosis. Biochem Cell Biol 95(1):31–33 Klaric M, Mandic V, Lovric S, e al., 2017, Antimicrobial efficacy of probiotic-containing toothpastes: an in vitro evaluation. Med Glas, Zenica, 14(1):16–24 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1023 EP 1033 DI 10.1007/s12253-018-0457-x PG 11 ER PT J AU Mlak, R Krawczyk, P Homa-Mlak, I Powrozek, T Ciesielka, M Koziol, P Milanowski, J Malecka-Massalska, T AF Mlak, Radoslaw Krawczyk, Pawel Homa-Mlak, Iwona Powrozek, Tomasz Ciesielka, Marzanna Koziol, Piotr Milanowski, Janusz Malecka-Massalska, Teresa TI Predictive Value of Single Nucleotide Polymorphisms of ERCC1, XPA, XPC, XPD and XPG Genes, Involved in NER Mechanism in Patients with Advanced NSCLC Treated with Cisplatin and Gemcitabine SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; Polymorphism; DNA repair; XPD; chemotherapy ID Non-small cell lung cancer; Polymorphism; DNA repair; XPD; chemotherapy AB The combination of cisplatin and gemcitabine is still one of the most frequently used first-line chemotherapy scheme in patients with advanced non-small cell lung cancer (NSCLC), in which tyrosine kinase inhibitors (TKIs) cannot be administered. Unfortunately, more than half of the patients have no benefit from chemotherapy but are still exposed to its toxic effects. Therefore, single nucleotide polymorphisms (SNPs) in the genes involved in nucleotide excision repair (NER) mechanism may be a potential predictive factor of efficiency of cytostatic based chemotherapy. The aim of the study was to evaluate the correlation between SNPs of the genes involved in NER mechanism and the effectiveness of chemotherapy based on cisplatin and gemcitabine in patients with advanced NSCLC. The study group included 91 NSCLC patients treated with first-line chemotherapy using cisplatin and gemcitabine. Genotyping was carried out using a mini-sequencing technique (SNaPshot™ PCR). The median progression-free survival (PFS) was significantly shorter in carriers of CC genotype of the XPD/ERCC2 (2251A > C) gene compared to patients with AA/AC genotypes (2 vs. 4.5 months; p = 0.0444; HR = 3.19, 95% CI:1.03–9.91). Rare CC genotype of XPD/ERCC2 gene, may be considered as an unfavorable predictive factor for chemotherapy based on cisplatin and gemcitabine in patients with advanced NSCLC. C1 [Mlak, Radoslaw] Medical University of Lublin, Department of Human Physiology, Radziwillowska 11, 20-080 Lublin, Poland. [Krawczyk, Pawel] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Homa-Mlak, Iwona] Medical University of Lublin, Department of Human Physiology, Radziwillowska 11, 20-080 Lublin, Poland. [Powrozek, Tomasz] Medical University of Lublin, Department of Human Physiology, Radziwillowska 11, 20-080 Lublin, Poland. [Ciesielka, Marzanna] Medical University of Lublin, Department of Forensic Medicine, Jaczewskiego 8b, 20-090 Lublin, Poland. [Koziol, Piotr] Medical University of Lublin, Department of Forensic Medicine, Jaczewskiego 8b, 20-090 Lublin, Poland. [Milanowski, Janusz] Medical University of Lublin, Department of Pneumonology, Oncology and Allergology, Jaczewskiego 8, 20-954 Lublin, Poland. [Malecka-Massalska, Teresa] Medical University of Lublin, Department of Human Physiology, Radziwillowska 11, 20-080 Lublin, Poland. RP Mlak, R (reprint author), Medical University of Lublin, Department of Human Physiology, 20-080 Lublin, Poland. EM radoslaw.mlak@gmail.com CR Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66(1):7–30 NCCN Clinical Practice Guidelines in Oncology, NCCN Guidelines®, Non-Small Cell Lung Cancer Version 4.2016 www. nccn.org Accessed 1 July 2017 Blumenthal GM, Karuri SW, Zhang H, Zhang L, Khozin S, Kazandjian D, Tang S, Sridhara R, Keegan P, Pazdur R, 2015, Overall response rate, progression-free survival, and overall survival with targeted and standard therapies in advanced non-small-cell lung cancer: US Food and Drug Administration trial-level and patient-level analyses. J Clin Oncol 33(9):1008–1014 Gridelli C, de Marinis F, Cappuzzo F, di Maio M, Hirsch FR, Mok T, Morgillo F, Rosell R, Spigel DR, Yang JCH, Ciardiello F, 2014, Treatment of advanced non-small-cell lung cancer with epidermal growth factor receptor, EGFR, mutation or ALK gene rearrangement: results of an international expert panel meeting of the Italian Association of Thoracic Oncology. Clin Lung Cancer 15:173–181 Simon GR, Somaiah N, 2014, A tabulated summary of targeted and biologic therapies for non-small-cell lung cancer. Clin Lung Cancer 15:21–51 La-Beck NM, Jean GW, Huynh C et al, 2015, Immune checkpoint inhibitors: new insights and current place in Cancer therapy. Pharmacotherapy 35(10):963–976 Peters GJ, Avan A, Ruiz MG, Orsini V, Avan A, Giovannetti E, Smit EF, 2014, Predictive role of repair enzymes in the efficacy of cisplatin combinations in pancreatic and lung cancer. Anticancer Res 34(1):435–442 Simon GR, Ismail-Khan R, Bepler G, 2007, Nuclear excision repairbased personalized therapy for non-small cell lung cancer: from hypothesis to reality. Int J Biochem Cell Biol 39(7–8):1318–1328 Sullivan I, Salazar J, Majem M, Pallares C, del Rio E, Paez D, Baiget M, Barnadas A, 2014, Pharmacogenetics of the DNA repair pathways in advanced non-small cell lung cancer patients treated with platinum-based chemotherapy. Cancer Lett 353(2):160–166 Simon GR, Schell MJ, Begum M, Kim J, Chiappori A, Haura E, Antonia S, Bepler G, 2012, Preliminary indication of survival benefit from ERCC1 and RRM1-tailored chemotherapy in patients with advanced nonsmall cell lung cancer: evidence from an individual patient analysis. Cancer 118:2525–2531 Zhang Q, Zhu X, Zhang L, Sun S, Huang J, Lin Y, 2014, A prospective study of biomarker-guided chemotherapy in patients with non-small cell lung cancer. Cancer Chemother Pharmacol 74:839–846 JoergerM, Burgers JA, Baas P, DoodemanVD, Smits PHM, Jansen RS, Vainchtein LD, Rosing H, Huitema ADR, Beijnen JH, Schellens JHM, 2012, Gene polymorphisms, pharmacokinetics, and hematological toxicity in advanced non-small-cell lung cancer patients receiving cisplatin/gemcitabine. Cancer Chemother Pharmacol 69(1):25–33 Murphy A, Chu JH, Xu M, Carey VJ, Lazarus R, Liu A, Szefler SJ, Strunk R, DeMuth K, Castro M, Hansel NN, Diette GB, Vonakis BM, Franklin Adkinson N Jr, Klanderman BJ, Senter- Sylvia J, Ziniti J, Lange C, Pastinen T, Raby BA, 2010, Mapping of numerous disease-associated expression polymorphisms in primary peripheral blood CD4+ lymphocytes. Hum Mol Genet 19:4745–4757 Zhao W, Hu XJ et al, 2013, Polymorphisms in the base excision repair pathway modulate prognosis of platinum-based chemotherapy in advanced non-small cell lung cancer. Cancer Chemother Pharmacol 71(5):1287–1295 Mazzoni F, Cecere FL, Meoni G, Giuliani C, Boni L, Camerini A, Lucchesi S, Martella F, Amoroso D, Lucherini E, Torricelli F, di Costanzo F, 2013, Phase II trial of customized first line chemotherapy according to ERCC1 and RRM1 SNPs in patients with advanced non-small-cell lung cancer. Lung Cancer 82:288–293 Camps C, Sarries C, Roig B, Javier Sanchez J, Queralt C, Sancho E, Martinez N, Taron M, Rosell R, 2003, Assessment of nucleotide excision repair XPD polymorphisms in the peripheral blood of gemcitabine/cisplatin-treated advanced non-small-cell lung cancer patients. Clin Lung Cancer 4(4):237–241 Liao WY, Shih JY, Chang GC, Cheng YK, Yang JCH, Chen YM, Yu CJ, 2012, Genetic polymorphism of XRCC1 Arg399Gln is associated with survival in non-small-cell lung cancer patients treated with gemcitabine/platinum. J Thorac Oncol 7(6):973–981 Tibaldi C, Giovannetti E, Vasile E, Mey V, Laan AC, Nannizzi S, di Marsico R, Antonuzzo A, Orlandini C, Ricciardi S, del Tacca M, Peters GJ, Falcone A, Danesi R, 2008, Correlation of CDA, ERCC1, and XPD polymorphisms with response and survival in gemcitabine/cisplatin-treated advanced non-small cell lung cancer patients. Clin Cancer Res 14(6):1797–1803 de las Penas R, Sanchez-Ronco M, Alberola V et al, 2006, Polymorphisms in DNA repair genes modulate survival in cisplatin/gemcitabine-treated non-small-cell lung cancer patients. Ann Oncol 17(4):668–675 Zhang ZY, Tian X,Wu R, Liang Y, Jin XY, 2012, Predictive role of ERCC1 and XPD genetic polymorphisms in survival of Chinese non-small cell lung cancer patients receiving chemotherapy. Asian Pac J Cancer Prev 13(6):2583–2586 Qin Q, Zhang C, Yang X, Zhu H, Yang B, Cai J, Cheng H, Ma J, Lu J, Zhan L, Liu J, Liu Z, Xu L, Sun X, 2013, Polymorphisms in XPD gene could predict clinical outcome of platinum-based chemotherapy for non-small cell lung cancer patients: a meta-analysis of 24 studies. PLoS One 8(11):e79864 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1035 EP 1045 DI 10.1007/s12253-018-0459-8 PG 11 ER PT J AU Carvalho, CdA Melendez, EM Sabato, dSC Palmero, IE Arantes, MRBL Neto, SC Carvalho, LA AF Carvalho, Carolina de Ana Melendez, Eliseo Matias Sabato, da Silva Cristina Palmero, Inez Edenir Arantes, Maria Rebolho Batista Lidia Neto, Scapulatempo Cristovam Carvalho, Lopes Andre TI Clinical and Molecular Characterization of Surgically Treated Oropharynx Squamous Cell Carcinoma Samples SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oropharyngeal cancer; Human papillomavirus; Methylation; Mutation; Prognosis ID Oropharyngeal cancer; Human papillomavirus; Methylation; Mutation; Prognosis AB A better understanding of the clinical and molecular features of oropharyngeal squamous cell carcinomas (OPSCC) may help in the development of strategies for a better patient management, improving survival rates. This retrospective study conducted a clinical and molecular characterization of surgically treated OPSCC samples. Paraffin-embedded samples from a series of cases were screened for high-risk (HR) human papillomavirus (HPV) infection, methylation of a 5-gene panel, p53 expression, and TP53 mutation. The study was conducted at Barretos Cancer Hospital. Twenty-five surgically treated OPSCC with available tissue were included in the study. Samples were classified according to HPV status and molecular features and some of these characteristics were associated to clinical data. Twenty percent of the cases were HR-HPV positive and 62.5% presented TP53 mutations. DAPK hypermethylation was associated with HPV status (p = 0.023), while methylated CCNA1 was inversely related to TP53 mutations in primary tumors (p = 0.042) and associated with a better disease-free survival (22.3% vs. 100.0%; p = 0.028) and overall survival (8.0% vs. 100.0%; p = 0.012). The results show differences regarding molecular and clinical characteristics in the oropharynx cases identified that should be validated in more cases to confirm whether these differences are able to classify patients according to outcome and help in a more thorough patient management. C1 [Carvalho, Carolina de Ana] Barretos Cancer Hospital, Molecular Oncology Research Center, Rua Antenor Duarte Vilela, 1331, 14784-400 Barretos, SP, Brazil. [Melendez, Eliseo Matias] Barretos Cancer Hospital, Molecular Oncology Research Center, Rua Antenor Duarte Vilela, 1331, 14784-400 Barretos, SP, Brazil. [Sabato, da Silva Cristina] Barretos Cancer Hospital, Molecular Oncology Research Center, Rua Antenor Duarte Vilela, 1331, 14784-400 Barretos, SP, Brazil. [Palmero, Inez Edenir] Barretos Cancer Hospital, Molecular Oncology Research Center, Rua Antenor Duarte Vilela, 1331, 14784-400 Barretos, SP, Brazil. [Arantes, Maria Rebolho Batista Lidia] Barretos Cancer Hospital, Molecular Oncology Research Center, Rua Antenor Duarte Vilela, 1331, 14784-400 Barretos, SP, Brazil. [Neto, Scapulatempo Cristovam] Barretos Cancer Hospital, Molecular Oncology Research Center, Rua Antenor Duarte Vilela, 1331, 14784-400 Barretos, SP, Brazil. [Carvalho, Lopes Andre] Barretos Cancer Hospital, Molecular Oncology Research Center, Rua Antenor Duarte Vilela, 1331, 14784-400 Barretos, SP, Brazil. RP Carvalho, CdA (reprint author), Barretos Cancer Hospital, Molecular Oncology Research Center, 14784-400 Barretos, Brazil. EM anacarol.oak@gmail.com CR Gillison ML, D'Souza G, Westra W, Sugar E, Xiao W, Begum S, Viscidi R, 2008, Distinct risk factor profiles for human papillomavirus type 16-positive and human papillomavirus type 16-negative head and neck cancers. J Natl Cancer Inst 100(6):407–420. https:// doi.org/10.1093/jnci/djn025 Marur S, D'Souza G, Westra WH, Forastiere AA, 2010, HPVassociated head and neck cancer: a virus-related cancer epidemic. Lancet Oncol 11(8):781–789. , DOI 10.1016/S1470- 2045(10)70017-6 Marklund L, Hammarstedt L, 2011, Impact of HPV in oropharyngeal Cancer. J Oncol 2011:509036. , DOI 10.1155/2011/ 509036 Gillison ML, Chaturvedi AK, Anderson WF, Fakhry C, 2015, Epidemiology of human papillomavirus-positive head and neck squamous cell carcinoma. J Clin Oncol Off J Am Soc Clin Oncol 33(29):3235–3242. , DOI 10.1200/JCO.2015.61.6995 Kreimer AR, Clifford GM, Boyle P, Franceschi S, 2005, Human papillomavirus types in head and neck squamous cell carcinomas worldwide: a systematic review. Cancer Epidemiol Biomarkers Prev 14(2):467–475. , DOI 10.1158/1055-9965.EPI-04- 0551 Zhen W, Karnell LH, Hoffman HT, Funk GF, Buatti JM, Menck HR, 2004, The National Cancer Data Base report on squamous cell carcinoma of the base of tongue.HeadNeck 26(8):660–674. https:// doi.org/10.1002/hed.20064 Lybak S, Ljokjel B, Haave H, Karlsdottir A, Vintermyr OK, Aarstad HJ, 2017, Primary surgery results in no survival benefit compared to primary radiation for oropharyngeal cancer patients stratified by high-risk human papilloma virus status. Eur Arch Otorhinolaryngol 274(1):477–487. , DOI 10.1007/ s00405-016-4203-2 Chaturvedi AK, Engels EA, Pfeiffer RM, Hernandez BY, Xiao W, Kim E, Jiang B, Goodman MT, Sibug-Saber M, Cozen W, Liu L, Lynch CF, Wentzensen N, Jordan RC, Altekruse S, Anderson WF, Rosenberg PS, Gillison ML, 2011, Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol Off J Am Soc Clin Oncol 29(32):4294–4301. https://doi. org/10.1200/JCO.2011.36.4596 GillisonML, KochWM, Capone RB, SpaffordM,WestraWH,Wu L, Zahurak ML, Daniel RW, Viglione M, Symer DE, Shah KV, Sidransky D, 2000, Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J Natl Cancer Inst 92(9):709–720 Mellin H, Friesland S, Lewensohn R, Dalianis T, Munck-Wikland E, 2000, Human papillomavirus, HPV, DNA in tonsillar cancer: clinical correlates, risk of relapse, and survival. Int J Cancer 89(3): 300–304 Hafkamp HC, Manni JJ, Haesevoets A, Voogd AC, Schepers M, Bot FJ, Hopman AH, Ramaekers FC, Speel EJ, 2008, Marked differences in survival rate between smokers and nonsmokers with HPV 16-associated tonsillar carcinomas. Int J Cancer 122(12): 2656–2664. , DOI 10.1002/ijc.23458 Goldenberg D, Begum S, Westra WH, Khan Z, Sciubba J, Pai SI, Califano JA, Tufano RP, Koch WM, 2008, Cystic lymph node metastasis in patients with head and neck cancer: an HPVassociated phenomenon. Head Neck 30(7):898–903. https://doi. org/10.1002/hed.20796 Ang KK, Harris J,Wheeler R,Weber R, Rosenthal DI, Nguyen-Tan PF, Westra WH, Chung CH, Jordan RC, Lu C, Kim H, Axelrod R, Silverman CC, Redmond KP, Gillison ML, 2010, Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 363(1):24–35. , DOI 10.1056/NEJMoa0912217 Gillison ML, Zhang Q, Jordan R, Xiao W, Westra WH, Trotti A, Spencer S, Harris J, Chung CH, Ang KK, 2012, Tobacco smoking and increased risk of death and progression for patients with p16- positive and p16-negative oropharyngeal cancer. J Clin Oncol Off J Am Soc Clin Oncol 30(17):2102–2111. , DOI 10.1200/ JCO.2011.38.4099 Deschler DG, Richmon JD, Khariwala SS, Ferris RL, Wang MB, 2014, The “new” head and neck cancer patient-young, nonsmoker, nondrinker, and HPV positive: evaluation. Otolaryngol Head Neck Surg 151(3):375–380. , DOI 10.1177/0194599814538605 Dansky Ullmann C, Harlan LC, Shavers VL, Stevens JL, 2012, A population-based study of therapy and survival for patients with head and neck cancer treated in the community. Cancer 118(18): 4452–4461. , DOI 10.1002/cncr.27419 Huang SH, Xu W, Waldron J, Siu L, Shen X, Tong L, Ringash J, Bayley A, Kim J, Hope A, Cho J, Giuliani M, Hansen A, Irish J, Gilbert R, Gullane P, Perez-Ordonez B, Weinreb I, Liu FF, O'Sullivan B, 2015, Refining American Joint Committee on Cancer/Union for International Cancer Control TNM stage and prognostic groups for human papillomavirus-related oropharyngeal carcinomas. J Clin Oncol 33(8):836–845. , DOI 10.1200/ JCO.2014.58.6412 Lindel K, Beer KT, Laissue J, Greiner RH, Aebersold DM, 2001, Human papillomavirus positive squamous cell carcinoma of the oropharynx: a radiosensitive subgroup of head and neck carcinoma. Cancer 92(4):805–813 Dahlstrand HM, Dalianis T, 2005, Presence and influence of human papillomaviruses, HPV, in tonsillar cancer. Adv Cancer Res 93:59–89. , DOI 10.1016/S0065-230X(05)93002-9 Shimura E, Hama T, Suda T, Ikegami M, Urashima M, Kojima H, 2017, The presence of HPV DNA in neck lymph node metastasis correlates with improved overall survival of patients with oropharyngeal cancer undergoing surgical treatment. Oncology 92(2):87– 93. , DOI 10.1159/000452420 Weinberger PM, Yu Z, Haffty BG, Kowalski D, Harigopal M, Brandsma J, Sasaki C, Joe J, Camp RL, Rimm DL, Psyrri A, 2006, Molecular classification identifies a subset of human papillomavirus–associated oropharyngeal cancers with favorable prognosis. J Clin Oncol Off J Am Soc Clin Oncol 24(5):736–747. , DOI 10.1200/JCO.2004.00.3335 Begum S, Gillison ML, Ansari-Lari MA, Shah K, Westra WH, 2003, Detection of human papillomavirus in cervical lymph nodes: a highly effective strategy for localizing site of tumor origin. Clin Cancer Res 9(17):6469–6475 Singhi AD, Westra WH, 2010, Comparison of human papillomavirus in situ hybridization and p16 immunohistochemistry in the detection of human papillomavirus-associated head and neck cancer based on a prospective clinical experience. Cancer 116(9): 2166–2173. , DOI 10.1002/cncr.25033 Keswani RN, Noffsinger A, Waxman I, Bissonnette M, 2006, Clinical use of p53 in Barrett’s esophagus. Cancer Epidemiol Biomarkers Prev 15(7):1243–1249. , DOI 10.1158/1055- 9965.EPI-06-0010 Rettori MM, de Carvalho AC, Bomfim Longo AL, de Oliveira CZ, Kowalski LP, Carvalho AL, Vettore AL, 2013, Prognostic significance of TIMP3 hypermethylation in post-treatment salivary rinse from head and neck squamous cell carcinoma patients. Carcinogenesis 34(1):20–27. , DOI 10.1093/carcin/bgs311 Brabender J, Usadel H, Danenberg KD, Metzger R, Schneider PM, Lord RV,Wickramasinghe K, LumCE, Park J, Salonga D, Singer J, Sidransky D, Holscher AH, Meltzer SJ, Danenberg PV, 2001, Adenomatous polyposis coli gene promoter hypermethylation in non-small cell lung cancer is associated with survival. Oncogene 20(27):3528–3532. , DOI 10.1038/sj.onc.1204455 Chaturvedi AK, Anderson WF, Lortet-Tieulent J, Curado MP, Ferlay J, Franceschi S, Rosenberg PS, Bray F, Gillison ML, 2013, Worldwide trends in incidence rates for oral cavity and oropharyngeal cancers. J Clin Oncol Off J Am Soc Clin Oncol 31(36):4550–4559. , DOI 10.1200/JCO.2013.50.3870 Howard J, Masterson L, Dwivedi RC, Riffat F, Benson R, Jefferies S, Jani P, Tysome JR, Nutting C, 2016)Minimally invasive surgery versus radiotherapy/chemoradiotherapy for small-volume primary oropharyngeal carcinoma. Cochrane Database Syst Rev 12: CD010963. , DOI 10.1002/14651858.CD010963.pub2 Parsons JT, Mendenhall WM, Stringer SP, Amdur RJ, Hinerman RW, Villaret DB, Moore-Higgs GJ, Greene BD, Speer TW, Cassisi NJ, Million RR, 2002, Squamous cell carcinoma of the oropharynx: surgery, radiation therapy, or both. Cancer 94(11):2967–2980. , DOI 10.1002/cncr.10567 Blanchard P, Hill C, Guihenneuc-Jouyaux C, Baey C, Bourhis J, Pignon JP, 2011, Mixed treatment comparison meta-analysis of altered fractionated radiotherapy and chemotherapy in head and neck cancer. J Clin Epidemiol 64(9):985–992. , DOI 10. 1016/j.jclinepi.2010.10.016 Lee J, Chang JS, Kwon HJ, Kim SH, Shin SJ, Keum KC, 2016, Impact of p16 expression in oropharyngeal cancer in the postoperative setting: the necessity of re-evaluating traditional risk stratification. Jpn J Clin Oncol 46(10):911–918. , DOI 10.1093/ jjco/hyw099 Dahlstrom KR, Calzada G, Hanby JD, Garden AS, Glisson BS, Li G, Roberts DB, Weber RS, Sturgis EM, 2013, An evolution in demographics, treatment, and outcomes of oropharyngeal cancer at a major cancer center: a staging system in need of repair. Cancer 119(1):81–89. , DOI 10.1002/cncr.27727 D'Souza G, Kreimer AR, Viscidi R, Pawlita M, Fakhry C, Koch WM,WestraWH,GillisonML(2007, Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 356(19): 1944–1956. , DOI 10.1056/NEJMoa065497 O'Sullivan B, Huang SH, Siu LL, Waldron J, Zhao H, Perez- Ordonez B, Weinreb I, Kim J, Ringash J, Bayley A, Dawson LA, HopeA, Cho J, Irish J, Gilbert R, Gullane P, Hui A, Liu FF, Chen E, Xu W, 2013, Deintensification candidate subgroups in human papillomavirus-related oropharyngeal cancer according to minimal risk of distant metastasis. J Clin Oncol Off J Am Soc Clin Oncol 31(5):543–550. , DOI 10.1200/JCO.2012.44.0164 Rischin D,Young RJ, Fisher R, Fox SB, Le QT, Peters LJ, Solomon B, Choi J, O'Sullivan B, Kenny LM, McArthur GA, 2010, Prognostic significance of p16INK4A and human papillomavirus in patients with oropharyngeal cancer treated on TROG02.02 phase III trial. J Clin Oncol Off J Am Soc Clin Oncol 28(27):4142–4148. , DOI 10.1200/JCO.2010.29.2904 Huang SH, Perez-Ordonez B, Weinreb I, Hope A, Massey C, Waldron JN, Kim J, Bayley AJ, Cummings B, Cho BC, Ringash J, Dawson LA, Siu LL, Chen E, Irish J, Gullane P, Hui A, Liu FF, Shen X, Xu W, O'Sullivan B, 2013, Natural course of distant metastases following radiotherapy or chemoradiotherapy in HPVrelated oropharyngeal cancer. Oral Oncol 49(1):79–85. https://doi. org/10.1016/j.oraloncology.2012.07.015 McIlwain WR, Sood AJ, Nguyen SA, Day TA, 2014, Initial symptoms in patients with HPV-positive and HPV-negative oropharyngeal cancer. JAMA Otolaryngol Head Neck Surg 140(5):441–447. , DOI 10.1001/jamaoto.2014.141 Lui VW, HedbergML, Li H, Vangara BS, Pendleton K, Zeng Y, Lu Y, Zhang Q, Du Y, Gilbert BR, Freilino M, Sauerwein S, Peyser ND, Xiao D, Diergaarde B, Wang L, Chiosea S, Seethala R, Johnson JT, Kim S, Duvvuri U, Ferris RL, Romkes M, Nukui T, Kwok-Shing Ng P, Garraway LA, Hammerman PS, Mills GB, Grandis JR, 2013, Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers. Cancer Discov 3(7): 761–769. , DOI 10.1158/2159-8290.CD-13-0103 Stransky N, Egloff AM, Tward AD, Kostic AD, Cibulskis K, Sivachenko A, Kryukov GV, Lawrence MS, Sougnez C, McKenna A, Shefler E, Ramos AH, Stojanov P, Carter SL, Voet D, Cortes ML, Auclair D, Berger MF, Saksena G, Guiducci C, Onofrio RC, Parkin M, Romkes M, Weissfeld JL, Seethala RR, Wang L, Rangel-Escareno C, Fernandez-Lopez JC, Hidalgo- Miranda A, Melendez-Zajgla J, Winckler W, Ardlie K, Gabriel SB, Meyerson M, Lander ES, Getz G, Golub TR, Garraway LA, Grandis JR, 2011, The mutational landscape of head and neck squamous cell carcinoma. Science 333(6046):1157–1160. https:// doi.org/10.1126/science.1208130 Pickering CR, Zhang J, Yoo SY, Bengtsson L, Moorthy S, Neskey DM, Zhao M, Ortega AlvesMV, Chang K, Drummond J, Cortez E, Xie TX, Zhang D, Chung W, Issa JP, Zweidler-McKay PA, Wu X, El-Naggar AK, Weinstein JN, Wang J, Muzny DM, Gibbs RA, Wheeler DA, Myers JN, Frederick MJ, 2013, Integrative genomic characterization of oral squamous cell carcinoma identifies frequent somatic drivers. Cancer Discov 3(7):770–781. , DOI 10. 1158/2159-8290.CD-12-0537 Agrawal N, Frederick MJ, Pickering CR, Bettegowda C, Chang K, Li RJ, Fakhry C, Xie TX, Zhang J, Wang J, Zhang N, El-Naggar AK, Jasser SA, Weinstein JN, Trevino L, Drummond JA, Muzny DM, Wu Y, Wood LD, Hruban RH, Westra WH, Koch WM, Califano JA, Gibbs RA, Sidransky D, Vogelstein B, Velculescu VE, Papadopoulos N, Wheeler DA, Kinzler KW, Myers JN, 2011, Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 333(6046):1154–1157. , DOI 10.1126/science.1206923 Mountzios G, Rampias T, Psyrri A, 2014, The mutational spectrum of squamous-cell carcinoma of the head and neck: targetable genetic events and clinical impact. Ann Oncol 25(10):1889–1900. , DOI 10.1093/annonc/mdu143 Cancer Genome Atlas Network, 2015, Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 517, 7536):576–582. , DOI 10.1038/nature14129 Dakubo GD, Jakupciak JP, Birch-Machin MA, Parr RL, 2007, Clinical implications and utility of field cancerization. Cancer Cell Int 7:2. , DOI 10.1186/1475-2867-7-2 Leemans CR, Braakhuis BJ, Brakenhoff RH, 2011, The molecular biology of head and neck cancer. Nat Rev Cancer 11(1):9–22. , DOI 10.1038/nrc2982 Poeta ML, Manola J, Goldwasser MA, Forastiere A, Benoit N, Califano JA, Ridge JA, Goodwin J, Kenady D, Saunders J, Westra W, Sidransky D, Koch WM, 2007, TP53 mutations and survival in squamous-cell carcinoma of the head and neck. N Engl J Med 357(25):2552–2561. , DOI 10.1056/ NEJMoa073770 van Houten VM, Tabor MP, van den Brekel MW, Kummer JA, Denkers F, Dijkstra J, Leemans R, van der Waal I, Snow GB, Brakenhoff RH, 2002, Mutated p53 as a molecular marker for the diagnosis of head and neck cancer. J Pathol 198(4):476–486. , DOI 10.1002/path.1242 Balz V, Scheckenbach K, Gotte K, Bockmuhl U, Petersen I, Bier H, 2003, Is the p53 inactivation frequency in squamous cell carcinomas of the head and neck underestimated? Analysis of p53 exons 2- 11 and human papillomavirus 16/18 E6 transcripts in 123 unselected tumor specimens. Cancer Res 63(6):1188–1191 Kutler DI, Auerbach AD, Satagopan J, Giampietro PF, Batish SD, Huvos AG, Goberdhan A, Shah JP, Singh B, 2003, High incidence of head and neck squamous cell carcinoma in patients with Fanconi anemia. Arch Otolaryngol Head Neck Surg 129(1):106–112 Hopkins J, Cescon DW, Tse D, Bradbury P, Xu W, Ma C, Wheatley-Price P, Waldron J, Goldstein D, Meyer F, Bairati I, Liu G, 2008, Genetic polymorphisms and head and neck cancer outcomes: a review. Cancer Epidemiol Biomarkers Prev 17(3):490– 499. , DOI 10.1158/1055-9965.EPI-07-2714 Smith RB, Sniezek JC, Weed DT, Wax MK, 2007, Utilization of free tissue transfer in head and neck surgery. Otolaryngol Head Neck Surg 137(2):182–191. , DOI 10.1016/j.otohns.2007. 04.011 Koch WM, Brennan JA, Zahurak M, Goodman SN, Westra WH, Schwab D, Yoo GH, Lee DJ, Forastiere AA, Sidransky D, 1996, p53 mutation and locoregional treatment failure in head and neck squamous cell carcinoma. J Natl Cancer Inst 88(21):1580–1586 Graveland AP, Golusinski PJ, Buijze M, Douma R, Sons N, Kuik DJ, Bloemena E, Leemans CR, Brakenhoff RH, Braakhuis BJ, 2011, Loss of heterozygosity at 9p and p53 immunopositivity in surgical margins predict local relapse in head and neck squamous cell carcinoma. Int J Cancer 128(8):1852–1859. , DOI 10. 1002/ijc.25523 Ying J, Li H, Seng TJ, Langford C, Srivastava G, Tsao SW, Putti T, Murray P, Chan AT, Tao Q, 2006, Functional epigenetics identifies a protocadherin PCDH10 as a candidate tumor suppressor for nasopharyngeal, esophageal and multiple other carcinomas with frequent methylation. Oncogene 25(7):1070–1080. , DOI 10.1038/sj.onc.1209154 Don KR, Ramani P, Ramshankar V, Sherlin HJ, Premkumar P, Natesan A, 2014, Promoter hypermethylation patterns of P16, DAPK and MGMT in oral squamous cell carcinoma: a systematic review and meta-analysis. Indian J Dent Res 25(6):797–805. , DOI 10.4103/0970-9290.152208 Sanchez-Cespedes M, Esteller M, Wu L, Nawroz-Danish H, Yoo GH, Koch WM, Jen J, Herman JG, Sidransky D, 2000, Gene promoter hypermethylation in tumors and serum of head and neck cancer patients. Cancer Res 60(4):892–895 Noorlag R, van Kempen PM, Moelans CB, de Jong R, Blok LE, Koole R, GrolmanW, van Diest PJ, van Es RJ,Willems SM, 2014, Promoter hypermethylation using 24-gene array in early head and neck cancer: better outcome in oral than in oropharyngeal cancer. Epigenetics 9(9):1220–1227. , DOI 10.4161/epi.29785 van Kempen PM, Noorlag R, Braunius WW, Stegeman I, Willems SM, Grolman W, 2014, Differences in methylation profiles between HPV-positive and HPV-negative oropharynx squamous cell carcinoma: a systematic review. Epigenetics 9(2):194–203. https:// doi.org/10.4161/epi.26881 SartorMA, Dolinoy DC, Jones TR, Colacino JA, Prince ME, Carey TE, Rozek LS, 2011, Genome-wide methylation and expression differences in HPV(+, and HPV(−, squamous cell carcinoma cell lines are consistent with divergent mechanisms of carcinogenesis. Epigenetics 6(6):777–787 Choudhury JH, Ghosh SK, 2015, Promoter hypermethylation profiling identifies subtypes of head and neck cancer with distinct viral, environmental, genetic and survival characteristics. PLoS One 10(6):e0129808. , DOI 10.1371/journal.pone.0129808 Kostareli E, Holzinger D, Bogatyrova O, Hielscher T, Wichmann G, Keck M, Lahrmann B, Grabe N, Flechtenmacher C, Schmidt CR, Seiwert T, Dyckhoff G, Dietz A, Hofler D, Pawlita M, Benner A, Bosch FX, Plinkert P, Plass C, Weichenhan D, Hess J, 2013, HPV-related methylation signature predicts survival in oropharyngeal squamous cell carcinomas. J Clin Invest 123(6):2488–2501. , DOI 10.1172/JCI67010 Tokumaru Y, Yamashita K, Osada M, Nomoto S, Sun DI, Xiao Y, Hoque MO, Westra WH, Califano JA, Sidransky D, 2004, Inverse correlation between cyclin A1 hypermethylation and p53 mutation in head and neck cancer identified by reversal of epigenetic silencing. Cancer Res 64(17):5982–5987. , DOI 10.1158/0008- 5472.CAN-04-0993 Perdomo S, Martin Roa G, Brennan P, Forman D, SierraMS, 2016, Head and neck cancer burden and preventive measures in Central and South America. Cancer Epidemiol 44(Suppl 1):S43–S52. , DOI 10.1016/j.canep.2016.03.012 Lopez CR, Antoni MH, Pereira D, Seay J, Whitehead N, Potter J, O'Sullivan M, Fletcher MA, 2013, Stress management, depression and immune status in lower income racial/ethnic minority women co-infected with HIVand HPV. J Appl Biobehav Res 18(1):37–57. , DOI 10.1111/jabr.12003 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1047 EP 1058 DI 10.1007/s12253-018-0462-0 PG 12 ER PT J AU Temraz, S Shamseddine, A Mukherji, D Charafeddine, M Tfayli, A Assi, H Hammoud, SM Makki, I Nassif, S AF Temraz, Sally Shamseddine, Ali Mukherji, Deborah Charafeddine, Maya Tfayli, Arafat Assi, Hazem Hammoud, Salim Miza Makki, Iman Nassif, Samer TI Ki67 and P53 in Relation to Disease Progression in Metastatic Pancreatic Cancer: a Single Institution Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic cancer; Ki67; P53; CA19–9; Prognosis ID Pancreatic cancer; Ki67; P53; CA19–9; Prognosis AB We investigated the expression patterns of Ki67 and p53 in metastatic pancreatic adenocarcinomas and analyzed their relationship with disease progression-free survival (PFS) and overall survival (OS) in the overall study population and in patients treated with a gemcitabine-containing chemotherapy versus FOLFIRINOX chemotherapy. Patients with histologically confirmed stage IV adenocarcinoma of the pancreas treated at AUBMC were included after obtaining institutional review board approval (IRB ID: IM.ST.05). The ROC was plotted to identify the threshold Ki-67, p53 and CA19–9 value for disease progression, the identified value was further used in Kaplan Meier curves to compare PFS for both groups (gemcitabine versus FOLFIRINOX). A value of p < 0.05 was considered significant in all analyses. On univariate analysis, patients who had a Ki- 67 > 12.5% or a p53 > 15% had significantly shorter PFS (p = 0.034 and p = 0.016, respectively). This effect was restricted to Gemcitabine or gemcitabine-combination treated patients. A decrease in CA19–9 levels 6–8 weeks after chemotherapy of >58% had significantly longer PFS (p = 0.027). On multivariate analysis after controlling for grade, age and P53, Ki-67 remained significant, for every one unit increase in Ki-67 the progression risk increases by 1.017 times. Our study highlights the negative impact of high P53 expression and Ki67 proliferation index on PFS in patients with metastatic pancreatic cancer. C1 [Temraz, Sally] American University of Beirut Medical Center, Department of Internal Medicine, Riad El Solh, 110 72020 Beirut, Lebanon. [Shamseddine, Ali] American University of Beirut Medical Center, Department of Internal Medicine, Riad El Solh, 110 72020 Beirut, Lebanon. [Mukherji, Deborah] American University of Beirut Medical Center, Department of Internal Medicine, Riad El Solh, 110 72020 Beirut, Lebanon. [Charafeddine, Maya] American University of Beirut Medical Center, Data Management and Clinical Research Unit, Riad El Solh, 110 72020 Beirut, Lebanon. [Tfayli, Arafat] American University of Beirut Medical Center, Department of Internal Medicine, Riad El Solh, 110 72020 Beirut, Lebanon. [Assi, Hazem] American University of Beirut Medical Center, Department of Internal Medicine, Riad El Solh, 110 72020 Beirut, Lebanon. [Hammoud, Salim Miza] American University of Beirut Medical Center, Department of Internal Medicine, Riad El Solh, 110 72020 Beirut, Lebanon. [Makki, Iman] American University of Beirut Medical Center, Department of Internal Medicine, Riad El Solh, 110 72020 Beirut, Lebanon. [Nassif, Samer] American University of Beirut Medical Center, Department of Pathology and Laboratory Medicine, Riad El Solh, 110 72020 Beirut, Lebanon. RP Temraz, S (reprint author), American University of Beirut Medical Center, Department of Internal Medicine, 110 72020 Beirut, Lebanon. EM st29@aub.edu.lb CR Siegel R, Ma J, Zou Z, Jemal A, 2014, Cancer statistics, 2014. CA Cancer J Clin 64(1):9–29. , DOI 10.3322/caac.21208 Saiki Y, Horii A, 2014, Molecular pathology of pancreatic cancer. Pathol Int 64(1):10–19. , DOI 10.1111/pin.12114 Shimamura A, Fisher DE, 1996, p53 in life and death. Clin Cancer Res 2(3):435–440 Striefler JK, Sinn M, Pelzer U, Juhling A, Wislocka L, Bahra M, Sinn BV, Denkert C, Dorken B, Oettle H, Riess H, Blaker H, Lohneis P, 2016, P53 overexpression and Ki67-index are associated with outcome in ductal pancreatic adenocarcinoma with adjuvant gemcitabine treatment. Pathol Res Pract 212(8): 726–734. , DOI 10.1016/j.prp.2016.06.001 Scholzen T, Gerdes J, 2000, The Ki-67 protein: from the known and the unknown. J Cell Physiol 182(3):311–322. , DOI 10.1002/(SICI)1097-4652(200003)182:3<311::AID-JCP1>3.0. CO;2-9 Jalava P, Kuopio T, Juntti-Patinen L, Kotkansalo T, Kronqvist P, Collan Y, 2006, Ki67 immunohistochemistry: a valuable marker in prognostication but with a risk of misclassification: proliferation subgroups formed based on Ki67 immunoreactivity and standardized mitotic index. Histopathology 48(6):674– 682. , DOI 10.1111/j.1365-2559.2006.02402.x Gentile V, Vicini P, Giacomelli L, Cardillo MR, Pierangeli A, Degener AM, 2006, Detection of human papillomavirus DNA, p53 and ki67 expression in penile carcinomas. Int J Immunopathol Pharmacol 19(1):209–215 Lee CS, 1996, Differences in cell proliferation and prognostic significance of proliferating cell nuclear antigen and Ki-67 antigen immunoreactivity in in situ and invasive carcinomas of the extrahepatic biliary tract. Cancer 78(9):1881–1887 Hruban RH, Iacobuzio-Donahue C,Wilentz RE, Goggins M, Kern SE, 2001, Molecular pathology of pancreatic cancer. Cancer J 7(4): 251–258 Wang B, Matsuoka S, Carpenter PB, Elledge SJ, 2002, 53BP1, a mediator of the DNA damage checkpoint. Science 298(5597):1435–1438. , DOI 10.1126/science. 1076182 Smith RA, Tang J, Tudur-Smith C, Neoptolemos JP, Ghaneh P, 2011, Meta-analysis of immunohistochemical prognostic markers in resected pancreatic cancer. Br J Cancer 104(9):1440–1451. , DOI 10.1038/bjc.2011.110 Bold RJ, Hess KR, Pearson AS, Grau AM, Sinicrope FA, Jennings M, McConkey DJ, Bucana CD, Cleary KR, Hallin PA, Chiao PJ, Abbruzzese JL, Evans DB, 1999, Prognostic factors in resectable pancreatic cancer: p53 and bcl-2. J Gastrointest Surg 3(3):263–277 Dergham ST, Dugan MC, Joshi US, Chen YC, Du W, Smith DW, Arlauskas P, Crissman JD, Vaitkevicius VK, Sarkar FH, 1997, The clinical significance of p21(WAF1/CIP-1, and p53 expression in pancreatic adenocarcinoma. Cancer 80(3):372– 381 Nio Y, Dong M, Uegaki K, Hirahara N, Minari Y, Sasaki S, Takamura M, Iguchi C, Tamura K, 1999, Comparative significance of p53 andWAF/1-p21 expression on the efficacy of adjuvant chemotherapy for resectable invasive ductal carcinoma of the pancreas. Pancreas 18(2):117–126 Nio Y, Dong M, Iguchi C, Yamasawa K, Toga T, Itakura M, Tamura K, 2001, Expression of Bcl-2 and p53 protein in resectable invasive ductal carcinoma of the pancreas: effects on clinical outcome and efficacy of adjuvant chemotherapy. J Surg Oncol 76(3):188–196 Makinen K, Hakala T, Lipponen P, Alhava E, Eskelinen M, 1998, Clinical contribution of bcl-2, p53 and Ki-67 proteins in pancreatic ductal adenocarcinoma. Anticancer Res 18(1B): 615–618 DiGiuseppe JA, Hruban RH, Goodman SN, Polak M, van den Berg FM, Allison DC, Cameron JL, Offerhaus GJ, 1994, Overexpression of p53 protein in adenocarcinoma of the pancreas. Am J Clin Pathol 101(6):684–688 Ahrendt SA, Brown HM, Komorowski RA, Zhu YR, Wilson SD, Erickson BA, Ritch PS, Pitt HA, Demeure MJ, 2000, p21WAF1 expression is associated with improved survival after adjuvant chemoradiation for pancreatic cancer. Surgery 128(4):520– 530. , DOI 10.1067/msy.2000.108052 Vitellius C, Eymerit-Morin C, Luet D, Fizanne L, Foubert F, Bertrais S, Rousselet MC, Caroli-Bosc FX, 2017, Relationship between the expression of O6-Methylguanine-DNA methyltransferase, MGMT, and p53, and the clinical response in metastatic pancreatic adenocarcinoma treated with FOLFIRINOX. Clin Drug Investig 37:669–677. , DOI 10.1007/s40261-017-0522-3 Galmarini CM, Clarke ML, Falette N, Puisieux A, Mackey JR, Dumontet C, 2002, Expression of a non-functional p53 affects the sensitivity of cancer cells to gemcitabine. Int J Cancer 97(4): 439–445 Fiorini C, Cordani M, Padroni C, Blandino G, Di Agostino S, Donadelli M, 2015, Mutant p53 stimulates chemoresistance of pancreatic adenocarcinoma cells to gemcitabine. Biochim Biophys Acta 1853(1):89–100. , DOI 10.1016/j.bbamcr.2014.10.003 Venkatasubbarao K, Peterson L, Zhao S, Hill P, Cao L, Zhou Q, Nawrocki ST, Freeman JW, 2013, Inhibiting signal transducer and activator of transcription-3 increases response to gemcitabine and delays progression of pancreatic cancer. Mol Cancer 12(1):104. , DOI 10.1186/1476-4598-12-104 Ko AH, Hwang J, Venook AP, Abbruzzese JL, Bergsland EK, Tempero MA, 2005, Serum CA19-9 response as a surrogate for clinical outcome in patients receiving fixed-dose rate gemcitabine for advanced pancreatic cancer. Br J Cancer 93(2):195–199. , DOI 10.1038/sj.bjc.6602687 Halm U, Schumann T, Schiefke I, Witzigmann H, Mossner J, Keim V, 2000, Decrease of CA 19-9 during chemotherapy with gemcitabine predicts survival time in patients with advanced pancreatic cancer. Br J Cancer 82(5):1013–1016. , DOI 10.1054/bjoc.1999.1035 Ziske C, Schlie C, Gorschluter M, Glasmacher A, Mey U, Strehl J, Sauerbruch T, Schmidt-Wolf IG, 2003, Prognostic value of CA 19-9 levels in patients with inoperable adenocarcinoma of the pancreas treated with gemcitabine. Br J Cancer 89(8):1413–1417. , DOI 10.1038/sj.bjc.6601263 Saad ED, Machado MC, Wajsbrot D, Abramoff R, Hoff PM, Tabacof J, Katz A, Simon SD, Gansl RC, 2002, Pretreatment CA 19-9 level as a prognostic factor in patients with advanced pancreatic cancer treated with gemcitabine. Int J Gastrointest Cancer 32(1):35–41. , DOI 10.1385/IJGC:32:1:35 Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul J-L, Gourgou-Bourgade S, de la Fouchardiere C, Bennouna J, Bachet J-B, Khemissa-Akouz F, Pere-Verge D, Delbaldo C, Assenat E, Chauffert B, Michel P, Montoto-Grillot C, Ducreux M, 2011, FOLFIRINOX versus gemcitabine for metastatic pancreatic Cancer. N Engl J Med 364(19):1817–1825. , DOI 10.1056/NEJMoa1011923 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1059 EP 1066 DI 10.1007/s12253-018-0464-y PG 8 ER PT J AU Nedjadi, T Salem, N Khayyat, D Al-Sayyad, A Al-Ammari, A Al-Maghrabi, J AF Nedjadi, Taoufik Salem, Nada Khayyat, Dareen Al-Sayyad, Ahmed Al-Ammari, Adel Al-Maghrabi, Jaudah TI Sonic Hedgehog Expression is Associated with Lymph Node Invasion in Urothelial Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; Sonic hedgehog; Expression; Lymph node; Prognosis ID Bladder cancer; Sonic hedgehog; Expression; Lymph node; Prognosis AB Bladder cancer (BC) is a deadly disease characterized by high recurrence rates and frequent progression to an aggressive phenotype. Dysregulation of various signaling pathways have been implicated in BC tumorigenesis, however, the clinical relevance of sonic hedgehog pathway (Shh) remains under investigated. The aim of the current study was to analyze the prognostic value of Shh expression in patients with bladder carcinoma. Immunohistochemical expression of Shh was performed using tissue microarray with 128 specimens from bladder cancer patients. Kaplan-meier survival was analysed and correlation between Shh protein expression and patients’ clinicopathological parameters wasexamined using Fisher’s exact test. The immuno-staining results revealed that Shh protein exhibits cytoplasmic localization and is expressed in 49% of the analyzed bladder cancer cohort. Our data indicated that high Shh expression significantly correlated with increased lymph node metastasis (p = 0.02), however no association was reported between Shh expression and other clinicopatholigical parameters. High expression of sonic hedgehog was associated with lymph node invasion which may indicate that Shh might play an important role in progression and metastasis of bladder cancer. C1 [Nedjadi, Taoufik] Ministry of National Guard-Health Affairs, King Abdulaziz Medical City, King Abdullah International Medical Research CentreJeddah, Saudi Arabia. [Salem, Nada] King Abdulaziz University, Center of Excellence in Genomic Medicine ResearchJeddah, Saudi Arabia. [Khayyat, Dareen] King Abdulaziz University, King Fahd Medical Research CentreJeddah, Saudi Arabia. [Al-Sayyad, Ahmed] King Abdulaziz University, Department of UrologyJeddah, Saudi Arabia. [Al-Ammari, Adel] King Faisal Specialist Hospital & Research Center, Department of UrologyJeddah, Saudi Arabia. [Al-Maghrabi, Jaudah] King Abdulaziz University, Department of PathologyJeddah, Saudi Arabia. RP Nedjadi, T (reprint author), Ministry of National Guard-Health Affairs, King Abdulaziz Medical City, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia. EM nedjadita@ngha.med.sa CR Antoni S, Ferlay J, Soerjomataram I, Znaor A, Jemal A, Bray F, 2017, Bladder Cancer incidence and mortality: a global overview and recent trends. Eur Urol 71(1):96–108 Sanli O, Dobruch J, Knowles MA, Burger M, Alemozaffar M, Nielsen ME, Lotan Y, 2017, Bladder cancer. Nat Rev Dis Primers 3:17022 Morgan TM, Clark PE, 2010, Bladder cancer. Curr Opin Oncol 22(3):242–249 Kaufman DS, ShipleyWU, Feldman AS, 2009;18, Bladder cancer. Lancet 374(9685):239–249 Millan-Rodriguez F, Chechile-Toniolo G, Salvador-Bayarri J, Palou J, Algaba F, Vicente-Rodriguez J, 2000, Primary superficial bladder cancer risk groups according to progression, mortality and recurrence. J Urol 164(3 Pt 1):680–684 Cheung G, Sahai A, Billia M, Dasgupta P, KhanMS, 2013, Recent advances in the diagnosis and treatment of bladder cancer. BMC Med 11:13 Netto GJ, Tafe LJ, 2016, Emerging bladder Cancer biomarkers and targets of therapy. Urol Clin North Am 43(1):63–76 Cheng L, Davison DD, Adams J, Lopez-Beltran A, Wang L, Montironi R, Zhang S, 2014, Biomarkers in bladder cancer: translational and clinical implications. Crit Rev Oncol Hematol 89(1): 73–111 Mbeutcha A, Lucca I, Mathieu R, Lotan Y, Shariat SF, 2016, Current status of urinary biomarkers for detection and surveillance of bladder Cancer. Urol Clin North Am 43(1):47–62 Ehlen HW, Buelens LA,Vortkamp A, 2006, Hedgehog signaling in skeletal development. Birth Defects Res C Embryo Today 78(3): 267–279 Song J, Zhang J, Wang J, Guo X, Dong W, 2015, β1 integrin mediates colorectal cancer cell proliferation and migration through regulation of the hedgehog pathway. Tumour Biol 36(3):2013–2021 Kanaya K, Ii M, Okazaki T et al, 2015, Sonic hedgehog signaling regulates vascular differentiation and function in human CD34 positive cells: vasculogenic CD34(+, cells with sonic hedgehog. Stem Cell Res 14(2):165–176 Velcheti V, Govindan R, 2007, Hedgehog signaling pathway and lung cancer. J Thorac Oncol 2(1):7–10 Statkiewicz M, Maryan N, Lipiec A, Grecka E, Grygorowicz MA, Omiotek M, Gorska A, Mikula M, Malecki M, 2014, The role of the SHH gene in prostate cancer cell resistance to paclitaxel. Prostate 74(11):1142–1152 Tao Y, Mao J, Zhang Q, Li L, 2011, Overexpression of hedgehog signaling molecules and its involvement in triple-negative breast cancer. Oncol Lett 2(5):995–1001 Yoshimoto AN, Bernardazzi C, Carneiro AJ et al, 2012, Hedgehog pathway signaling regulates human colon carcinoma HT-29 epithelial cell line apoptosis and cytokine secretion. PLoS One 7(9): e45332 Liao X, Siu MK, Au CWet al, 2009, Aberrant activation of hedgehog signaling pathway in ovarian cancers: effect on prognosis, cell invasion and differentiation. Carcinogenesis 30(1):131–140 Marechal R, Bachet JB, Calomme A et al, 2015, Sonic hedgehog and Gli1 expression predict outcome in resected pancreatic adenocarcinoma. Clin Cancer Res 21(5):1215–1224 Giakoustidis A, Giakoustidis D, Mudan S, Sklavos A, Williams R, 2015, Molecular signalling in hepatocellular carcinoma: role of and crosstalk among WNT/ß-catenin, sonic hedgehog, notch and Dickkopf-1. Can J Gastroenterol Hepatol 29(4):209–217 Mullor JL, Dahmane N, Sun T, Ruiz i Altaba A, 2001, Wnt signals are targets and mediators of Gli function. Curr Biol 11(10):769–773 Bosco-Clement G, Zhang F, Chen Z, Zhou HM, Li H, Mikami I, Hirata T, Yagui-Beltran A, Lui N, Do HT, Cheng T, Tseng HH, Choi H, Fang LT, Kim IJ, Yue D, Wang C, Zheng Q, Fujii N, Mann M, Jablons DM, He B, 2014, Targeting Gli transcription activation by small molecule suppresses tumor growth. Oncogene 33(16):2087–2097 Shevde LA, Samant RS, 2014, Nonclassical hedgehog-GLI signaling and its clinical implications. Int J Cancer 135(1):1–6 Thievessen I, Wolter M, Prior A, Seifert HH, Schulz WA, 2005, Hedgehog signaling in normal urothelial cells and in urothelial carcinoma cell lines. J Cell Physiol 203(2):372–377 Fei DL, Sanchez-Mejias A, Wang Z, Flaveny C, Long J, Singh S, Rodriguez-Blanco J, Tokhunts R, Giambelli C, Briegel KJ, Schulz WA, Gandolfi AJ, Karagas M, Zimmers TA, Jorda M, Bejarano P, Capobianco AJ, Robbins DJ, 2012, Hedgehog signaling regulates bladder cancer growth and tumorigenicity. Cancer Res 72(17): 4449–4458 Sverrisson EF, Zens MS, Fei DL et al, 2014, Clinicopathological correlates of Gli1 expression in a population-based cohort of patients with newly diagnosed bladder cancer. Urol Oncol 32(5):539–545 Islam SS, Mokhtari RB, Noman AS, Uddin M, RahmanMZ, Azadi MA, Zlotta A, van der Kwast T, Yeger H, Farhat WA, 2016, Sonic hedgehog, Shh, signaling promotes tumorigenicity and stemness via activation of epithelial-to-mesenchymal transition, EMT, in bladder cancer. Mol Carcinog 55:537–551 Shin K, Lim A, Odegaard JI, Honeycutt JD, Kawano S, HsiehMH, Beachy PA, 2014, Cellular origin of bladder neoplasia and tissue dynamics of its progression to invasive carcinoma. Nat Cell Biol 16(5):469–478 Kelleher FC, 2011, Hedgehog signaling and therapeutics in pancreatic cancer. Carcinogenesis 32(4):445–451 DiMagno L, Coni S, Di Marcotullio L, Canettieri G, 2015, Digging a hole under hedgehog: downstream inhibition as an emerging anticancer strategy. Biochim Biophys Acta 1856(1):62–72 ChenM, HildebrandtMA, Clague J et al, 2010, Genetic variations in the sonic hedgehog pathway affect clinical outcomes in nonmuscle- invasive bladder cancer. Cancer Prev Res, Phila, 3(10): 1235–1245 Shin K, Lee J, Guo N, Kim J, Lim A, Qu L, Mysorekar IU, Beachy PA, 2011, Hedgehog/Wnt feedback supports regenerative proliferation of epithelial stem cells in bladder. Nature 472(7341):110–114 Buhmeida A, Dallol A, Merdad A, al-Maghrabi J, GariMA, Abu- ElmagdMM, Chaudhary AG, AbuzenadahAM, Nedjadi T, Ermiah E, al-Thubaity F, al-Qahtani MH, 2014, High fibroblast growth factor 19, FGF19, expression predicts worse prognosis in invasive ductal carcinoma of breast. Tumour Biol 35(3):2817–2824 Ingham PW, McMahon AP, 2001, Hedgehog signaling in animal development: paradigms and principles. Genes Dev 15(23):3059– 3087 DeSouza KR, Saha M, Carpenter AR, Scott M, McHugh KM, 2013, Analysis of the sonic hedgehog signaling pathway in normal and abnormal bladder development. PLoS One 8(1):e53675 Gupta S, Takebe N, Lorusso P, 2010, Targeting the hedgehog pathway in cancer. Ther Adv Med Oncol 2(4):237–250 Aboulkassim TO, LaRue H, Lemieux P, Rousseau F, Fradet Y, 2003, Alteration of the PATCHED locus in superficial bladder cancer. Oncogene 22(19):2967–2971 Pignot G, Vieillefond A, Vacher S, ZerbibM, Debre B, Lidereau R, Amsellem-Ouazana D, Bieche I, 2012, Hedgehog pathway activation in human transitional cell carcinoma of the bladder. Br J Cancer 106(6):1177–1186 He HC, Chen JH, Chen XB, Qin GQ, Cai C, Liang YX, Han ZD, Dai QS, Chen YR, Zeng GH, Zhu JG, Jiang FN, ZhongWD(2012, Expression of hedgehog pathway components is associated with bladder cancer progression and clinical outcome. Pathol Oncol Res 18(2):349–355 van der Horst G, Bos L, van der Pluijm G, 2012, Epithelial plasticity, cancer stem cells, and the tumor-supportive stroma in bladder carcinoma. Mol CancerRes 10(8):995–1009 Li C, Du Y, Yang Z, et al., 2015, GALNT1-mediated glycosylation and activation of sonic hedgehog signaling maintains the selfrenewal and tumor-initiating capacity of bladder Cancer stem cells. Cancer Res Syed IS, Pedram A, Farhat WA, 2016, Role of sonic hedgehog, Shh, signaling in bladder Cancer Stemness and tumorigenesis. Curr Urol Rep 17(2):11 Wang D, Kong X, Li Y, Qian W, Ma J, Wang D, Yu D, Zhong C, 2017, Curcumin inhibits bladder cancer stem cells by suppressing sonic hedgehog pathway. Biochem Biophys Res Commun 493(1): 521–527 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1067 EP 1073 DI 10.1007/s12253-018-0477-6 PG 7 ER PT J AU Han, RL Wang, J Zhang, FJ Zhao, N Gao, BL AF Han, Ruo-Ling Wang, Jin Zhang, Feng-Juan Zhao, Na Gao, Bu-Lang TI Ultrasound Risk Assessment Combined with Molecular Markers of Galectin-3, c-MET, HBME-1 and CK19 for Diagnosis of Malignant and Benign Thyroid Nodules SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ultrasound; Malignancy risk; Thyroid nodules; Molecular marker; Diagnosis ID Ultrasound; Malignancy risk; Thyroid nodules; Molecular marker; Diagnosis AB To investigate the effect of ultrasound combined with expression of Galectin-3, c-Met, HBME-1 and CK19 in differentiating malignant from benign thyroid nodules. Forty-six patients with thyroid nodules were studied with ultrasound and immunohistochemical staining of excised thyroid nodules. The data were classified and compared. The immunohistochemical staining revealed 8 benign and 41 malignant thyroid lesions. In ultrasound risk assessment, the malignancy risk was low in four nodules, medium in five and high in 37 with lymphatic metastasis in 26. A significant (P < 0.05) association existed in the expression of Galectin-3 with nodule boundary and lymphatic metastasis, in HBME-1 with nodule micro-calcification and in c-Met with nodule micro-calcification and lymphatic metastasis. CK19 expression was not significantly (P > 0.05) associated with any of ultrasound features of nodule. Galectin-3, c-Met, HBME-1 and CK19 were significantly (P < 0.05) different in malignant and benign thyroid lesions, with a significant (P < 0.01) tendency in all the molecular markers in predicting the malignant from benign lesions. The ultrasound characteristics could significantly (P < 0.001) predict malignant nodules with a significant (P < 0.05) prediction tendency. The scores of Galectin-3, c-Met and CK19 significantly (P < 0.05) increased with increase of ultrasound malignancy risk degree. In malignant and benign lesions differentiated by ultrasound, no significant (P > 0.05) difference existed in HBME-1 expression, however, with ultrasound malignancy risk increase, the score of HBME-1 expression increased significantly (P = 0.03). Galectin-3, c-Met, HBME-1 and CK19 have significantly greater expressions in thyroid malignant than benign lesions and their expression increases with increase of ultrasound malignancy risk. The combination of both ultrasound and molecular markers can be used to differentiate malignant and benign thyroid lesions. C1 [Han, Ruo-Ling] The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Department of Ultrasound, 12 Jiankang Road, 050011 Shijiazhuang, Hebei Province, China. [Wang, Jin] The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Department of Ultrasound, 12 Jiankang Road, 050011 Shijiazhuang, Hebei Province, China. [Zhang, Feng-Juan] The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Department of Ultrasound, 12 Jiankang Road, 050011 Shijiazhuang, Hebei Province, China. [Zhao, Na] The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Department of Ultrasound, 12 Jiankang Road, 050011 Shijiazhuang, Hebei Province, China. [Gao, Bu-Lang] Henan Provincial People’s Hospital, Department of Interventional TherapyZhengzhou, Henan Province, China. RP Han, RL (reprint author), The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Department of Ultrasound, 050011 Shijiazhuang, China. EM hrl-63523@163.com CR Chi J, Walia E, Babyn P, Wang J, Groot G, Eramian M, 2017, Thyroid nodule classification in ultrasound images by fine-tuning deep convolutional neural network. J Digit Imaging 30:477–486 Kwak JY, HanKH, Yoon JH, MoonHJ, Son EJ, Park SH, Jung HK, Choi JS, Kim BM, Kim EK, 2011, Thyroid imaging reporting and data system for us features of nodules: a step in establishing better stratification of cancer risk. Radiology 260:892–899 Lingam RK, Qarib MH, Tolley NS, 2013, Evaluating thyroid nodules: predicting and selecting malignant nodules for fine-needle aspiration, fna, cytology. Insights Imaging 4:617–624 Koo BS, Kim JM, Seo ST, Yoon YH, Kwon KR, Kim SH, Kwon HW, Bae WJ, Lim YC, 2014, Upregulation of hgf and c-met is associated with subclinical central lymph node metastasis in papillary thyroid microcarcinoma. Ann Surg Oncol 21:2310–2317 Park YJ, Kwak SH, Kim DC, Kim H, Choe G, Park DJ, Jang HC, Park SH, Cho BY, Park SY, 2007, Diagnostic value of galectin-3, hbme-1, cytokeratin 19, high molecular weight cytokeratin, cyclin d1 and p27(kip1, in the differential diagnosis of thyroid nodules. J Korean Med Sci 22:621–628 Gharib H, Papini E, Paschke R, Duick DS, Valcavi R, Hegedus L, Vitti P, AACE/AME/ETA Task Force on Thyroid Nodules, 2010, American association of clinical endocrinologists, associazione medici endocrinologi, and european thyroid association medical guidelines for clinical practice for the diagnosis and management of thyroid nodules: Executive summary of recommendations. Endocrine practice 16(3):468–475 Reading CC, Charboneau JW, Hay ID, Sebo TJ, 2005, Sonography of thyroid nodules: a "classic pattern" diagnostic approach. Ultrasound Q 21:157–165 Kim EK, Park CS, Chung WY, Oh KK, Kim DI, Lee JT, Yoo HS, 2002, New sonographic criteria for recommending fine-needle aspiration biopsy of nonpalpable solid nodules of the thyroid. AJR Am J Roentgenol 178:687–691 Moon WJ, Jung SL, Lee JH,Na DG, Baek JH, Lee YH, KimJ,Kim HS, Byun JS, Lee DH, Thyroid Study Group, Korean Society of Neuro- and Head and Neck Radiology, 2008, Benign and malignant thyroid nodules: us differentiation–multicenter retrospective study. Radiology 247:762–770 Hong YJ, Son EJ, Kim EK, Kwak JY, Hong SW, Chang HS, 2010, Positive predictive values of sonographic features of solid thyroid nodule. Clin Imaging 34:127–133 Rago T, Vitti P, Chiovato L,Mazzeo S, De Liperi A,Miccoli P et al, 1998, Role of conventional ultrasonography and color flowdoppler sonography in predicting malignancy in 'cold' thyroid nodules. Eur J Endocrinol 138:41–46 Sohn YM, Kwak JY, Kim EK, Moon HJ, Kim SJ, Kim MJ, 2010, Diagnostic approach for evaluation of lymph node metastasis from thyroid cancer using ultrasound and fine-needle aspiration biopsy. AJR Am J Roentgenol 194:38–43 Krzeslak A, Lipinska A, 2004, Galectin-3 as a multifunctional protein. Cell Mol Biol Lett 9:305–328 deMatos PS, Ferreira AP, de Oliveira FF, Assumpcao LV,Metze K, Ward LS, 2005, Usefulness of hbme-1, cytokeratin 19 and galectin- 3 immunostaining in the diagnosis of thyroid malignancy. Histopathology 47:391–401 Prasad ML, Pellegata NS, Huang Y, Nagaraja HN, de la Chapelle A, Kloos RT, 2005, Galectin-3, fibronectin-1, cited-1, hbme1 and cytokeratin-19 immunohistochemistry is useful for the differential diagnosis of thyroid tumors. Modern pathology 18(1):48–57 Cheung CC, Ezzat S, Freeman JL, Rosen IB, Asa SL, 2001, Immunohistochemical diagnosis of papillary thyroid carcinoma. Modern pathology 14(4):338–342 Mase T, Funahashi H, Koshikawa T, Imai T, Nara Y, Tanaka Yet al, 2003, Hbme-1 immunostaining in thyroid tumors especially in follicular neoplasm. Endocr J 50:173–177 Sheibani K, Esteban JM, Bailey A, Battifora H, Weiss LM, 1992, Immunopathologic and molecular studies as an aid to the diagnosis of malignant mesothelioma. Hum Pathol 23:107–116 Sahoo S, Hoda SA, Rosai J, DeLellis RA, 2001, Cytokeratin 19 immunoreactivity in the diagnosis of papillary thyroid carcinoma: a note of caution. Am J Clin Pathol 116:696–702 MiettinenM, Kovatich AJ, Karkkainen P, 1997, Keratin subsets in papillary and follicular thyroid lesions. A paraffin section analysis with diagnostic implications. Virchows Archiv 431:407–413 Raphael SJ, McKeown-Eyssen G, Asa SL, 1994, High-molecularweight cytokeratin and cytokeratin-19 in the diagnosis of thyroid tumors. Modern pathology 7(3):295–300 Bottaro DP, Rubin JS, Faletto DL, Chan AM, Kmiecik TE, Vande Woude GF, Aaronson S, 1991, Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science 251:802–804 Ruggeri RM, Vitarelli E, Barresi G, Trimarchi F, Benvenga S, Trovato M, 2012, Hgf/c-met system pathways in benign and malignant histotypes of thyroid nodules: an immunohistochemical characterization. Histol Histopathol 27:113–121 Gentile A, Trusolino L, Comoglio PM, 2008, The met tyrosine kinase receptor in development and cancer. Cancer Metastasis Rev 27:85–94 Alexander EK, Kennedy GC, Baloch ZW, Cibas ES, Chudova D, Diggans J, Friedman L, Kloos RT, LiVolsi VA, Mandel SJ, Raab SS, Rosai J, Steward DL,Walsh PS,Wilde JI, ZeigerMA, Lanman RB, Haugen BR, 2012, Preoperative diagnosis of benign thyroid nodules with indeterminate cytology. N Engl J Med 367:705–715 Trimboli P, Guidobaldi L, Amendola S, Nasrollah N, Romanelli F, Attanasio D, Ramacciato G, Saggiorato E, Valabrega S, Crescenzi A, 2016, Galectin-3 and hbme-1 improve the accuracy of core biopsy in indeterminate thyroid nodules. Endocrine 52:39–45 DA Bhatia P, Friedlander P, Aslam R, Kandil E, 2015, Diagnostic potential of ancillary molecular testing in differentiation of benign and malignant thyroid nodules. Antiucancer Res 35:1237–1241 Cohen YRE, Clark DP, Zeiger MA, Umbricht CB, Tufano RP, Sidransky D,WestraWH, 2004)Mutational analysis of braf in fine needle aspiration biopsies of the thyroid: a potential application for the preoperative assessment of thyroid nodules. Clin Cancer Res 10:2761–2765 Esapa CTJS, Kendall-Taylor P, Lennard TW, Harris PE, 1999, Prevalence of ras mutations in thyroid neoplasia. Clin Endocrinol 50:529–535 Nikiforova MNLR, Biddinger PW, Alexander EK, Dorn GW 2nd, Tallini G, Kroll TG, Nikiforov YE, 2003, Ras point mutations and pax8-ppar gamma rearrangement in thyroid tumors: evidence for distinct molecular pathways in thyroid follicular carcinoma. J Clin Endocrinol Metab 88:2318–2326 Min HSLC, Jung KC, 2013, Correlation of immunohistochemical markers and braf mutation status with histological variants of papillary thyroid carcinoma in the korean population. J KoreanMed Sci 28:534–541 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1075 EP 1081 DI 10.1007/s12253-018-0485-6 PG 7 ER PT J AU Miyazawa, H Kato, K Kobayashi, Y Hirai, M Kimura, I Kitahara, H Noguchi, N Nakamura, H Kawashiri, Sh AF Miyazawa, Hiroki Kato, Koroku Kobayashi, Yutaka Hirai, Mariko Kimura, Iyo Kitahara, Hiroko Noguchi, Natsuyo Nakamura, Hiroyuki Kawashiri, Shuichi TI Clinicopathological Significance of the ET Axis in Human Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endothelin; ET-1; ETAR; ET-axis; Oral squamous cell carcinoma; Prognosis ID Endothelin; ET-1; ETAR; ET-axis; Oral squamous cell carcinoma; Prognosis AB The interaction between cancer cells and the surrounding microenvironment in malignant tumor tissue is known to be closely associated with cancer cell invasion and proliferation. Endothelin (ET) present in the microenvironment surrounding tumors has been reported to play a role in cancer cell invasion and proliferation by binding to receptors on the cell membrane of cancer cells. Here, we immunohistologically detected the expression of ET-1 and its receptor ETAR in oral squamous cell carcinoma (OSCC) and evaluated the association between the expression of each as well as their co-expression (ET-axis expression) and clinicopathological factors. A significant difference was observed between the invasion pattern as a parameter of cancer cellmalignancy and the expressions of ET-1 and ETAR. The survival rates were significantly lower among the patients who were strongly positive for ET-1 and the ETAR-positive patients compared to negative patients. There was also a significant difference between ET-axis expression and the degree of histological differentiation and mode of invasion, and the survival rate of the positive cases was significantly lower than that of the negative cases. Our findings suggested that ET-axis assessments are important for assessing the malignancy of cancer cells and predicting the prognoses of OSCC patients. C1 [Miyazawa, Hiroki] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Kato, Koroku] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Kobayashi, Yutaka] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Hirai, Mariko] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Kimura, Iyo] Nanto Municipal Hospital, Department of Oral and Maxillofacial Surgery, 938 Inami, 932-0211 Nanto, Japan. [Kitahara, Hiroko] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Noguchi, Natsuyo] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Nakamura, Hiroyuki] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Kawashiri, Shuichi] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. RP Kato, K (reprint author), Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 920-8641 Kanazawa, Japan. EM k-koroku@oral.m.kanazawa-u.ac.jp CR Nagao M, Tsugane S, 2016, Cancer in Japan: prevalence, prevention and the role of heterocyclic amines in human carcinogenesis. Genes Environ 38:16 Karnoub AE, Dash AB, Vo AP, Sullivan A, BrooksMW, Bell GW, Richardson AL, Polyak K, Tubo R, Weinberg RA, 2007, Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 449:557–563 Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL,Weinberg RA, 2005, Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through SDF-1/CXCL12 serection. Cell 121:335–348 Orimo A, Weinberg RA, 2006, Stromal fibroblasts in Cancer: a novel tumor-promoting cell type. Cell Cycle 5:1597–1601 De Wever O, Mareel M, 2003, Role of tissue stroma in cancer cell invasion. J Pathol 200:429–447 Salani D, Taraboletti G, Rosano L,Di Castro L, Borsotti P, Giavazzi R, Bagnato A, 2000, Endothelin-1 induces an Angiogenic phenotype in cultured endothelial cells and stimulates neovascularization in vivo. Am J Pathol 157:1703–1711 Grant K, Loizidou M, Taylor I, 2003, Endothelin-1: a multifunctional molecule in cancer. Br J Cancer 88:163–166 Wulfing P, Kersting C, Tio J, Fischer RJ, Wulfing C, Poremba C, Diallo R, Bocker W, Kiesel L, 2004, Endothelin-1, endothelin-A-, and endothelin-B- receptor expression is correlated with vascular endothelial growth factor expression and angiogenesis in breast Cancer. Clin Cancer Res 10:2393–2400 Kiowski W, Kim J, Oechslin E, Sutsch G, Hunziker P, Muller P, Bertel O, Schmitt R, Jones R, 1995, Evidence for endothelin-1- mediated vasoconstriction in severe chronic heart failure. Lancet 346:732–736 Ohkita M, Takaoka M, Matsumura Y, 2007, Endothelin-1 production and its involvement in cardiovascular diseases. Yakugaku Zasshi 127:1319–1329 Nelson JB, Chan-Tack K, Hedican SP, Magnuson SR, Opgenorth TJ, Steven Bova G, Simons JW, 1996, Endothelin-1 production and decreased endothelin B receptor expression in advanced prostate Cancer. Cancer Res 56:663–668 Gohji K, Kitazawa S, Tamada H, Katsuoka Y, Nakajima M, 2001, Expression of endothelin receptor A associated with prostate cancer progression. J Urol 165:1033–1036 Rosano L, Spinella F, Bagnato A, 2013, Endothelin 1 in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer 13:637–651 Nelson J, Bagnato A, Battistini B, Nisen P, 2003, The endothelin axis: emerging role in cancer. Nat Rev Cancer 3:110–116 Nelson JB, Hedican SP, George DJ, Reddi AH, Piantadosi S, Eisenberger MA, Simons JW, 1995, Identification of endothelin- 1 in the pathophysiology of metastatic adenocarcinoma of the prostate. Nat Med 1:944–949 Bagnato A, Tecce R, Moretti C, Di Castro V, Spergel D, Catt KJ, 1995, Autocrine actions of endothelin-1 as a growth factor in human ovarian carcinoma cells. Clin Cancer Res 1:1059–1066 (2009, International Union against cancer. In: Sobin LH, Gospodrowicz MK, Wittekind CH, eds, TNM classification of malignant tumours, 7th edn. Wiley-Liss, New York Yamamoto E, Kohama G, Sunagawa H, Iwai M, Hiratsuka H, 1983, Mode of invasion, bleomycin sensitivity, and clinical course in squamous cell carcinoma of the oral cavity. Cancer 51:2175–2180 Kato K, Kawashiri S, Tanaka A, Noguchi N, Nakaya H, Hase T, Yamamoto E, 2008, Predictive value of measuring p53 labeling index at the invasive front of oral squamous cell carcinomas. Pathol Oncol Res 14:57–61 Inoue A,YanagisawaM, Kimura S,KasuyaY, Miyauchi T, Goto K, Masaki T, 1989, The human endothelin family; three structurally and pharmacologically distinct isopeptides predicted by three separate genes. PNAS 86:2863–2867 Zhang ZY, Chen LL, Xu W, Sigdel K, Jiang XT, 2017, Effects of silencing endothelin-1 on invasion and vascular formation in lung cancer. Oncol Lett 13:4390–4396 Cong N, Li Z, ShaoW, Li J, Yu S, 2016, Activation of ETA receptor by Endothelin-1 induces hepatocellular carcinoma cell migration and invasion via ERK1/2 and AKT signaling pathways. J Membr Biol 249:119–128 Wulfing P, Diallo R, Kersting C,Wulfing C, Poremba C, Greb RR, Bocker W, Kiesel L, 2004, Endothelin-1, endothelin-A- and endothelin-B-receptor expression in preinvasive and invasive breast disease. Oncol Rep 11:791–796 McKenzie GA, Hinsley EE, Hunter K, Lambert DW, 2014, The endothelin axis in head and neck cancer: a promising therapeutic opportunity? J Oral Pathol Med 43:395–404 Hoffmann RR, Yurgel LS, Campos MM, 2010, Endothelins and their receptors as biological markers for oral cancer. Oral Oncol 46: 644–647 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1083 EP 1089 DI 10.1007/s12253-018-0514-5 PG 7 ER PT J AU Liu, Y Guan, J Chen, X AF Liu, Yang Guan, Jianzhong Chen, Xiaotian TI Identification of Differentially Expressed Genes under the Regulation of Transcription Factors in Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; Microarray dataset; Differentialy expressed genes; Transcription factors ID Osteosarcoma; Microarray dataset; Differentialy expressed genes; Transcription factors AB The present study was to investigate and identify the differentially expressed genes (DEGs) in the transcriptional regulatory network of osteosarcoma (OS). The gene expression dataset from Gene Expression Omnibus (GEO) datasets was downloaded. DEGs were identified and their functional annotation was also conducted. In addition, differentially expressed transcription factors (TFs) and the regulatory genes were identified. The electronic validation was used to verify the expression of selected genes. The integrated analysis led to 932 DEGs. The results of functional annotation indicated that these DEGs significantly enriched in the p53 signaling pathway, Jak-STAT signaling pathway and Wnt signaling pathway. ZNF354C, NFIC, NFATC2, SP2, FOXO3, EGR1, ZEB1, RREB1, EGR2 and SRF were covered by most TFs. The expression levels of NFIC and EGR2 in electronic validation were compatible with our bio-informatics result. In conclusion, the deregulation of these genes may provide valuable information in understanding the underlying molecular mechanism in the OS. C1 [Liu, Yang] The First Affiliated Hospital of Bengbu Medical College, Department of Orthopaedics, No. 287, ChangHuai Road, 233004 Bengbu, Anhui Province, China. [Guan, Jianzhong] The First Affiliated Hospital of Bengbu Medical College, Department of Orthopaedics, No. 287, ChangHuai Road, 233004 Bengbu, Anhui Province, China. [Chen, Xiaotian] The First Affiliated Hospital of Bengbu Medical College, Department of Orthopaedics, No. 287, ChangHuai Road, 233004 Bengbu, Anhui Province, China. RP Guan, J (reprint author), The First Affiliated Hospital of Bengbu Medical College, Department of Orthopaedics, 233004 Bengbu, China. EM guanjianzhongah@163.com CR Koyani CN, Kitz K, Rossmann C, Bernhart E, Huber E, Trummer C, Windischhofer W, Sattler W, Malle E, 2016, Activation of the MAPK/Akt/Nrf2-Egr1/HO-1-GCLc axis protects MG-63 osteosarcoma cells against 15d-PGJ2-mediated cell death. Biochem Pharmacol 104:29–41. , DOI 10.1016/j.bcp.2016.01.011 Mirabello L, Troisi RJ, Savage SA, 2009, Osteosarcoma incidence and survival rates from 1973 to 2004: data from the surveillance, epidemiology, and end results program. Cancer 115(7):1531–1543. , DOI 10.1002/cncr.24121 Ottaviani G, Jaffe N, 2009, The epidemiology of osteosarcoma. Cancer Treat Res 152:3–13. , DOI 10.1007/978-1-4419- 0284-9_1 Longhi A, Errani C, De Paolis M, Mercuri M, Bacci G, 2006, Primary bone osteosarcoma in the pediatric age: state of the art. Cancer Treat Rev 32(6):423–436. , DOI 10.1016/j.ctrv. 2006.05.005 Gorlick R, 2009, Current concepts on the molecular biology of osteosarcoma. Cancer Treat Res 152:467–478. , DOI 10. 1007/978-1-4419-0284-9_27 Mirabello L, Koster R,Moriarity BS, Spector LG, Meltzer PS,Gary J,MachielaMJ, Pankratz N, PanagiotouOA, Largaespada D,Wang Z, Gastier-Foster JM, Gorlick R, Khanna C, de Toledo SR, Petrilli AS, Patino-Garcia A, Sierrasesumaga L, Lecanda F, Andrulis IL, Wunder JS, Gokgoz N, Serra M, Hattinger C, Picci P, Scotlandi K, Flanagan AM, Tirabosco R, Amary MF, Halai D, Ballinger ML, Thomas DM, Davis S, Barkauskas DA, Marina N, Helman L, Otto GM, Becklin KL, Wolf NK, Weg MT, Tucker M, Wacholder S, Fraumeni JF Jr, Caporaso NE, Boland JF, Hicks BD, Vogt A, Burdett L, Yeager M, Hoover RN, Chanock SJ, Savage SA, 2015, A genome-wide scan identifies variants in NFIB associated with metastasis in patients with osteosarcoma. Cancer Discov 5(9): 920–931. , DOI 10.1158/2159-8290.cd-15-0125 Johnson DS, Mortazavi A, Myers RM, Wold B, 2007, Genomewide mapping of in vivo protein-DNA interactions. Science, New York, NY, 316(5830):1497–1502. , DOI 10.1126/science. 1141319 Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, Yefanov A, Lee H, Zhang N, Robertson CL, Serova N, Davis S, Soboleva A, 2013, NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res 41(Database issue): D991–D995. , DOI 10.1093/nar/gks1193 Knuppel R, Dietze P, Lehnberg W, Frech K, Wingender E, 1994, TRANSFAC retrieval program: a network model database of eukaryotic transcription regulating sequences and proteins. J Comput Biol 1(3):191–198 Gibbs CP Jr, Weber K, Scarborough MT, 2002, Malignant bone tumors. Instr Course Lect 51:413–428 Scotlandi K, Picci P, Kovar H, 2009, Targeted therapies in bone sarcomas. Curr Cancer Drug Targets 9(7):843–853 Ragland BD, Bell WC, Lopez RR, Siegal GP, 2002, Cytogenetics and molecular biology of osteosarcoma. Lab Investig 82(4):365– 373 Clark JC, Dass CR, Choong PF, 2008, A review of clinical and molecular prognostic factors in osteosarcoma. J Cancer Res Clin Oncol 134(3):281–297. , DOI 10.1007/s00432-007-0330- x Meng F, Zhu L, Huang W, Irwin DM, Zhang S, 2016, Bats: body mass index, forearm mass index, blood glucose levels and SLC2A2 genes for diabetes. Sci Rep 6:29960. , DOI 10.1038/ srep29960 Fei Q, Lin J, Meng H,Wang B, Yang Y, Wang Q, Su N, Li J, Li D, 2016, Identification of upstream regulators for synovial expression signature genes in osteoarthritis. Joint Bone Spine 83(5):545–551. , DOI 10.1016/j.jbspin.2015.09.001 Luo Y, Deng Z, Chen J, 2013, Pivotal regulatory network and genes in osteosarcoma. Arch Med Sci 9(3):569–575. https://doi. org/10.5114/aoms.2012.30956 Horsley V, Friday BB,Matteson S, Kegley KM, Gephart J, Pavlath GK, 2001, Regulation of the growth of multinucleated muscle cells by an NFATC2-dependent pathway. J Cell Biol 153(2):329–338 Wang J, Gardner BM, Lu Q, Rodova M, Woodbury BG, Yost JG, Roby KF, Pinson DM, Tawfik O, Anderson HC, 2009, Transcription factor Nfat1 deficiency causes osteoarthritis through dysfunction of adult articular chondrocytes. J Pathol 219(2):163– 172. , DOI 10.1002/path.2578 Bauer W, Rauner M, Haase M, Kujawski S, Arabanian LS, Habermann I, Hofbauer LC, Ehninger G, Kiani A, 2011, Osteomyelosclerosis, anemia and extramedullary hematopoiesis in mice lacking the transcription factor NFATc2. Haematologica 96(11):1580–1588. , DOI 10.3324/haematol.2011.042515 Kingsley C,Winoto A, 1992, Cloning of GT box-binding proteins: a novel Sp1 multigene family regulating T-cell receptor gene expression. Mol Cell Biol 12(10):4251–4261 Kim TH, Chiera SL, Linder KE, Trempus CS, Smart RC, Horowitz JM, 2010, Overexpression of transcription factor sp2 inhibits epidermal differentiation and increases susceptibility to wound- and carcinogen-induced tumorigenesis. Cancer Res 70(21):8507– 8516. , DOI 10.1158/0008-5472.can-10-1213 Ambrogini E, AlmeidaM, Martin-MillanM, Paik JH, Depinho RA, Han L, Goellner J,Weinstein RS, Jilka RL, O'Brien CA,Manolagas SC, 2010, FoxO-mediated defense against oxidative stress in osteoblasts is indispensable for skeletal homeostasis in mice. Cell Metab 11(2):136–146. , DOI 10.1016/j.cmet.2009.12.009 Almeida M, 2011, Unraveling the role of FoxOs in bone–insights from mouse models. Bone 49(3):319–327. , DOI 10.1016/ j.bone.2011.05.023 Turrel-Davin F, Tournadre A, Pachot A, Arnaud B, Cazalis MA, Mougin B,Miossec P, 2010, FoxO3a involved in neutrophil and T cell survival is overexpressed in rheumatoid blood and synovial tissue. Ann Rheum Dis 69(4):755–760. , DOI 10.1136/ ard.2009.109991 Lee JC, Espeli M, Anderson CA, Linterman MA, Pocock JM, Williams NJ, Roberts R, Viatte S, Fu B, Peshu N, Hien TT, Phu NH, Wesley E, Edwards C, Ahmad T, Mansfield JC, Gearry R, Dunstan S, Williams TN, Barton A, Vinuesa CG, Parkes M, Lyons PA, Smith KG, 2013, Human SNP links differential outcomes in inflammatory and infectious disease to a FOXO3- regulated pathway. Cell 155(1):57–69. , DOI 10.1016/j. cell.2013.08.034 Matsunoshita Y, Ijiri K, Ishidou Y, Nagano S, Yamamoto T, Nagao H, Komiya S, Setoguchi T, 2011, Suppression of osteosarcoma cell invasion by chemotherapy is mediated by urokinase plasminogen activator activity via up-regulation of EGR1. PLoS One 6(1): e16234. , DOI 10.1371/journal.pone.0016234 Liu Y, Zhang N,Wang Y, Xu M, Liu N, Pang X, Cao J, Ma N, Pang H, Liu L, Zhang H, 2012, Zinc finger E-box binding homeobox 1 promotes invasion and bone metastasis of small cell lung cancer in vitro and in vivo. Cancer Sci 103(8):1420–1428. , DOI 10.1111/j.1349-7006.2012.02347.x Shen A, Zhang Y, Yang H, Xu R, Huang G, 2012, Overexpression of ZEB1 relates to metastasis and invasion in osteosarcoma. J Surg Oncol 105(8):830–834. , DOI 10.1002/jso.23012 Nitz MD, Harding MA, Smith SC, Thomas S, Theodorescu D, 2011, RREB1 transcription factor splice variants in urologic cancer. Am J Pathol 179(1):477–486. , DOI 10.1016/j.ajpath. 2011.03.038 Liu H, Hew HC, Lu ZG, Yamaguchi T, Miki Y, Yoshida K, 2009, DNA damage signalling recruits RREB-1 to the p53 tumour suppressor promoter. Biochem J 422(3):543–551. , DOI 10. 1042/bj20090342 Li X, Zhang Z, Yu M, Li L, Du G, Xiao W, Yang H, 2013, Involvement of miR−20a in promoting gastric cancer progression by targeting early growth response 2, EGR2). Int J Mol Sci 14(8): 16226–16239. , DOI 10.3390/ijms140816226 Unoki M, Nakamura Y, 2001, Growth-suppressive effects of BPOZ and EGR2, two genes involved in the PTEN signaling pathway. Oncogene 20(33):4457–4465. , DOI 10.1038/sj.onc. 1204608 Bradley E, Ruan M, Oursler MJ, 2008, Novel pro-survival functions of the Kruppel-like transcription factor Egr2 in promotion of macrophage colony-stimulating factor-mediated osteoclast survival downstream of the MEK/ERK pathway. J Biol Chem 283(12): 8055–8064 Levi G, Topilko P, Schneider-Maunoury S, Lasagna M, Mantero S, Cancedda R, Charnay P, 1996, Defective bone formation in Krox- 20 mutant mice. Development 122(1):113–120 Miano JM, 2003, Serum response factor: toggling between disparate programs of gene expression. J Mol Cell Cardiol 35(6):577– 593 Li S, Czubryt MP, McAnally J, Bassel-Duby R, Richardson JA, Wiebel FF, Nordheim A, Olson EN, 2005, Requirement for serum response factor for skeletal muscle growth and maturation revealed by tissue-specific gene deletion in mice. Proc Natl Acad Sci U S A 102(4):1082–1087. , DOI 10.1073/pnas.0409103102 Chen J, Yuan K, Mao X, Miano JM, Wu H, Chen Y, 2012, Serum response factor regulates bone formation via IGF-1 and Runx2 signals. J Bone Miner Res Off J Am Soc Bone Miner Res 27(8): 1659–1668. , DOI 10.1002/jbmr.1607 Chandar N, Saluja R, Lamar PC, Kolman K, ProzialeckWC, 2005, P53 and beta-catenin activity during estrogen treatment of osteoblasts. Cancer Cell Int 5(24). , DOI 10.1186/1475-2867-5- 24 Mendoza S, Konishi T, DernellWS,Withrow SJ, MillerCW(1998, Status of the p53, Rb and MDM2 genes in canine osteosarcoma. Anticancer Res 18(6a):4449–4453 Bongiovanni L, Mazzocchetti F, Malatesta D, Romanucci M, Ciccarelli A, Buracco P, De Maria R, Palmieri C, Martano M, Morello E, Maniscalco L, Della Sa l d a L, 2012, Immunohistochemical investigation of cell cycle and apoptosis regulators, survivin, beta-catenin, p53, caspase 3, in canine appendicular osteosarcoma. BMC Vet Res 8:78. , DOI 10.1186/ 1746-6148-8-78 Angulo P, Kaushik G, Subramaniam D, Dandawate P, Neville K, Chastain K, Anant S, 2017, Natural compounds targeting major cell signaling pathways: a novel paradigm for osteosarcoma therapy. J Hematol Oncol 10(1):10. , DOI 10.1186/s13045-016- 0373-z Thomas DM, 2010, Wnts, bone and cancer. J Pathol 220(1):1–4. , DOI 10.1002/path.2635 Iwaya K, Ogawa H, KurodaM, Izumi M, Ishida T,Mukai K, 2003, Cytoplasmic and/or nuclear staining of beta-catenin is associated with lung metastasis. Clin Exp Metastasis 20(6):525–529 Kansara M, Tsang M, Kodjabachian L, Sims NA, Trivett MK, Ehrich M, Dobrovic A, Slavin J, Choong PF, Simmons PJ, Dawid IB, Thomas DM, 2009, Wnt inhibitory factor 1 is epigenetically silenced in human osteosarcoma, and targeted disruption accelerates osteosarcomagenesis in mice. J Clin Invest 119(4):837–851. , DOI 10.1172/jci37175 Hoang BH, Kubo T, Healey JH, Yang R, Nathan SS, Kolb EA, Mazza B, Meyers PA, Gorlick R, 2004, Dickkopf 3 inhibits invasion and motility of Saos-2 osteosarcoma cells by modulating the Wnt-beta-catenin pathway. Cancer Res 64(8):2734–2739 Guo Y, Zi X, Koontz Z, Kim A, Xie J, Gorlick R, Holcombe RF, Hoang BH, 2007, Blocking Wnt/LRP5 signaling by a soluble receptor modulates the epithelial to mesenchymal transition and suppresses met and metalloproteinases in osteosarcoma Saos-2 cells. J Orthop Res 25(7):964–971. , DOI 10.1002/jor.20356 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1091 EP 1102 DI 10.1007/s12253-018-0519-0 PG 12 ER PT J AU Lendvai, G Szekerczes, T Gyongyosi, B Schlachter, K Kontsek, E Pesti, A Patonai, A Werling, K Kovalszky, I Schaff, Zs Kiss, A AF Lendvai, Gabor Szekerczes, Timea Gyongyosi, Benedek Schlachter, Krisztina Kontsek, Endre Pesti, Adrian Patonai, Attila Werling, Klara Kovalszky, Ilona Schaff, Zsuzsa Kiss, Andras TI MicroRNA Expression in Focal Nodular Hyperplasia in Comparison with Cirrhosis and Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Focal nodular hyperplasia; Hepatic cirrhosis; Hepatocellular carcinoma; microRNA; Chronic hepatitis C ID Focal nodular hyperplasia; Hepatic cirrhosis; Hepatocellular carcinoma; microRNA; Chronic hepatitis C AB The liver disease focal nodular hyperplasia (FNH) has several histological features that resemble hepatic cirrhosis. Since cirrhosis may develop further into hepatocellular carcinoma (HCC) contrary to FNH, the aim of the present study was to identify microRNAs (miRNA), which, by their altered expression levels, may be associated with the benign, tumor-like nature of FNH. Altogether 106 surgically removed formalin-fixed paraffin-embedded liver samples were selected, including 22 FNH, 45 cirrhosis, 24 HCC and 15 normal liver tissues. Etiology of the cases of cirrhosis and HCC includes hepatitis C and alcoholism and the HCC cases developed in cirrhotic livers. Relative expression levels of 14 miRNAs were determined using TaqMan MicroRNA Assays. In comparison to normal liver, the levels of miR-34a and miR-224 were elevated not only in FNH but also in cirrhosis and HCC, while the expression of miR-17-5p, miR-18a and miR-210 was decreased in FNH. Further, the levels of miR- 21 and miR-222 were increased in cirrhosis and HCC but were decreased in FNH and the expression of miR-17-5p, miR-18a, miR-195 and miR-210 was decreased in FNH as compared with cirrhosis and/or HCC. In conclusion, the elevation of miR-34a and miR-224 may be associated with both benign and malignant proliferative processes, nevertheless the increased expression of oncomiRs miR-21 and miR-222 in cirrhosis and HCC but not in FNH may be related to malignant processes of the liver. The decreased levels of miR-18a, miR-195 and miR-210 may further differentiate FNH from cirrhosis, reflecting the different pathogenesis of these two entities contrary to some histologically similar features. C1 [Lendvai, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Szekerczes, Timea] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Gyongyosi, Benedek] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Schlachter, Krisztina] National Institute of Oncology, Department of Diagnostic Pathology, 1122 Budapest, Hungary. [Kontsek, Endre] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Pesti, Adrian] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Patonai, Attila] Semmelweis University, Department of Transplantation and Surgery, 1082 Budapest, Hungary. [Werling, Klara] Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. RP Lendvai, G (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM lendvai.gabor@med.semmelweis-univ.hu CR Chen SL, Zheng MH, Shi KQ, Yang T, Chen YP, 2013, A new strategy for treatment of liver fibrosis: letting MicroRNAs do the job. BioDrugs 27(1):25–34 Mormone E, George J, Nieto N, 2011, Molecular pathogenesis of hepatic fibrosis and current therapeutic approaches. Chem Biol Interact 193(3):225–231 Roncalli M, Sciarra A, Tommaso LD, 2016, Benign hepatocellular nodules of healthy liver: focal nodular hyperplasia and hepatocellular adenoma. Clin Mol Hepatol 22(2):199–211 Balabaud C, Al-Rabih WR, Chen PJ, Evason K, Ferrell L, Hernandez-Prera JC, Huang SF, Longerich T, Park YN, Quaglia A, Schirmacher P, Sempoux C, Thung SN, Torbenson M, Wee A, Yeh MM, Yeh SH, Le Bail B, Zucman-Rossi J, Bioulac-Sage P, 2013, Focal nodular hyperplasia and hepatocellular adenoma around the world viewed through the scope of the Immunopathological classification. Int J Hepatol 2013:268625 Kondo F, Fukusato T, Kudo M, 2014, Pathological diagnosis of benign hepatocellular nodular lesions based on the new World Health Organization classification. Oncology 87(Suppl 1):37–49 Sempoux C, Balabaud C, Bioulac-Sage P, 2014, Pictures of focal nodular hyperplasia and hepatocellular adenomas.World J Hepatol 6(8):580–595 Venturi A, Piscaglia F, Vidili G, Flori S, Righini R, Golfieri R, Bolondi L, 2007, Diagnosis and management of hepatic focal nodular hyperplasia. J Ultrasound 10(3):116–127 Nahm CB, Ng K, Lockie P, Samra JS, Hugh TJ, 2011, Focal nodular hyperplasia–a review of myths and truths. J Gastrointest Surg 15(12):2275–2283 Sohrabpour AA,MohamadnejadM,Malekzadeh R, 2012, Review article: the reversibility of cirrhosis. Aliment Pharmacol Ther 36(9): 824–832 Liou IW, 2014, Management of end-stage liver disease. Med Clin North Am 98(1):119–152 Murakami Y, Kawada N, 2017, MicroRNAs in hepatic pathophysiology. Hepatol Res 47(1):60–69 Bandiera S, Pfeffer S, Baumert TF, Zeisel MB, 2015, miR-122–a key factor and therapeutic target in liver disease. J Hepatol 62(2): 448–457 Hu J, Xu Y, Hao J, Wang S, Li C, Meng S, 2012, MiR-122 in hepatic function and liver diseases. Protein Cell 3(5):364–371 Lee CH, Kim JH, Lee SW, 2014, The role of microRNAs in hepatitis C virus replication and related liver diseases. J Microbiol 52(6):445–451 Huan L, Liang LH, He XH, 2016, Role of microRNAs in inflammation-associated liver cancer. Cancer Biol Med 13(4): 407–425 Lendvai G, Jarmay K, Karacsony G, Halasz T, Kovalszky I, Baghy K, Wittmann T, Schaff Z, Kiss A, 2014, Elevated miR-33a and miR-224 in steatotic chronic hepatitis C liver biopsies. World J Gastroenterol 20(41):15343–15350 GyugosM, Lendvai G, Kenessey I, Schlachter K, Halasz J, Nagy P, GaramiM, Jakab Z, Schaff Z, KissA, 2014)MicroRNAexpression might predict prognosis of epithelial hepatoblastoma. Virchows Arch 464(4):419–427 Szabo G, Bala S, 2013, MicroRNAs in liver disease. Nat Rev Gastroenterol Hepatol 10(9):542–552 Halasz T, Horvath G, Par G,Werling K, Kiss A, Schaff Z, Lendvai G, 2015, miR-122 negatively correlates with liver fibrosis as detected by histology and FibroScan. World J Gastroenterol 21(25): 7814–7823 DoleshalM,Magotra AA, Choudhury B, Cannon BD, Labourier E, Szafranska AE, 2008, Evaluation and validation of total RNA extraction methods for microRNA expression analyses in formalinfixed, paraffin-embedded tissues. J Mol Diagn 10(3):203–211 Borel F, Konstantinova P, Jansen PL, 2012, Diagnostic and therapeutic potential of miRNA signatures in patients with hepatocellular carcinoma. J Hepatol 56(6):1371–1383 Ladeiro Y, Couchy G, Balabaud C, Bioulac-Sage P, Pelletier L, Rebouissou S, Zucman-Rossi J, 2008, MicroRNA profiling in hepatocellular tumors is associated with clinical features and oncogene/tumor suppressor gene mutations. Hepatology 47(6): 1955–1963 Murakami Y, Yasuda T, Saigo K, Urashima T, Toyoda H, Okanoue T, Shimotohno K, 2006, Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene 25(17):2537–2545 Andersen CL, Jensen JL, Orntoft TF, 2004, Normalization of realtime quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res 64(15):5245–5250 Bioulac-Sage P, Balabaud C, Bedossa P, Scoazec JY, Chiche L, Dhillon AP, Ferrell L, Paradis V, Roskams T, Vilgrain V, Wanless IR, Zucman-Rossi J, 2007, Pathological diagnosis of liver cell adenoma and focal nodular hyperplasia: Bordeaux update. J Hepatol 46(3):521–527 Maillette de Buy Wenniger L, Terpstra V, Beuers U, 2010, Focal nodular hyperplasia and hepatic adenoma: epidemiology and pathology. Dig Surg 27(1):24–31 Pinzani M, 2015, Pathophysiology of liver fibrosis. Dig Dis 33(4): 492–497 He Y, Huang C, Zhang SP, Sun X, Long XR, Li J, 2012, The potential of microRNAs in liver fibrosis. Cell Signal 24(12): 2268–2272 Roderburg C, Luedde T, 2014, Circulating microRNAs as markers of liver inflammation, fibrosis and cancer. J Hepatol 61(6):1434– 1437 Tian XF, Ji FJ, Zang HL, Cao H, 2016, Activation of the miR-34a/ SIRT1/p53 signaling pathway contributes to the Progress of liver fibrosis via inducing apoptosis in hepatocytes but not in HSCs. PLoS One 11(7):e0158657 Ding J, Li M, Wan X, Jin X, Chen S, Yu C, Li Y, 2015, Effect of miR-34a in regulating steatosis by targeting PPARalpha expression in nonalcoholic fatty liver disease. Sci Rep 5:13729 Pineau P, Volinia S, McJunkin K,Marchio A, Battiston C, Terris B, Mazzaferro V, Lowe SW, Croce CM, Dejean A, 2010, miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci U S A 107(1):264–269 Han Y, Meng F, Venter J, Wu N, Wan Y, Standeford H, Francis H, Meininger C, Greene J Jr, Trzeciakowski JP, Ehrlich L, Glaser S, Alpini G, 2016, miR-34a-dependent overexpression of Per1 decreases cholangiocarcinoma growth. J Hepatol 64(6):1295–1304 Yan G, Li B, Xin X, Xu M, Ji G, Yu H, 2015, MicroRNA-34a promotes hepatic stellate cell activation via targeting ACSL1. Med Sci Monit 21:3008–3015 Sato Y, Harada K, Ikeda H, Fijii T, Sasaki M, Zen Y, Nakanuma Y, 2009, Hepatic stellate cells are activated around central scars of focal nodular hyperplasia of the liver–a potential mechanism of central scar formation. Hum Pathol 40(2):181–188 Cheng J, Zhou L, Xie QF, Xie HY,Wei XY, Gao F, Xing CY, Xu X, Li LJ, Zheng SS, 2010, The impact of miR-34a on protein output in hepatocellular carcinoma HepG2 cells. Proteomics 10(8):1557– 1572 Ma D, Tao X, Gao F, Fan C,Wu D, 2012, miR-224 functions as an onco-miRNA in hepatocellular carcinoma cells by activating AKT signaling. Oncol Lett 4(3):483–488 Wang Y, Ren J, Gao Y, Ma JZ, Toh HC, Chow P, Chung AY, Ooi LL, Lee CG, 2013, MicroRNA-224 targets SMAD family member 4 to promote cell proliferation and negatively influence patient survival. PLoS One 8(7):e68744 Li Q, Ding C, Chen C, Zhang Z, Xiao H, Xie F, Lei L, Chen Y, Mao B, Jiang M, Li J, Wang D, Wang G, 2014, miR-224 promotion of cell migration and invasion by targeting Homeobox D 10 gene in human hepatocellular carcinoma. J Gastroenterol Hepatol 29(4): 835–842 Scisciani C, Vossio S, Guerrieri F, Schinzari V, De Iaco R, de Meo D’Onorio P, Cervello M, Montalto G, Pollicino T, Raimondo G, Levrero M, Pediconi N, 2012, Transcriptional regulation of miR- 224 upregulated in human HCCs by NFkappaB inflammatory pathways. J Hepatol 56(4):855–861 Gyongyosi B, Vegh E, Jaray B, Szekely E, Fassan M, Bodoky G, Schaff Z, Kiss A, 2014, Pretreatment MicroRNA level and outcome in Sorafenib-treated hepatocellular carcinoma. J Histochem Cytochem 62(8):547–555 Iorio MV, Croce CM, 2012, microRNA involvement in human cancer. Carcinogenesis 33(6):1126–1133 WongQW, Ching AK, Chan AW, ChoyKW, To KF, Lai PB,Wong N, 2010, MiR-222 overexpression confers cell migratory advantages in hepatocellular carcinoma through enhancing AKT signaling. Clin Cancer Res 16(3):867–875 He C, Dong X, Zhai B, Jiang X, Dong D, Li B, Jiang H, Xu S, Sun X, 2015, MiR-21 mediates sorafenib resistance of hepatocellular carcinoma cells by inhibiting autophagy via the PTEN/Akt pathway. Oncotarget 6(30):28867–28881 Bao L, Yan Y, Xu C, Ji W, Shen S, Xu G, Zeng Y, Sun B, Qian H, Chen L, Wu M, Su C, Chen J, 2013, MicroRNA-21 suppresses PTEN and hSulf-1 expression and promotes hepatocellular carcinoma progression through AKT/ERK pathways. Cancer Lett 337(2):226–236 Yang YF,Wang F, Xiao JJ, SongY, Zhao YY, Cao Y, Bei YH, Yang CQ, 2014, MiR-222 overexpression promotes proliferation of human hepatocellular carcinoma HepG2 cells by downregulating p27. Int J Clin Exp Med 7(4):893–902 Melnik BC, 2015, MiR-21: an environmental driver of malignant melanoma? J Transl Med 13:202 Tili E, Michaille JJ, Croce CM, 2013, MicroRNAs play a central role in molecular dysfunctions linking inflammation with cancer. Immunol Rev 253(1):167–184 Ogawa T, Enomoto M, Fujii H, Sekiya Y, Yoshizato K, Ikeda K, Kawada N, 2012, MicroRNA-221/222 upregulation indicates the activation of stellate cells and the progression of liver fibrosis. Gut 61(11):1600–1609 Kim Y, Jho EH, 2017, Deubiquitinase YOD1: the potent activator of YAP in hepatomegaly and liver cancer. BMB Rep 50(6):281– 282 Patel SH, Camargo FD, Yimlamai D, 2017, Hippo signaling in the liver regulates organ size, cell fate, and Carcinogenesis. Gastroenterology 152(3):533–545 Cloonan N, Brown MK, Steptoe AL, Wani S, Chan WL, Forrest AR, Kolle G, Gabrielli B, Grimmond SM, 2008, The miR-17-5p microRNAis a key regulator of the G1/S phase cell cycle transition. Genome Biol 9(8):R127 Liu L, Cai X, Liu E, Tian X, Tian C, 2017, MicroRNA-18a promotes proliferation and metastasis in hepatocellular carcinoma via targeting KLF4. Oncotarget 8(40):68263–68269 Wang M, Zhang J, Tong L, Ma X, Qiu X, 2015, MiR-195 is a key negative regulator of hepatocellular carcinoma metastasis by targeting FGF2 and VEGFA. Int J Clin Exp Pathol 8(11):14110– 14120 Song LY, Ma YT, Wu CF, Wang CJ, Fang WJ, Liu SK, 2017, MicroRNA-195 activates hepatic stellate cells in vitro by targeting Smad7. Biomed Res Int 2017:1945631 Ivan M, Huang X, 2014, miR-210: fine-tuning the hypoxic response. Adv Exp Med Biol 772:205–227 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1103 EP 1109 DI 10.1007/s12253-018-0528-z PG 7 ER PT J AU Yang, XH Liu, L Hu, YJ Zhang, P Hu, QG AF Yang, Xi-Hu Liu, Liu Hu, Yong-Jie Zhang, Ping Hu, Qin-Gang TI Co-expression of XIAP and CIAP1 Play Synergistic Effect on Patient’s Prognosis in Head and Neck Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Head and neck; Squamous cell carcinoma; XIAP and CIAP1; Prognosis ID Head and neck; Squamous cell carcinoma; XIAP and CIAP1; Prognosis AB To explore the influence of chemotherapy on prognosis of Head and Neck Squamous Cell Carcinoma (HNSCC) and the relationship between XIAP and CIAP1 co-expression and the prognosis in HNSCC. 129 patients were recruited in our study, they were divided into two groups, neoadjuvant group (n = 60) and non-neoadjuvant group (n = 69). Expression level of XIAP and CIAP1 were examed in neoadjuvant group, and was correlated with clinical outcomes of the patients. The unselected patients were not benefit from neoadjuvant chemotherapy. Moreover, the patients whose tumors co-express high level of XIAP and CIAP1 presented poorer overall and disease-free survival rates than those whose tumors co-express low level of XIAP and CIAP1 (overall survival P < 0.001, disease-free survival P < 0.001). Our results validate that individual chemotherapy is important for HNSCC, and co-expression of XIAP and CIAP1 prompted a worse prognosis. C1 [Yang, Xi-Hu] Medical School of Nanjing University, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial Surgery, 210000 Nanjing, Jiangsu, China. [Liu, Liu] Shanghai Jiao Tong University School of Medicine, Ninth People’s Hospital, Department of Oral and Maxillofacial Surgery, 200011 Shanghai, China. [Hu, Yong-Jie] Shanghai Jiao Tong University School of Medicine, Ninth People’s Hospital, Department of Oral and Maxillofacial Surgery, 200011 Shanghai, China. [Zhang, Ping] Boston University, Henry M. Goldman School of Dental Medicine, 02125 Boston, MA, USA. [Hu, Qin-Gang] Medical School of Nanjing University, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial Surgery, 210000 Nanjing, Jiangsu, China. RP Hu, QG (reprint author), Medical School of Nanjing University, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial Surgery, 210000 Nanjing, China. EM qinganghu@hotmail.com CR Greenlee RT, Hill-Harmon MB, Murray T, Thun M, 2001, Cancer statistics. CA Cancer J Clin 51(1):15–36 Chen Z, Seimiya H, Naito M, Mashima T, Kizaki A, Dan S, Imaizumi M, Ichijo H, Miyazono K, Tsuruo T, 1999, ASK1 mediates apoptotic cell death induced by genotoxic stress. Oncogene 18(1):173–180 Mase H, Sasaki A, Alcalde RE et al, 2000, Regulation of apoptasis reduction in the cisplatin-resistant cell line by Bcl-2 and CPP32. Chemotherapy 46:69–76 Nakano Y, Bilim V, Yuuki K, Muto A, Kato T, Nagaoka A, Tomita Y, 2009, Molecular targeting of Bcl-2 overcomes prostate cancer cell adaptation to XIAP gene down regulation. Prostate Cancer Prostatic Dis 12(1):34–40 McManus DC, Lefebvre CA, Cherton-Horvat G, St-Jean M, Kandimalla ER, Agrawal S, Morris SJ, Durkin JP, LaCasse EC, 2004, Loss of XIAP protein expression by RNAi and antisense approaches sensitizes cancer cells to functionally diverse chemotherapeutics. Oncogene 23:8105–8117 Tamm I, Trepel M, Cardo-Vila M et al, 2003, Peptides targeting caspase inhibitors. J Biol Chem 278:14401–14405 Ma J-j, Chen B-l, Xin X-y, 2009, XIAP gene downregulation by small interfering RNA inhibits proliferation, induces apoptosis, and reverses the cisplatin resistance of ovarian carcinoma. Eur J Obstet Gynecol Reprod Biol 146:222–226 LaCasse EC, Mahoney DJ, Cheung HH et al, 2008, IAP-targeted therapies for cancer. Oncogene 27:6252–6275 Reed JC, 2001, The survivin saga goes in vivo. J Clin Invest 108: 965–969 Morizane Y, Honda R, Fukami K, Yasuda H, 2005, X-linked inhibitor of apoptosis functions as ubiquitin ligase toward mature caspase-9 and cytosolic Smac/DIABLO. J Biochem, Tokyo, 137: 125–132 Scott FL, Denault JB, Riedl SJ, Shin H, Renatus M, Salvesen GS, 2005, XIAP inhibits caspase-3 and-7 using two binding sites: evolutionarily conserved mechanism of IAPs. EMBO J 24:645–655 Suzuki Y, Nakabayashi Y, Takahashi R, 2001, Ubiquitin-protein ligase activity of X-linked inhibitor of apoptosis protein promotes proteasomal degradation of caspase-3 and enhances its antiapoptotic effect in Fas-induced cell death. Proc Natl Acad Sci U S A 98:8662–8667 Holcik M, Gibson H, Korneluk RG, 2001, XIAP: apoptotic brake and pro-mising therapeutic target. Apoptosis 6:253–261 LaCasse EC, Baird S, Korneluk RG, MacKenzie AE, 1998, The inhibitors of apoptosis, IAPs, and their emerging role in cancer. Oncogene 17:3247–3259 Hu YP, Cherton-Horvat G, Dragowska V et al, 2003, Antisense oligonucleotides targeting XIAP induce apoptosis and enhance chemotherapeutic activity against human lung cancer cells in vitro and in vivo. Clin Cancer Res 9:2826–2836 Amantana A, London CA, Iversen PL, Devi GR, 2004, X-linked inhibitor of apoptosis protein inhibition induces apoptosis and enhances chemotherapy sensitivity in human prostate cancer cells. Mol Cancer Ther 3(6):699–707 Tirro E, Consoli ML, Massimino M, Manzella L, Frasca F, Sciacca L, Vicari L, Stassi G, Messina L, Messina A, Vigneri P, 2006, Altered expression of c-IAP1, survivin, and Smac contributes to chemotherapy resistance in thyroid cancer cells. Cancer Res 66(8):4263–4272 Yang XH, Feng ZE, Yan M, Hanada S, Zuo H, Yang CZ, Han ZG, GuoW, Chen WT, Zhang P, 2012, XIAP is a predictor of cisplatinbased chemotherapy response and prognosis for patients with advanced head and neck cancer. PLoS One 7(3):e31601 Licitra L, Grandi C, Guzzo M, Mariani L, Lo Vullo S et al, 2003, Primary chemotherapy in resectable oral cavity squamous cell cancer: a randomized controlled trial. J Clin Oncol 21:327–333 Liston P, Young SS, Mackenzie AE et al, 2004, Life and death decisions: the role of the IAPs in modulating programmed cell death. Apoptosis 2:423–441 Li Y, Jian Z, Xia K, Li X, Lv X, Pei H, Chen Z, Li J, 2006, XIAP is related to the chemoresistance and inhibited its expression by RNA interferenc sensitize pancreatic carcinoma cells to chemotherapeutics. Pancreas 32:288–296 Holcik M, Yeh C, Korneluk RG, Chow T, 2000, Translational upregulation of X-linked inhibitor of apoptosis, XIAP, increases resistance to radiation induced cell death. Oncogene 19:4174–4177 Guoan X, Xiaomin W, Hanning W, Kaiyun C, Hao L, 2009, Xlinked inhibitor of apoptosis protein in human colorectal cancer and its correlation with prognosis. J Surg Oncol 100:708–712 Mizutani Y, Nakanishi H, Li YN, Matsubara H, Yamamoto K, Sato N, Shiraishi T, Nakamura T, Mikami K, Okihara K, Takaha N, Ukimura O, Kawauchi A, Nonomura N, Bonavida B, Miki T, 2007, Overexpression of XIAP expression in renal cell carcinoma predicts a worse prognosis. Int J Oncol 30(4):919–925 Yang YF, Du H, Huang P et al, 2008, XIAP expression and its predictive significance of prognosis in stage IIB osteosarcomas. Chin-Ger J Clin Oncol 7:416–419 Tanimoto T, Tsuda H, Imazeki N, Ohno Y, Imoto I, Inazawa J, Matsubara O, 2005, Nuclear expression of cIAP-1, an apoptosis inhibiting protein, predicts lymph node metastasis and poor patient prognosis in head and neck squamous cell carcinomas. Cancer Lett 224(1):141–151 Li J, Feng Q, Kim JM, Schneiderman D, Liston P, Li M, Vanderhyden B, Faught W, Fung MFK, Senterman M, Korneluk RG, Tsang BK, 2001, Human ovarian cancer and cisplatin resistance: possible role of inhibitor of apoptosis proteins. Endocrinology 142:370–380 Matsumiya T, Imaizumi T, Yoshida H, Kimura H, Satoh K, 2001, Cisplatin inhibits the expression of XIAP in an oral carcinoma cell line. Oral Oncol 37:296–300 Dynek JN, Vucic D, 2013, Antagonists of IAP proteins as cancer therapeutics. Cancer Lett 332(2):206–214 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1111 EP 1116 DI 10.1007/s12253-018-0533-2 PG 6 ER PT J AU Lambert, A Salleron, J Lion, M Rouyer, M Lozano, N Leroux, A Merlin, JL Harle, A AF Lambert, Aurelien Salleron, Julia Lion, M Rouyer, Marie Lozano, Nicolas Leroux, Agnes Merlin, Jean-Louis Harle, Alexandre TI Comparison of Three Real-Time PCR Assays for the Detection of PIK3CA Somatic Mutations in Formalin-Fixed Paraffin Embedded Tissues of Patients with Breast Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PIK3CA; Real-time PCR; Breast cancer; PI3K; ARMS; HRM ID PIK3CA; Real-time PCR; Breast cancer; PI3K; ARMS; HRM AB Breast cancer is the leading cause of cancer-related death in women worldwide. Mutations of the PIK3CA gene are found in approximately 25% of breast carcinomas and are reported as activators of the PI3K/AKT/mTOR pathway. This study aims to compare three assays for the somatic mutation detection of PIK3CA gene in FFPE tissues of patients with breast cancer. We compared Cobas® PIK3CA Mutation Test (Roche Diagnostics, Meylan, France), PCR amplification-refractory mutation system Scorpions® (ARMS) and High-Resolution Melting PCR assay (HRM) for the detection of PIK3CA mutations. Discrepant samples were assessed using Next Generation Sequencing (NGS). 46 FFPE breast carcinomas samples of patients treated for breast cancer have been assessed for PIK3CA mutations using the three PCR assays. Among the 46 samples, 17 (37.8%), 13 (28.36%) and 19 (41.3%) had a PIK3CA mutation, with Cobas®, ARMS and HRM assays respectively. Three different mutations of PIK3CA have been detected for one sample. Calculated kappa were 0.95[0.86;1] between Cobas® and HRM, 0.75[0.55;0.95] between Cobas® and ARMS and 0.72[0.51;0.92] between HRM and ARMS. Five samples were found with discrepant results. Our study shows that the Cobas® assay is suitable for PIK3CA mutation assessment in patients with breast cancer. HRM assay is also suitable for PIK3CA mutation assessment but requires a mutation characterization with a specific assay. C1 [Lambert, Aurelien] Institut de Cancerologie de Lorraine, Departement d’Oncologie Medicale, 54519 Vandoeuvre les Nancy, France. [Salleron, Julia] Institut de Cancerologie de Lorraine, Departement de Biostatistiques, 54519 Vandoeuvre les Nancy, France. [Lion, M] Universite de Lorraine, Institut de Cancerologie de Lorraine, Service de Biopathologie, CNRS UMR 7039 CRAN, 6 avenue de Bourgogne, 54519 Vandoeuvre les Nancy, France. [Rouyer, Marie] Universite de Lorraine, Institut de Cancerologie de Lorraine, Service de Biopathologie, CNRS UMR 7039 CRAN, 6 avenue de Bourgogne, 54519 Vandoeuvre les Nancy, France. [Lozano, Nicolas] Universite de Lorraine, Institut de Cancerologie de Lorraine, Service de Biopathologie, CNRS UMR 7039 CRAN, 6 avenue de Bourgogne, 54519 Vandoeuvre les Nancy, France. [Leroux, Agnes] Universite de Lorraine, Institut de Cancerologie de Lorraine, Service de Biopathologie, CNRS UMR 7039 CRAN, 6 avenue de Bourgogne, 54519 Vandoeuvre les Nancy, France. [Merlin, Jean-Louis] Universite de Lorraine, Institut de Cancerologie de Lorraine, Service de Biopathologie, CNRS UMR 7039 CRAN, 6 avenue de Bourgogne, 54519 Vandoeuvre les Nancy, France. [Harle, Alexandre] Universite de Lorraine, Institut de Cancerologie de Lorraine, Service de Biopathologie, CNRS UMR 7039 CRAN, 6 avenue de Bourgogne, 54519 Vandoeuvre les Nancy, France. RP Harle, A (reprint author), Universite de Lorraine, Institut de Cancerologie de Lorraine, Service de Biopathologie, CNRS UMR 7039 CRAN, 54519 Vandoeuvre les Nancy, France. EM a.harle@nancy.unicancer.fr CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127(12):2893–2917. https://doi. org/10.1002/ijc.25516 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108. , DOI 10.3322/caac.21262 Cossu-Rocca P, Orru S, Muroni MR, Sanges F, Sotgiu G, Ena S, Pira G, Murgia L, Manca A, Uras MG, Sarobba MG, Urru S, De Miglio MR, 2015, Analysis of PIK3CA mutations and activation pathways in triple negative breast Cancer. PLoS One 10(11): e0141763. , DOI 10.1371/journal.pone.0141763 Loibl S, Majewski I, Guarneri V, Nekljudova V, Holmes E, Bria E, Denkert C, Schem C, Sotiriou C, Loi S, Untch M, Conte P, Bernards R, Piccart M, von Minckwitz G, Baselga J, 2016, PIK3CA mutations are associated with reduced pathological complete response rates in primary HER2-positive breast cancer: pooled analysis of 967 patients from five prospective trials investigating lapatinib and trastuzumab. Ann Oncol 27(8):1519–1525. , DOI 10.1093/annonc/mdw197 Baselga J, Cortes J, Im SA, Clark E, Ross G, Kiermaier A, Swain SM, 2014, Biomarker analyses in CLEOPATRA: a phase III, placebo-controlled study of pertuzumab in human epidermal growth factor receptor 2-positive, first-line metastatic breast cancer. J Clin Oncol 32(33):3753–3761. , DOI 10.1200/jco.2013. 54.5384 Harle A, Lion M, Lozano N, Husson M, Harter V, Genin P, Merlin JL, 2013, Analysis of PIK3CA exon 9 and 20 mutations in breast cancers using PCR-HRM and PCR-ARMS: correlation with clinicopathological criteria. Oncol Rep 29(3):1043–1052. https://doi. org/10.3892/or.2013.2229 Briffa R, Um I, Faratian D, Zhou Y, Turnbull AK, Langdon SP, Harrison DJ, 2015, Multi-scale genomic, transcriptomic and proteomic analysis of colorectal Cancer cell lines to identify novel biomarkers. PLoS One 10(12):e0144708. , DOI 10.1371/ journal.pone.0144708 Nam SK, Yun S, Koh J, Kwak Y, Seo AN, Park KU, Kim DW, Kang SB, Kim WH, Lee HS, 2016, BRAF, PIK3CA, and HER2 oncogenic alterations according to KRAS mutation status in advanced colorectal cancers with distant metastasis. PLoS One 11(3):e0151865. , DOI 10.1371/journal.pone.0151865 Hinrichs JW, van Blokland WT, Moons MJ, Radersma RD, Radersma-van Loon JH, de Voijs CM, Rappel SB, Koudijs MJ, Besselink NJ, Willems SM, de Weger RA, 2015, Comparison of next-generation sequencing and mutation-specific platforms in clinical practice. Am J Clin Pathol 143(4):573–578. https://doi. org/10.1309/ajcp40xetvyamjpy Harle A, Busser B, RouyerM, Harter V, Genin P, Leroux A, Merlin JL, 2013, Comparison of COBAS 4800 KRAS, TaqMan PCR and high resolution melting PCR assays for the detection of KRAS somatic mutations in formalin-fixed paraffin embedded colorectal carcinomas. Virchows Arch 462(3):329–335. , DOI 10. 1007/s00428-013-1380-x Little S, 2001, Amplification-refractory mutation system, ARMS, analysis of point mutations. Curr Protoc Hum Genet Chapter 9:Unit 9.8. , DOI 10.1002/0471142905.hg0908s07 Pichler M, Balic M, Stadelmeyer E, Ausch C, Wild M, Guelly C, Bauernhofer T, Samonigg H, Hoefler G, Dandachi N, 2009, Evaluation of high-resolution melting analysis as a diagnostic tool to detect the BRAF V600E mutation in colorectal tumors. J Mol Diagn 11(2):140–147. , DOI 10.2353/jmoldx.2009. 080100 Willmore-Payne C, Holden JA, Tripp S, Layfield LJ, 2005, Human malignant melanoma: detection of BRAF- and c-kit-activating mutations by high-resolution amplicon melting analysis. Hum Pathol 36(5):486–493. , DOI 10.1016/j.humpath.2005.03.015 Untergasser A, Nijveen H, Rao X, Bisseling T, Geurts R, Leunissen JA, 2007, Primer3Plus, an enhanced web interface to Primer3. Nucleic Acids Res 35(Web Server issue):W71–W74. https://doi. org/10.1093/nar/gkm306 Harle A, Filhine-Tresarrieu P, Husson M, Boidot R, Rouyer M, Dubois C, Leroux A, Merlin JL, 2016, Rare RAS mutations in metastatic colorectal Cancer detected during routine RAS genotyping using next generation sequencing. Target Oncol 11(3):363–370. , DOI 10.1007/s11523-015-0404-7 Landis JR, Koch GG, 1977, The measurement of observer agreement for categorical data. Biometrics 33(1):159–174 Deguchi Y, Okabe H, Oshima N, Hisamori S, Minamiguchi S, Muto M, Sakai Y, 2017, PTEN loss is associated with a poor response to trastuzumab in HER2-overexpressing gastroesophageal adenocarcinoma. Gastric Cancer 20(3):416–427. , DOI 10. 1007/s10120-016-0627-z Weigelt B, Warne PH, Downward J, 2011, PIK3CA mutation, but not PTEN loss of function, determines the sensitivity of breast cancer cells to mTOR inhibitory drugs. Oncogene 30(29):3222– 3233. , DOI 10.1038/onc.2011.42 Zhang X, Park JS, Park KH, KimKH, Jung M, Chung HC, Rha SY, Kim HS, 2015, PTEN deficiency as a predictive biomarker of resistance to HER2-targeted therapy in advanced gastric cancer. Oncology 88(2):76–85. , DOI 10.1159/000366426 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1117 EP 1123 DI 10.1007/s12253-018-0538-x PG 7 ER PT J AU Zhu, Y Sun, X Lin, J Zhang, T Liu, X Shen, X AF Zhu, Yi Sun, Xiangwei Lin, Ji Zhang, Teming Liu, Xin Shen, Xian TI Investigating Potential Molecular Mechanisms of Carcinogenesis and Genes as Biomarkers for Prognosis of Gastric Cancer Based on Integrated Bioinformatics Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; GEO data; lncRNAs; Integrated bioinformatics; Regulatory network ID Gastric cancer; GEO data; lncRNAs; Integrated bioinformatics; Regulatory network AB Gastric cancer, as the fifth most common malignancy worldwide, is a deadly disease afflicting nearly a million people. Researchers have devoted much to study the mechanisms of carcinogenesis and progression, but the exact information of tumor initiation and progression is remained largely unknown. Here, we hypothesized several differentially expressed genes and possible pathways by employing integrated bioinformatics analysis. We fully analyzed four gastric cancer-related microarray datasets to screen differentially expressed mRNAs (DEMs), miRNAs (DEMis) and lncRNAs (DELs). The functional enrichment analysis was deeply construed, PPI network and ceRNA regulatory network were constructed to investigate potential mechanisms of tumorigenesis and progression. Furthermore, survival analysis was performed to identify critical lncRNAs that may significantly affect pathogenesis of gastric cancer. QRT-PCR was applied to verify our result.We identified two hub subnetworks that may explain the progression, metastasis and poor prognosis of gastric cancer. Meanwhile, several potential significant lncRNAs were identified. In summary, we ascertained several significantly changed KEGG pathways in the tumor initiation and progression. We also hypothesized several lncRNAs that contribute to poor prognosis of gastric cancer via integrated bioinformatics, which deserve further investigation. C1 [Zhu, Yi] Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital, No. 109, Xueyuan West Road, Lucheng District, 325000 Wenzhou, Zhejiang, China. [Sun, Xiangwei] Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital, No. 109, Xueyuan West Road, Lucheng District, 325000 Wenzhou, Zhejiang, China. [Lin, Ji] Wenzhou Medical University, First Affiliated HospitalWenzhou, Zhejiang, China. [Zhang, Teming] Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital, No. 109, Xueyuan West Road, Lucheng District, 325000 Wenzhou, Zhejiang, China. [Liu, Xin] Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital, No. 109, Xueyuan West Road, Lucheng District, 325000 Wenzhou, Zhejiang, China. [Shen, Xian] Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital, No. 109, Xueyuan West Road, Lucheng District, 325000 Wenzhou, Zhejiang, China. RP Shen, X (reprint author), Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital, 325000 Wenzhou, China. EM 13968888872@126.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Russo A, Li P, Strong VE, 2017, Differences in the multimodal treatment of gastric cancer: east versus west. J Surg Oncol 115(5): 603–614 Tseng CW, Lin CC, Chen CN, Huang HC, Juan HF, 2011, Integrative network analysis reveals active microRNAs and their functions in gastric cancer. BMC Syst Biol 5:99 Song YX, Sun JX, Zhao JH, Yang YC, Shi JX,Wu ZH, Chen XW, Gao P,Miao ZF,Wang ZN, 2017, Non-coding RNAs participate in the regulatory network of CLDN4 via ceRNA mediated miRNA evasion. Nat Commun 8(1):289 Diboun I, Wernisch L, Orengo C, Koltzenburg M, 2006, Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma. BMC Genomics 7:252 da Huang W, Sherman BT, Lempicki RA, 2009, Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 37(1):1–13 Bader GD, Hogue CW, 2003, An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics 4:2 Brugge J, Hung MC, Mills GB, 2007, A new mutational AKTivation in the PI3K pathway. Cancer Cell 12(2):104–107 Cicenas J, 2008, The potential role of Akt phosphorylation in human cancers. Int J Biol Markers 23(1):1–9 Hao NB, Tang B,Wang GZ, Xie R, Hu CJ,Wang SM,Wu YY, Liu E, Xie X, Yang SM, 2015, Hepatocyte growth factor, HGF, upregulates heparanase expression via the PI3K/Akt/NF-kappaB signaling pathway for gastric cancer metastasis. Cancer Lett 361(1):57–66 Ge Y, Liu H, Qiu X, Ma G, Wang H, du M, Wang M, Zhao Q, Tao G, Chu H, Zhang Z, 2018, Genetic variants in PI3K/Akt/mTOR pathway genes contribute to gastric cancer risk. Gene 670:130–135 Xie X et al, 2013, Inhibition of the PI3K/Akt pathway increases the chemosensitivity of gastric cancer to vincristine. Oncol Rep 30(2): 773–782 Raman D, Baugher PJ, Thu YM, Richmond A, 2007, Role of chemokines in tumor growth. Cancer Lett 256(2):137–165 Lacalle RA et al, 2017, Chemokine receptor signaling and the hallmarks of Cancer. Int Rev Cell Mol Biol 331:181–244 Jiang C,Ma S, Hu R,Wang X, Li M, Tian F, JiangW, Zhu L, Bian Z, 2018, Effect of CXCR4 on apoptosis in osteosarcoma cells via the PI3K/Akt/NF-kappabeta signaling pathway. Cell Physiol Biochem 46(6):2250–2260 Zheng F, Zhang Z, Flamini V, JiangWG, Cui Y, 2017, The Axis of CXCR4/SDF-1 plays a role in Colon Cancer cell adhesion through regulation of the AKT and IGF1R Signalling pathways. Anticancer Res 37(8):4361–4369 Peng C, Zhou K, An S, Yang J, 2015, The effect of CCL19/CCR7 on the proliferation andmigration of cell in prostate cancer. Tumour Biol 36(1):329–335 Ma XJ, Dahiya S, Richardson E, Erlander M, Sgroi DC, 2009, Gene expression profiling of the tumor microenvironment during breast cancer progression. Breast Cancer Res 11(1):R7 Jensen BV, Johansen JS, Skovsgaard T, Brandt J, Teisner B, 2002, Extracellular matrix building marked by the N-terminal propeptide of procollagen type I reflect aggressiveness of recurrent breast cancer. Int J Cancer 98(4):582–589 MengQ, Yang BY, Liu B,Yang JX, SunY(2018, Long non-coding RNA SNHG6 promotes glioma tumorigenesis by sponging miR- 101-3p. Int J Biol Markers 33(2):148–155 Pei X,Wang X, Li H, 2018, LncRNA SNHG1 regulates the differentiation of Treg cells and affects the immune escape of breast cancer via regulating miR-448/IDO. Int J Biol Macromol 118(Pt A):24–30 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1125 EP 1133 DI 10.1007/s12253-018-0523-4 PG 9 ER PT J AU Bellolio, E Riquelme, I Riffo-Campos, LA Rueda, C Ferreccio, C Villaseca, M Brebi, P Munoz, S Araya, CJ AF Bellolio, Enrique Riquelme, Ismael Riffo-Campos, L Angela Rueda, Carlos Ferreccio, Catterina Villaseca, Miguel Brebi, Priscilla Munoz, Sergio Araya, Carlos Juan TI Assessment of Gastritis and Gastric Cancer Risk in the Chilean Population Using the OLGA System SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE The OLGA system; Helicobacter pylori infection; Gastric atrophy; Metaplasia; Gastric cancer; Chilean population ID The OLGA system; Helicobacter pylori infection; Gastric atrophy; Metaplasia; Gastric cancer; Chilean population AB Gastric cancer (GC) is the first cancer-related cause of death in Chile; however, no plan for GC early detection has been implemented in this country. The OLGA system characterizes gastritis from stages 0 to IV according to the risk of developing GC based on H. pylori infection, atrophy, metaplasia and GC. In this study, the performance of the OLGA system was evaluated in 485 Chilean patients receiving routine endoscopy to improve the detection of early GC or preneoplastic lesions. The results showed that OLGA scores, atrophy, metaplasia and GC increased significantly with age (p < 0.001). Conversely, H. pylori infection was higher in younger groups (p < 0.05). All gastric lesions were more frequent in men than women. The majority of patients with atrophy also had metaplasia (99%, p < 0.0001). Patients with H. pylori infection had more gastric atrophy and metaplasia than those without infection (p < 0.05). Of the 485 patients, 21 (4.3%) had GC, being 2.3 times more frequent among men than women and about 2/3 (14) were in OLGA stage ≥2. In addition, 19 (90%) GC patients had atrophy and 18 (85%) had metaplasia (p < 0.001). In conclusion, the OLGA system facilitated the evaluation of GC precursor lesions particularly in patients with an OLGA score > 2 between 45 and 56 years old, because this group showed atrophy and intestinal metaplasia more frequently. Therefore, biennial endoscopic surveillance of patients with an OLGA >2 can be an important health policy in Chile for diagnosing GC in its early stages and reducing mortality over the next two decades. C1 [Bellolio, Enrique] Universidad de La Frontera, School of Medicine, Department of Pathological Anatomy, Avenida Alemania 0458, 4810296 Temuco, Chile. [Riquelme, Ismael] Universidad Autonoma de Chile, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomedicas, Avenida Alemania 01090, 4810101 Temuco, Chile. [Riffo-Campos, L Angela] Universidad de La Frontera, School of Medicine, Department of Pathological Anatomy, Avenida Alemania 0458, 4810296 Temuco, Chile. [Rueda, Carlos] Gastroenterologist, Avenida Apoquindo 2880, 7550000 Santiago, Chile. [Ferreccio, Catterina] Pontificia Universidad Catolica de Chile, School of MedicineSantiago, Chile. [Villaseca, Miguel] Universidad de La Frontera, School of Medicine, Department of Pathological Anatomy, Avenida Alemania 0458, 4810296 Temuco, Chile. [Brebi, Priscilla] Universidad de La Frontera, School of Medicine, Department of Pathological Anatomy, Avenida Alemania 0458, 4810296 Temuco, Chile. [Munoz, Sergio] Universidad de La Frontera, School of Medicine, Department of Public Health, General Jose Miguel Carrera 228, 4781135 Temuco, Chile. [Araya, Carlos Juan] Universidad de La Frontera, School of Medicine, Department of Pathological Anatomy, Avenida Alemania 0458, 4810296 Temuco, Chile. RP Bellolio, E (reprint author), Universidad de La Frontera, School of Medicine, Department of Pathological Anatomy, 4810296 Temuco, Chile. EM enrique.bellolio@ufrontera.cl CR Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63:11–30. , DOI 10.3322/caac.21166 Brenner H, Rothenbacher D, Arndt V, 2009, Epidemiology of stomach cancer. Methods Mol Biol 472:467–477. , DOI 10.1007/978-1-60327-492-0_23 Heise K, Bertran E, Andia ME, Ferreccio C, 2009, Incidence and survival of stomach cancer in a high-risk population of Chile. World J Gastroenterol 15:1854–1862. , DOI 10.3748/wjg.15.1854 Ferreccio C, Rollan A, Harris PR et al, 2007, Gastric cancer is related to early helicobacter pylori infection in a high-prevalence country. Cancer Epidemiol Biomark Prev 16:662–667. https://doi. org/10.1158/1055-9965.EPI-06-0514 Department of Statistics and Health Information. Ministry of Health. Chile http://www.deis.cl/defunciones-y-mortalidad-porcausas/ Mickevicius A, Ignatavicius P, Markelis R, Parseliunas A, Butkute D, Kiudelis M, Endzinas Z, Maleckas A, Dambrauskas Z, 2014, Trends and results in treatment of gastric cancer over last two decades at single east European Centre: a cohort study. BMC Surg 14: 98. , DOI 10.1186/1471-2482-14-98 Riquelme I, Letelier P, Riffo-Campos A, Brebi P, Roa J, 2016, Emerging role of miRNAs in the drug resistance of gastric Cancer. Int J Mol Sci 17:424. , DOI 10.3390/ijms17030424 IARC, 2014, Helicobacter pylori Eradication as a Strategy for Preventing Gastric Cancer, IARC Working Group Reports Volume 8) Lim SM, Lim JY, Cho JY, 2014, Targeted therapy in gastric cancer: personalizing cancer treatment based on patient genome. World J Gastroenterol 20:2042–2050. , DOI 10.3748/wjg.v20.i8.2042 Riquelme I, Tapia O, Leal P et al, 2015, miR-101-2, miR-125b-2 and miR-451a act as potential tumor suppressors in gastric cancer through regulation of the PI3K/AKT/mTOR pathway. Cell Oncol, Dordr, 39(1):23–33. , DOI 10.1007/s13402-015-0247-3 Uemura N, Okamoto S, Yamamoto S, Matsumura N, Yamaguchi S, Yamakido M, Taniyama K, Sasaki N, Schlemper RJ, 2001, Helicobacter pylori infection and the development of gastric cancer. N Engl JMed 345:784–789. , DOI 10.1056/NEJMoa001999 Correa P, Piazuelo MB, 2011, Helicobacter pylori infection and gastric adenocarcinoma. US Gastroenterol Hepatol Rev 7:59–64 Correa P, 1992, Human gastric carcinogenesis: a multistep and multifactorial process–first American Cancer Society award lecture on Cancer epidemiology and prevention. Cancer Res 52:6735–6740 Pasechnikov V, Chukov S, Fedorov E, Kikuste I, Leja M, 2014, Gastric cancer: prevention, screening and early diagnosis. World J Gastroenterol 20:13842–13862. , DOI 10.3748/wjg.v20. i38.13842 Pasechnikov VD, Chukov SZ, Kotelevets SM, Mostovov AN, Mernova VP, Polyakova MB, 2005, Invasive and non-invasive diagnosis of helicobacter pylori-associated atrophic gastritis: a comparative study. Scand J Gastroenterol 40:297–301 Rugge M, 2007, Secondary prevention of gastric cancer. Gut 56: 1646–1647. , DOI 10.1136/gut.2007.133926 Zhou Y, Li H-Y, Zhang J-J, Chen XY, Ge ZZ, Li XB, 2016, Operative link on gastritis assessment stage is an appropriate predictor of early gastric cancer.World J Gastroenterol 22:3670–3678. , DOI 10.3748/wjg.v22.i13.3670 Rugge M, Correa P, Di Mario F et al, 2008, OLGA staging for gastritis: a tutorial. Dig Liver Dis 40:650–658. , DOI 10. 1016/j.dld.2008.02.030 RuggeM, Meggio A, Pennelli G, Piscioli F, Giacomelli L, de Pretis G, Graham DY, 2007, Gastritis staging in clinical practice: the OLGA staging system. Gut 56:631–636. , DOI 10.1136/ gut.2006.106666 Dixon MF, Genta RM, Yardley JH, Correa P, 1996, Classification and grading of gastritis. The updated Sydney system. International workshop on the histopathology of gastritis, Houston 1994. Am J Surg Pathol 20:1161–1181 Rugge M, Genta RM, 2005, Staging and grading of chronic gastritis. Hum Pathol 36:228–233. , DOI 10.1016/j.humpath. 2004.12.008 Ohata H, Kitauchi S, Yoshimura N, Mugitani K, Iwane M, Nakamura H, Yoshikawa A, Yanaoka K, Arii K, Tamai H, Shimizu Y, Takeshita T, Mohara O, Ichinose M, 2004, Progression of chronic atrophic gastritis associated withHelicobacter pylori infection increases risk of gastric cancer. Int J Cancer 109:138–143. , DOI 10.1002/ijc.11680 Cho S-J, Choi IJ, Kook M-C, Nam BH, Kim CG, Lee JY, Ryu KW, Kim YW, 2013, Staging of intestinal- and diffuse-type gastric cancers with the OLGA and OLGIM staging systems. Aliment Pharmacol Ther 38:1292–1302. , DOI 10.1111/apt.12515 Nam JH, Choi IJ, Kook M-C, Lee JY, Cho SJ, Nam SY, Kim CG, 2014, OLGA and OLGIM stage distribution according to age and Helicobacter pylori status in the Korean population. Helicobacter 19:81–89. , DOI 10.1111/hel.12112 Satoh K, Osawa H, Yoshizawa M, Nakano H, Hirasawa T, Kihira K, Sugano K, 2008, Assessment of atrophic gastritis using the OLGA system. Helicobacter 13:225–229. , DOI 10.1111/ j.1523-5378.2008.00599.x Capelle LG, de Vries AC, Haringsma J, ter Borg F, de Vries RA, Bruno MJ, van Dekken H, Meijer J, van Grieken NCT, Kuipers EJ, 2010, The staging of gastritis with the OLGA system by using intestinal metaplasia as an accurate alternative for atrophic gastritis. Gastrointest Endosc 71:1150–1158. , DOI 10.1016/j.gie. 2009.12.029 Ricci C, Holton J, Vaira D et al, 2007, Diagnosis of helicobacter pylori: invasive and non-invasive tests. Best Pract Res Clin Gastroenterol 21: 299–313. , DOI 10.1016/j.bpg.2006.11.002 Rollan A, Ferreccio C, Gederlini A, Serrano C, Torres J, Harris P, 2006, Non-invasive diagnosis of gastric mucosal atrophy in an asymptomatic population with high prevalence of gastric cancer. World J Gastroenterol 12:7172–7178. , DOI 10.3748/ WJG.V12.I44.7172 Namekata T, Miki K, Kimmey M et al, 2000, Chronic atrophic gastritis and helicobacter pylori infection among Japanese Americans in Seattle. Am J Epidemiol 151(8):820–830 Correa P, Haenszel W, Cuello C et al, 1975, A model for gastric cancer epidemiology. Lancet 306:58–60. , DOI 10.1016/ S0140-6736(75)90498-5 Tatsuta M, Iishi H, Nakaizumi A, Okuda S, Taniguchi H,Hiyama T, Tsukuma A, Oshima A, 1993, Fundal atrophic gastritis as a risk factor for gastric cancer. Int J Cancer 53:70–74. , DOI 10. 1002/ijc.2910530114 Sipponen P, Graham DY, 2007, Importance of atrophic gastritis in diagnostics and prevention of gastric cancer: application of plasma biomarkers. Scand J Gastroenterol 42:2–10. , DOI 10. 1080/00365520600863720 Graham DY, Nurgalieva ZZ, El-Zimaity HMT et al, 2006, Noninvasive versus histologic detection of gastric atrophy in a Hispanic population in North America. Clin Gastroenterol Hepatol 4:306–314. , DOI 10.1016/j.cgh.2005.11.003 Naylor GM, Gotoda T, Dixon M, Shimoda T, Gatta L, Owen R, Tompkins D, Axon A, 2006, Why does Japan have a high incidence of gastric cancer? Comparison of gastritis between UK and Japanese patients. Gut 55:1545–1552. , DOI 10.1136/gut. 2005.080358 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1135 EP 1142 DI 10.1007/s12253-018-0532-3 PG 8 ER PT J AU Zhang, Y Xu, J Li, D Wan, T Hu, Q AF Zhang, Yunmei Xu, Jieru Li, Dairong Wan, Tao Hu, Qianfang TI Immunocytochemistry Based on a Cell-Type-Specific Aptamer for Rapid Immunostaining of Adenocarcinoma Cells in Clinical Serosal Fluids SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Aptamer; Immunocytochemistry; Adenocarcinoma cells; Serosal fluid ID Aptamer; Immunocytochemistry; Adenocarcinoma cells; Serosal fluid AB All too often, conventional immunocytochemistry (ICC) via an antibody on cytological samples is limited to a few smears due to scant cellularity. To circumvent these limitations, this study employed a cell-type-specific aptamer as the core tool in ICC protocols for a timely and highly specific ICC diagnosis. S6, an aptamer against A549 lung carcinoma cells, was adopted instead of antibodies in this study for differentiating cancer cells in serosal fluids. Here, we developed three different strategies for discriminating the adenocarcinoma cells in effusion cytology specimens using the S6 aptamer in ICC. These strategies included a biotin-labeled S6 aptamer, an FAM-labeled S6 aptamer, and an activatable S6 aptamer. A total of 112 serosal fluid specimens with known diagnoses were evaluated by all three modes of use of the S6 aptamer. ICC procedures based on biotin-labeled or FAM-labeled S6 aptamers required time-consuming washing to avoid interference from nonspecific adsorption. ICC procedures based on an activatable S6 aptamer probe showed a weak fluorescence signal in the absence of target cells, but the procedures showed a strong fluorescence signal due to alteration of the conformation without any complicated washing steps, in the presence of targets. The specificity and sensitivity are higher in all three different ICC protocols based on the S6 aptamer than those for antibody protocols for differentiating adenocarcinoma cells in clinical effusion cytology. ICC based on cell-type-specific aptamers, instead of on a panel of a set of antibodies, is promising as an auxiliary method for the diagnosis of cancer. C1 [Zhang, Yunmei] Chongqing Medical University, The Nursing College, 400016 Chongqing, China. [Xu, Jieru] Chongqing Medical University, The First Affiliated Hospital, Department of Respiratory and Critical Care Medicine, No. 1 YouYi Road, Yuzhong District, 400016 Chongqing, China. [Li, Dairong] Chongqing Medical University, The First Affiliated Hospital, Department of Respiratory and Critical Care Medicine, No. 1 YouYi Road, Yuzhong District, 400016 Chongqing, China. [Wan, Tao] Chongqing Medical University, The First Affiliated Hospital, Department of Respiratory and Critical Care Medicine, No. 1 YouYi Road, Yuzhong District, 400016 Chongqing, China. [Hu, Qianfang] Chongqing Medical University, The First Affiliated Hospital, Department of Respiratory and Critical Care Medicine, No. 1 YouYi Road, Yuzhong District, 400016 Chongqing, China. RP Li, D (reprint author), Chongqing Medical University, The First Affiliated Hospital, Department of Respiratory and Critical Care Medicine, 400016 Chongqing, China. EM lidairong-e@yeah.net CR Wu GP, Zhang SS, Fang CQ, Liu SL, Wang EH, 2008, Immunocytochemical panel for distinguishing carcinoma cells from reactive mesothelial cells in pleural effusions. Cytopathology 19:212–217. , DOI 10.1111/j.1365-2303. 2008.00559.x Sriramoju B, Kanwar R, Veedu RN, Kanwar JR, 2015, Aptamer-targeted oligonucleotide theranostics: a smarter approach for brain delivery and the treatment of neurological diseases. Curr Top Med Chem 15:1115–1124. , DOI 10.2174/1568026615666150413153928 Sun H, Zu Y, 2015, A highlight of recent advances in aptamer technology and its application. Molecules 20:11959–11980. , DOI 10.3390/molecules200711959 Zhang L, Oho AUID, Wan S, Jiang Y, Wang Y, Fu T, Liu Q, Oho AUID, Cao Z, Qiu L, Tan W, Oho AUID, 2017, Molecular elucidation of disease biomarkers at the Interface of chemistry and biology. J Am Chem Soc 139:2532–2540. , DOI 10.1021/jacs. 6b10646 Lee KA, Ahn JY, Lee SH, Singh SS, Kim DG, Min J, Kim YH, 2015, Aptamer-based Sandwich assay and its clinical outlooks for detecting Lipocalin-2 in hepatocellular carcinoma, HCC). Sci Rep 5:10897. , DOI 10.1038/srep10897 Bukari BA, Citartan M, Ch'ng ES, Bilibana MP, Rozhdestvensky T, Tang TH, 2017, Aptahistochemistry in diagnostic pathology: technical scrutiny and feasibility. Histochem Cell Biol 147:545–553. , DOI 10.1007/s00418-017-1561-9 Kunii T, Ogura S, Mie M, Kobatake E, 2011, Selection of DNA aptamers recognizing small cell lung cancer using living cell- SELEX. Analyst 136:1310–1312. , DOI 10.1039/ c0an00962h Zhao Z, Xu L, Shi X, Tan W, Fang X, Shangguan D, 2009, Recognition of subtype non-small cell lung cancer by DNA aptamers selected from living cells. Analyst 134:1808–1814. , DOI 10.1039/b904476k Shangguan D, Li Y, Tang Z, Cao ZC, Chen HW, Mallikaratchy P, Sefah K, Yang CJ, TanW(2006, Aptamers evolved from live cells as effective molecular probes for cancer study. Proc Natl Acad Sci U S A 103:11838–11843. , DOI 10.1073/pnas. 0602615103 Shum KT, Zhou J, Rossi JJ, 2013, Nucleic acid aptamers as potential therapeutic and diagnostic agents for lymphoma. J Cancer Ther 4:872–890. , DOI 10.4236/jct.2013.44099 Van Simaeys D, Lopez-Colon D, Sefah K, Sutphen R, Jimenez E, Tan W, 2010, Study of the molecular recognition of aptamers selected through ovarian cancer cell-SELEX. PLoS One 5:e13770. , DOI 10.1371/journal.pone.0013770 Li WM, Bing T, Wei JY, Chen ZZ, Shangguan DH, Fang J, 2014, Cell-SELEX-based selection of aptamers that recognize distinct targets on metastatic colorectal cancer cells. Biomaterials 35: 6998–7007. , DOI 10.1016/j.biomaterials.2014.04.112 Zhao B, Wu P, Zhang H, Cai C, 2015, Designing activatable aptamer probes for simultaneous detection of multiple tumorrelated proteins in living cancer cells. Biosens Bioelectron 68: 763–770. , DOI 10.1016/j.bios.2015.02.004 Kim M, Kim DM, Kim KS, JungW, 0000–0003–4740-0004 AO , Kim DE, 2018, Applications of Cancer Cell-Specific Aptamers in Targeted Delivery of Anticancer Therapeutic Agents. Molecules 23: , DOI 10.3390/molecules23040830 Zeng Z, Zhang P, Zhao N, Sheehan AM, Tung CH, Chang CC, Zu Y, 2010, Using oligonucleotide aptamer probes for immunostaining of formalin-fixed and paraffin-embedded tissues. Mod Pathol 23:1553–1558. , DOI 10.1038/modpathol.2010.151 Shi H, Cui W, He X, Guo Q,Wang K, Ye X, Tang J, 2013)Whole cell-SELEX aptamers for highly specific fluorescence molecular imaging of carcinomas in vivo. PLoS One 8:e70476. https://doi. org/10.1371/journal.pone.0070476 Wang S, Kong H, Gong X, Zhang S, Zhang X, 2014, Multicolor imaging of cancer cells with fluorophore-tagged aptamers for single cell typing. Anal Chem 86:8261–8266. , DOI 10.1021/ ac501657g Shi H, He X, Wang K, Wu X, Ye X, Guo Q, Tan W, Qing Z, Yang X, Zhou B, 2011, Activatable aptamer probe for contrast-enhanced in vivo cancer imaging based on cell membrane protein-triggered conformation alteration. Proc Natl Acad Sci U S A 108:3900–3905. , DOI 10.1073/pnas.1016197108 Tang J, Shi H, He X, Lei Y, Guo Q, Wang K, Yan L, He D, 2016, Tumor cell-specific split aptamers: target-driven and temperature-controlled self-assembly on the living cell surface. Chem Commun, Camb, 52:1482–1485. , DOI 10.1039/C5CC08977H Yan L, Shi H, He X, Wang K, Tang J, Chen M, Ye X, Xu F, Lei Y, 2014, A versatile activatable fluorescence probing platform for cancer cells in vitro and in vivo based on self-assembled aptamer/ carbon nanotube ensembles. Anal Chem 86:9271–9277. https://doi. org/10.1021/ac5024149 Kubik MJ, Bovbel A, Goli H, Saremian J, Siddiqi A, Masood S, 2015, Diagnostic value and accuracy of imprint cytology evaluation during image-guided core needle biopsies: review of our experience at a large academic center. Diagn Cytopathol 43:773–779. , DOI 10.1002/dc.23300 Porfyridis I, Georgiadis G, Michael M, Frangopoulos F, Vogazianos P, Papadopoulos A, Kara P, Charalampous C, Georgiou A, 2016, Rapid on-site evaluation with the Hemacolor rapid staining method of medical thoracoscopy biopsy specimens for the management of pleural disease. Respirology 21:1106–1112. , DOI 10. 1111/resp.12799 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1143 EP 1152 DI 10.1007/s12253-018-0555-9 PG 10 ER PT J AU Zombori, T Cserni, G AF Zombori, Tamas Cserni, Gabor TI Patterns of Regression in Breast Cancer after Primary Systemic Treatment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Primary systemic therapy; Breast cancer; Regression pattern; Molecular subtypes ID Primary systemic therapy; Breast cancer; Regression pattern; Molecular subtypes AB Despite national guidelines, the evaluation of effects of primary systemic treatment (PST) in breast cancer is a complex challenge. Our aims were to evaluate the response patterns focusing on correlations of radiological and pathological tumor size, regression heterogeneity in different molecular subtypes, cellularity changes and the incidence of enlarged, multinucleated neoplastic cells related to therapy. Slides of pretreatment biopsies and resection specimens of consecutive cases were reevaluated focusing on heterogeneity of regression per whole slide, and 40x or 100x magnification fields. Alteration in cellularity and the presence of multinucleated tumor giant cells were noted. The correlation of pathological and radiological sizes and their alterations were analyzed by Spearman rank correlation. The present study included 106 tumors. A decrease in size (84.9%) and cellularity (76.4%) was noted in all molecular subtypes. Inhomogeneous regression was found in 45.3%, with minor inhomogeneity in the majority. Scatter pattern regression was seen only in 8 cases (7.5%). Significant correlations were found between the pathological and radiological sizes (p = 0.02), and between the alterations of cellularity and pathological and radiological size (p = 0.04; p = 0.03, respectively). Multinucleated tumor giant cells were noted in 17.9% (n = 19), nearly exclusively in cases treated with PST including taxanes. Regression inhomogeneity following PST is present in about half of the cases, and is not related to molecular subtypes. The evaluation of the maximum area of the tumor bed is recommended for the proper evaluation of regression. Multinucleated tumor giant cells are related to PST including taxane derivate, and may cause upgrading. C1 [Zombori, Tamas] University of Szeged, Department of Pathology, Allomas u. 1., 6725 Szeged, Hungary. [Cserni, Gabor] University of Szeged, Department of Pathology, Allomas u. 1., 6725 Szeged, Hungary. RP Zombori, T (reprint author), University of Szeged, Department of Pathology, 6725 Szeged, Hungary. EM zomtam@gmail.com CR Thompson AM, Moulder-Thompson SL, 2012, Neoadjuvant treatment of breast cancer. Ann Oncol 10:x231–x236. , DOI 10. 1093/annonc/mds324 Royal College of Pathologists of Australasia, 2012, Invasive Breast cancer structured reporting protocol, 2nd Edition, https://www. rcpa.edu.au/getattachment/7b70b3e5-5dca-403f-893e- 638815f487b1/Protocol-invasive-breast-cancer.aspx. Accessed 26 June 2018 Lambein K, Van de Vijver K, Faverly D, Colpaert C, 2011, Belgian guidelines for laboratory handling and pathology reporting of breast carcinoma after neoadjuvant therapy. Belg JMed Oncol 5:144–153 Arbeitsgemeinschaft Gynakologische Onkologie Studiengruppe, AGO, https://www.ago-online.de/fileadmin/downloads/leitlinien/ mamma/2018-03/EN/Gesamt_PDF_Englisch/Updated_ Guidelines_2018.pdf. Accessed 26 June 2018 NHS Breast Screening Program, 2005, Pathology reporting of breast disease: a joint document incorporating the third edition of the NHS breast screening programs guidelines for pathology reporting in breast cancer screening and the second edition of the Royal College of pathologists’ minimum dataset for breast cancer histopathology. NHSBSP publication no 58. NHS Cancer screening programs jointly with the Royal College of pathologists Integraal Kankercentrum Nederland, 2012, Beoordeling na neoadjuvante chemo- of endocriene therapie. http://www. oncoline.nl/breastcancer 2012. Accessed 26 June 2018 College of American Pathologists. Protocol for the examination of specimens from patients with invasive carcinoma of the breast. http://www.cap.org/ShowProperty?nodePath=/UCMCon/ Contribution%20Folders/WebContent/pdf/cp-breast-invasive- 16protocol-3300.pdf Accessed 36 June 2018 Cserni G, Kulka J, Francz M, Jaray B, Kalman E, Kovacs I, Krenacs T, Udvarhelyi N, Vass L, 2016, Pathological diagnosis, work-up and reporting of breast cancer. Recommendations of the 3rd Hungarian consensus conference on breast Cancer. Magy Onkol 60:209-28, Hungarian) Park CK, JungWH, Koo JS, 2016, Pathologic evaluation of breast cancer after neoadjuvant therapy. J Pathol TranslMed 50:173–180. , DOI 10.4132/jptm.2016.02.02 Provenzano E, Bossuyt V, Viale G, Cameron D, Badve S, Denkert C, MacGrogan G, Penault-Llorca F, Boughey J, Curigliano G, Dixon JM, Esserman L, Fastner G, Kuehn T, Peintinger F, von Minckwitz G, White J, Yang W, Symmans WF, 2015, Residual disease characterization working Group of the Breast International Group-North American Breast Cancer Group Collaboration. Standardization of pathologic evaluation and reporting of postneoadjuvant specimens in clinical trials of breast cancer: recommendations from an international working group. Mod Pathol 28:1185–1201. , DOI 10.1038/modpathol.2015.74 Tot T, 2012, The role of large-format histopathology in assessing subgross morphological prognostic parameters: a single institution report of 1000 consecutive breast cancer cases. Int J Breast Cancer 2012:395415. , DOI 10.1155/2012/395415 U.S. Food and Drug Administration, 2014, Guidance for Industry: Pathological complete response in neoadjuvant treatment of highrisk early-stage breast cancer: use as an endpoint to support accelerated approval. http://www.fda.gov/downloads/Drugs/ GuidanceComplianceRegulatoryInformation/Guidances/ UCM305501.pdf Accessed 26 June 2018 AJCC, 2016, Cancer staging manual, 8th edn. Springer, Chicago Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, Bonnefoi H, Cameron D, Gianni L, Valagussa P, Swain SM, Prowell T, Loibl S, Wickerham DL, Bogaerts J, Baselga J, Perou C, Blumenthal G, Blohmer J, Mamounas EP, Bergh J, Semiglazov V, Justice R, Eidtmann H, Paik S, Piccart M, Sridhara R, Fasching PA, Slaets L, Tang S, Gerber B, Geyer CE Jr, Pazdur R, Ditsch N, Rastogi P, Eiermann W, von Minckwitz G, 2014, Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet 384:164–172. https://doi. org/10.1016/S0140-6736(13)62422-8 von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, Gerber B, EiermannW, Hilfrich J, Huober J, Jackisch C,Kaufmann M,Konecny GE, Denkert C,Nekljudova V,Mehta K, Loibl S, 2012, Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol 30:1796–1804. , DOI 10.1200/JCO.2011.38.8595 Mamounas EP, Anderson SJ, Dignam JJ, Bear HD, Julian TB, Geyer CE Jr, Taghian A, Wickerham DL, Wolmark N, 2012, Predictors of locoregional recurrence after neoadjuvant chemotherapy: results from combined analysis of National Surgical Adjuvant Breast and bowel project B-18 and B-27. J Clin Oncol 30:3960– 3966. , DOI 10.1200/JCO.2011.40.8369 Pinder SE, Provenzano E, Earl H, Ellis IO, 2007, Laboratory handling and histology reporting of breast specimens from patients who have received neoadjuvant chemotherapy. Histopathology 50:409– 417. , DOI 10.1111/j.1365-2559.2006.02419.x Denkert C, Schickling O, von Minckwitz G, 2006, Preoperative chemotherapy in breast cancer and the development of new predictive markers. Verh Dtsch Ges Pathol 114-23(German):90 Sataloff DM, Mason BA, Prestipino AJ, Seinige UL, Lieber CP, Baloch Z, 1995, Pathologic response to induction chemotherapy in locally advanced carcinoma of the breast: a determinant of outcome. J Am Coll Surg 180:297–306 Wells CA, Amendoeira I, Bellocq JP, Bianchi S, Boecker W, Borisch B, Bruun Rasmussen B, Callagy GM, Chmielik E, Cordoba A, Cserni G, Decker T, DeGaetano J, Drijkoningen M, Ellis IO, Faverly DR, Foschini MP, Frkovic-Grazio S, Grabau D, Heikkila P, Iacovou E, Jacquemier J, Kaya H, Kulka J, Lacerda M, Liepniece-Karele I, Martinez-Penuela J, Quinn CM, Rank F, Regitnig P, Reiner-Concin A, Sapino A, Tot T, Van Diest PJ, Varga Z, Wesseling J, Zolota V, Zozaya-Alvarez E., 2012, Pathology update. Quality assurance guidelines for pathology. In: European guidelines for quality assurance in breast cancer screening and diagnosis. Fourth edition, Supplements. Perry N, Broeders M, de Wolf C, Tornberg S, Holland R, von Karsa L, eds.). European Commission, Office for Official Publications of the European Union, Luxembourg, pp. 73–120 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, Senn HJ, 2013, Panel members, 2013, personalizing the treatment of women with early breast cancer: highlights of the St Gallen international expert consensus on the primary therapy of early breast cancer. Ann Oncol 24:2206–2223. https://doi. org/10.1093/annonc/mdt303 Giordano SH, Temin S, Chandarlapaty S, Crews JR, Esteva FJ, Kirshner JJ, Krop IE, Levinson J, Lin NU, Modi S, Patt DA, Perlmutter J, Ramakrishna N, Winer EP, Davidson NE, 2018, Systemic therapy for patients with advanced human epidermal growth factor receptor 2-positive breast cancer: ASCO clinical practice guideline update. J Clin Oncol [Epub ahead of print]. , DOI 10.1200/JCO.2018.79.2697 Kaufmann M, von Minckwitz G, Smith R, Valero V, Gianni L, Eiermann W, Howell A, Costa SD, Beuzeboc P, Untch M, Blohmer JU, Sinn HP, Sittek R, Souchon R, Tulusan AH, Volm T, Senn HJ, 2003, International expert panel on the use of primary, preoperative, systemic treatment of operable breast cancer: review and recommendations. J Clin Oncol 21:2600–2608. , DOI 10.1200/JCO.2003.01.136 Kaufmann M, Hortobagyi GN, Goldhirsch A, Scholl S, Makris A, Valagussa P, Blohmer JU, Eiermann W, Jackesz R, Jonat W, Lebeau A, Loibl S, Miller W, Seeber S, Semiglazov V, Smith R, Souchon R, Stearns V, Untch M, von Minckwitz G, 2006, Recommendations from an international expert panel on the use of neoadjuvant, primary, systemic treatment of operable breast cancer: an update. J Clin Oncol 24:1940–1949. https:// doi.org/10.1200/JCO.2005.02.6187 von Minckwitz G, Blohmer JU, Raab G, Lohr A, Gerber B, Heinrich G, Eidtmann H, Kaufmann M, Hilfrich J, Jackisch C, Zuna I, Costa SD, German Breast Group, 2005, In vivo chemosensitivity-adapted preoperative chemotherapy in patients with early-stage breast cancer: the GEPARTRIO pilot study. Ann Oncol 16:56–63. , DOI 10.1093/annonc/mdi001 Chollet P, Amat S, Cure H, de Latour M, Le Bouedec G, Mouret- Reynier MA, Ferriere JP, Achard JL, Dauplat J, Penault-Llorca F, 2002, Prognostic significance of a complete pathological response after induction chemotherapy in operable breast cancer. Br J Cancer 86:1041–1046. , DOI 10.1038/sj.bjc.6600210 Marinovich ML, Macaskill P, Irwig L, Sardanelli F, Mamounas E, von Minckwitz G, Guarneri V, Partridge SC, Wright FC, Choi JH, Bhattacharyya M, Martincich L, Yeh E, Londero V, Houssami N, 2015, Agreement between MRI and pathologic breast tumor size after neoadjuvant chemotherapy, and comparison with alternative tests: individual patient data meta-analysis. BMC Cancer 15:662. , DOI 10.1186/s12885-015-1664-4 Lee SC, Grant E, Sheth P, Garcia AA, Desai B, Ji L, Groshen S, Hwang D, Yamashita M, Hovanessian-Larsen L, 2017, Accuracy of contrast-enhanced ultrasound compared with magnetic resonance imaging in assessing the tumor response after neoadjuvant chemotherapy for breast Cancer. J Ultrasound Med 36:901–911. , DOI 10.7863/ultra.16.05060 Rajan R, Poniecka A, Smith TL, Yang Y, Frye D, Pusztai L, Fiterman DJ, Gal-Gombos E, Whitman G, Rouzier R, Green M, Kuerer H, Buzdar AU, Hortobagyi GN, Symmans WF, 2004, Change in tumor cellularity of breast carcinoma after neoadjuvant chemotherapy as a variable in the pathologic assessment of response. Cancer 100:1365–1373. , DOI 10.1002/cncr.20134 Abrial SC, Penault-Llorca F, Delva R, Bougnoux P, Leduc B, Mouret-Reynier MA, Mery-Mignard D, Bleuse JP, Dauplat J, Cure H, Chollet P, 2005, High prognostic significance of residual disease after neoadjuvant chemotherapy: a retrospective study in 710 patients with operable breast cancer. Breast Cancer Res Treat 94:255–263. , DOI 10.1007/s10549-005-9008-8 Diaz J, Stead L, Shapiro N, Newell R, Loudig O, Lo Y, Sparano J, Fineberg S, 2013)Mitotic counts in breast cancer after neoadjuvant systemic chemotherapy and development of metastatic disease. Breast Cancer Res Treat 138:91–97. , DOI 10.1007/ s10549-013-2411-7 Boughey JC, Peintinger F, Meric-Bernstam F, Perry AC, Hunt KK, Babiera GV, Singletary SE, Bedrosian I, Lucci A, Buzdar AU, Pusztai L, Kuerer HM, 2006, Impact of preoperative versus postoperative chemotherapy on the extent and number of surgical procedures in patients treated in randomized clinical trials for breast cancer. Ann Surg 244:464–470. , DOI 10.1097/01.sla. 0000234897.38950.5c Carey LA, Metzger R, Dees EC, Collichio F, Sartor CI, Ollila DW, Klauber-DeMore N, Halle J, Sawyer L, Moore DT, Graham ML, 2005)American joint committee on Cancer tumor-node-metastasis stage after neoadjuvant chemotherapy and breast cancer outcome. J Natl Cancer Inst 97:1137–1142. , DOI 10.1093/jnci/dji206 Neoadjuvant response-guided treatment of HER2 positive breast cancer, PREDIX HER2). ClinicalTrials.gov Identifier: NCT02568839 https://clinicaltrials.gov/ct2/show/NCT02568839?cond=breast+ neoadjuvant+treatment&rank=31 Accessed 2 August 2018 Efficacy and safety of Cabazitaxel versus weekly Paclitaxel as neoadjuvant treatment in patients with triple negative or luminal B/ HER2 normal BC, GENEVIEVE). ClinicalTrials.gov Identifier: NCT01779479 https://clinicaltrials.gov/ct2/show/NCT01779479? cond=breast+neoadjuvant+treatment&rank=69 Accessed 2 August 2018 Neoadjuvant Letrozole plus Metformin vs Letrozole plus Placebo for ER-positive postmenopausal breast cancer ClinicalTrials.gov Identifier: NCT01589367 https://clinicaltrials.gov/ct2/show/ NCT01589367?cond=breast+neoadjuvant+treatment&rank=97 Accessed 2 August 2018 Neoadjuvant Tamoxifen in locally advanced breast cancer in a low/ middle income country. ClinicalTrials.gov Identifier: NCT02806544 https://clinicaltrials.gov/ct2/show/NCT02806544? cond=breast+neoadjuvant+treatment&draw=2&rank=101 Accessed 2 August 2018 Leary A, Evans A, Johnston SR, A'Hern R, Bliss JM, Sahoo R, Detre S, Haynes BP, Hills M, Harper-Wynne C, Bundred N, Coombes G, Smith I, Dowsett M, 2015, Antiproliferative effect of Lapatinib in HER2-positive and HER2-negative/HER3-high breast cancer: results of the presurgical randomized MAPLE trial, CRUKE/06/039). Clin Cancer Res 21:2932–2940. , DOI 10.1158/1078-0432.CCR-14-1428 Honkoop AH, Pinedo HM, De Jong JS, Verheul HM, Linn SC, Hoekman K,Wagstaff J, van Diest PJ, 1997, Effects of chemotherapy on pathologic and biologic characteristics of locally advanced breast cancer. Am J Clin Pathol 107:211–218 Sharkey FE, Addington SL, Fowler LJ, Page CP, Cruz AB, 1996, Effects of preoperative chemotherapy on the morphology of resectable breast carcinoma. Mod Pathol 9:893–900 Valent A, Penault-Llorca F, Cayre A, Kroemer G, 2013, Change in HER2, ERBB2, gene status after taxane-based chemotherapy for breast cancer: polyploidization can lead to diagnostic pitfalls with potential impact for clinical management. Cancer Genet 206:37–41. , DOI 10.1016/j.cancergen.2012.12.001 Fukada I, Araki K, Kobayashi K, Shibayama T, Takahashi S, Gomi N, Kokubu Y, Oikado K, Horii R, Akiyama F, Iwase T, Ohno S, Hatake K, Sata N, Ito Y, 2018, Pattern of tumor shrinkage during neoadjuvant chemotherapy is associated with prognosis in lowgrade luminal early breast cancer. Radiology 286:49–57. https:// doi.org/10.1148/radiol.2017161548 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1153 EP 1161 DI 10.1007/s12253-018-0557-7 PG 9 ER PT J AU Carvalho, JM Laranjo, M Abrantes, MA Casalta-Lopes, J Sarmento-Santos, D Costa, T Serambeque, B Almeida, N Goncalves, T Mamede, C Encarnacao, J Oliveira, R Paiva, A de Carvalho, R Botelho, F Oliveira, C AF Carvalho, Joao Maria Laranjo, Mafalda Abrantes, Margarida Ana Casalta-Lopes, Joao Sarmento-Santos, Daniela Costa, Tania Serambeque, Beatriz Almeida, Nuno Goncalves, Telmo Mamede, Catarina Encarnacao, Joao Oliveira, Rui Paiva, Artur de Carvalho, Rui Botelho, Filomena Oliveira, Carlos TI Endometrial Cancer Spheres Show Cancer Stem Cells Phenotype and Preference for Oxidative Metabolism SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial neoplasms; Neoplastic stem cells; Glucose metabolism ID Endometrial neoplasms; Neoplastic stem cells; Glucose metabolism AB This study aimed to characterize endometrial cancer regarding cancer stem cells (CSC) markers, regulatory and differentiation pathways, tumorigenicity and glucose metabolism. Endometrial cancer cell line ECC1 was submitted to sphere forming protocols. The first spheres generation (ES1) was cultured in adherent conditions (G1). This procedure was repeated and was obtained generations of spheres (ES1, ES2 and ES3) and spheres-derived cells in adherent conditions (G1, G2 and G3). Populations were characterized regarding CD133, CD24, CD44, aldehyde dehydrogenase (ALDH), hormonal receptors, HER2, P53 and β- catenin, fluorine-18 fluorodeoxyglucose ([18F]FDG) uptake and metabolism by NMR spectroscopy. An heterotopic model evaluated differential tumor growth. The spheres self-renewal was higher in ES3. The putative CSC markers CD133, CD44 and ALDH expression were higher in spheres. The expression of estrogen receptor (ER)α and P53 decreased in spheres, ERβ and progesterone receptor had no significant changes and β-catenin showed a tendency to increase. There was a higher 18F-FDG uptake in spheres, which also showed a lower lactate production and an oxidative cytosol status. The tumorigenesis in vivo showed an earlier growth of tumours derived from ES3. Endometrial spheres presented self-renewal and differentiation capacity, expressed CSC markers and an undifferentiated phenotype, showing preference for oxidative metabolism. C1 [Carvalho, Joao Maria] University of Coimbra, Universitary Clinic of GynecologyCoimbra, Portugal. [Laranjo, Mafalda] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Abrantes, Margarida Ana] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Casalta-Lopes, Joao] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Sarmento-Santos, Daniela] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Costa, Tania] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Serambeque, Beatriz] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Almeida, Nuno] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Goncalves, Telmo] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Mamede, Catarina] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Encarnacao, Joao] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Oliveira, Rui] Coimbra Hospital and University Centre, Clinical Pathology DepartmentCoimbra, Portugal. [Paiva, Artur] Coimbra Hospital and Universitary Centre, Flow Cytometry UnitCoimbra, Portugal. [de Carvalho, Rui] University of Coimbra, Faculty of Science and Technology, Department of Life Sciences, Centre for Functional EcologyCoimbra, Portugal. [Botelho, Filomena] University of Coimbra, Faculty of Medicine, Biophysics InstituteCoimbra, Portugal. [Oliveira, Carlos] University of Coimbra, Faculty of Medicine, CIMAGOCoimbra, Portugal. RP Carvalho, JM (reprint author), University of Coimbra, Universitary Clinic of Gynecology, Coimbra, Portugal. EM mariajoaosflcarvalho@gmail.com CR Allegra A, Alonci A, Penna G, Innao V, Gerace D, Rotondo F, Musolino C, 2014, The cancer stem cell hypothesis: a guide to potential molecular targets. Cancer Investig 32:470–495. , DOI 10.3109/07357907.2014.958231 Chan RWS, Schwab KE, Gargett CE, 2004, Clonogenicity of human endometrial epithelial and stromal cells. Biol Reprod 70:1738– 1750. , DOI 10.1095/biolreprod.103.024109 Kato K, Yoshimoto M, Kato K, Adachi S, Yamayoshi A, Arima T, Asanoma K, Kyo S, Nakahata T, Wake N, 2007, Characterization of side-population cells in human normal endometrium. Hum Reprod 22:1214–1223. , DOI 10.1093/humrep/del514 Chan RWS, Gargett CE, 2006, Identification of label-retaining cells in mouse endometrium. Stem Cells, Dayton, Ohio, 24:1529–1538. , DOI 10.1634/stemcells.2005-0411 Carvalho MJ, LaranjoM, Abrantes AM, Torgal I, Botelho MF, Oliveira CF, 2015, Clinical translation for endometrial cancer stem cells hypothesis. Cancer Metastasis Rev 34:401– 416. , DOI 10.1007/s10555-015-9574-0 Hubbard SA, Friel AM, Kumar B et al, 2009, Evidence for cancer stem cells in human endometrial carcinoma. Cancer Res 69:8241– 8248. , DOI 10.1158/0008-5472.CAN-08-4808 Kato K, Takao T, Kuboyama A, Tanaka Y, Ohgami T, Yamaguchi S, Adachi S, Yoneda T, Ueoka Y, Kato K, Hayashi S, Asanoma K, Wake N, 2010, Endometrial cancer sidepopulation cells show prominent migration and have a potential to differentiate into the mesenchymal cell lineage. Am J Pathol 176:381–392. , DOI 10.2353/ajpath.2010. 090056 Kusunoki S, Kato K, Tabu K, Inagaki T, Okabe H, Kaneda H, Suga S, Terao Y, Taga T, Takeda S, 2013, The inhibitory effect of salinomycin on the proliferation, migration and invasion of human endometrial cancer stem-like cells. Gynecol Oncol 129:598–605. , DOI 10.1016/j.ygyno.2013.03.005 GotteM, Greve B, Kelsch R et al, 2011, The adult stem cell marker Musashi-1 modulates endometrial carcinoma cell cycle progression and apoptosis via Notch-1 and p21 WAF1/CIP1. Int J Cancer 129: 2042–2049. , DOI 10.1002/ijc.25856 Zhou X, Zhou Y-P, Huang G-R et al, 2011, Expression of the stem cell marker, Nanog, in human endometrial adenocarcinoma. Int J Gynecol Pathol 33:262–270. , DOI 10.1097/PGP. 0b013e3182055a1f Rutella S, Bonanno G, Procoli A et al, 2009, Cells with characteristics of cancer stem/progenitor cells express the CD133 antigen in human endometrial tumors. Clin Cancer Res 15:4299–4311. https:// doi.org/10.1158/1078-0432.CCR-08-1883 Friel AM, Zhang L, Curley MD et al, 2010, Epigenetic regulation of CD133 and tumorigenicity of CD133 positive and negative endometrial cancer cells. Reprod Biol Endocrinol 8:147. , DOI 10.1186/1477-7827-8-147 Rahadiani N, Ikeda J, Mamat S,Matsuzaki S, Ueda Y, Umehara R, Tian T, Wang Y, Enomoto T, Kimura T, Aozasa K, Morii E, 2011, Expression of aldehyde dehydrogenase 1, ALDH1, in endometrioid adenocarcinoma and its clinical implications. Cancer Sci 102:903–908. , DOI 10.1111/j.1349-7006. 2011.01864.x Tang DG, 2012, Understanding cancer stem cell heterogeneity and plasticity. Cell Res 22:457–472. , DOI 10.1038/cr.2012.13 Dontu G, Abdallah WM, Foley JM, Jackson KW, Clarke MF, Kawamura MJ, Wicha MS, 2003, In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev 17:1253–1270. , DOI 10.1101/gad. 1061803 Ponti D, Costa A, Zaffaroni N, Pratesi G, Petrangolini G, Coradini D, Pilotti S, PierottiMA, Daidone MG(2005, Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res 65:5506–5511. , DOI 10.1158/ 0008-5472.CAN-05-0626 Wilson H, Huelsmeyer M, Chun R, Young KM, Friedrichs K, Argyle DJ, 2008, Isolation and characterisation of cancer stem cells from canine osteosarcoma. Vet J 175:69–75. , DOI 10. 1016/j.tvjl.2007.07.025 Franken N a P, Rodermond HM, Stap J et al, 2006, Clonogenic assay of cells in vitro. Nat Protoc 1:2315–2319. , DOI 10. 1038/nprot.2006.339 Santos K, Laranjo M, Abrantes AM, Brito AF, Goncalves C, Sarmento Ribeiro AB, Botelho MF, Soares MIL, Oliveira ASR, Pinho e MeloTMVD(2014, Targeting triple-negative breast cancer cells with 6,7-bis(hydroxymethyl)-1H,3H-pyrrolo[1,2-c]thiazoles. Eur J Med Chem 79:273–281. , DOI 10.1016/j.ejmech. 2014.04.008 Abrantes AM, Serra MES, Goncalves AC, Rio J, Oliveiros B, Laranjo M, Rocha-Gonsalves AM, Sarmento-Ribeiro AB, Botelho MF, 2010, Hypoxia-induced redox alterations and their correlation with 99mTc-MIBI and 99mTc-HL-91 uptake in colon cancer cells. Nucl Med Biol 37:125–132. , DOI 10.1016/j. nucmedbio.2009.11.001 Carvalho RA, Rodrigues TB, Zhao P et al, 2004, A13C isotopomer kinetic analysis of cardiac metabolism: influence of altered cytosolic redox and [Ca2+]o.AmJ Phys Heart Circ Phys 287:H889–H895. , DOI 10.1152/ajpheart.00976.2003 Sherry AD, Jeffrey FMH, Malloy CR, 2004, Analytical solutions for 13C isotopomer analysis of complex metabolic conditions: substrate oxidation, multiple pyruvate cycles, and gluconeogenesis. Metab Eng 6:12–24. , DOI 10.1016/j.ymben.2003.10.007 Visvader JE, Lindeman GJ, 2008, Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 8:755–768. , DOI 10.1038/nrc2499 Nieto-Estevez V, Pignatelli J, Arauzo-Bravo MJ, Hurtado-Chong A, Vicario-Abejon C, 2013, A global transcriptome analysis reveals molecular hallmarks of neural stem cell death, survival, and differentiation in response to partial FGF-2 and EGF deprivation. PLoS One 8:e53594. , DOI 10.1371/journal.pone.0053594 Weiswald L-B, Bellet D, Dangles-Marie V, 2015, Spherical Cancer models in tumor biology. Neoplasia 17:1–15. , DOI 10. 1016/j.neo.2014.12.004 Prasetyanti PR, Zimberlin C, De Sousa E, Melo F, Medema JP, 2013, Isolation and propagation of colon cancer stem cells. Methods Mol Biol, Clifton, NJ, 1035:247–259. , DOI 10. 1007/978-1-62703-508-8_21 Bortolomai I, Canevari S, Facetti I, de Cecco L, Castellano G, Zacchetti A, Alison MR, Miotti S, 2010, Tumor initiating cells: development and critical characterization of a model derived from the A431 carcinoma cell line forming spheres in suspension. Cell Cycle 9:1194–1206. , DOI 10.4161/cc.9.6.11108 Liu Y, Nenutil R, Appleyard MV, Murray K, BoylanM, Thompson AM, Coates PJ, 2014, Lack of correlation of stem cell markers in breast cancer stem cells. Br J Cancer 110:2063–2071. https://doi. org/10.1038/bjc.2014.105 Chung L, Tang S, Wu Y et al, 2015, Galectin-3 augments tumor initiating property and tumorigenicity of lung cancer through interaction with β-catenin. Oncotarget 6:4936–4952. , DOI 10. 18632/oncotarget.3210 Kryczek I, Liu S, Roh M, Vatan L, Szeliga W, Wei S, Banerjee M, Mao Y, Kotarski J,Wicha MS, Liu R, ZouW(2012, Expression of aldehyde dehydrogenase and CD133 defines ovarian cancer stem cells. Int J Cancer 130:29–39. , DOI 10.1002/ijc.25967 Tirino V, Desiderio V, Paino F, de Rosa A, Papaccio F, la Noce M, Laino L, de Francesco F, Papaccio G, 2013, Cancer stem cells in solid tumors: an overview and new approaches for their isolation and characterization. FASEB J 27:13–24. , DOI 10.1096/fj. 12-218222 Hartomo T, Van Huyen Pham T, Yamamoto N et al, 2014, Involvement of aldehyde dehydrogenase 1A2 in the regulation of cancer stem cell properties in neuroblastoma. Int J Oncol 46:1089– 1098. , DOI 10.3892/ijo.2014.2801 Rivlin N, Koifman G, Rotter V, 2014, P53 orchestrates between Normal differentiation and Cancer. Semin Cancer Biol 32:10–17. , DOI 10.1016/j.semcancer.2013.12.006 Cui J, Li P, Liu X et al, 2015, Abnormal expression of the notch and Wnt/β-catenin signaling pathways in stem-like ALDHhiCD44+ cells correlates highly with Ki-67 expression in breast cancer. Oncol Lett 9:1600–1606. , DOI 10.3892/ol.2015.2942 Wang Y, van der Zee M, Fodde R, Blok LJ, 2010, Wnt/Β-catenin and sex hormone signaling in endometrial homeostasis and cancer. Oncotarget 1:674–684. , DOI 10.18632/oncotarget.101007 Gargett CE, Chan RWS, Schwab KE, 2008, Hormone and growth factor signaling in endometrial renewal: role of stem/progenitor cells. Mol Cell Endocrinol 288:22–29. , DOI 10.1016/j. mce.2008.02.026 Hapangama DK, Kamal aM, Bulmer JN, 2014, Estrogen receptor : the guardian of the endometrium. Hum Reprod Update 21:174– 193. , DOI 10.1093/humupd/dmu053 R a G, Gillies RJ, 2004, Why do cancers have high aerobic glycolysis? Nat Rev Cancer 4:891–899. , DOI 10.1038/nrc1478 Morfouace M, Lalier L, Bahut M, Bonnamain V, Naveilhan P, Guette C, Oliver L, Gueguen N, Reynier P, Vallette FM, 2012, Comparison of spheroids formed by rat glioma stem cells and neural stem cells reveals differences in glucose metabolism and promising therapeutic applications. J Biol Chem 287:33664–33674. , DOI 10.1074/jbc.M111. 320028 Palorini R, Votta G, Balestrieri C, Monestiroli A, Olivieri S, Vento R, Chiaradonna F, 2014, Energy metabolism characterization of a novel cancer stem cell-like line 3AB-OS. J Cell Biochem 115:368– 379. , DOI 10.1002/jcb.24671 Jang H, Yang J, Lee E, Cheong J-H, 2015, Metabolism in embryonic and cancer stemness. Arch Pharm Res 38:381–388. , DOI 10.1007/s12272-015-0558-y NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1163 EP 1174 DI 10.1007/s12253-018-0535-0 PG 12 ER PT J AU Helbig, G Chromik, K Wozniczka, K Kopinska, JA Boral, K Dworaczek, M Koclega, A Armatys, A Panz-Klapuch, M Markiewicz, M AF Helbig, Grzegorz Chromik, Karolina Wozniczka, Krzysztof Kopinska, J Anna Boral, Kinga Dworaczek, Martyna Koclega, Anna Armatys, Anna Panz-Klapuch, Marta Markiewicz, Miroslaw TI Real Life Data on Efficacy and Safety of Azacitidine Therapy for Myelodysplastic Syndrome, Chronic Myelomonocytic Leukemia and Acute Myeloid Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Azacitidine; Myelodysplastic syndrome; Chronic myelomonocytic leukemia; Acute myeloid leukemia; Results ID Azacitidine; Myelodysplastic syndrome; Chronic myelomonocytic leukemia; Acute myeloid leukemia; Results AB The administration of azacitidine (AZA) was found to be more effective than conventional care regimen (CCR) in patients with higher-risk myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia (CMML) and acute myeloid leukemia (AML) with lower blast count. We designed a study to determine efficacy and safety of AZA therapy in "real life" patients with MDS, CMML and AML. The study included 83 patients (65% male) with a median age at diagnosis of 68 years. 43 patients were diagnosed with higher-risk MDS, 30 had AML and 10- CMML. Median AZA dose was comparable between treated groups. AZA dose reduction was required for 44% of MDS, 17% of AML and 25% of CMML patients. Complete remission (CR) was achieved in 14% of MDS, 7% of AML and 10% of CMML patients. Overall response rate was following: 27% for MDS, 20% for AML and 20% for CMML. Estimated OS at 12 months was 75% for MDS, 60% for AML and 75% for CMML. Median follow-up for MDS/AML/CMML from AZA initiation to last follow-up was 9.0, 9.4 and 9.4 months, respectively. The most common toxicity of AZA therapy was myelosuppression and infections. AZA treatment was effective in a limited number of patients with acceptable safety profile. C1 [Helbig, Grzegorz] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street, 25, 40-032 Katowice, Poland. [Chromik, Karolina] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street, 25, 40-032 Katowice, Poland. [Wozniczka, Krzysztof] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street, 25, 40-032 Katowice, Poland. [Kopinska, J Anna] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street, 25, 40-032 Katowice, Poland. [Boral, Kinga] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street, 25, 40-032 Katowice, Poland. [Dworaczek, Martyna] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street, 25, 40-032 Katowice, Poland. [Koclega, Anna] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street, 25, 40-032 Katowice, Poland. [Armatys, Anna] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street, 25, 40-032 Katowice, Poland. [Panz-Klapuch, Marta] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street, 25, 40-032 Katowice, Poland. [Markiewicz, Miroslaw] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Dabrowski street, 25, 40-032 Katowice, Poland. RP Helbig, G (reprint author), Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, 40-032 Katowice, Poland. EM ghelbig@o2.pl CR Stresemann C, Lyko F, 2008, Modes of action of the DNA methyltransferase inhibitors azacitidine and decitabine. Int J Cancer 123: 8–13 Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A, Schoch R, Gattermann N, Sanz G, List A, Gore SD, Seymour JF, Bennett JM, Byrd J, Backstrom J, Zimmerman L, McKenzie D, Beach C, Silverman LR, 2009, International Vidaza High-Risk MDS Survival Study Group. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomized, open-label, phase III study. Lancet Oncol 10:223–232 Dombret H, Seymour JF, Butrym A, Wierzbowska A, Selleslag D, Jang JH, Kumar R, Cavenagh J, Schuh AC, Candoni A, Recher C, Sandhu I, Bernal del Castillo T, al-Ali HK,Martinelli G, Falantes J, Noppeney R, StoneRM, MindenMD, McIntyre H, Songer S, Lucy LM, Beach CL, Dohner H, 2015, International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood 126:291–299 AlfonsoA, Montalban-BravoG, Takahashi K, Jabbour EJ, Kadia T, Ravandi F, Cortes J, Estrov Z, Borthakur G, Pemmaraju N, Konopleva M, Bueso-Ramos C, Pierce S, Kantarjian H, Garcia- Manero G, 2017 Mar. 28, Natural history of chronic myelomonocytic leukemia treated with hypomethylating agents. Am J Hematol 92:599–606. , DOI 10.1002/ajh.24735 [epub ahead of print]. Cabrero M, Jabbour E, Ravandi F, Bohannan Z, Pierce S, Kantarjian HM, Garcia-Manero G, 2015, Discontinuation of hypomethylating agent therapy in patients with myelodysplastic syndromes or acute myelogenous leukemia in complete remission or partial response: retrospective analysis of survival after longterm follow-up. Leuk Res 39:520–524 Santini V, Fenaux P, Ghulam M et al, 2010, Management and supportive care measures for adverse events in patients with myelodysplastic syndromes treated with azacitidine. Eur J Haematol 85:130138 Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, Harris NL, le Beau MM, Hellstrom-Lindberg E, Tefferi A, Bloomfield CD, 2009, The 2008 revision of the WHO classification of myeloid neoplasms and acute leukemia, Rationale and important changes. Blood 114:937–951 Cheson BD, Greenberg PL, Bennet JM et al, 2006, Clinical application and proposal for modification of the international working group, IWG, response criteria in myelodysplasia. Blood 108:419–425 Silverman L, Demakos E, Peterson B et al, 2002, Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol 20:2429–2440 Navada SC, Silveraman LR, 2017, Safety and efficacy of azacitidine in elderly patients with intermediate to high-risk myelodysplastic syndromes. Ther Adv Hematol 8:21–27 Ozbalak M, Cetiner M, Bekoz H, Atesoglu EB, Ar C, Salihoglu A, Tuzuner N, Ferhanoglu B, 2012, Azacitidine has limited activity in Breal life^ patients withMDS andAML: a single Centre experience. Hematol Oncol 30:76–81 Isabella C, Avanzini P, Merli F. 5-Azacitidine in patients with myelodysplasia and acute myeloid leukemia: a single centre experience. Blood., ASH Annual Meeting Abstracts, 2010; 116: Abstract 4969. Beguin Y, Selleslag D, Mers S et al, 2015, Safety and efficacy of azacitidine in Belgian patients with high-risk myelodysplastic syndromes, acute myeloid leukaemia, or chronic myelomonocytic leukaemia: results of a real-life, non-interventional post-marketing survey. Acta Clin Belg 70:34–43 Voso MT, Niscola P, Piciocchi A et al, 2015, Standard dose and prolonged administration of azacitidine are associated with improved efficacy in a real-world group of patients with myelodysplastic syndrome or low blast count acute myeloid leukemia. Eur J Haematol 96:344–351 DiNardo CD, Pratz KW, LetaiA et al, 2018, Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a nonrandomised, open-label, phase 1b study. Lancet Oncol 19:216–228 Trubiano JA, Dickinson M, Thursky KA, Spelman T, Seymour JF, Slavin MA, Worth LJ, 2017, Incidence, etiology and timing of infections following azacitidine therapy for myelodysplastic syndromes. Leuk Lymphoma 58:2379–2386 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1175 EP 1180 DI 10.1007/s12253-018-00574-0 PG 6 ER PT J AU Zheng, J Xu, T Feng, Ch Zhang, Y AF Zheng, Jing Xu, Tingting Feng, Chen Zhang, Ying TI MiRNA-195-5p Functions as a Tumor Suppressor and a Predictive of Poor Prognosis in Non-small Cell Lung Cancer by Directly Targeting CIAPIN1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; miR-195-5p; Overall survival; Prognosis; CIAPIN1 ID Non-small cell lung cancer; miR-195-5p; Overall survival; Prognosis; CIAPIN1 AB Accumulating evidence suggests that microRNAs (miRNAs) has been proven to be a critical regulator in the tumor progression, of which miR-195-5p was reported to function as tumor suppressor in prostate cancer and oral squamous cell carcinoma. However, studies on the clinical significance and biological function of miR-195-5p in non-small cell lung cancer (NSCLC) were still unavailable. Here, we reported that the expression of miR-195-5p was decreased in NSCLC tissues and cell lines. Downregulation of miR-195-5p was significantly associated with TNM stage, tumor size and lymph node metastasis. The Kaplan-Meier survival analysis demonstrated that the survival time of NSCLC patients with high expression of miR-195-5p was longer than those with low expression during the 5-year follow up period (p = 0.0410). COX regression analysis indicated that miR-195-5p expression was an independent prognostic indicator for the survival of NSCLC patients (HR = 2.45, 95% CI: 1.53–4.63; p = 0.007). Results of functional analyses revealed that overexpression of miR-195-5p in A549 cells inhibited cell proliferation, induced cell cycle G0/G1 phase arrest and apoptosis using MTT and flow cytometry analysis. Furthermore, bioinformatics and luciferase reporter assays demonstrated that cytokine-induced apoptosis inhibitor 1 (CIAPIN1), an antiapoptotic molecule was a direct target of miR-195-5p in NSCLC cells. Meta-analysis based on Oncomine database showed CIAPIN1 was significantly up-regulated in human lung cancer tissues. Consistently, knockdown of CIAPIN1 phenocopied the inhibitory effects of miR-195-5p overexpression in NSCLC cell function. These findings suggest that miR-195-5p could be used as a potential prognostic predictor and tumor suppressor in NSCLC. C1 [Zheng, Jing] Taizhou Hospital, Department of Respiratory Medicine, 381 East Zhongshan Road, Jiaojiang District, 318000 Taizhou, Zhejiang, China. [Xu, Tingting] Taizhou Hospital, Department of Respiratory Medicine, 381 East Zhongshan Road, Jiaojiang District, 318000 Taizhou, Zhejiang, China. [Feng, Chen] Taizhou Hospital, Department of Respiratory Medicine, 381 East Zhongshan Road, Jiaojiang District, 318000 Taizhou, Zhejiang, China. [Zhang, Ying] Taizhou Hospital, Department of Respiratory Medicine, 381 East Zhongshan Road, Jiaojiang District, 318000 Taizhou, Zhejiang, China. RP Xu, T (reprint author), Taizhou Hospital, Department of Respiratory Medicine, 318000 Taizhou, China. EM Xu_tingting09@126.com CR Garcia-Campelo R, Bernabe R, CoboM, Corral J, Coves J, Domine M, Nadal E, Rodriguez-Abreu D, Vinolas N, Massuti B, 2015, SEOM clinical guidelines for the treatment of non-small cell lung cancer, NSCLC, 2015. Clin Transl Oncol 17(12):1020–1029. , DOI 10.1007/s12094-015-1455-z Ettinger DS, Wood DE, Akerley W, Bazhenova LA, Borghaei H, Camidge DR, Cheney RT, Chirieac LR, D'Amico TA, Demmy TL, Dilling TJ, Dobelbower MC, Govindan R, Grannis FWJr, Horn L, JahanTM, Komaki R, Krug LM, Lackner RP, Lanuti M, Lilenbaum R, Lin J, LooBWJr,Martins R, OttersonGA, Patel JD, PistersKM, Reckamp K, Riely GJ, Rohren E, Schild SE, Shapiro TA, Swanson SJ, Tauer K, Yang SC, Gregory K, Hughes M, National comprehensive cancer n, 2015, Non-small cell lung Cancer, version 6.2015. J Natl Compr Cancer Netw 13(5):515–524 Wang L, Qu J, Zhou L, Liao F, Wang J, 2018, MicroRNA-373 inhibits cell proliferation and invasion via targeting BRF2 in human non-small cell lung Cancer A549 cell line. Cancer Res Treat : official journal of Korean Cancer Association 50(3):936–949. https:// doi.org/10.4143/crt.2017.302 Guo H, Ingolia NT, Weissman JS, Bartel DP, 2010, Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature 466(7308):835–840. , DOI 10.1038/nature09267 Xie B, Ding Q, Han H, Wu D, 2013, miRCancer: a microRNAcancer association database constructed by text mining on literature. Bioinformatics 29(5):638–644. , DOI 10.1093/ bioinformatics/btt014 Zhang JG, Wang JJ, Zhao F, Liu Q, Jiang K, Yang GH, 2010, MicroRNA-21, miR-21, represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer, NSCLC). Clin Chim Acta 411(11–12):846–852. , DOI 10.1016/j.cca.2010.02.074 Wang R, Wang ZX, Yang JS, Pan X, De W, Chen LB, 2011, MicroRNA-451 functions as a tumor suppressor in human nonsmall cell lung cancer by targeting ras-related protein 14, RAB14). Oncogene 30(23):2644–2658. , DOI 10.1038/ onc.2010.642 Crawford M, Brawner E, Batte K, Yu L, HunterMG, Otterson GA, Nuovo G, Marsh CB, Nana-Sinkam SP, 2008, MicroRNA-126 inhibits invasion in non-small cell lung carcinoma cell lines. Biochem Biophys Res Commun 373(4):607–612. , DOI 10.1016/j.bbrc.2008.06.090 Kumarswamy R, Mudduluru G, Ceppi P, Muppala S, Kozlowski M, Niklinski J, Papotti M, Allgayer H, 2012, MicroRNA-30a inhibits epithelial-to-mesenchymal transition by targeting Snai1 and is downregulated in non-small cell lung cancer. Int J Cancer 130(9): 2044–2053. , DOI 10.1002/ijc.26218 LiY, Jiang Q, Xia N,Yang H,Hu C, 2012, Decreased expression of microRNA-375 in nonsmall cell lung cancer and its clinical significance. J Int Med Res 40(5):1662–1669. , DOI 10.1177/ 030006051204000505 Zhang HH, Zhang ZY, Che CL, Mei YF, Shi YZ, 2013, Array analysis for potential biomarker of gemcitabine identification in non-small cell lung cancer cell lines. Int J Clin Exp Pathol 6(9): 1734–1746 Luo Q,Wei C, Li X, Li J, Chen L, Huang Y, Song H, Li D, Fang L, 2014, MicroRNA-195-5p is a potential diagnostic and therapeutic target for breast cancer. Oncol Rep 31(3):1096–1102. https://doi. org/10.3892/or.2014.2971 Yu X, Zhang Y, Cavazos D, Ma X, Zhao Z, Du L, Pertsemlidis A, 2018, miR-195 targets cyclin D3 and survivin to modulate the tumorigenesis of non-small cell lung cancer. Cell Death Dis 9(2): 193. , DOI 10.1038/s41419-017-0219-9 Marzuk O, Peretz G, Bakhrat A, Abdu U, 2013, Drosophila CIAPIN1 homologue is required for follicle cell proliferation and survival. Dev Dyn 242(6):731–737. , DOI 10.1002/dvdy. 23951 Shibayama H, Takai E, Matsumura I, Kouno M, Morii E, Kitamura Y, Takeda J, Kanakura Y, 2004, Identification of a cytokineinduced antiapoptotic molecule anamorsin essential for definitive hematopoiesis. J Exp Med 199(4):581–592. , DOI 10. 1084/jem.20031858 Shi H, Zhou Y, Liu H, Chen C, Li S, Li N, Li X, ZhangX, Zhang H, Wang W, Zhao Q, 2010, Expression of CIAPIN1 in human colorectal cancer and its correlation with prognosis. BMC Cancer 10: 477. , DOI 10.1186/1471-2407-10-477 Hao Z, Li X, Qiao T, Du R, Hong L, Fan D, 2006, CIAPIN1 confers multidrug resistance by upregulating the expression of MDR-1 and MRP-1 in gastric cancer cells. Cancer Biol Ther 5(3):261–266 He L,Wang H, Jin H, Guo C, Xie H, Yan K, Li X, Shen Q, Qiao T, Chen G, Chai N, Zhao L, Dong Q, Zheng Y, Liu J, Fan D, 2009, CIAPIN1 inhibits the growth and proliferation of clear cell renal cell carcinoma. Cancer Lett 276(1):88–94. , DOI 10.1016/ j.canlet.2008.10.044 Wang Y, Qi HW, Li XH, Chen XX, Liu J, 2008, CIAPIN1 expression in human lung cancer tissues and inhibitory effects of the gene on human pulmonary carcinomaNCI-H446 cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 24(5):434–437 Selamat SA, Chung BS, Girard L, Wei Z, Ying Z, Campan M, Siegmund KD, Koss MN, Hagen JA, Wan LL, 2012, Genomescale analysis of DNA methylation in lung adenocarcinoma and integration with mRNA expression. Genome Res 22(7):1197–1211 Garber ME, Troyanskaya OG, Schluens K, Petersen S, Thaesler Z, Pacynagengelbach M, Van dRM, Rosen GD, Perou CM, Whyte RI, 2001, Diversity of gene expression in adenocarcinoma of the lung. Proc Natl Acad Sci U S A 98(24):13784–13789 Hou J, Aerts J, Den HB, Van IW, Den BM, Riegman P, Van dLC, Van dSP, Foekens JA, Hoogsteden HC, 2010, Gene expression-based classification of non-small cell lung carcinomas and survival prediction. PLoS One 5(4):e10312 Landi MT, Dracheva T, Rotunno M, Figueroa JD, Liu H, Dasgupta A, Mann FE, Fukuoka J, Hames M, Bergen AW, 2008, Gene expression signature of cigarette smoking and its role in lung adenocarcinoma development and survival. PLoS One 3(2):e1651 Okayama H, Kohno T, Ishii Y, Shimada Y, Shiraishi K, Iwakawa R, Furuta K, Tsuta K, Shibata T, Yamamoto S, 2012, Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas. Cancer Res 72(1):100–111 Jacqueline WP, Lin CH, Liang SC, Lin CJ, Chen KC, Wu YC, Chang CW, Su LJ, Shih-Lan H, Chen CH, 2007, Selection of DDX5 as a novel internal control for Q-RT-PCR from microarray data using a block bootstrap re-sampling scheme. BMC Genomics 8(1):140 Wachi S, Yoneda K, Wu R, 2005, Interactome-transcriptome analysis reveals the high centrality of genes differentially expressed in lung cancer tissues. Bioinformatics 21(23):4205–4208 Chai L, Kang XJ, Sun ZZ, Zeng MF, Yu SR, Ding Y, Liang JQ, Li TT, Zhao J, 2018)MiR-497-5p, miR-195-5p andmiR-455-3p function as tumor suppressors by targeting hTERT in melanoma A375 cells. Cancer Manag Res 10:989–1003. , DOI 10.2147/ CMAR.S163335 Wang T, Ren Y, Liu R, Ma J, Shi Y, Zhang L, Bu R, 2017, miR- 195-5p suppresses the proliferation,migration, and invasion of Oral squamous cell carcinoma by targeting TRIM14. Biomed Res Int 2017:7378148. , DOI 10.1155/2017/7378148 Luo Q, Zhang Z, Dai Z, Basnet S, Li S, Xu B, Ge H, 2016, Tumorsuppressive microRNA-195-5p regulates cell growth and inhibits cell cycle by targeting cyclin dependent kinase 8 in colon cancer. Am J Transl Res 8(5):2088–2096 Xu H, HuYW, Zhao JY,Hu XM, Li SF,WangYC, Gao JJ, ShaYH, Kang CM, Lin L, Huang C, Zhao JJ, Zheng L, Wang Q, 2015, MicroRNA-195-5p acts as an anti-oncogene by targeting PHF19 in hepatocellular carcinoma. Oncol Rep 34(1):175–182. https://doi. org/10.3892/or.2015.3957 Nana-Sinkam SP, Croce CM, 2013, Clinical applications for microRNAs in cancer. Clin Pharmacol Ther 93(1):98–104. https:// doi.org/10.1038/clpt.2012.192 Rupaimoole R, Slack FJ, 2017, MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov 16(3):203–222. , DOI 10.1038/nrd.2016.246 Li X, Pan Y, Fan R, Jin H, Han S, Liu J, Wu K, Fan D, 2008, Adenovirus-delivered CIAPIN1 small interfering RNA inhibits HCC growth in vitro and in vivo. Carcinogenesis 29(8):1587– 1593. , DOI 10.1093/carcin/bgn052 Yan K, He LJ, ChengW, Ji ZZ, Zhao BX, Hui XL, Cao SS, Chen B, He L, Liang SH,Miao Y, 2009, Inhibiting gastric cancer-associated angiogenesis by CIAPIN1 siRNA. Cancer Biol Ther 8(11):1058– 1063 Nymoen DA, Holth A, Hetland Falkenthal TE, Trope CG, Davidson B, 2015, CIAPIN1 and ABCA13 are markers of poor survival in metastatic ovarian serous carcinoma.Mol Cancer 14:44. , DOI 10.1186/s12943-015-0317-1 Chen X, Li X, Chen J, Zheng P, Huang S, Ouyang X, 2012, Overexpression of CIAPIN1 inhibited pancreatic cancer cell proliferation and was associated with good prognosis in pancreatic cancer. Cancer Gene Ther 19(8):538–544. , DOI 10.1038/cgt. 2012.28 Zheng X, Zhao Y, Wang X, Li Y, Wang R, Jiang Y, Gong T, Li M, Sun L, Hong L, Li X, Liang J, Luo G, Jin B, Yang J, Zhang H, Fan D, 2010, Decreased expression of CIAPIN1 is correlated with poor prognosis in patients with esophageal squamous cell carcinoma. Dig Dis Sci 55(12):3408–3414. , DOI 10.1007/s10620- 010-1212-7 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1181 EP 1190 DI 10.1007/s12253-018-0552-z PG 10 ER PT J AU Molnar, Cs Molnar, S Bedekovics, J Mokanszki, A Gyory, F Nagy, E Mehes, G AF Molnar, Csaba Molnar, Sarolta Bedekovics, Judit Mokanszki, Attila Gyory, Ferenc Nagy, Endre Mehes, Gabor TI Thyroid Carcinoma Coexisting with Hashimoto’s Thyreoiditis: Clinicopathological and Molecular Characteristics Clue up Pathogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thyroid; Carcinoma; Hashimoto thyreoiditis; Clinicopathology; Molecular pathology ID Thyroid; Carcinoma; Hashimoto thyreoiditis; Clinicopathology; Molecular pathology AB Thyroid cancer (TC) coexisting with Hashimoto’s thyroiditis (HT) presents with several characteristic features including multifocality and lower clinical stages compared to de novo carcinomas but its exact biology is still not understood. We reexamined clinico-pathological and molecular correlations between Hashimoto’s thyroditis and papillary thyroid cancer. A total of 262 patients with TC was evaluated who underwent thyroidectomy at the Surgical Department of the University of Debrecen. Clinical data, histology and molecular data were evaluated. Our cohort included 43 patients (16.4%) with (5 male, 38 female) and 219 (83.6%) patients without coexisting HT (48 male, 171 female). Hashimoto’s thyroiditis related thyroid cancer presented predominantly (93.0% of the cases) with the papillary histological type. Multifocality was observed more frequently with coexisting HT (16/40; 40.0%) compared to cases uninvolved (45/190; 23.7%)(p = 0.034). In contrast, lymphatic metastasis (pN1) with a significantly reduced frequency in patients with HT (4/11; 36.4%) then without HT (34/41 pN1; 82.9%)(p = 0.002). BRAF V600E mutation could be demonstrated at significantly lower rates in cases of PTC + HT (32.1 vs 60.7%, p < 0.005). High incidence, multifocality and papillary morphology strongly support a causal relation between TC and preexisting Hashimoto’s thyroiditis, the latter to be considered as a preneoplastic condition promoting thyroid carcinogenesis. C1 [Molnar, Csaba] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4042 Debrecen, Hungary. [Molnar, Sarolta] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4042 Debrecen, Hungary. [Bedekovics, Judit] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4042 Debrecen, Hungary. [Mokanszki, Attila] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4042 Debrecen, Hungary. [Gyory, Ferenc] University of Debrecen, Department of Surgery and Operative TechniquesDebrecen, Hungary. [Nagy, Endre] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, H-4042 Debrecen, Hungary. RP Molnar, Cs (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, H-4042 Debrecen, Hungary. EM molnar.csaba@med.unideb.hu CR Dong YH, Fu DG, 2014, Autoimmune thyroid disease: mechanism, genetics and current knowledge. Eur Rev Med Pharmacol Sci 18(23):3611–3618 Ajjan RA, Weetman AP, 2015, The pathogenesis of Hashimoto’s thyreoiditis: further developments in our understanding. Horm Metab Res 47(10):702–710 Pyzik A, Grywalska E, Matyjaszek-Matuszek B, Rolinski J, 2015, Immune disorders in Hashimoto’s thyroiditis: what do we know so far? J Immunol Res 2015:1–8. , DOI 10.1155/2015/ 979167 Sipos JA, Mazzaferri EL, 2010, Thyroid cancer epidemiology and prognostic variables. Clin Oncol 22(6):395–404 Lee JC, Sidhu SB, 2013, Papillary thyroid cancer: the most common endocrine malignancy. Endocrinology Today 2(3):15–20 Cardis E, Howe G, Ron E, Bebeshko V, Bogdanova T, Bouville A, Carr Z, Chumak V, Davis S, Demidchik Y, Drozdovitch V, Gentner N, Gudzenko N, Hatch M, Ivanov V, Jacob P, Kapitonova E, Kenigsberg Y, Kesminiene A, Kopecky KJ, Kryuchkov V, Loos A, Pinchera A, Reiners C, Repacholi M, Shibata Y, Shore RE, Thomas G, Tirmarche M, Yamashita S, Zvonova I, 2006, Cancer consequences of the Chernobyl accident: 20 years on. J Radiol Prot 26(2):127–140 Ron E, Lubin JH, Shore RE et al, 1995, Thyroid cancer after exposure to external radiation: a pooled analysis of seven studies. Radiat Res 178(2):43–60 Nikiforov YE, 2006, Radiation-induced thyroid cancer: what we have learned from Chernobyl. Endocr Pathol 17(4):307–317 Dailey ME, Lindsay S, Skahen R, 1955, Relation of thyroid neoplasms to Hashimoto disease of thyroid gland. Arch Surg 70:291– 297 Chesky VE, Hellwig CA, Welch JV, 1962, Cancer of the thyroid associated with Hashimoto’s disease: an analysis of forty-eight cases. Am Surg 28:678–685 Meier DW, Woolner LB, Beahrs OH et al, 1959, Parenchymal findings in thyroidal carcinoma: pathologic study of 256 cases. J Clin Endocrinol Metab 19(1):162–171 Shands WC, 1960, Carcinoma of the thyroid in association with struma lymphomatosa, Hashimoto’s disease). Ann Surg 151(5): 675–681 Crile G, Hazard JB, 1962, Incidence of cancer in struma lymphomatosa. Surg Gynecol Obstet 115:101–103 Crile G, 1978, Struma lymphomatosa and carcinoma of the thyroid. Surg Gynecol Obstet 147(3):350–352 Holm LE, Blomgren HL, Lowhagen T, 1985, Cancer risks in patients with chronic lymphocytic thyroiditis. N Engl J Med 312(10): 601–604 Kim KW, Park YJ, Kim EH, Park SY, Park DJ, Ahn SH, Park DJ, Jang HC, Cho BY, 2011, Elevated risk of papillary thyroid cancer in Korean patients with Hashimoto’s thyroiditis. Head Neck 33(5): 691–695 Consorti F, Loponte M, Milazzo F, Potasso L, Antonaci A, 2010, Risk of malignancy from thyroid nodular disease as an element of clinical management of patients with Hashimoto’s thyroiditis. Eur Surg Res 45(3–4):333–337 Fiore E, Rago T, Scutari M, Ugolini C, Proietti A, di Coscio G, Provenzale MA, Berti P, Grasso L, Mariotti S, Pinchera A, Vitti P, 2009, Papillary thyroid cancer, although strongly associated with lymphocytic infiltration on histology, is only weakly predicted by serum thyroid auto-antibodies in patients with nodular thyroid diseases. J Endocrinol Investig 32(4):344–351 Kurukahvecioglu O, Taneri F, Yuksel O, Aydin A, Tezel E, Onuk E, 2007, Total thyreoidectomy for the treatment of Hashimoto’s thyroiditis coexisting with papillary thyroid carcinoma. Adv Ther 24(3):510–516 Cipolla C, Sandonato L, Graceffa G, Fricano S, Torcivia A,Vieni S, Latteri S, Latteri MA, 2005, Hashimoto thyroiditis coexistent with papillary thyroid carcinoma. Am Surg 71(10):874–878 Tamimi DM, 2002, The association between chronic lymphocytic thyroiditis and thyroid tumors. Int J Surg Pathol 10(2):141–146 Okayashu I, FujiwaraM, Hara Yet al, 1995, Association of chronic lymphocytic thyroiditis and thyroid papillary carcinoma. A study of surgical cases among Japanese, and white and African Americans. Cancer 76(11):2312–2318 Lee JH, Kim Y, Choi JW, Kim YS, 2013, The association between papillary thyroid carcinoma and histologically proven Hashimoto’s thyroiditis: a meta-analysis. Eur J Endocrinol 168(3):343–349 Mazokopakis EE, Tzortzinis AA, Dalieraki-Ott EI, Tsartsalis A, Syros P, Karefilakis C, Papadomanolaki M, Starakis I, 2010, Coexistence of Hashimoto’s thyroiditis with papillary thyroid carcinoma. A retrospective study. Hormones 9(4):312–317 Huang BY, Hseuh C, Chao TC, Lin KJ, Lin JD, 2011, Welldifferentiated thyroid carcinoma with concomitant Hashimoto’s thyroiditis present with less aggressive clinical stage and low recurrence. Endocr Pathol 22(3):144–149 Repplinger D, Bargren A, Zhang YW, Adler JT, Haymart M, Chen H, 2008, Is Hashimoto’s thyroiditis a risk factor for papillary thyroid cancer? J Surg Res 150(1):49–52 Van Savell H, Hughes SM, Bower C et al, 2004, Lymphocytic infiltration in pediatric thyroid carcinomas. Pediatr Dev Pathol 7(5):487–492 Harach HR, Escalante DA, Day ES, 2002, Thyroid cancer and thyroiditis in Salta, Argentina: a 40-yr study in relation to iodine prophylaxis. Endocr Pathol 13(3):175–181 Loh KC, Greenspan FS, Dong F, Miller TR, Yeo PPB, 1999, Influence of lymphocytic thyroiditis on the prognostic outcome of patients with papillary thyroid carcinoma. J Clin EndocrinolMetab 84(2):458–463 Kebebew E, Treseler PA, Ituarte PH et al, 2001, Coexisting chronic lymphocytic thyroiditis and papillary thyroid cancer revisited. World J Surg 25(5):632–637 Soares P, Trovisco V, Rocha AS, Feijao T, Rebocho AP, Fonseca E, Vieira de Castro I, Cameselle-Teijeiro J, Cardoso-Oliveira M, Sobrinho-Simoes M, 2004, BRAF mutations typical of papillary thyroid carcinoma are most frequently detected in undifferentiated than is insular and insular-like poorly differentiated carcinomas. Virchows Arch 444:572–576 Nikiforov YE, Nikiforova MN, 2011, Molecular genetics and diagnosis of thyroid cancer. Nat Rev Endocrinol 7:569–580 Nikiforov YE, 2002, RET/PTC rearrangement in thyroid tumors. Endocr Pathol 13(1):3–16 Li F, Chen G, Sheng C, Gusdon AM, Huang Y, Lv Z, Xu H, Xing M, Qu S, 2015, BRAFV600E mutation in papillary thyroid microcarcinoma: a meta-analysis. Endocr Relat Cancer 22(2): 159–168 Liu X, Yan K, Lin X, Zhao L, An W, Wang C, Liu X, 2014, The association between BRAF V600E mutation and pathological features in PTC. Eur Arch Otorhinolaryngol 271:3041–3052 Ma H, Yan J, Zhang C et al, 2014, Expression of papillary thyroid carcinoma-associated molecular markers and their significance in follicular epithelial dysplasia with papillary thyroid carcinoma-like nuclear alterations in Hashimoto’s thyroiditis. Int J Clin Exp Pathol. 15;7, 11): 7999–8007 Kim SJ, Myong JP, Jee HG, Chai YJ, Choi JY, Min HS, Lee KE, Youn YK, 2016, Combined effect of Hashimoto’s thyroiditis and BRAF(V600E, mutation status on aggressiveness in papillary thyroid cancer. Head Neck 38(1):95–101 Fakhruddin N, JabbourM, NowyMet al, 2017, BRAF and NRAS mutations in papillary thyroid carcinoma and concordance in BRAF mutations between primary and corresponding lymph node metastases. Sci Report 7:4666. , DOI 10.1038/s41598-017- 04948-3 Chen H, Luthra R, Routbort MJ et al, 2017, Molecular profile of advanced thyroid carcinomas by next-generation sequencing: characterizing tumors beyond diagnosis for targeted therapy. Mol Cancer Ther 17(7):1575–1584 Wirtschafter A, Schmidt R, Rosen D, Kundu N, Santoro M, Fusco A, Multhaupt H, Atkins JP, Rosen MR, Keane WM, Rothstein JL, 1997, Expression of RET/PTC fusion gene as a marker for papillary carcinoma in Hashimoto’s thyroiditis. Laryngoscope 107(1): 95–100 Larson SD, Jackson LN, Riall TS, Uchida T, Thomas RP, Qiu S, Evers BM, 2007, Increased incidence of well-differentiated thyroid cancer associated with Hashimoto thyroiditis and the role of the PI3K/Akt pathway. J Am Coll Surg 204(5):764–773 Unger P, Ewart M, Wang BY, Gan L, Kohtz DS, Burstein DE, 2003, Expression of p63 in papillary thyroid carcinoma and in Hashimoto’s thyroiditis: a pathobiologic link? Hum Pathol 34(8): 764–769 Szekanecz Z, Szekanecz E, Bako G, Shoenfeld Y, 2011, Malignancies in autoimmune rheumatic diseases – a mini review. Gerontology 57(1):3–10 Marotta V, Guerra A, Zatelli MC, Uberti ED, Di Stasi V, Faggiano A, Colao A, Vitale M, 2013, BRAF mutation positive papillary thyroid carcinoma is less advanced when Hashimoto's thyroiditis lymphocytic infiltration is present. Clin Endocrinol 79(5):733–738 Kim WW, Ha TK, Bae SK, 2018, Clinical implications of the BRAF mutation in papillary thyroid carcinoma and chronic lymphocytic thyroiditis. J Otolaryngol 47:4. , DOI 10.1186/ s40463-017-0247-6 Yoon YH, Kim HJ, Lee JW, Kim JM, Koo BS, 2012, The clinicopathologic differences in papillary thyroid carcinoma with or without coexisting chronic lymphocytic thyroiditis. Eur Arch Otorhinolaryngol 269(3):1013–1017 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1191 EP 1197 DI 10.1007/s12253-019-00580-w PG 7 ER PT J AU Chiang, MR Kuo, ChW Wang, WCh Hou, TCh Kuo, ChY Lu, MY Lai, YCh AF Chiang, Ming-Ru Kuo, Chi-Wen Wang, Wen-Chung Hou, Tai-Cheng Kuo, Chen-Yun Lu, Meng-Yao Lai, Yen-Chein TI Correlations between Histological and Array Comparative Genomic Hybridization Characterizations of Wilms Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Wilms tumor; Nephroblastoma; Array comparative genomic hybridization; Molecular pathogenesis ID Wilms tumor; Nephroblastoma; Array comparative genomic hybridization; Molecular pathogenesis AB Wilms tumor, or nephroblastoma, is the most common pediatric renal malignancy. Its diagnosis is principally based on histology. Several genetic loci have been shown to be associated with Wilms tumor formation, including WT1, WT2, FWT1, FWT2, CTNNB1, WTX, and TP53. Other loci, such as 1p, 2q, 7p, 9q, 12q, 14q, 16q, 17p, and 22, have also been implicated in the etiology of Wilms tumor. The aim of this study is to elucidate the molecular pathogenesis of this tumor. In the present study, we analyzed the histological appearance and copy number aberrations using array comparative genomic hybridization of six Wilms tumors without somatic mutation in the WT1 gene. Many chromosomal aberrations on array comparative genomic hybridization analysis revealed that the genetics of Wilms tumors are extremely complex. Amplifications and deletions of large DNA fragments were observed in some samples. Amplifications of NDUFV1, ZIC2, SIX1, NR2F2,MIR1469, SOX9, JAG1, MIR6870, and GNAS were found in all six Wilms tumors. Moreover, amplifications of five genes were identified in the Wilms tumors of stromal type and amplifications of at least 10 genes were identified in the Wilms tumors of epithelial type. Our results indicated that amplifications of nine genes are the essential events in the tumorigenesis of Wilms tumor, which may inform its clinical and therapeutic management. In addition, mixed type Wilms tumor may be the heterogeneous group able to be classified using genetic results of epithelial and stromal components based on immunohistochemistry. C1 [Chiang, Ming-Ru] Jen-Ai Hospital, Department of Pediatrics, 483 Dong Rong Road, 412 Taichung, Taiwan, Republic of China. [Kuo, Chi-Wen] Jen-Ai Hospital, Department of Pharmacy, 483 Dong Rong Road, 412 Taichung, Taiwan, Republic of China. [Wang, Wen-Chung] Jen-Ai Hospital, Department of Obstetrics and Gynecology, 483 Dong Rong Road, 412 Taichung, Taiwan, Republic of China. [Hou, Tai-Cheng] Jen-Ai Hospital, Department of Pathology, 483 Dong Rong Road, 412 Taichung, Taiwan, Republic of China. [Kuo, Chen-Yun] Jen-Ai Hospital, Department of Pathology, 483 Dong Rong Road, 412 Taichung, Taiwan, Republic of China. [Lu, Meng-Yao] National Taiwan University Hospital, Department of Pediatrics, 8 Chung-Shan South Road, 100 Taipei, Taiwan, Republic of China. [Lai, Yen-Chein] Chung Shan Medical University, Department of Medical Laboratory and Biotechnology, No.110, Sec 1, Chien Kuo North Road, 402 Taichung, Taiwan, Republic of China. RP Lai, YCh (reprint author), Chung Shan Medical University, Department of Medical Laboratory and Biotechnology, 402 Taichung, Taiwan, Republic of China. EM yenchein@csmu.edu.tw CR Brown KW, Malik KT, 2001, The molecular biology of Wilms tumour. Expert Rev Mol Med 2001:1–16 Pastore G, Znaor A, Spreafico F, Graf N, Pritchard-Jones K, Steliarova-Foucher E, 2006, Malignant renal tumours incidence and survival in European children, 1978-1997): report from the Automated Childhood Cancer Information System project. Eur J Cancer 42:2103–2114 Steliarova-Foucher E, Colombet M, Ries LAG, Moreno F, Dolya A, Bray F et al, 2017, International incidence of childhood cancer, 2001-10: a population-based registry study. Lancet Oncol 18:719– 731 Stiller CA, Parkin DM, 1990, International variations in the incidence of childhood renal tumours. Br J Cancer 62:1026–1030 Hung IJ, Chang WH, Yang CP, Jaing TH, Liang DC, Lin KH et al, 2004, Epidemiology, clinical features and treatment outcome of Wilms' tumor in Taiwan: a report from Taiwan Pediatric Oncology Group. J Formos Med Assoc 103:104–111 Coppes MJ, Egeler RM, 1999, Genetics of Wilms' tumor. Semin Urol Oncol 17:2–10 Rahman N, Arbour L, Houlston R, Bonaiti-Pellie C, Abidi F, Tranchemontagne J et al, 2000, Penetrance of mutations in the familial Wilms tumor gene FWT1. J Natl Cancer Inst 92:650–652 Al-Hussain T, Ali A, Akhtar M, 2014, Wilms tumor: an update. Adv Anat Pathol 21:166–173 Menke A, McInnes L, Hastie ND, Schedl A, 1998, The Wilms' tumor suppressor WT1: approaches to gene function. Kidney Int 53:1512–1518 Ruteshouser EC, Robinson SM, Huff V, 2008, Wilms tumor genetics: mutations in WT1, WTX, and CTNNB1 account for only about one-third of tumors. Genes Chromosomes Cancer 47:461– 470 Kaneko Y, Homma C, Maseki N, Sakurai M, Hata J, 1991, Correlation of chromosome abnormalities with histological and clinical features in Wilms' and other childhood renal tumors. Cancer Res 51:5937–5942 Lu MY, Wang WC, Lin CW, Chang A, Lai YC, 2014, Identification of a constitutional mutation in the WT1 gene in Taiwanese patients with Wilms tumor. Adv Biosci Biotechnol 5: 230–234 Rassekh SR, Chan S, Harvard C, Dix D, Qiao Y, Rajcan-Separovic E, 2008, Screening for submicroscopic chromosomal rearrangements in Wilms tumor using whole-genome microarrays. Cancer Genet Cytogenet 182:84–94 Natrajan R, Little SE, Sodha N, Reis-Filho JS,Mackay A, Fenwick K et al, 2007, Analysis by array CGH of genomic changes associated with the progression or relapse ofWilms' tumour. J Pathol 211: 52–59 Natrajan R, Williams RD, Grigoriadis A, Mackay A, Fenwick K, Ashworth A et al, 2007, Delineation of a 1Mb breakpoint region at 1p13 in Wilms tumors by fine-tiling oligonucleotide array CGH. Genes Chromosomes Cancer 46:607–615 Md Zin R, Murch A, Charles A, 2011, Pathology, genetics and cytogenetics of Wilms' tumour. Pathology 43:302–312 Kullendorff CM, SollerM,Wiebe T, Mertens F, 2003, Cytogenetic findings and clinical course in a consecutive series of Wilms tumors. Cancer Genet Cytogenet 140:82–87 Hing S, Lu YJ, Summersgill B, King-Underwood L, Nicholson J, Grundy P et al, 2001, Gain of 1q is associated with adverse outcome in favorable histologyWilms' tumors. Am J Pathol 158:393– 398 Gow KW,Murphy JJ, 2002, Cytogenetic and histologic findings in Wilms' tumor. J Pediatr Surg 37:823–827 KrepischiACV, Maschietto M, Ferreira EN, SilvaAG, Costa SS, da Cunha IW et al, 2016, Genomic imbalances pinpoint potential oncogenes and tumor suppressors inWilms tumors.Mol Cytogenet 9: 20 Say B, Balci S, Tuncbilek E, 1971, 45,XO Turner's syndrome, Wilm's tumor and imperforate anus. Humangenetik 12:348–350 Schaub R, Burger A, Bausch D, Niggli FK, Schafer BW, Betts DR, 2007, Array comparative genomic hybridization reveals unbalanced gain of the MYCN region in Wilms tumors. Cancer Genet Cytogenet 172:61–65 Williams RD, Al-Saadi R, Chagtai T, Popov S, Messahel B, Sebire N et al, 2010, Subtype-specific FBXW7mutation andMYCN copy number gain in Wilms' tumor. Clin Cancer Res 16:2036–2045 Cresswell GD, Apps JR, Chagtai T, Mifsud B, Bentley CC, Maschietto M et al, 2016, Intra-tumor genetic heterogeneity in Wilms tumor: clonal evolution and clinical implications. EBioMedicine 9:120–129 Huang M, Weiss WA, 2013, Neuroblastoma and MYCN. Cold Spring Harb Perspect Med 3:a014415 Gadd S, Huff V, Walz AL, Ooms A, Armstrong AE, Gerhard DS et al, 2017, A Children's Oncology Group and TARGET initiative exploring the genetic landscape of Wilms tumor. Nat Genet 49: 1487–1494 Kahlert C, Lerbs T, Pecqueux M, Herpel E, Hoffmeister M, Jansen L et al, 2015, Overexpression of SIX1 is an independent prognostic marker in stage I-III colorectal cancer. Int J Cancer 137:2104–2113 Tian T, Li A, Lu H, Luo R, Zhang M, Li Z, 2015, Six1 promotes glioblastoma cell proliferation and invasion by upregulation of connective tissue growth factor. Am J Cancer Res 5:1823–1830 Emadi-Baygi M, Nikpour PEmadi-Andani E, 2015, SIX1 overexpression in diffuse-type and grade III gastric tumors: features that are associated with poor prognosis. Adv Biomed Res 4:139 Lee CT, Li L, Takamoto N, Martin JF, Demayo FJ, Tsai MJ et al, 2004, The nuclear orphan receptorCOUP-TFII is required for limb and skeletal muscle development. Mol Cell Biol 24:10835–10843 Pereira FA, Qiu Y, Zhou G, Tsai MJ, Tsai SY, 1999, The orphan nuclear receptor COUP-TFII is required for angiogenesis and heart development. Genes Dev 13:1037–1049 Bao Y, Gu D, Feng W, Sun X, Wang X, Zhang X et al, 2014, COUP-TFII regulatesmetastasis of colorectal adenocarcinoma cells by modulating Snail1. Br J Cancer 111:933–943 Yang B, Jing C, Wang J, Guo X, Chen Y, Xu R et al, 2014, Identification of microRNAs associated with lymphangiogenesis in human gastric cancer. Clin Transl Oncol 16:374–379 White NM, Khella HW, Grigull J, Adzovic S, Youssef YM, Honey RJ et al, 2011, miRNA profiling in metastatic renal cell carcinoma reveals a tumour-suppressor effect for miR-215. Br J Cancer 105: 1741–1749 Agatheeswaran S, Pattnayak NC, Chakraborty S, 2016, Identification and functional characterization of the miRNA-gene regulatory network in chronic myeloid leukemia lineage negative cells. Sci Rep 6:32493 Pathak E, Bhavya MD, Atri N, Mishra R, 2017, Deciphering the role of microRNAs in BRD4-NUT fusion gene induced NUT midline carcinoma. Bioinformation 13:209–213 Hong X, SinWC, Harris AL, Naus CC, 2015, Gap junctions modulate glioma invasion by direct transfer of microRNA. Oncotarget 6:15566–15577 Yan Z, Xiong Y, Xu W, Gao J, Cheng Y, Wang Z et al, 2012, Identification of hsa-miR-335 as a prognostic signature in gastric cancer. PLoS One 7:e40037 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1199 EP 1206 DI 10.1007/s12253-019-00601-8 PG 8 ER PT J AU Jiang, W Li, Z Guo, Q Wang, H Ma, M Sun, J Chen, Ch AF Jiang, Wentao Li, Zhang Guo, Qingjun Wang, Honghai Ma, Ming Sun, Jisan Chen, Chiyi TI Identification of the Pathogenic Biomarkers for Hepatocellular Carcinoma Based on RNA-seq Analyses SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma (HCC); Early diagnosis; RNA-seq ID Hepatocellular carcinoma (HCC); Early diagnosis; RNA-seq AB The purpose of this study was to explore potential biomarkers in the diagnosis of hepatocellular carcinoma (HCC) based on RNA-seq. The microarray data GSE98269 were downloaded from the GEO database, including the miRNA, mRNA and lncRNA expression profiles of 3 HCC tissues and 3 normal liver tissues from 3 HCC patients. The limma package was used to identify the differentially expressed miRNAs (DEMs) and the differentially expressed lncRNAs in HCC tissues compared with normal liver tissues. Database of DAVID, KEGG PATHWAY and Reactome were used to perform the functional and pathway enrichment. Putative targets for DEMs, and the miRNA-gene pairs were predicted via the miRWalk V2.0 database. The proteinprotein pairs of DEGs were screened via String software. The expression features of the differentially expressed lncRNAs were analyzed. The regulated network of DEGs and DEMs were constructed, and related genes and miRNAs were detected in the HCC tissues and normal liver samples with Q-PCR. A total of 678 DEGs, 32 DEMs and 411 differential expressed lncRNAs were identified. The DEGs were enriched in 196 GO terms and 79 pathways. 38 negative regulation miRNA-gene pairs and 1205 protein-protein interactions were screened out, and the regulated network was constructed based on them. KNG1, CDK1, EHHADH, CYP3A4, hsa-miR-199a-5p and hsa-miR-455-3p might be biomarkers in the occurrence of HCC. C1 [Jiang, Wentao] Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 24 Fukang Road, Nankai District, 300192 Tianjin, China. [Li, Zhang] Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 24 Fukang Road, Nankai District, 300192 Tianjin, China. [Guo, Qingjun] Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 24 Fukang Road, Nankai District, 300192 Tianjin, China. [Wang, Honghai] Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 24 Fukang Road, Nankai District, 300192 Tianjin, China. [Ma, Ming] Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 24 Fukang Road, Nankai District, 300192 Tianjin, China. [Sun, Jisan] Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 24 Fukang Road, Nankai District, 300192 Tianjin, China. [Chen, Chiyi] Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 24 Fukang Road, Nankai District, 300192 Tianjin, China. RP Jiang, W (reprint author), Tianjin First Center Hospital, Key Lab for Critical Care Medicine of the Ministry of Health, 300192 Tianjin, China. EM wentaojiang23@163.com CR McGuire S, 2016, World Cancer Report 2014. Geneva, Switzerland: World Health Organization, International Agency for Research on Cancer, WHO Press, 2015. Advances in Nutrition, Bethesda, Md, 7:418–419 Ferlay J, Soerjomataram I, Dikshit R et al, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. International Journal of Cancer 136:E359– E386 Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(9):7–30 Diaz-Gonzalez A, Forner A, Rodriguez de Lope C, Varela M, 2016, New challenges in clinical research on hepatocellular carcinoma. Revista Espanola De Enfermedades Digestivas : Organo Oficial De La Sociedad Espanola De Patologia Digestiva 108: 485–493 Li D, Satomura S, 2015, Biomarkers for hepatocellular carcinoma, HCC): an update. Adv Exp Med Biol 867:179–193 Kiyokawa H, Yasuda H, 2017, Serum Monomeric Laminin- Gamma2 as a Novel Biomarker for Hepatocellular Carcinoma. Cancer Science 108:1432–1439 Shaker O, Alhelf M, Morcos G, Elsharkawy A, 2017, miRNA-101-1 and miRNA-221 expressions and their polymorphisms as biomarkers for early diagnosis of hepatocellular carcinoma. Infection, Genetics and Evolution : Journal of Molecular Epidemiology and Evolutionary Genetics in Infectious Diseases 51:173–181 Shen Q, Fan J, Yang XR et al, 2012, Serum DKK1 as a protein biomarker for the diagnosis of hepatocellular carcinoma: a largescale, multicentre study. Lancet Oncol 13:817–826 Huang S, Jiang F, Wang Yet al, 2017, Diagnostic performance of tumor markers AFP and PIVKA-II in Chinese hepatocellular carcinoma patients. Tumour Biology : the Journal of the International Society for Oncodevelopmental Biology and Medicine 39: 1010428317705763 Saitta C, Raffa G, AlibrandiA et al, 2017, PIVKA-II is a useful tool for diagnostic characterization of ultrasound-detected liver nodules in cirrhotic patients. Medicine 96:e7266 Iarovaia GA, 2001, Kallikrein-kinin system: novel facts and concepts, literature review). Vopr Med Khim 47:20–42 Colman RW, 2006, Regulation of angiogenesis by the kallikreinkinin system. Curr Pharm Des 12:2599–2607 Quesada-Calvo F, Massot C, Bertrand V et al, 2017, OLFM4, KNG1 and Sec24C identified by proteomics and immunohistochemistry as potential markers of early colorectal cancer stages. Clin Proteomics 14:9 Ren Y, Ji N, Kang X et al, 2016, Aberrant ceRNA-mediated regulation of KNG1 contributes to glioblastoma-induced angiogenesis. Oncotarget Cho SJ, Lee SS, Kim YJ et al, 2010, Xylocydine, a novel Cdk inhibitor, is an effective inducer of apoptosis in hepatocellular carcinoma cells in vitro and in vivo. Cancer Lett 287:196–206 Luo HB, Chen GW, Shao YX, Li Z, Liu M, Liu PQ, 2012, 3DQSAR studies on pyrimidine analogues as cyclin-dependent kinase 1 inhibitors. Shenzhen Daxue Xuebao 29:438–443 Zhao J, Han SX, Ma JL et al, 2013, The role of CDK1 in apoptininduced apoptosis in hepatocellular carcinoma cells. Oncol Rep 30: 253–259 Yi Z,Wei H, Yan R et al, 2015, miR-582-5p inhibits proliferation of hepatocellular carcinoma by targeting CDK1 and AKT3. Tumor Biol 36:8309–8316 Baes M, Van Veldhoven PP, 2016, Hepatic dysfunction in peroxisomal disorders. BBA - Molecular Cell Research 1863:956–970 Houten SM, Denis S, Argmann CA, 2012, Peroxisomal Lbifunctional enzyme, Ehhadh, is essential for the production of medium-chain dicarboxylic acids. J Lipid Res 53:1296–1303 Suto K, Kajihara-Kano H, Yokoyama Y et al, 1999, Decreased expression of the peroxisomal bifunctional enzyme and carbonyl reductase in human hepatocellular carcinomas. J Cancer Res Clin Oncol 125:83–88 Zanger UM, Schwab M(2013, Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther 138:103–141 Ali GT, Al-Azhary NM, Mokhtar DA, 2014, Frequency and prognostic significant of CYP3A4-a-290G polymorphism in acute myeloid leukemia. J Adv Res 5:657–661 Rodrigues RM, De Kock J, Doktorova TY, Rogiers V, Vanhaecke T, 2015, Measurement of cytochrome P450 enzyme induction and inhibition in human hepatoma cells. Methods Mol Biol 1250:279–285 Williams AE, 2008, Functional aspects of animal microRNAs. Cell Mol Life Sci 65:545–562 Eulalio A, Huntzinger E, Nishihara T, Rehwinkel J, Fauser M, Izaurralde E, 2009, Deadenylation is a widespread effect of miRNA regulation. RNA, New York, NY, 15:21–32 Hawkins PG, Morris KV, 2008, RNA and transcriptional modulation of gene expression. Cell cycle, Georgetown, Tex, 7:602–607 Schultz S, Neubauer H, Bartsch H et al, 2014, Hsa-miR-199a- 5p is functionally relevant in progression from ductal carcinoma in situ to invasive breast tumours. Oncology Research & Treatment 37:68–68 Shen Q, Cicinnati VR, Zhang X et al, 2010, Role of microRNA- 199a-5p and discoidin domain receptor 1 in human hepatocellular carcinoma invasion. Mol Cancer 9:227 Sand M, Skrygan M, Sand D et al, 2012, Expression of microRNAs in basal cell carcinoma. Br J Dermatol 167:847–855 Chickooree D, Zhu K, Ram V, Wu HJ, He ZJ, Zhang S, 2016, A preliminary microarray assay of the miRNA expression signatures in buccal mucosa of oral submucous fibrosis patients. J Oral Pathol Med 45:691–697 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1207 EP 1213 DI 10.1007/s12253-019-00596-2 PG 7 ER PT J AU Samadani, AA Nikbakhsh, N Taheri, H Shafaee, Sh Fattahi, S Langroudi, PM Hajian, K Akhavan-Niaki, H AF Samadani, Akbar Ali Nikbakhsh, Novin Taheri, Hassan Shafaee, Shahriyar Fattahi, Sadegh Langroudi, Pilehchian Maryam Hajian, Karimollah Akhavan-Niaki, Haleh TI CDX1/2 and KLF5 Expression and Epigenetic Modulation of Sonic Hedgehog Signaling in Gastric Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; DNA methylation; Gene expression; Epigenetics ID Gastric cancer; DNA methylation; Gene expression; Epigenetics AB Gastric cancer is among the commonplace causes of cancer death worldwide. Sonic hedgehog (Shh) signaling is an important pathway which may be dysregulated in many cancers.CDX1/2, and KLF5are key transcription factors involved in Shh pathway and cancer stem cells. The aim of this study was to investigate the expression and epigenetic alterations of these genes in gastric cancer patients. DNA methylation’s modifications of CDX1, KLF5 and CDX2 genes alongside with the expressions of these genes in gastric cancer tissues and their non-tumoral counterparts (margin tissues) were analyzed using methylation specific sequencing, and Real time PCR Taq man assays, respectively. The expression of CDX1 (P = 0.002) and KLF5 (P = 0.010) were decreased significantly, but it was considerably increased for CDX2 (P = 0.001). Relatively, the results for the regulatory region methylation status of each CpG site had shown a notable fluctuation in these genes with no significant difference in most places. The creation of metastatic lymph nodes in patients was significantly associated with increased expression of CDX2 gene. The modifications of these genes expression can be considered as a cancer biomarker in future studies. Methylation of the investigated genes is not the main mechanism of gastric cancer development. C1 [Samadani, Akbar Ali] Babol University of Medical Sciences, Health Research Institute, Cellular and Molecular Biology Research CenterBabol, Iran. [Nikbakhsh, Novin] Babol University of Medical Sciences, Cancer Research Center, Department of SurgeryBabol, Iran. [Taheri, Hassan] Babol University of Medical Sciences, Department of GastroenterologyBabol, Iran. [Shafaee, Shahriyar] Babol University of Medical Sciences, Department of PathologyBabol, Iran. [Fattahi, Sadegh] Babol University of Medical Sciences, Health Research Institute, Cellular and Molecular Biology Research CenterBabol, Iran. [Langroudi, Pilehchian Maryam] Babol University of Medical Sciences, Health Research Institute, Cellular and Molecular Biology Research CenterBabol, Iran. [Hajian, Karimollah] Babol University of Medical Sciences, Faculty of Medicine, Department of Social medicine and statisticsBabol, Iran. [Akhavan-Niaki, Haleh] Babol University of Medical Sciences, Health Research Institute, Cellular and Molecular Biology Research CenterBabol, Iran. RP Akhavan-Niaki, H (reprint author), Babol University of Medical Sciences, Health Research Institute, Cellular and Molecular Biology Research Center, Babol, Iran. EM halehakhavan@yahoo.com CR Akhavan-Niaki H, Samadani AA, 2014, Molecular insight in gastric cancer induction: an overview of cancer stemness genes. Cell Biochem Biophys 68(3):463–473 Nikbakhsh N, Shafahi S, Taheri H, Samadani AA, Noroullahi SE, Pilecian M, Saromeh SH, Kolagar A, Ghasemzadeh M, Esmailzadeh Tahmtan S, 2016, Histopathological investigation in gastric cancer. J Int Res Med Pharm Sci 8(2):52–56 Singh SR, 2013, Gastric cancer stem cells: a novel therapeutic target. Cancer Lett 338(1):110–119 Li K, Dan Z, Nie YQ, 2014, Gastric cancer stem cells in gastric carcinogenesis, progression, prevention and treatment. World J Gastroenterol 20(18):5420–5426 Xu Z-Y, Tang J-N, Xie H-X, Du Y-A, Huang L,Yu P-F, Cheng X-D, 2015, 5-fluorouracil chemotherapy of gastric Cancer generates residual cells with properties of Cancer stemcells. Int J Biol Sci 11(3): 284–294 SamadaniAA, Akhavan-NiakiH(2015, Interaction of sonic hedgehog, SHH, pathway with cancer stem cell genes in gastric cancer. Med Oncol 32(3):48–55 Fujii Y, Yoshihashi K, Suzuki H, Tsutsumi S, Mutoh H, Maeda S, Yamagata Y, Seto Y, Aburatani H, Hatakeyama M, 2012, CDX1 confers intestinal phenotype on gastric epithelial cells via induction of stemness-associated reprogramming factors SALL4 and KLF5. Proc Natl Acad Sci U S A 09(50):20584–20589 Bornschein J, Tothk, Selgrad M, Kuestel D, Wex T, Molnar B, Tulassay Z, Malfertheiner P, 2013, Dysregulation of CDX1, CDX2 and SOX2 in patients with gastric cancer also affects the non-malignant mucosa. J Clin Pathol 66(9):819–822 Hryniuk A, Grainger S, Savory JG, Lohnes D, 2014, CDX1 and CDX2 function as tumor suppressors. J Biol Chem 289(48):33343– 33354 Samadani AA, Nikbakhsh N, Pilehchian M, Fattahi S, Akhavan- Niaki H, 2016, Epigenetic changes of CDX2 in gastric adenocarcinoma. J Cell Commun Signal 10(4):267–272 Chia NY, Deng N, Das K, Huang D, Hu L, Zhu Y, Lim KH, Lee MH,Wu J, SamXX, Tan GS,WanWK, YuW, Gan A, Tan AL, Tay ST, Soo KC, Wong WK, Dominguez LT, Ng HH, Rozen S, Goh LK, Teh BT, Tan P, 2015, Regulatory crosstalk between lineagesurvival oncogenes KLF5, GATA4 and GATA6 cooperatively promotes gastric cancer development. Gut 64(5):707–719 Kwak MK, Lee HJ, Hur K, Park DJ, Lee HS, Kim WH, Lee KU, Choe KJ, Guilford P, 2008, Expression of Kruppel-like factor 5 in human gastric carcinomas. J Cancer Res Clin Oncol 134(2):163–167 Kang JM, Lee BH, Kim N, Lee HS, Lee HE, Park JH, KimJS, Jung HC, Song IS, 2011, CDX1 and CDX2 expression in intestinal metaplasia, dysplasia and gastric cancer. J Korean Med Sci 26(5): 647–653 Mutoh H, Sakurai S, Satoh K, Osawa H, Hakamata Y, Takeuchi T, Sugano K, 2004, Cdx1 induced intestinal metaplasia in the transgenic mouse stomach: comparative study with Cdx2 transgenic mice. Gut 53(10):1416–1423 Visvader JE, Linderman GJ, 2008, Cancer stem cells in solid tumors: accumulating evidence and unresolved questions. Nat Rev Cancer 8(10):755–768 Akhavan-Niaki H, Samadani AA, 2013, DNA methylation and cancer development: molecular mechanism. Cell Biochem Biophys 67(2):501–513 Hur K, Niwa T, Toyoda T, Tsukamoto T, Tatematsu M, Yang HK, Ushijima T, 2011, Insufficient role of cell proliferation in aberrant DNA methylation induction and involvement of specific types of inflammation. Carcinogenesis 32(1):35–41 Fattahi S, Amirbozorgi G, Lotfi M, Navaei BA, Kavoosian S, Asouri M, Akhavan-Niaki H, 2018, Development of a universal Taqman probe for mRNA gene expression analysis. Iran J Sci Technol Trans A Sci 42(2):363–370 Fattahi S, Pilehchian Langroudi M, Samadani AA, Nikbakhsh N, Asouri M, Akhavan-Niaki H, 2017, Application of unique sequence index, USI, barcode to gene expression profiling in gastric adenocarcinoma. J Cell Commun Signal 11(1):97–104 Chia N-Y, Deng N, Das K, Huang D, Hu L, Zhu Y, Lim KH, Lee MH, Wu J, Sam XX, Tan GS, Wan WK, Yu W, Gan A, Tan ALK, Tay ST, Soo KC, Wong WK, Dominguez LTM, Ng HH, Rozen S, Goh LK, Teh BT, Tan P, 2015, Regulatory crosstalk between lineage-survival oncogenes KLF5, GATA4 and GATA6 cooperatively promotes gastric cancer development. Gut 64(5):707–719 Eda A, Osawa H, Yanaka I, Satoh K,Mutoh H, Kihira K, Sugano K, 2002, Expression of homeobox gene CDX2 precedes that ofCDX1 during the progression of intestinal metaplasia. J Gastroenterol 37: 94–100 Baba Y, Nosho K et al, 2009, Ralationship of CDX2 loss with molecular features and prognosis in colorectal cancer. Clin Cancer Res 15:4665–4673 Mizoshita T et al, 2001, Expression of CDX1 and CDX2 mRNAs and relevance of this expression to differentiation in human gastrointestinal mucosa with special emphasis on participation in intestinal metaplasia of the human stomach. Gastric Cancer 4(4):185–191 Chan CW et al, 2009, Gastrointestial differentiation marker cytokeratin 20 is regulated by homeobox CDX1. Proc Natl Acad Sci 16:1936–1941 Silberg DG, Sullivan J, Kang E, Swain GP, Moffett J, Sund NJ, Sackett SD, Kaestner KH, 2002, CDX2 ectopic expression induces gastric intestinal metaplasia in transgenic mice. Gastroenterology 122(3):689–696 Liu Y, Chidgey M, Yang VW1, Bialkowska AB, 2017, Kruppellike factor 5 is essential for maintenance of barrier function in mouse colon. Am J Physiol Gastrointest Liver Physiol 313(5): G478–G491 Hryniuk A, Grainger S, Savory JG, Lohnes D, 2014, Cdx1 and Cdx2 function as tumor suppressors. J Biol Chem 289(48): 33343–33354 Pilozzi E, OnelliMR, Ziparo V, Mercantini P, Ruco L, 2004, CDX1 expression is reduced in colorectal carcinoma and is associated with promoter hypermethylation. J Pathol 204:289–295 Zeraati H, Amiri Z, 2016, Estimating postoperative survival of gastric cancer patients and factors affecting it in Iran: based on a TNM-7 staging system. Acta Med Iran 54(2):114–8.5 Binto L, Yong J, Antebi YE, McCue K, Kazuki Y, Uno N, Oshimura M, Elowitz MB, 2016, Dynamics of epigenetic regulation at the single-cell level. Acta Med Iran 351(6274):720–724 Cropley JE, Dang TH,Martin DI, Suter CM, 2012, The penetrance of an epigenetic trait in mice is progressively yet reversibly increased by selection and environment. Proc Boil Sci 279(1737): 2347–2353 Lee JY, Kong G, 2016, Roles and epigenetic regulation of epithelial-mesenchymal transition and its transcription factors in cancer initiation and progression. Proc Biol Sci 73(24):4643–4660 Chen Z1, Wu Q, Ding Y, Zhou W, Liu R, Chen H, Zhou J, Feng J, 2018, Discovery of novel mifepristone derivatives via suppressing KLF5 expression for the treatment of triple-negative breast cancer. Eur J Med Chem 146:354–367 Bialkowska A, Crisp M, Madoux F, Spicer T, Knapinska A,Mercer B, Bannister TD, He Y, Chowdhury S, Cameron M, Yang VW, Hodder P. ML264: an antitumor agent that potently and selectively inhibits Kruppel-like factor five, KLF5, expression: a probe for studying Colon Cancer development and progression. Probe reports from the NIH molecular libraries program [internet]. Bethesda, MD): National Center for biotechnology information, US); 2010–2011 Oct 31 [updated 2013 Mar 7] Fattahi S, Pilehchian Langroudi M, Akhavan-Niaki H, 2018, Hedgehog signaling pathway: epigenetic regulation and role in disease and cancer development. J Cell Physiol 233(8):5726–5735 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1215 EP 1222 DI 10.1007/s12253-019-00594-4 PG 8 ER PT J AU Mohamed, G Talima, S Li, L Wei, W Rudzki, Z Allam, MR Simmons, W Tao, Q Murray, GP AF Mohamed, Ghada Talima, Soha Li, Lili Wei, Wenbin Rudzki, Zbigniew Allam, Mahmoud Rasha Simmons, William Tao, Qian Murray, G Paul TI Low Expression and Promoter Hypermethylation of the Tumour Suppressor SLIT2, are Associated with Adverse Patient Outcomes in Diffuse Large B Cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SLIT2; DLBCL; Hypermethylation; Patient survival ID SLIT2; DLBCL; Hypermethylation; Patient survival AB SLIT2 has been classified as a major tumour suppressor gene due to its frequent inactivation in different cancer types. However, alterations of SLIT2 expression and relation to patient outcomes in diffuse large B cell lymphoma (DLBCL) remain undefined. The aim of this study was to investigate the expression and the methylation status of SLIT2 gene as well as its relation to patient outcomes in DLBCL. Immunohistochemical (IHC) staining was carried out to detect the expression of SLIT2 in a series of 108 DLBCL cases. Re-analysis of previously published dataset (GSE10846) that measured gene expression in DLBCL patients who had received CHOP or R-CHOP therapy was performed to identify associations between SLIT2 and patients survival. Laser capture microdissection was performed to isolate GC B cells and DLBCL primary tumor cells. Bisulfite treatment and methylation-specific PCR (MSP) analysis were done to assess SLIT2 promotor methylation status.We report that the expression of SLIT2 protein was reduced in a subset of DLBCL cases and this was significantly correlated with advanced clinical stage (p = 0.041) and was an independent predictor of worse overall survival (OS) (p = 0.012). Re-analysis of published gene expression data showed that reduced SLIT2 mRNA expression was significantly correlated with worse OS in R-CHOP-treated ABC DLBCL patients (p = <0.01). Hypermethylation of the SLIT2 promotor was significantly correlated with low SLIT2 expression (p = 0.009). Our results provide a novel evidence of reduced expression of SLIT2 that is associated with promoter hypermethylation and adverse outcomes in patients with DLBCL. C1 [Mohamed, Ghada] Cairo University, National Cancer Institute, Department of PathologyGiza, Egypt. [Talima, Soha] Cairo University, Faculty of Medicine, Clinical Oncology DepartmentGiza, Egypt. [Li, Lili] The Chinese University of Hong Kong, State Key Laboratory of Oncology in South China, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health SciencesHong Kong, Hong Kong. [Wei, Wenbin] University of Birmingham, Institute of Cancer and Genomic Sciences, Vincent Drive, Edgbaston, B15 2TT Birmingham, UK. [Rudzki, Zbigniew] University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Department of Cellular PathologyBirmingham, UK. [Allam, Mahmoud Rasha] Cairo University, National Cancer Institute, Biostatistic and Cancer Epidemiology DepartmentGiza, Egypt. [Simmons, William] University of Birmingham, Institute of Cancer and Genomic Sciences, Vincent Drive, Edgbaston, B15 2TT Birmingham, UK. [Tao, Qian] The Chinese University of Hong Kong, State Key Laboratory of Oncology in South China, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health SciencesHong Kong, Hong Kong. [Murray, G Paul] University of Birmingham, Institute of Cancer and Genomic Sciences, Vincent Drive, Edgbaston, B15 2TT Birmingham, UK. RP Mohamed, G (reprint author), Cairo University, National Cancer Institute, Department of Pathology, Giza, Egypt. EM dr.ghada.elshafaee@gmail.com CR Hunt KE, Reichard KK, 2008, Diffuse large B-cell lymphoma. Arch Pathol Lab Med 132(1):118–124 Teras LR, DeSantis CE, Cerhan JR, Morton LM, Jemal A, Flowers CR, 2016, US lymphoid malignancy statistics by World Health Organization subtypes. CA Cancer J Clin 66(6):443–459. https:// doi.org/10.3322/caac.21357 Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, Boldrick JC, Sabet H, Tran T, Yu X, Powell JI, 2000, Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 403(6769):503. , DOI 10.1038/ 35000501 Sujobert P, Salles G, Bachy E, 2016, Molecular classification of diffuse large B-cell lymphoma: what is clinically relevant? Hematol Oncol ClinN30(6):1163–1177. , DOI 10.1016/j.hoc.2016. 07.001 McKelvey EM, Gottlieb JA, Wilson HE, Haut A, Talley RW, Stephens R, Lane M, Gamble JF, Jones SE, Grozea PN, Gutterman J, 1976, Hydroxyldaunomycin, Adriamycin, combination chemotherapy in malignant lymphoma. Cancer 38(4):1484– 1493. , DOI 10.1002/1097-0142(197610)38:4%3C1484:: aid-cncr2820380407%3E3.0.co;2-i Fisher RI, Gaynor ER, Dahlberg S, Oken MM, Grogan TM, Mize EM, Glick JH, Coltman CA Jr, Miller TP, 1993, Comparison of a standard regimen, CHOP, with three intensive chemotherapy regimens for advanced non-Hodgkin's lymphoma. New England Journal of Medicine 328(14):1002-6. , DOI 10.1056/ nejm199304083281404 Coiffier B, Lepage E, Briere J, Herbrecht R, Tilly H, Bouabdallah R, Morel P, Van Den Neste E, Salles G, Gaulard P, Reyes F, 2002, CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med 346(4):235–242. , DOI 10.1056/nejmoa011795 Coiffier B, Thieblemont C, Van Den Neste E, Lepeu G, Plantier I, Castaigne S, Lefort S, Marit G, Macro M, Sebban C, Belhadj K, 2010, Long-term outcome of patients in the LNH-98.5 trial, the first randomized study comparing rituximab-CHOP to standard CHOP chemotherapy in DLBCL patients: a study by the Groupe d'Etudes des Lymphomes de l'Adulte. Blood 1:blood-2010. https:// doi.org/10.1182/blood-2010-03-276246 Cerhan JR, Berndt SI, Vijai J, Ghesquieres H, McKay J,Wang SS, Wang Z, Yeager M, Conde L, de Bakker PI et al, 2014, Genomewide association study identifies multiple susceptibility loci for diffuse large B cell lymphoma. Nat. Genet 46:1233–1238. https:// doi.org/10.1158/1538-7445.am2014-lb-272 Skibola CF, Curry JD, Nieters A, 2007, Genetic susceptibility to lymphoma. Haematologica 92(7):960–969 Grone J, Doebler O, Loddenkemper C, Hotz B, Buhr HJ, Bhargava S, 2006, Robo1/Robo4: differential expression of angiogenic markers in colorectal cancer. Oncol Rep 15(6):1437–1443. https:// doi.org/10.3892/or.15.6.1437 Xian J, Clark KJ, Fordham R, Pannell R, Rabbitts TH, Rabbitts PH, 2001, Inadequate lung development and bronchial hyperplasia in mice with a targeted deletion in the Dutt1/Robo1 gene. Proc Natl Acad Sci 98(26):15062–15066 Dallol A, Da Silva NF, Viacava P, Minna JD, Bieche I, Maher ER, Latif F, 2002, SLIT2, a human homologue of the Drosophila Slit2 gene, has tumor suppressor activity and is frequently inactivated in lung and breast cancers. Cancer Res 62(20):5874–5880 Qiu H, Zhu J, Yu J, Pu H, Dong R, 2011, SLIT2 is epigenetically silenced in ovarian cancers and suppresses growth when activated. Asian Pac J Cancer Prev 12(3):791–795 Prasad A, Paruchuri V, Preet A, Latif F, Ganju RK, 2008, Slit-2 induces a tumor-suppressive effect by regulating β-catenin in breast cancer cells. J Biol Chem 283(39):26624–26633. , DOI 10. 1074/jbc.m800679200 Appe AJ, Aggerholm A, Hansen MC, Ebbesen LH, Hokland P, Bentzen HH, Nyvold CG, 2017, Differential expression levels and methylation status of ROBO1 in mantle cell lymphoma and chronic lymphocytic leukaemia. Int J Lab Hematol 39(3):e70– e73. , DOI 10.1111/ijlh.12615 Dunwell TL, Dickinson RE, Stankovic T, Dallol A, Weston V, Austen B, Catchpoole D, Maher ER, Latif F, 2009, Frequent epigenetic inactivation of the SLIT2 gene in chronic and acute lymphocytic leukemia. Epigenetics 4(4):265–269. , DOI 10. 4161/epi.9137 Enjuanes A, Fernandez V, Hernandez L, Navarro A, Bea S, Pinyol M, Lopez-Guillermo A, Rosenwald A, Ott G, Campo E, Jares P, 2011, Identification of methylated genes associated with aggressive clinicopathological features in mantle cell lymphoma. PLoS One 6(5):e19736. , DOI 10.1371/journal.pone.0019736 Hans CP,Weisenburger DD, Greiner TC, Gascoyne RD, Delabie J, Ott G,Muller-Hermelink HK, Campo E, Braziel RM, Jaffe ES, Pan Z, 2004, Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood 103(1):275–282. , DOI 10.1182/blood- 2003-05-1545 Lenz G,Wright G, Dave SS, XiaoW, Powell J, Zhao H, XuW, Tan B, Goldschmidt N, Iqbal J,Vose J, 2008, Stromal gene signatures in large-B-cell lymphomas. N Engl J Med 359(22):2313–2323. , DOI 10.1056/nejmoa0802885 Bolstad BM, Irizarry RA, Astrand M, Speed TP, 2003, A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19(2):185–193. , DOI 10.1093/bioinformatics/19.2.185 Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP, 2003, Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Research 31(4):e15. , DOI 10.1093/nar/ gng015 Tao Q, Swinnen LJ, Yang J, Srivastava G, Robertson KD, Ambinder RF, 1999, Methylation status of the Epstein-Barr virus major latent promoter C in iatrogenic B cell lymphoproliferative disease: application of PCR-based analysis. Am J Pathol 155(2): 619–625. , DOI 10.1016/s0002-9440(10)65157-7 Tao Q, Huang H, Geiman TM, Lim CY, Fu L, Qiu GH, Robertson KD, 2002, Defective de novo methylation of viral and cellular DNA sequences in ICF syndrome cells. Hum Mol Genet 11(18): 2091–2102. , DOI 10.1093/hmg/11.18.2091 Murray PG, Fan Y, Davies G, Ying J, Geng H, Ng KM, Li H, Gao Z, Wei W, Bose S, Anderton J, 2010, Epigenetic silencing of a proapoptotic cell adhesion molecule, the immunoglobulin superfamily member IGSF4, by promoter CpG methylation protects Hodgkin lymphoma cells from apoptosis. Am J Pathol 177(3): 1480–1490 , DOI 10.2353/ajpath.2010.100052 Andrews W, Liapi A, Plachez C, Camurri L, Zhang J, Mori S, Murakami F, Parnavelas JG, Sundaresan V, Richards LJ, 2006, Robo1 regulates the development of major axon tracts and interneuron migration in the forebrain. Development 133(11):2243– 2252 , DOI 10.1242/dev.02379 Qin F, Zhang H, Ma L, Liu X, Dai K, Li W, Gu F, Fu L, Ma Y, 2015, Low expression of Slit2 and Robo1 is associated with poor prognosis and brain-specific metastasis of breast cancer patients. Sci Rep 5:14430. , DOI 10.1038/srep14430 Tseng RC, Chang JM, Chen JH, HuangWR, TangYA, Kuo IY, Yan JJ, Lai WW, Wang YC, 2015, Deregulation of SLIT2-mediated Cdc42 activity is associated with esophageal cancer metastasis and poor prognosis. J Thorac Oncol 10(1):189–198. https://doi. org/10.1097/jto.0000000000000369 Kim HK, Zhang H, Li H, Wu TT, Swisher S, He D, Wu L, Xu J, Elmets CA, Athar M, Xu XC, 2008, Slit2 inhibits growth and metastasis of fibrosarcoma and squamous cell carcinoma. Neoplasia 10(12):1411–1420. , DOI 10.1593/neo.08804 Mitra S, Mazumder-Indra D, Mondal RK, Basu PS, Roy A, Roychoudhury S, Panda CK, 2012, Inactivation of SLIT2- ROBO1/2 pathway in premalignant lesions of uterine cervix: clinical and prognostic significances. PLoS One 7(6):e38342. https:// doi.org/10.1371/journal.pone.0038342 TsengRC, Lee SH, Hsu HS, Chen BH, TsaiWC, Tzao C,Wang YC, 2010, SLIT2 attenuation during lung cancer progression deregulates β-catenin and E-cadherin and associates with poor prognosis. Cancer Res 12:0008–5472. , DOI 10.1158/0008-5472.can- 09-2084 Dickinson RE, Duncan WC, 2010, The SLIT-ROBO pathway: a regulator of cell function with implications for the reproductive system. Reproduction 139:697–704. , DOI 10.1530/rep- 10-0017 Wu WJ, Tu S, Cerione RA, 2003, Activated Cdc42 sequesters c- Cbl and prevents EGF receptor degradation. Cell 114:715–725. , DOI 10.1016/s0092-8674(03)00688-3 Wong K, Ren XR, Huang YZ et al, 2001, Signal transduction in neuronal migration: roles of GTPase activating proteins and the small GTPase Cdc42 in the Slit-Robo pathway. Cell 107:209– 221. , DOI 10.1016/s0092-8674(01)00530-x Park KW, Morrison CM, Sorensen LK, Jones CA, Rao Y, Chien CB,Wu JY, Urness LD, Li DY, 2003, Robo4 is a vascular-specific receptor that inhibits endothelial migration. Dev Biol 261(1):251– 267. , DOI 10.1016/s0012-1606(03)00258-6 Chang PH, Hwang-Verslues WW, Chang YC, Chen CC, Hsiao M, JengYM, Chang KJ, Lee EY, Shew JY, LeeWH, 2012, Activation of Robo1 signaling of breast cancer cells by Slit2 from stromal fibroblast restrains tumorigenesis via blocking PI3K/Akt/β-catenin pathway. Cancer Res 23:canres-0877. , DOI 10.1158/ 0008-5472.can-12-0877 Legg JA, Herbert JM, Clissold P, Bicknell R, 2008, Slits and roundabouts in cancer, tumour angiogenesis and endothelial cell migration. Angiogenesis 11(1):13–21. , DOI 10.1007/s10456- 008-9100-x Maiti GP, Ghosh A, Mondal P, Ghosh S, Chakraborty J, Roy A, Roychowdhury S, Panda CK, 2015, Frequent inactivation of SLIT2 and ROBO1 signaling in head and neck lesions: clinical and prognostic implications. Oral surgery, oral medicine, oral pathology and oral radiology1 119(2):202–212. , DOI 10. 1016/j.oooo.2014.09.029 Dallol A, Forgacs E, Martinez A, Sekido Y, Walker R, Kishida T, Rabbitts P, Maher ER, Minna JD, Latif F, 2002, Tumour specific promoter region methylation of the human homologue of the Drosophila roundabout gene DUTT1, ROBO1, in human cancers. Oncogene 21(19):3020. , DOI 10.1038/sj.onc.1205421 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1223 EP 1231 DI 10.1007/s12253-019-00600-9 PG 9 ER PT J AU Martin-Manzo, VM Lara, C Vargas-de-Leon, C Carrero, J Queipo, G Fonseca-Sanchez, M Mejia-Dominguez, RN Kershenobich, D Mummidi, S Zentella-Dehesa, A Hernandez, J AF Martin-Manzo, Victoria Miriam Lara, Carlos Vargas-de-Leon, Cruz Carrero, Julio Queipo, Gloria Fonseca-Sanchez, Miguel Mejia-Dominguez, R Nancy Kershenobich, David Mummidi, Srinivas Zentella-Dehesa, Alejandro Hernandez, Joselin TI Interaction of Breast Cancer and Insulin Resistance on PD1 and TIM3 Expression in Peripheral Blood CD8 T Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Insulin resistance; TIM3; PD1 ID Breast cancer; Insulin resistance; TIM3; PD1 AB Epidemiological evidence points to a link between insulin resistance (IR) and breast cancer (BrCA). Insulin plays a role in CD8+ Tcells (CD8T) differentiation and function and affects adipocytokines levels. CD8Tactivity in BrCA is associated with favorable outcome; while PD1 and TIM3 are markers of CD8T exhaustion and play critical roles in the negative regulation of T cell responses. Patients with (BrCA) have high expression levels of PD1 on circulating. Therefore, we hypothesized that BrCA and IR could affect PD1 and/or TIM3 expression on circulating CD8T. We determine PD1 and TIM3 expression on CD8T and analyze the relationship of CD8T phenotype with serum insulin and plasma adipocytokines levels in the different groups. We enrolled four groups of treatment-naive patients: women without neoplasms (Neo-)/without IR (IR-), Neo−/with IR (IR+), BrCa/ IR- and BrCa/IR+.We found interactions between BrCA and IR with respect to TIM3 on naive and central memory (CM) CD8T subsets. Furthermore, BrCA had a greater PD1 + TIM3- CD8T frequency in CD8T subsets than Neo-. IR+ presented a significantly lower PD1 + TIM3- frequency in CD8Tsubsets compare to Non-IR. In addition, we found a negative correlation between insulin levels, HOMA and frequency of PD1 + TIM3- in CD8T and a positive correlation between adiponectin levels and the frequency PD1 + TIM3- in CD8T. The increased expression of PD1 on different subsets of CD8T from BrCa patients is consistent with immunological tolerance, whereas IR has a contrary effect. IR could have a deleterious role in the activation of CD8T that can be relevant to new BrCa immunotherapy. C1 [Martin-Manzo, Victoria Miriam] Universidad Nacional Autonoma de Mexico, Programa de Doctorado en Ciencias BiomedicasMexico City, Mexico. [Lara, Carlos] Hospital General de Mexico Dr. Eduardo Liceaga, Servicio de OncologiaMexico City, Mexico. [Vargas-de-Leon, Cruz] Instituto Politecnico Nacional, Escuela Superior de MedicinaMexico City, Mexico. [Carrero, Julio] Universidad Nacional Autonoma de Mexico, Instituto de Investigaciones Biomedicas, Departamento de InmunologiaMexico City, Mexico. [Queipo, Gloria] Hospital General de Mexico Dr. Eduardo Liceaga, Departamento de GeneticaMexico City, Mexico. [Fonseca-Sanchez, Miguel] Hospital General de Mexico Dr. Eduardo Liceaga, Departamento de GeneticaMexico City, Mexico. [Mejia-Dominguez, R Nancy] CIC-Universidad Nacional Autonoma de Mexico, Red de Apoyo a la Investigacion (RAI)Mexico City, Mexico. [Kershenobich, David] Universidad Nacional Autonoma de Mexico, Facultad de Medicina, HIPAM, Unidad de Investigacion en Medicina ExperimentalMexico City, Mexico. [Mummidi, Srinivas] University of Texas Rio Grande Valley, South Texas Diabetes and Obesity InstituteEdinburg, TX, USA. [Zentella-Dehesa, Alejandro] Universidad Nacional Autonoma de Mexico, Departamento de Bioquimica INCMNSZ; Centro de Cancer, CM ABMexico City, Mexico. [Hernandez, Joselin] Universidad Nacional Autonoma de Mexico, Facultad de Medicina, HIPAM, Unidad de Investigacion en Medicina ExperimentalMexico City, Mexico. RP Hernandez, J (reprint author), Universidad Nacional Autonoma de Mexico, Facultad de Medicina, HIPAM, Unidad de Investigacion en Medicina Experimental, Mexico City, Mexico. EM joselin.hernandezruiz@utrgv.edu CR Sun Y-S, Zhao Z, Yang Z-N, Xu F, Lu HJ, Zhu ZY, ShiW, Jiang J, Yao PP, Zhu HP, 2017, Risk Factors and Preventions of Breast Cancer. Int J Biol Sci 13:1387–1397 SunW, Lu J,Wu S, Bi Y, Mu Y, Zhao J, Liu C, Chen L, Shi L, Li Q, Yang T, Yan L, Wan Q, Liu Y, Wang G, Luo Z, Tang X, Chen G, Huo Y, Gao Z, Su Q, Ye Z,Wang Y, Qin G, Deng H, Yu X, Shen F, Chen L, Zhao L, Wang T, Sun J, Xu M, Xu Y, Chen Y, Dai M, Zhang J, Zhang D, Lai S, Li D, Ning G, Wang W, 2016, Association of insulin resistance with breast, ovarian, endometrial and cervical cancers in non-diabetic women. Am J Cancer Res 6: 2334–2344 Gunter MJ, Xie X, Xue X, Kabat GC, Rohan TE, Wassertheil- Smoller S, Ho GYF, Wylie-Rosett J, Greco T, Yu H, Beasley J, Strickler HD, 2015, Breast cancer risk in metabolically healthy but overweight postmenopausal women. Cancer Res 75:270–274, Breast Cancer Risk in Metabolically Healthy but Overweight Postmenopausal Women Nam S, Park S, Park HS, Kim S, Kim JY, Kim SI, 2016, Association between insulin resistance and luminal B subtype breast cancer in postmenopausal women. Medicine, Baltimore, 95:e2825 Shanik MH, Xu Y, Skrha J, Dankner R, Zick Y, Roth J, 2008, Insulin resistance and hyperinsulinemia: is hyperinsulinemia the cart or the horse? Diabetes Care Suppl 2:S262-S268 Arcidiacono B, Iiritano S, Nocera A et al, 2012, Insulin resistance and cancer risk: AnOverview of the pathogenetic mechanisms. Exp Diabetes Res 2012:789174 Yadav A, Kataria MA, Saini V, Yadav A, 2013, Role of leptin and adiponectin in insulin resistance. Clin Chim Acta 18(417):80–84 Paz-Filho G, Mastronardi C, Wong ML, Licinio J, 2012, Leptin therapy, insulin sensitivity, and glucose homeostasis. Indian J Endocrinol Metab 16(Suppl 3):S549–S555 Paz-Filho G, Lim EL, Wong ML, Licinio J, 2011, Associations between adipokines and obesity-related cancer. Front Biosci, Landmark Ed, 16:1634–1650 Ali HR, Provenzano E, Dawson SJ, Blows FM, Liu B, Shah M, Earl HM, Poole CJ, Hiller L, Dunn JA, Bowden SJ, Twelves C, Bartlett JMS, Mahmoud SMA, Rakha E, Ellis IO, Liu S, Gao D, Nielsen TO, Pharoah PDP, Caldas C, 2014, Association between CD8+ infiltration and breast cancer survival in 12, 439 patients. Ann Oncol 25:1536–1543 Matsumot H, Thike AA, Li H et al, 2016, Increased CD4 and CD8- positive T cell infiltrate signifies good prognosis in a subset of triple-negative breast cancer. Breast Cancer Res Treat 156:237–247 Fischer HJ, Sie C, Schumnn E et al, 2017, The insulin receptor plays a critical role in T cell function and adaptive immunity. J Immunol 198:1910–1920 Han JM, Patterson SJ, Speck M, Ehses JA, Levings MK, 2014, Insulin inhibits IL-10-mediated regulatory T cell function: implications for obesity. J Immunol 192:623–629 GranadosHM, Draghi A 2nd, TsurutaniN et al, 2017, Programmed cell death-1, PD-1, is dysregulated in Tcells from children with new onset type 1 diabetes. PLoS One 12:e0183887 Buchbinder EI, Desai A, 2016, CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol 39:98–106 Jin HT, Ahmed R, Okazaki T, 2011, Role of PD-1 in regulating Tcell immunity. Curr Top Microbiol Immunol 350:17–37 Keir ME, Butte MJ, Freeman GJ, Sharpe AH, 2008, PD-1 and its ligands in tolerance and immunity.Annu Rev Immunol 26:677–704 Sharpe AH, Pauken KE, 2018, The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol 18:153–167 Muenst S, Soysal SD, Gao F, Obermann EC, Oertli D, Gillanders WE, 2013, The presence of programmed death 1, pd-1, positive tumor-infiltrating lymphocytes is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat 139:667–676 Ghebeh H, Mohammed S, Al-Omair A et al, 2006, The B7-H1, PD-L1, T lymphocyte-inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: correlation with important high-risk prognostic factors. Neoplasia 8:190–198 Poschke I, De Boniface J, Mao Y, Kiessling R, 2012, Tumorinduced changes in the phenotype of blood-derived and tumorassociated T cells of early stage breast cancer patients. Int J Cancer 131:1611–1620 Wherry EJ, Kurachi M, 2015, Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 15:486–499 Okoye IS, Houghton M, Tyrrell L, Barakat K, Elahi S, 2017, Coinhibitory Receptor Expression and Immune Checkpoint Blockade: Maintaining a Balance in CD8+ T Cell Responses to Chronic Viral Infections and Cancer. Front Immunol 8:1215 Fourcade J, Sun Z, Benallaoua M, Guillaume P, Luescher IF, Sander C, Kirkwood JM, Kuchroo V, Zarour HM, 2010, Upregulation of TIM3 and PD-1 expression is associated with tumor antigen-specific CD8+T cell dysfunction in melanoma patients. J Exp Med 207:2175–2186 Gorman JV, Colgan JD, 2014, Regulation of Tcell responses by the receptor molecule Tim-3. Immunol Res 59:56–65 Fuertes-Marraco SA, Neubert NJ, Verdeil G, Speiser DE, 2015, Inhibitory receptors beyond T cell exhaustion. Front Immunol 6:310 Kuss I, Schaefer C, Godfrey TE, Ferris RL, Harris JM, GoodingW, Whiteside TL, 2005, Recent thymic emigrants and subsets of naive and memory Tcells in the circulation of patients with head and neck cancer. Clin Immunol 116:27–36 Anderson AC, 2012, Tim-3, a negative regulator of anti-tumor immunity. Curr Opin Immunol 24:213–216 ZhangY, Cai P, Li L, Shi L, Chang P, Liang T, Yang Q, Liu Y,Wang L, Hu L, 2017, Co-expression of TIM-3 and CEACAM1 promotes T cell exhaustion in colorectal cancer patients. Int Immunopharmacol 43:210–218 Liu H, Zhi L, Duan N, Su P, 2016, Abnormal expression of Tim-3 antigen on peripheral blood T cells is associated with progressive disease in osteosarcoma patients. FEBS Open Bio 6:807–815 Wu J, Liu C, Qin S, Hou H, 2013, The expression of Tim3 in peripheral blood of ovarian cancer. DNA Cell Biol 32:648–653 Avery L, Filderman J, Szymczak-Worman AL, Kane LP, 2018, Tim-3 co-stimulation promotes short-term effector T cells, restricts memory precursors and is dispensable for T cell exhaustion. Proc Natl Acad Sci U S A 115(10):2455–2460 Qiu Y, Chen J, Liao H et al, 2012, Tim-3-expressing CD4(+, and CD8(+, T cells in human tuberculosis, TB, exhibit polarized effector memory phenotypes and stronger anti-TB effector functions. PLoS Pathog 8:e1002984 Gorman JV, Starbeck-Miller G, Pham NL et al, 2014, Tim-3 directly enhances CD8 T cell responses to acute Listeria monocytogenes infection. J Immunol 192:3133–3142 Meggyes M, Miko E, Polgar B, Bogar B, Farkas B, Illes Z, Szereday L, 2014, Peripheral blood TIM-3 positive NK and CD8+ T cells throughout pregnancy: TIM3/galectin-9 interaction and its possible role during pregnancy. PLoS One 9:e92371 Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, Otsu M, Hara K, Ueki K, Sugiura S, Yoshimura K, Kadowaki T, Nagai R, 2009, CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 15:914–920 DeNardo DG, Coussens LM, 2007, Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression. Breast Cancer Res 9:212 Pierce BL, Ballard-Barbash R, Bernstein L, Baumgartner RN, Neuhouser ML, Wener MH, Baumgartner KB, Gilliland FD, Sorensen BE, McTiernan A, Ulrich CM, 2009, Elevated biomarkers of inflammation are associated with reduced survival among breast cancer patients. J Clin Oncol 27:3437–3444 Patsoukis N, Bardhan K, Chatterjee P, Sari D, Liu B, Bell LN, Karoly ED, Freeman GJ, Petkova V, Seth P, Li L, Boussiotis VA, 2015, PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun 6:6692 MaedaN, Shimomura I,KishidaK et al, 2002, Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nat Med 8(7): 731–737 Yadav A, Jyoti P, Jain SK, Bhattacharjee J, 2011, Correlation of adiponectin and leptin with insulin resistance: a pilot study in healthy north Indian population. Indian J Clin Biochem 26(2): 193–196 Osegbe I, Okpara H, Azinge E, 2016, Relationship between serum leptin and insulin resistance among obese Nigerian women. Ann Afr Med 15(1):14–9 Carbone F, La Rocca C, Matarese G, 2012, Immunological functions of leptin and adiponectin. Biochimie 94(10):2082–8, 2088 Tsang JY, Li D, Ho D, Peng J et al, 2011, Novel immunomodulatory effects of adiponectin on dendritic cell functions. Int Immunopharmacol 11(5):604–609 Rodriguez L, Graniel J, Ortiz R, 2007, Effect of leptin on activation and cytokine synthesis in peripheral blood lymphocytes of malnourished infected children. Clin Exp Immunol 148(3):478–485 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1233 EP 1243 DI 10.1007/s12253-019-00610-7 PG 11 ER PT J AU Zhao, W Chen, Sh Hou, X Liao, Q Chen, G Zhao, Y AF Zhao, Wenjing Chen, Shaobo Hou, Xianming Liao, Quan Chen, Ge Zhao, Yupei TI Predictive Factors of Lateral Lymph Node Metastasis in Papillary Thyroid Microcarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary thyroid microcarcinoma; Lateral lymph node metastasis; Skip metastasis; Predictors ID Papillary thyroid microcarcinoma; Lateral lymph node metastasis; Skip metastasis; Predictors AB This study was designed to determine the incidence and predictive factors for lateral lymph node metastasis (LNM) in patients with papillary thyroid microcarcinoma (PTMC). From January 2014 to July 2015, a retrospective review was conducted of 215 patients with PTMC who underwent total thyroidectomy and central lymph node dissection (LND) with lateral LND. Correlations of lateral LNM with clinicopathological features were examined using univariate analyses. Risk factors for lateral LNM were identified by multivariate analysis. Lateral LNM was observed in 163(75.8%) cases of 215 patients and often involved in level III (82.2%) and level IV (65.6%), with most found in two-levels (41.1%) and single-level (33.7%) models. Multivariate analyses showed that central LNM(odds ratio [OR]: 8.28, 95%confidence interval [CI]: 3.43–19.98, p < 0.001) and upper portion location (OR: 2.87 [CI: 1.34–6.09]; p = 0.007) were independent predictive factors for lateral LNM. The incidence of skip metastasis-Lateral LNM with central Lymph nodes negative-was 8.6% (14/163). Age ≥ 45 years old (OR: 4.37 [CI: 1.14– 16.66]; p = 0.031) and upper portion location (OR: 4.34 [CI: 1.27–14.78]; p = 0.019) were independent risk factors for skip metastasis by multivariate analyses. Taken together, patients with PTMC with central LNM and tumor in the upper pole were more likely to present with lateral LNM. Even if there was no central LNM, patients with an age ≥ 45 years old and tumors in the upper portion of the thyroid should be evaluated carefully for possible lateral LNM. C1 [Zhao, Wenjing] Chinese Academy of Medical Science & Peking Union Medical College, Peking Union Medical College Hospital, Central Laboratory, 1 Shuai Fu Yuan Hu Tong, 100730 Beijing, China. [Chen, Shaobo] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuai Fu Yuan Hu Tong, 100730 Beijing, China. [Hou, Xianming] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuai Fu Yuan Hu Tong, 100730 Beijing, China. [Liao, Quan] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuai Fu Yuan Hu Tong, 100730 Beijing, China. [Chen, Ge] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuai Fu Yuan Hu Tong, 100730 Beijing, China. [Zhao, Yupei] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 1 Shuai Fu Yuan Hu Tong, 100730 Beijing, China. RP Chen, G (reprint author), Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, 100730 Beijing, China. EM pumchchenge@163.com CR Yu XM, Wan Y, Sippel RS, Chen H, 2011, Should all papillary thyroid microcarcinomas be aggressively treated? An analysis of 18,445 cases. Ann Surg 254:653–660 Chow SM, Law SC, Chan JK, Au SK, Yau S, Lau WH, 2003, Papillary microcarcinoma of the thyroid: prognostic significance of lymph node metastasis and multifocality. Cancer 98:31–40 Pisanu A, Reccia I, Nardello O,UcchedduA, 2009, Risk factors for nodal metastasis and recurrence among patients with papillary thyroid microcarcinoma: differences in clinical relevance between nonincidental and incidental tumors. World J Surg 33:460–468 Hay ID, Hutchinson ME, Gonzalez Losada T, 2008, Papillary thyroid microcarcinoma: a study of 900 cases observed in a 60-year period. Surgery 144:980–988 Machens A, Hinze R, Thomusch O, Dralle H, 2002, Pattern of nodal metastasis for primary and reoperative thyroid cancer. World J Surg 26:22–28 Shaha AR, 1998, Management of the neck in thyroid cancer. Otolaryngol Clin N Am 31:823–831 Machens A, Holzhausen H, Dralle H, 2004, Skip metastases in thyroid cancer leaping the. Central lymph node compartment. Arch Surg 139:43–45 Lim YC, Koo BS, 2012, Predictive factors of skip metastases to lateral neck compartment. Leaping central neck compartment in papillary thyroid carcinoma. Oral Oncol 48:262–265 Lee YS, Shin SC, Lim YS et al, 2014, Tumor location–dependent skip lateral cervical lymph node metastasis in papillary thyroid cancer. Head Neck 36:887–891 Wada N, Duh QY, Sugino K et al, 2003, Lymph node metastasis from 259 papillary. Thyroidmicrocarcinomas: frequency, pattern of occurrence and recurrence, and optimal strategy for neck dissection. Ann Surg 237:399–407 Kim YS, 2012, Patterns and predictive factors of lateral lymph node metastasis in papillary thyroid microcarcinoma. Otolaryngol Head Neck Surg 147:15–19 Chung YS, Kim JY, Bae JS et al, 2009, Lateral lymph node metastasis in papillary thyroid carcinoma: results of therapeutic lymph node dissection. Thyroid 19:241–246 Kim TY, Hong SJ, Kim JM et al, 2008, Prognostic parameters for recurrence of papillary thyroid microcarcinoma. BMC Cancer 8:296 Pelizzo MR, Boschin IM, Toniato A et al, 2004, Natural history, diagnosis, treatment and outcome of papillary thyroid microcarcinoma, PTMC): a mono-institutional 12-year experience. Nucl Med Commun 25:547–552 Roh JL, Kim JM, Park CI, 2008, Lateral cervical lymph node metastases from papillary thyroid carcinoma: pattern of nodal metastases and optimal strategy for neck dissection. Ann Surg Oncol 15:1177–1182 Kwak JY, Kim EK, Kim MJ et al, 2009, Papillary microcarcinoma of the thyroid: predicting factors of lateral neck node metastasis. Ann Surg Oncol 16:1348–1355 Lee KJ, Cho YJ, Kim SJ et al, 2011, Analysis of the clinicopathologic features of papillary thyroid microcarcinoma based on 7-mm tumor size. World J Surg 35:318–323 Yang Y, Chen C, Chen Z et al, 2014, Prediction of central compartment lymph node metastasis in papillary thyroid microcarcinoma. Clin Endocrinol 81:282–288 Zeng RC, Li Q, Lin KL et al, 2012, Predicting the factors of lateral lymph node metastasis in papillary microcarcinoma of the thyroid in eastern China. Clin Transl Oncol 14:842–847 Kwak JY, Kim EK, Youk JH et al, 2008, Extrathyroid extension of well-differentiated papillary thyroid microcarcinoma on US. Thyroid 18:609–614 Lim YS, Lee JC, Lee YS et al, 2011, Lateral cervical lymph node metastases from papillary thyroid carcinoma: predictive factors of nodal metastasis. Surgery 150:116–121 Lin KL,Wang OC, Zhang XH, DaiXX, Hu XQ, Qu JM, 2010, The BRAF mutation is predictive of aggressive Clinicopathological characteristics in papillary thyroid microcarcinoma. Ann Surg Oncol 17:3294–3300 Ito Y, Tomoda C, Uruno T et al, 2004, Papillary microcarcinoma of the thyroid: how should it be treated? World J Surg 28:1115–1121 Chung YS, Kim JY, Bae JS et al, 2009, Lateral lymph node metastasis in papillary thyroid carcinoma: results of therapeutic lymph node dissection. Thyroid 19:241–245 Verburg FA, Mader U, Tanase K et al, 2013, Life expectancy is reduced in differentiated thyroid cancer patients≥45 years old with extensive local tumor invasion, lateral lymph node, or distant metastases at diagnosis and normal in all other DTC patients. J Clin Endocrinol Metab 98:172–180 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1245 EP 1251 DI 10.1007/s12253-018-0511-8 PG 7 ER PT J AU Khemka, R Gupta, M Jena, KN AF Khemka, Rashi Gupta, Monica Jena, Kanta Nehar TI CML with Megakaryocytic Blast Crisis: Report of 3 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chronic Myeloid Leukemia; Megakaryocytic blast crisis; Flowcytometric immunophenotyping ID Chronic Myeloid Leukemia; Megakaryocytic blast crisis; Flowcytometric immunophenotyping AB Chronic myelogenous leukemia (CML) is a chronic myeloproliferative neoplasm consistently associated with the BCR-ABL1 fusion gene located in the Philadelphia chromosome. The Blast Phase is diagnosed when blasts are ≥20% of the peripheral blood white cell count or of bone marrow nucleated cells or when there is an extramedullary blast proliferation. Megakaryocytic blast crisis as the presenting manifestation of CML is extremely rare and only 7 reported cases were found in the literature. Out of 34 cases of CML-Blast Phase between April 2015 and June 2016, 3 cases showed megakaryocytic differentiation. 2 of these presented in Blast phase as the first manifestation of CML and the third case was a known case of CML-Chronic phase. Flow cytometric immunophenotyping was performed on peripheral blood/bone marrow using 6- color flow cytometer Navios. On CD45 vs SSC two distinct populations of blasts were seen in two cases and single population in the third case. All the 3 cases were positive for CD61, cCD41, cCD61 confirming the megakaryocytic lineage. The clinical features, morphologic and cytogenetic findings help in the identification and distinction of megakaryocytic blast phase of CML from Acute Megakayoblastic Leukemia. The diagnosis of such rare presentation of CML is essential for determining the choice of treatment. Therefore including a megakaryocytic marker in the primary flow cytometry panel is important so that these cases are not underdiagnosed as Acute myeloid leukemia because of expression of CD13 and CD33 only. C1 [Khemka, Rashi] Delhi State Cancer Institute, Department of Oncopathology, Dilshad Garden, 110095 Delhi, India. [Gupta, Monica] Delhi State Cancer Institute, Department of Oncopathology, Dilshad Garden, 110095 Delhi, India. [Jena, Kanta Nehar] Delhi State Cancer Institute, Department of Clinical Oncology, Dilshad Garden, 110095 Delhi, India. RP Gupta, M (reprint author), Delhi State Cancer Institute, Department of Oncopathology, 110095 Delhi, India. EM drmonica123@gmail.com CR Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW, 2016, The 2016 revision to the World Health Organization, WHO, classification of myeloid neoplasms and acute leukemia. Blood 127:2391–2405. , DOI 10.1182/blood-2016-03-643544 Wu CD, Medeiros LJ, Miranda RN, Mark HF, Rintels P, 1996, Chronicmyeloid leukemiamanifested duringmegakaryoblastic crisis. South Med J 89:422–427 Pelloso LA, Baiocchi OC, ChauffailleML, Yama-motoM, Hungria VT, Bordin JO, 2002, Megakaryocytic blast crisis as a first presentation of chronic myeloid leukemia. Eur J Haematol 69:58–61 Bryant BJ, Alperin JB, Elghetany MT, 2007, Paraplegia as the presentingmanifestation of extramedullary megakaryoblastic transformation of previously undiagnosed chronic myelogenous leukemia. Am J Hematol 82:150–154 Campiotti L, GrandiAM, BiottiMG, Ultori C, Solbiati F, Codari R, Venco A, 2007, Megakaryocytic blast crisis as first presentation of chronic myeloid leukemia. Am J Hematol 82:231–233 Pullarkat ST, Vardiman JW, SlovakML, Rao DS, Rao NP, BedellV, Said JW, 2008, Megakaryocytic blast crisis as a presenting manifestation of chronic myeloid leukemia. Leuk Res 32:1770–1775 Al-Shehri A, Al-Seraihy A, Owaidah TM, Belgaumi AF, 2010, Megakaryocytic blast crisis at presentation in a pediatric patient with chronic myeloid leukemia. Hematol Oncol Stem Cell Ther 3(1):42–46 Karkuzhali P, ShanthiV, Usha TA(2013)Acase of chronicmyeloid leukaemia presenting as megakaryocytic blast crisis, AML M7). ecancermedicalscience 7:375 Cherian S, Wood B, 2012, Case 31. In: Cherian S, Wood B, eds, Flow cytometry in evaluation of hematopoietic neoplasms: A case based approach. College of American Pathologists, Northfield, pp 149–151 Soupir CP, Vergilio JA, Dal Cin P, 2007, Philadelphia chromosome-positive acute myeloid leukemia: a rare aggressive leukemia with clinicopathologic features distinct from chronic myeloid leukemia in myeloid blast crisis. Am J Clin Pathol 127(4): 642–650 Jarmuz M, Kroll R, Przybylowicz-Chalecka A, Ratajczak B, Gniot M, Szyfter K, Komarnicki M, 2010, Megakaryocytic blast crisis in a chronic myeloid leukaemia patient with a rare variant of Philadelphia rearrangement t(9;22;22, and a constitutional translocation t(3;7). Cancer Genet Cytogenet 199(1):45–47 Morris EL, Dutcher JP, 2005, Blastic phase of chronic myelogenous leukemia. Clin Adv Hematol Oncol 3:547–552 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1253 EP 1258 DI 10.1007/s12253-018-0484-7 PG 6 ER PT J AU da Rocha Boeira, Th Wolf, MJ Coser, J Grivicich, I Simon, D Lunge, RV AF da Rocha Boeira, Thais Wolf, Michel Jonas Coser, Janaina Grivicich, Ivana Simon, Daniel Lunge, Ricardo Vagner TI Polymorphisms in Genes Related to Cervical Cancer in A Brazilian Population: A Case-Control Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [da Rocha Boeira, Thais] Universidade Luterana do Brasil, PPG Biologia Celular e Molecular Aplicada a SaudeCanoas, RS, Brazil. [Wolf, Michel Jonas] Universidade Luterana do Brasil, PPG Biologia Celular e Molecular Aplicada a SaudeCanoas, RS, Brazil. [Coser, Janaina] Universidade de Cruz Alta / Universidade Regional do Noroeste do Estado do Rio Grande do Sul (UNICRUZ/UNIJUÍ), Programa de Pos-Graduacao em Atencao Integral a SaudeCruz Alta / Ijui, RS, Brazil. [Grivicich, Ivana] Universidade Luterana do Brasil, PPG Biologia Celular e Molecular Aplicada a SaudeCanoas, RS, Brazil. [Simon, Daniel] Universidade Luterana do Brasil, PPG Biologia Celular e Molecular Aplicada a SaudeCanoas, RS, Brazil. [Lunge, Ricardo Vagner] Universidade Luterana do Brasil, PPG Biologia Celular e Molecular Aplicada a SaudeCanoas, RS, Brazil. RP Wolf, MJ (reprint author), Universidade Luterana do Brasil, PPG Biologia Celular e Molecular Aplicada a Saude, Canoas, Brazil. EM jonasmwolf@gmail.com CR IARC, International Agency for Research on Cancer. Pharmaceuticals: a review of human carcinogens. Lyon, 2012., IARC monographs on the evaluation of carcinogenic risks to humans, v. 100E) Magnusson PK, Sparen P, Gyllensten UB, 1999). Genetic link to cervical tumours. Nature. 1;400(6739):29–30 Wang SS, BrattiMC, Rodriguez AC, Herrero R, Burk RD, Porras C, Gonzalez P, Sherman ME, Wacholder S, Lan ZE, Schiffman M, Chanock SJ, Hildesheim A, 2009, Common variants in immune and DNA repair genes and risk for human papillomavirus persistence and progression to cervical cancer. J Infect Dis 199(1):20–30 Wang SS, Gonzalez P, Yu K, Porras C, Li Q, SafaeianM, Rodriguez AC, Sherman ME, Bratti C, Schiffman M, Wacholder S, Burk RD, Herrero R, Chanock SJ, Hildesheim A, 2010, Common genetic variants and risk for HPV persistence and progression to cervical cancer. PLoS One 5(1):e8667 Coser J, Boeira Tda R, Wolf JM et al, 2016, Cervical human papillomavirus infection and persistence: a clinic-based study in the countryside from South Brazil. Braz J Infect Dis 20(1):61–68 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1259 EP 1261 DI 10.1007/s12253-018-0414-8 PG 3 ER PT J AU Mandal, P AF Mandal, Paramita TI Recent advances of Blood telomere length (BTL) shortening: A potential biomarker for development of cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE Telomere length; Shortening; Cancer; Cisease; Biomarker ID Telomere length; Shortening; Cancer; Cisease; Biomarker AB Telomeres, the specific DNA–protein structures remains at both ends of each chromosome and are crucial in the maintenance of chromosome integrity and genomic stability with protection of the chromosome from damage and degradation.. Increasing evidences suggest the correlation between telomere length and the development of cancers, but the findings remain obscure. Generally, the average length of telomere repeats at the ends of chromosomes that gives a clue in providing indirect information about their mitotic history. It plays immense role in preventing genome from nucleolytic degradation, unnecessary recombination, repair, and interchromosomal fusion. It has major role in storing the information in the genome. Telomere attrition during successive cell divisions induces chromosomal instability and contributes significantly to genomic rearrangements that can result in tumorigenesis. Convincing evidence documented that a meagre portion of telomeric DNA is expelled out during mitotic stage of cell division. But accelerated shortening telomere length at critical level triggers senescence and/or apoptosis. Various harmful agents with bad lifestyles are responsible in inducing shortening of telomere length with damage of DNA resulting to occurrence of disease with shortening of lifespan. Besides, telomerases, the specialized polymerase that synthesizes new telomere repeats and is strongly associated with cancer facilitating malignant transformation. Therefore, in the study, it is highlighted that the telomeres may play diverse roles in different cancers whereas shortening of telomere length may be risk factors for the development of tumors. C1 [Mandal, Paramita] The University of Burdwan, Department of ZoologyBurdwan, West Bengal, India. RP Mandal, P (reprint author), The University of Burdwan, Department of Zoology, Burdwan, India. EM paramita.mandal2@gmail.com CR Hirashima K, Migita T, Sato S, MuramatsuY, Ishikawa Y, Seimiyaa H, 2013, Telomere Length Influences Cancer Cell Differentiation In Vivo. Molecular and Cellular Biology 33(15):2988–2995 Leteurtre F, Li X, Guardiola P, Le Roux G, Sergere J-C, Richard P, Carosella ED, Gluckman E, 1999, Accelerated telomere shortening and telomerase activation in Fanconi’s anaemia. Br J Haematol 105(4):883–893 Willeit P, Willeit J, Mayr A, Oberhollenzer F, Brandstatter A, Kronenberg F, Kiechl S, 2010, 2010. Telomere Length and Risk of Incident Cancer and Cancer Mortality JAMA 304(1):69–75 Jafri MA, Ansari SA, Alqahtani MH, Shay JW, 2016, Roles of telomeres and telomerase in cancer, and advances in telomerasetargeted therapies. Genome Medicine 8:69 Moon IK, Jarstfer MB, 2007, The human telomere and its relationship to human disease, therapy, and tissue engineering. Frontiers in Bioscience 12(8-12):4595 Zhu X, Han W, Xue W, Zou Y, Xie C, Du J, Jin G, 2016, The association between telomere length and cancer risk in population studies. Scientific Reports 6(1) Shay JW, 2013, Are Short Telomeres Predictive of Advanced Cancer? Cancer Discovery 3(10):1096–1098 Greenberg RA, Chin L, Femino A, Lee KH, Gottlieb GJ, Singer RH, Greider CW, DePinho RA, 1999, Short dysfunctional telomeres impair tumourigenesis in the INK4a(delta2/3, cancer-prone mouse. Cell 97:515–525 Nasir NF, Kannan TP, Sulaiman SA, Shamsuddin S, Azlina A, Stangaciu S, 2015, The relationship between telomere length and beekeeping among Malaysians. AGE 37(3) Khan S, Chuturgoon AA, Naidoo DP, 2012, Telomeres and atherosclerosis. Cardiovascular journal of Africa 23(10):563–571 Van Steensel B, Smogorzewska A, de Lange T, 1998, TRF2 protects human telomeres from end-to-end fusions. Cell 92:401–413 Stewart JA, Chaiken MF, Wang F, Price CM, 2012, Maintaining the end: roles of telomere proteins in end-protection, telomere replication and length regulation. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis 730(1):12–19 Martinez P, Blasco MA, 2011, Telomeric and extra-telomeric roles for telomerase and the telomere-binding proteins. Nat Rev Cancer 11:161–176 Ma H, Zhou Z, Wei S, Liu Z, Pooley KA, Dunning AM, Svenson U, Goran R, Dean Hosgood H, Shen M, Wei Q, Toland AE, 2011, Shortened Telomere Length Is Associated with Increased Risk of Cancer: A Meta-Analysis. PLoS ONE 6(6):e20466 Cheung AL, Deng W, 2008, Telomere dysfunction, genome instability and cancer. Front Biosci 13(13):2075 Cottliar AS, Slavutsky IR, 2001, Telomeres and telomerase activity: their role in aging and in neoplastic development. Medicina, B Aires). 61(3):335–342 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1263 EP 1265 DI 10.1007/s12253-018-0425-5 PG 3 ER PT J AU Sarode, SG Sarode, CS Gadbail, RA Gondivkar, Sh Patil, Sh AF Sarode, S Gargi Sarode, C Sachin Gadbail, R Amol Gondivkar, Shailesh Patil, Shankargouda TI Tumor Associated Tissue Eosinophilia in Ameloblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Sarode, S Gargi] Dr. D. Y. Patil Dental College and Hospital, Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Vidyapeeth, Sant-Tukaram nagar, Pimpri, 411018 Pune, Maharashtra, India. [Sarode, C Sachin] Dr. D. Y. Patil Dental College and Hospital, Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Vidyapeeth, Sant-Tukaram nagar, Pimpri, 411018 Pune, Maharashtra, India. [Gadbail, R Amol] Indira Gandhi Government Medical College and Hospital, Department of DentistryNagpur, Maharashtra, India. [Gondivkar, Shailesh] Government Dental College & Hospital, Department of Oral Medicine and RadiologyNagpur, Maharashtra, India. [Patil, Shankargouda] Jazan University, College of Dentistry, Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral PathologyJazan, Saudi Arabia. RP Sarode, CS (reprint author), Dr. D. Y. Patil Dental College and Hospital, Department of Oral Pathology and Microbiology, 411018 Pune, India. EM drsachinsarode@gmail.com CR Lowe D, Jorizzo J, Hutt MS, 1981, Tumour-associated eosinophilia: a review. J Clin Pathol 34:1343–1348 Lorena SCM, Oliveira DT, Dorta RG, Landman G, Kowalski LP, 2003, Eotaxin expression in oral squamous cell carcinomas with andwithout tumour associated tissue eosinophilia.OralDis 9:279–283 Debta P, Debta FM, Chaudhary M,Wadhwan V, 2010, Evaluation of infiltration of immunological cells, tissue eosinophil and mast cell, in odontogenic cysts by using special stains. J Clin Cell Immunol 1:103 Tasaki T, Matsuyama A, Tabata T, Suzuki H, Yamada S, Sasaguri Y, HisaokaM, 2013, Sclerosing mucoepidermoid carcinoma with eosinophilia of the salivary gland: case report and review of the literature. Pathol Int 63(2):125–131 Fridlender ZG, Simon H-U, Shalit M, 2003, Metastatic carcinoma presenting with concomitant eosinophilia and thromboembolism. Am J Med Sci 326:98–101 Mingomataj EC, 2008, Eosinophil-induced prognosis improvement of solid tumors could be enabled by their vesicle-mediated barrier permeability induction. Med Hypotheses 70:582–584 Moezzi J, Gopalswamy N, Haas RJ Jr, Markert RJ, Suryaprasad S, Bhutani MS, 2000, Stromal eosinophilia in colonic epithelial neoplasms. Am J Gastroenterol 95(2):520–523 Uitto VJ, Ylipaavalniemi P, 1977, Alterations in collagenolytic activity caused by jaw fluids. Proc Finn Dent Soc 73:197–202 de Assis Caldas Pereira F, Gurgel CA, Ramos EA, Vidal MT, Pinheiro AL, Jurisic V, Sales CB, Cury PR, dos Santos JN, 2012, Distribution of mast cells in benign odontogenic tumors. Tumour Biol 33(2):455–461 Stewart S, EdwardEM(2001, Immunology, immunopathology and immunity, 6th edn. ASM, Washington D.C NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2019 VL 25 IS 3 BP 1267 EP 1268 DI 10.1007/s12253-018-0499-0 PG 2 ER PT J AU Fucic, A Aghajanyan, A Culig, Z Le Novere, N AF Fucic, Aleksandra Aghajanyan, Anna Culig, Zoran Le Novere, Nicolas TI Systems Oncology: Bridging Pancreatic and Castrate Resistant Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Pancreatic cancer; Castrate resistant prostate cancer; Cancer marker; Systems oncology; Cancer profiling ID Pancreatic cancer; Castrate resistant prostate cancer; Cancer marker; Systems oncology; Cancer profiling AB Large investments by pharmaceutical companies in the development of new antineoplastic drugs have not been resulting in adequate advances of new therapies. Despite the introduction of new methods, technologies, translational medicine and bioinformatics, the usage of collected knowledge is unsatisfactory. In this paper, using examples of pancreatic ductal adenocarcinoma (PaC) and castrate-resistant prostate cancer (CRPC), we proposed a concept showing that, in order to improve applicability of current knowledge in oncology, the re-clustering of clinical and scientific data is crucial. Such an approach, based on systems oncology, would include bridging of data on biomarkers and pathways between different cancer types. Proposed concept would introduce a new matrix, which enables combining of already approved therapies between cancer types. Paper provides a (a) detailed analysis of similarities in mechanisms of etiology and progression between PaC and CRPC, (b) diabetes as common hallmark of both cancer types and (c) knowledge gaps and directions of future investigations. Proposed horizontal and vertical matrix in cancer profiling has potency to improve current antineoplastic therapy efficacy. Systems biology map using Systems Biology Graphical Notation Language is used for summarizing complex interactions and similarities of mechanisms in biology of PaC and CRPC. C1 [Fucic, Aleksandra] Institute for Medical Research and Occupational Health, Ksaverska c 2, 10000 Zagreb, Croatia. [Aghajanyan, Anna] Peoples’ Friendship University of Russia, Institute of MedicineMoscow, Russian Federation. [Culig, Zoran] Medical University of Innsbruck, Department of UrologyInnsbruck, Austria. [Le Novere, Nicolas] Babraham InstituteCambridge, UK. RP Fucic, A (reprint author), Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia. EM afucic@imi.hr CR Jemal A, Bray F, CenterMM, Ferlay J,Ward E, Forman D, 2011, Global Cancer statistics CA. Cancer J Clin 61:69–90 Xiao AY, Tan ML, Wu LM, Asrani VM, Windsor JA, Yadav D, Petrov MS, 2016, Global incidence and mortality of pancreatic diseases: a systematic review, meta-analysis, and meta-regression of population-based cohort studies. Lancet Gastroenterol Hepatol 1(1):45–55 Hirst CJ, Cabrera C, Kirby M, 2012, Epidemiology of castration resistant prostate cancer: a longitudinal analysis using a UK primary care database. Cancer Epidemiol 36(6):e349–e353 SteinkampMP, O'Mahony OA, BrogleyM, Rehman H, Lapensee EW, Dhanasekaran S, HoferMD, Kuefer R, Chinnaiyan A, Rubin MA, Pienta KJ, Robins DM, 2009, Treatment-dependent androgen receptor mutations in prostate cancer exploit multiple mechanisms to evade therapy. Cancer Res 69(10):4434–4442 So A, GleaveM, Hurtado-Col A, Nelson C, 2005)Mechanisms of the development of androgen independence in prostate cancer. World J Urol 23(1):1–9 Mostaghel EA, Page ST, Lin DW, Fazli L, Coleman IM, True LD, Knudsen B, Hess DL, Nelson CC, Matsumoto AM, BremnerWJ, Gleave ME, Nelson PS, 2007, Intraprostatic androgens and androgen-regulated gene expression persist after testosterone suppression: therapeutic implications for castration-resistant prostate cancer. Cancer Res 67(10):5033–5041 Montgomery RB, Mostaghel EA, Vessella R, Hess DL, Kalhorn TF, Higano CS, True LD, Nelson PS, 2008, Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res 68(11):4447– 4454 Locke JA, Guns ES, Lubik AA, Adomat HH, Hendy SC, Wood CA, Ettinger SL, GleaveME, Nelson CC, 2008, Androgen levels increase by intratumoral de novo steroidogenesis during progression of castration-resistant prostate cancer. Cancer Res 68(15):6407–6415 Leon CG, Locke JA, Adomat HH, Etinger SL, Twiddy AL, Neumann RD, Nelson CC, Guns ES, Wasan KM, 2010, Alterations in cholesterol regulation contribute to the production of intratumoral androgens during progression to castrationresistant prostate cancer in a mouse xenograft model. Prostate 70(4):390–400 Kolodecik T, Shugrue C, Ashat M, Thrower EC, 2014, Risk factors for pancreatic cancer: underlying mechanisms and potential targets. Front Physiol 4:415 Bardeesy N, DePinho RA, 2002, Pancreatic cancer biology and genetics. Nat Rev Cancer 2:897–909 Polireddy K, Chen Q, 2016, Cancer of the pancreas: molecular pathways and current advancement in treatment. J Cancer 7(11): 1497–1514 Becker AE, Hernandez YG, Frucht H, Lucas AL, 2014, Pancreatic ductal adenocarcinoma: risk factors, screening, and early detection. World J Gastroenterol 20(32):11182–11198 Rhee H, Gunter JH, Heathcote P, Ho K, Stricker P, Corcoran NM, Nelson CC, 2015, Adverse effects of androgen-deprivation therapy in prostate cancer and their management. BJU Int 5(115 Supplement):3–13 Vermeulen A, Kaufman JM, Goemaere S, van Pottelberg I, 2002, Estradiol in elderly men. Aging Male 5:98–102 Navarro Silvera SA, Miller AB, Rohan TE, 2005, Hormonal and reproductive factors and pancreatic cancer risk: a prospective cohort study. Pancreas 30(4):369–374 Sun F, ZhangZW, Tan EM, Lim ZL, Li Y,Wang XC, Chua SE, Li J, Cheung E, Yong EL, 2016, Icaritin suppresses development of neuroendocrine differentiation of prostate cancer through inhibition of IL-6/STAT3 and aurora kinase a pathways in TRAMP mice. Carcinogenesis 37(7):701–711 Kovvali G, 2014, Systems oncology: a new paradigm in cancer research. J Carcinog 13:6 Chung WS, Stainier DY, 2008, Intra-endodermal interactions are required for pancreatic beta cell induction. Dev Cell 14(4):582– 593 Habener JF, Kemp DM, Thomas MK, 2005, Minireview: transcriptional regulation in pancreatic development. Endocrinology 146(3):1025–1034 Kim SK,MacDonald RJ, 2002, Signaling and transcriptional control of pancreatic organogenesis. Curr Opin Genet Dev 12(5):540– 547 Sugiyama T, Benitez CM, Ghodasara A, Liu L, McLean GW, Lee J, Blauwkamp TA, Nusse R, Wright CV, Gu G, Kim SK, 2013, Reconstituting pancreas development from purified progenitor cells reveals genes essential for islet differentiation. Proc Natl Acad Sci U S A 110(31):12691–12696 Kim MS, Lee DY, 2015, Insulin-like growth factor, IGF)-I and IGF binding proteins axis in diabetes mellitus. Ann Pediatr Endocrinol Metab 20(2):69–73 Murtaugh LC, 2008, The what, where, when and how of Wnt/β- catenin signaling in pancreas development. Organogenesis 4(2): 81–86 Lee SH, Johnson DT, Luong R,Yu EJ, Cunha GR, Nusse R, Sun Z, 2015, Wnt/β-catenin-responsive cells in prostatic development and regeneration. Stem Cells 33(11):3356–3367 Prins GS, Putz O, 2008, Review molecular signaling pathways that regulate prostate gland development. Differentiation 76(6): 641–659 Ma F, Ye H, He HH, Gerrin SJ, Chen S, Tanenbaum BA, Cai C, Sowalsky AG, He L, Wang H, Balk SP, Yuan X, 2016, SOX9 drives WNT pathway activation in prostate cancer. J Clin Invest 126(5):1745–1758 Su Q, Xin L, 2016, Notch signaling in prostate cancer: refining a therapeutic opportunity. Histol Histopathol 31(2):149–157 Gajula RP, Chettiar ST,Williams RD, Nugent K, Kato Y,Wang H, Malek R, Taparra K, Cades J, Annadanam A, Yoon AR, Fertig E, Firulli BA, Mazzacurati L, Burns TF, Firulli AB, An SS, Tran PT, 2015, Structure-function studies of the bHLH phosphorylation domain of TWIST1 in prostate cancer cells. Neoplasia 17(1):16– 31 Zhu G, Zhau HE, He H, Zhang L, Shehata B,Wang X, Cerwinka WH, Elmore J, He D, 2007, Sonic and desert hedgehog signaling in human fetal prostate development. Prostate 67(6):674–684 Jo A, Denduluri S, Zhang B,Wang Z, Yin L, Yan Z, Kang R, Shi LL,Mok J, LeeMJ, Haydon RC, 2014, The versatile functions of Sox9 in development, stem cells, and human diseases. Genes Diseases 1(2):149–161 Yu M, Gipp J, Yoon JW, Iannaccone P, Walterhouse D, Bushman W(2009, Sonic hedgehog-responsive genes in the fetal prostate. J Biol Chem 284(9):5620–5629 Verras M, Brown J, Li X et al, 2004)Wnt3a growth factor induces androgen receptor-mediated transcription and enhances cell growth in human prostate cancer cells. Cancer Res 64(24):8860– 8866 Nakamoto M, HisaokaM(2016, Clinicopathological implications of wingless/int1, WNT, signaling pathway in pancreatic ductal adenocarcinoma. J UOEH 38(1):1–8 Terris B, Cavard C, 2014, Diagnosis and molecular aspects of solid- pseudopapillary neoplasms of the pancreas. Semin Diagn Pathol 31(6):484–490 Corbishley TP, Iqbal MJ, Wilkinson ML et al, 1986, Androgen receptor in human normal and malignant pancreatic tissue and cell lines. Cancer 57(10):1992–1995 Glass JP, Parasher G, Arias-Pulido H et al, 2011, Mesothelin and GPR30 staining among a spectrum of pancreatic epithelial neoplasms. Int J Surg Pathol 19(5):588–596 Yeh S, Miyamoto H, Shima H et al, 1998, From estrogen to androgen receptor: a new pathway for sex hormones in prostate. Proc Natl Acad Sci U S A 95(10):5527–5532 Zhang Y, Coogan PF, Palmer JR et al, 2010, A case–control study of reproductive factors, female hormone use, and risk of pancreatic cancer. Cancer Causes Control 21(3):473–478 Jansa R, Prezelj J, Kocijancic A et al, 1996, Androstanediol glucuronide in patients with pancreatic cancer and in those with chronic pancreatitis. Horm Metab Res 28(8):381–383 Hsieh CL, Fei T, Chen Yet al, 2014, RNAs participate in androgen receptor-driven looping that selectively enhances gene activation. Proc Natl Acad Sci U S A 111(20):7319–7324 Dong Y, Matigian N et al, 2008, Tissue-specific promoter utilisation of the kallikrein-related peptidase genes, KLK5 and KLK7, and cellular localisation of the encoded proteins suggest roles in exocrine pancreatic function. Biol Chem 389:99–109 Iakovlev V, Siegel ER, Tsao MS et al, 2012, Expression of kallikrein-related peptidase 7 predicts poor prognosis in patients with unresectable pancreatic ductal adenocarcinoma. Cancer Epidemiol Biomark Prev 21(7):1135–1142 Sandhu V, Wedge DC, 2016, The genomic landscape of pancreatic and Periampullary adenocarcinoma. Cancer Res 76(17): 5092–5102 Parikh H, Wang Z, Pettigrew KA et al, 2011, Fine mapping the KLK3 locus on chromosome 19q13.33 associated with prostate cancer susceptibility and PSA levels. Hum Genet 129(6):675–685 Raju I, Kaushal GP, Haun RS, 2016, Epigenetic regulation of KLK7 gene expression in pancreatic and cervical cancer cells. Biol Chem 397(11):1135–1146 Konduri S, SchwarzMA, Cafasso D et al, 2007, Androgen receptor blockade in experimental combination therapy of pancreatic cancer. J Surg Res 142(2):378–386 Qu Y, Dai B, Ye D et al, 2015, Constitutively active AR-V7 plays an essential role in the development and progression of castrationresistant prostate cancer. Sci Rep 5:7654 Zhang Z, Chen L,Wang H et al, 2015, Inhibition of Plk1 represses androgen signaling pathway in castration-resistant prostate cancer. Cell Cycle 14(13):2142–2148 Mao Y, Xi L, Li Q et al, 2016, Regulation of cell apoptosis and proliferation in pancreatic cancer through PI3K/Akt pathway via polo-like kinase 1. Oncol Rep 36(1):49–56 Song B, Liu XS, Rice SJ et al, 2013, Plk1 phosphorylation of orc2 and hbo1 contributes to gemcitabine resistance in pancreatic cancer. Mol Cancer Ther 12(1):58–68 Shao C, Ahmad N, Hodges K et al, 2015, Inhibition of polo-like kinase 1, Plk1, enhances the antineoplastic activity of metformin in prostate Cancer. J Biol Chem 290(4):2024–2033 Sharifi N, 2012, The 5α-androstanedione pathway to dihydrotestosterone in castration-resistant prostate cancer. J Investig Med 60(2). , DOI 10.2310/JIM.0b013e31823874a4 Fernandez-del Castillo C, Robles-Diaz G et al, 1990, Pancreatic cancer and androgen metabolism: high androstenedione and low testosterone serum levels. Pancreas 5(5):515–518 Chang TC, Lin H, Rogers KA et al, 2013, Expression of aldo-keto reductase family 1 member C3, AKR1C3, in neuroendocrine tumors & adenocarcinomas of pancreas, gastrointestinal tract, and lung. Int J Clin Exp Pathol 6(11):2419–2429 Zhu X, Leav I, Leung YK et al, 2004, Dynamic regulation of estrogen receptor-β expression by DNA methylation during prostate cancer development and metastasis. Am J Pathol 164:2003– 2012 Yeh TS, Jan YY, Chiu CT et al, 2002, Characterisation of oestrogen receptor, progesterone receptor, trefoil factor 1, and epidermal growth factor and its receptor in pancreatic cystic neoplasms and pancreatic ductal adenocarcinoma. Gut 51(5):712– 716 Di Zazzo E, Galasso G et al, 2016, Prostate cancer stem cells: the role of androgen and estrogen receptors. Oncotarget 7(1):193–208 Sinha et al, 2016, Concurrent androgen and estrogen ablation and inhibition of steroid biosynthetic enzyme treatment for castrationresistant prostate Cancer. Anticancer Res 36(8):3847–3854 Kanda T, Jiang X, Yokosuka O, 2014, Androgen receptor signaling in hepatocellular carcinoma and pancreatic cancers. World J Gastroenterol 20(28):9229–9236 Culig Z, Pencik J,Merkel O et al, 2016, Breaking a paradigm: IL- 6/STAT3 signaling suppresses metastatic prostate cancer upon ARF expression. Mol Cell Oncol 3(2):e1090048 Matei DV, Renne G, Pimentel M et al, 2012, Neuroendocrine differentiation in castration-resistant prostate cancer: a systematic diagnostic attempt. Clin Genitourin Cancer 10(3):164–173 Parimi V, Goyal R, Poropatich K et al, 2014, Neuroendocrine differentiation of prostate cancer: a review. Am J Clin Exp Urol 2(4):273–285 Buchler P, Gukovskaya AS, Mouria M et al, 2003, Prevention of metastatic pancreatic cancer growth in vivo by induction of apoptosis with genistein, a naturally occurring is flavonoid. Pancreas 26(3):264–273 Ma HP, Ming LG, Ge BF et al, 2011, Icariin is more potent than genistein in promoting osteoblast differentiation and mineralization in vitro. J Cell Biochem 112(3):916–923 Chun JY, Nadiminty N, Dutt S et al, 2009, Interleukin-6 regulates androgen synthesis in prostate Cancer cells. Clin Cancer Res 15(15):4815–4822 Rojas A, Liu G, Coleman I et al, 2011, IL-6 promotes prostate tumorigenesis and progression through autocrine cross-activation of IGF-IR. Oncogene 30(20):2345–2355 Chandrasekar T, Yang JC, Gao AC et al, 2015, Evans CP. Mechanisms of resistance in castration-resistant prostate cancer, CRPC). Transl Androl Urol 4(3):365–380 Ono H, Basson MD, Ito H, 2016, P300 inhibition enhances gemcitabine-induced apoptosis of pancreatic cancer. Oncotarget 7(32):51301–51310 Paladino D, Yue P, Furuya H et al, 2016, A novel nuclear Src and p300 signaling axis controls migratory and invasive behavior in pancreatic cancer. Oncotarget 7(6):7253–7267 Muniraj T, Chari ST, 2012, Diabetes and pancreatic cancer. Minerva Gastroenterol Dietol 58(4):331–345 Barnard RJ, Aronson WJ, Tymchuk CN et al, 2002, Prostate cancer: another aspect of the insulin-resistance syndrome? Obes Rev 3(4):303–308 Shevach J, Gallagher EJ, Kochukoshy T et al, 2015, Concurrent diabetes mellitus may negatively influence clinical progression and response to androgen deprivation therapy in patients with advanced prostate. Cancer Front Oncol 5:129 Daka B, Rosen T, Jansson PA et al, 2012, Inverse association between serum insulin and sex hormone-binding globulin in a population survey in Sweden. Endocr Connect 2(1):18–22 Monteiro C, Sousa MV, Ribeiro R et al, 2013, Genetic variants in AR and SHBG and resistance to hormonal castration in prostate cancer. Med Oncol 30(1):490 Chuu CP, Kokontis JM, Hiipakka RA et al, 2011, Androgens as therapy for androgen receptor-positive castration-resistant prostate cancer. J Biomed Sci 23:18–63 Mayer MJ, Klotz LH, Venkateswaran V, 2015, Metformin and prostate cancer stem cells: a novel therapeutic target. Prostate Cancer Prostatic Dis 18:303–309 Duan W, Chen K et al, 2017, Desmoplasia suppression by metformin-mediated AMPK activation inhibits pancreatic cancer progression. Cancer Lett 385:225–233 Schiewer MJ, Knudsen KEAM, 2014, Ped up to treat prostate cancer: novel AMPK activators emerge for cancer therapy. EMBO Mol Med 6(4):439–441 Ko AH, Wang F, Holly EA, 2007, Pancreatic cancer and medical history in a population-based case-control study in the San Francisco Bay Area. California. Cancer Causes Control 18(8):809–819 Sarosiek K, Gandhi AV, Saxena S, Kang CY, Chipitsyna GI, Yeo CJ, Arafat HA, 2016, Hypothyroidism in pancreatic cancer: role of exogenous thyroid hormone in tumor invasion-preliminary observations. J Thyroid Res 2016:2454989. , DOI 10.1155/ 2016/2454989 Morote J, Esquena S, Orsola A et al, 2005, Effect of androgen deprivation therapy in the thyroid function test of patients with prostate cancer. Anti-Cancer Drugs 16(8):863–866 Heidegger I, Nagele U, Pircher A et al, 2014, Latent hypothyreosis as a clinical biomarker for therapy response under abiraterone acetate therapy. Anticancer Res 34(1):307–311 Yip YL, Novothy J, EdwardsMet al, 2003, Structural analysis of the erbb-2 receptor using monoclonal antibodies: implications for receptor signaling. Int J Cancer 104:303–309 Craft N, Shostak Y, CareyMet al, 1999, A mechanism for hormoneindependent prostate cancer throughmodulation of androgen receptor signaling by the HER-2/neu tyrosine kinase. Nat Med 5:280–285 Choi HJ, Hong JK, Sung SY et al, 2007, Expression of c-erbB-2 and Cyclooxygenase-2 in pancreatic ductal adenocarcinoma. Korean J Pathol 41:171–175 Hemi R, Paz K,Wertheim N et al, 2002, Transactivation of ErbB2 and ErbB3 by tumor necrosis factor-alpha and anisomycin leads to impaired insulin signaling through serine/threonine phosphorylation of IRS proteins. J Biol Chem 277(11):8961–8969 Komoto M, Nakata B, Amano R et al, 2009, HER2 overexpression correlates with survival after curative resection of pancreatic cancer. Cancer Sci 100(7):1243–1247 Murray NP, Reyes E, Fuentealba C et al, 2015, Possible role of HER-2 in the progression of prostate Cancer from primary tumor to androgen independence. Asian Pac J Cancer Prev 16(15):6615– 6619 Fyssas I, Syrigos KN, Konstandoulakis MM et al, 1997, Sex hormone levels in the serum of patients with pancreatic adenocarcinoma. Horm Metab Res 29(3):115–118 Robles-Diaz G, Duarte-Rojo A, 2001, Pancreas: a sex steroiddependent tissue. Isr Med Assoc J 3(5):364–368 Hoare D, Skinner TA, Black A et al, 2015, Serum folliclestimulating hormone levels predict time to development of castration-resistant prostate cancer. Can Urol Assoc J 9(3–4): 122–127 Pinthus JH, 2015, Follicle-stimulating hormone: a potential surrogate marker for androgen deprivation therapy oncological and systemic effects. Can Urol Assoc J 9(3–4):E226–E227 Lepor H, Neal D, Shore ND, 2012, LHRH agonists for the treatment of prostate Cancer: 2012. Rev Urol 14(1–2):1–12 Bernardo GM, Keri RA, 2012, FOXA1: a transcription factor with parallel functions in development and cancer. Biosci Rep 32(2):113–130 Jin HJ, Zhao JC,Wu L et al, 2014, Cooperativity and equilibrium with FOXA1 define the androgen receptor transcritptional program. Nature Comm 5:3972 Wang J, Nikhil K, Viccaro K, Chang L, JacobsenM, Sandusky G, Shah K, 2017, The Aurora-ATwist1 axis promotes highly aggressive phenotypes in pancreatic carcinoma. J Cell Sci 130(6):1078– 1093 Sen-Yo M, Suehiro Y, Kaino S et al, 2013, TWIST1 hypermethylation is observed in pancreatic cancer. Biomed Rep 1(1):31–33 Shiota M, Yokomizo A, Tada Yet al, 2010, Castration resistance of prostate cancer cells caused by castration-induced oxidative stress through Twist1 and androgen receptor overexpression. Oncogene 29:237–250 Takeuchi A, Shiota M, Beraldi E et al, 2014, Insulin-like growth factor-I induces CLU expression through Twist1 to promote prostate cancer growth. Mol Cell Endocrinol 384(1–2):117–125 Shiota M, Itsumi M, Takeuchi A et al, 2015, Crosstalk between epithelial-mesenchymal transition and castration resistance mediated by Twist1/AR signaling in prostate cancer. Endocr Relat Cancer 22(6):889–900 Mahajan K, Coppola D, Chen YA et al, 2012, Ack1 tyrosine kinase activation correlates with pancreatic cancer progression. Am J Pathol 180(4):1386–1393 Wu X, Cao Y, Het al X, 2016, Bazedoxifene as a novel GP130 inhibitor for pancreatic Cancer therapy. Mol Cancer Ther 15(11): 2609–2619 Yanagisawa N, Ichinoe M, Mikami Tet al, 2012, High expression of L-type amino acid transporter 1, LAT1, predicts poor prognosis in pancreatic ductal adenocarcinomas. J Clin Pathol 65(11):1019– 1023 Wang Q, Bailey CG, Ng C et al, 2011, Androgen receptor and nutrient signaling pathways coordinate the demand for increased amino acidtransport during prostate cancer progression. Cancer Res 71(24):7525–7536 Tursynbay Y, Zhang J, Li Z et al, 2016, Tokay T, Zhumadilov Z, Wu D, Xie Y.Pim-1 kinase as cancer drug target: an update. Biomed Rep 4(2):140–146 Xu J, Xiong G, Cao Z et al, 2016, PIM-1 contributes to the malignancy of pancreatic cancer and displays diagnostic and prognostic value. J Exp Clin Cancer Res 35(1):133 Block KM, Hanke NT, Maine EA et al, 2012, IL-6 stimulates STAT3 and Pim-1 kinase in pancreatic cancer cell lines. Pancreas 41(5):773–781 Linn DE, Yang X, Xie Y et al, 2012, Differential regulation of androgen receptor by PIM-1 kinases via phosphorylation-dependent recruitment of distinct ubiquitin E3 ligases. J Biol Chem 287(27):22959–22968 Holder SL, Abdulkadir SA, 2014, PIM1 kinase as a target in prostate cancer: roles in tumorigenesis, castration resistance, and docetaxel resistance. Curr Cancer Drug Targets 14(2):105–114 Wang J, Quan CY, ChangWLet al, 2015, Correlation between the expression of Pim-1 and androgen-deprivation therapy for prostate cancer. Zhonghua Nan Ke Xue 21(9):775–781 Jin UH, Kim SB, Safe S, 2015, Omeprazole inhibits pancreatic Cancer cell invasion through a nongenomic aryl hydrocarbon receptor pathway. Chem Res Toxicol 28(5):907–918 Le Novere N, HuckaM,Mi H,Moodie S, Schreiber F, Sorokin A, Demir E, 2009, The systems biology graphical notation. Nat Biotechnol 27(8):735–741 Szende B, Srkalovic G, Timar J et al, 1991, Localization of receptors for luteinizing hormone-releasing hormone in pancreatic and mammary cancer cells. Proc Natl Acad Sci U S A 88(10):4153– 4156 Salonia A, Abdollah F, Capitanio U et al, 2012, Serum sex steroids depict a nonlinear u-shaped association with high-risk prostate cancer at radical prostatectomy. Clin Cancer Res 18(13): 3648–3657 Iqbal MJ, Greenway B, Wilkinson ML et al, 1983, Sex-steroid enzymes, aromatase and 5 alpha-reductase in the pancreas: a comparison of normal adult, foetal and malignant tissue. Clin Sci, Lond, 65(1):71–75 Shiota M, Fujimoto N, Yokomizo A et al, 2015, SRD5A gene polymorphism in Japanese men predicts prognosis of metastatic prostate cancer with androgen-deprivation therapy. Eur J Cancer 51(14):1962–1969 Cai C, Chen S, Ng P et al, 2011, Intratumoral de novo steroid synthesis activates androgen receptor in castration-resistant prostate cancer and is upregulated by treatment with CYP17A1 inhibitors. Cancer Res 71(20):6503–6513 Latil A, Bieche I, Vidaud D et al, 2001, Evaluation of androgen, estrogen, ERα and ERβ), and progesterone receptor expression in human prostate cancer by real-time quantitative reverse transcription-polymerase chain reaction assays. Cancer Res 61(5):1919–1926 Greenway B, Iqbal MJ, Johnson PJ et al, 1981, Oestrogen receptor proteins in malignant and fetal pancreas. Br Med J, Clin Res Ed, 283(6294):751–753 Christoforou P, Christopoulos PF, Koutsilieris M, 2014, The role of estrogen receptor β in prostate Cancer. Mol Med 20(1):427– 434 Ellem SJ, Schmitt JF et al, 2004, Local aromatase expression in human prostate is altered in malignancy. J Clin Endocrinol Metab 89:2434–2441 Balk SP, Ko YJ, Bubley GJ, 2003, Biology of prostate-specific antigen. J Clin Oncol 21(2):383–391 Ren H, Zhang H,Wang X et al, 2014, Prostate-specific membrane antigen as a marker of pancreatic cancer cells. Med Oncol 31(3): 857 Chou A, Waddell N, Cowley MJ et al, 2013, Clinical and molecular characterization of HER2 amplified-pancreatic cancer. Genome Med 5(8):78 Vaishampayan U, Thakur A, Rathore R, Kouttab N, Lum LG, 2015, Phase I Study of anti-CD3 x anti-Her2 bispecific antibody in metastatic castrate resistant prostate cancer patients. Prostate Cancer 2015:285193. , DOI 10.1155/2015/285193 Tien JC, Liu Z, Liao L et al, 2013, The steroid receptor Coactivator-3 is required for the development of castrationresistant prostate Cancer. Cancer Res 73(13):3997–4008 Lam HM, Ouyang B, Chen J et al, 2014, Targeting GPR30 with G-1: a new therapeutic target for castration-resistant prostate cancer. Endocr Relat Cancer 21(6):903–914 Kimbara S, Kondo S, 2016, Immune checkpoint and inflammation as therapeutic targets in pancreatic carcinoma. World J Gastroenterol 22(33):7440–7452 Hustinx SR, Leoni LM, Yeo CJ et al, 2005, Concordant loss of MTAP and p16/CDKN2A expression in pancreatic intraepithelial neoplasia: evidence of homozygous deletion in a noninvasive precursor lesion. Mod Pathol 18(7):959–963 Collins CC, Volik SV, Lapuk AV et al, 2012, Next generation sequencing of prostate Cancer from a patient identifies a deficiency of Methylthioadenosine phosphorylase, MTAP), an exploitable tumor target. Mol Cancer Ther 11(3):775–783 Azzopardi S, Pang S, Klimstra DS et al, 2016, p53 and p16Ink4a/ p19Arf Loss Promotes Different Pancreatic Tumor Types from PyMT-Expressing Progenitor Cells. Neoplasia 18(10):610–617 Deng Y, Lu J, 2015, Targeting hexokinase 2 in castration-resistant prostate cancer. Mol Cell Oncol 2(3):e974465 Cozzi PJ, Wang J, Delprado W et al, 2005, MUC1, MUC2, MUC4, MUC5AC and MUC6 expression in the progression of prostate cancer. Clin Exp Metastasis 22(7):565–573 Park JY, Hiroshima Y, Lee JY, MUC1 et al, 2015, Selectively targets human pancreatic Cancer in Orthotopic nude mouse models. PLoS One 10(3):e0122100 Drivdahl R, Haugk KH et al, 2004, Suppression of growth and tumorigenicity in the prostate tumor cell line M12 by overexpression of the transcription factor SOX9. Oncogene 23:4584–4593 Nagathihalli NS, Castellanos JA, VanSaun MN et al, 2016, Pancreatic stellate cell secreted IL-6 stimulates STAT3 dependent invasiveness of pancreatic intraepithelial neoplasia and cancer cells. Oncotarget 7(40):65982–65992 Schweizer L, Rizzo CA, Spires TE et al, 2008, The androgen receptor can signal through Wnt/beta-catenin in prostate cancer cells as an adaptation mechanism to castration levels of androgens. BMC Cell Biol 9:4 The Cancer Genome Atlas Research, 2017, Network Integrated Genomic Characterisation of PAncreatic Ductal Adenocarcinom. Cancer Cell 32(3):185–203 e13 Kasina S, Macoska JA, 2012, The CXCL12/CXCR4 axis promotes ligand-independent activation of the androgen receptor. Mol Cell Endocrinol 351(2):249–263 Deng L, Shang Y, Guo S et al, 2014, Ran GTPase protein promotes metastasis and invasion in pancreatic cancer by deregulating the expression of AR and CXCR4. Cancer Biol Ther 15(8):1087–1093 Yang YA, Yu J, 2015, Current perspectives on FOXA1 regulation of androgen receptor signaling and prostate cancer. Genes Dis 2(2):144–151 Behnsawy HM, Miyake H, Harada K et al, 2013, Expression patterns of epithelial-mesenchymal transition markers in localized prostate cancer: significance in clinicpathological outcomes following radical prostatectomy. BJU 111(1):30–37 Chen S, Chen JZ, Zhang JQ et al, 2016, Hypoxia induces TWISTactivated epithelial-mesenchymal transition and proliferation of pancreatic cancer cells in vitro and in nude mice. Cancer Lett 383(1):73–84 Gao S, Sun Y, Zhang X et al, 2016, IGFBP2 activates the NF-κB pathway to drive epithelial-mesenchymal transition and invasive character in pancreatic ductal adenocarcinoma. Cancer Res 76(22):6543–6554 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1269 EP 1277 DI 10.1007/s12253-018-0467-8 PG 9 ER PT J AU Popiel, A Kobierzycki, Ch Dziegiel, P AF Popiel, Aneta Kobierzycki, Christopher Dziegiel, Piotr TI The Role of Testin in Human Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Testin; Cancerogenesis; Breast cancer; Ovarian cancer; Gastrointestinal cancers ID Testin; Cancerogenesis; Breast cancer; Ovarian cancer; Gastrointestinal cancers AB Testin is a protein expressed in almost all normal human tissues. It locates in the cytoplasm along stress fibers being recruited to focal adhesions. Together with zyxin and vasodilator stimulated protein it forms complexes with various cytoskeleton proteins such as actin, talin and paxilin. They jointly play significant role in cell motility and adhesion. In addition, their involvement in the cell cycle has been demonstrated. Expression of testin protein level correlates positively with percentage of cells in G1 phase, while overexpression can induce apoptosis and decreased colony forming ability. Decreased testin expression associate with loss by cells epithelial morphology and gain migratory and invasive properties of mesenchymal cells. Latest reports indicate that TES is a tumor suppressor gene which can contribute to cancerogenesis but the mechanism of loss TES gene expression is still unknown. Some authors point out hypermethylation of the CpG island as a main factor, however loss of heterozygosity may also play an important role [4, 5]. The altered expression of testin was found in malignant neoplasm, i.a. ovarian, lung, head and neck squamous cell cancer, breast, endometrial, colorectal, prostate and gastric cancers [1–9]. Testin participate in the processes of tumor growth, angiogenesis, and metastasis [10]. Many researchers stated involvement of testin in tumor progression, what suggest its potential usage in immunotherapy [7, 11]. Understanding the molecular functions of testin may be crucial in development personalized treatment. In the present manuscript up-to-date review of literature can be found. C1 [Popiel, Aneta] Wroclaw Medical University, Department of Human Morphology and EmbryologyWroclaw, Poland. [Kobierzycki, Christopher] Wroclaw Medical University, Department of Human Morphology and EmbryologyWroclaw, Poland. [Dziegiel, Piotr] Wroclaw Medical University, Department of Human Morphology and EmbryologyWroclaw, Poland. RP Popiel, A (reprint author), Wroclaw Medical University, Department of Human Morphology and Embryology, Wroclaw, Poland. EM aneta.popiel@live.com CR Qiu H, Zhu J, Yuan C, Yan S, Yang Q, Kong B, 2010, Frequent hypermethylation and loss of heterozygosity of the testis derived transcript gene in ovarian cancer. Cancer Sci 101:1255–1260. , DOI 10.1111/j.1349-7006.2010.01497.x Dong R, Pu H, Wang Y, Yu J, Lian K, Mao C, 2015, TESTIN was commonly hypermethylated and involved in the epithelialmesenchymal transition of endometrial cancer. APMIS 123:394– 400. , DOI 10.1111/apm.12361 Ma H,Weng D, Chen Y, HuangW, Pan K,Wang H, Sun J,Wang Q, Zhou Z, Wang H, Xia J, 2010, Extensive analysis of D7S486 in primary gastric cancer supports TESTIN as a candidate tumor suppressor gene. Mol Cancer 9:1–10 Chene L, Giroud C, Desgrandchamps F, Boccon-Gibod L, Cussenot O, Berthon P, Latil A, 2004, Extensive analysis of the 7q31 region in human prostate tumors supports TES as the best candidate tumor suppressor gene. Int J Cancer 111:798–804. , DOI 10.1002/ijc.20337 Li H, Huang K, Gao L, Wang L, Niu Y, Liu H, 2016, TES inhibits colorectal cancer progression through activation of p38. Oncotagret 7 Sarti M, Sevignani C, Calin GA, Aqeilan R, Shimizu M, Pentimalli F, Picchio MC, Godwin A, Rosenberg A, Drusco A, Negrini M, Croce CM, 2005, Adenoviral transduction of TESTIN gene into breast and uterine Cancer cell lines promotes apoptosis and tumor reduction in vivo. Clin Cancer Res 11:806–813 MingW, QianW, PengW-J, Hu J-F,Wang Z-Y, Liu H, Huang L-N, 2017, Testin is a tumor suppressor in non-small cell lung cancer. Oncol Rep 37:1027–1035. , DOI 10.3892/or.2016.5316 Gunduz E, GunduzM, Beder L, Nagatsuka H, Fukushima K, Sutcu R, Delibas N, Yamanaka N, Shimizu K, Nagai N, CM C, AG K,M E,DMP, JM C, MI E, JC L, V S, S T, XLW, JC Z, JS Z, JC Z, JC Z, T O, M G, C T, M S, A D, L C, W M, F B, B T, 2009, Downregulation of TESTIN and its association with Cancer history and a tendency toward poor survival in head and neck squamous cell carcinoma. Arch Otolaryngol Neck Surg 135:254. https://doi. org/10.1001/archoto.2008.560, 260 Li S, Xue M HL, 2016, Relationship between Testin expression and clinicopathological characteristics in nasopharyngeal carcinoma patients. 310–3 Steponaitis G, Kazlauskas A, Skiriute D, Valiulyte I, Skauminas K, Tamasauskas A, Vaitkiene P, 2016, Testin, TES, as a candidate tumour suppressor and prognostic marker in human astrocytoma. Oncol Lett 12:3305–3311. , DOI 10.3892/ol.2016.5077 Prieto-Garcia E, Diaz-Garcia CV, Garcia-Ruiz I, Agullo-Ortuno MT, 2017, Epithelial-to-mesenchymal transition in tumor progression. Med Oncol 34:122. , DOI 10.1007/s12032-017- 0980-8 Sala S, Van Troys M, Medves S, Catillon M, Timmerman E, Staes A, Schaffner-Reckinger E, Gevaert K, Ampe C, 2017, Expanding the interactome of TES by exploiting TES modules with different subcellular localizations. J Proteome Res acs.jproteome.7b00034. , DOI 10.1021/acs.jproteome.7b00034 Zhong Y, Zhu J, Wang Y, Zhou J, Ren K, Ding X, Zhang J, 2009, LIM domain protein TES changes its conformational states in different cellular compartments. Mol Cell Biochem 320:85–92. , DOI 10.1007/s11010-008-9901-7 Khurana T, Khurana B, Noegel a a, 2002, LIMproteins: association with the actin cytoskeleton. Protoplasma 219:1–12. , DOI 10.1007/s007090200000 Sala S, Catillon M, Hadzic E, Schaffner-reckinger E, Troys MVan, Ampe C, 2017, The PET and LIM1–2 domains of testin contribute to intramolecular and homodimeric interactions. 1:1–21 Weeks RJ, Ludgate JL, LeMee G, Morison IM, 2016, TESTIN induces rapid death and suppresses proliferation in childhood B acute lymphoblastic Leukaemia cells. PLoS One 11:e0151341. , DOI 10.1371/journal.pone.0151341 Oldenburg J, van der Krogt G, Twiss F, Bongaarts A, Habani Y, Slotman JA, Houtsmuller A, Huveneers S, de Rooij J, 2015, VASP, zyxin and TES are tension-dependent members of focal Adherens junctions independent of the α-catenin-vinculin module. Sci Rep 5: 17225. , DOI 10.1038/srep17225 Coutts AS, 2003, TES is a novel focal adhesion protein with a role in cell spreading. J Cell Sci 116:897–906. , DOI 10.1242/ jcs.00278 Tatarelli C, Linnenbach A, Mimori K, Croce CM, 2000, Characterization of the human TESTIN gene localized in the FRA7G region at 7q31. 2 Genomics 12:1–12. , DOI 10. 1006/geno.2000.6272 Gu Z, Ding G, Liang K, Zhang H, Guo G, Zhang L, Cui J, 2014, TESTIN suppresses tumor growth and invasion via manipulating cell cycle progression in endometrial carcinoma. Med SciMonit 20: 980–987. , DOI 10.12659/MSM.890544 Boeda B, Briggs DC, Higgins T, Garvalov BK, Fadden AJ, McDonald NQ, Way M, 2007, Tes, a specific Mena interacting partner, breaks the rules for EVH1 binding. Mol Cell 28:1071– 1082. , DOI 10.1016/j.molcel.2007.10.033 Griffith E, Coutts AS, Black DM, 2004, Characterisation of chicken TES and its role in cell spreading and motility. Cell Motil Cytoskeleton 57:133–142. , DOI 10.1002/cm.10162 Drusco A, Zanesi N, Roldo C, Trapasso F, Farber JL, Fong LY, Croce CM, 2005, Knockout mice reveal a tumor suppressor function for Testin. PNAS 102:10947–10951 Zhu J, Li X, Kong X, Moran MS, Su P, Haffty BG, Yang Q, 2012, Testin is a tumor suppressor and prognostic marker in breast cancer. Cancer Sci 103:2092–2101. , DOI 10.1111/cas.12020 Sarti M, Pinton S, Limoni C, Carbone GM, Pagani O, Cavalli F, Catapano CV, 2013, Differential expression of testin and survivin in breast cancer subtypes. Oncol Rep 30:824–832. , DOI 10.3892/or.2013.2502 Tobias ES, Hurlstone AFL, Mackenzie E, Mcfarlane R, Black DM, 2001, The TES gene at 7q31 . 1 is methylated in tumours and encodes a novel growth-suppressing LIM domain protein. Oncogene 20:2844–2853 Mueller W, Nutt CL, Ehrich M, Riemenschneider MJ, von Deimling A, van den Boom D, Louis DN, 2007, Downregulation of RUNX3 and TES by hypermethylation in glioblastoma. Oncogene 26:583–593. , DOI 10.1038/sj.onc.1209805 Han SY, Druck T, Huebner K, 2003, Candidate tumor suppressor genes at FRA7G are coamplified with MET and do not suppress malignancy in a gastric cancer. Genomics 81:105–107. https://doi. org/10.1016/S0888-7543(02)00029-0 Frey MK, Pothuri B, 2017, Homologous recombination deficiency, HRD, testing in ovarian cancer clinical practice: a review of the literature. Gynecol Oncol Res Pract 4:4. , DOI 10.1186/ s40661-017-0039-8 Bieche I, Khodja a DK, Lidereau R, 1997, Genetic alteration mapping on chromosome 7 in primary breast cancer. Clin Cancer Res 3: 1009–1016 Zenklusen JC, Thompson JC, Conti CJ, Klein-Szanto AJP, 1995, Frequent loss of heterozygosity in human primary squamous cell and Colon carcinomas at 7q31.1: evidence for a broad range tumor suppressor gene. Cancer Res 55:1347–1350 Jia X, Shanmugam C, Paluri RK, Jhala NC, Behring P, Katkoori VR, Sugandha SP, Bae S, 2017, Prognostic value of loss of heterozygosity and sub-cellular localization of SMAD4 varies with tumor stage in colorectal cancer. 8:20198–20212. , DOI 10. 18632/oncotarget.15560 Matsuura K, Shiga K, Yokoyama J, Saijo S, Miyagi T, Takasaka T, 1998, Loss of heterozygosity of chromosome 9p21 and 7q31 is correlated with high incidence of recurrent tumor in head and neck squamous cell carcinoma. Anticancer Res 18:453–458 Koike M, Tasaka T, Spira S, Tsuruoka N, Phillip Koeffler H, 1999, Allelotyping of acute myelogenous leukemia: loss of heterozygosity at 7q31.1, D7S486, and q33-34, D7S498, D7S505). Leuk Res 23:307–310. , DOI 10.1016/S0145-2126(98)00159-3 Weeks RJ, Kees UR, Song S, Morison IM, 2010, Silencing of TESTIN by dense biallelic promoter methylation is the most common molecular event in childhood acute lymphoblastic leukaemia. Mol Cancer 9:163. , DOI 10.1186/1476-4598-9-163 Shridhar V, Sun QC,Miller OJ, Kalemkerian GP, Petros J, Smith DI, 1997, Loss of heterozygosity on the long arm of human chromosome 7 in sporadic renal cell carcinomas. Oncogene 15: 2727–2733. , DOI 10.1038/sj.onc.1201448 Achille A, Biasi MO, Zamboni G, Bogina G, Magalini AR, Pederzoli P, Perucho M, Scarpa A, 1996, Chromosome 7q allelic losses in pancreatic carcinoma. Cancer Res 56:3808–3813 Hino O, Kobayashi T, 2017, Mourning Dr. Alfred G. Knudson: the two-hit hypothesis, tumor suppressor genes, and the tuberous sclerosis complex. Cancer Sci 108:5–11. , DOI 10.1111/cas. 13116 Nasir A, Holzer TR, Man M, Benjamin LE, Melemed AS, Schade AE, 2015, Differential expression of VEGFR2 protein in HER2 positive primary human breast cancer: potential relevance to newer anti-angiogenic therapies. Cancer Res 75:1–12. , DOI 10. 1186/s12935-017-0427-5 Iyer RP, Patterson NL, Fields GB, Lindsey ML, 2012, The history of matrix metalloproteinases : milestones , myths , and misperceptions. Trabslational Physiol. , DOI 10.1152/ajpheart. 00577.2012 Fang J, Zhou H, Zeng C,Yang J, LiuY, HuangX, Zhang J,Guan X, Zhuang S, 2011, MicroRNA-29b suppresses tumor angiogenesis, invasion, and metastasis by regulating matrix metalloproteinase 2 expression. Hepatology 54:1729–1740. , DOI 10.1002/ hep.24577 Holm K, Hegardt C, Staaf J, Vallon-Christersson J, Jonsson G, Olsson H, Borg A, RingnerM(2010)Molecular subtypes of breast cancer are associated with characteristic DNAmethylation patterns. Breast Cancer Res 12:R36. , DOI 10.1186/bcr2590 Kalluri R, R a W, 2009, Review series the basics of epithelialmesenchymal transition. J Clin Invest 119:1420–1428. https://doi. org/10.1172/JCI39104.1420 Grossi V, Peserico A, Tezil T, Simone C, 2014, p38α MAPK pathway: a key factor in colorectal cancer therapy and chemoresistance. World J Gastroenterol 20:9744–9758. , DOI 10.3748/wjg. v20.i29.9744 Zhang Y, Guo Z, Du T, Chen J, Wang W, Xu K, Lin T, Huang H, 2013, Prostate specific membrane antigen, PSMA): a novel modulator of p38 for proliferation, migration, and survival in prostate cancer cells. Prostate 73:835–841. , DOI 10.1002/pros. 22627 Hui L, Bakiri L, Stepniak E, Wagner EF, 2007, p38alpha: a suppressor of cell proliferation and tumorigenesis. Cell Cycle 6:2429– 2433. , DOI 10.4161/cc.6.20.4774 HuangW,Weng DS, Pan ZZ, Pan K, Ding PR, Zhou JW,Wang H, Zhang HK, Li JJ, Xia JC, 2008, Expression and clinical significance of TESTIN in primary gastric cancer. Chinese J Cancer 27: 267–270 Wang M,Wang Q, PengW, Hu J, 2017, Testin is a tumor suppressor in non-small cell lung cancer. 1027–1035. , DOI 10. 3892/or.2016.5316 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1279 EP 1284 DI 10.1007/s12253-018-0488-3 PG 6 ER PT J AU Koohini, Z Koohini, Z Teimourian, Sh AF Koohini, Zahra Koohini, Zohreh Teimourian, Shahram TI Slit/Robo Signaling Pathway in Cancer; a New Stand Point for Cancer Treatment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Slite; Robo; Tumor angiogenesis; Tumor markers ID Slite; Robo; Tumor angiogenesis; Tumor markers AB Angiogenesis and metastasis are two critical steps for cancer cells survival and migration. The microenvironment of tumor sphere induces new blood vessels formation for enhancing tumor mass. Preexisting capillaries and postcapillary venules in tumors bring about new blood vessels. ROBO1-ROBO4 are transmembrane receptors family which act as guidance molecules of the nervous system. The SLITs family is secreted glycoproteins that bind to these receptors. SLIT-ROBO signaling pathway plays an important role in neurogenesis and immune response. Linkage between ROBOs and their ligands (SLITs) induce chemorepllent signal for regulation of axon guidance and leukocyte cell migration, recent finding shows that it is also involved in endothelial cell migration and angiogenesis in various type of cancers. In this article we review recent finding of SLIT-ROBO pathway in angiogenesis and metastasis. C1 [Koohini, Zahra] Iran University of Medical Sciences, School of Medicine, Department of Medical GeneticsTehran, Iran. [Koohini, Zohreh] Mazandaran University of Medical Sciences, School of Medicine, Department of ImmunologySari, Iran. [Teimourian, Shahram] Iran University of Medical Sciences, School of Medicine, Department of Medical GeneticsTehran, Iran. RP Teimourian, Sh (reprint author), Iran University of Medical Sciences, School of Medicine, Department of Medical Genetics, Tehran, Iran. EM teimourian.sh@iums.ac.ir CR Ballard MS, Hinck L, 2012, A roundabout way to cancer. Adv Cancer Res 114:187–235 Greenberg JM, Thompson FY, Brooks SK, Shannon JM, Akeson AL, 2004, Slit and robo expression in the developing mouse lung. Dev Dyn 230(2):350–360 Hinck L, 2004, The versatile roles of "axon guidance" cues in tissue morphogenesis. Dev Cell 7(6):783–793 Medioni C, Bertrand N, Mesbah K, Hudry B, Dupays L, Wolstein O, Washkowitz AJ, Papaioannou VE, Mohun TJ, Harvey RP, Zaffran S, 2010, Expression of slit and Robo genes in the developing mouse heart. Dev Dyn 239(12):3303–3311 Brose K, Bland KS,Wang KH, Arnott D, HenzelW, Goodman CS et al, 1999, Slit proteins bind Robo receptors and have an evolutionarily conserved role in repulsive axon guidance. Cell 6(6):795– 806 Legg JA, Herbert JM, Clissold P, Bicknell R, 2008, Slits and roundabouts in cancer, tumour angiogenesis and endothelial cell migration. Angiogenesis 11(1):13–21 London NR, Li DY, 2011, Robo4-dependent slit signaling stabilizes the vasculature during pathologic angiogenesis and cytokine storm. Curr Opin Hematol 18(3):186–190 Wu JY, Feng L, Park HT, Havlioglu N,Wen L, Tang H, Bacon KB, Jiang ZH, Zhang XC, Rao Y, 2001, The neuronal repellent slit inhibits leukocyte chemotaxis induced by chemotactic factors. Nature 410(6831):948–952 Hanahan D, Folkman J, 1996, Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86(3):353–364 Jain RK, 2002, Tumor angiogenesis and accessibility: role of vascular endothelial growth factor. Semin Oncol 29(6 Suppl 16):3–9 Nusslein-Volhard C, Wieschaus E, Kluding H, 1984, Mutations affecting the pattern of the larval cuticle inDrosophila melanogaster : I. zygotic loci on the second chromosome. Wilehm Roux Arch Dev Biol 193(5):267–282 Chedotal A, 2007, Slits and their receptors. Adv Exp Med Biol 621:65–80 Blockus H, Chedotal A, 2016, Slit-Robo signaling. Development 143(17):3037–3044 Rothberg JM, Artavanis-Tsakonas S, 1992, Modularity of the slit protein. Characterization of a conserved carboxy-terminal sequence in secreted proteins and a motif implicated in extracellular protein interactions. J Mol Biol 227(2):367–370 Zhang B, Dietrich UM, Geng JG, Bicknell R, Esko JD, Wang L, 2009, Repulsive axon guidance molecule Slit3 is a novel angiogenic factor. Blood 114(19):4300–4309 Wu MF, Liao CY,Wang LY, Chang JT, 2017, The role of slit-Robo signaling in the regulation of tissue barriers. Tissue Barriers 5(2): e1331155 Kidd T, Bland KS, Goodman CS, 1999, Slit is the midline repellent for the robo receptor in Drosophila. Cell 96(6):785–794 Dickson BJ, Gilestro GF, 2006, Regulation of commissural axon pathfinding by slit and its Robo receptors. Annu Rev Cell Dev Biol 22:651–675 Kidd T, Brose K, Mitchell KJ, Fetter RD, Tessier-Lavigne M, Goodman CS, Tear G, 1998, Roundabout controls axon crossing of the CNS midline and defines a novel subfamily of evolutionarily conserved guidance receptors. Cell 92(2):205–215 Yadav SS, Narayan G, 2014, Role of ROBO4 signalling in developmental and pathological angiogenesis. Biomed Res Int 2014: 683025 Okada Y, Yano K, Jin E, Funahashi N, KitayamaM, Doi T, Spokes K, Beeler DL, Shih SC, Okada H, Danilov TA, Maynard E, Minami T, Oettgen P, Aird WC, 2007, A three-kilobase fragment of the human Robo4 promoter directs cell type-specific expression in endothelium. Circ Res 100(12):1712–1722 Zelina P, Blockus H, Zagar Y, Peres A, Friocourt F, Wu Z et al, 2014, Signaling switch of the axon guidance receptor Robo3 during vertebrate evolution. Neuron 84(6):1258–1272 Zhang F,Moniz HA,Walcott B, Moremen KW, Linhardt RJ,Wang L, 2013, Characterization of the interaction between Robo1 and heparin and other glycosaminoglycans. Biochimie 95(12):2345– 2353 Seki M, Watanabe A, Enomoto S, Kawamura T, Ito H, Kodama T, Hamakubo T, Aburatani H, 2010, Human ROBO1 is cleaved by metalloproteinases and gamma-secretase and migrates to the nucleus in cancer cells. FEBS Lett 584(13):2909–2915 Coleman HA, Labrador JP, Chance RK, Bashaw GJ, 2010, The Adam family metalloprotease Kuzbanian regulates the cleavage of the roundabout receptor to control axon repulsion at the midline. Development 137(14):2417–2426 Nguyen Ba-Charvet KT, Brose K, Ma L, Wang KH, Marillat V, Sotelo C et al, 2001, Diversity and specificity of actions of Slit2 proteolytic fragments in axon guidance. J Neurosci 21(12):4281– 4289 Wang KH, Brose K, Arnott D, Kidd T, Goodman CS, Henzel W, Tessier-Lavigne M, 1999, Biochemical purification of a mammalian slit protein as a positive regulator of sensory axon elongation and branching. Cell 96(6):771–784 Delloye-Bourgeois C, Jacquier A, Charoy C, Reynaud F, Nawabi H, Thoinet K, Kindbeiter K, Yoshida Y, Zagar Y, Kong Y, Jones YE, Falk J, Chedotal A, Castellani V, 2015, PlexinA1 is a new slit receptor and mediates axon guidance function of slit C-terminal fragments. Nat Neurosci 18(1):36–45 Wright KM, Lyon KA, Leung H, Leahy DJ, Ma L, Ginty DD, 2012, Dystroglycan organizes axon guidance cue localization and axonal pathfinding. Neuron 76(5):931–944 Svensson KJ, Long JZ, JedrychowskiMP, Cohen P, Lo JC, Serag S, Kir S, Shinoda K, Tartaglia JA, Rao RR, Chedotal A, Kajimura S, Gygi SP, Spiegelman BM, 2016, A secreted Slit2 fragment regulates adipose tissue thermogenesis and metabolic function. Cell Metab 23(3):454–466 Dascenco D, Erfurth ML, Izadifar A, Song M, Sachse S, Bortnick R, Urwyler O, Petrovic M, Ayaz D, He H, Kise Y, Thomas F, Kidd T, Schmucker D, 2015, Slit and receptor tyrosine phosphatase 69D confer spatial specificity to axon branching via Dscam1. Cell 162(5):1140–1154 Holmes GP, Negus K, Burridge L, Raman S, Algar E, Yamada T, Little MH, 1998, Distinct but overlapping expression patterns of two vertebrate slit homologs implies functional roles in CNS development and organogenesis. Mech Dev 79(1–2):57–72 Latil A, Chene L, Cochant-Priollet B, Mangin P, Fournier G, Berthon P et al, 2003, Quantification of expression of netrins, slits and their receptors in human prostate tumors. Int J Cancer 103(3): 306–315 Wang B, Xiao Y, Ding BB, Zhang N, Yuan X, Gui L, Qian KX, Duan S, Chen Z, Rao Y, Geng JG, 2003, Induction of tumor angiogenesis by slit-Robo signaling and inhibition of cancer growth by blocking Robo activity. Cancer Cell 4(1):19–29 Yuan W, Zhou L, Chen JH, Wu JY, Rao Y, Ornitz DM, 1999, The mouse SLIT family: secreted ligands for ROBO expressed in patterns that suggest a role in morphogenesis and axon guidance. Dev Biol 212(2):290–306 Dallol A, Morton D, Maher ER, Latif F, 2003, SLIT2 axon guidance molecule is frequently inactivated in colorectal cancer and suppresses growth of colorectal carcinoma cells. Cancer Res 63(5):1054–1058 Park KW, Morrison CM, Sorensen LK, Jones CA, Rao Y, Chien CB,Wu JY, Urness LD, Li DY, 2003, Robo4 is a vascular-specific receptor that inhibits endothelial migration. Dev Biol 261(1):251– 267 Pircher A, FieglM, Untergasser G, Heidegger I, Medinger M, Kern J et al, 2013, Favorable prognosis of operable non-small cell lung cancer [NSCLC] patients harboring an increased expression of tumor endothelial markers [TEMs]. Lung cancer, Amsterdam, Netherlands, 81(2):252–258 Jones CA, London NR, Chen H, Park KW, Sauvaget D, Stockton RA, Wythe JD, Suh W, Larrieu-Lahargue F, Mukouyama YS, Lindblom P, Seth P, Frias A, Nishiya N, Ginsberg MH, Gerhardt H, Zhang K, Li DY, 2008, Robo4 stabilizes the vascular network by inhibiting pathologic angiogenesis and endothelial hyperpermeability. Nat Med 14(4):448–453 Huminiecki L, Gorn M, Suchting S, Poulsom R, Bicknell R, 2002, Magic roundabout is a new member of the roundabout receptor family that is endothelial specific and expressed at sites of active angiogenesis. Genomics 79(4):547–552 Howitt JA, Clout NJ, Hohenester E, 2004, Binding site for Robo receptors revealed by dissection of the leucine-rich repeat region of slit. EMBO J 23(22):4406–4412 Kaur S, Castellone MD, Bedell VM, Konar M, Gutkind JS, Ramchandran R, 2006, Robo4 signaling in endothelial cells implies attraction guidance mechanisms. J Biol Chem 281(16): 11347–11356 Garrett TA, Van Buul JD, Burridge K, 2007, VEGF-induced Rac1 activation in endothelial cells is regulated by the guanine nucleotide exchange factor Vav2. Exp Cell Res 313(15):3285–3297 Gavard J, Gutkind JS, 2006, VEGF controls endothelial-cell permeability by promoting the beta-arrestin-dependent endocytosis of VE-cadherin. Nat Cell Biol 8(11):1223–1234 Jones CA, NishiyaN, London NR, ZhuW, Sorensen LK, Chan AC, LimCJ, ChenH, Zhang Q, Schultz PG, HayallahAM, Thomas KR, Famulok M, Zhang K, Ginsberg MH, Li DY, 2009, Slit2-Robo4 signalling promotes vascular stability by blocking Arf6 activity. Nat Cell Biol 11(11):1325–1331 Grossmann AH, Yoo JH, Clancy J, Sorensen LK, Sedgwick A, Tong Z et al, 2013, The small GTPase ARF6 stimulates betacatenin transcriptional activity during WNT5A-mediated melanoma invasion and metastasis. Sci Signal 6(265):ra14 ZhuW, London NR, Gibson CC, Davis CT, Tong Z, Sorensen LK, Shi DS, Guo J, Smith MCP, Grossmann AH, Thomas KR, Li DY, 2012, Interleukin receptor activates a MYD88-ARNO-ARF6 cascade to disrupt vascular stability. Nature 492(7428):252–255 Davis CT, Zhu W, Gibson CC, Bowman-Kirigin JA, Sorensen L, Ling J et al, 2014, ARF6 inhibition stabilizes the vasculature and enhances survival during endotoxic shock. J Immunol 192(12): 6045–6052 Bolton SJ, Anthony DC, Perry VH, 1998, Loss of the tight junction proteins occludin and zonula occludens-1 from cerebral vascular endothelium during neutrophil-induced blood-brain barrier breakdown in vivo. Neuroscience 86(4):1245–1257 Salcedo R, Ponce ML, Young HA, Wasserman K, Ward JM, Kleinman HK, Oppenheim JJ, Murphy WJ, 2000, Human endothelial cells express CCR2 and respond to MCP-1: direct role of MCP-1 in angiogenesis and tumor progression. Blood 96(1):34–40 Stamatovic SM, Keep RF, Kunkel SL, Andjelkovic AV, 2003, Potential role of MCP-1 in endothelial cell tight junction 'opening': signaling via rho and rho kinase. J Cell Sci 116(Pt 22):4615–4628 Voronov E, Shouval DS, Krelin Y, Cagnano E, Benharroch D, Iwakura Y, Dinarello CA, Apte RN, 2003, IL-1 is required for tumor invasiveness and angiogenesis. Proc Natl Acad Sci U S A 100(5):2645–2650 Martin TA, JiangWG, 2009, Loss of tight junction barrier function and its role in cancer metastasis. Biochim Biophys Acta 1788(4): 872–891 ZhouW, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, Yu Y, Chow A,O’Connor STF, ChinAR,YenY,WangY, Marcusson EG, Chu P, Wu J, Wu X, Li AX, Li Z, Gao H, Ren X, Boldin MP, Lin PC, Wang SE, 2014, Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 25(4):501– 515 Zhao H, Ahirwar DK, Oghumu S, Wilkie T, Powell CA, Nasser MW, Satoskar AR, Li DY, Ganju RK, 2016, Endothelial Robo4 suppresses breast cancer growth and metastasis through regulation of tumor angiogenesis. Mol Oncol 10(2):272–281 Cai H, Xue Y, Li Z, HuY,Wang Z, LiuWet al, 2015, Roundabout4 suppresses glioma-induced endothelial cell proliferation, migration and tube formation in vitro by inhibiting VEGR2-mediated PI3K/ AKT and FAK signaling pathways. Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology 35(5):1689–1705 Dickinson RE, Fegan KS, Ren X, Hillier SG, Duncan WC, 2011, Glucocorticoid regulation of SLIT/ROBO tumour suppressor genes in the ovarian surface epithelium and ovarian cancer cells. PloS one 6(11):e27792 Avci ME, Konu O, Yagci T, 2008, Quantification of SLIT-ROBO transcripts in hepatocellular carcinoma reveals two groups of genes with coordinate expression. BMC Cancer 8:392 Kaur S, Samant GV, Pramanik K, Loscombe PW, Pendrak ML, Roberts DD, Ramchandran R, 2008, Silencing of directional migration in roundabout4 knockdown endothelial cells. BMC Cell Biol 9:61 Koch AW, Mathivet T, Larrivee B, Tong RK, Kowalski J, Pibouin- Fragner L et al, 2011, Robo4 maintains vessel integrity and inhibits angiogenesis by interacting with UNC5B. Dev Cell 20(1):33–46 Klagsbrun M, Eichmann A, 2005, A role for axon guidance receptors and ligands in blood vessel development and tumor angiogenesis. Cytokine Growth Factor Rev 16(4–5):535–548 Yiin JJ, Hu B, Jarzynka MJ, Feng H, Liu KW, Wu JY, Ma HI, Cheng SY, 2009, Slit2 inhibits glioma cell invasion in the brain by suppression of Cdc42 activity. Neuro-Oncology 11(6):779–789 Wong K, Ren XR, Huang YZ, Xie Y, Liu G, Saito H, Tang H,Wen L, Brady-Kalnay SM, Mei L, Wu JY, Xiong WC, Rao Y, 2001, Signal transduction in neuronal migration: roles of GTPase activating proteins and the small GTPase Cdc42 in the slit-Robo pathway. Cell 107(2):209–221 Hu H, Li M, Labrador JP, McEwen J, Lai EC, Goodman CS, Bashaw GJ, 2005, Cross GTPase-activating protein [CrossGAP]/ Vilse links the roundabout receptor to Rac to regulate midline repulsion. Proc Natl Acad Sci U S A 102(12):4613–4618 Lundstrom A, Gallio M, Englund C, Steneberg P, Hemphala J, Aspenstrom P et al, 2004, Vilse, a conserved Rac/Cdc42 GAP mediating Robo repulsion in tracheal cells and axons. Genes Dev 18(17):2161–2171 Tole S,Mukovozov IM, Huang YW, MagalhaesMA, YanM, Crow MR et al, 2009, The axonal repellent, Slit2, inhibits directional migration of circulating neutrophils. J Leukoc Biol 86(6):1403– 1415 Martinez-Quiles N, Rohatgi R, Anton IM, Medina M, Saville SP, Miki H et al, 2001, WIP regulates N-WASP-mediated actin polymerization and filopodium formation. Nat Cell Biol 3(5):484–491 Krugmann S, Jordens I, Gevaert K, Driessens M, Vandekerckhove J, Hall A, 2001, Cdc42 induces filopodia by promoting the formation of an IRSp53:Mena complex. Curr Biol 11(21):1645–1655 Han SP, Yap AS, 2012, The cytoskeleton and classical cadherin adhesions. Subcell Biochem 60:111–135 Ratheesh A, Yap AS, 2012, A bigger picture: classical cadherins and the dynamic actin cytoskeleton. Nat Rev Mol Cell Biol 13(10): 673–679 Yilmaz M, Christofori G, 2010, Mechanisms of motility in metastasizing cells. Mol Cancer Res 8(5):629–642 Zhong C, Mei Z, Yong X, 2017, Cadherin switching induced by P120-catenin can promote the migration and invasion of oral squamous cell cancer cells. Hua Xi Kou Qiang Yi Xue Za Zhi 35(2): 183–186 Bashaw GJ, Kidd T, Murray D, Pawson T, Goodman CS, 2000, Repulsive axon guidance: Abelson and enabled play opposing roles downstream of the roundabout receptor. Cell 101(7):703–715 Wills Z, Emerson M, Rusch J, Bikoff J, Baum B, Perrimon N, van Vactor D, 2002, A Drosophila homolog of cyclase-associated proteins collaborates with the Abl tyrosine kinase to control midline axon pathfinding. Neuron 36(4):611–622 O'Donnell MP, Bashaw GJ, 2013, Distinct functional domains of the Abelson tyrosine kinase control axon guidance responses to netrin and slit to regulate the assembly of neural circuits. Development [Cambridge, England] 140(13):2724–2733 Rhee J, Buchan T, Zukerberg L, Lilien J, Balsamo J, 2007, Cables links Robo-bound Abl kinase to N-cadherin-bound beta-catenin to mediate slit-induced modulation of adhesion and transcription. Nat Cell Biol 9(8):883–892 ZhouWJ, Geng ZH, Chi S, ZhangW, Niu XF, Lan SJ,Ma L, Yang X, Wang LJ, Ding YQ, Geng JG, 2011, Slit-Robo signaling induces malignant transformation through Hakai-mediated Ecadherin degradation during colorectal epithelial cell carcinogenesis. Cell Res 21(4):609–626 Prasad A, Paruchuri V, Preet A, Latif F, Ganju RK, 2008, Slit-2 induces a tumor-suppressive effect by regulating beta-catenin in breast cancer cells. J Biol Chem 283(39):26624–26633 TsengRC, Lee SH, Hsu HS, Chen BH, TsaiWC, Tzao C,Wang YC, 2010, SLIT2 attenuation during lung cancer progression deregulates beta-catenin and E-cadherin and associates with poor prognosis. Cancer Res 70(2):543–551 FengY, Feng L,Yu D, Zou J, Huang Z, 2016, srGAP1mediates the migration inhibition effect of Slit2-Robo1 in colorectal cancer. J Exp Clin Cancer Res 35(1):191 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1285 EP 1293 DI 10.1007/s12253-018-00568-y PG 9 ER PT J AU Ying, J Qiu, X Yu, L Zhang, M AF Ying, Jie Qiu, Xiaoyan Yu, Lu Zhang, Miaomiao TI SOCS1 and its Potential Clinical Role in Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE SOCS1; Cell signaling; Ubiquitination; Tumors ID SOCS1; Cell signaling; Ubiquitination; Tumors AB Suppressor of cytokine signaling1 (SOCS1), as a member of SOCS family, has been widely studied in recent years. It has been found that SOCS1 not only participates in cell signaling, but also in ubiquitination mediated protein degradation process. Both of these two functions play an important role in the growth and proliferation of cells. Therefore, researchers speculated that SOCS1 also played an important role in tumors. This review mainly focuses on the structure, transcriptional regulation and functions of SOCS1 protein, and finally describes the possible clinical role of SOCS1 protein in tumors. C1 [Ying, Jie] Xuyi People’s Hospital, Department of Clinical Research Center, 211700 Xuyi, Jiangsu, China. [Qiu, Xiaoyan] Xuyi People’s Hospital, Department of Clinical Research Center, 211700 Xuyi, Jiangsu, China. [Yu, Lu] Xuyi People’s Hospital, Department of Clinical Research Center, 211700 Xuyi, Jiangsu, China. [Zhang, Miaomiao] Xuyi People’s Hospital, Department of Clinical Research Center, 211700 Xuyi, Jiangsu, China. RP Zhang, M (reprint author), Xuyi People’s Hospital, Department of Clinical Research Center, 211700 Xuyi, China. EM caotiantian198709@163.com CR Lemmon MA, Schlessinger J, 2000, Cell signaling by receptor tyrosine kinases. Cell 141(7):1117 Endo TA, Masuhara M, Yokouchi M, Suzuki R, Sakamoto H, Mitsui K, Matsumoto A, Tanimura S, Ohtsubo M, Misawa H, Miyazaki T, Leonor N, Taniguchi T, Fujita T, Kanakura Y, Komiya S, Yoshimura A, 1997, A new protein containing an SH2 domain that inhibits JAK kinases. Nature 387(6636):921–924 Hilton DJ, 1999, Negative regulators of cytokine signal transduction. Cell Mol Life Sci 55(12):1568–1577 Okochi O, Hibi K, Sakai M, Inoue S, Takeda S, Kaneko T et al, 2003, Methylation-mediated silencing of SOCS-1 gene in hepatocellular carcinoma derived from cirrhosis. Clin Cancer Res 9(14): 5295–5298 Chu PY, Yeh CM, Hsu NC, Chang YS, Chang JG, Yeh KT, 2010, Epigenetic alteration of the SOCS1 gene in hepatocellular carcinoma. Swiss Med Wkly 140(3):w13065 Li Z, Metze D, Nashan D, Mullertidow C, Serve HL, Poremba C et al, 2004, Expression of SOCS-1, suppressor of cytokine Signalling-1, in human melanoma. J Investig Dermatol 123(4): 737–745 Huang FJ, Steeg PS, Price JE, ChiuWT, Chou PC, Xie K, Sawaya R, Huang S, 2008, Molecular basis for the critical role of suppressor of cytokine signaling-1 in melanoma brain metastasis. Cancer Res 68(23):9634–9642 Oshimo Y, Kuraoka K, Nakayama H, Kitadai Y, Yoshida K, Chayama K, Yasui W, 2004, Epigenetic inactivation of SOCS-1 by CpG island hypermethylation in human gastric carcinoma. Int J Cancer 112(6):1003–1009 Shao N, Ma G, Zhang J, Zhu W, 2018, miR-221-5p enhances cell proliferation and metastasis through post-transcriptional regulation of SOCS1 in human prostate cancer. BMC Urol 18:14. https://doi. org/10.1186/s12894-018-0325-8 Linossi EM, Nicholson SE, 2012, The SOCS box-adapting proteins for ubiquitination and proteasomal degradation. IUBMB Life 64(4):316–323 Kamura T, Sato S, Haque D, Liu L, Jr KW, Conaway RC et al, 1998, The Elongin BC complex interacts with the conserved SOCS-box motif present in members of the SOCS, ras, WD-40 repeat, and ankyrin repeat families. Genes Dev 12(24):3872–3881 Babon JJ, Sabo JK, Soetopo A, Yao S, Bailey MF, Zhang JG, Nicola NA, Norton RS, 2008, The SOCS box domain of SOCS3: structure and interaction with the elonginBC-cullin5 ubiquitin ligase. J Mol Biol 381(4):928–940 Kamura T, Maenaka K, Kotoshiba S, Matsumoto M, Kohda D, Conaway RC et al, 2004, VHL-box and SOCS-box domains determine binding specificity for Cul2-Rbx1 and Cul5-Rbx2 modules of ubiquitin ligases. Genes Dev 18(24):3055–3065 Bullock AN, Debreczeni JE, Edwards AM, Sundstrom M, Knapp S, 2006, Crystal structure of the SOCS2-Elongin C-Elongin B complex defines a prototypical SOCS box ubiquitin ligase. Proc Natl Acad Sci U S A 103(20):7637–7642 Babon JJ,Mcmanus EJ,Yao S,Desouza DP, Mielke LA, Sprigg NS et al, 2006, The structure of SOCS3 reveals the basis of the extended SH2 domain function and identifies an unstructured insertion that regulates stability. Mol Cell 22(2):205–216 Yoshimura A, Ohkubo T, Kiguchi T, Jenkins NA, Gilbert DJ, Copeland NG, Hara T, Miyajima A, 1995, A novel cytokine-inducible gene CIS encodes an SH2-containing protein that binds to tyrosine-phosphorylated interleukin 3 and erythropoietin receptors. EMBO J 14(12):2816–2826 Kershaw NJ, Murphy JM, Liau NP, Varghese LN, Laktyushin A, Whitlock EL et al, 2013, SOCS3 binds specific receptor-JAK complexes to control cytokine signaling by direct kinase inhibition. Nat Struct Mol Biol 20(4):469–476 Babon JJ, Kershaw NJ, Murphy JM, Varghese LN, Laktyushin A, Young SN, Lucet IS, Norton RS, Nicola NA, 2012, Suppression of cytokine Signalling by SOCS3: characterisation of the mode of inhibition and the basis of its specificity. Immunity 36(2):239–250 Yandava CN, Pillari A, Drazen JM, 1999, Radiation hybrid and cytogenetic mapping of SOCS1 and SOCS2 to chromosomes 16p13 and 12q, respectively. Genomics 61(1):108–111 Naka T, Narazaki M, Hirata M, Matsumoto T, Minamoto S, Aono A, Nishimoto N, Kajita T, Taga T, Yoshizaki K, Akira S, Kishimoto T, 1997, Structure and function of a new STAT-induced STAT inhibitor. Nature 387(6636):924–929 Jegalian AG, Wu H, 2002, Regulation of Socs gene expression by the proto-oncoprotein GFI-1B: two routes for STAT5 target gene induction by erythropoietin. J Biol Chem 277(3):2345–2352 Sporri B,Kovanen PE, Sasaki A,Yoshimura A, LeonardWJ, 2001, JAB/SOCS1/SSI-1 is an interleukin-2-induced inhibitor of IL-2 signaling. Blood 97(1):221–226 Losman JA, Chen XP, Hilton D, Rothman P, 1999, Cutting edge: SOCS-1 is a potent inhibitor of IL-4 signal transduction. J Immunol 162(7):3770–3774 Dickensheets H, Vazquez N, Sheikh F, Gingras S,Murray PJ, Ryan JJ, Donnelly RP, 2007, Suppressor of cytokine signaling-1 is an IL- 4-inducible gene in macrophages and feedback inhibits IL-4 signaling. Genes Immun 8(1):21–27 Dickensheets HL, Venkataraman C, Schindler U, Donnelly RP, 1999, Interferons inhibit activation of STAT6 by interleukin 4 in human monocytes by inducing SOCS-1 gene expression. Proc Natl Acad Sci U S A 96(19):10800–10805 Hebenstreit D, Luft P, Schmiedlechner A, Regl G, Frischauf AM, Aberger F, Duschl A, Horejs-Hoeck J, 2003, IL-4 and IL-13 induce SOCS-1 gene expression in A549 cells by three functional STAT6- binding motifs located upstream of the transcription initiation site. J Immunol 171(11):5901–5907 Starr R,Willson TA, Viney EM, Murray LJ, Rayner JR, Jenkins BJ et al, 1997, A family of cytokine-inducible inhibitors of signalling. Nature 387(6636):917–921 Qing Y, Costapereira AP, Watling D, Stark GR, 2005, Role of tyrosine 441 of interferon-gamma receptor subunit 1 in SOCS-1- mediated attenuation of STAT1 activation. J Biol Chem 280(3): 1849–1853 Starr R, Fuchsberger M, Lau LS, Uldrich AP, Goradia A, Willson TA, Verhagen AM, Alexander WS, Smyth MJ, 2009, SOCS-1 binding to tyrosine 441 of IFN-gamma receptor subunit 1 contributes to the attenuation of IFN-gamma signaling in vivo. J Immunol 183(7):4537–4544 Song M, Shuai K, 1998, The suppressor of cytokine signaling, SOCS, 1 and SOCS3 but not SOCS2 proteins inhibit interferonmediated antiviral and antiproliferative activities. J Biol Chem 273(52):35056–35062 Crespo A, Filla MB, Russell SW, Murphy WJ, 2000, Indirect induction of suppressor of cytokine signalling-1 in macrophages stimulated with bacterial lipopolysaccharide: partial role of autocrine/ paracrine interferon-alpha/beta. Biochem J 349(Pt 1):99–104 Ram PA, Waxman DJ, 1999, SOCS/CIS protein inhibition of growth hormone-stimulated STAT5 signaling by multiple mechanisms. J Biol Chem 274(50):35553–35561 Hansen JA, Lindberg K, Hilton DJ, Nielsen JH, Billestrup N, 1999, Mechanism of inhibition of growth hormone receptor signaling by suppressor of cytokine signaling proteins. Mol Endocrinol 13(11): 1832–1843 Zhang J, Li H, Yu JP,Wang SE, Ren XB, 2012, Role of SOCS1 in tumor progression and therapeutic application. Int J Cancer 130(9): 1971–1980 Narazaki M, Fujimoto M, Matsumoto T, Morita Y, Saito H, Kajita T,Yoshizaki K, Naka T, Kishimoto T, 1998, Three distinct domains of SSI-1/SOCS-1/JAB protein are required for its suppression of interleukin 6 signaling. Proc Natl Acad Sci U S A 95(22):13130– 13134 Chen XP, Losman JA, Cowan S, Donahue E, Fay S, Vuong BQ, Nawijn MC, Capece D, Cohan VL, Rothman P, 2002, Pim serine/ threonine kinases regulate the stability of Socs-1 protein. Proc Natl Acad Sci U S A 99(4):2175–2180 Toniato E, Chen XP, Losman J, Flati V, Donahue L, Rothman P, 2002, TRIM8/GERP RING finger protein interacts with SOCS-1. J Biol Chem 277(40):37315–37322 Wu T, Xie M, Wang X, Jiang X, Li J, Huang H, 2012, miR-155 modulates TNF-α-inhibited osteogenic differentiation by targeting SOCS1 expression. Bone 51(3):498–505 Lu LF, Thai TH, Calado DP, Chaudhry A, Kubo M, Tanaka K, Loeb GB, Lee H, Yoshimura A, Rajewsky K, Rudensky AY, 2009, Foxp3-dependent microRNA155 confers competitive fitness to regulatory T cells through targeting SOCS1. Immunity 30(1):80–91 Yin Y, Liu W, Dai Y, 2015, SOCS3 and its role in associated diseases. Hum Immunol 76(10):775–780 Ilangumaran S, Ramanathan S, Rottapel R, 2004, Regulation of the immune system by SOCS family adaptor proteins. Semin Immunol 16(6):351–365 Hilton DJ, Richardson RT, AlexanderWS, Viney EM,Willson TA, Sprigg NS, Starr R, Nicholson SE, Metcalf D, Nicola NA, 1998, Twenty proteins containing a C-terminal SOCS box form five structural classes. Proc Natl Acad Sci U S A 95(1):114–119 Nicholson SE, Willson TA, Farley A, Starr R, Zhang JG, Baca M et al, 1999, Mutational analyses of the SOCS proteins suggest a dual domain requirement but distinct mechanisms for inhibition of LIF and IL-6 signal transduction. EMBO J 18(2):375–385 Ulevitch RJ, Tobias PS, 2003, Receptor-dependent mechanisms of cell stimulation by bacterial endotoxin. Annu Rev Immunol 13(1): 437–457 Aderem A, Ulevitch RJ, 2000, Toll-like receptors in the induction of the innate immune response. Nature 406(6797):782–787 Yuk JM, Jo EK, 2011, Toll-like receptors and innate immunity. J Bacteriol Virol 41(4):225 Akira S, Takeda K, Kaisho T, 2001, Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol 2(8): 675–680 Ziegler-HeitbrockHW(1995)Molecular mechanism in tolerance to lipopolysaccharide. J Inflamm 45(1):13–26 Nakagawa R, Naka T, Tsutsui H, Fujimoto M, Kimura A, Abe T, Seki E, Sato S, Takeuchi O, Takeda K, Akira S, Yamanishi K, Kawase I, Nakanishi K, Kishimoto T, 2002, SOCS-1 participates in negative regulation of LPS responses. Immunity 17(5):677–687 Mansell A, Smith R, Doyle SL, Gray P, Fenner JE, Crack PJ, Nicholson SE, Hilton DJ, O'Neill LAJ, Hertzog PJ, 2006, Suppressor of cytokine signaling 1 negatively regulates toll-like receptor signaling by mediating mal degradation. Nat Immunol 7(2):148–155 Mostecki J, Showalter BM, Rothman PB, 2005, Early growth Response-1 regulates lipopolysaccharide-induced suppressor of cytokine Signaling-1 transcription. J Biol Chem 280(4):2596–2605. , DOI 10.1074/jbc.M408938200 Babon J, Sabo J, Zhang JG, Nicola N, Norton R, 2009, The SOCS box encodes a hierarchy of affinities for Cullin5: implications for ubiquitin ligase formation and cytokine signalling suppression. J Mol Biol 387(1):162–174 Sepulveda PD, Ilangumaran S, Rottapel R, 2000, Suppressor of cytokine Signaling-1 inhibits VAV function through protein degradation. J Biol Chem 275(19):14005–14008 Monni R, Santos SC, Mauchauffe M, Berger R, Ghysdael J, Gouilleux F et al, 2001, The TEL-Jak2 oncoprotein induces Socs1 expression and altered cytokine response in Ba/F3 cells. Oncogene 20(7):849–858 Frantsve J, Schwaller J, Sternberg DW, Kutok J, Gilliland DG, 2001, Socs-1 inhibits TEL-JAK2-mediated transformation of hematopoietic cells through inhibition of JAK2 kinase activity and induction of proteasome-mediated degradation. Mol Cell Biol 21(10):3547–3557 De SP, Okkenhaug K, Rose JL, Hawley RG, Dubreuil P, Rottapel R, 1999, Socs1 binds to multiple signalling proteins and suppresses steel factor-dependent proliferation. EMBO J 18(4):904–915 Yasukawa H, Misawa H, Sakamoto H, Masuhara M, Sasaki A, Wakioka T et al, 1999, The JAK-binding protein JAB inhibits Janus tyrosine kinase activity through binding in the activation loop., Doctoral dissertation, Rennes 1) Linossi EM, Babon JJ, Hilton DJ, Nicholson SE, 2013, Suppression of cytokine signaling: the SOCS perspective. Cytokine Growth Factor Rev 24(3):241–248 Callus BA, Matheyprevot B, 1998, Interleukin-3-induced activation of the JAK/STAT pathway is prolonged by proteasome inhibitors. Blood 91(9):3182 Verdier F, Chretien S, Muller O, Varlet P, Yoshimura A, Gisselbrecht S et al, 1998, Proteasomes regulate erythropoietin receptor and signal transducer and activator of transcription 5, STAT5, activation. Possible involvement of the ubiquitinated Cis protein. J Biol Chem 273(43):28185 Wu QY, Zhu YY, Liu Y, Wei F, Tong YX, Cao J et al, 2018, CUEDC2, a novel interacting partner of the SOCS1 protein, plays important roles in the leukaemogenesis of acute myeloid leukaemia. Cell Death & Disease 9(7):774 Wu LM, Zhang F, Zhou L, Yang Z, Xie HY, Zheng SS, 2010, Predictive value of CpG island methylator phenotype for tumor recurrence in hepatitis B virus-associated hepatocellular carcinoma following liver transplantation. BMC Cancer 10(1):1–8 Nomoto S, Kinoshita T, Kato K, Otani S, Kasuya H, Takeda S, Kanazumi N et al, 2007, Hypermethylation of multiple genes as clonal markers in multicentric hepatocellular carcinoma. Br J Cancer 97(9):1260 Hussain S, Singh N, Salam I, Bandil K, Yuvaraj M, Akbar BM et al, 2011, Methylation-mediated gene silencing of suppressor of cytokine signaling-1, SOCS-1, gene in esophageal squamous cell carcinoma patients of Kashmir valley. J Recept Signal Transduct Res 31(2):147–156 Sobti RC, Singh N, Hussain S, Suri V, Nijhawan R, Bharti AC, Bharadwaj M, Das BC, 2011, Aberrant promoter methylation and loss of suppressor of cytokine signalling-1 gene expression in the development of uterine cervical carcinogenesis. Cell Oncol 34(6): 533–543 Chen CY, TsayW, Tang JL, Shen HL, Lin SW, Huang SY, Yao M, Chen YC, Shen MC, Wang CH, Tien HF, 2003, SOCS1 methylation in patients with newly diagnosed acute myeloid leukemia. Genes Chromosom Cancer 37(3):300–305 ZhangXH,Yang L, Liu XJ, Zhan Y, Pan YX,Wang XZ et al, 2018, Association between methylation of tumor suppressor gene SOCS1 and acute myeloid leukemia. Oncol Rep. , DOI 10.3892/or. 2018.6508 Galm O, Yoshikawa H, Esteller M, Osieka R, Herman JG, 2003, SOCS-1, a negative regulator of cytokine signaling, is frequently silenced by methylation in multiple myeloma. Blood 101(7):2784– 2788 Depil S, Saudemont A, Quesnel B, 2003, SOCS-1 gene methylation is frequent but does not appear to have prognostic value in patients with multiple myeloma. Leukemia 17(8):1678–1679 Komazaki T, Nagai H, EmiM, Terada Y, Yabe A, Jin E et al, 2004, Hypermethylation-associated inactivation of the SOCS-1 gene, a JAK/STAT inhibitor, in human pancreatic cancers. Jpn J Clin Oncol 34(4):191–194 Liu S, Ren S, Howell P, Fodstad O, Riker AI, 2008, Identification of novel epigenetically modified genes in human melanoma via promoter methylation gene profiling. Pigment Cell Melanoma Res 21(5):545–558 Sakamoto LH, De CB, Cajaiba M, Soares FA, Vettore AL, 2015, MT1G hypermethylation: a potential prognostic marker for hepatoblastoma. Pediatr Res 67(4):387–393 Zhang JG, Metcalf D, Rakar S, Asimakis M, Greenhalgh CJ, Willson TA, Starr R, Nicholson SE, Carter W, Alexander WS, Hilton DJ, Nicola NA, 2001, The SOCS box of suppressor of cytokine signaling-1 is important for inhibition of cytokine action in vivo. Proc Natl Acad Sci U S A 98(23):13261–13265 Shen L, Evelkabler K, Strube R, Chen SY, 2004, Silencing of SOCS1 enhances antigen presentation by dendritic cells and antigen-specific anti-tumor immunity. Nat Biotechnol 22(12): 1546–1553 Evelkabler K, Song XT, Aldrich M, Huang XF, Chen SY, 2006, SOCS1 restricts dendritic cells' ability to break self tolerance and induce antitumor immunity by regulating IL-12 production and signaling. J Clin Investig 116(1):90–100 Hong B, Ren W, Song XT, Evelkabler K, Chen SY, Huang XF, 2009, Human suppressor of cytokine signaling 1 controls immunostimulatory activity of monocyte-derived dendritic cells. Cancer Res 69(20):8076–8084 Hanada T, Tanaka K, Matsumura Y, Yamauchi M, Nishinakamura H, Aburatani H, Mashima R, Kubo M, Kobayashi T, Yoshimura A, 2005, Induction of hyper Th1 cell-type immune responses by dendritic cells lacking the suppressor of cytokine signaling-1 gene. J Immunol 174(7):4325–4332 Chikuma S, Kanamori M, Mise-Omata S, Yoshimura A, 2017, Suppressors of cytokine signaling: potential immune checkpoint molecules for cancer immunotherapy. Cancer Sci 108(4):574–580 Hashimoto M, Ayada T, Kinjyo I, Hiwatashi K, Yoshida H, Okada Y, Kobayashi T, Yoshimura A, 2009, Silencing of SOCS1 in macrophages suppresses tumor development by enhancing antitumor inflammation. Cancer Sci 100(4):730–736 Lesinski GB, Zimmerer JM, Kreiner M, Trefry J, Bill MA, Young GS et al, 2010, Modulation of SOCS protein expression influences the interferon responsiveness of human melanoma cells. BMC Cancer 10(1):142 Zitzmann K, Brand S, De Toni EN, Baehs S, Goke B, Meinecke J et al, 2007, SOCS1 silencing enhances antitumor activity of type I IFNs by regulating apoptosis in neuroendocrine tumor cells. Cancer Res 67(10):5025–5032 Sasi W, Wen GJ, Sharma A, Mokbel K, 2010, Higher expression levels of SOCS 1,3,4,7 are associated with earlier tumour stage and better clinical outcome in human breast cancer. BMC Cancer 10(1): 1–13 Roman-Gomez J, Jimenez-Velasco A, Castillejo J, Cervantes F, Barrios M, Colomer D et al, 2004, The suppressor of cytokine signaling-1 is constitutively expressed in chronic myeloid leukemia and correlates with poor cytogenetic response to interferon-alpha. Haematologica 89(1):42–48 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1295 EP 1301 DI 10.1007/s12253-019-00612-5 PG 7 ER PT J AU Liu, H Yin, Q Yang, G Qie, P AF Liu, Huining Yin, Qifan Yang, Guang Qie, Peng TI Prognostic Impact of Tumor Spread Through Air Spaces in Non-small Cell Lung Cancers: a Meta-Analysis Including 3564 Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Spread through air spaces; Non-small cell lung cancer; Prognosis; Meta-analysis ID Spread through air spaces; Non-small cell lung cancer; Prognosis; Meta-analysis AB Surgical resection is the most effective treatment for early stage lung adenocarcinoma. However, the rate of 5-year postoperative recurrence reaches 30%, Spread Through Air Spaces(STAS) is a recently described novel invasive pattern of lung cancer, According to the 2015 WHO classification. STAS is defined as micropapillary clusters, solid nests, or single cells spreading within air spaces beyond the edge of the main tumor, However, the prognostic role of STAS in lung cancer is not known, The aim of the current study is to evaluate the association between STAS and clinical outcome of lung cancer patients after surgical resection through ameta-analysis. Systematic research was conducted using three online databases to search for studies published before August 1, 2018. The 5-year RFS and OS for non-small cell lung cancer patients after surgical resection with or without STAS were compared. The studies were selected according to rigorous inclusion and exclusion criteria. Meta-analysis was performed using hazard ratio (HR) and 95% confidence intervals (CIs) as effective measures. Included in our meta-analysis were 12 studies, published from 2015 to 2018, with a total of 3564 patients. Our results clearly depicted that the presence of STAS predicted a worse outcome for 5-year RFS with the combined HR of 1.84(95% CI: 1.59–2.12). Meta-analysis of these 8 studies showed that patients with the presence of STAS were associated with shorter 5-year OS (the pooled HR:1.78, 95% CI: 1.51– 2.11). This meta-analysis illuminated that the presence of STAS might be a unfavorable prognostic factor for patients with NSCLC. it should be paid sufficient attention and recorded in pathologic reports, which can indicate treatment choice and prognosis of patients. In future, more studies with well-designed and large-scale are needed to confirm the conclusion. C1 [Liu, Huining] Hebei Provincal General Hospital, Department of Thoracic Surgery, 348,West He-Ping Road, 050051 Shijiazhuang, Hebei Province, China. [Yin, Qifan] Hebei Provincal General Hospital, Department of Thoracic Surgery, 348,West He-Ping Road, 050051 Shijiazhuang, Hebei Province, China. [Yang, Guang] Hebei Provincal General Hospital, Department of Thoracic Surgery, 348,West He-Ping Road, 050051 Shijiazhuang, Hebei Province, China. [Qie, Peng] Hebei Provincal General Hospital, Department of Thoracic Surgery, 348,West He-Ping Road, 050051 Shijiazhuang, Hebei Province, China. RP Liu, H (reprint author), Hebei Provincal General Hospital, Department of Thoracic Surgery, 050051 Shijiazhuang, China. EM liuhuining1972@163.com CR Brundage MD, Davies D, Mackillop WJ, 2002, Prognostic factors in non-small cell lung cancer: a decade of progress. Chest 122(3): 1037–1057 Siegel RL, Miller KD, Jemal A, 2015, Cancer statistics, 2015. CA Cancer J Clin 65(1):5–29 Mino-Kenudson M, Mark EJ, 2011, Reflex testing for epidermal growth factor receptor mutation and anaplastic lymphoma kinase fluoresce nce in situ hybridization in non-small cell lung cancer. Arch Pathol Lab Med 135(5):655–664 Hung J-J, Jeng W-J, Hsu W-H, Chou T-Y, Huang B-S, Wu Y-C, 2012, Predictors of death, local recurrence, and distant metastasis in completely resected pathological stage-I non–small-cell lung Cancer. J Thorac Oncol 7(7):1115–1123 Shiono S, Yanagawa N, 2016, Spread through air spaces is a predictive factor of recurrence and a prognostic factor in stage I lung adenocarcinoma. Interact Cardiovasc Thorac Surg 23(4):567–572 Kadota K, Nitadori J-I, Sima CS et al, 2015, Tumor spread through air spaces is an important pattern of invasion and impacts the frequency and location of recurrences after limited resection for small stage I lung adenocarcinomas. J Thorac Oncol 10(5):806–814 Warth A, Muley T, Kossakowski CA, Goeppert B, Schirmacher P, Dienemann H, Weichert W, 2015, Prognostic impact of intraalveolar tumor spread in pulmonary adenocarcinoma. Am J Surg Pathol 39(6):793–801 Lu S, Tan KS, Kadota K et al, 2016, Spread through air spaces, STAS, is an independent predictor of recurrence in lung squamous cell carcinoma. Mod Pathol 29:475A-A Morales-Oyarvide V,Mino-KenudsonM(2016, Tumor islands and spread through air spaces: distinct patterns of invasion in lung adenocarcinoma. Pathol Int 66(1):1–7 Dai C, Xie H, Su H, She Y, Zhu E, Fan Z, Zhou F, Ren Y, Xie D, Zheng H, Kadeer X, Chen D, Zhang L, Jiang G, Wu C, Chen C, 2017, Tumor spread through air spaces affects the recurrence and overall survival in patients with lung adenocarcinoma > 2 to 3 cm. J Thorac Oncol 12(7):1052–1060 Uruga H, Fujii T, Fujimori S, Kohno T, Kishi K, 2017, Semiquantitative assessment of tumor spread through air spaces, STAS, in early-stage lung adenocarcinomas. J Thorac Oncol 12(7):1046–1051 Kadota K, Kushida Y, Katsuki N, Ishikawa R, Ibuki E, Motoyama M, Nii K, Yokomise H, Bandoh S, Haba R, 2017, Tumor spread through air spaces is an independent predictor of recurrence-free survival in patients with resected lung squamous cell carcinoma. Am J Surg Pathol 41(8):1077–1086 Toyokawa G, Yamada Y, Tagawa T, Kozuma Y, Matsubara T, Haratake N, Takamori S, Akamine T, Oda Y, Maehara Y, 2018, Significance of spread through air spaces in resected pathological stage I lung adenocarcinoma. Ann Thorac Surg 105(6):1655–1663 Yanagawa N, Shiono S, Endo M, Ogata S-Y, 2018, Tumor spread through air spaces is a useful predictor of recurrence and prognosis in stage I lung squamous cell carcinoma, but not in stage II and III. Lung Cancer, Amsterdam, Netherlands, 120:14–21 Toyokawa G, Yamada Y, Tagawa T, Kinoshita F, Kozuma Y, Matsubara T, Haratake N, Takamori S, Akamine T, Hirai F, Oda Y, Maehara Y, 2018, Significance of spread through air spaces in resected lung adenocarcinomas with lymph node metastasis. Clin Lung Cancer 19:395–400.e1 Shiono S, Endo M, Suzuki K, Yarimizu K, Hayasaka K, Yanagawa N, 2018, Spread through air spaces is a prognostic factor in sublobar resection of non-small cell lung Cancer. Ann Thorac Surg 106(2):354–360 Toyokawa G, Yamada Y, Tagawa T, Oda Y, 2018, Significance of spread through air spaces in early-stage lung adenocarcinomas undergoing limited rese ction. Thorac Cancer 9(10):1255–1261 Lu S, Tan KS, Kadota K, Eguchi T, Bains S, Rekhtman N, Adusumilli PS, Travis WD, 2017, Spread through air spaces, STAS, is an independent predictor of recurrence and lung cancer-specific death in squamous cell carcinoma. J Thorac Oncol 12(2):223–234 Eguchi T, Kameda K, Lu S et al, 2017, OA07.06 in early-stage lung adenocarcinomas, survival by tumor size, T, is further stratified by tumor spread through air spaces. J Thorac Oncol 12(1, Supplement):S270-S1 Lu S, Eguchi T, Tan KS, Bains S, Kadota K, Rekhtman N, Adusumilli P, Travis W, 2017, Tumor spread through air spaces, STAS, in lung squamous cell cancer is an independent risk factor: a competing risk analysis. J Thorac Oncol 12(1): S412–S4S3 Yanagawa N, Shiono S, Abiko M, Suzuki K, Yarimizu K, 2017, The clinical impact of spread through air spaces, STAS, in surgically resected pStage I lung squamous cell carcinoma. J Thorac Oncol 12(1):S1119–S1S20 Yanagawa N, Shiono S, Endo M, Ogata SY, 2018, Tumor spread through air spaces is a useful predictor of recurrence and prognosis in stage I lung squamous cell carcinoma, but not in stage II and III. Lung Cancer, Amsterdam, Netherlands, 120:14–21 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1303 EP 1310 DI 10.1007/s12253-019-00616-1 PG 8 ER PT J AU Smolarz, B Brys, M Forma, E Zadrozny, M Bienkiewicz, J Romanowicz, H AF Smolarz, Beata Brys, Magdalena Forma, Ewa Zadrozny, Marek Bienkiewicz, Jan Romanowicz, Hanna TI Data on Single Nucleotide Polymorphism of DNA Repair Genes and Breast Cancer Risk from Poland SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DNA repair; Single nucleotide polymorphism; Breast cancer ID DNA repair; Single nucleotide polymorphism; Breast cancer AB Single nucleotide polymorphisms (SNPs) may modify the risk of cancer. They may be then regarded as potential markers of carcinogenesis. The aim of this study was to analyze the frequency of genotypes and alleles of SNPs in DNA repair genes and to investigate the influence this genetic variation exerts on breast cancer in Polish females. The test group comprised 600 females with breast cancer and 600 healthy controls. Genomic DNA was isolated and the SNPs in DNA repair genes were determined by High-Resolution Melter (HRM) technique. Following polymorphisms were analysed: Arg399Gln (rs25487) of the XRCC1, Gly322Asp (rs4987188) of the hMSH2, Lys751Gln (rs13181) of the XPD, Arg188His (rs3218536) of the XRCC2, P871L (rs799917) of the BRCA1 and N372H (rs144848) of the BRCA2 gene. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated for each genotype and allele. Statistically significant correlations were identified between 4 single nucleotide polymorphisms and the breast cancer risk: rs25487 rs4987188 rs13181 and rs799917. The alleles XRCC1-Gln (OR 5.11; 95% CI 5.68–11.64, p < .0001), hMSH2-Asp (OR 4.66; 95% CI 3.90–5.56, p < .0001), XPD-Gln (OR 2.65; 95% CI 2.24–3.14, p < .0001) and BRCA1-L (OR 1.45; 95% CI 1.24–1.71, p < .0001) genes were strongly correlated with this malignancy. No correlation was found between the studied SNPs and tumor grading nor the lymph node status. Further research on larger groups is warranted to determine the influence of above-mentioned genetic variants on breast cancer risk. C1 [Smolarz, Beata] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. [Brys, Magdalena] Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, Pomorska 141/143, 90-237 Lodz, Poland. [Forma, Ewa] Medical University of Lodz, Department of Chemistry and Clinical Biochemistry, Pomorska 141/143, 90-237 Lodz, Poland. [Zadrozny, Marek] Polish Mother’s Memorial Hospital - Research Institute, Department of Oncological Surgery and Breast Diseases, Rzgowska 281/289, 93-338 Lodz, Poland. [Bienkiewicz, Jan] Polish Mothers’ Memorial Hospital-Research Institute, Department of Surgical & Endoscopic Gynecology and Gynecologic Oncology, Rzgowska 281/289, 93-338 Lodz, Poland. [Romanowicz, Hanna] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. RP Smolarz, B (reprint author), Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, 93-338 Lodz, Poland. EM smolbea@wp.pl CR Jemal A, Siegel R,Ward E,HaoY, Xu J,Murray T, ThunMJ, 2008, Cancer statistics, 2008. CA Cancer J Clin 58:71–96 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D, Bray F, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403 Francisci S, Minicozzi P, Pierannunzio D, Ardanaz E, Eberle A, Grimsrud TK, Knijn A, Pastorino U, Salmeron D, Trama A, Sant M; EUROCARE-5 Working Group, 2015, Survival patterns in lung and pleural cancer in Europe 1999-2007: Results from the EUROCARE-5 study. Eur J Cancer 51:2242–2253 Coleman MP, Gatta G, Verdecchia A, Esteve J, Sant M, Storm H, Allemani C, Ciccolallo L, Santaquilani M, Berrino F, EUROCARE Working Group, 2003, EUROCARE-3 summary: cancer survival in Europe at the end of the 20th century. Ann Oncol 14:128–149 Martin AM,Weber BL, 2000, Genetic and hormonal risk factors in breast cancer. J Natl Cancer Inst 92:1126–1135 Kelley MR, Logsdon D, Fishel ML, 2014, Targeting DNA repair pathways for cancer treatment: what's new? Future Oncol 10:1215– 1237 Jackson SP, Bartek J, 2009, The DNA-damage response in human biology and disease. Nature 461:1071–1078 McCullough LE, Santella RM, Cleveland RJ,Millikan RC, Olshan AF, North KE, Bradshaw PT, Eng SM, Terry MB, Shen J, Crew KD, Rossner P Jr, Teitelbaum SL, Neugut AI, GammonMD, 2014, Polymorphisms in DNA repair genes, recreational physical activity and breast cancer risk. Int J Cancer 134: 654–663 Pruthi S, Gostout BS, Lindor NM, 2010, Identification and Management of Women with BRCA mutations or hereditary predisposition for breast and ovarian cancer. Mayo Clin Proc 85:1111– 1120 Pal T, Vadaparampil ST, 2012, Genetic risk assessments in individuals at high risk for inherited breast cancer in the breast oncology care setting. Cancer Control 19:255–266 Dowty JG, Win AK, Buchanan DD, Lindor NM, Macrae FA, Clendenning M, Antill YC, Thibodeau SN, Casey G, Gallinger S, Marchand LL, Newcomb PA, Haile RW, Young GP, James PA, Giles GG, Gunawardena SR, Leggett BA, Gattas M, Boussioutas A, Ahnen DJ, Baron JA, Parry S, Goldblatt J, Young JP, Hopper JL, Jenkins MA, 2013, Cancer risks for MLH1 and MSH2 mutation carriers. Hum Mutat 34:490–497 Wu S, Chen J, Ji Y, Liu Y, Gao L, Chen G, Shen K, Huang B, 2011, Association between the hMSH2 IVS12–6 T>C polymorphism and cancer risk: A meta-analysis. Exp Ther Med 2:1193–1198 Bansidhar BJ, 2012, Extracolonic manifestations of lynch syndrome. Clin Colon Rectal Surg 25:103–110 Maynard S, Schurman SH, Harboe C, de Souza-PintoNC, BohrVA, 2009, Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis 30:2–10 Huang JC, Hsu DS, Kazantsev A, Sancar A, 1994, Substrate spectrum of human excinuclease: repair of abasic sites, methylated bases, mismatches, and bulky adducts. Proc Natl Acad Sci U S A 91: 12213–12217 Tse D, Zhai R, Zhou W, Heist RS, Asomaning K, Su L, Lynch TJ, Wain JC, Christiani DC, Liu G, 2008, Polymorphisms of the NER pathway genes, ERCC1 and XPD are associated with esophageal adenocarcinoma risk. Cancer Causes Control 19:1077–1083 Kumar A, Pant MC, Singh HS, Khandelwal S, 2012, Associated risk of XRCC1 and XPD cross talk and life style factors in progression of head and neck cancer in north Indian population. Mutat Res 729:24–34 Davis JD, Lin SY, 2011, DNA damage and breast cancer. World J Clin Oncol 2:329–338 Abramson VG, Lehmann BD, Ballinger TJ, Pietenpol JA Subtyping of triple-negative breast cancer: implications for therapy. Cancer 121:8–16 Lieber MR, 2010, The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu Rev Biochem 79:181–211 Li X, HeyerWD, 2008, Homologous recombination in DNArepair and DNA damage tolerance. Cell Res 18:99–113 GorbunovaV, Seluanov A, Mao Z, Hine C, 2007, Changes inDNA repair during aging. Nucleic Acids Res 35:7466–7474 Khanna KK, Jackson SP, 2001, DNA double-strand breaks: signaling, repair and the cancer connection. Nat Genet 27:247–254 Zhao M, Chen P, Dong Y, Zhu X, Zhang X, 2014, Relationship between Rad51 G135C and G172T variants and the susceptibility to cancer: a meta-analysis involving 54 case-control studies. PLoS One 9:e87259 Al Zoubi MS, Zavaglia K, Mazanti C, Al Hamad M, Al Batayneh K, Aljabali AA, Bevilacqua G, 2017, Polymorphisms and mutations in GSTP1, RAD51, XRCC1 and XRCC3 genes in breast cancer patients. Int J Biol Markers. , DOI 10.5301/ijbm. 5000258 Romanowicz-Makowska H, Smolarz B, Zadrozny M, Westfa B, Baszczynski J, Kokolaszwili G, Burzyfiski M, Polac I, Sporny S, 2012, The association between polymorphisms of the RAD51- G135C, XRCC2-Arg188His and XRCC3-Thr241Met genes and clinico-pathologic features in breast cancer in Poland. Eur J Gynaecol Oncol 33:145–150 Romanowicz-Makowska H, Smolarz B, Zadrozny M, Westfal B, Baszczynski J, Polac I, Sporny S, 2011, Single nucleotide polymorphisms in the homologous recombination repair genes and breast cancer risk in Polish women. Tohoku J Exp Med 224:201–208 Michalska MM, Samulak D, Romanowicz H, Smolarz B, 2015, Single nucleotide polymorphisms, SNPs, of RAD51-G172T and XRCC2-41657C/T homologous recombination repair genes and the risk of triple- negative breast cancer in polish women. Pathol Oncol Res 21:935–940 Antoniou AC, Sinilnikova OM, Simard J, Leone M, Dumont M, Neuhausen SL, Struewing JP, Stoppa-Lyonnet D, Barjhoux L, Hughes DJ, Coupier I, Belotti M, Lasset C, Bonadona V, Bignon YJ, Genetic Modifiers of Cancer Risk in BRCA1/2 Mutation Carriers Study, GEMO), Rebbeck TR, Wagner T, Lynch HT, Domchek SM, Nathanson KL, Garber JE, Weitzel J, Narod SA, Tomlinson G, Olopade OI, Godwin A, Isaacs C, Jakubowska A, Lubinski J, Gronwald J, Gorski B, Byrski T, Huzarski T, Peock S, Cook M, Baynes C, Murray A, Rogers M, Daly PA, Dorkins H, Epidemiological Study of BRCA1 and BRCA2 Mutation Carriers, EMBRACE), Schmutzler RK, Versmold B, Engel C, Meindl A, Arnold N, Niederacher D, Deissler H, German Consortium for Hereditary Breast and Ovarian Cancer, GCHBOC), Spurdle AB, Chen X,Waddell N, Cloonan N, Kathleen Cuningham Consortium for Research into Familial Breast Cancer, kConFab), Kirchhoff T, Offit K, Friedman E, Kaufmann B, Laitman Y, Galore G, Rennert G, Lejbkowicz F, Raskin L, Andrulis IL, Ilyushik E, Ozcelik H, Devilee P, Vreeswijk MP, Greene MH, Prindiville SA, Osorio A, Benitez J, Zikan M, Szabo CI, Kilpivaara O, Nevanlinna H, Hamann U, Durocher F, Arason A, Couch FJ, Easton DF, Chenevix-Trench G, Consortium of Investigators of Modifiers of BRCA1/2, CIMBA,, 2007)RAD51 135G–>Cmodifies breast cancer risk among BRCA2 mutation carriers: results from a combined analysis of 19 studies. Am J Hum Genet 81:1186–1200 Kadouri L, Kote-Jarai Z, Hubert A, Durocher F, Abeliovich D, Glaser B, Hamburger T, Eeles RA, Peretz T, 2004, A singlenucleotide polymorphism in the RAD51 gene modifies breast cancer risk in BRCA2 carriers, but not in BRCA1 carriers or noncarriers. Br J Cancer 90:2002–2005 Wang WW, Spurdle AB, Kolachana P, Bove B, Modan B, Ebbers SM, Suthers G, TuckerMA, Kaufman DJ, DoodyMM, Tarone RE, Daly M, Levavi H, Pierce H, Chetrit A, kConFab, ABCFS/ CFRBCS, AJBCS, NISOC, Yechezkel GH, Chenevix-Trench G, Offit K, Godwin AK, Struewing JP, 2001, A single nucleotide polymorphism in the 5′ untranslated region of RAD51 and risk of cancer among BRCA1/2 mutation carriers. Cancer Epidemiol Biomark Prev 10:955–960 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1311 EP 1317 DI 10.1007/s12253-017-0370-8 PG 7 ER PT J AU Schneider, B Riedel, K Zhivov, A Huehns, M Zettl, H Guthoff, FR Junemann, A Erbersdobler, A Zimpfer, A AF Schneider, Bjoern Riedel, Katrin Zhivov, Andrey Huehns, Maja Zettl, Heike Guthoff, F Rudolf Junemann, Anselm Erbersdobler, Andreas Zimpfer, Annette TI Frequent and Yet Unreported GNAQ and GNA11 Mutations are Found in Uveal Melanomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Uveal melanoma; GNAQ/GNA11 mutation; Driver mutation frequency; Survival ID Uveal melanoma; GNAQ/GNA11 mutation; Driver mutation frequency; Survival AB Malignant melanoma of the uvea is the most common primary malignant tumor in the eye. We aimed to analyze GNAQ and GNA11 mutations in uveal melanomas using formalin-fixed, paraffin-embedded material and correlate the results with clinicopathological parameters. Tumor tissue was microdissected followed by amplification of GNAQ exon 4 and 5, GNA11 exon 4 and 5, and finally analyzed by Sanger sequencing.A total of 64.4GNA11/GNAQ mutations, including ten yet unreported, were found. Two cases showed multiple mutations. Overall survival was significantly shorter in the uveal melanoma cohort with GNAQ exon 5 mutation. In concordance with previous studies, high frequencies of mutations in GNAQ or GNA11 were detected. Interestingly, in about 20% of UM, not yet reported mutations in GNAQ or GNA11 were seen. Rarely, uveal melanoma may harbor double mutations in GNAQ and/or GNA11. Recent data imply, that implementation of GNAQ/GNA11 mutation analysis in routine diagnostic procedures might be helpful for future therapeutic decisions. C1 [Schneider, Bjoern] University Medicine Rostock, Institute of Pathology, Strempelstraße 14, 18055 Rostock, Germany. [Riedel, Katrin] University Medicine Rostock, Department of OphthalmologyRostock, Germany. [Zhivov, Andrey] OSG MVZ Betriebs GmbH, Ophtalmology, OMS, AnestheticsBamberg, Germany. [Huehns, Maja] University Medicine Rostock, Institute of Pathology, Strempelstraße 14, 18055 Rostock, Germany. [Zettl, Heike] University Medicine Rostock, Clinical Cancer RegistryRostock, Germany. [Guthoff, F Rudolf] University Medicine Rostock, Department of OphthalmologyRostock, Germany. [Junemann, Anselm] University Medicine Rostock, Department of OphthalmologyRostock, Germany. [Erbersdobler, Andreas] University Medicine Rostock, Institute of Pathology, Strempelstraße 14, 18055 Rostock, Germany. [Zimpfer, Annette] University Medicine Rostock, Institute of Pathology, Strempelstraße 14, 18055 Rostock, Germany. RP Schneider, B (reprint author), University Medicine Rostock, Institute of Pathology, 18055 Rostock, Germany. EM bjoern.schneider@med.uni-rostock.de CR Font RL, Croxatto JO, Narsing AR, 2006, Tumors of the eye and ocular adnexa. AFIP Atlas Tumor Pathol 4(5):48–60 Damato B, 2010, Does ocular treatment of uveal melanoma influence survival? Br J Cancer 103:285–290. , DOI 10.1038/sj. bjc.6605765 Curtin JA, Fridlyand J, Kageshita T et al, 2005, Distinct sets of genetic alterations in melanoma. N Engl J Med 353:2135–2147. , DOI 10.1056/NEJMoa050092 Zuidervaart W, van Nieuwpoort F, Stark M et al, 2005, Activation of the MAPK pathway is a common event in uveal melanomas although it rarely occurs through mutation of BRAF or RAS. Br J Cancer 92:2032–2038. , DOI 10.1038/sj.bjc.6602598 Weber A, Hengge UR, Urbanik D et al, 2003, Absence of mutations of the BRAF gene and constitutive activation of extracellularregulated kinase in malignant melanomas of the uvea. Lab Investig 83:1771–1776. , DOI 10.1097/01.LAB.0000101732. 89463.29 Onken MD, Worley LA, Long MD et al, 2008, Oncogenic mutations in GNAQ occur early in uveal melanoma. Invest Ophthalmol Vis Sci 49:5230–5234. , DOI 10.1167/iovs.08-2145 Bauer J, Kilic E, Vaarwater J et al, 2009, Oncogenic GNAQ mutations are not correlated with disease-free survival in uveal melanoma. Br J Cancer 101:813–815. , DOI 10.1038/sj.bjc. 6605226 Van Raamsdonk CD, Bezrookove V, Green G et al, 2009, Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature 457:599–602. , DOI 10.1038/nature07586 Van Raamsdonk CD, Griewank KG, Crosby MB et al, 2010, Mutations in GNA11 in uveal melanoma. N Engl J Med 363: 2191–2199. , DOI 10.1056/NEJMoa1000584 Populo H, Vinagre J, Lopes JM et al, 2011, Analysis of GNAQ mutations, proliferation and MAPK pathway activation in uveal melanomas. Br J Ophthalmol 95:715–719. , DOI 10. 1136/bjo.2009.174417 Daniels AB, Lee JE, MacConaill LE et al, 2012, High throughput mass spectrometry-based mutation profiling of primary uveal melanoma. Invest Ophthalmol Vis Sci 53:6991–6996. , DOI 10.1167/iovs.12-10427 Koopmans AE, Vaarwater J, Paridaens D, Rotterdam Ocular Melanoma Study group et al, 2013, Patient survival in uveal melanoma is not affected by oncogenic mutations in GNAQ and GNA11. Br J Cancer 109:493–496. , DOI 10.1038/bjc. 2013.299 Griewank KG, van de Nes J, Schilling B et al, 2014, Genetic and clinico-pathologic analysis of metastatic uveal melanoma. Mod Pathol 27:175–183. , DOI 10.1038/modpathol.2013.138 DonoM, Angelini G, CecconiMet al, 2014, Mutation frequencies of GNAQ, GNA11, BAP1, SF3B1, EIF1AX and TERT in uveal melanoma: detection of an activating mutation in the TERT gene promoter in a single case of uveal melanoma. Br J Cancer 110: 1058–1065. , DOI 10.1038/bjc.2013.804 Van Raamsdonk CD, Barsh GS, Wakamatsu K et al, 2009, Independent regulation of hair and skin color by two G proteincoupled pathways. Pigment Cell Melanoma Res 22:819–826. , DOI 10.1111/j.1755-148X.2009.00609.x FX Y, Luo J, Mo JS et al, 2014, Mutant Gq/11 promote uveal melanoma tumorigenesis by activating YAP. Cancer Cell 25:822– 830. , DOI 10.1016/j.ccr.2014.04.017 Cohen Y, Goldenberg-Cohen N, Parrella P et al, 2003, Lack of BRAF mutation in primary uveal melanoma. Invest Ophthalmol Vis Sci 44:2876–2878. , DOI 10.1167/iovs.02-1329 Cruz F 3rd, Rubin BP,Wilson D et al, 2003, Absence of BRAF and NRAS mutations in uveal melanoma. Cancer Res 63:5761–5766 Edmunds SC, Cree IA, Di Nicolantonio F et al, 2003, Absence of BRAF gene mutations in uveal melanomas in contrast to cutaneous melanomas. Br J Cancer 88:1403–1405. , DOI 10.1038/sj. bjc.6600919 Rimoldi D, Salvi S, Lienard D et al, 2003, Lack of BRAF mutations in uveal melanoma. Cancer Res 63:5712–5715 Kilic E, Bruggenwirth HT, Verbiest MM et al, 2004, The RASBRAF kinase pathway is not involved in uveal melanoma. Melanoma Res 14:203–205. , DOI 10.1097/01.cmr. 0000130006.46885.a0 Henriquez F, Janssen C, Kemp EG et al, 2007, The T1799A BRAF mutation is present in irismelanoma. Invest Ophthalmol Vis Sci 48: 4897–4900. , DOI 10.1167/iovs.07-0440 Mellen PL, Morton SJ, Shields CL, 2013, American joint committee on cancer staging of uveal melanoma. Oman J Ophthalmol 6: 116–118 O'Hayre M,Vazquez-Prado J,Kufareva I et al, 2013, The emerging mutational landscape of G proteins and G-protein-coupled receptors in cancer. Nat Rev Cancer 13:412–424. , DOI 10. 1038/nrc3521 Field MG, Harbour JW, 2014, Recent developments in prognostic and predictive testing in uveal melanoma. Curr Opin Ophthalmol 25:234–239. , DOI 10.1097/ICU.0000000000000051 Giannakis M, XJ M, Shukla SA et al, 2016, Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Rep 15:857– 865. , DOI 10.1016/j.celrep.2016.03.075 Feng X, Degese MS, Iglesias-Bartolome R et al, 2014, Hippoindependent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated rho GTPase signaling circuitry. Cancer Cell 25:831–845. , DOI 10.1016/j.ccr.2014.04.016 Opel KL, Chung D, McCord BR, 2010, A study of PCR inhibition mechanisms using real time PCR. J Forensic Sci 55:25–33. https:// doi.org/10.1111/j.1556-4029.2009.01245.x Metz CH, Lohmann D, ZeschnigkMet al, 2013, Uveal melanoma: current insights into clinical relevance of genetic testing. Klin Monatsbl Augenheilkd 230:686–691. , DOI 10.1055/s- 0033-1350628 Aalto Y, Eriksson L, Seregard S et al, 2001, Concomitant loss of chromosome 3 and whole armlosses and gains of chromosome 1, 6, or 8 in metastasizing primary uveal melanoma. Invest Ophthalmol Vis Sci 42:313–317 Harbour JW, 2012, The genetics of uveal melanoma: an emerging framework for targeted therapy. Pigment Cell Melanoma Res 25: 171–181. , DOI 10.1111/j.1755-148X.2012.00979.x Komatsubara KM, Manson DK, Carvajal RD, 2016, Selumetinib for the treatment of metastatic uveal melanoma: past and future perspectives. Future Oncol 12:1331–1344. , DOI 10. 2217/fon-2015-0075 Lyubasyuk V, Ouyang H, FX Yet al, 2015, YAP inhibition blocks uveal melanogenesis driven by GNAQ or GNA11 mutations. Mol Cell Oncol 2:e970957. , DOI 10.4161/23723548.2014. 970957 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1319 EP 1325 DI 10.1007/s12253-017-0371-7 PG 7 ER PT J AU Singh, A Singh, N Behera, D Sharma, S AF Singh, Amrita Singh, Navneet Behera, Digambar Sharma, Siddharth TI Genetic Investigation of Polymorphic OGG1 and MUTYH Genes Towards Increased Susceptibility in Lung Adenocarcinoma and its Impact on Overall Survival of Lung Cancer Patients Treated with Platinum Based Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Polymorphism; Lung cancer; Overall survival; Chemotherapy; OGG1; MUTHY ID Polymorphism; Lung cancer; Overall survival; Chemotherapy; OGG1; MUTHY AB Genes OGG1 and MUTYH are the two primary genes in Base excision repair pathway. OGG1 hydrolyzes the sugar phosphate backbone and remove the damaged base creating abasic site. MUTYH complements OGG1 as it particularly remove adenine mispaired with 8-oxo-G. Both OGG1 and MUTYH act as a check for the mis-incorporation of bases may be due to damages incurred on DNA. DNA isolation for 326 lung cancer cases and 330 controls was followed by genotyping making use of PCRRFLP. Logistic regression was done to analyze the risk towards lung cancer. Patients were followed through telephonic conversation. Kaplan meier and Cox-regression were used for survival analysis. OGG1 presented a high risk towards lung cancer (CG: OR = 2.44, p = 0.0003; CG + GG: OR = 1.88, p = 0.0093). On the same lines adenocarcinoma for OGG1 were potent risk factors towards lung cancer (CG: OR = 4.72, p = 0.0002; CG + GG: OR = 3.63, p = 0.0018). Single allelic carriers for MUTYH gene imposed a high risk towards overall lung susceptibility and for all the three histology. Stratified analysis for chemotherapeutic drugs revealed administration of Cisplatin/Carboplatin + Pemtrexed for OGG1Ser 326 Cys showed a better survival (MST CG vs. CC: 9.1 vs. 0.56, p = <0.0001; HR =0.051, p = 0.0025). Whereas, MUTYH Gln324His showed a smaller survival for mutant genotype (CC) (MST CC vs. GG: 4.0 vs. 9.4, p = 0.05; HR = 1.75, p = 0.26). Single allelic carriers for both OGG1 and MUTYH were risk factors towards lung cancer. The risk was amplified on combining both OGG1 and MUTYH. C1 [Singh, Amrita] Thapar University, Department of Biotechnology, 147002 Patiala, Punjab, India. [Singh, Navneet] Post Graduate Institute of Medical Education & Research (PGIMER), Sector 14, Department of Pulmonary MedicineChandigarh, India. [Behera, Digambar] Post Graduate Institute of Medical Education & Research (PGIMER), Sector 14, Department of Pulmonary MedicineChandigarh, India. [Sharma, Siddharth] Thapar University, Department of Biotechnology, 147002 Patiala, Punjab, India. RP Sharma, S (reprint author), Thapar University, Department of Biotechnology, 147002 Patiala, India. EM siddharthsharma.phd@thapar.edu CR Parikh PM, RanadeAA, Babu G, Ghadyalpatil N, Singh R, Bharath R, Bhattacharyya GS, Koyande S, Singhal M, Vora A, Verma A, Hingmire S, 2016, Lung cancer in India: current status and promising strategies. South Asian J Cancer 5(3):93–95 Peng Y, Li Z, Zhang S, Xiong Y, Cun Y, Qian C, Li M, Ren T, Xia L, Cheng Y, Wang D, 2014, Association of DNA base excision repair genes, OGG1, APE1 and XRCC1, polymorphisms with outcome to platinum-based chemotherapy in advanced non small-cell lung cancer patients. Int J Cancer 135:2687–2696 Butkiewicz D, Rusin M, Sikora B, Lach A, Chorazy M, 2011, An association between DNA repair gene polymorphisms and survival in patients with resected non-small cell lung cancer. Mol Biol Rep 38:5231–5241 Zheng L, GuanW, Li M-x, Zhong Z-y, Qian C-y, Yang X-q, Liao L, Li Z-p, Wang D, 2011, Genetic polymorphism of DNA Baseexcision repair genes, APE1, OGG1 and XRCC1, and their correlation with risk of lung cancer in a Chinese population. Arch Med Res 42(3):226–234 Sliwinski T, Przybylowska K, Markiewicz L, Rusin P, Pietruszewska W, Zelinska-Blizniewska H, Olszewski J, Morawiec-Sztandera A, Mlynarski W, Majsterek I, 2011, MUTYH Tyr165Cys, OGG1 Ser326Cys and XPD Lys751Gln polymorphisms and head neck cancer susceptibility: a case control study. Mol Biol Rep 38:1251–1261 DuanW-X, Hua R-X, YiW, Shen L-J, Jin Z-X, Zhao Y-H, Yi D-H, Chen W-S, Yu S-Q, 2012, The association between OGG1 Ser326Cys polymorphism and lung cancer susceptibility: a meta analysis of 27 studies. PLoS One 7(4):e35970 Poulsen MLM, Bisgaard ML, 2008, MUTYH Associated Polyposis, MAP). Curr Genomics 9(6):420–435 KasaharaM, Osawa K, Yoshida K,Miyaishi A, Osawa Y, Inoue N, Tsutou A, Tabuchi Y, Tanaka K, Yamamoto M, Shimada E, Takahashi J, 2008, Association of MUTYH Gln324His and APEX1 Asp148Glu with colorectal cancer and smoking in a Japanese population. J Exp Clin Cancer Res 27:49 Yan Y, Chen X, Li T, Li M, Liang H, 2014, Association of OGG1 Ser326Cys polymorphism and pancreatic cancer susceptibility: evidence from a meta-analysis. Tumor Biol 35:2397–2402 Nielsen M, van Steenbergen LN, Jones N, Vogt S, Vasen HFA, Morreau H, Aretz S, Sampson JR, Dekkers OM, Janssen-Heijnen MLG, Hes FJ, 2010, Survival of MUTYH-associated polyposis patients with colorectal cancer and matched control colorectal cancer patients. Natl Cancer Inst 102:1724–1730 Singh A, Singh N, Behera D, Sharma S, 2017, Polymorphism in XRCC1 gene modulates survival and clinical outcomes of advanced north Indian lung cancer patients treated with platinumbased doublet chemotherapy. Med Oncol 34:64 Singh A, Singh N, Behera D, Sharma S, 2016, Association and multiple interaction analysis among five XRCC1 polymorphic variants in modulating lung cancer risk in north Indian population. DNA Repair 47:30–41 Ding X, Wang K, Wu Z, Yao A, Li J, Jiao C, Qian J, Bai D, Li X, 2015, The Ser326Cys polymorphism of hOGG1 is associated with intrahepatic cholangiocarcinoma susceptibility in a Chinese population. Int J Clin Exp Med 8(9):16294–16300 Synowiec E, Wojcik KA, Czubatka A, Polakowski P, Izdebska J, Szaflik J, Blasiak J, Szaflik JP, 2015, Lack of association between polymorphisms of the DNA base excision repair genes MUTYH and hOGG1 and keratoconus in a Polish subpopulation. Arch Med Sci 11(5):1101–1110 Nohmi T, Kim SR, Yamada M, 2005, Modulation of oxidative mutagenesis and carcinogenesis by polymorphic forms of human DNA repair enzymes. Mutat Res 591:60–73 Qian B, Zhang H, Zhang L, Zhou X, Yu H, Chen K, 2011, Association of genetic polymorphisms in DNA repair pathway genes with non-small cell lung cancer risk. Lung Cancer 73:138– 146 Kohno T, Kunitoh H, Mimaki S, Shiraishi K, Kuchiba A, Yamamoto S, Yokota J, 2011, Contribution of the TP53, OGG1, CHRNA3, and HLA-DQA1 genes to the risk for lung squamous cell carcinoma. J Thorac Oncol 6:4 Li Z, GuanW, Li M-x, Zhong Z-y, Qian C-y, Yang X-q, Liao L, Li Z-p,Wang D, 2011, Genetic polymorphism of DNA Base-excision repair genes, APE1, OGG1 and XRCC1, and their correlation with risk of lung cancer in a Chinese population. Arch Med Res 42:226– 234 Le Marchand L, Donlon T, Lum-Jones A, Seifried A, Wilkens LR, 2002, Association of the hOGG1 Ser326Cys polymorphism with lung cancer risk. Cancer Epidemiol Biomark Prev 11:409–412 Couto PG, Bastos-Rodrigues L, Carneiro JG, Guieiro F, Bicalho MA, Leidenz FB, Bicalho AJ, Friedman E, De Marco L, 2015, DNA Base-excision repair genes OGG1 and NTH1 in Brazilian lung cancer patients. Mol Diagn Ther 19:389–395 Lai C-Y, Hsieh L-L, Tang R, Santella RM, Chang-Chieh CR, Yeh C-C, 2016, Association between polymorphisms of APE1 and OGG1 and risk of colorectal cancer in Taiwan. World J Gastroenterol 22(12):3372–3380 Letkova L, Matakova T, Musak L, Sarlinova M, Krutakova M, Slovakova P, Kavcova E, Jakusova V, Janickova M, Drgova A, Berzinec P, Halasova E, 2013, DNA repair genes polymorphism and lung cancer risk with the emphasis to sex differences.Mol Biol Rep 40(9):5261–5273 Mittal RD, Mandal RK, Gang R, 2012, Base excision repair pathway genes polymorphism in prostate and bladder cancer risk in north Indian population. Mech Ageing Dev 133:127–132 Karahalil B, Emerce E, Kocer B, Han S, Alkis N, Karakaya AE, 2008, The association of OGG1 ser326cys polymorphism and urinary 8-ohdg levels with lung cancer susceptibility: a hospital based case-control study in Turkey. Arh Hig Rada Toksikol 59: 241–250 De Ruyck K, Szaumkessel M, De Rudder I, Dehoorne A, Vral A, Claes K, Velghe A, Van Meerbeeck J, Thierens H, 2007, Polymorphisms in base-excision repair and nucleotide-excision repair genes in relation to lung cancer risk. Mutat Res 631:101–110 Kohno T, Kunitoh H, Toyama K,Yamamoto S, Kuchiba A, Saito D, Yanagitani N, Ishihara S-i, Saito R, Yokota J, 2006, Association of the OGG1-Ser326Cys polymorphism with lung adenocarcinoma risk. Cancer Sci 97:724–728 Ito H, Hamajima N, Takezaki T, Matsuo K, Tajima K, Hatooka S, Mitsudomi T, Suyama M, Sato S, Ueda R, 2002, A limited association of OGGI Ser326Cys polymorphism for adenocarcinoma of the lung. J Epidemiol 12(3):258–265 Miyaishi A, Osawa K, Osawa Y, Inoue N, Yoshida K, Kasahara M, Tsutou A, Tabuchi Y, Sakamoto K, Tsubota N, Takahashi J, 2009, MUTYH Gln324His gene polymorphism and genetic susceptibility for lung cancer in a Japanese population. J Exp Clin Cancer Res 28: 10 Przybylowska K, Kabzinski J, Sygut A, Dziki L, Dziki A, Majsterek I, 2013, An association selected polymorphisms of XRCC1, OGG1 and MUTYH gene and the level of efficiency oxidative DNA damage repair with a risk of colorectal cancer. Mutat Res 745–746:6–15 Szaflik JP, Cuchra M, Przybylowska-Sygut K, Dziki L, Kurowska AK, Gacek M, Drzewoski J, Szaflik J, Majsterek I, 2013, Association of the 399Arg/Gln XRCC1, the 194 Arg/Trp XRCC1, the 326Ser/Cys OGG1, and the 324Gln/His MUTYH gene polymorphisms with clinical parameters and the risk for development of primary open-angle glaucoma. Mutat Res 753:12–22 Li Z, GuanW, Li M-x, Zhong Z-y, Qian C-y, Yang X-q, Liao L, Li Z-p,Wang D, 2011, Genetic polymorphism of DNA Base-excision repair genes, APE1, OGG1 and XRCC1, and their correlation with risk of lung cancer in a Chinese population. Arch Med Res 42: 226e234 Sua Y, Zhanga H, Xua F, Konga J, Yub H, Qian B, 2015, DNA repair gene polymorphisms in relation to non-small cell lung cancer survival. Cell Physiol Biochem 36(4):1419–1429 Liddiard K, Hills R, Burnett AK, Darley RL, Tonks A, 2010, OGG1 is a novel prognostic indicator in acute myeloid leukaemia. Oncogene 29:2005–2012 Cecchin E, Agostini M, Pucciarelli S, De Paoli A, Canzonieri V, Sigon R, De Mattia E, Friso ML, Biason P, Visentin M, Nitti D, Toffoli G, 2011, Tumor response is predicted by patient genetic profile in rectal cancer patients treated with neo-adjuvant chemoradiotherapy. Pharmacogenomics J 11:214–226 Monteiro E, Varzim G, Silva R, da Costa L, 2005, Polymorphisms of the human OGG1 gene in laryngeal cancer: implications in radiotherapy response and survival. Rev Laryngol Otol Rhinol 126(3):135–140 Shinmura K, Goto M, Suzuki M, Tao H, Yamada H, Igarashi H, Matsuura S, Maeda M, Konno H, Matsuda T, Sugimura H, 2011, Reduced expression of MUTYH with suppressive activity against mutations caused by 8-hydroxyguanine is a novel predictor of a poor prognosis in human gastric cancer. J Pathol 225:414–423 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1327 EP 1340 DI 10.1007/s12253-017-0372-6 PG 14 ER PT J AU Raphael, J Nofech-Mozes, Sh Paramsothy, Th Li, N Gandhi, S AF Raphael, Jacques Nofech-Mozes, Sharon Paramsothy, Thivaher Li, N Gandhi, Sonal TI Tumor Infiltrating Lymphocytes in Breast Cancer Patients with Progressive Disease during Neoadjuvant Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Disease progression; Neoadjuvant chemotherapy; Tumor infiltrating lymphocytes ID Breast cancer; Disease progression; Neoadjuvant chemotherapy; Tumor infiltrating lymphocytes AB A minority of breast cancer (BC) patients progress during neoadjuvant chemotherapy (NCT). The aim of this study was to assess the value of Tumor infiltrating lymphocytes (TILs) in such a high-risk population where valid biomarkers are eagerly needed. A retrospective review identified BC patients who either progressed during NCT or achieved a pathologic complete response (pCR). An experienced BC pathologist semi-quantified stromal TILs in pre-treatment core biopsies using hematoxylin and eosin stained slides. The primary outcome was to compare the levels of TILs between the 2 groups as a continuous and categorical variable using the t-test and X2 test as appropriate. The secondary outcome was to compare survival outcomes between patients with high versus low TILs level using the log-rank test. Fifty patients were successfully identified and assessed for TILs: 21 progressed during NCT and 29 had a pCR. Patients with progressive disease were older with more advanced disease (p = 0.03, p = 0.0001 respectively). A significantly lower mean level of TILs was found in patients with progressive disease compared to patients with pCR: 14.3% (Standard Deviation (SD): 16.9) versus 32.8% (SD: 31), p = 0.01). The level of TILs was neither associated with baseline characteristics nor with survival outcomes. BC patients progressing during NCT have low TILs levels compared to patients with pCR. Prospective studies are needed to establish the utility of TILs as early biomarkers of tumor response, particularly in patients with disease progression who need novel treatment approaches. C1 [Raphael, Jacques] Sunnybrook Health Sciences Centre, Department of Medicine, Division of Medical Oncology, 2075 Bayview Avenue, M4N3M5 Toronto, ON, Canada. [Nofech-Mozes, Sharon] Sunnybrook Health Sciences Centre, Department of PathologyToronto, ON, Canada. [Paramsothy, Thivaher] Sunnybrook Health Sciences Centre, Department of Medicine, Division of Medical Oncology, 2075 Bayview Avenue, M4N3M5 Toronto, ON, Canada. [Li, N] Sunnybrook Research InstituteToronto, ON, Canada. [Gandhi, Sonal] Sunnybrook Health Sciences Centre, Department of Medicine, Division of Medical Oncology, 2075 Bayview Avenue, M4N3M5 Toronto, ON, Canada. RP Raphael, J (reprint author), Sunnybrook Health Sciences Centre, Department of Medicine, Division of Medical Oncology, M4N3M5 Toronto, Canada. EM raphaeljack13@hotmail.com CR Kong X, Moran MS, Zhang N et al, 2011, Meta-analysis confirms achieving pathological complete response after neoadjuvant chemotherapy predicts favourable prognosis for breast cancer patients. Eur J Cancer 47:2084–2090 Broglio KR, Quintana M, Foster M et al, 2016, Association of Pathologic Complete Response to Neoadjuvant therapy in HER2- positive breast cancer with long-term outcomes: a meta-analysis. JAMA Oncol 2(6):751–760 Cortazar P, Zhang L, Untch M et al, 2014, Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet 384(9938):164–172 De Mattos-Arruda L, Shen R, Reis-Filho JS, Cortes J, 2016, Translating neoadjuvant therapy into survival benefits: one size does not fit all. Nat Rev Clin Oncol 13(9):566–579 Weigelt B et al, 2005, Breast cancer metastasis: markers and models. Nat Rev Cancer 5:591–602 Von Minckwitz G, Blohmer JU, Costa SD et al, 2013, Responseguided neoadjuvant chemotherapy for breast cancer. J Clin Oncol 31(29):3623–3630 Kim KI, Lee KH, Kim TR et al, 2014, Ki-67 as a predictor of response to Neoadjuvant chemotherapy in breast cancer patients. J Breast Cancer 17(1):40–46 Denkert C, Loibl S, Noske A et al, 2010, Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol 28(1):105–113 Von Minckwitz G, Untch M, Nuesch E et al, 2011, Impact of treatment characteristics on response of different breast cancer phenotypes: pooled analysis of the German neo-adjuvant chemotherapy trials. Breast Cancer Res Treat 125:145–156 Mamounas EP, Cortazar P, Zhang L, et al, 2014, Loco-regional recurrence after neoadjuvant chemotherapy: Pooled analysis results from the Collaborative Trials in Neoadjuvant Breast Cancer, CTNeoBC). J Clin Oncol 32(Suppl 26; Abstr 61) Zambetti M, Mansutti M, Gomez P et al, 2012, Pathological complete response rates following different neoadjuvant chemotherapy regimens for operable breast cancer according to ER status, in two parallel, randomized phase II trials with an adaptive study design, ECTO II). Breast Cancer Res Treat 132(3):843–851 Straver ME, Rutger EJ, Rodenhuis S et al, 2010, The relevance of breast cancer subtypes in the outcome of neoadjuvant chemotherapy. Ann Surg Oncol 17:2411–2418 CaudleAS,Gonzalez-Angulo AM,Hunt KK et al, 2010, Predictors of tumor progression during neoadjuvant chemotherapy in breast cancer. J Clin Oncol 28(11):1821–1828 Raphael J, Paramsothy T, Li N, Lee J, Gandhi SA, 2017, Singleinstitution experience of salvage therapy for patients with early and locally advanced breast cancer who progress during neoadjuvant chemotherapy. Breast Cancer Res Treat 163(1):11–19 Denkert C, von Minckwitz G, Brase JC et al, 2015, Tumorinfiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J Clin Oncol 33:983–991 West NR, Milne K, Truong PT et al, 2011, Tumor-infiltrating lymphocytes predict response to anthracycline-based chemotherapy in estrogen receptor-negative breast cancer. Breast Cancer Res 13:R126 Yamaguchi R, Tanaka M, Yano A et al, 2012, Tumor-infiltrating lymphocytes are important pathologic predictors for neoadjuvant chemotherapy in patients with breast cancer. Hum Pathol 43: 1688–1694 Loi S, Sirtaine N, Piette F et al, 2013, Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98. J Clin Oncol 31:860–867 Adams S, Gray RJ, Demaria S et al, 2014, Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J Clin Oncol 32:2959–2966 Loi S, Michiels S, Salgado R et al, 2014, Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann Oncol 25(8):1544–1550 Edge SB, Byrd DR, Compton CC et al, eds,, 2010, AJCC Cancer Staging Manual, 7th edn. Springer, New York, pp 347–376 Hammond ME, Hayes DF, Dowsett M et al, 2010, ASCO/CAP guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol 28: 2784–2795 Wolff AC, Hammond ME, Hicks DG et al, 2013, Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: ASCO/CAP clinical practice guideline update. J Clin Oncol 31(31):3997–4013 Pennisi A, Kieber-Emmons T, Makhoul I, Hutchins L, 2016, Relevance of pathological complete response after Neoadjuvant therapy for breast cancer. Breast Cancer, Auckl, 10:103–106 Salgado R, Denkert C, Demaria S et al, 2015, The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: recommendations by an international TILs working group 2014. Ann Oncol 26(2):259–271 Ingold Heppner B, Loibl S, Denkert C, 2016, Tumor-infiltrating lymphocytes: a promising biomarker in breast cancer. Breast Care, Basel, 11(2):96–100 Schemper M, Smith TLA, 1996, Note on quantifying follow-up in studies of failure time. Control Clin Trials 17:343–346 Kaplan EL, Meier P, 1958, Nonparametric estimation from incomplete observations. J Am Stat Assoc 53(282):457–481 Yu X, Zhang Z, Wang Z et al, 2016, Prognostic and predictive value of tumor-infiltrating lymphocytes in breast cancer: a systematic review and meta-analysis. Clin Transl Oncol 18(5):497–506 Harris LN, Ismaila N, McShane LM et al, 2016, Use of biomarkers to guide decisions on adjuvant systemic therapy for women with early-stage invasive breast cancer: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol 34(10):1134–1150 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1341 EP 1347 DI 10.1007/s12253-017-0368-2 PG 7 ER PT J AU Matyas, P Postyeni, E Komlosi, K Szalai, R Bene, J Magyari, L Melegh, B Hadzsiev, K AF Matyas, Petra Postyeni, Etelka Komlosi, Katalin Szalai, Renata Bene, Judit Magyari, Lili Melegh, Bela Hadzsiev, Kinga TI Age-Related Hearing Impairment Associated NAT2, GRM7, GRHL2 Susceptibility Gene Polymorphisms and Haplotypes in Roma and Hungarian Populations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NAT2; GRM7; GRHL2; ARHI; Roma; Hungarian ID NAT2; GRM7; GRHL2; ARHI; Roma; Hungarian AB Age-related hearing impairment (ARHI) is the most frequent sensory disease in the elderly, which is caused by an interaction between genetic and environmental factors. Here we examined the ethnic differences, allele and genotype frequencies of the NAT2, GRM7, and GRHL2 genes pooled samples of healthy Hungarian and healthy and hearing impaired Roma people. Study populations of healthy Hungarian and Roma subjects were characterized for the rs1799930 NAT2, rs11928865 GRM7, rs10955255, rs13263539, and rs1981361 GRHL2 polymorphisms and deaf Roma subjects were characterized for the rs1799930 NAT2, rs13263539, and rs1981361 GRHL2 using a PCR-RFLP method. We found significant differences in minor allele frequencies for GRHL2 rs13263539 and rs1981361 polymorphism between healthy Roma and Hungarian samples (37.9% vs. 51.0% and 43.6% vs. 56.2%, respectively; p < 0.05). The differences of homozygous genotype of GRHL2 rs13263539 and rs1981361 variants, values were also significantly different (13.0% vs. 25.3% and 16.5 vs. 32.3%; p < 0.05). The NAT2 rs1799930 homozygous genotype was 14.0% in healthy Romas and 7.7% in Hungarians, while the minor A allele frequency was 38.0% and 26.7% in Roma and Hungarian population, respectively (p < 0.05). Furthermore, the frequency of GGT, GAC and GAT haplotypes was significantly higher in the Hungarian population than in healthy Roma (1.87 vs. 4.47%, 0.91 vs. 2.07% and 1.15 vs. 5.51%, respectively; p < 0.008). Present study revealed significant interethnic differences in allele polymorphisms of NAT2, GRM7 and GRHL2 exhibit quite marked ethnic differences in Roma populations that might have important implications for the preventive and therapeutic treatments in this population. C1 [Matyas, Petra] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Postyeni, Etelka] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Komlosi, Katalin] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Szalai, Renata] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Bene, Judit] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Magyari, Lili] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Melegh, Bela] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. [Hadzsiev, Kinga] University of Pecs, Department of Medical Genetics and Child Development, Szigeti 12, H-7624 Pecs, Hungary. RP Melegh, B (reprint author), University of Pecs, Department of Medical Genetics and Child Development, H-7624 Pecs, Hungary. EM melegh.bela@pte.hu CR Staecker H, ZhengQY, Van DeWater TR, 2001, Oxidative stress in aging in the C57B16/J mouse cochlea. Acta Otolaryngol 121(6): 666–672 Cruickshanks KJ,Wiley TL, Tweed TS, Klein BE, Klein R,Mares- Perlman JA, Nondahl DM, 1998, Prevalence of hearing loss in older adults in beaver dam, Wisconsin. The epidemiology of hearing loss study. Am J Epidemiol 148(9):879–886 Unal M, Tamer L, Dogruer ZN, Yildirim H, Vayisoglu Y, Camdeviren H, 2005, N-acetyltransferase 2 gene polymorphism and presbycusis. Laryngoscope 115(12):2238–2241 Yamasoba T, Lin FR, Someya S, Kashio A, Sakamoto T, Kondo K, 2013, Current concepts in age-related hearing loss: epidemiology and mechanistic pathways. Hear Res 303:30–38 Seidman MD, Khan MJ, TangWX, Quirk WS, 2002, Influence of lecithin on mitochondrial DNA and age-related hearing loss. Otolaryngol Head Neck Surg 127(3):138–144 Van Eyken E, Van Camp G, Fransen E, Topsakal V, Hendrickx JJ, Demeester K, Van de Heyning P, Maki-Torkko E, Hannula S, Sorri M, JensenM, Parving A, BilleM, BaurM, PfisterM, Bonaconsa A, Mazzoli M, Orzan E, Espeso A, Stephens D, Verbruggen K, Huyghe J, Dhooge I, Huygen P, Kremer H, Cremers CW, Kunst S,ManninenM, Pyykko I, Lacava A, SteffensM,Wienker TF, Van Laer L, 2007, Contribution of the N-acetyltransferase 2 polymorphism NAT2*6A to age-related hearing impairment. J Med Genet 44(9):570–578 Bared A, Ouyang X, Angeli S, Du LL, Hoang K, Yan D, Liu XZ, 2010, Antioxidant enzymes, presbycusis, and ethnic variability. Otolaryngol Head Neck Surg 143(2):263–268 Pujol R, Rebillard G, Puel JL, Lenoir M, Eybalin M, Recasens M, 1990, Glutamate neurotoxicity in the cochlea: a possible consequence of ischaemic or anoxic conditions occurring in ageing. Acta Otolaryngol Suppl 476:32–36 Friedman RA, Van Laer L, Huentelman MJ, Sheth SS, Van Eyken E, Corneveaux JJ, TembeWD, Halperin RF, Thorburn AQ, Thys S, Bonneux S, Fransen E, Huyghe J, Pyykko I, Cremers CW, Kremer H, Dhooge I, Stephens D, Orzan E, Pfister M, Bille M, Parving A, Sorri M, Van de Heyning PH, Makmura L, Ohmen JD, Linthicum FH, Jr., Fayad JN, Pearson JV, Craig DW, Stephan DA, Van Camp G, 2009, GRM7 variants confer susceptibility to age-related hearing impairment. Hum Mol Genet 18, 4):785–796 Vona B, Nanda I, Neuner C,Muller T, Haaf T, 2013, Confirmation of GRHL2 as the gene for the DFNA28 locus. Am J Med Genet A 161A(8):2060–2065 Peters LM, AndersonDW, Griffith AJ, GrundfastKM, San Agustin TB, Madeo AC, Friedman TB, Morell RJ, 2002, Mutation of a transcription factor, TFCP2L3, causes progressive autosomal dominant hearing loss, DFNA28. Hum Mol Genet 11(23):2877–2885 Van Laer L, Van Eyken E, Fransen E, Huyghe JR, Topsakal V, Hendrickx JJ, Hannula S, Maki-Torkko E, Jensen M, Demeester K, Baur M, Bonaconsa A, Mazzoli M, Espeso A, Verbruggen K, Huyghe J, Huygen P, Kunst S, Manninen M, Konings A, Diaz- Lacava AN, Steffens M, Wienker TF, Pyykko I, Cremers CW, Kremer H, Dhooge I, Stephens D, Orzan E, Pfister M, Bille M, Parving A, Sorri M, Van de Heyning PH, Van Camp G, 2008, The grainyhead like 2 gene, GRHL2), alias TFCP2L3, is associated with age-related hearing impairment. Hum Mol Genet 17(2):159–169 Morar B, Gresham D, Angelicheva D, Tournev I, Gooding R, Guergueltcheva V, Schmidt C, Abicht A, Lochmuller H, Tordai A, Kalmar L, Nagy M, Karcagi V, Jeanpierre M, Herczegfalvi A, Beeson D, Venkataraman V, Warwick Carter K, Reeve J, de Pablo R, Kucinskas V, Kalaydjieva L, 2004, Mutation history of the roma/gypsies. Am J Hum Genet 75(4):596–609 Gresham D, Morar B, Underhill PA, Passarino G, Lin AA,Wise C, Angelicheva D, Calafell F, Oefner PJ, Shen P, Tournev I, de Pablo R, Kucinskas V, Perez-Lezaun A, Marushiakova E, Popov V, Kalaydjieva L, 2001, Origins and divergence of the Roma, gypsies). Am J Hum Genet 69(6):1314–1331 Hajioff S, McKee M, 2000, The health of the Roma people: a review of the published literature. J Epidemiol Community Health 54(11):864–869 Seidman MD, Khan MJ, Dolan DF, Quirk WS, 1996, Age-related differences in cochlear microcirculation and auditory brain stem response. Arch Otolaryngol Head Neck Surg 122(11):1221–1226 Gates GA, Couropmitree NN, Myers RH, 1999, Genetic associations in age-related hearing thresholds. Arch Otolaryngol Head Neck Surg 125(6):654–659 ColingDE,Yu KC, Somand D, Satar B, Bai U, Huang TT, Seidman MD, Epstein CJ, Mhatre AN, Lalwani AK, 2003, Effect of SOD1 overexpression on age- and noise-related hearing loss. Free Radic Biol Med 34(7):873–880 Ates NA, UnalM, Tamer L, Derici E, Karakas S, Ercan B, Pata YS, Akbas Y, Vayisoglu Y, Camdeviren H, 2005, Glutathione Stransferase gene polymorphisms in presbycusis. Otol Neurotol 26(3):392–397 Pujol R, Puel JL, Gervais d'Aldin C, Eybalin M, 1993, Pathophysiology of the glutamatergic synapses in the cochlea. Acta Otolaryngol 113(3):330–334 Steinbach S, Lutz J, 2007, Glutamate induces apoptosis in cultured spiral ganglion explants. Biochem Biophys Res Commun 357(1): 14–19 Sipeky C, Matyas P, Melegh M, Janicsek I, Szalai R, Szabo I, Varnai R, Tarlos G, Ganczer A, Melegh B, 2014, Lower carrier rate of GJB2 W24X ancestral Indian mutation in Roma samples from Hungary: implication for public health intervention. Mol Biol Rep 41(9):6105–6110 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1349 EP 1355 DI 10.1007/s12253-018-0388-6 PG 7 ER PT J AU Sastre-Heres, JA Iglesias, I Alaguero-Calero, M Ruiz-Sanchez, D Garcia-Diaz, B Pena-Diaz, J AF Sastre-Heres, Jose Alejandro Iglesias, Irene Alaguero-Calero, Miguel Ruiz-Sanchez, Daniel Garcia-Diaz, Benito Pena-Diaz, Jaime TI Comparative Study of Different Classification Models in Renal-Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Renal cell cancer; Classification; Predictive; Prognostic ID Renal cell cancer; Classification; Predictive; Prognostic AB The aim of this study was to compare the Memorial Sloan-Kettering Cancer Center (MSKCC) and the Cleveland Clinic Foundation (CCF) models of classification of aRCC patients. In addition, the model developed from the pivotal trial of temsirolimus and those proposed by Motzer et al. in 2004, Escudier et al., Heng et al., Choueiri et al. and Bamias et al. were examined. An observational, retrospective study of patients starting first-line systemic therapy was conducted between 2008 and 2011. The variables used to evaluate the classification models were median overall survival (mOS) and median progression-free survival (mPFS). The comparison of different classification models was performed by comparing the area under the ROC (Receiver Operating Characteristic) curve (AUC) for time-dependent variables proposed by Heagerty. Eighty-eight patients were included. When the different models were compared, it was found that although based on the mOS, the Escudier model had better short-term (1-year) prognostic value, followed by the Heng model; in the long term, the models that presented a higher prognosis capacity were the Hudes and CCF models, closely followed by the Heng model. In addition, the Heng model had a slightly higher predictive ability than the other models. Based on the results, and in line with the European society for medical oncology (ESMO) guidelines, it appears that the model of Heng could be the best model to classify patients with aRCC and combines good shortand long-term prognostics while possessing better predictive ability and a more equal distribution of patients. C1 [Sastre-Heres, Jose Alejandro] Hospital San Juan de Dios de Zaragoza, Servicio de Farmacia, C/ Paseo Colon n°14, 50006 Zaragoza, Spain. [Iglesias, Irene] Ciudad Universitaria, University Complutense of Madrid, Faculy of Pharmacy, Plaza Ramon y Cajal, 28040 Madrid, Spain. [Alaguero-Calero, Miguel] Hospital Universitario Central de Asturias, Hospital Pharmacy Department, C/ Julian Claveria s/n, 33006 Oviedo, Asturias, Spain. [Ruiz-Sanchez, Daniel] Hospital Universitario Central de Asturias, Hospital Pharmacy Department, C/ Julian Claveria s/n, 33006 Oviedo, Asturias, Spain. [Garcia-Diaz, Benito] Hospital Universitario Severo Ochoa, C/ Avenida de Orellana, s/n, 28911 Leganes, Madrid, Spain. [Pena-Diaz, Jaime] University of Granada, Spanish Society of Hospital Pharmacy, Faculty of Pharmacy, Campus Universitario de Cartuja, 18071 Granada, Spain. RP Sastre-Heres, JA (reprint author), Hospital San Juan de Dios de Zaragoza, Servicio de Farmacia, 50006 Zaragoza, Spain. EM asastre@ohsjd.es CR Ljungberg B, Hanbury DC, Kuczyk MA et al, 2007, Renal cell carcinoma guideline. Eur Urol 51:1502–1510 Sanchez Folgueras MV, Palacio Vazquez IP, 2009, Registro hospitalario de tumores del Servicio de Salud del Principado de Asturias. Ano 2009. Astursalud website. http://www.hca.es/huca/ web/contenidos/servicios/rt/rt2012/rt2012.pdf. Accessed 05 Oct 2015 ChowW, Gridley G, Fraumeni J et al, 2000, Obesity, hypertension and the risk of kidney cancer inmen. N Engl J Med 343:1305–1311 Escudier B, Eisen T, Porta C et al, 2014, Clinical practice guidelines renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up clinical practice guidelines. Ann Oncol 25(3):49–56 NCNN Clinical practice guidelines in oncology. Version 1.2013. Kidney cancer. The National Comprehensive Cancer Network website http://www.nccn.org/professionals/physician_gls/f_ guidelines_nojava.asp#site. Accessed 05 Oct 2015 Motzer RJ, Bacik J, Murphy BA et al, 2002, Interferon-alfa as a comparative treatment for clinical trials of new therapies against advanced renal cell carcinoma. J Clin Oncol 20(1):289–296 Motzer RJ, Bacik J, Schwartz LH et al, 2004, Prognostic factors for survival in previously treated patients with metastatic renal cell carcinoma. J Clin Oncol 22(3):454–463 Mekhail TM,Abou-JawdeRM, Boumerhi Get al, 2005)Validation and extension of the memorial Sloan- Kettering prognostic factors model for survival in patients with previously untreated metastatic renal cell carcinoma. J Clin Oncol 23(4):832–841 Hudes G, Carducci M, Tomczak P et al, 2007, Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 356:2271–2281 Escudier B, Choueiri TK, Oudard S et al, 2007, Prognostic factors of metastatic renal cell carcinoma after failure of immunotherapy: new paradigm from a large phase III trial with shark cartilage extract AE 941. J Urol 178(5):1901–1905 Choueiri TK, Garcia JA, Elson P et al, 2007, Clinical factors associated with outcome in patients with metastatic clear-cell renal cell carcinoma treated with vascular endothelial growth factor-targeted therapy. Cancer 110(3):543–550 Heng DY, Xie W, Regan MM et al, 2009, Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol 27(34):5794–5799 Bamias A, Karadimou A, Lampaki S et al, 2010, Prognostic stratification of patients with advanced renal cell carcinoma treated with sunitinib: comparison with the memorial Sloan-Kettering prognostic factors model. BMC Cancer 2(10):45 Patil S, Figlin RA, Hutson TE et al, 2011, Prognostic factors for progression-free and overall survival with sunitinib targeted therapy and with cytokine as first-line therapy in patients with metastatic renal cell carcinoma. Ann Oncol 22(2):295–300 Heagerty PJ, Zheng Y, 2005, Survival model predictive accuracy and ROC curves. Biometrics 61(1):92–105 Sachin S, 2010, Pharmacotherapy self-assessment program 6th edition. American College of Clinical Pharmacy website. http://www. accp.com/bookstore/p6_se.aspx. Accessed 10 Oct 2015 Aguilo M, Alba F, Barrio J et al, 2007, Renal cell cancer. Pharm Lett 9(10):8592, eLetter, http://www.dicaf.es/pharmletter.php#. VgQutn1kbGQ. Accessed 10 Oct 2015 Karakiewcz P, Sun M, Bellmunt J et al, 2011, Prediction of progression-free survival rates after bevacizumab plus interferon alone in patients with metastatic renal cell carcinoma: comparion of a normogram to the Motzer criteria. Eur Urol 60:48–56 Sun M, Shariat SF, Cheng C et al, 2011, Prognostic factors and predictive models in renal cell carcinoma: a contemporary review. Eur Urol 60:644–661 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1357 EP 1362 DI 10.1007/s12253-018-0385-9 PG 6 ER PT J AU Santos Cirqueira, C Sousa Felipe-Silva, A Wakamatsu, A Vieira Marins, L Cavalcanti Rocha, E Sobroza de Mello, E Avancini Ferreira Alves, V AF Santos Cirqueira, Cinthya Sousa Felipe-Silva, Aloisio Wakamatsu, Alda Vieira Marins, Lidiane Cavalcanti Rocha, Eziel Sobroza de Mello, Evandro Avancini Ferreira Alves, Venancio TI Immunohistochemical Assessment of the Expression of Biliary Transportation Proteins MRP2 and MRP3 in Hepatocellular Carcinoma and in Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ATP-binding cassette transporters; Hepatocellular carcinoma; Cholangiocarcinoma; Immunohistochemistry ID ATP-binding cassette transporters; Hepatocellular carcinoma; Cholangiocarcinoma; Immunohistochemistry AB Multidrug resistance-associated protein 2 (MRP2) is a multi-specific organic anion transporter predominantly expressed in the canalicular membrane of hepatocytes, epithelial cells from gallbladder and apical membranes of proximal tubular kidney epithelium whereas multidrug resistance-associated protein 3 (MRP3) is present in the basolateral membrane of hepatocytes and cholangiocytes. This study aims to detect the expression of these transporters in hepatocellular carcinoma (HCC) and in cholangiocarcinoma (CC), searching for evidences for future studies on differential diagnosis and on clinical essays. The immunohistochemical reactivity (IHC) of these transporters was assessed in tissue microarrays of 80 HCC and 56 CC cases using monoclonal antibodies and compared with anatomopathological (AP) variables. The positivity of MRP2 was observed in 92.3% of HCC and in 96.3% of CC. The detection of high MRP2 expression in HCC was not significantly different (p > 0.05) according to the size, number of nodules architectural pattern and growth pattern of HCC and CC. Regarding histological grades, 22/22 well moderately differentiated HCC versus 50/56 poorly differentiated HCC were positive for MRP2. A trend for lower expression in poor differentiation HCC was found. And 50/50 well/moderately differentiated CC versus 2/4 poorly/undifferentiated CC were positive for MRP2. This result showed a reduced expression (p = 0,0004) in poorly differentiated CC. MRP3 positivity was observed in 18.8% of HCC and was not significantly different according to AP parameters. MRP3 was expressed in 44.5% CC, with a trend for lower expression in less differentiated CC and significantly lower rates in the ductular histological subtype (p = 0.023). The high expression of MRP2 in HCC and in CC is conserved regardlessmost of the anatomopathological parameters, except for a trend of lower expression in less differentiated HCC and CC. The observation of lower MRP3 expression in less differentiated CC and, especially, in the histological subtype with expression of hepatic progenitor cell phenotypes leads to future opportunities to evaluate the expression of this marker in cholangiocarcinomas. C1 [Santos Cirqueira, Cinthya] Instituto Adolfo Lutz, Centro de Patologia, Nucleo de Anatomia PatologicaSao Paulo, Brazil. [Sousa Felipe-Silva, Aloisio] Sao Paulo University School of Medicine, Department of PathologySao Paulo, Brazil. [Wakamatsu, Alda] Sao Paulo University School of Medicine, Department of PathologySao Paulo, Brazil. [Vieira Marins, Lidiane] Sao Paulo University School of Medicine, Department of PathologySao Paulo, Brazil. [Cavalcanti Rocha, Eziel] Sao Paulo University School of Medicine, Department of PathologySao Paulo, Brazil. [Sobroza de Mello, Evandro] Sao Paulo University School of Medicine, Department of PathologySao Paulo, Brazil. [Avancini Ferreira Alves, Venancio] Sao Paulo University School of Medicine, Department of PathologySao Paulo, Brazil. RP Santos Cirqueira, C (reprint author), Instituto Adolfo Lutz, Centro de Patologia, Nucleo de Anatomia Patologica, Sao Paulo, Brazil. EM cinthyacirqueira@yahoo.com.br CR Vasiliou V, Vasiliou K, Nebert DW, 2009, Human ATP-binding cassette, ABC, transporter family. Hum Genomics 3:281–290. , DOI 10.1186/1479-7364-3-3-281 Stefkova J, Poledne R, Hubacek JA, 2004, ATP-binding cassette, ABC, transporters in human metabolism and diseases. Physiol Res 53(3):235–243 Tarling EJ, de Aguiar Vallim TQ, Edwards PA, 2013, Role of ABC transporters in lipid transport and human disease. Trends Endocrinol Metab 24(7):342–350. , DOI 10.1016/j.tem. 2013.01.006 Lagana SM, Salomao M, Remotti HE, Knisely AS, Moreira RK, 2015, Bile salt export pump: a sensitive and specific immunohistochemical marker of hepatocellular carcinoma. Histopathology 66(4):598–602. , DOI 10.1111/his.12601 Nguyen T, Phillips D, Jain D, Torbenson M,Wu T-T, YehMMet al, 2015, Comparison of 5 immunohistochemical markers of hepatocellular differentiation for the diagnosis of hepatocellular carcinoma. Arch Pathol Lab Med 139(8):1028–1034. , DOI 10. 5858/arpa.2014-0479-OA Fujikura K, Yamasaki T, Otani K, Kanzawa M, Fukumoto T, Ku Y et al, 2016, BSEP and MDR3 – useful immunohistochemical markers to discriminate hepatocellular carcinomas from intrahepatic cholangiocarcinomas and hepatoid carcinomas. Am J Surg Pathol 40(5):689–696. , DOI 10.1097/PAS. 0000000000000585 Trauner M, Boyer JL, 2003, Bile salt transporters: molecular characterization, function, and regulation. Physiol Rev 83(2):633–671. , DOI 10.1152/physrev.00027.2002 ABCC2: ATP-binding cassette, sub-family C, CFTR/MRP), member 2. [Internet]. The Human Protein Atlas. 2017. http://www.proteinatlas.org/ENSG00000023839-ABCC2/tissue. Accessed 31 Jan 2017 Taniguchi K, Wada M, Kohno K, Nakamura T, Kawabe T, Kawakami M et al, 1996, A human canalicular multispecific organic anion transporter, cMOAT, gene is overexpressed in cisplatin-resistant human cancer cell lines with decreased drug accumulation. Cancer Res 56(18):4124–4129 Chen Z, Shi T, Zhang L, Zhu P, Deng M, Huang C et al, 2016, Mammalian drug efflux transporters of the ATP binding cassette, ABC, family in multidrug resistance: a review of the past decade. Cancer Lett 370(1):153–164. , DOI 10.1016/j.canlet.2015. 10.010 Nies AT, Konig J, Cui Y, Brom M, Spring H, Keppler D, 2002, Structural requirements for the apical sorting of human multidrug resistance protein 2, ABCC2). Eur J Biochem 269(7):1866–1876. , DOI 10.1007/s00424-006-0109-y Jedlitschky G, Hoffmann U, Kroemer HK, 2006, Structure and function of the MRP2, ABCC2, protein and its role in drug disposition. Expert Opin Drug Metab Toxicol 2(3):351–366 Alrefai WA, Gill RK, 2007, Bile acid transporters: structure, function, regulation and pathophysiological implications. Pharm Res 10:1803–1823. , DOI 10.1517/17425255.2.3.351 Erlinger S, Arias IM, Dhumeaux D, 2014, Inherited disorders of bilirubin transport and conjugation: new insights into molecular mechanisms and consequences. Gastroenterology 146(7):1625– 1638. , DOI 10.1053/j.gastro.2014.03.047 Keppler D, 2014, The roles of MRP2, MRP3, OATP1B1, and OATP1B3 in conjugated hyperbilirubinemia. Drug Metab Dispos 42(4):561–565. , DOI 10.1124/dmd.113.055772 Kool M, van der Linden M, de Haas M, Scheffer GL, de Vree JM, Smith AJ et al, 1999, MRP3, an organic anion transporter able to transport anti-cancer drugs. Proc Natl Acad Sci U S A 96(12): 6914–6919. , DOI 10.1073/pnas.96.12.6914 Scheffer GL, Kool M, Heijn M, de Haas M, Pijnenborg AC, Wijnholds J et al, 2000, Specific detection of multidrug resistance proteinsMRP1, MRP2, MRP3, MRP5, and MDR3 P-glycoprotein with a panel of monoclonal antibodies. Cancer Res 60(18):5269– 5277 Klaassen CD, Aleksunes LM, 2010, Xenobiotic, bile acid, and cholesterol transporters: function and regulation. Pharmacol Rev 62(1):1–96. , DOI 10.1124/pr.109.002014 ABCC3: ATP-binding cassette, sub-family C, CFTR/MRP), member 3 [Internet]. The Human Protein Atlas. 2017. http://www. proteinatlas.org/ENSG00000108846-ABCC3/tissue. Accessed 31 Jan 2017 ABCC3: ATP-binding cassette, sub-family C, CFTR/MRP), member 3 [Internet]. GenAtlas - Universite Paris Descartes. 2017. http:// genatlas.medecine.univ-paris5.fr/fiche.php?symbol=ABCC3. Accessed 31 Jan 2017 Konig J, Rost D, Cui Y, Keppler D, 1999, Characterization of the human multidrug resistance protein isoform MRP3 localized to the basolateral hepatocyte membrane. Hepatology 29(4):1156–1163. , DOI 10.1002/hep.510290404 Ballatori N, Hammond CL, Cunningham JB, Krance SM,Marchan R, 2005, Molecular mechanisms of reduced glutathione transport: role of the MRP/CFTR/ABCC and OATP/SLC21A families of membrane proteins. Toxicol Appl Pharmacol 204(3):238–255. , DOI 10.1016/j.taap.2004.09.008 Belinsky MG, Dawson PA, Shchaveleva I, Bain LJ, Wang R, Ling V et al, 2005, Analysis of the in vivo functions of Mrp3. Mol Pharmacol 68(1):160–168. , DOI 10.1124/mol.104. 010587 Halilbasic E, Claudel T, Trauner M, 2013, Bile acid transporters and regulatory nuclear receptors in the liver and beyond. J Hepatol 58(1):155–168. , DOI 10.1016/j.jhep.2012.08.002 Sodani K, Patel A, Kathawala RJ, Chen Z-S, 2012, Multidrug resistance associated proteins in multidrug resistance. Chin J Cancer 31(2):58–72. , DOI 10.5732/cjc.011.10329 Rau S, Autschbach F, Riedel HD, Konig J, Kulaksiz H, Stiehl A et al, 2008, Expression of the multidrug resistance proteins MRP2 and MRP3 in human cholangiocellular carcinomas. Eur J Clin Investig 38(2):134–142. , DOI 10.1111/j.1365-2362.2007. 01916.x Tomonari T, Takeishi S, Taniguchi T, Tanaka T, Tanaka H, Fujimoto S et al, 2016, MRP3 as a novel resistance factor for sorafenib in hepatocellular carcinoma. Oncotarget 7(6):7207– 7215. , DOI 10.18632/oncotarget.6889 Zollner G, Wagner M, Fickert P, Silbert D, Fuchsbichler A, Zatloukal K et al, 2005, Hepatobiliary transporter expression in human hepatocellular carcinoma. Liver Int, 2):367–379. https:// doi.org/10.1111/j.1478-3231.2005.01033.x Nies AT, Konig J, Pfannschmidt M, Klar E, Hofmann WJ, Keppler D, 2001, Expression of the multidrug resistance proteins MRP2 and MRP3 in human hepatocellular carcinoma. Int J Cancer 94(4):492–499. , DOI 10.1111/j.1478-3231.2005.01033.x Borght VS, Libbrecht L, Blokzijl H, Faber NK, Moshage H, Aerts R et al, 2005, Diagnostic and pathogenetic implications of the expression of hepatic transporters in focal lesions occurring in normal liver. J Pathol 207(4):471–482. , DOI 10.1002/path.1852 Apte U, Krisnamurthy P, 2010, Molecular pathology of liver diseases. In: Monga SPS, ed, Molecular pathology of liver diseases 1st edn. Springer, Boston, pp 147–164 Korita PV, Wakai T, Shirai Y, Matsuda Y, Sakata J, Takamura M et al, 2010, Multidrug resistance-associated protein 2 determines the efficacy of cisplatin in patients with hepatocellular carcinoma. Oncol Rep 4:965–972. , DOI 10.3892/ijmm.2012.1173 Atilano-Roque A, Roda G, Fogueri U, Kiser JJ, JoyMS, 2016, Effect of disease pathologies on transporter expression and function. J Clin Pharmacol 56(Suppl7):S205–S221. , DOI 10.1002/jcph.768 Buettner S, van Vugt JLA, IJzermans J, Groot Koerkamp B, 2017, Intrahepatic cholangiocarcinoma: current perspectives. Onco Targets Ther 10:1131–1142. , DOI 10.2147/OTT.S93629 Nakanuma Y, Kakuda Y, 2015, Pathologic classification of cholangiocarcinoma: new concepts. Best Pract Res Clin Gastroenterol 29(2):277–293. , DOI 10.1016/j.bpg.2015.02.006 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1363 EP 1371 DI 10.1007/s12253-018-0386-8 PG 9 ER PT J AU Srividya, A Giridhar, H Vishwanath, S Chakraborty, A AF Srividya, Arjuna Giridhar, B. Hosmane Vishwanath, Silpa Chakraborty, Anirban TI Profiling of Germline Mutations in Major Hotspot Codons of TP53 Using PCR-RFLP SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tumor suppressor protein TP53; Mutation; Hotspot codon; Homoallelic mutation; Heteroallelic mutation; Cancer ID Tumor suppressor protein TP53; Mutation; Hotspot codon; Homoallelic mutation; Heteroallelic mutation; Cancer AB Tumor suppressor protein, TP53 also known as the "guardian of the genome" plays a key role in preventing malignant transformation. Almost 50% of human tumors carry mutations in this gene; in the remaining tumors, the TP53 network is functionally inoperative. The majority of TP53 mutations are missense mutations and more than 90% of the missense mutations affect specific codons in the DNA-binding domain, called "hotspot codons". The present study was aimed at analyzing the germline mutation status of four hotspot codons in TP53 namely, codon 175, codon 245, codon 248 (within the DNA binding domain) and codon 72 (outside the DNA binding domain) in cancer cases encountered in a tertiary care hospital in South India by PCR-RFLP. The casecontrol study included 85–10 subjects respectively. The results of the study indicated that majority of the cancer cases did not harbor germline mutations in the four hot spot codons of TP53. The study further highlights the usefulness of PCR-RFLP as a simple and cost effective tool for checking gene mutations. C1 [Srividya, Arjuna] Nitte University Centre for Science Education and Research (NUCSER), Division of Molecular Genetics and Cancer, Kotekar-Beeri Road, Deralakatte, 575018 Mangaluru, India. [Giridhar, B. Hosmane] K S Hegde Medical Hospital, Department of Pulmonary Medicine, 575018 Mangaluru, India. [Vishwanath, Silpa] K S Hegde Medical Hospital, Department of Surgery, 575018 Mangaluru, India. [Chakraborty, Anirban] Nitte University Centre for Science Education and Research (NUCSER), Division of Molecular Genetics and Cancer, Kotekar-Beeri Road, Deralakatte, 575018 Mangaluru, India. RP Chakraborty, A (reprint author), Nitte University Centre for Science Education and Research (NUCSER), Division of Molecular Genetics and Cancer, 575018 Mangaluru, India. EM anirban@nitte.edu.in CR Chakraborty A, Uechi T, Kenmochi N, 2011, Guarding the ‘translation apparatus’: defective ribosome biogenesis and the p53 signaling pathway. WIREs RNA 2:507–522 Levine AJ, Momand J, Finlay CA, 1991, The p53 tumour suppressor gene. Nature 351:453–456 Zilfou J, Lowe S, 2009, Tumor suppressive functions of p53. CSH Perspectives a001883 Pecorino L, 2012, Text book of molecular biology of cancer. Oxford University Press, USA Alex S, Varda R, 2000, Oncogenic mutations of the p53 tumor suppressor: the demons of the guardian of the genome. Cancer Res 60:6788–6793 Hainaut P, Hollstein M, 2000, P53 and human cancer: the first ten thousand mutations. Adv Cancer Res 77:81–137 Matei C, Negura L, Liliac L, Negura A, Azoicai D, 2012, Validation of PCR-RFLP techniques for the evaluation of codon 72 of p53 and CYP1A1 gene’s polymorphisms in relation ovarian cancer in Romanian population. Romanian J Morphol Embryol 53(1):47–54 Chen P, Lin S, Wang C, Chen Y, Chen T, Chang J, 1993, BHotSpots^ mutation analysis of p53 gene in gastro intestinal cancers by amplification of naturally occurring and artificially created restriction sites. Clin Chem 39:2186–2191 Chittmittrapap S, Chieochansin T, Chaiteerakij R, Treeprasertsuk S, Klaikaew N, Tangkijvanich P, Komolmit P, Poovorawan Y, 2013, Prevalence of aflatoxin induced P53 mutation at codon 249, R249s, in hepatocellular carcinoma patients with and without hepatitis B surface antigen, HBsAg). Asian Pac J Cancer Prev 14(12):7675– 7679 Liming O, Chongtao G, Haizhen W, Suxia L, Huizhan Z, 2009, PCR- RFLP to detect codon 248 mutation in exon 7 of p53 tumor suppressor gene. Biochem Mol Biol Educ 37:106–109 Lane DP, Crawford LV, 1979, T antigen is bound to a host protein in SY40-transformed cells. Nature 278:261–263 Linzer DI, Levine AJ, 1979, Characterization of a 54K Dalton cellular SV40 tumor antigen present in SV40-transformed cells and uninfected embryonal carcinoma cells. Cell 17:43–52 Soussi T, 2000, p53 Antibodies in the Sera of Patients with Various Types of Cancer: A Review. Cancer 60:1777–1788 Nigro JM, Baker SJ, Preisinger AC, Jessup J,Hostetter R, Cleary K, Bigner SH,Davidson N, Baylin S, Devilees P, Glover T, Collins FS, Weston A, Modali R, Harris C, Vogelstein B, 1989, Mutations in the p53 gene occur in diverse human tumour types. Nature 342: 705–708 Agorastos T, Masouridou S, Lambropoulos AF, Chrisafi S, Miliaras D, Pantazis K, Constantinides TC, Kotsis A, Bontis I, 2004, p53 codon 72 polymorphism and correlation with ovarian and endometrial cancer in Greek women. Eur J Cancer Prev 13:277–280 Freed-Pastor W, Prives C, 2012, Mutant p53: one name, many proteins. Genes Dev 26:1268–1286 Brown CJ, Lain S, Verma CS, Fersht AR, Lane DP, 2009, Awakening guardian angels: drugging the p53 pathway. Nat Rev Cancer 9:862–873 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1373 EP 1377 DI 10.1007/s12253-018-0394-8 PG 5 ER PT J AU Sticz, T Molnar, A Danko, T Hujber, Z Petovari, G Nagy, N Vegso, Gy Kopper, L Sebestyen, A AF Sticz, Tamas Molnar, Anna Danko, Titanilla Hujber, Zoltan Petovari, Gabor Nagy, Noemi Vegso, Gyula Kopper, Laszlo Sebestyen, Anna TI The Effects of Different mTOR Inhibitors in EGFR Inhibitor Resistant Colon Carcinoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon carcinoma; EGFR inhibitor; mTOR inhibitor; Resistance ID Colon carcinoma; EGFR inhibitor; mTOR inhibitor; Resistance AB Several monoclonal antibodies and inhibitors targeting signalling pathways are being used in personalised medicine. Anti-EGFR antibodies seem to be effective, however, therapy resistance often occurs in colon carcinoma cases. mTOR inhibitors (mTORIs) could have a potential role in the breakthrough of therapy resistance. The mTOR activity related protein expression patterns and the in vitro effects of EGFR inhibitors (EGFRIs), mTORIs and their combinations were studied in different colon carcinoma cell lines (with different genetic backgrounds). Alamar Blue test and flow cytometry were used to analyse the in vitro proliferation and apoptotic effects of cetuximab, gefitinib, cisplatin, rapamycin, PP242 and NVP-BEZ235. The expressions of mTOR activity related proteins (p-70S6K, p-S6, Rictor, p-mTOR, Raptor) were studied by Western blot, immunocytochemistry and Duolink staining. The EGFRI resistance of the studied colon carcinoma cell lines related to their known mutations were confirmed, neither gefitinib nor cetuximab inhibited the proliferation or induced apoptosis in vitro. Individual differences in Rictor and Raptor expressions were detected by Western blot and immunocytochemistry beside elevated mTOR activity of these different colon carcinoma cell lines. These expression patterns correlated to the mTORIs sensitivity differences, moreover, mTORIs could enhance the effects of EGFRIs and other in vitro treatments. Our results suggest that mTORI combinations could be helpful in both EGFRI and platinum-based therapy of colon carcinomas. Moreover, we suggest determining both mTOR complex activity and mutations in Akt/mTOR signalling pathways for selecting the appropriate mTORIs and patients in potential future combination treatments. C1 [Sticz, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Molnar, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Danko, Titanilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Hujber, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Petovari, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Nagy, Noemi] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Vegso, Gyula] Semmelweis University, Department of Transplantation and Surgery, Ulloi ut 26, H-1085 Budapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. RP Sebestyen, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM hsebanna@gmail.com;anna@korb1.sote.hu CR Tsimberidou D, 2015, AM: targeted therapy in cancer. Cancer Chemother Pharmacol 76(6):1113–1132 Herzig DO, Tsikitis VL, 2015, Molecular markers for colon diagnosis, prognosis and targeted therapy. J Surg Oncol 111(1):96–102 Kalia M, 2015, Biomarkers for personalized oncology: recent advances and future challenges. Metabolism 64:S16–S21 Dietel M, Johrens K, Laffert MV, Hummel M, Blaker H, Pfitzner BM, Lehmann A, Denkert C, Darb-Esfahani S, Lenze D, Heppner FL, Koch A, Sers C, Klauschen F, Anagnostopoulos I, 2015, A 2015 update on predictive molecular pathology and its role in targeted cancer therapy: a review focusing on clinical relevance. Cancer Gene Ther 22(9):417–430. , DOI 10.1038/cgt.2015.39 Chong CR, Janne PA, 2013, The quest to overcome resistance to EGFR-targeted therapies in cancer. Nat Med 19(11):1389–1400 Khamisipour G, Jadidi-Niaragh F, Jahromi AS, Zandi K, Hojjat- Farsangi M, 2016, Mechanisms of tumor cell resistance to the current targeted-therapy agents. Tumour Biol:1–19 Zhao Y, Butler EB, Tan M, 2013, Targeting cellular metabolism to improve cancer therapeutics. Cell Death Dis 4(3):e532 Francipane MG, Lagasse E, 2014, mTOR pathway in colorectal cancer: an update. Oncotarget 5(1):49–66 Chiarini F, Evangelisti C, McCubrey JA, Martelli AM, 2015, Current treatment strategies for inhibiting mTOR in cancer. Trends Pharmacol Sci 36(2):124–135 FrancipaneMG, Lagasse E, 2015 Nov 26, Therapeutic potential of mTOR inhibitors for targeting cancer stem cells. Br J Clin Pharmacol 82:1180–1188. , DOI 10.1111/bcp.12844. Sticz T, Molnar A, Mark A, Hajdu M, Nagy N, Gy V, Micsik T, Kopper L, Sebestyen A, 2016, mTOR activity and its prognostic significance in human colorectal carcinoma depending on C1 and C2 complex related protein expression. Clin Pathol 70:410–416. , DOI 10.1136/jclinpath-2016-203913 Huang Z,Wu Y, Zhou X, Qian J, ZhuW, Shu Y, Liu P, 2015, Clinical efficacy of mTOR inhibitors in solid tumors: a systematic review. Future Oncol 11(11):1687–1699. , DOI 10.2217/fon.15.70 Lamming DW, 2016, Inhibition of the mechanistic target of rapamycin, mTOR)-rapamycin and beyond. Cold Spring Harb Perspect Med 6(5). , DOI 10.1101/cshperspect.a025924 Ahmed D, Eide PW, Eilertsen IA, Danielsen SA, Eknaes M, Hektoen M, Lind GE, Lothe RA, 2013 Sep 16, Epigenetic and genetic features of 24 colon cancer cell lines. Oncogene 2:e71. , DOI 10.1038/oncsis.2013.35 Roohi A, Hojjat-Farsangi M, 2016, Recent advances in targeting mTOR signaling pathway using small molecule inhibitors. J Drug Target 3:1–13 PubMedPMID: 27632356 Spindler KL, Sorensen MM, Pallisgaard N, Andersen RF, Havelund BM, Ploen J, Lassen U, Jakobsen AK, 2013, Phase II trial of temsirolimus alone and in combination with irinotecan for KRAS mutant metastatic colorectal cancer: outcome and results of KRAS mutational analysis in plasma. Acta Oncol 52(5):963–970 Harada K,Miyake H, KumanoM, FujisawaM(2013 [Epub ahead of print], Acquired resistance to temsirolimus in human renal cell carcinoma cells is mediated by the constitutive activation of signal transduction pathways through mTORC2. Br J Cancer 109:2389–2395 Sebestyen A, Mark A, Hajdu M, Nagy N, Molnar A, Vegso G, Barna G, Kopper L, 2015, Rapamycin can restore the negative regulatory function of transforming growth factor beta 1 in high grade lymphomas. Cytokine 73(2):219–224 Mark A, Hajdu M, Varadi Z, Sticz TB, Nagy N, Csomor J, Berczi L, Varga V, CsokaM, Kopper L, Sebestyen A, 2013, Characteristic mTOR activity in Hodgkin-lymphomas offers a potential therapeutic target in high risk disease–a combined tissue microarray, in vitro and in vivo study. BMC Cancer 13:250. , DOI 10.1186/ 1471-2407-13-250 Gao F, Huang C, Zhang Y, Sun R, Zhang Y, Wang H, Zhang S, 2016, Combination targeted therapy of VEGFR inhibitor, sorafenib, with an mTOR inhibitor, sirolimus induced a remakable response of rapid progressive Uterine PEComa. Cancer Biol Ther 17(6):595–598 Martin R, Desponds C, Eren RO, Quadroni M, Thome M, Fasel N, 2016, Caspase-mediated cleavage of raptor participates in the inactivation of mTORC1 during cell death. Cell Death Discov 2: 16024. , DOI 10.1038/cddiscovery.2016.24.eCollection Weigelt B, Warne PH, Downward J, 2011, PIK3CA mutation, but not PTEN loss of function, determines the sensitivity of breast cancer cells to mTOR inhibitor drugs. Oncogene 30(29):3222–3233 Wang Z, Martin D, Molinolo AA, Patel V, Iglesias-Bartolome R, Degese MS, Vitale-Cross L, Chen Q, Gutkind JS, 2014, mTOR co-targeting in cetuximab resistance in head and neck cancers harboring PIK3CA and RAS mutations. J Natl Cancer Inst 106(9). , DOI 10.1093/jnci/dju215 Li B, Gao S,Wei F, Bellail AC, Hao C, Liu T, 2012, Simultaneous targeting of EGFR and mTOR inhibits the growth of colorectal carcinoma cells. Oncol Rep 28(1):15–20 Leisching GR, Loos B, Botha MH, Engelbrecht AM, 2015, The role of mTOR during cisplatin treatment in an in vitro and ex vivo model of cervical cancer. Toxicology 335:72–78 Liu J, Zhang L, Zhang X, Xing X, 2015, Rapamycin enhanced the antitumor efficacy of oxaliplatin in cisplatin-resistant ovarian cancer cells A2780cis both in vitro and in vivo. J Chemother 27(6): 358–364. , DOI 10.1179/1973947815Y.0000000021 Costello BA, BoradMJ, QiY, KimGP, NorthfeltDW, Erlichman C, Alberts SR, 2014, Phase I trial of everolimus, gemcitabine and cisplatin in patients with solid tumors. Investig New Drugs 32(4): 710–716. , DOI 10.1007/s10637-014-0096-3 Im-Aram A, Farrand L, Bae SM, Song G, Song YS, Han JY, Tsang BK, 2013, ThemTORC2 component Rictor contributes to cisplatin resistance in human ovarian Cancer cells. PLoS One 8(9):e75455 Kim A, Lee JE, Lee SS, Kim C, Lee SJ, Jang WS, Park S, 2013, Coexistent mutations of KRAS and PIK3CA affect the efficacy of NVP-BEZ235, a dual PI3K/MTOR inhibitor, in regulating the PI3K/ MTOR pathway in colorectal cancer. Int J Cancer 133:984–996 Yang F, QianXJ,QinW, Deng R, Wu XQ,Qin J, FengGK, Zhu XF, 2013, Dual phosphoinositide 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 has a therapeutic potential and sensitizes cisplatin in nasopharyngeal carcinoma. PLoS One 8(3):e59879 Hecht JR, Reid TR, Garrett CR, Beck JT, Davidson SJ, Mackenzie MJ, Brandt U, Rizvi S, Sharma S, 2015, Phase I study of everolimus, cetuximab and irinotecan as second-line therapy in metastatic colorectal cancer. Anticancer Res 35(3):1567–1573 Severyn B, Nguyen T, Altman MD, Li L, Nagashima K, Naumov GN, Sathyanarayanan S, Cook E, Morris E, Ferrer M, Arthur B, Benita Y, Watters J, Loboda A, Hermes J, Gilliland DG, Cleary MA, Carroll PM, Strack P, Tudor M, Andersen JN, 2016, Development of a highthroughput gene expression screen for modulators of RAS-MAPK signaling in a mutant RAS cellular context. J Biomol Screen 21(9): 989–997. , DOI 10.1177/1087057116658646 Pohl M, Schmiegel W, 2016, Therapeutic strategies in diseases of the digestive tract- 2015 and beyond targeted therapies in Colon Cancer today and tomorrow. Dig Dis 34(5): 574–579. , DOI 10.1159/000445267 Kim ST, Kim SY, Klempner SJ, Yoon J, Kim N, Ahn S, Bang H, Kim KM, Park W, Park SH, Park JO, Park YS, Lim HY, Lee SH, Park K, Kang WK, Lee J, 2016, Rapamycininsensitive companion of mTOR, RICTOR, amplification defines a subset of advanced gastric Cancer and is sensitive to AZD2014-mediated mTORC1/2 inhibition. Ann Oncol: mdw669. , DOI 10.1093/annonc/mdw669 McCoach CE, Bivona TG, Blakely CM, Doebele RC, 2016, Neoadjuvant oncogene-targeted therapy in early stage non-smallcell lung Cancer as a strategy to improve clinical outcome and identify early mechanisms of resistance. Clin Lung Cancer 17(5): 466–469. , DOI 10.1016/j.cllc.2016.05.025 Chen DH, Zhang XS, 2015, Targeted therapy: resistance and resensitization. Chin J Cancer 34(11):496–501 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1379 EP 1386 DI 10.1007/s12253-018-0434-4 PG 8 ER PT J AU Ansari, H Shahrisa, A Tahmasebi Birgani, Y Tahmasebi Birgani, M Hajjari, M Mohammadi Asl, J AF Ansari, Hossein Shahrisa, Arman Tahmasebi Birgani, Yaser Tahmasebi Birgani, Maryam Hajjari, Mohammadreza Mohammadi Asl, Javad TI Long Noncoding RNAs in Colorectal Adenocarcinoma; an in silico Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer; Biomarker; Long noncoding RNAs; SNHG6; PVT1; ZFAS1 ID Cancer; Biomarker; Long noncoding RNAs; SNHG6; PVT1; ZFAS1 AB Long noncoding RNAs (lncRNAs) are lengthy noncoding transcripts which are involved in critical signaling pathways including cell cycle and apoptosis so it is not surprising to see their altered expression in human tumors. Colorectal adenocarcinoma is one the most frequent malignancies worldwide. The role of lncRNAs in colorectal adenocarcinoma is not well understood. To study the significance of lncRNAs in colorectal adenocarcinoma, we retrieved 189 approved lncRNAs from HGNC. The genes were imported into the cBioPortal database for transcriptomic analyses. We queried all the samples from TCGA provisional colorectal adenocarcinoma with RNA-seq v2 data in our study and considered RNA dysregulation with Z-score: ±2. The lncRNA which was altered in most of the patients were considered as "significant lncRNA" for further analyses. We considered the association of candidate lncRNAs with clinicopathologic parameters of samples including tumor disease anatomic site, neoplasm histologic types, tumor stage and survival. We also compute the specificity of the significant lncRNAs expression in colorectal adenocarcinoma comparing with other human cancers in cancer portal. Our analysis showed that lncRNAs SNHG6, PVT1 and ZFAS1 allocated the maximum alteration among the colorectal cases. The expression of SNHG6 and ZFAS1 was more in rectal adenocarcinoma than the colon carcinoma while the PVT1 showed the same expression levels in both tissues. However, we found that upregulation of PVT1 has been reduced the overall survival in patients. Altogether these data showed SNHG6, PVT1 and ZFAS1, are promising candidates for experimental research on colorectal adenocarcinoma to discover novel biomarker for this prevalent cancer. C1 [Ansari, Hossein] Islamic Azad University, Ahvaz Branch, Department of BiotechnologyAhvaz, Iran. [Shahrisa, Arman] Tarbiat Modares University, Faculty of Biosciences, Department of Molecular GeneticsTehran, Iran. [Tahmasebi Birgani, Yaser] Ahvaz Jundishapur University of Medical Sciences, School of Public Health, Department of Environmental Health EngineeringAhvaz, Iran. [Tahmasebi Birgani, Maryam] Ahvaz Jundishapur University of Medical Sciences, School of Medicine, Department of Medical GeneticsAhvaz, Iran. [Hajjari, Mohammadreza] Shahid Chamran University of Ahvaz, Faculty of Sciences, Department of GeneticsAhvaz, Iran. [Mohammadi Asl, Javad] Ahvaz Jundishapur University of Medical Sciences, School of Medicine, Department of Medical GeneticsAhvaz, Iran. RP Shahrisa, A (reprint author), Tarbiat Modares University, Faculty of Biosciences, Department of Molecular Genetics, Tehran, Iran. EM shahrisa.arman@hotmail.com CR Ge X, Chen Y, Liao X, Liu D, Li F, Ruan H, Jia W, 2013, Overexpression of long noncoding RNA PCAT-1 is a novel biomarker of poor prognosis in patients with colorectal cancer. Med Oncol 30:588. , DOI 10.1007/s12032-013-0588-6 Hrasovec S, Glavac D, 2012, MicroRNAs as Novel Biomarkers in Colorectal Cancer. Front Genet 3:180. , DOI 10.3389/ fgene.2012.00180 Cheetham SW, Gruhl F, Mattick JS, Dinger ME, 2013, Long noncoding RNAs and the genetics of cancer. Br J Cancer 108:2419– 2425. , DOI 10.1038/bjc.2013.233 Gibb EA, Brown CJ, Lam WL, 2011, The functional role of long non-coding RNA in human carcinomas. Mol Cancer 10:38. https:// doi.org/10.1186/1476-4598-10-38 Struhl K, 2007, Transcriptional noise and the fidelity of initiation by RNA polymerase II. Nat Struct Mol Biol 14:103–105. https:// doi.org/10.1038/nsmb0207-103 Gutschner T, Diederichs S, 2012, The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol 9:703–719. https://doi. org/10.4161/rna.20481 Huang Y, Liu N, Wang JP, Wang YQ, Yu XL, Wang ZB, Cheng XC, Zou Q, 2012, Regulatory long non-coding RNA and its functions. J Physiol Biochem 68:611–618. , DOI 10.1007/ s13105-012-0166-y Costa FF, 2010, Non-coding RNAs: Meet thy masters. Bioessays 32:599–608. , DOI 10.1002/bies.200900112 CabiliMN, Trapnell C, Goff L, KoziolM, Tazon-Vega B, Regev A, Rinn JL, 2011, Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 25:1915–1927. , DOI 10.1101/gad.17446611 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, Antipin Y, Reva B, Goldberg AP, Sander C, Schultz N, 2012, The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2:401–404. , DOI 10. 1158/2159-8290.CD-12-0095 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, Cerami E, Sander C, Schultz N, 2013, Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6:pl1. https:// doi.org/10.1126/scisignal.2004088 Shahrisa A, Tahmasebi BirmaniM(2017, cbaf:Multiple automated functions for cbioportal.org. R package version 1.0.2. https://doi. org/10.18129/B9.bioc.cbaf Petryszak R, Keays M, Tang YA, Fonseca NA, Barrera E, Burdett T, Fullgrabe A, Fuentes AM, Jupp S, Koskinen S, Mannion O, Huerta L, Megy K, Snow C, Williams E, Barzine M, Hastings E, Weisser H, Wright J, Jaiswal P, Huber W, Choudhary J, Parkinson HE, Brazma A, 2016, Expression Atlas update–an integrated database of gene and protein expression in humans, animals and plants. Nucleic Acids Res 44:D746–D752. , DOI 10.1093/nar/ gkv1045 Petryszak R, Burdett T, Fiorelli B, Fonseca NA, Gonzalez-Porta M, Hastings E, Huber W, Jupp S, Keays M, Kryvych N, McMurry J, Marioni JC, Malone J, Megy K, Rustici G, Tang AY, Taubert J, Williams E, Mannion O, Parkinson HE, Brazma A, 2014, Expression Atlas update–a database of gene and transcript expression from microarray- and sequencing-based functional genomics experiments. Nucleic Acids Res 42:D926–D932. , DOI 10. 1093/nar/gkt1270 Kapushesky M, Adamusiak T, Burdett T, Culhane A, Farne A, Filippov A, Holloway E, Klebanov A, Kryvych N, Kurbatova N, Kurnosov P, Malone J, Melnichuk O, Petryszak R, Pultsin N, Rustici G, Tikhonov A, Travillian RS, Williams E, Zorin A, Parkinson H, Brazma A, 2012, Gene Expression Atlas update–a value-added database of microarray and sequencing-based functional genomics experiments. Nucleic Acids Res 40:D1077– D1081. , DOI 10.1093/nar/gkr913 Consortium GT, 2015, Human genomics. The Genotype-Tissue Expression, GTEx, pilot analysis: multitissue gene regulation in humans. Science 348:648–660. , DOI 10.1126/science. 1262110 Yuan J,WuW, Xie C, Zhao G, Zhao Y, Chen R, 2013, NPInter v2. 0: an updated database of ncRNA interactions. Nucleic Acids Res 42:D104–D108 Schmitt AM, Chang HY, 2016, Long Noncoding RNAs in Cancer Pathways. Cancer Cell 29:452–463. , DOI 10.1016/j.ccell. 2016.03.010 Thorenoor N, Faltejskova-Vychytilova P, Hombach S, Mlcochova J, Kretz M, Svoboda M, Slaby O, 2016, Long non-coding RNA ZFAS1 interacts with CDK1 and is involved in p53-dependent cell cycle control and apoptosis in colorectal cancer. Oncotarget 7:622– 637. , DOI 10.18632/oncotarget.5807 Wang W, Xing C, 2016, Upregulation of long noncoding RNA ZFAS1 predicts poor prognosis and prompts invasion and metastasis in colorectal cancer. Pathol Res Pract 212:690–695. https://doi. org/10.1016/j.prp.2016.05.003 Picelli S, Vandrovcova J, Jones S, Djureinovic T, Skoglund J, Zhou XL, Velculescu VE, Vogelstein B, Lindblom A, 2008, Genomewide linkage scan for colorectal cancer susceptibility genes supports linkage to chromosome 3q. BMC Cancer 8:87. https://doi. org/10.1186/1471-2407-8-87 Kemp Z, Carvajal-Carmona L, Spain S, Barclay E, Gorman M, Martin L, Jaeger E, Brooks N, Bishop DT, Thomas H, Tomlinson I, Papaemmanuil E, Webb E, Sellick GS, Wood W, Evans G, Lucassen A, Maher ER, Houlston RS, ColoRectal tumour Gene Identification Study C, 2006, Evidence for a colorectal cancer susceptibility locus on chromosome 3q21-q24 from a high-density SNP genome-wide linkage scan. Hum Mol Genet 15:2903–2910. , DOI 10.1093/hmg/ddl231 Guo K, Yao J, Yu Q, Li Z, Huang H, Cheng J, Wang Z, Zhu Y, 2017, The expression pattern of long non-coding RNA PVT1 in tumor tissues and in extracellular vesicles of colorectal cancer correlates wi t h cancer progression. Tumour Biol 39: 1010428317699122. , DOI 10.1177/1010428317699122 Kawai M, Komiyama H, Hosoya M, Okubo H, Fujii T, Yokoyama N, Sato C, Ueyama T, Okuzawa A, Goto M, Kojima Y, Takahashi M, Sugimoto K, Ishiyama S, Munakata S, Ogura D, Niwa SI, Tomiki Y, Ochiai T, Sakamoto K, 2016, Impact of chromosome 17q deletion in the primary lesion of colorectal cancer on liver metastasis. Oncol Lett 12:4773–4778. , DOI 10.3892/ol. 2016.5271 Chen Y, Fang L, Zhang J, Li G, Ma M, Li C, Lyu J, Meng QH, 2017, Blockage of Glyoxalase I Inhibits Colorectal Tumorigenesis and Tumor Growth via Upregulation of STAT1, p53, and Bax and Downregulation of c-Myc and Bcl-2. Int J Mol Sci 18:570 Wang W, Deng J, Wang Q, Yao Q, Chen W, Tan Y, Ge Z, Zhou J, Zhou Y, 2017, Synergistic role of Cul1 and c-Myc: Prognostic and predictive biomarkers in colorectal cancer. Oncol Rep 38(1):245– 252. , DOI 10.3892/or.2017.5671 Rodrigues NR, Rowan A, Smith M, Kerr IB, BodmerWF, Gannon JV, Lane DP, 1990, p53 mutations in colorectal cancer. Proc Natl Acad Sci 87:7555–7559 Hao H, Liu L, Zhang D, Wang C, Xia G, Zhong F, Hu X, 2017, Diagnostic and prognostic value of miR-106a in colorectal cancer. Oncotarget 8:5038 Xiao G, Tang H,WeiW, Li J, Ji L, Ge J, 2014, Aberrant expression of microRNA-15a and microRNA-16 synergistically associates with tumor progression and prognosis in patients with colorectal cancer. Gastroenterol Res Pract. , DOI 10.1155/2014/ 364549 Chen H-Y, Lin Y-M, Chung H-C, Lang Y-D, Lin C-J, Huang J, Wang W-C, Lin F-M, Chen Z, Huang H-D, 2012, miR-103/ 107 promote metastasis of colorectal cancer by targeting the metastasis suppressors DAPK and KLF4. Cancer Res 72: 3631–3641 Birgani MT, Hajjari M, Shahrisa A, Khoshnevisan A, Shoja Z, Motahari P, Farhangi B, 2017, Long Non-Coding RNA SNHG6 as a Potential Biomarker for Hepatocellular Carcinoma. Pathol Oncol Res. , DOI 10.1007/s12253-017-0241-3 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1387 EP 1394 DI 10.1007/s12253-018-0428-2 PG 8 ER PT J AU Ghali, MR Mahjoub, S Zaied, S Bhiri, H Bahia, W Mahjoub, T Almawi, YW AF Ghali, M Rabeb Mahjoub, Sana Zaied, Sonia Bhiri, Hanen Bahia, Wael Mahjoub, Touhami Almawi, Y Wassim TI Association of Genetic Variants in NF-kB with Susceptibility to Breast Cancer: a Case Control Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Allele; Breast cancer; Haplotype; Metastasis; NF-κB ID Allele; Breast cancer; Haplotype; Metastasis; NF-κB AB Insofar as altered NF-κB signaling stemming from the presence of specific genetic variants in NF-κB gene contribute to cancer pathogenesis, this study evaluated the association between NF-κB rs147574894/I552V, rs148626207/M860T rs3774937 and rs1598859 variants and breast cancer and associated features and complications. This was a retrospective case-control study, which involved 207 women with breast cancer, and 214 cancer-free women who served as controls. NF-κB genotyping was done by real-time PCR. Significantly higher rs3774937 minor allele frequencies (MAF), and lower rs147574894 MAF were seen among breast cancer patients, thereby imparting disease susceptibility and protective nature to these variants, respectively. Significant association of rs3774937 and rs147574894 genotypes with breast cancer was seen under the dominant model. Histological type and grade, molecular type, Her2 positivity and ER+/Her2- correlated positively, while distant metastasis negatively correlated with rs3774937. On the other hand, rs147574894 negatively correlated with histological type and grade, tumor size, Her2 positivity, molecular type, and ER+/Her2-, while rs148626207 correlated positively with histological grade, but negatively with distant metastasis and triple-negative status. Breast cancer-susceptible and –protective 4-locus haplotypes were also identified. This is the first report that addresses the contribution of NF-κB variants to the pathogenesis of breast cancer in Middle Eastern-North African population, and the first to document positive association of rs3774937 with breast cancer. C1 [Ghali, M Rabeb] University of Monastir, Faculty of Pharmacy of Monastir, Laboratory of Human Genome and Multifactorial Diseases (LR12ES07)Monastir, Tunisia. [Mahjoub, Sana] University of Monastir, Faculty of Pharmacy of Monastir, Laboratory of Human Genome and Multifactorial Diseases (LR12ES07)Monastir, Tunisia. [Zaied, Sonia] CHU Fattouma Bourguiba, Department of Clinical OncologyMonastir, Tunisia. [Bhiri, Hanen] CHU Fattouma Bourguiba, Department of Clinical OncologyMonastir, Tunisia. [Bahia, Wael] University of Monastir, Faculty of Pharmacy of Monastir, Laboratory of Human Genome and Multifactorial Diseases (LR12ES07)Monastir, Tunisia. [Mahjoub, Touhami] University of Monastir, Faculty of Pharmacy of Monastir, Laboratory of Human Genome and Multifactorial Diseases (LR12ES07)Monastir, Tunisia. [Almawi, Y Wassim] El Manar University, Faculty of Sciences of TunisTunis, Tunisia. RP Almawi, YW (reprint author), El Manar University, Faculty of Sciences of Tunis, Tunis, Tunisia. EM wassim.almawi@outlook.com CR Salim EI, Moore MA, Al-Lawati JA et al, 2009, Cancer epidemiology and control in the Arab world - past, present and future.Asian Pac J Cancer Prev 10:3–16 Abdulrahman GO Jr, Rahman GA, 2012, Epidemiology of breast cancer in Europe and Africa. J Cancer Epidemiol 2012:915610 Perou CM, Sorlie T, Eisen MB et al, 2000, Molecular portraits of human breast tumours. Nature 406:747–752 Purdie CA, Baker L, Ashfield A et al, 2010, Increased mortality in HER2 positive, oestrogen receptor positive invasive breast cancer: a population-based study. Br J Cancer 103:475–481 Nyante SJ, Gammon MD, Kaufman JS et al, 2015, Genetic variation in estrogen and progesterone pathway genes and breast cancer risk: an exploration of tumor subtype-specific effects. Cancer Causes Control 26:121–131 Journe F, Durbecq V, Chaboteaux C et al, 2009, Association between farnesoid X receptor expression and cell proliferation in estrogen receptor-positive luminal-like breast cancer from postmenopausal patients. Breast Cancer Res Treat 115:523–535 Mantovani A, Garlanda C, Allavena P, 2010, Molecular pathways and targets in cancer-related inflammation. Ann Med 42:161–170 Murray JL, Thompson P, Yoo SY et al, 2013, Prognostic value of single nucleotide polymorphisms of candidate genes associated with inflammation in early stage breast cancer. Breast Cancer Res Treat 138:917–924 Wang Z, Liu QL, Sun W, Yang CJ, Tang L, Zhang X, Zhong XM, 2014, Genetic polymorphisms in inflammatory response genes and their associations with breast cancer risk. Croat Med J 55: 638–646 Teschendorff AE, Miremadi A, Pinder SE, Ellis IO, Caldas C, 2007, An immune response gene expression module identifies a good prognosis subtype in estrogen receptor negative breast cancer. Genome Biol 8:R157 Hayden MS, Ghosh S, 2012, NF-κB, the first quarter-century: remarkable progress and outstanding questions. Genes Dev 26:203– 234 NelsonDE, Ihekwaba AE, ElliottMet al, 2004, Oscillations in NFkappaB signaling control the dynamics of gene expression. Science 306:704–708 Oeckinghaus A, Ghosh S, 2009, The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb Perspect Biol 1:a000034 Vallabhapurapu S, KarinM, 2009, Regulation and function of NFkappaB transcription factors in the immune system. Annu Rev Immunol 27:693–733 DiDonato JA, Mercurio F, KarinM(2012, NF-kappaB and the link between inflammation and cancer. Immunol Rev 246:379–400 Beinke S, Ley SC, 2004, Functions of NF-kappaB1 and NFkappaB2 in immune cell biology. Biochem J 382:393–409 Nakshatri H, Bhat-Nakshatri P, Martin DA, Goulet RJ Jr, Sledge GW Jr, 1997, Constitutive activation of NF-kappaB during progression of breast cancer to hormone-independent growth. Mol Cell Biol 17:3629–3639 Curran JE,Weinstein SR, Griffiths LR, 2002, Polymorphic variants of NFKB1 and its inhibitory protein NFKBIA, and their involvement in sporadic breast cancer. Cancer Lett 188:103–107 Shojaie N, Ghaffari SM, 2016, Simultaneous analysis of Wnt and NF-κB signaling pathways in doxorubicin sensitive and methotrexate resistant PLC/ PRF/5 cells. Cell J 17:730–739 Jiao X,Wood LD, LindmanMet al, 2012, Somatic mutations in the notch, NF-KB, PIK3CA, and hedgehog pathways in human breast cancers. Genes Chromosomes Cancer 51:480–489 Brantley DM, Chen CL, Muraoka RS et al, 2001, Nuclear factorkappaB, NF-kappaB, regulates proliferation and branching in mouse mammary epithelium. Mol Biol Cell 12:1445–1455 Cao YX, Bonizzi G, Seagroves TN, Greten FR, Johnson R, Schmidt EV, Karin M, 2001, IKK alpha provides an essential link between RANK signaling and cyclin D1 expression during mammary gland development. Cell 107:763–775 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Resler AJ, Malone KE, Johnson LG, Malkki M, Petersdorf EW, McKnight B, Madeleine MM, 2013, Genetic variation in TLR or NFkappaB pathways and the risk of breast cancer: a case-control study. BMC Cancer 13:219 Jamshidi M, Fagerholm R, Khan S et al, 2015, SNP-SNP interaction analysis of NF-kappaB signaling pathway on breast cancer survival. Oncotarget 6:37979–37994 Alasmael N, Mohan R, Meira LB, Swales KE, Plant NJ, 2016, Activation of the Farnesoid X-receptor in breast cancer cell lines results in cytotoxicity but not increased migration potential. Cancer Lett 370:250–259 Giordano C, Barone I, Vircillo V et al, 2016, Activated FXR inhibits leptin signaling and counteracts tumor-promoting activities of cancer-associated fibroblasts in breast malignancy. Sci Rep 6:21782 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1395 EP 1400 DI 10.1007/s12253-018-0452-2 PG 6 ER PT J AU Evans, M O’Sullivan, B Smith, M Hughes, F Mullis, T Trim, N Taniere, P AF Evans, Matthew O’Sullivan, Brendan Smith, Matthew Hughes, Frances Mullis, Tina Trim, Nicola Taniere, Philippe TI Large-Scale EGFR Mutation Testing in Clinical Practice: Analysis of a Series of 18,920 Non-Small Cell Lung Cancer Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR; Lung cancer; Molecular pathology; TKI ID EGFR; Lung cancer; Molecular pathology; TKI AB We make use of a very large dataset of non-small cell lung cancer specimens to examine the molecular epidemiology of EGFR mutations, particularly with respect to rare and compound mutations, and to non-adenocarcinoma histological subtypes. We also demonstrate the feasibility of large-scale EGFR mutation screening using the full range of specimens encountered in routine practice. We retrospectively reviewed 18,920 unselected EGFR mutation results from our centre between July 2009 and October 2016, using Qiagen’s therascreen EGFR RGQ PCR Kit. Mutation rates were correlated with patient demographics and tumour histology. Our testing success rate was 93.9%, with similar success rates using histological and cytological specimens. Rare, potentially-targetable mutations accounted for 9.5% of all mutations detected. We identified a 2.5% mutation rate in tumours diagnosed as squamous cell carcinomas. There was a trend towards increasing EGFR mutation rates with increasing age, and while Del19 was the commonest mutation in the young, L858R predominated in the elderly. We found that EGFR mutation heterogeneity is rare within tumours and between primary and metastatic deposits. Our data demonstrate that large-scale, reflex EGFR mutation testing is feasible and affordable in the context of a publicly-funded health system. Furthermore, we have shown that the use of techniques sensitive only to classical mutations and selection of patients on the grounds of age, sex and histology denies patients access to potentially beneficial TKI therapy. C1 [Evans, Matthew] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2GW Birmingham, UK. [O’Sullivan, Brendan] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2GW Birmingham, UK. [Smith, Matthew] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2GW Birmingham, UK. [Hughes, Frances] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2GW Birmingham, UK. [Mullis, Tina] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2GW Birmingham, UK. [Trim, Nicola] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2GW Birmingham, UK. [Taniere, Philippe] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2GW Birmingham, UK. RP Evans, M (reprint author), Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, B15 2GW Birmingham, UK. EM matthew.evans7@nhs.net CR Cancer Genome Atlas Research N, 2012, Comprehensive genomic characterization of squamous cell lung cancers. Nature 489(7417): 519–525. , DOI 10.1038/nature11404 Marchetti A, Martella C, Felicioni L, Barassi F, Salvatore S, Chella A, Camplese PP, Iarussi T, Mucilli F, Mezzetti A, Cuccurullo F, Sacco R, Buttitta F, 2005, EGFR mutations in non-small-cell lung cancer: analysis of a large series of cases and development of a rapid and sensitive method for diagnostic screening with potential implications on pharmacologic treatment. J Clin Oncol 23(4):857– 865. , DOI 10.1200/JCO.2005.08.043 Dearden S, Stevens J, Wu YL, Blowers D, 2013, Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology, mutMap). Ann Oncol 24(9):2371– 2376. , DOI 10.1093/annonc/mdt205 Hata A, Katakami N, Yoshioka H, 2014, How sensitive are epidermal growth factor receptor-tyrosine kinase inhibitors for squamous cell carcinoma of the lung harboring EGFR gene-sensitive mutations? J Thorac Oncol 9(2):e20. , DOI 10.1097/JTO. 0000000000000098 Liu Y, Zhang Y, Zhang L, Liu B, Zhou X, Li Y, Zhou L, Gong Y, Wang Y, Lu Y, 2016, 167P: efficacy of EGFR-TKIs for lung squamous carcinomas harboring EGFR mutation: a multicenter study and pooled analysis of published reports. J Thorac Oncol 11(4 Suppl):S130. , DOI 10.1016/S1556-0864(16)30277-5 Shukuya T, Takahashi T, Kaira R, Ono A, Nakamura Y, Tsuya A, Kenmotsu H, Naito T, Kaira K, Murakami H, Endo M, Takahashi K, Yamamoto N, 2011, Efficacy of gefitinib for nonadenocarcinoma non-small-cell lung cancer patients harboring epidermal growth factor receptor mutations: a pooled analysis of published reports. Cancer Sci 102(5):1032–1037. , DOI 10. 1111/j.1349-7006.2011.01887.x Jia XL, Chen G, 2011, EGFR and KRAS mutations in Chinese patients with adenosquamous carcinoma of the lung. Lung Cancer 74(3):396–400. , DOI 10.1016/j.lungcan.2011.04.005 Tochigi N, Dacic S, Nikiforova M, Cieply KM, Yousem SA, 2011, Adenosquamous carcinoma of the lung: a microdissection study of KRAS and EGFR mutational and amplification status in a western patient population. Am J Clin Pathol 135(5):783–789. https://doi. org/10.1309/AJCP08IQZAOGYLFL Baik CS, Pritchard CC, Eaton KD, Chow LQ, 2013, EGFR mutations in squamous cell lung cancer in never-smokers. J Thorac Oncol 8(1):e6–e7. , DOI 10.1097/JTO. 0b013e3182762d49 Paik PK, Varghese AM, Sima CS, Moreira AL, Ladanyi M, Kris MG, Rekhtman N, 2012, Response to erlotinib in patients with EGFR mutant advanced non-small cell lung cancers with a squamous or squamous-like component. Mol Cancer Ther 11(11):2535– 2540. , DOI 10.1158/1535-7163.MCT-12-0163 Hata A, Katakami N, Fujita S, Kaji R, Imai Y, Takahashi Y, Nishimura T, Tomii K, Ishihara K, 2010, Frequency of EGFR and KRAS mutations in Japanese patients with lung adenocarcinoma with features of the mucinous subtype of bronchioloalveolar carcinoma. J Thorac Oncol 5(8):1197–1200. , DOI 10. 1097/JTO.0b013e3181e2a2bc Kadota K, Yeh YC, D'Angelo SP, Moreira AL, Kuk D, Sima CS, Riely GJ, Arcila ME, Kris MG, Rusch VW, Adusumilli PS, Travis WD, 2014, Associations between mutations and histologic patterns of mucin in lung adenocarcinoma: invasive mucinous pattern and extracellular mucin are associated with KRAS mutation. Am J Surg Pathol 38(8):1118–1127. , DOI 10.1097/PAS. 0000000000000246 Tsuta K, Kawago M, Inoue E, Yoshida A, Takahashi F, Sakurai H, Watanabe S, Takeuchi M, Furuta K, Asamura H, Tsuda H, 2013, The utility of the proposed IASLC/ATS/ERS lung adenocarcinoma subtypes for disease prognosis and correlation of driver gene alterations. Lung Cancer 81(3):371–376. , DOI 10.1016/j. lungcan.2013.06.012 Bell DW, Lynch TJ, Haserlat SM, Harris PL, Okimoto RA, Brannigan BW, Sgroi DC, Muir B, Riemenschneider MJ, Iacona RB, Krebs AD, JohnsonDH, Giaccone G, Herbst RS,Manegold C, Fukuoka M, Kris MG, Baselga J, Ochs JS, Haber DA, 2005, Epidermal growth factor receptor mutations and gene amplification in non-small-cell lung cancer: molecular analysis of the IDEAL/ INTACT gefitinib trials. J Clin Oncol 23(31):8081–8092. https:// doi.org/10.1200/JCO.2005.02.7078 Girard N, Sima CS, Jackman DM, Sequist LV, Chen H, Yang JC, Ji H, Waltman B, Rosell R, Taron M, Zakowski MF, Ladanyi M, Riely G, Pao W, 2012, Nomogram to predict the presence of EGFR activating mutation in lung adenocarcinoma. Eur Respir J 39(2):366–372. , DOI 10.1183/09031936.00010111 Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, Majem M, Lopez-Vivanco G, Isla D, Provencio M, Insa A, Massuti B, Gonzalez-Larriba JL, Paz-Ares L, Bover I, Garcia-Campelo R, Moreno MA, Catot S, Rolfo C, Reguart N, Palmero R, Sanchez JM, Bastus R, Mayo C, Bertran- Alamillo J, Molina MA, Sanchez JJ, Taron M, Spanish Lung Cancer G, 2009, Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med 361(10):958–967. , DOI 10.1056/NEJMoa0904554 Vallee A, Sagan C, Le Loupp AG, Bach K, Dejoie T, Denis MG, 2013, Detection of EGFR gene mutations in non-small cell lung cancer: lessons from a single-institution routine analysis of 1,403 tumor samples. Int J Oncol 43(4):1045–1051. , DOI 10. 3892/ijo.2013.2056 Sugiyama E, Goto K, Ishii G, Umemura S, Yoh K, Niho S, Ohmatsu H, Nagai K, Ohe Y, 2012, Higher incidence of EGFR exon 19 deletion in younger, age 40 or younger, patients with adenocarcinoma of the lung. J Clin Oncol 30(15_suppl):7044– 7044. , DOI 10.1200/jco.2012.30.15_suppl.7044 Lee B, Lee T, Lee SH, Choi YL, Han J, 2016, Clinicopathologic characteristics of EGFR, KRAS, and ALK alterations in 6,595 lung cancers. Oncotarget 7(17):23874–23884. , DOI 10.18632/ oncotarget.8074 Yang JC, Wu YL, Schuler M, Sebastian M, Popat S, Yamamoto N, Zhou C, Hu CP, O'Byrne K, Feng J, Lu S, Huang Y, Geater SL, Lee KY, Tsai CM, Gorbunova V, Hirsh V, Bennouna J, Orlov S, Mok T, Boyer M, Su WC, Lee KH, Kato T, Massey D, Shahidi M, Zazulina V, Sequist LV, 2015, Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma, LUX-Lung 3 and LUX-lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol 16(2):141–151. , DOI 10.1016/S1470-2045(14)71173-8 Ding P, Galla R, Inoue A, Lord S, Pavlakis N, Mitsudomi T, Links M, Gebski V, Yang J, Lee C, 2013, Exon 19 deletions, smoking history and gender as additional predictive factors for treatment benefit with EGFR tyrosine kinase inhibitors in patients harbouring activating EGFR mutations: a meta-analysis of 1432 patients in six randomised trials. J Thorac Oncol 8(11):S593–S594 Wang H, Huang J, Yu X, Han S, Yan X, Sun S, Zhu X, 2014, Different efficacy of EGFR tyrosine kinase inhibitors and prognosis in patients with subtypes of EGFR-mutated advanced non-small cell lung cancer: a meta-analysis. J Cancer Res Clin Oncol 140(11):1901–1909. , DOI 10.1007/s00432-014-1709-0 Jackman DM, Yeap BY, Lindeman NI, Fidias P, Rabin MS, Temel J, Skarin AT, Meyerson M, Holmes AJ, Borras AM, Freidlin B, Ostler PA, Lucca J, Lynch TJ, Johnson BE, Janne PA, 2007, Phase II clinical trial of chemotherapy-naive patients > or = 70 years of age treated with erlotinib for advanced non-small-cell lung cancer. J Clin Oncol 25(7):760–766. , DOI 10.1200/JCO. 2006.07.5754 Nishii T, Yokose T, Miyagi Y, Daigo Y, Ito H, Isaka T, Imai K, Murakami S, Kondo T, Saito H, Oshita F, Yamada K, Matsukuma S, Tsuboi M, Nakayama H, Masuda M, 2014, Clinicopathological features and EGFR gene mutation status in elderly patients with resected non-small-cell lung cancer. BMC Cancer 14:610. https:// doi.org/10.1186/1471-2407-14-610 Zhou C,Wu YL, Chen G, Feng J, LiuXQ,Wang C, Zhang S,Wang J, Zhou S, Ren S, Lu S, Zhang L, Hu C, Hu C, Luo Y, Chen L, Ye M, Huang J, Zhi X, Zhang Y, Xiu Q, Ma J, Zhang L, You C, 2011, Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer, OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 12(8):735–742. https:// doi.org/10.1016/S1470-2045(11)70184-X Yatabe Y, Matsuo K, Mitsudomi T, 2011, Heterogeneous distribution of EGFRmutations is extremely rare in lung adenocarcinoma. J Clin Oncol 29(22):2972–2977. , DOI 10.1200/JCO.2010. 33.3906 Zito Marino F, Liguori G, Aquino G, La Mantia E, Bosari S, Ferrero S, Rosso L, Gaudioso G, De Rosa N, Scrima M, Martucci N, La Rocca A, Normanno N, Morabito A, Rocco G, Botti G, Franco R, 2015, Intratumor heterogeneity of ALKrearrangements and homogeneity of EGFR-mutations in mixed lung adenocarcinoma. PLoS One 10(9):e0139264. https://doi. org/10.1371/journal.pone.0139264 Hsu KH, Chen KC, Yang TY, Yeh YC, Chou TY, Chen HY, Tsai CR, Chen CY, Hsu CP, Hsia JY, Chuang CY, Tsai YH, Chen KY, Huang MS, Su WC, Chen YM, Hsiung CA, Chang GC, Chen CJ, Yang PC, 2011, Epidermal growth factor receptor mutation status in stage I lung adenocarcinoma with different image patterns. J Thorac Oncol 6(6):1066–1072. , DOI 10.1097/JTO. 0b013e31821667b0 Taniguchi K, Okami J, Kodama K, HigashiyamaM, Kato K, 2008, Intratumor heterogeneity of epidermal growth factor receptor mutations in lung cancer and its correlation to the response to gefitinib. Cancer Sci 99(5):929–935. , DOI 10.1111/j.1349-7006. 2008.00782.x NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1401 EP 1409 DI 10.1007/s12253-018-0460-2 PG 9 ER PT J AU Cui, M Chang, Y Fang, QG Du, W Wu, JF Wang, JH Liu, ShT Luo, SX AF Cui, Meng Chang, Yu Fang, Qi-Gen Du, Wei Wu, Jun-Fu Wang, Ji-Heng Liu, Shan-Ting Luo, Su-Xia TI Non-Coding RNA Pvt1 Promotes Cancer Stem Cell–Like Traits in Nasopharyngeal Cancer via Inhibiting miR-1207 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PVT1; Nasopharyngeal carcinoma; miR-1207; PI3K/AKT; Tumor stemcell ID PVT1; Nasopharyngeal carcinoma; miR-1207; PI3K/AKT; Tumor stemcell AB Nasopharyngeal carcinoma (NPC) is a kind of head-neck malignant tumor. lncRNA-PVT1 can promote the proliferation of carcinoma cells, and induce cells to have stem cell-like potentials. However, the function of PVT1 in NPC cells is not clear. The expressions of lncRNA-PVT1 and the expressions of the stem cell markers in NPC tissues or cell lines were investigated by qRTPCR or western blot. The cell proliferation, and the ability of NPC cells to form spherical, clonal colonies were investigated by MTT assay, colony formation assay, and tumor-sphere formation assay. Cancer stem cells surface markers were detected by flow cytometry and western blot. PI3K/AKT signal activation in NPC cells was determined by western blot. PVT1 was significantly up-regulated in both NPC tissues and cell lines and associated with poor prognosis. PVT1 knockdown reduced NPC cells viability, clonogenicity, the cell surface CD44+/CD24− stem phenotype, and the expressions of the stem cell markers in NPC cells, including Oct4, c-Myc, SOX2, and ALDH. Furthermore, PVT1 negatively regulates the expression levels of miR-1207 in NPC cells and spheres cells, which is critical for NPC stemness. Knockdown of miR-1207 promoted stem phenotype and the expressions of the stem cell markers in NPC cells. Moreover, phosphor-PI3K (p-PI3K) and phosphor-AKT (p-AKT) were found to be down-regulated after PVT1 siRNAs transfection in NPC cells. And miR-1207 inhibitor transfection reversed the all the effects brought by PVT1 knockdown. Pvt1 promotes cancer stem cell–like properties in NPC cells via inhibiting miR-1207 and activating the PI3K/AKT signal pathway. C1 [Cui, Meng] Affiliated Cancer Hospital of Zhengzhou University, Department of Head & Neck and Thyroid, 450008 Zhengzhou, China. [Chang, Yu] First Affiliated Hospital of Zhengzhou University, Department of Oncology, 450008 Zhengzhou, China. [Fang, Qi-Gen] Affiliated Cancer Hospital of Zhengzhou University, Department of Head & Neck and Thyroid, 450008 Zhengzhou, China. [Du, Wei] Affiliated Cancer Hospital of Zhengzhou University, Department of Head & Neck and Thyroid, 450008 Zhengzhou, China. [Wu, Jun-Fu] Affiliated Cancer Hospital of Zhengzhou University, Department of Head & Neck and Thyroid, 450008 Zhengzhou, China. [Wang, Ji-Heng] Affiliated Cancer Hospital of Zhengzhou University, Department of Head & Neck and Thyroid, 450008 Zhengzhou, China. [Liu, Shan-Ting] Affiliated Cancer Hospital of Zhengzhou University, Department of Head & Neck and Thyroid, 450008 Zhengzhou, China. [Luo, Su-Xia] Affiliated Cancer Hospital of Zhengzhou University, Department of Medical Oncology, 450008 Zhengzhou, China. RP Luo, SX (reprint author), Affiliated Cancer Hospital of Zhengzhou University, Department of Medical Oncology, 450008 Zhengzhou, China. EM luosuxia8275@sina.com CR Chou J et al, 2008, Nasopharyngeal carcinoma–review of the molecular mechanisms of tumorigenesis. Head Neck 30(7):946–963 Xu T et al, 2013, Recurrent nasopharyngeal carcinoma: a clinical dilemma and challenge. Curr Oncol 20(5):e406–e419 Albini A et al, 2015, Cancer stem cells and the tumor microenvironment: interplay in tumor heterogeneity. Connect Tissue Res 56(5):414–425 Baccelli I, Trumpp A, 2012, The evolving concept of cancer and metastasis stem cells. J Cell Biol 198(3):281–293 Visvader JE, Lindeman GJ, 2008, Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 8(10):755–768 Han Yet al, 2015, Advanced applications of RNA sequencing and challenges. Bioinform Biol Insights 9(Suppl 1):29–46 Cech TR, Steitz JA, 2014, The noncoding RNA revolutiontrashing old rules to forge new ones. Cell 157(1):77–94 Huang T et al, 2013, Noncoding RNAs in cancer and cancer stem cells. Chin J Cancer 32(11):582–593 HuarteM(2015, The emerging role of lncRNAs in cancer. Nat Med 21(11):1253–1261 Schmitt AM, Chang HY, 2016, Long noncoding RNAs in Cancer pathways. Cancer Cell 29(4):452–463 Wang Y et al, 2015, A novel long non-coding RNA, hypoxiainducible factor-2alpha promoter upstream transcript, functions as an inhibitor of osteosarcoma stem cells in vitro. Mol Med Rep 11(4):2534–2540 Zhou M et al, 2016, LncRNA-Hh strengthen Cancer stem cells generation in twist-positive breast Cancer via activation of hedgehog signaling pathway. Stem Cells 34(1):55–66 Colombo T et al, 2015, PVT1: a rising star among oncogenic long noncoding RNAs. Biomed Res Int 2015:304208 GrahamM, Adams JM(1986, Chromosome 8 breakpoint far 3′ of the c-myc oncogene in a Burkitt's lymphoma 2;8 variant translocation is equivalent to the murine pvt-1 locus. EMBO J 5(11):2845–2851 Iaccarino I, 2017, lncRNAs and MYC: An Intricate Relationship. Int J Mol Sci 18(7):1497 Tseng YY et al, 2014, PVT1 dependence in cancer with MYC copy-number increase. Nature 512(7512):82–86 Huppi K et al, 2008, The identification of microRNAs in a genomically unstable region of human chromosome 8q24. Mol Cancer Res 6(2):212–221 Cui M et al, 2016, Long non-coding RNA PVT1 and cancer. Biochem Biophys Res Commun 471(1):10–14 Conte F et al, 2017, Role of the long non-coding RNA PVT1 in the dysregulation of the ceRNA-ceRNA network in human breast cancer. PLoS One 12(2):e0171661 Cui D et al, 2016, Long non-coding RNA PVT1 as a novel biomarker for diagnosis and prognosis of non-small cell lung cancer. Tumour Biol 37(3):4127–4134 Ding J et al, 2014, Expression and clinical significance of the long non-coding RNA PVT1 in human gastric cancer. Onco Targets Ther 7:1625–1630 Gao YL et al, 2017, Long non-coding RNA PVT1 facilitates cervical Cancer progression via negative regulating ofmiR-424. Oncol Res 25:1391–1398 Guo K et al, 2017, The expression pattern of long non-coding RNA PVT1 in tumor tissues and in extracellular vesicles of colorectal cancer correlates with cancer progression. Tumour Biol 39(4): 1010428317699122 Liu E et al, 2015, Overexpression of long non-coding RNA PVT1 in ovarian cancer cells promotes cisplatin resistance by regulating apoptotic pathways. Int J Clin Exp Med 8(11):20565–20572 Zhang XWet al, 2015, Overexpression of long non-coding RNA PVT1 in gastric cancer cells promotes the development of multidrug resistance. Biochem Biophys Res Commun 462(3):227–232 Zhuang C et al, 2015, Tetracycline-inducible shRNA targeting long non-coding RNA PVT1 inhibits cell growth and induces apoptosis in bladder cancer cells. Oncotarget 6(38):41194–41203 Wang F et al, 2014, Oncofetal long noncoding RNA PVT1 promotes proliferation and stem cell-like property of hepatocellular carcinoma cells by stabilizingNOP2. Hepatology 60(4):1278–1290 Alvarez ML et al, 2013, Role of microRNA 1207-5P and its host gene, the long non-coding RNA Pvt1, as mediators of extracellular matrix accumulation in the kidney: implications for diabetic nephropathy. PLoS One 8(10):e77468 Marur S, Forastiere AA, 2016, Head and neck squamous cell carcinoma: update on epidemiology, diagnosis, and treatment. Mayo Clin Proc 91(3):386–396 Hu S, Shan G, 2016, LncRNAs in Stem Cells. Stem Cells Int 2016: 2681925 Rosa A, Ballarino M, 2016, Long noncoding RNA regulation of pluripotency. Stem Cells Int 2016:1797692 Wang Y et al, 2013, Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Dev Cell 25(1):69–80 Yang YR et al, 2014, Increased expression of the lncRNA PVT1 promotes tumorigenesis in non-small cell lung cancer. Int J Clin Exp Pathol 7(10):6929–6935 Franca GS, Vibranovski MD, Galante PA, 2016, Host gene constraints and genomic context impact the expression and evolution of human microRNAs. Nat Commun 7:11438 Barsotti AM et al, 2012, p53-dependent induction of PVT1 and miR-1204. J Biol Chem 287(4):2509–2519 Riquelme E et al, 2014, Frequent coamplification and cooperation between C-MYC and PVT1 oncogenes promote malignant pleural mesothelioma. J Thorac Oncol 9(7):998–1007 Chen L et al, 2014, miR-1207-5p and miR-1266 suppress gastric cancer growth and invasion by targeting telomerase reverse transcriptase. Cell Death Dis 5:e1034 Zhang S, Cui W, 2014, Sox2, a key factor in the regulation of pluripotency and neural differentiation. World J Stem Cells 6(3): 305–311 Rizzino A, 2013, Concise review: the Sox2-Oct4 connection: critical players in a much larger interdependent network integrated at multiple levels. Stem Cells 31(6):1033–1039 Hadjimichael C et al, 2015, Common stemness regulators of embryonic and cancer stem cells. World J Stem Cells 7(9):1150–1184 Leis O et al, 2012, Sox2 expression in breast tumours and activation in breast cancer stem cells. Oncogene 31(11):1354–1365 Ginestier C et al, 2007, ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1(5):555–567 Xia P, Xu XY, 2015, PI3K/Akt/mTOR signaling pathway in cancer stem cells: from basic research to clinical application. Am J Cancer Res 5(5):1602–1609 Sunayama J et al, 2010, Crosstalk between the PI3K/mTOR and MEK/ERK pathways involved in the maintenance of self-renewal and tumorigenicity of glioblastoma stem-like cells. Stem Cells 28(11):1930–1939 Ruggeri BA et al, 1998, Amplification and overexpression of the AKT2 oncogene in a subset of human pancreatic ductal adenocarcinomas. Mol Carcinog 21(2):81–86 Wen Wet al, 2013, Cyclin G1 expands liver tumor-initiating cells by Sox2 induction via Akt/mTOR signaling. Mol Cancer Ther 12(9):1796–1804 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1411 EP 1422 DI 10.1007/s12253-018-0453-1 PG 12 ER PT J AU Boldrini, L Bartoletti, R Giordano, M Manassero, F Selli, C Panichi, M Galli, L Farci, F Faviana, P AF Boldrini, Laura Bartoletti, Riccardo Giordano, Mirella Manassero, Francesca Selli, Cesare Panichi, Marco Galli, Luca Farci, Fabiola Faviana, Pinuccia TI C-MYC, HIF-1α, ERG, TKT, and GSTP1: an Axis in Prostate Cancer? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; C-myc; HIF-1α; GSTP1; TKT; ERG ID Prostate cancer; C-myc; HIF-1α; GSTP1; TKT; ERG AB To analyze putative biomarkers for prostate cancer (PCA) characterization, the second leading cause of cancer-associated mortality in men. Quantification of the expression level of c-myc and HIF-1α was performed in 72 prostate cancer specimens. A cohort of 497 prostate cancer patients from The Cancer Genome Atlas (TCGA) database was further analyzed, in order to test our hypothesis.We found that high c-myc level was significantly associated with HIF-1α elevated expression (p = 0.008) in our 72 samples. Statistical analysis of 497 TCGA prostate cancer specimens confirmed the strong association (p = 0.0005) of c-myc and HIF-1α expression levels, as we found in our series. Moreover, we found high c-myc levels significantly associated with low Glutatione S-transferase P1 (GSTP1) expression (p = 0.01), with high Transketolase (TKT) expression (p < 0.0001). High TKT levels were found in TCGA samples with low GSTP1 mRNA (p < 0.0001), as shown for c-myc, and with ERG increased expression (p = 0.02). Finally, samples with low GSTP1 expression displayed higher ERG mRNA levels than samples with high GSTP1 score (p < 0.0001), as above shown for c-myc. Our study emphasizes the notion of a potential value of HIF-1α and c-myc as putative biomarkers in prostate cancer; moreover TCGA data analysis showed a putative crosstalk between c-myc, HIF-1α, ERG, TKT, and GSTP1, suggesting a potential use of this axis in prostate cancer. C1 [Boldrini, Laura] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, Via Roma 57, 56126 Pisa, Italy. [Bartoletti, Riccardo] University of Pisa, Department of Translational Research and New TechnologiesPisa, Italy. [Giordano, Mirella] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, Via Roma 57, 56126 Pisa, Italy. [Manassero, Francesca] Pisa University, Division of UrologyPisa, Italy. [Selli, Cesare] University of Pisa, Department of Translational Research and New TechnologiesPisa, Italy. [Panichi, Marco] Pisa University, Department of RadiotherapyPisa, Italy. [Galli, Luca] Pisa University, Division of Medical OncologyPisa, Italy. [Farci, Fabiola] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, Via Roma 57, 56126 Pisa, Italy. [Faviana, Pinuccia] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, Via Roma 57, 56126 Pisa, Italy. RP Boldrini, L (reprint author), University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, 56126 Pisa, Italy. EM laura.boldrini@med.unipi.it CR Gurel B, Iwata T, Koh CM et al, 2008, Nuclear MYC protein overexpression is an early alteration in human prostate carcinogenesis. Mod Pathol 21:1156–1167 Yang G, Timme TL, Frolov A,Wheeler TM, Thompson TC, 2005, Combined c-Myc and caveolin-1 expression in human prostate carcinoma predicts prostate carcinoma progression. Cancer 103:1186– 1194 Koh CM, Bieberich CJ, Dang CV, Nelson WG, Yegnasubramanian S, De Marzo AM, 2010, MYC and prostate cancer. Genes Cancer 1:617–628 Vergis R, Corbishley CM, Norman AR, Bartlett J, Jhavar S, Borre M, Heeboll S, Horwich A, Huddart R, Khoo V, Eeles R, Cooper C, Sydes M, DearnaleyD, Parker C, 2008, Intrinsicmarkers of tumour hypoxia and angiogenesis in localised prostate cancer and outcome of radical treatment: a retrospective analysis of two randomised radiotherapy trials and one surgical cohort study. Lancet Oncol 9: 342–351 Gravdal K, Halvorsen OJ, Haukaas SA, Akslen LA, 2009, Proliferation of immature tumor vessels is a novel marker of clinical progression in prostate cancer. Cancer Res 69:4708–4715 Ranasinghe WK, Xiao L, Kovac S, Chang M, Michiels C, Bolton D, Shulkes A, Baldwin GS, Patel O, 2013, The role of hypoxiainducible factor 1alpha in determining the properties of castrateresistant prostate cancers. PLoS One 8:e54251 Semenza GL, 2003, Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3:721–732 Wang XX, Jia HT, Yang H, LuoMH, Sun T, 2017, Overexpression of glutathione S-transferase P1 inhibits the viability and motility of prostate cancer via targeting MYC and inactivating the MEK/ ERK1/2 pathways. Oncol Res Benito A, Polat IH, Noe V, Ciudad CJ,Marin S, CascanteM(2017, Glucose-6-phosphate dehydrogenase and transketolase modulate breast cancer cell metabolic reprogramming and correlate with poor patient outcome. Oncotarget 8:106693–106706 Hernandez S, Font-Tello A, Juanpere N, de Muga S, Lorenzo M, Salido M, Fumado L, Serrano L, Cecchini L, Serrano S, Lloreta J, 2016, Concurrent TMPRSS2-ERG and SLC45A3-ERG rearrangements plus PTEN loss are not found in low grade prostate cancer and define an aggressive tumor subset. Prostate 76:854–865 Clark J, Attard G, Jhavar S, Flohr P, Reid A, De-Bono J, Eeles R, Scardino P, Cuzick J, Fisher G, Parker MD, Foster CS, Berney D et al, 2008, Complex patterns of ETS gene alteration arise during cancer development in the human prostate. Oncogene 27:1993– 2003 Perner S, Mosquera JM, Demichelis F, Hofer MD, Paris PL, Simko J, Collins C, Bismar TA, Chinnaiyan AM, De Marzo AM, Rubin MA, 2007, TMPRSS2-ERG fusion prostate cancer: an early molecular event associated with invasion. Am J Surg Pathol 31:882– 888 Park K, Dalton JT, Narayanan R, Barbieri CE, Hancock ML, Bostwick DG, Steiner MS, Rubin MA, 2014, TMPRSS2: ERG gene fusion predicts subsequent detection of prostate cancer in patients with high-grade prostatic intraepithelial neoplasia. J Clin Oncol 32:206–211 Demichelis F, Fall K, Perner S, Andren O, Schmidt F, Setlur SR, Hoshida Y, Mosquera JM, Pawitan Y, Lee C, Adami HO, Mucci LA, Kantoff PWet al, 2007, TMPRSS2: ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene 26:4596–4599 Saramaki OR, Harjula AE, Martikainen PM, Vessella RL, Tammela TL, Visakorpi T, 2008, TMPRSS2: ERG fusion identifies a subgroup of prostate cancers with a favorable prognosis. Clin Cancer Res 14:3395–3400 Hoogland AM, Jenster G, van Weerden WM, Trapman J, van der Kwast T, Roobol MJ, Schroder FH, Wildhagen MF, van Leenders GJ, 2012, ERG immunohistochemistry is not predictive for PSA recurrence, local recurrence or overall survival after radical prostatectomy for prostate cancer. Mod Pathol 25:471–479 Hagglof C, Hammarsten P, Stromvall K, Egevad L, Josefsson A, Stattin P, Granfors T, Bergh A, 2014, TMPRSS2- ERG expression predicts prostate cancer survival and associates with stromal biomarkers. PLoS One 9:e86824 Font-Tello A, Juanpere N, de Muga S, Lorenzo M, Lorente JA, Fumado L, Serrano L, Serrano S, Lloreta J, Hernandez S, 2015, Association of ERG and TMPRSS2-ERG with grade, stage, and prognosis of prostate cancer is dependent on their expression levels. Prostate 75:1216–1226 Mohyeldin A, Garzon-Muvdi T, Quinones-Hinojosa, 2010, Oxygen in stem cell biology: a critical component of the stem cell niche. Cell Stem Cell 7:150–161 Keith B, Simon MC, 2007, Hypoxia-inducible factors, stem cells, and cancer. Cell 129:465–472 Palma CS, Tannous MA, Malta TM, Russo EMS, Covas DT, Picanco-Castro V, 2013, Forced expression of OCT4 influences the expression of pluripotent genes in human mesenchymal stem cells and fibroblasts. Genet Mol Res 12:1054–1060 Pelengaris S, Khan M, Evan GI, 2002, Suppression of Myc induced apoptosis in beta cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression. Cell 109:321–334 Vafa O, Wade M, Kern S, Beeche M, Pandita TK, Hampton GM et al, 2002, C-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: a mechanism for oncogene-induced genetic instability. Mol Cell 9:1031–1044 Pouyssegur J, Dayan F, Mazure NM, 2006, Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 441: 437–443 Covello KL, Kehler J, Yu H, Gordan JD, Arsham AM, Hu CJ et al, 2006, HIF-2alpha regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth. Genes Dev 20:557–570 Dang CV, Kim JW, Gao P, Yustein J, 2008, The interplay between MYC and HIF in cancer. Nat Rev Cancer 8:51–56 Gabay M, Li Y, Felsher DW, 2014, MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb Perspect 4 Arvanitis C, Felsher DW, 2006, Conditional transgenic models define how MYC initiates and maintains tumorigenesis. Semin Cancer Biol 16:313–317 Shachaf CM, Kopelman AM, Arvanitis C, Karlsson A, Beer S, Mandl S et al, 2004, MYC inactivation uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer. Nature 431:1112–1117 Felsher DW, Bishop JM, 1999, Reversible tumorigenesis by MYC in hematopoietic lineages. Mol Cell 4:199–207 Jain M, Arvanitis C, Chu K, Dewey W, Leonhardt E, Trinh M et al, 2002, Sustained loss of a neoplastic phenotype by brief inactivation of MYC. Science 297:102–104 Karlsson A, Giuriato S, Tang F, Fung-Weier J, Levan G, Felsher DW, 2003, Genomically complex lymphomas undergo sustained tumor regression upon MYC inactivation unless they acquire novel chromosomal translocations. Blood 101:2797–2803 Beer S, Zetterberg A, Ihrie RA, McTaggart RA, Yang Q, Bradon N et al, 2004, Developmental context determines latency of MYCinduced tumorigenesis. PLoS Biol 2:e332 Nilsson JA, Cleveland JL, 2003, Myc pathways provoking cell suicide and cancer. Oncogene 22:9007–9021 Doe MR, Ascano J, Kaur M, Cole MD, 2012, Myc posttranscriptionally induces HIF1 protein and target gene expression in normal and cancer cells. Cancer Res 72:949–957 Huang LE, Bindra RS, Glazer PM, Harris AL, 2007, Hypoxiainduced genetic instability—a calculated mechanism underlying tumor progression. J Mol Med 85:139–148 Bristow RG, Hill RP, 2008, Hypoxia and metabolism. Hypoxia, DNA repair and genetic instability. Nat Rev Cancer 8:180–192 Reynolds TY, Rockwell S, Glazer PM, 1996, Genetic instability induced by the tumor microenvironment. Cancer Res 56:5754– 5757 Grandori C, Cowley SM, James LP, Eisenman RN, 2000, The Myc/Max/Mad network and the transcriptional control of cell behavior. Annu Rev Cell Dev Biol 16:653–699 Nasi S, Ciarapica R, Jucker R, Rosati J, Soucek L, 2001, Making decisions through Myc. FEBS Lett 490:153–162 Albihn A, Johnsen JI, HenrikssonMA, 2010)MYC in oncogenesis and as a target for cancer therapies. Adv Cancer Res 107:163–224 Eilers M, Eisenman RN, 2008, Myc’s broad reach. Genes Dev 22: 2755–2766 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Xu IM, Lai RK, Lin SH, Tse AP, Chiu DK, Koh HY, Law CT, Wong CM, Cai Z, Wong CC, Ng IO, 2016, Transketolase counteracts oxidative stress to drive cancer development. Proc Natl Acad Sci U S A 113:E725–E734 Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, Varambally S, Cao X, Tchinda J, Kuefer R et al, 2005, Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310:644–648 Adamo P, Porazinski S, Rajatileka S, Jumbe S, Hagen R, Cheung MK, Wilson I, Ladomery MR, 2017, The oncogenic transcription factor ERG represses the transcription of the tumour suppressor gene PTEN in prostate cancer cells. Oncol Lett 14:5605–5610 Adamo P, Ladomery MR, 2016, The oncogene ERG: a key factor in prostate cancer. Oncogene 35:403–414 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1423 EP 1429 DI 10.1007/s12253-018-0479-4 PG 7 ER PT J AU Comert, KG Basaran, D Akkoz, EH Celik, B Sinaci, S Turkmen, O Karalok, A Kandemir, O Turan, T AF Comert, Kimyon Gunsu Basaran, Derman Akkoz, Ergin Hayriye Celik, Burcin Sinaci, Selcan Turkmen, Osman Karalok, Alper Kandemir, Olcay Turan, Taner TI Blood Vessel Invasion in Endometrial Cancer Is One of the Mechanisms of Spread to the Cervix SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lympho-vascular invasion; Podoplanin; CD31; Blood vessel; Endometrial cancer ID Lympho-vascular invasion; Podoplanin; CD31; Blood vessel; Endometrial cancer AB To evaluate the association between type of invaded vessels (blood or lymphatic) and cervical involvement in endometrial cancer (EC). Pathological slides of 93 patients with EC who had vascular space invasion in hematoxylin-eosin staining underwent immunohistochemical assay with CD31 and podoplanin. CD31 and podoplanin were used to identify blood and lymphatic invaded vessels, respectively. Cervical stromal invasion (CSI) was determined in 21 (30%) patients. The rate of CD31- positivity was significantly higher in patients with CSI than without (76.2 and 34.7%, p = 0.001; respectively). Podoplaninpositivity was determined in 47.6 and 81.6% of patients with and without CSI, respectively (p = 0.005). Age, myometrial invasion and the combination of CD31-positivity with podoplanin-negativity were found as independent predictors for CSI. Blood vessel invasion is an important factor for CSI in EC. Blood vessel invasion rather than lymphatic vessel invasion is one of the predominant ways by which EC spreads to the cervix. C1 [Comert, Kimyon Gunsu] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology DepartmentAnkara, Turkey. [Basaran, Derman] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology DepartmentAnkara, Turkey. [Akkoz, Ergin Hayriye] Dr. Abdurrahman Yurtarslan Ankara Oncology Education and Research Hospital, Department of PathologyAnkara, Turkey. [Celik, Burcin] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Pathology DivisionAnkara, Turkey. [Sinaci, Selcan] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology DepartmentAnkara, Turkey. [Turkmen, Osman] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology DepartmentAnkara, Turkey. [Karalok, Alper] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology DepartmentAnkara, Turkey. [Kandemir, Olcay] Dr. Abdurrahman Yurtarslan Ankara Oncology Education and Research Hospital, Department of PathologyAnkara, Turkey. [Turan, Taner] Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology DepartmentAnkara, Turkey. RP Comert, KG (reprint author), Etlik Zubeyde Hanim Women’s Health Teaching and Research Hospital, Gynecologic Oncology Department, Ankara, Turkey. EM gunsukimyon@gmail.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:359–386. , DOI 10. 1002/ijc.29210 Bigelow B, Vekshtein V, Demopoulos RI, 1983, Endometrial carcinoma, stage II: route and extent of spread to the cervix. Obstet Gynecol 62:363–366 Jordan LB, Al-Nafussi A, 2002, Clinicopathological study of the pattern and significance of cervical involvement in cases of endometrial adenocarcinoma. Int J Gynecol Cancer 12:42–48 Tambouret R, Clement PB, Young RH, 2003, Endometrial endometrioid adenocarcinoma with a deceptive pattern of spread to the uterine cervix: a manifestation of stage IIb endometrial carcinoma liable to bemisinterpreted as an independent carcinoma or a benign lesion. Am J Surg Pathol 27:1080–1088 Fanning J, Alvarez PM, Tsukada Y, Piver MS, 1991, Prognostic significance of the extent of cervical involvement by endometrial cancer. Gynecol Oncol 40:46–47 Orezzoli JP, Sioletic S, Olawaiye A, Oliva E, del Carmen MG, 2009, Stage II endometrioid adenocarcinoma of the endometrium: clinical implications of cervical stromal invasion. Gynecol Oncol 113:316–323. , DOI 10.1016/j.ygyno.2009.03.007 Taskin S, Ortac F, Kahraman K, Goc G, Oztuna D, Gungor M, 2013, Cervical stromal involvement can predict survival in advanced endometrial carcinoma: a review of 67 patients. Int J Clin Oncol 18:105–109. , DOI 10.1007/s10147-011-0351-y Ozgul N, Boyraz G, Salman MC, Gultekin M, Yuce K, Ibrahimov A, Erturk A, Gungorduk K, Gulseren V, Sanci M, Turkmen O, Karalok A, Kimyon G, Turan T, Ozkan NT, Meydanli MM, Gungor T, Ayik H, Simsek T, 2018, Oncological outcomes of stage II endometrial Cancer: a retrospective analysis of 250 cases. Int J Gynecol Cancer 28:161–167. , DOI 10.1097/igc. 0000000000001133 Kadar NR, Kohorn EI, LiVolsi VA, Kapp DS, 1982, Histologic variants of cervical involvement by endometrial carcinoma. Obstet Gynecol 59:85–92 Fanning J, Alvarez PM, Tsukada Y, Piver MS, 1991, Cervical implantation metastasis by endometrial adenocarcinoma. Cancer 68: 1335–1339 Mohammed RA, Ellis IO, Lee AH, Martin SG, 2009, Vascular invasion in breast cancer; an overview of recent prognostic developments and molecular pathophysiological mechanisms. Histopathology 55:1–9. , DOI 10.1111/j.1365-2559.2008. 03169.x. Fan L, Mac MT, Frishberg DP, Fan X, Dhall D, Balzer BL, Geller SA, Wang HL, 2010, Interobserver and intraobserver variability in evaluating vascular invasion in hepatocellular carcinoma. J Gastroenterol Hepatol 25:1556–1561. , DOI 10.1111/j. 1440-1746.2010.06304.x Folkins AK, Nevadunsky NS, Saleemuddin A, Jarboe EA, Muto MG, Feltmate CM, Crum CP, Hirsch MS, 2010, Evaluation of vascular space involvement in endometrial adenocarcinomas: laparoscopic vs abdominal hysterectomies. Mod Pathol 23:1073–1079. , DOI 10.1038/modpathol.2010.91 Kryvenko ON, Epstein JI, 2012, Histologic criteria and pitfalls in the diagnosis of lymphovascular invasion in radical prostatectomy specimens. Am J Surg Pathol 36:1865–1873. , DOI 10. 1097/PAS.0b013e318262c3d0 Alexander-Sefre F, Singh N, Ayhan A, Salveson HB, Wilbanks G, Jacobs IJ, 2003, Detection of tumour lymphovascular space invasion using dual cytokeratin and CD31 immunohistochemistry. J Clin Pathol 56:786–788 Birner P, Schindl M, Obermair A, Breitenecker G, Kowalski H, Oberhuber G, 2001, Lymphatic microvessel density as a novel prognostic factor in early-stage invasive cervical cancer. Int J Cancer 95:29–33 Vandenput I, Vanhove T, Calster BV, Gorp TV, Moerman P, Verbist G, Vergote I, Amant F, 2010, The use of lymph vessel markers to predict endometrial cancer outcome. Int J Gynecol Cancer 20:363– 367. , DOI 10.1111/IGC.0b013e3181d4a0b9 Weber SK, Sauerwald A, Polcher M, Braun M, Debald M, Serce NB, Kuhn W, Brunagel-Walgenbach G, Rudlowski C, 2012, Detection of lymphovascular invasion by D2-40, podoplanin, immunoexpression in endometrial cancer. Int J Gynecol Cancer 22:1442–1448. , DOI 10.1097/IGC.0b013e318269139b Yoshida BA, Sokoloff MM, Welch DR, Rinker-Schaeffer CW, 2000, Metastasis-suppressor genes: a review and perspective on an emerging field. J Natl Cancer Inst 92:1717–1730 Guset G, Costi S, Lazar E, Dema A, CornianuM, Vernic C, Paiusan L, 2010, Expression of vascular endothelial growth factor, VEGF, and assessment of microvascular density with CD34 as prognostic markers for endometrial carcinoma. Romanian J Morphol Embryol 51:677–682 Mohammed ZM, McMillan DC, Edwards J, Mallon E, Doughty JC, Orange C, Going JJ, 2013, The relationship between lymphovascular invasion and angiogenesis, hormone receptors, cell proliferation and survival in patients with primary operable invasive ductal breast cancer. BMC Clin Pathol 13:31. , DOI 10. 1186/1472-6890-13-31 Chaffer CL, Weinberg RA, 2011, A perspective on cancer cell metastasis. Science 331:1559–1564. , DOI 10.1126/ science.1203543 Rubin SC, Hoskins WJ, Saigo PE, Nori D, Mychalczak B, Chapman D, Lewis JL Jr, 1992, Management of endometrial adenocarcinoma with cervical involvement. Gynecol Oncol 45:294– 298 Clement PB, Young RH, 2002, Endometrioid carcinoma of the uterine corpus: a review of its pathology with emphasis on recent advances and problematic aspects. Adv Anat Pathol 9:145–184 Birner P, Obermair A, Schindl M, Kowalski H, Breitenecker G, Oberhuber G, 2001, Selective immunohistochemical staining of blood and lymphatic vessels reveals independent prognostic influence of blood and lymphatic vessel invasion in early-stage cervical cancer. Clin Cancer Res 7:93–97 Martini T, Strobel P, Steidler A, Petrakopoulou N, Erben P, Bolenz C, 2015, Detection of lymphovascular invasion in urothelial carcinoma of the bladder through D2-40 immunostaining. Urologe A 54:70–75. , DOI 10.1007/s00120-014-3646-6 Van den Eynden GG, Van der Auwera I, Van Laere SJ, Colpaert CG, van Dam P, Dirix LY, Vermeulen PB, Van Marck EA, 2006, Distinguishing blood and lymph vessel invasion in breast cancer: a prospective immunohistochemical study. Br J Cancer 94:1643– 1649 Obermair A,Wanner C, Bilgi S, Speiser P, Reisenberger K, Kaider A, Kainz C, Leodolter S, Breitenecker G, Gitsch G, 1998, The influence of vascular space involvement on the prognosis of patients with stage IB cervical carcinoma: correlation of results from hematoxylin and eosin staining with results from immunostaining for factor VIII-related antigen. Cancer 82:689–696 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1431 EP 1436 DI 10.1007/s12253-018-0498-1 PG 6 ER PT J AU Yu, D Cheng, J Xue, K Zhao, X Wen, L Xu, Ch AF Yu, Dan Cheng, Jinzhang Xue, Kai Zhao, Xue Wen, Lianji Xu, Chengbi TI Expression of Programmed Death-Ligand 1 in Laryngeal Carcinoma and its Effects on Immune Cell Subgroup Infiltration SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PDL-1; TAMs; Laryngeal squamous cell carcinoma; Immunosuppression ID PDL-1; TAMs; Laryngeal squamous cell carcinoma; Immunosuppression AB To study the expression of programmed death-ligand 1 (PD-L1), and its effects on CD8+ tumor infiltrating lymphocytes (TILs) and tumor associated macrophages (TAMs) in human laryngeal squamous cell carcinoma. Sixty-nine patients with laryngeal carcinoma and 10 with vocal cord leukoplakia received tumor resection at Neck Surgery Department in the Second Affiliation Hospital of Jilin University (Changchun, Jilin) from Jan. 2010 to Dec. 2015. The expressions of PD-L1, CD8, CD16 and CD206 in laryngeal carcinoma, paracancerous and vocal cord leukoplakia tissues were detected with immunohistochemistry. The associations between PD-L1 expression and clinicopathologic features, expression of TAMs and CD8+ T cell infiltration were analyzed. Expression of PD-L1 is significantly higher in laryngeal carcinoma than in paracancerous or leukoplakia tissue. The expression of PD-L1 is closely associated with stage of laryngeal cancer, histological differentiation and neck lymphatic metastasis. PD-L1 expression is negatively correlated with the number of CD8+ TILs and CD16+ cells (M1 type TAMs), while is positively associated with CD206+ (M2 type TAMs). PD-L1 is highly expressed in the laryngeal cancer with the tumor microenvironment immunosuppression. C1 [Yu, Dan] Jilin University, The Second Hospital, Department of Otolaryngology Head and Neck Surgery, No. 218 Ziqiang Street, 130041 Changchun, China. [Cheng, Jinzhang] Jilin University, The Second Hospital, Department of Otolaryngology Head and Neck Surgery, No. 218 Ziqiang Street, 130041 Changchun, China. [Xue, Kai] Jilin University, The Second Hospital, Department of Otolaryngology Head and Neck Surgery, No. 218 Ziqiang Street, 130041 Changchun, China. [Zhao, Xue] Jilin University, The Second Hospital, Department of Otolaryngology Head and Neck Surgery, No. 218 Ziqiang Street, 130041 Changchun, China. [Wen, Lianji] Jilin University, The Second Hospital, Department of Otolaryngology Head and Neck Surgery, No. 218 Ziqiang Street, 130041 Changchun, China. [Xu, Chengbi] Jilin University, The Second Hospital, Department of Otolaryngology Head and Neck Surgery, No. 218 Ziqiang Street, 130041 Changchun, China. RP Xu, Ch (reprint author), Jilin University, The Second Hospital, Department of Otolaryngology Head and Neck Surgery, 130041 Changchun, China. EM xuchengbi@163.com CR Ferris RL, Whiteside TL, Ferrone S, 2006, Immune escape associated with functional defects in antigen-processing machinery in head and neck cancer. Clin Cancer Res 12(13):3890–3895 Zandberg DP, Strome SE, 2014, The role of the PD-L1:PD-1 pathway in squamous cell carcinoma of the head and neck. Oral Oncol 50(7):627–632 Jinushi M, Komohara Y, 2015, Tumor-associated macrophages as an emerging target against tumors: creating a new path from bench to bedside. Biochim Biophys Acta 1855(2):123–130 Lee YH,Martin-Orozco N, Zheng P, Li J, Zhang P, Tan H, Park HJ, Jeong M, Chang SH, Kim BS, Xiong W, Zang W, Guo L, Liu Y, Dong ZJ, OverwijkWW, Hwu P, Yi Q, Kwak L, Yang Z,Mak TW, Li W, Radvanyi LG, Ni L, Liu D, Dong C, 2017, Inhibition of the B7-H3 immune checkpoint limits tumor growth by enhancing cytotoxic lymphocyte function. Cell Res 27:1034–1045 Liu Z, Zhao Y, Fang J, Cui R, Xiao Yet al, 2017, SHP2 negatively regulates HLA-ABC and PD-L1 expression via STAT1 phosphorylation in prostate cancer cells. Oncotarget Fife BT, Pauken KE, Eagar TN, Obu T, Wu J, Tang Q, Azuma M, Krummel MF, Bluestone JA, 2009, Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nat Immunol 10(11):1185–1192 Zhang L, Li Z, Ding G, La X, Yang P et al, 2017, GRP78 plays an integral role in tumor cell inflammation-related migration induced by M2 macrophages. Cell Signal 37:136–148 Chen J, Li G, Meng H, Fan Y, Song Y, Wang S, Zhu F, Guo C, Zhang L, Shi Y, 2012, Upregulation of B7-H1 expression is associated with macrophage infiltration in hepatocellular carcinomas. Cancer Immunol Immunother 61(1):101–108 Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, Gupta R, Tsai JM, Sinha R, Corey D, Ring AM, Connolly AJ, Weissman IL, 2017, PD-1 expression by tumourassociated macrophages inhibits phagocytosis and tumour immunity. Nature 545(7655):495–499 Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, KuchrooVK, Sharpe AH, 2009, PD-L1 regulates the development, maintenance, and function of induced regulatory Tcells. J Exp Med 206(13):3015–3029 YvorelV, Patoir A, Casteillo F, Tissot C, Fournel P, StachowiczML, Karpathiou G, Tiffet O, Peoc’h M, Forest F, 2017, PD-L1 expression in pleomorphic, spindle cell and giant cell carcinoma of the lung is related to TTF-1, p40 expression and might indicate a worse prognosis. PLoS One 12(7):e0180346 Roper E, Lum T, Palme CE, Ashford B, Ch'ng S, RansonM, Boyer M, Clark J, Gupta R, 2017, PD-L1 expression predicts longer disease free survival in high risk head and neck cutaneous squamous cell carcinoma. Pathology 49:499–505 Milstein O, Hagin D, Lask A, Reich-Zeliger S, Shezen E, Ophir E, Eidelstein Y, Afik R, Antebi YE, Dustin ML, Reisner Y, 2011, CTLs respond with activation and granule secretion when serving as targets for T-cell recognition. Blood 117(3):1042–1052 Mischinger J, Comperat E, Schwentner C, Stenzl A, Gakis G, 2015, Inflammation and Cancer: what can we therapeutically expect from checkpoint inhibitors? Curr Urol Rep 16(9):59 Ager CR, Reilley MJ, Nicholas C, Bartkowiak T, Jaiswal AR, Curran MA, 2017, Intratumoral STING activation with T cell checkpoint modulation generates systemic antitumor immunity. Cancer Immunol Res 5:676–684 Fujii N, Shomori K, Shiomi T, Nakabayashi M, Takeda C, Ryoke K, Ito H, 2012, Cancer-associated fibroblasts and CD163-positive macrophages in oral squamous cell carcinoma: their clinicopathological and prognostic significance. J Oral Pathol Med 41(6):444– 451 Chen C, Qu QX, Shen Y, Mu CY, Zhu YB, Zhang XG, Huang JA, 2012, Induced expression of B7-H4 on the surface of lung cancer cell by the tumor-associated macrophages: a potential mechanism of immune escape. Cancer Lett 317(1):99–105 Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, Wu C, Zheng L, 2009, Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med 206(6):1327–1337 Kang FB,Wang L, LiD, ZhangYG, Sun DX(2015, Hepatocellular carcinomas promote tumor-associated macrophage M2- polarization via increased B7-H3 expression. Oncol Rep 33(1): 274–282 Chen W, Jiang J, Xia W, Huang J, 2017, Tumor-related exosomes contribute to tumor-promoting microenvironment: an immunological perspective. J Immunol Res 2017:1073947 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1437 EP 1443 DI 10.1007/s12253-018-0501-x PG 7 ER PT J AU Foj, L Filella, X AF Foj, Laura Filella, Xavier TI Identification of Potential miRNAs Biomarkers for High-Grade Prostate Cancer by Integrated Bioinformatics Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE miRNAs; Bioinformatics analysis; Differentially expressed genes; Protein–protein interaction network; Prostate cancer ID miRNAs; Bioinformatics analysis; Differentially expressed genes; Protein–protein interaction network; Prostate cancer AB The increasing number of datasets available in the GEO database offers a new approach to identify new miRNAs related to PCa. The aim of our study was to suggest a miRNA signature for the detection of high-grade PCa (Gleason score ≥ 7) using bioinformatics tools. Three mRNA datasets (GSE26022, GSE30521, GSE46602) were selected to identify the differentially expressed genes (DEGs) in high-grade PCa. Furthermore, two miRNA datasets (GSE45604, GSE46738) were analyzed to select the differentially expressed miRNAs (DEMs). Functional and pathway enrichment analysis was performed using DAVID and a protein-protein interaction network (PPI) was constructed through STRING. Besides, miRNAs which regulate hub genes were predicted using microRNA.org. A total of 973 DEGs were identified after the analyses of the mRNA datasets, enriched in key mechanisms underlying PCa development. Furthermore, we identified 10 hub genes (EGFR, VEGFA, IGF1, PIK3R1, CD44, ITGB4, ANXA1, BCL2, LPAR3, LPAR1). The most significant KEGG Pathway was PI3K-Akt signaling pathway, involved in cell proliferation and survival. Moreover, we identified 30 common miRNAs between significant DEMs and the predicted hub gene regulators. Twelve of these miRNAs (miR-1, −365, −132, −195, −133a, −133b, −200c, −339, −222, −21, −221, −708) regulate two or more hub genes identified in our study. We suggested a signature including these 12 miRNAs for high-grade PCa detection. These miRNAs have been associated with aggressive PCa, poor survival and resistance to treatment in the last years. C1 [Foj, Laura] SYNLAB Group, Labco DiagnosticsBarcelona, Catalonia, Spain. [Filella, Xavier] Hospital Clinic, IDIBAPS, Department of Biochemistry and Molecular Genetics (CDB), C/ Villarroel, 170, 08036 Barcelona, Catalonia, Spain. RP Filella, X (reprint author), Hospital Clinic, IDIBAPS, Department of Biochemistry and Molecular Genetics (CDB), 08036 Barcelona, Spain. EM xfilella@clinic.cat CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:E359–E386 Lepor H, Donin NM, 2014, Gleason 6 prostate cancer: serious malignancy or toothless lion? Oncology, Williston Park, 28:16–22 Ambs S, Prueitt RL, Yi M, Hudson RS, Howe TM, Petrocca F, Wallace TA, Liu CG, Volinia S, Calin GA, Yfantis HG, Stephens RM, Croce CM, 2008, Genomic profiling of microRNA and messenger RNA reveals deregulated microRNA expression in prostate cancer. Cancer Res 68:6162–6170 Scher HI, Sawyers CL, 2005, Biology of progressive, castrationresistant prostate cancer: directed therapies targeting the androgenreceptor signaling axis. J Clin Oncol 23:8253–8261 Gao L, Alumkal J, 2010, Epigenetic regulation of androgen receptor signaling in prostate cancer. Epigenetics 5:100–104 Filella X, Foj L, 2017, miRNAs as novel biomarkers in the management of prostate cancer. Clin Chem Lab Med 55:715–736 Agell L, Hernandez S, Nonell L, Lorenzo M, Puigdecanet E, de Muga S, Juanpere N, Bermudo R, Fernandez PL, Lorente JA, Serrano S, Lloreta J, 2012, A 12-gene expression signature is associated with aggressive histological in prostate cancer: SEC14L1 and TCEB1 genes are potential markers of progression. Am J Pathol 181:1585–1594 MortensenMM, Hoyer S, Lynnerup AS, Orntoft TF, Sorensen KD, BorreM, Dyrskjot L, 2015, Expression profiling of prostate cancer tissue delineates genes associated with recurrence after prostatectomy. Sci Rep 5:16018 Casanova-Salas I, Rubio-Briones J, Calatrava A, Mancarella C, Masia E, Casanova J, Fernandez-Serra A, Rubio L, Ramirez- Backhaus M, Arminan A, Dominguez-Escrig J, Martinez F, Garcia-Casado Z, Scotlandi K, Vicent MJ, Lopez-Guerrero JA, 2014, Identification of miR-187 and miR-182 as biomarkers of early diagnosis and prognosis in patients with prostate cancer treated with radical prostatectomy. J Urol 192:252–259 Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, Yefanov A, Lee H, Zhang N, Robertson CL, Serova N, Davis S, Soboleva A, 2013, NCBI GEO: archive for functional genomics data sets-update. Nucleic Acids Res 41:D991–D995 Da Huang W, Sherman BT, Lempicki RA, 2009, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4:44–57 Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G, 2000, Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 25:25–29 Kanehisa M, 2002, The KEGG database. Novartis Found Symp 247:91–101 Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, Kuhn M, Bork P, Jensen LJ, von Mering C, 2015, STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43:D447–D452 Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T, 2003, Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13:2498–2504 Betel D, Koppal A, Agius P, Sander C, Leslie C, 2010, Comprehensive modeling of microRNA targets predicts functional non-conserved and non-canonical sites. Genome Biol 11:R90 Jorissen RN, Walker F, Pouliot N, Garrett TP, Ward CW, Burgess AW, 2003, Epidermal growth factor receptor: mechanisms of activation and signalling. Exp Cell Res 284:31–53 Di Lorenzo G, Tortora G, D'Armiento FP, De Rosa G, Staibano S, Autorino R et al, 2002, Expression of epidermal growth factor receptor correlates with disease relapse and progression to androgen-independence in human prostate cancer. Clin Cancer Res 8:3438–3444 Mendelsohn J, Baselga J, 2000, The EGF receptor family as targets for cancer therapy. Oncogene 19:6550–6565 Baek KH, HongME, JungYY, Lee CH, Lee TJ, Park ES, KimMK, Yoo JH, Lee SW, 2012, Correlation of AR, EGFR, and HER2 expression levels in prostate Cancer: Immunohistochemical analysis and chromogenic in situ hybridization. Cancer Res Treat 44:50–56 Sridhar SS, Hotte SJ, Chin JL, Hudes GR, Gregg R, Trachtenberg J, Wang L, Tran-Thanh D, Pham NA, Tsao MS, Hedley D, Dancey JE, Moore MJ, 2010, A multicenter phase II clinical trial of lapatinib, GW572016, in hormonally untreated advanced prostate cancer. Am J Clin Oncol 33:609–613 Brizzolara A, Benelli R, Vene R, Barboro P, Poggi A, Tosetti F, Ferrari N, 2017, The ErbB family and androgen receptor signaling are targets of celecoxib in prostate cancer. Cancer Lett 400:9–17 Thamilselvan V, Menon M, Stein GS, Valeriote F, Thamilselvan S, 2017, Combination of Carmustine and selenite inhibits EGFR mediated growth signaling in androgen-independent prostate Cancer cells. J Cell Biochem 118:4331–4340 Jiang X, Chen S, Asara JM, Balk SP, 2010, Phosphoinositide 3- kinase pathway activation in phosphate and tensin homolog, PTEN)-deficient prostate cancer cells is independent of receptor tyrosine kinases and mediated by the p110beta and p110delta catalytic subunits. J Biol Chem 285:14980–14989 Mithal P, Allott E, Gerber L, Reid J,WelbournW, Tikishvili E, Park J, Younus A, Sangale Z, Lanchbury JS, Stone S, Freedland SJ, 2014, PTEN loss in biopsy tissue predicts poor clinical outcomes in prostate cancer. Int J Urol 21:1209–1214 Xie H, Xie B, Liu C, Wang J, Xu Y, 2017, Association of PTEN expression with biochemical recurrence in prostate cancer: results based on previous reports. Onco Targets Ther 10:5089–5097 Liu L, Dong X, 2014, Complex impacts of PI3K/AKTinhibitors to androgen receptor gene expression in prostate cancer cells. PLoS One 9:e108780 Carver BS, Chapinski C, Wongvipat J, Hieronymus H, Chen Y, Chandarlapaty S, Arora VK, le C, Koutcher J, Scher H, Scardino PT, Rosen N, Sawyers CL, 2011, Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell 19:575–586 WebsterRJ, GilesKM,Price KJ, ZhangPM,Mattick JS, Leedman PJ, 2009, Regulation of epidermal growth factor receptor signaling in human cancer cells by microRNA-7. J Biol Chem 284: 5731–5741 Reddy SD, Ohshiro K, Rayala SK, Kumar R, 2008, MicroRNA-7, a homeobox D10 target, inhibits p21-activated kinase 1 and regulates its functions. Cancer Res 68:8195–8200 Chang YL, Zhou PJ,Wei L, LiW, Ji Z, Fang YX, GaoWQ, 2015, MicroRNA-7 inhibits the stemness of prostate cancer stem-like cells and tumorigenesis by repressing KLF4/PI3K/Akt/p21 pathway. Oncotarget 6:24017–24031 Kalinowski FC, Giles KM, Candy PA, Ali A, Ganda C, Epis MR, Webster RJ, Leedman PJ, 2012, Regulation of epidermal growth factor receptor signaling and erlotinib sensitivity in head and neck cancer cells by miR-7. PLoS One 7:e47067 Liu Z, Jiang Z, Huang J, Huang S, Li Y, Yu S et al, 2014, miR-7 inhibits glioblastoma growth by simultaneously interfering with the PI3K/ATK and Raf/MEK/ERK pathways. Int J Oncol 44: 1571–1580 Folini M, Gandellini P, Longoni N, Profumo V, Callari M, Pennati M, Colecchia M, Supino R, Veneroni S, Salvioni R, Valdagni R, Daidone M, Zaffaroni N, 2010, miR-21: an oncomir on strike in prostate cancer. Mol Cancer 9:12 LiT,LiD,Sha JJ, SunP,HuangYR(2009)MicroRNA-21 directly targetsMARCKSand promotes apoptosis resistance and invasion in prostate cancer cells. Biochem Biophys Res Commun 383: 280–285 Coppola V, Musumeci M, Patrizii M, Cannistraci A, Addario A, Maugeri-Sacca M, Biffoni M, Francescangeli F, Cordenonsi M, Piccolo S, Memeo L, Pagliuca A, Muto G, Zeuner A, de Maria R, Bonci D, 2013, BTG2 loss and miR-21 upregulation contribute to prostate cell transformation by inducing luminal markers expression and epithelial–mesenchymal transition. Oncogene 32: 1843–1853 Ayub SG, Kaul D, Ayub T, 2015, Microdissecting the role of micro-RNAs in the pathogenesis of prostate cancer. Cancer Gene 208:289–302 QuW, Ding SM, Cao G,Wang SJ, Zheng XH, Li GH, 2016, miR- 132 mediates a metabolic shift in prostate cancer cells by targeting Glut1. FEBS Open Bio 6:735–741 Zhang X, Tao T, Liu C, Guan H, Huang Y, Xu B et al, 2016, Downregulation of miR-195 promotes prostate cancer progression by targeting HMGA1. Oncol Rep 36:376–382 McDonald AC, Vira M, Shen J, Sanda M, Raman JD, Liao J et al, 2018, Circulating microRNAs in plasma as potential biomarkers for the early detection of prostate cancer. Prostate 78:411–418 Culig Z, 2014, Proinflammatory cytokine interleukin-6 in prostate carcinogenesis. Am J Clin Exp Urol 2:231–238 Goto Y, Kojima S, Nishikawa R, Kurozumi A, KatoM, Enokida H, Matsushita R, Yamazaki K, Ishida Y, Nakagawa M, Naya Y, Ichikawa T, Seki N, 2015, MicroRNA expression signature of castration-resistant prostate cancer: the microRNA-221/222 cluster functions as a tumour suppressor and disease progression marker. Br J Cancer 113:1055–1065 Sun T, Wang X, He HH, Sweeney CJ, Liu SX, Brown M, Balk S, Lee GSM, Kantoff PW, 2014, MiR-221 promotes the development of androgen independence in prostate cancer cells via downregulation of HECTD2 and RAB1A. Oncogene 33:2790–2800 Yang X, Yang Y, Gan R, Zhao L, Li W, Zhou H, Wang X, Lu J, Meng QH, 2014, Downregulation of mir-221 and mir-222 restrain prostate cancer cell proliferation and migration that is partly mediated by activation of SIRT1. PLoS One 9:e98833 Medina R, Zaidi SK, Liu CG, Stein JL, van Wijnen AJ, Croce CM et al, 2008, MicroRNAs 221 and 222 bypass quiescence and compromise cell survival. Cancer Res 68:2773–2780 Kojima S, Chiyomaru T, Kawakami K, Yoshino H, Enokida H, Nohata N, Fuse M, Ichikawa T, Naya Y, Nakagawa M, Seki N, 2012, Tumour suppressors miR-1 and miR-133a target the oncogenic function of purine nucleoside phosphorylase, PNP, in prostate cancer. Br J Cancer 106:405–413 Chang YS, Chen WY, Yin JJ, Sheppard-Tillman H, Huang J, Liu YN, 2015, EGF receptor promotes prostate Cancer bone metastasis by downregulating miR-1 and activating TWIST1. Cancer Res 75: 3077–3086 Matin F, Jeet V, Clements JA, Yousef GM, Batra J, 2016, MicroRNA Theranostics in prostate Cancer precision medicine. Clin Chem 62:1318–1333 Chen C, Huang X,Wang Y, Lin L, Liu L, Li G,Wu S, Xu C, Zhou J, ShuaiX(2017, Polymeric vector-mediated delivery of anmiR-21 inhibitor for prostate cancer treatment. RSC Adv 7:11057–11066 Mercatelli N, Coppola V, Bonci D, Miele F, Costantini A, Guadagnoli M, Bonanno E, Muto G, Frajese GV, de Maria R, Spagnoli LG, Farace MG, Ciafre SA, 2008, The inhibition of the highly expressed miR-221 and miR-222 impairs the growth of prostate carcinoma xenografts in mice. PLoS One 3:e4029 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1445 EP 1456 DI 10.1007/s12253-018-0508-3 PG 12 ER PT J AU Chmelarova, M Baranova, I Ruszova, E Laco, J Hrochova, K Dvorakova, E Palicka, V AF Chmelarova, Marcela Baranova, Ivana Ruszova, Ema Laco, Jan Hrochova, Katerina Dvorakova, Eva Palicka, Vladimir TI Importance of Cadherins Methylation in Ovarian Cancer: a Next Generation Sequencing Approach SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Methylation; Cadherins; Ovarian cancer; Epigenetics; Next generation sequencing ID Methylation; Cadherins; Ovarian cancer; Epigenetics; Next generation sequencing AB Epigenetic aberrations are well known to play an important role in carcinogenesis, and also have a great potential to serve as biomarkers in many types of cancers, including ovarian cancer in which sensitive and specific biomarkers and detection methods are critically needed. The aim of this study was to investigate methylation of cadherin genes CDH10, CDH13 and CDH18 in ovarian cancer tissue by comparison with control tissue. The study group consisted of 38 patients with ovarian cancer and 25 control patients. For detection of epigenetic events we used next generation sequencing, the most important data were confirmed using high-resolution melting analysis and real-time PCR. We observed significantly higher methylation in CDH13, sporadic methylation in CDH10 and loss of methylation in CDH18 in the ovarian cancer group compared with the control group. These observations suggest that changes in methylation of cadherin genes may be one of the major mechanisms associated with ovarian cancer progression. In addition, because of the high frequency of methylation of the CDH13 gene in the early stages of ovarian cancer, the analyzed CpG sites might be good targets for next study of potential ovarian cancer screening biomarkers. C1 [Chmelarova, Marcela] Charles University, Faculty of Medicine and University Hospital in Hradec Kralove, Institute for Clinical Biochemistry and DiagnosticsHradec Kralove, Czech Republic. [Baranova, Ivana] Charles University, Faculty of Medicine and University Hospital in Hradec Kralove, Institute for Clinical Biochemistry and DiagnosticsHradec Kralove, Czech Republic. [Ruszova, Ema] University Hospital Hradec Kralove, Department of Clinical GeneticsHradec Kralove, Czech Republic. [Laco, Jan] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, The Fingerland Department of PathologyHradec Kralove, Czech Republic. [Hrochova, Katerina] Charles University, Faculty of Medicine and University Hospital in Hradec Kralove, Institute for Clinical Biochemistry and DiagnosticsHradec Kralove, Czech Republic. [Dvorakova, Eva] Charles University, Faculty of Medicine and University Hospital in Hradec Kralove, Department of Obstetrics and GynecologyHradec Kralove, Czech Republic. [Palicka, Vladimir] Charles University, Faculty of Medicine and University Hospital in Hradec Kralove, Institute for Clinical Biochemistry and DiagnosticsHradec Kralove, Czech Republic. RP Baranova, I (reprint author), Charles University, Faculty of Medicine and University Hospital in Hradec Kralove, Institute for Clinical Biochemistry and Diagnostics, Hradec Kralove, Czech Republic. EM ivana.baranova@fnhk.cz CR Brinton LA, Sahasrabuddhe V, Trabert B, 2013, Epidemiology of gynecologic cancers. In: Barakat RR, Berchuck A, Markman M, RandfallME(eds, Principles and practice of gynecologic oncology, 6th edn. Lippincott Williams & Wilkins, Philadelphia, pp 1–29 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JWW, Comber H, Forman D, Bray F, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49(6):1374–1403. , DOI 10. 1016/j.ejca.2012.12.027 Karlan-Beth Y, 2009, Patients at high risk for ovarian Cancer should undergo routine screening. Clinical Ovarian Cancer 2:83–89 Kurman RJ, Shih Ie M, 2010, The origin and pathogenesis of epithelial ovarian cancer: a proposed unifying theory. Am J Surg Pathol 34(3):433–443. , DOI 10.1097/PAS. 0b013e3181cf3d79 Barton CA, Hacker NF, Clark SJ, O'Brien PM, 2008, DNA methylation changes in ovarian cancer: implications for early diagnosis, prognosis and treatment. Gynecol Oncol 109(1):129–139. https:// doi.org/10.1016/j.ygyno.2007.12.017 Warton K, Samimi G, 2015, Methylation of cell-free circulating DNA in the diagnosis of cancer. Front Mol Biosci 2:13. https:// doi.org/10.3389/fmolb.2015.00013 Koukoura O, Spandidos DA, Daponte A, Sifakis S, 2014, DNA methylation profiles in ovarian cancer: implication in diagnosis and therapy, review). Mol Med Rep 10(1):3–9. , DOI 10.3892/ mmr.2014.2221 Zhang Q, Burdette JE, Wang J-P, 2014, Integrative network analysis of TCGA data for ovarian cancer. BMC Syst Biol 8(1):1–18. , DOI 10.1186/s12918-014-0136-9 Berx G, van Roy F, 2009, Involvement of members of the cadherin superfamily in cancer. Cold Spring Harb Perspect Biol 1(6): a003129. , DOI 10.1101/cshperspect.a003129 Angst BD, Marcozzi C, Magee AI, 2001, The cadherin superfamily: diversity in form and function. J Cell Sci 114(Pt 4):629–641 Asiaf A, Ahmad ST, Aziz SA, Malik AA, Rasool Z, Masood A, Zargar MA, 2014, Loss of expression and aberrant methylation of the CDH1, E-cadherin, gene in breast cancer patients from Kashmir. Asian Pac J Cancer Prev 15(15):6397–6403 Wu X, ZhuangYX, Hong CQ, Chen JY,YouYJ, Zhang F, Huang P, Wu MY, 2014, Clinical importance and therapeutic implication of E-cadherin gene methylation in human ovarian cancer. Med Oncol 31(8):100. , DOI 10.1007/s12032-014-0100-y Chmelarova M, Krepinska E, Spacek J, Laco J, Nekvindova J, Palicka V, 2012, Methylation analysis of tumour suppressor genes in ovarian cancer using MS-MLPA. Folia Biol, Praha, 58(6):246–250 Lin YL, Gui SL, Ma JG, 2015, Aberrant methylation of CDH11 predicts a poor outcome for patients with bladder cancer. Oncol Lett 10(2):647–652. , DOI 10.3892/ol.2015.3337 Mostafavi-Pour Z, Kianpour S, Dehghani M, Mokarram P, Torabinejad S, Monabati A, 2015, Methylation of integrin alpha4 and E-cadherin genes in human prostate Cancer. Pathol Oncol Res 21(4):921–927. , DOI 10.1007/s12253-015-9917-8 Dvorakova E, Chmelarova M, Laco J, Palicka V, Spacek J, 2013, Methylation analysis of tumor suppressor genes in endometroid carcinoma of endometrium using MS-MLPA. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 157(4):298–303. , DOI 10.5507/bp.2013.035 Chmelarova M, Ruszova E, Laco J, Dvorakova E, Smetana J et al, 2015, Monitoring of DNA methylation in ovarian cancer using microarrays. Klin Biochem Metab 23(3):100–104 Kurman RJ, Carcangiu ML, Herrington CS, Young RH, 2014, WHO classification of Tumours of female reproductive organs. Fourth edn. IARC Press, Lyon Bubancova I, Kovarikova H, Laco J, Ruszova E, Dvorak O, Palicka V, Chmelarova M, 2017, Next-generation sequencing approach in methylation analysis of HNF1B and GATA4 genes: searching for biomarkers in ovarian Cancer. Int J Mol Sci 18(2). , DOI 10.3390/ijms18020474 Breier G, Grosser M, Rezaei M, 2014, Endothelial cadherins in cancer. Cell Tissue Res 355(3):523–527. , DOI 10.1007/ s00441-014-1851-7 Rajwar YC, Jain N, Bhatia G, Sikka N, Garg B, Walia E, 2015, Expression and significance of Cadherins and its subtypes in development and progression of Oral cancers: a review. J Clin Diagn Res 9(5):ZE05–ZE07. , DOI 10.7860/JCDR/2015/11964.5907 Andreeva AV, Kutuzov MA, 2010, Cadherin 13 in cancer. Genes ChromosomCancer 49(9):775–790. , DOI 10.1002/gcc.20787 Bol GM, Suijkerbuijk KP, Bart J, Vooijs M, van der Wall E, van Diest PJ, 2010, Methylation profiles of hereditary and sporadic ovarian cancer. Histopathology 57(3):363–370. , DOI 10. 1111/j.1365-2559.2010.03642.x Ho CM, Huang CJ, Huang CY, Wu YY, Chang SF, Cheng WF, 2012, Promoter methylation status of HIN-1 associated with outcomes of ovarian clear cell adenocarcinoma. Mol Cancer 11:53. , DOI 10.1186/1476-4598-11-53 Ozdemir F, Altinisik J, Karateke A, Coksuer H, Buyru N, 2012, Methylation of tumor suppressor genes in ovarian cancer. Exp Ther Med 4(6):1092–1096. , DOI 10.3892/etm.2012.715 Feng Q, Deftereos G, Hawes SE, Stern JE, Willner JB, Swisher EM, Xi L, Drescher C, Urban N, Kiviat N, 2008, DNA hypermethylation, Her-2/neu overexpression and p53 mutations in ovarian carcinoma. Gynecol Oncol 111(2):320–329. , DOI 10. 1016/j.ygyno.2008.07.036 Rathi A, Virmani AK, Schorge JO, Elias KJ, Maruyama R, Minna JD, Mosk SC, Girard L, Fishman DA, Gazdar AF, 2002, Methylation profiles of sporadic ovarian tumors and nonmalignant ovaries from high-risk women. Clin Cancer Res 8, 11):3324–3331 Walker MM, Ellis SM, Auza MJ, Patel A, Clark P, 2008, The intercellular adhesionmolecule, cadherin-10, is a marker for human prostate luminal epithelial cells that is not expressed in prostate cancer. Mod Pathol 21(2):85–95. , DOI 10.1038/ modpathol.3800988 An CH, Je EM, Yoo NJ, Lee SH, 2015, Frameshift mutations of cadherin genes DCHS2, CDH10 and CDH24 genes in gastric and colorectal cancers with high microsatellite instability. Pathol Oncol Res 21(1):181–185. , DOI 10.1007/s12253-014-9804-8 Li C, Gao Z, Li F, Li X, Sun Y,Wang M, Li D,Wang R, Li F, Fang R, Pan Y, Luo X, He J, Zheng L, Xia J, Qiu L, He J, Ye T, Zhang R, HeM, ZhuM, Hu H, Shi T, Zhou X, Sun M, Tian S, Zhou Y,Wang Q, Chen L, Yin G, Lu J, Wu R, Guo G, Li Y, Hu X, Li L, Asan, Wang Q, Yin Y, Feng Q, Wang B, Wang H, Wang M, Yang X, Zhang X, Yang H, Jin L, Wang CY, Ji H, Chen H, Wang J, Wei Q, 2015, Whole exome sequencing identifies frequent somatic mutations in cell-cell adhesion genes in Chinese patients with lung squamous cell carcinoma. Sci Rep 5:14237. , DOI 10.1038/ srep14237 Venkatachalam R, Verwiel ET, Kamping EJ, Hoenselaar E, Gorgens H et al, 2011, Identification of candidate predisposing copy number variants in familial and early-onset colorectal cancer patients. Int J Cancer 129(7):1635–1642. , DOI 10.1002/ ijc.25821 Heikinheimo K, Jee KJ, Morgan PR, Nagy B, Knuutila S, Leivo I, 2007, Genetic changes in sporadic keratocystic odontogenic tumors, odontogenic keratocysts). J Dent Res 86(6):544–549 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1457 EP 1465 DI 10.1007/s12253-018-0500-y PG 9 ER PT J AU Ghazi-Khanloosani, M Bandegi, RA Kokhaei, P Barati, M Pakdel, A AF Ghazi-Khanloosani, Mousa Bandegi, Reza Ahmad Kokhaei, Parviz Barati, Mehdi Pakdel, Abbas TI CRP and LOX-1: a Mechanism for Increasing the Tumorigenic Potential of Colorectal Cancer Carcinoma Cell Line SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Carcinoembryonic antigen; Matrix metalloproteinase; C-reactive protein; Oxidized lowdensity lipoprotein; LOX-1 receptor; Tumorigenic potential ID Carcinoembryonic antigen; Matrix metalloproteinase; C-reactive protein; Oxidized lowdensity lipoprotein; LOX-1 receptor; Tumorigenic potential AB Chronic inflammation and dyslipidemia are associated with an increase in the incidence of colorectal cancer (CRC). Serum Creactive protein (CRP) and oxidized low-density lipoprotein (oxLDL), as Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) ligands, increase during inflammation and dyslipidemia, respectively. To evaluate the effects of CRP on the expression of important genes involved in the development of CRC, the CRC cell line, LS174T, was treated with the commercial CRP. Based on the Real-time PCR data, in the presence of CRP, LOX-1, CEA, MMP1, and MMP2 mRNA expression significantly increased, compared to the control group. Moreover, in the presence of CRP, secretion, and expression of CEA in the cell lysate and conditioned media increased in a concentration-dependent manner. The results of flow cytometry showed that expression of LOX-1 receptors at the cell surface increased significantly in the presence of 10 mg/L of CRP. However, inhibition of LOX-1 receptors with a specific monoclonal antibody reduced the effects of CRP on protein/mRNA expression. In conclusion, Increased CRP level, can potentially elevate the expression of important genes in CRC by stimulating LOX-1 receptors. C1 [Ghazi-Khanloosani, Mousa] Semnan University of Medical Sciences, Faculty of Medicine, Department of BiochemistrySemnan, Iran. [Bandegi, Reza Ahmad] Semnan University of Medical Sciences, Faculty of Medicine, Department of BiochemistrySemnan, Iran. [Kokhaei, Parviz] Semnan University of Medical Sciences, Cancer Research Center and Department of ImmunologySemnan, Iran. [Barati, Mehdi] Semnan University of Medical Sciences, Cancer Research Center and Department of ImmunologySemnan, Iran. [Pakdel, Abbas] Semnan University of Medical Sciences, Faculty of Medicine, Department of BiochemistrySemnan, Iran. RP Pakdel, A (reprint author), Semnan University of Medical Sciences, Faculty of Medicine, Department of Biochemistry, Semnan, Iran. EM pakdel@semums.ac.ir CR Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(1):7–30. , DOI 10.3322/caac.21387 Grivennikov SI, 2013, Inflammation and colorectal cancer: colitisassociated neoplasia. Semin Immunopathol 35(2):229–244. https:// doi.org/10.1007/s00281-012-0352-6 Morrison WB, 2012, Inflammation and cancer: a comparative view. J Vet Intern Med 26(1):18–31. , DOI 10.1111/j. 1939-1676.2011.00836.x Ernst PB, Gold BD, 2000, The disease spectrum of helicobacter pylori: the immunopathogenesis of gastroduodenal ulcer and gastric cancer. Annu Rev Microbiol 54:615–640. , DOI 10.1146/ annurev.micro.54.1.615 Swidnicka-Siergiejko AK, Gomez-Chou SB, Cruz-Monserrate Z, Deng D, Liu Y, Huang H, Ji B, Azizian N, Daniluk J, LuW,Wang H, Maitra A, Logsdon CD, 2017, Chronic inflammation initiates multiple forms of K-Ras-independent mouse pancreatic cancer in the absence of TP53. Oncogene 36(22):3149–3158. , DOI 10.1038/onc.2016.461 Rasch S, Algul H, 2014, A clinical perspective on the role of chronic inflammation in gastrointestinal cancer. Clin Exp Gastroenterol 7:261–272. , DOI 10.2147/CEG.S43457 Chen M, Masaki T, Sawamura T, 2002, LOX-1, the receptor for oxidized low-density lipoprotein identified from endothelial cells: implications in endothelial dysfunction and atherosclerosis. Pharmacol Ther 95(1):89–100 Shih HH, Zhang SW, CaoW, Hahn A,Wang J, Paulsen JE, Harnish DC, 2009, CRP is a novel ligand for the oxidized LDL receptor LOX-1. Am J Physiol Heart Circ Physiol 296(5):H1643–H1650. , DOI 10.1152/ajpheart.00938.2008 Guo YZ, Pan L, Du CJ, Ren DQ, Xie XM, 2013, Association between C-reactive protein and risk of cancer: a meta-analysis of prospective cohort studies. Asian Pac J Cancer Prev 14(1):243–248 Lin MS, Huang JX, Zhu JY, Shen HZ, 2013, Elevated pretreatment levels of high sensitivity C-reactive protein as a potential prognosticator in patients with colorectal cancer. Exp Ther Med 6(6):1369–1374. , DOI 10.3892/etm.2013.1350 Pakdel A, Malekzadeh M, Naghibalhossaini F, 2016, The association between preoperative serum CEA concentrations and synchronous liver metastasis in colorectal cancer patients. Cancer Biomark 16(2):245–252. , DOI 10.3233/Cbm-150561 Saito G, Sadahiro S, Kamata H, Miyakita H, Okada K, Tanaka A, Suzuki T, 2017, Monitoring of serum carcinoembryonic antigen levels after curative resection of Colon Cancer: cutoff values determined according to preoperative levels enhance the diagnostic accuracy for recurrence. Oncology 92(5):276–282. , DOI 10. 1159/000456075 Aarons CB, Bajenova O, Andrews C, Heydrick S, Bushell KN, Reed KL, Thomas P, Becker JM, Stucchi AF, 2007, Carcinoembryonic antigen-stimulated THP-1macrophages activate endothelial cells and increase cell-cell adhesion of colorectal cancer cells. Clin Exp Metastasis 24(3):201–209. , DOI 10.1007/ s10585-007-9069-7 Gangopadhyay A, Lazure DA, Thomas P, 1997, Carcinoembryonic antigen induces signal transduction in Kupffer cells. Cancer Lett 118(1):1–6. , DOI 10.1016/S0304- 3835(97)00216-4 Kim IH, Lee JE, Yang JH, Jeong JW, Ro S, LeeMA(2017, Clinical significance of changes in systemic inflammatory markers and carcinoembryonic antigen levels in predicting metastatic colorectal cancer prognosis and chemotherapy response. Asia-Pacific journal of clinical oncology 14:239–246. , DOI 10.1111/ajco. 12784 Liang M, Zhang P, Fu J, 2007, Up-regulation of LOX-1 expression by TNF-alpha promotes trans-endothelial migration of MDA-MB- 231 breast cancer cells. Cancer Lett 258(1):31–37. , DOI 10.1016/j.canlet.2007.08.003 Khaidakov M, Mehta JL, 2012, Oxidized LDL triggers prooncogenic signaling in human breastmammary epithelial cells partly via stimulation of MiR-21. PLoS One 7(10):e46973. https://doi. org/10.1371/journal.pone.0046973 Gonzalez-Chavarria I, Cerro RP, Parra NP, Sandoval FA, Zuniga FA, Omazabal VA, Lamperti LI, Jimenez SP, Fernandez EA, Gutierrez NA, Rodriguez FS, Onate SA, Sanchez O, Vera JC, Toledo JR, 2014, Lectin-Like Oxidized LDL Receptor-1 Is an Enhancer of Tumor Angiogenesis in Human Prostate Cancer Cells PLoS One 9, 8). , DOI 10.1371/Journalpone. 0106219 ARTN e106219 Gonzalez-Chavarria I, Fernandez E, Gutierrez N, Gonzalez-Horta EE, Sandoval F, Cifuentes P, Castillo C, Cerro R, Sanchez O, Toledo JR, 2017, LOX-1 activation by oxLDL triggers an epithelial mesenchymal transition and promotes tumorigenic potential in prostate cancer cells. Cancer Lett 414:34–43. , DOI 10. 1016/j.canlet.2017.10.035 Li C, Zhang J,Wu H, Li L,Yang C, Song S, Peng P, ShaoM, Zhang M, Zhao J, Zhao R, Wu W, Ruan Y, Wang L, Gu J, 2017, Lectinlike oxidized low-density lipoprotein receptor-1 facilitates metastasis of gastric cancer through driving epithelial-mesenchymal transition and PI3K/Akt/GSK3beta activation. Sci Rep 7:45275. https:// doi.org/10.1038/srep45275 Murdocca M, Mango R, Pucci S, Biocca S, Testa B, Capuano R, Paolesse R, Sanchez M, Orlandi A, di Natale C, Novelli G, Sangiuolo F, 2016, The lectin-like oxidized LDL receptor-1: a new potential molecular target in colorectal cancer. Oncotarget 7(12):14765–14780. , DOI 10.18632/oncotarget.7430 Fujita Y, Kakino A, Nishimichi N, Yamaguchi S, Sato Y, Machida S, Cominacini L, Delneste Y, Matsuda H, Sawamura T, 2009, Oxidized LDL receptor LOX-1 binds to C-reactive protein and mediates its vascular effects. Clin Chem 55(2):285–294. https:// doi.org/10.1373/clinchem.2008.119750 Rizzacasa B, Morini E, Pucci S, Murdocca M, Novelli G, Amati F, 2017, LOX-1 and its splice variants: a new challenge for atherosclerosis and Cancer-targeted therapies. Int J Mol Sci 18, 2)., DOI , DOI 10.3390/Ijms18020290 Artn 290 Zampino MG, Magni E, Ravenda PS, Cella CA, Bonomo G, Della Vigna P, Galdy S, Spada F, Varano GM, Mauri G, Fazio N, Orsi F, 2016, Treatments for colorectal liver metastases: a new focus on a familiar concept. Crit Rev OncolHematol 108:154–163. https://doi. org/10.1016/j.critrevonc.2016.11.005 Heikkila K, Ebrahim S, Rumley A, Lowe G, Lawlor DA, 2007, Associations of circulating C-reactive protein and interleukin-6 with survival in women with and without cancer: findings from the British Women's heart and health study. Cancer Epidemiol Biomark Prev 16(6):1155–1159. , DOI 10.1158/1055- 9965.EPI-07-0093 Trichopoulos D, Psaltopoulou T, Orfanos P, Trichopoulou A, Boffetta P, 2006, Plasma C-reactive protein and risk of cancer: a prospective study from Greece. Cancer Epidemiol Biomark Prev 15(2):381–384. , DOI 10.1158/1055-9965.EPI-05-0626 Heikkila K, Ebrahim S, Lawlor DA, 2007, A systematic review of the association between circulating concentrations of C reactive protein and cancer. J Epidemiol Community Health 61(9):824– 833. , DOI 10.1136/jech.2006.051292 Heikkila K, Harris R, Lowe G, Rumley A, Yarnell J, Gallacher J, Ben-Shlomo Y, Ebrahim S, Lawlor DA, 2009, Associations of circulating C-reactive protein and interleukin-6 with cancer risk: findings from two prospective cohorts and a meta-analysis. Cancer Causes Control 20(1):15–26. , DOI 10.1007/ s10552-008-9212-z Thomsen M, Kersten C, Sorbye H, Skovlund E, Glimelius B, Pfeiffer P, Johansen JS, Kure EH, Ikdahl T, Tveit KM, Christoffersen T, Guren TK, 2016, Interleukin-6 and C-reactive protein as prognostic biomarkers in metastatic colorectal cancer. Oncotarget 7(46):75013–75022. , DOI 10.18632/ oncotarget.12601 Woo HD, Kim K, Kim J, 2015, Association between preoperative C-reactive protein level and colorectal cancer survival: a meta-analysis. Cancer Causes Control 26(11):1661–1670. , DOI 10. 1007/s10552-015-0663-8 Lu JJ, Mitra S, Wang XW, Khaidakov M, Mehta JL, 2011, Oxidative stress and lectin-like ox-LDL-receptor LOX-1 in Atherogenesis and tumorigenesis. Antioxid Redox Signal 15(8): 2301–2333. , DOI 10.1089/ars.2010.3792 Zhang J, Zhang L, Li C, Yang C, Li L, Song S,Wu H, Liu F,Wang L, Gu J, 2017, LOX-1 is a poor prognostic indicator and induces epithelial-mesenchymal transition and metastasis in pancreatic cancer patients. Cell Oncol, Dordr, 41:73–84. , DOI 10.1007/ s13402-017-0360-6 Pakdel A,Naghibalhossaini F, Mokarram P, JaberipourM, Hosseini A, 2012, Regulation of carcinoembryonic antigen release from colorectal cancer cells. Mol Biol Rep 39(4):3695–3704. https://doi. org/10.1007/s11033-011-1144-0 Naghibalhossaini F, Ebadi P, 2006, Evidence for CEA release from human colon cancer cells by an endogenous GPI-PLD enzyme. Cancer Lett 234(2):158–167. , DOI 10.1016/j.canlet. 2005.03.028 Kim YW, Ko YT, Kim NK, Chung HC, Min BS, Lee KY, Park JP, Kim H, 2010, A comparative study of protein expression in primary colorectal cancer and synchronous hepatic metastases: the significance of matrix metalloproteinase-1 expression as a predictor of liver metastasis. Scand J Gastroenterol 45(2):217–225. https://doi. org/10.3109/00365520903453158 Zhu XH,Wang JM, Yang SS,Wang FF, Hu JL, Xin SN,Men H, Lu GF, Lan XL, Zhang D, Wang XY, Liao WT, Ding YQ, Liang L, 2017, Down-regulation of DAB2IP promotes colorectal cancer invasion and metastasis by translocating hnRNPK into nucleus to enhance the transcription of MMP2. Int J Cancer 141(1):172–183. , DOI 10.1002/ijc.30701 Hirsch HA, Iliopoulos D, Joshi A, Zhang Y, Jaeger SA, Bulyk M, Tsichlis PN, Shirley Liu X, Struhl K, 2010, A transcriptional signature and common gene networks link cancer with lipid metabolism and diverse human diseases. Cancer Cell 17(4):348–361. , DOI 10.1016/j.ccr.2010.01.022 Wang TC, Cardiff RD, Zukerberg L, Lees E, Arnold A, Schmidt EV, 1994, Mammary hyperplasia and carcinoma in MMTV-cyclin D1 transgenic mice. Nature 369(6482):669–671. , DOI 10. 1038/369669a0 Ninomiya I, Yonemura Y, Matsumoto H, Sugiyama K, Kamata T, Miwa K, Miyazaki I, Shiku H, 1991, Expression of c-myc gene product in gastric carcinoma. Oncology 48(2):149–153 Wang W, Xue L, Wang P, 2011, Prognostic value of beta-catenin, c-myc, and cyclin D1 expressions in patients with esophageal squamous cell carcinoma. Med Oncol 28(1):163–169. , DOI 10.1007/s12032-010-9436-0 Lee KS, KwakY, NamKH, KimDW, Kang SB, Choe G, KimWH, Lee HS, 2016, Favorable prognosis in colorectal cancer patients with co-expression of c-MYC and ss-catenin. BMC Cancer 16(1): 730. , DOI 10.1186/s12885-016-2770-7 Chu YC, Huang KT, 2016, CRP/oxLDL co-incubates impair endothelial functions through CD32, LOX-1, and keratin 1 with dependence on their ratio. J Taiwan Inst Chem E 64:16–21. https:// doi.org/10.1016/j.jtice.2016.03.048 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1467 EP 1475 DI 10.1007/s12253-018-0507-4 PG 9 ER PT J AU Zajkowska, M Lubowicka, E Fiedorowicz, W Szmitkowski, M Jamiolkowski, J Lawicki, S AF Zajkowska, Monika Lubowicka, Emilia Fiedorowicz, Wojciech Szmitkowski, Maciej Jamiolkowski, Jacek Lawicki, Slawomir TI Human Plasma Levels of VEGF-A, VEGF-C, VEGF-D, their Soluble Receptor - VEGFR-2 and Applicability of these Parameters as Tumor Markers in the Diagnostics of Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE VEGF family members; Diagnostic utility; AUC; ROC ID VEGF family members; Diagnostic utility; AUC; ROC AB VEGF family members are important factors in promoting angio- and lymphangiogenesis. The aim of this study was to investigate concentrations, diagnostic utility and power of VEGF-A, VEGF-C, VEGF-D and VEGFR-2 in comparison to CA15–3 in breast cancer (BC) patients. The study included 120 BC patients and 60 control patients (28 with benign breast tumors and 32 healthy women). Plasma levels of tested parameters were determined by ELISA, CA15–3 by CMIA. Concentrations of all parameters showed statistical significance when compared BC patients to controls. VEGF-D showed the highest SE (82.50%) in total BC group. Highest SP and PPV in total BC group showed VEGF-A(76.67%;84.78%,respectively), but lower than CA15–3. Highest NPV showed VEGF-C(52.33%), but it was lower than CA15–3. VEGF-C was also the best parameter which had statistically significant AUC in total cancer group (0.7672), but also stages I(0.7684) and II(0.7772). In the total group of BC almost all tested parameters showed statistically significant AUC, but a maximum range was obtained for the combination of VEGF-C + CA15–3(0.8476). The combined analysis of tested parameters and CA15–3 resulted in increase in SE and AUC values, which provides hope for developing a new panel of biomarkers that may be used in the diagnosis of BC in the future. C1 [Zajkowska, Monika] Medical University of Bialystok, Department of Biochemical Diagnostics, Waszyngtona 15A, 15-269 Bialystok, Poland. [Lubowicka, Emilia] Medical University of Bialystok, Department of Esthetic Medicine, 15-267 Bialystok, Poland. [Fiedorowicz, Wojciech] Maria Sklodowska-Curie Oncology Center, Department of Oncological Surgery, 15-001 Bialystok, Poland. [Szmitkowski, Maciej] Medical University of Bialystok, Department of Biochemical Diagnostics, Waszyngtona 15A, 15-269 Bialystok, Poland. [Jamiolkowski, Jacek] Medical University of Bialystok, Department of Population Medicine and Civilization Diseases Prevention, 15-269 Bialystok, Poland. [Lawicki, Slawomir] Medical University of Bialystok, Department of Biochemical Diagnostics, Waszyngtona 15A, 15-269 Bialystok, Poland. RP Lawicki, S (reprint author), Medical University of Bialystok, Department of Biochemical Diagnostics, 15-269 Bialystok, Poland. EM slawicki@umb.edu.pl CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108 Oeffinger KC, Fontham ET, Etzioni R, Herzig A, Michaelson JS, Shih YC, Walter LC, Church TR, Flowers CR, LaMonte S, Wolf AM, DeSantis C, Lortet-Tieulent J, Andrews K, Manassaram- Baptiste D, Saslow D, Smith RA, Brawley OW, Wender R, American Cancer Society, 2015, Breast Cancer screening for women at average risk: 2015 guideline update from the American Cancer Society. JAMA 314(15):1599–1614 Siegel RL, Miller KD, Jemal A, 2018, Cancer statistics, 2018. CA Cancer J Clin 68(1):7–30 Zajkowska M, Glazewska EK, Bedkowska GE, Chorazy P, Szmitkowski M, Lawicki S, 2016, Diagnostic power of vascular endothelial growth factor and macrophage Colony-stimulating factor in breast Cancer patients based on ROC analysis. Mediat Inflamm 2016:5962946 Harris L, Fritsche H, Mennel R, Norton L, Ravdin P, Taube S, Somerfield MR, Hayes DF, Bast RC Jr, American Society of Clinical Oncology, 2007, American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J Clin Oncol 25(33):5287–5312 Lawicki S, Zajkowska M, Glazewska EK, Bedkowska GE, Szmitkowski M, 2016, Plasma levels and diagnostic utility of VEGF, MMP-9, and TIMP-1 in the diagnosis of patients with breast cancer. Onco Targets Ther 9:911–919 Egeblad M, Werb Z, 2002, New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2(3):161–174 Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N, 1989, Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 246(4935):1306–1309 Zyla MM, Kostrzewa M, Litwinska E, Szpakowski A, Wilczynski JR, Stetkiewicz T, 2014, The role of angiogenic factors in endometrial cancer. Prz Menopauzalny 13(2):122–126 Werynska B, Dziegiel P, Jankowska R, 2009, Role of lymphangiogenesis in lung cancer. Folia Histochem Cytobiol 47(3):333–342 Morfoisse F, Renaud E, Hantelys F, Prats AC, Garmy-Susini B, 2015, Role of hypoxia and vascular endothelial growth factors in lymphangiogenesis. Mol Cell Oncol 2(4):e1024821 Karkkainen MJ, Haiko P, Sainio K, Partanen J, Taipale J, Petrova TV, Jeltsch M, Jackson DG, Talikka M, Rauvala H, Betsholtz C, Alitalo K, 2004, Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol 5(1):74–80 Baranska P, Jerczynska H, Pawlowska Z, 2005, Vascular endothelial growth factor–structure and functions. Postepy Biochem 51(1): 12–21 Costache MI, Ioana M, Iordache S, Ene D, Costache CA, Saftoiu A, 2015, VEGF expression in pancreatic Cancer and other malignancies: a review of the literature. Rom J Intern Med 53(3):199–208 Kopfstein L, Veikkola T, Djonov VG, Baeriswyl V, Schomber T, Strittmatter K, Stacker SA, Achen MG, Alitalo K, Christofori G, 2007, Distinct roles of vascular endothelial growth factor-D in lymphangiogenesis andmetastasis. Am J Pathol 170(4):1348–1361 Della-Morte D, Riondino S, Ferroni P, Palmirotta R, Pastore D, Lauro D, Guadagni F, Roselli M, 2015, Impact of VEGF gene polymorphisms in elderly cancer patients: clinical outcome and toxicity. Pharmacogenomics 16(1):61–78 Stacker SA, Achen MG, 2008, From anti-angiogenesis to antilymphangiogenesis: emerging trends in cancer therapy. Lymphat Res Biol 6(3–4):165–172 Carmeliet P, Ruiz de Almodovar C, Carmen RA, 2013, VEGF ligands and receptors: implications in neurodevelopment and neurodegeneration. Cell Mol Life Sci 70(10):1763–1778 Caballero B, Sherman SJ, Falk T, 2017, Insights into the mechanisms involved in protective effects of VEGF-B in dopaminergic neurons. Parkinsons Dis 2017:4263795 Hicklin DJ, Ellis LM, 2005, Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol 23(5):1011–1027 Lawicki S, Bedkowska GE, Gacuta-Szumarska E, Szmitkowski M, 2013, The plasma concentration of VEGF, HE4 and CA125 as a new biomarkers panel in different stages and sub-types of epithelial ovarian tumors. J Ovarian Res 6(1):45 Bedkowska GE, Gacuta E, ZajkowskaM, Glazewska EK, Osada J, Szmitkowski M, Chrostek L, Dabrowska M, Lawicki S, 2017, Plasma levels of MMP-7 and TIMP-1 in laboratory diagnostics and differentiation of selected histological types of epithelial ovarian cancers. J Ovarian Res 10(1):39 Huang J, Li Y, Xue G, ZhangW, Li S, Zhang J et al, 2014, Value of VEGF-C, VEGF-D and VEGFR-3 levels combined with serum TSH in diagnosis of papillary thyroid carcinoma. Nan Fang Yi Ke Da Xue Xue Bao 34(12):1814–7, 21 Xia H, Shen J, Chen S, Huang H, Xu Y, Ma H, 2016, Overexpression of VEGF-C correlates with a poor prognosis in esophageal cancer patients. Cancer Biomark 17(2):165–170 Raica M, Cimpean AM, Ceausu R, Ribatti D, 2011, Lymphatic microvessel density, VEGF-C, and VEGFR-3 expression in different molecular types of breast cancer. Anticancer Res 31(5):1757– 1764 Thielemann A, Baszczuk A, Kopczynski Z, Kopczynski P, Grodecka-Gazdecka S, 2013, Clinical usefulness of assessing VEGF and soluble receptors sVEGFR-1 and sVEGFR-2 in women with breast cancer. Ann Agric Environ Med 20(2):293–297 Kotowicz B, Fuksiewicz M, Jonska-Gmyrek J, Berezowska A, Radziszewski J, Bidzinski M, Kowalska M, 2017, Clinical significance of pretreatment serum levels of VEGF and its receptors, IL- 8, and their prognostic value in type I and II endometrial cancer patients. PLoS One 12(10):e0184576 Jensen MR, Simonsen L, Karlsmark T, Lanng C, Bulow J, 2015, Higher vascular endothelial growth factor-C concentration in plasma is associated with increased forearm capillary filtration capacity in breast cancer-related lymphedema. Phys Rep 3(6):e12403 Kummel S, Eggemann H, Luftner D, Thomas A, Jeschke S, Zerfel N, Heilmann V, Emons G, Zeiser T, Ulm K, Kobl M, Korlach S, Schmid P, Sehouli J, Elling D, Blohmer JU, 2006, Changes in the circulating plasma levels of VEGF and VEGF-D after adjuvant chemotherapy in patients with breast cancer and 1 to 3 positive lymph nodes. Anticancer Res 26(2C):1719–1726 Wu DW, Chang WA, Liu KT, Yen MC, Kuo PL, 2017, Vascular endothelial growth factor and protein level in pleural effusion for differentiating malignant from benign pleural effusion. Oncol Lett 14(3):3657–3662 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1477 EP 1486 DI 10.1007/s12253-018-0527-0 PG 10 ER PT J AU Liu, H Wei, Sh Zhang, L Yuan, Ch Duan, Y Qingwei, W AF Liu, Hong Wei, Shufang Zhang, Lei Yuan, Chenxi Duan, Yuanyuan Qingwei, Wang TI Secreted Phosphoprotein 1 Promotes the Development of Small Cell Lung Cancer Cells by Inhibiting Autophagy and Apoptosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SPP1; Small cell lung cancer cells; Autophagy; Apoptosis ID SPP1; Small cell lung cancer cells; Autophagy; Apoptosis AB This study aimed to investigate the expression of secreted phosphoprotein 1 (SPP1) on lung cancer cells and explore its underlying mechanism on autophagy and apoptosis which effect the development of lung cancer cells. GSE19804 related to lung cancer cells was screened from Gene Expression Omnibus (GEO) database, and we screened the 47 pairs of differential expressed mRNAs in lung cancer cells and adjacent tissues using microarray analysis. The expression of the core gene SPP1 was detected by qRT-PCR and western-blot. The transfection efficiency of lung cancer cells was detected by qRT-PCR and the expression of transfected group was tested by western-blot. Cell proliferation after transfection was tested by MTT assay and plate cloning experiment. The apoptosis rate of each transfection group was detected by flow cytometry.We use western-blot to test protein expression of autophagy-related proteins Beclin-1, LC3-I, LC3-II and p62 of each transfected group. Through analysis of GSE19804,the heat map showed SPP1 was the highest expressed in tumor tissues. qRT-PCR and western-blot detected SPP1 expression in lung cancer tissues was higher than that in normal adjacent tissues and was significantly increased in lung cancer cell lines. After transfection with pcDNA3.1-SPP1 (p-SPP1 group), siRNA1-SPP1 (siRNA1 group) and siRNA2- SPP1 (siRNA2 group), showed different expression of SPP1. Up-regulation of SPP1 enhanced cell viability and promoted tumor cell proliferation, while knockdown of SPP1 inhibited tumor cell proliferation. From the results of apoptosis rate, SPP1 inhibited the tumor cell apoptosis. However, in normal lung cell, SPP1 had no effect on cell proliferation and apoptosis. And to test autophagy-related proteins, we found that overexpression of SPP1 inhibited autophagy. High expression of SPP1 inhibited autophagy and apoptosis to promote the development of small cell lung cancer cells. C1 [Liu, Hong] Qilu Hospital of Shandong University, Cancer Research Center, No. 107 Cultural West Road, 250012 Jinan, Shandong, China. [Wei, Shufang] Qilu Hospital of Shandong University, No.2 Comprehensive Department, 250012 Jinan, Shandong, China. [Zhang, Lei] Taian City Central Hospital, Department of Thoracic Surgery, 271000 Taian, Shandong, China. [Yuan, Chenxi] Qilu Hospital of Shandong University, Cancer Research Center, No. 107 Cultural West Road, 250012 Jinan, Shandong, China. [Duan, Yuanyuan] Qilu Hospital of Shandong University, Cancer Research Center, No. 107 Cultural West Road, 250012 Jinan, Shandong, China. [Qingwei, Wang] Qilu Hospital of Shandong University, Cancer Research Center, No. 107 Cultural West Road, 250012 Jinan, Shandong, China. RP Qingwei, W (reprint author), Qilu Hospital of Shandong University, Cancer Research Center, 250012 Jinan, China. EM xjsnxwj@126.com CR Nanavaty P, Alvarez MS, Alberts WM(2014, Lung cancer screening: advantages, controversies, and applications. Cancer Control 21:9–14. , DOI 10.1177/107327481402100102 Hasan N, Kumar R, Kavuru MS, 2014, Lung cancer screening beyond low-dose computed tomography: the role of novel biomarkers. Lung 192:639–648. , DOI 10.1007/s00408-014- 9636-z Malhotra J, Malvezzi M, Negri E, La Vecchia C, Boffetta P, 2016, Risk factors for lung cancer worldwide. Eur Respir J 48:889–902. , DOI 10.1183/13993003.00359-2016 Zhu J, Zeng Y, Xu C, Qin H, Lei Z, Shen D, Liu Z, Huang JA, 2015, Expression profile analysis of microRNAs and downregulated miR-486-5p and miR-30a-5p in non-small cell lung cancer. Oncol Rep 34:1779–1786. , DOI 10.3892/or.2015.4141 He C, Klionsky DJ, 2009, Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet 43:67–93. , DOI 10.1146/annurev-genet-102808-114910 Nikoletopoulou V, Markaki M, Palikaras K, Tavernarakis N, 2013, Crosstalk between apoptosis, necrosis and autophagy. Biochim Biophys Acta 1833:3448–3459. , DOI 10.1016/j.bbamcr. 2013.06.001 Yang ZJ, Chee CE, Huang S, Sinicrope F, 2011, Autophagy modulation for cancer therapy. Cancer Biol Ther 11:169–176. https:// doi.org/10.4161/cbt.11.2.14663 Mathew R, Karantza-Wadsworth V, White E, 2007, Role of autophagy in cancer. Nat Rev Cancer 7:961–967. , DOI 10. 1038/nrc2254 Gordy C, He YW, 2012, The crosstalk between autophagy and apoptosis: where does this lead? Protein Cell 3:17–27. https://doi. org/10.1007/s13238-011-1127-x Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65:87–108. , DOI 10.3322/caac.21262 Heymann JJ, BulmanWA, Swinarski D, Pagan CA, Crapanzano JP, Haghighi M, Fazlollahi L, StooplerMB, Sonett JR, SacherAG, Shu CA, Rizvi NA, Saqi A, 2017, PD-L1 expression in non-small cell lung carcinoma: comparison among cytology, small biopsy, and surgical resection specimens. Cancer 125:896–907. , DOI 10.1002/cncy.21937 Gozuacik D, Kimchi A, 2004, Autophagy as a cell death and tumor suppressor mechanism. Oncogene 23:2891–2906. , DOI 10.1038/sj.onc.1207521 Ogier-Denis E, Codogno P, 2003, Autophagy: a barrier or an adaptive response to cancer. Biochim Biophys Acta 1603:113–128 Marino G, Niso-Santano M, Baehrecke EH, Kroemer G, 2014, Self-consumption: the interplay of autophagy and apoptosis. Nat Rev Mol Cell Biol 15:81–94. , DOI 10.1038/nrm3735 Marino G, Lopez-Otin C, 2004, Autophagy: molecular mechanisms, physiological functions and relevance in human pathology. Cell Mol Life Sci 61:1439–1454. , DOI 10.1007/s00018- 004-4012-4 Zhang M, Jiang M, Bi Y, Zhu H, Zhou Z, Sha J, 2012, Autophagy and apoptosis act as partners to induce germ cell death after heat stress inmice. PLoS One 7:e41412. , DOI 10.1371/journal. pone.0041412 Maiuri MC, Le Toumelin G, Criollo A, Rain JC, Gautier F, Juin P, Tasdemir E, Pierron G, Troulinaki K, Tavernarakis N, Hickman JA, Geneste O, Kroemer G, 2007, Functional and physical interaction between Bcl-X(L, and a BH3-like domain in Beclin-1. EMBO J 26: 2527–2539. , DOI 10.1038/sj.emboj.7601689 Wirawan E, Lippens S, Vanden Berghe T, Romagnoli A, Fimia GM, Piacentini M, Vandenabeele P, 2012, Beclin1: a role in membrane dynamics and beyond. Autophagy 8:6–17. , DOI 10. 4161/auto.8.1.16645 Tanida I, Ueno T, Kominami E, 2004, LC3 conjugation system in mammalian autophagy. Int J Biochem Cell Biol 36:2503–2518. , DOI 10.1016/j.biocel.2004.05.009 Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, Kominami E, Ohsumi Y, Yoshimori T, 2000, LC3, amammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J 19:5720–5728. , DOI 10. 1093/emboj/19.21.5720 Ichimura Y, Komatsu M, 2010, Selective degradation of p62 by autophagy. Semin Immunopathol 32:431–436. , DOI 10. 1007/s00281-010-0220-1 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1487 EP 1495 DI 10.1007/s12253-018-0504-7 PG 9 ER PT J AU Huang, HF Fan, XR Ji, ZG AF Huang, Hou-Feng Fan, Xin-Rong Ji, Zhi-Gang TI The Effectiveness of Sorafenib over Other Targeted Agents in the Second-Line Treatment of Metastatic Renal Cell Carcinoma: a Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sorafenib; Metastatic renal cell carcinoma; Second-line therapy; Meta-analysis ID Sorafenib; Metastatic renal cell carcinoma; Second-line therapy; Meta-analysis AB The aim of the study was to perform a meta-analysis to compare the therapeutic effects and adverse events (AEs) of sorafenib in second-line treatments of metastatic renal cell carcinoma (mRCC). We searched online electronic databases: Pubmed, Embase, Cochrane library updated on November 2017.Trials of the effectiveness of sorafenib in second-line treatments of advanced RCC were included, of which the main outcomes were objective response rate (ORR), progression-free survival (PFS), overall survival (OS) and grade 3/4 AE. Other TAs significantly reduced the risk of PFS compared to sorafenib with respect to second-line treatment (HR = 0.74; 95% CI, 0.65–0.83; p < 0.00001). No significant differences were, however, found in patients in terms of the ORR (HR = 1.82; 95% CI, 0.98–3.35; p = 0.06). Frequencies of the most common toxicities were overall similar and adverse events differed only in sensitivity analysis in rash with exclusion of other TAs (HR = 0.16; 95% CI, 0.05–0.52; p = 0.002). Overall survival was not debated between groups. In patients with mRCC, second-line sorafenib is associated with similar ORR as other target agents. While, sorafenib did not demonstrate a PFS advantage compared with other target agents, suggests sorafenib may not benefit patients with mRCC. Tolerability due to toxicities is similar compared sorafenib with other target agents. Further characterization of the RCC oncogenic pathway, and the ongoing clinical trials should help optimize the treatment option for second-line therapy of advanced renal cell carcinoma. C1 [Huang, Hou-Feng] Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Department of Urology, No.1 Shuaifuyuan, Dongcheng District, 100730 Beijing, China. [Fan, Xin-Rong] Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Department of Urology, No.1 Shuaifuyuan, Dongcheng District, 100730 Beijing, China. [Ji, Zhi-Gang] Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Department of Urology, No.1 Shuaifuyuan, Dongcheng District, 100730 Beijing, China. RP Ji, ZG (reprint author), Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Department of Urology, 100730 Beijing, China. EM jzhgpumch@163.com CR Eble JN, Sauter G, Epstein JI, 2004, Pathology and genetics of tumors of the urinary system and male genital organs. Lyon, France. IARC Press Skinner DG, Colvin RB, Vermillion CD, Pfister RC, Leadbetter WF, 1971, Diagnosis and management of renal cell carcinoma. A clinical and pathologic study of 309 cases. Cancer 28:1165–1177 Kumar R, Kapoor A, 2017, Current management of metastatic renal cell carcinoma: evolving new therapies. Curr Opin Support Palliat Care 11:231–237 Mariani RL, Jorge MP, Pereira SS, Melione LP, Carvalho-Oliveira R, Ma TH, Saldiva PH, 2009, Association between micronuclei frequency in pollen mother cells of Tradescantia and mortality due to cancer and cardiovascular diseases: a preliminary study in Sao Jose dos Campos, Brazil. Environ Pollut 157:1767–1770 Hutson TE, Escudier B, Esteban E, Bjarnason GA, Lim HY, Pittman KB, Senico P, Niethammer A, Lu DR, Hariharan S, Motzer RJ, 2014, Randomized phase III trial of temsirolimus versus sorafenib as second-line therapy after sunitinib in patients with metastatic renal cell carcinoma. J cElinical Oncol : Off J Am Soc Clin Oncol. 32:760–767 Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, Michaelson MD, Gorbunova VA, Gore ME, Rusakov IG, Negrier S, Ou YC, Castellano D, Lim HY, Uemura H, Tarazi J, Cella D, Chen C, Rosbrook B, Kim S, Motzer RJ, 2011, Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma, AXIS): a randomised phase 3 trial. Lancet 378:1931–1939 Iacovelli R, Verri E, Cossu Rocca M, Aurilio G, Cullura D, Santoni M, de Cobelli O, Nole F, 2016, Is there still a role for sorafenib in metastatic renal cell carcinoma? A systematic review and metaanalysis of the effectiveness of sorafenib over other targeted agents. Crit Rev Oncol Hematol 99:324–331 Heng DY, Signorovitch J, Swallow E, Li N, Zhong Y, Qin P, Zhuo DY, Wang X, Park J, Stergiopoulos S, Kollmannsberger C, 2014, Comparative effectiveness of second-line targeted therapies for metastatic renal cell carcinoma: a systematic review and metaanalysis of real-world observational studies. PLoS One 9:e114264 Borregales LD,AdibiM, ThomasAZ,Wood CG, KaramJA, 2016, The role of neoadjuvant therapy in the management of locally advanced renal cell carcinoma. Ther Adv Urol 8:130–141 Higgins JP, Thompson SG, 2002, Quantifying heterogeneity in a meta-analysis. Stat Med 21:1539–1558 Higgins JP, Thompson SG, Deeks JJ, Altman DG, 2003, Measuring inconsistency in meta-analyses. BMJ 327:557–560 Motzer RJ, Escudier B, Tomczak P, Hutson TE, Michaelson MD, Negrier S, Oudard S, Gore ME, Tarazi J, Hariharan S, Chen C, Rosbrook B, Kim S, Rini BI, 2013, Axitinib versus sorafenib as second-line treatment for advanced renal cell carcinoma: overall survival analysis and updated results from a randomised phase 3 trial. Lancet Oncol 14:552–562 Qin S, Bi F, Jin J, Cheng Y, Guo J, Ren X, Huang Y, Tarazi J, Tang J, Chen C, Kim S, Ye D, 2015, Axitinib versus sorafenib as a second-line therapy in Asian patients with metastatic renal cell carcinoma: results from a randomized registrational study. Onco Targets Ther 8:1363–1373 Capri S, Porta C, Delea TE, 2017, Cost-effectiveness of Pazopanib versus Sunitinib as first-line treatment for locally advanced or metastatic renal cell carcinoma from an Italian National Health Service Perspective. Clin Ther 39:567–580 e2 Wilkerson J, Fojo T, 2009, Progression-free survival is simply a measure of a drug's effect while administered and is not a surrogate for overall survival. Cancer J 15:379–385 Booth CM, Eisenhauer EA, 2012, Progression-free survival: meaningful or simply measurable? J Clin Oncol : Off J Am Soc Clin Oncol 30:1030–1033 Kang SK, Volodarskiy A, Ohmann EL, Balar AV, Bangalore S, 2016, Efficacy and safety of selective vascular endothelial growth factor receptor inhibitors compared with Sorafenib for metastatic renal cell carcinoma: a meta-analysis of randomised controlled trials. Clin Oncol 28:334–341 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1497 EP 1503 DI 10.1007/s12253-018-0516-3 PG 7 ER PT J AU Sipos, F Kiss, LA Constantinovits, M Tulassay, Zs Muzes, Gy AF Sipos, Ferenc Kiss, L Anna Constantinovits, Miklos Tulassay, Zsolt Muzes, Gyorgyi TI Modified Genomic Self-DNA Influences In Vitro Survival of HT29 Tumor Cells via TLR9- and Autophagy Signaling SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HT29; Colon cancer; TLR9; Autophagy; Colonosphere; Self-DNA ID HT29; Colon cancer; TLR9; Autophagy; Colonosphere; Self-DNA AB In relation of immunobiology, the consequence of the crosstalk between TLR9-signaling and autophagy is poorly documented in HT29 cancer cells. To assess the TLR9-mediated biologic effects ofmodified self-DNA sequences on cell kinetics and autophagy response HT29 cells were incubated separately with intact genomic (g), hypermethylated (m), fragmented (f), and hypermethylated/fragmented (m/f) self-DNAs. Cell viability, apoptosis, cell proliferation, colonosphere-formation were determined. Moreover, the relation of TLR9-signaling to autophagy response was assayed by real-time RT-PCR, immunocytochemistry and transmission electron microscopy (TEM). After incubation with g-, m-, and m/f-DNAs cell viability and proliferation decreased, while apoptosis increased. F-DNA treatment resulted in an increase of cell survival.Methylation of self-DNA resulted in decrease of TLR9 expression, while it did not influence the positive effect of DNA fragmentation on MyD88 and TRAF6 overexpression, and TNFα downregulation. Fragmentation of DNA abrogated the positive effect of methylation on IRAK2, NFκB and IL-8 mRNA upregulations. In case of the autophagy genes and proteins, g- and f-DNAs caused significant upregulation of Beclin1, Atg16L1, and LC3B. According to TEM analyses, autophagy was present in each group of tumor cells, but to a varying degree. Incubation with m-DNA suppressed tumor cell survival by inducing features of apoptotic cell death, and activated mitophagy. F-DNA treatment enhanced cell survival, and activated macroautophagy and lipophagy. Colonospheres were only present after m-DNA incubation. Our data provided evidence for a close existing interplay between TLR9-signaling and the autophagy response with remarkable influences on cell survival in HT29 cells subjected to modified self-DNA treatments. C1 [Sipos, Ferenc] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Kiss, L Anna] Semmelweis University, 1st Department of Anatomy, 1094 Budapest, Hungary. [Constantinovits, Miklos] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Tulassay, Zsolt] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. [Muzes, Gyorgyi] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi street 46, 1088 Budapest, Hungary. RP Sipos, F (reprint author), Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. EM dr.siposf@gmail.com CR Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674. , DOI 10.1016/j.cell.2011. .013 2. Yu L, Chen S, 2008, Toll-like receptors expressed in tumor cells: targets for therapy. Cancer Immunol Immunother 57:1271–1278. , DOI 10.1007/s00262-008-0459-8 Alexopoulou L, Holt AC, Medzhitov R, Flavell RA, 2001, Recognition of double-stranded RNA and activation of NFkappaB by Toll-like receptor 3. Nature 413:732–738 Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, Lipford G, Wagner H, Bauer S, 2004, Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science 303:1526–1529 Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, Matsumoto M, Hoshino K, Wagner H, Takeda K, Akira S, 2000, AToll-like receptor recognizes bacterial DNA. Nature 408:740–745 Takeshita F, Leifer CA, Gursel I, Ishii KJ, Takeshita S, Gursel M, Klinman DM, 2001, Cutting edge: Role of Toll-like receptor 9 in CpG DNA-induced activation of human cells. J Immunol 167:3555–3558 Krieg AM, 2002, CpG motifs in bacterial DNA and their immune effects. Annu Rev Immunol 20:709–760 Latz E, Schoenemeyer A, Visintin A, Fitzgerald KA, Monks BG, Knetter CF, Lien E, Nilsen NJ, Espevik T, Golenbock DT, 2004, TLR9 signals after translocating from the ER to CpG DNA in the lysosome. Nat Immunol 5:190–198 Blasius AL, Beutler B, 2010, Intracellular toll-like receptors. Immunity 32:305–315. , DOI 10.1016/j.immuni.2010. 03.012 Marshak-Rothstein A, Rifkin IR, 2007, Immunologically active autoantigens: the role of toll-like receptors in the development of chronic inflammatory disease. Annu Rev Immunol 25:419–441 Li X, Jiang S, Tapping RI, 2010, Toll-like receptor signaling in cell proliferation and survival. Cytokine 49:1–9. , DOI 10. 1016/j.cyto.2009.08.010. Vallabhapurapu S, KarinM, 2009, Regulation and function of NFkappaB transcription factors in the immune system. Annu Rev Immunol 27:693–733. , DOI 10.1146/annurev.immunol. 021908.132641 Pedersen G, Andresen L, Matthiessen MW, Rask-Madsen J, Brynskov J, 2005, Expression of Toll-like receptor 9 and response to bacterial CpG oligodeoxynucleotides in human intestinal epithelium. Clin Exp Immunol 141:298–306 Leon SA, Shapiro B, Sklaroff DM, Yaros MJ, 1977, Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 37:646–650 Sozzi G, Conte D, Mariani L, Lo Vullo S, Roz L, Lombardo C, Pierotti MA, Tavecchio L, 2001, Analysis of circulating tumor DNA in plasma at diagnosis and during follow-up of lung cancer patients. Cancer Res 61:4675–4678 Mouliere F, Robert B, Arnau Peyrotte E, Del Rio M, Ychou M, Molina F, Gongora C, Thierry AR, 2011, High fragmentation characterizes tumour-derived circulating DNA. PLoS One 6:e23418. , DOI 10.1371/journal.pone.0023418 Copeland RA, Olhava EJ, Scott MP, 2010, Targeting epigenetic enzymes for drug discovery. Curr Opin Chem Biol 14:505–510. , DOI 10.1016/j.cbpa.2010.06.174 Ivanov S, Dragoi AM, Wang X, Dallacosta C, Louten J, Musco G, Sitia G, Yap GS, Wan Y, Biron CA, Bianchi ME, Wang H, Chu WM, 2007, A novel role for HMGB1 in TLR9-mediated inflammatory responses to CpG-DNA. Blood 110:1970–1981 Tian J, Avalos AM, Mao SY, Chen B, Senthil K, Wu H, Parroche P, Drabic S, Golenbock D, Sirois C, Hua J, An LL, Audoly L, La Rosa G, Bierhaus A, Naworth P, Marshak- Rothstein A, Crow MK, Fitzgerald KA, Latz E, Kiener PA, Coyle AJ, 2007, Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat Immunol 8:487–496 Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, 2001, Knippers R. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 61:1659–1665 Tuomela J, Sandholm J, Kaakinen M, Patel A, Kauppila JH, Ilvesaro J, Chen D, Harris KW, Graves D, Selander KS, 2013, DNA from dead cancer cells induces TLR9-mediated invasion and inflammation in living cancer cells. Breast Cancer Res Treat 142:477–487. , DOI 10.1007/s10549-013-2762-0 Moga E, Alvarez E, Canto E, Vidal S, Rodriguez-Sanchez JL, Sierra J, Briones J, 2008, NK cells stimulated with IL-15 or CpG ODN enhance rituximab-dependent cellular cytotoxicity against Bcell lymphoma. Exp Hematol 36:69–77 Vollmer J, Krieg AM, 2009, Immunotherapeutic applications of CpG oligodeoxynucleotide TLR9 agonists. Adv Drug Deliv Rev 61:195–204. , DOI 10.1016/j.addr.2008.12.008 Furi I, Sipos F, Spisak S, Kiszner G, Wichmann B, Scholler A, Tulassay Z, Muzes G, Molnar B, 2013, Association of self-DNA mediated TLR9-related gene, DNA methyltransferase, and cytokeratin protein expression alterations in HT29-cells to DNA fragment length and methylation status. ScientificWorldJournal 2013:293296. , DOI 10.1155/2013/293296 Levine B, Klionsky DJ, 2004, Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell 6:463–477 Levine B, Deretic V, 2007, Unveiling the roles of autophagy in innate and adaptive immunity. Nat Rev Immunol 7:767–777 Mizushima N, Levine B, Cuervo AM, Klionsky DJ, 2008, Autophagy fights disease through cellular self-digestion. Nature 451:1069–1075. , DOI 10.1038/nature06639 Levine B, Kroemer G, 2008, Autophagy in the pathogenesis of disease. Cell 132:27–42. , DOI 10.1016/j.cell.2007.12.018 Rabinowitz JD, White E, 2010, Autophagy and metabolism. Science 330:1344–1348. , DOI 10.1126/science.1193497 Levine B, 2007, Cell biology: autophagy and cancer. Nature 446: 745–747 Mathew R, Karantza-Wadsworth V, White E, 2007, Role of autophagy in cancer. Nat Rev Cancer 7:961–967 Dalby KN, Tekedereli I, Lopez-Berestein G, Ozpolat B, 2010, Targeting the prodeath and prosurvival functions of autophagy as novel therapeutic strategies in cancer. Autophagy 6:322–329 Lorin S, Hamai A, Mehrpour M, Codogno P, 2013, Autophagy regulation and its role in cancer. Semin Cancer Biol 23:361–379. , DOI 10.1016/j.semcancer.2013.06.007 Delgado MA, Elmaoued RA, Davis AS, Kyei G, Deretic V, 2008, Toll-like receptors control autophagy. EMBO J 27:1110–1121. , DOI 10.1038/emboj.2008.31 Sanjuan MA, Dillon CP, Tait SW, Moshiach S, Dorsey F, Connell S, Komatsu M, Tanaka K, Cleveland JL, Withoff S, Green DR, 2007, Toll-like receptor signalling in macrophages links the autophagy pathway to phagocytosis. Nature 450:1253–1257 Mehta P, Henault J, Kolbeck R, Sanjuan MA, 2014, Noncanonical autophagy: one small step for LC3, one giant leap for immunity. Curr Opin Immunol 26C:69–75. , DOI 10.1016/j.coi.2013. 10.012 Shin DM, Yuk JM, Lee HM, Lee SH, Son JW, Harding CV, Kim JM, Modlin RL, Jo EK, 2010, Mycobacterial lipoprotein activates autophagy via TLR2/1/CD14 and a functional vitamin D receptor signalling. Cell Microbiol 12:1648–1665. , DOI 10.1111/j. 1462-5822.2010.01497.x Deretic V, Saitoh T, Akira S, 2013, Autophagy in infection, inflammation and immunity. Nat Rev Immunol 13:722–737. https://doi. org/10.1038/nri3532 Into T, Inomata M, Takayama E, Takigawa T, 2012, Autophagy in regulation of Toll-like receptor signaling. Cell Signal 24:1150– 1162. , DOI 10.1016/j.cellsig.2012.01.020 Zhan Z, Xie X, Cao H, Zhou X, Zhang XD, Fan H, Liu Z, 2014, Autophagy facilitates TLR4- and TLR3-triggered migration and invasion of lung cancer cells through the promotion of TRAF6 ubiquitination. Autophagy 10:257–268. , DOI 10.4161/ auto.27162 Chen GY, Chen CL, Tuan HY, Yuan PX, Li KC, Yang HJ, Hu YC, 2014, Graphene oxide triggers toll-like receptors/autophagy responses in vitro and inhibits tumor growth in vivo. Adv Healthc Mater 3(9):1486–1495. , DOI 10.1002/adhm.201300591 Bertin S, Samson M, Pons C, Guigonis JM, Gavelli A, Baque P, Brossette N, Pagnotta S, Ricci JE, Pierrefite-Carle V, 2008, Comparative proteomics study reveals that bacterial CpG motifs induce tumor cell autophagy in vitro and in vivo. Mol Cell Proteomics 7:2311–2322. , DOI 10.1074/mcp.M800100- MCP200 Bertin S, Pierrefite-Carle V, 2008, Autophagy and toll-like receptors: a new link in cancer cells. Autophagy 4:1086–1089 Schmid D, Pypaert M, Munz C, 2007, Antigen-loading compartments for major histocompatibility complex class II molecules continuously receive input from autophagosomes. Immunity 26:79–92 Pradere JP, Dapito DH, Schwabe RF, 2014, The Yin and Yang of Toll-like receptors in cancer. Oncogene 33:3485–3395. https://doi. org/10.1038/onc.2013.302 Widlak P, Garrard WT, 2009, Roles of the major apoptotic nuclease-DNA fragmentation factor-in biology and disease. Cell Mol Life Sci 66:263–274. , DOI 10.1007/ s00018-008-8472-9 Guiducci C, Ott G, Chan JH, Damon E, Calacsan C, Matray T, Lee KD, Coffman RL, Barrat FJ, 2006, Properties regulating the nature of the plasmacytoid dendritic cell response to Toll-like receptor 9 activation. J Exp Med 203:1999–2008 Shimosato T, Kimura T, Tohno M, Iliev ID, Katoh S, Ito Y, Kawai Y, Sasaki T, Saito T, Kitazawa H, 2006, Strong immunostimulatory activity of AT-oligodeoxynucleotide requires a six-base loop with a self-stabilized 5'-C...G-3' stem structure. Cell Microbiol 8:485–495 Cortez-Gonzalez X, Pellicciotta I, Gerloni M, Wheeler MC, Castiglioni P, Lenert P, Zanetti M, 2006, TLR9-independent activation of B lymphocytes by bacterial DNA. DNA Cell Biol 25: 253–261 AkhtarM,Watson JL, Nazli A,McKay DM, 2003, Bacterial DNA evokes epithelial IL-8 production by a MAPK-dependent, NFkappaB- independent pathway. FASEB J 17:1319–1321 Khorramizadeh MR, Hosseinzadeh S, Safavifar F, Saadat F, Aalizadeh N, Falak R, Jadali Z, Pezeshki M, 2007, Interaction of CpG-oligodeoxynucleotides with Toll like receptor 9 induces apoptosis and modulates metaloproteinase-2 activity in human intestinal epithelium. Iran J Allergy Asthma Immunol 6(3):107–114 El Andaloussi A, Sonabend AM, Han Y, Lesniak MS, 2006, Stimulation of TLR9 with CpG ODN enhances apoptosis of glioma and prolongs the survival of mice with experimental brain tumors. Glia 54(6):526–535 Qi XF, Zheng L, Kim CS, Lee KJ, Kim DH, Cai DQ, Qin JW, Yu YH, Wu Z, Kim SK, 2013, CpG oligodeoxynucleotide induces apoptosis and cell cycle arrest in A20 lymphoma cells via TLR9- mediated pathways. Mol Immunol 54:327–337. , DOI 10. 1016/j.molimm.2013.01.001 Zhang Y, Lin A, Zhang C, Tian Z, Zhang J, 2014, Phosphorothioate-modified CpG oligodeoxynucleotide, CpG ODN, induces apoptosis of human hepatocellular carcinoma cells independent of TLR9. Cancer Immunol Immunother 63:357–367. , DOI 10.1007/s00262-014-1518-y Arunkumar N, Liu C, Hang H, Song W, 2013, Toll-like receptor agonists induce apoptosis in mouse B-cell lymphoma cells by altering NF-κB activation. Cell Mol Immunol 10:360–372. https://doi. org/10.1038/cmi.2013.14 Barnie PA, Zhang P, Lu P, Chen X, Su Z, Wang S, Xu H, 2014, CpG-oligodeoxynucleotides suppress the proliferation of A549 lung adenocarcinoma cells via toll-like receptor 9 signaling and upregulation of Runt-related transcription factor 3 expression. Biomed Rep 2(3):374–377 Ruckdeschel K, Mannel O, Schrottner P, 2002, Divergence of apoptosis-inducing and preventing signals in bacteria-faced macrophages through myeloid differentiation factor 88 and IL-1 receptorassociated kinase members. J Immunol 168(9):4601–4611 Xie K, 2001, Interleukin-8 and human cancer biology. Cytokine Growth Factor Rev 12:375–391 Ghadimi D, Md V, Heller KJ, Schrezenmeir J, 2010, Effect of natural commensal-origin DNA on toll-like receptor 9, TLR9, signaling cascade, chemokine IL-8 expression, and barrier integritiy of polarized intestinal epithelial cells. Inflamm Bowel Dis 16:410– 427. , DOI 10.1002/ibd.21057 Huang B, Zhao J, Li H, He KL, Chen Y, Chen SH, Mayer L, Unkeless JC, Xiong H, 2005, Toll-like receptors on tumor cells facilitate evasion of immune surveillance. Cancer Res 65:5009–5014 MerrellMA, Ilvesaro JM, Lehtonen N, Sorsa T, Gehrs B, Rosenthal E, Chen D, Shackley B, Harris KW, Selander KS, 2006, Toll-like receptor 9 agonists promote cellular invasion by increasing matrix metalloproteinase activity. Mol Cancer Res 4:437–447 Xu L, Wang C, Wen Z, Yao X, Liu Z, Li Q, Wu Z, Xu Z, Liang Y, Ren T, 2010, Selective up-regulation of CDK2 is critical for TLR9 signaling stimulated proliferation of human lung cancer cell. Immunol Lett 127:93–99. , DOI 10. 1016/j.imlet.2009.10.002 Kuo CC, Liang SM, Liang CM, 2006, CpG-B oligodeoxynucleotide promotes cell survival via up-regulation of Hsp70 to increase Bcl-xL and to decrease apoptosis-inducing factor translocation. J Biol Chem 281:38200–38207 Yao F, Han Q, Zhong C, Zhao H, 2013, TRAF6 promoted the tumorigenicity of esophageal squamous cell carcinoma. Tumour Biol 34:3201–3207. , DOI 10.1007/s13277-013-0890-0 Zhong L, Cao F, You Q, 2013, Effect of TRAF6 on the biological behavior of human lung adenocarcinoma cell. Tumour Biol 34: 231–239. , DOI 10.1007/s13277-012-0543-8 Peng Z, Shuangzhu Y, Yongjie J, Xinjun Z, 2013, Ying L. TNF receptor-associated factor 6 regulates proliferation, apoptosis, and invasion of glioma cells.Mol Cell Biochem 377:87–96. https://doi. org/10.1007/s11010-013-1573-2. Meng Q, Zheng M, Liu H, Song C, ZhangW, Yan J, Qin L, Liu X, 2012, TRAF6 regulates proliferation, apoptosis, and invasion of osteosarcoma cell.Mol Cell Biochem 371:177–186. , DOI 10.1007/s11010-012-1434-4 Yamashita M, Fatyol K, Jin C, Wang X, Liu Z, Zhang YE, 2008, TRAF6 mediates Smad-independent activation of JNK and p38 by TGF-beta. Mol Cell 31:918–924. , DOI 10.1016/j.molcel. 2008.09.002. Linares JF, Duran A, Yajima T, PasparakisM, Moscat J, Diaz-Meco MT, 2013, K63 polyubiquitination and activation of mTOR by the p62-TRAF6 complex in nutrient-activated cells. Mol Cell 51:283– 296. , DOI 10.1016/j.molcel.2013.06.020 Wei J, Yuan Y, Jin C, Chen H, Leng L, He F, Wang J, 2012, The ubiquitin ligase TRAF6 negatively regulates the JAK-STAT signaling pathway by binding to STAT3 and mediating its ubiquitination. PLoS One 7:e49567. , DOI 10.1371/journal.pone.0049567 Sun H, Li X, Fan L, Wu G, Li M, Fang J, 2014, TRAF6 is upregulated in colon cancer and promotes proliferation of colon cancer cells. Int J Biochem Cell Biol 53:195–201. , DOI 10.1016/ j.biocel.2014.04.010 Chuang TH, Ulevitch RJ, 2000, Cloning and characterization of a sub-family of human toll-like receptors: hTLR7, hTLR8 and hTLR9. Eur Cytokine Netw 11:372–378 McKelvey KJ, Highton J, Hessian PA, 2011, Cell-specific expression of TLR9 isoforms in inflammation. J Autoimmun 36:76–86. , DOI 10.1016/j.jaut.2010.11.001 Mochida K, Oikawa Y, Kimura Y, Kirisako H, Hirano H, Ohsumi Y, Nakatogawa H, 2015, Receptor-mediated selective autophagy degrades the endoplasmic reticulum and the nucleus. Nature 522: 359–362. , DOI 10.1038/nature14506 Behrends C, Sowa ME, Gygi SP, Harper JW, 2010, Network organization of the human autophagy system. Nature 466:68–76. , DOI 10.1038/nature09204 He C, Klionsky DJ, 2009, Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet 43:67–93. , DOI 10.1146/annurev-genet-102808-114910 Gordy C, He YW, 2012, The crosstalk between autophagy and apoptosis: where does this lead? Protein Cell 3(1):17–27. https:// doi.org/10.1007/s13238-011-1127-x Guertin DA, Sabatini DM, 2007, Defining the role of mTOR in cancer. Cancer Cell 12(1):9–22 Hamacher-Brady A, BradyNR, 2016, Mitophagy programs: mechanisms and physiological implications of mitochondrial targeting by autophagy. Cell Mol Life Sci 73:775–795. , DOI 10. 1007/s00018-015-2087-8 Radogna F, Cerella C, Gaigneaux A, Christov C, Dicato M, Diederich M, 2016, Cell type-dependent ROS and mitophagy response leads to apoptosis or necroptosis in neuroblastoma. Oncogene 35:3839–3853. , DOI 10.1038/onc.2015.455 Farias N, Ho N, Butler S, Delaney L, Morrison J, Shahrzad S, Coomber BL, 2015, The effects of folic acid on global DNA methylation and colonosphere formation in colon cancer cell lines. J Nutr Biochem 26:818–826. , DOI 10.1016/j.jnutbio.2015.02.002 Xue F, Hu L, Ge R, Yang L, Liu K, Li Y, Sun Y, Wang K, 2016, Autophagy-deficiency in hepatic progenitor cells leads to the defects of stemness and enhances susceptibility to neoplastic transformation. Cancer Lett 371:38–47. , DOI 10.1016/j.canlet.2015.11.022 Kantara C, O'ConnellM, Sarkar S, Moya S, Ullrich R, Singh P, 2014, Curcumin promotes autophagic survival of a subset of colon cancer stem cells, which are ablated by DCLK1-siRNA. Cancer Res 74:2487–2498. , DOI 10.1158/0008-5472. CAN-13-3536 Ojha R, Bhattacharyya S, Singh SK, 2015, Autophagy in Cancer Stem Cells: A Potential Link Between Chemoresistance, Recurrence, and Metastasis. Biores Open Access 4(1):97–108. , DOI 10.1089/biores.2014.0035 Gomez de Cedron M, Ramirez deMolina A, 2016)Microtargeting cancer metabolism: opening new therapeutic windows based on lipid metabolism. J Lipid Res 57(2):193–206. , DOI 10. 1194/jlr.R061812 Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M, Tanaka K, Cuervo AM, CzajaMJ, 2009, Autophagy regulates lipid metabolism. Nature 458:1131–1135. , DOI 10.1038/ nature07976 Johansen T, Lamark T, 2011, Selective autophagy mediated by autophagic adapter proteins. Autophagy 7:279–296 Amir M, Czaja MJ, 2011, Autophagy in nonalcoholic steatohepatitis. Expert Rev Gastroenterol Hepatol 5(2):159–166. , DOI 10.1586/egh.11.4 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1505 EP 1517 DI 10.1007/s12253-018-0544-z PG 13 ER PT J AU Kowalewski, A Szylberg, Tyloch, J Antosik, P Neska-Dlugosz, I Frackowski, Tyloch, D Purpurowicz, P Grzanka, D AF Kowalewski, Adam Szylberg, Lukasz Tyloch, Janusz Antosik, Paulina Neska-Dlugosz, Izabela Frackowski, Lukasz Tyloch, Dominik Purpurowicz, Piotr Grzanka, Dariusz TI Caspase 3 as a Novel Marker to Distinguish Chromophobe Renal Cell Carcinoma from Oncocytoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chromophobe; ChRCC; Oncocytoma; Caspase 3; CASP3; Cyclin D1; p16; Survivin; CD138; Ki-67 ID Chromophobe; ChRCC; Oncocytoma; Caspase 3; CASP3; Cyclin D1; p16; Survivin; CD138; Ki-67 AB Despite advances in our understanding of the biology of chromophobe renal cell carcinoma (ChRCC) and renal oncocytoma (RO), the differential diagnosis among these tumors remains one of the most problematic in renal pathology. Today, CK7 is the most recommended marker to distinguish these entities, however it appears insufficiently accurate by itself. This study aimed to find an easily accessible IHC stain that might out-compete CK7 in this field. Expressions of CK7, cyclin D1, p16, survivin, CD138, Ki-67 and caspase 3 (CASP3) were analyzed in a total of 27 cases (20 ROs and 7 ChRCCs). Immunoreactivity was assessed based on a combined score of the extent and intensity of staining. Compared to RO, a higher percentage of the total ChRCCs stained positive for CK7 (67% vs. 22%, respectively) and CASP3 (86% vs. 25%) (P < 0.005). The differences in staining with cyclin D1, p16, survivin, CD138 and Ki-67 turned out to be statistically insignificant in differentiating ChRCC from RO. CASP3 is a promising marker in distinguishing ChRCC from RO and may represent an alternative for CK7. Cyclin D1, p16, survivin, CD138 and Ki-67 cannot be used to distinguish these neoplasms. C1 [Kowalewski, Adam] Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Department of Clinical Pathomorphology, ul. Sklodowskiej-Curie Str. 9, 85-094 Bydgoszcz, Poland. [Szylberg, Lukasz] Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Department of Clinical Pathomorphology, ul. Sklodowskiej-Curie Str. 9, 85-094 Bydgoszcz, Poland. [Tyloch, Janusz] Nicolaus Copernicus University, Department of General and Oncologic UrologyBydgoszcz, Poland. [Antosik, Paulina] Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Department of Clinical Pathomorphology, ul. Sklodowskiej-Curie Str. 9, 85-094 Bydgoszcz, Poland. [Neska-Dlugosz, Izabela] Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Department of Clinical Pathomorphology, ul. Sklodowskiej-Curie Str. 9, 85-094 Bydgoszcz, Poland. [Frackowski, Lukasz] Nicolaus Copernicus University, Department of General and Oncologic UrologyBydgoszcz, Poland. [Tyloch, Dominik] Nicolaus Copernicus University, Department of General and Oncologic UrologyBydgoszcz, Poland. [Purpurowicz, Piotr] Nicolaus Copernicus University, Department of General and Oncologic UrologyBydgoszcz, Poland. [Grzanka, Dariusz] Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Department of Clinical Pathomorphology, ul. Sklodowskiej-Curie Str. 9, 85-094 Bydgoszcz, Poland. RP Kowalewski, A (reprint author), Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Department of Clinical Pathomorphology, 85-094 Bydgoszcz, Poland. EM kowalewskiresearch@gmail.com CR Differential diagnosis - renal oncocytoma - surgical pathology criteria - Stanford university school of medicine. at . Accessed 4 Dec 2017 Williamson SR, Gadde R, Trpkov K, Hirsch MS, Srigley JR, Reuter VE, Cheng L, Kunju LP, Barod R, Rogers CG, Delahunt B, Hes O, Eble JN, Zhou M, McKenney JK, Martignoni G, Fleming S, Grignon DJ, Moch H, Gupta NS, 2017, Diagnostic criteria for oncocytic renal neoplasms: a survey of urologic pathologists. Hum Pathol 63:149–156 Zokalj I, Marotti M, Kolaric B, 2014, Pretreatment differentiation of renal cell carcinoma subtypes by CT: the influence of different tumor enhancementmeasurement approaches. Int Urol Nephrol 46:1089–1100 MacLennan S, Imamura M, Lapitan MC, Omar MI, Lam TB, Hilvano-Cabungcal AM, Royle P, Stewart F, MacLennan G, MacLennan S, Canfield SE, McClinton S, Griffiths TR, Ljungberg B, N'Dow J, UCAN Systematic Review Reference Group, EAU Renal Cancer Guideline Panel, 2012, Systematic review of oncological outcomes following surgical Management of Localised Renal Cancer. Eur Urol 61:972–993 Bellmunt J, Dutcher J, 2013, Targeted therapies and the treatment of non-clear cell renal cell carcinoma. Ann Oncol 24:1730–1740 Bodnar M, Szylberg L, Kazmierczak W, Marszalek A, 2015, Tumor progression driven by pathways activating matrix metalloproteinases and their inhibitors. J Oral Pathol Med 44:437–443 Bodnar M, Luczak M, Bednarek K, Szylberg L, Marszalek A, Grenman R, Szyfter K, Jarmuz-Szymczak M, Giefing M, 2016, Proteomic profiling identifies the inorganic pyrophosphatase, PPA1, protein as a potential biomarker of metastasis in laryngeal squamous cell carcinoma. Amino Acids 48:1469–1476 Ng KL, Rajandram R, Morais C, Yap NY, Samaratunga H, Gobe GC,Wood ST, 2014, Differentiation of oncocytoma from chromophobe renal cell carcinoma, RCC): can novel molecular biomarkers help solve an old problem? J Clin Pathol 67:97–104 Liu L, Qian J, Singh H, Meiers I, Zhou X, Bostwick DG, 2007, Immunohistochemical analysis of chromophobe renal cell carcinoma, renal oncocytoma, and clear cell carcinoma: an optimal and practical panel for differential diagnosis. Arch Pathol LabMed 131:1290–1297 Durinck S, Stawiski EW, Pavia-Jimenez A, Modrusan Z, Kapur P, Jaiswal BS, Zhang N, Toffessi-Tcheuyap V, Nguyen TT, Pahuja KB, Chen YJ, Saleem S, Chaudhuri S, Heldens S, Jackson M, Pena-Llopis S, Guillory J, Toy K, Ha C, Harris CJ, Holloman E, HillHM, Stinson J, Rivers CS, Janakiraman V,WangW, Kinch LN, Grishin NV, Haverty PM, Chow B, Gehring JS, Reeder J, Pau G, Wu TD,Margulis V, Lotan Y, Sagalowsky A, Pedrosa I, de Sauvage FJ, Brugarolas J, Seshagiri S, 2015, Spectrum of diverse genomic alterations define non–clear cell renal carcinoma subtypes. Nat Genet 47:13–21 Davis CF, Ricketts CJ, Wang M, Yang L, Cherniack AD, Shen H, Buhay C, Kang H, Kim SC, Fahey CC, Hacker KE, Bhanot G, Gordenin DA, Chu A, Gunaratne PH, Biehl M, Seth S, Kaipparettu BA, Bristow CA, Donehower LA, Wallen EM, Smith AB, Tickoo SK, Tamboli P, Reuter V, Schmidt LS, Hsieh JJ, Choueiri TK, Hakimi AA, Chin L, Meyerson M, Kucherlapati R, Park WY, Robertson AG, Laird PW, Henske EP, Kwiatkowski DJ, Park PJ, Morgan M, Shuch B, Muzny D, Wheeler DA, Linehan WM, Gibbs RA, RathmellWK, Creighton CJ, Creighton CJ, Davis CF, Morgan M, Gunaratne PH, Donehower LA, Kaipparettu BA, Wheeler DA, Gibbs RA, Signoretti S, Cherniack AD, Robertson AG, Chu A, Choueiri TK, Henske EP, Kwiatkowski DJ, Reuter V, Hsieh JJ, Hakimi AA, Tickoo SK, Ricketts C, Linehan WM, Schmidt LS, Gordenin DA, Bhanot G, Seiler M, Tamboli P, Rathmell WK, Fahey CC, Hacker KE, Smith AB, Wallen EM, Shen H, Laird PW, Shuch B, Muzny D, Buhay C, Wang M, Chao H, Dahdouli M, Xi L, Kakkar N, Reid JG, Downs B, Drummond J, Morton D, Doddapaneni H, Lewis L, English A,Meng Q, Kovar C, Wang Q, Hale W, Hawes A, Kalra D, Walker K, Murray BA, Sougnez C, Saksena G, Carter SL, Schumacher SE, Tabak B, Zack TI, Getz G, Beroukhim R, Gabriel SB, Meyerson M, Ally A, Balasundaram M, Birol I, Brooks D, Butterfield YSN, Chuah E, Clarke A, Dhalla N, Guin R, Holt RA, Kasaian K, Lee D, Li HI, Lim E, Ma Y, Mayo M, Moore RA, Mungall AJ, Schein JE, Sipahimalani P, Tam A, Thiessen N, Wong T, Jones SJM, Marra MA, Auman JT, Tan D, Meng S, Jones CD, Hoadley KA, Mieczkowski PA, Mose LE, Jefferys SR, Roach J, Veluvolu U, Wilkerson MD, Waring S, Buda E, Wu J, Bodenheimer T, Hoyle AP, Simons JV, Soloway MG, Balu S, Parker JS, Hayes DN, Perou CM,Weisenberger DJ, BootwallaMS, Triche T Jr, Lai PH, van den Berg DJ, Baylin SB, Chen F, Coarfa C, Noble MS, DiCara D, Zhang H, Cho J, Heiman DI, Gehlenborg N, Voet D, Lin P, Frazer S, Stojanov P, Liu Y, Zou L, Kim J, Lawrence MS, Chin L, Yang L, Seth S, Bristow CA, Protopopov A, Song X, Zhang J, Pantazi A, Hadjipanayis A, Lee E, Luquette LJ, Lee S, Parfenov M, Santoso N, Seidman J, Xu AW, Kucherlapati R, Park PJ, Kang H, Lee J, Roberts SA, Klimczak LJ, Fargo D, Lang M, Choi YL, Kim SC, Lee JK, ParkWY,WangW, Fan Y, Ahn J, Akbani R,Weinstein JN, Haussler D, Ma S, Radenbaugh A, Zhul J, Biehl M, Lichtenberg TM, Zmuda E, Black AD, Hanf B, Ramirez NC, Wise L, Bowen J, Leraas KM, Hall TM, Gastier-Foster JM, Kaelin WG, Thorne L, Boice L, Huang M, Vocke C, Peterson J, Worrell R, Merino M, Czerniak BA, Aldape KD, Wood CG, Spellman PT, Atkins MB, Cheville J, Thompson RH, Jensen MA, Pihl T, Wan Y, Ayala B, Baboud J, Velaga S, Walton J, Liu J, Chudamani S, Wu Y, Sheth M, Mills Shaw KR, Demchok JA, Davidsen T, Yang L, Wang Z, Tarnuzzer RW, Zhang J, Eley G, Felau I, Zenklusen JC, Hutter CM, Guyer MS, Ozenberger BA, Sofia HJ, 2014, The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell 26:319–330 Gerlinger M, Rowan AJ, Horswell S, Math M, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, Varela I, Phillimore B, Begum S,McDonald N, Butler A, Jones D, Raine K, Latimer C, Santos CR, Nohadani M, Eklund AC, Spencer-Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA, Swanton C, 2012, Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366:883–892 Flanigan RC, Mickisch G, Sylvester R, Tangen C, van Poppel H, Crawford ED, 2004, Cytoreductive nephrectomy in patients with metastatic renal Cancer: a combined analysis. J Urol 171:1071–1076 Flanigan RC, Salmon SE, Blumenstein BA, Bearman SI, Roy V, McGrath PC, Caton JR Jr, Munshi N, Crawford ED, 2001, Nephrectomy followed by interferon alfa-2b compared with interferon alfa-2b alone for metastatic renal-cell Cancer. N Engl J Med 345:1655–1659 AdamAC, Scriba A, OrtmannM, Huss S, Kahl P, Steiner S, Storkel S, Buttner R, 2015, Immunohistochemical analysis of cytochrome c oxidase facilitates differentiation between Oncocytoma and Chromophobe renal cell carcinoma. Appl Immunohistochem Mol Morphol 23:54–59 Ohe C, Kuroda N, Takasu K, Senzaki H, Shikata N, Yamaguchi T, Miyasaka C, Nakano Y, Sakaida N, Uemura Y, 2012, Utility of immunohistochemical analysis of KAI1, epithelial-specific antigen, and epithelial-related antigen for distinction of chromophobe renal cell carcinoma, an eosinophilic variant from renal oncocytoma. Med Mol Morphol 45:98–104 Memeo L, Jhang J, Assaad AM, Mckiernan JM, Murty VVVS, Hibshoosh H, Tong GX, Mansukhani MM, 2007, Immunohistochemical analysis for cytokeratin 7, KIT, and PAX2. Am J Clin Pathol 127:225–229 Leroy X,Moukassa D, CopinMC, Saint F,Mazeman E, Gosselin B, 2000, Utility of cytokeratin 7 for distinguishing chromophobe renal cell carcinoma from renal oncocytoma. Eur Urol 37:484–487 Mathers ME, Pollock AM, Marsh C, O’Donnell M, 2002, Cytokeratin 7: a useful adjunct in the diagnosis of chromophobe renal cell carcinoma. Histopathology 40:563–567 Geramizadeh B, Ravanshad M, Rahsaz M, 2008, Useful markers for differential diagnosis of oncocytoma, chromophobe renal cell carcinoma and conventional renal cell carcinoma. Indian J Pathol Microbiol 51:167–171 Adley BP, Papavero V, Sugimura J, Teh BT, Yang XJ, 2006, Diagnostic value of cytokeratin 7 and parvalbumin in differentiating chromophobe renal cell carcinoma from renal oncocytoma. Anal Quant Cytol Histol 28:228–236 Wu SL, Kothari P, Wheeler TM, Reese T, Connelly JH, 2002, Cytokeratins 7 and 20 immunoreactivity in Chromophobe renal cell carcinomas and renal Oncocytomas. Mod Pathol 15:712–717 Bing Z, Lal P, Lu S, Ziober A, Tomaszewski JE, 2013, Role of carbonic anhydrase IX, α-methylacyl coenzyme a racemase, cytokeratin 7, and galectin-3 in the evaluation of renal neoplasms: a tissue microarray immunohistochemical study. Ann Diagn Pathol 17:58–62 Huang W, Kanehira K, Drew S, Pier T, 2009, Oncocytoma can be differentiated from its renal cell carcinoma mimics by a panel of markers. Appl Immunohistochem Mol Morphol 17:12–17 Garcia E, Li M, 2006, Caveolin-1 Immunohistochemical analysis in differentiating Chromophobe renal cell carcinoma from renal Oncocytoma. Am J Clin Pathol 125:392–398 Musgrove EA, Caldon CE, Barraclough J, Stone A, Sutherland RL, 2011, Cyclin D as a therapeutic target in cancer. Nat Rev Cancer 11:558–572 Romagosa C, Simonetti S, Lopez-Vicente L, Mazo A, LleonartME, Castellvi J, Ramon y Cajal S, 2011, p16Ink4a overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors. Oncogene 30:2087–2097 Garg H, Suri P, Gupta JC, Talwar GP, Dubey S, 2016, Survivin: a unique target for tumor therapy. Cancer Cell Int 16(49):49 Gotte M, Kersting C, Radke I, Kiesel L, Wulfing P, 2007, An expression signature of syndecan-1, CD138), E-cadherin and c-met is associated with factors of angiogenesis and lymphangiogenesis in ductal breast carcinoma in situ. Breast Cancer Res 9:R8 Shimada K, Nakamura M, de Velasco MA, Tanaka M, Ouji Y, Miyake M, Fujimoto K, Hirao K, Konishi N, 2010, Role of syndecan-1, CD138, in cell survival of human urothelial carcinoma. Cancer Sci 101:155–160 Richard PO, Jewett MAS, Bhatt JR, Evans AJ, Timilsina N, Finelli A, 2016, Active surveillance for renal neoplasms with Oncocytic features is safe. J Urol 195:581–586 Li L, Jiang G, Chen Q, Zheng J, 2014, Ki67 is a promising molecular target in the diagnosis of cancer, review). Mol Med Rep 11: 1566–1572 Flanagan L, Meyer M, Fay J, Curry S, Bacon O, Duessmann H, John K, Boland KC, McNamara DA, Kay EW, Bantel H, Schulze- Bergkamen H, Prehn JHM, 2016, Low levels of Caspase-3 predict favourable response to 5FU-based chemotherapy in advanced colorectal cancer: Caspase-3 inhibition as a therapeutic approach. Cell Death Dis 7:e2087 Li F et al, 2010, Apoptotic cells activate the "phoenix rising" pathway to promote wound healing and tissue regeneration. Sci Signal 3:ra13–ra13 Hu Q, Peng J, LiuW, He X, Cui L, Chen X, YangM, Liu H, Liu S, WangH, 2014, Elevated cleaved caspase-3 is associatedwith shortened overall survival in several cancer types. Int J Clin Exp Pathol 7:5057–5070 Huang Q, Li F, Liu X, Li W, Shi W, Liu FF, O'Sullivan B, He Z, Peng Y, Tan AC, Zhou L, Shen J, Han G, Wang XJ, Thorburn J, Thorburn A, Jimeno A, Raben D, Bedford JS, Li CY, 2011, Caspase 3–mediated stimulation of tumor cell repopulation during cancer radiotherapy. Nat Med 17:860–866 Takata T, Tanaka F, Yamada T, Yanagihara K, Otake Y, Kawano Y, Nakagawa T, Miyahara R, Oyanagi H, Inui K, Wada H, 2001, Clinical significance of caspase-3 expression in pathologic-stage I, nonsmall-cell lung cancer. Int J Cancer 96:54 Dunnick, R., Sandler, C. M. & Newhouse, J. H. Textbook of Uroradiology., Wolters Kluwer Health, 2012). at . Accessed 4 Dec 2017 Van der Kwast T, Perez-Ordonez B, 2007, Renal oncocytoma, yet another tumour that does not fit in the dualistic benign/malignant paradigm? J Clin Pathol 60:585–586 Hes O, Petersson F, Kuroda N, Hora M, Michal M, 2013, Renal hybrid oncocytic/chromophobe tumors - a review. Histol Histopathol 28:1257–1264 Delongchamps NB, Galmiche L, Eiss D, Rouach Y, Vogt B, Timsit MO, Vieillefond A, Mejean A, 2009, Hybrid tumour ‘oncocytomachromophobe renal cell carcinoma’ of the kidney: a report of seven sporadic cases. BJU Int 103:1381–1384 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1519 EP 1524 DI 10.1007/s12253-018-0548-8 PG 6 ER PT J AU Sun, G Meng, J Duan, H Zhang, D Tang, Y AF Sun, Gongping Meng, Jin Duan, He Zhang, Dewei Tang, Yuanxin TI Diagnostic Assessment of septin9 DNA Methylation for Colorectal Cancer Using Blood Detection: A Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SEPT9 methylation; Colorectal cancer; Positive likelihood ratio; Negative likelihood ratio; Summary receiver operating characteristic ID SEPT9 methylation; Colorectal cancer; Positive likelihood ratio; Negative likelihood ratio; Summary receiver operating characteristic AB This meta-analysis aimed to assess the diagnostic efficiency of blood-based septin 9 (SEPT9) methylation assay for the detection of colorectal cancer (CRC). Studies were searched in the Springer, Wiley, Cochrane Library, PubMed, Ovid, Embase, Web of Science, China BioMedicine,Wanfang and China National Knowledge Infrastructure databases until July 2017. Methodological quality assessment was performed based on the guidelines of the Quality Assessment of Diagnostic Accuracy Studies. According to 1/3 and 2/3 algorithms, the meta-analyses for the diagnostic effect of SEPT9 in CRC were compared with healthy subjects and subjects with polyps, adenoma, and non-CRC, respectively. The random effects model was applied and publication bias was evaluated. The included 29 studies comprised 10,486 subjects (3202 patients with CRC and 7284 controls). In comparison with healthy subjects, the pooled sensitivity with 95% confidence intervals (CIs) of SEPT9 methylation for the diagnosis of CRC was 0.74 (95% CI: 0.61–0.84) in the 1/3 algorithm group, whereas the specificity was 0.96 (95% CI: 0.95–0.97) in the 2/3 algorithm group. Additionally, positive likelihood ratio was less than 10 and negative likelihood ratio more than 0.1 in the 2/3 algorithm group for patients with CRC vs. polyps and adenoma. The P value of Deeks’ funnel plot was 0.36, suggesting that there was no publication bias. SEPT9 methylation can be used to diagnose CRC in healthy individuals under the 2/3 algorithm. The determination of SEPT9 methylation does not distinguish well between CRC and polyps or adenoma. C1 [Sun, Gongping] China Medical University, The 4th Affiliated Hospital, Department of Surgery, No. 4 Chongshan Road, 110032 Shenyang, China. [Meng, Jin] China Medical University, Shengjing Hospital, No. 9 North of Nanjing Street, Heping District, 110000 Shenyang, China. [Duan, He] China Medical University, The 4th Affiliated Hospital, Department of Surgery, No. 4 Chongshan Road, 110032 Shenyang, China. [Zhang, Dewei] China Medical University, The 4th Affiliated Hospital, Department of Surgery, No. 4 Chongshan Road, 110032 Shenyang, China. [Tang, Yuanxin] China Medical University, The 4th Affiliated Hospital, Department of Surgery, No. 4 Chongshan Road, 110032 Shenyang, China. RP Zhang, D (reprint author), China Medical University, The 4th Affiliated Hospital, Department of Surgery, 110032 Shenyang, China. EM zhdeweizh@sina.com CR Aran V, Victorino AP, Thuler LC, Ferreira CG, 2016, Colorectal cancer: epidemiology, disease mechanisms and interventions to reduce onset and mortality. Clin Colorectal Cancer 15(3):195–203 Christensen TD, Escoffery C, Plath J, Son HJ, Edwards SKE, Xu R, 2017, Advances in colorectal neoplasms, pp 334 Brenner H, Kloor M, Pox CP, 2014, Colorectal cancer. Lancet 383(9927):1490–1502 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J, 2016, Cancer statistics in China, 2015. CA Cancer J Clin 66(2):115–132 Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RG, Barzi A, Jemal A, 2017, Colorectal cancer statistics, 2017. CA Cancer J Clin 67(3):177–193 Bae JM, Kim JH, Kang GH, 2016, Molecular subtypes of colorectal cancer and their clinicopathologic features, with an emphasis on the serrated neoplasia pathway. Arch Pathol Lab Med 140(5):406– 412 El Bairi K, Tariq K, Himri I, Jaafari A, Smaili W, Kandhro AH, Gouri A, Ghazi B, 2017, Decoding colorectal cancer epigenomics. Cancer Genet 220:49–76 Shen L, Catalano PJ, Benson AB 3rd, O'Dwyer P, Hamilton SR, Issa JP, 2007, Association between DNA methylation and shortened survival in patients with advanced colorectal cancer treated with 5-fluorouracil based chemotherapy. Clin Cancer Res 13(20): 6093–6098 Church TR, Wandell M, Lofton-Day C, Mongin SJ, Burger M, Payne SR, Castanos-Velez E, Blumenstein BA, Rosch T, Osborn N, 2014, Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut 63(2):317– 325 Esteller M, 2003, Relevance of DNA methylation in the management of cancer. Lancet Oncol 4(6):351–358 Dong L, Ren H, 2018, Blood-based DNA methylation biomarkers for early detection of colorectal Cancer. J Proteomics Bioinform 11(6):120–126 Ganepola GA, Nizin J, Rutledge JR, Chang DH, 2014, Use of blood-based biomarkers for early diagnosis and surveillance of colorectal cancer. World J Gastrointest Oncol 6(4):83 Wasserkort R, Kalmar A, Valcz G, Spisak S, Krispin M, Toth K, Tulassay Z, Sledziewski AZ, Molnar B, 2013, Aberrant septin 9 DNA methylation in colorectal cancer is restricted to a single CpG island. BMC Cancer 13(398):1471–2407 Ravegnini G, Zolezzi Moraga JM, Maffei F, Musti M, Zenesini C, Simeon V, Sammarini G, Festi D, Hrelia P, Angelini S, 2015, Simultaneous analysis of SEPT9 promoter methylation status, micronuclei frequency, and folate-related gene polymorphisms: the potential for a novel blood-based colorectal cancer biomarker. Int J Mol Sci 16(12):28486–28497 Johnson DA, Barclay RL, Mergener K, Weiss G, Konig T, Beck J, Potter NT, 2014, Plasma Septin9 versus fecal immunochemical testing for colorectal cancer screening: a prospective multicenter study. PLoS One 9(6):e98238 Molnar B, Toth K, Bartak BK, Tulassay Z, 2015, Plasma methylated septin 9: a colorectal cancer screeningmarker. Expert Rev Mol Diagn 15(2):171–184 Yan S, Liu Z, Yu S, Bao Y, 2016, Diagnostic value of methylated septin9 for colorectal cancer screening: a meta-analysis. Med Sci Monit 22:3409–3418 Song L, Jia J, Peng X, XiaoW, Li Y, 2017, The performance of the SEPT9 gene methylation assay and a comparison with other CRC screening tests: a meta-analysis. Sci Rep 7(1):3032 Nian J, Xu Sun SM, Yan C, MaY, FengY, Yang L, Yu M, Zhang G, Wang X, 2017, Diagnostic accuracy of methylated SEPT9 for blood-based colorectal Cancer detection: a systematic review and meta-analysis. Clin Transl Gastroenterol 8(1):e216 Galanopoulos M, Tsoukalas N, Papanikolaou IS, Tolia M, Gazouli M, Mantzaris GJ, 2017, Abnormal DNA methylation as a cell-free circulating DNA biomarker for colorectal cancer detection: a review of literature. World J Gastrointest Oncol 9(4):142–152 Grutzmann R, Molnar B, Pilarsky C, Habermann JK, Schlag PM, Saeger HD, Miehlke S, Stolz T, Model F, Roblick UJ, 2008, Sensitive detection of colorectal cancer in peripheral blood by septin 9 DNA methylation assay. PLoS One 3(11):e3759 Baratloo A, Elfil M, Negida A, 2015, Part 3: positive and negative likelihood ratios of diagnostic tests. Emergency 3(4):170–171 He NaCW, Yongqi L, LipingY, Huahong X, BiaoluoW, Yongzhan N, Xin W, Xuegang G, Kaichun W, 2014, Clinical significance of detecting serum methylated Sept9 gene in diagnosis of coloretal cancer. Chin J Dig 34(11):726–731 ChenCH,Yan SL, Yang TH, Chen SF,Yeh YH, Ou JJ, Lin CH, Lee YT, Chen CH, 2017, The relationship between the methylated Septin-9 DNA blood test and stool occult blood test for diagnosing colorectal Cancer in Taiwanese people. J Clin Lab Anal 31(1) Devos T, Tetzner R, Model F, Weiss G, Schuster M, Distler J, Steiger KV, Grutzmann R, Pilarsky C, Habermann JK, 2009, Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem 55(7):1337–1346 HeQ, ChenHY, Bai EQ, LuoYX, Fu RJ, HeYS, Jiang J,WangHQ, 2010, Development of a multiplex MethyLight assay for the detection of multigene methylation in human colorectal cancer. Cancer Genet Cytogenet 202(1):1–10 Qiong HE, Wang M, Zhou JW, Liu N, Lai YR, Pathology DO, 2015, Detection of ALX4 and SEPT9 methylation in human colorectal cancer byMultiplexMethyLight assay. J Sun Yat-Sen Univ, Med Sci, 36:657–662 Herbst A, Rahmig K, Stieber P, Philipp A, Jung A, Ofner A, Crispin A, Neumann J, Lamerz R, Kolligs FT, 2011, Methylation of NEUROG1 in serum is a sensitive marker for the detection of early colorectal Cancer. Am J Gastroenterol 106(6):1110–1118 Jin P, Kang Q,Wang X, Yang L, Yu Y, Li N, He YQ, Han X, Hang J, Zhang J, 2015, Performance of a second generation methylated SEPT9 test in detecting colorectal neoplasm. J Gastroenterol Hepatol 30(5):830–833 Kang Q, Jin P, Yang L,Wang X, An H, Liu L, Li N, Sheng J, 2014, Significance of Septin9 gene methylation detection of plasma circulation DNA in colorectal cancer screening. Zhonghua Yi Xue Za Zhi 94(48):3839 Lee HS, Hwang SM, Kim TS, Kim DW, Park DJ, Kang SB, Kim HH, ParkKU(2013, Circulating methylated septin 9 nucleic acid in the plasma of patients with gastrointestinal cancer in the stomach and colon. Transl Oncol 6(3):290–296 Liu Y, Tham CK, Ong SYK, Ho KS, Lim JF, Min HC, Che KL, Yi Z, Tang CL, KongWE, 2013, Serum methylation levels of TAC1. SEPT9 and EYA4 as diagnostic markers for early colorectal cancers: a pilot study. Biomarkers 18(5):399–405 Lofton-Day C, Model F, Devos T, Tetzner R, Distler J, Schuster M, Song X, Lesche R, Liebenberg V, Ebert M, 2008, DNA methylation biomarkers for blood-based colorectal cancer screening. Clin Chem 54(2):414–423 Potter NT, Hurban P, White MN, Whitlock KD, Loftonday CE, Tetzner R, Koenig T, Quigley NB, Weiss G, 2014, Validation of a real-time PCR–based qualitative assay for the detection of methylated SEPT9 DNA in human plasma. Clin Chem 60(9):1183–1191 Song L, Li Y, Jia J, Zhou G,Wang J, Kang Q, Jin P, Sheng J, Cai G, Cai S, 2016, Algorithm optimization in methylation detection with multiple RT-qPCR. PLoS One 11(11):e0163333 Su XL,Wang YF, Li SJ, Zhang F, CuiHW(2014, High methylation of the SEPT9 gene in Chinese colorectal cancer patients. Genet Mol Res 13(2):2513–2520 Tanzer M, Balluff B, Distler J, Hale K, Leodolter A, Rocken C, Molnar B, Schmid R, Lofton-Day C, Schuster T, 2010, Performance of epigenetic markers SEPT9 and ALX4 in plasma for detection of colorectal precancerous lesions. PLoS One 5(2):e9061 Toth K, Wasserkort R, Sipos F, Kalmar A, Wichmann B, Leiszter K, Valcz G, Juhasz M, Miheller P, Patai AV, 2014, Detection of methylated septin 9 in tissue and plasma of colorectal patients with neoplasia and the relationship to the amount of circulating cell-free DNA. PLoS One 9(12):e115415 Warren JD, Xiong W, Bunker AM, Vaughn CP, Furtado LV, Roberts WL, Fang JC, Samowitz WS, Heichman KA, 2011, Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer. BMC Med 9(1):133 Wu Dong YH, Yue L, Jingnan L, Jiaming Q, 2016, Detecting plasma methylated Septin9 gene combined with fecal immunochemical test in screening colorectal cancer and adenoma in outpatients. Chin J Dig 36(2):107–112 Wu D, Zhou G, Jin P, Zhu J, Li S,Wu Q,Wang G, Sheng J,Wang J, Song L, 2016, Detection of colorectal Cancer using a simplified SEPT9 gene methylation assay is a reliable method for opportunistic screening. J Mol Diagn 18(4):535–545 Qingqing D, Hao Z, Haichen X, Xiaolin L, Yun S, Qiping X, Weihao S, 2015, Value of methylated Septin 9 detection in screening colorectal cancer in elderly people. Chinese Journal of Geriatrics 34(12):1348–1350 Shijie L, Yangang L, Jing W, Changqi C, Yan Y, Peng Y, Qi W, 2015, The application of Septin9 methylation in screening of colorectal cancer using peripheral blood. Chinese Journal of General Surgery 24(12):1756–1760 Zhen W, Jiachang C, Qiong H, Huanyu P, Zhenyu Z, Huayun C, Ming L, 2012, The methylation SEPT9 detection in the early diagnosis for colorectal cancer using MS-HRM. Guangdong Medical Journal 33(12):1732–1734 Orntoft MB, Nielsen HJ, Orntoft TF, Andersen CL, 2015, Performance of the colorectal cancer screening marker Sept9 is influenced by age, diabetes and arthritis: a nested case-control study. BMC Cancer 15(819):015–1832 Toth K, Sipos F, Kalmar A, Patai AV, Wichmann B, Stoehr R, Golcher H, Schellerer V, Tulassay Z, Molnar B, 2012, Detection of methylated SEPT9 in plasma is a reliable screening method for both left-and right-sided colon cancers. PLoS One 7(9):e46000 Powrozek T, Krawczyk P, Kucharczyk T, Milanowski J, 2014, Septin 9 promoter region methylation in free circulating DNA— potential role in noninvasive diagnosis of lung cancer: preliminary report. Med Oncol 31(4):917 BeneishMD(1999, The detection of earnings manipulation. Financ Anal J 55(5):24–36 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1525 EP 1534 DI 10.1007/s12253-018-0559-5 PG 10 ER PT J AU Allameh, A Moazeni-Roodi, A Harirchi, I Ravanshad, M Motiee-Langroudi, M Garajei, A Hamidavi, A Mesbah-Namin, AS AF Allameh, Abdolamir Moazeni-Roodi, Abdolkarim Harirchi, Iraj Ravanshad, Mehrdad Motiee-Langroudi, Maziar Garajei, Ata Hamidavi, Azin Mesbah-Namin, Alireza Seyed TI Promoter DNA Methylation and mRNA Expression Level of p16 Gene in Oral Squamous Cell Carcinoma: Correlation with Clinicopathological Characteristics SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE OSCC; HPV; Smoking; P16 promoter methylation; P16 gene expression; Iran ID OSCC; HPV; Smoking; P16 promoter methylation; P16 gene expression; Iran AB The aim of this study was to investigate the relationship between p16 methylation and its expression in oral squamous cell carcinoma (OSCC). Also the contribution of clinicopathological factors, HPV infection and smoking in p16 expression and promoter methylation has been investigated. In this study 67 consecutive OSCC patients and 59 normal individuals were enrolled. All patients were candidates for surgery of oral cavity and fresh tumor biopsies were collected and processed for DNA and RNA extraction. Normal gingival tissues were collected from individuals referred to dentistry clinic and considered as controls. All the cases and controls were checked for HPV infection and then promoter methylation and expression of p16 gene were determined using Methylation-specific PCR (MSP) and real-time PCR (QPCR), respectively. Methylation of p16 in tumors and normal tissues were 59.7 and 38.9%, respectively. Most of hypermethylated samples (>82%) were in high grades. P16 methylation was comparable in HPV+ and HPV- patients or smokers. P16 was overexpressed (~3 fold; p = 0.044) in HPV+ tumors, but it was significantly down-regulated in smoker patients (40% of all tumors). Comparison of P16 expression in OSCC tumors with different degrees of promoter methylation further suggest the relationship of methylation rate and downregulation of P16 expression. The p16 methylation and expression was differentially affected in patients with HPV infection and the smoker cases. Regardless of the influence of environmental factors, it appears that P16 status is useful for classifying patients with OSCC and for influencing treatment strategies in accordance with this classification. Moreover, targeting the upregulation of p16 could be a promising therapeutic option. C1 [Allameh, Abdolamir] Tarbiat Modares University, Faculty of Medical Sciences, Department of Clinical BiochemistryTehran, Iran. [Moazeni-Roodi, Abdolkarim] Tarbiat Modares University, Faculty of Medical Sciences, Department of Clinical BiochemistryTehran, Iran. [Harirchi, Iraj] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Ravanshad, Mehrdad] Tarbiat Modares University, Faculty of Medical Sciences, Department of Medical VirologyTehran, Iran. [Motiee-Langroudi, Maziar] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Garajei, Ata] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Hamidavi, Azin] Tarbiat Modares University, Faculty of Medical Sciences, Department of Clinical BiochemistryTehran, Iran. [Mesbah-Namin, Alireza Seyed] Tarbiat Modares University, Faculty of Medical Sciences, Department of Clinical BiochemistryTehran, Iran. RP Allameh, A (reprint author), Tarbiat Modares University, Faculty of Medical Sciences, Department of Clinical Biochemistry, Tehran, Iran. EM allameha@modares.ac.ir CR Rivera C, 2015, Essentials of oral cancer. Int J Clin Exp Pathol 8: 11884–11894 Karsai S, Abel U, Roesch-Ely M, Affolter A, Hofele C, Joos S et al, 2007, Comparison of p16(INK4a, expression with p53 alterations in head and neck cancer by tissue microarray analysis. J Pathol 211: 314–322 Al-Kaabi A, van Bockel LW, Pothen AJ, Willems SM, 2014, p16INK4A and p14ARF gene promoter hypermethylation as prognostic biomarker in oral and oropharyngeal squamous cell carcinoma: a review. Dis Markers 2014:260549 Petti S, 2009, Lifestyle risk factors for oral cancer. Oral Oncol 45: 340–350 Laco J, Nekvindova J, Novakova V, Celakovsky P, Dolezalova H, Tucek L et al, 2012, Biologic importance and prognostic significance of selected clinicopathological parameters in patients with oral and oropharyngeal squamous cell carcinoma, with emphasis on smoking, protein p16(INK4a, expression, and HPV status. Neoplasma 59:398–408 Kreimer AR, Clifford GM, Boyle P, Franceschi S, 2005, Human papillomavirus types in head and neck squamous cell carcinomas worldwide: a systematic review. Cancer Epidemiol Biomark Prev 14:467–475 Nobori T, Miura K, Wu DJ, Lois A, Takabayashi K, Carson DA, 1994, Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Nature 368:753–756 Stone S, Jiang P, Dayananth P, Tavtigian SV, Katcher H, Parry D et al, 1995, Complex structure and regulation of the P16, MTS1, locus. Cancer Res 55:2988–2994 SerranoM, Hannon GJ, Beach D, 1993, A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature 366:704–707 Rocco JW, Sidransky D, 2001, p16(MTS-1/CDKN2/INK4a, in cancer progression. Exp Cell Res 264:42–55 Xu R,Wang F,Wu L,Wang J, Lu C, 2013, A systematic review of hypermethylation of p16 gene in esophageal cancer. Cancer Biomark 13:215–226 Jablonowski Z, Reszka E, Gromadzinska J, Wasowicz W, SosnowskiM(2011, Hypermethylation of p16 andDAPK promoter gene regions in patients with non-invasive urinary bladder cancer. Arch Med Sci 7:512–516 Sharma G, Mirza S, Prasad CP, Srivastava A, Gupta SD, Ralhan R, 2007, Promoter hypermethylation of p16INK4A, p14ARF, CyclinD2 and Slit2 in serum and tumor DNA from breast cancer patients. Life Sci 80:1873–1881 Shaw RJ, Liloglou T, Rogers SN, Brown JS, Vaughan ED, Lowe D et al, 2006, Promoter methylation of P16, RARbeta, E-cadherin, cyclin A1 and cytoglobin in oral cancer: quantitative evaluation using pyrosequencing. Br J Cancer 94:561–568 Khor GH, Froemming GR, Zain RB, Abraham MT, Omar E, Tan SK et al, 2013, DNA methylation profiling revealed promoter hypermethylation-induced silencing of p16, DDAH2 and DUSP1 in primary oral squamous cell carcinoma. Int J Med Sci 10:1727–1739 Demokan S, Chuang A, Suoglu Y, UlusanM, Yalniz Z, Califano JA et al, 2012, Promoter methylation and loss of p16(INK4a, gene expression in head and neck cancer. Head Neck 34:1470–1475 Todd R, Hinds PW, Munger K, Rustgi AK, Opitz OG, Suliman Y et al, 2002, Cell cycle dysregulation in oral cancer. Crit Rev Oral Biol Med 13:51–61 1 8 . Yuen PW, Man M, Lam KY, Kwong YL, 2002, Clinicopathological significance of p16 gene expression in the surgical treatment of head and neck squamous cell carcinomas. J Clin Pathol 55:58–60 19. Ai L, Stephenson KK, LingW, Zuo C, Mukunyadzi P, Suen JYet al, 2003, The p16, CDKN2a/INK4a, tumor-suppressor gene in head and neck squamous cell carcinoma: a promoter methylation and protein expression study in 100 cases. Mod Pathol 16:944–950 20. Alves MK, Ferrasi AC, Lima VP, Ferreira MV, de Moura Campos Pardini MI, Rabenhorst SH, 2011, Inactivation of COX-2,HMLH1 and CDKN2A gene by promoter methylation in gastric cancer: relationship with histological subtype, tumor location and helicobacter pylori genotype. Pathobiology 78:266–276 21. Banzai C, Nishino K, Quan J, Yoshihara K, Sekine M, Yahata T et al, 2014, Promoter methylation of DAPK1, FHIT, MGMT, and CDKN2A genes in cervical carcinoma. Int J Clin Oncol 19:127– 132 22. Celebiler Cavusoglu A, Sevinc AI, Saydam S, Canda T, Baskan Z, Kilic Yet al, 2010, Promoter methylation and expression changes of CDH1 and P16 genes in invasive breast cancer and adjacent normal breast tissue. Neoplasma 57:465–472 23. Jitani AK, Raphael V, Mishra J, Shunyu NB, Khonglah Y, Medhi J, 2015, Analysis of human papilloma virus 16/18 DNA and its correlation with p16 expression in Oral cavity squamous cell carcinoma in north-eastern India: a chromogenic in-situ hybridization based study. J Clin Diagn Res 9:EC04–EC07 24. Salehinejad J, Sharifi N, Amirchaghmaghi M, Ghazi N, Shakeri MT, Ghazi A, 2014, Immunohistochemical expression of p16 protein in oral squamous cell carcinoma and lichen planus. Ann Diagn Pathol 18:210–213 25. Lingen MW, Kalmar JR, Karrison T, Speight PM, 2008, Critical evaluation of diagnostic aids for the detection of oral cancer. Oral Oncol 44:10–22 26. Kochl S, Niederstatter H, 2005, Parson W. DNA extraction and quantitation of forensic samples using the phenol-chloroform method and real-time PCR. Methods Mol Biol 297:13–30 27. Sailasree R, Abhilash A, Sathyan KM, Nalinakumari KR, Thomas S, Kannan S, 2008, Differential roles of p16INK4A and p14ARF genes in prognosis of oral carcinoma. Cancer Epidemiol Biomark Prev 17:414–420 28. Cao J, Zhou J, Gao Y, Gu L, Meng H, Liu H et al, 2009, Methylation of p16 CpG island associated with malignant progression of oral epithelial dysplasia: a prospective cohort study. Clin Cancer Res 15:5178–5183 29. Ha PK, Califano JA, 2006, Promoter methylation and inactivation of tumour-suppressor genes in oral squamous-cell carcinoma. Lancet Oncol 7:77–82 30. Ishida E, Nakamura M, Ikuta M, Shimada K, Matsuyoshi S, Kirita T et al, 2005, Promotor hypermethylation of p14ARF is a key alteration for progression of oral squamous cell carcinoma. Oral Oncol 41:614–622 31. Papadimitrakopoulou VA, Izzo J,Mao L, Keck J, Hamilton D, Shin DM et al, 2001, Cyclin D1 and p16 alterations in advanced premalignant lesions of the upper aerodigestive tract: role in response to chemoprevention and cancer development. Clin Cancer Res 7: 3127–3134 32. Perez-Sayans M, Suarez-Penaranda JM, Padin-Iruegas ME, Gayoso-Diz P, Reis-De Almeida M, Barros-Angueira F et al, 2015, The loss of p16 expression worsens the prognosis of OSCC. Appl Immunohistochem Mol Morphol 23:724–732 33. Bhatia V, Goel MM, Makker A, Tewari S, Yadu A, Shilpi P et al, 2014, Promoter region hypermethylation and mRNA expression of MGMT and p16 genes in tissue and blood samples of human premalignant oral lesions and oral squamous cell carcinoma. Biomed Res Int 2014:248419 34. Schlecht NF, Ben-Dayan M, Anayannis N, Lleras RA, Thomas C, Wang Yet al, 2015, Epigenetic changes in the CDKN2A locus are associated with differential expression of P16INK4A and P14ARF in HPV-positive oropharyngeal squamous cell carcinoma. Cancer Med 4:342–353 35. Sargolzaei S, Farhadi S, Kazemi B, Bandehpour M, KharazifardMJ, 2014, The correlation between p16 expression and INK4a locus mutation with grades and stages in oral squamous cell carcinoma. Indian J Pathol Microbiol 57:24–30 36. Dona MG, Spriano G, Pichi B, Rollo F, Laquintana V, Covello R et al, 2015, Human papillomavirus infection and p16 overexpression in oropharyngeal squamous cell carcinoma: a case series from 2010 to 2014. Future Microbiol 10:1283–1291 37. Talikka M, Sierro N, Ivanov NV, Chaudhary N, Peck MJ, Hoeng J, Coggins CRE, 2012, ECR Peitsch MCgenomic impact of cigarette smoke, with application to three smoking-related diseases. Crit Rev Toxicol 42(10):877–889 38. Antonsson A, Neale RE, Boros S, Lampe G, ComanWB, Pryor DI et al, 2015, Human papillomavirus status and p16(INK4A, expression in patients with mucosal squamous cell carcinoma of the head and neck in Queensland, Australia. Cancer Epidemiol 39:174–181 39. Mai S, Welzel G, Ottstadt M, Lohr F, Severa S, Prigge ES et al, 2015, Prognostic relevance of HPVinfection and p16 overexpression in squamous cell anal Cancer. Int J Radiat Oncol Biol Phys 93: 819–827 40. Li J, Poi MJ, Tsai MD, 2011, Regulatory mechanisms of tumor suppressor P16(INK4A, and their relevance to cancer. Biochemistry 50:5566–5582 41. Konig F, Krekeler G, Honig JF, Cordon-Cardo C, Fischer G, Korabiowska M, 2007, Relation between human papillomavirus positivity and p16 expression in head and neck carcinomas–a tissue microarray study. Anticancer Res 27:283–288 42. El-Naggar AK, Westra WH, 2012, p16 expression as a surrogate marker for HPV-related oropharyngeal carcinoma: a guide for interpretative relevance and consistency. Head Neck 34:459–461 43. Wang S, Deng Z, Seneviratne CJ, Cheung GS, Jin L, Zhao B et al, 2015, Enterococcus faecalis promotes osteoclastogenesis and semaphorin 4D expression. Innate Immun 21:726–735 44. Shaw RJ, Hobkirk AJ, Nikolaidis G, Woolgar JA, Triantafyllou A, Brown JS et al, 2013)Molecular staging of surgical margins in oral squamous cell carcinoma using promoter methylation of p16(INK4A), cytoglobin, E-cadherin, and TMEFF2. Ann Surg Oncol 20:2796–2802 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1535 EP 1543 DI 10.1007/s12253-018-0542-1 PG 9 ER PT J AU Zhang, L Huang, Y Ling, J Zhuo, W Yu, Z Luo, Y Zhu, Y AF Zhang, Liang Huang, Yi Ling, JunJun Zhuo, Wenlei Yu, Zhen Luo, Yunbo Zhu, Yi TI Is Integrin Subunit Alpha 2 Expression a Prognostic Factor for Liver Carcinoma? A Validation Experiment Based on Bioinformatics Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Liver carcinoma; ITGA2; Tissue microarray; Immunohistochemistry; Prognosis ID Liver carcinoma; ITGA2; Tissue microarray; Immunohistochemistry; Prognosis AB ITGA2 (Integrin alpha-2) has been detected to be over-expressed in a number of cancers and has been suggested to be involved in cell adhesion and cell-surface mediated signaling. Our previous study using bioinformatic analyses has shown that ITGA2 might be a key gene being involved in the Cadmium-induced malignant transformation of liver cells. In the present study, we firstly aimed to learn the possible functions of ITGA2 via bioinformatics analysis, and then test its expression and clinical significance in liver carcinoma specimens through laboratory experiments. Gene ontology (GO) and pathway enrichment analysis, as well as protein-protein interaction (PPI) analysis has been conducted in Genecards. Then, a tissue microarray containing 90 cases of liver cancer and 90 paired adjacent non-cancerous samples was used for detection of ITGA2 expression by immunohistochemistry assay. Consequently, ITGA2 may be enriched in pathways regarding cell adhesion and migration. PPI analysis suggests that ITGA1, ITGB2, FLT4, LAMB1 and AGRN may have a close relationship with ITGA2. No association between ITGA2 expression and clinical parameters was observed. However, the data showed that ITGA2 might be an independent prognostic factor for liver cancer patients. In conclusion, the data suggest that ITGA2 over-expression might be a potential unfavorable prognostic factor and a potential therapeutic target for liver carcinoma. C1 [Zhang, Liang] China Agricultural University, College of food science and nutritional engineeringBeijing, China. [Huang, Yi] Affiliated hospital of Guizhou Medical University, Department of Internal MedicineGuiyang, China. [Ling, JunJun] Third Military Medical University, Xinqiao Hospital, Institute of CancerChongqing, China. [Zhuo, Wenlei] Third Military Medical University, Xinqiao Hospital, Institute of CancerChongqing, China. [Yu, Zhen] China Agricultural University, College of food science and nutritional engineeringBeijing, China. [Luo, Yunbo] China Agricultural University, College of food science and nutritional engineeringBeijing, China. [Zhu, Yi] China Agricultural University, College of food science and nutritional engineeringBeijing, China. RP Zhu, Y (reprint author), China Agricultural University, College of food science and nutritional engineering, Beijing, China. EM zhuyi@cau.edu.cn CR Satarug S, Vesey DA, Gobe GC, 2017, Current health risk assessment practice for dietary cadmium: data from different countries. Food Chem Toxicol 106(Pt A):430–445. , DOI 10.1016/j. fct.2017.06.013 Sabolic I, Breljak D, Skarica M, Herak-Kramberger CM, 2010, Role of metallothionein in cadmium traffic and toxicity in kidneys and other mammalian organs. Biometals 23(5):897–926. https:// doi.org/10.1007/s10534-010-9351-z Cartularo L, Kluz T, Cohen L, Shen SS, CostaM(2016, Molecular mechanisms of malignant transformation by low dose cadmium in Normal human bronchial epithelial cells. PLoS One 11(5): e0155002. , DOI 10.1371/journal.pone.0155002 Garcia-Esquinas E, Pollan M, Tellez-Plaza M, Francesconi KA, Goessler W, Guallar E, Umans JG, Yeh J, Best LG, Navas-Acien A, 2014, Cadmium exposure and cancer mortality in a prospective cohort: the strong heart study. Environ Health Perspect 122(4):363– 370. , DOI 10.1289/ehp.1306587 Ledda C, Loreto C, Zammit C, Marconi A, Fago L, Matera S, Costanzo V, Fuccio Sanza G, Palmucci S, Ferrante M, Costa C, Fenga C, Biondi A, Pomara C, Rapisarda V, 2017, Noninfective occupational risk factors for hepatocellular carcinoma: a review, review). Mol Med Rep 15(2):511–533. , DOI 10.3892/ mmr.2016.6046 Ma X,YangY, Li HL, ZhengW, Gao J, ZhangW, Yang G, ShuXO, Xiang YB, 2017, Dietary trace element intake and liver cancer risk: results from two population-based cohorts in China. Int J Cancer 140(5):1050–1059. , DOI 10.1002/ijc.30522 Stelzer G, Rosen N, Plaschkes I, Zimmerman S, Twik M, Fishilevich S, Stein TI, Nudel R, Lieder I, Mazor Y, Kaplan S, Dahary D, Warshawsky D, Guan-Golan Y, Kohn A, Rappaport N, Safran M, Lancet D, 2016, The GeneCards suite: from gene data mining to disease genome sequence analyses. Curr Protoc Bioinformatics 54(1):30 31–31 30 33. , DOI 10.1002/cpbi.5 Mo Z, Zheng S, Lv Z, Zhuang Y, Lan X, Wang F, Lu X, Zhao Y, Zhou S, 2016, Senescencemarker protein 30, SMP30, serves as a potential prognostic indicator in hepatocellular carcinoma. Sci Rep 6:39376. , DOI 10.1038/srep39376 Yang Q, Bavi P, Wang JY, Roehrl MH, 2017, Immuno-proteomic discovery of tumor tissue autoantigens identifies olfactomedin 4, CD11b, and integrin alpha-2 as markers of colorectal cancer with liver metastases. J Proteome 168:53–65. , DOI 10.1016/j. jprot.2017.06.021 Dong J,Wang R, Ren G, Li X,Wang J, Sun Y, Liang J, Nie Y, Wu K, Feng B, Shang Y, Fan D, 2017, HMGA2-FOXL2 Axis regulates metastases and epithelial-to-mesenchymal transition of chemoresistant gastric cancer. Clin Cancer Res 23(13):3461– 3473. , DOI 10.1158/1078-0432.CCR-16-2180 DingW, Fan XL, Xu X, Huang JZ, Xu SH, Geng Q, Li R, Chen D, Yan GR, 2015, Epigenetic silencing of ITGA2 by MiR-373 promotes cell migration in breast cancer. PLoS One 10(8):e0135128. , DOI 10.1371/journal.pone.0135128 Wong KF, Liu AM, Hong W, Xu Z, Luk JM, 2016, Integrin alpha2beta1 inhibits MST1 kinase phosphorylation and activates Yes-associated protein oncogenic signaling in hepatocellular carcinoma. Oncotarget 7(47):77683–77695. , DOI 10.18632/ oncotarget.12760 Zhao X, Wu Y, Lv Z, 2015, miR-128 modulates hepatocellular carcinoma by inhibition of ITGA2 and ITGA5 expression. Am J Transl Res 7(9):1564–1573 Chin SP, Marthick JR, West AC, Short AK, Chuckowree J, Polanowski AM, Thomson RJ, Holloway AF, Dickinson JL, 2015, Regulation of the ITGA2 gene by epigenetic mechanisms in prostate cancer. Prostate 75(7):723–734. , DOI 10.1002/ pros.22954 Gharibi A, La Kim S, Molnar J, Brambilla D, Adamian Y, Hoover M, Hong J, Lin J, Wolfenden L, Kelber JA, 2017, ITGA1 is a pre-malignant biomarker that promotes therapy resistance and metastatic potential in pancreatic cancer. Sci Rep 7(1):10060. , DOI 10.1038/s41598-017-09946-z Laskowska J, Lewandowska-Bieniek J, Szczepanek J, Styczynski J, Tretyn A, 2016, Genomic and transcriptomic profiles and in vitro resistance to mitoxantrone and idarubicin in pediatric acute leukemias. J Gene Med 18(8):165–179. , DOI 10.1002/jgm. 2889 Lee JY, Hong SH, Shin M, Heo HR, Jang IH, 2016, Blockade of FLT4 suppresses metastasis of melanoma cells by impaired lymphatic vessels. Biochem Biophys Res Commun 478(2):733– 738. , DOI 10.1016/j.bbrc.2016.08.017 Virga J, Bognar L, Hortobagyi T, Zahuczky G, Csosz E, Kallo G, Toth J, Hutoczki G, Remenyi-Puskar J, Steiner L, Klekner A, 2017, Prognostic role of the expression of invasion-related molecules in Glioblastoma. J Neurol Surg A Cent Eur Neurosurg 78(1):12–19. , DOI 10.1055/s-0036-1584920 Lin Q, Lim HS, Lin HL, Tan HT, Lim TK, CheongWK, Cheah PY, Tang CL, Chow PK, Chung MC, 2015, Analysis of colorectal cancer glyco-secretome identifies laminin beta-1, LAMB1, as a potential serological biomarker for colorectal cancer. Proteomics 15(22):3905–3920. , DOI 10.1002/pmic.201500236 Chakraborty S, Lakshmanan M, Swa HL, Chen J, Zhang X, Ong YS, Loo LS, Akincilar SC, Gunaratne J, Tergaonkar V, Hui KM, Hong W, 2015, An oncogenic role of Agrin in regulating focal adhesion integrity in hepatocellular carcinoma. Nat Commun 6: 6184. , DOI 10.1038/ncomms7184 Chakraborty S, Njah K, Pobbati AV, Lim YB, Raju A, Lakshmanan M, Tergaonkar V, Lim CT, Hong W, 2017, Agrin as a Mechanotransduction signal regulating YAP through the hippo pathway. Cell Rep 18(10):2464–2479. , DOI 10.1016/j. celrep.2017.02.041 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1545 EP 1552 DI 10.1007/s12253-018-0551-0 PG 8 ER PT J AU Xie, B Nie, Sh Hu, G Xiong, L Hu, F Li, M Peng, T Nie, J He, Y AF Xie, Biao Nie, Shaolin Hu, Gui Xiong, Li Hu, Fan Li, Mei Peng, Tianshu Nie, Jing He, Yongheng TI The Involvement of NF-κB/Klotho Signaling in Colorectal Cancer Cell Survival and Invasion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE LPS; NF-κB; Colorectal cancer; Klotho ID LPS; NF-κB; Colorectal cancer; Klotho AB Lipopolysaccharide significantly increased invasion, cell proliferation, and phospho-NF-κB p65 and phospho-IGF-1R protein, but decreased klotho protein expression, cell apoptosis, and the percentage of sub G0/G1 cells in SW480 and HT29 colorectal cancer cells. In contrast, NF-κB inhibitor exhibited a counteract effect of lipopolysaccharide. Transfection of Toll-like receptor 4 shRNA significantly decreased phospho-NF-κB p65 and phospho-IGF-1R protein levels, invasion, but significantly increased klotho protein expression, cell apoptosis, and the percentage of sub G0/G1 in SW480 and HT29 cells. In conclusion, inflammation inhibits klotho gene expression in colorectal cancer cells through activation of Toll-like receptor 4/NF-κB signal pathway. C1 [Xie, Biao] The Second Affiliated Hospital of Hunan University of Chinese Medicine, Department One of Anorectal Surgery, 139 Renmin Road, 410005 Changsha, Hunan, China. [Nie, Shaolin] Hunan Province Tumor Hospital, Department of Colorectal Surgery, 410013 Changsha, Hunan, China. [Hu, Gui] The Third Xiangya Hospital, Central South University, Department of Gastrointestinal Surgery, 410013 Changsha, Hunan, China. [Xiong, Li] Central South University, the Second Xiangya Hospital, Department of Hepatobiliary Surgery, 410008 Changsha, Hunan, China. [Hu, Fan] The Second Affiliated Hospital of Hunan University of Chinese Medicine, Department One of Anorectal Surgery, 139 Renmin Road, 410005 Changsha, Hunan, China. [Li, Mei] The Second Affiliated Hospital of Hunan University of Chinese Medicine, Department One of Anorectal Surgery, 139 Renmin Road, 410005 Changsha, Hunan, China. [Peng, Tianshu] Hunan Slack King Laboratory Animal Co., Ltd., 4100125 Changsha, Hunan, China. [Nie, Jing] The Second Affiliated Hospital of Hunan University of Chinese Medicine, Department Four of Anorectal Surgery, 410005 Changsha, Hunan, China. [He, Yongheng] The Second Affiliated Hospital of Hunan University of Chinese Medicine, Department One of Anorectal Surgery, 139 Renmin Road, 410005 Changsha, Hunan, China. RP He, Y (reprint author), The Second Affiliated Hospital of Hunan University of Chinese Medicine, Department One of Anorectal Surgery, 410005 Changsha, China. EM heyongheng1964@163.com CR Cancer-World Health Organization. http://www.who.int/ mediacentre/factsheets/fs297/en/ Colorectal Cancer Statistics, 2017, CA: Cancer J Clin. https://doi. org/10.3322/caac.21395 American Cancer Society, 2014, Colorectal cancer facts & figures 2014-2016. Atlanta, Ga: American Cancer Society Terzic J, Grivennikov S, Karin E, Karin M, 2010, Inflammation and colon cancer. Gastroenterology 138:2101–2114.e5 Viennois E, Chen F, Merlin D, 2013, NF-κB pathway in colitis-associated cancers. Transl Gastrointest Cancer 2:21– 29 Zhu QC, Gao RY, Wu W, Qin HL, 2013, Epithelial-mesenchymal transition and its role in the pathogenesis of colorectal cancer. Asian Pac J Cancer Prev 14:2689–2698 Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ, 2010, HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol 28:367–388 Zhang G, Ghosh S, 2000, Molecular mechanisms of NF-kappaB activation induced by bacterial lipopolysaccharide through toll-like receptors. J Endotoxin Res 6:453–457 Zhou X, Wang X, 2015, Klotho: a novel biomarker for cancer. J Cancer Res Clin Oncol 141:961–969 Tang X,Wang Y, Fan Z, Ji G,Wang M, Lin J, Huang S, Meltzer SJ, 2016, Klotho: a tumor suppressor and modulator of the Wnt/β- catenin pathway in human hepatocellular carcinoma. Lab Investig 96:197–205 Wang L, Wang X,Wang X, Jie P, Lu H, Zhang S, Lin X, Lam EK, Cui Y, Yu J, Jin H, 2011, Klotho is silenced through promoter hypermethylation in gastric cancer. Am J Cancer Res 1:111–119 Xie B, Chen J, Liu B, Zhan J, 2013, Klotho acts as a tumor suppressor in cancers. Pathol Oncol Res 19:611–617 Wu C, Lv C, Chen F, MaX, Shao Y, Wang Q, 2015, The function of miR-199a-5p/klotho regulating TLR4/NF-κB p65/NGAL pathways in rat mesangial cells cultured with high glucose and the mechanism. Mol Cell Endocrinol 417:84–93 Roy A, Srivastava M, Saqib U, Liu D, Faisal SM, Sugathan S, Bishnoi S, Baig MS, 2016, Potential therapeutic targets for inflammation in toll-like receptor 4, TLR4)-mediated signaling pathways. Int Immunopharmacol 40:79–89 Liu B, Sun L, Liu Q, Gong C, Yao Y, Lv X, Lin L, Yao H, Su F, Li D, Zeng M, Song E, 2015, A cytoplasmic NF-κB interacting long noncoding RNA blocks IκB phosphorylation and suppresses breast cancer metastasis. Cancer Cell 27:370–381 Xie B, Zhou J, Yuan L, Ren F, Liu DC, Li Q, Shu G, 2013, Epigenetic silencing of klotho expression correlateswith poor prognosis of human hepatocellular carcinoma. Hum Pathol 44:795–801 Xie B, Zhou J, Shu G, Liu DC, Zhou J, Chen J, Yuan L, 2013, Restoration of klotho gene expression induces apoptosis and autophagy in gastric cancer cells: tumor suppressive role of klotho in gastric cancer. Cancer Cell Int 13:18 Shu G, Xie B, Ren F, Liu DC, Zhou J, Li Q, Chen J, Yuan L, Zhou J, 2013, Restoration of klotho expression induces apoptosis and autophagy in hepatocellular carcinoma cells. Cell Oncol, Dordr, 36: 121–129 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1553 EP 1565 DI 10.1007/s12253-018-0493-6 PG 13 ER PT J AU Luo, Y He, Y Ye, X Song, J Wang, Q Li, Y Xie, X AF Luo, Yongyun He, Yaqin Ye, Xiaoping Song, Jianjun Wang, Qi Li, Yukui Xie, Xiaoliang TI High Expression of Long Noncoding RNA HOTAIRM1 is Associated with the Proliferation and Migration in Pancreatic Ductal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HOTAIRM1; Pancreatic ductal adenocarcinoma; Proliferation; Migration ID HOTAIRM1; Pancreatic ductal adenocarcinoma; Proliferation; Migration AB Pancreatic ductal adenocarcinoma (PDAC) is an incurable malignancy. Long noncoding RNA (LncRNA) HOTAIRM1 (HOX antisense intergenic RNA myeloid 1) has been shown to play important roles in the progression of several type cancers. However, the exact role of HOTAIRM1 in PDAC development remains largely unknown. This study aims to evaluate the potential function of HOTAIRM1 in the development and progress of PDAC. HOTAIRM1 expression was measured by RT-qPCR in forty seven paired human PDAC tissues and five PDAC cell lines. SW1990 and PANC-1 cells were transfected with siHOTAIRM1 to achieve HOTAIRM1 silence. MTT assay and colony formation assay were used to detect the effect of HOTAIRM1 knockdown on cell proliferation. The impact of HOTAIRM1 silence on cell cycle and apoptosis was assessed by flow cytometry assay. Transwell migration assay was performed to explore the influence of HOTAIRM1 downregulation on the migratory potential of PDAC cells.Western blot assay was applied to determine the expression changes of cell cycle, apoptosis, and migration-related genes before and after downregulating HOTAIRM1. HOTAIRM1 expression was abnormally upregulated in PDAC tissues and cells when compared with the control samples, and was positively associated with the expression of KRAS gene mutation. In vitro functional experiments, HOTAIRM1 expression was significantly downregulated by transfection with siHOTAIRM1 in SW1990 and PANC cell lines. HOTAIRM1 knockdown attenuated cell proliferation by inducing cell cycle arrest at G0/G1 phase, promoted cell apoptosis, and inhibited cell migration in PDAC cells by regulating related-genes expression. In conclusion, HOTAIRM1 plays a critical role in PDAC progression, which may be a novel diagnostic and rational therapeutic target for the treatment of pancreatic ductal adenocarcinoma. C1 [Luo, Yongyun] Ningxia Medical University, General Hospital, Department of Hepatobiliary Surgery, 750004 Yinchuan, China. [He, Yaqin] Ningxia Medical University, General Hospital, Surgery Laboratory, 804 South Shengli Street, 750004 Yinchuan, China. [Ye, Xiaoping] Ningxia Medical University, General Hospital, Department of Colorectal Surgery, 804 South Shengli Street, 750004 Yinchuan, China. [Song, Jianjun] Ningxia Medical University, General Hospital, Department of Hepatobiliary Surgery, 750004 Yinchuan, China. [Wang, Qi] Ningxia Medical University, General Hospital, Department of Hepatobiliary Surgery, 750004 Yinchuan, China. [Li, Yukui] Ningxia Medical University, General Hospital, Surgery Laboratory, 804 South Shengli Street, 750004 Yinchuan, China. [Xie, Xiaoliang] Ningxia Medical University, General Hospital, Department of Colorectal Surgery, 804 South Shengli Street, 750004 Yinchuan, China. RP Li, Y (reprint author), Ningxia Medical University, General Hospital, Surgery Laboratory, 750004 Yinchuan, China. EM zaimo571@163.com CR Oncology TL, 2014, Pancreatic cancer in the spotlight. Lancet Oncol 15:241 Taher MA, Khan ZR, Chowdhury MM, Nureelahi M, Chowdhury AK, Faruque MS, Wahiduzzaman M, Haque MA, 2015, Pylorus preserving pancreaticoduodenectomy vs. standard Whipple's procedure in case of carcinoma head of the pancreas and periampullary carcinoma. Mymensingh Med J 24:319–325 Xu YF, Lu Y, Cheng H, Jiang J, Xu J, Long J, Liu L, Ni Q, Liu C, Yu XJ, 2015, High expression of human leukocyte antigen-G is associated with a poor prognosis in patients with PDAC. Curr Mol Med 15:360–367 Ma J, Jemal A, 2013, The rise and fall of cancer mortality in the USA: why does pancreatic cancer not follow the trend? Future Oncol 9:917–919 Mercer TR, Dinger ME, Mattick JS, 2009, Long non-coding RNAs: insights into functions. Nat Rev Genet 10:155–159 Ponting CP, Oliver PL, Reik W, 2009, Evolution and functions of long noncoding RNAs. Cell 136:629–641 Leveille N, Melo CA, Rooijers K, Diazlagares A, Melo SA, Korkmaz G, Lopes R, Akbari MF, Maia AR, Wijchers PJ, 2015, Genome-wide profiling of p53-regulated enhancer RNAs uncovers a subset of enhancers controlled by a lncRNA. Nat Commun 6: 6520 Yin Y, Yan P, Lu J, Song G, Zhu Y, Li Z, Zhao Y, Shen B, Huang X, Zhu H, 2015, Opposing roles for the lncRNA haunt and its genomic locus in regulating HOXA gene activation during embryonic stem cell differentiation. Cell Stem Cell 16:504–516 Wapinski O, Chang HY, 2011, Long noncoding RNAs and human disease. Trends Cell Biol 21:354–361 Gong C, Maquat LE, 2011, lncRNAs transactivate STAU1- mediated mRNA decay by duplexing with 3' UTRs via Alu elements. Nature 470:284–288 Crea F, Watahiki A, Quagliata L, Xue H, Pikor L, Parolia A, Wang Y, Lin D, Wan LL, Farrar WL, 2014, Identification of a long noncoding RNA as a novel biomarker and potential therapeutic target for metastatic prostate cancer. Oncotarget 5:764 Shi Y, Lu J, Zhou J, Tan X, He Y, Ding J, Tian Y,Wang L,Wang K, 2014, Long non-coding RNA Loc554202 regulates proliferation and migration in breast cancer cells. Biochem Biophys Res Commun 446:448–453 Zhuang M, Gao W, Xu J, Wang P, Shu Y, 2014, The long noncoding RNA H19-derived miR-675 modulates human gastric cancer cell proliferation by targeting tumor suppressor RUNX1. Biochem Biophys Res Commun 448:315–322 Ding YC, Yu W, Ma C, Wang Q, Huang CS, Huang T, 2014, Expression of long non-coding RNA LOC285194 and its prognostic significance in human pancreatic ductal adenocarcinoma. Int J Clin Exp Pathol 7:8065–8070 Peng W, Gao W, Feng J, 2014, Long noncoding RNA HULC is a novel biomarker of poor prognosis in patients with pancreatic cancer. Med Oncol 31:346 Jiao F, Hu H, Han T, Yuan C,Wang L, Jin Z, Guo Z,Wang L, 2015, Long noncoding RNA MALAT-1 enhances stem cell-like phenotypes in pancreatic cancer cells. Int J Mol Sci 16:6677–6693 Kim K, Jutooru I, Chadalapaka G, Johnson G, Frank J, Burghardt R, Kim S, Safe S, 2013, HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer. Oncogene 32:1616–1625 Zhang X, Lian Z, Padden C, GersteinMB, Rozowsky J, Snyder M, Gingeras TR, Kapranov P,Weissman SM, Newburger PE, 2009, A myelopoiesis-associated regulatory intergenic noncoding RNA transcript within the human HOXA cluster. Blood 113:2526–2534 Zhou Y, Gong B, Jiang ZL, Zhong S, Liu XC, Dong K, Wu HS, Yang HJ, Zhu SK, 2015, Microarray expression profile analysis of long non-coding RNAs in pancreatic ductal adenocarcinoma. Int J Oncol 48:670 Marina DB, Salut B, Josep N, Marta P, Anna C, David G, Lourdes E , Mar T, Inmaculada H, Maria RJ, 2015 , The lincRNAHOTAIRM1, located in theHOXAgenomic region, is expressed in acute myeloid leukemia, impacts prognosis in patients in the intermediate-risk cytogenetic category, and is associated with a distinctive microRNA signature. Oncotarget 6:31613 Chen Y,Wu JJ, Lin XB, Bao Y, Chen ZH, Zhang CR, Cai Z, Zhou JY, DingMH, DifferentialWXJ, 2015, lncRNA expression profiles in recurrent gliomas compared with primary gliomas identified by microarray analysis. Int J Clin Exp Med 8:5033–5043 KimST, LimDH, Jang KT, LimT, Lee J, Choi YL, JangHL,Yi JH, Baek KK, Park SH, 2011, Impact of KRAS mutations on clinical outcomes in pancreatic cancer patients treated with first-line gemcitabine-based chemotherapy. Mol Cancer Ther 10:1993–1999 Rasola A, GeunaM(2001, A flow cytometry assay simultaneously detects independent apoptotic parameters. Cytometry 45:151–157 Yi Z, Shi M, Hao C, Gu J, Zhang J, Shen B, Deng X, Xie J, Xi Z, Peng C, 2015, NPM1 activates metabolic changes by inhibiting FBP1 while promoting the tumorigenicity of pancreatic cancer cells. Oncotarget 6:21443–21451 Daniel Z, Yuliya PG, Der CJ, Bryant KL, 2016, KRAS mutant pancreatic cancer: no lone path to an effective treatment. Cancers 8:45 T G and S D, 2012, The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol 9:703–719 Wan L, Kong J, Tang J, Wu Y, Xu E, Lai M, Zhang H, 2016, HOTAIRM1 as a potential biomarker for diagnosis of colorectal cancer functions the role in the tumour suppressor. J Cell Mol Med 20:2036–2044 Su X, Malouf GG, Chen Y, Zhang J, Yao H, Valero V, Weinstein JN, Spano JP, Meric-Bernstam F, Khayat D, 2014, Comprehensive analysis of long non-coding RNAs in human breast cancer clinical subtypes. Oncotarget 5:9864–9876 Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA, 2015, Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 29:595–608 Hilgers W, Rosty C, Hahn SA, 2002, Molecular pathogenesis of pancreatic cancer. Hematol Oncol Clin North Am 16:17–35 Schubbert S, Shannon K, Bollag G, 2007, Hyperactive Ras in developmental disorders and cancer. Nat Rev Cancer 7:295–308 Ijichi H, Chytil A, Gorska AE, Aakre ME, Fujitani Y, Fujitani S, Wright CVE, Moses HL, 2006, Aggressive pancreatic ductal adenocarcinoma in mice caused by pancreas-specific blockade of transforming growth factor-β signaling in cooperation with active Kras expression. Genes Dev 20:3147–3160 Thayer SP, Magliano MPD, Heiser PW, Nielsen CM, Roberts DJ, Lauwers GY, Yan PQ, Gysin S, Castillo CF, Yajnik V, 2003, Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 425:851–856 MaglianoMPD, Biankin AV, Heiser PW, Cano DA, Gutierrez PJA, Deramaudt T, Segara D, Dawson AC, Kench JG, Henshall SM, 2007, Common activation of canonicalWnt signaling in pancreatic adenocarcinoma. PLoS One 2:e1155 Weijzen S, Rizzo P, Braid M, Vaishnav R, Jonkheer SM, Zlobin A, Osborne BA, Gottipati S, Aster JC, HahnWC, 2002, Activation of Notch-1 signaling maintains the neoplastic phenotype in human Ras-transformed cells. Nat Med 8:979–986 Mikayama Y, Morinaga S, Watanabe T, Numata M, Tamagawa H, Yamamoto N, Shiozawa M, Akaike M, Yoichi K, Matsukuma S, 2012, Association of KRAS G12D mutation with outcome in patients with pancreatic cancer after surgery. J Clin Oncol 30:221 Zhi X, ChenW, Xue F, Liang C, Chen BW, Zhou Y,Wen L, Hu L, Shen J, Bai X, 2015, OSI-027 inhibits pancreatic ductal adenocarcinoma cell proliferation and enhances the therapeutic effect of gemcitabine bothin vitroandin vivo. Oncotarget 6:26230– 26241 Coqueret O, 2002, Linking cyclins to transcriptional control. Gene 299:35–55 Aix E, Sanchezferrer C, Aguado T, Flores I, 2016, Postnatal telomere dysfunction induces cardiomyocyte cell-cycle arrest through p21 activation. J Cell Biol 213:571–583 Cannell I, Merrick K, Morandell S, Zhu CQ, Braun C, Grant R, Cameron E, Tsao MS, Hemann M, Yaffe M, 2015, A pleiotropic RNA-binding protein controls distinct cell cycle checkpoints to drive resistance of p53 -defective tumors to chemotherapy. Cancer Cell 28:623–637 Cully M, 2015, Trial watch: Combinations with checkpoint inhibitors at wavefront of cancer immunotherapy. Nat Rev Drug Discov 14:374 Zhang X, Weissman SM, Newburger PE, 2014, Long intergenic non-coding RNA HOTAIRM1 regulates cell cycle progression during myeloid maturation in NB4 human promyelocytic leukemia cells. RNA Biol 11:777–787 Besson A, Dowdy SF, Roberts JM, 2008, CDK inhibitors: cell cycle regulators and beyond. Dev Cell 14:159–169 Gartel AL, Radhakrishnan SK, 2005, Lost in transcription: p21 repression, mechanisms, and consequences. Cancer Res 65:3980– 3985 JM B, LD A, 2005, The role of apoptosis in cancer development and treatment response. Nat Rev Cancer 5:231–237 Nollet F, Berx G, Van RF, 1999, The role of the E-cadherin/catenin adhesion complex in the development and progression of cancer. Mol Cell Biol Res Commun 2:77–85 Mcinroy L, Maatta A, 2007, Down-regulation of vimentin expression inhibits carcinoma cell migration and adhesion. Biochem Biophys Res Commun 360:109–114 Nagi C, Guttman M, Jaffer S, Qiao R, Keren R, Triana A, Li M, Godbold J, Bleiweiss IJ, Hazan RB, 2005, N-cadherin expression in breast cancer: correlation with an aggressive histologic variant– invasive micropapillary carcinoma. Breast Cancer Res Treat 94: 225–235 Cheng CW, Chang CW, Chu HW, Chen CY, Yu JC, Chao JI, Liu HF, Ding SL, Shen CY, 2012, MicroRNA-30a inhibits cell migration and invasion by downregulating vimentin expression and is a potential prognostic marker in breast cancer. Breast Cancer Res Treat 134:1081–1093 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1567 EP 1577 DI 10.1007/s12253-018-00570-4 PG 11 ER PT J AU Lalosevic, SM Coric, MV Pekmezovic, DT Simic, PT Ercegovac, PM Markovic, PA Krivokapic, VZ AF Lalosevic, Lj. Stojkovic Milica Coric, M Vesna Pekmezovic, D Tatjana Simic, P Tatjana Ercegovac, S. Pljesa Marija Markovic, R. Pavlovic Aleksandra Krivokapic, V Zoran TI Deletion and Single Nucleotide Polymorphisms in Common Glutathione-S Transferases Contribute to Colorectal Cancer Development SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Glutathione-S transferases; GST polymorphism; Carcinogenesis; Colorectal cancer development ID Colorectal cancer; Glutathione-S transferases; GST polymorphism; Carcinogenesis; Colorectal cancer development AB Glutathione-S transferases (GSTs) are xenobiotic-conjugation enzymes involved in the detoxification process of heterocyclic aromatic amines and polycyclic aromatic hydrocarbons, widely recognized risk factors of colorectal cancer (CRC) development. Polymorphism in GSTs often leads to alteration or complete lack of enzyme activity, which might have an effect on CRC carcinogenesis. Aim of this study was to investigate GST gene variants as risk factors in patients with CRC. A total of 523 CRC patients administered for surgical resection and 400 matched controls were included. Deletion polymorphism of GSTs M1 and T1 was investigated by polymerase chain reaction. Single nucleotide polymorphism of GSTA1 and P1 was investigated by restriction fragment length polymorphism method. The association between GST genotype and risk of CRC development was found in carriers of GSTT1-null and GSTP1-variant genotypes individually (p = 0.050 and p = 0.016, respectively). Furthermore, statistically significant association was found when combination of GSTP1-variant genotype with any of other three common GST genotypes was analyzed with respect to CRC susceptibility. Additionally, patients with combined GSTM1-null/GSTT1-null/GSTA1 low-activity/GSTP1-variant genotype showed 2.71-fold increased risk of developing CRC (p = 0.037). This study supports hypothesis that GST polymorphisms might have an important role in the process of the CRC development. Additionally, GSTM1-null/ GSTT1-null/ GSTA1 low-activity/ GSTP1-variant genotype could be combination of GST genotypes whose carriers are more prone to CRC development. C1 [Lalosevic, Lj. Stojkovic Milica] Clinical center of Serbia, Clinic of gastroenterology and hepatology, 11000 Belgrade, Serbia. [Coric, M Vesna] Belgrade University, Faculty of Pharmacy, Institute of Medical Biochemistry, 11000 Belgrade, Serbia. [Pekmezovic, D Tatjana] University of Belgrade, Faculty of Medicine, 11000 Belgrade, Serbia. [Simic, P Tatjana] Belgrade University, Faculty of Pharmacy, Institute of Medical Biochemistry, 11000 Belgrade, Serbia. [Ercegovac, S. Pljesa Marija] Belgrade University, Faculty of Pharmacy, Institute of Medical Biochemistry, 11000 Belgrade, Serbia. [Markovic, R. Pavlovic Aleksandra] Clinical center of Serbia, Clinic of gastroenterology and hepatology, 11000 Belgrade, Serbia. [Krivokapic, V Zoran] University of Belgrade, Faculty of Medicine, 11000 Belgrade, Serbia. RP Krivokapic, VZ (reprint author), University of Belgrade, Faculty of Medicine, 11000 Belgrade, Serbia. EM jagodica85@gmail.com CR Siegel RL, Miller KD, Jemal A, 2017 Jan, Cancer statistics, 2017. CA Cancer J Clin 67(1):7–30 Cunningham D, Atkin W, Lenz H-J, Lynch HT, Minsky B, Nordlinger B, Starling N, 2010, Colorectal cancer. Lancet Lond Engl 375(9719):1030–1047 Fleming M, Ravula S, Tatishchev SF, Wang HL, 2012, Colorectal carcinoma: pathologic aspects. J Gastrointest Oncol 3(3):153–173 Giglia MD, Chu DI, 2016, Familial colorectal Cancer: understanding the alphabet soup. Clin Colon Rectal Surg 29(3):185–195 Haggar FA, Boushey RP, 2009, Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg 22(4):191–197 Ramsey SD, Holmes RS, McDermott CL, Blough DK, Petrin KL, Poole EM et al, 2012, A comparison of approaches for association studies of polymorphisms and colorectal Cancer risk. Color Dis Off J Assoc Coloproctol G B Irel 14(9):e573–e586 Song Q-B, Wang Q, Hu W-G, 2014, A systemic review of glutathione S-transferase P1 Ile105Val polymorphism and colorectal cancer risk. Chin J Cancer Res Chung-Kuo Yen Cheng Yen Chiu 26(3):255–267 Mannervik B, Danielson UH, 1988, Glutathione transferases– structure and catalytic activity. CRC Crit Rev Biochem 23(3): 283–337 Koh W-P, Nelson HH, Yuan J-M, Van den Berg D, Jin A, Wang R et al, 2011, Glutathione S-transferase, GST, gene polymorphisms, cigarette smoking and colorectal cancer risk among Chinese in Singapore. Carcinogenesis 32(10):1507–1511 Edge SB, Compton CC, 2010, The American joint committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol 17(6):1471–1474 Abdel-Rahman SZ, el-Zein RA, Anwar WA, Au WW, 1996, A multiplex PCR procedure for polymorphic analysis of GSTM1 and GSTT1 genes in population studies. Cancer Lett 107(2):229– 233 Ping J, Wang H, Huang M, Liu Z-S, 2006, Genetic analysis of glutathione S-transferase A1 polymorphism in the Chinese population and the influence of genotype on enzymatic properties. Toxicol Sci Off J Soc Toxicol 89(2):438–443 Zhunussova G, Zhunusbekova B, Djansugurova L, 2015, Association between glutathione S-transferase M1 and T1 polymorphisms and colorectal cancer risk in patients from Kazakhstan. Clin Lab 61(1–2):161–168 Polimanti R, Carboni C, Baesso I, Piacentini S, Iorio A, De Stefano GF et al, 2013, Genetic variability of glutathione S-transferase enzymes in human populations: functional inter-ethnic differences in detoxification systems. Gene 512(1):102–107 Economopoulos KP, Sergentanis TN, 2010, GSTM1, GSTT1, GSTP1, GSTA1 and colorectal cancer risk: a comprehensive meta- analysis. Eur J Cancer Oxf Engl 46(9):1617–1631 van der Logt EMJ, Bergevoet SM, Roelofs HMJ, van Hooijdonk Z, te Morsche RHM, Wobbes T, de Kok JB, Nagengast FM, Peters WH, 2 0 0 4 , G e n e t i c p o l y m o r p h i s m s i n U D P - glucuronosyltransferases and glutathione S-transferases and colorectal cancer risk. Carcinogenesis 25(12):2407–2415 Martinez C, Martin F, Fernandez JM, Garcia-Martin E, Sastre J, Diaz-Rubio M, Agundez JAG, Ladero JM, 2006, Glutathione Stransferases mu 1, theta 1, pi 1, alpha 1 and mu 3 genetic polymorphisms and the risk of colorectal and gastric cancers in humans. Pharmacogenomics 7(5):711–718 Kury S, Buecher B, Robiou-du-Pont S, Scoul C, Colman H, Le Neel Tet al, 2008, Low-penetrance alleles predisposing to sporadic colorectal cancers: a French case-controlled genetic association study. BMC Cancer 8:326 Hezova R, Bienertova-Vasku J, SachlovaM, Brezkova V, Vasku A, Svoboda M, Radova L, Kiss I, Vyzula R, Slaby O, 2012, Common polymorphisms in GSTM1, GSTT1, GSTP1, GSTA1 and susceptibility to colorectal cancer in the central European population. Eur J Med Res 17(1):17 Tijhuis MJ, Wark PA, Aarts JMMJG, Visker MHPW, Nagengast FM, Kok FJ et al, 2005, GSTP1 and GSTA1 polymorphisms interact with cruciferous vegetable intake in colorectal adenoma risk. Cancer Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol 14(12):2943–2951 Little J, Sharp L, Masson LF, Brockton NT, Cotton SC, Haites NE, Cassidy J, 2006, Colorectal cancer and genetic polymorphisms of CYP1A1, GSTM1 and GSTT1: a case-control study in the Grampian region of Scotland. Int J Cancer 119(9):2155–2164 Li J, Xu W, Liu F, Huang S, He M, 2015, GSTM1 polymorphism contribute to colorectal cancer in Asian populations: a prospective meta-analysis. Sci Rep 5:12514 Available from: https://www.ncbi. nlm.nih.gov/pmc/articles/PMC4649893/ de Bruin WC, Wagenmans MJ, Peters WH, 2000, Expression of glutathione S-transferase alpha, P1-1 and T1-1 in the human gastrointestinal tract. Jpn J Cancer Res Gann 91(3):310–316 Liao C, Cao Y, Wu L, Huang J, Gao F, 2010, An updating metaanalysis of the glutathione S-transferase T1 polymorphisms and colorectal cancer risk: a HuGE review. Int J Color Dis 25(1):25–37 Qin X, Zhou Y, Chen Y, Li N, Chen B, Yang P, Wu XT, 2013, Glutathione S-transferase T1 gene polymorphism and colorectal cancer risk: an updated analysis. Clin Res Hepatol Gastroenterol 37(6):626–635 Wan H, Zhou Y, Yang P, Chen B, Jia G, Wu X, 2010, Genetic polymorphism of glutathione S-transferase T1 and the risk of colorectal cancer: a meta-analysis. Cancer Epidemiol 34(1):66–72 Masood N,Mubashar A, Yasmin A, 2015, Epidemiological factors related to GSTM1 and GSTT1 genes deletion in colon and rectum cancers: a case-control study. Cancer Biomark Sect Dis Markers 15(5):583–589 Strange RC, Fryer AA, 1999, The glutathione S-transferases: influence of polymorphismon cancer susceptibility. IARC Sci Publ 148: 231–249 Deng Q, He B, Pan Y, Sun H, Liu X, Chen J et al, 2015, Polymorphisms of GSTA1 contribute to elevated cancer risk: evidence from 15 studies. J BUON off J balk union. Oncologia 20(1): 287–295 Dogru-Abbasoglu S, Mutlu-Turkoglu U, Turkoglu S, Erbil Y, Barbaros U, Uysal M, Aykac-Toker G, 2002, Glutathione Stransferase- pi in malignant tissues and plasma of human colorectal and gastric cancers. J Cancer Res Clin Oncol 128(2):91–95 Tan Z, Feng M, Luo Y, Sun C, Fan Z, Tan Y, Fu B, Lang J, 2013, GSTP1 Ile105Val polymorphism and colorectal cancer risk: an updated analysis. Gene 527(1):275–282 Matakova T, Sivonova M, Halasova E, Mistuna D, Dzian A, Masar J, Dobrota D, 2009, Polymorphisms of biotransforming enzymes, GSTs, and their association with colorectal cancer in the Slovak population. Neoplasma 56(5):422–427 Wang J, Jiang J, Zhao Y, Gajalakshmi V, Kuriki K, Suzuki S, Nagaya T, Nakamura S, Akasaka S, Ishikawa H, Tokudome S, 2011, Genetic polymorphisms of glutathione S-transferase genes and susceptibility to colorectal cancer: a case-control study in an Indian population. Cancer Epidemiol 35(1):66–72 Kassab A, Msolly A, Lakhdar R, Gharbi O, Miled A, 2014, Polymorphisms of glutathione-S-transferases M1, T1, P1 and susceptibility to colorectal cancer in a sample of the Tunisian population. Med Oncol Northwood Lond Engl 31(1):760 Ates NA, Tamer L, Ates C, Ercan B, Elipek T, Ocal K et al, 2005, Glutathione S-transferaseM1, T1, P1 genotypes and risk for development of colorectal cancer. Biochem Genet 43(3–4):149–163 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1579 EP 1587 DI 10.1007/s12253-019-00589-1 PG 9 ER PT J AU Eiro, N Carrion, FJ Cid, S Andicoechea, A Garcia-Muniz, LJ Gonzalez, OL Vizoso, JF AF Eiro, Noemi Carrion, Francisco Juan Cid, Sandra Andicoechea, Alejandro Garcia-Muniz, Luis Jose Gonzalez, O Luis Vizoso, J Francisco TI Toll-Like Receptor 4 and Matrix Metalloproteases 11 and 13 as Predictors of Tumor Recurrence and Survival in Stage II Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prognostic factor; Stage II colorectal cancer; Survival; TLR4; MMP11; MMP13 ID Prognostic factor; Stage II colorectal cancer; Survival; TLR4; MMP11; MMP13 AB Current clinical-pathologic stratification factors do not allow clear identification of high-risk stage II colorectal cancer (CRC) patients. Therefore, the identification of additional prognostic markers is desirable. Toll-like receptor (TLR)-4 is activated during tumorigenesis and matrix metalloproteases (MMPs) are involved in invasion and metastasis.We aimed to evaluate the expression and clinical relevance of TLR4, MMP11 and MMP13 for patients with stage II CRC. Immunohistochemistry was used to study the expression of TLR4, MMP11 and MMP13 in 96 patients with stage II CRC. We measured the global expression and the expression by different cell types (tumor cells, cancer-associated fibroblasts (CAFs) and mononuclear inflammatory cells (MICs)). The potential relationship between expressions of factors and different prognostic variables were evaluated. Our results show significant relationships between either TLR4 expression by tumor cells and MMP11 expression by CAFs and high risk of tumor recurrence. In addition, the concurrence of age ≥ 75 years and the non-expression of MMP11 by CAFs identify a subgroup of patients with a good prognosis. Our results show that TLR4 expression by tumor cells and MMP11 expression by CAFs may to improve the identification of patients with stage II CRC with a high-risk of relapse. C1 [Eiro, Noemi] Fundacion Hospital de Jove, Unidad de Investigacion, Avda. Eduardo Castro 161, 33290 Gijon, Asturias, Spain. [Carrion, Francisco Juan] Fundacion Hospital de Jove, Unidad de Investigacion, Avda. Eduardo Castro 161, 33290 Gijon, Asturias, Spain. [Cid, Sandra] Fundacion Hospital de Jove, Unidad de Investigacion, Avda. Eduardo Castro 161, 33290 Gijon, Asturias, Spain. [Andicoechea, Alejandro] Fundacion Hospital de Jove, Servicio de Cirugia General, 33290 Gijon, Asturias, Spain. [Garcia-Muniz, Luis Jose] Fundacion Hospital de Jove, Unidad de Investigacion, Avda. Eduardo Castro 161, 33290 Gijon, Asturias, Spain. [Gonzalez, O Luis] Fundacion Hospital de Jove, Unidad de Investigacion, Avda. Eduardo Castro 161, 33290 Gijon, Asturias, Spain. [Vizoso, J Francisco] Fundacion Hospital de Jove, Unidad de Investigacion, Avda. Eduardo Castro 161, 33290 Gijon, Asturias, Spain. RP Vizoso, JF (reprint author), Fundacion Hospital de Jove, Unidad de Investigacion, 33290 Gijon, Spain. EM investigacion@hospitaldejove.com CR Tenesa A, Dunlop MG, 2009, New insights into the aetiology of colorectal cancer from genome-wide association studies. Nat Rev Genet 10(6):353–358 Jemal A, Siegel R, Xu J,Ward E, 2010, Cancer statistics, 2010. CA Cancer J Clin 60(5):277–300 Sobin LH, Gospodarowicz MK,Wittekind C, 2011, TNM classification of malignant tumours. Wiley Bosman FT, Carneiro F, Hruban RH, Theise ND, 2010, WHO classification of tumours of the digestive system. vol Ed. 4. World Health Organization, Jessup JM,McGinnis LS, Steele GD Jr,Menck HR,Winchester DP, 1996, The National Cancer Data Base. Report on colon cancer. Cancer 78(4):918–926 Edge SB, Compton CC, 2010, The American joint committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol 17(6):1471–1474 O'Connell JB, Maggard MA, Ko CY, 2004, Colon cancer survival rates with the new American joint committee on Cancer sixth edition staging. J Natl Cancer Inst 96(19):1420–1425 La TorreM, Lorenzon L, Pilozzi E, Barucca V, CavalliniM, Ziparo V, Ferri M, 2012, Number of harvested lymph nodes is the main prognostic factor in stage IIa colorectal cancer patients. J Surg Oncol 106(4):469–474 Santos C, Lopez-Doriga A, Navarro M, Mateo J, Biondo S, Martinez Villacampa M, Soler G, Sanjuan X, Paules MJ, Laquente B, Guino E, Kreisler E, Frago R, Germa JR, Moreno V, Salazar R, 2013, Clinicopathological risk factors of stage II colon cancer: results of a prospective study. Color Dis 15(4):414–422 Hari DM, Leung AM, Lee JH, Sim MS, Vuong B, Chiu CG, Bilchik AJ, 2013, AJCC Cancer staging manual 7th edition criteria for colon cancer: do the complex modifications improve prognostic assessment? J Am Coll Surg 217(2):181–190 Fang SH, Efron JE, Berho ME, Wexner SD, 2014, Dilemma of stage II colon cancer and decision making for adjuvant chemotherapy. J Am Coll Surg 219(5):1056–1069 Compton C, Fenoglio-PreiserCM, Pettigrew N, Fielding LP, 2000, American joint committee on Cancer prognostic factors consensus conference: colorectal working group. Cancer 88(7):1739–1757 Akagi Y, Shirouzu K, Kinugasa T, 2013, Extramural extension as indicator for postoperative adjuvant chemotherapy in stage IIA, pT3N0, colon cancer. J Surg Oncol 108(6):358–363 Stocchi L, Fazio VW, Lavery I, Hammel J, 2011, Individual surgeon, pathologist, and other factors affecting lymph node harvest in stage II colon carcinoma. Is a minimum of 12 examined lymph nodes sufficient? Ann Surg Oncol 18(2):405–412 Harris EI, Lewin DN, Wang HL, Lauwers GY, Srivastava A, Shyr Y, Shakhtour B, Revetta F, Washington MK, 2008, Lymphovascular invasion in colorectal cancer: an interobserver variability study. Am J Surg Pathol 32(12):1816–1821 Rakoff-Nahoum S, Medzhitov R, 2009, Toll-like receptors and cancer. Nat Rev Cancer 9(1):57–63 Mantovani A, Allavena P, Sica A, Balkwill F, 2008, Cancer-related inflammation. Nature 454(7203):436–444 Castro FA, Forsti A, Buch S, Kalthoff H, Krauss C, Bauer M, Egberts J, Schniewind B, Broering DC, Schreiber S, Schmitt M, Hampe J, Hemminki K, Schafmayer C, 2011, TLR-3 polymorphism is an independent prognostic marker for stage II colorectal cancer. Eur J Cancer 47(8):1203–1210 O'Leary DP, Bhatt L,Woolley JF, Gough DR,Wang JH, Cotter TG, Redmond HP, 2012, TLR-4 signalling accelerates colon cancer cell adhesion via NF-kappaB mediated transcriptional up-regulation of Nox-1. PLoS One 7(10):e44176 GrimmM,KimM, Rosenwald A,HeemannU, Germer CT,Waaga- Gasser AM, GasserM(2010, Toll-like receptor, TLR, 7 and TLR8 expression on CD133+ cells in colorectal cancer points to a specific role for inflammation-induced TLRs in tumourigenesis and tumour progression. Eur J Cancer 46(15):2849–2857 Rayburn ER, Wang W, Zhang R, Wang H, 2007, Experimental therapy for colon cancer: anti-cancer effects of TLR9 agonism, combination with other therapeutic modalities, and dependence upon p53. Int J Oncol 30(6):1511–1519 Eiro N, Gonzalez L, Gonzalez LO, Fernandez-Garcia B, Andicoechea A, Barbon E, Garcia-Muniz JL, Vizoso FJ, 2013, Toll-like receptor-4 expression by stromal fibroblasts is associated with poor prognosis in colorectal cancer. J Immunother 36(6):342– 349 Nelson AR, Fingleton B, Rothenberg ML, Matrisian LM, 2000, Matrix metalloproteinases: biologic activity and clinical implications. J Clin Oncol 18(5):1135–1149 Manes S, Llorente M, Lacalle RA, Gomez-Mouton C, Kremer L, Mira E, Martinez AC, 1999, The matrix metalloproteinase-9 regulates the insulin-like growth factor-triggered autocrine response in DU-145 carcinoma cells. J Biol Chem 274(11):6935–6945 Noe V, Fingleton B, Jacobs K, Crawford HC, Vermeulen S, Steelant W, Bruyneel E, Matrisian LM, Mareel M, 2001, Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci 114(Pt 1):111–118 Egeblad M, Werb Z, 2002, New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2(3):161–174 Turk V, Kos J, Turk B, 2004, Cysteine cathepsins, proteases)–on the main stage of cancer? Cancer Cell 5(5):409–410 Fingleton B, Vargo-Gogola T, Crawford HC, Matrisian LM, 2001, Matrilysin [MMP-7] expression selects for cells with reduced sensitivity to apoptosis. Neoplasia 3(6):459–468 Chen R, Cui J, Xu C, Xue T, Guo K, Gao D, Liu Y, Ye S, Ren Z, 2012, The significance ofMMP-9 overMMP-2 in HCC invasiveness and recurrence of hepatocellular carcinoma after curative resection. Ann Surg Oncol 19(Suppl 3):S375–S384 Koskensalo S, Louhimo J, Nordling S, Hagstrom J, Haglund C, 2011, MMP-7 as a prognostic marker in colorectal cancer. Tumour Biol 32(2):259–264 Escaff S, Fernandez JM, Gonzalez LO, Suarez A, Gonzalez-Reyes S, Gonzalez JM, Vizoso FJ, 2010, Study of matrix metalloproteinases and their inhibitors in prostate cancer. Br J Cancer 102(5):922– 929 Gonzalez LO, Pidal I, Junquera S, CorteMD, Vazquez J, Rodriguez JC, Lamelas ML, Merino AM, Garcia-Muniz JL, Vizoso FJ, 2007, Overexpression of matrix metalloproteinases and their inhibitors in mononuclear inflammatory cells in breast cancer correlates with metastasis-relapse. Br J Cancer 97(7):957–963 Pesta M, Topolcan O, Holubec L Jr, Rupert K, Cerna M, Holubec LS, Treska V, Finek J, Cerny R, 2007, Clinicopathological assessment and quantitative estimation of the matrix metalloproteinases MMP-2 and MMP-7 and the inhibitors TIMP-1 and TIMP-2 in colorectal carcinoma tissue samples. Anticancer Res 27(4A): 1863–1867 Asano T, Tada M, Cheng S, Takemoto N, Kuramae T, Abe M, Takahashi O, Miyamoto M, Hamada J, Moriuchi T, Kondo S, 2008, Prognostic values of matrix metalloproteinase family expression in human colorectal carcinoma. J Surg Res 146(1):32–42 Murray GI, DuncanME, O'Neil P,MelvinWT, Fothergill JE, 1996, Matrix metalloproteinase-1 is associated with poor prognosis in colorectal cancer. Nat Med 2(4):461–462 Curran S, Murray GI, 1999, Matrix metalloproteinases in tumour invasion and metastasis. J Pathol 189(3):300–308 Lyall MS, Dundas SR, Curran S, Murray GI, 2006, Profiling markers of prognosis in colorectal cancer. Clin Cancer Res 12(4): 1184–1191 Jensen SA, Vainer B, Bartels A, Brunner N, Sorensen JB, 2010, Expression of matrix metalloproteinase 9, MMP-9, and tissue inhibitor of metalloproteinases 1, TIMP-1, by colorectal cancer cells and adjacent stroma cells–associations with histopathology and patients outcome. Eur J Cancer 46(18):3233–3242 Chu D, Zhao Z, Zhou Y, Li Y, Li J, Zheng J, Zhao Q, Wang W, 2012, Matrix metalloproteinase-9 is associated with relapse and prognosis of patients with colorectal cancer. Ann Surg Oncol 19(1):318–325 Akishima-Fukasawa Y, Ishikawa Y, Akasaka Y, Uzuki M, Inomata N,Yokoo T, Ishii R, Shimokawa R,Mukai K,KiguchiH, Suzuki K, Fujiwara M, Ogata K, Niino H, Sugiura H, Ichinose A, Kuroda Y, Kuroda D, Ishii T, 2011, Histopathological predictors of regional lymph node metastasis at the invasive front in early colorectal cancer. Histopathology 59(3):470–481 Gonzalez L, Eiro N, Gonzalez LO, Andicoechea A, Barbon E, Garcia-Muniz JL, Vizoso FJ, 2012, Effect of the expression of matrix metalloproteases and their tissue inhibitors on survival of patients with resectable colorectal cancer. Dig Dis Sci 57(8): 2063–2071 Curran S, Dundas SR, Buxton J, Leeman MF, Ramsay R, Murray GI, 2004, Matrix metalloproteinase/tissue inhibitors of matrix metalloproteinase phenotype identifies poor prognosis colorectal cancers. Clin Cancer Res 10(24):8229–8234 Eiro N, Fernandez-Garcia B, Vazquez J, Del Casar JM, Gonzalez LO, Vizoso FJ, 2015, A phenotype from tumor stroma based on the expression of metalloproteases and their inhibitors, associated with prognosis in breast cancer. Oncoimmunology 4(7):e992222 Eiro N, Bermudez-Fernandez S, Fernandez-Garcia B, Atienza S, Beridze N, Escaf S, Vizoso FJ, 2014, Analysis of the expression of interleukins, interferon β, and nuclear factor-κ B in prostate Cancer and their relationship with biochemical recurrence. J Immunother 37(7):366–373 Fernandez-Garcia B, Eiro N, Marin L,Gonzalez-Reyes S, Gonzalez LO, Lamelas ML, Vizoso FJ, 2014, Expression and prognostic significance of fibronectin and matrix metalloproteases in breast cancer metastasis. Histopathology 64(4):512–522 Gonzalez-Reyes S, Marin L, Gonzalez L, Gonzalez LO, del Casar JM, Lamelas ML, Gonzalez-Quintana JM, Vizoso FJ, 2010, Study of TLR3, TLR4 and TLR9 in breast carcinomas and their association with metastasis. BMC Cancer 10(1):665 Nunez NG, Andreani V, Crespo MI, Nocera DA, Breser ML, Moron G, Dejager L, Libert C, Rivero V, Maccioni M, 2012, IFNbeta produced by TLR4-activated tumor cells is involved in improving the antitumoral immune response. Cancer Res 72(3): 592–603 Huang B, Zhao J, Unkeless JC, Feng ZH, Xiong H, 2008, TLR signaling by tumor and immune cells: a double-edged sword. Oncogene 27(2):218–224 MerrellMA, Ilvesaro JM, Lehtonen N, Sorsa T, Gehrs B, Rosenthal E, Chen D, Shackley B, Harris KW, Selander KS, 2006, Toll-like receptor 9 agonists promote cellular invasion by increasing matrix metalloproteinase activity. Mol Cancer Res 4(7):437–447 Lee Y, Kim H, Kim S, Kim KH, Chung JH, 2010, Activation of toll-like receptors 2, 3 or 5 induces matrixmetalloproteinase-1 and - 9 expression with the involvement of MAPKs and NF-kappaB in human epidermal keratinocytes. Exp Dermatol 19(8):e44–e49 Agarwal S, Misra R, Aggarwal A, 2010, Induction of metalloproteinases expression by TLR ligands in human fibroblast like synoviocytes from juvenile idiopathic arthritis patients. Indian J Med Res 131:771–779 Rath T, Stockle J, Roderfeld M, Tschuschner A, Graf J, Roeb E, 2011, Matrix metalloproteinase-13 is regulated by toll-like receptor- 9 in colorectal cancer cells and mediates cellular migration. Oncol Lett 2(3):483–488 Eiro N, Fernandez-Gomez J, Sacristan R, Fernandez-Garcia B, Lobo B, Gonzalez-Suarez J, Quintas A, Escaf S, Vizoso FJ, 2017, Stromal factors involved in human prostate cancer development, progression and castration resistance. J Cancer Res Clin Oncol 143(2):351–359 Delebecq TJ, Porte H, Zerimech F, Copin MC, Gouyer V, Dacquembronne E, Balduyck M, Wurtz A, Huet G, 2000, Overexpression level of stromelysin 3 is related to the lymph node involvement in non-small cell lung cancer. Clin Cancer Res 6(3): 1086–1092 Zhao ZS, Chu YQ, Ye ZY, Wang YY, Tao HQ, 2010, Overexpression of matrix metalloproteinase 11 in human gastric carcinoma and its clinicopathologic significance. Hum Pathol 41(5):686–696 Kessenbrock K, Plaks V,Werb Z, 2010, Matrix metalloproteinases: regulators of the tumor microenvironment. Cell 141(1):52–67 Bhowmick NA, Neilson EG, Moses HL, 2004, Stromal fibroblasts in cancer initiation and progression. Nature 432(7015):332–337 Peruzzi D, Mori F, Conforti A, Lazzaro D, De Rinaldis E, Ciliberto G, La Monica N, Aurisicchio L, 2009, MMP11: a novel target antigen for cancer immunotherapy. Clin Cancer Res 15(12):4104– 4113 McMillan DC, Hole DJ, McArdle CS, 2008, The impact of old age on cancer-specific and non-cancer-related survival following elective potentially curative surgery for dukes a/B colorectal cancer. Br J Cancer 99(7):1046–1049 Oliphant R, Horgan PG, Morrison DS, McMillan DC, 2015, Validation of a modified clinical risk score to predict cancerspecific survival for stage II colon cancer. Cancer Med 4(1):84–89 Lemmens VE, Janssen-Heijnen ML, Verheij CD, Houterman S, Repelaer van Driel OJ, Coebergh JW, 2005, Co-morbidity leads to altered treatment and worse survival of elderly patients with colorectal cancer. Br J Surg 92(5):615–623 Roxburgh CS, McMillan DC, 2010, Role of systemic inflammatory response in predicting survival in patients with primary operable cancer. Future Oncol 6(1):149–163 Turner N, Wong HL, Templeton A, Tripathy S, Whiti Rogers T, CroxfordM, Jones I, Sinnathamby M, Desai J, Tie J, Bae S, Christie M, Gibbs P, Tran B, 2016, Analysis of local chronic inflammatory cell infiltrate combined with systemic inflammation improves prognostication in stage II colon cancer independent of standard clinicopathologic criteria. Int J Cancer 138(3):671–678 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1589 EP 1597 DI 10.1007/s12253-019-00611-6 PG 9 ER PT J AU Yaghobi Joybari, A Azadeh, P Babaei, S Hosseini Kamal, F AF Yaghobi Joybari, Ali Azadeh, Payam Babaei, Siamak Hosseini Kamal, Farnaz TI Comparison of Capecitabine (Xeloda) vs. Combination of Capecitabine and Oxaliplatin (XELOX) as Neoadjuvant CRT for Locally Advanced Rectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chemotherapy; Capecitabine; Oxaliplatin; Rectal cancer ID Chemotherapy; Capecitabine; Oxaliplatin; Rectal cancer AB We decided to compare pathologic complete response (pCR) and disease-free survival (DFS) in rectal adenocarcinoma patients who received neoadjuvant chemoradiotherapy (CRT) with capecitabine plus oxaliplatin (XELOX) or capecitabine (Xeloda). In this study, patients with non-metastatic locally advanced rectal cancer (tumor stages of T2, T3, or T4) with or without lymph node involvement were retrospectively included. Patients received concomitant radiation (50.4–54 Gy external beam radiation in 28 to 30 fractions) and neoadjuvant therapy as either Xeloda (capecitabine, 2500 mg/m2 concomitantly with radiation therapy) (42patients) or XELOX [(oxaliplatin (50 mg/m2 intravenously once a week for five weeks) and capecitabine)] (72 patients). Surgery was done eight weeks after CRT. The endpoints were pCR (defined as no evidence of viable tumoral cells) and DFS (the interval from the initial treatment to the first tumor recurrence). Rectal sphincter preservation via low-anterior resection (LAR) was achieved in 73.8% of Xeloda group which was similar to XELOX group (70.8%), P = 0.61. pCR was documented in 11 (26.9%) of Xeloda group and 26 patients (36.1%) of XELOX group (P = 0.27). Tumor recurrence was recorded in 97 patients (85.1%). Mean (±SD) DFS was 52.13 (±31.92) months (median = 48 months). Mean (95% CI) DFS was 129.42 (110.19 to 148.64) in Xeloda group vs. 122.77 (110.72 to 134.83) in XELOX group (P = 0.74). Addition of oxaliplatin to capecitabine as neoadjuvant CRT for locally advanced rectal cancer did not result in improved pCR or better DFS. C1 [Yaghobi Joybari, Ali] Shahid Beheshti University of Medical Sciences, School of Medicine, Department of Radiation Oncology, Arabi Ave., Velenjak, 1983963113 Tehran, Iran. [Azadeh, Payam] Shahid Beheshti University of Medical Sciences, School of Medicine, Department of Radiation Oncology, Arabi Ave., Velenjak, 1983963113 Tehran, Iran. [Babaei, Siamak] Shahid Beheshti University of Medical Sciences, School of Medicine, Department of Radiation Oncology, Arabi Ave., Velenjak, 1983963113 Tehran, Iran. [Hosseini Kamal, Farnaz] Shahid Beheshti University of Medical Sciences, School of Medicine, Department of Radiation Oncology, Arabi Ave., Velenjak, 1983963113 Tehran, Iran. RP Azadeh, P (reprint author), Shahid Beheshti University of Medical Sciences, School of Medicine, Department of Radiation Oncology, 1983963113 Tehran, Iran. EM payazad@yahoo.com CR Lorimer PD, Motz BM, Kirks RC, Boselli DM,Walsh KK, Prabhu RS, Hill JS, Salo JC, 2017, Pathologic complete response rates after neoadjuvant treatment in rectal Cancer: An analysis of the National Cancer Database. Ann Surg Oncol 24(8):2095–2103 Mariette C, Brouquet A, Tzanis D, Laurenzi A, de la Rochefordiere A, Mariani P et al, 2017, What is the impact of neoadjuvant chemoradiation on outcomes in gastro-intestinal cancer? J Visc Surg 154(3):185–195 Bosset JF, Collette L, Calais G, Mineur L, Maingon P, Radosevic- Jelic L et al, 2006, Chemotherapy with preoperative radiotherapy in rectal cancer. N Engl J Med 355(11):1114–1123 Wasserberg N, Kundel Y, Purim O, Keidar A, Kashtan H, Sadot E, Fenig E, Brenner B, 2014, Sphincter preservation in distal CT2N0 rectal cancer after preoperative chemoradiotherapy. Radiat Oncol 9: 233 Kye BH, Kim HJ, Kim JG, Kim SH, Shim BY, Lee NS, Cho HM, 2013, Short-term effects of neoadjuvant chemoradiation therapy on anorectal function in rectal cancer patients: a pilot study. Radiat Oncol 8:203 Schrag D, 2013, Evolving role of neoadjuvant therapy in rectal cancer. Curr Treat Options Oncol 14(3):350–364 Sauer R, Becker H, HohenbergerW, Rodel C,Wittekind C, Fietkau R et al, 2004, Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med 351(17):1731–1740 Zhao L, Bai C, Shao Y, Guan M, Jia N, Xiao Y, Qiu H, Zhang F, Yang T, Zhong G, Chen S, 2011, A phase II study of neoadjuvant chemoradiotherapy with oxaliplatin and capecitabine for rectal cancer. Cancer Lett 310(2):134–139 Hirsch BR, Zafar SY, 2011, Capecitabine in the management of colorectal cancer. Cancer Manag Res 3:79–89 Haddad P, Miraie M, Farhan F, Fazeli MS, Alikhassi A, Maddah- Safaei A, Aghili M, Kalaghchi B, Babaei M, 2017, Addition of oxaliplatin to neoadjuvant radiochemotherapy in MRI-defined T3, T4 or N+ rectal cancer: a randomized clinical trial. Asia Pac J Clin Oncol 13:416–422 Schmoll HJ, Tabernero J, Maroun J, de Braud F, Price T, Van Cutsem E et al, 2015, Capecitabine plus Oxaliplatin compared with fluorouracil/Folinic acid as adjuvant therapy for stage III Colon Cancer: final results of the NO16968 randomized controlled phase III trial. J Clin Oncol 33(32):3733–3740 Hospers GA, Punt CJ, Tesselaar ME, Cats A, Havenga K, Leer JW et al, 2007, Preoperative chemoradiotherapy with capecitabine and oxaliplatin in locally advanced rectal cancer. A phase I-II multicenter study of the Dutch colorectal Cancer group. Ann Surg Oncol 14(10):2773–2779 Gao YH, An X, Sun WJ et al, 2014, Evaluation of capecitabine and oxaliplatin administered prior to and then concomitant to radiotherapy in high risk locally advanced rectal cancer. J Surg Oncol 109(5): 478–482 Ricardi U, Racca P, Franco P, Munoz F, Fanchini L, Rondi N et al, 2013, Prospective phase II trial of neoadjuvant chemoradiotherapy with Oxaliplatin and Capecitabine in locally advanced rectal cancer, XELOXART). Med Oncol 30(2):581 Koeberle D, Burkhard R, von Moos R, Winterhalder R, Hess V, Heitzmann F, Ruhstaller T, Terraciano L, Neuweiler J, Bieri G, Rust C, Toepfer M, 2008, Phase II study of capecitabine and oxaliplatin given prior to and concurrently with preoperative pelvic radiotherapy in patients with locally advanced rectal cancer. Br J Cancer 98(7):1204–1209 Zorcolo L, Rosman AS, Restivo A, Pisano M, Nigri GR, Fancellu A, Melis M, 2012, Complete pathologic response after combined modality treatment for rectal cancer and long-term survival: a metaanalysis. Ann Surg Oncol 19(9):2822–2832 Maas M, Nelemans PJ, Valentini V, Das P, Rodel C, Kuo LJ et al, 2010, Long-term outcome in patients with a pathological complete response after chemoradiation for rectal cancer: a pooled analysis of individual patient data. Lancet Oncol 11(9):835–844 Carlomagno C, Farella A, Bucci L,D'Armiento FP, Pesce G, Pepe S et al, 2009, Neo-adjuvant treatment of rectal cancer with capecitabine and oxaliplatin in combination with radiotherapy: a phase II study. Ann Oncol 20(5):906–912 Rodel C, Liersch T, HermannRM, Arnold D, Reese T, HippMet al, 2007)Multicenter phase II trial of chemoradiation with oxaliplatin for rectal cancer. J Clin Oncol 25(1):110–117 O’Connell MJ, Colangelo LH, Beart RW, Petrelli NJ, Allegra CJ, Sharif S et al, 2014, Capecitabine and oxaliplatin in the preoperative multimodality treatment of rectal cancer: surgical end points from National Surgical Adjuvant Breast and bowel project trial R-04. J Clin Oncol 32(18):1927–1934 Gerard JP, Azria D, Gourgou-Bourgade S, Martel-Laffay I, Hennequin C, Etienne PL et al, 2010, Comparison of two neoadjuvant chemoradiotherapy regimens for locally advanced rectal cancer: results of the phase III trial ACCORD 12/0405-Prodige 2. J Clin Oncol 28(10):1638–1644 Midgley RS, Yanagisawa Y, Kerr DJ, 2009, Evolution of nonsurgical therapy for colorectal cancer. Nat Clin Pract Gastroenterol Hepatol 6(2):108–120 Sha A, Abadi S, Gill S, 2017, Utilization of capecitabine plus oxaliplatin and 5-fluorouracil/folinic acid plus oxaliplatin in the adjuvant treatment of stage IIB and stage III colon cancer: a multi-Centre, retrospective, chart review study. J Oncol Pharm Pract 1078155217718381 Ostwal V, Engineer R, Ramaswamy A, Sahu A, Zanwar S, Arya S, Chopra S, Bal M, Patil P, Desouza A, Saklani A, 2016, Surgical outcomes of post chemoradiotherapy unresectable locally advanced rectal cancers improve with interim chemotherapy, is FOLFIRINOX better than CAPOX? J Gastrointest Oncol 7(6): 958–967 Liu F, Yang L, Wu Y et al, 2016, CapOX as neoadjuvant chemotherapy for locally advanced operable colon cancer patients: a prospective single-arm phase II trial. Chin J Cancer Res 28(6):589–597 Yaghoubi A, Azadeh P, Sheibani K et al, 2011, Comparison of 5FU-base Chemoradiation with and without Eloxatin on pathologic complete response in neoadjuvant Chemoradiation of rectal cancer. Iran J Pathol 6(3):110–116 Das P, Skibber JM, Rodriguez-Bigas MA, Feig BW, Chang GJ, Wolff RA, Eng C, Krishnan S, Janjan NA, Crane CH, 2007, Predictors of tumor response and downstaging in patients who receive preoperative chemoradiation for rectal cancer. Cancer 109(9): 1750–1755 Wallin U, Rothenberger D, Lowry A, Luepker R, Mellgren A, 2013, CEA - a predictor for pathologic complete response after neoadjuvant therapy for rectal cancer. Dis Colon Rectum 56(7):859–868 Zeng WG, Liang JW, Wang Z, Zhang XM, Hu JJ, Hou HR, Zhou HT, Zhou ZX, 2015, Clinical parameters predicting pathologic complete response following neoadjuvant chemoradiotherapy for rectal cancer. Chin J Cancer 34(10):468–474 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1599 EP 1605 DI 10.1007/s12253-019-00587-3 PG 7 ER PT J AU Smolarz, B Michalska, MM Samulak, D Romanowicz, H Wojcik, L AF Smolarz, Beata Michalska, M Magdalena Samulak, Dariusz Romanowicz, Hanna Wojcik, Luiza TI Polymorphism of DNA Repair Genes via Homologous Recombination (HR) in Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ovarian cancer; DNA repair; Polymorphism; Gene ID Ovarian cancer; DNA repair; Polymorphism; Gene AB Ovarian cancer is one of the most common types of cancer in women. The repair system via homologous recombination repairs double-strand breaks (DSB) of DNA, which are the most mortal for cell, out of all DNA damages. The genes, which encode the double-strand break repairing proteins, are highly polymorphic and, taking into account the significance of the repaired defects for cancer development, it seems important to learn the role of the polymorphisms in ovarian cancer development. The aim of the study was to determine the relationship between DNA repair genes via homologous recombination (HR) and modulation of the risk of ovarian cancer. The following polymorphisms were analysed: XRCC3-Thr241Met (rs861539), XRCC2–41657C/T (rs718282), XRCC2-Arg188His (rs3218536), BRCA1-Q356R (rs1799950) and RAD51–135 G/C (rs1801320). The study group included 600 patients with ovarian cancer and 600 healthy controls. The PCR-RFLP (PCR-based restriction fragment length polymorphism) technique was applied for polymorphism analysis. Allele XRCC3-241Met (OR 0.85, 95% CI 0.72–0.99, p < 0.045), XRCC2-41657 T (OR 1.67, 95% CI 1.42–1.96, p < .0001), BRCA1-356R (OR 1.61; % CI 1.37–1.90, p < .0001) and RAD51–135C (OR 5.16; 95% CI 4.29–6.20, p < .0001) strongly correlated with the neoplastic disease. No relationship was observed between the studied polymorphisms and the cancer progression stage according to FIGO classification. The results indicate that polymorphisms of DNA repair genes via homologous recombination may be associated with the incidence of ovarian cancer. Further research on larger groups is warranted to determine the influence of above-mentioned genetic variants on ovarian cancer risk. C1 [Smolarz, Beata] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. [Michalska, M Magdalena] Regional Hospital in Kalisz, Department of Obstetrics and GynaecologyKalisz, Poland. [Samulak, Dariusz] Regional Hospital in Kalisz, Department of Obstetrics and GynaecologyKalisz, Poland. [Romanowicz, Hanna] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. [Wojcik, Luiza] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, Rzgowska 281/289, 93-338 Lodz, Poland. RP Smolarz, B (reprint author), Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular Genetics, 93-338 Lodz, Poland. EM smolbea@wp.pl CR Edmondson RJ, Monaghan JM, 2001, The epidemiology of ovarian cancer. Int J Gynecol Cancer 11:423–429 Hakem R, 2008, DNA-damage repair; the good, the bad, and the ugly. EMBO J 27:589–605 Mitra S, Boldogh I, Izumi T, Hazra TK, 2001, Complexities of the DNA base excision repair pathway for repair of oxidative DNA damage. Environ Mol Mutagen 38:180–190 Broustas CG, Lieberman HB, 2014, DNA damage response genes and the development of cancer metastasis. Radiat Res 181:111–130 Davis JD, Lin SY, 2011, DNA damage and breast cancer. World J Clin Oncol 2:329–338 Slyskova J, Lorenzo Y, Karlsen A, Carlsen MH, Novosadova V, Blomhoff R, Vodicka P, CollinsAR, 2014, Both genetic and dietary factors underlie individual differences in DNA damage levels and DNA repair capacity. DNA Repair, Amst, 16:66–73 Jasin M, Rothstein R, 2013, Repair of Strand breaks by homologous recombination. Cold Spring Harb Perspect Biol 5:a012740 Lieber MR, 2010, The mechanism of double-Strand DNA break repair by the nonhomologousDNAend joining pathway. Annu Rev Biochem 79:181–211 Davis AJ, ChenDJ, 2013, DNA double strand break repair via nonhomologous end-joining. Transl Cancer Res 2:130–143 Genois MM, Paquet ER, Laffitte MCN, Maity R, Rodrigue A, Ouellette M, Masson JY, 2014, DNA Repair Pathways in Trypanosomatids: from DNA Repair to Drug Resistance. Microbiol Mol Biol Rev 78:40–73 Wood RD, Mitchell M, Sgouros J, Lindahl T, 2001, Human DNA repair genes. Science 291:1284–1289 Sielska J, Matecka M, Dabrowska E, Jakubek E, Urbaniak M, 2015, What do women know about breast cancer prophylaxis and a healthy style of life? Rep Pract Oncol Radiother 20:321–327 Harris HR, Eke AC, Chavarro JE, Missmer SA, 2018, Fruit and vegetable consumption and risk of endometriosis. Hum Reprod 33: 715–727. , DOI 10.1093/humrep/dey014 Ling KS, Chen GD, Tsai HJ, Lee MS, Wang PH, Liu FS, 2005, Genetic changes in ovarian cancer. Taiwanese J Obstet Gynecol 44: 218–231 Toss A, Tomasello C, Razzaboni E, Contu G, Grandi G, Cagnacci A, Schilder RJ, Cortesi L, 2015, Hereditary ovarian Cancer: not only BRCA 1 and 2 genes. Biomed Res Int 2015:341723 Orsulic S, Li Y, Soslow RA, Vitale-Cross LA, Gutkind JS, Varmus HE, 2002, Induction of ovarian cancer by defined multiple genetic changes in a mouse model system. Cancer Cell 1:53–62 Hollis RL, Gourley C, 2016, Genetic and molecular changes in ovarian cancer. Cancer Biol Med 13:236–247 Kappil MA, Liao Y, Terry MB, Santella RM, 2016, DNA repair gene expression levels as indicators of breast cancer in the breast cancer family registry. Anticancer Res 36:4039–4044 Kidane D, Chae WJ, Czochor J, Eckert KA, Glazer PM, Bothwell ALM, Sweasy JB, 2014, Interplay between DNA repair and inflammation, and the link to cancer. Crit Rev Biochem Mol Biol 49:116–139 Hemminki K, Forsti A, Bermejo JL, 2005, Single nucleotide polymorphisms, SNPs, are inherited from parents and they measure heritable events. J Carcinog 4:2 Erichsen HC, Chanock SJ, 2004, SNPs in cancer research and treatment. Br J Cancer 90:747–751 Helleday T, 2003, Pathways for mitotic homologous recombination in mammalian cells. Mutat Res 532:103–115 Jackson SP, 2002, Sensing and repairing DNA double-strand breaks. Carcinogenesis 23:687–696 Li X, HeyerWD, 2008, Homologous recombination in DNArepair and DNA damage tolerance. Cell Res 18:99–113 GorbunovaV, Seluanov A, Mao Z, Hine C, 2007, Changes inDNA repair during aging. Nucleic Acids Res 35:7466–7474 Khanna KK, Jackson SP, 2001, DNA double-strand breaks: signaling, repair and the cancer connection. Nat Genet 27:247–254 Trzeciak A, Blasiak J, 2001, DNA repair in mammalian cells: mechanisms of homologous recombination. Postepy Biochem 47: 38–51 Talar-Wojnarowska R, Gasiorowska A, Olakowski M, Dranka- Bojarowska D, Lampe P, Smolarz B, Malecka-Panas E, 2016, Analysis of XRCC2 and XRCC3 gene polymorphisms in pancreatic cancer. Biomed Rep 4:236–240 Michalska MM, Samulak D, Romanowicz H, Jablonski F, Smolarz B, 2016, Association between single nucleotide polymorphisms, SNPs, of XRCC2 and XRCC3 homologous recombination repair genes and ovarian cancer in Polish women. Exp Mol Pathol 100: 243–247 Qureshi Z, Mahjabeen I, Baig R, Kayani M, 2014, Correlation between selected XRCC2, XRCC3 and RAD51 gene polymorphisms and primary breast cancer in women in Pakistan. Asian Pac J Cancer Prev 15:10225–10229 Smolarz B, Makowska M, Samulak D, Michalska MM, Mojs E, Wilczak M, Romanowicz H, 2015, Association between single nucleotide polymorphisms, SNPs, of XRCC2 and XRCC3 homologous recombination repair genes and triple-negative breast cancer in Polish women. Clin Exp Med 15:151–157 Du L,Xiong T, He Q,WangY, Shen J, PengY, Jia Q,Yang J, Zhang Y, Huang J, 2014, The Thr241Met polymorphism in the XRCC3 gene is associated with increased risk of cancer in Chinese mainland populations. Tumour Biol 35:1371–1376 KayaniMA, Khan S, Baig RM,Mahjabeen I, 2014, Association of RAD 51 135 G/C, 172 G/T and XRCC3 Thr241Met gene polymorphisms with increased risk of head and neck cancer. Asian Pac J Cancer Prev 15:10457–10462 ZhaiM,Wang Y, JiangMF(2015, Arg188His polymorphism in the XRCC2 gene and the risk of ovarian cancer: a meta-analysis. Genet Mol Res 14:10808–10815 Zhang Y,Wang H, Peng Y, Liu Y, Xiong T, Xue P, Du L, 2014, The Arg188His polymorphism in the XRCC2 gene and the risk of cancer. Tumour Biol 35:3541–3549 Michalska MM, Samulak D, Bienkiewicz J, Romanowicz H, Smolarz B, 2015, Association between -41657C/T single nucleotide polymorphism of DNA repair gene XRCC2 and endometrial cancer risk in Polish women. Pol J Pathol 66:67–71 Wang N, Dong XJ, Li Y, Guo W, Zhou RM, Zhang XJ, Wang SJ, 2007, The association of XRCC2 gene polymorphism with susceptibility to esophageal squamous cell carcinoma and gastric cardiac adenocarcinoma in China. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 24:538–543 Michalska MM, Samulak D, Romanowicz H, Smolarz B, 2015, Single Nucleotide Polymorphisms, SNPs, of RAD51-G172T and XRCC2-41657C/T Homologous Recombination Repair Genes and the Risk of Triple- Negative Breast Cancer in PolishWomen. Pathol Oncol Res 21:935–940 Michalska MM, Samulak D, Smolarz B, 2014, An association between the -41657 C/T polymorphism of X-ray repair crosscomplementing 2, XRCC2, gene and ovarian cancer. Med Oncol 31:300 Ricks-Santi LJ, Sucheston LE, Yang Y, Freudenheim JL, Isaacs CJ, Schwartz MD, Dumitrescu RG, Marian C, Nie J, Vito D, Edge SB, Shields PG, 2011, Association of Rad51 polymorphism with DNA repair in BRCA1 mutation carriers and sporadic breast cancer risk. BMC Cancer 11:278 Ricks-Santi LJ, Nie J, Marian C, Ochs-Balcom HM, Trevisan M, Edge SB, Kanaan Y, Freudenheim JL, Shields PG, 2013, BRCA1 polymorphisms and breast cancer epidemiology in the Western New York exposures and breast cancer, WEB, study. Genet Epidemiol 37:504–511 Lakhani SR, Manek S, Penault-Llorca F, Flanagan A, Arnout L, Merrett S, McGuffog L, Steele D, Devilee P, Klijn JG, Meijers- Heijboer H, Radice P, Pilotti S, Nevanlinna H, Butzow R, Sobol H, Jacquemier J, Lyonet DS, Neuhausen SL, Weber B, Wagner T, Winqvist R, Bignon YJ, Monti F, Schmitt F, Lenoir G, Seitz S, Hamman U, Pharoah P, Lane G, Ponder B, Bishop DT, Easton DF, 2004, Pathology of ovarian cancers in BRCA1 and BRCA2 carriers. Clin Cancer Res 10:2473–2481 Neff RT, Senter L, Salani R, 2017, BRCA mutation in ovarian cancer: testing, implications and treatment considerations. Ther Adv Med Oncol 9:519–531 KayaniMA, Khan S, Baig RM,Mahjabeen I, 2014, Association of RAD51 135G/C, 172G/T and XRCC3 Thr241Met gene polymorphisms with increased risk of head and neck cancer. Asian Pac J Cancer Prev 5:10457–10462 Zhou GW, Hu J, Peng XD, Li Q, 2011, RAD51 135G>C polymorphism and breast cancer risk: a metaanalysis. Breast Cancer Res Treat 125:529–535 Hu X, Sun S, 2015, RAD51 gene 135G/C polymorphism and ovarian cancer risk: ameta-analysis. Int J Clin ExpMed 8:22365–22370 Wang W, Li JL, He XF, Li AP, Cai YL, Xu N, Sun SM, Wu BY, 2013, Association between the RAD51 135 G>C polymorphism and risk of cancer: a meta-analysis of 19,068 cases and 22,630 controls. PLoS One 8:e75153 Zhang BB, Wang DG, Xuan C, Sun GL, Deng KF, 2014, Genetic 135G/C polymorphism of RAD51 gene and risk of cancer: a metaanalysis of 28,956 cases and 28,372 controls. Familial Cancer 13: 515–526 Wang WW, Spurdle AB, Kolachana P, Bove B, Modan B, Ebbers SM, Suthers G, TuckerMA, Kaufman DJ, DoodyMM, Tarone RE, Daly M, Levavi H, Pierce H, Chetrit A, kConFab, ABCFS/ CFRBCS, AJBCS, NISOC, Yechezkel GH, Chenevix-Trench G, Offit K, Godwin AK, Struewing JP, 2001, A single nucleotide polymorphism in the 5′ untranslated region of RAD51 and risk of cancer among BRCA1/2 mutation carriers. Cancer Epidemiol Biomark Prev 10:955–960 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1607 EP 1614 DI 10.1007/s12253-019-00604-5 PG 8 ER PT J AU Varga, G Nagy, Zs Demeter, J Kosztolanyi, Sz Szomor, Hussain, A Deak, B Schneider, T Plander, M Szendrei, T Varoczy, L Illes, Batai, Peto, M Mikala, G AF Varga, Gergely Nagy, Zsolt Demeter, Judit Kosztolanyi, Szabolcs Szomor, Arpad Hussain, Alizadeh Deak, Beata Schneider, Tamas Plander, Mark Szendrei, Tamas Varoczy, Laszlo Illes, Arpad Batai, Arpad Peto, Monika Mikala, Gabor TI Real World Efficacy and Safety Results of Ixazomib Lenalidomide and Dexamethasone Combination in Relapsed/Refractory Multiple Myeloma: Data Collected from the Hungarian Ixazomib Named Patient Program SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Multiple myeloma; Relapsed; Ixazomib; Named patient program ID Multiple myeloma; Relapsed; Ixazomib; Named patient program AB Ixazomib-Revlimid-Dexamethasone is an all-oral treatment protocol for multiple myeloma with a manageable tolerability profile which was available through a named patient program for Hungarian patients from December 2015 to April 2017.We analyzed the clinical characteristics and survival of 77 patients treated at 7 centers within this program. The majority of patients responded, we found complete response in 9, very good partial response in 8, partial response in 32, minor response or stable disease in 13 and progressive disease in 11 patients. Progression free survival was 11.4 months. There was a trend of longer progression free survival in those with 1 vs. >1 prior treatment, with equally good effectivity in standard risk and high risk cytogenetic groups. The adverse events were usually mild, none leading to permanent drug interruptions. There were 5 fatalities: 3 infections and 2 pulmonary embolisms. Our real word data support the use of Ixazomib-Revlimid-Dexamethasone as a highly effective and well tolerated oral treatment protocol for relapsed myeloma. C1 [Varga, Gergely] Semmelweis University, 3rd Department of Internal Medicine, Kutvolgyi ut 4, H-1125 Budapest, Hungary. [Nagy, Zsolt] Semmelweis University, 1st Department of Internal MedicineBudapest, Hungary. [Demeter, Judit] Semmelweis University, 1st Department of Internal MedicineBudapest, Hungary. [Kosztolanyi, Szabolcs] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Szomor, Arpad] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Hussain, Alizadeh] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Deak, Beata] National Institute of OncologyBudapest, Hungary. [Schneider, Tamas] National Institute of OncologyBudapest, Hungary. [Plander, Mark] Vas County Markusovszky HospitalSzombathely, Hungary. [Szendrei, Tamas] Vas County Markusovszky HospitalSzombathely, Hungary. [Varoczy, Laszlo] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Batai, Arpad] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Peto, Monika] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Mikala, Gabor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. RP Varga, G (reprint author), Semmelweis University, 3rd Department of Internal Medicine, H-1125 Budapest, Hungary. EM vargager@gmail.com CR Moreau P, Masszi T, Grzasko N, Bahlis NJ, Hansson M, Pour L, Sandhu I, Ganly P, Baker BW, Jackson SR, Stoppa AM, Simpson DR, Gimsing P, Palumbo A, Garderet L, Cavo M, Kumar S, Touzeau C, Buadi FK, Laubach JP, Berg DT, Lin J, di Bacco A, Hui AM, van de Velde H, Richardson PG, 2016, Oral Ixazomib, Lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med 374(17):1621–1634 Mateos MV, Masszi T, Grzasko N, Hansson M, Sandhu I, Pour L, Viterbo L, Jackson SR, Stoppa AM, Gimsing P, Hamadani M, Borsaru G, Berg D, Lin J, di Bacco A, van de Velde H, Richardson PG, Moreau P, 2017, Impact of prior therapy on the efficacy and safety of oral ixazomib-lenalidomide-dexamethasone vs. placebo-lenalidomide-dexamethasone in patients with relapsed/ refractory multiple myeloma in TOURMALINE-MM1. Haematologica 102(10):1767–1775 Al-Salama ZT, Garnock-Jones KP, Scott LJ, 2017, Ixazomib: a review in relapsed and/or refractory multiple myeloma. Target Oncol 12(4):535–542 Terpos E, Maouche N, Minarik J et al, 2017, "Real world" data on the efficacy and safety of Ixazomib in combination with Lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: a combined study from the Greek, Czech and UK databases. Blood, 130(Suppl 1), 3087 Ziff M, Cheesman S, Kyriakou C, 2017, Real world use of ixazomib with lenalidomide and dexamethasone for patients with relapsed and relapsed refractory multiple myeloma. EHA 2017 Abstract 1975 Terpos E, Katodritou E, Kotsopoulou M, 2017)BReal world^ data on the efficacy and safety of ixazomib in combination with lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: a study of the Greek myeloma study group. EHA 2017 Abstract 1249 Durie BG, Harousseau JL, Miguel JS et al, 2006, International uniform response criteria for multiple myeloma. Leukemia 20: 1467–1473 Leleu X, Masszi T, Bahlis NJ, Viterbo L, Baker B, Gimsing P, Maisnar V, Samoilova O, Rosinol L, Langer C, Song K, Izumi T, Cleeland C, Berg D, Lin HM, Zhu Y, Skacel T, Moreau P, Richardson PG, 2018, Patient-reported health-related quality of life from the phase III TOURMALINE-MM1 study of ixazomiblenalidomide- dexamethasone versus placebo-lenalidomidedexamethasone in relapsed/refractory multiple myeloma. Am J Hematol [Epub ahead of print] 93:985–993 Armoiry X, Connock M, Tsertsvadze A, Cummins E, Melendez- Torres GJ, Royle P, Clarke A, 2018, Ixazomib for relapsed or refractory multiple myeloma: review from an evidence review group on a NICE single technology appraisal. Pharmacoeconomics [Epub ahead of print] 36:1073–1081 Hari P, Lin HM, Zhu Yet al, 2018, Healthcare resource utilization with ixazomib or placebo plus lenalidomide-dexamethasone in the randomized, double-blind, phase 3 TOURMALINE-MM1 study in relapsed/refractory multiple myeloma. J Med Econ 29:1–6 [Epub ahead of print] Avet-Loiseau H, Bahlis NJ, ChngWJ, Masszi T, Viterbo L, Pour L, Ganly P, Palumbo A, Cavo M, Langer C, Pluta A, Nagler A, Kumar S, Ben-Yehuda D, Rajkumar SV, San-Miguel J, Berg D, Lin J, van de Velde H, Esseltine DL, di Bacco A, Moreau P, Richardson PG, 2017, Ixazomib significantly prolongs progression-free survival in high-risk relapsed/refractory myeloma patients. Blood 130(24): 2610–2618 Dimopoulos MA, Kaufman JL,White D, Cook G, Rizzo M, Xu Y, Fahrbach K, Gaudig M, Slavcev M, Dearden L, Lam A, 2018, A comparison of the efficacy of immunomodulatory-containing regimens in relapsed/refractory multiple myeloma: a network metaanalysis. Clin Lymphoma Myeloma Leuk 18(3):163–173 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1615 EP 1620 DI 10.1007/s12253-019-00607-2 PG 6 ER PT J AU Liu, Z Yang, M Wang, S Chen, Hp Guan, X Zhao, Zx Jiang, Z Quan, Jch Yang, Rk Wang, Xsh AF Liu, Zheng Yang, Ming Wang, Song Chen, Hai-peng Guan, Xu Zhao, Zhi-xun Jiang, Zheng Quan, Ji-chuan Yang, Run-kun Wang, Xi-shan TI GGN Promotes Tumorigenesis by Regulating Proliferation and Apoptosis in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Gametogenetin (GGN); Proliferation; Apoptosis ID Colorectal cancer; Gametogenetin (GGN); Proliferation; Apoptosis AB Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths worldwide. GGN is a germ cell-specific gene, but its function in CRC has been rarely reported to date. The aim of this study was to investigate the potential role of GGN in CRC tumorigenesis. Therefore, in this study, we examined the expression of GGN in CRC cell lines and tissues and its effects on cellular proliferation and apoptosis. We then explored the underlying mechanism. Our results showed that GGN was significantly overexpressed in both CRC cell lines and tissues. Silencing GGN robustly inhibited proliferation of CRC cells, and it also promoted apoptosis of CRC cells.Moreover, knockdown of GGN inhibited the expression of p-Akt in CRC cells. Taken together, these results showed that knockdown of GGN inhibits proliferation and promotes apoptosis of CRC cells through the PI3K/Akt signaling pathway. Our findings revealed for the first time a potential oncogenic role for GGN in CRC progress. This finding may provide a unique perspective on how a germ cell-specific gene might serve as a biomarker, or even as a therapeutic target, for CRC. C1 [Liu, Zheng] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Department of Colorectal Surgery, 17 Panjiayuan Nanli, Chaoyang Dist., 100021 Beijing, China. [Yang, Ming] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Department of Colorectal Surgery, 17 Panjiayuan Nanli, Chaoyang Dist., 100021 Beijing, China. [Wang, Song] The Second Affiliated Hospital of Harbin Medical University, Department of Colorectal Cancer SurgeryHarbin, China. [Chen, Hai-peng] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Department of Colorectal Surgery, 17 Panjiayuan Nanli, Chaoyang Dist., 100021 Beijing, China. [Guan, Xu] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Department of Colorectal Surgery, 17 Panjiayuan Nanli, Chaoyang Dist., 100021 Beijing, China. [Zhao, Zhi-xun] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Department of Colorectal Surgery, 17 Panjiayuan Nanli, Chaoyang Dist., 100021 Beijing, China. [Jiang, Zheng] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Department of Colorectal Surgery, 17 Panjiayuan Nanli, Chaoyang Dist., 100021 Beijing, China. [Quan, Ji-chuan] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Department of Colorectal Surgery, 17 Panjiayuan Nanli, Chaoyang Dist., 100021 Beijing, China. [Yang, Run-kun] The Second Affiliated Hospital of Harbin Medical University, Department of Colorectal Cancer SurgeryHarbin, China. [Wang, Xi-shan] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Department of Colorectal Surgery, 17 Panjiayuan Nanli, Chaoyang Dist., 100021 Beijing, China. RP Wang, Xsh (reprint author), Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Department of Colorectal Surgery, 100021 Beijing, China. EM wxshan1208@126.com CR Siegel RL, Miller KD, Jemal A, 2018, Cancer statistics, 2018. CA Cancer J Clin 68:7–30 ChenW, Zheng R, Baade PD et al, 2016, Cancer statistics in China, 2015. CA Cancer J Clin 66:115–132 Kuipers EJ, Grady WM, Lieberman D et al, 2015, Colorectal cancer. Nat Rev Dis Primers 1:15065 Fakih MG, 2015, Metastatic colorectal cancer: current state and future directions. J Clin Oncol 33:1809–1824 Brenner H, KloorM, Pox CP, 2014, Colorectal cancer. Lancet 383: 1490–1502 Colussi D, Brandi G, Bazzoli F, Ricciardiello L, 2013, Molecular pathways involved in colorectal cancer: implications for disease behavior and prevention. Int J Mol Sci 14:16365–16385 Johnston PG, 2014, Identification of clinically relevant molecular subtypes in colorectal cancer: the dawning of a new era. Oncologist 19:568–573 Lu B, Bishop CE, 2003, Mouse GGN1 and GGN3, two germ cellspecific proteins from the single gene GGN, interact with mouse POG and play a role in spermatogenesis. J Biol Chem 278:16289–16296 Jamsai D, Sarraj MA, Merriner DJ, Drummond AE, Jones KT, McLachlan RI, O’Bryan MK, 2011, GGN1 in the testis and ovary and its variance within the Australian fertile and infertile male population. Int J Androl 34:624–632 WangW, LiC,Chen Y, Teng L, CaoY,XuY, Pan H, AnR, 2018, Increased expression of GGN promotes tumorigenesis in bladder cancer and is correlated with poor prognosis. Gene 652:7–15 Lan ZJ, Hu Y, Zhang S, Li X, Zhou H, Ding J, Klinge CM, Radde BN, Cooney AJ, Zhang J, Lei Z, 2016, GGNBP2 acts as a tumor suppressor by inhibiting estrogen receptor α activity in breast cancer cells. Breast Cancer Res Treat 158:263–276 Yin F, Liu L, Liu X et al, 2014, Downregulation of tumor suppressor gene ribonuclease T2 and gametogenetin binding protein 2 is associated with drug resistance in ovarian cancer. Oncol Rep 32: 362–372 Tariq K, Ghias K, 2016, Colorectal cancer carcinogenesis: a review of mechanisms. Cancer Biol Med 13:120–135 Okugawa Y, Grady WM, Goel A, 2015, Epigenetic alterations in colorectal Cancer: emerging biomarkers. Gastroenterology 149: 1204–1225.e12 Chen A, Li J, Song L, Ji C, Boing M, Chen J, Brand-Saberi B, 2017, GGNBP2 is necessary for testis morphology and sperm development. Sci Rep 7:2998 Zhao Q, Zhou Y, Cao Z, Zhu H, Huang P, Lu B, 2005, Germ-cell specific protein gametogenetin protein 2, GGN2), expression in the testis, and association with intracellular membrane. Mol Reprod Dev 72:31–39 Stoian M, State N, Stoica V, Radulian G, 2014, Apoptosis in colorectal cancer. J Med Life 7:160–164 Galluzzi L, Lopez-Soto A, Kumar S, Kroemer G, 2016, Caspases connect cell-death signaling to organismal homeostasis. Immunity 44:221–231 Malinowsky K, Nitsche U, Janssen KP, Bader FG, Spath C, Drecoll E, Keller G, Hofler H, Slotta-Huspenina J, Becker KF, 2014, Activation of the PI3K/AKT pathway correlates with prognosis in stage II colon cancer. Br J Cancer 110:2081–2089 Danielsen SA, Eide PW, Nesbakken A et al, 2015, Portrait of the PI3K/AKT pathway in colorectal cancer. Biochim Biophys Acta 1855:104–121 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1621 EP 1626 DI 10.1007/s12253-019-00595-3 PG 6 ER PT J AU Esfandi, F Ghafouri-Fard, S Kholghi Oskooei, V Taheri, M AF Esfandi, Farbod Ghafouri-Fard, Soudeh Kholghi Oskooei, Vahid Taheri, Mohammad TI β-Secretase 1 and its Naturally Occurring Anti-Sense RNA are Down-Regulated in Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BACE1; BACE1-AS; Gastric cancer ID BACE1; BACE1-AS; Gastric cancer AB β-secretase (BACE1) and its naturally occurring anti-sense RNA (BACE1-AS) have established role in the pathologic process leading to Alzheimer’s disease. Their possible implication in the neoangiogenesis suggests that they might be involved in the tumorigenesis events as well. In the present study, we compared transcript levels of these genes in 30 gastric cancer samples and their adjacent non-cancerous tissues (ANCTs) to find whether their altered expression might facilitate discrimination of these two sets of samples. Expressions of both genes were associated with site of primary tumor. Both genes were significantly downregulated in tumoral tissues compared with ANCTs. Significant correlations were detected between transcript levels of these genes in both sets of samples. Transcript levels of BACE1 and BACE1-AS had the diagnostic power of 75% based on Receiver operating characteristic curve analysis. The current study provides evidences for contribution of BACE1 and BACE1-AS in gastric cancer evolution and suggests their potential as diagnostic markers. C1 [Esfandi, Farbod] Shahid Beheshti University of Medical Sciences, Department of Medical GeneticsTehran, Iran. [Ghafouri-Fard, Soudeh] Shahid Beheshti University of Medical Sciences, Department of Medical GeneticsTehran, Iran. [Kholghi Oskooei, Vahid] Shahid Beheshti University of Medical Sciences, Department of Medical GeneticsTehran, Iran. [Taheri, Mohammad] Shahid Beheshti University of Medical Sciences, Urogenital Stem Cell Research CenterTehran, Iran. RP Taheri, M (reprint author), Shahid Beheshti University of Medical Sciences, Urogenital Stem Cell Research Center, Tehran, Iran. EM mohammad_823@yahoo.com CR Willem M, Garratt AN, Novak B, Citron M, Kaufmann S, Rittger A, DeStrooper B, Saftig P, Birchmeier C, Haass C, 2006, Control of peripheral nerve myelination by the beta-secretase BACE1. Science 314:664–666 John V, 2006, Human beta-secretase, BACE, and BACE inhibitors: progress report. Curr Top Med Chem 6:569–578 Faghihi MA, Modarresi F, Khalil AM, Wood DE, Sahagan BG, Morgan TE, Finch CE, Laurent GS III, Kenny PJ, Wahlestedt C, 2008, Expression of a noncoding RNA is elevated in Alzheimer’s disease and drives rapid feed-forward regulation of β-secretase. Nat Med 14:723–730 Paris D, Quadros A, Patel N, DelleDonne A, Humphrey J, Mullan M, 2005, Inhibition of angiogenesis and tumor growth by beta and gamma-secretase inhibitors. Eur J Pharmacol 514:1–15 Nienhuser H, Schmidt T, 2018, Angiogenesis and anti-angiogenic therapy in gastric cancer. Int J Mol Sci 19(1):43 Sun X, He G, Qing H, Zhou W, Dobie F, Cai F, Staufenbiel M, Huang LE, SongW(2006, Hypoxia facilitates Alzheimer’s disease pathogenesis by up-regulating BACE1 gene expression. Proc Natl Acad Sci U S A 103:18727–18732 Zhang X, Zhou K,Wang RS, Cui JK, Lipton SA, Liao FF, Xu HX, ZhangYW(2007, Hypoxia-inducible factor 1 alpha, HIF-1 alpha)- mediated hypoxia increases BACE1 expression and beta-amyloid generation. J Biol Chem 282:10873–10880 Kitajima Y, Miyazaki K, 2013, The critical impact of HIF-1a on gastric cancer biology. Cancers, Basel, 5:15–26 Urano N, Fujiwara Y, Doki Y, Tsujie M, Yamamoto H, Miyata H, Takiguchi S,Yasuda T,YanoM,MondenM(2006)Overexpression of hypoxia-inducible factor-1 alpha in gastric adenocarcinoma. Gastric Cancer 9:44–49 Rossner S, Sastre M, Bourne K, Lichtenthaler SF, 2006, Transcriptional and translational regulation of BACE1 expression–implications for Alzheimer’s disease. Prog Neurobiol 79:95–111 Lange-Dohna C, Zeitschel U, Gaunitz F, Perez-Polo JR, Bigl V, Rossner S, 2003, Cloning and expression of the rat BACE1 promoter. J Neurosci Res 73:73–80 Huang T, Kang W, Zhang B, Wu F, Dong Y, Tong JH, Yang W, Zhou Y, Zhang L, Cheng AS, Yu J, K.F. To, 2016, miR- 508-3p concordantly silences NFKB1 and RELA to inactivate canonical NF-kappaB signaling in gastric carcinogenesis. Mol Cancer 15:9 Sharma SA, Tummuru MK, Blaser MJ, Kerr LD, 1998, Activation of IL-8 gene expression by helicobacter pylori is regulated by transcription factor nuclear factor-kappa B in gastric epithelial cells. J Immunol 160:2401–2407 Akiyama Y, Watkins N, Suzuki H, Jair KW, van Engeland M, Esteller M, Sakai H, Ren CY, Yuasa Y, Herman JG, Baylin SB, 2003, GATA-4 and GATA-5 transcription factor genes and potential downstream antitumor target genes are epigenetically silenced in colorectal and gastric cancer. Mol Cell Biol 23:8429–8439 Mei Z, Yan P, Tan X, Zheng S, Situ B, 2015, Transcriptional regulation of BACE1 by NFAT3 leads to enhanced amyloidogenic processing. Neurochem Res 40:829–836 Robbs BK, Cruz AL, Werneck MB, Mognol GP, Viola JP, 2008, Dual roles for NFAT transcription factor genes as oncogenes and tumor suppressors. Mol Cell Biol 28:7168–7181 Zali H, Rezaei-Tavirani M, Vafaee R, Rezaei-Tavirani M, 2013, Gastric cardia adenocarcinoma pathway analysis. Gastroenterol Hepatol Bed Bench 6:S11–S18 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1627 EP 1633 DI 10.1007/s12253-019-00621-4 PG 7 ER PT J AU Miyamura, Y Kagara, N Miyake, T Tanei, T Naoi, Y Shimoda, M Shimazu, K Kim, JS Noguchi, Sh AF Miyamura, Yukiko Kagara, Naofumi Miyake, Tomohiro Tanei, Tomonori Naoi, Yasuto Shimoda, Masafumi Shimazu, Kenzo Kim, Jin Seung Noguchi, Shinzaburo TI Drainage of Tumor-Derived DNA into Sentinel Lymph Nodes in Breast Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Circulating tumor DNA; Sentinel lymph node; PIK3CA mutation; Apoptosis ID Circulating tumor DNA; Sentinel lymph node; PIK3CA mutation; Apoptosis AB Circulating tumor DNA (ctDNA) is released from cancer cells by apoptosis or other mechanisms, and as tumor tissue contains both blood and lymphatic vessels, ctDNA can spread to local lymph nodes (LNs). We aimed to detect the tumor-derived free DNA in metastasis-free LNs in patients with breast cancers harboring the PIK3CA-H1047R mutation. One hundred twenty-three patients were evaluated and the PIK3CA-H1047R mutation was assayed in sentinel LNs (SLNs), non-SLNs without metastasis, and serum by digital PCR. The mutant DNA was more frequent in metastasis-free SLNs (21.6%) than in metastasis-free non- SLNs (8.6%; P = 0.038), and patients with mutation-positive SLNs were more likely to be positive for serum mutant DNA. Apoptosis in primary breast tumors was determined by TUNEL assay. The apoptotic index was significantly higher (P = 0.003) in patients with mutation-positive SLNs without metastasis (mean, 1.17%) than those with mutation-negative SLNs without metastasis (mean, 0.79%). It was also significantly higher (P = 0.006) in those with mutation-positive serum (mean, 1.41%) than in those with mutation-negative serum (mean, 0.86%). Furthermore, fragment size of PIK3CA-H1047R mutant DNA in metastatic-free SLN lysate used for the one-step nucleic acid amplification (OSNA) assay was short (<500 bp). These results support the theory that DNA is released from the primary tumor via apoptotic fragmentation. In conclusion, ctDNA is detectable in metastasis-free LNs and significantly more frequent in SLNs from patients with breast tumors harboring a high apoptotic index, consistent with drainage of ctDNA from apoptotic primary tumor cells into SLNs. C1 [Miyamura, Yukiko] Osaka University, Graduate School of Medicine, Department of Breast and Endocrine Surgery, 2-2-E10, Yamadaoka, Suita, 565-0871 Osaka, Japan. [Kagara, Naofumi] Osaka University, Graduate School of Medicine, Department of Breast and Endocrine Surgery, 2-2-E10, Yamadaoka, Suita, 565-0871 Osaka, Japan. [Miyake, Tomohiro] Osaka University, Graduate School of Medicine, Department of Breast and Endocrine Surgery, 2-2-E10, Yamadaoka, Suita, 565-0871 Osaka, Japan. [Tanei, Tomonori] Osaka University, Graduate School of Medicine, Department of Breast and Endocrine Surgery, 2-2-E10, Yamadaoka, Suita, 565-0871 Osaka, Japan. [Naoi, Yasuto] Osaka University, Graduate School of Medicine, Department of Breast and Endocrine Surgery, 2-2-E10, Yamadaoka, Suita, 565-0871 Osaka, Japan. [Shimoda, Masafumi] Osaka University, Graduate School of Medicine, Department of Breast and Endocrine Surgery, 2-2-E10, Yamadaoka, Suita, 565-0871 Osaka, Japan. [Shimazu, Kenzo] Osaka University, Graduate School of Medicine, Department of Breast and Endocrine Surgery, 2-2-E10, Yamadaoka, Suita, 565-0871 Osaka, Japan. [Kim, Jin Seung] Osaka University, Graduate School of Medicine, Department of Breast and Endocrine Surgery, 2-2-E10, Yamadaoka, Suita, 565-0871 Osaka, Japan. [Noguchi, Shinzaburo] Osaka University, Graduate School of Medicine, Department of Breast and Endocrine Surgery, 2-2-E10, Yamadaoka, Suita, 565-0871 Osaka, Japan. RP Kagara, N (reprint author), Osaka University, Graduate School of Medicine, Department of Breast and Endocrine Surgery, 565-0871 Osaka, Japan. EM kagaran@onsurg.med.osaka-u.ac.jp CR Lyman GH, Giuliano AE, Somerfield MR, Benson AB 3rd, Bodurka DC, Burstein HJ, Cochran AJ, Cody HS 3rd, Edge SB, Galper S, Hayman JA, Kim TY, Perkins CL, Podoloff DA, Sivasubramaniam VH, Turner RR, Wahl R, Weaver DL, Wolff AC, Winer EP, 2005, American Society of Clinical Oncology guideline recommendations for sentinel lymph node biopsy in early-stage breast cancer. J Clin Oncol 23:7703–7720. https://doi. org/10.1200/jco.2005.08.001 Morton DL, Wen DR, Wong JH, Economou JS, Cagle LA, Storm FK, Foshag LJ, Cochran AJ, 1992, Technical details of intraoperative lymphatic mapping for early stage melanoma. Arch Surg 127: 392–399 Giuliano AE, Kirgan DM, Guenther JM, Morton DL, 1994, Lymphatic mapping and sentinel lymphadenectomy for breast cancer. Ann Surg 220:391–398 discussion 398-401 Nathanson SD, 2003, Insights into the mechanisms of lymph node metastasis. Cancer 98:413–423. , DOI 10.1002/cncr.11464 Teramoto A, Shimazu K, Naoi Y, Shimomura A, Shimoda M, Kagara N, Maruyama N, Kim SJ, Yoshidome K, Tsujimoto M, Tamaki Y, Noguchi S, 2014, One-step nucleic acid amplification assay for intraoperative prediction of non-sentinel lymph node metastasis in breast cancer patients with sentinel lymph nodemetastasis. Breast 23:579–585. , DOI 10.1016/j.breast.2014.05.026 Shimazu K, Sato N, Ogiya A, Sota Y, Yotsumoto D, Ishikawa T, Nakamura S, Kinoshita T, Tsuda H, Ohi Y, Akiyama F, Noguchi S, 2018, Intraoperative nomograms, based on one-step nucleic acid amplification, for prediction of non-sentinel node metastasis and four or more axillary nodemetastases in breast Cancer patients with sentinel node metastasis. Ann Surg Oncol 25:2603–2611. https:// doi.org/10.1245/s10434-018-6633-0 Veronesi U, Paganelli G, Viale G, Luini A, Zurrida S, Galimberti V, IntraM,VeronesiP,Maisonneuve P, Gatti G,Mazzarol G, DeCicco C, Manfredi G, Fernandez JR, 2006, Sentinel-lymph-node biopsy as a staging procedure in breast cancer: update of a randomised controlled study. Lancet Oncol 7:983–990. , DOI 10. 1016/s1470-2045(06)70947-0 Krag DN, Anderson SJ, Julian TB, Brown AM, Harlow SP, Ashikaga T, Weaver DL, Miller BJ, Jalovec LM, Frazier TG, Noyes RD, Robidoux A, Scarth HM, Mammolito DM, McCready DR, Mamounas EP, Costantino JP, Wolmark N, 2007, Technical outcomes of sentinel-lymph-node resection and conventional axillary-lymph-node dissection in patients with clinically node-negative breast cancer: results from the NSABP B-32 randomised phase III trial. Lancet Oncol 8:881–888. https://doi. org/10.1016/s1470-2045(07)70278-4 Wong SL, Edwards MJ, Chao C, Tuttle TM, Noyes RD, Carlson DJ, Cerrito PB,McMastersKM(2001, Sentinel lymph node biopsy for breast cancer: impact of the number of sentinel nodes removed on the false-negative rate. J Am Coll Surg 192:684–689 discussion 689-691 Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A, 2013, Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 10:472–484. , DOI 10.1038/nrclinonc.2013.110 Diaz LA Jr, Bardelli A, 2014, Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol 32:579–586. , DOI 10. 1200/jco.2012.45.2011 Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, Thornton K, Agrawal N, Sokoll L, Szabo SA, Kinzler KW, Vogelstein B, Diaz LA Jr, 2008, Circulating mutant DNA to assess tumor dynamics. Nat Med 14:985–990. , DOI 10.1038/nm.1789 Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, Antonarakis ES, Azad NS, Bardelli A, Brem H, Cameron JL, Lee CC, Fecher LA, Gallia GL, Gibbs P, Le D, Giuntoli RL, Goggins M, Hogarty MD, Holdhoff M, Hong SM, Jiao Y, Juhl HH, Kim JJ, Siravegna G, Laheru DA, Lauricella C, Lim M, Lipson EJ, Marie SK, Netto GJ, Oliner KS, Olivi A, Olsson L, Riggins GJ, Sartore-Bianchi A, Schmidt K, Shih lM, Oba-Shinjo SM, Siena S, Theodorescu D, Tie J, Harkins TT, Veronese S, Wang TL, Weingart JD, Wolfgang CL, Wood LD, Xing D, Hruban RH, Wu J, Allen PJ, Schmidt CM, Choti MA, Velculescu VE, Kinzler KW, Vogelstein B, Papadopoulos N, Diaz LA Jr, 2014, Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 6:224ra224. , DOI 10.1126/scitranslmed.3007094 Oshiro C, Kagara N, Naoi Y, Shimoda M, Shimomura A, Maruyama N, Shimazu K, Kim SJ, Noguchi S, 2015, PIK3CA mutations in serum DNA are predictive of recurrence in primary breast cancer patients. Breast Cancer Res Treat 150:299–307. , DOI 10.1007/s10549-015-3322-6 Stroun M, Lyautey J, Lederrey C, Olson-Sand A, Anker P, 2001, About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin Chim Acta 313:139–142 Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R, 2001, DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 61:1659–1665 van der Vaart M, Pretorius PJ, 2008, Circulating DNA. Its origin and fluctuation. Ann N YAcad Sci 1137:18–26. , DOI 10. 1196/annals.1448.022 The Cancer Genome Atlas Network, 2012, Comprehensive molecular portraits of human breast tumours. Nature 490:61–70. https:// doi.org/10.1038/nature11412 Maruyama N, Miyoshi Y, Taguchi T, Tamaki Y, Monden M, Noguchi S, 2007, Clinicopathologic analysis of breast cancers with PIK3CA mutations in Japanese women. Clin Cancer Res 13:408– 414. , DOI 10.1158/1078-0432.ccr-06-0267 Pereira B, Chin SF, Rueda OM, Vollan HK, Provenzano E, Bardwell HA, Pugh M, Jones L, Russell R, Sammut SJ, Tsui DW, Liu B, Dawson SJ, Abraham J, Northen H, Peden JF, Mukherjee A, Turashvili G, Green AR, McKinney S, Oloumi A, Shah S, Rosenfeld N, Murphy L, Bentley DR, Ellis IO, Purushotham A, Pinder SE, Borresen-Dale AL, Earl HM, Pharoah PD, Ross MT, Aparicio S, Caldas C, 2016, The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes. Nat Commun 7:11479. , DOI 10.1038/ncomms11479 Shimazu K, Tamaki Y, Taguchi T, Takamura Y, Noguchi S, 2002, Comparison between periareolar and peritumoral injection of radiotracer for sentinel lymph node biopsy in patients with breast cancer. Surgery 131:277–286 Tamaki Y, Akiyama F, Iwase T, Kaneko T, Tsuda H, Sato K, Ueda S, Mano M, Masuda N, Takeda M, Tsujimoto M, Yoshidome K, Inaji H, Nakajima H, Komoike Y, Kataoka TR, Nakamura S, Suzuki K, Tsugawa K, Wakasa K, Okino T, Kato Y, Noguchi S, Matsuura N, 2009, Molecular detection of lymph node metastases in breast cancer patients: results of a multicenter trial using the onestep nucleic acid amplification assay. Clin Cancer Res 15:2879– 2884. , DOI 10.1158/1078-0432.ccr-08-1881 Heitzer E, Ulz P, Geigl JB, 2015, Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem 61:112–123. , DOI 10.1373/clinchem.2014.222679 Tamaki Y, Sato N, Homma K, Takabatake D, Nishimura R, Tsujimoto M, Yoshidome K, Tsuda H, Kinoshita T, Kato H, Taniyama K, Kamio T, Nakamura S, Akiyama F, Noguchi S, 2012, Routine clinical use of the one-step nucleic acid amplification assay for detection of sentinel lymph node metastases in breast cancer patients: results of a multicenter study in Japan. Cancer 118: 3477–3483. , DOI 10.1002/cncr.26683 Mouliere F, Robert B, Arnau Peyrotte E, Del Rio M, Ychou M, Molina F, Gongora C, Thierry AR, 2011, High fragmentation characterizes tumour-derived circulating DNA. PLoS One 6:e23418. , DOI 10.1371/journal.pone.0023418 Underhill HR, Kitzman JO, Hellwig S, Welker NC, Daza R, Baker DN, GligorichKM, Rostomily RC, BronnerMP, Shendure J, 2016, Fragment length of circulating tumor DNA. PLoS Genet 12: e1006162. , DOI 10.1371/journal.pgen.1006162 Lo YM, Chan KC, Sun H, Chen EZ, Jiang P, Lun FM, Zheng YW, Leung TY, Lau TK, Cantor CR, Chiu RW, 2010, Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus. Sci Transl Med 2:61ra91. , DOI 10. 1126/scitranslmed.3001720 Martin-Sanchez E, Pernaut-Leza E, Mendaza S, Cordoba A, Vicente-Garcia F, Monreal-Santesteban I, Vizcaino JP, De Cerio MJ, Perez-Janices N, Blanco-Luquin I, Escors D, Ulazia- Garmendia A, Guerrero-Setas D, 2016, Gene promoter hypermethylation is found in sentinel lymph nodes of breast cancer patients, in samples identified as positive by one-step nucleic acid amplification of cytokeratin 19mRNA.Virchows Arch 469:51–59. https:// doi.org/10.1007/s00428-016-1941-x Wang Y, Waters J, Leung ML, Unruh A, Roh W, Shi X, Chen K, Scheet P, Vattathil S, Liang H, Multani A, Zhang H, Zhao R, Michor F, Meric-Bernstam F, Navin NE, 2014, Clonal evolution in breast cancer revealed by single nucleus genome sequencing. Nature 512:155–160. , DOI 10.1038/nature13600 Jahn K, Kuipers J, Beerenwinkel N, 2016, Tree inference for single-cell data. Genome Biol 17:86. , DOI 10.1186/ s13059-016-0936-x NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1635 EP 1643 DI 10.1007/s12253-019-00618-z PG 9 ER PT J AU Liu, B Li, X Liu, F Li, F Wei, Sh Liu, J Lv, Y AF Liu, Bo Li, Xiujuan Liu, Fengxi Li, Fengyu Wei, Shuxia Liu, Junchao Lv, Yang TI Expression and Significance of TRIM 28 in Squamous Carcinoma of Esophagus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal squamous cell carcinoma; TRIM28; Immunohistochemistry; Western blot; Immunofluorescence; Overall survival ID Esophageal squamous cell carcinoma; TRIM28; Immunohistochemistry; Western blot; Immunofluorescence; Overall survival AB Tripartite motif-containing protein 28 (TRIM28) has been proved to accelerate cell proliferation and metastasis in a variety of human cancers. However, the role of TRIM28 in esophageal squamous cell carcinoma (ESCC) remains unclear. In this study, to compare the biological effect and significance of TRIM28 expression in ESCC, immunohistochemistry (streptavidin-perosidase, S-P) method was used firstly to examine the expression of TRIM28 in 136 cases of ESCC, 35 cases of high grade intraepithelial neoplasia (HGIN), 29 cases of low grade intraepithelial neoplasia (LGIN) and 37 cases of normal esophageal epithelium (NEE). Then the associations of TRIM28 expression with clinicopathological data and overall survival (OS) were also analyzed. Western blot was performed to evaluate TRIM28 protein in a total of 20 matched human ESCC and NEE tissues. Moreover, the localization of TRIM28 protein in ESCC and NEE tissues was also detected by immunofluorescence. TRIM28 protein was mainly distributed in the nucleus of ESCC. The expression of TRIM28 increased progressively from NEE to LGIN, to HGIN, and to ESCC, and it was also related to invasive depth, pTNM stage and lymph node metastasis in ESCC (P < 0.05). The results of western blot and immunofluorescence all showed that the relative expression of TRIM28 protein was markedly upregulated in ESCC compared with the NEE tissues (P < 0.01). However, prognostic analysis showed that TRIM28 may not be a prognostic factor of patients with ESCC. In conclusion, the overexpression of TRIM28 may play an important role for development and metastasis in ESCC. C1 [Liu, Bo] First Affiliated Hospital of Hebei North University, Department of Pathology, 075000 Zhangjiakou, Hebei, China. [Li, Xiujuan] Hebei North University, Department of Histology and Embryology, 11 Zuanshi South Road, 075000 Zhangjiakou, Hebei, China. [Liu, Fengxi] Hebei North University, Department of Histology and Embryology, 11 Zuanshi South Road, 075000 Zhangjiakou, Hebei, China. [Li, Fengyu] No.251 Hospital of the Chinese People’s Liberation Army, Department of Oncology, 075000 Zhangjiakou, Hebei, China. [Wei, Shuxia] No.251 Hospital of the Chinese People’s Liberation Army, Department of Oncology, 075000 Zhangjiakou, Hebei, China. [Liu, Junchao] First Affiliated Hospital of Hebei North University, Department of Pathology, 075000 Zhangjiakou, Hebei, China. [Lv, Yang] Hebei North University, Department of Histology and Embryology, 11 Zuanshi South Road, 075000 Zhangjiakou, Hebei, China. RP Lv, Y (reprint author), Hebei North University, Department of Histology and Embryology, 075000 Zhangjiakou, China. EM hbbfxy_lvyang@163.com CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics 2012. CA Cancer J Clin 65:87–108 Chen W, Zheng RS, Zhang SW, Zheng RS, Zhang SW, Zeng HM, Zou XN, Gu XY, He J, 2016). Report of Cancer incidence and mortality in China, 2012. China Cancer 25:1–8. [In Chinese, English abstract] Worni M, Martin J, Gloor B, Pietrobon R, D'Amico TA, Akushevich I, Berry MF, 2012, Does surgery improve outcomes for esophageal squamous cell carcinoma? An analysis using the surveillance epidemiology and end results registry from 1998 to 2008. J Am Coll Surg 215:643–651 Dubecz A, Gall I, Solymosi N, Schweigert M, Peters JH, Feith M, Stein HJ, 2012, Temporal trends in long-term survival and cure rates in esophageal cancer: a SEER database analysis. J Thorac Oncol 7:443–447 Luo LL, Zhao L, Xi M, He LR, Shen JX, Li QQ, Liu SL, Zhang P, Xie D, Liu MZ, 2015, Association of insulin-like growth factorbinding protein-3 with radiotherapy response and prognosis of esophageal squamous cell carcinoma. Chin J Cancer 34:514–521 Qi YJ, Wang M, Liu RM, Wei H, Chao WX, Zhang T, Lou Q, Li XM, Ma J, Zhu H, Yang ZH, Liu HQ, Ma YF, 2014, Downregulation of 14-3-3σ correlates with multistage carcinogenesis and poor prognosis of esophageal squamous cell carcinoma. PLoS One 9:e95386 K I, Inoue S, 2012, TRIM proteins as RING finger E3 ubiquitin ligases. Adv Exp Med Biol 770:27–37 Friedman JR, FredericksWJ, Jensen DE, Speicher DW, Huang XP, Neilson EG, Rauscher FJ 3rd, 1996, KAP-1, a novel corepressor for the highly conserved KRAB repression domain. Genes Dev 10: 2067–2078 Liu L, Zhang L,Wang J, Zhao X, Xu Q, Lu Y, ZuoY, Chen L, Du J, Lian Y, Zhang Q, 2018, Downregulation of TRIM28 inhibits growth and increases apoptosis of nude mice with non-small cell lung cancer xenografts. Mol Med Rep 17:835–842 Liu L, Xiao L, Liang X, Chen L, Cheng L, Zhang L,Wu X, Xu Q, Ma C, 2017, TRIM28 knockdown increases sensitivity to etoposide by upregulating E2F1 in non-small cell lung cancer. Oncol Rep 37:3597–3605 Hao L, Leng J, Xiao R, Kingsley T, Li X, Tu Z, Yang X, Deng X, Xiong M, Xiong J, Zhang Q, 2017, Bioinformatics analysis of the prognostic value of tripartite motif 28 in breast cancer. Oncol Lett 13:2670–2678 Qi ZX, Cai JJ, Chen LC, Yue Q, Gong Y, Yao Y, Mao Y, 2016, TRIM28 as an independent prognostic marker plays critical roles in glioma progression. J Neuro-Oncol 126:19–26 Lu X, Ye K, Zou K, Chen J, 2014, Identification of copy number variation-driven genes for liver cancer via bioinformatics analysis. Oncol Rep 32:1845–1852 Liu B, Yang L, Li XJ, Li R, Sun W, Chen XY, Liu JC, 2018, Expression and significance of ASPP2 in squamous carcinoma of esophagus [J]. Kaohsiung. J Med Sci 34:321–329 Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, eds,, 2010, AJCC Cancer staging manual, 7th edn. Springer- Verlag, New York Wang CN, Li ZT, Shao F, Yang X, Feng X, Shi S, Gao Y, He J, 2017, High expression of collagen triple Helix repeat containing 1, CTHRC1, facilitates progression of oesophageal squamous cell carcinoma through MAPK/MEK/ERK/FRA-1 activation. J Exp Clin Cancer Res 36:84 Gushchina LV, Kwiatkowski TA, Bhattacharya S, Weisleder NL, 2018, Conserved structural and functional aspects of the tripartite motif gene family point towards therapeutic applications inmultiple diseases. Pharmacol Ther 185:12–25 Mandell MA, Jain A, Arko-Mensah J, Chauhan S, Kimura T, Dinkins C, Silvestri G, Munch J, Kirchhoff F, Simonsen A, Wei Y, Levine B, Johansen T, Deretic V, 2014, TRIM proteins regulate autophagy and can target autophagic substrates by direct recognition. Dev Cell 30:394–409 Cambiaghi V, Giuliani V, Lombardi S, Marinelli C, Toffalorio F, Pelicci PG, 2012, TRIM proteins in cancer. Adv Exp Med Biol 770:77–91 Song Y, Guo Q, Gao S, Hua K, 2018). Tripartite motif-containing protein 3 plays a role of tumor inhibitor in cervical cancer. Biochem Biophys Res Commun 498:686–692 Liu J, Rao J, Lou X, Zhai J, Ni Z, Wang X, 2017, Upregulated TRIM11 exerts its oncogenic effects in hepatocellular carcinoma through inhibition of P53. Cell Physiol Biochem 44:255–266 Crawford LJ, Johnston CK, Irvine AE, 2018, TRIM proteins in blood cancers. J Cell Commun Signal 12:21–29 Guo P,Ma X, ZhaoW, HuaiW, Li T, Qiu Y, Zhang Y, Han L, 2018, TRIM31 is upregulated in hepatocellular carcinoma and promotes disease progression by inducing ubiquitination of TSC1-TSC2 complex. Oncogene 37:478–488 Kawabata H, Azuma K, Ikeda K, Sugitani I, Kinowaki K, Fujii T, Osaki A, Saeki T, Horie-Inoue K, Inoue S, 2017, TRIM44 is a poor prognostic factor for breast cancer patients as amodulator of NF-κB signaling. Int J Mol Sci 18:1931 Sun Y, Ji B, Feng Y, Zhang Y, Ji D, Zhu C, Wang S, Zhang C, Zhang D, Sun Y, 2017, TRIM59 facilitates the proliferation of colorectal cancer and promotes metastasis via the PI3K/AKT pathway. Oncol Rep 38:43–52 Lin WY, Wang H, Song X, Zhang SX, Zhou PS, Sun JM, Li JS, 2016, Knockdown of tripartite motif 59, TRIM59, inhibits tumor growth in prostate cancer. Eur Rev Med Pharmacol Sci 20:4864–4873 Czerwinska P, Mazurek S,Wiznerowicz M, 2017, The complexity of TRIM28 contribution to cancer. J Biomed Sci 24:63 Li F, Wang Z, Lu G, 2018, TRIM28 promotes cervical cancer growth through the mTOR signaling pathway. Oncol Rep 39: 1860–1866 Wang YY, Li L, Zhao ZS, Wang HJ, 2013, Clinical utility of measuring expression levels of KAP1, TIMP1 and STC2 in peripheral blood of patients with gastric cancer. World J Surg Oncol 11:81 Cui Y, Yang S, Fu X, Feng J, Xu S, Ying G, 2014, High levels of KAP1 expression are associated with aggressive clinical features inovarian cancer. Int J Mol Sci 16:363–377 Wang Y, Jiang J, Li Q, Ma H, Xu Z, Gao Y, 2016, KAP1 is overexpressed in hepatocellular carcinoma and its clinical significance. Int J Clin Oncol 21:927–933 Martins MB, Marcello MA, Morari EC, Cunha LL, Soares FA, Vassallo J, Ward LS, 2013, Clinical utility of KAP-1 expression in thyroid lesions. Endocr Pathol 24:77–82 Chen L, Chen DT, Chen T, Rawal B, Fulp WJ, Haura EB, Cress WD, 2012, Tripartite motif containing 28, Trim28, can regulate cell proliferation by bridging HDAC1/E2F interactions. J Biol Chem 287:40106–40118 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1645 EP 1652 DI 10.1007/s12253-018-0558-6 PG 8 ER PT J AU Wang, Y Li, X Zhang, J Liu, Q Gao, P Li, D Zhang, Sh Liu, J AF Wang, Yue Li, Xiaorui Zhang, Jiahao Liu, Qiang Gao, Peng Li, Di Zhang, Shijie Liu, Ju TI CIZ1 Expression Is Upregulated in Hemangioma of the Tongue SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CIZ1; Hemangioma; Cell proliferation; Migration ID CIZ1; Hemangioma; Cell proliferation; Migration AB Hemangioma is a vascular neoplasm and one of the most common benign tumors. The pathogenesis of hemangioma has not been fully understood. CIZ1 (Cip1-interacting zinc finger protein 1) is a nuclear protein and the binding partner of p21. Dysregulation of CIZ1 expression has been reported in various types of cancerous tissues. In this study, we examined CIZ1 expression in hemangioma of the tongue and explored its function in vascular endothelial cells, the proliferative cell type in hemangioma. Immunohistochemistry showed that CIZ1 was highly expressed in hemangioma of the tongue while its expression is minimal in the normal tongue tissues. In vitro, knockdown of CIZ1 expression by shRNA transfection significantly reduced the proliferation and migration of human umbilical vein endothelium cells (HUVECs), suggesting a positive role of CIZ1 in endothelial cell proliferation and migration. Therefore, CIZ1 might involve in pathogenesis of hemangioma of the tongue by regulation of endothelial cell functions. C1 [Wang, Yue] Shandong University, School of Dentistry, Department of OrthodonticsJinan, China. [Li, Xiaorui] Shandong University, Shandong Provincial Qianfoshan Hospital, Medical Research CenterJinan, China. [Zhang, Jiahao] Shandong University, School of Dentistry, Department of OrthodonticsJinan, China. [Liu, Qiang] Shandong University, Shandong Provincial Qianfoshan Hospital, Medical Research CenterJinan, China. [Gao, Peng] Shandong University, Shandong Provincial Qianfoshan Hospital, Medical Research CenterJinan, China. [Li, Di] Pennsylvania State University, Eberly College of Sciences, 16801 University Park, PA, USA. [Zhang, Shijie] Qilu Hospital of Shandong University, Department of StomatologyJinan, China. [Liu, Ju] Shandong University, Shandong Provincial Qianfoshan Hospital, Medical Research CenterJinan, China. RP Liu, J (reprint author), Shandong University, Shandong Provincial Qianfoshan Hospital, Medical Research Center, Jinan, China. EM ju.liu@sdu.edu.cn CR Correa PH, Nunes LC, Johann AC, Aguiar MC, Gomez RS,Mesquita RA(2007, Prevalence of oral hemangioma, vascular malformation and varix in a Brazilian population. Braz Oral Res 21(1):40–45 Pranitha V, Puppala N, Deshmukh SN, Jagadesh B, Anuradha S, 2014, Cavernous hemangioma of tongue: management of two cases. J Clin Diagn Res 8(10):ZD15–ZD17 El-Raggal NM, El-Farrash RA, Saad AA, Attia EAS, Saafan HA, Shaaban IS, 2018, Circulating levels of vascular endothelial growth factor and basic fibroblastic growth factor in infantile hemangioma versus vascular malformations. Clin Appl Thromb Hemost 24(4): 663–668 El-Hashemite N, Walker V, Kwiatkowski DJ, 2005, Estrogen enhances whereas tamoxifen retards development of Tsc mouse liver hemangioma: a tumor related to renal Angiomyolipoma and pulmonary lymphangioleiomyomatosis. Cancer Res 65(6):2474–2481 Liu Q, Niu N,WadaY, Liu J, 2016, The role of Cdkn1A-interacting zinc finger protein 1, CIZ1, in DNA replication and pathophysiology. Int J Mol Sci 17(2):212 Wang DQ, Wang K, Yan DW, Liu J, Wang B, Li MX, Wang XW, Liu J, Peng ZH, Li GX, Yu ZH, 2014, Ciz1 is a novel predictor of survival in human colon cancer. Exp Biol Med, Maywood, 239(7): 862–870 Zhou X, Liu Q,Wada Y, Liao L, Liu J, 2017, CDKN1Ainteracting zinc finger protein 1 is a novel biomarker for lung squamous cell carcinoma. Oncol Lett 5(1):183–188 ZhangD,WangY, Dai Y,Wang J,SuoT, PanH,LiuH, ShenS, Liu H, 2015, CIZ1 promoted the growth and migration of gallbladder cancer cells. Tumour Biol 36(4):2583–2591 Reggiani Bonetti L, Boselli F, Lupi M, Bettelli S, Schirosi L, Bigiani N, Sartori G, Rivasi F, 2009, Expression of estrogen receptor in hemangioma of the uterine cervix: reports of three cases and review of the literature. Arch Gynecol Obstet 280(3):469–472 den Hollander P, Rayala SK, Coverley D, Kumar R, 2006, Ciz1, a novel DNA-binding coactivator of the estrogen receptor alpha, confers hypersensitivity to estrogen action. Cancer Res 66(22):11021– 11029 Mai HM, Zheng JW, Wang YA, Yang XJ, Zhou Q, Qin ZP, Li KL, 2013, CD133 selected stem cells from proliferating infantile hemangioma and establishment of an in vivo mice model of hemangioma. Chin Med J 126(1):88–94 Yu Y, Varughese J, Brown LF, Mulliken JB, Bischoff J, 2001, Increased Tie2 expression, enhanced response to angiopoietin-1, and dysregulated angiopoietin-2 expression in hemangiomaderived endothelial cells. Am J Pathol 159(6):2271–2280 Ou JM, Yu ZY, Qiu MK, Dai YX, Dong Q, Shen J, Wang XF, Liu YB, Quan ZW, Fei ZW, 2014, Knockdown of VEGFR2 inhibits proliferation and induces apoptosis in hemangioma-derived endothelial cells. Eur J Histochem 58(1):2263 Wu JK, Adepoju O, de Silva D, Baribault K, Boscolo E, Bischoff J, Kitajewski J, 2010, A switch in notch gene expression parallels stem cell to endothelial transition in infantile hemangioma. Angiogenesis 13(1):15–23 Ji Y, Chen S, Li K, Li L, Xu C, Xiang B, 2014, Signaling pathways in the development of infantile hemangioma. J Hematol Oncol 7:13 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1653 EP 1658 DI 10.1007/s12253-018-0495-4 PG 6 ER PT J AU Villamanan, L Alcaraz, E Pinna, AL Ruzzene, M Itarte, E Arus, C Plana, M Candiota, PA AF Villamanan, Lucia Alcaraz, Estefania Pinna, A Lorenzo Ruzzene, Maria Itarte, Emilio Arus, Carles Plana, Maria Candiota, Paula Ana TI Up-Regulation of the Alpha Prime Subunit of Protein Kinase CK2 as a Marker of Fast Proliferation in GL261 Cultured Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cell cycle; GL261 glioma; Cyclin D1; Preclinical brain tumour model; CK2 alpha prime ID Cell cycle; GL261 glioma; Cyclin D1; Preclinical brain tumour model; CK2 alpha prime AB Glioblastoma (GB) is the most prevalent malignant primary brain tumor in adults. The preclinical glioblastoma model GL261 is widely used for investigating new therapeutic strategies. GL261 cultured cells are used for assessing preliminary in vitro data for this model although very little is known about the molecular characteristics of this cell line. Protein Kinase CK2 is a pleiotropic serine-threonine kinase and its inhibition may be a promising therapeutic strategy for GB treatment. In our group we follow treatment response with CK2 inhibitors in vivo using the GL261 murine model. For that, it is of our interest to assess the differential expression of α, α’, β CK2 subunits as well as CK2 activity in the GL261 GB model. CK2α’ expression changed along the growth curve of GL261 cells, undergoing downregulation in postconfluent phase cells, whereas CK2α and CK2β expression remained essentially unchanged. Furthermore, a marked decrease in CK2 activity in slowly proliferating postconfluent phase GL261 cells was observed. Finally, CK2α’ expression in orthotopic GL261 tumors was intermediate between CK2α’ expression found in cultured cells in exponentially growing or postconfluent phase, reflecting the heterogeneous nature of GL261 tumours growing in vivo. The results obtained suggest that, in the GL261 cell line, CK2α’ could play a specific role in highly proliferative cells. Also, the decrease in CK2 activity in slowly proliferating GL261 cells could imply a differential susceptibility to subunit-specific CK2 inhibitors in this cell line, although further studies are needed to confirm this hypothesis. C1 [Villamanan, Lucia] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, Cerdanyola del VallesBarcelona, Spain. [Alcaraz, Estefania] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, Cerdanyola del VallesBarcelona, Spain. [Pinna, A Lorenzo] University of Padova, and CNR Institute of Neurosciences, Department of Biomedical Sciences, via U. Bassi 58B, 35131 Padova, Italy. [Ruzzene, Maria] University of Padova, and CNR Institute of Neurosciences, Department of Biomedical Sciences, via U. Bassi 58B, 35131 Padova, Italy. [Itarte, Emilio] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, Cerdanyola del VallesBarcelona, Spain. [Arus, Carles] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, Cerdanyola del VallesBarcelona, Spain. [Plana, Maria] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, Cerdanyola del VallesBarcelona, Spain. [Candiota, Paula Ana] Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, Cerdanyola del VallesBarcelona, Spain. RP Candiota, PA (reprint author), Universitat Autonoma de Barcelona, Departament de Bioquimica i Biologia Molecular, Barcelona, Spain. EM AnaPaula.Candiota@uab.cat CR Stupp R,Mason WP, van den BentMJ,WellerM, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E,Mirimanoff RO, 2005, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352(10):987–996. , DOI 10.1056/NEJMoa043330 Di Maira G, Salvi M, Arrigoni G, Marin O, Sarno S, Brustolon F, Pinna LA, Ruzzene M, 2005, Protein kinase CK2 phosphorylates and upregulates Akt/PKB. Cell Death Differ 12(6):668–677. , DOI 10.1038/sj.cdd.4401604 Rowse AL, Gibson SA, Meares GP, Rajbhandari R, Nozell SE, Dees KJ, Hjelmeland AB, McFarland BC, Benveniste EN, 2017, Protein kinase CK2 is important for the function of glioblastoma brain tumor initiating cells. J Neuro-Oncol 132(2):219–229. https:// doi.org/10.1007/s11060-017-2378-z Stacey DW(2003, Cyclin D1 serves as a cell cycle regulatory switch in actively proliferating cells. Current opinion in cell biology 15, 2): 158-163. , DOI 10.1016/S0955-0674(03)00008-5 Ferrer-Font L, Alcaraz E, Plana M, Candiota AP, Itarte E, Arus C, 2016, Protein kinase CK2 content in GL261 mouse glioblastoma. Pathol Oncol Res 22(3):633–637. , DOI 10.1007/s12253- 015-9987-7 Ferrer-Font L, Villamanan L, Arias-Ramos N, Vilardell J, Plana M, Ruzzene M, Pinna LA, Itarte E, Arus C, Candiota AP, 2017, Targeting protein kinase CK2: evaluating CX-4945 potential for GL261 glioblastoma therapy in immunocompetent mice. Pharmaceuticals 10(1). , DOI 10.3390/ph10010024 OrlandiniM, Semplici F, Ferruzzi R, Meggio F, Pinna LA, Oliviero S, 1998, Protein kinaseCK2alpha' is induced by serumas a delayed early gene and cooperates with Ha-ras in fibroblast transformation. J Biol Chem 273, 33):21291–21297. , DOI 10.1074/jbc. 273.33.21291 Ortega-Martorell S, Lisboa PJG, Vellido A, Simoes RV, Pumarola M, Julia-Sape M, Arus C, 2012, Convex Non-Negative Matrix Factorization for Brain Tumor Delimitation from MRSI Data. PLoS One 7, 10):e47824. , DOI 10.1371/journal.pone.0047824 Janeczko M, Orzeszko A, Kazimierczuk Z, Szyszka R, Baier A, 2012, CK2alpha and CK2alpha' subunits differ in their sensitivity to 4,5,6,7-tetrabromo- and 4,5,6,7-tetraiodo-1H-benzimidazole derivatives. Eur J Med Chem 47(1):345–350. , DOI 10.1016/ j.ejmech.2011.11.002 Bollacke A, Nienberg C, Borgne ML, Jose J, 2016, Toward selective CK2alpha and CK2alpha' inhibitors: development of a novel wholecell kinase assay by autodisplay of catalytic CK2alpha'. J Pharm Biomed Anal 121:253–260. , DOI 10.1016/j.jpba.2016.01.011 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1659 EP 1663 DI 10.1007/s12253-018-00567-z PG 5 ER PT J AU Mormile, R AF Mormile, Raffaella TI EGFR-TK1-Associated Interstitial Pneumonitis in Nivolumab-Treated Patients with NSCLC: When Is it Worth The Risk? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Mormile, Raffaella] Moscati Hospital, Division of Pediatrics and Neonatology, Via A. Gramsci, 81031 Aversa, Italy. RP Mormile, R (reprint author), Moscati Hospital, Division of Pediatrics and Neonatology, 81031 Aversa, Italy. EM raffaellamormile@alice.it CR OshimaY, Tanimoto T, Yuji K, Tojo A, 2018, EGFR-TKI-associated interstitial pneumonitis inNivolumab-treated patients with non-small cell lung Cancer. JAMA Oncol 4(8):1112–1115 Santabarbara G, Maione P, Rossi A, Palazzolo G, Gridelli C, 2016, Novel immunotherapy in the treatment of advanced non-small cell lung cancer. Expert Rev Clin Pharmacol 9(12):1571–1581 Ahn MJ, Sun JM, Lee SH, Ahn JS, Park K, 2017, EGFR TKI combination with immunotherapy in non-small cell lung cancer. Expert Opin Drug Saf 16(4):465–469 Gettinger S, Hellmann MD, Chow LQM, Borghaei H, Antonia S, Brahmer JR, Goldman JW, Gerber DE et al, 2018, Nivolumab plus erlotinib in patients with EGFR-mutant advanced NSCLC. J Thorac Oncol 13(9):1363–1372 Haratani K, Hayashi H, Tanaka T, Kaneda H, Togashi Y, Sakai K, Hayashi K et al, 2017, Tumor immune microenvironment and nivolumab efficacy in EGFR mutation-positive non-small-cell lung cancer based on T790M status after disease progression during EGFR-TKI treatment. Ann Oncol 28(7):1532–1539 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1665 EP 1666 DI 10.1007/s12253-019-00599-z PG 2 ER PT J AU Li, Xl Dai, Shd AF Li, Xiao-li Dai, Shun-dong TI Melanotic Schwannoma: Two Cases of Rare Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Li, Xiao-li] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of PathologyShenyang, Liaoning, China. [Dai, Shun-dong] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of PathologyShenyang, Liaoning, China. RP Dai, Shd (reprint author), China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, Shenyang, China. EM sddai@cmu.edu.cn CR Louis DN, 2007, WHO classification of tumours of the central nervous system[M]. In: International Agency for Research on Cancer Mahato D, Vivasbuitrago T, Gassie K et al, 2017, Intracranial melanotic schwannomas: a rare variant with unusual adherent features[J]. J Neuro-Oncol:136(1) Cheng X, Liu J, Le J et al, 2017, Invasive intramedullary melanotic schwannoma: case report and review of the literature[J]. Eur Spine J: 1–6 Torresmora J, Dry S, Li X et al, 2014, Malignant melanotic schwannian tumor: a clinicopathologic, immunohistochemical, and gene expression profiling study of 40 cases, with a proposal for the reclassification of "melanotic schwannoma".[J]. Am J Surg Pathol 38(1):94–105 Lee JK, Rho YJ, Dong MJ et al, 2017, Diagnostic clue of meningeal melanocytoma: case report and review of literature[J]. Yonsei Med J 58(2):467–470 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1667 EP 1670 DI 10.1007/s12253-018-0417-5 PG 4 ER PT J AU Altundag, K AF Altundag, Kadri TI Significant Family History of Breast Cancer may Further Increase Risk of Pre-malignant and Malignant Lesions in Specimens from Breast Reduction Surgery SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Altundag, Kadri] MKA Breast Cancer Clinic, Tepe Prime, Cankaya, 06800 Ankara, Turkey. RP Altundag, K (reprint author), MKA Breast Cancer Clinic, 06800 Ankara, Turkey. EM altundag66@yahoo.com CR Uson Junior PLS, Callegaro Filho D, Bugano DDG, Geyer FC, de Nigro Corpa MV, Goncalves PDS, Simon SD, Kaliks RA, 2018, Incidental Findings in Reduction Mammoplasty Specimens in Patients with No Prior History of Breast Cancer. An Analysis of 783 Specimens. Pathol Oncol Res 24(1):95–99 Metcalfe KA, Finch A, Poll A, Horsman D, Kim-Sing C, Scott J, Royer R, Sun P, Narod SA, 2009, Breast cancer risks in women with a family history of breast or ovarian cancer who have tested negative for a BRCA1 or BRCA2 mutation. Br J Cancer 100(2):421–425 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1671 EP 1671 DI 10.1007/s12253-018-0437-1 PG 1 ER PT J AU Raj, Th Patil, Sh Sarode, CS Sarode, SG AF Raj, A. Thirumal Patil, Shankargouda Sarode, C Sachin Sarode, S Gargi TI Oral Potentially Malignant Disorder Based Customized Epithelial Dysplasia Grading: a Need of the Hour SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Raj, A. Thirumal] Sri Venkateswara Dental College and Hospital, Department of Oral Pathology and Microbiology, Thalambur, 600130 Chennai, India. [Patil, Shankargouda] Jazan University, College of Dentistry, Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral PathologyJazan, Saudi Arabia. [Sarode, C Sachin] Dr. D. Y. Patil Dental College and Hospital, Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Vidyapeeth, PimpriPune, India. [Sarode, S Gargi] Dr. D. Y. Patil Dental College and Hospital, Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Vidyapeeth, PimpriPune, India. RP Raj, Th (reprint author), Sri Venkateswara Dental College and Hospital, Department of Oral Pathology and Microbiology, 600130 Chennai, India. EM thirumalraj666@gmail.com CR Gale N, Pilch BZ, Sidransky D, 2005, Epithelial precursor lesions. In: Barnes L, Eveson J, Reichart P, Sidransky D, eds, World Health Organization classification of tumors: pathology and genetics of tumors of the head and neck. IARC Press, Lyon Reibel J, Gale N, Hille J et al, 2017, Oral potentially malignant disorders and oral epithelial dysplasias. In: El-Naggar AK, Chan JKC, Gradis JR, Takata T, Slootweg PJ, eds, WHO classification of tumours of the head and neck. Tumors of the oral cavity and mobile tongue, 4th edn. IARC Press, Lyon Warnakulasuriya S, Tilakaratne WM, Ranganathan K, Kuriakose MA, 2018, Report of a consensus meeting of a group of oral and general pathologists in India on the grading of oral epithelial dysplasia. Oral Oncol. , DOI 10.1016/j.oraloncology. 2018.03.018 Muller S, 2017, Update from the 4th edition of the world health organization of head and neck tumors: tumors of the oral cavity and mobile tongue. Head Neck Pathol 11:33–40. , DOI 10. 1007/s12105-017-0794-1 Sarode SC, Chaudhary M, Gadbail A, Tekade S, Patil S, Sarode GS, 2018, Dysplastic features relevant to malignant transformation in the atrophic epithelium of oral submucous fibrosis: a preliminary study. J Oral Pathol Med. , DOI 10.1111/jop.12699 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1673 EP 1674 DI 10.1007/s12253-018-0455-z PG 2 ER PT J AU Son, JH Kim, SM Yoo, JN Lee, HS AF Son, Ji Hyun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Absence of Promoter Mutation in TBC1D12 Gene in Solid and Hematologic Neoplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Son, Ji Hyun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Oguchi ME, Noguchi K, Fukuda M, 2017, TBC1D12 is a novel Rab11-binding protein that modulates neurite outgrowth of PC12 cells. PLoS One 12:e0174883 Rheinbay E, Parasuraman P, Grimsby J, Tiao G, Engreitz JM, Kim J, Lawrence MS, Taylor-Weiner A, Rodriguez-Cuevas S, Rosenberg M, Hess J, Stewart C, Maruvka YE, Stojanov P, Cortes ML, Seepo S, Cibulskis C, Tracy A, Pugh TJ, Lee J, Zheng Z, Ellisen LW, Iafrate AJ, Boehm JS, Gabriel SB, Meyerson M, Golub TR, Baselga J, Hidalgo-Miranda A, Shioda T, Bernards A, Lander ES, Getz G, 2017, Recurrent and functional regulatory mutations in breast cancer. Nature 547:55–60 Heng J, Guo X,WuW,Wang Y, Li G, Chen M, Peng L,Wang S, Dai L, Tang L, Wang J, 2017, Integrated analysis of promoter mutation, methylation and expression of AKT1 gene in Chinese breast cancer patients. PLoS One 12:e0174022 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2019 VL 25 IS 4 BP 1675 EP 1676 DI 10.1007/s12253-018-0468-7 PG 2 ER PT J AU Kopper, L Timar, J AF Kopper, Laszlo Timar, Jozsef TI 25th Anniversary of Pathology Oncology Research SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Editorial C1 [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Timar, Jozsef] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Kopper, L (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM kopper@korb1.sote.hu CR Kopper L, 1995, Prologue. Pathol Oncol Res 1:6 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 1 EP 1 DI 10.1007/s12253-020-00795-2 PG 1 ER PT J AU Lendvai, G Szekerczes, T Illyes, I Dora, R Kontsek, E Gogl, A Kiss, A Werling, K Kovalszky, I Schaff, Zs Borka, K AF Lendvai, Gabor Szekerczes, Timea Illyes, Ildiko Dora, Reka Kontsek, Endre Gogl, Aliz Kiss, Andras Werling, Klara Kovalszky, Ilona Schaff, Zsuzsa Borka, Katalin TI Cholangiocarcinoma: Classification, Histopathology and Molecular Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Cholangiocarcinoma; Liver cancer; Biliary markers; Stem cells; MicroRNA ID Cholangiocarcinoma; Liver cancer; Biliary markers; Stem cells; MicroRNA AB Cholangiocarcinoma (CC) is the second most common tumor of the liver, originating from the biliary system with increasing incidence and mortality worldwide. Several new classifications review the significance of tumor localization, site of origin, proliferation and biomarkers in the intrahepatic, perihilar and distal forms of the lesion. Based on growth pattern mass-forming, periductal-infiltrating, intraductal, undefined and mixed types are differentiated. There are further subclassifications which are applied for the histological features, in particular for intrahepatic CC. Recognition of the precursors and early lesions of CC including biliary intraepithelial neoplasia (BilIN), intraductal papillary neoplasm of the bile ducts (IPNB), biliary mucinous cystic neoplasm (MCNB) and the candidate precursors, such as bile duct adenoma and von Meyenburg complex is of increasing significance. In addition to the previously used biliary markers detected by immunohistochemistry, several new markers have been added to the differentiation of both the benign and malignant lesions, which can be used to aid in the subclassification in association with the outcome of CC. Major aspects of biliary carcinogenesis have been revealed, yet, the exact way of this diverse process is still unclear. The factors contributing to molecular cholangiocarcinogenesis include various risk factors, different anatomical localizations, multiple cellular origins, genetic and epigenetic alterations, tumor microenvironment, heterogeneity and clonal evolution. Driver mutations have been identified, implying that they are optimal candidates for targeted therapy. The most promising therapeutic candidates have entered clinical trials. C1 [Lendvai, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Szekerczes, Timea] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Illyes, Ildiko] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Dora, Reka] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kontsek, Endre] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Gogl, Aliz] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Werling, Klara] Semmelweis University, 2nd Department of Internal Medicine, H-1085 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Borka, Katalin] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM schaff.zsuzsa@med.semmelweis-univ.hu CR Bridgewater J, Galle PR, Khan SA, Llovet JM, Park JW, Patel T, Pawlik TM, Gores GJ, 2014, Guidelines for the diagnosis and management of intrahepatic cholangiocarcinoma. J Hepatol 60(6):1268–1289 Dodson RM, Weiss MJ, Cosgrove D, Herman JM, Kamel I, Anders R, Geschwind JF, Pawlik TM, 2013, Intrahepatic cholangiocarcinoma: management options and emerging therapies. J Am Coll Surg 217(4):736–750 e734 Rizvi S, Gores GJ, 2013, Pathogenesis, diagnosis, and management of cholangiocarcinoma. Gastroenterology 145(6):1215– 1229 Javle M, Bekaii-Saab T, Jain A, Wang Y, Kelley RK, Wang K, Kang HC, Catenacci D, Ali S, Krishnan S, Ahn D, Bocobo AG, Zuo M, Kaseb A, Miller V, Stephens PJ, Meric-BernstamF, Shroff R, Ross J, 2016, Biliary cancer: utility of next-generation sequencing for clinical management. Cancer 122(24):3838–3847 Nakanuma Y, Sato Y, Harada K, Sasaki M, Xu J, Ikeda H, 2010, Pathological classification of intrahepatic cholangiocarcinoma based on a new concept. World J Hepatol 2(12):419–427 Nakanuma Y, Tsutsui A, Ren XS, Harada K, Sato Y, Sasaki M, 2014, What are the precursor and early lesions of peripheral intrahepatic cholangiocarcinoma? Int J Hepatol 2014:805973 Maemura K, Natsugoe S, Takao S, 2014, Molecular mechanism of cholangiocarcinoma carcinogenesis. J Hepatobiliary Pancreat Sci 21(10):754–760 Cai Y, Cheng N, Ye H, Li F, Song P, Tang W, 2016, The current management of cholangiocarcinoma: a comparison of current guidelines. Biosci Trends 10(2):92–102 Marcano-Bonilla L, Mohamed EA, Mounajjed T, Roberts LR, 2016, Biliary tract cancers: epidemiology, molecular pathogenesis and genetic risk associations. Chin Clin Oncol 5(5):61 Razumilava N, Gores GJ, 2014, Cholangiocarcinoma. Lancet 383(9935):2168–2179 Nakanuma Y, Harada K, Sasaki M, Sato Y, 2014, Proposal of a new disease concept "biliary diseases with pancreatic counterparts". Anatomical and pathological bases. Histol Histopathol 29(1):1–10 Simbolo M, Fassan M, Mafficini A, Lawlor RT, Ruzzenente A, Scarpa A, 2016, New genomic landscapes and therapeutic targets for biliary tract cancers. Front Biosci, Landmark Ed, 21:707–718 Serafini FM, Radvinsky D, 2016, The pathways of genetic transformation in cholangiocarcinogenesis. Cancer Genet 209(12): 554–558 Jain A, Kwong LN, Javle M, 2016, Genomic profiling of biliary tract cancers and implications for clinical practice. Curr Treat Options in Oncol 17(11):58 Edge SB, Byrd DR, Compton CC, 2010, AJCC Cancer staging manual, 7th edn. Springer, New York Sobin LH, Gospodarowicz MK, Wittekind C, 2009, TNM classification of malignant tumors, 7th edn. Wiley-Blackwell, Oxford Gandou C, Harada K, Sato Y, Igarashi S, Sasaki M, Ikeda H, Nakanuma Y, 2013, Hilar cholangiocarcinoma and pancreatic ductal adenocarcinoma share similar histopathologies, immunophenotypes, and development-related molecules. Hum Pathol 44(5):811–821 Nemeth Z, Szasz AM, Somoracz A, Tatrai P, Nemeth J, Gyorffy H, Szijarto A, Kupcsulik P, Kiss A, Schaff Z, 2009, Zonula occludens- 1, occludin, and E-cadherin protein expression in biliary tract cancers. Pathol Oncol Res 15(3):533–539 Nemeth Z, Szasz AM, Tatrai P, Nemeth J, Gyorffy H, Somoracz A, Szijarto A,Kupcsulik P, Kiss A, Schaff Z, 2009, Claudin-1, −2, −3, −4, −7, −8, and −10 protein expression in biliary tract cancers. J Histochem Cytochem 57(2):113–121 Zhou Y, Zhao Y, Li B, Huang J,Wu L, Xu D, Yang J, He J, 2012, Hepatitis viruses infection and risk of intrahepatic cholangiocarcinoma: evidence from a meta-analysis. BMC Cancer 12:289 Hu J, Yin B, 2016, Advances in biomarkers of biliary tract cancers. Biomed Pharmacother 81:128–135 Rizvi S, Khan SA, Hallemeier CL, Kelley RK, Gores GJ, 2018, Cholangiocarcinoma - evolving concepts and therapeutic strategies. Nat Rev Clin Oncol 15(2):95–111 Dong LQ, Shi Y, Ma LJ, Yang LX,Wang XY, Zhang S,Wang ZC, Duan M, Zhang Z, Liu LZ, Zheng BH, Ding ZB, Ke AW, Gao DM, Yuan K, Zhou J, Fan J, Xi R, Gao Q, 2018, Spatial and temporal clonal evolution of intrahepatic cholangiocarcinoma. J Hepatol 69(1):89–98 Palmer WC, Patel T, 2012, Are common factors involved in the pathogenesis of primary liver cancers? A meta-analysis of risk factors for intrahepatic cholangiocarcinoma. J Hepatol 57(1):69– 76 Wei M, Lu L, Lin P, Chen Z, Quan Z, Tang Z, 2016, Multiple cellular origins andmolecular evolution of intrahepatic cholangiocarcinoma. Cancer Lett 379(2):253–261 Raggi C, Invernizzi P, Andersen JB, 2015, Impact of microenvironment and stem-like plasticity in cholangiocarcinoma: molecular networks and biological concepts. J Hepatol 62(1):198–207 Moeini A, Sia D, Zhang Z, Camprecios G, Stueck A, Dong H, Montal R, Torrens L, Martinez-Quetglas I, Fiel MI, Hao K, Villanueva A, Thung SN, Schwartz ME, Llovet JM, 2017, Mixed hepatocellular cholangiocarcinoma tumors: Cholangiolocellular carcinoma is a distinct molecular entity. J Hepatol 66(5):952–961 Mertens JC, Rizvi S, Gores GJ, 2018, Targeting cholangiocarcinoma. Biochim Biophys Acta 1864(4 Pt B):1454–1460 Aishima S, Oda Y, 2015, Pathogenesis and classification of intrahepatic cholangiocarcinoma: different characters of perihilar large duct type versus peripheral small duct type. J Hepatobiliary Pancreat Sci 22(2):94–100 Carpino G, Cardinale V, Onori P, Franchitto A, Berloco PB, Rossi M,Wang Y, Semeraro R, Anceschi M, Brunelli R, Alvaro D, Reid LM, Gaudio E, 2012, Biliary tree stem/progenitor cells in glands of extrahepatic and intraheptic bile ducts: an anatomical in situ study yielding evidence of maturational lineages. J Anat 220(2): 186–199 Nakanuma Y, Crurado MP, Franceschi S, Gores GJ, Paradis V, Sripa B, Tsui WMS, Wee A, 2010, Intrahepatic cholangiocarcinoma. In: Bosman FT, Carneiro F, Hruban RH, Theise ND, eds, WHO classification of tumors of the digestive system, 4th edn. IARC, Lyon, pp 217–224 Fernandez Moro C, Fernandez-Woodbridge A, Alistair D'souza M, Zhang Q, Bozoky B, Kandaswamy SV, Catalano P, Heuchel R, Shtembari S, Del Chiaro M, Danielsson O, Bjornstedt M, Lohr JM, I s a k s son B, Ve r b e k e C, Bozoky B, 2016, Immunohistochemical typing of adenocarcinomas of the Pancreatobiliary system improves diagnosis and prognostic stratification. PLoS One 11(11):e0166067 Lagana S, Hsiao S, Bao F, Sepulveda A, Moreira R, Lefkowitch J, Remotti H, 2015, HepPar-1 and Arginase-1 immunohistochemistry in adenocarcinoma of the small intestine and Ampullary region. Arch Pathol Lab Med 139(6):791–795 Lodi C, Szabo E, Holczbauer A, Batmunkh E, Szijarto A, Kupcsulik P, Kovalszky I, Paku S, Illyes G, Kiss A, Schaff Z, 2006, Claudin-4 differentiates biliary tract cancers from hepatocellular carcinomas. Mod Pathol 19(3):460–469 Borka K, Kaliszky P, Szabo E, Lotz G,Kupcsulik P, Schaff Z, Kiss A, 2007, Claudin expression in pancreatic endocrine tumors as compared with ductal adenocarcinomas. Virchows Arch 450(5): 549–557 Kloppel G, Adsay V, Konukiewitz B, Kleeff J, Schlitter AM, Esposito I, 2013, Precancerous lesions of the biliary tree. Best Pract Res Clin Gastroenterol 27(2):285–297 Sato Y, Sasaki M, Harada K, Aishima S, Fukusato T, Ojima H, Kanai Y, Kage M, Nakanuma Y, Tsubouchi H, Hepatolithiasis Subdivision of Intractable Hepatobiliary Diseases Study Group of J, 2014, Pathological diagnosis of flat epithelial lesions of the biliary tract with emphasis on biliary intraepithelial neoplasia. J Gastroenterol 49(1):64–72 Sato Y, Harada K, Sasaki M, Nakanuma Y, 2013, Histological characteristics of biliary intraepithelial neoplasia-3 and intraepithelial spread of cholangiocarcinoma. Virchows Arch 462(4):421–427 Bosman FT, Carneiro F, Hruban RH, Theise ND, 2010, WHO classification of tumors of the digestive system, 4th edn. IARC, Lyon Ohtsuka M, Shimizu H, Kato A, Yoshitomi H, Furukawa K, Tsuyuguchi T, Sakai Y, Yokosuka O, Miyazaki M, 2014, Intraductal papillary neoplasms of the bile duct. Int J Hepatol 2014(459091):1–10 NakanumaY, UesakaK, Miyayama S, Yamaguchi H, OhtsukaM, 2017, Intraductal neoplasms of the bile duct. A new challenge to biliary tract tumor pathology. Histol Histopathol 32(10):1001– 1015 Zen Y, Adsay NV, Bardadin K, Colombari R, Ferrell L, Haga H, Hong SM, Hytiroglou P, Kloppel G, Lauwers GY, van Leeuwen DJ, Notohara K, Oshima K, Quaglia A, Sasaki M, Sessa F, Suriawinata A, Tsui W, Atomi Y, Nakanuma Y, 2007, Biliary intraepithelial neoplasia: an international interobserver agreement study and proposal for diagnostic criteria. Mod Pathol 20(6):701– 709 Hajosi-Kalcakosz S, Dezso K, Bugyik E, Bodor C, Paku S, Pavai Z, Halasz J, Schlachter K, Schaff Z, Nagy P, 2012, Enhancer of zeste homologue 2, EZH2, is a reliable immunohistochemical marker to differentiate malignant and benign hepatic tumors. Diagn Pathol 7:86 Sasaki M, Matsubara T, Kakuda Y, Sato Y, Nakanuma Y, 2014, Immunostaining for polycomb group protein EZH2 and senescent marker p16INK4a may be useful to di fferent i a t e cholangiolocellular carcinoma fromductular reaction and bile duct adenoma. Am J Surg Pathol 38(3):364–369 Fukumura Y, Nakanuma Y, Kakuda Y, Takase M, Yao T, 2017, Clinicopathological features of intraductal papillary neoplasms of the bile duct: a comparison with intraductal papillary mucinous neoplasm of the pancreas with reference to subtypes. Virchows Arch 471(1):65–76 Wan XS, Xu YY, Qian JY, Yang XB, Wang AQ, He L, Zhao HT, Sang XT, 2013, Intraductal papillary neoplasm of the bile duct. World J Gastroenterol 19(46):8595–8604 Nakanuma Y, Sato Y, Ojima H, Kanai Y, Aishima S, Yamamoto M, Ariizumi S, Furukawa T, Hayashi H, Unno M, Ohta T, Hepatolithiasis Subdivision of Intractable Hepatobiliary Diseases Study Group of J, 2014, Clinicopathological characterization of so-called "cholangiocarcinoma with intraductal papillary growth" with respect to "intraductal papillary neoplasm of bile duct, IPNB)". Int J Clin Exp Pathol 7(6):3112–3122 Zen Y, Fujii T, Itatsu K, Nakamura K, Minato H, Kasashima S, Kurumaya H, Katayanagi K, Kawashima A, Masuda S, Niwa H, Mitsui T, Asada Y, Miura S, Ohta T, Nakanuma Y, 2006, Biliary papillary tumors share pathological features with intraductal papillary mucinous neoplasm of the pancreas. Hepatology 44(5): 1333–1343 Nakanuma Y, Kakuda Y, Uesaka K, Miyata T, Yamamoto Y, Fukumura Y, Sato Y, Sasaki M, Harada K, Takase M, 2016, Characterization of intraductal papillary neoplasm of bile duct with respect to histopathologic similarities to pancreatic intraductal papillary mucinous neoplasm. Hum Pathol 51:103–113 Fujikura K, Fukumoto T, Ajiki T, Otani K, KanzawaM, Akita M, Kido M, Ku Y, Itoh T, Zen Y, 2016, Comparative clinicopathological study of biliary intraductal papillary neoplasms and papillary cholangiocarcinomas. Histopathology 69(6):950–961 Aishima S, Tanaka Y, Kubo Y, Shirabe K, Maehara Y, Oda Y, 2014, Bile duct adenoma and von Meyenburg complex-like duct arising in hepatitis and cirrhosis: pathogenesis and histological characteristics. Pathol Int 64(11):551–559 Zimmermann A, 2017, Tumors and tumor-like lesions of the hepatobiliary tract. General and surgical pathology. Vol. 1. Springer, Switzerland Bertram S, Padden J, Kalsch J, Ahrens M, Pott L, Canbay A, Weber F, Fingas C, Hoffmann AC, Vietor A, Schlaak JF, Eisenacher M, Reis H, Sitek B, Baba HA, 2016, Novel immunohistochemicalmarkers differentiate intrahepatic cholangiocarcinoma from benign bile duct lesions. J Clin Pathol 69(7):619–626 Tsokos CG, Krings G, Yilmaz F, Ferrell LD, Gill RM, 2016, Proliferative index facilitates distinction between benign biliary lesions and intrahepatic cholangiocarcinoma. Hum Pathol 57: 61–67 Song JS, Lee YJ, Kim KW, Huh J, Jang SJ, Yu E, 2008, Cholangiocarcinoma arising in von Meyenburg complexes: report of four cases. Pathol Int 58(8):503–512 Orii T, Ohkohchi N, Sasaki K, Satomi S, Watanabe M, Moriya T, 2003, Cholangiocarcinoma arising from preexisting biliary hamartoma of liver–report of a case. Hepatogastroenterology 50(50):333–336 O'Dell MR, Huang JL, Whitney-Miller CL, Deshpande V, Rothberg P, Grose V, Rossi RM, Zhu AX, Land H, Bardeesy N, Hezel AF, 2012, Kras(G12D, and p53 mutation cause primary intrahepatic cholangiocarcinoma. Cancer Res 72(6):1557–1567 Sato Y, Harada K, Sasaki M, Nakanuma Y, 2014, Cystic and micropapillary epithelial changes of peribiliary glands might represent a precursor lesion of biliary epithelial neoplasms. Virchows Arch 464(2):157–163 Cardinale V,Wang Y, Carpino G, Mendel G, Alpini G, Gaudio E, Reid LM, Alvaro D, 2012, The biliary tree–a reservoir of multipotent stem cells. Nat Rev Gastroenterol Hepatol 9(4):231– 240 Sutton ME, op den Dries S, Koster MH, Lisman T, Gouw AS, Porte RJ, 2012, Regeneration of human extrahepatic biliary epithelium: the peribiliary glands as progenitor cell compartment. Liver Int 32(4):554–559 Gibiino G, Fabbri C, Fagiuoli S, Ianiro G, Fornelli A, Cennamo V, 2017, Defining the biology of intrahepatic cholangiocarcinoma: molecular pathways and early detection of precursor lesions. Eur Rev Med Pharmacol Sci 21(4):730–741 Walter D, Hartmann S, Waidmann O, 2017, Update on cholangiocarcinoma: potential impact of genomic studies on clinical management. Z Gastroenterol 55(6):575–581 Valle JW, Lamarca A, Goyal L, Barriuso J, Zhu AX, 2017, New horizons for precision medicine in biliary tract cancers. Cancer Discov 7(9):943–962 Wise C, Pilanthananond M, Perry BF, Alpini G, McNeal M, Glaser SS, 2008, Mechanisms of biliary carcinogenesis and growth. World J Gastroenterol 14(19):2986–2989 Xie D, Ren Z, Fan J, Gao Q, 2016, Genetic profiling of intrahepatic cholangiocarcinoma and its clinical implication in targeted therapy. Am J Cancer Res 6(3):577–586 Tshering G, Dorji PW, ChaijaroenkulW, Na-Bangchang K, 2018, Biomarkers for the diagnosis of cholangiocarcinoma: a systematic review. Am J Trop Med Hyg 98(6):1788–1797 Nakanuma Y, Sasaki M, Sato Y, Ren X, Ikeda H, Harada K, 2009, Multistep carcinogenesis of perihilar cholangiocarcinoma arising in the intrahepatic large bile ducts. World J Hepatol 1(1):35–42 Marks EI, Yee NS, 2016, Molecular genetics and targeted therapeutics in biliary tract carcinoma. World J Gastroenterol 22(4): 1335–1347 Sasaki M, Matsubara T, Nitta T, Sato Y, Nakanuma Y, 2013, GNAS and KRAS mutations are common in intraductal papillary neoplasms of the bile duct. PLoS One 8(12):e81706 HsuM, Sasaki M, Igarashi S, Sato Y, Nakanuma Y, 2013, KRAS and GNAS mutations and p53 overexpression in biliary intraepithelial neoplasia and intrahepatic cholangiocarcinomas. Cancer 119(9):1669–1674 Churi CR, Shroff R, Wang Y, Rashid A, Kang HC, Weatherly J, Zuo M, Zinner R, Hong D, Meric-BernstamF, Janku F, Crane CH, Mishra L, Vauthey JN, Wolff RA, Mills G, Javle M, 2014, Mutation profiling in cholangiocarcinoma: prognostic and therapeutic implications. PLoS One 9(12):e115383 Nakaoka T, Saito Y, Saito H, 2017, Aberrant DNA methylation as a biomarker and a therapeutic target of cholangiocarcinoma. Int J Mol Sci 18(6):1111 Udali S, Guarini P, Moruzzi S, Ruzzenente A, Tammen SA, Guglielmi A, Conci S, Pattini P, Olivieri O, Corrocher R, Choi SW, Friso S, 2015, Global DNA methylation and hydroxymethylation differ in hepatocellular carcinoma and cholangiocarcinoma and relate to survival rate. Hepatology 62(2): 496–504 Chiang NJ, Shan YS, Hung WC, Chen LT, 2015, Epigenetic regulation in the carcinogenesis of cholangiocarcinoma. Int J Biochem Cell Biol 67:110–114 Lee H, Wang K, Johnson A, Jones DM, Ali SM, Elvin JA, Yelensky R, Lipson D, Miller VA, Stephens PJ, Javle M, Ross JS, 2016, Comprehensive genomic profiling of extrahepatic cholangiocarcinoma reveals a long tail of therapeutic targets. J Clin Pathol 69(5):403–408 Yoo KH, Kim NK, Kwon WI, Lee C, Kim SY, Jang J, Ahn J, Kang M, Jang H, Kim ST, Ahn S, Jang KT, Park YS, Park WY, Lee J, Heo JS, Park JO, 2016, Genomic alterations in biliary tract Cancer using targeted sequencing. Transl Oncol 9(3):173–178 Simbolo M, Fassan M, Ruzzenente A, Mafficini A, Wood LD, Corbo V, Melisi D, Malleo G, Vicentini C, Malpeli G, Antonello D, Sperandio N, Capelli P, Tomezzoli A, Iacono C, Lawlor RT, Bassi C, Hruban RH, Guglielmi A, Tortora G, de Braud F, Scarpa A, 2014, Multigene mutational profiling of cholangiocarcinomas identifies actionable molecular subgroups. Oncotarget 5(9):2839– 2852 Ross JS, Wang K, Gay L, Al-Rohil R, Rand JV, Jones DM, Lee HJ, Sheehan CE, Otto GA, Palmer G, Yelensky R, Lipson D, Morosini D, Hawryluk M, Catenacci DV, Miller VA, Churi C, Ali S, Stephens PJ, 2014, New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing. Oncologist 19(3):235–242 Ruzzenente A, Fassan M, Conci S, Simbolo M, Lawlor RT, Pedrazzani C, Capelli P, D'Onofrio M, Iacono C, Scarpa A, Guglielmi A, 2016, Cholangiocarcinoma heterogeneity revealed by multigene mutational profiling: clinical and prognostic relevance in surgically resected patients. Ann Surg Oncol 23(5): 1699–1707 Farshidfar F, Zheng S, Gingras MC, Newton Y, Shih J, 2017, Integrative genomic analysis of cholangiocarcinoma identifies distinct IDH-mutant molecular profiles. Cell Rep 18(11):2780–2794 Jiao Y, Pawlik TM, Anders RA, Selaru FM, Streppel MM, 2013, Exome sequencing identifies frequent inactivating mutations in BAP 1 , ARID1A a n d PBRM1 i n i n t r a h e p a t i c cholangiocarcinomas. Nat Genet 45(12):1470–1473 Putra J, de Abreu FB, Peterson JD, Pipas JM, Mody K, 2015, Molecular profiling of intrahepatic and extrahepatic cholangiocarcinoma using next generation sequencing. Exp Mol Pathol 99(2): 240–244 Zou S, Li J, Zhou H, Frech C, Jiang X, 2014, Mutational landscape of intrahepatic cholangiocarcinoma. Nat Commun 5:5696 Nakamura H, Arai Y, Totoki Y, Shirota T, Elzawahry A, Kato M, Hama N, Hosoda F, Urushidate T, Ohashi S, Hiraoka N, Ojima H, Shimada K, Okusaka T, Kosuge T, Miyagawa S, Shibata T, 2015, Genomic spectra of biliary tract cancer. Nat Genet 47(9):1003– 1010 Sia D, Losic B, Moeini A, Cabellos L, Hao K, 2015, Massive parallel sequencing uncovers actionable FGFR2-PPHLN1 fusion and ARAF mutations in intrahepatic cholangiocarcinoma. Nat Commun 6:6087 Fujimoto A, FurutaM, Shiraishi Y, Gotoh K, Kawakami Y, 2015, Whole-genome mutational landscape of liver cancers displaying biliary phenotype reveals hepatitis impact andmolecular diversity. Nat Commun 6:6120 Rizvi S, Gores GJ, 2017, Emerging molecular therapeutic targets for cholangiocarcinoma. J Hepatol 67(3):632–644 Lee H, Ross JS, 2017, The potential role of comprehensive genomic profiling to guide targeted therapy for patients with biliary cancer. Ther Adv Gastroenterol 10(6):507–520 Moeini A, Sia D, Bardeesy N, Mazzaferro V, Llovet JM, 2016, Molecular pathogenesis and targeted therapies for intrahepatic cholangiocarcinoma. Clin Cancer Res 22(2):291–300 Chong DQ, Zhu AX, 2016, The landscape of targeted therapies for cholangiocarcinoma: current status and emerging targets. Oncotarget 7(29):46750–46767 Maroni L, Pierantonelli I, Banales JM, Benedetti A, Marzioni M, 2013, The significance of genetics for cholangiocarcinoma development. Ann Transl Med 1(3):28 Dalmasso C, Carpentier W, Guettier C, Camilleri-Broet S, Borelli WV, Campos Dos Santos CR, Castaing D, Duclos-Vallee JC, Broet P, 2015, Patterns of chromosomal copy-number alterations in intrahepatic cholangiocarcinoma. BMC Cancer 15:126 Arnold A, Bahra M, Lenze D, Bradtmoller M, Guse K, Gehlhaar C, Blaker H, Heppner FL, Koch A, 2015, Genome wide DNA copy number analysis in cholangiocarcinoma using high resolution molecular inversion probe single nucleotide polymorphism assay. Exp Mol Pathol 99(2):344–353 Oliveira IS, Kilcoyne A, Everett JM, Mino-Kenudson M, Harisinghani MG, Ganesan K, 2017, Cholangiocarcinoma: classification, diagnosis, staging, imaging features, and management. Abdom Radiol, NY, 42(6):1637–1649 Andersen JB, Spee B, Blechacz BR, Avital I, Komuta M, Barbour A, Conner EA, Gillen MC, Roskams T, Roberts LR, Factor VM, Thorgeirsson SS, 2012, Genomic and genetic characterization of cholangiocarcinoma identifies therapeutic targets for tyrosine kinase inhibitors. Gastroenterology 142(4):1021–1031 e1015 Sia D, Hoshida Y, Villanueva A, Roayaie S, Ferrer J, Tabak B, Peix J, SoleM, Tovar V, Alsinet C, Cornella H, Klotzle B, Fan JB, Cotsoglou C, Thung SN, Fuster J, Waxman S, Garcia-Valdecasas JC, Bruix J, Schwartz ME, Beroukhim R, Mazzaferro V, Llovet JM, 2013, Integrative molecular analysis of intrahepatic cholangiocarcinoma reveals 2 classes that have different outcomes. Gastroenterology 144(4):829–840 Guest RV, Boulter L, Kendall TJ, Minnis-Lyons SE, Walker R, Wigmore SJ, Sansom OJ, Forbes SJ, 2014, Cell lineage tracing reveals a biliary origin of intrahepatic cholangiocarcinoma. Cancer Res 74(4):1005–1010 Kongpetch S, Jusakul A, Ong CK, Lim WK, Rozen SG, Tan P, Teh BT, 2015, Pathogenesis of cholangiocarcinoma: from genetics to signalling pathways. Best Pract Res Clin Gastroenterol 29(2):233–244 Oikawa T, 2016, Cancer stem cells and their cellular origins in primary liver and biliary tract cancers. Hepatology 64(2):645–651 Vijgen S, Terris B, Rubbia-Brandt L, 2017, Pathology of intrahepatic cholangiocarcinoma. Hepatobiliary Surg Nutr 6(1):22–34 Terada M, Horisawa K,Miura S, Takashima Y, Ohkawa Y, Sekiya S, Matsuda-Ito K, Suzuki A, 2016, Kupffer cells induce notchmediated hepatocyte conversion in a common mouse model of intrahepatic cholangiocarcinoma. Sci Rep 6:34691 Shibata M, Shen MM, 2013, The roots of cancer: stem cells and the basis for tumor heterogeneity. Bioessays 35(3):253–260 Brandi G, Farioli A, Astolfi A, Biasco G, Tavolari S, 2015, Genetic heterogeneity in cholangiocarcinoma: a major challenge for targeted therapies. Oncotarget 6(17):14744–14753 Walter D, Doring C, Feldhahn M, Battke F, Hartmann S, Winkelmann R, Schneider M, Bankov K, Schnitzbauer A, Zeuzem S, Hansmann ML, Peveling-Oberhag J, 2017, Intratumoral heterogeneity of intrahepatic cholangiocarcinoma. Oncotarget 8(9):14957–14968 Chan-On W, Nairismagi ML, Ong CK, Lim WK, Dima S, Pairojkul C, Lim KH, McPherson JR, Cutcutache I, Heng HL, Ooi L, Chung A, Chow P, Cheow PC, Lee SY, Choo SP, Tan IB, Duda D, Nastase A, Myint SS, Wong BH, Gan A, Rajasegaran V, Ng CC, Nagarajan S, Jusakul A, Zhang S, Vohra P, Yu W, Huang D, Sithithaworn P, Yongvanit P, Wongkham S, Khuntikeo N, Bhudhisawasdi V, Popescu I, Rozen SG, Tan P, Teh BT, 2013, Exome sequencing identifies distinct mutational patterns in liver fluke-related and non-infection-related bile duct cancers. Nat Genet 45(12):1474–1478 Cardinale V, Carpino G, Reid L, Gaudio E, Alvaro D, 2012, Multiple cells of origin in cholangiocarcinoma underlie biological, epidemiological and clinical heterogeneity. World J Gastrointest Oncol 4(5):94–102 Li Z, Shen J, Chan MT,WuWK, 2017, The role of microRNAs in intrahepatic cholangiocarcinoma. J Cell Mol Med 21(1):177–184 Wang N, Xia S, Chen K, Xiang X, Zhu A, 2015, Genetic alteration regulated by microRNAs in biliary tract cancers. Crit Rev Oncol Hematol 96(2):262–273 Esparza-Baquer A, Labiano I, Bujanda L, Perugorria MJ, Banales JM, 2016, MicroRNAs in cholangiopathies: potential diagnostic and therapeutic tools. Clin Res Hepatol Gastroenterol 40(1):15–27 Olaizola P, Lee-Law PY, Arbelaiz A, Lapitz A, Perugorria MJ, Bujanda L, Banales JM, 2018, MicroRNAs and extracellular vesicles in cholangiopathies. Biochim Biophys Acta 1864(4 Pt B): 1293–1307 Loosen SH, Schueller F, Trautwein C, Roy S, Roderburg C, 2017, Role of circulating microRNAs in liver diseases. World J Hepatol 9(12):586–594 Mazzocca A, Ferraro G, Misciagna G, Carr BI, 2016, A systemic evolutionary approach to cancer: Hepatocarcinogenesis as a paradigm. Med Hypotheses 93:132–137 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 3 EP 15 DI 10.1007/s12253-018-0491-8 PG 13 ER PT J AU Doma, V Barbai, T Beleaua, MA Kovalszky, I Raso, E Timar, J AF Doma, Viktoria Barbai, Tamas Beleaua, M-A Kovalszky, Ilona Raso, Erzsebet Timar, Jozsef TI KIT Mutation Incidence and Pattern of Melanoma in Central Europe SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Skin melanoma; Mucosal melanoma; KIT mutation and sanger sequencing ID Skin melanoma; Mucosal melanoma; KIT mutation and sanger sequencing AB Data on the KIT mutation rate in melanoma in the central European region is missing. Accordingly, in a cohort of 79 BRAF/ NRAS double wild type cutaneous melanoma and 17 mucosal melanoma KIT mutation was assessed by Sanger sequencing of exons 9,11,13,17 and 18. In this cutaneous melanoma cohort KIT mutation frequency was found to be 34/79 (43.04%) with a significantly higher rate in acrolentiginous melanoma (ALM) as compared to UV-induced common variants (20/34, 58.8% versus 14/45, 31.1%, p = 0.014). In the double wild type mucosal melanoma cohort the KIT mutation frequency was found to be comparable (41.2%). The actual frequency of KIT mutation in the original 227 patient cutaneous melanoma cohort was 34/ 227, 14.9%. Exon 11 was the most frequent mutation site (44.7%) followed by exon 9 (21.1%) equally characterizing UV induced common histotypes and ALM tumors. In mucosal melanoma exon 9 was the most frequently involved exon followed by exon 13 and 17. KIT mutation hotspots were identified in exon 9 (c482/491/492), in exon 11 (c559,c572, c570), in exon 13 (c642), in exon 17 (c822) and in exon 18 (c853). The relatively high KIT mutation rate in cutaneous melanoma in this central- European cohort justifies regular testing of this molecular target in this entity, not only in mucosal variants. C1 [Doma, Viktoria] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Barbai, Tamas] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Beleaua, M-A] University of Medicine, Department of Pathology, Pharmacy and ScienceTargu-Mures, Romania. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Raso, Erzsebet] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM jtimar@gmail.com CR Mei L, Du W, Idowu M, von Mehren M, Boikos SA, 2018, Advances and challenges on management of gastrointestinal stromal tumors. Front Oncol 8:135 Scherber RM, Borate U, 2018, How we diagnose and treat systemic mastocytosis in adults. Br J Haematol 180:11–23 Christen F, Hoyer K, Yoshida K, Hou HA, Waldhueter N et al, 2019, Genomic landscape and clonal evolution of acute myeloid leukemia with t(8;21): an international study on 331 patients. Blood 133:1140–1151 Curtin JA, Busam K, Pinkel D et al, 2006, Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol 24:4340–4346 Timar J, Vizkeleti L, Doma V, Barbai T, Raso E, 2016, Genetic progression ofmalignantmelanoma. CancerMetastasis Rev 35:93– 107 Reddy BY, Miller DM, Tsao H, 2017, Somatic driver mutations in melanoma. Cancer 123:2104–2117 Kong Y, Si L, Zhu Y, Xu X, Christopher L et al, 2011, Large-scale analysis of KIT aberrations in Chinese patients with melanoma. Clin Cancer Res 17:1684–1691 Newel F, Kong Y, Wilmott JS, Johansson PA, Ferguson PM. et al., 2019, Whole-genome landscape of mucosal melanoma reveals diverse drivers and therapeutic targets. Nature Comm10:3163 Zhou R, Shi C, TaoW, Li J,Wu J et al, 2019, Analysis of mucosal melanoma whole-genome landscapes reveals clinically relevant genomic aberrations. Clin Cancer Res 25:3548–3560 Niinuma T, Suzuki H, Sugai T, 2018, Molecular characterization and pathogenesis of gastrointestinal stromal tumor. Transl Gastroenterol Hepatol 3:2–15 Moltara ME, Novakovic S, Boc M, Bucic M, Rebersek M et al, 2018, Prevalence of BRAF, NRAS and c-KIT mutations in Slovenian patients with advanced melanoma. Radiol Oncol 52: 289–295 Doma V, Karpati S, Raso E, Barbai T, Timar J, 2019, Dynamic and unpredictable changes in mutant allele fractions of BRAF and NRAS during visceral progression of cutaneous malignant melanoma. BMC Cancer 19:786 Gong ZH, Zheng HY, Li J, 2018, The clinical significance of KIT mutations in melanoma: a meta-analysis. Melanoma Res 28:259– 270 Ponti G, Manfredini M, Greco S, Pellacani G, Depenni R et al, 2017, BRAF,NRAS and c-KIT advanced melanoma: clinicopathological features, target-therapy strategies and survival. Anticancer Res 37:7043–7048 Cinotti E, Chevallier J, Labeille B, Cambazard F, Thomas L et al, 2017, Mucosal melanoma: clinical, histological and c-kit gene mutational profile of 86 Frech cases. J Eur Acad Dermatol Venerol 31:1834–1840 Babei MA, Kamalidehghan B, Saleem M, Huri HZ, Ahmadipour F, 2016, Receptor tyrosine kinase, c-KIT, inhibitors: a potential therapeutic target in cancer cells. Drug. Design Dev Ther 10:2443– 2459 Carvajal RD, Antonescu CR, Wolchok JD, Chapman PB, Roman RA et al, 2011, KIT as a therapeutic target in metastatic melanoma. JAMA 305:2327–2334 Hodi FS, Corless CL, Jonathan AG-H, Fletcher JA, Zhu M et al, 2013, Imatinib for melanomas harbouring mutationally activated or amplified KIT arising on mucosal, acral and chronically sundamaged skin. J Clin Oncol 31:3182–3190 Guo J, Carvajal RD, Dummer R, Hauschild A, Daud A et al, 2017, Efficacy and safety of nilotinib in patients with KIT-mutated metastatic or inoperable melanoma: final results from the global, single arm, phase II. TEAM trial. Ann Oncol 28:1380–1387 Meng D, Carvajal RD, 2019, KIT as an oncogenic driver in melanoma: an update on clinical development. Am J Clin Dermatol 20: 315–323 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 17 EP 22 DI 10.1007/s12253-019-00788-w PG 6 ER PT J AU Petovari, G Danko, T Krencz, I Hujber, Z Rajnai, H Vetlenyi, E Raffay, R Papay, J Jeney, A Sebestyen, A AF Petovari, Gabor Danko, Titanilla Krencz, Ildiko Hujber, Zoltan Rajnai, Hajnalka Vetlenyi, Eniko Raffay, Regina Papay, Judit Jeney, Andras Sebestyen, Anna TI Inhibition of Metabolic Shift can Decrease Therapy Resistance in Human High-Grade Glioma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; Metabolism; Metabolic shift; mTORC2; Combination therapy ID Glioma; Metabolism; Metabolic shift; mTORC2; Combination therapy AB The high-grade brain malignancy, glioblastoma multiforme (GBM), is one of the most aggressive tumours in central nervous system. The developing resistance against recent therapies and the recurrence rate of GBMs are extremely high. In spite several new ongoing trials, GBM therapies could not significantly increase the survival rate of the patients as significantly. The presence of inter- and intra-tumoral heterogeneity of GBMs arise the problem to find both the pre-existing potential resistant clones and the cellular processes which promote the adaptation mechanisms such as multidrug resistance, stem cell-ness or metabolic alterations, etc. In our work, the in situ metabolic heterogeneity of high-grade human glioblastoma cases were analysed by immunohistochemistry using tissue-microarray. The potential importance of the detected metabolic heterogeneity was tested in three glioma cell lines (grade III-IV) using protein expression analyses (Western blot and WES Simple) and therapeutic drug (temozolomide), metabolic inhibitor treatments (including glutaminase inhibitor) to compare the effects of rapamycin (RAPA) and glutaminase inhibitor combinations in vitro (Alamar Blue and SRB tests). The importance of individual differences and metabolic alterations were observed in mono-therapeutic failures, especially the enhanced Rictor expressions after different monotreatments in correlation to lower sensitivity (temozolomide, doxycycline, etomoxir, BPTES). RAPA combinations with other metabolic inhibitors were the best strategies except for RAPA+glutaminase inhibitor. These observations underline the importance of multi-targeting metabolic pathways. Finally, our data suggest that the detected metabolic heterogeneity (the high mTORC2 complex activity, enhanced expression of Rictor, p-Akt, p-S6, CPT1A, and LDHA enzymes in glioma cases) and the microenvironmental or treatment induced metabolic shift can be potential targets in combination therapy. Therefore, it should be considered to map tissue heterogeneity and alterations with several cellular metabolism markers in biopsy materials after applying recently available or new treatments. C1 [Petovari, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Danko, Titanilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Krencz, Ildiko] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Hujber, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Rajnai, Hajnalka] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Vetlenyi, Eniko] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Raffay, Regina] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Papay, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Jeney, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. RP Sebestyen, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM anna@korb1.sote.hu CR Abrams DA, Hanson JA, Brown JM, Hsu FP, Delashaw JB Jr, Bota DA, 2015, Timing of surgery and bevacizumab therapy in neurosurgical patients with recurrent high grade glioma. J Clin Neurosci 22(1):35–39 Omuro A, Chan TA, Abrey LE, Khasraw M, Reiner AS, Kaley TJ, Deangelis LM, Lassman AB, Nolan CP, Gavrilovic IT, Hormigo A, Salvant C, Heguy A, Kaufman A, Huse JT, Panageas KS, Hottinger AF, Mellinghoff I, 2013, Phase II trial of continuous low-dose temozolomide for patients with recurrent malignant glioma. Neuro-Oncology 15(2):242–250. , DOI 10.1093/neuonc/nos295 Daniel P, Sabri S, Chaddad A, Meehan B, Jean-Claude B, Rak J, Abdulkarim BS, 2019, Temozolomide induced Hypermutation in glioma: evolutionary mechanisms and therapeutic opportunities. Front Oncol 9:41. , DOI 10.3389/fonc.2019.00041 Wang J, Cazzato E, Ladewig E, Frattini V, Rosenbloom DI, Zairis S, Abate F, Liu Z, Elliott O, Shin YJ, Lee JK, Lee IH, ParkWY, Eoli M, Blumberg AJ, Lasorella A, Nam DH, Finocchiaro G, Iavarone A, Rabadan R, 2016, Clonal evolution of glioblastoma under therapy. Nat Genet 48:768–776. , DOI 10.1038/ng.3590 Wu A, Maxwell R,XiaY, Cardarelli P, Oyasu M, Belcaid Z, KimE, Hung A, Luksik AS, Garzon-Muvdi T, Jackson CM, Mathios D, Theodros D, Cogswell J, Brem H, Pardoll DM, Lim M, 2019, Combination anti-CXCR4 and anti-PD-1 immunotherapy provides survival benefit in glioblastoma through immune cell modulation of tumor microenvironment. J Neuro-Oncol. , DOI 10.1007/ s11060-019-03172-5 Hodges TR, OttM, Xiu J, Gatalica Z, Swensen J, Zhou S, Huse JT, deGroot J, Li S, OverwijkWW, SpetzlerD, Heimberger AB, 2017, Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro-Oncology 19:1047–1057. , DOI 10.1093/ neuonc/nox026 Jain KK, 2018, A critical overview of targeted therapies for glioblastoma. Front Oncol. , DOI 10.3389/fonc.2018.00419 Hu LS, Ning S, Eschbacher JM, Baxter LC, Gaw N, Ranjbar S, Plasencia J, Dueck AC, Peng S, Smith KA, Nakaji P, Karis JP, Quarles CC, Wu T, Loftus JC, Jenkins RB, Sicotte H, Kollmeyer TM, O'Neill BP, Elmquist W, Hoxworth JM, Frakes D, Sarkaria J, Swanson KR, Tran NL, Li J,Mitchell JR, 2017, Radiogenomics to characterize regional genetic heterogeneity in glioblastoma. Neuro- Oncology 19(1):128–137. , DOI 10.1093/neuonc/now135 Reinartz R, Wang S, Kebir S, Silver DJ, Wieland A, Zheng T, Kupper M, Rauschenbach L, Fimmers R, Shepherd TM, Trageser D, Till A, Schafer N, Glas M, Hillmer AM, Cichon S, Smith AA, Pietsch T, Liu Y, Reynolds BA, Yachnis A, Pincus DW, Simon M, Brustle O, Steindler DA, Scheffler B, 2017, Functional subclone profiling for prediction of treatment-induced intratumor population shifts and discovery of rational drug combinations in human glioblastoma. Clin Cancer Res. 15;23(2):562–574. , DOI 10. 1158/1078-0432.CCR-15-2089 Parker NR, Hudson AL, Khong P, Parkinson JF, Dwight T, Ikin RJ, Zhu Y, Cheng ZJ, Vafaee F, Chen J, Wheeler HR, Howell VM, 2016, Intratumoral heterogeneity identified at the epigenetic, genetic and transcriptional level in glioblastoma. Sci Rep 4(6):22477. , DOI 10.1038/srep22477 Uribe D, Torres A, Rocha JD, Niechi I, Oyarzun C, Sobrevia L, Martin RS, Quezada C, 2017, Multidrug resistance in glioblastoma stem-like cells: role of the hypoxic microenvironment and adenosine signaling. Mol Asp Med 55:140–151. , DOI 10.1016/ j.mam.2017.01.009 Roos A, Ding Z, Loftus JC, Tran NL, 2017, Molecular and microenvironmental determinants of glioma stem-like cell survival and invasion. Front Oncol 7:120. , DOI 10. 3389/fonc.2017.00120 Agnihotri S, Zadeh G, 2016, Metabolic reprogramming in glioblastoma: the influence of cancer metabolism on epigenetics and unanswered questions. Neuro-Oncology 18(2):160–172. , DOI 10.1093/neuonc/nov125 Libby CJ, Tran AN, Scott SE, Griguer C, Hjelmeland AB, 2018, The pro-tumorigenic effects of metabolic alterations in glioblastoma including brain tumor initiating cells. Biochim Biophys Acta 1869(2):175–188. , DOI 10.1016/j.bbcan.2018.01.004 Jhanwar-UniyalM,Wainwright JV, Mohan AL, Tobias ME, Murali R, Gandhi CD, SchmidtMH, 2019, Diverse signaling mechanisms of mTOR complexes: mTORC1 and mTORC2 in forming a formidable relationship. Adv Biol Regul 72:51–62. , DOI 10. 1016/j.jbior.2019.03.003 Kim LC, Cook RS, Chen J, 2017, mTORC1 and mTORC2 in cancer and the tumor microenvironment. Oncogene 36(16):2191– 2201. , DOI 10.1038/onc.2016.363 Saxton RA, Sabatini DM, 2017, mTOR signaling in growth, metabolism, and disease. Cell 169(2):361–371. , DOI 10. 1016/j.cell.2017.03.035 Guerra F, Arbini AA, Moro L, 2017, Mitochondria and cancer chemoresistance. Biochim Biophys Acta 1858(8):686–699. , DOI 10.1016/j.bbabio.2017.01.012 Melone MAB, Valentino A, Margarucci S, Galderisi U, Giordano A, Peluso G, 2018, The carnitine system and cancer metabolic plasticity. Cell Death Dis 9(2):228. , DOI 10.1038/ s41419-018-0313-7 Pezze PD, Ruf S, Sonntag AG, Langelaar-Makkinje M, Hall P, Heberle AM, Navas PR, van Eunen K, Tolle RC, Schwarz JJ, Wiese H, Warscheid B, Deitersen J, Stork B, Faßler E, Schauble S, Hahn U, Horvatovich P, Shanley DP, Thedieck K, 2016, A systems study reveals concurrent activation of AMPK and mTOR by amino acids. Nature Comm volume 7, Article number: 13254 KeenanMM, Chi JT, 2015, Alternative fuels for cancer cells. Cancer J 21(2):49–55. , DOI 10.1097/PPO.0000000000000104 Petovari G, Hujber Z, Krencz I, Danko T, Nagy N, Toth F, Raffay R,MeszarosK, Rajnai H, Vetlenyi E, Takacs-Vellai K, Jeney A, Sebestyen A, 2018, Targeting cellular metabolism using rapamycin and/or doxycycline enhances anti-tumour effects in human glioma cells. Cancer Cell Int 18:211. https://doi. org/10.1186/s12935-018-0710-0 Bo C, Hong-gang Z, Wei W, Wen-guang G, Dong Z, Peng W, 2016, Studies on antitumor activity spectrum of doxycycline. J Solid Tumors. , DOI 10.5430/jst.v6n1p103 Lin H, Patel S, Affleck VS, Wilson I, Turnbull DM, Joshi AR, Maxwell R, Stoll EA, 2017, Fatty acid oxidation is required for the respiration and proliferation of malignant glioma cells. Neuro- Oncology 19(1):43–54. , DOI 10.1093/neuonc/now128 Ryu CH, Yoon WS, Park KY, Kim SM, Lim JY, Woo JS, Jeong CH, Hou Y, Jeun SS, 2012, Valproic acid downregulates the expression of MGMT and sensitizes temozolomide-resistant glioma cells. J Biomed Biotechnol 2012:987495. , DOI 10.1155/2012/987495 KimEL,Wustenberg R, RubsamA, Schmitz-Salue C,Warnecke G, Bucker EM, Pettkus N, Speidel D, Rohde V, Schulz-Schaeffer W, DeppertW, Giese A, 2010, Chloroquine activates the p53 pathway and induces apoptosis in human glioma cells. Neuro-Oncology 12(4):389–400. , DOI 10.1093/neuonc/nop046 Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW, 2016, The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol 131(6):803– 820. , DOI 10.1007/s00401-016-1545-1 Krencz I, Sebestyen A, Fabian K, Mark A, Moldvay J, Khoor A, Kopper L, Papay J, 2016, Expression of mTORC1/2-related proteins in primary and brain metastatic lung adenocarcinoma. Hum Pathol 62:66–73. , DOI 10.1016/j.humpath.2016.12.012 Masui K, Onizuka H, Cavenee WK, Mischel PS, Shibata N, 2019, Metabolic reprogramming in the pathogenesis of glioma: update. Neuropathology 39(1):3–13. , DOI 10.1111/neup.12535 Parada LF, Dirks PB, Wechsler-Reya RJ, 2017, Brain tumor stem cells remain in play. J Clin Oncol. 20;35(21):2428– 2431. , DOI 10.1200/JCO.2017.73.9540 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell. 4;144(5):646–674. , DOI 10.1016/j.cell. 2011.02.013 Hujber Z, Petovari G, Szoboszlai N, Danko T, Nagy N, Kriston C, Krencz I, Paku S, Ozohanics O, Drahos L, Jeney A, Sebestyen A, 2017, Rapamycin, mTORC1 inhibitor, reduces the production of lactate and 2-hydroxyglutarate oncometabolites in IDH1 mutant fibrosarcoma cells. J Exp Clin Cancer Res. 2;36(1):74. , DOI 10.1186/s13046-017-0544-y Liu B, Huang ZB, Chen X, See YX, Chen ZK, Yao HK, 2019, Mammalian target of rapamycin 2, MTOR2, and C-MYCmodulate glucosamine-6-phosphate synthesis in glioblastoma, GBM, cells through glutamine: Fructose-6-phosphate aminotransferase 1, GFAT1). Cell Mol Neurobiol 39(3):415–434. , DOI 10. 1007/s10571-019-00659-7 Basho RK, GilcreaseM,Murthy RK, Helgason T, KarpDD,Meric- Bernstam F, Hess KR, Herbrich SM, Valero V, Albarracin C, Litton JK, Chavez-MacGregor M, Ibrahim NK, Murray JL, Koenig KB, Hong D, Subbiah V, Kurzrock R, Janku F, Moulder SL, 2017, Targeting the PI3K/AKT/mTOR pathway for the treatment of mesenchymal triple-negative breast cancer: evidence from a phase 1 trial of mTOR inhibition in combination with liposomal doxorubicin and bevacizumab. JAMA Oncol. 1;3(4):509–515. https://doi. org/10.1001/jamaoncol.2016.5281 Stepanenko AA, Andreieva SV, Korets KV, Mykytenko DO, Baklaushev VP, Huleyuk NL, Kovalova OA, Kotsarenko KV, Chekhonin VP, Vassetzky YS, Avdieiev SS, Dmitrenko VV, 2016, Temozolomide promotes genomic and phenotypic changes in glioblastoma cells. Cancer Cell Int 16:36. , DOI 10. 1186/s12935-016-0311-8 ShiY, LimSK, Liang Q, Iyer SV,WangHY,Wang Z, Xie X, Sun D, Chen YJ, Tabar V, Gutin P, Williams N, De Brabander JK, Parada LF, 2019, Gboxin is an oxidative phosphorylation inhibitor that targets glioblastoma. Nature 567(7748):341–346. , DOI 10.1038/s41586-019-0993-x Scatena C, Roncella M, Di Paolo A, Aretini P, Menicagli M, Fanelli G, Marini C, Mazzanti CM, Ghilli M, Sotgia F, Lisanti MP, Naccarato AG, 2018, Doxycycline, an inhibitor of mitochondrial biogenesis, effectively reduces cancer stem cells, CSCs, in early breast cancer patients: a clinical pilot study. Front Oncol 12(8): 452. , DOI 10.3389/fonc.2018.00452 Wang X, Qiu Y, Yu Q, Li H, Chen X, Li M, Long Y, Liu Y, Lu L, Tang J, Zhang Z, He Q, 2017, Enhanced glioma therapy by synergistic inhibition of autophagy and tyrosine kinase activity. Int J Pharm. 30;536(1):1–10. , DOI 10.1016/j.ijpharm.2017.09.007 Padmakrishnan CJ, EaswerHV, VijayakurupV, Menon GR, Nair S, Gopala S, 2019, High LC3/Beclin expression correlates with poor survival in glioma: a definitive role for autophagy as evidenced by in vitro autophagic flux. Pathol Oncol Res 25:137. , DOI 10.1007/s12253-017-0310-7 Scalise M, Pochini L, Galluccio M, Console L, Indiveri C, 2017, Glutamine transport and mitochondrial metabolism in cancer cell growth. Front Oncol 11(7):306. , DOI 10. 3389/fonc.2017.00306 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 23 EP 33 DI 10.1007/s12253-019-00677-2 PG 11 ER PT J AU Krencz, I Sebestyen, A Khoor, A AF Krencz, Ildiko Sebestyen, Anna Khoor, Andras TI mTOR in Lung Neoplasms SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Lung neoplasms; mTORC1; mTORC2; mTOR inhibitors; mTOR signaling ID Lung neoplasms; mTORC1; mTORC2; mTOR inhibitors; mTOR signaling AB With the discovery of rapamycin 45 years ago, studies in the mechanistic target of rapamycin (mTOR) field started 2 decades before the identification of the mTOR kinase. Over the years, studies revealed that the mTOR signaling is a master regulator of homeostasis and integrates a variety of environmental signals to regulate cell growth, proliferation, and metabolism. Deregulation of mTOR signaling, particularly hyperactivation, frequently occurs in human tumors. Recent advances in molecular profiling have identified mutations or amplification of certain genes coding proteins involved in the mTOR pathway (e.g. PIK3CA, PTEN, STK11, and RICTOR) as the most common reasons contributing to mTOR hyperactivation. These genetic alterations of the mTOR pathway are frequently observed in lung neoplasms and may serve as a target for personalized therapy. mTOR inhibitor monotherapy has met limited clinical success so far; however, rational drug combinations are promising to improve efficacy and overcome acquired resistance. A better understanding of mTOR signaling may have the potential to help translation of mTOR pathway inhibitors into the clinical setting. C1 [Krencz, Ildiko] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Khoor, Andras] Mayo Clinic, Department of Laboratory Medicine and Pathology, 4500 San Pablo Road, 32224 Jacksonville, FL, USA. RP Khoor, A (reprint author), Mayo Clinic, Department of Laboratory Medicine and Pathology, 32224 Jacksonville, USA. EM Khoor.Andras@mayo.edu CR Vezina C, Kudelski A, Sehgal SN, 1975, Rapamycin, AY-22, 989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. J Antibiot, Tokyo, 28(10):721–726. , DOI 10.7164/antibiotics.28. 721 Eng CP, Sehgal SN, Vezina C, 1984, Activity of rapamycin, AY- 22,989, against transplanted tumors. J Antibiot, Tokyo, 37(10): 1231–1237. , DOI 10.7164/antibiotics.37.1231 Martel RR, Klicius J, Galet S, 1977, Inhibition of the immune response by rapamycin, a new antifungal antibiotic. Can J Physiol Pharmacol 55(1):48–51. , DOI 10.1139/y77-007 Sabatini DM, Erdjument-Bromage H, Lui M, Tempst P, Snyder SH, 1994, RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs. Cell 78(1):35–43. , DOI 10.1016/0092-8674(94)90570-3 Sabers CJ, Martin MM, Brunn GJ, Williams JM, Dumont FJ, Wiederrecht G, Abraham RT, 1995, Isolation of a protein target of the FKBP12-rapamycin complex in mammalian cells. J Biol Chem 270(2):815–822. , DOI 10.1074/jbc.270.2.815 Laplante M, Sabatini DM, 2012, mTOR signaling in growth control and disease. Cell 149(2):274–293. , DOI 10.1016/j. cell.2012.03.017 Gkountakos A, Pilotto S, Mafficini A, Vicentini C, Simbolo M, Milella M, Tortora G, Scarpa A, Bria E, Corbo V, 2018, Unmasking the impact of Rictor in cancer: novel insights of mTORC2 complex. Carcinogenesis. , DOI 10.1093/ carcin/bgy086 Saxton RA, Sabatini DM, 2017, mTOR signaling in growth, metabolism, and disease. Cell 168(6):960–976. , DOI 10. 1016/j.cell.2017.02.004 Shimobayashi M, Hall MN, 2014, Making new contacts: the mTOR network in metabolism and signalling crosstalk. Nat Rev Mol Cell Biol 15(3):155–162. , DOI 10.1038/nrm3757 Tian T, Li X, Zhang J, 2019, mTOR signaling in cancer and mTOR inhibitors in solid tumor targeting therapy. Int J Mol Sci 20(3). , DOI 10.3390/ijms20030755 Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, Abraham RT, 2017, The PI3K pathway in human disease. Cell 170(4):605–635. , DOI 10.1016/j.cell.2017.07.029 Yip CK, Murata K, Walz T, Sabatini DM, Kang SA, 2010, Structure of the human mTOR complex I and its implications for rapamycin inhibition. Mol Cell 38(5):768–774. https://doi. org/10.1016/j.molcel.2010.05.017 Shaw RJ, 2009, LKB1 and AMP-activated protein kinase control of mTOR signalling and growth. Acta Physiol, Oxf, 196(1):65– 80. , DOI 10.1111/j.1748-1716.2009.01972.x Kim E, Goraksha-Hicks P, Li L, Neufeld TP, Guan KL, 2008, Regulation of TORC1 by Rag GTPases in nutrient response. Nat Cell Biol 10(8):935–945. , DOI 10.1038/ncb1753 Mossmann D, Park S, Hall MN, 2018, mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat Rev Cancer 18(12):744–757. , DOI 10.1038/s41568-018- 0074-8 Fingar DC, Salama S, Tsou C, Harlow E, Blenis J, 2002, Mammalian cell size is controlled by mTOR and its downstream targets S6K1 and 4EBP1/eIF4E. Genes Dev 16(12):1472–1487. , DOI 10.1101/gad.995802 Peterson TR, Sengupta SS, Harris TE, Carmack AE, Kang SA, Balderas E, Guertin DA, Madden KL, Carpenter AE, Finck BN, Sabatini DM, 2011, mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell 146(3):408–420. https:// doi.org/10.1016/j.cell.2011.06.034 Porstmann T, Santos CR, Griffiths B, Cully M,Wu M, Leevers S, Griffiths JR, Chung YL, Schulze A, 2008, SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab 8(3):224–236. , DOI 10.1016/j.cmet. 2008.07.007 Ben-Sahra I, Howell JJ, Asara JM, Manning BD, 2013, Stimulation of de novo pyrimidine synthesis by growth signaling through mTOR and S6K1. Science 339(6125):1323–1328. , DOI 10.1126/science.1228792 Ben-Sahra I, Hoxhaj G, Ricoult SJH, Asara JM, Manning BD, 2016, mTORC1 induces purine synthesis through control of the mitochondrial tetrahydrofolate cycle. Science 351(6274):728– 733. , DOI 10.1126/science.aad0489 Kim J, Kundu M, Viollet B, Guan KL, 2011, AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 13(2):132–141. , DOI 10.1038/ncb2152 Martina JA, Chen Y, Gucek M, Puertollano R, 2012, MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB. Autophagy 8(6):903–914. https://doi. org/10.4161/auto.19653 Cunningham JT, Rodgers JT, Arlow DH, Vazquez F,Mootha VK, Puigserver P, 2007, mTOR controls mitochondrial oxidative function through a YY1-PGC-1alpha transcriptional complex. Nature 450(7170):736–740. , DOI 10.1038/nature06322 Cam H, Easton JB, High A, Houghton PJ, 2010, mTORC1 signaling under hypoxic conditions is controlled by ATM-dependent phosphorylation of HIF-1alpha. Mol Cell 40(4):509–520. https:// doi.org/10.1016/j.molcel.2010.10.030 Dodd KM, Yang J, Shen MH, Sampson JR, Tee AR, 2015, mTORC1 drives HIF-1alpha and VEGF-A signalling via multiple mechanisms involving 4E-BP1, S6K1 and STAT3. Oncogene 34(17):2239–2250. , DOI 10.1038/onc.2014.164 Duvel K, Yecies JL,Menon S, Raman P, Lipovsky AI, Souza AL, Triantafellow E, Ma Q, Gorski R, Cleaver S, Vander Heiden MG, MacKeigan JP, Finan PM, Clish CB, Murphy LO, Manning BD, 2010, Activation of a metabolic gene regulatory network downstream of mTOR complex 1.Mol Cell 39(2):171–183. https://doi. org/10.1016/j.molcel.2010.06.022 Tsang CK, ChenM, Cheng X, Qi Y, Chen Y, Das I, Li X, Vallat B, Fu LW, Qian CN, Wang HY, White E, Burley SK, Zheng XFS, 2018, SOD1 phosphorylation bymTORC1 couples nutrient sensing and redox regulation. Mol Cell 70(3):502–515.e508. https:// doi.org/10.1016/j.molcel.2018.03.029 Stuttfeld E, Aylett CH, Imseng S, Boehringer D, Scaiola A, Sauer E, Hall MN, Maier T, Ban N, 2018, Architecture of the human mTORC2 core complex. Elife 7. , DOI 10.7554/eLife. 33101 Sarbassov DD, Guertin DA, Ali SM, Sabatini DM, 2005, Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 307(5712):1098–1101. , DOI 10.1126/ science.1106148 Kazyken D,Magnuson B, Bodur C, Acosta-Jaquez HA, Zhang D, Tong X, Barnes TM, Steinl GK, Patterson NE, Altheim CH, Sharma N, Inoki K, Cartee GD, Bridges D, Yin L, Riddle SM, Fingar DC, 2019, AMPK directly activates mTORC2 to promote cell survival during acute energetic stress. Sci Signal 12(585). , DOI 10.1126/scisignal.aav3249 Luo Y, Xu W, Li G, Cui W, 2018, Weighing In on mTOR Complex 2 Signaling: The Expanding Role in Cell Metabolism. Oxid Med Cell Longev 2018:7838647. , DOI 10.1155/ 2018/7838647 Chen CH, Shaikenov T, Peterson TR, Aimbetov R, Bissenbaev AK, Lee SW, Wu J, Lin HK Sarbassov dos D, 2011, ER stress inhibits mTORC2 and Akt signaling through GSK-3betamediated phosphorylation of rictor. Sci Signal 4(161):ra10. , DOI 10.1126/scisignal.2001731 Dibble CC, Asara JM, Manning BD, 2009, Characterization of Rictor phosphorylation sites reveals direct regulation of mTOR complex 2 by S6K1. Mol Cell Biol 29(21):5657–5670. https:// doi.org/10.1128/mcb.00735-09 Koo J,Wu X,Mao Z, Khuri FR, Sun SY, 2015, Rictor Undergoes Glycogen Synthase Kinase 3, GSK3)-dependent, FBXW7- mediated Ubiquitination and Proteasomal Degradation. J Biol Chem 290(22):14120–14129. , DOI 10.1074/jbc.M114. 633057 Zinzalla V, Stracka D, OppligerW, Hall MN, 2011, Activation of mTORC2 by association with the ribosome. Cell 144(5):757–768. , DOI 10.1016/j.cell.2011.02.014 Engelman JA, 2009, Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer 9(8):550– 562. , DOI 10.1038/nrc2664 Fumarola C, Bonelli MA, Petronini PG, Alfieri RR, 2014, Targeting PI3K/AKT/mTOR pathway in non small cell lung cancer. Biochem Pharmacol 90(3):197–207. , DOI 10.1016/ j.bcp.2014.05.011 Yehia L, Ngeow J, Eng C, 2019, PTEN-opathies: from biological insights to evidence-based precision medicine. J Clin Invest 129(2):452–464. , DOI 10.1172/jci121277 Shorning BY, Griffiths D, Clarke AR, 2011, Lkb1 and Pten synergise to suppress mTOR-mediated tumorigenesis and epithelial-mesenchymal transition in the mouse bladder. PLoS One 6(1):e16209. , DOI 10.1371/journal.pone.0016209 Harris TK, 2003, PDK1 and PKB/Akt: ideal targets for development of new strategies to structure-based drug design. IUBMB L i f e 5 5, 3 , : 11 7 – 1 2 6 . h t t p s : / / d o i . o rg / 1 0 . 1 0 8 0 / 1521654031000115951 Hart JR, Vogt PK, 2011, Phosphorylation of AKT: a mutational analysis. Oncotarget 2(6):467–476 Arsham AM, Howell JJ, Simon MC, 2003, A novel hypoxiainducible factor-independent hypoxic response regulating mammalian target of rapamycin and its targets. J Biol Chem 278(32): 29655–29660. , DOI 10.1074/jbc.M212770200 Brinkhuizen T, Weijzen CA, Eben J, Thissen MR, van Marion AM, Lohman BG, Winnepenninckx VJ, Nelemans PJ, van Steensel MA, 2014, Immunohistochemical analysis of the mechanistic target of rapamycin and hypoxia signalling pathways in basal cell carcinoma and trichoepithelioma. PLoS One 9(9): e106427. , DOI 10.1371/journal.pone.0106427 Brugarolas J, Lei K, Hurley RL, Manning BD, Reiling JH, Hafen E, Witters LA, Ellisen LW, Kaelin WG Jr, 2004, Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/ TSC2 tumor suppressor complex. Genes Dev 18(23):2893–2904. , DOI 10.1101/gad.1256804 Kim J, Guan KL, 2019, mTOR as a central hub of nutrient signalling and cell growth. Nat Cell Biol 21(1):63–71. , DOI 10.1038/s41556-018-0205-1 Roccio M, Bos JL, Zwartkruis FJ, 2006, Regulation of the small GTPase Rheb by amino acids. Oncogene 25(5):657–664. https:// doi.org/10.1038/sj.onc.1209106 Bar-Peled L, Chantranupong L, Cherniack AD, ChenWW, Ottina KA, Grabiner BC, Spear ED, Carter SL, Meyerson M, Sabatini DM, 2013, A Tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1. Science 340(6136):1100–1106. , DOI 10.1126/science. 1232044 Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC, Bar-Peled L, Sabatini DM, 2008, The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 320(5882):1496–1501. , DOI 10.1126/science. 1157535 Cheng H, Zou Y, Ross JS,Wang K, Liu X, Halmos B,Ali SM, Liu H, Verma A, Montagna C, Chachoua A, Goel S, Schwartz EL, Zhu C, Shan J, Yu Y, Gritsman K, Yelensky R, Lipson D, Otto G, Hawryluk M, Stephens PJ, Miller VA, Piperdi B, Perez-Soler R, 2015, RICTOR amplification defines a novel subset of patients with lung cancer who may benefit from treatment with mTORC1/ 2 inhibitors. Cancer Discov 5(12):1262–1270. , DOI 10. 1158/2159-8290.Cd-14-0971 Janku F, Yap TA, Meric-Bernstam F, 2018, Targeting the PI3K pathway in cancer: are we making headway? Nat Rev Clin Oncol 15(5):273–291. , DOI 10.1038/nrclinonc.2018.28 Conde E, Angulo B, Tang M,MorenteM, Torres-Lanzas J, Lopez- Encuentra A, Lopez-Rios F, Sanchez-Cespedes M, 2006, Molecular context of the EGFR mutations: evidence for the activation of mTOR/S6K signaling. Clin Cancer Res 12(3 Pt 1):710– 717. , DOI 10.1158/1078-0432.Ccr-05-1362 Papadimitrakopoulou V, 2012, Development of PI3K/AKT/ mTOR pathway inhibitors and their application in personalized therapy for non-small-cell lung cancer. J Thorac Oncol 7(8): 1315–1326. , DOI 10.1097/JTO.0b013e31825493eb Dobashi Y, Watanabe Y, Miwa C, Suzuki S, Koyama S, 2011, Mammalian target of rapamycin: a central node of complex signaling cascades. Int J Clin Exp Pathol 4(5):476–495 Chen B, Tan Z, Gao J,WuW, Liu L, JinW, Cao Y, Zhao S, Zhang W, Qiu Z, Liu D, Mo X, Li W, 2015, Hyperphosphorylation of ribosomal protein S6 predicts unfavorable clinical survival in nonsmall cell lung cancer. J Exp Clin Cancer Res 34:126. https://doi. org/10.1186/s13046-015-0239-1 Krencz I, Sebestyen A, Fabian K, Mark A, Moldvay J, Khoor A, Kopper L, Papay J, 2017, Expression of mTORC1/2-related proteins in primary and brain metastatic lung adenocarcinoma. Hum Pathol 62:66–73. , DOI 10.1016/j.humpath.2016.12.012 Seki N, Takasu T,Mandai K, NakataM, Saeki H, Heike Y, Takata I, Segawa Y, Hanafusa T, Eguchi K, 2002, Expression of eukaryotic initiation factor 4E in atypical adenomatous hyperplasia and adenocarcinoma of the human peripheral lung. Clin Cancer Res 8(10):3046–3053 Yoshizawa A, Fukuoka J, Shimizu S, Shilo K, Franks TJ, Hewitt SM, Fujii T, Cordon-Cardo C, Jen J, Travis WD, 2010, Overexpression of phospho-eIF4E is associated with survival through AKT pathway in non-small cell lung cancer. Clin Cancer Res 16(1):240–248. , DOI 10.1158/1078-0432. Ccr-09-0986 Ekman S, Wynes MW, Hirsch FR, 2012, The mTOR pathway in lung cancer and implications for therapy and biomarker analysis. J Thorac Oncol 7(6):947–953. , DOI 10.1097/JTO. 0b013e31825581bd Facchinetti F, BluthgenMV, Tergemina-Clain G, Faivre L, Pignon JP, Planchard D, Remon J, Soria JC, Lacroix L, Besse B, 2017, LKB1/STK11 mutations in non-small cell lung cancer patients: Descriptive analysis and prognostic value. Lung Cancer 112:62– 68. , DOI 10.1016/j.lungcan.2017.08.002 Spoerke JM, O'Brien C, Huw L, Koeppen H, Fridlyand J, Brachmann RK, Haverty PM, Pandita A, Mohan S, Sampath D, Friedman LS, Ross L, Hampton GM, Amler LC, Shames DS, Lackner MR, 2012, Phosphoinositide 3-kinase, PI3K, pathway alterations are associated with histologic subtypes and are predictive of sensitivity to PI3K inhibitors in lung cancer preclinical models. Clin Cancer Res 18(24):6771–6783. , DOI 10. 1158/1078-0432.Ccr-12-2347 Imielinski M, Berger AH, Hammerman PS, Hernandez B, Pugh TJ, Hodis E, Cho J, Suh J, Capelletti M, Sivachenko A, Sougnez C, Auclair D, Lawrence MS, Stojanov P, Cibulskis K, Choi K, de Waal L, Sharifnia T, Brooks A, Greulich H, Banerji S, Zander T, Seidel D, Leenders F, Ansen S, Ludwig C, Engel-Riedel W, Stoelben E, Wolf J, Goparju C, Thompson K, Winckler W, Kwiatkowski D, Johnson BE, Janne PA, Miller VA, Pao W, Travis WD, Pass HI, Gabriel SB, Lander ES, Thomas RK, Garraway LA, Getz G, Meyerson M, 2012, Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150(6):1107–1120 Vasan N, Boyer JL, Herbst RS, 2014, A RAS renaissance: emerging targeted therapies for KRAS-mutated non-small cell lung cancer. Clin Cancer Res 20(15):3921–3930 Travis WD, Brambilla E, Burke AP, Marx A, Nicholson AG, 2015)WHO classification of tumours of the lung, pleura, thymus and heart. International Agency for Research on Cancer, Lyon Gazdar AF, Bunn PA, Minna JD, 2017, Small-cell lung cancer: what we know, what we need to know and the path forward. Nat Rev Cancer 17(12):725–737. , DOI 10.1038/nrc.2017.87 Miyoshi T, Umemura S, Matsumura Y, Mimaki S, Tada S, Makinoshima H, Ishii G, Udagawa H, Matsumoto S, Yoh K, Niho S, Ohmatsu H, Aokage K, Hishida T, Yoshida J, Nagai K, Goto K, Tsuboi M, Tsuchihara K, 2017, Genomic profiling of large-cell neuroendocrine carcinoma of the lung. Clin Cancer Res 23(3):757–765. , DOI 10.1158/1078-0432.Ccr-16- 0355 SimboloM, Mafficini A, Sikora KO, FassanM, Barbi S, Corbo V, Mastracci L, Rusev B, Grillo F, Vicentini C, Ferrara R, Pilotto S, Davini F, Pelosi G, Lawlor RT, Chilosi M, Tortora G, Bria E, Fontanini G, Volante M, Scarpa A, 2017, Lung neuroendocrine tumours: deep sequencing of the four World Health Organization histotypes reveals chromatin-remodelling genes as major players and a prognostic role for TERT, RB1, MEN1 and KMT2D. J Pathol 241(4):488–500 Hendifar AE, Marchevsky AM, Tuli R, 2017, Neuroendocrine tumors of the lung: current challenges and advances in the diagnosis and management of well-differentiated disease. J Thorac Oncol 12(3):425–436. , DOI 10.1016/j.jtho.2016.11. 2222 George J, Lim JS, Jang SJ, Cun Y, Ozretic L, Kong G, Leenders F, Lu X, Fernandez-Cuesta L, Bosco G, Muller C, Dahmen I, Jahchan NS, Park KS, Yang D, Karnezis AN, Vaka D, Torres A, Wang MS, Korbel JO,Menon R, Chun SM, Kim D,Wilkerson M, Hayes N, Engelmann D, Putzer B, Bos M, Michels S, Vlasic I, Seidel D, Pinther B, Schaub P, Becker C, Altmuller J, Yokota J, Kohno T, Iwakawa R, Tsuta K, Noguchi M, Muley T, Hoffmann H, Schnabel PA, Petersen I, Chen Y, Soltermann A, Tischler V, Choi CM, Kim YH, Massion PP, Zou Y, Jovanovic D, Kontic M, Wright GM, Russell PA, Solomon B, Koch I, Lindner M, Muscarella LA, la Torre A, Field JK, Jakopovic M, Knezevic J, Castanos-Velez E, Roz L, Pastorino U, Brustugun OT, Lund- Iversen M, Thunnissen E, Kohler J, Schuler M, Botling J, Sandelin M, Sanchez-Cespedes M, Salvesen HB, Achter V, Lang U, Bogus M, Schneider PM, Zander T, Ansen S, Hallek M, Wolf J, Vingron M, Yatabe Y, Travis WD, Nurnberg P, Reinhardt C, Perner S, Heukamp L, Buttner R, Haas SA, Brambilla E, Peifer M, Sage J, Thomas RK, 2015, Comprehensive genomic profiles of small cell lung cancer. Nature 524(7563):47–53 Rudin CM, Durinck S, Stawiski EW, Poirier JT, Modrusan Z, Shames DS, Bergbower EA, Guan Y, Shin J, Guillory J, Rivers CS, Foo CK, Bhatt D, Stinson J, Gnad F, Haverty PM, Gentleman R, Chaudhuri S, Janakiraman V, Jaiswal BS, Parikh C, Yuan W, Zhang Z, Koeppen H, Wu TD, Stern HM, Yauch RL, Huffman KE, Paskulin DD, Illei PB, Varella-Garcia M, Gazdar AF, de Sauvage FJ, Bourgon R, Minna JD, Brock MV, Seshagiri S, 2012, Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat Genet 44(10):1111–1116. , DOI 10.1038/ng.2405 Umemura S, Mimaki S, Makinoshima H, Tada S, Ishii G, Ohmatsu H, Niho S, Yoh K, Matsumoto S, Takahashi A, Morise M, Nakamura Y, Ochiai A, Nagai K, Iwakawa R, Kohno T, Yokota J, Ohe Y, Esumi H, Tsuchihara K, Goto K, 2014, Therapeutic priority of the PI3K/AKT/mTOR pathway in small cell lung cancers as revealed by a comprehensive genomic analysis. J Thorac Oncol 9(9):1324–1331 Krencz I, Sebestyen A, Papay J, Lou Y, Lutz GF, Majewicz TL, Khoor A, 2019, Correlation between immunohistochemistry and RICTOR FISH amplification in small cell lung carcinoma. Hum Pathol. , DOI 10.1016/j.humpath.2019.08.018 Ross JS, Wang K, Elkadi OR, Tarasen A, Foulke L, Sheehan CE, Otto GA, Palmer G, Yelensky R, Lipson D, Chmielecki J, Ali SM, Elvin J, Morosini D, Miller VA, Stephens PJ, 2014, Nextgeneration sequencing reveals frequent consistent genomic alterations in small cell undifferentiated lung cancer. J Clin Pathol 67(9):772–776 Sakre N,Wildey G, Behtaj M, Kresak A,Yang M, Fu P, DowlatiA, 2017, RICTOR amplification identifies a subgroup in small cell lung cancer and predicts response to drugs targeting mTOR. Oncotarget 8(4):5992–6002 Schmid K, Bago-Horvath Z, Berger W, Haitel A, Cejka D, Werzowa J, Filipits M, Herberger B, Hayden H, Sieghart W, 2010, Dual inhibition of EGFR and mTOR pathways in small cell lung cancer. Br J Cancer 103(5):622–628. , DOI 10. 1038/sj.bjc.6605761 Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, Chirieac LR, Dacic S, Duhig E, Flieder DB, Geisinger K, Hirsch FR, Ishikawa Y, Kerr KM, Noguchi M, Pelosi G, Powell CA, Tsao MS, Wistuba I, 2015, The 2015 World Health Organization Classification of Lung Tumors: Impact of Genetic, Clinical and Radiologic Advances Since the 2004 Classification. J Thorac Oncol 10(9):1243–1260. https://doi. org/10.1097/jto.0000000000000630 Glasgow CG, Steagall WK, Taveira-Dasilva A, Pacheco- Rodriguez G, Cai X, El-Chemaly S, Moses M, Darling T, Moss J, 2010, Lymphangioleiomyomatosis, LAM): molecular insights lead to targeted therapies. Respir Med 104(Suppl 1):S45–S58 Henske EP, McCormack FX(2012, Lymphangioleiomyomatosis - a wolf in sheep's clothing. J Clin Invest 122(11):3807–3816 Smolarek TA, Wessner LL, McCormack FX, Mylet JC, Menon AG, Henske EP, 1998, Evidence that lymphangiomyomatosis is caused by TSC2 mutations: chromosome 16p13 loss of heterozygosity in angiomyolipomas and lymph nodes from women with lymphangiomyomatosis. Am J Hum Genet 62(4):810–815 Badri KR, Gao L, Hyjek E, Schuger N, Schuger L, Qin W, Chekaluk Y, Kwiatkowski DJ, Zhe X, 2013, Exonic mutations of TSC2/TSC1 are common but not seen in all sporadic pulmonary lymphangioleiomyomatosis. Am J Respir Crit Care Med 187(6):663–665. , DOI 10.1164/ajrccm.187.6.663 Carsillo T, Astrinidis A, Henske EP, 2000, Mutations in the tuberous sclerosis complex gene TSC2 are a cause of sporadic pulmonary lymphangioleiomyomatosis. Proc Natl Acad Sci U S A 97(11):6085–6090. , DOI 10.1073/pnas.97.11.6085 Sato T, Seyama K, Fujii H,Maruyama H, SetoguchiY, Iwakami S, Fukuchi Y, Hino O, 2002, Mutation analysis of the TSC1 and TSC2 genes in Japanese patients wi th pulmonary lymphangioleiomyomatosis. J Hum Genet 47(1):20–28. https:// doi.org/10.1007/s10038-002-8651-8 Adachi K, Miki Y, Saito R, Hata S, Yamauchi M, Mikami Y, Okada Y, Seyama K, Kondo T, Sasano H, 2015, Intracrine steroid production and mammalian target of rapamycin pathways in pulmonary lymphangioleiomyomatosis. Hum Pathol 46(11):1685– 1693. , DOI 10.1016/j.humpath.2015.02.019 Hayashi T, Kumasaka T, Mitani K, Okada Y, Kondo T, Date H, Chen F, Oto T,Miyoshi S, Shiraishi T, Iwasaki A, Hara K, Saito T, Ando K, Kobayashi E, Gunji-Niitsu Y, Kunogi M, Takahashi K, Yao T, SeyamaK(2016, Bronchial involvement in advanced stage lymphangioleiomyomatosis: histopathologic and molecular analyses. Hum Pathol 50:34–42. , DOI 10.1016/j.humpath. 2015.11.002 Krencz I, Sebestyen A, Papay J, Jeney A, Hujber Z, Burger CD, Keller CA, Khoor A, 2018, In situ analysis of mTORC1/2 and ce l l u l a r me t a b o l i s m - r e l a t e d pr o t e i n s in human lymphangioleiomyomatosis. Hum Pathol 79:199–207. https://doi. org/10.1016/j.humpath.2018.05.018 McCormack FX, Gupta N, Finlay GR, Young LR, Taveira- DaSilva AM, Glasgow CG, Steagall WK, Johnson SR, Sahn SA, Ryu JH, Strange C, Seyama K, Sullivan EJ, Kotloff RM, Downey GP, Chapman JT, Han MK, D'Armiento JM, Inoue Y, Henske EP, Bissler JJ, Colby TV, Kinder BW, Wikenheiser- Brokamp KA, Brown KK, Cordier JF, Meyer C, Cottin V, Brozek JL, Smith K,Wilson KC,Moss J, 2016, Official american thoracic society/Japanese respiratory society clinical practice guidelines: lymphangioleiomyomatosis diagnosis and management. Am J Respir Crit Care Med 194(6):748–761 Conciatori F, Ciuffreda L, Bazzichetto C, Falcone I, Pilotto S, Bria E, Cognetti F, Milella M, 2018, mTOR cross-talk in cancer and potential for combination therapy. Cancers, Basel, 10(1). https:// doi.org/10.3390/cancers10010023 Zheng Y, Jiang Y, 2015, mTOR inhibitors at a glance. Mol Cell Pharmacol 7(2):15–20 Soria JC, Shepherd FA, Douillard JY, Wolf J, Giaccone G, Crino L, Cappuzzo F, Sharma S, Gross SH, Dimitrijevic S, Di Scala L, Gardner H, Nogova L, Papadimitrakopoulou V, 2009, Efficacy of everolimus, RAD001, in patients with advanced NSCLC previously treated with chemotherapy alone or with chemotherapy and EGFR inhibitors. Ann Oncol 20(10):1674–1681. , DOI 10.1093/annonc/mdp060 Fazio N, Granberg D, Grossman A, Saletan S, Klimovsky J, Panneerselvam A, Wolin EM, 2013, Everolimus plus octreotide long-acting repeatable in patients with advanced lung neuroendocrine tumors: analysis of the phase 3, randomized, placebocontrolled radiant-2 study. Chest 143(4):955–962. https://doi. org/10.1378/chest.12-1108 Pavel ME, Baudin E, Oberg KE, Hainsworth JD, Voi M, Rouyrre N, Peeters M, Gross DJ, Yao JC, 2017, Efficacy of everolimus plus octreotide LAR in patients with advanced neuroendocrine tumor and carcinoid syndrome: final overall survival from the randomized, placebo-controlled phase 3 RADIANT-2 study. Ann Oncol 28(7):1569–1575. , DOI 10.1093/annonc/ mdx193 Yao JC, Fazio N, Singh S, Buzzoni R, Carnaghi C, Wolin E, Tomasek J, Raderer M, Lahner H, Voi M, Pacaud LB, Rouyrre N, Sachs C, Valle JW, Fave GD, Van Cutsem E, Tesselaar M, Shimada Y, Oh DY, Strosberg J, Kulke MH, Pavel ME, 2016, Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract, RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet 387(10022):968–977. , DOI 10.1016/s0140- 6736(15)00817-x Ferolla P, BrizziMP,Meyer T,MansoorW,Mazieres J, Do Cao C, Lena H, Berruti A, Damiano V, Buikhuisen W, Gronbaek H, Lombard-Bohas C, Grohe C, Minotti V, Tiseo M, De Castro J, Reed N,Gislimberti G, SinghN, StankovicM, Oberg K, Baudin E, 2017, Efficacy and safety of long-acting pasireotide or everolimus alone or in combination in patients with advanced carcinoids of the lung and thymus, LUNA): an open-label, multicentre, randomised, phase 2 trial. Lancet Oncol 18(12):1652–1664. , DOI 10.1016/s1470-2045(17)30681-2 Pandya KJ, Dahlberg S, HidalgoM, Cohen RB, LeeMW, Schiller JH, Johnson DH, 2007, A randomized, phase II trial of two dose levels of temsirolimus, CCI-779, in patients with extensive-stage small-cell lung cancer who have responding or stable disease after induction chemotherapy: a trial of the Eastern Cooperative Oncology Group, E1500). J Thorac Oncol 2(11):1036–1041. , DOI 10.1097/JTO.0b013e318155a439 Tarhini A, Kotsakis A, GoodingW, Shuai Y, Petro D, Friedland D, Belani CP, Dacic S, Argiris A, 2010, Phase II study of everolimus, RAD001, in previously treated small cell lung cancer. Clin Cancer Res 16(23):5900–5907. , DOI 10.1158/1078- 0432.Ccr-10-0802 Ando K, KuriharaM, Kataoka H, Ueyama M, Togo S, Sato T, Doi T, Iwakami S, Takahashi K, Seyama K, Mikami M, 2013, Efficacy and safety of low-dose sirolimus for treatment of lymphangioleiomyomatosis. Respir Investig 51(3):175–183. , DOI 10.1016/j.resinv.2013.03.002 Goldberg HJ, Harari S, Cottin V, Rosas IO, Peters E, Biswal S, Cheng Y, Khindri S, Kovarik JM,Ma S, McCormack FX, Henske E P, 2 0 1 5 , E v e r o l i m u s f o r t h e t r e a t m e n t o f lymphangioleiomyomatosis: a phase II study. Eur Respir J 46(3): 783–794. , DOI 10.1183/09031936.00210714 McCormack FX, Inoue Y, Moss J, Singer LG, Strange C, Nakata K, Barker AF, Chapman JT, Brantly ML, Stocks JM, Brown KK, Lynch JP 3rd, Goldberg HJ, Young LR, Kinder BW, Downey GP, Sullivan EJ, Colby TV, McKay RT, Cohen MM, Korbee L, Taveira-DaSilva AM, Lee HS, Krischer JP, Trapnell BC, 2011, Efficacy and safety of sirolimus in lymphangioleiomyomatosis. N Engl J Med 364(17):1595–1606 Yao J, Taveira-DaSilva AM, Jones AM, Julien-Williams P, StylianouM,Moss J, 2014, Sustained effects of sirolimus on lung function and cystic lung lesions in lymphangioleiomyomatosis. Am J Respir Crit Care Med 190(11):1273–1282 Xu KF, Tian X, Yang Y, Zhang H, 2018, Rapamycin for lymphangioleiomyomatosis: optimal timing and optimal dosage. Thorax 73(4):308–310. , DOI 10.1136/thoraxjnl-2017- 211135 Tang Y, El-Chemaly S, Taveira-Dasilva A, Goldberg HJ, Bagwe S, Rosas IO, Moss J, Priolo C, Henske EP, 2019, Metabolic changes in patients with lymphangioleiomyomatosis treated with sirolimus and hydroxychloroquine. Chest. , DOI 10.1016/ j.chest.2019.05.038 Yoon HY, Hwang JJ, Kim DS, Song JW, 2018, Efficacy and safety of low-dose Sirolimus in Lymphangioleiomyomatosis. Orphanet J Rare Dis 13(1):204 Basu B, Krebs MG, Sundar R, Wilson RH, Spicer J, Jones R, Brada M, Talbot DC, Steele N, Ingles Garces AH, Brugger W, Harrington EA, Evans J, Hall E, Tovey H, de Oliveira FM, Carreira S, Swales K, Ruddle R, Raynaud FI, Purchase B, Dawes JC, Parmar M, Turner AJ, Tunariu N, Banerjee S, de Bono JS, Banerji U, 2018, Vistusertib, dual m-TORC1/2 inhibitor, in combination with paclitaxel in patients with high-grade serous ovarian and squamous non-small-cell lung cancer. Ann Oncol 29(9):1918–1925 Moore KN, Bauer TM, Falchook GS, Chowdhury S, Patel C, Neuwirth R, Enke A, Zohren F, Patel MR, 2018, Phase I study of the investigational oral mTORC1/2 inhibitor sapanisertib, TAK-228): tolerability and food effects of a milled formulation in patients with advanced solid tumours. ESMO Open 3(2): e000291 Jones AT, Yang J, Narov K, Henske EP, Sampson JR, Shen MH, 2019, Allosteric and ATP-competitive inhibitors of mTOR effectively suppress tumor progression-associated epithelial-mesenchymal transition in the kidneys of Tsc2(+/-, mice. Neoplasia 21(8): 731–739 Langer CJ, RedmanMW,Wade JL 3rd, Aggarwal C, Bradley JD, Crawford J, Stella PJ, Knapp MH, Miao J, Minichiello K, Herbst RS, Kelly K, Gandara DR, Papadimitrakopoulou VA, 2019, SWOG S1400B, NCT02785913), a Phase II study of GDC- 0032, Taselisib, for previously treated PI3K-positive patients with stage IV squamous cell lung cancer, Lung-MAP Sub-Study). J Thorac Oncol 14(10):1839–1846. , DOI 10.1016/j.jtho. 2019.05.029 Adjei AA, Bennouna J, Leighl NB, Felip E, Cortinovis DL, Alt J, Schaefer ES, Thomas M, Chouaid C, Morabito A, De Castro J, Grossi F, Paz-Ares L, De Pas TM, Maier J, Chakravartty A, Chol M, Aimone P, Planchard D, 2016, Safety and efficacy of buparlisib, BKM120, and chemotherapy in advanced, squamous non-small cell lung cancer, sqNSCLC): Results from the phase Ib/ II BASALT-2 and BASALT-3 studies. Journal of Clinical Oncology 34, 15_suppl):e20522-e20522. , DOI 10. 1200/JCO.2016.34.15_suppl.e20522 Vansteenkiste JF, Canon JL, De Braud F, Grossi F, De Pas T, Gray JE, Su WC, Felip E, Yoshioka H, Gridelli C, Dy GK, Thongprasert S, Reck M, Aimone P, Vidam GA, Roussou P, Wang YA, Di Tomaso E, Soria JC, 2015, Safety and efficacy of buparlisib, BKM120, in patients with PI3K pathway-activated non-small cell lung cancer: results from the phase II BASALT-1 study. J Thorac Oncol 10(9):1319–1327 David O, Jett J, LeBeau H, Dy G, Hughes J, Friedman M, Brody AR, 2004, Phospho-Akt overexpression in non-small cell lung cancer confers significant stage-independent survival disadvantage. Clin Cancer Res 10(20):6865–6871 Oh MH, Lee HJ, Yoo SB, Xu X, Choi JS, Kim YH, Lee SY, Lee CT, Jheon S, Chung JH, 2012, Clinicopathological correlations of mTOR and pAkt expression in non-small cell lung cancer. Virchows Arch 460(6):601–609. , DOI 10.1007/s00428- 012-1239-6 Yip PY, Cooper WA, Kohonen-Corish MR, Lin BP, McCaughan BC, Boyer MJ, Kench JG, Horvath LG, 2014, Phosphorylated Akt expression is a prognosticmarker in early-stage non-small cell lung cancer. J Clin Pathol 67(4):333–340. , DOI 10.1136/ jclinpath-2013-201870 Jacobsen K, Bertran-Alamillo J, Molina MA, Teixido C, Karachaliou N, Pedersen MH, Castellvi J, Garzon M, Codony- Servat C, Codony-Servat J, Gimenez-Capitan A, Drozdowskyj A, Viteri S, Larsen MR, Lassen U, Felip E, Bivona TG, Ditzel HJ, Rosell R, 2017, Convergent Akt activation drives acquired EGFR inhibitor resistance in lung cancer. Nat Commun 8(1):410. https:// doi.org/10.1038/s41467-017-00450-6 Puglisi M, Thavasu P, Stewart A, de Bono JS, O'BrienME, Popat S, Bhosle J, Banerji U, 2014, AKT inhibition synergistically enhances growth-inhibitory effects of gefitinib and increases apoptosis in non-small cell lung cancer cell lines. Lung Cancer 85(2): 141–146. , DOI 10.1016/j.lungcan.2014.05.008 Lara PN Jr, Longmate J, Mack PC, Kelly K, Socinski MA, Salgia R, Gitlitz B, Li T, Koczywas M, Reckamp KL, Gandara DR, 2015, Phase II study of theAKTinhibitorMK-2206 plus erlotinib in patients with advanced non-small cell lung cancer who Previously progressed on erlotinib. Clin Cancer Res 21(19): 4321–4326. , DOI 10.1158/1078-0432.CCR-14-3281 Owonikoko TK, Khuri FR, 2013, Targeting the PI3K/AKT/ mTOR pathway: biomarkers of success and tribulation. Am Soc Clin Oncol Educ Book. , DOI 10.1200/EdBook_AM. 2013.33.e395 Gonzalez-Angulo AM, Blumenschein GR Jr, 2013, Defining biomarkers to predict sensitivity to PI3K/Akt/mTOR pathway inhibitors in breast cancer. Cancer Treat Rev 39(4):313–320. https://doi. org/10.1016/j.ctrv.2012.11.002 Serra V, Markman B, Scaltriti M, Eichhorn PJ, Valero V, Guzman M, Botero ML, Llonch E, Atzori F, Di Cosimo S, Maira M, Garcia-Echeverria C, Parra JL, Arribas J, Baselga J, 2008, NVPBEZ235, a dual PI3K/mTOR inhibitor, prevents PI3K signaling and inhibits the growth of cancer cells with activating PI3K mutations. Cancer Res 68(19):8022–8030. , DOI 10.1158/ 0008-5472.Can-08-1385 Yang J, Nie J,Ma X,Wei Y, Peng Y,Wei X, 2019, Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol Cancer 18(1):26. , DOI 10.1186/s12943-019-0954-x Lim SM, Park HS, Kim S, Kim S, Ali SM, Greenbowe JR, Yang IS, Kwon NJ, Lee JL, RyuMH, Ahn JH, Lee J, LeeMG, Kim HS, Kim H, Kim HR,Moon YW, Chung HC, Kim JH, Kang YK, Cho BC, 2016, Next-generation sequencing reveals somatic mutations that confer exceptional response to everolimus. Oncotarget 7(9): 10547–10556. , DOI 10.18632/oncotarget.7234 HymanDM, Smyth LM,DonoghueMTA,Westin SN, Bedard PL, Dean EJ, Bando H, El-Khoueiry AB, Perez-Fidalgo JA, Mita A, Schellens JHM, Chang MT, Reichel JB, Bouvier N, Selcuklu SD, Soumerai TE, Torrisi J, Erinjeri JP, Ambrose H, Barrett JC, Dougherty B, Foxley A, Lindemann JPO, McEwen R, Pass M, Schiavon G, Berger MF, Chandarlapaty S, Solit DB, Banerji U, Baselga J, Taylor BS, 2017, AKT inhibition in solid tumors with AKT1 mutations. Journal of Clinical Oncology 35(20):2251– 2259. , DOI 10.1200/JCO.2017.73.0143 Gkountakos A, Pilotto S, Simbolo M, Vicentini C, Mafficini A, del Curatolo A, Scarpa A, Tortora G, Corbo V, Bria E, 2018, Potential role of RICTOR copy number gain, CNG, as a key biomarker ofmTOR activity: a comprehensive preclinical analysis in squamous cell lung cancer. Ann Oncol 29(suppl_8):viii649– viii669. , DOI 10.1093/annonc/mdy303.049 O'Reilly T,McSheehy PM, 2010, Biomarker development for the clinical activity of the mtor inhibitor everolimus, RAD001): processes, limitations, and further proposals. Transl Oncol 3(2):65– 79. , DOI 10.1593/tlo.09277 Kwei KA, Baker JB, Pelham RJ, 2012, Modulators of sensitivity and resistance to inhibi t ion of PI3K identified in a pharmacogenomic screen of the NCI-60 human tumor cell line collection. PLoS One 7(9):e46518. , DOI 10.1371/ journal.pone.0046518 Lin J, Sampath D, Nannini MA, Lee BB, Degtyarev M, Oeh J, Savage H, Guan Z, Hong R, Kassees R, Lee LB, Risom T, Gross S, Liederer BM, Koeppen H, Skelton NJ, Wallin JJ, Belvin M, Punnoose E, Friedman LS, Lin K, 2013, Targeting activated Akt with GDC-0068, a novel selective Akt inhibitor that is efficacious in multiple tumor models. Clin Cancer Res 19(7):1760–1772. , DOI 10.1158/1078-0432.Ccr-12-3072 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 35 EP 48 DI 10.1007/s12253-020-00796-1 PG 14 ER PT J AU Matyasi, B Farkas, Zs Kopper, L Sebestyen, A Boissan, M Mehta, A Takacs-Vellai, K AF Matyasi, Barbara Farkas, Zsolt Kopper, Laszlo Sebestyen, Anna Boissan, Mathieu Mehta, Anil Takacs-Vellai, Krisztina TI The Function of NM23-H1/NME1 and Its Homologs in Major Processes Linked to Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE NDPK; Metastasis inhibitor; NM23; Phagocytosis; Apoptosis; Cell migration; Dynamin; Phosphohistidine; Cftr ID NDPK; Metastasis inhibitor; NM23; Phagocytosis; Apoptosis; Cell migration; Dynamin; Phosphohistidine; Cftr AB Metastasis suppressor genes (MSGs) inhibit different biological processes during metastatic progression without globally influencing development of the primary tumor. The first MSG, NM23 (non-metastatic clone 23, isoform H1) or now called NME1 (stands for nonmetastatic) was identified some decades ago. Since then, ten human NM23 paralogs forming two groups have been discovered. Group INM23 genes encode enzymes with evolutionarily highly conserved nucleoside diphosphate kinase (NDPK) activity. In this review we summarize how results from NDPKs in model organisms converged on human NM23 studies. Next, we examine the role of NM23-H1 and its homologs within the metastatic cascade, e.g. cell migration and invasion, proliferation and apoptosis. NM23-H1 homologs are well known inhibitors of cell migration. Drosophila studies revealed that AWD, the fly counterpart of NM23-H1 is a negative regulator of cell motility by modulating endocytosis of chemotactic receptors on the surface of migrating cells in cooperation with Shibire/ Dynamin; this mechanism has been recently confirmed by human studies. NM23-H1 inhibits proliferation of tumor cells by phosphorylating the MAPK scaffold, kinase suppressor of Ras (KSR), resulting in suppression of MAPK signaling. This mechanism was also observed with the C. elegans homolog, NDK-1, albeit with an inverse effect on MAPK activation. Both NM23-H1 and NDK-1 promote apoptotic cell death. In addition, NDK-1, NM23-H1 and their mouse counterpart NM23-M1 were shown to promote phagocytosis in an evolutionarily conserved manner. In summary, inhibition of cell migration and proliferation, alongside actions in apoptosis and phagocytosis are all mechanisms through which NM23-H1 acts against metastatic progression. C1 [Matyasi, Barbara] Eotvos Lorand University, Institute of Biology, Department of Biological Anthropology, Pazmany Peter stny. 1/C, H-1117 Budapest, Hungary. [Farkas, Zsolt] Eotvos Lorand University, Institute of Biology, Department of Biological Anthropology, Pazmany Peter stny. 1/C, H-1117 Budapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Boissan, Mathieu] Sorbonne Universite, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France. [Mehta, Anil] Ninewells Hospital Medical School, Centre for CVS and Lung Biology, DD19SY Dundee, UK. [Takacs-Vellai, Krisztina] Eotvos Lorand University, Institute of Biology, Department of Biological Anthropology, Pazmany Peter stny. 1/C, H-1117 Budapest, Hungary. RP Takacs-Vellai, K (reprint author), Eotvos Lorand University, Institute of Biology, Department of Biological Anthropology, H-1117 Budapest, Hungary. EM krisztina.takacs@ttk.elte.hu CR Stafford LJ, Vaidya KS,Welch DR, 2008)Metastasis suppressors genes in cancer. Int J Biochem Cell Biol 40:874–891. https://doi. org/10.1016/j.biocel.2007.12.016 Khan I, Steeg PS, 2018, Metastasis suppressors: functional pathways. Lab Invest 98:198–210. , DOI 10.1038/labinvest. 2017.104 Steeg PS, Bevilacqua G, Kopper L et al, 1988, Evidence for a novel gene associated with low tumor metastatic potential. J Natl Cancer Inst 80:200–204. , DOI 10.1093/jnci/80.3.200 Fiore LS, Ganguly SS, Sledziona J et al, 2014, c-Abl and Arg induce cathepsin-mediated lysosomal degradation of the NM23- H1 metastasis suppressor in invasive cancer. Oncogene 33:4508– 4520. , DOI 10.1038/onc.2013.399 Pal S, Vishwanath SN, Erdjument-Bromage H et al, 2004, Human SWI/SNF-associated PRMT5 methylates histone H3 arginine 8 and negatively regulates expression of ST7 andNM23 tumor suppressor genes. Mol Cell Biol 24:9630–9645. , DOI 10. 1128/MCB.24.21.9630-9645.2004 Hartsough MT, Clare SE, Mair M et al, 2001, Elevation of breast carcinoma Nm23-H1 metastasis suppressor gene expression and reduced motility by DNA methylation inhibition. Cancer Res 61: 2320–2327 Chen W, Xiong S, Li J et al, 2015, The ubiquitin E3 ligase SCFFBXO24 recognizes deacetylated nucleoside diphosphate kinase A to enhance its degradation. Mol Cell Biol 35:1001–1013. , DOI 10.1128/MCB.01185-14 AlmeidaMI, Nicoloso MS, Zeng L et al, 2012, Strand-specific miR- 28-5p and miR-28-3p have distinct effects in colorectal cancer cells. Gastroenterology 142. , DOI 10.1053/j.gastro.2011.12.047 Boissan M, Dabernat S, Peuchant E et al, 2009, The mammalian Nm23/NDPK family: from metastasis control to cilia movement. Mol Cell Biochem 329:51–62. , DOI 10.1007/s11010- 009-0120-7 Desvignes T, Pontarotti P, Fauvel C, Bobe J, 2009, Nme protein family evolutionary history, a vertebrate perspective. BMC Evol Biol 9:256. , DOI 10.1186/1471-2148-9-256 Bevilacqua G, Sobel ME, Liotta LA, Steeg PS, 1989, Association of low nm23 RNA levels in human primary infiltrating ductal breast carcinomas with lymph node involvement and other histopathological indicators of high metastatic potential. Cancer Res 49:5185–5190 Florenes VA, Aamdal S,Myklebost O et al, 1992, Levels of nm23 messenger RNA in metastatic malignant melanomas: inverse correlation to disease progression. Cancer Res 52:6088–6091 Xerri L, Grob JJ, Battyani Z et al, 1994, NM23 expression in metastasis of malignant melanoma is a predictive prognostic parameter correlated with survival. Br J Cancer 70:1224–1228. , DOI 10.1038/bjc.1994.477 Boissan M, Lacombe M-L, 2006, Nm23/NDP kinases in hepatocellular carcinoma. J Bioenerg Biomembr 38:169–175. https://doi. org/10.1007/s10863-006-9031-4 An R, Meng J, Shi Q et al, 2010, Expressions of nucleoside diphosphate kinase, nm23, in tumor tissues are related with metastasis and length of survival of patients with hepatocellular carcinoma. Biomed Environ Sci 23:267–272. , DOI 10. 1016/S0895-3988(10)60062-1 Hartsough MT, Steeg PS, 2000, Nm23/nucleoside diphosphate kinase in human cancers. J Bioenerg Biomembr 32:301–308. , DOI 10.1023/a:1005597231776 Lacombe M-L, Boissan M, 2013, NME1, NME/NM23 nucleoside diphosphate kinase 1). Atlas Genet Cytogenet Oncol Haematol 17:526–538 Liu L, Li M, Zhang C et al, 2018, Prognostic value and clinicopathologic significance of nm23 in various cancers: A systematic review and meta-analysis. Int J Surg 60:257–265. , DOI 10.1016/j.ijsu.2018.10.035 Leonard MK,McCorkle JR, Snyder DE et al, 2018, Identification of a gene expression signature associated with the metastasis suppressor function of NME1: Prognostic value in human melanoma. Lab Investig 98:327–338. , DOI 10.1038/labinvest.2017.108 Howlett AR, Petersen OW, Steeg PS, Bissell MJ, 1994, A novel function for the nm23-H1 gene: overexpression in human breast carcinoma cells leads to the formation of basement membrane and growth arrest. J Natl Cancer Inst 86:1838–1844. , DOI 10. 1093/jnci/86.24.1838 Harlozinska A, Bar JK, Gerber J, 1996, nm23 expression in tissue sections and tumor effusion cells of ovarian neoplasms. Int J Cancer 69:415–9. , DOI 10.1002/(SICI)1097- 0215(19961021)69:5<415::AID-IJC11>3.0.CO;2-1 Niitsu N, Nakamine H, Okamoto M et al, 2003, Expression of nm23-H1 is associated with poor prognosis in peripheral T-cell lymphoma. Br J Haematol 123:621–630. , DOI 10.1046/ j.1365-2141.2003.04668.x Andolfo I, De Martino D, Liguori L et al, 2011, Correlation of NM23-H1 cytoplasmic expression with metastatic stage in human prostate cancer tissue. Naunyn Schmiedebergs Arch Pharmacol 384:489–498. , DOI 10.1007/s00210-011-0645-7 Chang CL, Zhu XX, Thoraval DH et al, 1994, Nm23-H1 mutation in neuroblastoma. Nature 370:335–336. , DOI 10. 1038/370335a0 Garcia I, Mayol G, Rios J et al, 2012, AThree-Gene Expression Signature Model for Risk Stratification of Patients with Neuroblastoma. Clin Cancer Res 18:2012–2023. , DOI 10.1158/1078-0432.CCR-11-2483 Tan C-Y, Chang CL, 2018, NDPKA is not just a metastasis suppressor - be aware of its metastasis-promoting role in neuroblastoma. Lab Invest 98:219–227. , DOI 10.1038/labinvest. 2017.105 Kapitanovic S, Cacev T, Berkovic M et al, 2004, nm23-H1 expression and loss of heterozygosity in colon adenocarcinoma. J Clin Pathol 57:1312–1318. , DOI 10.1136/jcp.2004. 017954 Boissan M, De Wever O, Lizarraga F et al, 2010, Implication of metastasis suppressor NM23-H1 in maintaining adherens junctions and limiting the invasive potential of human cancer cells. Cancer Res 70:7710–7722. , DOI 10.1158/0008-5472.CAN-10- 1887 Marino N, Marshall J-C, Steeg PS, 2011, Protein-protein interactions: a mechanism regulating the anti-metastatic properties of Nm23-H1. Naunyn Schmiedebergs Arch Pharmacol 384:351– 362. , DOI 10.1007/s00210-011-0646-6 Lee M-Y, JeongW-J, Oh J-W, Choi K-Y, 2009)NM23H2 inhibits EGF- and Ras-induced proliferation of NIH3T3 cells by blocking the ERK pathway. Cancer Lett 275:221–226. , DOI 10. 1016/j.canlet.2008.10.018 Mochizuki T, Bilitou A, Waters CT et al, 2009, Xenopus NM23- X4 regulates retinal gliogenesis through interaction with p27Xic1. Neural Dev 4:1. , DOI 10.1186/1749-8104-4-1 Postel EH, Berberich SJ, Flint SJ, Ferrone CA, 1993, Human cmyc transcription factor PuF identified as nm23-H2 nucleoside diphosphate kinase, a candidate suppressor of tumor metastasis. Science 261:478–480. , DOI 10.1126/science.8392752 Thakur RK, Kumar P, HalderK et al, 2009, Metastases suppressor NM23-H2 interaction with G-quadruplex DNA within c-MYC promoter nuclease hypersensitive element induces c-MYC expression. Nucleic Acids Res 37:172–183. , DOI 10.1093/nar/ gkn919 Fan Z, Beresford PJ, Oh DY et al, 2003, Tumor suppressor NM23-H1 is a granzyme A-activated DNase during CTLmediated apoptosis, and the nucleosome assembly protein SET is its inhibitor. Cell 112:659–672. , DOI 10.1016/s0092- 8674(03)00150-8 Klumpp S, Krieglstein J, 2009, Reversible phosphorylation of histidine residues in proteins from vertebrates. Sci Signal 2:pe13. , DOI 10.1126/scisignal.261pe13 Khan I, Steeg PS, 2018, The relationship of NM23, NME, metastasis suppressor histidine phosphorylation to its nucleoside diphosphate kinase, histidine protein kinase and motility suppression activities. Oncotarget 9:10185–10202. , DOI 10. 18632/oncotarget.23796 Hartsough MT, Morrison DK, Salerno M et al, 2002, Nm23-H1 Metastasis Suppressor Phosphorylation of Kinase Suppressor of Ras via a Histidine Protein Kinase Pathway. J Biol Chem 277: 32389–32399. , DOI 10.1074/jbc.M203115200 Adam K, Hunter T, 2018, Histidine kinases and the missing phosphoproteome from prokaryotes to eukaryotes. Lab Invest 98:233–247. , DOI 10.1038/labinvest.2017.118 Ma D, Xing Z, Liu B et al, 2002, NM23-H1 and NM23-H2 repress transcriptional activities of nuclease-hypersensitive elements in the platelet-derived growth factor-A promoter. J Biol Chem 277:1560–1567. , DOI 10.1074/jbc.M108359200 Zhang Q,McCorkle JR, NovakMet al, 2011)Metastasis suppressor function of NM23-H1 requires its 3’-5’ exonuclease activity. Int J cancer 128:40–50. , DOI 10.1002/ijc.25307 Tokarska-Schlattner M, Boissan M, Munier A et al, 2008, The nucleoside diphosphate kinase D, NM23-H4, binds the inner mitochondrial membrane with high affinity to cardiolipin and couples nucleotide transfer with respiration. J Biol Chem 283:26198– 26207. , DOI 10.1074/jbc.M803132200 Boissan M, Schlattner U, Lacombe ML, 2018, The NDPK/NME superfamily: State of the art. Lab Investig 98:164–174 Annesley SJ, Bago R, Bosnar MH et al, 2011, Dictyostelium discoideum nucleoside diphosphate kinase C plays a negative regulatory role in phagocytosis, macropinocytosis and exocytosis. PLoS One 6:e26024. , DOI 10.1371/journal.pone. 0026024 Kunzelmann K, Mehta A, 2013, CFTR: a hub for kinases and crosstalk of cAMP and Ca2+. FEBS J 280:4417–4429. https:// doi.org/10.1111/febs.12457 Madsen RR, Vanhaesebroeck B, 2020, Cracking the contextspecific PI3K signaling code. Sci Signal 13. , DOI 10. 1126/scisignal.aay2940 Takacs-Vellai K, Vellai T, Farkas Z, Mehta A, 2015, Nucleoside diphosphate kinases, NDPKs, in animal development. Cell Mol Life Sci 72:1447–1462. , DOI 10.1007/s00018-014-1803-0 Hunter P, 2008, The paradox of model organisms. The use of model organisms in research will continue despite their shortcomings. EMBO Rep 9:717–720. , DOI 10.1038/embor.2008.142 Bilitou A, Watson J, Gartner A, Ohnuma S, 2009, The NM23 family in development. Mol Cell Biochem 329:17–33. https:// doi.org/10.1007/s11010-009-0121-6 Sturtevant AH, 1956, A Highly Specific Complementary Lethal System in Drosophila Melanogaster. Genetics 41:118–123 Biggs J, Tripoulas N, Hersperger E et al, 1988, Analysis of the lethal interaction between the prune and Killer of prune mutations of Drosophila. Genes Dev 2:1333–1343. , DOI 10.1101/ gad.2.10.1333 Dearolf CR, Hersperger E, Shearn A, 1988, Developmental consequences of awdb3, a cell-autonomous lethal mutation of Drosophila induced by hybrid dysgenesis. Dev Biol 129:159– 168. , DOI 10.1016/0012-1606(88)90170-4 Rosengard AM, Krutzsch HC, Shearn A et al, 1989, Reduced Nm23/Awd protein in tumour metastasis and aberrant Drosophila development. Nature 342:177–180. , DOI 10. 1038/342177a0 Biggs J, Hersperger E, Steeg PS et al, 1990, A Drosophila gene that is homologous to a mammalian gene associated with tumor metastasis codes for a nucleoside diphosphate kinase. Cell 63: 933–940. , DOI 10.1016/0092-8674(90)90496-2 Wallet V,Mutzel R, Troll H et al, 1990, Dictyostelium nucleoside diphosphate kinase highly homologous to Nm23 and Awd proteins involved in mammalian tumor metastasis and Drosophila development. J Natl Cancer Inst 82:1199–1202. , DOI 10.1093/jnci/82.14.1199 Xu J, Liu LZ, Deng XF et al, 1996, The enzymatic activity of Drosophila AWD/NDP kinase is necessary but not sufficient for its biological function. Dev Biol 177:544–557. , DOI 10. 1006/dbio.1996.0184 Masse K, Dabernat S, Bourbon P-M et al, 2002, Characterization of the nm23-M2, nm23-M3 and nm23-M4 mouse genes: comparison with their human orthologs. Gene 296:87–97. , DOI 10.1016/s0378-1119(02)00836-3 Arnaud-Dabernat S, Bourbon PM, Dierich A et al, 2003, Knockout mice as model systems for studying nm23/NDP kinase gene functions. Application to the nm23-M1 gene. J Bioenerg Biomembr 35:19–30. , DOI 10.1023/a:1023561821551 Di L, Srivastava S, Zhdanova O et al, 2010, Nucleoside diphosphate kinase B knock-outmice have impaired activation of theK+ channel KCa3.1, resulting in defective T cell activation. J Biol Chem 285:38765–38771. , DOI 10.1074/jbc.M110. 168070 Postel EH, Wohlman I, Zou X et al, 2009, Targeted deletion of Nm23/nucleoside diphosphate kinase A and B reveals their requirement for definitive erythropoiesis in the mouse embryo. Dev Dyn 238:775–787. , DOI 10.1002/dvdy.21887 Vogel P, Read R, Hansen GM et al, 2010, Situs inversus in Dpcd/Poll-/-, Nme7-/-, and Pkd1l1-/- mice. Vet Pathol 47:120– 131. , DOI 10.1177/0300985809353553 Deplagne C, Peuchant E, Moranvillier I et al, 2011, The antimetastatic nm23-1 gene is needed for the final step of mammary duct maturation of the mouse nipple. PLoS One 6:e18645. https:// doi.org/10.1371/journal.pone.0018645 Srivastava S, Li Z, Ko K et al, 2006, Histidine phosphorylation of the potassium channel KCa3.1 by nucleoside diphosphate kinase B is required for activation of KCa3.1 and CD4 T cells. Mol Cell 24:665–675. , DOI 10.1016/j.molcel.2006.11.012 Vogel P, Hansen G, Fontenot G, Read R, 2010, Tubulin tyrosine ligase-like 1 deficiency results in chronic rhinosinusitis and abnormal development of spermatid flagella in mice. Vet Pathol 47: 703–712. , DOI 10.1177/0300985810363485 Vogel P, Read RW, Hansen GM et al, 2012, Congenital hydrocephalus in genetically engineered mice. Vet Pathol 49:166–181. , DOI 10.1177/0300985811415708 Farkas Z, Fancsalszky L, Saskoi E et al, 2018, The dosagedependent effect exerted by the NM23-H1/H2 homolog NDK-1 on distal tip cell migration in C. elegans. Lab Investig 98:182– 189. , DOI 10.1038/labinvest.2017.99 Boissan M,Wendum D, Arnaud-Dabernat S et al, 2005, Increased lung metastasis in transgenic NM23-Null/SV40 mice with hepatocellular carcinoma. J Natl Cancer Inst 97:836–845. https://doi. org/10.1093/jnci/dji143 Liu F, Zhang Y, Zhang X-Y, Chen H-L, 2002, Transfection of the nm23-H1 gene into human hepatocarcinoma cell line inhibits the expression of sialyl Lewis X, alpha1,3 fucosyltransferase VII, and metastatic potential. J Cancer Res Clin Oncol 128:189–196. , DOI 10.1007/s00432-001-0314-1 Baba H, Urano T, Okada K et al, 1995, Two isotypes of murine nm23/nucleoside diphosphate kinase, nm23-M1 and nm23-M2, are involved in metastatic suppression of a murine melanoma line. Cancer Res 55:1977–1981 Parhar RS, Shi Y, ZouMet al, 1995, Effects of cytokine-mediated modulation of nm23 expression on the invasion and metastatic behavior of B16F10 melanoma cells. Int J cancer 60:204–210. , DOI 10.1002/ijc.2910600213 Leone A, Flatow U, VanHoutte K, Steeg PS, 1993, Transfection of human nm23-H1 into the human MDA-MB-435 breast carcinoma cell line: effects on tumor metastatic potential, colonization and enzymatic activity. Oncogene 8:2325–2333 Otsuki Y, Tanaka M, Yoshii S et al, 2001, Tumor metastasis suppressor nm23H1 regulates Rac1 GTPase by interaction with Tiam1. Proc Natl Acad Sci U S A 98:4385–4390. , DOI 10.1073/pnas.071411598 MurakamiM,Meneses PI, Knight JS et al, 2008, Nm23-H1 modulates the activity of the guanine exchange factor Dbl-1. Int J cancer 123:500–510. , DOI 10.1002/ijc.23568 Horak CE, Lee JH, Elkahloun AG et al, 2007, Nm23-H1 suppresses tumor cell motil i ty by down-regulat ing the lysophosphatidic acid receptor EDG2. Cancer Res 67:7238– 7246. , DOI 10.1158/0008-5472.CAN-07-0962 Okabe-Kado J, Hagiwara-Watanabe Y, Niitsu N et al, 2018, NM23 downregulation and lysophosphatidic acid receptor EDG2/lpa1 upregulation during myeloid differentiation of human leukemia cells. Leuk Res 66:39–48. , DOI 10.1016/j. leukres.2018.01.003 Marino N, Marshall J-C, Collins JWet al, 2013, Nm23-H1 Binds to Gelsolin and Inactivates ItsActin-Severing Capacity to Promote Tumor Cell Motility and Metastasis. Cancer Res 73:5949–5962. , DOI 10.1158/0008-5472.CAN-13-0368 D’Angelo A, Garzia L, Andre A et al, 2004, Prune cAMP phosphodiesterase binds nm23-H1 and promotes cancer metastasis. Cancer Cell 5:137–149. , DOI 10.1016/s1535-6108(04, 00021-2 Garzia L, D’Angelo A, Amoresano A et al, 2008, Phosphorylation of nm23-H1 by CKI induces its complex formation with h-prune and promotes cell motility. Oncogene 27:1853– 1864. , DOI 10.1038/sj.onc.1210822 Suzuki E, Ota T, Tsukuda K et al, 2004, nm23-H1 reduces in vitro cell migration and the liver metastatic potential of colon cancer cells by regulating myosin light chain phosphorylation. Int J cancer 108:207–211. , DOI 10.1002/ijc.11546 Zhao R, Gong L, Li L et al, 2013, Nm23-H1 is a negative regulator of TGF-Β1-dependent induction of epithelial-mesenchymal transition. Exp Cell Res 319:740–749. , DOI 10.1016/j. yexcr.2012.10.013 AffolterM, Caussinus E, Emoto H et al, 2008, Tracheal branching morphogenesis in Drosophila: new insights into cell behaviour and organ architecture. Development 135:2055–2064. https:// doi.org/10.1242/dev.014498 Schottenfeld J, Song Y, Ghabrial AS, 2010, Tube continued: morphogenesis of the Drosophila tracheal system. Curr Opin Cell Biol 22:633–639. , DOI 10.1016/j.ceb.2010.07.016 Dammai V, Adryan B, Lavenburg KR, Hsu T, 2003, Drosophila awd, the homolog of human nm23, regulates FGF receptor levels and functions synergistically with shi/dynamin during tracheal development. Genes Dev 17:2812–2824. , DOI 10.1101/ gad.1096903 Montell DJ, YoonWH, Starz-Gaiano M(2012, Group choreography: mechanisms orchestrating the collective movement of border cells. Nat Rev Mol Cell Biol 13:631–645. , DOI 10.1038/ nrm3433 Nallamothu G,Woolworth JA, Dammai V, Hsu T, 2008, Awd, the homolog of metastasis suppressor gene Nm23, regulates Drosophila epithelial cell invasion. Mol Cell Biol 28:1964– 1973. , DOI 10.1128/MCB.01743-07 Duchek P, Somogyi K, Jekely G et al, 2001, Guidance of cell migration by the Drosophila PDGF/VEGF receptor. Cell 107: 17–26. , DOI 10.1016/s0092-8674(01)00502-5 McDonald JA, Pinheiro EM, Montell DJ, 2003, PVF1, a PDGF/VEGF homolog, is sufficient to guide border cells and interacts genetically with Taiman. Development 130:3469–3478. , DOI 10.1242/dev.00574 Silver DL, Geisbrecht ER, Montell DJ, 2005, Requirement for JAK/STAT signaling throughout border cell migration in Drosophila. Development 132:3483–3492. , DOI 10. 1242/dev.01910 KrishnanKS, Rikhy R, Rao S et al, 2001, Nucleoside diphosphate kinase, a source of GTP, is required for dynamin-dependent synaptic vesicle recycling. Neuron 30:197–210. , DOI 10. 1016/s0896-6273(01)00273-2 Ferguson SM, De Camilli P, 2012, Dynamin, a membraneremodelling GTPase. Nat Rev Mol Cell Biol 13:75–88. https:// doi.org/10.1038/nrm3266 Boissan M, Montagnac G, Shen Q et al, 2014, Membrane trafficking. Nucleoside diphosphate kinases fuel dynamin superfamily proteins with GTP for membrane remodeling. Science 344: 1510–1515. , DOI 10.1126/science.1253768 Khan I, Gril B, Steeg PS, 2019, Metastasis Suppressors NME1 and NME2 Promote Dynamin 2 Oligomerization and Regulate Tumor Cell Endocytosis, Motility and Metastasis. Cancer Res canres.0492.2019. , DOI 10.1158/0008-5472.CAN-19- 0492 McKayMM,Morrison DK, 2007, Integrating signals from RTKs to ERK/MAPK. Oncogene 26:3113–3121. , DOI 10. 1038/sj.onc.1210394 Karnoub AE, Weinberg R, 2008, Ras oncogenes: split personalities. Nat Rev Mol Cell Biol 9:517–531. , DOI 10.1038/ nrm2438 Morrison DK, 2001, KSR: aMAPK scaffold of the Ras pathway? J Cell Sci 114:1609–1612 Roy F, Laberge G, Douziech M et al, 2002, KSR is a scaffold required for activation of the ERK/MAPKmodule.Genes Dev 16: 427–438. , DOI 10.1101/gad.962902 Alexa A, Varga J, Remenyi A, 2010, Scaffolds are “active” regulators of signaling modules. FEBS J 277:4376–4382. https://doi. org/10.1111/j.1742-4658.2010.07867.x Muller J, Ory S, Copeland T et al, 2001, C-TAK1 regulates Ras signaling by phosphorylating the MAPK scaffold, KSR1. Mol Cell 8:983–993. , DOI 10.1016/s1097-2765(01)00383-5 Ory S, Zhou M, Conrads TP et al, 2003, Protein phosphatase 2A positively regulates Ras signaling by dephosphorylating KSR1 and Raf-1 on critical 14-3-3 binding sites. Curr Biol 13:1356– 1364. , DOI 10.1016/s0960-9822(03)00535-9 Masoudi N, Fancsalszky L, Pourkarimi E et al, 2013, The NM23- H1/H2 homolog NDK-1 is required for full activation of Ras signaling in C. elegans. Development 140:3486–3495. https:// doi.org/10.1242/dev.094011 Sundaram MV, 2013, Canonical RTK-Ras-ERK signaling and related alternative pathways. WormBook 1–38. , DOI 10.1895/wormbook.1.80.2 Sternberg PW, 2005, Vulval development. WormBook 1–28. , DOI 10.1895/wormbook.1.6.1 Eisenmann DM, Kim SK, 2000, Protruding vulva mutants identify novel loci and Wnt signaling factors that function during Caenorhabditis elegans vulva development. Genetics 156:1097– 1116 OhmachiM, Rocheleau CE, Church D et al, 2002, C. elegans ksr- 1 and ksr-2 have both unique and redundant functions and are required for MPK-1 ERK phosphorylation. Curr Biol 12:427– 433. , DOI 10.1016/s0960-9822(02)00690-5 Salerno M, Palmieri D, Bouadis A et al, 2005, Nm23-H1 metastasis suppressor expression level influences the binding properties, stability, and function of the kinase suppressor of Ras1, KSR1, Erk scaffold in breast carcinoma cells. Mol Cell Biol 25:1379– 1388. , DOI 10.1128/MCB.25.4.1379-1388.2005 Aguirre-Ghiso JA, 2007, Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer 7:834–846. https:// doi.org/10.1038/nrc2256 Holmgren L, O’reilly MS, Folkman J, 1995, Dormancy of micrometastases: Balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1:149–153. https:// doi.org/10.1038/nm0295-149 Horak CE, Lee JH,Marshall JC et al, 2008, The role ofmetastasis suppressor genes in metastatic dormancy. Apmis 116:586–601. , DOI 10.1111/j.1600-0463.2008.01213.x Aguirre-Ghiso JA, Estrada Y, Liu D, Ossowski L, 2003, ERKMAPK activity as a determinant of tumor growth and dormancy; regulation by p38SAPK. Cancer Res 63:1684–1695. , DOI 10.1016/j.urolonc.2003.12.012 Marshall J-CA, Collins JW, Nakayama J et al, 2012, Effect of inhibition of the lysophosphatidic acid receptor 1 on metastasis and metastatic dormancy in breast cancer. J Natl Cancer Inst 104:1306–1319. , DOI 10.1093/jnci/djs319 Marino N, Nakayama J, Collins JW, Steeg PS, 2012, Insights into the biology and prevention of tumor metastasis provided by the Nm23 metastasis suppressor gene. Cancer Metastasis Rev 31: 593–603. , DOI 10.1007/s10555-012-9374-8 Hanahan D,Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674. , DOI 10.1016/j.cell.2011.02.013 Adams JM, Cory S, 2007, The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene 26:1324–1337. https://doi. org/10.1038/sj.onc.1210220 Lowe SW, Cepero E, Evan G, 2004, Intrinsic tumour suppression. Nature 432:307–315. , DOI 10.1038/nature03098 Tran AT, Chapman EM, FlamandMNet al, 2019)MiR-35 buffers apoptosis thresholds in the C. elegans germline by antagonizing both MAPK and core apoptosis pathways. Cell Death Differ. , DOI 10.1038/s41418-019-0325-6 Rutkowski R, Dickinson R, Stewart G et al, 2011, Regulation of Caenorhabditis elegans p53/CEP-1-dependent germ cell apoptosis by Ras/MAPK signaling. PLoSGenet 7:e1002238. , DOI 10.1371/journal.pgen.1002238 Perrin AJ, Gunda M, Yu B et al, 2013, Noncanonical control of C. elegans germline apoptosis by the insulin/IGF-1 and Ras/MAPK signaling pathways. Cell Death Differ 20:97–107. , DOI 10.1038/cdd.2012.101 Chowdhury D, Beresford PJ, Zhu P et al, 2006, The Exonuclease TREX1 Is in the SET Complex and Acts in Concert with NM23-H1 to Degrade DNA during Granzyme A-Mediated Cell Death. Mol Cell 23:133–142. , DOI 10.1016/j.molcel.2006.06.005 Lu N, Zhou Z, 2012, Membrane trafficking and phagosome maturation during the clearance of apoptotic cells. Int Rev Cell Mol Biol 293:269–309. , DOI 10.1016/B978-0-12-394304-0. 00013-0 Poon IKH, Lucas CD, Rossi AG, Ravichandran KS, 2014, Apoptotic cell clearance: basic biology and therapeutic potential. Nat Rev Immunol 14:166–180. , DOI 10.1038/nri3607 Coussens LM, Werb Z, 2002, Inflammation and cancer. Nature 420:860–867. , DOI 10.1038/nature01322.Inflammation Fancsalszky L, Monostori E, Farkas Z et al, 2014, NDK-1, the homolog of NM23-H1/H2 regulates cell migration and apoptotic engulfment in C. elegans. PLoS One 9:e92687. , DOI 10. 1371/journal.pone.0092687 Farkas Z, PetricM, Liu X et al, 2019, The nucleoside diphosphate kinase NDK-1/NME1 promotes phagocytosis in concert with DYN-1/Dynamin. FASEB J fj201900220R. , DOI 10. 1096/fj.201900220R De Palma M, Hanahan D, 2012, The biology of personalized cancer medicine: facing individual complexities underlying hallmark capabilities. Mol Oncol 6:111–127. , DOI 10.1016/ j.molonc.2012.01.011 Martinez J, Almendinger J, Oberst A et al, 2011, Microtubuleassociated protein 1 light chain 3 alpha, LC3)-associated phagocytosis is required for the efficient clearance of dead cells. Proc Natl Acad Sci U S A 108:17396–17401. , DOI 10.1073/ pnas.1113421108 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 49 EP 61 DI 10.1007/s12253-020-00797-0 PG 13 ER PT J AU Deli, T Orosz, M Jakab, A AF Deli, Tamas Orosz, Monika Jakab, Attila TI Hormone Replacement Therapy in Cancer Survivors – Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Hormone replacement therapy; HRT; Menopausal hormone therapy; MHT; Estrogen replacement; Estrogen-progestin replacement; Oncologic patient; Cancer survivor; Gynecologic cancer; Non-gynecologic cancer; Breast cancer; Recurrence risk ID Hormone replacement therapy; HRT; Menopausal hormone therapy; MHT; Estrogen replacement; Estrogen-progestin replacement; Oncologic patient; Cancer survivor; Gynecologic cancer; Non-gynecologic cancer; Breast cancer; Recurrence risk AB Rapid advance in oncology leads to increasing survival of oncologic patients. More and more of them live long enough to reach either the natural age of menopause or, as a side effect of their oncotherapy, experience the cessation of gonadal function, leading to premature ovarian insufficiency, with disturbing vasomotor symptoms and long-term negative cardiovascular and skeletal effects. Thus, an ever increasing number of cancer survivors search endocrinologic help in the form of hormone replacement therapy (HRT). The misinterpretation of the WHI (Women's Health Initiative) Study has lead to an irrational fear of female hormone replacement, both by the general population and medical professionals. It has seemed the logical and safe conclusion to many physicians to avoid HRT, supposing that this attitude definitely causes no harm, whereas the decision of prescribing estrogen alone or with progestins might bear oncologic and thromboembolic risks and may even lead to litigation in case of a potentially related complication. However, it was known even before the WHI results that premature menopause and hypogonadism decreases the life expectancy of women by years through its skeletal and cardiovascular effects, and this negative effect correlates with the length of the hypoestrogenaemic period. Therefore, the denial of HRT also needs to be supported by evidence and should be weighed against the risks of HRT. Yet, the oncologic risk of HRT is extremely difficult to assess. In this work we review the latest evidence from in vitro experiments to clinical studies, regarding HRT in survivors of gynecologic and non-gynecologic cancers. Based on our literature research, we group tumours regarding the oncologic risk of properly chosen female hormone replacement therapy in cancer survivors as follows: ’HRT is advantageous’ (e.g. endometrial cancer type I, cervical adenocarcinoma, haematologic malignancies, local cutaneous malignant melanoma, colorectal cancer, hepatocellular cancer); ’HRT is neutral’ (e.g. BRCA 1/2mutation carriers without cancer, endometrial cancer type II, uterinal carcinosarcoma and adenosarcoma, certain types of ovarian cancer, cervical, vaginal and vulvar squamous cell carcinoma, prolactinoma, kidney cancer, pancreatic cancer, thyroid cancer); ’HRT is relatively contraindicated’ for various reasons (e.g. leiomyosarcoma, certain types of ovarian tumours, brain tumours, advanced metastatic malignant melanoma, lung cancer, gastric cancer, bladder cancer); ’HRT is disadvantageous and thus contraindicated’ (e.g. breast cancer, endometrial stroma sarcoma, meningioma, glioma, hormone receptor positive gastric and bladder cancer). C1 [Deli, Tamas] University Medical School of Debrecen, Department of Gynecology and ObstetricsDebrecen, Hungary. [Orosz, Monika] University Medical School of Debrecen, Department of Gynecology and ObstetricsDebrecen, Hungary. [Jakab, Attila] University Medical School of Debrecen, Department of Gynecology and ObstetricsDebrecen, Hungary. RP Deli, T (reprint author), University Medical School of Debrecen, Department of Gynecology and Obstetrics, Debrecen, Hungary. EM tamasdeli@yahoo.com CR Baber RJ, Panay N, Fenton A, Apr, IMS Writing Group, 2016, IMS Recommendations on women's midlife health and menopause hormone therapy. Climacteric(2):109–150 Lumsden MA, Davies M, Sarri G, Guideline Development Group for Menopause: Diagnosis and Management, NICE Clinical Guideline No. 23,, 2016, Diagnosis and Management of Menopause: The National Institute of Health and Care Excellence, NICE, Guideline. JAMA Intern Med 176(8):1205– 1206 ACOG Practice Bulletin No. 141: management of menopausal symptoms., 2014, Obstet Gynecol. 123(1):202-16. Ossewaarde ME, Bots ML, Verbeek AL, Peeters PH, van der Graaf Y, Grobbee DE, van der Schouw YT, 2005, Age at menopause, cause-specific mortality and total life expectancy. Epidemiology 16(4):556–562 Prossnitz ER, Barton M, 2014, Estrogen biology: new insights into GPER function and clinical opportunities. Mol Cell Endocrinol 389(1-2):71–83 Prossnitz ER, Hathaway HJ, 2015, What have we learned about GPER function in physiology and disease from knockout mice? J Steroid Biochem Mol Biol 153:114–126 Yamaga R, Ikeda K, Horie-Inoue K, Ouchi Y, Suzuki Y, Inoue S, 2013, RNA sequencing of MCF-7 breast cancer cells identifies novel estrogen-responsive genes with functional estrogen receptor-binding sites in the vicinity of their transcription start sites. Horm Cancer 4(4):222–232 Bourdeau V, Deschenes J, Metivier R, Nagai Y, Nguyen D, Bretschneider N, Gannon F, White JH, Mader S, 2004, Genome-wide identification of high-affinity estrogen response elements in human and mouse. Mol Endocrinol 18(6):1411–1427 Kamalakaran S1, Radhakrishnan SK, Beck WT, 2005, Identification of estrogen-responsive genes using a genomewide analysis of promoter elements for transcription factor binding sites. J Biol Chem 280(22):21491–21497 Sotoca AM, Vervoort J, Rietjens I MCM, Gustafsson JA., 2012, Human ERα and ERβ Splice Variants: Understanding Their Domain Structure in Relation to Their Biological Roles in Breast Cancer Cell Proliferation. Biochemistry, Prof. Deniz Ekinci, Ed.), ISBN: 978-953-51-0076-8, InTech. Xu Z, Liu J, Gu L, Ma X, Huang B, Pan X, 2016, Research progress on the reproductive and non-reproductive endocrine tumors by estrogen-related receptors. J Steroid Biochem Mol Biol 158:22–30 Li M, Chai HF, Peng F, Meng YT, Zhang LZ, Zhang L, Zou H, Liang QL, Li MM, Mao KG, Sun DX, Tong MY, Deng ZQ, Hou ZJ, Zhao Y, Li J, Wang XC, Lv SS, Zhang QQ, Yu X, Lam EW, Liu Q, Cui XN, Xu J, 2018, Estrogen receptor β upregulated by lncRNA-H19 to promote cancer stem-like properties in papillary thyroid carcinoma. Cell Death Dis 9(11):1120 Rubio GA, Catanuto P, Glassberg MK, Lew JI, Elliot SJ, 2018, Estrogen receptor subtype expression and regulation is altered in papillary thyroid cancer after menopause. Surgery 163(1):143– 149 Zhang Y, Wei F, Zhang J, Hao L, Jiang J, Dang L, Mei D, Fan S, Yu Y, Jiang L, 2017, Bisphenol A and estrogen induce proliferation of human thyroid tumor cells via an estrogen-receptordependent pathway. Arch Biochem Biophys 633:29–39 Eldien MMS, Abdou AG, Rageh T, Abdelrazek E, Elkholy E, 2017, Immunohistochemical expression of ER-α and PRin papillary thyroid carcinoma. Ecancermedicalscience 11:748. https:// doi.org/10.3332/ecancer.2017.748 Pierdominici M, Maselli A, Locatelli SL, Ciarlo L, Careddu G, Patrizio M, Ascione B, Tinari A, Carlo-Stella C, Malorni W, Matarrese P, Ortona E, 2017, Estrogen receptor β ligation inhibits Hodgkin lymphoma growth by inducing autophagy. Oncotarget 8(5):8522–8535 Maia DM1, Sciarrotta J, Abendroth K, Blatt J, 2000, Sex steroid receptors in Hodgkin's disease. Leuk Lymphoma 39(3-4):365– 371 Teske E1, Besselink CM, Blankenstein MA, Rutteman GR, Misdorp W, 1987, The occurrence of estrogen and progestin receptors and anti-estrogen binding sites, AEBS, in canine non- Hodgkin's lymphomas. Anticancer Res 7(4B):857–860 Ladikou EE, Kassi E, 2017, The emerging role of estrogen in B cell malignancies. Leuk Lymphoma 58(3):528–539 Yakimchuk K, Jondal M, Okret S, 2013, Estrogen receptor α and β in the normal immune system and in lymphoid malignancies. Mol Cell Endocrinol 375(1-2):121–129 Mukhopadhyay M, Das C, Kumari M, Sen A, Mukhopadhyay B, Mukhopadhyay B, 2017, Spectrum of meningioma with special reference to prognostic utility of ER,PR and Ki67 expression. J Lab Physicians 9(4):308–313 Hsu DW, Efird JT, Hedley-Whyte ET, 1997, Progesterone and estrogen receptors in meningiomas: prognostic considerations. J Neurosurg 86(1):113–120 Kuroi Y, Matsumoto K, Shibuya M, Kasuya H, 2018, Progesterone Receptor Is Responsible for Benign Biology of Skull Base Meningioma. World Neurosurg 118:e918–e924 Wolfsberger S, Doostkam S, Boecher-Schwarz HG, Roessler K, van Trotsenburg M, Hainfellner JA, Knosp E, 2004, Progesterone-receptor index in meningiomas: correlation with clinico-pathological parameters and review of the literature. Neurosurg Rev 27(4):238–245 Liu M, Zhang K, Zhao Y, Guo Q, Guo D, Zhang J, 2015, Evidence for involvement of steroid receptors and coactivators in neuroepithelial and meningothelial tumors. Tumour Biol 36(5):3251–3261 Jaffrain-Rea ML, Petrangeli E, Ortolani F, Fraioli B, Lise A, Esposito V, Spagnoli LG, Tamburrano G, Frati L, Gulino A, 1996, Cellular receptors for sex steroids in human pituitary adenomas. J Endocrinol 151(2):175–184 Caronti B, Palladini G, Bevilacqua MG, Petrangeli E, Fraioli B, Cantore G, Tamburrano G, Carapella CM, Jaffrain-Rea ML, 1993, Effects of 17 beta-estradiol, progesterone and tamoxifen on in vitro proliferation of human pituitary adenomas: correlation with specific cellular receptors. Tumour Biol 14(1):59–68 Natale CA, Li J, Zhang J, Dahal A, Dentchev T, Stanger BZ, Ridky TW., 2018, Activation of G protein-coupled estrogen receptor signaling inhibits melanoma and improves response to immune checkpoint blockade. Elife. 7. pii: e31770. TianW, PangW, Ge Y, He X, Wang D, Li X, Hou H, Zhou D, Feng S, Chen Z, Yang Y, 2018, Hepatocyte-generated 27- hydroxycholesterol promotes the growth of melanoma by activation of estrogen receptor alpha. J Cell Biochem 119(3):2929–2938 Dika E, Fanti PA, Vaccari S, Capizzi E, Degiovanni A, Gobbi A, Piraccini BM, Ribero S, Baraldi C, Ravaioli GM, Fiorentino M, Patrizi A, 2017, Oestrogen and progesterone receptors in melanoma and nevi: an immunohistochemical study. Eur J Dermatol 27(3):254–259 Bredow L, Stutzel L, Bohringer D,Gundlach E, Reinhard T, Auw- Haedrich C, 2014, Progesterone and estrogen receptors in conjunctival melanoma and nevi. Graefes Arch Clin Exp Ophthalmol 252(2):359–365 Hiramitsu S, Ishikawa T, Lee WR, Khan T, Crumbley C, Khwaja N, Zamanian F, Asghari A, Sen M, Zhang Y, Hawse JR, Minna JD, Umetani M, 2018, Estrogen Receptor Beta-Mediated Modulation of Lung Cancer Cell Proliferation by 27- Hydroxycholesterol. Front Endocrinol, Lausanne, 9:470 Ding X, Li L, Tang C, Meng C, XuW,Wei X, Guo Z, Zhang T, Fu Y, Zhang L, Wang X, Lin L, Liang J, 2018, Cytoplasmic expression of estrogen receptor β may predict poor outcome of EGFRTKI therapy in metastatic lung adenocarcinoma. Oncol Lett 16(2): 2382–2390 Stevanato Filho PR, Aguiar Junior S, Begnami MD, Ferreira FO, Nakagawa WT, Spencer RMSB, Bezerra TS, Boggiss PE, Lopes A, 2018, Estrogen Receptor β as a Prognostic Marker of Tumor Progression in Colorectal Cancer with Familial Adenomatous Polyposis and Sporadic Polyps. Pathol Oncol Res 24(3):533–540 Liu D, 2016, Gene signatures of estrogen and progesterone receptor pathways predict the prognosis of colorectal cancer. FEBS J 283(16):3115–3133 Kuo CT, Lee WS, 2016, Progesterone receptor activation is required for folic acid-induced anti-proliferation in colorectal cancer cell lines. Cancer Lett 378(2):104–110 Ge H, Yan Y, Tian F, Wu D, Huang Y, 2018, Prognostic value of estrogen receptor α and estrogen receptor β in gastric cancer based on a meta-analysis and The Cancer Genome Atlas, TCGA, datasets. Int J Surg 53:24–31 Iyer JK, Kalra M, Kaul A, Payton ME, Kaul R, 2017, Estrogen receptor expression in chronic hepatitis C and hepatocellular carcinoma pathogenesis. World J Gastroenterol 23(37):6802–6816 Tsai HW, Ho CL, Cheng SW, Lin YJ, Chen CC, Cheng PN, Yen CJ, Chang TT, Chiang PM, Chan SH, Ho CH, Chen SH, Wang YW, Chow NH, Lin JC., 2018, Progesterone receptor membrane component 1 as a potential prognostic biomarker for hepatocellular carcinoma. World J Gastroenterol 2018 Mar 14;24(10):1152- 1166. Garg D, Man Ng SS, Baig KM, Driggers P, Segars J, 2017, Progesterone-Mediated Non-Classical Signaling. Trends Endocrinol Metab 28(9):656–668 WetendorfM, DeMayo FJ, 2014, Progesterone receptor signaling in the initiation of pregnancy and preservation of a healthy uterus. Int J Dev Biol 58(2-4):95–106 Mani SK, Oyola MG, 2012, Progesterone signaling mechanisms in brain and behavior. Front Endocrinol, Lausanne, 3:7 American Cancer Society., 2016, Cancer Facts & Figures 2016. Atlanta: American Cancer Society https://www.cancer.org/ content/dam/cancer-org/research/cancer-facts-and-statistics/ annual-cancer-facts-and-figures/2016/cancer-facts-and-figures- 2016.pdf Murthy V, Chamberlain RS, 2012, Menopausal symptoms in young survivors of breast cancer: a growing problem without an ideal solution. Cancer Control 19(4):317–329 Cobleigh MA, Berris RF, Bush T, 1994, Estrogen replacement therapy in breast cancer survivors. A time for change. JAMA 272(7):540–545 Vassilopoulou-Sellin R, Asmar L, Hortobagyi GN, Klein MJ, McNeese M, Singletary SE, Theriault RL, 1999, Estrogen replacement therapy after localized breast cancer: clinical outcome of 319 women followed prospectively. J Clin Oncol 17(5):1482– 1487 Vassilopoulou-Sellin R, Theriault R, Klein MJ, 1997, Estrogen Replacement Therapy in Women with Prior Diagnosis and Treatment for Breast Cancer. Gynecol Oncol 65(1):89–93 O'Meara ES, Rossing MA, Daling JR, Elmore JG, Barlow WE, Weiss NS, 2001, Hormone replacement therapy after a diagnosis of breast cancer in relation to recurrence and mortality. J Natl Cancer Inst 93(10):754–762 Holmberg L, Anderson H, HABITS steering and data monitoring committees, 2004, HABITS, hormonal replacement therapy after breast cancer–is it safe?), a randomised comparison: trial stopped. Lancet 363(9407):453–455 Lupo M, Dains JE, Madsen LT, 2015, Hormone Replacement Therapy: An Increased Risk of Recurrence and Mortality for Breast Cancer Patients? J Adv Pract Oncol 6(4):322–330 Brewster A, Do KA, Thompson PA, Hahn KA, Sahin AA, Cao Y, Stewart MM, Murray JL, Hortobagyi GN, Bondy ML, 2007, Relationship between epidemiologic risk factors and breast cancer recurrence. J Clin Oncol 25(28):4438–4444 Holmberg L, Iversen OE, Rudenstam CM, Hammar M, Kumpulainen E, Jaskiewicz J, Jassem J, Dobaczewska D, Fjosne HE, Peralta O, Arriagada R, Holmqvist M, Maenpaa J, HABITS Study Group, 2008, Increased risk of recurrence after hormone replacement therapy in breast cancer survivors. J Natl Cancer Inst 100(7):475–482 Fahlen M, Fornander T, Johansson H, Johansson U, Rutqvist LE, Wilking N, von Schoultz E, 2013, Hormone replacement therapy after breast cancer: 10 year follow up of the Stockholm randomised trial. Eur J Cancer 49(1):52–59 Cummings SR, Ettinger B, Delmas PD, Kenemans P, Stathopoulos V, Verweij P, Mol-Arts M, Kloosterboer L, Mosca L, Christiansen C, Bilezikian J, Kerzberg EM, Johnson S, Zanchetta J, Grobbee DE, Seifert W, Eastell R, Trial Investigators LIFT, 2008, The Effects of Tibolone in Older Postmenopausal Women. N Engl J Med 359(7):697–708 Kenemans P, Bundred NJ, Foidart JM, Kubista E, von Schoultz B, Sismondi P, Vassilopoulou-Sellin R, Yip CH, Egberts J, Mol-Arts M, Mulder R, van Os S, Beckmann MW, LIBERATE Study Group, 2009, Safety and efficacy of tibolone in breast-cancer patients with vasomotor symptoms: a double-blind, randomised, non-inferiority trial. Lancet Oncol 10(2):135–146 Bundred NJ, Kenemans P, Yip CH, Beckmann MW, Foidart JM, Sismondi P, Schoultz B, Vassilopoulou-Sellin R, Galta RE, Lieshout EV, Mol-Arts M, Planellas J, Kubista E, 2012, Tibolone increases bone mineral density but also relapse in breast cancer survivors: LIBERATE trial bone substudy. Breast Cancer Res 14(1):R13 Sismondi P1, Kimmig R, Kubista E, Biglia N, Egberts J, Mulder R, Planellas J, Moggio G, Mol-Arts M, Kenemans P, 2011, Effects of tibolone on climacteric symptoms and quality of life in breast cancer patients–data from LIBERATE trial. Maturitas 70(4):365–372 Pruthi S1, Simon JA, Early AP, 2011, Current overview of the management of urogenital atrophy in women with breast cancer. Breast J 17(4):403–408 Durna EM1, Wren BG, Heller GZ, Leader LR, Sjoblom P, Eden JA, 2002, Hormone replacement therapy after a diagnosis of breast cancer: Cancer recurrence and mortality. Med J Aust 177(7):347–351 Fahlen M, Fornander T, Johansson H, Johansson U, Rutqvist LE, WilkingM, von Schoultz E, 2013, Hormone replacement therapy after breast cancer: 10 year follow up of the Stockholm randomised trial. Eur J Cancer 49:52–59 Decker DA, Pettinga JE, VanderVelde N, Huang RR, Kestin L, Burdakin JH, 2003, Estrogen replacement therapy in breast cancer survivors: A matched-controlled series. Menopause 10(4):277– 285 DiSaia PJ, Brewster WR, Ziogas A, Anton-Culver H, 2000, Breast cancer survival and hormone replacement therapy: A cohort analysis. Am J Clin Oncol 23(6):541–545 Finch A, Evans G, Narod SA, 2012, BRCA carriers, prophylactic salpingo-oophorectomy and menopause: clinical management considerations and recommendations. Women's Health, Lond, 8(5):543–555 Kotsopoulos J, Gronwald J, Karlan BY, Huzarski T, Tung N, Moller P, Armel S, Lynch HT, Senter L, Eisen A, Singer CF, Foulkes WD, Jacobson MR, Sun P, Lubinski J, Narod SA, Hereditary Breast Cancer Clinical Study Group, 2018, Hormone Replacement Therapy After Oophorectomy and Breast Cancer Risk Among BRCA1 Mutation Carriers. JAMA Oncol 4(8): 1059–1065 Guidozzi F1, Daponte A, 1999, Estrogen replacement therapy for ovarian carcinoma survivors: A randomized controlled trial. Cancer 86(6):1013–1018 Mascarenhas C1, Lambe M, Bellocco R, Bergfeldt K, Riman T, Persson I, Weiderpass E, 2006, Use of hormone replacement therapy before and after ovarian cancer diagnosis and ovarian cancer survival. Int J Cancer 119(12):2907–2915 Eeles RA, Morden JP, Gore M, Mansi J, Glees J, Wenczl M, Williams C, Kitchener H, Osborne R, Guthrie D, Harper P, Bliss JM., 2015, Adjuvant hormone therapy may improve survival in epithelial ovarian cancer: results of the AHT randomized trial. J Clin Oncol 2015 Dec 10;33(35):4138-4144. Pergialiotis V, Pitsouni E, Prodromidou A, Frountzas M, Perrea DN, Vlachos GD, 2016, Hormone therapy for ovarian cancer survivors: systematic review and meta-analysis. Menopause 23(3):335–342 Eeles RA, Tan S, Wiltshaw E, Fryatt I, A’Hern RP, Shepherd JH, Harmer CL, Blake RP, Chilvers CE, 1991, Hormone replacement therapy and survival after surgery for ovarian. BMJ 302(6771): 259–262 Ursic-Vrscaj M, Bebar S, Zakelj MP, 2001, Hormone replacement therapy after invasive ovarian serous cystadeno carcinoma treatment: the effect on survival. Menopause 8(1):70–75 Besevic J, Gunter MJ, Fortner RT, Tsilidis KS, Weiderpass E, Onland-Moret NE, Dossus L, Tjonneland A, Hansen L, Overvad K, Mesrine S, Baglietto L, Clavel-Chapelon F, Kaaks R, Aleksandrova K, Boeing H, Trichopoulou A, Lagiou P, Bamia C, Masala G, Agnoli C, Tumino R, Ricceri F, Panico S, HB, as, Bueno-de-Mesquita, Peeters PH, Jareid M, Quiros JR, Duell EJ, Sanchez MJ, Larranaga N, Chirlaque MD, Barricarte A, Dias JA, Sonestedt E, Idahl A, Lundin E, Wareham NJ, Khaw KT, Travis RC, Rinaldi S, Romieu I, Riboli E, Merritt MA., 2015, Reproductive factors and epithelial ovarian cancer survival in the EPIC cohort study. Br J Cancer 113(11):1622-1631. Pergialiotis V, Pitsouni E, Prodromidou A, Frountzas M, Perrea DN, Vlachos GD, 2016, Hormone therapy for ovarian cancer survivors: systematic reviewand meta-analysis. Menopause 23(3):335–342 Kuhle CL, Kapoor E, Sood R, Thielen JM, Jatoi A, Faubion SS, 2016, Menopausal hormone therapy in cancer survivors: A narrative review of the literature. Maturitas 92:86–96 Biglia N, Bounous VE, Sgro LG, D'Alonzo M, Gallo M, 2015, Treatment of climacteric symptoms in survivors of gynaecological cancer. Maturitas 82(3):296–298 Del Carmen MG, Rice LW, 2017, Management of menopausal symptoms in women with gynecologic cancers. Gynecol Oncol 146(2):427–435 Singh P, Oehler MK., 2010, Hormone replacement after gynaecological cancer. Maturitas 2010 Mar;65(3):190-197. Creasman WT, Henderson D, Hinshaw W, Clarke-Pearson DL, 1986, Estrogen replacement therapy in the patient treated for endometrial cancer. Obstet Gynecol 67(3):326–330 Lee RB, Burke TW, Park RC, 1990, Estrogen replacement therapy following treatment for stage I endometrial carcinoma. Gynecol Oncol 36(2):189–191 Chapman JA, DiSaia PJ, Osann K, Roth PD, Gillotte DL, Berman ML, 1996, Estrogen replacement in surgical stage I and II endometrial cancer survivors. Am J Obstet Gynecol 175(5):1195–1200 Suriano KA, McHale M, McLaren CE, Li KT, Re A, DiSaia PJ, 2001, Estrogen replacement therapy in endometrial cancer patients: a matched control study. Obstet Gynecol 97(4):555–560 Ayhan A, Taskiran C, Simsek S, Sever A, 2006, Does immediate hormone replacement therapy affect the oncologic outcome in endometrial cancer survivors? Int J Gynecol Cancer 16(2):805– 808 Barakat RR, Bundy BN, Spirtos NM, Bell J, Mannel RS, Gynecologic Oncology Group Study, 2006, Randomized double-blind trial of estrogen replacement therapy versus placebo in stage I or II endometrial cancer: a Gynecologic Oncology group study. J Clin Oncol 24(4):587–592 Shim SH, Lee SJ, Kim SN, 2014, Effects of hormone replacement therapy on the rate of recurrence in endometrial cancer survivors: a meta-analysis. Eur J Cancer 50(9):1628–1637 Pink D, Lindner T, Mrozek A, Kretzschmar A, Thuss-Patience PC, Dorken B, Reichardt P, 2006, Harm or benefit of hormone replacement inmetastatic low-grade endometrial stromal sarcoma: single centre experience with 10 cases and review of literature. Gynecol Oncol 101(3):464–469 Kelley TW, Borden EC, Goldblum JR, 2004, Estrogen and progesterone receptor expression in uterine and extrauterine leiomyosarcomas: an immunohistochemical study. Appl Immunohistochem Mol Morphol 12(4):338–341 Kapp DS, Shin JY, Chan JK, 2008, Prognostic factors and survival in 1396 patients with uterine leiomyosarcomas: emphasis on impact of lymphadenectomy and oophorectomy. Cancer 112(4): 820–830 Guidozzi F, 2013, Estrogen therapy in gynecological cancer survivors. Climacteric 16(6):611–617 Ploch E., 1987, Hormonal replacement therapy in patients after cervical cancer treatment. Gynecol Oncol 1987 Feb;26(2):169- 177. Madsen BS1, Jensen HL, van den Brule AJ,Wohlfahrt J, FrischM, 2008, Risk factors for invasive squamous cell carcinoma of the vulva and vagina–population-based case-control study in Denmark. Int J Cancer 122(12):2827–2834 Sherman KJ, Daling JR, McKnight B, Chu J, 1994, Hormonal factors in vulvar cancer. A case–control study. J Reprod Med 39(11):857–861 Somoye GO, Gull S, 2005, Adenocarcinoma of the vaginal vault following prolonged unopposed estrogen hormone replacement therapy. J Obstet Gynaecol 25(2):220–221 Kincade PW, Medina KL, Payne KJ, Rossi MI, Tudor KS, Yamashita Y, Kouro T, 2000, Early B-lymphocyte precursors and their regulation by sex steroids. Immunol Rev 175:128–137 Kauss MA, Reiterer G, Bunaciu RP, Yen A, 2008, Human myeloblastic leukemiacells, HL-60, express a membrane receptor for estrogen that signals and modulates retinoic acid-induced cell differentiation. Exp Cell Res 314(16):2999–3006 Li Q, Kopecky KJ, Mohan A, Willman CL, Appelbaum FR, Weick JK, Issa JP, 1999, Estrogen receptor methylation is associated with improved survival in adult acute myeloid leukemia. Clin Cancer Res 5(5):1077–1084 Tavani A, Pregnolato A, La Vecchia C, Franceschi S, 1997, A case-control study of reproductive factors and risk of lymphomas and myelomas. Leuk Res 21(9):885–888 Nelson RA, Levine AM, Bernstein L, 2001, Reproductive factors and risk of intermediate-or high-grade B-cell non-Hodgkin’s lymphoma in women. J Clin Oncol 19(5):1381–1387 Medina KL, Strasser A, Kincade PW, 2000, Estrogen influences the differentiation, proliferation, and survival of early B-lineage precursors. Blood 95(6):2059–2067 Preti HA, Cabanillas F, Talpaz M, Tucker SL, Seymour JF, Kurzrock R, 1997, Prognostic value of serum interleukin-6 in diffuse large-cell lymphoma. Ann Intern Med 127(3):186–194 Cheleuitte D, Mizuno S, Glowacki J, 1998, In vitro secretion of cytokines by human bone marrow: effects of age and estrogen status. J Clin Endocrinol Metab 83(6):2043–2051 Yang X, Wang C, He X, Wei J, Wang Y, Li X, Xu LP, 2017, Hormone therapy for premature ovarian insufficiency patients with malignant hematologic diseases. Climacteric 20(3):268–273 Tauchmanova L, Selleri C, De Rosa G, Sammartino A, Di Carlo C, Musella T, Martorelli C, Lombardi G, Rotoli B, Nappi C, Colao A, 2007, Estrogen-progestin therapy in women after stem cell transplant: our experience and literature review. Menopause 14(2):320–330 Black PM, 1997, Hormones, radiosurgery and virtual reality: new aspects of meningiomamanagement. Can J Neurol Sci 24(4):302– 306 Carroll RS, Brown M, Zhang J, DiRenzo J, Font DeMora J, McL. Black P., 2000, Expression of a subset of steroid receptor cofactors is associated with progesterone receptor expression in meningiomas. Clin Cancer Res 6:3570–3575. Hsu DW, Efird JT, Hedley-Whyte ET, 1997, Progesterone and estrogen receptors in meningiomas: prognostic considerations. J Neurosurg 86(1):113–120 Hilbig A, Barbosa-Coutinho LM, 1998, Meningiomas and hormonal receptors. Immunohistochemical study in typical and nontypical tumors. Arq Neuropsiquiatr 56(2):193–199 Pines A, 2011, Hormon therapy and brain tumors. Climacteric 14(2):215–216 O'Shea T, Crowley RK, Farrell M, MacNally S, Govender P, Feeney J, Gibney J, SherlockM., 2016, Growth of a progesterone receptor-positive meningioma in a female patient with congenital adrenal hyperplasia. Endocrinol Diabetes Metab Case Rep. 2016 pii: 16-0054. Benson VS, Kirichek O, Beral V, Green J, 2015, Menopausal hormone therapy and central nervous system tumor risk: large UK prospective study and meta-analysis. Int J Cancer 136(10): 2369–2377 Qi ZY, Shao C, Huang YL, Hui GZ, Zhou YX, Wang Z, 2013, Reproductive and exogenous hormone factors in relation to risk of meningioma in women: a meta-analysis. PLoS One 8(12):e83261 Harland TA, Freeman JL, Davern M, McCracken DJ, Celano EC, Lillehei K, Olson JJ, Ormond DR, 2018, Progesterone-only contraception is associated with a shorter progression-free survival in premenopausal women withWHO Grade I meningioma. J Neuro- Oncol 136(2):327–333 Christin-Maitre S, Delemer B, Touraine P, Young J, 2007, Prolactinoma and estrogens: pregnancy, contraception and hormonal replacement therapy. Ann Endocrinol, Paris, 68(2-3): 106–112 Sathyapalan T, Gonzalez S, Atkin SL, 2009, Effect of long-term, high-dose estrogen treatment on prolactin levels: a retrospective analysis. Climacteric 12(5):427–430 De Giorgi V, Gori A, Savarese I, D’Errico A, Scarfi F, Papi F, Maio V, Covarelli P, Massi D, Gandini S, 2017, Role of BMI and hormone therapy in melanoma risk: a case–control study. J Cancer Res Clin Oncol 143(7):1191–1197 MacKie RM, Bray CA, 2004, Hormone replacement therapy after surgery for stage 1 or 2 cutaneous melanoma. Br J Cancer 90(4): 770–772 Botteri E, Stoer NC, Sakshaug S, Graff-Iversen S, Vangen S, Hofvind S, Ursin G, Weiderpass E, 2017, Menopausal hormone therapy and risk of melanoma: Do estrogens and progestins have a different role? Int J Cancer 141(9):1763–1770 Marzagalli M, Marelli MM, Casati L, Fontana F, Moretti RM, Limonta P, 2016, Estrogen Receptor β in Melanoma: From Molecular Insights to Potential Clinical Utility. Front Endocrinol, Lausanne, 7:140 Marzagalli M, Casati L, Moretti RM, Montagnani Marelli M, Limonta P, 2015, Estrogen receptor beta agonists differentially affect the growth of human melanoma cell lines. PLoS One 10(7):e0134396 Thomas C, Gustafsson JA, 2011, The different roles of ER subtypes in cancer biology and therapy. Nat Rev Cancer 11(8):597– 608 de Giorgi V, Gori A, Grazzini M, Rossari S, Scarfi F, Corciova S, Verdelli A, Lotti T, Massi D, 2011, Estrogens, estrogen receptors and melanoma. Expert Rev Anticancer Ther 11(5):739–747 de Giorgi V, Gori A, Gandini S, Papi F, Grazzini M, Rossari S, Simoni A, Maio V, Massi D, 2013, Oestrogen receptor beta and melanoma: a comparative study. Br J Dermatol 168(3):513–519 MacKieRM, Bray CA, 2004, Hormone replacement therapy after surgery for stage 1 or 2 cutaneous melanoma. Br J Cancer 90(4): 770–772 Hsu LH, Chu NM, Kao SH, 2017, Estrogen, Estrogen Receptor and Lung Cancer. Int J Mol Sci 18(8):1713 Miki Y, Abe K, Suzuki S, Suzuki T, Sasano H, 2011, Suppression of estrogen actions in human lung cancer. Mol Cell Endocrinol 340(2):168–174 Chlebowski RT, Anderson GL, Manson JE, Schwartz AG, Wakelee H, Gass M, Rodabough RJ, Johnson KC, Wactawski- Wende J, Kotchen JM, Ockene JK, O'Sullivan MJ, Hubbell FA, Chien JW, Chen C, Stefanick ML., 2010, Lung cancer among postmenopausal women treated with estrogen alone inthe women’s health initiative randomized trial. J Natl Cancer Inst 2010 Sep 22;102(18):1413-1421. Clague J, Reynolds P, Henderson KD, Sullivan-Halley J, Ma H, Lacey JV Jr, Chang S, Delclos GL, Du XL, Forman MR, Bernstein L, 2014, Menopausal hormone therapy and lung cancer-specific mortality following diagnosis:the California teachers study. PLoS One 9(7):e103735 Ganti AK1, Sahmoun AE, Panwalkar AW, Tendulkar KK, Potti A, 2016, Hormone replacement therapy is associated with decreased survival in women with lung cancer. J Clin Oncol 24(1):59–63 Miki Y, Abe K, Suzuki S, Suzuki T, Sasano H, 2011, Suppression of estrogen actions in human lung cancer. Mol Cell Endocrinol 340(2):168–174 Regula J, Rupinski M, Kraszewska E, Polkowski M, Pachlewski J, Orlowska J, Nowacki MP, Butruk E, 2006, Colonoscopy in colorectal-cancerscreening for detection of advanced neoplasia. N Engl J Med 355(18):1863–1872 Koo JH, Jalaludin B, Wong SK, Kneebone A, Connor SJ et al, 2008, Improved survival in young women with colorectal cancer. Am J Gastroenterol 103(6):1488–1495 McArdle CS, McMillan DC, Hole DJ, 2003, Male gender adversely affects survival following surgery for colorectal cancer. Br J Surg 90(6):711–715 Murphy N, Strickler HD, Stanczyk FZ, Xue X, Wassertheil- Smoller S, Rohan TE, Ho GY, Anderson GL, Potter JD, Gunter MJ., 2015, A prospective evaluation of endogenous sex hormone levels and colorectal cancer risk in postmenopausal women. J Natl Cancer Inst.107(10). pii: djv210. Limsui D, Vierkant RA, Tillmans LS, Wang AH, Weisenberger DJ, Laird PW, Lynch CF, Anderson KE, French AJ, Haile RW, Harnack LJ, Potter JD, Slager SL, Smyrk TC, Thibodeau SN, Cerhan JR, Limburg PJ, 2012, Postmenopausal hormone therapy and colorectal cancer risk by molecularly defined subtypes among older women. Gut 61(9):1299–1305 Newcomb PA, Zheng Y, Chia VM, Morimoto LM, Doria-Rose VP, Templeton A, Thibodeau SN, Potter JD, 2007, Estrogen plus progestin use,microsatellite instability, and the risk of colorectal cancer in women. Cancer Res 67(15):7534–7539 Grodstein F, Newcomb PA, Stampfer MJ, 1999, Postmenopausal hormone therapy and the risk of colorectal cancer: a review and meta-analysis. Am J Med 106(5):574–582 Nanda K, Bastian LA, Hasselblad V, Simel DL, 1999, Hormone replacement therapy and the risk of colorectal cancer: a meta-analysis. Obstet Gynecol 93(5 Pt 2):880–888 Lin KJ, CheungWY, Lai JY, Giovannucci EL, 2012, The effect of estrogen vs.combined estrogen-progestogen therapy on the risk of colorectal cancer. Int J Cancer 130(2):419–430 Caiazza F1, Ryan EJ1, Doherty G1, Winter DC2, Sheahan K3, 2015, Estrogen receptors and their implications in colorectal carcinogenesis. Front Oncol 5:19 Stevanato Filho PR, Aguiar S Jr, Begnami MD, Kuasne H, Spencer RM, Nakagawa WT, Bezerra TS, Kupper BC, Takahashi RM, Filho MR, Rogatto SR, Lopes A, 2017, Oestrogen receptor beta isoform expression in sporadic colorectal cancer, familial adenomatous polyposis and progressive stages of colorectal cancer. BMC Cancer 17:754 Stevanato Filho PR, Aguiar Junior S, Begnami MD, Ferreira FO, Nakagawa WT, Spencer RMSB, Bezerra TS, Boggiss PE, Lopes A., 2018, Estrogen Receptor β as a Prognostic Marker of Tumor Progression in Colorectal Cancer with Familial Adenomatous Polyposis and Sporadic Polyps. Pathol Oncol Res, 3):533-540. Topi G, Ehrnstrom R, Jirstrom K, Palmquist I, Lydrup ML, Sjolander A, 2017, Association of the oestrogen receptor beta with hormone status and prognosis in a cohort of female patients with colorectal cancer. Eur J Cancer 83:279–289 Karami S, Daugherty SE, Schonfeld SJ, Park Y, Hollenbeck AR, Grubb RL 3rd, Hofmann JN, Chow WH, Purdue MP, 2013, Reproductive factors and kidney cancer risk in 2 US cohort studies, 1993-2010. Am J Epidemiol 177(12):1368–1377 Zucchetto A, Talamini R, Dal Maso L, Negri E, Polesel J, Ramazzotti V, Montella M, Canzonieri V, Serraino D, La Vecchia C, Franceschi S, 2008, Reproductive, menstrual, and other hormone-related factors and risk of renal cell cancer. Int J Cancer 123(9):2213–2216 Lee JE, Hankinson SE, Cho E, 2009, Reproductive factors and risk of renal cell cancer: the Nurses’ Health Study. Am J Epidemiol 169(10):1243–1250 Molokwu JC, Prizment AE, Folsom AR, 2007, Reproductive characteristics and risk of kidney cancer: IowaWomen’s. Maturitas 58(2):156–163 McCredie M, Stewart JH, 1992, Risk factors for kidney cancer in New South Wales, Australia. II. Urologic disease, hypertension, obesity, and hormonal factors. Cancer Causes Control 3(4):323– 331 Zhao XH, Gu SZ, Liu SX, Pan BR, 2003, Expression of estrogen receptor and estrogen receptor messenger RNA in gastric carcinoma tissues. World J Gastroenterol 9(4):665–669 Matsui M, Kojima O, Kawakami S, Uehara Y, Takahashi T, 1992, The prognosis of patients with gastric cancer possessing sex hormone receptors. Surg Today 22(5):421–425 Ur RahmanMS, Cao J, 2016, Estrogen receptors in gastric cancer: Advances and perspectives. World J Gastroenterol 22(8):2475– 2482 Tang W, Liu R, Yan Y, Pan X, Wang M, Han X, Ren H, Zhang Z, 2017, Expression of estrogen receptors and androgen receptor and their clinical significance in gastric cancer. Oncotarget 8(25): 40765–40777 Kim HW, Kim JH, Lim BJ, Kim H, Kim H, Park JJ, Youn YH, Park H, Noh SH, Kim JW, Choi SH, 2016, Sex Disparity in Gastric Cancer: Female Sex is a Poor Prognostic Factor for Advanced Gastric Cancer. Ann Surg Oncol 23(13):4344–4351 Brusselaers N, Maret-Ouda J, Konings P, El-Serag HB, Lagergren J, 2017, Menopausal hormone therapy and the risk of esophageal and gastric cancer. Int J Cancer 140(7):1693–1699 Adami HO1, Persson I, Hoover R, Schairer C, Bergkvist L, 1989, Risk of cancer in women receiving hormone replacement therapy. Int J Cancer 44(5):833–839 Shi L, Feng Y, Lin H, Ma R, Cai X, 2014, Role of estrogen in hepatocellular carcinoma: is inflammation the key? J Transl Med 12:93 Montella M, D’Arena G, Crispo A, Capunzo M, Nocerino F, Grimaldi M, Barbieri A, D’Ursi AM, Tecce MF, Amore A, Galdiero M, Ciliberto G, Giudice A, 2015, Role of Sex Hormones in the Development and Progression of Hepatitis B Virus-Associated Hepatocellular Carcinoma. Int J Endocrinol 2015:854530 Hassan MM, Botrus G, Abdel-Wahab R, Wolff RA, Li D, Tweardy D, Phan AT, Hawk E, Javle M, Lee JS, Torres HA, Rashid A, Lenzi R, Hassabo HM, Abaza Y, Shalaby AS, Lacin S, Morris J, Patt YZ, Amos CI, Khaderi SA, Goss JA, Jalal PK, Kaseb AO, 2017, Estrogen Replacement Reduces Risk and Increases Survival Times of Women With Hepatocellular Carcinoma. Clin Gastroenterol Hepatol 15(11):1791–1799 Basakci A, Kirkali Z, Tuzel E, Yorukoglu K, Mungan MU, Sade M, 2002, Prognostic significance of estrogen receptor expression in superficial transitional cell carcinoma of the urinary bladder. Eur Urol 41(3):342–345 Fernandez E, Gallus S, Bosetti C, Franceschi S, Negri E, La Vecchia C, 2003, Hormone replacement therapy and cancer risk: a systematic analysis from a network of case-control studies. Int J Cancer 105(3):408–412 Godoy G, Gakis G, Smith CL, Fahmy O, 2016, Effects of Androgen and Estrogen Receptor Signaling Pathways on Bladder Cancer Initiation and Progression. Bladder Cancer 2(2): 127–137 Dellagrammaticas D, Bryden AA, Collins GN, 2001, Regression of metastatic transitional cell carcinoma in response to tamoxifen. J Urol 165(5):1631 Hsu CH, Chen J,Wu CY, Cheng AL, Pu YS, 2001, Combination chemotherapy of cisplatin, methotrexate, vinblastine, , and highdose tamoxifen for transitional cell carcinoma. Anticancer Res 21(1B):711–715 Shang Z, Li Y, Hsu I, Zhang M, Tian J, Wen S, Han R, Messing EM, Chang C, Niu Y, Yeh S, 2016, Targeting estrogen/estrogen receptor alpha enhances Bacillus Calmette-Guerin efficacy in bladder cancer. Oncotarget 7(19):27325–27335 Moleti M, Sturniolo G, Di Mauro M, Russo M, Vermiglio F, 2017, Female Reproductive Factors and Differentiated Thyroid Cancer. Front Endocrinol, Lausanne, 8:111 Galanti MR, Hansson L, Lund E, Bergstrom R, Grimelius L, Stalsberg H, Carlsen E, Baron JA, Persson I, Ekbom A, 1996, Reproductive history and cigarette smoking as risk factors for thyroid cancer in women: a population-based case-control study. Cancer Epidemiol Biomark Prev 5(6):425–431 Mack WJ, Preston-Martin S, Bernstein L, Qian D, Xiang M., 1999, Reproductive and hormonal risk factors for thyroid cancer in Los Angeles county females. Cancer Epidemiol Biomarkers Prev. 8(11):991-997. Rossing MA, Voigt LF, Wicklund KG, Daling JR, 2000, Reproductive factors and risk of papillary thyroid cancer in women. Am J Epidemiol 151(8):765–772 Sakoda LC, Horn-Ross PL, 2002, Reproductive and menstrual history and papillary thyroid cancer risk: the San Francisco bay area thyroid cancer study. Cancer Epidemiol Biomark Prev 11(1): 51–57 Memon A, Darif M, Al-Saleh K, Suresh A, 2002, Epidemiology of reproductive and hormonal factors in thyroid cancer: evidence from a case-control study in the Middle East. Int J Cancer 97(1): 82–89 Truong T, Orsi L, Dubourdieu D, Rougier Y, Hemon D, Guenel P, 2005, Role of goiter and of menstrual and reproductive factors in thyroid cancer: a population- based case-control study in New Caledonia, South Pacific), a very high incidence area. Am J Epidemiol 161(11):1056–1065 Sturniolo G, Zafon C, Moleti M, Castellvi J, Vermiglio F,Mesac J, 2016, Immunohistochemical Expression of Estrogen Receptor-α and Progesterone Receptor in Patients with Papillary Thyroid Cancer. Eur Thyroid J 5(4):224–230 Yi JW, Kim SJ, Kim JK, Seong CY, Yu HW, Chai YJ, Choi JY, Lee KE, 2017, Upregulation of the ESR1 Gene and ESR Ratio, ESR1/ESR2, is Associated with a Worse Prognosis in Papillary Thyroid Carcinoma: The Impact of the Estrogen Receptor α/β Expression on Clinical Outcomes in Papillary Thyroid Carcinoma Patients. Ann Surg Oncol 24(12):3754–3762 Jastrzebska H, Gietka-Czernel M, Zgliczynski S, 2001, Hormonal replacement therapy in women after surgery for thyroid cancer treated with suppressive doses of L-thyroxine. Wiad Lek 54(Suppl 1):383–388 Tang B, Lv J, Li Y, Yuan S, Wang Z, He S, 2015, Relationship between female hormonal and menstrual factors and pancreatic cancer: a meta-analysis of observational studies. Medicine, Baltimore, 94(7):e177 Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, Kotchen JM, Ockene J, Writing Group for the Women's Health Initiative Investigators, 2002, Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women's Health Initiative randomized controlled trial. JAMA 288(3): 321–333 van der Schouw YT, van der Graaf Y, Steyerberg EW, Eijkemans JC, Banga JD, 1996, Age at menopause as a risk factor for cardiovascular mortality. Lancet 347(9003):714–718 de Villiers TJ, Gass ML, Haines CJ, Hall JE, Lobo RA, Pierroz DD, Rees M, 2013, Global Consensus Statement on Menopausal Hormone Therapy. Climacteric 16(2):203–204 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 63 EP 78 DI 10.1007/s12253-018-00569-x PG 16 ER PT J AU Evans, M O’Sullivan, B Hughes, F Mullis, T Smith, M Trim, N Taniere, P AF Evans, Matthew O’Sullivan, Brendan Hughes, Frances Mullis, Tina Smith, Matthew Trim, Nicola Taniere, Philippe TI The Clinicopathological and Molecular Associations of PD-L1 Expression in Non-small Cell Lung Cancer: Analysis of a Series of 10,005 Cases Tested with the 22C3 Assay SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; PD-L1; Immunotherapy; Pembrolizumab ID Lung cancer; PD-L1; Immunotherapy; Pembrolizumab AB PD-L1 expression testing is mandatory prior to pembrolizumab prescription in non-small cell lung cancer. Our service offers PDL1 testing using the PD-L1 IHC 22C3 pharmDx assay, in parallel with EGFR, ALK, ROS1 and (in some cases) KRAS testing. We correlate PD-L1 expression in 10,005 tumours with patient age and sex, with tumour histological subtypes, with the sampling modality and type of tissue, and with the presence of other molecular alterations. PD-L1 expression testing was performed using the aforementioned assay; tumour proportion scores (TPS) of 1 and 50% were taken as cut-offs for low and high positivity, respectively. EGFR testing was performed using the cobas® EGFR Mutation Test v2. ALK testing was performed using the VENTANA ALK (D5F3) CDx Assay. KRAS testing was performed using pyrosequencing. TPS <1% was seen in 44.4% of tumours, 1–49% in 25.0% and ≥ 50% in 30.6%.We identified no significant relationship with age. Female patients were slightly more likely to express PD-L1. Poorly-differentiated tumour histology and ALK translocation were significantly associated with PD-L1 expression. Rare EGFR mutations tended to be associated with PD-L1 expression. Pleural and nodal metastases were more likely to express PD-L1 than primary tumours, but biopsy and cytological specimens did not show different PD-L1 expression rates. Our data show that the means of acquiring a tumour sample (biopsy versus cytology) does not have a significant impact on PD-L1 expression. However, we found that certain metastatic sites were associated with significantly higher expression rates, which has substantial implications for selection of tissue for testing. C1 [Evans, Matthew] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2TH Birmingham, UK. [O’Sullivan, Brendan] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2TH Birmingham, UK. [Hughes, Frances] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2TH Birmingham, UK. [Mullis, Tina] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2TH Birmingham, UK. [Smith, Matthew] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2TH Birmingham, UK. [Trim, Nicola] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2TH Birmingham, UK. [Taniere, Philippe] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Mindelsohn Way, B15 2TH Birmingham, UK. RP Evans, M (reprint author), Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, B15 2TH Birmingham, UK. EM matthew.evans7@nhs.net CR Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, Gottfried M, Peled N, Tafreshi A, Cuffe S, O'Brien M, Rao S, Hotta K, Leiby MA, Lubiniecki GM, Shentu Y, Rangwala R, Brahmer JR, Investigators K, 2016, Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung Cancer. N Engl J Med 375(19):1823–1833. , DOI 10.1056/ NEJMoa1606774 Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, Molina J, Kim JH, Arvis CD, Ahn MJ, Majem M, Fidler MJ, de Castro G Jr, Garrido M, Lubiniecki GM, Shentu Y, Im E, Dolled- Filhart M, Garon EB, 2016, Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer, KEYNOTE-010): a randomised controlled trial. Lancet 387(10027):1540–1550. , DOI 10.1016/S0140-6736(15, 01281-7 Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, GubensM, Horn L, Carcereny E, AhnMJ, Felip E, Lee JS, Hellmann MD, Hamid O, Goldman JW, Soria JC, Dolled-Filhart M, Rutledge RZ, Zhang J, Lunceford JK, Rangwala R, Lubiniecki GM, Roach C, Emancipator K, Gandhi L, Investigators K, 2015, Pembrolizumab for the treatment of nonsmall- cell lung cancer. N Engl J Med 372(21):2018–2028. https:// doi.org/10.1056/NEJMoa1501824 Cho J, ZhouW, Choi YL, Sun JM, Choi H, Kim TE, Dolled-Filhart M, Emancipator K, Rutkowski MA, Kim J, 2015, 26OMolecular epidemiology study of PD-L1 expression in patients, pts, with EGFR-mutant NSCLC. Ann Oncol 26(suppl_9):ix8. https://doi. org/10.1093/annonc/mdv518.01 Salanova R, Pereyra JCC, Leguina L, Bena A, Barberis M, Vargas R, RJd T, Kreimberg K, Powazniak Y, 2017, Epidemiology of PDL1 and ALK expression and EGFR mutational status in Argentinian patients with lung cancer. J Clin Oncol 35(15_suppl): e20565. , DOI 10.1200/JCO.2017.35.15_suppl.e20565 Sorensen SF, Zhou W, Dolled-Filhart M, Georgsen JB, Wang Z, Emancipator K, Wu D, Busch-Sorensen M, Meldgaard P, Hager H, 2016, PD-L1 expression and survival among patients with advanced non-small cell lung Cancer treated with chemotherapy. Transl Oncol 9(1):64–69. , DOI 10. 1016/j.tranon.2016.01.003 Pan Y, Zheng D, Li Y, Cai X, Zheng Z, Jin Y, Hu H, Cheng C, Shen L,Wang J, Ji H, Sun Y, ZhouX, Chen H, 2017, Unique distribution of programmed death ligand 1, PD-L1, expression in east Asian non-small cell lung cancer. J Thorac Dis 9(8):2579–2586. https:// doi.org/10.21037/jtd.2017.08.61 Sun JM, Zhou W, Choi YL, Choi SJ, Kim SE, Wang Z, Dolled- Filhart M, Emancipator K, Wu D, Weiner R, Frisman D, Kim HK, Choi YS, Shim YM, Kim J, 2016, Prognostic significance of PDL1 in patients with non-small cell lung Cancer: a large cohort study of surgically resected cases. J Thorac Oncol 11(7):1003–1011. , DOI 10.1016/j.jtho.2016.04.007 Li C, Huang C, Mok TS, ZhuangW, Xu H, Miao Q, Fan X, ZhuW, Huang Y, Lin X, Jiang K, Hu D, Chen X, Huang P, Lin G, 2017, Comparison of 22C3 PD-L1 expression between surgically resected specimens and paired tissue microarrays in non-small cell lung Cancer. J Thorac Oncol 12(10):1536–1543. , DOI 10. 1016/j.jtho.2017.07.015 Cooper WA, Tran T, Vilain RE, Madore J, Selinger CI, Kohonen- Corish M, Yip P, Yu B, O'Toole SA, McCaughan BC, Yearley JH, Horvath LG, Kao S, Boyer M, Scolyer RA, 2015, PD-L1 expression is a favorable prognostic factor in early stage non-small cell carcinoma. Lung Cancer 89(2):181–188. , DOI 10.1016/j. lungcan.2015.05.007 Rangachari D, VanderLaan PA, Shea M, Le X, Huberman MS, Kobayashi SS, Costa DB, 2017, Correlation between classic driver oncogene mutations in EGFR, ALK, or ROS1 and 22C3-PD-L1 >/=50% expression in lung adenocarcinoma. J Thorac Oncol 12(5): 878–883. , DOI 10.1016/j.jtho.2016.12.026 Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, Miller ML, Rekhtman N, Moreira AL, Ibrahim F, Bruggeman C, Gasmi B, Zappasodi R, Maeda Y, Sander C, Garon EB, Merghoub T, Wolchok JD, Schumacher TN, Chan TA, 2015, Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348(6230):124–128. https:// doi.org/10.1126/science.aaa1348 Shimoji M, Shimizu S, Sato K, Suda K, Kobayashi Y, Tomizawa K, Takemoto T, Mitsudomi T, 2016, Clinical and pathologic features of lung cancer expressing programmed cell death ligand 1, PD-L1). Lung Cancer 98:69–75. https://doi. org/10.1016/j.lungcan.2016.04.021 Azuma K, Ota K, Kawahara A, Hattori S, Iwama E, Harada T, Matsumoto K, Takayama K, Takamori S, Kage M, Hoshino T, Nakanishi Y, Okamoto I, 2014, Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann Oncol 25(10):1935–1940. https:// doi.org/10.1093/annonc/mdu242 McLaughlin J, Han G, Schalper KA, Carvajal-Hausdorf D, Pelekanou V, Rehman J, Velcheti V, Herbst R, LoRusso P, Rimm DL, 2016, Quantitative assessment of the heterogeneity of PD-L1 expression in non-small-cell lung Cancer. JAMA Oncol 2(1):46– 54. , DOI 10.1001/jamaoncol.2015.3638 Cha YJ, Kim HR, Lee CY, Cho BC, Shim HS, 2016, Clinicopathological and prognostic significance of programmed cell death ligand-1 expression in lung adenocarcinoma and its relationship with p53 status. Lung Cancer 97:73–80. , DOI 10.1016/j.lungcan.2016.05.001 Vieira T, AntoineM, Hamard C, Fallet V, Duruisseaux M, Rabbe N, Rodenas A, Cadranel J, Wislez M, 2016, Sarcomatoid lung carcinomas show high levels of programmed death ligand-1, PD-L1, and strong immune-cell infiltration by TCD3 cells and macrophages. Lung Cancer 98:51–58. , DOI 10.1016/j.lungcan. 2016.05.013 Ilie M, Long-Mira E, Bence C, Butori C, Lassalle S, Bouhlel L, Fazzalari L, Zahaf K, Lalvee S,Washetine K,Mouroux J, Venissac N, Poudenx M, Otto J, Sabourin JC, Marquette CH, Hofman V, Hofman P, 2016, Comparative study of the PD-L1 status between surgically resected specimens and matched biopsies of NSCLC patients reveal major discordances: a potential issue for anti-PDL1 therapeutic strategies. Ann Oncol 27(1):147–153. https://doi. org/10.1093/annonc/mdv489 Huynh TG, Morales-Oyarvide V, CampoMJ, Gainor JF, Bozkurtlar E, Uruga H, Zhao L, Gomez-Caraballo M, Hata AN, Mark EJ, Lanuti M, Engelman JA, Mino-Kenudson M, 2016, Programmed cell death ligand 1 expression in resected lung adenocarcinomas: association with immune microenvironment. J Thorac Oncol 11(11):1869–1878. , DOI 10.1016/j.jtho.2016.08.134 FangW, Hong S, Chen N, He X, Zhan J, Qin T, Zhou T, Hu Z, Ma Y, Zhao Y, Tian Y, Yang Y, Xue C, Tang Y, Huang Y, Zhao H, Zhang L, 2015, PD-L1 is remarkably over-expressed in EBVassociated pulmonary lymphoepithelioma-like carcinoma and related to poor disease-free survival. Oncotarget 6(32):33019–33032. , DOI 10.18632/oncotarget.5028 Calles A, Liao X, Sholl LM, Rodig SJ, Freeman GJ, Butaney M, Lydon C, Dahlberg SE, Hodi FS, Oxnard GR, Jackman DM, Janne PA, 2015, Expression of PD-1 and its ligands, PD-L1 and PD-L2, in smokers and never smokers with KRAS-mutant lung Cancer. J Thorac Oncol 10(12):1726–1735. , DOI 10.1097/JTO. 0000000000000687 Koh J, Go H, Keam B, Kim MY, Nam SJ, Kim TM, Lee SH, Min HS, Kim YT, Kim DW, Jeon YK, Chung DH, 2015, Clinicopathologic analysis of programmed cell death-1 and programmed cell death-ligand 1 and 2 expressions in pulmonary adenocarcinoma: comparison with histology and driver oncogenic alteration status. Mod Pathol 28(9):1154–1166. , DOI 10. 1038/modpathol.2015.63 Kitazono S, Fujiwara Y, Tsuta K, Utsumi H, Kanda S, Horinouchi H, Nokihara H, Yamamoto N, Sasada S, Watanabe S, Asamura H, Tamura T, Ohe Y, 2015, Reliability of small biopsy samples compared with resected specimens for the determination of programmed death-ligand 1 expression in non–small-cell lung Cancer. Clin Lung Cancer 16(5):385–390. , DOI 10.1016/ j.cllc.2015.03.008 Zhang Y,Wang L, Li Y, Pan Y,Wang R, Hu H, Li H, Luo X, Ye T, Sun Y, Chen H, 2014, Protein expression of programmed death 1 ligand 1 and ligand 2 independently predict poor prognosis in surgically resected lung adenocarcinoma. Onco Targets Ther 7:567– 573. , DOI 10.2147/OTT.S59959 Uruga H, Bozkurtlar E, Huynh TG, Muzikansky A, Goto Y, Gomez-Caraballo M, Hata AN, Gainor JF, Mark EJ, Engelman JA, Lanuti MD, Mino-KenudsonM(2017, Programmed cell death ligand, PD-L1, expression in stage II and III lung adenocarcinomas and nodal metastases. J Thorac Oncol 12(3):458–466. https://doi. org/10.1016/j.jtho.2016.10.015 Tsao MS, Le Teuff G, Shepherd FA, Landais C, Hainaut P, Filipits M, Pirker R, Le Chevalier T, Graziano S, Kratze R, Soria JC, Pignon JP, Seymour L, Brambilla E, 2017, PD-L1 protein expression assessed by immunohistochemistry is neither prognostic nor predictive of benefit from adjuvant chemotherapy in resected nonsmall cell lung cancer. Ann Oncol 28, 4):882–889. , DOI 10.1093/annonc/mdx003 Toyokawa G, Takada K, Okamoto T, Kawanami S, Kozuma Y, Matsubara T, Haratake N, Takamori S, Akamine T, Katsura M, Yamada Y, Shoji F, Baba S, Kamitani T, Oda Y, Honda H, Maehara Y, 2017, Relevance between programmed death ligand 1 and radiologic invasiveness in pathologic stage I lung adenocarcinoma. Ann Thorac Surg 103(6):1750–1757. , DOI 10. 1016/j.athoracsur.2016.12.025 Wu S, Shi X, Sun J, Liu Y, Luo Y, Liang Z,Wang J, Zeng X, 2017, The significance of programmed cell death ligand 1 expression in resected lung adenocarcinoma. Oncotarget 8(10):16421–16429. , DOI 10.18632/oncotarget.14851 Cho JH, Zhou W, Choi YL, Sun JM, Choi H, Kim TE, Dolled- Filhart M, Emancipator K, Rutkowski MA, Kim J, 2017, Retrospective molecular epidemiology study of PD-L1 expression in patients with EGFR-mutant non-small cell lung Cancer. Cancer Res Treat 50:95–102. , DOI 10.4143/crt.2016.591 Zhang M, Li G, Wang Y, Wang Y, Zhao S, Haihong P, Zhao H, Wang Y, 2017, PD-L1 expression in lung cancer and its correlation with driver mutations: a meta-analysis. Sci Rep 7(1):10255. https:// doi.org/10.1038/s41598-017-10925-7 Ota K, Azuma K, Kawahara A, Hattori S, Iwama E, Tanizaki J, Harada T, Matsumoto K, Takayama K, Takamori S, Kage M, Hoshino T, Nakanishi Y, Okamoto I, 2015, Induction of PD-L1 expression by the EML4-ALK Oncoprotein and downstream signaling pathways in non-small cell lung Cancer. Clin Cancer Res 21(17):4014–4021. , DOI 10.1158/1078-0432.CCR-15- 0016 Pan ZK, Ye F, Wu X, An HX, Wu JX, 2015, Clinicopathological and prognostic significance of programmed cell death ligand1, PDL1, expression in patients with non-small cell lung cancer: a metaanalysis. J Thorac Dis 7(3):462–470. , DOI 10.3978/j.issn. 2072-1439.2015.02.13 Yang H, Chen H, Luo S, Li L, Zhou S, Shen R, Lin H, Xie X, 2017, The correlation between programmed death-ligand 1 expression and driver gene mutations in NSCLC. Oncotarget 8(14):23517– 23528. , DOI 10.18632/oncotarget.15627 D'Incecco A, Andreozzi M, Ludovini V, Rossi E, Capodanno A, Landi L, Tibaldi C, Minuti G, Salvini J, Coppi E, Chella A, Fontanini G, Filice ME, Tornillo L, Incensati RM, Sani S, Crino L, Terracciano L, Cappuzzo F, 2015, PD-1 and PD-L1 expression in molecularly selected non-small-cell lung cancer patients. Br J Cancer 112(1):95–102. , DOI 10.1038/bjc.2014.555 Li J, Chen Y, Shi X, Le X, Feng F, Chen J, Zhou C, Chen Y,Wen S, Zeng H, Chen AM, Zhang Y, 2017, A systematic and genomewide correlation meta-analysis of PD-L1 expression and targetable NSCLC driver genes. J Thorac Dis 9(8):2560–2571. , DOI 10.21037/jtd.2017.07.117 Wang A,Wang HY, Liu Y, Zhao MC, Zhang HJ, Lu ZY, Fang YC, Chen XF, LiuGT, 2015, The prognostic value of PD-L1 expression for non-small cell lung cancer patients: a meta-analysis. Eur J Surg Oncol 41(4):450–456. , DOI 10.1016/j.ejso.2015.01.020 Kim MY, Koh J, Kim S, Go H, Jeon YK, Chung DH, 2015, Clinicopathological analysis of PD-L1 and PD-L2 expression in pulmonary squamous cell carcinoma: comparison with tumor-infiltrating T cells and the status of oncogenic drivers. Lung Cancer 88(1):24–33. , DOI 10.1016/j.lungcan. 2015.01.016 Okita R, Maeda A, Shimizu K, Nojima Y, Saisho S, Nakata M, 2017, PD-L1 overexpression is partially regulated by EGFR/ HER2 signaling and associated with poor prognosis in patients with non-small-cell lung cancer. Cancer Immunol Immunother 66(7): 865–876. , DOI 10.1007/s00262-017-1986-y Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, Chen L, Pardoll DM, Topalian SL, Anders RA, 2014, Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 20(19):5064–5074. , DOI 10.1158/1078-0432.CCR-13- 3271 Kerr KM, Tsao MS, Nicholson AG, Yatabe Y, Wistuba II, Hirsch FR, Committee IP, 2015, Programmed death-ligand 1 immunohistochemistry in lung Cancer: in what state is this art? J Thorac Oncol 10(7):985–989. , DOI 10.1097/JTO.0000000000000526 Hong S, Chen N, Fang W, Zhan J, Liu Q, Kang S, He X, Liu L, Zhou T, Huang J, Chen Y, Qin T, Zhang Y, Ma Y, Yang Y, Zhao Y, Huang Y, Zhang L, 2016, Upregulation of PD-L1 by EML4-ALK fusion protein mediates the immune escape in ALK positive NSCLC: implication for optional anti-PD-1/PD-L1 immune therapy for ALK-TKIs sensitive and resistant NSCLC patients. Oncoimmunology 5(3):e1094598. , DOI 10.1080/ 2162402X.2015.1094598 Chen N, FangW, Lin Z, Peng P,Wang J, Zhan J, Hong S, Huang J, Liu L, Sheng J, Zhou T, Chen Y, Zhang H, Zhang L, 2017, KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma. Cancer Immunol Immunother 66: 1175–1187. , DOI 10.1007/s00262-017-2005-z Li D, Zhu X, Wang H, Li N, 2017, Association between PD-L1 expression and driven gene status in NSCLC: a meta-analysis. Eur J Surg Oncol 43(7):1372–1379. , DOI 10.1016/j.ejso.2017. 02.008 Haratani K, Hayashi H, Tanaka T, Kaneda H, Togashi Y, Sakai K, Hayashi K, Tomida S, Chiba Y, Yonesaka K, Nonagase Y, Takahama T, Tanizaki J, Tanaka K, Yoshida T, Tanimura K, Takeda M, Yoshioka H, Ishida T, Mitsudomi T, Nishio K, Nakagawa K, 2017, Tumor immune microenvironment and nivolumab efficacy in EGFR mutation-positive non-small-cell lung cancer based on T790M status after disease progression during EGFR-TKI treatment. Ann Oncol 28(7):1532–1539. https://doi. org/10.1093/annonc/mdx183 Madore J, Strbenac D, Vilain R,Menzies AM, Yang JY, Thompson JF, Long GV, Mann GJ, Scolyer RA, Wilmott JS, 2016, PD-L1 negative status is associated with lower mutation burden, differential expression of immune-related genes, and worse survival in stage III melanoma. Clin Cancer Res 22(15):3915–3923. https:// doi.org/10.1158/1078-0432.CCR-15-1714 Akbay EA, Koyama S, Carretero J, Altabef A, Tchaicha JH, Christensen CL, Mikse OR, Cherniack AD, Beauchamp EM, Pugh TJ, Wilkerson MD, Fecci PE, Butaney M, Reibel JB, Soucheray M, Cohoon TJ, Janne PA, Meyerson M, Hayes DN, Shapiro GI, Shimamura T, Sholl LM, Rodig SJ, Freeman GJ, Hammerman PS, Dranoff G, Wong KK, 2013, Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov 3(12):1355–1363. , DOI 10.1158/ 2159-8290.CD-13-0310 Chen N, FangW, Zhan J, Hong S, Tang Y, Kang S, Zhang Y, He X, Zhou T, Qin T, Huang Y, Yi X, Zhang L, 2015, Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFRdriven NSCLC: implication for optional immune targeted therapy for NSCLC patients with EGFR mutation. J Thorac Oncol 10(6): 910–923. , DOI 10.1097/JTO.0000000000000500 Lastwika KJ,WilsonW, 3rd, Li QK, Norris J, Xu H, Ghazarian SR, Kitagawa H, Kawabata S, Taube JM, Yao S, Liu LN, Gills JJ, Dennis PA, 2016, Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung Cancer. Cancer Res 76, 2):227–238. , DOI 10.1158/0008-5472. CAN-14-3362 O'Kane GM, Bradbury PA, Feld R, Leighl NB, Liu G, Pisters KM, Kamel-Reid S, Tsao MS, Shepherd FA, 2017, Uncommon EGFR mutations in advanced non-small cell lung cancer. Lung Cancer 109:137–144. , DOI 10.1016/j.lungcan.2017.04.016 Peters S, Creelan BD, Hellmann M, Socinski M, Reck M, Bhagavatheeswaran P, Chang HJ, Geese W, Paz-Ares L, Carbone D, 2017, Abstract CT082: Impact of tumor mutation burden on the efficacy of first-line nivolumab in stage iv or recurrent non-small cell lung cancer: An exploratory analysis of CheckMate 026, 77. , DOI 10.1158/1538- 7445.AM2017-CT082 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 79 EP 89 DI 10.1007/s12253-018-0469-6 PG 11 ER PT J AU Du, J Wang, S Wang, R Wang, SY Han, Q Xu, HT Yang, P Liu, Y AF Du, Jiang Wang, Si Wang, Rui Wang, Si-Yao Han, Qiang Xu, Hong-Tao Yang, Peng Liu, Yang TI Identifying Secondary Mutations in Chinese Patients with Imatinib-Resistant Gastrointestinal Stromal Tumors (GISTs) by Next Generation Sequencing (NGS) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastrointestinal stromal tumors; Secondary mutation; Acquired resistance; Next generation sequencing ID Gastrointestinal stromal tumors; Secondary mutation; Acquired resistance; Next generation sequencing AB The aim of this study was to characterize secondary kinase mutations in Chinese patients with imatinib-resistant gastrointestinal stromal tumors (GISTs).Mutations in receptor tyrosine kinase (KIT; exons 9, 11, 13, 14, 17, and 18) and platelet-derived growth factor-alpha (PDGFRA; exons 12, 14, and 18) were analyzed by direct sequencing. After imatinib treatment, 425 cancer-related target genes were analyzed by next generation sequencing (NGS) in imatinib-resistant patients. Correlation of sequencing results with clinicopathologic features were analyzed. We identified 320 patients with secondary acquired resistance. We determined that 65.63% (210/320) of resistant patients had secondary KIT mutations in exon 13 (n = 134), exon 14 (n =10), or exon 17 (n = 66), and 4.38%(14/320) had additional PDGFRA mutations in exon 14 (n = 3) or exon 18 (n = 11). All secondary KIT mutations were missense mutations and were mostly located in kinase domains. Ninety-six imatinib-resistant GIST patients did not have secondary KIT or PDGFRA mutations. Common independent mutation events were found in retinoblastoma protein 1 (RB1) (18/96 cases), SWI/SNF-related matrix associated actin-dependent regulator of chromatin subfamily B member 1 (SMARCB1) (16/96 cases), and myc-associated factor X (MAX) (10/96 cases). RB1 or SMARCB1 mutations coexisted with activation of other oncogenes in 6 or 15 cases, respectively. Multiple mutations were also seen in cases with MAX mutations. These mutations are frequently associated with clinicopathological factors. Secondary mutations of KIT/PDGFRA were the most important contributors in GISTs developing resistance to imatinib treatment. Additional genetic events including RB1, SMARCB1, and MAX except secondary KIT/PDGFRA mutations are the most common for GISTs to evolve into resistant disease. Clinical assessment of the effect of these mutations may benefit existing risk assessment models and selection of adjuvant therapies in GIST patients. C1 [Du, Jiang] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110122 Shenyang, China. [Wang, Si] the College of Basic Medical Sciences of China Medical University, Department of Medical Microbiology and Human Parasitology, 110122 Shenyang, China. [Wang, Rui] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110122 Shenyang, China. [Wang, Si-Yao] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110122 Shenyang, China. [Han, Qiang] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110122 Shenyang, China. [Xu, Hong-Tao] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110122 Shenyang, China. [Yang, Peng] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110122 Shenyang, China. [Liu, Yang] China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110122 Shenyang, China. RP Liu, Y (reprint author), China Medical University, The First Affiliated Hospital and College of Basic Medical Sciences, Department of Pathology, 110122 Shenyang, China. EM liuyanglovebee@163.com CR Di Vita M, Zanghi A, Cavallaro A et al, 2019, Gastric GIST and prognostic models. Which is the best to predict survival after surgery? Ann Ital Chir 90:31–40 Kalfusova A, Kodet R, 2017, Molecular mechanisms of primary and secondary resistance, molecular-genetic features and characteristics of KIT/PDGFRA non-mutated GISTs. Cesk Patol 53(4):167– 173 Miettinen M, Monihan JM, Sarlomo-Rikala M et al, 1999, Gastrointestinal stromal tumors/smoothmuscle tumors, GISTs, primary in the omentum and mesentery: clinicopathologic and immunohistochemical study of 26 cases. Am J Surg Pathol 23(9):1109– 1118 Li J, 2016, Molecular mechanism of gastrointestinal stromal tumors and progress in drug research. Zhonghua Wei Chang Wai Ke Za Zhi 19(11):1316–1320 Corless CL, Schroeder A, Griffith D et al, 2005, PDGFRA mutations in gastrointestinal stromal tumors: frequency, spectrum and in vitro sensitivity to imatinib. J Clin Oncol 23(23):5357–5364 Lasota J, Miettinen M, 2006, KIT and PDGFRA mutations in gastrointestinal stromal tumors, GISTs). Semin Diagn Pathol 23(2): 91–102 Li J, Zhang H, Lu Yet al, 2015, Presence of PDGFRA and DOG1 mutations in gastrointestinal stromal tumors among Chinese population. Int J Clin Exp Pathol 8(5):5721–5726 Ahmad F, Lad P, Bhatia S et al, 2015)Molecular spectrum of c-KIT and PDGFRA gene mutations in gastro intestinal stromal tumor: determination of frequency, distribution pattern and identification of novel mutations in Indian patients. Med Oncol 32(1):424 Rossi G, Valli R, Bertolini F et al, 2005, PDGFR expression in differential diagnosis between KIT-negative gastrointestinal stromal tumours and other primary soft-tissue tumours of the gastrointestinal tract. Histopathology. 46(5):522–531 Heinrich MC, Corless CL, Duensing A et al, 2003, PDGFRA activating mutations in gastrointestinal stromal tumors. Science. 299(5607):708–710 Kang HJ, Nam SW, Kim H et al, 2005, Correlation of KIT and platelet-derived growth factor receptor alpha mutations with gene activation and expression profiles in gastrointestinal stromal tumors. Oncogene. 24(6):1066–1074 von Mehren M, Joensuu H, 2018, Gastrointestinal stromal tumors. J Clin Oncol 36(2):136–143 Saito S, Nakata K, Kajiura S et al, 2013, Long-term follow-up outcome of imatinib mesylate treatment for recurrent and unresectable gastrointestinal stromal tumors. Digestion 87(1):47– 52 Rodriquenz MG, Rossi S, Ricci R et al, 2016, Gastrointestinal stromal tumors, GISTs, and second malignancies: a novel “sentinel tumor”? A monoinstitutional, STROBE-compliant observational analysis. Medicine, Baltimore, 95(38):e4718 Comandone A, Boglione A, 2015, The importance of mutational status in prognosis and therapy of GIST. Recenti Prog Med 106(1): 17–22 Huang WK, Akcakaya P, Gangaev A et al, 2018, miR-125a-5p regulation increases phosphorylation of FAK that contributes to imatinib resistance in gastrointestinal stromal tumors. Exp Cell Res 371(1):287–296 Boyar MS, Taub RN, 2007, New strategies for treating GISTwhen imatinib fails. Cancer Investig 25(5):328–335 Atay S, Wilkey DW, Milhem M et al, 2018, Insights into the proteome of gastrointestinal stromal tumors-derived exosomes reveals new potential diagnostic biomarkers. Mol Cell Proteomics 17(3): 495–515 Serrano C, Marino-Enriquez A, Tao DL et al, 2019, Complementary activity of tyrosine kinase inhibitors against secondary kit mutations in imatinib-resistant gastrointestinal stromal tumours. Br J Cancer 120(6):612–620 Tsai HJ, Jiaang WT, Shih NY et al, 2018, BPR1J373, a novel multitargeted kinase inhibitor, effectively suppresses the growth of gastrointestinal stromal tumor. Cancer Sci 109(11):3591–3601 Li J, Ye Y, Wang J et al, 2017, Chinese consensus guidelines for diagnosis and management of gastrointestinal stromal tumor. Chin J Cancer Res 29(4):281–293 Bolger AM, Lohse M, Usadel B, 2014, Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 30(15):2114– 2120 Li H, Durbin R, 2009, Fast and accurate short read alignment with burrows-wheeler transform. Bioinformatics. 25(14):1754–1760 McKenna A, Hanna M, Banks E et al, 2010, The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNAsequencing data. Genome Res 20(9):1297–1303 Van der Auwera GA, Carneiro MO, Hartl C et al, 2013, From FastQ data to high confidence variant calls: the genome analysis toolkit best practicespipeline. Curr Protoc Bioinformatics 43: 11.10.1–11.1033 Koboldt DC, Zhang Q, Larson DE et al, 2012, VarScan 2: somatic mutation and copy number alteration discovery in cancer by exomesequencing. Genome Res 22(3):568–576 Robinson JT, Thorvaldsdottir H, Wenger AM et al, 2017, Variant review with the integrative genomics viewer. Cancer Res 77(21): e31–e34 Amarasinghe KC, Li J, Hunter SM et al, 2014, Inferring copy number and genotype in tumour exome data. BMC Genomics 15: 732 Chen W, Kuang Y, Qiu HB et al, 2017, Dual targeting of insulin receptor and KIT in Imatinib-resistant gastrointestinal stromal tumors. Cancer Res 77(18):5107–5117 Obata Y, Horikawa K, Shiina I et al, 2018, Oncogenic Kit signalling on the Golgi is suppressed by blocking secretory trafficking with M-COPA in gastrointestinal stromal tumours. Cancer Lett 415: 1–10 Boichuk S, GalembikovaA, Dunaev P et al, 2017, Anovel receptor tyrosine kinase switch promotes gastrointestinal stromal tumor drug resistance. Molecules. 5:22(12) Heinrich MC, Maki RG, Corless CL et al, 2008, Primary and secondary kinase genotypes correlate with the biological and clinical activity of sunitinib in imatinib-resistant gastrointestinal stromal tumor. J Clin Oncol 26(33):5352–5359 Liegl B, Kepten I, Le C et al, 2008, Heterogeneity of kinase inhibitor resistance mechanisms in GIST. J Pathol 216(1):64–74 Yu K, Liu X, Jiang Z et al, 2017, Discovery of a highly selective KIT kinase primary V559D mutant inhibitor for gastrointestinal stromal tumors, GISTs). Oncotarget. 8(67):111110–111118 Tamborini E, Pricl S, Negri T et al, 2006, Functional analyses and molecularmodeling of two c-Kitmutations responsible for imatinib secondary resistance in GIST patients. Oncogene. 25(45):6140– 6146 Guo T, Agaram NP, Wong GC et al, 2007, Sorafenib inhibits the imatinib-resistant KITT670I gatekeeper mutation in gastrointestinal stromal tumor. Clin Cancer Res 13(16):4874–4881 Antonescu CR, Besmer P, Guo Tet al, 2005, Acquired resistance to imatinib in gastrointestinal stromal tumor occurs through secondary gene mutation. Clin Cancer Res 11:4182–4190 Lau J, Zhou Q, Sutton SE et al, 2014, Inhibition of c-Kit is not required for reversal of hyperglycemia by imatinib in NOD mice. PLoS One 9(1):e84900 Pricl S, Fermeglia M, Ferrone M et al, 2005, T315I-mutated Bcr- Abl in chronic myeloid leukemia and imatinib: insights from a computational study. Mol Cancer Ther 4(8):1167–1174 Tutone M, Lauria A, Almerico AM, 2011, Study of the role of “gatekeeper” mutations V654A and T670I of c-kit kinase in the interaction withinhibitors by means mixed molecular dynamics/ docking approach. Bioinformation. 7(6):296–298 Ihle MA, Huss S, Jeske W et al, 2018, Expression of cell cycle regulators and frequency of TP53 mutations in high risk gastrointestinal stromal tumors prior to adjuvant imatinib treatment. PLoS One 13(2):e0193048 Haller F, Gunawan B, von Heydebreck A et al, 2005, Prognostic role of E2F1 and members of the CDKN2A network in gastrointestinal stromal tumors. Clin Cancer Res 11(18):6589–6597 Wang X, Lee RS, Alver BH et al, 2017, SMARCB1-mediated SWI/SNF complex function is essential for enhancer regulation. Nat Genet 49(2):289–295 Lu C, Allis CD, 2017, SWI/SNF complex in cancer. Nat Genet 49(2):178–179 Nakayama RT, Pulice JL, Valencia AM et al, 2017, SMARCB1 is required for widespread BAF complex-mediated activation of enhancers and bivalent promoters. Nat Genet 49(11):1613–1623 Kohashi K, Yamamoto H, Yamada Y et al, 2018, SWI/SNF chromatin-remodeling complex status in SMARCB1/INI1- preserved epithelioid sarcoma. Am J Surg Pathol 42(3):312–318 Smith MJ, Walker JA, Shen Y et al, 2012, Expression of SMARCB1, INI1, mutations in familial schwannomatosis. Hum Mol Genet 21(24):5239–5245 Beliakova-Bethell N, Mukim A, White CH et al, 2019, Histone deacetylase inhibitors induce complex host responses that contribute to differential potencies of these compounds in HIV reactivation. J Biol Chem 294(14):5576–5589 Mora-Blanco EL, Mishina Y, Tillman EJ et al, 2014, Activation of β-catenin/TCF targets following loss of the tumor suppressor SNF5. Oncogene. 33(7):933–938 Kim KH, Roberts CW, 2014, Mechanisms by which SMARCB1 loss drives rhabdoid tumor growth. Cancer Genetics 207(9):365–372 Tang CM, Lee TE, Syed SA et al, 2016, Hedgehog pathway dysregulation contributes to the pathogenesis of human gastrointestinal stromal tumors via GLI-mediated activation of KIT expression. Oncotarget. 7(48):78226–78241 Lo WW, Wunder JS, Dickson BC et al, 2014, Involvement and targeted intervention of dysregulated Hedgehog signaling in osteosarcoma. Cancer. 120(4):537–547 Szczepny A, Rogers S, Jayasekara WSN et al, 2017, The role of canonical and non-canonical Hedgehog signaling in tumor progression in a mouse model of small cell lung cancer. Oncogene. 36(39): 5544–5550 Pelczar P, Zibat A, van Dop WA et al, 2013, Inactivation of Patched1 in mice leads to development of gastrointestinal stromal-like tumors that express Pdgfrα but not kit. Gastroenterology 144(1):134–144. e6 Castell A, Yan Q, Fawkner K et al, 2018, A selective high affinity MYC-binding compound inhibits MYC:MAX interaction and MYC-dependent tumor cell proliferation. Sci Rep 8(1) Melnik S,Werth N, Boeuf S et al, 2019, Impact of c-MYC expression on proliferation, differentiation, and risk of neoplastic transformation of human mesenchymal stromal cells. Stem Cell Res Ther 10(1):73 Boyd SR, Young DW, 2019, Max-imizing the Attenuation of Myc Using Small Molecules. Trends Pharmacol Sci 40(9):608–612 Struntz NB, Chen A, Deutzmann A et al, 2019, Stabilization of the Max Homodimer with a Small Molecule Attenuates Myc-Driven Transcription. Cell Chem Biol 26(5):711–723.e14 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 91 EP 100 DI 10.1007/s12253-019-00770-6 PG 10 ER PT J AU Kocsis-Deak, B Arvai, K Balla, B Tobias, B Kohanka, A Jaray, B Horanyi, J Podani, J Takacs, I Putz, Zs Kosa, J Lakatos, P AF Kocsis-Deak, Barbara Arvai, Kristof Balla, Bernadett Tobias, Balint Kohanka, Andrea Jaray, Balazs Horanyi, Janos Podani, Janos Takacs, Istvan Putz, Zsuzsanna Kosa, Janos Lakatos, Peter TI Targeted Mutational Profiling and a Powerful Risk Score as Additional Tools for the Diagnosis of Papillary Thyroid Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary thyroid cancer; New hotspot panel; Multivariate statistical method; Risk score; Next generation sequencing ID Papillary thyroid cancer; New hotspot panel; Multivariate statistical method; Risk score; Next generation sequencing AB Nowadays, the complementary diagnostics based on the suspicious thyroid lesion specific mutational state analysis is indispensable in the clinical practice. We aimed to test and validate our novel 568-mutational hotspot panel (23 cancer-related genes) on papillary thyroid cancers (PTCs) and their tumor-free pairs to find the most powerful mutation pattern related to PTC. The sequencing method was carried on with Ion Torrent PGM on 67 thyroid tissue samples. The most commonly detected mutation was the BRAF c.1799 T > A in all non-classical PTC cases. We utilized a multivariate statistical method (CVA) to determine a discrimination score based on mutational data array and to assess malignancy risk. Based on variants, the BRAF gene has by far the highest indicative power, followed by TSHR and APC. We highlighted novel aspects of the mutational profile and genetic markers of PTC. CVA has correctly assigned most of the samples based on the mutation frequencies and different variables of the selected genes, with high analytical probabilities. The final goal is to set up a new comprehensive rule-in and rule-out test to support the clinical decision making mainly in inconclusive fine-needle aspiration biopsy cases. C1 [Kocsis-Deak, Barbara] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor Street 2/a, 1083 Budapest, Hungary. [Arvai, Kristof] PentaCore LaboratoryBudapest, Hungary. [Balla, Bernadett] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor Street 2/a, 1083 Budapest, Hungary. [Tobias, Balint] PentaCore LaboratoryBudapest, Hungary. [Kohanka, Andrea] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor Street 2/a, 1083 Budapest, Hungary. [Jaray, Balazs] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Horanyi, Janos] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Podani, Janos] Eotvos Lorand University, Institute of BiologyBudapest, Hungary. [Takacs, Istvan] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor Street 2/a, 1083 Budapest, Hungary. [Putz, Zsuzsanna] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor Street 2/a, 1083 Budapest, Hungary. [Kosa, Janos] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor Street 2/a, 1083 Budapest, Hungary. [Lakatos, Peter] Semmelweis University, 1st Department of Internal Medicine, Koranyi Sandor Street 2/a, 1083 Budapest, Hungary. RP Kocsis-Deak, B (reprint author), Semmelweis University, 1st Department of Internal Medicine, 1083 Budapest, Hungary. EM kocsisd.barbi@gmail.com CR Institute NC: Cancer stat facts: Thyroid cancer. Available from: https://seer.cancer.gov/statfacts/html/thyro.html. Accessed 12 Sept 2017 Tobias B, Halaszlaki C, Balla B, Kosa JP, Arvai K, Horvath P, Takacs I, Nagy Z, Horvath E, Horanyi J, Jaray B, Szekely E, Szekely T, Gyori G, Putz Z, Dank M, Valkusz Z, Vasas B, Ivanyi B, Lakatos P, 2016, Genetic alterations in Hungarian patients with papillary thyroid Cancer. Pathol Oncol Res 22:27–33 Carpi A, Mechanick JI, Saussez S, Nicolini A, 2010, Thyroid tumor marker genomics and proteomics: diagnostic and clinical implications. J Cell Physiol 224:612–619 Cancer Genome Atlas Research N: Integrated genomic characterization of papillary thyroid carcinoma. Cell 159: 676–690, 2014 Halaszlaki C, Tobias B, Balla B, Kosa JP, Horanyi J, Bolony E, Nagy Z, Speer G, Jaray B, Szekely E, Istok R, Szekely T, Putz Z, Dank M, Lakatos P, Takacs I, 2016, Predictive value of somatic mutations for the development of malignancy in thyroid nodules by cytopathology. Endocr Pract 22:1081–1087 Etzel M, Happel C, von Muller F, Ackermann H, Bojunga J, 2013, Grunwald F: [palpation and elastography of thyroid nodules in comparison]. Nuklearmedizin 52:97–100 Misiakos EP, Margari N, Meristoudis C, Machairas N, Schizas D, Petropoulos K, Spathis A, Karakitsos P, Machairas A, 2016, Cytopathologic diagnosis of fine needle aspiration biopsies of thyroid nodules. World J Clin Cases 4:38–48 Kargi AY, Bustamante MP, Gulec S, 2017, Genomic profiling of thyroid nodules: current role for ThyroSeq next-generation sequencing on clinical decision-making. Mol Imaging Radionucl Ther 26:24–35 Nikiforova MN, Nikiforov YE, 2009, Molecular diagnostics and predictors in thyroid cancer. Thyroid 19:1351–1361 Kim MI, Alexander EK, 2012, Diagnostic use of molecular markers in the evaluation of thyroid nodules. Endocr Pract 18: 796–802 Nikiforov YE, Carty SE, Chiosea SI, Coyne C, Duvvuri U, Ferris RL, GoodingWE, LeBeau SO, Ohori NP, Seethala RR, TublinME, Yip L, Nikiforova MN, 2015, Impact of the multi-gene ThyroSeq next-generation sequencing assay on Cancer diagnosis in thyroid nodules with Atypia of undetermined significance/follicular lesion of undetermined significance cytology. Thyroid 25:1217–1223 McIver B, Castro MR, Morris JC, Bernet V, Smallridge R, Henry M, Kosok L, Reddi H, 2014, An independent study of a gene expression classifier, Afirma, in the evaluation of cytologically indeterminate thyroid nodules. J Clin Endocrinol Metab 99:4069– 4077 Cha YJ, Koo JS, 2016, Next-generation sequencing in thyroid cancer. J Transl Med 14:322 Podani J: SYN-TAX 2000. Available from: http://ramet.elte.hu/ ~podani/SYN2000.html. Accessed 9 Aug 2017 Torregrossa L, Viola D, Sensi E, Giordano M, Piaggi P, Romei C, Materazzi G,Miccoli P, Elisei R, Basolo F, 2016, Papillary thyroid carcinoma with rare exon 15 BRAFmutation has indolent behavior: a single-institution experience. J Clin EndocrinolMetab 101:4413– 4420 Zou M, Baitei EY, Alzahrani AS, BinHumaid FS, Alkhafaji D, Al- Rijjal RA, Meyer BF, Shi Y, 2014, Concomitant RAS, RET/PTC, or BRAF mutations in advanced stage of papillary thyroid carcinoma. Thyroid 24:1256–1266 Sastre-Perona A, Santisteban P, 2012, Role of the wnt pathway in thyroid cancer. Front Endocrinol, Lausanne, 3:31 Latchford A, Volikos E, Johnson V, Rogers P, Suraweera N, Tomlinson I, Phillips R, Silver A, 2007, APC mutations in FAPassociated desmoid tumours are non-random but not 'just right'. Hum Mol Genet 16:78–82 Dalal KM, Moraitis D, Iwamoto C, Shaha AR, Patel SG, Ghossein RA, 2006, Clinical curiosity: cribriform-morular variant of papillary thyroid carcinoma. Head Neck 28:471–476 Gerber TS, Schad A, Hartmann N, Springer E, Zechner U,Musholt TJ, 2018, Targeted next-generation sequencing of cancer genes in poorly differentiated thyroid cancer. Endocr Connect 7:47–55 Smallridge RC,Marlow LA, Copland JA, 2009, Anaplastic thyroid cancer: molecular pathogenesis and emerging therapies. Endocr Relat Cancer 16:17–44 Xing M, 2010, Genetic alterations in the phosphatidylinositol-3 kinase/Akt pathway in thyroid cancer. Thyroid 20:697–706 PanW, Zhou L, GeM, Zhang B, Yang X, Xiong X, Fu G, Zhang J, Nie X, Li H, Tang X,Wei J, Shao M, Zheng J, Yuan Q, TanW,Wu C, Yang M, Lin D, 2016, Whole exome sequencing identifies lncRNA GAS8-AS1 and LPAR4 AS novel papillary thyroid carcinoma driver alternations. Hum Mol Genet 25:1875–1884 Liu C, Liu Z, Chen T, Zeng W, Guo Y, Huang T, 2016, TERT promoter mutation and its association with Clinicopathological features and prognosis of papillary thyroid Cancer: a meta-analysis. Sci Rep 6:36990 Alzahrani AS, Alsaadi R, Murugan AK, Sadiq BB, 2016, TERT promoter mutations in thyroid Cancer. Horm Cancer 7:165–177 Vinagre J, Almeida A, Populo H, Batista R, Lyra J, Pinto V, Coelho R, Celestino R, Prazeres H, Lima L, Melo M, da Rocha AG, Preto A, Castro P, Castro L, Pardal F, Lopes JM, Santos LL, Reis RM, Cameselle-Teijeiro J, Sobrinho-Simoes M, Lima J, Maximo V, Soares P, 2013, Frequency of TERT promoter mutations in human cancers. Nat Commun 4:2185 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 101 EP 108 DI 10.1007/s12253-019-00772-4 PG 8 ER PT J AU Tian, P Cheng, X Zhao, Z Zhang, Y Bao, C Wang, Y Cai, Sh Ma, G Huang, Y AF Tian, Panwen Cheng, Xiangyang Zhao, Zhengyi Zhang, Yuzi Bao, Celimuge Wang, Yanyan Cai, Shangli Ma, Guowei Huang, Ying TI Spectrum of Pathogenic Germline Mutations in Chinese Lung Cancer Patients through Next-Generation Sequencing SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Germline mutation; Next-generation sequencing; Lung cancer; DNA damage repair pathway ID Germline mutation; Next-generation sequencing; Lung cancer; DNA damage repair pathway AB Lung cancer is currently a leading cause of cancer-associated mortality worldwide. Despite the increasing evidences of variants that were associated with lung cancer risk, investigations of genetic factors and their roles in genetic susceptibility to lung cancer were limited. Here we systematically investigated the spectrum of pathogenic germline mutations in Chinese population with lung cancer. Genomic profiling of DNA was performed through next-generation sequencing (NGS) on tissue biopsy from 1764 Chinese lung cancer patients with a 381 cancer gene panel between January 01, 2017 and May 07, 2019. Patients with germline mutations were identified, and their clinical information were collected. Of 1764 patients with lung cancer, 67 (3.8%) patients were identified to carry pathogenic or likely pathogenic germline mutations in 25 cancer predisposition genes, with a frequency of 3.6% in lung adenocarcinoma (49/1349), 4.3% in squamous cell lung cancer (14/322), 5.6% in small cell lung cancer (4/72), and none in lung adenosquamous carcinoma (0/21), respectively. The highest pathogenic germline mutational prevalence were found in BRCA2 (0.79%), CHEK2 (0.40%), BRCA1 (0.34%), and TP53 (0.34%). Two splice mutations were reported for the first time in this study. Notably, a majority (85.5%) of the detected germline mutations fell in DNA damage repair pathways. C1 [Tian, Panwen] Sichuan University, West China Hospital, Department of Respiratory MedicineChengdu, Sichuan, China. [Cheng, Xiangyang] Guangzhou Medical College, First Affiliated HospitalGuangzhou, China. [Zhao, Zhengyi] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Zhang, Yuzi] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Bao, Celimuge] 3D Medicines Inc., The Information System DepartmentShanghai, China. [Wang, Yanyan] 3D Medicines Inc., The Information System DepartmentShanghai, China. [Cai, Shangli] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Ma, Guowei] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Thoracic Surgery, 510060 Guangzhou, China. [Huang, Ying] First Affiliated Hospital of Army Military Medical University, Department of Respiratory, 30 Gaotanyan Zhengjie, Shapingba District, 400038 Chongqing, China. RP Huang, Y (reprint author), First Affiliated Hospital of Army Military Medical University, Department of Respiratory, 400038 Chongqing, China. EM hga_83@sina.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6):394–424. , DOI 10.3322/caac. 21492 Tibaldi C, Giovannetti E, Vasile E, Boldrini L, Gallegos-Ruiz MI, Bernardini I, Incensati R, Danesi R, Cappuzzo F, Peters GJ, Fontanini G, 2011, Inherited germline T790Mmutation and somatic epidermal growth factor receptormutations in non-small cell lung cancer patients. J Thorac Oncol 6(2):395–396. , DOI 10. 1097/JTO.0b013e3182059a6f Lu S, Yu Y, Li Z, Yu R, Wu X, Bao H, Ding Y, Shao YW, Jian H, 2019, EGFR and ERBB2 Germline mutations in Chinese lung Cancer patients and their roles in genetic susceptibility to Cancer. J Thorac Oncol 14(4):732–736. , DOI 10.1016/j.jtho.2018. 12.006 Oxnard GR, Nguyen KS, Costa DB, 2014, Germline mutations in driver oncogenes and inherited lung cancer risk independent of smoking history. J Natl Cancer Inst 106(1):djt361. , DOI 10.1093/jnci/djt361 Parry EM, Gable DL, Stanley SE, Khalil SE, Antonescu V, Florea L, ArmaniosM(2017, Germline mutations in DNA repair genes in lung adenocarcinoma. J Thorac Oncol 12(11):1673–1678. https:// doi.org/10.1016/j.jtho.2017.08.011 Couto PP, Bastos-Rodrigues L, Schayek H,Melo FM, Lisboa RGC, MirandaDM,VilhenaA, Bale AE, Friedman E, DeMarco L, 2017, Spectrum of germline mutations in smokers and non-smokers in Brazilian non-small-cell lung cancer, NSCLC, patients. Carcinogenesis 38(11):1112–1118. , DOI 10.1093/carcin/ bgx089 Su D, Zhang D, Chen K, Lu J, Wu J, Cao X, Ying L, Jin Q, Ye Y, Xie Z, Xiong L, MaoW, Li F, 2017, High performance of targeted next generation sequencing on variance detection in clinical tumor specimens in comparison with current conventional methods. J Exp Clin Cancer Res 36(1):121. , DOI 10.1186/s13046-017- 0591-4 Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, Voelkerding K, Rehm HL, Committee ALQA, 2015, Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med 17(5):405– 424. , DOI 10.1038/gim.2015.30 Ismail IH, Davidson R, Gagne JP, Xu ZZ, Poirier GG, Hendzel MJ, 2014, Germline mutations in BAP1 impair its function in DNA double-strand break repair. Cancer Res 74(16):4282–4294. https:// doi.org/10.1158/0008-5472.CAN-13-3109 Gou LY, Wu YL, 2014, Prevalence of driver mutations in nonsmall- cell lung cancers in the People's Republic of China. Lung Cancer, Auckland, NZ, 5:1–9. , DOI 10.2147/LCTT. S40817 Oxnard GR, Miller VA, Robson ME, Azzoli CG, Pao W, Ladanyi M, Arcila ME, 2012, Screening for germline EGFR T790M mutations through lung cancer genotyping. J Thorac Oncol 7(6):1049– 1052. , DOI 10.1097/JTO.0b013e318250ed9d Daly MB, Pilarski R, Berry M, Buys SS, Farmer M, Friedman S, Garber JE, Kauff ND, Khan S, Klein C, Kohlmann W, Kurian A, Litton JK, Madlensky L, Merajver SD, Offit K, Pal T, Reiser G, Shannon KM, Swisher E, Vinayak S, Voian NC, Weitzel JN, Wick MJ, Wiesner GL, Dwyer M, Darlow S, 2017, NCCN guidelines insights: genetic/familial high-risk assessment: breast and ovarian, version 2.2017. J Natl Compr Cancer Netw 15(1):9–20 Gourley C, Balmana J, Ledermann JA, Serra V, Dent R, Loibl S, Pujade-Lauraine E, Boulton SJ, 2019, Moving from poly, ADPribose, polymerase inhibition to targeting DNA repair and DNA damage response in Cancer therapy. J Clin Oncol: JCO1802050. , DOI 10.1200/JCO.18.02050 Sen T, Rodriguez BL, Chen L, Corte CMD,Morikawa N, Fujimoto J, Cristea S, Nguyen T, Diao L, Li L, Fan Y, Yang Y, Wang J, Glisson BS, Wistuba II, Sage J, Heymach JV, Gibbons DL, Byers LA, 2019, Targeting DNA damage response promotes antitumor immunity through STING-mediated T-cell activation in small cell lung Cancer. Cancer Discov 9(5):646–661. , DOI 10.1158/ 2159-8290.CD-18-1020 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 109 EP 114 DI 10.1007/s12253-019-00771-5 PG 6 ER PT J AU Yu, L Yang, JSh AF Yu, Lu Yang, Jing Shou TI Primary Osteosarcoma of the Liver: Case Report and Literature Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Mesenchymal tumor; Hepatic; Osteoid; Extraskeletal osteosarcoma ID Mesenchymal tumor; Hepatic; Osteoid; Extraskeletal osteosarcoma AB Extraskeletal osteosarcoma is a rare, highly malignant, osteoid formation mesenchymal neoplasm in the absence of bone involvement, associated with exceptionally poor prognosis. It frequently arises in the soft tissues of the extremities or in the retroperitoneum, but rarely in visceral organ. We describe a primary osteosarcoma of the liver in a 70-year-old man who presented with an episode of fever, accompanied by abdominal discomfort, after an accident abdominal strike. Ultrasonography and computed tomography revealed a large heterogeneous mass with areas of dense calcification involving most of the right lobe of liver. Radiography did not show evidence of primary tumor or primary bone lesion at any other site. Histologically, the tumor showed an essentially similar appearance as osteosarcoma originating in the skeleton, comprised of polygonal or spindle shaped cells, along with abundant eosinophilic lace-like osteoids, or irregularly arranged bone trabeculae. Immunohistochemistry showed that the tumor cells were positive for vimentin, CD10, and focally for SMA and CD56, but negative for other lineage-specific markers. Thus, the findings favored a primary hepatic osteosarcoma. This patient received palliative chemotherapy to ease the signs of his sickness due to the large size of the tumor and he died 4 months later. C1 [Yu, Lu] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, No. 169 Chang Le Xi Road, 710032 Xi’an, Shaanxi, China. [Yang, Jing Shou] Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, No. 169 Chang Le Xi Road, 710032 Xi’an, Shaanxi, China. RP Yang, JSh (reprint author), Fourth Military Medical University, Xijing Hospital, State Key Laboratory of Cancer Biology and Department of Pathology, 710032 Xi’an, China. EM yangsj@fmmu.edu.cn CR Bane BL, Evans HL, Ro JY, Carrasco CH, Grignon DJ, Benjamin RS, Ayala AG, 1990, Extraskeletal osteosarcoma. A clinicopathologic review of 26 cases. Cancer 65(12):2762–2770 Chung EB, Enzinger FM, 1987, Extraskeletal osteosarcoma. Cancer 60(5):1132–1142 Lee JS, Fetsch JF, Wasdhal DA, Lee BP, Pritchard DJ, Nascimento AG, 1995, Areview of 40 patients with extraskeletal osteosarcoma. Cancer 76(11):2253–2259 Lidang Jensen M, Schumacher B, Myhre Jensen O, Steen Nielsen O, Keller J, 1998, Extraskeletal osteosarcomas: a clinicopathologic study of 25 cases. Am J Surg Pathol 22(5):588–594 McCarter MD, Lewis JJ, Antonescu CR, Brennan MF, 2000, Extraskeletal osteosarcoma: analysis of outcome of a rare neoplasm. Sarcoma 4(3):119–123 Allan CJ, Soule EH, 1971, Osteogenic sarcoma of the somatic soft tissues. Clinicopathologic study of 26 cases and review of literature. Cancer 27(5):1121–1133 Hoch M, Ali S, Agrawal S, Wang C, Khurana JS, 2013, Extraskeletal osteosarcoma: a case report and review of the literature. J Radiol Case Rep 7(7):15–23 Govender D, Rughubar KN, 1998, Primary hepatic osteosarcoma: case report and literature review. Pathology 30(3):323–325 Maynard JH, Fone DJ, 1969, Haemochromatosis with osteogenic sarcoma in the liver. Med J Aust 2(25):1260–1263 Sumiyoshi A, Niho Y, 1971, Primary osteogenic sarcoma of the liver–report of an autopsy case. Acta Pathol Jpn 21(2):305–312 von Hochstetter AR, Hattenschwiler J, Vogt M, 1987, Primary osteosarcoma of the liver. Cancer 60(9):2312–2317 Craig JR, Peters RL, Edmondson HA, 1989, Atlas of tumor pathology: tumors of the liver and intrahepatic bile ducts. Armed Forces Institute of Pathology, Washington, pp 223–255 Kitayama Y, Sugimura H, Arai T, Nagamatsu K, Kino I, 1995, Primary osteosarcoma arising from cirrhotic liver. Pathol Int 45(4):320–325 Krishnamurthy VN, Casillas VJ, Bejarano P, Saurez M, Franceschi D, 2004, Primary osteosarcoma of liver. European Journal of Radiology Extra 50(1):31–36 Park SH, Choi SB, Kim WB, Song TJ, 2009, Huge primary osteosarcoma of the liver presenting an aggressive recurrent pattern following surgical resection. J Dig Dis 10(3):231–235 Nawabi A, Rath S, Nissen N, Forscher C, Colquhoun S, Lee J, Geller S,Wong A, Klein AS, 2009, Primary hepatic osteosarcoma. J Gastrointest Surg 13(8):1550–1553 Jiang RD, Zhang ZL, Li T, 2015, Abdominal pain caused by spontaneous rupture of a liver tumor. Gastroenterology 149(1):35–36 Tamang TG, Shuster M, Chandra AB, 2016, Primary hepatic osteosarcoma: a rare cause of primary liver tumor. Clin Med Insights Case Rep 9:31–33 Mechtersheimer G, Moller P, 1989, Expression of the common acute lymphoblastic leukemia antigen, CD10, in mesenchymal tumors. Am J Pathol 134(5):961–965 Hasegawa T, Hirose T, Kudo E, Hizawa K, Usui M, Ishii S, 1991, Immunophenotypic heterogeneity in osteosarcomas. Hum Pathol 22(6):583–590 Ojima A, Sugiyama T, Takeda T, Hazama F, Nakakuki F, Uesugi Y et al, 1964, Six cases of rare malignant tumors of the liver. Acta Pathol Jpn 14:95–102 Leger-Ravet MB, Borgonovo G, Amato A, Lemaigre G, Franco D, 1996, Carcinosarcoma of the liver with mesenchymal differentiation: a case report. Hepato-Gastroenterology 43(7):255–259 Carter R, 1969, Hepatoblastoma in the adult. Cancer 23(1): 191–197 Haratake J, Horie A, 1991, An immunohistochemical study of sarcomatoid liver carcinomas. Cancer 68(1):93–97 Kakizoe S, Kojiro M, Nakashima T, 1987, Hepatocellular carcinoma with sarcomatous change. Clinicopathologic and immunohistochemical studies of 14 autopsy cases. Cancer 59(2):310–316 Stout AP, 1948, Mesenchymoma, the mixed tumor of mesenchymal derivatives. Ann Surg 127(2):278–290 Heukamp LC, Knoblich A, Rausch E, Friedrichs N, Schildhaus HU, Kahl P, Tismer R, Schneider B, Buttner R, Houshdaran F, 2007, Extraosseous osteosarcoma arising from the small intestinal mesentery. Pathol Res Pract 203(6):473–477 Sordillo PP, Hajdu SI, Magill GB, Golbey RB, 1983, Extraosseous osteogenic sarcoma. A review of 48 patients. Cancer 51(4):727–734 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 115 EP 120 DI 10.1007/s12253-018-0483-8 PG 6 ER PT J AU Sun, X Guo, Sh AF Sun, Xiaojun Guo, Shilei TI The Prognostic and Predictive Value of Dihydropyrimidine Dehydrogenase-Related Indicators in Clinical Outcomes of Chemotherapy in Colorectal Cancer Patients: a Systematic Review and Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Dihydropyrimidine dehydrogenase; Thymidylate phosphorylase; Colorectal cancer; Meta-analysis ID Dihydropyrimidine dehydrogenase; Thymidylate phosphorylase; Colorectal cancer; Meta-analysis AB Colorectal cancer (CRC) is the third most common cancer worldwide. Predictive biomarkers are needed to predict patients’ outcomes and to select a chemotherapy regimen. We assessed whether dihydropyrimidine dehydrogenase (DPD)-related indicators can predict CRC patients’ outcomes. We searched the studies in PubMed, EmBase, and the Cochrane Library up to March 4, 2018.We mainly analyzed different CRC patients’ outcomes according to specific DPD-related indicators. Twenty-five articles were included in the meta-analysis. The results showed that for disease-free survival (DFS), low DPD expression was significantly superior to high expression (I2 = 72%; HR: 1.59; 95%CI: 1.21–2.09; p = 0.001). However, this result had a potential publication bias (Begg’s test: p = 0.007; Egger’s test: p = 0.004). Among patients treated with chemotherapy, a high thymidylate phosphorylase (TP)/DPD ratio was advantageous for DFS (I2 = 63.7%; HR: 0.65; 95%CI: 0.46–0.92; p = 0.015), and this result did not have a publication bias. For overall survival (OS), low DPD expression was superior to high expression (I2 = 74.4%;HR: 2.11; 95%CI: 1.48–3.00; p < 0.001), although this result had a publication bias (Egger’s test: p = 0.003; Begg’s test: p = 0.010). There was no difference in OS according to the TP/DPD ratio (I2 = 0%; HR: 0.92; 95%CI: 0.75–1.13; p = 0.420). DFS and OS were better in CRC patients with low DPD expression than in those with high DPD expression. However, because of publication bias, more DPD indicator-related studies, especially with negative results, are still needed. Patients with a high TP/DPD ratio have better DFS but not OS. C1 [Sun, Xiaojun] Nanjing Qi-xia Xi-gang community health service centers, Inpatients department, 210033 Nanjing, Jiangsu, China. [Guo, Shilei] Nanjing Regenerative Medicine Engineering and Technology Research Center, R&D department, No.108, Ganjiabian East, Qixia District, 210046 Nanjing, Jiangsu, China. RP Guo, Sh (reprint author), Nanjing Regenerative Medicine Engineering and Technology Research Center, R&D department, 210046 Nanjing, China. EM guoshilei1986@aliyun.com CR Grothey A, Sobrero AF, Shields AF, Yoshino T, Paul J, Taieb J, Souglakos J, Shi Q, Kerr R, Labianca R, Meyerhardt JA, Vernerey D, Yamanaka T, Boukovinas I, Meyers JP, Renfro LA, Niedzwiecki D,Watanabe T, Torri V, Saunders M, Sargent DJ, Andre T, Iveson T, 2018, Duration of adjuvant chemotherapy for stage III colon cancer. N Engl J Med 378(13):1177–1188 Wu DM,Wang YJ, Fan SH, Zhuang J, Zhang ZF, Shan Q, Han XR, Wen X, Li MQ, Hu B, Sun CH, Bao YX, Xiao HJ, Yang L, Lu J, ZhengYL, 2017, Networkmeta-analysis of the efficacy of first-line chemotherapy regimens in patients with advanced colorectal cancer. Oncotarget 8(59):100668–100677 Iveson TJ, Kerr RS, Saunders MP, Cassidy J, Hollander NH, Tabernero J, Haydon A, Glimelius B, Harkin A, Allan K, McQueen J, Scudder C, Boyd KA, Briggs A, Waterston A, Medley L, Wilson C, Ellis R, Essapen S, Dhadda AS, Harrison M, Falk S, Raouf S, Rees C, Olesen RK, Propper D, Bridgewater J, Azzabi A, Farrugia D, Webb A, Cunningham D, Hickish T, Weaver A, Gollins S, Wasan HS, Paul J, 2018, 3 versus 6 months of adjuvant oxaliplatin-fluoropyrimidine combination therapy for colorectal cancer, SCOT): an international, randomised, phase 3, non-inferiority trial. Lancet Oncol 19(4):562–578 Ofverholm A, Arkblad E, Skrtic S, Albertsson P, Shubbar E, Enerback C, 2010, Two cases of 5-fluorouracil toxicity linked with gene variants in the DPYD gene. Clin Biochem 43(3):331–334 Cubero DI, Del Giglio A, 2013, Tegafur-uracil, UFT, in lower doses is safe for the treatment of colorectal cancer in patients with partial dihydropyrimidine dehydrogenase deficiency: a proof of principle. Ther Adv Med Oncol 5(1):93–94 van Kuilenburg AB, Hausler P, Schalhorn A, Tanck MW, Proost JH, Terborg C et al, 2012, Evaluation of 5-fluorouracil pharmacokinetics in cancer patients with a c.1905+1G>A mutation in DPYD by means of a Bayesian limited sampling strategy. Clin Pharmacokinet 51(3):163–174 Higgins JP, Altman DG, Gotzsche PC, Juni P, Moher D, Oxman AD et al, 2011, The Cochrane Collaboration's tool for assessing risk of bias in randomised trials. BMJ 343:d5928 Stang A, 2010, Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in metaanalyses. Eur J Epidemiol 25(9):603–605 Ades AE, Lu G, Higgins JP, 2005, The interpretation of randomeffects meta-analysis in decision models. Med Decis Mak 25(6): 646–654 Duval S, Tweedie R, 2000, Trim and fill: a simple funnel-plotbased method of testing and adjusting for publication bias in meta- analysis. Biometrics 56(2):455–463 Koda K, Miyauchi H, Kosugi C, Kaiho T, Takiguchi N, Kobayashi S, Maruyama T, Matsubara H,, Boso Clinical Oncology Group,, 2016, Tumor 5-FU-related mRNA expression and efficacy of Oral Fluoropyrimidines in adjuvant chemotherapy of colorectal Cancer. Anticancer Res 36(10):5325–5331 Schmoll HJ, Tabernero J, Maroun J, de Braud F, Price T, Van Cutsem E et al, 2015, Capecitabine plus oxaliplatin compared with fluorouracil/Folinic acid as adjuvant therapy for stage III colon cancer: final results of the NO16968 randomized controlled phase III trial. J Clin Oncol 33(32):3733–3740 Mori T, Ohue M, Takii Y, Hashizume T, Kato T, Kotake K et al, 2013, Factors predicting the response to oral fluoropyrimidine drugs: a phase II trial on the individualization of postoperative adjuvant chemotherapy using oral fluorinated pyrimidines in stage III colorectal cancer treated by curative resection, ACT-01 study). Oncol Rep 29(2):437–444 Soong R, Shah N, Salto-Tellez M, Tai BC, Soo RA, Han HC et al, 2008, Prognostic significance of thymidylate synthase, dihydropyrimidine dehydrogenase and thymidine phosphorylase protein expression in colorectal cancer patients treated with or without 5-fluorouracil-based chemotherapy. Ann Oncol 19(5):915–919 Lassmann S, Hennig M, Rosenberg R, Nahrig J, Schreglmann J, Krause F et al, 2006, Thymidine phosphorylase, dihydropyrimidine dehydrogenase and thymidylate synthase mRNA expression in primary colorectal tumors-correlation to tumor histopathology and clinical follow-up. Int J Color Dis 21(3):238–247 Tsuji T, Sawai T, Takeshita H, Nakagoe T, Hidaka S, Atsushi N et al, 2004, Tumor dihydropyrimidine dehydrogenase in stage II and III colorectal cancer: low level expression is a beneficial marker in oral-adjuvant chemotherapy, but is also a predictor for poor prognosis in patients treated with curative surgery alone. Cancer Lett 204(1):97–104 Kataoka K, Kanazawa A, Nakajima A, Yamaguchi A, Arimoto A, 2015, Prognostic value of biomarkers in metastatic colorectal cancer patients. J Surg Res 194(2):343–350 Shigeta K, Ishii Y, Hasegawa H, Okabayashi K, Kitagawa Y, 2014, Evaluation of 5-fluorouracil metabolic enzymes as predictors of response to adjuvant chemotherapy outcomes in patients with stage II/III colorectal cancer: a decision-curve analysis. World J Surg 38(12):3248–3256 Koumarianou A, Tzeveleki I, Mekras D, Eleftheraki AG, Bobos M, Wirtz R, Fountzilas E, Valavanis C, Xanthakis I, Kalogeras KT, Basdanis G, Pentheroudakis G, Kotoula V, Fountzilas G, 2014, Prognostic markers in early-stage colorectal cancer: significance of TYMS mRNA expression. Anticancer Res 34(9):4949–4962 Ochiai T, Umeki M, Miyake H, Iida T, Okumura M, Ohno K et al, 2014, Impact of 5-fluorouracil metabolizing enzymes on chemotherapy in patients with resectable colorectal cancer. Oncol Rep 32(3):887–892 Donada M, Bonin S, Nardon E, De Pellegrin A, Decorti G, StantaG, 2011, Thymidilate synthase expression predicts longer survival in patients with stage II colon cancer treated with 5-flurouracil independently of microsatellite instability. J Cancer Res Clin Oncol 137(2):201–210 Jensen SA, Vainer B, Kruhoffer M, Sorensen JB, 2009, Microsatellite instability in colorectal cancer and association with thymidylate synthase and dihydropyrimidine dehydrogenase expression. BMC Cancer 9:25 Gustavsson B, Kaiser C, Carlsson G, Wettergren Y, Odin E, Lindskog EB, Niyikiza C, Ma D, 2009, Molecular determinants of efficacy for 5-FU-based treatments in advanced colorectal cancer: mRNA expression for 18 chemotherapy-related genes. Int J Cancer 124(5):1220–1226 Yamada H, Iinuma H, Watanabe T, 2008, Prognostic value of 5- fluorouracil metabolic enzyme genes in Dukes' stage B and C colorectal cancer patients treated with oral 5-fluorouracil-based adjuvant chemotherapy. Oncol Rep 19(3):729–735 Tokunaga Y, Sasaki H, Saito T, 2007, Clinical role of orotate phosphoribosyl transferase and dihydropyrimidine dehydrogenase in colorectal cancer treated with postoperative fluoropyrimidine. Surgery 141(3):346–353 Ochiai T, Nishimura K, Noguchi H, Kitajima M, Tsukada A, Watanabe E, Nagaoka I, Futagawa S, 2006, Prognostic impact of orotate phosphoribosyl transferase among 5-fluorouracil metabolic enzymes in resectable colorectal cancers treated by oral 5- fluorouracil-based adjuvant chemotherapy. Int J Cancer 118(12): 3084–3088 Hotta T, Takifuji K, Taniguchi K, Sahara M, Yokoyama S, Matsuda K, Higashiguchi T, Tominaga T, Oku Y, Yamaue H, 2006, The relat ionship between survival and the expression of dihydropyrimidine dehydrogenase in patients with colorectal cancer. Oncol Rep 16(1):177–182 Ciaparrone M, Quirino M, Schinzari G, Zannoni G, Corsi DC, Vecchio FM, Cassano A, la Torre G, Barone C, 2006, Predictive role of thymidylate synthase, dihydropyrimidine dehydrogenase and thymidine phosphorylase expression in colorectal cancer patients receiving adjuvant 5-fluorouracil. Oncology 70(5):366–377 Westra JL, Hollema H, Schaapveld M, Platteel I, Oien KA, Keith WN, Mauritz R, Peters GJ, Buys CHCM, Hofstra RMW, Plukker JTM, 2005, Predictive value of thymidylate synthase and dihydropyrimidine dehydrogenase protein expression on survival in adjuvantly treated stage III colon cancer patients. Ann Oncol 16(10):1646–1653 Tsuji T, Sawai T, Takeshita H, Nakagoe T, Hidaka S, Yamaguchi H, Yasutake T, Nagayasu T, Tagawa Y, 2004, Tumor dihydropyrimidine dehydrogenase expression is a useful marker in adjuvant therapy with oral fluoropyrimidines after curative resection of colorectal cancer. Cancer Chemother Pharmacol 54(6):531– 536 Oi K, Makino M, Ozaki M, Takemoto H, Yamane N, Nakamura S, Ikeguchi M, Kaibara N, 2004, Immunohistochemical dihydropyrimidine dehydrogenase expression is a good prognostic indicator for patients with Dukes' C colorectal cancer. Anticancer Res 24(1):273–279 Kornmann M, Schwabe W, Sander S, Kron M, Strater J, Polat S et al, 2003, Thymidylate synthase and dihydropyrimidine dehydrogenase mRNA expression levels: predictors for survival in colorectal cancer patients receiving adjuvant 5-fluorouracil. Clin Cancer Res 9(11):4116–4124 IchikawaW, Uetake H, Shirota Y, Yamada H, Takahashi T, Nihei Z e t a l, 2 0 0 3 , Bot h g e n e e x p r e s s i o n f o r or o t a t e phosphoribosyltransferase and its ratio to dihydropyrimidine dehydrogenase influence outcome following fluoropyrimidine-based chemotherapy for metastatic colorectal cancer. Br J Cancer 89(8): 1486–1492 Ichikawa W, Uetake H, Shirota Y, Yamada H, Nishi N, Nihei Z, Sugihara K, Hirayama R, 2003, Combination of dihydropyrimidine dehydrogenase and thymidylate synthase gene expressions in primary tumors as predictive parameters for the efficacy of fluoropyrimidine-based chemotherapy for metastatic colorectal cancer. Clin Cancer Res 9(2):786–791 Ikeguchi M, Makino M, Kaibara N, 2002, Thymidine phosphorylase and dihydropyrimidine dehydrogenase activity in colorectal carcinoma and patients prognosis. Langenbeck's Arch Surg 387(5–6):240–245 Yoshisue K, Kanie S, Nishimura T, Chikamoto J, Nagayama S, 2009, Effect of dimethylnitrosamine-induced liver dysfunction on the pharmacokinetics of 5-fluorouracil after administration of S-1, an antitumour drug, to rats. J Pharm Pharmacol 61(12):1643– 1651 Bronckaers A, Gago F, Balzarini J, Liekens S, 2009, The dual role of thymidine phosphorylase in cancer development and chemotherapy. Med Res Rev 29(6):903–953 Elamin YY, Rafee S, Osman N, O Byrne KJ, Gately K, 2016, Thymidine phosphorylase in cancer; enemy or friend? Cancer Microenviron 9(1):33–43 Ranieri G, Grammatica L, Patruno R, Zito AF, Valerio P, Iacobellis S et al, 2007, A possible role of thymidine phosphorylase expression and 5-fluorouracil increased sensitivity in oropharyngeal cancer patients. J Cell Mol Med 11(2): 362–368 Amatori F, Di Paolo A, Del Tacca M, Fontanini G, Vannozzi F, Boldrini L et al, 2006, Thymidylate synthase, dihydropyrimidine dehydrogenase and thymidine phosphorylase expression in colorectal cancer and normal mucosa in patients. Pharmacogenet Genomics 16(11):809–816 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 121 EP 131 DI 10.1007/s12253-018-00563-3 PG 11 ER PT J AU Roberto, MG Delsin, EL Vieira, MG Silva, OM Hakime, GR Gava, FN Engel, EE Scrideli, AC Tone, GL Brassesco, SM AF Roberto, Molinari Gabriela Delsin, Elis Lara Vieira, Maciel Gabriela Silva, Oliveira Marcela Hakime, Guedes Rodrigo Gava, Fabricio Nelson Engel, Eduard Edgard Scrideli, Alberto Carlos Tone, Gonzaga Luiz Brassesco, Sol Maria TI ROCK1-PredictedmicroRNAs Dysregulation Contributes to Tumor Progression in Ewing Sarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Rock; microRNA; Ewing sarcoma; Invasion; Motility ID Rock; microRNA; Ewing sarcoma; Invasion; Motility AB Over the last decade, the rho-associated kinases and several metastasis-associated microRNAs have emerged as important contributors of tumor invasion. However, despite prominence, our understanding of their involvement in the metastatic potential of Ewing Sarcoma (EWS) is incomplete. The expression profiles of ROCK1 or ROCK2 and miR- 124-3p, miR-138-5p, miR-139-5p, miR-335-5p and miR-584-5p (all of which were previously predicted or validated to regulate these kinases) were evaluated through qRT-PCR and associated with clinical parameters. In vitro assays to evaluate colony formation and invasion/migration capacities were performed on SK-ES-1 cells transfected with premiR mimics. ROCK1 expression was significantly reduced in EWS tissues, though there was no association with pathological parameters. miR-124-3p, miR-139-5p and miR-335-3p were also found significantly downregulated and positively correlated with ROCK1. Stratification indicated an association between lower levels of miR-139-5p and miR- 584-5p with disease progression (p < 0.05), while reduced expression of the former and miR-124-3p were associated with reduced survival. In vitro miR-139-5p overexpression yielded inconsistent results: while mir-139-5p restoration significantly reduced invasion, the clonogenic capacity of cells was increased. Our study demonstrated that downregulation of miR-124-3p, miR-139-5p and miR-584-5p are associated with disease progression in EWS and may serve as a risk assessment biomarkers though, as seen for mir-139-5p, their specific role remain to be elucidated for considering tailoring treatment options. C1 [Roberto, Molinari Gabriela] University of Sao Paulo, Ribeirao Preto School of Medicine, Regional Blood CenterSao Paulo, Brazil. [Delsin, Elis Lara] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of GeneticsSao Paulo, Brazil. [Vieira, Maciel Gabriela] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of GeneticsSao Paulo, Brazil. [Silva, Oliveira Marcela] University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell BiologySao Paulo, Brazil. [Hakime, Guedes Rodrigo] University of Sao Paulo, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, Department of BiologySao Paulo, Brazil. [Gava, Fabricio Nelson] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of Biomechanics, Medicine and Rehabilitation of the Locomotor SystemSao Paulo, Brazil. [Engel, Eduard Edgard] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of Biomechanics, Medicine and Rehabilitation of the Locomotor SystemSao Paulo, Brazil. [Scrideli, Alberto Carlos] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of PediatricsSao Paulo, Brazil. [Tone, Gonzaga Luiz] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of PediatricsSao Paulo, Brazil. [Brassesco, Sol Maria] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of Biomechanics, Medicine and Rehabilitation of the Locomotor SystemSao Paulo, Brazil. RP Brassesco, SM (reprint author), University of Sao Paulo, Ribeirao Preto School of Medicine, Department of Biomechanics, Medicine and Rehabilitation of the Locomotor System, Sao Paulo, Brazil. EM solbrassesco@usp.br CR Gonzales EHH, Betancourt GM, Martinez OQ, Cabezaz IH, 2013, Sarcoma de Ewing. AMC 17:n5:623–640 Rodriguez-Galindo C, Spunt SL, Pappo AS, 2003, Treatment of Ewing sarcoma family of tumors: current status and outlook for the future. Med Pediatr Oncol 40:276–287 Kim JY, Nam JK, Lee SA, Lee MS, Cho SK, Park ZYet al, 2011, Proteasome inhibition causes epithelial-mesenchymal transition upon TM4SF5 expression. J Cell Biochem 112:782–792 Tseliou M, Al-Qahtani A, Alarifi S, Alkahtani SH, Stournaras C, Sourvinos G, 2016, The role of RhoA, RhoB and RhoCGTPases in cell morphology, proliferation and migration in human cytomegalovirus, HCMV, infected glioblastoma cells. Cell Physiol Biochem 38:94–109 Amano M, Nakayama M, Kaibuchi K, 2010, Rho-kinase/ROCK. A key regulator of the cytoskeleton and cell polarity. Cytoskeleton, Hoboken, 67:545–554 Kamai T, Tsujii T, Arai K, Takagi K, Asami H, Ito Y et al, 2003, Significant association of rho/ROCK pathway with invasion and metastasis of bladder cancer. Clin Cancer Res 9: 2632–2641 Bottino J, Gelaleti GB, Maschio LB, Jardim-Perassi BV, de Campos Zuccari DA, 2014, Immunoexpression of ROCK-1 and MMP-9 as prognostic markers in breast cancer. Acta Histochem 116:1367–1373 Wong CC, Wong CM, Tung EK, SL A, Lee JM, Poon RT et al, 2011, The microRNAmiR-139 suppressesmetastasis and progression of hepatocellular carcinoma by down-regulating rho-kinase 2. Gastroenterology 140:322–331 Akagi EM, Lavorato-Rocha AM, 2014, Maia BeM, Rodrigues IS, Carvalho KC, Stiepcich MM et al. ROCK1 as a novel prognostic marker in vulvar cancer. BMC Cancer 14:822 Raimo M, Orso F, Grassi E, Cimino D, Penna E, De Pitta C et al, 2016, miR-146a exerts differential effects on melanoma growth and Metastatization. Mol Cancer Res 14:548–562 Loirand G, 2015, Rho kinases in health and disease: from basic science to translational research. Pharmacol Rev 67:1074–1095 SH L, Jiang XJ, Xiao GL, Liu DY, Yuan XR, 2014, miR-124a restoration inhibits glioma cell proliferation and invasion by suppressing IQGAP1 and β-catenin. Oncol Rep 32:2104–2110 Agirre X, Vilas-Zornoza A, Jimenez-Velasco A, Martin-Subero JI, Cordeu L, Garate L et al, 2009, Epigenetic silencing of the tumor suppressor microRNA Hsa-miR-124a regulates CDK6 expression and confers a poor prognosis in acute lymphoblastic leukemia. Cancer Res 69:4443–4453 Furuta M, Kozaki KI, Tanaka S, Arii S, Imoto I, Inazawa J, 2010, miR-124 and miR-203 are epigenetically silenced tumor-suppressive microRNAs in hepatocellular carcinoma. Carcinogenesis 31:766–776 Pierson J, Hostager B, Fan R, Vibhakar R, 2008, Regulation of cyclin dependent kinase 6 by microRNA 124 in medulloblastoma. J Neuro-Oncol 90:1–7 Wilting SM, van Boerdonk RA, Henken FE, Meijer CJ, Diosdado B, Meijer GA et al, 2010, Methylation-mediated silencing and tumour suppressive function of hsa-miR-124 in cervical cancer. Mol Cancer 9:167 Chen X, He D, Dong XD, Dong F, Wang J, Wang L et al, 2013, MicroRNA-124a is epigenetically regulated and acts as a tumor suppressor by controlling multiple targets in uveal melanoma. Invest Ophthalmol Vis Sci 54:2248–2256 CB H, Li QL, JF H, Zhang Q,Xie JP, 2014, Deng.miR-124 inhibits growth and invasion of gastric cancer by targeting ROCK1. Asian Pac J Cancer Prev 15:6543–6546 An L, Liu Y, Wu A, Guan Y, 2013, microRNA-124 inhibits migration and invasion by down-regulating ROCK1 in glioma. PLoS One 8:e69478 Ueno K, Hirata H, Shahryari V, Chen Y, Zaman MS, Singh K et al, 2011, Tumour suppressor microRNA-584 directly targets oncogene Rock-1 and decreases invasion ability in human clear cell renal cell carcinoma. Br J Cancer 104:308–315 Xiang J, Wu Y, Li DS, Wang ZY, Shen Q, Sun TQ et al, 2015, miR-584 suppresses invasion and cell migration of thyroid carcinoma by regulating the target oncogene ROCK1. Oncol Res Treat 38:436–440 Xue H, Guo X, Han X, Yan S, Zhang J, Xu S et al, 2016, MicroRNA-584-3p, a novel tumor suppressor and prognostic marker, reduces the migration and invasion of human glioma cells by targeting hypoxia-induced ROCK1. Oncotarget 7:4785–4805 Hua W, Sa KD, Zhang X, Jia LT, Zhao J, Yang AG et al, 2015, MicroRNA-139 suppresses proliferation in luminal type breast cancer cells by targeting topoisomerase II alpha. Biochem Biophys Res Commun 463:1077–1083 Luo HN, Wang ZH, Sheng Y, Zhang Q, Yan J, Hou J et al, 2014, MiR-139 targets CXCR4 and inhibits the proliferation and metastasis of laryngeal squamous carcinoma cells. Med Oncol 31:789 Song M, Yin Y, Zhang J, Zhang B, Bian Z, Quan C et al, 2014, MiR-139-5p inhibitsmigration and invasion of colorectal cancer by downregulating AMFR and NOTCH1. Protein Cell 5:851–861 Sun C, Sang M, Li S, Sun X, Yang C, Xi Yet al, 2015, Hsa-miR- 139-5p inhibits proliferation and causes apoptosis associated with down-regulation of c-met. Oncotarget 6:39756–39792 Krowiorz K, Ruschmann J, Lai C, Ngom M, Maetzig T, Martins V et al, 2016, MiR-139-5p is a potent tumor suppressor in adult acute myeloid leukemia. Blood Cancer J 6:e508 Bao W, Fu HJ, Xie QS, Wang L, Zhang R, Guo ZY et al. HER2 interacts with CD44 to up-regulate CXCR4 via epigenetic silencing of microRNA-139 in gastric cancer cells. Gastroenterology. 2011; 141:2076–2087.e2076 Buhnemann C, Li S, Yu H, Branford WH, Schafer KL, Llombart- Bosch A et al, 2014, Quantification of the heterogeneity of prognostic cellular biomarkers in ewing sarcoma using automated image and random survival forest analysis. PLoS One 9:e107105 Komuro H, Saihara R, Shinya M, Takita J, Kaneko S, Kaneko M et al, 2007, Identification of side population cells, stem-like cell population, in pediatric solid tumor cell lines. J Pediatr Surg 42: 2040–2045 Yang M, Zhang R, Yan M, Ye Z, LiangW, Luo Z, 2010, Detection and characterization of side population in Ewing's sarcoma SK-ES- 1 cells in vitro. Biochem Biophys Res Commun 391:1062–1066 Gao CF, Xie Q, Su YL, Koeman J, Khoo SK, Gustafson M et al, 2005, Proliferation and invasion: plasticity in tumor cells. Proc Natl Acad Sci USA 102:10528–10533 Hecht I, Natan S, Zaritsky A, Levine H, Tsarfaty I, Ben-Jacob E, 2015, The motility-proliferation-metabolism interplay during metastatic invasion. Sci Rep 5:13538 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 133 EP 139 DI 10.1007/s12253-017-0374-4 PG 7 ER PT J AU Ghali, MR Al-Mutawa, AM Ebrahim, HB Jrah, HH Zaied, S Bhiri, H Hmila, F Mahjoub, T Almawi, YW AF Ghali, M Rabeb Al-Mutawa, A Maryam Ebrahim, H Bashayer Jrah, H Hanen Zaied, Sonia Bhiri, Hanen Hmila, Fahmi Mahjoub, Touhami Almawi, Y Wassim TI Progesterone Receptor (PGR) Gene Variants Associated with Breast Cancer and Associated Features: a Case-Control Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Haplotypes; Mutation; Progesterone; Progesterone receptor ID Breast cancer; Haplotypes; Mutation; Progesterone; Progesterone receptor AB Insofar as altered estrogen receptor-progesterone receptor (PR) expression contribute to breast cancer pathogenesis, previous studies examined the association of genetic variation in PR gene (PGR) with breast cancer, but with mixed outcome. We evaluated the association between PGR variants, and breast cancer and associated features. A retrospective case-control study involving 183 female breast cancer patients, and 222 control women. PGR genotyping was done by real-time PCR. Minor allele frequencies of rs1042838, rs590688, and rs10895068 PGR gene polymorphisms were significantly higher in breast cancer patients compared to controls. Patients carrying rs1042838 G/T, rs590688 C/C, and rs10895068 G/A genotypes had higher risk of breast cancer, while carriage of rs3740753 G/G genotype was associated with marginal reduction in breast cancer risk. In addition, carriage of rs1042839, rs3740753, and rs10895068 minor allele was associated with Her2 status, while rs3740753 and rs10895068 were associated with effective hormone replacement therapy. Furthermore, carriage of rs10895068 minor allele in breast cancer women were also associated with age at first pregnancy, hormone receptor (RH) status, and previous use of oral contraceptives. PGR haploview analysis documented moderate-strong linkage disequilibrium (non-random association of alleles at different loci) between 7 of the 8 tested PGR SNPs, thus allowing construction of 7-locus PGR haplotypes. Two haplotypes, ATGCCGA and GTGCCGA, both containing rs590688, were positively associated with breast cancer, thus assigning a breast cancer-susceptible nature to these haplotypes. PGR rs1042838, rs590688, and rs10895068, and ATGCCGA and GTGCCGA haplotypes are related with increased breast cancer susceptibility in Tunisian women. C1 [Ghali, M Rabeb] University of Monastir, Faculty of Pharmacy of Monastir, Laboratory of Human Genome and Multifactorial Diseases (LR12ES07)Monastir, Tunisia. [Al-Mutawa, A Maryam] El Manar University, Faculty of Sciences of Tunis, Manar II, 2092 Tunis, Tunisia. [Ebrahim, H Bashayer] El Manar University, Faculty of Sciences of Tunis, Manar II, 2092 Tunis, Tunisia. [Jrah, H Hanen] University of Monastir, Faculty of Pharmacy of Monastir, Laboratory of Human Genome and Multifactorial Diseases (LR12ES07)Monastir, Tunisia. [Zaied, Sonia] CHU Fattouma Bourguiba, Department of Clinical OncologyMonastir, Tunisia. [Bhiri, Hanen] CHU Fattouma Bourguiba, Department of Clinical OncologyMonastir, Tunisia. [Hmila, Fahmi] CHU Farhat Hached, Department of SurgerySousse, Tunisia. [Mahjoub, Touhami] University of Monastir, Faculty of Pharmacy of Monastir, Laboratory of Human Genome and Multifactorial Diseases (LR12ES07)Monastir, Tunisia. [Almawi, Y Wassim] El Manar University, Faculty of Sciences of Tunis, Manar II, 2092 Tunis, Tunisia. RP Almawi, YW (reprint author), El Manar University, Faculty of Sciences of Tunis, 2092 Tunis, Tunisia. EM wassim.almawi@outlook.com CR Fan L, Goss PE, Strasser-Weippl K, 2015, Current status and future projections of breast cancer in Asia. Breast Care, Basel, 10:372– 378. , DOI 10.1007/s10552-014-0491-2 Abdulrahman GO Jr, Rahman GA, 2012, Epidemiology of breast cancer in Europe and Africa. J Cancer Epidemiol 2012:915610. , DOI 10.1155/2012/915610 Mojgan H, Massoud H, Ahmad E, 2012, ERCC1 intron 1 was associated with breast cancer risk. Arch Med Sci 8:655–658. , DOI 10.5114/aoms.2012.30289 GallegosMP, Figuera LE, RamosMC, Salas E, PueblaAM, Peralta V, Gaecia JE, Davalos IP, Zuniga GM, 2014, The association between the 844ins68 polymorphism in the CBS gene and breast cancer. Arch Med Sci 10:1214–1224. , DOI 10.5114/ aoms.2014.47830 Gomez Flores-Ramos L, Escoto-De Dios A, Puebla-Perez AM, Figuera-Villanueva LE, Ramos-Silva A, Ramirez-Patino R, Delgado-Saucedo JI, Salas-Gonzalez E et al, 2013, Association of the tumor necrosis factor-alpha –308G>A polymorphism with breast cancer in Mexican women. Genet Mol Res 12:5680–5693. , DOI 10.4238/2013 Xu YL, Sun Q, Shan GL, Zhang J, Liao HB, Li SY, Jiang J, Shao ZMet al, 2012)A case-control study on risk factors of breast cancer in China. Arch Med Sci 8:303–309. , DOI 10.5114/aoms. 2012.28558 Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DTet al, 2000)Molecular portraits of human breast tumours. Nature 406:747–752 Nyante SJ, Gammon MD, Kaufman JS, Bensen JT, Lin DY, Barnholtz-Sloan JS, Hu Y, He Q, Luo J, Millikan RC, 2015, Genetic variation in estrogen and progesterone pathway genes and breast cancer risk: an exploration of tumor subtype-specific effects. Cancer Causes Control 26:121–131. , DOI 10. 1007/s10552-014-0491-2 Eiro N, Fernandez-Garcia B, Vazquez J, Del Casar JM, Gonzalez LO, Vizoso FJ, 2015, A phenotype from tumor stroma based on the expression of metalloproteases and their inhibitors, associated with prognosis in breast cancer. Oncoimmunology 4:e992222 Hawkins RA, White G, Bundred NJ, Dixon JM, Miller WR, Stewart HJ, Forrest AP, 1987, Prognostic significance of oestrogen and progestogen receptor activities in breast cancer. Br J Surg 74: 1009–1013 Savouret JF, Misrahi M, Milgrom E, 1990, Molecular action of progesterone. Int J BioChemiPhysics 22:579–594 Graham JD, Clarke CL, 2002, Expression and transcriptional activity of progesterone receptor A and progesterone receptor B in mammalian cells. Breast Cancer Res 4:187–190 Mani SK, OyolaMG, 2012, Progesterone signalingmechanisms in brain and behavior. Front Endocrinol, Lausanne, 3:7. https://doi. org/10.3389/fendo.2012.00007 Fisher B, Redmond C, Fisher ER, Caplan R, 1988, Relative worth of estrogen or progesterone receptor and pathologic characteristics of differentiation as indicators of prognosis in node negative breast cancer patients: findings from National Surgical Adjuvant Breast and Bowel Project Protocol B-06. J Clin Oncol 6:1076–1087 Mote PA, Bartow S, Tran N, Clarke CL, 2002, Loss of co-ordinate expression of progesterone receptors A and B is an early event in breast carcinogenesis. Breast Cancer Res Treat 72:163–172 Gabriel CA,Mitra N, Demichele A, Rebbeck T, 2013, Association of progesterone receptor gene, PGR, variants and breast cancer risk in African American women. Breast Cancer Res Treat 139:833– 843. , DOI 10.1007/s10549-013-2592-0 Pooley KA, Healey CS, Smith PL, Pharoah PD, Thompson D, Tee L, West J, Jordan C et al, 2006, Association of the progesterone receptor gene with breast cancer risk: a single-nucleotide polymorphism tagging approach. Cancer Epidemiol Biomark Prev 15:675– 682 Pearce CL, Hirschhorn JN,Wu AH, Burtt NP, Stram DO, Young S, Kolonel LN, Henderson BE et al, 2005, Clarifying the PROGINS allele association in ovarian and breast cancer risk: a haplotypebased analysis. J Natl Cancer Inst 97:51–59 Gold B, Kalush F, Bergeron J, Scott K,Mitra N,Wilson K, Ellis N, Huang H et al, 2004, Estrogen receptor genotypes and haplotypes associated with breast cancer risk. Cancer Res 64:8891–8900 Johnatty SE, Spurdle AB, Beesley J, Chen X, Hopper JL, Duffy DL, Chenevix-Trench G, 2008, Progesterone receptor polymorphisms and risk of breast cancer: results from two Australian breast cancer studies. Breast Cancer Res Treat 109:91–99 Fernandez LP, Milne RL, Barroso E, Cuadros M, Arias JI, Ruibal A, Benitez J, Ribas G, 2006, Estrogen and progesterone receptor gene polymorphisms and sporadic breast cancer risk: a Spanish case control study. Int J Cancer 119:467–471 Romano A, Delvoux B, Fischer DC, Groothuis P, 2007, The PROGINS polymorphism of the human progesterone receptor diminishes the response to progesterone. J Mol Endocrinol 38:331– 350 Clendenen T, Zeleniuch-Jacquotte A, Wirgin I, Koenig KL, Afanasyeva Y, Lundin E, Arslan AA, Axelsson T et al, 2013, Genetic variants in hormone-related genes and risk of breast cancer. PLoS One 8:e69367. , DOI 10.1371/journal.pone. 0069367 Diergaarde B, Potter JD, Jupe ER, Manjeshwar S, Shimasaki CD, Pugh TW, Defreese DC, Gramling BA et al, 2008, Polymorphisms in genes involved in sex hormone metabolism, estrogen plus progestin hormone therapy use, and risk of postmenopausal breast cancer. Cancer Epidemiol Biomark Prev 17:1751–1759. https:// doi.org/10.1158/1055-9965.EPI-08-0168 Reding KW, Li CI, Weiss NS, Chen C, Carlson CS, Duggan D, Thummel KE, Daling JR, Malone KE, 2009, Genetic variation in the progesterone receptor and metabolism pathways and hormone therapy in relation to breast cancer risk. Am J Epidemiol 170:1241– 1249. , DOI 10.1093/aje/kwp298 Spurdle AB, Hopper JL, Chen X, McCredieMR, Giles GG, Venter DJ, Southey MC, Chenevix-Trench G, 2002, The progesterone receptor exon 4 Val660Leu G/T polymorphism and risk of breast cancer in Australian women. Cancer Epidemiol Biomark Prev 11: 439–443 Gaudet MM, Milne RL, Cox A, Camp NJ, Goode EL, Humphreys MK, Dunning AM, Morrison J et al, 2009, Five polymorphisms and breast cancer risk: results from the Breast Cancer Association Consortium. Cancer Epidemiol Biomark Prev 18:1610–1616. , DOI 10.1158/1055-9965 Zhang B, Beeghly-Fadiel A, Long J, Zheng W, 2011, Genetic variants associated with breast-cancer risk: comprehensive research synopsis, meta-analysis, and epidemiological evidence. Lancet Oncol 12:477–488. , DOI 10.1016/S1470-2045(11, 70076-6 De Vivo I, Hankinson SE, Colditz GA, Hunter DJ, 2004, The progesterone receptor Val660–>Leu polymorphism and breast cancer risk. Breast Cancer Res 6:R636–R639 Huggins GS, Wong JY, Hankinson SE, De Vivo I, 2006, GATA5 activation of the progesterone receptor gene promoter in breast cancer cells is influenced by the +331G/A polymorphism. Cancer Res 66:1384–1390 Feigelson HS, Rodriguez C, Jacobs EJ, DiverWR, Thun MJ, Calle EE, 2004, No association between the progesterone receptor gene +331G/A polymorphism and breast cancer. Cancer Epidemiol Biomark Prev 13:1084–1085 Yu KD, Chen AX, Shao ZM, 2010, No association between a progesterone receptor gene promoter polymorphism, +331G>A, and breast cancer risk in Caucasian women: evidence from a literature-based meta-analysis. Breast Cancer Res Treat 122:853– 858 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 141 EP 147 DI 10.1007/s12253-017-0379-z PG 7 ER PT J AU Dobi, Fodor, E Maraz, A Egyud, Zs Cserhati, A Tiszlavicz, L Reisz, Z Barzo, P Varga, Z Hideghety, K AF Dobi, Agnes Fodor, Emese Maraz, Aniko Egyud, Zsofia Cserhati, Adrienne Tiszlavicz, Laszlo Reisz, Zita Barzo, Pal Varga, Zoltan Hideghety, Katalin TI Boost Irradiation Integrated to Whole Brain Radiotherapy in the Management of Brain Metastases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE WBRT; Simultaneous integrated boost; Brain metastasis .3DCRT; IMRT ID WBRT; Simultaneous integrated boost; Brain metastasis .3DCRT; IMRT AB Our retrospective analysis aimed to evaluate the clinical value of dose intensification schemes: WBRT and consecutive, delayed, or simultaneous integrated boost (SIB) in brain metastasis (BM) management. Clinical data and overall survival (OS) of 468 patients with BM from various primaries treated with 10 × 3 Gy WBRT (n = 195), WBRT+ 10 × 2 Gy boost (n = 125), or simultaneously 15 × 2.2 Gy WBRT+0.7 Gy boost (n = 148) during a 6-year period were statistically analysed. Significant difference in OS could be detected with additional boost to WBRT (3.3 versus 6.5 months) and this difference was confirmed for BMs of lung cancer and melanoma and both for oligo- and multiplex lesions. The OS was prolonged for the RPA 2 and RPA3 categories, if patients received escalated dose, 4.0 vs. 7.7 months; (p = 0.002) in class RPA2 and 2.6 vs. 4.2 months; (p < 0.0001) in the class RPA 3 respectively. The significant difference in OS was also achieved with SIB. The shortened overall treatment time of SIB with lower WBRT fraction dose exhibited survival benefit over WBRT alone, and could be applied for patients developing BM even with unfavourable prognostic factors. These results warrant for further study of this approach with dose escalation using the lately available solutions for hippocampus sparing and fractionated stereotactic irradiation. The simultaneous delivery of WBRT with reduced fraction dose and boost proved to be advantageous prolonging the OS with shortened treatment time and reduced probability for cognitive decline development even for patients with poor performance status and progressing extracranial disease. C1 [Dobi, Agnes] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Fodor, Emese] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Maraz, Aniko] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Egyud, Zsofia] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Cserhati, Adrienne] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Tiszlavicz, Laszlo] University of Szeged, Department of Pathology, Allomas utca 1, H-6725 Szeged, Hungary. [Reisz, Zita] University of Szeged, Department of Pathology, Allomas utca 1, H-6725 Szeged, Hungary. [Barzo, Pal] University of Szeged, Department of Neurosurgery, Semmelweis utca 8, H-6725 Szeged, Hungary. [Varga, Zoltan] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Hideghety, Katalin] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. RP Dobi, (reprint author), University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. EM dobiagnes@gmail.com CR Nayak L, Lee EQ,Wen PY, 2012, Epidemiology of brain metastases. Curr Oncol Rep 14(1):48–54 Singh M, Manoranjan B, Mahendram S, McFarlane N, Venugopal C, Singh SK, 2014, Brain metastasis-initiating cells: survival of the fittest. Int J Mol Sci 15(5):9117–9133 Taillibert S, Le Rhun E, 2015, Epidemiology of brain metastases. Cancer Radiother 19(1):3–9, Article in French) Soffietti R, Ruda R, Mutani R, 2002, Management of brain metastases. J Neurol 249(10):1357–1369 Ellis TL, Neal MT, Chan MD, 2012, The role of surgery, radiosurgery and whole brain radiation therapy in the management of patients with metastatic brain tumors. Int J Surg Oncol 952345 Khuntia D, Brown P, Li J et al, 2006, Whole-brain radiotherapy in the management of brain metastasis. J Clin Oncol 24:1295–1304 Patchell RA, Tibbs PA,Walsh JWet al, 1990, A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med 322:494–500 Weber DC, Caparrotti F, Laouiti M, Malek K, 2011, Simultaneous in-field boost for patients with 1 to 4 brainmetastasis/es treated with volumetric modulated arc therapy: a prospective study on qualityof- life. Radiat Oncol 6:79 Linskey ME, Andrews DW, Asher AL et al, 2010, The role of stereotactic radiosurgery in the management of patients with newly diagnosed brain metastases: a systematic review and evidencebased clinical practice guideline. J Neuro-Oncol 96(1):45–68 Gaspar LE, Mehta MP, Patchell RA et al, 2010, The role of whole brain radiation therapy in the management of newly diagnosed brain metastases: a systematic review and evidencebased clinical practice guideline. J Neuro-Oncol 96(1):17–32 Kalkanis SN, Kondziolka D, Gaspar LE et al, 2010, The role of surgical resection in the management of newly diagnosed brain metastases: a systematic review and evidencebased clinical practice guideline. J Neuro-Oncol 96(1):33–43 Sahgal A, Aayama H, Kocher M et al, 2015, Phase 3 trials of stereotactic radiosurgery with or without whole-brain radiation therapy for 1 to 4 brain metastasis: individual patient data meta-analysis. Int J Radiat Oncol Biol Phys 91(4):710–717 Aayoma H, Tago M, Shirato H, 2015, Stereotactic radiosurgery with or without upfront whole brain radiation therapy for one to four brain metastases. Secondary analysis of the diagnosis-specific graded prognostic assessment. JAMA Oncol 1(4):457–464 Yuan X, Liu WJ, Li B, Shen ZT, Shen JS, Zhu XX, 2017, A Bayesian network meta-analysis of whole brain radiotherapy and stereotactic radiotherapy for brain metastasis.Medicine, Baltimore, 96(34):e7698. , DOI 10.1097/MD.0000000000007698 Chung SY, Chang JH, Kim HR, Cho BC, Lee CG, Suh CO, 2017, Optimal dose and volume for postoperative radiotherapy in brain oligometastases from lung cancer: a retrospective study. Radiat Oncol J 35(2):153–162 Casanova N, Mazouni Z, Bieri S, Combescure C, Pica A, Weber DC, 2010, Whole brain radiotherapy with a conformational external beam radiation boost for lung cancer patients with 1-3 brain metastasis: a multi institutional study. Radiat Oncol 5:13 Bruzzaniti V, Abate A, Pedrini M, Benassi M, Strigari L, 2011, IsoBED: a tool for automatic calculation of biologically equivalent fractionation schedules in radiotherapy using IMRTwith a simultaneous integrated boost, SIB, technique. J Exp Clin Cancer Res 30: 52 Borghetti P, Pedretti S, Spiazzi L, Avitabile R, UrpisM, Foscarini F, Tesini G, Trevisan F, Ghirardelli P, Pandini SA, Triggiani L, Magrini SM, Buglione M, 2016, Whole brain radiotherapy with adjuvant or concomitant boost in brain metastasis:dosimetric comparison between helical and volumetric IMRT technique. Radiat Oncol 11:59. , DOI 10.1186/s13014-016-0634-6 Hall EJ, Brenner DJ, 1993, The radiobiology of radiosurgery: rationale for different treatment regimes for AVMs and malignancies. Int J Radiat Oncol Biol Phys 25:381–385 Chao JH, Phillips R, Nickson JJ, 1954, Roentgen-ray therapy of cerebral metastases. Cancer 7:682–689 Nieder C, Spanne O, Mehta MP, Grosu AL, Geinitz H, 2011, Presentation, patterns of care, and survival in patients with brain metastases: what has changed in the last 20 years? Cancer 117(11): 2505–2512 Kocher M, Soffietti R, Abacioglu U et al, 2011, Adjuvant wholebrain radiotherapy versus observation after radiosurgery or surgical resection of one to three cerebral metastases: results of the EORTC 22952-26001 study. J Clin Oncol 29(2):134–141 Tsao MN(2015, Brain metastases: advances over the decades. Ann Palliat Med 4(4):225–232 Andrews DW, Scott CB, Sperduto PW et al, 2004, Whole brain radiation therapy with or without stereotactic radiosurgery boost for patients with one to three brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet 363:1665–1672 Kondziolka D, Patel A, Lunsford LD, Kassam A, Flickinger JC, 1999, Stereotactic radiosurgery plus whole brain radiotherapy versus radiotherapy alone for patients with multiple brain metastases. Int J Radiat Oncol Biol Phys 45:427–434 Aoyama H, Shirato H, Tago M et al, 2006, Stereotactic radiosurgery plus whole-brain radiation therapy vs stereotactic radiosurgery alone for treatment of brain metastases: a randomized controlled trial. JAMA 295:2483–2491 Kocher M, Soffietti R, Abacioglu U et al, 2011, Adjuvant wholebrain radiotherapy versus observation after radiosurgery or surgical resection of one to three cerebral metastases: results of the EORTC 22952-26001 study. J Clin Oncol 29:134–141 Buglione M, Pedretti S, Gipponi S et al, 2015, The treatment of patients with 1-3 brain metastases: is there a place for whole brain radiotherapy alone, yet? A retrospective analysis. Radiol Med 120(12):1146–1152 Brown PD, Asher AL, Ballman KV et al, 2015, NCCTG N0574, Alliance): A phase III randomized trial of whole brain radiation therapy, WBRT, in addition to radiosurgery, SRS, in patients with 1 to 3 brain metastases. J Clin Oncol 33(15_suppl):LBA4 Meyers CA, Smith JA, Bezjak A et al, 2004, Neurocognitive function and progression in patients with brain metastases treated with whole-brain radiation and motexafin gadolinium: results of a randomized phase III trial. J Clin Oncol 22:157–165 Weiss SE, Kelly PJ, 2010, Neurocognitive function after WBRT plus SRS or SRS alone. Lancet Oncol 11:220–221 Soffietti R, Kocher M, Abacioglu UM et al, 2013, A European Organisation for Research and Treatment of Cancer phase III trial of adjuvant whole-brain radiotherapy versus observation in patients with one to three brain metastases from solid tumors after surgical resection or radiosurgery: quality-of-life results. J Clin Oncol 31: 65–72 Tsao M, Xu W, Sahgal A, 2012, A meta-analysis evaluating stereotactic radiosurgery, whole-brain radiotherapy, or both for patients presenting with a limited number of brain metastases. Cancer 118:2486–2493 Soliman H, Das S, Larson DA, Sahgal A, 2016, Stereotactic radiosurgery, SRS, in the modern management of patients with brain metastases. Oncotarget 7(11):12318–12330 Mehta M, 2015, The controversy surrounding the use of wholebrain radiotherapy in brain metastases patients. Neuro-Oncology 17(7):919–923 Dye NB, Gondi V, Mehta MP, 2015, Strategies for preservation of memory function in patients with brainmetastases. Chin Clin Oncol 4(2):24 Brown PD, Pugh S, Laack NN et al, 2013, Memantine for the prevention of cognitive dysfunction in patients receiving wholebrain radiotherapy: a randomized, double-blind, placebocontrolled trial. Neuro-Oncology 15(10):1429–1437 Filley CM, Kleinschmidt-DeMasters BK, 2001, Toxic leukoencephalopathy. N Engl J Med 345:425–432 Arriagada R, Le Chevalier T, Borie F et al, 1995, Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. J Natl Cancer Inst 87:183–190 Komaki R, Meyers CA, Shin DM et al, 1995, Evaluation of cognitive function in patients with limited small cell lung cancer prior to and shortly following prophylactic cranial irradiation. Int J Radiat Oncol Biol Phys 33:179–182 Tiwari V, Pande SC, Verma K, Goel S, 2015, Simultaneous integrated boost with intensity modulated radiation therapy in brain oligometastases: a feasible technique for developing countries. South Asian J Cancer 4(1):11–14 Rodrigues G, Zindler J, Warner A, Bauman G, Senan S, Lagerwaard F, 2013, Propensity-score matched pair comparison of whole brain with simultaneous in-field boost radiotherapy and stereotactic radiosurgery. Radiother Oncol 106(2):206–209 Patchell RA, Tibbs PA, Regine WF et al, 1998, Postoperative radiotherapy in the treatment of single metastases to the brain: a randomized trial. JAMA 280(17):1485–1489 Wang TJ, Saad S, Qureshi YH et al, 2015, Outcomes of gamma knife radiosurgery, bi-modality & tri-modality treatment regimens for patients with one or multiple brain metastases: the Columbia University Medical Center experience. J Neuro-Oncol 122(2):399–408 Lin NU, Lee EQ, Aoyama H et al, 2013, Challenges relating to solid tumour brain metastases in clinical trials, part 1: patient population, response, and progression. A report from the RANO group. Lancet Oncol 14:e396–e406 Lin NU,Wefel JS, Lee EQ et al, 2013, Challenges relating to solid tumour brain metastases in clinical trials, part 2: neurocognitive, neurological, and quality-of-life outcomes. A report from the RANO group. Lancet Oncol 14:e407–e416 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 149 EP 157 DI 10.1007/s12253-018-0383-y PG 9 ER PT J AU Wang, JX Wu, HL Zhu, M Zhou, R AF Wang, Jian-Xiang Wu, Hai-Long Zhu, Meng Zhou, Rui TI Role of Anti-Epidermal Growth Factor Receptor Therapy Compared with Anti-Vascular Endothelial Growth Factor Therapy for Metastatic Colorectal Cancer: an Update Meta-Analysis of Randomized Clinical Trials SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Anti-EGFR; Anti-VEGF; Chemotherapy; Meta-analysis ID Colorectal cancer; Anti-EGFR; Anti-VEGF; Chemotherapy; Meta-analysis AB Monoclonal antibodies targeting epidermal growth factor receptor (EGFR) or vascular endothelial growth factor (VEGF) have showed clinical benefit in combination with chemotherapeutic cytotoxic drugs in the first-line therapy of metastatic colorectal cancer (mCRC). Data from randomized studies comparing these monoclonal antibodies as initial therapy is conflicting, and their comparative efficacy remains unknown. This study aimed to evaluate the impact of the combination of anti-epidermal growth factor receptor (anti-EGFR) therapy and anti-vascular endothelial growth factor therapy on mCRC patient outcomes by combining the data from randomized clinical trials. Three trials meeting the eligibility criteria, and four randomized studies were included in the meta-analysis. For MCRC patients with KRAS wild type (KRAS-WT), the ORR was superior in patients treated with anti-EGFR compared with those who treated with anti-VEGF therapy. This effect was even better for all RAS-WT patients. Progression-free survival (PFS) rates were not significantly different for KRAS-WT mCRC and all RAS-WT mCRC between the two groups. The overall survival (OS) was higher for RAS wild-type (RAS-WT) mCRC patients who received anti-EGFR, but the KRAS-WT patients compared to the anti-VEGF therapy. The results of our research indicate that superior ORR and OS between the addition of anti-EGFR therapy VS anti-VEGF therapy in all RAS-WT patients with MCRC. There was no significant difference in OS and PFS between the two groups for KRAS-WT mCRC. These results suggest that anti- EGFR monoclonal antibodies can achieve an equivalent efficacy when compared with anti-VEGF therapy of all RAS-WT mCRC patients. C1 [Wang, Jian-Xiang] Huazhong University of Science and Technology, Puai Hospital, Department of General Surgery, No. 473 Hanzheng Street, Qiaokou District, 430033 Wuhan, China. [Wu, Hai-Long] Huazhong University of Science and Technology, Puai Hospital, Department of General Surgery, No. 473 Hanzheng Street, Qiaokou District, 430033 Wuhan, China. [Zhu, Meng] Huazhong University of Science and Technology, Puai Hospital, Department of General Surgery, No. 473 Hanzheng Street, Qiaokou District, 430033 Wuhan, China. [Zhou, Rui] Huazhong University of Science and Technology, Puai Hospital, Department of General Surgery, No. 473 Hanzheng Street, Qiaokou District, 430033 Wuhan, China. RP Zhou, R (reprint author), Huazhong University of Science and Technology, Puai Hospital, Department of General Surgery, 430033 Wuhan, China. EM zhourui0815@126.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386. https://doi. org/10.1002/ijc.29210 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J. Clin. 65(2):87 Van Cutsem E, Nordlinger B, Adam R, Kohne CH, Pozzo C, Poston G, Ychou M, Rougier P, European Colorectal Metastases Treatment G, 2006, Towards a pan-European consensus on the treatment of patients with colorectal liver metastases. Eur J Cancer 42(14): 2212–2221. , DOI 10.1016/j.ejca.2006.04.012 Petrelli F, Borgonovo K, Cabiddu M, Ghilardi M, Barni S, 2011, Cetuximab and panitumumab in KRAS wild-type colorectal cancer: a meta-analysis. Int J Color Dis 26(7):823–833. , DOI 10.1007/s00384-011-1149-0 Vale CL, Tierney JF, Fisher D,Adams RA, Kaplan R,Maughan TS, Parmar MK, Meade AM, 2012, Does anti-EGFR therapy improve outcome in advanced colorectal cancer? A systematic review and meta-analysis. Cancer Treat Rev 38(6):618–625. , DOI 10. 1016/j.ctrv.2011.11.002 Bokemeyer C, Bondarenko I, Makhson A, Hartmann JT, Aparicio J, de Braud F, Donea S, Ludwig H, Schuch G, Stroh C, Loos AH, Zubel A, Koralewski P, 2009, Fluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment ofmetastatic colorectal cancer. J Clin Oncol 27(5):663–671. https:// doi.org/10.1200/JCO.2008.20.8397 Van Cutsem E, Kohne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, D'Haens G, Pinter T, Lim R, Bodoky G, Roh JK, Folprecht G, Ruff P, Stroh C, Tejpar S, Schlichting M, Nippgen J, Rougier P, 2009, Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med 360(14):1408–1417. , DOI 10.1056/NEJMoa0805019 Van Cutsem E, Kohne CH, Lang I, Folprecht G, Nowacki MP, Cascinu S, Shchepotin I, Maurel J, Cunningham D, Tejpar S, Schlichting M, Zubel A, Celik I, Rougier P, Ciardiello F, 2011, Cetuximab plus irinotecan, fluorouracil, and leucovorin as firstline treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol 29(15):2011–2019. , DOI 10.1200/ JCO.2010.33.5091 Bokemeyer C, Van Cutsem E, Rougier P, Ciardiello F, Heeger S, SchlichtingM, Celik I, Kohne CH, 2012, Addition of cetuximab to chemotherapy as first-line treatment for KRAS wild-typemetastatic colorectal cancer: pooled analysis of the CRYSTAL and OPUS randomised clinical trials. Eur J Cancer 48(10):1466–1475. , DOI 10.1016/j.ejca.2012.02.057 Hurwitz H, Fehrenbacher L, NovotnyW, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E, Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F, 2004, Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350(23):2335–2342. , DOI 10. 1056/NEJMoa032691 Macedo LT, da Costa Lima AB, Sasse AD, 2012, Addition of bevacizumab to first-line chemotherapy in advanced colorectal cancer: a systematic review and meta-analysis, with emphasis on chemotherapy subgroups. BMC Cancer 12:89. , DOI 10.1186/ 1471-2407-12-89 Saltz LB, Clarke S, Diaz-Rubio E, Scheithauer W, Figer A, Wong R, Koski S, Lichinitser M, Yang TS, Rivera F, Couture F, Sirzen F, Cassidy J, 2008, Bevacizumab in combination with oxaliplatinbased chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol 26(12):2013– 2019. , DOI 10.1200/JCO.2007.14.9930 Hurwitz HI, Yi J, Ince W, Novotny WF, Rosen O, 2009, The clinical benefit of bevacizumab in metastatic colorectal cancer is independent of K-ras mutation status: analysis of a phase III study of bevacizumab with chemotherapy in previously untreated metastatic colorectal cancer. Oncologist 14(1):22–28. , DOI 10.1634/ theoncologist.2008-0213 Heinemann V, von Weikersthal LF, Decker T, Kiani A, Vehling- Kaiser U, Al-Batran SE, Heintges T, Lerchenmuller C, Kahl C, Seipelt G, Kullmann F, Stauch M, Scheithauer W, Hielscher J, Scholz M, Muller S, Link H, Niederle N, Rost A, Hoffkes HG, Moehler M, Lindig RU, Modest DP, Rossius L, Kirchner T, Jung A, Stintzing S, 2014, FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer, FIRE-3): a randomised, open-label, phase 3 trial. Lancet Oncol 15(10):1065–1075. , DOI 10.1016/S1470- 2045(14)70330-4 Stintzing S,Modest DP, Rossius L, LerchMM, vonWeikersthal LF, Decker T, Kiani A, Vehling-Kaiser U, Al-Batran SE, Heintges T, Lerchenmuller C, Kahl C, Seipelt G, Kullmann F, Stauch M, Scheithauer W, Held S, Giessen-Jung C, Moehler M, Jagenburg A, Kirchner T, Jung A, Heinemann V, investigators F-, 2016, FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab for metastatic colorectal cancer, FIRE-3): a post-hoc analysis of tumour dynamics in the final RAS wild-type subgroup of this randomised open-label phase 3 trial. Lancet Oncol 17, 10):1426– 1434. , DOI 10.1016/S1470-2045(16)30269-8 Schwartzberg LS, Rivera F, Karthaus M, Fasola G, Canon JL, Hecht JR, Yu H, Oliner KS, Go WY, 2014, PEAK: a randomized, multicenter phase II study of panitumumab plus modified fluorouracil, leucovorin, and oxaliplatin, mFOLFOX6, or bevacizumab plus mFOLFOX6 in patients with previously untreated, unresectable, wild-type KRAS exon 2 metastatic colorectal cancer. J Clin Oncol 32(21):2240–2247. , DOI 10.1200/JCO. 2013.53.2473 Venook AP, Niedzwiecki D, Lenz HJ, Innocenti F, Fruth B, Meyerhardt JA, Schrag D, Greene C, O'Neil BH, Atkins JN, Berry S, Polite BN, O'Reilly EM, Goldberg RM, Hochster HS, Schilsky RL, Bertagnolli MM, El-Khoueiry AB, Watson P, Benson AB 3rd, Mulkerin DL, Mayer RJ, Blanke C, 2017, Effect of First-Line Chemotherapy Combined With Cetuximab or Bevacizumab on Overall Survival in Patients With KRAS Wild- Type Advanced or Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA 317(23):2392–2401. , DOI 10.1001/ jama.2017.7105 Higgins JP, Thompson SG, 2002, Quantifying heterogeneity in a meta-analysis. Stat Med 21(11):1539–1558. , DOI 10. 1002/sim.1186 Lenz H, Niedzwiecki D, Innocenti F, Blanke C, Mahony MR, O'Neil BH, Shaw JE, Polite B, Hochster H, Atkins J, Goldberg R, Mayer R, Schilsky RL, Bertagnolli M, Venook A, 2014, 501OCALGB/SWOG 80405: PHASE I II TRIAL OF IRINOTECAN/5-FU/LEUCOVORIN, FOLFIRI, OR OXALIPLATIN/5-FU/LEUCOVORIN, MFOLFOX6, WITH BEVACIZUMAB, BV, OR CETUXIMAB, CET, FOR PATIENTS, PTS, WITH EXPANDED RAS ANALYSES UNTREATED METASTATIC ADENOCARCINOMA OF THE COLON OR RECTUM, MCRC). Ann Oncol 25(suppl_4): mdu438.413–mdu438.413. , DOI 10.1093/annonc/ mdu438.13 Venook AP, Niedzwiecki D, Lenz H-J, Innocenti F, Mahoney MR, O'Neil BH, Shaw JE, Polite BN, Hochster HS, Atkins JN, Goldberg RM,Mayer RJ, Schilsky RL, BertagnolliMM, Blanke CD, Cancer, Leukemia Group B S, ECOG, 2014, CALGB/SWOG 80405: phase III trial of irinotecan/5-FU/leucovorin, FOLFIRI, or oxaliplatin/5-FU/leucovorin, mFOLFOX6, with bevacizumab, BV, or cetuximab, CET, for patients, pts, with KRAS wild-type, wt, untreated metastatic adenocarcinoma of the colon or rectum, MCRC). J Clin Oncol 32(15_suppl):LBA3–LBA3. , DOI 10.1200/jco.2014.32.15_suppl.lba3 Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, Rivera F, Kocakova I, Ruff P, Blasinska-Morawiec M, Smakal M, Canon JL, Rother M, Oliner KS, TianY, Xu F, Sidhu R, 2014, Final results from PRIME: randomized phase III study of panitumumab with FOLFOX4 for first-line treatment of metastatic colorectal cancer. Ann Oncol 25(7):1346–1355. , DOI 10.1093/annonc/ mdu141 Khattak MA, Martin H, Davidson A, Phillips M, 2015, Role of first-line anti-epidermal growth factor receptor therapy compared with anti-vascular endothelial growth factor therapy in advanced colorectal cancer: a meta-analysis of randomized clinical trials. Clin Colorectal Cancer 14(2):81–90 Maughan TS, Adams RA, Smith CG, Meade AM, Seymour MT, Wilson RH, Idziaszczyk S, Harris R, Fisher D, Kenny SL, Kay E, Mitchell JK, Madi A, Jasani B, JamesMD, Bridgewater J, Kennedy MJ, Claes B, Lambrechts D, Kaplan R, Cheadle JP, Investigators MCT, 2011, Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer: results of the randomised phase 3 MRC COIN trial. Lancet 377(9783):2103–2114. , DOI 10.1016/S0140- 6736(11)60613-2 Chan D, Pavlakis N, Price TJ, Karapetis CS, Tebbutt NC, Shapiro JD, Segelov E, 2014, Impact of chemotherapy partner on efficacy of targeted therapy in metastatic colorectal cancer, mCRC): a metaanalysis. Asia Pac J Clin Oncol 10:104–104 Piessevaux H, Buyse M, Schlichting M, Van CE, Bokemeyer C, Heeger S, Tejpar S, 2013, Use of early tumor shrinkage to predict long-term outcome in metastatic colorectal cancer treated with cetuximab. Journal of clinical oncology official journal of the. Proc Am Soc Clin Oncol 31(30):3764 Mansmann UR, Sartorius U, Laubender RP, Giessen CA, Esser R, Heinemann V, 2013, Deepness of response: a quantitative analysis of its impact on post-progression survival time after first-line treatment in patients with mCRC. J Clin Oncol 31(4) Ciardiello F, Bianco RR, Caputo R, Damiano V, Troiani T, Melisi D, De VF, De PS, Bianco AR, Tortora G, 2004, Antitumor activity of ZD6474, a vascular endothelial growth factor receptor tyrosine kinase inhibitor, in human cancer cells with acquired resistance to antiepidermal growth factor receptor therapy. Clin Cancer Res 10(2):784 Viloria-Petit A, Crombet T, Jothy S, Hicklin D, Bohlen P, Schlaeppi JM, Rak J, Kerbel RS, 2001, Acquired resistance to the antitumor effect of epidermal growth factor receptor-blocking antibodies in vivo: a role for altered tumor angiogenesis. Cancer Res 61(13): 5090–5101 Yang YH, Lin JK, Chen WS, Lin TC, Yang SH, Jiang JK, Lan YT, Lin CC, Yen CC, Tzeng CH, 2014, Comparison of cetuximab to bevacizumab as the first-line bio-chemotherapy for patients with metastatic colorectal cancer: superior progression-free survival is restricted to patients with measurable tumors and objective tumor response—a retrospective stu. J Cancer Res Clin Oncol 140(11): 1927–1936 Ye LC, Liu TS, Ren L,Wei Y, ZhuDX, Zai SY,Ye QH,Yu Y, Xu B, Qin XY, 2013, Randomized controlled trial of cetuximab plus chemotherapy for patients with KRAS wild-type unresectable colorectal liver-limited metastases. Journal of clinical oncology official journal of the. Proc Am Soc Clin Oncol 31(16):1931 Okines A, Del PO, Cunningham D, Chau I, Van CE, Saltz L, Cassidy J, 2009, Surgery with curative-intent in patients treated with first-line chemotherapy plus bevacizumab for metastatic colorectal cancer first BEAT and the randomised phase-III NO16966 trial. Br J Cancer 101(7):1033 Folprecht G, Gruenberger T, Bechstein WO, Raab HR, Lordick F, Hartmann JT, Lang H, Frilling A, Stoehlmacher J, Weitz J, 2010, Tumour response and secondary resectability of colorectal liver metastases following neoadjuvant chemotherapy with cetuximab: the CELIM randomised phase 2 trial. Lancet Oncol 11(1):38–47 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 159 EP 166 DI 10.1007/s12253-017-0365-5 PG 8 ER PT J AU Fernandez-Vega, I Santos-Juanes, J Camacho-Urkaray, E Lorente-Gea, L Garcia, B Gutierrez-Corres, BF Quiros, ML Guerra-Merino, I Aguirre, JJ AF Fernandez-Vega, Ivan Santos-Juanes, Jorge Camacho-Urkaray, Emma Lorente-Gea, Laura Garcia, Beatriz Gutierrez-Corres, Borja Francisco Quiros, M Luis Guerra-Merino, Isabel Aguirre, Javier Jose TI Miki (Mitotic Kinetics Regulator) Immunoexpression in Normal Liver, Cirrhotic Areas and Hepatocellular Carcinomas: a Preliminary Study with Clinical Relevance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Miki; Immunohistochemistry; Liver; Immunoglobulin superfamily ID Hepatocellular carcinoma; Miki; Immunohistochemistry; Liver; Immunoglobulin superfamily AB Hepatocellular carcinoma (HCC) is the most common type of primary malignant tumor in the liver. One of the main features of cancer survival is the generalized loss of growth control exhibited by cancer cells, and Miki is a protein related to the immunoglobulin superfamily that plays an important role in mitosis. We aim to study protein expression levels of Miki in non-tumoral liver and 20 HCCs recruited from a Pathology Department. Clinical information was also obtained. A tissue microarray was performed, and immunohistochemical techniques applied to study protein expression levels of Miki. In normal liver, Miki was weakly expressed, showing nuclear staining in the hepatocytes. Cirrhotic areas and HCCs showed a variety of staining patterns. Most HCC samples showed positive expression, with three different staining patterns being discernible: nuclear, cytoplasmic and mixed. Statistical analysis showed a significant association between grade of differentiation, Ki-67 proliferative index, survival rates and staining patterns. This study has revealed the positive expression of Miki in normal liver, cirrhotic areas and HCCs. Three different staining patterns of Miki expression with clinical relevance were noted in HCCs. C1 [Fernandez-Vega, Ivan] Hospital Universitario de Araba-Txagorritxu, Department of PathologyVitoria-Gasteiz, Spain. [Santos-Juanes, Jorge] Hospital Universitario Central de Asturias, Department of PathologyOviedo, Spain. [Camacho-Urkaray, Emma] Hospital Universitario de Araba-Txagorritxu, Department of PathologyVitoria-Gasteiz, Spain. [Lorente-Gea, Laura] Hospital Universitario de Araba-Txagorritxu, Department of PathologyVitoria-Gasteiz, Spain. [Garcia, Beatriz] Instituto Universitario Fernandez-VegaOviedo, Spain. [Gutierrez-Corres, Borja Francisco] Hospital Universitario de Araba-Txagorritxu, Department of PathologyVitoria-Gasteiz, Spain. [Quiros, M Luis] Instituto Universitario Fernandez-VegaOviedo, Spain. [Guerra-Merino, Isabel] Hospital Universitario de Araba-Txagorritxu, Department of PathologyVitoria-Gasteiz, Spain. [Aguirre, Javier Jose] Hospital Universitario de Araba-Txagorritxu, Department of PathologyVitoria-Gasteiz, Spain. RP Fernandez-Vega, I (reprint author), Hospital Universitario de Araba-Txagorritxu, Department of Pathology, Vitoria-Gasteiz, Spain. EM ivan_fernandez_vega@hotmail.com CR Stoot JH, Coelen RJ, De Jong MC, Dejong CH, 2010, Malignant transformation of hepatocellular adenomas into hepatocellular carcinomas: a systematic review including more than 1600 adenoma cases. HPB 12:509–522 McGlynn KA, London WT, 2011, The global epidemiology of hepatocellular carcinoma: present and future. Clin Liver Dis 15: 223–243 vii-x Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Ozaki Y, Matsui H, Asou H, Nagamachi A, Aki D, Honda H et al, 2012, Poly-ADP ribosylation of Miki by tankyrase-1 promotes centrosome maturation. Mol Cell 47:694–706 Chang P, Jacobson MK, Mitchison TJ, 2004, Poly(ADP-ribose, is required for spindle assembly and structure. Nature 432:645–649 Asou H, Matsui H, Ozaki Y, Nagamachi A, Nakamura M, Aki D et al, 2009, Identification of a common microdeletion cluster in 7q21.3 subband among patients with myeloid leukemia and myelodysplastic syndrome. Biochem Biophys Res Commun 383: 245–251 Nigg EA, 2002, Centrosome aberrations: cause or consequence of cancer progression? Nat Rev Cancer 2:815–825 Chung Moh M, Hoon Lee L, Shen S, 2005, Cloning and characterization of hepaCAM, a novel Ig-like cell adhesion molecule suppressed in human hepatocellular carcinoma. J Hepatol 42:833–841 Bordeaux J, Welsh A, Agarwal S, Killiam E, Baquero M, Hanna J et al, 2010, Antibody validation. BioTechniques 48:197–209 Kondo Y, 1985, Histologic features of hepatocellular carcinoma and allied disorders. Pathol Annu 20(Pt 2):405–430 Sugihara S, Nakashima O, Kojiro M, Majima Y, Tanaka M, Tanikawa K, 1992, The morphologic transition in hepatocellular carcinoma. A comparison of the individual histologic features disclosed by ultrasound-guided fine-needle biopsy with those of autopsy. Cancer 70:1488–1492 Hidaka T, Hama S, Shrestha P, Saito T, Kajiwara Y, Yamasaki F et al, 2009, The combination of low cytoplasmic and high nuclear expression of p27 predicts a better prognosis in high-grade astrocytoma. Anticancer Res 29:597–603 Matsuda Y, Yoshimura H, Ishiwata T, Sumiyoshi H, Matsushita A, Nakamura Y, et al, 2016, Mitotic index and multipolar mitosis in routine histologic sections as prognostic markers of pancreatic cancers: a clinicopathological study. Pancreatology : official journal of the international association of Pancreatology16:127–132 Batistatou A, 2004, Mitoses and cancer. Med Hypotheses 63(2): 281 Gisselsson D, HakansonU, Stoller P,Marti D, Jin Y, RosengrenAH et al, 2008, When the genome plays dice: circumvention of the spindle assembly checkpoint and near-random chromosome segregation in multipolar cancer cell mitoses. PLoS One 3:e1871 Chan KS, Koh CG, Li HY, 2012, Mitosis-targeted anti-cancer therapies: where they stand. Cell Death Dis 3:e411 Hallett RM, Huang C, Motazedian A, Auf der Mauer S, Pond GR, Hassell JA et al, 2015, Treatment-induced cell cycle kinetics dictate tumor response to chemotherapy. Oncotarget 6:7040–7052 Weaver BA, 2014, How Taxol/paclitaxel kills cancer cells. Mol Biol Cell 25:2677–2681 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 167 EP 173 DI 10.1007/s12253-018-0387-7 PG 7 ER PT J AU da Silva, NLJ Ranzi, DA Carvalho, TC Scheide, VT Strey, ThMY Graziottin, MT Bica, GC AF da Silva, Niederauer Leote Jessica Ranzi, Durayski Alana Carvalho, Trainotti Caroline Scheide, Vicente Tales Strey, Thome Machado Yuri Graziottin, Meyer Tulio Bica, Giuliano Claudia TI Cell Cycle Markers in the Evaluation of Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; Cell cycle markers; Prognosis; Immunohistochemistry ID Bladder cancer; Cell cycle markers; Prognosis; Immunohistochemistry AB Bladder cancer (BC) is a heterogeneous neoplasia characterized by a high number of recurrences. Standardized clinical and morphological parameters are not always sufficient to predict individual tumor behavior. The aim of this study was to evaluate the expression of cell cycle regulators proteins as potential adjuvant in prognosis and monitoring of this disease. Block paraffin samples from patients with urothelial bladder carcinoma treated by transurethral resection (TUR) were collected to immunohistochemistry analysis for proteins p16, p21, p27, p53, pRb and Ki-67. Chi square, logistic regression and Kaplan-Meier curve were used to analyze the prognostic value of these markers. Of the 93 patients included in the study, the main categories of staging observed were T1 (53%) and Ta (29%), and the distribution between tumor grades was 58% of patients with low grade to 42% of patients with high grade. The expressions of p16, p21, p27, p53, pRb and Ki-67 were altered in 31%, 42%, 60%, 91%, 27% and 56% of patients, respectively. The immunohistochemical expression of Ki-67 was associated with tumor histological grade (p = 0.016), and expression of pRb with recurrence-free survival (p = 0.035), but no isolated marker was significant associated with recurrence and progression in multivariate analysis. More than two markers abnormally expressed were associated with presence of recurrence (p = 0.005) and lower recurrence-free survival (p = 0.004). Our panel marker has important prognostic value for BC, especially when more than two have altered expression predicting good clinical recurrence implication. C1 [da Silva, Niederauer Leote Jessica] Health Sciences Federal University of Porto Alegre (UFCSPA), Research Laboratory of Pathology, 245 Sarmento Leite, RS 90050-170 Porto Alegre, Brazil. [Ranzi, Durayski Alana] Health Sciences Federal University of Porto Alegre (UFCSPA), Research Laboratory of Pathology, 245 Sarmento Leite, RS 90050-170 Porto Alegre, Brazil. [Carvalho, Trainotti Caroline] Health Sciences Federal University of Porto Alegre (UFCSPA), Research Laboratory of Pathology, 245 Sarmento Leite, RS 90050-170 Porto Alegre, Brazil. [Scheide, Vicente Tales] Health Sciences Federal University of Porto Alegre (UFCSPA), Research Laboratory of Pathology, 245 Sarmento Leite, RS 90050-170 Porto Alegre, Brazil. [Strey, Thome Machado Yuri] Health Sciences Federal University of Porto Alegre (UFCSPA), Research Laboratory of Pathology, 245 Sarmento Leite, RS 90050-170 Porto Alegre, Brazil. [Graziottin, Meyer Tulio] Health Sciences Federal University of Porto Alegre (UFCSPA), Research Laboratory of Pathology, 245 Sarmento Leite, RS 90050-170 Porto Alegre, Brazil. [Bica, Giuliano Claudia] Health Sciences Federal University of Porto Alegre (UFCSPA), Research Laboratory of Pathology, 245 Sarmento Leite, RS 90050-170 Porto Alegre, Brazil. RP Bica, GC (reprint author), Health Sciences Federal University of Porto Alegre (UFCSPA), Research Laboratory of Pathology, RS 90050-170 Porto Alegre, Brazil. EM claudia@ufcspa.edu.br CR Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2014, GLOBOCAN 2012 v1.0, cancer incidence and mortality worldwide: IARC CancerBase no. 11 [internet]. International Agency for Research on Cancer, Lyon. Available from: http://globocan.iarc.fr. Accessed 22 Apr 2016 Larre S, Catto JWF, Cookson MS, Messing EM, Shariat SF, Soloway MS et al, 2013, Screening for bladder cancer: rationale, limitations, whom to target, and perspectives. Eur Urol 63:1049– 1058 Kumar V, Abbas AK, Fausto N, 2010, Robbins & Cotran pathologic basis of disease, 8th edn. Saunders, Philadelphia Netto GJ, 2012, Molecular biomarkers in urothelial carcinoma of the bladder: are we there yet? Nat Rev Urol 9:41–51 Parekh DJ, Bochner BH, Dalbagni G, 2006, Superficial and muscle-invasive bladder cancer: principles of management for outcomes assessments. J Clin Oncol 24:5519–5527 Babjuk M, Burger M, Zigeuner R, Shariat SF, Van Rhijn BWG, Comperat E et al, 2013, EAU guidelines on non– muscle-invasive urothelial carcinoma of the bladder: update 2013. Eur Urol 64:639–653 Goodison S, Rosser CJ, Urquidi V, 2013, Bladder cancer detection and monitoring: assessment of urine- and blood-based marker tests. Mol Diagn Ther 17:71–84 Wang LC, Xylinas E, Kent MT, Kluth LA, Rink M, Jamzadeh A et al, 2013, Combining smoking information and molecular markers improves prognostication in patients with urothelial carcinoma of the bladder. Urol Oncol 4:433–440 Xylinas E, Kluth LA, Lotan Y, Daneshmand S, Rieken M, Karakiewicz PI et al, 2014, Blood- and tissue-based biomarkers for prediction of outcomes in urothelial carcinoma of the bladder. Urol Oncol 32:230–242 Netto GJ, Cheng L, 2012, Emerging critical role of molecular testing in diagnostic genitourinary pathology. Arch Pathol Lab Med 136:372–390 Matsushita K, Cha EK, Matsumoto K, Baba S, Chromecki TF, Fajkovic H et al, 2011, Immunohistochemical biomarkers for bladder cancer prognosis. Int J Urol 18:616–629 Kamat AM, Hegartyet RK, Gee JR, Clark PE, Svatek RS, Hegarty N et al, 2013, ICUD-EAU international consultation on bladder cancer 2012: screening, diagnosis, and molecular markers. Eur Urol 63:4–15 Cheng L, Davison DD, Adams J, Lopez-Beltran A, Wang L, Montironi R et al, 2014, Biomarkers in bladder cancer: translational and clinical implications. Crit Rev Oncol Hemat 89:73–111 Prophet EB,Mills B, Arrington JB et al, 1992, Laboratory methods in Histotechnology. Armed forces institute of pathology, Washington, DC Zavalhia LS, Romitti M, Netto GC, Dos Santos GT, Meurer RT, Hilbig A et al, 2012, Evaluation of the expression of C-kit, CD117, in ependymomas and oligodendrogliomas. Dis Markers 33:61–68 Lotan Y, Bagrodia A, Passoni N, Rachakonda V, Kapur P, Arriaga Y et al, 2013, Prospective evaluation of a molecular marker panel for prediction of recurrence and cancer-specific survival after radical cystectomy. Eur Urol 64:465–471 Olsson H, Hultman P, Monsef N, Rosell J, Jahnson S, 2012, Immunohistochemical evaluation of cell cycle regulators: impact on predicting prognosis in stage t1 urinary bladder cancer. ISRN Urol:12:379081 Montironi R, Lopez-Beltran A, Mazzucchelli R, Bostwick DG, 2003, Classification and grading of the non-invasive urothelial neoplasms: recent advances and controversies. J Clin Pathol 56: 91–95 Tanagho EA, Mcaninch JW, 2010, Urologia Geral de Smith, 17th edn. Porto Alegre, Artmed Poyet C, Jentsch B, Hermanns T, Schweckendiek D, Seifert H-H, Schmidtpeter M, Sulser T et al, 2014, Expression of histone deacetylases 1, 2 and 3 in urothelial bladder cancer. BMC Clin Pathol 14:10 Maeng Y-H, Eun S-Y, Huh J-S, 2010, Expression of fibroblast growth factor receptor 3 in the recurrence of non-muscle-invasive urothelial carcinoma of the bladder 2010. Korean J Urol 51:94–100 Wang L, Feng C, Ding G, Ding Q, Zhou Z, Jiang H et al, 2014, Ki67 and TP53 expressions predict recurrence of non-muscleinvasive bladder cancer. Tumor Biol 35:2989–2995 Rajcani J, Kajo K, AdamkovM, Moravekova LL, Felcanova Det al, 2013, Immunohistochemical characterization of urothelial carcinoma. Bratisl Lek Listy 114:431–438 Mitra AP, Bartsch CC, Cote RJ, 2009, Strategies for molecular expression profiling in bladder cancer. Cancer Metastasis Rev 28: 317–326 Lee K, Jung ES, Choi Y-J, Lee KY, Lee A, 2010, Expression of pRb, p53, p16 and cyclin D1 and their clinical implications in urothelial carcinoma. J Korean Med Sci, 10):1449–1455 Kruger S, Mahnken A, Kausch I, Feller AC, 2005, P16 Immunoreactivity is an independent predictor of tumor progression in minimally invasive urothelial bladder carcinoma. Eur Urol 47: 463–467 Weihong D, Gou Y, Sun C, Xia G,Wang H, Chen Z et al, 2014, Ki- 67 is an independent indicator in non–muscle invasive bladder cancer, NMIBC); combination of EORTC risk scores and Ki-67 expression could improve the risk stratification of NMIBC. Urol Oncol-Semin Ori 32:42.e13–42.e19 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 175 EP 181 DI 10.1007/s12253-018-0389-5 PG 7 ER PT J AU Gkouveris, I Nikitakis, N Sklavounou, A AF Gkouveris, Ioannis Nikitakis, Nikolaos Sklavounou, Alexandra TI p38 Expression and Modulation of STAT3 Signaling in Oral Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p38; STAT3; Oral cancer; Tumor grade ID p38; STAT3; Oral cancer; Tumor grade AB p38 protein belongs to Mitogen-activated protein kinases family that link extracellular stimuli with intracellular responses participating in numerous of fundamental cell processes. Persistent activation of STAT3 has been associated with cell proliferation, differentiation and apoptosis in oral squamous cell carcinoma (OSCC). This study examines the effects of p38 modulation on STAT3 signaling and cellular activities in OSCC cells and investigates possible correlation of p38 expression with tumor degree of differentiation. Phosphop38 immunostaining was performed in 60 OSCC including well, moderately and poorly differentiated tumors. Semiquantitative analysis was used, by calculating intensity, percentage and combined scores. Protein expression levels of STAT3 (total, tyrosine and serine phosphorylated), p38 and cyclin D1 were assessed in two OSCC cell lines. p38 inhibition was achieved by pharmacological agent (SB2023580). Cell proliferation and viability rates were also evaluated. Phospho-p38 immunoexpression was intense in almost all tumor specimens, nevertheless, did not correlate with tumor differentiation. Inhibition of p38 with SB203580 did not appear to affect tyrosine or serine phosphorylated STAT3 as well as cyclin D1 levels in both cell lines. Moreover, p38 inhibition resulted in mild dose dependent decreases in cell growth and viability in both cell lines. p38 is highly expressed in OSCC but does not seem to mediate the oncogenic STAT3 pathway. However, changes found in proliferation and viability may suggest that p38 functions as potent regulator of HNSCC. Understanding the complexity of p38 signaling and crosstalk between other major molecules, may guide the development of novel pharmacologic therapies for cancer treatment and prevention. C1 [Gkouveris, Ioannis] National and Kapodistrian University of Athens, Dental School, Department of Oral Pathology and Medicine, 2 Thivon Str. Goudi, 11527 Athens, Greece. [Nikitakis, Nikolaos] National and Kapodistrian University of Athens, Dental School, Department of Oral Pathology and Medicine, 2 Thivon Str. Goudi, 11527 Athens, Greece. [Sklavounou, Alexandra] National and Kapodistrian University of Athens, Dental School, Department of Oral Pathology and Medicine, 2 Thivon Str. Goudi, 11527 Athens, Greece. RP Gkouveris, I (reprint author), National and Kapodistrian University of Athens, Dental School, Department of Oral Pathology and Medicine, 11527 Athens, Greece. EM igkouver@gmail.com CR El Jamal SM, Taylor EB, Abd Elmageed ZY, Alamodi AA, Selimovic D, Alkhateeb A, Hannig M, Hassan SY, Santourlidis S, Friedlander PL, Haikel Y, Vijaykumar S, Kandil E, Hassan M, 2016, Interferon gamma-induced apoptosis of head and neck squamous cell carcinoma is connected to indoleamine-2,3-dioxygenase via mitochondrial and ER stress-associated pathways. Cell Div 11: 11. , DOI 10.1186/s13008-016-0023-4 Gkouveris I, Nikitakis N, KaranikouM, Rassidakis G, Sklavounou A, 2016, JNK1/2 expression andmodulation of STAT3 signaling in oral cancer. Oncol Lett 12(1):699–706. , DOI 10.3892/ol. 2016.4614 Molinolo AA, Amornphimoltham P, Squarize CH, Castilho RM, Patel V, Gutkind JS, 2009, Dysregulated molecular networks in head and neck carcinogenesis. Oral Oncol 45(4–5):324–334. , DOI 10.1016/j.oraloncology.2008.07.011 Chin D, Boyle GM, Porceddu S, Theile DR, Parsons PG, Coman WB, 2006, Head an neck cancer: past, present and future. Expert Rev Anticancer Ther 6(7):1111–1118. , DOI 10.1586/ 14737140.6.7.1111 Gkouveris I, Nikitakis N, Sauk J, 2015, STAT3 signaling in cancer. J Cancer Ther 6(8):709–726 Mali SB, 2015, Review of STAT3, signal transducers and activators of transcription, in head and neck cancer. Oral Oncol 51(6): 565–569. , DOI 10.1016/j.oraloncology.2015.03.004 Macha MA, Matta A, Kaur J, Chauhan SS, Thakar A, Shukla NK, Gupta SD, Ralhan R, 2011, Prognostic significance of nuclear pSTAT3 in oral cancer. Head Neck 33(4):482–489 Rane SG, Reddy EP, 2000, Janus kinases: components of multiple signaling pathways. Oncogene 19(49):5662–5679. , DOI 10.1038/sj.onc.1203925 Sen M, Joyce S, Panahandeh M, Li C, Thomas SM, Maxwell J, Wang L, Gooding WE, Johnson DE, Grandis JR, 2012, Targeting Stat3 abrogates EGFR inhibitor resistance in cancer. Clin Cancer Res 18(18):4986–4996. , DOI 10.1158/1078-0432.ccr-12- 0792 Xiong A, Yang Z, Shen Y, Zhou J, Shen Q, 2014, Transcription factor STAT3 as a novel molecular target for Cancer prevention. Cancers, Basel, 6(2):926–957. , DOI 10.3390/ cancers6020926 Shah NG, Trivedi TI, Tankshali RA, Goswami JV, Jetly DH, Shukla SN, Shah PM, Verma RJ, 2009, Prognostic significance of molecular markers in oral squamous cell carcinoma: a multivariate analysis. Head Neck 31(12):1544–1556. , DOI 10.1002/hed.21126 Trivedi TI, Tankshali RA, Goswami JV, Shukla SN, Shah PM, Shah NG, 2011, Identification of site-specific prognostic biomarkers in patients with oral squamous cell carcinoma. Neoplasma 58(3):217– 226 Bromberg J, 2002, Stat proteins and oncogenesis. J Clin Invest 109(9):1139–1142. , DOI 10.1172/jci15617 Jewett A, Head C, Cacalano NA, 2006, Emerging mechanisms of immunosuppression in oral cancers. J Dent Res 85:1061–1073 Lai SY, Johnson FM, 2010, Defining the role of the JAK-STAT pathway in head and neck and thoracic malignancies: implications for future therapeutic approaches. Drug Resist Updat 13(3):67–78. , DOI 10.1016/j.drup.2010.04.001 Naher L, Kiyoshima T, Kobayashi I, Wada H, Nagata K, Fujiwara H, Ookuma YF, Ozeki S, Nakamura S, Sakai H, 2012, STAT3 signal transduction through interleukin-22 in oral squamous cell carcinoma. Int J Oncol 41:1577–1586 Johnson GL, Lapadat R, 2002, Mitogen-activated protein kinase pathwaysmediated by ERK, JNK, and p38 protein kinases. Science 298(5600):1911–1912. , DOI 10.1126/science.1072682 Aguzzi A,Maggioni D, Nicolini G, TrediciG, GainiRM, Garavello W, 2009, MAP kinase modulation in squamous cell carcinoma of the oral cavity. Anticancer Res 29(1):303–308 Dhillon AS, Hagan S, Rath O, Kolch W, 2007, MAP kinase signalling pathways in cancer. Oncogene 26(22):3279–3290. https:// doi.org/10.1038/sj.onc.1210421 Kim JY, An JM, Chung WY, Park KK, Hwang JK, Kim d S, Seo SR, Seo JT, 2013, Xanthorrhizol induces apoptosis through ROSmediatedMAPK activation in human oral squamous cell carcinoma cells and inhibits DMBA-induced oral carcinogenesis in hamsters. Phytother Res 27(4):493–498. , DOI 10.1002/ptr.4746 Li B, Lu L, Zhong M, Tan XX, Liu CY, Guo Y, Yi X, 2013, Terbinafine inhibits KSR1 and suppresses Raf-MEK-ERK signaling in oral squamous cell carcinoma cells. Neoplasma 60(4):406– 412. , DOI 10.4149/neo_2013_052 Maggioni D, Gaini R, Nicolini G, Tredici G, Garavello W, 2011, MAPKs activation in head and neck squamous cell carcinomas. Oncol Rev 5:223–231 Doganer F, Turgut Cosan D,GunesHV, Degirmenci I, Bal C, 2014, The effects of p38 gene silencing on breast cancer cells. Mol Biol Rep 41(5):2923–2927. , DOI 10.1007/s11033-014-3148-z Junttila MR, Ala-Aho R, Jokilehto T, Peltonen J, Kallajoki M, Grenman R, Jaakkola P, Westermarck J, Kahari VM, 2007, p38alpha and p38delta mitogen-activated protein kinase isoforms regulate invasion and growth of head and neck squamous carcinoma cells. Oncogene 26(36):5267–5279. , DOI 10.1038/sj.onc. 1210332 Leelahavanichkul K, Amornphimoltham P, Molinolo AA, Basile JR, Koontongkaew S, Gutkind JS, 2014, A role for p38 MAPK in head and neck cancer cell growth and tumor-induced angiogenesis and lymphangiogenesis. Mol Oncol 8(1):105–118. https://doi. org/10.1016/j.molonc.2013.10.003 Riebe C, Pries R, Kemkers A, Wollenberg B, 2007, Increased cytokine secretion in head and neck cancer upon p38 mitogenactivated protein kinase activation. Int J Mol Med 20(6):883–887 Yen CY, Liang SS, Han LY, Chou HL, Chou CK, Lin SR, Chiu CC, 2013, Cardiotoxin III inhibits proliferation and migration of oral cancer c e l l s through MAPK and MMP s i g n a l i ng. ScientificWorldJournal 2013:650946. , DOI 10.1155/ 2013/650946 Koul HK, Pal M, Koul S, 2013, Role of p38 MAP kinase signal transduction in solid tumors. Genes Cancer 4(9–10):342–359. , DOI 10.1177/1947601913507951 Gkouveris I, Nikitakis N, Karanikou M, Rassidakis G, Sklavounou A, 2014, Erk1/2 activation and modulation of STAT3 signaling in oral cancer. Oncol Rep 32(5):2175–2182. , DOI 10.3892/ or.2014.3440 19 Ahmed ST, Mayer A, Ji JD, Ivashkiv LB, 2002, Inhibition of IL-6 signaling by a p38-dependent pathway occurs in the absence of new protein synthesis. J Leukoc Biol 72(1):154–162 Tkach M, Rosemblit C, Rivas MA, Proietti CJ, Diaz Flaque MC, Mercogliano MF, Beguelin W, Maronna E, Guzman P, Gercovich FG, Deza EG, Elizalde PV, Schillaci R, 2013, p42/p44 MAPKmediated Stat3Ser727 phosphorylation is required for progestininduced full activation of Stat3 and breast cancer growth. Endocr Relat Cancer 20(2):197–212. , DOI 10.1530/erc-12-0194 Xue P, Zhao Y, Liu Y, Yuan Q, Xiong C, Ruan J, 2014, A novel compound RY10-4 induces apoptosis and inhibits invasion via inhibiting STAT3 through ERK-, p38-dependent pathways in human lung adenocarcinoma A549 cells. Chem Biol Interact 209:25– 34. , DOI 10.1016/j.cbi.2013.11.014 Park JI, Lee MG, Cho K, Park BJ, Chae KS, Byun DS, Ryu BK, Park YK, Chi SG, 2003, Transforming growth factor-beta1 activates interleukin-6 expression in prostate cancer cells through the synergistic collaboration of the Smad2, p38-NF-kappaB, JNK, and Ras signaling pathways. Oncogene 22(28):4314–4332. https://doi. org/10.1038/sj.onc.1206478 Khandrika L, Lieberman R, Koul S, Kumar B, Maroni P, Chandhoke R, Meacham RB, Koul HK, 2009, Hypoxiaassociated p38 mitogen-activated protein kinase-mediated androgen receptor activation and increased HIF-1alpha levels contribute to emergence of an aggressive phenotype in prostate cancer. Oncogene 28(9):1248–1260. , DOI 10.1038/onc.2008.476 Maroni PD, Koul S, Meacham RB, KoulHK(2004, Mitogen activated protein kinase signal transduction pathways in the prostate. Cell Commun Signal 2(1):5. , DOI 10.1186/1478-811x-2-5 Suarez-Cuervo C, Merrell MA, Watson L, Harris KW, Rosenthal EL, Vaananen HK, Selander KS, 2004, Breast cancer cells with inhibition of p38alpha have decreased MMP-9 activity and exhibit decreased bone metastasis in mice. Clin Exp Metastasis 21(6):525– 533 Kumar B, Sinclair J, Khandrika L, Koul S, Wilson S, Koul HK, 2009, Differential effects of MAPKs signaling on the growth of invasive bladder cancer cells. Int J Oncol 34(6):1557–1564 Kumar B, Koul S, Petersen J, Khandrika L, Hwa JS, Meacham RB, Wilson S, Koul HK, 2010, p38 mitogen-activated protein kinasedriven MAPKAPK2 regulates invasion of bladder cancer by modulation ofMMP-2 andMMP-9 activity. Cancer Res 70(2):832–841. , DOI 10.1158/0008-5472.can-09-2918 Huang Q, Lan F,Wang X, Yu Y, Ouyang X, Zheng F, Han J, Lin Y, Xie Y, Xie F, Liu W, Yang X, Wang H, Dong L, Wang L, Tan J, 2014, IL-1beta-induced activation of p38 promotes metastasis in gastric adenocarcinoma via upregulation of AP-1/c-fos,MMP2 and MMP9. Mol Cancer 13:18. , DOI 10.1186/1476-4598-13- 18 Cui XP, Qin CK, Zhang ZH, Su ZX, Liu X, Wang SK, Tian XS, 2014, HOXA10 promotes cell invasion and MMP-3 expression via TGFbeta2-mediated activation of the p38 MAPK pathway in pancreatic cancer cells. Dig Dis Sci 59(7):1442–1451. https://doi. org/10.1007/s10620-014-3033-6 Cheng TL, Symons M, Jou TS, 2004, Regulation of anoikis by Cdc42 and Rac1. Exp Cell Res 295(2):497–511. , DOI 10.1016/j.yexcr.2004.02.002 Avisetti DR, Babu KS, Kalivendi SV, 2014, Activation of p38/JNK pathway is responsible for embelin induced apoptosis in lung cancer cells: transitional role of reactive oxygen species. PLoS One 9(1):e87050. , DOI 10.1371/journal.pone.0087050 Bulavin DV, Fornace AJ Jr, 2004, p38MAP kinase's emerging role as a tumor suppressor. Adv Cancer Res 92:95–118. , DOI 10.1016/s0065-230x(04)92005-2 Vega GG, Aviles-Salas A, Chalapud JR, Martinez-Paniagua M, Pelayo R, Mayani H, Hernandez-Pando R, Martinez-Maza O, Huerta-Yepez S, Bonavida B, Vega MI, 2015, P38 MAPK expression and activation predicts failure of response to CHOP in patients with diffuse large B-cell lymphoma. BMC Cancer 15:722. https:// doi.org/10.1186/s12885-015-1778-8 Martinez-Useros J, Georgiev-Hristov T, Borrero-Palacios A, Fernandez-Acenero MJ, Rodriguez-Remirez M, del Puerto- Nevado L, Cebrian A, Gomez del Pulgar MT, Cazorla A, Vega- Bravo R, Perez N, Celdran A, Garcia-Foncillas J, 2015, Identification of poor-outcome Biliopancreatic carcinoma patients with two-marker signature based on ATF6alpha and p-p38 "STARD compliant". Medicine, Baltimore, 94(45):e1972. https:// doi.org/10.1097/md.0000000000001972 Li C, Johnson DE, 2012, Bortezomib induces autophagy in head and neck squamous cell carcinoma cells via JNK activation. Cancer Lett 314(1):102–107. , DOI 10.1016/j.canlet.2011.09.020 Hour MJ, Lee KT, Wu YC, Wu CY, You BJ, Chen TL, Lee HZ, 2013, A novel antitubulin agent, DPQZ, induces cell apoptosis in human oral cancer cells through Ras/Raf inhibition and MAP kinases activation. Arch Toxicol 87(5):835–846. , DOI 10. 1007/s00204-012-0991-3 Zhang S,Wang XL, Gan YH, Li SL, 2010, Activation of c-Jun Nterminal kinase is required for mevastatin-induced apoptosis of salivary adenoid cystic carcinoma cells. Anti-Cancer Drugs 21(7): 678–686 Shen F, Fan X, Liu B, Jia X, Gao A, Du H, Ye M, You B, Huang C, Shi X, 2008, Downregulation of cyclin D1-CDK4 protein in human embryonic lung fibroblasts, HELF, induced by silica is mediated through the ERK and JNK pathway. Cell Biol Int 32(10): 1284–1292. , DOI 10.1016/j.cellbi.2008.07.015 Stegeman H, Kaanders JH, Verheijen MM, Peeters WJ, Wheeler DL, Iida M, Grenman R, van der Kogel AJ, Span PN, Bussink J, 2013, Combining radiotherapy with MEK1/2, STAT5 or STAT6 inhibition reduces survival of head and neck cancer lines. Mol Cancer 12(1):133. , DOI 10.1186/1476-4598-12-133 Fletcher EV, Love-Homan L, Sobhakumari A, Feddersen CR, Koch AT, Goel A, Simons AL, 2013, EGFR inhibition induces proinflammatory cytokines via NOX4 in HNSCC. Mol Cancer Res 11(12): 1574–1584. , DOI 10.1158/1541-7786.mcr-13-0187 Duffey D, Dolgilevich S, Razzouk S, Li L, Green R, Gorti GK, 2011, Activating transcription factor-2 in survival mechanisms in head and neck carcinoma cells. Head Neck 33(11):1586–1599. , DOI 10.1002/hed.21648 KimJE, Lee JI, JinDH, Lee WJ, Park GB, KimS, KimYS,Wu TC, Hur DY, Kim D, 2014, Sequential treatment of HPV E6 and E7- expressing TC-1 cells with bortezomib and celecoxib promotes apoptosis through p-p38 MAPK-mediated downregulation of cyclin D1 and CDK2. Oncol Rep 31(5):2429–2437. , DOI 10.3892/or.2014.3082 Park SW, Kim HS, Hah JW, JeongWJ, Kim KH, SungMW(2010, Celecoxib inhibits cell proliferation through the activation of ERK and p38 MAPK in head and neck squamous cell carcinoma cell lines. Anti-Cancer Drugs 21(9):823–830. , DOI 10.1097/ CAD.0b013e32833dada8 Lin Y, Mallen-St Clair J, Wang G, Luo J, Palma-Diaz F, Lai C, Elashoff DA, Sharma S, Dubinett SM, St John M, 2016, p38 MAPK mediates epithelial-mesenchymal transition by regulating p38IP and Snail in head and neck squamous cell carcinoma. Oral Oncol 60:81–89. , DOI 10.1016/j.oraloncology.2016.06. 0106. Deraz EM, Kudo Y, Yoshida M, Obayashi M, Tsunematsu T, Tani H, Siriwardena SB, Keikhaee MR, Qi G, Iizuka S, Ogawa I, Campisi G, Lo Muzio L, Abiko Y, Kikuchi A, Takata T, 2011, MMP-10/stromelysin-2 promotes invasion of head and neck cancer. PLoS One 6(10):e25438 Chang CM, Chang PY, Tu MG, Lu CC, Kuo SC, Amagaya S, Lee CY, Jao HY, Chen MY, Yang JS, 2012, Epigallocatechin gallate sensitizes CAL-27 human oral squamous cell carcinoma cells to the anti-metastatic effects of gefitinib, Iressa, via synergistic suppression of epidermal growth factor receptor and matrix metalloproteinase- 2. Oncol Rep 28(5):1799–1807. , DOI 10.3892/or. 2012.1991 Schuettler D, Piontek G,Wirth M, Haller B, Reiter R, Brockhoff G, Pickhard A, 2015, Selective inhibition of EGFR downstream signaling reverses the irradiation-enhanced migration ofHNSCC cells. Am J Cancer Res 5(9):2660–2672 Platanias LC, 2003, The p38 mitogen-activated protein kinase pathway and its role in interferon signaling. Pharmacol Ther 98(2):129–142 Tanabe K, Kozawa O, IidaH(2016, cAMP/PKA enhances interleukin- 1beta-induced interleukin-6 synthesis through STAT3 in glial cells. Cell Signal 28(1):19–24. , DOI 10.1016/j.cellsig.2015.10.009 Riebe C, Pries R, Schroeder KN, Wollenberg B, 2011, Phosphorylation of STAT3 in head and neck cancer requires p38 MAPKinase, whereas phosphorylation of STAT1 occurs via a different signaling pathway. Anticancer Res 31(11):3819–3825 Enslen H, Raingeaud J, Davis RJ, 1998, Selective activation of p38 mitogen-activated protein, MAP, kinase isoforms by the MAP kinase kinases MKK3 and MKK6. J Biol Chem 273(3):1741–1748 Hu MC, Wang YP, Mikhail A, Qiu WR, Tan TH, 1999, Murine p38-delta mitogen-activated protein kinase, a developmentally regulated protein kinase that is activated by stress and proinflammatory cytokines. J Biol Chem 274(11):7095–7102 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 183 EP 192 DI 10.1007/s12253-018-0405-9 PG 10 ER PT J AU Damodaran, M Paul, FDS Venkatesan, V AF Damodaran, Mohan Paul, F D Solomon Venkatesan, Vettriselvi TI Genetic Polymorphisms in miR-146a, miR-196a2 and miR-125a Genes and its Association in Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microRNA; Prostate cancer; Genotyping; Haplotypes ID microRNA; Prostate cancer; Genotyping; Haplotypes AB The increase in incidence of prostate cancer in the Indian Population stresses the need to identify genetic markers for susceptibility and prognosis. Recent studies show that microRNAs play an important role in tumorigenesis by altering proliferation, differentiation and cell death. Gene polymorphisms not only in promoter region but also within miRNA gene have been shown to affect expression. The present study was aimed to analyze the role of miR-146a, miR-196a2 and miR-125a gene polymorphisms in prostate cancer. Genotyping of three SNPs rs73318382, rs57095329, rs2910164 in miRNA146a, rs11614913 in miR-196a2 and rs41275794, rs12976445, rs10404453 and rs1297533 in miR-125a was performed in 100 cases and 100 controls. Statistical analysis revealed the heterozygous AG genotype of the rs57095329 was significantly decreased in the cases when compared to the controls (OR-0.45, CI -0.24 to 0.85, p value-0.02) indicating an inverse association of this genotype with prostate cancer. Further the heterozygous CT of miR-196a2 (rs11614913) (OR-1.88, CI-1.06 to 3.35, p-0.02) and homozygous CC of miR-125a (rs12976445) (OR-2.55, CI -1.15 to 4.65, p-0.03) showed increased risk for prostate cancer. Combined analysis of all the genotypes revealed that the haplotype combination AGGCGTGG (OR = 0.09 at CI 95% (0.01–0.65) showed an inverse association with prostate cancer. Stratified analysis based on the age and tumor grade revealed no significant association. C1 [Damodaran, Mohan] Sri Ramachandra University, Department of Human Genetics, 600116 Chennai, India. [Paul, F D Solomon] Sri Ramachandra University, Department of Human Genetics, 600116 Chennai, India. [Venkatesan, Vettriselvi] Sri Ramachandra University, Department of Human Genetics, 600116 Chennai, India. RP Venkatesan, V (reprint author), Sri Ramachandra University, Department of Human Genetics, 600116 Chennai, India. EM vettriselviv@yahoo.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Lalitha K, Suman G, Pruthvish S, Mathew A, Murthy NS, 2012, Estimation of time trends of incidence of prostate canner-an Indian scenario. Asian Pac J Cancer Prev 13(12):6245–6250 Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T, 2001, Identification of novel genes coding for small expressed RNAs. Science 294(5543):853–858 Esquela-Kerscher A, Slack FJ, 2006, Oncomirs—microRNAs with a role in cancer. Nat Rev Cancer 6(4):259–269 Cho WC, 2007, OncomiRs: the discovery and progress of microRNAs in cancers. Mol Cancer 6(1):60 Sun J, Lu H, Wang X, Jin H, 2013, MicroRNAs in hepatocellular carcinoma: regulation, function, and clinical implications. Sci World J 2013:1–14 Lu J, Gu H, Tang Q,WuW, Yuan B, Guo D, Hu L, 2016, Common SNP in hsa-miR-196a-2 increases hsa-miR-196a-5p expression and predisposes to idiopathic male infertility in Chinese Han population. Sci Rep 6 Sun Q, Zhao X, Liu X,Wang Y, Huang J, Jiang B et al, 2014, miR- 146a functions as a tumor suppressor in prostate cancer by targeting Rac1. Prostate 74(16):1613–1621 Lin SL, Chiang A, Chang D, Ying SY, 2008, Loss of mir-146a function in hormone-refractory prostate cancer. RNA 14(3):417–424 Chen C, Zhang Y, Zhang L, Weakley SM, Yao Q, 2011, MicroRNA-196: critical roles and clinical applications in development and cancer. J Cell Mol Med 15(1):14–23 Hoffman AE, Zheng T, Yi C, Leaderer D,Weidhaas J, Slack F et al, 2009, microRNA miR-196a-2 and breast cancer: a genetic and epigenetic association study and functional analysis. Cancer Res 69(14):5970–5977 Hu Z, Liang J,Wang Z, Tian T, Zhou X, Chen J, Miao R,Wang Y, Wang X, Shen H, 2009, Common genetic variants in premicroRNAs were associated with increased risk of breast cancer in Chinese women. Hum Mutat 30:79–84 Wang J,Wang Q, Liu H, Shao N, Tan B, Zhang G, Cheng Y, 2012, The association of miR-146a rs2910164 and miR-196a2 rs11614913 polymorphisms with cancer risk: a meta-analysis of 32 studies. Mutagenesis 27(6):779–788 Scott GK, Goga A, Bhaumik D, Berger CE, Sullivan CS, Benz CC, 2007, Coordinate suppression of ERBB2 and ERBB3 by enforced expression of micro-RNA miR-125a or miR-125b. J Biol Chem 282(2):1479–1486 Bi Q, Tang S, Xia L, Du R, Fan R, Gao L, Nie Y, 2012, Ectopic expression of MiR-125a inhibits the proliferation and metastasis of hepatocellular carcinoma by targeting MMP11 and VEGF. PLoS One 7(6):e40169 Ninio-Many L, Grossman H, Levi M, Zilber S, Tsarfaty I, Shomron N et al, 2014)MicroRNA miR-125a-3p modulates molecular pathway of motility and migration in prostate cancer cells. Oncoscience 1(4):250 Luo X, Yang W, Ye DQ, Cui H, Zhang Y, Hirankarn N, Qian X, Tang Y, Lau YL, De Vries N, Tak PP, 2011, A functional variant in microRNA-146a promoter modulates its expression and confers disease risk for systemic lupus erythematosus. PLoS Genet 7(6): e1002128 Zhou B, Rao L, Peng Y,Wang Y, Chen Y, Song Y, Zhang L, 2010, Common genetic polymorphisms in pre-microRNAs were associated with increased risk of dilated cardiomyopathy. Clin Chim Acta 411(17):1287–1290 Williams, A. E., Perry,M. M.,Moschos, S. A., Larner-Svensson, H. M., & Lindsay, M. A., 2008). Role of miRNA-146a in the regulation of the innate immune response and cancer Sole X, Guino E, Valls J, Iniesta R, Moreno V, 2006, SNPStats: a web tool for the analysis of association studies. Bioinformatics 22(15):1928–1929 Lu, J., Getz, G.,Miska, E. A., Alvarez-Saavedra, E., Lamb, J., Peck, D. … & Downing, J. R., 2005). MicroRNA expression profiles classify human cancers nature, 435(7043), 834–838 Lv M, Dong W, Li L, Zhang L, Su X, Wang L, Gao L, Zhang L, 2013, Association between genetic variants in pre-miRNA and colorectal cancer risk in a Chinese population. J Cancer Res Clin Oncol 139(8):1405–1410 Zhang X, Gu Y, Liu X, Yu Y, Shi J, Yu Q, Sun H, Kanu JS, Zhan S, Liu Y, 2015, Association of pre-miR-146a rs2910164 polymorphism with papillary thyroid cancer. Int J Endocrinol 2015:1–9 Chae YS, Kim JG, Lee SJ, Kang BW, Lee YJ, Park JY, Jeon HS, Park JS, Choi GS, 2013, A miR-146a polymorphism, rs2910164, predicts risk of and survival from colorectal cancer. Anticancer Res 33(8):3233–3239 Williams AE, Perry MM, Moschos SA, Larner-Svensson HM, Lindsay MA, 2008, Role of miRNA-146a in the regulation of the innate immune response and cancer. Biochem Soc Trans 36:1211– 1215 Kati PP, Minja JP,Waltering KK et al, 2007, microRNA expression profiling in prostate cancer. Cancer Res 67(13):6130–6135 Shi-Lung L, Angela C, Donald C, Shao-Yao Y, 2008, Loss ofmiR- 146a function in hormone-refractory prostate cancer. RNA 14:417– 424 Xu B,Wang N,Wang X, Tong N, Shao N, Tao J, Li P, Niu X, Feng N, Zhang L, Hua L,Wang Z, ChenM, 2012, miR-146a suppresses tumor growth and progression by targeting EGFR pathway and in a p-ERK- dependent manner in castration-resistant prostate cancer. Prostate 72(11):1171–1178 George GP, Gangwar R, Mandal RK, Sankhwar SN, Mittal RD, 2011, Genetic variation in microRNA genes and prostate cancer risk in north Indian population. Mol Biol Rep 38(3):1609–1615 Xu B, Feng NH, Li PC, Tao J,Wu D, Zhang ZD, Tong N,Wang JF, Song NH, ZhangW, Hua LX, 2010, A functional polymorphism in pre-miR-146a gene is associated with prostate cancer risk and mature miR-146a expression in vivo. Prostate 70(5):467–472 Zhu L, Chu H, Gu D, Ma L, Shi D, Zhong D, Tong N, Zhang Z, Wang M, 2012, A functional polymorphism in miRNA-196a2 is associated with colorectal cancer risk in a Chinese population.DNA Cell Biol 31(3):350–354 Rodriguez A, Griffiths-Jones S, Ashurst JL, Bradley A, 2004, Identification of mammalian microRNA host genes and transcription units. Genome Res 14(10a):1902–1910 Mattie MD, Benz CC, Bowers J, Sensinger K, Wong L, Scott GK, Fedele V, Ginzinger D, Getts R, Haqq C, 2006, Optimized highthroughput microRNA expression profiling provides novel biomarker assessment of clinical prostate and breast cancer biopsies. Mol Cancer 5:24 Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, Menard S, Palazzo JP, Rosenberg A,Musiani P, Volinia S, Nenci I, Calin GA, Querzoli P, Negrini M, Croce CM, 2005, MicroRNA gene expression deregulation in human breast cancer. Cancer Res 65:7065–7070 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 193 EP 200 DI 10.1007/s12253-018-0412-x PG 8 ER PT J AU Cui, K Zou, H Shi, M Ou, Y Han, L Zhang, B Hu, D Li, Sh AF Cui, Kai Zou, Hongsheng Shi, Mingliang Ou, Yang Han, Lu Zhang, Bo Hu, Dawei Li, Sheng TI Gene Expression Profiles in Chemokine (C-C Motif) Ligand 21-Overexpressing Pancreatic Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chemokine (C-C motif) ligand 21; Pancreatic cancer cell; Lentiviral transduction; DNA microarray; Matrix metallopeptidase-9 ID Chemokine (C-C motif) ligand 21; Pancreatic cancer cell; Lentiviral transduction; DNA microarray; Matrix metallopeptidase-9 AB Chemokine (C-C Motif) ligand 21 (CCL21) plays an important role in tumor immunity. However, the molecular mechanisms by which CCL21 regulates tumor immunity remain largely unknown. In this study, we successfully generated a lentiviral vector expressing human CCL21 (Lenti-hCCL21), which was confirmed by biological assays. The Lenti-hCCL21 was transduced into PANC-1 cells, a chemokine (C-C motif) receptor 7 (CCR7)-positive human pancreatic cancer cell line. We used the scratch wound and transwell assays to measure cell migration of the CCL21-overexpressing PANC-1 cells. A DNA microarray assay was performed to determine gene expression profiles. The results showed that CCL21 lentiviral transduction significantly up- or down-regulated a panel of tumor-associated genes, although CCL21 appeared to have no effect on PANC-1 cell migration. Importantly, CCL21 promoted matrix metallopeptidase-9 (MMP-9) expression in PANC-1 cells. CCL21 regulates pancreatic cancer immunity possibly through governing the expression of a panel of tumor-associated genes, including MMP-9. C1 [Cui, Kai] Shandong Cancer Hospital Affiliated to Shandong University, 250117 Jinan, Shandong, China. [Zou, Hongsheng] People’s Hospital of Rongcheng, 264300 Rongcheng, Shandong, China. [Shi, Mingliang] People’s Hospital of Laiwu City, 271199 Laiwu, Shandong, China. [Ou, Yang] Shandong Cancer Hospital Affiliated to Shandong University, 250117 Jinan, Shandong, China. [Han, Lu] Shandong Cancer Hospital Affiliated to Shandong University, 250117 Jinan, Shandong, China. [Zhang, Bo] Shandong Cancer Hospital Affiliated to Shandong University, 250117 Jinan, Shandong, China. [Hu, Dawei] Shandong Cancer Hospital Affiliated to Shandong University, 250117 Jinan, Shandong, China. [Li, Sheng] Shandong Cancer Hospital Affiliated to Shandong University, 250117 Jinan, Shandong, China. RP Hu, D (reprint author), Shandong Cancer Hospital Affiliated to Shandong University, 250117 Jinan, China. EM ckjason@sohu.com CR Yadav D, Lowenfels AB, 2013, The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology 144(6):1252–1261. https:// doi.org/10.1053/j.gastro.2013.01.068 Sozzani S, Locati M, Allavena P, Van Damme J, Mantovani A, 1996, Chemokines: a superfamily of chemotactic cytokines. Int J Clin Lab Res 26(2):69–82 Griffith JW, Sokol CL, Luster AD, 2014, Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu Rev Immunol 32:659–702. , DOI 10.1146/annurevimmunol- 032713-120145 Gunn MD, Tangemann K, Tam C, Cyster JG, Rosen SD, Williams LT, 1998, A chemokine expressed in lymphoid high endothelial venules promotes the adhesion and chemotaxis of naive T lymphocytes. Proc Natl Acad Sci U S A 95(1):258–263 Warnock RA, Campbell JJ, DorfME,Matsuzawa A,McEvoy LM, Butcher EC, 2000, The role of chemokines in the microenvironmental control of T versus B cell arrest in Peyer's patch high endothelial venules. J Exp Med 191(1):77–88 Willimann K, Legler DF, Loetscher M, Roos RS, Delgado MB, Clark-Lewis I, Baggiolini M, Moser B, 1998, The chemokine SLC is expressed in T cell areas of lymph nodes and mucosal lymphoid tissues and attracts activated T cells via CCR7. Eur J Immunol 28(6):2025–2034. , DOI 10.1002/(SICI)1521- 4141(199806)28:06<2025::AID-IMMU2025>3.0.CO;2-C Kirk CJ, Hartigan-O'Connor D, Mule JJ, 2001, The dynamics of the T-cell antitumor response: chemokine-secreting dendritic cells can prime tumor-reactive T cells extranodally. Cancer Res 61(24): 8794–8802 Ahn GO, Brown JM, 2008, Matrix metalloproteinase-9 is required for tumor vasculogenesis but not for angiogenesis: role of bone marrow-derived myelomonocytic cells. Cancer Cell 13(3):193– 205. , DOI 10.1016/j.ccr.2007.11.032 Nagakawa Y, Aoki T, Kasuya K, Tsuchida A, Koyanagi Y, 2002, Histologic features of venous invasion, expression of vascular endothelial growth factor and matrix metalloproteinase-2 and matrix metalloproteinase-9, and the relation with liver metastasis in pancreatic cancer. Pancreas 24(2):169–178 Wang J, Zhang X, Thomas SM, Grandis JR, Wells A, Chen ZG, Ferris RL, 2005, Chemokine receptor 7 activates phosphoinositide- 3 kinase-mediated invasive and prosurvival pathways in head and neck cancer cells independent of EGFR. Oncogene 24(38):5897– 5904. , DOI 10.1038/sj.onc.1208740 Sanchez-Sanchez N, Riol-Blanco L, de la Rosa G, Puig-Kroger A, Garcia-Bordas J, Martin D, Longo N, Cuadrado A, Cabanas C, Corbi AL, Sanchez-Mateos P, Rodriguez-Fernandez JL, 2004, Chemokine receptor CCR7 induces intracellular signaling that inhibits apoptosis of mature dendritic cells. Blood 104(3):619–625. , DOI 10.1182/blood-2003-11-3943 Sancho M, Vieira JM, Casalou C, Mesquita M, Pereira T, Cavaco BM, Dias S, Leite V, 2006, Expression and function of the chemokine receptor CCR7 in thyroid carcinomas. J Endocrinol 191(1): 229–238. , DOI 10.1677/joe.1.06688 Sperveslage J, Frank S, Heneweer C, Egberts J, Schniewind B, Buchholz M, Bergmann F, Giese N, Munding J, Hahn SA, Kalthoff H, Kloppel G, Sipos B, 2012, Lack of CCR7 expression is rate limiting for lymphatic spread of pancreatic ductal adenocarcinoma. Int J Cancer 131(4):E371–E381. https://doi. org/10.1002/ijc.26502 Roy I, Zimmerman NP, Mackinnon AC, Tsai S, Evans DB, Dwinell MB, 2014, CXCL12 chemokine expression suppresses human pancreatic cancer growth and metastasis. PLoS One 9(3):e90400. , DOI 10.1371/journal.pone.0090400 Zhao B, Cui K, Wang CL, Wang AL, Zhang B, Zhou WY, Zhao WH, Li S, 2011, The chemotactic interaction between CCL21 and its receptor, CCR7, facilitates the progression of pancreatic cancer via induction of angiogenesis and lymphangiogenesis. J Hepatobiliary Pancreat Sci 18(6):821–828. , DOI 10. 1007/s00534-011-0395-4 Lin Y, Sharma S, John MS, 2014, CCL21 cancer immunotherapy. Cancers, Basel, 6(2):1098–1110. , DOI 10.3390/ cancers6021098 Turnquist HR, Lin X, Ashour AE, Hollingsworth MA, Singh RK, Talmadge JE, Solheim JC, 2007, CCL21 induces extensive intratumoral immune cell infiltration and specific anti-tumor cellular immunity. Int J Oncol 30(3):631–639 Zhang L,Wang D, Li Y, Liu Y, Xie X,Wu Y, Zhou Y, Ren J, Zhang J, Zhu H, Su Z, 2016, CCL21/CCR7 Axis contributed to CD133+ pancreatic cancer stem-like cell metastasis via EMT and Erk/NFkappaB pathway. PLoS One 11(8):e0158529. , DOI 10. 1371/journal.pone.0158529 Xu Y, Li Z, Jiang P,Wu G, Chen K, Zhang X, Li X, 2015, The coexpression of MMP-9 and Tenascin-C is significantly associated with the progression and prognosis of pancreatic cancer. Diagn Pathol 10:211. , DOI 10.1186/s13000-015-0445-3 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 201 EP 208 DI 10.1007/s12253-018-0390-z PG 8 ER PT J AU Wang, Z Zhang, L Wan, Z He, Y Huang, H Xiang, H Wu, X Zhang, K Liu, Y Goodin, S Du, Z Zheng, X AF Wang, Zhenshi Zhang, Lanyue Wan, Zheng He, Yan Huang, Huarong Xiang, Hongping Wu, Xiaofeng Zhang, Kun Liu, Yang Goodin, Susan Du, Zhiyun Zheng, Xi TI Atorvastatin and Caffeine in Combination Regulates Apoptosis, Migration, Invasion and Tumorspheres of Prostate Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Atorvastatin; Apoptosis; Caffeine; Combination treatment; Prostate cancer ID Atorvastatin; Apoptosis; Caffeine; Combination treatment; Prostate cancer AB Atorvastatin is the most prescribed cholesterol-lowering statin, while caffeine enhances chemo-sensitivity and induces apoptosis of tumor cells through its DNA repair-inhibiting effect. The present study investigated the effects and mechanisms of atorvastatin and caffeine in combination on human prostate cancer cells cultured in vitro. Cell growth were determined by the trypan blue exclusion assay. The cell apoptosis and colony formation were determined by morphological assessment. The ability of cell migration and invasion were performed using a scratch wound-healing and Transwell assay. Tumorspheres were formed in suspension under the condition of non-adherence and serum-free medium. Finally, the western blot assay was used to determine the levels of proteins. The combination synergistically suppressed proliferation and induced apoptotic death. Meanwhile, the migration, invasion, and the formation of tumorspheres were significantly inhibited by the combination. We found that atorvastatin and caffeine in combination downregulated phospho-Akt, phospho-Erk1/2, anti-apoptotic Bcl-2 and Survivin protein levels. Results of the present study indicate treatment with the combination of caffeine and atorvastatin may be an effective strategy for inhibiting the growth of prostate cancer and should be evaluated clinically. C1 [Wang, Zhenshi] Guangdong University of Technology, Allan H. Conney Laboratory for Anticancer Research, 510006 Guangzhou, China. [Zhang, Lanyue] Guangdong University of Technology, Allan H. Conney Laboratory for Anticancer Research, 510006 Guangzhou, China. [Wan, Zheng] Rutgers, The State University of New Jersey, Ernest Mario School of Pharmacy, Department of Chemical Biology, 08854 Piscataway, NJ, USA. [He, Yan] Guangdong University of Technology, Allan H. Conney Laboratory for Anticancer Research, 510006 Guangzhou, China. [Huang, Huarong] Guangdong University of Technology, Allan H. Conney Laboratory for Anticancer Research, 510006 Guangzhou, China. [Xiang, Hongping] Guangdong University of Technology, School of Materials and Energy, Guangdong Provincial Key Laboratory of Functional Soft Condensed Matter, 510006 Guangzhou, China. [Wu, Xiaofeng] Guangdong University of Technology, Allan H. Conney Laboratory for Anticancer Research, 510006 Guangzhou, China. [Zhang, Kun] Guangdong University of Technology, Allan H. Conney Laboratory for Anticancer Research, 510006 Guangzhou, China. [Liu, Yang] Xiamen University, Medical college, Department of Nursing, 361000 Xiamen, China. [Goodin, Susan] Rutgers Cancer Institute of New Jersey, 08903 New Brunswick, NJ, USA. [Du, Zhiyun] Guangdong University of Technology, Allan H. Conney Laboratory for Anticancer Research, 510006 Guangzhou, China. [Zheng, Xi] Guangdong University of Technology, Allan H. Conney Laboratory for Anticancer Research, 510006 Guangzhou, China. RP Du, Z (reprint author), Guangdong University of Technology, Allan H. Conney Laboratory for Anticancer Research, 510006 Guangzhou, China. EM zhiyundugdut@foxmail.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics. CA Cancer J Clin 66(1):7–30 Yap TA, Zivi A, Omlin A, De Bono JS, 2011, The changing therapeutic landscape of castration-resistant prostate cancer. Nat Rev Clin Oncol 8(10):597–610 Francini E, Sweeney CJ, 2016, Docetaxel activity in the era of lifeprolonging hormonal therapies for metastatic castration-resistant prostate Cancer. Eur Urol 70:410–412 Amable L, 2016, Cisplatin resistance and opportunities for precision medicine. Pharmacol Res 106:27–36 Zhang L, Li L, Jiao M, Wu D, Wu K, Li X, Zhu G, Yang L, Wang X, Wang X, Hsieh J, He D, 2012, Genistein inhibits the stemness properties of prostate cancer cells through targeting hedgehog–Gli1 pathway. Cancer Lett 323(1):48–57 Peng M, Darko KO, Tao T, Huang Y, Su Q, He C, Yin T, Liu Z, Yang X, 2017, Combination of metformin with chemotherapeutic drugs via different molecular mechanisms. Cancer Treat Rev 54: 24–33. , DOI 10.1016/j.ctrv.2017.01.005 Bansal D, Undela K, D'Cruz S, Schifano F, 2012, Statin use and risk of prostate cancer: a meta-analysis of observational studies. PLoS One 7(10):e46691 Flick ED, Habel LA, Chan KA, Van Den Eeden SK, Quinn VP, Haque R, Orav EJ, Seeger JD, Sadler MC, Quesenberry Jr CP, Sternfeld B, Jacobsen SJ, Whitmer RA, Caan BJ, 2007, Statin use and risk of prostate cancer in the California Men's health study cohort. Cancer Epidemiol Prev Biomarkers 16(11):2218–2225 Lustman A, Nakar S, Cohen AD, Vinker S, 2014, Statin use and incident prostate cancer risk: does the statin brand matter? A population-based cohort study. Prostate Cancer Prostatic Dis 17: 6–9 Boudreau DM, Yu O, Buist DS, Miglioretti DL, 2008, Statin use and prostate cancer risk in a large population-based setting. Cancer Causes Control 19:767–774 Allott EH, Howard LE, Cooperberg MR, Kane CJ, Aronson WJ, Terris MK, Amling CL, Freedland SJ, 2014, Postoperative statin use and risk of biochemical recurrence following radical prostatectomy: results from the shared equal access regional Cancer hospital, SEARCH, database. BJU Int 114:661–666 Yu O, Eberg M, Benayoun S, Aprikian A, Batist G, Suissa S, Azoulay L, 2013, Use of statins and the risk of death in patients with prostate cancer. J Clin Oncol 32(1):5–11 Wang C, Tao W, Wang Y, Bikow J, Lu B, Keating A, Verma S, Parker TG, Han R,Wen XY, 2010, Rosuvastatin, identified from a zebrafish chemical genetic screen for antiangiogenic compounds, suppresses the growth of prostate cancer. Eur Urol 58(3):418–426 Xie F, Liu J, Li C, Zhao Y, 2016, Simvastatin blocks TGF-β1- induced epithelial-mesenchymal transition in human prostate cancer cells. Oncol Lett 11(5):3377–3383 Chen X, Liu Y,Wu J, Huang H, Du Z, Zhang K, Zhou D, Hung K, Goodin S, Zheng X, 2016, Mechanistic study of inhibitory effects of atorvastatin and docetaxel in combination on prostate Cancer. Cancer Genomics Proteomics 13:151–160 Zheng X, Cui XX, Gao Z, Zhao Y, Liu Y, ShihWJ, HuangMT, Liu Y, Rabson A, Reddy B, Yang CS, Conney AH, 2010, Atorvastatin and celecoxib in combination inhibits the progression of androgendependent LNCaP xenograft prostate tumors to androgen independence. Cancer Prev Res 3:114–124 Zheng X, Cui XX, Avila GE, HuangMT, Liu Y, Patel J, Kong AN, Paulino R, Shih WJ, Lin Y, Rabson AB, Reddy BS, Conney AH, 2007, Atorvastatin and celecoxib inhibit prostate PC-3 tumors in immunodeficient mice. Clin Cancer Res 13:5480–5487 Sabisz M, Skladanowski A, 2008, Modulation of cellular response to anticancer treatment by caffeine: inhibition of cell cycle checkpoints, DNA repair and more. Curr Pharm Biotechnol 9(4):325– 336 Tomita K, Tsuchiya H, Sasaki T, 1989, DNA repair and drug resistance: enhancement of the effects of anticancer agents by DNA repair inhibitors. Gan To Kagaku Ryoho Cancer Chemother 16(3Pt2):576–584 Li A,Wu N, Zou H, Zhu B, Xiong S, Xiao G, 2016, Low concentration of caffeine inhibits cell viability,migration and invasion, and induces cell apoptosis of B16F10 melanoma cells. Int J Clin Exp Pathol 9(11):11206–11213 Okano J, Nagahara T, Matsumoto K, Murawaki Y, 2008, Caffeine inhibits the proliferation of liver cancer cells and activates the MEK/ERK/EGFR signalling pathway. Basic Clin Pharmacol Toxicol 102(6):543–551 ZhengX, Cui X, HuangM, LiuY,Wagner GC, LinY, ShihWJ, Lee M, Yang CS, Conney AH, 2012, Inhibition of progression of androgen-dependent prostate LNCaP tumors to androgen independence in SCID mice by oral caffeine and voluntary exercise. Nutr Cancer 64(7):1029–1037 Chen Z, Chen S, Lu G, Chen X, 2012, Phosphorus limitation for the colony formation, growth and photosynthesis of an edible cyanobacterium, Nostoc sphaeroides. Biotechnol Lett 34(1):137–143 Wang ZS, Huang HR, Zhang LY, Kim S, He Y, Li DL, Farischon C, Zhang K, Zheng X, Du ZY, Goodin S, 2017, Mechanistic Study of Inhibitory Effects of Metformin and Atorvastatin in Combination on Prostate Cancer Cells in Vitro and in Vivo. Biol Pharm Bull 40(8):1247 Yu L,Wu X, ChenM, Huang H, He Y,Wang H, Li D, Du Z, Zhang K, Goodin S, Zheng X, 2017, The effects and mechanism of YK-4- 279 in combination with docetaxel on prostate Cancer. Int J Med Sci 14(4):356–366 Farnier M, Davignon J, 1998, Current and future treatment of hyperlipidemia: the role of statins. Am J Cardiol 82:3J–10J Trialists CT, 2005, Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90056 participants in 14 randomised trials of statins. Lancet 366:1267–1278 McFarlane SI, Muniyappa R, Francisco R, Sowers JR, 2002, Pleiotropic effects of statins: lipid reduction and beyond. J Clin Endocrinol Metab 87:1451–1458 Kim JH, Cox ME, Wasan KM, 2014, Effect of simvastatin on castration-resistant prostate cancer cells. Lipids Health Dis 13:56 Goldstein JL, Brown MS, 1990, Regulation of the mevalonate pathway. Nature 343:425–430 Pruitt K, Der CJ, 2001, Ras and rho regulation of the cell cycle and oncogenesis. Cancer Lett 171:1–10 Sarkaria JN, Busby EC, Tibbetts RS, Roos P, Taya Y, Karnitz LM, Abraham RT, 1999, Inhibition of ATM and ATR kinase activities by the radiosensitizing agent, caffeine. Cancer Res 59(17):4375– 4382 Miwa S, Sugimoto N, Yamamoto N, Shirai T, Nishida H, Hayashi K,Kimura H, Takeuchi A, IgarashiK, Yachie A, TsuchiyaH, 2012, Caffeine induces apoptosis of osteosarcoma cells by inhibiting AKT/mTOR/S6K, NF-κB and MAPK pathways. Anticancer Res 32(9):3643–3649 He Z, Ma WY, Hashimoto T, Bode AM, Yang CS, Dong Z, 2003, Induction of apoptosis by caffeine is mediated by the p53, Bax, and caspase 3 pathways. Cancer Res 63(15):4396–4401 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 209 EP 216 DI 10.1007/s12253-018-0415-7 PG 8 ER PT J AU Zidi, S Sahli, M Mezlini, A Yacoubli-Loueslati, B AF Zidi, Sabrina Sahli, Mariem Mezlini, Amel Yacoubli-Loueslati, Besma TI Association of Combined Tobacco Smoking, Hormonal Contraceptive use and Status Matrimonial with Cervical Cancer Evolution in Tunisian Women SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; FIGO staging; Status matrimonial; Smoking; Hormonal contraceptive ID Cervical cancer; FIGO staging; Status matrimonial; Smoking; Hormonal contraceptive AB Status matrimonial, cigarette smoking and hormonal contraceptive (HC) use have been associated with cervical cancer (CC) establishment by influencing the CC carcinogenesis process. In the present study, we aim to confirm this correlation between these factors and the risk of CC occurrence among Tunisian population. To evaluate the role of matrimonial status, smoking and HC as cofactors of CC installation, we performed a random selection of 600 women from Salah Azeiz institute in Tunisia and a questionnaire was conducted by doctors for each patient. Logistic regression after adjustment for potential confounding factors, relative excess risk due to interaction (RERI) and synergy index (S) were used to evaluate the additive interaction. Subgroup analysis was conducted to examine whether the relative risks changed with CC stages. There were an excess risk among smoker patients and patient with HC use (p < 0.001) for CC installation. Women who are smokers have a 14 times greater risk of suffering from cervical cancer and approximately 24 times greater to develop an advanced form of CC malignancy. Having a history of using birth control pills increase CC occurrence and aggravation (OR~2). The matrimonial status seems an important factor for CC appearance (OR = 3.58 and 2.46) among CC Tunisian patient. However, no significant biological interaction from this three joint exposure was observed in the early FIGO stages but the risk increase in advanced FIGO stages. In our Tunisian cohort, oral contraception, smoking habit and matrimonial status are associated with an overall increased risk of CC development. Indeed, it may damage the local immunity system and may affect the disease severity in patient carriers of some genetic risk biomarkers. The balance of cancer risks may vary among Tunisian CC patient, depending on some environmental co-factors. C1 [Zidi, Sabrina] El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 2092 Tunis, Tunisia. [Sahli, Mariem] Charles Nicole HospitalTunis, Tunisia. [Mezlini, Amel] Salah Azeiz Oncology InstituteTunis, Tunisia. [Yacoubli-Loueslati, Besma] El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 2092 Tunis, Tunisia. RP Zidi, S (reprint author), El Manar University, Faculty of Sciences of Tunis, Department of Biology, Laboratory of Mycology, Pathologies and Biomarkers: LR16ES05, 2092 Tunis, Tunisia. EM ZIDISABRINA86@gmail.com CR Harper DM, Demars LR, 2014, Primary strategies for HPV infection and cervical cancer prevention. Clin Obstet Gynecol 57:256– 258 IARC, 2003, Monographs. International Agency for Research on Cancer, Lyon Castle PE,Wacholder S, LorinczAT, Scott DR, ShermanME, Glass AG et al, 2002, A prospective study of high-grade cervical neoplasia risk among human papillomavirus-infected women. J Natl Cancer Inst 94:1406–1414 Longatto-Filho A, Hammes LS, Sarian LO, Roteli-Martins C, Derchain SF, Erzen M et al, 2011, Hormonal contraceptives and the length of their use are not independent risk factors for high-risk HPVinfections or high-grade CIN. Gynecol Obstet Investig 71:93– 103 Harris TG, Miller L, Kulasingam SL, Feng Q, Kiviat NB, Schwartz SMet al, 2009, Depot-medroxyprogesterone acetate and combined oral contraceptive use and cervical neoplasia among women with oncogenic human papillomavirus infection. Am J Obstet Gynecol 200:489.e1–489.e8 Frega A, Scardamaglia P, Piazze J, Cerekja A, Pacchiarotti A, Verrico M et al, 2008, Oral contraceptives and clinical recurrence of human papillomavirus lesions and cervical intraepithelial neoplasia following treatment. Int J Gynaecol Obstet 100:175–178 Syrjanen K, Shabalova I, Petrovichev N, KozachenkoV, Zakharova T, Pajanidi J et al, 2006, Oral contraceptives are not an independent risk factor for cervical intraepithelial neoplasia or high-risk human papillomavirus infections. Anticancer Res 26:4729–4740 Gadducci A, Barsotti C, Cosio S, Domenici L, Riccardo Genazzani A, 2011, Smoking habit, immune suppression, oral contraceptive use, and hormone replacement therapy use and cervical carcinogenesis: a review of the literature. Gynecol Endocrinol 27:597–604 La Vecchia C, Boccia S. Oral contraceptives, human papillomavirus and cervical cancer. Eur J Cancer Prev 2014; 23:110–112 Poppe WA, Ide PS, Drijkoningen MP, Lauweryns JM, Van Assche FA, 1995, Tobacco smoking impairs the local immunosurveillance in the uterine cervix. An immunohistochemical study. Gynecol Obstet Investig 39:34–38 Poppe WA, Peeters R, Drijkoningen M, Ide PS, Daenens P, Lauweryns JM et al, 1996, Cervical cotinine and macrophage- Langerhans cell density in the normal human uterine cervix. Gynecol Obstet Investig 41:253–259 Oh HY, Kim MK, Seo S-S, Lee J-K, 2016, Association of Combined Tobacco Smoking and Oral Contraceptive use with Cervical Intraepithelial Neoplasia 2 or 3 in Korean Women. J Epidemiol 26(1):22–29 Zidi1 S, Sghaier1 I, Gazouani E, Mezlini A, Yacoubi-Loueslati B, 2015, Evaluation of Toll-Like Receptors 2/3/4/9 Gene Polymorphisms in Cervical Cancer Evolution. Pathol. Oncol. Res. , DOI 10.1007/s12253-015-0009-6 Pandey S, Mittal B, Srivastava M, Singh S, Srivastava K, Lal P, Mittal RD, 2011, Evaluation of toll-like receptors 3, c 1377C/T, and 9, G2848A, gene polymorphisms in cervical cancer susceptibility. Mol Biol Rep 38:4715–4721 Kim J, Kim BK, Lee CH, Seo SS, Park SY, Roh JW, 2012, Human papillomavirus genotypes and cofactors causing cervical intraepithelial neoplasia and cervical cancer in Korean women. Int J Gynecol Cancer 22:1570–1576 Kim JW, Song SH, Jin CH, Lee JK, Lee NW, Lee KW, 2012, Factors affecting the clearance of high-risk human papillomavirus infection and the progression of cervical intraepithelial neoplasia. J Int Med Res 40:486–496 Tay SK, Tay KJ, 2004, Passive cigarette smoking is a risk factor in cervical neoplasia. Gynecol Oncol 93:116–120 Tsai HT, Tsai YM, Yang SF, Wu KY, Chuang HY, Wu TN et al, 2007, Lifetime cigarette smoke and second-hand smoke and cervical intraepithelial neoplasm—a community-based case-control study. Gynecol Oncol 105:181–188 Natphopsuk S, Settheetham-Ishida W, Sinawat S, Pientong C, Yuenyao P, Ishida T, 2012, Risk factors for cervical cancer in northeastern Thailand: detailed analyses of sexual and smoking behavior. Asian Pac J Cancer Prev 13:5489–5495 Alam S, Conway MJ, Chen HS, Meyers C, 2008, The cigarette smoke carcinogen benzo[a]pyrene enhances human papillomavirus synthesis. J Virol 82:1053–1058 Stampfli MR, Anderson GP, 2009, How cigarette smoke skews immune responses to promote infection, lung disease and cancer. Nat Rev Immunol 9:377–384 Sundberg K, Johansson AS, Stenberg G, Widersten M, Seidel A, Mannervik B et al, 1998, Differences in the catalytic efficiencies of allelic variants of glutathione transferase P1-1 towards carcinogenic diol epoxides of polycyclic aromatic hydrocarbons. Carcinogenesis 19:433–436 Jee SH, Lee JE, Kim S, Kim JH, UmSJ, Lee SJ et al, 2002, GSTP1 polymorphism, cigarette smoking and cervical cancer risk in Korean women. Yonsei Med J 43:712–716 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 217 EP 222 DI 10.1007/s12253-018-0442-4 PG 6 ER PT J AU Vuletic, A Jovanic, I Jurisic, V Milovanovic, Z Nikolic, S Spurnic, I Konjevic, G AF Vuletic, Ana Jovanic, Irena Jurisic, Vladimir Milovanovic, Zorka Nikolic, Srdan Spurnic, Igor Konjevic, Gordana TI IL-2 And IL-15 Induced NKG2D, CD158a and CD158b Expression on T, NKT- like and NK Cell Lymphocyte Subsets from Regional Lymph Nodes of Melanoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Regional lymph nodes; IL-2; IL-15; T cells; NKT-like cells; NK cells ID Regional lymph nodes; IL-2; IL-15; T cells; NKT-like cells; NK cells AB Regional lymph nodes (LN)s represent important immunological barriers in spreading of malignant tumors. However, they are the most frequent early metastatic site in melanoma. Immunomodulatory agents including cytokines have been included in therapy of melanoma and have shown severe side effects and toxicity. In this sense, there is a growing need for bringing these agents to further in vitro testing that may enlighten aspects of their regional application. Therefore, the aim of this study was to investigate the effect of interleukin (IL)-2 and IL-15, the two cytokines with similar immune-enhancing effects, on the expression of activating NKG2D, inhibitory CD158a and CD158b receptors on CD8+ T, NKT-like and NK cell lymphocyte subsets from regional LNs of melanoma patients. In this study, we showed significant effects of IL-2 and IL-15 cytokine treatments on the expression of activating NKG2D and on inhibitory CD158a and CD158b receptors on lymphocytes, CD8+ T, NKT-like and NK cell lymphocyte subsets originating from regional LNs of melanoma patients. Furthermore, IL-2 and IL-15 by inducing the expression of NKG2D activating receptor on innate and on adaptive lymphocyte subsets and by augmenting NK cell antitumor cytotoxicity that correlated with the cytokine-induced NKG2D expression, increased antitumor potential of immune cells in regional LNs of melanoma patients irrespective of LN involvement. These findings indicate the importance of immune cell population from regional LNs of melanoma patients in the development of immune intervention strategies that may if applied locally increase antitumor potential to the level that controls tumor progressions. C1 [Vuletic, Ana] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Jovanic, Irena] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Jurisic, Vladimir] University of Kragujevac, Faculty of MedicineKragujevac, Serbia. [Milovanovic, Zorka] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Nikolic, Srdan] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Spurnic, Igor] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. [Konjevic, Gordana] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia. RP Vuletic, A (reprint author), Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia. EM radovanovica@ncrc.ac.rs CR Umansky V, Sevko A, 2012, Melanoma-induced immunosuppression and its neutralization. Semin Cancer Biol 22:319–326 Grotz TE, Mansfield AS, Jakub JW,Markovic SN, 2012, Regional lymphatic immunity in melanoma. Melanoma Res 22:9–18 Zheng R, Kjaergaard J, Lee WT, Cohen PA, Shu S, 2007, Significance of regional draining lymph nodes in the development of tumor immunity: implications for cancer immunotherapy. Cancer Treat Res 135:223–237 Pulliam SR, Uzhachenko RV, Adunyah SE, Shanker A, 2016, Common gamma chain cytokines in combinatorial immune strategies against cancer. Immunol Lett 169:61–72 Fewkes NM, Mackall CL, 2010, Novel gamma-chain cytokines as candidate immune modulators in immune therapies for cancer. Cancer J 16:392–398 Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM, Ugolini S, 2011, Innate or adaptive immunity? The example of natural killer cells. Science 331:44–49 Zafirova B,Wensveen FM, Gulin M, Polic B, 2011, Regulation of immune cell function and differentiation by the NKG2D receptor. Cell Mol Life Sci 68:3519–3529 Snyder MR, Weyand CM, Goronzy JJ, 2004, The double life of NK receptors: stimulation or co-stimulation? Trends Immunol 25: 25–32 Vivier E, Anfossi N, 2004, Inhibitory NK-cell receptors on T cells: witness of the past, actors of the future. Nat Rev Immunol 4:190– 198 Patterson S, Chaidos A, Neville DC, Poggi A, Butters TD, Roberts IA, Karadimitris A, 2008, Human invariant NKT cells display alloreactivity instructed by invariant TCR-CD1d interaction and killer Ig receptors. J Immunol 181:3268–3276 Jackson A, Warner N, 1986, Preparation, staining and analysis by flow cytometry of peripheral blood leukocytes. In: Rose N, Friedman H, Fahey J, eds, Manual of Clin Lab immunology, 3rd edn. American Society for Microbiology,Washington, pp 226–235 Vuletic AM, Jovanic IP, Jurisic VB, Milovanovic ZM, Nikolic SS, Tanic NT, Konjevic GM, 2015, In-vitro activation of natural killer cells from regional lymph nodes of melanoma patients with interleukin-2 and interleukin-15. Melanoma Res 25:22–34 Cochran AJ, Huang RR, Lee J, Itakura E, Leong SP, Essner R, 2006, Tumour-induced immune modulation of sentinel lymph nodes. Nat Rev Immunol 6:659–670 Farzad Z, Cochran AJ, McBrideWH GJD, Wong V, Morton L, 1990, Lymphocyte subset alterations in nodes regional to human melanoma. Cancer Res 50:3585–35888 Vuletic A, Jovanic I, Jurisic V, Milovanovic Z, Nikolic S, Spurnic I, Konjevic G, 2015, Decreased interferon γ production in CD3+ and CD3- CD56+ lymphocyte subsets in metastatic regional lymph nodes of melanoma patients. Pathol Oncol Res 21:1109–1114 Balch CM, Soong SJ, Gershenwald JE, Thompson JF, Reintgen DS, Cascinelli N, Urist M, McMasters KM, Ross MI, Kirkwood JM, Atkins MB, Thompson JA, Coit DG, Byrd D, Desmond R, ZhangY, Liu PY, Lyman GH,MorabitoA(2001, Prognostic factors analysis of 17,600 melanoma patients: validation of the American joint committee on Cancer melanoma staging system. J Clin Oncol 19:3622–3634 Vuletic A, Jurisic V, Jovanic I, Milovanovic Z, Nikolic S, Konjevic G, 2013, Distribution of several activating and inhibitory receptors onCD3(−)CD56(+, NK cells in regional lymph nodes ofmelanoma patients. J Surg Res 183:860–868 Konjevic G, Mirjacic Martinovic K, Vuletic A, Jurisic V, Spuzic I, 2009, Distribution of several activating and inhibitory receptors on CD3-CD16+ NK cells and their correlationwithNKcell function in healthy individuals. J Membr Biol 230:113–123 Ali TH, Pisanti S, Ciaglia E, Mortarini R, Anichini A, Garofalo C, Tallerico R, Santinami M, Gulletta E, Ietto C, GalganiM, Matarese G, Bifulco M, Ferrone S, Colucci F, Moretta A, Karre K, Carbone E, 2014, Enrichment of CD56(dim)KIR + CD57 + highly cytotoxic NK cells in tumour-infiltrated lymph nodes of melanoma patients. Nat Commun 5:5639. , DOI 10.1038/ncomms6639 Keskin DB, Allan DS, Rybalov B, Andzelm MM, Stern JN, Kopcow HD, Koopman LA, Strominger JL, 2007, TGFbeta promotes conversion of CD16+ peripheral blood NK cells into CD16- NK cells with similarities to decidual NK cells. Proc Natl Acad Sci U S A 104:3378–3383 Fransen MF, Arens R, Melief CJ, 2013, Local targets for immune therapy to cancer: tumor draining lymph nodes and tumor microenvironment. Int J Cancer 132:1971–1976 Konjevic G, Vuletic A, Mirjacic Martinovic K, 2016, Natural killer cell receptors: alterations and therapeutic targeting in malignancies. Immunol Res 64:25–35 Zhang C, Zhang J, Niu J, Zhang J, Tian Z, 2008, Interleukin-15 improves cytotoxicity of natural killer cells via up-regulating NKG2D and cytotoxic effector molecule expression as well as STAT1 and ERK1/2 phosphorylation. Cytokine 42:128–136 Verhoeven DH, de Hooge AS, Mooiman EC, Santos SJ, ten Dam MM, Gelderblom H, Melief CJ, Hogendoorn PC, Egeler RM, van Tol MJ, Schilham MW, Lankester AC, 2008, NK cells recognize and lyse Ewing sarcoma cells through NKG2D and DNAM-1 receptor dependent pathways. Mol Immunol 45:3917–3925 Hromadnikova I, Li S, Kotlabova K, Dickinson AM, 2016, Influence of in vitro IL-2 or IL-15 alone or in combination with Hsp 70 derived 14-Mer peptide, TKD, on the expression ofNK cell Activatory and inhibitory receptors on peripheral blood T cells, B cells and NKT cells. PLoS One 11:e0151535 Correia MP, Costa AV, Uhrberg M, Cardoso EM, Arosa FA, 2011, IL-15 induces CD8+ T cells to acquire functional NK receptors capable of modulating cytotoxicity and cytokine secretion. Immunobiology 216:604–612 Decot V, Voillard L, Latger-Cannard V, Aissi-Rothe L, Perrier P, Stoltz JF, Bensoussan D, 2010, Natural-killer cell amplification for adoptive leukemia relapse immunotherapy: comparison of three cytokines, IL-2, IL-15, or IL-7 and impact on NKG2D, KIR2DL1, and KIR2DL2 expression. Exp Hematol 38:351–362 Romagnani C, Juelke K, Falco M,Morandi B, D'Agostino A, Costa R, Ratto G, Forte G, Carrega P, Lui G, Conte R, Strowig T,Moretta A, Munz C, Thiel A, Moretta L, Ferlazzo G, 2007, CD56brightCD16- killer Ig-like receptor- NK cells display longer telomeres and acquire features of CD56dim NK cells upon activation. J Immunol 178:4947–4955 Ferlazzo G, Thomas D, Lin SL, Goodman K, Morandi B, Muller WA, Moretta A, Munz C, 2004, The abundant NK cells in human secondary lymphoid tissues require activation to express killer cell Ig-like receptors and become cytolytic. J Immunol 172:1455–1462 Cichocki F, Hanson RJ, Lenvik T, Pitt M, McCullar V, Li H, Anderson SK, Miller JS, 2009, The transcription factor c-Myc enhances KIR gene transcription through direct binding to an upstream distal promoter element. Blood 113:3245–3253 Juelke K, Killig M, Thiel A, Dong J, Romagnani C, 2009, Education of hyporesponsive NK cells by cytokines. Eur J Immunol 39:2548–2555 Konjevic G, Jurisic V, Jovic V, Vuletic A, Mirjacic Martinovic K, Radenkovic S, Spuzic I, 2012, Investigation of NK cell function and their modulation in different malignancies. Immunol Res 52: 139–156 Konjevic G, Mirjacic Martinovic K, Vuletic A, Jovic V, Jurisic V, Babovic N, Spuzic I, 2007, Lowexpression ofCD161 andNKG2D activating NK receptor is associated with impaired NK cell cytotoxicity inmetastatic melanoma patients. Clin ExpMetastasis 24:1– 11 Seidel MG, Freissmuth M, Pehamberger H, Micksche M, 1998, Stimulation of natural killer activity in peripheral blood lymphocytes of healthy donors and melanoma patients in vitro: synergism between interleukin, IL)-12 and IL-15 or IL-12 and IL-2. Naunyn Schmiedeberg's Arch Pharmacol 358:382–389 Sabry M, LowdellMW(2013, Tumor-primed NK cells: waiting for the green light. Front Immunol 4:408 Konjevic G, Mirjacic Martinovic K, Vuletic A, Babovic N, 2010, In-vitro IL-2 or IFN-α- induced NKG2D and CD161 NK cell receptor expression indicates novel aspects of NK cell activation in metastatic melanoma patients. Melanoma Res 20:459–467 Topham NJ, Hewitt EW(2009, Natural killer cell cytotoxicity: how do they pull the trigger? Immunology 128:7–15 Guia S, Jaeger BN, Piatek S, Mailfert S, Trombik T, Fenis A, Chevrier N, Walzer T, Kerdiles YM, Marguet D, Vivier E, Ugolini S, 2011, Confinement of activating receptors at the plasma membrane controls natural killer cell tolerance. Sci Signal 4:ra21 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 223 EP 231 DI 10.1007/s12253-018-0444-2 PG 9 ER PT J AU Pako, J Bikov, A Barta, I Matsueda, H Puskas, R Galffy, G Kerpel-Fronius, A Antus, B Horvath, I AF Pako, Judit Bikov, Andras Barta, Imre Matsueda, Hideyo Puskas, Rita Galffy, Gabriella Kerpel-Fronius, Anna Antus, Balazs Horvath, Ildiko TI Assessment of the circulating klotho protein in lung cancer patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; Biomarkers; Tumor suppressor; Premature aging; Insuline-like growth factor 1 ID Lung cancer; Biomarkers; Tumor suppressor; Premature aging; Insuline-like growth factor 1 AB The anti-aging factor, klotho has been identified as a tumor suppressor in various human cancers, including lung cancer. In vitro studies provided evidence that klotho expression influences the characteristics of lung cancer cells, however, in vivo results are lacking. The aim of our study was to evaluate whether circulating klotho protein might serve as a potential biomarker of lung cancer. Blood samples were taken from 45 newly diagnosed lung cancer patients (31 NSCLC, 14 SCLC) and 43 control subjects. Plasma klotho concentration was measured using ELISA. No difference in plasma klotho values was detected between patients and control subjects (366.3 (257.9–486.8) vs. 383.5 (304.6–489.7) pg/ml respectively (median (IQR)); p > 0.05). Plasma klotho levels in patients with distant metastasis did not differ from less advanced stage disease (354.2 (306.9–433.3 vs. 328.5 (242.5–419.7) pg/ml, p > 0.05). In contrast, analyzed with one-way ANOVA, significant difference (p = 0.04) was found between the examined histological types of lung cancer: adenocarcinoma (353 (329.4–438.5) pg/ml), squamous cell carcinoma (308 (209.6–348.1) pg/ml) and small cell lung cancer (388.8 (289.9–495.4) pg/ml). However, Tukey’s post hoc test did not reveal significant difference between any pairs of histological groups. There was no difference between any histological subtype and health either. Our results suggest that circulating klotho protein cannot be considered as a biomarker for lung cancer. Further studies are warranted in order to examine the relationship between klotho expression in lung tissue and circulating levels of the protein, and to explore its mechanism of action in lung cancer. C1 [Pako, Judit] National Koranyi Institute of Pulmonology, Piheno ut 1, 1121 Budapest, Hungary. [Bikov, Andras] Semmelweis University, Department of Pulmonology, Dios arok utca 1/c, 1125 Budapest, Hungary. [Barta, Imre] National Koranyi Institute of Pulmonology, Piheno ut 1, 1121 Budapest, Hungary. [Matsueda, Hideyo] Saitama Medical University Medical Center, 1981 KamodaKawagoe-shi, Saitama, Japan. [Puskas, Rita] Semmelweis University, Department of Pulmonology, Dios arok utca 1/c, 1125 Budapest, Hungary. [Galffy, Gabriella] Semmelweis University, Department of Pulmonology, Dios arok utca 1/c, 1125 Budapest, Hungary. [Kerpel-Fronius, Anna] National Koranyi Institute of Pulmonology, Piheno ut 1, 1121 Budapest, Hungary. [Antus, Balazs] National Koranyi Institute of Pulmonology, Piheno ut 1, 1121 Budapest, Hungary. [Horvath, Ildiko] National Koranyi Institute of Pulmonology, Piheno ut 1, 1121 Budapest, Hungary. RP Pako, J (reprint author), National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary. EM tidujok@gmail.com CR Chang B, Kim J, Jeong D, Jeong Y, Jeon S, Jung SI et al, 2012, Klotho inhibits the capacity of cell migration and invasion in cervical cancer. Oncol Rep 28:1022–1028 Yan Y, Wang Y, Xiong Y, Lin X, Zhou P, Chen Z, 2017, Reduced Klotho expression contributes to poor survival rates in human patients with ovarian cancer, and overexpression of Klotho inhibits the progression of ovarian cancer partly via the inhibition of systemic inflammation nude mice. Mol Med Rep 15:1777–1785 Wolf I, Levanon-Cohen S, Bose S, Ligumsky H, Sredni B, Kanety H et al, 2008, Klotho: a tumor suppressor and a modulator of the IGF-1 and FGF pathways in human breast cancer. Oncogene 27: 7094–7105 Rubinek T, Wolf I, 2016, The role of alpha-klotho as a universal tumor supressor. Vitam Horm 101:197–214 Zhou X, Wang X, 2015, Klotho: a novel biomarker for cancer. J Cancer Res Clin Oncol 141:961–969 Kuro-o M, 2010, Klotho. Eur J Physiol 459:333–343 Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P et al, 2005, Suppression of aging in mice by the Hormone Klotho. Science 5742:1829–1833 Liu H, Fergusson MM, Castilho RM, Liu J, Cao L, Chen J et al, 2007, Augmented Wnt signaling in a mammalian model of accelerated aging. Science 317:803–806 Velcheti V, Ramaswamy G, 2006, Insulin-like growth factor and lung cancer. J Thorac Oncol 1:607–610 He B, Barg RN,You L, Xu Z, Reguart N, Mikami I et al, 2005)Wnt signaling in stem cells and non-small-cell lung cancer. Clin Lung Cancer 7:54–60 FidlerMJ, Shersher DD, Borgia JA, Bonomi P, 2012, Targeting the insulin-like growth factor receptor pathway in lung cancer: problems and pitfalls. Ther Adv Med Oncol 4:51–60 Yang J, Chen J,He J, Li J, Shi J, ChoWCet al, 2016)Wnt signaling as potential therapeutic target in lung cancer. Expert Opin Ther Targets 20:999–1015 Chen B, Ma X, Liu S, Zhao W, Wu J, 2012, Inhibition of lung cancer cells growth, motility and induction of apoptosis by Klotho, a novel secreted Wnt antagonist in a dose-dependent manner. Cancer Biol Ther 13:1221–1228 Usuda J, Ichinose S, Ishizumi T, Ohtani K, Inoue T, Saji H et al, 2011, Klotho predicts good clinical outcome in patients with limited-disease small cell lung cancer who received surgery. Lung Cancer 74:332–337 Usuda J, Ichinose S, Ishizumi T, Ohtani K, Inoue T, Saji H et al, 2011, Klotho is a novel biomarker for good survival in resected large cell neuroendocrine carcinoma of the lung. Lung Cancer 72:355–359 Chen B,Wang X, ZhaoW,Wu J, 2010, Klotho inhibits growth and promotes apoptosis in human lung cancer cell line A549. J Exp Clin Cancer Res 29:99 Chen T, Ren H, Thakur A, Yang T, Li Y, Zhang S et al, 2016, Decreased level of klotho contributes to drug resistance in lung cancer cells: involving in klotho-mediated cell autophagy. DNA Cell Biol 35:751–757 Wang Y, Chen L, Huang G, He D, He J, Xu Wet al, 2013, Klotho sensitizes human lung cancer cell line to cisplatin via PI3k/Akt pathway. PLoS One 8:e57391 Tang X, Fan Z, Huang S, 2013, Can soluble klotho protein be a potential tumor biomarker? Am J Clin Pathol 139:259–262 Gigante M, Lucarelli G, Divella C, Netti GS, Pontrelli P, Cafiero C et al, 2015, Soluble serum αklotho is a potential predictive marker of disease progression in clear cell renal cell carcinoma. Medicine 94:e1917 Pedersen L, Pedersen SM, Brasen CL, Rasmussen LM, 2013, Soluble serum klotho levels in healthy subjects. Comparison of two different immunoassays. Clin Biochem 46:1079–1083 Pako J, Barta I, Balogh Z, Kerti M, Drozdovszky O, Bikov A et al, 2017, Assessment of the anti-aging klotho protein in patients with COPD undergoing pulmonary rehabilitation. COPD 14:176–180 Patel MS, Donaldson AV, Lewis A, Natanek SA, Lee JY, Andersson YM et al, 2016, Klotho and smoking – An interplay influencing the skeletal muscle functions deficits that occur in COPD. Respir Med 113:50–56 Yamazaki Y, Imura A, Urakawa I, Shimada T, Murakami J, Aono Y et al, 2010, Establishment of sandwich ELISA for soluble alphaklotho measurement: Age-dependent change of soluble alphaklotho levels in healthy subjects. Biochem Biophys Res Commun 398:513–518 Rubinek T, Shulman M, Israeli S, Bose S, Avraham A, Zundelevich A et al, 2012, Epigenetic silencing of the tumor suppressor klotho in human breast cancer. Breast Cancer Res Treat 133:649–657 Xie B, Zhou J, Yuan L, Ren F, Liu DC, Li Q et al, 2013, Epigenetic silencing of klotho expression correlates with poor prognosis of human hepatocellular carcinoma. Hum Pathol 44:795–801 Usugi T, Ohno T, Ohyama Y, Uchiyama T, Saito Y,Matsumura Yet al, 2000, Decreased insulin production and increased insulin sensitivity in the klotho mutant mouse, a novel animal model for human aging. Metabolism 49:1118–1123 Neidert MC, Sze L, Zwimpfer C, Sarnthein J, Seifert B, Frei K et al, 2013, Soluble α-klotho: a novel serum biomarker for the activity of GH-producing pituitary adenomas. Eur J Endocrinol 168:575–583 Sze L, Bernays RL, Zwimpfer C, Wiesli P, Brandle M, Schmid C, 2012, Excessively high soluble Klotho in patients with acromegaly. J Intern Med 272:93–97 Kohler S, Tschopp O, Sze L, Neidert M, Bernays RL, Spanaus KS et al, 2013, Monitoring for potential residual disease activity by serum insulin-like growth factor 1 and soluble Klotho in patients with acromegaly after pituitary surgery. Gen Comp Endocrinol 188:282–287 Yu H, Spitz MR, Mistry J, Gu J, Hong WK, Wu X, 1999, Plasma levels of insulin-like growth factor-1 and lung cancer risk: a casecontrol analysis. J Natl Cancer Inst 91:151–156 Spitz MR, Barnett MJ, Goodman GE, Thornquist MD, Wu X, Pollak M, 2002, Serum insulin-like growth factor, IGF, and IGFbinding protein levels and risk of lung cancer: a case-control study nested in the beta-carotene and retinol efficacy trial cohort. Cancer Epidemiol Biomarkers Prev 11:1413–1418 Zhang M, Li X, Zhang X, Yang Y, Feng Z, Liu X, 2014, Association of serum hemoglobin A1c, C-peptide and insulin-like growth factor-1 levelswith the occurrence and development of lung cancer. Mol Clin Oncol 4:506–508 Tas F, Bilgin E, Tastekin D, Erturk K, Duranyildiz D, 2016, Serum IGF-1 and IGFBP-3 levels as clinicalmarkers for patients with lung cancer. Biomed Rep 4:609–614 Minuto F, Del Monte P, Barreca A, Alama A, Cariola G, Giordano G, 1988, Evidence for autocrine mitogenic stimulation by somatomedin- C/insuline-like growth factor I on an established human lung cancer cell line. Cancer Res 48:3716–3719 Toshifumi S, TakeshitaY,Murohara T, SasakiK, Egami K, Shintani S et al, 2004, Angiogenesis and vasculogenesis are impaired in the precocious-aging klotho mouse. Circulation 110:1148–1155 Fukino K, Suzuki T, Saito Y, Shindo T, Amaki T, Kurabayashi M et al, 2002, Regulation of angiogenesis by the aging suppressor gene klotho. Biochem Biophys Res Commun 293:332–337 Pan JY, Sun CC, Li SJ, Huang J, Li DJ, 2015, Role of miR-10b in non-small cell lung cancer, NSCLC, cells by targeting klotho. Cancer Cell Microenviron 2:e936 Qi HW, Shen Z, Fan LH, 2011, Combined inhibition of insulin-like growth factor-1 receptor enhances the effects of gefitinib in a human non-small cell lung cancer resistant cell line. Exp Ther Med 2: 1091e1095 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 233 EP 238 DI 10.1007/s12253-018-0441-5 PG 6 ER PT J AU Prieto-Granada, C Castner, N Chen, A Li, J Yue, B Wong, YJ Iyengar, S Sondak, KV Zager, SJ Messina, LJ AF Prieto-Granada, Carlos Castner, Nicholas Chen, Ann Li, Jiannong Yue, Binglin Wong, Y Joyce Iyengar, Sanjana Sondak, K Vernon Zager, S Jonathan Messina, L Jane TI Behavior of Cutaneous Adnexal Malignancies: a Single Institution Experience SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cutaneous adnexal neoplasms; Eccrine carcinoma; Sentinel lymph node biopsy ID Cutaneous adnexal neoplasms; Eccrine carcinoma; Sentinel lymph node biopsy AB Cutaneous adnexal malignancies are biologically and pathologically diverse, and associated with a range of clinical outcomes. Given their rarity, the prognosis and optimal treatment of these neoplasms remains unclear. A single institution database from a tertiary care cancer center of patients treated for malignant cutaneous adnexal tumors was retrospectively analyzed. Clinicopathologic variables and outcome measures were analyzed in patients undergoing wide excision with or without sentinel node biopsy. 103 patients were analyzed; the majority of tumors were of eccrine sweat gland derivation (n = 69, 70%), and these exhibited a higher rate of nodal involvement and overall worse outcome. Sixteen patients (16%) demonstrated nodal metastasis, which included 10 (10%) with nodal disease at presentation and 6 who developed nodal metastasis during follow-up. 20 patients underwent sentinel node biopsy, and 2 (10%) had a positive sentinel node. 62% of nodal metastases occurred in patients with porocarcinoma. Seven patients died of disease (7%) with a median time from diagnosis to death of 48 months (range, 10–174). After a median follow up of 44.7 months, age > 70 years and larger tumor size were significantly associated with worse overall survival. Adnexal malignancies are rare tumors, and there is a paucity of information to guide the clinician in determining optimum surgical and medical treatment. Tumors of eccrine derivation, especially porocarcinomas, have a high risk of nodal involvement and may be considered for sentinel node biopsy. C1 [Prieto-Granada, Carlos] Moffitt Cancer Center, Department of Anatomic Pathology, 12902 Magnolia Drive, 33612 Tampa, FL, USA. [Castner, Nicholas] Moffitt Cancer Center, Department of Cutaneous OncologyTampa, FL, USA. [Chen, Ann] Moffitt Cancer Center, Department of BiostatisticsTampa, FL, USA. [Li, Jiannong] Moffitt Cancer Center, Department of BiostatisticsTampa, FL, USA. [Yue, Binglin] Moffitt Cancer Center, Department of BiostatisticsTampa, FL, USA. [Wong, Y Joyce] Moffitt Cancer Center, Department of Anatomic Pathology, 12902 Magnolia Drive, 33612 Tampa, FL, USA. [Iyengar, Sanjana] Moffitt Cancer Center, Department of Cutaneous OncologyTampa, FL, USA. [Sondak, K Vernon] Moffitt Cancer Center, Department of Cutaneous OncologyTampa, FL, USA. [Zager, S Jonathan] Moffitt Cancer Center, Department of Cutaneous OncologyTampa, FL, USA. [Messina, L Jane] Moffitt Cancer Center, Department of Anatomic Pathology, 12902 Magnolia Drive, 33612 Tampa, FL, USA. RP Messina, LJ (reprint author), Moffitt Cancer Center, Department of Anatomic Pathology, 33612 Tampa, USA. EM jane.messina@moffitt.org CR Edge SB, 2010, American Joint Committee on Cancer. In: AJCC cancer staging manual, 7th edn. Springer, New York Rogers HW, Weinstock MA, Harris AR et al, 2010, Incidence estimate of nonmelanoma skin cancer in the United States, 2006. Arch Dermatol 146(3):283–287 Blake PW, Bradford PT, Devesa SS, Toro JR, 2010, Cutaneous appendageal carcinoma incidence and survival patterns in the United States: a population-based study. Arch Dermatol 146(6): 625–632 Martinez SR, Barr KL, Canter RJ, 2011, Rare tumors through the looking glass: an examination of malignant cutaneous adnexal tumors. Arch Dermatol 147(9):1058–1062 Mallone S, De Vries E, GuzzoM et al, 2012, Descriptive epidemiology ofmalignantmucosal and uveal melanomas and adnexal skin carcinomas in Europe. Eu J Ca 48(8):1167–1175 Stam H, Lohuis PJ, Zupan-Kajcovski B, Wouters MW, van der Hage JA, Visser O, 2013, Increasing incidence and survival of a rare skin cancer in the Netherlands. A population-based study of 2,220 cases of skin adnexal carcinoma. J Surg Oncol 107(8): 822–827 AmbeCMSV, 2014, Sentinel lymph node biopsy in melanoma and other cutaneous malignancies. Am J Hematol Oncol 10(3):3–9 Bogner PN, Fullen DR, Lowe L et al, 2003, Lymphatic mapping and sentinel lymph node biopsy in the detection of early metastasis from sweat gland carcinoma. Cancer 97(9):2285– 2289 Delgado R, Kraus D, Coit DG, Busam KJ, 2003, Sentinel lymph node analysis in patients with sweat gland carcinoma. Cancer 97(9): 2279–2284 Breslow A, 1970, Thickness, cross-sectional areas and depth of invasion in the prognosis of cutaneous melanoma. Ann Surg 172(5):902–908 Wright JR, Jr. Albert C., 2012, Broders' paradigm shifts involving the prognostication and definition of cancer. Arch Pathol Lab Med 136(11):1437–1446 Morton DL, Wen DR, Wong JH et al, 1992, Technical details of intraoperative lymphatic mapping for early stage melanoma. Arch Surg, Chicago, Ill: 1960, 127(4):392–399 Ross MI, Reintgen D, Balch CM, 1993, Selective lymphadenectomy: emerging role for lymphatic mapping and sentinel node biopsy in the management of early stage melanoma. Semin Surg Oncol 9(3):219–223 Ponti G, Losi L, Di Gregorio C et al, 2005, Identification of Muir- Torre syndrome among patients with sebaceous tumors and keratoacanthomas: role of clinical features, microsatellite instability, and immunohistochemistry. Cancer 103(5):1018–1025 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 239 EP 244 DI 10.1007/s12253-018-0427-3 PG 6 ER PT J AU Zhang, Y Ding, Sh Yang, J Chen, X Huang, W AF Zhang, Yanping Ding, Shi Yang, Jie Chen, Xingfeng Huang, Weilin TI Identification of miR-146a is Associated with the Aggressiveness and Suppresses Proliferation via Targeting CDKN2A in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE miR-146a; Biomarker; Breast cancer ID miR-146a; Biomarker; Breast cancer AB There is emerging evidence that some microRNAs can promote or suppress several human cancer development and progression. However, the profile and molecular mechanism of microRNAs for human breast cancer is poorly unknown. We used bioinformatics approaches to find new candidate diagnostic and therapeutic miRNAs in human breast cancer via analysis of TCGA RNA sequencing data and publicly GEO microarray data, in order to provide theoretical basis for the future investigations of breast cancer. Decreased expression miR-146a was identified as a key regulator of human breast cancer development and progression. Interestingly, we founded that miR-146a expression levels dependent on tumor size and pathological grading in breast cancer patients, but not associated with other factors including age, T-classification. Kaplan-Meier survival analysis showed that patients with high miR-146a expression had a longer survival rate than those low miR-146a expressions. In vitro assays of overexpression miR-146a induces cell cycle arrest and inhibits MDA-MB-231 cell proliferation. Furthermore, luciferase reporter gene assays demonstrated that miR-146a directly combine the 3-untranslated region of CDKN2A mRNA. In conclusion, we demonstrated miR-146a play an important role in breast cancer development and progression. C1 [Zhang, Yanping] Changde Vocational and Technical College, Department of Clinical Medicine, #4253 Renmin Road, Wuling District, 415000 Changde City, Hunan Province, China. [Ding, Shi] Changde Vocational and Technical College, Department of Clinical Medicine, #4253 Renmin Road, Wuling District, 415000 Changde City, Hunan Province, China. [Yang, Jie] Changde Vocational and Technical College, Department of Clinical Medicine, #4253 Renmin Road, Wuling District, 415000 Changde City, Hunan Province, China. [Chen, Xingfeng] Changde Vocational and Technical College, Department of Clinical Medicine, #4253 Renmin Road, Wuling District, 415000 Changde City, Hunan Province, China. [Huang, Weilin] Changde Vocational and Technical College, Department of Clinical Medicine, #4253 Renmin Road, Wuling District, 415000 Changde City, Hunan Province, China. RP Zhang, Y (reprint author), Changde Vocational and Technical College, Department of Clinical Medicine, 415000 Changde City, China. EM ypzhcd@126.com CR Barton MK, 2016, Use of posttreatment imaging and biomarker testing for survivors of breast cancer. CA Cancer J Clin 66(3): 175–176 Ward EM, DeSantis CE, Lin CC, Kramer JL, Jemal A, Kohler B, Brawley OW, Gansler T, 2015, Cancer statistics: Breast cancer in situ. CA Cancer J Clin 65(6):481–495 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87–108 Gotzsche PC, Jorgensen KJ, 2013, Screening for breast cancer with mammography. Cochrane Database Syst Rev 6:CD001877 Siu AL, 2016, Force USPST: screening for breast cancer: U.S. preventive services task force recommendation statement. Ann Intern Med 164(4):279–296 Lacroix M, 2006, Significance, detection and markers of disseminated breast cancer cells. Endocr Relat Cancer 13(4):1033–1067 Hayes J, Richardson A, Frampton C, 2013, Population attributable risks for modifiable lifestyle factors and breast cancer in New Zealand women. Intern Med J 43(11):1198–1204 Collaborative Group on Hormonal Factors in Breast C, 2002, Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet 360(9328):187–195 Yager JD, Davidson NE, 2006, Estrogen carcinogenesis in breast cancer. N Engl J Med 354(3):270–282 Lee IM, Shiroma EJ, Lobelo F, Puska P, Blair SN, Katzmarzyk PT, 2012, Lancet physical activity series working G: effect of physical inactivity on major non-communicable diseases worldwide: an analysis of burden of disease and life expectancy. Lancet 380(9838):219–229 Casey PM, Cerhan JR, Pruthi S, 2008, Oral contraceptive use and risk of breast cancer. Mayo Clin Proc 83(1):86–90 quiz 90-81 Bartel DP, 2009, MicroRNAs: target recognition and regulatory functions. Cell 136(2):215–233 Fabian MR, Sonenberg N, Filipowicz W, 2010, Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem 79:351–379 Lewis BP, Burge CB, Bartel DP, 2005, Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120(1):15–20 Valdmanis PN, Gu S, Chu K, Jin L, Zhang F, Munding EM, Zhang Y, Huang Y, Kutay H, Ghoshal K et al, 2016, RNA interferenceinduced hepatotoxicity results from loss of the first synthesized isoform of microRNA-122 in mice. Nat Med 22(5):557–562 Wu H, Mo YY, 2009, Targeting miR-205 in breast cancer. Expert Opin Ther Targets 13(12):1439–1448 Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ, 2008, The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol 10(5):593–601 Wen SY, Lin Y, Yu YQ, Cao SJ, Zhang R, Yang XM, Li J, Zhang YL, Wang YH, Ma MZ et al, 2015, miR-506 acts as a tumor suppressor by directly targeting the hedgehog pathway transcription factor Gli3 in human cervical cancer. Oncogene 34(6):717–725 Vosa U, Vooder T, Kolde R, Fischer K, Valk K, Tonisson N, Roosipuu R, Vilo J, Metspalu A, Annilo T, 2011, Identification of miR-374a as a prognostic marker for survival in patients with early-stage nonsmall cell lung cancer. Genes Chromosom Cancer 50(10):812–822 Akcakaya P, Ekelund S, Kolosenko I, Caramuta S, Ozata DM, Xie H, Lindforss U, Olivecrona H, Lui WO, 2011, miR-185 and miR- 133b deregulation is associated with overall survival and metastasis in colorectal cancer. Int J Oncol 39(2):311–318 Mavrakis KJ,McDonald ER 3rd, Schlabach MR, Billy E, Hoffman GR, deWeck A, Ruddy DA, Venkatesan K, Yu J,McAllister G et al, 2016, Disordered methionine metabolism in MTAP/CDKN2Adeleted cancers leads to dependence on PRMT5. Science 351(6278):1208–1213 Boughton B, 2015, BRAF and CDKN2A mutations in secondary high-grade glioma. Lancet Oncol 16(5):e201 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 245 EP 251 DI 10.1007/s12253-018-0430-8 PG 7 ER PT J AU Krafft, U Reis, H Ingenwerth, M Kovalszky, I Becker, M Niedworok, Ch Darr, Ch Nyirady, P Hadaschik, B Szarvas, T AF Krafft, Ulrich Reis, Henning Ingenwerth, Marc Kovalszky, Ilona Becker, Markus Niedworok, Christian Darr, Christopher Nyirady, Peter Hadaschik, Boris Szarvas, Tibor TI Nuclear Localization of Robo is Associated with Better Survival in Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder Cancer; Robo1; Robo4; Nuclear translocation; Slit2; Prognosis; Angiogenesis ID Bladder Cancer; Robo1; Robo4; Nuclear translocation; Slit2; Prognosis; Angiogenesis AB The Slit-Robo pathway has shown to be altered in several malignant diseases. However, its role in bladder cancer is poorly understood. Therefore, we aimed to assess the tissue expression of Robo1 and Robo4 as well as their ligand Slit2 in different stages of bladder cancer to explore possible changes of Slit-Robo signaling during the progression of bladder cancer. Robo1, Robo4 and Slit2 gene expressions were analyzed in 92 frozen bladder cancer tissue samples by using reverse transcription quantitative real-time PCR. Immunohistochemical analyses were performed on 149 formalin-fixed and paraffin-embedded bladder cancer tissue samples. Results were correlated with the clinical and follow-up data by performing both univariable and multivariable analyses. Robo1 and Robo4 nuclear staining intensity was significantly higher in low stage and low grade bladder cancer. Elevated Robo1 nuclear staining was associated with better disease-specific survival (DSS) (p = 0.045). Similarly, stronger Robo4 nuclear staining tended to be associated with longer DSS (p = 0.061). We found higher Robo1 and Slit2 gene expression levels in advanced stages of bladder cancer (p = 0.007 and p < 0.001). High Slit2 gene expression was correlated with significantly shorter DSS (p < 0.005), while Robo1 and Robo4 gene expressions were not associated with patients’ prognosis. Our results demonstrate that the nuclear expression of Robo1 and Robo4 is associated with a favourable prognosis suggesting that its translocation into the nucleus represent a posttranslational regulation process which may exhibit an antitumor effect in bladder cancer. C1 [Krafft, Ulrich] University of Duisburg-Essen, Department of Urology, Hufelandstr 55, 45147 Essen, Germany. [Reis, Henning] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Ingenwerth, Marc] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Becker, Markus] University of Duisburg-Essen, Department of Urology, Hufelandstr 55, 45147 Essen, Germany. [Niedworok, Christian] University of Duisburg-Essen, Department of Urology, Hufelandstr 55, 45147 Essen, Germany. [Darr, Christopher] University of Duisburg-Essen, Department of Urology, Hufelandstr 55, 45147 Essen, Germany. [Nyirady, Peter] Semmelweis University, Department of Urology, Ulloi ut 78/b, 1082 Budapest, Hungary. [Hadaschik, Boris] University of Duisburg-Essen, Department of Urology, Hufelandstr 55, 45147 Essen, Germany. [Szarvas, Tibor] University of Duisburg-Essen, Department of Urology, Hufelandstr 55, 45147 Essen, Germany. RP Szarvas, T (reprint author), University of Duisburg-Essen, Department of Urology, 45147 Essen, Germany. EM sztibusz@gmail.com CR Stein JP, Skinner DG, 2006, Radical cystectomy for invasive bladder cancer: long-term results of a standard procedure.World J Urol 24(3):296–304. , DOI 10.1007/s00345-006-0061-7 Robertson AG, Kim J, Al-Ahmadie H, Bellmunt J, Guo G, Cherniack AD, Hinoue T, Laird PW, Hoadley KA, Akbani R, Castro MAA, Gibb EA, Kanchi RS, Gordenin DA, Shukla SA, Sanchez-Vega F, Hansel DE, Czerniak BA, Reuter VE, Su X, de Sa Carvalho B, Chagas VS, Mungall KL, Sadeghi S, Pedamallu CS, Lu Y, Klimczak LJ, Zhang J, Choo C, Ojesina AI, Bullman S, Leraas KM, LichtenbergTM,Wu CJ, Schultz N, Getz G, Meyerson M, Mills GB, McConkey DJ, Weinstein JN, Kwiatkowski DJ, Lerner SP, 2017, Comprehensive molecular characterization of muscle-invasive bladder Cancer. Cell 171(3):540–556.e525. , DOI 10.1016/j.cell.2017.09.007 Wang B, Xiao Y, Ding BB, Zhang N, Yuan X, Gui L, Qian KX, Duan S, Chen Z, Rao Y, Geng JG, 2003, Induction of tumor angiogenesis by Slit-Robo signaling and inhibition of cancer growth by blocking Robo activity. Cancer Cell 4(1):19–29 Dickinson RE, Dallol A, Bieche I, Krex D, Morton D, Maher ER, Latif F, 2004, Epigenetic inactivation of SLIT3 and SLIT1 genes in human cancers. Br J Cancer 91(12):2071–2078. , DOI 10. 1038/sj.bjc.6602222 Dickinson RE, Duncan WC, 2010, The SLIT-ROBO pathway: a regulator of cell function with implications for the reproductive system. Reproduction, Cambridge, England, 139(4):697–704. , DOI 10.1530/rep-10-0017 Kaur S, Castellone MD, Bedell VM, Konar M, Gutkind JS, Ramchandran R, 2006, Robo4 signaling in endothelial cells implies attraction guidance mechanisms. J Biol Chem 281(16): 11347–11356. , DOI 10.1074/jbc.M508853200 Sundaresan V, Heppell-Parton A, Coleman N,Miozzo M, Sozzi G, Ball R, Cary N, Hasleton P, Fowler W, Rabbitts P, 1995, Somatic genetic changes in lung cancer and precancerous lesions. Ann Oncol 6(Suppl 1):27–31 discussion 31-22 Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK,Wilson PJ, PatchAM,Wu J, Chang DK, Cowley MJ, Gardiner BB, Song S, Harliwong I, Idrisoglu S, Nourse C, Nourbakhsh E, Manning S, Wani S, Gongora M, Pajic M, Scarlett CJ, Gill AJ, Pinho AV, Rooman I, Anderson M, Holmes O, Leonard C, Taylor D, Wood S, Xu Q, Nones K, Fink JL, Christ A, Bruxner T, Cloonan N, Kolle G, Newell F, Pinese M, Mead RS, Humphris JL, Kaplan W, Jones MD, Colvin EK, Nagrial AM, Humphrey ES, Chou A, Chin VT, Chantrill LA, Mawson A, Samra JS, Kench JG, Lovell JA, Daly RJ, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Kakkar N, Zhao F, Wu YQ,WangM,MuznyDM, FisherWE, Brunicardi FC, Hodges SE, Reid JG, Drummond J, Chang K, Han Y, Lewis LR, Dinh H, Buhay CJ, Beck T, Timms L, Sam M, Begley K, Brown A, Pai D, Panchal A, Buchner N, De Borja R, Denroche RE, Yung CK, Serra S, Onetto N, Mukhopadhyay D, Tsao MS, Shaw PA, Petersen GM, Gallinger S, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Schulick RD, Wolfgang CL, Morgan RA, Lawlor RT, Capelli P, CorboV, Scardoni M, Tortora G, TemperoMA,MannKM, Jenkins NA, Perez-Mancera PA, Adams DJ, Largaespada DA, Wessels LF, Rust AG, Stein LD, Tuveson DA, Copeland NG, Musgrove EA, Scarpa A, Eshleman JR, Hudson TJ, Sutherland RL, Wheeler DA, Pearson JV, McPherson JD, Gibbs RA, Grimmond SM, 2012, Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491(7424):399–405. , DOI 10. 1038/nature11547 Gara RK, Kumari S, Ganju A, YallapuMM, Jaggi M, Chauhan SC, 2015, Slit/Robo pathway: a promising therapeutic target for cancer. Drug Discov Today 20(1):156–164. , DOI 10.1016/j. drudis.2014.09.008 Prasad A, Fernandis AZ, Rao Y, Ganju RK, 2004, Slit proteinmediated inhibition of CXCR4-induced chemotactic and chemoinvasive signaling pathways in breast cancer cells. J Biol Chem 279(10):9115–9124. , DOI 10.1074/jbc. M308083200 Prasad A, Paruchuri V, Preet A, Latif F, Ganju RK, 2008, Slit-2 induces a tumor-suppressive effect by regulating beta-catenin in breast cancer cells. J Biol Chem 283(39):26624–26633. https:// doi.org/10.1074/jbc.M800679200 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), method. Methods, San Diego, Calif, 25(4):402–408. https:// doi.org/10.1006/meth.2001.1262 Huang T, Kang W, Cheng AS, Yu J, To KF, 2015, The emerging role of Slit-Robo pathway in gastric and other gastro intestinal cancers. BMC Cancer 15:950. , DOI 10.1186/ s12885-015-1984-4 Legg JA, Herbert JM, Clissold P, Bicknell R, 2008, Slits and roundabouts in cancer, tumour angiogenesis and endothelial cell migration. Angiogenesis 11(1):13–21. , DOI 10. 1007/s10456-008-9100-x ZhouWJ, Geng ZH, Chi S, ZhangW, Niu XF, Lan SJ,Ma L, Yang X, Wang LJ, Ding YQ, Geng JG, 2011, Slit-Robo signaling induces malignant transformation through Hakai-mediated Ecadherin degradation during colorectal epithelial cell carcinogenesis. Cell Res 21(4):609–626. , DOI 10.1038/cr.2011.17 Stella MC, Trusolino L, Comoglio PM, 2009, The Slit/Robo system suppresses hepatocyte growth factor-dependent invasion and morphogenesis. Mol Biol Cell 20(2):642–657. , DOI 10. 1091/mbc.E08-03-0321 Marlow R, Strickland P, Lee JS,Wu X, PebenitoM, Binnewies M, Le EK, Moran A, Macias H, Cardiff RD, Sukumar S, Hinck L, 2008, SLITs suppress tumor growth in vivo by silencing Sdf1/ Cxcr4 within breast epithelium. Cancer Res 68(19):7819–7827. , DOI 10.1158/0008-5472.can-08-1357 Schmid BC, Rezniczek GA, Fabjani G, Yoneda T, Leodolter S, Zeillinger R, 2007, The neuronal guidance cue Slit2 induces targeted migration and may play a role in brain metastasis of breast cancer cells. Breast Cancer Res Treat 106(3):333–342. https://doi. org/10.1007/s10549-007-9504-0 Li Y, Cheng H, Xu W, Tian X, Li X, Zhu C, 2015, Expression of Robo protein in bladder cancer tissues and its effect on the growth of cancer cells by blocking Robo protein. Int J Clin Exp Pathol 8(9): 9932–9940 Fujiwara M, Ghazizadeh M, Kawanami O, 2006, Potential role of the Slit/Robo signal pathway in angiogenesis. Vasc Med, London, England, 11(2):115–121 Seki M, Watanabe A, Enomoto S, Kawamura T, Ito H, Kodama T, Hamakubo T, Aburatani H, 2010, Human ROBO1 is cleaved by metalloproteinases and gamma-secretase and migrates to the nucleus in cancer cells. FEBS Lett 584(13):2909–2915. , DOI 10.1016/j.febslet.2010.05.009 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 253 EP 261 DI 10.1007/s12253-018-0447-z PG 9 ER PT J AU Sato, K Miyamoto, M Takano, M Furuya, K Tsuda, H AF Sato, Kimiya Miyamoto, Morikazu Takano, Masashi Furuya, Kenichi Tsuda, Hitoshi TI Different Prognostic Implications of Aquaporin-1 and Aquaporin-5 Expression among Different Histological Types of Ovarian Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Aquaporin; Water channel; Ovarian carcinoma; Immunohistochemistry; Tissue microarray ID Aquaporin; Water channel; Ovarian carcinoma; Immunohistochemistry; Tissue microarray AB The aquaporins (AQPs) are a family of transmembrane water channel proteins that are distributed in various human tissues. Recent studies have suggested that AQP expression correlates with various aspects of cancer biology that determine the aggressiveness of different cancers. Ovarian carcinoma is one of the most lethal gynecological cancers. Some studies have suggested that AQPs are expressed in ovarian carcinoma, and are associated with cancer cell growth and migration. In this study, we immunohistochemically evaluated the expression of AQP1, 3, 5, and 9 in a total of 300 ovarian carcinomas using tissue microarrays. In our analyses of correlations between aquaporin expression and overall survival, high AQP5 expression was significantly associated with poorer prognosis (P = 0.029). For AQP1, the low expression group trended towards poorer prognosis than the high expression group, but the difference was not statistically significant. When ovarian carcinomas were divided by histological types, high AQP5 expression correlated with poorer prognosis in serous carcinoma (P = 0.015), and low AQP1 expression correlated with poorer prognosis in clear cell carcinomas (P = 0.0055). By contrast, high AQP1 expression correlated with poorer prognosis in mucinous carcinoma (P = 0.0001) and endometrioid carcinoma (P = 0.021). Our studies suggest that AQPs can be useful prognostic markers in ovarian carcinoma, but their correlation with prognosis depends on the histological type of ovarian carcinoma. C1 [Sato, Kimiya] National Defense Medical College, Department of Basic Pathology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Miyamoto, Morikazu] National Defense Medical College, Department of Obstetrics and GynecologyTokorozawa, Japan. [Takano, Masashi] National Defense Medical College, Department of Clinical OncologyTokorozawa, Japan. [Furuya, Kenichi] National Defense Medical College, Department of Obstetrics and GynecologyTokorozawa, Japan. [Tsuda, Hitoshi] National Defense Medical College, Department of Basic Pathology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. RP Sato, K (reprint author), National Defense Medical College, Department of Basic Pathology, 359-8513 Tokorozawa, Japan. EM kimiya@ndmc.ac.jp CR Verkman AS, 2005, More than just water channels: unexpected cellular roles of aquaporins. J Cell Sci 118:3225–3232 Agre P, King LS, YasuiM et al, 2002, Aquaporin water channels – from atomic structure to clinical medicine. J Physiol 542:3–16 Saadoun S, Papadopoulos MC, Hara-Chikuma M, Verkman AS, 2005, Impairment of angiogenesis and cell migration by targeted aquaporin-1 gene disruption. Nature 434:786–792 Papadopoulos MC, Saadoun S, Verkman AS, 2008, Aquaporins and cell migration. Pflugers Arch 456:693–700 Woo J, Lee J, Chae YK et al, 2008, Overexpression of AQP5, a putative oncogene, promotes cell growth and transformation. Cancer Lett 264:54–62 Verkman AS, Hara-Chikuma M, Papadopoulos MC, 2008, Aquaporins – new players in cancer biology. J Mol Med, Berl, 86:523–529 Hu J, Verkman AS, 2006, Increased migration and metastatic potential of tumor cells expressing aquaporin water channels. FASEB J 20:1892–1894 Wang J, Feng L, Zhu Z et al, 2015, Aquaporins as diagnostic and therapeutic targets in cancer: how far we are. J Transl Med 13:96. , DOI 10.1186/s12967-015-0439-7 Machida Y, Ueda Y, Shimasaki M et al, 2011, Relationship of aquaporin 1, 3, and 5 expression in lung cancer cells to cellular differentiation, invasive growth, and metastasis potential. Hum Pathol 42:669–678 Yoshida T, Hojo S, Sekine S et al, 2013, Expression of aquaporin-1 is a poor prognostic factor for stage II and III colon cancer. Mol Clin Oncol 1:953–958 Guo X, Sun T, Yang M, Li Z, Li Z, Gao Y, 2013, Prognostic value of combined aquaporin 3 and aquaporin 5 overexpression in hepatocellular carcinoma. Biomed Res Int 206525. , DOI 10. 1155/2013/206525 El Hindy N, Bankfalvi A, Herring A et al, 2013, Correlation of aquaporin-1 water channel protein expressionwith tumor angiogenesis in human astrocytoma. Anticancer Res 33:609–613 Jung HJ, Park JY, Jeon HS, Kwon TH, 2011, Aquaporin-5: a marker protein for proliferation and migration of human breast cancer cells. PLoS One 6:e28492. , DOI 10.1371/ journal.pone.0028492 Cannistra SA, 2004, Cancer of the ovary. N Engl J Med 351: 2519–2529 ZhangXY, Zhang PY, 2016, Recent perspectives of epithelial ovarian carcinoma. Oncol Lett 12:3055–3058 Yang JH, Shi YF, Cheng Q, Deng L, 2006, Expression and localization of aquaporin-5 in the epithelial ovarian tumors. Gynecol Oncol 100:294–299 Yang JH, Yu YQ, Yan CX, 2011, Localisation and expression of aquaporin subtypes in epithelial ovarian tumours. Histol Histopathol 26:1197–1205 Yang JH, Yan CX, Chen XJ, Zhu YS, 2011, Expression of aquaglyceroporins in epithelial ovarian tumours and their clinical significance. J Int Med Res 39:702–711 K. Sato et al Takal MK, Baykal C, Baser E et al, 2013, Does Aquaporin-1 expression have clinical significance in serous epithelial ovarian cancer? J Turk Ger Gynecol Assoc 14:130–135 Yang JH, Shi YF, Chen XD, Qi WJ, 2006, The influence of aquaporin-1 and microvessel density on ovarian carcinogenesis and ascites formation. Int J Gynecol Cancer 16(Suppl 1):400–405 Mobasheri A, Airley R, Hewitt SM, Marples D, 2005, Heterogeneous expression of the aquaporin 1, AQP1, water channel in tumors of the prostate, breast, ovary, colon and lung: a study using high density multiple human tumor tissue microarrays. Int J Oncol 26:1149–1158 Frede J, Fraser SP, Oskay-Ozcelik G et al, 2013, Ovarian cancer: ion channel and aquaporin expression as novel targets of clinical potential. Eur J Cancer 49:2331–2344 Hoque MO, Soria JC, Woo J et al, 2006, Aquaporin 1 is overexpressed in lung cancer and stimulates NIH-3 T3 cell proliferation and anchorage-independent growth. Am J Pathol 168: 1345–1353 Wu Z, Li S, Liu J et al, 2015, RNAi-mediated silencing of AQP1 expression inhibited the proliferation, invasion and tumorigenesis of osteosarcoma cells. Cancer Biol Ther 16:1332–1340 Meng F, Rui Y, Xu L,Wan C, Jiang X, Li G, 2014, Aqp1 enhances migration of bone marrow mesenchymal stem cells through regulation of FAK and β-catenin. Stem Cells Dev 23:66–75 Tomita Y, Dorward H, Yool AJ et al, 2017, Role of aquaporin 1 Signalling in Cancer development and progression. Int J Mol Sci 18:E299. , DOI 10.3390/ijms18020299 Zhang Z, Chen Z, Song Y, Zhang P, Hu J, Bai C, 2010, Expression of aquaporin 5 increases proliferation and metastasis potential of lung cancer. J Pathol 221:210–220 Woo J, Lee J, Kim MS, Jang SJ, Sidransky D, Moon C, 2008, The effect of aquaporin 5 overexpression on the Ras signaling pathway. Biochem Biophys Res Commun 367:291–298 Yan C, Zhu Y, Zhang X, Chen X, Zheng W, Yang J, 2014, Downregulated aquaporin 5 inhibits proliferation and migration of human epithelial ovarian cancer 3AO cells. J Ovarian Res 7:78. https://doi. org/10.1186/s13048-014-0078-2 Tiwari A, Hadley JA, Ramachandran R, 2014, Aquaporin 5 expression is altered in ovarian tumors and ascites-derived ovarian tumor cells in the chicken model of ovarian tumor. J Ovarian Res 7:99. , DOI 10.1186/s13048-014-0099-x Aishima S, Kuroda Y, Nishihara Yet al, 2007, Down-regulation of aquaporin-1 in intrahepatic cholangiocarcinoma is related to tumor progression and mucin expression. Hum Pathol 38:1819–1825 Takenawa J, Kaneko Y, Kishishita M et al, 1998, Transcript levels of aquaporin 1 and carbonic anhydrase IV as predictive indicators for prognosis of renal cell carcinoma patients after nephrectomy. Int J Cancer 79:1–7 Huang Y, Murakami T, Sano F et al, 2009, Expression of aquaporin 1 in primary renal tumors: a prognostic indicator for clear-cell renal cell carcinoma. Eur Urol 56:690–698 Hayashi Y, EdwardsNA, ProescholdtMA,Oldfield EH, MerrillMJ, 2007, Regulation and function of aquaporin-1 in glioma cells. Neoplasia 9:777–787 Ji C, Cao C, Lu S et al, 2008, Curcumin attenuates EGF-induced AQP3 up-regulation and cell migration in human ovarian cancer cells. Cancer Chemother Pharmacol 62:857–865 Huang YT, Zhou J, Shi S et al, 2015, Identification of estrogen response element in Aquaporin-3 gene that mediates estrogeninduced cell migration and invasion in estrogen receptor-positive breast Cancer. Sci Rep 5:12484. , DOI 10.1038/srep12484 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 263 EP 271 DI 10.1007/s12253-018-0456-y PG 9 ER PT J AU Li, L Fu, LQ Wang, HJ Wang, YY AF Li, Li Fu, Luo-Qin Wang, Hui-Ju Wang, Yuan-Yu TI CAP2 is a Valuable Biomarker for Diagnosis and Prognostic in Patients with Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; CAP2; Biomarker; Diagnosis; Prognosis ID Gastric cancer; CAP2; Biomarker; Diagnosis; Prognosis AB Cyclase-associated protein 2 (CAP2) protein is reported to be upregulated in hepatocellular carcinoma (HCC), human breast cancer, and malignant melanoma. However, its expression in gastric cancer remains unknown, this study was to investigate CAP2 expression and its prognostic significance in gastric cancer. Firstly, we analyzed the Oncomine databases to compare CAP2 mRNA expression in gastric cancer and normal tissues. CAP2 protein expression was analyzed in gastric cancer samples and non-tumor mucosa by RT-PCR and immunohistochemical analysis. Consequently, statistical analyses were performed to evaluate the clinicopathological significance of CAP2 expression in gastric cancer. CAP2 expression was significant higher in gastric cancer tissues than that in non-tumor mucosa at protein levels. CAP2 was up-regulated in 57.8% (252/436) of gastric cancer samples, while detected in only 10.9% (10/92) of non-tumor mucosa. Statistical analysis shows that the expression of CAP2 was correlated with tumor size, Lauren’s classification, depth of invasion, lymph node and distant metastases, and regional lymph node stage, TNM stage, but not with age, sex, histology classification, and histologic differentiation. Kaplan-Meier analysis indicated that high CAP2 expression was associated with poor overall survival (78.7%) in 203 of 252 gastric cancer patients. In stage I, II, and III tumors, the 5-year survival rate was lower in those with high expression of CAP2 than those with low expression. In stage IV tumors, the expression of CAP2 did not correlate with the 5-year survival rate. Multiple Cox regression analysis indicated CAP2 as an independent predictor for overall survival [hazard ratio (HR) = 2.045, 95% confidence interval: 1.445–2.895, p < 0.01], while Lauren’s classification, TNM stage, and expression of CAP2 were independent prognostic factors in patients with gastric cancer. For the first time, we found that CAP2 was upregulated in gastric cancer, and was associated with lymph node and distant metastases. CAP2 may serve as a prognostic indicator for patients with gastric cancer. C1 [Li, Li] People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People’s Hospital, Clinical Research Institute, 310014 Hangzhou, Zhejiang, China. [Fu, Luo-Qin] People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People’s Hospital, Clinical Research Institute, 310014 Hangzhou, Zhejiang, China. [Wang, Hui-Ju] People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People’s Hospital, Clinical Research Institute, 310014 Hangzhou, Zhejiang, China. [Wang, Yuan-Yu] Key Laboratory of Gastroenterology of Zhejiang ProvinceHangzhou, Zhejiang, China. RP Wang, YY (reprint author), Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, China. EM lywyy1979@126.com CR Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J, 2016, Cancer statistics in China, 2015. CA Cancer J Clin 66(2):115–132 Field J, Vojtek A, Ballester R, Bolger G, Colicelli J, Ferguson K, Gerst J, Kataoka T, Michaeli T, Powers S et al, 1990, Cloning and characterization of CAP, the S. Cerevisiae gene encoding the 70 kd adenylyl cyclase-associated protein. Cell 61(2):319–327 Fedor-Chaiken M, Deschenes RJ, Broach JRSRV, 1990, A gene required for RAS activation of adenylate cyclase in yeast. Cell 61(2):329–340 Peche V, Shekar S, Leichter M, Korte H, Schroder R, Schleicher M, Holak TA, Clemen CS, Ramanath-Y B, Pfitzer G, Karakesisoglou I, Noegel AA, 2007, CAP2, cyclase-associated protein 2, is a dual compartment protein. Cell Mol Life Sci 64(19–20):2702–2715 YilmazM, Christofori GEMT, 2009, The cytoskeleton. and cancer cell invasion Cancer Metastasis Rev 28(1–2):15–33 Xu L, Peng S, Huang Q, Liu Y, Jiang H, Li X, Wang J, 2016, Expression status of cyclase associated protein 2 as a prognostic marker for human breast cancer. Oncol Rep 36(4):1981–1988 Masugi Y, Tanese K, Emoto K, Yamazaki K, Effendi K, Funakoshi T, Mori M, Sakamoto M, 2015, Overexpression of adenylate cyclase-associated protein 2 is a novel prognostic marker in malignant melanoma. Pathol Int 65(12):627–634 Fu J, Li M, Wu DC, Liu LL, Chen SL, Yun JP, 2015, Increased expression of CAP2 indicates poor prognosis in hepatocellular carcinoma. Transl Oncol 8(5):400–406 Chen M, Zheng T, Han S, Zhang L, Bai Y, Fang X, Ding SZ, Yang Y, 2015, A preliminary study of plasma cyclase-associated protein 2 as a novel biomarker for early stageand alpha-fetoprotein negative hepatocellular carcinoma patients. Clin Res Hepatol Gastroenterol 39(2):215–221 Zhao ZS,Wang YY, Chu YQ et al, 2010, SPARC is associated with gastric cancer progression and poor survival of patients. Clin Cancer Res 16(1):260–268 Yu G, Swiston J, Young D, 1994, Comparison of human CAP and CAP2, homologs of the yeast adenylyl cyclase-associatedproteins. J Cell Sci 107(Pt 6):1671–1678 Effendi K, Yamazaki K,Mori T, Masugi Y, Makino S, SakamotoM, 2013, Involvement of hepatocellular carcinoma biomarker, cyclase-associated protein 2 in zebrafish body development and cancer progression. Exp Cell Res 319(1):35–44 Stockigt F, Peche VS, Linhart M, Nickenig G, Noegel AA, Schrickel JW, 2016, Deficiency of cyclase-associated protein 2 promotes arrhythmias associated with connexin43 maldistribution and fibrosis. Arch Med Sci 12(1):188–198 Field J, Ye DZ, Shinde M, Liu F, Schillinger KJ, Lu M, Wang T, Skettini M, Xiong Y, Brice AK, Chung DC, Patel VV, 2015, CAP2 in cardiac conduction, sudden cardiac death and eye development. Sci Rep 5:17256 Sakamoto M, Effendi K, Masugi Y, 2010, Molecular diagnosis of multistage hepato carcinogenesis. Jpn J Clin Oncol 40(9):891–896 Sakamoto M, 2009, Early HCC: diagnosis and molecular markers. J Gastroenterol 44(Suppl 19):108–111 Shibata R, Mori T, Du W, Chuma M, Gotoh M, Shimazu M, Ueda M, Hirohashi S, Sakamoto M, 2006, Overexpression of cyclaseassociated protein 2 in multistage hepatocarcinogenesis. Clin Cancer Res 12(18):5363–5368 Miao H, Jia Y, Xie S,Wang X, Zhao J, Chu Y, Zhou Z, Shi Z, Song X, Li L, 2014 Dec 5, Structural insights into the C1q domain of Caprin-2 in canonicalWnt signaling. J Biol Chem 289(49):34104– 34113 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 273 EP 279 DI 10.1007/s12253-018-0450-4 PG 7 ER PT J AU Liu, M Du, K Jiang, B Wu, X AF Liu, Maoxi Du, Kunli Jiang, Bo Wu, Xingye TI High Expression of PhospholipaseD2 Induced by Hypoxia Promotes Proliferation of Colon Cancer Cells through Activating NF- κ Bp65 Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon cancer; PLD2; Proliferation; NF-κBp65; Hypoxia ID Colon cancer; PLD2; Proliferation; NF-κBp65; Hypoxia AB Hypoxia is a typical feature of colon cancer occurrence and progression. We have reported that high expression and activity of PhospholipaseD2 (PLD2) induced by hypoxia in colon cancer cells. In order to further investigate the role of PLD2 in colon cancer under hypoxic conditions. MTT assay was used to detect the proliferation of human colon cancer cells (SW480 and SW620) under hypoxic conditions by decrease the PLD2 gene expression or inhibit the activity of PLD2. Expression level of p-P65/T-P65 and Cyclin D1 were detected in those cells treated as above through using western blot and RT-PCR analysis. Effect of NF-κBp65 inhibitor (BAY-117082) on the proliferation and expression level of Cyclin D1 and PLD2 of colon cancer cells under hypoxic conditions were further analyzed. As a result, decreased the expression of PLD2 or inhibited the activity of PLD2 leaded to the proliferation of hypoxia colon cancer cells reduced, and along with the expression level of p-P65/T-P65 and Cyclin D1 reduced. However, inhibition the expression level of p-P65/T-P65 lead to the proliferation and expression of Cyclin D1 in those hypoxia colon cancer cells also reduced. In vivo growth decreased in response to PLD2 and NF-κBp65 inhibition. Our study indicates that high expression of PLD2 induced by hypoxia promotes the proliferation of colon cancer cells, and it may elevate the expression level of Cyclin D1 through activating NF-κBp65 signaling pathway. Inhibition of the PLD2 expression may provide a new clue for treatment for colon cancer. C1 [Liu, Maoxi] Shanxi Cancer Hospital, Department of Anorectal Surgery, 030013 Taiyuan, China. [Du, Kunli] Xijing Hospital, Department of Anorectal Surgery, 710032 Xi’an, China. [Jiang, Bo] Shanxi Cancer Hospital, Department of Anorectal Surgery, 030013 Taiyuan, China. [Wu, Xingye] The First Affiliated Hospital of Chongqing Medical University, Department of Gastrointestinal Surgery, 400016 Chongqing, China. RP Jiang, B (reprint author), Shanxi Cancer Hospital, Department of Anorectal Surgery, 030013 Taiyuan, China. EM 13834567839@163.com CR Siegel R, Naishadham D, Jemal A, 2012, Cancer statistics, 2012. CA Cancer J Clinic 62:10–29 Wang Z, Sun Y, 2010, Targeting P53 for novel anticancer therapy. Transl Oncol 3(1):1–12 Rupnarain C, Dlamini Z, Naicker S, Bhoola K, 2004, Colon cancer: genomics and apoptotic events. Biol Chem 385(6):449–464 Uchida N, Okamura S, Kuwano H, 1999, Phospholipase D activity in human gastric carcinoma. Anticancer Res 19(1B):77–86 AhnMJ, Park SY, KimWK, Cho JH, Chang BJ, Kim DJ, 2012, A single nucleotide polymorphism in the phospholipase D1 gene is associated with risk of non-small lung cancer. Int J Biomed Sci 8(2):121–128 Park MH, do Min S, 2011, Quercetin-induced downregulation of PLD1 inhibits proliferation and invasion in U87 glioma cells. Biochem Biophys Res Commun 412(4):710–715 Toschi A, Edelstein J, Rockwell P, Ohh M, Foster DA, 2008, HIF alpha expression in VHL-deficient renal cancer cells is dependent on phospholipase D. Oncogene 27(19):2746–2753 Noh DY, Ahn SJ, Lee RA, Park IA, Kim JH, Suh PG, Ryu SH, Lee KH, Han JS, 2000, Overexpression of phospholipase D in human breast cancer tisues. Cancer Lett 161(2):207–214 Oshimoto H, Okamura S, Yoshida M, Mori M, 2003, Increased activity and expression of Phospholipase D2 in human colorectal cancer. Oncol Res 14(1):31–37 Saito M, Iwadate M, Higashimoto M, Ono K, 2007, Expression of phospholipase D2 in human colorectal carcinoma. Oncol Rep 18(5):1329–1334 Yamada Y, Hamajima N, Kato T, Iwata H, 2003, Association of a polymorphism of the phospholipase D2 gene with the prevalence of colorectal cancer. J Mol Med 81(2):126–131 Maoxi L, Kunli D, Zhongxue F, Shouru Z, Wu X, 2015, Hypoxiainducible factor 1-alpha up-regulates the expression of phospholipase D2 in colon cancer cells under hypoxic conditions.Med Oncol 32(1):394 Liu M, Zhongxue F, XongyeW, Kunli D, Shouru Z, Li Z Inhibition of phospholipaseD2 increases hypoxia-induced human colon cancer cell apoptosis through inactivating of the PI3K/AKT signaling pathway. Tumor Biol. , DOI 10.1007/s13277-015-4348-4 Liu T, Liu D, Liu J, Song JT, Gao SL, Li H, Hu LH, Lin BR, 2012, Effact of NF-kappa B inhibitors on the chemotherapy-induced apoptosis of the colon cancer cellHT-29. Exp Ther Med 4(4):716–722 Venkateswararao E, Sharma VK, Yun J, Kim Y, Jung SH, 2014, Anti-proliferative effect of chalcone derivatives through inactivation of NF-κB in human cancer cell. Bioorg Med Chem 22(13): 3386–3392 Kwun HJ, Lee JH, Min DS et al, 2003, Transcriptional repression of cyclin-dependent kinase inhibitor p21 gene by phospholipase D1 and D2[J]. FEBS Lett 544(1–3):38–44 Foster DA, Xu L, 2003, Phospholipase D in cell proliferation and cancer. Mol Cancer Res 1(11):789–800 zhao Y, Ehara H, Akao Y, Shamoto M, Nakagawa Y et al, 2000, Increased activity and intranyclear expression of phospholipase D2 in human renal cancer. Biochem Biophys Res Commun 278(1): 140–143 Kang DW, Min DS, 2109, Positive feedback regulation between phospholipase D and wnt signaling promotes wnt-driven anchorage- independent growth of colorectal cancer cells. PLoS One 5(8): 2010 Ye Q, Kantonen S, Gomez-Cambronero J, 2012, Serum deprivation confers the MDA-MB-231breast cancer line with an EGFR/ JANK3/PLD2 system that maximizes cancer cell invasion. J Mol Biol 425(4):755–766 Bruntz RC, Taylor HE, Lindsley CW, Brown H, 2014, Phospholipase D2 mediates survival signaling through direct regulation of AKT in glioblastoma cells. J Biol Chem 289(2):600–616 Shen Q, StantonML, FengW, RodriquezME, Ramondetta L, Chen L et al, 2010, Morphoproteomic analysis reveals an overexpressed and constitutively activated phospholipase D1-m TOR pathway in endometrial carcinoma. Int J Clin Exp Pathol 4(1):13–21 Wang Q, Zhou Y, Evers BM, 2006, Pkc delta-mediated regulation of FLIP expression in human cancer cells. Int J Cancer 118:326– 334 Park MH, Ahn B-H, Hong Y-K, Min DS Overexpression of phospholipase D enhances matrix metalloproteinase-2 expression and glioma cell invasion via protein kinase C and protein kinase a/ NF-κB/SP1-mdiated signaling pathways. Carcinogenesis 30(2): 356–365 Yamada M, Banno Y, Takuwa Y, Koda M, Hara A, Nozawa Y, 2004, Overxpression of phospholipase D prevents actinomycin D-induced apoptosis through potentiation of phosphoinositide 3- kinase signalling pathways in Chinese-hamster ovary cells. Biochem J 378(Pt2):649–656 Oh JW, Kim EY, koo BS, Lee HB, Kim KS, Han JS Der f 2 activates phospholipase D in human T lymphocytes from Dermatophagoides farina specific allergic individuals: involvement of protein kinase C-α. Exp Mol Med 36(2004):486–492 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta DeltaC(T), method. Methods 25:402–408 M. Liu et al NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 281 EP 290 DI 10.1007/s12253-018-0429-1 PG 10 ER PT J AU Kiss, K Regos, E Rada, K Firneisz, G Baghy, K Kovalszky, I AF Kiss, Katalin Regos, Eszter Rada, Kristof Firneisz, Gabor Baghy, Kornelia Kovalszky, Ilona TI Chronic Hyperglycaemia Induced Alterations of Hepatic Stellate Cells Differ from the Effect of TGFB1, and Point toward Metabolic Stress SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hyperglycaemia; TGFB1; Hepatic stellate cells; Fibrogenesis; Endoplasmic stress ID Hyperglycaemia; TGFB1; Hepatic stellate cells; Fibrogenesis; Endoplasmic stress AB The deleterious effect of hyperglycemia on the biology of the liver is supported by clinical evidence. It can promote the development of fatty liver, liver fibrosis, even liver cancer as complication of diabetes mellitus. As liver fibrosis is the consequence of hepatic stellate cell (HSC) activation, the questions were addressed whether alterations induced by high glucose concentration are directly related to TGFB1 effect, or other mechanisms are activated. In order to obtain information on the response of HSC for high glucose, LX-2 cells (an immortalized human HSC cell lineage) were cultured in 15.3 mM glucose containing medium for 21 days. The effect of glucose was compared to that of TGFB1. Our data revealed that chronic exposure of high glucose concentration initiated profound alteration of LX-2 cells and the effect is different from those observed upon interaction with TGFB1. Whereas TGFB1 induced the production of extracellular matrix proteins, high glucose exposure resulted in decreased MMP2 activity, retardation of type I collagen in the endoplasmic reticulum, with decreased pS6 expression, pointing to development of endoplasmic stress and sequestration of p21CIP1/WAF1 in the cytoplasm which can promote the proliferation of LX2 cells. C1 [Kiss, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Regos, Eszter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Rada, Kristof] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Firneisz, Gabor] Semmelweis University, 2nd Department of Internal Medicine, Szentkiralyi utca 46, H-1085 Budapest, Hungary. [Baghy, Kornelia] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. RP Kovalszky, I (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM koval@korb1.sote.hu CR Yokogawa K,Matsui-Yuasa I, Tamura A, Terada M, Kojima-Yuasa A, 2011, Inhibitory effects of Ecklonia cava extract on high glucose-induced hepatic stellate cell activation. Mar Drugs 9(12): 2793–2808. , DOI 10.3390/md9122793 Firneisz G, 2014, Non-alcoholic fatty liver disease and type 2 diabetes mellitus: the liver disease of our age? World J Gastroenterol 20(27):9072–9089. , DOI 10.3748/wjg.v20.i27.9072 Schuppan D, Afdhal NH, 2008, Liver cirrhosis. Lancet 371(9615): 838–851. , DOI 10.1016/S0140-6736(08)60383-9 Tolman KG, Fonseca V, Dalpiaz A, Tan MH, 2007, Spectrum of liver disease in type 2 diabetes and management of patients with diabetes and liver disease. Diabetes Care 30(3):734–743. https:// doi.org/10.2337/dc06-1539 Adams LA, Harmsen S, St Sauver JL, Charatcharoenwitthaya P, Enders FB, Therneau T, Angulo P, 2010, Nonalcoholic fatty liver disease increases risk of death among patients with diabetes: a community-based cohort study. Am J Gastroenterol 105(7):1567– 1573. , DOI 10.1038/ajg.2010.18 Picardi A, D'Avola D, Gentilucci UV, Galati G, Fiori E, Spataro S, Afeltra A, 2006, Diabetes in chronic liver disease: from old concepts to new evidence. Diabetes Metab Res Rev 22(4):274–283. , DOI 10.1002/dmrr.636 Paradis V, Perlemuter G, Bonvoust F, Dargere D, Parfait B, Vidaud M, Conti M, Huet S, Ba N, Buffet C, Bedossa P, 2001, High glucose and hyperinsulinemia stimulate connective tissue growth factor expression: a potential mechanism involved in progression to fibrosis in nonalcoholic steatohepatitis. Hepatology 34(4 Pt 1):738– 744. , DOI 10.1053/jhep.2001.28055 Brownlee M, 2001, Biochemistry and molecular cell biology of diabetic complications. Nature 414(6865):813–820. https://doi. org/10.1038/414813a Neyrinck AM, Cani PD, Dewulf EM, De Backer F, Bindels LB, Delzenne NM, 2009, Critical role of Kupffer cells in the management of diet-induced diabetes and obesity. Biochem Biophys Res Commun 385(3):351–356. , DOI 10.1016/j.bbrc.2009.05. 070 Friedman SL, 2000, Molecular regulation of hepatic fibrosis, an integrated cellular response to tissue injury. J Biol Chem 275(4): 2247–2250 Knittel T, Janneck T, Muller L, Fellmer P, Ramadori G, 1996, Transforming growth factor beta 1-regulated gene expression of Ito cells. Hepatology 24(2):352–360. , DOI 10.1053/jhep. 1996.v24.pm0008690404 Knittel T, Schuppan D, Meyer zum Buschenfelde KH, Ramadori G, 1992, Differential expression of collagen types I, III, and IV by fatstoring, Ito, cells in vitro. Gastroenterology 102(5):1724–1735 Lanthier N, Horsmans Y, Leclercq IA, 2009, The metabolic syndrome: how it may influence hepatic stellate cell activation and hepatic fibrosis. Curr Opin Clin Nutr Metab Care 12(4):404–411. , DOI 10.1097/MCO.0b013e32832c7819 Kiss K, Baghy K, Spisak S, Szanyi S, Tulassay Z, Zalatnai A, Lohr JM, Jesenofsky R, Kovalszky I, Firneisz G, 2015, Chronic hyperglycemia induces trans-differentiation of human pancreatic stellate cells and enhances the malignant molecular communication with human pancreatic cancer cells. PLoS One 10(5):e0128059. https:// doi.org/10.1371/journal.pone.0128059 Xu L, Hui AY, Albanis E, Arthur MJ, O'Byrne SM, Blaner WS, Mukherjee P, Friedman SL, Eng FJ, 2005, Human hepatic stellate cell lines, LX-1 and LX-2: new tools for analysis of hepatic fibrosis. Gut 54(1):142–151. , DOI 10.1136/gut.2004.042127 Bradford MM, 1976, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72:248–254 Moreira RK, 2007, Hepatic stellate cells and liver fibrosis. Arch Pathol Lab Med 131(11):1728–1734. , DOI 10.1043/1543- 2165(2007)131[1728:HSCALF]2.0.CO;2 Yin C, Evason KJ, Asahina K, Stainier DY, 2013, Hepatic stellate cells in liver development, regeneration, and cancer. J Clin Invest 123(5):1902–1910. , DOI 10.1172/JCI66369 Kordes C, Sawitza I, Gotze S, Herebian D, Haussinger D, 2014, Hepatic stellate cells contribute to progenitor cells and liver regeneration. J Clin Invest 124(12):5503–5515. , DOI 10.1172/ JCI74119 Li JT, Liao ZX, Ping J, Xu D, Wang H, 2008, Molecular mechanism of hepatic stellate cell activation and antifibrotic therapeutic strategies. J Gastroenterol 43(6):419–428. , DOI 10.1007/ s00535-008-2180-y Friedman SL, 2010, Evolving challenges in hepatic fibrosis. Nat Rev Gastroenterol Hepatol 7(8):425–436. , DOI 10.1038/ nrgastro.2010.97 MarkiewskiMM, DeAngelis RA, Lambris JD, 2006, Liver inflammation and regeneration: two distinct biological phenomena or parallel pathophysiologic processes? Mol Immunol 43(1–2):45–56. , DOI 10.1016/j.molimm.2005.06.019 Garcia-Compean D, Jaquez-Quintana JO, Gonzalez-Gonzalez JA, Maldonado-Garza H, 2009, Liver cirrhosis and diabetes: risk factors, pathophysiology, clinical implications and management. World J Gastroenterol 15(3):280–288 Hong F, Tuyama A, Lee TF, Loke J, Agarwal R, Cheng X, Garg A, Fiel MI, Schwartz M, Walewski J, Branch A, Schecter AD, Bansal MB(2009, Hepatic stellate cells express functional CXCR4: role in stromal cell-derived factor-1alpha-mediated stellate cell activation. Hepatology 49(6):2055–2067. , DOI 10.1002/hep.22890 Saiman Y, Jiao J, Fiel MI, Friedman SL, Aloman C, Bansal MB, 2015, Inhibition of the CXCL12/CXCR4 chemokine axis with AMD3100, a CXCR4 small molecule inhibitor, worsens murine hepatic injury. Hepatol Res 45(7):794–803. , DOI 10. 1111/hepr.12411 TsaiWC, Liang FC, Cheng JW, Lin LP, Chang SC, Chen HH, Pang JH, 2013, High glucose concentration up-regulates the expression of matrix metalloproteinase-9 and -13 in tendon cells. BMC Musculoskelet Disord 14:255. , DOI 10.1186/1471-2474- 14-255 Kim ES, Kim MS,Moon A, 2004, TGF-beta-induced upregulation of MMP-2 and MMP-9 depends on p38 MAPK, but not ERK signaling in MCF10A human breast epithelial cells. Int J Oncol 25(5):1375–1382 Nakagawa T, Lan HY, Glushakova O, Zhu HJ, Kang DH, Schreiner GF, Bottinger EP, Johnson RJ, Sautin YY, 2005, Role of ERK1/2 and p38 mitogen-activated protein kinases in the regulation of thrombospondin-1 by TGF-beta1 in rat proximal tubular cells and mouse fibroblasts. J Am Soc Nephrol 16(4):899–904. https://doi. org/10.1681/ASN.2004080689 Hugo C, 2003, The thrombospondin 1-TGF-beta axis in fibrotic renal disease. Nephrol Dial Transplant 18(7):1241–1245 Friedman SL, 2008, Mechanisms of hepatic fibrogenesis. Gastroenterology 134(6):1655–1669. , DOI 10.1053/j. gastro.2008.03.003 Vincent EE, Elder DJ, Thomas EC, Phillips L, Morgan C, Pawade J, Sohail M, MayMT, HetzelMR, Tavare JM(2011, Akt phosphorylation on Thr308 but not on Ser473 correlates with Akt protein kinase activity in human non-small cell lung cancer. Br J Cancer 104(11):1755–1761. , DOI 10.1038/bjc.2011.132 Bergmann C, Akhmetshina A, Dees C, Palumbo K, Zerr P, Beyer C, Zwerina J, Distler O, Schett G, Distler JH, 2011, Inhibition of glycogen synthase kinase 3beta induces dermal fibrosis by activation of the canonical Wnt pathway. Ann Rheum Dis 70(12):2191– 2198. , DOI 10.1136/ard.2010.147140 Dooley S, ten Dijke P, 2012, TGF-beta in progression of liver disease. Cell Tissue Res 347(1):245–256. , DOI 10.1007/ s00441-011-1246-y Lai WC, Zhou M, Shankavaram U, Peng G, Wahl LM, 2003, Differential regulation of lipopolysaccharide-induced monocyte matrix metalloproteinase, MMP)-1 and MMP-9 by p38 and extracellular signal-regulated kinase 1/2 mitogen-activated protein kinases. J Immunol 170(12):6244–6249 Singh R, Song RH, Alavi N, Pegoraro AA, Singh AK, Leehey DJ, 2001, High glucose decreases matrix metalloproteinase-2 activity in rat mesangial cells via transforming growth factor-beta1. Exp Nephrol 9(4):249–257. , DOI 10.1159/000052619 Bujor AM, Pannu J, Bu S, Smith EA, Muise-Helmericks RC, Trojanowska M, 2008, Akt blockade downregulates collagen and upregulates MMP1 in human dermal fibroblasts. J Invest Dermatol 128(8):1906–1914. , DOI 10.1038/jid.2008.39 Li X, Talts U, Talts JF, Arman E, Ekblom P, Lonai P, 2001, Akt/ PKB regulates laminin and collagen IV isotypes of the basement membrane. Proc Natl Acad Sci U S A 98(25):14416–14421. https:// doi.org/10.1073/pnas.251547198 Danne T, SpiroMJ, Spiro RG, 1993, Effect of high glucose on type IV collagen production by cultured glomerular epithelial, endothelial, and mesangial cells. Diabetes 42(1):170–177 Son G, Hines IN, Lindquist J, Schrum LW, Rippe RA, 2009, Inhibition of phosphatidylinositol 3-kinase signaling in hepatic stellate cells blocks the progression of hepatic fibrosis. Hepatology 50(5):1512–1523. , DOI 10.1002/hep.23186 Lee SJ, Seo KW, Kim CD, 2015, LPS increases 5-LO expression on monocytes via an activation of Akt-Sp1/NF-kappaB pathways. Korean J Physiol Pharmacol 19(3):263–268. , DOI 10. 4196/kjpp.2015.19.3.263 Du K, Montminy M, 1998, CREB is a regulatory target for the protein kinase Akt/PKB. J Biol Chem 273(49):32377–32379 Patel S, Santani D, 2009, Role of NF-kappa B in the pathogenesis of diabetes and its associated complications. Pharmacol Rep 61(4): 595–603 Suzuki A, Tsutomi Y, Akahane K, Araki T, Miura M, 1998, Resistance to Fas-mediated apoptosis: activation of caspase 3 is regulated by cell cycle regulator p21WAF1 and IAP gene family ILP. Oncogene 17(8):931–939. , DOI 10.1038/sj.onc. 1202021 Zhou BP, Liao Y, Xia W, Spohn B, Lee MH, Hung MC, 2001, Cytoplasmic localization of p21Cip1/WAF1 by Akt-induced phosphorylation in HER-2/neu-overexpressing cells. Nat Cell Biol 3(3): 245–252. , DOI 10.1038/35060032 Li Y, Dowbenko D, Lasky LA, 2002, AKT/PKB phosphorylation of p21Cip/WAF1 enhances protein stability of p21Cip/WAF1 and promotes cell survival. J Biol Chem 277(13):11352–11361. https:// doi.org/10.1074/jbc.M109062200 Tam AB, Mercado EL, Hoffmann A, Niwa M, 2012, ER stress activates NF-kappaB by integrating functions of basal IKK activity, IRE1 and PERK. PLoS One 7(10):e45078. , DOI 10.1371/ journal.pone.0045078 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 291 EP 299 DI 10.1007/s12253-018-0458-9 PG 9 ER PT J AU Li, Z Zhang, B You, W Li, P Youlin, K AF Li, Zhang Zhang, Bingqiang You, Wenxian Li, Pan Youlin, Kuang TI Rab23 Promotes Hepatocellular Carcinoma Cell Migration Via Rac1/TGF-β Signaling SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma cell; Rab23; Rac1; TGF-β; Migration ID Hepatocellular carcinoma cell; Rab23; Rac1; TGF-β; Migration AB Rab23 is a member of Ras-related small GTPase family, which plays a critical role in the progression of wide range of tumors. However, its biological function in hepatocellular carcinoma still remains unclear. Here, we investigated the effects of Rab23 on proliferation and migration in hepatocellular carcinoma cell and its potential mechanisms. We found over-expression of Rab23 promoted the Hep3B hepatocellular carcinoma cell migration, which could be reversed by Rab23 silencing. Rab23 induced Rac1 activation and followed progression of epithelial-mesenchymal transition (EMT) along with upregulation of N-cadherin, snail as well as vimentin and downregulation of E-cadherin via upregulating Transforming Growth Factor-β (TGF-β). Silencing Rac1 significantly attenuated Rab23-induced HepG2 migration and TGF-β. Moreover, knockdown of TGF-β effectively attenuated Rab23-induced EMT. Taken together, we demonstrated a mechanistic cascade of Rab23 enhancing Rac1 activation and subsequent TGF-β expression, leading to hepatocellular carcinoma cell migration. C1 [Li, Zhang] The First Affiliated Hospital of Chongqing Medical University, Gastroenterology, 400016 Chongqing, China. [Zhang, Bingqiang] The First Affiliated Hospital of Chongqing Medical University, Gastroenterology, 400016 Chongqing, China. [You, Wenxian] The First Affiliated Hospital of Chongqing Medical University, Gastroenterology, 400016 Chongqing, China. [Li, Pan] The First Affiliated Hospital of Chongqing Medical University, Gastroenterology, 400016 Chongqing, China. [Youlin, Kuang] Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 400016 Chongqing, China. RP Youlin, K (reprint author), Chongqing Medical University, The First Affiliated Hospital, Department of Urology, 400016 Chongqing, China. EM kyl361@163.com CR Massarweh NN, El-Serag HB, 2017, Epidemiology of hepatocellular carcinoma and intrahepatic cholangiocarcinoma. Cancer Control 24:1073274817729245 Forner A, Llovet JM, Bruix J, 2012, Hepatocellular carcinoma. In: Lancet, vol 379,, London), pp 1245–1255 Delevoye C, Goud B, 2015, Rab GTPases and kinesin motors in endosomal trafficking. Methods Cell Biol 130:235–246 Eggenschwiler JT, Espinoza E, Anderson KV, 2001, Rab23 is an essential negative regulator of the mouse sonic hedgehog signalling pathway. Nature 412:194–198 Cai ZZ, Xu LB, Cai JL, Wang JS, Zhou B, Hu H, 2015, Inactivation of Rab23 inhibits the invasion and motility of pancreatic duct adenocarcinoma. Genet Mol Res 14:2707–2715 Jiang Y, Han Y, Sun C,Han C, Han N, ZhiW, Qiao Q, 2016, Rab23 is overexpressed in human bladder cancer and promotes cancer cell proliferation and invasion. Tumour Biol 37:8131–8138 Wang M, Dong Q, Wang Y, 2016, Rab23 is overexpressed in human astrocytoma and promotes cell migration and invasion through regulation of Rac1. Tumour Biol 37:11049–11055 Hou Q, Wu YH, Grabsch H, Zhu Y, Leong SH, Ganesan K, Cross D, Tan LK, Tao J, Gopalakrishnan V, Tang BL, Kon OL, Tan P, 2008, Integrative genomics identifies RAB23 as an invasion mediator gene in diffuse-type gastric cancer. Cancer Res 68:4623– 4630 Denning KM, Smyth PC, Cahill SF, Finn SP, Conlon E, Li J, Flavin RJ, Aherne ST, Guenther SM, Ferlinz A, O'Leary JJ, Sheils OM, 2007, A molecular expression signature distinguishing follicular lesions in thyroid carcinoma using preamplification RT-PCR in archival samples. Mod Pathol 20:1095–1102 Liu YJ,Wang Q, LiW, HuangXH, ZhenMC, Huang SH, Chen LZ, Xue L, Zhang HW, 2007, Rab23 is a potential biological target for treating hepatocellular carcinoma. World J Gastroenterol 13:1010– 1017 Jansen S, Gosens R, Wieland T, Schmidt M, 2017, Paving the rho in cancer metastasis: rho GTPases and beyond. Pharmacol Ther Bid HK, Roberts RD,Manchanda PK, Houghton PJ, 2013, RAC1: an emerging therapeutic option for targeting cancer angiogenesis and metastasis. Mol Cancer Ther 12:1925–1934 Melzer C, Hass R, von der Ohe J, Lehnert H, Ungefroren H, 2017, The role of TGF-beta and its crosstalk with RAC1/RAC1b signaling in breast and pancreas carcinoma. Cell Commun Signal 15:19 Liu Y, Zeng C, Bao N, Zhao J, Hu Y, Li C, Chi S, 2015, Effect of Rab23 on the proliferation and apoptosis in breast cancer. Oncol Rep 34:1835–1844 Cheng L, Yang F, Zhou B, Yang H, Yuan Y, Li X, Han S, 2016, RAB23, regulated by miR-92b, promotes the progression of esophageal squamous cell carcinoma. Gene 595:31–38 Jian Q,Miao Y, Tang L, HuangM, Yang Y, BaW, Liu Y, Chi S, Li C, 2016, Rab23 promotes squamous cell carcinoma cell migration and invasion via integrin beta1/Rac1 pathway. Oncotarget 7:5342– 5352 Zhang XY,Mu JH, Liu LY, Zhang HZ, 2017, Upregulation of miR- 802 suppresses gastric cancer oncogenicity via targeting RAB23 expression. Eur Rev Med Pharmacol Sci 21:4071–4078 Dong C, Du Q, Wang Z, Wang Y, Wu S, Wang A, 2016, MicroRNA-665 suppressed the invasion and metastasis of osteosarcoma by directly inhibiting RAB23. Am J Transl Res 8:4975– 4981 Katz LH, Li Y, Chen JS,Munoz NM,Majumdar A, Chen J, Mishra L, 2013, Targeting TGF-beta signaling in cancer. Expert Opin Ther Targets 17:743–760 Mishra L, Banker T, Murray J, Byers S, Thenappan A, He AR, Shetty K, Johnson L, Reddy EP, 2009, Liver stem cells and hepatocellular carcinoma. Hepatology, Baltimore, MD, 49:318–329 Meindl-Beinker NM, Matsuzaki K, Dooley S, 2012, TGF-beta signaling in onset and progression of hepatocellular carcinoma. In: Digestive Diseases, vol 30,, Basel), pp 514–523 Wendt MK, Tian M, Schiemann WP, 2012, Deconstructing the mechanisms and consequences of TGF-beta-induced EMT during cancer progression. Cell Tissue Res 347:85–101 Caja L, Bertran E, Campbell J, Fausto N, Fabregat I, 2011, The transforming growth factor-beta, TGF-beta)mediates acquisition of a mesenchymal stem cell-like phenotype in human liver cells. J Cell Physiol 226:1214–1223 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 301 EP 306 DI 10.1007/s12253-018-0463-z PG 6 ER PT J AU Yang, Sh Liu, L Xu, D Li, X AF Yang, Shasha Liu, Lan Xu, Dongyuan Li, Xiangdan TI The Relationship of the TLR9 and TLR2 Genetic Polymorphisms with Cervical Cancer Risk: a Meta-Analysis of Case-Control Studies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TLR9; TLR2; Meta-analysis; Cervical cancer; Polymorphism ID TLR9; TLR2; Meta-analysis; Cervical cancer; Polymorphism AB This meta-analysis aimed to assess the association of common TLR9 and TLR2 gene polymorphisms (TLR9 1486 T/C, TLR9 G2848A, and TLR2–196 to −174 del/ins) with cervical cancer risk. Studies were searched in Scopus, Pubmed, Embase, and CNKI until December 2017. Both fixed-effects and random-effects models were applied to combine odds ratio (OR) and 95% confidence intervals (95% CI). A total of 11 studies including 7856 participants were identified. The pooled estimation revealed an increased risk of cervical cancer in Caucasian subjects carrying the C allele of the TLR9 1486 T/C polymorphism (OR = 1.46, 95% CI: 1.11–1.92, p = 0.007), while there was a decreased risk in Mixed subjects carrying the C allele (OR = 0.35, 95% CI: 0.15–0.82, p = 0.016). Concerning the TLR9 G2848A polymorphism, the A allele was associated with an increased risk of cervical cancer in Caucasians (OR = 1.19, 95% CI: 1.02–1.40, p = 0.030), whereas Asian and Mixed subjects showed no significant associations. No significant associations were demonstrated between the TLR2–196 to −174 del/ins polymorphism and cervical cancer. Our findings suggest that the TLR9 1486 T/C and G2848A polymorphisms contribute to cervical cancer risk, but there is no association of the TLR2–196 to −174 del/ins polymorphism with cervical cancer. C1 [Yang, Shasha] Medical College of Yanbian University, Center of Morphological Experiment, 133000 Yanji, China. [Liu, Lan] Affiliated Hospital of Yanbian University, Department of Pathology, 133000 Yanji, China. [Xu, Dongyuan] Medical College of Yanbian University, Center of Morphological Experiment, 133000 Yanji, China. [Li, Xiangdan] Medical College of Yanbian University, Center of Morphological Experiment, 133000 Yanji, China. RP Li, X (reprint author), Medical College of Yanbian University, Center of Morphological Experiment, 133000 Yanji, China. EM lixiangdan@ybu.edu.cn CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386 Patel JP, Lee EH, Mena CI,Walker CN, 2017, Effects ofmetformin on endothelial health and erectile dysfunction. Transl Androl Urol 6(3):556–565 Yu L, Chen S, 2008, Toll-like receptors expressed in tumor cells: targets for therapy. Cancer Immunol Immunother 57(9):1271–1278 Hasan UA, Bates E, Takeshita F, Biliato A, Accardi R, Bouvard V, MansourM, Vincent I, Gissmann L, Iftner T, SideriM, Stubenrauch F, TommasinoM(2007, TLR9 expression and function is abolished by the cervical cancer-associated human papillomavirus type 16. J Immunol 178(5):3186–3197 Lee JW, Choi JJ, Seo ES, Kim MJ, Kim WY, Choi CH, Kim TJ, Kim BG, Song SY, Bae DS, 2007, Increased toll-like receptor 9 expression in cervical neoplasia. Mol Carcinog 46(11):941–947 Fehri E, Ennaifer E, Ardhaoui M, Ouerhani K, Laassili T, Bel Haj Rhouma R, Guizani I, Boubaker S, 2014, Expression of toll-like receptor 9 increases with progression of cervical neoplasia in Tunisian women–a comparative analysis of condyloma, cervical intraepithelial neoplasia and invasive carcinoma. Asian Pac J Cancer Prev 15(15):6145–6150 Ghosh A, Dasgupta A, Bandyopadhyay A, Ghosh T, Dalui R, Biswas S, Banerjee U, Basu A, 2015, A study of the expression and localization of toll-like receptors 2 and 9 in different grades of cervical intraepithelial neoplasia and squamous cell carcinoma. Exp Mol Pathol 99(3):720–724 de Matos LG, Candido EB, Vidigal PV, Bordoni PH, Lamaita RM, Carneiro MM, da Silva-Filho AL, 2017, Association between tolllike receptor and tumor necrosis factor immunological pathways in uterine cervical neoplasms. Tumori 103(1):81–86 Hasimu A, Ge L, Li QZ, Zhang RP, Guo X, 2011, Expressions of toll-like receptors 3, 4, 7, and 9 in cervical lesions and their correlation with HPV16 infection in Uighur women. Chin J Cancer 30(5):344–350 DeCarlo CA, Rosa B, Jackson R, Niccoli S, Escott NG, Zehbe I, 2012, Toll-like receptor transcriptome in the HPV-positive cervical cancer microenvironment. Clin Dev Immunol 2012:785825 Mantel N, Haenszel W, 1959, Statistical aspects of the analysis of data from retrospective studies of disease. J Natl Cancer Inst 22(4): 719–748 DerSimonian R, Laird N, 1986, Meta-analysis in clinical trials. Control Clin Trials 7(3):177–188 Pandey S, Mittal RD, Srivastava M, Srivastava K, Singh S, Srivastava S, Mittal B, 2009, Impact of toll-like receptors [TLR] 2, −196 to −174 del, and TLR 4, Asp299Gly, Thr399Ile, in cervical cancer susceptibility in north Indian women. Gynecol Oncol 114(3):501–505 Pandey S, Mittal B, Srivastava M, Singh S, Srivastava K, Lal P, Mittal RD, 2011, Evaluation of toll-like receptors 3, c.1377C/T, and 9, G2848A, gene polymorphisms in cervical cancer susceptibility. Mol Biol Rep 38(7):4715–4721 Chen X, Wang S, Liu L, Chen Z, Qiang F, Kan Y, Shen Y, Wu J, Shen H, Hu Z, 2012, A genetic variant in the promoter region of toll-like receptor 9 and cervical cancer susceptibility. DNA Cell Biol 31(5):766–771 Roszak A, Lianeri M, Sowinska A, Jagodzinski PP, 2012, Involvement of toll-like receptor 9 polymorphism in cervical cancer development. Mol Biol Rep 39(8):8425–8430 Lai ZZ, Ni-Zhang PXL, Song L, 2013, Toll-like receptor 9, TLR9, gene polymorphisms associated with increased susceptibility of human papillomavirus-16 infection in patients with cervical cancer. J Int Med Res 41(4):1027–1036 Bodelon C, Madeleine MM, Johnson LG, Du Q, Galloway DA, Malkki M, Petersdorf EW, Schwartz SM, 2014, Genetic variation in the TLR and NF-κB pathways and cervical and vulvar cancer risk: a population-based case-control study. Int J Cancer 134(2): 437–444 Bi X, Xu X, Yu X,Wang N, Yin F,Wang Y, 2014, Study on TLRs polymorphisms and cervical cancer susceptibility. Prog Obstet Gynecol 23(7):520–523 Zidi S, Sghaier I, Gazouani E, Mezlini A, Yacoubi-Loueslati B, 2016, Evaluation of toll-like receptors 2/3/4/9 gene polymorphisms in cervical Cancer evolution. Pathol Oncol Res 22(2): 323–330 Xu X, Yin C,Yan Z, Hu Y, Yang H, 2017, The correlation of TLR9 gene polymorphisms with occurence and development of cervical cancer in Yunnan Han population. J Guizhou Med Univ 42(1):21–25 Martinez-Campos C, Bahena-Roman M, Torres-Poveda K, Burguete-Garcia AI, Madrid-Marina V, 2017, TLR9 gene polymorphism -1486T/C, rs187084, is associated with uterine cervical neoplasm in Mexican female population. J Cancer Res Clin Oncol 143(12):2437–2445 Jin Y, Qiu S, Shao N, Zheng J, 2017, Association of toll-like receptor gene polymorphisms and its interaction with HPV infection in determining the susceptibility of cervical cancer in Chinese Han population. Mamm Genome 28(5–6):213–219 Nasu K, Narahara H, 2010, Pattern recognition via the toll-like receptor system in the human female genital tract. Mediat Inflamm 2010:976024 Daud II, ScottME, Ma Y, Shiboski S, Farhat S, Moscicki AB, 2011, Association between toll-like receptor expression and human papillomavirus type 16 persistence. Int J Cancer 128(4):879–886 Mu X, Zhao J, Yuan X, Zhao X, Yao K, Liu Y, Zhao X, 2015, Gene polymorphisms of toll-like receptor 9 -1486T/C and 2848G/a in cervical Cancer risk. Int J Gynecol Cancer 25(7):1173–1178 Ding C, Yu H, Yu H, Qin H, 2012, TP53 codon 72 polymorphism with hepatocellular carcinoma: ametaanalysis. J IntMed Res 40(2): 446–454 Wang HL, Lu X, Yang X, Xu N, 2016, Association of MBL2 exon1 polymorphisms with high-risk human papillomavirus infection and cervical cancers: a meta-analysis. Arch Gynecol Obstet 294(6):1109–1116 Tan SC, Ismail MP, Duski DR, Othman NH, Ankathil R, 2017, FAS c.-671A>G polymorphism and cervical cancer risk: a casecontrol study and meta-analysis. Cancer Genet 211:18–25 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 307 EP 315 DI 10.1007/s12253-018-0465-x PG 9 ER PT J AU Balatoni, T Ladanyi, A Frohlich, G Czirbesz, K Kovacs, P Panczel, G Bence, E Plotar, V Liszkay, G AF Balatoni, Timea Ladanyi, Andrea Frohlich, Georgina Czirbesz, Kata Kovacs, Peter Panczel, Gitta Bence, Eszter Plotar, Vanda Liszkay, Gabriella TI Biomarkers Associated with Clinical Outcome of Advanced Melanoma Patients Treated with Ipilimumab SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Melanoma; Ipilimumab; Predictive factors; Lactate dehydrogenase ID Melanoma; Ipilimumab; Predictive factors; Lactate dehydrogenase AB Ipilimumab was the first immunotherapy approved for metastatic melanoma in decades and is currently registered as a second line treatment. However, new immunotherapies, in combination with ipilimumab, offer even better clinical outcomes for patients compared with single-agent treatments, at the expense of improved toxicity. The aim of this study was to evaluate the feasibility of ipilimumab outside the clinical trials and to identify survival predictors for treatment benefit. Data were collected on 47 advanced melanoma patients treated with ipilimumab between 2010 and 2015 at a single center. Association of clinical characteristics (including primary tumor characteristics), serum lactate dehydrogenase (LDH), erythrocyte sedimentation rate, absolute eosinophil, lymphocyte, and neutrophil count, neutrophil/lymphocyte and eosinophil/lymphocyte ratio with toxicity and clinical outcome were assessed using univariate and multivariate analysis. Median progression-free survival at a median follow-up of 10 months was 2.7 months and median overall survival was 9.8 months. Objective response was observed in 17%of patients and the disease control rate at week 24 was 40%. The 1- and 2-year survival rates documented were 40 and 28%, respectively. Significant association between high LDH level (>1.5× upper limit of normal) and decreased overall survival was demonstrated in uni- and multivariate analysis (hazard ratio [HR]: 3.554, 95% CI: 1.225–10.306, p = 0.019). Neither biomarkers nor clinical outcome were associated with toxicity. Using baseline serum LDH to identify patients most likely to benefit from ipilimumab therapy could serve as a simple and inexpensive biomarker of clinical outcome. C1 [Balatoni, Timea] National Institute of Oncology, Department of Dermatology, 7-9. Rath Gy. u., H-1122 Budapest, Hungary. [Ladanyi, Andrea] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Frohlich, Georgina] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Czirbesz, Kata] National Institute of Oncology, Department of Dermatology, 7-9. Rath Gy. u., H-1122 Budapest, Hungary. [Kovacs, Peter] National Institute of Oncology, Department of Dermatology, 7-9. Rath Gy. u., H-1122 Budapest, Hungary. [Panczel, Gitta] National Institute of Oncology, Department of Dermatology, 7-9. Rath Gy. u., H-1122 Budapest, Hungary. [Bence, Eszter] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Plotar, Vanda] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Liszkay, Gabriella] National Institute of Oncology, Department of Dermatology, 7-9. Rath Gy. u., H-1122 Budapest, Hungary. RP Balatoni, T (reprint author), National Institute of Oncology, Department of Dermatology, H-1122 Budapest, Hungary. EM balatoni.timea@hotmail.com CR Tsao H, Atkins MB, Sober AJ, 2004, Management of cutaneous melanoma. N Engl J Med 351:998–1012 Agarwala SS, 2009, Current systemic therapy for metastatic melanoma. Expert Rev Anticancer Ther 9:587–595 Hodi FS, O’Day SJ, McDermott DF et al, 2010, Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363:711–723 Schadendorf D, Hodi FS, Robert C,Weber JS, Margolin K, Hamid O, Patt D, Chen TT, Berman DM, Wolchok JD, 2015, Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol 33:1889–1894 Postow MA, Chesney J, Pavlik AC et al, 2015, Nivolumab and ipilimumab versus ipiliumab in untreated melanoma. N Engl J Med 372:2006–2017 Eggermont AM, Chiarion-Sileni V, Grob JJ et al, 2015, Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma, EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol 16:522–530 Ku GY, Yuan J, Page DB, Schroeder SE, Panageas KS, Carvajal RD, Chapman PB, Schwartz GK, Allison JP, Wolchok JD, 2010, Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: lymphocyte count after 2 doses correlates with survival. Cancer 116:1767–1775 Di Giacomo AM, Danielli R, Calabro L, Bertocci E, Nannicini C, Giannarelli D et al, 2011, Ipilimumab experience in heavily pre-treated patients with melanoma in an expanded access program at the University Hospital of Siena, Italy). Cancer Immunol Immunother 60:467–477 Di Giacomo AM, Calabro L, Danielli R et al, 2013, Long-term survival and immunological parameters in metastatic melanoma patients who responded to ipilimumab 10 mg/kg within an expanded access programme. Cancer Immunol Immunother 62:1021–1028 Delyon J, Mateus C, Lefeuvre D, Lanoy E, Zitvogel L, Chaput N, Roy S, Eggermont AM, Routier E, Robert C, 2013, Experience in daily practice with ipilimumab for the treatment of patients with metastatic melanoma: an early increase in lymphocyte and eosinophil counts is associated with improved survival. Ann Oncol 24: 1697–1703 Wilgenhof S, Du Four S, Vandenbroucke F, Everaert H, Salmon I, Lienard D, Marmol VD, Neyns B, 2013, Single-center experience with ipilimumab in an expanded access program for patients with pre-treated advanced melanoma. J Immunother 36:215–222 Kelderman S, Heemskerk B, van Tinteren H, van den Brom R, Hospers GA, van den Eertwegh A, Kapiteijn EW, de Groot JW, Soetekouw P, Jansen RL, Fiets E, Furness AJ, Renn A, Krzystanek M, Szallasi Z, Lorigan P, Gore ME, Schumacher TN, Haanen JB, Larkin JM, Blank CU, 2014, Lactate dehydrogenase as a selection criterion for ipilimumab treatment in metastatic melanoma. Cancer Immunol Immunother 63:449–458 Simeone E, Gentilcore G, Giannarelli D, Grimaldi AM, Caraco C, Curvietto M, Esposito A, Paone M, Palla M, Cavalcanti E, Sandomenico F, Petrillo A, Botti G, Fulciniti F, Palmieri G, Queirolo P, Marchetti P, Ferraresi V, Rinaldi G, Pistillo MP, Ciliberto G, Mozzillo N, Ascierto PA, 2014, Immunological and biological changes during ipilimumab treatment and their potential correlation with clinical response and survival in patients with advanced melanoma. Cancer Immunol Immunother 63:675–683 Berrocal A, Arance A, Lopez Martin JA, Soriano V, Munoz E, Alonso L, Espinosa E, Lopez Criado P, Valdivia J, Martin Algarra S, Spanish Melanoma Group, 2014, Ipilimumab for advanced melanoma: experience from the Spanish Expanded Access Program. Melanoma Res 24:577–583 Diem S, Kasenda B, Martin-Liberal J, Lee A, Chauhan D, Gore M, Larkin J, 2015, Prognostic score for patients with advanced melanoma treated with ipilimumab. Eur J Cancer 51:2785–2791 Ferrucci PF, Gandini S, Battaglia A, Alfieri S, di Giacomo AM, Giannarelli D, Cappellini GCA, de Galitiis F, Marchetti P, Amato G, Lazzeri A, Pala L, Cocorocchio E, Martinoli C, 2015, Baseline neutrophil-to-lymphocyte ratio is associated with outcome of ipilimumab-treated metastatic melanoma patients. Br J Cancer 112:1904–1910 Gebhardt C, Sevko A, Jiang H, Lichtenberger R, Reith M, Tarnanidis K, Holland-Letz T, Umansky L, Beckhove P, Sucker A, Schadendorf D, Utikal J, Umansky V, 2015, Myeloid cells and related chronic inflammatory factors as novel predictive markers in melanoma treatment with ipilimumab. Clin Cancer Res 21:5453–5459 Hannani D, Vetizou M, Enot D et al, 2015, Anticancer immunotherapy by CTLA-4 blockade: obligatory contribution of IL-2 receptors and negative prognostic impact of soluble CD25. Cell Res 25:208–224 Valpione S,Martinoli C, Fava P,Mocellin S, Campana LG, Quaglino P, Ferrucci PF, Pigozzo J, Astrua C, Testori A, Chiarion-Sileni V, 2015, Personalised medicine: Development and external validation of a prognostic model for metastatic melanoma patients treated with ipilimumab. Eur J Cancer 51:2086–2094 Khoja L, Atenafu EG, TempletonA, QyeY, ChappellMA, Saibil S, Hogg D, Butler MO, Joshua AM, 2016, The full blood count as a biomarker of outcome and toxicity in ipilimumab-treated cutaneous metastatic melanoma. Cancer Med 5:2792–2799 Martens A, Wistuba-Hamprecht K, Geukes Foppen M et al, 2016, Baseline peripheral blood biomarkers associated with clinical outcome of advanced melanoma patients treated with ipilimumab. Clin Cancer Res 22:2908–2918 Zaragoza J, Caille A, Beneton N, Bens G, Christiann F, Maillard H, Machet L, 2016, High neutrophil to lymphocyte ratio measured before starting ipilimumab treatment is associated with reduced overall survival in patients with melanoma. Br J Dermatol 174: 146–151 Attia P, Phan GQ, Maker AV, Robinson MR, Quezado MM, Yang JC, Sherry RM, Topalian SL, Kammula US, Royal RE, Restifo NP, Haworth LR, Levy C, Mavroukakis SA, Nichol G, Yellin MJ, Rosenberg SA, 2005, Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anticytotoxic T-lymphocyte antigen-4. J Clin Oncol 23:6043–6053 Downey SG, Klapper JA, Smith FO, Yang JC, Sherry RM, Royal RE, Kammula US, Hughes MS, Allen TE, Levy CL, Yellin M, Nichol G, White DE, Steinberg SM, Rosenberg SA, 2007, Prognostic factors related to clinical response in patients with metastatic melanoma treated by CTL-associated antigen-4 blockade. Clin Cancer Res 13:6681–6688 Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix- Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JWC, Leung SY, Yuen ST,Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA, 2002, Mutations of the BRAF gene in human cancer. Nature 417:949–954 Platz A, Egyhazi S, Ringborg U, Hansson J, 2008, Human cutaneous melanoma; a review of NRAS and BRAF mutation frequencies in relation to histogenic subclass and body site. Mol Oncol 1:394–405 Johnson DB, Lovly CM, Flavin M, Panageas KS, Ayers GD, Zhao Z, Iams WT, Colgan M, DeNoble S, Terry CR, Berry EG, Iafrate AJ, Sullivan RJ, Carvajal RD, Sosman JA, 2015, Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer Immunol Res 3:288–295 Ascierto PA, Simeone E, Sileni VC, Pigozzo J, Maio M, AltomonteM, del VecchioM, di Guardo L,Marchetti P, Ridolfi R, Cognetti F, Testori A, Bernengo M, Guida M, Marconcini R, Mandala M, Cimminiello C, Rinaldi G, Aglietta M, Queirolo P, 2014, Clinical experience with ipilimumab 3 mg/kg: real-world efficacy and safety data from an expanded access programme cohort. J Transl Med 12:116 Wolchok JD, Hoos A, O’Day S, Weber JS, Hamid O, Lebbe C, Maio M, Binder M, Bonsack O, Nichol G, Humphrey R, Hodi FS, 2009, Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res 15: 7412–7420 Margolin K, Ernstoff MS, Hamid O, Lawrence D, McDermott D, Puzanov I, Wolchok JD, Clark JI, Sznol M, Logan TF, Richards J, Michener T, Balogh A, Heller KN, Hodi FS, 2012, Ipilimumab in patients with melanoma and brain metastases: an open-label, phase 2 trial. Lancet Oncol 13:459–465 Queirolo P, Spagnolo F, Ascierto PA, Simeone E, Marchetti P, Scoppola A, del Vecchio M, di Guardo L, Maio M, di Giacomo AM, Antonuzzo A, Cognetti F, Ferraresi V, Ridolfi L, Guidoboni M, Guida M, Pigozzo J, Chiarion Sileni V, 2014, Efficacy and safety of ipilimumab in patients with advancedmelanoma and brain metastases. J Neuro-Oncol 118:109–116 Ugurel S, Thirumaran RK, Bloethner S, Gast A, Sucker A et al, 2007, B-RAF and N-RAS mutations are preserved during short time in vitro propagation and differentially impact prognosis. PLoS One 2(2):e236 Jakob JA, Basset RL, Cs N, Curry JL, Joseph RW et al, 2012, NRAS mutation status is an independent prognostic factor in metastatic melanoma. Cancer 118(16):4014–4023 Devitt B, Liu W, Salemi R, Wolfe R, Kelly J et al, 2011, Clinical outcome and pathological features associated with NRAS mutation in cutaneous melanoma. Pigment Cell Melanoma Res 24(4):666–672 CarlinoMS, Haydu LE, Kakavand H, Menzies AM, Hamilton AL, Yu B, Ng CC, CooperWA, Thompson JF, Kefford RF, O'Toole SA, Scolyer RA, Long GV, 2014, Correlation of BRAF and NRAS mutation status with outcome, site of distant metastasis and response to chemotherapy in metastatic melanoma. Br J Cancer 111(2):292–299 Ellehorst JA, Greene VR, Ekmekcioglu S, Warneke CL, Johnson MMet al, 2011, Clinical correlates of NRAS and BRAF mutations in primary human melanoma. Clin Cancer Res 17(2):229–235 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Guthrie GJ, Charles KA, Roxburgh CS, Horgan PG, McMillan DC, Clarke SJ, 2013, The systemic inflammation-based neutrophil-lymphocyte ratio: experience in patients with cancer. Crit Rev Oncol Hematol 88:218–230 Templeton AJ, McNamara MG, Seruga B et al, 2014, Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: a systematic review and meta-analysis. J Natl Cancer Inst 106:dju124 Petrulio CA, Kim-Schulze S, Kaufman HL, 2006, The tumor microenvironment and implications for cancer immunotherapy. Expert Opin Biol Ther 6:671–684 Vasievich EA, Huang L, 2011, The suppressive tumor microenvironment: a challenge in cancer immunotherapy. Mol Pharm 8:635–641 Schmidt H, Bastholt L, Geertsen P, Christensen IJ, Larsen S, Gehl J, von der Maase H, 2005, Elevated neutrophil and monocyte counts in peripheral blood are associated with poor survival in patients with metastatic melanoma: a prognostic model. Br J Cancer 93: 273–278 Gooden MJ, de Bock GH, Leffers N, Daemen T, Nijman HW, 2011, The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br J Cancer 105: 93–103 Ladanyi A, 2015, Prognostic and predictive significance of immune cells infiltrating cutaneous melanoma. Pigment Cell Melanoma Res 28:490–500 Simson L, Ellyard JI, Dent LA, Matthaei KI, Rothenberg ME, Foster PS, Smyth MJ, Parish CR, 2007, Regulation of carcinogenesis by IL-5 and CCL11: a potential role for eosinophils in tumor immune surveillance. J Immunol 178:4222–4229 Carretero R, Sektioglu IM, Garbi N, Salgado OC, Beckhove P, Hammerling GJ, 2015, Eosinophils orchestrate cancer rejection by normalizing tumor vessels and enhancing infiltration of CD8+ T cells. Nat Immunol 16:609–617 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 317 EP 325 DI 10.1007/s12253-018-0466-9 PG 9 ER PT J AU Inomata, M Hirai, T Seto, Z Tokui, K Taka, Ch Okazawa, S Kambara, K Ichikawa, T Imanishi, Sh Yamada, T Miwa, T Hayashi, R Tobe, K AF Inomata, Minehiko Hirai, Takahiro Seto, Zenta Tokui, Kotaro Taka, Chihiro Okazawa, Seisuke Kambara, Kenta Ichikawa, Tomomi Imanishi, Shingo Yamada, Toru Miwa, Toshiro Hayashi, Ryuji Tobe, Kazuyuki TI Clinical Parameters for Predicting the Survival in Patients with Squamous and Non-squamous-cell NSCLC Receiving PD-1 Inhibitor Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; Programmed death 1 inhibitor; Squamous cell lung cancer ID Non-small cell lung cancer; Programmed death 1 inhibitor; Squamous cell lung cancer AB We explored the associations between progression-free survival (PFS) after the initiation of PD-1 inhibitor therapy and the clinical parameters in patients with NSCLC. We reviewed the clinical data of patients with NSCLC treated with PD-1 inhibitor. Data of a total of 36 patients, including 16 patients with squamous cell NSCLC and 20 patients with non-squamous cell NSCLC were reviewed. Multivariate analyses identified EGFR status, C-reactive protein (CRP), and PFS following previous therapy as being significantly associated with the PFS after initiation of PD-1 inhibitor therapy in patients with NSCLC. In patients with squamous cell NSCLC, the blood neutrophil/lymphocyte ratio (NLR), serum lactate dehydrogenase (LDH), serum C-reactive protein (CRP), and PFS following previous therapy were identified as being significantly associated with the PFS after initiation of PD-1 inhibitor therapy. However, none of these associations, except for PFS following previous therapy, were found in patients with non-squamous cell NSCLC. NLR, LDH and CRP were associated with the PFS after initiation of PD-1 inhibitor therapy in patients with squamous cell NSCLC, and PFS following previous therapy was the common parameter associated with the PFS after initiation of PD-1 inhibitor therapy in both squamous-cell NSCLC and non-squamous-cell NSCLC patients. C1 [Inomata, Minehiko] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Hirai, Takahiro] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Seto, Zenta] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Tokui, Kotaro] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Taka, Chihiro] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Okazawa, Seisuke] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Kambara, Kenta] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Ichikawa, Tomomi] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Imanishi, Shingo] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Yamada, Toru] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Miwa, Toshiro] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. [Hayashi, Ryuji] Toyama University Hospital, Department of Medical OncologyToyama City, Japan. [Tobe, Kazuyuki] Toyama University Hospital, First Department of Internal Medicine, Sugitani 2630, 930-0194 Toyama City, Japan. RP Inomata, M (reprint author), Toyama University Hospital, First Department of Internal Medicine, 930-0194 Toyama City, Japan. EM minomata@med.u-toyama.ac.jp CR Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, Waterhouse D, Ready N, Gainor J, Aren Frontera O, Havel L, Steins M, Garassino MC, Aerts JG, Domine M, Paz-Ares L, Reck M, Baudelet C, Harbison CT, Lestini B, Spigel DR, 2015, Nivolumab versus docetaxel in advanced squamous-cell nonsmall- cell lung cancer. N Engl J Med 373:123–135 Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, Barlesi F, Kohlhaufl M, Arrieta O, Burgio MA, Fayette J, Lena H, Poddubskaya E, Gerber DE, Gettinger SN, Rudin CM, Rizvi N, Crino L, Blumenschein GR Jr, Antonia SJ, Dorange C, Harbison CT, Graf Finckenstein F, Brahmer JR, 2015, Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 373:1627– 1639 Taniguchi Y, Tamiya A, Isa SI, Nakahama K, Okishio K, Shiroyama T, Suzuki H, Inoue T, Tamiya M, Hirashima T, Imamura F, Atagi S, 2017, Predictive factors for poor progression-free survival in patients with non-small cell lung cancer treated with nivolumab. Anticancer Res 37:5857–5862 Oya Y, Yoshida T, Kuroda H, Mikubo M, Kondo C, Shimizu J, Horio Y, Sakao Y, Hida T, Yatabe Y, 2017, Predictive clinical parameters for the response of nivolumab in pretreated advanced non-small-cell lung cancer. Oncotarget 8:103117–103128 Mezquita L, Auclin E, Ferrara R, Charrier M, Remon J, Planchard D, Ponce S, Ares LP, Leroy L, Audigier-Valette C, Felip E, Zeron- Medina J, Garrido P, Brosseau S, Zalcman G, Mazieres J, Caramela C, Lahmar J, Adam J, Chaput N, Soria JC, Besse B, 2018, Association of the lung immune prognostic index with immune checkpoint inhibitor outcomes in patients with advanced nonsmall cell lung cancer. JAMA Oncol 4:351–357 Bagley SJ, Kothari S, Aggarwal C, Bauml JM, Alley EW, Evans TL, Kosteva JA, Ciunci CA, Gabriel PE, Thompson JC, Stonehouse-Lee S, Sherry VE, Gilbert E, Eaby-Sandy B, Mutale F, DiLullo G, Cohen RB, Vachani A, Langer CJ, 2017, Pretreatment neutrophil-to-lymphocyte ratio as a marker of outcomes in nivolumab-treated patients with advanced non-small-cell lung cancer. Lung Cancer 106:1–7 Kobayashi H, Omori S, Nakashima K, Wakuda K, Ono A, Kenmotsu H, Naito T, Murakami H, Endo M, Takahashi T, 2017, Response to the treatment immediately before nivolumab monotherapy may predict clinical response to nivolumab in patients with non-small cell lung cancer. Int J Clin Oncol 22(4):690–697 Inomata M, Hayashi R, Tokui K, Taka C, Okazawa S, Kambara K, Imanishi S, Suzuki K, Yamada T,Miwa T, Kashii T, Matsui S, Tobe K, 2014, Outcome and prognostic factors in patients with small cell lung cancer who receive third-line chemotherapy. Tumori 100:507– 511 Koukourakis MI, Kambouromiti G, Pitsiava D, Tsousou P, Tsiarkatsi M, Kartalis G, 2009, Serum C-reactive protein, CRP, levels in cancer patients are linked with tumor burden and are reduced by anti-hypertensive medication. Inflammation 32:169– 175 Lippitz BE, Harris RA, 2016, Cytokine patterns in cancer patients: a review of the correlation between interleukin 6 and prognosis. Oncoimmunology 5:e1093722 Lo CW, ChenMW, HsiaoM,Wang S, Chen CA, Hsiao SM, Chang JS, Lai TC, Rose-John S, Kuo ML, Wei LH, 2011, IL-6 transsignaling in formation and progression of malignant ascites in ovarian cancer. Cancer Res 71:424–434 Wehbe H, Henson R, Meng F, Mize-Berge J, Patel T, 2006, Interleukin-6 contributes to growth in cholangiocarcinoma cells by aberrant promoter methylation and gene expression. Cancer Res 66:10517–10524 Ulas A, Turkoz FP, Silay K, Tokluoglu S, Avci N, Oksuzoglu B, Alkis N, 2014, A laboratory prognostic index model for patients with advanced non-small cell lung cancer. PLoS One 9:e114471 Koukourakis MI, Giatromanolaki A, Sivridis E, Bougioukas G, Didilis V et al, 2003, Lactate dehydrogenase-5, LDH-5, overexpression in non-small-cell lung cancer tissues is linked to tumour hypoxia, angiogenic factor production and poor prognosis. Br J Cancer 89:877–885 Danner BC, Didilis VN,Wiemeyer S, Stojanovic T, Kitz J, Emmert A, Fuzesi L, Schondube FA, 2010, Long-term survival is linked to serum LDH and partly to tumour LDH-5 in NSCLC. Anticancer Res 30:1347–1351 Gottfried E, Kunz-Schughart LA, Ebner S, Mueller-Klieser W, Hoves S, Andreesen R, Mackensen A, Kreutz M, 2006, Tumorderived lactic acid modulates dendritic cell activation and antigen expression. Blood 107:2013–2021 Fischer K, Hoffmann P, Voelkl S, Meidenbauer N, Ammer J, Edinger M, Gottfried E, Schwarz S, Rothe G, Hoves S, Renner K, Timischl B, Mackensen A, Kunz-Schughart L, Andreesen R, Krause SW, Kreutz M, 2007, Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 109:3812– 3819 Templeton AJ, McNamara MG, Seruga B, Vera-Badillo FE, Aneja P, Ocana A, Leibowitz-Amit R, Sonpavde G, Knox JJ, Tran B, Tannock IF, Amir E, 2014, Prognostic role of neutrophil-tolymphocyte ratio in solid tumors: a systematic review and metaanalysis. J Natl Cancer Inst 106:dju124 Gooden MJ, de Bock GH, Leffers N, Daemen T, Nijman HW, 2011, The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br J Cancer 105: 93–103 Petrie HT, Klassen LW, Kay HD, 1985, Inhibition of human cytotoxic T lymphocyte activity in vitro by autologous peripheral blood granulocytes. J Immunol 134:230–234 el-Hag A, Clark RA, 1987, Immunosuppression by activated human neutrophils. Dependence on the myeloperoxidase system. J Immunol 139:2406–2413 Hiramatsu S, Tanaka H, Nishimura J, Sakimura C, Tamura T, Toyokawa T, Muguruma K, Yashiro M, Hirakawa K, Ohira M, 2018, Neutrophils in primary gastric tumors are correlated with neutrophil infiltration in tumor-draining lymph nodes and the systemic inflammatory response. BMC Immunol 19(13):16 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 327 EP 333 DI 10.1007/s12253-018-0473-x PG 7 ER PT J AU Eftekhari, A Peivand, Z Saadat, I Saadat, M AF Eftekhari, Alireza Peivand, Zahra Saadat, Iraj Saadat, Mostafa TI Association between Genetic Polymorphisms in Superoxide Dismutase Gene Family and Risk of Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; SOD1; SOD2; SOD3; Polymorphism ID Gastric cancer; SOD1; SOD2; SOD3; Polymorphism AB To determine the association between the SOD1 (Ins/Del), SOD2 (rs2758339, rs5746136), and SOD3 (rs2536512) polymorphisms and the risk of gastric cancer the present study performed. This is a case-control study, including 159 patients with gastric cancer and 242 healthy controls. All subjects were Persian Muslims living in Shiraz (south west Iran). Frequency matching by age and gender was performed. Genomic DNA was extracted from whole blood. Genotypes of the study polymorphism were determined using polymerase chain reaction based methods. The SOD1 Ins/Del and SOD3 rs2536512 polymorphisms did not appear to have relationship with gastric cancer risk. Both SOD2 polymorphisms (rs2758339, rs5746136) showed significant association with the risk of gastric cancer, under assumption that the variant alleles act as dominant alleles. There was significant association between smoking habit and the risk of gastric cancer (OR = 2.54, 95% CI = 1.61–4.02, P < 0.001). Smoker individuals having two putative high-risk genotypes showed elevated risk of gastric cancer compared with nonsmokers without high-risk genotypes, (OR = 5.75, 95% CI = 1.59–20.6, P = 0.007). Assuming that smoking habit and the genotypes are independent risk factors, there was a significant linear trend for the numbers of risk factors and gastric cancer risk (χ2 = 22.9, P < 0.001). This study indicates that the SOD2 polymorphism (rs2758339, rs5746136) is associated with increased risk of gastric cancer, especially in smoker individuals. C1 [Eftekhari, Alireza] Shiraz University, College of Sciences, Department of Biology, 71467-13565 Shiraz, Iran. [Peivand, Zahra] Shiraz University, College of Sciences, Department of Biology, 71467-13565 Shiraz, Iran. [Saadat, Iraj] Shiraz University, College of Sciences, Department of Biology, 71467-13565 Shiraz, Iran. [Saadat, Mostafa] Shiraz University, College of Sciences, Department of Biology, 71467-13565 Shiraz, Iran. RP Saadat, I (reprint author), Shiraz University, College of Sciences, Department of Biology, 71467-13565 Shiraz, Iran. EM isaadat@shirazu.ac.ir CR Gao S, Zhang X, Wang P, Dai L, Zhang J,Wang K, 2011, Genetic epidemiological analysis reveals a multi-gene additive model for gastric cancer. Familial Cancer 10:119–125 Draghicescu T, Roman IC, Paltanea L, 1998, Epidemiologic research on the genetic risk factors in gastric and colorectal cancer. Romanian J Morphol Embryol 44:149–152 Corso G, Seruca R, Roviello F, 2012, Gastric cancer carcinogenesis and tumor progression. Ann Ital Chir 83:172–176 Sjodahl K, Lu Y, Nilsen TI, YeW, Hveem K, Vatten L, Lagergren J, 2007, Smoking and alcohol drinking in relation to risk of gastric cancer: a population-based, prospective cohort study. Int J Cancer 120:128–132 Handa O, Naito Y, Yoshikawa T, 2011, Redox biology and gastric carcinogenesis: the role of helicobacter pylori. Redox Rep 16:1–7 Konat GW, 2003, H2O2-induced higher order chromatin degradation: a novel mechanism of oxidative genotoxicity. J Biosci 28:57–60 Berg D, Youdim MB, Riederer P, 2004, Redox imbalance. Cell Tissue Res 318:201–213 Nordberg J, Arner ES, 2001, Reactive oxygen species, antioxidants, and the mammalian thioredoxin system. Free Radic Biol Med 31:1287–1312 Kinnula VL, Crapo JD, 2004, Superoxide dismutases in malignant cells and human tumors. Free Radic Biol Med 36:718–744 Zelko IN, Mariani TJ, Folz RJ, 2002, Superoxide dismutase multigene family: a comparison of the CuZn-SOD, SOD1), Mn- SOD, SOD2), and EC-SOD, SOD3, gene structures, evolution, and expression. Free Radic Biol Med 33:337–349 Broom WJ, Greenway M, Sadri-Vakili G, Russ C, Auwarter KE, Glajch KE, Dupre N, Swingler RJ, Purcell S, Hayward C, Sapp PC, McKenna-Yasek D, Valdmanis PN, Bouchard JP, Meininger V, Hosler BA, Glass JD, Polack M, Rouleau GA, Cha JH, Hardiman O, Brown RH Jr, 2008, 50bp deletion in the promoter for superoxide dismutase 1, SOD1, reduces SOD1 expression in vitro and may correlate with increased age of onset of sporadic amyotrophic lateral sclerosis. Amyotroph Lateral Scler 9:229–237 Saify K, Saadat M, 2017, Influence of a 50bp Ins/Del polymorphism at promoter of the superoxide dismutase-1 on gene expression and risk of heroin dependency. Environ Health Prev Med 22:4 Wan XS, Devalaraja MN, St Clair DK, 1994, Molecular structure and organization of the human manganese superoxide dismutase gene. DNA Cell Biol 13:1127–1136 Hernandez-Saavedra D, McCord JM, 2009, Association of a new intronic polymorphism of the SOD2 gene, G1677T, with cancer. Cell Biochem Funct 27:223–227 Rosta K, Molvarec A, Enzsoly A, Nagy B, Ronai Z, Fekete A, Sasvari-Szekely M, Rigo J Jr, Ver A, 2009, Association of extracellular superoxide dismutase, SOD3, Ala40Thr gene polymorphism with pre-eclampsia complicated by severe fetal growth restriction. Eur J Obstet Gynecol Reprod Biol 142:134–138 Liu YY, Chen HY, Zhang ML, Tian D, Li S, Lee JY, 2012, Loss of fragile histidine triad and amplification of 1p36.22 and 11p15.5 in primary gastric adenocarcinomas. World J Gastroenterol 18:4522– 4532 Kimura Y, Noguchi T, Kawahara K, Kashima K, Daa T, Yokoyama S, 2004, Genetic alterations in 102 primary gastric cancers by comparative genomic hybridization: gain of 20q and loss of 18q are associated with tumor progression. Mod Pathol 17:1328–1337 Nakanishi M, Sakakura C, Fujita Y, Yasuoka R, Aragane H, Koide K, Hagiwara A, Yamaguchi T, Nakamura Y, Abe T, Inazawa J, Yamagishi H, 2000, Genomic alterations in primary gastric cancers analyzed by comparative genomic hybridization and clinicopathological factors. Hepatogastroenterology 47:658–662 Fan B, Dachrut S, Coral H, Yuen ST, Chu KM, Law S, Zhang L, Ji J, Leung SY, Chen X, 2012, Integration of DNA copy number alterations and transcriptional expression analysis in human gastric cancer. PLoS One 7:e29824 Liu XP, Li DY, Liu XL, Xu JD, Furuya T, Kawauchi S, Oga A, Sasaki K, 2009, Comparison of chromosomal aberrations between primary tumors and their synchronous lymph-node metastases in intestinal-type gastric carcinoma. Pathol Res Pract 205:105–111 Hidaka S, Yasutake T, Kondo M, Takeshita H, Yano H, Haseba M, Tsuji T, Sawai T, Nakagoe T, Tagawa Y, 2003, Frequent gains of 20q and losses of 18q are associated with lymph node metastasis in intestinal-type gastric cancer. Anticancer Res 23:3353–3357 WuMS, Chang MC, Huang SP, Tseng CC, Sheu JC, Lin YW, Shun CT, Lin MT, Lin JT, 2001, Correlation of histologic subtypes and replication error phenotype with comparative genomic hybridization in gastric cancer. Genes Chromosom Cancer 30:80–86 Aoki M, Yamamoto K, Noshiro H, Sakai K, Yokota J, Kohno T, Tokino T, Ishida S, Ohyama S, Ninomiya I, Uesaka K, Kitajima M, Shimada S, Matsuno S, Yano M, HiratsukaM, Sugimura H, Itoh F, Minamoto T, Maehara Y, Takenoshita S, Aikou T, Katai H, Yoshimura K, Takahashi T, Akagi K, Sairenji M, Yamamura Y, Sasazuki T, 2005, A full genome scan for gastric cancer. J Med Genet 42:83–87 Takeno A, Takemasa I, Doki Y, Yamasaki M, Miyata H, Takiguchi S, Fujiwara Y, Matsubara K, Monden M, 2008, Integrative approach for differentially over-expressed genes in gastric cancer by combining large-scale gene expression profiling and network analysis. Br J Cancer 99:1307–1315 Hendrickson DJ, Fisher JH, Jones C, Ho YS, 1990, Regional localization of human extracellular superoxide dismutase gene to 4pter-q21. Genomics 8:736–738 Huret JL, Delabar JM, Marlhens F, Aurias A, Nicole A, Berthier M, Tanzer J, Sinet PM, 1987, Down syndrome with duplication of a region of chromosome 21 containing the CuZn superoxide dismutase gene without detectable karyotypic abnormality. HumGenet 75: 251–257 Church SL, Grant JW,Meese EU, Trent JM, 1992, Sublocalization of the gene encoding manganese superoxide dismutase, MnSOD/ SOD2, to 6q25 by fluorescence in situ hybridization and somatic cell hybrid mapping. Genomics 14:823–825 Shin VY, Cho CH, 2005, Nicotine and gastric cancer. Alcohol 35: 259–264 Liu Y, Zha L, Li B, Zhang L, Yu T, Li L, 2014, Correlation between superoxide dismutase 1 and 2 polymorphisms and susceptibility to oral squamous cell carcinoma. Exp Ther Med 7:171–178 Ding G, Liu F, Shen B, Feng C, Xu J, Ding Q, 2012, The association between polymorphisms in prooxidant or antioxidant enzymes, myeloperoxidase, SOD2, and CAT, and genes and prostate cancer risk in the Chinese population of Han nationality. Clin Genitourin Cancer 10:251–255 Hubackova M, Vaclavikova R, Ehrlichova M, Mrhalova M, Kodet R, Kubackova K, Vrana D, Gut I, Soucek P, 2012, Association of superoxide dismutases and NAD(P)H quinone oxidoreductases with prognosis of patients with breast carcinomas. Int J Cancer 130:338–348 Rafiee L, Saadat I, Saadat M, 2010, Glutathione S-transferase genetic polymorphisms, GSTM1, GSTT1 and GSTO2, in three Iranian populations. Mol Biol Rep 37:155–158 Saadat M, 2015, Distribution of ACE insertion/deletion, I/D, polymorphism in Iranian populations. Mol Biol Res Commun 4:63–66 Fallahzadeh-Abarghooei L, Zahedi T,Mirabedi F, SaadatM(2015, Allelic prevalence of intron 3 insertion/deletion genetic polymorphism of DNA double-strand break repair gene XRCC4 in four Iranian populations. Egypt J Med Hum Genet 16:215–218 Russo M, Cocco S, Secondo A, Adornetto A, Bassi A, Nunziata A, Polichetti G, De Felice B, Damiano S, Seru R, Mondola P, Di Renzo G, 2011, Cigarette smoke condensate causes a decrease of the gene expression of cu-Zn superoxide dismutase, Mn superoxide dismutase, glutathione peroxidase, catalase, and free radicalinduced cell injury in SH-SY5Y human neuroblastoma cells. Neurotox Res 19:49–54 Esakky P, Hansen DA, Drury AM, Moley KH. Cigarette smoke condensate induces aryl hydrocarbon receptor-dependent changes in gene expression in spermatocytes. Reprod NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 335 EP 339 DI 10.1007/s12253-018-0470-0 PG 5 ER PT J AU Rosenberg, EJ Albersheim, AJ Sathianathen, JN Murugan, P Weight, JCh AF Rosenberg, E Joel Albersheim, A Jacob Sathianathen, J Niranjan Murugan, Paari Weight, J Christopher TI Five New Cases of Primary Renal Carcinoid Tumor: Case Reports and Literature Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Urology; Oncology; Kidney; Renal; Carcinoid; Neuroendocrine; Surgical pathology ID Urology; Oncology; Kidney; Renal; Carcinoid; Neuroendocrine; Surgical pathology AB Carcinoid tumors, a slow-growing NET, most commonly arise in the gastrointestinal tract (73.7%), followed by the bronchopulmonary system (25.1%), and least commonly in the genitourinary system (<1%). Primary carcinoid tumors of the kidney—with approximately 100 cases reported in the literature since the first described case in 1966—are thought to be so rare because neuroendocrine cells are not typically found in the renal parenchyma. Here we present a series of five cases at our institution with primary carcinoid tumors of the kidney followed by a literature review. In the literature we describe the diagnostic stains used to determine renal carcinoid tumors. We also describe why partial nephrectomies are the gold standard treatment in these cases, while radical nephrectomy can be used in certain circumstances. Limited research on treatment of metastasis of these tumors exists, but we summarize the results of existing treatments. Major prognostic factors and survival of patients with these tumors is described as well as the increased prevalence of this tumors in patients with horseshoe kidneys. This study presents five new cases of primary renal carcinoid tumors and a comprehensive review of the previously published cases.We are able to make limited prognostic predictions from currently published literature, but we will continue to learn from our patients’ long-term courses to draw conclusions about biological behavior, treatment outcomes, and recurrence of rare disease. C1 [Rosenberg, E Joel] University of Minnesota Medical School, Department of Urology, 420 Delaware St. SE MMC 394, 55455 Minneapolis, MN, USA. [Albersheim, A Jacob] University of Minnesota Medical School, Department of Urology, 420 Delaware St. SE MMC 394, 55455 Minneapolis, MN, USA. [Sathianathen, J Niranjan] University of Minnesota Medical School, Department of Urology, 420 Delaware St. SE MMC 394, 55455 Minneapolis, MN, USA. [Murugan, Paari] University of Minnesota Medical School, Department of Urology, 420 Delaware St. SE MMC 394, 55455 Minneapolis, MN, USA. [Weight, J Christopher] University of Minnesota Medical School, Department of Urology, 420 Delaware St. SE MMC 394, 55455 Minneapolis, MN, USA. RP Weight, JCh (reprint author), University of Minnesota Medical School, Department of Urology, 55455 Minneapolis, USA. EM cjweight@umn.edu CR Sun K, You Q, Zhao M, Yao H, Xiang H, Wang L, 2013, Concurrent primary carcinoid tumor arising within mature teratoma and clear cell renal cell carcinoma in the horseshoe kidney: report of a rare case and review of the literature. Int J Clin Exp Pathol 6(11):2578–2584 Seker KG, Sam E, Sahin S et al, 2017, Partial nephrectomy in horseshoe kidney: primary carcinoid tumor. Arch Ital Urol Androl 89(4):316–318 Korkmaz T, Seber S, Yavuzer D, Gumus M, Turhal NS, 2013, Primary renal carcinoid: treatment and prognosis. Crit Rev Oncol Hematol 87(3):256–264 Resnick ME, Unterberger H, McLoughlin PT, 1966, Renal carcinoid producing the carcinoid syndrome.Med Times 94(8):895–896 Litwinowicz R, Szpor J, Janus G, Worek M, Okon K, 2011, Primary carcinoid tumour in horseshoe kidney. Pol J Pathol 62(1): 72–74 Omiyale AO, Venyo AK, 2013, Primary carcinoid tumour of the kidney: a review of the literature. Adv Urol 2013:579396 Kulke MH, Mayer RJ, 1999, Carcinoid tumors. N Engl J Med 340(11):858–868 Xiang L, Kong B, 2013, PAX8 is a novel marker for differentiating between various types of tumor, particularly ovarian epithelial carcinomas. Oncol Lett 5(3):735–738 Romero FR, Rais-Bahrami S, Permpongkosol S, Fine SW, Kohanim S, Jarrett TW, 2006, Primary carcinoid tumors of the kidney. J Urol 176(6 Pt 1):2359–2366 Singh PP, Malhotra AS, Kashyap V, 2009, Carcinoid tumor of the kidney: an unusual renal tumor. Indian J Urol 25(4):537–538 Gedaly R, Jeon H, Johnston TD, McHugh PP, RowlandRG, Ranjan D, 2008, Surgical treatment of a rare primary renal carcinoid tumor with liver metastasis. World J Surg Oncol 6:41 IsmailM, Nielsen TK, Lagerveld B et al, 2018, Renal cryoablation: multidisciplinary, collaborative and perspective approach. Cryobiology 83:90-94 Chakravarthy A, Abrams RA, 1995, Radiation therapy in the management of patients withmalignant carcinoid tumors. Cancer 75(6): 1386–1390 Bondanelli M, Ambrosio MR, Zatelli MC, Cavazzini L, Al Jandalirifa’y L, degli Uberti EC, 2005, Regression of liver metastases of occult carcinoid tumorwith slow release lanreotide therapy. World J Gastroenterol 11(13):2041–2044 Sarmiento JM, Heywood G, Rubin J, Ilstrup DM, Nagorney DM, Que FG, 2003, Surgical treatment of neuroendocrine metastases to the liver: a plea for resection to increase survival. J Am Coll Surg 197(1):29–37 Mocellin S, Nitti D, 2013, Gastrointestinal carcinoid: epidemiological and survival evidence from a large population-based study, n = 25 531). Ann Oncol 24(12):3040–3044 Society TAC, 2016, Survival rates for lung carcinoid tumors. https://www.cancer.org/cancer/lung-carcinoid-tumor/detectiondiagnosis- staging/survival-rates.html#written_by. Accessed 5 May 2018 Krishnan B, Truong LD, Saleh G, SirbaskuDM, SlawinKM(1997, Horseshoe kidney is associated with an increased relative risk of primary renal carcinoid tumor. J Urol 157(6):2059–2066 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 341 EP 346 DI 10.1007/s12253-018-0481-x PG 6 ER PT J AU Koo, DH Do, IG Oh, S Lee, YGy Kim, K Sohn, HJ Park, SK Yang, HJ Jung, SY Park, ID Jeong, UK Kim, OH Kim, H Serrero, G Chun, HK AF Koo, Dong-Hoe Do, In-Gu Oh, Sukjoong Lee, Yun-Gyoo Kim, Kyungeun Sohn, Hee Jin Park, Soo-Kyung Yang, Hyo-Joon Jung, Suk Yoon Park, Il Dong Jeong, Uk Kyung Kim, Ook Hyung Kim, Hungdai Serrero, Ginette Chun, Ho-Kyung TI Prognostic Value of Progranulin in Patients with Colorectal Cancer Treated with Curative Resection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Progranulin; Colorectal neoplasms; Recurrence; Prognosis ID Progranulin; Colorectal neoplasms; Recurrence; Prognosis AB Progranulin (PGRN) has been characterized as an autocrine growth and survival factor and is known to stimulate tumorigenesis and proliferation of several types of cancer cell. However, little is known about the prognostic role of PGRN in colorectal cancer (CRC). A retrospective analysis was performed for patients with colorectal cancer who underwent curative resection between May 2013 and June 2015. PGRN expression in tumor cells was semi-quantitatively categorized (no expression, 0; weak/focal, 1+; moderate/focal or diffuse, 2+; strong/diffuse, 3+), and high expression was considered for tumors graded ≥2+ staining intensity. A total of 109 patients (28 stage I, 32 stage II, and 49 stage III) were analyzed. Thirty-eight patients (35%) had tumors with high PGRN expression, and there was a trend of elevated pre-operative CEA and CA19–9 levels in patients with high PGRN expressing tumors compared to those with low PGRN-expressing tumors (CEA, 49% vs. 21%; CA19–9, 21% vs. 7%). The 3–year recurrence-free survival (3Y–RFS) and overall survival rates were 83.7%(95% CI, 76.8–90.6) and 96.0%(95% CI, 92.3–99.7), respectively. Patients with high PGRN-expressing tumors had a worse rate of 3Y–RFS (66.8%) compared to those with low PGRN-expressing tumors (92.4%; p = 0.010). Multivariate analysis showed that high PGRN expression, age (>66 years), stage (III), and perineural invasion (+) were independent prognostic factors associated with poor RFS after adjusting for confounding factors including sex, MSI, tumor location, KRAS, and lympho-vascular invasion. PGRN overexpression was significantly associated with poor RFS in patients with CRC who have undergone curative resection. C1 [Koo, Dong-Hoe] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Division of Hematology/Oncology, Department of Internal Medicine, 29 Saemunan-ro, Jongno-gu, 03181 Seoul, South Korea. [Do, In-Gu] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of PathologySeoul, South Korea. [Oh, Sukjoong] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Division of Hematology/Oncology, Department of Internal Medicine, 29 Saemunan-ro, Jongno-gu, 03181 Seoul, South Korea. [Lee, Yun-Gyoo] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Division of Hematology/Oncology, Department of Internal Medicine, 29 Saemunan-ro, Jongno-gu, 03181 Seoul, South Korea. [Kim, Kyungeun] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of PathologySeoul, South Korea. [Sohn, Hee Jin] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of PathologySeoul, South Korea. [Park, Soo-Kyung] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Division of Gastroenterology, Department of Internal MedicineSeoul, South Korea. [Yang, Hyo-Joon] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Division of Gastroenterology, Department of Internal MedicineSeoul, South Korea. [Jung, Suk Yoon] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Division of Gastroenterology, Department of Internal MedicineSeoul, South Korea. [Park, Il Dong] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Division of Gastroenterology, Department of Internal MedicineSeoul, South Korea. [Jeong, Uk Kyung] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Surgery, 29 Saemunan-ro, Jongno-gu, 03181 Seoul, South Korea. [Kim, Ook Hyung] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Surgery, 29 Saemunan-ro, Jongno-gu, 03181 Seoul, South Korea. [Kim, Hungdai] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Surgery, 29 Saemunan-ro, Jongno-gu, 03181 Seoul, South Korea. [Serrero, Ginette] A&G Pharmaceutical, Inc.Columbia, MD, USA. [Chun, Ho-Kyung] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Surgery, 29 Saemunan-ro, Jongno-gu, 03181 Seoul, South Korea. RP Oh, S (reprint author), Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Division of Hematology/Oncology, Department of Internal Medicine, 03181 Seoul, South Korea. EM sukjoong.oh@samsung.com CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65:87–108. , DOI 10.3322/caac.21262 Ciombor KK, Bekaii-Saab T, 2018, A comprehensive review of sequencing and combination strategies of targeted agents in metastatic colorectal Cancer. Oncologist 23:25–34. , DOI 10. 1634/theoncologist.2017-0203 Serrero G, 2003, Autocrine growth factor revisited: PC-cellderived growth factor, progranulin), a critical player in breast cancer tumorigenesis. Biochem Biophys Res Commun 308:409–413 Yang D, Wang LL, Dong TT, Shen YH, Guo XS, Liu CY, Liu J, Zhang P, Li J, Sun YP, 2015, Progranulin promotes colorectal cancer proliferation and angiogenesis through TNFR2/Akt and ERK signaling pathways. Am J Cancer Res 5:3085–3097 LeeKW, Park SK,Yang HJ, Jung YS, ChoiKY, KimKE, Jung KU, Kim HO, Kim H, Chun HK, Park DI, 2016, Microsatellite instability status of interval colorectal cancers in a Korean population. Gut Liver 10:781–785. , DOI 10.5009/gnl15376 Lee J, Jang KT, Ki CS, Lim T, Park YS, Lim HY, Choi DW, Kang WK, Park K, Park JO, 2007, Impact of epidermal growth factor receptor, EGFR, kinase mutations, EGFR gene amplifications, and KRAS mutations on survival of pancreatic adenocarcinoma. Cancer 109:1561–1569. , DOI 10.1002/cncr.22559 Kim JH, Do IG, Kim K, Sohn JH, Kim HJ, JeonWK, Lee SR, Son BH, Shin JH, Nam H, Kwon HJ, Kim MS, Hong HP, Serrero G, Koo DH, 2016, Progranulin as a predictive factor of response to chemotherapy in advanced biliary tract carcinoma. Cancer Chemother Pharmacol 78:1085–1092. , DOI 10.1007/ s00280-016-3170-z Serrero G, Hawkins DM, Yue B, Ioffe O, Bejarano P, Phillips JT, Head JF, Elliott RL, Tkaczuk KR, Godwin AK,Weaver J, Kim WE, 2012, Progranulin, GP88, tumor tissue expression is associated with increased risk of recurrence in breast cancer patients diagnosed with estrogen receptor positive invasive ductal carcinoma. Breast Cancer Res 14:R26. , DOI 10.1186/bcr3111 Arechavaleta-Velasco F, Perez-Juarez CE, Gerton GL, Diaz-Cueto L, 2017, Progranulin and its biological effects in cancer. Med Oncol 34:194. , DOI 10.1007/s12032-017-1054-7 Serrero G, Ioffe OB, 2003, Expression of PC-cell-derived growth factor in benign and malignant human breast epithelium. Hum Pathol 34:1148–1154 Davidson B, Alejandro E, Florenes VA, Goderstad JM, Risberg B, Kristensen GB, Trope CG, Kohn EC, 2004, Granulin-epithelin precursor is a novel prognostic marker in epithelial ovarian carcinoma. Cancer 100:2139–2147. , DOI 10.1002/cncr.20219 Ho JC, Ip YC, Cheung ST, Lee YT, Chan KF, Wong SY, Fan ST, 2008, Granulin-epithelin precursor as a therapeutic target for hepatocellular carcinoma. Hepatology 47:1524–1532. , DOI 10.1002/hep.22191 Wang H, Sun Y, Liu S, Yu H, Li W, Zeng J, Chen C, Jia J, 2011, Upregulation of progranulin by helicobacter pylori in human gastric epithelial cells via p38MAPK and MEK1/2 signaling pathway: role in epithelial cell proliferation and migration. FEMS Immunol Med Microbiol 63:82–92. , DOI 10.1111/j.1574-695X.2011. 00833.x Edelman MJ, Feliciano J, Yue B, Bejarano P, Ioffe O, Reisman D, Hawkins D, Gai Q, Hicks D, Serrero G, 2014, GP88, progranulin): a novel tissue and circulating biomarker for non-small cell lung carcinoma. Hum Pathol 45:1893–1899. , DOI 10.1016/j. humpath.2014.05.011 Elkabets M, Gifford AM, Scheel C, Nilsson B, Reinhardt F, Bray MA, Carpenter AE, Jirstrom K, Magnusson K, Ebert BL, Ponten F, Weinberg RA, McAllister SS, 2011, Human tumors instigate granulin-expressing hematopoietic cells that promote malignancy by activating stromal fibroblasts in mice. J Clin Invest 121:784– 799. , DOI 10.1172/jci43757 Koo DH, Park CY, Lee ES, Ro J, Oh SW, 2012, Progranulin as a prognostic biomarker for breast cancer recurrence in patients who had hormone receptor-positive tumors: a cohort study. PLoS One 7: e39880. , DOI 10.1371/journal.pone.0039880 Han JJ, Yu M, Houston N, Steinberg SM, Kohn EC, 2011, Progranulin is a potential prognostic biomarker in advanced epithelial ovarian cancers. Gynecol Oncol 120:5–10. , DOI 10. 1016/j.ygyno.2010.09.006 Cheung ST, Cheung PF, Cheng CK, Wong NC, Fan ST, 2011, Granulin-epithelin precursor and ATP-dependent binding cassette, ABC)B5 regulate liver cancer cell chemoresistance. Gastroenterology 140:344–355. , DOI 10.1053/j.gastro. 2010.07.049 Giovannucci E, Harlan DM, Archer MC, Bergenstal RM, Gapstur SM, Habel LA, Pollak M, Regensteiner JG, Yee D, 2010, Diabetes and cancer: a consensus report. CA Cancer J Clin 60:207–221. , DOI 10.3322/caac.20078 Matsubara T, Mita A, Minami K, Hosooka T, Kitazawa S, Takahashi K, Tamori Y, Yokoi N, Watanabe M, Matsuo E, Nishimura O, Seino S, 2012, PGRN is a key adipokine mediating high fat diet-induced insulin resistance and obesity through IL-6 in adipose tissue. Cell Metab 15:38–50. , DOI 10.1016/j. cmet.2011.12.002 Tang W, Lu Y, Tian QY, Zhang Y, Guo FJ, Liu GY, Syed NM, Lai Y, Lin EA, Kong L, Su J, Yin F, Ding AH, Zanin-Zhorov A, Dustin ML, Tao J, Craft J, Yin Z, Feng JQ, Abramson SB, Yu XP, Liu CJ, 2011, The growth factor progranulin binds to TNF receptors and is therapeutic against inflammatory arthritis in mice. Science 332: 478–484. , DOI 10.1126/science.1199214 Song Z, Zhang X, Zhang L, Xu F, Tao X, Zhang H, Lin X, Kang L, Xiang Y, Lai X, Zhang Q, Huang K, Dai Y, Yin Y, Cao J, 2016, Progranulin plays a central role in host defense during Sepsis by promoting macrophage recruitment. Am J Respir Crit Care Med 194:1219–1232. , DOI 10.1164/rccm.201601-0056OC Limburg PJ, Vierkant RA, Fredericksen ZS, Leibson CL, Rizza RA, Gupta AK, Ahlquist DA, Melton LJ 3rd, Sellers TA, Cerhan JR, 2006, Clinically confirmed type 2 diabetes mellitus and colorectal cancer risk: a population-based, retrospective cohort study. Am J Gastroenterol 101:1872–1879. , DOI 10.1111/j. 1572-0241.2006.00725.x Shebl FM, Andreotti G, Meyer TE, Gao YT, Rashid A, Yu K, Shen MC, Wang BS, Han TQ, Zhang BH, Stanczyk FZ, Hsing AW, 2011, Metabolic syndrome and insulin resistance in relation to biliary tract cancer and stone risks: a population-based study in Shanghai, China. Br J Cancer 105:1424–1429. , DOI 10. 1038/bjc.2011.363 Peairs KS, Barone BB, Snyder CF, Yeh HC, Stein KB, Derr RL, Brancati FL, Wolff AC, 2011, Diabetes mellitus and breast cancer outcomes: a systematic review and meta-analysis. J Clin Oncol 29: 40–46. , DOI 10.1200/jco.2009.27.3011 Lee JY, Jeon I, Kim JW, Song YS, Yoon JM, Park SM, 2013, Diabetes mellitus and ovarian cancer risk: a systematic review and meta-analysis of observational studies. Int J Gynecol Cancer 23:402–412. , DOI 10.1097/IGC.0b013e31828189b2 Turati F, Polesel J, Di Maso M,Montella M, LibraM, Grimaldi M, Tavani A, Serraino D, La Vecchia C, Bosetti C, 2015, Diabetes mellitus and the risk of bladder cancer: an Italian case-control study. Br J Cancer 113:127–130. , DOI 10.1038/bjc.2015.178 Buraschi S, Xu SQ, Stefanello M, Moskalev I, Morcavallo A, Genua M, Tanimoto R, Birbe R, Peiper SC, Gomella LG, Belfiore A, Black PC, Iozzo RV, Morrione A, 2016, Suppression of progranulin expression inhibits bladder cancer growth and sensitizes cancer cells to cisplatin. Oncotarget 7:39980–39995. https:// doi.org/10.18632/oncotarget.9556 Qu H, Deng H, Hu Z, 2013, Plasma progranulin concentrations are increased in patients with type 2 diabetes and obesity and correlated with insulin resistance. Mediat Inflamm 2013:360190. https://doi. org/10.1155/2013/360190 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 343 EP 350 DI 10.1007/s12253-018-0520-7 PG 8 ER PT J AU Argyropoulou, M Veskoukis, SA Karanatsiou, PM Manolakelli, A Kostoglou-Athanassiou, I Vilaras, G Karameris, A Liadaki, K AF Argyropoulou, Marilena Veskoukis, S Aristidis Karanatsiou, Pagona-Maria Manolakelli, Aikaterini Kostoglou-Athanassiou, Ifigenia Vilaras, George Karameris, Andreas Liadaki, Kalliopi TI Low Prevalence of TERT Promoter, BRAF and RAS Mutations in Papillary Thyroid Cancer in the Greek Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TERT; BRAF; RAS; Mutations; Papillary thyroid cancer ID TERT; BRAF; RAS; Mutations; Papillary thyroid cancer AB Thyroid cancer is a common endocrine malignancy and displays a variety of histological patterns ranging from adenoma to malignant tumors. Molecular diagnostics have given significant insights into the genetic basis of thyroid tumorigenesis, known to be linked with signaling pathways affected by oxidative stress. We report for the first time a genotype study of TERT promoter combined with BRAF and RAS mutations in Papillary Thyroid Cancer (PTC) cases in the Greek population. Polymerase Chain Reaction and sequencing were used to identify TERT promoter (C228T, C250T, CC243-243TT) mutations, the BRAF (T1799A) mutation and mutations in codons 12, 13, 61 of the HRAS, KRAS and NRAS genes. The most common C228T TERT promoter mutation was identified in 2 PTC cases co-existing with the BRAF mutation. The BRAF T1799A mutation was detected in 10 PTC cases, while two different NRAS mutations in codon 61 (C181A and A182G) were found in 2 PTC cases. These mutations occur in a mutually exclusive manner. Our results indicate that despite the low frequencies, the study of the specific mutations should be encouraged because they are indicative of aggressive forms of thyroid cancer of the papillary histotype in this patient cohort, thus providing insights towards their therapeutic management. C1 [Argyropoulou, Marilena] University of Thessaly, Department of Biochemistry and Biotechnology, Viopolis, Mezourlo, 41500 Larissa, Greece. [Veskoukis, S Aristidis] University of Thessaly, Department of Biochemistry and Biotechnology, Viopolis, Mezourlo, 41500 Larissa, Greece. [Karanatsiou, Pagona-Maria] University of Thessaly, Department of Biochemistry and Biotechnology, Viopolis, Mezourlo, 41500 Larissa, Greece. [Manolakelli, Aikaterini] University of Thessaly, Department of Biochemistry and Biotechnology, Viopolis, Mezourlo, 41500 Larissa, Greece. [Kostoglou-Athanassiou, Ifigenia] General Hospital Korgialenio-Benakio National Red Cross, Department of Endocrinology, Erythrou Stavrou 1, 11526 Athens, Greece. [Vilaras, George] 417 Veterans Administration Hospital (Ν.Ι.Μ.Τ.S.), Department of Pathology, Section of Molecular Pathology, Monis Petraki 10, 11521 Athens, Greece. [Karameris, Andreas] 417 Veterans Administration Hospital (Ν.Ι.Μ.Τ.S.), Department of Pathology, Section of Molecular Pathology, Monis Petraki 10, 11521 Athens, Greece. [Liadaki, Kalliopi] University of Thessaly, Department of Biochemistry and Biotechnology, Viopolis, Mezourlo, 41500 Larissa, Greece. RP Liadaki, K (reprint author), University of Thessaly, Department of Biochemistry and Biotechnology, 41500 Larissa, Greece. EM kliad@bio.uth.gr CR Fletcher CDM(2013, Diagnostic histopathology of tumors. 4th edition, Elsevier Saunders Vol 2: 1177–1272 Nikiforov YE, Nikiforova MN, 2011, Molecular genetics and diagnosis of thyroid cancer. Nat Rev Endocrinol 7:569–580 Xing M, Haugen BR, Schlumberger M, 2013, Progress in molecular-based management of differentiated thyroid cancer. Lancet 381:1058–1069 Landa I, Ganly I, Chan TA,Mitsutake N,MatsuseM, Ibrahimpasic T, Ghossein RA, Fagin JA, 2013, TERT promoter mutations in thyroid Cancer: higher prevalence in advanced forms of the disease. J Clin Endocrinol Metab 98:E1562–E1566 Greider CW, Blackburn EH, 2004, Tracking telomerase. Cell 116: S83–S86 Heidenreich B, Rachakonda P, Hemminki K, Kumar R, 2014, TERT promoter mutations in cancer development. Curr Opin Genet Dev 24:30–37 Vinagre J, Almeida A, Populo H, Batista R, Lyra J, Pinto V et al, 2013, Frequency of TERT promoter mutations in human cancers. Nat Commun 4:2185 Horn S, Figl A, Rachakonda PS, Fischer C, Sucker A, Gast A, Kadel S, Moll I, Nagore E, Hemminki K, Schadendorf D, Kumar R, 2013, TERT promoter mutations in familial and sporadic melanoma. Science 339:959–961 Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA, 2013, Highly recurrent TERT promoter mutations in human melanoma. Science 339:957–959 Liu R, XingM(2016, TERT promoter mutations in thyroid cancer. Endocr Relat Cancer 23:R143–R155 Liu X, Bishop J, Shan Y, Pai S, Liu D, Murugan AK, Sun H, el-Naggar AK, Xing M, 2013, Highly prevalent TERT promoter mutations in aggressive thyroid cancers. Endocr Relat Cancer 20:603–610 Jin L, Chen E, Dong S, Cai Y, Zhang X, Zhou Y, Zeng R, Yang F, Pan C, Liu Y,WuW, XingM, Zhang X,Wang O, 2016, BRAF and TERT promotermutations in the aggressiveness of papillary thyroid carcinoma-a study of 653 patients. Oncotarget 7:18346–18355 Xing M, Liu R, Liu X, Murugan AK, Zhu G, Zeiger MA, Pai S, Bishop J, 2014, BRAF V600E and TERT promoter mutations cooperatively identify the most aggressive papillary thyroid cancer with highest recurrence. J Clin Oncol 32:2718–2726 Liu X, Qu S, Liu R, Sheng C, Shi X, Zhu G,Murugan AK, Guan H, Yu H, Wang Y, Sun H, Shan Z, Teng W, Xing M, 2014, TERT promoter mutations and their association with BRAF V600E mutation and aggressive clinicopathological characteristics of thyroid cancer. J Clin Endocrinol Metab 99:E1130–E1136 Xing M, 2005, BRAF mutation in thyroid cancer. Endocr Relat Cancer 12:245–262 Ciampi R, Nikiforov YE, 2005, Alterations of the BRAF gene in thyroid tumors. Endocr Pathol 16:163–172 Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM et al, 2004, Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 116:855–867 Azouzi N, Cailloux J, Cazarin JM, Knauf JA, Cracchiolo J, Al Ghuzlan A et al, 2017, NADPH oxidase NOX4 is a critical mediator of BRAFV600E-induced downregulation of the sodium/iodide symporter in papillary thyroid carcinomas. Antioxid Redox Signal 26:864–877 Nikiforova MN, Lynch RA, Biddinger PW, Alexander EK, Dorn IIGW, Tallini G et al, 2003, RAS point mutations and PAX8-PPAR gamma rearrangement in thyroid tumors: evidence for distinct molecular pathways in thyroid follicular carcinoma. J Clin Endocrinol Metab 88:2318–2326 Prior IA, Lewis PD, Mattos C, 2012, A comprehensive survey of RAS mutations in cancer. Cancer Res 72:2457–2467 Rosai J, Zampi G, Carcangiu ML, 1983, Papillary carcinoma of the thyroid. A discussion of its several morphologic expressions, with particular emphasis on the follicular variant. Am J Surg Pathol 7 :809–817 Song YS, Lim JA, Park YJ, 2015, Mutation profile of welldifferentiated thyroid Cancer in Asians. Endocrinol Metab 30: 252–262 Goutas N, Vlachodimitropoulos D, Bouka M, Lazaris AC, Nasioulas G, Gazouli M, 2008, BRAF and K-RAS mutation in a Greek papillary and medullary thyroid carcinoma cohort. Anticancer Res 28:305–308 Jung CK, LittleMP, Lubin JH, Brenner AV,Wells SA Jr, Sigurdson AJ et al, 2014, The increase in thyroid cancer incidence during the last four decades is accompanied by a high frequency of BRAF mutations and a sharp increase in RAS mutations. J Clin Endocrinol Metab 99:E276–E285 Guan H, Ji M, Bao R,Yu H,Wang Y, Hou P, ZhangY, Shan Z, Teng W, Xing M, 2009, Association of high iodine intake with the T1799A BRAF mutation in papillary thyroid cancer. J Clin Endocrinol Metab 94:1612–1617 Frasca F, Nucera C, Pellegriti G, Gangemi P, Attard M, Stella M, Loda M, Vella V, Giordano C, Trimarchi F, Mazzon E, Belfiore A, Vigneri R, 2008, BRAF(V600E, mutation and the biology of papillary thyroid cancer. Endocr Relat Cancer 15:191–205 Schulten HJ, Salama S, Al-Mansouri Z, Alotibi R, Al- Ghamdi K, Al-Hamour OA et al, 2012, BRAF mutations in thyroid tumors from an ethnically diverse group. Hered Cancer Clin Pract 10:10–17 Trovisco V, Soares P, Preto A, de Castro IV, Lima J, Castro P, Maximo V, Botelho T, Moreira S, Meireles AM, Magalhaes J, Abrosimov A, Cameselle-Teijeiro J, Sobrinho-Simoes M, 2005, Type and prevalence of BRAF mutations are closely associated with papillary thyroid carcinoma histotype and patients' age but not with tumour aggressiveness. Virchows Arch 446:589–595 Nikiforova MN, Kimura ET, Gandhi M, Biddinger PW, Knauf JA, Basolo F, Zhu Z, Giannini R, Salvatore G, Fusco A, Santoro M, Fagin JA, Nikiforov YE, 2003, BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas. J Clin Endocrinol Metab 88:5399–5404 Murugan AK, Qasem E, Al-Hindi H, Shi Y, Alzahrani AS, 2016, Classical V600E and other non-hotspot BRAF mutations in adult differentiated thyroid cancer. J Transl Med 14:204 Schulten HJ, Salama S, Al-Ahmadi A, Al-Mansouri Z, Mirza Z, Al-Ghamdi K et al, 2013, Comprehensive survey of HRAS, KRAS, and NRAS mutations in proliferative thyroid lesions from an ethnically diverse population. Anticancer Res 33:4779–4784 Zhu Z, Gandhi M, Nikiforova MN, Fischer AH, Nikiforov YE, 2003, Molecular profile and clinical-pathologic features of the follicular variant of papillary thyroid carcinoma: an unusually high prevalence of ras mutations. Am J Clin Pathol 120:71–77 Jang EK, Song DE, Sim SY, Kwon H, Choi YM, Jeon MJ, Han JM, Kim WG, Kim TY, Shong YK, Kim WB, 2014, NRAS codon 61 mutation is associated with distant metastasis in patients with follicular thyroid carcinoma. Thyroid 24:1275–1281 Carta C, Moretti S, Passeri L, Barbi F, Avenia N, Cavaliere A, Monacelli M, Macchiarulo A, Santeusanio F, Tartaglia M, Puxeddu E, 2006, Genotyping of an Italian papillary thyroid carcinoma cohort revealed high prevalence of BRAF mutations, absence of RAS mutations and allowed the detection of a new mutation of BRAF oncoprotein, BRAF(V599lns)). Clin Endocrinol 64:105–109 Melo M, Gaspar da Rocha A, Vinagre J et al, 2014, TERT promoter mutations are a major Indicator of poor outcome in differentiated thyroid carcinomas. J Clin Endocrinol Metab 99:E754–E765 George JR, Henderson YC,Williams MD, Roberts DB, Hei H, Lai SY, Clayman GL, 2015, Association of TERT promoter mutation, but not BRAF mutation, with increased mortality in PTC. J Clin Endocrinol Metab 100:E1550–E1559 Zhang Q, Liu SZ, Zhang Q, Guan YX, Chen QJ, Zhu QY, 2016, Meta-analyses of association between BRAF(V600E, mutation and clinicopathological features of papillary thyroid carcinoma. Cell Physiol Biochem 38:763–776 Xing M, Alzahrani AS, Carson KA, Shong YK, Kim TY, Viola D, Elisei R, Bendlova B, Yip L, Mian C, Vianello F, Tuttle RM, Robenshtok E, Fagin JA, Puxeddu E, Fugazzola L, Czarniecka A, Jarzab B, O'Neill CJ, Sywak MS, Lam AK, Riesco-Eizaguirre G, Santisteban P, Nakayama H, Clifton-Bligh R, Tallini G, Holt EH, Sykorova V, 2015, Association between BRAF V600E mutation and recurrence of papillary thyroid cancer. J Clin Oncol 33:42–50 Xing M, Alzahrani AS, Carson KA, Viola D, Elisei R, Bendlova B, Yip L, Mian C, Vianello F, Tuttle RM, Robenshtok E, Fagin JA, Puxeddu E, Fugazzola L, Czarniecka A, Jarzab B, O’Neill CJ, Sywak MS, Lam AK, Riesco-Eizaguirre G, Santisteban P, Nakayama H, Tufano RP, Pai SI, Zeiger MA, Westra WH, Clark DP, Clifton-Bligh R, Sidransky D, Ladenson PW, Sykorova V, 2013, Association between BRAF V600E mutation and mortality in patients with papillary thyroid cancer. JAMA 309:1493–1501 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 347 EP 354 DI 10.1007/s12253-018-0497-2 PG 8 ER PT J AU Yang, X Liu, G Zang, L Li, D Yu, F Xiang, X Li, W AF Yang, Xiaolin Liu, Geling Zang, Luyang Li, Ding Yu, Fang Xiang, Xiuxiu Li, Weijuan TI ZNF703 is Overexpressed in Papillary Thyroid Carcinoma Tissues and Mediates K1 Cell Proliferation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ZNF703; Papillary thyroid cancer; siRNA; Proliferation; Apoptosis; Invasion ID ZNF703; Papillary thyroid cancer; siRNA; Proliferation; Apoptosis; Invasion AB Zinc finger protein 703 (ZNF703), a member of the NET family of transcription factors, has recently emerged as an important player in the development of several types of cancers, though its role in papillary thyroid cancer (PTC) has not been characterized. We investigated the expression of ZNF703, its association with the most common genetic mutation in PTC, BRAF V600E, and its potential use as a therapeutic target. Real-time PCR, immunohistochemical staining, and western blot analysis of ZNF703 expression were performed for 36 cases of PTC and corresponding normal thyroid tissues. ZNF703 mRNA and protein expression was found to be significantly higher in PTC compared to normal thyroid tissues (P < 0.05). Furthermore, expression was associated with the tumor size, lymph node metastasis, and advanced disease stage. Immunohistochemical results showed that there was no correlation between ZNF703 protein levels and BRAF V600E mutation. The human PTC cell line K1, which has a BRAF V600E mutation, was selected for further investigation. Using small interfering RNA (siRNA), ZNF703 was shown to contribute to the proliferation, apoptosis, and invasion of K1 cells. ZNF703-siRNA downregulated E2F1 and MMP9 protein expression and enhanced the expression of p27 protein (P < 0.05), but had no effects on BRAF V600E protein levels. These results suggest that ZNF703 may be of potential use as a new marker for PTC prognosis and therapy that functions independent of BRAF V600E expression. C1 [Yang, Xiaolin] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Liu, Geling] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Zang, Luyang] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Li, Ding] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Yu, Fang] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Xiang, Xiuxiu] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. [Li, Weijuan] Tangshan Workers Hospital, Department of Endocrinology (Section I)Tangshan, China. RP Liu, G (reprint author), Tangshan Workers Hospital, Department of Endocrinology (Section I), Tangshan, China. EM tsliugeling@126.com CR Pellegriti G, Frasca F, Regalbuto C et al, 2013)Worldwide increasing incidence of thyroid cancer: update on epidemiology and risk factors. J Cancer Epidemiol 2013(1):965212 Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM, 2014, Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 74(11):2913–2921 Pathak KA, Leslie WD, Klonisch TC et al, 2013, The changing face of thyroid cancer in a population-based cohort. Cancer Med 2(4):537–544 Davies L,Welch HG, 2006, Increasing incidence of thyroid cancer in the United States, 1973-2002. JAMA 295(18):2164–2167 Allen JG, Gale S, Zoeller RT, Spengler JD, Birnbaum L, McNeely E, 2016, PBDE flame retardants, thyroid disease, and menopausal status in U.S. women. Environ Health 15(1):60 Aschebrookkilfoy B, DellaValle CT, Purdue M et al, 2015, Polybrominated diphenyl ethers and thyroid cancer risk in the prostate, colorectal, lung, and ovarian Cancer screening trial cohort.Am J Epidemiol 181(11):883–888 Ward MH, Kilfoy BA, Weyer PJ, Anderson KE, Folsom AR, Cerhan JR, 2010, Nitrate intake and the risk of thyroid cancer and thyroid disease. Epidemiology 21(3):389–395 Ostroumova E, Gudzenko N, Brenner A, Gorokh Y, Hatch M, Prysyazhnyuk A, Mabuchi K, Bazyka D, 2014, Thyroid cancer incidence in Chornobyl liquidators in Ukraine: SIR analysis, 1986-2010. Eur J Epidemiol 29(5):337–342 Katanoda K, Kamo K, Tsugane S, 2016, Quantification of the increase in thyroid cancer prevalence in Fukushima after the nuclear disaster in 2011–a potential overdiagnosis? Jpn J Clin Oncol 46(3):284–286 Sinnott B, Ron E, Schneider AB, 2010, Exposing the thyroid to radiation: a review of its current extent, risks, and implications. Endocr Rev 31(5):756–773 Camargo RY, Tomimori EK, Neves SC et al, 2008, Thyroid and the environment: exposure to excessive nutritional iodine increases the prevalence of thyroid disorders in Sao Paulo, Brazil. Eur J Endocrinol 159(3):293–299 Leenhardt L, Grosclaude P, 2011, Epidemiology of thyroid carcinoma over the world. Ann Endocrinol, Paris, 72(2):136–148 Rinaldi S, LiseM, Clavel-Chapelon F, Boutron-RuaultMC, Guillas G, Overvad K, Tjonneland A, Halkjaer J, Lukanova A, Kaaks R, Bergmann MM, Boeing H, Trichopoulou A, Zylis D, Valanou E, Palli D, Agnoli C, Tumino R, Polidoro S, Mattiello A, Bas Buenode- Mesquita H, Peeters PH, Weiderpass E, Lund E, Skeie G, Rodriguez L, Travier N, Sanchez MJ, Amiano P, Huerta JM, Ardanaz E, Rasmuson T, Hallmans G, Almquist M, Manjer J, Tsilidis KK, Allen NE, Khaw KT, Wareham N, Byrnes G, Romieu I, Riboli E, Franceschi S, 2012, Body size and risk of differentiated thyroid carcino mas: findings from the EPIC study. Int J Cancer 131(6):E1004–E1014 Oberman B, Khaku A, Camacho F, Goldenberg D, 2015, Relationship between obesity, diabetes and the risk of thyroid cancer. Am J Otolaryngol 36(4):535–541 Malaguarnera R, Vella V, Nicolosi ML, Belfiore A, 2017, Insulin resistance: any role in the changing epidemiology of thyroid Cancer? Front Endocrinol 8:314 Zane M, Parello C, Pennelli G et al, 2016, Estrogen and thyroid cancer is a stem affair: A preliminary study. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 2016: 85 Dong W, Zhang H, Li J et al, 2013, Estrogen induces metastatic potential of papillary thyroid cancer cells through estrogen receptor alpha and beta. Int J Endocrinol 2013:941568 Ahmed RA, Aboelnaga EM, 2015, Thyroid cancer in Egypt: histopathological criteria, correlation with survival and oestrogen receptor protein expression. Pathol Oncol Res 21:793–802 Caini S, Gibelli B, Palli D, Saieva C, Ruscica M, Gandini S, 2015, Menstrual and reproductive history and use of exogenous sex hormones and risk of thyroid cancer among women: a meta-analysis of prospective studies. Cancer Causes Control 26:511–518 Xing M, Alzahrani AS, Carson KA, Viola D, Elisei R, Bendlova B, Yip L, Mian C, Vianello F, Tuttle RM, Robenshtok E, Fagin JA, Puxeddu E, Fugazzola L, Czarniecka A, Jarzab B, O’Neill CJ, Sywak MS, Lam AK, Riesco-Eizaguirre G, Santisteban P, Nakayama H, Tufano RP, Pai SI, Zeiger MA, Westra WH, Clark DP, Clifton-Bligh R, Sidransky D, Ladenson PW, Sykorova V, 2013, Association between BRAF V600E mutation and mortality in patients with papillary thyroid cancer. JAMA 309:1493–1501 Siroy AE, Boland GM, Milton DR, Roszik J, Frankian S, Malke J, Haydu L, Prieto VG, Tetzlaff M, Ivan D,WangWL, Torres-Cabala C, Curry J, Roy-Chowdhuri S, Broaddus R, Rashid A, Stewart J, Gershenwald JE, Amaria RN, Patel SP, Papadopoulos NE, Bedikian A, Hwu WJ, Hwu P, Diab A, Woodman SE, Aldape KD, Luthra R, Patel KP, Shaw KR, Mills GB, Mendelsohn J, Meric-Bernstam F, Kim KB, Routbort MJ, Lazar AJ, Davies MA, 2015, Beyond BRAF(V600): clinical mutation panel testing by next-generation sequencing in advanced melanoma. J Invest Dermatol 135(2):508–515 Dvorak K, Higgins A, Palting J et al, 2017, Immunohistochemistry with Anti-BRAF V600E, VE1, mouse monoclonal antibody is a sensitive method for detection of the BRAF V600E mutation in colon cancer: evaluation of 120 Cases with and without KRAS mutation and literature review. Pathol Oncol Res 2017:1–11 Ghossein RA, Katabi N, Fagin JA, 2013, Immunohistochemical detection of mutated BRAF V600E supports the clonal origin of BRAF-induced thyroid cancers along the spectrum of disease progression. J Clin Endocrinol Metab 98(8):E1414–E1421 Dovrat TO, Sokol E, Frampton G et al, 2018, Unusually long-term responses to vemurafenib in BRAF V600E mutated colon and thyroid cancers followed by the development of rare RAS activating mutations. Cancer Biol Ther 5:1–4 Antonello ZA, Hsu N, Bhasin M, Roti G, Joshi M, van Hummelen P, Ye E, Lo AS, Karumanchi SA, Bryke CR, Nucera C, 2017, Vemurafenib-resistance via de novo RBM genes mutations and chromosome 5 aberrations is overcome by combined therapy with palbociclib in thyroid carcinoma with BRAFV600E. Oncotarget 8(49):84743–84760 Nakamura M, Runko AP, Sagerstrom CG, 2004, A novel subfamily of zinc finger genes involved in embryonic development. J Cell Biochem 93:887–895 Brown JD, Dutta S, Bharti K, Bonner RF, Munson PJ, Dawid IB, Akhtar AL, Onojafe IF, Alur RP, Gross JM, Hejtmancik JF, Jiao X, Chan WY, Brooks BP, 2009, Expression profiling during ocular development identifies 2 Nlz genes with a critical role in optic fissure closure. Proc Natl Acad Sci U S A 106:1462–1467 Slorach EM, Chou J, Werb Z, 2011, Zeppo1 is a novel metastasis promoter that represses E-cadherin expression and regulates p120-catenin isoform expression and localization. Genes Dev 25:471–484 Sircoulomb F, Nicolas N, Ferrari A, Finetti P, Bekhouche I, Rousselet E, Lonigro A, Adelaide J, Baudelet E, Esteyries S, Wicinski J, Audebert S, Charafe-Jauffret E, Jacquemier J, Lopez M, Borg JP, Sotiriou C, Popovici C, Bertucci F, Birnbaum D, Chaffanet M, Ginestier C, 2011, ZNF703 gene amplification at 8p12 specifies luminal B breast cancer. EMBO Mol Med 3(3):153–166 Shahi P, Slorach EM,Wang CY, Chou J, Lu A, Ruderisch A,Werb Z, 2015, The transcriptional repressor ZNF503/Zeppo2 promotes mammary epithelial cell proliferation and enhances cell invasion. J Biol Chem 290(6):3803–3813 Yang G, Ma F, Zhong M et al, 2014, ZNF703 acts as an oncogene that promotes progression in gastric cancer. Oncol Rep 31:1877–1882 Li K, Wang J, Han J et al, 2016, Overexpression of ZNF703 facilitates tumorigenesis and predicts unfavorable prognosis in patients with cholangiocarcinoma. Oncotarget 7:76108–76117 HaoW, Xubin D, Jinshan Z et al, 2017, Elevated expression of zinc finger protein 703 promotes cell proliferation and metastasis through PI3K/AKT/GSK-3β Signalling in Oral squamous cell carcinoma. Cell Physiol Biochem 44:920–934 Morrison JA, Pike LA, Lund G, Zhou Q, Kessler BE, Bauerle KT, Sams SB, Haugen BR, Schweppe RE, 2015, Characterization of thyroid Cancer cell lines in murine Orthotopic and Intracardiac metastasis models. Hormones & Cancer 6(2–3):87–99 Pereira-Castro I, Costa AMS, Oliveira MJ, Barbosa I, Rocha AS, Azevedo L, da Costa LT, 2013, Characterization of human NLZ1/ ZNF703 identifies conserved domains essential for proper subcellular localization and transcriptional repression. J Cell Biochem 114:120–133 Dorfman R, Glazer L, Weihe U, Wernet MF, Shilo BZ, 2002, Elbow and Noc define a family of zinc finger proteins controlling morphogenesis of specific tracheal branches. Development 129: 3585–3596 Weihe U, Dorfman R, Wernet MF, Cohen SM, Milan M, 2004, Proximodistal subdivision of Drosophila legs and wings: the elbow-no ocelli gene complex. Development 131:767–774 Zhao XJ, Yang Y, Fitch DHA, Herman MA, 2002, TLP-1 is an asymmetric cell fate determinant that responds to Wnt signals and controls male tail tip morphogenesis in C-elegans. Development 129:1497–1508 Brown JD, Dutta S, Bharti K, Bonner RF, Munson PJ, Dawid IB, Akhtar AL, Onojafe IF, Alur RP, Gross JM, Hejtmancik JF, Jiao X, Chan WY, Brooks BP, 2009, Expression profiling during ocular development identifies 2 Nlz genes with acritical role in optic fissure closure. Proc Natl Acad Sci U S A 106:1462–1467 Ji SJ, Periz G, Sockanathan S, 2009, Nolz1 is induced by retinoid signals and controls motoneuron subtype identity through distinct repressor activities. Development 136:231–240 Andreazzoli M, Broccoli V, Dawid IB, 2001, Cloning and expression of noz1, a zebrafish zinc finger gene related to Drosophila nocA. Mech Dev 104:117–120 Baykara O, Dalay N, Kaynak K, Buyru N, 2016, ZNF703 overexpression may act as an oncogene in non-small cell lung cancer. Cancer Med 5:2873–2878 Shackney SE, Chowdhury SA, Schwartz R, 2014, A novel subset of human tumors that simultaneously overexpress multiple E2Fresponsive genes found in breast, ovarian, and prostate cancers. Cancer Inform 13:89–100 Chu I, Sun J, Arnaout A, Kahn H, Hanna W, Narod S, Sun P, Tan CK, Hengst L, Slingerland J, 2007, p27 phosphorylation by Src regulates inhibition of cyclin E-CDK2. Cell 128:281–294 Grimmler M, Wang Y, Mund T, Cilensek Z, Keidel EM, Waddell MB, Jakel H, Kullmann M, Kriwacki RW, Hengst L, 2007, CDKinhibitory activity and stability of p27Kip1 are directly regulated by oncogenic tyrosine kinases. Cell 128:269–280 Lipsa RS, Sanjib S, 2016, Mechanism of p27 unfolding for CDK2 reactivation. Sci Rep 6:1 Hung JJ, Hsueh CT, Chen KH, Hsu WH, Wu YC, 2012, Clinical significance of E2F1 protein expression in non-small cell lung cancer. Exp Hematol Oncol 1(1):18 Zhang D, Wang Y, Liang Y, Zhang M, Wei J, Zheng X, Li F, Meng Y, Zhu NW, Li J, Wu XR, Huang C, 2014, Loss of p27 upregulates MnSOD in a STAT3-dependent manner, disrupts intracellular redox activity and enhances cell migration. J Cell Sci 127(13):2920–2933 Jeannot P, Nowosad A, Perchey RT, Callot C, Bennana E, Katsube T, Mayeux P, Guillonneau F, Manenti S, Besson A, 2017, p27Kip1 promotes invadopodia turnover and invasion through the regulation of the PAK1/Cortactin pathway. eLife 6:e22207 Bondar T, Kalinina A, Khair L, Kopanja D, Nag A, Bagchi S, Raychaudhuri P, 2006, Cul4A and DDB1 associate with Skp2 to target p27Kip1 for proteolysis involving the COP9 signalosome. Mol Cell Biol 26:2531–2539 Huang J, Zhang F, Jiang L, Hu G, Sun W, Zhang C, Ding X, 2017, Inhibition of SKP2 sensitizes bromocriptine-induced apoptosis in human Prolactinoma cells. Cancer Res Treat 49(2):358–373 Baldini E, Arlot-Bonnemains Y, Sorrenti S et al, 2011, Aurora kinases are expressed in medullary thyroid carcinoma, MTC, and their inhibition suppresses in vitro growth and tumorigenicity of the MTC derived cell line TT. BMC Cancer 11:411 Li Z, Guo Y, Jiang H, Zhang T, Jin C, Young CYF, Yuan H, 2014, Differential regulation of MMPs by E2F1, Sp1 and NF-kappa B controls the small cell lung cancer invasive phenotype. BMC Cancer 14:276 Johnson JL, Pillai S, Pernazza D, Sebti SM, Lawrence NJ, Chellappan SP, 2012, Regulation of matrix metalloproteinase genes by E2F transcription factors: Rb-Raf-1 interaction as a novel target for metastatic disease. Cancer Res 72:516–526 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 355 EP 364 DI 10.1007/s12253-018-0494-5 PG 10 ER PT J AU Zhang, G Xu, Y Zhou, H AF Zhang, Geng Xu, Yi Zhou, Huifang TI The Infiltration of ICOS+ Cells in Nasopharyngeal Carcinoma is Beneficial for Improved Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Nasopharyngeal carcinoma; Survival; Inducible T cell co-stimulator; IFNγ; Prognosis ID Nasopharyngeal carcinoma; Survival; Inducible T cell co-stimulator; IFNγ; Prognosis AB Nasopharyngeal carcinoma (NPC) is a highly malignant tumor, associated with poor patient prognoses, and high rates of morbidity and mortality. Currently, immune checkpoint therapy has brought new treatment strategy for NPC. The inducible T cell co-stimulator (ICOS) belongs to the B7-CD28 immunoglobulin superfamily, which is currently the subject of intense study due to great successes gained in treatment of different malignancies by disrupting their family members. However, the role of ICOS played in NPC remains poorly understood. Immunohistochemistry (IHC) was stained with the ICOS specific antibody and ICOS expression is decreased in patients with either lymphatic or distant metastasis and inversely associated with TNM stage of NPC patients. Importantly, high ICOS expression is significantly correlated with overall survival (OS) of NPC patients (N = 185, p < 0.001), and ICOS expression is also proved to be an independent prognostic factor by multivariate analysis. Surgical excised fresh NPC specimens (N = 185) were homogenized to analyze the specific cytokine expression by ELISA assay. ICOS expression level is associated with increased cytotoxic T lymphocyte number and high interferon IFNγ expression, the characteristics of Th1 cells. In addition, the correlation between the percentage of ICOS+ T cells in tumor tissue and survival was detected. Conclusively, expression of ICOS is associated with improved survival in NPC and percentage of ICOS+ cells acting as Th1 cells in primary tumor tissue may be a clinical biomarker for good prognosis of NPC patients. C1 [Zhang, Geng] Tianjin Medical University General Hospital, Department of Otorhinolaryngology, No. 154 Anshan Road, Heping District, 300052 Tianjin, China. [Xu, Yi] Tianjin Medical University General Hospital, Department of Otorhinolaryngology, No. 154 Anshan Road, Heping District, 300052 Tianjin, China. [Zhou, Huifang] Tianjin Medical University General Hospital, Department of Otorhinolaryngology, No. 154 Anshan Road, Heping District, 300052 Tianjin, China. RP Zhou, H (reprint author), Tianjin Medical University General Hospital, Department of Otorhinolaryngology, 300052 Tianjin, China. EM huifzhou667@163.com CR Lee AW, Ma BB, Ng WT, Chan AT, 2015, Management of nasopharyngeal carcinoma: current practice and future perspective. J Clin Oncol 33(29):3356–3364. , DOI 10.1200/JCO.2015. 60.9347 Mao YP, Xie FY, Liu LZ, Sun Y, Li L, Tang LL, Liao XB, Xu HY, Chen L, Lai SZ, Lin AH, LiuMZ,Ma J, 2009, Re-evaluation of 6th edition of AJCC staging system for nasopharyngeal carcinoma and proposed improvement based on magnetic resonance imaging. Int J Radiat Oncol Biol Phys 73(5):1326–1334. , DOI 10.1016/ j.ijrobp.2008.07.062 Zheng L, Cao C, Cheng G, Hu Q, Chen X, 2017, Cytomembranic PD-L1 expression in locoregionally advanced nasopharyngeal carcinoma. Onco Targets Ther 10:5483–5487. , DOI 10.2147/ OTT.S152007 Hua YJ,Han F, Lu LX, MaiHQ,Guo X, HongMH, Lu TX, Zhao C, 2012, Long-term treatment outcome of recurrent nasopharyngeal carcinoma treated with salvage intensity modulated radiotherapy. Eur J Cancer 48(18):3422–3428. , DOI 10.1016/j.ejca. 2012.06.016 Kong L, Wang L, Shen C, Hu C, Wang L, Lu JJ, 2016, Salvage Intensity-Modulated Radiation Therapy, IMRT, for locally recurrent nasopharyngeal cancer after definitive IMRT: a novel scenario of the modern era. Sci Rep 6:32883. , DOI 10.1038/ srep32883 Chen C, FeeW, Chen J, Chan C, Khong B, HaraW, Goffinet D, Li D, Le QT, 2014, Salvage treatment for locally recurrent nasopharyngeal carcinoma, NPC). Am J Clin Oncol 37(4):327–331. https:// doi.org/10.1097/COC.0b013e318277d804 Karam I, Huang SH, McNiven A, Su J, Xu W, Waldron J, Bayley AJ, Kim J, Cho J, Ringash J, Hope A, Chen E, Chan B, Goldstein D, O'Sullivan B, Giuliani ME, 2016, Outcomes after reirradiation for recurrent nasopharyngeal carcinoma: north American experience. Head Neck 38(Suppl 1):E1102–E1109. , DOI 10. 1002/hed.24166 Zhang Y, Luo Y, Qin SL, Mu YF, Qi Y, Yu MH, Zhong M, 2016, The clinical impact of ICOS signal in colorectal cancer patients. Oncoimmunology 5(5):e1141857. , DOI 10.1080/ 2162402X.2016.1141857 Sharpe AH, Freeman GJ, 2002, The B7-CD28 superfamily. Nat Rev Immunol 2(2):116–126. , DOI 10.1038/nri727 Ceeraz S, Nowak EC, Noelle RJ, 2013, B7 family checkpoint regulators in immune regulation and disease. Trends Immunol 34(11): 556–563. , DOI 10.1016/j.it.2013.07.003 Swallow MM, Wallin JJ, Sha WC, 1999, B7h, a novel costimulatory homolog of B7.1 and B7.2, is induced by TNFalpha. Immunity 11(4):423–432 Faget J, Bendriss-Vermare N, Gobert M, Durand I, Olive D, Biota C, Bachelot T, Treilleux I, Goddard-Leon S, Lavergne E, Chabaud S, Blay JY, Caux C,Menetrier-Caux C, 2012, ICOS-ligand expression on plasmacytoid dendritic cells supports breast cancer progression by promoting the accumulation of immunosuppressive CD4+ T cells. Cancer Res 72(23):6130–6141. , DOI 10.1158/ 0008-5472.CAN-12-2409 Faget J, Sisirak V, Blay JY, Caux C, Bendriss-Vermare N, Menetrier-Caux C, 2013, ICOS is associated with poor prognosis in breast cancer as it promotes the amplification of immunosuppressive CD4+ T cells by plasmacytoid dendritic cells. Oncoimmunology 2(3):e23185. , DOI 10.4161/onci. 23185 Hutloff A, Dittrich AM, Beier KC, Eljaschewitsch B, Kraft R, Anagnostopoulos I, Kroczek RA, 1999, ICOS is an inducible Tcell co-stimulator structurally and functionally related to CD28. Nature 397(6716):263–266. , DOI 10.1038/16717 McAdam AJ, Chang TT, Lumelsky AE, Greenfield EA, Boussiotis VA, Duke-Cohan JS, Chernova T, Malenkovich N, Jabs C, Kuchroo VK, Ling V, Collins M, Sharpe AH, Freeman GJ, 2000, Mouse inducible costimulatory molecule, ICOS, expression is enhanced by CD28 costimulation and regulates differentiation of CD4+ T cells. J Immunol 165(9):5035–5040 Akbari O, Stock P, Meyer EH, Freeman GJ, Sharpe AH, Umetsu DT, DeKruyff RH, 2008, ICOS/ICOSL interaction is required for CD4+ invariant NKT cell function and homeostatic survival. J Immunol 180(8):5448–5456 Fu T, He Q, Sharma P, 2011, The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res 71(16):5445–5454. , DOI 10.1158/ 0008-5472.CAN-11-1138 CoyleAJ, Lehar S, Lloyd C, Tian J, Delaney T, Manning S, Nguyen T, Burwell T, Schneider H, Gonzalo JA, Gosselin M, Owen LR, Rudd CE, Gutierrez-Ramos JC, 2000, The CD28-related molecule ICOS is required for effective T cell-dependent immune responses. Immunity 13(1):95–105 Yoshinaga SK, Whoriskey JS, Khare SD, Sarmiento U, Guo J, Horan T, Shih G, Zhang M, Coccia MA, Kohno T, Tafuri-Bladt A, Brankow D, Campbell P, Chang D, Chiu L, Dai T, Duncan G, Elliott GS, Hui A, McCabe SM, Scully S, Shahinian A, Shaklee CL, Van G, Mak TW, Senaldi G, 1999, T-cell co-stimulation through B7RP-1 and ICOS. Nature 402(6763):827–832. https:// doi.org/10.1038/45582 Zhang Y, Lv D, Kim HJ, Kurt RA, Bu W, Li Y, Ma X, 2013, A novel role of hematopoietic CCL5 in promoting triple-negative mammary tumor progression by regulating generation of myeloidderived suppressor cells. Cell Res 23(3):394–408. , DOI 10.1038/cr.2012.178 Burris HA, Callahan MK, Tolcher AW, Kummar S, Falchook GS, Pachynski RK, Tykodi SS, Gibney GT, Seiwert TY, Gainor JF, LoRusso P, Hilbert J, Apgar JF, Hua F, Burke JM, Lazaro M, Clancy M, Ding B, Trehu EG, Yap TA, 2017, Phase 1 safety of ICOS agonist antibody JTX-2011 alone and with nivolumab, nivo, in advanced solid tumors; predicted vs observed pharmacokinetics, PK, in ICONIC. J Clin Oncol 35(15_suppl):3033–3033. https:// doi.org/10.1200/JCO.2017.35.15_suppl.3033 Angevin E, Bauer TM, Ellis CE, Gan H, Hall R, Hansen A, Hoos A, Jewell RC, Katz J, Martin-Liberal J, Maio M, Mayes PA, Mazumdar J, Millward M, Rischin D, Schellens JH, Yadavilli S, Zhou H, 2017, Abstract CT039: INDUCE- 1: a phase I open-label study of GSK3359609, an ICOS agonist antibody, administered alone and in combination with pembrolizumab in patients with selected, advanced solid tumors. Cancer Res 77(13 Supplement):CT039–CT039. , DOI 10.1158/1538-7445.Am2017-ct039 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 365 EP 370 DI 10.1007/s12253-018-0509-2 PG 6 ER PT J AU Chopra, S Goel, S Thakur, B Bhatia, A AF Chopra, Sucheta Goel, Sumit Thakur, Banita Bhatia, Alka TI Do Different Stemness Markers Identify Different Pools of Cancer Stem Cells in Malignancies: A Study on ER+ and ER-Breast Cancer Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Stemness; Breast cancer; Cancer stem cells; Estrogen receptor ID Stemness; Breast cancer; Cancer stem cells; Estrogen receptor AB In view of popularity of cancer stem cell (CSC) model all events in evolution of cancer are being explained in that context. Breast cancer is first solid tumor in which CSCs were identified. We aimed to compare stemness profile of two major subtypes [Estrogen receptor positive (ER+) and negative (ER−)] breast cancer using different sets of markers. Expression of CD44/CD24, CK/Vimentin, E-Cadherin/Fibronectin and percentage of side population (SP) was studied in ER+ (T47D) and ER−(MDA-MB-231) cell lines by flow cytometry. Breast CSCs (BCSCs) were sorted using CD44+/CD24−/low expression and SP analysis and cultured. BCSCs were then compared with Non-CSCs (NCSCs) for response to drugs (Paclitaxel and Cisplatin), Ki67 and ER expression. Results showed higher expression of stemness markers (CD44+/CD24−/low, CK+/Vimentin+ and ECadherin−/FibrinectinF+) in MDA-MB-231 cells. Percentage SP representing BCSCs was found to be significantly more in later (3.20 ± 0.002 cf. T47D 1.25%± 0.0007). BCSCs were found to be more resistant to drugs as compared to NCSCs in both cell lines. ER expression was weak in BCSCs sorted from T47D as compared to NCSCs. Ki67 was expressed in both BCSCs and NCSCs. Differences in expression of stemness markers help to explain aggressive behavior, higher recurrence rate and metastatic potential of MDA-MB-231 cells. However, no correlation amongst different markers used suggests that they may be identifying varied populations of cells in tumor hierarchy. A weak ER expression in BCSCs may be strategy used by BCSCs to escape effect of hormone therapy in ER+ breast cancers. C1 [Chopra, Sucheta] Postgraduate Institute of Medical Education and Research, Department of Experimental Medicine & Biotechnology, 160012 Chandigarh, India. [Goel, Sumit] Postgraduate Institute of Medical Education and Research, Department of Experimental Medicine & Biotechnology, 160012 Chandigarh, India. [Thakur, Banita] Postgraduate Institute of Medical Education and Research, Department of Experimental Medicine & Biotechnology, 160012 Chandigarh, India. [Bhatia, Alka] Postgraduate Institute of Medical Education and Research, Department of Experimental Medicine & Biotechnology, 160012 Chandigarh, India. RP Bhatia, A (reprint author), Postgraduate Institute of Medical Education and Research, Department of Experimental Medicine & Biotechnology, 160012 Chandigarh, India. EM alkabhatia@ymail.com CR Gay L, Baker AM, Graham TA: Tumour Cell Heterogeneity. F1000Res 5, 2016 Gasch C, Ffrench B, O'Leary JJ et al, 2017, Catching moving targets: cancer stem cell hierarchies, therapy-resistance & considerations for clinical intervention. Mol Cancer 16:43 Badve S, Nakshatri H, 2012, Breast-cancer stem cells-beyond semantics. Lancet Oncol 13:e43–e48 Bozorgi A, Khazaei M, KhazaeiMR, 2015, New findings on breast Cancer stem cells: a review. J Breast Cancer 18:303–312 Sledge GW,Mamounas EP, HortobagyiGN, Burstein HJ, Goodwin PJ, Wolff AC, 2014, Past, present, and future challenges in breast cancer treatment. J Clin Oncol 32:1979–1986 Dai M, Zhang C, Ali A, et al: CDK4 regulates cancer stemness and is a novel therapeutic target for triple-negative breast cancer. Scientific Reports 6, Article number: 35383, 2016 Chekhun S, Bezdenezhnykh N, Shvets J, Lukianova N, 2013, Expression of biomarkers related to cell adhesion, metastasis and invasion of breast cancer cell lines of different molecular subtype. Exp Oncol 35:174–179 Prasetyanti PR, Medema JP, 2017, Intra-tumor heterogeneity from a cancer stem cell perspective. Mol Cancer 16:41 Visvader JE, Lindeman GJ, 2008, Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 8:755–768 Leung EL, Fiscus RR, Tung JW et al, 2010, Non-small cell lung cancer cells expressing CD44 are enriched for stem cell-like properties. PLoS One 5:e14062 Godar S, Ince TA, BellGW, Feldser D, Donaher JL, Bergh J, Liu A, Miu K,Watnick RS, Reinhardt F, McAllister SS, Jacks T,Weinberg RA, 2008, Growth-inhibitory and tumor- suppressive functions of p53 depend on its repression of CD44 expression. Cell 134:62–73 Zheng J, Li Y, Yang J et al, 2011, NDRG2 inhibits hepatocellular carcinoma adhesion, migration and invasion by regulating CD24 expression. BMC Cancer 11(251):1–9 Cichy J, Pure E, 2003, The liberation of CD44. J Cell Biol 161: 839–843 Akrap N, Andersson D, Bom E, Gregersson P, Stahlberg A, Landberg G, 2016, Identification of distinct breast Cancer stem cell populations based on single-cell analyses of functionally enriched stem and progenitor pools. Stem Cell Reports 6:121–136 Horimoto Y, Arakawa A, Sasahara N, Tanabe M, Sai S, Himuro T, Saito M, 2016, Combination of Cancer stem cell markers CD44 and CD24 is superior to ALDH1 as a prognostic Indicator in breast Cancer patients with distant metastases. PLoS One 11:e0165253 Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA, 2008, The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133:704–715 Morel AP, Lievre M, Thomas C et al, 2008, Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS One 3:e2888 Hwang-Verslues WW, Kuo WH, Chang PH, Pan CC, Wang HH, Tsai ST, JengYM, Shew JY, Kung JT, Chen CH, Lee EYHP, Chang KJ, LeeWH, 2009)Multiple lineages of human breast cancer stem/ progenitor cells identified by profilingwith stem cellmarkers. PLoS One 4:e8377 Oh TG, Wang SC, Acharya BR, Goode JM, Graham JD, Clarke CL, Yap AS, Muscat GEO, 2016, The nuclear receptor, RORgamma. Regulates Pathways Necessary for Breast Cancer Metastasis EBioMedicine 6:59–72 Walia V, Yu Y, Cao D, Sun M, McLean JR, Hollier BG, Cheng J, Mani SA, Rao K, Premkumar L, Elble RC, 2012, Loss of breast epithelial marker hCLCA2 promotes epithelial-to-mesenchymal transition and indicates higher risk of metastasis. Oncogene 31: 2237–2246 Armstrong AJ, Marengo MS, Oltean S, Kemeny G, Bitting RL, Turnbull JD, Herold CI, Marcom PK, George DJ, Garcia-Blanco MA, 2011, Circulating tumor cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers. Mol Cancer Res 9:997–1007 Polioudaki H, Agelaki S, Chiotaki R, Politaki E, Mavroudis D, Matikas A, Georgoulias V, Theodoropoulos PA, 2015, Variable expression levels of keratin and vimentin reveal differential EMT status of circulating tumor cells and correlation with clinical characteristics and outcome of patients with metastatic breast cancer. BMC Cancer 15:399 Christofori G, Semb H, 1999, The role of the cell-adhesion molecule E-cadherin as a tumour-suppressor gene. Trends Biochem Sci 24:73–76 Takeichi M, 1993, Cadherins in cancer: implications for invasion and metastasis. Curr Opin Cell Biol 5:806–811 Geiger B, Yamada KM, 2011, Molecular architecture and function of matrix adhesions. Cold Spring Harb Perspect Biol 3 Sun X, Fa P, Cui Z, Xia Y, Sun L, Li Z, Tang A, Gui Y, Cai Z, 2014, The EDA-containing cellular fibronectin induces epithelialmesenchymal transition in lung cancer cells through integrin alpha9beta1-mediated activation of PI3-K/AKT and Erk1/2. Carcinogenesis 35:184–191 GoodellMA: Stem cell identification and sorting using the Hoechst 33342 side population, SP). Curr Protoc Cytom Chapter 9:Unit9 18, 2005 Britton KM, Eyre R, Harvey IJ, Stemke-Hale K, Browell D, Lennard TWJ, Meeson AP, 2012, Breast cancer, side population cells and ABCG2 expression. Cancer Lett 323:97–105 Nakanishi T, Chumsri S, Khakpour N, Brodie AH, Leyland-Jones B, Hamburger AW, Ross DD, Burger AM, 2010, Side-population cells in luminal-type breast cancer have tumour-initiating cell properties, and are regulated by HER2 expression and signalling. Br J Cancer 102:815–826 Jin C, Zou T, Li J et al, 2015, Side population cell level in human breast cancer and factors related to disease-free survival. Asian Pac J Cancer Prev 16:991–996 Liu Y, Nenutil R, Appleyard MV, Murray K, BoylanM, Thompson AM, Coates PJ, 2014, Lack of correlation of stem cell markers in breast cancer stem cells. Br J Cancer 110(8):2063–2071 Dontu G, Abdallah WM, Foley JM, Jackson KW, Clarke MF, Kawamura MJ, Wicha MS, 2003, In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev 17:1253–1270 Ponti D, Costa A, Zaffaroni N, Pratesi G, Petrangolini G, Coradini D, Pilotti S, PierottiMA, Daidone MG(2005, Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res 65:5506–5511 Manuel Iglesias J, Beloqui I, Garcia-Garcia F, Leis O, Vazquez- Martin A, Eguiara A, Cufi S, Pavon A, Menendez JA, Dopazo J, Martin AG, 2013, Mammosphere formation in breast carcinoma cell lines depends upon expression of E-cadherin. PLoS One 8: e77281 Shafee N, Smith CR, Wei S, Kim Y, Mills GB, Hortobagyi GN, Stanbridge EJ, Lee EYHP, 2008, Cancer stem cells contribute to cisplatin resistance in Brca1/p53-mediated mouse mammary tumors. Cancer Res 68:3243–3250 Dean M, Fojo T, Bates S, 2005, Tumour stem cells and drug resistance. Nat Rev Cancer 5:275–284 Phillips TM, McBride WH, Pajonk F, 2006, The response of CD24(−/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancer Inst 98:1777–1785 Cidado J, Wong HY, Rosen DM, Cimino-Mathews A, Garay JP, Fessler AG, Rasheed ZA, Hicks J, Cochran RL, Croessmann S, Zabransky DJ, Mohseni M, Beaver JA, Chu D, Cravero K, Christenson ES, Medford A, Mattox A, de Marzo AM, Argani P, Chawla A, Hurley PJ, Lauring J, Park BH, 2016, Ki-67 is required for maintenance of cancer stem cells but not cell proliferation. Oncotarget 7:6281–6293 Guttilla IK, Phoenix KN, Hong X, Tirnauer JS, Claffey KP, White BA, 2012, Prolonged mammosphere culture of MCF-7 cells induces an EMT and repression of the estrogen receptor by microRNAs. Breast Cancer Res Treat 132:75–85 Voutsadakis IA, 2016, Epithelial-Mesenchymal transition, EMT, and regulation of EMT factors by steroid nuclear receptors in breast Cancer: a review and in Silico investigation. J Clin Med 5 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 371 EP 378 DI 10.1007/s12253-018-0503-8 PG 8 ER PT J AU Benesova, L Halkova, T Bunganic, B Belsanova, B Zavoral, M Traboulsi, E Minarik, M AF Benesova, Lucie Halkova, Tereza Bunganic, Bohus Belsanova, Barbora Zavoral, Miroslav Traboulsi, Eva Minarik, Marek TI Comparison of Native Aspirates and Cytological Smears Obtained by EUS-Guided Biopsies for Effective DNA/RNA Marker Testing in Pancreatic Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EUS-FNA; Pancreatic cancer; KRAS; miR-21 ID EUS-FNA; Pancreatic cancer; KRAS; miR-21 AB We compare two types of pancreatic carcinoma samples obtained by EUS-guided fine needle biopsy (EUS-FNB) in terms of the success rates and clinical validity of analysis of two most commonly investigated DNA/RNA pancreatic cancer markers, KRAS mutations and miR-21 expression. 118 patients with pancreatic ductal adenocarcinoma underwent EUS-FNB. The collected sample was divided, one part was stored in a stabilizing solution as native aspirate (EUS-FNA) and second part was processed into the cytological smear (EUS-FNC). DNA/RNA extraction was followed by analysis of KRAS mutations and miR-21 expression. For both sample types, the yields of DNA/RNA extraction and success rates of KRAS mutation and miRNA expression were evaluated. Finally, the resulting KRAS mutation frequency and miR-21 prognostic role were compared to literature data from tissue resections. The overall amount of isolated DNA/RNA from EUS-FNC was lower compared to the EUS-FNA, average yield 10 ng vs 147 ng for DNA and average yield 164 vs. 642 ng for RNA, but the success rates for KRAS and miR-21 analysis was 100% for both sample types. The KRAS-mutant detection frequency in EUS-FNC was 12% higher than in EUS-FNA (90 vs 78%). The prognostic role of miR-21 was confirmed in EUS-FNC (p = 0.02), but did not reach statistical significance in EUS-FNA (p = 0.06). Although both types of EUS-FNB samples are suitable for DNA/RNA extraction and subsequent DNA mutation and miRNA expression analysis, reliable results with clinical validity were only obtained for EUS-FNC. C1 [Benesova, Lucie] Genomac Research Institute, Centre for Applied Genomics of Solid Tumors (CEGES), 161 00 Prague, Czech Republic. [Halkova, Tereza] Genomac Research Institute, Centre for Applied Genomics of Solid Tumors (CEGES), 161 00 Prague, Czech Republic. [Bunganic, Bohus] Charles University in Prague and General University Hospital in Prague, First Faculty of Medicine, 1st Department of Medicine—Department of Haematology, 169 02 Prague, Czech Republic. [Belsanova, Barbora] Genomac Research Institute, Centre for Applied Genomics of Solid Tumors (CEGES), 161 00 Prague, Czech Republic. [Zavoral, Miroslav] Charles University in Prague and General University Hospital in Prague, First Faculty of Medicine, 1st Department of Medicine—Department of Haematology, 169 02 Prague, Czech Republic. [Traboulsi, Eva] Military University Hospital, Pathology department, 169 02 Prague, Czech Republic. [Minarik, Marek] Genomac Research Institute, Centre for Applied Genomics of Solid Tumors (CEGES), 161 00 Prague, Czech Republic. RP Minarik, M (reprint author), Genomac Research Institute, Centre for Applied Genomics of Solid Tumors (CEGES), 161 00 Prague, Czech Republic. EM mminarik@email.com CR Ballehaninna UK, Chamberlain RS, 2012, The clinical utility of serum CA 19-9 in the diagnosis, prognosis and management of pancreatic adenocarcinoma: an evidence based appraisal. J Gastrointest Oncol 3:105–119 Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL et al, 2012, Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491:399–405 Salek C, Benesova L, Zavoral M, Nosek V, Kasperova L, Ryska M, Strnad R, Traboulsi E, Minarik M, 2007, Evaluation of clinical relevance of examining K-ras, p16 and p53 mutations along with allelic losses at 9p and 18q in EUS-guided fine needle aspiration samples of patients with chronic pancreatitis and pancreatic cancer. World J Gastroenterol 13:3714–3720 Ginesta MM, Mora J, Mayor R, Farre A, Peinado MA, Busquets J et al, 2013, Genetic and epigenetic markers in the evaluation of pancreatic masses. J Clin Pathol 66:192–197 Thompson H, 2013, US National Cancer Institute's new Ras project targets an old foe. Nat Med 19:949–950 Eser S, Schnieke A, Schneider G, Saur D, 2014, Oncogenic KRAS signalling in,pancreatic cancer. Br J Cancer 111:817–822 Minarik M, Halkova T, Belsanova B, Bunganic B, Zavoral M, Benesova L, 2016, Detection of specific KRAS mutation type, Gly12Asp, GGT > GAT), in EUS-guided fine needle aspiration cytology, EUS-FNAC, identifies pancreatic cancer patients with poor prognosis. AACR 107th Annual Meeting on Bioinformatics and Systems Biology. Cancer Res 76(Suppl 14):4943 Halkova T, Cuperkova R, Minarik M, Benesova L, 2015, MicroRNAs in pancreatic cancer: involvement in carcinogenesis and potential use for diagnosis and prognosis. Gastroenterol Res Pract 2015:892903 Dillhoff M, Liu J, Frankel W, Croce C, Bloomston M, 2008, MicroRNA-21 is overexpressed in pancreatic cancer and a potential predictor of survival. J Gastrointest Surg 12:2171–2176 du Rieu MC, Torrisani J, Selves J, Al Saati T, Souque A, Dufresne M et al, 2010, MicroRNA-21 is induced early in pancreatic ductal adenocarcinoma precursor lesions. Clin Chem 56:603–612 Jamieson NB, Morran DC, Morton JP, Ali A, Dickson EJ, Carter CR, Sansom OJ, Evans TRJ, McKay CJ, Oien KA, 2012, MicroRNA molecular profiles associated with diagnosis, clinicopathologic criteria, and overall survival in patients with resectable pancreatic ductal adenocarcinoma. Clin Cancer Res 18:534–545 Yu J, Li A, Hong SM, Hruban RH, Goggins M, 2012, MicroRNA alterations of pancreatic intraepithelial neoplasias. Clin Cancer Res 18:981–992 Papaconstantinou IG, Manta A, Gazouli M, Lyberopoulou A, Lykoudis PM, Polymeneas G, Voros D, 2013, Expression of microRNAs in patients with pancreatic cancer and its prognostic significance. Pancreas 42:67–71 Vychytilova-Faltejskova P, Kiss I, Klusova S, Hlavsa J, Prochazka V, Kala Z, Mazanec J, Hausnerova J, Kren L, Hermanova M, Lenz J, Karasek P, Vyzula R, Slaby O, 2015, MiR-21, miR-34a, miR-198 and miR-217 as diagnostic and prognostic biomarkers for chronic pancreatitis and pancreatic ductal adenocarcinoma. Diagn Pathol 10:38 Frampton AE, Krell J, Jamieson NB, Gall TM, Giovannetti E, Funel N et al, 2015, MicroRNAs with prognostic significance in pancreatic ductal adenocarcinoma: a meta-analysis. Eur J Cancer 51:1389–1404 Hwang JH,Voortman J,Giovannetti E, Steinberg SM, Leon LG, Kim YT, Funel N, Park JK, Kim MA, Kang GH, Kim SW, Chiaro MD, Peters GJ, Giaccone G, 2010, Identification of microRNA-21 as a biomarker for chemoresistance and clinical outcome following adjuvant therapy in resectable pancreatic cancer. PLoS One 5:e10630 Nelsen EM, Buehler D, Soni AV, Gopal DV, 2015, Endoscopic ultrasound in the evaluation of pancreatic neoplasms-solid and cystic: a review. World J Gastrointest Endosc 7:318–327 Salek C,Minarikova P, Benesova L, Nosek V, Strnad R, Zavoral M, MinarikM(2009, Mutation status of K-ras, p53 and allelic losses at 9p and 18q are not prognostic markers in patients with pancreatic cancer. Anticancer Res 29:1803–1810 Khalid A, Dewitt J, Ohori NP, Chen JH, Fasanella KE, Sanders M, McGrath KM, NikiforovaM(2011, EUS-FNA mutational analysis in differentiating autoimmune pancreatitis and pancreatic cancer. Pancreatology 11:482–486 Tada M, Komatsu Y, Kawabe T, Sasahira N, Isayama H, Toda N, Shiratori Y, Omata M, 2002, Quantitative analysis of K-ras gene mutation in pancreatic tissue obtained by endoscopic ultrasonography-guided fine needle aspiration: clinical utility for diagnosis of pancreatic tumor. Am J Gastroenterol 97:2263–2270 PelliseM, CastellsA, Gines A, SoleM,Mora J, Castellvi-Bel S et al, 2003, Clinical usefulness of KRAS mutational analysis in the diagnosis of pancreatic adenocarcinoma by means of endosonography-guided fine-needle aspiration biopsy. Aliment Pharmacol Ther 17:1299–1307 Wang X, Gao J, Ren Y, Gu J, Du Y, Chen J et al, 2011, Detection of KRAS gene mutations in endoscopic ultrasound-guided fine-needle aspiration biopsy for improving pancreatic cancer diagnosis. Am J Gastroenterol 106:2104–2111 Takahashi K, Yamao K, Okubo K, Sawaki A,Mizuno N, Ashida R, Koshikawa T, Ueyama Y, Kasugai K, Hase S, Kakumu S, 2005, Differential diagnosis of pancreatic cancer and focal pancreatitis by using EUS-guided FNA. Gastrointest Endosc 61:76–79 Szafranska AE, Doleshal M, Edmunds HS, Gordon S, Luttges J, Munding JB, Barth RJ, Gutmann EJ, Suriawinata AA, Marc Pipas J, Tannapfel A, Korc M, Hahn SA, Labourier E, Tsongalis GJ, 2008, Analysis of microRNAs in pancreatic fine-needle aspirates can classify benign and malignant tissues. Clin Chem 54:1716–1724 Torrisani J, Bournet B, du Rieu MC, Bouisson M, Souque A, Escourrou J, Buscail L, Cordelier P, 2009, Let-7 MicroRNA transfer in pancreatic cancer-derived cells inhibits in vitro cell proliferation but fails to alter tumor progression. Hum Gene Ther 20:831–844 Brand RE, Adai AT, Centeno BA, Lee LS, Rateb G, Vignesh S, Menard C, Wiechowska–Kozlowska A, Boldys H, Hartleb M, Sanders MK, Munding JB, Tannapfel A, Hahn SA, Stefanczyk L, Tsongalis GJ, Whitcomb DC, Conwell DL, Morisset JA, Gardner TB, Gordon SR, Suriawinata AA, Lloyd MB, Wylie D, Labourier E, Andruss BF, Szafranska–Schwarzbach AE, 2014, A microRNAbased test improves endoscopic ultrasound-guided cytologic diagnosis of pancreatic cancer. Clin Gastroenterol Hepatol 12:1717–1723 Labourier E, Shifrin A, Busseniers AE, Lupo MA, ManganelliML, Andruss B, Wylie D, Beaudenon-Huibregtse S, 2015, Testing for miRNA, mRNA, and DNA on fine-needle aspiration improves the preoperative diagnosis of thyroid nodules with indeterminate cytology. J Clin Endocrinol Metab 100:2743–2750 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), method. Methods 25:402–408 Neesse A, Michl P, Frese KK, Feig C, Cook N, Jacobetz MA, Lolkema MP, Buchholz M, Olive KP, Gress TM, Tuveson DA, 2011, Stromal biology and therapy in pancreatic cancer. Gut 60: 861–868 Saito KC, Fuziwara CS, Kimura ET, 2013, Nucleic acid recovery from thyroid fine-needle cytology slides. Arq Bras Endocrinol Metabol 57:490–491 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 379 EP 385 DI 10.1007/s12253-018-0490-9 PG 7 ER PT J AU Li, K Liu, Y Xu, Sh Wang, J AF Li, Ke Liu, Ying Xu, Shuning Wang, Jufeng TI PPM1D Functions as Oncogene and is Associated with Poor Prognosis in Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Protein phosphatase Mg2+/Mn2+ dependent1D; Esophageal squamous cell carcinoma; Tumor metastasis; Prognosis ID Protein phosphatase Mg2+/Mn2+ dependent1D; Esophageal squamous cell carcinoma; Tumor metastasis; Prognosis AB Mounting evidence has demonstrated that PPM1D participates in the development and progression of a wide variety of tumors. However, its precise roles in esophageal squamous cell carcinoma (ESCC) remain under investigation. Here, UALCAN, an interactive web-portal to perform the expression analyses of PPM1D using TCGA gene expression data, and PPM1D high expression was exhibited in primary esophageal cancer. Further investigation revealed that PPM1D expression was obviously higher in ESCC tissues than in normal tissues (P < 0.01), which was consistent with the results from real-time qPCR and Western blotting in ESCC tissues and paired normal esophageal tissues. Besides, PPM1D expression was closely correlated with TNM staging, tumor differentiation and lymph node metastasis (P < 0.01), but not related to the patients’ gender and age (P > 0.05). Notably, PPM1D expression in metastatic ESCC patients was markedly higher than that in non-metastatic ESCC patients (P < 0.01), and the ESCC patients with high PPM1D expression predicted poor prognosis. Multivariate assay demonstrated that PPM1D and lymph node metastasis were considered as independent prognostic factors for the ESCC patients. These findings suggest PPM1D plays a pivotal important role in onset and progression of ESCC, and may be a new biomarker for metastasis and prognosis of the ESCC patients. C1 [Li, Ke] Affiliated Cancer Hospital of Zhengzhou University, Department of Medical Oncology, 127th Dongming Rd, 450008 Zhengzhou, Henan Province, China. [Liu, Ying] Affiliated Cancer Hospital of Zhengzhou University, Department of Medical Oncology, 127th Dongming Rd, 450008 Zhengzhou, Henan Province, China. [Xu, Shuning] Affiliated Cancer Hospital of Zhengzhou University, Department of Medical Oncology, 127th Dongming Rd, 450008 Zhengzhou, Henan Province, China. [Wang, Jufeng] Affiliated Cancer Hospital of Zhengzhou University, Department of Medical Oncology, 127th Dongming Rd, 450008 Zhengzhou, Henan Province, China. RP Wang, J (reprint author), Affiliated Cancer Hospital of Zhengzhou University, Department of Medical Oncology, 450008 Zhengzhou, China. EM wangjf1101@126.com CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127:2893–2917. , DOI 10.1002/ijc.25516 Jemal A, CenterMM, DeSantis C,Ward EM(2010)Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol Biomark Prev 19:1893–1907. , DOI 10.1158/ 1055-9965.EPI-10-0437 Wang Z, Tang L, Sun G, TangY, XieY,Wang S, Hu X, GaoW, Cox SB,Wang JS, 2006, Etiological study of esophageal squamous cell carcinoma in an endemic region: a population-based case control study in Huaian, China. BMC Cancer 6:287. , DOI 10. 1186/1471-2407-6-287 Khuroo MS, Zargar SA, Mahajan R, Banday MA, 1992, High incidence of oesophageal and gastric cancer in Kashmir in a population with special personal and dietary habits. Gut 33:11–15 Mahboubi E, Kmet J, Cook PJ, Day NE, Ghadirian P, Salmasizadeh S, 1973, Oesophageal cancer studies in the Caspian Littoral of Iran: the Caspian cancer registry. Br J Cancer 28:197–214 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65:87–108. , DOI 10.3322/caac.21262 van Hagen P, Hulshof MC, van Lanschot JJ, Steyerberg EW, van Berge Henegouwen MI, Wijnhoven BP, Richel DJ, Nieuwenhuijzen GA, Hospers GA, Bonenkamp JJ, Cuesta MA, Blaisse RJ, Busch OR, ten Kate FJ, Creemers GJ, Punt CJ, Plukker JT, Verheul HM, Spillenaar Bilgen EJ, van Dekken H, van der Sangen MJ, Rozema T, Biermann K, Beukema JC, Piet AH, van Rij CM, Reinders JG, Tilanus HW, van der Gaast A, 2012, Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366:2074–2084. , DOI 10.1056/ NEJMoa1112088 Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, Scarffe JH, Lofts FJ, Falk SJ, Iveson TJ, Smith DB, Langley RE, Verma M, Weeden S, Chua YJ, Participants MT, 2006, Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med 355: 11–20. , DOI 10.1056/NEJMoa055531 Vousden KH, Lu X, 2002, Live or let die: the cell's response to p53. Nat Rev Cancer 2:594–604. , DOI 10.1038/nrc864 Levine AJ, 1997, p53, the cellular gatekeeper for growth and division. Cell 88:323–331 Chen J, 2016, The cell-cycle arrest and apoptotic functions of p53 in tumor initiation and progression. Cold Spring Harb Perspect Med 6. , DOI 10.1101/cshperspect.a026104 Zhu YH, Zhang CW, Lu L, Demidov ON, Sun L, Yang L, Bulavin DV, Xiao ZC, 2009, Wip1 regulates the generation of new neural cells in the adult olfactory bulb through p53-dependent cell cycle control. Stem Cells 27:1433–1442. , DOI 10.1002/stem.65 Fiscella M, Zhang H, Fan S, Sakaguchi K, Shen S, Mercer WE, Vande Woude GF, O'Connor PM, Appella E, 1997, Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc Natl Acad Sci U S A 94: 6048–6053 Choi J, Appella E, Donehower LA, 2000, The structure and expression of the murine wildtype p53-induced phosphatase 1, Wip1, gene. Genomics 64:298–306. , DOI 10.1006/geno.2000. 6134 Li J, Yang Y, Peng Y, Austin RJ, van EyndhovenWG, Nguyen KC, Gabriele T, McCurrach ME, Marks JR, Hoey T, Lowe SW, Powers S, 2002, Oncogenic properties of PPM1D located within a breast cancer amplification epicenter at 17q23. Nat Genet 31:133–134. , DOI 10.1038/ng888 Bulavin DV, Demidov ON, Saito S, Kauraniemi P, Phillips C, Amundson SA, Ambrosino C, Sauter G, Nebreda AR, Anderson CW, Kallioniemi A, Fornace AJ Jr, Appella E, 2002, Amplification of PPM1D in human tumors abrogates p53 tumor-suppressor activity. Nat Genet 31:210–215. , DOI 10.1038/ng894 Nannenga B, Lu X, Dumble M, Van Maanen M, Nguyen TA, Sutton R, Kumar TR, Donehower LA, 2006, Augmented cancer resistance and DNA damage response phenotypes in PPM1D null mice. Mol Carcinog 45:594–604. , DOI 10.1002/mc. 20195 Wu B, Guo BM, Kang J, Deng XZ, Fan YB, Zhang XP, Ai KX, 2016, PPM1D exerts its oncogenic properties in human pancreatic cancer through multiple mechanisms. Apoptosis 21:365–378. , DOI 10.1007/s10495-015-1211-4 Kozakai Y, Kamada R, Kiyota Y, Yoshimura F, Tanino K, Sakaguchi K, 2014, Inhibition of C-terminal truncated PPM1D enhances the effect of doxorubicin on cell viability in human colorectal carcinoma cell line. Bioorg Med Chem Lett 24:5593–5596. , DOI 10.1016/j.bmcl.2014.10.093 Yang H, Gao XY, Li P, Jiang TS, 2015, PPM1D overexpression predicts poor prognosis in non-small cell lung cancer. Tumour Biol 36:2179–2184. , DOI 10.1007/s13277-014-2828-6 Wang W, Zhu H, Zhang H, Zhang L, Ding Q, Jiang H, 2014, Targeting PPM1D by lentivirus-mediated RNA interference inhibits the tumorigenicity of bladder cancer cells. Braz J Med Biol Res 47:1044–1049 Liu S, Qi L, HanW,Wan X, Jiang S, LiY, Xie Y, Liu L, Zeng F, Liu Z, Zu X, 2014, Overexpression of wip1 is associated with biologic behavior in human clear cell renal cell carcinoma. PLoS One 9: e110218. , DOI 10.1371/journal.pone.0110218 Richter M, Dayaram T, Gilmartin AG, Ganji G, Pemmasani SK, Van Der Key H, Shohet JM, Donehower LA, Kumar R, 2015, WIP1 phosphatase as a potential therapeutic target in neuroblastoma. PLoS One 10:e0115635. , DOI 10.1371/journal.pone. 0115635 Peng TS, He YH, Nie T, Hu XD, Lu HY, Yi J, Shuai YF, Luo M, 2014, PPM1D is a prognostic marker and therapeutic target in colorectal cancer. Exp Ther Med 8:430–434. , DOI 10. 3892/etm.2014.1762 Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi B, Varambally S, 2017, UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia 19:649–658. , DOI 10. 1016/j.neo.2017.05.002 Liu Y, Li K, Ren Z, Li S, Zhang H, Fan Q, 2012, Clinical implication of elevated human cervical cancer oncogene-1 expression in esophageal squamous cell carcinoma. J Histochem Cytochem 60: 512–520. , DOI 10.1369/0022155412444437 Liu HT, Wang N, Wang X, Li SL, 2010, Overexpression of Pim-1 is associated with poor prognosis in patients with esophageal squamous cell carcinoma. J Surg Oncol 102:683–688. , DOI 10.1002/jso.21627 Lu Z, Liu H, Xue L, Xu P, Gong T, Hou G, 2008, An activated Notch1 signaling pathway inhibits cell proliferation and induces apoptosis in human esophageal squamous cell carcinoma cell line EC9706. Int J Oncol 32:643–651 Lu X, Nguyen TA, Moon SH, Darlington Y, Sommer M, Donehower LA, 2008, The type 2C phosphatase Wip1: an oncogenic regulator of tumor suppressor and DNA damage response pathways. Cancer Metastasis Rev 27:123–135. , DOI 10. 1007/s10555-008-9127-x LambrosMB, Natrajan R, Geyer FC, Lopez-GarciaMA, Dedes KJ, Savage K, Lacroix-Triki M, Jones RL, Lord CJ, Linardopoulos S, Ashworth A, Reis-Filho JS, 2010, PPM1D gene amplification and overexpression in breast cancer: a qRT-PCR and chromogenic in situ hybridization study. Mod Pathol 23:1334–1345. , DOI 10.1038/modpathol.2010.121 Tan DS, Lambros MB, Rayter S, Natrajan R, Vatcheva R, Gao Q, Marchio C, Geyer FC, Savage K, Parry S, Fenwick K, Tamber N, Mackay A, Dexter T, Jameson C, McCluggage WG, Williams A, Graham A, Faratian D, El- Bahrawy M, Paige AJ, Gabra H, Gore ME, Zvelebil M, Lord CJ, Kaye SB, Ashworth A, Reis-Filho JS, 2009, PPM1D is a potential therapeutic target in ovarian clear cell carcinomas. Clin Cancer Res 15:2269–2280. , DOI 10.1158/1078-0432.CCR-08-2403 Hofstetter W, Swisher SG, Correa AM, Hess K, Putnam JB Jr, Ajani JA, Dolormente M, Francisco R, Komaki RR, Lara A, Martin F, Rice DC, Sarabia AJ, Smythe WR, Vaporciyan AA, Walsh GL, Roth JA, 2002, Treatment outcomes of resected esophageal cancer. Ann Surg 236:376–384; discussion 384-5. https://doi. org/10.1097/01.SLA.0000027925.23604.5C Urschel JD, Vasan H, Blewett CJ, 2002, A meta-analysis of randomized controlled trials that compared neoadjuvant chemotherapy and surgery to surgery alone for resectable esophageal cancer. Am J Surg 183:274–279 Zhang H, Chen W, Duan CJ, Zhang CF, 2013, Overexpression of HSPA2 is correlated with poor prognosis in esophageal squamous cell carcinoma. World J Surg Oncol 11:141. , DOI 10. 1186/1477-7819-11-141 Li GB, Zhang XL, Yuan L, Jiao QQ, Liu DJ, Liu J, 2013, Protein phosphatase magnesium-dependent 1delta, PPM1D, mRNA expression is a prognosis marker for hepatocellular carcinoma. PLoS One 8:e60775. , DOI 10.1371/journal.pone.0060775 Hirasawa A, Saito-Ohara F, Inoue J,Aoki D, Susumu N, Yokoyama T, Nozawa S, Inazawa J and Imoto I, 2003, Association of 17q21- q24 gain in ovarian clear cell adenocarcinomas with poor prognosis and identification of PPM1D and APPBP2 as likely amplification Targets Clin Cancer Res 9:1995–2004 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 387 EP 395 DI 10.1007/s12253-018-0518-1 PG 9 ER PT J AU Imredi, E Liszkay, G Kenessey, I Plotar, V Godeny, M Toth, B Fedorcsak, I Timar, J AF Imredi, Eleonora Liszkay, Gabriella Kenessey, Istvan Plotar, Vanda Godeny, Maria Toth, Bela Fedorcsak, Imre Timar, Jozsef TI Aquaporin-1 Protein Expression of the Primary Tumor May Predict Cerebral Progression of Cutaneous Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE AQP1 protein; Cutaneous melanoma; Melanoma brain metastases; Metastatic melanoma ID AQP1 protein; Cutaneous melanoma; Melanoma brain metastases; Metastatic melanoma AB Brain metastasis is a frequent complication of the progression of malignant melanoma. In a previous study aquaporin 1 (AQP1) protein expression was found to be associated with increased mortality and decreased progression free survival in cutaneous melanoma. To explore further the potential of this marker we studied the AQP1 protein expression in 67 metastatic melanoma patients using immunohistochemistry. Primary tumor samples were acquired from patients with brain (BR) (n = 44) and extracranial (EC) (n = 23) metastases, while brain metastatic samples were collected during neurosurgical resection (n = 5). Patients with brain metastases had shorter overall survival (p = 0.02) and significantly higher AQP1 expression in the primary tumors (median H-score = 250 vs. 140, p = 0.044) as compared to patients of the EC metastasis group. AQP1 expression was found to be significantly lower in the brain metastases compared to the corresponding primary tumors (median H-score = 35 vs. 300 p = 0.01). However, in brain metastases AQP1 expression was heterogenous, AQP1 protein was more abundant in the melanoma cells far away from the capillaries as compared to tumor cells adjacent to vessels indicating a hypoxia-driven expression of AQP1. We suggest that AQP1 expression could well be a prognostic marker of brain metastatic potential of human cutaneous melanoma. C1 [Imredi, Eleonora] Semmelweis University, 2nd Department of Pathology, Ulloi Str 93, H-1091 Budapest, Hungary. [Liszkay, Gabriella] National Institute of OncologyBudapest, Hungary. [Kenessey, Istvan] Semmelweis University, 2nd Department of Pathology, Ulloi Str 93, H-1091 Budapest, Hungary. [Plotar, Vanda] National Institute of OncologyBudapest, Hungary. [Godeny, Maria] National Institute of OncologyBudapest, Hungary. [Toth, Bela] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Fedorcsak, Imre] National Institute of Clinical NeurosciencesBudapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, Ulloi Str 93, H-1091 Budapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM jtimar@gmail.com CR Franceschini D, Franzese C, Navarria P, Ascolese AM, De Rose F, Del Vecchio M et al, 2016, Radiotherapy and immunotherapy: can this combination change the prognosis of patients with melanoma brain metastases? Cancer Treat Rev 50:1–8 Spagnolo F, Picasso V, Lambertini M, Ottaviano V, Dozin B, Queirolo P, 2016, Survival of patients with metastatic melanoma and brain metastases in the era of map-kinase inhibitors and immunologic checkpoint blockade antibodies: a systematic review. Cancer Treat Rev 45:38–45 Staudt M, Lasithiotakis K, Leiter U, Meier F, Eigentler T, Bamberg M, Tatagiba M, Brossart P, Garbe C, 2010, Determinants of survival in patients with brain metastases from cutaneous melanoma. Br J Cancer 102:1213–1218 Weidle UH, Birzele F, Kollmorgen G, Ruger R, 2016, Dissection of the process of brain metastasis reveals targets and mechanisms for molecular-based intervention. Cancer Genomics Proteomics 13: 245–258 Park ES, Kim SJ, Kim SW, Yoon SL, Leem SH, Kim SB, Kim SM, Park YY, Cheong JH, Woo HG, Mills GB, Fidler IJ, Lee JS, 2011, Cross-species hybridization of microarrays for studying tumor transcriptome of brain metastasis. PNAS 108:17456–17461 Nygaard V, Prasmickaite L, Vasiliauskaite K, Clancy T, Hovig E, 2014, Melanoma brain colonization involves the emergence of a brain-adaptive phenotype. Oncoscience 1:82–94 Wilhelm I,Molnar J, Fazakas C,Hasko J, Krizbai IA, 2013, Role of the blood-brain barrier in the formation of brain metastases. Int J Mol Sci 14:1383–1411 Dome B, Paku S, Somlai B, Timar J, 2002, Vascularization of cutaneous melanoma involves vessel co-option and has clinical significance. J Pathol 197:355–362 Kusters B, Leenders WP, Wesseling P, Smits D, Verrijp K, Ruiter DJ et al, 2002, Vascular endothelial growth factor-a(165, induces progression of melanoma brain metastases without induction of sprouting angiogenesis. Cancer Res 62:341–345 Nicchia GP, Stigliano C, Sparaneo A, Rossi A, Frigeri A, Svelto M, 2013, Inhibition of aquaporin-1 dependent angiogenesis impairs tumour growth in a mouse model of melanoma. J Mol Med 91: 613–623 Hu J, Verkman AS, 2006, Increased migration and metastatic potential of tumor cells expressing aquaporin water channels. FASEB J 20:1892–1894 Timar JPL, Raso E, 2007, Identification of the metastasis gene signature of human melanoma: dissection of tumor- and host components. Clin Exp Metastasis 24:211–316 Jaeger J, Koczan D, Thiesen HJ, Ibrahim SM, Gross G, Spang R, Kunz M, 2007, Gene expression signatures for tumor progression, tumor subtype, and tumor thickness in laser-microdissected melanoma tissues. Clin Cancer Res 13:806–815 Riker AI, Enkemann SA, Fodstad O, Liu S, Ren S, Morris C, Xi Y, Howell P, Metge B, Samant RS, Shevde LA, Li W, Eschrich S, Daud A, Ju J, Matta J, 2008, The gene expression profiles of primary and metastatic melanoma yields a transition point of tumor progression and metastasis. BMC Med Genet 1:13 Imredi E, Toth B, Doma V, Barbai T, Raso E, Kenessey I et al, 2016, Aquaporin 1 protein expression is associated with braf v600 mutation and adverse prognosis in cutaneous melanoma. Melanoma Res 26:254–260 Gershenwald JE, Scolyer RA, Hess KR, Sondak VK, Long GV, Ross MI, Lazar AJ, Faries MB, Kirkwood JM, McArthur G, Haydu LE, Eggermont AMM, Flaherty KT, Balch CM, Thompson JF, for members of the American Joint Committee on Cancer Melanoma Expert Panel and the International Melanoma Database and Discovery Platform, 2017, Melanoma staging: evidence-based changes in the American joint committee on cancer eighth edition cancer staging manual. CA Cancer J Clin 67:472–492 Fidler IJ, Schackert G, Zhang RD, Radinsky R, Fujimaki T, 1999, The biology of melanoma brain metastasis. Cancer Metastasis Rev 18:387–400 Paku S, Dome B, Toth R, Timar J, 2000, Organ-specificity of the extravasation process: an ultrastructural study. Clinical ExpMet 18: 481–492 Jilaveanu LB, Parisi F, Barr ML, Zito CR, Cruz-Munoz W, Kerbel RS, Rimm DL, Bosenberg MW, Halaban R, Kluger Y, Kluger HM, 2015, Plekha5 as a biomarker and potential mediator of melanoma brain metastasis. Clin Cancer Res 21:2138–2147 Gaziel-Sovran A, Osman I, Hernando E, 2013, In vivo modeling and molecular characterization: a path toward targeted therapy of melanoma brain metastasis. Front Oncol 3:127 Barbai T, Fejos Z, Puskas LG, Timar J, Raso E, 2015, The importance of microenvironment: the role of ccl8 in metastasis formation of melanoma. Oncotarget 6:29111–29128 Langley RR, Fidler IJ, 2011, The seed and soil hypothesis revisited–the role of tumor-stroma interactions in metastasis to different organs. Int J Cancer 128:2527–2535 Kenessey I, Banki B, Mark A, Varga N, Tovari J, Ladanyi A et al, 2012, Revisiting cb1 receptor as drug target in human melanoma. Pathol Oncol Res 18:857–866 Hasko J, Fazakas C, Molnar J, Nyul-Toth A, Herman H, Hermenean A et al, 2014, Cb2 receptor activation inhibits melanoma cell transmigration through the blood-brain barrier. Int J Mol Sci 15:8063–8074 Chen G, Chakravarti N, Aardalen K, Lazar AJ, Tetzlaff MT, Wubbenhorst B, Kim SB, Kopetz S, Ledoux AA, Gopal YNV, Pereira CG, DengW, Lee JS, Nathanson KL, Aldape KD, Prieto VG, Stuart D, Davies MA, 2014, Molecular profiling of patient-matched brain and extracranial melanoma metastases implicates the pi3k pathway as a therapeutic target. Clin Cancer Res 20:5537–5546 Rakosy Z, Vizkeleti L, Ecsedi S, Voko Z, Begany A, BarokMet al, 2007, EGFR gene copy number alterations in primary cutaneous malignant melanomas are associated with poor prognosis. Int J Cancer 121:1729–1737 Abreu-Rodriguez I, Sanchez Silva R, Martins AP, Soveral G, Toledo-Aral JJ, Lopez-Barneo J et al, 2011, Functional and transcriptional induction of aquaporin-1 gene by hypoxia; analysis of promoter and role of hif-1alpha. PLoS One 6:e28385 Simone L, Gargano CD, Pisani F, Cibelli A, Mola MG, Frigeri A, Svelto M, Nicchia GP, 2018, Aquaporin-1 inhibition reduces metastatic formation in a mouse model of melanoma. J Cell Mol Med 22:904–912 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 405 EP 410 DI 10.1007/s12253-018-0513-6 PG 6 ER PT J AU Ahmed, AHM Ali, HM Abbas, HH Elatrash, AG Foda, AMA AF Ahmed, A H Mohamed Ali, Hassan Mohamed Abbas, Hafez Hashem Elatrash, Ali Gamal Foda, AlRahman Mohammad Abd TI Expression of TOMM34 and Its Clinicopathological Correlations in Urothelial Carcinoma of the Bladder SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TOMM34; Urothelial; Mitochondrial; Markers ID TOMM34; Urothelial; Mitochondrial; Markers AB The substantial difference between normal cells and cancer cells in terms of their energy metabolism in mitochondria provides an interesting basis for the development of novel therapeutic agents targeting energy machinery of tumour cells. TOMM34 is one of the Tom (translocase of the outer membrane of mitochondria) family that was found to be overexpressed in colorectal, hepatocellular, lung and early invasive breast carcinomas. The expression profile of mitochondrial translocases in bladder cancer compared to normal urinary bladder tissues has not been investigated yet. Therefore, the aim of the current study is to investigate the expression pattern of TOMM34 in bladder cancer tissues and explore its correlation with the clinicopathological parameters of those cases. Sixty patients who underwent either transurethral resection or radical cystectomy for bladder cancer were included in this study with revision of all their clinicopathological data and tumor slides. Ten histologically normal urothelial biopsies were also included. Immunohistochemical staining for TOMM34 was done and semi-quantitatively scored using the modified H-score. All relations were analysed using established statistical methodologies. TOMM34 overexpression was significantly associated with high tumour stage, muscle invasion and high grade. Significant positive association was observed between TOMM34 expression and poor outcome in terms of shorter disease-specific survival. This study suggests TOMM34 as a biomarker of progression and poor prognosis in urothelial cell carcinoma patients. Furthermore, we suggest a role played by mitochondrial machinery in urothelial cell carcinoma progression, which is a potential target for the newly-discovered vaccine therapy for urothelial cell carcinoma. C1 [Ahmed, A H Mohamed] Suez Canal University, Faculty of Medicine, Department of PathologyIsmailia, Egypt. [Ali, Hassan Mohamed] Suez Canal University, Faculty of Medicine, Department of UrologyIsmailia, Egypt. [Abbas, Hafez Hashem] Suez Canal University, Faculty of Medicine, Department of UrologyIsmailia, Egypt. [Elatrash, Ali Gamal] Suez Canal University, Faculty of Medicine, Department of UrologyIsmailia, Egypt. [Foda, AlRahman Mohammad Abd] Mansoura University, Faculty of Medicine, Department of Pathology, 35516 Mansoura, Egypt. RP Foda, AMA (reprint author), Mansoura University, Faculty of Medicine, Department of Pathology, 35516 Mansoura, Egypt. EM abdofoda@mans.edu.eg CR Badar F, Sattar A, Meerza F et al, 2009, Carcinoma of the urinary bladder in a tertiary care setting in a developing country. Asian Pac J Cancer Prev 10(3):449–452 Rogers CG, Palapattu GS, Shariat SF et al, 2006, Clinical outcomes following radical cystectomy for primary nontransitional cell carcinoma of the bladder compared to transitional cell carcinoma of the bladder. J Urol 175(6):2048–2053 discussion 2053 Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63(1):11–30 Kirkali Z, Chan T, Manoharan M et al, 2005, Bladder cancer: epidemiology, staging and grading, and diagnosis. Urology 66(6 Suppl 1):4–34 Stenzl A, Cowan NC, De Santis M et al, 2011, Treatment of muscle-invasive and metastatic bladder cancer: update of the EAU guidelines. Eur Urol 59(6):1009–1018 Neupert W, 1997, Protein import into mitochondria. Annu Rev Biochem 66:863–917 Hoogenraad NJ, Ward LA, Ryan MT, 2002, Import and assembly of proteins into mitochondria of mammalian cells. Biochim Biophys Acta 1592(1):97–105 Trachootham D, Zhang H, ZhangWet al, 2008, Effective elimination of fludarabine-resistant CLL cells by PEITC through a redoxmediated mechanism. Blood 112(5):1912–1922 Trachootham D, Alexandre J, Huang P, 2009, Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat Rev Drug Discov 8(7):579–591 LuW, OgasawaraMA, Huang P, 2007)Models of reactive oxygen species in cancer. Drug Discov Today Dis Model 4(2):67–73 Chewawiwat N, Yano M, Terada K et al, 1999, Characterization of the novel mitochondrial protein import component, Tom34, in mammalian cells. J Biochem 125(4):721–727 Mukhopadhyay A, Avramova LV, Weiner H, 2002, Tom34 unlike Tom20 does not interact with the leader sequences ofmitochondrial precursor proteins. Arch Biochem Biophys 400(1):97–104 Shimokawa T, Matsushima S, Tsunoda T et al, 2006, Identification of TOMM34, which shows elevated expression in the majority of human colon cancers, as a novel drug target. Int J Oncol 29(2):381– 386 Aleskandarany MA, Negm OH, Rakha EA et al, 2012, TOMM34 expression in early invasive breast cancer: a biomarker associated with poor outcome. Breast Cancer Res Treat 136(2):419–427 Matsushita N, Yamamoto S, Inoue Yet al, 2017, RT-qPCR analysis of the tumor antigens TOMM34 and RNF43 in samples extracted from paraffin-embedded specimens of colorectal cancer. Oncol Lett 14(2):2281–2287 McCarty KS Jr, Miller LS, Cox EB et al, 1985, Estrogen receptor analyses. Correlation of biochemical and immunohistochemical methods using monoclonal antireceptor antibodies. Arch Pathol Lab Med 109(8):716–721 Camp RL, Dolled-Filhart M, Rimm DL, 2004, X-tile: a new bioinformatics tool for biomarker assessment and outcome-based cutpoint optimization. Clin Cancer Res 10(21):7252–7259 de Moura MB, dos Santos LS, Van Houten B, 2010, Mitochondrial dysfunction in neurodegenerative diseases and cancer. Environ Mol Mutagen 51(5):391–405 Kroemer G, 2006, Mitochondria in cancer. Oncogene 25(34): 4630–4632 Zhao Z, Han F, He Y et al, 2014, Stromal-epithelial metabolic coupling in gastric cancer: stromal MCT4 and mitochondrial TOMM20 as poor prognostic factors. Eur J Surg Oncol 40:1361– 1368 Sotgia F, Whitaker-Menezes D, Martinez-Outschoorn UE et al, 2012, Mitochondrial metabolism in cancer metastasis: visualizing tumor cell mitochondria and the "reverse Warburg effect" in positive lymph node tissue. Cell Cycle 11(7):1445–1454 Johnson JM, Lai SY, Cotzia P et al, 2015, Mitochondrial metabolism as a treatment target in anaplastic thyroid cancer. Semin Oncol 42(6):915–922 Mikkilineni L, Whitaker-Menezes D, Domingo-Vidal M et al, 2017, Hodgkin lymphoma: a complex metabolic ecosystem with glycolytic reprogramming of the tumormicroenvironment. Semin Oncol 44(3):218–225 Gooptu M, Whitaker-Menezes D, Sprandio J et al, 2017, Mitochondrial and glycolytic metabolic compartmentalization in diffuse large B-cell lymphoma. Semin Oncol 44(3):204–217 Asmarinah A, Paradowska-Dogan A, Kodariah R et al, 2014, Expression of the Bcl-2 family genes and complexes involved in the mitochondrial transport in prostate cancer cells. Int J Oncol 45(4):1489–1496 Liu Y, Lan L, Huang K et al, 2014, Inhibition of Lon blocks cell proliferation, enhances chemosensitivity by promoting apoptosis and decreases cellular bioenergetics of bladder cancer potential roles of Lon as a prognostic marker and therapeutic target in bladder cancer. Oncotarget 5:11209–11224 Mo M, Peng F, Wang L et al, 2013, Roles of mitochondrial transcription factor A and microRNA-590-3p in the development of bladder cancer. Oncol Lett 6:617–623 Nie X, Li M, Lu B et al, 2013, Down-regulating overexpressed human Lon in cervical cancer suppresses cell proliferation and bioenergetics. PLoS One 8(11):e81084 Gibellini L, Pinti M, Boraldi F et al, 2014, Silencing of mitochondrial Lon protease deeply impairs mitochondrial proteome and function in colon cancer cells. FASEB J 28(12):5122–5135 Gibellini L, Pinti M, Bartolomeo R et al, 2015, Inhibition of Lon protease by triterpenoids alters mitochondria and is associated to cell death in human cancer cells. Oncotarget 6:25466–25483 Jin B, Fu G, Pan H et al, 2011, Anti-tumour efficacy of mitofusin-2 in urinary bladder carcinoma. Med Oncol 28 Suppl 1:S373–S380 Oyama T, Ishikawa Y, Hayashi M et al, 2007, The effects of fixation, processing and evaluation criteria on immunohistochemical detection of hormone receptors in breast cancer. Breast Cancer 14(2):182–188 Espina V, Wulfkuhle J, Calvert VS et al, 2008, Reverse phase protein microarrays for theranostics and patient-tailored therapy. Methods Mol Biol 441:113–128 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 411 EP 418 DI 10.1007/s12253-018-0524-3 PG 8 ER PT J AU Pal, I Illes, Varoczy, L AF Pal, Ildiko Illes, Arpad Varoczy, Laszlo TI Multiple Myeloma of the Young – a Single Center Experience Highlights Future Directions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Multiple myeloma; Young; 40 years; Progression-free survival; Overall survival ID Multiple myeloma; Young; 40 years; Progression-free survival; Overall survival AB Multiple myeloma is quite uncommon in the young population. We performed a retrospective review in our database from 2006 to 2015 to examine the clinical features, outcomes and survival of multiple myeloma patients ≤40 years old. Among 312 newly diagnosed patients we found sixteen (5.1%) who were 40 years old or younger. Their characteristics including M-protein type, genetical alterations, clinical symptoms and disease stage were as various as those in the older population. All but two young patients underwent autologous stem cell transplantation after the induction treatment. Their response to treatment did not differ markedly from the older patients. We also compared the survival data of patients ≤40 years and > 40 years old. The 5-year progression-free survival were 48% and 35%, the 5-year overall survival were 83% and 53% respectively, the latter showing a significant advantage for the younger population. 70% of the young patients received maintenance or consolidation therapy after the initial treatment. Although several effective new therapies have been introduced recently, there is still an unmet need for curative treatment options for young and fit multiple myeloma patients. C1 [Pal, Ildiko] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Varoczy, Laszlo] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. RP Varoczy, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, H-4032 Debrecen, Hungary. EM laszlo.varoczy@gmail.com CR Kazandijan D, 2016, Multiple myeloma epidemiology and survival: a unique malignancy. Semin Oncol 43(6):676–681 Rajkumar SV, Dimopoulos MA, Palumbo A et al, 2014, International myeloma working group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol 15:538–548 Kumar S, 2017, Emerging options in multiple myeloma: targeted, immune and epigenetic therapies. Hematology Am Soc Hematol Educ Program 2017(1):518–524 Chng WJ, Dispenzieri A, Chim CS et al, 2014, IMWG consensus on risk stratification in multiple myeloma. Leukemia 28:269–277 Kyle RA, GertzMA,Witzig TE, Lust JA, LacyMQ, Dispenzieri A, Fonseca R, Rajkumar SV, Offord JR, Larson DR, Plevak ME, Therneau TM, Greipp PR, 2003, Review of 1072 patients with newly diagnosed multiple myeloma. Mayo Clin Proc 78:21–33 Cheema PK, Zadeh S, Kukreti V, Reece D, Chen C, Trudel S, Mikhael J, 2009, Age 40 years and under does not confer superior prognosis in patients with multiple myeloma undergoing upfront autologous stem cell transplant. Biol Blood Marrow Transplant. 15: 686–693 Geetha N, Jayaparkash M, Rekhenair A et al, 1999, Plasma cell neoplasms in young. Br J Radiol 72:1012–1015 Ludwig H, Durie BGM, Bolejack et al, 2008)Myeloma in patients younger than age 50 yearsa presents with more favorable features and shows better survival: an analysis of 10.549 patients from the Internationaly Myeloma Working Group. Blood. 111: 4039–4047 Blade J, Kyle RA, Griepp PR, 1996, Presenting features and prognosis in 72 patients with multiple myeloam who were younger than 40 years. Br J Haematol 93:345–351 Jurczyszyn A, Nahi H, Avivi I, Gozzetti A, Niesvizky R, Yadlapati S, Jayabalan DS, Robak P, Pika T, Andersen KT, Rasche L, Madry K, Woszczyk D, Razny M, Usnarska-Zubkiewicz L, Knopinska- Posluszny W, Wojciechowska M, Guzicka-Kazimierczak R, Joks M, Grosicki S, Ciepluch H, Rymko M, Vesole DH, Castillo JJ, 2016, Characteristics and outcomes of patients with multiple myeloma aged 21-40 years versus 41-60 years: a multi-institutional case-control study. Br J Haematol 175:884–891 Jurczyszyn A, Davila J, Kortum KMet al, 2018)Multiplemyeloma in patients up to 30 years of age: amulticenter retrospective study of 52 cases. Leuk Lymphoma 22:1–6 Mateos MV, San Miguel JV, 2017, Management of multiple myeloma in the newly diagnosed patient. Hematology Am Soc Hematol Educ Program. 2017(1):498–507 Sonneveld P, 2017, Management of multiple myeloma in the relapsed / refarcory patient. Hematology Am Soc Hematol Educ Program. 2017(1):508–517 Chan HSH, Chen CI, Reece DE, 2017, Current review on high-risk myeloma. Curr Hematol Mal Rep 12:96–108 Ludwig H, Zojer H, 2017, Fixed duration vs coninuous therapy in multiple myeloma. Hematology Am Soc Hematol Educ Program. 2017(1):212–222 Mir MA, Kapoor P, Kumar S, Pandey S, Dispenzieri A, Lacy MQ, Dingli D, Hogan W, Buadi F, Hayman S, Gandhi M, Gertz MA, 2015, Trends and outcomes in allogeneic hematopoietic stem cell transplant for multiple myeloma at Mayo Clinic. Clin Ly Myeloma & Leuk 15:349–357 Htut M, D1Souza A, Krishnan A et al, 2017, Autologous / Allogeneic hematopoietic Cell Transplant versus tandem autologous transplantation for multiple myeloma: comparison of longterm post-relapse survival. Biol Blood Marrow Transplant pii: S1083–8791(17)30793–0., DOI , DOI 10.1016/j.bbmt. 2017.10.024. [Epub ahead of print] NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 419 EP 424 DI 10.1007/s12253-018-0526-1 PG 6 ER PT J AU An, N Cheng, D AF An, Ning Cheng, Donghui TI The Long Noncoding RNA HOST2 Promotes Gemcitabine Resistance in Human Pancreatic Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic neoplasms; LncRNA; HOST2; Gemcitabine ID Pancreatic neoplasms; LncRNA; HOST2; Gemcitabine AB Our study was aimed to identify the fundamental role of lncRNA HOST2 in gemcitabine resistance regulation in human pancreatic cancer cells. The levels of HOST2 in pancreatic cancer cell lines were measured by quantitative real-time PCR (qRT-PCR). Due to high expression and strong gemcitabine resistance, Hs766T and AsPC-1 cell lines were selected to be knockdown the expression of HOST2 by transfection sh-HOST2. After manipulation of HOST2, the cell proliferation induced by gemcitabine was examined by CCK-8 assay. Next, colony formation ability of Hs766Tand AsPC-1 cell lines was determined by clone-forming assay. At last, the relationship between HOST2 and cell apoptosis in Hs766T and AsPC-1 cell lines was evaluated by flow cytometry. QRT-PCR revealed that HOST2 was overexpressed in six pancreas neoplasm cell lines compared with normal cell lines HPDE6-C7. HOST2 expression levels in group resistant to gemcitabine were higher than the group sensitive to gemcitabine. Additionally, CCK-8 assay verified that cell proliferation was inhibited by sh-HOST2 with or without gemcitabine treatment. Furthermore, clone-forming assay revealed that colony formation ability was weakened by downregulated HOST2 with or without gemcitabine treatment. Flow cytometry revealed that cell apoptosis induced by gemcitabine was promoted by sh-HOST2. In conclusion, down-regulated HOST2 inhibited proliferation and promoted apoptosis of pancreas cancer cells with or without gemcitabine treatment. Thus, HOST2 is a potential therapeutic target for gemcitabine chemoresistance in pancreatic neoplasms. C1 [An, Ning] Sichuan Medical Academy & Sichuan People′s Hospital, Department of Hepatobiliary Surgery, No 32 Western Third Section of First Ring Road, 610072 Chengdu, Sichuan, China. [Cheng, Donghui] Sichuan Medical Academy & Sichuan People′s Hospital, Department of Hepatobiliary Surgery, No 32 Western Third Section of First Ring Road, 610072 Chengdu, Sichuan, China. RP Cheng, D (reprint author), Sichuan Medical Academy & Sichuan People′s Hospital, Department of Hepatobiliary Surgery, 610072 Chengdu, China. EM chengyongqin111111@163.com CR Moschovis D, Gazouli M, Tzouvala M, Vezakis A, Karamanolis G, 2017, Long non-coding RNA in pancreatic adenocarcinoma and pancreatic neuroendocrine tumors. Ann Gastroenterol 30(6):622– 628. , DOI 10.20524/aog.2017.0185 Huang X, Zhi X, Gao Y, Ta N, Jiang H, Zheng J, 2016, LncRNAs in pancreatic cancer. Oncotarget 7(35):57379–57390. https://doi. org/10.18632/oncotarget.10545 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108. , DOI 10.3322/caac.21262 Klimstra DS, 2007, Nonductal neoplasms of the pancreas. Mod Pathol 20(Suppl 1):S94–S112. , DOI 10.1038/ modpathol.3800686 Mayor S, 2015, Immunotherapy improves overall survival in pancreatic cancer. Lancet Oncol 16(2):e58. , DOI 10.1016/ S1470-2045(15)70017-3 Bergmann L, Maute L, Heil G, Russel J, Weidmann E, Koberle D, Fuxius S, Weigang-Kohler K, AulitzkyWE,Wormann B, Hartung G,Moritz B, Edler L, Burkholder I, Scheulen ME, Richly H, 2015, A prospective randomised phase-II trial with gemcitabine versus gemcitabine plus sunitinib in advanced pancreatic cancer: a study of the CESAR Central European Society for Anticancer Drug Research-EWIV. Eur J Cancer 51(1):27–36. , DOI 10. 1016/j.ejca.2014.10.010 Seufferlein T, Bachet JB, Van Cutsem E, Rougier P, Group EGW, 2012, Pancreatic adenocarcinoma: ESMO-ESDO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 23(Suppl 7):vii33–vii40. , DOI 10.1093/annonc/mds224 Stathis A, Moore MJ, 2010, Advanced pancreatic carcinoma: current treatment and future challenges. Nat Rev Clin Oncol 7(3):163– 172. , DOI 10.1038/nrclinonc.2009.236 Wang L, Dong P, Wang W, Huang M, Tian B, 2017, Gemcitabine treatment causes resistance and malignancy of pancreatic cancer stem-like cells via induction of lncRNA HOTAIR. Exp Ther Med 14(5):4773–4780. , DOI 10.3892/etm.2017.5151 Wang Z, LiY, Ahmad A, Banerjee S, Azmi AS, KongD, Sarkar FH, 2011, Pancreatic cancer: understanding and overcoming chemoresistance. Nat Rev Gastroenterol Hepatol 8(1):27–33. , DOI 10.1038/nrgastro.2010.188 O'Reilly EM, Abou-Alfa GK, 2007, Cytotoxic therapy for advanced pancreatic adenocarcinoma. Semin Oncol 34(4):347–353. , DOI 10.1053/j.seminoncol.2007.05.009 Zhou M, Ye Z, Gu Y, Tian B, Wu B, Li J, 2015, Genomic analysis of drug resistant pancreatic cancer cell line by combining long noncoding RNA and mRNA expression profling. Int J Clin Exp Pathol 8(1):38–52 Mittal A, Chitkara D, Behrman SW, Mahato RI, 2014, Efficacy of gemcitabine conjugated and miRNA-205 complexed micelles for treatment of advanced pancreatic cancer. Biomaterials 35(25): 7077–7087. , DOI 10.1016/j.biomaterials.2014.04.053 Di Gesualdo F, Capaccioli S, Lulli M, 2014, A pathophysiological view of the long non-coding RNAworld. Oncotarget 5(22):10976– 10996. , DOI 10.18632/oncotarget.2770 Chandra Gupta S, Nandan Tripathi Y, 2017, Potential of long non-coding RNAs in cancer patients: from biomarkers to therapeutic targets. Int J Cancer 140(9):1955–1967. , DOI 10.1002/ijc.30546 Rangel LB, Sherman-Baust CA, Wernyj RP, Schwartz DR, Cho KR, Morin PJ, 2003, Characterization of novel human ovarian cancer-specific transcripts, HOSTs, identified by serial analysis of gene expression. Oncogene 22(46):7225–7232. , DOI 10. 1038/sj.onc.1207008 Gao Y, Meng H, Liu S, Hu J, ZhangY, Jiao T, Liu Y, Ou J,Wang D, Yao L, Liu S, Hui N, 2015, LncRNA-HOST2 regulates cell biological behaviors in epithelial ovarian cancer through a mechanism involving microRNA let-7b. Hum Mol Genet 24(3):841–852. , DOI 10.1093/hmg/ddu502 Lu PW, Li L, Wang F, Gu YT, 2018, Effects of long non-coding RNA HOST2 on cell migration and invasion by regulating MicroRNA let-7b in breast cancer. J Cell Biochem 119(6):4570– 4580. , DOI 10.1002/jcb.26606 Wang W, Li X, Meng FB, Wang ZX, Zhao RT, Yang CY, 2017, Effects of the long non-coding RNA HOST2 on the proliferation, migration, invasion and apoptosis of human osteosarcoma cells. Cell Physiol Biochem 43(1):320–330. , DOI 10.1159/000480412 Liu RT, Cao JL, Yan CQ,Wang Y, An CJ, Lv HT, 2017, Effects of LncRNA-HOST2 on cell proliferation, migration, invasion and apoptosis of human hepatocellular carcinoma cell line SMMC-7721. Biosci Rep 37(2):BSR20160532. , DOI 10.1042/ BSR20160532 Arumugam T, Ramachandran V, Fournier KF,Wang H,Marquis L, Abbruzzese JL, Gallick GE, Logsdon CD, McConkey DJ, ChoiW, 2009, Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res 69(14):5820–5828. , DOI 10.1158/0008-5472.CAN-08-2819 Muller S, Raulefs S, Bruns P, Afonso-Grunz F, Plotner A, Thermann R, Jager C, Schlitter AM, Kong B, Regel I, Roth WK, Rotter B, Hoffmeier K, Kahl G, Koch I, Theis FJ, Kleeff J,Winter P, Michalski CW(2015, Next-generation sequencing reveals novel differentially regulated mRNAs, lncRNAs, miRNAs, sdRNAs and a piRNA in pancreatic cancer. Mol Cancer 14:94. , DOI 10.1186/s12943-015-0358-5 You L, Chang D, Du HZ, Zhao YP, 2011, Genome-wide screen identifies PVT1 as a regulator of gemcitabine sensitivity in human pancreatic cancer cells. Biochem Biophys Res Commun 407(1):1– 6. , DOI 10.1016/j.bbrc.2011.02.027 Li C, Zhao Z, Zhou Z, Liu R, 2016, Linc-ROR confers gemcitabine resistance to pancreatic cancer cells via inducing autophagy and modulating the miR-124/PTBP1/PKM2 axis. Cancer Chemother Pharmacol 78(6):1199–1207. https://doi. org/10.1007/s00280-016-3178-4 Li Z, Zhao X, Zhou Y, Liu Y, Zhou Q, Ye H,Wang Y, Zeng J, Song Y, Gao W, Zheng S, Zhuang B, Chen H, Li W, Li H, Li H, Fu Z, Chen R, 2015, The long non-coding RNA HOTTIP promotes progression and gemcitabine resistance by regulatingHOXA13 in pancreatic cancer. J Transl Med 13:84. , DOI 10.1186/s12967- 015-0442-z Zhang Y, Tang X, Shi M, Wen C, Shen B, 2017, MiR-216a decreasesMALAT1 expression, induces G2/Marrest and apoptosis in pancreatic cancer cells. Biochem Biophys Res Commun 483(2): 816–822. , DOI 10.1016/j.bbrc.2016.12.167 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 425 EP 431 DI 10.1007/s12253-018-0486-5 PG 7 ER PT J AU de Souza, GM de Jesus, FS Santos, ME Gomes, SBE Filho, dPSA Santos, MSE da Silveira, HL Santos, HSS de Paula, MBA Farias, CL Guimaraes, LSA AF de Souza, Goncalves Marcela de Jesus, Ferreira Sabrina Santos, Mangabeira Eloa Gomes, Stenio Batista Emisael Filho, de Paulo Santiago Arlen Santos, Macedo Sobrinho Eliane da Silveira, Henrique Luiz Santos, Henrique Sousa Sergio de Paula, Mauricio Batista Alfredo Farias, Conceicao Lucyana Guimaraes, Luiz Sena Andre TI Radiation Therapy Reduced Blood Levels of LDH, HIF-1α, and miR-210 in OSCC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Radiotherapy; Warburg effect; OSCC ID Radiotherapy; Warburg effect; OSCC AB Radiation Therapy (RT) is a treatment option for a large number of neoplasias. However, the effect of RT on the level of hypoxia markers is poorly understood. The present study aimed to investigate the effect of RT on the levels of hypoxic markers in Oral squamous cell carcinoma (OSCC). Evaluation of HIF-1α and miR-210 levels in OSCC was performed. Then a proteomic analysis was performed to identify candidate hypoxic targets of RT. To validate proteomic studies, the effect of RT on HIF-1α, miR-210, PDH-A and LDH-A levels under hypoxia was assessed by qRT-PCR. The impact of RT in hypoxia markers was evaluated in patients to confirm in vitro results. An increase in the HIF-1α levels was observed in OSCC. RT reduced OSCC cell proliferation and migration. Interestingly, hypoxia could revert the effect of radiation on OSCC phenotype. However, proteomics analyses suggested that LDH is one of the critical targets of RT even in hypoxia. Moreover, RT decreased HIF-1α, miR-210, and LDH even in hypoxia. The current study demonstrated that hypoxia could revert the effects of RT in the OSCC context. However, RT reduces the levels HIF-1α, miR-210 and LDH in vivo and in vitro. The consequences of RT in blood should be carefully investigated. C1 [de Souza, Goncalves Marcela] Universidade Estadual de Montes Claros, Department of DentistryMontes Claros, Minas Gerais, Brazil. [de Jesus, Ferreira Sabrina] Universidade Estadual de Montes Claros, Department of DentistryMontes Claros, Minas Gerais, Brazil. [Santos, Mangabeira Eloa] Universidade Estadual de Montes Claros, Department of DentistryMontes Claros, Minas Gerais, Brazil. [Gomes, Stenio Batista Emisael] Universidade Estadual de Montes Claros, Department of DentistryMontes Claros, Minas Gerais, Brazil. [Filho, de Paulo Santiago Arlen] Hospital Dilson GodinhoMontes Claros, Minas Gerais, Brazil. [Santos, Macedo Sobrinho Eliane] Universidade Estadual de Montes Claros, Department of DentistryMontes Claros, Minas Gerais, Brazil. [da Silveira, Henrique Luiz] Universidade Estadual de Montes Claros, Department of DentistryMontes Claros, Minas Gerais, Brazil. [Santos, Henrique Sousa Sergio] Universidade Federal de Minas Gerais (UFMG), Food Engineering College, Institute of Agricultural SciencesMontes Claros, Minas Gerais, Brazil. [de Paula, Mauricio Batista Alfredo] Universidade Estadual de Montes Claros, Department of DentistryMontes Claros, Minas Gerais, Brazil. [Farias, Conceicao Lucyana] Universidade Estadual de Montes Claros, Department of DentistryMontes Claros, Minas Gerais, Brazil. [Guimaraes, Luiz Sena Andre] Universidade Estadual de Montes Claros, Department of DentistryMontes Claros, Minas Gerais, Brazil. RP Guimaraes, LSA (reprint author), Universidade Estadual de Montes Claros, Department of Dentistry, Montes Claros, Brazil. EM andreluizguimaraes@gmail.com CR Zhang S, Tian L, Ma P, Sun Q, Zhang K, 2015, GuanchaoWang, et al. potential role of differentially expressed lncRNAs in the pathogenesis of oral squamous cell carcinoma. Arch Oral Biol 60(10): 1581–1587 Gupta A, Baxi S, Hoyne C, 2017, Assessing feasibility, compliance and toxicity of concomitant chemo-radiotherapy in head and neck cancers in the Northern Territory: initial experience and challenges. J Med Radiat Sci 64(2):131–137 Fraga CA, de Oliveira MV, de Oliveira ES, Barros LO, Santos FB, Gomez RS et al, 2012, A high HIF-1alpha expression genotype is associated with poor prognosis of upper aerodigestive tract carcinoma patients. Oral Oncol 48(2):130–135 Gruber G, Greiner RH, Hlushchuk R,Aebersold DM, Altermatt HJ, Berclaz G, Djonov V, 2004, Hypoxia-inducible factor 1 alpha in high-risk breast cancer: an independent prognostic parameter? Breast Cancer Res : BCR 6(3):R191–R198 Thongchot S, Yongvanit P, Loilome W, Seubwai W, Phunicom K, TassaneeyakulW, Pairojkul C, PromkotraW, Techasen A, Namwat N, 2014, High expression of HIF-1alpha, BNIP3 and PI3KC3: hypoxia-induced autophagy predicts cholangiocarcinoma survival andmetastasis. Asian Pacific journal of Cancer Prevention : APJCP 15(14):5873–5878 Warnakulasuriya S, 2009, Global epidemiology of oral and oropharyngeal cancer. Oral Oncol 45(4–5):309–316 Stelzle F, Knipfer C, Schuster M, Bocklet T, Noth E, Adler W, Schempf L, Vieler P, Riemann M, Neukam FW, Nkenke E, 2013, Factors influencing relative speech intelligibility in patients with oral squamous cell carcinoma: a prospective study using automatic, computer-based speech analysis. Int J Oral Maxillofac Surg 42(11): 1377–1384 Nordsmark M, Bentzen SM, RudatV, Brizel D, Lartigau E, Stadler P, BeckerA, AdamM, MollsM,Dunst J, Terris DJ,Overgaard J, 2005, Prognostic value of tumor oxygenation in 397 head and neck tumors after primary radiation therapy. An international multi-center study. Radiother Oncol 77(1):18–24 Barker HE, Paget JT, Khan AA, Harrington KJ, 2015, The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer 15(7):409–425 Guimaraes TA, Farias LC, Santos ES, de Carvalho Fraga CA, Orsini LA, de Freitas TL et al, 2016, Metformin increases PDH and suppresses HIF-1alpha under hypoxic conditions and induces cell death in oral squamous cell carcinoma. Oncotarget 7(34): 55057–55068 Otto AM, 2016, Warburg effect(s)—a biographical sketch of Otto Warburg and his impacts on tumor metabolism. Cancer Metab 4(1): 5 Luc R, Tortorella SM, Ververis K, Karagiannis TC, 2015, Lactate as an insidious metabolite due to theWarburg effect. Mol Biol Rep 42(4):835–840 Loeffelbein DJ, Eiber M, Mayr P, Souvatzoglou M, Mucke T, von Bomhard A et al, 2015, Loco-regional recurrence after surgical treatment of oral squamous cell carcinoma: proposals for followup imaging based on literature, national guidelines and institutional experience. J Craniomaxillofac Surg 43(8):1546–1552 Allison SJ,Knight JR, Granchi C, Rani R, Minutolo F, Milner J et al, 2014, Identification of LDH-A as a therapeutic target for cancer cell killing via, i, p53/NAD(H)-dependent and, ii, p53-independent pathways. Oncogene 3:e102 Read JA, Winter VJ, Eszes CM, Sessions RB, Brady RL, 2001, Structural basis for altered activity of M- and H-isozyme forms of human lactate dehydrogenase. Proteins 43(2):175–185 Fantin VR, St-Pierre J, Leder P, 2006, Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell 9(6):425–434 Semenza GL, 1999, Regulation of mammalian O2 homeostasis by hypoxia-inducible factor 1. Annu Rev Cell Dev Biol 15:551–578 de Carvalho Fraga CA, Alves LR, Marques-Silva L, de Sousa AA, Jorge AS, de Jesus SF et al, 2013, High HIF-1alpha expression genotypes in oral lichen planus. Clin Oral Investig 17(9):2011–2015 Hoffmann AC, Mori R, Vallbohmer D, Brabender J, Drebber U, Baldus SE, Klein E, AzumaM, Metzger R, Hoffmann C, Hoelscher AH, Danenberg KD, Prenzel KL, Danenberg PV, 2008, High expression of heparanase is significantly associated with dedifferentiation and lymph node metastasis in patients with pancreatic ductal adenocarcinomas and correlated to PDGFA and via HIF1a to HBEGF and bFGF. J Gastrointest Surg 12(10):1674–1681 discussion 81-2 Tuomisto A, Garcia-Solano J, Sirnio P, Vayrynen J, Perez- Guillermo M, Makinen MJ et al, 2016, HIF-1alpha expression and high microvessel density are characteristic features in serrated colorectal cancer. Virchows Arch 469(4):395–404 van derGroep P, Bouter A, Menko FH, van derWall E, van Diest PJ, 2008, High frequency of HIF-1α overexpression in BRCA1 related breast cancer. Breast Cancer Res Treat 111(3):475–480 Aebersold DM, Burri P, Beer KT, Laissue J, Djonov V, Greiner RH, Semenza GL, 2001, Expression of hypoxia-inducible factor-1alpha: a novel predictive and prognostic parameter in the radiotherapy of oropharyngeal cancer. Cancer Res 61(7):2911–2916 Silva P, Slevin NJ, Sloan P,Valentine H, Cresswell J, Ryder D, Price P, Homer JJ, West CML, 2008, Prognostic significance of tumor hypoxia inducible factor-1alpha expression for outcome after radiotherapy in oropharyngeal cancer. Int J Radiat Oncol Biol Phys 72(5):1551–1559 Langhammer S, Najjar M, Hess-Stumpp H, Thierauch KH, 2011, LDH-A influences hypoxia-inducible factor 1alpha, HIF1 alpha, and is critical for growth of HT29 colon carcinoma cells in vivo. Target Oncol 6(3):155–162 Pouyssegur J, Dayan F, Mazure NM, 2006, Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 441(7092):437–443 Karatas OF, Suer I, Yuceturk B, Yilmaz M, Oz B, Guven G et al, 2016, Identification of microRNA profile specific to cancer stemlike cells directly isolated from human larynx cancer specimens. BMC Cancer 16(1):853 PuissegurMP, Mazure NM, Bertero T, Pradelli L, Grosso S, Robbe- Sermesant K et al, 2011, miR-210 is overexpressed in late stages of lung cancer and mediates mitochondrial alterations associated with modulation of HIF-1 activity. Cell Death Differ 18(3):465–478 Camps C, Buffa FM, Colella S, Moore J, Sotiriou C, Sheldon H, Harris AL, Gleadle JM, Ragoussis J, 2008, Hsa-miR-210 is induced by hypoxia and is an independent prognostic factor in breast cancer. Clin Cancer Res 14(5):1340–1348 Gee HE, Camps C, Buffa FM, Patiar S,Winter SC, Betts G, Homer J, Corbridge R, Cox G, West CM, Ragoussis J, Harris AL, 2010, Hsa-mir-210 is a marker of tumor hypoxia and a prognostic factor in head and neck cancer. Cancer 116(9):2148–2158 Fasanaro P, D'Alessandra Y, Di Stefano V, Melchionna R, Romani S, Pompilio G et al, 2008, MicroRNA-210 modulates endothelial cell response to hypoxia and inhibits the receptor tyrosine kinase ligand Ephrin-A3. J Biol Chem 283(23):15878–15883 HockelM, Vaupel P, 2001, Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 93(4): 266–276 Okunieff P, Hoeckel M, Dunphy EP, Schlenger K, Knoop C,Vaupel P, 1993, Oxygen tension distributions are sufficient to explain the local response of human breast tumors treated with radiation alone. Int J Radiat Oncol Biol Phys 26(4):631–636 Yang X, Zhu H, Ge Y, Liu J, Cai J, Qin Q, Zhan L, Zhang C, Xu L, Liu Z, Yang Y, Yang Y, Ma J, Cheng H, Sun X, 2014, Melittin enhances radiosensitivity of hypoxic head and neck squamous cell carcinoma by suppressing HIF-1alpha. Tumor Biol 35(10):10443– 10448 Moeller BJ, Dreher MR, Rabbani ZN, Schroeder T, Cao Y, Li CY, Dewhirst MW, 2005, Pleiotropic effects of HIF-1 blockade on tumor radiosensitivity. Cancer Cell 8(2):99–110 Liu J, Zhang J,Wang X, Li Y, Chen Y, Li K, Zhang J, Yao L, Guo G, 2010, HIF-1 and NDRG2 contribute to hypoxia-induced radioresistance of cervical cancer Hela cells. Exp Cell Res 316(12):1985–1993 Dong G, Chen Q, Jiang F, Yu D,Mao Q, XiaW, Shi R,Wang J, Xu L, 2016, Diisopropylamine dichloroacetate enhances radiosensitization in esophageal squamous cell carcinoma by increasing mitochondria-derived reactive oxygen species levels. Oncotarget 7(42):68170–68178 Lall R, Ganapathy S, Yang M, Xiao S, Xu T, Su H, Shadfan M, Asara JM, Ha CS, Ben-Sahra I, Manning BD, Little JB, Yuan ZM, 2014, Low-dose radiation exposure induces a HIF-1-mediated adaptive and protective metabolic response. Cell Death Differ 21(5):836–844 Ferlay J, Soerjomataram I, Dikshit R, Eser S,Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386 Rosner B, 2011, Fundamentals of biostatistics. Brooks/Cole, Cengage Learning, Boston Katabi N, Lewis JS, 2017, Update from the 4th edition of theWorld Health Organization classification of head and neck Tumours: what is new in the 2017 WHO blue book for tumors and tumor-like lesions of the neck and lymph nodes. Head Neck Pathol 11(1): 48–54 El-Naggar AK, Chan JKC, Grandis JR, Takata T, 2017, Slootweg PJ. WHO Classification of Head and Neck Tumours, International Agency for Research on Cancer Guimaraes TA, Farias LC, Fraga CA, Feltenberger JD, Melo GA, Coletta RD, Souza Santos SH, de Paula AMB, Guimaraes AL, 2016, Evaluation of the antineoplastic activity of gallic acid in oral squamous cell carcinoma under hypoxic conditions. Anti-Cancer Drugs 27(5):407–416 Helman LJ, Gazdar AF, Park JG, Cohen PS, Cotelingam JD, Israel MA, 1988, Chromogranin a expression in normal and malignant human tissues. J Clin Invest 82(2):686–690 Domingos PLB, Souza MG, Guimaraes TA, Santos ES, Farias LC, de Carvalho Fraga CA et al, 2017, Hypoxia reduces the E-cadherin expression and increases OSCC cell migration regardless of the Ecadherin methylation profile. Pathol Res Pract 213(5):496–501 Aragao AZ, Belloni M, Simabuco FM, Zanetti MR, Yokoo S, Domingues RR et al, 2012, Novel processed form of syndecan-1 shed from SCC-9 cells plays a role in cell migration. PLoS One 7(8):e43521 Xia J, Wishart DS, 2010, MSEA: a web-based tool to identify biologically meaningful patterns in quantitative metabolomic data. Nucleic Acids Res 38(Web Server issue):W71–W77 Xia J, Sinelnikov IV, Han B, Wishart DS, 2015, MetaboAnalyst 3.0–making metabolomics more meaningful. Nucleic Acids Res 43(W1):W251–W257 Vaupel P, Harrison L, 2004, Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist 9(Suppl 5):4–9 Schneider CA, Rasband WS, Eliceiri KW, 2012, NIH image to ImageJ: 25 years of image analysis. Nat Meth 9(7):671–675 Mithani SK, Mydlarz WK, Grumbine FL, Smith IM, Califano JA, 2007, Molecular genetics of premalignant oral lesions. Oral Dis 13(2):126–133 Speksnijder CM, van der Glas HW, van der Bilt A, van Es RJ, van der Rijt E, Koole R, 2010, Oral function after oncological intervention in the oral cavity: a retrospective study. Journal of oral and maxillofacial surgery : official journal of the American Association of Oral and Maxillofacial Surgeons 68(6):1231–1237 Coutinho-Camillo CM, Lourenco SV, de Araujo LL, Kowalski LP, Soares FA, 2015, Expression of apoptosis-regulating miRNAs and target mRNAs in oral squamous cell carcinoma. Cancer Genet 208(7–8):382–389 Scapoli L, Palmieri A, Lo Muzio L, Pezzetti F, Rubini C, Girardi A et al, 2010, MicroRNA expression profiling of oral carcinoma identifies new markers of tumor progression. Int J Immunopathol Pharmacol 23(4):1229–1234 Golias T, Papandreou I, Sun R, Kumar B, Brown NV, Swanson BJ, Pai R, Jaitin D, le QT, Teknos TN, Denko NC, 2016, Hypoxic repression of pyruvate dehydrogenase activity is necessary for metabolic reprogramming and growth of model tumours. Sci Rep 6:31146 Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC, 2006, HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab 3(3):187–197 De Saedeleer CJ, Copetti T, Porporato PE, Verrax J, Feron O, Sonveaux P, 2012, Lactate activates HIF-1 in oxidative but not in Warburg-phenotype human tumor cells. PLoS One 7(10):e46571 Saito K, Matsumoto S, Takakusagi Y, Matsuo M,Morris HD, Lizak MJ,Munasinghe JP, Devasahayam N, Subramanian S,Mitchell JB, Krishna MC, 2015, 13C-MR spectroscopic imaging with hyperpolarized [1-13C]pyruvate detects early response to radiotherapy in SCC tumors and HT-29 tumors. Clinical cancer research : an official journal of the American Association for Cancer Research 21(22):5073–5081 Hosokawa Y, Okumura K, Terashima S, Sakakura Y, 2012, Radiation protective effect of hypoxia-inducible factor-1alpha, HIF-1alpha, on human oral squamous cell carcinoma cell lines. Radiat Prot Dosimetry 152(1–3):159–163 Leung E, Cairns RA, Chaudary N, Vellanki RN, Kalliomaki T, Moriyama EH, Mujcic H, Wilson BC, Wouters BG, Hill R, Milosevic M, 2017, Metabolic targeting of HIF-dependent glycolysis reduces lactate, increases oxygen consumption and enhances response to high-dose single-fraction radiotherapy in hypoxic solid tumors. BMC Cancer 17(1):418 Koukourakis MI, Giatromanolaki A, 2018, Warburg effect, lactate dehydrogenase, and radio/chemo-therapy efficacy. Int J Radiat Biol:1–19. , DOI 10.1080/09553002.2018.1490041 Jackson SP, Bartek J, 2009, The DNA-damage response in human biology and disease. Nature 461(7267):1071–1078 Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, Formenti SC, 2004, Ionizing radiation inhibition of distant untreated tumors, abscopal effect, is immune mediated. Int J Radiat Oncol Biol Phys 58(3):862–870 Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM et al, 2010, Inhibition of lactate dehydrogenase a induces oxidative stress and inhibits tumor progression. Proc Natl Acad Sci U S A 107(5):2037–2042 Serganova I, Cohen IJ, Vemuri K, Shindo M, Maeda M, Mane M, Moroz E, Khanin R, Satagopan J, Koutcher JA, Blasberg R, 2018, LDH-A regulates the tumor microenvironment via HIF-signaling and modulates the immune response. PLoS One 13(9):e0203965 Herskind C, Ma L, Liu Q, Zhang B, Schneider F, Veldwijk MR, Wenz F, 2017, Biology of high single doses of IORT: RBE, 5 R's, and other biological aspects. Radiat Oncol 12(1):24 Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS, Formenti SC, Demaria S, 2009, Fractionated but not singledose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clinical cancer research : an official journal of the American Association for Cancer Research. 15(17):5379–5388 Golden EB, Chhabra A, Chachoua A, Adams S, Donach M, Fenton-Kerimian M, Friedman K, Ponzo F, Babb JS, Goldberg J, Demaria S, Formenti SC, 2015, Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. Lancet Oncol 16(7):795–803 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 433 EP 442 DI 10.1007/s12253-018-0517-2 PG 10 ER PT J AU Li, Y Bai, W Zhang, L AF Li, Yuanhang Bai, Weijun Zhang, Linlin TI The Overexpression of CD80 and ISG15 Are Associated with the Progression and Metastasis of Breast Cancer by a Meta-Analysis Integrating Three Microarray Datasets SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Microarray data; Meta-analysis; Breast cancer; Metastasis ID Microarray data; Meta-analysis; Breast cancer; Metastasis AB Breast cancer is a common cancer and could result in a substantial mortality. The study aimed to screen gene signatures associated with the development and metastasis of breast cancer and explore their regulation mechanisms. Three datasets of GSE10797, GSE8977 and GSE3744 were downloaded from GEO (Gene Expression Omnibus) database, containing 55 breast cancer samples and 27 normal samples. After data preprocessing using limma software and RMA (robust multiarray average) algorithm, DEGs (differentially expressed genes) between breast tumor and normal tissues in three individual experiments were identified using MADAM package. Function and pathway enrichment analyses were performed for the DEGs. Transcription factors and TAGs (tumor associated genes) among the DEGs were recognized and the PPI (protein-protein-interaction) network for the DEGs was constructed using Cytoscape software. The mRNA expression was analyzed via real-time quantitative PCR and protein expression was measured by western blotting. Totally, 100 DEGs were identified, including 33 up-regulated genes and 67 down-regulated genes. Among them, up-regulated DEGs such as CD80 was enriched in toll-like receptor (TLR) interaction pathway and the TAG, ISG15 was related to RIG-I-like receptor signaling pathway, while CXCL10 was involved in both of the two pathways. Whereas, the down-regulated DEG, CXCL12 was significantly associated with axon guidance pathway. Additionally, these DEGs were also pivotal nodes in the PPI network with high degrees. Besides, CXCL10 and CD80 were both interacted with IFNG. The mRNA expression of ISG15 was obviously enhanced in human breast cancer cells MCF-7, while no significant difference of CXCL10 mRNA level was found between MCF10A and MCF-7 cells. Moreover, the proteins expression levels of CD80 and ISG15 were significantly increased in MCF-7, MDA-MB-468 and MDA-MB-231 breast cancer cells than in normal MCF10A cells. CD80 might be responsible for the breast cancer’s progression and metastasis via regulating innate immune system. In addition, ISG15 is identified as a crucial gene signature associated with breast cancer development and metastasis via RIGI-like receptor signaling pathway. C1 [Li, Yuanhang] Cancer Hospital of China Medical University, Medical Department, No. 44 Xiaoheyan Road, Dadong District, 110042 Shenyang, Liaoning Province, China. [Bai, Weijun] Cancer Hospital of China Medical University, Medical Department, No. 44 Xiaoheyan Road, Dadong District, 110042 Shenyang, Liaoning Province, China. [Zhang, Linlin] Cancer Hospital of China Medical University, Medical Department, No. 44 Xiaoheyan Road, Dadong District, 110042 Shenyang, Liaoning Province, China. RP Li, Y (reprint author), Cancer Hospital of China Medical University, Medical Department, 110042 Shenyang, China. EM yuanhangli619@hotmail.com CR Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF, 2003, Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci 100:3983–3988 Arthur A, Shi S, Zannettino AC, Fujii N, Gronthos S, Koblar SA, 2009, Implanted adult human dental pulp stem cells induce endogenous axon guidance. Stem Cells 27:2229–2237 AshburnerMet al, 2000, Gene Ontology: tool for the unification of biology. Nat Genet 25:25–29 Bektas N et al, 2008, The ubiquitin-like molecule interferonstimulated gene 15, ISG15, is a potential prognostic marker in human breast cancer. Breast Cancer Res 10:R58 Boyle P, Levin B, 2008, World cancer report 2008. International Agency for Research on Cancer Lyon, France:330 Casey T et al, 2009, Molecular signatures suggest a major role for stromal cells in development of invasive breast cancer. Breast Cancer Res Treat 114:47–62 Chen X, Armstrong MA, Li G, 2006, Mesenchymal stem cells in immunoregulation. Immunol Cell Biol 84:413–421 Chen K, Huang J, Gong W, Iribarren P, Dunlop NM, Wang JM, 2007, Toll-like receptors in inflammation, infection and cancer. Int Immunopharmacol 7:1271–1285 Chen J-S, Hung W-S, Chan H-H, Tsai S-J, Sun HS, 2013, In silico identification of oncogenic potential of fyn-related kinase in hepatocellular carcinoma. Bioinformatics 29:420–427 Cui X-F, Imaizumi T, Yoshida H, Borden EC, Satoh K, 2004, Retinoic acid-inducible gene-I is induced by interferon-γ and regulates the expression of interferon-γ stimulated gene 15 in MCF-7 cells. Biochem Cell Biol 82:401–405 Datta D et al, 2006, Ras-induced modulation of CXCL10 and its receptor splice variant CXCR3-B in MDA-MB-435 and MCF-7 cells: relevance for the development of human breast cancer. Cancer Res 66:9509–9518 Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA, 2003, DAVID: database for annotation, visualization, and integrated discovery. Genome Biol 4:P3 Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, Luster AD, 2002, IFN-γ-inducible protein 10, IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking. J Immunol 168:3195–3204 Franceschini A et al, 2013, STRING v9. 1: protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res 41:D808–D815 Gautier L, Cope L, Bolstad BM, Irizarry RA, 2004, Affy—analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 20: 307–315 Goldberg-Bittman L, Neumark E, Sagi-Assif O, Azenshtein E, Meshel T, Witz IP, Ben-Baruch A, 2004, The expression of the chemokine receptor CXCR3 and its ligand, CXCL10, in human breast adenocarcinoma cell lines. Immunol Lett 92:171–178 Hardaker EL et al, 2004, Regulation of TNF-α-and IFN-γ-induced CXCL10 expression: participation of the airway smooth muscle in the pulmonary inflammatory response in chronic obstructive pulmonary disease. FASEB J 18:191–193 Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP, 2003, Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4:249–264 Kanehisa M, Goto S, 2000, KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res 28:27–30 Karnoub AE et al, 2007, Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 449:557–563 Kobayashi K, Hernandez LD, Galan JE, Janeway CAJr, Medzhitov R, Flavell RA, 2002, IRAK-M is a negative regulator of toll-like receptor signaling. Cell 110:191–202 Kohl M, Wiese S, Warscheid B, 2011, Cytoscape: software for visualization and analysis of biological networks. In: Data Mining in Proteomics. Springer, pp 291–303 Kugler KG, Mueller LA, Graber A, 2010, MADAM-an open source meta-analysis toolbox for R and bioconductor. Source Code Biol Med 5:3 Liu R et al, 2007, The prognostic role of a gene signature from tumorigenic breast-cancer cells. N Engl J Med 356:217–226 Ma X-J, Dahiya S, Richardson E, Erlander M, Sgroi DC, 2009, Gene expression profiling of the tumor microenvironment during breast cancer progression. Breast Cancer Res 11:R7 Rhodes DR, Barrette TR, Rubin MA, Ghosh D, Chinnaiyan AM, 2002, Meta-analysis of microarrays interstudy validation of gene expression profiles reveals pathway dysregulation in prostate cancer. Cancer Res 62:4427–4433 Richardson AL et al, 2006, X chromosomal abnormalities in basallike human breast cancer. Cancer Cell 9:121–132 Ross JS et al, 2003, The Her-2/neu gene and protein in breast cancer 2003: biomarker and target of therapy. Oncologist 8:307– 325 Rutkowski R, Moniuszko T, Stasiak-Barmuta A, Kosztyla-Hojna B, AlifierM, Rutkowski K, Tatarczuk-Krawiel A, 2003, CD80 and CD86 expression on LPS-stimulated monocytes and the effect of CD80 and CD86 blockade on IL-4 and IFN-gamma production in Nanotopic bronchial asthma. Arch Immunol Ther Exp 51:421–428 Sant M et al, 2003, Stage at diagnosis is a key explanation of differences in breast cancer survival across Europe. Int J Cancer 106:416–422 Schreibelt G, Tel J, Sliepen KH, Benitez-Ribas D, Figdor CG, Adema GJ, de Vries IJM, 2010, Toll-like receptor expression and function in human dendritic cell subsets: implications for dendritic cell-based anti-cancer immunotherapy. Cancer Immunol Immunother 59:1573–1582 Siegel R, Ma J, Zou Z, Jemal A, 2014, Cancer statistics, 2014. CA Cancer J Clin 64:9–29 SmidMet al, 2006, Genes associated with breast cancer metastatic to bone. J Clin Oncol 24:2261–2267 Turnbull C et al, 2010, Genome-wide association study identifies five new breast cancer susceptibility loci. Nat Genet 42:504–507 Zhao M, Sun J, Zhao Z, 2013, TSGene: a web resource for tumor suppressor genes. Nucleic Acids Res 41:D970–D976 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 443 EP 452 DI 10.1007/s12253-018-0478-5 PG 10 ER PT J AU Lee, S Ahn, YM Kim, JY Cho, YE Park, JH AF Lee, Sun Ahn, Yong-Min Kim, Jee-Youn Cho, Young-Eun Park, Jae-Hoon TI Downregulation of NOP53 Ribosome Biogenesis Factor Leads to Abnormal Nuclear Division and Chromosomal Instability in Human Cervical Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NOP53; Mitosis; Abnormal nucleus; Chromosomal instability ID NOP53; Mitosis; Abnormal nucleus; Chromosomal instability AB NOP53 ribosome biogenesis factor (NOP53) is a nucleolar protein involved in oncogenesis/tumor suppression, cell cycle regulation, and cell death. Here, we investigated the role of NOP53 in the maintenance of normal nuclear shape and chromosomal stability. Depletion of NOP53 by shRNA caused abnormal nuclear morphology, including large nucleus, irregular nucleus, and multinucleated cells, and chromosomal instability resulting in micronucleus or nuclear bud formation. The abnormal nuclear shape and chromosomal instability were restored by re-expression of NOP53. We further showed that NOP53 was involved in chromosome congression in metaphase. Downregulation of NOP53 induced aberrant chromosome congression and spindle checkpoint activation, resulting in delayed mitosis and mitotic arrest. Thus, our findings demonstrated that the nucleolar protein NOP53 participated in mitotic progression and that dysregulated NOP53 expression caused chromosomal instability in cancer cells. C1 [Lee, Sun] Kyung Hee University, College of Medicine, Department of Pathology, 02453 Seoul, South Korea. [Ahn, Yong-Min] Kyung Hee University, College of Medicine, Department of Pathology, 02453 Seoul, South Korea. [Kim, Jee-Youn] Kyung Hee University, College of Medicine, Department of Pathology, 02453 Seoul, South Korea. [Cho, Young-Eun] Kyung Hee University, College of Medicine, Department of Pathology, 02453 Seoul, South Korea. [Park, Jae-Hoon] Kyung Hee University, College of Medicine, Department of Pathology, 02453 Seoul, South Korea. RP Park, JH (reprint author), Kyung Hee University, College of Medicine, Department of Pathology, 02453 Seoul, South Korea. EM jhpark@khu.ac.kr CR Boulon S,Westman BJ, Hutten S, Boisvert FM, Lamond AI, 2010, The nucleolus under stress. Mol Cell 40:216–227 Amin MA, Matsunaga S, Uchiyama S, Fukui K, 2008, Depletion of nucleophosmin leads to distortion of nucleolar and nuclear structures in HeLa cells. Biochem J 415:345–351 Amin MA, Matsunaga S, Ma N, Takata H, Yokoyama M, Uchiyama S, Fukui K, 2007, Fibrillarin, a nucleolar protein, is required for normal nuclear morphology and cellular growth in HeLa cells. Biochem Biophys Res Commun 360:320–326 Kalt I, Levy A, Borodianskiy-Shteinberg T, Sarid R, 2012, Nucleolar localization of GLTSCR2/PICT-1 is mediated by multiple unique nucleolar localization sequences. PLoS One 7:e30825 Kim JY, Seok KO, Kim YJ, Bae WK, Lee S, Park JH, 2011, Involvement of GLTSCR2 in the DNA damage response. Am J Pathol 179:1257–1264 Lee S, Kim JY, Kim YJ, Seok KO, Kim JH, Chang YJ, Kang HY, Park JH, 2012, Nucleolar protein GLTSCR2 stabilizes p53 in response to ribosomal stresses. Cell Death Differ 19:1613–1622 Yim JH, Kim YJ, Ko JH, Cho YE, Kim SM, Kim JY, Lee S, Park JH, 2007, The putative tumor suppressor gene GLTSCR2 induces PTEN-modulated cell death. Cell Death Differ 14:1872–1879 Shibamoto Y, Streffer C, Fuhrmann C, Budach V, 1991, Tumor radiosensitivity prediction by the cytokinesis-block micronucleus assay. Radiat Res 128:293–300 Kim JY, Cho YE, Park JH, 2015, The nucleolar protein GLTSCR2 is an upstream negative regulator of the oncogenic nucleophosmin- MYC axis. Am J Pathol 185:2061–2068 Ugrinova I, Monier K, Ivaldi C, Thiry M, Storck S, Mongelard F, Bouvet P, 2007, Inactivation of nucleolin leads to nucleolar disruption, cell cycle arrest and defects in centrosome duplication. BMC Mol Biol 8:66 Ma N, Matsunaga S, Takata H, Ono-Maniwa R, Uchiyama S, Fukui K, 2007, Nucleolin functions in nucleolus formation and chromosome congression. J Cell Sci 120:2091–2105 Tsai RY, Pederson T, 2014, Connecting the nucleolus to the cell cycle and human disease. FASEB J 28:3290–3296 Takada H, Kurisaki A, 2015, Emerging roles of nucleolar and ribosomal proteins in cancer, development, and aging. Cell Mol Life Sci 72:4015–4025 Amin MA, Matsunaga S, Uchiyama S, Fukui K, 2008, Nucleophosmin is required for chromosome congression, proper mitotic spindle formation, and kinetochore-microtubule attachment in HeLa cells. FEBS Lett 582:3839–3844 Maraldi NM, Lattanzi G, Capanni C, Columbaro M, Merlini L, Mattioli E, Sabatelli P, Squarzoni S, Manzoli FA, 2006, Nuclear envelope proteins and chromatin arrangement: a pathogenic mechanism for laminopathies. Eur J Histochem 50:1–8 Vogt E, Kirsch-Volders M, Parry J, Eichenlaub-Ritter U, 2008, Spindle formation, chromosome segregation and the spindle checkpoint in mammalian oocytes and susceptibility to meiotic error. Mutat Res 651:14–29 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 453 EP 459 DI 10.1007/s12253-018-0531-4 PG 7 ER PT J AU Farsani, AM Kamel, M Mehrpouri, M Heris, ShR Hamidpour, M Salari, S Mohamadi, HM AF Farsani, Allahbakhshian Mehdi Kamel, Masomeh Mehrpouri, Mahdieh Heris, Shiri Reza Hamidpour, Mohsen Salari, Sina Mohamadi, Hosien Mohamad TI The Expression of Interferon Gamma (IFN-γ) and Interleukin 6 (IL6) in Patients with Acute Lymphoblastic Leukemia (ALL) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ALL; IFN-γ; IL-6 ID ALL; IFN-γ; IL-6 AB Interferon gamma (IFN-γ) and interleukin 6 (IL-6) are including the most important cytokines which have been associated with the biological behavioral and immune responses in malignancies. Based on the critical roles which these two cytokines play against tumor cells, the present study was aimed to investigate the genes expression level of IL6 and IFN-γ in patients with Acute Lymphoblastic Leukemia and compare with normal controls. Fifty-two patients with ALL and 13 healthy volunteer were under studied. The peripheral blood mononuclear cells of all patients and normal controls were separated by ficoll. The expression of interferon gamma and interleukin 6 genes were determined by RQ-PCR. Finally all data were analyzes using T student, one way ANOVA and Mann-Whitney tests were use to analyze all samples data. Our finding showed that the level of IFN-γ gene expression was significant decreased in patients with All as compared with healthy controls (83 change fold, p < 0.0001). The level of IL-6 Gene expression was not changeable in B-ALL patients as compared with healthy control (p = 0.4), but in T-ALL patients, was significantly reduced (p < 0.01). The results of present study indicated that IFN-γ gene expression reduced in ALL patients. It provides a valuable insight that immune system may disrupted in patients with ALL, which cause tumor cells escape from immune surveillance. C1 [Farsani, Allahbakhshian Mehdi] Shahid Beheshti University of Medical Sciences, School of Allied Medical Sciences, Department of Hematology and Blood BankingTehran, Iran. [Kamel, Masomeh] Shahid Beheshti University of Medical Sciences, School of Allied Medical Sciences, Department of Hematology and Blood BankingTehran, Iran. [Mehrpouri, Mahdieh] Shahid Beheshti University of Medical Sciences, School of Allied Medical Sciences, Department of Hematology and Blood BankingTehran, Iran. [Heris, Shiri Reza] Shahid Beheshti University of Medical Sciences, School of Allied Medical Sciences, Department of Hematology and Blood BankingTehran, Iran. [Hamidpour, Mohsen] Shahid Beheshti University of Medical Sciences, School of Allied Medical Sciences, Department of Hematology and Blood BankingTehran, Iran. [Salari, Sina] Shahid Beheshti University of Medical Sciences, Taleghani General Hospital, Department of Internal MedicineTehran, Iran. [Mohamadi, Hosien Mohamad] Shahid Beheshti University of Medical Sciences, School of Allied Medical Sciences, Department of Hematology and Blood BankingTehran, Iran. RP Hamidpour, M (reprint author), Shahid Beheshti University of Medical Sciences, School of Allied Medical Sciences, Department of Hematology and Blood Banking, Tehran, Iran. EM mohsenhp@sbmu.ac.ir CR Harrison CJ, 2013, Targeting signaling pathways in acute lymphoblastic leukemia: new insights. American Society of Hematology, Education Program 2013(1):118–125 Cardoso B, GirioA, Henriques C,Martins L, Santos C, SilvaAet al, 2008, Aberrant signaling in T-cell acute lymphoblastic leukemia: biological and therapeutic implications. Braz J Med Biol Res 41(5): 344–350 Brady MT, Lee J, Ferrone S et al, 2011, Interferon-γ secretion by t(9;22, acute lymphoblastic leukemia- derived dendritic cells. American Society of Hematology, Education Program 35(2):275– 277 Kholoussi SM, Bayoumi FS, El-Nady H, 2008, Estimation of serum interferon-gamma level in childhood acut lymphoblastic leukemia patients. J Med Sci 8(1):68–72 Marth C, Fiegl H, Zeimet AGet al, 2004, Interferon-g expression is an independent prognostic factor in ovarian cancer. Am J Obstet Gynecol 191:1598–1605 Tian G, Mi J, Wei X, Zhao D, Qiao L, Yang C, Li X et al, 2015, Circulating interleukin-6 and cancer: a meta-analysis using Mendelian randomization. Sci Rep Heikkila K, Ebrahim S, Lawlor DA, 2008, Systematic review of the association between circulating interleukin-6, IL-6, and cancer. Eur J Cancer 44:937–945 Sugiyama H, Inoue K, Ogawa H, Yamagami T, Soma T, Miyake S, Hirata M, Kishimoto T, 1995, The expression of IL-6 and its related genes in acute leukemia. Leuk Lymphoma 21:1 Heinrich PC, Behrmann I, Haan S, Hermanns HM, Muller-Newen G, Schaper F, 2003, Principles of interleukin, IL)-6-type cytokine signalling and its regulation. J Biochem 374(1):1–20 Bauvois B, Nguyen J, Tang R, Billard C, Kolb J-P, 2009, Types I and II interferon supregulate the co-stimulatory CD80 molecule in monocytes via interferon regulatory factor. Biochem Pharmacol 8(5):514–522 Sato A, Ohtsuki M, HataM et al, 2006, Antitumor activity of IFNin murine tumor models. J Immunol 176:7686–7694 Fujii H, Trudeau JD, Teachey DT, Fish JD, Stephan A et al, 2007, In vivo control of acute lymphoblastic leukemia by immuno stimulatory Cp Goligonucleotides. Blood 1090:5.2008–5.2013 Yin HQ, Qiao ZH, Zhu L, Zhang L, Su LP, Lu YJ, 2006, Levels of intracellular IL-6 and IFN-gamma in children with acute lymphoblastic leukemia. American Society of Hematology, Education Program 8(6):461–463 HeePark H, Kim M, Lee B-H et al, 2006, Intracellular IL-4, IL-10, and IFN-γ levels of leukemic cells and bone marrow Tcells in acute leukemia. Ann Clin Lab Sci 36:15–17 Zhang X-I, Komada Y, Joe C, 2000, Intracellular cytokine pro®le of T cells with acute lymphoblastic leukemia. Cancer Immunol Immunother 49(3):165–172 Strander H, 1986, Interferon treatment of human neoplasia. Adv Cancer Res 46:1 De Maeyer E, De Maeyer-Guignard J, 1988, Interferons and other regulatory cytokines. Wiley, New York, pp 134–334 Grander D, Heyman M, Brondum-Nielsen K, Liu Y, Lundgren E, Soderhall S, Einhorn S, 1992, Interferon system in primary acute lymphocytic leukemia cells with or without deletions of the − 0.5). The mean survival time was 72.7 ± 7.82 months. Survival rates for 1, 3 and 5 years were 93%, 65% and 45%, respectively. The mean LODDS value was −1.0466 ± 0.51. LODDS subgroups show strong correlation with Overall Survival (OS). The mean survival were 114.8, 81.8, 56.6 and 25.6 months in LODDS subgroups 1, 2, 3 and 4, respectively (Log-rank; p = 0.002), in addition LOODS values shows correlation with perineural invasion and micro vascular invasion (p = 0.015 and p = 0.001 respectively). Findings in our patient group support the hypothesis that LODDS subgroups correlate with OS, and that value of LODDS has considerable role in prediction of OS as well. C1 [Agalar, Cihan] Dokuz Eylul University, Faculty of Medicine, Department of General Surgery, Balcova, Inciralti, 35340 Izmir, Turkey. [Aysal, Anil] Dokuz Eylul University, Faculty of Medicine, Department of PathologyIzmir, Turkey. [Unek, Tarkan] Dokuz Eylul University, Faculty of Medicine, Department of General Surgery, Balcova, Inciralti, 35340 Izmir, Turkey. [Egeli, Tufan] Dokuz Eylul University, Faculty of Medicine, Department of General Surgery, Balcova, Inciralti, 35340 Izmir, Turkey. [Ozbilgin, Mucahit] Dokuz Eylul University, Faculty of Medicine, Department of General Surgery, Balcova, Inciralti, 35340 Izmir, Turkey. [Akturk, Nesrin] Dokuz Eylul University, Faculty of Medicine, Department of Radiation OncologyIzmir, Turkey. [Semiz, Salih Huseyin] Dokuz Eylul University, Institute of OncologyIzmir, Turkey. [Unek, Tugba] Dokuz Eylul University, Institute of OncologyIzmir, Turkey. [Akarsu, Mesut] Dokuz Eylul University School of Medicine, The Department of GastroenterologyIzmir, Turkey. [Soyturk, Mujde] Dokuz Eylul University School of Medicine, The Department of GastroenterologyIzmir, Turkey. [Ellidokuz, Hulya] Dokuz Eylul University, Institute of OncologyIzmir, Turkey. [Sagol, Ozgul] Dokuz Eylul University, Faculty of Medicine, Department of PathologyIzmir, Turkey. [Astarcioglu, Ibrahim] Dokuz Eylul University, Faculty of Medicine, Department of General Surgery, Balcova, Inciralti, 35340 Izmir, Turkey. RP Agalar, C (reprint author), Dokuz Eylul University, Faculty of Medicine, Department of General Surgery, 35340 Izmir, Turkey. EM cihan.agalar@deu.edu.tr CR Albores-Saavedra J, Schwartz AM, Batich K, Henson DE, 2009, Cancers of the ampulla of vater: demographics, morphology, and survival based on 5,625 cases from the SEER program. J Surg Oncol 100:598–605 BenhamicheAM, Jouve JL, Manfredi S, Prost P, Isambert N, Faivre J, 2000, Cancer of the ampulla of Vater: results of a 20-year population- based study. Eur J Gastroenterol Hepatol 12:75–79 Beger HG, Treitschke F, Gansauge F, Harada N, Hiki N,Mattfeld T, 1999, Tumor of the ampulla of Vater: experience with local or radical resection in 171 consecutively treated patients. Arch Surg 134:526–532 O’Connell JB,MaggardMA,Manunga J et al, 2008, Survival after resection of Ampullary carcinoma: a National Population-Based Study. Ann Surg Oncol 15:1820–1827 Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, 2010, AJCC Cancer staging manual, 7th edn. Springer, New York Falconi M, Crippa S, Dominguez I et al, 2008, Prognostic relevance of lymph node ratio and number of resected nodes after curative resection of ampulla of Vater carcinoma. Ann Surg Oncol 15:3178–3186 Sierzega M, Nowak K, Kulig J, Matyja A, Nowak W, Popiela T, 2009, Lymph node involvement in ampullary cancer: the importance of the number, ratio, and location of metastatic nodes. J Surg Oncol 100:19–24 Balci S, Basturk O, Saka B et al, 2015, Substaging nodal status in Ampullary carcinomas has significant prognostic value: proposed revised staging based on an analysis of 313 well-characterized cases. Ann Surg Oncol 22:4392–4401 Kang HJ, Eo SH, Kim SC et al, 2014, Increased number of metastatic lymph nodes in adenocarcinoma of the ampulla of Vater as a prognostic factor: a proposal of new nodal classification. Surgery 155:74–84 AminM.B, Edge S, Greene F, et al. AJCC Cancer Staging Manual, 8th. Edition, Chicago. 2017 Sun Z, Xu Y, Li DMet al, 2010, Log odds of positive lymph nodes: a novel prognostic indicator superior to the number-based and the ratio-based n category for gastric cancer patients with R0 resection. Cancer 116:2571–2580 Wang J, Hassett JM, Dayton MT, Kulaylat MN, 2008, The prognostic superiority of log odds of positive lymph nodes in stage III colon cancer. J Gastrointest Surg 12:1790–1796 Chen LJ, Chung KP, Chang YJ, Chang YJ, 2015, Ratio and log odds of positive lymph nodes in breast cancer patients with mastectomy. Surg Oncol 24:239–247 Huang B, Chen C, Ni M, Mo S, Cai G, Cai S, 2016, Log odds of positive lymph nodes is a superior prognostic indicator in stage III rectal cancer patients: a retrospective analysis of 17,632 patients in the SEER database. Int J Surg 32:24–30 Lahat G, Lubezky N, Gerstenhaber F et al, 2016, Number of evaluated lymph nodes and positive lymph nodes, lymph node ratio, and log odds evaluation in early-stage pancreatic ductal adenocarcinoma: numerology or valid indicators of patient outcome?World J Surg Onc 14:254 Manabe T, Ohshio G, Baba N et al, 1989, Radical pancreatectomy for ductal cell carcinoma of the head of the pancreas. Cancer 64: 1132–1137 Tol JA, Gouma DJ, Bassi C et al, 2014, Definition of a standard lymphadenectomy in surgery for pancreatic ductal adenocarcinoma: a consensus statement by the international study group on pancreatic surgery, ISGPS). Surgery 156:591–600 Clavien PA, Barkun J, de Oliveira ML et al, 2009, The Clavien- Dindo classification of surgical complications: five-year experience. Ann Surg 250:187–196 Bassi C, Dervenis C, Butturini G et al, 2005, Postoperative pancreatic fistula: an international study group, ISGPF, definition. Surgery 138:8–13 Louvet C, Andre T, Hammel P et al, 2001, Phase II trial of bimonthly leucovorin, 5-fluorouracil and gemcitabine for advanced pancreatic adenocarcinoma, FOLFUGEM). Ann Oncol 12:675–679 Goodman KA, Regine WF, Dawson LA et al, 2012, Radiation therapy oncology group consensus panel guidelines for the delineation of the clinical target volume in the postoperative treatment of pancreatic head cancer. Int J Radiat Oncol Biol Phys 83:901–908 Arslan NC, Sokmen S, Canda AE, Terzi C, Sarioglu S, 2014, The prognostic impact of the log odds of positive lymph nodes in colon cancer. Color Dis 16:386–392 Chen S-C, Shyr Y-M, Chou S-C, Wang S-E, 2015, The role of lymph nodes in predicting the prognosis of ampullary carcinoma after curative resection. World J Surg Onc 13:224 Sessa F, Furlan D, Zampatti C et al, 2007, Prognostic factors for ampullary adenocarcinomas: tumor stage, tumor histology, tumor location, immunohistochemistry and microsatellite instability. Virchows Arch 451:649–657 Showalter TN, Zhan T, Anne PR, Carnevali I, Franzi F, Capella C, 2011, The influence of prognostic factors and adjuvant Chemoradiation on survival after Pancreaticoduodenectomy for Ampullary carcinoma. J Gastrointest Surg 15:1411–1416 Sakata J, Shirai Y, Wakai T, Ajioka Y, Akazawa K, Hatakeyama K, 2011, Assessment of the nodal status in ampullary carcinoma: the number of positive lymph nodes versus the lymph node ratio.World J Surg 35:2118–2124 Chang DK, Jamieson NB, Johns AL et al, 2013, Histomolecular phenotypes and outcome in adenocarcinoma of the ampulla of vater. J Clin Oncol 31:1348–1356 Choi SB, Kim WB, Song TJ, Suh SO, Kim YC, Choi SY, 2011, Surgical outcomes and prognostic factors for ampulla of vater cancer. Scand J Surg 100:92–98 de Paiva Haddad LB, Patzina RA, Penteado S et al, 2010, Lymph node involvement and not the histophatologic subtype is correlated with outcome after resection of adenocarcinoma of the ampulla of vater. J Gastrointest Surg 14:719–728 Sakata J, Shirai Y, Wakai T et al, 2007, Number of positive lymph nodes independently affects long-term survival after resection in patients with ampullary carcinoma. Eur J Surg Oncol 33:346–351 Sommerville CAM, Limongelli P, PaiMet al, 2009, Survival analysis after pancreatic resection for ampullary and pancreatic head carcinoma: an analysis of clinicopathological factors. J Surg Oncol 100:651–656 Lee JH, Lee KG, Ha TK, Jun YJ, Paik SS, Park HK, 2011, Pattern analysis of lymph nodemetastasis and the prognostic importance of number of metastatic nodes in ampullary adenocarcinoma. Am Surg 77:322–329 Zhou J, Zhang Q, Li P, Shan Y, Zhao D, Cai J, 2013, Prognostic relevance of number and ratio ofmetastatic lymph nodes in resected carcinoma of the ampulla of Vater. Chin J Cancer Res 25:735–742 La Torre M, Nigri G, Petrucciani N et al, 2014, Prognostic assessment of different lymph node staging methods for pancreatic cancer with R0 resection: PN staging, lymph node ratio, log odds of positive lymph nodes. Pancreatology 14:289–294 Kwon J, Kim K, Chie EK et al, 2017, Prognostic relevance of lymph node status for patientswith ampullary adenocarcinoma after radical resection followed by adjuvant treatment. Eur J Surg Oncol 43:1690–1696 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 467 EP 473 DI 10.1007/s12253-019-00584-6 PG 7 ER PT J AU Fang, J Chen, W Meng, XL AF Fang, Jun Chen, Wei Meng, Xiang-Ling TI LncRNA CASC9 Suppressed the Apoptosis of Gastric Cancer Cells through Regulating BMI1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lncRNA CASC9; BMI1; Gastric cancer; Ubiquitination; Degradation ID lncRNA CASC9; BMI1; Gastric cancer; Ubiquitination; Degradation AB Long noncoding RNAs (lncRNAs) play important roles in regulating the apoptosis of gastric cancer (GC) cells. This study aims to investigate the underlying mechanism of lncRNA CASC9 in regulating the apoptosis of GC cells. The expressions of lncRNA and protein in GC tissues and cell lines were detected by qRT-PCR and western blot. GC cell apoptosis was detected by flow cytometry analysis. RNA pull-down and RNA immunoprecipitation (RIP) assays were conducted to verify the interaction between CASC9 and BMI1. LncRNA CASC9 was upregulated in GC tissue and GC cells, and high CASC9 expression was positively correlated with TNM stage and lymph node metastasis. Silencing CASC9 promoted the apoptosis of GC cells. LncRNA CASC9 could interact with BMI1 and positively regulate BMI1 expression. Silencing CASC9 promoted the ubiquitination of BMI1. In addition, lncRNA CASC9 regulated the apoptosis of GC cells through BMI1. Furthermore, interfering CASC9 inhibited the tumor growth of GC. LncRNA CASC9 could interact with BMI1 to regulate the degradation of BMI1, thus to affect the apoptosis of GC cells and suppressed tumor growth. C1 [Fang, Jun] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 218 Jixi Rd., 230022 Hefei, Anhui, China. [Chen, Wei] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 218 Jixi Rd., 230022 Hefei, Anhui, China. [Meng, Xiang-Ling] The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 218 Jixi Rd., 230022 Hefei, Anhui, China. RP Fang, J (reprint author), The First Affiliated Hospital of Anhui Medical University, Department of General Surgery, 230022 Hefei, China. EM 102473526@qq.com CR Siegel RL, Miller KD, Jemal A, 2018, Cancer statistics, 2018. CA Cancer J Clin 68(1):7–30 Ge S et al, 2012, Association between habitual dietary salt intake and risk of gastric Cancer: a systematic review of observational studies. Gastroenterol Res Pract 2012:808120–808120 Seo AN et al, 2019, Exon 9 mutation of PIK3CA associated with poor survival in patients with Epstein-Barr virus-associated gastric Cancer. Anticancer Res 39(4):2145–2154 Aoyama T, Yoshikawa T, 2017, Adjuvant therapy for locally advanced gastric cancer. Surg Today 47(11):1295–1302 Allemani C, Weir HK, Carreira H, Harewood R, Spika D, Wang XS, Bannon F, Ahn JV, Johnson CJ, Bonaventure A, Marcos- Gragera R, Stiller C, Azevedo e Silva G, Chen WQ, Ogunbiyi OJ, Rachet B, Soeberg MJ, You H, Matsuda T, Bielska-Lasota M, Storm H, Tucker TC, Coleman MP, 2015, Global surveillance of cancer survival 1995-2009: analysis of individual data for 25,676, 887 patients from 279 population-based registries in 67 countries, CONCORD-2). Lancet, London, England, 385(9972):977–1010 Wang MW, Liu J, Liu Q, Xu QH, Li TF, Jin S, Xia TS, 2017, LncRNA SNHG7 promotes the proliferation and inhibits apoptosis of gastric cancer cells by repressing the P15 and P16 expression. Eur Rev Med Pharmacol Sci 21(20):4613–4622 Li P, Zhou X, SunW, ShengW, Tu Y, Yu Y, Dong J,Ye B, Zheng Z, Lu M, 2017, Elemene induces apoptosis of human gastric Cancer cell line BGC-823 via extracellular signal-regulated kinase, ERK, 1/2 signaling pathway. Med Sci Monitor 23:809–817 Zhang S, Zhang X, Sun Q, Zhuang C, Li G, Sun L,Wang H, 2019, LncRNA NR2F2-AS1 promotes tumourigenesis through modulating BMI1 expression by targeting miR-320b in non-small cell lung cancer. J Cell Mol Med 23(3):2001–2011 Zhao T, Chen Y, Sheng S, Wu Y, Zhang T, 2018, Upregulating microRNA-498 inhibits gastric cancer proliferation invasion and chemoresistance through inverse interaction of Bmi1. Cancer Gene Ther Chen Y, Lian G, Zhang Q, Zeng L, Qian C, Chen S, Huang K, 2013, Overexpression of Bmi-1 induces the malignant transformation of gastric epithelial cells in vitro. Oncol Res 21(1):33–41 Gao F-L, Li WS, Liu CL, Zhao GQ, 2013, Silencing Bmi-1 enhances the senescence and decreases the metastasis of human gastric cancer cells. World J Gastroenterol 19(46):8764–8769 Liu P, Zhang M, Niu Q, Zhang F, Yang Y, Jiang X, 2018, Knockdown of long non-coding RNA ANRIL inhibits tumorigenesis in human gastric cancer cells via microRNA-99a-mediated down-regulation of BMI1. Braz J Med Biol Res 51(10):e6839– e6839 Sahasrabuddhe AA, Dimri M, Bommi PV, Dimri GP, 2011, βTrCP regulates BMI1 protein turnover via ubiquitination and degradation. Cell cycle, Georgetown, Tex, 10(8):1322–1330 Rinn JL, Chang HY, 2012, Genome regulation by long noncoding RNAs. Annu Rev Biochem 81:145–166 PengWX, Koirala P,Mo YY, 2017, LncRNA-mediated regulation of cell signaling in cancer. Oncogene 36(41):5661–5667 Zhang E, He X, Zhang C, Su J, Lu X, Si X, Chen J, Yin D, Han L, de W, 2018, A novel long noncoding RNA HOXC-AS3 mediates tumorigenesis of gastric cancer by binding to YBX1. Genome Biol 19(1):154–154 Xue M, Chen LY,WangWJ, Su TT, Shi LH,Wang L, ZhangW, Si JM, Wang LJ, Chen SJ, 2018, HOTAIR induces the ubiquitination of Runx3 by interacting with Mex3b and enhances the invasion of gastric cancer cells. Gastric Cancer 21(5):756–764 Klingenberg M, Groß M, Goyal A, Polycarpou-Schwarz M, Miersch T, Ernst AS, Leupold J, Patil N, Warnken U, Allgayer H, Longerich T, Schirmacher P, Boutros M, Diederichs S, 2018, The long noncoding RNA Cancer susceptibility 9 and RNA binding protein heterogeneous nuclear ribonucleoprotein L form a complex and Coregulate genes linked to AKT signaling. Hepatology 68(5): 1817–1832 Jiang F, Qi W, Wang Y, Wang W, Fan L, 2019, lncRNA PEG10 promotes cell survival, invasion and migration by sponging miR- 134 in human bladder cancer. Biomed Pharmacother 114:108814 Zhai W et al, 2019, A positive feed-forward loop between LncRNA-URRCC and EGFL7/P-AKT/FOXO3 signaling promotes proliferation andmetastasis of clear cell renal cell carcinoma. Mol Cancer 18(1):81 Shao G et al, 2019, lncRNA CASC9 positively regulates CHK1 to promote breast cancer cell proliferation and survival through sponging the miR-195/497 cluster. Int J Oncol 54(5):1665–1675 Yang Y, Chen D, Liu H, Yang K, 2019, Increased expression of lncRNA CASC9 promotes tumor progression by suppressing autophagy-mediated cell apoptosis via the AKT/mTOR pathway in oral squamous cell carcinoma. Cell Death Dis 10(2):41–41 Li X, Chen B, Chi D, Zhang Y, Jiang W, 2019, lncRNA CASC9 regulates cell migration and invasion in hemangioma endothelial cells by targeting miR-125a-3p/Nrg1. OncoTargets Ther 12:423– 432 Wang KC, Chang HY, 2011, Molecular mechanisms of long noncoding RNAs. Mol Cell 43(6):904–914 Liu Z, Chen Z, Fan R, Jiang B, Chen X, Chen Q, Nie F, Lu K, Sun M, 2017, Over-expressed long noncoding RNA HOXA11-AS promotes cell cycle progression and metastasis in gastric cancer. Mol Cancer 16(1):82–82 LiW, Yu X, Xia Z, Yu X, Xie L, Ma X, Zhou H, Liu L,Wang J, Yang Y, Liu H, 2018, Repression of Noxa by Bmi1 contributes to deguelin-induced apoptosis in non-small cell lung cancer cells. J Cell Mol Med 22(12):6213–6227 Kim M, Lee S, ParkWH, Suh DH, Kim K, Kim YB, No JH, 2018, Silencing Bmi1 expression suppresses cancer stemness and enhances chemosensitivity in endometrial cancer cells. Biomed Pharmacother 108:584–589 Wang Y, Lv H, Xu Z, Sun J, Ni Y, Chen Z, Cheng X, 2019, Huaier n-butanol extract suppresses proliferation and metastasis of gastric cancer via c-Myc-Bmi1 axis. Sci Rep 9(1):447–447 Zhang X-W, Sheng YP, Li Q, Qin W, Lu YW, Cheng YF, Liu BY, Zhang FC, Li J, Dimri GP, Guo WJ, 2010, BMI1 and Mel-18 oppositely regulate carcinogenesis and progression of gastric cancer. Mol Cancer 9:40–40 Li W, Li Y, Tan Y, Ma K, Cui J, 2010, Bmi-1 is critical for the proliferation and invasiveness of gastric carcinoma cells. J Gastroenterol Hepatol 25(3):568–575 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 475 EP 482 DI 10.1007/s12253-019-00703-3 PG 8 ER PT J AU Aguin, N Rodriguez-Alonso, A Lopez-Trigo, N Castuera, P Luis, RJ Caeiro, B AF Aguin, Nerea Rodriguez-Alonso, Andres Lopez-Trigo, Nuria Castuera, I. Perez Luis, Rodriguez Javier Caeiro, Blazquez TI Association Between the Interleukin-17 Gene Polymorphism -197G>A and the Risk of Prostate Cancer in a Galician Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Interleukin-17A (IL-17A); SNP; Prostate cancer (PCa); Inflammation ID Interleukin-17A (IL-17A); SNP; Prostate cancer (PCa); Inflammation AB A case-control study was carried out in which the role of the Single Nucleotide Polymorphism rs2275913 in the pathogenesis of prostate cancer was analysed for the first time. This polymorphism is located in −197 position of IL-17A gene and implies a A>G change. The sample consists of 433 Galician men, 241 of whom are prostate cancer patients and 192 are healthy men with no tumours. Besides the influence of this marker, directly involved in the inflammatory process, other variables that were described as prostate cancer risk factors were also studied: age, smoking and Body Mass Index (BMI). By the analysis of Odds Ratio (OR) (CI 95%) a protective effect of heterozygous genotype AG was observed in comparison with homozygous genotypes AA and GG. As regards other risk factors, a significant increased risk was observed in smokers homozygous between 10 and 32 packyears (p = 0.032). Age and BMI show interesting patterns, but not significant ones. This study shows a possible link between the rs2275913 and the onset of PCa which could be influenced by age, BMI and above all, smoking. C1 [Aguin, Nerea] Universidade de Santiago de Compostela, Facultade de Bioloxia, Area de Antropoloxia Fisica, 15702 Santiago de Compostela, Galicia, Spain. [Rodriguez-Alonso, Andres] Hospital Arquitecto Marcide, Servicio de Uroloxia, 15405 Ferrol, Galicia, Spain. [Lopez-Trigo, Nuria] Universidade de Santiago de Compostela, Facultade de Bioloxia, Area de Antropoloxia Fisica, 15702 Santiago de Compostela, Galicia, Spain. [Castuera, I. Perez] Universidade de Santiago de Compostela, Facultade de Enfermeria, 15702 Santiago de Compostela, Galicia, Spain. [Luis, Rodriguez Javier] Universidade de Santiago de Compostela, Facultade de Bioloxia, Area de Antropoloxia Fisica, 15702 Santiago de Compostela, Galicia, Spain. [Caeiro, Blazquez] Universidade de Santiago de Compostela, Facultade de Bioloxia, Area de Antropoloxia Fisica, 15702 Santiago de Compostela, Galicia, Spain. RP Aguin, N (reprint author), Universidade de Santiago de Compostela, Facultade de Bioloxia, Area de Antropoloxia Fisica, 15702 Santiago de Compostela, Spain. EM Nerea.aguin@gmail.com CR de Oncologia Medica SE, SEOM,, 2016, Las cifras del cancer en Espana. Ed. Sociedad Espanola de Oncologia Medica, Madrid Asociacion Espanola Contra el Cancer, AECC, https://www.aecc.es. Accessed 26 March 2018 Books ES, 2015, Eurostat regional yearbook 2015. Publications Office of the European Union, Luxembourg Cayuela A, Rodriguez-Dominguez S, Martin VE, Candau BR, 2008, Recent changes in prostate cancer mortality in Spain. Trends analysis from 1991 to 2005. Actas Urol Esp 32:184–189 Cabanes A, Perez-Gomez B, Aragones N, PollanM, Lopez-Abente G, 2009, La situacion del cancer en Espana, 1975–2006. Instituto de Salud Carlos III, Madrid, pp 20–83 Sun J, Turner A, Xu J, Gronberg H, Isaacs W, 2007, Genetic variability in inflammation pathways and prostate cancer risk. Urol Oncol 25:250–259. , DOI 10.1016/j.urolonc.2006.10.001 Sfanos DF, De Marzo AM, 2012, Prostate cancer and inflammation: the evidence. Histopathology 60:199–215. , DOI 10. 1111/j.1365-2559.2011.04033.x Duan Y, Shi JN, Pan C, Chen HL, Zhang SZ, 2014, Association between the interleukin-17A -197G>A, rs2275913, polymorphism and risk of digestive cancer. Asian Pac J Cancer Prev 15:9295–9300 Howell WM, Rose-Zerilli MJ, 2006, Interleukin-10 polymorphisms, cancer susceptibility and prognosis. Familial Cancer 5: 143–149. , DOI 10.1007/s10689-005-0072-3 KawaguchiM, AdachiM, OdaN, Kokobu K, Huang SK, 2004, IL- 17 cytokine family. J Allergy Clin Immunol 116:1265–1273. , DOI 10.1016/j.jaci.2004.10.019 Murugaiyan G, Saha B, 2009, Protumor vs antitumor functions of IL-17. J Immunol 183:4169–4175. , DOI 10.4049/ jimmunol.0901017 Gaffen SL, 2011, Recent advances in the IL-17 cytokine family. Curr Opin Immunol 23:613–619. , DOI 10.1016/j.coi. 2011.07.006 Lv Q, Zhu D, Zhang J, Yi Y, Yang S, ZhangW, 2015, Association between six genetic variants of IL-17A and IL-17F and cervical cancer risk: a case-control study. Tumor Biol 36:3979–3984. , DOI 10.1007/s13277-015-3041-y Wang N, Yang J, Lu J, Qiao Q, Bao G, Wu T, Ho W, 2014, IL-17 gene polymorphism is associated with susceptibility to gastric cancer. Tumor Biol 35:10025–10030. , DOI 10.1007/s13277- 014-2255-8 Huppler AR, Bishu S, Gaffen SL, 2012, Mucocutaneous candidiasis: the IL-17 pathway and implications for targeted immunotherapy. Arthritis Res Ther 14:217. , DOI 10.1186/ar3893 Yang L-J, GaoW, Bai J-X, Zhang X-K, Han X, Sun Y-H, Zhang LL, Zhang M-M, 2016, Correlation between Interleukin-17 gene polymorphism and gastric cancer susceptibility in Han China population. Eur Rev Med Pharmacol Sci 20:1271–1282 Wang L, Jiang Y, Zhang Y, Wang Y, Huang S, Wang Z, Tian B, Yang Y, Jiang W, Pang D, 2012, Association analysis of IL-17A and IL-17F polymorphisms in Chinese Han women with breast cancer. PLoS One 7:e34400. , DOI 10.1371/journal.pone. 0034400 Zhou B, Zhang P,Wang Y, Shi S, ZhangK, Liao H, Zhang L, 2013, Interleukin-17 gene polymorphisms are associated with bladder cancer in a Chinese Han population. Mol Carcinog 53:871–878. , DOI 10.1002/mc.21928 Omrane I,Marrakchi R, Baroudi O, MezliniA,Ayari H, Medimegh I, Stambouli N, Kourda N, Bouzaienne H, Uhrhammer N, Bougatef K, Bignon Y-J, Benammar-Elgaaied A, 2014, Significant association between interleukin-17A polymorphism and colorectal cancer. Tumor Biol 35:6627–6632. , DOI 10.1007/s13277-014- 1890-4 Hou C, Yang F, 2015, Interleukin-17A gene polymorphism is associated with susceptibility to gastric cancer. Int J Clin Exp Pathol 8:7378–7384 PMC4525973 Li L, Tian YL, Lv XM, Yu HF, Xie YY, Wang JD, Shi W, 2015, Association analysis of IL-17A and IL-17F polymorphisms in Chinese women with cervical cancer. Genet Mol Res 14:12178– 12183. , DOI 10.4238/2015.October.9.6 Li Z, Liu D, Cao M, Jiang M, Luo F, 2015, IL-17A and IL-17F polymorphisms and gastric cancer risk: a meta-analysis. Genet Mol Res 14:7008–7017. , DOI 10.4238/2015.June.26.10 Dai ZM, Zhang TS, Lin S, Zhang WG, Liu J, Cao XM, Li HB, Wang M, Liu XH, Liu K, Li SL, Dai ZJ, 2016, Role of IL-17A rs2275913 and IL-17F rs763780 polymorphisms in risk of cancer development: an updated meta-analysis. Sci Rep 6:20439. https:// doi.org/10.1038/srep20439 Oeth P, Beaulieu M, Park C, Kosman D, del Mistro G, van den Boom D, Jurinke C, 2005, iPLEX™ assay: increased plexing efficiency and flexibility for MassARRAY® System through single base primer extension with mass-modified terminators. SEQUENOM Application Note 27 Sokal RR, Rohlf FJ, Lahoz Leon M, 1972, Biometria. Principios y metodos estadisticos en la investigacion biologica. Ed H Blume. Madrid Sole X, Guino E, Valls J, Iniesta R, Moreno V, 2006, SNPStats: a web tool for the analysis of association studies. Bioinformatics 22: 1928–1929. , DOI 10.1093/bioinformatics/btl268 Sherry ST,WordMH, KholodovM, Baker J, Phan L, Smigielski EM et al, 2001, dbNSP: the NCBI database of genetic variation. Nucleic Acids Res 29(1):308–311. http://www.ncbi.nlm.nih.gov/snp. Accessed 26 March 2018 Finkel T, Serrano M, Blasco MA, 2007, The common biology of cancer and ageing. Nature 448:767–774. , DOI 10.1038/ nature05985 Rook GAW, Dalgleish A, 2011, Infection, immunoregulation, and cancer. Immunol Rev 240:141–159. , DOI 10.1111/j.1600- 065X.2010.00987.x Lavie CJ,Milani RV, Ventura HO, 2009, Obesity and cardiovascular disease. Risk factor, paradox, and impact of weight loss. J Am Coll Cardiol 53:1925–1932. , DOI 10.1016/j.jacc.2008.12.068 Zeyda M, Stulnig TM(2007, Adipose tissue macrophages. Immunol Lett 112:61–67. , DOI 10.1016/j.imlet.2007.07.003 Shibata T, Tahara T, Hirata I, Arisawa T Genetic polymorphism of interleukin-17A and -17F genes in gastric carcinogenesis. Hum Immunol 70:547–551. , DOI 10.1016/j.humimm.2009.04.030 WuX, Zeng Z, ChenB,Yu J,XueL,HaoY, ChenM, Sung JJY, Hu P, 2010, Association between polymorphisms in interleukin-17A and interleukin-17F genes and risks of gastric cancer. Int J Cancer 127:86–92. , DOI 10.1002/ijc.25027 Arisawa T, Tahara T, Shiroeda H, Matsue Y, Minato T, Nomura T, Yamada H, Hayashi R, Saito T, Matsunaga K, Fukuyama T, Hayashi N, Otsuka T, Fukumura A, Nakamura M, Shibata T, 2012, Genetic polymorphisms of IL17A and pri-microRNA-938, targeting IL17A 3’-UTR, influence susceptibility to gastric cancer. Hum Immunol 73:747–752. , DOI 10.1016/j.humimm. 2012.04.011 Quan Y, Zhou B,Wang Y, Duan R,Wang K, Gao Q, Shi S, Yaping S, Zhang L, Xi M, 2012, Association between IL17 polymorphisms and risk of cervical cancer in Chinese women. Clin Dev Immunol 258293:1–6. , DOI 10.1155/2012/258293 Kutikhin AG, Yuzhalin AE, Volkov AN, Zhivotovskiy AS, Brusina EB, 2014, Correlation between genetic polymorphisms within IL- 1B and TLR4 genes and cancer risk in a Russian population: a casecontrol study. Tumor Biol 35:4821–4830. , DOI 10.1007/ s13277-014-1633-6 Qinghai Z, Yanying W, Yunfang C, Xuui Z, Xiaoqiao Z, 2014, Effect of interleukin-17A and interleukin-17F gene polymorphisms on the risk of gastric cancer in a Chinese population. Gene 537: 328–332. , DOI 10.1016/j.gene.2013.11.007 Zhang X, Zheng L, Sun Y, Zhang X, 2014, Analysis of the association of interleukin-17 gene polymorphisms with gastric cancer risk and interaction with Helicobacter pylori infection in a Chinese population. Tumor Biol 35:1575–1580. , DOI 10.1007/ s13277-013-1217-x Cong J, Liu R,Wang X, Sheng L, Jiang H,WangW, ZhangY, Yang S, Li C, 2015, Association between interleukin-17 gene polymorphisms and the risk of cervical cancer in a Chinese population. Int J Clin Exp Pathol 8:9567–9573 Gao Y-W, Xu M, Xu Y, Li D, Zhou S, 2015, Effect of three common IL-17 single nucleotide polymorphisms on the risk of developing gastric cancer. Oncol Lett 9:1398–1402. , DOI 10. 3892/ol.2014.2827 Lee YC, Chung JH, Kim SK, Rhee SY, Chon S, Oh SJ, Hong IK, Eun YG, 2015, Association between interleukin 17/interleukin 17 receptor gene polymorphisms and papillary thyroid cancer in Korean population. Cytokine 71:283–288. , DOI 10.1016/ j.cyto.2014.11.011 Sun LX, Wang XB, Huang XJ, 2015, Association analysis of rs2275913G>A and rs763780T>C interleukin 17 polymorphisms in Chinese women with cervical cancer. GenetMol Res 14:13612– 13617. , DOI 10.4238/2015.October.28.22 Xi XE, Liu Y, Lu Y, Huang L, Qin X, Li S, 2015, Interleukin-17A and interleukin-17F gene polymorphisms and hepatitis B virusrelated hepatocellular carcinoma risk in a Chinese population. Med Oncol 32:355. , DOI 10.1007/s12032-014-0355-3 Eiro N, Vizoso FJ, 2912, Inflammation and cancer. World J Gastrointestinal Surg 4:62–72. , DOI 10.4240/wjgs.v4.i3.62 Eder T, Mayer R, Langsenlehner U, Renner W, Krippl P, Wascher TC, Pummer K, Kapp KS, 2009, Interleukin-10 [ATA] promoter haplotype and prostate cancer risk: a population-based study. Eur J Cancer 43:472–475. , DOI 10.1016/j.ejca.2006.11.003 Zabaleta J, Lin H-Y, Sierra RA, HallMC, Clark PE, Sartor OA, Hu JJ, Ochoa AC, 2008, Interactions of cytokine gene polymorphisms in prostate cancer risk. Carcinogenesis 29:573–578. , DOI 10.1093/carcin/bgm277 Michaud DS, Daugherty SE, Berndt S, Platz EA, Yeager M, Crawford ED, Hsing A, Huang W-Y, Hayes RB, 2006, Genetic polymorphisms of interleukin-1B, IL-1B), IL-6, IL-8, and IL-10, and risk of prostate cancer. Cancer Res 66:4525–4530. https://doi. org/10.1158/0008-5472.CAN-05-3987 Iwakura Y, Ishigame H, Saijo S, Nakae S, 2011, Functional specialization of interleukin-17 family members. Immunity 34:149– 162. , DOI 10.1016/j.immuni.2011.02.012 Chen CH, Gong M,Yi QT,Guo JH, 2015, Role of interleukin-6 gene polymorphisms in the development of prostate cancer. Genet Mol Res 14:13370–13374. , DOI 10.4238/2015.October.26.34 Faupel-Bagder JM, Kidd LCR, Albanes D, Virtamo J,Woodson K, Tangrea JA, 2008, Association of IL-10 polymorphisms with prostate cancer risk and grade of disease. Cancer Causes Control 19: 119–124. , DOI 10.1007/s10552-007-9077-6 Mandal S, Abebe F, Chaudhary J, 2014, -174G/C polymorphism in the interleukin-6 promoter is differently associated with prostate cancer incidence depending on race. Genet Mol Res 13:139–151. , DOI 10.4238/2014.January.10.5 Espinoza JL, Takami A, Nakata K, Onizuka M, Kawase T, Akiyama H, Miyamura K, Morishima Y, Fukuda T, Kodera Y, Nakao S, 2011, A genetic variant in the IL-17 promoter is functionally associated with acute graft-versus-host disease after unrelated bone marrow transplantation. PLoS ONE 6:e26229. https:// doi.org/10.1371/journal.pone.0026229 Wrobel T, Gḙbura K, Wysoczariska B, Jaẑwiec V, Dobrzyriska O, Mazur G, Kuliczkowski K, Bogunia-Kubik K, 2014, IL-17F gene polymorphism is associated with susceptibility to acute myeloid leukaemia. J Cancer Res Clin Oncol 140:1551–1555. https://doi. org/10.1007/s00432-014-1674-7 Nemati K, Golmoghaddam H, Hosseini SV, Ghaderi A, Doroudchi M, 2015, Interleukin-17FT7488 allele is associated with a decreased risk of colorectal cancer and tumour progression. Gene 561:88–94. , DOI 10.1016/j.gene.2015.02.014 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 483 EP 489 DI 10.1007/s12253-018-0537-y PG 7 ER PT J AU Liu, Y Yang, B Zhang, X Huang, Q Liu, H AF Liu, Yanhui Yang, Bo Zhang, Xiaoyan Huang, Quanfei Liu, Hailiang TI The Gene Mutation Spectrum of Breast Cancer Analyzed by Semiconductor Sequencing Platform SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Semiconductor sequencing platform (SSP); Breast cancer; Mutate spectrum; Genetic screening ID Semiconductor sequencing platform (SSP); Breast cancer; Mutate spectrum; Genetic screening AB To use the semiconductor sequencing platform (SSP) to analyze the gene mutate spectrum of breast cancer patients. We recruited 46 breast cancer patients, and detected the ER/PR/HER2 expression level of the tumor tissue by immunohistochemistry. In addition, combined with SSP technology, we detected 207 hot mutation regions of 50 breast cancer related genes with multiple polymerase chain reaction (PCR) technology. There were 8 cases of grade I, 18 cases of grade II, 20 cases of grade III in 46 breast cancer patients according to histological grade and 12 cases of ER/PR + HER2 +, 18 cases of ER/PR + HER2−, 13 cases of ER/PR - HER2 +, 20 cases of ER/PR − HER2− according to ER/PR/HER2 status classification. Moreover, we found that there were 33 gene locus mutations of 8 genes including AKT1, APC, BRAF, CDKN2A, KRAS, PTEN, PIK3CA and TP53, but difference was not statistically significant (P > 0.05) when compared these gene mutations (except for PIK3CA) in each groups according to the histological classification of breast cancer and the ER/PR/HER2 classification. PIK3CA mutation rate of grade I was obviously higher than that of grade II ~ III histological grading in breast cancer patients (P < 0.05). Based on our results, we drew a conclusion that the occurrence and development of breast cancer was a process involved multiple genes. Here, we found that PIK3CA played a role in the development of the early stage of breast cancer, which could provide clinical basis for treatment of breast cancer. Moreover, SSP technology could be an effective and sensitive method for detection of gene mutation spectrum in breast cancer. C1 [Liu, Yanhui] Dongguan Maternal and Child Health Hospital, Dongguan Institute of Reproductive and Genetic Research, 523000 Dongguan, Guangdong, China. [Yang, Bo] Dongguan Maternal and Child Health Hospital, Dongguan Institute of Reproductive and Genetic Research, 523000 Dongguan, Guangdong, China. [Zhang, Xiaoyan] Dongguan Maternal and Child Health Hospital, Dongguan Institute of Reproductive and Genetic Research, 523000 Dongguan, Guangdong, China. [Huang, Quanfei] CapitalBio Technology Inc., 101111 Beijing, China. [Liu, Hailiang] CapitalBio Technology Inc., 101111 Beijing, China. RP Liu, Y (reprint author), Dongguan Maternal and Child Health Hospital, Dongguan Institute of Reproductive and Genetic Research, 523000 Dongguan, China. EM liuliang71215@163.com CR Cancer Genome Atlas Network, 2012, Comprehensive molecular portraits of human breast tumours. Nature 490:61–70 Fan L, Strasser-Weippl K, Li JJ, St Louis J, Finkelstein DM, Yu KD, Chen WQ, Shao ZM, Goss PE, 2014, Breast cancer in China. Lancet Oncol 15: e279–289 Banin Hirata BK, Oda JM, Losi Guembarovski R, Ariza CB, de Oliveira CE, Watanabe MA, 2014, Molecular markers for breast cancer: prediction on tumor behavior. Dis Markers 2014:513158 van de Vijver MJ, He YD, van't Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, Parrish M, Atsma D, Witteveen A, Glas A, Delahaye L, van der Velde T, Bartelink H, Rodenhuis S, Rutgers ET, Friend SH, Bernards R, 2002, A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 347:1999–2009 Kittaneh M, Montero AJ, Gluck S, 2013, Molecular profiling for breast cancer: a comprehensive review. Biomark Cancer 5:61–70 Roy-Chowdhuri S, de Melo Gagliato D, Routbort MJ, Patel KP, Singh RR, Broaddus R, Lazar AJ, Sahin A, Alvarez RH, Moulder S, Wheler JJ, Janku F, Gonzalez-Angulo AM, Chavez-MacGregor M, Valero V, Ueno NT, Mills G, Mendelsohn J, Yao H, Aldape K, Luthra R, Meric-Bernstam F, 2015, Multigene clinical mutational profiling of breast carcinoma using next-generation sequencing. Am J Clin Pathol 144:713–721 Lundin J, Lundin M, Holli K, Kataja V, Elomaa L, Pylkkanen L, Turpeenniemi-Hujanen T, JoensuuH, 2001, Omission of histologic grading from clinical decision making may result in overuse of adjuvant therapies in breast cancer: results from a nationwide study. J Clin Oncol 19:28–36 van 't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA,Mao M, Peterse HL, van der Kooy K, Marton MJ,Witteveen AT, Schreiber GJ, Kerkhoven RM, Roberts C, Linsley PS, Bernards R, Friend SH, 2002, Gene expression profiling predicts clinical outcome of breast cancer. Nature 415:530–536 IvshinaAV, George J, Senko O,MowB, Putti TC, Smeds J, Lindahl T, Pawitan Y, Hall P, Nordgren H, Wong JE, Liu ET, Bergh J, Kuznetsov VA, Miller LD, 2006, Genetic reclassification of histologic grade delineates new clinical subtypes of breast cancer. Cancer Res 66:10292–10301 Lips EH, Michaut M, Hoogstraat M, Mulder L, Besselink NJ, Koudijs MJ, Cuppen E, Voest EE, Bernards R, Nederlof PM, Wesseling J, Rodenhuis S, Wessels LF, 2015, Next generation sequencing of triple negative breast cancer to find predictors for chemotherapy response. Breast Cancer Res 17:134 Almendro V, Fuster G, 2011, Heterogeneity of breast cancer: etiology and clinical relevance. Clin Transl Oncol 13:767–773 Ariga R, Zarif A, Korasick J, Reddy V, Siziopikou K, Gattuso P, 2005, Correlation of her-2/neu gene amplification with other prognostic and predictive factors in female breast carcinoma. Breast J 11:278–280 Sneige N, Hess KR, Multani AS, Gong Y, Ibrahim NK, 2017, Prognostic significance of equivocal human epidermal growth factor receptor 2 results and clinical utility of alternative chromosome 17 genes in patients with invasive breast cancer: a cohort study. Cancer 123:1115–1123 Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, Baehner FL, Walker MG, Watson D, Park T, Hiller W, Fisher ER, Wickerham DL, Bryant J, Wolmark N, 2004, A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 351:2817–2826 Cantley LC, 2002, The phosphoinositide 3-kinase pathway. Science 296:1655–1657 Berns K, Horlings HM, Hennessy BT, Madiredjo M, Hijmans EM, Beelen K, Linn SC, Gonzalez-Angulo AM, Stemke-Hale K, Hauptmann M, Beijersbergen RL, Mills GB, van de Vijver MJ, Bernards R, 2007, A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell 12:395–402 Bhat-Nakshatri P, Goswami CP, Badve S, Magnani L, Lupien M, Nakshatri H, 2016, Molecular insights of pathways resulting from two common PIK3CA mutations in breast Cancer. Cancer Res 76: 3989–4001 Mayer IA, Arteaga CL, 2014, PIK3CA activating mutations: a discordant role in early versus advanced hormone-dependent estrogen receptor-positive breast cancer? J Clin Oncol 32:2932–2934 von Minckwitz G, Schneeweiss A, Loibl S, Salat C, Denkert C, Rezai M, Blohmer JU, Jackisch C, Paepke S, Gerber B, Zahm DM, Kummel S, Eidtmann H, Klare P, Huober J, Costa S, Tesch H, Hanusch C, Hilfrich J, Khandan F, Fasching PA, Sinn BV, Engels K, Mehta K, Nekljudova V, Untch M, 2014, Neoadjuvant carboplatin in patients with triple-negative and HER2-positive early breast cancer, GeparSixto; GBG 66): a randomised phase 2 trial. Lancet Oncol 15:747–756 Payne SN, Maher ME, Tran NH, Van De Hey DR, Foley TM, Yueh AE, Leystra AA, Pasch CA, Jeffrey JJ, Clipson L, MatkowskyjKA, Deming DA, 2015, PIK3CA mutations can initiate pancreatic tumorigenesis and are targetable with PI3K inhibitors. Oncogenesis 4:e169 Liang X, Lau QC, Salto-Tellez M, Putti TC, Loh M, Sukumar S, 2006, Mutational hotspot in exon 20 of PIK3CA in breast cancer among Singapore Chinese. Cancer Biol Ther 5:544–548 Liedtke C, Cardone L, Tordai A, Yan K, Gomez HL, Figureoa LJ, Hubbard RE, Valero V, Souchon EA, Symmans WF, Hortobagyi GN, Bardelli A, Pusztai L, 2008, PIK3CA-activating mutations and chemotherapy sensitivity in stage II-III breast cancer. Breast Cancer Res 10:R27 Huszno J, Grzybowska E, 2018, TP53 mutations and SNPs as prognostic and predictive factors in patients with breast cancer. Oncol Lett 16:34–40 Schayek H, De Marco L, Starinsky-Elbaz S, Rossette M, Laitman Y, Bastos-Rodrigues L, da Silva Filho AL, Friedman E, 2016, The rate of recurrent BRCA1, BRCA2, and TP53 mutations in the general population, and unselected ovarian cancer cases. In: Belo Horizonte, Brazil, Cancer Genet, 209, pp 50–52 Lee DS, Yoon SY, Looi LM, Kang P, Kang IN, Sivanandan K, Ariffin H, Thong MK, Chin KF, Mohd Taib NA, Yip CH, Teo SH, 2012, Comparable frequency of BRCA1, BRCA2 and TP53 germline mutations in a multi-ethnic Asian cohort suggests TP53 screening should be offered together with BRCA1/2 screening to early-onset breast cancer patients. Breast Cancer Res 14:R66 Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB, 2005, Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov 4:988–1004 Samuels Y, Velculescu VE, 2004, Oncogenic mutations of PIK3CA in human cancers. Cell Cycle 3:1221–1224 M. Lapierre, S. Bonnet, C. Bascoul-Mollevi, I. Ait-Arsa, S. Jalaguier, M. Del Rio, M. Plateroti, P. Roepman, M. Ychou, J. Pannequin, F. Hollande, M. Parker, V. Cavailles, RIP140 increases APC expression and controls intestinal homeostasis and tumorigenesis, J Clin Invest, 124, 2014, 1899–1913 Clevers H, Nusse R, 2012)Wnt/beta-catenin signaling and disease. Cell 149:1192–1205 Sanchez-Munoz A, Gallego E, de Luque V, Perez-Rivas LG, Vicioso L, Ribelles N, Lozano J, Alba E, 2010, Lack of evidence for KRAS oncogenic mutations in triple-negative breast cancer. BMC Cancer 10:136 Premalatha BR, Patil S, Rao RS, Reddy NP, Indu M, 2013, Odontogenic tumor markers - an overview. J Int Oral Health 5: 59–69 Nagore E,Montoro A, Garcia-Casado Z, Botella-Estrada R, Insa A, Lluch A, Lopez-Guerrero JA, Guillen C, 2009, Germline mutations in CDKN2A are infrequent in female patients with melanoma and breast cancer. Melanoma Res 19:211–214 Debniak T, Cybulski C, Gorski B, Huzarski T, Byrski T, Gronwald J, Jakubowska A, Kowalska E, Oszurek O, Narod SA, Lubinski J, 2007, CDKN2A-positive breast cancers in young women from Poland. Breast Cancer Res Treat 103:355–359 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 491 EP 497 DI 10.1007/s12253-018-0522-5 PG 7 ER PT J AU Wang, Xe Wang, Yh Zhou, Q Peng, M Zhang, J Chen, M Ma, Lj Xie, GM AF Wang, Xi-en Wang, You-hui Zhou, Qiang Peng, Min Zhang, Jing Chen, Mi Ma, Li-juan Xie, Guo-Ming TI Immunomodulatory Effect of Lentinan on Aberrant T Subsets and Cytokines Profile in Non-small Cell Lung Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; Chemo-immunotherapy; Lentinan; T cell subsets ID Non-small cell lung cancer; Chemo-immunotherapy; Lentinan; T cell subsets AB As a purified active component from traditional Chinese medicine, lentinan administration can be applied as beneficial chemoimmunotherapy for anti-tumor. In this study, the immunomodulatory effects of lentinan on aberrant T subsets and cytokines profile were evaluated for non-small cell lung cancer (NSCLC). Of all NSCLC patients treated with NP chemotherapeutic protocol (combination of vinorelbin and cisplatin), 73 cases were recruited in this retrospective cohort trial study, of which 38 cases received additional lentinan. The changes of aberrant T subsets and cytokines profile were compared between two groups (chemotherapy in combination with lentinan vs. conserved single chemotherapy) by flow cytometry and molecular biology. Higher subset ratio of CD3+CD8+ cytotoxic T cells was confirmed in the peripheral blood of NSCLC patients. Chemoimmunotherapy of lentinan resulted in a significant increase of CD3 + CD56+ NKT cells (15.7 ± 3.1%), compared with 8.6 ± 1.4% of NKT cells in single chemotherapy group, and up-regulated CD3+CD8+ and CD3+CD4+ subsets as well, but caused the decrease of CD4+CD25+ Tregs induction, accompanied by significant alleviation of IL-10 and TGF-β1, and elevation of IFN-γ, TNF-α, and IL-12 (P < 0.05). It could be confirmed that lentinan could not only enhance the cellular immunity and promote the beneficial of anti-tumor by associated immunotherapy, but also had the ability to inhibit the expansion of immune suppressive Tregs in the NSCLC patients, in whom there was a raised Tregs induction compared to health control. Lentinan-based chemoimmunotherapy is a promising strategy for anti-tumor via enhancing the proliferation of cytotoxic T cells, followed by the elevation of inflammatory chemokines/cytokines. Meanwhile, the percentage of CD4+ CD25+ Tregs is down-regulated, leading to a shift in the inflammatory status from Th2 to Th1 in NSCLC patients treated with lentinan. C1 [Wang, Xi-en] Ningbo Yinzhou People Hospital, Department of Integrated Traditional Chinese and Western Medicine on Oncology, 315040 Ningbo, Zhejiang, China. [Wang, You-hui] Ningbo Yinzhou People Hospital, Department of Integrated Traditional Chinese and Western Medicine on Oncology, 315040 Ningbo, Zhejiang, China. [Zhou, Qiang] Ningbo Yinzhou People Hospital, Department of Integrated Traditional Chinese and Western Medicine on Oncology, 315040 Ningbo, Zhejiang, China. [Peng, Min] Ningbo Yinzhou People Hospital, Department of Integrated Traditional Chinese and Western Medicine on Oncology, 315040 Ningbo, Zhejiang, China. [Zhang, Jing] Ningbo Yinzhou People Hospital, Department of Integrated Traditional Chinese and Western Medicine on Oncology, 315040 Ningbo, Zhejiang, China. [Chen, Mi] Ningbo Yinzhou People Hospital, Department of Integrated Traditional Chinese and Western Medicine on Oncology, 315040 Ningbo, Zhejiang, China. [Ma, Li-juan] Ningbo Yinzhou People Hospital, Department of Integrated Traditional Chinese and Western Medicine on Oncology, 315040 Ningbo, Zhejiang, China. [Xie, Guo-Ming] Ningbo Yinzhou People Hospital, Department of Integrated Traditional Chinese and Western Medicine on Oncology, 315040 Ningbo, Zhejiang, China. RP Wang, Xe (reprint author), Ningbo Yinzhou People Hospital, Department of Integrated Traditional Chinese and Western Medicine on Oncology, 315040 Ningbo, China. EM XEnWang@tom.com CR Tolcher AW, Patnaik A, Papadopoulos KP, RascoDW, Becerra CR, Allred AJ, Orford K, Aktan G, Ferron-Brady G, Ibrahim N, Gauvin J, Motwani M, Cornfeld M, 2015, Phase I study of the MEK inhibitor trametinib in combination with the AKTinhibitor afuresertib in patients with solid tumors and multiple myeloma. Cancer Chemother Pharmacol 75:183–189 Guo Q, Li J, Lin H, 2015, Effect and molecular mechanisms of traditional Chinese medicine on regulating tumor immunosuppressive microenvironment. Biomed Res Int 2015:261620 Tanaka K, Ishikawa S, Matsui Y, Tamesada M, Harashima N, Harada M, 2011, Oral ingestion of Lentinula edodes mycelia extract inhibits B16 melanoma growth via mitigation of regulatory T cell-mediated immunosuppression. Cancer Sci 102:516–521 Nagashima Y, Sanpei N, Yamamoto S, Yoshino S, Tangoku A, Oka M, 2005, Evaluation of host immunity and side effects in breast cancer patients treated with adjuvant chemotherapy, FEC therapy). Gan To Kagaku Ryoho 32:1550–1552 Okuno K, Uno K, 2011, Efficacy of orally administered Lentinula edodesmycelia extract for advanced gastrointestinal cancer patients undergoing cancer chemotherapy: a pilot study. Asian Pac J Cancer Prev 12:1671–1674 Nakano H, Namatame K, Nemoto H, Motohashi H, Nishiyama K, Kumada K, 1999, A multi-institutional prospective study of lentinan in advanced gastric cancer patients with unresectable and recurrent diseases: effect on prolongation of survival and improvement of quality of life. Kanagawa Lentinan Research Group. Hepato-Gastroenterology 46:2662–2668 Wang W, Dai X, Ouyang X, 2006, Efficacy of lentinan combined with chemotherapy in advanced non-small cell lung cancer. Zhongguo Fei Ai Za Zhi 9:78–81 Piao BK, Wang YX, Xie GR, Mansmann U, Matthes H, Beuth J, Lin HS, 2004, Impact of complementary mistletoe extract treatment on quality of life in breast, ovarian and non-small cell lung cancer patients. A prospective randomized controlled clinical trial. Anticancer Res 24:303–309 Kidd PM, 2000, The use of mushroom glucans and proteoglycans in cancer treatment. Altern Med Rev 5:4–27 Yamaguchi Y, Miyahara E, Hihara J, 2011, Efficacy and safety of orally administered Lentinula edodes mycelia extract for patients undergoing cancer chemotherapy: a pilot study.AmJ ChinMed 39: 451–459 Murata Y, Shimamura T, Tagami T, Takatsuki F, Hamuro J, 2002, The skewing to Th1 induced by lentinan is directed through the distinctive cytokine production by macrophages with elevated intracellular glutathione content. Int Immunopharmacol 2:673–689 Chehimi J, Trinchieri G, 1994, Interleukin-12: a bridge between innate resistance and adaptive immunity with a role in infection and acquired immunodeficiency. J Clin Immunol 14: 149–161 Hsieh CS, Heimberger AB, Gold JS, O'Garra A, Murphy KM, 1992, Differential regulation of T helper phenotype development by interleukins 4 and 10 in an alpha beta T-cell-receptor transgenic system. Proc Natl Acad Sci U S A 89:6065–6069 Murata Y, Shimamura T, Hamuro J, 2002, The polarization of T(h)1/T(h)2 balance is dependent on the intracellular thiol redox status of macrophages due to the distinctive cytokine production. Int Immunol 14:201–212 Karpus WJ, Swanborg RH, 1989, CD4+ suppressor cells differentially affect the production of IFN-gamma by effector cells of experimental autoimmune encephalomyelitis. J Immunol 143:3492–3497 D'Andrea A, Aste-Amezaga M, Valiante NM, Ma X, Kubin M, Trinchieri G, 1993, Interleukin 10, IL-10, inhibits human lymphocyte interferon gamma-production by suppressing natural killer cell stimulatory factor/IL-12 synthesis in accessory cells. J Exp Med 178:1041–1048 Suzuki M, Kikuchi T, Takatsuki F, Hamuro J, 1994, Curative effects of combination therapy with lentinan and interleukin-2 against established murine tumors, and the role of CD8-positive T cells. Cancer Immunol Immunother 38:1–8 Chen C, Chen D, ZhangY, Chen Z, ZhuW, Zhang B,Wang Z, Le H, 2014, Changes of CD4+CD25+FOXP3+ and CD8+CD28- regulatory T cells in non-small cell lung cancer patients undergoing surgery. Int Immunopharmacol 18:255–261 Filaci G, Fenoglio D, Fravega M, Ansaldo G, Borgonovo G, Traverso P, 2007, CD8+ CD28- T regulatory lymphocytes inhibiting T cell proliferative and cytotoxic functions infiltrate human cancers. J Immunol 179:4323–4334 Tsukishiro T, Donnenberg AD, Whiteside TL, 2003, Rapid turnover of the CD8(+)CD28(−, T-cell subset of effector cells in the circulation of patients with head and neck cancer. Cancer Immunol Immunother 52:599–607 Karagoz B, Bilgi O, Gumus M, Erikci AA, Sayan O, Turken O, Kandemir EG, Ozturk A, Yaylaci M, 2010, CD8+CD28- cells and CD4+CD25+ regulatory Tcells in the peripheral blood of advanced stage lung cancer patients. Med Oncol 27:29–33 Meloni F, MorosiniM, Solari N, Passadore I, Nascimbene C, Novo M, 2006, Foxp3 expressing CD4+CD25+ and CD8+CD28- T regulatory cells in the peripheral blood of patients with lung cancer and pleural mesothelioma. Hum Immunol 67:1–12 Giroux M, Denis F, 2005, CD1d-unrestricted human NKT cells release chemokines upon Fas engagement. Blood 105:703–710 Sathyanarayanan V, Neelapu SS, 2015, Cancer immunotherapy: strategies for personalization and combinatorial approaches. Mol Oncol 9:2043–2053 Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DA, Wherry EJ, 2009, Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol 10:29–37 Wherry EJ, 2011, T cell exhaustion. Nat Immunol 12:492–499 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 499 EP 505 DI 10.1007/s12253-018-0545-y PG 7 ER PT J AU Khan, SM Qadri, Q Makhdoomi, JM Wani, AM Malik, AA Niyaz, M Masoodi, RSh Andrabi, IK Ahmad, R Mudassar, S AF Khan, S Mosin Qadri, Qurteeba Makhdoomi, J Mudasir Wani, A Muneer Malik, A Aejaz Niyaz, Madiha Masoodi, R Shariq Andrabi, I Khurshid Ahmad, Rauf Mudassar, Syed TI RET/PTC Gene Rearrangements in Thyroid Carcinogenesis: Assessment and Clinico-Pathological Correlations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Thyroid Cancer; TSH; Papillary thyroid cancer; Kashmir Valley; RET/PTC rearrangement ID Thyroid Cancer; TSH; Papillary thyroid cancer; Kashmir Valley; RET/PTC rearrangement AB Rearranged during transfection (RET) is a proto oncogene implicated in thyroid carcinogenesis of papillary type (PTC). The RET proto-oncogene in PTC is constitutively activated by fusion of its tyrosine kinase domain with the 5 ´region of another gene thereby generating chimeric products collectively named RET/PTCs. RET/PTC1 and RET/PTC3 are best characterized among all RET/PTC rearrangements. Kashmir valley has witnessed an alarming increase in thyroid cancer incidence in young women. Therefore, we investigated the occurrence of RET/PTC 1 & 3 rearrangements by semi quantitative and qPCR in thyroid cancer patients (n = 48) of Kashmiri population and interrelated results with various clinicopathological characteristics. We observed that all the RET/PTC rearrangements were confined to PTC cases (10/40). Presence of RET/PTC rearrangement significantly correlated with gender, elevated TSH levels and lymph node metastasis. Overall, our study advocates that RET/PTC3 rearrangement is a frequent event in the carcinogenesis of thyroid gland in Kashmiri population although a study with a larger sample size is needed to get a clear scenario. C1 [Khan, S Mosin] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Soura, Srinagar, India. [Qadri, Qurteeba] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Soura, Srinagar, India. [Makhdoomi, J Mudasir] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Soura, Srinagar, India. [Wani, A Muneer] Sher-I-Kashmir Institute of Medical Sciences, Department of General and Minimal Invasive Surgery, 190011 Soura, Srinagar, India. [Malik, A Aejaz] Sher-I-Kashmir Institute of Medical Sciences, Department of General and Minimal Invasive Surgery, 190011 Soura, Srinagar, India. [Niyaz, Madiha] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Soura, Srinagar, India. [Masoodi, R Shariq] Sher-I-Kashmir Institute of Medical Sciences, Department of Endocrinology, 190011 Soura, Srinagar, India. [Andrabi, I Khurshid] University of Kashmir, Department of Biotechnology, 190006 Hazratbal, Srinagar, India. [Ahmad, Rauf] SMHS Govt. Superspeciality Hospital, Department of ENT, 190010 Karan Nagar, Srinagar, India. [Mudassar, Syed] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Soura, Srinagar, India. RP Mudassar, S (reprint author), Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Soura, India. EM syed.mudassar@skims.ac.in CR Pellegriti G, Frasca F, Regalbuto C, Squatrito S, Vigneri R, 2013, Worldwide increasing incidence of thyroid cancer: update on epidemiology and risk factors. J Cancer Epidemiol 2013:1–10. https:// doi.org/10.1155/2013/965212 La Vecchia C, Malvezzi M, Bosetti C et al, 2015, Thyroid cancer mortality and incidence: a global overview. Int J Cancer 136:2187– 2195. , DOI 10.1002/ijc.29251 Liu Y, Su L, Xiao H, 2017, Review of factors related to the thyroid Cancer epidemic. Int J Endocrinol 2017:1–9. , DOI 10. 1155/2017/5308635 XingM(2013)Molecular pathogenesis and mechanisms of thyroid cancer. Nat Rev Cancer 13:184–199. , DOI 10.1038/ nrc3431 Marotta V, Guerra A, Sapio MR, Vitale M, 2011, RET/PTC rearrangement in benign and malignant thyroid diseases: a clinical standpoint. Eur J Endocrinol 165:499–507 , DOI 10. 1530/EJE-11-0499 Santoro M, Carlomagno F, 2013, Central role of RET in thyroid cancer. Cold Spring Harb Perspect Biol 5:1–17. , DOI 10. 1101/cshperspect.a009233 Kurokawa K, Kawai K, Hashimoto M, Ito Y, Takahashi M, 2003, Cell signalling and gene expression mediated by RET tyrosine kinase. J Intern Med 253:627–633 Nikiforov YE, 2002, RET/PTC rearrangement in thyroid tumors. Endocr Pathol 13:3–16. , DOI 10.1385/EP:13:1:03 Tong G-X, Mody K, Wang Z, Hamele-Bena D, Nikiforova MN, Nikiforov YE, 2015, Mutations of TSHR and TP53 genes in an aggressive clear cell follicular carcinoma of the thyroid. Endocr Pathol 26:315–319 , DOI 10.1007/s12022-015-9388-1 Khan MS, Pandith AA, Azad N, Hussain MU, Masoodi SR, Wani KA, Andrabi KI, Mudassar S, 2014, Impact of molecular alterations of BRAF in the pathogenesis of thyroid cancer. Mutagenesis 29:131–137 Khan MS, Pandith AA, HussainMU, Iqbal M, Khan NP,Wani KA, Masoodi SR,Mudassar S, 2013, Lack of mutational events of RAS genes in sporadic thyroid cancer but high risk associated with HRAS T81C singlenucleotide polymorphism, case–control study). Tumor Biol 34:521–529 Jhiang SM, 2000, The RET proto-oncogene in human cancers. Oncogene 19:5590–5597. , DOI 10.1038/sj.onc.1203857 Romei C, Ciampi R, Elisei R, 2016, A comprehensive overview of the role of the RET proto-oncogene in thyroid carcinoma. Nat Rev Endocrinol 12:192–202 , DOI 10.1038/nrendo.2016.11 Thomas GA, Bunnell H, Cook HA et al, 1999, High prevalence of RET/PTC rearrangements in Ukrainian and Belarussian post- Chernobyl thyroid papillary carcinomas: a strong correlation between RET/PTC3 and the solid-follicular variant. J Clin Endocrinol Metab 84:4232–4238. , DOI 10.1210/jcem.84. 11.6129 HamataniK, EguchiH, Ito R,MukaiM, Takahashi K, TagaM, Imai K, Cologne J, Soda M, Arihiro K, Fujihara M, Abe K, Hayashi T, Nakashima M, Sekine I, Yasui W, Hayashi Y, Nakachi K, 2008, RET/PTC rearrangements preferentially occurred in papillary thyroid Cancer among atomic bomb survivors exposed to high radiation dose. Cancer Res 68:7176–7182 , DOI 10.1158/0008- 5472.CAN-08-0293 Bounacer A, Wicker R, Schlumberger M, Sarasin A, Suarez HG, 1997, Oncogenic rearrangements of the ret proto-oncogene in thyroid tumors induced after exposure to ionizing radiation. Biochimie 79:619–623. , DOI 10.1016/S0300-9084(97)82012-3 Mayr B, Potter E, Goretzki P, Ruschoff J, Dietmaier W, Hoang-Vu C, Dralle H, Brabant G, 1998, Expression of ret/PTC1, −2, −3, −delta3 and −4 in German papillary thyroid carcinoma. Br J Cancer 77:903–906 Zou M, Shi Y, Farid NR, 1994, Low rate of ret proto-oncogene activation, PTC/retTPC, in papillary thyroid carcinomas from Saudi Arabia. Cancer 73:176–180 Lee CH, Hsu LS, ChiCWet al, 1998, High frequency of rearrangement of the RET protooncogene, RET/PTC, in Chinese papillary thyroid carcinomas. J Clin Endocrinol Metab 83:1629–1632. , DOI 10.1210/jcem.83.5.4774 Chua EL, Wu WM, Tran KT et al, 2000, Prevalence and distribution of ret/ptc 1, 2, and 3 in papillary thyroid carcinoma in New Caledonia and Australia. J Clin Endocrinol Metab 85:2733–2739. , DOI 10.1210/jcem.85.8.6722 Detection of RET/PTC oncogene rearrangements in Korean papillary thyroid carcinomas. - PubMed - NCBI. https://www.ncbi.nlm. nih.gov/pubmed/10646664. Accessed 5 Apr 2018 Wajjwalku W, Nakamura S, Hasegawa Y, Miyazaki K, Satoh Y, Funahashi H, Matsuyama M, Takahashi M, 1992, Low frequency of rearrangements of the ret and trk proto-oncogenes in Japanese thyroid papillary carcinomas. Jpn J Cancer Res Gann 83:671–675 Smida J, Salassidis K, Hieber L, Zitzelsberger H, Kellerer AM, Demidchik EP, Negele T, Spelsberg F, Lengfelder E, Werner M, Bauchinger M, 1999, Distinct frequency of ret rearrangements in papillary thyroid carcinomas of children and adults from Belarus. Int J Cancer 80:32–38 Gandhi M, Dillon LW, Pramanik S, Nikiforov YE, Wang YH, 2010, DNA breaks at fragile sites generate oncogenic RET/PTC rearrangements in human thyroid cells. Oncogene 29:2272–2280 , DOI 10.1038/onc.2009.502 Adeniran AJ, Zhu Z, Gandhi M, Steward DL, Fidler JP, Giordano TJ, Biddinger PW, Nikiforov YE, 2006, Correlation between genetic alterations and microscopic features, clinical manifestations, and prognostic characteristics of thyroid papillary carcinomas. Am J Surg Pathol 30:216–222 Su X, Li Z, He C, Chen W, Fu X, Yang A, 2016, Radiation exposure, young age, and female gender are associated with high prevalence of RET/PTC1 and RET/PTC3 in papillary thyroid cancer: a meta-analysis. Oncotarget 7:16716–16730. , DOI 10. 18632/oncotarget.7574 Tallini G, Asa SL, 2001, RET oncogene activation in papillary thyroid carcinoma. Adv Anat Pathol 8:345–354 Grubbs EG, Ng PK-S, Bui J, Busaidy NL, Chen K, Lee JE, Lu X, Lu H, Meric-Bernstam F, Mills GB, Palmer G, Perrier ND, Scott KL, Shaw KR, Waguespack SG, Williams MD, Yelensky R, Cote GJ, 2015, RET fusion as a novel driver of medullary thyroid carcinoma. J Clin Endocrinol Metab 100:788–793. , DOI 10. 1210/jc.2014-4153 Guerra A, Sapio MR, Marotta V et al, 2011, Prevalence of RET/ PTC rearrangement in benign and malignant thyroid nodules and its clinical application. Endocr J 58:31–38 Wirtschafter A, Schmidt R, Rosen D, Kundu N, Santoro M, Fusco A, Multhaupt H, Atkins JP, Rosen MR, Keane WM, Rothstein JL, 1997, Expression of the RET/PTC fusion gene as a marker for papillary carcinoma in Hashimoto’s thyroiditis. Laryngoscope 107:95–100 Sheils OM, O’Leary JJ, Uhlmann Vet al, 2000, Ret/PTC-1 activation in Hashimoto thyroiditis. Int J Surg Pathol 8:185–189. https:// doi.org/10.1177/106689690000800305 Basolo F, Molinaro E, Agate L, Pinchera A, Pollina L, Chiappetta G,Monaco C, Santoro M, Fusco A, Miccoli P, Elisei R, Capezzone M, Pacini F, 2001, RET protein expression has no prognostic impact on the long-term outcome of papillary thyroid carcinoma. Eur J Endocrinol 145:599–604 Puxeddu E, Moretti S, Giannico A, MartinelliM,Marino C, Avenia N, Cristofani R, Farabi R, Reboldi G, Ribacchi R, Pontecorvi A, Santeusanio F, 2003, Ret/PTC activation does not influence clinical and pathological features of adult papillary thyroid carcinomas. Eur J Endocrinol 148:505–513 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 507 EP 513 DI 10.1007/s12253-018-0540-3 PG 7 ER PT J AU El-Bolkainy, T Mohamed, G Badawy, O AF El-Bolkainy, Tarek Mohamed, Ghada Badawy, Omnia TI Precursor Lesions of the Vocal Cord: a Study on the Diagnostic Role of Histomorphology, Histometry and Ki-67 Proliferation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Laryngeal cancer; Dysplasia; Dyskeratosis; Histometry; Ki-67 labeling index ID Laryngeal cancer; Dysplasia; Dyskeratosis; Histometry; Ki-67 labeling index AB The precise typing of precursor lesions of squamous cell carcinoma of vocal cord is of vital importance since it determines the line of therapy and prognosis. The aim of the present study is to evaluate the possible value of the types of dyskeratosis, histometry and cell proliferation rate in discriminating these lesions. The present retrospective study was based on 145 patients, classified according to the updated 2017 WHO system and included: Low-grade dysplasia (24 cases), high-grade dysplasia (53 cases), carcinoma in situ (33 cases) and microinvasive carcinomas (35 cases). Cell proliferation was assessed by immunoreactivity to Ki-67. For histometry and quantitation of Ki-67 proliferation rate, an image analysis system was used (Leica LAS, Wetzlar, Germany). Epithelial pearls (cell nests) were commonly observed in microinvasive carcinoma (82.9%) than high-grade dysplasia (5.9%). The median epithelial thickness, as well as, proliferation rate showed a significant increase according to the grade of the lesion. It is concluded that dyskeratosis pattern, histometry and Ki-67proliferation rate are valuable parameters to characterize precursor lesions. The presence of epithelial pearls, thickness > 450 μm and Ki-67 > 40% denote high risk lesions that require adequate excision and/or radiotherapy. C1 [El-Bolkainy, Tarek] Cairo University, National Cancer Institute, Department of PathologyGiza, Egypt. [Mohamed, Ghada] Cairo University, National Cancer Institute, Department of PathologyGiza, Egypt. [Badawy, Omnia] Cairo University, National Cancer Institute, Department of PathologyGiza, Egypt. RP Badawy, O (reprint author), Cairo University, National Cancer Institute, Department of Pathology, Giza, Egypt. EM omniabadawy75@gmail.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:E359–E386 Globocan, 2012, Estimated Cancer incidence, mortality and prevalence worldwide: Sources, methods and major patterns. http:// globocan.iarc.fr/Pages/fact_sheets_population.aspx?country=818 Kumar V, Abbas A, Aster J, 2018, Robbins basic pathology, 10th edn. Elsevier Inc, Philadelphia, pp 546–547 Fletcher C, ed,, 2013, Diagnostic histopathology of tumors, 4th edn. Elsevier, Saunders, Philadelphia, p 171 Nayar R,WilburD, eds,, 2015, The Bethesda system for Reporting cervical cytology, ed 3. Definitions, criteria, and explanatory notes. Springer, New York Barnes L, Eveson IW, Reichart P, Sidransky D, eds,, 2005, WHO pathology and genetics, head and neck tumors, 3rd edition, p120, IARC. Lyon, France El-Naggar AK, Chan JKC, Grandis JR, Takata T, Slootweg PJ, 2017, WHO classification of head and neck tumors, 4th edition, p 91–93. IARC, Lyon Gnepp DR, ed,, 2009, Diagnostic surgical pathology of head and neck, 2nd edn. Saunders Elsevier, Philadelphia Gale N, Michaels L, Luzar B, Poljak M, Zidar N, Fischinger J, Cardesa A, 2009, Current review on squamous intraepithelial lesions of the larynx. Histopathology 54:639–656 Gale N, Blagus R, El-Mofty S et al, 2014, Evaluation of a new grading system for laryngeal squamous intraepithelial lesions—a proposed unified classification. Histopathology 65:456–464 Grissman JD, Zarbo RJ, Drozdowicz S, 1988, Carcinoma in situ and microinvasive squamous carcinoma of the laryngeal glottis. Arch Otolaryngol Head Neck Surg 114:299–307 Barnes L, ed,, 2009, Surgical pathology of head and neck, 3rd edn. Informa Health care, New York Arens C, Glanz H, Wonckhaus J, Hersemeyer K, Kraft M, 2007, Histologic assessment of epithelial thickness in early laryngeal cancer or precursor lesions and its impact on endoscopic imaging. Eur Arch Otorhinolaryngol 264:645–649 Mondal D, Saha K, Datta C, Chatterjee U, Sengupta A, 2013, Ki- 67, p27, p53 expression in squamous epithelial lesions of larynx. Indian J Otolaryngol Head Neck Surg 65:126–133 Amin MB et al, 2017, AJCC cancer staging manual. Springer- Verlag, New York Weller MD, Nankivell PC, McConkey C, Paleri V, Mehanna HM, 2012, The risk and interval to malignancy of patients with laryngeal dysplasia; a systematic review of case series andmeta-analysis. Clin Otolaryngol 35(5):364–372 Fleskens SA, Bergshoeff VE, Voogd AC et al, 2011, Interobserver variability of laryngeal mucosal premalignant lesions: a histopathological evaluation. Mod Pathol 24:892–898 Sarioglu S, Cakalagaoglu F, Elagoz S et al, 2010, Inter-observer agreement in laryngeal pre-neoplastic lesions. Head Neck Pathol 4: 276–280 McLaren KM, Burnett RA, Goodlad JR et al, 2000, Consistency of histopathological reporting of laryngeal dysplasia. Histopathology 37:460–463 Hirano M, 1974, Morphological structure of the vocal cord as a vibrator and its variants as a vibrator and its variations. Folia Phoniatr, Basel, 26:89–94 Kalter OP, Delemarre JFM, Alons CL, Veldhuizen RW, Meyer C, Snow GB, 1985, Quantitative morphometry of squamous cell hyperplasia of the larynx. J Clin Pathol 38:489–495 COr A, Gale N, Kambic V, 1997, Quantitative pathology of laryngeal epithelial hyperplastic lesions. Acta Otolaryngol Suppl 527: 57–61 Ashraf MJ, Maghbul M, Azapira N et al, 2010, Expression of Ki- 67 and p53 in primary squamous cell carcinoma of the larynx. Indian J Pathol Microbiol 53:661–665 Sengiz S, Pabuccuoglu U, Sarioglu S, 2004, Immunohistological comparison of the World Health Organisation, WHO, and Ljubljana classifications on the grading of preneoplastic lesions of the larynx. Pathol Res Pract 200:181–188 Pavlovic B, Djukic V, Milovanovic J, Tomanovic N, Milovanovic A, Trivic A, 2013, Morphometric analysis of Ki-67 and p16 expression in laryngeal precursor lesions. Eur Arch Otorhinolaryngol 270:1405–1410 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 515 EP 520 DI 10.1007/s12253-018-0560-z PG 6 ER PT J AU Yan, Y Zhang, L Tan, L Ma, X Yong, Z Shao, Sh Liu, J Xue, Ch Li, Z Zhang, X Drokow, KE Shi, X Ren, J AF Yan, Yanli Zhang, Long Tan, Li Ma, Xiaowei Yong, Zhang Shao, Shuai Liu, Jiaxin Xue, Chaofan Li, Zongfang Zhang, Xiaozhi Drokow, Kwateng Emmanuel Shi, Xiaoting Ren, Juan TI Endocrine Therapy for Ductal Carcinoma In Situ (DCIS) of the Breast with Breast Conserving Surgery (BCS) and Radiotherapy (RT): a Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ductal carcinoma in situ (DCIS); Endocrine therapy; Meta-analysis ID Ductal carcinoma in situ (DCIS); Endocrine therapy; Meta-analysis AB The management of ductal carcinoma in situ (DCIS) with endocrine therapy remains controversial. A meta-analysis was conducted to evaluate the role of endocrine therapy for DCIS with breast conserving surgery (BCS) and radiotherapy (RT). A total of 7 articles with randomized controlled trials were included. Five articles compared the effects of BCS and RT followed by tamoxifen (TAM) or not (BCS + RT + TAM vs BCS + RT) and 2 compared the effects of TAM and anastrozole (ANA). TAM obviously reduced the rates of recurrence of ipsilateral breast cancer (IBCR), recurrence of contralateral breast cancer (CBCR), recurrence of ipsilateral invasive breast cancer (IBCR-INV) and recurrence of contralateral DCIS (CBCR-DCIS), and increased the rate of event-free survival (EFS). While ANA reduced the rates of CBCR and recurrence of contralateral invasive breast cancer (CBCR-INV). Patients with ANA had higher incidence of arthralgia, osteoporosis, hypercholesteremia, headache and vaginal dryness, but lower incidence of deep-vein thrombosis, pulmonary embolism, vasomotor or gynaecological, hot flushes, vaginal haemorrhage, vaginal discharge and vaginal candidiasis than TAM. In conclusion, DCIS patients with positive hormone receptors should be recommended to receive endocrine therapy. Selection of TAM or ANA is based on clinical characteristics and underlying disease of patients, as well as the side-effects of drugs. C1 [Yan, Yanli] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi Province, China. [Zhang, Long] First Affliated Hospital, Xi’an Jiaotong University, Department of Oncology Radiotherapy, 710061 Xi’an, Shaanxi Province, China. [Tan, Li] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi Province, China. [Ma, Xiaowei] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi Province, China. [Yong, Zhang] Xi’an Children Hospital, Department of Orthopedics, 710000 Xi’an, Shaanxi Province, China. [Shao, Shuai] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi Province, China. [Liu, Jiaxin] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi Province, China. [Xue, Chaofan] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi Province, China. [Li, Zongfang] Second Affiliated Hospital of Xi’an Jiaotong University, Department of Surgery, 710004 Xi’an, Shaanxi Province, China. [Zhang, Xiaozhi] First Affliated Hospital, Xi’an Jiaotong University, Department of Oncology Radiotherapy, 710061 Xi’an, Shaanxi Province, China. [Drokow, Kwateng Emmanuel] Xi’an Jiaotong University, First Affiliated Hospital of Medical College, 710061 Xi’an, Shaanxi Province, China. [Shi, Xiaoting] First Affliated Hospital, Xi’an Jiaotong University, Department of Oncology Radiotherapy, 710061 Xi’an, Shaanxi Province, China. [Ren, Juan] First Affliated Hospital, Xi’an Jiaotong University, Department of Oncology Radiotherapy, 710061 Xi’an, Shaanxi Province, China. RP Ren, J (reprint author), First Affliated Hospital, Xi’an Jiaotong University, Department of Oncology Radiotherapy, 710061 Xi’an, China. EM 869491533@qq.com CR James TA, Wade JE, Sprague BL, 2016, The impact of mammographic screening on the surgical management of breast cancer. J Surg Oncol 113:496–500. , DOI 10.1002/jso.24184 Torre LA, Sauer AM, Chen MS Jr, Kagawa-Singer M, Jemal A, Siegel RL, 2016, Cancer statistics for Asian Americans, native Hawaiians, and Pacific islanders, 2016: converging incidence in males and females. CA Cancer J Clin 66:182–202. , DOI 10.3322/caac.21335 Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66(1):7–30. , DOI 10.3322/caac.21332 Mitchell KB, Kuerer H, 2015, Ductal carcinoma in situ: treatment update and current trends. Curr Oncol Rep 17(11):48. https://doi. org/10.1007/s11912-015-0473-x Lebeau A, Kuhn T, 2016, Updates in the treatment of ductal carcinoma in situ of the breast. Curr Opin Obstet Gynecol 28(1):49–58. , DOI 10.1097/GCO.0000000000000237 Staley H, McCallum I, Bruce J, 2014, Postoperative tamoxifen for ductal carcinoma in situ: Cochrane systematic review and metaanalysis. Breast, Edinburgh, Scotland, 23(5):546–551. https://doi. org/10.1016/j.breast.2014.06.015 Houghton J, GeorgeWD, Cuzick J, Duggan C, Fentiman IS, Spittle M, 2003, Radiotherapy and tamoxifen in women with completely excised ductal carcinoma in situ of the breast in the UK, Australia, and New Zealand: randomised controlled trial. Lancet 362(9378): 95–102 Cuzick J, Sestak I, Pinder SE, Ellis IO, Forsyth S, Bundred NJ, Forbes JF, Bishop H, Fentiman IS, George WD, 2011, Effect of tamoxifen and radiotherapy in women with locally excised ductal carcinoma in situ: long-term results from the UK/ANZ DCIS trial. Lancet Oncol. 12(1):21–29. , DOI 10.1016/S1470- 2045(10)70266-7 Baum M, Budzar AU, Cuzick J, Forbes J, Houghton JH, Klijn JG, Sahmoud T, Group AT, 2002, Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomised trial. Lancet 359(9324):2131–2139 Howell A, Cuzick J, Baum M, Buzdar A, Dowsett M, Forbes JF, Hoctin-Boes G, Houghton J, Locker GY, Tobias JS, Group AT, 2005, Results of the ATAC, Arimidex, tamoxifen, alone or in combination, trial after completion of 5 years' adjuvant treatment for breast cancer. Lancet 365(9453):60–62. , DOI 10.1016/ S0140-6736(04)17666-6 Breast International Group 1-98 Collaborative G, Thurlimann B, Keshaviah A, Coates AS, Mouridsen H, Mauriac L, Forbes JF, Paridaens R, Castiglione-Gertsch M, Gelber RD, Rabaglio M, Smith I,Wardley A, Price KN, Goldhirsch A, 2005, A comparison of letrozole and tamoxifen in postmenopausal women with early breast cancer. N Engl J Med 353(26):2747–2757. , DOI 10.1056/NEJMoa052258 Gradishar WJ, Anderson BO, Balassanian R, Blair SL, Burstein HJ, Cyr A, Elias AD, Farrar WB, Forero A, Giordano SH, 2017, NCCN guidelines insights: breast Cancer, version 1.2017. Journal of the National Comprehensive Cancer Network. Jnccn 15(4):433– 451 Margolese RG, Cecchini RS, Julian TB, Ganz PA, Costantino JP, Vallow LA, Albain KS, Whitworth PW, Cianfrocca ME, Brufsky AM, Gross HM, Soori GS, Hopkins JO, Fehrenbacher L, Sturtz K, Wozniak TF, Seay TE, Mamounas EP, Wolmark N, 2015, Anastrozole versus tamoxifen in postmenopausal women with ductal carcinoma in situ undergoing lumpectomy plus radiotherapy, NSABP B-35): a randomised, double-blind, phase 3 clinical trial. Lancet 387:849–856. , DOI 10.1016/S0140-6736(15, 01168-X Forbes JF, Sestak I, Howell A, Bonanni B, Bundred N, Levy C, von Minckwitz G, Eiermann W, Neven P, Stierer M, Holcombe C, Coleman RE, Jones L, Ellis I, Cuzick J, investigators I-I, 2015, Anastrozole versus tamoxifen for the prevention of locoregional and contralateral breast cancer in postmenopausal women with locally excised ductal carcinoma in situ, IBIS-II DCIS): a doubleblind, randomised controlled trial. Lancet. , DOI 10.1016/ S0140-6736(15)01129-0 KF S, DG A, D M, Group C, 2010, CONSORT 2010 statement: updated guidelines for reporting parallel group randomized trials. Ann Intern Med 8(5):18 Lu J, ZhaoQ, Zhai YJ, He HR, Yang LH,Gao F, Zhou RS, Zheng J, Ma XC, 2015, Genetic polymorphisms of CYP1A1 and risk of leukemia: a meta-analysis. OncoTargets Ther 8:2883–2902. , DOI 10.2147/OTT.S92259 Fisher B, Dignam J, Wolmark N, Wickerham DL, Fisher ER, Mamounas E, Smith R, Begovic M, Dimitrov NV, Margolese RG, Kardinal CG, Kavanah MT, Fehrenbacher L, Oishi RH, 1999, Tamoxifen in treatment of intraductal breast cancer: National Surgical Adjuvant Breast and bowel project B-24 randomised controlled trial. Lancet, London, England, 353(9169):1993–2000. , DOI 10.1016/S0140-6736(99, 05036-9 Fisher B, Land S, Mamounas E, Dignam J, Fisher ER,Wolmark N, 2001, Prevention of invasive breast cancer in women with ductal carcinoma in situ: an update of the National Surgical Adjuvant Breast and bowel project experience. Semin Oncol 28(4):400–418 Wapnir IL, Dignam JJ, Fisher B, Mamounas EP, Anderson SJ, Julian TB, Land SR, Margolese RG, Swain SM, Costantino JP, Wolmark N, 2011, Long-term outcomes of invasive ipsilateral breast tumor recurrences after lumpectomy in NSABP B-17 and B-24 randomized clinical trials for DCIS. J Natl Cancer Inst 103(6):478–488. , DOI 10.1093/jnci/djr027 Barnes NL, Boland GP, Davenport A, Knox WF, Bundred NJ, 2005, Relationship between hormone receptor status and tumour size, grade and comedo necrosis in ductal carcinoma in situ. Br J Surg 92(4):429–434. , DOI 10.1002/bjs.4878 Claus EB, Chu P, Howe CL, Davison TL, Stern DF, Carter D, DiGiovanna MP, 2001, Pathobiologic findings in DCIS of the breast: morphologic features, angiogenesis, HER-2/neu and hormone receptors. Exp Mol Pathol 70(3):303–316. , DOI 10.1006/exmp.2001.2366 Allred DC, Anderson SJ, Paik S, Wickerham DL, Nagtegaal ID, Swain SM, Mamounas EP, Julian TB, Geyer CE Jr, Costantino JP, Land SR, Wolmark N, 2012, Adjuvant tamoxifen reduces subsequent breast cancer in women with estrogen receptor-positive ductal carcinoma in situ: a study based on NSABP protocol B-24. J Clin Oncol 30(12):1268–1273. , DOI 10.1200/JCO.2010. 34.0141 Cuzick J, Sestak I, Baum M, Buzdar A, Howell A, Dowsett M, Forbes JF, 2010, Effect of anastrozole and tamoxifen as adjuvant treatment for early-stage breast cancer: 10-year analysis of the ATAC trial. Lancet Oncol 11(12):1135–1141. , DOI 10. 1016/s1470-2045(10)70257-6 Menopausal hormone use and ovarian cancer risk: individual participant meta-analysis of 52 epidemiological studies, 2015). The Lancet 385, 9980):1835–1842. , DOI 10.1016/s0140- 6736(14)61687-1 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 521 EP 531 DI 10.1007/s12253-018-0553-y PG 11 ER PT J AU Li, W Meng, Z Zou, T Wang, G Su, Y Yao, Sh Sun, X AF Li, Weichao Meng, Zengdong Zou, Tiannan Wang, Gang Su, Yijing Yao, Shaoping Sun, Xianrun TI MiR-374a Activates Wnt/β-Catenin Signaling to Promote Osteosarcoma Cell Migration by Targeting WIF-1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma (OS); miR-374a; Migration; Wnt inhibitory factor-1 (WIF-1); Wnt/β-catenin; Signaling pathway ID Osteosarcoma (OS); miR-374a; Migration; Wnt inhibitory factor-1 (WIF-1); Wnt/β-catenin; Signaling pathway AB MiR-374a was proved to take part in the initiation and development of several cancers. However, the molecular mechanism of miR-374a in osteosarcoma (OS) cells remains unclear. The aim of our research was to investigate the role of miR-374a in OS cells migration and clarify the potential mechanisms. Quantitative real-time PCR (qRT-PCR) and western blot analysis were applied to evaluate the expression of miR-374a and Wnt inhibitory factor-1 (WIF-1). Bioinformatical methods and luciferase reporter assay were carried out to predict and confirm the combination of miR-374a and WIF-1. Transwell and wound healing assays were performed to detect the migration capacity of OS cells. Lithium chloride (LiCl) was used to investigate the role of LiCl-activated Wnt/β-catenin signaling pathway in regulating cell migration. Our studies revealed that miR-374a was upregulated whereas WIF-1 was down-regulated in OS cells. Besides, WIF-1 was the target of miR-374a by performing luciferase reporter assay. By transfection of miR-374a inhibitor and/or WIF-1 siRNA to OS cells, we found that miR-374a promoted the migration of OS cells. In addition, the inhibition of WIF-1 abolished the miR-374a inhibitor-induced migration suppression of OS cells. LiCl experiment revealed that miR-374a promoted OS cells migration by regulating Wnt/β-catenin signaling. In conclusion, miR-374a promotes OS cells migration by activating Wnt/β-catenin signaling pathway via targeting WIF-1. C1 [Li, Weichao] Kunming University of Science and Technology, Medical School, 650500 Kunming, China. [Meng, Zengdong] The First Affiliated Hospital of Kunming Medical College, Department of Surgery, No. 157 Jinbi Road, 650032 Kunming, China. [Zou, Tiannan] The First Affiliated Hospital of Kunming Medical College, Department of Surgery, No. 157 Jinbi Road, 650032 Kunming, China. [Wang, Gang] The First Affiliated Hospital of Kunming Medical College, Department of Surgery, No. 157 Jinbi Road, 650032 Kunming, China. [Su, Yijing] The First Affiliated Hospital of Kunming Medical College, Department of Surgery, No. 157 Jinbi Road, 650032 Kunming, China. [Yao, Shaoping] The First Affiliated Hospital of Kunming Medical College, Department of Surgery, No. 157 Jinbi Road, 650032 Kunming, China. [Sun, Xianrun] The First Affiliated Hospital of Kunming Medical College, Department of Surgery, No. 157 Jinbi Road, 650032 Kunming, China. RP Yao, Sh (reprint author), The First Affiliated Hospital of Kunming Medical College, Department of Surgery, 650032 Kunming, China. EM gukeyshp@sina.com CR Misaghi A, Goldin A, Awad M, Kulidjian AA, 2018, Osteosarcoma: a comprehensive review. SICOT J 4:12 Namlos HM, Meza-Zepeda LA, Baroy T, Ostensen IH, Kresse SH, Kuijjer ML, Serra M, Burger H, Cleton-Jansen AM, Myklebost O, 2012, Modulation of the osteosarcoma expression phenotype by microRNAs. PLoS One 7:e48086 Kumar R, Kumar M, Malhotra K, Patel S, 2018, Primary osteosarcoma in the elderly revisited: current concepts in diagnosis and treatment. Curr Oncol Rep 20:13 Simpson S, Dunning MD, de Brot S, Grau-Roma L, Mongan NP, Rutland CS, 2017, Comparative review of human and canine osteosarcoma: morphology, epidemiology, prognosis, treatment and genetics. Acta Vet Scand 59:71 Zhang Y, Yang Q, Wang S, 2014, MicroRNAs: a new key in lung cancer. Cancer Chemother Pharmacol 74:1105–1111 Yang X, Wang L, Wang Q, Li L, Fu Y, Sun J, 2018, MiR-183 inhibits osteosarcoma cell growth and invasion by regulating LRP6-Wnt/beta-catenin signaling pathway. Biochem Biophys Res Commun 496:1197–1203 Yang Z, Li X, Yang Y, He Z, Qu X, Zhang Y, 2016, Long noncoding RNAs in the progression, metastasis, and prognosis of osteosarcoma. Cell Death Dis 7:e2389 Chen Y, Jiang J, Zhao M, Luo X, Liang Z, Zhen Y, Fu Q, Deng X, Lin X, Li L, 2016, microRNA-374a suppresses colon cancer progression by directly reducing CCND1 to inactivate the PI3K/AKT pathway. Oncotarget 7:41306–41319 Cai J, Guan H, Fang L, Yang Y, Zhu X, Yuan J,Wu J, Li M, 2013, MicroRNA-374a activates Wnt/beta-catenin signaling to promote breast cancer metastasis. J Clin Invest 123:566–579 Wang Y, Xin H, Han Z, Sun H, Gao N, Yu H, 2015, MicroRNA- 374a promotes esophageal cancer cell proliferation via Axin2 suppression. Oncol Rep 34:1988–1994 Lee SH, Koo BS, Kim JM, Huang S, Rho YS, Bae WJ, Kang HJ, Kim YS, Moon JH, Lim YC, 2014, Wnt/beta-catenin signalling maintains self-renewal and tumourigenicity of head and neck squamous cell carcinoma stem-like cells by activating Oct4. J Pathol 234:99–107 MohammedMK, Shao C,Wang J,Wei Q,Wang X, Collier Z, Tang S, Liu H, Zhang F, Huang J, Guo D, Lu M, Liu F, Liu J, Ma C, Shi LL, Athiviraham A, He TC, Lee MJ, 2016, Wnt/beta-catenin signaling plays an ever-expanding role in stem cell self-renewal, tumorigenesis and cancer chemoresistance. Genes Dis 3:11–40 Yang F, Zeng Q, Yu G, Li S, Wang CY, 2006, Wnt/beta-catenin signaling inhibits death receptor-mediated apoptosis and promotes invasive growth of HNSCC. Cell Signal 18:679–687 Lin B, Hong H, Jiang X, Li C, Zhu S, TangN,Wang X, She F, Chen Y, 2017)WNT inhibitory factor 1 promoter hypermethylation is an early event during gallbladder cancer tumorigenesis that predicts poor survival. Gene 622:42–49 Tang Q, Zhao H, Yang B, Li L, Shi Q, Jiang C, Liu H, 2017, WIF-1 gene inhibition and Wnt signal transduction pathway activation in NSCLC tumorigenesis. Oncol Lett 13:1183–1188 Malinauskas T, Aricescu AR, Lu W, Siebold C, Jones EY, 2011, Modular mechanism ofWnt signaling inhibition byWnt inhibitory factor 1. Nat Struct Mol Biol 18:886–893 Huang Y, Du Q,WuW, She F, Chen Y, 2016, Rescued expression of WIF-1 in gallbladder cancer inhibits tumor growth and induces tumor cell apoptosis with altered expression of proteins. Mol Med Rep 14:2573–2581 Ma Y, Zhu B, Liu X, Yu H, Yong L, Liu X, Shao J, Liu Z, 2015, Inhibition of oleandrin on the proliferation show and invasion of osteosarcoma cells in vitro by suppressingWnt/beta-catenin signaling pathway. J Exp Clin Cancer Res 34:115 Yu M, Guo D, Cao Z, Xiao L, Wang G, 2018, Inhibitory effect of MicroRNA-107 on osteosarcoma malignancy through regulation of Wnt/beta-catenin signaling in vitro. Cancer Investig 36:175–184 Jiang Z, Jiang C, Fang J, 2018, Up-regulated lnc-SNHG1 contributes to osteosarcoma progression through sequestration ofmiR-577 and activation of WNT2B/Wnt/beta-catenin pathway. Biochem Biophys Res Commun 495:238–245 Poos K, Smida J, Maugg D, Eckstein G, Baumhoer D, Nathrath M, Korsching E, 2015, Genomic heterogeneity of osteosarcoma - shift from single candidates to functional modules. PLoS One 10: e0123082 Li X, Lu Q, Xie W, Wang Y, Wang G, 2018, Anti-tumor effects of triptolide on angiogenesis and cell apoptosis in osteosarcoma cells by inducing autophagy via repressing Wnt/beta-catenin signaling. Biochem Biophys Res Commun 496:443–449 He W, Feng L, Xia D, Han N, 2014, MiR-374a promotes the proliferation of human osteosarcoma by downregulating FOXO1 expression. Int J Clin Exp Med 8:3482–3489 Yiyu Chen JJ, Zhao M, Luo X, Liang Z, Zhen Y, Qiaofen F, Deng X, Lin X, Li L, Luo R, Liu Z, Fang W, 2016, microRNA-374a suppresses colon cancer progression by directly reducing CCND1 to inactivate the PI3K/AKT pathway. Oncotarget 7:41306–41319 Vosa U, Vooder T, Kolde R, Fischer K, Valk K, Tonisson N, Roosipuu R, Vilo J, Metspalu A, Annilo T, 2011, Identification of miR-374a as a prognostic marker for survival in patients with early-stage nonsmall cell lung cancer. Genes Chromosomes Cancer 50:812–822 Wei B, Guo Y, Zhai J, Su J, Han L, Kang C, Zhang Q, 2013, A study of the relationship between the Wnt/betacatenin signaling pathway and the gastrointestinal development of rat embryonic and perinatal periods. Exp Ther Med 5:1598–1602 Tang L, Wang D, Gu D, 2018, Knockdown of Sox2 inhibits OS cells invasion and migration via modulating Wnt/beta-catenin signaling pathway. Pathol Oncol Res 24:907–913 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 533 EP 539 DI 10.1007/s12253-018-0556-8 PG 7 ER PT J AU Xuan, Z Huang, J Gao, L Wang, Y Wang, J Sun, Y AF Xuan, Zhuoqi Huang, Jianming Gao, Lin Wang, Yong Wang, Jiandong Sun, Yueming TI Receptor Tyrosine Kinase EphB3: a Prognostic Indicator in Colorectal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; EphB3; Hypermethylation; Lymph node metastasis; TNM stage ID Colorectal cancer; EphB3; Hypermethylation; Lymph node metastasis; TNM stage AB Although EphB3 expression is down-regulated in colorectal cancer (CRC) cells compared with normal intestinal epithelial cells, the relationship between EphB3 expression and clinicopathological parameters in CRC is unclear. We examined EphB3 expression in 128 CRC tissue specimens and in 19 adenoma specimens using immunohistochemistry. The relationships between EphB3 expression and clinicopathological parameters, KRAS mutations, BRAF V600E mutation, MSI and survival were evaluated using Spearman’s rank correlation and Kaplan–Meier survival analyses, respectively. CpG methylation in the EphB3 promoter was examined in four human CRC cell lines and tissues. EphB3 was strongly expressed in all normal intestinal epithelial cells (128/128) and adenoma cells (19/19). In CRC tumor cells, EphB3 expression was negative or weak in 41.4% (53/128), moderate in 26.6% (34/128), and strong in 32.0% (41/128) of samples. EphB3 expression was negatively associated with invasive depth (P = 0.016, rs = −0.213), lymph node metastasis (P = 0.000, rs = −0.490), and TNM stage (P = 0.000, rs = −0.390), and was positively associated with poor differentiation (P = 0.001, rs = 0.290), BRAF V600E mutation (P = 0.008, rs = 0.235), and longer overall survival (P < 0.001). In multivariate analysis, EphB3 expression (P = 0.007) and lymph node metastasis (P < 0.001) were independent prognostic factors for poor survival. Hypermethylation of the EphB3 promoter was detected in cell lines and CRC tissues. EphB3 is down-regulated in CRC compared to normal mucosa. Hypermethylation of CpG island is contributed to downregulation of EphB3 in CRC. EphB3 expression in tumor cells may be a useful prognostic indicator for patients with CRC. C1 [Xuan, Zhuoqi] The First Affiliated Hospital of Nanjing Medical University, Department of Colorectal Surgery, 210029 Nanjing, Jiangsu, China. [Huang, Jianming] The Jiangyin People’s Hospital, Department of Gastrointestinal Surgery, 214400 Jinagsu, China. [Gao, Lin] The Jiangyin People’s Hospital, Department of Gastrointestinal Surgery, 214400 Jinagsu, China. [Wang, Yong] The First Affiliated Hospital of Nanjing Medical University, Department of Colorectal Surgery, 210029 Nanjing, Jiangsu, China. [Wang, Jiandong] Nanjing University School of Medicine, Jinling Hospital, Department of Pathology, No.305, Zhong San Dong Lu, 210002 Nanjing, China. [Sun, Yueming] The First Affiliated Hospital of Nanjing Medical University, Department of Colorectal Surgery, 210029 Nanjing, Jiangsu, China. RP Wang, J (reprint author), Nanjing University School of Medicine, Jinling Hospital, Department of Pathology, 210002 Nanjing, China. EM jd_wang@outlook.com CR (1997, Unified nomenclature for Eph family receptors and their ligands, the ephrins. Eph Nomenclature Committee. Cell 90(3): 403–404 Hirai H, Maru Y, Hagiwara K, Nishida J, Takaku F, 1987, A novel putative tyrosine kinase receptor encoded by the eph gene. Science 238(4834):1717–1720 Wilkinson DG, 2001, Multiple roles of EPH receptors and ephrins in neural development. Nat Rev Neurosci 2(3):155–164. https://doi. org/10.1038/35058515 Xu Q, Mellitzer G, Robinson V, Wilkinson DG, 1999, In vivo cell sorting in complementary segmental domains mediated by Eph receptors and ephrins. Nature 399(6733):267–271. , DOI 10.1038/20452 Pasquale EB, 2008, Eph-ephrin bidirectional signaling in physiology and disease. Cell 133(1):38–52. , DOI 10.1016/j.cell. 2008.03.011 Zeng G, Hu Z, Kinch MS, Pan CX, Flockhart DA, Kao C, Gardner TA, Zhang S, Li L, Baldridge LA, Koch MO, Ulbright TM, Eble JN, Cheng L, 2003, High-level expression of EphA2 receptor tyrosine kinase in prostatic intraepithelial neoplasia. Am J Pathol 163(6):2271–2276. , DOI 10.1016/S0002-9440(10, 63584-5 Thaker PH, Deavers M, Celestino J, Thornton A, Fletcher MS, Landen CN, Kinch MS, Kiener PA, Sood AK, 2004, EphA2 expression is associated with aggressive features in ovarian carcinoma. Clin Cancer Res 10(15):5145–5150. , DOI 10.1158/ 1078-0432.CCR-03-0589 Wang J, Kataoka H, Suzuki M, Sato N, Nakamura R, Tao H, Maruyama K, Isogaki J, Kanaoka S, Ihara M, Tanaka M, Kanamori M, Nakamura T, Shinmura K, Sugimura H, 2005, Downregulation of EphA7 by hypermethylation in colorectal cancer. Oncogene 24(36):5637–5647. , DOI 10.1038/sj.onc. 1208720 Chiu ST, Chang KJ, Ting CH, Shen HC, Li H, Hsieh FJ, 2009, Over-expression of EphB3 enhances cell-cell contacts and suppresses tumor growth in HT-29 human colon cancer cells. Carcinogenesis 30(9):1475–1486. , DOI 10.1093/carcin/ bgp133 Oricchio E, Nanjangud G, Wolfe AL, Schatz JH, Mavrakis KJ, Jiang M, Liu X, Bruno J, Heguy A, Olshen AB, Socci ND, Teruya-Feldstein J, Weis-Garcia F, Tam W, Shaknovich R, Melnick A, Himanen JP, Chaganti RS, Wendel HG, 2011, The Eph-receptor A7 is a soluble tumor suppressor for follicular lymphoma. Cell 147(3):554–564. , DOI 10.1016/j.cell.2011. 09.035 Saito T, Masuda N, Miyazaki T, Kanoh K, Suzuki H, Shimura T, Asao T, Kuwano H, 2004, Expression of EphA2 and E-cadherin in colorectal cancer: correlation with cancer metastasis. Oncol Rep 11(3):605–611 GaoW, Zhang Q,Wang Y,Wang J, Zhang S, 2017, EphB3 protein is associated with histological grade and FIGO stage in ovarian serous carcinomas. APMIS 125(2):122–127. , DOI 10. 1111/apm.12646 Herrem CJ, Tatsumi T, Olson KS, Shirai K, Finke JH, Bukowski RM, Zhou M, Richmond AL, Derweesh I, Kinch MS, Storkus WJ, 2005, Expression of EphA2 is prognostic of disease-free interval and overall survival in surgically treated patients with renal cell carcinoma. Clin Cancer Res 11(1):226–231 Brannan JM, DongW, Prudkin L, Behrens C, Lotan R, Bekele BN, Wistuba I, Johnson FM, 2009, Expression of the receptor tyrosine kinase EphA2 is increased in smokers and predicts poor survival in non-small cell lung cancer. Clin Cancer Res 15(13):4423–4430. , DOI 10.1158/1078-0432.CCR-09-0473 Wang L, Hu H, Tian F, ZhouW, Zhou S,Wang J, 2015, Expression of EphA2 protein is positively associated with age, tumor size and Fuhrman nuclear grade in clear cell renal cell carcinomas. Int J Clin Exp Pathol 8(10):13374–13380 Bai YQ, Zhang JY, Bai CY, Xu XE,Wu JY, Chen B,Wu ZY,Wang SH, Shen J, Shen JH, Yao XD, Gao LZ,Wu B, Gu HL, Liu XH, Li X, Li EM, Xu LY, 2015, Low EphA7 expression correlated with lymph node metastasis and poor prognosis of patients with esophageal squamous cell carcinoma. Acta Histochem Cytochem 48(3): 75–81. , DOI 10.1267/ahc.14054 Li S, Wu Z, Ma P, Xu Y, Chen Y, Wang H, He P, Kang Z, Yin L, Zhao Y, Zhang X, Xu X, Ma X, Guan M, 2017, Ligand-dependent EphA7 signaling inhibits prostate tumor growth and progression. Cell Death Dis 8(10):e3122. , DOI 10.1038/cddis.2017.507 Dawson DW, Hong JS, Shen RR, French SW, Troke JJ, Wu YZ, Chen SS, Gui D, Regelson M,Marahrens Y, Morse HC 3rd, Said J, Plass C, Teitell MA, 2007, Global DNA methylation profiling reveals silencing of a secreted form of Epha7 in mouse and human germinal center B-cell lymphomas. Oncogene 26(29):4243–4252. , DOI 10.1038/sj.onc.1210211 Bohme B, Holtrich U,Wolf G, Luzius H, Grzeschik KH, Strebhardt K, Rubsamen-Waigmann H, 1993, PCR mediated detection of a new human receptor-tyrosine-kinase, HEK 2. Oncogene 8(10): 2857–2862 Baker RK, Vanderboom AK, Bell GW, Antin PB, 2001, Expression of the receptor tyrosine kinase gene EphB3 during early stages of chick embryo development. Mech Dev 104(1–2):129–132 Kang JU, Koo SH, Kwon KC, Park JW, Kim JM, 2009, Identification of novel candidate target genes, including EPHB3, MASP1 and SST at 3q26.2-q29 in squamous cell carcinoma of the lung. BMC Cancer 9:237. , DOI 10.1186/1471-2407-9-237 Ji XD, Li G, Feng YX, Zhao JS, Li JJ, Sun ZJ, Shi S, Deng YZ, Xu JF, Zhu YQ, Koeffler HP, Tong XJ, Xie D, 2011, EphB3 is overexpressed in non-small-cell lung cancer and promotes tumor metastasis by enhancing cell survival and migration. Cancer Res 71(3):1156–1166. , DOI 10.1158/0008-5472.CAN-10- 0717 Li G, Ji XD, Gao H, Zhao JS, Xu JF, Sun ZJ, Deng YZ, Shi S, Feng YX, Zhu YQ,Wang T, Li JJ, Xie D, 2012, EphB3 suppresses nonsmall- cell lung cancer metastasis via a PP2A/RACK1/Akt signalling complex. Nat Commun 3:667. , DOI 10.1038/ ncomms1675 Batlle E, Henderson JT, Beghtel H, van den Born MM, Sancho E, Huls G, Meeldijk J, Robertson J, van de Wetering M, Pawson T, Clevers H, 2002, Beta-catenin and TCF mediate cell positioning in the intestinal epithelium by controlling the expression of EphB/ephrinB. Cell 111(2):251–263 Jagle S, Ronsch K, Timme S, Andrlova H, Bertrand M, Jager M, Proske A, SchremppM, Yousaf A,Michoel T, Zeiser R,Werner M, Lassmann S, Hecht A, 2014, Silencing of the EPHB3 tumorsuppressor gene in human colorectal cancer through decommissioning of a transcriptional enhancer. Proc Natl Acad Sci U S A 111(13):4886–4891. , DOI 10.1073/pnas. 1314523111 Ronsch K, Jager M, Schopflin A, Danciu M, Lassmann S, Hecht A, 2011, Class I and III HDACs and loss of active chromatin features contribute to epigenetic silencing of CDX1 and EPHB tumor suppressor genes in colorectal cancer. Epigenetics 6(5):610–622 Batlle E, Bacani J, Begthel H, Jonkheer S, Gregorieff A, van de Born M,Malats N, Sancho E, Boon E, Pawson T, Gallinger S, Pals S, Clevers H, 2005, EphB receptor activity suppresses colorectal cancer progression. Nature 435(7045):1126–1130. , DOI 10.1038/nature03626 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 541 EP 549 DI 10.1007/s12253-018-0562-x PG 9 ER PT J AU Niyaz, M Khan, SM Wani, AR Shah, JO Mudassar, S AF Niyaz, Madiha Khan, S Mosin Wani, A Rauf Shah, J Omar Mudassar, Syed TI Sonic Hedgehog Protein is Frequently Up-Regulated in Pancreatic Cancer Compared to Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sonic hedgehog; Hedgehog signalling pathway; Protein expression; Western blotting; Colorectal cancer; Pancreatic cancer ID Sonic hedgehog; Hedgehog signalling pathway; Protein expression; Western blotting; Colorectal cancer; Pancreatic cancer AB Sonic hedgehog (SHH) is a secreted protein which functions in autocrine or paracrine fashion on target cells to activate hedgehog (HH) signalling cascade responsible for growth and proliferation. This study is an attempt to understand the expression dynamics of SHH protein in colon, rectal and pancreatic cancers. Protein expression of SHH was studied by Western Blotting in the histologically confirmed colon, rectum and pancreatic cancer tissue samples along with their adjacent normal tissues. Only 31.4% (11 of 35) and 26.9% (7 of 26) of colon and rectal cancer cases respectively showed an increase in SHH expression in tumours compared to 72.7% (24 of 33) of the pancreatic cancer cases when compared with their adjacent normal tissues. Our results suggest that SHH may have a strong role in the predisposition of Pancreatic cancer and could possibly be used as a diagnostic or prognostic biomarker. C1 [Niyaz, Madiha] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Soura, Srinagar, India. [Khan, S Mosin] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Soura, Srinagar, India. [Wani, A Rauf] Sher-I-Kashmir Institute of Medical Sciences, Department of General and Minimal Invasive Surgery, 190011 Soura, Srinagar, India. [Shah, J Omar] Sher-I-Kashmir Institute of Medical Sciences, Department of Surgical Gastroenterology, 190011 Soura, Srinagar, India. [Mudassar, Syed] Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Soura, Srinagar, India. RP Mudassar, S (reprint author), Sher-I-Kashmir Institute of Medical Sciences, Department of Clinical Biochemistry, 190011 Soura, India. EM syed.mudassar@skims.ac.in CR Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F, 2017, Global patterns and trends in colorectal cancer incidence and mortality. Gut 66:683–691. , DOI 10.1136/ gutjnl-2015-310912 Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:E359–E386. , DOI 10.1002/ijc.29210 Ilic M, Ilic I, 2016, Epidemiology of pancreatic cancer. World J Gastroenterol 22:9694–9705. , DOI 10.3748/wjg.v22.i44. 9694 Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67:7–30. , DOI 10.3322/caac.21387 Chapnick DA,Warner L, Bernet J, Rao T, Liu X, 2011, Partners in crime: the TGFβ and MAPK pathways in cancer progression. Cell Biosci 1:42. , DOI 10.1186/2045-3701-1-42 Markowitz SD, Bertagnolli MM, 2009, Molecular origins of cancer: molecular basis of colorectal cancer. N Engl J Med 361:2449– 2460. , DOI 10.1056/NEJMra0804588 Segditsas S, Tomlinson I, 2006, Colorectal cancer and genetic alterations in theWnt pathway. Oncogene 25:7531–7537. https://doi. org/10.1038/sj.onc.1210059 Kuzhandaivel A, Schultz SW, Alkhori L, Alenius M, 2014, Ciliamediated hedgehog signaling in Drosophila. Cell Rep 7:672–680. , DOI 10.1016/j.celrep.2014.03.052 Varjosalo M, Taipale J, 2008, Hedgehog: functions and mechanisms. Genes Dev 22:2454–2472. , DOI 10.1101/gad. 1693608 Merchant JL, 2012, Hedgehog signalling in gut development, physiology and cancer. J Physiol 590:421–432. , DOI 10. 1113/jphysiol.2011.220681 Briscoe J, Therond PP, 2013, The mechanisms of hedgehog signalling and its roles in development and disease. Nat Rev Mol Cell Biol 14:416–429. , DOI 10.1038/nrm3598 Shi X, Zhang Z, Zhan X, Cao M, Satoh T, Akira S, Shpargel K, Magnuson T, Li Q, Wang R, Wang C, Ge K, Wu J, 2014, An epigenetic switch induced by Shh signalling regulates gene activation during development and medulloblastoma growth. Nat Commun 5:5425–5456. , DOI 10.1038/ncomms6425 Xie J,MuroneM, Luoh S-M, Ryan A, Gu Q, Zhang C, Bonifas JM, Lam CW, Hynes M, Goddard A, Rosenthal A, Jr EHE, de Sauvage FJ, 1998, Activating smoothened mutations in sporadic basal-cell carcinoma. Nature 391:90–92. , DOI 10.1038/34201 Satheesha S,Manzella G, Bovay A, Casanova EA, Bode PK, Belle R, Feuchtgruber S, Jaaks P, Dogan N, Koscielniak E, Schafer BW, 2016, Targeting hedgehog signaling reduces self-renewal in embryonal rhabdomyosarcoma. Oncogene 35:2020–2030. https://doi. org/10.1038/onc.2015.267 Thayer SP, di Magliano MP, Heiser PW, Nielsen CM, Roberts DJ, Lauwers GY, Qi YP, Gysin S, Castillo CFD, Yajnik V, Antoniu B, McMahon M, Warshaw AL, Hebrok M, 2003, Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 425:851–856. , DOI 10.1038/nature02009 Bailey JM, Mohr AM, Hollingsworth MA, 2009, Sonic hedgehog paracrine signaling regulates metastasis and lymphangiogenesis in pancreatic cancer. Oncogene 28:3513–3525. , DOI 10. 1038/onc.2009.220 Noman AS, UddinM, RahmanMZ, NayeemMJ, AlamSS, Khatun Z, Wahiduzzaman M, Sultana A, Rahman ML, Ali MY, Barua D, Ahmed I, Islam MS, Aboussekhra A, Yeger H, Farhat WA, Islam SS, 2016, Overexpression of sonic hedgehog in the triple negative breast cancer: clinicopathological characteristics of high burden breast cancer patients from Bangladesh. Sci Rep 6:18830. https:// doi.org/10.1038/srep18830 Lee S-Y, Han HS, Lee KY, Hwang TS, Kim JH, Sung IK, Park HS, Jin CJ, Choi KW, 2007, Sonic hedgehog expression in gastric cancer and gastric adenoma. Oncol Rep 17:1051–1055 Karhadkar SS, Steven Bova G, Abdallah N, Dhara S, Gardner D, Maitra A, Isaacs JT, Berman DM, Beachy PA, 2004, Hedgehog signalling in prostate regeneration, neoplasia and metastasis. Nature 431:707–712. , DOI 10.1038/nature02962 Park K-S, Martelotto LG, Peifer M, Sos ML, Karnezis AN, Mahjoub MR, Bernard K, Conklin JF, Szczepny A, Yuan J, Guo R, Ospina B, Falzon J, Bennett S, Brown TJ, Markovic A, Devereux WL, Ocasio CA, Chen JK, Stearns T, Thomas RK, Dorsch M, Buonamici S, Watkins DN, Peacock CD, Sage J, 2011, A crucial requirement for hedgehog signaling in small cell lung cancer. Nat Med 17:1504–1508. , DOI 10.1038/nm. 2473 Guo L-Y, Liu P, Wen Y, Cui W, Zhou Y, 2014, Sonic hedgehog signaling pathway in primary liver cancer cells. Asian Pac J Trop Med 7:735–738. , DOI 10.1016/S1995-7645(14)60126-7 Williamson AJ, Doscas ME, Ye J, Heiden KB, Xing M, Li Y, Prinz RA, XuX, 2016, The sonic hedgehog signaling pathway stimulates anaplastic thyroid cancer cell motility and invasiveness by activating Akt and c-Met. Oncotarget 7:10472–10485. , DOI 10. 18632/oncotarget.7228 Shin K, Lim A, Zhao C, Sahoo D, Pan Y, Spiekerkoetter E, Liao JC, Beachy PA, 2014, Hedgehog signaling restrains bladder cancer progression by eliciting stromal production of urothelial differentiation factors. Cancer Cell 26:521–533. , DOI 10.1016/j. ccell.2014.09.001 Bhattacharya R, Kwon J, Ali B, Wang E, Patra S, Shridhar V, Mukherjee P, 2008, Role of hedgehog signaling in ovarian cancer. Clin Cancer Res 14:7659–7666. , DOI 10.1158/1078- 0432.CCR-08-1414 Varnat F, Duquet A, MalerbaM, ZbindenM,Mas C, Gervaz P, Ruiz i Altaba A, 2009, Human colon cancer epithelial cells harbour active HEDGEHOG-GLI signalling that is essential for tumour growth, recurrence, metastasis and stem cell survival and expansion. EMBO Mol Med 1:338–351. , DOI 10.1002/emmm. 200900039 Pandith AA, Siddiqi MA, 2012, Burden of cancers in the valley of Kashmir: 5 year epidemiological study reveals a different scenario. Tumour Biol 33:1629–1637. , DOI 10.1007/s13277-012- 0418-z Yang L, Xie G, Fan Q, Xie J, 2009, Activation of the hedgehogsignaling pathway in human cancer and the clinical implications. Oncogene 29:469–481. , DOI 10.1038/onc.2009.392 Saqui-Salces M, Merchant JL, 2010, Hedgehog signaling and gastrointestinal cancer. BiochimBiophysActa 1803:786–795. https:// doi.org/10.1016/j.bbamcr.2010.03.008 Mazumdar T, DeVecchio J, Shi T, Jones J, Agyeman A, Houghton JA, 2011, Hedgehog signaling drives cellular survival in human colon carcinoma cells. Cancer Res 71:1092–1102. , DOI 10.1158/0008-5472.CAN-10-2315 Gerling M, Buller NVJA, Kirn LM et al, 2016, Erratum: Stromal Hedgehog signalling is downregulated in colon cancer and its restoration restrains tumour growth. Nat Commun 7:12936. https:// doi.org/10.1038/ncomms12936 Chatel G, Ganeff C, Boussif N, Delacroix L, Briquet A, Nolens G, Winkler R, 2007, Hedgehog signaling pathway is inactive in colorectal cancer cell lines. Int J Cancer 121:2622–2627. , DOI 10.1002/ijc.22998 Douard R, Moutereau S, Pernet P, ChimingqiM, Allory Y, Manivet P, Conti M, Vaubourdolle M, Cugnenc PH, Loric S, 2006, Sonic hedgehog-dependent proliferation in a series of patients with colorectal cancer. Surgery 139:665–670. , DOI 10.1016/j.surg. 2005.10.012 Ohuchida K, Mizumoto K, Fujita H, Yamaguchi H, Konomi H, Nagai E, Yamaguchi K, Tsuneyoshi M, Tanaka M, 2006, Sonic hedgehog is an early developmental marker of intraductal papillary mucinous neoplasms: clinical implications of mRNA levels in pancreatic juice. J Pathol 210:42–48. , DOI 10.1002/path.2019 Marechal R, Bachet J-B, Calomme A et al, 2015, Sonic hedgehog and Gli1 expression predict outcome in resected pancreatic adenocarcinoma. Clin Cancer Res 21:1215–1224. , DOI 10. 1158/1078-0432.CCR-14-0667 Onishi H, KatanoM, 2014, Hedgehog signaling pathway as a new therapeutic target in pancreatic cancer.World J Gastroenterol:WJG 20:2335–2342. , DOI 10.3748/wjg.v20.i9.2335 Ertao Z, Jianhui C, Chuangqi C et al, 2016, Autocrine Sonic hedgehog signaling promotes gastric cancer proliferation through induction of phospholipase Cγ1 and the ERK1/2 pathway. J Exp Clin Cancer Res 35:63. , DOI 10. 1186/s13046-016-0336-9 Ma X, Sheng T, ZhangY, Zhang X, He J, Huang S, Chen K, Sultz J, Adegboyega PA, Zhang H, Xie J, 2006, Hedgehog signaling is activated in subsets of esophageal cancers. Int J Cancer 118:139– 148. , DOI 10.1002/ijc.21295 Mohelnikova-Duchonova B, Kocik M, Duchonova B, Brynychova V, Oliverius M, Hlavsa J, Honsova E, Mazanec J, Kala Z, Ojima I, Hughes DJ, Doherty JE, Murray HA, Crockard MA, Lemstrova R, Soucek P, 2017, Hedgehog pathway overexpression in pancreatic cancer is abrogated by new-generation taxoid SB-T-1216. Pharmacogenomics J 17:452–460. , DOI 10.1038/tpj. 2016.55 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 551 EP 557 DI 10.1007/s12253-018-00564-2 PG 7 ER PT J AU Zhang, K Li, C Liu, J Li, Z Ma, Ch AF Zhang, Ke Li, Cuilin Liu, Jianqiu Li, Zhi Ma, Chao TI Down-Regulation of APTR and it’s Diagnostic Value in Papillary and Anaplastic Thyroid Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE APTR; Papillary thyroid cancer (PTC); Anaplastic thyroid cancer (ATC); ROC; Diagnosis ID APTR; Papillary thyroid cancer (PTC); Anaplastic thyroid cancer (ATC); ROC; Diagnosis AB APTR has been employed as a potential biomarker attributing to it was involved in carcinogenesis and malignancy’s progression. However, the roles of APTR in papillary thyroid cancer (PTC) and anaplastic thyroid cancer (ATC) are unclear. In the present study, we aimed to explore the relative expression of APTR in PTC and ATC tissues and the relation between APTR expression and PTC clinicopathological features. We analyzed APTR expression in PTC and ATC by investigating data obtained from the Gene Expression Omnibus (GEO) database. Then, we tested 76-pair PTC and adjacent normal samples by qRT-PCR, and the result was in accordance with the analysis in GEO datasets. Chi-square (χ2) analysis was employed to evaluate the association between APTR and PTC clinical features. These results showed that APTR was negatively related to TNM stages, distant metastasis. In addition, we further evaluated the feasibility of using APTR to detect PTC and ATC patients by the receiver operating characteristic (ROC) and the area under curve (AUC). These findings implied that down-regulation of APTR is correlated with tumorigenesis, also indicated that the potential diagnostic value of APTR for detecting PTC and ATC patients. C1 [Zhang, Ke] Central South University, Xiangya Hospital, Department of Clinical Pharmacology, 410008 Changsha, China. [Li, Cuilin] ZhuZhou Central Hospital, Department of Pharmacy, 410078 ZhuZhou, China. [Liu, Jianqiu] Central South University, Xiangya Hospital, Department of Clinical Pharmacology, 410008 Changsha, China. [Li, Zhi] Central South University, Xiangya Hospital, Department of Clinical Pharmacology, 410008 Changsha, China. [Ma, Chao] Zhengzhou Central Hospital Affiliated to Zhengzhou University, Department of Hepatopancreatobiliary Surgery, Tongbai Road #195, 450007 Zhengzhou, Henan, China. RP Ma, Ch (reprint author), Zhengzhou Central Hospital Affiliated to Zhengzhou University, Department of Hepatopancreatobiliary Surgery, 450007 Zhengzhou, China. EM machao813@163.com CR Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(1):7–30 Davies L,Welch HG, 2006, Increasing incidence of thyroid cancer in the United States, 1973–2002. 2 JAMA 295(18):2164–2167 Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63:11–30 McHenry CR, Stulberg JJ, 2014, Prophylactic central compartment neck dissection for papillary thyroid cancer. Surg Clin North Am 94(3):529–540 Are C, Ann SAR, 2006, Anaplastic thyroid carcinoma: biology, pathogenesis, prognostic factors, and treatment approaches. Surg Oncol 13(4):453–464 Schlumberger M, Sherman SI, 2012, Approach to the patient with advanced differentiated thyroid cancer. Eur J Endocrinol 166(1):5– 11 Rinn JL, Chang HY, 2012, Genome regulation by long noncoding RNAs. Annu Rev Biochem 81:145–166 Batista PJ, Chang HY, 2013, Long noncoding RNAs: cellular address codes in development and disease. Cell 152(6):1298–1307 Schmitt AM, Chang HY, 2016, Long noncoding RNAs in Cancer pathways. Cancer Cell 29(4):452–463 Hou S, Lin Q, Guan F, Lin C, 2018). LncRNA TNRC6C-AS1 regulates UNC5B in thyroid cancer to influence Cell Prolif, migration, and invasion as a competing endogenous RNA of miR-129- 5p. J Cell Biochem 2018 Jun 12 Zhang H, Cai Y, Zheng L et al., 2018, LncRNA BISPR promotes the progression of thyroid papillary carcinoma by regulating miR- 21-5p. Int J Immunopathol Pharmacol. 2018 Jan-Dec;32: 2058738418772652 Qin Y, SunW, Zhang H, Zhang P,Wang Z, DongW,He L, Zhang T, Shao L, Zhang W, Wu C, 2018, LncRNA GAS8-AS1 inhibits cell proliferation through ATG5-mediated autophagy in papillary thyroid cancer. Endocrine 59(3):555–564 Ma G, Tang M, Wu Y, Xu X, Pan F, Xu R, 2016, LncRNAs and miRNAs: potential biomarkers and therapeutic targets for prostate cancer. Am J Transl Res 8(12):5141–5150 Zhang D, Liu X, Wei B, Qiao G, Jiang T, Chen Z, 2017, Plasma lncRNA GAS8-AS1 AS a potential biomarker of papillary thyroid carcinoma in Chinese patients. Int J Endocrinol 2017(2645904):1– 6 Liao D, Lv G, Wang T, Min J, Wang Y, Liu S, 2018, Prognostic value of long non-coding RNA BLACAT1 in patients with papillary thyroid carcinoma. Cancer Cell Int 18:47 WangXM, Liu Y, Fan YX, Liu Z, Yuan QL, Jia M, Geng ZS, Gu L, Lu XB, 2018, LncRNA PTCSC3 affects drug resistance of anaplastic thyroid cancer through STAT3/INO80 pathway. Cancer Biol Ther 19(7):590–597 Negishi M, Wongpalee SP, Sarkar S et al, 2014, A new lncRNA, APTR, associates with and represses the CDKN1A/p21 promoter by recruiting polycomb proteins. PLoS One 9(4):e95216–e95216 Yu F, Zheng J, Mao Y, Dong P, Li G, Lu Z, Guo C, Liu Z, Fan X, 2015, Long non-coding RNA APTR promotes the activation of hepatic stellate cells and the progression of liver fibrosis. Biochem Biophys Res Commun 463(4):679–685 von Roemeling CA, Marlow LA, Pinkerton AB, Crist A, Miller J, Tun HW, Smallridge RC, Copland JA, 2015, Aberrant lipid metabolism in anaplastic thyroid carcinoma reveals stearoyl CoA desaturase 1 as a novel therapeutic target. J Clin Endocrinol Metab 100:E697–E709 Lan X, Zhang H, Wang Z, Dong W, Sun W, Shao L, Zhang T, Zhang D, 2015, Genome-wide analysis of long noncoding RNA expression profile in papillary thyroid carcinoma. Gene 569(1): 109–117 Tomas G, Tarabichi M, Gacquer D, Hebrant A, Dom G, Dumont JE, Keutgen X, Fahey TJ,Maenhaut C, Detours V, 2012, A general method to derive robust organ-specific gene expression-based differentiation indices: application to thyroid cancer diagnostic. Oncogene 31(41):4490–4498 Li C, Zou H,Wang Z, Tang X, Fan X, Zhang K, Liu J, Li Z, 2018, REST, not REST4, is a risk factor associated with radiotherapy plus chemotherapy efficacy in glioma. Drug Des Devel Ther 12:1363– 1371 Schmittgen TD, Livak KJ, 2008, Analyzing real-time PCR data by the comparative C(T, method. Nat Protoc 3:1101–1108 Yu S, Qi Y, Jiang J,Wang H, Zhou Q, 2018, APTR is a prognostic marker in cirrhotic patients with portal hypertension during TIPS procedure. Gene 645:30–33 Abbas T, Dutta A, 2009, p21 in cancer: intricate networks and multiple activities. Nat Rev Cancer 9:400–414 LaBaer J, Garrett MD, Stevenson LF, Slingerland JM, Sandhu C, Chou HS, Fattaey A, Harlow E, 1997, New functional activities for the p21 family of CDK inhibitors. Genes Dev 11:847–862 Melck A, Masoudi H, Griffith OL, Rajput A, Wilkins G, Bugis S, Jones SJM, Wiseman SM, 2007, Cell cycle regulators show diagnostic and prognostic utility for differentiated thyroid cancer. Ann Surg Oncol 14:3403–3411 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 559 EP 565 DI 10.1007/s12253-018-0561-y PG 7 ER PT J AU Nterma, P Panopoulou, E Papadaki-Petrou, E Assimakopoulou, M AF Nterma, Pinelopi Panopoulou, Eleni Papadaki-Petrou, Eleni Assimakopoulou, Martha TI Immunohistochemical Profile of Tumor Suppressor Proteins RASSF1A and LATS1/2 in Relation to p73 and YAP Expression, of Human Inflammatory Bowel Disease and Normal Intestine SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE RASSF1A; LATS1/2; HIPPO pathway; Human inflammatory bowel disease; Ulcerative colitis; Crohn’s disease ID RASSF1A; LATS1/2; HIPPO pathway; Human inflammatory bowel disease; Ulcerative colitis; Crohn’s disease AB The intestinal neoplastic transformation is a possible risk of chronic inflammatory bowel disease (IBD). Previous evidence in mice IBD provides a role for the RAS-association domain family tumor suppressor protein 1 A (RASSF1A), in the repairing process following mucosa epithelium damage, through cooperation with the HIPPO-signaling molecules p73, and YAP. HIPPO pathway which has been implicated in stem cell activity includes as key components for signal transduction the large tumor suppressor homology Ser/Thr kinases LATS1/2. The aim of this study was to assess immunohistochemically, using specific antibodies, the RASSF1A and LATS1/2 expression patterns in a cohort of patients with IBD including 52 ulcerative colitis (UC), 24 Crohn’s disease (CD) and 24 IBD unclassified (IBD-U), compared with normal intestine from non-IBD patients (control group). The relationship between subtypes of IBD and RASSF1A and LATS1/2 expression, both individually and related to p73 and YAP/ pYAP(Ser127) proteins was also investigated. Quantitative analyses of the immunohistochemical findings in mucosa cells revealed a significantly decreased expression in UC and IBD-U for RASSF1A expression and a significantly elevated expression in UC, IBD-U, and CD for LATS1/2 expression compared with normal mucosa (P < 0.05).However, ROC curve analysis showed that only LATS1/2 could differentiate IBD from control group. RASSF1A expression was significantly correlated with LATS1/2 in UC with dysplasia (P < 0.0001), and p73 in UC (P < 0.001), and IBD-U(P < 0.02). The expression of all proteins did not differ significantly between subtypes of IBD (P ≥ 0.05). RASSF1A-LATS1/2 co-expression was mainly observed in IBD samples. These findings suggest that tumor suppression proteins RASSF1A and LATS1/2 may be involved in the pathogenesis of human IBD and imply a potential cooperation of RASSF1A, and HIPPO signaling pathways in human bowel inflammation. C1 [Nterma, Pinelopi] University of Patras, School of Medicine, Department of Anatomy, Histology and Embryology, Biomedical Sciences Research Building, 1 Asklipiou, Gr-26504 Rion, Greece. [Panopoulou, Eleni] General Hospital Agios Andreas, Department of PathologyPatras, Greece. [Papadaki-Petrou, Eleni] University of Patras, School of Medicine, Department of Anatomy, Histology and Embryology, Biomedical Sciences Research Building, 1 Asklipiou, Gr-26504 Rion, Greece. [Assimakopoulou, Martha] University of Patras, School of Medicine, Department of Anatomy, Histology and Embryology, Biomedical Sciences Research Building, 1 Asklipiou, Gr-26504 Rion, Greece. RP Assimakopoulou, M (reprint author), University of Patras, School of Medicine, Department of Anatomy, Histology and Embryology, Gr-26504 Rion, Greece. EM massim@upatras.gr CR Maher MM, 2012, Inflammatory bowel disease: review and future view. Front Biosci, Elite Ed, 4:1638–1647 Conrad K, Roggenbuck D, Laass MW, 2014, Diagnosis and classification of ulcerative colitis. Autoimmun Rev 13(4–5):463–466 Laass MW, Roggenbuck D, Conrad K, 2014, Diagnosis and classification of Crohn's disease. Autoimmun Rev 13(4–5):467–471 Feakins RM, 2014, Ulcerative colitis or Crohn's disease? Pitfalls and problems. Histopathology 64(3):317–335 Martland GT, Shepherd NA, 2007, Indeterminate colitis: definition, diagnosis, implications and a plea for nosological sanity. Histopathology 50(1):83–96 Okayasu I, 2012, Development of ulcerative colitis and its associated colorectal neoplasia as a model of the organ-specific chronic inflammation-carcinoma sequence. Pathol Int 62:368–380 Rogler G, 2014, Chronic ulcerative colitis and colorectal cancer. Cancer Lett 345:235–241 Donninger H, Vos MD, Clark GJ, 2007, The RASSF1A tumor suppressor. J Cell Sci 120:3163–3172 Overmeyer JH, Maltese WA, 2011, Death pathways triggered by activated Ras in cancer cells. Front Biosci, Landmark Ed, 16:1693– 1713 Song MS, Song SJ, Kim SY, Oh HJ, Lim DS, 2008, The tumour suppressor RASSF1A promotes MDM2 self-ubiquitination by disrupting the MDM2–DAXX–HAUSP complex. EMBO J 27: 1863–1874 Agathanggelou A, Cooper WN, Latif F, 2005, Role of the Rasassociation domain family 1 tumor suppressor gene in human cancers. Cancer Res 65:3497–3508 Abouzeid HE, Kassem AM, Abdel Wahab AH, El-mezayen HA, Sharad H, Abdel Rahman S, 2011, Promoter hypermethylation of RASSF1A, MGMT, and HIC-1 genes in benign and malignant colorectal tumors. Tumour Biol 32:845–852 Sakamoto N, Terai T, Ajioka Y, Abe S, Kobayasi O, Hirai S, Hino O,Watanabe H, Sato N, Shimoda T, Fujii H, 2004, Frequent hypermethylation of RASSF1A in early flat-type colorectal tumors. Oncogene 23(55):8900–8907 Cao D, Chen Y, Tang Y, Peng XC, Dong H, Li LH, Cheng K, Ge J, Liu JY, 2013, Loss of RASSF1A expression in colorectal cancer and its association with K-ras status. Biomed Res Int 2013:976765 Gordon M, El-Kalla M, Zhao Y, Fiteih Y, Law J, Volodko N, Anwar-Mohamed A, El-Kadi AO, Liu L, Odenbach J, Thiesen A, Onyskiw C, Ghazaleh HA, Park J, Lee SB, Yu VC, Fernandez- Patron C, Alexander RT, Wine E, Baksh S, 2013, The tumor suppressor gene, RASSF1A, is essential for protection against inflammation-induced injury. PLoS One 8(10):e75483 Hiemer SE, Varelas X, 2013, Stem cell regulation by the Hippo pathway. Biochim Biophys Acta 1830(2):2323–2334 Ramos A, Camargo FD, 2012, The Hippo signaling pathway and stem cell biology. Trends Cell Biol 22(7):339–346 Hong W, Guan KL, 2012, The YAP and TAZ transcription coactivators: key downstream effectors of the mammalian Hippo pathway. Semin Cell Dev Biol 23(7):785–793 Levy D, Adamovich Y, Reuven N, Shaul Y, 2008, Yap1 phosphorylation by c-Abl is a critical step in selective activation of proapoptotic genes in response to DNA damage. Mol Cell 29(3): 350–361 Matallanas D, Romano D, Yee K, Meissl K, Kucerova L, Piazzolla D, Baccarini M, Vass JK, Kolch W, O'neill E, 2007, RASSF1A elicits apoptosis through an MST2 pathway directing proapoptotic transcription by the p73 tumor suppressor protein. Mol Cell 27(6):962–975 Li VS, Clevers H, 2013, Intestinal regeneration: YAP-tumor suppressor and oncoprotein? Curr Biol 23(3):R110–R112 Cai J, Zhang N, Zheng Y, deWilde RF, Maitra A, Pan D, 2010, The Hippo signaling pathway restricts the oncogenic potential of an intestinal regeneration program. Genes Dev 24(21):2383–2388 Barry ER, Morikawa T, Butler BL, Shrestha K, de la Rosa R, Yan KS, Fuchs CS, Magness ST, Smits R, Ogino S, Kuo CJ, Camargo FD, 2013, Restriction of intestinal stem cell expansion and the regenerative response by YAP. Nature 493(7430):106–110 Avruch J, Zhou D, Bardeesy N, 2012, YAP oncogene overexpression supercharges colon cancer proliferation. Cell Cycle 11(6):1090–1096 Wang L, Shi S, Guo Z, Zhang X, Han S, Yang A, Wen W, Zhu Q, 2013, Overexpression of YAP and TAZ is an independent predictor of prognosis in colorectal cancer and related to the proliferation and metastasis of colon cancer cells. PLoS One 8(6):e65539 Wang Y, Dong Q, Zhang Q, Li Z, Wang E, Qiu X, 2010, Overexpression of yes-associated protein contributes to progression and poor prognosis of non-small-cell lung cancer. Cancer Sci 101:1279–1285 Ji T, Liu D, Shao W, Yang W, Wu H, Bian X, 2012, Decreased expression of LATS1 is correlated with the progression and prognosis of glioma. J Exp Clin Cancer Res 31:67 Puig P, Capodieci P, Drobnjak M, Verbel D, Prives C, Cordon- Cardo C, Di Como CJ, 2003, p73 expression in human normal and tumor tissues: loss of p73 expression is associated with tumor progression in bladder cancer. Clin Cancer Res 9:5642–5651 Guan M, Peng HX, Yu B, Lu Y, 2003, p73 Overexpression and angiogenesis in human colorectal carcinoma. Jpn J Clin Oncol 33(5):215–220 Fernandes MS, Carneiro F, Oliveira C, Seruca R, 2013, Colorectal cancer and RASSF family–a special emphasis on RASSF1A. Int J Cancer 132(2):251–258 Wagner KJ, Cooper WN, Grundy RG, Caldwell G, Jones C, Wadey RB, Morton D, Schofield PN, Reik W, Latif F, Maher ER, 2002, Frequent RASSF1A tumour suppressor gene promoter methylation inWilms' tumour and colorectal cancer. Oncogene 21(47):7277–7282 Visser S, Yang X, 2010, LATS tumor suppressor: a new governor of cellular homeostasis. Cell Cycle 9(19):3892–3903 Wierzbicki PM, Adrych K, Kartanowicz D, Stanislawowski M, Kowalczyk A, Godlewski J, Skwierz-Bogdanska I, Celinski K, Gach T, Kulig J, Korybalski B, Kmiec Z, 2013, Underexpression of LATS1 TSG in colorectal cancer is associated with promoter hypermethylation. World J Gastroenterol 19(27):4363–4373 Xia H, Qi H, LiY, Pei J, Barton J, Blackstad M, Xu T, TaoW(2002, LATS1 tumor suppressor regulates G2/M transition and apoptosis. Oncogene 21(8):1233–1241 Villanacci V, Antonelli E, Geboes K, Casella G, Bassotti G, 2013, Histological healing in inflammatory bowel disease: a still unfulfilled promise. World J Gastroenterol 19(7):968–978 Fausti F, Di Agostino S, Sacconi A, Strano S, Blandino G, 2012, Hippo and rassf1a pathways: a growing affair. Mol Biol Int 2012:307628 Arijs I, Li K, Toedter G, Quintens R, Van Lommel L, Van Steen K, Leemans P, De Hertogh G, Lemaire K, Ferrante M, Schnitzler F, Thorrez L, Ma K, Song XY,Marano C, Van Assche G, Vermeire S, Geboes K, Schuit F, Baribaud F, Rutgeerts P, 2009, Mucosal gene signatures to predict response to infliximab in patients with ulcerative colitis. Gut 58(12):1612–1619 Arijs I, Quintens R, Van Lommel L, Van Steen K, De Hertogh G, Lemaire K, Schraenen A, Perrier C, Van Assche G, Vermeire S, Geboes K, Schuit F, Rutgeerts P, 2010, Predictive value of epithelial gene expression profiles for response to infliximab in Crohn's disease. Inflamm Bowel Dis 16(12):2090–2098 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 567 EP 574 DI 10.1007/s12253-018-00575-z PG 8 ER PT J AU He, D Liu, L Wang, Y Sheng, M AF He, Dongyun Liu, Li Wang, Yang Sheng, Minjia TI A Novel Genes Signature Associated with the Progression of Polycystic Ovary Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Polycystic ovary syndrome; Microarray; WGCNA; Interaction network ID Polycystic ovary syndrome; Microarray; WGCNA; Interaction network AB To identify genes involving in the pathogenesis of polycystic ovary syndrome (PCOS). In this study, the comprehensive analysis of GSE8157 was downloaded. Overlapping genes of differentially expressed genes (DEGs) were identified, and enrichment analysis for these genes was performed. A modular network of differentially expressed genes was constructed by weighted gene co-expression network analyses (WGCNA), and a total of 322 differentially expressed genes in 5 stable modules were screened. The correlations of genes of the stable modules in BioGRID 3.4, STRING 10.5, HPRD9 databases were screened, and the interaction network of 104 DEGs was constructed. In addition, some genes and the key words were searched in CTD. A total of 596 differentially expressed genes were screened, including 379 genes that were up-regulated in case group and down-regulated in control group and treat group, and 217 genes that were down-regulated in case group and up-regulated in control group and treat group. The differentially expressed genes were enriched in PPAR signaling pathway, Neuroactive ligand-receptor interaction, cAMP signaling pathway, of which pathways were involved in the cancer development. Finally, 7 important target genes were identified, such as APOC3 was interacted with pioglitazone, ADCY2 involved in cAMP signaling pathway, and the genes (C3AR1, HRH2, GRIA1, MLNR and TAAR2) involved in neuroactive ligand-receptor interaction. In addition, the important target genes were significantly differential expression. These results implied that the 7 important target genes were played an important role in the development and progression of PCOS. Our study implied that genes had played a key role in the development and progression of PCOS, the results showed that microarray can be use as a method for the discovery of new biomarkers and therapeutic targets for PCOS. C1 [He, Dongyun] China-Japan Union Hospital of Jilin University, Department of Gynecology and Obstetrics, No.126, Xiantai Road, 130031 Changchun, China. [Liu, Li] China-Japan Union Hospital of Jilin University, Department of Gynecology and Obstetrics, No.126, Xiantai Road, 130031 Changchun, China. [Wang, Yang] The Affiliated Hospital of Changchun University of Chinese Medicine, Department of Dermatology, 130031 Changchun, China. [Sheng, Minjia] China-Japan Union Hospital of Jilin University, Department of Gynecology and Obstetrics, No.126, Xiantai Road, 130031 Changchun, China. RP Sheng, M (reprint author), China-Japan Union Hospital of Jilin University, Department of Gynecology and Obstetrics, 130031 Changchun, China. EM angela83666@163.com CR Shahebrahimi K, Jalilian N, Bazgir N, Rezaei M, 2016, Comparison clinical and metabolic effects of metformin and pioglitazone in polycystic ovary syndrome. Indian J EndocrinolMetab 20(6):805–809 Guido M, Romualdi D, Suriano R, Giuliani M, Costantini B, Apa R, Lanzone A, 2004, Effect of pioglitazone treatment on the adrenal androgen response to corticotrophin in obese patients with polycystic ovary syndrome. Hum Reprod 19(3):534–539 Schroder AK, Tauchert S, Ortmann O, Diedrich K, Weiss JM, 2004, Insulin resistance in patients with polycystic ovary syndrome. Ann Med 36(6):426–439 De LV, La MA, Petraglia F, 2003, Insulin-lowering agents in the management of polycystic ovary syndrome. Endocr Rev 24(5): 633–667 Skov V,Glintborg D, Knudsen S, Tan Q, Jensen T, Kruse TA, Beck- Nielsen H, Hojlund K, 2008, Pioglitazone enhances mitochondrial biogenesis and ribosomal protein biosynthesis in skeletal muscle in polycystic ovary syndrome. PLoS One 3(6):e2466 Tang T, Norman RJ, Balen AH, Lord JM, 2003, Insulin-sensitising drugs, metformin, troglitazone, rosiglitazone, pioglitazone, Dchiro- inositol, for polycystic ovary syndrome. Cochrane Database Syst Rev 3(3):CD003053 Glueck CJ, Moreira A, Goldenberg N, Sieve L, Wang P, 2003, Pioglitazone and metformin in obese women with polycystic ovary syndrome not optimally responsive to metformin. Hum Reprod 18(8):1618–1625 Barrett T, Troup DB,Wilhite SE, Ledoux P, Evangelista C, Kim IF, TomashevskyM, Marshall KA, Phillippy KH, Sherman PM, 2011, NCBI GEO: archive for functional genomics data sets—10 years on. Nucleic Acids Res 39(Database issue):D1005 Li Y, Ngom A, Rueda L, 2010, Missing value imputation methods for gene-sample-time microarray data analysis. IEEE Symposium on Computational Intelligence in Bioinformatics & Computational Biology:1–7 Smyth GK, 2011, Limma: linear models for microarray data. In: Bioinformatics & Computational Biology Solutions Using R & Bioconductor, pp 397–420. , DOI 10.1007/0-387-29362- 0_23 Sherlock G, 2000, Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 25(1):25–29 Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K, 2017, KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res 45(Database issue): D353–D361 Dennis G, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA, 2003, DAVID: database for annotation, visualization, and integrated discovery. Genome Biol 4(5):P3 Langfelder P, Horvath S, 2008)WGCNA: an R package for weighted correlation network analysis. Bmc Bioinformatics 9(1):559 Medina IR, Lubovacpilav Z, 2016, Gene co-expression network analysis for identifying modules and functionally enriched pathways in type 1 diabetes. PLoS One 11(6):e0156006 BJ B, C S TR, L B AB, M L RO, DH L, J B VW, 2008, The BioGRID interaction database: 2008 update. Nucleic Acids Res 36(Database issue):637–640 Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, Santos A, Doncheva NT, Roth A, Bork P, 2017, The STRING database in 2017: quality-controlled protein–protein association networks, made broadly accessible. Nucleic Acids Res 45(Database issue):D362–D368 Mohmood R, Ramachandra YI, KrishnaV, Rahiman BA, Mohan S, Ranganathan P, Ramabadran S, Kashyap MK, 2009, Human protein reference database - 2009 update Davis AP, Wiegers TC, Wiegers J, Johnson RJ, Sciaky D, Grondin CJ, Mattingly CJ, 2018, Chemical-induced phenotypes at CTD help inform the pre-disease state and construct adverse outcome pathways. Toxicological Sciences An Official Journal of the Society of Toxicology Jong MC, Hofker MH, Havekes LM, 1999, Role of ApoCs in lipoprotein metabolism: functional differences between ApoC1, ApoC2, and ApoC3. Arterioscler Thromb Vasc Biol 19(3):472 Jong MC, Hofker MH, Havekes LM, 1999, Role of ApoCs in lipoprotein metabolism. Arterioscler Thromb Vasc Biol 19(3): 472–484 Jackson CL, Breslow JL, 1984, Isolation and sequence of a human apolipoprotein CII cDNA clone and its use to isolate and map to human chromosome 19 the gene for apolipoprotein CII. Proc Natl Acad Sci U S A 81(10):2945–2949 Shachter NS, Hayek T, Leff T, Smith JD, RosenbergDW,Walsh A, Ramakrishnan R, Goldberg IJ, Ginsberg HN, Breslow JL, 1994, Overexpression of apolipoprotein CII causes hypertriglyceridemia in transgenic mice. J Clin Investig 93(4):1683–1690 Braun-Falco O, Plewig G, Wolff HH, Burgdorf WHC, 2000, Disorders of lipoprotein and lipid metabolism Silva HVD, Lauer SJ, Wang J, Simonet WS, Weisgraber KH, Mahley RW, Taylor JM, 1994, Overexpression of human apolipoprotein C-III in transgenic mice results in an accumulation of apolipoprotein B48 remnants that is corrected by excess apolipoprotein E. J Biol Chem 269(3):2324 Shulman RS, Herbert PN, Wehrly K, Fredrickson DS, 1975, Thf complete amino acid sequence of C-I, apoLp-Ser), an apolipoprotein fromhuman very lowdensity lipoproteins. J Biol Chem 250(1): 182–190 JongMC, Gijbels MJ, Dahlmans VE, Gorp PJ, Koopman SJ, Ponec M, ., Hofker MH, Havekes LM(1998, Hyperlipidemia and cutaneous abnormalities in transgenic mice overexpressing human apolipoprotein C1. J Clin Investig 101, 1):145 Jong MC, Dahlmans VE, Gorp PJ, Van DKW, Van BML, Hofker MH, Havekes LM, 1996, In the absence of the low density lipoprotein receptor, human apolipoprotein C1 overexpression in transgenic mice inhibits the hepatic uptake of very low density lipoproteins via a receptor-associated protein-sensitive pathway. J Clin Investig 98(10):2259–2267 Shachter NS, Ebara TR, Steiner G, Breslow JL, Ginsberg HN, Smith JD, 1996, Combined hyperlipidemia in transgenic mice overexpressing human apolipoprotein Cl. J Clin Investig 98(3):846 Samanci C,Alis D,Ustabasioglu FE, Ozmen E, Ucar AK, Aslan M, Habibi HA, Bakan S, Ozcabi B, Evliyaoglu SO, 2016, Apparent diffusion coefficient measurement of ovarian stroma: a potential tool for the diagnosis of polycystic ovary syndrome. Diagn Interv Imaging 98(1):57–61 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 575 EP 582 DI 10.1007/s12253-019-00676-3 PG 8 ER PT J AU Jo, SY Kim, SM Yoo, JN Lee, HS AF Jo, Sol Yun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Inactivating Frameshift Mutations of HACD4 and TCP10L Tumor Suppressor Genes in Colorectal and Gastric Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Ikeda M, Kanao Y, Yamanaka M, Sakuraba H, Mizutani Y, Igarashi Y, Kihara A, 2008, Characterization of four mammalian 3- hydroxyacyl-CoA dehydratases involved in very long-chain fatty acid synthesis. FEBS Lett 582:2435–2440 Zhu S, Wang Z, Zhang Z, Wang J, Li Y, Yao L, Mei Q, Zhang W, 2014, PTPLAD2 is a tumor suppressor in esophageal squamous cell carcinogenesis. FEBS Lett 588:981–989 Nord H, Hartmann C, Andersson R, Menzel U, Pfeifer S, Piotrowski A, Bogdan A, Kloc W, Sandgren J, Olofsson T, Hesselager G, Blomquist E, Komorowski J, von Deimling A, Bruder CE, Dumanski JP, Diaz de Stahl T, 2009, Characterization of novel and complex genomic aberrations in glioblastoma using a 32K BAC array. Neuro-Oncology 11:803–818 Zuo J, Cai H,Wu Y, Ma H, JiangW, Liu C, Han D, Ji G, Yu L, 2014, TCP10L acts as a tumor suppressor by inhibiting cell proliferation in hepatocellular carcinoma. Biochem Biophys Res Commun 446:61– 67 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Choi YJ, Kim MS, An CH, Yoo NJ, Lee SH, 2014, Regional bias of intratumoral genetic heterogeneity of nucleotide repeats in colon cancers with microsatellite instability. Pathol Oncol Res 20:965–971 Jo YS, Choi MR, Song SY, Kim MS, Yoo NJ, Lee SH, 2016, Frameshift mutations of HSPA4 and MED13 in gastric and colorectal cancers. Pathol Oncol Res 22:769–772 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 583 EP 584 DI 10.1007/s12253-018-0409-5 PG 2 ER PT J AU Jo, SY Kim, SM Yoo, JN Lee, HS AF Jo, Sol Yun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Intratumoral Heterogeneity for Inactivating Frameshift Mutation of CYB5R2 Gene in Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Jo, Sol Yun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Devaney JM, Wang S, Funda S, Long J, Taghipour DJ, Furbert- Harris P, Ittmann M, Kwabi-Addo B, 2013, Identification of novel DNA-methylated genes that correlate with human prostate cancer and high-grade prostatic intraepithelial neoplasia. Molecular cloning and characterization of p56dok-2 defines a new family of RasGAPbinding proteins. Prostate Cancer Prostatic Dis 16:292–300 X X, Zhao W, Tian F, Zhou X, Zhang J, Huang T, Hou B, Du C, Wang S, Mo Y, Yu N, Zhou S, You J, Zhang Z, Huang G, Zeng X, 2014, Cytochrome b5 reductase 2 is a novel candidate tumor suppressor gene frequently inactivated by promoter hypermethylation in human nasopharyngeal carcinoma. Tumour Biol 35:3755–3763 Choi YJ, Kim MS, An CH, Yoo NJ, Lee SH, 2014, Regional bias of intratumoral genetic heterogeneity of nucleotide repeats in colon cancers with microsatellite instability. Pathol Oncol Res 20:965–971 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Oh HR, An CH, Yoo NJ, Lee SH, 2015, Frameshift mutations of MUC15 gene in gastric and its regional heterogeneity in gastric and colorectal cancers. Pathol Oncol Res 21:713–718 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 585 EP 586 DI 10.1007/s12253-018-0406-8 PG 2 ER PT J AU Lee, HJ An, HCh Kim, SM Yoo, JN Lee, HS AF Lee, Hwa Ju An, Hyeok Chang Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Intratumoral Heterogeneity of RPL22 Frameshift Mutation in Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Lee, Hwa Ju] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [An, Hyeok Chang] The Catholic University of Korea, College of Medicine, Department of General SurgerySeoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Rao S, Lee SY, Gutierrez A, Perrigoue J, Thapa RJ, Tu Z, Jeffers JR, Rhodes M, Anderson S, Oravecz T, Hunger SP, Timakhov RA, Zhang R, Balachandran S, Zambetti GP, Testa JR, Look AT, Wiest DL, 2012, Inactivation of ribosomal protein L22 promotes transformation by induction of the stemness factor, Lin28B. Blood 120: 3764–3773 Ferreira AM, Tuominen I, van Dijk-Bos K, Sanjabi B, van der Sluis T, van der Zee AG, Hollema H, Zazula M, Sijmons RH, Aaltonen LA, Westers H, Hofstra RM, 2014, High frequency of RPL22 mutations in microsatellite-unstable colorectal and endometrial tumors. Hum Mutat 35:1442–1445 Nagarajan N, Bertrand D, Hillmer AM, Zang ZJ, Yao F, Jacques PE, Teo AS, Cutcutache I, Zhang Z, LeeWH, Sia YY, Gao S, Ariyaratne PN, Ho A,Woo XY, Veeravali L, Ong CK, Deng N, Desai KV, Khor CC, Hibberd ML, Shahab A, Rao J,Wu M, Teh M, Zhu F, Chin SY, Pang B, So JB, Bourque G, Soong R, SungWK, Tean Teh B, Rozen S, Ruan X, Yeoh KG, Tan PB, Ruan Y, 2012, Whole-genome reconstruction and mutational signatures in gastric cancer. Genome Biol 13:R115 Marusyk A, Almendro V, Polyak K, 2012, Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 12:323–334 Choi EJ, Kim MS, Song SY, Yoo NJ, Lee SH, 2017, Intratumoral Heterogeneity of Frameshift Mutations in MECOM Gene is Frequent in Colorectal Cancers with High Microsatellite Instability. Pathol Oncol Res 23:145–149 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 587 EP 588 DI 10.1007/s12253-018-0438-0 PG 2 ER PT J AU Zhang, Pb Cui, J AF Zhang, Peng-bo Cui, Jing TI Letter to the Editor Regarding the Article "Long Non-Coding RNA SPRY4-IT1 Can Predict Poor Prognosis in Digestive System Malignancies: a Meta-Analysis" SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Zhang, Peng-bo] Kunming University of Science and Technology, Medical SchoolKunming, Chenggong, China. [Cui, Jing] Kunming General Hospital, Department of PathologyKunming, Chenggong, China. RP Zhang, Pb (reprint author), Kunming University of Science and Technology, Medical School, Kunming, China. EM 1518918082@qq.com CR Sun C, Ding Y, Wang S, Hu W, 2017, Long Non-Coding RNA SPRY4-IT1 Can Predict Poor Prognosis in Digestive System Malignancies: aMeta-Analysis. Pathology & Oncology Research:1–7 Egger M, Smith GD, Phillips AN, 1997, Meta-analysis: principles and procedures. Bmj British Medical Journal 315(7121):1533–1537 Lin KK, Sewell JL, 2013, The effects of race and socioeconomic status on immunomodulator and anti-tumor necrosis factor use among ambulatory patients with inflammatory bowel disease in the United States. Am J Gastroenterol 108(12):1824 Peng W, Wu G, Fan H, Wu J, Feng J, 2015, Long noncoding RNA SPRY4-IT1 predicts poor patient prognosis and promotes tumorigenesis in gastric cancer. Tumor Biol 36(9):6751–6758 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 589 EP 590 DI 10.1007/s12253-018-0432-6 PG 2 ER PT J AU Plander, M Szendrei, T Vadvari, Ivanyi, J AF Plander, Mark Szendrei, Tamas Vadvari, Arpad Ivanyi, Janos TI Standard Dose of Ibrutinib is Effective in the Treatment of Bing-Neel Syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Plander, Mark] Vas County Markusovszky Hospital, Department of Hematology, Markusovszky str 5, 9700 Szombathely, Hungary. [Szendrei, Tamas] Vas County Markusovszky Hospital, Department of Hematology, Markusovszky str 5, 9700 Szombathely, Hungary. [Vadvari, Arpad] Markusovszky University Teaching Hospital, Department of RadiologySzombathely, Hungary. [Ivanyi, Janos] Vas County Markusovszky Hospital, Department of Hematology, Markusovszky str 5, 9700 Szombathely, Hungary. RP Plander, M (reprint author), Vas County Markusovszky Hospital, Department of Hematology, 9700 Szombathely, Hungary. EM planderm@yahoo.com CR Simon L, Fitsiori A, Lemal R, Dupuis J, Carpentier B, Boudin L et al, 2015, Bing-Neel syndrome, a rare complication of Waldenstrom’s macroglobulinemia: analysis of 44 cases and review of the literature. A study on behalf of the French innovative leukemia organization, FILO). Haematologica 100:1587–1594 Castillo JJ, D’Sa S, Lunn MP, Minnema MC, Tedeschi A, Lansigan F et al, 2016, Central nervous system involvement by Waldenstrom macroglobulinaemia, Bing-Neel syndrome): a multi-institutional retrospective study. Br J Haematol 172:709–715 Treon SP, Tripsas CK, Meid K, Warren D, Varma G, Green R et al, 2015, Ibrutinib in previously treated patients with Waldenstrom’s Macroglobulinemia. N Engl J Med 372:1430–1440 Bernard S, Goldwirt L, Amorim S, Brice P, Briere J, De Kerviler E et al, 2015, Activity of ibrutinib in mantle cell lymphoma patients with central nervous system relapse. Blood 126:1695–1698 Mason C, Savona S, Rini JN, Castillo JJ, Xu L, Hunter ZR et al, 2017, Ibrutinib penetrates the blood brain barrier and shows efficacy in the therapy of Bing Neel syndrome. Br J Haematol 179(2):339– 341 Cabannes-Hamy A, Lemal R, Goldwirt L, Poulain S, Amorin S, Perignon R et al, 2016, Efficacy of ibrutinib in the treatment of Bing-Neel syndrome. Am J Hematol 91:E17–E19 Boudin L, Patient M, Romeo E, Blade JS, de Jaureguiberry JP, 2018, Efficacy of ibrutinib as first-line treatment of tumoral Bing-Neel syndrome. Leuk Lymphoma 23:1–3 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 591 EP 592 DI 10.1007/s12253-018-0482-9 PG 2 ER PT J AU Stein, KM Morris, KL Martin, GM AF Stein, K Matthew Morris, K Lindsay Martin, G Mike TI Next-Generation Sequencing Identifies Novel RTK VUSs in Breast Cancer with an Emphasis on ROS1, ERBB4, ALK and NTRK3 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Stein, K Matthew] West Cancer CenterMemphis, TN, USA. [Morris, K Lindsay] University of Tennessee, Health Science Center, College of MedicineMemphis, TN, USA. [Martin, G Mike] West Cancer CenterMemphis, TN, USA. RP Stein, KM (reprint author), West Cancer Center, Memphis, USA. EM mkstein@westclinic.com CR Blau CA, Ramirez AB, Blau S, Pritchard CC, Dorschner MO, Schmechel SC, Martins TJ, Mahen EM, Burton KA, Komashko VM, Radenbaugh AJ, Dougherty K, Thomas A, Miller CP, Annis J, Fromm JR, Song C, Chang E, Howard K, Austin S, Schmidt RA, Linenberger ML, Becker PS, Senecal FM, Mecham BH, Lee SI, Madan A, Ronen R, Dutkowski J, Heimfeld S, Wood BL, Stilwell JL, Kaldjian EP, Haussler D, Zhu J, 2016, A distributed network for intensive longitudinal monitoring in metastatic triple-negative breast Cancer. J Natl Compr Cancer Netw 14(1):8–17 Kurppa KJ, Denessiouk K, Johnson MS, Elenius K, 2016, Activating ERBB4 mutations in non-small cell lung cancer. Oncogene 35(10):1283–1291 Holla VR, Elamin YY, Bailey AM, Johnson AM, Litzenburger BC, Khotskaya YB, Sanchez NS, Zeng J, Shufean MA, Shaw KR, Mendelsohn J, Mills GB, Meric-Bernstam F, Simon GR, 2017, ALK: a tyrosine kinase target for cancer therapy. Cold Spring Harb Mol Case Stud 3(1):a001115 Stephens P, Edkins S, Davies H, Greenman C, Cox C, Hunter C, Bignell G, Teague J, Smith R, Stevens C, O'Meara S, Parker A, Tarpey P, Avis T, Barthorpe A, Brackenbury L, Buck G, Butler A, Clements J, Cole J, Dicks E, Edwards K, Forbes S, Gorton M, Gray K, Halliday K, Harrison R, Hills K, Hinton J, Jones D, Kosmidou V, Laman R, Lugg R, Menzies A, Perry J, Petty R, Raine K, Shepherd R, Small A, Solomon H, Stephens Y, Tofts C, Varian J, Webb A, West S, Widaa S, Yates A, Brasseur F, Cooper CS, Flanagan AM, Green A, Knowles M, Leung SY, Looijenga LHJ, Malkowicz B, Pierotti MA, Teh B, Yuen ST, Nicholson AG, Lakhani S, Easton DF,Weber BL, StrattonMR, Futreal PA,Wooster R, 2005, A screen of the complete protein kinase gene family identifies diverse patterns of somatic mutations in human breast cancer. Nat Genet 37:590–592 Wood LD, Calhoun ES, Silliman N, Ptak J, Szabo S, Powell SM, Riggins GJ, Wang TL, Yan H, Gazdar A, Kern SE, Pennacchio L, Kinzler KW, Vogelstein B, Velculescu VE, 2006, Somatic mutations of GUCY2F, EPHA3, and NTRK3 in human cancers. Hum Mutat 27:1060–1061 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 593 EP 595 DI 10.1007/s12253-018-0550-1 PG 3 ER PT J AU Darre, T Djiwa, T Dare, S Alassani, F Napo-Koura, G AF Darre, Tchin Djiwa, Toukilnan Dare, Sassil Alassani, Fousseni Napo-Koura, Gado TI Difficult Causality Relationship between Colorectal Cancer and Schistosomiasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE Colorectal cancer; Schistosomiasis; HIV; Carcinoma; Lymphoma ID Colorectal cancer; Schistosomiasis; HIV; Carcinoma; Lymphoma AB The purpose of the study was to determine the nature of the association between colorectal cancer and schistosomiasis infection. This was a retrospective and descriptive study of colorectal cancers and to identify cases associated with schistosomiasis. A total of 814 cases of rectal colorectal cancer were collected and 3 cases were associated with schistosomiasis. These are 2 cases of Liberkunian adenocarcinoma and one case of malt lymphoma. The patients were all farmers and from rural areas. A co-infection with HIV was found in his 3 patients. Our data show an extreme rarity of the association colorectal cancer and schistosomiasis; it seems difficult to conceive of a causal relationship. C1 [Darre, Tchin] University Teaching Hospital of Lome, Department of PathologyLome, Togo. [Djiwa, Toukilnan] University Teaching Hospital of Lome, Department of PathologyLome, Togo. [Dare, Sassil] University Teaching Hospital of Lome, Department of Visceral SurgeryLome, Togo. [Alassani, Fousseni] University Teaching Hospital of Lome, Department of Visceral SurgeryLome, Togo. [Napo-Koura, Gado] University Teaching Hospital of Lome, Department of PathologyLome, Togo. RP Darre, T (reprint author), University Teaching Hospital of Lome, Department of Pathology, Lome, Togo. EM paolodarre@yahoo.fr CR Plummer M, de Martel C, Vignat J, Ferlay J, Bray F, Franceschi S, 2016, Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob Health 4(9):e609–e616 Lapoile E, Bellaiche G, Boucard M et al, 2008, Cancer du rectum associe a une infection a Schistosoma hematobium. Gastroenterol Clin Biol 30(10):1225–1226 Darre T, Kpatcha K, Tchaou M et al, 2015, Histological aspect of urinary schistosomiasis in Togo: results of a cohort of 192 cases. Bull Soc Pathol Exot 108(2):124–125 Ferguson AR, 1911, Associated bilharziosis and primary malignant disease of the urinary bladder with observations on a series of forty cases. J Pathol Bacteriol 16(1):76–94 Gaye AM, Doh K, Thiam I, Bentefouet L, Woto-Gaye G, 2016, Schistosomiasis and cancer: a fortuitous association or relationships cause and effect. Bull Cancer 103(9):806–807 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2020 VL 26 IS 1 BP 597 EP 598 DI 10.1007/s12253-018-00566-0 PG 2 ER PT J AU Luo, P Wu, S Yu, Y Ming, X Li, Sh Zuo, X Tu, J AF Luo, Ping Wu, Sanyun Yu, Yalan Ming, Xinliang Li, Shuo Zuo, Xuelan Tu, Jiancheng TI Current Status and Perspective Biomarkers in AFP Negative HCC: Towards Screening for and Diagnosing Hepatocellular Carcinoma at an Earlier Stage SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE HCC; Biomarker; AFP-negative; Diagnose ID HCC; Biomarker; AFP-negative; Diagnose AB Hepatocellular carcinoma (HCC) is one of the most malignant cancer with high morbidity and mortality which lead to a serious burden to society. AFP (alpha-fetoprotein) is the most widely used serum biomarker to detect HCC worldwide. However, no AFP elevation have been found in many HCC and AFP analysis can’t be used to screen HCC in these cases. Currently, many studies have been carried out to find reliable biomarker in diagnosing AFP-negative HCC. Such biomarker would help the diagnosis of AFP-negative HCC, ensuring the timely initiation of treatment. In this review, we highlight the important role of biomarkers that can differentiate AFP-negative HCCs, and discuss their potential clinical applications as biomarkers for the diagnosis of AFP-negative HCC. C1 [Luo, Ping] Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory MedicineWuhan, China. [Wu, Sanyun] Zhongnan Hospital of Wuhan University, Department of HematologyWuhan, China. [Yu, Yalan] Zhongnan Hospital of Wuhan University, Department of HematologyWuhan, China. [Ming, Xinliang] Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory MedicineWuhan, China. [Li, Shuo] Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory MedicineWuhan, China. [Zuo, Xuelan] Zhongnan Hospital of Wuhan University, Department of HematologyWuhan, China. [Tu, Jiancheng] Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory MedicineWuhan, China. RP Tu, J (reprint author), Zhongnan Hospital of Wuhan University, Department of Clinical Laboratory Medicine, Wuhan, China. EM jianchengtu@whu.edu.cn CR Gao T, Zhi J, Mu C, Gu S, Xiao J, Yang J,Wang Z, Xiang Y, 2018, One-step detection for two serological biomarker species to improve the diagnostic accuracy of hepatocellular carcinoma. Talanta 178:89–93 Cadier B, Bulsei J, Nahon P, Seror O, Laurent A, Rosa I, Layese R, Costentin C, Cagnot C, Durand-Zaleski I, Chevreul K, 2017, Early detection and curative treatment of hepatocellular carcinoma: a cost-effectiveness analysis in france and in the united states. Hepatology 65:1237–1248 Guo S, ChenW, Luo Y, Ren F, Zhong T, Rong M, Dang Y, Feng Z, Chen G, 2015, Clinical implication of long non-coding rna neat1 expression in hepatocellular carcinoma patients. Int J Clin Exp Pathol 8:5395–5402 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90 Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66:7–30 Waghray A, Murali AR, Menon KN, 2015, Hepatocellular carcinoma: From diagnosis to treatment. World J Hepatol 7:1020–1029 ChenKW, Ou TM, HsuCW, Horng CT, Lee CC, Tsai YY, Tsai CC, Liou YS, Yang CC, Hsueh CW, Kuo WH, 2015, Current systemic treatment of hepatocellular carcinoma: a review of the literature. World J Hepatol 7:1412–1420 Uygun Ilikhan S, Bilici M, Sahin H, Demir Akca AS, CanM, Oz II, Guven B, Buyukuysal MC, Ustundag Y, 2015, Assessment of the correlation between serum prolidase and alpha-fetoprotein levels in patients with hepatocellular carcinoma. World J Gastroenterol 21: 6999–7007 Quagliata L,MatterMS, Piscuoglio S, Arabi L, Ruiz C, Procino A, Kovac M, Moretti F, Makowska Z, Boldanova T, Andersen JB, Hammerle M, Tornillo L, Heim MH, Diederichs S, Cillo C, Terracciano LM, 2014, Long noncoding rna hottip/hoxa13 expression is associated with disease progression and predicts outcome in hepatocellular carcinoma patients. Hepatology 59:911–923 Benson AB 3rd, Abrams TA, Ben-Josef E, Bloomston PM, Botha JF, Clary BM, Covey A, Curley SA, D'Angelica MI, Davila R, Ensminger WD, Gibbs JF, Laheru D, Malafa MP, Marrero J, Meranze SG, Mulvihill SJ, Park JO, Posey JA, Sachdev J, Salem R, Sigurdson ER, Sofocleous C, Vauthey JN, Venook AP, Goff LW, Yen Y, Zhu AX, 2009, Nccn clinical practice guidelines in oncology: hepatobiliary cancers. J Natl Compr Cancer Netw 7:350–391 Villanueva A, Minguez B, Forner A, Reig M, Llovet JM, 2010, Hepatocellular carcinoma: novel molecular approaches for diagnosis, prognosis, and therapy. Annu Rev Med 61:317–328 Li J, Cheng ZJ, Liu Y, Yan ZL, Wang K, Wu D, Wan XY, Xia Y, LauWY,WuMC, Shen F, 2015, Serum thioredoxin is a diagnostic marker for hepatocellular carcinoma. Oncotarget 6:9551–9563 Johnson PJ, 1999, Role of alpha-fetoprotein in the diagnosis and management of hepatocellular carcinoma. J Gastroenterol Hepatol 14(Suppl):S32–S36 Trevisani F, D'Intino PE, Morselli-Labate AM, Mazzella G, Accogli E, Caraceni P, Domenicali M, De Notariis S, Roda E, Bernardi M, 2001, Serum alpha-fetoprotein for diagnosis of hepatocellular carcinoma in patients with chronic liver disease: influence of hbsag and anti-hcv status. J Hepatol 34:570–575 Han LL, Lv Y, Guo H, Ruan ZP, Nan KJ, 2014, Implications of biomarkers in human hepatocellular carcinoma pathogenesis and therapy. World J Gastroenterol 20:10249–10261 Chen S, Chen H, Gao S, Qiu S, Zhou H, Yu M, Tu J, 2017, Differential expression of plasma microrna-125b in hepatitis b virus-related liver diseases and diagnostic potential for hepatitis b virus-induced hepatocellular carcinoma. Hepatol Res 47:312–320 Bruix J, ShermanM(2011)Management of hepatocellular carcinoma: an update. Hepatology 53:1020–1022 Oda K, Ido A, Tamai T, Matsushita M, Kumagai K, Mawatari S, Saishoji A, Kure T, Ohno K, Toyokura E, Imanaka D, Moriuchi A, Uto H, Oketani M, Hashiguchi T, Tsubouchi H, 2011, Highly sensitive lens culinaris agglutinin-reactive alpha-fetoprotein is useful for early detection of hepatocellular carcinoma in patients with chronic liver disease. Oncol Rep 26:1227–1233 Oeda S, Iwane S, Takasaki M, Furukawa NE, Otsuka T, Eguchi Y, Anzai K, 2016, Optimal follow-up of patients with viral hepatitis improves the detection of early-stage hepatocellular carcinoma and the prognosis of survival. Intern Med 55:2749–2758 Berhane S, Toyoda H, Tada T, Kumada T, Kagebayashi C, Satomura S, Schweitzer N, Vogel A, Manns MP, Benckert J, Berg T, Ebker M, Best J, Dechene A, Gerken G, Schlaak JF, Weinmann A,Worns MA, Galle P, Yeo W, Mo F, Chan SL, Reeves H, Cox T, Johnson P, 2016, Role of the galad and balad-2 serologic models in diagnosis of hepatocellular carcinoma and prediction of survival in patients. Clin Gastroenterol Hepatol 14:875–886.e876 Liebman HA, Furie BC, Tong MJ, Blanchard RA, Lo KJ, Lee SD, ColemanMS, Furie B, 1984, Des-gamma-carboxy, abnormal, prothrombin as a serummarker of primary hepatocellular carcinoma. N Engl J Med 310:1427–1431 Furie B, Furie BC, 1990, Molecular basis of vitamin k-dependent gamma-carboxylation. Blood 75:1753–1762 Seo SI, Kim HS, KimWJ, Shin WG, Kim DJ, Kim KH, Jang MK, Lee JH, Kim JS, Kim HY, Kim DJ, Lee MS, Park CK, 2015, Diagnostic value of pivka-ii and alpha-fetoprotein in hepatitis b virus-associated hepatocellular carcinoma. World J Gastroenterol 21:3928–3935 Yu R, Tan Z, Xiang X, Dan Y, Deng G, 2017, Effectiveness of pivka-ii in the detection of hepatocellular carcinoma based on real-world clinical data. BMC Cancer 17:608 Ji J,WangH, LiY, Zheng L,YinY, Zou Z, Zhou F, ZhouW, Shen F, Gao C, 2016, Diagnostic evaluation of des-gamma-carboxy prothrombin versus alpha-fetoprotein for hepatitis b virus-related hepatocellular carcinoma in China: a large-scale, multicentre study. PLoS One 11:e0153227 Wang X, ZhangW, Liu Y, GongW, Sun P, Kong X, Yang M,Wang Z, 2017, Diagnostic value of prothrombin induced by the absence of vitamin k or antagonist-ii, pivka-ii, for early stage hbv related hepatocellular carcinoma. Infect Agent Cancer 12:47 Oka H, Saito A, Ito K, Kumada T, Satomura S, Kasugai H, Osaki Y, Seki T, KudoM, TanakaM(2001, Multicenter prospective analysis of newly diagnosed hepatocellular carcinoma with respect to the percentage of lens culinaris agglutinin-reactive alpha-fetoprotein. J Gastroenterol Hepatol 16:1378–1383 Kumada T, Toyoda H, Kiriyama S, Tanikawa M, Hisanaga Y, Kanamori A, Tada T, Tanaka J, Yoshizawa H, 2011, Predictive value of tumor markers for hepatocarcinogenesis in patients with hepatitis c virus. J Gastroenterol 46:536–544 Toyoda H, Kumada T, Tada T, Kaneoka Y, Maeda A, Kanke F, Satomura S, 2011, Clinical utility of highly sensitive lens culinaris agglutinin-reactive alpha-fetoprotein in hepatocellular carcinoma patients with alpha-fetoprotein <20 ng/ml. Cancer Sci 102:1025–1031 Best J, Bilgi H, Heider D, Schotten C, Manka P, Bedreli S, Gorray M, Ertle J, van Grunsven LA, Dechene A, 2016, The galad scoring algorithm based on afp, afp-l3, and dcp significantly improves detection of bclc early stage hepatocellular carcinoma. Z Gastroenterol 54:1296–1305 Zhang Z, ZhangY,Wang Y, Xu L, XuW(2016, Alpha-fetoproteinl3 and golgi protein 73 may serve as candidate biomarkers for diagnosing alpha-fetoprotein-negative hepatocellular carcinoma. OncoTargets Ther 9:123–129 Khraiwesh B, Zhu JK, Zhu J, 1819, Role of mirnas and sirnas in biotic and abiotic stress responses of plants. Biochim Biophys Acta 2012:137–148 ShiM, Jiang Y, Yang L, Yan S,Wang YG, Lu XJ, 2018, Decreased levels of serum exosomal mir-638 predict poor prognosis in hepatocellular carcinoma. J Cell Biochem 119:4711–4716 Liu HN,Wu H, Chen YJ, Tseng YJ, Bilegsaikhan E, Dong L, Shen XZ, Liu TT, 2017, Serum microrna signatures and metabolomics have high diagnostic value in hepatocellular carcinoma. Vet Comp Oncol 8:108810–108824 Lai YC, Ushio N, Rahman MM, Katanoda Y, Ogihara K, Naya Y, Moriyama A, Iwanaga T, Saitoh Y, Sogawa T, Sunaga T, Momoi Y, Izumi H, Miyoshi N, Endo Y, Fujiki M, Kawaguchi H, Miura N, 2018, Aberrant expression of microRNAs and the mir-1/met pathway in canine hepatocellular carcinoma. Vet Comp Oncol 16:288– 296 Lyra-Gonzalez I, Flores-Fong LE, Gonzalez-Garcia I, Medina- Preciado D, Armendariz-Borunda J, 2015, Micrornas dysregulation in hepatocellular carcinoma: insights in genomic medicine. World J Hepatol 7:1530–1540 Wu XM, Xi ZF, Liao P, Huang HD, Huang XY,Wang C,Ma Y, Xia Q, Yao JG, Long XD, 2017, Diagnostic and prognostic potential of serum microrna-4651 for patients with hepatocellular carcinoma related to aflatoxin b1. Oncotarget 8:81235–81249 Guo X, Lv X, Lv X, Ma Y, Chen L, Chen Y, 2017, Circulating mir- 21 serves as a serum biomarker for hepatocellular carcinoma and correlated with distant metastasis. Oncotarget 8:44050–44058 Zuo D, Chen L, Liu X, Wang X, Xi Q, Luo Y, Zhang N, Guo H, 2016, Combination of mir-125b and mir-27a enhances sensitivity and specificity of afp-based diagnosis of hepatocellular carcinoma. Tumour Biol 37:6539–6549 Lin XJ, Chong Y, Guo ZW, Xie C, Yang XJ, Zhang Q, Li SP, Xiong Y, Yuan Y, Min J, Jia WH, Jie Y, Chen MS, Chen MX, Fang JH, Zeng C, Zhang Y, Guo RP, Wu Y, Lin G, Zheng L, Zhuang SM, 2015, A serum microrna classifier for early detection of hepatocellular carcinoma: a multicentre, retrospective, longitudinal biomarker identification study with a nested case-control study. Lancet Oncol 16:804–815 Wang X, Song X, Zhuo W, Fu Y, Shi H, Liang Y, Tong M, Chang G, Luo Y, 2009, The regulatory mechanism of hsp90alpha secretion and its function in tumor malignancy. ProcNatlAcad SciUS A 106:21288–21293 Fu Y, Xu X, Huang D, Cui D, Liu L, Liu J, He Z, Liu J, Zheng S, Luo Y, 2017, Plasma heat shock protein 90alpha as a biomarker for the diagnosis of liver cancer: an official, large-scale, andmulticenter clinical trial. EBioMedicine 24:56–63 Fan Y, Li Y, Chen Y, Zhao YJ, Liu LW, Li J,Wang SL, Alolga RN, YinY,WangXM, Zhao DS, Shen JH, Meng FQ, Zhou X, Xu H, He GP, Lai MD, Li P, Zhu W, Qi LW, 2016, Comprehensive metabolomic characterization of coronary artery diseases. J Am Coll Cardiol 68:1281–1293 Luo P, Yin P, Hua R, Tan Y, Li Z, Qiu G, Yin Z, Xie X, Wang X, ChenW, Zhou L,Wang X, Li Y, Chen H, Gao L, Lu X,Wu T,Wang H, Niu J, Xu G, 2017, A large-scale, multi-center serum metabolite biomarkers identification study for the early detection of hepatocellular carcinoma. Hepatology , DOI 10.1002/hep.29561 Xu H, Dong X, Chen Y, Wang X, 2017, Serum exosomal hnRNPH1 mRNA as a novel marker for hepatocellular carcinoma. Clin Chem Lab Med 56:479–484 Caviglia GP, Abate ML, Petrini E, Gaia S, Rizzetto M, Smedile A, 2016, Highly sensitive alpha-fetoprotein, lens culinaris agglutininreactive fraction of alpha-fetoprotein and des-gammacarboxyprothrombin for hepatocellular carcinoma detection. Hepatol Res 46:E130–E135 Song P, Feng X, InagakiY, Song T, Zhang K,Wang Z, Zheng S, Ma K, Li Q, Kong D,Wu Q, Zhang T, Zhao X, Hasegawa K, Sugawara Y, Kokudo N, TangW(2014, Clinical utility of simultaneous measurement of alpha-fetoprotein and des-gamma-carboxy prothrombin for diagnosis of patients with hepatocellular carcinoma in China: a multi-center case-controlled study of 1,153 subjects. Biosci Trends 8:266–273 Tang J, Jiang R, Deng L, Zhang X, Wang K, Sun B, 2015, Circulation long non-coding rnas act as biomarkers for predicting tumorigenesis and metastasis in hepatocellular carcinoma. Oncotarget 6:4505–4515 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 599 EP 603 DI 10.1007/s12253-019-00585-5 PG 5 ER PT J AU Yuan, P Ge, Y Liu, X Wang, Sh Ye, Z Xu, H Chen, Z AF Yuan, Peng Ge, Yue Liu, Xiao Wang, Shen Ye, Zhangqun Xu, Hua Chen, Zhiqiang TI The Association of Androgen Receptor Expression with Renal Cell Carcinoma Risk: a Systematic Review and Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Renal cell carcinoma; Androgen receptor; Prognosis; Targeted therapy ID Renal cell carcinoma; Androgen receptor; Prognosis; Targeted therapy AB The relationship between androgen receptor expression and renal cell carcinoma risk remains controversial. This study is aimed to investigate the clinical significance of androgen receptor expression in renal cell carcinoma. A computerized bibliographic search of Embase, the PubMed, and Web of Science combined with manual research between 1977 and 2017 was conducted to explore the association between androgen receptor expression and clinicopathological features of renal cell carcinoma. Data were analyzed by a meta-analysis using RevMan 5.3 analysis software. Eleven retrospective studies with 1839 renal cell carcinoma cases were finally included according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines. It was found that there was no significant difference between androgen receptor expression and susceptibility, pathological type, metastatic status, metastatic type (lymph or distant metastasis) and cancer-specific survival of renal cell carcinoma (P > 0.05). However, positive androgen receptor expression was demonstrated to be significantly associated with male patients, lower pathological grade, and earlier tumor stage of renal cell carcinoma (OR = 1.69, 95% CI = 1.30–2.19, P < 0.0001; OR = 2.06, 95% CI = 1.49–2.85, P < 0.0001; OR = 2.81, 95% CI = 1.30–6.12, P = 0.009; respectively). In conclusion, higher androgen receptor expression was correlated with male patients, low tumor grade and early stage of renal cell carcinoma. Based on current results, androgen receptor-inhibited target therapy for renal cell carcinoma patients may be of limited benefits and should be taken into more evaluations. C1 [Yuan, Peng] Huazhong University of Science and Technology, Tongji Medical School, Tongji Hospital, Department of Urology, No.1095 Jie Fang Avenue, Hankou, 430030 Wuhan, China. [Ge, Yue] Huazhong University of Science and Technology, Tongji Medical School, Tongji Hospital, Department of Urology, No.1095 Jie Fang Avenue, Hankou, 430030 Wuhan, China. [Liu, Xiao] Huazhong University of Science and Technology, Tongji Medical School, Tongji Hospital, Department of Urology, No.1095 Jie Fang Avenue, Hankou, 430030 Wuhan, China. [Wang, Shen] Huazhong University of Science and Technology, Tongji Medical School, Tongji Hospital, Department of Urology, No.1095 Jie Fang Avenue, Hankou, 430030 Wuhan, China. [Ye, Zhangqun] Huazhong University of Science and Technology, Tongji Medical School, Tongji Hospital, Department of Urology, No.1095 Jie Fang Avenue, Hankou, 430030 Wuhan, China. [Xu, Hua] Huazhong University of Science and Technology, Tongji Medical School, Tongji Hospital, Department of Urology, No.1095 Jie Fang Avenue, Hankou, 430030 Wuhan, China. [Chen, Zhiqiang] Huazhong University of Science and Technology, Tongji Medical School, Tongji Hospital, Department of Urology, No.1095 Jie Fang Avenue, Hankou, 430030 Wuhan, China. RP Chen, Z (reprint author), Huazhong University of Science and Technology, Tongji Medical School, Tongji Hospital, Department of Urology, 430030 Wuhan, China. EM urol_chen@163.com CR Siegel RL, Miller KD, Jemal A, 2018, Cancer statistics, 2018. CA Cancer J Clin 68:7–30 Campbell S, Uzzo RG, AllafME, Bass EB, Cadeddu JA, Chang A, Clark PE, Davis BJ, Derweesh IH, Giambarresi L, Gervais DA, Hu SL, Lane BR, Leibovich BC, Pierorazio PM, 2017, Renal mass and localized renal Cancer: AUA guideline. J Urol 198:520–529 Delahunt B, Cheville JC, Martignoni G, Humphrey PA, Magi- Galluzzi C, McKenney J, Egevad L, Algaba F, Moch H, Grignon DJ, Montironi R, Srigley JR, Members of the ISUP Renal Tumor Panel, 2013, The International Society of Urological Pathology, ISUP, grading system for renal cell carcinoma and other prognostic parameters. Am J Surg Pathol 37:1490–1504 Kuthi L, Jenei A, Hajdu A, Nemeth I, Varga Z, Bajory Z, Pajor L, Ivanyi B, 2017, Prognostic factors for renal cell carcinoma subtypes diagnosed according to the 2016 WHO renal tumor classification: a study involving 928 patients. Pathol Oncol Res 23:689– 698 He D, Li L, Zhu G, Liang L, Guan Z, Chang L, Chen Y, Yeh S, Chang C, 2014, ASC-J9 suppresses renal cell carcinoma progression by targeting an androgen receptor-dependent HIF2α/ VEGF signaling pathway. Cancer Res 74:4420–4430 Zhang HL, Sheng XN, Li XS,Wang HK, Chi ZH, He ZS, Ye DW, Guo J, 2017, Sorafenib versus sunitinib as first-line treatment agents in Chinese patients with metastatic renal cell carcinoma: the largest multicenter retrospective analysis of survival and prognostic factors. BMC Cancer 17:16 Chiarini F, Evangelisti C, McCubrey JA, Martelli AM, 2015, Current treatment strategies for inhibiting mTOR in cancer. Trends Pharmacol Sci 36:124–135 Ciccarese C, Brunelli M, Montironi R, Fiorentino M, Iacovelli R, Heng D, Tortora G, Massari F, 2016, The prospect of precision therapy for renal cell carcinoma. Cancer Treat Rev 49:37–44 Ravaud A, Motzer RJ, Pandha HS, George DJ, Pantuck AJ, Patel A, Chang YH, Escudier B, Donskov F, Magheli A, Carteni G, Laguerre B, Tomczak P, Breza J, Gerletti P, Lechuga M, Lin X, Martini JF, Ramaswamy K, Casey M, Staehler M, Patard JJ, STRAC Investigators, 2016, Adjuvant Sunitinib in high-risk renalcell carcinoma after nephrectomy. N Engl J Med 375:2246–2254 Matsumoto T, Sakari M, Okada M, Yokoyama A, Takahashi S, Kouzmenko A, Kato S, 2013, The androgen receptor in health and disease. Annu Rev Physiol 75:201–224 Pihlajamaa P, Sahu B, Janne OA, 2015, Determinants of receptorand tissue-specific actions in androgen signaling. Endocr Rev 36: 357–384 Bennett NC, Gardiner RA, Hooper JD, Johnson DW, Gobe GC, 2010, Molecular cell biology of androgen receptor signalling. Int J Biochem Cell Biol 42:813–827 Yonekura S, Terauchi F, Hoshi K, Yamaguchi T, Kawai S, 2018, Androgen receptor predicts first and multiple recurrences in nonmuscle invasive urothelial carcinoma of the bladder. Pathol Oncol Res. , DOI 10.1007/s12253-018-0431-7 Olsen JR, Azeem W, Hellem MR, Marvyin K, Hua Y, Qu Y, Li L, Lin B, Ke XIS, Oyan AM, Kalland KH, 2016, Context dependent regulatory patterns of the androgen receptor and androgen receptor target genes. BMC Cancer 16:377 Liang L, Li L, Tian J, Lee SO, DangQ, HuangCK, Yeh S, Erturk E, Bushinsky D, Chang LS, He D, Chang C, 2014, Androgen receptor enhances kidney stone-CaOx crystal formation via modulation of oxalate biosynthesis & oxidative stress. Mol Endocrinol 28:1291– 1303 Li JY, Zhou T, Gao X, Xu C, Sun Y, Peng Y, Chang Z, Zhang Y, Jiang J,Wang L, Hou J, 2010, Testosterone and androgen receptor in human nephrolithiasis. J Urol 184:2360–2363 Chow WH, Dong LM, Devesa SS, 2010, Epidemiology and risk factors for kidney cancer. Nat Rev Urol 7:245–257 Bennett NC, Rajandram R, Ng KL, Gobe GC, 2014, Evaluation of steroid hormones and their receptors in development and progression of renal cell carcinoma. J Kidney Cancer VHL 1:17–25 Langner C, Ratschek M, Rehak P, Schips L, Zigeuner R, 2004, Steroid hormone receptor expression in renal cell carcinoma: an immunohistochemical analysis of 182 tumors. J Urol 171:611–614 Noh SJ, Kang MJ, Kim KM et al, 2013, Acetylation status of P53 and the expression of DBC1, SIRT1, and androgen receptor are associated with survival in clear cell renal cell carcinoma patients. Pathology 45:574–580 Huang Q, Sun Y, Ma X, Gao Y, Li X, Niu Y, Zhang X, Chang C, 2017, Androgen receptor increases hematogenous metastasis yet decreases lymphatic metastasis of renal cell carcinoma. Nat Commun 8:918 Foersch S, Schindeldecker M, Keith M, Tagscherer KE, Fernandez A, Stenzel PJ, Pahernik S, Hohenfellner M, Schirmacher P, RothW, Macher-Goeppinger S, 2017, Prognostic relevance of androgen receptor expression in renal cell carcinomas. Oncotarget 8:78545– 78555 Vrabel M, 2015, Preferred reporting items for systematic reviews and meta-analyses. Oncol Nurs Forum 42:552–554 Stang A, 2010, Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in metaanalyses. Eur J Epidemiol 25:603–605 Ha YS, Lee GT, Modi P, Kwon YS, Ahn H, Kim WJ, Kim IY, 2015, Increased expression of androgen receptor mRNA in human renal cell carcinoma cells is associated with poor prognosis in patients with localized renal cell carcinoma. J Urol 194:1441–1448 Williams EM, Higgins JP, Sangoi AR, McKenney JK, Troxell ML, 2015, Androgen receptor immunohistochemistry in genitourinary neoplasms. Int Urol Nephrol 4:81–85 Zhu G, Liang L, Li L, Dang Q, Song W, Yeh S, He D, Chang C, 2014, The expression and evaluation of androgen receptor in human renal cell carcinoma. Urology 83:510 e19–5510e24 Nakano E, Tada Y, Fujioka H, Matsuda M, Osafune M, Kotake T, Sato B, TakahaM, Sonoda T, 1984, Hormone receptor in renal cell carcinoma and correlation with clinical response to endocrine therapy. J Urol 132:240–245 Klotzl G, Otto U, Becker H, Klosterhalfen H, 1987, Determination of androgen, progestin and estrogen receptors with two different assays in renal cell carcinoma. Urol Int 42:100–104 Ronchi E, Pizzocaro G,Miodini P, Piva L, Salvioni R, Di Fronzo G, 1984, Steroid hormone receptors in normal and malignant human renal tissue: relationship with progestin therapy. J Steroid Biochem 21:329–335 Concolino G,Marocchi A, Toscano V, Di Silverio F, 1981, Nuclear androgen receptor as marker of responsiveness to medroxyprogesterone acetate in human renal cell carcinoma. J Steroid Biochem 15:397–402 KoryakinaY, TaHQ, Gioeli D(2014, Androgen receptor phosphorylation: biological context and functional consequences. Endocr Relat Cancer 21:T131–T145 Lin WJ, Izumi K, 2014, Androgen receptor, ccl2, and epithelialmesenchymal transition: a dangerous affair in the tumor microenvironment. Oncoimmunology 3:e27871 Tonelli F, Alossaimi M, Williamson L, Tate RJ, Watson DG, Chan E, Bittman R, Pyne NJ, Pyne S, 2013, The sphingosine kinase inhibitor 2-(p-hyroxyanilino)-4-(p-chlorophenyl, thiazole reduces androgen receptor expression via an oxidative stress- dependent mechanism. Br J Pharmacol 168:1497–1505 Carver BS, 2014, Strategies for targeting the androgen receptor axis in prostate cancer. Drug Discov Today 19:1493–1497 Miyamoto H, Yang Z, Chen YT, Ishiguro H, Uemura H, Kubota Y, Nagashima Y, Chang YJ, Hu YC, Tsai MY, Yeh S, Messing EM, Chang C, 2007, Promotion of bladder cancer development and progression by androgen receptor signals. J Natl Cancer Inst 99: 558–568 Chang C, Lee SO, Yeh S, Chang TM, 2014, Androgen receptor, AR, differential roles in hormone-related tumors including prostate, bladder, kidney, lung, breast and liver. Oncogene 33:3225– 3234 Schweizer MT, Yu EY, 2017, AR-signaling in human malignancies: prostate cancer and beyond. Cancers, Basel, 9:7 Vera-Badillo FE, Templeton AJ, de Gouveia P, Diaz-Padilla I, Bedard PL, al-Mubarak M, Seruga B, Tannock IF, Ocana A, Amir E, 2014, Androgen receptor expression and outcomes in early breast cancer: a systematic review and meta-analysis. J Natl Cancer Inst 106:djt319 Grogg A, Trippel M, Pfaltz K, Ladrach C, Droeser RA, Cihoric N, Salhia B, Zweifel M, Tapia C, 2015, Androgen receptor status is highly conserved during tumor progression of breast cancer. BMC Cancer 15:872 Gucalp A, Tolaney S, Isakoff SJ, Ingle JN, Liu MC, Carey LA, Blackwell K, Rugo H, Nabell L, Forero A, Stearns V, Doane AS, Danso M, Moynahan ME, Momen LF, Gonzalez JM, Akhtar A, Giri DD, Patil S, Feigin KN, Hudis CA, Traina TA, on behalf of the Translational Breast Cancer Research Consortium, TBCRC 011,, 2013, Phase II trial of bicalutamide in patients with androgen receptor- positive, estrogen receptor-negative metastatic breast Cancer. Clin Cancer Res 19:5505–5512 MaWL, Lai HC, Yeh S, Cai X, Chang C, 2014, Androgen receptor roles in hepatocellular carcinoma, fatty liver, cirrhosis and hepatitis. Endocr Relat Cancer 21:R165–R182 Ljungberg B, Bensalah K, Canfield S, Dabestani S, Hofmann F, Hora M, Kuczyk MA, Lam T, Marconi L, Merseburger AS, Mulders P, Powles T, Staehler M, Volpe A, Bex A, 2015, EAU guidelines on renal cell carcinoma: 2014 update. Eur Urol 67: 913–924 Tosco L, Van Poppel H, Frea B, Gregoraci G, Joniau S, 2013, Survival and impact of clinical prognostic factors in surgically treated metastatic renal cell carcinoma. Eur Urol 63:646–652 Kim SH, Kim S, Joo J, Seo HK, Joung JY, Lee KH, Chung J, 2016, A retrospective study of predictive factors for unexpectedly prolonged or shortened progression-free survival and overall survival among patients with metastatic renal cell carcinoma who received first-line targeted therapy. BMC Cancer 16:577 Wang K, Sun Y, TaoW, Fei X, Chang C, 2017, Androgen receptor, AR, promotes clear cell renal cell carcinoma, ccRCC, migration and invasion via altering the circHIAT1/miR-195-5p/29a-3p/29c- 3p/CDC42 signals. Cancer Lett 394:1–12 ZhaiW, SunY, Guo C, Hu G,WangM, Zheng J, LinWY, Huang Q, Li G, Zheng J, Chang C, 2017, LncRNA-SARCC suppresses renal cell carcinoma, RCC, progression via altering the androgen receptor, AR)/miRNA-143-3p signals. Cell Death Differ 24:1502–1517 Czarnecka AM, Niedzwiedzka M, Porta C, Szczylik C, 2016, Hormone signaling pathways as treatment targets in renal cell cancer, review). Int J Oncol 48:2221–2235 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 605 EP 614 DI 10.1007/s12253-019-00650-z PG 10 ER PT J AU Moosavi, MS Elham, Y AF Moosavi, Mahdieh-Sadat Elham, Yalda TI Aquaporins 1, 3 and 5 in Different Tumors, their Expression, Prognosis Value and Role as New Therapeutic Targets SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Aquaporin; Cancer; Prognosis ID Aquaporin; Cancer; Prognosis AB All different types of metabolism of tumors are dependent on the flow of water molecules through the biological membrane, where fluid transfer interceded by aquaporins (AQPs) are the basis means for water entrance into the cells or outside them. Aquaporins play other roles including cellular migration, cellular expansion and cellular adhesion facilitation. Therefore, regulators of AQPs may be useful anticancer agents. Medline, Scopus, Embase, and Web of Sciences were searched. From among the papers found, 106 were related to the subject. All of the examined cancers in relation to AQP1 included adenoid cystic carcinoma, bladder, breast, cervical, colon, colorectal, hepatocellular, lung, ovarian, plural mesothelioma, prostate, renal cell carcinoma and squamous cell carcinoma. All of the studied cancers in relation with AQP3 included gastric, breast, prostate, lung, pancreas, skin, bladder, squamous cell carcinoma, cervical, adenoid cystic carcinoma, colon, colorectal, ovarian, and hepatocellular cancers and with regard to AQP5 were lung, squamous cell carcinoma, ovarian, adenoid cystic carcinoma, breast, colon, colorectal, hepatic, pancreas, gallbladder, prostate, and gastric cancers. Over or under-expression of AQP1, 3 and is exist in the mentioned cancers across different studies. Over-expression of AQP1, AQP3 and AQP5 is clearly associated with carcinogenesis, metastasis, reduced survival rate, lymph node metastasis, poorer prognosis, and cellular migration. Also, cancer treatments in relation to these markers suggest AQP reduction during the treatment. C1 [Moosavi, Mahdieh-Sadat] Tehran University of Medical Sciences, Faculty of Dentistry, Dental Research Center, Dentistry Research Institute, Department of Oral MedicineTehran, Iran. [Elham, Yalda] Tehran University of Medical Sciences, Faculty of Dentistry, Dental Research Center, Dentistry Research Institute, Department of Oral MedicineTehran, Iran. RP Elham, Y (reprint author), Tehran University of Medical Sciences, Faculty of Dentistry, Dental Research Center, Dentistry Research Institute, Department of Oral Medicine, Tehran, Iran. EM y-elham@razi.tums.ac.ir CR Wang L, Zhang Y, Wu X, Yu G, 2016, Aquaporins: new targets for cancer therapy. Technol Cancer Res Treat 15(6):821–828 Verkman AS, Anderson MO, Papadopoulos MC, 2014, Aquaporins: important but elusive drug targets. Nat Rev Drug Discov 13(4):259–277 Papadopoulos MC, Saadoun S, 2015, Key roles of aquaporins in tumor biology. Biochim Biophys Acta Biomembr 1848(10): 2576–2583 Direito I, Madeira A, Brito MA, Soveral G, 2016, Aquaporin-5: from structure to function and dysfunction in cancer. CellMol Life Sci 73(8):1623–1640 Takata K, Matsuzaki T, Tajika Y, 2004, Aquaporins: water channel proteins of the cell membrane. Prog Histochem Cytochem 39(1):1–83 Tomita Y, Dorward H, Yool A, Smith E, Townsend A, Price T, Hardingham J, 2017, Role of aquaporin 1 signalling in cancer development and progression. Int J Mol Sci 18(2):299 Niu D, Kondo T, Nakazawa T, Yamane T, Mochizuki K, Kawasaki T, Matsuzaki T, Takata K, Katoh R, 2012, Expression of aquaporin3 in human neoplastic tissues. Histopathology. 61(4): 543–551 Jensen HH, Login FH, Koffman JS, Kwon T-H, Nejsum LN, 2016, The role of aquaporin-5 in cancer cell migration: a potential active participant. Int J Biochem Cell Biol 79:271–276 TanM, Shao C, Bishop JA, Feng Z, Trock BJ,WestraWH, Ha PK, 2014)Aquaporin-1 promoter hypermethylation is associated with improved prognosis in salivary gland adenoid cystic carcinoma. Otolaryngol Head Neck Surg 150(5):801–807 Zhang X, Chen Y, Dong L, Shi B, 2018, Effect of selective inhibition of aquaporin 1 on chemotherapy sensitivity of J82 human bladder cancer cells. Oncol Lett 15(3):3864–3869 Zou W, Yang Z, Li D, Liu Z, Zou Q, Yuan Y, 2019, AQP1 and AQP3 expression are associated with severe symptoms and poorprognosis of the pancreatic ductal adenocarcinoma. Appl Immunohistochem Mol Morphol 27(1):40–47 Luo L,Yang R, Zhao S, ChenY, Hong S,Wang K,Wang T, Cheng J, Zhang T, Chen D, 2018, Decreased miR-320 expression is associated with breast cancer progression, cell migration, and invasiveness via targeting aquaporin 1. Acta Biochim Biophys Sin 50(5):473–480 Qin F, Zhang H, Shao Y, Liu X, Yang L, Huang Y et al, 2016, Expression of aquaporin1, a water channel protein, in cytoplasm is negatively correlated with prognosis of breast cancer patients. Oncotarget. 7(7):8143 Yin T, Yu S, Xiao L, Zhang J, Liu C, Lu Yet al, 2008, Correlation between the expression of aquaporin 1 and hypoxia-inducible factor 1 in breast cancer tissues. J Huazhong Univ Sci Technolog Med Sci 28(3):346 Shi Y-H, Chen R, Talafu T, Nijiati R, Lalai S, 2012, Significance and expression of aquaporin 1, 3, 8 in cervical carcinoma in Xinjiang Uygur women of China. Asian Pac J Cancer Prev 13(5):1971–1975 Dorward HS, Du A, Bruhn MA,Wrin J, Pei JV, Evdokiou A et al, 2016, Pharmacological blockade of aquaporin-1 water channel by AqB013 restricts migration and invasiveness of colon cancer cells and prevents endothelial tube formation in vitro. J Exp Clin Cancer Res 35(1):36 Jiang Y, 2009, Aquaporin-1 activity of plasma membrane affects HT20 colon cancer cell migration. IUBMB Life 61(10):1001– 1009 Kang BW, Kim JG, Lee SJ, Chae YS, Jeong JY, Yoon GS, Park SY, Kim HJ, Park JS, Choi GS, Jeong JY, 2015, Expression of aquaporin-1, aquaporin-3, and aquaporin-5 correlates with nodal metastasis in colon cancer. Oncology. 88(6):369–376 Yoshida T, Hojo S, Sekine S, Sawada S, Okumura T, Nagata Tet al, 2013, Expression of aquaporin-1 is a poor prognostic factor for stage II and III colon cancer. Mol Clin Oncol 1(6):953–958 Imaizumi H, Ishibashi K, Takenoshita S, Ishida H, 2018, Aquaporin 1 expression is associated with response to adjuvant chemotherapy in stage II and III colorectal cancer. Oncol Lett 15(5):6450–6456 Luo LM, Xia H, Shi R, Zeng J, Liu XR, Wei M, 2017, The association between aquaporin-1 expression, microvessel density and the clinicopathological features of hepatocellular carcinoma. Oncol Lett 14(6):7077–7084 Bellezza G, Vannucci J, Bianconi F, Metro G, Del Sordo R, Andolfi M et al, 2017, Prognostic implication of aquaporin 1 overexpression in resected lung adenocarcinoma. Interact Cardiovasc Thorac Surg 25(6):856–861 Hoque MO, Soria J-C,Woo J, Lee T, Lee J, Jang SJ, Upadhyay S, Trink B, Monitto C, Desmaze C, Mao L, Sidransky D, Moon C, 2006, Aquaporin 1 is overexpressed in lung cancer and stimulates NIH-3T3 cell proliferation and anchorage-independent growth. Am J Pathol 168(4):1345–1353 Liu Y, ZhuW(2015, Effects of cetuximab combined with afatinib on the expression of KDR and AQP1 in lung cancer. Genet Mol Res 14(4):16652–16661 Takal MK, Baykal C, Baser E, Kaya MD, Dursun P, Ozen O et al, 2013, Does Aquaporin-1 expression have clinical significance in serous epithelial ovarian cancer? J Turk Ger Gynecol Assoc 14(3): 130–135 Sato K, Miyamoto M, Takano M, Furuya K, Tsuda H, 2018, Different prognostic implications of Aquaporin-1 and Aquaporin-5 expression among different histological types of ovarian carcinoma. Pathol Oncol Res:1–9 Wang Y, Fan Y, Zheng C, Zhang X, 2017, Knockdown of AQP1 inhibits growth and invasion of human ovarian cancer cells. Mol Med Rep 16(4):5499–5504 Angelico G, Caltabiano R, Loreto C, Ieni A, Tuccari G, Ledda C, Rapisarda V, 2018, Immunohistochemical expression of Aquaporin-1 in Fluoro-Edenite-induced malignant mesothelioma: a preliminary report. Int J Mol Sci 19(3):685 Jagirdar RM, Apostolidou E, Molyvdas PA, Gourgoulianis KI, Hatzoglou C, Zarogiannis SG, 2016, Influence of AQP1 on cell adhesion, migration, and tumor sphere formation in malignant pleural mesothelioma is substratum-and histological-type dependent. Am J Phys Lung Cell Mol Phys 310(6):L489–LL95 Kao SCH, Armstrong N, Condon B, Griggs K, McCaughan B, Maltby S,Wilson A, HendersonDW, Klebe S, 2012, Aquaporin 1 is an independent prognostic factor in pleural malignant mesothelioma. Cancer. 118(11):2952–2961 Pan X-Y, Guo H, Han J, Hao F, An Y, Xu Y, Xiaokaiti Y, Pan Y, Li XJ, 2012, Ginsenoside Rg3 attenuates cell migration via inhibition of aquaporin 1 expression in PC-3M prostate cancer cells. Eur J Pharmacol 683(1–3):27–34 Tie L, Lu N, Pan X-Y, Pan Y, An Y, Gao J-W, Lin YH, Yu HM, Li XJ, 2012, Hypoxia-induced up-regulation of aquaporin-1 protein in prostate cancer cells in a p38-dependent manner. Cell Physiol Biochem 29(1–2):269–280 Huang Y, Murakami T, Sano F, Kondo K, Nakaigawa N, Kishida T, Kubota Y, Nagashima Y, Yao M, 2009, Expression of aquaporin 1 in primary renal tumors: a prognostic indicator for clear-cell renal cell carcinoma. Eur Urol 56(4):690–699 Morrissey JJ, Kharasch ED, 2013, The specificity of urinary aquaporin 1 and perilipin 2 to screen for renal cell carcinoma. J Urol 189(5):1913–1920 Morrissey JJ, Mobley J, Song J, Vetter J, Luo J, Bhayani S et al, 2014, Urinary concentrations of aquaporin-1 and perilipin-2 in patients with renal cell carcinoma correlate with tumor size and stage but not grade. Urology 83(1):256. e9–256. e14 Morrissey JJ, Mobley J, Figenshau RS, Vetter J, Bhayani S, Kharasch ED, eds, 2015, Urine aquaporin 1 and perilipin 2 differentiate renal carcinomas from other imaged renal masses and bladder and prostate cancer. Mayo Clinic Proceedings. Elsevier Morrissey JJ, Mellnick VM, Luo J, Siegel MJ, Figenshau RS, Bhayani S, Kharasch ED, 2015, Evaluation of urine aquaporin-1 and perilipin-2 concentrations as biomarkers to screen for renal cell carcinoma: a prospective cohort study. JAMA Oncol 1(2): 204–212 Sreedharan S, Petros JA,Master VA, Ogan K, Pattaras JG, Roberts DL, Lian F, Arnold RS, 2014, Aquaporin-1 protein levels elevated in fresh urine of renal cell carcinoma patients: potential use for screening and classification of incidental renal lesions. Dis Markers 2014:1–6 Ticozzi-Valerio D, Raimondo F, Pitto M, Rocco F, Bosari S, Perego R, Sarto C, di Fonzo A, Bosso N, Mocarelli P, Galli- Kienle M, Magni F, 2007, Differential expression of AQP1 in microdomain-enriched membranes of renal cell carcinoma. Proteomics Clin Appl 1(6):588–597 Lehnerdt G, BachmannH, Adamzik M, Panic A, Koksal E,Weller P et al, 2015, AQP1, AQP5, Bcl-2 and p16 in pharyngeal squamous cell carcinoma. J Laryngol Otol 129(6):580–586 Yamazato Y, Shiozaki A, Ichikawa D, Kosuga T, Shoda K, Arita T et al, 2018, Aquaporin 1 suppresses apoptosis and affects prognosis in esophageal squamous cell carcinoma. Oncotarget. 9(52): 29957 Imredi E, Toth B, Doma V, Barbai T, Raso E, Kenessey I, Timar J, 2016, Aquaporin 1 protein expression is associated with BRAF V600 mutation and adverse prognosis in cutaneous melanoma. Melanoma Res 26(3):254–260 Imredi E, Liszkay G, Kenessey I, Plotar V, Godeny M, Toth B et al, 2018, Aquaporin-1 protein expression of the primary tumor may predict cerebral progression of cutaneous melanoma. Pathol Oncol Res:1–6 Zhou Y, Wang Y, Wen J, Zhao H, Dong X, Zhang Z et al, 2016, Aquaporin 3 promotes the stem-like properties of gastric cancer cells via Wnt/GSK-3β/β-catenin pathway. Oncotarget. 7(13):16529 Chen J, Wang T, Zhou Y-C, Gao F, Zhang Z-H, Xu H, Wang SL, Shen LZ, 2014, Aquaporin 3 promotes epithelial-mesenchymal transition in gastric cancer. J Exp Clin Cancer Res 33(1):38 Zhao H, Wen J, Dong X, He R, Gao C, Zhang W et al, 2017, Identification of AQP3 and CD24 as biomarkers for carcinogenesis of gastric intestinal metaplasia. Oncotarget. 8(38):63382 Zhao H, Yang X, Zhou Y, Zhang W, Wang Y, Wen J et al, 2015, Potential role of aquaporin 3 in gastric intestinal metaplasia. Oncotarget. 6(36):38926 Wang J, Gui Z, Deng L, Sun M, Guo R, ZhangW, Shen L, 2012, C-met upregulates aquaporin 3 expression in human gastric carcinoma cells via the ERK signalling pathway. Cancer Lett 319(1): 109–117 Xu H, Xu Y, ZhangW, Shen L, Yang L, Xu Z, 2011, Aquaporin-3 positively regulates matrix metalloproteinases via PI3K/AKT signal pathway in human gastric carcinoma SGC7901 cells. J Exp Clin Cancer Res 30(1):86 Wang G, Gao F, Zhang W, Chen J, Wang T, Zhang G, Shen L, 2012, Involvement of aquaporin 3 in helicobacter pylori-related gastric diseases. PLoS One 7(11):e49104 Li Z, Li B, Zhang L, Chen L, Sun G, Zhang Q, Wang J, Zhi X, Wang L,Xu Z, Xu H, 2016, The proliferation impairment induced by AQP3 deficiency is the result of glycerol uptake and metabolism inhibition in gastric cancer cells. Tumor Biol 37(7):9169– 9179 Jiang B, Li Z, Zhang W, Wang H, Zhi X, Feng J, Chen Z, Zhu Y, Yang L, Xu H, Xu Z, 2014, miR-874 inhibits cell proliferation, migration and invasion through targeting aquaporin-3 in gastric cancer. J Gastroenterol 49(6):1011–1025 Zhou Y, Wang Y, Wang S, Shen L, 2015, Hyperglycemia promotes human gastric carcinoma progression via aquaporin 3. Dig Dis Sci 60(8):2338–2345 Kang S, Chae YS, Lee SJ, Kang BW, Kim JG, KimWWet al, 2015, Aquaporin 3 expression predicts survival in patients with HER2-positive early breast cancer. Anticancer Res 35(5):2775–2782 Cao X-C, Zhang W-R, Cao W-F, Liu B-W, Zhang F, Zhao H-M, Meng R, Zhang L, Niu RF, Hao XS, Zhang B, 2013, Aquaporin3 is required for FGF-2-induced migration of human breast cancers. PLoS One 8(2):e56735 Huang Y-T, Zhou J, Shi S, Xu H-Y, Qu F, Zhang D, Chen YD, Yang J, Huang HF, Sheng JZ, 2015, Identification of estrogen response element in aquaporin-3 gene that mediates estrogeninduced cell migration and invasion in estrogen receptor-positive breast cancer. Sci Rep 5:12484 Satooka H, Hara-Chikuma M, 2016, Aquaporin-3 controls breast cancer cell migration by regulating hydrogen peroxide transport and its downstream cell signaling.Molecular and cellular biology. MCB. 00971–15 Arif M, Kitchen P, Conner MT, Hill EJ, Nagel D, Bill RM et al, 2018, Downregulation of aquaporin 3 inhibits cellular proliferation, migration and invasion in the MDA-MB-231 breast cancer cell line. Oncol Lett 16(1):713–720 Hwang I, Jung S-I, Hwang E-C, Song SH, Lee H-S, Kim S-O, Kang TW, Kwon D, Park K, 2012, Expression and localization of aquaporins in benign prostate hyperplasia and prostate cancer. Chonnam Med J 48(3):174–178 Ismail M, Bokaee S, Davies J, Harrington K, Pandha H, 2009, Inhibition of the aquaporin 3 water channel increases the sensitivity of prostate cancer cells to cryotherapy. Br J Cancer 100(12):1889–1895 Chen J, Wang Z, Xu D, Liu Y, Gao Y, 2015, Aquaporin 3 promotes prostate cancer cell motility and invasion via extracellular signal-regulated kinase 1/2-mediated matrix metalloproteinase-3 secretion. Mol Med Rep 11(4):2882–2888 Wang J, Tanji N, Kikugawa T, Shudou M, Song X, Yokoyama M, 2007, Expression of aquaporin 3 in the human prostate. Int J Urol 14(12):1088–1092 Xiong G, Chen X, Zhang Q, Fang Y, Chen W, Li C, Zhang J, 2017, RNA interference influenced the proliferation and invasion of XWLC-05 lung cancer cells through inhibiting aquaporin 3. Biochem Biophys Res Commun 485(3):627–634 Hou S-Y, Li Y-P,Wang J-H,Yang S-L,WangY,WangY, KuangY, 2016, Aquaporin-3 inhibition reduces the growth of NSCLC cells induced by hypoxia. Cell Physiol Biochem 38(1):129–140 Direito I, Paulino J, Vigia E, Brito MA, Soveral G, 2017, Differential expression of aquaporin-3 and aquaporin-5 in pancreatic ductal adenocarcinoma. J Surg Oncol 115(8):980–996 Huang X, Huang L, ShaoM, 2017, Aquaporin 3 facilitates tumor growth in pancreatic cancer by modulating mTOR signaling. Biochem Biophys Res Commun 486(4):1097–1102 Liu W, Wang K, Gong K, Li X, Luo K, 2012, Epidermal growth factor enhances MPC-83 pancreatic cancer cell migration through the upregulation of aquaporin 3. Mol Med Rep 6(3):607–610 RamadanWM, Gheida SF, El-AshmawyAA, ShareefMM(2017, Aquaporin-3 expression in common hyperproliferative skin disorders: an immunohistochemical study. J Egypt Women Dermatol Soc 14(2):128–136 Seleit I, Bakry OA, Sharaky DA, Ragheb E, 2015, Evaluation of aquaporin-3 role in nonmelanoma skin cancer: an immunohistochemical study. Ultrastruct Pathol 39(5):306–317 OttoW, Rubenwolf PC, Burger M, Fritsche H-M, RoßlerW, May M, Hartmann A, Hofstadter F, Wieland WF, Denzinger S, 2012, Loss of aquaporin 3 protein expression constitutes an independent prognostic factor for progression-free survival: an immunohistochemical study on stage pT1 urothelial bladder cancer. BMC Cancer 12(1):459 Rubenwolf P, Thomas C, Denzinger S, Hartmann A, Burger M, Georgopoulos NT, Otto W, 2015, Loss of AQP3 protein expression is associated with worse progression-free and cancer-specific survival in patients with muscle-invasive bladder cancer. World J Urol 33(12):1959–1964 Ishimoto S,WadaK, UsamiY, Tanaka N, Aikawa T, OkuraMet al, 2012, Differential expression of aquaporin 5 and aquaporin 3 in squamous cell carcinoma and adenoid cystic carcinoma. Int J Oncol 41(1):67–75 Soland TM, Ulekleiv CH, Galtung HK, 2015, Hypoxia influence aquaporin 3, AQP3, and migration in oral squamous cell carcinoma, OSCC). Oral Surg Oral Med Oral Pathol Oral Radiol 119(3):e181 Kusayama M, Wada K, Nagata M, Ishimoto S, Takahashi H, Yoneda M, Nakajima A, Okura M, Kogo M, Kamisaki Y, 2011, Critical role of aquaporin 3 on growth of human esophageal and oral squamous cell carcinoma. Cancer Sci 102(6):1128–1136 Chen R, Shi Y, Amiduo R, Tuokan T, Suzuk L, 2014, Expression and prognostic value of aquaporin 1, 3 in cervical carcinoma in women of Uygur ethnicity from Xinjiang, China. PloS One 9(6):e98576 Li A, Lu D, ZhangY, Li J, Fang Y, Li F et al, 2013, Critical role of aquaporin-3 in epidermal growth factor-induced migration of colorectal carcinoma cells and its clinical significance. Oncol Rep 29(2):535–540 Xuejun C, Weimin C, Xiaoyan D, Wei Z, Qiong Z, Jianhua Y, 2014, Effects of aquaporins on chemosensitivity to cisplatin in ovarian cancer cells. Arch Gynecol Obstet 290(3):525–532 Ji C, Cao C, Lu S, Kivlin R, Amaral A, Kouttab N, Yang H, Chu W, Bi Z, di W, Wan Y, 2008, Curcumin attenuates EGF-induced AQP3 up-regulation and cell migration in human ovarian cancer cells. Cancer Chemother Pharmacol 62(5):857–865 Peng R, Zhang Y, Zhao G, Li J, Shen X, Wang J, Sun JY, 2016, Differential regulation of the expression of aquaporins 3 and 9 by Auphen and dbcAMP in the SMMC-7721 hepatocellular carcinoma cell line. Biotech Histochem 91(5):333–341 Chen G, Shi Y, Liu M, Sun J, 2018, circHIPK3 regulates cell proliferation and migration by sponging miR-124 and regulating AQP3 expression in hepatocellular carcinoma. Cell Death Dis 9(2):175 Machida Y, Ueda Y, ShimasakiM, Sato K, Sagawa M, Katsuda S, Sakuma T, 2011, Relationship of aquaporin 1, 3, and 5 expression in lung cancer cells to cellular differentiation, invasive growth, and metastasis potential. Hum Pathol 42(5):669–678 Jo YM, Park TI, Lee HY, Jeong JY, Lee WK, 2016, Prognostic significance of aquaporin 5 expression in non-small cell lung cancer. J Pathol Transl Med 50(2):122–128 Song T, Yang H, Ho JCM, Tang SCW, Sze SCW, Lao L et al, 2015, Expression of aquaporin 5 in primary carcinoma and lymph node metastatic carcinoma of non-small cell lung cancer. Oncol Lett 9(6):2799–2804 Chae YK, Woo J, Kim M-J, Kang SK, Kim MS, Lee J, Lee SK, Gong G, Kim YH, Soria JC, Jang SJ, Sidransky D, Moon C, 2008, Expression of aquaporin 5, AQP5, promotes tumor invasion in human non small cell lung cancer. PLoS One 3(5):e2162 Liu S, Zhang S, Jiang H, Yang Y, Jiang Y, 2013, Co-expression of AQP3 and AQP5 in esophageal squamous cell carcinoma correlates with aggressive tumor progression and poor prognosis. Med Oncol 30(3):636 Shimizu H, Shiozaki A, Ichikawa D, Fujiwara H, Konishi H, Ishii H, Komatsu S, Kubota T, Okamoto K, Kishimoto M, Otsuji E, 2014, The expression and role of aquaporin 5 in esophageal squamous cell carcinoma. J Gastroenterol 49(4):655–666 Yan C, Yang J, Shen L, Chen X, 2012, Inhibitory effect of epigallocatechin gallate on ovarian cancer cell proliferation associated with aquaporin 5 expression. Arch Gynecol Obstet 285(2):459–467 Yang J, Yan C, Zheng W, Chen X, 2012, Proliferation inhibition of cisplatin and aquaporin 5 expression in human ovarian cancer cell CAOV3. Arch Gynecol Obstet 285(1):239–245 Chen X, Yang J, Zheng W, 2009, Effect of topotecan on expression of aquaporin protein 5 and nuclear factorB! B in ovarian cancer SKOV3 cells. Chi J Cancer 28(8) Yang J-H, Shi Y-F, Cheng Q, Deng L, 2006, Expression and localization of aquaporin-5 in the epithelial ovarian tumors. Gynecol Oncol 100(2):294–299 Yan C, Zhu Y, Zhang X, Chen X, ZhengW, Yang J, 2014, Downregulated aquaporin 5 inhibits proliferation and migration of human epithelial ovarian cancer 3AO cells. J Ovarian Res 7(1):78 Jung HJ, Park J-Y, Jeon H-S, Kwon T-H, 2011, Aquaporin-5: a marker protein for proliferation and migration of human breast cancer cells. PLoS One 6(12):e28492 Jensen HH, Login FH, Park J-Y, Kwon T-H, Nejsum LN, 2017, Immunohistochemical evalulation of activated Ras and Rac1 as potential downstream effectors of aquaporin-5 in breast cancer in vivo. Biochem Biophys Res Commun 493(3):1210–1216 Lee SJ, Chae YS, KimJG, KimWW, Jung JH, Park HY, Jeong JY, Park JY, Jung HJ, Kwon TH, 2014, AQP5 expression predicts survival in patients with early breast cancer. Ann Surg Oncol 21(2):375–383 Zhu Z, Jiao L, Li T,Wang H,WeiW, Qian H, 2018, Expression of AQP3 and AQP5 as a prognostic marker in triple-negative breast cancer. Oncol Lett 16(2):2661–2667 Guo K, Jin F, 2015, NFAT5 promotes proliferation and migration of lung adenocarcinoma cells in part through regulating AQP5 expression. Biochem Biophys Res Commun 465(3):644–649 Zhang Z-Q, Zhu Z-X, Bai C-X, Chen Z-H, 2011, Aquaporin 5 expression increases mucin production in lung adenocarcinoma. Oncol Rep 25(6):1645–1650 Moon C, Soria J-C, Jang SJ, Lee J, Hoque MO, Sibony M, Trink B, Chang YS, Sidransky D, Mao L, 2003, Involvement of aquaporins in colorectal carcinogenesis. Oncogene. 22(43):6699–6703 Chen C, Ma T, Zhang C, Zhang H, Bai L, Kong L, Luo J, 2017, Down-regulation of aquaporin 5-mediated epithelialmesenchymal transition and anti-metastatic effect by natural product Cairicoside E in colorectal cancer. Mol Carcinog 56(12):2692–2705 Shi X, Wu S, Yang Y, Tang L, Wang Y, Dong J, Lu B, Jiang G, Zhao W, 2014, AQP5 silencing suppresses p38 MAPK signaling and improves drug resistance in colon cancer cells. Tumor Biol 35(7):7035–7045 Guo X, Sun T, Yang M, Li Z, Li Z, Gao Y, 2013, Prognostic value of combined aquaporin 3 and aquaporin 5 overexpression in hepatocellular carcinoma. Biomed Res Int 2013:1–7 He Z, Dong W, Hu J, Ren X, 2017, AQP5 promotes hepatocellular carcinoma metastasis via NF-κB-regulated epithelialmesenchymal transition. Biochem Biophys Res Commun 490(2):343–348 Kudou M, Shiozaki A, Kosuga T, Shimizu H, Ichikawa D, Konishi H, Morimura R, Komatsu S, Ikoma H, Fujiwara H, Okamoto K, Marunaka Y, Otsuji E, 2017, Heat shock exerts anticancer effects on liver cancer via autophagic degradation of aquaporin 5. Int J Oncol 50(5):1857–1867 Pust A, Kylies D, Hube-Magg C, Kluth M, Minner S, Koop C, Grob T, Graefen M, Salomon G, Tsourlakis MC, Izbicki J, Wittmer C, Huland H, Simon R, Wilczak W, Sauter G, Steurer S, Krech T, Schlomm T, Melling N, 2016, Aquaporin 5 expression is frequent in prostate cancer and shows a dichotomous correlation with tumor phenotype and PSA recurrence. Hum Pathol 48:102–110 Watanabe T, Fujii T, Oya T, Horikawa N, Tabuchi Y, Takahashi Y, Morii M, Takeguchi N, Tsukada K, Sakai H, 2009, Involvement of aquaporin-5 in differentiation of human gastric cancer cells. J Physiol Sci 59(2):113–122 HuangY-H, Zhou X-Y,Wang H-M, Xu H, Chen J, Lv N-H, 2013, Aquaporin 5 promotes the proliferation and migration of human gastric carcinoma cells. Tumor Biol 34(3):1743–1751 Verkman A, 2009, Aquaporins: translating bench research to human disease. J Exp Biol 212(11):1707–1715 Nico B, Ribatti D, 2010, Aquaporins in tumor growth and angiogenesis. Cancer Lett 294(2):135–138 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 615 EP 625 DI 10.1007/s12253-019-00646-9 PG 11 ER PT J AU Singh, G Mishra, S Chander, H AF Singh, Garima Mishra, Sarthak Chander, Harish TI KIBRA Team Up with Partners to Promote Breast Cancer Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Breast Cancer; Metastasis; Kibra; DDR1; DLC1&PKCζ ID Breast Cancer; Metastasis; Kibra; DDR1; DLC1&PKCζ AB Among women, breast cancer is the most frequently diagnosed cancer. Most of the breast cancers represent metastasis to distant organs at the time of diagnosis and accounts for the majority of deaths. Metastasis is characterized by many genetic aberrations including mutations, overexpression of oncogenes etc. KIBRA (KIdney/BRAin protein), a scaffolding protein is recently described as an important player in the process of invasion and metastasis. The Kidney/BRAin protein through its different domains interacts with various proteins to couple cytoskeleton arrangement, cell polarity and migration. N terminal and C terminal of the protein contains the WW, Internal C2 & putative class III PDZ domain that interacts with DDR1, DLC1 & PKCζ. These protein-protein interactions equip the breast cancer cells to invade and metastasize. Here, we discuss a comprehensive knowledge about the KIBRA protein, its domains and the interacting partners involved in metastasis of breast cancer. C1 [Singh, Garima] Central University of Punjab, Department of Human Genetics and Molecular Medicine, Laboratory of Molecular Medicine, 151001 Bathinda, India. [Mishra, Sarthak] Central University of Punjab, Department of Human Genetics and Molecular Medicine, Laboratory of Molecular Medicine, 151001 Bathinda, India. [Chander, Harish] Central University of Punjab, Department of Human Genetics and Molecular Medicine, Laboratory of Molecular Medicine, 151001 Bathinda, India. RP Chander, H (reprint author), Central University of Punjab, Department of Human Genetics and Molecular Medicine, Laboratory of Molecular Medicine, 151001 Bathinda, India. EM harish.chander@cup.edu.in CR Friedenreich CM, 2011, Physical activity and breast Cancer: review of the epidemiologic evidence and biologic mechanisms. In: Senn H-J, Otto F, eds, Clinical Cancer prevention. Springer Berlin Heidelberg, Berlin, pp 125–139 Zhang X, Liu X, Luo J, Xiao W, Ye X, Chen M, Li Y, Zhang GJ, 2016, Notch3 inhibits epithelial–mesenchymal transition by activating Kibra-mediated hippo/YAP signaling in breast cancer epithelial cells. Oncogenesis 5(11):e269. , DOI 10.1038/ oncsis.2016.67 Kang Y, Massague J, 2004, Epithelial-mesenchymal transitions: twist in development and metastasis. [Short Survey]. Cell 118(3): 277–279. , DOI 10.1016/j.cell.2004.07.011 Thiery JP,MorganM(2004, Breast cancer progression with a twist. Nat Med 10:777–778. , DOI 10.1038/nm0804-777 Thiery JP, Sleeman JP, 2006, Complex networks orchestrate epithelial–mesenchymal transitions. [review article]. Nat Rev Mol Cell Biol 7:131. , DOI 10.1038/nrm1835 Ahmad A, Sarkar SH, Bitar B, Ali S, Aboukameel A, Sethi S, Li Y, Bao B, Kong D, Banerjee S, Padhye SB, Sarkar FH, 2012, Garcinol Regulates EMT and Wnt Signaling Pathways In Vitro and In Vivo, Leading to Anticancer Activity against Breast Cancer Cells.Mol Cancer Ther 11(10):2193–2201. , DOI 10.1158/ 1535-7163.mct-12-0232-t Debies MT, Gestl SA, Mathers JL,Mikse OR, Leonard TL,Moody SE, Chodosh LA, Cardiff RD, Gunther EJ, 2008, Tumor escape in a Wnt1-dependent mouse breast cancer model is enabled by p19(Arf)/p53 pathway lesions but not p16(Ink4a, loss. J Clin Invest 118(1):51–63. , DOI 10.1172/JCI33320 Li Y,Wicha MS, Schwartz SJ, Sun D, 2011, Implications of cancer stem cell theory for cancer chemoprevention by natural dietary compounds. [Review]. J Nutr Biochem 22(9):799–806. https:// doi.org/10.1016/j.jnutbio.2010.11.001 Bao B, Ahmad A, Li Y, Azmi AS, Ali S, Banerjee S, Kong D, Sarkar FH, 2012, Targeting CSCs within the tumor microenvironment for cancer therapy: a potential role ofmesenchymal stem cells. Expert Opin Ther Targets 16(10):1041–1054. , DOI 10. 1517/14728222.2012.714774 Wang Z, LiY, Ahmad A, Azmi AS, Kong D, Banerjee S, Sarkar FH, 2010, Targeting miRNAs involved in cancer stem cell and EMT regulation: an emerging concept in overcoming drug resistance. [Article]. Drug Resist Updat 13(4–5):109–118. , DOI 10. 1016/j.drup.2010.07.001 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. [Review]. Cell 144(5):646–674. , DOI 10. 1016/j.cell.2011.02.013 Suman P, Mishra S, Chander H, 2018, High expression of FBP17 in invasive breast cancer cells promotes invadopodia formation. Med Oncol 35(5):71 Binayke A, Mishra S, Suman P, Das S, Chander H, 2019, Awakening the "guardian of genome": reactivation of mutant p53. Cancer Chemother Pharmacol 83(1):1–15. , DOI 10.1007/ s00280-018-3701-x Chander H, Truesdell P, Meens J, Craig AW, 2013, Transducer of Cdc42-dependent actin assembly promotes breast cancer invasion and metastasis. Oncogene 32(25):3080–3090 Kremerskothen J, Plaas C, Buther K, Finger I, Veltel S, Matanis T, Liedtke T, Barnekow A, 2003, Characterization of KIBRA, a novel WW domain-containing protein. [Article]. Biochem Biophys Res Commun 300(4):862–867. , DOI 10.1016/S0006- 291X(02)02945-5 Hilton HN, Stanford PM, Harris J, Oakes SR, Kaplan W, Daly RJ, Ormandy CJ, 2008, KIBRA interacts with discoidin domain receptor 1 to modulate collagen-induced signalling. [Article]. Biochim Biophys Acta, Mol Cell Res 1783(3):383–393. , DOI 10. 1016/j.bbamcr.2007.12.007 Schneider A, Huentelman M, Kremerskothen J, Duning K, Spoelgen R, Nikolich K, 2010, KIBRA: a new gateway to learning andmemory? [Review]. Front Aging Neurosci 2(4). , DOI 10.3389/neuro.24.004.2010 Yoshihama Y, Chida K, Ohno S, 2012, The KIBRA-aPKC connection. Commun Integr Biol 5(2):146–151. , DOI 10.4161/ cib.18849 Duning K, Schurek E-M, Schluter M, Bayer M, Reinhardt H-C, Schwab A, Schaefer L, Benzing T, Schermer B, Saleem MA, Huber TB, Bachmann S, Kremerskothen J, Weide T, Pavenstadt H, 2008, KIBRA modulates directional migration of podocytes. J Am Soc Nephrol 19(10):1891–1903. , DOI 10.1681/asn. 2007080916 Herb A, Wisden W, Catania MV, Marechal D, Dresse A, Seeburg PH, 1996, Prominent dendritic localization in forebrain neurons of a novel mRNA and its product, dendrin. [Article]. Mol Cell Neurosci 8(5):367–374. , DOI 10.1006/mcne.1996.0594 Neuner-Jehle M, Denizot JP, Borbely AA, Mallet J, 1996, Characterization and sleep deprivation-induced expression modulation of dendrin, a novel dendritic protein in rat brain neurons. J Neurosci Res 46(2):138–151. , DOI 10.1002/(SICI)1097- 4547(19961015)46:2<138::AID-JNR2>3.0.CO;2-I Rayala SK, den Hollander P, Manavathi B, Talukder AH, Song C, Peng S, Barnekow A, Kremerskothen J, Kumar R, 2006, Essential role of KIBRA in co-activator function of dynein light chain 1 in mammalian cells. J Biol Chem 281(28):19092–19099. https://doi. org/10.1074/jbc.M600021200 Ji M, Yang S, Chen Y, Xiao L, Zhang L, Dong J, 2012, Phosphoregulation of KIBRA by CDK1 and CDC14 phosphatase controls cell-cycle progression. Biochem J 447(1):93–102. , DOI 10.1042/bj20120751 Xiao L, Chen Y, Ji M, Volle DJ, Lewis RE, Tsai M-Y, Dong J, 2011, KIBRA protein phosphorylation is regulated by mitotic kinase Aurora and protein phosphatase 1. J Biol Chem 286(42): 36304–36315. , DOI 10.1074/jbc.M111.246850 Yang S, JiM, Zhang L, Chen Y,Wennmann DO, Kremerskothen J, Dong J, 2014, Phosphorylation of KIBRA by the extracellular signal-regulated kinase, ERK)–ribosomal S6 kinase, RSK, cascade modulates cell proliferation and migration. Cell Signal 26(2):343– 351. , DOI 10.1016/j.cellsig.2013.11.012 Mavuluri J, Beesetti S, Surabhi R, Kremerskothen J, Venkatraman G, Rayala SK, 2016, Phosphorylation dependent regulation of DNA damage response of adaptor protein KIBRA in cancer cells. Mol Cell Biol 36(9):1354–1365. , DOI 10.1128/MCB. 01004-15 Dobrosotskaya I, Guy RK, James GL, 1997, MAGI-1, a membrane-associated guanylate kinase with a unique arrangement of protein-protein interaction domains. [Article]. J Biol Chem 272(50):31589–31597. , DOI 10.1074/jbc.272.50.31589 Buther K, Plaas C, Barnekow A, Kremerskothen J, 2004, KIBRA is a novel substrate for protein kinase Cζ. [Article]. Biochem Biophys Res Commun 317(3):703–707. , DOI 10.1016/j. bbrc.2004.03.107 Rizo J, Sudhof TC, 1998, C2-domains, structure and function of a universal Ca2+−binding domain. J Biol Chem 273(26):15879– 15882. , DOI 10.1074/jbc.273.26.15879 Fanning AS, Anderson JM, 1999, PDZ domains: fundamental building blocks in the organization of protein complexes at the plasma membrane. J Clin Investig 103(6):767–772 Kremerskothen J, Plaas C, Kindler S, Frotscher M, Barnekow A, 2005, Synaptopodin, a molecule involved in the formation of the dendritic spine apparatus, is a dual actin/α-actinin binding protein. J Neurochem 92(3):597–606. , DOI 10.1111/j.1471-4159. 2004.02888.x Kremerskothen J, Kindler S, Finger I, Veltel S, Barnekow A, 2006, Postsynaptic recruitment of Dendrin depends on both dendritic mRNA transport and synaptic anchoring. J Neurochem 96(6): 1659–1666. , DOI 10.1111/j.1471-4159.2006.03679.x Lauriat TL, Dracheva S, Kremerskothen J, Duning K, Haroutunian V, Buxbaum JD, Hyde TM, Kleinman JE, AlisonMcInnes L, 2006, Characterization of KIAA0513, a novel signaling molecule that interacts with modulators of neuroplasticity, apoptosis, and the cytoskeleton. Brain Res 1121(1):1–11. , DOI 10.1016/j. brainres.2006.08.099 Shin K, Straight S, Margolis B, 2005, PATJ regulates tight junction formation and polarity in mammalian epithelial cells. J Cell Biol 168(5):705–711. , DOI 10.1083/jcb.200408064 Shin K, Wang Q, Margolis B, 2007, PATJ regulates directional migration of mammalian epithelial cells. EMBO Rep 8(2):158– 164. , DOI 10.1038/sj.embor.7400890 Traer CJ, Rutherford AC, Palmer KJ,Wassmer T, Oakley J,Attar N, et al., 2007, SNX4 coordinates endosomal sorting of TfnR with dynein-mediated transport into the endocytic recycling compartment. Nat Cell Biol 9(12):1370. , DOI 10.1038/ncb1656 Weigelt B, Peterse JL, Van't Veer LJ, 2005, Breast cancer metastasis: markers and models. Nat Rev Cancer 5(8):591–602 Minn AJ, Gupta GP, Siegel PM, Bos PD, ShuW, Giri DD, Viale A, Olshen AB, Gerald WL, Massague J, 2005, Genes that mediate breast cancer metastasis to lung. Nature 436(7050):518–524 Bos PD, Zhang XH-F, Nadal C, Shu W, Gomis RR, Nguyen DX, Minn AJ, van de Vijver MJ, Gerald WL, Foekens JA, Massague J, 2009, Genes that mediate breast cancer metastasis to the brain. Nature 459(7249):1005–1009 Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordon- Cardo C, Guise TA, Massague J, 2003, A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3(6):537– 549 Hilton HN, Stanford PM, Harris J, Oakes SR, Kaplan W, Daly RJ, Ormandy CJ, 2008, KIBRA interacts with discoidin domain receptor 1 to modulate collagen-induced signalling. Biochimica et Biophysica Acta, BBA)-Mol Cell Res 1783(3):383–393. https:// doi.org/10.1016/j.bbamcr.2007.12.007 Duning K, Schurek E-M, Schluter M, Bayer M, Reinhardt H-C, Schwab A, Schaefer L, Benzing T, Schermer B, Saleem MA, Huber TB, Bachmann S, Kremerskothen J, Weide T, Pavenstadt H, 2008, KIBRA modulates directional migration of podocytes. J Am Soc Nephrol: JASN 19(10):1891–1903. , DOI 10. 1681/ASN.2007080916 Barker KT, Martindale JE, Mitchell PJ, Kamalati T, Page MJ, Phippard DJ et al, 1995, Expression patterns of the novel receptor-like tyrosine kinase, DDR, in human breast tumours. [Article]. Oncogene 10(3):569–575 Vogel WF, Aszodi A, Alves F, Pawson T, 2001, Discoidin domain receptor 1 tyrosine kinase has an essential role in mammary gland development. [Article]. Mol Cell Biol 21(8):2906–2917. https:// doi.org/10.1128/MCB.21.8.2906-2917.2001 Fata JE, Werb Z, Bissell MJ, 2004, Regulation of mammary gland branchingmorphogenesis by the extracellular matrix and its remodeling enzymes. [Review]. Breast Cancer Res 6(1):1–11 Faraci-Orf E, McFadden C, Vogel WF, 2006, DDR1 signaling is essential to sustain Stat5 function during lactogenesis. J Cell Biochem 97(1):109–121. , DOI 10.1002/jcb.20618 Sun R, Gao P, Chen L, Ma D, Wang J, Oppenheim JJ, Zhang N, 2005, Protein kinase C ζ is required for epidermal growth factor– induced chemotaxis of human breast Cancer cells. Cancer Res 65(4):1433–1441. , DOI 10.1158/0008-5472.can-04-1163 Wu J, Zhang B, Wu M, Li H, Niu R, Ying G, Zhang N, 2010, Screening of a PKC ζ-specific kinase inhibitor PKCzI257.3 which inhibits EGF-induced breast cancer cell chemotaxis. [journal article]. Investig New Drugs 28(3):268–275. , DOI 10.1007/ s10637-009-9242-8 Zhang F, Zhang X, Li M, Chen P, Zhang B, Guo H, CaoW,Wei X, Cao X, Hao X, Zhang N, 2010, mTOR complex component Rictor interacts with PKCζ and regulates Cancer cell metastasis. Cancer Res 70(22):9360–9370. , DOI 10.1158/0008-5472.can-10- 0207 Urtreger AJ, Grossoni, VC, Falbo, KB, Kazanietz, MG, & Bal de Kier Joffe, ED, 2005, Atypical protein kinase C-ζ modulates clonogenicity, motility, and secretion of proteolytic enzymes in murine mammary cells.Molecular Carcinogenesis: Published in cooperation with the University of Texas MD Anderson Cancer Center 42(1):29–39. , DOI 10.1002/mc.20066 Curat CA, Vogel WF, 2002, Discoidin domain receptor 1 controls growth and adhesion of mesangial cells. [Article]. J Am Soc Nephrol 13(11):2648–2656. , DOI 10.1097/01.ASN. 0000032419.13208.0C Xie J, Haslam SZ, 1997, Extracellular matrix regulates ovarian hormone-dependent proliferation of mouse mammary epithelial cells. [Article]. Endocrinology 138(6):2466–2473. , DOI 10.1210/en.138.6.2466 Paul A, Danley M, Saha B, Tawfik O, Paul S, 2015, PKCζ promotes breast Cancer invasion by regulating expression of Ecadherin and zonula Occludens-1, ZO-1, via NFκB-p65. Sci Rep 5:12520. , DOI 10.1038/srep12520 Pfister KK, Fisher EMC, Gibbons IR, Hays TS, Holzbaur ELF, McIntosh JR, Porter ME, Schroer TA, Vaughan KT, Witman GB, King SM, Vallee RB, 2005, Cytoplasmic dynein nomenclature. J Cell Biol 171(3):411–413. , DOI 10.1083/jcb.200508078 Rayala SK, den Hollander P, Balasenthil S, Yang Z, Broaddus RR, Kumar R, 2005, Functional regulation of oestrogen receptor pathway by the dynein light chain 1. EMBO Rep 6(6):538–544. https:// doi.org/10.1038/sj.embor.7400417 Daniels TR, Delgado T, Rodriguez JA, Helguera G, Penichet ML, 2006, The transferrin receptor part I: biology and targeting with cytotoxic antibodies for the treatment of cancer. Clin Immunol 121(2):144–158. , DOI 10.1016/j.clim.2006.06.010 Shin K, Fogg VC, Margolis B, 2006, Tight junctions and cell polarity. Annu Rev Cell Dev Biol 22(1):207–235. , DOI 10. 1146/annurev.cellbio.22.010305.104219 Margolis B, Borg J-P, 2005, Apicobasal polarity complexes. J Cell Sci 118(22):5157–5159. , DOI 10.1242/jcs.02597 Knight JF, Sung VYC, Kuzmin E, Couzens AL, de Verteuil DA, Ratcliffe CDH, Coelho PP, Johnson RM, Samavarchi-Tehrani P, Gruosso T, Smith HW, Lee W, Saleh SM, Zuo D, Zhao H, Guiot MC, Davis RR, Gregg JP, Moraes C, Gingras AC, Park M, 2018, KIBRA, WWC1, is a metastasis suppressor gene affected by chromosome 5q loss in triple-negative breast Cancer. Cell Rep 22(12): 3191–3205. , DOI 10.1016/j.celrep.2018.02.095 Arivazhagan L, Surabhi RP, Kanakarajan A, Sundaram S, Pitani RS, et al., 2017, KIBRA attains oncogenic activity by repressing RASSF1A. Br J Cancer 117(4):553. , DOI 10.1038/bjc. 2017.192 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 627 EP 634 DI 10.1007/s12253-019-00660-x PG 8 ER PT J AU Assumpcao, P Khayat, A Araujo, T Barra, W Ishak, G Cruz, A Santos, S Santos, Demachki, S Assumpcao, P Calcagno, D Santos, N Assumpcao, M Moreira, F Santos, A Assumpcao, C Riggins, G Burbano, R AF Assumpcao, Paulo Khayat, Andre Araujo, Taissa Barra, Williams Ishak, Geraldo Cruz, Aline Santos, Sidney Santos, Andrea Demachki, Samia Assumpcao, Paula Calcagno, Danielle Santos, Ney Assumpcao, Monica Moreira, Fabiano Santos, Andre Assumpcao, Carolina Riggins, Gregory Burbano, Rommel TI The Small Bowel Cancer Incidence Enigma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Small bowel cancer; Low incidence; DNA repair; Enigma ID Small bowel cancer; Low incidence; DNA repair; Enigma AB Although the small bowel is a vast organ with a highly proliferative epithelium, the incidence of small bowel cancers is surprisingly low. Many factors could be involved in this unexpected cancer incidence, including difficult access to the exploration of the small bowel mucosa, which might lead to missed diagnoses of non-obstructive and non-bleeding small tumours. Moreover, possible factors that influence the low incidence include more efficient machinery of DNA replication and DNA repair enzymes, peculiarities in microbiota components, competence of the immune system, and the speed of intestinal transit. Importantly, the answer for the enigmatic risk of driver mutations caused by replication errors may be hidden in the small bowel, which is an obscure part of digestive tract that is usually inaccessible by endoscopic or colonoscopic conventional investigations. These observations warrant the necessity of an urgent exploration of small bowel features, including the evaluation of DNA replication controls and expression of DNA repair genes, in order to shed light on these obscure events. C1 [Assumpcao, Paulo] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Khayat, Andre] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Araujo, Taissa] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Barra, Williams] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Ishak, Geraldo] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Cruz, Aline] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Santos, Sidney] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Santos, Andrea] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Demachki, Samia] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Assumpcao, Paula] Universidade Federal do Para, Programa de Pos-Graduacao em Genetica e Biologia Molecular, 66075-110 Belem, Brazil. [Calcagno, Danielle] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Santos, Ney] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Assumpcao, Monica] Hospital Universitario Joao de Barros Barreto, Servico de Endoscopia Digestiva, 66073-000 Belem, Brazil. [Moreira, Fabiano] Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. [Santos, Andre] Universidade Federal do Para, Programa de Pos-Graduacao em Genetica e Biologia Molecular, 66075-110 Belem, Brazil. [Assumpcao, Carolina] Hospital Alemao Oswaldo Cruz, Servico de Cirurgia Oncologica, 01327-001 Sao Paulo, Brazil. [Riggins, Gregory] Johns Hopkins Hospital, Department of Neurosurgery, 21287 Baltimore, MD, USA. [Burbano, Rommel] Hospital Ophir Loyola, Laboratorio de Biologia Molecular, 66060-281 Belem, Brazil. RP Assumpcao, P (reprint author), Universidade Federal do Para, Nucleo de Pesquisas em Oncologia, 66073-000 Belem, Brazil. EM assumpcaopp@gmail.com CR Kato M, Kimura K, Hirakawa A, Kobayashi Y, Ishida R, Kamihira O, Majima T, Funahashi Y, Sassa N, Matsukawa Y, Hattori R, Gotoh M, Tsuzuki T, 2018, Prognostic parameter for high risk prostate cancer patients at initial presentation. Prostate 78:11–16. , DOI 10.1002/pros.23438 Sun YS, Zhao Z, Yang ZN, Xu F, Lu HJ, Zhu ZY, Shi W, Jiang J, Yao PP, Zhu HP, 2017, Risk factors and preventions of breast Cancer. Int J Biol Sci 13:1387–1397. , DOI 10.7150/ijbs. 21635 Khan N, Afaq F, Mukhtar H, 2010, Lifestyle as risk factor for cancer: evidence from human studies. Cancer Lett 293:133–143. , DOI 10.1016/j.canlet.2009.12.013 Tomasetti C, Li L, Vogelstein B, 2017, Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention. Science 355: 1330–1334. , DOI 10.1126/science.aaf9011 Eun K, Ham SW, Kim H, 2017, Cancer stem cell heterogeneity: origin and new perspectives on CSC targeting. BMB Rep 50:117– 125 Sell S, 2010, On the stem cell origin of cancer. Am J Pathol 176: 2584–2494. , DOI 10.2353/ajpath.2010.091064 Vassilev A, DePamphilis ML, 2017, Links between DNA replication, stem cells and Cancer. Genes, Basel, 8:45. , DOI 10. 3390/genes8020045 Kunkel TA, Bebenek K, 2000, DNA replication fidelity. Annu Rev Biochem 69:497–529. , DOI 10.1146/annurev.biochem. 69.1.497 Andrianova MA, Bazykin GA, Nikolaev SI, Seplyarskiy VB, 2017, Human mismatch repair system balances mutation rates between strands by removing more mismatches from the lagging strand. Genome Res 27:1336–1343. , DOI 10.1101/gr. 219915.116 Hass CS, Gakhar L,Wold MS(2010, Functional characterization of a cancer causing mutation in human replication protein a. Mol Cancer Res 8:1017–1026. , DOI 10.1158/1541-7786. MCR-10-0161 Taylor EM, Lindsay HD, 2016, DNA replication stress and cancer: cause or cure? Future Oncol 12:221–237. , DOI 10.2217/ fon.15.292 Torgovnick A, Schumacher B, 2015, DNA repair mechanisms in cancer development and therapy. Front Genet 6(157). https://doi. org/10.3389/fgene.2015.00157 Barbari SR, Shcherbakova PV, 2017, Replicative DNA polymerase defects in human cancers: consequences, mechanisms, and implications for therapy. DNA Repair, Amst, 56:16–25. https://doi. org/10.1016/j.dnarep.2017.06.003 Harris RS, 2013, Cancer mutation signatures, DNA damage mechanisms, and potential clinical implications. Genome Med 5:87. , DOI 10.1186/gm490 Rycaj K, Tang DG, 2015, Cell-of-origin of cancer versus cancer stem cells: assays and interpretations. Cancer Res 75:4003–4011. , DOI 10.1158/0008-5472.CAN-15-0798 Kreso A, Dick JE, 2014, Evolution of the cancer stem cell model. Cell Stem Cell 14:275–291. , DOI 10.1016/j.stem.2014. 02.006 Andersson-Rolf A, Zilbauer M, Koo BK, Clevers H, 2017, Stem cells in repair of gastrointestinal epithelia. Physiology, Bethesda, 32:278–289. , DOI 10.1152/physiol.00005.2017 Sarosiek T, Stelmaszuk M, 2018, Small intestine neoplasms. Pol Merkur Lekarski 44:45–48 Taylor S, Lobo AJ, 2016, Diagnosis and treatment of inflammatory bowel disease. Practitioner 260:19–23 Cloyd JM, George E, Visser BC, 2016, Duodenal adenocarcinoma: Advances in diagnosis and surgical management. World J Gastrointest Surg 8:212–221. , DOI 10.4240/wjgs.v8.i3. 212 Bhandari A, Woodhouse M, Gupta S, 2017, Colorectal cancer is a leading cause of cancer incidence andmortality among adults younger than 50 years in the USA: a SEER-based analysis with comparison to other young-onset cancers. J Investig Med 65:311–315. , DOI 10.1136/jim-2016-000229 Marley AR, Nan H, 2016, Epidemiology of colorectal cancer. Int J Mol Epidemiol Genet 7:105–114 Talseth-Palmer BA, 2017, The genetic basis of colonic adenomatous polyposis syndromes. Hered Cancer Clin Pract 15(5):5. https:// doi.org/10.1186/s13053-017-0065-x Leoz ML, Carballal S, Moreira L, Ocana T, Balaguer F, 2015, The genetic basis of familial adenomatous polyposis and its implications for clinical practice and risk management. Appl Clin Genet 8:95– 107. , DOI 10.2147/TACG.S51484 Jackstadt R, Sansom OJ, 2016, Mousemodels of intestinal cancer. J Pathol 238:141–151. , DOI 10.1002/path.4645 Kopacova M, Rejchrt S, Bures J, Tacheci I, 2013, Small intestinal Tumours. Gastroenterol Res Pract 2013(702536):1–7. https://doi. org/10.1155/2013/702536 Han YF, Zhang QZ, 1995, An analysis of 36 cases of primary tumor of the small bowel. Tumor 15:406–407 Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2013, GLOBOCAN 2012 v1.0, Cancer incidence and mortality worldwide: IARC CancerBase no. 11 [internet]. Lyon, France: International Agency for Research on. Cancer. Available from: http://globocan.iarc.fr. Accessed 11 February 2019 Lauren P, 1965, The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand 64: 31–49 Correa P, Piazuelo MB, 2012, The gastric precancerous cascade. J Dig Dis 13:2–9 Fu DG, 2015, Epigenetic alterations in gastric cancer, review). Mol Med Rep 12:3223–3230. , DOI 10.3892/mmr.2015.3816 Yoon H, Kim N, 2015, Diagnosis and management of high risk group for gastric cancer. Gut Liver 9:5–17. , DOI 10.5009/ gnl14118 Gonzalez CA, Sanz-Anquela JM, Companioni O, Bonet C, Berdasco M, Lopez C, Mendoza J, Martin-Arranz MD, Rey E, Poves E, Espinosa L, Barrio J, Torres MA, Cuatrecasas M, Elizalde I, Bujanda L, Garmendia M, Ferrandez A, Munoz G, Andreu V, Paules MJ, Lario S, Ramirez MJ, Study group, Gisbert JP, 2016, Incomplete type of intestinal metaplasia has the highest risk to progress to gastric cancer: results of the Spanish follow-up multicenter study. J Gastroenterol Hepatol 31:953–958. https://doi. org/10.1111/jgh.13249 Gomez JM,WangAY, 2014, Gastric intestinal metaplasia and early gastric cancer in the west: a changing paradigm. Gastroenterol Hepatol, N Y, 10:369–378 Correa P, Piazuelo MB, 2010, Wilson KT, 2010, pathology of gastric intestinal metaplasia: clinical implications. Am J Gastroenterol 105:493–498. , DOI 10.1038/ajg.2009.728 Park YH, Kim N, 2015, Review of atrophic gastritis and intestinal metaplasia as a premalignant lesion of gastric cancer. J Cancer Prev 20:25–40. , DOI 10.15430/JCP.2015.20.1.25 Law R, Varayil JE, WongKeeSong LM, Fidler J, Fletcher JG, Barlow J, Alexander J, Rajan E, Hansel S, Becker B, Larson JJ, Enders FT, Bruining DH, Coelho-Prabhu N, 2017, Assessment of multi-modality evaluations of obscure gastrointestinal bleeding. World J Gastroenterol 23:614–621. , DOI 10.3748/wjg. v23.i4.614 Kim BS, Li BT, Engel A, Samra JS, Clarke S, Norton ID, Li AE, 2014, Diagnosis of gastrointestinal bleeding: a practical guide for clinicians. World J Gastrointest Pathophysiol 5:467–478. https:// doi.org/10.4291/wjgp.v5.i4.467 Strik C, Stommel MW, Schipper LJ, van Goor H, Ten Broek RP, 2016, Risk factors for future repeat abdominal surgery. Langenbeck's Arch Surg 401:829–837. , DOI 10.1007/ s00423-016-1414-3 Ferguson HJ, Ferguson CI, Speakman J, Ismail T, 2015, Management of intestinal obstruction in advanced malignancy. Ann Med Surg, Lond, 4:264–270. , DOI 10.1016/j.amsu. 2015.07.018 Pironi L, Arends J, Baxter J, Bozzetti F, Pelaez RB, Cuerda C, Forbes A, S8 G, Gillanders L, Holst M, Jeppesen PB, Joly F, Kelly D, Klek S, Irtun O, Olde Damink SW, Panisic M, Rasmussen HH, Staun M, Szczepanek K,Van Gossum A,Wanten G, Schneider SM, Shaffer J Home Artificial Nutrition & Chronic Intestinal Failure; acute intestinal failure special interest groups of ESPEN, 2015, ESPEN endorsed recommendations. Definition and classification of intestinal failure in adults. Clin Nutr 34:171–180. https://doi. org/10.1016/j.clnu.2014.08.017 Thomas S, Izard J,Walsh E, Batich K, Chongsathidkiet P, Clarke G, Sela DA, Muller AJ, Mullin JM, Albert K, Gilligan JP, DiGuilio K, Dilbarova R, Alexander W, Prendergast GC, 2017, The host microbiome regulates and maintains human health: a primer and perspective for non-microbiologists. Cancer Res 77:1783–1812. , DOI 10.1158/0008-5472.CAN-16-2929 Shreiner AB, Kao JY, Young VB, 2015, The gut microbiome in health and in disease. Curr Opin Gastroenterol 31:69–75. https:// doi.org/10.1097/MOG.0000000000000139 Francescone R, Hou V, Grivennikov SI, 2014, Microbiome, inflammation, and cancer. Cancer J 20:181–189. , DOI 10. 1097/PPO.0000000000000048 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 635 EP 639 DI 10.1007/s12253-019-00682-5 PG 5 ER PT J AU Zalatnai, A Kis-Orha, V AF Zalatnai, Attila Kis-Orha, Viktoria TI Solid-pseudopapillary Neoplasms of the Pancreas is still an Enigma: a Clinicopathological Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Pancreas; Solid-pseudopapillary neoplasm; Histogenesis; Review ID Pancreas; Solid-pseudopapillary neoplasm; Histogenesis; Review AB The solid-pseudopapillary neoplasm of the pancreas is a rare but enigmatic entity occurring mainly in young women. Since the first description by V. Frantz in 1959 the terminology of this tumor has continuously changed but it has remained simply descriptive, because the exact histogenesis is still obscure. Although in majority of cases the survival is excellent, nevertheless, the expected prognosis is not exactly predictable. In this review the authors aim to summarize its clinicopathological features, the expected biological behavior, the molecular alterations, the immune phenotype and discuss the putative histogenesis. From diagnostic point of view, the salient histological characteristic findings are analyzed that would help to differentiate it from other, look-alike pancreatic tumors, and suggestions are made about the desirable content of the histological report. C1 [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Kis-Orha, Viktoria] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. RP Zalatnai, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM zalatnai@korb1.sote.hu CR Frantz VK, 1959, Tumors of the pancreas. Armed forces Institute of Pathology, Washington, DC Yoon DY, Hines OJ, Bilchik AJ, Lewin K, Cortina G, Reber HA, 2001, Solid and papillary epithelial neoplasms of the pancreas: aggressive resection for cure. Am Surg 67:1195–1199 Cai YQ, Xie SM, Ran X, Wang X, Mai G, Liu XB, 2014, Solid pseudopapillary tumor of the pancreas in male patients: report of 16 cases. World J Gastroenterol 20:6939–6945 Shorter NA, Glick RD, Klimstra DS, Brennan MF, Laquaglia MP, 2002, Malignant pancreatic tumors in childhood and adolescence: thememorial Sloan-Kettering experience, 1967 to present. J Pediatr Surg 37:887–892 Wang WB, Zhang TP, Sun MQ, Peng Z, Chen G, Zhao YP, 2014, Solid pseudopapillary tumor of the pancreas with liver metastasis: clinical features and management. Eur J Surg Oncol 40:1572–1577 Lieber MR, Lack EE, Roberts JR Jr, Merino MJ, Patterson K, Restrepo C, Solomon D, Chandra R, Triche TJ, 1987, Solid and papillary epithelial neoplasm of the pancreas. An ultrastructural and immunocytochemical study of six cases. Am J Surg Pathol 11:85– 93 Uppin SG, HuiM, Thumma V, Challa S, UppinMS, Bheerappa N, Sastry RA, Paul TR, Prayaga AK, 2015, Solid-pseudopapillary neoplasm of the pancreas: a clinicopathological and immunohistochemical study of 33 cases from a single institution in southern India. Indian J Pathol Microbiol 58:163–169 Nguyen NQ, Johns AL, Gill AJ, Ring N, Chang DK, Clarkson A, Merrett ND, Kench JG, Colvin EK, Scarlett CJ, Biankin AV, 2011, Clinical and immunohistochemical features of 34 solid pseudopapillary tumors of the pancreas. J Gastroenterol Hepatol 26:267–274 Tajima Y, Kohara N,Maeda J, Inoue K, Kitasato A, Natsuda K, Irie J, Adachi T, Kuroki T, Eguchi S, Kanematsu T, 2012, Peritoneal and nodal recurrence 7 years after the excision of a ruptured solid pseudopapillary neoplasm of the pancreas: report of a case. Surg Today 42:776–780 Huang SC, Wu TH, Chen CC, Chen TC, 2013, Spontaneous rupture of solid pseudopapillary neoplasm of the pancreas during pregnancy. Obstet Gynecol 121(2 Pt 2 Suppl 1):486–488 FukunagaM(2001, Pseudopapillary solid cystic tumor arising from an extrapancreatic site. Arch Pathol Lab Med 125:1368–1371 Zhu H, Xia D, Wang B, Meng H, 2013, Extrapancreatic solid pseudopapillary neoplasm: report of a case of primary retroperitoneal origin and review of the literature. Oncol Lett 5:1501–1504 Wu H, Huang YF, Liu XH, Xu MH, 2017, Extrapancreatic solid pseudopapillary neoplasm followed by multiple metastases: case report. World J Gastrointest Oncol 9:497–501 Li HX, Zhang Y, Du ZG, Tang F, Qi XQ, Yin B, Jiang YJ, Yang F, Subedi S, 2013, Multi-centric solid-pseudopapillary neoplasm of the pancreas. Med Oncol 30:330 Hirabayashi K, Kurokawa S, Maruno A, YamadaM, Kawaguchi Y, Nakagohri T, Mine T, Sugiyama T, Tajiri T, Nakamura N, 2015, Sex differences in immunohistochemical expression and capillary density in pancreatic solid pseudopapillary neoplasm. Ann Diagn Pathol 19:45–49 Chen C, Jing W, Gulati P, Vargas H, French SW, 2004, Melanocytic differentiation in a solid pseudopapillary tumor of the pancreas. J Gastroenterol 39:579–583 Daum O, Sima R, Mukensnabl P, Vanecek T, BrouckovaM, Benes Z, Michal M, 2005, Pigmented solid-pseudopapillary neoplasm of the pancreas. Pathol Int 55:280–284 Kempski HM, Austin N, Chatters SJ, Toomey SM, Chalker J, Anderson J, Sebire NJ, 2006, Previously unidentified complex cytogenetic changes found in a pediatric case of solidpseudopapillary neoplasm of the pancreas. Cancer Genet Cytogenet 164:54–60 Tiemann K, Heitling U, Kosmahl M, Kloppel G, 2007, Solid pseudopapillary neoplasms of the pancreas show an interruption of the Wnt-signaling pathway and express gene products of 11q. Mod Pathol 20:955–960 Park M, Kim M, Hwang D, Park M, Kim WK, Kim SK, Shin J, Park ES, Kang CM, Paik YK, Kim H, 2014, Characterization of gene expression and activated signaling pathways in solidpseudopapillary neoplasm of pancreas. Mod Pathol 27:580–593 Muller-Hocker J, Zietz CH, Sendelhofert A, 2001, Deregulated expression of cell cycle-associated proteins in solid pseudopapillary tumor of the pancreas. Mod Pathol 14:47–53 Abraham SC, Klimstra DS, Wilentz RE, Yeo CJ, Conlon K, Brennan M, Cameron JL, Wu TT, Hruban RH, 2002, Solidpseudopapillary tumors of the pancreas are genetically distinct from pancreatic ductal adenocarcinomas and almost always harbor betacatenin mutations. Am J Pathol 160:1361–1369 Zhang Y, Han X,Wu H, Zhou Y, 2017, Bioinformatics analysis of transcription profiling of solid pseudopapillary neoplasm of the pancreas. Mol Med Rep 16:1635–1642 Park M, Lim JS, Lee HJ, Na K, Lee MJ, Kang CM, Paik YK, Kim H, 2015, Distinct protein expression profiles of solid- Pseudopapillary neoplasms of the pancreas. J Proteome Res 14: 3007–3014 Xu Y, Zhao G, Pu N, Nuerxiati A, Ji Y, Zhang L, Rong Y, Lou W, Wang D, Kuang T, Xu X, Wu W, 2017, One hundred twenty-one resected solid Pseudopapillary tumors of the pancreas: an 8-year single-institution experience at Zhongshan hospital, Shanghai, China. Pancreas 46:1023–1028 Yu PF, Hu ZH, Wang XB, Guo JM, Cheng XD, Zhang YL, Xu Q, 2010, Solid pseudopapillary tumor of the pancreas: a review of 553 cases in Chinese literature. World J Gastroenterol 16:1209– 1214 Yu P, Cheng X, Du Y, Yang L, Xu Z, YinW, Zhong Z,Wang X, Xu H, Hu C, 2015, Solid Pseudopapillary neoplasms of the pancreas: a 19-year multicenter experience in China. J Gastrointest Surg 19: 1433–1440 Kim CW, Han DJ, Kim J, Kim YH, Park JB, Kim SC, 2011, Solid pseudopapillary tumor of the pancreas: can malignancy be predicted? Surgery 149:625–634 Yang F, Yu X, Bao Y, Du Z, Jin C, Fu D, 2016, Prognostic value of Ki-67 in solid pseudopapillary tumor of the pancreas: Huashan experience and systematic review of the literature. Surgery 159: 1023–1031 Hanada K, Kurihara K, Itoi T, Katanuma A, Sasaki T, Hara K, Nakamura M, Kimura W, Suzuki Y, Sugiyama M, Ohike N, Fukushima N, Shimizu M, Ishigami K, Gabata T, Okazaki K, 2018, Clinical and pathological features of solid pseudopapillary neoplasms of the pancreas: a nationwidemulticenter study in Japan. Pancreas 47:1019–1026 Serrano PE, Serra S, Al-Ali H, Gallinger S, Greig PD, McGilvray ID, Moulton CA,Wei AC, Cleary SP, 2014, Risk factors associated with recurrence in patients with solid pseudopapillary tumors of the pancreas. JOP 15:561–568 Meng Z, Cao M, Wang F, Zhang Y, Wu H, 2018, Tumor location correlates with Clinicopathological features and prognosis of the solid Pseudopapillary neoplasm. Gastroenterol Res Pract 9023947. , DOI 10.1155/2018/9023947 Honore C, Goere D, Dartigues P, Burtin P, Dumont F, Elias D, 2012, Peritoneal carcinomatosis from solid pseudopapillary neoplasm, Frantz's tumour, of the pancreas treated with HIPEC. Anticancer Res 32:1069–1073 Gomez P, Yorke R, Ayala AG, Ro JY, 2012, Solid-pseudopapillary neoplasm of pancreas with long delayed liver metastasis. Ann Diagn Pathol 16:380–384 Kato T, Egawa N, Kamisawa T, Tu Y, Sanaka M, Sakaki N, Okamoto A, Bando N, Funata N, Isoyama T, 2002, A case of solid pseudopapillary neoplasm of the pancreas and tumor doubling time. Pancreatology 2:495–498 Estrella JS, Li L, Rashid A, Wang H, Katz MH, Fleming JB, Abbruzzese JL, Wang H, 2014, Solid pseudopapillary neoplasm of the pancreas: clinicopathologic and survival analyses of 64 cases from a single institution. Am J Surg Pathol 38:147–157 Ercelep O, Ozdemir N, Turan N, Topcu TO, UysalM, Tanriverdi O, Demirci U, Taskoylu BY, Urakci Z, Duran AO, Aksoy A, Menekse S, Ozcelik M, Gumus M, 2019, Retrospective evaluation of patients diagnosed with solid pseudopapillary neoplasms of the pancreas. Curr Probl Cancer 43:26–32 Prakash PS, Chan DYS, Madhavan K, 2018, The stomach: a rare site formetastatic solid pseudopapillary neoplasm of the pancreas. J Gastrointest Surg 22:759–760 Gao H, Gao Y, Yin L,Wang G,Wei J, Jiang K,Miao Y, 2018, Risk factors of the recurrences of pancreatic solid Pseudopapillary tumors: a systematic review and meta-analysis. J Cancer 9:1905– 1914 Irtan S, Galmiche-Rolland L, Elie C, Orbach D, Sauvanet A, Elias D, Guerin F, Coze C, Faure-Conter C, Becmeur F, Demarche M, Galifer RB, Galloy MA, Podevin G, Aubert D, Piolat C, De Lagausie P, Sarnacki S, 2016, Recurrence of solid Pseudopapillary neoplasms of the pancreas: results of a Nationwide study of risk factors and treatment modalities. Pediatr Blood Cancer 63:1515–1521 Song H, Dong M, Zhou J, ShengW, Zhong B, GaoW(2017, Solid Pseudopapillary neoplasm of the pancreas: Clinicopathologic feature, risk factors of malignancy, and survival analysis of 53 cases from a single center. Biomed Res Int 2017:5465261 Shimizu M, Matsumoto T, Hirokawa M, Monobe Y, Iwamoto S, Tsunoda T, Manabe T, 1999, Solid-pseudopapillary carcinoma of the pancreas. Pathol Int 49:231–234 Kim EK, Jang M, Park M, Kim H, 2017, LEF1, TFE3 and AR are putative diagnostic markers of solid pseudopapillary neoplasms. Oncotarget. 8:93404–93413 Geers C, Moulin P, Gigot JF, Weynand B, Deprez P, Rahier J, Sempoux C, 2006, Solid and pseudopapillary tumor of the pancreas–review and new insights into pathogenesis. Am J Surg Pathol 30:1243–1249 Neill KG, Saller J, Al Diffalha S, CentenoBA, MalafaMP, Coppola D, 2018, EGFR L861Q mutation in a metastatic solidpseudopapillary neoplasm of the pancreas. Cancer Genomics Proteomics 15:201–205 Liszka L, Mrowiec S, Pajak J, Kostrzab-Zdebel A, Lampe P, Kajor M, 2014, Limited usefulness of histopathological features in identification of a clinically aggressive solid-pseudopapillary neoplasm of the pancreas. Pol J Pathol 65:182–193 Kim JH, Lee JM, 2016, Clinicopathologic review of 31 cases of solid Pseudopapillary pancreatic tumors: can we use the scoring system of microscopic features for suggesting clinically malignant potential? Am Surg 82:308–313 KosmahlM, Seada LS, Janig U, Harms D, Kloppel G, 2000, Solidpseudopapillary tumor of the pancreas: its origin revisited. Virchows Arch 436:473–480 Park JY, Kim SG, Park J, 2014, Solid pseudopapillary tumor of the pancreas in children: 15-year experience at a single institution with assays using an immunohistochemical panel. Ann Surg Treat Res 86:130–135 Calvani J, Lopez P, Sarnacki S, Molina TJ, Gibault L, Fabre M, Scharfmann R, Capito C, Galmiche L, 2019, Solid pseudopapillary neoplasms of the pancreas do not express major pancreatic markers in pediatric patients. Hum Pathol 83:29–31 Notohara K, Hamazaki S, Tsukayama C, Nakamoto S, Kawabata K, Mizobuchi K, Sakamoto K, Okada S, 2000, Solidpseudopapillary tumor of the pancreas: immunohistochemical localization of neuroendocrine markers and CD10. Am J Surg Pathol 24:1361–1371 Li L, Li J, Hao C, Zhang C, Mu K, Wang Y, Zhang T, 2011, Immunohistochemical evaluation of solid pseudopapillary tumors of the pancreas: the expression pattern of CD99 is highly unique. Cancer Lett 310:9–14 Ohiwa K, Igarashi M, Nagasue N, Nagasaki M, Harada T, 1997, Solid and cystic tumor, SCT, of the pancreas in an adult man. HPB Surg 10:315–321 Harrison G, Hemmerich A, Guy C, Perkinson K, Fleming D, McCall S, Cardona D, Zhang X, 2017, Overexpression of SOX11 and TFE3 in solid-Pseudopapillary neoplasms of the pancreas. Am J Clin Pathol 149:67–75 Jiang Y, Xie J, Wang B, Mu Y, Liu P, 2018, TFE3 is a diagnostic marker for solid pseudopapillary neoplasms of the pancreas. Hum Pathol 81:166–175 Lawlor RT, Dapra V, Girolami I, Pea A, Pilati C, Nottegar A, Piccoli P, Parolini C, Sperandio N, Capelli P, Scarpa A, Luchini C, 2019, CD200 expression is a feature of solid pseudopapillary neoplasms of the pancreas. Virchows Arch 474:105–109 HavM, De Potter A, Ferdinande L,Van BockstalM, LemD, Eav S, Pattyn P, Praet M, Cuvelier C, Libbrecht L, 2011, Glypican-3 is a marker for solid pseudopapillary neoplasm of the pancreas. Histopathology. 59:1278–1279 Shen Y, Wang Z, Zhu J, Chen Y, Gu W, Liu Q, 2014, α- Methylacyl-CoA racemase, P504S, is a useful marker for the differential diagnosis of solid pseudopapillary neoplasm of the pancreas. Ann Diagn Pathol 18:146–150 Klimstra DS,Wenig BM, Heffess CS, 2000, Solid-pseudopapillary tumor of the pancreas: a typically cystic carcinoma of low malignant potential. Semin Diagn Pathol 17:66–80 Wrba F, Chott A, Ludvik B, Schratter M, Spona J, Reiner A, Scjernthaner G, Krisch K, 1988, Solid and cystic tumour of the pancreas; a hormonal-dependent neoplasm? Histopathology 12: 338–340 Ueda N, Nagakawa T, Ohta T, Nakamura T, Ueno K, Miyazaki I, Kurachi M, Konishi I, Hirono T, Takayanagi N, Sodani H, Kanno S, 1991, Clinicopathological studies on solid and cystic tumors of the pancreas. Gastroenterol Jpn 26:497–502 Hamoudi AB, Misugi K, Grosfeld JL, Reiner CB, 1970, Papillary epithelial neoplasms of pancreas in a child. Cancer 26:1126–1134 Schlosnagle DC, Campbell WG, 1981, The papillary and solid neoplasm of the pancreas. A report ot two cases with electron microscopy, one containing neurosecretory granules. Cancer 47: 2603–2610 Cao D, Maitra A, Saavedra JA, Klimstra DS, Adsay NV, Hruban RH, 2005, Expression of novel markers of pancreatic ductal adenocarcinoma in pancreatic nonductal neoplasms: additional evidence of different genetic pathways. Mod Pathol 18:752–761 Kallichandra N, Tsai S, Stabile BE, Buslon V, Delgado DL, French SW, 2006, Histogenesis of solid pseudopapillary tumor of the pancreas: the case for the centroacinar cell of origin. Exp Mol Pathol 81:101–107 Matsunou H, Konishi F, 1990, Papillary-cystic neoplasm of the pancreas. A clinicopathologic study concerning the tumor aging and malignancy of nine cases. Cancer 65:283–291 Sun CD, Lee WJ, Choi JS, Oh JT, Choi SH, 2005, Solidpseudopapillary tumours of the pancreas: 14 years experience. ANZ J Surg 75:684–689 Escobar MA Jr, McClellan JM, Thomas W, 2017, Solid pseudopapillary tumour, Frantz's tumour, of the pancreas in childhood: successful management of late liver metastases with sunitinib and chemoembolisation. BMJ Case Rep pii: bcr-2017-221906. , DOI 10.1136/bcr-2017-221906 Lagiewska B, Pacholczyk M, Lisik W, Cichocki A, Nawrocki G, Trzebicki J, Chmura A, 2013, Liver transplantation for nonresectable metastatic solid pseudopapillary pancreatic cancer. Ann Transplant 18:651–653 Wojciak M, Gozdowska J, Pacholczyk M, LisikW, Kosieradzki M, Cichocki A, Tronina O, DurlikM(2018, Liver transplantation for a metastatic pancreatic solid-Pseudopapillary tumor, Frantz tumor): a case report. Ann Transplant 23:520–523 Foo WC, Harrison G, Zhang X, 2017, Immunocytochemistry for SOX-11 and TFE3 as diagnostic markers for solid pseudopapillary neoplasms of the pancreas in FNA biopsies. Cancer Cytopathol 125:831–837 Comper F, Antonello D, Beghelli S, Gobbo S, Montagna L, Pederzoli P, Chilosi M, Scarpa A, 2009, Expression pattern of claudins 5 and 7 distinguishes solid-pseudopapillary from pancreatoblastoma, acinar cell and endocrine tumors of the pancreas. Am J Surg Pathol 33:768–774 Bouassida M, Mighri MM, Bacha D, Chtourou MF, Touinsi H, Azzouz MM, Sassi S, 2012, Solid pseudopapillary neoplasm of the pancreas in an old man: age does not matter. Pan Afr Med J 13:8 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 641 EP 649 DI 10.1007/s12253-019-00671-8 PG 9 ER PT J AU Ding, L Lu, Sh Li, Y AF Ding, Lei Lu, Shengdi Li, Yanli TI Regulation of PD-1/PD-L1 Pathway in Cancer by Noncoding RNAs SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE PD-1/PD-L1 pathway; Immune checkpoint blockade; Noncoding RNA; microRNA ID PD-1/PD-L1 pathway; Immune checkpoint blockade; Noncoding RNA; microRNA AB Immune checkpoint blockade has demonstrated significant anti-tumor immunity in an array of cancer types, yet the underlying regulatory mechanism of it is still obscure, and many problems remain to be solved. As an inhibitory costimulatory signal of T-cells, the programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) pathway can paralyze T-cells at the tumor site, enabling the immune escape of tumor cells. Although many antibodies targeting PD-1/PD-L1 have been developed to block their interaction for the treatment of cancer, the reduced response rate and resistance to the therapies call for further comprehension of this pathway in the tumor microenvironment. MicroRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are two main types of noncoding RNAs that play critical parts in the regulation of immune response in tumorigenesis, including the PD-1/ PD-L1 pathway. Here we summarize the most recent studies on the control of this pathway by noncoding RNAs in cancer and hopefully will offer new insights into immune checkpoint blockade therapies. C1 [Ding, Lei] Shanghai University, School of Life Science, Lab for Noncoding RNA & Cancer, 200444 Shanghai, China. [Lu, Shengdi] Shanghai Jiaotong University, Shanghai Sixth People’s Hospital, 200233 Shanghai, China. [Li, Yanli] Shanghai University, School of Life Science, Lab for Noncoding RNA & Cancer, 200444 Shanghai, China. RP Li, Y (reprint author), Shanghai University, School of Life Science, Lab for Noncoding RNA & Cancer, 200444 Shanghai, China. EM liyanli@shu.edu.cn CR Teng MWet al, 2008, Immune-mediated dormancy: an equilibrium with cancer. J Leukoc Biol 84(4):988–993 Schneider H et al, 2006, Reversal of the TCR stop signal by CTLA-4. Science 313(5795):1972–1975 Ishida Yet al, 1992, Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 11(11):3887–3895 Dong H et al, 2002, Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 8(8): 793–800 Dong H et al, 1999, B7-H1, a third member of the B7 family, costimulates T-cell proliferation and interleukin-10 secretion. Nat Med 5(12):1365–1369 Freeman GJ et al, 2000, Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 192(7):1027–1034 Zhang X et al, 2004, Structural and functional analysis of the costimulatory receptor programmed death-1. Immunity 20(3): 337–347 LesterhuisWJ, Steer H, Lake RA, 2011, PD-L2 is predominantly expressed by Th2 cells. Mol Immunol 49(1–2):1–3 Chen L, 2004, Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol 4(5):336–347 Sheppard KA et al, 2004, PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. FEBS Lett 574(1–3):37–41 Francisco LM, Sage PT, Sharpe AH, 2010, The PD-1 pathway in tolerance and autoimmunity. Immunol Rev 236:219–242 ZamaniMR et al, 2016, PD-1/PD-L and autoimmunity: a growing relationship. Cell Immunol 310:27–41 Sharpe AH et al, 2007, The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol 8(3):239–245 Butte MJ et al, 2007, Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity 27(1):111–122 Schadendorf D et al, 2015, Pooled analysis of Long-term survival data from phase II and phase III trials of Ipilimumab in Unresectable or metastatic melanoma. J Clin Oncol 33(17):1889–1894 Wolchok JD et al, 2013, Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 369(2):122–133 Younes RN et al, 2011, Chemotherapy beyond first-line in stage IV metastatic non-small cell lung cancer. Rev Assoc Med Bras, 1992, 57(6):686–691 ReckMet al, 2016, Pembrolizumab versus chemotherapy for PDL1- positive non-small-cell lung Cancer. N Engl J Med 375(19): 1823–1833 D'Alterio C et al, 2016, CXCR4-CXCL12-CXCR7, TLR2-TLR4, and PD-1/PD-L1 in colorectal cancer liver metastases from neoadjuvant-treated patients. Oncoimmunology 5(12):e1254313 Sasaki S et al, 2018, EBV-associated gastric cancer evades T-cell immunity by PD-1/PD-L1 interactions. Gastric Cancer Saito H et al, 2018, Highly activated PD-1/PD-L1 pathway in gastric Cancer with PD-L1 expression. Anticancer Res 38(1): 107–112 Liu S et al, 2017, PD-1/PD-L1 interaction up-regulates MDR1/Pgp expression in breast cancer cells via PI3K/AKT and MAPK/ERK pathways. Oncotarget 8(59):99901–99912 Shi W et al, 2018, Follicular helper T cells promote the effector functions of CD8(+, T cells via the provision of IL-21, which is downregulated due to PD-1/PD-L1-mediated suppression in colorectal cancer. Exp Cell Res 372(1):35–42 Motzer RJ et al, 2015, Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J Clin Oncol 33(13): 1430–1437 Sharma P, Allison JP, 2015, The future of immune checkpoint therapy. Science 348(6230):56–61 Schutz F et al, 2017, PD-1/PD-L1 pathway in breast Cancer. Oncol Res Treat 40(5):294–297 Ge Y et al, 2013, Blockade of PD-1/PD-L1 immune checkpoint during DC vaccination induces potent protective immunity against breast cancer in hu-SCID mice. Cancer Lett 336(2):253–259 Strickland KC et al, 2016, Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget 7(12):13587– 13598 Tan D, Sheng L, Yi QH, 2018, Correlation of PD-1/PD-L1 polymorphisms and expressions with clinicopathologic features and prognosis of ovarian cancer. Cancer Biomark 21(2):287–297 Powles T et al, 2014, MPDL3280A, anti-PD-L1, treatment leads to clinical activity in metastatic bladder cancer. Nature 515(7528): 558–562 Kwok G et al, 2016, Pembrolizumab, Keytruda). Hum Vaccin Immunother 12(11):2777–2789 Topalian SL et al, 2012, Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366(26):2443–2454 Hodi FS et al, 2010, Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363(8):711–723 Topalian SL et al, 2014, Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 32(10):1020–1030 Bray F et al, 2018, Global Cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin Barlesi F et al, 2014)Maintenance bevacizumab-pemetrexed after first-line cisplatin-pemetrexed-bevacizumab for advanced nonsquamous nonsmall-cell lung cancer: updated survival analysis of the AVAPERL, MO22089, randomized phase III trial. Ann Oncol 25(5):1044–1052 Ettinger DS et al, 2012, Non-small cell lung cancer. J Natl Compr Cancer Netw 10(10):1236–1271 Shamai S, Merimsky O, 2018, Efficacy and safety of Nivolumab in non-small cell lung cancer patients in Tel-Aviv tertiary medical center: facing the reality. Mol Clin Oncol 9(4):419–422 Osa A et al, 2018, Clinical implications of monitoring nivolumab immunokinetics in non-small cell lung cancer patients. JCI Insight:3(19) Nizam A, Aragon-Ching JB, 2018, Frontline immunotherapy treatment with nivolumab and ipilimumab in metastatic renal cell cancer: a new standard of care. Cancer Biol Ther:1–2 Long GV et al, 2018, Assessment of nivolumab exposure and clinical safety of 480 mg every 4 weeks flat-dosing schedule in patients with cancer. Ann Oncol Hida T, 2018, Nivolumab for the treatment of Japanese patients with advanced metastatic non-small cell lung cancer: a review of clinical trial evidence for efficacy and safety. Ther Adv Respir Dis 12:1753466618801167 Grimm SE et al, 2018, Nivolumab for treating metastatic or Unresectable urothelial Cancer: An evidence review Group perspective of a NICE single technology appraisal. Pharmacoeconomics Keir ME et al, 2008, PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 26:677–704 AgataYet al, 1996, Expression of the PD-1 antigen on the surface of stimulatedmouse Tand B lymphocytes. Int Immunol 8(5):765– 772 Zaric B et al, 2018, PD-1 and PD-L1 protein expression predict survival in completely resected lung adenocarcinoma. Clin Lung Cancer 19:e957–e963 Karim R et al, 2009, Tumor-expressed B7-H1 and B7-DC in relation to PD-1+ T-cell infiltration and survival of patients with cervical carcinoma. Clin Cancer Res 15(20):6341–6347 PD-1 inhibitors raise survival in NSCLC. Cancer Discov, 2014. 4(1):6 Yang ZZ et al, 2015, PD-1 expression defines two distinct T-cell sub-populations in follicular lymphoma that differentially impact patient survival. Blood Cancer J 5:e281 Prasanth KV, Spector DL, 2007, Eukaryotic regulatory RNAs: an answer to the 'genome complexity' conundrum. Genes Dev 21(1): 11–42 Enfield KS et al, 2012, Mechanistic roles of noncoding RNAs in lung Cancer biology and their clinical implications. Genet Res Int 2012:737416 Mohr AM, Mott JL, 2015, Overview of microRNA biology. Semin Liver Dis 35(1):3–11 Bentwich I et al, 2005, Identification of hundreds of conserved and nonconserved human microRNAs. Nat Genet 37(7):766–770 Berezikov E et al, 2005, Phylogenetic shadowing and computational identification of human microRNA genes. Cell 120(1):21–24 Voorhoeve PM, 2010, MicroRNAs: oncogenes, tumor suppressors or master regulators of cancer heterogeneity? Biochim Biophys Acta 1805(1):72–86 Lee YS, Dutta A, 2006)MicroRNAs: small but potent oncogenes or tumor suppressors. Curr Opin Investig Drugs 7(6):560–564 Chen CZ, 2005, MicroRNAs as oncogenes and tumor suppressors. N Engl J Med 353(17):1768–1771 Ortholan C et al, 2009, MicroRNAs and lung cancer: new oncogenes and tumor suppressors, new prognostic factors and potential therapeutic targets. Curr Med Chem 16(9):1047–1061 Wozniak M, Mielczarek A, Czyz M, 2016, miRNAs in melanoma: tumor suppressors and oncogenes with prognostic potential. Curr Med Chem 23(28):3136–3153 Bergmann JH, Spector DL, 2014, Long non-coding RNAs: modulators of nuclear structure and function. Curr Opin Cell Biol 26: 10–18 Cabili MN et al, 2011, Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 25(18):1915–1927 Dinger ME et al, 2008, Long noncoding RNAs in mouse embryonic stem cell pluripotency and differentiation. Genome Res 18(9):1433–1445 Batista PJ, Chang HY, 2013, Long noncoding RNAs: cellular address codes in development and disease. Cell 152(6):1298–1307 Pei X, Wang X, Li H, 2018, LncRNA SNHG1 regulates the differentiation of Treg cells and affects the immune escape of breast cancer via regulating miR-448/IDO. Int J Biol Macromol 118(Pt A):24–30 Guo Q et al, 2015, Comprehensive analysis of lncRNA-mRNA co-expression patterns identifies immune-associated lncRNA biomarkers in ovarian cancer malignant progression. Sci Rep 5:17683 Rusek AM et al, 2015, MicroRNA modulators of epigenetic regulation, the tumor microenvironment and the immune system in lung cancer. Mol Cancer 14:34 Ray M, Ruffalo MM, Bar-Joseph Z, 2018, Construction of integrated microRNA and mRNA immune cell signatures to predict survival of patients with breast and ovarian cancer. Genes Chromosom Cancer Miao BP et al, 2015, Nasopharyngeal cancer-derived microRNA- 21 promotes immune suppressive B cells. Cell Mol Immunol 12(6):750–756 Li ZH et al., 2018, MicroRNA-92a promotes tumor growth and suppresses immune function through activation of MAPK/ERK signaling pathway by inhibiting PTEN in mice bearing U14 cervical cancer. Cancer Med Korsunsky I et al, 2017, Two microRNA signatures for malignancy and immune infiltration predict overall survival in advanced epithelial ovarian cancer. J Investig Med 65(7):1068–1076 Khorrami S et al, 2017, MicroRNA-146a induces immune suppression and drug-resistant colorectal cancer cells. Tumour Biol 39(5):1010428317698365 Afonso-Grunz F, Muller S, 2015, Principles of miRNA-mRNA interactions: beyond sequence complementarity. CellMol Life Sci 72(16):3127–3141 Kataoka K et al, 2016, Aberrant PD-L1 expression through 3'- UTR disruption in multiple cancers. Nature 534(7607):402–406 DuWet al, 2017, Variant SNPs at the microRNA complementary site in the B7-H1 3′-untranslated region increase the risk of nonsmall cell lung cancer. Mol Med Rep 16(3):2682–2690 Wang Wet al, 2012, A frequent somatic mutation in CD274 3'- UTR leads to protein over-expression in gastric cancer by disrupting miR-570 binding. Hum Mutat 33(3):480–484 Gong AYet al, 2009)MicroRNA-513 regulates B7-H1 translation and is involved in IFN-gamma-induced B7-H1 expression in cholangiocytes. J Immunol 182(3):1325–1333 Gong AY et al, 2010, Cryptosporidium parvum induces B7-H1 expression in cholangiocytes by down-regulating microRNA-513. J Infect Dis 201(1):160–169 Yee D et al, 2017, MicroRNA-155 induction via TNF-alpha and IFN-gamma suppresses expression of programmed death ligand-1, PD-L1, in human primary cells. J Biol Chem 292(50):20683–20693 Zhang J, BraunMY, 2014, PD-1 deletion restores susceptibility to experimental autoimmune encephalomyelitis in miR-155- deficient mice. Int Immunol 26(7):407–415 CioffiMet al, 2017, The miR-25-93-106b cluster regulates tumor metastasis and immune evasion via modulation of CXCL12 and PD-L1. Oncotarget 8(13):21609–21625 Takamizawa J et al, 2004, Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res 64(11):3753–3756 Yu SL et al, 2008, MicroRNA signature predicts survival and relapse in lung cancer. Cancer Cell 13(1):48–57 Wang DTet al, 2013, miR-150, p53 protein and relevant miRNAs consist of a regulatory network in NSCLC tumorigenesis. Oncol Rep 30(1):492–498 Fu Y et al, 2018, Silencing of Long non-coding RNA MIAT sensitizes lung Cancer cells to Gefitinib by epigenetically regulating miR-34a. Front Pharmacol 9:82 Shi Yet al, 2014, The microRNA miR-34a inhibits non-small cell lung cancer, NSCLC, growth and the CD44hi stem-like NSCLC cells. PLoS One 9(3):e90022 Wang J et al, 2014, Downregulation of miR-486-5p contributes to tumor progression and metastasis by targeting protumorigenic ARHGAP5 in lung cancer. Oncogene 33(9):1181–1189 Shao Y et al, 2016, Direct repression of the oncogene CDK4 by the tumor suppressor miR-486-5p in non-small cell lung cancer. Oncotarget 7(23):34011–34021 Gao ZJ et al, 2018, miR-486-5p functions as an oncogene by targeting PTEN in non-small cell lung cancer. Pathol Res Pract 214(5):700–705 Yu S, Geng S, Hu Y, 2018, miR-486-5p inhibits cell proliferation and invasion through repressing GAB2 in non-small cell lung cancer. Oncol Lett 16(3):3525–3530 Liang C et al, 2017)MicroRNA-18a-5p functions as an oncogene by directly targeting IRF2 in lung cancer. Cell Death Dis 8(5):e2764 Shen Z et al, 2015, Effect of miR-18a overexpression on the radiosensitivity of non-small cell lung cancer. Int J Clin Exp Pathol 8(1):643–648 Chen G et al, 2013, miR-146a inhibits cell growth, cell migration and induces apoptosis in non-small cell lung cancer cells. PLoS One 8(3):e60317 Park DH et al, 2015, MicroRNA-146a inhibits epithelial mesenchymal transition in non-small cell lung cancer by targeting insulin receptor substrate 2. Int J Oncol 47(4):1545–1553 Kapodistrias N, Bobori C, Theocharopoulou G, 2017, MiR-140- 3p Downregulation in Association with PDL-1 Overexpression in Many Cancers: A Review from the Literature Using Predictive Bioinformatics Tools. Adv Exp Med Biol 988:225–233 Ji X,Wang E, Tian F, 2018, MicroRNA-140 suppresses osteosarcoma tumor growth by enhancing anti-tumor immune response and blocking mTOR signaling. Biochem Biophys Res Commun 495(1):1342–1348 Xie WB et al, 2018, MiR-140 expression regulates cell proliferation and targets PD-L1 in NSCLC. Cell Physiol Biochem 46(2): 654–663 Boldrini L et al, 2017, Role of microRNA-33a in regulating the expression of PD-1 in lung adenocarcinoma. Cancer Cell Int 17:105 Jiang D et al, 2006, Duplication and expression analysis of multicopy miRNA gene family members in Arabidopsis and rice. Cell Res 16(5):507–518 Senfter D et al, 2016, The microRNA-200 family: still much to discover. Biomol Concepts 7(5–6):311–319 Wang R et al, 2013, Functional role of miR-34 family in human cancer. Curr Drug Targets 14(10):1185–1191 Vergani E et al, 2016, Overcoming melanoma resistance to vemurafenib by targeting CCL2-induced miR-34a, miR-100 and miR-125b. Oncotarget 7(4):4428–4441 HuarteMet al, 2010, A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell 142(3):409–419 Cortez MA et al, 2016, PDL1 Regulation by p53 via miR-34. J Natl Cancer Inst:108(1) FengX et al, 2014)MiR-200, a new starmiRNA in human cancer. Cancer Lett 344(2):166–173 Humphries B, Yang C, 2015, The microRNA-200 family: small molecules with novel roles in cancer development, progression and therapy. Oncotarget 6(9):6472–6498 Diaz Tet al, 2014, Role ofmiR-200 familymembers in survival of colorectal cancer patients treated with fluoropyrimidines. J Surg Oncol 109(7):676–683 Chen L et al, 2014, Metastasis is regulated via microRNA-200/ ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat Commun 5:5241 Fujita Y et al, 2015, The clinical relevance of the miR-197/ CKS1B/STAT3-mediated PD-L1 network in chemoresistant non-small-cell lung cancer. Mol Ther 23(4):717–727 Tang D et al, 2018, The miR-3127-5p/p-STAT3 axis up-regulates PD-L1 inducing chemoresistance in non-small-cell lung cancer. J Cell Mol Med 22:3847–3856 Audrito Vet al, 2017, PD-L1 up-regulation inmelanoma increases disease aggressiveness and is mediated through miR-17-5p. Oncotarget 8(9):15894–15911 Li Q et al, 2016, miR-28modulates exhaustive differentiation of T cells through silencing programmed cell death-1 and regulating cytokine secretion. Oncotarget 7(33):53735–53750 Rivera LB, Bergers G, 2013, Location, location, location: macrophage positioning within tumors determines pro- or antitumor activity. Cancer Cell 24(6):687–689 Jahangiri A et al, 2013, Gene expression profile identifies tyrosine kinase c-met as a targetable mediator of antiangiogenic therapy resistance. Clin Cancer Res 19(7):1773–1783 Biswas SK,Mantovani A, 2010)Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol 11(10):889–896 Liu G, Abraham E, 2013, MicroRNAs in immune response and macrophage polarization. Arterioscler Thromb Vasc Biol 33(2): 170–177 Choi B et al, 2015, The relevance of miRNA-21 in HSV-induced inflammation in a mouse model. Int J Mol Sci 16(4):7413–7427 Zhang G et al, 2015, microRNA-4717 differentially interacts with its polymorphic target in the PD1 3′ untranslated region: A mechanism for regulating PD-1 expression and function in HBVassociated liver diseases. Oncotarget 6(22):18933–18944 Zhang Z et al, 2008, miR-21 plays a pivotal role in gastric cancer pathogenesis and progression. Lab Investig 88(12):1358–1366 YangSMet al, 2013)miR-21 confers cisplatin resistance in gastric cancer cells by regulating PTEN. Toxicology 306:162–168 Xu Y et al, 2012, miR-21 Is a Promising Novel Biomarker for Lymph Node Metastasis in Patients with Gastric Cancer. Gastroenterol Res Pract 2012:640168 P LA et al, 2018)Up-Regulation ofmiR-21,miR-25, miR-93, and miR-106b in Gastric Cancer. Iran Biomed J 22(6):367–373 Zhang H et al, 2012, miR-21 downregulated TCF21 to inhibit KISS1 in renal cancer. Urology 80(6):1298–1302 e1 An F, Liu Y, Hu Y, 2017, miR-21 inhibition of LATS1 promotes proliferation and metastasis of renal cancer cells and tumor stem cell phenotype. Oncol Lett 14(4):4684–4688 Winther M et al, 2015, Evaluation of miR-21 and miR-375 as prognostic biomarkers in esophageal cancer. Acta Oncol 54(9): 1582–1591 Wen SW et al, 2015, Association of miR-21 with esophageal cancer prognosis: a meta-analysis. Genet Mol Res 14(2): 6578–6582 Fu C et al, 2014, The expression of miR-21 and miR-375 predict prognosis of esophageal cancer. Biochem Biophys Res Commun 446(4):1197–1203 Zhang J et al, 2012, miR-21, miR-17 and miR-19a induced by phosphatase of regenerating liver-3 promote the proliferation and metastasis of colon cancer. Br J Cancer 107(2):352–359 Yu Yet al, 2015, miR-21 and miR-145 cooperation in regulation of colon cancer stem cells. Mol Cancer 14:98 Deng J et al, 2014, Targeting miR-21 enhances the sensitivity of human colon cancer HT-29 cells to chemoradiotherapy in vitro. Biochem Biophys Res Commun 443(3):789–795 Xue X et al, 2016, MiR-21 and MiR-155 promote non-small cell lung cancer progression by downregulating SOCS1, SOCS6, and PTEN. Oncotarget 7(51):84508–84519 Xia H et al, 2017, miR-21 modulates the effect of EZH2 on the biological behavior of human lung cancer stem cells in vitro. Oncotarget 8(49):85442–85451 Su C et al, 2018, MiR-21 improves invasion and migration of drug-resistant lung adenocarcinoma cancer cell and transformation of EMT through targeting HBP1. Cancer Med 7(6): 2485–2503 Yang Y, Guo JX, Shao ZQ, 2017, miR-21 targets and inhibits tumor suppressor gene PTEN to promote prostate cancer cell proliferation and invasion: An experimental study. Asian Pac J Trop Med 10(1):87–91 Reis ST et al, 2012, miR-21 may acts as an oncomir by targeting RECK, a matrix metalloproteinase regulator, in prostate cancer. BMC Urol 12:14 Amankwah EK et al, 2013, miR-21, miR-221 and miR-222 expression and prostate cancer recurrence among obese and nonobese cases. Asian J Androl 15(2):226–230 Zadeh MM, Ranji N, Motamed N, 2015, Deregulation of miR-21 and miR-155 and their putative targets after silibinin treatment in T47D breast cancer cells. Iran J BasicMed Sci 18(12):1209–1214 FragniMet al, 2016, The miR-21/PTEN/Akt signaling pathway is involved in the anti-tumoral effects of zoledronic acid in human breast cancer cell lines. Naunyn Schmiedeberg's Arch Pharmacol 389(5):529–538 Yang CH et al, 2015, The oncogenic microRNA-21 inhibits the tumor suppressive activity of FBXO11 to promote tumorigenesis. J Biol Chem 290(10):6037–6046 Iliopoulos D et al, 2011, The negative costimulatory molecule PD-1 modulates the balance between immunity and tolerance via miR-21. Eur J Immunol 41(6):1754–1763 Xi J et al, 2018, miR-21 depletion in macrophages promotes tumoricidal polarization and enhances PD-1 immunotherapy. Oncogene 37(23):3151–3165 Tran E et al, 2015, Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 350(6266):1387–1390 Xie G et al, 2017, Helicobacter pylori promote B7-H1 expression by suppressing miR-152 and miR-200b in gastric Cancer cells. PLoS One 12(1):e0168822 Wang Y et al, 2017, MicroRNA-152 regulates immune response via targeting B7-H1 in gastric carcinoma. Oncotarget 8(17): 28125–28134 Zhao L et al, 2016, The tumor suppressor miR-138-5p targets PDL1 in colorectal cancer. Oncotarget 7(29):45370–45384 Xu C et al, 2014, Loss of Lkb1 and Pten leads to lung squamous cell carcinoma with elevated PD-L1 expression. Cancer Cell 25(5):590–604 Zhu J et al, 2014, MiR-20b, −21, and -130b inhibit PTEN expression resulting in B7-H1 over-expression in advanced colorectal cancer. Hum Immunol 75(4):348–353 Shlush LI, Mitchell A, 2015, AML evolution from preleukemia to leukemia and relapse. Best Pract Res Clin Haematol 28(2–3):81–89 Wang X et al, 2015, Tumor suppressor miR-34a targets PD-L1 and functions as a potential immunotherapeutic target in acute myeloid leukemia. Cell Signal 27(3):443–452 Pyzer AR et al, 2017, MUC1 inhibition leads to decrease in PD-L1 levels via upregulation of miRNAs. Leukemia 31(12): 2780–2790 Xu S et al, 2016, miR-424(322, reverses chemoresistance via Tcell immune response activation by blocking the PD-L1 immune checkpoint. Nat Commun 7:11406 Tao Z et al, 2018, MiR-195/−16 family enhances radiotherapy via T cell activation in the tumor microenvironment by blocking the PD-L1 immune checkpoint. Cell Physiol Biochem 48(2):801–814 Huang F et al, 2018, MicroRNA-374b inhibits liver cancer progression via down regulating programmed cell death-1 expression on cytokine-induced killer cells. Oncol Lett 15(4):4797–4804 Wu Q et al, 2018, miR-375 inhibits IFN-gamma-induced programmed death 1 ligand 1 surface expression in head and neck squamous cell carcinoma cells by blocking JAK2/STAT1 signaling. Oncol Rep 39(3):1461–1468 Wei J et al, 2016, MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints. Neuro-Oncology 18(5):639–648 Jia L et al, 2017, miR-142-5p regulates tumor cell PD-L1 expression and enhances anti-tumor immunity. Biochem Biophys Res Commun 488(2):425–431 Zou MX et al, 2018, Clinicopathologic implications of CD8(+ )/Foxp3(+, ratio and miR-574-3p/PD-L1 axis in spinal chordoma patients. Cancer Immunol Immunother 67(2):209–224 He B, Yan F, Wu C, 2018, Overexpressed miR-195 attenuated immune escape of diffuse large B-cell lymphoma by targeting PD-L1. Biomed Pharmacother 98:95–101 Gao L et al, 2019, MiR-873/PD-L1 axis regulates the stemness of breast cancer cells. EBioMedicine 41:395–407 Tang Yet al, 2017, Co-expression of AFAP1-AS1 and PD-1 predicts poor prognosis in nasopharyngeal carcinoma. Oncotarget 8(24):39001–39011 Haderk F et al, 2017, Tumor-derived exosomes modulate PD-L1 expression in monocytes. Science Immunology 2(13) Zhang C et al, 2019, Upregulation of long noncoding RNA SNHG20 promotes cell growth and metastasis in esophageal squamous cell carcinoma via modulating ATM-JAK-PD-L1 pathway. J Cell Biochem Gilligan KE, Dwyer RM, 2017, Engineering Exosomes for Cancer Therapy. Int J Mol Sci:18(6) Kazandjian D et al, 2016, FDA approval summary: Nivolumab for the treatment of metastatic non-small cell lung Cancer with progression on or after platinum-based chemotherapy. Oncologist 21(5):634–642 Tanaka K et al., 2013, Tumor-suppressive function of proteintyrosine phosphatase non-receptor type 23 in testicular germ cell tumors is lost upon overexpression of miR142-3p microRNA. J Biol Chem 288(33): p. 23990-9 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 651 EP 663 DI 10.1007/s12253-019-00735-9 PG 13 ER PT J AU Cserni, G Sejben, A AF Cserni, Gabor Sejben, Anita TI Grading Ductal Carcinoma In Situ (DCIS) of the Breast – What’s Wrong with It? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Breast cancer; Ductal carcinoma in situ; Grade ID Breast cancer; Ductal carcinoma in situ; Grade AB Ductal carcinoma in situ of the breast is a non-obligate precursor of invasive breast cancer, and at its lower risk endmight not need treatment, a hypothesis tested in several currently running randomized clinical trials. This review describes the heterogeneity of grading ductal carcinoma in situ (DCIS). First it considers differences between low and high grade DCIS, and then it looks at several grading schemes and highlights how different these are, not only in the features considered for defining a given grade but also in their wording of a given variable seen in the grade in question. Rather than being fully comprehensive, the review aims to illustrate the inconsistencies. Reproducibility studies on grading mostly suggestive of moderate agreement on DCIS differentiation are also illustrated. The need for a well structured, more uniformand widely accepted language for grading DCIS is urged to avoid misunderstanding based misclassifications and improper treatment selection. C1 [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, H-6000 Kecskemet, Hungary. [Sejben, Anita] University of Szeged, Department of Pathology, Allomas u. 1, H-6725 Szeged, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. EM cserni@freemail.hu CR Wilkerson PM, Dedes KJ, Lopez-Garcia MA, Geyer FC, Reis- Filho JS, 2011, The molecular evolution of breast cancer precursors and risk indicators. In: Kahan Z, Tot T, eds, Breast cancer, a heterogeneous disease entity. The very early stages. Springer Science+ Business Media, Dordrecht, pp 89–117 Elston CW, Ellis IO, Pathological prognostic factors in breast cancer. I, 1991, The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19:403–410 Amendoeira I, Apostolikas N, Bellocq JP, Bianchi S, Boecker W, Borisch B, Bussolati G, Connolly CE, Cserni G, Decker T, Dervan P, Drijkoningen M, Ellis IO, Elston CW, Eusebi V, Faverly D, Heikkila P, Holland R, Kerner H, Kulka J, Jacquemier J, Lacerda M, Martinez-Penuela J, De Miguel C, Nordgren H, Peterse JL, Rank F, Regitnig P, Reiner A, Sapino A, Sigal-Zafrani B, Tanous AM, Thorstenson S, Zozaya E, Wells CA, EC Working Group on Breast Screening Pathology, 2006, Quality assurance guidelines for pathology. In: Perry N, Broeders M, de Wolf C, Tornberg S, Holland R, von Karsa L, eds, European guidelines for quality assurance in breast cancer screening and diagnosis, 4th edn. European Comission, Luxemburg, pp 219–311 Ellis IO, Carder P, Hales S, Lee AHS, Pinder SE, Rakha E, Al-Sam S, Deb R, Hanby A, Liebmann R, Provenzano E, Rowlands D, Wells CA, Anderson N, Girling A, Ibrahim M, Mallon E, Quinn C, 2016, Pathology reporting of breast disease in surgical excision specimens incorporating the dataset for histological reporting of breast cancer. The Royal College of Pathologists. https://www. rcpath.org/uploads/assets/uploaded/c5a73c7a-50a4-4077- 8815c49abbcbc1f1.pdf. Accessed 13May 2019 Lester SC, Bose S, Chen YY, Connolly JL, de Baca ME, Fitzgibbons PL, Hayes DF, Kleer C, O'Malley FP, Page DL, Smith BL, Tan LK, Weaver DL, Winer E, 2009, Protocol for the examination of specimens from patients with ductal carcinoma in situ of the breast. Arch Pathol Lab Med 133:15–25 Schnitt SJ, Allred C, Britton P, Ellis IO, Lakhani SR, Morrow M, Palazzo J, Reynolds C, Rutgers E, Simpson J, van de Vijver MJ, Vincent-Salomon A, 2012, Ductal carcinoma in situ. In: Lakhani SR, Ellis IO, Schnitt SJ, Tan PH, van de Vijver MJ, eds, WHO classification of tumours of the breast, 4th edn. International Agency for Research on Cancer, Lyon, pp 90–94 Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, Nordgren H, Farmer P, Praz V, Haibe-Kains B, Desmedt C, Larsimont D, Cardoso F, Peterse H, Nuyten D, Buyse M, Van de Vijver MJ, Bergh J, Piccart M, Delorenzi M, 2006, Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst 98:262–272 Hannemann J, Velds A, Halfwerk JB, Kreike B, Peterse JL, van de VijverMJ, 2006, Classification of ductal carcinoma in situ by gene expression profiling. Breast Cancer Res 8:61 Balleine RL, Webster LR, Davis S, Salisbury EL, Palazzo JP, Schwartz GF, Cornfield DB, Walker RL, Byth K, Clarke CL, Meltzer PS, 2008, Molecular grading of ductal carcinoma in situ of the breast. Clin Cancer Res 14:8244–8252 Sanders ME, Schuyler PA, Simpson JF, Page DL, Dupont WD, 2015, Continued observation of the natural history of low-grade ductal carcinoma in situ reaffirms proclivity for local recurrence even after more than 30 years of follow-up. Mod Pathol 28:662– 669 Maxwell AJ, Clements K, Hilton B, Dodwell DJ, Evans A, Kearins O, Pinder SE, Thomas J, Wallis MG, Thompson AM, 2018, Risk factors for the development of invasive cancer in unresected ductal carcinoma in situ. Eur J Surg Oncol 44:429–435 Cserni G, 2002, Tumour histological grade may progress between primary and recurrent invasive mammary carcinoma. J Clin Pathol 55:293–297 Wang SY, Shamliyan T, Virnig BA, Kane R, 2011, Tumor characteristics as predictors of local recurrence after treatment of ductal carcinoma in situ: a meta-analysis. Breast Cancer Res Treat 127:1– 14. Elshof LE, Tryfonidis K, Slaets L, van Leeuwen-Stok AE, Skinner VP, Dif N, Pijnappel RM, Bijker N, Rutgers EJ,Wesseling J, 2015, Feasibility of a prospective, randomised, open-label, international multicentre, phase III, non-inferiority trial to assess the safety of active surveillance for low risk ductal carcinoma in situ - the LORD study. Eur J Cancer 51:1497–1510 Francis A, Thomas J, Fallowfield L,WallisM, Bartlett JM, Brookes C, Roberts T, Pirrie S, Gaunt C, Young J, BillinghamL, Dodwell D, Hanby A, Pinder SE, Evans A, Reed M, Jenkins V, Matthews L, Wilcox M, Fairbrother P, Bowden S, Rea D, 2015, Addressing overtreatment of screen detected DCIS; the LORIS trial. Eur J Cancer 51:2296–2303 ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/ NCT02926911. Accessed 13 May 2019 Toss M, Miligy I, Thompson AM, Khout H, Green AR, Ellis IO, Rakha EA, 2017, Current trials to reduce surgical intervention in ductal carcinoma in situ of the breast: critical review. Breast 35: 151–156 Rea D, Francis A, Wallis M, Thomas J, Bartlett J, Bowden S, Dodwell D, Fallowfield L, Gaunt C, Hanby A, Jenkins V, Matthews L, Pinder S, Pirrie S, Reed M, Wilcox M, Roberts T, Kirwan C, Brookes C, Fairbrother P, Billingham L, Evans A, Young J, 2017, Confusion over differences in registration and randomization criteria for the LORIS, low-risk DCIS, trial. Ann Surg Oncol 24(Suppl 3):566–567 Holland R, Peterse JL, Millis RR, Eusebi V, Faverly D, van de Vijver MJ, Zafrani B, 1994, Ductal carcinoma in situ: A proposal for a new classification. Semin Diagn Pathol 11:167–180 SilversteinMJ, LagiosMD, Craig PH,Waisman JR, Lewinsky BS, Colburn WJ, Poller DN, 1996, A prognostic index for ductal carcinoma in situ of the breast. Cancer 77:2267–2274 Consensus Conference Committee, 1998, Consensus Conference on the classification of ductal carcinoma in situ. Cancer 1;82:2293– 2295 National Breast and Ovarian Cancer Centre and Australian Cancer Network, 2008, The pathology reporting of breast cancer. A guide for pathologists, surgeons, radiologists and oncologists, 3rd edn. National Breast and Ovarian Cancer Centre, Surry Hills, NSW. https://www.cancer.org.au/content/pdf/HealthProfessionals/ ClinicalGuidelines/Pathology_reporting_breastcancer_3rdEd2008. pdf. Accessed 13 May 2019 Cutuli B, Arnould L, Barreau B, Bellocq JP, Bonnier P, Fignon A, Fondrinier E, Fourquet A, Lemanski C, Lesur A, Sigal-Zafrani B, Tunon de Lara C, 2009, Recommandations professionnelles Cancer du Sein in situ. Institut National du Cancer, Boulogne- Billancourt. https://www.ecancer.fr/content/download/95918/ 1021188/file/RECOSIS10.pdf. Accessed 13 May 2019 Integraal Kankercentrum Nederland, 2012, Breast cancer: Dutch guidelines, version 2.0; NABON 2012. Oncoline. https://www. oncoline.nl/uploaded/docs/mammacarcinoom/Dutch%20Breast% 20Cancer%20Guideline%202012.pdf. Accessed 13 May 2019 Leitlinienprogramm Onkologie, 2018, Interdisziplinare S3- Leitlinie fur die Fruherkennung, Diagnostik, Therapie und Nachsorge des Mammakarzinoms. Langversion 4.1. https://www. leitlinienprogramm-onkologie.de/leitlinien/mammakarzinom/. Accessed 13 May 2019 Fisher ER, Gregorio RM, Fisher B, Redmond C, Vellios F, Sommers SC, 1975, The Pathology of invasive breast cancer. A Syllabus derived from findings of the National Surgical Adjuvant Breast Project, Protocol No. 4). Cancer 36:1–85 Silverstein MJ, 2003, The University of Southern California/Van Nuys prognostic index for ductal carcinoma in situ of the breast. Am J Surg 186:337–343 Rudloff U, Brogi E, Reiner AS, Goldberg JI, Brockway JP, Wynveen CA, McCormick B, Patil S, Van Zee KJ, 2010, The influence of margin width and volume of disease near margin on benefit of radiation therapy for women with DCIS treated with breast-conserving therapy. Ann Surg 251(4):583–591 Sagara Y, Freedman RA, Vaz-Luis I, Mallory MA, Wong SM, Aydogan F, DeSantis S, Barry WT, Golshan M, 2016, Patient Prognostic Score and Associations With Survival Improvement Offered by Radiotherapy After Breast-Conserving Surgery for Ductal Carcinoma In Situ: A Population-Based Longitudinal Cohort Study. J Clin Oncol 10;34(11):1190–1196 Scott MA, Lagios MD, Axelsson K, Rogers LW, Anderson TJ, Page DL, 1997, Ductal carcinoma in situ of the breast: reproducibility of histological subtype analysis. Hum Pathol 28:967–973 Bethwaite P, Smith N, Delahunt B, Kenwright D, 1998, Reproducibility of new classification schemes for the pathology of ductal carcinoma in situ of the breast. J Clin Pathol 51:450–454 Sneige N, Lagios MD, Schwarting R, Colburn W, Atkinson E, Weber D, Sahin A, Kemp B, Hoque A, Risin S, Sabichi A, Boone C, Dhingra K, Kelloff G, Lippman S, 1999, Interobserver reproducibility of the Lagios nuclear grading system for ductal carcinoma in situ. Hum Pathol 30(3):257–262 Sloane JP, Amendoeira I, Apostolikas N, Bellocq JP, Bianchi S, Boecker W, Bussolati G, Coleman D, Connolly CE, Eusebi V, De Miguel C, Dervan P, Drijkoningen R, Elston CW, Faverly D, Gad A, Jacquemier J, Lacerda M, Martinez-Penuela J, Munt C, Peterse JL, Rank F, Sylvan M, Tsakraklides V, Zafrani B, 1999, Consistency achieved by 23 European pathologists in categorizing ductal carcinoma in situ of the breast using five classifications. European Commission Working Group on Breast Screening Pathology. Hum Pathol 29(10):1056–1062 Wells WA, Carney PA, Eliassen MS, Grove MR, Tosteson AN, 2000, Pathologists' agreement with experts and reproducibility of breast ductal carcinoma-in-situ classification schemes. Am J Surg Pathol 24(5):651-659 Douglas-Jones AG, Morgan JM, Appleton MAC, Attanoos RL, Caslin A, Champ CS, Cotter M, Dallimore NS, Dawson A, Fortt RW, Griffiths AP,HughesM, Kitching PA, O’Brien C, Rashid AM, Stock D, Verghese A, Williams DW, Williams NW, Williams S, 2000, Consistency in the observation of features used to classify duct carcinoma in situ, DCIS, of the breast. J Clin Pathol 53:596- 602 Gomes DS, Porto SS, Balabram D, Gobbi H, 2014, Inter-observer variability between general pathologists and a specialist in breast pathology in the diagnosis of lobular neoplasia, columnar cell lesions, atypical ductal hyperplasia and ductal carcinoma in situ of the breast. Diagn Pathol 19;9:121 Schuh F, Biazus JV, Resetkova E, Benfica CZ, de FreitasVentura A, Uchoa D, Graudenz M, Edelweiss MIA, 2015, Histopathological grading of breast ductal carcinoma in situ: validation of a webbased survey through intra-observer reproducibility analysis. Diagn Pathol 10:93 Van Bockstal M, Baldewijns M, Colpaert C, Dano H, Floris G, Galant C, Lambein K, Peeters D, Van Renterghem S, Van Rompuy AS, Verbeke S, Verschuere S, Van Dorpe J, 2018, Dichotomous histopathological assessment of ductal carcinoma in situ of the breast results in substantial interobserver concordance. Histopathology 73(6):923-932 Landis JR, Koch GG, 1977, The measurement of observer agreement for categorical data. Biometrics 33(1):159-174 Van Dooijeweert C, van Diest PJ, Willems SM, Kuijpers CCH, Deckers IAG(2019, Significant inter- and intra-laboratory variation in grading of ductal carcinoma in situ of the breast: a nationwide study of 4901 patients in the Netherlands. Breast Cancer Res Treat 174(2):479-488 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 665 EP 671 DI 10.1007/s12253-019-00760-8 PG 7 ER PT J AU Burian, Zs Ladanyi, A Barbai, T Piurko, V Garay, T Raso, E Timar, J AF Burian, Zsuzsanna Ladanyi, Andrea Barbai, Tamas Piurko, Violetta Garay, Tamas Raso, Erzsebet Timar, Jozsef TI Selective Inhibition of HIF1α Expression by ZnSO4 Has Antitumoral Effects in Human Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ZnSO4; Human melanoma; HIF1α; Metastasis inhibition ID ZnSO4; Human melanoma; HIF1α; Metastasis inhibition AB Zinc as an essential trace metal is a ubiquitous component of various molecules of the cell. Studies indicated that it may modulate functions of various cancer cell types, and can even inhibit metastasis formation in experimental models. In melanoma, zinc was shown to affect melanin production and to induce apoptosis. Using human melanoma cell lines, we have tested the effects of ZnSO4 on cell proliferation, survival, migration as well as in vivo on experimental liver colony formation. We have found that ZnSO4 has antiproliferative and proapoptotic effects in vitro. In SCID mice intraperitoneal administration of ZnSO4 specifically inhibited liver colony formation without affecting primary tumor growth. To reveal the molecular mechanisms of action of zinc in human melanoma, we have tested mRNA expression of zinc finger transcription factors and found a strong inhibitory effect on HIF1α, as compared to WT1 whereas HIF2α and MTF1 expression was unaffected. Immunohistochemical detection of HIF1α protein in liver metastases confirmed its decreased nuclear expression after in vivo ZnSO4 treatment. These data indicate that in human melanoma zinc administration may have an antimetastatic effect due to a selective downregulation of HIF1α. C1 [Burian, Zsuzsanna] National Institute of OncologyBudapest, Hungary. [Ladanyi, Andrea] National Institute of OncologyBudapest, Hungary. [Barbai, Tamas] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Piurko, Violetta] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Garay, Tamas] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Raso, Erzsebet] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM jtimar@gmail.com CR Vallee BL, Falchuk KH, 1993, The biochemical basis of zinc physiology. Phys Rev 73:79–118 Grattan BJ, Freake HC, 2012, Zinc and cancer: implications for LIV-1 in breast cancer. Nutrients 4:648–675 Intsuka S, Araki S, 1978, Plasma copper and zinc levels in patients with malignant tumors of digestive organs: clinical evaluation of the cu/Zn ratio. Cancer 42:626–631 Hisaki T, Furumoto T, Nozaka K, Koga S, 1988, Serum zinc and copper changes after gastrectomy in aged patients with gastric cancer. Jpn J Sur 18:158–163 Prasad AS, Beck FW, Doerr TD, Shamsa FH, Mathog RH, 1998, Nutritional and zinc status of head and neck cancer patients: an interpretive review. Am Coll Nutr 17:409–418 Yucel I, Arpaci F, Ozet A, Berk O, 1994, Serum copper and zinc levels and copper/zinc ratio in patients with breast cancer. Biol Trace Elem Res 40:31–38 Igic PG, Lee E, Harper W, Roach KW, 2012, Toxic effects associated with consumption of zinc. Mayo Clin Proc 77:713–716 Fosmire GJ, 1990, Zinc toxicity. Am J Clin Nutr 51:225–227 Timar J, Raso R, Paku S, Kopper L, 1998, Oral administration of a trace element preparation and zinc inhibit liver metastasis of 3LLHH murine tumor cells. Int J Mol Med 2:105–113 Dubi N, Gheber L, Fisgman D, Sekler I, Hershfinkel M, 2008, Extracellular zinc and zinc-citrate, acting through a putative zincsensing receptor, regulate growth and survival of prostate cancer cells. Carcinogenesis 29:1692–1702 Hwang JJ, Kim HN, Kim J, Cho DH, Kim MJ, Kim YS, Kim Y, Park SJ, Koh JY, 2010, Zinc(II, ion mediates tamoxifen-induced autophagy and cell death in MCF-7 breast cancer cell line. Biometals 23(6):997–1013 Yamada H, Suzuki K, Koizumi S, 2007, Gene expression profile in human cells exposed to zinc. J Toxicol Sci 32:193–196 Kindermann B, Doring F, Pfaffl M, Daniel H, 2004, Identification of genes responsive to intracellular zinc depletion in the human colon adenocarcinoma cell line HT-29. J Nutr 134:57–62 Schadendorf D, vanAkkoi ACJ, Berking C, Griewank KG, Gutzmer R, Hauschild A et al, 2018, Melanoma. Lancet 392: 971–984 Farmer PJ, Gidanian S, Shahandeh B, Di Bilio AJ, Tohidian N, Meyskens FL Jr., 2003, Melanin as a target for melanoma chemotherapy: pro-oxidant effect of oxygen and metals on melanoma viability. Pigment Cell Res 16:273–279 Ladanyi A, Timar J, Paku S, Molnar G, Lapis K, 1990, Selection and characterization of human melanoma lines with different livercolonizing capacity. Int J Cancer 46:456–461 Timar J, Raso E, Honn KV, Hagmann W, 1999, 12-lipoxygenase expression in human melanoma cell lines. Adv Exp Med Biol 469: 617–622 Garay, T., E. Juhasz, E. Molnar, M. Eisenbauer, A. Czirok, B. Dekan,et al., 2013, Cell migration or cytokinesis and proliferation? - revisiting the "go or grow" hypothesis in cancer cells in vitro. Exp Cell Res 319:3094–3103 Wellinghausen N, Kirchner H, Rink L, 1997, The immunobiology of zinc. Immunol Today 18:523–524 Franklin RB, Costello LC, 2009, The important role of the apoptotic effects of zinc in the development of cancers. J Cell Biochem 106:750–757 Kim I, Kim CH, Seo GH, Kim HS, Lee J, Kim DG, Ahn YS, 2008, Inhibitory effect of zinc on hypoxic HIF-1 activation in astrocytes. Neuroreport 19:1065–1068 Nardinocchi L, Pantisano V, Puca R, Porru M, Aiello A, Grasselli A, Leonetti C, SafranM, Rechavi G, Givol D, Farsetti A, D'Orazi G, 2010, Zinc downregulates HIF-1α and inhibits its activity in tumor cells in vitro and in vivo. PLoS One 5:e15048 Yuan Y, Hillind G, Ferguson T, Millhorn DE, 2003, Cobalt inhibits the interaction between hypoxia inducible factor-a and von Hippel Lindau protein by direct binding to hypoxia inducible factor-a. J Biol Chem 278:15911–15916 Chun YS, Choi E,Yeo EJ, Lee HJ, KimMS, Park JW(2001)Anew HIF1 alpha variant induced by zinc ion suppresses HIF1-mediated hypoxic responses. J Cell Sci 114:4051–4061 Nardinocchi L, Puca R, Sacchi A, Rechavi G, Givol D, D’Orazi G, 2009, Targeting hypoxia in cancer cells by restoring homeodomain interacting protein-kinase 2 and p53 activity and suppressing HIF1- alpha. PlosOne 4:e6819 Kuphal S, Winklmeier A, Warnecke C, Bosserhoff AK, 2010, Constitutive HIF-1 activity in malignant melanoma. Eur J Cancer 46:1159–1169 Zbytek B, Peacock DL, Seagroves TN, Slominski A, 2013, Putative role of HIF transcriptional activity in melanocytes and melanoma biology. Dermatoendocrinol 5:239–251 Valencak J, Kittler H, Schmid K, Schreiber M, Raderer M, Gonzalez-Inchaurraga M, Birner P, Pehamberger H, 2009, Prognostic relevance of hypoxia inducible factor-1alpha expression in patients with melanoma. Clin Exp Dermatol 34:e962–e964 Hanna SC, Krishnan B, Bailey ST, Moschos SJ, Kuan PF, Shimamura T, Osborne LD, Siegel MB, Duncan LM, O’Brien ET III, Superfine R, Miller CR, Simon MC, Wong KK, Kim WY, 2013, HIF1α and HIF2α independently activate SRC to promote melanoma metastases. J Clin Invest 123:2078–2093 Tatrai E, Bartal A,Gacs A, Paku S,Kenessey I, Garay T, Hegedus B, Molnar E, Cserepes MT, Hegedus Z, Kucsma N, Szakacs G, Tovari J, 2017, Cell-type dependent HIF1α-mediated effects of hypoxia on proliferation, migration and metastatic potential of human tumor cells. Oncotarget 8:44498–44510 Yeo EJ, Chun YS, Cho YS, Kim J, Lee JC, Kim MS, Park JW, 2003, YC-1: a potential anticancer drug targeting hypoxiainducible factor 1. J Natl Cancer Inst 95:516–525 Scheuermann TH, Li Q, Ma HW, Key J, Zhang L, Chen R, Garcia JA, Naidoo J, Longgood J, Frantz DE, Tambar UK, Gardner KH, Bruick RK, 2013, Allosteric inhibition of hypoxia inducible factor- 2 with small molecules. Nat Chem Biol 9:271–276 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 673 EP 679 DI 10.1007/s12253-018-00573-1 PG 7 ER PT J AU Nariman-Saleh-Fam, Z Saadatian, Z Nariman-Saleh-Fam, L Ouladsahebmadarek, E Tavakkoly-Bazzaz, J Bastami, M AF Nariman-Saleh-Fam, Ziba Saadatian, Zahra Nariman-Saleh-Fam, Lida Ouladsahebmadarek, Elaheh Tavakkoly-Bazzaz, Javad Bastami, Milad TI An Association and Meta-Analysis of Esophageal Squamous Cell Carcinoma Risk Associated with PLCE1 rs2274223, C20orf54 rs13042395 and RUNX1 rs2014300 Polymorphisms SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal cancer; Polymorphism; Iran; GWAS; Association; Meta-analysis ID Esophageal cancer; Polymorphism; Iran; GWAS; Association; Meta-analysis AB One of the highest risk of esophageal squamous cell carcinoma (ESCC) in the world has been reported in Iran, which is located in the Asian esophageal cancer belt. ESCC constitutes 90% of the esophageal cancer cases in Iran. Genome wide association studies (GWASs) in Chinese have identified a number of candidate variants, of which PLCE1rs2274223, C20orf54rs13042395 and RUNX1rs2014300 are studied in high risk populations including Chinese, Caucasians and Africans. However, results are inconsistent and it is unknown whether similar associations exist in Iranian population. We evaluated association of three GWAS identified variants with risk of ESCC in an Iranian cohort consisted of 200 ESCC patients and 300 healthy controls and conducted meta-analysis of ESCC risk associated with rs2274223 (involving 9810 cases and 13,128 controls) and rs13042395 (involving 2363 cases and 5329 controls). Logistic regression analysis showed that rs2274223 was associated with ESCC under codominant [GG/AA, 2.47(1.17–5.23), P:0.021], dominant [AG+GG/AA, 1.57(1.09–2.27), P:0.016], recessive [GG/AA+AG, 2.18(1.04–4.56), P:0.036] and log-additive models [1.51(1.12–2.02), P:0.006]. C20orf54 rs13042395 was not associated with ESCC under any genetic model. RUNX1 rs2014300 was associated with risk of ESCC assuming codominant [AG/GG, 0.63(0.41–0.97), P:0.018], dominant [AG+ AA/GG, 0.59 (0.39–0.89), P:0.010] and log-additive models [0.61 (0.42–0.87), P: 0.005]. Meta-analysis found significant associations between rs2274223 and ESCC under all analyzed genetic models. However, meta-analysis stratified by ethnicity showed a significant association in Asians but not non-Asian populations. No significant association was found for rs13042395 in meta-analysis. This study provided first evidence for association of GWAS-identified variants with risk of ESCC in an Iranian cohort. C1 [Nariman-Saleh-Fam, Ziba] Tabriz University of Medical Sciences, Women’s Reproductive Health Research CenterTabriz, Iran. [Saadatian, Zahra] Shahid Beheshti University of Medical Sciences, Faculty of Medicine, Student Research CommitteeTehran, Iran. [Nariman-Saleh-Fam, Lida] Tabriz University of Medical Sciences, Liver and Gastrointestinal Diseases Research CenterTabriz, Iran. [Ouladsahebmadarek, Elaheh] Tabriz University of Medical Sciences, Women’s Reproductive Health Research CenterTabriz, Iran. [Tavakkoly-Bazzaz, Javad] Shahid Beheshti University of Medical Sciences, Department of Medical GeneticsTehran, Iran. [Bastami, Milad] Tabriz University of Medical Sciences, Immunology Research CenterTabriz, Iran. RP Tavakkoly-Bazzaz, J (reprint author), Shahid Beheshti University of Medical Sciences, Department of Medical Genetics, Tehran, Iran. EM tavakkolybazzazj@tums.ac.ir CR Schweigert M, Dubecz A, Stein HJ, 2013, Oesophageal cancer–an overview. Nat Rev Gastroenterol Hepatol 10(4):230–244. https:// doi.org/10.1038/nrgastro.2012.236 Castro C, Bosetti C, Malvezzi M, Bertuccio P, Levi F, Negri E, La Vecchia C, LunetN, 2014, Patterns and trends in esophageal cancer mortality and incidence in Europe, 1980-2011, and predictions to 2015. Ann Oncol 25(1):283–290. , DOI 10.1093/annonc/ mdt486 Mahboubi E, Kmet J, Cook PJ, Day NE, Ghadirian P, Salmasizadeh S, 1973, Oesophageal cancer studies in the Caspian Littoral of Iran: the Caspian cancer registry. Br J Cancer 28(3):197–214 Zare M, Jazii FR, Alivand MR, Nasseri NK, Malekzadeh R, Yazdanbod M, 2009, Qualitative analysis of adenomatous polyposis coli promoter: Hypermethylation, engagement and effects on survival of patients with esophageal cancer in a high risk region of the world, a potential molecular marker. BMC Cancer 9(1):24. , DOI 10.1186/1471-2407-9-24 Sadjadi A, Marjani H, Semnani S, Nasseri-Moghaddam S, 2010, Esophageal Cancer in Iran: a review. Middle East J Cancer 1(1) Mosavi-Jarrahi A, Mohagheghi MA, 2006, Epidemiology of esophageal cancer in the high-risk population of Iran. Asian Pac J Cancer Prev 7(3):375–380 Wu C, Hu Z, He Z, Jia W, Wang F, Zhou Y, Liu Z, Zhan Q, Liu Y, Yu D, Zhai K, Chang J, Qiao Y, Jin G, Liu Z, Shen Y, Guo C, Fu J, Miao X, Tan W, Shen H, Ke Y, Zeng Y, Wu T, Lin D, 2011, Genome-wide association study identifies three new susceptibility loci for esophageal squamous-cell carcinoma in Chinese populations. Nat Genet 43(7):679–684. , DOI 10.1038/ng.849 Nariman-Saleh-Fam Z, Bastami M, Somi MH, Samadi N, Abbaszadegan MR, Behjati F, Ghaedi H, Tavakkoly-Bazzaz J, Masotti A, 2016, In silico dissection of miRNA targetome polymorphisms and their role in regulating miRNA-mediated gene expression in esophageal cancer. Cell Biochem Biophys 74:1–15. , DOI 10.1007/s12013-016-0754-5 Saadatian Z,Masotti A, Nariman Saleh Fam Z, Alipoor B, Bastami M, Ghaedi H, 2014, Single-nucleotide polymorphisms within MicroRNAs sequences and their 3' UTR target sites may regulate gene expression in gastrointestinal tract cancers. Iran Red Crescent Med J 16(7):e16659. , DOI 10.5812/ircmj.16659 Bastami M, Nariman-Saleh-Fam Z, Saadatian Z, Nariman-Saleh- Fam L,OmraniMD, Ghaderian SM,Masotti A, 2016, ThemiRNA targetome of coronary artery disease is perturbed by functional polymorphisms identified and prioritized by in-depth bioinformatics analyses exploiting genome-wide association studies. Gene 594(1):74–81. , DOI 10.1016/j.gene.2016.08.054 Lin Y, Totsuka Y, He Y, Kikuchi S, Qiao Y, Ueda J, Wei W, Inoue M, Tanaka H, 2013, Epidemiology of esophageal cancer in Japan and China. J Epidemiol 23(4):233–242 Abnet CC, FreedmanND, Hu N,Wang Z, Yu K, Shu XO,Yuan JM, ZhengW, Dawsey SM, Dong LM, Lee MP, Ding T, Qiao YL, Gao YT, Koh WP, Xiang YB, Tang ZZ, Fan JH, Wang C, Wheeler W, Gail MH, Yeager M, Yuenger J, Hutchinson A, Jacobs KB, Giffen CA, Burdett L, Fraumeni JF, Tucker MA, Chow WH, Goldstein AM, Chanock SJ, Taylor PR, 2010, A shared susceptibility locus in PLCE1 at 10q23 for gastric adenocarcinoma and esophageal squamous cell carcinoma. Nat Genet 42(9):764–767. , DOI 10. 1038/ng.649 Wu C,Wang Z, Song X, Feng XS, Abnet CC, 2014, Joint analysis of three genome-wide association studies of esophageal squamous cell carcinoma in Chinese populations. Nat Genet 46(9):1001– 1006. , DOI 10.1038/ng.3064 Wang LD, Zhou FY, Li XM et al, 2010, Genome-wide association study of esophageal squamous cell carcinoma in Chinese subjects identifies susceptibility loci at PLCE1 and C20orf54. Nat Genet 42(9):759–763. , DOI 10.1038/ng.648 Gu H, Ding G, Zhang W, Liu C, Chen Y, Chen S, Jiang P, 2012, Replication study of PLCE1 and C20orf54 polymorphism and risk of esophageal cancer in a Chinese population. Mol Biol Rep 39(9): 9105–9111. , DOI 10.1007/s11033-012-1782-x Palmer AJ, Lochhead P, Hold GL, Rabkin CS, Chow WH, Lissowska J, Vaughan TL, Berry S, Gammon M, Risch H, el- Omar EM, 2012, Genetic variation in C20orf54, PLCE1 and MUC1 and the risk of upper gastrointestinal cancers in Caucasian populations. Eur J Cancer Prev 21(6):541–544. , DOI 10. 1097/CEJ.0b013e3283529b79 Bye H, Prescott NJ, LewisCM, Matejcic M,Moodley L, Robertson B, Rensburg C, Parker MI, Mathew CG, 2012, Distinct genetic association at the PLCE1 locus with oesophageal squamous cell carcinoma in the south African population. Carcinogenesis 33(11):2155–2161. , DOI 10.1093/carcin/bgs262 Nariman-Saleh-Fam Z, BastamiM, Somi MH, Behjati F, Mansoori Y, Daraei A, Saadatian Z, Nariman-Saleh-Fam L, Mahmoodzadeh H, Makhdoumi Y, Tabrizi FV, Ebrahimi-Sharif B, Hezarian A, Naghashi S, Abbaszadegan MR, Tavakkoly-Bazzaz J, 2017, miRNA-related polymorphisms in miR-423, rs6505162, and PEX6, rs1129186, and risk of esophageal squamous cell carcinoma in an Iranian cohort. Genet Test Mol Biomarkers 21(6):382–390. , DOI 10.1089/gtmb.2016.0346 Umar M, Upadhyay R, Kumar S, Ghoshal UC, Mittal B, 2014, Role of novel and GWAS originated PLCE1 genetic variants in susceptibility and prognosis of esophageal cancer patients in northern Indian population. Tumour Biol 35(11):11667–11676. https:// doi.org/10.1007/s13277-014-2458-z Bastami M, Ghaderian SM, Omrani MD, Mirfakhraie R, Nariman- Saleh-Fam Z, Mansoori Y, Masotti A, 2015, Evaluating the association of common UBE2Z variants with coronary artery disease in an Iranian population. Cell Mol Biol, Noisy-le-grand, 61(7): 50–54 Bastami M, Ghaderian SM, Omrani MD, Mirfakhraie R, Vakili H, Parsa SA, Nariman-Saleh-Fam Z, Masotti A, 2016, MiRNArelated polymorphisms in miR-146a and TCF21 are associated with increased susceptibility to coronary artery disease in an Iranian population. Genet Test Mol Biomarkers 20(5):241–248. https:// doi.org/10.1089/gtmb.2015.0253 Graffelman J, 2015, Exploring Diallelic Genetic Markers: The HardyWeinberg Package. 2015 64(3):23. , DOI 10.18637/ jss.v064.i03 Gonzalez JR, Armengol L, Sole X, Guino E, Mercader JM, Estivill X, Moreno V, 2007, SNPassoc: an R package to perform whole genome association studies. Bioinformatics 23(5):644–645. https:// doi.org/10.1093/bioinformatics/btm025 Schwarzer G, 2007, meta: An R package for meta-analysis. R News, 3):40–45 DerSimonian R, Laird N, 1986, Meta-analysis in clinical trials. Control Clin Trials 7(3):177–188 Mantel N, Haenszel W, 1959, Statistical aspects of the analysis of data from retrospective studies of disease. J Natl Cancer Inst 22(4): 719–748 Begg CB, Mazumdar M, 1994, Operating characteristics of a rank correlation test for publication bias. Biometrics 50(4):1088–1101 Malik MA, Umar M, Gupta U, Zargar SA, Mittal B, 2014, Phospholipase C epsilon 1, PLCE1 rs2274223A>G, rs3765524C>Tand rs7922612C>T, polymorphisms and esophageal cancer risk in the Kashmir Valley. Asian Pac J Cancer Prev 15(10):4319–4323 Dong Y, Chen J, Chen Z, Tian C, Lu H, Ruan J, Yang W, 2015, Evaluating the Association of Eight Polymorphisms with Cancer susceptibility in a Han Chinese population. PLoS One 10(7): e0132797. , DOI 10.1371/journal.pone.0132797 Dura P, Bregitha CV, teMorsche RH et al, 2013)GWAS-uncovered SNPs in PLCE1 and RFT2 genes are not implicated in Dutch esophageal adenocarcinoma and squamous cell carcinoma etiology. Eur J Cancer Prev 22(5):417–419. , DOI 10.1097/CEJ. 0b013e32835c7f53 Piao JM, Shin MH, Kim HN, Song HR, Kweon SS, Choi JS, Shim HJ, Hwang JE, Bae WK, Kim SH, Choi YD, Cui LH, 2014, Replication of results of genome-wide association studies on esophageal squamous cell carcinoma susceptibility loci in a Korean population. Dis Esophagus 27(8):798–801. , DOI 10.1111/ dote.12155 Duan F, Xie W, Cui L, Wang P, Song C, Qu H, Wang K, Zhang J, Dai L, 2013, Novel functional variants locus in PLCE1 and susceptibility to esophageal squamous cell carcinoma: based on published genome-wide association studies in a central Chinese population. Cancer Epidemiol 37(5):647–652. , DOI 10.1016/j. canep.2013.04.009 Hu H, Yang J, Sun Y, Yang Y, Qian J, Jin L,Wang M, Bi R, Zhang R, Zhu M, Sun M, Ma H,Wei Q, Jiang G, Zhou X, Chen H, 2012, Putatively functional PLCE1 variants and susceptibility to esophageal squamous cell carcinoma, ESCC): a case-control study in eastern Chinese populations. Ann Surg Oncol 19(7):2403–2410. , DOI 10.1245/s10434-011-2160-y Yang J, Wu H, Wei S, Xiong H, Fu X, Qi Z, Jiang Q, Li W, Hu G, Yuan X, Liao Z, 2014, HPV seropositivity joints with susceptibility loci identified in GWASs at apoptosis associated genes to increase the risk of esophageal squamous cell carcinoma, ESCC). BMC Cancer 14(1). , DOI 10.1186/1471-2407-14-501 ZhouRM, Li Y,Wang N, Liu BC, Chen ZF, Zuo LF, 2012, PLC-ε1 gene polymorphisms significantly enhance the risk of esophageal squamous cell carcinoma in individuals with a family history of upper gastrointestinal cancers. Arch Med Res 43(7):578–584. , DOI 10.1016/j.arcmed.2012.09.006 Song X, Zhou FY, Zhang LQ, 2012, Correlation of PLCE1 gene polymorphism in the smoker/non-smokers, drinking /non-drinking, and BMI subgroups with esophageal squamous cell carcinoma. J Henan University 31:186–190 Chen YZ, Cui XB, Pang XL, Li L, Hu JM, Liu CX, CaoYW, Yang L, Li F, 2013, Relationship between rs2274223 and rs3765524 polymorphisms of PLCE1 and risk of esophageal squamous cell carcinoma in a kazakh Chinese population. Chin J Pathol 42(12): 795–800. , DOI 10.3760/cma.j.issn.0529-5807.2013.12. 002 Wei W, Ji A, Wang J, Wei Z, Lian C, Yang J, Ma L, Ma L, Qin X, Wang LD, 2013, Functional single nucleotide polymorphism in C20orf54 modifies susceptibility to esophageal squamous cell carcinoma. Dis Esophagus 26(1):97–103. , DOI 10.1111/j. 1442-2050.2012.01339.x Tan HZ,Wu ZY,Wu JY, Long L, Jiao JW, PengYH, XuYW, Li SS, Wang W, Zhang JJ, Li EM, Xu LY, 2016, Single nucleotide polymorphism rs13042395 in the SLC52A3 gene as a biomarker for regional lymph node metastasis and relapse-free survival of esophageal squamous cell carcinoma patients.BMC Cancer 16(560):560. , DOI 10.1186/s12885-016-2588-3 Xue W, Zhu M, Wang Y, He J, Zheng L, 2015, Association between PLCE1 rs2274223 a > G polymorphism and cancer risk: proof from a meta-analysis. Sci Rep 5(7986). , DOI 10. 1038/srep07986 Martins M, McCarthy A, Baxendale R, Guichard S, Magno L, Kessaris N, El-Bahrawy M, Yu P, Katan M, 2014, Tumor suppressor role of phospholipase C epsilon in Ras-triggered cancers. Proc Natl Acad Sci U S A 111(11):4239–4244. , DOI 10.1073/ pnas.1311500111 Danielsen SA, Cekaite L, Agesen TH, Sveen A, Nesbakken A, Thiis-Evensen E, Skotheim RI, Lind GE, Lothe RA, 2011, Phospholipase C isozymes are deregulated in colorectal cancer– insights gained from gene set enrichment analysis of the transcriptome. PLoS One 6(9):e24419. , DOI 10.1371/ journal.pone.0024419 Li M, Edamatsu H, Kitazawa R, Kitazawa S, Kataoka T, 2009, Phospholipase Cepsilon promotes intestinal tumorigenesis of Apc(min/+, mice through augmentation of inflammation and angiogenesis. Carcinogenesis 30(8):1424–1432. , DOI 10. 1093/carcin/bgp125 Zhao L, Wei ZB, Yang CQ, Chen JJ, Li D, Ji AF, Ma L, 2014, Effects of PLCE1 gene silencing by RNA interference on cell cycling and apoptosis in esophageal carcinoma cells. Asian Pac J Cancer Prev 15(13):5437–5442 Chen YZ, Cui XB, Hu JM, Zhang WJ, Li SG, Yang L, Shen XH, Liu CX, Pan QF, Yu SY, Yuan XL,Yang L, GuWY, Chen JZ,Wang LD, Li F, 2013, Overexpression of PLCE1 in Kazakh esophageal squamous cell carcinoma: implications in cancer metastasis and aggressiveness. Apmis 121(10):908–918. , DOI 10.1111/ apm.12095 Cui XB, Pang XL, Li S, Jin J, Hu JM, Yang L, Liu CX, Li L,Wen SJ, LiangWH, Chen YZ, Li F, 2014, Elevated expression patterns and tight correlation of the PLCE1 and NF-kappaB signaling in Kazakh patients with esophageal carcinoma. Med Oncol 31(1): 791. , DOI 10.1007/s12032-013-0791-5 Li Y, An J, Huang S, Liao H, Weng Y, Cai S, Zhang J, 2014, PLCE1 suppresses p53 expression in esophageal cancer cells. Cancer Investig 32(6):236–240. , DOI 10.3109/ 07357907.2014.905588 Cui XB, Chen YZ, Pang XL, LiuW, Hu JM, Li SG, Yang L, Zhang WJ, Liu CX, CaoYW, Jiang JF, GuWY, Pang J, Yang L, Yuan XL, Yu SY, Li F, 2013, Multiple polymorphisms within the PLCE1 are associated with esophageal cancer via promoting the gene expression in a Chinese Kazakh population. Gene 530(2):315–322. , DOI 10.1016/j.gene.2013.08.057 Duan F, Cui S, Song C, Zhao X, Dai L, Shen Y, 2015, Esophageal squamous cell carcinoma and gastric cardia adenocarcinoma shared susceptibility locus in C20orf54: evidence from published studies. Sci Rep 5(11961). , DOI 10.1038/srep11961 Wu H, Zheng J, Deng J, Zhang L, Li N, Li W, Li F, Lu J, Zhou Y, 2015, LincRNA-uc002yug.2 involves in alternative splicing of RUNX1 and serves as a predictor for esophageal cancer and prognosis. Oncogene 34(36):4723–4734. , DOI 10.1038/onc. 2014.400 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 681 EP 692 DI 10.1007/s12253-019-00579-3 PG 12 ER PT J AU Zhang, J Zhang, J Chen, W Li, H Li, M Li, L AF Zhang, Jianguo Zhang, Jianzhong Chen, Wenqi Li, Huiyu Li, Meiying Li, Lisha TI Bioinformatics Analysis Makes Revelation to Potential Properties on Regulation and Functions of Human Sox2 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bioinformatics analysis; SOX2; Protein-protein interactions; Proteomics; Protein regulation ID Bioinformatics analysis; SOX2; Protein-protein interactions; Proteomics; Protein regulation AB Sex determining region Y-box 2 (Sox2) is a transcription factor that is essential for maintaining self-renewal or pluripotency of undifferentiated embryonic stem cells. The expression and distribution of Sox2 in tumor tissues have been extensively recorded, which are related to the progression and metastasis of tumor. However, a complete mechanistic understanding of Sox2 regulation and function remains to be studied. Herein, we show new potential properties of Sox2 regulation and functions from bioinformatics analysis. We use numerous algorithms to characterize the Sox2 gene promoter elements and the Sox2 protein structure, physio-chemical, localization properties and its evolutionary relationships. The expression of Sox2 is regulated by a diverse set of transcription factors and associated with the levels of methylation of CpG Islands in promoters. The structural properties of Sox2 indicate that Sox2 expresses as a stem cell marker in a variety of stem cells. Sox2 together with other transcription factors or proteins regulate the expression of downstream target genes, which makes a great difference to the biological function of stem cells. Not only stem cells, Sox2 also play an important role in tumor cells. In conclusion, this information from bioinformatics analysis will help to understand Sox2 regulation and functions better in future attempts. C1 [Zhang, Jianguo] Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of PathobiologyChangchun, China. [Zhang, Jianzhong] Qingdao University, Department of MedicineQingdao, China. [Chen, Wenqi] Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of PathobiologyChangchun, China. [Li, Huiyu] Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of PathobiologyChangchun, China. [Li, Meiying] Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of PathobiologyChangchun, China. [Li, Lisha] Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of PathobiologyChangchun, China. RP Li, L (reprint author), Jilin University, Ministry of Education, Norman Bethune Medical College, The Key Laboratory of Pathobiology, Changchun, China. EM lilisha@jlu.edu.cn CR Sarkar A, Hochedlinger K, 2013, The sox family of transcription factors: versatile regulators of stem and progenitor cell fate. Cell Stem Cell 12:15–30 Rex M, Church R, Tointon K, Ichihashi RMA, Mokhtar S, Uwanogho D, Sharpe PT, Scotting PJ, 1998, Granule cell development in the cerebellum is punctuated by changes in Sox gene expression. Mol Brain Res 55:28–34 Adachi K, Nikaido I, Ohta H, Ohtsuka S, Ura H, Kadota M, Wakayama T, Ueda HR, Niwa H, 2013, Context-dependent wiring of Sox2 regulatory networks for self-renewal of embryonic and trophoblast stem cells. Mol Cell 52:380–392 Lin BY, Huang XF, Han X, Foltz G, 2011, SOX2, SRY, sex determining region Y)-box 2). Atlas Genet Cytogenet Oncol Haematol 15(12):1054–1057 Zhao X, Sun B, Sun D et al, 2015, Slug promotes hepatocellular cancer cell progression by increasing sox2 and nanog expression. Oncol Rep 33:149–156 Can T, 2014, Introduction to bioinformatics. Methods Mol Biol 1107:51–71 Eck RV, DayhoffMO(1966, Evolution of the structure of ferredoxin based on living relics of primitive amino acid sequences. Science 152:363–366 Reese MG, 2001, Application of a time-delay neural network to promoter annotation in the Drosophila melanogaster genome. Comput Chem 26:51–56 Li W, Cowley A, Uludag M, Gur T, McWilliam H, Squizzato S, Park YM, Buso N, Lopez R, 2015, The EMBL-EBI bioinformatics web and programmatic tools framework. Nucleic Acids Res 43: W580–W584 Li LC, Dahiya R, 2002, MethPrimer: designing primers for methylation PCRs. Bioinformatics 18:1427–1431 Messeguer X, Escudero R, Farre D, Nunez O, Martinez J, Alba M, 2002, PROMO: detection of known transcription regulatory elements using species-tailored searches. Bioinformatics 18:333–334 Wilkins MR, Gasteiger E, Bairoch A et al, 1999, Protein identification and analysis tools in the ExPASy server. Methods Mol Biol 112:531–552 Geourjon C, Deleage G, 1995, SOPMA: significant improvements in protein secondary structure prediction by consensus prediction from multiple alignments. Comput Appl Biosci : CABIOS 11:681– 684 Garnier J: GOR secondary structure prediction method version IV. Methods Enzymol, RF Doolittle Ed. 266: 540-553, 1998 Petersen TN, Brunak S, von Heijne G, Nielsen H, 2011, SignalP 4.0: discriminating signal peptides from transmembrane regions. Nat Methods 8:785–786 Nguyen Ba AN, Pogoutse A, Provart N, Moses AM, 2009, NLStradamus: a simple hidden Markov model for nuclear localization signal prediction. BMC Bioinf 10:202 Emanuelsson O, Nielsen H, Brunak S, von Heijne G, 2000, Predicting subcellular localization of proteins based on their Nterminal amino acid sequence. J Mol Biol 300:1005–1016 Horton P, Park KJ, Obayashi T, Fujita N, Harada H, Adams-Collier CJ, Nakai K, 2007, WoLF PSORT: protein localization predictor. Nucleic Acids Res 35:W585–W587 Finn RD, Attwood TK, Babbitt PC, Bateman A, Bork P, Bridge AJ, Chang HY, Dosztanyi Z, el-Gebali S, Fraser M, Gough J, Haft D, Holliday GL, Huang H, Huang X, Letunic I, Lopez R, Lu S, Marchler-Bauer A, Mi H, Mistry J, Natale DA, Necci M, Nuka G, Orengo CA, Park Y, Pesseat S, Piovesan D, Potter SC, Rawlings ND, Redaschi N, Richardson L, Rivoire C, Sangrador- Vegas A, Sigrist C, Sillitoe I, Smithers B, Squizzato S, Sutton G, Thanki N, Thomas PD, Tosatto SCE, Wu CH, Xenarios I, Yeh LS, Young SY, Mitchell AL, 2017, InterPro in 2017-beyond protein family and domain annotations. Nucleic Acids Res 45: D190–D199 Krogh A, Larsson B, von Heijne G, Sonnhammer EL, 2001, Predicting transmembrane protein topology with a hidden Markov model: application to complete genomes. J Mol Biol 305: 567–580 Biasini M, Bienert S, Waterhouse A, Arnold K, Studer G, Schmidt T, Kiefer F, Cassarino TG, Bertoni M, Bordoli L, Schwede T, 2014, SWISS-MODEL: modelling protein tertiary and quaternary structure using evolutionary information. Nucleic Acids Res 42:W252–W258 Larkin MA, Blackshields G, Brown NP, Chenna R, McGettigan PA, McWilliam H, Valentin F, Wallace IM, Wilm A, Lopez R, Thompson JD, Gibson TJ, Higgins DG, 2007, Clustal W and Clustal X version 2.0. Bioinformatics 23:2947–2948 Saldanha AJ, 2004, Java Treeview-extensible visualization of microarray data. Bioinformatics 20:3246–3248 Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, Kuhn M, Bork P, Jensen LJ, von Mering C, 2015, STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43:D447–D452 Johnson M, Zaretskaya I, Raytselis Y, Merezhuk Y, McGinnis S, Madden TL, 2008, NCBI BLAST: a better web interface. Nucleic Acids Res 36:W5–W9 Kumar S, Stecher G, Tamura K, 2016, MEGA7: molecular evolutionary genetics analysis version 7.0 for bigger datasets. Mol Biol Evol 33:1870–1874 Kanehisa M, Sato Y, Kawashima M, Furumichi M, Tanabe M, 2016, KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res 44:D457–D462 Hackenberg M, Previti C, Luque-Escamilla PL, Carpena P, Martinez-Aroza J, Oliver JL, 2006, CpGcluster: a distance-based algorithm for CpG-island detection. BMC Bioinf 7:446 Wu Y, Guo Z, Wu H, Wang X, Yang L, Shi X, et al, 2012, SUMOylation represses Nanog expression via modulating transcription factors Oct4 and Sox2. PLoS One 7(6):e39606 Orkin SH, 2005, Chipping away at the embryonic stem cell network. Cell 122:828–830 Zhao L, Zevallos SE, Rizzoti K, Jeong Y, Lovell-Badge R, Epstein DJ, 2012, Disruption of SoxB1-dependent sonic hedgehog expression in the hypothalamus causes septo-optic dysplasia. Dev Cell 22:585–596 Bora-Singhal N, Nguyen J, Schaal C, Perumal D, Singh S, Coppola D, Chellappan S, 2015, YAP1 regulates OCT4 activity and SOX2 expression to facilitate self-renewal and vascular mimicry of stemlike cells. Stem Cells 33:1705–1718 Verma NK, Gadi A, Maurizi G, Roy UB, Mansukhani A, Basilico C, 2017, Myeloid zinc finger 1 and GA binding protein co-operate with Sox2 in regulating the expression of yes-associated protein 1 in cancer cells. Stem Cells 35(12):2340–2350 Miranda CJ, Braun L, Jiang YY, Hester ME, Zhang L, Riolo M, Wang H, Rao M, Altura RA, Kaspar BK, 2012, Aging brain microenvironment decreases hippocampal neurogenesis throughWntmediated survivin signaling. Aging Cell 11:542–552 Seo E, Basu-Roy U, Zavadil J, Basilico C, Mansukhani A, 2011, Distinct functions of Sox2 control self-renewal and differentiation in the osteoblast lineage. Mol Cell Biol 31:4593–4608 Keramari M, Razavi J, Ingman KA, Patsch C, Edenhofer F, Ward CM, et al, 2010, Sox2 is essential for formation of trophectodermin the preimplantation embryo. PLoS One 5(11):e13952 PivaM, Domenici G, Iriondo O, Rabano M, Simoes BM, Comaills V, Barredo I, Lopez-Ruiz JA, Zabalza I, Kypta R, Vivanco MM, 2014, Sox2 promotes tamoxifen resistance in breast cancer cells. EMBO Mol Med 6:66–79 Du J, Li B, Fang Y et al, 2015, Overexpression of class III β- tubulin, Sox2, and nuclear Survivin is predictive of taxane resistance in patients with stage III ovarian epithelial cancer. BMC Cancer 15:536 Li D, Zhao L-N, Zheng X-L et al, 2014, Sox2 is involved in paclitaxel resistance of the prostate cancer cell line PC-3 via the PI3K/ Akt pathway. Mol Med Rep 10:3169–3176 Wen W, Han T, Chen C, Huang L, Sun W, Wang X, Chen SZ, Xiang DM, Tang L, Cao D, Feng GS, Wu MC, Ding J, Wang HY, 2013, Cyclin G1 expands liver tumor-initiating cells by Sox2 induction via Akt/mTOR signaling. Mol Cancer Ther 12:1796–1804 Chou M-Y, Hu F-W, Yu C-H, Yu C-C, 2015, Sox2 expression involvement in the oncogenicity and radiochemoresistance of oral cancer stem cells. Oral Oncol 51:31–39 Rudin CM, Durinck S, Stawiski EW, Poirier JT,Modrusan Z, Shames DS, Bergbower EA, Guan Y, Shin J, Guillory J, Rivers CS, Foo CK, Bhatt D, Stinson J, Gnad F, Haverty PM, Gentleman R, Chaudhuri S, Janakiraman V, Jaiswal BS, Parikh C, YuanW, Zhang Z, Koeppen H, Wu TD, Stern HM, Yauch RL, Huffman KE, Paskulin DD, Illei PB, Varella-Garcia M, Gazdar AF, de Sauvage FJ, Bourgon R, Minna JD, BrockMV, Seshagiri S, 2012, Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat Genet 44:1111–1116 Chen S, Li X, Lu D et al, 2013, SOX2 regulates apoptosis through MAP4K4-survivin signaling pathway in human lung cancer cells. Carcinogenesis 35:613–623 Ji J, Zheng PS, 2010, Expression of Sox2 in human cervical carcinogenesis. Hum Pathol 41:1438–1447 Huang X, XiongM, Jin Yet al, 2016, Evidence that high-migration drug-surviving MOLT4 leukemia cells exhibit cancer stem cell-like properties. Int J Oncol 49:343–351 Wang L, Yang H, Lei Z, Zhao J, Chen Y, Chen P, Li C, Zeng Y, Liu Z, Liu X, Zhang HT, 2016, Repression of TIF1gamma by SOX2 promotes TGF-beta-induced epithelial-mesenchymal transition in non-small-cell lung cancer. Oncogene 35:867–877 Tornin J, Martinez-Cruzado L, Santos L et al, 2016, Inhibition of SP1 by the mithramycin analog EC-8042 efficiently targets tumor initiating cells in sarcoma. Oncotarget 7:30935–30950 Marques-Torrejon MA, Porlan E, Banito A et al, 2013, Cyclindependent kinase inhibitor p21 controls adult neural stem cell expansion by regulating Sox2 gene expression. Cell Stem Cell 12:88–100 Alonso MM, Diez-Valle R, Manterola L, Rubio A, Liu D, Cortes- Santiago N, Urquiza L, Jauregi P, de Munain AL, Sampron N, Aramburu A, Tejada-Solis S, Vicente C, Odero MD, Bandres E, Garcia-Foncillas J, IdoateMA, Lang FF, Fueyo J, Gomez-Manzano C, 2011, Genetic and epigenetic modifications of Sox2 contribute to the invasive phenotype of malignant gliomas. PLoS One 6: e26740 Li X, Wang YK, Song ZQ, Du ZQ, Yang CX, 2016, Dimethyl sulfoxide perturbs cell cycle progression and spindle Organization in Porcine Meiotic Oocytes. PLoS One 11:e0158074 Jung K, Wu F, Wang P, Ye X, Abdulkarim BS, Lai R, 2014, YB-1 regulates Sox2 to coordinately sustain stemness and tumorigenic properties in a phenotypically distinct subset of breast cancer cells. BMC Cancer 14:328 Chien CS, Wang ML, Chu PY, Chang YL, Liu WH, Yu CC, Lan YT, Huang PI, Lee YY, Chen YW, Lo WL, Chiou SH, 2015, Lin28B/Let-7 regulates expression of Oct4 and Sox2 and reprograms oral squamous cell carcinoma cells to a stem-like state. Cancer Res 75:2553–2565 WuQQ, Zhang LS, Su P, Lei X, Liu X,Wang H, Lu L, BaiY, Xiong T, Li D, Zhu Z, Duan E, Jiang E, Feng S, Han M, Xu Y, Wang F, Zhou J, 2015, MSX2 mediates entry of human pluripotent stem cells into mesendoderm by simultaneously suppressing SOX2 and activating NODAL signaling. Cell Res 25:1314–1332 Lee Y, Kim KH, Kim DG, Cho HJ, Kim Y, Rheey J, Shin K, Seo YJ, Choi YS, Lee JI, Lee J, Joo KM, Nam DH, 2015, FoxM1 promotes stemness and radio-resistance of glioblastoma by regulating the master stem cell regulator Sox2. PLoS One 10:e0137703 Archer TC, Jin J, Casey ES, 2011, Interaction of Sox1, Sox2, Sox3 and Oct4 during primary neurogenesis. Dev Biol 350:429–440 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 693 EP 706 DI 10.1007/s12253-019-00581-9 PG 14 ER PT J AU Li, X Xu, X Chen, K Wu, H Wang, Y Yang, Sh Wang, K AF Li, Xinwei Xu, Xueying Chen, Keng Wu, Haijian Wang, Yirong Yang, Shuxu Wang, Kun TI miR-370 Sensitizes TMZ Response Dependent of MGMT Status in Primary Central Nervous System Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Primary central nervous system lymphoma; miR-370; MGMT; Temozolomide; Drug resistance ID Primary central nervous system lymphoma; miR-370; MGMT; Temozolomide; Drug resistance AB Primary central nervous system lymphoma (PCNSL) is an aggressive and rare subtype of non-Hodgkin lymphoma, arising exclusively in the CNS with a poor prognosis. Previous evidence has proved that MGMT was a promising target involving in TMZ resistance of PCNSL. Our study described a new miR-370-mediated mechanism of MGMT regulation in PCNSL. We first showed that miR-370 was downregulated in PCNSL tissues, while MGMT was inversely overexpressed. It was also observed that miR-370 suppressed the expression of MGMT. Additionally, upregulation of miR-370 significantly increased TMZ sensitivity dependent of MGMT, thus suppressed Raji cell proliferation and induced apoptosis in vitro. In conclusion, these results suggest that miR-370 is a potential target in PCNSL treatment. C1 [Li, Xinwei] Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. [Xu, Xueying] Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. [Chen, Keng] Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. [Wu, Haijian] Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. [Wang, Yirong] Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. [Yang, Shuxu] Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. [Wang, Kun] Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. RP Wang, K (reprint author), Zhejiang University, Medical College, Sir Run Run Shaw Hospital, Department of Neurosurgery, 310016 Hangzhou, China. EM simonkunwang@zju.edu.cn CR Batchelor T, Loeffler JS, 2006, Primary CNS lymphoma. J Clin Oncol 24(8):1281–1288 Korfel A, Schlegel U, 2013, Diagnosis and treatment of primary CNS lymphoma. Nat Rev Neurol 9(6):317–327 Olson JE, Janney CA, Rao RD, Cerhan JR, Kurtin PJ, Schiff D, Kaplan RS, O'Neill BP, 2002, The continuing increase in the incidence of primary central nervous system non-Hodgkin lymphoma: a surveillance, epidemiology, and end results analysis. Cancer 95(7):1504–1510 Ferreri AJ, Verona C, Politi LS, Chiara A, Perna L, Villa E, ReniM, 2011, Consolidation radiotherapy in primary central nervous system lymphomas: impact on outcome of different fields and doses in patients in complete remission after upfront chemotherapy. Int J Radiat Oncol Biol Phys 80(1):169–175 Moise L, Matta C, Hanna C, Pilorge S, Ferme C, Durrbach A, RibragV, 2011)Methotrexate- and/or cytarabine-based chemotherapy may be effective and safe in solid-organ transplant recipients with primary central nervous system lymphomas. Leuk Lymphoma 52(3):521–524 Shi X, Zhang X, Yi C, Wang X, Chen Z, Zhang B, 2013, The combination of 13N-ammonia and 18F-FDG in predicting primary central nervous system lymphomas in immunocompetent patients. Clin Nucl Med 38(2):98–102 Mead GM, Bleehen NM, Gregor A, Bullimore J, Shirley D, Rampling RP, Trevor J, Glaser MG, Lantos P, Ironside JW, Moss TH, Brada M, Whaley JB, Stenning SP, 2000, A medical research council randomized trial in patients with primary cerebral non- Hodgkin lymphoma: cerebral radiotherapy with and without cyclophosphamide, doxorubicin, vincristine, and prednisone chemotherapy. Cancer 89(6):1359–1370 Panageas KS, Elkin EB, DeAngelis LM, Ben-Porat L, Abrey LE, 2005, Trends in survival from primary central nervous system lymphoma, 1975–1999: a population-based analysis. Cancer 104(11):2466–2472 Herrlinger U, Kuker W, Platten M, 2002, First-line therapy with temozolomide induces regression of primary CNS lymphoma. Neurology 58(10):1573–1574 Reni M, Mason W, Zaja F, Perry J, Franceschi E, Bernardi D, Dell'Oro S, Stelitano C, Candela M, Abbadessa A, Pace A, Bordonaro R, Latte G, Villa E, Ferreri AJ, 2004, Salvage chemotherapy with temozolomide in primary CNS lymphomas: preliminary results of a phase II trial. Eur J Cancer 40(11):1682–1688 ReniM, Ferreri AJ, Landoni C, Villa E, 2000, Salvage therapy with temozolomide in an immunocompetent patient with primary brain lymphoma. J Natl Cancer Inst 92(7):575–576 Strik HM, Spreer A, Nagel H, Jacob S, Jung W, Kitze B, Bahr M, 2004, Clinical response following adjuvant temozolomide in a patient with primary cerebral lymphoma. Anticancer Res 24(6): 4121–4125 Reni M, Zaja F, Mason W, Perry J, Mazza E, Spina M, Bordonaro R, Ilariucci F, Faedi M, Corazzelli G, Manno P, Franceschi E, Pace A, Candela M, Abbadessa A, Stelitano C, Latte G, Ferreri AJ, 2007, Temozolomide as salvage treatment in primary brain lymphomas. Br J Cancer 96(6):864–867 Wang K, Xie D, Xie J,Wan Y, Ma L, Qi X, Yang S, 2015, MiR-27a regulates Wnt/beta-catenin signaling through targeting SFRP1 in glioma. Neuroreport 26(12):695–702 Wang K, Wang X, Zou J, Zhang A, Wan Y, Pu P, Song Z, Qian C, Chen Y, Yang S, Wang Y, 2013, miR-92b controls glioma proliferation and invasion through regulatingWnt/beta-catenin signaling via nemo-like kinase. Neuro-Oncology 15(5):578–588 Yang S,Wang K, Qian C, Song Z, Pu P, Zhang A,WangW, Niu H, Li X, Qi X, Zhu Y, Wang Y, 2012, A predicted miR-27a-mediated network identifies a signature of glioma. Oncol Rep 28(4):1249– 1256 Nass D, Rosenwald S, Meiri E, Gilad S, Tabibian-Keissar H, Schlosberg A, Kuker H, Sion-Vardy N, Tobar A, Kharenko O, Sitbon E, Lithwick Yanai G, Elyakim E, Cholakh H, Gibori H, Spector Y, Bentwich Z, Barshack I, Rosenfeld N, 2009, MiR-92b and miR-9/9* are specifically expressed in brain primary tumors and can be used to differentiate primary from metastatic brain tumors. Brain Pathol 19(3):375–383 DeAngelis LM, 2015, Neuro-oncology: primary CNS lymphoma treatment-the devil is in the details. Nat Rev Neurol 11(6):314–315 Omuro AM, Taillandier L, Chinot O, Carnin C, Barrie M, Hoang- Xuan K, 2007, Temozolomide and methotrexate for primary central nervous system lymphoma in the elderly. J Neuro-Oncol 85(2): 207–211 Reni M, Mazza E, Foppoli M, Ferreri AJ, 2007, Primary central nervous system lymphomas: salvage treatment after failure to high dose methotrexate. Cancer Lett 258(2):165–170 Kurzwelly D, Glas M, Roth P, Weimann E, Lohner H, Waha A, SchabetM, Reifenberger G,Weller M, HerrlingerU, 2010, Primary CNS lymphoma in the elderly: temozolomide therapy and MGMT status. J Neuro-Oncol 97(3):389–392 Enting RH, Demopoulos A, DeAngelis LM, Abrey LE, 2004, Salvage therapy for primary CNS lymphoma with a combination of rituximab and temozolomide. Neurology 63(5):901–903 Omuro AM, Taillandier L, Chinot O, Carnin C, Barrie M, Hoang- Xuan K, 2007, Temozolomide and methotrexate for primary central nervous system lymphoma in the elderly. J Neuro-Oncol 85(2): 207–211 YungangW, Xiaoyu L, Pang T,Wenming L, Pan X(2014)miR-370 targeted FoxM1 functions as a tumor suppressor in laryngeal squamous cell carcinoma, LSCC). Biomed Pharmacother 68(2):149– 154 Chen XP, Chen YG, Lan JY, Shen ZJ, 2014, MicroRNA-370 suppresses proliferation and promotes endometrioid ovarian cancer chemosensitivity to cDDP by negatively regulating ENG. Cancer Lett 353(2):201–210 Feng Y,Wang L, Zeng J, Shen L, Liang X, Yu H, Liu S, Liu Z, Sun Y, Li W, Chen C, Jia J, 2013, FoxM1 is overexpressed in helicobacter pylori-induced gastric carcinogenesis and is negatively regulated by miR-370. Mol Cancer Res 11(8):834–844 Zhang X, Zeng J, Zhou M, Li B, Zhang Y, Huang T,Wang L, Jia J, Chen C, 2012, The tumor suppressive role of miRNA-370 by targeting FoxM1 in acute myeloid leukemia. Mol Cancer 11:56 Fischer L, Hummel M, Korfel A, Lenze D, Joehrens K, Thiel E, 2011, Differential micro-RNA expression in primary CNS and nodal diffuse large B-cell lymphomas. Neuro-Oncology 13(10): 1090–1098 Zheng J, Xu J, Ma S, Sun X, Geng M, Wang L, 2013, Clinicopathological study of gene rearrangement and microRNA expression of primary central nervous system diffuse large B-cell lymphomas. Int J Clin Exp Pathol 6(10):2048–2055 Robertus JL, Harms G, Blokzijl T, Booman M, de Jong D, van Imhoff G, Rosati S, Schuuring E, Kluin P, van den Berg A, 2009, Specific expression of miR-17-5p and miR-127 in testicular and central nervous system diffuse large B-cell lymphoma. Mod Pathol 22(4):547–555 Sun G, Hou YB, Jia HY, Bi XH, Yu L, Chen DJ, 2016, MiR-370 promotes cell death of liver cancer cells by Akt/FoxO3a signalling pathway. Eur Rev Med Pharmacol Sci 20(10):2011–2019 Cao X, Liu D, Yan X, Zhang Y, Yuan L, Zhang T, Fu M, Zhou Y, Wang J, 2013, Stat3 inhibits WTX expression through upregulation of microRNA-370 in Wilms tumor. FEBS Lett 587(6): 639–644 Gao Y, Chen X, Liu H, 2016, Up-regulation of miR-370-3p restores glioblastoma multiforme sensitivity to temozolomide by influencing MGMT expression. Sci Rep 6:32972 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 707 EP 714 DI 10.1007/s12253-019-00605-4 PG 8 ER PT J AU Mehemmai, Ch Cherbal, F Hamdi, Y Guedioura, A Benbrahim, W Bakour, R Abdelhak, S AF Mehemmai, Chiraz Cherbal, Farid Hamdi, Yosr Guedioura, Abdelmoumene Benbrahim, Wassila Bakour, Rabah Abdelhak, Sonia TI BRCA1 and BRCA2 Germline Mutation Analysis in Hereditary Breast/Ovarian Cancer Families from the Aures Region (Eastern Algeria): First Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Algerian women; Aures region; HBOC; BRCA1; BRCA2; Genetic testing; NGS; Cancer panel ID Algerian women; Aures region; HBOC; BRCA1; BRCA2; Genetic testing; NGS; Cancer panel AB Breast cancer is currently the leading cause of cancer morbidity and mortality among Algerian women. In this study, we aimed to investigate the mutation spectrum of BRCA1 and BRCA2 genes in hereditary breast/ovarian cancer (HBOC) families from the Aures region (eastern Algeria). High risk breast/ovarian cancer families were selected from overall 1162 consecutive patients collected from cancer registry of anticancer center of Batna. Breast cancers were diagnosed between 2011 and 2015. Recurrent mutations on BRCA1 and BRCA2 previously found in Algerian patients were screened using PCR-direct sequencing in 113 HBOC families. In addition, for the first time in Algeria, HBOC patients were analyzed by NGS using a cancer panel of 30 hereditary cancer genes or BRCA1/2 genetic test. Six distinct deleterious mutations in BRCA1 and BRCA2 and a new VUS in PALB2 were detected in ten patients. Two distinct BRCA2 pathogenic variants c.1813dupA and c.8485C > T detected in two young female triple negative breast cancer (TNBC) patients, respectively, with a family history of male breast cancer, are reported here for the first time in Algerian population. Interestingly, we also detected a BRCA exon 15 deletion in two unrelated young female TNBC patients with strong family history of breast/ovarian cancer. Our study showed differences in the distribution of the mutation spectrum of BRCA genes between the Aures region and the north central region of Algeria. Our results will contribute in the implementation of genetic counseling and testing for patients and families at risk of hereditary breast and ovarian cancer. C1 [Mehemmai, Chiraz] USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, El Alia, Bab Ezzouar, 16111 Algiers, Algeria. [Cherbal, Farid] USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, El Alia, Bab Ezzouar, 16111 Algiers, Algeria. [Hamdi, Yosr] University of Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Biomedical Genomics and Oncogenetics (LRTI, IPT 05)Tunis, Tunisia. [Guedioura, Abdelmoumene] USTHB, Faculty of Biological Sciences, LOBEMAlgiers, Algeria. [Benbrahim, Wassila] Anti-cancer center of BatnaBatna, Algeria. [Bakour, Rabah] USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, El Alia, Bab Ezzouar, 16111 Algiers, Algeria. [Abdelhak, Sonia] University of Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Biomedical Genomics and Oncogenetics (LRTI, IPT 05)Tunis, Tunisia. RP Cherbal, F (reprint author), USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, 16111 Algiers, Algeria. EM farid.cherbal@gmail.com CR Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, et al., 2013, GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11 [Internet]. Lyon, France: International Agency for Research on Cancer. Available from: http://globocan.iarc.fr. Accessed on 12/1/2018 GLOBOCAN, 2012, Breast cancer incidence and mortality worldwide, 2012. International agency for research on cancer. http:// globocan.iarc.fr/Pages/fact_sheets_population.aspx. Accessed 4 Jan 2018 Perou CM, Sorlie T, Eisen MB, Van de Rijn M, Jeffrey SS, Rees CA et al, 2000)Molecular portraits of human breast tumors. Nature 406:747–752 Sotiriou C, Neo SY, McShane LM, Korn EL, Long PM, Jazaeri A et al, 2003, Breast cancer classification and prognosis based on gene expression profiles from a population-based study. Proc Natl Acad Sci U S A 100:10393–10398 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, Panel members, 2011, Strategies for subtypes—dealing with the diversity of breast cancer: highlights of the St Gallen international expert consensus on the primary therapy of early breast Cancer 2011. Ann Oncol 22:1736–1747. , DOI 10.1093/ annonc/mdr304 Cherbal F, Gaceb H, Mehemmai C, Saiah I, Bakour R, Rouis AO, Boualga K, Benbrahim W, Mahfouf H, 2015, Distribution of molecular breast cancer subtypes among Algerian women and correlation with clinical and tumor characteristics: a population-based study. Breast Dis 35(2):95–102. , DOI 10.3233/BD- 150398 Kuchenbaecker KB, Hopper JL, Barnes DR, Phillips KA, Mooij TM, Roos-Blom MJ, Jervis S, van Leeuwen FE, Milne RL, Andrieu N, Goldgar DE, Terry MB, Rookus MA, Easton DF, Antoniou AC, and the BRCA1 and BRCA2 Cohort Consortium, McGuffog L, Evans DG, Barrowdale D, Frost D, Adlard J, Ong KR, Izatt L, Tischkowitz M, Eeles R, Davidson R, Hodgson S, Ellis S, Nogues C, Lasset C, Stoppa-Lyonnet D, Fricker JP, Faivre L, Berthet P, Hooning MJ, van der Kolk LE, Kets CM, Adank MA, John EM, Chung WK, Andrulis IL, Southey M, Daly MB, Buys SS, Osorio A, Engel C, Kast K, Schmutzler RK, Caldes T, Jakubowska A, Simard J, Friedlander ML, McLachlan SA, Machackova E, Foretova L, Tan YY, Singer CF, Olah E, Gerdes AM, Arver B, Olsson H, 2017, BRCA1 and BRCA2 cohort consortium. Risks of breast, ovarian, and contralateral breast cancer for BRCA1 and BRCA2 mutation carriers. JAMA 317(23):2402–2416. , DOI 10.1001/jama.2017.7112 Purnomosari D, Pals G, Wahyono A, Aryandono T, Manuaba TW, Haryono SJ, van Diest PJ, 2007, BRCA1 and BRCA2 germline mutation analysis in the Indonesian population. Breast Cancer Res Treat 106(2):297–304 Villarreal-Garza C, Weitzel JN, Llacuachaqui M, Sifuentes E, Magallanes-Hoyos MC, Gallardo L, Alvarez-Gomez RM, Herzog J, Castillo D, Royer R, Akbari M, Lara-Medina F, Herrera LA, Mohar A, Narod SA, 2015, The prevalence of BRCA1 and BRCA2 mutations among young Mexican women with triplenegative breast cancer. Breast Cancer Res Treat 150(2):389–394. , DOI 10.1007/s10549-015-3312-8 Yang XR, Devi BCR, Sung H, Guida J, Mucaki EJ, Xiao Y, Best A, Garland L, Xie Y, Hu N, Rodriguez-Herrera M, Wang C, Jones K, Luo W, Hicks B, Tang TS, Moitra K, Rogan PK, Dean M, 2017, Prevalence and spectrum of germline rare variants in BRCA1/2 and PALB2 among breast cancer cases in Sarawak, Malaysia. Breast Cancer Res Treat 165(3):687–697. , DOI 10.1007/ s10549-017-4356-8 Palmero EI, Carraro DM, Alemar B et al, 2018, The germline mutational landscape of BRCA1 and BRCA2 in Brazil. Sci Rep 8(1):9188. , DOI 10.1038/s41598-018-27315-2 Zheng Y,Walsh T, Gulsuner S, Casadei S, LeeMK, Ogundiran TO, Ademola A, Falusi AG, Adebamowo CA, Oluwasola AO, Adeoye A, Odetunde A, Babalola CP, Ojengbede OA, Odedina S, Anetor I, Wang S, Huo D, Yoshimatsu TF, Zhang J, Felix GES, King MC, Olopade OI, 2018, Inherited breast Cancer in Nigerian women. J Clin Oncol 36(28):2820–2825. , DOI 10.1200/JCO.2018. 78.3977 Bekada A, Arauna LR, Deba T, Calafell F, Benhamamouch S, Comas D, 2015, Genetic heterogeneity in Algerian human populations. PLoS One 10(9):e0138453. , DOI 10.1371/journal. pone.0138453 Cherbal F, Bakour R, Adane S, Boualga K, 2012, BRCA1 and BRCA2 germline mutation spectrum in hereditary breast/ovarian cancer families from Maghrebian countries. Breast Dis 34:1–8 Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, eds,, 2010, AJCC Cancer staging manual, 7th edn. Springer, New York Hammond ME, Hayes DF, Wolff AC, Mangu PB, Temin S, 2010, American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Oncol Pract 6:195–197 Pennacchia I, Carbone A, Di Cerbo A, Vecchio FM, Arena V, 2015, 2013 ASCO/CAP updated guidelines for human epidermal growth factor receptor 2 testing: impact on routine practice. Breast 24:285e6 Cherbal F, Bakour R, Adane S, Boualga K, Benais-Pont G, Maillet P, 2010, BRCA1 and BRCA2 germline mutations screening in Algerian breast/ovarian cancer families. Dis Markers 28:377–384 Engert S, Wappenschmidt B, Betz B, Kast K, Kutsche M, Hellebrand H, Goecke TO, Kiechle M, Niederacher D, Schmutzler RK, Meindl A, 2008, MLPA screening in the BRCA1 gene from 1,506 German hereditary breast cancer cases: novel deletions, frequent involvement of exon 17, and occurrence in single early-onset cases. Hum Mutat 29:948–958 Tai YC, Domchek S, Parmigiani G, Chen S, 2007, Breast cancer risk among male BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 99(23):1811–1814 Silvestri V, Barrowdale D, Mulligan AM et al, 2016, Male breast cancer in BRCA1 and BRCA2 mutation carriers: pathology data from the consortium of investigators of modifiers of BRCA1/2. Breast Cancer Res 18(1):15. , DOI 10.1186/s13058-016- 0671-y Janavicius R, 2010, Founder BRCA1/2mutations in the Europe: implications for hereditary breast-ovarian cancer prevention and control. EPMA J 1, 397–412(3) Gonzalez-Angulo AM, Timms KM, Liu S, Chen H, Litton JK, Potter J, Lanchbury JS, Stemke-Hale K, Hennessy BT, Arun BK, Hortobagyi GN, Do KA, Mills GB, Meric-Bernstam F, 2011, Incidence and outcome of BRCA mutations in unselected patients with triple receptor-negative breast cancer. Clin Cancer Res 17: 1082–1089 Robertson L, Hanson H, Seal S, Warren-Perry M, Hughes D, Howell I, Turnbull C, Houlston R, Shanley S, Butler S, Evans DG, Ross G, Eccles D, Tutt A, Rahman N, TNT Trial TMG., BCSC, UK,, 2012, BRCA1 testing should be offered to individuals with triple-negative breast cancer diagnosed below 50 years. Br J Cancer 106:1234–1238 Meyer P, Landgraf K, Hogel B, Eiermann W, Ataseven B, 2012, BRCA2 mutations and triple- negative breast cancer. PLoS One 7(5):e38361. , DOI 10.1371/journal.pone. 0038361 Hartman AR, Kaldate RR, Sailer LM, Painter L, Grier CE, Endsley RR, Griffin M, Hamilton SA, Frye CA, Silberman MA, Wenstrup RJ, Sandbach JF, 2012, Prevalence of BRCA mutations in an unselected population of triple-negative breast cancer. Cancer 118: 2787–2782 Couch FJ, Hart SN, Sharma P, Toland AE, Wang X, Miron P, Olson JE, Godwin AK, Pankratz VS, Olswold C, Slettedahl S, Hallberg E, Guidugli L, Davila JI, Beckmann MW, Janni W, Rack B, Ekici AB, Slamon DJ, Konstantopoulou I, Fostira F, Vratimos A, Fountzilas G, Pelttari LM, Tapper WJ, Durcan L, Cross SS, Pilarski R, Shapiro CL, Klemp J, Yao S, Garber J, Cox A, Brauch H, Ambrosone C, Nevanlinna H, Yannoukakos D, Slager SL, Vachon CM, Eccles DM, Fasching PA, 2014, Inherited mutations in 17 breast cancer susceptibility genes among a large triple negative breast cancer cohort unselected for family history of breast cancer. J Clin Oncol 33:304–311. , DOI 10.1200/JCO.2014.57.1414 Wong-Brown MW, Meldrum CJ, Carpenter JE et al, 2015, Prevalence of BRCA1 and BRCA2 germline mutations in patients with triple-negative breast cancer. Breast Cancer Res Treat 50:71– 80 Hahnen E, Hauke J, Engel C, Neidhardt G, Rhiem K, Schmutzler RK, 2017, Germline mutations in triplenegative breast Cancer. Breast Care, Basel, 1:15–19. , DOI 10.1159/000455999 Chen H, Wu J, Zhang Z et al, 2018, Association between BRCA status and triple-negative breast Cancer: a meta-analysis. Front Pharmacol 9:909. , DOI 10.3389/fphar. 2018.00909 Cherbal F, Gaceb H,Mehemmai C, Bakour R, Yatta K, Boukoufa I, Mahfouf H, Boualga K, 2016, Abstract 2553 : prevalence of BRCA1 germline mutations in Algerian population: implications for genetic testing and counseling. Cancer Res 76(14 Suppl): 2553. , DOI 10.1158/1538-7445.AM2016-2553 Troudi W, Uhrhammer N, Sibille C, Dahan C, Mahfoudh W, Bouchlaka Souissi C, Jalabert T, Chouchane L, Bignon YJ, Ben Ayed F, Ben Ammar Elgaaied A, 2007, Contribution of the BRCA1 and BRCA2 mutations to breast cancer in Tunisia. J Hum Genet 52: 915–920 Tazzite A, Jouhadi H, Nadifi S, Aretini P, Falaschi E, Collavoli A, Benider A, Caligo MA, 2012, BRCA1 and BRCA2 germline mutations in Moroccan breast/ovarian cancer families: novel mutations and unclassified variants. Gynecol Oncol 125(3):687–692. https:// doi.org/10.1016/j.ygyno.2012.03.007 Blay P, Santamaria I, Pitiot AS, LuqueM, Alvarado MG, Lastra A, Fernandez Y, Paredes A, Freije JMP, Balbin M, 2013, Mutational analysis of BRCA1 and BRCA2 in hereditary breast and ovarian cancer families from Asturias, northern Spain). BMC Cancer 13: 243. , DOI 10.1186/1471-2407-13-243 Krajc M, Zadnik V, Novakovic S, Stegel V, Teugels E, Besic N, Hocevar M, Vakselj A, de Greve J, Zgajnar J, 2014, Geographical distribution of Slovenian BRCA1/2 families according to family origin: implications for genetic screening. Clin Genet 85(1):59– 63. , DOI 10.1111/cge.12119 Jara L, Morales S, de Mayo T, Gonzalez-Hormazabal P, Carrasco V, Godoy R, 2017)Mutations in BRCA1, BRCA2 and other breast and ovarian cancer susceptibility genes in central and south American populations. Biol Res 50(1):35. , DOI 10.1186/s40659- 017-0139-2 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 715 EP 726 DI 10.1007/s12253-019-00586-4 PG 12 ER PT J AU Mervai, Zs Reszegi, A Miklya, I Knoll, J Schaff, Zs Kovalszky, I Baghy, K AF Mervai, Zsolt Reszegi, Andrea Miklya, Ildiko Knoll, Jozsef Schaff, Zsuzsa Kovalszky, Ilona Baghy, Kornelia TI Inhibitory Effect of (2R)-1-(1-Benzofuran-2-yl)-N-propylpentan-2-amine on Lung Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BPAP; Lung adenocarcinoma; Cancer; Tumor inhibition; FVB/N; Geroconversion ID BPAP; Lung adenocarcinoma; Cancer; Tumor inhibition; FVB/N; Geroconversion AB BPAP is a potent enhancer substance with catecholaminergic and serotoninergic activity in the brain. It was discovered that it is also effective against certain types of experimental cancers, showing the most promising results in case of lung cancer. That is why we tested its efficacy in two different doses in a newly developed EGFR wild type mouse lung adenocarcinoma xenograft model. Experiments were conducted on FVB/N and SCID mouse strains treated with low and high dose of BPAP. Body weight, survival, and tumor volumes were recorded. Furthermore, the activity of major signaling pathways of NSCLC such as MAPK and Akt/mTOR as well as cell cycle regulation were determined. Significant inhibition of tumor growth was exerted by both doses, but the mechanism of action was different. High dose directly inhibited, whereas low dose activated the main signaling pathways. Exposure to low dose BPAP resulted in elevated activity of the mTOR pathway together with p16INK-induced cell cycle arrest, a typical feature of geroconversion, a senescent state characterized by loss of cell proliferation. Finally the events culminated in cell cycle inhibition point in case of both doses mirrored by the decrease of cyclin D1, CDK4 and PCNA. In addition, BPAP treatment had a beneficial effect on bodyweight suggesting that the compound at least in part is able to compensate the cancer-related wasting. In view of the low toxicity and confirmed antitumor effect of BPAP against experimental lung adenocarcinoma, this novel compound deserves further attention. C1 [Mervai, Zsolt] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Reszegi, Andrea] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Miklya, Ildiko] Semmelweis University, Department of Pharmacology and PharmacotherapyBudapest, Hungary. [Knoll, Jozsef] Semmelweis University, Department of Pharmacology and PharmacotherapyBudapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Baghy, Kornelia] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. RP Baghy, K (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM baghy.kornelia@med.semmelweis-univ.hu CR Knoll J, Magyar K, 1972, Some puzzling effects of monoamine oxidase inhibitors. Adv Biochem Psychopharmacol 5:393–408 Heinonen EH, Myllyla V, 1998, Safety of selegiline, deprenyl, in the treatment of Parkinson's disease. Drug Saf 19:11–22 Knoll J, 1988, The striatal dopamine dependency of life span in male rats. Longevity study with, −)deprenyl.Mech Ageing Dev 46: 237–262 Freisleben HJ, Lehr F, Fuchs J, 1994, Lifespan of immunosuppressed NMRI-mice is increased by deprenyl. J Neural Transm Suppl 41:231–236 Ruehl WW, Entriken TL, Muggenburg BA, Bruyette DS, Griffith WC, Hahn FF, 1997, Treatment with L-deprenyl prolongs life in elderly dogs. Life Sci 61:1037–1044 Knoll J, Knoll B, Torok Z, Timar J, Yasar S, 1992, The pharmacology of 1-phenyl-2-propylamino-pentane, PPAP), a deprenylderived new spectrum psychostimulant. Arch Int Pharmacodyn Ther 316:5–29 Knoll J, Yoneda F, Knoll B, Ohde H, Miklya I, 1999,, −)1-, Benzofuran-2-yl)-2-propylaminopentane, [(−)BPAP], a selective enhancer of the impulse propagation mediated release of catecholamines and serotonin in the brain. Br J Pharmacol 128:1723–1732 Maruyama W, Yi H, Takahashi T, Shimazu S, Ohde H, Yoneda F, Iwasa K, Naoi M, 2004, Neuroprotective function of R-(−)-1-, benzofuran-2-yl)-2-propylaminopentane, [R-(−)-BPAP], against apoptosis induced by N-methyl(R)salsolinol, an endogenous dopaminergic neurotoxin, in human dopaminergic neuroblastoma SHSY5Y cells. Life Sci 75:107–117 Gaszner P, Miklya I, 2006, Major depression and the synthetic enhancer substances,, −)-deprenyl and R-(−)-1-(benzofuran-2-yl)- 2-propylaminopentane. Prog Neuro-Psychopharmacol Biol Psychiatry 30:5–14 Knoll J, Miklya I, 2016, Longevity study with low doses of selegiline/(−)-deprenyl and, 2R)-1-(1-benzofuran-2-yl)-Npropylpentane- 2-amine, BPAP). Life Sci 167:32–38 Knoll J, Baghy K, Eckhardt S, Ferdinandy P, Garami M, Harsing LG Jr, Hauser P, Mervai Z, Pocza T, Schaff Z, Schuler D, Miklya I, 2017, A longevity study with enhancer substances, selegiline, BPAP, detected an unknown tumor-manifestation-suppressing regulation in rat brain. Life Sci 182:57–64 Mervai Z, Egedi K, Kovalszky I , Baghy K, 2018, Diethylnitrosamine induces lung adenocarcinoma in FVB/N mouse. BMC Cancer 18:157 Siegel R, Ma J, Zou Z, Jemal A, 2014, Cancer statistics, 2014. CA Cancer J Clin 64:9–29 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D, Bray F, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403 Molina JR, Yang P, Cassivi SD, Schild SE, Adjei AA, 2008, Nonsmall cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc 83:584–594 Subramanian J, Govindan R, 2007, Lung cancer in never smokers: a review. J Clin Oncol Off J Am Soc Clin Oncol 25:561–570 Mudduluru G, Walther W, Kobelt D, Dahlmann M, Treese C, Assaraf YG, Stein U, 2016, Repositioning of drugs for intervention in tumor progression and metastasis: old drugs for new targets. Drug Resist Updat 26:10–27 Bradford MM, 1976, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72:248–254 Chan BA, Hughes BG, 2015, Targeted therapy for non-small cell lung cancer: current standards and the promise of the future. Transl Lung Cancer Res 4:36–54 Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC et al, 2004, Activating mutations in the epidermal growth factor receptor underlying responsiveness of nonsmall- cell lung cancer to gefitinib. N Engl J Med 350:2129–2139 Pallis AG, 2012, A review of treatment in non-small-cell lung Cancer. Eur Oncol Haematol 8(4):208–212 Bosma MJ, Carroll AM, 1991, The SCID mouse mutant: definition, characterization, and potential uses. Annu Rev Immunol 9: 323–350 Brambilla E, Gazdar A, 2009, Pathogenesis of lung cancer signalling pathways: roadmap for therapies. Eur Respir J 33:1485–1497 Yip PY, 2015, Phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin, PI3K-Akt-mTOR, signaling pathway in nonsmall cell lung cancer. Transl Lung Cancer Res 4:165–176 Mendoza MC, Er EE, Blenis J, 2011, The Ras-ERK and PI3KmTOR pathways: cross-talk and compensation. Trends Biochem Sci 36:320–328 Fumarola C, Bonelli MA, Petronini PG, Alfieri RR, 2014, Targeting PI3K/AKT/mTOR pathway in non small cell lung cancer. Biochem Pharmacol 90:197–207 Blagosklonny MV, 2014, Geroconversion: irreversible step to cellular senescence. Cell Cycle 13:3628–3635 Demidenko ZN, Blagosklonny MV, 2008, Growth stimulation leads to cellular senescence when the cell cycle is blocked. Cell Cycle 7:3355–3361 Blagosklonny MV, 2012, Cell cycle arrest is not yet senescence, which is not just cell cycle arrest: terminology for TOR-driven aging. Aging, Albany NY, 4:159–165 Yoshida A, Diehl JA, 2015, CDK4/6 inhibitor: from quiescence to senescence. Oncoscience 2:896–897 Kovatcheva M, Liu DD, Dickson MA, Klein ME, O'Connor R, Wilder FO, Socci ND, Tap WD, Schwartz GK, Singer S, Crago AM, Koff A, 2015, MDM2 turnover and expression of ATRX determine the choice between quiescence and senescence in response to CDK4 inhibition. Oncotarget 6:8226–8243 Kollmann K, Heller G, Sexl V, 2011, c-JUN prevents methylation of p16(INK4a,, and Cdk6): the villain turned bodyguard. Oncotarget 2:422–427 Wisdom R, Johnson RS, Moore C, 1999, c-Jun regulates cell cycle progression and apoptosis by distinct mechanisms. EMBO J 18: 188–197 Szabo E, Riffe ME, Steinberg SM, Birrer MJ, Linnoila RI, 1996, Altered cJUN expression: an early event in human lung carcinogenesis. Cancer Res 56:305–315 Laplante M, Sabatini DM, 2012, mTOR signaling in growth control and disease. Cell 149:274–293 Saxton RA, Sabatini DM, 2017, mTOR signaling in growth, metabolism, and disease. Cell 169:361–371 KumarNB, Kazi A, Smith T, Crocker T,Yu D, Reich RR, Reddy K, Hastings S, Exterman M, Balducci L, Dalton K, Bepler G, 2010, Cancer cachexia: traditional therapies and novel molecular mechanism-based approaches to treatment. Curr Treat Options in Oncol 11:107–117 ZhouW, Jiang ZW, Tian J, Jiang J, Li N, Li JS, 2003, Role of NFkappaB and cytokine in experimental cancer cachexia. World J Gastroenterol 9:1567–1570 Denlinger CE, Rundall BK, Jones DR, 2004, Modulation of antiapoptotic cell signaling pathways in non-small cell lung cancer: the role of NF-kappaB. Semin Thorac Cardiovasc Surg 16:28–39 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 727 EP 734 DI 10.1007/s12253-019-00603-6 PG 8 ER PT J AU Naruse, T Yanamoto, S Okuyama, K Ohmori, K Tsuchihashi, H Furukawa, K Yamada, Shi Umeda, M AF Naruse, Tomofumi Yanamoto, Souichi Okuyama, Kohei Ohmori, Keisuke Tsuchihashi, Hiroki Furukawa, Kohei Yamada, Shin-ichi Umeda, Masahiro TI Immunohistochemical Study of PD-1/PD-L1 Axis Expression in Oral Tongue Squamous Cell Carcinomas: Effect of Neoadjuvant Chemotherapy on Local Recurrence SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PD-1; PD-L1; Local recurrence; NAC ID PD-1; PD-L1; Local recurrence; NAC AB While neoadjuvant chemotherapy (NAC) for patients with oral tongue squamous cell carcinoma (OTSCC) may improve tumor microenvironment, it may lead to local immune suppression caused by residual cancer cells. The efficacy of NAC is therefore controversial. In our study, we investigated tumor microenvironments after NAC using immune checkpoint molecules, and evaluated the association between tumor microenvironments, clinicopathological factors and outcomes. We reviewed the records of 121 patients who underwent radical surgery for OTSCC between April 2001 and March 2015. Patients with a positive surgical margin and a follow up period of less than 6 months were excluded. For these patients, programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1) expressions were immunohistochemically examined. The expression of PD-1 and PD-L1 were significantly associated with local recurrence in patients with OTSCC (P < 0.01 and P < 0.01, respectively). We found a significant decrease in 5-year disease specific survival rate for patients with combined PD-1+/PD-L1+ expressions (P < 0.05). In the subgroup analysis of local recurrence between the NAC treated group and those who received surgery alone, high levels of PD-1 and PD-L1 expressions were significantly found in the former, but not in the latter group. Local recurrence in the NAC treated group may contribute to local immune suppression in OTSCC. NAC lead to local immune suppression and immune checkpoint molecules play an important role in local recurrence in patients with OTSCC who received NAC. NAC modality can’t be recommended for patients with OTSCC at present. C1 [Naruse, Tomofumi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yanamoto, Souichi] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Okuyama, Kohei] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Ohmori, Keisuke] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Tsuchihashi, Hiroki] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Furukawa, Kohei] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. [Yamada, Shin-ichi] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 390-8621 Matsumoto, Nagano, Japan. [Umeda, Masahiro] Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 1-7-1 Sakamoto, 852-8588 Nagasaki, Japan. RP Naruse, T (reprint author), Nagasaki University, Graduate School of Biomedical Sciences, Department of Clinical Oral Oncology, 852-8588 Nagasaki, Japan. EM naruse12@nagasaki-u.ac.jp CR Jemal A, Bray F, Center MM et al, 2011, Global Cancer statistics: 2011. CA Cancer J Clin 61:69–90 Listl S, Jansen L, Stenzinger A et al, 2013, Survival of patients with oral cavity cancer in Germany. PLoS One 8:e53415 Rogers SN, Brown JS,Woolgar JA et al, 2009, Survival following primary surgery for oral cancer. Oral Oncol 45:201–211 National Comprehensive Cancer Network Clinical practice guidelines in oncology, 2017, Head and neck cancers, Version 2. https:// www.nccn.org/professionals/physician_gls/pdf/head-and-neck. pdf/. Accessed 31th January, 2018 Umeda M, Komatsubara H, Ojima Yet al, 2004, Lack of survival advantage in patients with advanced, resectable squamous cell carcinoma of the oral cavity receiving induction chemotherapy with cisplatin, CDDP), docetaxel, TXT, and 5-fluorouracil, 5FU). Kobe J Med Sci 50:189–196 Licitra L, Grandi C, GuzzoMet al, 2003, Primary chemotherapy in resectable oral cavity squamous cell cancer: a randomized controlled trial. J Clin Oncol 21:327–333 Sakuishi K, Apetoh L, Sullivan JM et al, 2010, Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore antitumor immunity. J Exp Med 207:2187–2194 Zandberg DP, Strome SE, 2014, The role of the PD-L1:PD-1 pathway in squamous cell carcinoma of the head and neck. Oral Oncol 50:627–632 Kerkar SP, Restifo NP, 2012, Cellular constituents of immune escape within the tumormicroenvironment. Cancer Res 72:3125–3130 Yanamoto S, Yamada S, Takahashi H et al, 2014, Expression of the cancer stem cell markers CD44v6 and ABCG2 in tongue cancer: effect of neoadjuvant chemotherapy on local recurrence. Int J Oncol 44:1153–1162 Peng J, Hamanishi J, Matsumura N et al, 2015, Chemotherapy induces programmed cell death-ligand 1 overexpression via the nuclear factor-κB to foster an immunosuppressive tumor microenvironment in ovarian Cancer. Cancer Res 75:5034–5045 Patel SG, Shah JP, 2005, TNM staging of cancers of the head and neck: striving for uniformity among diversity. CA Cancer J Clin 55: 242–258 Bryne M, Koppang HS, Lilleng R et al, 1992, Malignancy grading of the deep invasive margins of oral squamous cell carcinoma has high prognostic value. J Pathol 166:375–381 Hirakawa H, Hanai N, Ozawa T et al, 2016, Prognostic impact of pathological response to neoadjuvant chemotherapy followed by definitive surgery in sinonasal squamous cell carcinoma. Head Neck 47:1305–1311 Troeltzsch M,Woodlock T, Pianka A et al, 2017, Is there evidence for the presence and relevance of the PD-1/PD-L1 pathway in Oral squamous cell carcinoma? Hints from an Immunohistochemical study. J Oral Maxillofac Surg 75:969–977 Maruse Y, Kawano S, Jinno Tet al, 2018, Significant association of increased PD-L1 and PD-1 expression with nodal metastasis and a poor prognosis in oral squamous cell carcinoma. Int J Oral Maxillofac Surg 47:836–845 Wang B, Zhang S, Yue K, Wang X-D, 2013, The recurrence and survival of oral squamous cell carcinoma: a report of 275 cases. Chin J Cancer 32:614–618 Zhou Y, Miao J, Wu H et al, 2017, PD-1 and PD-L1 expression in 132 recurrent nasopharyngeal carcinoma: the correlation with anemia and outcomes. Oncotarget 8:51210–51223 Saigusa S, Toiyama Y, Tanaka K et al, 2016, Implication of programmed cell death ligand 1 expression in tumor recurrence and prognosis in rectal cancer with neoadjuvant chemoradiotherapy. Int J Clin Oncol 21:946–952 Muller T, Braun M, Dietrich D et al, 2017, PD-L1: a novel prognostic biomarker in head and neck squamous cell carcinoma. Oncotarget 8:52889–52900 Lin YM, SungWW, HsiehMJ et al, 2015, High PD-L1 expression correlates with metastasis and poor prognosis in oral squamous cell carcinoma. PLoS One 10:1–11 Hirai M,Kitahara H,Kobayashi Yet al, 2017, Regulation of PD-L1 expression in a high-grade invasive human oral squamous cell carcinoma microenvironment. Int J Oncol 50:41–48 Hanna GJ,Woo SB, Li YYet al, 2017, Tumor PD-L1 expression is associated with improved survival and lower recurrence risk in young women with oral cavity squamous cell carcinoma. Int J Oral Maxillofac Surg pii S0901-5027:31616–31618 Strati A, Koutsodontis G, Papaxoinis G, Angelidis I, Zavridou M, Economopoulou P, Kotsantis I, AvgerisM, Mazel M, Perisanidis C, Sasaki C, Alix-Panabieres C, Lianidou E, Psyrri A, 2017, Prognostic significance of PD-L1 expression on circulating tumor cells in patients with head and neck squamous cell carcinoma. Ann Oncol 28:1923–1933 Ock C-Y, Kim S, Keam B, Kim S, Ahn YO, Chung EJ, Kim JH, Kim TM, Kwon SK, Jeon YK, Jung KC, Kim DW, Wu HG, Sung MW, Heo DS, 2017, Changes in programmed death-ligand 1 expression during cisplatin treatment in patients with head and neck squamous cell carcinoma. Oncotarget 8:97920–97927 Shalapour S, Font-Burgada J, Di Caro G et al, 2015, Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy. Nature 521:94–98 Braakhuis BJM, Bloemena E, Leemans CR, Brakenhoff RH, 2010, Molecular analysis of surgical margins in head and neck cancer: more than a marginal issue. Oral Oncol 46:485–491 Ferris RL, Blumenschein G, Fayette J et al, 2016, Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med 375:1856–1867 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 735 EP 742 DI 10.1007/s12253-019-00606-3 PG 8 ER PT J AU Vukovic, V Karan-Djurasevic, T Antic, D Tosic, N Kostic, T Marjanovic, I Dencic-Fekete, M Djurasinovic, V Pavlovic, S Mihaljevic, B AF Vukovic, Vojin Karan-Djurasevic, Teodora Antic, Darko Tosic, Natasa Kostic, Tatjana Marjanovic, Irena Dencic-Fekete, Marija Djurasinovic, Vladislava Pavlovic, Sonja Mihaljevic, Biljana TI Association of SLC28A3 Gene Expression and CYP2B6*6 Allele with the Response to Fludarabine Plus Cyclophosphamide in Chronic Lymphocytic Leukemia Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chronic lymphocytic leukemia; Fludarabine; Cyclophosphamide; SLC28A3 expression; CYP2B6*6 allele; Response to therapy ID Chronic lymphocytic leukemia; Fludarabine; Cyclophosphamide; SLC28A3 expression; CYP2B6*6 allele; Response to therapy AB Fludarabine plus cyclophosphamide (FC) chemotherapy is the basis of treatment protocols used in management of chronic lymphocytic leukemia (CLL). In some patients, response to therapy may be affected by aberrant function of genes involved in pharmacokinetics and pharmacodynamics of the drugs. The aim of this research was to assess the impact of pharmacogenetic variability, namely expression of SLC28A3 gene and the presence of CYP2B6*6 variant allele, on the FC treatment efficacy. Forty-four CLL patients with functional TP53 gene at the time of FC initiation were enrolled in this study. CYP2B6 genotyping was performed by polymerase chain reaction and direct sequencing. SLC28A3 expression was measured by quantitative reverse transcriptase polymerase chain reaction. Significantly higher pretreatment levels of SLC28A3 mRNA were detected in patients who failed to respond to FC in comparison to patients who achieved complete and partial response (p = 0.01). SLC28A3 high expressing cases were almost ten times more likely not to respond to FC than low-expressing cases (OR = 9.8; p = 0.046). However, association of SLC28A3 expression with progression-free survival (PFS) and overall survival (OS) was not observed. CYP2B6*6 allele, detected in 24 patients (54.6%), exerted no association with the attainment of response to FC, as well as with PFS and OS. The results of this study demonstrate that SLC28A3 expression is a significant predictor of FC efficacy in CLL patients with intact TP53. Elevated SLC28A3 mRNA levels are associated with inferior short-term response to FC, suggesting that, if validated on larger cohorts, SLC28A3 expression may become a biomarker useful for pretreatment stratification of patients. C1 [Vukovic, Vojin] Clinical Center of Serbia, Clinic for HematologyBelgrade, Serbia. [Karan-Djurasevic, Teodora] University of Belgrade, Institute of Molecular Genetics and Genetic EngineeringBelgrade, Serbia. [Antic, Darko] Clinical Center of Serbia, Clinic for HematologyBelgrade, Serbia. [Tosic, Natasa] University of Belgrade, Institute of Molecular Genetics and Genetic EngineeringBelgrade, Serbia. [Kostic, Tatjana] University of Belgrade, Institute of Molecular Genetics and Genetic EngineeringBelgrade, Serbia. [Marjanovic, Irena] University of Belgrade, Institute of Molecular Genetics and Genetic EngineeringBelgrade, Serbia. [Dencic-Fekete, Marija] University of Belgrade, Faculty of Medicine, Institute for PathologyBelgrade, Serbia. [Djurasinovic, Vladislava] Clinical Center of Serbia, Clinic for HematologyBelgrade, Serbia. [Pavlovic, Sonja] University of Belgrade, Institute of Molecular Genetics and Genetic EngineeringBelgrade, Serbia. [Mihaljevic, Biljana] Clinical Center of Serbia, Clinic for HematologyBelgrade, Serbia. RP Vukovic, V (reprint author), Clinical Center of Serbia, Clinic for Hematology, Belgrade, Serbia. EM vojinvukovic@yahoo.com CR Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G, Dohner H, Hillmen P, KeatingMJ,Montserrat E, Rai KR, Kipps TJ International Workshop on Chronic Lymphocytic L, 2008, Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the international workshop on chronic lymphocytic leukemia updating the National Cancer Institute-working group 1996 guidelines. Blood 111(12):5446–5456. , DOI 10.1182/blood-2007-06-093906 Grever MR, Lucas DM, Dewald GW, Neuberg DS, Reed JC, Kitada S, Flinn IW, Tallman MS, Appelbaum FR, Larson RA, Paietta E, Jelinek DF, Gribben JG, Byrd JC, 2007, Comprehensive assessment of genetic and molecular features predicting outcome in patients with chronic lymphocytic leukemia: results from the US intergroup phase III trial E2997. J Clin Oncol 25(7):799–804. , DOI 10.1200/JCO.2006.08.3089 Catovsky D, Richards S,Matutes E, Oscier D, DyerM, Bezares RF, Pettitt AR, Hamblin T, Milligan DW, Child JA, Hamilton MS, Dearden CE, Smith AG, Bosanquet AG, Davis Z, Brito- Babapulle V, Else M, Wade R, Hillmen P, Group UKNCRIHOCS, Group NCLLW, 2007, Assessment of fludarabine plus cyclophosphamide for patients with chronic lymphocytic leukaemia, the LRF CLL4 trial): a randomised controlled trial. Lancet 370(9583):230–239. , DOI 10.1016/S0140- 6736(07)61125-8 Eichhorst BF, Busch R, Hopfinger G, Pasold R, Hensel M, Steinbrecher C, Siehl S, Jager U, Bergmann M, Stilgenbauer S, Schweighofer C, Wendtner CM, Dohner H, Brittinger G, Emmerich B, Hallek M, German CLLSG, 2006, Fludarabine plus cyclophosphamide versus fludarabine alone in first-line therapy of younger patients with chronic lymphocytic leukemia. Blood 107(3):885–891. , DOI 10.1182/blood-2005-06-2395 Hallek M, Fischer K, Fingerle-Rowson G, Fink AM, Busch R, Mayer J, Hensel M, Hopfinger G, Hess G, von Grunhagen U, Bergmann M, Catalano J, Zinzani PL, Caligaris-Cappio F, Seymour JF, Berrebi A, Jager U, Cazin B, Trneny M, Westermann A, Wendtner CM, Eichhorst BF, Staib P, Buhler A, Winkler D, Zenz T, Bottcher S, Ritgen M, Mendila M, Kneba M, Dohner H, Stilgenbauer S, International Group of I, German Chronic Lymphocytic Leukaemia Study G, 2010, Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial. Lancet 376(9747):1164–1174. , DOI 10.1016/ S0140-6736(10)61381-5 Ghielmini M, Vitolo U, Kimby E, Montoto S, Walewski J, Pfreundschuh M, Federico M, Hoskin P, McNamara C, Caligaris- Cappio F, Stilgenbauer S, Marcus R, Trneny M, Dreger P, Montserrat E, Dreyling M, Panel Members of the 1st ECCoML, 2013, ESMO guidelines consensus conference on malignant lymphoma 2011 part 1: diffuse large B-cell lymphoma, DLBCL), follicular lymphoma, FL, and chronic lymphocytic leukemia, CLL). Ann Oncol 24(3):561–576. , DOI 10.1093/annonc/ mds517 Committee EG, 2017, Appendix 4: chronic lymphocytic leukaemia: eUpdate published online 27 June 2017, www.esmo.org/ Guidelines/Haematological-Malignancies). Ann Oncol 28, suppl_ 4):iv149-iv152. , DOI 10.1093/annonc/mdx242 Flinn IW, Neuberg DS, Grever MR, Dewald GW, Bennett JM, Paietta EM, Hussein MA, Appelbaum FR, Larson RA, Moore DF Jr, Tallman MS, 2007, Phase III trial of fludarabine plus cyclophosphamide compared with fludarabine for patients with previously untreated chronic lymphocytic leukemia: US intergroup trial E2997. J Clin Oncol 25(7):793–798. , DOI 10.1200/JCO. 2006.08.0762 Robak T, Dmoszynska A, Solal-Celigny P, Warzocha K, Loscertales J, Catalano J, Afanasiev BV, Larratt L, Geisler CH, Montillo M, Zyuzgin I, Ganly PS, Dartigeas C, Rosta A, Maurer J, Mendila M, Saville MW, Valente N, Wenger MK, Moiseev SI, 2010, Rituximab plus fludarabine and cyclophosphamide prolongs progression-free survival compared with fludarabine and cyclophosphamide alone in previously treated chronic lymphocytic leukemia. J Clin Oncol 28(10):1756–1765. , DOI 10.1200/ JCO.2009.26.4556 Edelmann J, Gribben JG, 2017, Managing patients with TP53- deficient chronic lymphocytic leukemia. J Oncol Pract 13(6):371– 377. , DOI 10.1200/JOP.2017.023291 Zenz T, Eichhorst B, Busch R, Denzel T, Habe S, Winkler D, Buhler A, Edelmann J, Bergmann M, Hopfinger G, Hensel M, Hallek M, Dohner H, Stilgenbauer S, 2010, TP53 mutation and survival in chronic lymphocytic leukemia. J Clin Oncol 28(29): 4473–4479. , DOI 10.1200/JCO.2009.27.8762 Rossi D, CerriM, Deambrogi C, Sozzi E, Cresta S, Rasi S, De Paoli L, Spina V, Gattei V, Capello D, Forconi F, Lauria F, Gaidano G, 2009, The prognostic value of TP53 mutations in chronic lymphocytic leukemia is independent of Del17p13: implications for overall survival and chemorefractoriness. Clin Cancer Res 15(3):995– 1004. , DOI 10.1158/1078-0432.CCR-08-1630 Malcikova J, Smardova J, Rocnova L, Tichy B, Kuglik P, Vranova V, Cejkova S, Svitakova M, Skuhrova Francova H, Brychtova Y, Doubek M, Brejcha M, Klabusay M, Mayer J, Pospisilova S, Trbusek M, 2009, Monoallelic and biallelic inactivation of TP53 gene in chronic lymphocytic leukemia: selection, impact on survival, and response to DNA damage. Blood 114(26):5307–5314. , DOI 10.1182/blood-2009-07-234708 Dohner H, Stilgenbauer S, Benner A, Leupolt E, Krober A, Bullinger L, Dohner K, Bentz M, Lichter P, 2000, Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med 343(26):1910–1916. h t t p s : / / d o i . o rg/10.1056/ NEJM200012283432602 Zenz T, Habe S, Denzel T, Mohr J,Winkler D, Buhler A, Sarno A, Groner S, Mertens D, Busch R, Hallek M, Dohner H, Stilgenbauer S, 2009, Detailed analysis of p53 pathway defects in fludarabinerefractory chronic lymphocytic leukemia, CLL): dissecting the contribution of 17p deletion, TP53 mutation, p53-p21 dysfunction, and miR34a in a prospective clinical trial. Blood 114(13):2589–2597. , DOI 10.1182/blood-2009-05-224071 Landau DA, Tausch E, Taylor-Weiner AN, Stewart C, Reiter JG, Bahlo J, Kluth S, Bozic I, Lawrence M, Bottcher S, Carter SL, Cibulskis K, Mertens D, Sougnez CL, Rosenberg M, Hess JM, Edelmann J, Kless S, Kneba M, Ritgen M, Fink A, Fischer K, Gabriel S, Lander ES, Nowak MA, Dohner H, Hallek M, Neuberg D, Getz G, Stilgenbauer S,Wu CJ, 2015, Mutations driving CLL and their evolution in progression and relapse. Nature 526(7574):525–530. , DOI 10.1038/nature15395 Galmarini CM, Mackey JR, Dumontet C, 2001, Nucleoside analogues: mechanisms of drug resistance and reversal strategies. Leukemia 15(6):875–890 Emadi A, Jones RJ, Brodsky RA, 2009, Cyclophosphamide and cancer: golden anniversary. Nat Rev Clin Oncol 6(11):638–647. , DOI 10.1038/nrclinonc.2009.146 Young JD, 2016, The SLC28, CNT, and SLC29, ENT, nucleoside transporter families: a 30-year collaborative odyssey. Biochem Soc Trans 44(3):869–876. , DOI 10.1042/BST20160038 Molina-Arcas M, Bellosillo B, Casado FJ, Montserrat E, Gil J, Colomer D, Pastor-Anglada M, 2003, Fludarabine uptake mechanisms in B-cell chronic lymphocytic leukemia. Blood 101(6):2328– 2334. , DOI 10.1182/blood-2002-07-2236 Mackey JR, Galmarini CM, Graham KA, Joy AA, Delmer A, Dabbagh L, Glubrecht D, Jewell LD, Lai R, Lang T, Hanson J, Young JD, Merle-Beral H, Binet JL, Cass CE, Dumontet C, 2005, Quantitative analysis of nucleoside transporter and metabolismgene expression in chronic lymphocytic leukemia, CLL): identification of fludarabine-sensitive and -insensitive populations. Blood 105(2):767–774. , DOI 10.1182/blood-2004-03- 1046 Roy P, Yu LJ, Crespi CL, Waxman DJ, 1999, Development of a substrate-activity based approach to identify the major human liver P-450 catalysts of cyclophosphamide and ifosfamide activation based on cDNA-expressed activities and liver microsomal P-450 profiles. Drug Metab Dispos 27(6):655–666 Zanger UM, Klein K, 2013, Pharmacogenetics of cytochrome P450 2B6, CYP2B6): advances on polymorphisms, mechanisms, and clinical relevance. Front Genet 4:24. , DOI 10.3389/ fgene.2013.00024 Desta Z, Saussele T, Ward B, Blievernicht J, Li L, Klein K, Flockhart DA, Zanger UM, 2007, Impact of CYP2B6 polymorphism on he p a t i c efavirenz metabolism i n vi t r o . Pharmacogenomics 8(6):547–558. , DOI 10.2217/ 14622416.8.6.547 HofmannMH, Blievernicht JK, Klein K, Saussele T, Schaeffeler E, Schwab M, Zanger UM, 2008, Aberrant splicing caused by single nucleotide polymorphism c.516G>T [Q172H], a marker of CYP2B6*6, is responsible for decreased expression and activity of CYP2B6 in liver. J Pharmacol Exp Ther 325(1):284–292. , DOI 10.1124/jpet.107.133306 Tsuchiya K, Gatanaga H, Tachikawa N, Teruya K, Kikuchi Y, Yoshino M, Kuwahara T, Shirasaka T, Kimura S, Oka S, 2004, Homozygous CYP2B6 *6, Q172H and K262R, correlates with high plasma efavirenz concentrations in HIV-1 patients treated with standard efavirenz-containing regimens. Biochem Biophys Res Commun 319(4):1322–1326. , DOI 10.1016/j.bbrc.2004. 05.116 Lang T, Klein K, Fischer J, Nussler AK, Neuhaus P, Hofmann U, Eichelbaum M, Schwab M, Zanger UM, 2001, Extensive genetic polymorphism in the human CYP2B6 gene with impact on expression and function in human liver. Pharmacogenetics 11(5):399–415 Zhang H, Sridar C, Kenaan C, Amunugama H, Ballou DP, Hollenberg PF, 2011, Polymorphic variants of cytochrome P450 2B6, CYP2B6.4-CYP2B6.9, exhibit altered rates of metabolism for bupropion and efavirenz: a charge-reversal mutation in the K139E variant, CYP2B6.8, impairs formation of a functional cytochrome p450-reductase complex. J Pharmacol Exp Ther 338(3): 803–809. , DOI 10.1124/jpet.111.183111 Ariyoshi N, Ohara M, Kaneko M, Afuso S, Kumamoto T, Nakamura H, Ishii I, Ishikawa T, KitadaM(2011, Q172H replacement overcomes effects on the metabolism of cyclophosphamide and efavirenz caused by CYP2B6 variant with Arg262. Drug Metab Dispos 39(11):2045–2048. , DOI 10.1124/dmd. 111.039586 Xu C, Ogburn ET, GuoY, Desta Z, 2012, Effects of the CYP2B6*6 allele on catalytic properties and inhibition of CYP2B6 in vitro: implication for the mechanism of reduced efavirenz metabolism and other CYP2B6 substrates in vivo. Drug Metab Dispos 40(4): 717–725. , DOI 10.1124/dmd.111.042416 Xie HJ, Yasar U, Lundgren S, Griskevicius L, Terelius Y, Hassan M, Rane A, 2003, Role of polymorphic human CYP2B6 in cyclophosphamide bioactivation. Pharmacogenomics J 3(1):53–61. , DOI 10.1038/sj.tpj.6500157 Johnson GG, Lin K, Cox TF, Oates M, Sibson DR, Eccles R, Lloyd B, Gardiner LJ, Carr DF, PirmohamedM, Strefford JC, Oscier DG, Gonzalez de Castro D, Else M, Catovsky D, Pettitt AR, 2013, CYP2B6*6 is an independent determinant of inferior response to fludarabine plus cyclophosphamide in chronic lymphocytic leukemia. Blood 122(26):4253–4258. , DOI 10.1182/blood- 2013-07-516666 Cheson BD, Bennett JM, GreverM, Kay N,Keating MJ, O'Brien S, Rai KR, 1996, National Cancer Institute-sponsored working group guidelines for chronic lymphocytic leukemia: revised guidelines for diagnosis and treatment. Blood 87(12):4990–4997 Pospisilova S, Gonzalez D, Malcikova J, Trbusek M, Rossi D, Kater AP, Cymbalista F, Eichhorst B, Hallek M, Dohner H, Hillmen P, van Oers M, Gribben J, Ghia P, Montserrat E, Stilgenbauer S, Zenz T, European Research Initiative on CLL, 2012, ERIC recommendations on TP53 mutation analysis in chronic lymphocytic leukemia. Leukemia 26(7):1458–1461. , DOI 10.1038/leu.2012.25 Wood BL, ArrozM, Barnett D, DiGiuseppe J, Greig B, Kussick SJ, Oldaker T, Shenkin M, Stone E, Wallace P, 2007, 2006 Bethesda i n t e r n a t i on a l consensus recommendations on the immunophenotypic analysis of hematolymphoid neoplasia by flow cytometry: optimal reagents and reporting for the flow cytometric diagnosis of hematopoietic neoplasia. Cytometry B Clin Cytom 72(Suppl 1):S14–S22. , DOI 10.1002/cyto.b.20363 Davis BH, Holden JT, Bene MC, Borowitz MJ, Braylan RC, Cornfield D, Gorczyca W, Lee R, Maiese R, Orfao A, Wells D, Wood BL, Stetler-StevensonM(2007, 2006 Bethesda international consensus recommendations on the flow cytometric immunophenotypic analysis of hematolymphoid neoplasia: medical indications. Cytometry B Clin Cytom 72 Suppl 1:S5–13. https:// doi.org/10.1002/cyto.b.20365 Ghia P, Stamatopoulos K, Belessi C, Moreno C, Stilgenbauer S, Stevenson F, Davi F, Rosenquist R, European Research Initiative on CLL, 2007, ERIC recommendations on IGHV gene mutational status analysis in chronic lymphocytic leukemia. Leukemia 21(1): 1–3. , DOI 10.1038/sj.leu.2404457 Rosenquist R, Cortese D, Bhoi S, Mansouri L, Gunnarsson R, 2013, Prognostic markers and their clinical applicability in chronic lymphocytic leukemia: where do we stand? Leuk Lymphoma 54(11):2351–2364. , DOI 10.3109/10428194.2013. 783913 Stilgenbauer S, 2015, Prognostic markers and standard management of chronic lymphocytic leukemia. Hematology Am Soc Hematol Educ Program 2015:368–377. , DOI 10.1182/ asheducation-2015.1.368 Montserrat E, Dreger P, 2016, Treatment of chronic lymphocytic leukemia with del(17p)/TP53 mutation: allogeneic hematopoietic stem cell transplantation or BCR-signaling inhibitors? Clin Lymphoma Myeloma Leuk 16(Suppl):S74–S81. , DOI 10.1016/j.clml.2016.02.013 Justenhoven C, Pentimalli D, Rabstein S, Harth V, Lotz A, Pesch B, Bruning T, Dork T, Schurmann P, Bogdanova N, Park-Simon TW, Couch FJ, Olson JE, Fasching PA, Beckmann MW, Haberle L, Ekici A, Hall P, Czene K, Liu J, Li J, Baisch C, Hamann U, Ko YD, Brauch H, 2014, CYP2B6*6 is associated with increased breast cancer risk. Int J Cancer 134(2):426–430. , DOI 10.1002/ijc.28356 Berkoz M, Yalin S, 2009, Association of CYP2B6 G15631T polymorphism with acute leukemia susceptibility. Leuk Res 33(7):919– 923. , DOI 10.1016/j.leukres.2008.11.014 Yuan ZH, Liu Q, Zhang Y, Liu HX, Zhao J, Zhu P, 2011, CYP2B6 gene single nucleotide polymorphisms and leukemia susceptibility. Ann Hematol 90(3):293–299. , DOI 10.1007/s00277-010- 1085-z Daraki A, Zachaki S, Koromila T, Diamantopoulou P, Pantelias GE, Sambani C, Aleporou V, Kollia P, Manola KN, 2014, The G(5)(1)(6)T CYP2B6 germline polymorphism affects the risk of acutemyeloid leukemia and is associated with specific chromosomal abnormalities. PLoS One 9(2):e88879. , DOI 10.1371/ journal.pone.0088879 Daraki A, Kakosaiou K, Zachaki S, Sambani C, Aleporou-Marinou V, Kollia P, Manola KN, 2016, Polymorphisms and haplotypes of the CYP2B6 detoxification gene in the predisposition of acute myeloid leukemia, AML, and induction of its cytogenetic abnormalities. Cancer Genet 209(11):525–533. , DOI 10.1016/j. cancergen.2016.10.004 Gandhi M, Aweeka F, Greenblatt RM, Blaschke TF, 2004, Sex differences in pharmacokinetics and pharmacodynamics. Annu Rev Pharmacol Toxicol 44:499–523. , DOI 10.1146/ annurev.pharmtox.44.101802.121453 Lamba V, Lamba J, Yasuda K, Strom S, Davila J, Hancock ML, Fackenthal JD, Rogan PK, Ring B, Wrighton SA, Schuetz EG, 2003, Hepatic CYP2B6 expression: gender and ethnic differences and relationship to CYP2B6 genotype and CAR, constitutive androstane receptor, expression. J Pharmacol Exp Ther 307(3): 906–922. , DOI 10.1124/jpet.103.054866 Ren S, Kalhorn TF, McDonald GB, Anasetti C, Appelbaum FR, Slattery JT, 1998, Pharmacokinetics of cyclophosphamide and its metabolites in bone marrow transplantation patients. Clin Pharmacol Ther 64(3):289–301. , DOI 10.1016/S0009- 9236(98)90178-3 Chang TK, Yu L, Maurel P, Waxman DJ, 1997, Enhanced cyclophosphamide and ifosfamide activation in primary human hepatocyte cultures: response to cytochrome P-450 inducers and autoinduction by oxazaphosphorines. Cancer Res 57(10):1946– 1954 McCune JS, Salinger DH, Vicini P, Oglesby C, Blough DK, Park JR, 2009, Population pharmacokinetics of cyclophosphamide and metabolites in children with neuroblastoma: a report from the Children's oncology group. J Clin Pharmacol 49(1):88–102. , DOI 10.1177/0091270008325928 Tsang RY, Santos C, Ghosh S, Dabbagh L, King K, Young J, Cass CE, Mackey JR, Lai R, 2008, Immunohistochemistry for human concentrative nucleoside transporter 3 protein predicts fludarabine sensitivity in chronic lymphocytic leukemia. Mod Pathol 21(11): 1387–1393. , DOI 10.1038/modpathol.2008.110 Fernandez-Calotti P, Pastor-Anglada M, 2010, All-trans-retinoic acid promotes trafficking of human concentrative nucleoside transporter-3, hCNT3, to the plasma membrane by a TGF-beta1- mediated mechanism. J Biol Chem 285(18):13589–13598. https:// doi.org/10.1074/jbc.M109.055673 Fernandez-Calotti PX, Lopez-Guerra M, Colomer D, Pastor- Anglada M, 2012, Enhancement of fludarabine sensitivity by alltrans- retinoic acid in chronic lymphocytic leukemia cells. Haematologica 97(6):943–951. , DOI 10.3324/haematol. 2011.051557 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 743 EP 752 DI 10.1007/s12253-019-00613-4 PG 10 ER PT J AU Jia, Y Tian, Y An, Sh Yang, D AF Jia, Ying Tian, Ye An, Shuo Yang, Dong TI Effects of microRNA-195 on the Prognosis of Glioma Patients and the Proliferation and Apoptosis of Human Glioma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microRNA-195; Glioma; Prognosis; Cell cycle; Cell proliferation; Apoptosis ID microRNA-195; Glioma; Prognosis; Cell cycle; Cell proliferation; Apoptosis AB Glioma is the most common and aggressive intracranial malignant tumor with poor prognosis. Acts as a tumor suppressor, microRNA-195 (miR-195) plays important roles in a variety of cancers. However, the expression of miR-195 and role of miR- 195 in glioma are still not well understood. 186 patients with glioma were enrolled and the follow-up period ranges from 1 to 69months. MiR-195 was exogenously transfected into human glioma U87 cell line. The cell proliferation assay (CCK-8), colony formation assay, cell cycle analysis and cell apoptosis analysis were examined to investigate miR-195 effect on U87 cells. MiR- 195 levels were reversely correlated with pathological grades (r = −0.487, p = 0.003). For patients with low miR-195 levels, their median survival time was 15 months, whereas the median survival time in patients with high miR-195 levels was 56.53 months. Multi-factor Cox regression analysis showed that high level of miR-195 (Odds ratio (OR): 0.347, 95% CI: 0.121–0.992) was associated with decreased mortality risk of patients. Moreover, overexpression of miR-195 inhibits proliferation and colony formation, and induces apoptosis of U87 cells. MiR-195 could block the glioma cells in G0/G1 phase, reducing S phase cells and regulating apoptosis related proteins (Caspase-3, Caspase-8, Caspase-9 and Bcl-2). Downregulation of miR-195 was associated with poor prognosis in human glioma. MiR-195 acted as tumor suppressor through inhibiting cell proliferation and promoting cell apoptosis via blockade of cell cycle and regulation of apoptosis related proteins. C1 [Jia, Ying] Tianjin Medical University General Hospital, Department of Neurosurgery, 300052 Tianjin, China. [Tian, Ye] Tianjin Medical University General Hospital, Department of Neurosurgery, 300052 Tianjin, China. [An, Shuo] Tianjin Medical University General Hospital, Department of Neurosurgery, 300052 Tianjin, China. [Yang, Dong] Tianjin Medical University General Hospital, Department of Otorhinolaryngology, No.154 An Shan Dao, Heping District, 300052 Tianjin, China. RP Yang, D (reprint author), Tianjin Medical University General Hospital, Department of Otorhinolaryngology, 300052 Tianjin, China. EM yangdong_dr@163.com CR Li K, Lu D, Guo Y, Wang C, Liu X, Liu Y, Liu D, 2018, Trends and patterns of incidence of diffuse glioma in adults in the United States, 1973-2014. Cancer Med 7:5281–5290. , DOI 10.1002/cam4.1757 Xia Z, Qiu D, Deng J, Jiao X, Yang R, Sun Z, Wan X, Li J, 2018, Methylation-induced downregulation and tumor-suppressive role of microRNA-98 in glioma through targeting Sal-like protein 4. Int J Mol Med 41(5):2651–2659. , DOI 10.3892/ijmm.2018.3464 Siegel RL, Miller KD, Jemal A, 2018, Cancer statistics, 2018. CA Cancer J Clin 68(1):7–30. , DOI 10.3322/caac.21442 Ahir BK, Ozer H, Engelhard HH, Lakka SS, 2017, MicroRNAs in glioblastoma pathogenesis and therapy: a comprehensive review. Crit Rev Oncol Hematol 120:22–33. , DOI 10.1016/j. critrevonc.2017.10.003 Barciszewska AM, 2016, MicroRNAs as efficient biomarkers in high-grade gliomas. Folia Neuropathol 54(4):369–374. https://doi. org/10.5114/fn.2016.64812 Bartel DP, 2004, MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116(2):281–297 Zhang F, MengW, Tong B, 2016, Down-regulation of MicroRNA- 133b suppresses apoptosis of Lens epithelial cell by up-regulating BCL2L2 in age-related cataracts. Med Sci Monit 22:4139–4145 Zhang Y, Peng Z, Zhao Y, Chen L, 2016, microRNA-25 inhibits cell apoptosis of human gastric adenocarcinoma cell line AGS via regulating CCNE1 and MYC. Med Sci Monit 22:1415–1420 HeneghanHM,Miller N, Lowery AJ, Sweeney KJ, Newell J, Kerin MJ, 2010, Circulating microRNAs as novel minimally invasive biomarkers for breast cancer. Ann Surg 251(3):499–505. https:// doi.org/10.1097/SLA.0b013e3181cc939f Chen H, Untiveros GM, McKee LA, Perez J, Li J, Antin PB, Konhilas JP, 2012, Micro-RNA-195 and -451 regulate the LKB1/ AMPK signaling axis by targeting MO25. PLoS One 7(7):e41574. , DOI 10.1371/journal.pone.0041574 Busk PK, Cirera S, 2010, MicroRNA profiling in early hypertrophic growth of the left ventricle in rats. Biochem Biophys Res Commun 396(4):989–993. , DOI 10.1016/j.bbrc.2010.05.039 Joglekar MV, Parekh VS, Mehta S, Bhonde RR, Hardikar AA, 2007, MicroRNA profiling of developing and regenerating pancreas reveal post-transcriptional regulation of neurogenin3. Dev Biol 311(2):603–612. , DOI 10.1016/j.ydbio.2007.09.008 Yu S, Jing L, Yin XR,WangMC, Chen YM, Guo Y, Nan KJ, Han LL, 2017)MiR-195 suppresses the metastasis and epithelial-mesenchymal transition of hepatocellular carcinoma by inhibiting YAP. Oncotarget 8(59):99757–99771. , DOI 10.18632/oncotarget.20909 Jia LF, Wei SB, Gong K, Gan YH, Yu GY, 2013, Prognostic implications of micoRNA miR-195 expression in human tongue squamous cell carcinoma. PLoS One 8(2):e56634. , DOI 10. 1371/journal.pone.0056634 Cecene G, Ak S, Eskiler GG, Demirdogen E, Erturk E, Gokgoz S, Polatkan V, Egeli U, Tunca B, Tezcan G, Topal U, Tolunay S, Tasdelen I, 2016, Circulating miR-195 as a therapeutic biomarker in Turkish breast Cancer patients. Asian Pac J Cancer Prev 17(9): 4241–4246 Soon PS, Tacon LJ, Gill AJ, Bambach CP, Sywak MS, Campbell PR, Yeh MW, Wong SG, Clifton-Bligh RJ, Robinson BG, Sidhu SB, 2009, miR-195 and miR-483-5p identified as predictors of poor prognosis in adrenocortical Cancer. Clin Cancer Res 15(24): 7684–7692. , DOI 10.1158/1078-0432.CCR-09-1587 Wang R, Zhao N, Li S, Fang JH, Chen MX, Yang J, Jia WH, Yuan Y, Zhuang SM(2013)MicroRNA-195 suppresses angiogenesis and metastasis of hepatocellular carcinoma by inhibiting the expression of VEGF, VAV2, and CDC42. Hepatology 58(2):642–653. https:// doi.org/10.1002/hep.26373 Lakomy R, Sana J, Hankeova S, Fadrus P, Kren L, Lzicarova E, Svoboda M, Dolezelova H, Smrcka M, Vyzula R, Michalek J, Hajduch M, Slaby O, 2011, MiR-195, miR-196b, miR-181c, miR-21 expression levels and O-6-methylguanine-DNA methyltransferase methylation status are associated with clinical outcome in glioblastoma patients. Cancer Sci 102(12):2186–2190. https:// doi.org/10.1111/j.1349-7006.2011.02092.x Ilhan-Mutlu A, Wohrer A, Berghoff AS, Widhalm G, Marosi C, Wagner L, Preusser M, 2013, Comparison of microRNA expression levels between initial and recurrent glioblastoma specimens. J Neuro- Oncol 112(3):347–354. , DOI 10.1007/s11060-013-1078-6 Zhang QQ, Xu H, HuangMB,Ma LM, Huang QJ, Yao Q, Zhou H, Qu LH, 2012, MicroRNA-195 plays a tumor-suppressor role in human glioblastoma cells by targeting signaling pathways involved in cellular proliferation and invasion. Neuro-Oncology 14(3):278– 287. , DOI 10.1093/neuonc/nor216 Gaur A, Jewell DA, Liang Y, Ridzon D, Moore JH, Chen C, Ambros VR, IsraelMA(2007, Characterization ofmicroRNAexpression levels and their biological correlates in human cancer cell lines. Cancer Res 67(6):2456–2468. , DOI 10.1158/0008-5472.CAN-06-2698 Henriksen M, Johnsen KB, Olesen P, Pilgaard L, DurouxM(2014, MicroRNA expression signatures and their correlation with clinicopathological features in glioblastoma multiforme. NeuroMolecular Med 16(3):565–577. , DOI 10.1007/s12017-014-8309-7 Wang A, Ren M, Song Y,Wang X,Wang Q, Yang Q, Liu H, Du Z, Zhang G, Wang J, 2018, MicroRNA expression profiling of bone marrow mesenchymal stem cells in steroid-induced osteonecrosis of the femoral head associated with osteogenesis. Med Sci Monit 24:1813–1825 Liu L, Chen L, Xu Y, Li R, Du X, 2010, microRNA-195 promotes apoptosis and suppresses tumorigenicity of human colorectal cancer cells. Biochem Biophys Res Commun 400(2):236–240. https:// doi.org/10.1016/j.bbrc.2010.08.046 Li C, ZhaoQ, ZhangW, ChenM, JuW, WuL, HanM, Ma Y, ZhuX, Wang D, Guo Z, He X, 2017, MicroRNA-146b-5p identified in porcine liver donation model is associated with early allograft dysfunction in human liver transplantation. Med Sci Monit 23:5876–5884 Yu M, Zhang X, Li H, Zhang P, DongW(2017)MicroRNA-588 is downregulated and may have prognostic and functional roles in human breast cancer. Med Sci Monit 23:5690–5696 Li F, Liang A, Lv Y, Liu G, Jiang A, Liu P, 2017, MicroRNA-200c inhibits epithelial-mesenchymal transition by targeting the BMI-1 gene through the Phospho-AKT pathway in endometrial carcinoma cells in vitro. Med Sci Monit 23:5139–5149 Tian Z, Zhou H, Xu Y, Bai J, 2017, MicroRNA-495 inhibits new bone regeneration via targeting high mobility group AT-hook 2, HMGA2). Med Sci Monit 23:4689–4698 Zhang Z, Li H, Chen S, Li Y, Cui Z, Ma J, 2017, Knockdown of MicroRNA-122 protects H9c2 cardiomyocytes from hypoxia-induced apoptosis and promotes autophagy. Med Sci Monit 23:4284–4290 Fang DZ, Wang YP, Liu J, Hui XB, Wang XD, Chen X, Liu D, 2018, MicroRNA-129-3p suppresses tumor growth by targeting E2F5 in glioblastoma. Eur Rev Med Pharmacol Sci 22(4):1044– 1050. , DOI 10.26355/eurrev_201802_14387 Szemraj M, Oszajca K, Szemraj J, Jurowski P, 2017, MicroRNA expression analysis in serum of patients with congenital hemochromatosis and age-related macular degeneration, AMD). Med Sci Monit 23:4050–4060 Zhang J, Qin L, Han L, Zhao Y, Jing H, SongW, Shi H, 2017, Role of MicroRNA-93 I in pathogenesis of left ventricular remodeling via targeting cyclin-D1. Med Sci Monit 23:3981–3988 Gan TQ, Chen WJ, Qin H, Huang SN, Yang LH, Fang YY, Pan LJ, Li ZY, Chen G, 2017, Clinical value and prospective pathway signaling of MicroRNA-375 in lung adenocarcinoma: a study based on the Cancer genome atlas, TCGA), gene expression omnibus, GEO, and bioinformatics analysis. Med Sci Monit 23:2453–2464 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 753 EP 763 DI 10.1007/s12253-019-00622-3 PG 11 ER PT J AU Wang, Q Li, Ch Xie, Z Bu, Z Shi, L Wang, Ch Jiang, F AF Wang, Qianru Li, Chunjiao Xie, Zhongli Bu, Zhiguo Shi, Liwei Wang, Chuan Jiang, Feng TI The Development and Application of Virtual Reality Animation Simulation Technology: Take Gastroscopy Simulation System as an Example SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE VR animation; Medical simulation; Gastroscopy simulation; Operating system; Cross-border fusion ID VR animation; Medical simulation; Gastroscopy simulation; Operating system; Cross-border fusion AB Virtual reality (VR) technology has a great potential in the field of medical simulation due to its immersion, interactivity and autonomy. It provides a new direction for integration and application in various disciplines. Combination of VR technology and clinical practice brings great convenience for medical education and experiments. Modern VR simulators can create realistic environments that capture minute anatomical details with high accuracy and solves the problem of difficulty in mass productions with traditional devices. Taking gastroscopy simulation system as an example, this paper discusses the development and application of VR animation technology, together with its excellent performance and current research status in surgery, scientific research, training and education. C1 [Wang, Qianru] Film-Video-Animation School, Sichuan Fine Arts Institute, 401331 Chongqing, China. [Li, Chunjiao] Film-Video-Animation School, Sichuan Fine Arts Institute, 401331 Chongqing, China. [Xie, Zhongli] Film-Video-Animation School, Sichuan Fine Arts Institute, 401331 Chongqing, China. [Bu, Zhiguo] Film-Video-Animation School, Sichuan Fine Arts Institute, 401331 Chongqing, China. [Shi, Liwei] Chongqing General Hospital, Digestive Department, No. 104, Pipashan Main Street, Yuzhong District, 400014 Chongqing, China. [Wang, Chuan] Chongqing General Hospital, Digestive Department, No. 104, Pipashan Main Street, Yuzhong District, 400014 Chongqing, China. [Jiang, Feng] Chongqing General Hospital, Digestive Department, No. 104, Pipashan Main Street, Yuzhong District, 400014 Chongqing, China. RP Jiang, F (reprint author), Chongqing General Hospital, Digestive Department, 400014 Chongqing, China. EM FengJiang17@126.com CR Schroeder R, 1993, Virtual reality in the real world: history, applications and projections. Futures 25(9):963–973 Gatica-Rojas V, Mendez-Rebolledo G, 2014, Virtual reality interface devices in the reorganization of neural networks in the brain of patients with neurological diseases. Neural Regen Res 9(8):888–896 Harpham-Lockyer L, Laskaratos FM, Berlingieri P, Epstein O, 2015, Role of virtual reality simulation in endoscopy training. World J Gastrointest Endosc 7(18):1287–1294 WangW, Tang L, Zhao XF, Huang JS, 2015, About the thinking of high simulation equipment's application on clinical medical practice teaching. Pop Sci Tech 15(5):153–154,140, in Chinese) Guze PA, 2015, Using technology to meet the challenges of medical education. Trans Am Clin Climatol Assoc 126:260–270 Kin T, Nakatomi H, Shono N, Nomura S, Saito T, Oyama H, Saito N, 2017, Neurosurgical virtual reality simulation for brain tumor using high-definition computer graphics: a review of the literature. Neurol Med Chir, Tokyo, 57(10):513–520 Grantcharov TP, Kristiansen VB, Bendix J, Bardram L, Rosenberg J, Funch-Jensen P, 2004, Randomized clinical trial of virtual reality simulation for laparoscopic skills training. Br J Surg 91(2):146–150 Coleman J, Nduka CC, Darzi A, 1994, Virtual reality and laparoscopic surgery. Br J Surg 81(12):1709–1711 Mousnier A, Kubat N, Massias-Simon A, Segeral E, Rain JC, Benarous R, Emiliani S, Dargemont C, 2007, von Hippel Lindau binding protein 1-mediated degradation of integrase affects HIV-1 gene expression at a postintegration step. Proc Natl Acad Sci U S A 104(34):13615–13620 Alaker M, Wynn GR, Arulampalam T, 2016, Virtual reality training in laparoscopic surgery: a systematic review & meta-analysis. Int J Surg 29:85–94 Seymour NE, 2008, VR to OR: a review of the evidence that virtual reality simulation improves operating room performance. World J Surg 32(2):182–188 Kundhal PS, Grantcharov TP, 2009, Psychomotor performance measured in a virtual environment correlates with technical skills in the operating room. Surg Endosc 23(3):645–649 Hyltander A, Liljegren E, Rhodin PH, Lonroth H, 2002, The transfer of basic skills learned in a laparoscopic simulator to the operating room. Surg Endosc 16(9):1324–1328 Qi XY, Lu W, Han FT, Liu HQ, Cao YX, 2012, Researching and application of virtual simulation teaching inmedical imaging equipment teaching. Chin J Med Phys 29(1):3208–3210, in Chinese) de Visser H,WatsonMO, Salvado O, Passenger JD, 2011, Progress in virtual reality simulators for surgical training and certification. Med J Aust 194(4):S38–S40 Arnold SH, Svendsen MB, Konge L, Svendsen LB, Preisler L, 2015, Three-dimensional motion tracking correlates with skill level in upper gastrointestinal endoscopy. Endoscopy 47(9):825–828 Latypov NN, Latypov NN, 1999, Method for tracking and displaying user's spatial position and orientation, a method for representing virtual reality for a user, and systems of embodiment of such methods. US. US 6005548 A. Burdea GC, 1996, Force and touch feedback for virtual reality. John Wiley & Sons, Inc., New York Wang P, BeckerAA, Jones IA, Glover AT, Benford SD, Greenhalgh CM, Vloeberghs M, 2006, A virtual reality surgery simulation of cutting and retraction in neurosurgery with force-feedback. Comput Methods Prog Biomed 84(1):11–18 Maschuw K, Hassan I, Bartsch DK, 2010, Surgical training using simulator. Virtual reality. Chirurg 81(1):19–24 Badash I, Burtt K, Solorzano CA, Carey JN, 2016, Innovations in surgery simulation: a review of past, current and future techniques. Ann Transl Med 4(23):453 Gorman PJ,Meier AH, Krummel TM(1999, Simulation and virtual reality in surgical education: real or unreal? Arch Surg 134(11): 1203–1208 Zhang WB, Cao L, Xiong JJ, 2016, The realistic challenges of virtual reality technology. Sci Sin Inform 46(12):1779–1784, in Chinese) Burdea GC, Coiffet P, 2003, Virtual reality technology, 2nd edn. John Wiley & Sons, Inc., New Jersey, USA Sakakushev BE, Marinov BI, Stefanova PP, Kostianev SS, Georgiou EK, 2007, Striving for better medical education: the simulation approach. Folia Med, Plovdiv, 59(2):123–131 Zhang L, Grosdemouge C, Arikatla VS, AhnW, Sankaranarayanan G, De S, Jones D, Schwaitzberg S, Cao CG, 2012, The added value of virtual reality technology and forcefeedback for surgical training simulators. Work 41(Suppl 1):2288–2292 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 765 EP 769 DI 10.1007/s12253-019-00590-8 PG 5 ER PT J AU Zhu, W Ji, X AF Zhu, Wei Ji, Xiaoliang TI The Impact of MicroRNA-133a on Prognosis and Clinicopathological Parameters for Digestive System Cancers: a Comprehensive Study Based on Meta-Analysis and TCGA Database SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MicroRNA-133a; Digestive system Cancer; TCGA; Meta-analysis ID MicroRNA-133a; Digestive system Cancer; TCGA; Meta-analysis AB We conducted a meta-analysis on the impact of microRNA-133a (miR-133a) on digestive system cancers, and verified the results through The Cancer Genome Atlas (TCGA). Relevant studies were searched in English and Chinese database and meta-analysis was performed using Stata 12.0. The corresponding information of miR-133a and digestive system cancers were obtained from TCGA database and analysis was performed using SPSS. Increased miR-133a expression was linked with favorable overall survival (OS) in digestive system cancers (HR = 0.539, 95% CI: 0.416–0.698, P < 0.001), digestive tract cancers (HR =0.558, 95% CI: 0.406–0.767, P < 0.001), esophageal squamous cell carcinoma (ESCC) (HR = 0.427, 95% CI: 0.265–0.690, P = 0.001) and gastric cancer (HR = 0.541, 95%CI: 0.385–0.761, P < 0.001). The expression ofmiR-133a was significantly lower in cancer tissue compared with adjacent tissue for ESCC (P < 0.001), gastric cancer (P < 0.001), colorectal cancer (P < 0.001) and hepatocellular carcinoma (P = 0.002). Meanwhile, the area under the ROC curve (AUC) value for miR-133a was 0.836, 0.888, and 0.99 in ESCC, gastric cancer and colorectal cancer. MiR-133a is a tumor suppressor with prognostic and diagnostic values for digestive system cancers. High miR-133a expression was associated with better prognosis and less adverse clinicopathological parameters. More research should be performed to test these findings. C1 [Zhu, Wei] The First Hospital of Huzhou, Department of General SurgeryHuzhou, Zhejiang, China. [Ji, Xiaoliang] Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Department of General SurgeryHuzhou, Zhejiang, China. RP Ji, X (reprint author), Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Department of General Surgery, Huzhou, China. EM jixiaolianghz@163.com CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108. , DOI 10.3322/caac.21262 Pourhoseingholi MA, Vahedi M, Baghestani AR, 2015, Burden of gastrointestinal cancer in Asia; an overview. Gastroenterol Hepatol Bed Bench 8(1):19–27 Yuequan J, Shifeng C, Bing Z, 2010, Prognostic factors and family history for survival of esophageal squamous cell carcinoma patients after surgery. Ann Thorac Surg 90(3):908–913. , DOI 10. 1016/j.athoracsur.2010.05.060 Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, Pfeffer S, Rice A, Kamphorst AO, Landthaler M, Lin C, Socci ND, Hermida L, Fulci V, Chiaretti S, Foa R, Schliwka J, Fuchs U, Novosel A, Muller RU, Schermer B, Bissels U, Inman J, Phan Q, Chien M,Weir DB, Choksi R, De Vita G, Frezzetti D, Trompeter HI, Hornung V, Teng G, Hartmann G, PalkovitsM,Di Lauro R, Wernet P, Macino G, Rogler CE, Nagle JW, Ju J, Papavasiliou FN, Benzing T, Lichter P, TamW, BrownsteinMJ, Bosio A, Borkhardt A, Russo JJ, Sander C, Zavolan M, Tuschl T, 2007, A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 129(7):1401–1414. , DOI 10.1016/j.cell.2007.04.040 Gao SH, Liu J, Zhang HJ, Zhao N, Zhang J, 2016, Low miR-133a expression is a predictor of outcome in patients with esophageal squamous cell cancer. Eur Rev Med Pharmacol Sci 20(18):3788– 3792 Li CY, Liang GY, Yao WZ, Sui J, Shen X, Zhang YQ, Peng H, Hong WW, Ye YC, Zhang ZY, ZhangWH, Yin LH, Pu YP, 2017, Identification and functional characterization of microRNAs reveal a potential role in gastric cancer progression. Clin Transl Oncol 19(2):162–172. , DOI 10.1007/s12094-016-1516-y Wan TM, Lam CS, Ng L, Chow AK,Wong SK, Li HS,Man JH, Lo OS, Foo D, Cheung A, Yau T, Poon JT, Poon RT, Law WL, Pang RW, 2014, The clinicopathological significance of miR-133a in colorectal cancer. Dis Markers 2014:919283–919288. https://doi. org/10.1155/2014/919283 Qin Y, Dang X, Li W, Ma Q, 2013, miR-133a functions as a tumor suppressor and directly targets FSCN1 in pancreatic cancer. Oncol Res 21(6):353–363. , DOI 10.3727/096504014x14024160459122 Krieg A, Riemer JC, Telan LA, Gabbert HE, Knoefel WT, 2015, CXCR4–a prognostic and Clinicopathological biomarker for pancreatic ductal adenocarcinoma: a meta-analysis. PLoS One 10(6): e0130192. , DOI 10.1371/journal.pone.0130192 Stang A, 2010, Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in metaanalyses. Eur J Epidemiol 25(9):603–605. , DOI 10.1007/ s10654-010-9491-z Riley RD, Elia EG, Malin G, Hemming K, Price MP, 2015, Multivariate meta-analysis of prognostic factor studies with multiple cut-points and/or methods of measurement. Stat Med 34(17): 2481–2496. , DOI 10.1002/sim.6493 Suzuki S, Yokobori T, Tanaka N, Sakai M, Sano A, Inose T, Sohda M, Nakajima M, Miyazaki T, Kato H, Kuwano H, 2012, CD47 expression regulated by the miR-133a tumor suppressor is a novel prognostic marker in esophageal squamous cell carcinoma. Oncol Rep 28(2):465–472. , DOI 10.3892/or.2012.1831 Akanuma N, Hoshino I, Akutsu Y, Murakami K, Isozaki Y, Maruyama T, Yusup G, Qin W, Toyozumi T, Takahashi M, Suito H, Hu X, Sekino N, Matsubara H, 2014, MicroRNA-133a regulates the mRNAs of two invadopodia-related proteins, FSCN1 and MMP14, in esophageal cancer. Br J Cancer 110(1):189–198. , DOI 10.1038/bjc.2013.676 Chen G, Peng J, Zhu W, Tao G, Song Y, Zhou X, Wang W, 2014, Combined downregulation of microRNA-133a and microRNA- 133b predicts chemosensitivity of patients with esophageal squamous cell carcinoma undergoing paclitaxel-based chemotherapy. Med Oncol 31(11):263. , DOI 10.1007/s12032-014-0263-6 Cheng Z, Liu F,Wang G, Li Y, Zhang H, Li F, 2014, miR-133 is a key negative regulator of CDC42-PAK pathway in gastric cancer. Cell Signal 26(12):2667–2673. , DOI 10.1016/j.cellsig. 2014.08.012 Wang LL, Du LT, Li J, Liu YM, Qu AL, Yang YM, Zhang X, Zheng GX, Wang CX, 2014, Decreased expression of miR-133a correlates with poor prognosis in colorectal cancer patients.World J Gastroenterol 20(32):11340–11346. , DOI 10.3748/wjg. v20.i32.11340 Zhang W, Liu K, Liu S, Ji B, Wang Y, Liu Y, 2015, MicroRNA- 133a functions as a tumor suppressor by targeting IGF-1R in hepatocellular carcinoma. Tumour Biol 36(12):9779–9788. https://doi. org/10.1007/s13277-015-3749-8 Calin GA, Croce CM, 2006, MicroRNA signatures in human cancers. Nat Rev Cancer 6(11):857–866. , DOI 10.1038/ nrc1997 Kaluzna EM, 2014, MicroRNA-155 and microRNA-196b: promising biomarkers in hepatitis C virus infection? Rev Med Virol 24(3):169–185. , DOI 10.1002/rmv.1785 Han ZB, Chen HY, Fan JW,Wu JY, Tang HM, Peng ZH, 2012, Upregulation of microRNA-155 promotes cancer cell invasion and predicts poor survival of hepatocellular carcinoma following liver transplantation. J Cancer Res Clin Oncol 138(1):153–161. https:// doi.org/10.1007/s00432-011-1076-z Godfrey AC, Xu Z,Weinberg CR, Getts RC,Wade PA, DeRoo LA, Sandler DP, Taylor JA, 2013, Serum microRNA expression as an early marker for breast cancer risk in prospectively collected samples from the sister study cohort. Breast Cancer Res 15(3):R42. , DOI 10.1186/bcr3428 Wang LK, Hsiao TH, Hong TM, Chen HY, Kao SH,WangWL, Yu SL, Lin CW, Yang PC, 2014, MicroRNA-133a suppressesmultiple oncogenic membrane receptors and cell invasion in non-small cell lung carcinoma. PLoS One 9(5):e96765. , DOI 10.1371/ journal.pone.0096765 Li S, Xiao FY, Shan PR, Su L, Chen DL, Ding JY,Wang ZQ, 2015, Overexpression of microRNA-133a inhibits ischemia-reperfusioninduced cardiomyocyte apoptosis by targeting DAPK2. J Hum Genet 60(11):709–716. , DOI 10.1038/jhg.2015.96 Ji F, Zhang H, Wang Y, Li M, Xu W, Kang Y, Wang Z, Wang Z, Cheng P, Tong D, Li C, Tang H, 2013, MicroRNA-133a, downregulated in osteosarcoma, suppresses proliferation and promotes apoptosis by targeting Bcl-xL and Mcl-1. Bone 56(1):220–226. , DOI 10.1016/j.bone.2013.05.020 Fujiwara T, Katsuda T, Hagiwara K, Kosaka N, Yoshioka Y, Takahashi RU, Takeshita F, Kubota D, Kondo T, Ichikawa H, Yoshida A, Kobayashi E, Kawai A, Ozaki T, Ochiya T, 2014, Clinical relevance and therapeutic significance of microRNA- 133a expression profiles and functions in malignant osteosarcoma-initiating cells. Stem Cells 32(4):959–973. https:// doi.org/10.1002/stem.1618 Nohata N, Hanazawa T, Kikkawa N, Mutallip M, Fujimura L, Yoshino H, Kawakami K, Chiyomaru T, Enokida H, Nakagawa M, Okamoto Y, Seki N, 2011, Caveolin-1 mediates tumor cell migration and invasion and its regulation by miR-133a in head and neck squamous cell carcinoma. Int J Oncol 38(1):209–217 Zhao L,WangH, Liu C, Liu Y,Wang X,Wang S, SunX, Li J, Deng Y, Jiang Y, Ding Y, 2010, Promotion of colorectal cancer growth and metastasis by the LIM and SH3 domain protein 1. Gut 59(9): 1226–1235. , DOI 10.1136/gut.2009.202739 Chiyomaru T, Enokida H, Tatarano S, Kawahara K, Uchida Y, Nishiyama K, Fujimura L, Kikkawa N, Seki N, Nakagawa M, 2010, miR-145 and miR-133a function as tumour suppressors and directly regulate FSCN1 expression in bladder cancer. Br J Cancer 102(5):883–891. , DOI 10.1038/sj.bjc.6605570 Xiao J, Zou Y, Lu X, Xie B, Yu Q, He B, He B, Chen Q, 2016, Prognostic value of decreased microRNA-133a in solid cancers: a meta-analysis. OncoTargets Ther 9:5771–5779. , DOI 10. 2147/ott.s112358 Zhao Y, Huang J, Zhang L, Qu Y, Li J, Yu B, Yan M, Yu Y, Liu B, Zhu Z, 2014, MiR-133b is frequently decreased in gastric cancer and its overexpression reduces the metastatic potential of gastric cancer cells. BMC Cancer 14:34. , DOI 10.1186/1471- 2407-14-34 Li X, Wan X, Chen H, Yang S, Liu Y, Mo W, Meng D, Du W, Huang Y, Wu H, Wang J, Li T, Li Y, 2014, Identification of miR- 133b and RB1CC1 as independent predictors for biochemical recurrence and potential therapeutic targets for prostate cancer. Clin Cancer Res 20(9):2312–2325. , DOI 10.1158/1078-0432. ccr-13-1588 Chen XN,Wang KF, Xu ZQ, Li SJ, Liu Q, Fu DH,Wang X,Wu B, 2014, MiR-133b regulates bladder cancer cell proliferation and apoptosis by targeting Bcl-w and Akt1. Cancer Cell Int 14:70. , DOI 10.1186/s12935-014-0070-3 Qiu T, Zhou X,Wang J, Du Y, Xu J, Huang Z, ZhuW, Shu Y, Liu P, 2014, MiR-145, miR-133a and miR-133b inhibit proliferation, migration, invasion and cell cycle progression via targeting transcription factor Sp1 in gastric cancer. FEBS Lett 588(7):1168– 1177. , DOI 10.1016/j.febslet.2014.02.054 Kano M, Seki N, Kikkawa N, Fujimura L, Hoshino I, Akutsu Y, Chiyomaru T, Enokida H, NakagawaM, Matsubara H, 2010, miR- 145, miR-133a and miR-133b: tumor-suppressive miRNAs target FSCN1 in esophageal squamous cell carcinoma. Int J Cancer 127(12):2804–2814. , DOI 10.1002/ijc.25284 Zhou Y,Wu D, Tao J, Qu P, Zhou Z, Hou J, 2013)MicroRNA-133 inhibits cell proliferation, migration and invasion by targeting epidermal growth factor receptor and its downstream effector proteins in bladder cancer. Scand J Urol 47(5):423–432. , DOI 10. 3109/00365599.2012.748821 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 771 EP 781 DI 10.1007/s12253-019-00619-y PG 11 ER PT J AU Miyamoto, M Takano, M Tsuda, H Matsuura, H Sakamoto, T Takasaki, K Kato, K Soyama, H Aoyama, T Ishibashi, H Iwahashi, H Furuya, K AF Miyamoto, Morikazu Takano, Masashi Tsuda, Hitoshi Matsuura, Hiroko Sakamoto, Takahiro Takasaki, Kazuki Kato, Kento Soyama, Hiroaki Aoyama, Tadashi Ishibashi, Hiroki Iwahashi, Hideki Furuya, Kenichi TI The Haphazard Pattern in Grade-3 Endometrioid Carcinoma Is Associated with Poor Prognosis and Tumor Lymphocyte Infiltration SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial carcinoma; Endometrial endometrioid carcinoma; Invasive pattern; Tumor infiltrating lymphocyte ID Endometrial carcinoma; Endometrial endometrioid carcinoma; Invasive pattern; Tumor infiltrating lymphocyte AB The aim of this study was to examine the associations among the haphazard invasive patterns, defined as directionless infiltration into the myometrium; expression of key proteins; tumor infiltrative lymphocytes (TILs); and the prognosis of gade-3 endometrioid carcinoma (G3EC). Between 1990 and 2013, patients with G3EC who underwent surgery at our hospital were identified. Invasive patterns were classified into either haphazard, infiltrative, or expansile patterns. The estrogen, progesterone, androgen receptor, cytokeratin 5/6, epidermal growth factor receptor, E-cadherin, snail-2, vimentin, ZEB1, chromogranin A, synaptophysin, MLH1, MSH2, MSH6, and PMS2 levels were evaluated by immunochemical analysis. The degree of strong or weak lymphocyte infiltration (LI) were evaluated using zone formation of LI at the invasive front. Haphazard, infiltrative, and expansile patterns were discovered in 8 (18%), 6 (13%), and 31 (69%) cases, respectively. Cases with the haphazard patterns were diagnosed at a more advanced stage (p < 0.01) and recurred more frequently (p < 0.01). There were statistical differences in progression-free survival (PFS) and overall survival (OS) between the three groups (PFS; p < 0.01: OS; p < 0.01). In multivariate analysis, only the haphazard pattern was found to be an independent, worse prognostic factor of PFS (Hazard ratio (HR) =10.8, p < 0.01) and OS (HR= 23.3, p < 0.01). Furthermore, the haphazard invasive pattern was related with weak LI (p < 0.01) but not with the expression of all proteins analyzed. The haphazard pattern was found to be a worse prognostic factor and was associated with weak LI in G3EC. The aggressive feature of G3EC might be associated with LI but not tumor biology. C1 [Miyamoto, Morikazu] National Defense Medical College, Department of Obstetrics and Gynecology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Takano, Masashi] National Defense Medical College, Department of Clinical OncologyTokorozawa, Saitama, Japan. [Tsuda, Hitoshi] National Defense Medical College, Department of Basic PathologyTokorozawa, Saitama, Japan. [Matsuura, Hiroko] National Defense Medical College, Department of Obstetrics and Gynecology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Sakamoto, Takahiro] National Defense Medical College, Department of Obstetrics and Gynecology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Takasaki, Kazuki] National Defense Medical College, Department of Obstetrics and Gynecology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Kato, Kento] National Defense Medical College, Department of Obstetrics and Gynecology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Soyama, Hiroaki] National Defense Medical College, Department of Obstetrics and Gynecology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Aoyama, Tadashi] National Defense Medical College, Department of Obstetrics and Gynecology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Ishibashi, Hiroki] National Defense Medical College, Department of Obstetrics and Gynecology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Iwahashi, Hideki] National Defense Medical College, Department of Obstetrics and Gynecology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Furuya, Kenichi] National Defense Medical College, Department of Obstetrics and Gynecology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. RP Miyamoto, M (reprint author), National Defense Medical College, Department of Obstetrics and Gynecology, 359-8513 Tokorozawa, Japan. EM morikazu1118@hotmail.co.jp CR Prat J, 2015, Pathology of cancers of the female genital tract. Int J Gynaecol Obstet 131(Suppl 2):S132–S145. , DOI 10. 1016/j.ijgo.2015.06.010. Cancer Genome Atlas Research Network, Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, Shen H, RobertsonAG, Pashtan I, Shen R, Benz CC, Yau C, Laird PW, Ding L, Zhang W, Mills GB, Kucherlapati R,Mardis ER, Levine DA, 2013, Integrated genomic characterization of endometrial carcinoma. Nature 497:67–73. , DOI 10.1038/nature12113 Clinical Practice Guideline in Oncology. National Comprehensive Cancer Network. Http://www.nccn.org/professionals/physiciangls/ PDF/uterine.pdf. Accessed 04 Jan 2019 Morice P, Leary A, Creutzberg C, Abu-Rustum N, Darai E, 2016, Endometrial cancer. Lancet. 387:1094–1108. , DOI 10. 1016/S0140-6736(15)00130-0 Epub 2015 Sep 6 Kurman RJ, Carcangiu ML, Herrington CS, Young RH, 2014, WHO classification of tumours of female reproductive organs. International Agency for Research on Cancer, Lyon, pp 126–133 Lewin SN, Herzog TJ, Barrena Medel NI, Deutsch I, Burke WM, Sun X, Wright JD, 2010, Comparative performance of the 2009 international federation of gynecology and obstetrics' staging system for uterine corpus cancer. Obstet Gynecol 116:1141–1149. , DOI 10.1097/AOG.0b013e3181f39849 Wang J, Jia N, Li Q, Wang C, Tao X, Hua K, Feng W, 2016, Analysis of recurrence and survival rates in grade 3 endometrioid endometrial carcinoma. Oncol Lett 12:2860–2867. , DOI 10.3892/ol.2016.4918 Rasool N, Fader AN, Seamon L, Neubauer NL, Shahin FA, Alexander HA, Moore K, Moxley K, Secord AA, Kunos C, Rose PG, O'Malley DM, 2010, Stage I, grade 3 endometrioid adenocarcinoma of the endometrium: an analysis of clinical outcomes and patterns of recurrence. Gynecol Oncol 116:10–14. , DOI 10.1016/j.ygyno.2009.10.043 Epub 2009 Oct 28 MiyamotoM, TakanoM, Tsuda H,Matuura H, Aoyama T, Soyama H, Kato K, Iwahashi H, Ishibashi H,Yoshikawa T, SuzukiA, Hirata J, Furuya K, 2018, Zone formation of lymphocyte infiltration at invasive front as a biomarker of prognosis in endometrial carcinomas. Oncology 3:1–8. , DOI 10.1159/000493010 McIntyre JB, Nelson GS, Ghatage P, Morris D, Duggan MA, Lee CH, Doll CM, Kobel M, 2014, PIK3CA missense mutation is associated with unfavorable outcome in grade 3 endometrioid carcinoma but not in serous endometrial carcinoma. Gynecol Oncol 132:188–193. , DOI 10.1016/j.ygyno.2013.11.015 Meng B, Hoang LN, McIntyre JB, Duggan MA, Nelson GS, Lee CH, Kobel M, 2014, POLE exonuclease domain mutation predicts long progression-free survival in grade 3 endometrioid carcinoma of the endometrium. Gynecol Oncol 132:188–193. , DOI 10.1016/j.ygyno.2014.05.006. Suzuki C, Matsumoto T, Sonoue H, Arakawa A, Furugen Y, Kinoshita K, 2003, Prognostic significance of the infiltrative pattern invasion in endometrioid adenocarcinoma of the endometrium. Pathol Int 53:495–500 Longacre TA, Hendrickson MR, 1999, Diffusely infiltrative endometrial adenocarcinoma. An adenoma malignum pattern of myoinvasion. Am J Surg Pathol 23:69–78 Iwatsuki M, Mimori K, Yokobori T, Ishi H, Beppu T, Nakamori S, Baba H, Mori M, 2010, Epithelial-mesenchymal transition in cancer development and its clinical significance. Cancer Sci 101:293– 299. , DOI 10.1111/j.1349-7006.2009.01419.x Colas E, Pedrola N, Devis L, Ertekin T, Campoy I, Martinez E, Llaurado M, Rigau M, Olivan M, Garcia M, Cabrera S, Gil- Moreno A, Xercavins J, Castellvi J, Garcia A, Ramon y Cajal S, Moreno-Bueno G, Dolcet X, Alameda F, Palacios J, Prat J, Doll A, Matias-Guiu X, Abal M, Reventos J, 2012, The EMT signaling pathways in endometrial carcinoma. Clin Transl Oncol 14:715– 720. , DOI 10.1007/s12094-012-0866-3 Eichhorn JH, Young RH, 2001, Neuroendocrine tumors of the genital tract. Am J Clin Pathol 115(Suppl):S94–S112. , DOI 10.1309/64CW-WKGK-49EF-BYD1 van Hoeven KH, Hudock JA, Woodruff JM, Suhrland MJ, 1995, Small cell neuroendocrine carcinoma of the endometrium. Int J Gynecol Pathol 14:21–29 Niikura H, Sasano H, Matsunaga G, Watanabe K, Ito K, Sato S, Yajima A, 1995, Prognostic value of epidermal growth factor receptor expression in endometrioid endometrial carcinoma. Hum Pathol 26:892–896 Stefansson IM, Salvesen HB, Akslen LA, 2006, Loss of p63 and cytokeratin 5/6 expression is associated with more aggressive tumors in endometrial carcinoma patients. Int J Cancer 118:1227– 1233. , DOI 10.1002/ijc.21415 Piulats JM, Matias-Guiu X, 2016, Immunotherapy in endometrial Cancer: in the Nick of time. Clin Cancer Res 22:5623–5625. https:// doi.org/10.1158/1078-0432.CCR-16-1820 Alvarez T, Miller E, Duska L, Oliva E, 2012, Molecular profile of grade 3 endometrioid endometrial carcinoma: is it a type I or type II endometrial carcinoma? Am J Surg Pathol 36:753–761. https://doi. org/10.1097/PAS.0b013e318247b7bb NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 783 EP 790 DI 10.1007/s12253-019-00624-1 PG 8 ER PT J AU Lubowicka, E Zbucka-Kretowska, M Sidorkiewicz, I Zajkowska, M Gacuta, E Puchnarewicz, A Chrostek, L Szmitkowski, M Lawicki, S AF Lubowicka, Emilia Zbucka-Kretowska, Monika Sidorkiewicz, Iwona Zajkowska, Monika Gacuta, Ewa Puchnarewicz, Andrzej Chrostek, Lech Szmitkowski, Maciej Lawicki, Slawomir TI Diagnostic Power of Cytokine M-CSF, Metalloproteinase 2 (MMP-2) and Tissue Inhibitor-2 (TIMP-2) in Cervical Cancer Patients Based on ROC Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE M-CSF; MMP-2; TIMP-2; Cervical cancer; Tumor markers ID M-CSF; MMP-2; TIMP-2; Cervical cancer; Tumor markers AB Macrophage colony-stimulating factor (M-CSF), matrix metalloproteinase-2 (MMP-2) and its specific tissue inhibitor (TIMP-2) may play an important role in the pathogenesis of cancer disease. We investigated the plasma levels and diagnostic power (ROC curve analysis) of M-CSF, MMP-2, TIMP-2 and tumor markers CA 125 and SCC-Ag in cervical cancer (CC) patients as compared to control group. The study included 89 patients with cervical cancer. The control group consisted of 50 healthy, untreated women. The plasma levels of M-CSF, MMP-2 and TIMP-2 were determined using ELISA, CA 125 and SCC-Ag – by CMIA method. The median levels of M-CSF, TIMP-2, SCC-Ag and CA 125 in the entire group of CC were significantly different than compared to the healthy women group. MMP-2 showed the highest value of sensitivity from all examined parameters (in stage I of CC – 93.10%, II – 82.76%, III and IV – 96.88%, total group – 92.05%). The highest specificity was obtained by M-CSF (86%). The area under the ROC curve (AUC) of M-CSF (0.8051) was the largest of all the tested parameters (even higher than commonly used tumor markers) in the group of cervical cancer. The combination of M-CSF, MMP-2 or TIMP- 2 with SCC antigen resulted in an increase AUCs in all cases (0.8760;0.7880;0.8081;respectively). The findings of this study suggest the usefulness of all examined parameters in the diagnostics of CC patients. Out of the tested substances, M-CSF also appears to be the best candidate for cancer diagnostics in all stages of the disease, based on ROC analysis. C1 [Lubowicka, Emilia] Medical University of Bialystok, Department of Esthetic Medicine, Akademicka 3, 15-267 Bialystok, Poland. [Zbucka-Kretowska, Monika] Medical University of Bialystok, Department of Reproduction and Gynecological Endocrinology, 15-276 Bialystok, Poland. [Sidorkiewicz, Iwona] Medical University of Bialystok, Department of Reproduction and Gynecological Endocrinology, 15-276 Bialystok, Poland. [Zajkowska, Monika] Medical University of Bialystok, Department of Biochemical Diagnostics, 15-269 Bialystok, Poland. [Gacuta, Ewa] Medical University of Bialystok, Department of Perinatology, 15-276 Bialystok, Poland. [Puchnarewicz, Andrzej] Provincial Hospital, Department of Urology, 15-278 Bialystok, Poland. [Chrostek, Lech] Medical University of Bialystok, Department of Biochemical Diagnostics, 15-269 Bialystok, Poland. [Szmitkowski, Maciej] Medical University of Bialystok, Department of Biochemical Diagnostics, 15-269 Bialystok, Poland. [Lawicki, Slawomir] Medical University of Bialystok, Department of Population Medicine and Civilization Diseases Prevention, 15-269 Bialystok, Poland. RP Lubowicka, E (reprint author), Medical University of Bialystok, Department of Esthetic Medicine, 15-267 Bialystok, Poland. EM emila_lubowicka@wp.pl CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108. , DOI 10.3322/caac.21262 Jin L, Xu ZX, 2015, Recent advances in the study of HPVassociated carcinogenesis. Virol Sin 30(2):101–106. https://doi. org/10.1007/s12250-015-3586-3 Tsikouras P, Zervoudis S, Manav B, Tomara E, Iatrakis G, Romanidis C, Bothou A, Galazios G, 2016, Cervical cancer: screening, diagnosis and staging. J BUON 21(2):320–325 Lewis CE, Pollard JW, 2006, Distinct role of macrophages in different tumor microenvironments. Cancer Res 66(2):605–612. , DOI 10.1158/0008-5472.CAN-05-4005 Achkova D, Maher J, 2016, Role of the colony-stimulating factor, CSF)/CSF-1 receptor axis in cancer. Biochem Soc Trans 44(2): 333–341. , DOI 10.1042/BST20150245 Zajkowska M, Glazewska EK, Bedkowska GE, Chorazy P, Szmitkowski M, Lawicki S, 2016, Diagnostic power of vascular endothelial growth factor and macrophage Colony-stimulating factor in breast Cancer patients based on ROC analysis. Mediat Inflamm 2016:5962946. , DOI 10.1155/2016/5962946 Richardsen E, Uglehus RD, Johnsen SH, Busund LT, 2015, Macrophage-colony stimulating factor, CSF1, predicts breast cancer progression and mortality. Anticancer Res 35(2):865–874 Bedkowska GE, Lawicki S, Gacuta E, Pawlowski P, Szmitkowski M, 2015, M-CSF in a new biomarker panel with HE4 and CA 125 in the diagnostics of epithelial ovarian cancer patients. J Ovarian Res 8:27. , DOI 10.1186/s13048-015-0153-3 Harano K, Hirakawa A, Kato T, Suzuki K,Watanabe S, Katsumata N, 2014, Use of colony-stimulating factor in patients with ovarian cancer receiving paclitaxel and carboplatin in Japan. J Gynecol Oncol 25(2):124–129. , DOI 10.3802/jgo.2014.25.2.124 Poonawalla IB, Piller LB, Lairson DR, ChanW, Du XL, 2016, Use of hematopoietic growth factors and risk of thromboembolic and pulmonary toxicities in elderly patients with advanced ovarian Cancer. Womens Health Issues 26(5):574–583. , DOI 10. 1016/j.whi.2016.05.007 Aharinejad S, Salama M, Paulus P, Zins K, Berger A, Singer CF, 2013, Elevated CSF1 serum concentration predicts poor overall survival in women with early breast cancer. Endocr Relat Cancer 20(6):777–783. , DOI 10.1530/ERC-13-0198 Lawicki S, Glazewska EK, Sobolewska M, Bedkowska GE, SzmitkowskiM(2016, Plasma levels and diagnostic utility of macrophage Colony-stimulating factor, matrix Metalloproteinase-9, and tissue inhibitor of Metalloproteinases-1 as new biomarkers of breast Cancer. Ann Lab Med 36(3):223–229. , DOI 10. 3343/alm.2016.36.3.223 Rego SL, Helms RS, Dreau D, 2014, Breast tumor cell TACE-shed MCSF promotes pro-angiogenic macrophages through NF-κB signaling. Angiogenesis 17(3):573–585. , DOI 10.1007/ s10456-013-9405-2 Bjorklund M, Koivunen E, 2005, Gelatinase-mediated migration and invasion of cancer cells. Biochim Biophys Acta 1755(1):37– 69. , DOI 10.1016/j.bbcan.2005.03.001 Turpeenniemi-Hujanen T, 2005, Gelatinases, MMP-2 and -9, and their natural inhibitors as prognostic indicators in solid cancers. Biochimie 87(3–4):287–297. , DOI 10.1016/j.biochi. 2005.01.014 Remillard TC, Bratslavsky G, Jensen-Taubman S, Stetler- Stevenson WG, Bourboulia D, 2014, Molecular mechanisms of tissue inhibitor of metalloproteinase 2 in the tumor microenvironment. Mol Cell Ther 2:17. , DOI 10.1186/2052-8426-2-17 Walsh LA, Cepeda MA, Damjanovski S, 2012, Analysis of the MMP-dependent and independent functions of tissue inhibitor of metalloproteinase-2 on the invasiveness of breast cancer cells. J Cell Commun Signal 6(2):87–95. , DOI 10.1007/s12079- 011-0157-8 Chakraborti S, Mandal M, Das S, Mandal A, Chakraborti T, 2003, Regulation of matrix metalloproteinases: an overview. Mol Cell Biochem 253(1–2):269–285 Bourboulia D, Stetler-Stevenson WG, 2010, Matrix metalloproteinases, MMPs, and tissue inhibitors of metalloproteinases, TIMPs): positive and negative regulators in tumor cell adhesion. Semin Cancer Biol 20(3):161–168. , DOI 10.1016/j. semcancer.2010.05.002 Brew K, Nagase H, 2010, The tissue inhibitors of metalloproteinases, TIMPs): an ancient family with structural and functional diversity. Biochim Biophys Acta 1803(1):55–71. , DOI 10. 1016/j.bbamcr.2010.01.003 Kahari VM, Saarialho-Kere U, 1999, Matrix metalloproteinases and their inhibitors in tumour growth and invasion. Ann Med 31(1):34–45 Zola P, Macchi C, Cibula D, Colombo N, Kimmig R, Maggino T, Reed N, Kesic V, 2015, Follow-up in gynecological malignancies: a state of art. Int J Gynecol Cancer 25(7):1151–1164. https://doi. org/10.1097/IGC.0000000000000498 Eubank TD, GallowayM,MontagueCM,Waldman WJ, Marsh CB, 2003, M-CSF induces vascular endothelial growth factor production and angiogenic activity from human monocytes. J Immunol 171(5):2637–2643 Lawicki S, Bedkowska E, Gacuta-Szumarska E, Czygier M, Szmitkowski M, 2007, The plasma levels and diagnostic utility of stem cell factor and macrophage–colony stimulating factor in endometrial cancer patients. Przegl Lek 64(12):987–990 Lawicki S, Szmitkowski M, Wojtukiewicz M, 2006, The pretreatment plasma level and diagnostic utility of M-CSF in benign breast tumor and breast cancer patients. Clin Chim Acta 371(1–2):112– 116. , DOI 10.1016/j.cca.2006.02.033 Lawicki S, Gacuta-Szumarska E, Bedkowska GE, Szmitkowski M, 2012, Hematopoietic cytokines as tumor markers in gynecological malignancies. A multivariate analysis in epithelial ovarian cancer patients. Growth Factors 30(6):357–366. , DOI 10.3109/ 08977194.2012.724407 Mroczko B, Szmitkowski M, Wereszczynska-Siemiatkowska U, Okulczyk B, Kedra B, 2007, Pretreatment serum levels of hematopoietic cytokines in patients with colorectal adenomas and cancer. Int J Color Dis 22(1):33–38. , DOI 10.1007/s00384-006-0099-4 Liu H, Zhang H, Shen Z, Lin C,Wang X, Qin J, Qin X, Xu J, Sun Y, 2016, Increased expression of CSF-1 associates with poor prognosis of patients with gastric Cancer undergoing gastrectomy. Medicine, Baltimore, 95(9):e2675. , DOI 10.1097/MD. 0000000000002675 Fingleton B, 2006, Matrix metalloproteinases: roles in cancer and metastasis. Front Biosci 11:479–491 Pellikainen JM, Ropponen KM, Kataja VV, Kellokoski JK, Eskelinen MJ, Kosma VM, 2004, Expression of matrix metalloproteinase, MMP)-2 and MMP-9 in breast cancer with a special reference to activator protein-2, HER2, and prognosis. Clin Cancer Res 10(22): 7621–7628. , DOI 10.1158/1078-0432.CCR-04-1061 Crawford HC,Matrisian LM, 1994, Tumor and stromal expression of matrix metalloproteinases and their role in tumor progression. Invasion Metastasis 14(1–6):234–245 Lawicki S, Bedkowska GE, Gacuta-Szumarska E, Knapp P, SzmitkowskiM(2012, Pretreatment plasma levels and diagnostic utility of hematopoietic cytokines in cervical cancer or cervical intraepithelial neoplasia patients. Folia Histochem Cytobiol 50(2):213–219 Lawicki S, Zajkowska M, Glazewska EK, Bedkowska GE, Szmitkowski M, 2016, Plasma levels and diagnostic utility of MCSF, MMP-2 and its inhibitor TIMP-2 in the diagnostics of breast Cancer patients. Clin Lab 62(9):1661–1669 Vasiliades G, Kopanakis N, Vasiloglou M, Zografos G,Margaris H, Masselou K, Kokosi E, Liakakos T, 2012, Role of the hematopoietic cytokines SCF, IL-3, GM-CSF and M-CSF in the diagnosis of pancreatic and ampullary cancer. Int J Biol Markers 27(3):e186– e194. , DOI 10.5301/JBM.2012.9348 Lawicki S, Bedkowska GE, SzmitkowskiM(2013, VEGF,M-CSF and CA 15-3 as a new tumormarker panel in breast malignancies: a multivariate analysis with ROC curve. Growth Factors 31(3):98– 105. , DOI 10.3109/08977194.2013.797900 Ghosh A, Moirangthem A, Dalui R, Ghosh T, Bandyopadhyay A, Dasgupta A, Banerjee U, Jana N, Basu A, 2014, Expression of matrix metalloproteinase-2 and 9 in cervical intraepithelial neoplasia and cervical carcinoma among different age groups of premenopausal and postmenopausal women. J Cancer Res Clin Oncol 140(9):1585–1593. , DOI 10.1007/s00432-014-1695-2 Lawicki S, Bedkowska E, Gacuta-Szumarska E, Knapp P, Szmitkowski M, 2008, The plasma levels and diagnostic utility of stem cell factor, SCF, andmacrophage-colony stimulating factor, M-CSF, in cervical cancer patients. PolMerkur Lekarski 25(145): 38–42 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 791 EP 800 DI 10.1007/s12253-019-00626-z PG 10 ER PT J AU Wang, J Zhao, Y Xu, H Ma, J Liang, F Zou, Q Lin, F AF Wang, Jianfeng Zhao, Yan Xu, Hongyan Ma, Jun Liang, Feihai Zou, Qingxu Lin, Fengwu TI Silencing NID2 by DNA Hypermethylation Promotes Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung cancer; DNA methylation; NID2; Nude mice xenograft ID Lung cancer; DNA methylation; NID2; Nude mice xenograft AB To characterize the DNA methylation as well as exploring the relationship between NID2 methylation and the lung cancer development. Collecting chip data of 9 lung cancer samples and 11 adjacent normal samples from the Gene Expression Omnibus database. Tissues and cells NID2 gene methylation level was measured by methylation-specific PCR. NID2 mRNA level and protein level were validated by Real-Time PCR and Western blot separately. Functional study of lung cancer cells was performed with Cell Counting Kit-8 assay. Colony formation assay, Transwell assay, wound healing assay and low cytometry were performed. Finally, NID2 tumorigenesis in vivo was tested in nude mice xenograft models. Microarray analysis outcome present NID2 hypermethylation status in lung cancer tissues. High methylation and low mRNA expression levels of NID2 were detected. After NID2 demethylation or overexpression in cancer cells, cell viability, proliferation, migration as well as invasion ability decreased. Nevertheless, a significant enhancement in apoptosis rate were observed. Overexpressing NID2 or demethylation in lung cancer cells inhibited the tumorigenesis of lung cancer in nude mice. The mRNA and protein level of NID2 in tumors obtained from nude mice xenograft were unanimous with the in vitro assays’ outcome, which significantly decreased after overexpressing NID2 or demethylation. NID2 methylation reduces its expression level and promotes the development of lung cancer. C1 [Wang, Jianfeng] China-Japan Union Hospital of Jilin University, Department of Radiotherapy, 130031 Changchun, Jilin, China. [Zhao, Yan] The Second Hospital of Jilin University, Medical Examination Center, 130021 Changchun, Jilin, China. [Xu, Hongyan] Jilin Second People’s Hospital, Department of Oncology, 132011 Jilin, Jilin, China. [Ma, Jun] The Affiliated Hospital of Beihua University, Department of Radiotherapy, 132011 Jilin, Jilin, China. [Liang, Feihai] The Second Affiliated Hospital of Guangxi Medical University, Department of Cardiothoracic Surgery, 530007 Nanning, Guangxi, China. [Zou, Qingxu] China-Japan Union Hospital of Jilin University, Department of Thoracic Surgery, No. 126 freedom Avenue, 130031 Changchun, Jilin, China. [Lin, Fengwu] China-Japan Union Hospital of Jilin University, Department of Thoracic Surgery, No. 126 freedom Avenue, 130031 Changchun, Jilin, China. RP Lin, F (reprint author), China-Japan Union Hospital of Jilin University, Department of Thoracic Surgery, 130031 Changchun, China. EM fengwulin@hotmail.com CR Christopoulos P, Kirchner M, Bozorgmehr F, Endris V, Elsayed M, Budczies J, Ristau J, Penzel R, Herth FJ, Heussel CP, Eichhorn M, Muley T, Meister M, Fischer JR, Rieken S, Lasitschka F, Bischoff H, Sotillo R, Schirmacher P, Thomas M, Stenzinger A, 2018, Identification of a highly lethal V3(+, TP53(+, subset in ALK(+, lung adenocarcinoma. Int J Cancer. , DOI 10.1002/ijc. 31893 Morrow JD, Glass K, Cho MH, Hersh CP, Pinto-Plata V, Celli B, Marchetti N, Criner G, Bueno R, Washko G, Choi AMK, Quackenbush J, Silverman EK, DeMeo DL, 2018, Human Lung DNA methylation quantitative trait loci Colocalize with chronic obstructive pulmonary disease genome-wide association loci. Am J Respir Crit Care Med 197(10):1275–1284. , DOI 10. 1164/rccm.201707-1434OC Chen WQ, Li H, Sun KX, Zheng RS, Zhang SW, Zeng HM, Zou XN, Gu XY, He J, 2018, Report of Cancer incidence and mortality in China, 2014. Zhonghua zhong liu za zhi [Chinese journal of oncology] 40(1):5–13. , DOI 10.3760/cma.j.issn.0253- 3766.2018.01.002 Hong QY, Wu GM, Qian GS, Hu CP, Zhou JY, Chen LA, Li WM, Li SY, Wang K, Wang Q, Zhang XJ, Li J, Gong X, Bai CX, Lung Cancer Group of Chinese Thoracic S, Chinese Alliance Against Lung C, 2015, Prevention and management of lung cancer in China. Cancer 121 Suppl 17:3080–3088. , DOI 10.1002/ cncr.29584 Vlahopoulos S, Adamaki M, Khoury N, Zoumpourlis V, Boldogh I, 2018, Roles of DNA repair enzyme OGG1 in innate immunity and its significance for lung cancer. Pharmacol Ther. , DOI 10. 1016/j.pharmthera.2018.09.004 Feng X, Xie X, Zheng B, Peng C, Zhou H, Qin J, 2018, The more potential performance of nidogen 2 methylation by tissue or plasma DNAover brichoalveolar lavageDNAin diagnosis of nonsmall cell lung cancer. J Cancer Res Ther 14(Supplement):S341–S346. , DOI 10.4103/0973-1482.235352 Risch A, Plass C, 2008, Lung cancer epigenetics and genetics. Int J Cancer 123(1):1–7. , DOI 10.1002/ijc.23605 Schubeler D, 2015, Function and information content of DNA methylation. Nature 517(7534):321–326. , DOI 10.1038/ nature14192 Mehrmohamadi M, Mentch LK, Clark AG, Locasale JW, 2016, Integrative modelling of tumour DNA methylation quantifies the contribution of metabolism. Nat Commun 7:13666. , DOI 1038/ncomms13666 10. Gut P, Verdin E, 2013, The nexus of chromatin regulation and intermediary metabolism. Nature 502(7472):489–498. https://doi. org/10.1038/nature12752 Locasale JW, 2013, Serine, glycine and one-carbon units: cancer metabolism in full circle. Nat Rev Cancer 13(8):572–583. https:// doi.org/10.1038/nrc3557 Mentch SJ, Mehrmohamadi M, Huang L, Liu X, Gupta D, Mattocks D, Gomez Padilla P, Ables G, Bamman MM, Thalacker-Mercer AE, Nichenametla SN, Locasale JW, 2015, Histone methylation dynamics and gene regulation occur through the sensing of one-carbon metabolism. Cell Metab 22(5):861–873. , DOI 10.1016/j.cmet.2015.08.024 Anderson OS, Sant KE, Dolinoy DC, 2012, Nutrition and epigenetics: an interplay of dietary methyl donors, one-carbon metabolismandDNAmethylation. J Nutr Biochem23(8):853–859. https:// doi.org/10.1016/j.jnutbio.2012.03.003 Berman BP, Weisenberger DJ, Aman JF, Hinoue T, Ramjan Z, Liu Y, Noushmehr H, Lange CP, van Dijk CM, Tollenaar RA, Van Den Berg D, Laird PW, 2011, Regions of focal DNA hypermethylation and long-range hypomethylation in colorectal cancer coincide with nuclear lamina-associated domains. Nat Genet 44(1):40–46. https:// doi.org/10.1038/ng.969 LandauDA, Clement K, ZillerMJ, Boyle P, Fan J, Gu H, Stevenson K, Sougnez C, Wang L, Li S, Kotliar D, Zhang W, Ghandi M, Garraway L, Fernandes SM, Livak KJ, Gabriel S, Gnirke A, Lander ES, Brown JR, Neuberg D, Kharchenko PV, Hacohen N, Getz G, Meissner A,Wu CJ, 2014, Locally disordered methylation forms the basis of intratumor methylome variation in chronic lymphocytic leukemia. Cancer Cell 26(6):813–825. , DOI 10. 1016/j.ccell.2014.10.012 Geng J, Sun J, Lin Q, Gu J, Zhao Y, Zhang H, Feng X, He Y,Wang W, Zhou X, Yu J, 2012, Methylation status of NEUROG2 and NID2 improves the diagnosis of stage I NSCLC. Oncol Lett 3(4): 901–906. , DOI 10.3892/ol.2012.587 Vasiljevic N, Ahmad AS, Beesley C, Thorat MA, Fisher G, Berney DM, Moller H, Yu Y, Lu YJ, Cuzick J, Foster CS, Lorincz AT, 2013, Association betweenDNA methylation ofHSPB1 and death in low Gleason score prostate cancer. Prostate Cancer Prostatic Dis 16(1):35–40. , DOI 10.1038/pcan.2012.47 Wang Q, Williamson M, Bott S, Brookman-Amissah N, Freeman A, Nariculam J, Hubank MJ, Ahmed A, Masters JR, 2007, Hypomethylation ofWNT5A, CRIP1 and S100P in prostate cancer. Oncogene 26(45):6560–6565. , DOI 10.1038/sj.onc. 1210472 Chai AW, Cheung AK, Dai W, Ko JM, Ip JC, Chan KW, Kwong DL, NgWT, Lee AW, Ngan RK, Yau CC, Tung SY, Lee VH, Lam AK, Pillai S, Law S, Lung ML, 2016, Metastasis-suppressing NID2, an epigenetically-silenced gene, in the pathogenesis of nasopharyngeal carcinoma and esophageal squamous cell carcinoma. Oncotarget 7(48):78859–78871. , DOI 10.18632/ oncotarget.12889 Kohfeldt E, Sasaki T, Gohring W, Timpl R, 1998, Nidogen-2: a new basement membrane protein with diverse binding properties. J Mol Biol 282(1):99–109. , DOI 10.1006/jmbi.1998.2004 Mokkapati S, BechtelM, Reibetanz M, Miosge N, Nischt R, 2012, Absence of the basement membrane component nidogen 2, but not of nidogen 1, results in increased lung metastasis in mice. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society 60(4):280–289. , DOI 10. 1369/0022155412436586 Ulazzi L, Sabbioni S, Miotto E, Veronese A, Angusti A, Gafa R, Manfredini S, Farinati F, Sasaki T, Lanza G, Negrini M, 2007, Nidogen 1 and 2 gene promoters are aberrantly methylated in human gastrointestinal cancer. Mol Cancer 6:17. , DOI 10. 1186/1476-4598-6-17 Esteller M, Sanchez-Cespedes M, Rosell R, Sidransky D, Baylin SB, Herman JG, 1999, Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res 59(1):67–70 Li B, Lu Q, Song ZG, Yang L, Jin H, Li ZG, Zhao TJ, Bai YF, Zhu J, Chen HZ, Xu ZY, 2013, Functional analysis of DNA methylation in lung cancer. Eur Rev Med Pharmacol Sci 17(9):1191–1197 Hsiao SH, Huang TH, Leu YW, 2009, Excavating relics of DNA methylation changes during the development of neoplasia. Semin Cancer Biol 19(3):198–208. , DOI 10.1016/j.semcancer. 2009.02.015 Timpl R, Brown JC, 1996, Supramolecular assembly of basement membranes. BioEssays : news and reviews in molecular, cellular and developmental biology 18(2):123–132. , DOI 10. 1002/bies.950180208 Dong LJ, Hsieh JC, Chung AE, 1995, Two distinct cell attachment sites in entactin are revealed by amino acid substitutions and deletion of the RGD sequence in the cysteine-rich epidermal growth factor repeat 2. J Biol Chem 270(26):15838–15843 Wu C, Chung AE, McDonald JA, 1995, A novel role for alpha 3 beta 1 integrins in extracellular matrix assembly. J Cell Sci 108(Pt 6):2511–2523 Dedhar S, Jewell K, Rojiani M, Gray V, 1992, The receptor for the basement membrane glycoprotein entactin is the integrin alpha 3/beta 1. J Biol Chem 267(26):18908–18914 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 801 EP 811 DI 10.1007/s12253-019-00609-0 PG 11 ER PT J AU Regos, E Karaszi, K Reszegi, A Kiss, A Schaff, Zs Baghy, K Kovalszky, I AF Regos, Eszter Karaszi, Katalin Reszegi, Andrea Kiss, Andras Schaff, Zsuzsa Baghy, Kornelia Kovalszky, Ilona TI Syndecan-1 in Liver Diseases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Syndecan-1; Liver; Cirrhosis; Metastasis; Hepatocellular carcinoma ID Syndecan-1; Liver; Cirrhosis; Metastasis; Hepatocellular carcinoma AB Liver diseases such as liver cirrhosis, primary and metastatic liver cancers are still a major medical challenge. Syndecan-1 is one of the most important proteoglycans in the liver. Syndecan-1 is normally expressed on the surfaces of hepatocytes and cholangiocytes. Due to liver diseases the amount of syndecan-1 increases in the liver. Despite the emerging data of the biological function of syndecan-1, the clinical usefulness of this proteoglycan is still unknown. In our study we correlated syndecan-1 expression to clinicopathological data. We found that syndecan-1 proved to be a good marker for hepatitis C virus based hepatocellular carcinoma and increased with liver dysfunction. C1 [Regos, Eszter] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Street 26, 1085 Budapest, Hungary. [Karaszi, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Street 26, 1085 Budapest, Hungary. [Reszegi, Andrea] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Street 26, 1085 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi street 93, 1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi street 93, 1091 Budapest, Hungary. [Baghy, Kornelia] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Street 26, 1085 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi Street 26, 1085 Budapest, Hungary. RP Kovalszky, I (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM koval@korb1.sote.hu CR Schuppan D, Afdhal NH, 2008, Liver cirrhosis. Lancet 371(9615): 838–851. , DOI 10.1016/S0140-6736(08)60383-9 de Ridder J, de Wilt JHW, Simmer F, Overbeek L, Lemmens V, Nagtegaal I, 2016, Incidence and origin of histologically confirmed liver metastases: an explorative case-study of 23,154 patients. Oncotarget 7(34):55368–55376. , DOI 10.18632/ oncotarget.10552 Reddy SS, Civan JM(2016, From child-Pugh to model for end-stage liver disease: deciding who needs a liver transplant. Med Clin North Am 100(3):449–464. , DOI 10.1016/j.mcna.2015.12.002 Choi Y, Chung H, Jung H, Couchman JR, Oh ES, 2011, Syndecans as cell surface receptors: unique structure equates with functional diversity. Matrix Biol 30(2):93–99. , DOI 10.1016/j. matbio.2010.10.006 Sanderson RD, BorsetM(2002, Syndecan-1 in B lymphoid malignancies. Ann Hematol 81(3):125–135. , DOI 10.1007/ s00277-002-0437-8 Stepp MA, Pal-Ghosh S, Tadvalkar G, Pajoohesh-Ganji A, 2015, Syndecan-1 and its expanding list of contacts. Adv Wound Care 4(4):235–249. , DOI 10.1089/wound.2014.0555 Theocharis AD, Skandalis SS, Tzanakakis GN, Karamanos NK, 2010, Proteoglycans in health and disease: novel roles for proteoglycans in malignancy and their pharmacological targeting. FEBS J 277(19):3904–3923. , DOI 10.1111/j.1742-4658.2010. 07800.x Manon-Jensen T, Itoh Y, Couchman JR, 2010, Proteoglycans in health and disease: the multiple roles of syndecan shedding. FEBS J 277(19):3876–3889. , DOI 10.1111/j.1742-4658. 2010.07798.x Zvibel I, Halfon P, Fishman S, Penaranda G, Leshno M, Or AB, Halpern Z, Oren R, 2009, Syndecan 1, CD138, serum levels: a novel biomarker in predicting liver fibrosis stage in patients with hepatitis C. Liver Int 29(2):208–212. , DOI 10.1111/j. 1478-3231.2008.01830.x Yilmaz Y, Eren F, Colak Y, Senates E, Celikel CA, Imeryuz N, 2012, Hepatic expression and serum levels of syndecan 1, CD138, in patients with nonalcoholic fatty liver disease. Scand J Gastroenterol 47(12):1488–1493. , DOI 10.3109/ 00365521.2012.725093 Kovalszky I, Dudas J, Gallai M, Hollosi P, Tatrai P, Tatrai E, Zs S, 2004, Proteoglikanok a majban. Magy Onkol 48(3):207–213 Baghy K, Tatrai P, Regos E, Kovalszky I, 2016, Proteoglycans in liver cancer. World J Gastroenterol 22(1):379–393. , DOI 10.3748/wjg.v22.i1.379 Regos E, Abdelfattah HH, Reszegi A, Szilak L, Werling K, Szabo G, Kiss A, Schaff Z, Kovalszky I, Baghy K, 2018, Syndecan-1 inhibits early stages of liver fibrogenesis by interfering with TGFbeta1 action and upregulating MMP14. Matrix Biol. https:// doi.org/10.1016/j.matbio.2018.02.008 Deng Y, Foley EM, Gonzales JC, Gordts PL, Li Y, Esko JD, 2012, Shedding of syndecan-1 from human hepatocytes alters very low density lipoprotein clearance. HEPATOLOGY 55(1):277–286. , DOI 10.1002/hep.24626 Afratis NA, Nikitovic D, Multhaupt HA, Theocharis AD, Couchman JR, Karamanos NK, 2017, Syndecans - key regulators of cell signaling and biological functions. FEBS J 284(1):27–41. , DOI 10.1111/febs.13940 Rapraeger AC, 2013, Synstatin: a selective inhibitor of the syndecan-1-coupled IGF1R-alphavbeta3 integrin complex in tumorigenesis and angiogenesis. FEBS J 280(10):2207–2215. , DOI 10.1111/febs.12160 Mitsou I, Multhaupt HAB, Couchman JR, 2017, Proteoglycans, ion channels and cell-matrix adhesion. Biochem J 474(12):1965– 1979. , DOI 10.1042/bcj20160747 Shi Q, Jiang J, Luo G, 2013, Syndecan-1 serves as themajor receptor for attachment of hepatitis C virus to the surfaces of hepatocytes. J Virol 87(12):6866–6875. , DOI 10.1128/jvi.03475-12 Wiesner R, Lake JR, Freeman RB, Gish RG, 2006)Model for endstage liver disease, MELD, exception guidelines. Liver Transpl 12(12 Suppl 3):S85–S87. , DOI 10.1002/lt.20961 Nam EJ, Hayashida K, Aquino RS, Couchman JR, Kozar RA, Liu J, Park PW(2017, Syndecan-1 limits the progression of liver injury and promotes liver repair in acetaminophen-induced liver injury in mice. HEPATOLOGY 66(5):1601–1615. , DOI 10.1002/ hep.29265 Grigorov B, Reungoat E, Gentil Dit Maurin A, Varbanov M, Blaising J, Michelet M, Manuel R, Parent R, Bartosch B, Zoulim F, Ruggiero F, Pecheur EI, 2017, Hepatitis C virus infection propagates through interactions between Syndecan-1 and CD81 and impacts the hepatocyte glycocalyx. Cell Microbiol 19(5). https:// doi.org/10.1111/cmi.12711 Pap Z, Pavai Z, Denes L, Kovalszky I, Jung J, 2009, An immunohistochemical study of colon adenomas and carcinomas: Ecadherin, Syndecan-1, Ets-1. Pathol Oncol Res 15(4):579–587. , DOI 10.1007/s12253-009-9157-x NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 813 EP 819 DI 10.1007/s12253-019-00617-0 PG 7 ER PT J AU Sawicka, E Kratz, ME Szymanska, B Guzik, A Wesolowski, A Kowal, P Pawlik-Sobecka, L Piwowar, A AF Sawicka, Ewa Kratz, Maria Ewa Szymanska, Beata Guzik, Anna Wesolowski, Artur Kowal, Pawel Pawlik-Sobecka, Lilla Piwowar, Agnieszka TI Preliminary Study on Selected Markers of Oxidative Stress, Inflammation and Angiogenesis in Patients with Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; Cancer grade and stage; Oxidative stress; Inflammation; Angiogenesis ID Bladder cancer; Cancer grade and stage; Oxidative stress; Inflammation; Angiogenesis AB In recent years, bladder cancer (BC) has been reported as one of the most commonly occurring cancers among older people, and its detection is still difficult. Therefore, there is a need to search for additional useful markers of disease. Some studies indicate the important roles of inflammation and oxidative stress (OS) in bladder tumour pathogenesis. The aim of this study was to examine the levels of selected markers of OS, inflammation and angiogenesis in blood plasma/serum samples derived from patients with BC, and a healthy control group. Moreover the degrees of change and strength of correlation between values of the analysed markers and tumour stage or grade were estimated. Concentrations of: malondialdehyde (MDA) and advanced oxidation protein products (AOPP), and total antioxidant status (TAS) divided into slow (TAS-s) and fast (TAS-f) antioxidants (spectrophotometric measurement), angiogenin (ANG) (immunoenzymatic method) and C-reactive protein (CRP) (immunoturbidimetric method) were determined in both the studied groups. The majority of values of the examined parameters were significantly higher among patients, while subfractions of TAS were significantly lower in comparison to the control group. Moreover, different values and different strengths of correlation between the examined parameters and cancer stage or grade were noticed. The most significant changes for CRP were observed in T2 and for MDA in G3, while the lowest TAS-f activity was revealed in G1 patients. Increased values of OS parameters, angiogenesis and inflammation markers, in combination with reduced TAS subfractions activity in BC are important in its pathogenesis and will be helpful in estimation of patients’ condition. C1 [Sawicka, Ewa] Wroclaw Medical University, Faculty of Pharmacy with Division of Laboratory Diagnostics, Department of Toxicology, Borowska Street 211, 50-556 Wroclaw, Poland. [Kratz, Maria Ewa] Wroclaw Medical University, Faculty of Pharmacy with Division of Laboratory Diagnostics, Department of Laboratory Diagnostics, Borowska Street 211, 50-556 Wroclaw, Poland. [Szymanska, Beata] Wroclaw Medical University, Faculty of Pharmacy with Division of Laboratory Diagnostics, Department of Toxicology, Borowska Street 211, 50-556 Wroclaw, Poland. [Guzik, Anna] Wroclaw Medical University, Faculty of Pharmacy with Division of Laboratory Diagnostics, Department of Toxicology, Borowska Street 211, 50-556 Wroclaw, Poland. [Wesolowski, Artur] Wroclaw Medical University, Faculty of Pharmacy with Division of Laboratory Diagnostics, Students Scientific Society at the Department of Toxicology, Borowska Street 211, 50-556 Wroclaw, Poland. [Kowal, Pawel] Wroclaw Medical University, Department of Urology and Oncological Urology, H. M. Kamienskiego Street 73a, 51-124 Wroclaw, Poland. [Pawlik-Sobecka, Lilla] Wroclaw Medical University, Faculty of Pharmacy with Division of Laboratory Diagnostics, Department of Laboratory Diagnostics, Borowska Street 211, 50-556 Wroclaw, Poland. [Piwowar, Agnieszka] Wroclaw Medical University, Faculty of Pharmacy with Division of Laboratory Diagnostics, Department of Toxicology, Borowska Street 211, 50-556 Wroclaw, Poland. RP Kratz, ME (reprint author), Wroclaw Medical University, Faculty of Pharmacy with Division of Laboratory Diagnostics, Department of Laboratory Diagnostics, 50-556 Wroclaw, Poland. EM ewa.kratz@umed.wroc.pl CR Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JWW, Comber H, Forman D, Bray F, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403 Didkowska J,Wojciechowska U, Sosnowski R In-depth analysis of epidemiological data, geographic differentiation and differentiation of morbidity and mortality due to urothelial diseases National Cancer Registry, Center for Oncology - Maria Sklodowska-Curie Institute. http://onkologia.org.pl/raporty/. Access 15 Dec 2015 Didkowska J,Wojciechowska U Malignant neoplasms in Poland in 2013. National Cancer Registry, Center for Oncology - Maria Sklodowska-Curie Institute. http://onkologia.org.pl/raporty/. Access 3 Mar 2016 ValkoM, Izakovic M, Mazur M, Rhodes CJ, Telser J, 2004, Role of oxygen radicals in DNA damage and cancer incidence. Mol Cell Biochem 266:37–56 Kong Q, Beel JA, Lillehei KO, 2000, A threshold concept for cancer therapy. Med Hypotheses 5:529–535 Brown NS, Bicknell R, 2001, Hypoxia and oxidative stress in breast cancer: oxidative stress, its effects on the growth, metastatic potential, and response to therapy of breast cancer. Breast Cancer Res 3:323–327 Kumar B, Koul S, Khandrika L, Meacham RB, Koul HK, 2008, Oxidative stress in inherent in prostate cancer cells and is required for aggressive phenotype. Cancer Res 68:1777–1785 Toyokuni S, Okamoto K, Yodoi J, Hiai H, 1995, Persistent oxidative stress in cancer. FEBS Lett 358:1–3 SaedGM, Diamond MP, FletcherNM(2017, Updates of the role of oxidative stress in the pathogenesis of ovarian cancer. Gynecol Oncol 145:595–602 Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB, 2010, Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 49:1603–1616 Sawicka E, Lisowska A, Kowal P, Dlugosz A, 2015, The role of oxidative stress in bladder cancer. [Polish]. Postepy HigMed Dosw 69:744–752 Kattoor JK, Pothineni NV, Palagiri D, Mehta JL, 2017, Oxidative stress in atherosclerosis. Curr Atheroscler Rep 19:42–49 Tsikas D, 2017, Assessment of lipid peroxidation by measuring malondialdehyde, MDA, and relatives in biological samples: analytical and biological challenges. Anal Biochem 524:13–30 Liu Q, Luo Q, Halim A, Song G, 2017, Targeting lipid metabolism of cancer cells: a promising therapeutic strategy for cancer. Cancer Lett 401:39–45 Federico A, Morgillo F, Tuccillo C, Ciardiello F, Loguercio C, 2007, Chronic inflammation and oxidative stress in human carcinogenesis. Int J Cancer 121:2381–2386 Witko-Sarsat V, Gausson V, Descamps-Latscha B, 2003, Are advanced oxidation protein products potential uremic toxins? Kidney Int Suppl 84:11–14 Piwowar A, 2010, Advanced oxidation products of proteins. Part I. Mechanism of formation, structure and properties. [Polish]. Pol Merkur Lekarski 28:166–169 Scibior-Bentkowska D, Czeczot H, 2009, Cancer cells and oxidative stress. Adv Hyg Exp Med 63:58–72 Yalcini O, Karatas F, Erulas FA, Ozdemir E, 2003, The levels of glutathione peroxidase, vitamin A, E, C and lipid peroxidation in patients with transitional cell carcinoma of the bladder. BJU Int 93: 863–866 Mazdak H, Zia H, 2012, Vitamin E reduces superficial bladder cancer recurrence: a randomized controlled trial. Int J Prev Med 3:110–115 Fus LP, Gornicka B, 2016, Role of angiogenesis in urothelial bladder carcinoma. Cent European J Urol 69:258–263 Shariat SF, Youseff RF, Gupta A, Chade DC, Karakiewicz PI, Isbarn H, 2010, Association of angiogenesis related markers with bladder cancer outcomes and other molecular markers. J Urol 183: 1744–1750 Donmez G, Sullu Y, Baris S, Yildiz L, Aydin O, Karagoz F, Kandemir B, 2009, Vascular endothelial growth factor, VEGF), matrix metalloproteinase-9, MMP-9), and thrombospondin-1, TSP-1, expression in urothelial carcinomas. Pathol Res Pract 205:854–857 Inoue K, Slaton JW, Kim SJ, Perrotte P, Eve BY, Bar-Eli M, Radinsky R, Dinney CP, 2000, Interleukin 8 expression regulates tumorigenicity and metastasis in human bladder cancer. Cancer Res 60:2290–2299 Li S, Hu G, 2010, Angiogenin-mediated rRNA transcription in cancer and neurodegeneration. Int J Biochem Mol Biol 1:26–35 Eissa S, SwellamM, Labib RA, El-Zayat T, El Ahmady O, 2009, A panel of angiogenic factors for early bladder cancer detection: enzyme immunoassay and Western blot. J Urol 181:1353–1360 Shu J, Huang M, Tian Q, Shui Q, Zhou Y, Chen J, 2015, Downregulation of angiogenin inhibits the growth and induces apoptosis in human bladder cancer cells through regulating AKT/ mTOR signaling pathway. J Mol Histol 46:157–171 Grivennikov SI, Greten FR, Karin M, 2010, Immunity, inflammation and cancer. Cell 140:883–899 Grivennikov SI, Karin M, 2010, Inflammation and oncogenesis: a vicious connection. Curr Opin Genet Dev 20:65–71 Gonda TA, Tu S, Wang TC, 2009, Chronic inflammation, the tumor microenvironment and carcinogenesis. Cell Cycle 8:2005– 2013 Eblin KE, Bredfeldt TG, Gandolfi AJ, 2008, Immortalized human urothelial cells as a model of arsenic-induced bladder cancer. Toxicology 248:67–76 Meliker JR, Nriagu JO, 2007, Arsenic in drinking water and bladder cancer: review of epidemiological evidence. Trace Metals and other Contaminants in the Environment 9:551–584 Romanenko A, Kakehashi A, Morimura K, Wanibuchi H, Wei M, Vozianov A, Fukushima S, 2009, Urinary bladder carcinogenesis induced by chronic exposure to persistent low dose ionizing radiation after Chernobyl accident. Carcinogenesis 30:1821–1831 Schetter AJ, Heegaard NHH, Harris CC, 2010, Inflammation and cancer: interweaving micro RNA, free radical, cytokine and p53 patchways. Carcinogenesis 31:37–49 Murata M, Thanan R, Ma N, Kawanishi S, 2012, Role of nitrative and oxidative DNA damage in inflammation-related carcinogenesis. J Biomed Biotechnol 2012:623019. , DOI 10.1155/ 2012/623019 Bostrom PJ, van Rhijn BWG, Fleshner N, Finelli A, Jewett M, Thoms J, Hanna S, Kuk C, Zlotta AR, 2010, Staging and staging errors in bladder cancer. Eur Urol Suppl 9:2–9. , DOI 10. 1016/j.eursup.2010.01.005 Yoshioka T, Kawada K, Shimada T, Mori M, 1979, Lipid peroxidation in maternal and cord blood and protective mechanism against activated-oxygen toxicity in the blood. Am J Obstet Gynecol 135:372–376 Re R, Pellegrini N, Proteggente A, Pannala A, YangM, Rice-Evans C, 1999, Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free Radic Biol Med 26:1231–1237 Badjatia N, Satyam A, Singh P, Seth A, Sharma A, 2010, Altered antioxidant status and lipid peroxidation in Indian patients with urothelial bladder carcinoma. Urol Oncol 28:360–367 Nechuta S, Lu W, Chen Z, Zheng Y, Gu K, Cai H, Zheng W, Shu XO, 2011, Vitamin supplement use during breast cancer treatment and survival: a prospective cohort study. Cancer Epidemiol Biomark Prev 20:262–271 Gecit I, Aslan M, GunesM, Pirincci N, Esen R, Demir H, Ceylan K, 2012, Serum prolidase activity, oxidative stres and nitric oxide levels in patients with bladder cancer. J Cancer Res Clin Oncol 138:739–743 Kosova F, Cetin B, Akinici M et al, 2007, Advanced oxidation protein products, ferrous oxidation in xylenol orange and malondialdehyde levels in thyroid cancer. Ann Surg Oncol 14: 2016–2020 Gakis G, Todenhӧfer T, Renninger M et al, 2011, Development of a new outcome prediction model in carcinoma invading the bladder based on preoperative serum C-reactive protein and standard pathological risk factors the TNR-C score. BJU Int 108:1800–1805 Morka J, Drozdz J, 2006, CRP - a high risk indicator and a new therapeutic target, CRP – wskaznik podwyzszonego ryzyka i nowy cel terapii). Forum Kardiol 11:27–31 Saito K, Kihara K, 2013, Role of C-reactive protein in urological cancers: a useful biomarker for predicting outcomes. Int J Urol 20: 161–171 Zhao H, Grossman B, George L et al, 2005, Increased plasma levels of angiogenin and the risk of bladder carcinoma. Cancer 104:30–35 Urquidi V, Goodison S, Kim J, Chang M, Daib Y, Rosser CJ, 2012, VEGF, CA9 and angiogenin as a urinary biomarker for bladder cancer detection. Urology 79:1185.e1–1185.e6 Radomska-Lesniewska DM, Balan BJ, Skopinski P, 2017, Angiogenesis modulation by exogenous antioxidants. Cent Eur J Immunol 42:370–376 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 821 EP 831 DI 10.1007/s12253-019-00620-5 PG 11 ER PT J AU Arabkari, V Clancy, E Dwyer, MR Kerin, JM Kalinina, O Holian, E Newell, J Smith, JT AF Arabkari, Vahid Clancy, Eoin Dwyer, M Roisin Kerin, J Michael Kalinina, Olga Holian, Emma Newell, John Smith, J Terry TI Relative and Absolute Expression Analysis of MicroRNAs Associated with Luminal A Breast Cancer– A Comparison SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MicroRNAs; Luminal A breast cancer; Relative quantification; Absolute quantification ID MicroRNAs; Luminal A breast cancer; Relative quantification; Absolute quantification AB MicroRNAs, as small non-coding regulatory RNAs, play crucial roles in various aspects of breast cancer biology. They have prognostic and diagnostic value, which makes them very interesting molecules to investigate. Reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) is the gold standard method to analyse miRNA expression in breast cancer patients. This study investigated two RT-qPCR methods (absolute and relative) to determine the expression of ten miRNAs in whole blood samples obtained from luminal A breast cancer patients compared to healthy controls. Whole blood samples were collected from 38 luminal A breast cancer patients and 20 healthy controls in Paxgene blood RNA tubes. Total RNA was extracted and analysed by relative and absolute RT-qPCR. For relative RT-qPCR, miR-16 was used as an endogenous control. For absolute RT-qPCR, standard curves were generated using synthetic miRNA oligonucleotides to determine the absolute copy number of each miRNA. Of the ten miRNAs that were analysed, the absolute RT-qPCR method identified six miRNAs (miR-16, miR-145, miR-155, miR- 451a, miR-21 and miR-486) that were upregulated and one miRNA (miR-195) that was downregulated. ROC curve and AUC analysis of the data found that the combination of three miRNAs (miR-145, miR-195 and miR-486) had the best diagnostic value for luminal A breast cancer with an AUC of 0.875, with 76% sensitivity and 81% specificity. On the other hand, the relative RTqPCR method identified two miRNAs (miR-155 and miR-486) that were upregulated and miR-195, which was downregulated. Using this approach, the combination of three miRNAs (miR-155, miR-195 and miR-486) was showed to have an AUC of 0.657 with 65% sensitivity and 69% specificity. We conclude that miR-16 is not a suitable normalizer for the relative expression profiling of miRNAs in luminal A breast cancer patients. Compared to relative quantification, absolute quantification assay is a better method to determine the expression level of circulating miRNAs in Luminal A breast cancer. C1 [Arabkari, Vahid] NUI Galway, School of Natural Sciences, National Centre for Biomedical Engineering Science (NCBES), Molecular Diagnostics Research GroupGalway, Ireland. [Clancy, Eoin] NUI Galway, School of Natural Sciences, National Centre for Biomedical Engineering Science (NCBES), Molecular Diagnostics Research GroupGalway, Ireland. [Dwyer, M Roisin] NUI Galway, School of Medicine, Lambe Institute for Translational Research, Discipline of SurgeryGalway, Ireland. [Kerin, J Michael] NUI Galway, School of Medicine, Lambe Institute for Translational Research, Discipline of SurgeryGalway, Ireland. [Kalinina, Olga] NUI Galway, School of Mathematics, Statistics and Applied Mathematics, Clinical Research FacilityGalway, Ireland. [Holian, Emma] NUI Galway, School of Mathematics, Statistics and Applied Mathematics, Clinical Research FacilityGalway, Ireland. [Newell, John] NUI Galway, School of Mathematics, Statistics and Applied Mathematics, Clinical Research FacilityGalway, Ireland. [Smith, J Terry] NUI Galway, School of Natural Sciences, National Centre for Biomedical Engineering Science (NCBES), Molecular Diagnostics Research GroupGalway, Ireland. RP Arabkari, V (reprint author), NUI Galway, School of Natural Sciences, National Centre for Biomedical Engineering Science (NCBES), Molecular Diagnostics Research Group, Galway, Ireland. EM v.arabkari1@nuigalway.ie CR WHO.World Health Organization, 2015, Global health observatory data repository. 2015. Number of deaths, World, by cause. http:// wwww.hoint/mediacentre/factsheets/fs310/en/ Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer Journal international du cancer. 2015;136(5):E359–E386. , DOI 10.1002/ijc.29210 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H et al, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49(6):1374–1403. , DOI 10.1016/j.ejca.2012.12. 027 PerouCM, Sorlie T, EisenMB, van de Rijn M, Jeffrey SS, Rees CA et al, 2000, Molecular portraits of human breast tumours. Nature. 406(6797):747–752. , DOI 10.1038/35021093 Sorlie T, Wang Y, Xiao C, Johnsen H, Naume B, Samaha RR et al, 2006, Distinct molecular mechanisms underlying clinically relevant subtypes of breast cancer: gene expression analyses across three different platforms. BMC Genomics 7:127. , DOI 10.1186/1471-2164-7-127 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, 2011, Strategies for subtypes–dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol 22(8):1736–1747. , DOI 10.1093/annonc/ mdr304 Rouzier R, Perou CM, Symmans WF, Ibrahim N, Cristofanilli M, Anderson K et al, 2005, Breast cancer molecular subtypes respond differently to preoperative chemotherapy. Clin Cancer Res 11(16): 5678–5685. , DOI 10.1158/1078-0432.ccr-04-2421 Kumar MS, Lu J, Mercer KL, Golub TR, Jacks T, 2007, Impaired microRNA processing enhances cellular transformation and tumorigenesis. Nat Genet 39(5):673–677. , DOI 10.1038/ng2003 Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR, 2005, MicroRNA expression profiles classify human cancers. Nature. 435(7043):834–838. https:// doi.org/10.1038/nature03702 Zen K, Zhang C-Y, 2012, CirculatingMicroRNAs: a novel class of biomarkers to diagnose and monitor human cancers. Med Res Rev 32(2):326–348. , DOI 10.1002/med.20215 Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, Menard S, Palazzo JP, Rosenberg A, Musiani P, Volinia S, Nenci I, Calin GA, Querzoli P, Negrini M, Croce CM, 2005, MicroRNA gene expression deregulation in human breast cancer. Cancer Res 65(16):7065–7070. , DOI 10.1158/0008-5472.CAN-05-1783 Heneghan HM, Miller N, Lowery AJ, Sweeney KJ, Kerin MJ, 2009, 2009, MicroRNAs as Novel Biomarkers for Breast Cancer. J Oncol:950201. , DOI 10.1155/2010/950201 Tahiri A, Leivonen SK, Luders T, Steinfeld I, Ragle Aure M, Geisler J et al, 2014, Deregulation of cancer-related miRNAs is a common event in both benign and malignant human breast tumors. Carcinogenesis. 35(1):76–85. , DOI 10.1093/carcin/ bgt333 McDermott AM, Miller N, Wall D, Martyn LM, Ball G, Sweeney KJ et al, 2014, Identification and validation of oncologic miRNA biomarkers for luminal A-like breast cancer. PLoS One 9(1): e87032. , DOI 10.1371/journal.pone.0087032 Chan M, Liaw CS, Ji SM, Tan HH, Wong CY, Thike AA et al, 2013, Identification of circulating microRNA signatures for breast cancer detection. Clin Cancer Res 19(16):4477–4487. https://doi. org/10.1158/1078-0432.CCR-12-3401 Pfaffl MW, Horgan GW, Dempfle L, 2002, Relative expression software tool, REST, for group-wise comparison and statistical analysis of relative expression results in real-time PCR. Nucleic Acids Res 30(9):e36 Pfaffl MW, 2001, A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 29(9):e45 Agarwal V, Bell GW, Nam JW, Bartel DP, 2015, Predicting effective microRNA target sites in mammalian mRNAs. Elife 4. https:// doi.org/10.7554/eLife.05005 Klein D, 2002, Quantification using real-time PCR technology: applications and limitations. Trends Mol Med 8(6):257–260 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), Method. Methods 25(4):402–408. , DOI 10.1006/ meth.2001.1262 Mannhalter C, Koizar D, Mitterbauer G, 2000, Evaluation of RNA isolation methods and reference genes for RT-PCR analyses of rare target RNA. Clin Chem Lab Med 38(2):171–177. , DOI 10.1515/cclm.2000.026 Karge WH 3rd, Schaefer EJ, Ordovas JM, 1998, Quantification of mRNA by polymerase chain reaction, PCR, using an internal standard and a nonradioactive detection method. Methods Mol Biol 110:43–61. , DOI 10.1385/1-59259-582-0:43 Bustin SA, 2000, Absolute quantification ofmRNAusing real-time reverse transcription polymerase chain reaction assays. J Mol Endocrinol 25(2):169–193 Pfaffl MW, Hageleit M, 2001, Validities of mRNA quantification using recombinantRNAand recombinantDNA external calibration curves in real-time RT-PCR. Biotechnol Lett 23(4):275–282. , DOI 10.1023/a:1005658330108 Hindson CM, Chevillet JR, Briggs HA, Gallichotte EN, Ruf IK, Hindson BJ, Vessella RL, TewariM(2013, Absolute quantification by droplet digital PCR versus analog real-time PCR. Nat Methods 10(10):1003–1005. , DOI 10.1038/nmeth.2633 Pohl G, Shih IM, 2004, Principle and applications of digital PCR. Expert Rev Mol Diagn 4(1):41–47 Mar-Aguilar F, Mendoza-Ramirez JA, Malagon-Santiago I, Espino-Silva PK, Santuario-Facio SK, Ruiz-Flores P et al, 2013, Serum circulating microRNA profiling for identification of potential breast cancer biomarkers. Dis Markers 34(3):163–169 Kodahl AR, LyngMB, Binder H, Cold S, Gravgaard K, Knoop AS, Ditzel HJ, 2014, Novel circulating microRNA signature as a potential non-invasive multi-marker test in ER-positive early-stage breast cancer: a case control study. Mol Oncol 8(5):874–883. , DOI 10.1016/j.molonc.2014.03.002 Ng EK, Li R, Shin VY, Jin HC, Leung CP, Ma ES et al, 2013, Circulating microRNAs as specific biomarkers for breast cancer detection. PLoS One 8(1):e53141. , DOI 10.1371/journal. pone.0053141 Rask L, Balslev E, Sokilde R, Hogdall E, Flyger H, Eriksen J et al, 2014, Differential expression of miR-139, miR-486 and miR-21 in breast cancer patients sub-classified according to lymph node status. Cell Oncol, Dordr, 37(3):215–227. , DOI 10.1007/ s13402-014-0176-6 Sochor M, Basova P, Pesta M, Dusilkova N, Bartos J, Burda P, Pospisil V, Stopka T, 2014, Oncogenic microRNAs: miR-155, miR-19a, miR-181b, and miR-24 enable monitoring of early breast cancer in serum. BMC Cancer 14:448. , DOI 10.1186/ 1471-2407-14-448 Madhavan D, Zucknick M, Wallwiener M, Cuk K, Modugno C, Scharpff M et al, 2012, Circulating miRNAs as surrogate markers for circulating tumor cells and prognostic markers in metastatic breast cancer. Clin Cancer Res 18(21):5972–5982. , DOI 10.1158/1078-0432.ccr-12-1407 Godfrey AC, Xu Z,Weinberg CR, Getts RC,Wade PA, LA DR et al, 2013, Serum microRNA expression as an early marker for breast cancer risk in prospectively collected samples from the Sister Study cohort. Breast Cancer Res 15(3):R42. , DOI 10.1186/ bcr3428 Heneghan HM,Miller N, Lowery AJ, Sweeney KJ, Newell J, Kerin MJ, 2010, Circulating microRNAs as novel minimally invasive biomarkers for breast cancer. Ann Surg 251(3):499–505. https:// doi.org/10.1097/SLA.0b013e3181cc939f Park IH, Kang JH, Lee KS, Nam S, Ro J, Kim JH, 2014, Identification and clinical implications of circulating microRNAs for estrogen receptor-positive breast cancer. Tumour Biol 35: 12173–12180. , DOI 10.1007/s13277-014-2525-5 Schrauder MG, Strick R, Schulz-Wendtland R, Strissel PL, Kahmann L, Loehberg CR, Lux MP, Jud SM, Hartmann A, Hein A, Bayer CM, Bani MR, Richter S, Adamietz BR,Wenkel E, Rauh C, Beckmann MW, Fasching PA, 2012, Circulating micro-RNAs as potential blood-based markers for early stage breast cancer detection. PLoS One 7(1):e29770. , DOI 10.1371/journal. pone.0029770 Heneghan HM, Miller N, Kelly R, Newell J, Kerin MJ, 2010, Systemic miRNA-195 differentiates breast cancer from other malignancies and is a potential biomarker for detecting noninvasive and early stage disease. Oncologist 15(7):673–682. , DOI 10.1634/theoncologist.2010-0103 Ouyang M, Li Y, Ye S, Ma J, Lu L, Lv W, Chang G, Li X, Li Q, Wang S,WangW(2014)MicroRNAprofiling implies newmarkers of chemoresistance of triple-negative breast cancer. PLoS One 9(5): e96228. , DOI 10.1371/journal.pone.0096228 Ng EK, Chong WW, Jin H, Lam EK, Shin VY, Yu J et al, 2009, Differential expression of microRNAs in plasma of patients with colorectal cancer: a potentialmarker for colorectal cancer screening. Gut. 58(10):1375–1381. , DOI 10.1136/gut.2008.167817 Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O'Briant KC, Allen A, Lin DW, Urban N, Drescher CW, Knudsen BS, Stirewalt DL, Gentleman R, Vessella RL, Nelson PS, Martin DB, Tewari M, 2008, Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci 105(30):10513– 10518. , DOI 10.1073/pnas.0804549105 Liu J, Mao Q, Liu Y, Hao X, Zhang S, Zhang J, 2013, Analysis of miR-205 and miR-155 expression in the blood of breast cancer patients. Chin J Cancer Res 25(1):46–54. , DOI 10.3978/ j.issn.1000-9604.2012.11.04 Calvano Filho CM, Calvano-Mendes DC, Carvalho KC, Maciel GA, Ricci MD, Torres AP et al, 2014, Triple-negative and luminal A breast tumors: differential expression ofmiR-18a-5p,miR-17-5p, and miR-20a-5p. Tumour Biol 35(8):7733–7741. , DOI 10.1007/s13277-014-2025-7 Anfossi S, Giordano A, Gao H, Cohen EN, Tin S,Wu Q, Garza RJ, Debeb BG, Alvarez RH, Valero V, Hortobagyi GN, Calin GA, Ueno NT, Woodward WA, Reuben JM, 2014, High serum miR- 19a levels are associated with inflammatory breast cancer and are predictive of favorable clinical outcome in patients with metastatic HER2+ inflammatory breast cancer. PLoS One 9(1):e83113. , DOI 10.1371/journal.pone.0083113 Mangolini A, Ferracin M, ZanziMV, Saccenti E, Ebnaof SO, Poma VV, Sanz JM, Passaro A, Pedriali M, Frassoldati A, Querzoli P, Sabbioni S, Carcoforo P, Hollingsworth A, Negrini M, 2015, Diagnostic and prognostic microRNAs in the serum of breast cancer patients measured by droplet digital PCR. Biomarker Res 3:12. , DOI 10.1186/s40364-015-0037-0 Zhang G, Liu Z, Cui G, Wang X, Yang Z, 2014, MicroRNA-486- 5p targeting PIM-1 suppresses cell proliferation in breast cancer cells. Tumour Biol 35(11):11137–11145. , DOI 10.1007/ s13277-014-2412-0 Lerebours F, Cizeron-Clairac G, Susini A, Vacher S, Mouret- Fourme E, Belichard C, et al. miRNA expression profiling of inflammatory breast cancer identifies a 5-miRNA signature predictive of breast tumor aggressiveness. Int J Cancer Journal international du cancer. 2013;133(7):1614–1623. , DOI 10.1002/ ijc.28171 O'Connell RM, Rao DS, Chaudhuri AA, BoldinMP, Taganov KD, Nicoll J, Paquette RL, Baltimore D, 2008, Sustained expression of microRNA-155 in hematopoietic stem cells causes a myeloproliferative disorder. J Exp Med 205(3):585–594. , DOI 10. 1084/jem.20072108 Kluiver J, Poppema S, de Jong D, Blokzijl T, Harms G, Jacobs S, Kroesen BJ, van den Berg A, 2005, BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and diffuse large B cell lymphomas. J Pathol 207(2):243–249. , DOI 10.1002/path. 1825 Volinia S, Calin GA, Liu C-G, Ambs S, Cimmino A, Petrocca F et al, 2006, A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 103(7):2257–2261 Wang X, Tang S, Le SY, Lu R, Rader JS, Meyers C et al, 2008, Aberrant expression of oncogenic and tumor-suppressive microRNAs in cervical cancer is required for cancer cell growth. PLoS One 3(7):e2557. , DOI 10.1371/journal.pone. 0002557 Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M, Stephens RM, Okamoto A, Yokota J, Tanaka T, Calin GA, Liu CG, CroceCM, Harris CC, 2006, Unique microRNAmolecular profiles in lung cancer diagnosis and prognosis. Cancer Cell 9(3):189–198. , DOI 10.1016/j.ccr.2006.01.025 Kong W, He L, Coppola M, Guo J, Esposito NN, Coppola D et al, 2016, MicroRNA-155 regulates cell survival, growth, and chemosensitivity by targeting FOXO3a in breast cancer. J Biol Chem 291(43):22855. , DOI 10.1074/jbc.A110.101055 Zhao FL, Dou YC, Wang XF, Han DC, Lv ZG, Ge SL et al, 2014, Serum microRNA-195 is down-regulated in breast cancer: a potential marker for the diagnosis of breast cancer. Mol Biol Rep 41(9): 5913–5922. , DOI 10.1007/s11033-014-3466-1 Li D, Zhao Y, Liu C, Chen X, Qi Y, Jiang Yet al, 2011, Analysis of MiR-195 and MiR-497 expression, regulation and role in breast cancer. Clin Cancer Res 17(7):1722–1730. , DOI 10. 1158/1078-0432.ccr-10-1800 Luo Q,Wei C, LiX, Li J, Chen L,HuangYet al, 2014, MicroRNA- 195-5p is a potential diagnostic and therapeutic target for breast cancer. Oncol Rep 31(3):1096–1102. , DOI 10.3892/or. 2014.2971 Hu Z, Dong J,Wang LE, Ma H, Liu J, Zhao Y, Tang J, Chen X, Dai J,Wei Q, Zhang C, Shen H, 2012, Serum microRNA profiling and breast cancer risk: the use of miR-484/191 as endogenous controls. Carcinogenesis. 33(4):828–834. , DOI 10.1093/carcin/ bgs030 Iorio MV, Visone R, Di Leva G, Donati V, Petrocca F, Casalini P et al, 2007, MicroRNA signatures in human ovarian cancer. Cancer Res 67(18):8699–8707. , DOI 10.1158/0008-5472.can-07- 1936 Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S et al, 2008, MicroRNA-21, miR-21, posttranscriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 27(15):2128–2136. , DOI 10.1038/sj.onc. 1210856 Chan JA, Krichevsky AM, Kosik KS, 2005, MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res 65(14):6029–6033. , DOI 10.1158/0008-5472.can-05- 0137 Stuckrath I, Rack B, JanniW, Jager B, Pantel K, Schwarzenbach H, 2015, Aberrant plasma levels of circulating miR-16, miR-107, miR-130a and miR-146a are associated with lymph node metastasis and receptor status of breast cancer patients. Oncotarget. 6(15): 13387–13401. , DOI 10.18632/oncotarget.3874 Shin VY, Siu JM, Cheuk I, Ng EK, Kwong A, 2015, Circulating cell-free miRNAs as biomarker for triple-negative breast cancer. Br J Cancer 112(11):1751–1759. , DOI 10.1038/bjc.2015.143 Guo LJ, Zhang QY, 2012, Decreased serum miR-181a is a potential new tool for breast cancer screening. Int J Mol Med 30(3):680– 686. , DOI 10.3892/ijmm.2012.1021 McDermott AM, KerinMJ, Miller N, 2013, Identification and validation of miRNAs as endogenous controls for RQ-PCR in blood specimens for breast cancer studies. PLoS One 8(12):e83718 Lu J, ClarkAG(2012, Impact of microRNAregulation on variation in human gene expression. Genome Res 22(7):1243–1254. https:// doi.org/10.1101/gr.132514.111 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 833 EP 844 DI 10.1007/s12253-019-00627-y PG 12 ER PT J AU Soria-Comes, T Palomar-Abril, V Ureste, MM Guerola, TM Maiques, MIC AF Soria-Comes, Teresa Palomar-Abril, Vicente Ureste, Martin Maria Guerola, Tallon Monica Maiques, Maestu Inmaculada Concepcion TI Real-World Data of the Correlation between EGFR Determination by Liquid Biopsy in Non-squamous Non-small Cell Lung Cancer (NSCLC) and the EGFR Profile in Tumor Biopsy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR mutation status; Liquid biopsy; ctDNA; Cobas EGFR assay; Non-small cell lung cancer (NSCLC) ID EGFR mutation status; Liquid biopsy; ctDNA; Cobas EGFR assay; Non-small cell lung cancer (NSCLC) AB EGFR-mutated non-small cell lung cancer (NSCLC) has significant improved outcomes when treated with EGFR-tyrosine kinase inhibitors (TKI). Thus, EGFR-mutational status should be assessed at diagnosis and in the course of treatment with TKI. However, tissue samples are not always evaluable, and molecular profiling has been increasingly performed in cell-free tumor DNA (ctDNA) from blood samples. Our objective is to evaluate the reliability of ctDNA profiling in plasma samples in a real-world setting. We retrospectively analyzed the patients diagnosed with non-squamous NSCLC from May 2016 to December 2017 at Hospital Universitario Doctor Peset who had been tested for EGFR mutations in tissue and plasma samples. Both samples were sent to an external laboratory to perform the analysis by the cobas® EGFR assay. Percentage of agreement and concordance were calculated by kappa statistic. Of 102 patients reviewed, 89 were eligible. The overall EGFR mutation frequency was 18.6% for the evaluable tissue samples and 19.6% for evaluable plasma samples. Mutation status concordance between matched samples was 87.4%. Cohen’s kappa index (κ) = 0.6 (sensitivity 70.6%, specificity 91.7%, positive predictive value 66.7%, negative predictive value 93%). When concordance was stablished only in stage IV tumors κ = 0.7, suggesting a higher agreement in advanced disease. This real-world data suggest that plasma is a feasible sample for ctDNA EGFR mutation assessment. Results of ctDNA molecular profiling are reliable when using a validated technique such as the cobas® EGFR assay, especially in patients that cannot undergo a tissue biopsy. C1 [Soria-Comes, Teresa] Hospital Universitario Doctor Peset, Department of Medical Oncology, 46017 Valencia, Comunitat Valenciana, Spain. [Palomar-Abril, Vicente] Hospital Universitario Doctor Peset, Department of Medical Oncology, 46017 Valencia, Comunitat Valenciana, Spain. [Ureste, Martin Maria] Hospital Universitario Doctor Peset, Department of Medical Oncology, 46017 Valencia, Comunitat Valenciana, Spain. [Guerola, Tallon Monica] Hospital Universitario Doctor Peset, Department of Medical OncologyValencia, Comunitat Valenciana, Spain. [Maiques, Maestu Inmaculada Concepcion] Hospital Universitario Doctor Peset, Department of Medical Oncology, 46017 Valencia, Comunitat Valenciana, Spain. RP Soria-Comes, T (reprint author), Hospital Universitario Doctor Peset, Department of Medical Oncology, 46017 Valencia, Spain. EM t.soria.comes@gmail.com CR Cancer Incidence IARC, 2012, Mortality and prevalence worldwide. In: GLOBOCAN Malvezzi M, Carioli G, Bertuccio P, Boffetta P, Levi F, La Vecchia C et al, 2017, European cancer mortality predictions for the year 2017, with focus on lung cancer. Ann Oncol 28:1117–1123. https:// doi.org/10.1093/annonc/mdx033 Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, Majem M, Lopez-Vivanco G, Isla D, Provencio M, Insa A, Massuti B, Gonzalez-Larriba JL, Paz-Ares L, Bover I, Garcia- Campelo R, Moreno MA, Catot S, Rolfo C, Reguart N, Palmero R, Sanchez JM, Bastus R, Mayo C, Bertran-Alamillo J, Molina MA, Sanchez JJ, Taron M, 2009, Screening for epidermal growth factor receptor mutations in lung Cancer. N Engl J Med 361:958– 967. , DOI 10.1056/NEJMoa0904554 Janku F, Angenendt P, Tsimberidou AM, Fu S, Naing A, Falchook GS, Hong DS, Holley VR, Cabrilo G, Wheler JJ, Piha-Paul SA, Zinner RG, Bedikian AY, Overman MJ, Kee BK, Kim KB, Kopetz SE, Luthra R, Diehl F, Meric- Bernstam F, Kurzrock R, 2015, Actionable mutations in plasma cell-free DNA in patients with advanced cancers referred for experimental targeted therapies. Oncotarget 6: 12809–12821. , DOI 10.18632/oncotarget.3373 Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, Antonarakis ES, Azad NS, Bardelli A, Brem H, Cameron JL, Lee CC, Fecher LA, Gallia GL, Gibbs P, le D, Giuntoli RL, Goggins M, Hogarty MD, Holdhoff M, Hong SM, Jiao Y, Juhl HH, Kim JJ, Siravegna G, Laheru DA, Lauricella C, Lim M, Lipson EJ, Marie SKN, Netto GJ, Oliner KS, Olivi A, Olsson L, Riggins GJ, Sartore-Bianchi A, Schmidt K, Shih M, Oba-Shinjo SM, Siena S, Theodorescu D, Tie J, Harkins TT, Veronese S, Wang TL, Weingart JD, Wolfgang CL, Wood LD, Xing D, Hruban RH, Wu J, Allen PJ, Schmidt CM, Choti MA, Velculescu VE, Kinzler KW, Vogelstein B, Papadopoulos N, Diaz LA, 2014, Detection of circulating tumor DNA in early and late stage human malignancies. Sci Transl Med 6:224ra24. , DOI 10.1126/scitranslmed.3007094 Goldstraw P, Chansky K, Crowley J, Rami-Porta R, Asamura H, EberhardtWEE,NicholsonAG, Groome P,MitchellA, Bolejack V, Goldstraw P, Rami-Porta R, Asamura H, Ball D, Beer DG, Beyruti R, Bolejack V, Chansky K, Crowley J, Detterbeck F, Erich Eberhardt WE, Edwards J, Galateau-Salle F, Giroux D, Gleeson F, Groome P, Huang J, Kennedy C, Kim J, Kim YT, Kingsbury L, Kondo H, Krasnik M, Kubota K, Lerut A, Lyons G, Marino M, Marom EM, van Meerbeeck J, Mitchell A, Nakano T, Nicholson AG, Nowak A, Peake M, Rice T, Rosenzweig K, Ruffini E, Rusch V, Saijo N, van Schil P, Sculier JP, Shemanski L, Stratton K, Suzuki K, Tachimori Y, Thomas CF Jr, Travis W, Tsao MS, Turrisi A, Vansteenkiste J, Watanabe H, Wu YL, Baas P, Erasmus J, Hasegawa S, Inai K, Kernstine K, Kindler H, Krug L, Nackaerts K, Pass H, Rice D, Falkson C, Filosso PL, Giaccone G, Kondo K, Lucchi M, Okumura M, Blackstone E, Abad Cavaco F, Ansotegui Barrera E, Abal Arca J, Parente Lamelas I, Arnau Obrer A, Guijarro Jorge R, Ball D, Bascom GK, Blanco Orozco AI, Gonzalez Castro MA, Blum MG, Chimondeguy D, Cvijanovic V, Defranchi S, de Olaiz Navarro B, Escobar Campuzano I, Macia Vidueira I, Fernandez Araujo E, Andreo Garcia F, Fong KM, Francisco Corral G, Cerezo Gonzalez S, Freixinet Gilart J, Garcia Aranguena L, Garcia Barajas S, Girard P, Goksel T, Gonzalez Budino MT, Gonzalez Casaurran G, Gullon Blanco JA, Hernandez Hernandez J, Hernandez Rodriguez H, Herrero Collantes J, Iglesias Heras M, Izquierdo Elena JM, Jakobsen E, Kostas S, Leon Atance P, Nunez Ares A, Liao M, Losanovscky M, Lyons G, Magaroles R, de Esteban Julvez L, Marinan Gorospe M, McCaughan B, Kennedy C, Melchor Íniguez R, Miravet Sorribes L, Naranjo Gozalo S, Alvarez de Arriba C, Nunez Delgado M, Padilla Alarcon J, Penalver Cuesta JC, Park JS, Pass H, Pavon Fernandez MJ, Rosenberg M, Ruffini E, Rusch V, Sanchez de Cos Escuin J, Saura Vinuesa A, Serra Mitjans M, Strand TE, Subotic D, Swisher S, Terra R, Thomas C, Tournoy K, van Schil P, Velasquez M, Wu YL, Yokoi K, 2016, The IASLC lung cancer staging project: proposals for revision of the TNM stage groupings in the forthcoming, eighth, edition of the TNM classification for lung cancer. J Thorac Oncol 11:39–51. https:// doi.org/10.1016/j.jtho.2015.09.009 Viera AJ, Garrett JM, 2005, Understanding interobserver agreement: the kappa statistic. Fam Med 37:360–363 Planchard D, Popat S, Kerr K, Novello S, Smit E, Faivre-Finn C et al, 2018, Metastatic non-small-cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up†. Ann Oncol 27:v192–v237. , DOI 10.1093/annonc/mdw326. Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, Palmero R, Garcia-Gomez R, Pallares C, Sanchez JM, Porta R, Cobo M, Garrido P, Longo F, Moran T, Insa A, deMarinis F, Corre R, Bover I, Illiano A, Dansin E, de Castro J, Milella M, Reguart N, Altavilla G, Jimenez U, Provencio M, Moreno MA, Terrasa J, Munoz-Langa J, Valdivia J, Isla D, Domine M, Molinier O, Mazieres J, Baize N, Garcia-Campelo R, Robinet G, Rodriguez- Abreu D, Lopez-Vivanco G, Gebbia V, Ferrera-Delgado L, Bombaron P, Bernabe R, Bearz A, Artal A, Cortesi E, Rolfo C, Sanchez-Ronco M, Drozdowskyj A, Queralt C, de Aguirre I, Ramirez JL, Sanchez JJ, Molina MA, Taron M, Paz-Ares L, 2012, Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutationpositive non-small-cell lung cancer, EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 13:239–246. , DOI 10.1016/S1470-2045(11)70393-X Sequist LV, Yang JC-H, Yamamoto N, O’Byrne K, Hirsh V,Mok T et al, 2013, Phase III study of Afatinib or cisplatin plus Pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 31:3327–3334. , DOI 10.1200/JCO. 2012.44.2806 Oxnard GR, Arcila ME, Sima CS, Riely GJ, Chmielecki J, Kris MG, Pao W, Ladanyi M, Miller VA, 2011, Acquired resistance to EGFR tyrosine kinase inhibitors in EGFR-mutant lung cancer: distinct natural history of patients with tumors harboring the T790M mutation. Clin Cancer Res 17:1616–1622. , DOI 10.1158/ 1078-0432.CCR-10-2692 Goss G, Tsai CM, Shepherd FA, Bazhenova L, Lee JS, Chang GC, Crino L, Satouchi M, Chu Q, Hida T, Han JY, Juan O, Dunphy F, Nishio M, Kang JH, MajemM,Mann H, CantariniM, Ghiorghiu S, Mitsudomi T, 2016, Osimertinib for pretreated EGFR Thr790Metpositive advanced non-small-cell lung cancer, AURA2): a multicentre, open-label, single-arm, phase 2 study. Lancet Oncol 17:1643–1652. , DOI 10.1016/S1470-2045(16)30508-3 Rolfo C, Mack PC, Scagliotti GV, Baas P, Barlesi F, Bivona TG, Herbst RS, Mok TS, Peled N, Pirker R, Raez LE, Reck M, Riess JW, Sequist LV, Shepherd FA, Sholl LM, Tan DSW, Wakelee HA, Wistuba II, Wynes MW, Carbone DP, Hirsch FR, Gandara DR, 2018, Liquid biopsy for advanced non-small cell lung Cancer, NSCLC): a statement paper from the IASLC. J Thorac Oncol 13: 1248–1268. , DOI 10.1016/j.jtho.2018.05.030 Malapelle U, Sirera R, Jantus-Lewintre E, Reclusa P, Calabuig- Farinas S, Blasco A, Pisapia P, Rolfo C, Camps C, 2017, Profile of the Roche cobas®EGFR mutation test v2 for non-small cell lung cancer. Expert Rev Mol Diagn 17:209–215. , DOI 10. 1080/14737159.2017.1288568 Reckamp KL, Melnikova VO, Karlovich C, Sequist LV, Camidge DR, Wakelee H, Perol M, Oxnard GR, Kosco K, Croucher P, Samuelsz E, Vibat CR, Guerrero S, Geis J, Berz D, Mann E, Matheny S, Rolfe L, Raponi M, Erlander MG, Gadgeel S, 2016, A highly sensitive and quantitative test platform for detection of NSCLC EGFR mutations in urine and plasma. J Thorac Oncol 11:1690–1700. , DOI 10.1016/j.jtho.2016.05.035 Douillard JY, Ostoros G, Cobo M, Ciuleanu T, Cole R, McWalter G, Walker J, Dearden S, Webster A, Milenkova T, McCormack R, 2014, Gefitinib treatment in EGFR mutated caucasian NSCLC: circulating-free tumor DNA as a surrogate for determination of EGFR status. J Thorac Oncol 9:1345–1353. , DOI 10. 1097/JTO.0000000000000263 Soria J-C, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH et al, 2018, Osimertinib in untreated EGFR -mutated advanced non–small-cell lung Cancer. N Engl J Med 2017:NEJMoa1713137. , DOI 10.1056/ NEJMoa1713137 Han B, Tjulandin S, Hagiwara K, Normanno N, Wulandari L, Laktionov K, Hudoyo A, He Y, Zhang YP, Wang MZ, Liu CY, Ratcliffe M, McCormack R, Reck M, 2017, EGFR mutation prevalence in Asia-Pacific and Russian patients with advanced NSCLC of adenocarcinoma and non-adenocarcinoma histology: the IGNITE study. Lung Cancer 113:37–44. , DOI 10.1016/j. lungcan.2017.08.021 Wu Y-L, Sequist LV, Hu C-P, Feng J, Lu S, Huang Y, LiW, Hou M, Schuler M, Mok T, Yamamoto N, O'Byrne K, Hirsh V, Gibson N, Massey D, Kim M, Yang JCH, 2016, EGFR mutation detection in circulating cell-free DNA of lung adenocarcinoma patients: analysis of LUX-lung 3 and 6. Br J Cancer 116:175–185. , DOI 10.1038/bjc.2016.420 Reck M, Hagiwara K, Han B, Tjulandin S, Grohe C, Yokoi T, Morabito A, Novello S, Arriola E, Molinier O, McCormack R, Ratcliffe M, Normanno N, 2016, CtDNA determination of EGFR mutation status in European and Japanese patients with advanced NSCLC: the ASSESS study. J Thorac Oncol 11:1682–1689. https:// doi.org/10.1016/j.jtho.2016.05.036 Esteban E, Majem M, Martinez Aguillo M, Martinez Banaclocha N, Domine M, Gomez Aldaravi L, Juan O, Cajal R, Gonzalez Arenas MC, Provencio M, 2015, Prevalence of EGFR mutations in newly diagnosed locally advanced or metastatic non-small cell lung cancer Spanish patients and its association with histological subtypes and clinical features: the Spanish REASON study. Cancer Epidemiol 39:291–297. , DOI 10.1016/j.canep.2015.02.003 Normanno N, Denis MG, Thress KS, Ratcliffe M, Reck M, 2017, Guide to detecting epidermal growth factor receptor, EGFR, mutations in ctDNA of patients with advanced non-small-cell lung cancer. Oncotarget 8:12501–12516. , DOI 10.18632/ oncotarget.13915 Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A, 2013, Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 10:472–484. , DOI 10.1038/nrclinonc.2013.110 Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Liquid biopsies: genotyping circulating tumor DNA. Nat Med 2014;4:224ra24-224ra24. , DOI 10.1158/2159-8290.CD- 13-1014.Blood. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 845 EP 851 DI 10.1007/s12253-019-00628-x PG 7 ER PT J AU Zhao, R Li, L Yang, J Niu, Q Wang, H Qin, X Zhu, N Shi, A AF Zhao, Rui Li, Lei Yang, Jinbo Niu, Qinfeng Wang, Han Qin, Xiaodong Zhu, Ning Shi, Anchen TI Overexpression of Pyruvate Kinase M2 in Tumor Tissues Is Associated with Poor Prognosis in Patients with Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pyruvate kinase M2; Hepatocellular carcinoma; Prognosis; Overall survival rate; Reoccurrence ID Pyruvate kinase M2; Hepatocellular carcinoma; Prognosis; Overall survival rate; Reoccurrence AB Hepatocellular carcinoma (HCC) is one of the most common malignant tumors, with a high degree of malignancy and a poor prognosis. The aim of this study was to investigate the relationship between expression of pyruvate kinase M2 (PKM2) and prognosis in patients with HCC. The expression levels of PKM2 and PKM1 in 86 cases of HCC were detected by immunohistochemistry. An H score was used to evaluate the expression of PKM, and all patients were further divided into PKM high expression and PKM low-expression groups. The relationship between PKM2 expression and the clinicopathological parameters and prognosis of patients were subsequently analyzed. Our data suggested that the expression level of PKM2 was significantly higher in HCC tissues than in adjacent tissues and the negatively expression of PKM1 in HCC tissues. Kaplan-Meier analysis revealed that PKM2 expression was strongly associated with survival in HCC patients (P = 0.001). The patients in the PKM2 high-expression group had significantly shorter survival times than the patients in the PKM2 low-expression group (hazard ratio for death, 2.358; 95% confidence interval [1.156, 4.812]; P = 0.018). In conclusion, these data indicate that PKM2 expression in HCC tissue samples can be used as a prognostic factor for patients with HCC and that high PKM2 expression is correlated with a poor prognosis in HCC patients. C1 [Zhao, Rui] Lanzhou University, School of Life Sciences, 730000 Lanzhou, China. [Li, Lei] Lanzhou University First Hospital, Department of Interventional Radiology, 730030 Lanzhou, China. [Yang, Jinbo] Lanzhou University, School of Life Sciences, 730000 Lanzhou, China. [Niu, Qinfeng] Baoji Affiliated Hospi, Xi’an Medical College, Baoguang Branch, 721006 Baoji, China. [Wang, Han] Lanzhou University, School of Life Sciences, 730000 Lanzhou, China. [Qin, Xiaodong] Lanzhou University, School of Life Sciences, 730000 Lanzhou, China. [Zhu, Ning] Lanzhou University, School of Life Sciences, 730000 Lanzhou, China. [Shi, Anchen] Lanzhou University, School of Basic Medical Sciences, 730000 Lanzhou, China. RP Yang, J (reprint author), Lanzhou University, School of Life Sciences, 730000 Lanzhou, China. EM zhaor@lzu.edu.cn CR Altekruse SF, McGlynn KA, Dickie LA, Kleiner DE, 2012, Hepatocellular carcinoma confirmation, treatment, and survival in surveillance, epidemiology, and end results registries, 1992-2008. Hepatology 55:476–482. , DOI 10.1002/hep.24710 Li H, Huang W, Luo R, 2015, The microRNA-325 inhibits hepatocellular carcinoma progression by targeting high mobility group box 1. Diagn Pathol 10:117. , DOI 10.1186/s13000-015- 23-z 3. ZengQA,Qiu J, Zou R, LiY, Li S, Li B, Huang P, Hong J, ZhengY, Lao X, Yuan Y, 2012, Clinical features and outcome of multiple primary malignancies involving hepatocellular carcinoma: a longterm follow-up study. BMC Cancer 12:148. , DOI 10. 1186/1471-2407-12-148 Chen CJ, Yang HI, Su J, Jen CL, You SL, Lu SN, Huang GT, Iloeje UH, REVEAL-HBV Study Group, 2006, Risk of hepatocellular carcinoma across a biological gradient of serum hepatitis B virus DNA level. Jama 295:65–73. , DOI 10. 1001/jama.295.1.65 Bosch FX, Ribes J, Diaz M, Cleries R, 2004, Primary liver cancer: worldwide incidence and trends. Gastroenterology 127:S5–S16 Ang SF, Ng ES, Li H, Ong YH, Choo SP, Ngeow J, Toh HC, Lim KH, Yap HY, Tan CK, Ooi LL, Cheow PC, Chung AY, Chow PK, Foo KF, Tan MH, 2015, The Singapore liver cancer recurrence, SLICER, score for relapse prediction in patients with surgically resected hepatocellular carcinoma. PLoS ONE 10:e0118658. , DOI 10.1371/journal.pone.0128058 Llovet JM, Burroughs A, Bruix J, 2003, Hepatocellular carcinoma. Lancet 362:1907–1917. , DOI 10.1016/S0140-6736(03, 14964-1 Yu G, YuW, Jin G, Xu D, Chen Y, Xia T, Yu A, FangW, Zhang X, Li Z, Xie K, 2015, PKM2 regulates neural invasion of and predicts poor prognosis for human hilar cholangiocarcinoma. Mol Cancer 14:193. , DOI 10.1186/s12943-015-0462-6 Yamada S, Fuchs BC, Fujii T, Shimoyama Y, Sugimoto H, Nomoto S, Takeda S, Tanabe KK, Kodera Y, Nakao A, 2013, Epithelial-tomesenchymal transition predicts prognosis of pancreatic cancer. Surgery 154:946–954. , DOI 10.1016/j.surg.2013.05.004 MacDonald MJ, ChangCM(1985, Pancreatic islets contain theM2 isoenzyme of pyruvate kinase. Its phosphorylation has no effect on enzyme activity. Mol Cell Biochem 68:115–120 Dombrauckas JD, Santarsiero BD, Mesecar AD, 2005, Structural basis for tumor pyruvate kinase M2 allosteric regulation and catalysis. Biochemistry 44:9417–9429. , DOI 10.1021/ bi0474923 Vaupel P, Harrison L, 2004, Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist 9: 4–9. , DOI 10.1634/theoncologist.9-90005-4 Wong N, De Melo J, Tang D, 2013, PKM2, a central point of regulation in cancer metabolism. Int J Cell Biol 2013:242513:1– 11. , DOI 10.1155/2013/242513 Noguchi T, Inoue H, Tanaka T, 1986, The M1- and M2-type isozymes of rat pyruvate kinase are produced from the same gene by alternative RNA splicing. J Biol Chem 261:13807–13812 Eigenbrodt E, Reinacher M, Scheefers-Borchel U, Scheefers H, Friis R, 1992, Double role for pyruvate kinase type M2 in the expansion of phosphometabolite pools found in tumor cells. Crit Rev Oncog 3:91–115 Mazurek S, Boschek CB, Hugo F, Eigenbrodt E, 2005, Pyruvate kinase type M2 and its role in tumor growth and spreading. Semin Cancer Biol 15:300–308. , DOI 10.1016/j.semcancer. 2005.04.009 Steinberg P, Klingelhoffer A, Schafer A,Wust G,Weisse G, Oesch F, Eigenbrodt E, 1999, Expression of pyruvate kinase M2 in preneoplastic hepatic foci of N-nitrosomorpholine-treated rats. Virchows Arch 434:213–220. , DOI 10.1007/ s004280050330 David CJ, Chen M, Assanah M, Canoll P, Manley JL, 2010, HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature 63:364–368. , DOI 10. 1038/nature08697 Dong G,Mao Q, XiaW, XuY,Wang J,Xu L, Jiang F, 2016, PKM2 and cancer: the function of PKM2 beyond glycolysis. Oncol Lett 11:1980–1986. , DOI 10.1038/nature08697 Li Z, Yang P, Li Z, 2014, Themultifaceted regulation and functions of PKM2 in tumor progression. Biochim Biophys Acta 1846:285– 296. , DOI 10.1016/j.bbcan.2014.07.008 Christofk HR, Vander Heiden MG, Harris MH, Ramanathan A, Gerszten RE, Wei R, Fleming MD, Schreiber SL, Cantley LC, 2008, The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 452:230–233. , DOI 10.1038/nature06734 Tamada M, Suematsu M, Saya H, 2012, Pyruvate kinase M2: multiple faces for conferring benefits on cancer cells. Clin Cancer Res 18:5554–5561. , DOI 10.1158/1078-0432.CCR-12-0859 Schulze G, 2000, The tumor marker tumor M2-PK: an application in the diagnosis of gastrointestinal cancer. Anticancer Res 20:4961–4964 Wong N, Yan J, Ojo D, De Melo J, Cutz JC, Tang D, 2014, Changes in PKM2 associate with prostate cancer progression. Cancer Investig 32:330–338. , DOI 10.3109/07357907. 2014.919306 Hardt PD, Ngoumou BK, Rupp J, Schnell-Kretschmer H, Kloer HU, 2000, Tumor M2-pyruvate kinase: a promising tumor marker in the diagnosis of gastro-intestinal cancer. Anticancer Res 20: 4965–4968 Fan F, Wu H, Liu Z, Hou X, Chen W, Wang A, Lu Y, 2016, Nuclear PKM2 expression, an independent risk factor for ER after curative resection of hepatocellular carcinoma. Biomed Pharmacother 84:1858–1864. , DOI 10.1016/j.biopha. 2016.10.108 Chen Z, Lu X, Wang Z, Jin G, Wang Q, Chen D, Chen T, Li J, Fan J, Cong W, Gao Q, He X, 2015, Co-expression of PKM2 and TRIM35 predicts survival and recurrence in hepatocellular carcinoma. Oncotarget 6:2538–2548. , DOI 10.18632/ oncotarget.2991 Xu Q, Liu X, Zheng X, Yao Y, Liu Q, 2014, PKM2 regulates Gli1 expression in hepatocellular carcinoma. Oncol Lett 8:1973–1979. , DOI 10.3892/ol.2014.2441 Fan FT, Shen CS, Tao L, Tian C, Liu ZG, Zhu ZJ, Liu YP, Pei CS, Wu HY, Zhang L, Wang AY, Zheng SZ, Huang SL, Lu Y, 2014, PKM2 regulates hepatocellular carcinoma cell epithelialmesenchymal transition and migration upon EGFR activation. Asian Pac J Cancer Prev 15:1961–1970 Liu WR, Tian MX, Yang LX, Lin YL, Jin L, Ding ZB, Shen YH, Peng YF, Gao DM, Zhou J, Qiu SJ, Dai Z, He R, Fan J, Shi YH, 2015, PKM2 promotes metastasis by recruiting myeloid-derived suppressor cells and indicates poor prognosis for hepatocellular carcinoma. Oncotarget 6:846–861. , DOI 10.18632/ oncotarget.2749 LiuY,WuH,MeiY, DingX, YangX,Li C,DengM, GongJ, 2017, Clinicopathological and prognostic significance of PKM2 protein expression in cirrhotic hepatocellular carcinoma and non-cirrhotic hepatocellular carcinoma. Sci Rep 7:15294. , DOI 10. 1038/s41598-017-14813-y NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 853 EP 860 DI 10.1007/s12253-019-00630-3 PG 8 ER PT J AU Fernandez-Acenero, JM Cruz, M Sastre-Varela, J Casal, IJ Ceron Nieto, M del Puerto-Nevado, L Garcia-Foncillas, J Cebrian, A AF Fernandez-Acenero, Jesus Maria Cruz, Mari Sastre-Varela, Javier Casal, Ignacio Jose Ceron Nieto, Angeles Maria del Puerto-Nevado, Laura Garcia-Foncillas, Jesus Cebrian, Arancha TI TRIM72 Immunohistochemical Expression Can Predict Relapse in Colorectal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon carcinoma; Chemotherapy; Early stage disease; Ubiquitin ligase; TRIM72; Recurrence; Immunohistochemistry ID Colon carcinoma; Chemotherapy; Early stage disease; Ubiquitin ligase; TRIM72; Recurrence; Immunohistochemistry AB Large bowel adenocarcinoma is one of the most frequent human neoplasms and despite recent insights into the pathophysiology and molecular basis of this disease, mortality remains high in advanced and metastatic cases. Most guidelines recommend adjuvant chemotherapy for tumours involving lymph nodes, but not for patients with localized stage I or II disease. However, it is well known that approximately 20% of stage II colorectal carcinoma patients eventually recur, mainly with distant or peritoneal involvement and show bad prognosis. It would be important to predict which patients are at increased risk of recurrence to guide potential adjuvant therapy use in this controversial setting. In this sense, only microsatellite stability has been proposed as a predictive tool in some guidelines. The tripartite motif family protein 72 (TRIM72) is a ubiquitin ligase, involved in the cell membrane repair machinery and known to be associated to insulin resistance. Its potential role in colon cancer has recently been proposed. The aim of this study is to determine the potential predictive value of TRIM72 immunohistochemical expression in stage II colon carcinoma. We have retrospectively reviewed a series of 95 patients with stage II colon microsatellite stable carcinomas operated with a curative intent at a single large tertiary hospital in Madrid (Spain) between 2006 and 2012. None of the patients received adjuvant chemotherapy. We reviewed the histopathological slides and constructed a tissue microarray (TMA) of three representative areas to perform immunohistochemical staining for TRIM72. In our series 30 patients (31.7%) recurred after a median follow-up of 17.5 months. Lack of immunohistochemical expression of TRIM72 in the tumor was significantly and independently associated to recurrence. A recent report by Chen et al. has shown that TRIM72 can be measured in plasma for colon carcinoma detection as an alternative to CEA or CA19.9, with lower levels in patients with carcinoma. Our report is the first one to show that lower immunohistochemical expression of TRIM72 predicts recurrence in colon stage II carcinoma. We feel this predictive influence can be related to its crucial role as a central regulator in many signaling pathways (PI3K-AKT, ERK).As an ubiquitin ligase, the lack of TRIM72 could increase the levels of several potential oncogenic molecules and therefore lead to a more aggressive phenotype. It remains to be shown whether chemotherapy could change the clinical behaviour of this bad prognosis group. We propose TRIM72 immunohistochemical analysis as a potential tool to predict recurrence risk in stage II colon carcinoma patients. Our results should be confirmed in larger series, but could open the way to management strategies refinement in this early stage group of patients. C1 [Fernandez-Acenero, Jesus Maria] Hospital Clinico San Carlos, Department of Surgical Pathology, Avda Profesor Martin Lagos s, /n, 28040 Madrid, Spain. [Cruz, Mari] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology DivisionMadrid, Spain. [Sastre-Varela, Javier] Hospital Clinico San Carlos, Departments of OncologyMadrid, Spain. [Casal, Ignacio Jose] Centro de Investigaciones Biologicas (CIB)Madrid, Spain. [Ceron Nieto, Angeles Maria] Hospital Clinico San Carlos, Department of Surgical Pathology, Avda Profesor Martin Lagos s, /n, 28040 Madrid, Spain. [del Puerto-Nevado, Laura] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology DivisionMadrid, Spain. [Garcia-Foncillas, Jesus] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology DivisionMadrid, Spain. [Cebrian, Arancha] University Hospital Fundacion Jimenez Diaz, Oncohealth Institute, Health Research Institute FJD-UAM, Translational Oncology DivisionMadrid, Spain. RP Fernandez-Acenero, JM (reprint author), Hospital Clinico San Carlos, Department of Surgical Pathology, 28040 Madrid, Spain. EM mgg10167@gmail.com CR Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics 2016. CA Cancer J Clin 66:7–30 Shui L, Wu YS, Lin H, Shui P, Sun Q, Chen X, 2018, Triplet chemotherapy, FOLFOXIRI, plus bevacizumab versus doublet chemotherapy, FOLFOX/FOLFIRI, plus bevacizumab in conversion therapy for metastatic colorectal Cancer: a meta-analysis. Cell Physiol Biochem 48:1870–1881 Osterman E, Glimelius B, 2018, Recurrence risk after up-to-date Colon Cancer staging, surgery, and pathology: analysis of the entire Swedish population. Dis Colon rectum 61:1016–1025 Bockelman C, Engelmann BE, Kaprio T, Hansen TF, Glimelius B, 2015, Risk of recurrence in patients with colon cancer stage II and III: a systematic review and meta-analysis of recent literature. Acta Oncol 54:5–16 Maeda C, Hidaka E, Mori Y, Mukai S, Miyachi H, Sawada N, 2015, Tumor diameter is an easy and useful predictor of recurrence in stage II colorectal Cancer. Dig Surg 32:338–343 Lemekert FA, Bournazos A, Eckert DM, Kenzler M, Hawkes JM, Butler TL et al, 2016, Lack of MG53 in human heart precludes utility as a biomarker of myocardial injury or endogenous cardioprotective factor. Cardiovasc Res 110:178–187 Liu L,Wong CC, Gong B, Yu J, 2017, Functional significance and therapeutic implication of ring-type E3 ligases in colorectal cancer. Oncogene 37:148–159 Chen Z, Yin X, Li K, Chen S, Li H, Li Y, Zhang Q,Wang H, Qiu Y, 2018, Serum levels of TRIM72 are lower among patients with colon cancer: identification of a potential diagnostic marker. Tokohu J Exp Med 245:61–68 Wille-Jorgensen P, Syk I, Smedh K, Laurberg S, Nielsen DT, Petersen SH, Renehan AG, Horvath-Puho E, Pahlman L, Sorensen HT, For the COLOFOL Study Group, 2018, Effect of more vs less frequent follow-up testing on overall and colorectal Cancer-specific mortality in patients with stage II or III colorectal Cancer: the COLOFOL randomized clinical trial. JAMA 319: 2095–2103 Hu X, Li YQ, Li QG, Ma YL, Peng JJ, Cai S, 2018, Mucinous adenocarcinomas Histotype can also be a high-risk factor for stage II colorectal Cancer patients. Cell Physiol Biochem 47:630–640 Lee VWK, Chan KF, 2018, Tumor budding and poorlydifferentiated cluster in prognostication in stage II colon cancer. Pathol Res Pract 214:402–407 Yamadera M, Shinto E, Tsuda H, Kajiwara Y, Naito Y, Hase K, Yamamoto J, Ueno H, 2018, Sialyl Lewisx expression at the invasive front as a predictive marker of liver recurrence in stage IIcolorectal cancer. Oncol Lett 15:221–228 Li C, Cai S, Wang X, Jiang Z, 2015, Identification and characterization of ANO9 in stage II and III colorectal carcinoma. Oncotarget 6:29324–29334 Li C, Cai S, Wang X, Jiang Z, 2014, Hypomethylation-associated up-regulation of TCF3 expression and recurrence in stage II and III colorectal cancer. PLoS One 9(11):e112005 NCCN Guidelines Version 3.2018 Colon Cancer. Copyright National Comprehensive Cancer Network Inc. Last accessed online 9/20/2018 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 861 EP 865 DI 10.1007/s12253-019-00629-w PG 5 ER PT J AU Collak, KF Ummuhan, D Sagir, F AF Collak, Kisaayak Filiz Ummuhan, Demir Sagir, Fatma TI YAP1 Is Involved in Tumorigenic Properties of Prostate Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE YAP1; pYAP1; Prostate cancer; Androgen receptor ID YAP1; pYAP1; Prostate cancer; Androgen receptor AB The Yes Associated Protein 1 (YAP1) is a transcriptional cofactor negatively regulated by Hippo Pathway. The dysregulation of the pathway has been shown to have a role in tumorigenesis and metastasis in several cancers including prostate cancer (PCa). In this study, YAP1 expression was upregulated in the whole cell lysates and cytoplasmic/nuclear extracts of AR negative (PC3) compared to AR positive (LNCaP) prostate cancer cell lines and primary prostate epithelial cells (PrePEC). pYAP1 expression elevated in LNCaP compared to PC3 and PrePEC in whole cell lysates and cytoplasmic extracts. The treatment of LNCaP and PC3 with YAP1-targeting siRNA oligonucleotides (YAP1 siRNA) significantly reduced their proliferation in vitro. Furthermore, treatment with YAP1 siRNA diminished the clonogenicity, anchorage-independent growth on soft agar, migration and invasion of PC3 cells. Co-IP/WB experiments revealed that YAP1 and AR formed a complex and ChIP/PCR results confirmed that YAP1 was bound to androgen response elements (ARE) core region of the prostate specific antigen (PSA) promoter. The loss of function experiments in LNCaP and PC3 revealed that YAP1 regulates proliferation, colony formation as well as anchorage-independent growth and potentially plays an important role in migration and invasion. Finally, analysis of publicly available data sets indicated that LNCaP had no YAP1 copy number alteration whereas PC3 had gain of YAP1 which was also reflected as an increase in the mRNA level. Moreover, YAP1 copy number gain and elevated YAP1 mRNA expression were detected in clinical samples analyzed in publicly available data sets. Taken together, these results suggested that YAP1 has a role in PCa tumorigenesis. C1 [Collak, Kisaayak Filiz] Istanbul Medeniyet University, Faculty of Engineering and Natural Sciences, Molecular Biology and Genetics Department, Uskudar, 34700 Istanbul, Turkey. [Ummuhan, Demir] Istanbul Medeniyet University, Faculty of Engineering and Natural Sciences, Molecular Biology and Genetics Department, Uskudar, 34700 Istanbul, Turkey. [Sagir, Fatma] Istanbul Medeniyet University, Faculty of Engineering and Natural Sciences, Molecular Biology and Genetics Department, Uskudar, 34700 Istanbul, Turkey. RP Collak, KF (reprint author), Istanbul Medeniyet University, Faculty of Engineering and Natural Sciences, Molecular Biology and Genetics Department, 34700 Istanbul, Turkey. EM filiz.collak@medeniyet.edu.tr CR Harvey KF, Pfleger CM, Hariharan IK, 2003, The Drosophila Mst ortholog, hippo, restricts growth and cell proliferation and promotes apoptosis. Cell. 114:457–467 Mahoney JE, Mori M, Szymaniak AD, Varelas X, Cardoso W, 2014, The hippo pathway effector yap controls patterning and differentiation of airway epithelial progenitors. Dev Cell 30:137– 150 Zhao B,Wei X, Li W, Udan RS, Yang Q, Kim J, et al. Inactivation of YAP oncoprotein by the hippo pathway is involved in cell contact inhibition and tissue growth control. 2007;2747–61 Ehmer U, Sage J, 2016, Control of proliferation and Cancer growth by the hippo signaling pathway. Mol Cancer Res 14:127–140 Zhao B, Ye X, Yu J, Li L, Li W, Li S, Yu J, Lin JD, Wang CY, Chinnaiyan AM, Lai ZC, Guan KL, 2008, TEAD mediates YAPdependent gene induction and growth control. GenesDev 22:1962– 1971 Alarcon C, Zaromytidou A-I, Xi Q, Gao S, Yu J, Fujisawa S et al, 2009, Nuclear CDKs drive Smad transcriptional activation and turnover in BMP and TGF- pathways. Cell. 139:757–769 Strano S, Munarriz E, Rossi M, Castagnoli L, Shaul Y, Sacchi A, Oren M, Sudol M, Cesareni G, Blandino G, 2001, Physical interaction with yes-associated protein enhances p73 transcriptional activity. J Biol Chem 276:15164–15173 Haskins JW, Nguyen DX, Stern DF, 2014, Neuregulin 1-activated ERBB4 interacts with YAP to induce hippo pathway target genes and promote cell migration. Sci Signal 7:ra116 Attard G, Parker C, Eeles RA, Schroder F, Tomlins SA, Tannock I, Drake CG, de Bono JS, 2016, Prostate cancer. Lancet. 387:70–82 Disibio G, French SW, 2008, Metastatic patterns of cancers: results from a large autopsy study. Arch Pathol Lab Med 132:931–939 Schrecengost R, Knudsen KE, 2013, Molecular pathogenesis and progression of prostate Cancer. Semin Oncol 40:244–258 Hu X, Jia Y, Yu J, Chen J, Fu Q, 2015, Loss of YAP protein in prostate cancer is associated with Gleason score increase. Tumori. 101:189–193 Yuan M, Tomlinson V, Lara R, Holliday D, Chelala C, Harada T, Gangeswaran R, Manson-Bishop C, Smith P, Danovi SA, Pardo O, Crook T, Mein CA, Lemoine NR, Jones LJ, Basu S, 2008, Yesassociated protein, YAP, functions as a tumor suppressor in breast. Cell Death Differ 15:1752–1759 Wang Y, Dong Q, Zhang Q, Li Z, Wang E, Qiu X, 2010, Overexpression of yes-associated protein contributes to progression and poor prognosis of non-small-cell lung cancer. Cancer Sci 101:1279–1285 Xu MZ, Yao T-J, Lee NPY, Ng IOL, Chan Y-T, Zender L, Lowe SW, Poon RTP, Luk JM, 2009, Yes-associated protein is an independent prognostic marker in hepatocellular carcinoma. Cancer. 115:4576–4585 Hall CA,Wang R, Miao J, Oliva E, Shen X,Wheeler T, Hilsenbeck SG, Orsulic S, Goode S, 2010, Hippo pathway effector yap is an ovarian cancer oncogene. Cancer Res 70:8517–8525 Zhang C, Liang K, Zhou G, Zhang Q, Li J, 2014, Expression of hippo pathway in colorectal cancer. Saudi J Gastroenterol 20:188 Zhou G-X, Li X-Y, Zhang Q, Zhao K, Zhang C-P, Xue C-H, Yang K, Tian ZB, 2013, Effects of the hippo signaling pathway in human gastric cancer. Asian Pac J Cancer Prev 14:5199–5205 Ge L, Smail M, Meng W, Shyr Y, Ye F, Fan K-H, Li X, Zhou HM, Bhowmick NAYes-associated protein expression in head and neck squamous cell carcinoma nodal metastasis. Ramqvist T, editor. PLoS One 2011;6:e27529 Pei T, Li Y, Wang J,Wang H, Liang Y, Shi H et al, 2015, YAP is a critical oncogene in human cholangiocarcinoma. Oncotarget. 6: 17206–17220 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, Antipin Y, Reva B, Goldberg AP, Sander C, Schultz N, 2012, The cBio Cancer genomics portal: an open platform for exploring multidimensional Cancer genomics data. Cancer Discov 2(5):401–404 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO et al, 2013, Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6:pl1 Bao Y, Hata Y, Ikeda M, Withanage K, 2011, Mammalian hippo pathway: from development to cancer and beyond. J Biochem 149: 361–379 Zhang L, Yang S, Chen X, Stauffer S, Yu F, Lele SM et al, 2015, The hippo pathway effector YAP regulates motility , invasion , and castration-resistant growth of. Prostate Cancer Cells 35:1350–1362 Kuser-Abali G, Alptekin A, Lewis M, Garraway IP, Cinar B, 2015, YAP1 and AR interactions contribute to the switch from androgendependent to castration-resistant growth in prostate cancer. Nat Commun 6:8126 Sheng X, Bin LW,Wang DL, Chen KH, Cao JJ, Luo Z et al, 2015, YAP is closely correlated with castration-resistant prostate cancer, and downregulation of YAP reduces proliferation and induces apoptosis of PC-3 cells. Mol Med Rep 12:4867–4876 Gu S, Chen K, Yin M, Wu Z, Wu Y, 2016, Proteomic profiling of isogenic primary and metastatic medulloblastoma cell lines reveals differential expression of key metastatic factors. J Proteome 160:55–63 Lee HJ, Diaz MF, Price KM, Ozuna JA, Zhang S, Sevick-muraca EM, et al. Fluid shear stress activates YAP1 to promote cancer cell motility. 2017; 8:14122 Haemmerle M, Taylor ML, Gutschner T, Pradeep S, Cho MS, Sheng J, Lyons YM, Nagaraja AS, Dood RL, Wen Y, Mangala LS, Hansen JM, Rupaimoole R, Gharpure KM, Rodriguez- Aguayo C, Yim SY, Lee JS, Ivan C, Hu W, Lopez-Berestein G, Wong ST, Karlan BY, Levine DA, Liu J, Afshar-Kharghan V, Sood AK, 2017, Platelets reduce anoikis and promote metastasis by activating YAP1 signaling. Nat Commun 8:310 Chai J, Xu S, Guo F, 2017, TEAD1 mediates the oncogenic activities of hippo-YAP1 signaling in osteosarcoma. Biochem Biophys Res Commun 488:297–302 Grannas K, Arngarden L, Lonn P, Mazurkiewicz M, Blokzijl A, Zieba A et al, 2015, Crosstalk between hippo and TGFβ: subcellular localization of YAP/TAZ/Smad complexes. J Mol Biol 427: 3407–3415 Ferrigno O, Lallemand F, Verrecchia F, L’Hoste S, Camonis J, Atfi Aet al, 2002, Yes-associated protein, YAP65, interacts with Smad7 and potentiates its inhibitory activity against TGF-beta/Smad signaling. Oncogene 21:4879–4884 Vitolo MI, Anglin IE, Mahoney WM, Renoud KJ, Gartenhaus RB, Bachman KE, Passaniti A, 2007, The RUNX2 transcription factor cooperates with the YES-associated protein, YAP65, to promote cell transformation. Cancer Biol Ther 6:856–863 Omerovic J, Puggioni EM, Napoletano S,ViscoV, Fraioli R, Frati L et al, 2004, Ligand-regulated association of ErbB-4 to the transcriptional co-activator YAP65 controls transcription at the nuclear level. Exp Cell Res 294:469–479 Hoang DT, Iczkowski KA, Kilari D, SeeW, Nevalainen MT, 2017, Androgen receptor-dependent and -independent mechanisms driving prostate cancer progression: opportunities for therapeutic targeting from multiple angles. Oncotarget 8:3724–3745 Shiota M, Yokomizo A, Fujimoto N, Naito S, 2011, Androgen receptor cofactors in prostate Cancer: potential therapeutic targets of castration-resistant prostate Cancer. Curr Cancer Drug Targets 11:870–881 Nguyen, Liem T.l. Nguyen LT, Tretiakova MS, Silvis MR et al. EA the YTP and I the D of A-RPTCC 2015;27(6):797–808., DOI 10. 1016/j. ccell. 2015. 05. 005. Tretiakova MS, Silvis MR, Lucas J, Klezovitch O, Coleman I et al, 2015, ERG Activates the YAP1 Transcriptional Program and Induces the Development of Age-Related Prostate Tumors. Cancer Cell 27:797–808 Lehmann W, Mossmann D, Kleemann J, Mock K, Meisinger C, Brummer T, Herr R, Brabletz S, Stemmler MP, Brabletz T ZEB1 turns into a transcriptional activator by interacting with YAP1 in aggressive cancer types. Nat Commun ; 2016;7:10498 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 867 EP 876 DI 10.1007/s12253-019-00634-z PG 10 ER PT J AU Roberto, MG Lira, CR Delsin, EL Vieira, MG Silva, OM Hakime, GR Yamashita, EM Engel, EE Scrideli, AC Tone, GL Brassesco, SM AF Roberto, Molinari Gabriela Lira, Caroline Regia Delsin, Elis Lara Vieira, Maciel Gabriela Silva, Oliveira Marcela Hakime, Guedes Rodrigo Yamashita, Eiji Mauricio Engel, Eduard Edgard Scrideli, Alberto Carlos Tone, Gonzaga Luiz Brassesco, Sol Maria TI microRNA-138-5p as a Worse Prognosis Biomarker in Pediatric, Adolescent, and Young Adult Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE miR-138; ROCK; microRNA; Osteosarcoma; Prognosis ID miR-138; ROCK; microRNA; Osteosarcoma; Prognosis AB Osteosarcoma (OS) is the most common primary malignant bone tumor with two peaks of incidence, in early adolescence and the elderly. Patients affected with this malignancy often present metastatic disease at diagnosis, and despite multimodality therapy, survival has not improved substantially over the past 3 decades. Recently, miR-138-5p, proposed as a crucial intracellular mediator of invasion, has been recognized to target the Rho-associated coiled-coil containing protein kinase 2 (ROCK2). Dysregulation of ROCK1 and ROCK2 was also described in OS, being associated to higher metastasis incidence and worse prognosis. Nonetheless, the specific roles of miR-138-5p in pediatric and young adult OS and its ability to modulate these kinases remain to be established. Thus, in the present study, the expression levels miR- 138-5p were evaluated in a consecutive cohort of exclusively pediatric and young adult primary OS samples. In contrast to previous reports that included adult tissues, our results showed upregulation of miR-138-5p associated with reduced event-free survival and relapsed cases. In parallel, ROCK1 mRNA levels were significantly reduced in tumor samples and negatively correlated with miR-138-5p. Similar correlations were observed after studying the profiles of ROCK1 and ROCK2 by immunohistochemistry. Our data present miR-138-5p as a consistent prognostic factor in pediatric and young adult OS, reinforcing its participation in the post-transcriptional regulation of ROCK kinases. C1 [Roberto, Molinari Gabriela] University of Sao Paulo, Ribeirao Preto School of Medicine, Regional Blood CenterSao Paulo, Brazil. [Lira, Caroline Regia] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of PediatricsSao Paulo, Brazil. [Delsin, Elis Lara] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of GeneticsSao Paulo, Brazil. [Vieira, Maciel Gabriela] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of GeneticsSao Paulo, Brazil. [Silva, Oliveira Marcela] University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell BiologySao Paulo, Brazil. [Hakime, Guedes Rodrigo] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of Biomechanics, Medicine and Rehabilitation of the Locomotor SystemSao Paulo, Brazil. [Yamashita, Eiji Mauricio] University of Sao Paulo, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, Department of BiologySao Paulo, Brazil. [Engel, Eduard Edgard] University of Sao Paulo, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, Department of BiologySao Paulo, Brazil. [Scrideli, Alberto Carlos] University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell BiologySao Paulo, Brazil. [Tone, Gonzaga Luiz] University of Sao Paulo, Ribeirao Preto School of Medicine, Department Cell BiologySao Paulo, Brazil. [Brassesco, Sol Maria] University of Sao Paulo, Ribeirao Preto School of Medicine, Department of Biomechanics, Medicine and Rehabilitation of the Locomotor SystemSao Paulo, Brazil. RP Brassesco, SM (reprint author), University of Sao Paulo, Ribeirao Preto School of Medicine, Department of Biomechanics, Medicine and Rehabilitation of the Locomotor System, Sao Paulo, Brazil. EM solbrassesco@usp.br CR Benjamin RS, 2015, Osteosarcoma: better treatment through better trial design. Lancet Oncol 16(1):12–13 Ottaviani G, Jaffe N, 2009, The etiology of osteosarcoma. Cancer Treat Res 152:15–32 Mirabello L, Troisi RJ, Savage SA, 2009, Osteosarcoma incidence and survival rates from 1973 to 2004. Cancer 115:1531–1543 Gorlick R, Khanna C, 2010, Osteosarcoma. J Bone Miner Res 25: 683–691 Harting MT, Blakely ML, 2006, Management of osteosarcoma pulmonary metastases. Semin Pediatr Surg 15:25–29 Isakoff MS, Bielack SS, Meltzer P, Gorlick R, 2015, Osteosarcoma: current treatment and a collaborative pathway to success. J Clin Oncol 33:3029–3035 Massague J, Obenauf AC, 2016, Metastatic colonization by circulating tumour cells. Nature 529:298–306 Liu X, Choy E, Hornicek FJ, Yang S, Yang C, Harmon D, Mankin H, Duan Z, 2011, ROCK1 as a potential therapeutic target in osteosarcoma. J Orthop Res 29:1259–1266 Wang W, Zhou X, Wei M, 2015, MicroRNA-144 suppresses osteosarcoma growth and metastasis by targeting ROCK1 and ROCK2. Oncotarget 6:10297–10308 Fusella F, Ferretti R, Recupero D, Rocca S, Di Savino A, Tornillo G, Silengo L, Turco E, Cabodi S, Provero P, Pandolfi PP, Sapino A, Tarone G, BrancaccioM, 2014, Morgana acts as a proto-oncogene through inhibition of a ROCK-PTEN pathway. J Pathol 234(2): 152–163 Lochhead PA, Wickman G, Mezna M, Olson MF, 2010, Activating ROCK1 somatic mutations in human cancer. Oncogene 29:2591–2598 Zucchini C, Manara MC, Pinca RS, De Sanctis P, Guerzoni C, Sciandra M, Lollini P-L, Cenacchi G, Picci P, Valvassori L, Scotlandi K, 2014, CD99 suppresses osteosarcoma cell migration through inhibition of ROCK2 activity. Oncogene 33:1912–1921 Tan Y, Hu H, Tan W, Jin L, Liu J, Zhou H, 2016, MicroRNA-138 inhibits migration and invasion of non-small cell lung cancer cells by targeting LIMK1. Mol Med Rep 14:4422–4428 Zhang J, Liu D, Feng Z,Mao J, Zhang C, LuY, Li J, Zhang Q, Li Q, Li L, 2016, MicroRNA-138 modulates metastasis and EMT in breast cancer cells by targeting vimentin. Biomed Pharmacother 77:135–141 Jiang L, Liu X, Kolokythas A,Yu J,Wang A,Heidbreder CE, Shi F, Zhou X, 2010, Downregulation of the Rho GTPase signaling pathway is involved in the microRNA-138-mediated inhibition of cell migration and invasion in tongue squamous cell carcinoma. Int J Cancer 127:505–512 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), method. Methods 25:402–408 Varghese F, Bukhari AB,Malhotra R, De A, 2014, IHC profiler: an open source plugin for the quantitative evaluation and automated scoring of immunohistochemistry images of human tissue samples. PLoS One 9:e96801 Li B, Yang X-X, Wang D, Ji H-K, 2016, MicroRNA-138 inhibits proliferation of cervical cancer cells by targeting c-Met. Eur Rev Med Pharmacol Sci 20:1109–1114 Liu X, Jiang L, Wang A, Yu J, Shi F, Zhou X, 2009, MicroRNA- 138 suppresses invasion and promotes apoptosis in head and neck squamous cell carcinoma cell lines. Cancer Lett 286:217–222 Long L, Huang G, Zhu H, Guo Y, Liu Y, Huo J, 2013, Downregulation of miR-138 promotes colorectal cancer metastasis via directly targeting TWIST2. J Transl Med 11:275 Mitomo S, Maesawa C, Ogasawara S, Iwaya T, Shibazaki M, Yashima-Abo A, Kotani K, Oikawa H, Sakurai E, Izutsu N, Kato K, Komatsu H, Ikeda K, Wakabayashi G, Masuda T, 2008, Downregulation of miR-138 is associated with overexpression of human telomerase reverse transcriptase protein in human anaplastic thyroid carcinoma cell lines. Cancer Sci 99:280–286 Sun D-K, Wang J-M, Zhang P, Wang Y-Q, 2015, MicroRNA-138 regulatesmetastatic potential of bladder Cancer through ZEB2. Cell Physiol Biochem 37:2366–2374 Xiao L, Zhou H, Li X-P, Chen J, Fang C,Mao C-X, Cui J-J, Zhang W, Zhou H-H, Yin J-Y, Liu Z-Q, 2016, MicroRNA-138 acts as a tumor suppressor in non small cell lung cancer via targeting YAP1. Oncotarget 7:40038–40046 Yu C,WangM, Li Z, Xiao J, Peng F, Guo X,Deng Y, Jiang J, Sun C, 2015, MicroRNA-138-5p regulates pancreatic cancer cell growth through targeting FOXC1. Cell Oncol, Dordr, 38:173–181 Wang Q, Tang H, Yin S, Dong C, 2013, Downregulation of microRNA-138 enhances the proliferation, migration and invasion of cholangiocarcinoma cells through the upregulation of RhoC/p- ERK/MMP-2/MMP-9. Oncol Rep 29:2046–2052 Yeh Y-M, Chuang C-M, Chao K-C,Wang L-H, 2013, MicroRNA- 138 suppresses ovarian cancer cell invasion and metastasis by targeting SOX4 and HIF-1α. Int J Cancer 133:867–878 Jiang B, Mu W, Wang J, Lu J, Jiang S, Li L, Xu H, Tian H, 2016, MicroRNA-138 functions as a tumor suppressor in osteosarcoma by targeting differentiated embryonic chondrocyte gene 2. J Exp Clin Cancer Res 35:69 Lane J, Martin TA, Watkins G, Mansel RE, Jiang WG, 2008, The expression and prognostic value of ROCK I and ROCK II and their role in human breast cancer. Int J Oncol 33:585–593 Mertsch S, Thanos S, 2014, Opposing signaling of ROCK1 and ROCK2 determines the switching of substrate specificity and the mode of migration of glioblastoma cells.Mol Neurobiol 49:900–915 Wu Y, Tang Y, Li Z, Li Z, Zhao Y, Wu Z, Su Q, 2014, Expression and significance of Rac1, Pak1 and Rock1 in gastric carcinoma. Asia Pac J Clin Oncol 10:e33–e39 Takeba Y, Matsumoto N, Watanabe M, Takenoshita-Nakaya S, Ohta Y, Kumai T, Takagi M, Koizumi S, Asakura T, Otsubo T, 2012, The rho kinase inhibitor fasudil is involved in p53- mediated apoptosis in human hepatocellular carcinoma cells. Cancer Chemother Pharmacol 69:1545–1555 Yang X, Di J, ZhangY, Zhang S, Lu J, Liu J, ShiW(2012, The rhokinase inhibitor inhibits proliferation and metastasis of small cell lung cancer. Biomed Pharmacother 66:221–227 Storey JD, Madeoy J, Strout JL, Wurfel M, Ronald J, Akey JM, 2007, Gene-expression variation within and among human populations. Am J Hum Genet 80:502–509 Zhang W, Dolan ME, 2008, Ancestry-related differences in gene expression: findings may enhance understanding of health disparities between populations. Pharmacogenomics 9:489–492 Scotting PJ, Walker DA, Perilongo G, 2005, Opinion: childhood solid tumours: a developmental disorder. Nat Rev Cancer 5:481–488 Boissel N, Auclerc MF, Lheritier V, Perel Y, Thomas X, Leblanc T, Rousselot P, Cayuela JM, Gabert J, Fegueux N, Piguet C, Huguet- Rigal F, Berthou C, Boiron JM, Pautas C, Michel G, Fiere D, Leverger G, Dombret H, Baruchel A, 2003, Should adolescents with acute lymphoblastic leukemia be treated as old children or young adults? Comparison of the French FRALLE-93 and LALA-94 trials. J Clin Oncol 21(5):774–780 Korshunov A, RemkeM,WerftW, Benner A, Ryzhova M,Witt H, Sturm D, Wittmann A, Schottler A, Felsberg J, Reifenberger G, Rutkowski S, Scheurlen W, Kulozik AE, von Deimling A, Lichter P, Pfister S, 2010, Adult and pediatricMedulloblastomas are genetically distinct and require different algorithms for molecular risk stratification. J Clin Oncol 28:3054–3060 Northcott PA, Hielscher T, Dubuc A, Mack S, Shih D, Remke M, Al-Halabi H, Albrecht S, Jabado N, Eberhart CG, Grajkowska W, WeissWA, Clifford SC, Bouffet E, Rutka JT, Korshunov A, Pfister S, Taylor MD, 2011, Pediatric and adult sonic hedgehog medulloblastomas are clinically and molecularly distinct. Acta Neuropathol 122:231–240 Paugh BS, Qu C, Jones C, Liu Z, Adamowicz-Brice M, Zhang J, Bax DA, Coyle B, Barrow J, Hargrave D, Lowe J, Gajjar A, Zhao W, Broniscer A, Ellison DW, Grundy RG, Baker SJ, 2010, Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease. J Clin Oncol 28: 3061–3068 Sultan I, Qaddoumi I, Yaser S, Rodriguez-Galindo C, Ferrari A, 2009, Comparing adult and pediatric rhabdomyosarcoma in the surveillance, epidemiology and end results program, 1973 to 2005: an analysis of 2,600 patients. J Clin Oncol 27:3391–3397 Akagi EM, Lavorato-Rocha AM, Maia B d M, Rodrigues IS, Carvalho KC, Stiepcich MM, Baiocchi G, Sato-Kuwabara Y, Rogatto SR, Soares FA, Rocha RM, 2014, ROCK1 as a novel prognostic marker in vulvar cancer. BMC Cancer 14:822 Dourado MR, de Oliveira CE, Sawazaki-Calone I, Sundquist E, Coletta RD, Salo T, 2017, Clinicopathologic significance of ROCK2 expression in oral squamous cell carcinomas. J Oral Pathol Med 47:121–127. , DOI 10.1111/jop.12651 Amaya CN, Mitchell DC, Bryan BA, 2017, Rho kinase proteins display aberrant upregulation in vascular tumors and contribute to vascular tumor growth. BMC Cancer 17:485 Islam M, Datta J, Lang JC, Teknos TN, 2014, Down regulation of RhoC by microRNA-138 results in de-activation of FAK, Src and Erk1/2 signaling pathway in head and neck squamous cell carcinoma. Oral Oncol 50:448–456 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 877 EP 883 DI 10.1007/s12253-019-00633-0 PG 7 ER PT J AU Kamyab-Hesary, K Ghanadan, A Balighi, K Mousavinia, FS Nasimi, M AF Kamyab-Hesary, Kambiz Ghanadan, Alireza Balighi, Kamran Mousavinia, Faeze Seyede Nasimi, Maryam TI Immunohistochemical Staining in the Assessment of Melanoma Tumor Thickness SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Melanoma; Breslow thickness; Immunohistochemical staining ID Melanoma; Breslow thickness; Immunohistochemical staining AB Vertical tumor thickness has great influence in the prognosis and staging of melanoma. The aim of this study was determination of the differences between melanoma tumor thickness in conventional hematoxylin and eosin (H&E) and immunohistochemical techniques. Thirty-six biopsy specimens were included in our study. For each sample, four adjacent tissue sections were stained with H&E, in addition S-100, Melan- A and HMB-45 staining was performed on the next serial sections. The mean thickness of tumor invasion was 2.16, 2.38, 2.22 and 2.29 mm in H&E, S-100, HMB45 and Melan-A sections evaluation, respectively. The mean difference of the Breslow thickness between H&E and S-100 and also, between H&E and Melan-A stained slides were statistically significant (p˂0.05) while no difference was found in the tumor thickness of the H&E and HMB45 staining evaluation (p = 0.278). Greater tumor thickness was observed in 25 lesions (69.4%) with S-100, 20 lesions (55.5%) with Melan-A and 17 (47.2%) lesions in HMB-45 rather than H&E staining. Conclusively, it appears that H&E staining cannot prove the actual size of melanoma invasion in some cases and immunohistochemical examination can be a complementary method in this situations. Of the melanoma associated immunomarkers, the combination of S-100 and Melan-A staining may suffice to measure depth of tumor invasion. C1 [Kamyab-Hesary, Kambiz] Tehran University of Medical Sciences, Razi Hospital, Department of DermatopathologyTehran, Iran. [Ghanadan, Alireza] Tehran University of Medical Sciences, Razi Hospital, Department of DermatopathologyTehran, Iran. [Balighi, Kamran] Tehran University of Medical Sciences, Razi Hospital, Department of Dermatology, Vahdate Eslami Street, 1199663911 Tehran, Iran. [Mousavinia, Faeze Seyede] Shahid Beheshti University of Medical SciencesTehran, Iran. [Nasimi, Maryam] Tehran University of Medical Sciences, Razi Hospital, Department of Dermatology, Vahdate Eslami Street, 1199663911 Tehran, Iran. RP Nasimi, M (reprint author), Tehran University of Medical Sciences, Razi Hospital, Department of Dermatology, 1199663911 Tehran, Iran. EM Nsm.maryam@gmail.com CR Breslow A, 1980, Prognosis in cutaneous melanoma: tumor thickness as a guide to treatment. Pathol Annu 15:1–22 Shaikh WR, Dusza SW, Weinstock MA et al, 2015, Melanoma thickness and survival trends in the United States, 1989–2009. J Natl Cancer Inst 12:108(1) Scolyer RA, Shaw HM, Thompson JF, Li LXL, Colman MH, Lo SK,McCarthy SW, Palmer AA, Nicoll KD, Dutta B, Slobedman E, Watson GF, Stretch JR, 2003, Interobserver reproducibility of histopathologic prognostic variables in primary cutaneousmelanomas. Am J Surg Pathol 27:1571–1576 Haydu LE, Stollman JT, Scolyer RA, Spillane AJ, Quinn MJ, Saw RPM, Shannon KF, Stretch JR, Bonenkamp JJ, Thompson JF, 2016, Minimum safe pathologic excision margins for primary cutaneous melanomas, 1–2 mm in thickness): analysis of 2131 patients treated at a single center. Ann Surg Oncol 23:1071–1081 Hurt MA, Santa Cruz DJ, 1994, Malignant melanoma microstaging. History, premises, methods, problems, and recommendations—a call for standardization. Pathol Annu 29: 51–74 Ohsie SJ, Sarantopoulos GP, Cochran AJ, Binder SW, 2008, Immunohistochemical characteristics of melanoma. J Cutan Pathol 35:433–444 Plaza JA, Suster D, Perez-Montiel D, 2007, Expression of immunohistochemical markers in primary and metastatic malignant melanoma: a comparative study in 70 patients using a tissuemicroarray technique. Appl Immunohistochem Mol Morphol 15:421–425 Paradela S, Fonseca E, Pita S, KantrowSM, GoncharukVN,Diwan H, Prieto VG, 2009, Spitzoid melanoma in children: clinicopathological study and application of immunohistochemistry as an adjunct diagnostic tool. J Cutan Pathol 36:740–752 Buzaid AC, Gershenwald JE, Atkins MB, Ross ME, 2016, Tumor node metastasis, TNM, staging system and other prognostic factors in cutaneous melanoma. UpToDate. UpToDate, Waltham Drabeni M, Lopez-Vilaro L, Barranco C, Trevisan G, Gallardo F, Pujol RM, 2013, Differences in tumor thickness between hematoxylin and eosin and Melan-A immunohistochemically stained primary cutaneous melanomas. Am J Dermatopathol 35:56–63 Xia J,Wang Y, Li F,Wang J,Mu Y,Mei X, Li X, ZhuW, Jin X, Yu K, 2016, Expression of microphthalmia transcription factor, S100 protein, and HMB-45 in malignant melanoma and pigmented nevi. Biomed Rep 5:327–331 Orchard GE, 1998, Melan A, MART-1): a new monoclonal antibody for malignant melanoma diagnosis. Br J Biomed Sci 55:8–9 Morton DL, Davtyan DG, Wanek LA, Foshag LJ, Cochran AJ, 1993, Multivariate analysis of the relationship between survival and the microstage of primary melanoma by Clark level and Breslow thickness. Cancer. 71:3737–3743 Urist MM, Balch CM, Soong SJ et al, 1984, Head and neck melanoma in 534 clinical Stage I patients. A prognostic factors analysis and results of surgical treatment. Ann Surg 200:769–775 Dyson SW, Bass J, Pomeranz J, Jaworsky C, Sigel J, Somach S, 2005, Impact of thorough block sampling in the histologic evaluation of melanomas. Arch Dermatol 141:734–736 de Vries TJ, Smeets M, de Graaf R, Hou-Jensen K, Brocker EB, Renard N, Eggermont AMM, van Muijen GNP, Ruiter DJ, 2001, Expression of gp100, MART-1, tyrosinase, and S100 in paraffinembedded primary melanomas and locoregional lymph node, and visceral metastases: implications for diagnosis and immunotherapy. A study conducted by the EORTC melanoma cooperative group. J Pathol 193:13–20 Kim RH, Meehan SA, 2017, Immunostain use in the diagnosis of melanomas referred to a tertiarymedical center: a 15-year retrospective review, 2001–2015). J Cutan Pathol 44:221–227 Penneys NS, 1987, Microinvasive lentigo maligna melanoma. J Am Acad Dermatol 17:675–680 Flugge G, Rassner G, 1989, Demonstration of S 100 protein in malignant melanoma of the skin. Pattern of distribution and significance for determination of tumor thickness. Hautarzt. 40:290–295 Prade M, Sancho-Garnier H, Cesarini JP, Cochran A, 1980, Difficulties encountered in the application of Clark classification and the Breslow thickness measurement in cutaneous malignant melanoma. Int J Cancer 26:159–163 Prieto VG, Shea CR, 2008, Use of immunohistochemistry in melanocytic lesions. J Cutan Pathol 35:1–10 Megahed M, Schon M, Selimovic D, Schon MP, 2002, Reliability of diagnosis of melanoma in situ. Lancet. 359:1921–1922 Thum C, Hollowood K, Birch J, Goodlad JR, Brenn T, 2011, Aberrant Melan-a expression in atypical fibroxanthoma and undifferentiated pleomorphic sarcoma of the skin. J Cutan Pathol 38:954–960 Bax MJ, Johnson TM, Harms PW, Schwartz JL, Zhao L, Fullen DR, Chan MP, 2016, Detection of occult invasion in melanoma in situ. JAMA Dermatol 152:1201–1208 Kim J, Taube J, McCalmont TH, Glusac EJ, 2011, Quantitative comparison of MiTF, Melan-a, HMB-45 and Mel-5 in solar lentigines and melanoma in situ. J Cutan Pathol 38:775–779 Helm TN, Helm KF, 2014, Breslow thickness determined with the use of Immunohistochemical techniques could provide misleading information when used with prognostic models based on data obtained by conventional means. Am J Dermatopathol 36:757 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 885 EP 891 DI 10.1007/s12253-019-00635-y PG 7 ER PT J AU Huang, Q Li, X Huang, Z Yu, F Wang, X Wang, Sh He, Z Lin, J AF Huang, Qingshan Li, Xiaodong Huang, Zhen Yu, Fengqiang Wang, Xinwen Wang, Shenglin He, Zhizhen Lin, Jianhua TI ACTN4 Promotes the Proliferation, Migration, Metastasis of Osteosarcoma and Enhances its Invasive Ability through the NF-κB Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; ACTN4; Invasion; Metastasis; NF-κB ID Osteosarcoma; ACTN4; Invasion; Metastasis; NF-κB AB Alpha-actinin-4 (ACTN4) is associated with different types of tumors, but its role in osteosarcoma (OS) is not known. We aimed to investigate the effect of ACTN4 on the growth, migration, invasion and metastasis of OS. We further explored the possible mechanism of how ACTN4 affects the development of OS. First, the expression of ACTN4 in OS tissues and OS cell lines was analyzed by PCR. Second, the role of ACTN4 in the development of OS was explored by the proliferation, scratch, and invasion assays. We further explored the effect of ACTN4 on OS growth in an orthotopic xenograft model of nude mice. In addition, we used hematoxylin and eosin (HE) staining of lung tissues in nude mice to observe the effect of ACTN4 on lung metastasis of OS. Finally, rescue experiments further investigated the role of NF-κB on ACTN4 in the development of OS. ACTN4 was highly expressed in OS tissues and OS cell lines. In vitro experiments demonstrated that reducing ACTN4 expression inhibited the proliferation, migration, and invasion of OS. In contrast, overexpression of ACTN4 promotes these effects. In vivo experiments further validated that ACTN4 promoted the growth of OS. The HE staining of lungs in nude mice revealed that ACTN4 promoted lung metastasis of OS. In addition, we found that ACTN4 enhanced the ability of OS to invade, through the NF-κB pathway. ACTN4 promotes the proliferation, migration, metastasis of OS and enhances its invasion ability through the NF-κB pathway C1 [Huang, Qingshan] The First Affiliated Hospital of Fujian Medical University, Department of Orthopedics, 350005 Fuzhou, China. [Li, Xiaodong] The First Affiliated Hospital of Fujian Medical University, Department of Orthopedics, 350005 Fuzhou, China. [Huang, Zhen] The First Affiliated Hospital of Fujian Medical University, Department of Orthopedics, 350005 Fuzhou, China. [Yu, Fengqiang] The First Affiliated Hospital of Fujian Medical University, Department of Orthopedics, 350005 Fuzhou, China. [Wang, Xinwen] The First Affiliated Hospital of Fujian Medical University, Department of Orthopedics, 350005 Fuzhou, China. [Wang, Shenglin] The First Affiliated Hospital of Fujian Medical University, Department of Orthopedics, 350005 Fuzhou, China. [He, Zhizhen] The First Affiliated Hospital of Fujian Medical University, Department of Orthopedics, 350005 Fuzhou, China. [Lin, Jianhua] The First Affiliated Hospital of Fujian Medical University, Department of Orthopedics, 350005 Fuzhou, China. RP Lin, J (reprint author), The First Affiliated Hospital of Fujian Medical University, Department of Orthopedics, 350005 Fuzhou, China. EM jianhua1@126.com CR Mirabello L, Troisi RJ, Savage SA, 2009, Osteosarcoma incidence and survival rates from 1973 to 2004: data from the surveillance, epidemiology, and end results program. Cancer 115(7):1531–1543. , DOI 10.1002/cncr.24121 Ottaviani G, Jaffe N, 2009, The epidemiology of osteosarcoma. Cancer Treat Res 152:3–13. , DOI 10.1007/978-1-4419- 0284-9_1 Rogozhin DV, Bulycheva IV, Konovalov DM, Talalaev AG, Roshchin VY, Ektova AP, Bogoroditsky YS, Strykov VA, Kazakova AN, Olshanskaya YV, Kachanov DY, Tereshchenko GV, 2015, [Classical osteosarcoma in children and adolescent]. Arkh Patol 77(5):68–74. , DOI 10.17116/ patol201577568-74 Marko TA, Diessner BJ, Spector LG, 2016, Prevalence of metastasis at diagnosis of osteosarcoma: an international comparison. Pediatr Blood Cancer 63(6):1006–1011 Bielack SS, Kempf-Bielack B, Delling G, Exner GU, Flege S, Helmke K, Kotz R, Salzer-Kuntschik M, Werner M, Winkelmann W, Zoubek A, Jurgens H, Winkler K, 2002, Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol 20(3):776–790. https://doi. org/10.1200/JCO.2002.20.3.776 Briccoli A, Rocca M, Salone M, Guzzardella GA, Balladelli A, Bacci G, 2010, High grade osteosarcoma of the extremities metastatic to the lung: long-term results in 323 patients treated combining surgery and chemotherapy, 1985-2005. Surg Oncol 19(4):193– 199. , DOI 10.1016/j.suronc.2009.05.002 Martin JW, Squire JA, Zielenska M, 2012, The genetics of osteosarcoma. Sarcoma 2012:627254. , DOI 10.1155/2012/ 627254 Li Z, Dou P, Liu T, He S, 2017, Application of long noncoding RNAs in osteosarcoma: biomarkers and therapeutic targets. Cell Physiol Biochem 42(4):1407–1419. , DOI 10.1159/ 000479205 Honda K, Yamada T, Endo R, Ino Y, Gotoh M, Tsuda H,Yamada Y, Chiba H, Hirohashi S, 1998, Actinin-4, a novel actin-bundling protein associated with cell motility and cancer invasion. J Cell Biol 140(6):1383–1393 Honda K, 2015, The biological role of actinin-4, ACTN4, in malignant phenotypes of cancer. Cell Biosci 5:41. , DOI 10. 1186/s13578-015-0031-0 Hsu KS, Kao HY, 2013, Alpha-actinin 4 and tumorigenesis of breast cancer. Vitam Horm 93:323–351. , DOI 10.1016/ B978-0-12-416673-8.00005-8 Ehrlicher AJ, Krishnan R, Guo M, Bidan CM, Weitz DA, Pollak MR, 2015, Alpha-actinin binding kinetics modulate cellular dynamics and force generation. Proc Natl Acad Sci U S A 112(21): 6619–6624. , DOI 10.1073/pnas.1505652112 Hamill KJ, Hopkinson SB, Skalli O, Jones JC, 2013, Actinin-4 in keratinocytes regulates motility via an effect on lamellipodia stability and matrix adhesions. FASEB J 27(2):546–556. , DOI 10.1096/fj.12-217406 Liu X, Chu KM, 2017, alpha-Actinin-4 promotes metastasis in gastric cancer. Lab Investig 97(9):1084–1094. , DOI 10. 1038/labinvest.2017.28 Kakuya T, Mori T, Yoshimoto S, Watabe Y, Miura N, Shoji H, Onidani K, Shibahara T, Honda K, 2017, Prognostic significance of gene amplification of ACTN4 in stage I and II oral tongue cancer. Int J Oral Maxillofac Surg 46(8):968–976. , DOI 10.1016/ j.ijom.2017.03.001 Zhong Z, Mao S, Lin H, Lin JM, Lin J, 2018, Comparative proteomics of cancer stem cells in osteosarcoma using ultra-highperformance liquid chromatography and Orbitrap fusion mass spectrometer. Talanta 178:362–368. , DOI 10.1016/j.talanta. 2017.09.053 Hoesel B, Schmid JA, 2013, The complexity of NF-kappaB signaling in inflammation and cancer. Mol Cancer 12:86. https://doi. org/10.1186/1476-4598-12-86 Sun SC, 2011, Non-canonical NF-kappaB signaling pathway. Cell Res 21(1):71–85. , DOI 10.1038/cr.2010.177 Bian D, Gao C, Bao K, Song G, 2017, The long non-coding RNA NKILA inhibits the invasion-metastasis cascade of malignant melanoma via the regulation of NF-kB. Am J Cancer Res 7(1):28–40 Liu M, Du K, Jiang B, Wu X, 2018, High expression of PhospholipaseD2 induced by hypoxia promotes proliferation of colon cancer cells through activating NF- kappa Bp65 signaling pathway. Pathol Oncol Res. , DOI 10.1007/s12253-018- 0429-1 Lomert E, Turoverova L, Kriger D, Aksenov ND, Nikotina AD, Petukhov A,Mittenberg AG, Panyushev NV, KhotinM, Volkov K, Barlev NA, Tentler D, 2018, Co-expression of RelA/p65 and ACTN4 induces apoptosis in non-small lung carcinoma cells. Cell Cycle 17(5):616–626. , DOI 10.1080/15384101.2017. 1417709 Berlin O, Samid D, Donthineni-Rao R, Akeson W, Amiel D, Woods VL Jr, 1993, Development of a novel spontaneous metastasis model of human osteosarcoma transplanted orthotopically into bone of athymic mice. Cancer Res 53(20):4890–4895 Naito S, von Eschenbach AC, Giavazzi R, Fidler IJ, 1986, Growth and metastasis of tumor cells isolated from a human renal cell carcinoma implanted into different organs of nude mice. Cancer Res 46(8):4109–4115 Harting MT, Blakely ML, 2006, Management of osteosarcoma pulmonary metastases. Semin Pediatr Surg 15(1):25–29. https:// doi.org/10.1053/j.sempedsurg.2005.11.005 Agarwal N, Adhikari AS, Iyer SV, Hekmatdoost K, Welch DR, Iwakuma T, 2013, MTBP suppresses cell migration and filopodia formation by inhibiting ACTN4. Oncogene 32(4):462–470. https:// doi.org/10.1038/onc.2012.69 Liao D, Zhong L, Duan T, Zhang RH, Wang X, Wang G, Hu K, Lv X, Kang T, 2015, Aspirin suppresses the growth and metastasis of osteosarcoma through the NF-kappaB pathway. Clin Cancer Res 21(23):5349–5359. , DOI 10.1158/1078- 0432.CCR-15-0198 Kovac B, Makela TP, Vallenius T, 2018, Increased alpha-actinin-1 destabilizes E-cadherin-based adhesions and associates with poor prognosis in basal-like breast cancer. PLoS One 13(5):e0196986. , DOI 10.1371/journal.pone.0196986 Inoue S, Ide H, Mizushima T, Jiang G, Netto GJ, Gotoh M, Miyamoto H, 2018, Nuclear factor-kappaB promotes urothelial tumorigenesis and cancer progression via cooperation with androgen receptor signaling. Mol Cancer Ther 17(6):1303–1314. https:// doi.org/10.1158/1535-7163.MCT-17-0786 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 893 EP 904 DI 10.1007/s12253-019-00637-w PG 12 ER PT J AU Jorgo, K Polgar, Cs Major, T Stelczer, G Herein, A Pocza, T Gesztesi, L Agoston, P AF Jorgo, Kliton Polgar, Csaba Major, Tibor Stelczer, Gabor Herein, Andras Pocza, Tamas Gesztesi, Laszlo Agoston, Peter TI Acute and Late Toxicity after Moderate Hypofractionation with Simultaneous Integrated Boost (SIB) Radiation Therapy for Prostate Cancer. A Single Institution, Prospective Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Simultaneous integrated boost; Moderate hypofractionation; Intensity-modulated radiotherapy; Image-guided radiotherapy ID Prostate cancer; Simultaneous integrated boost; Moderate hypofractionation; Intensity-modulated radiotherapy; Image-guided radiotherapy AB To evaluate the acute and late toxicity using moderately hypofractionated, intensity-modulated radiotherapy (IMRT) with a simultaneous integrated boost (SIB) to prostate for patients with intermediate and high risk prostate cancer. From 2015 to 2017, 162 patients were treated with IMRT with SIB to the prostate. IMRT plans were designed to deliver 50.4Gy in 28 fractions (1.8 Gy/fraction) to the pelvic lymph nodes (whole pelvis radiotherapy, WPRT) while simultaneously delivering 57.4 Gy in 28 fractions (2.05 Gy/fraction) to the seminal vesicles and 70 Gy in 28 fractions (2.5 Gy/fraction) to the prostate for high risk patients. For intermediate risk patients the same technique was applied, without WPRT. Acute and cumulative late genitourinary (GU) and gastrointestinal (GI) toxicities were scored according to the Radiation Therapy Oncology Group (RTOG) scoring system. Of the 162 patients enrolled, 156 (96%) completed the treatment as planned. The median follow-up time was 30 months. Seventy-eight patients (48.2%) were treated with WPRT. The rate of acute grade ≥ 2 GI and GU toxicities in all patients were 22% and 58%, respectively. The rate of cumulative late grade ≥ 2 GI and GU toxicities were 11% and 17%, respectively. Acute grade 3 GI and GU toxicities occurred in 1% and 1%. Late grade 3 GI and GU side effects occurred in 5% and 4%, respectively. None of the patients developed grade ≥ 4 toxicity. IMRT with SIB technique using moderate hypofractionation to the prostate is feasible treatment option for intermediate and high risk patients, associated with low rate of severe GU and GI toxicities. C1 [Jorgo, Kliton] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Polgar, Csaba] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Major, Tibor] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Stelczer, Gabor] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Herein, Andras] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Pocza, Tamas] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Gesztesi, Laszlo] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Agoston, Peter] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. RP Jorgo, K (reprint author), National Institute of Oncology, Center of Radiotherapy, 1122 Budapest, Hungary. EM jorgokliton@gmail.com CR Kuban DA, Tucker SL, Dong L et al, 2008, Long-term results of the M.D. Anderson randomized dose-escalation trial for prostatecancer. Int J Radiat Oncol Biol Phys 70:67–74 Peeters ST, Heemsbergen WD, Koper PC et al, 2006, Doseresponsein radiotherapy for localized prostate cancer: results of theDutch multicenter randomized phase III trial comparing 68Gy of radiotherapy with 78 Gy. J Clin Oncol 24:1990–1996 Zietman AL, DeSilvio ML, Slater JD et al, 2005, Comparison of conventional-dose vs. high-dose conformal radiation therapyin clinically localized adenocarcinoma of the prostate: a randomized controlled trial. JAMA 294:1233–1239 Miralbell R, Roberts SA, Zubizarreta E et al, 2012, Dose-fractionation sensitivity of prostate cancer deduced from radiotherapy outcomes of 5, 969 patients in seven international institutional datasets:α/β= 1.4, 0.9– 2.2, Gy. Int J Radiat Oncol Biol Phys 82:1017–1024 Brenner DJ, Hall EJ, 1999, Fractionation and protraction for radiotherapy of prostate carcinoma. Int J Radiat Oncol Biol Phys 43:1095–1101 Tree AC,KhooVS, vanAsNJ et al, 2014, Is biochemical relapse-free survival after profoundly hypofractionated radiotherapy consistent with current radiobiological models? Clin Oncol 26:216–229 Lee WR, Koontz BF, 2018, Moderate hypofractionation for prostate cancer. Transl Androl Urol 7:321–329 RoachMIII, 1993, Re: the use of prostate specific antigen, clinical stage and Gleason score to predict pathological stage in men with localized prostate cancer. J Urol 150:1923–1924 Joniau S, Van den Bergh L, Lerut E et al, 2013, Mapping of pelvic lymph node metastases in prostate cancer. Eur Urol 63:450–458 Heidenreich A, Varga Z, Von Knobloch R, 2002, Extended pelvic lymphadenectomy in patients undergoing radical prostatectomy: high incidence of lymph node metastasis. J Urol 167:1681–1686 Bader P, Burkhard FC, Markwalder R et al, 2002, Is a limited lymph node dissection an adequate staging procedure for prostate cancer? J Urol 168:514–518 Wyler SF, Sulser T, Seifert HH, Ruszat R, Forster TH, Gasser TC, Bachmann A, 2006, Laparoscopic extended pelvic lymph node dissection for high-risk prostate cancer. Urology 68:883–887 Arenas LF, Fullhase C, Boemans P et al, 2010, Detecting lymph nodes metastasis in prostate cancer through extended vs. standard laparoscopic pelvic lymphadenectomy. Aktuelle Urol 41:S10–S14 Seaward SA,Weinberg V, Lewis P, Leigh B, Phillips TL, Roach M III, 1998, Improved freedom from PSA failure with whole pelvic irradiation for high-risk prostate cancer. Int J Radiat Oncol Biol Phys 42:1055–1062 Seaward SA,Weinberg V, Lewis P et al, 1998, Identification of a highrisk clinically localised prostate cancer subgroup receiving maximum benefit from whole pelvic irradiation. Cancer J Sci Am 4:370–377 Pan CC, Kim KY, Taylor JM et al, 2002, Influence of 3D-CRT pelvic irradiation on outcome in prostate cancer treated with external beam radiotherapy. Int J RadiatOncol Biol Phys 53:1139–1145 Aizer AA, Yu JB, McKeon AM et al, 2009, Whole pelvic radiotherapy versus prostate only radiotherapy in the management of locally advanced or aggressive prostate adenocarcinoma. Int J Radiat Oncol Biol Phys 75:1344–1349 Milecki P, Baczyk M, Skowronek J et al, 2009, Benefit of whole pelvis radiotherapy combined with neoadjuvant androgen deprivation for the high-risk prostate cancer. J Biomed Biotechnol 2009:625394 Mantini G, Tagliaferri L, Mattiucci GC et al, 2011, Effect of whole pelvic radiotherapy for patients with locally advanced prostate cancer treated with radiotherapy and long-term androgen deprivation therapy. Int J Radiat Oncol Biol Phys 81:721–726 Asbell SO, Martz KL, Shin KH et al, 1998, Impact of surgical staging in evaluating the radiotherapeutic outcome in RTOP 77- 06, a phase III study for T1bN0M0, A2, and T2N0M0, B, prostate carcinoma. Int J Radiat Oncol Biol Phys 40:769–782 Lawton CA, DeSilvio M, Roach M et al, 2007, An update of the phase III trial comparing whole-pelvic to prostate-only radiotherapy and neo-adjuvant to adjuvant total androgen suppression: updated analysis of RTOG 94-13, with emphasis on unexpected hormone/ radiation interactions. Int J Radiat Oncol Biol Phys 69:646–655 Pommier P, Chabaud S, Lagrange JL et al, 2007, Is there a role for pelvic irradiation in localized prostate adenocarcinoma? Preliminary results of GETUG-01. J Clin Oncol 25:5366–5373 D’Amico AV, Moul J, Carroll PR et al, 2003, Cancer-specific mortality after surgery orradiation for patients with clinically localized prostate cancer managed during the prostate-specific antigen era. J Clin Oncol 21:2163–2172 Koszo, R., Varga, L., Fodor, E., Kahan Z., Cserhati A., Hideghety K., Egyud Z., Szabo C., Borzasi E., Szabo D., Mullner K., Varga Z., Maraz A., 2018, Prone positioning on a belly board decreases rectal and bowel doses in pelvic intensity-modulated radiation therapy, IMRT, for prostate. Cancer Pathol Oncol Res , DOI 10. 1007/s12253-018-0436-2 Lawton CA, Michalski J, El-Naqa I et al, 2009, RTOG GU radiation oncology specialists reach consensus on pelvic lymph node volumes for high-risk prostate cancer. Int J Radiat Oncol Biol Phys 74:383–387 Chao KK, Goldstein NS, Yan D, Vargas CE, Ghilezan MI, Korman HJ, Kernen KM, Hollander JB, Gonzalez JA, Martinez AA, Vicini FA, Kestin LL, 2006, Clinicopathologic analysis of extracapsular extension in prostate cancer: should the clinical target volume be expanded posterolaterally to account for microscopic extension? Int J Radiat Oncol Biol Phys 65:999–1007 Arcangeli G, Saracino B, Arcangeli S et al, 2017, Moderate hypofractionation in high-risk, organ-confined prostate cancer: final results of a phase III randomized trial. J Clin Onco l35:1891–1897 Pollack A,Walker G, Horwitz EM, Price R, Feigenberg S, Konski AA, Stoyanova R, Movsas B, Greenberg RE, Uzzo RG, Ma C, Buyyounouski MK, 2013, Randomized trial of hypofractionated external beam radiotherapy for prostate cancer. J Clin Oncol 31:3860–3868 HoffmanKE, VoongKR, Levy LB, PughTJ, Choi S, duW, Frank SJ, Johnson JL, Kudchadker R, Nguyen QN, Lee A, Mahmood U, McGuire SE, Kuban DA, 2016, Randomized trial of hypofractionated dose-escalated intensity modulated radiation therapy versus conventionally fractionated intensity modulated radiation therapy for localized prostate cancer. Int J Radiat Oncol Biol Phys 96:S32 Incrocci L, Wortel RC, Alemayehu WG et al, 2016, Hypofractionated versus conventionally fractionated radiotherapy for patients with localized prostate cancer, HYPRO): final efficacy results from a randomized, multicentre, open-label, phase 3 trial. Lancet Oncol 17:1061–1069 Aluwini S, Pos F, Schimmel E et al, 2016, Hypofractionated versus conventionally fractionated radiotherapy for patients with prostate cancer, HYPRO): late toxicity results from a randomized, non-inferiority, phase 3 trial. Lancet Oncol 17:464–474 Lee WR, Dignam JJ, AminMB, Bruner DW, Low D, Swanson GP, Shah AB, D’Souza DP, Michalski JM, Dayes IS, Seaward SA, Hall WA, Nguyen PL, Pisansky TM, Faria SL, Chen Y, Koontz BF, Paulus R, Sandler HM, 2016, Randomized phase IIInoninferiority study comparing two radiotherapy fractionation schedules in patients with low-risk prostate cancer. J Clin Oncol 34:2325–2332 Dearnaley D, Syndikus I, Mossop H et al, 2016, Conventional versus hypofractionated high-dose intensity-modulated radiotherapy for prostate cancer: 5-year outcomes of the randomised, noninferiority, phase 3CHHiP trial. Lancet Oncol 17:1047–1060 Wilkins A, Mossop H, Syndikus I et al, 2015, Hypofractionated radiotherapy versus conventionally fractionated radiotherapy for patients with intermediate-risk localized prostate cancer: 2-year patient- reported outcomes of the randomized, non-inferiority, phase 3CHHiP trial. Lancet Oncol 16:1605–1616 Catton CN, Lukka H, Gu CS, Martin JM, Supiot S, Chung PWM, Bauman GS, Bahary JP, Ahmed S, Cheung P, Tai KH, Wu JS, Parliament MB, Tsakiridis T, Corbett TB, Tang C, Dayes IS, Warde P, Craig TK, Julian JA, LevineMN(2017, Randomized trial of a hypofractionated radiation regimen for the treatment of localized prostate cancer. J Clin Oncol 35:1884–1890 Roach M, DeSilvio M, Lawton C et al, 2003, Radiation therapy oncology group 9413. Phase III trial comparing whole pelvic versus prostate only radiotherapy and neoadjuvant versus adjuvant combined androgen suppression: radiation therapy oncology group 9413. J Clin Oncol 21:1904–1911 Lawton CA, DeSilvio M, Roach M et al, 2007, An update of the phase III trial comparing whole pelvic to prostate only radiotherapy and neoadjuvant to adjuvant total androgen suppression: updated analysis of RTOG 94-13, with emphasis on unexpected hormone/ radiation interactions. Int J Radiat Oncol Biol Phys 69:646–655 Cozzarini C, Fiorino C, Muzio D et al, 2007, Significant reduction of acute toxicity following pelvic irradiation with helical Tomotherapy in patients with localized prostate cancer. Radiother Oncol 84:164–170 Di Muzio N, Fiorino C, Cozzarini C et al, 2009, Phase I-II study of hypofractionated simultaneous integrated boost with tomotherapy for prostate cancer. Int J Radiat Oncol Biol Phys 74:392–398 Di Muzio NG, Fodor A, Noris B et al, 2016, Moderate Hypofractionation with simultaneous integrated boost in prostate Cancer: long-term results of a phase I/II study. ClinOncol 28:490–500 Saracino B, Petrongari MG, Marzi S et al, 2014, Intensitymodulatedpelvic radiation therapy and simultaneous integrated boost tothe prostate area in patients with high-risk prostate cancer: apreliminary report of disease control. Cancer Med 3:1313–1321 Arcangeli G, Saracino B, Petrongari MG et al, 2007, Analysis of toxicity in patients with high risk prostate cancer treated with intensity-modulated pelvic radiation therapy and simultaneous integrated dose escalation to prostate area. Radiother Oncol 84:148–155 Franzese C, Fogliata A, D'Agostino GR et al, 2017, Moderate hypofractionated radiotherapy with volumetric modulated arc therapy and simultaneous integrated boost for pelvic irradiation in prostate cancer. J Cancer Res Clin Oncol 143:1301–1309 McCammon R, Rusthoven KE, Kavanagh B et al, 2009, Toxicity assessment of pelvic intensity-modulated radiotherapy with hypofractionated simultaneous integrated boost to prostate for intermediate- and high-risk prostate cancer. Int J Radiat Oncol Biol Phys 75:413–420 46. Engels B, Soete G, Tournel K et al, 2009, Helical tomotherapy with simultaneous integrated boost for high risk and lymph node positiveprostate cancer: early report on acute and late toxicity. Technol Cancer Res Treat 8:353–9 48. Adkison JB, McHaffie DR, Bentzen SM et al, 2012, Phase I trial of pelvic nodal dose escalation with hypofractionated IMRT for highrisk prostate cancer. Int J Radiat Oncol Biol Phys 82:184–190 49. Alongi F, Fogliata A, Navarria P et al, 2012, Moderate hypofractionation and simultaneous integrated boost with volumetric modulated arc therapy, RapidArc, for prostate cancer. Report of feasibility and acute toxicity. Strahlenther Onkol 188:990–996 50. Chang MG, Mukhopadhyay N, Holdford D, Skinner V, Saraiya S, Moghanaki D, Anscher MS, 2018, Phase 1/2 study of hypofractionated intensity-modulated radiation therapy for prostate cancer including simultaneously integrated boost. Pract Radiat Oncol 8:e149–e157 51. Magli A, Moretti E, Tullio A, Giannarini G, Tonetto F, Urpis M, Crespi M, Foti C, Prisco A, Polsinelli M, de Giorgi G, Bravo G, Scalchi P, TrovoM(2018, Hypofractionated simultaneous integrated boost, IMRT-SIB, with pelvic nodal irradiation and concurrent androgen deprivation therapy for high-risk prostate cancer: results of a prospective phase II trial. Prostate Cancer Prostatic Dis 21:269–276 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 905 EP 912 DI 10.1007/s12253-019-00623-2 PG 8 ER PT J AU Thangarasu, R Pachaiappan, P Subbaiyan, Th AF Thangarasu, Rajakumar Pachaiappan, Pugalendhi Subbaiyan, Thilagavathi TI Anti-Estrogenic and Anti-Cell Proliferative Effect of Allyl Isothiocyanate in Chemoprevention of Chemically Induced Mammary Carcinogenesis in Rats SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE 7-12-dimethylbenz(a)anthracene; Allyl isothiocyanate; Cell proliferation; Chemoprevention; Mammary carcinogenesis; N-methyl-N-nitrosourea ID 7-12-dimethylbenz(a)anthracene; Allyl isothiocyanate; Cell proliferation; Chemoprevention; Mammary carcinogenesis; N-methyl-N-nitrosourea AB The anti-estrogenic and anti-cell proliferative effect of allyl isothiocyanate (AITC) was carried out by analyzing the status of sex hormones and its receptors and cell proliferative markers in chemically induced mammary carcinogenesis in rats. Mammary tumor was induced by a single dose of DMBA (25 mg/rat) and MNU (50 mg/kg bw) injected subcutaneously near mammary gland. RT-PCR, western blotting and immunohistochemical analysis of mammary tissues show an upregulation of ER-α, PR, aromatase, PCNA, cyclin D1 and AgNORs staining and down regulation of p53 expression as well as plasma estradiol, prolactin and testosterone levels increased in DMBA and MNU-induced tumor bearing rats. Oral administration of AITC at a dose of 20 mg/kg bw restored the levels of sex hormones and its receptors, aromatase, cell proliferative markers and AgNORs staining near to normal levels. Molecular docking studies also supported these findings. The results suggest that anti-estrogenic and anti-proliferative effect of AITC prevent the development of DMBA and MNU-induced mammary carcinogenesis in rat. C1 [Thangarasu, Rajakumar] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, 608 002 Tamil Nadu, India. [Pachaiappan, Pugalendhi] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, 608 002 Tamil Nadu, India. [Subbaiyan, Thilagavathi] Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, 608 002 Tamil Nadu, India. RP Pachaiappan, P (reprint author), Annamalai University, Faculty of Science, Department of Biochemistry and Biotechnology, 608 002 Tamil Nadu, India. EM pugalau@gmail.com CR Siegel R,Ma J, Zou Z, Jemal A, 2014, Cancer statistics. CA Cancer J Clin 64:9–29 Yager JD, Davidson NE, 2006, Estrogen carcinogenesis in breast cancer. N Engl J Med 354:270–282 Mirzaie S, Chupani L, Asadabadi EB, Shahverdi AR, Jamalan M, 2013, Novel inhibitor discovery against aromatase through virtual screening and molecular dynamic simulation: a computational approach in drug design. EXCLI J 12:168–183 Mense SM, Remotti F, Bhan A, Singh B, El-Tamer M, Hei TK, Bhat HK, 2008, Estrogen-induced breast cancer: alterations in breast morphology and oxidative stress as a function of estrogen exposure. Toxicol Appl Pharmacol 232:78–85 Mertani HC, Garcia-Caballero T, Lambert A, Gerard F, Palayer C, Boutin JM, Vonderhaar BK,Waters MJ, Lobie PE,Morel G, 1998, Cellular expression of growth hormone and prolactin receptors in human breast disorders. Int J Cancer 79:202–211 Storer RD, Kraynak AR, Mckelvey TW, Elia MC, Goodrow TL, Deluca JQ, 1997, Themouse lymphoma L5178Y TK+/− cell line is heterozygous for a codon 170 mutation in the p53 tumor suppressor gene. Mutat Res 373:157–165 Kasala ER, Bodduluru LN, Madana RM, Athira KV, Gogoi R, Barua CC, 2015, Chemopreventive and therapeutic potential of chrysin in cancer: mechanistic perspectives. Toxicol Lett 233: 214–225 Caldon CE, Sutherland RL, Musgrove E, 2010, Cell cycle proteins in epithelial cell differentiation: implications for breast cancer. Cell Cycle 9:1918–1928 Arivazhagan L, Sorimuthu Pillai S, 2014, Tangeretin, a citrus pentamethoxyflavone, exerts cytostatic effect via p53/p21 up-regulation and suppresses metastasis in 7,12- dimethylbenz(α)anthracene-induced rat mammary carcinoma. J Nutr Biochem 25:1140–1153 Allred CD, Allred KF, Ju YH, Clausen LM, Doerge DR, Schantz SL, Korol DL,Wallig MA, Helferich WG, 2004, Dietary genistein results in larger MNU-induced, estrogen-dependent mammary tumors following ovariectomy of Sprague-Dawley rats. Carcinogenesis 25:211–218 Kwon KH, Barve A, Yu S, Huang MT, Kong AN, 2007, Cancer chemoprevention by phytochemicals: potential molecular targets, biomarkers and animal models. Acta Pharmacol Sin 28:1409–1421 Wattenberg LW, 1987, Inhibitory effects of benzyl isothiocyanates administered shortly before diethylnitrosamine or benzo[a]pyrene on pulmonary and forestomach neoplasia in A/J mice. Carcinogenesis 8:1971–1973 Xiao D, Srivastava SK, Lew KL, Zeng Y, Hershberger P, Johnson CS, Trump DL, Singh SV, 2003, Allyl isothiocyanate, a constituent of cruciferous vegetables, inhibits proliferation of human prostate cancer cells by causing G2/M arrest and inducing apoptosis. Carcinogenesis 24:891–897 Rajakumar T, Pugalendhi P, Thilagavathi S, 2015, Dose response chemopreventive potential of allyl isothiocyanate against 7,12- dimethylbenz(a)anthracene induced mammary carcinogenesis in female Sprague-Dawley rats. Chem Biol Interact 231:35–43 Ploton D,MenagerM, Jeannesson P, Himber G, Pigeon F, Adnet JJ, 1986, Improvement in the staining and in the visualization of the argyrophilic proteins of the nucleolar organizer region at the optical level. Histochem J 18:5–14 Chomczynski P, Sacchi N, 1987, Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem 162:156–159 Lowry OH, Rosebrough NJ, Farr AL, Randall RJ, 1951, Protein measurement with Folin phenol reagent. J Biol Chem 193:265–275 Santen RJ, Allred DC, Ardoin SP, Archer DF, Boyd N, Braunstein GD, Burger HG, Colditz GA, Davis SR, Gambacciani M, Gower BA, Henderson VW, JarjourWN, Karas RH, Kleerekoper M, Lobo RA, Manson JE, Marsden J, Martin KA, Martin L, Pinkerton JV, Rubinow DR, Teede H, Thiboutot DM, Utian WH, Endocrine Society, 2010, Postmenopausal hormone therapy: an Endocrine Society scientific statement. J Clin Endocrinol Metab 95:S1–S66 el-AzizMA, Hassan HA, MohamedMH,Meki AR, Abdel-Ghaffar SK, Hussein MR, 2005, The biochemical and morphological alterations following administration of melatonin, retinoic acid and Nigella sativa in mammary carcinoma: an animal model. Int J Exp Pathol 86:383–396 Lakshmi A, Subramanian S, 2014, Chemotherapeutic effect of tangeretin, a polymethoxylated flavone studied in 7,12- dimethylbenz(a)anthracene induced mammary carcinoma in experimental rats. Biochimie 99:96–109 Kang L, Ding L, Wang ZY, 2009, Isothiocyanates repress estrogen receptor alpha expression in breast cancer cells. Oncol Rep 21:185–192 Powers CN, Setzer WN, 2015, A molecular docking study of phytochemical estrogen mimics from dietary herbal supplements. In Silico Pharmacol 3:4 Clevenger CV, Furth PA, Hankinson SE, Schuler LA, 2003, The role of prolactin in mammary carcinoma. Endocr Rev 24:1–27 Cos S, Gonzalez A, Guezmes A, Mediavilla MD, Martinez-Campa C, Alonso-Gonzalez C, Sanchez-Barcelo EJ, 2006, Melatonin inhibits the growth of DMBA-induced mammary tumors by decreasing the local biosynthesis of estrogens through the modulation of aromatase activity. Int J Cancer 118:274–278 Brueggemeier RW, Hackett JC, Diaz-Cruz ES, 2005, Aromatase inhibitors in the treatment of breast cancer. Endocr Rev 26:331–345 Munusami P, Vasavi CS, Divya G, 2014, Molecular docking studies on flavonoid compounds: an insight into aromatase inhibitors. Int J Pharm Pharm Sci 6:141–148 Al-Dhaheri WS, Hassouna I, Al-Salam S, Karam SM, 2008, Characterization of breast cancer progression in the rat. Ann N Y Acad Sci 1138:121–131 Derenzini M, Ploton D, 1991, Interphase nucleolar organizer regions in cancer cells. Int Rev Exp Pathol 32:149–192 Derenzini M, Pession A, Trere D, 1990, Quantity of nucleolar silver-stained proteins is related to proliferating activity in cancer cells. Lab Investig 63:137–140 Agarwal ML, Agarwal A, Taylor WR, Stark GR, 1995, p53 controls both the G2/Mand the G1 cell cycle checkpoints andmediates reversible growth arrest in human fibroblasts. Proc Natl Acad Sci U S A 92:8493–8497 Tor YS, Yazan LS, Foo JB, Wibowo A, Ismail N, Cheah YK, Abdullah R, Ismail M, Ismail IS, Yeap SK, 2015, Induction of apoptosis in MCF-7 cells via oxidative stress generation, mitochondria-dependent and caspase-independent pathway by ethyl acetate extract of Dillenia suffruticosa and its chemical profile. PLoS One 10:e0127441 Chen NG, Chen KT, Lu CC, Lan YH, Lai CH, Chung YT, Yang JS, Lin YC, 2010, Allyl isothiocyanate triggers G2/M phase arrest and apoptosis in human brain malignant glioma GBM 8401 cells through a mitochondria-dependent pathway. Oncol Rep 24:449–455 Rengarajan T, Nandakumar N, Balasubramanian MP, 2013, DPinitol prevents rat breast carcinogenesis induced by 7, 12- Dimethylbenz [a] anthracene through inhibition of Bcl-2 and induction of p53, caspase-3 proteins and modulation of hepatic biotransformation enzymes and antioxidants. Biomed Prev Nutr 3:31–41 Gowtham Kumar S, Ramakrishnan V, Madhusudhanan N, Thiyagarajan G, Langeswaran VK, Balasubramanian MP, 2011, Apoptosis induced in hepatoma cells, hepg2, by allyl isothiocyanate associated with expression of p53 and activation of caspase-3. IJBPR 2:19–26 Yang YM, Conaway CC, Chiao JW, Wang CX, Amin S, Whysner J, Dai W, Reinhardt J, Chung FL, 2002, Inhibition of benzo(a)pyrene-induced lung tumorigenesis in a/J mice by dietary N-acetylcysteine conjugates of benzyl and phenethyl isothiocyanates during the postinitiation phase is associated with activation of mitogen-activated protein kinases and p53 activity and induction of apoptosis. Cancer Res 62:2–7 Bishayee A, Mandal A, 2014, Trianthema portulacastrum Linn. Exerts chemoprevention of 7,12-dimethylbenz(a)anthracene-induced mammary tumorigenesis in rats. Mutat Res 768:107–118 Rajmani RS, Doley J, Singh PK, Kumar R, Barathidasan R, Kumar P, Verma PC, Tiwari AK, 2011, Induction of mammary gland tumour in rats using N-methyl-N-nitroso urea and their histopathology. Indian J Vet Pathol 35:142–146 Yu Q, Geng Y, Sicinski P, 2001, Specific protection against breast cancers by cyclin D1 ablation. Nature 411:1017–1021 Jang TJ, Kang MS, Kim H, Kim DH, Lee JI, Kim JR, 2000, Increased expression of cyclin D1, cyclin E and p21(Cip1, associated with decreased expression of p27(Kip1, in chemically induced rat mammary carcinogenesis. Jpn J Cancer Res 91:1222–1232 AshrafiM, Bathaie SZ, Abroun S, 2012, High expression of cyclin D1 and p21 in N-Nitroso-N-Methylurea-induced breast Cancer in Wistar albino female rats. Cell J Fall 14:193–202 Chiao JW, Chung FL, Kancherla R, Ahmed T, Mittelman A, Conaway CC, 2002, Sulforaphane and its metabolite mediate growth arrest and apoptosis in human prostate cancer cells. Int J Oncol 20:631–636 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 913 EP 925 DI 10.1007/s12253-019-00638-9 PG 13 ER PT J AU Sohn, YS Lee, JJ Lee, HJ AF Sohn, Young Seo Lee, Jeong-Ju Lee, Hyuk Jae TI Molecular Profile and Clinicopathologic Features of Follicular Variant Papillary Thyroid Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NIFTP; BRAF; RAS; Thyroid neoplasm ID NIFTP; BRAF; RAS; Thyroid neoplasm AB The non-invasive encapsulated follicular variant of papillary thyroid carcinoma (FVPTC) has an indolent clinical behavior. Recently, it was proposed that this tumor type should be reclassified as non-invasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP). To characterize NIFTPs, we evaluated the molecular and clinicopathologic characteristics of each FVPTC subtype. This study enrolled 29 patients with FVPTC who underwent thyroidectomy between January 2007 and June 2017. They were classified as non-invasive encapsulated FVPTC (NIFTP, n = 10), invasive encapsulated FVPTC (n = 11), and infiltrative FVPTC (n = 8) by two independent pathologists. Genetic alterations were analyzed by targeted next-generation sequencing using formalin-fixed, paraffin-embedded tissue samples and the clinicopathologic characteristics were retrospectively reviewed. There was no difference in preoperative cytologic classification between NIFTPs and invasive encapsulated FVPTCs, whereas infiltrative FVPTC was more likely to be Bethesda class VI than the encapsulated type (50% versus 9.5%; P = 0.033). Lymph node metastasis was not found in NIFTPs. There was no BRAFV600E mutation in NIFTPs, whereas one of 11 invasive encapsulated FVPTCs and three of 8 infiltrative FVPTCs harbored BRAFV600E. RAS mutations were frequently detected in encapsulated FVPTCs (5 of 10 NIFTPs and 4 of 11 invasive encapsulated FVPTCs) but were only detected in one case of the infiltrative type. There were no differences in molecular or clinicopathologic profiles between non-invasive and invasive encapsulated FVPTCs, except for lymph node metastasis and the presence of BRAFV600E. NIFTP has favorable pathologic characteristics with a high frequency of RAS mutations. C1 [Sohn, Young Seo] Hanyang University College of Medicine, Myongji Hospital, Department of Internal Medicine, Division of Endocrinology and MetabolismGoyang, South Korea. [Lee, Jeong-Ju] Hanyang University College of Medicine, Myongji Hospital, Department of PathologyGoyang, South Korea. [Lee, Hyuk Jae] Hanyang University College of Medicine, Myongji Hospital, Department of Internal Medicine, Division of Endocrinology and MetabolismGoyang, South Korea. RP Sohn, YS (reprint author), Hanyang University College of Medicine, Myongji Hospital, Department of Internal Medicine, Division of Endocrinology and Metabolism, Goyang, South Korea. EM drsohnsy@gmail.com CR JKC C, 2000, Tumors of the thyroid and parathyroid glands.: Churchill Livingstone Lam AK, Lo CY, Lam KS, 2005, Papillary carcinoma of thyroid: a 30-yr clinicopathological review of the histological variants. Endocr Pathol 16(4):323–330 Tielens ET, Sherman SI, Hruban RH, Ladenson PW, 1994, Follicular variant of papillary thyroid carcinoma. A clinicopathologic study. Cancer. 73(2):424–431 Passler C, Prager G, Scheuba C et al, 2003, Follicular variant of papillary thyroid carcinoma: a long-term follow-up. Arch Surg 138(12):1362–1366 S. L., 1960, Carcinoma of the thyroid gland: Aclinical and pathologic study of 293 patients at the University of California hospital Chan J, 2002, Strict criteria should be applied in the diagnosis of encapsulated follicular variant of papillary thyroid carcinoma. Am J Clin Pathol 117(1):16–18 Liu J, Singh B, Tallini G et al, 2006, Follicular variant of papillary thyroid carcinoma: a clinicopathologic study of a problematic entity. Cancer. 107(6):1255–1264 Rivera M, Ricarte-Filho J, Knauf J et al, 2010)Molecular genotyping of papillary thyroid carcinoma follicular variant according to its histological subtypes, encapsulated vs infiltrative, reveals distinct BRAF and RAS mutation patterns. Mod Pathol 23(9):1191–1200 Vivero M, Kraft S, Barletta JA, 2013, Risk stratification of follicular variant of papillary thyroid carcinoma. Thyroid. 23(3):273–279 Bychkov A, Hirokawa M, Jung CK et al, 2017, Low rate of noninvasive follicular thyroid neoplasm with papillary-like nuclear features in Asian practice. Thyroid. 27(7):983–984 Finnerty BM, KleimanDA, Scognamiglio Tet al, 2015, Navigating the management of follicular variant papillary thyroid carcinoma subtypes: a classic PTC comparison. Ann Surg Oncol 22(4): 1200–1206 Piana S, Frasoldati A, Di Felice E, Gardini G, Tallini G, Rosai J, 2010, Encapsulated well-differentiated follicular-patterned thyroid carcinomas do not play a significant role in the fatality rates from thyroid carcinoma. Am J Surg Pathol 34(6):868–872 Nikiforov YE, Seethala RR, Tallini G et al, 2016, Nomenclature revision for encapsulated follicular variant of papillary thyroid carcinoma: a paradigm shift to reduce overtreatment of indolent tumors. JAMA Oncol 2(8):1023–1029 Howitt BE, Jia Y, Sholl LM, Barletta JA, 2013, Molecular alterations in partially-encapsulated or well-circumscribed follicular variant of papillary thyroid carcinoma. Thyroid. 23(10):1256–1262 Kim TH, Lee M, Kwon AY et al, 2018, Molecular genotyping of the non-invasive encapsulated follicular variant of papillary thyroid carcinoma. Histopathology. 72(4):648–661 Howitt BE, Paulson VA, Barletta JA, 2015, Absence of BRAF V600E in non-infiltrative, non-invasive follicular variant of papillary thyroid carcinoma. Histopathology. 67(4):579–582 Kim M, Jeon MJ, Oh HS et al, 2018, BRAF and RAS mutational status in noninvasive follicular thyroid neoplasm with papillary-like nuclear features and invasive subtype of encapsulated follicular variant of papillary thyroid carcinoma in Korea. Thyroid. 28(4): 504–510 Zhao L, Dias-Santagata D, Sadow PM, Faquin WC, 2017, Cytological, molecular, and clinical features of noninvasive follicular thyroid neoplasm with papillary-like nuclear features versus invasive forms of follicular variant of papillary thyroid carcinoma. Cancer Cytopathol 125(5):323–331 Cibas ES, Ali SZ, 2009, The Bethesda system for reporting thyroid cytopathology. Thyroid. 19(11):1159–1165 Li H, Durbin R, 2009, Fast and accurate short read alignment with burrows-wheeler transform. Bioinformatics. 25(14):1754–1760 DePristo MA, Banks E, Poplin R et al, 2011, A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet 43(5):491–498 Wang K, Li M, Hakonarson H, 2010, ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res 38(16):e164 Cingolani P, Platts A, Wang le L et al, 2012, A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly, Austin, 6(2):80–92 Landrum MJ, Lee JM, Benson M et al, 2016, ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res 44(D1):D862–D868 Wei Z, Wang W, Hu P, Lyon GJ, Hakonarson H, 2011, SNVer: a statistical tool for variant calling in analysis of pooled or individual next-generation sequencing data. Nucleic Acids Res 39(19):e132 McFadden DG, Dias-Santagata D, Sadow PM et al, 2014, Identification of oncogenic mutations and gene fusions in the follicular variant of papillary thyroid carcinoma. J Clin Endocrinol Metab 99(11):E2457–E2462 Kim MJ, Won JK, Jung KC et al, 2018, Clinical characteristics of subtypes of follicular variant papillary thyroid carcinoma. Thyroid. 28(3):311–318 Cho U, Mete O, Kim MH, Bae JS, Jung CK, 2017, Molecular correlates and rate of lymph node metastasis of non-invasive follicular thyroid neoplasm with papillary-like nuclear features and invasive follicular variant papillary thyroid carcinoma: the impact of rigid criteria to distinguish non-invasive follicular thyroid neoplasm with papillary-like nuclear features. Mod Pathol 30(6):810–825 Nikiforov YE, Baloch ZW, Hodak SP et al, 2018, Change in diagnostic criteria for noninvasive follicular thyroid neoplasm with Papillarylike nuclear features. JAMA Oncol 4(8):1125–1126 Trovisco V, Soares P, Soares R, Magalhaes J, Sa-Couto P, Sobrinho- SimoesM(2005, A new BRAF gene mutation detected in a case of a solid variant of papillary thyroid carcinoma. Hum Pathol 36(6): 694–697 Lee SR, Jung CK, Kim TE et al, 2013, Molecular genotyping of follicular variant of papillary thyroid carcinoma correlates with diagnostic category of fine-needle aspiration cytology: values of RAS mutation testing. Thyroid. 23(11):1416–1422 Jang MA, Lee ST, Oh YL et al, 2012, Identification of a rare 3 bp BRAF gene deletion in a thyroid nodule bymutant enrichment with 3′-modified oligonucleotides polymerase chain reaction. Ann Lab Med 32(3):238–241 Jung SH, Kim MS, Jung CK et al, 2016, Mutational burdens and evolutionary ages of thyroid follicular adenoma are comparable to those of follicular carcinoma. Oncotarget. 7(43):69638–69648 Nicolson NG, Murtha TD, Dong W et al, 2018, Comprehensive genetic analysis of follicular thyroid carcinoma predicts prognosis independent of histology. J Clin EndocrinolMetab 103:2640–2650 NikiforovaMN,Wald AI, Roy S, Durso MB, Nikiforov YE, 2013, Targeted next-generation sequencing panel, ThyroSeq, for detection ofmutations in thyroid cancer. J Clin Endocrinol Metab 98(11): E1852–E1860 Kunstman JW, Juhlin CC, Goh G et al, 2015, Characterization of the mutational landscape of anaplastic thyroid cancer via wholeexome sequencing. Hum Mol Genet 24(8):2318–2329 Guerra A, Sapio MR, Marotta V et al, 2012, The primary occurrence of BRAF(V600E, is a rare clonal event in papillary thyroid carcinoma. J Clin Endocrinol Metab 97(2):517–524 Ustun B, Chhieng D, Prasad ML et al, 2014, Follicular variant of papillary thyroid carcinoma: accuracy of FNA diagnosis and implications for patient management. Endocr Pathol 25(3):257–264 Paulson VA, Shivdasani P, Angell TE et al, 2017, Noninvasive follicular thyroid neoplasm with papillary-like nuclear features accounts for more than half of "carcinomas" harboring RAS mutations. Thyroid. 27(4):506–511 Shi X, Liu R, Basolo F et al, 2016, Differential Clinicopathological risk and prognosis ofmajor papillary thyroid Cancer variants. J Clin Endocrinol Metab 101(1):264–274 Haugen BR, Sawka AM, Alexander EK et al, 2017, American Thyroid Association guidelines on the Management of Thyroid Nodules and Differentiated Thyroid Cancer Task Force Review and recommendation on the proposed renaming of encapsulated follicular variant papillary thyroid carcinoma without invasion to noninvasive follicular thyroid neoplasm with papillary-like nuclear features. Thyroid. 27(4):481–483 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 927 EP 936 DI 10.1007/s12253-019-00639-8 PG 10 ER PT J AU Qi, J Ni, W AF Qi, Junhui Ni, Wei TI Attenuation of MAMLD1 Expression Suppresses the Growth and Migratory Properties of Gonadotroph Pituitary Adenomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pituitary adenoma; MAMLD1; CXorf6; Notch ID Pituitary adenoma; MAMLD1; CXorf6; Notch AB Gonadotroph pituitary adenomas (GPAs) constitute approximately 15–40% of pituitary tumors. Some GPAs can be highly infiltrative, making full surgical resection challenging and increasing the risk of recurrence. The transcriptional co-activator Mastermind-Like Domain Containing 1 (MAMLD1, CXorf6, F18) is involved in regulating signaling pathways important in pituitary tumorigenesis, including the Notch signaling pathway. However, MAMLD1’s role in GPA remains unknown. GPA biopsies were collected from 96 patients following surgery, who were monitored until tumor recurrence. GPA tissue was used for immunohistochemistry. The murine GPA cell lines αT3 and LβT2 were used for in vitro experiments. Lentiviral constructs were employed for MAMLD1 knockdown (KD) and dominant negative (DN) mutant experiments. Quantitative real-time PCR (qPCR) and Western blotting of MAMLD1 and Notch2 were performed. MTT and Transwell assays were used to quantify proliferation and migration, respectively. An αT3 xenograft model was established in athymic nude mice followed by fluorescent IHC of xenograft tumors. MAMLD1 and Notch2 levels correlated positively with aggressive GPAs. Increased MAMLD1 levels correlated with shortened recurrence-free survival (RFS) in aggressive GPA patients. Moreover, MAMLD1 expression independently affected patient RFS according to multivariate Cox regression. In vitro, MAMLD1 KD in the murine GPA cell lines attenuated their proliferation and migration and Notch2 expression. Additionally, DN MAMLD1L210X lowered their proliferative and migratory capacity. MAMLD1 KD suppressed tumor growth and Notch2 expression in murine xenografts. MAMLD1 may serve as a predictor of GPA patient outcome and may also be leveraged as a possible therapeutic target for aggressive GPA tumors. C1 [Qi, Junhui] The Second Hospital of Yunnan Province, Department of NeurosurgeryKunming, Yunnan Province, China. [Ni, Wei] The Third Affiliated Hospital of Kunming Medical University, Department of Neurosurgery, No. 519, Kunzhou Road, 650118 Kunming, Yunnan Province, China. RP Ni, W (reprint author), The Third Affiliated Hospital of Kunming Medical University, Department of Neurosurgery, 650118 Kunming, China. EM niweikm2018@163.com CR Scheithauer BW, Gaffey TA, Lloyd RV, Sebo TJ, Kovacs KT, Horvath E, Yapicier O, Young WF Jr, Meyer FB, Kuroki T, Riehle DL, Laws ER Jr, 2006, Pathobiology of pituitary adenomas and carcinomas. Neurosurgery 59(2):341–353 Knosp E, Steiner E, Kitz K, Matula C, 1993, Pituitary adenomas with invasion of the cavernous sinus space: a magnetic resonance imaging classification compared with surgical findings. Neurosurgery 33(4):610–618 Donoho DA, Bose N, Zada G, Carmichael JD, 2017, Management of aggressive growth hormone secreting pituitary adenomas. Pituitary 20(1):169–178 Dhandapani S, Singh H, Negm HM, Cohen S, Anand VK, Schwartz TH, 2016, Cavernous sinus invasion in pituitary adenomas: systematic review and pooled data meta-analysis of radiologic criteria and comparison of endoscopic and microscopic surgery. World Neurosurg 96:36–46 Qian ZR, Sano T, Yoshimoto K, Asa SL, Yamada S, Mizusawa N, Kudo E, 2007, Tumor-specific downregulation and methylation of the CDH13, H-cadherin, and CDH1, E-cadherin, genes correlate with aggressiveness of human pituitary adenomas. Mod Pathol 20(12):1269–1277 Chaidarun SS, Klibanski A, editors. Gonadotropinomas. Seminars in reproductive medicine; 2002: Copyright© 2002 by Thiememedical publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA. Tel:+ 1, 212, 584–4662 Young JrWF, Scheithauer BW, Kovacs KT, Horvath E, Davis DH, Randall RV, editors. Gonadotroph adenoma of the pituitary gland: a clinicopathologic analysis of 100 cases. Mayo Clin Proc; 1996: Elsevier Fukami M, Wada Y, Okada M, Kato F, Katsumata N, Baba T, Morohashi KI, Laporte J, Kitagawa M, Ogata T, 2008, Mastermind-like domain-containing 1, MAMLD1 or CXorf6, transactivates the Hes3 promoter, augments testosterone production, and contains the SF1 target sequence. J Biol Chem 283(9):5525–5532 Andreiuolo F, Varlet P, Tauziede-Espariat A, Junger ST, Dorner E, Dreschmann V, Kuchelmeister K, Waha A, Haberler C, Slavc I, Corbacioglu S, Riemenschneider MJ, Leipold A, Rudiger T, Korholz D, Acker T, Russo A, Faber J, Sommer C, Armbrust S, Rose M, Erdlenbruch B, Hans VH, Bernbeck B, Schneider D, Lorenzen J, Ebinger M, Handgretinger R, Neumann M, van Buiren M, Prinz M, Roganovic J, Jakovcevic A, Park SH, Grill J, Puget S, Messing-Junger M, Reinhard H, Bergmann M, Hattingen E, Pietsch T, 2018, Childhood supratentorial ependymomas with YAP 1-MAMLD 1 fusion: an entity with characteristic clinical, radiological, cytogenetic and histopathological features. Brain Pathol Archer TC, Pomeroy SL, 2015, Defining the molecular landscape of ependymomas. Cancer Cell 27(5):613–615 Chen Y, Thai HT, Lundin J, Lagerstedt-Robinson K, Zhao S, Markljung E et al, 2010, Mutational study of the MAMLD1-gene in hypospadias. Eur J Med Genet 53(3):122–126 Ruiz-Arana I-L, Hubner A, Cetingdag C, Krude H, Gruters A, Fukami M et al, 2015, A novel hemizygous mutation of MAMLD1 in a patient with 46, XY complete gonadal dysgenesis. Sex Dev 9(2):80–85 McElhinny A, Li J, Wu L, 2008, Mastermind-like transcriptional co-activators: emerging roles in regulating cross talk among multiple signaling pathways. Oncogene 27(38):5138–5147 Liu Q, Wang J, Yang H, Gao H, Li C, Lan X, Zhang Y, 2018, Attenuation of EGFL7 expression inhibits growth hormone– producing pituitary adenomas growth and invasion. Hum Gene Ther 29:1396–1406 Wang J, Liu Q, Gao H, Wan D, Li C, Li Z et al, 2017, EGFL7 participates in regulating biological behavior of growth hormone– secreting pituitary adenomas via Notch2/DLL3 signaling pathway. Tumor Biol 39(7):1010428317706203 DongW, Zhu H, Gao H, ShiW, Zhang Y,Wang H et al Expression of cyclin E/Cdk2/p27Kip1 in growth hormone adenomas. World Neurosurg 2018 Shinoda K, Lei H, Yoshii H, Nomura M, Nagano M, Shiba H, Sasaki H, Osawa Y, Ninomiya Y, Niwa O, Morohashi KI, Li E, 1995, Developmental defects of the ventromedial hypothalamic nucleus and pituitary gonadotroph in the Ftz-F1 disrupted mice. Dev Dyn 204(1):22–29 Asa SL, Bamberger A, Cao B,WongM, Parker KL, Ezzat S, 1996, The transcription activator steroidogenic factor-1 is preferentially expressed in the human pituitary gonadotroph. J Clin Endocrinol Metab 81(6):2165–2170 Polidoro MA, Morace R, Arcella A, Esposito V, Giangaspero F, Jaffrain-Rea M-L, editors. Factors associated with SF1 gene expression in clinically non-functioning pituitary adenomas. 20th European Congress of Endocrinology; 2018: BioScientifica Chesnokova V, Zonis S, Zhou C, Ben-Shlomo A, Wawrowsky K, Toledano Y, Tong Y, Kovacs K, Scheithauer B, Melmed S, 2011, Lineage-specific restraint of pituitary gonadotroph cell adenoma growth. PLoS One 6(3):e17924 Camats N, Fernandez-Cancio M, Audi L, Mullis PE, Moreno F, Casado IG et al, 2015, Human MAMLD1 gene variations seem not sufficient to explain a 46, XY DSD phenotype. PLoS One 10(11):e0142831 Laporte J, Kioschis P, Hu L-J, Kretz C, Carlsson B, Poustka A, Mandel JL, Dahl N, 1997, Cloning and characterization of an alternatively spliced gene in proximal Xq28 deleted in two patients with intersexual genitalia and myotubular myopathy. Genomics 41(3):458–462 Nakamura M, Fukami M, Sugawa F, Miyado M, Nonomura K, Ogata T, 2011, Mamld1 knockdown reduces testosterone production and Cyp17a1 expression in mouse Leydig tumor cells. PLoS One 6(4):e19123 MiyadoM, Yoshida K, Miyado K, Katsumi M, Saito K, Nakamura S, Ogata T, FukamiM(2017, Knockout ofmurineMamld1 impairs testicular growth and daily spermproduction but permits Normal postnatal androgen production and fertility. Int J Mol Sci 18(6):1300 Miyado M, Miyado K, Katsumi M, Saito K, Nakamura A, Shihara D, Ogata T, Fukami M, 2015, Parturition failure in mice lacking Mamld1. Sci Rep 5:14705 Giachino C, Boulay J-L, Ivanek R, Alvarado A, Tostado C, Lugert S, Tchorz J, Coban M, Mariani L, Bettler B, Lathia J, Frank S, Pfister S, Kool M, Taylor V, 2015, A tumor suppressor function for Notch signaling in forebrain tumor subtypes. Cancer Cell 28(6): 730–742 Yuan X,Wu H, Xu H, Xiong H, Chu Q,Yu S,Wu GS,WuK, 2015, Notch signaling: an emerging therapeutic target for cancer treatment. Cancer Lett 369(1):20–27 Aung KL, El-Khoueiry AB, Gelmon K, Tran B, Bajaj G, He B et al, 2018, A multi-arm phase I dose escalating study of an oral NOTCH inhibitor BMS-986115 in patients with advanced solid tumours. Investig New Drugs:1–11 Mir O, Azaro A, Merchan J, Chugh R, Trent J, Rodon J, Ohnmacht U, Diener JT, Smith C,Yuen E, Oakley GJ III, le Cesne A, Soria JC, Benhadji KA, Massard C, 2018, Notch pathway inhibition with LY3039478 in soft tissue sarcoma and gastrointestinal stromal tumours. Eur J Cancer 103:88–97 Massard C,Azaro A, Soria J-C, Lassen U, Le Tourneau C, Sarker D et al, 2018, First-in-human study of LY3039478, an Oral Notch signaling inhibitor in advanced or metastatic cancer. Ann Oncol 29(9):1911–1917 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 937 EP 946 DI 10.1007/s12253-019-00615-2 PG 10 ER PT J AU Gao, C Shen, J Meng, ZX He, XF AF Gao, Cao Shen, Jiang Meng, Zhi-Xiu He, Xiao-Feng TI Sevoflurane Inhibits Glioma Cells Proliferation and Metastasis through miRNA-124-3p/ROCK1 Axis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sevoflurane; Glioma; miRNA-124-3p; ROCK1 ID Sevoflurane; Glioma; miRNA-124-3p; ROCK1 AB Malignant glioma is the most common primary malignancy in the brain. It is aggressive, highly invasive, and destructive. Studies have shown that sevoflurane can affect the invasion and migration of a variety of malignant tumors. However, its effects on human glioma cells and related mechanisms are not clear. Cultured U251 and U87 cells were pretreated with sevoflurane. The effect of sevoflurane on cell proliferation, migration, apoptosis and invasion ability were evaluated by MTT, wound healing assay, cell apoptosis and transwell assays, respectively. miRNA-124-3p and ROCK1 signaling pathway genes expression in sevoflurane treated cell lines was measured by quantitative real-time PCR (qRT-PCR) and western blotting analysis. The potential target genes of miRNA were predicted by online software. Luciferase reporter assay was employed to validate the direct targeting of ROCK1 by miRNA-124-3p. In present studies, sevoflurane inhibits glioma cells proliferation, invasion and migration. Additionally, inversely correlation between miR-124-3p and ROCK1 expression in sevoflurane treated glioma cells was observed. Furthermore, sevoflurane inhibits glioma cells proliferation, migration and invasion through miR-124-3p/ROCK1 axis. Taken together, our study revealed that sevoflurane can inhibit glioma cell proliferation, invasion and migration. Its mechanism may be related to the upregulation of miR-124-3p, which suppresses ROCK1 signaling pathway. The results of the study will help to understand the pharmacological effects of inhaled general anesthetics more comprehensively and help to provide an experimental basis for selecting more reasonable anesthetics for cancer patients. C1 [Gao, Cao] The Third Affiliated Hospital of Soochow University, Department of Anesthesiology, 213002 Changzhou, China. [Shen, Jiang] The Third Affiliated Hospital of Soochow University, Department of Anesthesiology, 213002 Changzhou, China. [Meng, Zhi-Xiu] The Third Affiliated Hospital of Soochow University, Department of Anesthesiology, 213002 Changzhou, China. [He, Xiao-Feng] The Third Affiliated Hospital of Soochow University, Department of Anesthesiology, 213002 Changzhou, China. RP He, XF (reprint author), The Third Affiliated Hospital of Soochow University, Department of Anesthesiology, 213002 Changzhou, China. EM hexiaofeng1567@163.com CR Sun Q, Xu R, Xu H,Wang G, Shen X, Jiang H, 2017, Extracranial metastases of high-grade glioma: the clinical characteristics and mechanism. World J Surg Oncol 15(1):181 Kim R, 2018, Effects of surgery and anesthetic choice on immunosuppression and cancer recurrence. J Transl Med 16(1):8 Yi W, Li D, Guo Y, Zhang Y, Huang B, Li X, 2016, Sevoflurane inhibits the migration and invasion of glioma cells by upregulating microRNA-637. Int J Mol Med 38(6):1857–1863 Ramassone A, Pagotto S, Veronese A, Visone R, 2018, Epigenetics and MicroRNAs in Cancer. Int J Mol Sci 19(2) Liu X, Kang J, Sun S, Luo Y, Ji X, Zeng X, Zhao S, 2018, iASPP, a microRNA124 target, is aberrantly expressed in astrocytoma and regulates malignant glioma cell migration and viability. Mol Med Rep 17(1):1970–1978 Ye X, Wei W, Zhang Z, He C, Yang R, Zhang J, Wu Z, Huang Q, Jiang Q(2017, Identification ofmicroRNAs associated with glioma diagnosis and prognosis. Oncotarget 8(16):26394–26403 Ye J, Zhang Z, Wang Y, Chen C, Xu X, Yu H, Peng M, 2016, Altered hippocampal microRNA expression profiles in neonatal rats caused by sevoflurane anesthesia: MicroRNA profiling and bioinformatics target analysis. Exp Ther Med 12(3):1299–1310 Tang Y, He Y, Zhang P,Wang J, Fan C, Yang L, Xiong F, Zhang S, Gong Z, Nie S, Liao Q, Li X, Li X, Li Y, Li G, Zeng Z, Xiong W, Guo C, 2018, LncRNAs regulate the cytoskeleton and related rho/ ROCK signaling in cancer metastasis. Mol Cancer 17(1):77 Xu H, Mei XP, Xu LX, 2018, The effect of pre- and after-treatment of sevoflurane on central ischemia tolerance and the underlying mechanisms. J Dent Anesth Pain Med 18(1):1–8 Kvolik S, Glavas-Obrovac L, Bares V, Karner I, 2005, Effects of inhalation anesthetics halothane, sevoflurane, and isoflurane on human cell lines. Life Sci 77(19):2369–2383 Muller-Edenborn B, Roth-Z'graggen B, Bartnicka K, Borgeat A, Hoos A, Borsig L, Beck-Schimmer B, 2012, Volatile anesthetics reduce invasion of colorectal cancer cells through down-regulation of matrix metalloproteinase-9. Anesthesiology 117(2):293–301 Liang H, Wang HB, Liu HZ, Wen XJ, Zhou QL, Yang CX, 2013, The effects of combined treatment with sevoflurane and cisplatin on growth and invasion of human adenocarcinoma cell line A549. Biomed Pharmacother 67(6):503–509 Liang H, Gu M, Yang C, Wang H, Wen X, Zhou Q, 2012, Sevoflurane inhibits invasion and migration of lung cancer cells by inactivating the p38 MAPK signaling pathway. J Anesth 26(3):381–392 WeiGH, Zhang J, Liao DQ, Li Z,Yang J, LuoNF, GuY(2014, The common anesthetic, sevoflurane, induces apoptosis in A549 lung alveolar epithelial cells. Mol Med Rep 9(1):197–203 Hayase T, Tachibana S, Yamakage M, 2016, Effect of sevoflurane anesthesia on the comprehensive mRNA expression profile of the mouse hippocampus. Med Gas Res 6(2):70–76 An F, Gong G,Wang Y, Bian M, Yu L,Wei C, 2017, MiR-124 acts as a target for Alzheimer's disease by regulating BACE1. Oncotarget 8(69):114065–114071 Wang X, Wang X, 2006, Systematic identification of microRNA functions by combining target prediction and expression profiling. Nucleic Acids Res 34(5):1646–1652 Silber J, Lim DA, Petritsch C, Persson AI, Maunakea AK, Yu M, Vandenberg SR, Ginzinger DG, James CD, Costello JF, Bergers G, Weiss WA, Alvarez-Buylla A, Hodgson JG, 2008, miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells. BMC Med 6:14 Pierson J, Hostager B, Fan R, Vibhakar R, 2008, Regulation of cyclin dependent kinase 6 by microRNA 124 in medulloblastoma. J Neuro-Oncol 90(1):1–7 Skalsky RL, Cullen BR, 2011, Reduced expression of brainenriched microRNAs in glioblastomas permits targeted regulation of a cell death gene. PLoS One 6(9):e24248 Massague J, 2008, TGFbeta in Cancer. Cell 134(2):215–230 Cancer Genome Atlas Research Network, 2014, Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513(7517):202–209 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 947 EP 954 DI 10.1007/s12253-019-00597-1 PG 8 ER PT J AU Thorpe, D Butler, R Sultani, M Vanhoecke, B Stringer, A AF Thorpe, Daniel Butler, Ross Sultani, Masooma Vanhoecke, Barbara Stringer, Andrea TI Irinotecan-Induced Mucositis Is Associated with Goblet Cell Dysregulation and Neural Cell Damage in a Tumour Bearing DA Rat Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mucositis; Chemotherapy; Enteric nervous system; Mucus ID Mucositis; Chemotherapy; Enteric nervous system; Mucus AB Irinotecan–induced mucositis is a major oncological problem. Goblet cells secrete mucus, protecting the intestinal mucosa, with secretion altered during mucositis. The enteric nervous system is involved in regulating gut motility and secretion. The aim of this study was to determine whether enteric neural cells and goblet cells are altered following irinotecan treatment. Tumour-bearing Dark Agouti rats were administered a single dose of 175 mg/kg of irinotecan intraperitoneally and 0.01 mg/kg atropine subcutaneously. Experimental and untreated control rats were killed at times 6, 24, 48, 72, 96 and 120 h after treatment. Jejunum and colon samples were formalin fixed. Haematoxylin and eosin staining, Alcian Blue-PAS staining, and immunohistochemistry with S-100 antibody (neural cell marker) were carried out. Statistical analyses were carried out using Kruskal-Wallis test with Dunns post test, Mann Whitney U test and nonlinear regression. Total goblet cells decreased at 72 h compared with controls in the colon (p < 0.05). The percentage of cavitated goblet cells decreased compared to all other time points at 120 h in the colon. The number of S-100 positive cells in the submucosal plexus decreased in the colon (p = 0.0046) and in the myenteric plexus of the jejunum and colon (p = 0.0058 and p = 0.0022, respectively), when comparing treated with control. Enteric ganglia in the myenteric plexus of the jejunum decreased at 24 h and 96 h. Irinotecan-induced mucositis is associated with increases in mucus secretion, and enteric neural cell change. These changes may contribute to the pathophysiology of mucositis through the dysregulation of neural signalling. C1 [Thorpe, Daniel] University of South Australia, School of Pharmacy and Medical Sciences, 5001 Adelaide, Australia. [Butler, Ross] University of South Australia, School of Pharmacy and Medical Sciences, 5001 Adelaide, Australia. [Sultani, Masooma] University of South Australia, School of Medical Sciences, 5001 Adelaide, Australia. [Vanhoecke, Barbara] University of Ghent, Center for Microbiology Ecology and TechnologyGhent, Belgium. [Stringer, Andrea] University of South Australia, School of Pharmacy and Medical Sciences, 5001 Adelaide, Australia. RP Thorpe, D (reprint author), University of South Australia, School of Pharmacy and Medical Sciences, 5001 Adelaide, Australia. EM Daniel.thorpe@unisa.edu.au CR Keefe DM, Brealey J, Goland GJ, Cummins AG, 2000, Chemotherapy for cancer causes apoptosis that precedes hypoplasia in crypts of the small intestine in humans. Gut 47(5):632–637 Keefe DM, Cummins AG, Dale BM, Kotasek D, Robb TA, Sage RE, 1997, Effect of high-dose chemotherapy on intestinal permeability in humans. Clin Sci, Lond, 92(4):385–389 Pico JL, Avila-Garavito A, Naccache P, 1998)Mucositis: its occurrence, consequences, and treatment in the oncology setting. Oncologist 3(6):446–451 Keefe DM, 2004, Gastrointestinal mucositis: a new biological model. Support Care Cancer 12(1):6–9. , DOI 10.1007/ s00520-003-0550-9 Sonis ST, Costa JW Jr, Evitts SM, Lindquist LE, Nicolson M, 1992, Effect of epidermal growth factor on ulcerative mucositis in hamsters that receive cancer chemotherapy. Oral Surg Oral Med Oral Pathol 74(6):749–755 Savarese DM, Hsieh C, Stewart FM, 1997, Clinical impact of chemotherapy dose escalation in patients with hematologic malignancies and solid tumors. J Clin Oncol 15(8):2981–2995 Elting LS, Cooksley C, Chambers M, Cantor SB, Manzullo E, Rubenstein EB, 2003, The burdens of cancer therapy. Clinical and economic outcomes of chemotherapy-induced mucositis. Cancer 98(7):1531–1539. , DOI 10.1002/cncr.11671 Elting LS, Cooksley CD, Chambers MS, Garden AS, 2007, Risk, outcomes, and costs of radiation-induced oral mucositis among patients with head-and-neck malignancies. Int J Radiat Oncol Biol Phys 68(4):1110–1120. , DOI 10.1016/j.ijrobp.2007.01.053 Gibson RJ, Keefe DM, Lalla RV, Bateman E, Blijlevens N, Fijlstra M, King EE, Stringer AM, van der Velden WJ, Yazbeck R, Elad S, Bowen JM(2013, Systematic review of agents for the management of gastrointestinal mucositis in cancer patients. Support Care Cancer 21(1):313–326. , DOI 10.1007/s00520-012-1644-z Lalla RV, Bowen J, Barasch A, Elting L, Epstein J, Keefe DM, McGuire DB, Migliorati C, Nicolatou-Galitis O, Peterson DE, Raber-Durlacher JE, Sonis ST, Elad S, 2014, MASCC/ISOO clinical practice guidelines for the management of mucositis secondary to cancer therapy. Cancer 120(10):1453–1461. , DOI 10. 1002/cncr.28592 Keefe DM, Schubert MM, Elting LS, Sonis ST, Epstein JB, Raber- Durlacher JE,Migliorati CA, McGuire DB, Hutchins RD, Peterson DE, 2007, Updated clinical practice guidelines for the prevention and treatment ofmucositis. Cancer 109(5):820–831. , DOI 10.1002/cncr.22484 Sonis ST, Elting LS, Keefe D, Peterson DE, Schubert M, Hauer- JensenM, Bekele BN, Raber-Durlacher J, Donnelly JP, Rubenstein EB, 2004, Perspectives on cancer therapy-induced mucosal injury: pathogenesis, measurement, epidemiology, and consequences for patients. Cancer 100(9 Suppl):1995–2025. , DOI 10.1002/ cncr.20162 Rubenstein EB, Peterson DE, Schubert M, Keefe D, McGuire D, Epstein J, Elting LS, Fox PC, Cooksley C, Sonis ST, 2004, Clinical practice guidelines for the prevention and treatment of cancer therapy-induced oral and gastrointestinal mucositis. Cancer 100(9 Suppl):2026–2046. , DOI 10.1002/cncr.20163 Saltz LB, 2003, Understanding and managing chemotherapy-induced diarrhea. J Support Oncol 1(1):35–46; discussion 38-41, 45-36 Wadler S, Benson AB 3rd, Engelking C, Catalano R, Field M, Kornblau SM, Mitchell E, Rubin J, Trotta P, Vokes E, 1998, Recommended guidelines for the treatment of chemotherapyinduced diarrhea. J Clin Oncol 16(9):3169–3178 Al-Dasooqi N, Sonis ST, Bowen JM, Bateman E, Blijlevens N, Gibson RJ, Logan RM, Nair RG, Stringer AM, Yazbeck R, Elad S, Lalla RV, 2013, Emerging evidence on the pathobiology of mucositis. Support Care Cancer 21(7):2075–2083. , DOI 10.1007/s00520-013-1810-y de Koning BA, Sluis M, Lindenbergh-Kortleve DJ, Velcich A, Pieters R, Buller HA, Einerhand AW, Renes IB, 2007, Methotrexate-induced mucositis in mucin 2-deficient mice. J Cell Physiol 210(1):144–152. , DOI 10.1002/jcp.20822 Stringer AM, Gibson RJ, Logan RM, Bowen JM, Yeoh AS, Laurence J, Keefe DM, 2009, Irinotecan-induced mucositis is associated with changes in intestinal mucins. Cancer Chemother Pharmacol 64(1): 123–132. , DOI 10.1007/s00280-008-0855-y Stringer AM, Gibson RJ, Logan RM, Bowen JM, Yeoh AS, Hamilton J, Keefe DM, 2009, Gastrointestinal microflora and mucins may play a critical role in the development of 5-fluorouracilinduced gastrointestinal mucositis. Exp Biol Med, Maywood, 234(4):430–441. , DOI 10.3181/0810-rm-301 Saegusa Y, Ichikawa T, Iwai T, Goso Y, Okayasu I, Ikezawa T, Shikama N, Saigenji K, Ishihara K, 2008, Changes in the mucus barrier of the rat during 5-fluorouracil-induced gastrointestinal mucositis. Scand J Gastroenterol 43(1):59–65 Verburg M, Renes IB, Meijer HP, Taminiau JA, Buller HA, Einerhand AW, Dekker J, 2000, Selective sparing of goblet cells and paneth cells in the intestine of methotrexate-treated rats. Am J Physiol Gastrointest Liver Physiol 279(5):G1037–G1047 Thorpe D, Stringer A, Butler R, 2013, Chemotherapy-induced mucositis: the role of mucin secretion and regulation, and the enteric nervous system. Neurotoxicology 38:101–105. , DOI 10. 1016/j.neuro.2013.06.007 Wood JD, 2011, Enteric nervous system: the brain-in-the-gut. In: Integrated systems physiology: from molecule to function to disease, vol 26. Morgan & Claypool Life Sciences, Ohio Branka JE, Vallette G, Jarry A, Laboisse CL, 1997, Stimulation of mucin exocytosis from human epithelial cells by nitric oxide: evidence for a cGMP-dependent and a cGMP-independent pathway. Biochem J 323(Pt 2):521–524 Plaisancie P, Barcelo A, Moro F, Claustre J, Chayvialle JA, Cuber JC, 1998, Effects of neurotransmitters, gut hormones, and inflammatory mediators on mucus discharge in rat colon. Am J Phys 275(5 Pt 1):G1073–G1084 Gibson RJ, Keefe DM, Thompson FM, Clarke JM, Goland GJ, Cummins AG, 2002, Effect of interleukin-11 on ameliorating intestinal damage after methotrexate treatment of breast cancer in rats. Dig Dis Sci 47(12):2751–2757 Gibson RJ, Keefe DM, Clarke JM, Regester GO, Thompson FM, Goland GJ, Edwards BG, Cummins AG, 2002, The effect of keratinocyte growth factor on tumour growth and small intestinal mucositis after chemotherapy in the rat with breast cancer. Cancer Chemother Pharmacol 50(1):53–58. , DOI 10.1007/ s00280-002-0460-4 Bowen JM, Gibson RJ, Cummins AG, Tyskin A, KeefeDM(2007, Irinotecan changes gene expression in the small intestine of the rat with breast cancer. Cancer Chemother Pharmacol 59(3):337–348. , DOI 10.1007/s00280-006-0275-9 Gibson RJ, Bowen JM, Keefe DM, 2005, Palifermin reduces diarrhea and increases survival following irinotecan treatment in tumorbearing DA rats. Int J Cancer 116(3):464–470. , DOI 10. 1002/ijc.21082 Barcelo A, Claustre J, Moro F, Chayvialle JA, Cuber JC, Plaisancie P, 2000, Mucin secretion is modulated by luminal factors in the isolated vascularly perfused rat colon. Gut 46(2):218–224 Ilg EC, Schafer BW, Heizmann CW, 1996, Expression pattern of S100 calcium-binding proteins in human tumors. Int J Cancer 68(3):325–332. , DOI 10.1002/(sici)1097- 0215(19961104)68:3<325::aid-ijc10>3.0.co;2-7 Bowen JM, Gibson RJ, KeefeDM, Cummins AG, 2005, Cytotoxic chemotherapy upregulates pro-apoptotic Bax and Bak in the small intestine of rats and humans. Pathology 37(1):56–62 Cohen J, 2013, Statistical power analysis for the behavioral sciences. Taylor & Francis, New York Logan RM, Gibson RJ, Bowen JM, Stringer AM, Sonis ST, Keefe DM, 2008, Characterisation of mucosal changes in the alimentary tract following administration of irinotecan: implications for the pathobiology of mucositis. Cancer Chemother Pharmacol 62(1): 33–41. , DOI 10.1007/s00280-007-0570-0 Gibson RJ, Bowen JM, Inglis MR, Cummins AG, Keefe DM, 2003, Irinotecan causes severe small intestinal damage, as well as colonic damage, in the rat with implanted breast cancer. J Gastroenterol Hepatol 18(9):1095–1100 Stringer AM, Gibson RJ, Bowen JM, Logan RM, Ashton K, Yeoh AS, Al-Dasooqi N, Keefe DM(2009, Irinotecan-induced mucositis manifesting as diarrhoea corresponds with an amended intestinal flora and mucin profile. Int J Exp Pathol 90(5):489–499. https:// doi.org/10.1111/j.1365-2613.2009.00671.x Takasuna K, Hagiwara T, Hirohashi M, KatoM, Nomura M, Nagai E, Yokoi T, Kamataki T, 1996, Involvement of beta-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride, CPT-11, in rats. Cancer Res 56(16):3752–3757 Takasuna K, Hagiwara T, Hirohashi M, KatoM, Nomura M, Nagai E, Yokoi T, Kamataki T, 1998, Inhibition of intestinal microflora beta-glucuronidase modifies the distribution of the active metabolite of the antitumor agent, irinotecan hydrochloride, CPT-11, in rats. Cancer Chemother Pharmacol 42(4):280–286 Takasuna K, Hagiwara T, Watanabe K, Onose S, Yoshida S, Kumazawa E, Nagai E, Kamataki T, 2006, Optimal antidiarrhea treatment for antitumor agent irinotecan hydrochloride, CPT-11)- induced delayed diarrhea. Cancer Chemother Pharmacol 58(4): 494–503. , DOI 10.1007/s00280-006-0187-8 Cao XJ,Wang WM, 2009, Histology and mucin histochemistry of the digestive tract of yellow catfish, Pelteobagrus fulvidraco. Anat Histol Embryol 38(4):254–261. , DOI 10.1111/j.1439- 0264.2009.00932.x Truter D, Strijdom H, Everson F, Kotze SH, 2016)Mucin secreting cells in the stomach and colon are altered by combination antiretroviral treatment in an obese rat model. Acta Histochem 119:122– 128. , DOI 10.1016/j.acthis.2016.11.014 Van Herreweghen F, Van den Abbeele P, De Mulder T, De Weirdt R, Geirnaert A, Hernandez-Sanabria E, Vilchez-Vargas R, Jauregui R, Pieper DH, Belzer C, De Vos WM, Van de Wiele T, 2016, In vitro colonisation of the distal colon by Akkermansia muciniphila is largely mucin and pH dependent. Benefic Microbes 8:1–16. , DOI 10.3920/bm2016.0013 Laburthe M, Augeron C, Rouyer-Fessard C, Roumagnac I, Maoret JJ, Grasset E, Laboisse C, 1989, Functional VIP receptors in the human mucus-secreting colonic epithelial cell line CL.16E. Am J Phys 256(3 Pt 1):G443–G450 Gutierrez MM, Alluisetti GB, Olabe JA, Amorebieta VT, 2008, Nitrosation of N-methylhydroxylamine by nitroprusside. A kinetic andmechanistic study. Dalton Trans 37:5025–5030. , DOI 10.1039/b805329d Phillips TE, Stanley CM, Wilson J, 1993, The effect of 16,16- dimethyl prostaglandin E2 on proliferation of an intestinal goblet cell line and its synthesis and secretion of mucin glycoproteins. Prostaglandins Leukot Essent Fat Acids 48(6):423–428 Logan RM, Stringer AM, Bowen JM, Gibson RJ, Sonis ST, Keefe DM, 2008, Serum levels of NFkappaB and pro-inflammatory cytokines following administration of mucotoxic drugs. Cancer Biol Ther 7(7):1139–1145 Specian RD, Neutra MR, 1980, Mechanism of rapid mucus secretion in goblet cells stimulated by acetylcholine. J Cell Biol 85(3): 626–640 Specian RD, Neutra MR, 1982, Regulation of intestinal goblet cell secretion. Am J Phys 242(4):G370–G379 Lake AM, Bloch KJ, Sinclair KJ,WalkerWA, 1980, Anaphylactic release of intestinal goblet cell mucus. Immunology 39(2):173–178 Deplancke B, Gaskins HR, 2001, Microbial modulation of innate defense: goblet cells and the intestinal mucus layer. Am J Clin Nutr 73(6):1131s–1141s Furness JB, Kunze WA, Clerc N, 1999, Nutrient tasting and signaling mechanisms in the gut. II. The intestine as a sensory organ: neural, endocrine, and immune responses. Am J Phys 277(5 Pt 1): G922–G928 Furness JB, 2012, The enteric nervous system and neurogastroenterology. Nat Rev Gastroenterol Hepatol 9(5):286– 294. , DOI 10.1038/nrgastro.2012.32 McCool DJ,MarconMA, Forstner JF, Forstner GG(1990, The T84 human colonic adenocarcinoma cell line produces mucin in culture and releases it in response to various secretagogues. Biochem J 267(2):491–500 Turner JE, Stockinger B, Helmby H, 2013, IL-22 mediates goblet cell hyperplasia and worm expulsion in intestinal helminth infection. PLoS Pathog 9(10):e1003698. , DOI 10.1371/journal. ppat.1003698 Sultani M, Stringer AM, Bowen JM, Gibson RJ, 2012, Antiinflammatory cytokines: important immunoregulatory factors contributing to chemotherapy-induced gastrointestinal mucositis. Chemother Res Pract 2012:490804–490811. , DOI 10. 1155/2012/490804 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 955 EP 965 DI 10.1007/s12253-019-00644-x PG 11 ER PT J AU Bai, Y Wang, J Gao, Z Dai, E AF Bai, Yuru Wang, Jiabao Gao, Zhihua Dai, Erqing TI Identification and Verification of Two Novel Differentially Expressed Proteins from Non-neoplastic Mucosa and Colorectal Carcinoma Via iTRAQ Combined with Liquid Chromatography-Mass Spectrometry SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Proteomics; Colorectal cancer; Isobaric mass tags for relative and absolute quantification; Multidimensional liquid chromatography and tandem mass spectrometry; Differential expressed proteins ID Proteomics; Colorectal cancer; Isobaric mass tags for relative and absolute quantification; Multidimensional liquid chromatography and tandem mass spectrometry; Differential expressed proteins AB Recurrence or metastasis of colorectal cancer (CRC) is common following surgery and/or adjuvant therapy, particularly in patients with an advanced stage of the cancer. Identifying key molecular markers of CRC is beneficial for early diagnosis and early treatment, which may eventually improve the prognosis of patients with CRC. Isobaric mass tags for relative and absolute quantification (iTRAQ) in combination with multidimensional liquid chromatography and tandem mass spectrometry (LC-MS/ MS) were used to identify differentially expressed proteins between CRC tissues and paired adjacent normal mucosa. Among the 105 patients, adenocarcinoma was the most common CRC subtype, stage III was the most common Tumor-Node-Metastasis stage and high levels of Ki-67 indicated the rapid proliferation of tumor cells in the samples. The LC-MS/MS-based iTRAQ technology identified 271 differentially expressed proteins, with 130 upregulated proteins and 141 downregulated proteins. Bioinformatics analysis revealed that golgin subfamily A member 2 (GOLGA2) and heterogeneous nuclear ribonucleoprotein D0 (hnRNPD) were located in the center of the upregulated protein network, and were closely associated with the development of CRC. The upregulation of GOLGA2 and hnRNPD was further verified in human tissues using western blotting and immunohistochemistry. GOLGA2 and hnRNPD were identified as two novels differentially expressed proteins in human CRC. Furthermore, the LCMS/ MS-based iTRAQ proteomic approach is a useful tool for searching and identifying differentially expressed proteins, and may be used to provide a comprehensive understanding of the processes that mediate the development of CRC. C1 [Bai, Yuru] Characteristic Medical Center of Chinese People’s Armed Police Forces, Department of Military Medical and Health Care, 300162 Tianjin, China. [Wang, Jiabao] Characteristic Medical Center of Chinese People’s Armed Police Forces, Department of Military Medical and Health Care, 300162 Tianjin, China. [Gao, Zhihua] Characteristic Medical Center of Chinese People’s Armed Police Forces, Department of Military Medical and Health Care, 300162 Tianjin, China. [Dai, Erqing] Characteristic Medical Center of Chinese People’s Armed Police Forces, Department of Military Medical and Health Care, 300162 Tianjin, China. RP Dai, E (reprint author), Characteristic Medical Center of Chinese People’s Armed Police Forces, Department of Military Medical and Health Care, 300162 Tianjin, China. EM erqingdai@163.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386 Karanikas M, Esebidis A, 2016, Increasing incidence of colon cancer in patients <50 years old: a new entity? Ann Transl Med 4(9):164 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87–108 Barton MK, 2012, Oxaliplatin in the adjuvant treatment of colon cancer. CA Cancer J Clin 62(1):3–4 Lote H, Spiteri I, Ermini L, Vatsiou A, Roy A, McDonald A, Maka N, Balsitis M, Bose N, Simbolo M, Mafficini A, Lampis A, Hahne JC, Trevisani F, Eltahir Z, Mentrasti G, Findlay C, Kalkman EAJ, Punta M, Werner B, Lise S, Aktipis A, Maley C, Greaves M, Braconi C, White J, Fassan M, Scarpa A, Sottoriva A, Valeri N, 2017, Carbon dating cancer: defining the chronology of metastatic progression in colorectal cancer. Ann Oncol 28:1243–1249 Chaffer CL, Weinberg RA, 2011, A perspective on cancer cell metastasis. Science 331(6024):1559–1564 Tao Y, Han T, Zhang T, Sun C, 2017, Sulfatase-2 promotes the growth and metastasis of colorectal cancer by activating Akt and Erk1/2 pathways. Biomed Pharmacother 89:1370–1377 Shen T, Yang Z, Cheng X, Xiao Y, Yu K, Cai X, Xia C, Li Y, 2017, CXCL8 induces epithelial-mesenchymal transition in colon cancer cells via the PI3K/Akt/NF-kappaB signaling pathway. Oncol Rep 37:2095–2100 Huang GL et al, 2017, Oncogenic retinoic acid receptor gamma knockdown reversesmulti-drug resistance of human colorectal cancer via Wnt/beta-catenin pathway. Cell Cycle:1–8 Wang Z et al, 2017, Endothelin promotes colorectal tumorigenesis by activating YAP/TAZ. Cancer Res 77(9):2413–2423 Chen CL, Chung T, Wu CC, Ng KF, Yu JS, Tsai CH, Chang YS, Liang Y, Tsui KH, Chen YT, 2015, Comparative tissue proteomics of microdissected specimens reveals novel candidate biomarkers of bladder Cancer. Mol Cell Proteomics 14(9):2466–2478 Bouchal P, Roumeliotis T, Hrstka R, Nenutil R, Vojtesek B, Garbis SD, 2009, Biomarker discovery in low-grade breast cancer using isobaric stable isotope tags and two-dimensional liquid chromatographytandem mass spectrometry, iTRAQ-2DLC-MS/MS, based quantitative proteomic analysis. J Proteome Res 8(1):362–373 Chen RX, SongHY, Dong YY, Hu C, ZhengQD, Xue TC, Liu XH, Zhang Y, Chen J, Ren ZG, Liu YK, Cui JF, 2014, Dynamic expression patterns of differential proteins during early invasion of hepatocellular carcinoma. PLoS One 9(3):e88543 Li Y, Wang X, Ao MH, Gabrielson E, Askin F, Zhang H, Li QK, 2013, Aberrant Mucin5B expression in lung adenocarcinomas detected by iTRAQ labeling quantitative proteomics and immunohistochemistry. Clin Proteomics 10(1):15 Sun CY, Xia GW, Xu K, Ding Q, 2010, Application of iTRAQ in proteomic study of prostate cancer. Zhonghua Nan Ke Xue 16(8): 741–744 Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G, 2000, Gene ontology: tool for the unification of biology. The gene ontology consortium. Nat Genet 25(1):25–29 Zhang B, Kirov S, Snoddy J, 2005, WebGestalt: an integrated system for exploring gene sets in various biological contexts. Nucleic Acids Res 33(Web Server issue):W741–W748 da Huang W, Sherman BT, Lempicki RA, 2009, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4(1):44–57 KanehisaM, Furumichi M, TanabeM, Sato Y, Morishima K, 2017, KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res 45(D1):D353–D361 Kanehisa M, Sato Y, Kawashima M, Furumichi M, Tanabe M, 2016, KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res 44(D1):D457–D462 Orchard S, Ammari M, Aranda B, Breuza L, Briganti L, Broackes- Carter F, Campbell NH, Chavali G, Chen C, del-Toro N, Duesbury M, Dumousseau M, Galeota E, Hinz U, Iannuccelli M, Jagannathan S, Jimenez R, Khadake J, Lagreid A, Licata L, Lovering RC, Meldal B, Melidoni AN, Milagros M, Peluso D, Perfetto L, Porras P, Raghunath A, Ricard-Blum S, Roechert B, Stutz A, Tognolli M, van Roey K, Cesareni G, Hermjakob H, 2014, The MIntAct project–IntAct as a common curation platform for 11 molecular interaction databases. Nucleic Acids Res 42(Database issue): D358–D363 Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T, 2003, Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13(11):2498–2504 Dai P,Wang Q,WangW, Jing R,WangW,Wang F, Azadzoi K, Yang JH, Yan Z, 2016, Unraveling molecular differences of gastric Cancer by label-free quantitative proteomics analysis. Int J Mol Sci 17(1) Witkos TM, LoweM(2017, Recognition and tethering of transport vesicles at the Golgi apparatus. Curr Opin Cell Biol 47:16–23 Potelle S, Klein A, Foulquier F, 2015, Golgi post-translational modifications and associated diseases. J Inherit Metab Dis 38(4):741–751 Chang SH, Hong SH, Jiang HL, Minai-Tehrani A, Yu KN, Lee JH, Kim JE, Shin JY, Kang B, Park S, Han K, Chae C, ChoMH, 2012, GOLGA2/GM130, cis-Golgi matrix protein, is a novel target of anticancer gene therapy. Mol Ther 20(11):2052–2063 Nakamura N, 2010, Emerging new roles of GM130, a cis-Golgi matrix protein, in higher order cell functions. J Pharmacol Sci 112(3):255–264 Medici D, Hay ED, Olsen BR, 2008, Snail and slug promote epithelial-mesenchymal transition through beta-catenin-T-cell factor-4-dependent expression of transforming growth factor-beta3. Mol Biol Cell 19(11):4875–4887 Tse JC, Kalluri R, 2007, Mechanisms of metastasis: epithelial-tomesenchymal transition and contribution of tumor microenvironment. J Cell Biochem 101(4):816–829 Kalluri R, Weinberg RA, 2009, The basics of epithelialmesenchymal transition. J Clin Invest 119(6):1420–1428 Godde NJ, Galea RC, Elsum IA, Humbert PO, 2010, Cell polarity in motion: redefining mammary tissue organization through EMT and cell polarity transitions. J Mammary Gland Biol Neoplasia 15(2):149–168 Lee SC, Kim OH, Lee SK, Kim SJ, 2015, IWR-1 inhibits epithelial-mesenchymal transition of colorectal cancer cells through suppressingWnt/beta-catenin signaling as well as survivin expression. Oncotarget 6(29):27146–27159 Nieto MA, 2013, Epithelial plasticity: a common theme in embryonic and cancer cells. Science 342(6159):1234850 Roy E, Bruyere J, Flamant P, Bigou S, Ausseil J, Vitry S, Heard JM, 2012, GM130 gain-of-function induces cell pathology in a model of lysosomal storage disease. Hum Mol Genet 21(7):1481–1495 Zhao J, Yang C, Guo S, Wu Y, 2015, GM130 regulates epithelialto- mesenchymal transition and invasion of gastric cancer cells via snail. Int J Clin Exp Pathol 8(9):10784–10791 Han N, Li W, Zhang M, 2013, The function of the RNA-binding protein hnRNP in cancer metastasis. J Cancer Res Ther 9(Suppl): S129–S134 Gouble A, Grazide S, Meggetto F, Mercier P, Delsol G, Morello D, 2002, A new player in oncogenesis: AUF1/hnRNPD overexpression leads to tumorigenesis in transgenic mice. Cancer Res 62(5): 1489–1495 Geng Y, Zhang L, Xu M, Sheng W, Dong A, Cao J, Cao J, 2015, The expression and significance of hnRNPD in esophageal squamous cell carcinoma cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 31(12):1659–1663 Kumar M, Matta A, Masui O, Srivastava G, Kaur J, Thakar A, Shukla NK, RoyChoudhury A, Sharma M, Walfish PG, Michael Siu KW, Chauhan SS, Ralhan R, 2015, Nuclear heterogeneous nuclear ribonucleoprotein D is associated with poor prognosis and interactome analysis reveals its novel binding partners in oral cancer. J Transl Med 13:285 Trojanowicz B, Brodauf L, Sekulla C, Lorenz K, Finke R, Dralle H, Hoang-Vu C, 2009, The role of AUF1 in thyroid carcinoma progression. Endocr Relat Cancer 16(3):857–871 Eder S, Arndt A, Lamkowski A, DaskalakiW, Rump A, Priller M, Genze F, Wardelmann E, Port M, Steinestel K, 2017, Baseline MAPK signaling activity confers intrinsic radioresistance to KRAS-mutant colorectal carcinoma cells by rapid upregulation of heterogeneous nuclear ribonucleoprotein K, hnRNP K). Cancer Lett 385:160–167 Zhang Z, Zhou C, Chang Y, Zhang Z, Hu Y, Zhang F, Lu Y, Zheng L, Zhang W, Li X, Li X, 2016, Long non-coding RNA CASC11 interacts with hnRNP-K and activates the WNT/beta-catenin pathway to promote growth and metastasis in colorectal cancer. Cancer Lett 376(1):62–73 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 967 EP 976 DI 10.1007/s12253-019-00651-y PG 10 ER PT J AU Kim, U Kim, CY Lee, MJ Oh, H Ryu, B Kim, J Park, JH AF Kim, Ukjin Kim, C-Yoon Lee, Min Ji Oh, Hanseul Ryu, Bokyeong Kim, Jin Park, Jae-Hak TI Phloretin Inhibits the Human Prostate Cancer Cells Through the Generation of Reactive Oxygen Species SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Phloretin; Prostate cancer; ROS; Redox homeostasis; Wnt/β-catenin signaling ID Phloretin; Prostate cancer; ROS; Redox homeostasis; Wnt/β-catenin signaling AB Phloretin is a flavonoid with known anticancer activities. However, we do not fully understand how phloretin mitigates prostate cancer on the molecular level. In the present study, we examined changes in proliferation, colony formation, and migration after phloretin treatment in human prostate cancer cells PC3 and DU145. We measured reactive oxygen species (ROS) and gene expression. Phloretin increased ROS and suppressed cell proliferation, migration, and colony formation in both cell lines. Additionally, phloretin treatment increased oxidative stress, as demonstrated through lower antioxidant enzymes (catalase, SOD2, Gpx1, Gpx3). In addition, their regulator CISD2 decreased in expression. We also found that increased ROS significantly downregulated multiple components of the Wnt/β-catenin signaling pathway (β-catenin, TCF4, FoxA2, c-Myc) and Twist1. Thus, anticancer activity of phloretin against human prostate cancer cells occurs through generating ROS to influence Wnt/β- catenin signaling. The results of this study suggest that phloretin has a therapeutic effect on prostate cancer in vitro, inhibiting the proliferation and migration of cancer cell lines PC3 and DU145. The mechanism of phloretin appears to be increasing ROS production. We thus recommend phloretin as a promising anticancer therapeutic agent. C1 [Kim, Ukjin] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. [Kim, C-Yoon] Konkuk University, School of Medicine, Department of Stem Cell Biology, 05029 Seoul, South Korea. [Lee, Min Ji] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. [Oh, Hanseul] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. [Ryu, Bokyeong] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. [Kim, Jin] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. [Park, Jae-Hak] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. RP Park, JH (reprint author), Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. EM pjhak@snu.ac.kr CR Kohler BA, Sherman RL, Howlader N, Jemal A, Ryerson AB, Henry KA, Boscoe FP, Cronin KA, Lake A, Noone A-M, 2015, Annual report to the nation on the status of cancer, 1975-2011, featuring incidence of breast cancer subtypes by race/ethnicity, poverty, and state. J Natl Cancer Inst 107(6):djv048 Center MM, Jemal A, Lortet-Tieulent J,Ward E, Ferlay J, Brawley O, Bray F, 2012, International variation in prostate cancer incidence and mortality rates. Eur Urol 61(6):1079–1092 Khandrika L, Kumar B, Koul S, Maroni P, Koul HK, 2009, Oxidative stress in prostate cancer. Cancer Lett 282(2):125–136 Cairns RA, Harris IS, Mak TW, 2011, Regulation of cancer cell metabolism. Nat Rev Cancer 11(2):85–95 Korswagen HC, 2006, Regulation of the Wnt/β-catenin pathway by redox signaling. Dev Cell 10(6):687–688 Yu X, Wang Y, Jiang M, Bierie B, Roy-Burman P, Shen MM, Taketo MM, Wills M, Matusik RJ, 2009, Activation of β-catenin in mouse prostate causes HGPIN and continuous prostate growth after castration. Prostate 69(3):249–262 Miller JR, 2001, The wnts. Genome Biol 3(1):reviews3001. 3001 Chesire DR, Ewing CM, Gage WR, Isaacs WB, 2002, In vitro evidence for complex modes of nuclear β-catenin signaling during prostate growth and tumorigenesis. Oncogene 21(17):2679–2694 Shin SY, Kim CG, Jho E-H, Rho M-S, Kim YS, Kim Y-H, Lee YH, 2004, Hydrogen peroxide negatively modulates Wnt signaling through downregulation of β-catenin. Cancer Lett 212(2):225–231 Boyer J, Liu RH, 2004, Apple phytochemicals and their health benefits. Nutr J 3(1):5 DeviMA,DasN, 1993, In vitro effects of natural plant polyphenols on the proliferation of normal and abnormal human lymphocytes and their secretions of interleukin-2. Cancer Lett 69(3):191–196 Zhu S-P, Liu G,Wu X-T, Chen F-X, Liu J-Q, Zhou Z-H, Zhang J-F, Fei S-J, 2013, The effect of phloretin on human γδ T cells killing colon cancer SW-1116 cells. Int Immunopharmacol 15(1):6–14 Nelson J, Falk R, 1992, The efficacy of phloridzin and phloretin on tumor cell growth. Anticancer Res 13(6A):2287–2292 Kypta RM, Waxman J, 2012, Wnt/β-catenin signalling in prostate cancer. Nature Reviews Urology 9(8):418–428 Prasannaraj G, Sahi SV, Benelli G, Venkatachalam P, 2017, Coating with active Phytomolecules enhances anticancer activity of bio-engineered ag Nanocomplex. J Clust Sci 28(4):2349–2367 Priyadharshini RI, Prasannaraj G, Geetha N, Venkatachalam P, 2014, Microwave-mediated extracellular synthesis of metallic silver and zinc oxide nanoparticles using macro-algae, Gracilaria edulis, extracts and its anticancer activity against human PC3 cell lines. Appl Biochem Biotechnol 174(8):2777–2790 Liu Y, Fan C, Pu L,Wei C, Jin H, Teng Y, Zhao M, Yu ACH, Jiang F, Shu J, 2016, Phloretin induces cell cycle arrest and apoptosis of human glioblastoma cells through the generation of reactive oxygen species. J Neuro-Oncol 128(2):217–223 Meyer F,Galan P, Douville P, Bairati I, Kegle P, Bertrais S, Estaquio C, Hercberg S, 2005, Antioxidant vitamin and mineral supplementation and prostate cancer prevention in the SU. VI.MAX trial. Int J Cancer 116(2):182–186 Kim J, Kim J, Bae J-S, 2016, ROS homeostasis and metabolism: a critical liaison for cancer therapy. Exp Mol Med 48(11):e269 Chang SN, Lee JM, Oh H, Park JH, 2016, Glutathione peroxidase 3 inhibits prostate tumorigenesis in TRAMP mice. Prostate 76(15): 1387–1398 Lill R, 2009, Function and biogenesis of iron–Sulphur proteins. Nature 460(7257):831–838 Chen Y-F, Kao C-H, Chen Y-T,Wang C-H,Wu C-Y, Tsai C-Y, Liu F-C, Yang C-W,Wei Y-H, Hsu M-T, 2009, Cisd2 deficiency drives premature aging and causesmitochondria-mediated defects in mice. Genes Dev 23(10):1183–1194 Chang NC, Nguyen M, Bourdon J, Risse P-A, Martin J, Danialou G, Rizzuto R, Petrof BJ, Shore GC, 2012, Bcl-2-associated autophagy regulator Naf-1 required for maintenance of skeletal muscle. Hum Mol Genet 21(10):2277–2287 Li S-M, Chen C-H, Chen Y-W, Yen Y-C, Fang W-T, Tsai F-Y, Chang J-L, Shen Y-Y, Huang S-F, Chuu C-P, 2017, Upregulation of CISD2 augments ROS homeostasis and contributes to tumorigenesis and poor prognosis of lung adenocarcinoma. Sci Rep 7(1): 11893 Ouyang X, DeWeese TL, Nelson WG, Abate-Shen C, 2005, Lossof- function of Nkx3. 1 promotes increased oxidative damage in prostate carcinogenesis. Cancer Res 65(15):6773–6779 Prendergast GC, 1999, Mechanisms of apoptosis by c-Myc. Oncogene 18(19):2967–2987 Cassinelli G, Supino R, Zuco V, Lanzi C, Scovassi AI, Semple SC, Zunino F, 2004, Role of c-myc protein in hormone refractory prostate carcinoma: cellular response to paclitaxel. Biochem Pharmacol 68(5):923–931 Li Q, Dang CV, 1999, C-Myc overexpression uncouples DNA replication from mitosis. Mol Cell Biol 19(8):5339–5351 Gajula RP, Chettiar ST,Williams RD, Thiyagarajan S, Kato Y, Aziz K, Wang R, Gandhi N, Wild AT, Vesuna F, 2013, The twist box domain is required for Twist1-induced prostate cancer metastasis. Molecular Cancer Research:molcanres. 0218.2013 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 977 EP 984 DI 10.1007/s12253-019-00643-y PG 8 ER PT J AU Oguh-Olayinka, L Agarwal, V Ranatunge, D Campbell, A Laufer, S Cawkwell, Ly Lind, JM AF Oguh-Olayinka, Lily Agarwal, Vijay Ranatunge, Dulani Campbell, Anne Laufer, Stefan Cawkwell, Lynn Lind, J Michael TI The Investigation of Lipoxygenases as Therapeutic Targets in Malignant Pleural Mesothelioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Arachidonic acid; Cyclooxygenase; Immunohistochemistry; Lipoxygenase; Mesothelioma ID Arachidonic acid; Cyclooxygenase; Immunohistochemistry; Lipoxygenase; Mesothelioma AB Advanced malignant pleural mesothelioma (MPM) has an extremely poor prognosis with limited chemotherapy options, therefore the identification of new therapeutic targets would aid in disease management. Arachidonic acid is metabolised by cyclooxygenase and lipoxygenase enzymes. The lipoxygenase isoenzymes 5-LOX and 12-LOX have been implicated in carcinogenesis. We aimed to examine 5-LOX and 12-LOX protein expression in a large retrospective series of mesothelioma samples. Further to this, the in vitro cytotoxic effects of lipoxygenase pathway inhibitors were investigated in mesothelioma cells. Archival samples from 83 patients with MPM were examined by immunohistochemistry for expression of the 5-LOX and 12-LOX proteins. The MTS assay was used to assess cell viability following 72 h treatment with the lipoxygenase pathway inhibitors baicalein, licofelone, MK-886 and zileuton in the MPM cell lines NCI-H2052, NCI-H2452 and MSTO-211H. Positive 12-LOX protein expression was recorded in 69/83 (83%) and positive 5-LOX expression was observed in 56/77 (73%) of MPM tissue samples. Co-expression of 5-LOX with 12-LOX was seen in 46/78 (58%) of MPM samples. Positive expression of 5-LOX, 12- LOX and COX-2 proteins was identified in the NCI-H2052, NCI-H2452 and MSTO-211H MPM cell lines. Baicalein (12-LOX and 15-LOX inhibitor) was effective in 3/3 MPM cell lines at low concentrations with an IC50 range of 9.6 μM to 20.7 μM. We have demonstrated that the 5-LOX and 12-LOX proteins are expressed in a significant proportion of MPM samples (73% and 83% respectively) and may represent novel therapeutic targets in this disease. We have demonstrated that the inhibition of the LOX pathway using baicalein may be effective as a novel treatment for MPM, however further human pharmacokinetic studies are required in order to establish whether the concentration used in vitro is clinically achievable. C1 [Oguh-Olayinka, Lily] Castle Hill Hospital, Hull York Medical School, Research Laboratories, Daisy Building, HU16 5JQ Hull, UK. [Agarwal, Vijay] Castle Hill Hospital, Hull York Medical School, Research Laboratories, Daisy Building, HU16 5JQ Hull, UK. [Ranatunge, Dulani] Castle Hill Hospital, Hull York Medical School, Research Laboratories, Daisy Building, HU16 5JQ Hull, UK. [Campbell, Anne] Hull and East Yorkshire NHS Trust, Histopathology DepartmentHull, UK. [Laufer, Stefan] Eberhard Karls University, Department of Pharmaceutical ChemistryTubingen, Germany. [Cawkwell, Lynn] Castle Hill Hospital, Hull York Medical School, Research Laboratories, Daisy Building, HU16 5JQ Hull, UK. [Lind, J Michael] Castle Hill Hospital, Hull York Medical School, Research Laboratories, Daisy Building, HU16 5JQ Hull, UK. RP Oguh-Olayinka, L (reprint author), Castle Hill Hospital, Hull York Medical School, Research Laboratories, HU16 5JQ Hull, UK. EM L.Cawkwell@hull.ac.uk CR Lanphear BP, Buncher CR, 1992, Latent period for malignant mesothelioma of occupational origin. J Occup Med 34:718–721 Carbone M, Kratzke RA, Testa JR, 2002, The pathogenesis of mesothelioma. Semin Oncol 29:2–17 Ceresoli GL, Locati LD, Ferreri AJ et al, 2001, Therapeutic outcome according to histologic subtype in 121 patients with malignant pleural mesothelioma. Lung Cancer 34:279–287 O’Kane SL, Cawkwell L, Campbell A, Lind MJ, 2005, Cyclooxygenase-2 expression predicts survival in malignant pleural mesothelioma. Eur J Cancer 41:1645–1648. , DOI 10. 1016/j.ejca.2005.04.019 Vogelzang NJ, Rusthoven JJ, Symanowski J, Denham C, Kaukel E, Ruffie P, Gatzemeier U, BoyerM, Emri S, Manegold C, Niyikiza C, Paoletti P, 2003, Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma. J Clin Oncol 21:2636–2644. , DOI 10. 1200/JCO.2003.11.136 Tsao AS,Wistuba I, Roth JA, Kindler HL, 2009)Malignant pleural mesothelioma. J Clin Oncol 27:2081–2090. , DOI 10. 1200/JCO.2008.19.8523 Ceresoli GL, Zucali PA, Gianoncelli L, Lorenzi E, Santoro A, 2010, Second-line treatment for malignant pleural mesothelioma. Cancer Treat Rev 36:24–32. , DOI 10.1016/j.ctrv. 2009.09.003 Grosso F, Scagliotti GV, 2012, Systemic treatment of malignant pleural mesothelioma. Future Oncol 8:293–305. , DOI 10. 2217/fon.12.14 Claria J, Romano M, 2005, Pharmacological intervention of cyclooxygenase-2 and 5-lipoxygenase pathways. Impact on inflammation and cancer. Curr Pharm Des 11:3431–3447. , DOI 2174/138161205774370753 10. Greenhough A, Smartt HJM, Moore AE, Roberts HR, Williams AC, Paraskeva C, Kaidi A, 2009, The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis 30:377–386. , DOI 10. 1093/carcin/bgp014 Salvado MD, Alfranca A, Haeggstrom JZ, Redondo JM, 2012, Prostanoids in tumor angiogenesis: therapeutic intervention beyond COX-2. Trends Mol Med 18:233–243. , DOI 10.1016/j. molmed.2012.02.002 Goossens L, Pommery N, Henichart JP, 2007, COX-2/5-LOX dual acting anti-inflammatory drugs in cancer chemotherapy. Curr Top Med Chem 7:283–296 Schneider C, Pozzi A, 2011, Cyclooxygenases and lipoxygenases in cancer. Cancer Metastasis Rev 30:277–294. , DOI 10. 1007/s10555-011-9310-3 Fu S,WuY, ZhangY, QiaoMM, ChenY, 2004, Anti-cancer effects of COX-2 inhibitors and their correlation with angiogenesis and invasion in gastric cancer. World J Gastroenterol 10:1971–1974 Cerella C, Sobolewski C, Chateauvieux S, Henry E, Schnekenburger M, Ghelfi J, Dicato M, Diederich M, 2011, COX-2 inhibitors block chemotherapeutic agent-induced apoptosis prior to commitment in hematopoietic cancer cells. Biochem Pharmacol 82:1277–1290. , DOI 10.1016/j.bcp.2011.06.028 Chan AT, Giovannucci EL, Meyerhardt JA, Schernhammer ES, Curhan GC, Fuchs CS, 2005, Long-term use of aspirin and nonsteroidal anti-inflammatory drugs and risk of colorectal cancer. JAMA 294:914–923. , DOI 10.1001/jama.294.8.914 O’Kane SL, Eagle GL, Greenman J, Lind MJ, Cawkwell L, 2010, COX-2 specific inhibitors enhance the cytotoxic effects of pemetrexed in mesothelioma cell lines. Lung Cancer 67:160–165. , DOI 10.1016/j.lungcan.2009.04.008 Orlando UD, Garona J, Ripoll GV, Maloberti PM, Solano AR, Avagnina A, Gomez DE, Alonso DF, Podesta EJ, 2012, The functional interaction between acyl-coa synthetase 4, 5-lipooxygenase and cyclooxygenase-2 controls tumor growth: a novel therapeutic target. PLoS One 7:e40794. , DOI 10.1371/journal.pone. 0040794 Moore GY, Pidgeon GP, 2017, Cross-talk between cancer cells and the tumour microenvironment: the role of the 5-lipoxygenase pathway. Int J Mol Sci 18 Wang D, DuBois R, 2010, Eicosanoids and cancer. Nat Rev Cancer 10:181–193. , DOI 10.1038/nrc2809 Tong W-G, Ding X-Z, Talamonti MS, Bell RH, Adrian TE, 2005, LTB4 stimulates growth of human pancreatic cancer cells via MAPK and PI-3 kinase pathways. Biochem Biophys Res Commun 335:949–956. , DOI 10.1016/j.bbrc.2005.07.166 Ihara A, Wada K, Yoneda M, Fujisawa N, Takahashi H, Nakajima A, 2007, Blockade of leukotriene B4 signaling pathway induces apoptosis and suppresses cell proliferation in colon cancer. J Pharmacol Sci 103:24–32. , DOI 10.1254/jphs.FP0060651 Kandouz M, Nie D, Pidgeon GP, Krishnamoorthy S, Maddipati KR, Honn KV, 2003, Platelet-type 12-lipoxygenase activates NF-kappaB in prostate cancer cells. Prostaglandins Other Lipid Mediat 71:189–204 Honn KV, Timar J, Rozhin J, Bazaz R, Sameni M, Ziegler G, Sloane BF, 1994, A lipoxygenase metabolite, 12-(S)-HETE, stimulates protein kinase C-mediated release of cathepsin B from malignant cells. Exp Cell Res 214:120–130. , DOI 10.1006/ excr.1994.1240 Hong SH, Avis I, Vos MD, Martinez A, Treston AM, Mulshine JL, 1999, Relationship of arachidonic acid metabolizing enzyme expression in epithelial cancer cell lines to the growth effect of selective biochemical inhibitors. Cancer Res 59:2223–2228 Hennig R, Ding X-Z, Tong W-G, Schneider MB, Standop J, Friess H, Buchler MW, Pour PM, Adrian TE, 2002, 5-lipoxygenase and leukotriene B(4, receptor are expressed in human pancreatic cancers but not in pancreatic ducts in normal tissue. Am J Pathol 161: 421–428. , DOI 10.1016/S0002-9440(10)64198-3 Gupta S, Srivastava M, Ahmad N, Sakamoto K, Bostwick DG, Mukhtar H, 2001, Lipoxygenase-5 is overexpressed in prostate adenocarcinoma. Cancer 91:737–743. , DOI 10.1002/ 1097-0142(20010215)91:4<737::AID-CNCR1059>3.0.CO;2-F Yoshimura R,Matsuyama M, TsuchidaKet al, 2003, Expression of lipoxygenase in human bladder carcinoma and growth inhibition by its inhibitors. J Urol 170:1994–1999. , DOI 10.1097/01.ju. 0000080296.54262.c8 Jiang WG, Douglas-Jones A, Mansel RE, 2003, Levels of expression of lipoxygenases and cyclooxygenase-2 in human breast cancer. Prostaglandins Leukot Essent Fat Acids 69:275–281. https:// doi.org/10.1016/S0952-3278(03)00110-8 Ohd JF, Nielsen CK, Campbell J, Landberg G, Lofberg H, Sjolander A, 2003, Expression of the leukotriene D4 receptor CysLT1, COX-2, and other cell survival factors in colorectal adenocarcinomas. Gastroenterology 124:57–70. , DOI 10. 1053/gast.2003.50011 Winer I, Normolle DP, Shureiqi I, Sondak VK, Johnson T, Su L, Brenner DE, 2002, Expression of 12-lipoxygenase as a biomarker for melanoma carcinogenesis. Melanoma Res 12:429–434. https:// doi.org/10.1097/00008390-200209000-00003 Chen X,Wang S,Wu N, Sood S,Wang P, Jin Z, Beer DG, Giordano TJ, Lin Y, ShihWC, Lubet RA, Yang CS, 2004, Overexpression of 5-lipoxygenase in rat and human esophageal adenocarcinoma and inhibitory effects of Zileuton and celecoxib on carcinogenesis. Clin Cancer Res 10:6703–6709. , DOI 10.1158/1078-0432. CCR-04-0838 Hoque A, Lippman SM, Wu T-TT et al, 2005, Increased 5- lipoxygenase expression and induction of apoptosis by its inhibitors in esophageal cancer: a potential target for prevention. Carcinogenesis 26:785–791. , DOI 10.1093/carcin/bgi026 Li N, Sood S,Wang S, Fang M,Wang P, Sun Z, Yang CS, Chen X, 2005, Overexpression of 5-lipoxygenase and cyclooxygenase 2 in hamster and human oral cancer and chemopreventive effects of zileuton and celecoxib. Clin Cancer Res 11:2089–2096. https:// doi.org/10.1158/1078-0432.CCR-04-1684 Romano M, Catalano A, Nutini M et al, 2001, 5-lipoxygenase regulates malignant mesothelial cell survival: involvement of vascular endothelial growth factor. FASEB J 15:2326–2336. https:// doi.org/10.1096/fj.01-0150com Cawkwell L, Gray S, Murgatroyd H, Sutherland F, Haine L, Longfellow M, O'Loughlin S, Cross D, Kronborg O, Fenger C, Mapstone N, Dixon M, Quirke P, 1999, Choice of management strategy for colorectal cancer based on a diagnostic immunohistochemical test for defective mismatch repair. Gut 45:409–415. , DOI 10.1136/gut.45.3.409 Steinhilber D, Hofmann B, 2014, Recent advances in the search for novel 5-lipoxygenase inhibitors. Basic Clin Pharmacol Toxicol 114: 70–77. , DOI 10.1111/bcpt.12114 Evans JF, Ferguson AD, Mosley RT, Hutchinson JH, 2008)What’s all the FLAP about?: 5-lipoxygenase-activating protein inhibitors for inflammatory diseases. Trends Pharmacol Sci 29:72–78. https:// doi.org/10.1016/j.tips.2007.11.006 Deschamps JD, Kenyon VA, Holman TR, 2006, Baicalein is a potent in vitro inhibitor against both reticulocyte 15-human and platelet 12-human lipoxygenases. Bioorg Med Chem 14:4295– 4301. , DOI 10.1016/j.bmc.2006.01.057 DeWitt DL, 1999, Cox-2-selective inhibitors: the new super aspirins. Mol Pharmacol 55:625–631 AlbrechtW, Unger A, Nussler AK, Laufer S, 2008, In vitro metabolism of 2-[6-(4-chlorophenyl)-2,2-dimethyl-7-phenyl-2,3- dihydro-1H-pyrrolizin-5-yl] acetic acid, licofelone, ML3000), an inhibitor of cyclooxygenase-1 and -2 and 5-lipoxygenase. Drug Metab Dispos 36:894–903. , DOI 10.1124/dmd.108. 020347 Chou T-C, Talalay P, 1984, Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzym Regul 22:27–55. , DOI 10.1016/ 0065-2571(84)90007-4 Edwards JG, Faux SP, Plummer SM et al, 2002, Cyclooxygenase-2 expression is a novel prognostic factor in malignant mesothelioma n. Clin Cancer Res 8:1857–1862 Baldi A, 2004, Prognostic significance of cyclooxygenase-2, COX-2, and expression of cell cycle inhibitors p21 and p27 in human pleural malignant mesothelioma. Thorax 59:428–433. , DOI 10.1136/thx.2003.008912 Mineo TC, Ambrogi V, Cufari ME, Pompeo E, 2010, May cyclooxygenase-2, COX-2), p21 and p27 expression affect prognosis and therapeutic strategy of patients with malignant pleural mesothelioma? Eur J Cardiothorac Surg 38:245–252. https://doi. org/10.1016/j.ejcts.2010.02.012 Nuvoli B, Galati R, 2013, Cyclooxygenase-2, epidermal growth factor receptor, and aromatase signaling in inflammation and mesothelioma. Mol Cancer Ther 12:844–852. , DOI 10.1158/ 1535-7163.MCT-12-1103 Lee HZ, Leung HWC, Lai MY, Wu CH, 2005, Baicalein induced cell cycle arrest and apoptosis in human lung squamous carcinoma CH27 cells. Anticancer Res 25:959–964 Chao J-I, Su W-C, Liu H-F, 2007, Baicalein induces cancer cell death and proliferation retardation by the inhibition of CDC2 kinase and survivin associated with opposite role of p38 mitogen-activated protein kinase and AKT. Mol Cancer Ther 6:3039–3048. https:// doi.org/10.1158/1535-7163.MCT-07-0281 Takahashi H, Chen MC, Pham H, Angst E, King JC, Park J, Brovman EY, Ishiguro H, Harris DM, Reber HA, Hines OJ, Gukovskaya AS, Go VLW, Eibl G, 2011, Baicalein, a component of Scutellaria baicalensis, induces apoptosis by Mcl-1 down-regulation in human pancreatic cancer cells. Biochim Biophys Acta 1813:1465–1474. , DOI 10.1016/j.bbamcr.2011.05.003 Chen K, Zhang S, Ji Y, Li J, An P, Ren H, Liang R, Yang J, Li Z, 2013, Baicalein inhibits the invasion and metastatic capabilities of hepatocellular carcinoma cells via down-regulation of the ERK pathway. PLoS One 8:e72927. , DOI 10.1371/journal. pone.0072927 Tavolari S, Bonafe M, Marini M, Ferreri C, Bartolini G, Brighenti E,Manara S, Tomasi V, Laufer S, Guarnieri T, 2008, Licofelone, a dual COX/5-LOX inhibitor, induces apoptosis in HCA-7 colon cancer cells through themitochondrial pathway independently from its ability to affect the arachidonic acid cascade. Carcinogenesis 29: 371–380. , DOI 10.1093/carcin/bgm265 Fischer AS, Metzner J, Steinbrink SD, Ulrich S, Angioni C, Geisslinger G, Steinhilber D, Maier TJ, 2010, 5-lipoxygenase inhibitors induce potent anti-proliferative and cytotoxic effects in human tumour cells independently of suppression of 5-lipoxygenase activity. Br J Pharmacol 161:936–949. https://doi. org/10.1111/j.1476-5381.2010.00915.x Aryal P, Kim K, Park P-H, Ham S, Cho J, Song K, 2014, Baicalein induces autophagic c e l l death through AMPK/ULK1 activation and downregulation of mTORC1 complex components in human cancer cells. FEBS J 281: 4644–4658. , DOI 10.1111/febs.12969 Melstrom LG, Bentrem DJ, Salabat MR, Kennedy TJ, Ding XZ, Strouch M, Rao SM, Witt RC, Ternent CA, Talamonti MS, Bell RH, Adrian TA, 2008, Overexpression of 5-lipoxygenase in colon polyps and cancer and the effect of 5-LOX inhibitors in vitro and in a murinemodel. Clin Cancer Res 14:6525–6530. , DOI 10. 1158/1078-0432.CCR-07-4631 Timar J, Raso E, Dome B, Li L, Grignon D, Nie D, Honn KV, Hagmann W, 2000, Expression, subcellular localization and putative function of platelet-type 12-lipoxygenase in human prostate cancer cell lines of different metastatic potential. Int J Cancer 87:37–43. , DOI 10.1002/1097-0215(20000701)87:1<37:: AID-IJC6>3.0.CO;2-L Li M, Shi A, Pang H, Xue W, Li Y, Cao G, Yan B, Dong F, Li K, Xiao W, He G, Du G, Hu X, 2014, Safety, tolerability, and pharmacokinetics of a single ascending dose of baicalein chewable tablets in healthy subjects. J Ethnopharmacol 156:210–215. https://doi. org/10.1016/j.jep.2014.08.031 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 985 EP 995 DI 10.1007/s12253-019-00652-x PG 11 ER PT J AU Kajary, K Lengyel, Zs Tokes, AM Kulka, J Dank, M Tokes, T AF Kajary, Kornelia Lengyel, Zsolt Tokes, Anna-Maria Kulka, Janina Dank, Magdolna Tokes, Timea TI Dynamic FDG-PET/CT in the Initial Staging of Primary Breast Cancer: Clinicopathological Correlations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Kinetics; PET-CT; Tumor-infiltrating lymphocytes ID Breast cancer; Kinetics; PET-CT; Tumor-infiltrating lymphocytes AB Our aim was to evaluate correlation between clinicopathological features (clinical T and clinical N stages; histological type; nuclear grade; hormone-receptor and HER2 status, proliferation activity and tumor subtypes) of breast cancer and kinetic parameters measured by staging dynamic FDG-PET/CT examinations. Following ethical approval and patients’ informed consent we included 34 patients with 35 primary breast cancers in our prospective study. We performed dynamic PET imaging, and assessed plasma activity noninvasively. To delineate primary tumors we applied a frame-by-frame semi-automatic software-based correction of motion artefacts. FDG two-compartment kinetic modelling was applied to assess K1, k2, k3 rate coefficients and to calculate Ki (tracer flux constant) and MRFDG (FDG metabolic rate). We found that k3, Ki and MRFDG were significantly higher in higher grade (p = 0.0246, 0.0089 and 0.0076, respectively), progesterone-receptor negative (p = 0.0344, 0.0217 and 0.0132) and highly-proliferating (p = 0.0414, 0.0193 and 0.0271) tumors as well as in triple-negative and hormone-receptor negative/HER2-positive subtypes (p = 0.0310, 0.0280 and 0.0186). Ki and MRFDG were significantly higher in estrogen receptor negative tumors (p = 0.0300 and 0.0247, respectively). Ki was significantly higher in node-positive than in node-negative disease (p = 0.0315). None of the assessed FDG-kinetic parameters showed significant correlation with stromal TIL. In conclusion, we confirmed a significant relationship between kinetic parameters measured by dynamic PET and the routinely assessed clinicopathological factors of breast cancer: high-grade, hormone-receptor negative tumors with high proliferation rate are characterized by higher cellular FDG-uptake and FDG-phosphorylation rate. Furthermore, we found that kinetic parameters based on the dynamic examinations are probably not influenced by stromal TIL infiltration. C1 [Kajary, Kornelia] Pozitron Diagnosztika Kft, Hunyadi J. Str. 9, H-1117 Budapest, Hungary. [Lengyel, Zsolt] Pozitron Diagnosztika Kft, Hunyadi J. Str. 9, H-1117 Budapest, Hungary. [Tokes, Anna-Maria] Semmelweis University, 2nd Department of Pathology, Ulloi str. 93, H-1091 Budapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi str. 93, H-1091 Budapest, Hungary. [Dank, Magdolna] Semmelweis University, Department of Oncology, Tomo utca 25-29, H-1083 Budapest, Hungary. [Tokes, Timea] Semmelweis University, Department of Oncology, Tomo utca 25-29, H-1083 Budapest, Hungary. RP Tokes, T (reprint author), Semmelweis University, Department of Oncology, H-1083 Budapest, Hungary. EM tokes.timea@med.semmelweis-univ.hu;timi.tokes@gmail.com CR International Atomic Energy Agency, 2013, Standard operation procedures for PET/CT: a practical approach for use in adult oncology. IAEA Human Health Series No.26, Vienna Groheux D, EspieM, Giacchetti S, Hindie E, 2013, Performance of FDG PET/CT in the clinical management of breast cancer. Radiology 266:388–405 Kajary K, Tokes T, Dank M, Kulka J, Szakall S Jr, Lengyel Z, 2015, Correlation of the value of 18F-FDG uptake, described by SUVmax, SUVavg, metabolic tumour volume and total lesion glycolysis, to clinicopathological prognostic factors and biological subtypes in breast cancer. Nucl Med Comm 36:28–37 Huang SC, 2000, Anatomy of SUV. Nucl Med Biol 27:643–646 Morris ED, Endres CJ, Schmidt KC, Christian BT, Muic RJ, Fisher RE, 2004, Kinetic modeling in positron emission tomography. In: Wernick MN, Aarsvold JN, eds, Emission tomography. The fundamentals of PET and SPECT. Elsevier Academic Press, San Diego, pp 499–540 Kelloff GJ, Hoffman JM, Johnson B, Scher HI, Siegel BA, Cheng EY et al, 2005, Progress and promise of FDG-PET imaging for cancer patient management and oncologic drug development. Clin Cancer Res 11:2785–2808 Curigliano G, Burstein HJ, Winer EP, Gnant M, Dubsky P, Loibl S et al, 2017, De-escalating and escalating treatments for early-stage breast cancer: the St Gallen international expert consensus conference on the primary therapy of early breast Cancer 2017. Ann Oncol 28:1700–1712 Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G et al, 2015, The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: recommendations by an International TILsWorking Group 2014. Ann Oncol 26:259–271 Tokes T, Kajary K, Dank M, Lengyel Z, 2018, Motion correction during dynamic FDG-PET/CT studies performed in the staging of breast cancer. Quarterly Journal of Nuclear Medicine and Molecular Imaging. , DOI 10.23736/S1824-4785.19. 03114-5 Soret M, Bacharach SL, Buvat I, 2007, Partial-volume effect in PET tumor imaging. J Nucl Med 48:932–945 Brierley JD, Gospodarowicz MK, Wittekind C, editors in chief, 2017, TNM classification of malignant tumours. Eighth Edition. Oxford: Wiley Allred DC, Harvey JM, Berardo M, Clark GM, 1998, Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11:155–168 Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ et al, 2007, American Society of Clinical Oncology/ College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol 25:118–145 Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH et al, 2013, Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol 31:3997–4013 DowsettM, Nielsen TO, A'Hern R, Bartlett J, Coombes RC, Cuzick J et al, 2011, Assessment of Ki67 in breast cancer: recommendations from the International Ki67 in Breast Cancer working group. J Natl Cancer Inst 103:1656–1664 Pathmanathan N, Balleine RL, 2013, Ki67 and proliferation in breast cancer. J Clin Pathol 66:512–516 Tokes AM, Szasz AM, Geszti F, Lukacs LV, Kenessey I, Turanyi E et al, 2015, Expression of proliferation markers Ki67, cyclin A, geminin and aurora-kinase A in primary breast carcinomas and corresponding distant metastases. J Clin Pathol 68:274–282 Shankar LK, Hoffman JM, Bacharach S, Graham MM, Karp J, Lammertsma AA et al, 2006, Consensus recommendations for the use of 18F-FDG PET as an indicator of therapeutic response in patients in National Cancer Institute Trials. J Nucl Med 47: 1059–1066 Boellaard R, O'Doherty MJ, Weber WA, Mottaghy FM, Lonsdale MN, Stroobants SG et al, 2010, FDG PET and PET/CT: EANM procedure guidelines for tumour PET imaging: version 1.0. Eur J Nucl Med Mol Imaging 37:181–200 Mankoff DA, Dunnwald LK, Gralow JR, Ellis GK, Charlop A, Lawton TJ et al, 2002, Blood flow and metabolism in locally advanced breast cancer: relationship to response to therapy. J Nucl Med 43:500–509 Dunnwald LK, Doot RK, Specht JM, Gralow JR, Ellis GK, Livingston RB et al, 2011, PET tumor metabolism in locally advanced breast cancer patients undergoing neoadjuvant chemotherapy: value of static versus kinetic measures of fluorodeoxyglucose uptake. Clin Cancer Res 17:2400–2409 Doot RK, Dunnwald LK, Schubert EK, Muzi M, Peterson LM, Kinahan PE et al, 2007, Dynamic and static approaches to quantifying 18F-FDG uptake for measuring cancer response to therapy, including the effect of granulocyte CSF. J Nucl Med 48:920–925 Partridge SC, Vanantwerp RK, Doot RK, Chai X, Kurland BF, Eby PR et al, 2010, Association between serial dynamic contrastenhanced MRI and dynamic 18F-FDG PET measures in patients undergoing neoadjuvant chemotherapy for locally advanced breast cancer. J Magn Reson Imaging 32:1124–1131 Tseng J, Dunnwald LK, Schubert EK, Link JM, Minoshima S, Muzi M et al, 2004, 18F-FDG kinetics in locally advanced breast cancer: correlation with tumor blood flow and changes in response to neoadjuvant chemotherapy. J Nucl Med 45:1829–1837 Dunnwald LK, Gralow JR, Ellis GK, Livingston RB, Linden HM, Specht JM et al, 2008, Tumor metabolism and blood flow changes by positron emission tomography: relation to survival in patients treated with neoadjuvant chemotherapy for locally advanced breast cancer. J Clin Oncol 26:4449–4457 Eby PR, Partridge SC, White SW, Doot RK, Dunnwald LK, Schubert EK et al, 2008, Metabolic and vascular features of dynamic contrast-enhanced breast magnetic resonance imaging and, 15)O-water positron emission tomography blood flow in breast cancer. Acad Radio 15:1246–1254 Wangerin KA, Muzi M, Peterson LM, Linden HM, Novakova A, Mankoff DA et al, 2017, A virtual clinical trial comparing static versus dynamic PET imaging in measuring response to breast cancer therapy. Phys Med Biol 62:3639–3655 Lee S, Choi S, Kim SY, Yun MJ, Kim HI, 2017, Potential utility of FDG PET-CT as a non-invasive tool for monitoring local immune responses. J Gastric Cancer 17:384–393 Lopci E, Toschi L, Grizzi F, Rahal D, Olivari L, Castino GF et al, 2016, Correlation of metabolic information on FDG-PET with tissue expression of immune markers in patients with non-small cell lung cancer, NSCLC, who are candidates for upfront surgery. Eur J Nucl Med Mol Imaging 43:1954–1961 Fujii T, Yanai K, Tokuda S, Nakazawa Y, Kurozumi S, Obayashi S et al, 2018, Relationship between FDG uptake and neutrophil/ lymphocyte ratio in patients with invasive ductal breast cancer. Anticancer Res 38:4927–4931 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 997 EP 1006 DI 10.1007/s12253-019-00641-0 PG 10 ER PT J AU Guo, Y Qiao, X Zhu, L Song, R AF Guo, Yan Qiao, Xue Zhu, Li Song, Rongbo TI MicroRNA-182-5p Modulates Oral Squamous Cell Carcinoma Migration and Invasion Via Targeting MTSS1 Gene SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral squamous cell carcinoma; miR-182-5p; MTSS1; Migration; Invasion ID Oral squamous cell carcinoma; miR-182-5p; MTSS1; Migration; Invasion AB Oral squamous cell carcinoma (OSCC) characterized with invasive growth, local metastasis and later stage diagnosis was a common malignancy in head and neck region. The aim of this study was to explore the relationship between miR-182-5p and OSCC, which will contribute to find potential biomarker for OSCC metastasis. MiR-182-5p expression level was detected by the quantitative real-time PCR (qRT-PCR). Cell migration and invasion ability were examined by scratch and Transwell assay. Loss of function together with luciferase reporter assay were used to verify the miR-182-5p modulated OSCC cells migration and metastasis was mediated by MTSS1. The expression of MTSS1 protein was examined by western blotting. MiR-182-5p upregulated in OSCC, was involved in the migration and invasion of OSCC and the increased miR-182-5p expression was correlated with lower OSCC differentiation grade, higher T and N stage. Bioinformatics analysis predicted MTSS1 gene was a potential target of miR-182-5p. Following co-transfection, qRT-PCR, luciferase activities assay and western blotting confirmed that MTSS1 gene was a direct target of miR-182-5p and silence of MTSS1 could reverse the effects of miR-182-5p on OSCC migration and invasion. MiR-182-5p was up-regulated in OSCC and the ability of miR-182-5p to promote MTSS1 repression may precipitate in the OSCC through bypassing cell migration and invasion control. C1 [Guo, Yan] China Medical University, School of Stomatology, Department of Central Laboratory, 117 North Nanjing Street, Heping District, 110002 Shenyang, Liaoning, China. [Qiao, Xue] China Medical University, School of Stomatology, Department of Central Laboratory, 117 North Nanjing Street, Heping District, 110002 Shenyang, Liaoning, China. [Zhu, Li] China Medical University, School of Stomatology, Department of Central Laboratory, 117 North Nanjing Street, Heping District, 110002 Shenyang, Liaoning, China. [Song, Rongbo] China Medical University, School of Stomatology, Department of Central Laboratory, 117 North Nanjing Street, Heping District, 110002 Shenyang, Liaoning, China. RP Guo, Y (reprint author), China Medical University, School of Stomatology, Department of Central Laboratory, 110002 Shenyang, China. EM yguo@cmu.edu.cn CR Krishna Rao SV, Mejia G, Roberts-Thomson K, Logan R, 2013, Epidemiology of oral cancer in Asia in the past decade–an update, 2000-2012). Asian Pac J Cancer Prev 14:5567–5577 de Morais EF, Mafra RP, Gonzaga AK,de Souza DL, Pinto LP, da Silveira EJ, 2016, Prognostic Factors of Oral Squamous Cell Carcinoma in Young Patients: A Systematic Review. J Oral Maxillofac Surg, pii: S0278–2391(16): 31286–31281 Kessler P, Grabenbauer G, Leher A, Bloch-Birkholz A, Vairaktaris E, Neukam FW, 2008, Neoadjuvant and adjuvant therapy in patients with oral squamous cell carcinoma Long-term survival in a prospective, non-randomized study. Br J Oral Maxillofac Surg 46(1):1–5 Leemans CR, Braakhuis BJ, Brakenhoff RH, 2011, The molecular biology of head and neck cancer. Nat Rev Cancer 11:9–22 Huang S, Tan X, Huang Z, Chen Z, Lin P, Fu SW, 2018, microRNA biomarkers in colorectal cancer liver metastasis. J Cancer 9(21):3867–3873 Shen J, ZhouW, Bi N, Song YM, Zhang FQ, Zhan QM,Wang LH, 2018, MicroRNA-886-3P functions as a tumor suppressor in small cell lung cancer. Cancer Biol Ther 19(12):1185–1192 Shang C, Guo Y, Hong Y, Xue YX, 2016, Long non-coding RNA TUSC7, a target of miR-23b, plays tumor-suppressing roles in human gliomas. Front Cell Neurosci 10:235 Kong P, Chen L, Yu M, Tao J, Liu J, Wang Y, Pan H, Zhou W, Wang S, 2018, miR-3178 inhibits cell proliferation and metastasis by targeting Notch1 in triple-negative breast cancer. Cell Death Dis 9(11):1059 Zhao D, Zhang Y, Song L, 2018, MiR-16-1 targeted silences far upstream element binding protein 1 to advance the Chemosensitivity to Adriamycin in gastric Cancer. Pathol Oncol Res 24(3):483–488 Shang C, TangW, Pan C, Hu XH, HongY, 2018, Long non-coding RNA TUSC7 inhibits temozolomide resistance by targeting miR- 10a in glioblastoma. Cancer Chemother Pharmacol 81(4):671–678 Sharifi M, Moridnia A, 2017, Apoptosis-inducing and antiproliferative effect by inhibition of miR-182-5p through the regulation of CASP9 expression in human breast cancer. Cancer Gene Ther 24(2):75–82 Li Y, Chen S, Shan Z, Bi L, Yu S, Li Y, Xu S, 2017, miR-182-5p improves the viability, mitosis, migration, and invasion ability of human gastric cancer cells by down-regulating RAB27A. Biosci Rep 37(3):pii: BSR20170136 Hirata H, Ueno K, Shahryari V, Deng G, Tanaka Y, Tabatabai ZL, Hinoda Y, Dahiya R, 2013, MicroRNA-182-5p promotes cell invasion and proliferation by down regulating FOXF2, RECK and MTSS1 genes in human prostate cancer. PLoS One 8(1):e55502 Suzuki R, Amatya VJ, Kushitani K, Kai Y, Kambara T, Takeshima Y, 2018, miR-182 and miR-183 promote cell proliferation and invasion by targeting FOXO1 in mesothelioma. Front Oncol 8:446 Golbakhsh MR, Boddouhi B, Hatami N, Goudarzi PK, Shakeri M, Yahaghi E, Taheriazam A, 2017, Down-regulation of microRNA- 182 and microRNA-183 predicts progression of osteosarcoma. Arch Med Sci 13(6):1352–1356 Xu X, Wu J, Li S, Hu Z, Xu X, Zhu Y, Liang Z, Wang X, Lin Y, Mao Y, Chen H, Luo J, Liu B, Zheng X, Xie L, 2014, Downregulation of microRNA-182-5p contributes to renal cell carcinoma proliferation via activating the AKT/FOXO3a signaling pathway. Mol Cancer 13:109 Guo Y, Ren MS, Shang C, Zhu L, Zhong M, 2015, MTSS1 gene regulated by miR-96 inhibits cell proliferation and metastasis in tongue squamous cellular carcinoma Tca8113 cell line. Int J Clin Exp Med 8(9):15441–15449 Marshall J, 2011, Transwell(®, invasion assays. Methods Mol Biol 769:97–110 Wang J, Li J, Shen J, Wang C, Yang L, Zhang X, 2012, MicroRNA-182 downregulates metastasis suppressor 1 and contributes tometastasis of hepatocellular carcinoma.BMC Cancer 12: 227 Lv Q, Hu JX, Li YJ, Xie N, Song DD, Zhao W, Yan YF, Li BS, Wang PY, Xie SY, 2017, MiR-320a effectively suppresses lung adenocarcinoma cell proliferation and metastasis by regulating STAT3 signals. Cancer Biol Ther 18(3):142–151 Rajabpour A, Afgar A, Mahmoodzadeh H, Radfar JE, Rajaei F, Teimoori-Toolabi L, 2017, MiR-608 regulating the expression of ribonucleotide reductase M1 and cytidine deaminase is repressed through induced gemcitabine chemoresistance in pancreatic cancer cells. Cancer Chemother Pharmacol 80(4):765–775 Wang J, Chen T, Shan G, 2017, miR-148b regulates proliferation and differentiation of neural stem cells via Wnt/β-catenin signaling in rat ischemic stroke model. Front Cell Neurosci 11:329 Xi Z,Wang P, Xue Y, Shang C, Liu X,Ma J, Li Z, Li Z, BaoM, Liu Y, 2017, Overexpression of miR-29a reduces the oncogenic properties of glioblastoma stem cells by downregulating quaking gene isoform 6. Oncotarget. 8(15):24949–24963 Tsuchiyama K, Ito H, Taga M, Naganuma S, Oshinoya Y, Nagano K, Yokoyama O, Itoh H, 2013, Expression of microRNAs associated with Gleason grading system in prostate cancer:miR-182-5p is a useful marker for high grade prostate cancer. Prostate. 73(8):827– 834 Xue J, Zhou A,WuY,Morris SA, LinK, Amin S,Verhaak R, Fuller G, Xie K, Heimberger AB, Huang S, 2016, miR-182-5p induced by STAT3 activation promotes glioma tumorigenesis. Cancer Res 76(14):4293–4304 Zeleniak AE, Huang W, Brinkman MK, Fishel ML, Hill R, 2017, Loss of MTSS1 results in increased metastatic potential in pancreatic cancer. Oncotarget. 8(10):16473–16487 Ling DJ, Chen ZS, Liao QD, Feng JX, Zhang XY, Yin TY, 2016, Differential effects of MTSS1 on invasion and proliferation in subtypes of non-small cell lung cancer cells. Exp Ther Med 12(2): 1225–1231 ShiW, Hasimu G,Wang Y, Li N, Chen M, Zhang H, 2015)MTSS1 is an independent prognostic biomarker for survival in intrahepatic cholangiocarcinoma patients. Am J Transl Res 7(10):1974–1983 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1007 EP 1013 DI 10.1007/s12253-019-00647-8 PG 7 ER PT J AU Shams, R Seifi-Alan, M Bandehpour, M Omrani, DM Ghafouri-Fard, S AF Shams, Roshanak Seifi-Alan, Mahnaz Bandehpour, Mojgan Omrani, Davood Mir Ghafouri-Fard, Soudeh TI C-X-C Chemokine Receptor Type 7 (CXCR-7) Expression in Invasive Ductal Carcinoma of Breast in Association with Clinicopathological Features SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CXCR-7; C-X-C chemokine receptor type 7; Breast cancer ID CXCR-7; C-X-C chemokine receptor type 7; Breast cancer AB C-X-C chemokine receptor type 7 (CXCR-7) is an atypical receptor for chemokines whose role in different stages of carcinogenesis has been evaluated in breast cancer cell lines and animal models. Moreover, it has been demonstrated to be a target of regulation by the tumor suppressor microRNA (miR)-100. In the present study, we assessed CXCR-7 expression in 60 breast cancer patients in association with clinicopathological and demographic data of patients. We also extracted the results of our previous work on miR-100 expression in the same cohort of patients to assess the correlation between miR-100 and CXCR-7 expression levels. Transcript levels of CXCR-7 were significantly higher in tumoral tissues compared with adjacent non-cancerous tissues (ANCTs) (Tumoral vs. ANCTs: 3.64 ± 1.8 vs. 0.73 ± 1.3, P = 0.000). A significant negative correlation was detected between CXCR-7 protein and miR-100 transcript levels (r = −0.526, P < 0.05). High CXCR-7mRNA levels were significantly associated with tumor size (P = 0.01). Besides, high protein levels were more prevalent in higher TNM stages (P = 0.000). Moreover, high CXCR-7 protein levels were significantly associated with ER (P = 0.005) and PR (P = 0.02) status. The present work provides further evidence for the role of CXCR-7 in breast cancer and proposes the elimination of inhibitory effects of miR-100 on CXCR-7 expression as a mechanism for its up-regulation in breast cancer tissues. C1 [Shams, Roshanak] Shahid Beheshti University of Medical Sciences, Department of Medical GeneticsTehran, Iran. [Seifi-Alan, Mahnaz] Shahid Beheshti University of Medical Sciences, Department of Medical GeneticsTehran, Iran. [Bandehpour, Mojgan] Shahid Beheshti University of Medical Sciences, School of Advanced Technologies in Medicine, Department of BiotechnologyTehran, Iran. [Omrani, Davood Mir] Shahid Beheshti University of Medical Sciences, Department of Medical GeneticsTehran, Iran. [Ghafouri-Fard, Soudeh] Shahid Beheshti University of Medical Sciences, Department of Medical GeneticsTehran, Iran. RP Ghafouri-Fard, S (reprint author), Shahid Beheshti University of Medical Sciences, Department of Medical Genetics, Tehran, Iran. EM s.ghafourifard@sbmu.ac.ir CR Burns JM et al, 2006, A novel chemokine receptor for SDF-1 and ITAC involved in cell survival, cell adhesion, and tumor development. J Exp Med 203:2201–2213. , DOI 10.1084/jem.20052144 TarnowskiMet al, 2010)Macrophagemigration inhibitory factor is secreted by rhabdomyosarcoma cells, modulates tumor metastasis by binding to CXCR4 and CXCR7 receptors and inhibits recruitment of Cancer-associated fibroblasts. Mol Cancer Res 8:1328– 1343. , DOI 10.1158/1541-7786.MCR-10-0288 Salazar N et al, 2014, The chemokine receptor CXCR7 interacts with EGFR to promote breast cancer cell proliferation.Mol Cancer 13, Artn 198. , DOI 10.1186/1476-4598-13-198 Miao Z et al, 2007, CXCR7, RDC1, promotes breast and lung tumor growth in vivo and is expressed on tumor-associated vasculature. ProcNatl Acad SciU S A 104:15735–15740. , DOI 10.1073/pnas.0610444104 Muir S. et al., AACR, 2008) Hernandez L, Magalhaes MAO, Coniglio SJ, Condeelis JS, Segall JE, 2011, Opposing roles of CXCR4 and CXCR7 in breast cancer metastasis. Breast Cancer Res 13, Artn R128. , DOI 10.1186/Bcr3074 Qian TT et al, 2018, CXCR7 regulates breast tumor metastasis and angiogenesis in vivo and in vitro. Mol Med Rep 17:3633–3639. , DOI 10.3892/mmr.2017.8286 Compton C et al, 2010, AJCC cancer staging manual. Chapter 14: 143–164 Barrett T, Edgar R, 2006, Gene expression omnibus: microarray data storage, submission, retrieval, and analysis. Method Enzymol 411:352–369. , DOI 10.1016/S0076-6879(06)11019-8 Tasharrofi B et al, 2016, Comparative expression analysis of hypoxia-inducible factor-alpha and its natural occurring antisense in breast cancer tissues and adjacent noncancerous tissues. Cell Biochem Funct 34:572–578. , DOI 10.1002/cbf.3230 Cao Yet al, 2018)MicroRNA-100 suppresses human gastric cancer cell proliferation by targeting CXCR7. Oncol Lett 15:453–458 Zhou S-M et al, 2016, miR-100 suppresses the proliferation and tumor growth of esophageal squamous cancer cells via targeting CXCR7. Oncol Rep 35:3453–3459 Zhou WH, Wu X, Hu WD, Du MR, 2015, Co-expression of CXCR4 and CXCR7 in human endometrial stromal cells is modulated by steroid hormones. Int J Clin Exp Pathol 8:2449–2460 Kerdivel G, Boudot A, Pakdel F, 2013, Estrogen represses CXCR7 gene expression by inhibiting the recruitment of NFκB transcription factor at the CXCR7 promoter in breast cancer cells. Biochem Biophys Res Commun 431:729–733 Boudot A et al, 2011, Differential estrogen-regulation of CXCL12 chemokine receptors, CXCR4 and CXCR7, contributes to the growth effect of estrogens in breast cancer cells. PLoS One 6:e20898 Liu Y, Beyer A, Aebersold R, 2016, On the dependency of cellular protein levels on mRNA abundance. Cell 165:535–550 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1015 EP 1020 DI 10.1007/s12253-019-00649-6 PG 6 ER PT J AU Zhou, XY Liu, H Ding, ZB Xi, HP Wang, GW AF Zhou, Xiang-Yang Liu, Hong Ding, Zheng-Bin Xi, Hai-Peng Wang, Guang-Wei TI lncRNA SNHG16 Exerts Oncogenic Functions in Promoting Proliferation of Glioma Through Suppressing p21 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Long non-coding RNA; SNHG16; p21; Glioma ID Long non-coding RNA; SNHG16; p21; Glioma AB Glioma is a malignant brain tumor that accounts for 30% of all brain tumors and 80% of malignant brain tumors. This poor clinical outcome makes the study of molecular mechanisms in glioma as an urgent subject. However, the certain mechanism remains unclear. Long non-coding RNAs (lncRNAs) plays a key role in glioma development and progression. In the present study, we aimed to explore the potential mechanisms of lncRNA SNHG16 in glioma. The levels of lncRNA SNHG16 were qualified in both glioma tissues and cell lines using qRT-PCR assay. The ability of cell proliferation was tested via CCK-8 and colony formation assays. Transfections were performed to knockdown SNHG16 and its target gene p21. The cell cycles and cell apoptosis were evaluated using flow cytometry, and the expression of SNHG16, p21 and apoptosis biomarkers were qualified with qRT-PCR and western blot assays. The expression of SNHG16 were up-regulated in both glioma tissues and cell lines. Knockdown of SNHG16 was associated with poor proliferation, decreased monoclonal formation rates, but increased apoptosis rates, which also caused the high expression of p21. Moreover, p21 could mediate cell proliferation and monoclonal formation, promote cell apoptosis in glioma, which was negatively correlated with lncRNA SNHG16. The molecule mechanism experiments revealed that SNHG16 could not only inhibit the expression of p21 but also suppressed the level of caspase 3 and 9, while promoted cyclinD1 and cyclinB1 expression. lncRNA SNHG16 could promote the cell proliferation and inhibit the apoptosis of glioma through suppressing p21, indicating that lncRNA SNHG16 might be quite vital for the diagnosis and progression of glioma and could even be a novel therapeutic target for glioma. C1 [Zhou, Xiang-Yang] The First Affiliated Hospital of University of South China, Department of Neurosurgery, No.69, Chuanshan Road, Shigu District, 421001 Hengyang, Hunan Province, China. [Liu, Hong] The First Affiliated Hospital of University of South China, Department of Neurosurgery, No.69, Chuanshan Road, Shigu District, 421001 Hengyang, Hunan Province, China. [Ding, Zheng-Bin] The First Affiliated Hospital of University of South China, Department of Neurosurgery, No.69, Chuanshan Road, Shigu District, 421001 Hengyang, Hunan Province, China. [Xi, Hai-Peng] The First Affiliated Hospital of University of South China, Department of Neurosurgery, No.69, Chuanshan Road, Shigu District, 421001 Hengyang, Hunan Province, China. [Wang, Guang-Wei] The First Affiliated Hospital of University of South China, Department of Neurosurgery, No.69, Chuanshan Road, Shigu District, 421001 Hengyang, Hunan Province, China. RP Wang, GW (reprint author), The First Affiliated Hospital of University of South China, Department of Neurosurgery, 421001 Hengyang, China. EM wangguangwei48@163.com CR Pines A, 2011, Hormone therapy and brain tumors. Climacteric 14(2):215–216 Yin CL et al, 2014, The role of glioma stem cells in glioma tumorigenesis. Front Biosci, Landmark Ed, 19:818–824 Chen JW, Zhou CF, Lin ZX, 2015, The influence of different classification standards of age groups on prognosis in high-grade hemispheric glioma patients. J Neurol Sci 356(1–2):148–152 Malzkorn B, Reifenberger G, 2016, Practical implications of integrated glioma classification according to the World Health Organization classification of tumors of the central nervous system 2016. Curr Opin Oncol 28(6):494–501 Liu G et al, 2018, Survival analysis for high-grade glioma patients who received comprehensive treatment. Zhong Nan Da Xue Xue Bao Yi Xue Ban 43(4):388–393 Li Y et al, 2015, Construction and analysis of lncRNA-lncRNA synergistic networks to reveal clinically relevant lncRNAs in cancer. Oncotarget 6(28):25003–25016 HuarteM(2015, The emerging role of lncRNAs in cancer. Nat Med 21(11):1253–1261 Li X et al, 2014, lncRNAs: insights into their function and mechanics in underlying disorders. Mutat Res Rev Mutat Res 762:1–21 Wang XP et al, 2017, Long non-coding RNA PAR5 inhibits the proliferation and progression of glioma through interaction with EZH2. Oncol Rep 38(5):3177–3186 Cui Yet al, 2017, Long noncoding RNA HOXA11-AS functions as miRNA sponge to promote the glioma tumorigenesis through targeting miR-140-5p. DNA Cell Biol 36(10):822–828 Ma CC et al, 2016, Long non-coding RNA ATB promotes glioma malignancy by negatively regulating miR-200a. J Exp Clin Cancer Res 35(1):90 Li H et al, 2016, Differential long noncoding RNA and mRNA expression in differentiated human glioblastoma stem cells. Mol Med Rep 14(3):2067–2076 Yang BYet al, 2018, Long non-coding RNA SNHG16 contributes to glioma malignancy by competitively binding miR-20a-5p with E2F1. J Biol Regul Homeost Agents 32(2):251–261 Yan L et al, 2017, Identification of lymph node metastasis-related microRNAs in lung adenocarcinoma and analysis of the underlying mechanisms using a bioinformatics approach. Exp Biol Med, Maywood, 242(7):709–717 Lu YF et al, 2018, LncRNA SNHG16 functions as an oncogene by sponging MiR-4518 and up-regulating PRMT5 expression in glioma. Cell Physiol Biochem 45(5):1975–1985 Liu K et al, 2017, LncRNA SNHG15 contributes to proliferation, invasion and autophagy in osteosarcoma cells by sponging miR- 141. J Biomed Sci 24(1):46 Chao Yet al, 2015, Mst1 regulates glioma cell proliferation via the AKT/mTOR signaling pathway. J Neuro-Oncol 121(2):279–288 Takano N et al, 2014, Decreased expression of cystathionine betasynthase promotes glioma tumorigenesis. Mol Cancer Res 12(10): 1398–1406 Sun J et al, 2018, RUNX3 inhibits glioma survival and invasion via suppression of the betacatenin/TCF-4 signaling pathway. J Neuro- Oncol 140(1):15–26 Hochberg FH et al, 2014, Glioma diagnostics and biomarkers: an ongoing challenge in the field of medicine and science. Expert Rev Mol Diagn 14(4):439–452 Hottinger AF, Hegi ME, Baumert BG, 2016, Current management of low-grade gliomas. Curr Opin Neurol 29(6):782–788 Goldbrunner R et al, 2018, The treatment of gliomas in adulthood. Dtsch Arztebl Int 115(20–21):356–364 Hart MG et al, 2013, Temozolomide for high grade glioma. Cochrane Database Syst Rev 4(4):CD007415 Zhang X et al, 2014, Combination of photodynamic therapy and temozolomide on glioma in a rat C6 glioma model. Photodiagn Photodyn Ther 11(4):603–612 Han X et al, 2018, LncRNATUG1 affects cell viability by regulating glycolysis in osteosarcoma cells. Gene 674:87–92 Yang Q et al, 2018, Long non-coding RNA PVT1 promotes cell proliferation and invasion through regulating miR-133a in ovarian cancer. Biomed Pharmacother 106:61–67 Almnaseer ZA, Mourtada-Maarabouni M, 2018, Long noncoding RNA MIAT regulates apoptosis and the apoptotic response to chemotherapeutic agents in breast cancer cell lines. Biosci Rep 38(4): BSR20180704 Kaikkonen MU, Lam MT, Glass CK, 2011, Non-coding RNAs as regulators of gene expression and epigenetics. Cardiovasc Res 90(3):430–440 Tsai MC et al, 2010, Long noncoding RNA as modular scaffold of histone modification complexes. Science 329(5992):689–693 Liu Yet al, 2015, Over-expression of lncRNA DANCR is associated with advanced tumor progression and poor prognosis in patients with colorectal cancer. Int J Clin Exp Pathol 8(9):11480– 11484 Zhang X et al, 2012, Long non-coding RNA expression profiles predict clinical phenotypes in glioma. Neurobiol Dis 48(1):1–8 Wei N,Wei H, Zhang H, 2018, Long non-coding RNA ZEB1-AS1 promotes glioma cell proliferation, migration and invasion through regulating miR-577. Eur Rev Med Pharmacol Sci 22(10):3085– 3093 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1021 EP 1028 DI 10.1007/s12253-019-00648-7 PG 8 ER PT J AU Liu, H Zhang, Q Lou, Q Zhang, X Cui, Y Wang, P Yang, F Wu, F Wang, J Fan, T Li, Sh AF Liu, Hongtao Zhang, Qing Lou, Qianqian Zhang, Xin Cui, Yunxia Wang, Panpan Yang, Fan Wu, Fan Wang, Jing Fan, Tianli Li, Shenglei TI Differential Analysis of lncRNA, miRNA and mRNA Expression Profiles and the Prognostic Value of lncRNA in Esophageal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Long chain non-coding RNA; MicroRNA; messenger RNA; Differential expression; ceRNA; Esophageal cancer ID Long chain non-coding RNA; MicroRNA; messenger RNA; Differential expression; ceRNA; Esophageal cancer AB Integrative central axis of lncRNA-miRNA-mRNA plays pivotal roles in tumor development and progression. However, the regulatory role of lncRNA-miRNA-mRNA in esophageal cancer remains elusive. TCGA database was utilized to investigate the differential expression of lncRNA, miRNA and mRNA in esophageal cancer (ESCA) and normal esophageal tissues, and GEO database was used to further validate the expression profile of key genes. Differential lncRNAs in TCGA database were submitted to Starbase, and lncRNAs related to overall survival were analyzed using Kaplan-Meier and log-rank test. We found 145 lncRNAs, 112 miRNAs and 2000 protein coding mRNAs were differentially expressed in ESCA samples, which were tightly involved in chromosome segregation, extracellular matrix assembly by GO assay, and KEGG assay revealed the correlation of differentially expressed genes with cell cycle, apoptosis and cGMP-PKG signaling pathway. Furthermore, there were 291 nodes in ceRNA network, which consisted of 40 lncRNAs, 28 miRNAs and 233 mRNAs, and formed 677 relations. Furthermore, 6 of 10 lncRNAs in TCGA database were consistent with GEO database, and expressions of 10 mRNAs in TCGA database all exhibited the same tendency with GEO database. Notably, we found 8 lncRNAs (WDFY3-AS2, CASC8, UGDH-AS1, RAP2C-AS1, AC007128.1, AC016205.1, AC092803.2 and AC079949.2) were correlated with overall survival of the patients with ESCA. The key differentially expressed genes participate in the development and progression of ESCA, and thus the elucidation of functions of lncRNA-miRNA-mRNA will provide new novel therapeutic target for the patients with ESCA. C1 [Liu, Hongtao] Zhengzhou University, College of Life Sciences, 100 Kexue Road, 450001 Zhengzhou, Henan, China. [Zhang, Qing] Zhengzhou University, College of Life Sciences, 100 Kexue Road, 450001 Zhengzhou, Henan, China. [Lou, Qianqian] Zhengzhou University, College of Life Sciences, 100 Kexue Road, 450001 Zhengzhou, Henan, China. [Zhang, Xin] Zhengzhou University, College of Life Sciences, 100 Kexue Road, 450001 Zhengzhou, Henan, China. [Cui, Yunxia] Zhengzhou University, College of Life Sciences, 100 Kexue Road, 450001 Zhengzhou, Henan, China. [Wang, Panpan] Zhengzhou University, College of Life Sciences, 100 Kexue Road, 450001 Zhengzhou, Henan, China. [Yang, Fan] Zhengzhou University, College of Life Sciences, 100 Kexue Road, 450001 Zhengzhou, Henan, China. [Wu, Fan] Zhengzhou University, College of Life Sciences, 100 Kexue Road, 450001 Zhengzhou, Henan, China. [Wang, Jing] Zhengzhou University, College of Life Sciences, 100 Kexue Road, 450001 Zhengzhou, Henan, China. [Fan, Tianli] Zhengzhou University, School of Basic Medicine, Department of Pharmacology, 100 Kexue Road, 450001 Zhengzhou, Henan, China. [Li, Shenglei] The First Affiliated Hospital of Zhengzhou University, Department of Pathology, 40 Daxue Road, 450052 Zhengzhou, Henan, China. RP Liu, H (reprint author), Zhengzhou University, College of Life Sciences, 450001 Zhengzhou, China. EM liuht1230@126.com CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90 Abnet CC,Arnold M,WeiWQ(2018, Epidemiology of esophageal squamous cell carcinoma. Gastroenterology 154(2):360–373 Arnold M, Soerjomataram I, Ferlay J, Forman D, 2015, Global incidence of oesophageal cancer by histological subtype in 2012. Gut 64(3):381–387 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108 Ohashi S, Miyamoto S, Kikuchi O, Goto T, Amanuma Y, Muto M, 2015, Recent advances from basic and clinical studies of esophageal squamous cell carcinoma. Gastroenterology 149(7):1700– 1715 Song G, Shen Y, Ruan Z, Li X, Chen Y, Yuan W, Ding X, Zhu L, Qian L, 2016, LncRNA-uc.167 influences cell proliferation, apoptosis and differentiation of P19 cells by regulating Mef2c. Gene 590(1):97–108 Zhao J, Liu Y, Zhang W, Zhou Z, Wu J, Cui P, Zhang Y, Huang G, 2015, Long non-coding RNA Linc00152 is involved in cell cycle arrest, apoptosis, epithelial to mesenchymal transition, cell migration and invasion in gastric cancer. Cell Cycle 14(19):3112–3123 Song R, Hou G, Yang J, Yuan J, Wang C, Chai T, Liu Z, 2018, Effects of PLK1 on proliferation, invasion and metastasis of gastric cancer cells through epithelial-mesenchymal transition. Oncol Lett 16(5):5739–5744 Morris KV,Mattick JS, 2014, The rise of regulatory RNA. Nat Rev Genet 15(6):423–437 Garzon R, Calin GA, Croce CM, 2009, MicroRNAs in Cancer. Annu Rev Med 60:167–179 HuarteM(2015, The emerging role of lncRNAs in cancer. Nat Med 21(11):1253–1261 Schmitt AM, Chang HY, 2016, Long noncoding RNAs in Cancer pathways. Cancer Cell 29(4):452–463 Evans JR, Feng FY, Chinnaiyan AM(2016, The bright side of dark matter: lncRNAs in cancer. J Clin Invest 126(8):2775–2782 Li DY, ChenWJ, Luo L,Wang YK, Shang J, Zhang Y, Chen G, Li SK, 2017, Prospective lncRNA-miRNA-mRNA regulatory network of long non-coding RNA LINC00968 in non-small cell lung cancer A549 cells: a miRNA microarray and bioinformatics investigation. Int J Mol Med 40(6):1895–1906 Yao K,Wang Q, Jia J, ZhaoH(2017)Acompeting endogenous RNA network identifies novel mRNA, miRNA and lncRNA markers for the prognosis of diabetic pancreatic cancer. Tumour Biol 39(6): 1010428317707882 Ye S, Yang L, Zhao X, Song W, Wang W, Zheng S, 2014, Bioinformatics method to predict two regulation mechanism: TFmiRNA- mRNAand lncRNA-miRNA-mRNA in pancreatic cancer. Cell Biochem Biophys 70(3):1849–1858 Liu Y, Zhu J, Ma X, Han S, Xiao D, Jia Y,Wang Y, 2018, ceRNA network construction and comparison of gastric cancer with or without Helicobacter pylori infection. J Cell Physiol Li R, Qu H, Wang S, Wei J, Zhang L, Ma R, Lu J, Zhu J, Zhong WD, Jia Z, 2018, GDCRNATools: an R/Bioconductor package for integrative analysis of lncRNA, miRNA and mRNA data in GDC. Bioinformatics 34(14):2515–2517 Anders S, Pyl PT, HuberW(2015, HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31(2): 166–169 Lun AT, Chen Y, Smyth GK, 2016, It's DE-licious: a recipe for differential expression analyses of RNA-seq experiments using quasi-likelihood methods in edgeR. Methods Mol Biol 1418:391– 416 Li JH, Liu S, Zhou H, Qu LH, Yang JH, 2014, starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res 42(Database issue):D92–D97 Jeggari A, Marks DS, Larsson E, 2012, miRcode: amap of putative microRNA target sites in the long non-coding transcriptome. Bioinformatics 28(15):2062–2063 Chou CH, Shrestha S, Yang CD, Chang NW, Lin YL, Liao KW, Huang WC, Sun TH, Tu SJ, Lee WH et al, 2018, miRTarBase update 2018: a resource for experimentally validated microRNAtarget interactions. Nucleic Acids Res 46(D1):D296–D302 Furio-Tari P, Tarazona S, Gabaldon T, Enright AJ, Conesa A, 2016, spongeScan: a web for detecting microRNA binding elements in lncRNA sequences. Nucleic Acids Res 44(W1):W176–W180 Yu G,Wang LG, Han Y, He QY, 2012, Clusterprofiler: an R package for comparing biological themes among gene clusters. OMICS 16(5):284–287 Ritchie ME, Phipson B,Wu D, Hu Y, Law CW, Shi W, Smyth GK, 2015, Limma powers differential expression analyses for RNAsequencing and microarray studies. Nucleic Acids Res 43(7):e47 Zhu H, Lu J, Zhao H, Chen Z, Cui Q, Lin Z,Wang X,Wang J,Dong H, Wang S et al, 2018, Functional long noncoding RNAs, lncRNAs, in clear cell kidney carcinoma revealed by reconstruction and comprehensive analysis of the lncRNA-miRNA-mRNA regulatory network. Med Sci Monit 24:8250–8263 Fan CN, Ma L, Liu N, 2018, Systematic analysis of lncRNAmiRNA- mRNA competing endogenous RNA network identifies four-lncRNA signature as a prognostic biomarker for breast cancer. J Transl Med 16(1):264 Lin P,Wen DY, Li Q, He Y, Yang H, Chen G, 2018, Genome-wide analysis of prognostic lncRNAs, miRNAs, and mRNAs forming a competing endogenous RNA network in hepatocellular carcinoma. Cell Physiol Biochem 48(5):1953–1967 Zhao J, Cheng W, He X, Liu Y, Li J, Sun J, Li J, Wang F, Gao Y, 2018, Construction of a specific SVM classifier and identification of molecular markers for lung adenocarcinoma based on lncRNAmiRNA- mRNA network. Onco Targets Ther 11:3129–3140 Chen X, Han H, Li Y, Zhang Q, Mo K, Chen S, 2016, Upregulation of long noncoding RNA HOTTIP promotes metastasis of esophageal squamous cell carcinoma via induction of EMT. Oncotarget 7(51):84480–84485 Song W, Zou SB, 2016, Prognostic role of lncRNA HOTAIR in esophageal squamous cell carcinoma. Clin Chim Acta 463:169– 173 Liu B, Pan CF, Yao GL, Wei K, Xia Y, Chen YJ, 2018, The long non-coding RNA AK001796 contributes to tumor growth via regulating expression of p53 in esophageal squamous cell carcinoma. Cancer Cell Int 18:38 Wang W, Zhu Y, Li S, Chen X, Jiang G, Shen Z, Qiao Y, Wang L, Zheng P, Zhang Y, 2016, Long noncoding RNA MALAT1 promotes malignant development of esophageal squamous cell carcinoma by targeting beta-catenin via Ezh2. Oncotarget 7(18):25668– 25682 Yao J, Huang JX, Lin M, Wu ZD, Yu H, Wang PC, Ye J, Chen P, Wu J, Zhao GJ, 2016, Microarray expression profile analysis of aberrant long non-coding RNAs in esophageal squamous cell carcinoma. Int J Oncol 48(6):2543–2557 CaoW,WuW, Shi F, Chen X,Wu L, Yang K, Tian F, Zhu M, Chen G,WangWet al, 2013, Integrated analysis of long noncoding RNA and coding RNA expression in esophageal squamous cell carcinoma. Int J Genomics 2013:480534 Wang W, Wei C, Li P, Wang L, Li W, Chen K, Zhang J, Zhang W, Jiang G, 2018, Integrative analysis of mRNA and lncRNA profiles identified pathogenetic lncRNAs in esophageal squamous cell carcinoma. Gene 661:169–175 Dong X, Kong C, Liu X, Bi J, Li Z, Li Z, Zhu Y, Zhang Z, 2018, GAS5 functions as a ceRNA to regulate hZIP1 expression by sponging miR-223 in clear cell renal cell carcinoma. Am J Cancer Res 8(8):1414–1426 Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP, 2011, A ceRNA hypothesis: the Rosetta stone of a hidden RNA language? Cell 146(3):353–358 Chi H, Yang R, Zheng X, Zhang L, Jiang R, Chen J, 2018, LncRNA RP11-79H23.3 Functions as a Competing Endogenous RNA to Regulate PTEN Expression through Sponging hsa-miR- 107 in the Development of Bladder Cancer. Int J Mol Sci 19(9) Wang P, Li J, Zhao W, Shang C, Jiang X,Wang Y, Zhou B, Bao F, Qiao H, 2018, A novel LncRNA-miRNA-mRNA triple network identifies LncRNA RP11-363E7.4 as an important regulator of miRNA and gene expression in gastric Cancer. Cell Physiol Biochem 47(3):1025–1041 Botti G, Marra L, Malzone MG, Anniciello A, Botti C, Franco R, Cantile M, 2017, LncRNA HOTAIR as prognostic circulating marker and potential therapeutic target in patients with tumor diseases. Curr Drug Targets 18(1):27–34 Wang Y, Li Y, Song HQ, Sun GW, 2018, Long non-coding RNA LINC00899 as a novel serum biomarker for diagnosis and prognosis prediction of acute myeloid leukemia. Eur Rev Med Pharmacol Sci 22(21):7364–7370 Yang L, Zhou JD, Zhang TJ, Ma JC, Xiao GF, Chen Q, Deng ZQ, Lin J, Qian J, Yao DM, 2018, Overexpression of lncRNA PANDAR predicts adverse prognosis in acute myeloid leukemia. Cancer Manag Res 10:4999–5007 Kiran M, Chatrath A, Tang X, Keenan DM, Dutta A, 2018, A prognostic signature for lower grade gliomas based on expression of long non-coding RNAs. Mol Neurobiol Mao Y, Fu Z, Zhang Y, Dong L, Zhang Y, Zhang Q, Li X, Liu J, 2018, A seven-lncRNA signature predicts overall survival in esophageal squamous cell carcinoma. Sci Rep 8(1):8823 Bao J, Zhou C, Zhang J, Mo J, Ye Q, He J, Diao J, 2018, Upregulation of the long noncoding RNA FOXD2-AS1 predicts poor prognosis in esophageal squamous cell carcinoma. Cancer Biomark 21(3):527–533 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1029 EP 1039 DI 10.1007/s12253-019-00655-8 PG 11 ER PT J AU Yang, Ch Huang, D Ma, C Ren, J Fu, L Cheng, Ch Li, B Shi, X AF Yang, Chenggang Huang, Di Ma, Cui Ren, Jing Fu, Lina Cheng, Cheng Li, Bangling Shi, Xiaofeng TI Identification of Pathogenic Genes and Transcription Factors in Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Osteosarcoma; Transcription factors; DEGs; Integrated analysis ID Osteosarcoma; Transcription factors; DEGs; Integrated analysis AB Osteosarcoma (OS) is an aggressive malignant tumor of the bones. Our study intended to identify and analyze potential pathogenic genes and upstream regulators for OS. We performed an integrated analysis to identify candidate pathogenic genes of OS by using three Gene Expression Omnibus (GEO) databases (GSE66673, GSE49003 and GSE37552). GO and KEGG enrichment analysis were utilized to predict the functional annotation and potential pathways of differentially expressed genes (DEGs). The OS-specific transcriptional regulatory network was established to study the crucial transcriptional factors (TFs) which target the DEGs in OS. From the three GEO datasets, we identified 759 DEGs between metastasis OS samples and non-metastasis OS samples. After GO and KEGG analysis, ‘cell adhesion’ (FDR = 1.27E-08), ‘protein binding’ (FDR = 1.13E-22), ‘cytoplasm’ (FDR = 5.63E-32) and ‘osteoclast differentiation’ (FDR = 0.000992221) were significantly enriched pathways for DEGs. HSP90AA1 exhibited a highest degree (degree = 32) and was enriched in ‘pathways in cancer’ and ‘signal transduction’. BMP6, regulated by Pax-6, was enriched in the ‘TGF-beta signaling pathway’. We indicated that BMP6 may be downregulated by Pax-6 in the non-metastasis OS samples. The up-regulated HSP90AA1 and down-regulated BMP6 and ‘pathways in cancer’ and ‘signal transduction’ were deduced to be involved in the pathogenesis of OS. The identified biomarkers and biological process in OS may provide foundation for further study. C1 [Yang, Chenggang] Gu’an Bojian Bio-Technology Co. LTDLangfang, China. [Huang, Di] Gu’an Bojian Bio-Technology Co. LTDLangfang, China. [Ma, Cui] Gu’an Bojian Bio-Technology Co. LTDLangfang, China. [Ren, Jing] Beijing Medintell Bioinformatic Technology Co. LTD, No. 1, Shanyuan Street, Haidian District, 100081 Beijing, China. [Fu, Lina] Beijing Medintell Bioinformatic Technology Co. LTD, No. 1, Shanyuan Street, Haidian District, 100081 Beijing, China. [Cheng, Cheng] Beijing Medintell Bioinformatic Technology Co. LTD, No. 1, Shanyuan Street, Haidian District, 100081 Beijing, China. [Li, Bangling] Jiangnan University, School of biotechnologyWuxi, China. [Shi, Xiaofeng] Gu’an Bojian Bio-Technology Co. LTDLangfang, China. RP Shi, X (reprint author), Gu’an Bojian Bio-Technology Co. LTD, Langfang, China. EM shi.xiaofeng@medintell.com CR ZhouW, Hao M, Du X, Chen K,Wang G, Yang J, 2014, Advances in targeted therapy for osteosarcoma. Discov Med 17(96):301–307 Bernardini, G., Geminiani, M., Gambassi, S., Orlandini, M., Petricci, E., & Marzocchi, B., et al., 2017). Novel smoothened antagonists as anti-neoplastic agents for the treatment of osteosarcoma. J Cell Physiol Yan H, Zhang B, Fang C, Chen L, 2018, Mir-340 alleviates chemoresistance of osteosarcoma cells by targeting zeb1. Anti- Cancer Drugs:1 Pang Y, Zhao J, Fowdur M, Liu Y,Wu H, HeM, 2018, To explore the mechanism of the grm4 gene in osteosarcoma by rna sequencing and bioinformatics approach. Med Sci Monit Basic Res 24:16– 25 Mitchell PJ, Tjian R, 1989, Transcriptional regulation in mammalian cells by sequence-specific dna binding proteins. Science 245(4916):371–378 ZhuM, Liu CC, Cheng C, 2013, Reactin: regulatory activity inference of transcription factors underlying human diseases with application to breast cancer. BMC Genomics 14(1):504 Yang L, Feng S, Yang Y, 2016, Identification of transcription factors, tfs, and targets involved in the cholangiocarcinoma, cca, by integrated analysis. Cancer Gene Ther 23(12):439–445 Diao C, Xi Y, Xiao T, 2018, Identification and analysis of key genes in osteosarcoma using bioinformatics. Oncol Lett 15(3): 2789–2794 Liu HY, Zhang CJ, 2017, Identification of differentially expressed genes and their upstream regulators in colorectal cancer. Cancer Gene Ther 24:244–250 Benjamini Y, Hochberg Y, 1995, Controlling the false discovery rate-a practical and powerful approach to multiple testing. J R Stat Soc 57(1):289–300 Li JJ, Wang BQ, Fei Q, Yang Y, Li D, 2016, Identification of candidate genes in osteoporosis by integrated microarray analysis. Bone Joint Res 5(12):594–601 Wang F,Wang R, Li Q, Qu X, Hao Y, Yang J, Zhao H,Wang Q, Li G, Zhang F, Zhang H, Zhou X, Peng X, Bian Y, Xiao W, 2017, A transcriptome profile in hepatocellular carcinomas based on integrated analysis of microarray studies. Diagn Pathol 12(1):4 Lee YS, Kim JK, Ryu SW, Bae SJ, Kwon K, Noh YH, Kim SY, 2015, Integrative meta-analysis of multiple gene expression profiles in acquired gemcitabine-resistant cancer cell lines to identify novel therapeutic biomarkers.Asian Pac J Cancer Prev 16(7):2793– 2800 Luetke A, Meyers PA, Lewis I, Juergens H, 2014, Osteosarcoma treatment - where do we stand? A state of the art review. Cancer Treat Rev 40(4):523–532 Guan D, Tian H, 2017, Integrated network analysis to explore the key genes regulated by parathyroid hormone receptor 1 in osteosarcoma. World J Sugr Oncol 15(1):177 Taipale M, Jarosz DF, Lindquist S, 2010, Hsp90 at the hub of protein homeostasis: emerging mechanistic insights. Nat Rev Mol Cell Biol 11(7):515–528 Coskunpinar E, Akkaya N, Yildiz P, Oltulu YM, Aynaci E, Isbir T, Yaylim I, 2014, The significance of hsp90aa1, hsp90ab1 and hsp90b1 gene polymorphisms in a turkish population with nonsmall cell lung cancer. Anticancer Res 34(2):753–757 Chu SH, Liu YW, Zhang L, Liu B, Li L, Shi JZ, Li L, 2013, Regulation of survival and chemoresistance by hsp90aa1 in ovarian cancer skov3 cells. Mol Biol Rep 40(1):1–6 Mckay, W., Boden, S., & Yoon, S., 2006). METHODS OF I N D U C I N G T H E E X P R E S S I O N O F B O N E MORPHOGENETIC PROTEI N S, BMP s , AND TRANSFORMING GROWTH FACTOR-beta-PROTEINS, TGF-betas, IN CELLS. CA, EP1629106 Honda Y, Knutsen R, StrongDD, Sampath TK, Baylink DJ, Mohan S, 1997, Osteogenic protein-1 stimulates mrna levels of bmp-6 and decreases mrna levels of bmp-2 and -4 in human osteosarcoma cells. Calcif Tissue Int 60(3):297–301 Shi, Y. J., & Pan, X. T., 2016). Bmp6 and bmp4 expression in patients with cancer-related anemia and its relationship with hepcidin and s-hjv. Genet Mol Res Gmr, 15(1) Hu F, Zhang Y, Li M, Zhao L, Chen J, Yang S et al, 2015, Bmp-6 inhibits the metastasis of mda-mb-231 breast cancer cells by regulating mmp-1 expression. Oncol Rep 35(3) Liu G, Liu YJ, Lian WJ, Zhao ZW, Yi T, Zhou HY, 2014, Reduced bmp6 expression by dna methylation contributes to emt and drug resistance in breast cancer cells. Oncol Rep 32(2): 581–588 Zong X, Yang H, Yu Y, Zou D, Ling Z, He X, Meng X, 2011, Possible role of pax-6 in promoting breast cancer cell proliferation and tumorigenesis. BMB Rep 44(9):595–600 Shyr CR, Tsai MY, Yeh S, Kang HY, Chang YC, Wong PL et al, 2010, Tumor suppressor pax6 functions as androgen receptor co-repressor to inhibit prostate cancer growth. Prostate 70(2):190–199 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1041 EP 1048 DI 10.1007/s12253-019-00645-w PG 8 ER PT J AU Gasinska, A Jaszczynski, J Rychlik, U Luczynska, E Pogodzinski, M Palaczynski, M AF Gasinska, Anna Jaszczynski, Janusz Rychlik, Urszula Luczynska, Elzbieta Pogodzinski, Marek Palaczynski, Mikolaj TI Prognostic Significance of Serum PSA Level and Telomerase, VEGF and GLUT-1 Protein Expression for the Biochemical Recurrence in Prostate Cancer Patients after Radical Prostatectomy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostatic carcinoma; Biochemical failure; PSA; GLUT-1; VEGF; hTERT ID Prostatic carcinoma; Biochemical failure; PSA; GLUT-1; VEGF; hTERT AB The aim of the study was to evaluate prognosis for biochemical recurrence (BR) by analysing the pathological and biological characteristics of prostate cancer (PCa) after radical prostatectomy (RP). There were 130 men with clinically localized PCa in whom pretreatment serum PSA level and Ki-67, prostate specific membrane antigen (PSMA), glucose transporter-1 (GLUT-1), vascular endothelial growth factor (VEGF), microvessel density (MVD) and human telomerase reverse transcriptase (hTERT) proteins expression, based on number of immunohistochemically positive cells (labelling index), were retrospectively studied. In order to assess the prognostic significance of analysed variables in univariate and multivariate Cox analysis, patients were dichotomized based on cut-off points chosen by receiver operating characteristic (ROC) curves. There were 83 males (63.8%) at pTstage 1–2 and 47 (36.1%) at pTstage 3–4, respectively, with median (range) age of 62.8 years (49–77), and median follow-up of 78.5 months (12–148). In 42 (32.3%) men BR was found. In univariate analysis, tumour biological features: PSA ≤8 ng/ mL (p = 0.006), Ki-67LI ≤ 12.7% (p = 0.015), VEGFLI>11.0% (p = 0.030), and hTERTLI>6.7% (p = 0.016), but not clinicopathological parameters, appeared to be positive prognosticators for BRFS. In the Cox analysis, Ki-67 lost its significance, and clinicopathological parameters appeared to be nonsignificant. The independent negative prognostic factors for BRFS were: PSA > 8.0 ng/mL, (Hazard ratio = 2.75, p = 0.003), GLUT-1 > 19.1% (HR = 2.1, p = 0.032), VEGF≤11.0% (HR = 1, p = 0.024) and hTERT≤6.7% (HR = 1, p = 0.017). High PSA level, and GLUT-1 expression and lower VEGF and nuclear hTERT expression may indicate the great role of hypoxia in BR induction in PCa. C1 [Gasinska, Anna] Maria Sklodowska - Curie Institute, Cracow Branch, Oncology Center, Department of Tumour Pathology, Garncarska 11, 31-115 Krakow, Poland. [Jaszczynski, Janusz] Maria Sklodowska - Curie Institute, Cracow Branch, Oncology Center, Department of SurgeryKrakow, Poland. [Rychlik, Urszula] Maria Sklodowska - Curie Institute, Cracow Branch, Oncology Center, Department of Clinical BiochemistryKrakow, Poland. [Luczynska, Elzbieta] Maria Sklodowska - Curie Institute, Cracow Branch, Oncology Center, Department of RadiologyKrakow, Poland. [Pogodzinski, Marek] Maria Sklodowska - Curie Institute, Cracow Branch, Oncology Center, Department of SurgeryKrakow, Poland. [Palaczynski, Mikolaj] Maria Sklodowska - Curie Institute, Cracow Branch, Oncology Center, Department of SurgeryKrakow, Poland. RP Gasinska, A (reprint author), Maria Sklodowska - Curie Institute, Cracow Branch, Oncology Center, Department of Tumour Pathology, 31-115 Krakow, Poland. EM z5gasins@cyf-kr.edu.pl CR Mahal BA, Yang DD, Wang NQ, Alshalalfa M, Davicioni E, Choeurng V, Schaeffer EM, Ross AE, Spratt DE, den RB, Martin NE, Mouw KW, Orio PF 3rd, Choueiri TK, Taplin ME, Trinh QD, Feng FY, Nguyen PL, 2011, Clinical and genomic characterization of low-prostate-specific antigen, high-grade prostate cancer. Eur Urol 74:146–154. , DOI 10.1016/j.eururo.2018.01.043 Bill-Axelson A, Holmberg L, Ruutu M, Haggman M, Andersson SO, Bratell S, Spangberg A, Busch C, Nordling S, Garmo H, Palmgren J, Adami HO, Norlen BJ, Johansson JE, 2005, Radical prostatectomy versus watchful waiting in early prostate cancer. N Engl J Med 352(19):1977–1984. , DOI 10.1056/ NEJMoa043739 Duffy MJ, 2014, PSA in screening for prostate cancer: more good than harm or more harm than good? Adv Clin Chem 66:1–23 Xia J, Trock B, Gulati R et al, 2014, Overdetection of recurrence after radical prostatectomy: estimates based on patient and tumor characteristics. Clin Cancer Res 20. , DOI 10.1158/1078- 0432.CCR-13-3366 Shao Y-H, Demissie K, ShihW, Mehta AR, SteinMN, Roberts CB, DiPaola RS, Lu-Yao GL, 2009, Contemporary risk profile of prostate cancer in the United States. J Natl Cancer Inst 101:1280–1283. , DOI 10.1093/jnci/djp262 Vergis R, Corbishley CM, Norman AR, Bartlett J, Jhavar S, Borre M, Heeboll S, Horwich A, Huddart R, Khoo V, Eeles R, Cooper C, Sydes M, DearnaleyD, Parker C, 2008, Intrinsicmarkers of tumour hypoxia and angiogenesis in localized prostate cancer and outcome of radical treatment: a retrospective analysis of two randomized radiotherapy trials and one surgical cohort study. Lancer Oncol 9: 342–351. , DOI 10.1016/S1470-2045(08)70076-7 Kim SH, Park WS, Park BR, Joo J, Joung JY, Seo HK, Chung J, Lee KH, 2017, PSCA, Cox-2, and Ki-67 are independent, predictive markers of biochemical recurrence in clinically localized prostate cancer: a retrospective study. Asian J Androl 19(4):458–462. , DOI 10.4103/1008-682X.180798 Bettencourt MC, Bauer JJ, Sesterhenn IA, Mostofi FK, McLeod DAG, Moul JW, 1996, Ki-67 expression is a prognostic marker of prostate cancer recurrence after radical prostatectomy. J Urol 156:1064–1068 Leon P, Cancel-Tassin G, Drouin S et al, 2018, Comparison of cell cycle progression score with two immunohistochemical markers, PTEN and Ki-67, for predicting outcome in prostate cancer after radical prostatectomy. World J Urol 36:1495–1500. , DOI 1007/s00345-018-2290-y 10. Kristiansen G, 2012, Diagnostic and prognostic molecular biomarkers for prostate cancer. Histopathol 60:125–141 Lalonde E, Ishkanian AS, Sykes J, Fraser M, Ross-Adams H, Erho N, Dunning MJ, Halim S, Lamb AD, Moon NC, Zafarana G, Warren AY, Meng X, Thoms J, Grzadkowski MR, Berlin A, Have CL, Ramnarine VR, Yao CQ,Malloff CA, Lam LL, Xie H, Harding NJ, Mak DYF, Chu KC, Chong LC, Sendorek DH, P'ng C, Collins CC, Squire JA, Jurisica I, Cooper C, Eeles R, Pintilie M, Dal Pra A, Davicioni E, Lam WL, Milosevic M, Neal DE, van der Kwast T, Boutros PC, Bristow RG, 2014, Tumour genomic and microenvironmental heterogeneity for integrated prediction of 5-year biochemical recurrence of prostate cancer: a retrospective cohort study. Lancet Oncol 15:1521–1532. , DOI 10.1016/S1470- 2045(14)71021-6 Shiradkar R, Ghose S, Jambor I, Taimen P, Ettala O, Purysko AS, Madabhushi A, 2018, Radiomic features from pretreatment biparametric MRI predict prostate cancer biochemical recurrence: preliminary findings. J Magn Reson Imaging 48:1626–1636. , DOI 10.1002/jmri.26178 Vandergrift LA, Decelle EA, Kurth J, Wu S, Fuss TL, DeFeo EM, Halpern EF, TaupitzM, McDougalWS, Olumi AF,Wu CL, Cheng LL, 2018, Metabolomic prediction of human prostate cancer aggressiveness: magnetic resonance spectroscopy of histologically benign tissue. Sci Rep 8:4997. , DOI 10.1038/s41598- 018-23177-w Luczynska E, Gasinska A, Wilk W, 2012, Expression of Ki-67, MIB-1, and GLUT-1 proteins in non-advanced prostatic Cancer. Pol J Pathol 63:272–277 Gasinska A, Luczynska E,WilkW, Cichocka A, 2013, Expression of telomerase and prostate-specific membrane antigen in nonadvanced prostatic Cancer. Folia Histochem Cytobiol 51:66–72. , DOI 10.5603/FHC.2013.0010 Luczynska E, Gasinska A,WilkW(2013, Microvessel density and vascular endothelial growth factor expression in clinically localized prostate cancer. Pol J Pathol 1:33–38 Gordan JD, Simon MC, 2007, Hypoxia-inducible factors: central regulatirs of the tumor phenotype. Curr Opin Genet Dev 17:71–77. , DOI 10.1016/j.gde.2006.12.006 Abou-Ouf H, Alshalalfa M, Takhar M, Erho N, Donnelly B, Davicioni E, Karnes RJ, Bismar TA, 2018, Validation of 10-gene molecular signature for predicting biochemical recurrence and clinical metastasis in localized prostate. J Cancer Res Clin Oncol 144: 883–891. , DOI 10.1007/s00432-018-2615-7 Stackhouse GB, Sesterhenn IA, Bauer JJ et al, 1999, p53 and BCL- 2 immunohistochemistry in pretreatment prostate needle biopsies to predict recurrence of prostate cancer after radical prostatectomy. J Urol 162:2040–2045 Tsan D-L, Fan K-H, Chen Y-C et al, 2013, Pre-radiotherapy PSA level as a predictor for biochemical control in prostate Cancer patients receiving radiotherapy after radical prostatectomy. Biom J 36: 71–76 Roehl KA, Han M, Ramos CG et al, 2004, Cancer progression and survival rates following anatomical radical retropubic prostatectomy in 3,478 consecutive patients: long-term results. J Urol 172: 910–914. , DOI 10.1097/01.ju.0000134888.22332.bb Minner S,Wittmer C, Graefen M, Salomon G, Steuber T, Haese A, Huland H, Bokemeyer C, Yekebas E, Dierlamm J, Balabanov S, Kilic E, Wilczak W, Simon R, Sauter G, Schlomm T, 2011, High level PSMA expression is associated with PSA recurrence in surgically treated prostate cancer. Prostate 71:281–288. , DOI 10.1002/pros.21241 De la Taille A,Katz AE, Bagiella E et al, 2000)Microvessel density as a predictor of PSA recurrence after radical prostatectomy. A comparison of CD34 and CD31. Am J Clin Pathol 113:555–562. , DOI 10.1309/02W2-KE50-PKEF-G2G4 Wen S, Niu Y, Lee SO, Chang C, 2014, Androgen receptor, AR, vs negative roles in prostate cancer cell deaths including apoptosis, anoikis, entosis, necrosis and autophage cell death. Cancer Treat Rev 40:31–40. , DOI 10.1016/j.ctrv.2013.07.008 Joseph IBJK, Nelson JB, Denmeade SR et al, 1997, Androgens regulate vascular endothelial growth factor content in normal and malignant prostatic tissue. Clin Cancer Res 3:2507–2511. https:// doi.org/10.1016/j.ctrv.2013.07.008 Mabjeesh NJ, Amir S, 2007, Hypoxia-inducible factor, HIF, in human tumorigenesis. Histol Histopathol 22:559–572. https://doi. org/10.14670/HH-22.559 Mao K, Badoual C, Camparo P et al, 2008, The prognostic value of vascular endothelial growth factor, VEGF)-A and its receptor in clinically localized prostate cancer: a prospective evaluation in 100 patients undergoing radical prostatectomy. Can J Urol 15: 4257–4262 Ahmed S, Passos JF, Birket MJ, Beckmann T, Brings S, Peters H, Birch-Machin MA, von Zglinicki T, Saretzki G, 2008, Telomerase does not counteract telomere shortening but protects mitochondrial function under oxidative stess. J Cell Sci 121:1046–1053. https:// doi.org/10.1242/jcs.019372 Saretzki G, 2014, Extra-telomeric functions of human telomerase: cancer, mitochondria and oxidative stress. Curr Pharm Des 20(41): 6386–6403 PMID:24975608 Moehren U, Papaionnou M, Reeb CA et al, 2008, Wild-type but not mutant androgen receptor inhibits expression of the hTERT telomerase subunit: a novel role of AR mutation for prostate cancer development. FASEB J 22:4–1267. , DOI 10.1096/fj.07- 9360com NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1049 EP 1056 DI 10.1007/s12253-019-00659-4 PG 8 ER PT J AU Li, Z Guo, Z AF Li, Zhi Guo, Zhiqiang TI Comparison of CDH1 Gene Hypermethylation Status in Blood and Serum among Gastric Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Hypermethylation; CDH1 gene; Prognosis ID Gastric cancer; Hypermethylation; CDH1 gene; Prognosis AB Hypermethylation is epigenetic alteration, well known for gene silencing. CHD1 gene is known as invasion and tumor suppressor gene, decreased expression due to hypermethylation could promote tumor cell invasion and metastasis. Present study designed to investigate the CDH1 gene promoter hypermethylation status by methylation specific polymerase chain reaction in 100 newly diagnosed gastric cancer patients. 53% of hypermethylation was observed in DNA extracted from blood in Gastric cancer patients while 66% was observed in serum-based DNA. Significant differences in CDH1gene promoter hypermethylation was observed in serum-based DNA extracted from Gastric cancer patients. Patients in early stage (I&II) vs advanced stage (III&IV), distant organ metastases vs no metastases had 60% vs 7% and 42% 24% of CDH1 promoter hypermethylation in serum DNA (p = 0.006, 0.001) respectively. Patients who were with lymph node invasion, loss of appetite, loss of weight had 55%, 47%, 61% CDH1 gene promoter hypermethylation compare to who were not with lymph node invasion, loss of appetite, loss of weight had 11%, 19%, 5% of hypermethylation and these differences was found to be significant. Strong association was observed with overall median survival of patients (p < 0.0001). Patients who had CDH1 gene promoter hypermethylation in serum-based DNA showed poor overall median survival (14.3 months) and unmethylated patients had better overall median survival (33.2 months). CDH1 hypermethylation status was found to be associated with advancement of disease, distant organ metastases and lymph node invasion in Gastric cancer patients. C1 [Li, Zhi] Lanzhou petrochemical general hospital, Gansu Gem Flower Hospital, Physical examination center, 730060 Lanzhou, Gansu, China. [Guo, Zhiqiang] Lanzhou petrochemical general hospital, Gansu Gem Flower Hospital, Medical section, 730060 Lanzhou, Gansu, China. RP Guo, Z (reprint author), Lanzhou petrochemical general hospital, Gansu Gem Flower Hospital, Medical section, 730060 Lanzhou, China. EM georgiadespori@yahoo.com CR Siegel R, Ma J, Zou Z, Jemal A, 2014, Cancer statistics, 2014. CA Cancer J Clin 64:9–29 Lee JH, Han SU, Cho H, Jennings B, Gerrard B, DeanM, Schmidt L, Zbar B, Vande Woude GF, 2000, A novel germ line juxtamembrane Met mutation in human gastric cancer. Oncogene 19(43):4947–4953 Yousuf A, Bhat MY, Pandith AA, Afroze D, Khan NP, Alam K, Shah P, Shah MA, Mudassar S, 2014, MGMT gene silencing by promoter hypermethylation in gastric cancer in a high incidence area. Cell Oncol 37:245–252 Goffin J, Eisenhauer E, 2002, DNA methyltransferase inhibitors— state of the art. Ann Oncol 13:1699–1716 Baylin SB, Belinsky SA, Herman JG, 2000, Aberrant methylation of gene promoters in cancer. Concepts, misconcepts and promise. J Natl Cancer Inst 92:1460–1461 Van RF, 2014, Beyond E-cadherin: roles of other cadherin superfamily members in cancer. Nat Rev Cancer 14:121–134 Karayiannakis AJ, Syrigos KN, Chatzigianni E, Papanikolaou S, Alexiou D, Kalahanis N et al, 1998, Aberrant E-cadherin expression associated with loss of differentiation and advanced stage in human pancreatic cancer. Anticancer Res 18:4177–4180 Humar B, Blair V, Charlton A,More H, Martin I, Guilford P, 2009, E-cadherin deficiency initiates gastric signet-ring cell carcinoma in mice and man. Cancer Res 69:2050–2056 Galanter JM, Christopher RG, Sam SO, Torgerson D, Pino-Yanes M, Thakur N et al, 2015, Methylation analysis reveals fundamental differences between ethnicity and genetic ancestry. Bio Rxiv. , DOI 10.1101/036822 Kang GH, Lee HJ, Hwang KS, Lee S, Kim JH, Kim JS, 2003, Aberrant CpG Island hypermethylation of chronic gastritis, in relation to aging, gender, intestinal metaplasia, and chronic inflammation. Am J Pathol 163:1551–1556 Choi IS, Wu TT, 2005, Epigenetic alterations in gastric Carcinogenesis. Cell Res 15:247–254 Nawroz H, Koch W, Anker P, Stroun M, Sidransky D, 1996, Microsatellite alterations in serum DNA of head and neck cancer patients. Nat Med 2(9):1035–1037 Hibi K, Robinson CR, Booker S,Wu L, Hamilton SR, Sidransky D, Jen J, 1998)Molecular detection of genetic alterations in the serum of colorectal cancer patients. Cancer Res 58:1405–1407 KopreskiMS, Benko FA, BorysDJ, Khan A,KweeCM,McGarrity TJ et al, 1998)Mutated KRAS DNA in plasma as a marker in early colorectal neoplasia. Proc Am Soc Clin Oncol 17:269 Lee TL, LeungWK, Chan MW, Ng EK, Tong JH, Lo KW, Chung SC, Sung JJ, To KF, 2002, Detection of gene promoter hypermethylation in the tumor and serum of patients with gastric carcinoma. Clin Cancer Res 8:1761–1766 Rashid H, Alam K, Afroze D, Yousuf A, Banday M, Kawoosa F, 2016, Hypermethylation status of E-Cadherin gene in gastric cancer patients in a high incidence area. APJCP 17(6):2757 Tamura G, Yin J, Wang S, Fleisher AS, Abraham JM, Kong D, 2000, E-Cadherin gene promoter hypermethylation in primary human gastric carcinomas. J Natl Cancer Inst 92:569–573 Benton G, Crooke E, George J, 2009, Laminin-1 induces Ecadherin expression in 3-dimensional cultured breast cancer cells by inhibiting DNA methyltransferase 1 and reversing promoter methylation status. FASEB J 23:3884–3895 Cheng CW,Wu PE, Yu JC, Huang CS, Yue CT,Wu CW, Shen CY, 2001)Mechanisms of inactivation of E-cadherin in breast carcinoma: modification of the two-hit hypothesis of tumor suppressor gene. Oncogene 20:3814–3823 Liu J, Sun X, qin S, Wang H, Ning D, Li Y et al, 2016, CDH1 promoter methylation correlates with decreased gene expression and poor prognosis in patients with breast cancer. Oncol Lett 11: 2635–2643 Caldeira JR, Prando EC, Quevedo FC, Neto FA, Rainho CA, Rogatto SR, 2006, CDH1 promoter hypermethylation and Ecadherin protein expression in infiltrating breast cancer. BMC Cancer 6:48 Lee S, Lee HJ, KimJH, Lee HS, Jang JJ, Kang GH, 2003, Aberrant CpG i s l a n d h y p e r m e t h y l a t i o n a l o n g m u l t i s t e p hepatocarcinogenesis. Am J Pathol 163(4):1371–1378 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1057 EP 1062 DI 10.1007/s12253-019-00658-5 PG 6 ER PT J AU Feng, Y Jiang, Y Wen, T Meng, F Shu, X AF Feng, Yueyi Jiang, Yiqing Wen, Tao Meng, Fang Shu, Xiaochen TI Identifying Potential Prognostic Markers for Muscle-Invasive Bladder Urothelial Carcinoma by Weighted Gene Co-Expression Network Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Muscle-invasive bladder urothelial carcinoma (MIBC); WGCNA; Prognostic marker; Hub gene ID Muscle-invasive bladder urothelial carcinoma (MIBC); WGCNA; Prognostic marker; Hub gene AB Muscle-invasive bladder urothelial carcinoma (MIBC) is characterized as a genetic heterogeneous cancer with a high percentage of recurrence and worse prognosis. Identify the prognostic potentials of novel genes for muscle-invasive urothelial bladder cancer could at least provide important information for early detection and clinical treatment. Weighted gene co-expression network analysis (WGCNA) algorithm, a powerful systems biology approach, was utilized to extract co-expressed gene networks from mRNA expression dataset to construct transcriptional modules in MIBC samples, which was associated with demographic and clinical traits of MIBC patients. The potential prognostic markers of MIBC were screened out in the discovery dataset and verified in an independent external validation dataset. A total of 8 co-expression modules were detected through the WGCNA algorithm in the discovery datasets based on 401 MIBC samples. One transcriptional module enriched in cell development was observed to be correlated with the MIBC prognosis in the discovery datasets (HR = 1.48, 95%CI = 1.04–2.11) and independently verified in an external dataset (HR = 3.59, 95%CI = 1.09–11.79). High expression of hub genes including discoidin domain receptor tyrosine kinase 2 (DDR2), PDZ and LIM domain 3 (PDLIM3), zinc finger protein 521 (ZNF521), methionine sulfoxide reductase B3 (MSRB3) were significantly associated with the unfavorable survival of MIBC patients. We identified and validated four novel potential biomarkers associated with prognosis of MIBC patients by constructing genes co-expression networks. The discovery of these genetic markers may provide a new target for the development of MIBC chemotherapeutic drugs. C1 [Feng, Yueyi] Medical College of Soochow University, School of Public Health, Department of Epidemiology, 215123 Suzhou, China. [Jiang, Yiqing] Harrison International Peace Hospital, Department of General Surgery, 053000 Hengshui, China. [Wen, Tao] Capital Medical University, Beijing Chao-Yang Hospital, Medical Research Centre, 100020 Beijing, China. [Meng, Fang] Chinese Academy of Medical Sciences, Centre of Systems Medicine, 100730 Beijing, China. [Shu, Xiaochen] Medical College of Soochow University, School of Public Health, Department of Epidemiology, 215123 Suzhou, China. RP Shu, X (reprint author), Medical College of Soochow University, School of Public Health, Department of Epidemiology, 215123 Suzhou, China. EM xcshu@suda.edu.cn CR Antoni S, Ferlay J, Soerjomataram I, Znaor A, Jemal A, Bray F, 2017, Bladder Cancer incidence and mortality: a global overview and recent trends. Eur Urol 71(1):96–108. , DOI 10.1016/j. eururo.2016.06.010 Kamat AM, Hahn NM, Efstathiou JA, Lerner SP, Malmstrom P-U, Choi W, Guo CC, Lotan Y, Kassouf W, 2016, Bladder cancer. Lancet 388(10061):2796–2810. , DOI 10.1016/s0140- 6736(16)30512-8 Migita T, Ueda A, Ohishi T, Hatano M, Seimiya H, Horiguchi SI, Koga F, Shibasaki F, 2017, Epithelial-mesenchymal transition promotes SOX2 and NANOG expression in bladder cancer. Lab Invest 97:567–576. , DOI 10.1038/labinvest.2017.17 Baumgart E, Cohen MS, Silva Neto B, Jacobs MA, Wotkowicz C, Rieger-Christ KM, Biolo A, Zeheb R, Loda M, Libertino JA, Summerhayes IC, 2007, Identification and prognostic significance of an epithelial-mesenchymal transition expression profile in human bladder tumors. Clinical cancer research : an official journal of the American association for. Cancer Res 13(6):1685–1694. , DOI 10.1158/1078-0432.CCR-06-2330 Grossman HB, Natale RB, Tangen CM, Speights VO, Vogelzang NJ, Trump DL, deVere White RW, Sarosdy MF, Wood DP Jr, Raghavan D, Crawford ED, 2003, Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N Engl J Med 349(9):859–866. , DOI 10. 1056/NEJMoa022148 Amir S, Mabjeesh NJ, 2017, microRNA expression profiles as decision-making biomarkers in the management of bladder cancer. Histol Histopathol 32(2):107–119. , DOI 10. 14670/HH-11-814 Dudek AM, van Kampen JGM, Witjes JA, Kiemeney L, Verhaegh GW, 2018, LINC00857 expression predicts and mediates the response to platinum-based chemotherapy inmuscle-invasive bladder cancer. Cancer Med 7:3342–3350. , DOI 10.1002/cam4. 1570 Siegel RL, Miller KD, Jemal A, 2018, Cancer statistics, 2018. CA Cancer J Clin 68(1):7–30. , DOI 10.3322/caac.21442 Kiselyov A, Bunimovich-Mendrazitsky S, Startsev V, 2016, Key signaling pathways in the muscle-invasive bladder carcinoma: clinical markers for disease modeling and optimized treatment. Int J Cancer 138(11):2562–2569. , DOI 10.1002/ijc.29918 Masson-Lecomte A, Rava M, Real FX, Hartmann A, Allory Y, Malats N, 2014, Inflammatory biomarkers and bladder cancer prognosis: a systematic review. Eur Urol 66(6):1078–1091. , DOI 10.1016/j.eururo.2014.07.033 Clarke C,Madden SF, Doolan P, Aherne ST, Joyce H, O'Driscoll L, Gallagher WM, Hennessy BT, Moriarty M, Crown J, Kennedy S, ClynesM(2013, Correlating transcriptional networks to breast cancer survival: a large-scale coexpression analysis. Carcinogenesis 34(10):2300–2308. , DOI 10.1093/carcin/bgt208 Oros Klein K, Oualkacha K, Lafond MH, Bhatnagar S, Tonin PN, Greenwood CM, 2016, Gene Coexpression analyses differentiate networks associated with diverse cancers harboring TP53 missense or null mutations. Front Genet 7:137. , DOI 10.3389/fgene. 2016.00137 Guo L, Zhang K, Bing Z, 2017, Application of a coexpression network for the analysis of aggressive and nonaggressive breast cancer cell lines to predict the clinical outcome of patients. Mol Med Rep 16(6):7967–7978. , DOI 10.3892/mmr.2017.7608 Robertson AG, Kim J, Al-Ahmadie H, Bellmunt J, Guo G, Cherniack AD, Hinoue T, Laird PW, Hoadley KA, Akbani R, Castro MAA, Gibb EA, Kanchi RS, Gordenin DA, Shukla SA, Sanchez-Vega F, Hansel DE, Czerniak BA, Reuter VE, Su X, de Sa Carvalho B, Chagas VS, Mungall KL, Sadeghi S, Pedamallu CS, Lu Y, Klimczak LJ, Zhang J, Choo C, Ojesina AI, Bullman S, LeraasKM, LichtenbergTM,Wu CJ, Schultz N,GetzG, Meyerson M, Mills GB, McConkey DJ, Network TR, Weinstein JN, Kwiatkowski DJ, Lerner SP, 2017, Comprehensive molecular characterization of muscle-invasive bladder Cancer. Cell 171(3): 540–556 e525. , DOI 10.1016/j.cell.2017.09.007 Rebouissou S, Bernard-Pierrot I, de Reynies A, Lepage ML, Krucker C, Chapeaublanc E, Herault A, Kamoun A, Caillault A, Letouze E, Elarouci N, Neuzillet Y, Denoux Y, Molinie V, Vordos D, Laplanche A,Maille P, Soyeux P, Ofualuka K, Reyal F, Biton A, Sibony M, Paoletti X, Southgate J, Benhamou S, Lebret T, Allory Y, Radvanyi F, 2014, EGFR as a potential therapeutic target for a subset of muscle-invasive bladder cancers presenting a basal-like phenotype. Sci Transl Med 6(244):244ra291. , DOI 10. 1126/scitranslmed.3008970 Mak MP, Tong P, Diao L, Cardnell RJ, Gibbons DL, William WN, Skoulidis F, Parra ER, Rodriguez-Canales J, Wistuba II, Heymach JV, Weinstein JN, Coombes KR, Wang J, Byers LA, 2016, A patient- derived, pan-Cancer EMT signature identifies global molecular alterations and immune target enrichment following epithelialto- mesenchymal transition. Clin Cancer Res 22(3):609–620. , DOI 10.1158/1078-0432.CCR-15-0876 Love MI, HuberW, Anders S, 2014, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15(12):550. , DOI 10.1186/s13059-014-0550-8 Langfelder P, Horvath S, 2008)WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9:559. , DOI 10.1186/1471-2105-9-559 Zhang B, Horvath S, 2005, A general framework for weighted gene co-expression network analysis. Stat Appl Genet Mol Biol 4: Article17. , DOI 10.2202/1544-6115.1128 Yuan L, Chen L, Qian K, Qian G,Wu CL,Wang X, Xiao Y, 2017, Co-expression network analysis identified six hub genes in association with progression and prognosis in human clear cell renal cell carcinoma, ccRCC). Genom Data 14:132–140. , DOI 10. 1016/j.gdata.2017.10.006 Yu G,Wang LG, Han Y, He QY, 2012, clusterProfiler: an R package for comparing biological themes among gene clusters. Omics : a Journal Of Integrative Biology 16(5):284–287. , DOI 10. 1089/omi.2011.0118 Achkar T, Parikh RA, 2018, Adjuvant therapy in muscle-invasive bladder Cancer and upper tract urothelial carcinoma. Urol Clin N Am 45(2):257–266. , DOI 10.1016/j.ucl.2017.12.010 Hermans TJN, Voskuilen CS, van der HeijdenMS, Schmitz-Drager BJ, Kassouf W, Seiler R, Kamat AM, Grivas P, Kiltie AE, Black PC, van Rhijn BWG, 2017, Neoadjuvant treatment for muscleinvasive bladder cancer: the past, the present, and the future. Urol Oncol 36:413–422. , DOI 10.1016/j.urolonc.2017.10.014 Tsai MC, Li WM, Huang CN, Ke HL, Li CC, Yeh HC, Chan TC, Liang PI, Yeh BW,Wu WJ, Lim SW, Li CF, 2016, DDR2 overexpression in urothelial carcinoma indicates an unfavorable prognosis: a large cohort study. Oncotarget 7(48):78918–78931. https:// doi.org/10.18632/oncotarget.12912 Corsa CA, Brenot A, GritherWR, Van Hove S, Loza AJ, Zhang K, Ponik SM, Liu Y, DeNardo DG, Eliceiri KW, Keely PJ, Longmore GD, 2016, The action of Discoidin domain receptor 2 in basal tumor cells and stromal Cancer-associated fibroblasts is critical for breast Cancer metastasis. Cell Rep 15(11):2510–2523. https:// doi.org/10.1016/j.celrep.2016.05.033 GritherWR, Divine LM,Meller EH,Wilke DJ, Desai RA, Loza AJ, Zhao P, Lohrey A, LongmoreGD, Fuh KC(2018, TWIST1 induces expression of discoidin domain receptor 2 to promote ovarian cancer metastasis. Oncogene 37(13):1714–1729. , DOI 10. 1038/s41388-017-0043-9 Kim D, Ko P, You E, Rhee S, 2014, The intracellular juxtamembrane domain of discoidin domain receptor 2, DDR2, is essential for receptor activation and DDR2-mediated cancer progression. Int J Cancer 135(11):2547–2557. , DOI 10.1002/ijc.28901 Sasaki S, UedaM, Iguchi T, Kaneko M, Nakayama H,Watanabe T, Sakamoto A,Mimori K, 2017, DDR2 expression is associated with a high frequency of peritoneal dissemination and poor prognosis in colorectal Cancer. Anticancer Res 37(5):2587–2591. https://doi. org/10.21873/anticanres.11603 Bagnall RD, Yeates L, Semsarian C, 2010, Analysis of the Z-disc genes PDLIM3 and MYPN in patients with hypertrophic cardiomyopathy. Int J Cardiol 145(3):601–602. , DOI 10.1016/j. ijcard.2010.08.004 Fagerberg L, Hallstrom BM, Oksvold P, Kampf C, Djureinovic D, Odeberg J, Habuka M, Tahmasebpoor S, Danielsson A, Edlund K, Asplund A, Sjostedt E, Lundberg E, Szigyarto CA, Skogs M, Takanen JO, Berling H, Tegel H, Mulder J, Nilsson P, Schwenk JM, Lindskog C, Danielsson F, Mardinoglu A, Sivertsson A, von Feilitzen K, Forsberg M, Zwahlen M, Olsson I, Navani S, Huss M, Nielsen J, Ponten F, UhlenM, 2014, Analysis of the human tissuespecific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteomics 13(2):397– 406. , DOI 10.1074/mcp.M113.035600 Germano G, Morello G, Aveic S, Pinazza M, Minuzzo S, Frasson C, Persano L, Bonvini P, Viola G, Bresolin S, Tregnago C, Paganin M, Pigazzi M, Indraccolo S, Basso G, 2017, ZNF521 sustains the differentiation block in MLL-rearranged acute myeloid leukemia. Oncotarget 8(16):26129–26141. , DOI 10.18632/oncotarget.15387 Spina R, Filocamo G, Iaccino E, Scicchitano S, Lupia M, Chiarella E, Mega T, Bernaudo F, Pelaggi D, Mesuraca M, Pazzaglia S, Semenkow S, Bar EE, Kool M, Pfister S, Bond HM, Eberhart CG, Steinkuhler C, Morrone G, 2013, Critical role of zinc finger protein 521 in the control of growth, clonogenicity and tumorigenic potential of medulloblastoma cells. Oncotarget 4(8):1280–1292. , DOI 10.18632/oncotarget.1176 Harder L, Eschenburg G, Zech A, Kriebitzsch N, Otto B, Streichert T, Behlich AS, Dierck K, Klingler B, Hansen A, Stanulla M, Zimmermann M, Kremmer E, Stocking C, Horstmann MA, 2013, Aberrant ZNF423 impedes B cell differentiation and is linked to adverse outcome of ETV6-RUNX1 negative B precursor acute lymphoblastic leukemia. J Exp Med 210(11):2289–2304. , DOI 10.1084/jem.20130497 Mega T, Lupia M, Amodio N, Horton SJ, Mesuraca M, Pelaggi D, Agosti V, Grieco M, Chiarella E, Spina R, Moore MA, Schuringa JJ, Bond HM, Morrone G, 2011, Zinc finger protein 521 antagonizes early B-cell factor 1 and modulates the B-lymphoid differentiation of primary hematopoietic progenitors. Cell Cycle 10(13): 2129–2139. , DOI 10.4161/cc.10.13.16045 Brot N, Weissbach L, Werth J, Weissbach H, 1981, Enzymatic reduction of protein-bound methionine sulfoxide. Proc Natl Acad Sci U S A 78(4):2155–2158 Kim HY, Gladyshev VN, 2004, Methionine sulfoxide reduction in mammals: characterization of methionine-R-sulfoxide reductases. Mol Biol Cell 15(3):1055–1064. , DOI 10. 1091/mbc.E03-08-0629 Kwak GH, Kim HY, 2017, MsrB3 deficiency induces cancer cell apoptosis through p53-independent and ER stress-dependent pathways. Arch Biochem Biophys 621:1–5. , DOI 10.1016/j. abb.2017.04.001 Kwak GH, Kim TH, Kim HY, 2017, Down-regulation of MsrB3 induces cancer cell apoptosis through reactive oxygen species production and intrinsic mitochondrial pathway activation. Biochem Biophys Res Commun 483(1):468–474. , DOI 10.1016/j. bbrc.2016.12.120 Lee E, Kwak GH, Kamble K, Kim HY, 2014, Methionine sulfoxide reductase B3 deficiency inhibits cell growth through the activation of p53-p21 and p27 pathways. Arch Biochem Biophys 547:1– 5. , DOI 10.1016/j.abb.2014.02.008 Morel AP, Ginestier C, Pommier RM, Cabaud O, Ruiz E,Wicinski J, Devouassoux-Shisheboran M, Combaret V, Finetti P, Chassot C, Pinatel C, Fauvet F, Saintigny P, Thomas E, Moyret-Lalle C, Lachuer J, Despras E, Jauffret JL, Bertucci F, Guitton J, Wierinckx A,Wang Q, Radosevic-RobinN, Penault-Llorca F, Cox DG, Hollande F, Ansieau S, Caramel J, Birnbaum D, Vigneron AM, Tissier A, Charafe-Jauffret E, Puisieux A, 2017, A stemness-related ZEB1- MSRB3 axis governs cellular pliancy and breast cancer genome stability. Nat Med 23(5):568–578. , DOI 10.1038/nm.4323 Mani SA, GuoW, LiaoMJ, Eaton EN,Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J,Weinberg RA, 2008, The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133(4): 704–715. , DOI 10.1016/j.cell.2008.03.027 Polyak K, Weinberg RA, 2009, Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer 9(4):265–273. , DOI 10.1038/nrc2620 Manzotti G, Torricelli F, Benedetta D, Lococo F, Sancisi V, Rossi G, Piana S, Ciarrocchi A, 2018, An epithelial-to-mesenchymal transcriptional switch triggers evolution of pulmonary Sarcomatoid carcinoma, PSC, and identifies Dasatinib as new therapeutic option. Clinical cancer research : an official journal of the American Association for Cancer Research. , DOI 10.1158/1078-0432.CCR-18-2364 Xie B, LinW, Ye J,Wang X, Zhang B, Xiong S, Li H, Tan G, 2015, DDR2 facilitates hepatocellular carcinoma invasion and metastasis via activating ERK signaling and stabilizing SNAIL1. J Exp Clin Cancer Res : CR 34:101. , DOI 10.1186/s13046-015-0218-6 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1063 EP 1072 DI 10.1007/s12253-019-00657-6 PG 10 ER PT J AU Shousha, S Anscombe, O McFarlane, T AF Shousha, Sami Anscombe, Oliver McFarlane, Taneisha TI All Benign and Malignant Apocrine Breast Lesions Over-Express Claudin 1 and 3 and Are Negative for Claudin 4 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast; Apocrine breast carcinoma; Triple negative carcinoma; Claudin 1; Claudin 3; Claudin 4 ID Breast; Apocrine breast carcinoma; Triple negative carcinoma; Claudin 1; Claudin 3; Claudin 4 AB Invasive apocrine carcinoma of the breast is an uncommon triple negative tumour that lacks a specific therapeutic target. Apocrine metaplasia of the breast shares common morphological features with apocrine carcinoma, and was previously found to consistently over-express claudin 1 and to lack claudin 4. This study was aimed at finding whether apocrine carcinoma, and other related apocrine breast lesions, have similar claudin profile. The immunohistochemical expression of claudin 1, 3 and 4 was studied in 11 cases of in situ and invasive apocrine breast carcinoma, 7 benign apocrine lesions and 45 consecutive morphologically non-apocrine triple negative breast carcinomas. All cases were also immunostained for Gross Cystic Disease Fluid Protein- 15 (GCDFP-15), a marker for apocrine differentiation. Apocrine breast lesions maintained their expression pattern from benign through DCIS to invasive carcinoma; all showing strong expression of claudin 1 and 3 and absence of claudin 4. The same pattern of expression was seen in 2 out of the 45 morphologically non-apocrine tumours, but both showed strong positive staining for GCDFP-15. It is concluded that all benign and malignant apocrine lesions of the breast have a consistent pattern of claudin 1, 3 and 4 expression, suggesting the presence of a specific pathway for the development of invasive apocrine carcinoma. The overexpression of claudin 1 and 3 may have therapeutic implications as targets for managing apocrine cancers. C1 [Shousha, Sami] Charing Cross Hospital and Imperial College, Department of Histopathology, Fulham Palace Road, W6 8RF London, UK. [Anscombe, Oliver] Charing Cross Hospital and Imperial College, Department of Histopathology, Fulham Palace Road, W6 8RF London, UK. [McFarlane, Taneisha] Charing Cross Hospital and Imperial College, Department of Histopathology, Fulham Palace Road, W6 8RF London, UK. RP Shousha, S (reprint author), Charing Cross Hospital and Imperial College, Department of Histopathology, W6 8RF London, UK. EM S.shousha@imperial.ac.uk CR KwonMJ, 2013, Emerging roles of claudins in human cancer. Int J Mol Sci 14:18148–18181 Bleich M, Gunzel D, 2017, Physiology, pathophysiology and clinical impact of claudins. Pflugers Arch 469:1–2 Tobaries S, Siegel PM, 2016, 15 August, The role of claudins in cancer metastasis. Oncogene. , DOI 10.1036/onc.216.289 Osanai M, Takasawa A, Murata M, Sawada N, 2017, Claudins in cancer. Pflugers Arch 469:55–67 Tokes A-M, Kulka J, Paku S, Szik A, Paska C, Novak PK et al, 2005, Claudin-1. -3 and −4 proteins and mRNA expression in benign and malignant breast lesions: a research study. Breast Cancer Res 7:R296–R305. , DOI 10.1186/bcr983 Kominsky SL, Argani P, Korz D, Evron E, Raman V, Garrett E, Rein A, Sauter G, Kallioniemi OP, Sukumar S, 2003, Loss of the tight junction protein claudin-7 correlates with histological grade in both ductal carcinoma in situ and invasive ductal cartcinoma of the breast. Oncogene 22:2021–2033 Blanchard AA, Skliris GP,Watson P,Murphy LC, Penner C, Tomes L et al, 2009, Claudin 1, 3, and 4 protein expression in ER negative breast cancer correlates with markers of the basal phenotype. Virchows Arch 454:647–656 Kulka J, Szasz AM, Nemeth Z, Madaras L, Schaff Z, Molnar IA et al, 2009, Expression of tight junction protein claudin-4 in basallike breast carcinoma. Pathol Oncol Res 15:59–64 Lanigan F, McKiernan E, Brennan DJ, Hegarty S, Millikan RC, McBryan J, Jirstrom K, Landberg G, Martin F, Duffy MJ, GallagherWM, 2009, Increased claudin-4 expression is associated with poor prognosis and high tumour grade in breast cancer. Int J Cancer 124:2088–2097 Myal Y, Leygue E, Blanchard AA, 2010 Article ID 956897, Claudin 1 in breast tumorigenesis: Revelation of a possible novel Bclaudin high^ subset of breast cancers. J Biomed Bioteh. https:// doi.org/10.1155/2010/956897 Gerhard R, Ricardo S, Albergaria A, Gomes M, Silva AR, Logullo AF, Cameselle-Teijeiro JF, Paredes J, Schmitt F, 2012, Immunohistochemical features of claudin-low intrinsic subtype in metaplastic breast carcinomas. Breast 21:354–360 Blanchard AA, Ma X, Dueck KJ, Penner C, Cooper SC, Mulhall D, Murphy LC, Leygue E, Myal Y, 2013, Cladin 1 expression in basal-like breast cancer is related to patient age. BMC Cancer 13: 268 Lu S, Singh K, Mangray S, Tavares R, Noble L, Resnick M et al, 2013, Claudin expression in high grade invasive ductal carcinoma of the breast: correlation with the molecular type. Modern Pathol 26:485–495 Van Voss MRH, van Diest PJ, Smolders YHCM, Bart J, van der Wal E, van der Groep. Distinct claudin expression characterizes BRCA1=related breast cancer. Histopathology 2014;65:814–827 Zhou B, Moodie A, Blanchard AA, Leygue E, Myal Y, 2015, Claudin 1 in breast cancer: new insights. J Clin Med 4:1960–1976 Dias K, Dvorkin-Gheva A, Hallett RM,Wu Y, Hassell J, Pond GR, Levine M, Whelan T, Bane AL, 2017, Claudin-low breast cancer, clinical & pathological characteristics. PLoS One 12:e0168669. , DOI 10.1371/journal.pone.0168669 Ma F, Ding X, Fan Y, Ying J, Zheng S, Lu N, Xu B, 2014, A CLDN1-negative phenotype predicts poor prognosis in triple negative breast cancer. PLoS One 9(11):e112765 Morohashi S, Kusumi T, Sato F, Odagiri h CH, Yoshihara S et al, 2007, Decreased expression of claudin-1 correlates with recurrence status in breast cancer. Int J Mol Med 20:139–143 Abd-Elazeem MA, Abd-Elazeem MA, 2015, Claudin 4 expression in triple negative breast cancer: correlation with androgen receptors and Ki-67 expression. Ann Diag Pathol 19:37–42 Soini Y, 2004, Claudin 2, 3, 4 and 5 in Paget’s disease and breast carcinoma. Hum Pathol 35:1531–1536 Lin X, Shang X, Manorek G, Howell SB, 2013, Regulation of the epithelial-mesenchymal transition by claudin-3 and claudi-4. PLoS One 8(6):e67496 Gromov P, Espinoza JA, Gromova I, 2015, Molecular and diagnostic features of apocrine breast lesions. Expert Rev Mol Diagn 15(8):1011–1022 Vranic S,Merchio C, Castellano I, Botta C, Scalzo MS, Bender RP et al, 2015, Immunohistochemical and molecular profiling of histologically defined apocrine carcinoma of the breast. Hum Pathol 45:1350–1359 Mills AM, Gottlieb CE, Wendroth SM, Brenin CM, Atkins KA, 2016, Pure apocrine carcinomas represent a clinicopathologically distinct androgen receptor-positive subset of triple-negative breast cancers. Am J Surg Pathol 40:1109–1116 Hoevel T, Macer R, Mundigl O, Swisshelm K, Kubbies M, 2002, Expression and targeting of the tight junction protein CLDN1 in CLDN1- negative human breast tumour cells. J Cell Physiol 191: 60–68 Moldvay J, Fabian K, Jacket M, Nemeth Z, Bogos K, Furak J et al, 2017, Claudin-1 protein expression is a good prognostic factor in non-small cell lung cacer, but only in squamous cell carcinoma cases. Pathol Oncol Res 23:151–156 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1073 EP 1078 DI 10.1007/s12253-019-00662-9 PG 6 ER PT J AU Liu, Y Liang, G Zhou, T Liu, Z AF Liu, Yang Liang, Guodong Zhou, Tingting Liu, Zengshan TI Silencing UHRF1 Inhibits Cell Proliferation and Promotes Cell Apoptosis in Retinoblastoma Via the PI3K/Akt Signalling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE UHRF1; Retinoblastoma; PI3K/Akt signalling pathway; Cell proliferation; Cell apoptosis ID UHRF1; Retinoblastoma; PI3K/Akt signalling pathway; Cell proliferation; Cell apoptosis AB This study aimed to investigate the effect of silencing ubiquitin-like with PHD and RING finger domains 1 (UHRF1) on the proliferation and apoptosis of retinoblastoma (RB) cells and to clarify the molecular mechanism of the UHRF1 gene in the development of RB. Human RBWERI-Rb-1 cells were selected and assigned into a blank group (WERI-Rb-1 cells with no transfection), NC-shRNA group (WERI-Rb-1 cells infected with NC-shRNA virus) and UHRF1-shRNA group (WERI-Rb-1 cells infected with pGC-UHRF1-shRNALV-GFP# (39–1) virus). The mRNA and protein expression of UHRF1 was detected by RT-qPCR and Western blot analysis. The effect of silencing UHRF1 on the proliferation and apoptosis of WERI-Rb-1 cells was assessed by MTT assay, EdU assay, flowcytometry, and Hoechst staining. Furthermore, the expression of cell cycle-related factor (cyclin D1), apoptosis-related factors (caspase-9, Bcl-2 and Bax), and PI3K/Akt signalling pathway-related factors (p-PI3K, PI3K, p-Akt and Akt) were measured via Western blot analysis. The RNA interference plasmid UHRF1-shRNA was successfully constructed. After WERI-Rb-1 cells were infected with UHRF1-shRNA, decreased mRNA and protein expression of UHRF1 was found. WERI-Rb-1 cells infected with UHRF1-shRNA showed inhibited proliferative ability and increased apoptosis. In the UHRF1-shRNA group, more cells arrested at the G0/G1 phase and less cells at the S and G2/M phases. WERI-Rb-1 cells infected with UHRF1-shRNA had increased expression of caspase-9 and Bax and decreased expression of Bcl-2 expression and decreased levels of p-PI3K and p-Akt. In conclusion, our study demonstrated that silencing UHRF1 could inhibit the proliferation of RB cells and promote apoptosis. The mechanism may be caused by the downregulation of the proportion of Bcl-2/Bax expression and the promotion of the expression of caspase-9 through the PI3K/Akt signalling pathway. C1 [Liu, Yang] Ministry of Education Institutes, Jilin University, College of Veterinary Medicine, Key Laboratory of Zoonosis, No. 5333 Xi’an Road, 130062 Changchun, China. [Liang, Guodong] Jilin Cancer Hospital, Department of Colorectal and Stomach Cancer Surgery, 130062 Changchun, China. [Zhou, Tingting] The Affiliated Hospital of Changchun University of Chinese Medicine, 130062 Changchun, China. [Liu, Zengshan] Ministry of Education Institutes, Jilin University, College of Veterinary Medicine, Key Laboratory of Zoonosis, No. 5333 Xi’an Road, 130062 Changchun, China. RP Liu, Z (reprint author), Ministry of Education Institutes, Jilin University, College of Veterinary Medicine, Key Laboratory of Zoonosis, 130062 Changchun, China. EM zengshanliu18@163.com CR Zhang J, Benavente CA, McEvoy J, Flores-Otero J, Ding L, Chen X, Ulyanov A, Wu G, Wilson M, Wang J, Brennan R, Rusch M, Manning AL, Ma J, Easton J, Shurtleff S, Mullighan C, Pounds S, Mukatira S, Gupta P, NealeG, Zhao D, Lu C, Fulton RS, Fulton LL, Hong X, Dooling DJ, Ochoa K, Naeve C, Dyson NJ, Mardis ER, Bahrami A, Ellison D, Wilson RK, Downing JR, Dyer MA, 2012, A novel retinoblastoma therapy from genomic and epigenetic analyses. Nature 481(7381):329–334. , DOI 10.1038/ nature10733 Jo DH, Kim JH, Cho CS, Cho YL, Jun HO, Yu YS, Min JK, Kim JH, 2014, STAT3 inhibition suppresses proliferation of retinoblastoma through down-regulation of positive feedback loop of STAT3/ miR-17-92 clusters. Oncotarget 5(22):11513–11525. https://doi. org/10.18632/oncotarget.2546 Busch M, Grosse-Kreul J, Wirtz JJ, Beier M, Stephan H, Royer- Pokora B, Metz K, Dunker N, 2017, Reduction of the tumorigenic potential of human retinoblastoma cell lines by TFF1 overexpression involves p53/caspase signaling and miR-18a regulation. Int J Cancer 141(3):549–560. , DOI 10.1002/ijc.30768 Rushlow DE,MolBM, Kennett JY, Yee S, Pajovic S, Theriault BL, Prigoda-Lee NL, Spencer C, Dimaras H, Corson TW, Pang R, Massey C, Godbout R, Jiang Z, Zacksenhaus E, Paton K, Moll AC, Houdayer C, Raizis A, Halliday W, Lam WL, Boutros PC, Lohmann D, Dorsman JC, Gallie BL, 2013, Characterisation of retinoblastomas without RB1 mutations: genomic, gene expression, and clinical studies. Lancet Oncol 14(4):327–334. , DOI 10.1016/S1470-2045(13)70045-7 Dimaras H, Corson TW, Cobrinik D,White A, Zhao J, Munier FL, Abramson DH, Shields CL, Chantada GL, Njuguna F, Gallie BL, 2015, Retinoblastoma. Nat Rev Dis Primers 1:15021. https://doi. org/10.1038/nrdp.2015.21 Yun J, Li Y, Xu CT, Pan BR, 2011, Epidemiology and Rb1 gene of retinoblastoma. Int J Ophthalmic Pract 4(1):103–109. https://doi. org/10.3980/j.issn.2222-3959.2011.01.24 Shields CL, Bianciotto CG, Jabbour P, Ramasubramanian A, Lally SE, Griffin GC, Rosenwasser R, Shields JA, 2011, Intra-arterial chemotherapy for retinoblastoma: report no. 1, control of retinal tumors, subretinal seeds, and vitreous seeds. Arch Ophthalmol 129(11):1399–1406. , DOI 10.1001/archophthalmol.2011.150 Allaman-Pillet N, Oberson A, Schorderet DF, 2015, BIRO1, a cellpermeable BH3 peptide, promotes mitochondrial fragmentation and death of retinoblastoma cells. Mol Cancer Res 13(1):86–97. , DOI 10.1158/1541-7786.MCR-14-0253 Unoki M, Daigo Y, Koinuma J, Tsuchiya E, Hamamoto R, Nakamura Y, 2010, UHRF1 is a novel diagnostic marker of lung cancer. Br J Cancer 103(2):217–222. , DOI 10.1038/sj.bjc. 6605717 Kim JK, Esteve PO, Jacobsen SE, Pradhan S, 2009, UHRF1 binds G9a and participates in p21 transcriptional regulation in mammalian cells. Nucleic Acids Res 37(2):493–505. , DOI 10.1093/ nar/gkn961 Sharif J, MutoM, Takebayashi S, Suetake I, Iwamatsu A, Endo TA, Shinga J, Mizutani-Koseki Y, Toyoda T, Okamura K, Tajima S, Mitsuya K, Okano M, Koseki H, 2007, The SRA protein Np95 mediates epigenetic inheritance by recruiting Dnmt1 to methylated DNA. Nature 450(7171):908–912. , DOI 10.1038/ nature06397 Tien AL, Senbanerjee S, Kulkarni A, Mudbhary R, Goudreau B, Ganesan S, Sadler KC, Ukomadu C, 2011, UHRF1 depletion causes a G2/M arrest, activation of DNA damage response and apoptosis. Biochem J 435(1):175–185. , DOI 10.1042/ BJ20100840 Benavente CA, Finkelstein D, Johnson DA, Marine JC, Ashery- Padan R, Dyer MA, 2014, Chromatin remodelers HELLS and UHRF1 mediate the epigenetic deregulation of genes that drive retinoblastoma tumor progression. Oncotarget 5(20):9594–9608. , DOI 10.18632/oncotarget.2468 Sadler KC, Krahn KN, Gaur NA, Ukomadu C, 2007, Liver growth in the embryo and during liver regeneration in zebrafish requires the cell cycle regulator, uhrf1. Proc Natl Acad Sci U S A 104(5):1570– 1575. , DOI 10.1073/pnas.0610774104 He H, Lee C, Kim JK, 2018, UHRF1 depletion sensitizes retinoblastoma cells to chemotherapeutic drugs via downregulation of XRCC4. Cell Death Dis 9(2):164. , DOI 10.1038/s41419- 017-0203-4 Zhang HB, Lu P, Guo QY, Zhang ZH, Meng XY, 2013, Baicalein induces apoptosis in esophageal squamous cell carcinoma cells through modulation of the PI3K/Akt pathway. Oncol Lett 5(2): 722–728. , DOI 10.3892/ol.2012.1069 Moreno F, Sinaki B, Fandino A, Dussel V, Orellana L, Chantada G, 2014, A population-based study of retinoblastoma incidence and survival in argentine children. Pediatr Blood Cancer 61(9):1610– 1615. , DOI 10.1002/pbc.25048 Boutrid H, JockovichME,Murray TG, PinaY, Feuer WJ, Lampidis TJ, Cebulla CM, 2008, Targeting hypoxia, a novel treatment for advanced retinoblastoma. Invest Ophthalmol Vis Sci 49(7):2799– 2805. , DOI 10.1167/iovs.08-1751 Smith G, Sala R, Carroll L, Behan K, Glaser M, Robins E, Nguyen QD, Aboagye EO, 2012, Synthesis and evaluation of nucleoside radiotracers for imaging proliferation. Nucl Med Biol 39(5):652– 665. , DOI 10.1016/j.nucmedbio.2011.12.002 Kan G, He H, Zhao Q, Li X, Li M, Yang H, Kim JK, 2017, Functional dissection of the role of UHRF1 in the regulation of retinoblastoma methylome. Oncotarget 8(24):39497–39511. , DOI 10.18632/oncotarget.17078 Matsushita R, Yoshino H, Enokida H, Goto Y, Miyamoto K, Yonemori M, Inoguchi S, Nakagawa M, Seki N, 2016, Regulation of UHRF1 by dual-strand tumor-suppressor microRNA-145, miR-145-5p and miR-145-3p): inhibition of bladder cancer cell aggressiveness. Oncotarget 7(19):28460–28487. , DOI 10.18632/oncotarget.8668 Fang L, Shanqu L, Ping G, Ting H, XiW, Ke D, Min L, JunxiaW, Huizhong Z, 2012, Gene therapy with RNAi targeting UHRF1 driven by tumor-specific promoter inhibits tumor growth and enhances the sensitivity of chemotherapeutic drug in breast cancer in vitro and in vivo. Cancer Chemother Pharmacol 69(4):1079– 1087. , DOI 10.1007/s00280-011-1801-y Mori T, Li Y, Hata H, Ono K, Kochi H, 2002, NIRF, a novel RING finger protein, is involved in cell-cycle regulation. Biochem Biophys Res Commun 296(3):530–536 Mori T, LiY, Hata H, KochiH(2004, NIRF is a ubiquitin ligase that is capable of ubiquitinating PCNP, a PEST-containing nuclear protein. FEBS Lett 557(1–3):209–214 Li Y, Mori T, Hata H, Homma Y, Kochi H, 2004, NIRF induces G1 arrest and associates with Cdk2. Biochem Biophys Res Commun 319(2):464–468. , DOI 10.1016/j.bbrc.2004.04.190 Mori T, Ikeda DD, Fukushima T, Takenoshita S, Kochi H, 2011, NIRF constitutes a nodal point in the cell cycle network and is a candidate tumor suppressor. Cell Cycle 10(19):3284–3299. https:// doi.org/10.4161/cc.10.19.17176 Qin Y,Wang J,GongW, ZhangM, Tang Z, Zhang J,Quan Z, 2014, UHRF1 depletion suppresses growth of gallbladder cancer cells through induction of apoptosis and cell cycle arrest. Oncol Rep 31(6):2635–2643. , DOI 10.3892/or.2014.3145 Vermeulen K, Van Bockstaele DR, Berneman ZN, 2005, Apoptosis: mechanisms and relevance in cancer. Ann Hematol 84(10):627–639. , DOI 10.1007/s00277-005-1065-x Wang B, Shen J,Wang J, 2017)UNBS5162 inhibits proliferation of human retinoblastoma cells by promoting cell apoptosis. OncoTargets and Therapy 10:5303–5309. , DOI 10.2147/ OTT.S145518 Liu D, Hou P, Liu Z, Wu G, Xing M, 2009, Genetic alterations in the phosphoinositide 3-kinase/Akt signaling pathway confer sensitivity of thyroid cancer cells to therapeutic targeting of Akt and mammalian target of rapamycin. Cancer Res 69(18):7311–7319. , DOI 10.1158/0008-5472.CAN-09-1077 Cohen Y, Merhavi-Shoham E, Avraham-Lubin BC, Savetsky M, Frenkel S, Pe'er J, Goldenberg-Cohen N, 2009, PI3K/Akt pathway mutations in retinoblastoma. Invest Ophthalmol Vis Sci 50(11): 5054–5056. , DOI 10.1167/iovs.09-3617 Cui Y, Chen X, Zhang J, Sun X, Liu H, Bai L, Xu C, Liu X, 2016, Uhrf1 controls iNKT cell survival and differentiation through the Akt-mTOR Axis. Cell Rep 15(2):256–263. , DOI 10.1016/ j.celrep.2016.03.016 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1079 EP 1088 DI 10.1007/s12253-019-00656-7 PG 10 ER PT J AU Boldrini, L Giordano, M Melfi, F Lucchi, M Fontanini, G AF Boldrini, Laura Giordano, Mirella Melfi, Franca Lucchi, Marco Fontanini, Gabriella TI Distinct Angiogenic microRNA-mRNA Expression Profiles Among Subtypes of Lung Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microRNAs; Angiogenesis; Lung adenocarcinoma subtypes; VEGF ID microRNAs; Angiogenesis; Lung adenocarcinoma subtypes; VEGF AB Adenocarcinoma (ADC) represents the most common histological type of non-small cell lung cancer (NSCLC), with a heterogeneous pattern of growth classified as lepidic, acinar, papillary, solid, and micropapillary. For ADC patients there are few available therapeutic options and a valuable therapeutic strategy is represented by angiogenesis inhibitors; however, new reliable biomarkers to identify patients with benefit from anti-angiogenic drugs are needed. We designed a panel of sixteen miRNAs together with six their mRNA targets involved in the angiogenesis pathway and expression analysis was performed by the nCounter System® (NanoString Technologies) in 88 ADC patients: 29 were predominantly lepidic (33%), 26 solid (29.5%), 22 acinar (25%), and for 11 patients the prevalent pattern was papillary (12.5%). When we compared mRNA expression levels with the different histological ADC subtypes we found a significant higher expression of VEGF in papillary and solid than in other subtypes (p = 0.008). Among 16 miRNAs that target the angiogenic mRNA, 4 were significantly downregulated in papillary/solid compared to other groups. Our data suggest a distinct angiogenic miRNA-mRNA expression profile among the subtypes of ADC, with a putative clinical application to stratify patients for anti-angiogenetic drugs. Moreover, the regulation of angiogenic mRNA factors by miRNAs could provide a novel therapeutic approach based on their expression pattern specific for distinct ADC subtypes. Further studies are needed in a larger cohort of patients to confirm our results. C1 [Boldrini, Laura] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, Via Roma 57, 56126 Pisa, Italy. [Giordano, Mirella] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, Via Roma 57, 56126 Pisa, Italy. [Melfi, Franca] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, Via Roma 57, 56126 Pisa, Italy. [Lucchi, Marco] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, Via Roma 57, 56126 Pisa, Italy. [Fontanini, Gabriella] University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, Via Roma 57, 56126 Pisa, Italy. RP Boldrini, L (reprint author), University of Pisa, Department of Molecular, Medical and Surgical Pathology and of the Critical Area, 56126 Pisa, Italy. EM laura.boldrini@med.unipi.it CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65:87–108 Travis WD, Brambilla E, Noguchi M, Nicholson A, Geisinger K, Yatabe Y, Ishikawa Y, Wistuba I, Flieder DB, Franklin W, Gazdar A, Hasleton PS, Henderson DW, Kerr KM, Petersen I, Roggli V, Thunnissen E, Tsao M, 2012, Diagnosis of lung cancer in small biopsies and cytology: implications of the 2011 International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society Classification. Arch Pathol Lab Med; Epub ahead of print TravisWD, Brambilla E, Burke AP, Marx A, Nicholson AG, 2015, WHO classification of Tumours of the lung, pleura, thymus and heart. International Agency for Research on Cancer, Lyon von der Thusen JH, Tham YS, Pattenden H et al, 2013, Prognostic significance of predominant histologic pattern and nuclear grade in resected adenocarcinoma of the lung potential parameters for a grading system. J Thorac Oncol 8:37–44 Clay TD, Do H, Sundararajan V, Moore MM, Conron M, Wright GM, McLachlan SA, Dobrovic A, Russell PA, 2014, The clinical relevance of pathologic subtypes in metastatic lung adenocarcinoma. J Thorac Oncol 9:654–663 Cha MJ, Lee HY, Lee KS, Jeong JY, Han J, Shim YM, Hwang HS, 2014, Micropapillary and solid subtypes of invasive lung adenocarcinoma: clinical predictors of histopathology and outcome. J Thorac Cardiovasc Surg 147:921–928 Urer HN, Kocaturk CI, Gunluoglu MZ, Arda N, Bedirhan MA, Fener N, Dincer SI, 2014, Relationship between lung adenocarcinoma histological subtype and patient prognosis. Ann Thorac Cardiovasc Surg 20:12–18 Iachina M, Green A, Jakobsen E, 2014, The direct and indirect impact of comorbidity on the survival of patients with non-small cell lung cancer: a combination of survival, staging and resection models with missing measurements in covariates. BMJ Open 4: e003846 Alshangiti A, Chandhoke G, Ellis PM, 2018, Antiangiogenic therapies in non-small-cell lung cancer. Curr Oncol 25(Suppl 1):S45–S58 Ferrara N, Kerbel RS, 2005, Angiogenesis as a therapeutic target. Nature 438:967–974 Wang J, Chen J, Guo Y,Wang B, Chu H, 2017, Strategies targeting angiogenesis in advanced non-small cell lung cancer. Oncotarget 8: 53854–53872 Qu J, Zhang Y, Chen X, Yang H, Zhou C, Yang N, 2017, Newly developed anti-angiogenic therapy in non-small cell lung cancer. Oncotarget 9:10147–10163 Fabian MR, Sonenberg N, Filipowicz W, 2010, Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem 79:351–379 Fontanella C, Ongaro E, Bolzonello S, Guardascione M, Fasola G, Aprile G, 2014, Clinical advances in the development of novel VEGFR2 inhibitors. Ann Transl Med 2:123 Manzo A, Montanino A, Carillio G, Costanzo R, Sandomenico C, Normanno N, Piccirillo MC, Daniele G, Perrone F, Rocco G, Morabito A, 2017, Angiogenesis inhibitors in NSCLC. Int J Mol Sci 18(10) Rajabi M, Mousa SA, 2017, The role of angiogenesis in cancer treatment. Biomedicine 5:2 Viallard C, Larrivee B, 2017, Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis 20: 409–426 Urbich C, Kuehbacher A, Dimmeler S, 2008, Role of microRNAs in vascular diseases, inflammation, and angiogenesis. Cardiovasc Res 79:581–588 Joshi P, Middleton J, Jeon YJ, Garofalo M, 2014, MicroRNAs in lung cancer. World J Methodol 4:59–72 Castro D, Moreira M, Gouveia AM, Pozza DH, De Mello RA, 2017, MicroRNAs in lung cancer. Oncotarget 8:81679–81685 Fish JE, Santoro MM, Morton SU, Yu S, Yeh RF, Wythe JD, Ivey KN, Bruneau BG, Stainier DY, Srivastava D, 2008, miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell 15: 272–284 Chen Y, Gorski DH, 2008, Regulation of angiogenesis through a microRNA, miR-130a, that down-regulates antiangiogenic homeobox genes GAX and HOXA5. Blood 111:1217–1226 Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, Richardson JA, Bassel-Duby R, Olson EN, 2008, The endothelial-specificmicroRNAmiR-126 governs vascular integrity and angiogenesis. Dev Cell 15:261–271 Anand S, Majeti BK, Acevedo LM, Murphy EA, Mukthavaram R, Scheppke L, Huang M, Shields DJ, Lindquist JN, Lapinski PE, King PD, Weis SM, Cheresh DA, 2010, MicroRNA-132- mediated loss of p120RasGAP activates the endothelium to facilitate pathological angiogenesis. Nat Med 16:909–914 Landskroner-Eiger S, Moneke I, Sessa WC, 2013, miRNAs as modulators of angiogenesis. Cold Spring Harb Perspect Med 3: a006643 Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M,Wojcik SE, Aqeilan RI, Zupo S, Dono M, Rassenti L, Alder H, Volinia S, Liu CG, Kipps TJ, Negrini M, Croce CM, 2005, miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A 102:13944–13949 Hua Z, Lv Q, YeW,Wong CK, Cai G, Gu D, Ji Y, Zhao C,Wang J, Yang BB, Zhang Y, 2006, MiRNA-directed regulation of VEGF and other angiogenic factors under hypoxia. PLoS One 1:e116 Kuehbacher A, Urbich C, Dimmeler S, 2008, Targeting microRNA expression to regulate angiogenesis. Trends Pharmacol Sci 29:12–15 Fasanaro P, D'Alessandra Y, Di Stefano V, Melchionna R, Romani S, Pompilio G, Capogrossi MC, Martelli F, 2008, MicroRNA-210 modulates endothelial cell response to hypoxia and inhibits the receptor tyrosine kinase ligand Ephrin-A3. J Biol Chem 283: 15878–15883 Suarez Y, Fernandez-Hernando C, Yu J, Gerber SA, Harrison KD, Pober JS, Iruela-Arispe ML, Merkenschlager M, Sessa WC, 2008, Dicer-dependent endothelial microRNAs are necessary for postnatal angiogenesis. Proc Natl Acad Sci U S A 105:14082–14087 Suarez Y, Sessa WC, 2009, MicroRNAs as novel regulators of angiogenesis. Circ Res 104:442–454 Ghosh A, Dasgupta D, Ghosh A, Roychoudhury S, Kumar D, Gorain M, Butti R, Datta S, Agarwal S, Gupta S, Krishna Dhali G, Chowdhury A, Schmittgen TD, Kundu GC, Banerjee S, 2017, MiRNA199a-3p suppresses tumor growth, migration, invasion and angiogenesis in hepatocellular carcinoma by targeting VEGFA, VEGFR1, VEGFR2, HGF and MMP2. Cell Death Dis 8:e2706 Rahmani F, Avan A, Hashemy SI, Hassanian SM, 2018, Role of Wnt/β-catenin signaling regulatory microRNAs in the pathogenesis of colorectal cancer. J Cell Physiol 233:811–817 Pan JY, Sun CC, Bi ZY, Chen ZL, Li SJ, Li QQ,Wang YX, Bi YY, Li DJ, 2017, miR-206/133b cluster: a weapon against lung cancer? Mol Ther Nucleic Acids 8:442–449 Korde A, Jin L, Zhang JG, Ramaswamy A, Hu B, Kolahian S, Guardela BJ, Herazo-Maya J, Siegfried JM, Stabile L, Pisani MA, Herbst RS, Kaminski N, Elias JA, Puchalski JT, Takyar SS, 2017, Lung endothelial MicroRNA-1 regulates tumor growth and angiogenesis. Am J Respir Crit Care Med 196: 1443–1455 Zhou Y, Li S, Li J, Wang D, Li Q, 2017, Effect of microRNA-135a on cell proliferation, migration, invasion, apoptosis and tumor angiogenesis through the IGF-1/PI3K/ Akt signaling pathway in non-small cell lung Cancer. Cell Physiol Biochem 42:1431–1446 Liu L, Bi N,Wu L, Ding X,Men Y, ZhouW, Li L, ZhangW, Shi S, Song Y, Wang L, 2017, MicroRNA-29c functions as a tumor suppressor by targeting VEGFA in lung adenocarcinoma. Mol Cancer 16:50 Ho CS, Noor SM, Nagoor NH, 2018, MiR-378 and MiR-1827 regulate tumor invasion, migration and angiogenesis in human lung adenocarcinoma by targeting RBX1 and CRKL, respectively. J Cancer 9:331–345 Zombori T, Nyari T, Tiszlavicz L, Palfoldi R, Csada E, Geczi T, Ottlakan A, Pecsy B, Cserni G, Furak J, 2018, The more the micropapillary pattern in stage I lung adenocarcinoma, the worse the prognosis-a retrospective study on digitalized slides. Virchows Arch 472:949–958 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1089 EP 1096 DI 10.1007/s12253-019-00664-7 PG 8 ER PT J AU de Araujo, BA Infanger Serrano, LTh Mota Pedroso, CM Mariano, VF Altemani, A Gripp, MF Crespo, NA Chone, TC AF de Araujo, Bueno Amanda Infanger Serrano, Luis Thiago Mota Pedroso, Claudia Maria Mariano, Viviane Fernanda Altemani, Albina Gripp, Mignone Flavio Crespo, Nubiato Agricio Chone, Takahiro Carlos TI Clinicopathologic Diagnostic and Prognostic Factors of Spindle Cell Carcinoma of Upper Airway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Spindle cell carcinoma; Sarcomatoid carcinoma; Head and neck cancer; Immunohistochemistry; Prognostic factors ID Spindle cell carcinoma; Sarcomatoid carcinoma; Head and neck cancer; Immunohistochemistry; Prognostic factors AB Spindle cell carcinoma (SpCC) is a rare tumor, which occurs in upper respiratory tract, mainly in larynx. This study aimed to review the clinical and pathological characteristics for diagnosis and prognosis. Retrospective cohort study. All patients with SpCC in upper respiratory tract treated for curative intent was included. All patients were reviewed in search of epithelial component and immunohistochemistry when not found. It was evaluated rate of recurrence and disease-free survival with univariate and multivariate analysis with Kaplan Meier and Cox Regression model adjusted to propensity score indexes (PSI) according to age, gender, site of tumor, stage, surgical treatment, status of margins of surgical resection, lymphatic invasion. There were 16 cases of SpCC.31% were diagnosed with light microscopy and others with immunohistochemistry for epithelial marker. Disease-free survival was higher in early stage disease in univariate and multivariate analysis, as the main prognostic factor. Surgical treatment increases in 2.54 the rate of survival. The SpCC is a rare tumor considered a highly malignant variant of squamous cell carcinoma. It has male predominance and tobacco use as risk factors. Its treatment should follow the same recommendations for squamous cell carcinoma, with surgery as the maintain treatment. Immunohistochemistry is an adjuvant important tool for diagnosis of SpCC. C1 [de Araujo, Bueno Amanda] University of Campinas, UNICAMP, Department of Otorhinolaryngology, Head Neck, 13081-970 Campinas, Sao Paulo, Brazil. [Infanger Serrano, Luis Thiago] University of Campinas, UNICAMP, Department of Otorhinolaryngology, Head Neck, 13081-970 Campinas, Sao Paulo, Brazil. [Mota Pedroso, Claudia Maria] University of Campinas, UNICAMP, Department of Otorhinolaryngology, Head Neck, 13081-970 Campinas, Sao Paulo, Brazil. [Mariano, Viviane Fernanda] UNICAMP - University of Campinas, School of Medicine, Department of PathologyCampinas, Sao Paulo, Brazil. [Altemani, Albina] UNICAMP - University of Campinas, School of Medicine, Department of PathologyCampinas, Sao Paulo, Brazil. [Gripp, Mignone Flavio] University of Campinas, UNICAMP, Department of Otorhinolaryngology, Head Neck, 13081-970 Campinas, Sao Paulo, Brazil. [Crespo, Nubiato Agricio] University of Campinas, UNICAMP, Department of Otorhinolaryngology, Head Neck, 13081-970 Campinas, Sao Paulo, Brazil. [Chone, Takahiro Carlos] University of Campinas, UNICAMP, Department of Otorhinolaryngology, Head Neck, 13081-970 Campinas, Sao Paulo, Brazil. RP de Araujo, BA (reprint author), University of Campinas, UNICAMP, Department of Otorhinolaryngology, Head Neck, 13081-970 Campinas, Brazil. EM mandibueno@hotmail.com CR Virchow V. Die Krankhaften Geschwfilste, vol 2. Berlin, Germany: A. Hirschwald; 1864-1865:181-182. Gupta R, Singh S, Hedau S, Nigam S, Das BC, Sigh I, Mandal AK, 2007, Spindle cell carcinoma of head and neck: an immunohistochemical and molecular approach to its pathogenesis. J Clin Pathol 60:472–475 Figi FA, 1933, Sarcoma of the larynx. Arch Otolaryngol Head Neck Surg 18:21–33 Gerry D, Fritsch VA, Lentsch EJ, 2014, Spindle cell carcinoma of upper aerodigestive tract: an analysis of 341 cases with comparison to conventional squamous cell carcinoma. Ann Otol Rhinol Laryngol 123(8):576–583 Olsen K, Lewis JE, Suman VJ, 1997, Spindle cell carcinoma of the larynx and hypopharynx. Otolaryngol Head Neck Surg 116(1):47–52 Bishop JA, Montgomery EA, Westra WH, 2014, Use of p40 and p63 immunohistochemistry and human papillomavirus testing as ancillary tools for the recognition of head and neck sarcomatoid carcinoma and its distinction from benign and malignant mesenchymal processes. Am J Surg Pathol 38(2):257–264. https://doi. org/10.1097/PAS.0000000000000119 Choi HR, Sturgis EM, Rosenthal DI, Luna MA, Batsakis JG, El- Naggar AK, 2003, Sarcomatoid carcinoma of the head and neck: molecular evidence for evolution and progression fromconventional squamous cell carcinomas. Am J Surg Pathol 27(9):1216–1220 Choi HR, Roberts DB, Johnigan RH, Sturgis EM, Rosenthal DI, Weber RS, Luna MA, Batsakis JG, El-Naggar AK, 2004, Molecular and clinicopathologic comparisons of head and neck squamous carcinoma variants: common and distinctive features of biological significance. Am J Surg Pathol 28(10):1299–1310 Chang NJ, Kao DS, Lee LY, Chang JW, HouMM, LamWL, Cheng MH, 2013, Sarcomatoid carcinoma in head and neck: a review of 30 years of experience–clinical outcomes and reconstructive results. Ann Plast Surg 71(Suppl 1):S1–S7. , DOI 10.1097/ SAP.0000000000000069. Lewis JS, Ritter JH, El-Mofty S, 2005 Nov, Alternative epithelial markers in sarcomatoid carcinomas of the head and neck, lung, and bladder-p63, MOC-31, and TTF-1. Mod Pathol 18(11):1471–1481 Viswanathan S, Rahman K, Pallavi S, Sachin J, Patil A, Chaturvedi P, D'Cruz A, Agarwal J, Kane SV, 2010, Sarcomatoid, spindle cell, carcinoma of the head and neck mucosal region: a clinicopathologic review of 103 cases froma tertiary referral cancer Centre. Head Neck Pathol 4(4):265–275. , DOI 10.1007/s12105-010-0204-4 Dubal PM, Marchiano E, Kam D, Dutta R, Kalyoussef E, Baredes S, Eloy JA, 2015, Laryngeal spindle cell carcinoma: a populationbased analysis of incidence and survival. Laryngoscope. 125(12): 2709–2714. , DOI 10.1002/lary.25383 Epub 2015 May 21 13. Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, 2010, Head and neck. In: Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, eds, Cancer staging handbook from the AJCC Cancer stagingmanual, 7th edn. Springer,NewYork, pp 21–100 14. Ha Lan TT, Chen SJ, Arps DP, Fullen DR, Patel RM, Siddiqui J, Carskadon S, Palanisamy N, Harms PW, 2014, Expression of the p40 isoform of p63 has high specificity for cutaneous sarcomatoid squamous cell carcinoma. J Cutan Pathol 41(11):831–838. https:// doi.org/10.1111/cup.12387 15. Virchow V. Die Krankhaften Geschwfilste, vol 2. Berlin, Germany: A. Hirschwald; 1864-1865:181-182. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1097 EP 1104 DI 10.1007/s12253-019-00654-9 PG 8 ER PT J AU Yeh, YS Chen, YT Tsai, HL Huang, ChW Ma, ChJ Su, WCh Huang, ChM Huang, MY Hu, HM Lu, ChY Wang, JY AF Yeh, Yung-Sung Chen, Yi-Ting Tsai, Hsiang-Lin Huang, Ching-Wen Ma, Cheng-Jen Su, Wei-Chih Huang, Chun-Ming Huang, Ming-Yii Hu, Huang-Ming Lu, Chien-Yu Wang, Jaw-Yuan TI Predictive Value of ERCC1, ERCC2, and XRCC Expression for Patients with Locally Advanced or Metastatic Gastric Cancer Treated with Neoadjuvant mFOLFOX-4 Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE locally advanced gastric cancer; metastatic gastric cancer; mFOLFOX-4; neoadjuvant; ERCC1; ERCC2 ID locally advanced gastric cancer; metastatic gastric cancer; mFOLFOX-4; neoadjuvant; ERCC1; ERCC2 AB The dismal outcome in patients with locally advanced or metastatic gastric cancer (GC) highlights the need for effective systemic neoadjuvant chemotherapy to improve clinical results. This study evaluated the correlation between the expression of three DNA repair genes, namely the excision repair cross-complementing group 1 (ERCC1), excision repair cross-complementing group 2 (ERCC2), and X-ray repair cross-complementing protein 1 (XRCC1) and the clinical outcome of patients with locally advanced or metastatic GC treated with mFOLFOX-4 neoadjuvant chemotherapy. Fifty-eight patients with histologically confirmed locally advanced or metastatic GC following neoadjuvant mFOLFOX-4 chemotherapy were enrolled between January 2009 and January 2018. We analyzed clinicopathological features and ERCC1, ERCC2, and XRCC1 expression to identify potential predictors of clinical response. Among the 58 patients, 16 (27.6%) were categorized into the response group (partial response) and 42 into the nonresponse group (stable disease in 24 patients and progressive disease in 18 patients). A multivariate analysis showed that ERCC1 overexpression (P = 0.003), ERCC2 overexpression (P = 0.049), and either ERCC1 or ERCC2 overexpression (P = 0.002) were independent predictors of response following mFOLFOX-4 neoadjuvant chemotherapy. Additionally, ERCC1 and ERCC2 overexpression did not only predict the response but also progression-free survival (both P < 0.05) and overall survival (both P < 0.05). ERCC1 and ERCC2 overexpression are promising predictive biomarkers for patients with locally advanced or metastatic GC receiving neoadjuvant mFOLFOX-4 chemotherapy and the potential clinical implication is mandatory for further investigation. C1 [Yeh, Yung-Sung] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Trauma and Surgical Critical Care, Department of SurgeryKaohsiung, Taiwan, Republic of China. [Chen, Yi-Ting] Kaohsiung Medical University, College of Medicine, Department of PathologyKaohsiung, Taiwan, Republic of China. [Tsai, Hsiang-Lin] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, No. 100 Tzyou 1st Road, San-Ming District, 807 Kaohsiung, Taiwan, Republic of China. [Huang, Ching-Wen] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, No. 100 Tzyou 1st Road, San-Ming District, 807 Kaohsiung, Taiwan, Republic of China. [Ma, Cheng-Jen] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, No. 100 Tzyou 1st Road, San-Ming District, 807 Kaohsiung, Taiwan, Republic of China. [Su, Wei-Chih] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, No. 100 Tzyou 1st Road, San-Ming District, 807 Kaohsiung, Taiwan, Republic of China. [Huang, Chun-Ming] Kaohsiung Medical University, College of Medicine, Graduate Institute of Clinical MedicineKaohsiung, Taiwan, Republic of China. [Huang, Ming-Yii] Kaohsiung Medical University, Faculty of Medicine, College of Medicine, Department of Radiation OncologyKaohsiung, Taiwan, Republic of China. [Hu, Huang-Ming] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Gastroenterology, Department of Internal MedicineKaohsiung, Taiwan, Republic of China. [Lu, Chien-Yu] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Gastroenterology, Department of Internal MedicineKaohsiung, Taiwan, Republic of China. [Wang, Jaw-Yuan] Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, No. 100 Tzyou 1st Road, San-Ming District, 807 Kaohsiung, Taiwan, Republic of China. RP Wang, JY (reprint author), Kaohsiung Medical University, Kaohsiung Medical University Hospital, Division of Colorectal Surgery, Department of Surgery, 807 Kaohsiung, Taiwan, Republic of China. EM cy614112@ms14.hinet.net;jayuwa@cc.kmu.edu.tw CR Yeh YS, Tsai HL, Ma CJ,Wu DC, Lu CY, Wu IC, Hou MF,Wang JY, 2015, A retrospective study of the safety and efficacy of a firstline treatment with modified FOLFOX-4 in unresectable advanced or recurrent gastric cancer patients. Chemotherapy 58:411–418 Al-Batran SE, Hartmann JT, Probst S, Schmalenberg H, Hollerbach S, Hofheinz R, Rethwisch V, Seipelt G, Homann N, Wilhelm G, Schuch G, Stoehlmacher J, Derigs HG, Hegewisch-Becker S, Grossmann J, Pauligk C, Atmaca A, Bokemeyer C, Knuth A, Jager E, Arbeitsgemeinschaft Internistische Onkologie, 2008, Phase III trial in metastatic gastroesophageal adenocarcinoma with fluorouracil, leucovorin plus either oxaliplatin or cisplatin: a study of the Arbeitsgemeinschaft Internistische Onkologie. J Clin Oncol 26:1435–1342 Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, Scarffe JH, Lofts FJ, Falk SJ, Iveson TJ, Smith DB, Langley RE, Verma M, Weeden S, Chua YJ, Trial Participants MAGIC, 2006, Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med 355:11–20 HirakawaM, Sato Y, Ohnuma H, Takayama T, Sagawa T, Nobuoka T, Harada K, Miyamoto H, Sato Y, Takahashi Y, Katsuki S, Hirayama M, Takahashi M, Ono M, Maeda M, Takada K, Hayashi T, Sato T, Miyanishi K, Takimoto R, Kobune M, Hirata K, Kato J, 2013, A phase II study of neoadjuvant combination chemotherapy with docetaxel, cisplatin, and S-1 for locally advanced resectable gastric cancer: nucleotide excision repair, NER, as potential chemoresistance marker. Cancer Chemother Pharmacol 71:789–797 Colton B, HartleyM,ManningMA, Carroll JE, Xiu J, Smaglo BG, Mikhail S, Salem ME, 2016, Exceptional Response to Systemic Therapy in Advanced Metastatic Gastric Cancer: A Case Report. Cureus 8:e457 HuangMY, Tsai HL, Lin CH, HuangCW, Ma CJ, Huang CM, Chai CY, Wang JY, 2013, Predictive value of ERCC1, ERCC2, and XRCC1 overexpression for stage III colorectal cancer patients receiving FOLFOX-4 adjuvant chemotherapy. J Surg Oncol 108: 457–464 Raymond E, Faivre S, Chaney S, Woynarowski J, Cvitkovic E, 2002, Cellular and molecular pharmacology of oxaliplatin. Mol Cancer Ther 1:227–235 Huang MY, Huang ML, Chen MJ, Lu CY, Chen CF, Tsai PC, Chuang SC, Hou MF, Lin SR, Wang JY, 2011, Multiple genetic polymorphisms in the prediction of clinical outcome of metastatic colorectal cancer patients treated with first-line FOLFOX-4 chemotherapy. Pharmacogenet Genomics 21:18–25 Zheng DL, Tang GD, Chen YN, Zhang T, Qin MB, 2016, Genetic variability of ERCC1 and ERCC2 genes involved in the nucleotide excision repair pathway influences the treatment outcome of gastric cancer. Genet Mol Res 15:gmr.15027384 Edge SB Compton, 2010, The American Joint Committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. CC. Ann Surg Oncol 17:1471–1474 Shia J, Klimstra DS, Nafa K, Offit K, Guillem JG, Markowitz AJ, Gerald WL, Ellis NA, 2005, Value of Immunohistochemical detection of DNA mismatch repair proteins in predicting germline mutation in hereditary colorectal neoplasms. Am J Surg Pathol 29:96– 104 Zhang YY, Gu KS, Wu HY, Yang F, Bu LJ, Zhao CC, Zhang YR, 2015, Correlation of ERCC1 expression in peripheral blood lymphocytes with outcomes of patients with gastric cancer treated with oxaliplatin-based adjuvant chemotherapy. Genet Mol Res 14: 15921–15929 Raymond E, Chaney S, Taamma A, Cvitkovic E, 1998, Oxaliplatin: a review of preclinical and clinical studies. Ann Oncol 9:1053–1071 Rosell R, Mendez P, Isla D, Taron M, 2007, Platinum resistance related to a functional NER pathway. J Thorac Oncol 2:1063–1066 Bai Y, Wang L, Li G, Fang X, Li Y, Yang S, 2015, Genetic variability of ERCC1 genes in NER pathway influences the treatment outcome of gastric cancer. Int J Clin Exp Pathol 8:13367–13373 Popanda O, Schattenberg T, Phong CT, Butkiewicz D, Risch A, Edler L, Kayser K, Dienemann H, Schulz V, Drings P, Bartsch H, Schmezer P, 2004, Specific combinations of DNA repair gene variants and increased risk for non-small cell lung cancer. Carcinogenesis 25:2433–2441 Xue MH, Li GY, Wu XJ, Zhang CX, Zhang CF, Zhu KX, 2015, Genetic variability of genes in NER pathway influences the treatment outcome of gastric cancer. Int J Clin Exp Pathol 8:5563–5569 Joshi MB, Shirota Y, Danenberg KD, Conlon DH, Salonga DS, Herndon JE 2nd, Danenberg PV, Harpole DH Jr, 2005, High gene expression of TS1, GSTP1, and ERCC1 are risk factors for survival in patients treated with trimodality therapy for esophageal cancer. Clin Cancer Res 11:2215–2221 Shirota Y, Stoehlmacher J, Brabender J, Xiong YP, Uetake H, Danenberg KD, Groshen S, Tsao-Wei DD, Danenberg PV, Lenz HJ, 2001, ERCC1 and thymidylate synthase mRNA levels predict survival for colorectal cancer patients receiving combination oxaliplatin and fluorouracil chemotherapy. J Clin Oncol 19:4298– 4304 Huang MY, Huang JJ, Huang CM, Lin CH, Tsai HL, Huang CW, Chai CY, Lin CY, Wang JY, 2017, Relationship Between Expression of Proteins ERCC1, ERCC2, and XRCC1 and Clinical Outcomes in Patients with Rectal Cancer Treated with FOLFOX-Based Preoperative Chemoradiotherapy. World J Surg 41:2884–2897 Simon G, Sharma S, Cantor A, Smith P, Bepler G, 2005, ERCC1 expression is a predictor of survival in resected patients with nonsmall cell lung cancer. Chest 127:978–983 Dabholkar M, Vionnet J, Bostick-Bruton FYJ, Reed E, 1994, Messenger RNA levels of XPAC and ERCC1 in ovarian cancer tissue correlate with response to platinum-based chemotherapy. J Clin Invest 94:703–708 Bellmunt J, Paz-Ares L, CuelloM, Cecere FL, Albiol S, Guillem V, Gallardo E, Carles J, Mendez P, de la Cruz JJ, Taron M, Rosell R, Baselga J, Spanish Oncology Genitourinary Group, 2007, Gene expression of ERCC1 as a novel prognostic marker in advanced bladder cancer patients receiving cisplatin-based chemotherapy. Ann Oncol 18:522–528 Fareed KR, Al-Attar A, Soomro IN, Kaye PV, Patel J, Lobo DN, Parsons SL, Madhusudan S, 2010, Tumour regression and ERCC1 nuclear protein expression predict clinical outcome in patients with gastro-oesophageal cancer treated with neoadjuvant chemotherapy. Br J Cancer 102:1600–1607 LiW, Qin J, SunYH, Liu TS, 2010)Neoadjuvant chemotherapy for advanced gastric cancer: a meta-analysis.World J Gastroenterol 16: 5621–5628 De Vita F, Giuliani F, Galizia G, Belli C, Aurilio G, Santabarbara G, Ciardiello F, Catalano G, Orditura M, 2007, Neo-adjuvant and adjuvant chemotherapy of gastric cancer. Ann Oncol 18:vi120– vi123 Mezhir JJ, Tang LH, Coit DG, 2010, Neoadjuvant therapy of locally advanced gastric cancer. J Surg Oncol 101:305–314 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1105 EP 1116 DI 10.1007/s12253-019-00666-5 PG 12 ER PT J AU Szentkereszty, M Komlosi, IZs Szucs, G Barna, G Tamasi, L Losonczy, Gy Galffy, G AF Szentkereszty, Marton Komlosi, Istvan Zsolt Szucs, Gergo Barna, Gabor Tamasi, Lilla Losonczy, Gyorgy Galffy, Gabriella TI Effect of COPD on Inflammation, Lymphoid Functions and Progression-Free Survival during First-Line Chemotherapy in Advanced Non-small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Advanced NSCLC; COPD; Inflammation; Lymphopenia; VEGF; MDSC; Neutrophils; CRP; T cell exhaustion ID Advanced NSCLC; COPD; Inflammation; Lymphopenia; VEGF; MDSC; Neutrophils; CRP; T cell exhaustion AB Chronic obstructive pulmonary disease (COPD) is a common comorbidity of non-small cell lung cancer (NSCLC). COPD is characterized by systemic inflammation and lymphocyte dysfunction, mechanisms that are also known to accelerate progression of advanced (IIIB-IV) stage NSCLC. We aimed to find out whether COPD exerts an influence on tumor induced inflammatory and lymphoid responses and progression-free survival (PFS) after first-line treatment in advanced NSCLC. Patients suffering from NSCLC (n = 95), COPD (n = 54), NSCLC+COPD (n = 80) and healthy controls (n = 60) were included. PFS, neutrophil granulocyte and lymphocyte cell counts were recorded. Serum IFNγ, TNFα, VEGF concentrations were measured by using multiplex cytometric bead-based immunoassay. Prevalence of myeloid-derived suppressor cell populations (MDSC-s), and signs of T cell exhaustion were tested by using flow cytometry. Median PFS increased in the NSCLC+COPD group compared to NSCLC patients without COPD (7.4 vs 4.9 months, p < 0.01). NSCLC+COPD patients had 1.7 times (1.2–2.4) more likely to have longer PFS compared to NSCLC patients without COPD (Cox analysis, p<0.01). Neutrophil cell counts, CRP, IFNγ and TNFα concentrations were all reduced in NSCLC+COPD (all p < 0.05 vs NSCLC). NSCLC+COPD was also associated with reduced serum IL-10 concentration and increased granzyme-B positive CD8 cell counts compared to NSCLC without COPD. The effects of VEGF and MDSC-s on systemic inflammation appeared to be blunted by COPD in patients suffering from advanced NSCLC. Concomitant COPD moderates tumor-induced inflammation and supports some effector lymphoid functions and thereby may be an independent positive predictive factor of longer PFS after first-line therapy in advanced NSCLC. C1 [Szentkereszty, Marton] Semmelweis University, Department of Pulmonology, Dios arok 1/C, H-1121 Budapest, Hungary. [Komlosi, Istvan Zsolt] Semmelweis University, Department of Pulmonology, Dios arok 1/C, H-1121 Budapest, Hungary. [Szucs, Gergo] Semmelweis University, Department of Pulmonology, Dios arok 1/C, H-1121 Budapest, Hungary. [Barna, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Tamasi, Lilla] Semmelweis University, Department of Pulmonology, Dios arok 1/C, H-1121 Budapest, Hungary. [Losonczy, Gyorgy] Semmelweis University, Department of Pulmonology, Dios arok 1/C, H-1121 Budapest, Hungary. [Galffy, Gabriella] Semmelweis University, Department of Pulmonology, Dios arok 1/C, H-1121 Budapest, Hungary. RP Losonczy, Gy (reprint author), Semmelweis University, Department of Pulmonology, H-1121 Budapest, Hungary. EM losonczygyrgy@gmail.com CR Garon EB, 2012, Issues surrounding clinical trial endpoints in solid malignancies with a focus on metastatic non-small cell lung cancer. Lung Cancer 77(3):475–481. , DOI 10.1016/j.lungcan. 2012.06.007 Mary K, Gospodarowicz BOS, Sobin LH, 2006, Lung cancer. In: Prognostic factors in cancer. 3rd edn. Wiley-Liss, New York Trajkovic-Vidakovic M, de Graeff A, Voest EE, Teunissen SCCM, 2012, Symptoms tell it all: a systematic review of the value of symptom assessment to predict survival in advanced cancer patients. Critical Reviews in Oncology Hematology 84(1):130– 148. , DOI 10.1016/j.critrevonc.2012.02.011 Teramukai S, Kitano T, Kishida Y, Kawahara M, Kubota K, Komuta K, MinatoK, Mio T, Fujita Y, Yonei T, Nakano K, Tsuboi M, Shibata K, Furuse K, FukushimaM(2009, Pretreatment neutrophil count as an independent prognostic factor in advanced non-small-cell lung cancer: an analysis of Japan multinational trial organisation LC00-03. Eur J Cancer 45(11):1950–1958. , DOI 10.1016/j.ejca.2009.01.023 Wherry EJ, Kurachi M, 2015, Molecular and cellular insights into Tcell exhaustion. Nat Rev Immunol 15(8):486–499. , DOI 10.1038/nri3862 Ray-Coquard I, Cropet C,Van GlabbekeM, Sebban C, Le Cesne A, Judson I, Tredan O, Verweij J, Biron P, Labidi I, Guastalla J-P, Bachelot T, Perol D, Chabaud S, Hogendoorn PCW, Cassier P, Dufresne A, Blays J-Y, European Org R, Treatment C, 2009, Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res 69(13):5383– 5391. , DOI 10.1158/0008-5472.can-08-3845 YaoY,YuanD, Liu H, Gu X, SongY(2013, Pretreatment neutrophil to lymphocyte ratio is associated with response to therapy and prognosis of advanced non-small cell lung cancer patients treated with first-line platinum-based chemotherapy. Cancer Immunology Immunotherapy 62(3):471–479. , DOI 10.1007/s00262-012-1347-9 Berardi R, Rinaldi S, Santoni M, Newsom-Davis T, Tiberi M, Morgese F, Caramanti M, Savini A, Ferrini C, Torniai M, Fiordoliva I, Bower M, Cascinu S, 2016, Prognostic models to predict survival in patients with advanced non-small cell lung cancer treated with first-line chemo- or targeted therapy. Oncotarget 7(18):26916–26924. , DOI 10.18632/oncotarget.8309 Kocher F, Hilbe W, Seeber A, Pircher A, Schmid T, Greil R, Auberger J, Nevinny-Stickel M, Sterlacci W, Tzankov A, Jamnig H, Kohler K, Zabernigg A, Frotscher J, Oberaigner W, Fiegl M, 2015, Longitudinal analysis of 2293 NSCLC patients: a comprehensive study from the TYROL registry. Lung Cancer 87(2):193– 200. , DOI 10.1016/j.lungcan.2014.12.006 Czyzykowski R, Nowak D, Janiak A, Wlodarczyk A, Sarniak A, Krakowska M, Potemski P, 2016, A retrospective evaluation of associations between chronic obstructive pulmonary disease, smoking, and efficacy of chemotherapy and selected laboratory parameters in patients with advanced non-small cell lung cancer. Wspolczesna Onkol 20(5): 407–413. , DOI 10.5114/wo.2016.64605 Omote N, Hashimoto N, Morise M, Sakamoto K, Miyazaki S, Ando A, Nakahara Y, Hasegawa Y, 2017, Impact of mild to moderate COPD on feasibility and prognosis in non-small cell lung cancer patients who received chemotherapy. Int J COPD 12: 3541–3547. , DOI 10.2147/copd.s149456 Izquierdo JL, Resano P, El Hachem A, Graziani D, Almonacid C, Sanchez IM, 2014, Impact of COPD in patients with lung cancer and advanced disease treatedwith chemotherapy and/or tyrosine kinase inhibitors. Int J COPD 9:1053–1058. , DOI 10.2147/copd. s68766 Arca JA, Lamelas IP, Ortega RA, Perez JB, Navarro MET, Velazquez PM, 2009, Lung Cancer and COPD: a common combination. Arch Bronconeumol 45(10):502–507. , DOI 10. 1016/j.arbres.2009.07.005 Mark NM, Kargl J, Busch SE, Yang GHY, Metz HE, Zhang HJ, Hubbard JJ, Pipavath SNJ, Madtes DK, Houghton AM, 2018, Chronic obstructive pulmonary disease alters immune cell composition and immune checkpoint inhibitor efficacy in non-small cell lung Cancer. Am J Respir Crit Care Med 197(3):325–336. https:// doi.org/10.1164/rccm.201704-0795OC Young RP, Hopkins RJ, Christmas T, Black PN, Metcalf P, Gamble GD, 2009, COPD prevalence is increased in lung cancer, independent of age, sex and smoking history. Eur Respir J 34(2):380–386. , DOI 10.1183/09031936.00144208 MacNee W VJ, Agusti A, 2016, COPD: pathogenesis and natural history. In: Murray & Nadel’s textbook of respiratory medicine. 6th edn. Elsevier Saunders, pp 751–766 Mok T, Gorbunova V, Juhasz E, Szima B, Burdaeva O, Orlov S, Yu CJ, Archer V, Hilton M, Delmar P, Pallaud C, Reck M, 2014, A correlative biomarker analysis of the combination of bevacizumab and carboplatin-based chemotherapy for advanced nonsquamous non-small-cell lung Cancer results of the phase II randomized ABIGAIL study, BO21015). J Thorac Oncol 9(6):848–855 Hu PP, Liu WD, Wang LG, Yang MM, Du JJ, 2013, High circulating VEGF level predicts poor overall survival in lung cancer. J Cancer Res Clin Oncol 139(7):1157–1167. , DOI 10.1007/ s00432-013-1425-1 Kaya A, Ciledag A, Gulbay BE, PoyrazBM, Celik G, Sen E, Savas H, Savas I, 2004, The prognostic significance of vascular endothelial growth factor levees in sera of non-small cell lung cancer patients. Respir Med 98(7):632–636. , DOI 10.1016/j.rmed. 2003.12.017 Johnson B, Osada T, Clay T, Lyerly H,MorseM(2009, Physiology and therapeutics of vascular endothelial growth factor in tumor immunosuppression. Curr Mol Med 9(6):702–707 Talmadge JE, Gabrilovich DI, 2013, History of myeloid-derived suppressor cells. Nat Rev Cancer 13(10):739–U779. https://doi. org/10.1038/nrc3581 Zarour HM, 2016, Reversing T-cell dysfunction and exhaustion in Cancer. Clin Cancer Res 22(8):1856–1864. , DOI 10.1158/ 1078-0432.ccr-15-1849 Kamphorst AO, Pillai RN, Yang S, Nasti TH, Akondy RS,Wieland A, Sica GL, Yu K, Koenig L, Patel NT, Behera M, Wu H, McCausland M, Chen Z, Zhang C, Khuri FR, Owonikoko TK, Ahmed R, Ramalingam SS, 2017, Proliferation of PD-1+CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A 114(19):4993–4998. https://doi. org/10.1073/pnas.1705327114 Huang AC, Postow MA, Orlowski RJ,Mick R, Bengsch B,Manne S, Xu W, Harmon S, Giles JR, Wenz B, Adamow M, Kuk D, Panageas KS, Carrera C, Wong P, Quagliarello F, Wubbenhorst B, D'Andrea K, Pauken KE, Herati RS, Staupe RP, Schenkel JM, McGettigan S, Kothari S, George SM, Vonderheide RH, Amaravadi RK, Karakousis GC, Schuchter LM, Xu X, Nathanson KL, Wolchok JD, Gangadhar TC, Wherry EJ, 2017, T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 545(7652):60–6+. , DOI 10.1038/nature22079 Scrimini S, Pons J, Agusti A, Clemente A, Crespi SallanM,Miquel Bauca J, Soriano JB, Cosio BG, Lopez M, Crespi C, Sauleda J, 2015, Expansion of myeloid-derived suppressor cells in chronic obstructive pulmonary disease and lung cancer: potential link between inflammation and cancer. Cancer Immunol Immunother 64(10):1261–1270. , DOI 10.1007/s00262-015-1737-x Kalathil SG, Lugade AA, Pradhan V, Miller A, Parameswaran GI, Sethi S, Thanavala Y, 2014, T-regulatory cells and programmed death 1(+, T cells contribute to effector T-cell dysfunction in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 190(1):40–50. , DOI 10.1164/rccm.201312-2293OC Quint LE, 2007, Staging non-small cell lung cancer. Cancer imaging : the official publication of the international Cancer imaging society 7:148–159. , DOI 10.1102/1470-7330.2007.0026 Buccheri G, Ferrigno D, Tamburini M, 1996, Karnofsky and ECOG performance status scoring in lung cancer: a prospective, longitudinal study of 536 patients from a single institution. Eur J Cancer 32A(7):1135–1141. , DOI 10.1016/0959-8049(95, 00664-8 http://www.goldcopd.org/Guidelines/guideline-2010-gold-report. html., 2010) Komlosi ZI, Kovacs N, van de Veen W, Kirsch A, Fahrner HB, Wawrzyniak M, Rebane A, Stanic B, Palomares O, Ruckert B, Menz G, Akdis M, Losonczy G, Akdis CA, 2017, Human CD40Lexpressing type 3 innate lymphoid cells induce IL-10-producing immature transitional regulatory B cells. J Allergy Clin Immunol 142: 178–194.e11. , DOI 10.1016/j.jaci.2017.07.046 Bronte V, Brandau S, Chen S-H, Colombo MP, Frey AB, Greten TF, Mandruzzato S, Murray PJ, Ochoa A, Ostrand-Rosenberg S, Rodriguez PC, Sica A, Umansky V, Vonderheide RH, Gabrilovich DI, 2016, Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun 7. , DOI 10.1038/ncomms12150 Voron T, Marcheteau E, Pernot S, Colussi O, Tartour E, Taieb J, Terme M, 2014, Control of the immune response by proangiogenic factors. Front Oncol 4:70. , DOI 10.3389/ fonc.2014.00070 Abdallah A, Belal M, El Bastawisy A, Gaafar R, 2014, Plasma vascular endothelial growth factor 165 in advanced non-small cell lung cancer. Oncol Lett 7(6):2121–2129. , DOI 10.3892/ol. 2014.2016 TamiyaM, Tamiya A, Yasue T, Nakao K, Omachi N, Shiroyama T, Tani E, Hamaguchi M, Morishita N, Suzuki H, Okamoto N, Okishio K, Kawaguchi T, Atagi S, Hirashima T, 2016, Vascular endothelial growth factor in plasma and pleural effusion is a biomarker for outcome after bevacizumab plus carboplatin-paclitaxel Treatment for non-small cell lung Cancer with malignant pleural effusion. Anticancer Res 36(6):2939–2944 Di Stefano A, Caramori G, Gnemmi I, Contoli M, Vicari C, Capelli A, Magno F, D'Anna SE, Zanini A, Brun P, Casolari P, Chung KF, Barnes PJ, Papi A, Adcock I, Balbi B, 2009, T helper type 17- related cytokine expression is increased in the bronchial mucosa of stable chronic obstructive pulmonary disease patients. Clin Exp Immunol 157(2):316–324. , DOI 10.1111/j.1365-2249. 2009.03965.x Wang HY, Peng WD, Weng YS, Ying HJ, Li HQ, Xia DJ, Yu WJ, 2012, Imbalance of Th17/Treg cells in mice with chronic cigarette smoke exposure. Int Immunopharmacol 14(4):504–512. https://doi. org/10.1016/j.intimp.2012.09.011 Baraldo SZM, Zuin R, Saetta M, 2007, Lymphocytes. In: Stockley RRS, Rabe K, Celli B, eds, Chronic obstructive pulmonary disease. Blackwell Publishing, pp 269–274 Hoenderdos K, Condliffe A, 2013, The neutrophil in chronic obstructive pulmonary disease too little, too late or too much, too soon? Am J Respir Cell Mol Biol 48(5):531–539. , DOI 10.1165/rcmb.2012-0492TR Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD, 2004, The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med 350(26):2645–2653. https:// doi.org/10.1056/NEJMoa032158 Ruffell B, DeNardo DG, Affara NI, Coussens LM, 2010, Lymphocytes in cancer development: polarization towards protumor immunity. Cytokine Growth Factor Rev 21(1):3–10. , DOI 10.1016/j.cytogfr.2009.11.002 Chang SH,Mirabolfathinejad SG, Katta H, Cumpian AM, Gong L, CaetanoMS, Moghaddam SJ, Dong C, 2014, T helper 17 cells play a critical pathogenic role in lung cancer. Proc Natl Acad Sci U S A 111(15):5664–5669. , DOI 10.1073/pnas.1319051111 Eide HA, Halvorsen AR, Sandhu V, Fane A, Berg J, Haakensen VD, Kure EH, Brustugun OT, Kiserud CE, Kyte JA, Helland A, 2016, Non-small cell lung cancer is characterised by a distinct inflammatory signature in serum compared with chronic obstructive pulmonary disease. Clin Transl Immunol 5:7. , DOI 10.1038/cti.2016.65 Zhai R, Yu X, Shafer A,Wain JC, Christiani DC, 2014, The impact of coexisting COPD on survival of patients with early-stage non-small cell lung Cancer undergoing surgical resection. Chest 145(2): 346–353. , DOI 10.1378/chest.13-1176 Qiang G, Liang C, Xiao F, Yu Q, Wen H, Song Z, Tian Y, Shi B, Guo Y, Liu D, 2016, Impact of chronic obstructive pulmonary disease on postoperative recurrence in patients with resected nonsmall- cell lung cancer. Int J COPD 11:43–49. , DOI 10. 2147/copd.s97411 Engstrom G, Segelstorm N, Ekberg-Aronsson M, Nilsson PM, Lindgarde F, Lofdahl CG, 2009, Plasma markers of inflammation and incidence of hospitalisations for COPD: results from a population-based cohort study. Thorax 64(3):211–215. https://doi. org/10.1136/thx.2008.102079 Bridges JP, Davis HW, Damodarasamy M, Kuroki Y, Howles G, Hui DY, McCormack FX, 2000, Pulmonary surfactant proteins a and D are potent endogenous inhibitors of lipid peroxidation and oxidative cellular injury. J Biol Chem 275(49):38848–38855. , DOI 10.1074/jbc.M005322200 Ambade VN, Sontakke AN, Barthwal MS, Tyagi R, Basannar DR, 2015, Diagnostic utility of biomarkers in COPD. Respir Care 60(12):1729–1742. , DOI 10.4187/respcare.03753 Morissette MC, Jobse BN, Thayaparan D, Nikota JK, Shen P, Labiris NR, Kolbeck R, Nair P, Humbles AA, Staempfli MR, 2014, Persistence of pulmonary tertiary lymphoid tissues and anti-nuclear antibodies following cessation of cigarette smoke exposure. Respir Res 15. , DOI 10.1186/1465-9921-15-49 Basu A, Hoerning A, Datta D, Edelbauer M, Stack MP, Calzadilla K, Pal S, Briscoe DM, 2010, Cutting edge: vascular endothelial growth factor-mediated signaling in human CD45RO(+, CD4(+, T cells promotes Akt and ERK activation and Costimulates IFNgamma production. J Immunol 184(2):545–549. , DOI 10. 4049/jimmunol.0900397 Lee Y, Kim SH, Han J-Y, Kim HT, Yun T, Lee JS, 2012, Early neutrophil-to-lymphocyte ratio reduction as a surrogate marker of prognosis in never smokers with advanced lung adenocarcinoma receiving gefitinib or standard chemotherapy as first-line therapy. J Cancer Res Clin Oncol 138(12):2009–2016. , DOI 10. 1007/s00432-012-1281-4 Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, Walsh LA, Postow MA, Wong P, Ho TS, Hollmann TJ, Bruggeman C, Kannan K, Li Y, Elipenahli C, Liu C, Harbison CT, Wang L, Ribas A, Wolchok JD, Chan TA, 2014, Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 371(23):2189–2199. , DOI 10.1056/ NEJMoa1406498 Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, Miller ML, Rekhtman N, Moreira AL, Ibrahim F, Bruggeman C, Gasmi B, Zappasodi R, Maeda Y, Sander C, Garon EB, Merghoub T, Wolchok JD, Schumacher TN, Chan TA, 2015, Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348(6230):124–128. , DOI 10.1126/science.aaa1348 de Biasi AR, Villena-Vargas J, Adusumilli PS, 2014, Cisplatininduced antitumor immunomodulation: a review of preclinical and clinical evidence. Clin Cancer Res 20(21):5384–5391. https:// doi.org/10.1158/1078-0432.ccr-14-1298 Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G, 2013, Mechanism of action of conventional and targeted anticancer therapies: reinstating Immunosurveillance. Immunity 39(1):74–88. , DOI 10.1016/j.immuni.2013.06.014 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1117 EP 1128 DI 10.1007/s12253-019-00661-w PG 12 ER PT J AU Hegedus, A Trzaskoma, L Soldos, P Tuza, K Katona, P Greger, Zs Zsarnoczky-Dulhazi, F Kopper, B AF Hegedus, Adam Trzaskoma, Lukasz Soldos, Peter Tuza, Kornelia Katona, Peter Greger, Zsolt Zsarnoczky-Dulhazi, Fanni Kopper, Bence TI Adaptation of Fatigue Affected Changes in Muscle EMG Frequency Characteristics for the Determination of Training Load in Physical Therapy for Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer treatment; physical rehabilitation; neuromuscular fatigue; Isometric; Motor unit activation ID Cancer treatment; physical rehabilitation; neuromuscular fatigue; Isometric; Motor unit activation AB Cancer patients often experience loss in body weight and also a decrease in muscle mass, which results in the reduction of physical activity and mobilization of the patient. To decelerate the loss of muscle mass, as part of the cancer treatment patients frequently undergo physical therapy and considering the physical capabilities of the patients, with moderate loads. Moreover, frequent studies also observed for cancer patients, together with the decrease in muscle mass a shift into fast-twitch muscle fibers from slow-twitch fibers. The aim of our study therefore was to determine how motor fibers behave under moderate isometric load executed until total exhaustion. 11 university students (G1), and 14 elite athletes (G2) participated in the study. 65% of the maximal voluntary contraction (MVC) was determined for the biceps brachii muscle, and with this load holding a weight, participants had to sustain a 90 deg. isometric elbow flexion in a standing posture until complete fatigue occurred. EMG activity for the biceps brachii muscle was measured and frequency analysis was performed. 3 windows were determined in the fatiguing protocol: the first (W1), middle (W2), and last (W3) 5 s, and also frequency analysis for MVC was performed (MAX) between 0 and 260 Hz with 20Hz wide frequency bands. The results indicate, that as the protocol progressed in time and the effect of fatigue increased (from W1 to W3) the activity of low frequency muscle fibers significantly increased (0-40 Hz) while activity of high frequency muscle fibers (60-260 Hz) significantly decreased for G1 and G2 groups identically. We can conclude, that training applied with constant moderate tension as fatigue increases will result in the increased activation of the lower frequency slow– twitch muscle fibers, but the increase of fatigue in the lower frequency fibers will not result in the increase in the activation level of the higher frequency fast-twitch fibers. Consequently, because as slow-twitch fibers are being used at moderate loads and even when fatigue occurs in these fibers the fast-twitch fibers will not work, higher muscle loads are needed if the aim is to activate fast-twitch fibers. Considering the shift into fast-twitch muscle fibers from slow-twitch fibers for cancer patients, in some cases if the patient’s age and physical status allows during the physical treatment, higher loads and consequently higher levels of activation might be beneficial for the retardment of loss concerning the fast-twitch fiber mass. C1 [Hegedus, Adam] University of Physical Education, Department of KinesiologyBudapest, Hungary. [Trzaskoma, Lukasz] University of Physical Education, Department of KinesiologyBudapest, Hungary. [Soldos, Peter] University of Physical Education, Department of KinesiologyBudapest, Hungary. [Tuza, Kornelia] University of Physical Education, Department of KinesiologyBudapest, Hungary. [Katona, Peter] University of Physical Education, Department of KinesiologyBudapest, Hungary. [Greger, Zsolt] University of Physical Education, Department of KinesiologyBudapest, Hungary. [Zsarnoczky-Dulhazi, Fanni] University of Physical Education, Department of KinesiologyBudapest, Hungary. [Kopper, Bence] University of Physical Education, Department of KinesiologyBudapest, Hungary. RP Kopper, B (reprint author), University of Physical Education, Department of Kinesiology, Budapest, Hungary. EM kopper.tf@gmail.com CR Muscaritoli M, Molfino A, Gioia G, Laviano A, Rossi Fanelli F, 2011, The "parallel pathway": a novel nutritional and metabolic approach to cancer patients. Intern Emerg Med 6(2):105–112 Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, Jatoi A, Loprinzi C, MacDonald N, Mantovani G, Davis M, Muscaritoli M, Ottery F, Radbruch L, Ravasco P,Walsh D,Wilcock A, Kaasa S, Baracos VE, 2011, Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 12(5): 489–495 Prado CM, Baracos VE, McCargar LJ, Reiman T, Mourtzakis M, Tonkin K, Mackey JR, Koski S, Pituskin E, Sawyer MB, 2009, Sarcopenia as a determinant of chemotherapy toxicity and time to tumor progression in metastatic breast cancer patients receiving capecitabine treatment. Clin Cancer Res 15(8):2920–2926 Tisdale MJ, 2010, Cancer cachexia. Curr Opin Gastroenterol 26(2): 146–151 Jackson W, Alexander N, Schipper M, Fig L, Feng F, Jolly S Characterization of changes in total body composition for patients with head and neck cancer undergoing chemoradiotherapy using dual-energy x-ray absorptiometry. Head Neck 36(9):1356–1362 Antoun S, Birdsell L, Sawyer MB, Venner P, Escudier B, Baracos VE, 2010, Association of skeletal muscle wasting with treatment with sorafenib in patients with advanced renal cell carcinoma: results from a placebo-controlled study. J Clin Oncol 28(6):1054–1060 MuscaritoliM, BossolaM, Aversa Z, Bellantone R, Rossi Fanelli F, 2006, Prevention and treatment of cancer cachexia: new insights into an old problem. Eur J Cancer 42(1):31–41 Muscaritoli M, Molfino A, Lucia S, Rossi Fanelli F, 2015, Cachexia: a preventable comorbidity of cancer. A T.A.R.G.E.T. approach. Crit Rev Oncol Hematol 94(2):251–259 Segal RJ, Reid RD, Courneya KS, Malone SC, Parliament MB, Scott CG, Venner PM, Quinney HA, Jones LW, D'Angelo ME, Wells GA, 2003, Resistance exercise in men receiving androgen deprivation therapy for prostate cancer. J Clin Oncol 21(9):1653–1659 CameraDM, SmilesWJ, Hawley JA, 2016, Exercise-induced skeletal muscle signaling pathways and human athletic performance. Free Radic Biol Med 98:131–143 Argiles JM, Busquets S, Lopez-Soriano FJ, Costelli P, Penna F, 2012, Are there any benefits of exercise training in cancer cachexia? J Cachexia Sarcopenia Muscle 3(2):73–76 Enoka RM(2015, Neuromechanical Basis of Kinesiology, 5th edn. Humaniknetics, Leeds Toth MJ, Callahan DM, Miller MS, Tourville TW, Hackett SB, Couch ME, Dittus K, 2016, Skeletal muscle fiber size and fiber type distribution in human cancer: effects of weight loss and relationship to physical function. Clin Nutr 35(6):1359–1365 Knudson D, 2003, Fundamentals of biomechanics, Second edn. Springer Science Buisiness Media, LLC, New York Denny-Brown D, Pennybacker JB, 1938, Fibrillation and fascilitation in voluntary muscle. Brain 61:311–344 Henneman E, Somjen G, Carpenter DO, 1965, Excitability and inhibitability of motoneurons of different sizes. J Neurophysiol 28:599–620 Luff AR, Atwood HL, 1972)Membrane properties and contraction of single muscle fibers in the mouse. Am J Physiol 222:1435–1440 Sale D Neural adaptation to strength training. In: Komi P, ed, Strength and power in sport. Blackwell Scientific Publications, London, pp 249–265 Gerdle B, Karlsson S, Crenshaw AG, Elert J, Friden J, 2000, The influences of muscle fibre proportions and areas upon EMG during maximal dynamic knee extensions. Eur J Appl Physiol 81:2–10 Wakeling JM, Syme DA, 2002)Wave properties of action potential from fast and slow motor units. Muscle Nerve 26:659–668 Hodson-Tole EF, Wakeling JM, 2007, Variations in motor unit recruitment patterns occur within and between muscles in the running rat, Rattus norvegicus). J Exp Biol 210:2333–2345 Lee SSM, Miara MB, Arnold AS, Biewener AA, Wakeling JM, 2011, EMG analysis tuned for determining the timing and level of activation in different motor units. J Electromyogr Kinesiol 21: 557–565 Horvath M, Fazekas G Mozgaskarosodas felmerese elektromiografiaval – a kineziologiai EMG. Ideggyogyaszati Szemle 56(11–12):360–369 Hermens HJ, Freriks B, Merletti R, Stegeman D, Blok J, Rau G, Disselhorst-Klug C, et al., 1999, SENIAM 8 European Recommendations for Surface ElectroMyoGraphy Results of the SENIAM project, Roessingh Research and Development, Enschede, the Netherlands Diefenthaeler F, Bini RR, VazMA, 2012, Frequency band analysis of muscle activation during cycling to exhaustion. Brasilian Journal of Kinanthropometry 14:243–253 McManus L, Hu X, Rymer WZ, Lowery MM, Suresh NL, 2015, Changes in motor unit behavior following isometric fatigue of the first dorsal interosseous muscle. J Neurophysiol 113(9):3186–3196 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1129 EP 1135 DI 10.1007/s12253-019-00668-3 PG 7 ER PT J AU Zhang, L Ren, HW Wu, QL Wu, YJ Song, X AF Zhang, Lei Ren, Hong-Wei Wu, Qi-Long Wu, Yan-Juan Song, Xiang TI The Effect of Next-Generation TKI in Non-Small Cell Lung Cancer after Failure of First-Line Treatment: a Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NSCLC; EGFR-TKIs; Pretreated patients; Meta-analysis ID NSCLC; EGFR-TKIs; Pretreated patients; Meta-analysis AB Resistance develops against first-generation tyrosine kinase inhibitors (TKIs), which target the epidermal growth factor receptor (EGFR), after a while for non-small-cell lung cancer (NSCLC). Recently, researchers have developed specific inhibitors against them. Among those inhibitors, next-generation EGFR-TKIs have gained prominence due to the greater efficacy and more favorable tolerability. Today, the efficacy of next-generation EGFR-TKIs in patients with advanced NSCLC after failure on first-generation EGFR-TKIs still remains under investigation. The aim of this meta-analysis was to systematically assess the efficacy and safety profiles of next-generation EGFR-TKIs in advanced NSCLC after failure on first-generation EGFR-TKIs. We performed a comprehensive search of the several electronic databases to September, 2018 to identify clinical trials. The primary endpoint was overall survival (OS), progression-free survival (PFS), disease controlled rate (DCR), objective response rate (ORR), and adverse events (AEs). Severe adverse events (AEs) (grade ≥ 3) based on the EGFR-TKIs were analysed. Odds Ratio (OR) along with 95% confidence interval (95% CI) were utilized for the main outcome analysis. In total, we had 3 randomized controlled trials in this analysis. The group of next-generation EGFR-TKIs was significantly improved PFS (OR = 0.34,95%CI = 0.29–0.40, P < 0.00001), as well with the ORR (OR = 10.48,95%CI = 3.87–28.34, P < 0.00001) and DCR (OR = 6.03,95%CI = 4.41–8.25, P < 0.00001), respectively. However, there is no significant difference in overall survival with next-generation EGFR-TKIs (OR = 1.05,95%CI = 0.85–1.31, P = 0.66). While, the OR for the treatment-related AEs of grade 3 or 4 (diarrhoea, rash/acne, nausea, vomiting, anemia) between the patients who received next-generation EGFR-TKIs and chemotherapy did not show safety benefit (P>0.05). Next-generation EGFR-TKIs was shown to be the better agent to achieve higher response rate and the longer PFS in NSCLC patients as the later-line therapy for previously treated patients with first generation EGFR-TKIs. While, the benefit of the OS and safety compared with the chemotherapy did not achieved. Further research is needed to develop a database of all EGFR mutations and their individual impact on the differing treatments. C1 [Zhang, Lei] The Second Hospital of Shanxi Medical University, Department of Oncology, No. 382 Wuyi Road, Xinghualing District, 030013 Taiyuan, Shanxi, China. [Ren, Hong-Wei] The Second Hospital of Shanxi Medical University, Department of Oncology, No. 382 Wuyi Road, Xinghualing District, 030013 Taiyuan, Shanxi, China. [Wu, Qi-Long] The Second Hospital of Shanxi Medical University, Department of Oncology, No. 382 Wuyi Road, Xinghualing District, 030013 Taiyuan, Shanxi, China. [Wu, Yan-Juan] The Second Hospital of Shanxi Medical University, Department of Cardio-Thoracic SurgeryTaiyuan, Shanxi, China. [Song, Xiang] The Second Hospital of Shanxi Medical University, Department of Oncology, No. 382 Wuyi Road, Xinghualing District, 030013 Taiyuan, Shanxi, China. RP Song, X (reprint author), The Second Hospital of Shanxi Medical University, Department of Oncology, 030013 Taiyuan, China. EM zhanglei790114@sina.com CR Jemal A, Bray F, Center MM et al, 2011, Global cancer statistics.[J]. CA Cancer J Clin 61(2):69 Kazaz SN, Oztop I, 2017, Treatment after first-generation epidermal growth factor receptor tyrosine kinase inhibitor resistance in non-small-cell lung Cancer[J]. Turk Thorac J 18(3):66–71 Masters GA, Temin S, Azzoli CG et al, 2017, Systemic therapy for stage IV non–small-cell lung Cancer: American Society of Clinical Oncology clinical practice guideline update[J]. J Clin Oncol 33(30): 832–837 Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, Gemma A, Harada M, Yoshizawa H, Kinoshita I, Fujita Y, Okinaga S, Hirano H,Yoshimori K, Harada T, Ogura T, Ando M, Miyazawa H, Tanaka T, Saijo Y, Hagiwara K, Morita S, Nukiwa T, North-East Japan Study Group, 2010, Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR[J]. N Engl J Med 362(25):2380–2388 Mitsudomi T, Morita S, Yatabe Y et al, 2010, Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor, WJTOG3405): an open label, randomised phase 3 trial.[J]. Lancet Oncol 11(2):104–105 Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, Palmero R, Garcia-Gomez R, Pallares C, Sanchez JM, Porta R, Cobo M, Garrido P, Longo F, Moran T, Insa A, deMarinis F, Corre R, Bover I, Illiano A, Dansin E, de Castro J, Milella M, Reguart N, Altavilla G, Jimenez U, Provencio M, Moreno MA, Terrasa J, Munoz-Langa J, Valdivia J, Isla D, Domine M, Molinier O, Mazieres J, Baize N, Garcia-Campelo R, Robinet G, Rodriguez- Abreu D, Lopez-Vivanco G, Gebbia V, Ferrera-Delgado L, Bombaron P, Bernabe R, Bearz A, Artal A, Cortesi E, Rolfo C, Sanchez-Ronco M, Drozdowskyj A, Queralt C, de Aguirre I, Ramirez JL, Sanchez JJ, Molina MA, Taron M, Paz-Ares L, Spanish Lung Cancer Group in collaboration with Groupe Francais de Pneumo-Cancerologie and Associazione Italiana Oncologia Toracica, 2012, Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer, EURTAC): a multicentre, open-label, randomised phase 3 trial.[J]. Lancet Oncol 13(3):239–246 Sos ML, Rode HB, Heynck S, Peifer M, Fischer F, Kluter S, Pawar VG, Reuter C, Heuckmann JM,Weiss J, Ruddigkeit L, Rabiller M, Koker M, Simard JR, Getlik M, Yuza Y, Chen TH, Greulich H, Thomas RK, Rauh D, 2010, Chemogenomic profiling provides insights into the limited activity of irreversible EGFR inhibitors in tumor cells expressing the T790M EGFR resistance mutation[J]. Cancer Res 70(3):868–874 Cross DAE, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, Orme JP, Finlay MRV, Ward RA, Mellor MJ, Hughes G, Rahi A, Jacobs VN, Brewer MR, Ichihara E, Sun J, Jin H, Ballard P, al-Kadhimi K, Rowlinson R, Klinowska T, Richmond GHP, Cantarini M, Kim DW, Ranson MR, Pao W, 2014, AZD9291, an irreversible EGFR TKI, overcomes T790Mmediated resistance to EGFR inhibitors in lung Cancer[J]. Cancer Discovery 4(9):1046–1061 Higgins JP, Thompson SG, 2002, Quantifying heterogeneity in a meta-analysis. Stat Med 21(11):1539–1558 Miller VA, Hirsh V, Cadranel J, Chen YM, Park K, Kim SW, Zhou C, Su WC, Wang M, Sun Y, Heo DS, Crino L, Tan EH, Chao TY, Shahidi M, Cong XJ, Lorence RM, Yang JCH, 2012, Afatinib versus placebo for patients with advanced, metastatic non-smallcell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy, LUX-lung 1): a phase 2b/3 randomised trial[J]. Lancet Oncol 13(5):528–538 Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, Shepherd FA, He Y, Akamatsu H, Theelen WS, Lee CK, Sebastian M, Templeton A, Mann H, Marotti M, Ghiorghiu S, Papadimitrakopoulou VA, AURA3 Investigators, 2017, Osimertinib or platinum-Pemetrexed in EGFR T790M-positive lung Cancer.[J]. N Engl J Med 376(7):629–640 Nie K, Zhang Z, Zhang C, Geng C, Zhang L, Xu X, Liu S,Wang S, Zhuang X, Lan K, Ji Y, 2018, Osimertinib compared docetaxelbevacizumab as third-line treatment in EGFRT790Mmutated nonsmall- cell lung Cancer[J]. Lung Cancer 121:5–11 Yu Z, Boggon TJ, Kobayashi S, Jin C, Ma PC, Dowlati A, Kern JA, Tenen DG, Halmos B, 2007, Resistance to an irreversible epidermal growth factor receptor, EGFR, inhibitor in EGFR-mutant lung cancer reveals novel treatment strategies[J]. Cancer Res 67(21): 10417–10427 Kobayashi S, Ji H, Yuza Y, Meyerson M, Wong KK, Tenen DG, Halmos B, 2005, An alternative inhibitor overcomes resistance caused by a mutation of the epidermal growth factor receptor.[J]. Cancer Res 65(16):7096–7101 Romanidou O, Landi L, Cappuzzo F, Califano R, 2016, Overcoming resistance to first/second generation epidermal growth factor receptor tyrosine kinase inhibitors and ALK inhibitors in oncogene-addicted advanced non-small cell lung cancer[J]. Therapeutic Advances in Medical Oncology 8(3):176–187 Jackman D, Pao W, Riely GJ, Engelman JA, Kris MG, Janne PA, Lynch T, Johnson BE, Miller VA, 2010, Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer.[J]. J Clin Oncol 28(2): 357–360 Pao W, Chmielecki J, 2010, Rational, biologically based treatment of EGFR-mutant non-small-cell lung cancer[J]. Nat Rev Cancer 10(11):760–774 Heigener DF, Reck M, 2011, Mutations in the epidermal growth factor receptor gene in non-small cell lung cancer: impact on treatment beyond gefitinib and erlotinib.[J]. Adv Ther 28(2):126–133 Oxnard GR, Arcila ME, Sima CS, Riely GJ, Chmielecki J, Kris MG, Pao W, Ladanyi M, Miller VA, 2011, Acquired resistance to EGFR tyrosine kinase inhibitors in EGFR mutant lung cancer: distinct natural history of patients with tumors harboring the T790M mutation[J]. Clinical Cancer Research An Official Journal of the American Association for Cancer Research 17(6):1616–1622 Zhou C, Yao LD, 2016, Strategies to improve outcomes of patients with EGRF-mutant non-small cell lung Cancer: review of the literature[J]. J Thorac Oncol 11(2):174–186 Li D, Ambrogio L, Shimamura T, Kubo S, Takahashi M, Chirieac LR, Padera RF, Shapiro GI, Baum A, Himmelsbach F, Rettig WJ, Meyerson M, Solca F, Greulich H, Wong KK, 2008, BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models[J]. Oncogene 27(34):4702–4711 Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, Dechaphunkul A, Imamura F, Nogami N, Kurata T, Okamoto I, Zhou C, Cho BC, Cheng Y, Cho EK, Voon PJ, Planchard D, Su WC, Gray JE, Lee SM, Hodge R, Marotti M, Rukazenkov Y, Ramalingam SS, FLAURA Investigators, 2018, Osimertinib in untreated EGFR-mutated advanced non-small-cell lung Cancer.[J]. N Engl J Med 378(2):113– 125 Goss G, Tsai CM, Shepherd FA et al, 2018, CNS response to osimertinib in patients with T790M-positive advanced NSCLC: pooled data from two phase II trials[J]. Ann Oncol 12(1):S440– S441 Davies RS, Nelmes DJ, Butler R, Lester JF, 2016, Non-small cell lung Cancer in South Wales: are exon 19 deletions and L858R different?[J]. Anticancer Res 36(8):4267–4271 Koyama N, Watanabe Y, Iwai Y, Kawamura R,Miwa C, Nagai Y, Hagiwara K, Koyama S, 2017, Distinct benefit of overall survival between patients with non-small-cell lung Cancer harboring EGFR exon 19 deletion and exon 21 L858R substitution[J]. Chemotherapy 62(3):151–158 Ke EE, Zhou Q, Zhang QY, Su J, Chen ZH, Zhang XC, Xu CR, Yang JJ, Tu HY, Yan HH, Zhang YC, Niu FY, Wu YL, 2017, A higher proportion of the EGFR, T790Mmutation may contribute to the better survival of patients with exon 19 deletions compared with those with L858R[J]. J Thorac Oncol 12(9):1368–1375 Solca F, Dahl G, Zoephel A, Bader G, Sanderson M, Klein C, Kraemer O, Himmelsbach F, Haaksma E, Adolf GR, 2012, Target binding properties and cellular activity of afatinib, BIBW 2992), an irreversible ErbB family blocker.[J]. Journal of Pharmacology & Experimental Therapeutics 343(2):342–350 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1137 EP 1143 DI 10.1007/s12253-019-00669-2 PG 7 ER PT J AU Celik, B Bulut, T Yalcin, DA AF Celik, Betul Bulut, Tangul Yalcin, Didem Arzu TI Tissue HE4 Expression Discriminates the Ovarian Serous Carcinoma but Not the Uterine Serous Carcinoma Patients. A New Adjunct to the Origin of the Tumor Site SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HE4; Serous; Carcinoma; Ovary; Endometrium; HER2 ID HE4; Serous; Carcinoma; Ovary; Endometrium; HER2 AB Both uterine serous carcinoma (USC) and ovarian serous carcinoma (OSC) are presented at advanced stage at the first admission and disseminated disease makes the anatomical site of the tumor origin impossible. CA125 and p53 are reliable markers that are useful for differentiating both uterine serous and ovarian serous carcinoma from their most common subtypes (endometrioid type carcinoma of ovary and uterus) but so far there is no histopathologic marker that differentiates USC from OSC. On the other hand, Trastuzumab (Herceptin) increases progression-free survival among USC patients, but not OSC patients and makes the histopathologically assigning the origin of the tumor important. So, the aim of this study was to evaluate the immunohistopathological discriminative value of the human epididymis secretory protein 4 (HE4) between OSC and USC patients. Patients with a diagnosis of OSC and UTC were enrolled. HE4 expression was evaluated by immunohistochemistry. The results were compared between groups. Of the tumor tissues studied, HE4 immunostaining was seen in the majority of ovarian serous carcinoma cases (89.65%), while endometrial serous carcinoma cases were devoid of HE4 immunostaining. HE4 immunostaining was seen in 39.1% uterine serous carcinoma cases and this difference was statistically significant (p = 0.001). Our study demonstrated for the first time the potential of HE4 expression to predict the anatomical site of tumor origin. HE4 is a novel tumor marker that differentiates USC from OSC. C1 [Celik, Betul] Antalya Training and Research Hospital, Department of PathologyAntalya, Turkey. [Bulut, Tangul] Antalya Training and Research Hospital, Department of PathologyAntalya, Turkey. [Yalcin, Didem Arzu] Health Science University, Antalya Hospital, Immunology UnitAntalya, Turkey. RP Celik, B (reprint author), Antalya Training and Research Hospital, Department of Pathology, Antalya, Turkey. EM bet_celik@yahoo.com CR Havrilesky LJ, Secord AA, Bae-Jump Vet al, 2007, Outcomes in surgical stage I uterine papillary serous carcinoma. Gynecol Oncol 105:677–682 Slomovitz BM, Burke TW, Eifel PJ et al, 2003, Uterine papillary serous carcinoma, UPSC): a single institution review of 129 cases. Gynecol Oncol 91:463–469 Soliman PT, Slomovitz BM, Broaddus RR, Sun CC, Oh JC, Eifel PJ, Gershenson DM, Lu KH, 2004 Aug, Synchronous primary cancers of the endometrium and ovary: a single institution review of 84 cases. Gynecol Oncol 94(2):456–462 Taskin OC, Onder S, Topuz S et al, 2017, A selected Immunohistochemical panel aids in differential diagnosis and prognostic stratification of subtypes of high-grade endometrial carcinoma: a Clinicopathologic and Immunohistochemical study at a single institution. Appl Immunohistochem Mol Morphol 25(10): 696–702 ZhangY, Garcia-Buitrago MT, Koru-Sengul T, Schuman S, Ganjei- Azar P, 2013 Sep, An immunohistochemical panel to distinguish ovarian from uterine serous papillary carcinomas. Int J Gynecol Pathol 32(5):476–481. , DOI 10.1097/PGP. 0b013e31826ddc4e Teplinsky E, Muggia F EGFR and HER2: is there a role in ovarian cancer? Transl Cancer Res 2015(1):4, 107–117. , DOI 10. 3978/j.issn.2218-676X.2015.01.01 Ma Q, Wang Q, Zhong D, 2015, Advances of human epididymis protein 4 in lung cancer. Zhongguo Fei Ai Za Zhi 18(3):184–186 Kirchhoff C, 1998, Molecular characterization of epididymal proteins. Rev Reprod 3(2):86–95 Kirchhoff C, Habben I, Ivell R, Krull N, 1991, A major human epididymis-specific cDNA encodes a protein with sequence homology to extracellular proteinase inhibitors. Biol Reprod 45(2):350– 357 Lu R, Sun X, Xiao R, Zhou L, Gao X, Guo L, 2012, Human epididymis protein 4, HE4, plays a key role in ovarian cancer cell adhesion and motility. Biochem Biophys Res Commun 419(2): 274–280 Lokich E, Singh RK, Han A et al, 2014, HE4 expression is associated with hormonal elements and mediated by importin-dependent nuclear translocation. Sc Rep 4:5500 Zhu YF, He LS, Zhang ZD, Huang QS, 2012, Expression of serum human epididymal secretory protein E4 at low grade and high grade serouscarcinomas. Asian Pac J Trop Med 5(12):925–930. https:// doi.org/10.1016/S1995-7645(12)60175-8 Georgakopoulos P, Mehmood S, Akalin A, Shroyer KR, 2012, Immunohistochemical localization of HE4 in benign, borderline, and malignant lesions of the ovary. Int J Gynecol Pathol 31(6): 517–523. , DOI 10.1097/PGP.0b013e31824fe269 LiX, Gao Y, Tan M, Zhuang H, Gao J,Hu Z,Wang H, Zhu L, Liu J, Lin B, 2015, Expression of HE4 in Endometrial Cancer and Its Clinical Significance. Biomed Res Int 2015:437468. https://doi. org/10.1155/2015/437468 Bulut T, Celik B, Yalcin AD, Keser S, 2017, Tissue expression of HE4 and its correlation with CA125 and p53 in high grade serous ovarian carcinoma. Eur J Gyneacol Oncol 38(5):745–749. https:// doi.org/10.12892/ejgo3992.2017 Bulut T, Celik B, 2018, Tissue HE4 expression distinguishes pulmonary adenocarcinoma from squamous cell carcinoma and small cell carcinoma. Correlation with TTF-1 and Napsin-A. Anal Quant Cytopathol Histopathol 40(1):1–8 Celik B, Bulut T, Yalcin AD, 2018, The expression of human epididymis protein 4a, HE4, in the Normal gastric epithelia and its role in the development of intestinal metaplasia and gastric Cancer. J Adv Med Med Res 27(2):1–10. , DOI 10.9734/ JAMMR/2018/41815 Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(1):7–30. , DOI 10.3322/caac.21387 Boruta DM 2nd, Gehrig PA, Fader AN, Olawaiye AB, 2009, Management of women with uterine papillary serous cancer: a Society of Gynecologic Oncology, SGO, review. Gynecol Oncol 115(1):142–153. , DOI 10.1016/j.ygyno.2009.06.011 Lim D, Murali R, Murray MP, Veras E, Park KJ, Soslow RA, 2016, Morphological and Immunohistochemical reevaluation of tumors initially diagnosed as ovarian Endometrioid carcinoma with emphasis on high-grade tumors. Am J Surg Pathol 40(3):302–312. https:// doi.org/10.1097/PAS.0000000000000550 Gultekin M, Dundar S, Kucukyildiz I, KaracaMZ, Boztas G, Turan SH, Hacikamiloglu E, Keskinkilic B, 2017, Survival of gynecological cancers in Turkey: where are we at? J Gynecol Oncol 28(6): e85. , DOI 10.3802/jgo.2017.28.e85 Zhu YF, Gao GL, Tang SB, Zhang ZD, Huang QS, 2013, Effect of WFDC 2 silencing on the proliferation, motility and invasion of human serous ovarian cancer cells in vitro. Asian Pac J Trop Med 6(4):265–272. , DOI 10.1016/S1995-7645(13)60055-3 Kong X, Chang X, Cheng H, Ma R, Ye X, Cui H, 2014, Human epididymis protein 4 inhibits proliferation of human ovarian cancer cells via the mitogen-activated protein kinase and phosphoinositide 3-kinase/AKT pathways. Int J Gynecol Cancer 24(3):427–436 Zhou L, Xiao R, Chen Y, Zhang J, Lu RQ, Guo L, 2013, Effect of down-regulation of HE4 gene expression on biologic behavior of ovarian cancer cells. Zhonghua Bing Li Xue Za Zhi 42(10):687– 690 Gao L, Cheng HY, Dong L, Ye X, Liu YN, Chang XH, Cheng YX, Chen J, Ma RQ, Cui H, 2011, The role of HE4 in ovarian cancer: inhibiting tumour cell proliferation and metastasis. J Int Med Res 39(5):1645–1660 Zou SL, Chang XH, Ye X et al, 2011, Effect of human epididymis protein 4 gene silencing on the malignant phenotype in ovarian cancer. Chin Med J 124(19):3133–3140 Devan SM, Pailoor J, Sthaneshwar P, Narayanan V, 2013, Pattern of tissue expression of CA-125 and HE4 in primary epithelial ovarian tumours and correlation with serum CA-125 levels. Asian Pac J Cancer Prev 14(8):4545–4548 Li J, Chen H, Mariani A et al, 2013, HE4, WFDC2, Promotes Tumor Growth in Endometrial Cancer Cell Lines. Int J Mol Sci 14(3):6026–6043. , DOI 10.3390/ijms14036026 Kemik P, Saatli B, Yildirim N, Kemik VD, Deveci B, Terek MC, Kocturk S, Koyuncuoglu M, Saygili U, 2016, Diagnostic and prognostic values of preoperative serum levels of YKL-40, HE-4 and DKK-3 in endometrial cancer. Gynecol Oncol 140(1):64–69. , DOI 10.1016/j.ygyno.2015.11.020 Minar L, Klabenesova I, Jandakova E, 2015, The importance of HE4 in differential diagnosis of endometrial cancer. Ceska Gynekol 80(4):256–263 Abbink K, Zusterzeel PL, Geurts-Moespot AJ et al, 2018, HE4 is superior to CA125 in the detection of recurrent disease in high-risk endometrial cancer p a t i e n t s . Tumour Bi o l 40(2): 1010428318757103. , DOI 10.1177/1010428318757103 Mutz-Dehbalaie I, Egle D, Fessler S et al, 2012, HE4 is an independent prognostic marker in endometrial cancer patients. Gynecol Oncol 126(2):186–191 Angioli R, Capriglione S, Scaletta G, Aloisi A, Miranda A, de Cicco Nardone C, Terranova C, Plotti F, 2016, The role of HE4 in endometrial cancer recurrence: how to choose the optimal follow-up program. Tumour Biol 37(4):4973–4978 Acs G, Pasha T, Zhang PJ, 2004, WT1 is differentially expressed in serous, endometrioid, clear cell, and mucinous carcinomas of the peritoneum, fallopian tube, ovary, and endometrium. Int J Gynecol Pathol 23:110–118 Zorn KK, Bonome T, Gangi L, Chandramouli GV, Awtrey CS, Gardner GJ, Barrett JC, Boyd J, Birrer MJ, 2005, Gene expression profiles of serous, endometrioid, and clear cell subtypes of ovarian and endometrial cancer. Clin Cancer Res 11:6422–6430 Fadare O, James S, Desouki MM, Khabele D, 2013, Coordinate patterns of estrogen receptor, progesterone receptor, and Wilms tumor 1 expression in the histopathologic distinction of ovarian from endometrial serous adenocarcinomas. Ann Diagn Pathol 17(5):430–433 Santin AD, Bellone S, Van Stedum S et al, 2005, Determination of HER2/neu status in uterine serous papillary carcinoma: comparative analysis of immunohistochemistry and fluorescence in situ hybridization. Gynecol Oncol 98(1):24–30 Buza N, Roque DM, Santin AD, 2014, HER2/neu in endometrial cancer: a promising therapeutic target with diagnostic challenges. Arch Pathol Lab Med 138(3):343–350 Fader AN, Roque DM, Siegel E et al, 2018, Randomized phase II trial of carboplatin-paclitaxel versus carboplatin-paclitaxel- Trastuzumab in uterine serous carcinomas that overexpress human epidermal growth factor receptor 2/neu. J Clin Oncol 36(20):2044– 2051. , DOI 10.1200/JCO.2017.76.5966 Serrano-Olvera A, Duenas-Gonzalez A, Gallardo-Rincon D, Candelaria M, de la Garza-Salazar J, 2006, Prognostic, predictive and therapeutic implications of HER2 in invasive epithelial ovarian cancer. Cancer Treat Rev 32:180–190 Mohammed RAA, Makboul R, Elsers DAH, Elsaba TMAM, Thalab AMAB, Shaaban OM, 2017, Pattern of HER-2 gene amplification and protein expression in benign, borderline, and malignant ovarian serous and mucinous neoplasms. Int J Gynecol Pathol 36(1):50–57 Bookman MA, Darcy KM, Clarke-Pearson D, Boothby RA, Horowitz IR, 2003, Evaluation of monoclonal humanized anti- HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the Gynecologic Oncology Group. J Clin Oncol 21:283–290 Jia L, Yuan Z, Wang Y, Cragun JM, Kong B, Zheng W, 2015, Primary sources of pelvic serous cancer in patients with endometrial intraepithelial carcinoma. Mod Pathol 28(1):118–127. https://doi. org/10.1038/modpathol.2014.76 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1145 EP 1151 DI 10.1007/s12253-019-00675-4 PG 7 ER PT J AU Ren, ZQ Yan, WJ Zhang, XZ Zhang, Pb Zhang, Ch Chen, ShK AF Ren, Ze-Qiang Yan, Wen-Jing Zhang, Xiu-Zhong Zhang, Peng-bo Zhang, Chong Chen, Shou-Kun TI CUL1 Knockdown Attenuates the Adhesion, Invasion, and Migration of Triple-Negative Breast Cancer Cells via Inhibition of Epithelial-Mesenchymal Transition SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CUL1; Breast Cancer; Epithelial-mesenchymal transition; Akt; GSK3β; EGFR ID CUL1; Breast Cancer; Epithelial-mesenchymal transition; Akt; GSK3β; EGFR AB Cullin-1 (CUL1) is an important factor for tumor growth and a potential therapeutic target for breast cancer therapy, but the molecular mechanism in triple-negative breast cancer (TNBC) is unknown. In the present study, CUL1 shRNA was transfected into BT549 and MDA-MB-231 breast cancer cells. Cell morphology, adhesion, invasion, and migration assays were carried out in the CUL1 knockdown cells. Additionally, protein expression levels of epithelial-mesenchymal transition (EMT)-related factors, Akt phosphorylation at S473 (pAkt), glycogen synthase kinase-3β phosphorylation at ser9 (pGSK3β), cytoplasmic and nuclear β-catenin, and epidermal growth factor receptor phosphorylation at Tyr1068 (pEGFR) were detected by Western blot analysis. CUL1 knockdown significantly suppressed the adhesion, invasion and migration capabilities of the cells, and decreased the expression of Snail1/2, ZEB1/2, Twist1/2, Vimentin, and increased the expression of Cytokeratin 18 (CK18). Moreover, CUL1 knockdown significantly downregulated the phosphorylated levels of Akt, GSK3β, and EGFR, inhibiting the translocation of β-catenin from the cytoplasm to the nucleus. The results indicate that CUL1 knockdown prohibited the metastasis behaviors of breast cancer cells through downregulation (dephosphorylation) of the EMT signaling pathways of EGFR and Akt/GSK3β/β-catenin in breast cancer. These results strongly suggested that reinforcement of the EMT might be a key for CUL1 to accelerate TNBC metastasis. C1 [Ren, Ze-Qiang] Affiliated Hospital of Xuzhou Medical University, General Surgery, 99 West Huaihai Road, 221006 Xuzhou, China. [Yan, Wen-Jing] Xuzhou Medical University, School of Nursing, 209 Tongshan Road, 221004 Xuzhou, China. [Zhang, Xiu-Zhong] Affiliated Hospital of Xuzhou Medical University, General Surgery, 99 West Huaihai Road, 221006 Xuzhou, China. [Zhang, Peng-bo] Affiliated Hospital of Xuzhou Medical University, General Surgery, 99 West Huaihai Road, 221006 Xuzhou, China. [Zhang, Chong] Affiliated Hospital of Xuzhou Medical University, General Surgery, 99 West Huaihai Road, 221006 Xuzhou, China. [Chen, Shou-Kun] Affiliated Hospital of Xuzhou Medical University, General Surgery, 99 West Huaihai Road, 221006 Xuzhou, China. RP Ren, ZQ (reprint author), Affiliated Hospital of Xuzhou Medical University, General Surgery, 221006 Xuzhou, China. EM rzq0805@163.com CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108. , DOI 10.3322/caac.21262 Carey LA, PerouCM, Livasy CA, Dressler LG, Cowan D, Conway K, Karaca G, Troester MA, Tse CK, Edmiston S, Deming SL, Geradts J, Cheang MC, Nielsen TO, Moorman PG, Earp HS, Millikan RC, 2006, Race, breast cancer subtypes, and survival in the Carolina breast Cancer study. JAMA 295(21):2492–2502. , DOI 10.1001/jama.295.21.2492 Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D, 2000, Molecular portraits of human breast tumours. Nature 406(6797):747–752. https:// doi.org/10.1038/35021093 Sarikas A, Hartmann T, Pan ZQ, 2011, The cullin protein family. Genome Biol 12(4):220. , DOI 10.1186/gb-2011-12-4-220 Nakayama KI, Nakayama K, 2006, Ubiquitin ligases: cell-cycle control and cancer. Nat Rev Cancer 6(5):369–381. , DOI 10.1038/nrc1881 Chen GD, Li G, 2010, Increased Cul1 expression promotes melanoma cell proliferation through regulating p27 expression. Int J Oncol 37(5):1339–1344. , DOI 10.3892/Ijo_00000786 Zhang Q, Chen Q, Lu X, Zhou Z, Zhang H, Lin HY, Duan E, Zhu C, Tan Y,Wang H, 2013, CUL1 promotes trophoblast cell invasion at the maternal-fetal interface. Cell Death Dis 4. ARTN e502. , DOI 10.1038/cddis.2013.1 Xu M, Yang X, Zhao J, Zhang J, Zhang S, Huang H, Liu Y, Liu J, 2014, High expression of Cullin1 indicates poor prognosis for NSCLC patients. Pathol Res Pract 210(7):397–401. https://doi. org/10.1016/j.prp.2014.01.015 Bai J, Zhou Y, Chen GD, Zeng JY, Ding JJ, Tan YF, Zhou JW, Li G, 2011, Overexpression of Cullin1 is associated with poor prognosis of patients with gastric cancer. Hum Pathol 42(3):375–383. https:// doi.org/10.1016/j.humpath.2010.09.003 Bai J, Yong HM, Chen FF, Mei PJ, Liu H, Li C, Pan ZQ, Wu YP, Zheng JN, 2013, Cullin1 is a novel marker of poor prognosis and a potential therapeutic target in human breast cancer. Ann Oncol 24(8):2016–2022. , DOI 10.1093/annonc/mdt147 Zhang J, Liu D, Feng Z,Mao J, Zhang C, LuY, Li J, ZhangQ, Li Q, Li L, 2016, MicroRNA-138 modulates metastasis and EMT in breast cancer cells by targeting vimentin. Biomed Pharmacother 77:135–141. , DOI 10.1016/j.biopha.2015.12.018 Jung H, Kim B, Moon BI, Oh ES, 2016, Cytokeratin 18 is necessary for initiation of TGF-beta1-induced epithelial-mesenchymal transition in breast epithelial cells. Mol Cell Biochem 423(1–2): 21–28. , DOI 10.1007/s11010-016-2818-7 Avizienyte E, Frame MC, 2005, Src and FAK signalling controls adhesion fate and the epithelial-to-mesenchymal transition. Curr Opin Cell Biol 17(5):542–547. , DOI 10.1016/j.ceb.2005.08.007 Janda E, Lehmann K, Killisch I, Jechlinger M, Herzig M, Downward J, Beug H, Grunert S, 2002, Ras and TGF[beta] cooperatively regulate epithelial cell plasticity and metastasis: dissection of Ras signaling pathways. J Cell Biol 156(2):299–313. https://doi. org/10.1083/jcb.200109037 Yan D, Avtanski D, Saxena NK, Sharma D, 2012, Leptin-induced epithelial-mesenchymal transition in breast cancer cells requires beta-catenin activation via Akt/GSK3- and MTA1/Wnt1 proteindependent pathways. J Biol Chem 287(11):8598–8612. https:// doi.org/10.1074/jbc.M111.322800 Zhao J, Ou B, Han D, Wang P, Zong Y, Zhu C, Liu D, Zheng M, Sun J, Feng H, Lu A, 2017, Tumor-derived CXCL5 promotes human colorectal cancer metastasis through activation of the ERK/Elk-1/snail and AKT/GSK3beta/beta-catenin pathways. Mol Cancer 16(1):70. , DOI 10.1186/s12943-017-0629-4 Kenny PA, Bissell MJ, 2007, Targeting TACE-dependent EGFR ligand shedding in breast cancer. J Clin Invest 117(2):337–345. , DOI 10.1172/JCI29518 Buchholz TA, Tu X, Ang KK, Esteva FJ, Kuerer HM, Pusztai L, Cristofanilli M, Singletary SE, Hortobagyi GN, Sahin AA, 2005, Epidermal growth factor receptor expression correlates with poor survival in patients who have breast carcinoma treated with doxorubicin-based neoadjuvant chemotherapy. Cancer 104(4): 676–681. , DOI 10.1002/cncr.21217 Davison Z, de Blacquiere GE,Westley BR,May FE, 2011, Insulinlike growth factor-dependent proliferation and survival of triplenegative breast cancer cells: implications for therapy. Neoplasia 13(6):504–515 Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V, 2007, Descriptive analysis of estrogen receptor, ER)-negative, progesterone receptor, PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a populationbased study from the California cancer registry. Cancer 109(9): 1721–1728. , DOI 10.1002/cncr.22618 LiX, MaQ, XuQ, Liu H, Lei J, Duan W, BhatK, Wang F, Wu E, Wang Z, 2012, SDF-1/CXCR4 signaling induces pancreatic cancer cell invasion and epithelial-mesenchymal transition in vitro through non-canonical activation of hedgehog pathway. Cancer Lett 322(2): 169–176. , DOI 10.1016/j.canlet.2012.02.035 Wu J, Liu W, Williams JP, Ratner N, 2017, EGFR-Stat3 signalling in nerve glial cells modifies neurofibroma initiation. Oncogene 36(12):1669–1677. , DOI 10.1038/onc.2016.386 Liu Y, Ji R, Li J, Gu Q, Zhao X, Sun T, Wang J, Li J, Du Q, Sun B, 2010, Correlation effect of EGFR and CXCR4 and CCR7 chemokine receptors in predicting breast cancer metastasis and prognosis. J Exp Clin Cancer Res 29:16. , DOI 10.1186/1756-9966-29-16 Yamashita N, Tokunaga E, Kitao H, Hisamatsu Y, Taketani K, Akiyoshi S, Okada S, Aishima S, Morita M, Maehara Y, 2013, Vimentin as a poor prognostic factor for triple-negative breast cancer. J Cancer Res Clin 139(5):739–746. , DOI 10.1007/ s00432-013-1376-6 Jeong H, Ryu YJ, An J, Lee Y, Kim A, 2012, Epithelialmesenchymal transition in breast cancer correlates with high histological grade and triple-negative phenotype. Histopathology 60(6B):E87–E95. , DOI 10.1111/j.1365-2559.2012. 04195.x Ghahhari NM, Babashah S, 2015, Interplay between microRNAs and WNT/beta-catenin signalling pathway regulates epithelialmesenchymal transition in cancer. Eur J Cancer 51(12):1638– 1649. , DOI 10.1016/j.ejca.2015.04.021 Weyemi U, Redon CE, Sethi TK, Burrell AS, Jailwala P, Kasoji M, Abrams N, Merchant A, Bonner WM, 2016, Twist1 and slug mediate H2AX-regulated epithelial-mesenchymal transition in breast cells. Cell Cycle 15(18):2398–2404. , DOI 10.1080/ 15384101.2016.1198864 Pan Y, Zhang J, Fu H, Shen L, 2016, miR-144 functions as a tumor suppressor in breast cancer through inhibiting ZEB1/2-mediated epithelial mesenchymal transition process. Onco Targets Ther 9: 6247–6255. , DOI 10.2147/OTT.S103650 Kokkinos MI,Wafai R,Wong MK, Newgreen DF, Thompson EW, WalthamM(2007, Vimentin and epithelial-mesenchymal transition in human breast cancer–observations in vitro and in vivo. Cells Tissues Organs 185(1–3):191–203. , DOI 10.1159/ 000101320 Kumar A, Pandurangan AK, Lu F, Fyrst H, Zhang M, Byun HS, Bittman R, Saba JD, 2012, Chemopreventive sphingadienes downregulate Wnt signaling via a PP2A/Akt/ GSK3beta pathway in colon cancer. Carcinogenesis 33(9): 1726–1735. , DOI 10.1093/carcin/bgs174 Fang D, Hawke D, Zheng Y, Xia Y, Meisenhelder J, Nika H, Mills GB, Kobayashi R, Hunter T, Lu Z, 2007, Phosphorylation of betacatenin by AKT promotes beta-catenin transcriptional activity. J Biol Chem 282(15):11221–11229. , DOI 10.1074/jbc. M611871200 Wang C, Jin H, Wang N, Fan S, Wang Y, Zhang Y, Wei L, Tao X, Gu D, Zhao F, Fang J, Yao M, Qin W, 2016, Gas6/ Axl Axis contributes to Chemoresistance and metastasis in breast Cancer through Akt/GSK-3beta/beta-catenin signaling. Theranostics 6(8):1205–1219. , DOI 10.7150/thno. 15083 Geyer FC, Lacroix-Triki M, Savage K, Arnedos M, Lambros MB, MacKay A, Natrajan R, Reis-Filho JS, 2011, Betacatenin pathway activation in breast cancer is associated with triple-negative phenotype but not with CTNNB1 mutation. Modern Pathol 24(2):209–231. , DOI 10.1038/ modpathol.2010.205 Li J, Zhou BP, 2011, Activation of beta-catenin and Akt pathways by twist are critical for the maintenance of EMT associated cancer stem cell-like characters. BMC Cancer 11:49. , DOI 10. 1186/1471-2407-11-49 Garrett TP, McKern NM, Lou M, Elleman TC, Adams TE, Lovrecz GO, Zhu HJ, Walker F, Frenkel MJ, Hoyne PA, Jorissen RN, Nice EC, Burgess AW, Ward CW, 2002, Crystal structure of a truncated epidermal growth factor receptor extracellular domain bound to transforming growth factor alpha. Cell 110(6):763–773 Corkery B, Crown J, Clynes M, O'Donovan N, 2009, Epidermal growth factor receptor as a potential therapeutic target in triple-negative breast cancer. Ann Oncol 20(5):862– 867. , DOI 10.1093/annonc/mdn710 Xu W, Yang Z, Lu N, 2015, A new role for the PI3K/Akt signaling pathway in the epithelial-mesenchymal transition. Cell Adhes Migr 9(4):317–324. , DOI 10.1080/ 19336918.2015.1016686 Patil PU, D'Ambrosio J, Inge LJ, Mason RW, Rajasekaran AK, 2015, Carcinoma cells induce lumen filling and EMT in epithelial cells through soluble E-cadherin-mediated activation of EGFR. J Cell Sci 128(23):4366–4379. , DOI 10.1242/jcs.173518 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1153 EP 1163 DI 10.1007/s12253-019-00681-6 PG 11 ER PT J AU Jaberie, H Hosseini, VS Naghibalhossaini, F AF Jaberie, Hajar Hosseini, Vahid Seyed Naghibalhossaini, Fakhraddin TI Evaluation of Alpha 1-Antitrypsin for the Early Diagnosis of Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Alpha 1-antitrypsin; Colorectal Cancer; CEA; Promoter methylation ID Alpha 1-antitrypsin; Colorectal Cancer; CEA; Promoter methylation AB Previous proteomic studies have identified alpha 1-antitrypsin (A1AT) as a potential serum biomarker for colorectal cancer (CRC). In this case-control study, we evaluated plasma A1AT concentration and activity as a biomarker for the early diagnosis of colorectal cancer in a group of 113 sporadic CRC patients. We also analyzed A1AT gene promoter methylation, and genotypes in this group of CRC patients. The plasma A1AT and CEA concentrations were measured using the nephelometric and ELISA methods, respectively. A1AT activity was determined by Trypsin Inhibitor Capacity assay. The genomic DNA from blood samples were subjected to Z and S genotype analysis using PCR-RFLP method and the gene promoter methylation in tumors and their adjacent normal tissues was determined by methylation specific-PCR assay. The plasma levels of A1AT and CEA in patients (median, 2.3 g/L and 5.96 ng/ml, respectively) were significantly higher than those in healthy controls (medians, 1.43 g/L and 2.57 ng/ml, respectively) (p = 0.0001). The plasma A1AT activity and concentrations were positively correlated with the tumor stage and well-discriminated between early and advanced stages. The A1AT activity in plasma was the most useful marker for CRC diagnosis (median 4.8 mmol/min/ml in cases vs 1.91 mmol/min/ml in controls, p = 0.0001). No deficient Z or S alleles of A1AT was observed in patients’ genotype and the gene promoter tends to be more methylated in normal mucosa than in tumor tissues. We conclude that plasma A1AT activity has better sensitivity and specificity than CEA measurement for the early detection of CRC. Promoter demethylation might play a role in increasing plasma A1AT levels in CRC patients. C1 [Jaberie, Hajar] Shiraz University of Medical Sciences, Department of Biochemistry, Zand Street, 7134845794 Shiraz, Iran. [Hosseini, Vahid Seyed] Shiraz University of Medical Sciences, Colorectal Research CenterShiraz, Iran. [Naghibalhossaini, Fakhraddin] Shiraz University of Medical Sciences, Department of Biochemistry, Zand Street, 7134845794 Shiraz, Iran. RP Naghibalhossaini, F (reprint author), Shiraz University of Medical Sciences, Department of Biochemistry, 7134845794 Shiraz, Iran. EM fakhraddin.naghibalhossaini@mail.mcgill.ca CR Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F, 2017, Global patterns and trends in colorectal cancer incidence and mortality. Gut 66:683–691 Dolatkhah R, Somi MH, Bonyadi MJ, Asvadi Kermani I, Farassati F, Dastgiri S, 2015, Colorectal cancer in Iran: molecular epidemiology and screening strategies. J Cancer Epidemiol 2015:643020 Duffy MJ, van Dalen A, Haglund C, Hansson L, Holinski-Feder E, Klapdor R, Lamerz R, Peltomaki P, Sturgeon C, Topolcan O, 2007, Tumour markers in colorectal cancer: European Group On Tumour Markers, EGTM, guidelines for clinical use. Eur J Cancer 43:1348–1360 Pakdel A, Malekzadeh M, Naghibalhossaini F, 2016, The association between preoperative serum CEA concentrations and synchronous liver metastasis in colorectal cancer patients. Cancer Biomark 16:245–252 Chuang SC, Su YC, Lu CY, Hsu HT, Sun LC, Shih YL, Ker CG, Hsieh JS, Lee KT, Wang JY, 2011, Risk factors for the development of metachronous liver metastasis in colorectal cancer patients after curative resection. World J Surg 35:424–429 Duffy MJ, Lamerz R, Haglund C, Nicolini A, Kalousova M, Holubec L, Sturgeon C, 2014, Tumor markers in colorectal cancer, gastric cancer and gastrointestinal stromal cancers: European group on tumor markers 2014 guidelines update. Int J Cancer 134:2513–2522 Balkwill F, Mantovani A, 2001, Inflammation and cancer: back to Virchow? Lancet. 357:539–545 Wang F, Arun P, Friedman J, Chen Z, Van Waes C, 2009, Current and potential inflammation targeted therapies in head and neck cancer. Curr Opin Pharmacol 9:389–395 Ghuman S, Van HemelrijckM, Garmo H, Holmberg L,Malmstrom H, Lambe M, Hammar N, Walldius G, Jungner I, Wulaningsih W, 2017, Serum inflammatory markers and colorectal cancer risk and survival. Br J Cancer 116:1358–1365 Slattery ML, Wolff RK, Herrick J, Caan BJ, Samowitz W, 2009, Tumor markers and rectal cancer: support for an inflammationrelated pathway. Int J Cancer 125:1698–1704 Kittas C, Aroni K, Kotsis L, Papadimitriou CS, 1982, Distribution of lysozyme, alpha-I-antichymotrypsin and alpha-I-antitrypsin in adenocarcinomas of the stomach and large intestine. An immunohistochemical study. Virchows Arch [A] 398:139–147 Greene CM, McElvaney NG, 2009, Proteases and antiproteases in chronic neutrophilic lung disease - relevance to drug discovery. Br J Pharmacol 158:1048–1058 Shahaf G, Moser H, Ozeri E, Mizrahi M, Abecassis A, Lewis EC, 2011, Alpha-1-antitrypsin gene delivery reduces inflammation, increases T-regulatory cell population size and prevents islet allograft rejection. Mol Med, Cambridge, Mass, 17:1000–1011 McCarthy C, Saldova R,WormaldMR, Rudd PM,McElvaney NG, Reeves EP, 2014, The role and importance of glycosylation of acute phase proteins with focus on alpha-1 antitrypsin in acute and chronic inflammatory conditions. J Proteome Res 13:3131–3143 El-Akawi ZJ, Al-Hindawi FK, Bashir NA, 2008, Alpha-1 antitrypsin, alpha1-AT, plasma levels in lung, prostate and breast cancer patients. Neuro Endocrinol Lett 29:482–484 Propst T, Propst A, Dietze O, Judmaier G, Braunsteiner H, VogelW, 1994, Prevalence of hepatocellular carcinoma in alpha-1- antitrypsin deficiency. J Hepatol 21:1006–1011 Benkmann HG, Hanssen HP, Ovenbeck R, Goedde HW, 1987, Distribution of alpha-1-antitrypsin and haptoglobin phenotypes in bladder cancer patients. Hum Hered 37:290–293 Ganji SM, Sahebghadam-Lotfi A, Rastgar-Jazii F, Yazdanbod M, Mota A, Mohsenifar A, Kazemnejad A, 2010, Alpha-1 antitrypsin deficient squamous cell carcinoma of esophagus in the Azeri population of Iran. Lab Med 41:613–618 Trachte AL, Suthers SE, Lerner MR, Hanas JS, Jupe ER, Sienko AE, Adesina AM, Lightfoot SA, Brackett DJ, Postier RG, 2002, Increased expression of alpha-1-antitrypsin, glutathione Stransferase pi and vascular endothelial growth factor in human pancreatic adenocarcinoma. Am J Surg 184:642–647 discussion 647-8 Xie LQ, Zhao C, Cai SJ, XuY, Huang LY, Bian JS, Shen CP, Lu HJ, Yang PY, 2010, Novel proteomic strategy reveal combined alpha1 antitrypsin and cathepsin D as biomarkers for colorectal cancer early screening. J Proteome Res 9:4701–4709 Kwon CH, Park HJ, Choi JH, Lee JR, Kim HK, Jo HJ, Kim HS, Oh N, Song GA, Park DY, 2015, Snail and serpinA1 promote tumor progression and predict prognosis in colorectal cancer. Oncotarget 6:20312–20326 Perez-Holanda S, Blanco I, MenendezM, Rodrigo L, 2014, Serum concentration of alpha-1 antitrypsin is significantly higher in colorectal cancer patients than in healthy controls. BMC Cancer 14:355 Yang P, Cunningham JM, Halling KC, Lesnick TG, Burgart LJ, Wiegert EM, Christensen ER, Lindor NM, Katzmann JA, Thibodeau SN, 2000, Higher risk of mismatch repair-deficient colorectal cancer in alpha, 1)-antitrypsin deficiency carriers and cigarette smokers. Mol Genet Metab 71:639–645 Lindor NM, Yang P, Evans I, Schowalter K, De Andrade M, Li J, Jeavons E, Peterson G, Gallinger S, Bapat B, Hopper J, Jass J, Jenkins M, Templeton A, Potter J, Newcomb PA, Lemarchand L, Grove J, Haile R, Baron J, Seminara D, Limburg P, Thibodeau SN, 2010, Alpha-1-antitrypsin deficiency and smoking as risk factors for mismatch repair deficient colorectal cancer: a study from the colon cancer family registry. Mol Genet Metab 99:157–159 Peltier J, Roperch JP, Audebert S, Borg JP, Camoin, 2016, Quantitative proteomic analysis exploring progression of colorectal cancer: modulation of the serpin family. J Proteome 148:139–148 Bujanda L, Sarasqueta C, Cosme A, Hijona E, Enriquez-Navascues JM, Placer C, Villarreal E, Herreros-Villanueva M, Giraldez MD, Gironella M, Balaguer F, Castells A, 2013, Evaluation of alpha 1- antitrypsin and the levels of mRNA expression of matrix metalloproteinase 7, urokinase type plasminogen activator receptor and COX-2 for the diagnosis of colorectal cancer. PLoS One 8:e51810 Topic A, Juranic Z, Jelic S, Magazinovic IG, 2009, Polymorphism of alpha-1-antitrypsin in hematological malignancies. Genet Mol Biol 32:716–719 Yang P, Sun Z, Krowka MJ, AubryMC, BamletWR,Wampfler JA, Thibodeau SN, Katzmann JA, Allen MS, Midthun DE, Marks RS, de AndradeM(2008, Alpha1-antitrypsin deficiency carriers, tobacco smoke, chronic obstructive pulmonary disease, and lung cancer risk. Arch Intern Med 168:1097–1103 Qiu W, Baccarelli A, Carey VJ, Boutaoui N, Bacherman H, Klanderman B, Rennard S, Agusti A, Anderson W, Lomas DA, DeMeo DL, 2012, Variable DNA methylation is associated with chronic obstructive pulmonary disease and lung function. Am J Respir Crit Care Med 185:373–381 Barton DE, Francke U, 1987, Activation of human alpha 1- antitrypsin genes in rat hepatoma x human fibroblast hybrid cell lines is correlated with demethylation. Somat Cell Mol Genet 13: 635–644 Jaberie H, Naghibalhossaini F, 2016, Recombinant production of native human α-1-antitrypsin protein in the liver HepG2 cells. Biotechnol Lett 38:1683–1690 Sarabi MM, Naghibalhossaini F, 2015, Association of DNA methyltransferases expression with global and gene-specific DNA methylation in colorectal cancer cells. Cell Biochem Funct 33:427–433 Laffranchi M, Elliston ELK, Gangemi F, Berardelli R, Lomas DA, Irving JA, Fra A, 2019, Characterisation of a type II functionallydeficient variant of alpha-1-antitrypsin discovered in the general population. PLoS One 14:e0206955 Haq I, Irving JA, Saleh AD, Dron L, Regan-Mochrie GL, Motamedi-Shad N, Hurst JR, Gooptu B, Lomas DA, 2016, Deficiency mutations of Alpha-1 antitrypsin. Effects on folding, function, and polymerization. Am J Respir CellMol Biol 54:71–80 Ward DG, Suggett N, Cheng Y,WeiW, Johnson H, Billingham LJ, Ismail T,Wakelam MJ, Johnson PJ, Martin A, 2006, Identification of serum biomarkers for colon cancer by proteomic analysis. Br J Cancer 94:1898–1905 Karashima S, Kataoka H, Itoh H, Maruyama R, Koono M, 1990, Prognostic significance of alpha-1-antitrypsin in early stage of colorectal carcinomas. Int J Cancer 45:244–250 Tahara E, Ito H, Taniyama K, Yokozaki H, Hata J, 1984, Alpha 1- antitrypsin, alpha 1-antichymotrypsin, and alpha 2-macroglobulin in human gastric carcinomas: a retrospective immunohistochemical study. Hum Pathol 15:957–964 Weissmann G, Zurier RB, Hoffstein S, 1970, Leucocytic proteases and the immunological release of lysosomal enzymes. Am J Pathol 68:539–560 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1165 EP 1173 DI 10.1007/s12253-019-00679-0 PG 9 ER PT J AU Alsaegh, AM Altaie, MA Zhu, Sh AF Alsaegh, Amjed Mohammed Altaie, Muayad Alaa Zhu, Shengrong TI p63 Expression and its Relation to Epithelial Cells Proliferation in Dentigerous Cyst, Odontogenic Keratocyst, and Ameloblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p63; Ki-67; Odontogenic; Dentigerous; Keratocyst; Ameloblastoma ID p63; Ki-67; Odontogenic; Dentigerous; Keratocyst; Ameloblastoma AB The current controversy about the classification of odontogenic keratocyst reflects the shortage in the understanding of the odontogenic cysts and tumors. The aim of the present study was to investigate p63 immunoexpression and its relation to the proliferation of the epithelial lining in dentigerous cyst (DC), odontogenic keratocyst (OKC), and follicular type of ameloblastoma (AB). The study involved 36 samples, which are DC (n = 12), OKC (n=9), and AB (n = 15). p63 protein expression was evaluated by immunohistochemistry. The results on Ki-67 expression were obtained from our previous studies and correlated with p63 expressions. p63 was expressed differently in the studied lesions with various distribution in different study samples. Statistical analysis using Kruskal-Wallis test showed a significant difference in the expression of p63 protein among DC, OKC, and AB (p = 0.048). Subsequently, Mann- Whitney U test revealed the expression of p63 protein was significantly higher in OKC than DC (p = 0.018). Interestingly, Spearman’s correlation analysis showed a positive correlation between the expression of p63 and Ki-67 in the odontogenic epithelium of DC (σ = 0.757, P = 0.004) and OKC (σ=0.741, P = 0.022).While no such a positive correlation was found between the two studied markers in AB group (σ = 0.006, P = 0.983). In conclusion, the present results indicated various expression and correlation of p63 with the proliferation of odontogenic epithelial cells in DC, OKC, and AB. This diversity could reflect a different role and pathways of ΔNp63 in odontogenic tumor than that in odontogenic cyst. These together will help in better understanding the pathogenesis and biological behavior of odontogenic cysts and tumors. C1 [Alsaegh, Amjed Mohammed] University of Science and Technology of Fujairah, College of Dentistry, Department of Oral and Maxillofacial Surgery, Al-Hulifat, 2202 Fujairah, United Arab Emirates. [Altaie, Muayad Alaa] Sharjah University, Medical College, Sharjah Medical Research InstituteSharjah, United Arab Emirates. [Zhu, Shengrong] Huazhong University of Science and Technology, Tongji Hospital of Tongji Medical College, Department of Oral and Maxillofacial Surgery, 430030 Wuhan, Hubei, China. RP Alsaegh, AM (reprint author), University of Science and Technology of Fujairah, College of Dentistry, Department of Oral and Maxillofacial Surgery, 2202 Fujairah, United Arab Emirates. EM mohammedalsaegh@yahoo.com CR Soluk-Tekkesin M, Wright JM, 2018, The World Health Organization classification of odontogenic lesions: a summary of the changes of the 2017, 4th, edition. Turk Patoloji Derg 34:1–18 Jiang Q, Xu GZ, Yang C, Yu CQ, He DM, Zhang ZY, 2011, Dentigerous cysts associated with impacted supernumerary teeth in the anterior maxilla. Exp Ther Med 2:805–809 Stoelinga PJW, 2018, Keratocystic odontogenic tumour, KCOT, has again been renamed odontogenic keratocyst, OKC). Int J Oral Maxillofac Surg S0901-5027(18)30313–30318 de Molon RS, Verzola MH, Pires LC, Mascarenhas VI, da Silva RB, Cirelli JA, Barbeiro RH, 2015, Five years follow-up of a keratocyst odontogenic tumor treated by marsupialization and enucleation: a case report and literature review. Contemp Clin Dent 6: S106–S110 Al-Moraissi EA, Dahan AA, Alwadeai MS, Oginni FO, Al-Jamali JM, Alkhutari AS, Al-Tairi NH, Almaweri AA, Al-Sanabani JS, 2017, What surgical treatment has the lowest recurrence rate following the management of keratocystic odontogenic tumor?: a large systematic review and meta-analysis. J Craniomaxillofac Surg 45:131–144 Sham E, Leong J, Maher R, Schenberg M, Leung M,Mansour AK, 2009, Mandibular ameloblastoma: clinical experience and literature review. ANZ J Surg 79:739–744 Alsaegh MA, Miyashita H, Zhu SR, 2015, Expression of human papillomavirus is correlated with Ki-67 and COX-2 expressions in keratocystic odontogenic tumor. Pathol Oncol Res 21:65–71 Alsaegh MA, Miyashita H, Taniguchi T, Zhu SR, 2017, Odontogenic epithelial proliferation is correlated with COX-2 expression in dentigerous cyst and ameloblastoma. Exp Ther Med 13: 247–253 Scholzen T, Gerdes J, 2000, The Ki-67 protein: from the known and the unknown. J Cell Physiol 182:311–322 YangAN, KaghadM,WangYM,Gillett E, FlemingMD, DotschV, Andrews NC, Caput D, McKeon F, 1998, p63, a p53 homolog at 3q27- 29, encodes multiple products with transactivating, deathinducing, and dominant-negative activities. Mol Cell 2:305–316 Mangiulli M, Valletti A, Caratozzolo MF, Tullo A, Sbisa E, Pesole G, D'Erchia AM, 2009, Identification and functional characterization of two new transcriptional variants of the human p63 gene. Nucleic Acids Res 37:6092–6104 Chen YK, Hsue SS, Lin LM(2003, Immunohistochemical demonstration of p63 in DMBA-induced hamster buccal pouch squamous cell carcinogenesis. Oral Dis 9:235–240 Goncalves CK, Fregnani ER, Leon JE, Silva-Sousa YT, Perez DE, 2012, Immunohistochemical expression of p63, epidermal growth factor receptor, EGFR, and notch-1 in radicular cysts, dentigerous cysts and keratocystic odontogenic tumors. Braz Dent J 23:337– 343 Kumamoto H, Ohki K, Ooya K, 2005, Expression of p63 and p73 in ameloblastomas. J Oral Pathol Med 34:220–226 Brkic A, Mutlu S, Kocak-Berberoglu H, Olgac V, 2010, Pathological changes and immunoexpression of p63 gene in dental follicles of asymptomatic impacted lower third molars: an immunohistochemical study. J Craniofac Surg 21:854–857 De Laurenzi V, Rossi A, Terrinoni A, Barcaroli D, Levrero M, Costanzo A, Knight RA, Guerrieri P, Melino G, 2000, p63 and p73 transactivate differentiation gene promoters in human keratinocytes. Biochem Biophys Res Commun 273:342–276 Bergholz J, Xiao ZX, 2012, Role of p63 in development, tumorigenesis and Cancer progression. Cancer Microenviron 5:311–322 Venkatanarayan A, Raulji P, Norton W, Chakravarti D, Coarfa C, Su X, Sandur SK, Ramirez MS, Lee J, Kingsley CV, Sananikone EF, Rajapakshe K, Naff K, Parker-Thornburg J, Bankson JA, Tsai KY, Gunaratne PH, Flores ER, 2014, IAPP-driven metabolic reprogramming induces regression of p53-deficient tumours in vivo. Nature 517:626–630 Lo Muzio L, Santarelli A, Caltabiano R, Rubini C, Pieramici T, Fior A, Trevisiol L, Carinci F, Leonardi R, Bufo P, Lanzafame S, Piattelli A, 2005, p63 expression in odontogenic cysts. Int J Oral Maxillofac Surg 34:668–673 Foschini MP, Cocchi R, Marucci G, Pennesi MG, Magrini E, Ligorio C, Lombardini F, Tosi AL, Marchetti C, 2006, High DeltaN p63 isoform expression favours recurrences in odontogenic keratocyst–odontogenic keratocystic tumour. Int J Oral Maxillofac Surg 35:673–675 Lo Muzio L, Santarelli A, Caltabiano R, Rubini C, Pieramici T, Giannone N, Carinci F, Leonardi R, Lanzafame S, Piattelli A, 2006, p63 expression correlates with pathological features and biological behaviour of odontogenic tumours. Histopathology 49:211–214 Gurgel CA, Ramos EA, Azevedo RA, Sarmento VA, da Silva Carvalho AM, dos Santos JN, 2008, Expression of Ki-67, p53 and p63 proteins in keratocyst odontogenic tumours: an immunohistochemical study. J Mol Histol 39:311–316 Dong Q, Pan S, Sun LS, Li TJ, 2010)Orthokeratinized odontogenic cyst: a clinicopathologic study of 61 cases. Arch Pathol Lab Med 134:271–275 Seyedmajidi M, Shafaee S, Shafigh E, Bijani A, Hamidi H, 2011, p63 expression in randomized odontogenic cysts. Saudi Med J 32: 463–466 Atarbashi Moghadam S, Atarbashi Moghadam F, Mokhtari S, Eini E, 2013, Immunohistochemical analysis of P63 expression in odontogenic lesions. Biomed Res Int 2013:624176 Varsha B, Gharat AL, Nagamalini B, Jyothsna M, Mothkur ST, Swaminathan U, 2014, Evaluation and comparison of expression of p63 in odontogenic keratocyst, solid ameloblastoma and unicystic ameloblastoma. J Oral Maxillofac Pathol 18:223–228 de Brito Monteiro BV, Cavalcante RB, Maia Nogueira RL, da Costa Miguel MC, Weege Nonaka CF, da Silveira EJ, 2015, Participation of hMLH1, p63, and MDM2 proteins in the pathogenesis of syndromic and nonsyndromic keratocystic odontogenic tumors. Oral Surg Oral Med Oral Pathol Oral Radiol 120:52–527 Jaafari-Ashkavandi Z, Geramizadeh B, Ranjbar MA, 2015, P63 and Ki-67 expression in Dentigerous cyst and Ameloblastomas. J Dent, Shiraz, 16:323–328 Chandrangsu S, Sappayatosok K, 2016, p53, p63 and p73 expression and angiogenesis in keratocystic odontogenic tumors. J Clin Exp Dent 8:e505–e511 Zhou Y, Xu Q, Ling B, Xiao W, Liu P, 2011, Reduced expression of ΔΝp63α in cervical squamous cell carcinoma. Clin Invest Med 34:E184–E191 Narahashi T, Niki T, Wang T, Goto A, Matsubara D, Funata N, Fukayama M, 2006, Cytoplasmic localization of p63 is associated with poor patient survival in lung adenocarcinoma. Histopathology 49:349–357 Hsiao YH, Su YA, Tsai HD, Mason JT, Chou MC, Man YG, 2010, Increased invasiveness and aggressiveness in breast epithelia with cytoplasmic p63 expression. Int J Biol Sci 6:428–442 Dhillon PK, Barry M, Stampfer MJ, Perner S, Fiorentino M, Fornari A, Ma J, Fleet J, Kurth T, Rubin MA, Mucci LA, 2009, Aberrant cytoplasmic expression of p63 and prostate cancer mortality. Cancer Epidemiol Biomark Prev 18:595–600 Sbisa E, Mastropasqua G, Lefkimmiatis K, Caratozzolo MF, D'Erchia AM, Tullo A, 2006, Connecting p63 to cellular proliferation: the example of the adenosine deaminase target gene. Cell Cycle 5:205–212 de Oliveira MG, Lauxen Ida S, Chaves AC, Rados PV, Sant'Ana FilhoM(2011, Odontogenic epithelium: immunolabeling of Ki-67, EGFR and survivin in pericoronal follicles, dentigerous cysts and keratocystic odontogenic tumors. Head Neck Pathol 5:1–7 Singh HP, 2016)Need to reclassify keratocystic odontogenic tumor into cyst and neoplasm. Natl J Maxillofac Surg 7:111 Yugawa T, Narisawa-Saito M, Yoshimatsu Y, Haga K, Ohno S, Egawa N, Fujita M, Kiyono T, 2010, DeltaNp63alpha repression of the Notch1 gene supports the proliferative capacity of normal human keratinocytes and cervical cancer cells. Cancer Res 70:4034–4044 Zhu L, Rorke EA, Eckert RL, 2007, DeltaNp63alpha promotes apoptosis of human epidermal keratinocytes. J Invest Dermatol 127:1980–1991 Venkatanarayan A, Raulji P, Norton W, Flores ER, 2016, Novel therapeutic interventions for p53-altered tumors through manipulation of its family members. p63 and p73 Cell Cycle 15:164–171 Napoli M, Venkatanarayan A, Raulji P, Meyers BA, Norton W, Mangala LS, Sood AK, Rodriguez-Aguayo C, Lopez-Berestein G, Vin H, Duvic M, Tetzlaff MB, Curry JL, Rook AH, Abbas HA, Coarfa C, Gunaratne PH, Tsai KY, Flores ER, 2016, ΔNp63/DGCR8-dependent MicroRNAs mediate therapeutic efficacy of HDAC inhibitors in Cancer. Cancer Cell 29:874–888 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1175 EP 1182 DI 10.1007/s12253-019-00680-7 PG 8 ER PT J AU Zaky, HA Elsers, D Bakry, R Abdelwanis, M Nabih, O Hafez, R Rezk, M AF Zaky, H Amen Elsers, Dalia Bakry, Rania Abdelwanis, Mostafa Nabih, Ola Hafez, Rania Rezk, Mahmoud TI Prognostic Value of Accumulative Expression of COX-2 and p53 in Small and Diffuse Large B Cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE p53; COX-2; Lymphoma ID p53; COX-2; Lymphoma AB Cyclooxygenase-2 (COX-2) plays an important role in carcinogenesis, which catalyzes the conversion of arachidonic acid into prostaglandins. P53 is a tumor suppressor gene that contributes to apoptosis and cell cycle control. There is functional interaction between p53 and COX-2, which lead to abrogation of apoptosis and progression of malignancy. To assess the relationship between COX-2, p53 expression and the clinicopathologic features in SLL and DLBCL. We immunohistochemically examined the expression of COX-2 and p53 in non-neoplastic lymphoid cells, lymph nodal low-grade (50 cases of SLL), intermediate and high-grade lymphomas (100 cases of DLBCL) and their corresponding bone marrow specimens. The expression of COX-2 and p53 was absent in the in non-neoplastic lymphoid cells. In contrast, their expression values increased progressively with the advancing grade of lymphoma (p < 0.001). COX-2 expression was significantly associated with advanced disease stage, high grade lymphomas, and disease relapse and p53 expression. The p53was detected in 64.5% in patients positive for COX-2. The expressions of COX-2 and p53 proteins, were significantly associated with shorter overall-survival and progression free survival. Here we report up-regulation of COX-2and p53 protein expression in SLL and DLBCL indicating their interactive involvement in the pathogenesis of lymphoma. Our data provide a rationale for further investigation of COX-2 expression in lymphomas for potential prognostic, chemopreventive and chemotherapeutic purposes. C1 [Zaky, H Amen] Asyut University, South Egypt Cancer Institute, Medical Oncology & Hematology DepartmentAssiut, Egypt. [Elsers, Dalia] Asyut University, Faculty of Medicine, Pathology DepartmentAssiut, Egypt. [Bakry, Rania] Asyut University, South Egypt Cancer Institute, Clinical Pathology DepartmentAssiut, Egypt. [Abdelwanis, Mostafa] Asyut University, South Egypt Cancer Institute, Radiotherapy DepartmentAssiut, Egypt. [Nabih, Ola] Asyut University, Clinical Oncology DepartmentAssiut, Egypt. [Hafez, Rania] Asyut University, Internal Medicine, Hematology UnitAssiut, Egypt. [Rezk, Mahmoud] Asyut University, Faculty of Medicine, Pathology DepartmentAssiut, Egypt. RP Zaky, HA (reprint author), Asyut University, South Egypt Cancer Institute, Medical Oncology & Hematology Department, Assiut, Egypt. EM amenzaky74@yahoo.com CR Jelic S, Jovanovic V, Milanovic N et al, 1997, Richter syndrome with emphasis on large-cell non-Hodgkin lymphoma in previously unrecognized subclinical chronic lymphocytic leukemia. Neoplasma. 44:63–68 Matolcsy A, Casali P, Knowles DM, 1995, Different clonal origin of B cell populations of chronic lymphocytic leukemia and largecell lymphoma in Richter’s syndrome. Ann N YAcad Sci 764:496– 503 Traweek ST, Liu J, JohnsonRM,Winberg CD, Rappaport H, 1993, High-grade transformation of chronic lymphocytic leukemia and low-grade non-Hodgkin’s lymphoma. Genotypic confirmation of clonal identity. Am J Clin Pathol 100:519–526 Brynes RK, McCourty A, Sun NC, Koo CH, 1995, Trisomy 12 in Richter’s transformation of chronic lymphocytic leukemia. Am J Clin Pathol 104:199–203 Bea S, Lopez-Guillermo A, Ribas M et al, 2002, Genetic imbalances in progressed B cell chronic lymphocytic leukemia and transformed large-cell lymphoma, Richter’s syndrome). Am J Pathol 161:957–968 d’Amore ES, Wick MR, Geisinger KR, Frizzera G, 1990, Primary malignant lymphoma arising in postmastectomy lymphedema. Another facet of the Stewart-Treves syndrome. Am J Surg Pathol 14:456–463 Copie-Bergman C, Niedobitek G, Mangham DC et al, 1997, Epstein-Barr virus in B cell lymphomas associated with chronic suppurative inflammation. J Pathol 183:287–292 Aozasa K, Takakuwa T, Nakatsuka S, 2005, Pyothorax-associated lymphoma: a lymphoma developing in chronic inflammation. Adv Anat Pathol 12:324–331 Sehn LH, Scott DW, Chhanabhai M et al, 2011, Impact of concordant and discordant bone marrow involvement on outcome in diffuse large B cell lymphoma treated with R-CHOP. J Clin Oncol Off J Am Soc Clin Oncol 29:1452–1457 Cabanillas F, Velasquez WS, Hagemeister FB, McLaughlin P, Redman JR, 1992, Clinical, biologic, and histologic features of late relapses in diffuse large cell lymphoma. Blood. 79:1024–1028 Hodges GF, Lenhardt TM, Cotelingam JD, 1994, Bone marrow involvement in large-cell lymphoma. Prognostic implications of discordant disease. Am J Clin Pathol 101:305–311 Robertson LE, Redman JR, Butler JJ et al, 1991, Discordant bone marrow involvement in diffuse large-cell lymphoma: a distinct clinical-pathologic entity associated with a continuous risk of relapse. J Clin Oncol Off J Am Soc Clin Oncol 9:236–242 Barisik NO, Bozkurt S, Gumus M et al, 2010, Expression and prognostic significance of Cox-2 and p-53 in Hodgkin lymphomas: a retrospective study. Diagn Pathol 5:19 Ohsawa M, Fukushima H, Ikura Y et al, 2006, Expression of cyclooxygenase-2 in Hodgkin’s lymphoma: its role in cell proliferation and angiogenesis. Leuk Lymphoma 47:1863–1871 Zhi H, Wang L, Zhang J et al, 2006, Significance of COX-2 expression in human esophageal squamous cell carcinoma. Carcinogenesis. 27:1214–1221 Romero M, Artigiani R, Costa H, Oshima CT,Miszputen S, Franco M, 2008, Evaluation of the immunoexpression of COX-1, COX-2 and p53 in Crohn’s disease. Arq Gastroenterol 45:295–300 Asproni P, Vignoli M, Cancedda S, Millanta F, Terragni R, Poli A, 2014, Immunohistochemical expression of cyclooxygenase-2 in normal, hyperplastic and neoplastic canine lymphoid tissues. J Comp Pathol 151:35–41 Konturek PC, Konturek SJ, Pierzchalski P et al, 2002, Gastric MALT-lymphoma, gastrin and cyclooxygenases. Acta Gastro- Enterol Belg 65:17–23 Al-Salam S, Awwad A, Sudhadevi M et al, 2013, Epstein-Barr virus infection correlates with the expression of COX-2, p16(INK4A, and p53 in classic Hodgkin lymphoma. Int J Clin Exp Pathol 6:2765–2777 Paul AG, Sharma-Walia N, Chandran B, 2011, Targeting KSHV/ HHV-8 latency with COX-2 selective inhibitor nimesulide: a potential chemotherapeutic modality for primary effusion lymphoma. PLoS One 6:e24379 Paydas S, Ergin M, Erdogan S, Seydaoglu G, 2007, Cyclooxygenase-2 expression in non-Hodgkin’s lymphomas. Leuk Lymphoma 48(2):389–395 Mestre F, Gutierrez A, Ramos R et al, 2012, Expression of COX-2 on Reed-Sternberg cells is an independent unfavorable prognostic factor in Hodgkin lymphoma treated with ABVD. Blood. 119: 6072–6079 Hsum YW, Yew WT, Hong PL et al, 2011, Cancer chemopreventive activity ofmaslinic acid: suppression of COX-2 expression and inhibition of NF-kappaB and AP-1 activation in Raji cells. Planta Med 77:152–157 Paydas S, Ergin M, Seydaoglu G, Erdogan S, Yavuz S, 2009, Prognos t i c [corrected] significance of angiogenic/ lymphangiogenic, anti-apoptotic, inflammatory and viral factors in 88 cases with diffuse large B cell lymphoma and review of the literature. Leuk Res 33(12) Wun T, McKnight H, Tuscano JM, 2004, Increased cyclooxygenase-2, COX-2): a potential role in the pathogenesis of lymphoma. Leuk Res 28:179–190 Bernard MP, Bancos S, Sime PJ, Phipps RP, 2008, Targeting cyclooxygenase-2 in hematological malignancies: rationale and promise. Curr Pharm Des 14:2051–2060 Hussein MR, Haemel AK, Wood GS, 2003, p53-related pathways and the molecular pathogenesis of melanoma. Eur J Cancer Prev 12:93–100 Hussein MR, Al-Sabae TM, Georgis MN, 2004, Analysis of Bcl-2 and p53 protein expression in non-Hodgkin’s lymphoma. Ann Oncol 15:1849–1850 Hussein MR, Al-Sabae TM, Georgis MN, 2005, Analysis of the Bcl-2 and p53 protein expression in the lymphoproliferative lesions in the upper Egypt. Cancer Biol Ther 4:324–328 Li HL, Sun BZ, Ma FC, 2004, Expression of COX-2, iNOS, p53 and Ki-67 in gastricmucosa-associated lymphoid tissue lymphoma. World J Gastroenterol 10:1862–1866 Holmila R, Cyr D, Luce D et al, 2008, COX-2 and p53 in human sinonasal cancer: COX-2 expression is associated with adenocarcinoma histology and wood-dust exposure. Mutat Res 122:2154– 2159 Kardosh A, Wang W, Uddin J et al, 2005, Dimethyl-celecoxib, DMC), a derivative of celecoxib that lacks cyclooxygenase-2- inhibitory function, potently mimics the anti-tumor effects of celecoxib on Burkitt’s lymphoma in vitro and in vivo. Cancer Biology & Therapy 4:571–582 Witkowska M, Smolewski P, 2013, Helicobacter pylori infection, chronic inflammation, and genomic transformations in gastric MALT lymphoma. Mediat Inflamm 2013:523170 Loong F, Chan AC, Ho BC et al, 2010, Diffuse large B cell lymphoma associated with chronic inflammation as an incidental finding and new clinical scenarios. Cancer Biol Ther 23:493–501 Valli R, Froio E, Alvarez de Celis MI, Mandato VD, Piana S, 2014, Diffuse large B cell lymphoma occurring in an ovarian cystic teratoma: expanding the spectrum of large B cell lymphoma associated with chronic inflammation. Hum Pathol 45:2507–2511 Sagaert X, Van Cutsem E, De Hertogh G, Geboes K, Tousseyn T, 2010, Gastric MALT lymphoma: a model of chronic inflammation-induced tumor development. Nat Rev Gastroenterol Hepatol 7:336–346 Baecklund E, Iliadou A, Askling J et al, 2006, Association of chronic inflammation, not its treatment, with increased lymphoma risk in rheumatoid arthritis. Arthritis Rheum 54:692–701 Valli R, Piana S, Capodanno I, Cavazza A, 2011, Diffuse large B cell lymphoma associated with chronic inflammation arising in a renal pseudocyst. Int J Surg Pathol 19:117–119 Hoeft B, Becker N, Deeg E, Beckmann L, Nieters A, 2008, Joint effect between regular use of non-steroidal anti-inflammatory drugs, variants in inflammatory genes and risk of lymphoma. Cancer Causes Control 19(2):163–173 Katkoori VR, Manne K, Vital-Reyes VS et al, 2013, Selective COX-2 inhibitor, celecoxib, decreases cellular growth in prostate cancer cell lines independent of p53. Biotechnic & Histochemistry: Int J Surg Pathol 88:38–46 Piplani H, VaishV, Rana C, Sanyal SN, 2013, Up-regulation of p53 andmitochondrial signaling pathway in apoptosis by a combination of COX-2 inhibitor, Celecoxib and Dolastatin 15, a marine mollusk linear peptide in experimental colon carcinogenesis. Mol Carcinog 52:845–858 Hazar B, Ergin M, Seyrek E, Erdogan S, Tuncer I, Hakverdi S, 2004, Cyclooxygenase-2, Cox-2, expression in lymphomas. Leuk Lymphoma 45:1395–1399 Zweifel BS, Davis TW, Ornberg RL, Masferrer JL, 2002, Direct evidence for a role of cyclooxygenase 2-derived prostaglandin E2 in human head and neck xenograft tumors. Cancer Res 62:6706– 6711 Hosomi Y, Yokose T, Hirose Yet al, 2000, Increased cyclooxygenase 2, COX-2, expression occurs frequently in precursor lesions of human adenocarcinoma of the lung. Lung Cancer 30:73–81 Miyata Y, Koga S, Kanda S, Nishikido M, Hayashi T, Kanetake H, 2003, Expression of cyclooxygenase-2 in renal cell carcinoma: correlation with tumor cell proliferation, apoptosis, angiogenesis, expression of matrix metalloproteinase-2, and survival. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research 9:1741–1749 Kim MH, Seo SS, Song YS et al, 2003, Expression of cyclooxygenase-1 and -2 associated with expression of VEGF in primary cervical cancer and at metastatic lymph nodes. Gynecol Oncol 90:83–90 Ermert L, Dierkes C, Ermert M, 2003, Immunohistochemical expression of cyclooxygenase isoenzymes and downstream enzymes in human lung tumors. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research 9:1604– 1610 Sharma S, Stolina M, Yang SC et al, 2003, Tumor cyclooxygenase 2-dependent suppression of dendritic cell function. Gynecol Oncol 9:961–968 Zhang H, Sun XF, 2002, Overexpression of cyclooxygenase-2 correlates with advanced stages of colorectal cancer. Am J Gastroenterol 97:1037–1041 Liu X, Li P, Zhang ST, You H, Jia JD, Yu ZL, 2008, COX-2mRNA expression in esophageal squamous cell carcinoma, ESCC, and effect by NSAID. Clin Cancer Res 21:9–14 Chiarugi V, Magnelli L, Gallo O, 1998, Cox-2, iNOS and p53 as play-makers of tumor angiogenesis, review). Int JMolMed 2:715– 719 Gallo O, Schiavone N, Papucci L et al, 2003, Down-regulation of nitric oxide synthase-2 and cyclooxygenase-2 pathways by p53 in squamous cell carcinoma. Am J Pathol 163:723–732 Han JA, Kim JI, Ongusaha PP et al, 2002, P53-mediated induction of Cox-2 counteracts p53- or genotoxic stress-induced apoptosis. EMBO J 21:5635–5644 Subbaramaiah K, Altorki N, Chung WJ, Mestre JR, Sampat A, Dannenberg AJ, 1999, Inhibition of cyclooxygenase-2 gene expression by p53. J Biol Chem 274:10911–10,915 Choi EM, Heo JI, Oh JYet al, 2005, COX-2 regulates p53 activity and inhibits DNA damage-induced apoptosis. Biochem Biophys Res Commun 328:1107–1112 deMoraes E, Dar NA, deMoura Gallo CV, Hainaut P, 2007, Crosstalks between cyclooxygenase-2 and tumor suppressor protein p53: Balancing life and death during inflammatory stress and carcinogenesis. Biochem Biophys Res Commun 121:929–937 Khalifeh I, Munkarah AR, Lonardo F et al, 2004, Expression of Cox-2, CD34, Bcl-2, and p53 and survival in patients with primary peritoneal serous carcinoma and primary ovarian serous carcinoma. Int J Cancer 23:162–169 Hussain SP, Harris CC, 1999, p53 mutation spectrum and load: the generation of hypotheses linking the exposure of endogenous or exogenous carcinogens to human cancer. Mutat Res 428:23–32 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1183 EP 1190 DI 10.1007/s12253-019-00674-5 PG 8 ER PT J AU Ekalaksananan, T Wongjampa, W Phusingha, P Chuerduangphui, J Vatanasapt, P Promthet, S Patarapadungkit, N Pientong, Ch AF Ekalaksananan, Tipaya Wongjampa, Weerayut Phusingha, Pensiri Chuerduangphui, Jureeporn Vatanasapt, Patravoot Promthet, Supannee Patarapadungkit, Natcha Pientong, Chamsai TI Comprehensive Data of P53 R282 Gene Mutation with Human Papillomaviruses (HPV)-Associated Oral Squamous Cell Carcinoma (OSCC) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Human papillomavirus; HPV; P53mutation; P53 exon 8 codon 282; Oral squamous cell carcinoma; OSCC; Allele specific oligonucleotide; ASO ID Human papillomavirus; HPV; P53mutation; P53 exon 8 codon 282; Oral squamous cell carcinoma; OSCC; Allele specific oligonucleotide; ASO AB Alterations of the P53 gene and human papillomavirus (HPV) infection are associated with development of oral squamous cell carcinoma (OSCC). We aimed to identify mutation of P53 exon 8 codon 282 in OSCC and correlate these with HPV infection as well as histopathological grade of OSCC. Samples of known HPV infection status were studied including oral lesion cells, formalin-fixed paraffin embedded (FFPE) tissues from OSCC and exfoliated oral cells of matched age-sex controls. P53 exon 8 mutation was detected using the polymerase chain reaction (PCR). Mutation of codon 282 was identified by allele-specific oligonucleotide typing (ASO) using EvaGreen real-time PCR. The PCR products were analyzed by gel electrophoresis and melting curve analysis. Mutation of P53 exon 8 was seen in 81.7% and 69.6% of FFPE OSCC tissues and oral lesion cells, respectively. This was significantly higher than in controls (16.7%). Frequency of mutation did not differ between HPV-positive samples (62.5% and 81.8% in oral lesion cells and FFPE tissue samples, respectively) and HPV-negative samples (73.3% and 81.5% in oral lesion cells and FFPE tissue samples, respectively). This finding is similar to P53 codon 282 mutation that was found only in FFPE tissues (35.0%) and oral lesion cells (32.6%) from both HPV-positive and negative OSCC. Interestingly, frequency of mutation was higher in well-differentiated OSCC with HPV-infection (28.1%) than without HPV (14.8%). This result demonstrated a mutation hot spot in P53 associated with oral carcinogenesis and might be useful to guide chemotherapeutic modality for HPV-associated OSCC in northeast Thailand. C1 [Ekalaksananan, Tipaya] Khon Kaen University, Faculty of Medicine, Department of MicrobiologyKhon Kaen, Thailand. [Wongjampa, Weerayut] Khon Kaen University, Faculty of Medicine, Department of MicrobiologyKhon Kaen, Thailand. [Phusingha, Pensiri] Khon Kaen University, Faculty of Medicine, Department of MicrobiologyKhon Kaen, Thailand. [Chuerduangphui, Jureeporn] Khon Kaen University, Faculty of Medicine, Department of MicrobiologyKhon Kaen, Thailand. [Vatanasapt, Patravoot] Khon Kaen University, HPV & EBV and Carcinogenesis Research GroupKhon Kaen, Thailand. [Promthet, Supannee] Khon Kaen University, HPV & EBV and Carcinogenesis Research GroupKhon Kaen, Thailand. [Patarapadungkit, Natcha] Khon Kaen University, HPV & EBV and Carcinogenesis Research GroupKhon Kaen, Thailand. [Pientong, Chamsai] Khon Kaen University, Faculty of Medicine, Department of MicrobiologyKhon Kaen, Thailand. RP Pientong, Ch (reprint author), Khon Kaen University, Faculty of Medicine, Department of Microbiology, Khon Kaen, Thailand. EM chapie@kku.ac.th CR Parkin DM, Pisani P, Ferlay J, 1999, Estimates of the worldwide incidence of 25 major cancers in 1990. Int J Cancer 80:827–841. , DOI 10.1002/(SICI)1097-0215(19990315)80:6<827:: AID-IJC6>3.0.CO;2-P Tsantoulis P, Kastrinakis N, Tourvas A, Laskaris G, Gorgoulis V, 2007, Advances in the biology of oral cancer. Oral Oncol 43:523– 534. , DOI 10.1016/j.oraloncology.2006.11.010 Krishna A, Singh S, Kumar V, Pal U, 2015, Molecular concept in human oral cancer. Natl J Maxillofac Surg 6:9. , DOI 10. 4103/0975-5950.168235 Kashiwazaki H, Tonoki H, Tada M, Chiba I, Shindoh M, TotsukaY, Iggo R, Moriuchi T, 1997, High frequency of p53 mutations in human oral epithelial dysplasia and primary squamous cell carcinoma detected by yeast functional assay. Oncogene 15:2667. https:// doi.org/10.1038/sj.onc.1201438 Greenblatt M, BennettWP, Hollstein M, Harris C, 1994, Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res 54:4855–4878 Fei P, El-Deiry WS, 2003, P53 and radiation responses. Oncogene 22:5774. , DOI 10.1038/sj.onc.1206677 Petitjean A, Mathe E, Kato S, Ishioka C, Tavtigian SV, Hainaut P, Olivier M, 2007, Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat 28:622– 629. , DOI 10.1002/humu.20495 Xu J,Wang J, Hu Y, Qian J, Xu B, Chen H, ZouW, Fang JY, 2014, Unequal prognostic potentials of p53 gain-of-function mutations in human cancers associate with drug-metabolizing activity. Cell Death Dis 5:e1108. , DOI 10.1038/cddis.2014.75 Suzuki H, Komiya A, Aida S, Ito H, Yatani R, Shimazaki J, 1996, Detection of human papillomavirus DNA and p53 gene mutations in human prostate cancer. Prostate 28:318–324. , DOI 10.1002/, SICI)1097-0045(199605)28:5<318::AID-PROS8>3.0.CO;2-7 He D, Zhang DK, Lam KY, Ma L, Ngan HYS, Liu SS, Tsao SW, 1997, Prevalence of HPV infection in esophageal squamous cell carcinoma in Chinese patients and its relationship to the p53 gene mutation. Int J Cancer 72:959–964. , DOI 10.1002/(SICI, 1097-0215(19970917)72:6<959::AID-IJC7>3.0.CO;2-O Busby-Earle R, Steel C, Williams A, Cohen B, Bird C, 1994, p53 mutations in cervical carcinogenesis–low frequency and lack of correlation with human papillomavirus status. Br J Cancer 69:732. , DOI 10.1038/bjc.1994.138 Oda D, Bigler L, Mao EJ, Disteche CM, 1996, Chromosomal abnormalities in HPV-16-immortalized oral epithelial cells. Carcinogenesis 17:2003–2008. , DOI 10.1093/carcin/17. 9.2003 Lo K, Mok C, Chung G, Huang D,Wong F, Chan M, Lee J, Tsao S, 1992, Presence of p53 mutation in human cervical carcinomas associated with HPV-33 infection. Anticancer Res 12:1989–1994 Phusingha P, Ekalaksananan T, Vatanasapt P, Loyha K, Promthet S, Kongyingyoes B, Patarapadungkit N, Chuerduangphui J, Pientong C, 2017, Human papillomavirus, HPV, infection in a case-control study of oral squamous cell carcinoma and its increasing trend in northeastern Thailand. J Med Virol 89:1096–1101. , DOI 10.1002/jmv.24744 Snijders AM, Schmidt BL, Fridlyand J, Dekker N, Pinkel D, Jordan RC, Albertson DG, 2005, Rare amplicons implicate frequent deregulation of cell fate specification pathways in oral squamous cell carcinoma. Oncogene 24:4232. , DOI 10.1038/sj.onc. 1208601 Trivedy C, Warnakulasuriya K, Tavassoli M, Steingrimsdottir H, Penhallow J, Maher R, Johnson N, 1998, p53 aberrations in oral submucous fibrosis and oral squamous cell carcinoma detected by immunocytochemistry and PCR-SSCP. J Oral Pathol Med 27:72– 77. , DOI 10.1111/j.1600-0714.1998.tb02097.x Nylander K, Schildt E, Eriksson M, Magnusson A, Mehle C, Roos G, 1996, A non-random deletion in the p53 gene in oral squamous cell carcinoma. Br J Cancer 73:1381. , DOI 10.1038/bjc. 1996.262 Ostwald C, Gogacz P, Hillmann T, Schweder J, Gundlach K, Kundt G, Barten M, 2000, p53 mutational spectra are different between squamous-cell carcinomas of the lip and the oral cavity. Int J Cancer 88:82–86. , DOI 10.1002/1097-0215(20001001)88:1<82:: AID-IJC13>3.0.CO;2-N Chuerduangphui J, Pientong C, Patarapadungkit N, Chotiyano A, Vatanasapt P, Kongyingyoes B, Promthet S, Swangphon P, Bumrungthai S, Pimson C, 2017, Amplification of EGFR and cyclin D1 genes associated with human papillomavirus infection in oral squamous cell carcinoma. Med Oncol 34:148. , DOI 10.1007/s12032-017-1010-6 Lee YS, Bae SM, Kwak SY, Park DC, Kim YW, Hur SY, Park EK, Han BD, Lee YJ, Kim CK, 2006, Cell cycle regulatory protein expression profiles by adenovirus p53 infection in human papilloma virus-associated cervical cancer cells. Cancer Res Treat 38:168. , DOI 10.4143/crt.2006.38.3.168 Muller PA, Vousden KH, 2014, Mutant p53 in cancer: new functions and therapeutic opportunities. Cancer Cell 25:304–317. , DOI 10.1016/j.ccr.2014.01.021 Javid J, Masroor M, Rashid Mir A, Ahamad I, Farooq S, 2012, Clinical and prognostic significance of R282W p53 gene mutation in North India patients with non small cell lung Cancer. Transl Med 2:2161–1025.1000110. , DOI 10.4172/2161-1025. 1000110 Acharya S, Ekalaksananan T, Vatanasapt P, Loyha K, Phusingha P, Promthet S, Kongyingyoes B, Pientong C, 2015, Association of Epstein-Barr virus infection with oral squamous cell carcinoma in a case–control study. J Oral Pathol Med 44:252–257 Kang CJ, Lin CY, Wang HM, Fan KH, Ng SH, Lee LY, Chen IH, Huang SF, Liao CT, Yen TC, 2011, The number of pathologically positive lymph nodes and pathological tumor depth predicts prognosis in patients with poorly differentiated squamous cell carcinoma of the oral cavity. Int J Radiat Oncol Biol Phys 81:e223–e230. , DOI 10.1016/j.ijrobp.2011.03.060 Montoro JRMC, Hicz HA, Souza L, Livingstone D, Melo DH, Tiveron RC,Mamede RCM(2008, Prognostic factors in squamous cell carcinoma of the oral cavity. Rev Bras Otorrinolaringol 74: 861–866. , DOI 10.1590/S0034-72992008000600008 Mishra A, Bharti AC, Varghese P, Saluja D, Das BC, 2006, Differential expression and activation of NF-κB family proteins during oral carcinogenesis: role of high risk human papillomavirus infection. Int J Cancer 119:2840–2850. , DOI 10.1002/ijc.22262 Wong YK, Liu TY, Chang KW, Lin SC, Chao TW, Li PL, Chang CS, 1998, p53 alterations in betel quid- and tobacco-associated oral squamous cell carcinomas from Taiwan. J Oral PatholMed 27:243– 248. , DOI 10.1111/j.1600-0714.1998.tb01950.x HeatonCM, Durr ML, Tetsu O, Zante A,Wang SJ, 2014, TP53 and CDKN2a mutations in never-smoker oral tongue squamous cell carcinoma. Laryngoscope 124. , DOI 10.1002/lary.24595 Thongsuksai P, Boonyaphiphat P, Sriplung H, Sudhikaran W, 2003, p53 mutations in betel-associated oral cancer from Thailand. Cancer Lett 201:1–7. , DOI 10.1016/S0304- 3835(03)00417-8 Zhang Y, Coillie S, Fang J, Xu J, 2016, Gain of function of mutant p53: R282W on the peak? Oncogenesis 5:e196. , DOI 10. 1038/oncsis.2016.8 Hsieh LL, Wang PF, Chen I-H, Liao CT, Wang HM, Chen MC, Chang JTC, Cheng AJ, 2001, Characteristics of mutations in the p53 gene in oral squamous cell carcinoma associated with betel quid chewing and cigarette smoking in Taiwanese. Carcinogenesis 22: 1497–1503. , DOI 10.1093/carcin/22.9.1497 Sharan RN, Mehrotra R, Choudhury Y, Asotra K, 2012, Association of betel nut with carcinogenesis: revisit with a clinical perspective. PLoS One 7:e42759 Pfeifer GP, Denissenko MF, Olivier M, Tretyakova N, Hecht SS, Hainaut P, 2002, Tobacco smoke carcinogens, DNA damage and p53 mutations in smoking-associated cancers. Oncogene 21:7435 Scheidt JHG, Yurgel LS, Cherubini K, FigueiredoMAZ, Salum FG, 2012, Characteristics of oral squamous cell carcinoma in users or non users of tobacco and alcohol. RevistaOdonto Ciencia 27:69–73 Paquette RL, Lee YY, Wilczynski SP, Karmakar A, Kizaki M, Miller CW, Koeffler HP, 1993, Mutations of p53 and human papillomavirus infection in cervical carcinoma. Cancer 72:1272–1280. , DOI 10.1002/1097-0142(19930815)72:4<1272::AIDCNCR2820720420> 3.0.CO;2-Q Dai M, Clifford GM, Le Calvez F, Castellsague X, Snijders PJ, Pawlita M, Herrero R, Hainaut P, Franceschi S, 2004, Human papillomavirus type 16 and TP53 mutation in oral cancer: matched analysis of the IARC multicenter study. Cancer Res 64:468–471. , DOI 10.1158/0008-5472 Shima K, Kobayashi I, Saito I, Kiyoshima T, Matsuo K, Ozeki S, Ohishi M, Sakai H, 2000, Incidence of human papillomavirus 16 and 18 infection and p53 mutation in patients with oral squamous cell carcinoma in Japan. Br J Oral Maxillofac Surg 38:445–450. , DOI 10.1054/bjom.2000.0162 Singh RD, Patel KR, Patel PS, 2016, p53mutation spectrum and its role in prognosis of oral cancer patients: a study from Gujarat,West India. Mutat Res-Fund Mol M 783:15–26. , DOI 10.1016/ j.mrfmmm.2015.12.001 Schwartz SR, Yueh B, McDougall JK, Daling JR, Schwartz SM, 2001, Human papillomavirus infection and survival in oral squamous cell cancer: a population-based study. Otolaryngol Head Neck Surg 125:1–9. , DOI 10.1067/mhn.2001.116979 Chiba I, Shindoh M, Yasuda M, Yamazaki Y, Amemiya A, Sato Y, Fujinaga K, K-i N, Fukuda H, 1996, Mutations in the p53 gene and human papillomavirus infection as significant prognostic factors in squamous cell carcinomas of the oral cavity. Oncogene 12:1663–1668 Attner P, Du J, Nasman A, Hammarstedt L, Ramqvist T, Lindholm J, Marklund L, Dalianis T, Munck-Wikland E, 2011, Human papillomavirus and survival in patients with base of tongue cancer. Int J Cancer 128:2892–2897. , DOI 10.1002/ijc.25625 Klozar J, Kratochvil V, Salakova M, Smahelova J, Vesela E, Hamsikova E, Betka J, Tachezy R, 2008, HPV status and regional metastasis in the prognosis of oral and oropharyngeal cancer. Eur Arch Otorhinolaryngol 265:75–82. , DOI 10.1002/ijc.25625 BrandweinM, Zeitlin J, Nuovo G,MacConnell P, Bodian C, Urken M, Biller H, 1994, HPV detection using “hot start” polymerase chain reaction in patients with oral cancer: a clinicopathological study of 64 patients. Mod Pathol 7:720–727 Sisk EA, Soltys SG, Zhu S, Fisher SG, Carey TE, Bradford CR, 2002, Human papillomavirus and p53 mutational status as prognostic factors in head and neck carcinoma. Head Neck 24:841–849. , DOI 10.1002/hed.10146 Huang C, Taki T, Adachi M, Konishi T, HigashiyamaM,MiyakeM, 1998, Mutations in exon 7 and 8 of p53 as poor prognostic factors in patients with non-small cell lung cancer. Oncogene 16:2469. , DOI 10.1038/sj.onc.1201776 Lee LN, Shew JY, Sheu JC, Lee YC, Lee WC, Fang MT, Chang HF, Yu CJ, Yang PC, Luh KT, 1994, Exon 8 mutation of p53 gene associated with nodal metastasis in non-small-cell lung cancer. Am J Respir Crit Care Med 150:1667–1671. , DOI 10.1164/ ajrccm.150.6.7952630 Kozomara R, Brankovic-Magic M, Jovic N, Stosic S, Magic Z, 2007, Prognostic significance of TP53 mutations in oral squamous cell carcinoma with human papilloma virus infection. Int J Biol Markers 22:252–257. , DOI 10.1177/ 172460080702200403 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1191 EP 1199 DI 10.1007/s12253-019-00673-6 PG 9 ER PT J AU Zhang, W Liu, M Li, Y Song, Sh Li, K Ma, Y AF Zhang, Wei Liu, Mingkai Li, Yue Song, Shichao Li, Kai Ma, Yongliang TI The Role of O6-methylguanine-DNA Methyltransferase Polymorphisms in Prostate Cancer Susceptibility: a Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MGMT; PCa; Prostate cancer; Risk; Meta-analysis ID MGMT; PCa; Prostate cancer; Risk; Meta-analysis AB To assess the associations between O6-methylguanine-DNA methyltransferase (MGMT) polymorphisms and prostate cancer risk. We retrieved PubMed, Cochrane Library and Embase electronic database to search for all eligible studies published from Jan 1, 1970 to Sep 31, 2017 to conduct a Meta-analysis. We identified 11 independent studies in 5 eligible reports, including 5143 cases and 8118 controls. The data suggested that rs12917 was associated with higher PCa risk under the contrast of TT vs CC and recessive model in overall population (TT vs CC: OR = 1.599, 95%CI: 1.007–2.539, P = 0.047; TT vs CC + CT: OR = 1.627, 95%CI: 1.026–2.580, P = 0.038). In subgroup analyses stratified by ethnicity, the remarkable association with higher PCa risk was detected under allelic model, dominant model, the contrast of TC vs CC, and the contrast of TC vs CC + TT in Asian population. (T vs C: OR = 1.911, 95%CI: 1.182–3.090, P = 0.008; TC vs CC: OR = 1.948, 95%CI: 1.152–3.295, P = 0.013; TC + TT vs CC: OR = 1.994, 95%CI: 1.190–3.342, P = 0.009; TC vs CC + TT: OR = 1.926, 95%CI: 1.140–3.255, P = 0.014). However, the data suggest the rs2308327 and rs2308321 polymorphisms of the MGMT gene were nor associated with the susceptibility of prostate cancer. Based on the meta-analysis, MGMT rs12917 polymorphism increase the susceptibility to prostate cancer, which can be taken for a diagnosis and screening molecular biomarker for prostate cancer patients. C1 [Zhang, Wei] Affiliated Hospital of Hebei University, Department of Urology, 071000 Baoding, China. [Liu, Mingkai] Affiliated Hospital of Hebei University, Department of Urology, 071000 Baoding, China. [Li, Yue] Affiliated Hospital of Hebei University, Department of Urology, 071000 Baoding, China. [Song, Shichao] Affiliated Hospital of Hebei University, Department of Urology, 071000 Baoding, China. [Li, Kai] Affiliated Hospital of Hebei University, Department of Urology, 071000 Baoding, China. [Ma, Yongliang] Fourth Hospital of Hebei Medical University, Department of Urology, 050011 Shijiazhuang, China. RP Ma, Y (reprint author), Fourth Hospital of Hebei Medical University, Department of Urology, 050011 Shijiazhuang, China. EM 284569670@qq.com CR Hung RJ, Baragatti M, Thomas D, McKay J, Szeszenia-Dabrowska N, Zaridze D, Lissowska J, Rudnai P, Fabianova E, Mates D, Foretova L, Janout V, Bencko V, Chabrier A, Moullan N, Canzian F, Hall J, Boffetta P, Brennan P, 2007, Inherited predisposition of lung cancer: a hierarchical modeling approach to DNA repair and cell cycle control pathways. Cancer Epidem Biomar 16(12):2736– 2744 Chae MH, Jang JS, Kang HG, Park JH, Park JM, LeeWK, Kam S, Lee EB, Son JW, Park JY, 2006)O6-alkylguanine-DNA alkyltransferase gene polymorphisms and the risk of primary lung cancer. Mol Carcinog 45(4):239–249 Daniels DS, Woo TT, Luu KX, Noll DM, Clarke ND, Pegg AE, Tainer JA, 2004, DNA binding and nucleotide flipping by the human DNA repair protein AGT. Nat Struct Mol Biol 11(8):714–720 CalegariMA, Inno A, Monterisi S, Orlandi A, Santini D, Basso M, Cassano A, Martini M, Cenci T, de Pascalis I, Camarda F, Barbaro B, Larocca LM, Gori S, Tonini G, Barone C, 2017, A phase 2 study of temozolomide in pretreated metastatic colorectal cancer with MGMT promoter methylation. Br J Cancer 116(10):1279–1286 Chen GD, Qian DY, Li ZG, Fan GY, You KL, Wu YL, 2017)Down-regulation of p16 and MGMT promotes the antiproliferative and pro-apoptotic effects of 5-Aza-dC and radiation on cervical cancer cells. Cell Biochem Funct 35(8):488–496 Tomaszowski KH, Hellmann N, Ponath V, Takatsu H, Shin HW, Kaina B, 2017, Uptake of glucose-conjugated MGMTinhibitors in cancer cells: role of flippases and type IV P-type ATPases. Sci Rep 7(1):13925 Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016. CA Cancer J Clin 66(1):7–30 Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(1):7–30 Liu TZ, Guo ZQ,Wang T, CaoY, Huang D,Wang XH(2017)Metaanalysis of the role of IL-6 rs1800795 polymorphism in the susceptibility to prostate cancer: evidence based on 17 studies. Medicine 96(11):e6126 Zhang L, Zhang Y, Zhang X, Hong B, 2015)Prostaglandinendoperoxide synthase 2, PTGS2, rs20417 polymorphism and prostate cancer risk: a meta analysis. Int J Clin Exp Med 8(11): 20454–20462 Agalliu I, Kwon EM, Salinas CA, Koopmeiners JS, Ostrander EA, Stanford JL, 2010, Genetic variation in DNA repair genes and prostate cancer risk: results from a population-based study. Cancer Causes & Control 21(2):289–300 Loh YH, Mitrou PN, Bowman R, Wood A, Jeffery H, Luben RN, Lentjes MA, Khaw KT, Rodwell SA, 2010, MGMT Ile143Val polymorphism, dietary factors and the risk of breast, colorectal and prostate cancer in the European prospective investigation into cancer and nutrition, EPIC)-Norfolk study. DNA Repair 9(4):421–428 Loh YH,Mitrou PN,Wood A, Luben RN, McTaggart A, Khaw KT, Rodwell SA, 2011, SMAD7 and MGMT genotype variants and cancer incidence in the European prospective investigation into Cancer and nutrition, EPIC)-Norfolk study. Cancer Epidemiol 35(4):369–374 Matullo G, Dunning AM, Guarrera S, Baynes C, Polidoro S, Garte S, Autrup H, Malaveille C, PelusoM, Airoldi L, Veglia F, Gormally E, Hoek G, Krzyzanowski M, Overvad K, Raaschou-Nielsen O, Clavel-Chapelon F, Linseisen J, Boeing H, Trichopoulou A, Palli D, Krogh V, Tumino R, Panico S, Bueno-De-Mesquita HB, Peeters PH, Lund E, Pera G, Martinez C, Dorronsoro M, Barricarte A, Tormo MJ, Quiros JR, Day NE, Key TJ, Saracci R, Kaaks R, Riboli E, Vineis P, 2006, DNA repair polymorphisms and cancer risk in non-smokers in a cohort study. Carcinogenesis 27(5):997–1007 Ritchey JD, Huang WY, Chokkalingam AP, Gao YT, Deng J, Levine P, Stanczyk FZ, Hsing AW, 2005, Genetic variants of DNA repair genes and prostate cancer: a population-based study. Cancer Epidem Biomar 14(7):1703–1709 Chuang LY, Chang HW, Lin MC, Yang CH, 2012, Chaotic particle swarm optimization for detecting SNP-SNP interactions for CXCL12-related genes in breast cancer prevention. European journal of cancer prevention : the official journal of the European Cancer Prevention Organisation, ECP, 21(4):336–342 Goodman JE, Mechanic LE, Luke BT, Ambs S, Chanock S, Harris CC, 2006, Exploring SNP-SNP interactions and colon cancer risk using polymorphism interaction analysis. Int J Cancer 118(7): 1790–1797 Schwender H, Selinski S, Blaszkewicz M, Marchan R, Ickstadt K, Golka K, Hengstler JG, 2012, Distinct SNP combinations confer susceptibility to urinary bladder cancer in smokers and nonsmokers. PLoS One 7(12):e51880 Hu Z,Wang H, Shao M, Jin G, SunW,WangY, Liu H,WangY, Ma H, Qian J, Jin L, Wei Q, Lu D, Huang W, Shen H, 2007, Genetic variants in MGMTand risk of lung cancer in southeastern Chinese: a haplotype-based analysis. Hum Mutat 28(5):431–440 Huang J, Ye F, Chen H, Lu W, Xie X, 2007, Amino acid substitution polymorphisms of the DNA repair gene MGMT and the susceptibility to cervical carcinoma. Carcinogenesis 28(6):1314–1322 Krzesniak M, Butkiewicz D, Samojedny A, Chorazy M, Rusin M, 2004, Polymorphisms in TDG and MGMT genes - epidemiological and functional study in lung cancer patients from Poland. Ann Hum Genet 68(Pt 4):300–312 Li C, Liu J, Li A, Qian L,Wang X,Wei Q, Zhou J, Zhang Z, 2005, Exon 3 polymorphisms and haplotypes of O6-methylguanine-DNA methyltransferase and risk of bladder cancer in southern China: a case-control analysis. Cancer Lett 227(1):49–57 Liu Y, Scheurer ME, El-Zein R, Cao Y, Do KA, Gilbert M, Aldape KD, Wei Q, Etzel C, Bondy ML, 2009, Association and interactions between DNA repair gene polymorphisms and adult glioma. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology 18(1):204–214 Shen J, TerryMB, GammonMD, GaudetMM, TeitelbaumSL, Eng SM, Sagiv SK, Neugut AI, Santella RM, 2005, MGMT genotype modulates the associations between cigarette smoking, dietary antioxidants and breast cancer risk. Carcinogenesis 26(12): 2131–2137 Shen M, Purdue MP, Kricker A, Lan Q, Grulich AE, Vajdic CM, Turner J,Whitby D, Chanock S, Rothman N, ArmstrongBK, 2007, Polymorphisms in DNA repair genes and risk of non-Hodgkin's lymphoma in New South Wales, Australia. Haematologica 92(9): 1180–1185 Huang WY, Chow WH, Rothman N, Lissowska J, Llaca V, Yeager M, Zatonski W, Hayes RB, 2005, Selected DNA repair polymorphisms and gastric cancer in Poland. Carcinogenesis 26(8):1354–1359 Wang L, Liu H, Zhang Z, Spitz MR, Wei Q, 2006, Association of genetic variants of O6-methylguanine-DNAmethyltransferase with risk of lung cancer in non-Hispanic whites. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology 15(12):2364–2369 Liu J, Zhang R, Chen F, Yu C, Sun Y, Jia C, Zhang L, Salahuddin T, Li X, Lang J, Song X, 2013, MGMT Leu84Phe polymorphism contributes to cancer susceptibility: evidence from 44 case-control studies. PLoS One 8(9):e75367 Qiu ZX, Xue F, Shi XF, He X, Ma HN, Chen L, Chen PZ, 2014, MGMT Leu84Phe gene polymorphism and lung cancer risk: a meta- analysis. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine 35(5): 4381–4387 Zhong Y, Huang Y, Huang Y, Zhang T, Ma C, Zhang S, Fan W, Chen H, Qian J, Lu D, 2010, Effects of O6-methylguanine-DNA methyltransferase, MGMT, polymorphisms on cancer: a metaanalysis. Mutagenesis 25(1):83–95 Hirschhorn JN, Lohmueller K, Byrne E, Hirschhorn K, 2002, A comprehensive review of genetic association studies. Genetics in medicine : official journal of the American College of Medical Genetics 4(2):45–61 Rodriguez C, Calle EE, Miracle-McMahill HL, TathamLM,Wingo PA, Thun MJ, Heath CW Jr, 1997, Family history and risk of fatal prostate cancer. Epidemiology, Cambridge, Mass, 8(6):653–657 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1201 EP 1209 DI 10.1007/s12253-019-00672-7 PG 9 ER PT J AU Stec, R Cierniak, Sz Lubas, A Brzoskowska, U Syrylo, T Zielinski, H Semeniuk-Wojtas, A AF Stec, Rafal Cierniak, Szczepan Lubas, Arkadiusz Brzoskowska, Urszula Syrylo, Tomasz Zielinski, Henryk Semeniuk-Wojtas, Aleksandra TI Intensity of Nuclear Staining for Ki-67, p53 and Survivin as a New Prognostic Factor in Non-muscle Invasive Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; NMIBC; Ki-67; p53; Survivin ID Bladder cancer; NMIBC; Ki-67; p53; Survivin AB The aim of the study was to determine the prognostic value of expression levels of biomarkers selected on the basis of the literature: p53, Ki-67, survivin, β-catenin, E-cadherin and N-cadherin in patients with non-muscle invasive bladder cancer. Immunohistochemistry was performed on sections of primary papillary carcinoma of the bladder removed during transurethral resection of the tumor in 134 patients. The expression of β-catenin and E-cadherin was found in all analyzed cases and Ncadherin expression was demonstrated in 3.73% of the tissues examined. The expression of the p53 protein was confirmed in 96.27% of tissues examined. The expression of the Ki-67 protein was demonstrated in all analyzed cases. Survivin expression was found in 95.52% of the study group. Multivariate analysis confirmed the relationship between the recurrence-free survival (RFS) and the intensity of the nuclear reaction for p53 (HR 1417, 95% CI 1.001–2.007, p = 0.049) and survivin (HR 1.451; 95% CI 1.078–1.955; p = 0.014), the expression level of the Ki-67 protein expressed by the TS index (HR 1.146, 95%CI 1.116–1.823, p = 0.005) and the use of adjuvant BCG therapy (HR 0.218, 95% CI 0.097–0.489, p = 0.0002). The evaluation of Ki-67 expression and the intensity of nuclear staining for survivin and p53 may provide additional information that will allow more accurate stratification of the risk of NMIBC recurrence after TURBT. C1 [Stec, Rafal] Medical University of Warsaw, Oncology Department, 19/25 Stepinska St., 00-739 Warsaw, Poland. [Cierniak, Szczepan] Military Institute of Medicine, Pathomorphology Department, 128 Szaserow St., 04-141 Warsaw, Poland. [Lubas, Arkadiusz] Military Institute of Medicine, Internal Diseases, Nephrology and Dialysis Department, 128 Szaserow St., 04-141 Warsaw, Poland. [Brzoskowska, Urszula] Military Institute of Medicine, Pathomorphology Department, 128 Szaserow St., 04-141 Warsaw, Poland. [Syrylo, Tomasz] Military Institute of Medicine, General, Functional and Oncological Urology Department, 128 Szaserow St., 04-141 Warsaw, Poland. [Zielinski, Henryk] Military Institute of Medicine, General, Functional and Oncological Urology Department, 128 Szaserow St., 04-141 Warsaw, Poland. [Semeniuk-Wojtas, Aleksandra] Military Institute of Medicine, Oncology Department, 128 Szaserow St., 04-141 Warsaw, Poland. RP Semeniuk-Wojtas, A (reprint author), Military Institute of Medicine, Oncology Department, 04-141 Warsaw, Poland. EM osemeniuk@wp.pl CR Alsheikh A,Mohamedali Z, Jones E, Masterson J, Gilks CB, 2001, Comparison of the WHO/ISUP classification and cytokeratin 20 expression in predicting the behavior of low-grade papillary urothelial tumors. Mod Pathol 14:267–272 Sylvester RJ, van der Meijden AP, Oosterlinck W et al, 2006, Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials. Eur Urol 49(3): 466–477 Zieger K,Wolf H, Olsen PR, Hojgaard K, 2000, Long-term followup of noninvasive bladder tumours, stage Ta): recurrence and progression. BJU Int 85(7):824–828 Fernandez-Gomez J, Madero R, Solsona E, Unda M, Martinez- Pineiro L, Gonzalez M, Portillo J, Ojea A, Pertusa C, Rodriguez- Molina J, Camacho JE, Rabadan M, Astobieta A, Montesinos M, Isorna S, Muntanola P, Gimeno A, Blas M, Martinez-Pineiro JA, 2009, Predicting nonmuscle invasive bladder cancer recurrence and progression in patients treated with bacillus Calmetie-Guerin: the CUETO scoring model. J Urol 182(5):2195–2203 Xylinas E, Kent M, Kluth L, Pycha A, Comploj E, Svatek RS, Lotan Y, Trinh QD, Karakiewicz PI, Holmang S, Scherr DS, Zerbib M, Vickers AJ, Shariat SF, 2013, Accuracy of the EORTC risk tables and of the CUETO scoring model to predict outcomes in non-muscle-invasive urothelial carcinoma of the bladder. Br J Cancer 109:1460–1466 Sznarkowska A, Olszewski R, Zawadzka-Pankau J, 2010, Farmakologiczna aktywacja supresora nowotworu, natywnego bialka p53 jako obiecujaca strategia zwalczania nowotworow. Postepy Hig Med Dosw 64:396–407 Giaccia AJ, Kastan MB, 1998, The complexity of p53 modulation: emerging patterns from divergent signals. Genes Dev 12(19):2973– 2983 Gao J, Huang HY, Pak J, 2004, i wsp. p53 deficiency provokes urothelial proliferation and synergizes with activated Ha-ras in promoting urothelial tumorigenesis. Oncogene 23:687–696 Wesierska-Gadek J, Bednarek J, Kilianska ZM, 2007, Nowe oblicze bialek antyapoptotycznych. II. Surwiwina. Postepy Biochem 53(3):239–253 Urbaniak J, 2004, Ekspresja surwiwiny w nowotworach ludzkich. Adv Clin Exp Med 13(6):1037–1046 Zhang B, Lu Z, Hou Y, Hu J,Wang C, 2014, The effects of STAT3 and Survivin silencing on the growth of human bladder carcinoma cells. Tumour Biol 35(6):5401–5407 Scholzen T, Gerdes J, 2000 Mar, The Ki-67 protein: from the known and the unknown. J Cell Physiol 182(3):311–322 Balcerak A, Wakula M, Trebinska A, Grzybowska EA, 2016, Migracja i inwazyjnosc komorek nowotworowych; rola plastycznosci komorek i udzial macierzy zewnatrzkomorkowej w tworzeniu przerzutow. NOWOTWORY Journal of Oncology 66(1):45–52 Lamparska-Przybysz M, Wieczorek M, Majorek M, Guzenda P, 2006, Rola szlaku Wnt/β-katenina w molekularnym mechanizmie procesow nowotworowych.Wspolczesna Onkologia 10(10):497–501 Tian X, Liu Z, Niu B et al, 2011, E-cadherin/β-catenin complex and the epithelial barrier. J Biomed Biotechnol 2011:567305 Ahmad I, Morton JP, Singh LB, Radulescu SM, Ridgway RA, Patel S, Woodgett J, Winton DJ, Taketo MM, Wu XR, Leung HY, Sansom OJ, 2011, B-Catenin activation synergizes with PTEN loss to cause bladder cancer formation. Oncogene 30:178–189 Allred DC, Harvey JM, Berardo M, Clark GM, 1998, Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11(2):155–168 Elsherif E, Elbaky TA, Elserafy F et al, 2016, β-Catenin and SKP2 proteins as predictors of grade and stage of non-muscle invasive urothelial bladder carcinoma. Chin Clin Oncol 5(1):6 Senol S, Yildirim A, Ceyran B et al, 2015, Prognostic significance of survivin, β-catenin and p53 expression in urothelial carcinoma. Bosn J Basic Med Sci 15(4):7–14 Koletsas N, Koletsa T, Choidas S et al, 2017, Immunohistochemical investigation of HER/AKT/mTOR pathway and cellular adhesion molecules in urothelial carcinomas. Pathol Res Int 2017:6794150 Singh R, Ansari JA, Maurya N, Mandhani A, Agrawal V, Garg M, 2017, Epithelial-to-mesenchymal transition and its correlation with Clinicopathologic features in patients with urothelial carcinoma of the bladder. Clin Genitourin Cancer 15(2):e187–e197 Muramaki M, Miyake H, Terakawa T, Kumano M, Sakai I, Fujisawa M, 2012, Expression profile of E-cadherin and Ncadherin in non-muscle-invasive bladder cancer as a novel predictor of intravesical recurrence following transurethral resection. Urol Oncol 30(2):161–166 AbufarajM, Shariat SF, Haitel A,Moschini M, Foerster B, Chlosta P, Gust K, Babjuk M, Briganti A, Karakiewicz PI, Albrecht W, 2017, Prognostic role of N-cadherin expression in patients with non-muscle-invasive bladder cancer. Urol Oncol 35(5):264–271 Chen JX, Deng N, Chen X, Chen LW, Qiu SP, Li XF, Li JP, 2012, A novel molecular grading model: combination of Ki67 and VEGF in predicting tumor recurrence and progression in non-invasive urothelial bladder cancer. Asian Pac J Cancer Prev 13(5):2229–2234 Vetterlein MW, Roschinski J, Gild P, Marks P, Soave A, Doh O, Isbarn H, Hoppner W, Wagner W, Shariat SF, Brausi M, Buscheck F, Sauter G, Fisch M, Rink M, 2017, Impact of the Ki-67 labeling index and p53 expression status on disease-free survival in pT1 urothelial carcinoma of the bladder. Transl Androl Urol 6(6): 1018–1026 Xi RC, Sheng YR, Chen WH, Sheng L, Gang JJ, Tong Z, Shan Z, Ying GH, Dong LC, 2013, Expression of survivin and livin predicts early recurrence in non-muscle invasive bladder cancer. J Surg Oncol 107(5):550–554 Skagias L, Politi E, Karameris A, Sambaziotis D, Archondakis A, Ntinis A, Moreas I, Vasou O, Koutselini H, Patsouris E, 2009, Survivin expression as a strong indicator of recurrence in urothelial bladder cancer. Anticancer Res 29:4163–4167 Jeon C, KimM,Kwak C, KimHH, Ku JH, 2013, Prognostic role of survivin in bladder cancer: a systematic review and meta-analysis. PLoS One 8(10):e76719 Van de Vijver MJ, 2001, Assessment of the need and appropriate method for testing for the human epidermal growth factor receptor- 2, HER2). Eur J Cancer 37:11–17 Wang L, Feng C, Ding G, Zhou Z, Jiang H, Wu Z, 2013, Relationship of TP53 and Ki67 expression in bladder cancer under WHO 2004 classification. J BUON 18(2):420–424 Ben Abdelkrim S, Rammeh S, Ziadi S, Tlili T, JaidaneM, MokniM, 2014, Expression of topoisomerase II alpha, ki67, and p53 in primary non-muscle-invasive urothelial bladder carcinoma. J Immunoass Immunochem 35(4):358–367 Thakur B, Kishore S, Dutta K, Kaushik S, Bhardwaj A, 2017, Role of p53 and Ki-67 immunomarkers in carcinoma of urinary bladder. Indian J Pathol Microbiol 60(4):505–509 Qamar S, Inam QA, Ashraf S et al, 2017, Prognostic value of p53 expression intensity in urothelial cancers. J Coll Physicians Surg Pak 27(4):232–236 Lv S, Turlova E, Zhao S, Kang H, Han M, Sun HS, 2014, Prognostic and clinicopathological significance of survivin expression in bladder cancer patients: a meta-analysis. Tumour Biol 35(2): 1565–1574 He Y, Wang N, Zhou X, Wang J, Ding Z, Chen X, Deng Y, 2018, Prognostic value of ki67 in BCG-treated non-muscle invasive bladder cancer: a meta-analysis and systematic review. BMI Open 8(4): e019635 Zhou X, Zhang G, Tian Y, 2015, p53 status correlates with the risk of recurrence in non-muscle invasive bladder cancers treated with Bacillus Calmette-Guerin: a meta-analysis. PLoS One 10(3): e0119476 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1211 EP 1219 DI 10.1007/s12253-019-00678-1 PG 9 ER PT J AU Mignon, S Willard-Gallo, K Van den Eynden, G Salgado, R Decoster, L Marien, MK Vansteenkiste, FJ Teugels, E De Greve, J AF Mignon, Sacha Willard-Gallo, Karen Van den Eynden, Gert Salgado, Roberto Decoster, Lore Marien, M Koen Vansteenkiste, F Johan Teugels, Erik De Greve, Jacques TI The Relationship Between Tumor-Infiltrating Lymphocytes, PD-L1 Expression, Driver Mutations and Clinical Outcome Parameters in Non-Small Cell Lung Cancer Adenocarcinoma in Patients with a Limited to no Smoking History SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung adenocarcinoma; Tumor infiltrating lymphocytes (TIL); Programmed death-ligand 1 (PD-L1); Programmed death-1 (PD-1); Driver mutations ID Lung adenocarcinoma; Tumor infiltrating lymphocytes (TIL); Programmed death-ligand 1 (PD-L1); Programmed death-1 (PD-1); Driver mutations AB Tumor infiltrating lymphocytes (TIL), programmed death 1 (PD-1) and programmed death-ligand 1 (PD-L1) expression are important prognostic markers. This study aimed to investigate these markers in lung adenocarcinoma (ADC) biopsies from patients with stage IIIB or IVADC with little or no smoking history, to investigate their prognostic value and to correlate these results with the presence of driver mutations in the tumors. TIL were retrospectively evaluated on hematoxylin and eosin stained slides from 152 tumor samples. PD-1/PD-L1 expression was retrospectively evaluated with PD-1/PD-L1 immunohistochemistry (IHC) double staining on 74 tumor samples with sufficient residual tissue. PD-L1 expression was analysed on stromal cells of the tumor compartment, the tumor cells and TIL and PD-1 on TIL. Median overall survival (OS) was longer in patients with high stromal TIL infiltration compared to patients with low stromal TIL infiltration (68 weeks vs. 35 weeks respectively; p = 0.003). This was observed most prominently in KRAS mutant tumors (95 weeks vs. 12 weeks; p = 0.003). Only PD-L1 expression on tumor stromal cells influenced OS and indicated a worse prognosis (77 weeks vs 25 weeks; p = 0.013). Stromal TIL counts nor PD-1/PD-L1 expression levels were associated with the presence of driver mutations. The results of the current study reinforce the prognostic role of TIL in lung ADC, which is most prominent in KRAS mutant cancers. The results of the PD-1/PD-L1 analysis suggest that stromal cells can effectively suppress the anti-tumor immune response via the PD-L1 pathway. C1 [Mignon, Sacha] Universitair Ziekenhuis Brussel, Oncologisch Centrum UZ Brussel, Departement of Medical Oncology, Laarbeeklaan 101, 1090 Brussels, Belgium. [Willard-Gallo, Karen] Institut Jules Bordet, Molecular Immunology Laboratory, Boulevard de Waterloo 121, 1000 Brussels, Belgium. [Van den Eynden, Gert] Institut Jules Bordet, Molecular Immunology Laboratory, Boulevard de Waterloo 121, 1000 Brussels, Belgium. [Salgado, Roberto] Institut Jules Bordet, Molecular Immunology Laboratory, Boulevard de Waterloo 121, 1000 Brussels, Belgium. [Decoster, Lore] Universitair Ziekenhuis Brussel, Oncologisch Centrum UZ Brussel, Departement of Medical Oncology, Laarbeeklaan 101, 1090 Brussels, Belgium. [Marien, M Koen] Universiteit Antwerpen, Prinsstraat 13, 2000 Antwerp, Belgium. [Vansteenkiste, F Johan] University Hospital KU Leuven, Department Respiratory Oncology, Herestraat 49, 3000 Leuven, Belgium. [Teugels, Erik] Universitair Ziekenhuis Brussel, Oncologisch Centrum UZ Brussel, Departement of Medical Oncology, Laarbeeklaan 101, 1090 Brussels, Belgium. [De Greve, Jacques] Universitair Ziekenhuis Brussel, Oncologisch Centrum UZ Brussel, Departement of Medical Oncology, Laarbeeklaan 101, 1090 Brussels, Belgium. RP Mignon, S (reprint author), Universitair Ziekenhuis Brussel, Oncologisch Centrum UZ Brussel, Departement of Medical Oncology, 1090 Brussels, Belgium. EM sacha.mignon@uzbrussel.be CR Reck M, Paz-Ares L, 2015, Immunologic checkpoint blockade in lung Cancer. Semin Oncol Elsevier 42(3):402–417 Wu P, Wu D, Li L, Chai Y, Huang J, 2015, PD-L1 and survival in solid tumors: a meta-analysis. PLoS One 10(6):e0131403 Scagliotti GV, Bironzo P, Vansteenkiste J, 2015, Addressing the unmet need in lung cancer: the potential of immuno-oncology. Cancer Treat Rev 41:465–475 Pan Z-K, Ye F,Wu X, An H-X,Wu J-X, 2015, Clinicopathological and prognostic significance of programmed cell death ligand1, PDL1, expression in patients with non-small cell lung cancer: a metaanalysis. J Thorac Dis 7(3):462–470 Schmidt LH, Kummel A, Gorlich D, Mohr M, Brockling S, Mikesch JH, Grunewald I, Marra A, Schultheis AM, Wardelmann E, Muller-Tidow C, Spieker T, Schliemann C, Berdel WE, Wiewrodt R, Hartmann W, 2015, PD-1 and PD-L1 expression in NSCLC indicate a favorable prognosis in defined subgroups. PLoS One 10(8):e0136023 Velcheti V, Schalper KA, Carvajal DE, Anagnostou VK, Syrigos KN, Sznol M, Herbst RS, Gettinger SN, Chen L, Rimm DL, 2014, Programmed death ligand-1 expression in non-small cell lung cancer. Lab Investig 94(1):107–116 Mittal D, Gubin MM, Schreiber RD, Smyth MJ, 2015, New insights into cancer immunoediting and its three component phases - elimation, equilibrium and escape. Curr Opin Immunol 27:16–25 Schalper KA, Brown J, Carvajal-Hausdorf D, McLaughlin J, Velcheti V, Syrigos KN et al, 2015, Objective measurement and clinical significance of TILs in non-small cell lung cancer. JNCI J Natl Cancer Inst 107(3):dju435–dju435 Bremnes RM, Busund L-T, Kilvaer TL, Andersen S, Richardsen E, Paulsen EE et al, 2016, The role of tumor infiltrating lymphocytes in development, progression and prognosis of non-small cell lung cancer. J Thorac Oncol 11(6): 789-800 Geng Y, Shao Y, HeW, HuW, Xu Y, Chen J,Wu C, Jiang J, 2015, Prognostic role of tumor-infiltrating lymphocytes in lung Cancer: a meta-analysis. Cell Physiol Biochem 37:1560–1571 Brambilla E, Le Teuff G, Marguet S, Lantuejoul S, Dunant A, Graziano S et al, 2016, Prognostic effect of tumor lymphocytic infiltration in Resectable non-small-cell lung Cancer. J Clin Oncol 34:1223–1230 De Greve J,Van Meerbeeck J, Van Steenkiste J, Teugels E, Geers C, Meert A et al, 2016, Prospective evaluation of first-line Erlotinib in advanced non-small cell lung Cancer, NSCLC, carrying an activating EGFR mutation: a multicenter academic phase II study in Caucasian patients, FIELT). PLoS One 11(3):e0147599 Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, van den Eynden G, Baehner FL, Penault-Llorca F, Perez EA, Thompson EA, SymmansWF, Richardson AL, Brock J, Criscitiello C, Bailey H, Ignatiadis M, Floris G, Sparano J, Kos Z, Nielsen T, Rimm DL, Allison KH, Reis-Filho JS, Loibl S, Sotiriou C, Viale G, Badve S, Adams S, Willard-Gallo K, Loi S, International TILs Working Group 2014, 2015, The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: recommendations by an international TILs working group 2014. Ann Oncol Buisseret L, Garaud S, deWind A, Van den Eynden G, Boisson A, Solinas C et al, 2017, Tumor infiltrating lymphocyte composition, organization and PD-1/PD-L1 expression are linked in breast cancer. Oncoimmunology. 6(1):e1257452 R core team. R: a language and environment for statistical computing. R Foundation for statistical computing, Vienna. 2015. Available from: Last https://www.r-project.org/. Accessed 30 July /2016 Gamer M, Lemon J, Fellows I, Singh P., 2012, Package ‘irr’. Available from:. Last https://cran.r-project.org/web/packages/irr/ irr.pdf. Accessed on 30 July 2016 Kirkegaard T, Edwards J, Tovey S, McGlynn LM, Krishna SN, Mukherjee R et al, 2006, Observer variation in immunohistochemical analysis of protein expression, time for a change? Histopathology. 48(7):787–794 Hendry S, Salgado R, Thomas G, 2017, Assessing tumorinfiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the international Immuno-oncology biomarkers working group: part 2: TILs in melanoma, gastrointestinal tract Carcinom. Adv Anat Pathol 24(6):311–335 Toki M, Mani N, Smithy J, Liu Y, Altan M, Wasserman B et al, 2018, Immune marker profiling and programmed death ligand 1 expression across NSCLCmutations. J Thorac Oncol 13(12):1884– 1896 Paz-Ares L, Horn L, Borghaei H, Spigel D, Steins M, Ready N et al, 2015, Phase III, randomized trial, CheckMate 057, of nivolumab, NIVO, versus docetaxel, DOC, in advanced non-squamous cell, non-SQ, non-small cell lung cancer, NSCLC). J Clin Oncol 33: LBA109 Akbay EA, Koyama S, Carretero J, Altabef A, Tchaicha JH, Christensen CL, Mikse OR, Cherniack AD, Beauchamp EM, Pugh TJ, Wilkerson MD, Fecci PE, Butaney M, Reibel JB, Soucheray M, Cohoon TJ, Janne PA, Meyerson M, Hayes DN, Shapiro GI, Shimamura T, Sholl LM, Rodig SJ, Freeman GJ, Hammerman PS, Dranoff G, Wong KK, 2013, Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov 3(12):1355–1363 Azuma K, Ota K, Kawahara a HS, Iwama E, Harada T et al, 2014, Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann Oncol 25:1935–1940 Wang A,Wang HY, Liu Y, ZhaoMC, Zhang HJ, Lu ZYet al, 2015, The prognostic value of PD-L1 expression for non-small cell lung cancer patients: a meta-analysis. J Cancer Surg 41:450–456 He J, Hu Y, Hu M, Li B, 2015, Development of PD-1/PD-L1 pathway in tumor immune microenvironment and treatment for non-small cell lung cancer. Sci Rep 5(august):13110 Lin C, Chen X, Li M, Liu J, Qi X, Yang W, et al., 2015, Programmed death-ligand 1 expression predicts tyrosine kinase inhibitor response and better prognosis in a cohort of patients with epidermal growth factor receptor mutation-positive lung adenocarcinoma. Clin Lung Cancer 16(5): e25-35 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1221 EP 1228 DI 10.1007/s12253-019-00670-9 PG 8 ER PT J AU Oscorbin, PI Shadrina, SA Kozlov, VV Voitsitsky, EV Filipenko, LM AF Oscorbin, P Igor Shadrina, S Alexandra Kozlov, V Vadim Voitsitsky, E Vladimir Filipenko, L Maxim TI Absence of EGFR C797S Mutation in Tyrosine Kinase Inhibitor-Naive Non–Small Cell Lung Cancer Tissues SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; EGFR; C797S; Osimertinib; Droplet digital PCR ID Non-small cell lung cancer; EGFR; C797S; Osimertinib; Droplet digital PCR AB EGFR tyrosine-kinase inhibitors (TKIs) are used as targeted therapeutics for the treatment of advanced non–small cell lung cancer (NSCLC) with EGFR-activating mutations. EGFR C797S is common causes of acquired resistance to third generation TKIs. There is wide-spread opinion that resistance-conferring mutation present even in a small proportion of cancer cells before the start of therapy could potentially predict poor response to a targeted drug. In our study, we tested whether C797S can be found in previously untreated NSCLCs. We analyzed DNA samples extracted from formalin-fixed paraffin-embedded (FFPE) tumor tissue sections of 470 lung adenocarcinoma patients, including 235 samples with activating EGFR mutations. Screening was performed using highly sensitive droplet digital PCR assay. No tumor samples with baseline C797S were identified. C797S does not occur in TKI-naive NSCLCs and provide evidence that screening for this mutation before TKIs administration may not be necessary. C1 [Oscorbin, P Igor] Institute of Chemical Biology and Fundamental Medicine, Laboratory of Pharmacogenomics, 8 Lavrentiev Avenue, 630090 Novosibirsk, Russian Federation. [Shadrina, S Alexandra] Novosibirsk State University, 2 Pirogova Street, 630090 Novosibirsk, Russian Federation. [Kozlov, V Vadim] Institute of Cytology and Genetics, 10 Lavrentiev Avenue, 630090 Novosibirsk, Russian Federation. [Voitsitsky, E Vladimir] Institute of Cytology and Genetics, 10 Lavrentiev Avenue, 630090 Novosibirsk, Russian Federation. [Filipenko, L Maxim] Institute of Chemical Biology and Fundamental Medicine, Laboratory of Pharmacogenomics, 8 Lavrentiev Avenue, 630090 Novosibirsk, Russian Federation. RP Oscorbin, PI (reprint author), Institute of Chemical Biology and Fundamental Medicine, Laboratory of Pharmacogenomics, 630090 Novosibirsk, Russian Federation. EM osc.igor@gmail.com CR Metro G, 2018, EGFR targeted therapy for lung cancer: are we almost there? Transl Lung Cancer Res 7:S142–S145. https://doi. org/10.21037/tlcr.2018.03.11 Bollinger MK, Agnew AS, Mascara GP, 2018, Osimertinib: a third-generation tyrosine kinase inhibitor for treatment of epidermal growth factor receptor-mutated non-small cell lung cancer with the acquired Thr790Met mutation. J Oncol Pharm Pract 24:379–388. , DOI 10.1177/1078155217712401 Lu X, Yu L, Zhang Z, Ren X, Smaill JB, Ding K, 2018, Targeting EGFR L858R/T790M and EGFR L858R/T790M/C797S resistance mutations in NSCLC: current developments in medicinal chemistry. Med Res Rev 38:1550–1581. , DOI 10.1002/med.21488 Camidge DR, Pao W, Sequist LV, 2014, Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nat Rev Clin Oncol 11:473–481. , DOI 10.1038/nrclinonc.2014.104 Soria J-C, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, Dechaphunkul A, Imamura F, Nogami N, Kurata T, Okamoto I, Zhou C, Cho BC, Cheng Y, Cho EK, Voon PJ, Planchard D, Su WC, Gray JE, Lee SM, Hodge R, Marotti M, Rukazenkov Y, Ramalingam SS, FLAURA Investigators, 2018, Osimertinib in untreated EGFR -mutated advanced non–small-cell lung cancer. N Engl J Med 378:113–125. , DOI 10.1056/NEJMoa1713137 Walter AO, Sjin RTT, Haringsma HJ, Ohashi K, Sun J, Lee K, Dubrovskiy A, Labenski M, Zhu Z, Wang Z, Sheets M, St Martin T, Karp R, van Kalken D, Chaturvedi P, Niu D, Nacht M, Petter RC, Westlin W, Lin K, Jaw-Tsai S, Raponi M, van Dyke T, Etter J, Weaver Z, Pao W, Singh J, Simmons AD, Harding TC, Allen A, 2013, Discovery of a mutant-selective covalent inhibitor of EGFR that overcomes T790M-mediated resistance in NSCLC. Cancer Discov 3:1404–1415. https:// doi.org/10.1158/2159-8290.CD-13-0314 Kim ES, 2016, Olmutinib: first global approval. Drugs 76:1153– 1157. , DOI 10.1007/s40265-016-0606-z Lelais G, Epple R, Marsilje TH, Long YO, McNeill M, Chen B, Lu W, Anumolu J, Badiger S, Bursulaya B, DiDonato M, Fong R, Juarez J, Li J, Manuia M, Mason DE, Gordon P, Groessl T, Johnson K, Jia Y, Kasibhatla S, Li C, Isbell J, Spraggon G, Bender S, Michellys PY, 2016, Discovery of, R , E )- N -(7- Chloro-1-(1-[4-(dimethylamino)but-2-enoyl]azepan-3-yl)-1 H - benzo[ d ]imidazol-2-yl)-2-methylisonicotinamide, EGF816), a novel, potent, and WT sparing covalent inhibitor of oncogenic, L858R, ex19del, and resistant, T790M, EGFR mutants for the treatment of EGFR mutant non-small-cell lung cancers. J Med Chem 59:6671–6689. , DOI 10.1021/acs.jmedchem. 5b01985 Yu HA, Spira A, Horn L, Weiss J, West H, Giaccone G, Evans T, Kelly RJ, Desai B, Krivoshik A, Moran D, Poondru S, Jie F, Aoyama K, Keating A, Oxnard GR, 2017, A phase I, dose escalation study of oral ASP8273 in patients with non–small cell lung cancers with epidermal growth factor receptor mutations. Clin Cancer Res 23:7467–7473. , DOI 10.1158/1078-0432. CCR-17-1447 Xu X, 2015, Parallel phase 1 clinical trials in the US and in China: accelerating the test of avitinib in lung cancer as a novel inhibitor selectively targeting mutated EGFR and overcoming T790Minduced resistance. Chin J Cancer 34:27. , DOI 10.1186/ s40880-015-0029-3 Thress KS, Paweletz CP, Felip E, Cho BC, Stetson D, Dougherty B, Lai Z, Markovets A, Vivancos A, Kuang Y, Ercan D,Matthews SE, Cantarini M, Barrett JC, Janne PA, Oxnard GR, 2015, Acquired EGFR C797S mutation mediates resistance to AZD9291 in nonsmall cell lung cancer harboring EGFR T790M. Nat Med 21:560– 562. , DOI 10.1038/nm.3854 Chen L, FuW, Zheng L, Liu Z, Liang G, 2018, Recent Progress of small-molecule epidermal growth factor receptor, EGFR, inhibitors against C797S resistance in non-small-cell lung cancer. J Med Chem 61:4290–4300. , DOI 10.1021/acs.jmedchem. 7b01310 InukaiM, Toyooka S, Ito S, Asano H, Ichihara S, Soh J, Suehisa H, Ouchida M, Aoe K, Aoe M, Kiura K, Shimizu N, Date H, 2006, Presence of Epidermal Growth Factor Receptor gene T790M mutation as a minor clone in non–small cell lung cancer. Cancer Res 66:7854–7858. , DOI 10.1158/0008-5472.CAN-06-1951 Yu HA, Arcila ME, Hellmann MD, KrisMG, LadanyiM, Riely GJ, 2014, Poor response to erlotinib in patients with tumors containing baseline EGFR T790M mutations found by routine clinical molecular testing. Ann Oncol 25:423–428. , DOI 10.1093/ annonc/mdt573 Bell DW, Gore I, Okimoto RA, Godin-Heymann N, Sordella R, Mulloy R, Sharma SV, Brannigan BW, Mohapatra G, Settleman J, Haber DA, 2005, Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR. Nat Genet 37:1315–1316. , DOI 10.1038/ng1671 Gazdar A, Robinson L, Oliver D, Xing C, Travis WD, Soh J, Toyooka S, Watumull L, Xie Y, Kernstine K, Schiller JH, 2014, Hereditary lung cancer syndrome targets never smokers with germline EGFR gene T790M mutations. J Thorac Oncol 9:456– 463. , DOI 10.1097/JTO.0000000000000130 Oxnard GR, Heng JC, Root EJ, Rainville IR, Sable-Hunt AL, Shane-Carson KP, Carbone DP, Wiesner GL, Garber JE, 2015, Initial results of a prospective, multicenter trial to study inherited lung cancer risk associated with germline EGFRT790M: INHERIT EGFR. J Clin Oncol 33:1505. , DOI 10.1200/jco.2015.33. 15_suppl.1505 Armbruster DA, Pry T, 2008, Limit of blank, limit of detection and limit of quantitation. Clin Biochem Rev 29(Suppl 1):S49–S52 , DOI citeulike-article-id:3416410. Accessed 29 Aug 2008. Han B, Tjulandin S, Hagiwara K, Normanno N, Wulandari L, Laktionov K, Hudoyo A, He Y, Zhang YP, Wang MZ, Liu CY, Ratcliffe M, McCormack R, Reck M, 2017, EGFR mutation prevalence in Asia-Pacific and Russian patients with advanced NSCLC of adenocarcinoma and non-adenocarcinoma histology: the IGNITE study. Lung Cancer 113:37–44. , DOI 10. 1016/j.lungcan.2017.08.021 Oxnard GR, Hu Y, Mileham KF, Husain H, Costa DB, Tracy P, Feeney N, Sholl LM, Dahlberg SE, Redig AJ, Kwiatkowski DJ, Rabin MS, Paweletz CP, Thress KS, Janne PA, 2018, Assessment of resistance mechanisms and clinical implications in patients with EGFR T790M-positive lung cancer and acquired resistance to Osimertinib. JAMA Oncol 4:1527–1534. , DOI 10.1001/ jamaoncol.2018.2969 Nagano T, Tachihara M, Nishimura Y, 2018, Mechanism of resistance to epidermal growth factor receptor-tyrosine kinase inhibitors and a potential treatment strategy. Cells 7:212. , DOI 10. 3390/cells7110212 Yang Z, Yang N, Ou Q, Xiang Y, Jiang T, Wu X, Bao H, Tong X, Wang X, Shao YW, Liu Y, Wang Y, Zhou C, 2018, Investigating novel resistance mechanisms to third-generation EGFR tyrosine kinase inhibitor Osimertinib in non-small cell lung cancer patients. Clin Cancer Res 24:3097–3107. , DOI 10.1158/1078- 0432.CCR-17-2310 Ho C-C, Liao W-Y, Lin C-A, Shih JY, Yu CJ, Chih-Hsin Yang J, 2017, Acquired BRAF V600E mutation as resistant mechanism after treatment with Osimertinib. J Thorac Oncol 12:567–572. , DOI 10.1016/j.jtho.2016.11.2231 S-HI O, Cui J, Schrock AB et al, 2017, Emergence of novel and dominant acquired EGFR solvent-front mutations at Gly796, G796S/R, together with C797S/R and L792F/H mutations in one EGFR, L858R/T790M, NSCLC patient who progressed on osimertinib. Lung Cancer 108:228–231. , DOI 10.1016/j. lungcan.2017.04.003 Knebel FH, Bettoni F, Shimada AK, Cruz M, Alessi JV, Negrao MV, Reis LFL, Katz A, Camargo AA, 2017, Sequential liquid biopsies reveal dynamic alterations of EGFR driver mutations and indicate EGFR amplification as a new mechanism of resistance to osimertinib in NSCLC. Lung Cancer 108:238–241. , DOI 10.1016/j.lungcan.2017.04.004 van Noesel J, van der Ven WH, van Os TAM, Kunst PWA, Weegenaar J, Reinten RJA, Kancha RK, Duyster J, van Noesel CJM, 2013, Activating germline R776H mutation in the epidermal growth factor receptor associated with lung cancer with squamous differentiation. J Clin Oncol 31:e161–e164. , DOI 10. 1200/JCO.2012.42.1586 Ruan Z, Kannan N, 2015, Mechanistic insights into R776H mediated activation of epidermal growth factor receptor kinase. Biochemistry 54:4216–4225. , DOI 10.1021/acs.biochem. 5b00444 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1229 EP 1234 DI 10.1007/s12253-019-00683-4 PG 6 ER PT J AU Mohammadpour-Gharehbagh, A Heidari, Z Eskandari, M Aryan, A Salimi, S AF Mohammadpour-Gharehbagh, Abbas Heidari, Zahra Eskandari, Moein Aryan, Abtin Salimi, Saeedeh TI Association between Genetic Polymorphisms in microRNA Machinery Genes and Risk of Papillary Thyroid Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary thyroid carcinoma; DICER1; DROSHA; XPO5; Polymorphism ID Papillary thyroid carcinoma; DICER1; DROSHA; XPO5; Polymorphism AB Evidence suggests that the microRNAs are involved in tumorigenesis and progression of various types of malignant tumors. Therefore, the aim of current research was to examine association between genetic variants in the miRNA machinery genes and risk of papillary thyroid carcinoma(PTC) in Iranian population. Peripheral blood samples were collected from 120 PTC patients and 130 healthy subjects. Genotyping of polymorphisms in miRNA Machinery genes (DICER1 rs3742330, DROSHA rs6877842 and XPO5 rs11077) polymorphisms was performed using PCR-RFLP method. Chi square and independent sample t tests were applied for categorical and continuous variables, respectively. In this study, we found that frequency of DICER1 rs3742330G allele was significantly higher in controls compared to PTC patients. In addition, the DICER1 rs3742330 polymorphism was associated with lower risk of PTC in dominant (AG + GG vs. AA, OR = 0.5, 95%CI = 0.3–0.9, P = 0.03) model. No association was found between DROSHA rs6877842 and XPO5 rs11077 polymorphisms and PTC neither in dominant nor in recessive and allelic models. The frequency of DROSHA rs6877842GC genotype was higher in PTC patients with smaller tumor size (<1). Therefore, this polymorphism could be a protective factor for tumor development in PTC patients (OR = 0.3, 95%CI = 0.1–1, P = 0. 04). The current study indicated that DICER1 rs3742330 polymorphism was associated with lower risk of PTC. Furthermore, DROSHA rs6877842 polymorphism could be a protective factor for tumor development in PTC patients. C1 [Mohammadpour-Gharehbagh, Abbas] Zahedan University of Medical Sciences, School of Medicine, Department of Clinical BiochemistryZahedan, Iran. [Heidari, Zahra] Zahedan University of Medical Sciences, Department of EndocrinologyZahedan, Iran. [Eskandari, Moein] Zahedan University of Medical Sciences, School of Paramedical Sciences, Department of Laboratory SciencesZahedan, Iran. [Aryan, Abtin] Zahedan University of Medical Sciences, Radiology departmentZahedan, Iran. [Salimi, Saeedeh] Zahedan University of Medical Sciences, School of Medicine, Department of Clinical BiochemistryZahedan, Iran. RP Salimi, S (reprint author), Zahedan University of Medical Sciences, School of Medicine, Department of Clinical Biochemistry, Zahedan, Iran. EM sasalimi@yahoo.com;salimis@zaums.ac.ir CR Ries LAG, Young Jr JL, Keel GE, Eisner MP, Lin YD, Horner MJD, 2007, Cancer survival among adults: US SEER program, 1988–2001. Patient and tumor characteristics SEER Survival Monograph Publication:07–6215 Lim H, Devesa SS, Sosa JA, Check D, Kitahara CM, 2017, Trends in thyroid cancer incidence and mortality in the United States, 1974-2013. Jama 317(13):1338–1348 Ning L, RaoW, Yu Y, Liu X, Pan Y, Ma Y, Liu R, Zhang S, Sun H, Yu Q, 2016, Association between the KRAS gene polymorphisms and papillary thyroid carcinoma in a Chinese Han population. J Cancer 7(15):2420–2426 Lun Y,Wu X, Xia Q, Han Y, Zhang X, Liu Z,Wang F, Duan Z, Xin S, Zhang J, 2013, Hashimoto’s thyroiditis as a risk factor of papillary thyroid cancer may improve cancer prognosis. Otolaryngol Head Neck Surg 148(3):396–402 Rago T, Fiore E, Scutari M, Santini F, Di Coscio G, Romani R, Piaggi P, Ugolini C, Basolo F, Miccoli P, 2010, Male sex, single nodularity, and young age are associated with the risk of finding a papillary thyroid cancer on fine-needle aspiration cytology in a large series of patients with nodular thyroid disease. Eur J Endocrinol 162(4):763–770 Zhang Q, Song F, Zheng H, Zhu X, Song F, Yao X, Zhang L, Chen K, 2013, Association between single-nucleotide polymorphisms of BRAF and papillary thyroid carcinoma in a Chinese population. Thyroid 23(1):38–44 Ning L, Yu Y, Liu X, Ai L, Zhang X, Rao W, Shi J, Sun H, Yu Q, 2015, Association analysis of MET gene polymorphism with papillary thyroid carcinoma in a Chinese population. Int J Endocrinol 2015(2015):1–5. , DOI 10.1155/2015/405217 Wei W-J, Lu Z-W, Li D-S, Wang Y, Zhu Y-X, Wang Z-Y, Wu Y, Wang Y-L, Ji Q-H, 2014, Association of the miR-149 Rs2292832 polymorphism with papillary thyroid Cancer risk and clinicopathologic characteristics in a Chinese population. Int J Mol Sci 15(11): 20968–20981 Heidari Z, Mohammadpour-Gharehbagh A, Eskandari M, Harati- Sadegh M, Salimi S, 2018, Genetic polymorphisms of miRNA let7a-2 and pri-mir-34b/c are associated with an increased risk of papillary thyroid carcinoma and clinical/pathological features. J Cell Biochem 120:8640–8647. , DOI 10.1002/jcb.28152 Ha T-Y, 2011, MicroRNAs in human diseases: from cancer to cardiovascular disease. Immune Netw 11(3):135–154 Zhang W, Dahlberg JE, Tam W, 2007, MicroRNAs in tumorigenesis: a primer. Am J Pathol 171(3):728–738 Lin S, Gregory RI, 2015, MicroRNA biogenesis pathways in cancer. Nat Rev Cancer 15(6):321–333 Jansson MD, Lund AH, 2012, MicroRNA and cancer. Mol Oncol 6(6):590–610 Paranjape T, Slack FJ, Weidhaas JB, 2009, MicroRNAs: tools for cancer diagnostics. Gut 58(11):1546–1554. , DOI 10.1136/ gut.2009.179531 Fuziwara CS, Kimura ET, 2017, MicroRNAs in thyroid development, function and tumorigenesis. Mol Cell Endocrinol 456:44–50. , DOI 10.1016/j.mce.2016.12.017 Krol J, Loedige I, Filipowicz W, 2010, The widespread regulation ofmicroRNA biogenesis, function and decay. Nat Rev Genet 11(9): 597–610 Song X, Zhong H,Wu Q,WangM, Zhou J, Zhou Y, Lu X, Ying B, 2017, Association between SNPs in microRNA machinery genes and gastric cancer susceptibility, invasion, and metastasis in Chinese Han population. Oncotarget 8(49):86435–86446. https:// doi.org/10.18632/oncotarget.21199 Rah H, Jeon YJ, Lee BE, Kim JO, Shim SH, Lee WS, Choi DH, Kim JH, Kim NK, 2013, Association of polymorphisms in microRNA machinery genes, DROSHA, DICER1, RAN, and XPO5, with risk of idiopathic primary ovarian insufficiency in Korean women. Menopause 20(10):1067–1073 Wang F, Jiang C, Sun Q, Yan F,Wang L, Fu Z, Liu T, Hu F, 2015, miR-195 is a key regulator of Raf1 in thyroid cancer. Onco Targets Ther 8:3021–3028. , DOI 10.2147/ott.s90710 Li D, Jian W, Wei C, Song H, Gu Y, Luo Y, Fang L, 2014, Downregulation of miR-181b promotes apoptosis by targeting CYLD in thyroid papillary cancer. Int J Clin Exp Pathol 7(11):7672–7680 Deng X, Wu B, Xiao K, Kang J, Xie J, Zhang X, Fan Y, 2015, MiR-146b-5p promotes metastasis and induces epithelialmesenchymal transition in thyroid cancer by targeting ZNRF3. Cell Physiol Biochem: international journal of experimental cellular physiology, biochemistry, and pharmacology 35(1):71–82. https:// doi.org/10.1159/000369676 Xiong F,WuC,Chang J, YuD, XuB, Yuan P, Zhai K, Xu J, TanW, Lin D, 2011, Genetic variation in an miRNA-1827 binding site in MYCL1 alters susceptibility to small-cell lung cancer. Cancer Res 71(15):5175–5181. , DOI 10.1158/0008-5472.can-10-4407 Robertson JC, Jorcyk CL, Oxford JT, 2018, DICER1 syndrome: DICER1 mutations in rare cancers. Cancers 10(5). , DOI 10.3390/cancers10050143 KarubeY, TanakaH, Osada H, Tomida S, TatematsuY, Yanagisawa K, Yatabe Y, Takamizawa J, Miyoshi S, Mitsudomi T, 2005, Reduced expression ofDicer associatedwith poor prognosis in lung cancer patients. Cancer Sci 96(2):111–115 Merritt WM, Lin YG, Han LY, Kamat AA, Spannuth WA, Schmandt R, Urbauer D, Pennacchio LA, Cheng J-F, Nick AM, 2008, Dicer, Drosha, and outcomes in patients with ovarian cancer. N Engl J Med 359(25):2641–2650 Erler P, Keutgen XM, Crowley MJ, Zetoune T, Kundel A, Kleiman D, Beninato T, Scognamiglio T, Elemento O, Zarnegar R, 2014, Dicer expression and microRNA dysregulation associate with aggressive features in thyroid cancer. Surgery 156(6):1342–1350 Kim MN, Kim JO, Lee SM, Park H, Lee JH, Rim KS, Hwang SG, Kim NK, 2016, Variation in the Dicer and RAN genes are associated with survival in patients with hepatocellular carcinoma. PLoS One 11(9):e0162279. , DOI 10.1371/journal.pone. 0162279 Osuch-Wojcikiewicz E, Bruzgielewicz A, Niemczyk K, Sieniawska-Buccella O, Nowak A, Walczak A, Majsterek I, 2015, Association of polymorphic variants of miRNA processing genes with larynx cancer risk in a polish population. Biomed Res Int 2015(2015):1–17. , DOI 10.1155/2015/298378 Zeng Y, Cullen BR, 2004, Structural requirements for premicroRNA binding and nuclear export by Exportin 5. Nucleic Acids Res 32(16):4776–4785. , DOI 10.1093/nar/gkh824 Leaderer D, Hoffman AE, Zheng T, Fu A,Weidhaas J, Paranjape T, Zhu Y, 2011, Genetic and epigenetic association studies suggest a role of microRNA biogenesis gene exportin-5, XPO5, in breast tumorigenesis. Int J Mol Epidemiol Genet 2(1):9–18 Ye Y, Wang KK, Gu J, Yang H, Lin J, Ajani JA, Wu X, 2008, Genetic variations in microRNA-related genes are novel susceptibility loci for esophageal cancer risk. Cancer Prev Res 1(6):460– 469 Wen J, Gao Q,Wang N, ZhangW, Cao K, Zhang Q, Chen S, Shi L, 2017, Association of microRNA-related gene XPO5 rs11077 polymorphism with susceptibility to thyroid cancer. Medicine 96(14):e6351. , DOI 10.1097/md.0000000000006351 Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Radmark O, Kim S, 2003, The nuclear RNase III Drosha initiates microRNA processing. Nature 425(6956):415–419 Yuan L, Chu H,Wang M,Gu X, ShiD, Ma L, Zhong D, Du M, Li P, Tong N, Fu G, Qin C, Yin C, Zhang Z, 2013, Genetic variation in DROSHA 3'UTR regulated by hsa-miR-27b is associated with bladder cancer risk. PLoS One 8(11):e81524. , DOI 10. 1371/journal.pone.0081524 Sung H, Jeon S, Lee KM, Han S, Song M, Choi JY, Park SK, Yoo KY, Noh DY, Ahn SH, Kang D, 2012, Common genetic polymorphisms of microRNA biogenesis pathway genes and breast cancer survival. BMC Cancer 12:195. , DOI 10.1186/1471-2407- 12-195 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1235 EP 1241 DI 10.1007/s12253-019-00688-z PG 7 ER PT J AU Krafft, U Tschirdewahn, S Hess, J Harke, N Hadaschik, AB Nyirady, P Szendroi, A Szucs, M Modos, O Olah, Cs Szekely, E Reis, H Szarvas, T AF Krafft, Ulrich Tschirdewahn, Stephan Hess, Jochen Harke, N. Nina Hadaschik, A Boris Nyirady, Peter Szendroi, Attila Szucs, Maria Modos, Orsolya Olah, Csilla Szekely, Eszter Reis, Henning Szarvas, Tibor TI STIP1 Tissue Expression Is Associated with Survival in Chemotherapy-Treated Bladder Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; STIP1; Cisplatin; Resistance ID Bladder cancer; STIP1; Cisplatin; Resistance AB To optimize treatment decisions in advanced bladder cancer (BC), we aimed to assess the therapy predictive value of STIP1 with regard to cisplatin therapy. Cisplatin-based chemotherapy represents the standard first-line systemic treatment of advanced bladder cancer. Since novel immuno-oncologic agents are already available for cisplatin-resistant or ineligible patients, biological markers are needed for the prediction of cisplatin resistance. STIP1 expression was analyzed in paraffin-embedded bladder cancer tissue samples of 98 patients who underwent adjuvant or salvage cisplatin-based chemotherapy by using immunohistochemistry. Furthermore, prechemotherapy serum STIP1 concentrations were determined in 48 BC patients by ELISA. Results were correlated with the clinicopathological and follow-up data. Stronger STIP1 nuclear staining was associated with worse OS in both the whole patient group (p = 0.034) and the subgroup of patients who received at least 2 cycles of chemotherapy (p = 0.043). These correlations remained significant also in the multivariable analyses (p =0.035 and p = 0.040). Stronger STIP1 cytoplasmatic immunostaining correlated with shorter PFS both in the whole cohort (p = 0.045) and in the subgroup of patients who received at least 2 cycles of chemotherapy (p = 0.026). Elevated STIP1 serum levels were associated with older patient’s age, but we found no correlation between STIP1 serum levels and patients’ outcome. Our results suggest that tissue STIP1 analysis might be used for the prediction of cisplatin-resistance in BC. In contrast, pretreatment STIP1 serum levels showed no predictive value for chemotherapy response and survival. C1 [Krafft, Ulrich] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Tschirdewahn, Stephan] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Hess, Jochen] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Harke, N. Nina] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Hadaschik, A Boris] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. [Nyirady, Peter] Semmelweis University, Department of UrologyBudapest, Hungary. [Szendroi, Attila] Semmelweis University, Department of UrologyBudapest, Hungary. [Szucs, Maria] Semmelweis University, Department of UrologyBudapest, Hungary. [Modos, Orsolya] Semmelweis University, Department of UrologyBudapest, Hungary. [Olah, Csilla] Semmelweis University, Department of UrologyBudapest, Hungary. [Szekely, Eszter] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Reis, Henning] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Szarvas, Tibor] University of Duisburg-Essen, Department of Urology, Hufelandstr. 55, 45147 Essen, Germany. RP Szarvas, T (reprint author), University of Duisburg-Essen, Department of Urology, 45147 Essen, Germany. EM sztibusz@gmail.com CR Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H et al, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur JCancer 49:1374–1403 Antoni S, Ferlay J, Soerjomataram I, Znaor A, Jemal A, Bray F, 2017, Bladder cancer incidence and mortality: a global overview and recent trends. Eur Urol 71:96–108 Stein JP, Skinner DG, 2006, Radical cystectomy for invasive bladder cancer: long-term results of a standard procedure.World J Urol 24:296–304 Stadler WM, Lerner SP, 2003, Perioperative chemotherapy in locally advanced bladder cancer. Lancet. 361:1922–1923 Leow JJ, Martin-DoyleW, Rajagopal PS, Patel CG, Anderson EM, Rothman ATet al, 2014, Adjuvant chemotherapy for invasive bladder cancer: a 2013 updated systematic review and meta-analysis of randomized trials. Eur Urol 66:42–54 Rouanne M, Roumiguie M, Houede N, Masson-Lecomte A, Colin P, Pignot G et al, 2018, Development of immunotherapy in bladder cancer: present and future on targeting PD(L)1 and CTLA-4 pathways. World J Urol 36(11):1727–1740 Seiler R, Oo HZ, Tortora D, Clausen TM,Wang CK, Kumar G et al, 2017, An Oncofetal glycosaminoglycan modification provides therapeutic access to cisplatin-resistant bladder Cancer. Eur Urol 72:142–150 Ozcan MF, Dizdar O, Dincer N, Balci S, Guler G, Gok B et al, 2013, Low ERCC1 expression is associated with prolonged survival in patients with bladder cancer receiving platinum-based neoadjuvant chemotherapy. Urol Oncol 31:1709–1715 Hemdan T, Malmstrom P-UU, Jahnson S, Segersten U, 2015, Emmprin expression predicts response and survival following cisplatin containing chemotherapy for bladder Cancer: a validation study. J Urol 194:1575–1581 Als AB, Dyrskjot L, von der Maase H, Koed K, Mansilla F, Toldbod HE et al, 2007, Emmprin and survivin predict response and survival following cisplatin-containing chemotherapy in patients with advanced bladder cancer. Clin Cancer Res 13:4407–4414 van Rhijn BW, Catto JW, Goebell PJ, Knuchel R, Shariat SF, van der Poel HG et al, 2014, Molecular markers for urothelial bladder cancer prognosis: toward implementation in clinical practice. Urol Oncol 32:1078–1087 Krafft U, Tschirdewahn S,Hess J,HarkeNN, Hadaschik B,Olah C, Krege S, Nyirady P, Szendroi A, Szucs M, Modos O, Szekely E, Reis H, Szarvas T, 2019, Validation of survivin and HMGA2 as biomarkers for cisplatin resistance in bladder cancer. Urol Oncol. , DOI 10.1016/j.urolonc.2019.04.015 Zhang S, Shao J, Su F, 2018, Prognostic significance of STIP1 expression in human cancer: a meta-analysis. Clin Chim Acta 486:168–176 Krege S, Rexer H, vom Dorp F, de Geeter P, Klotz T, Retz M et al, 2014, Prospective randomized double-blind multicentre phase II study comparing gemcitabine and cisplatin plus sorafenib chemotherapy with gemcitabine and cisplatin plus placebo in locally advanced and/or metastasized urothelial cancer: SUSE, AUO-AB 31/ 05). BJU Int 113:429–436 Nicolet CM, Craig EA, 1989, Isolation and characterization of STI1, a stress-inducible gene from Saccharomyces cerevisiae. Mol Cell Biol 9:3638–3646 Longshaw VM, Chapple JP, Balda MS, Cheetham ME, Blatch GL, 2004, Nuclear translocation of the Hsp70/Hsp90 organizing protein mSTI1 is regulated by cell cycle kinases. J Cell Sci 117:701–710 Odunuga OO, Longshaw VM, Blatch GL, 2004, Hop: more than an Hsp70/Hsp90 adaptor protein. Bioessays. 26:1058–1068 Johnson BD, Schumacher RJ, Ross ED, Toft DO, 1998, Hop modulates Hsp70/Hsp90 interactions in protein folding. J Biol Chem 273:3679–3686 Zanata SM, Lopes MH, Mercadante AF et al, 2002, Stressinducible protein 1 is a cell surface ligand for cellular prion that triggers neuroprotection. EMBO J 21(13):3307–3316 Wang TH, Chao A, Tsai CL, Chang CL et al, 2010, Stressinduced phosphoprotein 1 as a secreted biomarker for human ovarian cancer promotes cancer cell proliferation. Mol Cell Proteomics 9(9):1873–1884 Roffe M, Beraldo FH, Bester R, 2010, Prion protein interaction with stress-inducible protein 1 enhances neuronal protein synthesis via mTOR. Proc Natl Acad Sci U S A 107(29):13147–13152 Chen Z, Xu L, Su T, Ke Z, Peng Z, Zhang N et al, 2017, Autocrine STIP1 signaling promotes tumor growth and is associated with disease outcome in hepatocellular carcinoma. Biochem Biophys Res Commun 493:365–372 Su T, Liao J, Dai Z, Xu L, Chen S, Wang Y et al, 2018, Stressinduced phosphoprotein 1 mediates hepatocellular carcinoma metastasis after insufficient radiofrequency ablation. Oncogene. 37: 3514–3527 Skalnikova H, Martinkova J, Hrabakova R, Halada P, Dziechciarkova M, Hajduch M et al, 2011, Cancer drugresistance and a look at specific proteins: Rho GDP-dissociation inhibitor 2, Y-box binding protein 1, and HSP70/90 organizing protein in proteomics clinical application. J Proteome Res 10: 404–415 Kim S, Cho H, Nam EJ, Kim SW, Kim YT, Park YWet al, 2010, Autoantibodies against stress-induced phosphoprotein-1 as a novel biomarker candidate for ovarian cancer. Genes Chromosom Cancer 49:585–595 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1243 EP 1249 DI 10.1007/s12253-019-00689-y PG 7 ER PT J AU Rakhshan, A Esmaeili, HM Kahaei, SM Taheri, M Omrani, DM Noroozi, R Ghafouri-Fard, S AF Rakhshan, Azadeh Esmaeili, Hossein Mohammad Kahaei, Salar Mir Taheri, Mohammad Omrani, Davood Mir Noroozi, Rezvan Ghafouri-Fard, Soudeh TI A Single Nucleotide Polymorphism in GAS5 lncRNA is Associated with Risk of Bladder Cancer in Iranian Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Bladder cancer; GAS5; lncRNA ID Bladder cancer; GAS5; lncRNA AB Down-regulation of the long non-coding RNA (lncRNA) growth arrest-specific 5 (GAS5) has a pathogenic role in bladder cancer. Moreover, genomic variants of this lncRNA have been associated with risk of diverse cancers. In the present project, we genotyped two putative functional SNPs (rs2067079 and rs6790) in 122 bladder cancer patients and 150 age- and sex matched healthy subjects. The rs2067079 was associated risk of bladder cancer in recessive inheritance model (TT vs.CC + CT: OR (95% Confidence interval (CI)) = 2.67 (1.27–5.62), adjusted P value = 0.02). The T G haplotype (rs2067079 and rs6790) increased the risk of bladder cancer in the assessed population (OR (95% CI) = 1.73 (1.18–2.56), adjusted P value = 0.02). Consequently, in the current project we introduced a novel risk locus for bladder cancer in Iranian population. C1 [Rakhshan, Azadeh] Shahid Beheshti University of Medical Sciences, Urogenital Stem Cell Research CenterTehran, Iran. [Esmaeili, Hossein Mohammad] High Institute for Research and Education in Transfusion Medicine, Blood Transfusion Research CenterTehran, Iran. [Kahaei, Salar Mir] Shahid Beheshti University of Medical Sciences, Department of Medical GeneticsTehran, Iran. [Taheri, Mohammad] Shahid Beheshti University of Medical Sciences, Department of Medical GeneticsTehran, Iran. [Omrani, Davood Mir] Shahid Beheshti University of Medical Sciences, Urology and Nephrology Research CenterTehran, Iran. [Noroozi, Rezvan] Shahid Beheshti University of Medical Sciences, Phytochemistry Research CenterTehran, Iran. [Ghafouri-Fard, Soudeh] High Institute for Research and Education in Transfusion Medicine, Blood Transfusion Research CenterTehran, Iran. RP Taheri, M (reprint author), Shahid Beheshti University of Medical Sciences, Department of Medical Genetics, Tehran, Iran. EM mohammad_823@yahoo.com CR Rafiemanesh H, Lotfi Z, Bakhtazad S, Ghoncheh M, Salehiniya H, 2018, The epidemiological and histological trend of bladder cancer in Iran. J Cancer Res Ther 14(3):532–536 Gu J, Wu XF, 2011, Genetic susceptibility to bladder cancer risk and outcome. Pers Med 8(3):365–374 Taheri M, Omrani MD, Ghafouri-Fard S, 2018, Long non-coding RNA expression in bladder cancer. Biophys Rev 10(4):1205–1213 Minotti L, Agnoletto C, Baldassari F, Corra F, Volinia S, 2018, SNPs and somatic mutation on long non-coding RNA: new frontier in the cancer studies? High-throughput. 7(4) Zhang H, Guo Y, Song Y, Shang C, 2017, Long noncoding RNA GAS5 inhibits malignant proliferation and chemotherapy resistance to doxorubicin in bladder transitional cell carcinoma. Cancer Chemother Pharmacol 79(1):49–55 Mourtada-Maarabouni M, Pickard MR, Hedge VL, Farzaneh F, Williams GT, 2009, GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene. 28(2): 195–208 Mahdi Eftekharian M, Noroozi R, Komaki A, Mazdeh M, Taheri M, Ghafouri-Fard S, 2019, GAS5 genomic variants and risk of multiple sclerosis. Neurosci Lett 701:54–57 Smith CM, Steitz JA, 1998, Classification of gas5 as a multi-smallnucleolar- RNA, snoRNA, host gene and a member of the 5′-terminal oligopyrimidine gene family reveals common features of snoRNA host genes. Mol Cell Biol 18(12):6897–6909 Ghaforui-Fard S, Taheri M, 2019, Growth arrest specific transcript 5 in tumorigenesis process:An update on the expression pattern and genomic variants. Biomed Pharmacother 112 Li QJ,Ma G, Sun SH, Xu Y,Wang BJ, 2018, Polymorphism in the promoter region of lncRNA GAS5 is functionally associated with the risk of gastric cancer. Clin Res Hepatol Gas 42(5):478–482 Li WH, Huang K, Wen FB, Cui GH, Guo HZ, Zhao S, 2017, Genetic variation of lncRNA GAS5 contributes to the development of lung cancer. Oncotarget. 8(53):91025–91029 Zhu ZS, Xue YZ, Fu W, Li CY, Feng LJ, Xing YH et al, 2016, Functional indel polymorphism within lncRNA GAS5 and colorectal carcinoma risk. Int J Clin Exp Pathol 9(11):11767–11773 Guo Z,Wang YH, Zhao Y, Jin Y, An L,Wu B et al, 2017, Genetic polymorphisms of long non-coding RNA GAS5 predict platinumbased concurrent chemoradiotherapy response in nasopharyngeal carcinoma patients. Oncotarget. 8(37):62286–62297 Wang M, Guo C, Wang L, Luo G, Huang C, Li YW et al, 2018, Long noncoding RNA GAS5 promotes bladder cancer cells apoptosis through inhibiting EZH2 transcription. Cell Death Dis 9 Yoo J, Lee Y, Kim Y, Rha SY, Kim Y, 2008, SNPAnalyzer 2.0: a web-based integrated workbench for linkage disequilibrium analysis and association analysis. BMC Bioinf 9:290 Liu ZH,WangW, Jiang JT, Bao ED, Xu DL, Zeng YG et al, 2013, Downregulation of GAS5 promotes bladder cancer cell proliferation, partly by regulating CDK6. PLoS One 8(9) Ren C, An GL, Zhao CH, Ouyang ZY, Bo XC, Shu WJ, 2018, Lnc2Catlas: an atlas of long noncoding RNAs associated with risk of cancers. Sci Rep-Uk 8 Ward LD, Kellis M, 2012, HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res 40(D1):D930–D9D4 Zhang Q, Stovall DB, Inoue K, Sui G, 2011, The oncogenic role of Yin Yang 1. Crit Rev Oncog 16(3–4):163–197 Zaravinos A, Lambrou GI, Boulalas I, Delakas D, Spandidos DA, 2011, Identification of common differentially expressed genes in urinary bladder cancer. PLoS One 6(4):e18135 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1251 EP 1254 DI 10.1007/s12253-019-00693-2 PG 4 ER PT J AU Jin, G Fan, XM Li, KX Niu, ShH Zhang, QY AF Jin, Ge Fan, Xiao-Mei Li, Kui-Xiu Niu, Shu-Huai Zhang, Qian-Ying TI The Association between Epidermal Growth Factor Receptor Single Nucleotide Polymorphisms and Radiochemotherapy Response in Cervical Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; Epidermal growth factor receptor; Single nucleotide polymorphism; Radio-chemotherapy ID Cervical cancer; Epidermal growth factor receptor; Single nucleotide polymorphism; Radio-chemotherapy AB Emerging data reveal that epidermal growth factor receptor (EGFR) single nucleotide polymorphisms (SNPs) can act as efficacy indicators for tumor treatment. Here, the association between EGFR R497K (rs11543848) and -216G/T (rs712829) SNPs and radio-chemotherapy response in cervical cancer was investigated. EGFR R497K and -216G/T genotypes were analyzed by polymerase chain reaction-ligation detection reaction in 196 cervical cancer patients receiving radiotherapy alone, or in combination with chemotherapy. Compared with the 497G/G genotype, the A/A genotype significantly increased sensitivity to radio-chemotherapy treatment (adjusted OR = 0.244, 95% CI = 0.087–0.680). Sensitivity to radio-chemotherapy was not significantly different in carriers of the ‘T’ allele than that measured for the -216G/G genotype (adjusted OR = 2.412, 95%CI = 0.856– 6.979). Additionally, the 497A/A genotype conferred a reduced risk of recurrence or metastasis than did the G/G genotype (adjusted OR = 0.248, 95% CI = 0.078–0.786, P< 0.05). Moreover, carriers of the ‘T’ allele did not have significantly modified risk of recurrence or metastasis compared with those with the -216G/G genotype (adjusted OR = 1.027, 95% CI = 0.324–3.253). Multivariate analysis revealed an association between clinical stage and treatment response (adjusted OR = 3.575, 95% CI = 1.662–7.692) and between age and the risk of recurrence or metastasis (adjusted OR = 0.319, 95%CI = 0.148–0.691). Our results show that, in patients with cervical cancer, the R497K polymorphism is correlated with treatment response and the risk of recurrence or metastasis. The R497K SNP might be a genetic marker for prediction of radio-chemotherapy response and the risk of recurrence and/or metastasis in patients with cervical cancer. C1 [Jin, Ge] the Fourth Hospital of Hebei Medical University, Department of Gynecologic Oncology, #12, Jiankang Road, 050011 Shijiazhuang, Hebei province, China. [Fan, Xiao-Mei] the Fourth Hospital of Hebei Medical University, Department of Gynecologic Oncology, #12, Jiankang Road, 050011 Shijiazhuang, Hebei province, China. [Li, Kui-Xiu] the Fourth Hospital of Hebei Medical University, Department of Gynecologic Oncology, #12, Jiankang Road, 050011 Shijiazhuang, Hebei province, China. [Niu, Shu-Huai] the Fourth Hospital of Hebei Medical University, Department of Gynecologic Oncology, #12, Jiankang Road, 050011 Shijiazhuang, Hebei province, China. [Zhang, Qian-Ying] the Fourth Hospital of Hebei Medical University, Department of Gynecologic Oncology, #12, Jiankang Road, 050011 Shijiazhuang, Hebei province, China. RP Fan, XM (reprint author), the Fourth Hospital of Hebei Medical University, Department of Gynecologic Oncology, 050011 Shijiazhuang, China. EM fanxiaomei2006@sina.com CR Lee KB, Shim SH, Lee JM, 2018, Comparison between adjuvant chemotherapy and adjuvant radiotherapy/chemoradiotherapy after radical surgery in patients with cervical cancer: a meta-analysis. J Gynecol Oncol 29 Gupta S, Maheshwari A, Parab P, Mahantshetty U, Hawaldar R, Sastri Chopra S, Kerkar R, Engineer R, Tongaonkar H, Ghosh J, Gulia S, Kumar N, Shylasree TS, Gawade R, Kembhavi Y, Gaikar M, Menon S, Thakur M, Shrivastava S, Neoadjuvant BR, 2018, Chemotherapy followed by radical surgery versus concomitant chemotherapy and radiotherapy in patients with stage IB2, IIA, or IIB squamous cervical Cancer: a randomized controlled trial. J Clin Oncol:JCO2017759985 Benedetti-Panici P, Greggi S, Colombo A, Amoroso M, Smaniotto D, Giannarelli D, Amunni G, Raspagliesi F, Zola P, Mangioni C, Landoni F, 2002, Neoadjuvant chemotherapy and radical surgery versus exclusive radiotherapy in locally advanced squamous cell cervical cancer: results from the Italian multicenter randomized study. J Clin Oncol 20:179–188 Cerciello F, Riesterer O, Sherweif M, Odermatt B, Ciernik IF, 2007, Is EGFR a moving target during radiotherapy of carcinoma of the uterine cervix? Gynecol Oncol 106:394–399 Fukazawa EM, Baiocchi G, Soares FA, Kumagai LY, Faloppa CC, Badiglian-Filho L, Coelho FR, Goncalves WJ, Costa RL, Goes JC, 2014, Cox-2, EGFR, and ERBB-2 expression in cervical intraepithelial neoplasia and cervical cancer using an automated imaging system. Int J Gynecol Pathol 33:225–234 Hernowo BS, Suryanti S, Wibisono F, 2016, Correlation between EGFR expression and Radiosensitivity in cervical adenocarcinoma cases. Asian Pac J Cancer Prev 17:2535–2537 Hu S, FuW, Li T, Yuan Q,Wang F, Lv G, Lv Y, Fan X, Shen Y, Lin F, Tang Y, Ye X, Yang Y, Lei C, 2017, Antagonism of EGFR and notch limits resistance to EGFR inhibitors and radiation by decreasing tumor-initiating cell frequency. Sci Transl Med 9:eaag0339 Wang M, Han J, Marcar L, Black J, Liu Q, Li X, Nagulapalli K, Sequist LV, Mak RH, Benes CH, Hong TS, Gurtner K, Krause M, Baumann M, Kang JX, Whetstine JR, Willers H, 2017, Radiation resistance in KRAS-mutated lung Cancer is enabled by stem-like propertiesmediated by an Osteopontin-EGFR pathway. Cancer Res 77:2018–2028 Huang S, Li C, Armstrong EA, Peet CR, Saker J, Amler LC, Sliwkowski MX, Harari PM, 2013, Dual targeting of EGFR and HER3 withMEHD7945A overcomes acquired resistance to EGFR inhibitors and radiation. Cancer Res 73:824–833 Jin T, Yu CX, Jin T, Yu CX, Lei DP, LiuDY, Xu FL, LuYT, Pan XL, 2009, Identification of epidermal growth factor receptor, EGFR, exon 20 single nucleotide polymorphism in Chinese squamous cell carcinoma of head and neck, SCCHN). Acta Otolaryngol 129: 1306–1312 Feng X, Qin JJ, Zheng BS, Huang LL, Xie XY, Zhou HF, 2014, Association of epidermal growth factor receptor, EGFR, gene polymorphism with lung cancer risk: a systematic review. J Recept Signal Transduct Res 34:333–334 Moriai T, Kobrin MS, Korc M, 1993, Cloning of a variant epidermal growth factor receptor. Biochem Biophys Res Commun 191: 1034–1039 Moriai T, Kobrin MS, Hope C, Speck L, Korc M, 1994, Avariant epidermal growth factor receptor exhibits altered type alpha transforming growth factor binding and transmembrane signaling. Proc Natl Acad Sci U S A 91:10217–10221 Datta NR, Stutz E, Liu M, Rogers S, Klingbiel D, Siebenhuner A, Singh S, Bodis S, 2017, Concurrent chemoradiotherapy vs. radiotherapy alone in locally advanced cervix cancer: a systematic review and meta-analysis. Gynecol Oncol 145:374–385 Bandres E, Barricarte R, Cantero C, Honorato B, Malumbres R, Zarate R, Alcalde J, Garcia-Foncillas J, 2007, Epidermal growth factor receptor, EGFR, polymorphisms and survival in head and neck cancer patients. Oral Oncol 43:713–719 Stoehlmacher-Williams J, Obermann L, Ehninger G, Goekkurt E, 2012, Polymorphisms of the epidermal growth factor receptor, EGFR, and survival in patients with advanced cancer of the head and neck, HNSCC). Anticancer Res 32:421–425 Press OA, Zhang W, Gordon MA, Yang D, Lurje G, Iqbal S, El- Khoueiry A, Lenz HJ, 2008, Gender-related survival differences associated with EGFR polymorphisms in metastatic colon cancer. Cancer Res 68:3037–3042 Zhang W, Park DJ, Lu B, Yang DY, Gordon M, Groshen S, Yun J, Press OA, Vallbohmer D, Rhodes K, Lenz HJ, 2005, Epidermal growth factor receptor gene polymorphisms predict pelvic recurrence in patients with rectal cancer treated with chemoradiation. Clin Cancer Res 11:600–605 Ichihara S, Toyooka S, Fujiwara Y, Hotta K, Shigematsu H, Tokumo M, Soh J, Asano H, Ichimura K, Aoe K, Aoe M, Kiura K, Shimizu K, Date H, Shimizu N, 2007, The impact of epidermal growth factor receptor gene status on gefitinibtreated Japanese patients with non-small-cell lung cancer. Int J Cancer 120:1239–1247 Nomura M, Shigematsu H, Li L, Suzuki M, Takahashi T, Estess P, Siegelman M, Feng Z, Kato H, Marchetti A, Shay JW, Spitz MR, Wistuba II, Minna JD, Gazdar AF, 2007, Polymorphisms, mutations, and amplification of the EGFR gene in non-small cell lung cancers. PLoS Med 4:e125 Liu G, Gurubhagavatula S, ZhouW,Wang Z,Yeap BY, Asomaning K, Su L, Heist R, Lynch TJ, Christiani DC, 2008, Epidermal growth factor receptor polymorphisms and clinical outcomes in non-small-cell lung cancer patients treated with gefitinib. Pharmacogenomics J 8:129–138 Costa BM, Viana-PereiraM, Fernandes R, Costa S, Linhares P, Vaz R, Pinheiro C, Lima J, Soares P, Silva A, Pardal F, Amorim J, Nabico R, Almeida R, Alegria C, Pires MM, Pinheiro C, Carvalho E, Oliveira P, Lopes JM, Reis RM, 2011, Impact of EGFR genetic variants on glioma risk and patient outcome. Cancer Epidemiol Biomark Prev 20:2610–2617 Demirci S, Ozsaran Z, Ozsaran A, Yavas F, Demircioglu B, Hanhan M, Dikmen Y, Aras AB, 2012, Evaluation of treatment results and prognostic factors in early-stage cervical carcinoma patients treated with postoperative radiotherapy or radiochemotherapy. Eur J Gynaecol Oncol 33:62–67 Lee WM, Park SI, Kim BJ, Kim MH, Choi SC, Lee ED, Ryu SY, 2012, Clinicopathologic factors for central recurrence in patients with locally advanced bulky cervical cancer. Eur J Obstet Gynecol Reprod Biol 161:219–223 Huang YT,Wang CC, Tsai CS, Lai CH, Chang TC, Chou HH, Lee SP, Hong JH, 2012, Clinical behaviors and outcomes for adenocarcinoma or adenosquamous carcinoma of cervix treated by radical hysterectomy and adjuvant radiotherapy or chemoradiotherapy. Int J Radiat Oncol Biol Phys 84:420–427 Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, Jia M, Shepherd R, Leung K, Menzies A, Teague JW, Campbell PJ, Stratton MR, Futreal PA, 2011, COSMIC: mining complete cancer genomes in the catalogue of somatic mutations in Cancer. Nucleic Acids Res 39:D945–D950 Hodoglugil U, CarrilloMW, Hebert JM, Karachaliou N, Rosell RC, Altman RB, Klein TE, 2013, PharmGKB summary: very important pharmacogene information for the epidermal growth factor receptor. Pharmacogenet Genomics 23:636–642 Liu W, He L, Ramirez J, Krishnaswamy S, Kanteti R, Wang YC, Salgia R, Ratain MJ, 2011, Functional EGFR germline polymorphisms may confer risk for EGFR somatic mutations in non-small cell lung cancer, with a predominant effect on exon 19 microdeletions. Cancer Res 71:2423–2427 Sasaki H, Okuda K, Shimizu S, Takada M, Kawahara M, Kitahara N, Okumura M, Matsumura A, Iuchi K, Kawaguchi T, Kubo A, Kawano O, Yukiue H, Yano M, Fujii Y, 2009, EGFR R497K polymorphism is a favorable prognostic factor for advanced lung cancer. J Cancer Res Clin Oncol 135:313–318 BrennanCW,Verhaak RG,McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, Beroukhim R, Bernard B, Wu CJ, Genovese G, Shmulevich I, Barnholtz-Sloan J, Zou L, Vegesna R, Shukla SA, Ciriello G, Yung WK, Zhang W, Sougnez C, Mikkelsen T, Aldape K, Bigner DD, Van Meir EG, Prados M, Sloan A, Black KL, Eschbacher J, Finocchiaro G, Friedman W, Andrews DW, Guha A, Iacocca M, O'Neill BP, Foltz G, Myers J, Weisenberger DJ, Penny R, Kucherlapati R, Perou CM, Hayes DN, Gibbs R, Marra M, Mills GB, Lander E, Spellman P, Wilson R, Sander C, Weinstein J, Meyerson M, Gabriel S, Laird PW, Haussler D, Getz G, Chin L, Network TR, 2013, The somatic genomic landscape of glioblastoma. Cell 155:462–477 Binder ZA, Thorne AH, Bakas S,Wileyto EP, Bilello M, Akbari H, Rathore S, Ha SM, Zhang L, Ferguson CJ, Dahiya S, Bi WL, Reardon DA, Idbaih A, Felsberg J, Hentschel B, Weller M, Bagley SJ, Morrissette JJD, Nasrallah MP, Ma J, Zanca C, Scott AM, Orellana L, Davatzikos C, Furnari FB, O'Rourke DM, 2018, Epidermal growth factor receptor extracellular domain mutations in glioblastoma present opportunities for clinical imaging and therapeutic development. Cancer Cell 34:163–177 e167 Arena S, Bellosillo B, Siravegna G,Martinez A, Canadas I, Lazzari L, Ferruz N, Russo M, Misale S, Gonzalez I, IglesiasM, Gavilan E, Corti G, Hobor S, Crisafulli G, Salido M, Sanchez J, Dalmases A, Bellmunt J,De Fabritiis G, Rovira A, Di Nicolantonio F, Albanell J, Bardelli A, Montagut C, 2015, Emergence of multiple EGFR extracellular mutations during Cetuximab treatment in colorectal Cancer. Clin Cancer Res 21:2157–2166 Goncalves A, Esteyries S, Taylor-Smedra B, Lagarde A, Ayadi M, Monges G, Bertucci F, Esterni B, Delpero JR, Turrini O, Lelong B, Viens P, Borg JP, Birnbaum D, Olschwang S, Viret F, 2008, A polymorphism of EGFR extracellular domain is associated with progression free-survival in metastatic colorectal cancer patients receiving cetuximab-based treatment. BMC Cancer 8:169 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1255 EP 1261 DI 10.1007/s12253-019-00690-5 PG 7 ER PT J AU Okamoto, A Sakakura, K Takahashi, H Motegi, Si Kaira, K Yokobori-Kuwabara, Y Ishikawa, O Chikamatsu, K AF Okamoto, Ayako Sakakura, Koichi Takahashi, Hideyuki Motegi, Sei-ichiro Kaira, Kyoichi Yokobori-Kuwabara, Yuki Ishikawa, Osamu Chikamatsu, Kazuaki TI Immunological and Clinicopathological Significance of MFG-E8 Expression in Patients with Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Milk fat globule-epidermal growth factor 8 (MFG-E8); Oral squamous cell carcinoma (OSCC); Tumor microenvironment (TME); Immune suppression ID Milk fat globule-epidermal growth factor 8 (MFG-E8); Oral squamous cell carcinoma (OSCC); Tumor microenvironment (TME); Immune suppression AB Milk fat globule-epidermal growth factor 8 (MFG-E8) is a glycoprotein secreted by the activated macrophages and acts as a bridge between apoptotic cells and phagocytes. Aside from macrophages, a variety of malignant cells also express MFG-E8. The objective of this study is to elucidate the clinical relevance and significance of MFG-E8 in the tumor microenvironment (TME) of patients with oral squamous cell carcinoma (OSCC). We investigated MFG-E8 expression in 74 patients with OSCC by immunohistochemistry and evaluated the relationship between MFG-E8 expression and various clinicopathological factors including immune cell infiltration. MFG-E8 expression was detected in 34 of 74 (45.9%) patients with OSCC and a significant correlation was observed with levels of infiltrating T cells, macrophages, and immunosuppressive M2 macrophages. Furthermore, MFG-E8 expression was also associated with clinical stage, lymphatic/vascular invasion, and Ki-67+ tumor cells but not with survival. Our results suggest that MFG-E8 may play an important role in shaping the immune suppressive network in TME as well as tumor progression. C1 [Okamoto, Ayako] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck Surgery, 3718511 Gunma, Japan. [Sakakura, Koichi] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck Surgery, 3718511 Gunma, Japan. [Takahashi, Hideyuki] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck Surgery, 3718511 Gunma, Japan. [Motegi, Sei-ichiro] Gunma University, Graduate School of Medicine, Department of DermatologyMaebashi, Japan. [Kaira, Kyoichi] Gunma University, Graduate School of Medicine, Department of Oncology Clinical DevelopmentMaebashi, Japan. [Yokobori-Kuwabara, Yuki] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck Surgery, 3718511 Gunma, Japan. [Ishikawa, Osamu] Gunma University, Graduate School of Medicine, Department of DermatologyMaebashi, Japan. [Chikamatsu, Kazuaki] Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck Surgery, 3718511 Gunma, Japan. RP Chikamatsu, K (reprint author), Gunma University, Graduate School of Medicine, Department of Otolaryngology-Head and Neck Surgery, 3718511 Gunma, Japan. EM tikamatu@gunma-u.ac.jp CR Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, Vonderheide RH, Pittet MJ, Jain RK, Zou W, Howcroft TK, Woodhouse EC, Weinberg RA, Krummel MF, 2018, Understanding the tumor immune microenvironment, TIME, for effective therapy. Nat Med 24:541–550 Quail DF, Joyce JA, 2013, Microenvironmental regulation of tumor progression and metastasis. Nat Med 19:1423–1437 Balkwill FR, Capasso M, Hagemann T, 2012, The tumor microenvironment at a glance. J Cell Sci 125:5591–5596 Gajewski TF, Schreiber H, Fu Y-X, 2013, Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 14: 1014–1022 Munn DH, Bronte V, 2016, Immune suppressive mechanisms in the tumor microenvironment. Curr Opin Immunol 39:1–6 Chen DS, Mellman I, 2013, Oncology meets immunology: the cancer-immunity cycle. Immunity 39:1–10 Hanayama R, Tanaka M, Miwa K, Shinohara A, Iwamatsu A, Nagata S, 2002, Identification of a factor that links apoptotic cells to phagocytes. Nature 417:182–187 Asano K, Miwa M, Miwa K, Hanayama R, Nagase H, Nagata S, Tanaka M, 2004)Masking of phosphatidylserine inhibits apoptotic cell engulfment and induces autoantibody production in mice. J Exp Med 200:459–467 Jinushi M, Nakazaki Y, Dougan M, Carrasco DR, Mihm M, Dranoff G, 2007, MFG-E8-mediated uptake of apoptotic cells by APCs links the pro- and antiinflammatory activities of GM-CSF. J Clin Invest 117:1902–1913 Oba J, Moroi Y, Nakahara T, Abe T, Hagihara A, Furue M, 2011, Expression of milk fat globle epidermal growth factor-VIII may be an indicator of poor prognosis in malignant melanoma. Br J Dermatol 165:506–512 Yamazaki M, Maruyama S, Abe T, Essa A, Babkair H, Cheng J et al, 2014, MFG-E8 expression for progression of oral squamous cell carcinoma and for self-clearance of apoptotic cells. Lab Investig 94: 1260–1272 Jia M, Yao H, Chen C, Wang Y, Wang H, Cui T, Zhu J, 2017, Prognostic correlation between MFG-E8 expression level and colorectal cancer. Arch Med Res 48:270–275 Yamada K, Uchiyama A, Uehara A, Perera B, Ogino S, Yokoyama Y, Takeuchi Y, Udey MC, Ishikawa O, Motegi SI, 2016, MFG-E8 drives melanoma growth by stimulating mesenchymal stromal cellinduced angiogenesis and M2 polarization of tumor-associated macrophages. Cancer Res 76:4283–4292 Silvestre JS, Thery C, Hamard G, Boddaert J, Aguilar B, Delcayre A et al, 2005, Lactadherin promotes VEGF-dependent neovascularization. Nat Med 11:499–506 Reichert TE, Strauss L, Wagner EM, Gooding W, Whiteside TL, 2002, Signaling abnormalities, apoptosis, and reduced proliferation of circulating and tumor-infiltrating lymphocytes in patients with oral carcinoma. Clin Cancer Res 8:3137–3145 Buck AC, Schirrmeister HH, Guhlmann CA, Diederichs CG, Shen C, Buchmann I et al, 2001, Ki-67 immunostaining in pancreatic cancer and chronic active pancreatitis: does in vivo FDG uptake correlate with proliferative activity? J Nucl Med 42:721–725 Soki FN, Koh AJ, Jones JD, Kim YW, Dai J, Keller ET, Pienta KJ, Atabai K, Roca H, McCauley LK, 2014, Polarization of prostate cancer-associated macrophages is induced by milk fat globule-EGF factor 8, MFG-E8)-mediated efferocytosis. J Biol Chem 289: 24560–24572 Uchiyama A, Yamada K, Ogino S, Yokoyama Y, Takeuchi Y, Udey MC, Ishikawa O, Motegi SI, 2014, MFG-E8 regulates angiogenesis in cutaneous wound healing. Am J Pathol 184:1981–1990 Dai W, Li Y, Lv YN, Wei CD, Zheng HY, 2014, The roles of a novel anti-inflammatory factor, milk fat globule-epidermal growth factor 8, in patients with coronary atherosclerotic heart disease. Atherosclerosis 233:661–665 Motegi S, LeitnerWW, Lu M, Tada Y, Sardy M,Wu C et al, 2011, Pericyte-derived MFG-E8 regulates pathologic angiogenesis. Arterioscler Thromb Vasc Biol 31:2024–2034 Fonseca C, Dranoff G, 2008, Capitalizing on the immunogenicity of dying tumor cells. Clin Cancer Res 14:1603–1608 Quail DF, Joyce JA, 2013)Microenvironmental regulation of tumor progression and metastasis. Nat Med 19:1423–1437 Noy R, Pollard JW, 2014, Tumor-associated macrophages: from mechanisms to therapy. Immunity 41:49–61 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1263 EP 1268 DI 10.1007/s12253-019-00692-3 PG 6 ER PT J AU Zhou, Sh Tu, J Ding, Sh Lu, G Lin, Z Ding, Y Deng, B Zhang, Y Xiao, W Gong, W AF Zhou, Shuaiyang Tu, Jin Ding, Shizhen Lu, Guotao Lin, Zhijie Ding, Yanbing Deng, Bing Zhang, Yu Xiao, Weiming Gong, Weijuan TI High Expression of Angiopoietin-like Protein 4 in Advanced Colorectal Cancer and its Association with Regulatory T Cells and M2 Macrophages SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Angiopoietin-like protein 4; Treg; M2 macrophage; Colorectal cancer; Correlation ID Angiopoietin-like protein 4; Treg; M2 macrophage; Colorectal cancer; Correlation AB Colorectal cancer (CRC) is one of the most aggressive tumours in the human digestive system. Most CRC patients have poor prognosis due to metastasis and recurrence. Angiopoietin-like 4 (ANGPTL4) is involved in tumour development. Regulatory T (Treg) cells and M2 macrophages promote tumour growth and metastasis. Herein, we explored the changes of ANGPTL4 expression in CRC patients at different stages and observed whether in situ tumour-Treg and -M2 macrophages are correlated with ANGPTL4 expression. Serum ANGPTL4 (sANGPTL4) levels of 70 CRC patients and 10 healthy controls were detected by ELISA. ANGPTL4, Foxp3 and CD163 expression levels in CRC tissues were measured by immunohistochemistry. Recombinant ANGPTL4 (rANGPTL4) proteins were further added into cell-culture systems for induction of Treg cells and M2 macrophages. The results showed both sANGPTL4 and in situ tumour-ANGPTL4 expression levels increased in Dukes C–D stage CRC patients. Foxp3+ and CD163+ cells in tumour tissue sections were also more intensive in Dukes C–D stage patients than in Dukes A–B stage patients. Foxp3+ and CD163+ cells in tumour tissues were positively correlated with both tissue and sANGPTL4 expression (P < 0.01). Recombinant ANGPTL4 promoted the induction of murine Treg cells and M2 macrophages ex vivo. Therefore, elevated ANGPTL4 expression could be a marker for advanced CRC. Treg cell and M2 macrophage induction could be one of the mechanisms of tumour promotion mediated by ANGPTL4. C1 [Zhou, Shuaiyang] Yangzhou University, Affiliated Hospital, Department of Gastroenterology, 225001 Yangzhou, China. [Tu, Jin] Yangzhou University, Affiliated Hospital, Department of Gastroenterology, 225001 Yangzhou, China. [Ding, Shizhen] Yangzhou University, School of Medicine, Department of Basic Medicine, 225001 Yangzhou, China. [Lu, Guotao] Yangzhou University, Affiliated Hospital, Department of Gastroenterology, 225001 Yangzhou, China. [Lin, Zhijie] Yangzhou University, School of Medicine, Department of Basic Medicine, 225001 Yangzhou, China. [Ding, Yanbing] Yangzhou University, Affiliated Hospital, Department of Gastroenterology, 225001 Yangzhou, China. [Deng, Bing] Yangzhou University, Affiliated Hospital, Department of Gastroenterology, 225001 Yangzhou, China. [Zhang, Yu] Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, 225001 Yangzhou, China. [Xiao, Weiming] Yangzhou University, Affiliated Hospital, Department of Gastroenterology, 225001 Yangzhou, China. [Gong, Weijuan] Yangzhou University, Affiliated Hospital, Department of Gastroenterology, 225001 Yangzhou, China. RP Gong, W (reprint author), Yangzhou University, Affiliated Hospital, Department of Gastroenterology, 225001 Yangzhou, China. EM wjgong@yzu.edu.cn CR Mody K, Baldeo C, Bekaii-Saab T, 2018, Antiangiogenic therapy in colorectal Cancer. Cancer J 24:165–170 Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:E359–E386 Santulli G, 2014, Angiopoietin-like proteins: a comprehensive look. Front Endocrinol, Lausanne, 5: 4, eCollection 2014, 5 Zhu P, Goh YY, Chin HF, Kersten S, Tan NS, 2012, Angiopoietinlike 4: a decade of research. Biosci Rep 32:211–219 Sadeghabadi ZA, Nourbakhsh M, Alaee M, Larijani B, Razzaghy- Azar M, 2018, Peroxisome proliferator-activated receptor gamma expression in peripheral blood mononuclear cells and angiopoietinlike protein 4 levels in obese children and adolescents. J Endocrinol Investig 41:241–247 Tjeerdema N, Georgiadi A, Jonker JT, van Glabbeek M, Alizadeh Dehnavi R, Tamsma JT, Smit JW, Kersten S, Rensen PC, 2014, Inflammation increases plasma angiopoietin-like protein 4 in patients with the metabolic syndrome and type 2 diabetes. BMJ Open Diabetes Res Care 2:e000034 Tan MJ, Teo Z, Sng MK, Zhu P, Tan NS, 2012, Emerging roles of angiopoietin-like 4 in human cancer. Mol Cancer Res 10:677–688 Kim SH, Park YY, Kim SW, Lee JS, Wang D, DuBois RN, 2011, ANGPTL4 induction by prostaglandin E2 under hypoxic conditions promotes colorectal cancer progression. Cancer Res 71: 7010–7020 Zhu P, Tan MJ, Huang RL, Tan CK, Chong HC, Pal M, Lam CR, Boukamp P, Pan JY, Tan SH, 2011, Angiopoietin-like 4 protein elevates the prosurvival intracellular O2(−):H2O2 ratio and confers anoikis resistance to tumors. Cancer Cell 19:401–415 Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M, Zhu Y, Wei S, Kryczek I, Daniel B, Gordon A, Myers L, Lackner A, Disis ML, Knutson KL, Chen L, Zou W, 2004, Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 10:942–949 Roszer T, 2015, Understanding the mysterious M2 macrophage through activation markers and effector mechanisms. Mediat Inflamm 2015:816460 Hesketh M, Sahin KB, West ZE, Murray RZ, 2017, Macrophage phenotypes regulate scar formation and chronic wound healing. Int J Mol Sci 18 Nakayama T, Hirakawa H, Shibata K, Nazneen A, Abe K, Nagayasu T, Taguchi T, 2011, Expression of angiopoietin-like 4, ANGPTL4, in human colorectal cancer: ANGPTL4 promotes venous invasion and distant metastasis. Oncol Rep 25:929–935 XiaoW, YinM,Wu K, Lu G, Deng B, ZhangY, Qian L, Jia X, Ding Y, Gong W, 2017, High-dose wogonin exacerbates DSS-induced colitis by up-regulating effector T cell function and inhibiting Treg cell. J Cell Mol Med 21:286–298 Wang K, Vella AT, 2016, Regulatory T cells and Cancer: a twosided story. Immunol Investig 45:797–812 Young PP, Ardestani S, Li B, 2010, Myeloid cells in Cancer progression: unique subtypes and their roles in tumor growth, vascularity and host immune suppression. Cancer Microenviron 4:1–11 Goh YY, Pal M, Chong HC, Zhu P, Tan MJ, Punugu L, Tan CK, Huang RL, Sze SK, Tang MB, Ding JL, Kersten S, Tan NS, 2010, Angiopoietin-like 4 interacts with matrix proteins to modulate wound healing. J Biol Chem 285:32999–33009 Izraely S, Ben-Menachem S, Sagi-AssifO, Meshel T,Marzese DM, Ohe S, Zubrilov I, Pasmanik-Chor M, Hoon DSB, 2017, ANGPTL4 promotes the progression of cutaneous melanoma to brain metastasis. Oncotarget 8:75778–75796 Shafik NM, Mohamed DA, Bedder AE, El-Gendy AM, 2015, Significance of tissue expression and serum levels of angiopoietin-like protein 4 in breast Cancer progression: link to NF-κB /P65 activity and pro-inflammatory cytokines. Asian Pac J Cancer Prev 16:8579–8587 Ma T, Jham BC, Hu J, Friedman ER, Basile JR, Sodhi A, Montaner S, 2010, Upregulation of angiopoietin-like 4 by viral G protein-coupled receptor promotes angiogenesis and vascular permeability in Kaposi’s sarcoma. Infectious Agents and Cancer 5(Suppl 1):A84 Liao Y, LuW, Che Q, Yang T, Qiu H, Zhang H, He X,Wang J, Qiu M, Zou Y, GuW,Wan X, 2014, SHARP1 suppresses angiogenesis of endometrial cancer by decreasing hypoxia-inducible factor-1α level. PLoS One 9:e99907 Hata S, Nomura T, Iwasaki K, Sato R, YamasakiM, Sato F,Mimata H, 2017, Hypoxia-induced angiopoietin-like protein 4 as a clinical biomarker and treatment target for human prostate cancer. Oncol Rep 38:120–128 Yi J, Pan BZ, Xiong L, Song HZ, 2013, Clinical significance of angiopoietin-like protein 4 expression in tissue and serum of esophageal squamous cell carcinoma patients. Med Oncol 30:680 Huang Z, Xie J, Lin S, Li S, Huang Z, Wang Y, Ye J, 2016, The downregulation of ANGPTL4 inhibits the migration and proliferation of tongue squamous cell carcinoma. Arch Oral Biol 71:144– 149 El-Shal AS, Zidan HE, Rashad NM, Wadea FM, 2017, Angiopoietin-like protein 3 and 4 expression 4 and their serum levels in hepatocellular carcinoma. Cytokine 96:75–86 Chen JW, Luo YJ, Yang ZF,Wen LQ, Huang L, 2018, Knockdown of angiopoietin-like 4 inhibits the development of human gastric cancer. Oncol Rep 39:1739–1746 Kubo H, Kitajima Y, Kai K, Nakamura J,Miyake S, Yanagihara K, Morito K, Tanaka T, Shida M, Noshiro H, 2016, Regulation and clinical significance of the hypoxia-induced expression of ANGPTL4 in gastric cancer. Oncol Lett 11:1026–1034 Galaup A, Cazes A, Le Jan S, Philippe J, Connault E, Le Coz E, Mekid H, Mir LM, Opolon P, Corvol P, Monnot C, Germain S, 2006, Angiopoietin-like 4 prevents metastasis through inhibition of vascular permeability and tumor cell motility and invasiveness. Proc Natl Acad Sci U S A 103:18721–18726 Shang B, Liu Y, Jiang SJ, Liu Y, 2015, Prognostic value of tumorinfiltrating Foxp3 regulatory T cells in cancers: a systematic review and meta-analysis. Sci Rep 5:15179 Fujimoto H, Saito Y, OhuchidaK, Kawakami E, Fujiki S,Watanabe T, Ono R, Kaneko A, Takagi S, Najima Y, 2018, Deregulated mucosal immune surveillance through gut-associated regulatory T cells and PD-1 + T cells in human colorectal Cancer. J Immunol 200:3291–3303 Litwitz-Salomon E, Malyshkina A, Schimmer S, Dittmer U, 2018, The cytotoxic activity of natural killer cells is suppressed by IL-10 regulatory T cells during acute retroviral infection. Front Immunol 9:1947 Pandiyan P, Zheng L, Ishihara S, Reed J, LenardoMJ, 2007, CD4+ CD25+Foxp3+ regulatory T cells induce cytokine deprivationmediated apoptosis of effector CD4+ T cells. Nat Immunol 8: 1353–1362 Mempel TR, Pittet MJ, Khazaie K,WeningerW,Weissleder R, von Boehmer H, von Andrian UH, 2006, Regulatory T cells reversibly suppress cytotoxic T cell function independent of effector differentiation. Immunity 25:129–141 Xu P, Fan W, Zhang Z, Wang J, Wang P, Li Y, Yu M, 2017, The clinicopathological and prognostic implications of Foxp3+ regulatory Tcells in patients with colorectal cancer: a meta-analysis. Front Physiol 8:950 Correale P, Rotundo MS, Del Vecchio MT, Remondo C, Migali C, Ginanneschi C, Tsang KY, Licchetta A, Mannucci S, Loiacono L, Tassone P, Francini G, Tagliaferri P, 2010, Regulatory, Foxp3+, Tcell tumor infiltration is a favorable prognostic factor in advanced colon cancer patients undergoing chemo or chemoimmunotherapy. J Immunother 33:435–441 YoonHH, Orrock JM, Foster NR, Sargent DJ, Smyrk TC, Sinicrope FA, 2012, Prognostic impact of Foxp3+ regulatory T cells in relation to CD8+ T lymphocyte density in human colon carcinomas. PLoS One 7:e42274 Ladoire S, Martin F, Ghiringhelli F, 2011, Prognostic role of Foxp3+ regulatory T cells infiltrating human carcinomas: the paradox of colorectal cancer. Cancer Immunol Immunother 60:909–918 Lee CC, Lin JC, Hwang WL, Kuo YJ, Chen HK, Tai SK, Lin CC, Yang MH, 2018, Macrophage-secreted interleukin-35 regulates cancer cell plasticity to facilitate metastatic colonization. Nat Commun 9:3763 Lan J, Sun L, Xu F, Liu L, Hu F, Song D, Hou Z, Wu W, Luo X, Wang J, Yuan X, Hu J, Wang G, 2019, M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res 79:146–158 Zhou Q, Peng RQ, Wu XJ, Xia Q, Hou JH, Ding Y, Zhou QM, Zhang X, Pang ZZ, Wan DS, Zeng YX, Zhang XS, 2010, The density of macrophages in the invasive front is inversely correlated to liver metastasis in colon cancer. J Transl Med 8:13 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1269 EP 1278 DI 10.1007/s12253-019-00695-0 PG 10 ER PT J AU Gao, Bsh Rong, Chsh Xu, Hm Sun, T Hou, J Xu, Y AF Gao, Bao-shan Rong, Chun-shu Xu, Hong-mei Sun, Tao Hou, Jie Xu, Ying TI Peptidyl Arginine Deiminase, Type II (PADI2) Is Involved in Urothelial Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PADI2; Urothelial bladder cancer; Overexpressed; Tissue microarray analysis ID PADI2; Urothelial bladder cancer; Overexpressed; Tissue microarray analysis AB Peptidyl arginine deiminase, type II (PADI2) expression has been shown to potentiate multiple different carcinogenesis pathway including breast carcinoma and spontaneous skin neoplasia. The objective of this study was to examine the role of PADI2 in urothelial bladder cancer which has not been evaluated previously. Analysis of mutation and genome amplification of bladder cancer within The Cancer Genome Atlas (TCGA) showed that PADI2 is both mutated and amplified in a cohort of bladder cancer patients, with the largest number of mutations detected in urothelial bladder cancer. Even though PADI2 expression was not significantly correlated to survival in bladder cancer patients, it was significantly overexpressed at the mRNA and protein levels, as revealed by TCGA data and immunohistochemistry analysis, respectively. PADI2 showed wide expression pattern in bladder cancer tissues but was hardly detected in tumor adjacent normal tissue. RNAi mediated silencing of PADI2 in the bladder cancer cell line T24 did not result in a change of proliferation. Interestingly knockdown of PADI2 expression did not affect Snail1 protein, which is associated with metastatic progression, in these cells. However, PADI2 silencing remarkably attenuated both in vitro migration and invasion- in T24 cells indicating a Snail1-independent effect of PADI2 on invasive potential of urothelial bladder cancer. This was further corroborated by in vivo xenograft assays where PADI2 shRNA harboring T24 cells did not have detectable tumors by week 4 as compared to robust tumors in the control Luciferase shRNA harboring cells. PADI2 silencing did not affect proliferation rates and hence this would suggest that PADI2 knockdown is perhaps causing increased apoptosis as well as transition through the cell cycle, which needs to be confirmed in future studies. Our results reveal a yet undefined role of PADI2 as an oncogene in urothelial bladder cancer. C1 [Gao, Bao-shan] The First Hospital of Jilin University, Urology Center, No.71, Xinmin Street, 130021 Changchun, Jilin, China. [Rong, Chun-shu] The Affiliated Hospital to Changchun University of Chinese Medicine, Department of Encephalopathy Diseases, 130021 Changchun, Jilin, China. [Xu, Hong-mei] The First Hospital of Jilin University, Obstetric Department, 130021 Changchun, Jilin, China. [Sun, Tao] The First Hospital of Jilin University, Urology Center, No.71, Xinmin Street, 130021 Changchun, Jilin, China. [Hou, Jie] The First Hospital of Jilin University, Urology Center, No.71, Xinmin Street, 130021 Changchun, Jilin, China. [Xu, Ying] The First Hospital of Jilin University, Urology Center, No.71, Xinmin Street, 130021 Changchun, Jilin, China. RP Xu, Y (reprint author), The First Hospital of Jilin University, Urology Center, 130021 Changchun, China. EM yingxu_jilinuni@126.com CR Murta-Nascimento C, Schmitz-Drager BJ, ZeegersMP, Steineck G, Kogevinas M, Real FX, Malats N, 2007, Epidemiology of urinary bladder cancer: from tumor development to patient's death.World J Urol 25(3):285–295 Kogevinas M, Mannetje AT, Cordier S, Ranft U, Gonzalez CA, Vineis P, Chang-Claude J, Lynge E, Wahrendorf J, Tzonou A, 2003, Occupation and bladder cancer among men in Western Europe. Cancer Causes Control 14(10):907–914 Zeegers MPA, Tan FES, Dorant E, Van dB, Piet A, 2015, The impact of characteristics of cigarette smoking on urinary tract cancer risk. Cancer 89(3):630–639 Yin J, Wang C, Liang D, Vogel U, Li Y, Liu J, Qi R, Sun X, 2012, No evidence of association between the synonymous polymorphisms in XRCC1 and ERCC2 and breast cancer susceptibility among nonsmoking Chinese. Gene 503(1):118–122 Wei Q, Spitz MR, 1997, The role of DNA repair capacity in susceptibility to lung cancer: a review. Cancer Metastasis Rev 16(3–4): 295–307 Sturgis EM, Castillo EJ, Li L, Zheng R, Eicher SA, Clayman GL, Strom SS, Spitz MR,Wei Q, 1999, Polymorphisms of DNA repair gene XRCC1 in squamous cell carcinoma of the head and neck. Carcinogenesis 20(11):2125–2129 Iii MCM, Mohrenweiser HW, Bell DA, 2001, Genetic variability in susceptibility and response to toxicants. Toxicol Lett 120(1):259–268 Alberg AJ, Jorgensen TJ, Ingo R, LeeW,Yao SY, Yvette BS, Bailey K, Judith HB, Helzlsouer KJ, Kao WHL, 2013, DNA repair gene variants in relation to overall cancer risk: a population-based study. Carcinogenesis 34(1):86–92 Kury S, Buecher B, Robiou-Du-Pont S, Scoul C, Colman H, Neel TL, Houerou CL, Faroux R, Ollivry J, Lafraise B, 2008, Lowpenetrance alleles predisposing to sporadic colorectal cancers: a French case-controlled genetic association study. BMC Cancer 8(1):326–326 Mcelwee JL, Sunish M, Sachi H, Sams KL, Anguish LJ, Dalton ML, Iva C,Wakshlag JJ, Coonrod SA, 2014, PAD2 overexpression in transgenic mice promotes spontaneous skin neoplasia. Cancer Res 74(21):6306–6317 Mohanan S, Cherrington BD, Horibata S, Mcelwee JL, Thompson PR, Coonrod SA, 2012, Potential role of peptidylarginine deiminase enzymes and protein citrullination in cancer pathogenesis. Biochem Res Int 2012(5):895343 Cherrington BD, Mohanan S, Diep AN, Fleiss R, Sudilovsky D, Anguish LJ, Coonrod SA, Wakshlag JJ, 2012, Comparative analysis of Peptidylarginine Deiminase-2 expression in canine, feline and human mammary tumours. J Comp Pathol 147(2–3):139–146 Cherrington BD, Morency E, Struble AM, Coonrod SA,Wakshlag JJ, 2010, Potential role for peptidylarginine deiminase 2, PAD2, in citrullination of canine mammary epithelial cell histones. PLoS One 5(7):e11768 Cherrington BD, Xuesen Z, Mcelwee JL, Eric M, Anguish LJ, Coonrod SA, 2012, Potential role for PAD2 in gene regulation in breast cancer cells. PLoS One 7(7):e41242 Mcelwee JL, Mohanan S, Griffith OL, Breuer HC, Anguish LJ, Cherrington BD, Palmer AM, Howe LR, Subramanian V, Causey CP, 2012, Identification of PADI2 as a potential breast cancer biomarker and therapeutic target. BMC Cancer 12(1):500–500 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, Cerami E, Sander C, Schultz N, 2013, Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6(269):l1 Unberath P, Knell C, Prokosch HU, Christoph J, 2019, Developing new analysis functions for a translational research platform: extending the cBioPortal for cancer genomics. Stud Health Technol Inform 258:46–50 Shoji N, Yasuto U, Hiroshi O,Masataka M, Tetsuro S, Kiyokazu T, Yoshiaki K, Akihiko S, Testuhiro O, Sumiya I, 2007, Snail plays a key role in E-cadherin-preserved esophageal squamous cell carcinoma. Oncol Rep 17(3):517–523 Zheng H, ., Kang Y, 2014, Multilayer control of the EMT master regulators. Oncogene 33, 14):1755–1763 Kalluri R, 2009, EMT: when epithelial cells decide to become mesenchymal-like cells. J Clin Invest 119(6):1417–1419. https:// doi.org/10.1172/jci39675 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1279 EP 1285 DI 10.1007/s12253-019-00687-0 PG 7 ER PT J AU Kadletz, CL Brkic, FF Jank, JB Schneider, S Cede, J Seemann, R Gruber, SE Gurnhofer, E Heiduschka, G Kenner, L AF Kadletz, C Lorenz Brkic, F Faris Jank, J Bernhard Schneider, Sven Cede, Julia Seemann, Rudolf Gruber, S Elisabeth Gurnhofer, Elisabeth Heiduschka, Gregor Kenner, Lukas TI AF1q Expression Associates with CD44 and STAT3 and Impairs Overall Survival in Adenoid Cystic Carcinoma of the Head and Neck SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Adenoid cystic carcinoma; AF1q; Wnt; STAT3; Prognosis ID Adenoid cystic carcinoma; AF1q; Wnt; STAT3; Prognosis AB Salivary gland malignancies of the head and neck form a heterogeneous group. Adenoid cystic carcinomas are an aggressive entity of salivary gland malignancies characterized by frequent distant metastases and poor response to radio- and chemotherapy. AF1Q is a MLL fusion partner, which can activate Wnt and STAT3 signaling. Recently, overexpression of AF1q has been identified as a poor prognosticator in patients of different malignancies. A total of 46 patients with adenoid cystic carcinoma were immunohistochemically evaluated for expression of AF1q and clinical outcome was analyzed in this context. Additionally, STAT3 and the Wnt downstream target CD44 were investigated and correlated with AF1q. AF1q was overexpressed in 52.2%. Overexpression of AF1q was associated with poorer overall survival (p = 0.03). Additionally, lymph node metastases and solid tumor parts were more frequently observed in AF1qhigh patients (p = 0.07 and 0.05, respectively). AF1q did not influence the occurrence of distant metastases. Expression of AF1q was associated with higher levels of STAT3 and CD44 (p = 0.003 and 0.006, respectively). AF1q is a novel prognostic marker for poor overall survival in adenoid cystic carcinoma patients. The deleterious effects on survival may be a result of promotion of the STAT3 and Wnt pathway. C1 [Kadletz, C Lorenz] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck SurgeryVienna, Austria. [Brkic, F Faris] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck SurgeryVienna, Austria. [Jank, J Bernhard] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck SurgeryVienna, Austria. [Schneider, Sven] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck SurgeryVienna, Austria. [Cede, Julia] Medical University of Vienna, Department of Cranio-maxillo and Facial SurgeryVienna, Austria. [Seemann, Rudolf] Medical University of Vienna, Department of Cranio-maxillo and Facial SurgeryVienna, Austria. [Gruber, S Elisabeth] Medical University of Vienna, Department of SurgeryVienna, Austria. [Gurnhofer, Elisabeth] Medical University of Vienna, Department of Experimental Pathology and Laboratory Animal PathologyVienna, Austria. [Heiduschka, Gregor] Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck SurgeryVienna, Austria. [Kenner, Lukas] Medical University of Vienna, Department of Experimental Pathology and Laboratory Animal PathologyVienna, Austria. RP Kadletz, CL (reprint author), Medical University of Vienna, Department of Oto-rhino-laryngology and Head and Neck Surgery, Vienna, Austria. EM lorenz.kadletz@meduniwien.ac.at CR Sood S, McGurk M, Vaz F, 2016, J Larnygol Otol 130:S142 Barnes L, Eveson JW, Reichart P, Sidransky D, 2005, Pathology and genetics of head and neck Tumours. IARC Press Dockerty MB, Mayo CW, 1942, Surg Gynecol Obstet 1033 Lloyd S, Yu JB, Wilson LD, Decker RH, 2011, Am J Clin Oncol 34:76 van Weert S, Reinhard R, Bloemena E, Buter J, Witte BI, Vergeer MR, Leemans CR, 2016, Head Neck 39:456 Fordice J, Kershaw C, El-Naggar A, Goepfert H, 1999, Arch Otolaryngol Head Neck Surg 125:149 Park J, Schlederer M, Schreiber M, Ice R, Merkel O, Bilban M, Hofbauer S, Kim S, Addison J, Zou J, Ji C, Bunting ST, Wang Z, Shoham M, Huang G, Bago-Horvath Z, Gibson LF, Rojanasakul Y, Remick S, Ivanov A, Pugacheva E, Bunting KD, Moriggl R, Kenner L, Tse W, 2015, Oncotarget 6(20697) TSE W, ZHU WM, CHEN HS, COHEN A, 1995, Blood 85:650 Co NN, Tsang WP, Tsang TY, Yeung CLA, Yau PL, Kong SK, Kwok TT, 2010, Oncol Rep 24:547 Tiberio P, Cavadini E, Callari M, Daidone MG, Appierto V, 2012, PLoS One 7:e39968 Wang R, Geng N, Zhou Y, Zhang D, Li L, Li J, Ji N, ZhouM, Chen Y, Chen Q, 2015, Biomed Mater Eng 26:S2145 Schneider S, Thurnher D, Seemann R, Brunner M, Kadletz L, Ghanim B, Aumayr K, Heiduschka G, Lill C, 2015, Eur Arch Otorhinolaryngol 273:1283 Lill C, Schneider S, Seemann R, Kadletz L, Aumayr K, Ghanim B, Thurnher D, 2014, Head Neck 37:30 Park J, Kim S, Joh J, Remick SC, Miller DM, Yan J, Kanaan Z, Chao J-H, KremMM, Basu SK, Hagiwara S, Kenner L,Moriggl R, Bunting KD, Tse W, 2016, Oncotarget 7(43960) Bu L-L,DengW-W, Huang C-F, Liu B, ZhangW-F, Sun Z-J, 2015, Am J Cancer Res 5:1751 Spiro RH, Huvos AG, Strong EW, 1974, Am J Surg 128:512 Szanto PA, Luna MA, Tortoledo ME, White RA, 1984, Cancer 54: 1062 Perzin KH, Gullane P, Clairmont AC, 1978, Cancer 42:265 Hu J, Li G, Liu L, Wang Y, Li X, Gong J, 2017, Int J Mol Sci 18 Tiberio P, Lozneanu L, Angeloni V, Cavadini E, Pinciroli P, Callari M, Carcangiu ML, Lorusso D, Raspagliesi F, Pala V, DaidoneMG, Appierto V, 2017, Oncotarget 8(23246) Tse CO, KimS, Park J, 2017, BiochemBiophys Res Commun 484: 884 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1287 EP 1292 DI 10.1007/s12253-019-00696-z PG 6 ER PT J AU Liu, Q Pan, Lz Hu, M Ma, Jy AF Liu, Qin Pan, Liang-zhi Hu, Min Ma, Jian-ying TI Molecular Network-Based Identification of Circular RNA-Associated ceRNA Network in Papillary Thyroid Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Papillary thyroid cancer; circRNA; Competitive endogenous RNA; microRNA ID Papillary thyroid cancer; circRNA; Competitive endogenous RNA; microRNA AB Circular RNAs (circRNAs) have displayed dysregulated expression in several types of cancer. Nevertheless, their function and underlying mechanisms in papillary thyroid cancer (PTC) remains largely unknown. This study aimed to describe the regulatory mechanisms in PTC. The expression profile of circRNA was download from the Gene ExpressionOmnibus (GEO) database. The mRNA and miRNA data of PTC was downloaded from The Cancer Genome Atlas (TCGA) database. The circRNA-miRNA-mRNA network by Cytoscape. The interactions between proteins were analyzed using the STRING database and hubgenes were identified using MCODE plugin. Then, we conducted a circRNA-miRNA-hubgenes regulatory module. Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genomes (KEGG) pathway analysis were conducted using R packages “Clusterprofile”. We identified 14 differential expression circRNAs (DEcircRNA), 3106 differential expression mRNAs (DEmRNA), 142 differential expression miRNAs (DEmiRNA) and in PTC. Twelve circRNAs, 33 miRNAs, and 356 mRNAs were identified to construct the ceRNA network of PTC. PPI network and module analysis identified 5 hubgenes. Then, a circRNA-miRNA-hubgene subnetwork was constructed based on the 2 DEcircRNAs, 3 DEmiRNAs, and 4 DEmRNAs. GO and KEGG pathway analysis indicated DEmRNAs might be associated with PTC onset and progression. These ceRNAs are critical in the pathogenesis of PTC and may serve as future therapeutic biomarkers. C1 [Liu, Qin] Hubei Polytechnic University, Huangshi Central Hospital of Edong Healthcare Group, Department of Breast Surgery, Thyroid Surgery, No.141, Tianjin RoadHuangshi, Hubei, China. [Pan, Liang-zhi] Hubei Polytechnic University, Huangshi Central Hospital (Pu Ai Hospital) of Edong Healthcare Group, Personnel SectionHuangshi, Hubei, China. [Hu, Min] Hubei Polytechnic University, Huangshi Central Hospital (Pu Ai Hospital) of Edong Healthcare Group, Personnel SectionHuangshi, Hubei, China. [Ma, Jian-ying] Hubei Polytechnic University, Huangshi Central Hospital of Edong Healthcare Group, Department of Breast Surgery, Thyroid Surgery, No.141, Tianjin RoadHuangshi, Hubei, China. RP Ma, Jy (reprint author), Hubei Polytechnic University, Huangshi Central Hospital of Edong Healthcare Group, Department of Breast Surgery, Thyroid Surgery, Huangshi, China. EM majianying2015@sina.com CR Siegel RL, Miller KD, Jemal A, 2018, Cancer statistics, 2018. CA Cancer J Clin 68(1):7–30. , DOI 10.3322/caac.21442 Mao Y, XingM, 2016, Recent incidences and differential trends of thyroid cancer in the USA. Endocr Relat Cancer 23(4):313–322. , DOI 10.1530/erc-15-0445 Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM, Schlumberger M, Schuff KG, Sherman SI, Sosa JA, Steward DL, Tuttle RM, Wartofsky L, 2016, 2015 American Thyroid Association management guidelines for adult patients with thyroid nodules and differentiated thyroid Cancer: the American Thyroid Association guidelines task force on thyroid nodules and differentiated thyroid Cancer. Thyroid. 26(1):1–133. , DOI 10.1089/thy.2015. 0020 Liu FH, Kuo SF, Hsueh C, Chao TC, Lin JD, 2015, Postoperative recurrence of papillary thyroid carcinoma with lymph node metastasis. J Surg Oncol 112(2):149–154. , DOI 10.1002/jso. 23967 Grant CS, 2015, Recurrence of papillary thyroid cancer after optimized surgery. Gland Surg 4(1):52–62. , DOI 10.3978/j. issn.2227-684X.2014.12.06 Qu S, Yang X, Li X,Wang J, Gao Y, Shang R, SunW, Dou K, Li H, 2015, Circular RNA: a new star of noncoding RNAs. Cancer Lett 365(2):141–148. , DOI 10.1016/j.canlet.2015.06.003 Chen LL, 2016, The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol 17(4):205–211. , DOI 10. 1038/nrm.2015.32 Wang F, Nazarali AJ, Ji S, 2016, Circular RNAs as potential biomarkers for cancer diagnosis and therapy. Am J Cancer Res 6(6): 1167–1176 Li J, Yang J, Zhou P, le Y, Zhou C,Wang S, Xu D, Lin HK, Gong Z, 2015, Circular RNAs in cancer: novel insights into origins, properties, functions and implications. Am J Cancer Res 5(2):472–480 Liu M, Liu KD, Zhang L et al, 2018, Circ_0009910 regulates growth and metastasis and is associated with poor prognosis in gastric cancer. J Cell Biochem 22(23):8248–8256. , DOI 10.26355/eurrev_201812_16519 Zhang X, Xu Y, Qian Z et al, 2018, circRNA_104075 stimulates YAP-dependent tumorigenesis through the regulation of HNF4a and may serve as a diagnostic marker in hepatocellular carcinoma. Cell Death Dis 9(11):1091. , DOI 10.1038/s41419-018- 1132-6 Zhang G, SunW, Zhu L, Feng Y,Wu L, Li T, 2018, Overexpressed circ_0029426 in glioblastoma forecasts unfavorable prognosis and promotes cell progression by sponging miR-197. J Cell Biochem 120:10295–10302. , DOI 10.1002/jcb.28313 Liu G, Huang K, Jie Z, Wu Y, Chen J, Chen Z, Fang X, Shen S, 2018, CircFAT1 sponges miR-375 to promote the expression of yes-associated protein 1 in osteosarcoma cells. Mol Cancer 17(1): 170. , DOI 10.1186/s12943-018-0917-7 Li X, Zhang Z, Jiang H, Li Q,Wang R, Pan H, Niu Y, Liu F, Gu H, Fan X, Gao J, 2018, Circular RNA circPVT1 promotes proliferation and invasion through sponging miR-125b and activating E2F2 signaling in non-small cell lung Cancer. J Cell Biochem 51(5): 2324–2340. , DOI 10.1159/000495876 Wang R, Zhang S, Chen X et al, 2018, EIF4A3-induced circular RNA MMP9, circMMP9, acts as a sponge of miR-124 and promotes glioblastoma multiforme cell tumorigenesis. 17(1):166. , DOI 10.1186/s12943-018-0911-0 Wong N, Wang X, 2015, miRDB: an online resource for microRNA target prediction and functional annotations. Nucleic Acids Res 43(Database issue):D146–D152. , DOI 10. 1093/nar/gku1104 Fromm B, Billipp T, Peck LE, Johansen M, Tarver JE, King BL, Newcomb JM, Sempere LF, Flatmark K, Hovig E, Peterson KJ, 2015, A uniform system for the annotation of vertebrate microRNA genes and the evolution of the human microRNAome. Annu Rev Genet 49:213–242. , DOI 10.1146/annurevgenet- 120213-092023 Yu G,Wang LG, Han Y, He QY, 2012, clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 16(5):284–287. , DOI 10.1089/omi.2011.0118 Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J, Marzluff WF, Sharpless NE, 2013, Circular RNAs are abundant, conserved, and associated withALU repeats. RNA. 19(2):141–157. , DOI 10.1261/rna.035667.112 Rybak-Wolf A, Stottmeister C, Glazar P et al, 2015, Circular RNAs in the mammalian brain are highly abundant, conserved, and dynamically expressed. Mol Cell 58(5):870–885. , DOI 10. 1016/j.molcel.2015.03.027 Lux S, Bullinger L, 2018, Circular RNAs in Cancer. Adv Exp Med Biol 1087:215–230. , DOI 10.1007/978-981-13-1426-1_17 Chen S, Zhao Y, 2018, Circular RNAs: characteristics, function, and role in human cancer. Histol Histopathol 33(9):887–893. , DOI 10.14670/hh-11-969 Kristensen LS, Hansen TB, Veno MT, Kjems J, 2018, Circular RNAs in cancer: opportunities and challenges in the field. Oncogene. 37(5):555–565. , DOI 10.1038/onc.2017.361 Zhong Y, Du Y, Yang X et al, 2018, Circular RNAs function as ceRNAs to regulate and control human cancer progression. Mol Cancer 17(1):79. , DOI 10.1186/s12943-018-0827-8 Chen F, Feng Z, Zhu J, Liu P, Yang C, Huang R, Deng Z, 2018, Emerging roles of circRNA_NEK6 targeting miR-370-3p in the proliferation and invasion of thyroid cancer viaWnt signaling pathway. Cancer Biol Ther 19(12):1139–1152. , DOI 10.1080/ 15384047.2018.1480888 Wei H, Pan L, Tao D, Li R, 2018, Circular RNA circZFR contributes to papillary thyroid cancer cell proliferation and invasion by sponging miR-1261 and facilitating C8orf4 expression. Biochem Biophys Res Commun 503(1):56–61. , DOI 10.1016/j. bbrc.2018.05.174 Liu F, Zhang J, Qin L et al, 2018, Circular RNA EIF6, Hsa_circ_0060060, sponges miR-144-3p to promote the cisplatin-resistance of human thyroid carcinoma cells by autophagy regulation. Aging, Albany NY, 10(12):3806–3820. , DOI 10.18632/aging.101674 Jin X,Wang Z, PangWet al, 2018, Upregulated hsa_circ_0004458 contributes to progression of papillary thyroid carcinoma by inhibition of miR-885-5p and activation of RAC1. Med Sci Monit 24: 5488–5500. , DOI 10.12659/msm.911095 Jiang W, Wen D, Gong L, Wang Y, Liu Z, Yin F, 2018, Circular RNA hsa_circ_0000673 promotes hepatocellular carcinoma malignance by decreasing miR-767-3p targeting SET. Biochem Biophys Res Commun 500(2):211–216. , DOI 10.1016/j.bbrc.2018. 04.041 Chang P, Wang F, Li Y, 2018, Hsa_circ_0000673 is downregulated in gastric cancer and inhibits the proliferation and invasion of tumor cells by targetting miR-532-5p. Biosci Rep 38(5). , DOI 10.1042/bsr20180538 Bartel DP, 2018, Metazoan MicroRNAs. Cell. 173(1):20–51. , DOI 10.1016/j.cell.2018.03.006 Bartel DP, 2009, MicroRNAs: target recognition and regulatory functions. Cell. 136(2):215–233. , DOI 10.1016/j.cell. 2009.01.002 Liu C, Liu R, Zhang D, Deng Q, Liu B, Chao HP, Rycaj K, Takata Y, Lin K, Lu Y, Zhong Y, Krolewski J, Shen J, Tang DG, 2017, MicroRNA-141 suppresses prostate cancer stem cells and metastasis by targeting a cohort of pro-metastasis genes. Nat Commun 8: 14270 SongHM, LuoY, LiDF,Wei CK,HuaKY, Song JL, Xu H,Maskey N, Fang L, 2015, MicroRNA-96 plays an oncogenic role by targeting FOXO1 and regulating AKT/FOXO1/Bim pathway in papillary thyroid carcinoma cells. Int J Clin Exp Pathol 8(9): 9889–9900 Spitschak A, Meier C, Kowtharapu B, Engelmann D, Putzer BM, 2017, MiR-182 promotes cancer invasion by linking RET oncogene activated NF-kappaB to loss of the HES1/Notch1 regulatory circuit. Mol Cancer 16(1):24. , DOI 10.1186/s12943-016- 0563-x Wu ZY,Wang SM, Chen ZH, Huv SX, Huang K, Huang BJ, du JL, Huang CM, Peng L, Jian ZX, Zhao G, 2015, MiR-204 regulates HMGA2 expression and inhibits cell proliferation in human thyroid cancer. Cancer Biomark 15(5):535–542. , DOI 10.3233/ cbm-150492 Li J, Zheng X, Gao M, Zhao J, Li Y, Meng X, Qian B, Li J, 2017, Suberoyl bis-hydroxamic acid activates Notch1 signaling and induces apoptosis in anaplastic thyroid carcinoma through p53. Oncol Rep 37(1):458–464. , DOI 10.3892/or.2016.5281 Kim YH, Choi YW, Lee J, Soh EY, Kim JH, Park TJ, 2017, Senescent tumor cells lead the collective invasion in thyroid cancer. Nat Commun 8:15208. , DOI 10.1038/ncomms15208 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1293 EP 1299 DI 10.1007/s12253-019-00697-y PG 7 ER PT J AU Liu, Ch Xu, D Xue, B Liu, B Li, J Huang, J AF Liu, Chao Xu, Dingwei Xue, Bai Liu, Bolin Li, Jing Huang, Jie TI Upregulation of RUNX1 Suppresses Proliferation and Migration through Repressing VEGFA Expression in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; RUNX1; VEGFA; Tumor suppression ID Hepatocellular carcinoma; RUNX1; VEGFA; Tumor suppression AB Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, and occurs in people with chronic liver diseases. Current treatment methods include surgery, transplant, and chemotherapy. Our study demonstrates runt-related transcription factor 1 (RUNX1) as a novel molecule in the initiation and development of HCC, and the role of its interaction with vascular endothelial growth factor A (VEGFA) in HCC. We showed the suppressive role of RUNX1 in the proliferation and migration of hepatocytes. In addition, the repressor RUNX1 functioned as a transcription factor on the promoter of VEGFA to inhibit the expression of VEGFA. Study in the HCC cells demonstrated that the suppression of HCC proliferation and migration was masked in the presence of overexpressed VEGFA. Introduction of RUNX1 into HCC mice model significantly limited the tumor growth. In summary, our study demonstrated that RUNX1 functions as a repressor in the HCC and this suppressive function was dependent on its effect on VEGFA. C1 [Liu, Chao] Jiangsu College of Nursing, School of Medical Technology, 223005 Huainan, Jiangsu Province, China. [Xu, Dingwei] The Second Affiliation Hospital of Kunming Medical University, Department of Hepatobiliary Surgery, 243 Dianmian Avenue, Wuhua District, 650101 Kunming, Yunnan Province, China. [Xue, Bai] Jiangsu College of Nursing, School of Medical Technology, 223005 Huainan, Jiangsu Province, China. [Liu, Bolin] Jiangsu College of Nursing, School of Medical Technology, 223005 Huainan, Jiangsu Province, China. [Li, Jing] Jiangsu College of Nursing, School of Medical Technology, 223005 Huainan, Jiangsu Province, China. [Huang, Jie] The Second Affiliation Hospital of Kunming Medical University, Department of Hepatobiliary Surgery, 243 Dianmian Avenue, Wuhua District, 650101 Kunming, Yunnan Province, China. RP Huang, J (reprint author), The Second Affiliation Hospital of Kunming Medical University, Department of Hepatobiliary Surgery, 650101 Kunming, China. CR Tunissiolli NM, Castanhole-Nunes MMU, Biselli-Chicote PM, Pavarino EC, da Silva RF, da Silva RC, Goloni-Bertollo EM, 2017, Hepatocellular Carcinoma: a Comprehensive Review of Biomarkers, Clinical Aspects, and Therapy. Asian Pac J Cancer Prev 18(4):863– 872. , DOI 10.22034/APJCP.2017.18.4.863 Gomes MA, Priolli DG, Tralhao JG, Botelho MF, 2013, Hepatocellular carcinoma: epidemiology, biology, diagnosis, and therapies. Rev Assoc Med Bras 59(5):514–524. , DOI 10. 1016/j.ramb.2013.03.005 Forner A, Llovet JM, Bruix J, 2012, Hepatocellular carcinoma. Lancet 379(9822):1245–1255. , DOI 10.1016/S0140- 6736(11)61347-0 Forner A, Gilabert M, Bruix J, Raoul JL, 2014, Treatment of intermediate-stage hepatocellular carcinoma. Nat Rev Clin Oncol 11(9):525–535. , DOI 10.1038/nrclinonc.2014.122 Sood R, Kamikubo Y, Liu P, 2017, Role of RUNX1 in hematological malignancies. Blood 129(15):2070–2082. , DOI 10. 1182/blood-2016-10-687830 Ito Y, Bae SC, Chuang LS, 2015, The RUNX family: developmental regulators in cancer. Nat Rev Cancer 15(2):81–95. https://doi. org/10.1038/nrc3877 Wu J, Huang G, Ratner N, 2015, Runx1: a new driver in neurofibromagenesis. Oncoscience 2(11):904–905. , DOI 10.18632/oncoscience.266 Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson A, Kampf C, Sjostedt E, Asplund A, Olsson I, Edlund K, Lundberg E, Navani S, Szigyarto CA, Odeberg J,Djureinovic D, Takanen JO, Hober S, Alm T, Edqvist PH, Berling H, Tegel H, Mulder J, Rockberg J, Nilsson P, Schwenk JM, Hamsten M, von Feilitzen K, Forsberg M, Persson L, Johansson F, ZwahlenM, von Heijne G, Nielsen J, Ponten F, 2015, Proteomics. Tissue-based map of the human proteome. Science 347(6220):1260419. , DOI 10. 1126/science.1260419 Goyama S, Huang G, Kurokawa M, Mulloy JC, 2015, Posttranslational modifications of RUNX1 as potential anticancer targets. Oncogene 34(27):3483–3492. , DOI 1038/onc.2014.305 10. Carmeliet P, 2005, VEGF as a key mediator of angiogenesis in cancer. Oncology 69(Suppl 3):4–10. , DOI 10.1159/000088478 HansenW, Hutzler M, Abel S, Alter C, Stockmann C, Kliche S, Albert J, Sparwasser T, Sakaguchi S,Westendorf AM, Schadendorf D, Buer J, Helfrich I, 2012, Neuropilin 1 deficiency on CD4+Foxp3+ regulatory T cells impairs mouse melanoma growth. J Exp Med 209(11):2001– 2016. , DOI 10.1084/jem.20111497 Goel HL, Mercurio AM, 2013, VEGF targets the tumour cell. Nat Rev Cancer 13(12):871–882. , DOI 10.1038/nrc3627 Weis SM, Cheresh DA, 2005, Pathophysiological consequences of VEGF-induced vascular permeability. Nature 437(7058):497–504. , DOI 10.1038/nature03987 ShibuyaM(2013, Vascular endothelial growth factor and its receptor system: physiological functions in angiogenesis and pathological roles in various diseases. J Biochem 153(1):13–19. https://doi. org/10.1093/jb/mvs136 Voron T, Colussi O, Marcheteau E, Pernot S, Nizard M, Pointet AL, Latreche S, Bergaya S, Benhamouda N, Tanchot C, Stockmann C, Combe P, Berger A, Zinzindohoue F, Yagita H, Tartour E, Taieb J, Terme M, 2015, VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med 212(2):139– 148. , DOI 10.1084/jem.20140559 Horwitz E, Stein I, Andreozzi M, Nemeth J, Shoham A, Pappo O, Schweitzer N, Tornillo L, Kanarek N, Quagliata L, Zreik F, Porat RM, Finkelstein R, Reuter H, Koschny R, Ganten T, Mogler C, Shibolet O, Hess J, Breuhahn K, Grunewald M, Schirmacher P, Vogel A, Terracciano L, Angel P, Ben-Neriah Y, Pikarsky E, 2014, Human and mouse VEGFA-amplified hepatocellular carcinomas are highly sensitive to sorafenib treatment. Cancer Discov 4(6):730–743. , DOI 10.1158/2159-8290.CD-13-0782 Gascon E, Gaillard S, Malapert P, Liu Y, Rodat-Despoix L, Samokhvalov IM, Delmas P, Helmbacher F, Maina F, Moqrich A, 2010, Hepatocyte growth factor-Met signaling is required for Runx1 extinction and peptidergic differentiation in primary nociceptive neurons. J Neurosci 30(37):12414–12423. , DOI 10.1523/JNEUROSCI.3135-10.2010 Ter Elst A, Ma B, Scherpen FJ, de Jonge HJ, Douwes J,Wierenga AT, Schuringa JJ, Kamps WA, de Bont ES, 2011, Repression of vascular endothelial growth factor expression by the runt-related transcription factor 1 in acute myeloid leukemia. Cancer Res 71(7):2761–2771. , DOI 10.1158/0008-5472.CAN-10-0402 Bowers SR, Calero-Nieto FJ, Valeaux S, Fernandez-Fuentes N, Cockerill PN, 2010, Runx1 binds as a dimeric complex to overlapping Runx1 sites within a palindromic element in the human GMC CSF enhancer. Nucleic Acids Res 38(18):6124–6134. https://doi. org/10.1093/nar/gkq356 Klaunig JE, Pereira MA, Ruch RJ, Weghorst CM, 1988, Doseresponse relationship of diethylnitrosamine-initiated tumors in neonatal balb/cmice: effect of phenobarbital promotion. Toxicol Pathol 16(3):381–385. , DOI 10.1177/019262338801600310 Wang L, Brugge JS, Janes KA, 2011, Intersection of FOXO- and RUNX1-mediated gene expression programs in single breast epithelial cells during morphogenesis and tumor progression. Proc Natl Acad Sci U S A 108(40):E803–E812. , DOI 10. 1073/pnas.1103423108 Mercado-Matos J, Matthew-Onabanjo AN, Shaw LM, 2017, RUNX1 and breast cancer. Oncotarget 8(23):36934–36935. , DOI 10.18632/oncotarget.17249 Barnum KJ, O'Connell MJ, 2014, Cell cycle regulation by checkpoints. Methods Mol Biol 1170:29–40. , DOI 10.1007/ 978-1-4939-0888-2_2 Cai D, Chen SC, PrasadM, He L,Wang X, Choesmel-Cadamuro V, Sawyer JK, Danuser G, Montell DJ, 2014, Mechanical feedback through E-cadherin promotes direction sensing during collective cell migration. Cell 157(5):1146–1159. , DOI 10.1016/j. cell.2014.03.045 Battaglia RA, Delic S, Herrmann H, Snider NT, 2018, Vimentin on the move: new developments in cell migration. F1000Res 7. https:// doi.org/10.12688/f1000research.15967.1 Kidd ME, Shumaker DK, Ridge KM, 2014, The role of vimentin intermediate filaments in the progression of lung cancer. Am J Respir Cell Mol Biol 50(1):1–6. https://doi. org/10.1165/rcmb.2013-0314TR Xu X, Wang Y, Chen Z, Sternlicht MD, Hidalgo M, Steffensen B, 2005, Matrix metalloproteinase-2 contributes to cancer cell migration on collagen. Cancer Res 65(1):130–136 Srivastava VK, Gara RK, Rastogi N, Mishra DP, Ahmed MK, Gupta S, Goel MM, Bhatt ML, 2014, Serum vascular endothelial growth factor-A, VEGF-A, as a biomarker in squamous cell carcinoma of head and neck patients undergoing chemoradiotherapy. Asian Pac J Cancer Prev 15(7):3261–3265 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1301 EP 1311 DI 10.1007/s12253-019-00694-1 PG 11 ER PT J AU Foda, AMA Rafi, S Ikram, N Alam, SM Ayesha, S AF Foda, AlRahman Mohammad Abd Rafi, Samia Ikram, Nadeem Alam, Syed Mariya Ayesha, Sana TI Spinal Versus Intracranial Meningioma: Aberrant Expression of CD10 and Inhibin with Relation to Clinicopathological Features and Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD10; Inhibin; Spinal; Intracranial; Meningioma ID CD10; Inhibin; Spinal; Intracranial; Meningioma AB CD10 and inhibin are used mainly in CNS pathology to distinguish hemangioblastoma from metastatic clear cell renal cell carcinoma. Some meningiomas can mimic both tumors and so we aimed at this study to investigate the expression of both markers in a large number of meningioma cases. One hundred thirty-four meningioma samples were collected, 14 of them were spinal and 120 were intracranial. Manual TMA blocks were constructed using modified mechanical pencil tip method and immunohistochemistry for CD10 and inhibin was done. Intracranial meningioma occurred in significantly younger age than spinal ones. Most of spinal meningiomas were of transitional histology. CD10 was expressed in 14% of cases with significant positivity in spinal rather than intracranial cases. Transitional meningiomas showed the highest positivity for CD10 expression, while the least positive was the meningiotheliomatous type. Inhibin was expressed in 6% of cases with no significant relation to clinicopathological and histological features. There was no significant relationship between the expression of CD10 and inhibin expression in meningiomas. In conclusion, spinal meningiomas differ than intracranial ones in many clinicopathological and biological aspects. Among these differences is CD10 expression being more expressed in spinal meningiomas. However CD10 and inhibin are aberrantly expressed in a proportion of meningiomas, both have no relations to poor prognostic factors but more caution should be exerted during usage of these markers in diagnosis of hemangioblastoma and metastatic RCC. Further studies are suggested for exploring more biological differences between spinal and intracranial meningiomas. C1 [Foda, AlRahman Mohammad Abd] Mansoura University, Faculty of Medicine, Department of PathologyMansoura, Egypt. [Rafi, Samia] Batterjee Medical College for Sciences and TechnologyJeddah, Saudi Arabia. [Ikram, Nadeem] Batterjee Medical College for Sciences and TechnologyJeddah, Saudi Arabia. [Alam, Syed Mariya] Batterjee Medical College for Sciences and TechnologyJeddah, Saudi Arabia. [Ayesha, Sana] Tribhuvan University, Faculty of MedicineKirtipur, Nepal. RP Foda, AMA (reprint author), Mansoura University, Faculty of Medicine, Department of Pathology, Mansoura, Egypt. EM abdofoda@mans.edu.eg CR Riemenschneider MJ, Perry A, Reifenberger G, 2006, Histological classification and molecular genetics of meningiomas. Lancet Neurol 5(12):1045–1054 Langford LA, 1996, Pathology of meningiomas. J Neuro-Oncol 29:217–221. , DOI 10.1007/BF00165651 Backer-Grondahl T, Moen BH, Torp SH, 2012, The histopathological spectrum of human meningiomas. Int J Clin Exp Pathol 5(3): 231–242 Bi WL, Zhang M, Wu WW et al, 2016, Meningioma genomics: diagnostic, prognostic, and therapeutic applications. Front Surg 3: 40. , DOI 10.3389/fsurg.2016.00040 Harter P, Braun Y, Plate K, 2017, Classification of meningiomasadvances and controversies. Chinese Chin Clin Oncol 6(Suppl 1): S2. , DOI 10.21037/cco.2017.05.02 Domingues P, Gonzalez-Tablas M, Otero A et al, 2015, Genetic/ molecular alterations of meningiomas and the signaling pathways targeted. Oncotarget 6(13):10671–10688 Yuzawa S, Nishihara H, Tanaka S, 2016, Genetic landscape of meningioma. Brain Tumor Pathol 33(4):237–247 Han XY, Wang W, Wang LL, Wang XR, Li G, 2017, Genetic variants and increased risk of meningioma: an updated meta-analysis. OncoTargets Ther 10:1875–1888. , DOI 10.2147/ OTT.S130147 Sayagues JM, Tabernero MD, Maillo A, Trelles O, Espinosa AB, Sarasquete ME, Merino M, Rasillo A, Vera JF, Santos-Briz A, de Alava E, Garcia-Macias MC, Orfao A, 2006)Microarray-based analysis of spinal versus intracranial meningiomas: different clinical, biological, and genetic characteristics associated with distinct patterns of gene expression. J Neuropathol Exp Neurol 65(5):445–454 Yun JH, Roh JY, Park SH, Lee JR, 2013, CD10 expression in cutaneous squamous cell carcinoma and its precursor lesions: evaluation using tissue microarray. Ann Dermatol 25(4):515–517. , DOI 10.5021/ad.2013.25.4.515 Bahadir B, Behzatoglu K, Bektas S, Bozkurt ER, Ozdamar SO, 2009, CD10 expression in urothelial carcinoma of the bladder. Diagn Pathol 4:38. , DOI 10.1186/1746-1596-4-38 Bilalovic N, Sandstad B, Golouh R, Nesland JM, Selak I, Torlakovic EE, 2004, CD10 protein expression in tumor and stromal cells of malignant melanoma is associated with tumor progression. Mod Pathol 17(10):1251–1258 Jung SM, Kuo TT, 2005, Immunoreactivity of CD10 and inhibin alpha in differentiating hemangioblastoma of central nervous system from metastatic clear cell renal cell carcinoma. Mod Pathol 18(6):788–794 Makanji Y, Zhu J, Mishra R, Holmquist C, Wong WPS, Schwartz NB, Mayo KE, Woodruff TK, 2014, Inhibin at 90: from discovery to clinical application, a historical review. Endocr Rev 35(5):747– 794. , DOI 10.1210/er.2014-1003 Weissenbacher T, Bruning A, Kimmich T et al, 2010, Immunohistochemical labeling of the inhibin/activin βC subunit in normal human placental tissue and chorionic carcinoma cell lines. J Histochem Cytochem 58(8):751–757. , DOI 10. 1369/jhc.2010.956185 Foda AA, 2013, No-cost manual method for preparation of tissue microarrays having high quality comparable to semiautomated methods. Appl Immunohistochem Mol Morphol 21(3):271–274. , DOI 10.1097/PAI.0b013e318268a93f Boulagnon-Rombi C, Fleury C, Fichel C et al, 2017, Immunohistochemical approach to the differential diagnosis ofmeningiomas and their mimics. J Neuropathol ExpNeurol 76(4):289– 298. , DOI 10.1093/jnen/nlx008 Chu P, Arber AD, 2000, Paraffin-section detection of CD10 in 505 nonhematopoietic neoplasms: frequent expression in renal cell carcinoma and endometrial stromal sarcoma. Am J Clin Pathol 113(3): 374–382 Sanchez-Cespedes R, Suarez-Bonnet A, Millan Y, Guil-Luna S, Reymundo C, Herraez P, Espinosa de los Monteros A, Martin de las Mulas J, 2013, Use of CD10 as a marker of canine mammary myoepithelial cells. Vet J 195(2):192–199. , DOI 10.1016/ j.tvjl.2012.06.003 Sato Y, Itoh F, Hinoda Y, Ohe Y, Nakagawa N, Ueda R, Yachi A, Imai K, 1996, Expression of CD10/neutral endopeptidase in normal and malignant tissues of the human stomach and colon. J Gastroenterol 31(1):12–17. , DOI 10.1007/bf01211181 Ordi J, Romagosa C, Tavassoli FA, Nogales F, Palacin A, Condom E, Torne A, Cardesa A, 2003, CD10 expression in epithelial tissues and tumors of the gynecologic tract: a useful marker in the diagnosis of mesonephric, trophoblastic, and clear cell tumors. Am J Surg Pathol 27(2):178–186 Avery AK, Beckstead J, Renshaw AA, Corless CL, 2000, Use of antibodies to RCC and CD10 in the differential diagnosis of renal neoplasms. Am J Surg Pathol 24(2):203–210. , DOI 10. 1097/00000478-200002000-00006 Kristiansen G, Schluns K, Yongwei Y, Dietel M, Petersen I, 2002, CD10 expression in non-small cell lung cancer. Anal Cell Pathol 24(1):41–46 Prayson RA, Chamberlain WA, Angelov L, 2010, Clear cell meningioma: a Clinicopathologic study of 18 tumors and examination of the use of CD10, CA9, and RCC antibodies to distinguish between clear cell meningioma and metastatic clear cell renal cell carcinoma. Appl Immunohistochem Mol Morphol 18(5):422–428. , DOI 10.1097/PAI.0b013e3181dd35d2 Gurses I, Scheithauer BW, 2012, Inhibin-a immunoreactivity in nervous system lesions. Appl Immunohistochem Mol Morphol 20(3):277–284. , DOI 10.1097/PAI.0b013e31823e5ba8 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1313 EP 1318 DI 10.1007/s12253-019-00704-2 PG 6 ER PT J AU Li, T Li, H Xie, Sh Tan, Y Xie, ZP Li, WY Ai, F AF Li, Tao Li, Heng Xie, Sheng Tan, Yan Xie, Zi-Ping Li, Wen-Yi Ai, Fen TI Lactate Dehydrogenase-to-Lymphocyte Ratio Represents a Powerful Prognostic Tool of Metastatic Renal Cell Carcinoma Patients Treated with Tyrosine Kinase Inhibitors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE International metastatic renal cell carcinoma database consortium; Metastatic renal cell carcinoma; Lactate dehydrogenase to lymphocytes ratio; Tyrosine kinase inhibitors; Prognostic factors ID International metastatic renal cell carcinoma database consortium; Metastatic renal cell carcinoma; Lactate dehydrogenase to lymphocytes ratio; Tyrosine kinase inhibitors; Prognostic factors AB Inflammation parameters were verified to predict clinical outcomes of metastatic renal cell carcinoma (mRCC) patients treated with tyrosine kinase inhibitors (TKIs). Here, we developed a novel marker, lactate dehydrogenase (tumor burden marker) to lymphocytes (inflammation marker) ratio (LLR), aimed to reveal the prognostic role of LLR for mRCC patients treated with TKIs. We collected clinical data of mRCC patients treated with TKIs. Receiver operating curve analysis was used to determine the optimal cut-off value. The c-index method was used to determine the best predictive marker for overall survival (OS). Clinicopathological characteristics on OS and progression-free survival (PFS) were evaluated by univariate analysis, and multivariate analyses. LLR provided the greatest improvement in the c-index, and displayed the best marker of the prognostic accuracy for OS. Univariate analysis revealed that LLR, ECOG PS and IMDC risks were significant predictors of OS and PFS. However, multivariate analysis indicated that IMDC risks failed to predict PFS, and only showed predictor of OS. We finally stratified patients into low LLR (<150) and high LLR (≥150) group with different clinical outcomes. LLR represents a powerful prognostic tool of clinical outcome in mRCC patients treated with TKIs. C1 [Li, Tao] Hubei University of Medicine, Remin HospitalShiyan, Hubei Province, China. [Li, Heng] Hubei University of Medicine, Remin HospitalShiyan, Hubei Province, China. [Xie, Sheng] Hubei University of Medicine, Remin HospitalShiyan, Hubei Province, China. [Tan, Yan] Hubei University of Medicine, Remin HospitalShiyan, Hubei Province, China. [Xie, Zi-Ping] Hubei University of Medicine, Remin HospitalShiyan, Hubei Province, China. [Li, Wen-Yi] The First Affiliated Hospital of Kunming Medical CollegeKunming, Yunnan Province, China. [Ai, Fen] Huazhong University of Science and Technology, Tongji Medical College, The Central Hospital of Wuhan, Department of EmergencyWuhan, Hubei Province, China. RP Li, WY (reprint author), The First Affiliated Hospital of Kunming Medical College, Kunming, China. EM 942408619@qq.com CR Torre LA, Bray F, Siegel RL et al, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65:87–108 Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC, 1995, Results of treatment of 255 patients withmetastatic renal cell carcinoma who received high-doserecombinant interleukin-2 therapy. J Clin Oncol 13(3):688–696 Zahoor H, Barata PC, Jia X, Martin A, Allman KD, Wood LS, Gilligan TD, Grivas P, Ornstein MC, Garcia JA, Rini BI, 2018, Patterns, predictors and subsequent outcomes of disease progression in metastatic renal cell carcinoma patients treated with nivolumab. J Immunother Cancer 6(1):107 Lalani AA, XieW,Martini DJ et al, 2018, Change in neutrophil-tolymphocyte ratio, NLR, in response to immune checkpoint blockade formetastatic renal cell carcinoma. J Immunother Cancer 6(1):5 Heng DY, Xie W, Regan MM et al, 2009, Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol 27(34): 5794–5799 Chrom P, Zolnierek J, Bodnar L, Stec R, Szczylik C, 2019 in press, External validation of the systemic immune-inflammation index as a prognostic factor in metastatic renal cell carcinoma and its implementation within the international metastatic renal cell carcinoma database consortium model. Int J Clin Oncol 24:526–532 Yasuda Y, Saito K, Yuasa T, Uehara S, Kawamura N,Yokoyama M, Ishioka J, Matsuoka Y, Yamamoto S, Okuno T, Yonese J, Kihara K, Fujii Y, 2017, Early response of C-reactive protein as a predictor of survival in patients with metastatic renal cell carcinoma treated with tyrosine kinase inhibitors. Int J Clin Oncol 22(6):1081–1086 Hirofumi O, Keita U, Tatsuhiro K et al, 2017, Predictive value of themodified Glasgow prognostic score for the therapeutic effects of molecular-targeted drugs on advanced renal cell carcinoma. Mol Clin Oncol 6(5):669–675 Dirican A, Kucukzeybek Y, Somali I et al, 2013, The association of hematologic parameters on the prognosis of patients withmetastatic renal cell carcinoma. J BUON 18(2):413–419 Chrom P, Stec R, Semeniuk-Wojtas A, Bodnar L, Spencer NJ, Szczylik C, 2016, Fuhrman grade and neutrophil-to-lymphocyte ratio influence on survival in patients with metastatic renal cell carcinoma treated with first-line tyrosine kinase inhibitors. Clin Genitourin Cancer 14(5):457–464 Gu L, Ma X, Wang L et al, 2016, Prognostic value of a systemic inflammatory response index in metastatic renal cell carcinoma and construction of a predictivemodel. Oncotarget. 8(32):52094–52103 Fukuda H, Takagi T, Kondo T, Shimizu S, Tanabe K, 2018, Predictive value of inflammation-based prognostic scores in patients with metastatic renalcell carcinoma treated with cytoreductive nephrectomy. Oncotarget. 9(18):14296–14305 Huang AC, Postow MA, Orlowski RJ,Mick R, Bengsch B,Manne S, Xu W, Harmon S, Giles JR, Wenz B, Adamow M, Kuk D, Panageas KS, Carrera C, Wong P, Quagliarello F, Wubbenhorst B, D’Andrea K, Pauken KE, Herati RS, Staupe RP, Schenkel JM, McGettigan S, Kothari S, George SM, Vonderheide RH, Amaravadi RK, Karakousis GC, Schuchter LM, Xu X, Nathanson KL, Wolchok JD, Gangadhar TC, Wherry EJ, 2017, T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 545(7652):60–65 Escudier B, Porta C, Schmidinger M, Rioux-Leclercq N, Bex A, Khoo V, Gruenvald V, Horwich A, 2016, Renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow- up. Ann Oncol 27(suppl 5):v58–v68 Wang KF, Chang BY, Chen XQ, Liu PP, Wuxiao ZJ, Wang ZH, Li S, Jiang WQ, Xia ZJ, 2014, A prognostic model based on pretreatment platelet lymphocyte ratio for stage IE/IIE upper aerodigestive tract extranodal NK/T cell lymphoma, nasal type. Med Oncol 31(12):318 Proctor MJ, Horgan PG, Talwar D, Fletcher CD, Morrison DS, McMillan DC, 2013, Optimization of the systemic inflammationbased Glasgow prognostic score: a Glasgow inflammation outcome study. Cancer. 119:2325–2332 Zhang J, Yao YH, Li BG, Yang Q, Zhang PY, Wang HT, 2015, Prognostic value of pretreatment serum lactate dehydrogenase level in patients with solidtumors: a systematic review andmeta-analysis. Sci Rep 5:9800 Liu R, Cao J, Gao X, Zhang J, Wang L, Wang B, Guo L, Hu X, Wang Z, 2016, Overall survival of cancer patients with serum lactate dehydrogenase greater than 1000 IU/L. Tumour Biol 37(10): 14083–14088 Wang ZX, Yang LP, QiuMZ,Wang ZQ, Zhou YX,Wang F, Zhang DS, Wang FH, Li YH, Xu RH, 2016, Prognostic value of preoperative serum lactate dehydrogenase levels for resectable gastriccancer and prognostic nomograms. Oncotarget. 7(26): 39945–39956 Passardi A, Scarpi E, Tamberi S, Cavanna L, Tassinari D, Fontana A, Pini S, Bernardini I, Accettura C, Ulivi P, Frassineti GL, Amadori D, 2015, Impact of pre-treatment lactate dehydrogenase levels on prognosis and bevacizumab efficacy in patients with metastatic colorectal cancer. PLoS One 10(8):e0134732 Ji H, Niu X, Yin L,Wang Y, Huang L, Xuan Q, Li L, Zhang H, Li J, Yang Y, AnW, Zhang Q, 2018, Ratio of immune response to tumor burden predicts survival via regulating functions of lymphocytes and monocytes in diffuse large B-cell lymphoma. Cell Physiol Biochem 45(3):951–961 DaiW, Jia B, Yang J, Zhou S, Liu P, He X, Qin Y, Gui L, Zhang C, Han X, Sun Y, Shi Y, 2017, Development of new prognostic model based on pretreatment βLRI and LLRI for stage IE/IIE upper aerodigestive tract ENKTL, nasal type. Oncotarget. 8(21):34787– 34795 Semeniuk-Wojtas A, Lubas A, Stec R, Syrylo T, Niemczyk S, Szczylik C, 2018, Neutrophil-to-lymphocyte ratio, platelet-tolymphocyte ratio, and C-reactive protein as new and simple prognostic factors in patients with metastatic renal cell Cancer treated with tyrosine kinase inhibitors:a systemic review and meta-analysis. Clin Genitourin Cancer 16(3):e685–e693 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1319 EP 1324 DI 10.1007/s12253-019-00707-z PG 6 ER PT J AU Gallarin Salamanca, MI Espin Jaime, TM Moran-Penco, MJ Salas Martinez, J AF Gallarin Salamanca, Maria Isabel Espin Jaime, Teresa Maria Moran-Penco, Miguel Jose Salas Martinez, Jesus TI Role of Peritoneal Cytology in Patients with Early Stage Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Peritoneal lavage; Peritoneal cytology ID Colorectal cancer; Peritoneal lavage; Peritoneal cytology AB The main objective of this study was to investigate the incidence of free tumour cells in peritoneal lavage cytology performed using a standard method before and after the resection of the tumour, and to assess the usefulness of this technique in scheduled interventions on colorectal cancer patients. Peritoneal lavage cytology was performed on a homogeneous sample of 188 patients undergoing colorectal cancer curative resection before and after the resection of the tumour. The procedure was performed systematically in all cases. Malignant cells were detected in the peritoneal lavage cytology performed before the resection of the tumour in three patients. Lymph node affection was the variable most associated with the prognosis of these colorectal cancer patients. Peritoneal lavage cytology can provide additional information for a small group of patients who need to be closely monitored and studied to decide on the most effective type of chemotherapy. C1 [Gallarin Salamanca, Maria Isabel] Infanta Cristina Hospital, Department of Surgery, Carrillo Areanas 20A. Villafranca de los Barros, 06220 Badajoz, Spain. [Espin Jaime, Teresa Maria] Infanta Cristina Hospital, Department of Surgery, Carrillo Areanas 20A. Villafranca de los Barros, 06220 Badajoz, Spain. [Moran-Penco, Miguel Jose] University of Extremadura, Department of SurgeryBadajoz, Spain. [Salas Martinez, Jesus] Infanta Cristina Hospital, Department of Surgery, Carrillo Areanas 20A. Villafranca de los Barros, 06220 Badajoz, Spain. RP Gallarin Salamanca, MI (reprint author), Infanta Cristina Hospital, Department of Surgery, 06220 Badajoz, Spain. EM gallarinsa@gmail.com CR Ziselman EM, Harkavy SE, HoganM,WestW, Atkinson B, 1984, Peritoneal washing cytology. Uses and diagnostic criteria in gynecologic neoplasms. Acta Cytol 28:105–110 Lowe E, McKenna H, 1989, Peritoneal washing cytology: a retrospective analysis of 175 gynaecological patients. Aust N Z J Obstet Gynaecol 29:55–61 Miyashiro I, Takachi K, Doki Y, Ishikawa O, Ohigashi H, Murata K, Sasaki Y, Imaoka S, Nakaizumi A, Takenaka A, Furukawa H, HiratsukaM(2005)When is curative gastrectomy justified for gastric cancer with positive peritoneal lavage cytology but negative macroscopic peritoneal implant? World J Surg 29:1131–1134 Ishikawa O, Wada H, Ohigashi H, Doki Y, Yokoyama S, Noura S, Yamada T, Sasaki Y, Imaoka S, Kasugai T, Matsunaga T, Takenaka A, Nakaizumi A, 2003, Postoperative cytology for drained fluid from the pancreatic bed after “curative” resection of pancreatic cancers: does it predict both the patient’s prognosis and the site of cancer recurrence? Ann Surg 238:103–110 Doki Y, Kabuto T, Ishikawa O, Ohigashi H, Sasaki Y, Yamada T et al, 2001, Does pleural lavage cytology before thoracic closure predict both patient’s prognosis and site of cancer recurrence after resection of esophageal cancer? Surgery 130:792–797 Mohan HM, O'Connor DB, O'Riordan JM, Winter DC, 2013, Prognostic significance of detection of microscopic peritoneal disease in colorectal cancer: a systematic review. Surg Oncol 22:e1–e6 Bosanquet DC, Harris DA, Beynon J, 2013, Systematic review and meta-analysis of intraoperative peritoneal lavage for colorectal cancer staging. Br J Surg 100:853–862 Nishikawa T, Sunami E, Tanaka T, Tanaka J, Kiyomatsu T, Kawai K, 2015 Sep, el al. Incidence and prognostic significance of positive peritoneal lavage in colorectal cancer. Surg Today 45(9): 1073–1081 Altomare DF, Tedeschi M, Rotelli MT, Bocale D, Piscitelli D, RinaldiM, 2011, Lack of prognostic role of pre- and postoperative peritoneal cytology and cytokeratin PCR-expression on local recurrence after curative anterior resection for mid-low rectal cancer. Updat Surg 63:109–113 Noura S, Ohue M, Seki Y, Yano M, Ishikawa O, Kameyama M, 2009, Long-term prognostic value of conventional peritoneal lavage cytology in patients undergoing curative colorectal cancer resection. Dis Colon Rectum 52:1312–1320 Lemoine L, Sugarbaker P, Van der Speeten K, 2016, Pathophysiology of colorectal peritoneal carcinomatosis: role of the peritoneum. World J Gastroenterol 22(34):7692–7707 Temesi R, Sikorszki L, Bezsilla J, Botos A, Kovacs J, Tihanyi T, 2012, Impact of positive intraabdominal lavage cytology on the long-term prognosis of colorectal cancer patients.World J Surg 36: 2714e21 Lloyd JM, McIver CM, Stephenson SA, Hewett PJ, Rieger N, Hardingham JE, 2006, Identification of early-stage colorectal cancer patients at risk of relapse post-resection by immunobead reverse transcription-PCR analysis of peritoneal lavage fluid for malignant cells. Clin Cancer Res 12:417–423 Koppe MJ, Boerman OC, Oyen WJ, Bleichrodt RP, 2006, Peritoneal carcinomatosis of colorectal origin: incidence and current treatment strategies. Ann Surg 243:212–222 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1325 EP 1329 DI 10.1007/s12253-019-00706-0 PG 5 ER PT J AU Lei, Y Hu, Q Gu, J AF Lei, Yu Hu, Qiaoling Gu, Jiang TI Expressions of Carbohydrate Response Element Binding Protein and Glucose Transporters in Liver Cancer and Clinical Significance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ChREBP; Glucose transporters; Hepatocellular carcinoma; Diagnostic marker; Glycolysis ID ChREBP; Glucose transporters; Hepatocellular carcinoma; Diagnostic marker; Glycolysis AB Carbohydrate response element binding protein (ChREBP) is a glucose-sensing transcription factor that mediates the induction of glycolytic and lipogenic genes in response to glucose. We investigated the expression patterns of ChREBP and glucose transporters (GLUTs) in human hepatocellular carcinoma (HCC) and their association with HCC progression. ChREBP, GLUT2 and GLUT1 immunohistochemistry were performed on liver tissue array containing normal liver tissue, HCC adjacent tissue and cancer tissue of different HCC stages. The effect of HCC malignancy on protein expression was analyzed with one-way ANOVA. The correlations between protein expressions were analyzed with Pearson Correlation test. We found that ChREBP protein expression tended to be positively correlated to liver malignancy. GLUT2 protein expression was significantly reduced in human HCC as compared to normal liver tissue and its expression in HCC was inversely associated to malignancy (p < 0.001). In contrast, GLUT1 was significantly increased in cancer cells and its expression was positively correlated to malignancy (p < 0.001). Furthermore, GLUT1 expression was positively associated to ChREBP expression (r = 0.481, p < 0.0001, n = 70) but negatively correlated to GLUT2 expression (r = −0.320, p = 0.007, n = 70). Notably, ChREBP-expressing hepatocytes did not express GLUT2 but GLUT1. This is the first report unveiling expressions of ChREBP and GLUT2/GLUT1 and their relations in HCC. The expression patterns are related to malignancy and this information would facilitate evaluation of clinical behavior and treatment of HCC. C1 [Lei, Yu] Shantou University Medical College, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Collaborative and Creative Center, Department of Pathology and Pathophysiology, 515041 Shantou, Guangdong, China. [Hu, Qiaoling] Shantou University Medical College, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Collaborative and Creative Center, Department of Pathology and Pathophysiology, 515041 Shantou, Guangdong, China. [Gu, Jiang] Shantou University Medical College, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Collaborative and Creative Center, Department of Pathology and Pathophysiology, 515041 Shantou, Guangdong, China. RP Gu, J (reprint author), Shantou University Medical College, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Collaborative and Creative Center, Department of Pathology and Pathophysiology, 515041 Shantou, China. EM 2523381625@qq.com CR Trinchet JC, Chaffaut C, Bourcier V, Degos F, Henrion J, Fontaine H, Roulot D, Mallat A, Hillaire S, Cales P, Ollivier I, Vinel JP, Mathurin P, Bronowicki JP, Vilgrain V, N'Kontchou G, Beaugrand M, Chevret S, for the Groupe d'Etude et de Traitement du Carcinome Hepatocellula i r e, GRETCH,, 2011, Ultrasonographic surveillance of hepatocellular carcinoma in cirrhosis: a randomized trial comparing 3- and 6-month periodicities. Hepatology 54(6):1987–1997 Qin LX, Tang ZY, 2002, The prognostic significance of clinical and pathological features in hepatocellular carcinoma. World J Gastroenterol 8(2):193–199 Llovet JM, Chen Y,Wurmbach E, Roayaie S, FielMI, Schwartz M, Thung SN, Khitrov G, Zhang W, Villanueva A, Battiston C, Mazzaferro V, Bruix J, Waxman S, Friedman SL, 2006, A molecular signature to discriminate dysplastic nodules from early hepatocellular carcinoma in HCV cirrhosis. Gastroenterology 131(6): 1758–1767 Giovannucci E, Harlan DM, Archer MC, Bergenstal RM, Gapstur SM, Habel LA, Pollak M, Regensteiner JG, Yee D, 2010, Diabetes and cancer: a consensus report. Diabetes Care 33(7):1674–1685 Chang SC, Yang WV, 2016, Hyperglycemia, tumorigenesis, and chronic inflammation. Crit Rev Oncol Hematol 108:146–153 Xu CX, Zhu HH, Zhu YM, 2014, Diabetes and cancer: associations, mechanisms, and implications for medical practice. World J Diabetes 5(3):372–380 Kim JW, Dang CV, 2006, Cancer's molecular sweet tooth and the Warburg effect. Cancer Res 66(18):8927–8930 Tong X, Zhao F, Thompson CB, 2009, The molecular determinants of de novo nucleotide biosynthesis in cancer cells. Curr Opin Genet Dev 19(1):32–37 Christofk HR, Vander Heiden MG, Harris MH, Ramanathan A, Gerszten RE, Wei R, Fleming MD, Schreiber SL, Cantley LC, 2008, The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 452(7184):230– U74 Liu, Y.C., et al., Global Regulation of Nucleotide Biosynthetic Genes by c-Myc. PLoS One, 2008. 3(7) Zhu A, Lee D, Shim H, 2011, Metabolic positron emission tomography imaging in cancer detection and therapy response. Semin Oncol 38(1):55–69 MuecklerM, Thorens B, 2013, The SLC2, GLUT, family of membrane transporters. Mol Asp Med 34(2–3):121–138 Thorens B, Mueckler M, 2010, Glucose transporters in the 21st century. Am J Physiol Endocrinol Metab 298(2):E141–E145 Ganapathy V, Thangaraju M, Prasad PD, 2009, Nutrient transporters in cancer: relevance to Warburg hypothesis and beyond. Pharmacol Ther 121(1):29–40 Medina RA, Owen GI, 2002, Glucose transporters: expression, regulation and cancer. Biol Res 35(1):9–26 Endo M, Tateishi U, Seki K, Yamaguchi U, Nakatani F, Kawai A, Chuman H, Beppu Y, 2007, Prognostic implications of glucose transporter protein-1, Glut-1, overexpression in bone and softtissue sarcomas. Jpn J Clin Oncol 37(12):955–960 Semaan A, Munkarah AR, Arabi H, Bandyopadhyay S, Seward S, Kumar S, Qazi A, Hussein Y, Morris RT, Ali-Fehmi R, 2011, Expression of GLUT-1 in epithelial ovarian carcinoma: correlation with tumor cell proliferation, angiogenesis, survival and ability to predict optimal cytoreduction. Gynecol Oncol 121(1):181–186 Kim YH, Jeong DC, Pak K, Han ME, Kim JY, Liangwen L, Kim HJ, Kim TW, Kim TH, Hyun DW, Oh SO, 2017, SLC2A2, GLUT2, as a novel prognostic factor for hepatocellular carcinoma. Oncotarget 8(40):68381–68392 Noguchi Y, Marat D, Saito A, Yoshikawa T, Doi C, Fukuzawa K, Tsuburaya A, Satoh S, Ito T, 1999, Expression of facilitative glucose transporters in gastric tumors. Hepatogastroenterology 46(28): 2683–2689 Seino Yet al, 1993, Abnormal facilitative glucose transporter gene expression in human islet cell tumors. J Clin Endocrinol Metab 76(1):75–78 Yamashita H, Takenoshita M, Sakurai M, Bruick RK, Henzel WJ, Shillinglaw W, Arnot D, Uyeda K, 2001, A glucose-responsive transcription factor that regulates carbohydrate metabolism in the liver. Proc Natl Acad Sci U S A 98(16):9116–9121 Iizuka K, Bruick RK, Liang G, Horton JD, Uyeda K, 2004, Deficiency of carbohydrate response element-binding protein, ChREBP, reduces lipogenesis as well as glycolysis. Proc Natl Acad Sci U S A 101(19):7281–7286 Cairo S et al, 2001, WBSCR14, a gene mapping to the Williams– Beuren syndrome deleted region, is a new member of the mlx transcription factor network. Hum Mol Genet 10(6):617–627 Ishii S, IIzuka K, Miller BC, Uyeda K, 2004, Carbohydrate response element binding protein directly promotes lipogenic enzyme gene transcription. Proc Natl Acad Sci U S A 101(44):15597– 15602 Stoeckman AK, Ma L, Towle HC, 2004, Mlx is the functional heteromeric partner of the carbohydrate response element-binding protein in glucose regulation of lipogenic enzyme genes. J Biol Chem 279(15):15662–15669 Ma L, Robinson LN, Towle HC, 2006, ChREBP center dot mlx is the principal mediator of glucose-induced gene expression in the liver. J Biol Chem 281(39):28721–28730 Pedersen KB, Zhang P, Doumen C, Charbonnet M, Lu D, Newgard CB, Haycock JW, Lange AJ, Scott DK, 2007, The promoter for the gene encoding the catalytic subunit of rat glucose-6-phosphatase contains two distinct glucose-responsive regions. Am J Physiol Endocrinol Metab 292(3):E788–E801 Jeong YS, Kim D, Lee YS, Kim HJ, Han JY, Im SS, Chong HK, Kwon JK, Cho YH, Kim WK, Osborne TF, Horton JD, Jun HS, Ahn YH, Ahn SM, Cha JY, 2011, Integrated expression profiling and genome-wide analysis of ChREBP targets reveals the dual role for ChREBP in glucose-regulated gene expression. PLoS One 6(7): e22544 Tong X, Zhao F,Mancuso A, Gruber JJ, Thompson CB, 2009, The glucose-responsive transcription factor ChREBP contributes to glucose-dependent anabolic synthesis and cell proliferation. Proc Natl Acad Sci U S A 106(51):21660–21665 Airley RE, McHugh P, Evans AR, Harris B, Winchester L, Buffa FM, al-Tameemi W, Leek R, Harris AL, 2014, Role of carbohydrate response element-binding protein, ChREBP, in generating an aerobic metabolic phenotype and in breast cancer progression. Br J Cancer 110(3):715–723 Lei Y, Huang T, Su M, Luo J, Korteweg C, Li J, Chen Z, Qiu Y, Liu X, Yan M, Wang Y, Gu J, 2014, Expression and distribution of immunoglobulin G in the normal liver, hepatocarcinoma and postpartial hepatectomy liver. Lab Investig 94(11):1283–1295 Li J, Zhou Y, Gu J, 2014, Stain-decolorize-stain, SDS): a new technique for multiple staining. Histochem Cell Biol 141(3):251– 262 Evans A, Bates V, Troy H, Hewitt S, Holbeck S, Chung YL, Phillips R, Stubbs M, Griffiths J, Airley R, 2008, Glut-1 as a therapeutic target: increased chemoresistance and HIF-1-independent link with cell turnover is revealed through COMPARE analysis and metabolomic studies. Cancer Chemother Pharmacol 61(3):377– 393 Lunt SY, Vander Heiden MG, 2011, Aerobic Glycolysis: Meeting the Metabolic Requirements of Cell Proliferation. Annu Rev Cell Dev Biol 27(27):441–464 Heiden MGV, Cantley LC, Thompson CB, 2009, Understanding theWarburg effect: the metabolic requirements of cell proliferation. Science 324(5930):1029–1033 Yamamoto T, Seino Y, Fukumoto H, Koh G, Yano H, Inagaki N, Yamada Y, Inoue K,Manabe T, Imura H, 1990, Over-expression of facilitative glucose transporter genes in human cancer. Biochem Biophys Res Commun 170(1):223–230 Lu H, Forbes RA, Verma A, 2002, Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg effect in carcinogenesis. J Biol Chem 277(26):23111–23115 Warburg O, 1956, On the origin of cancer cells. Science 123(3191): 309–314 Higashi T, Tamaki N, Torizuka T, Nakamoto Y, Sakahara H, Kimura T, Honda T, Inokuma T, Katsushima S, Ohshio G, ImamuraM, Konishi J, 1998, FDG uptake, GLUT-1 glucose transporter and cellularity in human pancreatic tumors. J Nucl Med 39(10):1727–1735 Chung JK et al, 1999, Mechanisms related to [18F]fluorodeoxyglucose uptake of human colon cancers transplanted in nude mice. J Nucl Med 40(2):339–346 Rempel A, Bannasch P, Mayer D, 1994, Differences in expression and intracellular distribution of hexokinase isoenzymes in rat liver cells of different transformation stages. Biochim Biophys Acta 1219(3):660–668 Mathupala SP, Rempel A, Pedersen PL, 1995, Glucose catabolism in cancer cells. Isolation, sequence, and activity of the promoter for type II hexokinase. J Biol Chem 270(28):16918–16925 Younes M et al, 1995, GLUT1 expression in human breast carcinoma: correlation with known prognostic markers. Anticancer Res 15(6B):2895–2898 Chandler JD, Williams ED, Slavin JL, Best JD, Rogers S, 2003, Expression and localization of GLUT1 and GLUT12 in prostate carcinoma. Cancer 97(8):2035–2042 Kawamura T, Kusakabe T, Sugino T, Watanabe K, Fukuda T, Nashimoto A, Honma K, Suzuki T, 2001, Expression of glucose transporter-1 in human gastric carcinoma: association with tumor aggressiveness, metastasis, and patient survival. Cancer 92(3):634– 641 Pak K, Cheon GJ, Nam HY, Kim SJ, Kang KW, Chung JK, Kim EE, Lee DS, 2014, Prognostic value of metabolic tumor volume and total lesion glycolysis in head and neck cancer: a systematic review and meta-analysis. J Nucl Med 55(6):884–890 Mardones L, Ormazabal V, Romo X, Jana C, Binder P, Pena E, Vergara M, Zuniga FA, 2011, The glucose transporter-2, GLUT2, is a low affinity dehydroascorbic acid transporter. Biochem Biophys Res Commun 410(1):7–12 Wisniewski JR et al, 2016, In-depth quantitative analysis and comparison of the human hepatocyte and hepatoma cell line HepG2 proteomes. J Proteome 136:234–247 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1331 EP 1340 DI 10.1007/s12253-019-00708-y PG 10 ER PT J AU Chauhan, Sh Sen, S Singh, N Sharma, A Chawla, B Kashyap, S AF Chauhan, Sheetal Sen, Seema Singh, Neeta Sharma, Anjana Chawla, Bhavna Kashyap, Seema TI Human Papillomavirus Detection Strategies in Retinoblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Chauhan, Sheetal] All India Institute of Medical Sciences, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, Department of Ocular Pathology, Room no. 725, 110029 New Delhi, India. [Sen, Seema] All India Institute of Medical Sciences, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, Department of Ocular Pathology, Room no. 725, 110029 New Delhi, India. [Singh, Neeta] All India Institute of Medical Sciences, Department of Biochemistry, 110029 New Delhi, India. [Sharma, Anjana] All India Institute of Medical Sciences, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, Department of Ocular Microbiology, 110029 New Delhi, India. [Chawla, Bhavna] All India Institute of Medical Sciences, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, Ocular Oncology Service, 110029 New Delhi, India. [Kashyap, Seema] All India Institute of Medical Sciences, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, Department of Ocular Pathology, Room no. 725, 110029 New Delhi, India. RP Sen, S (reprint author), All India Institute of Medical Sciences, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, Department of Ocular Pathology, 110029 New Delhi, India. EM ssenop@rediffmail.com CR OrjuelaM, Castaneda VP, Ridaura C, Lecona E, Leal C, Abramson DH, Orlow I, Gerald W, Cordon-Cardo C, 2000, Presence of human papilloma virus in tumor tissue from children with retinoblastoma: an alternative mechanism for tumor development. Clin Cancer Res 6:4010–4016 Montoya-Fuentes H, de la Paz Ramirez-Munoz M, Villar-Calvo V et al, 2003, Identification of DNA sequences and viral proteins of 6 human papillomavirus types in retinoblastoma tissue. Anticancer Res 23:2853–2862 Bhuvaneswari A, Pallavi VR, Jayshree RS, Kumar RV, 2012, Maternal transmission of human papillomavirus in retinoblastoma: a possible route of transfer. Indian J Med Paediatr Oncol 33(4):210–215 Palazzi MA, Yunes JA, Cardinalli IA, Stangenhaus GP, Brandalise SR, Ferreira SA, Sobrinho JSP, Villa LL, 2003, Detection of oncogenic human papillomavirus in sporadic retinoblastoma. Acta Ophthalmol Scand 81:396–398 Antoneli CB, Ribeiro KB, Sredni STet al, 2011, Low prevalence of HPV in Brazilian children with retinoblastoma. J Med Virol 83: 115–118 Gillison ML, Chen R, Goshu E, Rushlow D, Chen N, Banister C, Creek KE, Gallie BL, 2007, Human retinoblastoma is not caused by known pRb-inactivating human DNA tumor viruses. Int J Cancer 120:1482–1490 Shukla S, Bharti AC, Mahata S, Hussain S, Kumar R, Hedau S, Das BC, 2009, Infection of human papillomaviruses in cancers of different human organ sites. Indian J Med Res 130:222–233 Mohan A, Venkatesan N, Kandalam M, Pasricha G, Acharya P, Khetan V, Gopal L, Sharma T, Biswas J, Krishnakumar S, 2009, Detection of human papillomavirus DNA in retinoblastoma samples: a preliminary study. J Pediatr Hematol Oncol 31:8–13 Anand B, Ramesh C, Appaji L et al, 2011, Prevalence of high-risk human papillomavirus genotypes in retinoblastoma. Br J Ophthalmol 95:014–1018 Shetty OA, Naresh KN, Banavali SD, Shet T, Joshi R, Qureshi S, Mulherkar R, Borges A, Desai SB, 2012, Evidence for the presence of high risk human papillomavirus in retinoblastoma tissue from nonfamilial retinoblastoma in developing countries. Pediatr Blood Cancer 58:185–190 Abreu ALP, Souza RP, Gimenes F, Consolaro MEL, 2012, A review of methods for detect human papillomavirus infection. Virol J 9:262 Lee SJ, LeeAW, Kang CS, Park JS, ParkDC, Ki EY, Lee KH, Yoon JH, Hur SY, Kim TJ, 2014, Clinicopathological implications of human papilloma virus, HPV, L1 capsid protein immunoreactivity in HPV16-positive cervical cytology. International Journal of Medical Sciences: Int J Med Sci 11:80–86 Dabic MM, Hlupic L, Babic D, Jukic S, Seiwerth S, 2004, Comparison of polymerase chain reaction and catalyzed signal amplification in situ hybridization methods for human papillomavirus detection in paraffin-embedded cervical preneoplastic and neoplastic lesions. Arch Med Res 35:511–516 Giuliani L, Coletti A, Syrjanen K, Favalli C, Ciotti M, 2006, Comparison of DNA sequencing and Roche linear array in human papillomavirus, HPV, genotyping. Anticancer Res 26:3939–3941 Ryoo NK, KimJE, Choung HK, KimN, Lee MJ,Khwarg SI, 2013, Human papilloma virus in retinoblastoma tissues from Korean patients. Korean J Ophthalmol 27:368–371 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1341 EP 1344 DI 10.1007/s12253-018-00577-x PG 4 ER PT J AU Mo, YH Jeon, HE Kim, SM Yoo, JN Lee, HS AF Mo, Yoon Ha Jeon, Ha Eun Kim, Sung Min Yoo, Jin Nam Lee, Hyung Sug TI Analysis of Promoter Mutation in Long Non-coding RNA NEAT1 in Acute Leukemias SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Mo, Yoon Ha] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Jeon, Ha Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Kim, Sung Min] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Huarte M, 2015, The emerging role of lncRNAs in cancer. Nat Med 21:1253–1261 Fang J, Qiao F, Tu J, Xu J, Ding F, Liu Y, Akuo BA, Hu J, Shao S, 2017, High expression of long non-coding RNA NEAT1 indicates poor prognosis of human cancer. Oncotarget 8:45918–45927 Rheinbay E, Parasuraman P, Grimsby J, Tiao G, Engreitz JM, Kim J, Lawrence MS, Taylor-Weiner A, Rodriguez-Cuevas S, Rosenberg M, Hess J, Stewart C, Maruvka YE, Stojanov P, Cortes ML, Seepo S, Cibulskis C, Tracy A, Pugh TJ, Lee J, Zheng Z, Ellisen LW, Iafrate AJ, Boehm JS, Gabriel SB, Meyerson M, Golub TR, Baselga J, Hidalgo-Miranda A, Shioda T, Bernards A, Lander ES, Getz G, 2017, Recurrent and functional regulatory mutations in breast cancer. Nature 547:55–60 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1345 EP 1346 DI 10.1007/s12253-019-00608-1 PG 2 ER PT J AU Tsiambas, E Mastronikolis, SN Fotiades, PP Ragos, V Kavantzas, N Lazaris, CA AF Tsiambas, Evangelos Mastronikolis, S Nicholas Fotiades, P Panagiotis Ragos, Vasileios Kavantzas, Nikolaos Lazaris, C Andreas TI Epstein-Barr Virus MicroRNAs in Nasopharyngeal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Tsiambas, Evangelos] 401 General Army Hospital, Department of Pathology-CytologyAthens, Greece. [Mastronikolis, S Nicholas] 401 General Army Hospital, Department of Pathology-CytologyAthens, Greece. [Fotiades, P Panagiotis] 424 General Army Hospital, Department of SurgeryThessaloniki, Greece. [Ragos, Vasileios] University of Ioannina, School of Health Sciences, Department of Maxillofacial and Department of MedicineIoannina, Greece. [Kavantzas, Nikolaos] University of Athens, Medical School, Department of PathologyAthens, Greece. [Lazaris, C Andreas] University of Athens, Medical School, Department of PathologyAthens, Greece. RP Tsiambas, E (reprint author), 401 General Army Hospital, Department of Pathology-Cytology, Athens, Greece. EM tsiambasecyto@yahoo.gr CR Lo KW, Chung GT, To KF, 2012, Deciphering themolecular genetic basis of NPC through molecular, cytogenetic, and epigenetic approaches. Semin Cancer Biol 22:79–86 Hiromu S, Maruyama R, Yamamoto E, Masahiro K, 2012, DNA methylation and microRNA dysregulation in cancer. Mol Oncol 6: 567–578 Zhao L, Fong AHW, Liu N, Cho WCS, 2018, Molecular subtyping of nasopharyngeal carcinoma, NPC, and a microRNA-based prognostic model for distant metastasis. J Biomed Sci 25(1):16–28 Wu Q, Han T, Sheng X, ZhangN,Wang P, 2018)Downregulation of EB virus miR-BART4 inhibits proliferation and aggressiveness while promoting radiosensitivity of nasopharyngeal carcinoma. Biomed Pharmacother 108:741–751 Lung RW, Hau PM, Yu KH et al, 2018, EBV-encoded miRNAs target ATM-mediated response in nasopharyngeal carcinoma. J Pathol 244(4):394–407 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1347 EP 1348 DI 10.1007/s12253-019-00631-2 PG 2 ER PT J AU Pinczes, L Magyari, F Remenyi, Gy Pfliegler, Gy Barna, S Bedekovics, J Illes, AF Pinczes, Laszlo Magyari, Ferenc Remenyi, Gyula Pfliegler, Gyorgy Barna, Sandor Bedekovics, Judit Illes, Arpad TI Intravascular Occlusion by Leukemic Blast Cells Causing Multiplex Hand Necrosis in a Patient with Acute Myeloid Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Pinczes, Laszlo] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Magyari, Ferenc] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Remenyi, Gyula] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Pfliegler, Gyorgy] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Barna, Sandor] Scanomed Kft.Debrecen, Hungary. [Bedekovics, Judit] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. RP Pinczes, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM pinczes.laszlo.imre@med.unideb.hu CR Yamamoto Y, Yamamoto S, 2017, Achenbach’s Syndrome. N Engl J Med 376:e53. , DOI 10.1056/NEJMicm1610146 Guzman-Uribe P, Vargas-Ruiz AG, 2015, Thrombosis in leukemia: incidence, causes, and practical management. Curr Oncol Rep 17:19. , DOI 10.1007/s11912-015-0444-2 Bakst RL, Tallman MS, Douer D, Yahalom J, 2011, How I treat extramedullary acute myeloid leukemia. Blood 118:3785–3793. , DOI 10.1182/blood-2011-04-347229 Porcu P, Cripe LD, Ng EW, Bhatia S, Danielson CM, Orazi A, McCarthy LJ, 2000, Hyperleukocytic Leukemias and Leukostasis: a review of pathophysiology, clinical presentation and management. Leuk Lymphoma 39:1–18. , DOI 10.3109/ 10428190009053534 Thepot S, Itzykson R, Seegers V, Recher C, Raffoux E, Quesnel B, Delaunay J, Cluzeau T, Marfaing Koka A, Stamatoullas A, Chaury MP, Dartigeas C, Cheze S, Banos A, Morel P, Plantier I, Taksin AL, Marolleau JP, Pautas C, ThomasX, Isnard F, Beve B, Chait Y, Guerci A, Vey N, Dreyfus F, Ades L, Ifrah N, Dombret H, Fenaux P, Gardin C, On behalf of the Groupe Francophone des Myelodysplasies, GFM), the Acute Leukemia French Association, ALFA); the Groupe Ouest-Est des Leucemies Aigues; Maladies du Sang, GOELAMS,, 2014, Azacitidine in untreated acute myeloid leukemia: a report on 149 patients. Am J Hematol 89:410–416. , DOI 10.1002/ajh.23654 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1349 EP 1351 DI 10.1007/s12253-019-00636-x PG 3 ER PT J AU Kumar, SS Sreedharan, NS Patil, Sh Raj, Th AF Kumar, Satish Sunil Sreedharan, Nellimad Sreena Patil, Shankargouda Raj, A. Thirumal TI The Potential Role of Organoids in Pathology and Oncology Research SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Kumar, Satish Sunil] Sai Srinivas Dental Care, SithalapakkamChennai, India. [Sreedharan, Nellimad Sreena] Corning Dental AssociatesCorning, NY, USA. [Patil, Shankargouda] Jazan University, College of Dentistry, Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral PathologyJazan, Saudi Arabia. [Raj, A. Thirumal] Sri Venkateswara Dental College and Hospital, Department of Oral Pathology and Microbiology, Thalambur, 600130 Chennai, India. RP Raj, Th (reprint author), Sri Venkateswara Dental College and Hospital, Department of Oral Pathology and Microbiology, 600130 Chennai, India. EM thirumalraj666@gmail.com CR LancasterMA, Knoblich JA, 2014, Organogenesis in a dish: modeling development and disease using organoid technologies. Science 345(6194):1247125. , DOI 10.1126/science.1247125 HuchM, Koo B-K, 2015)Modelingmouse and human development using organoid cultures. Development 142(18):3113–3125. https:// doi.org/10.1242/dev.118570 Duong HQ, Nemazanyy I, Rambow F, Tang SC, Delaunay S, Tharun L, Florin A, Buttner R, Vandaele D, Close P, Marine JC, Shostak K, ChariotA(2018, The endosomal protein CEMIP linksWnt signaling to MEK1-ERK1/2 activation in Selumetinib-resistant intestinal organoids. Cancer Res 78:4533–4548. , DOI 10.1158/ 0008-5472.CAN-17-3149 Ullah I, Subbarao RB, Rho GJ, 2015, Human mesenchymal stem cells - current trends and future prospective. Biosci Rep 35(2): e00191. , DOI 10.1042/BSR20150025 Jabs J, Zickgraf FM, Park J, Wagner S, Jiang X, Jechow K, Kleinheinz K, Toprak UH, Schneider MA, Meister M, Spaich S, Sutterlin M, Schlesner M, Trumpp A, Sprick M, Eils R, Conrad C, 2017, Screening drug effects in patient-derived cancer cells link organoid responses to genome alterations. Mol Syst Biol 13:955. , DOI 10.15252/msb.20177697 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1353 EP 1354 DI 10.1007/s12253-019-00642-z PG 2 ER PT J AU Mormile, R AF Mormile, Raffaella TI Immune Checkpoint Inhibitors in Small Cell Lung Cancer: Is It Also a Matter of Helios− Cells? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Mormile, Raffaella] Moscati Hospital, Division of Pediatrics and Neonatology, Via A. Gramsci, 81031 Aversa, Italy. RP Mormile, R (reprint author), Moscati Hospital, Division of Pediatrics and Neonatology, 81031 Aversa, Italy. EM raffaellamormile@alice.it CR Ready N, Farago AF, de Braud F, Atmaca A, Hellmann MD, Schneider JG, Spigel DR, Moreno V, Chau I, Hann CL, Eder JP, Steele NL, Pieters A, Fairchild J, Antonia SJ, 2018, Third-line nivolumab monotherapy in recurrent sclc: check mate 032. J Thorac Oncol. 2018 Oct 10. pii: S1556–0864(18)33181–2.0998079 Schmid S, Fruh M, 2018, Immune checkpoint inhibitors and small cell lung cancer: what's new? Thorac Dis 2018 May;10(Suppl 13): S1503–S1508 Wang W, Hodkinson P, McLaren F, MacKinnon A, Wallace W, Howie S, Sethi T, 2012 Sep 15, Small cell lung cancer tumour cells induce regulatory T lymphocytes, and patient survival correlates negatively with FOXP3+ cells in tumour infiltrate. Int J Cancer 131(6):E928–E937 Himmel ME1, MacDonald KG, Garcia RV, Steiner TS, Levings MK. Helios+ and Helios- cells coexist within the natural FOXP3+ T regulatory cell subset in humans. J Immunol 2013 Mar 1;190(5):2001–2008 Zhang X, Zeng Y, Qu Q, Zhu J, Liu Z, Ning W, Zeng H, Zhang N, DuW, Chen C, Huang JA, 2017 Dec, PD-L1 induced by IFN-γ from tumor-associated macrophages via the JAK/STAT3 and PI3K/AKT signaling pathways promoted progression of lung cancer. Int J Clin Oncol 22(6):1026–1033 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1355 EP 1356 DI 10.1007/s12253-019-00653-w PG 2 ER PT J AU Mormile, R AF Mormile, Raffaella TI Immune Checkpoint Inhibitor Therapy in HIV-Positive Patients with Advanced-Stage Cancer: a Golden Card to Be Played? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Mormile, Raffaella] Moscati Hospital, Division of Pediatrics and Neonatology, Via A. Gramsci, 81031 Aversa, Italy. RP Mormile, R (reprint author), Moscati Hospital, Division of Pediatrics and Neonatology, 81031 Aversa, Italy. EM raffaellamormile@alice.it CR Cook MR, Kim C, 2019, Safety and efficacy of immune checkpoint inhibitor therapy in patients with HIVinfection and advancedstage Cancer: a systematic review. JAMA Oncol. , DOI 10. 1001/jamaoncol.2018.6737 Ostios-Garcia L, Faig J, Leonardi GC, Adeni AE, Subegdjo SJ et al, 2018 Jul, Safety and efficacy of PD-1 inhibitors among HIVpositive patients with non-small cell lung Cancer. J Thorac Oncol 13(7):1037–1042 Seidel JA, Otsuka A, Kabashima K, 2018, Anti-PD-1 and anti-CTLA- 4 therapies in Cancer: mechanisms of action, efficacy, and limitations. Front Oncol 8:86. , DOI 10.3389/fonc.2018.00086 Jeon HS, Choi YY, Choi JE, LeeWK, Lee E, Yoo SS, Lee SY, Lee J, Cha SI, Kim CH, Park JY, 2014, Telomere length of tumor tissues and survival in patients with early stage non-small cell lung cancer. Mol Carcinog 53(4):272–279 Rachakonda S, Srinivas N, Mahmoudpour SH, Garcia-Casado Z, Requena C, TravesV, SorianoV, CardelliM, Pjanova D,Molven A, Gruis N, Nagore E, Kumar R, 2018, Telomere length and survival in primary cutaneous melanoma patients. Sci Rep 8(1):10947 Unemori P, Leslie KS, HuntPW, Sinclair E, Epling L,Mitsuyasu R, Effros RB, Dock J, Dollard SG, Deeks SG, Martin JN, Maurer TA, 2013, Immunosenescence is associated with presence of Kaposi's sarcoma in antiretroviral treated HIV infection. AIDS. 27(11):1735 Liu JCY, Leung JM, Ngan DA, Nashta NF, Guillemi S, HarrisMet al, 2015, Absolute leukocyte telomere length in HIV-infected and uninfected individuals: evidence of accelerated cell senescence in HIVassociated chronic obstructive pulmonary disease. PLoS One 10(4): e0124426. , DOI 10.1371/journal.pone.0124426 Lichterfeld M,Mou D, Cung TD,WilliamsKL,WaringMT, Huang J, Pereyra F, Trocha A, Freeman GJ, Rosenberg ES,Walker BD, Yu XG, 2008, Telomerase activity of HIV-1-specific CD8+ T cells: constitutive up-regulation in controllers and selective increase by blockade of PD ligand 1 in progressors. Blood. 112(9):3679–3687 ZhangW, Shi L, Zhao Z, Du P, Ye X, Li D, Cai Z, Han J, Cai J, 2019, Disruption of CTLA-4 expression on peripheral blood CD8 + T cell enhances anti-tumor efficacy in bladder cancer. Cancer Chemother Pharmacol. , DOI 10.1007/s00280-019-03800-x Aamdal E, Gaudernack G, Inderberg EM, Rasch W, Bjorheim J, Aamdal S, Guren TK, 2017, Telomerase peptide vaccine combined with ipilimumab in metastatic melanoma: reports from a phase I trial. Ann Oncol Volume 28(Issue suppl_5):1 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1357 EP 1358 DI 10.1007/s12253-019-00686-1 PG 2 ER PT J AU Mormile, R AF Mormile, Raffaella TI Body Fat and Risk of Breast Cancer in Postmenopausal Women: What Is the Truth? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Mormile, Raffaella] Moscati Hospital, Division of Pediatrics and Neonatology, Via A. Gramsci, 81031 Aversa, Italy. RP Mormile, R (reprint author), Moscati Hospital, Division of Pediatrics and Neonatology, 81031 Aversa, Italy. EM raffaellamormile@alice.it CR Iyengar NM, Arthur R, Manson JE, Chlebowski RT, Kroenke CH, Peterson L, Cheng TD, Feliciano EC, Lane D, Luo J, Nassir R, Pan K, Wassertheil-Smoller S, Kamensky V, Rohan TE, Dannenberg AJ, 2018, Association of body fat and risk of breast cancer in postmenopausal women with normal body mass index: a secondary analysis of a randomized clinical trial and observational study. JAMA Oncol 5:155. , DOI 10.1001/jamaoncol.2018.5327 Gomes JL, Fernandes T, Soci UP, Silveira AC, Barretti DL, Negrao CE, Oliveira EM, 2017, Obesity downregulates MicroRNA-126 inducing capillary rarefaction in skeletal muscle: effects of aerobic exercise training. Oxidative Med Cell Longev 2017:2415246 Wang CZ, Yuan P, Li Y, 2015 Jun 1, MiR-126 regulated breast cancer cell invasion by targeting ADAM9. Int J Clin Exp Pathol 8(6):6547–6553 Li P,Wei J, Li X, Cheng Y, ChenW, Cui Y, Simoncini T, Gu Z, Yang J, Fu X, 2017, 17β-estradiol enhances vascular endothelial Ets-1/ miR-126-3p expression: the possible mechanism for attenuation of atherosclerosis. J Clin Endocrinol Metab 102(2):594–603 Furlan A, Vercamer C, Heliot L, Wernert N, Desbiens X, Pourtier A, 2019, Ets-1 drives breast cancer cell angiogenic potential and interactions between breast cancer and endothelial cells. Int J Oncol 54(1):29–40 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1359 EP 1360 DI 10.1007/s12253-019-00691-4 PG 2 ER PT J AU Mormile, R AF Mormile, Raffaella TI Total Serum Cholesterol and Pancreatic Cancer Risk: What Is the Link? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Mormile, Raffaella] Moscati Hospital, Division of Pediatrics and Neonatology, Via A. Gramsci, 81031 Aversa, Italy. RP Mormile, R (reprint author), Moscati Hospital, Division of Pediatrics and Neonatology, 81031 Aversa, Italy. EM raffaellamormile@alice.it CR Chen WC, Boursi B, Mamtani R, Yang YX, 2018, Total serum cholesterol and pancreatic cancer: a nested case-control study. Cancer Epidemiol Biomark Prev 28(2):363-369 Zhou P, Li B, Liu B, Chen T, Xiao J, 2018, Prognostic role of serum total cholesterol and high-density lipoprotein cholesterol in cancer survivors: a systematic review and meta-analysis. Clin Chim Acta 477:94–104 Razidlo GL, Burton KM, McNiven MA, 2018, Interleukin-6 promotes pancreatic cancer cell migration by rapidly activating the small GTPase CDC42. J Biol Chem 293(28):11143–11153 Holmer R, Goumas FA,Waetzig GH, Rose-John S, Kalthoff H, 2014, Interleukin-6: a villain in the drama of pancreatic cancer development and progression. Hepatobiliary Pancreat Dis Int 13(4):371–380 Hashizume M, Mihara M, 2011, IL-6 and lipid metabolism. Inflammation and Regeneration 31:325–333 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1361 EP 1361 DI 10.1007/s12253-019-00698-x PG 1 ER PT J AU Mormile, R AF Mormile, Raffaella TI Aspirin Use and Risk of Glioma: a Double Track for a Single Goal? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Mormile, Raffaella] Moscati Hospital, Division of Pediatrics and Neonatology, Via A. Gramsci, 81031 Aversa, Italy. RP Mormile, R (reprint author), Moscati Hospital, Division of Pediatrics and Neonatology, 81031 Aversa, Italy. EM raffaellamormile@alice.it CR Gusyatiner O, Hegi ME, 2018 Aug, Glioma epigenetics: from subclassification to novel treatment options. Semin Cancer Biol 51:50– 58 Amirian ES, Ostrom QT, Armstrong GN, Lai RK, Gu X, Jacobs DI, Jalali A, Claus EB, Barnholtz-Sloan JS, Il'yasova D, Schildkraut JM, Ali-Osman F, Sadetzki S, Jenkins RB, LaChance DH, Olson SH, Bernstein JL, Merrell RT, Wrensch MR, Johansen C, Houlston RS, ScheurerME, Shete S,Amos CI, Melin B, BondyML, 2018 Nov 27. pii: cebp.0702.2018, Aspirin, non-steroidal anti-inflammatory drugs, NSAIDs), and glioma risk: original data from the glioma international case-control study and a meta-analysis. Cancer Epidemiol Biomark Prev. , DOI 10.1158/1055-9965.EPI-18-0702 Fujita M, Kohanbash G, Fellows-MayleW,Hamilton RL, Komohara Y, Decker SA, Ohlfest JR, Okada H, 2011 Apr 1, COX-2 blockade suppresses gliomagenesis by inhibiting myeloid-derived suppressor cells. Cancer Res 71(7):2664–2674 Zhang S, Zhang C, Song Y, Zhang J, Xu J, 2018, Prognostic role of survivin in patients with glioma.Medicine, Baltimore, 97(17):e0571 Zhang F, Chu J,Wang F, 2017, Expression and clinical significance of cyclooxygenase 2 and survivin in human gliomas. Oncol Lett 14(2):1303–1308 Nemeth K, Szucs N, Czirjak S, Reiniger L, Szabo B, Barna G, Karaszi K, Igaz P, Zivkovic V, Korbonits M, Patocs A, Butz H, 2018, Survivin as a potential therapeutic target of acetylsalicylic acid in pituitary adenomas. Oncotarget. 9(49):29180–29192 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1363 EP 1364 DI 10.1007/s12253-019-00699-w PG 2 ER PT J AU Mormile, R AF Mormile, Raffaella TI Statin Therapy and Survival among Women with Ovarian Cancer: how much of it Is True? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Mormile, Raffaella] Moscati Hospital, Division of Pediatrics and Neonatology, Via A. Gramsci, 81031 Aversa, Italy. RP Mormile, R (reprint author), Moscati Hospital, Division of Pediatrics and Neonatology, 81031 Aversa, Italy. EM raffaellamormile@alice.it CR Harding BN, Delaney JA, Urban RR, Weiss NS, 2019, Use of statinmedications following diagnosis in relation to survival among women with ovarian cancer. Cancer Epidemiol Biomark Prev 28: 1127–1133 Couttenier A, Lacroix O, Vaes E, Cardwell CR, De Schutter H, Robert A, 2017, Statin use is associated with improved survival in ovarian cancer: a retrospective population-based study. PLoS One 12(12):e0189233 Loppnow H, Zhang L, Buerke M, Lautenschlager M, Chen L, Frister A, Schlitt A, Luther T, Song N, Hofmann B, Rose-John S, Silber RE, Muller-Werdan U, Werdan K, 2011, Statins potently reduce the cytokine-mediated IL-6 release in SMC/MNC co cultures. J Cell Mol Med 15(4):994–1004 Hansen M, Kuhlman ACB, Sahl RE, Kelly B, Morville T, Dohlmann TL, Chrois KM, Larsen S, Helge JW, Dela F, 2019, Inflammatory biomarkers in patients in Simvastatin treatment: No effect of co-enzyme Q10 supplementation. Cytokine 113:393–399. , DOI 10.1016/j.cyto.2018.10.011 Davaro F, Forde SD, Garfield M, Jiang Z, Halmen K, Tamburro ND, Kurt-Jones E, Fitzgerald KA, Golenbock DT, Wang D, 2014, 3-Hydroxyl-3-methylglutaryl coenzyme a, HMG-CoA, reductase inhibitor, statin)-induced 28-kDa interleukin-1β interfereswithmature IL-1β signaling. J Biol Chem 289(23):16214–16222 Cahill CM, Rogers JT, 2008, Interleukin, IL, 1beta induction of IL- 6 is mediated by a novel phosphatidylinositol 3-kinase-dependent AKT/IkappaB kinase alpha pathway targeting activator protein-1. J Biol Chem 283(38):25900–25912 Schauer IG, Zhang J, Xing Z, Guo X, Mercado-Uribe I, Sood AK, Huang P, Liu J, 2013, Interleukin-1β promotes ovarian tumorigenesis through a p53/NF-κB-mediated inflammatory response in stromal fibroblasts. Neoplasia 15(4):409–420 Browning L, Patel MR, Horvath EB, Tawara K, Jorcyk CL, 2018, IL-6 and ovarian cancer: inflammatory cytokines in promotion of metastasis. Cancer Manag Res 10:6685–6693 Yousefi H, Momeny M, Ghaffari SH, Parsanejad N, Poursheikhani A, Javadikooshesh S, Zarrinrad G, Esmaeili F, Alishahi Z, Sabourinejad Z, Sankanian G, Shamsaiegahkani S, Bashash D, Shahsavani N, Tavakkoly-Bazzaz J, Alimoghaddam K, Ghavamzadeh A, 2019, IL-6/IL-6R pathway is a therapeutic target in chemoresistant ovarian cancer. Tumori 105(1):84–91. https://doi. org/10.1177/0300891618784790 Samavati L, Rastogi R, Du W, Huttemann M, Fite A, Franchi L, 2009, STAT3 tyrosine phosphorylation is critical for interleukin 1 beta and interleukin-6 production in response to lipopolysaccharide and live bacteria. Mol Immunol 46(8–9):1867–1877 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAY PY 2020 VL 26 IS 2 BP 1365 EP 1366 DI 10.1007/s12253-019-00729-7 PG 2 ER PT J AU Lu, ChTh Collins, L Cohen, P Jay, A Campbell, MJ O’Callaghan, M AF Lu, Chengxuan Thomas Collins, Luke Cohen, Penelope Jay, Alex Campbell, M Jared O’Callaghan, Michael TI Prognostic Differences in ISUP Grade Group 4: a Systematic Review and Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Prostate cancer; Gleason score; Grade group 4; Biopsy; Prostate cancer specific mortality ID Prostate cancer; Gleason score; Grade group 4; Biopsy; Prostate cancer specific mortality AB The ISUP (Internal Society of Urologic Pathology) recently adopted a five-tiered prognostication system. There is evidence to suggest that the ISUP grade group 4 is a heterogeneous entity regarding prognosis. Our aim was to systematically examine the existing evidence to determine if outcome differences exist within the ISUP grade group 4. A systematic search of the literature for all studies examining the heterogeneity of the ISUP grade group 4 was conducted. Available studies were combined with meta-analysis to evaluate prognostic differences within the ISUP grade group 4 measured by all cause mortality (ACM) and the prostate cancer-specific mortality (PCSM). Eight studies were identified and utilised a variety of outcome measures to answer the question of heterogeneity within the ISUP grade group 4. Four of these studies examined prognosis using both ACM and PCSM. These were combined into a meta-analysis. The combined group of 5 + 3/3 + 5 had statistically significant higher ACM (hazard ratio [HR] 1.23, 95% confidence internal [Cl] 1.08–1.41) when compared to the 4+4 group. There was no difference in the PCSM between the two groups (HR 1.34, 95% CI 0.89–2.01). However, heterogeneity was high for this analysis secondary to a range of methodological differences. Our meta-analysis showed that Gleason grade 3 + 5/5 + 3 had higher ACM than Gleason grade group 4 + 4. Measures of PCSM were statistically insignificant, although heterogeneity was high. Evidence suggests that heterogeneity is likely, although inconclusive. Further studies with consistent methodologies are required to answer this question. C1 [Lu, Chengxuan Thomas] UNSW, The George Institute for Global Health, 2052 Sydney, NSW, Australia. [Collins, Luke] Royal Adelaide Hospital, 5000 Adelaide, Australia. [Cohen, Penelope] Royal Adelaide Hospital, SA PathologyAdelaide, Australia. [Jay, Alex] Flinders Medical Centre, Urology Unit, Bedford ParkAdelaide, Australia. [Campbell, M Jared] UNSW, Graduate School of Biomedical Engineering, Centre for Nanoscale Biophotonics, 2052 Sydney, NSW, Australia. [O’Callaghan, Michael] Flinders Medical Centre, Urology Unit, Bedford ParkAdelaide, Australia. RP Lu, ChTh (reprint author), UNSW, The George Institute for Global Health, 2052 Sydney, Australia. EM thomascxlu@gmail.com CR Sanda MG, Cadeddu JA, Kirkby E, Chen RC, Crispino T, Fontanarosa J, Freedland SJ, Greene K, Klotz LH, Makarov DV, Nelson JB, Rodrigues G, Sandler HM, Taplin ME, Treadwell JR, 2017, Clinically localized prostate cancer: AUA/ASTRO/SUO guideline. Part I: risk stratification, shared decision making, and care options. J Urol 199:683–690. , DOI 10.1016/j.juro. 2017.11.095 Sanda MG, Cadeddu JA, Kirkby E, Chen RC, Crispino T, Fontanarosa J, Freedland SJ, Greene K, Klotz LH, Makarov DV, Nelson JB, Rodrigues G, Sandler HM, Taplin ME, Treadwell JR, 2018, Clinically localized prostate cancer: AUA/ASTRO/SUO guideline. Part II: recommended approaches and details of specific care options. J Urol 199(4):990–997. , DOI 10.1016/j.juro. 2018.01.002 Epstein JI, Allsbrook WC Jr, Amin MB, Egevad LL, 2005, The 2005 International Society of Urological Pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol 29(9):1228–1242 (NCCN, NCCN, 2018, Clinical practice guidelines on prostate cancer Epstein JI, Zelefsky MJ, Sjoberg DD, Nelson JB, Egevad L, Magi- Galluzzi C, Vickers AJ, Parwani AV, Reuter VE, Fine SW, Eastham JA,Wiklund P, Han M, Reddy CA, Ciezki JP, Nyberg T, Klein EA, 2016, A contemporary prostate Cancer grading system: a validated alternative to the Gleason score. Eur Urol 69(3): 428–435. , DOI 10.1016/j.eururo.2015.06.046 Burdick MJ, Reddy CA, Ulchaker J, Angermeier K, Altman A, Chehade N, Mahadevan A, Kupelian PA, Klein EA, Ciezki JP, 2009, Comparison of biochemical relapse-free survival between primary Gleason score 3 and primary Gleason score 4 for biopsy Gleason score 7 prostate cancer. Int J Radiat Oncol Biol Phys 73(5): 1439–1445. , DOI 10.1016/j.ijrobp.2008.07.033 Kang DE, Fitzsimons NJ, Presti JC Jr, Kane CJ, Terris MK, Aronson WJ, Amling CL, Freedland SJ, Group SDS, 2007, Risk stratification of men with Gleason score 7 to 10 tumors by primary and secondary Gleason score: results from the SEARCH database. Urology 70(2):277–282. , DOI 10.1016/j.urology.2007.03.059 Koontz BF, TsivianM, Mouraviev V, Sun L, Vujaskovic Z,Moul J, LeeWR, 2012, Impact of primary Gleason grade on risk stratification for Gleason score 7 prostate cancers. Int J Radiat Oncol Biol Phys 82(1):200–203. , DOI 10.1016/j.ijrobp.2010.11.023 Stark JR, Perner S, StampferMJ, Sinnott JA, Finn S, Eisenstein AS, Ma J, Fiorentino M, Kurth T, LodaM, Giovannucci EL, RubinMA, Mucci LA, 2009, Gleason score and lethal prostate cancer: does 3 + 4 = 4 + 3? J Clin Oncol 27(21):3459–3464. , DOI 10.1200/ jco.2008.20.4669 Tollefson MK, Leibovich BC, Slezak JM, Zincke H, Blute ML, 2006, Long-term prognostic significance of primary Gleason pattern in patients with Gleason score 7 prostate cancer: impact on prostate cancer specific survival. J Urol 175(2):547– 551. , DOI 10.1016/s0022-5347(05)00152-7 Wright JL, Salinas CA, Lin DW, Kolb S, Koopmeiners J, Feng Z, Stanford JL, 2009, Prostate cancer specific mortality and Gleason 7 disease differences in prostate cancer outcomes between cases with Gleason 4 + 3 and Gleason 3 + 4 tumors in a population based cohort. J Urol 182(6):2702–2707. , DOI 10.1016/j.juro. 2009.08.026 Huynh MA, Chen MH, Wu J, Braccioforte MH, Moran BJ, D'Amico AV, 2016, Gleason score 3 + 5 or 5 + 3 versus 4 + 4 prostate cancer: the risk of death. Eur Urol 69(6):976– 979. , DOI 10.1016/j.eururo.2015.08.054 Mahal BA, Muralidhar V, Chen YW, Choueiri TK, Hoffman KE, Hu JC, Sweeney CJ, Yu JB, Feng FY, Trinh QD, Nguyen PL, 2016, Gleason score 5 + 3 = 8 prostate cancer: much more like Gleason score 9? BJU Int 118(1):95–101. , DOI 10.1111/bju.13239 Thomas Chengxuan Lu MOC, 2017, Are there prognostic differences within the International Society of Urological Pathology Grade Group 4 comprised of Gleason pattern 4+4, 3+5, 5+3? A systematic review. http://www.crd.york.ac.uk/PROSPERO/ display_record.php?ID=CRD42017058923. Accessed 27 May 2017 Stang A, 2010, Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in metaanalyses. Eur J Epidemiol 25(9):603–605. , DOI 10.1007/ s10654-010-9491-z Lu TC, Moretti K, Beckmann K, Cohen P, O'Callaghan M, 2017, ISUP group 4 - a homogenous group of prostate cancers? Pathol Oncol Res 24:921–925. , DOI 10.1007/s12253-017-0331- 2 Rusthoven CG, Carlson JA,Waxweiler TV, Yeh N, Raben D, Flaig TW, Kavanagh BD, 2014, The prognostic significance of Gleason scores in metastatic prostate cancer. Urol Oncol 32(5):707–713. , DOI 10.1016/j.urolonc.2014.01.004 Rusthoven CG, Waxweiler TV, DeWitt PE, Flaig TW, Raben D, KavanaghBD, 2015, Gleason stratifications prognostic for survival in men receiving definitive external beam radiation therapy for localized prostate cancer. Urol Oncol 33(2):71 e11–71 e79. , DOI 10.1016/j.urolonc.2014.07.010 Harding-Jackson N, Kryvenko ON, Whittington EE, Eastwood DC, Tjionas GA, Jorda M, Iczkowski KA, 2016, Outcome of Gleason 3 + 5 = 8 prostate cancer diagnosed on needle biopsy: prognostic comparison with Gleason 4 + 4 = 8. J Urol 196(4): 1076–1081. , DOI 10.1016/j.juro.2016.05.105 Gandaglia G, Karnes RJ, Sivaraman A, Moschini M, Fossati N, Zaffuto E, Dell'Oglio P, Cathelineau X, Montorsi F, Sanchez- Salas R, Briganti A, 2017, Are all grade group 4 prostate cancers created equal? Implications for the applicability of the novel grade grouping. Urol Oncol 35(7):461 e467–461 e414. , DOI 10. 1016/j.urolonc.2017.02.012 van den Bergh RC, van der Kwast TH, de Jong J, Zargar H, Ryan AJ, Costello AJ, Murphy DG, van der Poel HG(2016)Validation of the novel International Society of Urological Pathology 2014 fivetier Gleason grade grouping: biochemical recurrence rates for 3+5 disease may be overestimated. BJU Int 118(4):502–505. https://doi. org/10.1111/bju.13478 JacksonW, Hamstra DA, Johnson S, Zhou J, Foster B, Foster C, Li D, Song Y, Palapattu GS, Kunju LP, Mehra R, Feng FY, 2013, Gleason pattern 5 is the strongest pathologic predictor of recurrence, metastasis, and prostate cancer-specific death in patients receiving salvage radiation therapy following radical prostatectomy. Cancer 119(18):3287–3294. , DOI 10.1002/cncr.28215 Sabolch A, Feng FY, Daignault-Newton S, Halverson S, Blas K, Phelps L, Olson KB, Sandler HM, Hamstra DA, 2011, Gleason pattern 5 is the greatest risk factor for clinical failure and death from prostate cancer after doseescalated radiation therapy and hormonal ablation. Int J Radiat Oncol Biol Phys 81(4):e351–e360. , DOI 10. 1016/j.ijrobp.2011.01.063 Antonarakis ES, Feng Z, Trock BJ, Humphreys EB, Carducci MA, Partin AW, Walsh PC, Eisenberger MA, 2012, The natural history of metastatic progression in men with prostate-specific antigen recurrence after radical prostatectomy: long-term follow-up. BJU Int 109(1):32–39. , DOI 10.1111/j.1464-410X.2011.10422.x Freedland SJ, Humphreys EB, Mangold LA, Eisenberger M, Dorey FJ, Walsh PC, Partin AW, 2005, Risk of prostate cancer-specific mortality following biochemical recurrence after radical prostatectomy. Jama 294(4):433–439. , DOI 10.1001/jama.294.4.433 Punnen S, Cooperberg MR, D'Amico AV, Karakiewicz PI, Moul JW, Scher HI, Schlomm T, Freedland SJ, 2013, Management of biochemical recurrence after primary treatment of prostate cancer: a systematic review of the literature. Eur Urol 64(6):905–915. , DOI 10.1016/j.eururo.2013.05.025 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1367 EP 1375 DI 10.1007/s12253-019-00632-1 PG 9 ER PT J AU Raffone, A Travaglino, A Saccone, G D’Alessandro, P Arduino, B Mascolo, M De Placido, G Insabato, L Zullo, F AF Raffone, Antonio Travaglino, Antonio Saccone, Gabriele D’Alessandro, Pietro Arduino, Bruno Mascolo, Massimo De Placido, Giuseppe Insabato, Luigi Zullo, Fulvio TI Diabetes Mellitus Is Associated with Occult Cancer in Endometrial Hyperplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Concurrent cancer; Endometrial cancer; Endometrial intraepithelial neoplasia; Glycemia; Occult cancer; Risk ID Concurrent cancer; Endometrial cancer; Endometrial intraepithelial neoplasia; Glycemia; Occult cancer; Risk AB In the management of women diagnosed with endometrial hyperplasia (EH), it is crucial to determine the risk of coexistent cancer. Diabetes mellitus has been recently suggested as a significant risk factor. However, results in this regard are conflicting. Our aim was to assess the association between diabetes mellitus and coexistent cancer in women diagnosed with endometrial hyperplasia. A systematic review and meta-analysis was performed by searching electronic databases from their inception to October 2018 for studies assessing the presence of coexistent cancer after a preoperative diagnosis of endometrial hyperplasia in women stratified for diabetes mellitus. Odds ratio was calculated with 95% confidence interval; a p value <0.05 was considered significant. Twelve retrospective studies with 1579 EH were included. Diabetes mellitus showed significant association with the presence of cancer coexistent with endometrial hyperplasia (OR = 1.96; 95% CI, 1.07–3.60; p = 0.03). Heterogeneity among studies was moderate (I2 = 55%). Funnel plot showed asymmetric distribution of OR values, with the large and accurate studies showing results stronger than small and less accurate one; this finding should exclude a publication bias. In women diagnosed with endometrial hyperplasia, diabetes mellitus is a risk factor for coexistent cancer, and thus may be included in a predictive algorithm for the risk stratification. In women conservatively treated, glycemic control may be required to prevent the risk of progression. Further studies are necessary to confirm the clinical significance of diabetes mellitus in this field. C1 [Raffone, Antonio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics UnitNaples, Italy. [Travaglino, Antonio] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Saccone, Gabriele] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics UnitNaples, Italy. [D’Alessandro, Pietro] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics UnitNaples, Italy. [Arduino, Bruno] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics UnitNaples, Italy. [Mascolo, Massimo] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [De Placido, Giuseppe] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics UnitNaples, Italy. [Insabato, Luigi] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Zullo, Fulvio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics UnitNaples, Italy. RP Travaglino, A (reprint author), University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, 80131 Naples, Italy. EM antonio.travaglino.ap@gmail.com CR Kurman R, Carcangiu M, Herrington C, Young R, 2014, World Health Organisation classification of tumors of female reproductive organs, 4th edn. International Agency for Research on Cancer, IARC, Press, Lyon Sanderson PA, Critchley HOD, Williams ARW, Arends MJ, Saunders PTK, 2017, New concepts for an old problem: the diagnosis of endometrial hyperplasia. Hum Reprod Update 23(2):232– 254 Travaglino A, Raffone A, Saccone G et al, 2018, Endometrial hyperplasia and risk of coexistent cancer: WHO vs EIN criteria. Histopathology. Baak JP, Mutter GL, 2005, EIN and WHO94. J Clin Pathol 58(1): 1–6 Ordi J, Bergeron C, Hardisson D, McCluggage WG, Hollema H, Felix A, Soslow RA, Oliva E, Tavassoli FA, Alvarado-Cabrero I, Wells M, Nogales FF, 2014, Reproducibility of current classifications of endometrial endometrioid glandular proliferations: further evidence supporting a simplified classification. Histopathology. 64(2):284–292 McCluggage WG, 2006, My approach to the interpretation of endometrial biopsies and curettings. J Clin Pathol 59(8):801–812 Travaglino A, Raffone A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2018, Loss of Bcl-2 immunohistochemical expression in endometrial hyperplasia: a specific marker of precancer and novel indication for treatment. A systematic review and meta-analysis. Acta Obstet Gynecol Scand 97:1415–1426 Raffone A, Travaglino A, Saccone G, Mascolo M, Insabato L, Mollo A, de Placido G, Zullo F, 2019, PAX2 in endometrial carcinogenesis and in differential diagnosis of endometrial hyperplasia. A systematic review and meta-analysis of diagnostic accuracy. Acta Obstet Gynecol Scand 98(3):287–299 Raffone A, Travaglino A, Saccone G, CampaninoMR, Mollo A, de Placido G, Insabato L, Zullo F, 2019, Loss of PTEN expression as diagnostic marker of endometrial precancer: a systematic review and meta-analysis. Acta Obstet Gynecol Scand 98(3):275–286 Travaglino A, Raffone A, Saccone G, de Luca C,Mollo A, Mascolo M, de Placido G, Insabato L, Zullo F, 2019, Immunohistochemical nuclear expression of β-catenin as a surrogate of CTNNB1 exon 3 mutation in endometrial Cancer. Am J Clin Pathol 151:529–538. , DOI 10.1093/ajcp/aqy178 Chen YL, Cheng WF, Lin MC, Huang CY, Hsieh CY, Chen CA, 2009, Concurrent endometrial carcinoma in patients with a curettage diagnosis of endometrial hyperplasia. J Formos Med Assoc 108(6):502–507 Chen YL,Wang KL, ChenMY,YuMH,Wu CH, KeYM, Chen YJ, Chang YY, Hsu KF, Yen MS, 2013, Risk factor analysis of coexisting endometrial carcinoma in patients with endometrial hyperplasia: a retrospective observational study of Taiwanese gynecologic oncology group. J Gynecol Oncol 24(1):14–20 Matsuo K, Ramzan AA, Gualtieri MR, Mhawech-Fauceglia P, Machida H, Moeini A, Dancz CE, Ueda Y, Roman LD, 2015, Prediction of concurrent endometrial carcinoma in women with endometrial hyperplasia. Gynecol Oncol 139(2):261–267 Moher D, Shamseer L, Clarke M et al, 2015, Preferred reporting items for systematic review and meta-analysis protocols, PRISMAP, 2015 statement. Syst Rev 4(1) Whiting PF, Rutjes AW, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, Leeflang MM, Sterne JA, Bossuyt PM, QUADAS-2 Group, 2011)QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med 155(8):529–536 Lacey JV Jr, Mutter GL, Nucci MR, Ronnett BM, Ioffe OB, Rush BB, Glass AG, Richesson DA, Chatterjee N, Langholz B, Sherman ME, 2008, Risk of subsequent endometrial carcinoma associated with endometrial intraepithelial neoplasia classification of endometrial biopsies. Cancer 113(8):2073–2081 Baak JP,Mutter GL, Robboy S, van Diest PJ, Uyterlinde AM, Orbo A, Palazzo J, Fiane B, Lovslett K, Burger C, Voorhorst F, Verheijen RH, 2005, The molecular genetics and morphometry-based endometrial intraepithelial neoplasia classification system predicts disease progression in endometrial hyperplasia more accurately than the 1994 World Health Organization classification system. Cancer. 103(11):2304–2312 Menke A, Casagrande S, Geiss L, Cowie CC, 2015, Prevalence of and trends in diabetes among adults in the United States, 1988- 2012. JAMA. 314(10):1021–1029 Widra EA, Dunton CJ, McHugh M, Palazzo JP, 1995 May, Endometrial hyperplasia and the risk of carcinoma. Int J Gynecol Cancer 5(3):233–235 Bilgin T, Ozuysal S, Ozan H, Atakan T, 2004, Coexisting endometrial cancer in patients with a preoperative diagnosis of atypical endometrial hyperplasia. J Obstet Gynaecol Res 30(3):205–209 Merisio C, Berretta R, De Ioris A et al, 2005, Endometrial cancer in patients with preoperative diagnosis of atypical endometrial hyperplasia. Eur J Obstet Gynecol Reprod Biol 122(1):107–111 Daud S, Jalil SS, Griffin M, Ewies AA, 2011 Nov, Endometrial hyperplasia - the dilemma of management remains: a retrospective observational study of 280 women. Eur J Obstet Gynecol Reprod Biol 159(1):172–175 Robbe EJ, van Kuijk SM, de Boed EM et al, 2012, Predicting the coexistence of an endometrial adenocarcinoma in the presence of atypical complex hyperplasia: immunohistochemical analysis of endometrial samples. Int J Gynecol Cancer 22(7):1264–1272 Touboul C, Piel B, Koskas M, Gonthier C, Ballester M, Cortez A, Darai E, 2014, Factors predictive of endometrial carcinoma in patients with atypical endometrial hyperplasia on preoperative histology. Anticancer Res 34(10):5671–5676 Zhou L, Meng Z, Wu Y, Zhu H, Wang X, 2014, Prediction of endometrial carcinogenesis probability while diagnosed as atypical endometrial hyperplasia: a new risk model based on age, CA199 and CA125 assay. Eur J Obstet Gynecol Reprod Biol 183:5–9 Dolanbay M, Kutuk MS, Uludag S, Bulut A, Ozgun M, Ozcelik B, Serin I, 2015, Concurrent endometrial carcinoma in hysterectomy specimens in patients with histopathological diagnosis of endometrial hyperplasia in curettage specimens. Ginekol Pol 86(10):753– 758 Kadirogullari P, Atalay CR, OzdemirO, SariME, 2015, Prevalence of co-existing endometrial carcinoma in patients with preoperative diagnosis of endometrial hyperplasia. J Clin Diagn Res 9(10): QC10–QC14 Ferenczy A, Gelfand MM, Tzipris F, 1983, The cytodynamics of endometrial hyperplasia and carcinoma. A review. Ann Pathol 3(3): 189–201 Travaglino A, Raffone A, Saccone G, Mollo A, de Placido G, Mascolo M, Insabato L, Zullo F, 2019, Complexity of glandular architecture should be reconsidered in the classification and management of endometrial hyperplasia. APMIS. 127:427–434. https:// doi.org/10.1111/apm.12945 Raffone A, Travaglino A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2019, Endometrial hyperplasia and progression to cancer: which classification system stratifies the risk better? A systematic review and meta-analysis. Arch Gynecol Obstet 299: 1233–1242. , DOI 10.1007/s00404-019-05103-1 Travaglino A, Raffone A, Saccone G, et al, 2019, PTEN immunohistochemistry in endometrial hyperplasia: which are the optimal criteria for the diagnosis of precancer? APMIS. , DOI 10. 1111/apm.12938 Raglan O, Kalliala I, Markozannes G, Cividini S, Gunter MJ, Nautiyal J, Gabra H, Paraskevaidis E, Martin-Hirsch P, Tsilidis KK, Kyrgiou M, 2018, Risk factors for endometrial Cancer: an umbrella review of the literature. Int J Cancer. , DOI 10. 1002/ijc.31961 Fader AN, Arriba LN, Frasure HE, von Gruenigen VE, 2009, Endometrial cancer and obesity: epidemiology, biomarkers, prevention and survivorship. Gynecol Oncol 114(1):121–127 PollakM(2008, Insulin and insulin-like growth factor signalling in neoplasia. Nat Rev Cancer 8:915–928 Sasaki LMP, Andrade KRC, Figueiredo ACMG, Wanderley MDS, Pereira MG, 2018, Factors associated with malignancy in Hysteroscopically resected endometrial polyps: a systematic review and meta-analysis. J Minim Invasive Gynecol 25(5):777–785 Management of Endometrial Hyperplasia Green-top Guideline No. 67 RCOG/BSGE joint guideline | February 2016 Zhang Q, Qi G, Kanis MJ, Dong R, Cui B, Yang X, Kong B, 2017 May 3, Comparison among fertility-sparing therapies for well differentiated early-stage endometrial carcinoma and complex atypical hyperplasia. Oncotarget. 8(34):57642–57653 Giampaolino P, Di Spiezio Sardo A, Mollo A et al, 2018, Hysteroscopic endometrial focal resection followed by Levonorgestrel intrauterine device insertion as a fertility-sparing treatment of atypical endometrial hyperplasia and early endometrial Cancer: a retrospective study. J Minim Invasive Gynecol Raffone A, Travaglino A, Saccone G, Alviggi C, Mascolo M, de Placido G, Insabato L, Mollo A, Zullo F, 2019, Management of women with atypical polypoid adenomyoma of the uterus: a quantitative systematic review. Acta Obstet Gynecol Scand. https://doi. org/10.1111/aogs.13553 Meireles CG, Pereira SA, Valadares LP, Rego DF, Simeoni LA, Guerra ENS, Lofrano-Porto A, 2017, Effects of metformin on endometrial cancer: systematic review and meta-analysis. Gynecol Oncol 147(1):167–180 Raffone A, Travaglino A, Saccone G, Mollo A, de Placido G, Insabato L, Zullo F, 2019, Should progesterone and estrogens receptors be assessed for predicting the response to conservative treatment of endometrial hyperplasia and cancer? A systematic review and meta-analysis. Acta Obstet Gynecol Scand. , DOI 10. 1111/aogs.13586 Travaglino A, Raffone A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2018, PTEN as a predictive marker of response to conservative treatment in endometrial hyperplasia and early endometrial cancer. A systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol 231:104–110 Travaglino A, Raffone A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2019, Immunohistochemical predictive markers of response to conservative treatment of endometrial hyperplasia and early endometrial cancer: a systematic review. Acta Obstet Gynecol Scand. , DOI 10.1111/aogs.13587 Raffone A, Travaglino A, Saccone G, Di Maio A, Mollo A, Mascolo M, De Rosa R, De Placido G, Insabato L, Zullo F,, 2019, Diabetes mellitus and responsiveness of endometrial hyperplasia and early endometrial cancer to conservative treatment. Gynecological Endocrinology:1-6 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1377 EP 1384 DI 10.1007/s12253-019-00684-3 PG 8 ER PT J AU Wong, D Yip, S Sorensen, HP AF Wong, Derek Yip, Stephen Sorensen, H Poul TI Methods for Identifying Patients with Tropomyosin Receptor Kinase (TRK) Fusion Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE NTRK gene fusions; TRK fusions; TRK inhibitors; Next-generation sequencing; NGS ID NTRK gene fusions; TRK fusions; TRK inhibitors; Next-generation sequencing; NGS AB NTRK gene fusions affecting the tropomyosin receptor kinase (TRK) protein family have been found to be oncogenic drivers in a broad range of cancers. Small molecule inhibitors targeting TRK activity, such as the recently Food and Drug Administration-approved agent larotrectinib (Vitrakvi®), have shown promising efficacy and safety data in the treatment of patients with TRK fusion cancers. NTRK gene fusions can be detected using several different approaches, including fluorescent in situ hybridization, reverse transcription polymerase chain reaction, immunohistochemistry, next-generation sequencing, and ribonucleic acid-based multiplexed assays. Identifying patients with cancers that harbor NTRK gene fusions will optimize treatment outcomes by providing targeted precision therapy. C1 [Wong, Derek] University of British Columbia, Department of PathologyVancouver, BC, Canada. [Yip, Stephen] University of British Columbia, Department of PathologyVancouver, BC, Canada. [Sorensen, H Poul] Provincial Health Services Authority, British Columbia Cancer AgencyVancouver, BC, Canada. RP Sorensen, HP (reprint author), Provincial Health Services Authority, British Columbia Cancer Agency, Vancouver, Canada. EM psor@mail.ubc.ca CR Maruyama I, 2014, Mechanisms of activation of receptor tyrosine kinases: monomers or dimers. Cells 3(2):304–330 Arevalo JC, Wu SH, 2006, Neurotrophin signaling: many exciting surprises! Cell Mol Life Sci 63(13):1523–1537. , DOI 10. 1007/s00018-006-6010-1 Nakagawara A, 2001, Trk receptor tyrosine kinases: a bridge between cancer and neural development. Cancer Lett 169(2):107–114 Huang EJ, Reichardt LF, 2003, Trk receptors: roles in neuronal signal transduction. Annu Rev Biochem 72:609–642. https://doi. org/10.1146/annurev.biochem.72.121801.161629 Amatu A, Sartore-Bianchi A, Siena S, 2016, NTRKgene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open 1(2):e000023. , DOI 10.1136/esmoopen- 2015-000023 Medves S, Demoulin JB, 2012, Tyrosine kinase gene fusions in cancer: translating mechanisms into targeted therapies. J Cell Mol Med 16(2):237–248. , DOI 10.1111/j.1582-4934.2011. 01415.x Wai DH, Knezevich SR, Lucas T, Jansen B, Kay RJ, Sorensen PHB, 2000, The ETV6-NTRK3 gene fusion encodes a chimeric protein tyrosine kinase that transforms NIH3T3 cells. Oncogene 19(7): 906–915. , DOI 10.1038/sj.onc.1203396 Vaishnavi A, Le AT, Doebele RC, 2015, TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov 5(1): 25–34. , DOI 10.1158/2159-8290.Cd-14-0765 Triche TJ, Hicks MJ, Sorensen PH, 2015, Diagnostic pathology of pediatric malignancies. In: Pizzo PA, Poplack DG, eds, Principles and practice of pediatric oncology, 7th Edition. Wolters Kluwer Health Martin-Zanca D, Hughes SH, Barbacid M, 1986, A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Nature 319(6056):743–748. https://doi. org/10.1038/319743a0 Knezevich SR, McFadden DE, TaoW, Lim JF, Sorensen PH, 1998, A novel ETV6-NTRK3 gene fusion in congenital fibrosarcoma. Nat Genet 18(2):184–187. , DOI 10.1038/ng0298-184 Knezevich SR, Garnett MJ, Pysher TJ, Beckwith JB, Grundy PE, Sorensen PH, 1998, ETV6-NTRK3 gene fusions and trisomy 11 establish a histogenetic link between mesoblastic nephroma and congenital fibrosarcoma. Cancer Res 58(22):5046–5048 Rubin BP, Chen CJ, Morgan TW, Xiao S, Grier HE, Kozakewich HP, Perez-Atayde AR, Fletcher JA, 1998, Congenital mesoblastic nephroma t(12;15, is associated with ETV6-NTRK3 gene fusion: cytogenetic and molecular relationship to congenital, infantile, fibrosarcoma. Am J Pathol 153(5):1451–1458. , DOI 10. 1016/S0002-9440(10)65732-X Tognon C, Knezevich SR, Huntsman D, Roskelley CD, Melnyk N, Mathers JA, Becker L, Carneiro F, MacPherson N, Horsman D, Poremba C, Sorensen PH, 2002, Expression of the ETV6- NTRK3 gene fusion as a primary event in human secretory breast carcinoma. Cancer Cell 2(5):367–376 Eguchi M, Eguchi-Ishimae M, Tojo A, Morishita K, Suzuki K, Sato Y, Kudoh S, Tanaka K, Setoyama M, Nagamura F, Asano S, Kamada N, 1999, Fusion of ETV6 to neurotrophin-3 receptor TRKC in acute myeloid leukemia with t(12;15)(p13;q25). Blood 93(4):1355–1363 Leeman-Neill RJ, Kelly LM, Liu P, Brenner AV, Little MP, Bogdanova TI, Evdokimova VN, Hatch M, Zurnadzy LY, Nikiforova MN, Yue NJ, Zhang M, Mabuchi K, Tronko MD, Nikiforov YE, 2014, ETV6-NTRK3 is a common chromosomal rearrangement in radiation-associated thyroid cancer. Cancer 120(6):799–807. , DOI 10.1002/cncr.28484 Wu G, Diaz AK, Paugh BS, Rankin SL, Ju B, Li Y, Zhu X, Qu C, Chen X, Zhang J, Easton J, Edmonson M, Ma X, Lu C, Nagahawatte P, Hedlund E, Rusch M, Pounds S, Lin T, Onar- Thomas A, Huether R, Kriwacki R, Parker M, Gupta P, Becksfort J, Wei L, Mulder HL, Boggs K, Vadodaria B, Yergeau D, Russell JC, Ochoa K, Fulton RS, Fulton LL, Jones C, Boop FA, Broniscer A, Wetmore C, Gajjar A, Ding L, Mardis ER, Wilson RK, Taylor MR, Downing JR, Ellison DW, Zhang J, Baker SJ, 2014, The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma. Nat Genet 46(5):444–450. , DOI 10.1038/ng.2938 El Demellawy D, Cundiff CA, Nasr A, Ozolek JA, Elawabdeh N, Caltharp SA, Masoudian P, Sullivan KJ, de Nanassy J, Shehata BM, 2016, Congenital mesoblastic nephroma: a study of 19 cases using immunohistochemistry and ETV6-NTRK3 fusion gene rearrangement. Pathology 48(1):47–50. , DOI 10.1016/j.pathol. 2015.11.007 Skalova A, Vanecek T, Sima R, Laco J, Weinreb I, Perez-Ordonez B, Starek I,GeierovaM, SimpsonRH, Passador-Santos F, Ryska A, Leivo I, Kinkor Z, Michal M, 2010, Mammary analogue secretory carcinoma of salivary glands, containing the ETV6-NTRK3 fusion gene: a hitherto undescribed salivary gland tumor entity. Am J Surg Pathol 34(5):599–608. , DOI 10.1097/PAS. 0b013e3181d9efcc Bourgeois JM, Knezevich SR, Mathers JA, Sorensen PH, 2000, Molecular detection of the ETV6-NTRK3 gene fusion differentiates congenital fibrosarcoma from other childhood spindle cell tumors. Am J Surg Pathol 24(7):937–946 Sartore-Bianchi A, Ardini E, Bosotti R, Amatu A, Valtorta E, Somaschini A, Raddrizzani L, Palmeri L, Banfi P, Bonazzina E, Misale S,Marrapese G, Leone A, Alzani R, Luo D, Hornby Z, Lim J, Veronese S, Vanzulli A, Bardelli A, Martignoni M, Davite C, Galvani A, Isacchi A, Siena S, 2016, Sensitivity to entrectinib associated with a novel LMNA-NTRK1 gene fusion in metastatic colorectal cancer. J Natl Cancer Inst 108(1). , DOI 10. 1093/jnci/djv306 Drilon A, Siena S, Ou SI, PatelM, AhnMJ, Lee J, Bauer TM, Farago AF, Wheler JJ,Liu SV, Doebele R, Giannetta L, Cerea G, Marrapese G, Schirru M, Amatu A, Bencardino K, Palmeri L, Sartore-Bianchi A, Vanzulli A, Cresta S, Damian S, Duca M, Ardini E, Li G, Christiansen J, Kowalski K, Johnson AD, Patel R, Luo D, Chow- Maneval E, Hornby Z, Multani PS, Shaw AT, De Braud FG, 2017, Safety and antitumor activity of the multitargeted pan-TRK, ROS1, and ALK inhibitor entrectinib: combined results from two phase I trials, ALKA-372-001 and STARTRK-1). Cancer Discov 7(4):400– 409. , DOI 10.1158/2159-8290.CD-16-1237 Drilon A, Laetsch TW, Kummar S, DuBois SG, Lassen UN, Demetri GD, Nathenson M, Doebele RC, Farago AF, Pappo AS, Turpin B, Dowlati A, Brose MS, Mascarenhas L, Federman N, Berlin J, El-Deiry WS, Baik C, Deeken J, Boni V, Nagasubramanian R, Taylor M, Rudzinski ER, Meric-Bernstam F, Sohal DPS, Ma PC, Raez LE, Hechtman JF, Benayed R, Ladanyi M, Tuch BB, Ebata K, Cruickshank S, Ku NC, Cox MC, Hawkins DS, Hong DS, Hyman DM, 2018, Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med 378(8):731–739. , DOI 10.1056/ NEJMoa1714448 Kummar S, Lassen UN, 2018, TRK inhibition: a new tumoragnostic treatment strategy. Target Oncol 13(5):545–556. https:// doi.org/10.1007/s11523-018-0590-1 Lezcano C, Shoushtari AN, Ariyan C, Hollmann TJ, Busam KJ, 2018, Primary and metastatic melanoma with NTRK fusions. Am J Surg Pathol 42(8):1052–1058. , DOI 10.1097/PAS. 0000000000001070 Prabhakaran N, Guzman MA, Navalkele P, Chow-Maneval E, Batanian JR, 2018, Novel TLE4-NTRK2 fusion in a ganglioglioma identified by array-CGH and confirmed by NGS: potential for a gene targeted therapy. Neuropathology 38:380–386. https://doi. org/10.1111/neup.12458 Chmielecki J, Bailey M, He J, Elvin J, Vergilio JA, Ramkissoon S, Suh J, Frampton GM, Sun JX, Morley S, Spritz D, Ali S, Gay L, Erlich RL, Ross JS, Buxhaku J, Davies H, Faso V, Germain A, Glanville B, Miller VA, Stephens PJ, Janeway KA, Maris JM, Meshinchi S, Pugh TJ, Shern JF, Lipson D, 2017, Genomic profiling of a large set of diverse pediatric cancers identifies known and novel mutations across tumor spectra. Cancer Res 77(2):509–519. , DOI 10.1158/0008-5472.CAN-16-1106 Hechtman JF, Benayed R, Hyman DM, Drilon A, Zehir A, Frosina D, Arcila ME, Dogan S, Klimstra DS, Ladanyi M, Jungbluth AA, 2017, Pan-Trk immunohistochemistry is an efficient and reliable screen for the detection of NTRK fusions.AmJ Surg Pathol 41(11): 1547–1551. , DOI 10.1097/PAS.0000000000000911 MilioneM, Ardini E, Christiansen J,Valtorta E,Veronese S, Bosotti R, Pellegrinelli A, Testi A, Pietrantonio F, Fuca G, Wei G, Murphy D, Siena S, Isacchi A, De Braud F, 2017, Identification and characterization of a novel SCYL3-NTRK1 rearrangement in a colorectal cancer patient. Oncotarget 8(33):55353–55360. , DOI 10.18632/oncotarget.19512 Edgren H OK, Ruusulehto A, Ganji G, 2015, Rapid pan-cancer identification of previously unidentified fusion genes to enable novel targeted therapeutics. Proceedings: AACR 106th annual meeting 2015. 75:suppl 15; abstract 4793 Kim J, Lee Y, Cho HJ, Lee YE, An J, Cho GH, Ko YH, Joo KM, Nam DH, 2014, NTRK1 fusion in glioblastoma multiforme. PLoS One 9(3):e91940. , DOI 10.1371/journal.pone.0091940 Sigal D, Tartar M, Xavier M, Bao F, Foley P, Luo D, Christiansen J, Hornby Z,Maneval EC, Multani P, 2017, Activity of Entrectinib in a patient with the first reported NTRK fusion in neuroendocrine Cancer. Journal of the National Comprehensive Cancer Network : JNCCN 15(11):1317–1322. , DOI 10.6004/jnccn.2017.7029 Stransky N, Cerami E, Schalm S, Kim JL, Lengauer C, 2014, The landscape of kinase fusions in cancer. Nat Commun 5:4846. https:// doi.org/10.1038/ncomms5846 ChurchAJ, CalicchioML, NardiV, Skalova A, Pinto A, DillonDA, Gomez-Fernandez CR, Manoj N, Haimes JD, Stahl JA, Dela Cruz FS, Tannenbaum-Dvir S, Glade-Bender JL, Kung AL, DuBois SG, Kozakewich HP, Janeway KA, Perez-Atayde AR, Harris MH, 2018, Recurrent EML4-NTRK3 fusions in infantile fibrosarcoma and congenital mesoblastic nephroma suggest a revised testing strategy. Modern pathology : an official journal of the United States and Canadian academy of. Pathology, Inc 31(3):463–473. , DOI 10.1038/modpathol.2017.127 Lee SJ, Li GG, Kim ST, Hong ME, Jang J, Yoon N, Ahn SM, Murphy D, Christiansen J, Wei G, Hornby Z, Lee DW, Park JO, Park YS, Lim HY, Hong SN, Kim SH, Kang WK, Park K, Park WY, Kim KM, Lee J, 2015, NTRK1 rearrangement in colorectal cancer patients: evidence for actionable target using patient-derived tumor cell line. Oncotarget 6(36):39028–39035. , DOI 10. 18632/oncotarget.5494 Brenca M, Rossi S, Polano M, Gasparotto D, Zanatta L, Racanelli D, Valori L, Lamon S, Dei Tos AP, Maestro R, 2016, Transcriptome sequencing identifies ETV6-NTRK3 as a gene fusion involved in GIST. J Pathol 238(4):543–549. , DOI 10.1002/path.4677 Yoshihara K,Wang Q, Torres-GarciaW, Zheng S, Vegesna R, Kim H, Verhaak RG, 2015, The landscape and therapeutic relevance of cancer-associated transcript fusions. Oncogene 34(37):4845–4854. , DOI 10.1038/onc.2014.406 Jones DT, Hutter B, Jager N, Korshunov A, Kool M, Warnatz HJ, Zichner T, Lambert SR, Ryzhova M, Quang DA, Fontebasso AM, Stutz AM, Hutter S, Zuckermann M, Sturm D, Gronych J, Lasitschka B, Schmidt S, Seker-Cin H, Witt H, Sultan M, Ralser M, Northcott PA, Hovestadt V, Bender S, Pfaff E, Stark S, Faury D, Schwartzentruber J, Majewski J,Weber UD, Zapatka M, Raeder B, Schlesner M, Worth CL, Bartholomae CC, von Kalle C, Imbusch CD, Radomski S, Lawerenz C, van Sluis P, Koster J, Volckmann R, Versteeg R, Lehrach H, Monoranu C, Winkler B, Unterberg A, Herold-Mende C, Milde T, Kulozik AE, Ebinger M, Schuhmann MU, Cho YJ, Pomeroy SL, von Deimling A, Witt O, Taylor MD, Wolf S, KarajannisMA, Eberhart CG, ScheurlenW, Hasselblatt M, Ligon KL, KieranMW, Korbel JO, Yaspo ML, Brors B, Felsberg J, Reifenberger G, Collins VP, Jabado N, Eils R, Lichter P, PfisterSM, International Cancer Genome Consortium PedBrain Tumor P, 2013, Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat Genet 45(8):927–932. , DOI 10.1038/ng.2682 Roberts KG, Li Y, Payne-Turner D, Harvey RC, Yang YL, Pei D, McCastlain K, Ding L, Lu C, Song G,Ma J, Becksfort J, Rusch M, Chen SC, Easton J, Cheng J, Boggs K, Santiago-Morales N, Iacobucci I, Fulton RS, Wen J, Valentine M, Cheng C, Paugh SW, Devidas M, Chen IM, Reshmi S, Smith A, Hedlund E, Gupta P, Nagahawatte P,Wu G, Chen X,Yergeau D,Vadodaria B,Mulder H, Winick NJ, Larsen EC, Carroll WL, Heerema NA, Carroll AJ, Grayson G, Tasian SK, Moore AS, Keller F, Frei-Jones M, Whitlock JA, Raetz EA, White DL, Hughes TP, Guidry Auvil JM, Smith MA, Marcucci G, Bloomfield CD, Mrozek K, Kohlschmidt J, Stock W, Kornblau SM, Konopleva M, Paietta E, Pui CH, Jeha S, Relling MV, Evans WE, Gerhard DS, Gastier- Foster JM, Mardis E, Wilson RK, Loh ML, Downing JR, Hunger SP,Willman CL, Zhang J, Mullighan CG, 2014, Targetable kinaseactivating lesions in Ph-like acute lymphoblastic leukemia. NEngl J Med 371(11):1005–1015. ht t p s : / /doi . o r g /10.1056/ NEJMoa1403088 George J, Walter V, Peifer M, Alexandrov LB, Seidel D, Leenders F, Maas L, Muller C, Dahmen I, Delhomme TM, Ardin M, Leblay N, Byrnes G, Sun R, De Reynies A, McLeer-Florin A, Bosco G, Malchers F,Menon R, Altmuller J, Becker C,Nurnberg P, Achter V, Lang U, Schneider PM, Bogus M, Soloway MG, Wilkerson MD, Cun Y, McKay JD, Moro-Sibilot D, Brambilla CG, Lantuejoul S, Lemaitre N, Soltermann A, Weder W, Tischler V, Brustugun OT, Lund-Iversen M, Helland A, Solberg S, Ansen S, Wright G, Solomon B, Roz L, Pastorino U, Petersen I, Clement JH, Sanger J, Wolf J, Vingron M, Zander T, Perner S, Travis WD, Haas SA, Olivier M, Foll M, Buttner R, Hayes DN, Brambilla E, Fernandez- Cuesta L, Thomas RK, 2018, Integrative genomic profiling of large-cell neuroendocrine carcinomas reveals distinct subtypes of high-grade neuroendocrine lung tumors. Nat Commun 9(1):1048. , DOI 10.1038/s41467-018-03099-x Ronsley R, Rassekh SR, Shen Y, Lee AF, Jantzen C, Halparin J, Albert C, Hawkins DS, Amed S, Rothstein R, Mungall AJ, Dix D, Blair G, Nadel H, Jones SJM, Laskin J, Marra MA, JD R, 2018, Application of genomics to identify therapeutic targets in recurrent pediatric papillary thyroid carcinoma. Cold Spring Harb Mol Case Stud 4(2):a002568. , DOI 10.1101/mcs.a002568 Hartmaier RJ, Albacker LA, Chmielecki J, Bailey M, He J, Goldberg ME, Ramkissoon S, Suh J, Elvin JA, Chiacchia S, Frampton GM, Ross JS, Miller V, Stephens PJ, Lipson D, 2017, High-throughput genomic profiling of adult solid tumors reveals novel insights into Cancer pathogenesis. Cancer Res 77(9):2464– 2475. , DOI 10.1158/0008-5472.CAN-16-2479 Ross JS,Wang K, Gay L, Al-Rohil R, Rand JV, Jones DM, Lee HJ, Sheehan CE, Otto GA, Palmer G, Yelensky R, Lipson D, Morosini D,Hawryluk M, Catenacci DV, Miller VA, Churi C, Ali S, Stephens PJ, 2014, New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing. Oncologist 19(3):235–242. , DOI 10.1634/theoncologist. 2013-0352 Shi E, Chmielecki J, Tang CM, Wang K, Heinrich MC, Kang G, Corless CL, Hong D, Fero KE, Murphy JD, Fanta PT, Ali SM, De Siena M, Burgoyne AM, Movva S, Madlensky L, Heestand GM, Trent JC, Kurzrock R, Morosini D, Ross JS, Harismendy O, Sicklick JK, 2016, FGFR1 and NTRK3 actionable alterations in "wild-type" gastrointestinal stromal tumors. J Transl Med 14(1): 339. , DOI 10.1186/s12967-016-1075-6 Wiesner T, He J, Yelensky R, Esteve-Puig R, Botton T, Yeh I, Lipson D, Otto G, Brennan K, Murali R, Garrido M, Miller VA, Ross JS, Berger MF, Sparatta A, Palmedo G, Cerroni L, Busam KJ, Kutzner H, Cronin MT, Stephens PJ, Bastian BC, 2014, Kinase fusions are frequent in Spitz tumours and spitzoid melanomas. Nat Commun 5:3116. , DOI 10.1038/ncomms4116 Zheng Z, LiebersM, Zhelyazkova B, Cao Y, Panditi D, Lynch KD, Chen J, Robinson HE, Shim HS, Chmielecki J, Pao W, Engelman JA, Iafrate AJ, Le LP, 2014, Anchored multiplex PCR for targeted next-generation sequencing. Nat Med 20(12):1479–1484. https:// doi.org/10.1038/nm.3729 Liang J, CaiW, Feng D, Teng H, Mao F, JiangY, Hu S, Li X, Zhang Y, Liu B, Sun ZS, 2018, Genetic landscape of papillary thyroid carcinoma in the Chinese population. J Pathol 244(2):215–226. , DOI 10.1002/path.5005 Vaishnavi A, Capelletti M, Le AT, Kako S, Butaney M, Ercan D, Mahale S, Davies KD, Aisner DL, Pilling AB, Berge EM, Kim J, Sasaki H, Park S, Kryukov G, Garraway LA, Hammerman PS, Haas J, Andrews SW, Lipson D, Stephens PJ, Miller VA, Varella- Garcia M, Janne PA, Doebele RC, 2013, Oncogenic and drugsensitive NTRK1 rearrangements in lung cancer. Nat Med 19(11): 1469–1472. , DOI 10.1038/nm.3352 Chiang S, Cotzia P, Hyman DM, Drilon A, Tap WD, Zhang L, Hechtman JF, Frosina D, Jungbluth AA, Murali R, Park KJ, Soslow RA, Oliva E, Iafrate AJ, Benayed R, Ladanyi M, Antonescu CR, 2018, NTRK fusions define a novel uterine sarcoma subtype with features of Fibrosarcoma. Am J Surg Pathol 42(6): 791–798. , DOI 10.1097/PAS.0000000000001055 Tognon C, Garnett M, Kenward E, Kay R, Morrison K, Sorensen PHB, 2001, The chimeric protein tyrosine kinase ETV6-NTRK3 requires both Ras-Erk1/2 and PI3-kinase-Akt signaling for fibroblast transformation. Cancer Res 61(24):8909–8916 Tognon CE, Mackereth CD, Somasiri AM, McIntosh LP, Sorensen PHB, 2004, Mutations in the SAM domain of the ETV6-NTRK3 chimeric tyrosine kinase block polymerization and transformation activity. Mol Cell Biol 24(11):4636–4650. , DOI 10.1128/ mcb.24.11.4636-4650.2004 Li Z, Tognon CE, Godinho FJ, Yasaitis L, Hock H, Herschkowitz JI, Lannon CL, Cho E, Kim SJ, Bronson RT, Perou CM, Sorensen PH, Orkin SH, 2007, ETV6-NTRK3 fusion oncogene initiates breast cancer from committed mammary progenitors via activation of AP1 complex. Cancer Cell 12(6):542–558. , DOI 10. 1016/j.ccr.2007.11.012 Morrison KB, Tognon CE, Garnett MJ, Deal C, Sorensen PH, 2002, ETV6-NTRK3 transformation requires insulin-like growth factor 1 receptor signaling and is associated with constitutive IRS-1 tyrosine phosphorylation. Oncogene 21(37):5684–5695. https:// doi.org/10.1038/sj.onc.1205669 Martin MJ, Melnyk N, Pollard M, BowdenM, Leong H, Podor TJ, Gleave M, Sorensen PH, 2006, The insulin-like growth factor I receptor is required for Akt activation and suppression of anoikis in cells transformed by the ETV6-NTRK3 chimeric tyrosine kinase. Mol Cell Biol 26(5):1754–1769. , DOI 10.1128/MCB.26. 5.1754-1769.2006 Tognon CE, Rafn B, Cetinbas NM, Kamura T, Trigo G, Rotblat B, Okumura F, Matsumoto M, Chow C, Davare M, Pollak M, Mayor T, Sorensen PH, 2018, Insulin-like growth factor 1 receptor stabilizes the ETV6-NTRK3 chimeric oncoprotein by blocking its KPC1/Rnf123-mediated proteasomal degradation. J Biol Chem 293(32):12502–12515. , DOI 10.1074/jbc.RA117.000321 Kheder ES, Hong DS, 2018, Emerging targeted therapy for tumors with NTRK fusion proteins. Clin Cancer Res:Clincanres 1156: 2018. , DOI 10.1158/1078-0432.ccr-18-1156 Liu D, OffinM, Harnicar S, Li BT, Drilon A, 2018, Entrectinib: an orally available, selective tyrosine kinase inhibitor for the treatment of NTRK, ROS1, and ALK fusion-positive solid tumors. Ther Clin Risk Manag 14:1247–1252. , DOI 10.2147/TCRM. S147381 Ziegler DS, Wong M, Mayoh C, Kumar A, Tsoli M, Mould E, Tyrrell V, Khuong-Quang DA, Pinese M, Gayevskiy V, Cohn RJ, Lau LMS, ReynoldsM, CoxMC, Gifford A, RodriguezM, Cowley MJ, Ekert PG, Marshall GM, Haber M, 2018, Brief report: potent clinical and radiological response to larotrectinib in TRK fusiondriven high-grade glioma. Br J Cancer 119(6):693–696. https://doi. org/10.1038/s41416-018-0251-2 Laetsch TW, DuBois SG, Mascarenhas L, Turpin B, Federman N, Albert CM, Nagasubramanian R, Davis JL, Rudzinski E, Feraco AM, Tuch BB, Ebata KT, Reynolds M, Smith S, Cruickshank S, Cox MC, Pappo AS, Hawkins DS, 2018, Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: phase 1 results from a multicentre, open-label, phase 1/2 study. The Lancet Oncology 19(5):705–714. , DOI 10.1016/s1470-2045(18, 30119-0 U.S. Food & Drug Administration. Drugs@FDA: FDA Approved Drug Products. https://www.accessdata.fda.gov/scripts/cder/daf/ index.cfm?event=overview.process&varApplNo=210861. [Accessed 12 March 2019] Cui C, ShuW, Li P, 2016, Fluorescence in situ hybridization: cellbased genetic diagnostic and research applications. Frontiers in cell and developmental biology 4:89. , DOI 10.3389/fcell. 2016.00089 Argani P, FritschM, Kadkol SS, Schuster A, Beckwith JB, Perlman EJ, 2000, Detection of the ETV6-NTRK3 chimeric RNA of infantile fibrosarcoma/cellular congenital mesoblastic nephroma in paraffin-embedded tissue: application to challenging pediatric renal stromal tumors. Modern pathology : an official journal of the United States and Canadian academy of Pathology, Inc 13, 1):29– 36. , DOI 10.1038/modpathol.3880006 Kumar S, Razzaq SK, Vo AD, Gautam M, Li H, 2016, Identifying fusion transcripts using next generation sequencing.Wiley interdisciplinary reviews RNA 7(6):811–823. , DOI 10.1002/ wrna.1382 Doshi S, Ray D, Stein K, Zhang J, Koduru P, Fogt F, Wellman A, Wat R, Mathews C, 2016, Economic analysis of alternative strategies for detection of ALK rearrangements in non small cell lung Cancer. Diagnostics, Basel, 6(1). , DOI 10.3390/ diagnostics6010004 Davies KD, Le AT, Sheren J, Nijmeh H, Gowan K, Jones KL, Varella-Garcia M, Aisner DL, Doebele RC, 2018, Comparison of molecular testing modalities for detection of ROS1 rearrangements in a cohort of positive patient samples. J Thorac Oncol 13(10): 1474–1482. , DOI 10.1016/j.jtho.2018.05.041 Godbey P, Myles JL, Black-Schaffer WS, Scott J Pathology payment – An overview of the 2018 proposed Medicare physician fee schedule. College of American Pathologists https://documents.cap. org/documents/pathology-payment.pdf. [Accessed 12March 2019] Helman E, Nguyen M, Karlovich CA, Despain D, Choquette AK, Spira AI, Yu HA, Camidge DR, Harding TC, Lanman RB, Simmons AD, 2018, Cell-free DNA Next-generation sequencing prediction of response and resistance to third-generation EGFR inhibitor. Clin Lung Cancer 19(6):518–530 e517. , DOI 10. 1016/j.cllc.2018.07.008 Bubendorf L, Buttner R, Al-Dayel F, Dietel M, Elmberger G, Kerr K, Lopez-Rios F, Marchetti A, Oz B, Pauwels P, Penault-Llorca F, Rossi G, Ryska A, Thunnissen E, 2016, Testing for ROS1 in nonsmall cell lung cancer: a review with recommendations. Virchows Arch 469(5):489–503. , DOI 10.1007/s00428-016-2000-3 Cheng DT, Mitchell TN, Zehir A, Shah RH, Benayed R, Syed A, Chandramohan R, Liu ZY, Won HH, Scott SN, Brannon AR, O'Reilly C, Sadowska J, Casanova J, Yannes A, Hechtman JF, Yao J, Song W, Ross DS, Oultache A, Dogan S, Borsu L, Hameed M, Nafa K, Arcila ME, Ladanyi M, Berger MF, 2015, Memorial Sloan Kettering-integrated mutation profiling of actionable Cancer targets, MSK-IMPACT). a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology J Mol Diagn 17(3):251–264. , DOI 10.1016/j. jmoldx.2014.12.006 Mullin E Next Generation sequencing in the clinical laboratory: in the trenches with early adopters. https://www.aacc.org/publications/ cln/articles/2015/may/next-generation-sequencing-in-the-clinicallaboratory. [Accessed 12 March 2019] Sabari JK, Santini F, Bergagnini I, Lai WV, Arbour KC, Drilon A, 2017, Changing the therapeutic landscape in non-small cell lung cancers: the evolution of comprehensive molecular profiling improves access to therapy. Curr Oncol Rep 19(4):24. https://doi. org/10.1007/s11912-017-0587-4 Paasinen-Sohns A, Koelzer VH, Frank A, Schafroth J, Gisler A, Sachs M, Graber A, Rothschild SI, Wicki A, Cathomas G, Mertz KD, 2017, Single-center experience with a targeted Next generation sequencing assay for assessment of relevant somatic alterations in solid tumors. Neoplasia 19(3):196–206. , DOI 10.1016/ j.neo.2017.01.003 Wegert J, Vokuhl C, Collord G, Del Castillo Velasco-Herrera M, Farndon SJ, Guzzo C, Jorgensen M, Anderson J, Slater O, Duncan C, Bausenwein S, Streitenberger H, Ziegler B, Furtwangler R, Graf N, Stratton MR, Campbell PJ, Jones DT, Koelsche C, Pfister SM, Mifsud W, Sebire N, Sparber-Sauer M, Koscielniak E, Rosenwald A, Gessler M, Behjati S, 2018, Recurrent intragenic rearrangements of EGFR and BRAF in soft tissue tumors of infants. Nat Commun 9(1):2378. , DOI 10.1038/s41467-018-04650-6 van den Akker J, Mishne G, Zimmer AD, Zhou AY, 2018, A machine learning model to determine the accuracy of variant calls in capture-based next generation sequencing. BMC Genomics 19(1):263. , DOI 10.1186/s12864-018-4659-0 Tkachuk DC,Westbrook CA, Andreeff M, Donlon TA, ClearyML, Suryanarayan K, Homge M, Redner A, Gray J, Pinkel D, 1990, Detection of bcr-abl fusion in chronic myelogeneous leukemia by in situ hybridization. Science 250(4980):559–562 Doebele RC, Davis LE, Vaishnavi A, Le AT, Estrada-Bernal A, Keysar S, Jimeno A, Varella-Garcia M, Aisner DL, Li Y, Stephens PJ, Morosini D, Tuch BB, Fernandes M, Nanda N, Low JA, 2015, An oncogenic NTRK fusion in a patient with soft-tissue sarcoma with response to the tropomyosin-related kinase inhibitor LOXO-101. Cancer Discov 5(10):1049–1057. , DOI 10. 1158/2159-8290.Cd-15-0443 Lyu X, Wang X, Zhang L, Chen Z, Zhao Y, Hu J, Fan R, Song Y, 2017, Detection of 22 common leukemic fusion genes using a single-step multiplex qRT-PCR-based assay. Diagn Pathol 12(1): 55. , DOI 10.1186/s13000-017-0634-3 Wu YC, Chang IC,Wang CL, Chen TD, Chen YT, Liu HP, Chu Y, Chiu YT, Wu TH, Chou LH, Chen YR, Huang SF, 2013, Comparison of IHC, FISH and RT-PCR methods for detection of ALK rearrangements in 312 non-small cell lung cancer patients in Taiwan. PLoS One 8(8):e70839. , DOI 10.1371/journal. pone.0070839 Beadling C,Wald AI,Warrick A, Neff TL, Zhong S, Nikiforov YE, Corless CL, Nikiforova MN, 2016, A multiplexed amplicon approach for detecting gene fusions by Next-generation sequencing. J Mol Diagn 18(2):165–175. , DOI 10.1016/j.jmoldx. 2015.10.002 Fehr A, Loning T, Stenman G, 2011)Mammary analogue secretory carcinoma of the salivary glands with ETV6-NTRK3 gene fusion. Am J Surg Pathol 35(10):1600–1602. , DOI 10.1097/PAS. 0b013e31822832c7 Gatalica Z, Xiu J, Swensen J, Vranic S, 2018, Molecular characterization of cancers with NTRK gene fusions. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc , DOI 10.1038/s41379-018-0118-3 Zhang J,WhiteNM, SchmidtHK, Fulton RS, Tomlinson C,Warren WC, Wilson RK, Maher CA, 2016, INTEGRATE: gene fusion discovery using whole genome and transcriptome data. Genome Res 26(1):108–118. , DOI 10.1101/gr.186114.114 Maher CA, Kumar-Sinha C, Cao X, Kalyana-Sundaram S, Han B, Jing X, Sam L, Barrette T, Palanisamy N, Chinnaiyan AM, 2009, Transcriptome sequencing to detect gene fusions in cancer. Nature 458(7234):97–101. , DOI 10.1038/nature07638 Farago AF, Le LP, Zheng Z, Muzikansky A, Drilon A, Patel M, Bauer TM, Liu SV, Ou SH, Jackman D, Costa DB, Multani PS, Li GG, Hornby Z, Chow-Maneval E, Luo D, Lim JE, Iafrate AJ, Shaw AT, 2015, Durable clinical response to entrectinib in NTRK1- rearranged non-small cell lung cancer. J Thorac Oncol 10(12): 1670–1674. , DOI 10.1097/01.JTO.0000473485.38553.f0 Alvarez-Breckenridge C, Miller JJ, Nayyar N, Gill CM, Kaneb A, D'Andrea M, Le LP, Lee J, Cheng J, Zheng Z, Butler WE, Multani P, Chow Maneval E, Ha Paek S, Toyota BD, Dias-Santagata D, Santagata S, Romero J, Shaw AT, Farago AF, Yip S, Cahill DP, Batchelor TT, Iafrate AJ, Brastianos PK, 2017, Clinical and radiographic response following targeting of BCAN-NTRK1 fusion in glioneuronal tumor. NPJ Precis Oncol 1(1):5. , DOI 10. 1038/s41698-017-0009-y Murphy DA, Ely HA, Shoemaker R, Boomer A, Culver BP, Hoskins I, Haimes JD, Walters RD, Fernandez D, Stahl JA, Lee J, Kim KM, Lamoureux J, Christiansen J, 2017, Detecting gene rearrangements in patient populations through a 2-step diagnostic test comprised of rapid IHC enrichment followed by sensitive nextgeneration sequencing. Appl Immunohistochem Mol Morphol 25(7):513–523. , DOI 10.1097/PAI.0000000000000360 Qadir MA, Zhan SH, Kwok B, Bruestle J, Drees B, Popescu OE, Sorensen PH, 2014, ChildSeq-RNA. a next-generation sequencingbased diagnostic assay to identify known fusion transcripts in childhood sarcomas J Mol Diagn 16(3):361–370. , DOI 10. 1016/j.jmoldx.2014.01.002 Chang KTE, Goytain A, Tucker T, Karsan A, Lee CH, Nielsen TO, Ng TL, 2018, Development and evaluation of a pan-sarcoma fusion gene detection assay using the NanoString nCounter platform. J Mol Diagn 20(1):63–77. , DOI 10.1016/j.jmoldx.2017.09.007 Lindquist KE, Karlsson A, Leveen P, Brunnstrom H, Reutersward C, Holm K, Jonsson M, Annersten K, Rosengren F, Jirstrom K, Kosieradzki J, Ek L, Borg A, Planck M, Jonsson G, Staaf J, 2017, Clinical framework for next generation sequencing based analysis of treatment predictive mutations and multiplexed gene fusion detection in non-small cell lung cancer. Oncotarget 8(21): 34796–34810. , DOI 10.18632/oncotarget.16276 Hyman DM, Solit DB, Arcila ME, Cheng DT, Sabbatini P, Baselga J, Berger MF, Ladanyi M, 2015, Precision medicine at memorial Sloan Kettering Cancer center: clinical next-generation sequencing enabling next-generation targeted therapy trials. Drug Discov Today 20(12):1422–1428. , DOI 10.1016/j.drudis.2015. 08.005 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1385 EP 1399 DI 10.1007/s12253-019-00685-2 PG 15 ER PT J AU Li, Z Liu, J Zhang, X Fang, L Zhang, C Zhang, Z Yan, L Tang, Y Fan, Y AF Li, Zeyan Liu, Jikai Zhang, Xiang Fang, Liang Zhang, Cong Zhang, Zhao Yan, Lei Tang, Yueqing Fan, Yidong TI Prognostic Significance of Cyclin D1 Expression in Renal Cell Carcinoma: a Systematic Review and Meta-analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE CyclinD1; Renal cell carcinoma; Prognosis; Meta-analysis ID CyclinD1; Renal cell carcinoma; Prognosis; Meta-analysis AB Previous studies indicated that cyclin D1 shown the potential as a tumor biomarker. However, the prognostic value of cyclin D1 in renal cell carcinoma (RCC) remains controversial. This study investigated the correlation of cyclin D1 expression with the prognostic and clinicopathological features in RCC patients. We systematically searched the database of PubMed, Embase, Cochrane, and Web of Science updated on November 26, 2017. Eighteen studies with 2282 patients satisfied the inclusion criteria. Results demonstrated that cyclin D1 overexpression in RCC showed significant favorable prognostic impact on disease-free survival (DFS) (HR 0.57, 95% CI: 0.43–0.74) and disease-specific survival (DSS) (HR 0.59, 95% CI 0.41–0.85) without significant heterogeneity. In subgroup of clear cell RCC, the prognostic effect on DFS was robust and the pooled HR was 0.39 (95% CI: 0.27–0.57). However, no association between overall survival (OS) and cyclin D1 expression was observed. Stratified analysis in DFS studies by sample size, staining patterns race and metastasis status showed similar results. Otherwise, cyclin D1 overexpression predicted a reduced prevalence of high TNM stage (T3 + T4) (OR 0.63, 95% CI: 0.40–0.99), high-grade tumor (G3 + G4) (OR 0.51, 95% CI: 0.31–0.81) and large tumor size (OR 0.35, 95% CI: 0.19–0.62). Our meta-analysis indicated that cyclin D1 overexpression could predict the favorable prognosis in patients with RCC. C1 [Li, Zeyan] Qilu Hospital of Shandong University, Department of Urology, Wenhuaxi Road 44, 250012 Jinan, Shandong, China. [Liu, Jikai] Qilu Hospital of Shandong University, Department of Urology, Wenhuaxi Road 44, 250012 Jinan, Shandong, China. [Zhang, Xiang] Qilu Hospital of Shandong University, Department of Urology, Wenhuaxi Road 44, 250012 Jinan, Shandong, China. [Fang, Liang] Qilu Hospital of Shandong University, Department of Urology, Wenhuaxi Road 44, 250012 Jinan, Shandong, China. [Zhang, Cong] Qilu Hospital of Shandong University, Department of Urology, Wenhuaxi Road 44, 250012 Jinan, Shandong, China. [Zhang, Zhao] Qilu Hospital of Shandong University, Department of Urology, Wenhuaxi Road 44, 250012 Jinan, Shandong, China. [Yan, Lei] Qilu Hospital of Shandong University, Department of Urology, Wenhuaxi Road 44, 250012 Jinan, Shandong, China. [Tang, Yueqing] Qilu Hospital of Shandong University, Department of Urology, Wenhuaxi Road 44, 250012 Jinan, Shandong, China. [Fan, Yidong] Qilu Hospital of Shandong University, Department of Urology, Wenhuaxi Road 44, 250012 Jinan, Shandong, China. RP Fan, Y (reprint author), Qilu Hospital of Shandong University, Department of Urology, 250012 Jinan, China. EM fanyd@sdu.edu.cn CR Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61(2):69–90. https://doi. org/10.3322/caac.20107 MacLennan S, Imamura M, Lapitan MC, Omar MI, Lam TB, Hilvano-Cabungcal AM, Royle P, Stewart F, MacLennan G, MacLennan SJ, Canfield SE, McClinton S, Griffiths TR, Ljungberg B, N’Dow J, 2012, Systematic review of oncological outcomes following surgical management of localised renal cancer. Eur Urol 61(5):972–993. , DOI 10.1016/j.eururo. 2012.02.039 Ljungberg B,Mehle C, Stenling R, Roos G, 1996, Heterogeneity in renal cell carcinoma and its impact no prognosis–a flow cytometric study. Br J Cancer 74(1):123–127 Diehl JA, 2002, Cycling to cancer with cyclin D1. Cancer Biol Ther 1(3):226–231 Shan J, ZhaoW, GuW(2009, Suppression of cancer cell growth by promoting cyclin D1 degradation.Mol Cell 36(3):469–476. https:// doi.org/10.1016/j.molcel.2009.10.018 Palmqvist R, Stenling R, Oberg A, Landberg G, 1998, Expression of cyclin D1 and retinoblastoma protein in colorectal cancer. Eur J Cancer, Oxford, England : 1990, 34(10):1575–1581 Aaltomaa S, Lipponen P, Ala-Opas M, Eskelinen M, Syrjanen K, Kosma VM, 1999, Expression of cyclins a and D and p21(waf1/cip1, proteins in renal cell cancer and their relation to clinicopathological variables and patient survival. Br J Cancer 80(12):2001–2007. , DOI 10.1038/sj.bjc.6690634 Hedberg Y, Ljungberg B, Roos G, Landberg G, 2003, Expression of cyclin D1, D3, E, and p27 in human renal cell carcinoma analysed by tissue microarray. Br J Cancer 88(9):1417–1423. , DOI 10.1038/sj.bjc.6600922 Xu XL, Chen SZ, Chen W, Zheng WH, Xia XH, Yang HJ, Li B, Mao WM, 2013, The impact of cyclin D1 overexpression on the prognosis of ER-positive breast cancers: a meta-analysis. Breast Cancer Res Treat 139(2):329–339. , DOI 10.1007/ s10549-013-2563-5 Li Y,Wei J, Xu C, Zhao Z, You T, 2014, Prognostic significance of cyclin D1 expression in colorectal cancer: a meta-analysis of observational studies. PLoS One 9(4):e94508. , DOI 10.1371/ journal.pone.0094508 Ren B, Li W, Yang Y, Wu S, 2014, The impact of cyclin D1 overexpression on the prognosis of bladder cancer: a meta-analysis. World J Surg Oncol 12:55. , DOI 10.1186/1477-7819-12- 55 Zhang LQ, Jiang F, Xu L,Wang J, Bai JL, Yin R,Wu YQ, Meng LJ, 2012, The role of cyclin D1 expression and patient's survival in non-small-cell lung cancer: a systematic review with meta-analysis. Clin Lung Cancer 13(3):188–195. , DOI 10.1016/j.cllc. 2011.10.003 Tobin NP, Sims AH, Lundgren KL, Lehn S, Landberg G, 2011, Cyclin D1, Id1 and EMT in breast cancer. BMC Cancer 11:417. , DOI 10.1186/1471-2407-11-417 Ishii Y, Pirkmaier A, Alvarez JV, FrankDA,Keselman I, Logothetis D, Mandeli J, O’ConnellMJ,Waxman S, Germain D, 2006, Cyclin D1 overexpression and response to bortezomib treatment in a breast cancer model. J Natl Cancer Inst 98(17):1238–1247. , DOI 10.1093/jnci/djj334 Moher D, Liberati A, Tetzlaff J, Altman DG, 2009, Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. J Clin Epidemiol 62(10):1006–1012. https:// doi.org/10.1016/j.jclinepi.2009.06.005 Furukawa J,Miyake H, FujisawaM(2016, GLI2 expression levels in radical nephrectomy specimens as a predictor of disease progression in patients with metastatic clear cell renal cell carcinoma following treatment with sunitinib. Mol Clin Oncol 5(3):186–192. , DOI 10.3892/mco.2016.950 HaddadAQ, Luo JH,Krabbe LM,Darwish O, Gayed B,Youssef R, Kapur P, Rakheja D, Lotan Y, Sagalowsky A, Margulis V, 2017, Prognostic value of tissue-based biomarker signature in clear cell renal cell carcinoma. BJU Int 119(5):741–747. , DOI 10. 1111/bju.13776 Chaves Portela S, Santos-Arrontes D, Fernandez-Acenero MJ, Garcia Gonzalez J, Paniagua P, 2011, Stage pT3a of renal clear cell carcinoma: do tumors with sinus fat involvement behave the same as those with perinephric fat involvement? Rom J Morphol Embryol = Revue Roumaine deMorphologie et EMBRYOLOGIE 52(2):569–574 Lima MS, Pereira RA, Costa RS, Tucci S, Dantas M, Muglia VF, Ravinal RC, Barros-Silva GE, 2014, The prognostic value of cyclin D1 in renal cell carcinoma. Int Urol Nephrol 46(5):905–913. https:// doi.org/10.1007/s11255-013-0602-0 Biswas S, Charlesworth PJ, Turner GD, Leek R, Thamboo PT, Campo L, Turley H, Dildey P, Protheroe A, Cranston D, Gatter KC, Pezzella F, Harris AL, 2012, CD31 angiogenesis and combined expression of HIF-1alpha and HIF-2alpha are prognostic in primary clear-cell renal cell carcinoma, CC-RCC), but HIFalpha transcriptional products are not: implications for antiangiogenic trials and HIFalpha biomarker studies in primary CC-RCC. Carcinogenesis 33(9):1717–1725. , DOI 10.1093/carcin/ bgs222 Phuoc NB, Ehara H, Gotoh T, Nakano M, Yokoi S, Deguchi T, Hirose Y, 2007, Immunohistochemical analysis with multiple antibodies in search of prognostic markers for clear cell renal cell carcinoma. Urology 69(5):843–848. , DOI 10.1016/j. urology.2007.01.069 Allory Y, Matsuoka Y, Bazille C, Christensen EI, Ronco P, Debiec H, 2005, The L1 cell adhesion molecule is induced in renal cancer cells and correlates with metastasis in clear cell carcinomas. Clin Cancer Res : an Official Journal of the American Association for Cancer Research 11(3):1190–1197 Stubbs C, Bardoli AD, Afshar M, Pirrie S, Miscoria M, Wheeley I, Porfiri E, 2017, A study of angiogenesis markers in patients with renal cell carcinoma undergoing therapy with Sunitinib. Anticancer Research 37(1):253–259. , DOI 10.21873/ anticanres.11315 Kusuda Y, Miyake H, Behnsawy HM, Fukuhara T, Inoue TA, FujisawaM(2013, Prognostic prediction in patients withmetastatic renal cell carcinoma treated with sorafenib based on expression levels of potential molecular markers in radical nephrectomy specimens. Urol Oncol 31(1):42–50. , DOI 10.1016/j. urolonc.2010.09.008 Bodnar L, Stec R, Cierniak S, Synowiec A, Wcislo G, Jesiotr M, Koktysz R, Kozlowski W, Szczylik C, 2015, Clinical usefulness of PI3K/Akt/mTOR genotyping in companion with other clinical variables in metastatic renal cell carcinoma patients treated with everolimus in the second and subsequent lines. Ann Oncol : OFFICIAL journal of the European Society for Medical Oncology 26(7):1385–1389. , DOI 10.1093/annonc/ mdv166 Su T, Han Y, Yu Y, Tan X, Li X, Hou J, Du Y, Shen J,Wang G, Ma L, Jiang S, Zhang H, Cao G, 2013, A GWAS-identified susceptibility locus on chromosome 11q13.3 and its putative molecular target for prediction of postoperative prognosis of human renal cell carcinoma. Oncol Lett 6(2):421–426. , DOI 10.3892/ol. 2013.1422 Dahinden C, Ingold B, Wild P, Boysen G, Luu VD, Montani M, Kristiansen G, Sulser T, Buhlmann P, Moch H, Schraml P, 2010, Mining tissuemicroarray data to uncover combinations of biomarker expression patterns that improve intermediate staging and grading of clear cell renal cell cancer. Clin Cancer Res : an Official Journal of the American Association for Cancer Research 16(1): 88–98. , DOI 10.1158/1078-0432.ccr-09-0260 Wang F, Guo CH, Sun W, Dong Y, Yang Z, 2012, Expressions of E-cadherin, β-catenin and Cyclin D1 in renal clear cell carcinoma. Chinese Journal of Cancer Prevention and Treatment 19(22): 1716–1718+1729 Migita T, Oda Y, Naito S, Tsuneyoshi M, 2002, Low expression of p27(Kip1, is associated with tumor size and poor prognosis in patients with renal cell carcinoma. Cancer 94(4):973–979 Ge JR, Ren GP, Yao HP, 2007, Expression of cyclin D1 and p27kip1 in renal cell carcinoma and its significance. Zhejiang Da Xue Xue Bao Yi Xue Ban = Journal of Zhejiang University Medical sciences 36(5):483–487 Hsu RJ, Ho JY, Cha TL, Yu DS,Wu CL, Huang WP, Chu P, Chen YH, Chen JT, Yu CP, 2012, WNT10A plays an oncogenic role in renal cell carcinoma by activating WNT/beta-catenin pathway. PLoS One 7(10):e47649. , DOI 10.1371/journal.pone. 0047649 Wells G, 2004, The Newcastle-Ottawa scale, NOS, for assessing the quality of nonrandomised studies in meta-analysis. Cancer. 2002 Feb 15;94(4):973-9. http://www.ohrica/programs/clinical_ epidemiologyoxford.htm Stroup DF, Berlin JA,Morton SC, Olkin I,Williamson GD, Rennie D, Moher D, Becker BJ, Sipe TA, Thacker SB, 2000, Metaanalysis of observational studies in epidemiology: a proposal for reporting. Meta-analysis of observational studies in epidemiology, MOOSE, group. JAMA 283(15):2008–2012 Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR, 2007, Practical methods for incorporating summary time-to-event data into meta-analysis. Trials 8:16. , DOI 10.1186/1745- 6215-8-16 Strauss M, Lukas J, Bartek J, 1995, Unrestricted cell cycling and cancer. Nat Med 1(12):1245–1246 Jirawatnotai S, Hu Y,MichowskiW, Elias JE, Becks L, Bienvenu F, Zagozdzon A, Goswami T, Wang YE, Clark AB, Kunkel TA, van Harn T, Xia B, Correll M, Quackenbush J, Livingston DM, Gygi SP, Sicinski P, 2011, A function for cyclin D1 in DNA repair uncovered by protein interactome analyses in human cancers. Nature 474(7350):230–234. , DOI 10.1038/nature10155 Han EK, Sgambato A, Jiang W, Zhang YJ, Santella RM, Doki Y, Cacace AM, Schieren I,Weinstein IB, 1995, Stable overexpression of cyclin D1 in a human mammary epithelial cell line prolongs the S-phase and inhibits growth. Oncogene 10(5):953–961 Parry D, Bates S, Mann DJ, Peters G, 1995, Lack of cyclin D-Cdk complexes in Rb-negative cells correlates with high levels of p16INK4/MTS1 tumour suppressor gene product. EMBO J 14(3):503–511 Atadja P, Wong H, Veillete C, Riabowol K, 1995, Overexpression of cyclin D1 blocks proliferation of normal diploid fibroblasts. Exp Cell Res 217(2):205–216. , DOI 10.1006/excr.1995.1080 Freeman RS, Estus S, Johnson EMJr, 1994, Analysis of cell cyclerelated gene expression in postmitotic neurons: selective induction of Cyclin D1 during programmed cell death. Neuron 12(2):343– 355. , DOI 10.1016/0896-6273(94)90276-3 Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP, 1998, Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4(7):844–847 Simon RM, Paik S, Hayes DF, 2009, Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst 101(21):1446–1452. , DOI 10.1093/jnci/djp335 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1401 EP 1409 DI 10.1007/s12253-019-00776-0 PG 9 ER PT J AU Travaglino, A Raffone, A Mascolo, M Guida, M Insabato, L Zannoni, FG Zullo, F AF Travaglino, Antonio Raffone, Antonio Mascolo, Massimo Guida, Maurizio Insabato, Luigi Zannoni, Franco Gian Zullo, Fulvio TI TCGA Molecular Subgroups in Endometrial Undifferentiated/Dedifferentiated Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Cancer; Treatment; Endometrium; Risk assessment; PROMISE ID Cancer; Treatment; Endometrium; Risk assessment; PROMISE AB We aimed to classify undifferentiated/dedifferentiated carcinoma (UDC/DDC) according to the four TCGA molecular subgroups of endometrial cancer: microsatellite-instable/hypermutated (MSI), POLE-mutant/ultramutated (POLE), copy-number-low/p53- wild-type (p53wt), and copy-number-high/p53-abnormal (p53abn), through a systematic review and meta-analysis. Electronic databases were searched from January 2013 to July 2019 for studies assessing the TCGA classification in endometrial UDC/ DDC series. Pooled prevalence of each TCGA subgroup on the total UDC/DDCs was calculated. Three studies with 73 patients were included. Pooled prevalence of the TCGA subgroups were: 12.4% for the POLE subgroup, 44% for the MSI subgroup, 18.6%for the p53abn subgroup, 25%for the p53wt group. All TCGA groups are represented in UDC/DDC, with a predominance of the MSI group, indicating a biological heterogeneity. Hypermutated/ultramutated cancers constitute the majority of UDC/ DDC, suggesting a crucial difference with other high-risk histologies of endometrial carcinoma. C1 [Travaglino, Antonio] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology UnitNaples, Italy. [Raffone, Antonio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Mascolo, Massimo] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology UnitNaples, Italy. [Guida, Maurizio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Insabato, Luigi] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology UnitNaples, Italy. [Zannoni, Franco Gian] Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic, Pathology Unit, Department of Woman and Child HealthRome, Italy. [Zullo, Fulvio] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology UnitNaples, Italy. RP Raffone, A (reprint author), University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, 80131 Naples, Italy. EM anton.raffone@gmail.com CR Siegel RL, Miller KD, Jemal A, 2015, Cancer statistics, 2015. CA Cancer J Clin 65(1):5–29 Travaglino A, Raffone A, Saccone G et al, 2019, Immunohistochemical nuclear expression of β-catenin as a surrogate of CTNNB1 exon 3 mutation in endometrial Cancer. Am J Clin Pathol 151(5):529–538 Raffone A, Travaglino A,MascoloMet al, 2019, TCGAmolecular groups of endometrial cancer: Pooled data about prognosis. Gynecol Oncol. , DOI 10.1016/j.ygyno.2019.08.019 Raffone A, Travaglino A, Santoro A et al, 2019, Accuracy of onestep nucleic acid amplification in detecting lymph node metastases in endometrial Cancer. Pathol Oncol Res. , DOI 10.1007/ s12253-019-00727-9 Travaglino A, Raffone A, Saccone G et al, 2019, Immunohistochemical predictive markers of response to conservative treatment of endometrial hyperplasia and early endometrial cancer: a systematic review. Acta Obstet Gynecol Scand. , DOI 10.1111/aogs.13587 Cancer Genome Atlas Research Network et al, 2013, Integrated genomic characterization of endometrial carcinoma. Nature 497(7447):67–73 Stelloo E, Nout RA, Osse EM et al, 2016, Improved risk assessment by integrating molecular and clinicopathological factors in early-stage endometrial cancer-combined analysis of the PORTEC cohorts. Clin Cancer Res 22(16):4215–4224 Bosse T, Nout RA, McAlpine JN et al, 2018, Molecular classification of grade 3 endometrioid endometrial cancers identifies distinct prognostic subgroups. Am J Surg Pathol 42(5):561–568 Cosgrove CM, Tritchler DL, Cohn DE et al, 2018 Jan, An NRG oncology/GOG study of molecular classification for risk prediction in endometrioid endometrial cancer. GynecolOncol. 148(1):174– 180 Talhouk A, McConechy MK, Leung S et al, 2015, A clinically applicable molecular-based classification for endometrial cancers. Br J Cancer 113(2):299–310 Talhouk A,McConechyMK, Leung S et al, 2017, Confirmation of ProMisE: a simple, genomics-based clinical classifier for endometrial cancer. Cancer. 123(5):802–813 Kommoss S, McConechy MK, Kommoss F et al, 2018 May 1, Final validation of the ProMisEmolecular classifier for endometrial carcinoma in a large population-based case series. AnnOncol. 29(5):1180–1188 Hoang LN, Kinloch MA, Leo JM et al, 2017 Feb, Interobserver agreement in endometrial carcinoma Histotype diagnosis varies depending on the Cancer genome atlas, TCGA)-based molecular subgroup. Am J Surg Pathol 41(2):245–252 Raffone A, Travaglino A, Saccone G et al, 2019, Management of women with atypical polypoid adenomyoma of the uterus: a quantitative systematic review. Acta Obstet Gynecol Scand. https://doi. org/10.1111/aogs.13553 Lionetti R, De Luca M, Travaglino A et al, 2019 Jul, Treatments and overall survival in patients with Krukenberg tumor. Arch Gynecol Obstet 300(1):15–23 Travaglino A, Raffone A, Saccone G et al, 2019, Nuclear expression of β-catenin in endometrial hyperplasia as marker of premalignancy. APMIS. , DOI 10.1111/apm.12988 Raffone A, Travaglino A, Saccone G et al, 2019, Should progesterone and estrogens receptors be assessed for predicting the response to conservative treatment of endometrial hyperplasia and cancer? A systematic review and meta-analysis. Acta Obstet Gynecol Scand. , DOI 10.1111/aogs.13586 Travaglino A, Raffone A, Saccone A et al, 2019, Immunophenotype of atypical Polypoid Adenomyoma of the uterus: diagnostic value and insight on pathogenesis. Appl Immunohistochem Mol Morphol. , DOI 10.1097/PAI. 0000000000000780 Moher D, Shamseer L, Clarke M et al, 2015, Preferred reporting items for systematic review and meta-analysis protocols, PRISMAP, 2015 statement. Systematic Reviews 4:1 Whiting PF, Rutjes AW,WestwoodME et al, 2011, QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med 155(8):529–536 Travaglino A, Raffone A, Saccone G et al, 2019, Congruence between 1994 WHO classification of endometrial hyperplasia and endometrial intraepithelial neoplasia system. Am J Clin Pathol. , DOI 10.1093/ajcp/aqz132 Raffone A, Travaglino A, Saccone G et al, 2019, PTEN expression in endometrial hyperplasia and risk of cancer: a systematic review and meta-analysis. Arch Gynecol Obstet 299(6):1511–1524 Travaglino A, Raffone A, Mascolo M et al, 2019, Clear cell endometrial carcinoma and the TCGA classification. Histopathology. , DOI 10.1111/his.13976 Travaglino A, Raffone A, Saccone G et al, 2019, PTEN immunohistochemistry in endometrial hyperplasia: which are the optimal criteria for the diagnosis of precancer? APMIS 127(4):161–169 Raffone A, Travaglino A, Saccone G et al, 2019, Endometrial hyperplasia and progression to cancer: which classification system stratifies the risk better? A systematic review and meta-analysis. Arch Gynecol Obstet 299(5):1233–1242 Travaglino A, Raffone A, Saccone G et al, 2019, Significant risk of occult cancer in complex non-atypical endometrial hyperplasia. Arch Gynecol Obstet. , DOI 10.1007/s00404-019-05299-2 Raffone A, Travaglino A, Saccone G et al, 201, Diabetesmellitus is associated with occult cancer in endometrial hyperplasia. Pathol Oncol Res. , DOI 10.1007/s12253-019-00684-3 Raffone A, Travaglino A, Saccone G et al, 2019, Diagnostic and prognostic value of ARID1A in endometrial hyperplasia: a novel marker of occult cancer. APMIS. , DOI 10.1111/apm. 12977 Raffone A, Travaglino A, Saccone G et al, 2019, Diabetes mellitus and responsiveness of endometrial hyperplasia and early endometrial cancer to conservative treatment. Gynecol Endocrinol 5:1–6. , DOI 10.1080/09513590.2019.1624716 Meng B, Hoang LN, McIntyre JB et al, 2014, POLE exonuclease domain mutation predicts long progression-free survival in grade 3 endometrioid carcinoma of the endometrium. Gynecol Oncol 134(1):15–19 Stewart CJ, Crook ML, 2015, SWI/SNF complex deficiency and mismatch repair protein expression in undifferentiated and dedifferentiated endometrial carcinoma. Pathology. 47(5):439–445 Coatham M, Li X, Karnezis AN et al, 2016, Concurrent ARID1A and ARID1B inactivation in endometrial and ovarian dedifferentiated carcinomas. Mod Pathol 29(12):1586–1593 Ramalingam P, Croce S, McCluggage WG, 2017, Loss of expression of SMARCA4, BRG1), SMARCA2, BRM, and SMARCB1, INI1, in undifferentiated carcinoma of the endometrium is not uncommon and is not always associated with rhabdoid morphology. Histopathology. 70(3):359–366 Rosa-Rosa JM, Leskela S, Cristobal-Lana E et al, 2016, Molecular genetic heterogeneity in undifferentiated endometrial carcinomas. Mod Pathol 29(11):1390–1398 Espinosa I, Lee CH, D’Angelo E, Palacios J, Prat J, 2017, Undifferentiated and dedifferentiated endometrial carcinomas with POLE exonuclease domain mutations have a favorable prognosis. Am J Surg Pathol 41(8):1121–1128 Kobel M, Hoang LN, Tessier-Cloutier B et al, 2018, Undifferentiated endometrial carcinomas show frequent loss of core switch/sucrose nonfermentable complex proteins. Am J Surg Pathol 42(1):76–83 Murali R, Davidson B, Fadare O et al, 2019, High-grade endometrial carcinomas: morphologic and Immunohistochemical features, diagnostic challenges and recommendations. Int J Gynecol Pathol 38 Suppl 1:S40–S63 Silverberg SG, Nogales F, Tavassoli FA, Devilee P, eds,, 2003, Tumours of the uterine corpus. Pathology and genetics: Tumours of the breast and female genital organs. IARC Press. World Health Organization Classification of Tumours, Lyon, pp 217–257 Zaino R, Carinelli SG, Eng C, Kurman RJ, Carcangiu ML, Herrington CS, Young RH, eds,, 2014, Tumours of the uterine corpus. WHO classification of tumours of female reproductive organs. IARC Press. World Health Organization Classification of Tumours, Lyon, pp 121–154 Karnezis AN, Hoang LN, Coatham M et al, 2016, Loss of switch/ sucrose non-fermenting complex protein expression is associated with dedifferentiation in endometrial carcinomas. Mod Pathol 29: 302–314 Abu-Rustum NR, Yashar CM, Bean S, et al, 2019, NCCN clinical practice guidelines in oncology, NCCN Guidelines®, – Uterine Neoplasms. Version 3.Feb 11 2019 Stelloo E, Bosse T, Nout RA et al, 2015, Refining prognosis and identifying targetable pathways for high-risk endometrial cancer; a TransPORTEC initiative. Mod Pathol 28(6):836–844 Cherniack AD, Shen H, Walter Vet al, 2017, Integrated molecular characterization of uterine Carcinosarcoma. Cancer Cell 31(3):411– 423 Hacking S, Jin C, Komforti M, Liang S, Nasim M, 2019, MMR deficient undifferentiated/dedifferentiated endometrial carcinomas showing significant programmed death ligand-1 expression, sp 142, with potential therapeutic implications. Pathol Res Pract 22: 152552. , DOI 10.1016/j.prp.2019.152552 GoodmanAM,Kato S, Bazhenova L et al, 2017, Tumormutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther 16(11):2598–2608 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1411 EP 1416 DI 10.1007/s12253-019-00784-0 PG 6 ER PT J AU Raffone, A Travaglino, A Cerbone, M Gencarelli, A Mollo, A Insabato, L Zullo, F AF Raffone, Antonio Travaglino, Antonio Cerbone, Marco Gencarelli, Annarita Mollo, Antonio Insabato, Luigi Zullo, Fulvio TI Diagnostic Accuracy of Immunohistochemistry for Mismatch Repair Proteins as Surrogate of Microsatellite Instability Molecular Testing in Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Mismatch repair; Microsatellite instability; Endometrial cancer; Risk assessment; ProMisE; TCGA ID Mismatch repair; Microsatellite instability; Endometrial cancer; Risk assessment; ProMisE; TCGA AB Microsatellite instability (MSI) defines one of the four molecular groups of endometrial carcinoma identified by The Cancer Genome Atlas (TCGA). Immunohistochemistry for mismatch repair (MMR) proteins (MLH1, MSH2, MSH6, PMS2) has been proposed as a widely applicable technique to identify this group in the common practice. However, the diagnostic accuracy of such approach has never been calculated. We aimed to assess: 1) the diagnostic accuracy of MMR proteins immunohistochemistry as surrogate of MSI molecular testing in endometrial carcinoma; 2) whether a combination of only two MMR proteins may be used as a still cheaper test. A systematic review and meta-analysis of was performed by searching electronic databases from their inception to September 2019. All studies assessing endometrial carcinoma with bothMMR proteins immunohistochemistry and MSI molecular testing were included. Diagnostic accuracy was assessed as sensitivity, specificity, positive and negative likelihood ratios (LR+, LR-), diagnostic odds ratio (DOR) and area under the curve (AUC) on SROC curves.A subgroup analysis was performed for a combination of only two MMR proteins (MLH1-MSH2 vs MSH6-PMS2). Ten studies with 3097 patients were included. Out of these, 1110 were suitable for the meta-analysis. Immunohistochemistry for all the four MMR proteins showed sensitivity = 0.96, specificity = 0.95, LR + =17.7, LR- = 0.05, DOR = 429.77, and high diagnostic accuracy (AUC = 0.988). The combination of MLH1 and MSH2 showed sensitivity = 0.88, specificity = 0.96, LR + =22.36, LR- = 0.15, DOR = 200.69, and high diagnostic accuracy (AUC = 0.9838). The combination of MSH6 and PMS2 showed the same results as the complete panel of four MMR proteins. In conclusion, MMR proteins immunohistochemistry is a highly accurate surrogate of MSI molecular testing in endometrial carcinoma. A combination of MSH6 and PMS2 may allow reducing the cost without decrease in the diagnostic accuracy. C1 [Raffone, Antonio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics UnitNaples, Italy. [Travaglino, Antonio] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Cerbone, Marco] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics UnitNaples, Italy. [Gencarelli, Annarita] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Mollo, Antonio] University of Salerno, Surgery and Dentistry “Scuola Medica Salernitana”, Department of MedicineSalerno, Baronissi, Italy. [Insabato, Luigi] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Zullo, Fulvio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics UnitNaples, Italy. RP Travaglino, A (reprint author), University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, 80131 Naples, Italy. EM antonio.travaglino@unina.it CR Siegel RL, Miller KD, Jemal A, 2015, Cancer statistics, 2015. CA Cancer J Clin 65(1):5–29 Raffone A, Travaglino A, Mascolo M, Carbone L, Guida M, Insabato L, Zullo F, 2019a, TCGA molecular groups of endometrial cancer: pooled data about prognosis. Gynecol Oncol 155(2): 374–383 Travaglino A, RaffoneA, Saccone G, de Luca C,Mollo A, Mascolo M, d e P l a c i d o G, I n s a b a t o L , Z u l l o F, 2019a , Immunohistochemical nuclear expression of β-catenin as a surrogate of CTNNB1 exon 3 mutation in endometrial Cancer. Am J Clin Pathol 151(5):529–538 Colombo N, Creutzberg C, Amant F, Bosse T, Gonzalez-Martin A, Ledermann J, Marth C, Nout R, Querleu D, Mirza MR, Sessa C, ESMO-ESGO-ESTRO Endometrial Consensus Conference Working Group, 2016, ESMO-ESGO-ESTRO consensus conference on endometrial Cancer: diagnosis, treatment and follow-up. Ann Oncol 27(1):16–41 Gilks CB, Oliva E, Soslow RA, 2013, Poor interobserver reproducibility in the diagnosis of high-grade endometrial carcinoma. Am J Surg Pathol 37:874–881 Hoang LN, McConechy MK, Kobel M et al, 2013, Histotypegenotype correlation in 36 high-grade endometrial carcinomas. Am J Surg Pathol 37:1421–1432 Talhouk A, McConechy MK, Leung S, Li-Chang HH, Kwon JS, Melnyk N, YangW, Senz J, Boyd N, Karnezis AN, Huntsman DG, Gilks CB, McAlpine J, 2015, A clinically applicable molecularbased classification for endometrial cancers. Br J Cancer 113(2): 299–310 Talhouk A, McConechy MK, Leung S, Yang W, Lum A, Senz J, Boyd N, Pike J, Anglesio M, Kwon JS, Karnezis AN, Huntsman DG, Gilks CB, McAlpine J, 2017, Confirmation of ProMisE: a simple, genomics-based clinical classifier for endometrial cancer. Cancer 123(5):802–813 Kommoss S, McConechy MK, Kommoss F, Leung S, Bunz A, Magrill J, Britton H, Kommoss F, Grevenkamp F, Karnezis A, Yang W, Lum A, Kramer B, Taran F, Staebler A, Lax S, Brucker SY, Huntsman DG, Gilks CB,McAlpine J, Talhouk A, 2018, Final validation of the ProMisE molecular classifier for endometrial carcinoma in a large population-based case series. Ann Oncol 29(5): 1180–1188 Network CGAR et al, 2013, Integrated genomic characterization of endometrial carcinoma. Nature 497(7447):67–73 Travaglino A, Raffone A, Mascolo M et al, 2019b, Clear cell endometrial carcinoma and the TCGA classification. Histopathology. , DOI 10.1111/his.13976 Stelloo E, Nout RA, Osse EM et al, 2016, Improved risk assessment by integrating molecular and Clinicopathological factors in early-stage endometrial Cancer. Clin Cancer Res 22(16):4215– 4224 Britton H, Huang L, Lum A et al, 2019, Molecular classification defines outcomes and opportunities in young women with endometrial carcinoma. Gynecol Oncol 153(3):487–495 Travaglino A, Raffone A, Saccone G, Mascolo M, Pignatiello S, Mollo A, de Placido G, Insabato L, Zullo F, 2019c, PTEN immunohistochemistry in endometrial hyperplasia: which are the optimal criteria for the diagnosis of precancer? APMIS 127(4):161–169 Raffone A, Travaglino A, Saccone G, Viggiani M, Giampaolino P, Insabato L, Mollo A, de Placido G, Zullo F, 2019b, PTEN expression in endometrial hyperplasia and risk of cancer: a systematic review and meta-analysis. Arch Gynecol Obstet 299(6):1511–1524 Raffone A, Travaglino A, Saccone G, Mascolo M, Insabato L, Mollo A, de Placido G, Zullo F, 2019c, PAX2 in endometrial carcinogenesis and in differential diagnosis of endometrial hyperplasia. A systematic review and meta-analysis of diagnostic accuracy. Acta Obstet Gynecol Scand 98(3):287–299 Travaglino A, Raffone A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2019d, Immunohistochemical predictive markers of response to conservative treatment of endometrial hyperplasia and early endometrial cancer: a systematic review. Acta Obstet Gynecol Scand 98(9):1086–1099 Raffone A, Travaglino A, Saccone G, Cieri M, Mascolo M, Mollo A, Insabato L, Zullo F, 2019d, Diagnostic and prognostic value of ARID1A in endometrial hyperplasia: a novel marker of occult cancer. APMIS 127(9):597–606 Travaglino A, Raffone A, Saccone G et al, 2019e, Nuclear expression of β-catenin in endometrial hyperplasia as marker of premalignancy. APMIS. , DOI 10.1111/apm.12988 Chao X, Li L, Wu M et al, 2019, Comparison of screening strategies for Lynch syndrome in patients with newly diagnosed endometrial cancer: a prospective cohort study in China. Cancer Commun, Lond, 39:42 Libera L, Craparotta I, Sahnane N, Chiaravalli AM, Mannarino L, Cerutti R, Riva C, Marchini S, Furlan D, 2018, Targeted gene sequencing of lynch syndrome-related and sporadic endometrial carcinomas. Hum Pathol 81:235–244 Bruegl AS, Ring KL, Daniels M et al, 2017, Clinical Challenges Associated with Universal Screening for Lynch Syndrome- Associated Endometrial Cancer. Cancer Prev Res, Phila, 10:108– 115 Goodfellow PJ, Billingsley CC, Lankes HA, Ali S, Cohn DE, Broaddus RJ, Ramirez N, Pritchard CC, Hampel H, Chassen AS, Simmons LV, Schmidt AP, Gao F, Brinton LA, Backes F, Landrum LM, Geller MA, DiSilvestro P, Pearl ML, Lele SB, Powell MA, Zaino RJ, Mutch D, 2015, Combined microsatellite instability, MLH1 methylation analysis, and immunohistochemistry for lynch syndrome screening in endometrial cancers from GOG210: an NRG oncology and gynecologic oncology group study. J Clin Oncol 33:4301–4308 Haraldsdottir S, Hampel H, Tomsic J et al, 2014, Colon and endometrial cancers with mismatch repair deficiency can arise from somatic, rather than germline, mutations. Gastroenterology 147: 1308–1316.e1 McConechy MK, Talhouk A, Li-Chang HH, Leung S, Huntsman DG, Gilks CB, McAlpine J, 2015, Detection of DNA mismatch repair, MMR, deficiencies by immunohistochemistry can effectively diagnose the microsatellite instability, MSI, phenotype in endometrial carcinomas. Gynecol Oncol 137:306–310 Ollikainen M, Abdel-Rahman WM, Moisio AL, Lindroos A, Kariola R, Jarvela I, Poyhonen M, Butzow R, Peltomaki P, 2005, Molecular analysis of familial endometrial carcinoma: a manifestation of hereditary nonpolyposis colorectal cancer or a separate syndrome. J Clin Oncol 23:4609–4616 Peterson LM, Kipp BR, Halling KC, Kerr SE, Smith DI, Distad TJ, Clayton AC, Medeiros F, 2012, Molecular characterization of endometrial cancer: a correlative study assessing microsatellite instability, MLH1 hypermethylation, DNA mismatch repair protein expression, and PTEN, PIK3CA, KRAS, and BRAF mutation analysis. Int J Gynecol Pathol 31:195–205 Choi YD, Choi J, Kim JH et al, 2008, Microsatellite instability at a tetranucleotide repeat in type I endometrial carcinoma. J Exp Clin Cancer Res 27:88 Stelloo E, Jansen AML, Osse EM, Nout RA, Creutzberg CL, Ruano D, Church DN, Morreau H, Smit VTHBM, van Wezel T, Bosse T, 2017, Practical guidance for mismatch repair-deficiency testing in endometrial cancer. Ann Oncol 28:96–102 Sotiriadis A, Papatheodorou SI, Martins WP, 2016, Synthesizing evidence from diagnostic accuracy tests: the SEDATE guideline. Ultrasound Obstet Gynecol 47(3):386–395 Moher D, Shamseer L, Clarke M et al, 2015, Preferred reporting items for systematic review and meta-analysis protocols, PRISMAP, 2015 statement. Systematic Rev 4:1 Whiting PF, Rutjes AW, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, Leeflang MM, Sterne JA, Bossuyt PM, QUADAS-2 Group, 2011)QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med 155(8):529–536 Murphy KM, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8(3): 305–311 Richard GF, Kerrest A, Dujon B, 2008, Comparative genomics and molecular dynamics of DNA repeats in eukaryotes. Microbiol Mol Biol Rev 72(4):686–727 Gulcher J, 2012)Microsatellite markers for linkage and association studies. Cold Spring Harb Protoc 4:425–432 Colle R, Cohen R, Cochereau D, Duval A, Lascols O, Lopez- Trabada D, Afchain P, Trouilloud I, Parc Y, Lefevre JH, Flejou JF, Svrcek M, Andre T, 2017, Immunotherapy and patients treated for cancer with microsatellite instability. Bull Cancer 104(1):42–51 Chang L, Chang M, Chang HM, Chang F, 2018, Microsatellite instability: a predictive biomarker for Cancer immunotherapy. Appl Immunohistochem Mol Morphol 26(2):e15–e21 Moroney MR, Davies KD, Wilberger AC, Sheeder J, Post MD, Berning AA, Fisher C, Lefkowits C, Guntupalli SR, Behbakht K, Corr BR, 2019, Molecular markers in recurrent stage I, grade 1 endometrioid endometrial cancers. Gynecol Oncol 153:517–520 Stelloo E, Bosse T, Nout RA, MacKay HJ, Church DN, Nijman HW, Leary A, Edmondson RJ, Powell ME, Crosbie EJ, Kitchener HC, Mileshkin L, Pollock PM, Smit VT, Creutzberg CL, 2015, Refining prognosis and identifying targetable pathways for highrisk endometrial cancer; a TransPORTEC initiative.Mod Pathol 28: 836–844 Fishel R, LescoeMK, RaoMR, Copeland NG, Jenkins NA, Garber J, Kane M, Kolodner R, 1993, The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell 75(5):1027–1038 Papadopoulos N, Nicolaides NC, Wei YF, Ruben SM, Carter KC, Rosen CA, Haseltine WA, Fleischmann RD, Fraser CM, Adams MD(1994)Mutation of a mutL homolog in hereditary colon cancer. Science 263(5153):1625–1629 Vilar E, Gruber SB, 2010, Microsatellite instability in colorectal cancer-the stable evidence. Nat Rev Clin Oncol 7(3):153–162. , DOI 10.1038/nrclinonc.2009.237 Sarode VR, Robinson L, 2019, Screening for lynch syndrome by immunohistochemistry of mismatch repair proteins: significance of indeterminate result and correlation with mutational studies. Arch Pathol Lab Med 143(10):1225–1233 Watkins JC, Nucci MR, Ritterhouse LL, Howitt BE, Sholl LM, 2016, Unusual mismatch repair Immunohistochemical patterns in endometrial carcinoma. Am J Surg Pathol 40(7):909–916 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1417 EP 1427 DI 10.1007/s12253-020-00811-5 PG 11 ER PT J AU Uhlyarik, A Piurko, V Vizkeleti, L Papai, Zs Raso, E Lahm, E Kiss, Sikter, M Vachaja, J Kenessey, I Timar, J AF Uhlyarik, Andrea Piurko, Violetta Vizkeleti, Laura Papai, Zsuzsanna Raso, Erzsebet Lahm, Erika Kiss, Eva Sikter, Marta Vachaja, Jozsef Kenessey, Istvan Timar, Jozsef TI EGFR Protein Expression of KRAS Wild-Type Colorectal Cancer: Predictive Value of the Sidedness for Efficacy of Anti-EGFR Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon cancer; EGFR protein; Sidedness; Anti-EGFR therapy ID Colon cancer; EGFR protein; Sidedness; Anti-EGFR therapy AB Right- and left-sided colorectal cancers (RSCRC and LSCRC, respectively) are different developmentally, genetically and prognostically. Clinical data also indicate that they respond differently to anti-EGFR therapies. The role of EGFR protein expression in KRAS wild type colorectal cancer is also controversial. Here we have used a cohort of anti-EGFR antibody treated KRAS-wild type colorectal cancer patients (n = 97) to analyse the prognostic role of EGFR protein expression in relation to sidedness. In our cohort EGFR copy number, determined by FISH, was not associated with the level of EGFR protein, assessed by immunohistochemistry and measured by H-scoring. There was a significantly higher EGFR H-score detected in RSCRC as compared to LSCRC in primary tumors (p = 0.04). Furthermore, in a proportion of cases (n = 31) metastatic tissues were also available and their analysis also found a significantly higher EGFR H-score in metastases of RSCRC compared to LSCRC (p = 0.018). Kaplan Meyer survival analysis demonstrated that anti-EGFR antibody therapies were more effective in case of LSCRC compared to RSCRC. Although in case of progression-free survival data just indicated a trend (p = 0.065), in case of overall survival the difference was significant favouring LSCRC (p = 0.047). These data demonstrated for the first time that the EGFR protein expression is significantly higher in KRAS wild type RSLCL as compared to LSCRC. Meanwhile it is somewhat unexpected that the lower EGFR protein expression was found to be associated with better efficacy of anti-EGFR antibody therapies of colorectal cancer, the finding of which must be further validated. C1 [Uhlyarik, Andrea] Honved Korhaz, Onkologiai OsztalyBudapest, Hungary. [Piurko, Violetta] Semmelweis University, 2nd Department of Pathology, 93 Ulloi str, 1091 Budapest, Hungary. [Vizkeleti, Laura] Semmelweis University, 2nd Department of Pathology, 93 Ulloi str, 1091 Budapest, Hungary. [Papai, Zsuzsanna] Honved Korhaz, Onkologiai OsztalyBudapest, Hungary. [Raso, Erzsebet] Semmelweis University, 2nd Department of Pathology, 93 Ulloi str, 1091 Budapest, Hungary. [Lahm, Erika] Honved Korhaz, Onkologiai OsztalyBudapest, Hungary. [Kiss, Eva] Honved Korhaz, Onkologiai OsztalyBudapest, Hungary. [Sikter, Marta] Honved Korhaz, Onkologiai OsztalyBudapest, Hungary. [Vachaja, Jozsef] Honved Korhaz, Onkologiai OsztalyBudapest, Hungary. [Kenessey, Istvan] Semmelweis University, 2nd Department of Pathology, 93 Ulloi str, 1091 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, 93 Ulloi str, 1091 Budapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM jtimar@gmail.com CR Petrelli F, Tomasello G, Borgonovo K, Ghidini M, Turati L, Dallera P, Passalacqua R, Sgroi G, Barni S, 2017, Prognostic survival associated with left-sided vs right-sided colon cancer: a systematic review and meta-analysis. JAMA Oncol 3:211–219 Warschkow R, SulzMC,Marti L, Tarantino I, Schmied BM, Cerny T, Guller U, 2016, Better survival in right-sided versus left-sided stage I - III colon cancer patients. BMC Cancer 16:554 Baran B, Ozupek NM, Tetik YN, Acar E, Bekcioglu O, Baskin Y, 2018, Difference between left-sided and right-sided colorectal cancer: a focused review of literature. Gastroenterol Res 11:264–273 Loree JM, Pereira AAL, Lam M, Willauer AN, Raghav K, Dasari A, Morris VK, Advani S, MenterDG, Eng C, Shaw K, Broaddus R, Routbort MJ, Liu Y, Morris JS, Luthra R, Meric-Bernstam F, Overman MJ, Maru D, Kopetz S, 2018, Classifying colorectal cancer by tumor location rather than sidedness highlights a continuum in mutation profiles and consensus molecular subtypes. Clin Cancer Res 24:1062–1072 Dienstmann R, Vermeulen L, Guinney J, Kopetz S, Tejpar S, Tabernero J, 2017, Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer 17: 79–92 Tejpar S, Stintzing S, Ciardiello F, Tabernero J, van Cutsem E, Beier F, Esser R, Lenz HJ, Heinemann V, 2017, Prognostic and predictive relevance of primary tumor location in patients with RAS wild-type metastatic colorectal cancer: retrospective analyses of the CRYSTAL and FIRE-3 trials. JAMA Oncol 3:194–201 Brule SY, Jonker DJ, Karapetis CS et al, 2015, Location of colon cancer, right-sided versus left-sided, as a prognostic factor and a predictor of benefit from cetuximab in NCIC CO.17. Eur J Cancer 51:1405–1414 Li D, Fu Q, LiM, Yin C, Zhao J, Li F, 2017, Primary tumor site and anti-EGFR monoclonal antibody benefit in metastatic colorectal cancer: a meta-analysis. Future Oncol 13:1115–1127 Cao DD, Xu HL, Xu XM, Ge W, 2017, The impact of primary tumor location on efficacy of cetuximab in metastatic colorectal cancer patients with different KRAS status: a systematic review and meta-analysis. Oncotarget 8:53631–53641 Salem ME, Weinberg BA, Xiu J et al, 2017, Comparative molecular analyses of left-sided colon, right-sided colon, and rectal cancers. Oncotarget 8:86356–86368 Cserepes M, Gy O, Lohinai Z et al, 2014, Subtype-specific KRAS mutations in advanced lung adenocarcinoma: a retrospective study of patients treated with platinum-based chemotherapy. Eur J Cancer 50:1819–1828 Hirsch FR, Varella-Garcia M, Bunn PA et al, 2003, Epidermal growth factor receptor in nonsmall-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol 21:3798–3807 Snyder M, Bottiglieri S, Almhanna K, 2018, Impact of primary tumor location on first-line bevacizumab or cetuximab inmetastatic colorectal cancer. Rev Recent Clin Trials 13:139–149 Yang Z-Y, Shen W-X, Hu X-F, Zheng DY, Wu XY, Huang YF, Chen JZ, Mao C, Tang JL, 2012, EGFR gene copy number as a predictive biomarker for the treatment of metastatic colorectal cancer with anti-EGFR monoclonal antibodies: a meta-analysis. J Hematol Oncol 5:52–62 Hecht JR,Mitchell E,NeubauerMA, Burris HA, Swanson P, Lopez T, Buchanan G, Reiner M, Gansert J, Berlin J, 2010, Lack of correlation between epidermal growth factor receptor status and response to Panitumumab monotherapy in metastatic colorectal cancer. Clin Cancer Res 16:2205–2213 Licitra L, Storkel S, Kerr KM, van Cutsem E, Pirker R, Hirsch FR, Vermorken JB, von Heydebreck A, Esser R, Celik I, Ciardiello F, 2013, Predicitive value of epidermal growth factor receptor expression for first-line chenmotherapy plus cetuximab in patients with head and neck and colorectal cancer: analysis of data from the EXTREME and CRYSTAL studies. Eur J Cancer 49:1161–1168 Chung KY, Shia J, Kemeny NE, Shah M, Schwartz GK, Tse A, Hamilton A, Pan D, Schrag D, Schwartz L, Klimstra DS, Fridman D, Kelsen DP, Saltz LB, 2005, Cetuximab shows activity in colorectal cancer patients with tumors that do not express the epidermal growth factor receptor by immunohistochemistry. J ClinOncol 23:1803–1810 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1429 EP 1434 DI 10.1007/s12253-018-00572-2 PG 6 ER PT J AU Tarancon-Diez, M Buttner, R Friedrichs, N AF Tarancon-Diez, Maria Buttner, Reinhard Friedrichs, Nicolaus TI Enhanced Tumoral MLH1-Expression in MLH1-/PMS2-Deficient Colon Cancer Is Indicative of Sporadic Colon Cancer and Not HNPCC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; HNPCC; Lynch syndrome; MLH1; Mismatch repair enzymes ID Colorectal cancer; HNPCC; Lynch syndrome; MLH1; Mismatch repair enzymes AB Hereditary Non-Polyposis Colorectal Cancer (HNPCC) is caused by germline mutations of mismatch-repair (MMR) genes MLH1, MSH2, MSH6 and PMS2. MLH1-/PMS2-deficient colorectal carcinomas might be HNPCC-associated but also caused by MLH1-promoter methylation in sporadic colon carcinoma. This study analyzed semiquantitatively whether the MLH1 staining pattern might be indicative of sporadic or HNPCC-associated colorectal cancer. Using a semi-quantitative score ranging from 0 (negative) to 12 (maximum immunopositivity) we analyzed MLH1 expression patterns in 130 MLH1-/PMS2-deficient colorectal cancers. The collective consisted of 70 HNPCC-associated colorectal cancers and 60 sporadic colon cancers. In tumor cells of 70 HNPCC-associated colorectal cancers, 64 cases (91.43%) showed no MLH1 staining, 5 cases weak (7.14%) and 1 case (1.43%) stronger staining intensity. In contrast, in tumor cells of 60 sporadic colorectal cancers 45 cases (75.0%) showed no MLH1 staining, 10 cases weak (16.67%) and 5 cases (8.33%) stronger staining intensity to a varying extent. In immuno-positive cases, MLH1 showed a characteristic dot-like nuclear staining pattern in the tumor cells. We compared cases with absent to weak MLH1-staining (immuno-scores 0 to 2) to cases with elevated immuno-scores (3 to 12) detecting a statistically significant difference between HNPCC-associated and sporadic colon cancers (p value = 0.0031, Fisher’s exact test). Taken together, enhanced tumoral MLH1 expression in MLH1-/PMS2-deficient colorectal carcinomas seems to be indicative of sporadic origin. In contrast, HNPCC-associated colorectal cancer showed absent or very weak MLH1 immunopositivity. Therefore, this semiquantitative and easy to exert MLH1 immuno-score might help to identify sporadic MLH1-/PMS2-deficient colorectal cancer cases prior to time-consuming methylation analysis. C1 [Tarancon-Diez, Maria] University of Cologne Medical School, Institute of Pathology, Kerpener Str. 62, 50937 Cologne, Germany. [Buttner, Reinhard] University of Cologne Medical School, Institute of Pathology, Kerpener Str. 62, 50937 Cologne, Germany. [Friedrichs, Nicolaus] University of Cologne Medical School, Institute of Pathology, Kerpener Str. 62, 50937 Cologne, Germany. RP Friedrichs, N (reprint author), University of Cologne Medical School, Institute of Pathology, 50937 Cologne, Germany. EM nicolaus.friedrichs@uk-koeln.de CR Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F, 2017, Global patterns and trends in colorectal cancer incidence and mortality. Gut 66(4):683–691 Haggar FA, Boushey RP, 2009, Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg 22(4):191–197 Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386 Aran V, Victorino AP, Thuler LC, Ferreira CG, 2016, Colorectal Cancer: epidemiology, disease mechanisms and interventions to reduce onset and mortality. Clin Colorectal Cancer 15(3):195–203 Lynch HT, de la Chapelle A, 2003, Hereditary colorectal cancer. N Engl J Med 348(10):919–932 Peltomaki P, 2003, Role of DNA mismatch repair defects in the pathogenesis of human cancer. J Clin Oncol 21(6):1174–1179 Cohen R, Buhard O, Cervera P, Hain E, Dumont S, Bardier A, Bachet JB, Gornet JM, Lopez-Trabada D, Dumont S, Kaci R, Bertheau P, Renaud F, Bibeau F, Parc Y, Vernerey D, Duval A, Svrcek M, Andre T, 2017, Clinical and molecular characterisation of hereditary and sporadic metastatic colorectal cancers harbouring microsatellite instability/DNA mismatch repair deficiency. Eur J Cancer 86:266–274 Poulogiannis G, Frayling IM, Arends MJ, 2010, DNA mismatch repair deficiency in sporadic colorectal cancer and Lynch syndrome. Histopathology 56(2):167–179 Lynch HT, Lanspa S, Shaw T, Casey MJ, Rendell M, Stacey M, Townley T, Snyder C, Hitchins M, Bailey-Wilson J, 2018, Phenotypic and genotypic heterogeneity of Lynch syndrome: a complex diagnostic challenge. Familial Cancer 17(3):403–414 Shia J, 2008, Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part I The utility of immunohistochemistry J Mol Diagn 10(4):293–300 Giuffre G et al, 2005, Microsatellite analysis of hereditary nonpolyposis colorectal cancer-associated colorectal adenomas by laser-assisted microdissection: correlation with mismatch repair protein expression provides new insights in early steps of tumorigenesis. J Mol Diagn 7(2):160–170 Steinke V, Holzapfel S, Loeffler M, Holinski-Feder E, Morak M, Schackert HK, Gorgens H, Pox C, Royer-Pokora B, von Knebel- Doeberitz M, Buttner R, Propping P, Engel C, on behalf of the German HNPCC Consortium, 2014, Evaluating the performance of clinical criteria for predicting mismatch repair gene mutations in Lynch syndrome: a comprehensive analysis of 3,671 families. Int J Cancer 135(1):69–77 Mihaylova VT, Bindra RS, Yuan J, Campisi D, Narayanan L, Jensen R, Giordano F, Johnson RS, Rockwell S, Glazer PM, 2003, Decreased expression of the DNA mismatch repair gene Mlh1 under hypoxic stress in mammalian cells. Mol Cell Biol 23(9):3265–3273 Kansikas M, KaselaM, Kantelinen J, NystromM(2014, Assessing howreduced expression levels of themismatch repair genesMLH1, MSH2, andMSH6 affect repair efficiency. HumMutat 35(9):1123– 1127 Kishi K, Doki Y, Yano M, Yasuda T, Fujiwara Y, Takiguchi S, Kim S, Higuchi I, Monden M, 2003, Reduced MLH1 expression after chemotherapy is an indicator for poor prognosis in esophageal cancers. Clin Cancer Res 9(12):4368–4375 Mangold E, Pagenstecher C, Friedl W, Mathiak M, Buettner R, Engel C, Loeffler M, Holinski-Feder E, Muller-Koch Y, Keller G, Schackert HK, Kruger S, Goecke T, Moeslein G, Kloor M, Gebert J, Kunstmann E, Schulmann K, Ruschoff J, Propping P, the German HNPCC Consortium, 2005, Spectrum and frequencies of mutations in MSH2 and MLH1 identified in 1,721 German families suspected of hereditary nonpolyposis colorectal cancer. Int J Cancer 116(5):692–702 Gullotti L, Czerwitzki J, Kirfel J, Propping P, Rahner N, Steinke V, Kahl P, Engel C, Schule R, Buettner R, Friedrichs N, 2011, FHL2 expression in peritumoural fibroblasts correlates with lymphatic metastasis in sporadic but not in HNPCC-associated colon cancer. Lab Investig 91(12):1695–1705 Al-Nomani L et al, 2015, Tumoral expression of nuclear cofactor FHL2 is associated with lymphatic metastasis in sporadic but not in HNPCC-associated colorectal cancer. Pathol Res Pract 211(2):171– 174 Farchoukh L, Kuan SF, Dudley B, Brand R, Nikiforova M, Pai RK, 2016)MLH1-deficient colorectal carcinoma with wild-type BRAF and MLH1 promoter Hypermethylation Harbor KRAS mutations and Arise from conventional adenomas. Am J Surg Pathol 40(10): 1390–1399 Nakamura H, Tanimoto K, Hiyama K, YunokawaM, Kawamoto T, Kato Y, Yoshiga K, Poellinger L, Hiyama E, Nishiyama M, 2008, Human mismatch repair gene,MLH1, is transcriptionally repressed by the hypoxia-inducible transcription factors, DEC1 and DEC2. Oncogene 27(30):4200–4209 Sameer AS, Nissar S, Fatima K, 2014, Mismatch repair pathway: molecules, functions, and role in colorectal carcinogenesis. Eur J Cancer Prev 23(4):246–257 Watson P, Vasen HFA, Mecklin JP, Bernstein I, Aarnio M, Jarvinen HJ, Myrhoj T, Sunde L, Wijnen JT, Lynch HT, 2008, The risk of extra-colonic, extra-endometrial cancer in the Lynch syndrome. Int J Cancer 123(2):444–449 Park JG, Vasen HF, Park Y, Park K, Peltomaki P, Ponzo de Leon M, Rodriguez-Bigas MA, Lubinski J, Beck NE, Bisgaard ML, Miyaki M, Wijnen JT, Baba S, Lindblom A, Madlensky L, Lynch HT, 2002, Suspected HNPCC and Amsterdam criteria II: evaluation of mutation detection rate, an international collaborative study. Int J Color Dis 17(2):109–114 Newton K, Jorgensen NM, Wallace AJ, Buchanan DD, Lalloo F, McMahon RFT, Hill J, Evans DG, 2014, Tumour MLH1 promoter region methylation testing is an effective prescreen for Lynch syndrome, HNPCC). J Med Genet 51(12):789–796 Bettstetter M, Dechant S, Ruemmele P, Vogel C, Kurz K, Morak M, Keller G, Holinski-Feder E, Hofstaedter F, Dietmaier W, 2008, MethyQESD, a robust and fast method for quantitative methylation analyses in HNPCC diagnostics using formalin-fixed and paraffinembedded tissue samples. Lab Investig 88(12):1367–1375 Yang HM, Mitchell JM, Sepulveda JL, Sepulveda AR, 2015, Molecular and histologic considerations in the assessment of serrated polyps. Arch Pathol Lab Med 139(6):730–741 Mangold E, Pagenstecher C, Friedl W, Fischer HP, Merkelbach- Bruse S, Ohlendorf M, Friedrichs N, Aretz S, Buettner R, Propping P, Mathiak M, 2005, Tumours from MSH2 mutation carriers show loss of MSH2 expression but many tumours from MLH1 mutation carriers exhibit weak positive MLH1 staining. J Pathol 207(4):385–395 Santos JC, Bastos AU, Cerutti JM, Ribeiro ML, 2013, Correlation of MLH1 and MGMT expression and promoter methylation with genomic instability in patients with thyroid carcinoma. BMC Cancer 13:79 Springuel L, Losdyck E, Saussoy P, Turcq B, Mahon FX, Knoops L, Renauld JC, 2016, Loss ofmutL homolog-1, MLH1, expression promotes acquisition of oncogenic and inhibitor-resistant point mutations in tyrosine kinases. Cell Mol Life Sci 73(24):4739–4748 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1435 EP 1439 DI 10.1007/s12253-018-00571-3 PG 5 ER PT J AU Aalijahan, H Ghorbian, S AF Aalijahan, Hamid Ghorbian, Saeid TI Clinical Application of Long Non-Coding RNA-UCA1 as a Candidate Gene in Progression of Esophageal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE LncRNA-UCA1; Candidate gene; Esophageal Cancer; LncRNAs ID LncRNA-UCA1; Candidate gene; Esophageal Cancer; LncRNAs AB Esophageal cancer (EC) is known as one of the most prevalent gastrointestinal cancers, and results in the seventh highest number of cancer-relevant deaths. Long non-coding RNAs (lncRNAs) have substantial roles in several biological processes. LncRNA human urothelial carcinoma associated 1 (UCA1) is announced to be enhanced in multiple types of human cancers. This survey was carried out to identify the potential role of the lncRNA-UCA1 in the progression of EC. A case-control investigation was performed on 140 FFPE tissues of EC patients consisting of 70 cancerous tissues and 70 marginal tissues samples. To determine the lncRNA-UCA1 gene expression changes, quantitative reverse-transcription polymerase chain reaction (qRT-PCR) method was utilized. In addition, the associations between the lncRNA-UCA1 gene expression and clinicopathological parameters were assessed. Our findings revealed that the lncRNA-UCA1 was notably up-regulated in EC tissues compared to adjacent normal tissues (P < 0.05). LncRNA-UCA1 expression was substantially correlated to alcohol drinking (P = 0.008) and socioeconomic status (P = 0.001), while shared no correlation with age, hot drinking status and stage (P > 0.05). Our data indicated that the lncRNA-UCA1 play an important role in the progression of EC and may be considered as a candidate gene in the pathogenesis of EC patients. C1 [Aalijahan, Hamid] Islamic Azad University, Ahar Branch, Department of Molecular GeneticsAhar, Iran. [Ghorbian, Saeid] Islamic Azad University, Ahar Branch, Department of Molecular GeneticsAhar, Iran. RP Ghorbian, S (reprint author), Islamic Azad University, Ahar Branch, Department of Molecular Genetics, Ahar, Iran. EM ghorbian20@yahoo.com;s_ghorbian@iau-ahar.ac.ir CR Zhang Y, 2013 14, Epidemiology of esophageal cancer. World J Gastroenterol: WJG 19(34):5598 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87–108 Ghorbian S, ArdekaniAM(2012, Non-invasive detection of esophageal Cancer using genetic changes in circulating cell-free DNA. Avicenna J Med Biotechnol 4(1):3–13 Song Q, Liu H, Wang J, Jia Y, Liu Y, Wang N, Tan B, Guan S, An D, Cheng Y, 2014, Dinner-to-bed time and post-dinner walk: new potential independent factors in esophageal cancer development. J Cancer Res Clin Oncol 140(5):817–821 Tang WR, Fang JY, Wu KS, Shi XJ, Luo JY, Lin K, 2014, Epidemiological characteristics and prediction of esophageal cancer mortality in China from 1991 to 2012. Asian Pac J Cancer Prev 15(16):6929–6934 Park S, Paeng JC, Kang CH, Cheon GJ, Kang KW, Chung JK, Lee DS, 2018, Dual-time point 18 F-FDG PET/CT for the staging of oesophageal cancer: the best diagnostic performance by retention index for N-staging in non-calcified lymph nodes. Eur J Nucl Med Mol Imaging:1–2 Lanzafame M, Bianco G, Terracciano L, Ng C, Piscuoglio S, 2018, The role of long non-coding RNAs in hepatocarcinogenesis. Int J Mol Sci 19(3):682 Aalijahan H, Ghorbian S, 2018, Long non-coding RNAs and cervical cancer. Exp Mol Pathol Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, Poliakov A, 2015, The landscape of long noncoding RNAs in the human transcriptome. Nat Genet 47(3):199–208 CabiliMN, Trapnell C, Goff L, KoziolM, Tazon-Vega B, Regev A, Rinn JL, 2011, Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 2 Martianov I, Ramadass A, Barros AS, Chow N, Akoulitchev A, 2007, Repression of the human dihydrofolate reductase gene by a non-coding interfering transcript. Nature 445(7128):666–670 Bayoumi AS, Sayed A, Broskova Z, Teoh JP,Wilson J, Su H, Tang YL, Kim IM, 2016, Crosstalk between long noncoding RNAs and microRNAs in health and disease. Int J Mol Sci 17(3):356 Wang KC, Yang YW, Liu B, Sanyal A, Corces-Zimmerman R, Chen Y, Lajoie BR, Protacio A, Flynn RA, Gupta RA, Wysocka J, 2011, A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature. 472(7341):120–124 Ye H, Lin J, Yao X, Li Y, Lin X, Lu H, 2018, Overexpression of long non-coding RNA NNT-AS1 correlates with tumor progression and poor prognosis in osteosarcoma. Cell Physiol Biochem 45(5): 1904–1914 XueM, ChenW, Li X, 2016, Urothelial cancer associated 1: a long noncoding RNA with a crucial role in cancer. J Cancer Res Clin Oncol 142(7):1407–1419 Liu FT, Dong Q, Gao H, Zhu ZM, 2017, The prognostic significance of UCA1 for predicting clinical outcome in patients with digestive system malignancies. Oncotarget 8(25):40620 Huang J, Zhou N, Watabe K, Lu Z, Wu F, Xu M, Mo YY, 2015, Long non-coding RNA UCA1 promotes breast tumor growth by suppression of p27, Kip1). Cell Death Dis 5(1):e1008 Yang YT, Wang YF, Lai JY, Shen SY, Wang F, Kong J, Zhang W, Yang HY, 2016, Long non-coding RNA UCA 1 contributes to the progression of oral squamous cell carcinoma by regulating the WNT/β-catenin signaling pathway. Cancer Sci 107(11):1581–1589 Bian Z, Jin L, Zhang J, Yin Y, Quan C, Hu Y, Feng Y, Liu H, Fei B, Mao Y, Zhou L, 2016, LncRNA—UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting miR-204-5p. Sci Rep 6:23892 Razavi M, Ghorbian S, 2019, Original article - up-regulation of long non-coding RNA-PCAT-1 promotes invasion and metastasis in esophageal squamous cell carcinoma. EXCLI 18:422–428 Sadeghpour S, Ghorbian S, 2019, Evaluation of the potential clinical prognostic value of lncRNA-BANCR gene in esophageal squamous cell carcinoma. Mol Biol Rep 46:991–995 Qian Y, Liu D, Cao S, Tao Y, Wei D, Li W, Li G, Pan X, Lei D, 2017, Upregulation of the long noncoding RNA UCA1 affects the proliferation, invasion, and survival of hypopharyngeal carcinoma. Mol Cancer 16(1):68 Zhao W, Sun C, Cui Z, 2017, A long noncoding RNA UCA1 promotes proliferation and predicts poor prognosis in glioma. Clin Transl Oncol 19(6):735–741 Chen P,Wan D, Zheng D, Zheng Q,Wu F, Zhi Q, 2016, Long noncoding RNA UCA1 promotes the tumorigenesis in pancreatic cancer. Biomed Pharmacother 83:1220–1226 WangHM, Lu JH, ChenWY, Gu AQ, 2015, Upregulated lncRNAUCA1 contributes to progression of lung cancer and is closely related to clinical diagnosis as a predictive biomarker in plasma. Int J Clin Exp Med 8(7):11824 Fang Z,Wu L,Wang L, Yang Y, Meng Y, Yang H, 2014, Increased expression of the long non-coding RNAUCA1 in tongue squamous cell carcinomas: a possible correlation with cancer metastasis. Oral Surg Oral Med Oral Pathol Oral Radiol 117(1):89–95 Huang J, Zhou N, Watabe K, Lu Z, Wu F, Xu M, Mo YY, 2015, Long non-coding RNA UCA1 promotes breast tumor growth by suppression of p27, Kip1). Cell Death Dis 5(1):e1008 Han Y, Yang YN, Yuan HH, Zhang TT, Sui H, Wei XL, Liu L, Huang P, Zhang WJ, Bai YX, 2014, UCA1, a long non-coding RNA up-regulated in colorectal cancer influences cell proliferation, apoptosis and cell cycle distribution. Pathology. 46(5): 396–401 Zheng Q,Wu F, DaiWY, ZhengDC, Zheng C,Ye H, Zhou B, Chen JJ, Chen P, 2015, Aberrant expression of UCA1 in gastric cancer and its clinical significance. Clin Transl Oncol 17(8):640–646 Wang F,Ying HQ,He BS, Pan YQ, DengQW, Sun HL, Chen J, Liu X, Wang SK, 2015, Upregulated lncRNA-UCA1 contributes to progression of hepatocellular carcinoma through inhibition of miR-216b and activation of FGFR1/ERK signaling pathway. Oncotarget 6(10):7899–7917 Tian Y, Zhang X, Hao Y, Fang Z, He Y, 2014, Potential roles of abnormally expressed long noncoding RNA UCA1 and Malat-1 in metastasis of melanoma. Melanoma Res 24(4):335–341 Liu SP, Yang JX, Cao DY, Shen K, 2013, Identification of differentially expressed long non-coding RNAs in human ovarian cancer cells with different metastatic potentials. Cancer Biology & Medicine 10(3):138 Li JY, Ma X, Zhang CB, 2014, Overexpression of long non-coding RNA UCA1 predicts a poor prognosis in patients with esophageal squamous cell carcinoma. Int J Clin Exp Pathol 7(11):7938 Wang X, Gao Z, Liao J, Shang M, Li X, Yin L, Pu Y, Liu R, 2016, lncRNA UCA1 inhibits esophageal squamous-cell carcinoma growth by regulating the Wnt signaling pathway. J Toxic Environ Health A 79(9–10):407–418 Jiao C, Song Z, Chen J, Zhong J, CaiW, Tian S, Chen S, Yi Y, Xiao Y, 2016, lncRNA-UCA1 enhances cell proliferation through functioning as a ceRNA of Sox4 in esophageal cancer. Oncol Rep 36(5): 2960–2966 Deng F, Zhou K, Cui W, Liu D, Ma Y, 2015, Clinicopathological significance of wnt/β-catenin signaling pathway in esophageal squamous cell carcinoma. Int J Clin Exp Pathol 8(3):3045 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1441 EP 1446 DI 10.1007/s12253-019-00711-3 PG 6 ER PT J AU Kranitz, N Szepesvary, Zs Kocsis, K Kullmann, T AF Kranitz, Noemi Szepesvary, Zsolt Kocsis, Karoly Kullmann, Tamas TI Neuroendocrine Cancer of the Prostate SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Neuroendocrine; Prostate cancer; Immunohistochemistry; Serum marker; Platinum ID Neuroendocrine; Prostate cancer; Immunohistochemistry; Serum marker; Platinum AB Neuroendocrine cancer of the prostate is considered to be a rare entity with bad prognosis and limited therapeutic options. We performed a prospective analysis of the patients treated in our hospital for prostate cancer between 1st January 2015 and 31rd December 2018. Neuroendocrine phenomena were tested by immunohistochemistry and laboratory chemistry on the request of the clinicians in the cases when a positive diagnosis was suspected. Clinical tableaux of high suspicion of neuroendocrine cancer included radiological progression of a metastatic disease without PSA rise, relatively extended metastatic disease associated to a low PSA, disease with non-pulmonary visceral metastases. 10 patients were diagnosed with neuroendocrine tumour out of 521 prostate cancers. Half of the patients had a survival over a year. 3 patients received 3 lines of efficacious palliative chemotherapy. 1 patient underwent prostatectomy after neoadjuvant chemotherapy for a localised disease. The incidence of neuroendocrine tumours among prostate cancer patients was higher than expected. Some of the patients had a relatively good outcome. C1 [Kranitz, Noemi] Petz Aladar Hospital, Department of PathologyGyor, Hungary. [Szepesvary, Zsolt] Petz Aladar Hospital, Department of UrologyGyor, Hungary. [Kocsis, Karoly] Petz Aladar Hospital, Department of Oncoradiology, Vasvari Pal u. 2-4, 9024 Gyor, Hungary. [Kullmann, Tamas] Petz Aladar Hospital, Department of Oncoradiology, Vasvari Pal u. 2-4, 9024 Gyor, Hungary. RP Kullmann, T (reprint author), Petz Aladar Hospital, Department of Oncoradiology, 9024 Gyor, Hungary. EM kullmanndoki@hotmail.com CR Honn KV, Aref A, Chen YQ et al, 1996, Prostate cancer - old problems and new approaches., Part II. Diagnostic and prognostic markers, Pathology and Biological Aspects). Pathol Oncol Res 2: 191–211 Parimi V, Goyal R, Poropatich K, Yang XJ, 2014, Neuroendocrine differenciation of prostate cancer: a review, 2014). Am J Clin Exp Urol 2:273–285 Li J, Wang Z, 2016, The pathology of unusual subtypes of prostate cancer. Chin J Cancer Res 28:130–143 Zaffuto E, Pompe R, ZanatyMet al, 2017, Contemporary incidence and cancer control outcomes of primary neuroendocrine prostate cancer: a SEER database analysis. Clin Genitourin Cancer 15: e793–e800 Kishore K, Rafeeq A, Chime C et al, 2018, Poorly differentiated small-cell-type neuroendocrine carcinoma of the prostate: a case report and literature review. Case Rep Oncol 11:676–681 Palmgren JS, Karavadia SS, Wakefield MR, 2007, Unusual and underappreciated: small cell cancer of the prostate. Semin Oncol 34:22–29 Beltran H, Oromendia C, Danila DC et al, 2019, A phase II trial of the Aurora kinase a inhibitor alisertib for patients with castrationresistant and neuroendocrine prostate cancer: efficacy and biomarkers. Clin Cancer Res 25:43–51 Monn MF, Cheng L, 2016, Emerging trend in the evaluationand management of small cell prostate cancer: a clinical and molecular perspective. Expert Rev Anticancer Ther 16:1029–1037 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1447 EP 1450 DI 10.1007/s12253-019-00712-2 PG 4 ER PT J AU Li, Y Yang, Sh Yue, H Yuan, D Li, L Zhao, J Zhao, L AF Li, Yan Yang, Shouyan Yue, Honggang Yuan, Dandi Li, Luxia Zhao, Jinghong Zhao, Lintao TI Unraveling LGALS1 as a Potential Immune Checkpoint and a Predictor of the Response to Anti-PD1 Therapy in Clear Cell Renal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Immunotherapy; PD-L1; LGALS1; Galectin1; Clear cell renal carcinoma; Immune checkpoint ID Immunotherapy; PD-L1; LGALS1; Galectin1; Clear cell renal carcinoma; Immune checkpoint AB Immunotherapy base on immune checkpoint inhibitor had obtained significant progress in extending the survival of clear cell renal carcinoma (ccRCC) patients. In order to further improve the efficiency of immunotherapy, novel immune checkpoint inhibitors needed to be developed. Differentially expressed genes (DEGs) between healthy kidney tissues and ccRCC tissues had been found from GSE68417 by GEO2R online analysis tool. Correlation analysis and Kaplan–Meier survival analyses were based on UALCAN database. Analyses of the outcome of anti-PD1 treatment had been found from GSE67501 dataset. At first, 9 genes with higher expression were associated with shorter overall survival time. More importantly, higher expression of LGALS1 was correlated with a profitable outcome of anti-PD1 treatment and the combined the expression level of PD-L1 and LGALS1 together could more efficiently predict the outcome of anti-PD1 treatment than using PD-L1 alone. At last, the genes which correlated with LGALS1 expression in ccRCC patients were enriched in TNF alpha Signaling Pathway which is mainly correlated with T cell apoptosis and survival. Together, these suggest LGALS1 could be a potential immune checkpoint, which could promote tumor progression through affecting T cell survival. C1 [Li, Yan] Army Medical University (Third Military Medical University), Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Department of Nephrology, 400037 Chongqing, China. [Yang, Shouyan] Army Medical University (Third Military Medical University), 958th Hospital, Southwest Hospital, Department of Oncology, 400020 Chongqing, China. [Yue, Honggang] Army Medical University (Third Military Medical University), 958th Hospital, Southwest Hospital, Department of Oncology, 400020 Chongqing, China. [Yuan, Dandi] Army Medical University (Third Military Medical University), 958th Hospital, Southwest Hospital, Department of Oncology, 400020 Chongqing, China. [Li, Luxia] Army Medical University (Third Military Medical University), 958th Hospital, Southwest Hospital, Department of Oncology, 400020 Chongqing, China. [Zhao, Jinghong] Army Medical University (Third Military Medical University), Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Department of Nephrology, 400037 Chongqing, China. [Zhao, Lintao] Army Medical University (Third Military Medical University), Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Department of Nephrology, 400037 Chongqing, China. RP Zhao, J (reprint author), Army Medical University (Third Military Medical University), Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Department of Nephrology, 400037 Chongqing, China. EM zhaojh@tmmu.edu.cn CR Frew IJ, Moch H, 2015, A clearer view of the molecular complexity of clear cell renal cell carcinoma. Annu Rev Pathol 10:263–289 Hecht JR. Current and emerging therapies for metastatic colorectal cancer: applying research findings to clinical practice. American journal of health-system pharmacy : AJHP : official journal of the American Society of Health-System Pharmacists. 2008;65:S15–21; quiz S2–4 Buczek M, Escudier B, Bartnik E, Szczylik C, Czarnecka A, 1845, Resistance to tyrosine kinase inhibitors in clear cell renal cell carcinoma: from the patient's bed to molecular mechanisms. Biochim Biophys Acta 2014:31–41 Ciccarese C, Massari F, Santoni M, Heng DY, Sotte V, Brunelli M et al, 2015, New molecular targets in non clear renal cell carcinoma: an overview of ongoing clinical trials. Cancer Treat Rev 41: 614–622 Vermassen T, De Meulenaere A, Van deWalle M, Rottey S, 2017, Therapeutic approaches in clear cell and non-clear cell renal cell carcinoma. Acta Clin Belg 72:12–18 Miao D,Margolis CA, GaoW, VossMH, LiW, Martini DJ, Norton C, Bosse D,Wankowicz SM, Cullen D, Horak C,Wind-Rotolo M, Tracy A, Giannakis M, Hodi FS, Drake CG, Ball MW, Allaf ME, Snyder A, Hellmann MD, Ho T, Motzer RJ, Signoretti S, Kaelin WG Jr, Choueiri TK, van Allen EM, 2018, Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science. 359:801–806 Choueiri TK, Fishman MN, Escudier B, McDermott DF, Drake CG, Kluger H et al, 2016, Immunomodulatory activity of Nivolumab in metastatic renal cell carcinoma. Clinical cancer research : an Official Journal of the American Association for Cancer Research 22:5461–5471 Motzer RJ, Tannir NM,McDermott DF, Aren Frontera O, Melichar B, Choueiri TK et al, 2018, Nivolumab plus Ipilimumab versus Sunitinib in advanced renal-cell carcinoma. N Engl J Med 378: 1277–1290 Sharpe AH, Pauken KE, 2018, The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol 18:153–167 Thibodeau BJ, Fulton M, Fortier LE, Geddes TJ, Pruetz BL, Ahmed S, et al. Characterization of clear cell renal cell carcinoma by gene expression profiling. Urologic oncology. 2016;34:168.e1– 9 Ascierto ML, McMiller TL, Berger AE, Danilova L, Anders RA, Netto GJ et al, 2016, The Intratumoral balance between metabolic and immunologic gene expression is associated with anti-PD-1 response in patients with renal cell carcinoma. Cancer immunology research 4:726–733 Yang W, Ma J, Zhou W, Li Z, Zhou X, Cao B et al, 2019, Identification of hub genes and outcome in colon cancer based on bioinformatics analysis. Cancer Manag Res 11:323–338 Jha PK, Vijay A, Sahu A, Ashraf MZ, 2016, Comprehensive gene expression meta-analysis and integrated bioinformatic approaches reveal shared signatures between thrombosis and myeloproliferative disorders. Sci Rep 6:37099 Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S, Jenkins SL, Jagodnik KM, Lachmann A, McDermottMG,Monteiro CD, Gundersen GW,Ma'ayan A, 2016, Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res 44:W90–W97 Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi B, et al. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses. Neoplasia, New York, NY). 2017;19:649–58 Lou W, Chen J, Ding B, Chen D, Zheng H, Jiang D, Xu L, Bao C, Cao G, Fan W, 2018, Identification of invasion-metastasisassociated microRNAs in hepatocellular carcinoma based on bioinformatic analysis and experimental validation. J Transl Med 16:266 Pomer S, Schirrmacher V, Thiele R, Lohrke H, Brkovic D, Staehler G, 1995, Tumor response and 4 year survival-data of patients with advanced renal-cell carcinoma treated with autologous tumor vaccine and subcutaneous R-IL-2 and IFN-alpha(2b). Int J Oncol 6: 947–954 Picarda E, Ohaegbulam KC, Zang X, 2016, Molecular pathways: targeting B7-H3, CD276, for human Cancer immunotherapy. Clinical Cancer Research : an Official Journal of the American Association for Cancer Research 22:3425–3431 He J, Baum LG, 2004, Presentation of galectin-1 by extracellular matrix triggers T cell death. J Biol Chem 279:4705–4712 Ramanan VK, Risacher SL, Nho K, Kim S, Shen L,McDonald BC et al, 2015, GWAS of longitudinal amyloid accumulation on 18Fflorbetapir PET in Alzheimer's disease implicates microglial activation gene IL1RAP. Brain J Neurol 138:3076–3088 Smith NL, Hankinson J, Simpson A, Denning DW, Bowyer P, 2014, Reduced expression of TLR3, TLR10 and TREM1 by human macrophages in chronic cavitary pulmonary aspergillosis, and novel associations of VEGFA, DENND1B and PLAT. Clinical Microbiology and Infection : Official Publication of the European Society of Clinical Microbiology and Infectious Diseases 20: O960–O968 Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, Miller ML, Rekhtman N, Moreira AL, Ibrahim F, Bruggeman C, Gasmi B, Zappasodi R, Maeda Y, Sander C, Garon EB, Merghoub T, Wolchok JD, Schumacher TN, Chan TA, 2015, Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 348:124–128 Castro MP, Goldstein N, 2015, Mismatch repair deficiency associated with complete remission to combination programmed cell death ligand immune therapy in a patient with sporadic urothelial carcinoma: immunotheranostic considerations. Journal for immunotherapy of cancer 3:58 Astorgues-Xerri L, Riveiro ME, Tijeras-Raballand A, Serova M, Neuzillet C, Albert S, Raymond E, Faivre S, 2014, Unraveling galectin-1 as a novel therapeutic target for cancer. Cancer Treat Rev 40:307–319 Cedeno-Laurent F, Dimitroff CJ. Galectin-1 research in T cell immunity: past, present and future. Clinical immunology, Orlando, Fla). 2012;142:107–16 Perillo NL, Pace KE, Seilhamer JJ, Baum LG, 1995, Apoptosis of T cells mediated by galectin-1. Nature. 378:736–739 Ishitani T, Takaesu G, Ninomiya-Tsuji J, Shibuya H, Gaynor RB, Matsumoto K, 2003, Role of the TAB2-related protein TAB3 in IL- 1 and TNF signaling. EMBO J 22:6277–6288 Wang L, FengW, Yang X, Yang F,Wang R, Ren Q, Zhu X, Zheng G, 2018, Fbxw11 promotes the proliferation of lymphocytic leukemia cells through the concomitant activation of NF-kappaB and beta-catenin/TCF signaling pathways. Cell Death Dis 9:427 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1451 EP 1458 DI 10.1007/s12253-019-00710-4 PG 8 ER PT J AU Liu, ZQ Lu, MY Liu, B AF Liu, Zhi-Qin Lu, Mei-Yin Liu, Bin TI Polymorphisms in XPC Gene and Risk of Uterine Leiomyoma in Reproductive Women SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Uterine leiomyoma; XPC; Polymorphism; Genetic susceptibility ID Uterine leiomyoma; XPC; Polymorphism; Genetic susceptibility AB XPC gene belongs to DNA repair pathway, which is involved in the development of uterine leiomyoma. However, its relationships with leiomyoma risk were never reported. We here hypothesized that XPC gene was associated with the risk of uterine leiomyoma. In this case–control study with a total of 391 leiomyoma cases and 493 tumor-free controls in a reproductive women population in South China, two missense polymorphisms rs2228001 A > C (Lys939Gln) and rs2228000 C > T (Ala499Val) were genotyped by quantitative polymerase chain reaction (qPCR). Then, the associations between these two polymorphisms and leiomyoma risk were investigated. It was revealed that the rs2228000 CT/TT variant genotypes had a decreased leiomyoma risk (adjusted odds ratio = 0.73, 95% confidence interval = 0.54–0.94) compared with rs2228000 CC genotype. Further stratified analysis also revealed that the protective effect of rs2228000 CT/TT on the risk of uterine leiomyoma was more evident among subjects who were younger than 35 years old compared with those with larger tumors (diameter of tumor >5 cm), and those with fewer number of myomas (only one). However, no significant association was observed for leiomyoma risk for rs2228001 A > C. This study indicated that genetic variations in XPC gene are associated with leiomyoma susceptibility in a reproductive women population. It warrants further confirmation in larger prospective studies with different populations. C1 [Liu, Zhi-Qin] Jinan University, Baoan Maternal and Child Health Hospital, Department of Obstetrics and GynecologyShenzhen, Guangdong, China. [Lu, Mei-Yin] Jinan University, Baoan Maternal and Child Health Hospital, Department of Biobank, 518102 Shenzhen, Guangdong, China. [Liu, Bin] Jinan University, Baoan Maternal and Child Health Hospital, Department of Biobank, 518102 Shenzhen, Guangdong, China. RP Liu, B (reprint author), Jinan University, Baoan Maternal and Child Health Hospital, Department of Biobank, 518102 Shenzhen, China. EM gz12liubin@163.com CR Stewart EA, Laughlin-Tommaso SK, CatherinoWH, Lalitkumar S, Gupta D, Vollenhoven B, 2016, Uterine fibroids. Nat Rev Dis Primers 2:16043–16043 Rice KE, Secrist JR, Woodrow EL, Hallock LM, Neal JL, 2012, Etiology, diagnosis, and management of uterine leiomyomas. J Midwifery Womens Health 57:241–247 Rein MS, 2000, Advances in uterine leiomyoma research: the progesterone hypothesis. Environ Health Perspect 108:791–793 Jeon YT, Kim JW, Park NH, Song YS, Kang SB, Lee HP, 2005, DNA repair gene XRCC1 Arg399Gln polymorphism is associated with increased risk of uterine leiomyoma. Hum Reprod 20:1586–1589 Chang WS, Tsai CW, Wang JY, Ying TH, Hsiao TS, Chuang CL, Yueh TC, Liao CH, Hsu CM, Liu SP, Gong CL, Tsai CH, Bau DT, 2015, Contribution of X-ray repair complementing defective repair in Chinese hamster cells 3, XRCC3, genotype to leiomyoma risk. Anticancer Res 35:4691–4696 Hsieh YY, Chang CC, Bau DT, Yeh LS, Tsai FJ, Tsai CH, 2008, Xray repair cross-complementing group 4, XRCC4, promoter −1394( *)T-related genotype, but not XRCC4 codon 247/intron 3 or xeroderma pigmentosum group D codon 312, 751/promoter−114, polymorphisms are correlated with higher susceptibility to myoma. Fertil Steril 90:1417–1423 Sugasawa K, Ng JM, Masutani C, Iwai S, van der Spek PJ, Eker AP, Hanaoka F, Bootsma D, Hoeijmakers JH, 1998, Xeroderma pigmentosum group C protein complex is the initiator of global genome nucleotide excision repair. Mol Cell 2:223–232 D'ErricoM, Parlanti E, TesonM, de Jesus BM, Degan P, Calcagnile A, Jaruga P, Bjoras M, Crescenzi M, Pedrini AM, Egly JM, Zambruno G, Stefanini M, Dizdaroglu M, Dogliotti E, 2006, New functions of XPC in the protection of human skin cells from oxidative damage. EMBO J 25:4305–4315 Chuang CL, Wang CH, Hsu CH, Hsiao CL, Chen GL, Yen ST, Li HT, Chang WS, Tsai CW, Wang SC, Bau DT, 2017, Contribution of double-strand break repair Gene Nijmegen breakage syndrome 1 genotypes, gender difference and smoking status to Taiwanese lung Cancer. Anticancer Res 37:2417–2423 Zhao M, Howard EW, Guo Z, Parris AB, Yang X, 2017, p53 pathway determines the cellular response to alcohol-induced DNA damage in MCF-7 breast cancer cells. PLoS One 12: e0175121 He J, Shi TY, Zhu ML, Wang MY, Li QX, Wei QY, 2013, Associations of Lys939Gln and Ala499Val polymorphisms of the XPC gene with cancer susceptibility: a meta-analysis. Int J Cancer 133:1765–1775 Hua RX, Zhu J, Jiang DH, Zhang SD, Zhang JB, XueWQ, Li XZ, Zhang PF, He J, JiaWH(2016, Association of XPC gene polymorphisms with colorectal Cancer risk in a southern Chinese population: a case-control study and meta-analysis. Genes, Basel, 7:73–73 He J, Qiu LX, Wang MY, Hua RX, Zhang RX, Yu HP, Wang YN, Sun MH, Zhou XY, Yang YJ,Wang JC, Jin L,Wei QY, Li J, 2012, Polymorphisms in the XPG gene and risk of gastric cancer in Chinese populations. Hum Genet 131:1235–1244 He J, Wang F, Zhu J, Zhang R, Yang T, Zou Y, Xia H, 2016, Association of potentially functional variants in the XPG gene with neuroblastoma risk in a Chinese population. J Cell Mol Med 20: 1481–1490 Hua RX, Zhuo ZJ, Shen GP, Zhu J, Zhang SD, Xue WQ, Li XZ, Zhang PF, He J, Jia WH, 2016, Polymorphisms in the XPC gene and gastric cancer susceptibility in a southern Chinese population. Onco Targets Ther 9:5513–5519 He BS, Xu T, Pan YQ, Wang HJ, Cho WC, Lin K, Sun HL, Gao TY, Wang SK, 2016, Nucleotide excision repair pathway gene polymorphisms are linked to breast cancer risk in a Chinese population. Oncotarget 7:84872–84882 Engin AB, Karahalil B, Engin A, Karakaya AE, 2011, DNA repair enzyme polymorphisms and oxidative stress in a Turkish population with gastric carcinoma. Mol Biol Rep 38:5379–5386 Wang Y, Li Z, Liu N, Zhang G, 2014, Association between CCND1 and XPC polymorphisms and bladder cancer risk: a meta-analysis based on 15 case-control studies. Tumour Biol 35: 3155–3165 Zheng Y, Deng Z, Yin J,Wang S, Lu D,Wen X, Li X, Xiao D, Hu C, Chen X, Zhang W, Zhou H, Liu Z, 2017, The association of genetic variations in DNA repair pathways with severe toxicities in NSCLC patients undergoing platinum-based chemotherapy. Int J Cancer 141:2336–2347 Dong J, Hu Z, Shu Y, Pan S, ChenW,Wang Y, Hu L, Jiang Y, Dai J, Ma H, Jin G, Shen H, 2012, Potentially functional polymorphisms in DNA repair genes and non-small-cell lung cancer survival: a pathway-based analysis. Mol Carcinog 51:546–552 Xue MH, Li GY, Wu XJ, Zhang CX, Zhang CF, Zhu KX, 2015, Genetic variability of genes in NER pathway influences the treatment outcome of gastric cancer. Int J Clin Exp Pathol 8:5563–5569 LongXD,Ma Y, Huang YZ, Yi Y, Liang QX, Ma AM, Zeng LP, Fu GH, 2010, Genetic polymorphisms in DNA repair genes XPC, XPD, and XRCC4, and susceptibility to helicobacter pylori infection-related gastric antrum adenocarcinoma in Guangxi population, China. Mol Carcinog 49:611–618 Paszkowska-Szczur K, Scott RJ, Serrano-Fernandez P, Mirecka A, Gapska P, Gorski B, Cybulski C, Maleszka R, Sulikowski M, Nagay L, Lubinski J, Debniak T, 2013, Xeroderma pigmentosum genes and melanoma risk. Int J Cancer 133:1094–1100 Zhao Z, Zhang A, Zhao Y, Xiang J, Yu D, Liang Z, Xu C, Zhang Q, Li J, Duan P, 2018, The association of polymorphisms in nucleotide excision repair genes with ovarian cancer susceptibility. Biosci Rep 38:BSR20180114 Wood RD, Mitchell M, Sgouros J, Lindahl T, 2001, Human DNA repair genes. Science 291:1284–1289 Watanabe K, Ikuno Y, Kakeya Y, Ikeno S, Taniura H, Kurono M, Minemori K, Katsuyama Y, Naka-Kaneda H, 2019, Age-related dysfunction of the DNA damage response in intestinal stem cells. Inflamm Regen 39:8–8 Anglada T, Repulles J, Espinal A, LaBarge MA, Stampfer MR, Genesca A, Martin M, 2019, Delayed γH2AX foci disappearance in mammary epithelial cells from aged women reveals an ageassociated DNA repair defect. Aging, Albany NY, 11:1510–1523 Paszkowska-Szczur K, Scott RJ, Gorski B, Cybulski C, Kurzawski G, Dymerska D, Gupta S, van de Wetering T, Masojc B, Kashyap A, Gapska P, Gromowski T, Kladny J, Lubinski J, Debniak T, 2015, Polymorphisms in nucleotide excision repair genes and susceptibility to colorectal cancer in the polish population. Mol Biol Rep 42:755–764 Ebili HO, Iyawe VO, Adeleke KR, Salami BA, Banjo AA, Nolan C, Rakha E, Ellis I, Green A, Agboola AOJ, 2018, Checkpoint kinase 1 expression predicts poor prognosis in Nigerian breast Cancer patients. Mol Diagn Ther 22:79–90 Kim DH, Lee H, Kim DH, Chae SW, Sohn JH, Kim K, Do SI, 2018, High excision repair cross-complementation group 1 expression is associated with favorable prognostic factors in breast cancer. Oncol Lett 1:4995–5003 Suh KJ, Ryu HS, Lee KH, Kim H, Min A, Kim TY, Yang Y, Lee HB, Moon HG, Han SW, Oh DY, Han W, Park IA, Noh DY, Im SA, 2019, Prognostic effects of abnormal DNA damage response protein expression in breast cancer. Breast Cancer Res Treat 175:117–127 Bray MJ, Torstenson ES, Jones SH, Edwards TL, Velez Edwards DR, 2018, Evaluating risk factors for differences in fibroid size and number using a large electronic health record population. Maturitas 114:9–13 Peddada SD, Laughlin SK, Miner K, Guyon JP, Haneke K, Vahdat HL, Semelka RC, Kowalik A, Armao D, Davis B, Baird DD, 2008, Growth of uterine leiomyomata among premenopausal blank and white women. Proc Natl Acad Sci U S A 105:19887–19892 Gupta S, Jose J, Manyonda I, 2008, Clinical presentation of fibroids. Best Pract Res Clin Obstet Gynaecol 22:615–626 Zepiridis LI, Grimbizis GF, Tarlatzis BC, 2015, Infertility and uterine fibroids. Best Pract Res Clin Obstet Gynaecol 34:66–73 Donnez J, DolmansMM(2016, Uterine fibroid management: from the present to the future. Hum Reprod Update 22:665–686 Yoo EH, Lee PI, Huh CY, Kim DH, Lee BS, Lee JK, Kim D, 2007, Predictors of leiomyoma recurrence after laparoscopic myomectomy. J Minim Invasive Gynecol 14:690–697 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1459 EP 1464 DI 10.1007/s12253-019-00720-2 PG 6 ER PT J AU Respondek, M Beberok, A Rzepka, Z Rok, J Wrzesniok, D AF Respondek, Michalina Beberok, Artur Rzepka, Zuzanna Rok, Jakub Wrzesniok, Dorota TI Mcl-1 Inhibitor Induces Cells Death in BRAF-Mutant Amelanotic Melanoma Trough GSH Depletion, DNA Damage and Cell Cycle Changes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Amelanotic melanoma; Apoptosis; BH3 mimetics; MIM1; Dacarbazine; image cytometry ID Amelanotic melanoma; Apoptosis; BH3 mimetics; MIM1; Dacarbazine; image cytometry AB Mcl-1 is a potent antiapoptotic protein and amplifies frequently in many human cancer. Currently, it is considered that the extensively expressed of Mcl-1 protein in melanoma cells is associated with rapid tumor progression, poor prognosis and low chemosensitivity. Therefore, the antiapoptotic protein Mcl-1 could be considered as a potential target for malignant melanoma treatment. The aim of this study was to assess the effect of MIM1 a specific low molecular Mcl-1 protein inhibitor and mixture of MIM1 and dacarbazine on the viability, cell cycle progression and apoptosis induction in amelanotic C32 melanoma cells. The cytotoxic activity of MIM1 towards C32 melanoma cells was examined by the WST-1 test. The Mcl-1 protein level as a drug target in amelanotic melanoma cells was defined by Western blot analysis. Cell cycle progression, DNA fragmentation as well as GSH depletion were determined by fluorescence image cytometer NucleoCounter NC-3000. The obtained results demonstrate that the specific Mcl-1 protein inhibitor - MIM1 decreases cell viability and induce apoptosis (S-phase arrest, DNA fragmentation and redox imbalance) in amelanotic melanoma cells and intensify the proapoptotic properties of DTIC, as a result of interactions with Mcl-1 protein. Taken together, the presented data suggest that Mcl-1 protein is a an important target in malignant melanoma treatment and provide for the first time convincing evidence that MIM1, which inhibits Mcl-1 antiapoptotic protein is able to induce apoptosis and sensitize melanoma cells to alkylating agent. C1 [Respondek, Michalina] Medical University of Silesia, Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, Jagiellonska 4, 41-200 Sosnowiec, Poland. [Beberok, Artur] Medical University of Silesia, Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, Jagiellonska 4, 41-200 Sosnowiec, Poland. [Rzepka, Zuzanna] Medical University of Silesia, Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, Jagiellonska 4, 41-200 Sosnowiec, Poland. [Rok, Jakub] Medical University of Silesia, Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, Jagiellonska 4, 41-200 Sosnowiec, Poland. [Wrzesniok, Dorota] Medical University of Silesia, Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, Jagiellonska 4, 41-200 Sosnowiec, Poland. RP Respondek, M (reprint author), Medical University of Silesia, Department of Pharmaceutical Chemistry, School of Pharmacy with the Division of Laboratory Medicine, 41-200 Sosnowiec, Poland. EM mrespondek@sum.edu.pl CR McKibbin T, 2015, Melanoma: understandind relevant molecular pathways as well as vailable and emerging therapies. Am J Manag Care 21:224–233 Rao AG, Babu VA, Koppada D, Haritha M, Chandana P, 2016, Amelanotic melanoma in the vicinity of acquired melanocytic nevi and not arising from agminated melanocytic nevi: masquerading as pyogenic granuloma. Indian J Dermatol 61(1):122–134 Thomas N, Kricker A,WaxweilerW, Dillon P, Busman K, From L, Groben PA, Armstrong BK, Anton-Culver H, Gruber SB, Marrett LD, Gallagher RP, Zanetti R, Rosso S, Dwyer T, Venn A, Kanetsky PA, Orlow I, Paine S, Ollila DW, Reiner AS, Luo L, Hao H, Frank JS, Begg CB, Berwick M, 2014, Comparison of clinicopathologic features and survival of histopathologically amelanotic and pigmented melanomas: a population-based study. JAMA Dermatol 150(12):1306–1314 Mattia G, Puglisi R, Ascione B, Malorni W, Care A, Matarrese P, 2018, Cell death-based treatments of melanoma: conventional treatments and new therapeutic strategies. Cell Death Dis 9(2):1–14 Bhatia S, Tykodi SA, Thompson JA, 2009, Treatment ofmetastatic melanoma: an overview. Oncology 23(6):488–496 Mohana-Kumaran N, Hill DS, Allen JD, Haass NK, 2014, Targeting the intrinsic apoptosis pathway as a strategy for melanoma therapy. Pigment Cell Melanoma Res 27(4):525–539 Billiard C, 2013, BH3 mimetics: status of the field and new developments. Mol Cancer Ther 12:1691–1700 Nikoletopoulou V, Markaki M, Palikaras K, Tavernarakis N, 2013, Crosstalk between apoptosis, necrosis and autophagy. Biochim Biophys Acta 1833(12):3448–3459 Montero J, Letai A, 2018, Why do BCL-2 inhibitors work and where should we use themin the clinic? CellDeath Differ 25:56–64 Plotz M, Eberle J, 2014, BH3-only proteins: possible proapoptotic triggers for melanoma therapy. Exp Dermatol 23(6):375–378 Mukherjee N, Lu Y, Almeida A, Lambert K, Shiau CW, Su JC, Luo Y, Fujita M, RobinsonWA, Robinson SE, NorrisDA, ShellmanYG, 2017, Use of a MCL-1 inhibitor alone to de-bulk melanoma and in combination to kill melanoma initiating cells. Oncotarget 8(29): 46801–46817 Bandarchi B, Jabbari CA, Vedadi A, Navab R, 2013, Molecular biology of normal melanocytes and melanoma cells. J Clin Pathol 66(8):644–648 Quinn BA, Dash R, Azab B, Sarkar S, Das SK, Kumar S, Oyesanya RA, Dasgupta S, Dent P, Grant S, Rahmani M, Curiel DT, Dmitriev I, Hedvat M, Wei J, Wu B, Stebbins JL, Reed JC, Pellecchia M, Sarkar D, Fisher PB, 2011, Targeting Mcl-1 for the therapy of cancer. Expert Opin Investig Drugs 20(10):1397–1411 Boisvert-Adamo K, LongmateW, Abel EW, Aplin AE, 2009)Mcl- 1 is required for melanoma cell resistance to anoikis. Mol Cancer Res 7(4):549–556 Zhao Y, He J, Li J, Peng X,Wang X, Dong Z, Zhao E, Liu Y,Wu Z, Cui H, 2017, Demethylzeylasteral inhibits cell proliferation and induces apoptosis through suppressing MCL1 in melanoma cells. Cell Death Dis 8(10):e3133 Reuland SN, Goldstein NB, Partyka KA, Smith S, Luo Y, Fujita M, Gonzalez R, Lewisn K, Norris DA, Shellman YG, 2012, ABT-737 synergizes with Bortezomib to killmelanoma cells. Biol Open 1(2): 92–100 Cohen NA, Stewart ML, Gavathiotis E, Tepper LJR, Bruekner SR, Koss B, Opferman JP, Walensky LD, 2012, A competitive stapled peptide screen identifies a selective small molecule that overcomes Mcl-1-dependent leukemia cell survival. Chem Biol 19(9):1175– 1186 Kritzer JA, 2012, The secret of MIM: a novel, Mcl-1-specific small molecule. Chem Biol 19:1082–1083 Wroblewski D, Jiang CC, Croft A, Farrelly ML, ZhangXD, Hersey P, 2013, OBATOCLAX and ABT-737 induce ER stress responses in human melanoma cells that limit induction of apoptosis. PLoS One 8(12):e84073 Thallinger C,Wolschek M,Wacheck V, Maierhofer H, Ginsberg P, Polterauer P, Pehamberger H, Monia BP, Selzer E,Wolff K, Jansen B, 2003, Mcl-1 antisense therapy chemosensitizes human melanoma in a SCID mouse xenotransplantation model. J Invest Dermatol 120(6):1081–1086 Circu ML, Aw TY, 2012, Glutathione and modulation of cell apoptosis. Biochim Biophys Acta 1823(10):1767–1777 Ortega AL, Mena S, Estrela JM, 2011, Glutathione in cancer cell death. Cancers, Basel, 3(1):1285–1310 NicolaMD, Ghibelli L, 2014, Glutathione depletion in survival and apoptotic pathways. Front Pharmacol 5:1–4 Beberok A, Wrzesniok D, Szlachta M, Rok J, Rzepka Z, Respondek M, Buszman E, 2017, Lomefloxacin induces oxidative stress and apoptosis in COLO829 melanoma cells. Int J Mol Sci 18(10):1–18 Pucci B, Kasten M, Giordano A, 2000, Cell cycle and apoptosis. Neoplasia 2(4):291–299 Alenzi FQ, 2004, Links between apoptosis, proliferation and the cell cycle. B J Biomed 61(2):99–102 Al-Qatati A, Aliwaini S, 2017, Combined pitavastatin and dacarbazine treatment activates apoptosis and autophagy resulting in synergistic cytotoxicity in melanoma cells. Oncol Lett 14(6): 7993–7999 Luedtke D, Niu X, Pan Yet al, 2017, Inhibition ofMcl-1 enhances cell death induced by the Bcl-2-selective inhibitor ABT-199 in acute myeloid leukemia cells. Signal Transduct Target Ther 2: 17012 Caenepeel S, Brown SP, Belmontes B et al, 2018, AMG 176, a selective MCL1 inhibitor, is effective in hematologic Cancer models alone and in combination with established therapies. Cancer Discov 8(12):1582–1597 Lucas KM, Mohana-Kumaran N, Lau D, Zhang XD, Hersey P, Huang DC, Weninger W, Haass NK, Allen JD, 2012, Modulation of NOXA and MCL-1 as a strategy for sensitizing melanoma cells to the BH3-mimetic ABT-737. Clin Cancer Res 18(3):783–795 Senft D, Berking C, Graf SA, Kammerbauer C, Ruzicka T, Besch R, 2012, Selective induction of cell death in melanoma cell lines through targeting of Mcl-1 and A1. PLoS One 7(1):e30821 Kelly PN, Strasser A, 2011, The role of Bcl-2 and its pro-survival relatives in tumourigenesis and cancer therapy. Cell Death Differ 18(9):1414–1424 Varadarajan S, Vogler M, Butterworth M, Dinsdale D, Walensky LD, Cohen GM, 2013, Evaluation and critical assessment of putative MCL-1 inhibitors. Cell Death Differ 20(11):1475–1484 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1465 EP 1474 DI 10.1007/s12253-019-00715-z PG 10 ER PT J AU Shao, Y Tao, X Lu, R Zhang, H Ge, J Xiao, B Ye, G Guo, J AF Shao, Yongfu Tao, Xueping Lu, Rongdan Zhang, Haiqiang Ge, Jiaxin Xiao, Bingxiu Ye, Guoliang Guo, Junming TI Hsa_circ_0065149 is an Indicator for Early Gastric Cancer Screening and Prognosis Prediction SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CircRNA; Hsa_circ_0065149; Exosomes; Gastric juice; Biomarker ID CircRNA; Hsa_circ_0065149; Exosomes; Gastric juice; Biomarker AB Circular RNAs (circRNAs) are an endogenous RNAs with a covalently closed cyclic structure. They have emerged recently as key regulators in the development and progression of human cancers. However, the clinical values of most circRNAs in gastric cancer (GC) are unknown. Hsa_circ_0065149, one of the dysregulated circRNAs in gastric carcinogenesis detected by circRNA microarray, was chose as a targeted circRNA in this study. We firstly enlarged sample size and identified the level changes of hsa_circ_0065149 among four stages of gastric tumorigenesis from healthy gastric mucosa, gastritis, intestinal metaplasia to GC. Then, the potential relationship between hsa_circ_0065149 expression levels and GC patients’ clinicopathological factors was investigated. Moreover, the clinical significance of hsa_circ_0065149 in plasma exosomes and gastric juice were explored. Receiver operating characteristic (ROC) curve and Kaplan-Meier survival curve were constructed to evaluate diagnostic and prognostic values. Finally, bioinformatics analysis was performed to excavate the potential functions of hsa_circ_0065149. Hsa_circ_0065149 expression was only significantly down-regulated in gastric cancer, not changed among healthy gastric mucosa and gastritis intestinal metaplasia. Low hsa_circ_0065149 expression levels in GC tissues were significantly associated with tumor diameter (P = 0.034) and perineural invasion (P = 0.037). GC patients with low hsa_circ_0065149 levels had a much longer overall survival than those in high group (P = 0.020). More important, hsa_circ_0065149 levels were significantly decreased in plasma exosomes of early GC patients. As a screening biomarker for early GC, hsa_circ_0065149 in plasma exosomes has higher sensitivity and specificity than traditional clinical biomarkers. Bioinformatics analysis suggest that the abnormal expression of hsa_circ_0065149 may play an important role during gastric carcinogenesis. Those results indicate that hsa_circ_0065149 in exosomes is an indicator for early GC screening and prognosis prediction. C1 [Shao, Yongfu] The Affiliated Hospital of Medical School of Ningbo University, Department of Gastroenterology, 315020 Ningbo, China. [Tao, Xueping] The Affiliated Hospital of Medical School of Ningbo University, Department of Gastroenterology, 315020 Ningbo, China. [Lu, Rongdan] The Affiliated Hospital of Medical School of Ningbo University, Department of Gastroenterology, 315020 Ningbo, China. [Zhang, Haiqiang] The Affiliated Hospital of Medical School of Ningbo University, Department of Gastroenterology, 315020 Ningbo, China. [Ge, Jiaxin] The Affiliated Hospital of Medical School of Ningbo University, Department of Gastroenterology, 315020 Ningbo, China. [Xiao, Bingxiu] Ningbo University School of Medicine, Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, 315211 Ningbo, China. [Ye, Guoliang] The Affiliated Hospital of Medical School of Ningbo University, Department of Gastroenterology, 315020 Ningbo, China. [Guo, Junming] Ningbo University School of Medicine, Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, 315211 Ningbo, China. RP Ye, G (reprint author), The Affiliated Hospital of Medical School of Ningbo University, Department of Gastroenterology, 315020 Ningbo, China. EM ndfyygl@126.com CR Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(4):7–30 Tian M, Chen R, Li T, Xiao B, 2018, Reduced expression of circRNA hsa_circ_0003159 in gastric cancer and its clinical significance. J Clin Lab Anal 32(3):e22281 Wang J, Qu J, Li Z, Che X, Liu J, Teng Y, Jin B, ZhaoM, Liu Y, Qu X, 2018, Pretreatment platelet-to-lymphocyte ratio is associated with the response to first-line chemotherapy and survival in patients with metastatic gastric cancer. J Clin Lab Anal 32(3):e22185 Xie Y, Shao Y, Sun W, Ye G, Zhang X, Xiao B, Guo J, 2018, Downregulated expression of hsa_circ_0074362 in gastric cancer and its potential diagnostic values. Biomark Med 12(1):11–20 Salzman J, 2016, Circular RNA expression: its potential regulation and function. Trends Genet 32(5):309–316 Zhang Y, Liang W, Zhang P, Chen J, Qian H, Zhang X, Xu W, 2017, Circular RNAs: emerging cancer biomarkers and targets. J Exp Clin Cancer Res 36(1):152 Fu L, Jiang Z, Li T, Hu Y, Guo J, 2018, Circular RNAs in hepatocellular carcinoma: functions and implications. Can Med 7:3101– 3109. , DOI 10.1002/cam4.1574 Qu S, Liu Z, Yang X, Zhou J, Yu H, Zhang R, Li H, 2018, The emerging functions and roles of circular RNAs in cancer. Cancer Lett 414:301–309 Kristensen LS, Hansen TB, Veno MT, Kjems J, 2018, Circular RNAs in cancer: opportunities and challenges in the field. Oncogene 37(5):555–565 Shao Y, Li J, Lu R, Li T, Yang Y, Xiao B, Guo J, 2017, Global circular RNA expression profile of human gastric cancer and its clinical significance. Can Med 6(6):1173–1180 Yu Z, Wei L, Peng Z, Chen J, Qian H, Zhang X, Xu W, 2017, Circular RNAs: emerging cancer biomarkers and targets. J Exp Clin Cancer Res 36(1):152 Li H, Hao X,Wang H, Liu Z, He Y, PuM, Zhang H, Yu H, Duan J, Qu S, 2016, Circular RNA expression profile of pancreatic ductal adenocarcinoma revealed by microarray. Cell Physiol Biochem 40(6):1334–1344 Fu L, Yao T, Chen Q, MoX, Hu Y, Guo J, 2017, Screening differential circular rna expression profiles reveals hsa_circ_0004018 is associated with hepatocellular carcinoma. Oncotarget 8(35):58405–58416 Vlachos IS, Zagganas K, Paraskevopoulou MD, Georgakilas G, Karagkouni D, Vergoulis T, Dalamagas T, Hatzigeorgiou AG, 2015, DIANA-miRPath v3.0: deciphering microRNA function with experimental support. Nucleic Acids Res 43(1):460–466 Liu J, Liu T, Wang X, He A, 2017, Circles reshaping the RNA world: from waste to treasure. Mol Cancer 16(1):58 Zhang Y, Liu H, LiW, Yu J, Li J, Shen Z, Ye G, Qi X, Li G, 2017, CircRNA_100269 is downregulated in gastric cancer and suppresses tumor cell growth by targeting miR-630. Aging 9(6): 1585–1594 Li T, ShaoY, Fu L, XieY,Zhu L, SunW,Yu R,Xiao B,Guo J, 2018, Plasma circular RNA profiling of patients with gastric cancer and their droplet digital RT-PCR detection. J Mol Med 96(1):85–96 Wang H, Huang C, Zheng C, Li P, Xie W, Wang J, Lin J, Lu J, 2012, Tumor size as a prognostic factor in patients with advanced gastric cancer in the lower third of the stomach. World J Gastroenterol 18(38):5470–5475 Deng J,YouQ, GaoY,Yu Q, Zhao P, ZhengY, FangW, Xu N, Teng L, 2014, Prognostic value of perineural invasion in gastric cancer: a systematic review and meta-analysis. PLoS One 9(2):e88907 Lu J, Huang C, Zheng C, Li P, Xie J, Wang J, Lin J, 2013, Consideration of tumor size improves the accuracy of TNM predictions in patients with gastric cancer after curative gastrectomy. Surg Oncol 22(3):167–171 Lu J, Huang C, Zheng C, Li P, Xie J,Wang J, Lin J, Chen Q, Cao L, Lin M, 2014, Prognostic value of tumor size in patients with remnant gastric cancer: is the seventh UICC stage sufficient for predicting prognosis? PLoS One 9(12):e115776 Zhou Z, Xu G, Zhang W, Zhao H, Wu Y, 2014, Reevaluating significance of perineural invasion in gastric cancer based on double immunohistochemical staining. Arch Pathol Lab Med 138(2): 229–234 Fan F, Tian Y, Xu G, Liu Z, Liu S, Zheng G, Guo M, Lian X, Fan D, ZhangH, 2017, Diagnostic and prognostic value of CEA, CA19–9, AFP and CA125 for early gastric cancer. BMC Cancer 17(1):737 Whiteside TL, 2015, The potential of tumor-derived exosomes for noninvasive cancer monitoring. Expert Rev Mol Diagn 15(10): 1293–1310 Greening DW, Gopal SK, Xu R, Simpson RJ, Chen W, 2015, Exosomes and their roles in immune regulation and cancer. Semin Cell Dev Biol 40:72–81 Fernando MR, Jiang C, Krzyzanowski GD, Ryan WL, 2017, New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes. PLoS One 12(8):e0183915 Cui L, Zhang X, Ye G, Zheng T, Song H, Deng H, Xiao B, Xia T, Yu X, Le Y, Guo J, 2013, Gastric juice MicroRNAs as potential biomarkers for the screening of gastric cancer. Cancer 119(9):1618–1626 Rong D, Sun H, Li Z, Liu S, Dong C, Fu K, TangW, Cao H, 2017, An emerging function of circRNA-miRNAs-mRNA axis in human diseases. Oncotarget 8(42):73271–73281 Cheng J, Zhuo H, Xu M,Wang L, Xu H, Peng J, Hou J, Lin L, Cai J, 2018, Regulatory network of circRNA-miRNA-mRNA contributes to the histological classification and disease progression in gastric cancer. J Transl Med 16(1):216 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1475 EP 1482 DI 10.1007/s12253-019-00716-y PG 8 ER PT J AU Picasso, R Tagliafico, A Calabrese, M Martinoli, C Pistoia, F Rossi, A Zaottini, F Derchi, L AF Picasso, Riccardo Tagliafico, Alberto Calabrese, Massimo Martinoli, Carlo Pistoia, Federico Rossi, Anna Zaottini, Federico Derchi, Lorenzo TI Primary and Secondary Breast Lymphoma: Focus on Epidemiology and Imaging Features SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast; Lymphoma; Epidemiology; Imaging ID Breast; Lymphoma; Epidemiology; Imaging AB Aim of this study was to select all the cases of Primary (PBL) and Secondary (SBL) Breast Lymphoma from our breast unit since 01/01/2000, to obtain up-to-date data on the prevalence of this rare pathology and to analyze imaging features, with a special focus on CT. All pathological reports of breast biopsies performed from 01/01/2000 to 01/01/2019 were at first screened. Among them, we performed two different researches, looking for key words suggesting either a diagnosis of lymphoma or any other malignant disease. Using the Wiseman criteria, we identify PBL and SBL. All imaging features of PBL and SBL were analyzed. Prevalence of lymphoma amongst suspicious breast masses and amongst all breast malignancies were calculated. Out of 42,505 histopathology reports from mammary nodule biopsies, we found 19,354 malignancies. We were able to identify 11 patients affected by PBL (0,03% of suspicious breast lesions, 0.06% of breast malignancies), and 23 cases of SBL (0,05% of suspicious breast lesions, 0,12%of breast malignancies).Most common isotype in PBL was DLBC lymphoma, whereas in SBL that resulted Follicular lymphoma. In PBL group, we were able to retrieve images 7 CT or CT-PET study performed at diagnosis 7 US, 1 mammography and 1 MR. In SBL group, we analyzed 14 CT/CT-PET examinations, 11 US studies and 3 mammography. PBL and SBL are rarer than considered until now. There is no definite imaging characteristic able to distinguish between these two pathologies and among them and breast cancer. C1 [Picasso, Riccardo] University of Genova, Department of Health Sciences (DISSAL), Radiology Section, Via Pastore, 1-16132 Genova, Italy. [Tagliafico, Alberto] University of Genova, Department of Health Sciences (DISSAL), Radiology Section, Via Pastore, 1-16132 Genova, Italy. [Calabrese, Massimo] Ospedale Policlinico San MartinoGenova, Italy. [Martinoli, Carlo] University of Genova, Department of Health Sciences (DISSAL), Radiology Section, Via Pastore, 1-16132 Genova, Italy. [Pistoia, Federico] University of Genova, Department of Health Sciences (DISSAL), Radiology Section, Via Pastore, 1-16132 Genova, Italy. [Rossi, Anna] Ospedale Policlinico San MartinoGenova, Italy. [Zaottini, Federico] University of Genova, Department of Health Sciences (DISSAL), Radiology Section, Via Pastore, 1-16132 Genova, Italy. [Derchi, Lorenzo] University of Genova, Department of Health Sciences (DISSAL), Radiology Section, Via Pastore, 1-16132 Genova, Italy. RP Picasso, R (reprint author), University of Genova, Department of Health Sciences (DISSAL), Radiology Section, 1-16132 Genova, Italy. EM Riccardo.picasso@gmail.com CR Sabate JM, Gomez A, Torrubia S et al, 2002, Lymphoma of the breast: clinical and radiologic features with pathologic correlation in 28 patients. Breast J 8:294–304 Wiseman C, Liao K, 1972, Primary lymphoma of the breast. Cancer 29:1705–1712 Hugh JC, Jackson F, Hanson J, Poppema S, 1990, Primary breast lymphoma. An immunohistologic study of 20 new cases. Cancer 66:2602–2611 Avenia N, Sanguinetti A, Cirocchi R, Bistoni G, Trastulli S, D'Ajello F, Barberini F, Cavallaro G, Rulli A, Sidoni A, Noya G, de Toma G, Sciannameo F, 2010, Primary breast lymphomas: a multi- centric experience. World J Surg Oncol 8:53 Giardini R, Piccolo C, Rilke F. Primary non-Hodgkin's lymphomas of the female breast. Cancer 1992;69:725±35, 735 Mattia AR, Ferry JA, Harris NL, 1993, Breast lymphoma: a B-cell spectrum including low-grade B-cell lymphoma of mucosaassociated lymphoid tissue. Am J Surg Pathol 17:574–587 Brogi E, Harris NL, 1999, Lymphomas of the breast: pathology and clinical behavior. Semin Oncol 26:357–364 Topalovski M, Cristan D, Mattson JC, 1999, Lymphoma of the breast: a clinicopathologic study of primary and secondary cases. Arch Pathol Lab Med 123:1208–1218 Freeman C, Berg JW, Cutler SJ, 1972, Occurrence and prognosis of extranodal lymphomas. Cancer 29:252–260 YhimHY,Kang HJ, ChoiYH, KimSJ,KimWS, ChaeYS, KimJS, ChoiCW, Oh SY, Eom HS, Kim JA, Lee JH,Won JH, Shim H, Lee JJ, Sung HJ, Kim HJ, Lee DH, Suh C, Kwak JY, 2010, Clinical outcomes and prognostic factors in patients with breast diffuse large B cell lymphoma: consortium for improving survival of lymphoma, CISL, study. BMC Cancer 10:321 Franco Perez F, Lavernia J et al, 2016, Primary breast lymphoma: analysis of 55 cases of the Spanish lymphoma oncology group. Clinical Lymphoma, Myeloma & Leukemia 17(3):186–191 Cheah CY, Campbell B, Seymour J, 2014, Primary breast lymphoma. Cancer Treat Rev 40:900–908 Aviv A, Tadmor T, Polliack A, 2013, Primary diffuse large B-cell lymphoma of the breast: looking at pathogenesis, clinical issues and therapeutic options. Ann Oncol 24:2236–2244 Caon J, Wai E, Hart J et al, 2012, Treatment and outcomes of primary breast lymphoma. Clin Breast Cancer 12:412–419 Surov A, Holzhausen H-J, Wienke A, Schmidt J, Thomssen C, Arnold D, Ruschke K, Spielmann R-P, 2012, Primary and secondary breast lymphoma: prevalence, clinical signs and radiological features. Br J Radiol 85:e195–e205 Validire P, Capovilla C, Asselain B et al, 2009, Primary breast non- Hodgkin’s lymphoma: a large single center study of initial characteristics, natural history, and prognostic factors. Am J Hematol 84: 133–139 Jeanneret-Sozzi W, Taghian A, Epelbaum R, 2008, Primary breast lymphoma: patient profile, outcome and prognostic factors. A multicentre rare cancer network study. BMC Cancer 8:86–92 Liu FF, Clark RM, 1986, Primary lymphoma of the breast. Clin Radiol 37:567–570 Yang WT, Lane DL, LePetross HT, Abruzzo LV, Macapinlac HA, 2007, Breast lymphoma: imaging findings of 32 tumors in 27 patients. Radiology 245:692–702 Balu-Maestro C, Bruneton JN, Rogopoulos A, Marcy PY, Guidicelli T, Raffaelli C et al, 1992, Mammographic and ultrasonographic appearances of lymphoma of the breast. Eur Radiol 2: 565–569 Liberman L, Giess CS, Dershaw DD, Louie DC, Deutch BM, 1994, Non-Hodgkin lymphoma of the breast: imaging characteristics and correlation with histopathologic findings. Radio- logy 192:157–160 Surov A, Holzhausen HJ, Wienke A, Schmidt J, Thomssen C, Arnold D, Ruschke K, Spielmann RP, 2012, Primary and secondary breast lymphoma: prevalence, clinical signs and radiological features. Br J Radiol 85:e195–e205 Jernstrom P, Sether JM, 1967, Primary lymphosarcoma of the mammary gland. JAMA 201:503–506 Inoue M, Sano R,Watal R et al, 2003, Dynamic multi- detector CT of breast tumors: diagnostic features and comparison with conventional techniques. AJR Am J Roentgenol 181:679–686 Harvey JA,MahoneyMC, NewellMS, Bailey L, Barke LD,D’Orsi C, Hayes MK, Jokich PM, Lee SJ, Lehman CD, Mainiero MB, Mankoff DA, Patel SB, Reynolds HE, Sutherland ML, Haffty BG, 2013, ACR appropriateness criteria palpable breast masses. J Am Coll Radiol 10:742–749 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1483 EP 1488 DI 10.1007/s12253-019-00730-0 PG 6 ER PT J AU Malik, UU Siddiqui, AI Ilyas, A Hashim, Z Staunton, L Kwasnik, A Pennington, RS Shamshad, Z AF Malik, Urooj Uzma Siddiqui, Ather Imtiaz Ilyas, Amber Hashim, Zehra Staunton, Lisa Kwasnik, Anna Pennington, R Stephen Shamshad, Zarina TI Identification of Differentially Expressed Proteins from Smokeless Tobacco Addicted Patients Suffering from Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral squamous cell carcinoma; Tissue proteomics; Differential expression; Biomarker; Smokeless tobacco ID Oral squamous cell carcinoma; Tissue proteomics; Differential expression; Biomarker; Smokeless tobacco AB Oral squamous cell carcinoma (OSCC) is the eight most common malignancy worldwide with an incidence rate of 40%in southeast Asia. Lack of effective diagnostic tools at early stage and disease recurrence despite extensive treatments are main reasons for high mortality and low survival rates. The aim of current study was to identify differentially expressed proteins to explore potential candidate biomarkers having diagnostic significance. We performed comparative proteomic analysis of paired protein samples (cancerous buccal mucosa and adjacent normal tissue) from OSCC patients using a combination of two dimensional gel electrophoresis and Mass spectrometric analysis. On the basis of spot intensity, seventeen proteins were found to be consistently differentially expressed among most of the samples which were identified through mass spectrometry. For validation of identified proteins, expression level of stratifin was determined using immuno-histochemistry and Western blot analysis. All identified proteins were analyzed by STRING to explore their interaction. Among uniquely identified proteins in this study, at least two candidate markers (Ig Kappa chain C region and Isoform 2 of fructose bisphosphate aldolase A) were found to be novel with respect to OSCC which can be explored further. Results presented in current study are likely to contribute in understanding the involvement of these molecules in carcinogenesis apart from their plausible role as diagnostic/prognostic markers. C1 [Malik, Urooj Uzma] University of Karachi, National Center for Proteomics, 75270 Karachi, Pakistan. [Siddiqui, Ather Imtiaz] Jinnah Postgraduate MedicalKarachi, Pakistan. [Ilyas, Amber] University of Karachi, National Center for Proteomics, 75270 Karachi, Pakistan. [Hashim, Zehra] University of Karachi, National Center for Proteomics, 75270 Karachi, Pakistan. [Staunton, Lisa] University College Dublin, School of Medicine and Medical Science, UCD Conway Institute of Biomolecular and Biomedical ResearchDublin, Ireland. [Kwasnik, Anna] University College Dublin, School of Medicine and Medical Science, UCD Conway Institute of Biomolecular and Biomedical ResearchDublin, Ireland. [Pennington, R Stephen] University College Dublin, School of Medicine and Medical Science, UCD Conway Institute of Biomolecular and Biomedical ResearchDublin, Ireland. [Shamshad, Zarina] University of Karachi, National Center for Proteomics, 75270 Karachi, Pakistan. RP Shamshad, Z (reprint author), University of Karachi, National Center for Proteomics, 75270 Karachi, Pakistan. EM szarina@uok.edu.pk CR Ferlay J, Soerjomataram R, Dikshit R, Eser S, Mathers C, RebeloM et al, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:E359–E386 Johnson NW, Jayasekara P, Amarasinghe AA, 2011, Squamous cell carcinoma and precursor lesions of the oral cavity: epidemiology and aetiology. Periodontol 2000 57:19–37 Ni YH, Ding L, Hu QG, Hua ZC, 2015, Potential biomarkers for oral squamous cell carcinoma: proteomics discovery and clinical validation. Proteomics Clin Appl 9:86–97 Zygogianni AG, Kyrgias G, Karakitsos P, Psyrri A, Kouvaris J, Kelekis N, Kouloulias V, 2011, Oral squamous cell cancer: early detection and the role of alcohol and smoking. Head Neck Oncol 3: 2 Liaquat N, Jaffar AM, Haroon MZ, Khan MB, Habib H, 2016, Knowledge and perception of areca/smokeless tobacco users about oral cancer. J Ayub Med Coll Abbottabad 28:164–167 Imam SZ, Nawaz H, Sepah YJ, Pabaney AH, Ilyas M, Ghaffar S, 2007, Use of smokeless tobacco among groups of Pakistani medical students - a cross sectional study. BMC Public Health 7:231– 231 de Camargo CM, Voti L, Guerra-Yi M, Chapuis F, Mazuir M, Curado MP, 2010, Oral cavity cancer in developed and in developing countries: population-based incidence. Head Neck 32:357– 367 Nair U, Bartsch H, Nair J, 2004, Alert for an epidemic of oral cancer due to use of the betel quid substitutes gutkha and pan masala: a review of agents and causative mechanisms. Mutagenesis 19:251–262 Alamgir M, Jamal Q, Jafarey N, Mi r z a T, 2013, Clinicopathological parameters of 50 oral squamous cell carcinoma cases in Karachi. Pak J Medic Dentis 2:3–8 Johann DJ, McGuigan MD, Patel AR, Tomov S, Ross S, Conrads TP et al, 2004, Clinical proteomics and biomarker discovery. Ann N YAcad Sci 1022:295–305 G.A.o.t.W.M. Association, 2014)World medical association declaration of Helsinki: ethical principles for medical research involving human subjects. J Amer College Dentists 81:14 Deschler DG, Day T, 2008, TNM staging of head and neck cancer and neck dissection classification. Amer Acad Otolaryngol–Head & Neck Surg Found:10–23 Shevchenko A, Tomas H, Havli J, Olsen JV,MannM(2006, In-gel digestion for mass spectrometric characterization of proteins and proteomes. Nat Protoc 1:2856–2860 Reed NA, Oh D-J, Czymmek KJ, Duncan MK, 2001, An immunohistochemical method for the detection of proteins in the vertebrate lens. J Immunol Methods 253:243–252 Wang Z, Jiang L, Huang C, Li Z, Chen L, Gou L, Chen P, Tong A, Tang M, Gao F, Shen J, Zhang Y, Bai J, Zhou M, Miao D, Chen Q, 2008, Comparative proteomics approach to screening of potential diagnostic and therapeutic targets for oral squamous cell carcinoma. Mol Cell Proteomics 7:1639–1650 Dorsey K, Agulnik M, 2013, Promising new molecular targeted therapies in head and neck cancer. Drugs 73:315–325 Pandey JP, Kistner-Griffin E, Black L, Namboodiri AM, Iwasaki I, Kasuga Y et al, 2014, IGKC and FcγR genotypes and humoral immunity to HER2 in breast cancer. Immunobiology 219:113–117 Whiteside TL, Ferrone S, 2012, For breast cancer prognosis, immunoglobulin kappa chain surfaces to the top. Clin Cancer Res 18: 2417–2419 Schmidt M, Hellwig B, Hammad S, Othman A, Lohr M, Chen Z, Boehm D, Gebhard S, Petry I, Lebrecht A, Cadenas C,Marchan R, Stewart JD, Solbach C, Holmberg L, Edlund K, Kultima HG, Rody A, Berglund A, LambeM, Isaksson A, Botling J, Karn T,Muller V, Gerhold-Ay A, Cotarelo C, Sebastian M, Kronenwett R, Bojar H, Lehr HA, Sahin U, Koelbl H, Gehrmann M, Micke P, Rahnenfuhrer J, Hengstler JG, 2012, A comprehensive analysis of human gene expression profiles identifies stromal immunoglobulin κ C as a compatible prognostic marker in human solid tumors. Clin Cancer Res 18:2695–2703 Chen X, Yang TT, Zhou Y, Wang W, Qiu XC, Gao J, Li CX, Long H, Ma BA, Ma Q, Zhang XZ, Yang LJ, Fan QY, 2014, Proteomic profiling of osteosarcoma cells identifies ALDOA and SULT1A3 as negative survival markers of human osteosarcoma. Mol Carcinog 53:138–144 Soga T, 2013, Cancer metabolism: key players in metabolic reprogramming. Cancer Sci 104:275–281 Ma D, Chen X, Zhang PY, Zhang H, Wei LJ, Hu S et al, 2017, Upregulation of the ALDOA/DNA-PK/p53 pathway by dietary restriction suppresses tumor growth. Oncogene 37:1041 Ye F, Chen Y, Xia L, Lian J, Yang S, 2018, Aldolase a overexpression is associated with poor prognosis and promotes tumor progression by the epithelial-mesenchymal transition in colon cancer. Biochem Biophys Res Commun 497:639–645 Li C, Xiao Z, Chen Z, Zhang X, Li J,Wu X, Li X, Yi H, LiM, Zhu G, Liang S, 2006, Proteome analysis of human lung squamous carcinoma. Proteomics 6:547–558 Du S, Guan Z, Hao L, Song Y, Wang L, Gong L et al, 2014, Fructose-bisphosphate aldolase a is a potential metastasisassociated marker of lung squamous cell carcinoma and promotes lung cell tumorigenesis and migration. PLoS One 9:e85804 Frei E, 2011, Albumin binding ligands and albumin conjugate uptake by cancer cells. Diabetol Metab Syndr 3:11 Stehle G, Sinn H, Wunder A, Schrenk HH, Stewart JC, Hartung G et al, 1997, Plasma protein, albumin, catabolism by the tumor itself—implications for tumor metabolism and the genesis of cachexia. Crit Rev Oncol Hematol 26:77–100 Gupta D, Lis CG, 2010, Pretreatment serum albumin as a predictor of cancer survival: a systematic review of the epidemiological literature. Nutr J 9:69 Thiel UJE, Feltens R, Adryan B, Gieringer R, Brochhausen C, Schuon R, Fillies T, Grus F, Mann WJ, Brieger J, 2011, Analysis of differentially expressed proteins in oral squamous cell carcinoma by MALDI-TOF MS. J Oral Pathol Med 40:369–379 Alam H, Gangadaran P, Bhate AV, Chaukar DA, Sawant SS, Tiwari R et al, 2011, Loss of keratin 8 phosphorylation leads to increased tumor progression and correlates with clinico-pathological parameters of OSCC patients. PLoS One 6:e27767 Wei KJ, Zhang L, Yang X, Zhong LP, Zhou XJ, Pan HY, Li J, Chen WT, Zhang ZY, 2009, Overexpression of cytokeratin 17 protein in oral squamous cell carcinoma in vitro and in vivo. Oral Dis 15:111– 117 Xu L, Chen S, Bergan R, 2006, MAPKAPK2 and HSP27 are downstream effectors of p38 MAP kinase-mediated matrix metalloproteinase type 2 activation and cell invasion in human prostate cancer. Oncogene 25:2987–2998 Lo Muzio L, Leonardi R, Mariggio M, Mignogna M, Rubini C, Vinella A et al, 2004, HSP 27 as possible prognostic factor in patients with oral squamous cell carcinoma. Histol Histopathol 19:119–128 Zamanian M, Veerakumarasivam A, Abdullah S, Rosli R, 2013, Calreticulin and cancer. Pathol & Oncol Res 19:149–154 Chiang WF, Hwang TZ, Hour TC, Wang LH, Chiu CC, Chen HR et al, 2013, Calreticulin, an endoplasmic reticulum-resident protein, is highly expressed and essential for cell proliferation and migration in oral squamous cell carcinoma. Oral Oncol 49:534–541 Benzinger A, Muster N, Koch HB, Yates JR, Hermeking H, 2005, Targeted proteomic analysis of 14-3-3ς, a p53 effector commonly silenced in cancer. Mol Cell Proteomics 4:785–795 Badea L, Herlea V, Dima SO, Dumitrascu T, Popescu I, 2008, Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia-the authors reported a combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia. Hepato-Gastroenterology 55: 2016 Perathoner A, Pirkebner D, Brandacher G, Spizzo G, Stadlmann S, Obrist P, Margreiter R, Amberger A, 2005, 14-3-3σ expression is an independent prognostic parameter for poor survival in colorectal carcinoma patients. Clin Cancer Res 11:3274–3279 Laimer K, Blassnig N, Spizzo G, Kloss F, Rasse M, Obrist P, Schafer G, Perathoner A, Margreiter R, Amberger A, 2009, Prognostic significance of 14-3-3σ expression in oral squamous cell carcinoma, OSCC). Oral Oncol 45:127–134 Hermeking H, Lengauer C, Polyak K, He TC, Zhang L, Thiagalingam S et al, 1997, 14-3-3σIs a p53-regulated inhibitor of G2/M progression. Mol Cell 1:3–11 Yang HY, Wen YY, Chen CH, Lozano G, Lee MH, 2003, 14-3-3σ positively regulates p53 and suppresses tumor growth. Mol Cell Biol 23:7096–7107 Rothenberg SM, Ellisen LW(2012, The molecular pathogenesis of head and neck squamous cell carcinoma. J Clin Invest 122:1951– 1957 Chanthammachat P, PromwikornW, Pruegsanusak K, Roytrakul S, Srisomsap C, Chokchaichamnankit D, Svasti J, Boonyaphiphat P, K S, Thongsuksai P, 2013, Comparative proteomic analysis of oral squamous cell carcinoma and adjacent non-tumour tissue from Thailand. Arch Oral Biol 58:1677–1685 Ralhan R, Masui O, DeSouza LV,Matta A, Macha M, Siu K, 2011, Identification of proteins secreted by head and neck cancer cell lines using LC-MS/MS: strategy for discovery of candidate serological biomarkers. Proteomics 11:2363–2376 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1489 EP 1497 DI 10.1007/s12253-019-00724-y PG 9 ER PT J AU Tokes, T Tokes, AM Szentmartoni, Gy Kiszner, G Muhl, D Molnar, B Kulka, J Krenacs, T Dank, M AF Tokes, Timea Tokes, Anna-Maria Szentmartoni, Gyongyver Kiszner, Gergo Muhl, Dorottya Molnar, Akos Bela Kulka, Janina Krenacs, Tibor Dank, Magdolna TI Prognostic and Clinicopathological Correlations of Cell Cycle Marker Expressions before and after the Primary Systemic Therapy of Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Primary systemic therapy; Cell cycle; Ki67; MCM; CyclinA; PHH3 ID Breast cancer; Primary systemic therapy; Cell cycle; Ki67; MCM; CyclinA; PHH3 AB We aimed to analyze the expression of cell-cycle regulation markers – minichromosome maintenance protein 2 (MCM2), Ki-67, Cyclin-A and phosphohistone-H3 (PHH3) − in pre-treatment core-biopsy samples of breast carcinomas in correlation with known predictive and prognostic factors. Totally 52 core biopsy samples obtained prior to neoadjuvant therapy were analyzed. Immunohistochemistry was performed to analyze the expression of MCM2, Ki- 67, Cyclin A and PHH3, which were correlated with the following clinicopathological parameters: clinical TNM, tumor grade, biological subtype, the presence of tumor infiltrating lymphocytes (TIL), pathological tumor response rate to the neoadjuvant therapy and patient survival. All investigated markers showed higher expression in high grade and in triple negative tumors (p < 0.01 and p < 0.05, respectively). Hormone receptor negative tumors showed significantly higher expression of Ki-67 (p < 0.01), MCM2 (p < 0.01) and Cyclin A (p < 0.01) than hormone receptor positive ones. Tumors with increased TIL showed significantly higher Ki-67 expression (p = 0.04). Pattern analysis suggested that novel cell-cycle marker-based subgrouping reveals predictive and prognostic potential. Tumors with high MCM2, Cyclin A or PHH3 expression showed significantly higher rate of pathological complete remission. Tumors with early relapse (progression-free survival ≤2 years) and shortened overall survival also show a higher rate of proliferation. Our cell cycle marker (Ki-67, MCM2, Cyclin A, PHH3) based testing could identify tumors with worse prognosis, but with a favorable response to primary systemic therapy. The pattern of cell-cycle activity could also be useful for predicting early relapse, but our findings need to be further substantiated in larger patient cohorts. C1 [Tokes, Timea] Semmelweis University, Department of Oncology, Tomo utca 25-29, 4th floor, H-1083 Budapest, Hungary. [Tokes, Anna-Maria] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Szentmartoni, Gyongyver] Semmelweis University, Department of Oncology, Tomo utca 25-29, 4th floor, H-1083 Budapest, Hungary. [Kiszner, Gergo] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Muhl, Dorottya] Semmelweis University, Department of Oncology, Tomo utca 25-29, 4th floor, H-1083 Budapest, Hungary. [Molnar, Akos Bela] Semmelweis University, 1st Department of Surgery, Ulloi ut 78/A, H-1083 Budapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Dank, Magdolna] Semmelweis University, Department of Oncology, Tomo utca 25-29, 4th floor, H-1083 Budapest, Hungary. RP Tokes, T (reprint author), Semmelweis University, Department of Oncology, H-1083 Budapest, Hungary. EM timi.tokes@gmail.com;tokes.timea@med.semmelweis-univ.hu CR Goldhirsch A, Wood WC, Gelber RD, Coates AS, Thurlimann B, Senn HJ, Gallen S, 2007, Progress and promise: highlights of the international expert consensus on the primary therapy of early breast cancer. Ann Oncol 18:1133–1144 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B et al, 2013, Personalizing the treatment of women with early breast cancer: highlights of the St Gallen international expert consensus on the primary therapy of early breast cancer. Ann Oncol 24:2206–2223 Kaufmann M, von Minckwitz G, Bear HD, Buzdar A, McGale P, Bonnefoi H et al, 2007, Recommendations from an international expert panel on the use of neoadjuvant, primary, systemic treatment of operable breast cancer: new perspectives. Ann Oncol 18:1927–1934 Colozza M, Azambuja E, Cardoso F, Sotiriou C, Larsimont D, Piccart MJ, 2005, Proliferative markers as prognostic and predictive tools in early breast cancer: where are we now? Ann Oncol 16:1723–1739 Bottini A, Berruti A, Bersiga A, Brizzi MP, Bruzzi P, Aguggini S et al, 2001, Relationship between tumour shrinkage and reduction in Ki67 expression after primary chemotherapy in human breast cancer. Br J Cancer 85:1106–1112 Polley MY, Leung SC, McShane LM, Gao D, Hugh JC, Mastropasqua MG et al, 2013, International Ki67 in Breast Cancer Working Group of the Breast International G, North American Breast Cancer G. An international Ki67 reproducibility study. J Natl Cancer Inst 105:1897–1906 Loddo M, Kingsbury SR, RashidM, Proctor I, Holt C,Young J et al, 2009, Cell-cycle-phase progression analysis identifies unique phenotypes of major prognostic and predictive significance in breast cancer. Br J Cancer 100:959–970 Shetty A, Loddo M, Fanshawe T, Prevost AT, Sainsbury R, Williams GH et al, 2005, DNA replication licensing and cell cycle kinetics of normal and neoplastic breast. Br J Cancer 93:1295–1300 Tokes AM, Szasz AM, Geszti F, Lukacs LV, Kenessey I, Turanyi E et al, 2015, Expression of proliferation markers Ki67, cyclin A, geminin and aurora-kinase A in primary breast carcinomas and corresponding distant metastases. J Clin Pathol 68:274–282 Whitfield ML, George LK, Grant GD, Perou CM, 2006, Common markers of proliferation. Nat Rev Cancer 6:99–106 Endl E, Gerdes J, 2000, The Ki-67 protein: fascinating forms and an unknown function. Exp Cell Res 257:231–237 Williams GH, Stoeber K, 2012, The cell cycle and cancer. J Pathol 226:352–364 Lei M, 2005, The MCM complex: its role in DNA replication and implications for cancer therapy. Curr Cancer Drug Targets 5:365–380 Vermeulen K, Van Bockstaele DR, Berneman ZN, 2003, The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer. Cell Prolif 36:131–149 Bossard C, Jarry A, Colombeix C, Bach-Ngohou K, Moreau A, Loussouarn D et al, 2006, Phosphohistone H3 labelling for histoprognostic grading of breast adenocarcinomas and computerassisted determination of mitotic index. J Clin Pathol 59:706–710 Skaland I, Janssen EA, Gudlaugsson E, Hui Ru Guo L, Baak JP, 2009, The prognostic value of the proliferation marker phosphohistone H3, PPH3, in luminal, basal-like and triple negative phenotype invasive lymph node-negative breast cancer. Cell Oncol 31:261–271 Skaland I, Janssen EA, Gudlaugsson E, Klos J, Kjellevold KH, Soiland H, Baak JP, 2009, Validating the prognostic value of proliferation measured by Phosphohistone H3, PPH3, in invasive lymph node-negative breast cancer patients less than 71 years of age. Breast Cancer Res Treat 114:39–45 de Azambuja E, Cardoso F, de Castro G Jr, Colozza M, Mano MS, Durbecq Vet al, 2007, Ki-67 as prognostic marker in early breast cancer: a meta-analysis of published studies involving 12,155 patients. Br J Cancer 96:1504–1513 Inwald EC, Klinkhammer-Schalke M, Hofstadter F, Zeman F, Koller M, Gerstenhauer M et al, 2013, Ki-67 is a prognostic parameter in breast cancer patients: results of a large population-based cohort of a cancer registry. Breast Cancer Res Treat 139:539–552 Balmativola D, Marchio C, Maule M, Chiusa L, Annaratone L, Maletta F et al, 2014, Pathological non-response to chemotherapy in a neoadjuvant setting of breast cancer: an inter-institutional study. Breast Cancer Res Treat 148:511–523 Tokes T, Tokes AM, Szentmartoni G, Kiszner G,Madaras L, Kulka J et al, 2016, Expression of cell cycle markers is predictive of the response to primary systemic therapy of locally advanced breast cancer. Virchows Archiv 468:675–686 Fasching PA, Heusinger K, Haeberle L, Niklos M, Hein A, Bayer CM et al, 2011, Ki67, chemotherapy response, and prognosis in breast cancer patients receiving neoadjuvant treatment. BMC Cancer 11:486 Acs B, Zambo V, Vizkeleti L, Szasz AM, Madaras L, Gy S et al, 2017, Ki-67 as a controversial predictive and prognostic marker in breast cancer patients treated with neoadjuvant chemotherapy. Diagn Pathol 12:20 Ahlin C, Zhou W, Holmqvist M, Holmberg L, Nilsson C, Jirstrom K et al, 2009, Cyclin A is a proliferative marker with good prognostic value in node-negative breast cancer. Cancer Epidemiol Biomarkers Prev 18:2501–2506 Strand C, Ahlin C, Bendahl PO, Fjallskog ML, Hedenfalk I, Malmstrom P et al, 2012, Combination of the proliferation marker cyclin A, histological grade, and estrogen receptor status in a new variable with high prognostic impact in breast cancer. Breast Cancer Res Treat 131:33–40 Poikonen P, Sjostrom J, Amini RM, Villman K, Ahlgren J, Blomqvist C, 2005, Cyclin A as a marker for prognosis and chemotherapy response in advanced breast cancer. Br J Cancer 93: 515–519 Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G et al, 2015, The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: recommendations by an International TILsWorking Group 2014. Ann Oncol 26:259–271 Cserni G, Francz M, Jaray B, Kalman E, Kovacs I, Kulka J, Orosz Z, Udvarhelyi N, Vass L, 2010, Pathological diagnostic, handling and histological findings in breast cancer. Magy Onkol 54:2017–2226 Pinder SE, Provenzano E, Earl H, Ellis IO, 2007, Laboratory handling and histology reporting of breast specimens from patients who have received neoadjuvant chemotherapy. Histopathology 50:409–417 Jones RL, Lakhani SR, Ring AE, Ashley S, Walsh G, Smith IE, 2006, Pathological complete response and residual DCIS following neoadjuvant chemotherapy for breast carcinoma. Br J Cancer 94:358–362 Allred DC, Harvey JM, Berardo M, Clark GM, 1998, Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11:155–168 Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ et al, 2007, American Society of Clinical Oncology/ College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol 25:118–145 Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH et al, 2013, Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol 31:3997–4013 DowsettM, Nielsen TO, A'Hern R, Bartlett J, Coombes RC, Cuzick J et al, 2011, Assessment of Ki67 in breast cancer: recommendations from the International Ki67 in Breast Cancer working group. J Natl Cancer Inst 103:1656–1664 Pathmanathan N, Balleine RL, 2013, Ki67 and proliferation in breast cancer. J Clin Pathol 66:512–516 Ali HR, Dawson SJ, Blows FM, Provenzano E, Pharoah PD, Caldas C, 2012, Aurora kinase A outperforms Ki67 as a prognostic marker in ER-positive breast cancer. Br J Cancer 106:1798–1806 Wiesner FG,Magener A, Fasching PA,Wesse J, Bani MR, Rauh C et al, 2009, Ki-67 as a prognostic molecular marker in routine clinical use in breast cancer patients. Breast 18:135–141 Haerslev T, Jacobsen GK, Zedeler K, 1996, Correlation of growth fraction by Ki-67 and proliferating cell nuclear antigen, PCNA, immunohistochemistry with histopathological parameters and prognosis in primary breast carcinomas. Breast Cancer Res Treat 37:101–113 Baak JP, Gudlaugsson E, Skaland I, Guo LH, Klos J, Lende TH et al, 2009, Proliferation is the strongest prognosticator in node-negative breast cancer: significance, error sources, alternatives and comparison with molecular prognostic markers. Breast Cancer Res Treat 115: 241–254 Ahlin C, Aaltonen K, Amini RM, Nevanlinna H, Fjallskog ML, Blomqvist C, 2007, Ki67 and cyclin A as prognostic factors in early breast cancer. What are the optimal cut-off values? Histopathology 51:491–498 Trihia H, Murray S, Price K, Gelber RD, Golouh R, Goldhirsch A et al, 2013, International Breast Cancer Study G. Ki-67 expression in breast carcinoma: its association with grading systems, clinical parameters, and other prognostic factors–a surrogate marker? Cancer 97:1321–1331 Cheang MC, Chia SK, Voduc D, Gao D, Leung S, Snider J et al, 2009, Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 101:736–750 Nishimura R, Osako T, Okumura Y, Hayashi M, Toyozumi Y, Arima N, 2010, Ki-67 as a prognostic marker according to breast cancer subtype and a predictor of recurrence time in primary breast cancer. Exp Ther Med 1:747–754 Keam B, Im SA, Lee KH, Han SW, Oh DY, Kim JH et al, 2011, Ki- 67 can be used for further classification of triple negative breast cancer into two subtypes with different response and prognosis. Breast Cancer Res 13:R22 Mrklic I, Capkun V, Pogorelic Z, Tomic S, 2013, Prognostic value of Ki-67 proliferating index in triple negative breast carcinomas. Pathol Res Pract 209:296–301 Stanton SE, Disis ML, 2016, Clinical significance of tumorinfiltrating lymphocytes in breast cancer. J Immunother Cancer 24:59 Chung YR, Kim HJ, JangMH, Park SY, 2017, Prognostic value of tumor infiltrating lymphocyte subsets in breast cancer depends on hormone receptor status. Breast Cancer Res Treat 161:409–420 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1499 EP 1510 DI 10.1007/s12253-019-00726-w PG 12 ER PT J AU Wangmo, Ch Charoen, N Jantharapattana, K Dechaphunkul, A Thongsuksai, P AF Wangmo, Chimi Charoen, Nattinee Jantharapattana, Kitti Dechaphunkul, Arunee Thongsuksai, Paramee TI Epithelial–Mesenchymal Transition Predicts Survival in Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Epithelial–mesenchymal transition; E-cadherin; Vimentin; p16; Oral squamous cell carcinoma; Prognosis; Immunohistochemistry ID Epithelial–mesenchymal transition; E-cadherin; Vimentin; p16; Oral squamous cell carcinoma; Prognosis; Immunohistochemistry AB Synergistic loss of E-cadherin and acquisition of vimentin are characteristic feature of epithelial–mesenchymal transition (EMT) which confers an invasive phenotype of epithelial cancer cells. The aim of the study was to evaluate the prognostic significance of E-cadherin and vimentin expression individually and in combination as a measure of epithelial–mesenchymal transition (EMT) in oral squamous cell carcinoma (OSCC). Expression of E-cadherin and vimentin through immunohistochemical analysis was examined in 200 patients with surgically resected OSCC. Combined E-cadherin and vimentin expression was evaluated to determine the EMT status. Kaplan–Meier curves and log-rank test were used to compare differences in survival. Cox regression analysis was performed to identify independent prognostic factors. E-cadherin expression was negative in 28 (14%) tumors, and vimentin expression was positive in 87 (43.5%) tumors. Moreover, 99 (49.5%), 87 (43.5%), and 14 (7.5%) tumors exhibited no, partial, and complete EMT, respectively. Both individual protein expression were significant prognostic factors [Negative E-cadherin, hazard ratio (HR) = 1.74, 95% confidence interval (CI) = 1.04–2.93; positive vimentin, HR = 1.64, 95% CI = 1.12–2.41]. For EMT status, the HR increased with EMT progression [partial EMT, HR = 1.64, 95% CI = 1.09–2.49; complete EMT, HR = 2.88, 95% CI = 1.44–5.79], of which, the complete EMT had higher HR than was individual protein expression. Combined E-cadherin and vimentin expression as a measure of EMT showed a superior prognostic significance compared with individual protein expression. C1 [Wangmo, Chimi] Prince of Songkla University, Faculty of Medicine, Department of Pathology, 90110 Hat Yai, Songkhla, Thailand. [Charoen, Nattinee] Prince of Songkla University, Faculty of Medicine, Department of Pathology, 90110 Hat Yai, Songkhla, Thailand. [Jantharapattana, Kitti] Prince of Songkla University, Faculty of Medicine, Department of Otolaryngology Head and Neck Surgery, 90110 Hat Yai, Songkhla, Thailand. [Dechaphunkul, Arunee] Prince of Songkla University, Faculty of Medicine, Division of Medical Oncology, Department of Internal Medicine, 90110 Hat Yai, Songkhla, Thailand. [Thongsuksai, Paramee] Prince of Songkla University, Faculty of Medicine, Department of Pathology, 90110 Hat Yai, Songkhla, Thailand. RP Thongsuksai, P (reprint author), Prince of Songkla University, Faculty of Medicine, Department of Pathology, 90110 Hat Yai, Thailand. EM tparamee@gmail.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68:394–424 Virani S, Bilheem S, Chansaard W, Chitapanarux I, Daoprasert K, Khuanchana S, Leklob A, Pongnikorn D, Rozek LS, Siriarechakul S, Suwanrungruang K, Tassanasunthornwong S, Vatanasapt P, Sriplung H, 2017, National and subnational population-based incidence of cancer in Thailand: assessing cancers with the highest burdens. Cancers 9:108 Pruegsanusak K, Peeravut S, Leelamanit V, Sinkijcharoenchai W, Jongsatitpaiboon J, Phungrassami T, Chuchart K, Thongsuksai P, Pruegsanusak K, Peeravut S, Leelamanit V et al, 2012, Survival and prognostic factors of different sites of head and neck cancer: an analysis from Thailand. Asian Pac J Cancer Prev 13:885–850 Chen TC, HsuCW, Lou PJ, Ko JY, Yang TL, Chen CN, Chang YL, Wang CP, 2013, The clinical predictive factors for subsequent distant metastasis in patients with locoregionally advanced oral squamous cell carcinoma. Oral Oncol 49:367–373 Thiery JP, Sleeman JP, 2006, Complex networks orchestrate epithelial–mesenchymal transitions. Nat Rev Mol Cell Biol 7: 131–142 Scanlon CS, Van Tubergen EA, Inglehart RC, D’Silva NJ, 2013, Biomarkers of epithelial-mesenchymal transition in squamous cell carcinoma. J Dent Res 92:114–121 Lamouille S, Xu J, Derynck R, 2014, Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 15:178–196 Sato R, Semba T, Saya H, Arima Y, 2016, Concise review: stem cells and epithelial-mesenchymal transition in cancer: biological implications and therapeutic targets. Stem Cells 34:1997–2007 Li QQ, Xu JD,WangWJ, Cao XX, Chen Q, Tang F, Chen ZQ, Liu XP, Xu ZD, 2009, Twist1-mediated adriamycin-induced epithelialmesenchymal transition relates to multidrug resistance and invasive potential in breast cancer cells. Clin Cancer Res 15:2657–2665 Liu LK, Jiang XY, Zhou XX, Wang DM, Song XL, Jiang HB, 2010, Upregulation of vimentin and aberrant expression of Ecadherin/ beta-catenin complex in oral squamous cell carcinomas: correlation with the clinicopathological features and patient outcome. Mod Pathol 23:213–224 Fan CC, Wang TY, Cheng YA, Jiang SS, Cheng CW, Lee AY, Kao TY, 2013, Expression of E-cadherin, twist, and p53 and their prognostic value in patients with oral squamous cell carcinoma. J Cancer Res Clin Oncol 139:1735–1744 Sawant SS, Vaidya MM, Chaukar DA Alam H, Dmello C, Gangadaran P, Kannan S, Kane S, Dange PP, Dey N, Ranganathan K, D'Cruz AK, 2014, Clinical significance of aberrant vimentin expression in oral premalignant lesions and carcinomas. Oral Dis 20:453–465 Liu PF, Kang BH,Wu YM, Sun JH, Yen LM, Fu TY, Lin YC, Liou HH, Lin YS, Sie HC, Hsieh IC, TsengYK, ShuCW, HsiehYD,Ger LP, 2017, Vimentin is a potential prognostic factor for tongue squamous cell carcinoma among five epithelial-mesenchymal transition- related proteins. PLoS One 12:e0178581 Pyo SW, Hashimoto M, Kim YS, Kim CH, Lee SH, Johnson KR, Wheelock MJ, Park JU, 2007, Expression of E-cadherin, Pcadherin and N-cadherin in oral squamous cell carcinoma: correlation with the clinicopathologic features and patient outcome. J Craniomaxillofac Surg 35:1–9 Wolf GT, Winter W, Bellile E, Nguyen A, Donnelly CR, McHugh JB, Thomas D, Amlani L, Rozek L, Lei YL, Head and Neck SPORE Program, 2018, Histologic pattern of invasion and epithelial-mesenchymal phenotype predict prognosis in squamous carcinoma of the head and neck. Oral Oncol 87:29–35 Aruga N, Kijima H, Masuda R, Onozawa H, Yoshizawa T, Tanaka M, Inokuchi S, Iwazaki M, 2018, Epithelial-mesenchymal transition, EMT, is correlated with patient's prognosis of lung squamous cell carcinoma. Tokai J Exp Clin Med 43:5–13 da Cunha IW, Souza MJ, da CostaWH, AmancioAM, Fonseca FP, Zequi Sde C, Lopes A, Guimaraes GC, Soares F, 2016, Epithelialmesenchymal transition, EMT, phenotype at invasion front of squamous cell carcinoma of the penis influences oncological outcomes. Urol Oncol 34:433.e19–433.e26 Ang KK,Harris J,Wheeler R,Weber R, Rosenthal DI, Nguyen-Tan PF, Westra WH, Chung CH, Jordan RC, Lu C, Kim H, Axelrod R, Silverman CC, Redmond KP, Gillison ML, 2010, Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 363:24–35 Satelli A, Li S, 2011, Vimentin in cancer and its potential as a molecular target for cancer therapy. Cell Mol Life Sci 68: 3033–3046 Bankfalvi A, Krassort M, Buchwalow IB, Vegh A, Felszeghy E, Piffko J, 2002, Gains and losses of adhesion molecules, CD44, Ecadherin, and beta-catenin, during oral carcinogenesis and tumour progression. J Pathol 198:343–351 Mandal M, Myers JN, Lippman SM, Johnson FM, Williams MD, Rayala S, Ohshiro K, Rosenthal DI, Weber RS, Gallick GE, El- Naggar AK, 2008, Epithelial to mesenchymal transition in head and neck squamous carcinoma: association of Src activation with E-cadherin down-regulation, vimentin expression, and aggressive tumor features. Cancer 112:2088–2100 Zhou J, Tao D, Xu Q, Gao Z, Tang D, 2015, Expression of Ecadherin and vimentin in oral squamous cell carcinoma. Int J Clin Exp Pathol 8:3150–3154 Ukpo OC, Thorstad WL, Zhang Q, Lewis JS Jr, 2012, Lack of association of cadherin expression and histopathologic type, metastasis, or patient outcome in oropharyngeal squamous cell carcinoma: a tissue microarray study. Head Neck Pathol 6:38–47 NijkampMM, Span PN, Hoogsteen IJ, van der Kogel AJ, Kaanders JH, Bussink J, 2011, Expression of E-cadherin and vimentin correlates with metastasis formation in head and neck squamous cell carcinoma patients. Radiother Oncol 99:344–348 LingenMW, XiaoW, Schmitt A, Jiang B, Pickard R, Kreinbrink P, Perez-Ordonez B, Jordan RC, Gillison ML, 2013, Low etiologic fraction for high-risk human papillomavirus in oral cavity squamous cell carcinomas. Oral Oncol 49:1–8 Loeschke S, Ohlmann AK, Brasen JH, Holst R,Warnke PH, 2016, Prognostic value of HMGA2, P16, and HPV in oral squamous cell carcinomas. J Craniomaxillofac Surg 44:1422–1429 Rodriguez-Santamarta T, Rodrigo JP, Garcia-Pedrero JM, Alvarez- Teijeiro S, Angeles Villaronga M, Suarez-Fernandez L, Alvarez- Arguelles ME, Astudillo A, 2016, Prevalence of human papillomavirus in oral squamous cell carcinomas in northern Spain. Eur Arch Otorhinolaryngol 273:4549–4559 Wang F, Zhang H, Xue Y, Wen J, Zhou J, Yang X,Wei J, 2017, A systematic investigation of the association between HPV and the clinicopathological parameters and prognosis of oral and oropharyngeal squamous cell carcinomas. Cancer Med 6:910–917 Khovidhunkit SO, Buajeeb W, Sanguansin S, Poomsawat S, Weerapradist W, 2008, Detection of human papillomavirus in oral squamous cell carcinoma, leukoplakia and lichen planus in Thai patients. Asian Pac J Cancer Prev 9:771–775 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1511 EP 1518 DI 10.1007/s12253-019-00731-z PG 8 ER PT J AU Marangon, GC de Bitencorte, TJ Michita, TR Lunge, RV dos Santos, CD Alvares-da-Silva, RM Simon, D AF Marangon, Guerra Camila de Bitencorte, Teixeira Joice Michita, Tomoya Rafael Lunge, Ricardo Vagner dos Santos, Cruz Deivid Alvares-da-Silva, Reis Mario Simon, Daniel TI Association between MICA rs2596542 Polymorphism with the Risk of Hepatocellular Carcinoma in Chronic Hepatitis C Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HCV; Hepatocellular carcinoma; Liver cirrhosis; MICA gene; rs2596542; HCC ID HCV; Hepatocellular carcinoma; Liver cirrhosis; MICA gene; rs2596542; HCC AB In this study we investigated the impact of rs2596542A/G single nucleotide polymorphism (SNP) in the major histocompatibility complex class I chain-related sequence A (MICA) gene on HCV-induced hepatocellular carcinoma (HCC) susceptibility in a Brazilian population. In total, 252 HCV-infected patients (98 with HCV-induced HCC and 154 non-malignant HCV-induced liver cirrhosis) were enrolled and 98 healthy control subjects (negative anti-HCV). The MICA rs2596542 SNP genotypes were determined by real-time PCR assay. No differences in MICA genotype frequencies between HCV-induced cirrhosis patients and controls were observed. However, genotype frequencies of rs2596542A/G SNP were statistically different between HCV induced HCC patients and controls (p = 0.048), and also between HCC and HCV-induced cirrhosis patients (p = 0.039). The highest frequency of the rs2596542AA genotype was observed in HCC patients (31.6%) when compared with HCV-induced cirrhosis patients (18.8%) and healthy controls (19.4%). Also, rs2596542AA genotype carriers have an increased risk for HCC when compared to HCV-induced cirrhosis status [odds ratio (OR) = 1.99; 95% confidence interval (CI) = 1.06–3.74, p = 0.020)] and healthy individuals (OR = 1.92, 95% CI = 1.00–3.70, p = 0.049). Taken together our study suggest that MICA rs2596542 SNP impacts HCV-induced HCC susceptibility suggesting that genetic variants in MICA are of clinical relevance to hepatocarcinogenesis by impacting host immune response in chronic HCV infection. C1 [Marangon, Guerra Camila] Universidade Luterana do Brasil, Programa de Pos Graduacao em Diagnostico Genetico e MolecularCanoas, RS, Brazil. [de Bitencorte, Teixeira Joice] Universidade Luterana do Brasil, Programa de Pos Graduacao em Diagnostico Genetico e MolecularCanoas, RS, Brazil. [Michita, Tomoya Rafael] Universidade Luterana do Brasil, Programa de Pos Graduacao em Diagnostico Genetico e MolecularCanoas, RS, Brazil. [Lunge, Ricardo Vagner] Universidade Luterana do Brasil, Programa de Pos Graduacao em Diagnostico Genetico e MolecularCanoas, RS, Brazil. [dos Santos, Cruz Deivid] Universidade Federal do Rio Grande do Sul (UFRGS), Hospital de Clinicas de Porto Alegre, Servico de Gastroenterologia, Departamento de Medicina InternaPorto Alegre, RS, Brazil. [Alvares-da-Silva, Reis Mario] Universidade Federal do Rio Grande do Sul (UFRGS), Hospital de Clinicas de Porto Alegre, Servico de Gastroenterologia, Departamento de Medicina InternaPorto Alegre, RS, Brazil. [Simon, Daniel] Universidade Luterana do Brasil, Programa de Pos Graduacao em Diagnostico Genetico e MolecularCanoas, RS, Brazil. RP Simon, D (reprint author), Universidade Luterana do Brasil, Programa de Pos Graduacao em Diagnostico Genetico e Molecular, Canoas, Brazil. EM daniel.simon@ulbra.br CR Tang H, Grise H, 2009, Cellular and molecular biology of HCV infection and hepatitis. Clin Sci, Lond, 117:49–65. , DOI 10.1042/CS20080631 El Khoury AC, Wallace C, Klimack WK, Razavi H, 2012, Economic burden of hepatitis C-associated diseases: Europe, Asia Pacific, and the Americas. JMed Econ 15:887–896. , DOI 10.3111/13696998.2012.681332 Wong RJ, Gish RG, 2016, Metabolic manifestations and complications associated with chronic hepatitis C virus infection. Gastroenterol Hepatol, N Y, 12:293–299 http://www.ncbi.nlm. nih.gov/pubmed/27499712 Lingala S, Ghany MG, 2015, Natural history of hepatitis C. Gastroenterol Clin N Am 44:717–734. , DOI 10.1016/j. gtc.2015.07.003 Sebastiani G, Gkouvatsos K, Pantopoulos K, 2014, Chronic hepatitis C and liver fibrosis. World J Gastroenterol 20:11033–11053. , DOI 10.3748/wjg.v20.i32.11033 Balogh J, Victor D, Asham EH, Burroughs SG, Boktour M, Saharia A, Li X, Ghobrial RM, Monsour HP, 2016, Hepatocellular carcinoma: a review. J Hepatocell Carcinoma 3:41–53. , DOI 10.2147/JHC.S61146 Miki D, Ochi H, Hayes CN, Abe H, Yoshima T, Aikata H, Ikeda K, Kumada H, Toyota J, Morizono T, Tsunoda T, Kubo M, Nakamura Y, Kamatani N, Chayama K, 2011, Variation in the DEPDC5 locus is associated with progression to hepatocellular carcinoma in chronic hepatitis C virus carriers. Nat Genet 43:797–800. , DOI 10.1038/ng.876 Patin E, Kutalik Z, Guergnon J, Bibert S, Nalpas B, Jouanguy E, Munteanu M, Bousquet L, Argiro L, Halfon P, Boland A, Mullhaupt B, Semela D, Dufour J-F, Heim MH, Moradpour D, Cerny A, Malinverni R, Hirsch H, Martinetti G, Suppiah V, Stewart G, Booth DR, George J, Casanova J-L, Brechot C, Rice CM, Talal AH, Jacobson IM, BourliereM, Theodorou I, Poynard T, Negro F, Pol S, Bochud P-Y, Abel L, I.H.C.G.C. Swiss Hepatitis C Cohort Study Group, 2012, French ANRS HC EP 26 Genoscan Study Group, Genome-wide association study identifies variants associated with progression of liver fibrosis from HCV infection. Gastroenterology 143:1244–1252.e12. , DOI 10.1053/j. gastro.2012.07.097 Urabe Y, Ochi H, Kato N, Kumar V, Takahashi A, Muroyama R, Hosono N, Otsuka M, Tateishi R, Lo PHY, Tanikawa C, Omata M, Koike K,Miki D, Abe H, Kamatani N, Toyota J, Kumada H, Kubo M, Chayama K, Nakamura Y, Matsuda K, 2013, A genome-wide association study of HCV-induced liver cirrhosis in the Japanese population identifies novel susceptibility loci at the MHC region. J Hepatol 58:875–882. , DOI 10.1016/j.jhep.2012.12.024 Kumar V, Kato N, Urabe Y, Takahashi A, Muroyama R, Hosono N, Otsuka M, Tateishi R, OmataM, Nakagawa H, Koike K, Kamatani N, Kubo M, Nakamura Y, Matsuda K, 2011, Genome-wide association study identifies a susceptibility locus for HCV-induced hepatocellular carcinoma. Nat Genet 43:455–458. , DOI 10.1038/ng.809 Bahram S, Bresnahan M, Geraghty DE, Spies T, 1994, A second lineage of mammalian major histocompatibility complex class I genes. Proc Natl Acad Sci U S A 91:6259–6263. , DOI 10.1073/pnas.91.14.6259, http://www.ncbi.nlm.nih.gov/pubmed/ 8022771 Bauer S, 1999, Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA, science, 80-. ), vol 285, pp 727–729. , DOI 10.1126/science.285.5428.727 Ghadially H, Brown L, Lloyd C, Lewis L, Lewis A, Dillon J, Sainson R, Jovanovic J, Tigue NJ, Bannister D, Bamber L, Valge- Archer V,Wilkinson RW(2017, MHC class I chain-related protein a and B, MICA and MICB, are predominantly expressed intracellularly in tumour and normal tissue. Br J Cancer 116:1208–1217. , DOI 10.1038/bjc.2017.79 Schrambach S, Ardizzone M, Leymarie V, Sibilia J, Bahram S, 2007, In vivo expression pattern of MICA and MICB and its relevance to auto-immunity and cancer. PLoS One 2:e518. https:// doi.org/10.1371/journal.pone.0000518 Chen D, Gyllensten U, 2014, MICA polymorphism: biology and importance in cancer. Carcinogenesis. 35:2633–2642. https://doi. org/10.1093/carcin/bgu215 Zhang J, Basher F,Wu JD, 2015, NKG2D ligands in tumor immunity: two sides of a coin. Front Immunol 6:97. , DOI 10. 3389/fimmu.2015.00097 Baranwal AK, Mehra NK, 2017, Major histocompatibility complex class I chain-related a, MICA, molecules: relevance in solid organ transplantation. Front Immunol 8:182. , DOI 10. 3389/fimmu.2017.00182 KumarV, Yi Lo PH, Sawai H, Kato N, Takahashi A, Deng Z, Urabe Y, Mbarek H, Tokunaga K, Tanaka Y, Sugiyama M, Mizokami M, Muroyama R, Tateishi R, Omata M, Koike K, Tanikawa C, Kamatani N, Kubo M, Nakamura Y, Matsuda K, 2012, Soluble MICA and a MICA variation as possible prognostic biomarkers for HBV-induced hepatocellular carcinoma. PLoS One 7:e44743. , DOI 10.1371/journal.pone.0044743 Wang L-P, Niu H, Xia Y-F, Han Y-L, Niu P, Wang H-Y, Zhou Q-L, 2015, Prognostic significance of serum sMICA levels in non-small cell lung cancer. Eur Rev Med Pharmacol Sci 19:2226–2230 http:// www.ncbi.nlm.nih.gov/pubmed/26166647 Zhao Y-K, Jia C-M, Yuan G-J, Liu W, Qiu Y, Zhu Q-G, 2015, Expression and clinical value of the soluble major histocompatibility complex class I-related chain a molecule in the serum of patients with renal tumors. Genet Mol Res 14:7233–7240. , DOI 10.4238/2015.June.29.16 Li H, Liu F, Zhu H, Zhou X, Lu J, Chang H, Hu J, 2016, Interaction between polymorphisms of IFN-γ and MICA correlated with hepatocellular carcinoma. Med SciMonit 22:549–553. , DOI 10.12659/MSM.895101 Motomura T, Ono Y, Shirabe K, Fukuhara T, Konishi H, Mano Y, Toshima T, Yoshiya S,Muto J, Ikegami T, Yoshizumi T, Maehara Y, 2012, Neither MICA Nor DEPDC5 Genetic Polymorphisms Correlate with Hepatocellular Carcinoma Recurrence following Hepatectomy. HPB Surg 2012:185496. , DOI 10.1155/ 2012/185496 Lange CM, Bibert S, Dufour J-F, Cellerai C, Cerny A, Heim MH, Kaiser L, Malinverni R, Mullhaupt B, Negro F, Semela D, Moradpour D, Kutalik Z, Bochud P-Y, 2013, Swiss hepatitis C cohort study group, comparative genetic analyses point to HCP5 as susceptibility locus forHCV-associated hepatocellular carcinoma. J Hepatol 59: 504–509. , DOI 10.1016/j.jhep.2013.04.032 BurzaMA, MottaBM, MancinaRM, Pingitore P, Pirazzi C, Lepore SM, Spagnuolo R, Doldo P, Russo C, Lazzaro V, Fischer J, Berg T, Aghemo A, Cheroni C, De Francesco R, Fargion S, Colombo M, Datz C, Stickel F, Valenti L, Romeo S, 2016, DEPDC5 variants increase fibrosis progression in Europeans with chronic hepatitis C virus infection. Hepatology. 63:418–427. , DOI 10.1002/ hep.28322 Mohamed AA, Elsaid OM, Amer EA, Elosaily HH, Sleem MI, Gerges SS, Saleh MA, El Shimy A, El Abd YS, 2017, Clinical significance of SNP, rs2596542, in histocompatibility complex class I-related gene a promoter region among hepatitis C virus related hepatocellular carcinoma cases. J Adv Res 8:343–349. https:// doi.org/10.1016/j.jare.2017.03.004 Bedossa P, Poynard T, 1996, An algorithm for the grading of activity in chronic hepatitis C. the METAVIR cooperative study group. Hepatology. 24:289–293. , DOI 10.1002/hep. 510240201 Alameri HF, Sanai FM, Al Dukhayil M, Azzam NA, Al-Swat KA, Hersi AS, Abdo AA, 2007, Six minute walk test to assess functional capacity in chronic liver disease patients. World J Gastroenterol 13:3996–4001. , DOI 10.3748/wjg.v13.i29.3996 Bruix J, Sherman M, 2011, American Association for the Study of Liver Diseases, management of hepatocellular carcinoma: an update. Hepatology. 53:1020–1022. , DOI 10.1002/hep.24199 Lahiri DK, Nurnberger JI, 1991, Arapid non-enzymaticmethod for the preparation of HMW DNA from blood for RFLP studies. Nucleic Acids Res 19:5444 http://www.ncbi.nlm.nih.gov/pubmed/ 1681511 Rodriguez S, Gaunt TR, Day INM, 2009, Hardy-Weinberg equilibrium testing of biological ascertainment for Mendelian randomization studies. Am J Epidemiol 169:505–514. , DOI 10. 1093/aje/kwn359 Vescovo T, Refolo G,Vitagliano G, FimiaGM, PiacentiniM(2016, Molecular mechanisms of hepatitis C virus-induced hepatocellular carcinoma. Clin Microbiol Infect 22:853–861. , DOI 10. 1016/j.cmi.2016.07.019 Lo PHY, Urabe Y, Kumar V, Tanikawa C, Koike K, Kato N, Miki D, Chayama K, Kubo M, Nakamura Y, Matsuda K, 2013, Identification of a functional variant in the MICA promoter which regulates MICA expression and increases HCV-related hepatocellular carcinoma risk. PLoS One 8:e61279. , DOI 10.1371/ journal.pone.0061279 Michita RT, Chies JAB, Schramm S, Horn PA, Heinemann FM, Wunsch A, Viebahn R, Schenker P, Rebmann V, 2018, A valine mismatch at position 129 of MICA is an independent predictor of Cytomegalovirus infection and acute kidney rejection in simultaneous pancreas−kidney transplantation recipients. Int J Mol Sci 19. , DOI 10.3390/ijms19092618 Machiela MJ, Chanock SJ, 2018, LDassoc: an online tool for interactively exploring genome-wide association study results and prioritizing variants for functional investigation. Bioinformatics. 34:887–889. , DOI 10.1093/bioinformatics/btx561 Huang C-F, Huang C-Y, Yeh M-L,Wang S-C, Chen K-Y, Ko Y-M, Lin C-C, Tsai Y-S, Tsai P-C, Lin Z-Y, Chen S-C, Dai C-Y, Huang JF, ChuangW-L, Yu M-L, 2017, Genetics variants and serum levels of MHC class I chain-related a in predicting hepatocellular carcinoma development in chronic hepatitis C patients post antiviral treatment. EBioMedicine. 15:81–89. , DOI 10.1016/j.ebiom. 2016.11.031 Huang C-F, Huang C-I, Yeh M-L,Wang S-C, Chen K-Y, Ko Y-M, Lin C-C, Tsai Y-S, Tsai P-C, Lin Z-Y, Chen S-C, Dai C-Y, Huang J-F, Chuang W-L, Yu M-L, 2017, Diversity of the association of serum levels and genetic variants of MHC class I polypeptide-related chain a with liver fibrosis in chronic hepatitis C. Oncotarget. 8:32618–32625. , DOI 10.18632/oncotarget.15941 Isernhagen A,Malzahn D, Bickeboller H, Dressel R, 2016, Impact of the MICA-129Met/Val dimorphism on NKG2D-mediated biological functions and disease risks. Front Immunol 7:588. https:// doi.org/10.3389/fimmu.2016.00588 Isernhagen A, Malzahn D, Viktorova E, Elsner L, Monecke S, von Bonin F, Kilisch M, Wermuth JM, Walther N, Balavarca Y, Stahl- Hennig C, Engelke M, Walter L, Bickeboller H, Kube D, Wulf G, Dressel R, 2015, The MICA-129 dimorphism affects NKG2D signaling and outcome of hematopoietic stem cell transplantation. EMBO Mol Med 7:1480–1502. , DOI 10.15252/emmm. 201505246 Vallian S, Rad MJ, Tavallaei M, Tavassoli M, 2012, Correlation of major histocompatibility complex class I related a, MICA, polymorphism with the risk of developing breast cancer.Med Oncol 29: 5–9. , DOI 10.1007/s12032-010-9776-9 Chen D, Hammer J, Lindquist D, Idahl A, Gyllensten U, 2014, A variant upstream of HLA-DRB1 and multiple variants in MICA influence susceptibility to cervical cancer in a Swedish population. Cancer Med 3:190–198. , DOI 10.1002/cam4.183 Tong HV, Toan NL, Song LH, Bock C-T, Kremsner PG, Velavan TP, 2013, Hepatitis B virus-induced hepatocellular carcinoma: functional roles of MICA variants. J Viral Hepat 20:687–698. , DOI 10.1111/jvh.12089 Chalupny NJ, Rein-Weston A, Dosch S, Cosman D, 2006, Downregulation of the NKG2D ligand MICA by the human cytomegalovirus glycoprotein UL142. Biochem Biophys Res Commun 346: 175–181. , DOI 10.1016/j.bbrc.2006.05.092 Ashiru O, Bennett NJ, Boyle LH, Thomas M, Trowsdale J, Wills MR, 2009, NKG2D ligand MICA is retained in the cis-Golgi apparatus by human cytomegalovirus protein UL142. J Virol 83: 12345–12354. , DOI 10.1128/JVI.01175-09 Moenkemeyer M, Heiken H, Schmidt RE, Witte T, 2009, Higher risk of cytomegalovirus reactivation in human immunodeficiency virus-1-infected patients homozygous forMICA5.1. HumImmunol 70:175–178. , DOI 10.1016/j.humimm.2009.01.005 Goto K, Kato N, 2015, MICA SNPs and the NKG2D system in virus-induced HCC. J Gastroenterol 50:261–272. , DOI 10.1007/s00535-014-1000-9 Goto K, Annan DA,Morita T, LiW,Muroyama R, MatsubaraY, Ito S, Nakagawa R, Tanoue Y, Jinushi M, Kato N, 2016, Novel chemoimmunotherapeutic strategy for hepatocellular carcinoma based on a genome-wide association study. Sci Rep 6:38407. , DOI 10.1038/srep38407 Poggi A, Catellani S, Garuti A, Pierri I, Gobbi M, Zocchi MR, 2009, Effective in vivo induction of NKG2D ligands in acute myeloid leukaemias by all-trans-retinoic acid or sodium valproate. Leukemia. 23:641–648. , DOI 10.1038/leu.2008.354 Kuper H, Tzonou A, Kaklamani E, Hsieh C-C, Lagiou P, Adami HO, Trichopoulos D, Stuver SO, 2000, Tobacco smoking, alcohol consumption and their interaction in the causation of hepatocellular carcinoma. Int J Cancer 85:498–502. , DOI 10.1002/(SICI, 1097-0215(20000215)85:4<498::AID-IJC9>3.0.CO;2-F Steinle A, Li P,Morris DL, Groh V, Lanier LL, Strong RK, Spies T, 2001, Interactions of human NKG2D with its ligands MICA, MICB, and homologs of the mouse RAE-1 protein family. Immunogenetics. 53:279–287. , DOI 10.1007/ s002510100325 Hayashi T, Imai K,Morishita Y, Hayashi I, Kusunoki Y, Nakachi K, 2006, Identification of the NKG2D haplotypes associated with natural cytotoxic activity of peripheral blood lymphocytes and cancer immunosurveillance. Cancer Res 66:563–570. , DOI 10.1158/0008-5472.CAN-05-2776 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1519 EP 1525 DI 10.1007/s12253-019-00738-6 PG 7 ER PT J AU Gregova, M Hojny, J Nemejcova, K Bartu, M Mara, M Boudova, B Laco, J Krbal, L Ticha, I Dundr, P AF Gregova, Maria Hojny, Jan Nemejcova, Kristyna Bartu, Michaela Mara, Michal Boudova, Barbora Laco, Jan Krbal, Lukas Ticha, Ivana Dundr, Pavel TI Leiomyoma with Bizarre Nuclei: a Study of 108 Cases Focusing on Clinicopathological Features, Morphology, and Fumarate Hydratase Alterations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Leiomyoma with bizarre nuclei; Usual leiomyoma; Leiomyosarcoma; Fumarate hydratase ID Leiomyoma with bizarre nuclei; Usual leiomyoma; Leiomyosarcoma; Fumarate hydratase AB Leiomyoma with bizarre nuclei (LBN) is an uncommon variant of uterine smooth muscle neoplasm. Involvement of fumarate hydratase (FH) has been suggested in the pathogenesis of a subset of LBN. The goal of our study is to assess the clinicopathological, morphological, immunohistochemical and molecular findings focusing on FH in LBNs (n = 108) and compare it with the findings in usual leiomyomas (UL; n = 50) and leiomyosarcomas (LMS; n = 42). Immunohistochemically, loss of FH expression was found in 67/108 of LBN, 1/50 of UL and in no LMS. Class 4/5 FH mutations were detected in 15/53 LBN with sufficient DNA quality for molecular analysis. Pathogenic variants of the FH gene were detected in neither UL nor LMS. Local recurrence after surgery was present in 18/92 of LBN patients, 7 of which were histologically verified and 2 of which were found to be LBN. Our results confirmed that LBN behave in a benign fashion, although they may relapse. FH gene mutations were a common finding only in LBN, but not in UL and LMS. Immunohistochemistry with an antibody against FH seems to have a good sensitivity (87%) and moderate specificity (58%) with regard to predicting FH gene mutations and could be used as a screening method in tumors with features suggestive of FH alterations to identify patients who are at risk for the FH aberrations. C1 [Gregova, Maria] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Hojny, Jan] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Nemejcova, Kristyna] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Bartu, Michaela] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Mara, Michal] Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Obstetrics and GynecologyPrague, Czech Republic. [Boudova, Barbora] Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Obstetrics and GynecologyPrague, Czech Republic. [Laco, Jan] Charles University, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, The Fingerland Department of PathologyPrague, Czech Republic. [Krbal, Lukas] Charles University, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, The Fingerland Department of PathologyPrague, Czech Republic. [Ticha, Ivana] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Dundr, Pavel] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. RP Dundr, P (reprint author), Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, 12800 Prague, Czech Republic. EM pavel.dundr@vfn.cz CR Bell SW, Kempson RL, Hendrickson MR, 1994, Problematic uterine smooth muscle neoplasms. A clinicopathologic study of 213 cases. Am J Surg Pathol 18(6):535–558 Kurman RJ, Carcangiu ML, Young RH, Herrington CSE, 2014, WHO Classification of tumours of female reproductive organs, 4th edn. IARC, Lyon:307 Liegl-Atzwanger B, Heitzer E, Flicker K, Muller S, Ulz P, Saglam O, Tavassoli F, Devouassoux-Shisheboran M, Geigl J, Moinfar F, 2016, Exploring chromosomal abnormalities and genetic changes in uterine smooth muscle tumors. Mod Pathol 29(10):1262–1277. , DOI 10.1038/modpathol.2016.107 Makinen N, Kampjarvi K, Frizzell N, Butzow R, Vahteristo P, 2017, Characterization of MED12, HMGA2, and FH alterations reveals molecular variability in uterine smooth muscle tumors. Mol Cancer 16(1):101. , DOI 10.1186/s12943-017-0672-1 Ubago JM, Zhang Q, Kim JJ, Kong B,Wei JJ, 2016, Two subtypes of atypical Leiomyoma: clinical, histologic, and molecular analysis. Am J Surg Pathol 40(7):923–933. , DOI 10.1097/PAS. 0000000000000646 Nibert M, Heim S, 1990, Uterine leiomyoma cytogenetics. Genes Chromosomes Cancer 2(1):3–13 Lehtonen R, Kiuru M, Vanharanta S, Sjoberg J, Aaltonen LM, Aittomaki K, Arola J, Butzow R, Eng C, Husgafvel-Pursiainen K, Isola J, Jarvinen H, Koivisto P, Mecklin JP, Peltomaki P, Salovaara R,Wasenius VM, Karhu A, Launonen V, Nupponen NN, Aaltonen LA, 2004, Biallelic inactivation of fumarate hydratase, FH, occurs in nonsyndromic uterine leiomyomas but is rare in other tumors. Am J Pathol 164(1):17–22. , DOI 10.1016/S0002- 9440(10)63091-X Harrison WJ, Andrici J, Maclean F, Madadi-Ghahan R, Farzin M, Sioson L, Toon CW, Clarkson A, Watson N, Pickett J, Field M, Crook A, Tucker K, Goodwin A, Anderson L, Srinivasan B, Grossmann P, Martinek P, Ondic O, Hes O, Trpkov K, Clifton- Bligh RJ, Dwight T, Gill AJ, 2016, Fumarate hydratase-deficient uterine leiomyomas occur in both the syndromic and sporadic settings. Am J Surg Pathol 40(5):599–607. , DOI 10.1097/ PAS.0000000000000573 Ylisaukko-oja SK, Kiuru M, Lehtonen HJ, Lehtonen R, Pukkala E, Arola J, Launonen V, Aaltonen LA, 2006, Analysis of fumarate hydratase mutations in a population-based series of early onset uterine leiomyosarcoma patients. Int J Cancer 119(2):283–287. , DOI 10.1002/ijc.21798 Barker KT, Spendlove HE, Banu NS, Bridge JA, Fisher C, Shipley J, Garrett M, Manyonda I, Houlston RS, 2006, No evidence for epigenetic inactivation of fumarate hydratase in leiomyomas and leiomyosarcomas. Cancer Lett 235(1):136–140. , DOI 10. 1016/j.canlet.2005.04.017 Alam NA, Olpin S, Rowan A, Kelsell D, Leigh IM, Tomlinson IP, Weaver T, 2005, Missense mutations in fumarate hydratase in multiple cutaneous and uterine leiomyomatosis and renal cell cancer. J Mol Diagn 7(4):437–443. , DOI 10.1016/S1525-1578(10, 60574-0 Llamas-Velasco M, Requena L, Adam J, Frizzell N, Hartmann A, Mentzel T, 2016, Loss of fumarate hydratase and aberrant protein succination detected with S-(2-Succino)-cysteine staining to identify patients with Multiple cutaneous and uterine Leiomyomatosis and hereditary Leiomyomatosis and renal cell cancer syndrome. Am J Dermatopathol 38(12):887–891. , DOI 10.1097/ DAD.0000000000000580 Wei JJ, 2016, Atypical leiomyoma with features suggesting of fumarate hydratase mutation. Int J Gynecol Pathol 35(6):531–536. , DOI 10.1097/PGP.0000000000000276 Linehan WM, Rouault TA, 2013, Molecular pathways: fumarate hydratase-deficient kidney cancer–targeting the Warburg effect in cancer. Clin Cancer Res 19(13):3345–3352. , DOI 10. 1158/1078-0432.CCR-13-0304 Tong WH, Sourbier C, Kovtunovych G, Jeong SY, Vira M, Ghosh M, Romero VV, Sougrat R, Vaulont S, Viollet B, Kim YS, Lee S, Trepel J, Srinivasan R, Bratslavsky G, Yang Y, Linehan WM, Rouault TA, 2011, The glycolytic shift in fumarate-hydratasedeficient kidney cancer lowers AMPK levels, increases anabolic propensities and lowers cellular iron levels. Cancer Cell 20(3): 315–327. , DOI 10.1016/j.ccr.2011.07.018 Tomlinson IP, Alam NA, Rowan AJ, Barclay E, Jaeger EE, Kelsell D, Leigh I, Gorman P, Lamlum H, Rahman S, Roylance RR, Olpin S, Bevan S, Barker K, Hearle N, Houlston RS, Kiuru M, Lehtonen R, Karhu A, Vilkki S, Laiho P, Eklund C, Vierimaa O, Aittomaki K, Hietala M, Sistonen P, Paetau A, Salovaara R, Herva R, Launonen V, Aaltonen LA, Multiple Leiomyoma C, 2002, Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat Genet 30(4):406– 410. , DOI 10.1038/ng849 Trpkov K, Hes O, Agaimy A, Bonert M, Martinek P, Magi-Galluzzi C, Kristiansen G, Luders C, Nesi G, Comperat E, Sibony M, Berney DM, Mehra R, Brimo F, Hartmann A, Husain A, Frizzell N, Hills K, Maclean F, Srinivasan B, Gill AJ, 2016, Fumarate hydratase-deficient renal cell carcinoma is strongly correlated with fumarate hydratase mutation and hereditary leiomyomatosis and renal cell carcinoma syndrome. Am J Surg Pathol 40(7):865–875. , DOI 10.1097/PAS.0000000000000617 Buelow B, Cohen J, Nagymanyoki Z, Frizzell N, Joseph NM, McCalmont T, Garg K, 2016, Immunohistochemistry for 2- Succinocysteine, 2SC, and Fumarate Hydratase, FH, in cutaneous leiomyomas may aid in identification of patients with HLRCC, Hereditary Leiomyomatosis and Renal Cell Carcinoma Syndrome). Am J Surg Pathol 40(7):982–988. , DOI 10. 1097/PAS.0000000000000626 Wei MH, Toure O, Glenn GM, Pithukpakorn M, Neckers L, Stolle C, Choyke P, Grubb R, Middelton L, Turner ML, Walther MM, Merino MJ, Zbar B, LinehanWM, Toro JR, 2006, Novelmutations in FH and expansion of the spectrum of phenotypes expressed in families with hereditary leiomyomatosis and renal cell cancer. J Med Genet 43(1):18–27. , DOI 10.1136/jmg.2005.033506 McLaren W, Gil L, Hunt SE, Riat HS, Ritchie GR, Thormann A, Flicek P, Cunningham F, 2016, The Ensembl variant effect predictor. Genome Biol 17(1):122. , DOI 10.1186/s13059-016- 0974-4 Bennett JA,Weigelt B, Chiang S, Selenica P, ChenYB, Bialik A, Bi R, Schultheis AM, Lim RS, Ng CKY, Morales-Oyarvide V, Young RH, Reuter VE, Soslow RA, Oliva E, 2017, Leiomyoma with bizarre nuclei: a morphological, immunohistochemical and molecular analysis of 31 cases. Mod Pathol 30(10):1476–1488. https:// doi.org/10.1038/modpathol.2017.56 Croce S, Young RH, Oliva E, 2014, Uterine leiomyomas with bizarre nuclei: a clinicopathologic study of 59 cases. Am J Surg Pathol 38(10):1330–1339. , DOI 10.1097/PAS. 0000000000000249 Dastranj TabriziA, Ghojazadeh M, Thagizadeh Anvar H,Vahedi A, Naji S, Mostafidi E, Berenjian S, 2015, Immunohistochemical profile of uterine leiomyoma with bizarre nuclei; comparison with conventional leiomyoma, smooth muscle tumors of uncertain malignant potential and leiomyosarcoma. Adv Pharm Bull 5(Suppl 1): 683–687. , DOI 10.15171/apb.2015.093 Kefeli M, Caliskan S, Kurtoglu E, Yildiz L, Kokcu A, 2017, Leiomyoma with bizarre nuclei: clinical and pathologic features of 30 patients. Int J Gynecol Pathol 37:379–387. , DOI 10.1097/PGP.0000000000000425 Liang Y, Zhang X, Chen X, Lu W, 2015, Diagnostic value of progesterone receptor, p16, p53 and pHH3 expression in uterine atypical leiomyoma. Int J Clin Exp Pathol 8(6):7196–7202 Ly A, Mills AM, McKenney JK, Balzer BL, Kempson RL, Hendrickson MR, Longacre TA, 2013, Atypical leiomyomas of the uterus: a clinicopathologic study of 51 cases. Am J Surg Pathol 37(5):643–649. , DOI 10.1097/PAS. 0b013e3182893f36 Sung CO, Ahn G, Song SY, Choi YL, Bae DS, 2009, Atypical leiomyomas of the uterus with long-term follow-up after myomectomy with immunohistochemical analysis for p16INK4A, p53, Ki- 67, estrogen receptors, and progesterone receptors. Int J Gynecol Pathol 28(6):529–534. , DOI 10.1097/PGP. 0b013e3181a2b8d3 Zhang Q, Poropatich K, Ubago J, Xie J, Xu X, Frizzell N, Kim J, Kong B, Wei JJ, 2017, Fumarate hydratase mutations and alterations in Leiomyoma with bizarre nuclei. Int J Gynecol Pathol 37: 421–430. , DOI 10.1097/PGP.0000000000000447 Kalogiannidis I, Stavrakis T, Dagklis T, Petousis S, Nikolaidou C, Venizelos I, Rousso D, 2016, A clinicopathological study of atypical leiomyomas: benign variant leiomyoma or smooth-muscle tumor of uncertain malignant potential. Oncol Lett 11(2):1425–1428. , DOI 10.3892/ol.2015.4062 Mills AM, Ly A, Balzer BL, Hendrickson MR, Kempson RL, McKenney JK, Longacre TA, 2013, Cell cycle regulatory markers in uterine atypical leiomyoma and leiomyosarcoma: immunohistochemical study of 68 cases with clinical follow-up. Am J Surg Pathol 37(5):634–642. , DOI 10.1097/PAS. 0b013e318287779c Pang SJ, Li CC, Shen Y, Liu YZ, Shi YQ, Liu YX, 2015, Value of counting positive PHH3 cells in the diagnosis of uterine smooth muscle tumors. Int J Clin Exp Pathol 8(5):4418–4426 Dundr P, Povysil C, Tvrdik D,MaraM(2007, Uterine leiomyomas with inclusion bodies: an immunohistochemical and ultrastructural analysis of 12 cases. Pathol Res Pract 203(3):145–151. https://doi. org/10.1016/j.prp.2006.12.008 Makinen N, Vahteristo P, Kampjarvi K, Arola J, Butzow R, Aaltonen LA, 2013)MED12 exon 2 mutations in histopathological uterine leiomyoma variants. Eur J Hum Genet 21(11):1300–1303. , DOI 10.1038/ejhg.2013.33 Makinen N, Aavikko M, Heikkinen T, Taipale M, Taipale J, Koivisto-Korander R, Butzow R, Vahteristo P, 2016, Exome sequencing of uterine leiomyosarcomas identifies frequent mutations in TP53,ATRX, andMED12. PLoS Genet 12(2):e1005850. https:// doi.org/10.1371/journal.pgen.1005850 Reyes C, Karamurzin Y, Frizzell N, Garg K, Nonaka D, Chen YB, Soslow RA, 2014, Uterine smooth muscle tumors with features suggesting fumarate hydratase aberration: detailed morphologic analysis and correlation with S-(2-succino)-cysteine immunohistochemistry. Mod Pathol 27(7):1020–1027. , DOI 10.1038/ modpathol.2013.215 Sanz-Ortega J, Vocke C, Stratton P, Linehan WM, Merino MJ, 2013, Morphologic and molecular characteristics of uterine leiomyomas in hereditary leiomyomatosis and renal cancer, HLRCC, syndrome. Am J Surg Pathol 37(1):74–80. https://doi. org/10.1097/PAS.0b013e31825ec16f Joseph NM, Solomon DA, Frizzell N, Rabban JT, Zaloudek C, Garg K, 2015, Morphology and immunohistochemistry for 2SC and FH aid in detection of fumarate hydratase gene aberrations in uterine leiomyomas from young patients. Am J Surg Pathol 39(11): 1529–1539. , DOI 10.1097/PAS.0000000000000520 Miettinen M, Felisiak-Golabek A, Wasag B, Chmara M, Wang Z, Butzow R, Lasota J, 2016, Fumarase-deficient uterine leiomyomas: an immunohistochemical, molecular genetic, and clinicopathologic study of 86 cases. Am J Surg Pathol 40(12):1661–1669. https://doi. org/10.1097/PAS.0000000000000703 Picaud S, Kavanagh KL, Yue WW, Lee WH, Muller-Knapp S, Gileadi O, Sacchettini J, Oppermann U, 2011, Structural basis of fumarate hydratase deficiency. J Inherit Metab Dis 34(3):671–676. , DOI 10.1007/s10545-011-9294-8 Chuang GS, Martinez-Mir A, Geyer A, Engler DE, Glaser B, Cserhalmi-Friedman PB, Gordon D, Horev L, Lukash B, Herman E, Cid MP, Brenner S, Landau M, Sprecher E, Garcia Muret MP, ChristianoAM, Zlotogorski A, 2005, Germline fumarate hydratase mutations and evidence for a founder mutation underlying multiple cutaneous and uterine leiomyomata. J Am Acad Dermatol 52(3 Pt 1):410–416. , DOI 10.1016/j.jaad.2004.08.051 Siegler L, Erber R, Burghaus S, Brodkorb T,Wachter D,Wilkinson N, Bolton J, StringfellowH,Haller F, BeckmannMW, Hartmann A, Agaimy A, 2018, Fumarate hydratase, FH, deficiency in uterine leiomyomas: recognition by histological features versus blind immunoscreening. Virchows Arch 472(5):789–796. https://doi. org/10.1007/s00428-018-2292-6 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1527 EP 1537 DI 10.1007/s12253-019-00739-5 PG 11 ER PT J AU Peters, AMM Meijer, C Fehrmann, SNR Walenkamp, MEA Kema, PI de Vries, GEE Hollema, H Oosting, FS AF Peters, A M Marloes Meijer, Coby Fehrmann, S N Rudolf Walenkamp, M E Annemiek Kema, P Ido de Vries, G E Elisabeth Hollema, Harry Oosting, F Sjoukje TI Serotonin and Dopamine Receptor Expression in Solid Tumours Including Rare Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Serotonin receptor 1B; Serotonin receptor 2B; Dopamine receptor D2; Dopamine receptor D1; Neovascularization; Neoplasms ID Serotonin receptor 1B; Serotonin receptor 2B; Dopamine receptor D2; Dopamine receptor D1; Neovascularization; Neoplasms AB In preclinical studies serotonin stimulates and dopamine inhibits tumour growth and angiogenesis. Information regarding serotonin and dopamine receptor (5-HTR and DRD) expression in human cancers is limited. Therefore, we screened a large tumour set for receptor mRNA overexpression using functional genomic mRNA (FGmRNA) profiling, and we analysed protein expression and location of 5-HTR1B, 5-HTR2B, DRD1, and DRD2 with immunohistochemistry in different tumour types. With FGmRNA profiling 11,756 samples representing 43 tumour types were compared to 3,520 normal tissue samples to analyse receptor overexpression. 5-HTR2B overexpression was present in many tumour types, most frequently in uveal melanomas (56%). Receptor overexpression in rare cancers included 5-HTR1B in nasopharyngeal carcinoma (17%), DRD1 in ependymoma (30%) and synovial sarcoma (21%), and DRD2 in astrocytoma (13%). Immunohistochemistry demonstrated high 5-HTR2B protein expression on melanoma and gastro-intestinal stromal tumour cells and endothelial cells of colon, ovarian, breast, renal and pancreatic tumours. 5-HTR1B expression was predominantly low. High DRD2 protein expression on tumour cells was observed in 48% of pheochromocytomas, and DRD1 expression ranged from 14% in melanoma to 57% in renal cell carcinoma. In conclusion, 5-HTR1B, 5-HTR2B, DRD1, and DRD2 show mRNA overexpression in a broad spectrum of common and rare cancers. 5-HTR2B protein is frequently highly expressed in human cancers, especially on endothelial cells. These findings support further investigation of especially 5HTR2B as a potential treatment target. C1 [Peters, A M Marloes] University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Hanzeplein 1, 9700 Groningen, RB, The Netherlands. [Meijer, Coby] University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Hanzeplein 1, 9700 Groningen, RB, The Netherlands. [Fehrmann, S N Rudolf] University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Hanzeplein 1, 9700 Groningen, RB, The Netherlands. [Walenkamp, M E Annemiek] University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Hanzeplein 1, 9700 Groningen, RB, The Netherlands. [Kema, P Ido] University of Groningen, University Medical Center Groningen, Department of Laboratory Medicine, 9700 Groningen, RB, The Netherlands. [de Vries, G E Elisabeth] University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Hanzeplein 1, 9700 Groningen, RB, The Netherlands. [Hollema, Harry] University of Groningen, University Medical Center Groningen, Department of Pathology, 9700 Groningen, RB, The Netherlands. [Oosting, F Sjoukje] University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Hanzeplein 1, 9700 Groningen, RB, The Netherlands. RP Oosting, FS (reprint author), University of Groningen, University Medical Center Groningen, Department of Medical Oncology, 9700 Groningen, The Netherlands. EM s.oosting@umcg.nl CR Beaulieu JM, Gainetdinov RR, 2011, The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev 63:182– 217 Jonnakuty C, Gragnoli C, 2008, What do we know about serotonin? J Cell Physiol 217:301–306 Qin L, Zhao D, Xu J, Ren X, Terwilliger EF, Parangi S, Lawler J, Dvorak HF, Zeng H, 2013, The vascular permeabilizing factors histamine and serotonin induce angiogenesis through TR3/Nur77 and subsequently truncate it through thrombospondin-1. Blood 121:2154–2164 Zamani A, Qu Z, 2012, Serotonin activates angiogenic phosphorylation signaling in human endothelial cells. FEBS Lett 586:2360– 2365 Asada M, Ebihara S, Yamanda S, Niu K, Okazaki T, Sora I, Arai H, 2009, Depletion of serotonin and selective inhibition of 2B receptor suppressed tumor angiogenesis by inhibiting endothelial nitric oxide synthase and extracellular signal-regulated kinase 1/2 phosphorylation. Neoplasia 11:408–417 Basu S, Nagy JA, Pal S, Vasile E, Eckelhoefer IA, Susan Bliss V, Manseau EJ, Dasgupta PS, Dvorak HF, Mukhopadhyay D, 2001, The neurotransmitter dopamine inhibits angiogenesis induced by vascular permeability factor/vascular endothelial growth factor. Nat Med 7:569–574 Sarkar C, Chakroborty D, Chowdhury UR, Dasgupta PS, Basu S, 2008, Dopamine increases the efficacy of anticancer drugs in breast and colon cancer preclinical models. Clin Cancer Res 14: 2502–2510 Moreno-Smith M, Lee SJ, Lu C, Nagaraja AS, He G, Rupaimoole R, Han HD, Jennings NB, Roh JW, Nishimura M, Kang Y, Allen JK, Armaiz GN, Matsuo K, Shahzad MMK, Bottsford-Miller J, Langley RR, Cole SW, Lutgendorf SK, Siddik ZH, Sood AK, 2013, Biologic effects of dopamine on tumor vasculature in ovarian carcinoma. Neoplasia 15:502–510 Borcherding DS, Tong W, Hugo ER et al, 2016, Expression and therapeutic targeting of dopamine receptor-1, D1R, in breast cancer. Oncogene 35:3103–3113 Peters MA, Walenkamp AM, Kema IP, Meijer C, De Vries EG, Oosting SF, 2014, Dopamine and serotonin regulate tumor behavior by affecting angiogenesis. Drug Resist Updat 17:96–104 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144:646–674 Jandaghi P, Najafabadi HS, Bauer AS, Papadakis AI, Fassan M, Hall A, Monast A, von Knebel Doeberitz M, Neoptolemos JP, Costello E, Greenhalf W, Scarpa A, Sipos B, Auld D, Lathrop M, Park M, BuchlerMW, StrobelO, Hackert T, GieseNA, Zogopoulos G, Sangwan V, Huang S, Riazalhosseini Y, Hoheisel JD, 2016, Expression of DRD2 is increased in human pancreatic ductal adenocarcinoma and inhibitors slow tumor growth in mice. Gastroenterology 151:1218–1231 Jordan K, Gralla R, Jahn F, Molassiotis A, 2014, International antiemetic guidelines on chemotherapy induced nausea and vomiting, CINV): content and implementation in daily routine practice. Eur J Pharmacol 722:197–202 Peterlin BL, Rapoport AM, 2007, Clinical pharmacology of the serotonin receptor agonist, zolmitriptan. Expert Opin Drug Metab Toxicol 3:899–911 Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, Yefanov A, Lee H, Zhang N, Robertson CL, Serova N, Davis S, Soboleva A, 2013, NCBI GEO: archive for functional genomics data sets – update. Nucleic Acids Res 41:D991–D995 Fehrmann RS, Karjalainen JM, Krajewska M et al, 2015, Gene expression analysis identifies global gene dosage sensitivity in cancer. Nat Genet 47:115–125 Van Berkel A, Lenders JW, Timmers HJ, 2014, Diagnosis of endocrine disease: biochemical diagnosis of phaeochromocytoma and paraganglioma. Eur J Endocrinol 170:R109–R119 Tharayil VS, Wouters MM, Stanich JE et al, 2010, Lack of serotonin 5-HT2B receptor alters proliferation and network volume of interstitial cells of Cajal in vivo. Neurogastroenterol Motil 22: 462–469 Pore M, Meijer C, de Bock GH, Boersma-van Ek W, Terstappen LWMM, Groen HJM, Timens W, Kruyt FAE, Hiltermann TJN, 2016, Cancer stem cells, epithelial to mesenchymal markers, and circulating tumor cells in small cell lung cancer. Clin Lung Cancer 17:535–542 Muller K, Gilbertz KP, Meineke V, 2012, Serotonin and ionizing radiation synergistically affect proliferation and adhesion molecule expression of malignant melanoma cells. J Dermatol Sci 68:89–98 Henriksen R, Dizeyi N, Abrahamsson PA, 2012, Expression of serotonin receptors 5-HT1A, 5-HT1B, 5-HT2B and 5-HT4 in ovary and ovarian tumours. Anticancer Res 32:1361–1366 Kopparapu PK, Tinzl M, Anagnostaki L, Persson JL, Dizeyi N, 2013, Expression and localization of serotonin receptors in human breast cancer. Anticancer Res 33:363–370 Saveanu A, Sebag F, Guillet B, Archange C, EssametW, Barlier A, Palazzo FF, Taieb D, 2013, Targeting dopamine receptors subtype 2, D2DR, in pheochromocytomas: head-to-head comparison between in vitro and in vivo findings. J Clin Endocrinol Metab 98: E1951–E1955 Saveanu A, Muresan M, De Micco C et al, 2011, Expression of somatostatin receptors, dopamine D2 receptors, noradrenaline transporters, and vesicular monoamine transporters in 52 pheochromocytomas and paragangliomas. Endocr Relat Cancer 18:287–300 Stein MN, Bertino JR, Kaufman HL, Mayer T, Moss R, Silk A, Chan N,Malhotra J, Rodriguez L, Aisner J, Aiken RD, Haffty BG, DiPaola RS, Saunders T, Zloza A, Damare S, Beckett Y, Yu B, Najmi S, Gabel C, Dickerson S, Zheng L, el-Deiry WS, Allen JE, Stogniew M, Oster W, Mehnert JM, 2017, First-in-human clinical trial of oral ONC201 in patients with refractory solid tumors. Clin Cancer Res 23:4163–4169 Arrillaga-Romany I, Chi AS, Allen JE, OsterW,Wen PY, Batchelor TT, 2017, A phase 2 study of the imipridone ONC201, a selective DRD2 antagonist for oncology, administered every three weeks in recurrent glioblastoma. Oncotarget 8:79298–79304 Wang Y, Li J, Tohti M, Hu Y, Wang S, Li W, Lu Z, Ma C, 2014, The expression profile of dopamine D2 receptor, MGMT and VEGF in different histological subtypes of pituitary adenomas: a study of 197 cases and indications for medical therapy. J Exp Clin Cancer Res 33:56 Hyman DM, Taylor BS, Baselga J, 2017, Implementing genomedriven oncology. Cell 168:584–599 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1539 EP 1547 DI 10.1007/s12253-019-00734-w PG 9 ER PT J AU Sun, QX Wang, RR Liu, N Liu, Ch AF Sun, Qing-Xiu Wang, Rong-Rong Liu, Na Liu, Chao TI Dysregulation of miR-204-3p Driven by the Viability and Motility of Retinoblastoma via Wnt/β-catenin Pathway In Vitro and In Vivo SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Long non-coding RNA-BANCR; microRNA-204-3p; Retinoblastoma; WNT signaling pathway; Viability; Motility ID Long non-coding RNA-BANCR; microRNA-204-3p; Retinoblastoma; WNT signaling pathway; Viability; Motility AB Retinoblastoma (RB) is a malignant intraocular tumor that frequently occurs in infants and toddlers. Although the most of RB patients in the developed countries could survival from this cancer, the patients in undeveloped areas are still suffering. The human retinal pigment epithelial cell line ARPE-19 and human retinoblastoma (RB) cell lines HXO-RB44, Y79, and WERI-Rb1 were cultured. The mRNA levels of BANCR and miR-204-3p in these cell lines were measured by qRT-PCR. After transfection with sh-BANCR or treatment with miR-204-3p inhibitor in Y79 cells, the cell proliferation rate, growth, invasion, migration, apoptosis and Wnt/β-catenin signaling pathway activity were measured. The regular Y79 and Y79 cells stably expressed sh- BANCR were injected subcutaneously into nude mice, respectively. The volumes and pathohistological futures of tumors were compared. The biochemical features similar to the cell culture were detected and compered. The mRNA measurements showed that BANCR negatively modulate miR-204-3p expression via directly integration with it. Besides, miR-204-3p and Wnt/β- catenin signalling pathway were found to participate in the oncogenic effects of BANCR on RB cell line by Hoechst staining, cell Counting Kit-8 (CCK-8) assay, wound healing assay, Transwell assay, and Western blot analysis in vitro. In addition, an in vivo tumorigenesis experiment in nude mice injected with Y79 cells stably expressed sh-BANCR conformed in the effects of BANCR on RB. Taken together, the knockdown of BANCR inhibited cell proliferation, apoptosis, invasion, and migration in RB via targeting miR-204-3p, the mechanism may involve inhibiting Wnt/β-catenin signaling pathway. C1 [Sun, Qing-Xiu] The Second Clinical Medical College of Qingdao University, Department of OphthalmologyQingdao, China. [Wang, Rong-Rong] The Second Clinical Medical College of Qingdao University, Department of OphthalmologyQingdao, China. [Liu, Na] The Second Clinical Medical College of Qingdao University, Department of OphthalmologyQingdao, China. [Liu, Chao] The Second Clinical Medical College of Qingdao University, Department of OphthalmologyQingdao, China. RP Liu, Ch (reprint author), The Second Clinical Medical College of Qingdao University, Department of Ophthalmology, Qingdao, China. EM cha0liu@yandex.com CR Mendoza PR, Grossniklaus HE, 2015, The biology of retinoblastoma. Prog Mol Biol Transl Sci 134:503–516. , DOI 10. 1016/bs.pmbts.2015.06.012 Rodriguez-Galindo C, Orbach DB, VanderVeen D, 2015, Retinoblastoma. Pediatr Clin N Am 62:201–223. , DOI 10.1016/j.pcl.2014.09.014 Fabian ID, Onadim Z, Karaa E, Duncan C, Chowdhury T, Scheimberg I, Ohnuma SI, Reddy MA, Sagoo MS, 2018, The management of retinoblastoma. Oncogene 37:1551–1560. https:// doi.org/10.1038/s41388-017-0050-x Metz KA, Westerwick D, Driever F, Schmid KW, Le Guin CHD, 2017, Retinoblastoma and retinocytoma, retinoma). Pathologe 38: 507–514. , DOI 10.1007/s00292-017-0384-8 Kivela TT, Hadjistilianou T, 2017, Neonatal Retinoblastoma. Asia Pac J Oncol Nurs 4:197–204. , DOI 10.4103/apjon.apjon_18_17 Manjandavida FP, Honavar SG, Shields CL, Shields JA, 2013, Retinoblastoma: recent update and management Frontiers. Asia Pac J Ophthalmol, Phila, 2:351–353. , DOI 10.1097/ APO.0000000000000026 Lin P, O'Brien JM, 2009, Frontiers in the management of retinoblastoma. Am J Ophthalmol 148:192–198. , DOI 10.1016/ j.ajo.2009.04.004 Jabbour P, Chalouhi N, Tjoumakaris S, Gonzalez LF, Dumont AS, Chitale R, Rosenwasser R, Bianciotto CG, Shields C, 2012, Pearls and pitfalls of intraarterial chemotherapy for retinoblastoma. J Neurosurg Pediatr 10:175–181. , DOI 10.3171/2012.5.PEDS1277 Gatta G, Capocaccia R, Stiller C, Kaatsch P, Berrino F, Terenziani M, EUROCAREWorking Group, 2005, Childhood cancer survival trends in Europe: a EUROCARE working group study. J Clin Oncol 23:3742–3751. , DOI 10.1200/JCO.2005.00.554 Lanzafame M, Bianco G, Terracciano LM, Ng CKY, Piscuoglio S, 2018, The role of long non-coding RNAs in Hepatocarcinogenesis. Int J Mol Sci 19. , DOI 10. 3390/ijms19030682 Clark RJ, Craig MP, Agrawal S, Kadakia M, 2018, microRNA involvement in the onset and progression of Barrett's esophagus: a systematic review. Oncotarget 9:8179–8196. , DOI 10. 18632/oncotarget.24145 Zhou S,WangL, YangQ, LiuH,MengQ, JiangL,Wang S, JiangW, 2018, Systematical analysis of lncRNA-mRNA competing endogenous RNA network in breast cancer subtypes. Breast Cancer Res Treat 169:267–275. , DOI 10.1007/s10549-018-4678-1 Koleckova M, Janikova M, Kolar Z, 2018, MicroRNAs in triplenegative breast cancer. Neoplasma 65:1–13. , DOI 10. 4149/neo_2018_170115N36 Momen-Heravi F, Bala S, 2018, Emerging role of non-coding RNA in oral cancer. Cell Signal 42:134–143. , DOI 10.1016/j. cellsig.2017.10.009 Li H, Chen S, Liu J, Guo X, Xiang X, Dong T, Ran P, Li Q, Zhu B, Zhang X,Wang D, Xiao C, Zheng S, 2018, Long non-coding RNA PVT1-5 promotes cell proliferation by regulating miR-126/ SLC7A5 axis in lung cancer. Biochem Biophys Res Commun 495:2350–2355. , DOI 10.1016/j.bbrc.2017.12.114 Zhang M, Li Q, Pan Y, Wang H, Liu G, Yin H, 2018, MicroRNA- 655 attenuates the malignant biological behaviours of retinoblastoma cells by directly targeting PAX6 and suppressing the ERK and p38MAPK signalling pathways. Oncol Rep 39:2040–2050. https:// doi.org/10.3892/or.2018.6264 Wu X, Zeng Y, Wu S, Zhong J, Wang Y, Xu J, 2015, MiR-204, down-regulated in retinoblastoma, regulates proliferation and invasion of human retinoblastoma cells by targeting CyclinD2 and MMP-9. FEBS Lett 589:645–650. , DOI 10.1016/j. febslet.2015.01.030 Su S, Gao J, Wang T, Wang J, Li H, Wang Z, 2015, Long noncoding RNA BANCR regulates growth and metastasis and is associated with poor prognosis in retinoblastoma. Tumour Biol 36: 7205–7211. , DOI 10.1007/s13277-015-3413-3 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T), method. Methods, San Diego, Calif., 25:402–408. https:// doi.org/10.1006/meth.2001.1262 Rion N, Ruegg MA, 2017, LncRNA-encoded peptides: more than translational noise? Cell Res 27:604–605. https://doi. org/10.1038/cr.2017.35 Dey BK, Mueller AC, Dutta A, 2014, Long non-coding RNAs as emerging regulators of differentiation, development, and disease. Transcription 5:e944014. , DOI 10.4161/21541272.2014.944014 Deng H, Zhang J, Shi J, Guo Z, He C, Ding L, Tang JH, Hou Y, 2016, Role of long non-coding RNA in tumor drug resistance. Tumour Biol 37:11623–11631. , DOI 10.1007/s13277- 016-5125-8 Vikram R, Ramachandran R, Abdul KS, 2014, Functional significance of long non-coding RNAs in breast cancer. Breast Cancer 21: 515–521. , DOI 10.1007/s12282-014-0554-y Osielska MA, Jagodzinski PP, 2018, Long non-coding RNA as potential biomarkers in non-small-cell lung cancer: what do we know so far? Biomed Pharmacother 101:322–333. , DOI 10.1016/j.biopha.2018.02.099 Zhuang J, He S, Wang G, Wang G, Ni J, Zhang S, Ye Y, Xia W, 2018, Long non-coding RNA FGFR3-AS1 promotes hepatocellular carcinoma carcinogenesis via modulating PI3K/AKT pathway. Oncol Res 26:1257–1265. , DOI 10.3727/ 096504018X15172756878992 Peng Z, Liu C, Wu M, 2018, New insights into long noncoding RNAs and their roles in glioma. Mol Cancer 17:61. , DOI 10.1186/s12943-018-0812-2 Flockhart RJ, Webster DE, Qu K, Mascarenhas N, Kovalski J, Kretz M, Khavari PA, 2012, BRAFV600E remodels the melanocyte transcriptome and induces BANCR to regulate melanoma cell migration. Genome Res 22:1006–1014. , DOI 10.1101/gr.140061.112 Liu XF, Hao JL, Xie T, Pant OP, Lu CB, Lu CW, Zhou DD, 2018, The BRAF activated non-coding RNA: a pivotal long non-coding RNA in human malignancies. Cell Prolif 51:e12449. https://doi. org/10.1111/cpr.12449 Shen X, Bai Y, Luo B, Zhou X, 2017, Upregulation of lncRNA BANCR associated with the lymph node metastasis and poor prognosis in colorectal cancer. Biol Res 50:32. , DOI 10.1186/ s40659-017-0136-5 Liao T, Qu N, Shi RL, Guo K, Ma B, Cao YM, Xiang J, Lu ZW, Zhu YX, Li DS, Ji QH, 2017, BRAF-activated LncRNA functions as a tumor suppressor in papillary thyroid cancer. Oncotarget 8: 238–247. , DOI 10.18632/oncotarget.10825 Liu A, Liu S, 2016, Noncoding RNAs in growth and death of Cancer cells. Adv Exp Med Biol 927:137–172. , DOI 10. 1007/978-981-10-1498-7_5 Review Khorshidi A, Dhaliwal P, Yang BB, 2016, Noncoding RNAs in tumor angiogenesis. Adv Exp Med Biol 927:217–241. https://doi. org/10.1007/978-981-10-1498-7_8 Slattery ML, Mullany LE, Sakoda LC, Samowitz WS, Wolff RK, Stevens JR, Herrick JS, 2018, Expression of Wnt-signaling pathway genes and their associations with miRNAs in colorectal cancer. Oncotarget 9:6075–6085. , DOI 10.18632/oncotarget.23636 Wei H, Zhang JJ, Tang QL, 2017)MiR-638 inhibits cervical cancer metastasis through Wnt/beta-catenin signaling pathway and correlates with prognosis of cervical cancer patients. Eur Rev Med Pharmacol Sci 21:5587–5593. , DOI 10.26355/eurrev_ 201712_13999 Huang J, He Y,McLeod HL, Xie Y, Xiao D, Hu H, Chen P, Shen L, Zeng S, Yin X, Ge J, Li L, Tang L, Ma J, Chen Z, 2017, miR-302b inhibits tumorigenesis by targeting EphA2 via Wnt/ beta-catenin/ EMT signaling cascade in gastric cancer. BMC Cancer 17:886. , DOI 10.1186/s12885-017-3875-3 Krishnamurthy N, Kurzrock R, 2018, Targeting the Wnt/beta-catenin pathway in cancer: update on effectors and inhibitors. Cancer Treat Rev 62:50–60. , DOI 10.1016/j.ctrv.2017.11.002 Majidinia M, Aghazadeh J, Jahanban-Esfahlani R, Yousefi B, 2018, The roles of Wnt/beta-catenin pathway in tissue development and regenerative medicine. J Cell Physiol 233:5598–5612. , DOI 10.1002/jcp.26265 Peng Y, Zhang X, Feng X, Fan X, Jin Z, 2017, The crosstalk between microRNAs and the Wnt/beta-catenin signaling pathway in cancer. Oncotarget 8:14089–14106. , DOI 10.18632/ oncotarget.12923 Onyido EK, Sweeney E, Nateri AS, 2016, Wnt-signalling pathways and microRNAs network in carcinogenesis: experimental and bioinformatics approaches. Mol Cancer 15:56. , DOI 10.1186/s12943-016-0541-3 Zhang K, Zhang J, Han L, Pu P, Kang C, 2012, Wnt/beta-catenin signaling in glioma. J NeuroImmune Pharmacol 7:740–749. https:// doi.org/10.1007/s11481-012-9359-y He H, Chen K, Wang F, Zhao L, Wan X, Wang L, Mo Z, 2015, miR-204-5p promotes the adipogenic differentiation of human adipose-derived mesenchymal stem cells by modulating DVL3 expression and suppressing Wnt/beta-catenin signaling. Int J Mol Med 35:1587–1595. , DOI 10.3892/ijmm.2015.2160 Wang L, Tian H, Yuan J, Wu H, Wu J, Zhu X, 2015, CONSORT: Sam68 is directly regulated by MiR-204 and promotes the selfrenewal potential of breast Cancer cells by activating the Wnt/ Beta-catenin signaling pathway. Medicine, Baltimore, 94:e2228. , DOI 10.1097/MD.0000000000002228 Zou Y, Li J, Chen Y, Xiao H, Zhang F, Yu D, Luo K, 2017, BANCR: a novel oncogenic long non-coding RNA in human cancers. Oncotarget 8:94997–95004. , DOI 10.18632/ oncotarget.22031 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1549 EP 1558 DI 10.1007/s12253-019-00722-0 PG 10 ER PT J AU Liu, L Zhang, JW Zhu, NSh Zhu, Y Guo, B Yang, XH AF Liu, Liu Zhang, Jin-Wei Zhu, Ni-Sha Zhu, Yun Guo, Bing Yang, Xi-Hu TI Clear Cell Odontogenic Carcinoma: a Clinicopathological and Immunocytochemical Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Head and neck; Clear cell odontogenic carcinoma; Oral cancer; Prognosis ID Head and neck; Clear cell odontogenic carcinoma; Oral cancer; Prognosis AB Clear cell odontogenic carcinoma (CCOC) is a rare odontogenic tumor associated with aggressive clinical behavior, metastasis and low survival. To date, only 67 cases have been described in the English language literature, and an understanding of the behavior of CCOC has been based on limited case reports. The aim of the research was to further reveal the features of CCOC. We report 5 new cases of CCOC, with a mean age of 52.4 years. The clinical and histopathologic data of the disease obtained from earlier literature (95 cases) and the 5 new cases were analyzed. Data were extracted, including demographics, histopathologic findings, clinical presentation, primary treatment and outcomes. Immunohistochemical results revealed that the cancer is positive for AE1/AE3, EMA and CK19, negative for smooth muscle actin SMA, Vim and S-100. EWSR1 translocation was also observed in the new cases, which may help in the diagnosis of CCOC. Metastases of CCOC were rare, but the local recurrence rate of CCOC rose to 42%. The best treatment for patients with CCOC is wide local excision combined with regional lymph node dissection. C1 [Liu, Liu] Shanghai Jiao Tong University School of Medicine, Ninth People’s Hospital, Department of Oral and Maxillofacial-Head & Neck Oncology, 200011 Shanghai, China. [Zhang, Jin-Wei] Third Central Hospital, Branch of Tianjin, Department of Stomatology, 300000 Tianjin, China. [Zhu, Ni-Sha] Medical School of Nanjing University, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial Surgery, 210000 Nanjing, Jiangsu, China. [Zhu, Yun] Shanghai Jiao Tong University School of Medicine, Ninth People’s Hospital, Department of Oral and Maxillofacial-Head & Neck Oncology, 200011 Shanghai, China. [Guo, Bing] Shanghai Jiao Tong University School of Medicine, Ninth People’s Hospital, Department of Oral and Maxillofacial-Head & Neck Oncology, 200011 Shanghai, China. [Yang, Xi-Hu] Affiliated Hospital of Jiangsu University, Department of Oral and Maxillofacial Surgery, 212000 Zhenjiang, Jiangsu, China. RP Guo, B (reprint author), Shanghai Jiao Tong University School of Medicine, Ninth People’s Hospital, Department of Oral and Maxillofacial-Head & Neck Oncology, 200011 Shanghai, China. EM doctorguo117@sina.com CR Piattelli A, Sesenna E, Trisi P et al, 1994, Clear cell odontogenic carcinoma: report of a case with lymph node and pulmonary metastases. Eur J Cancer B Oral Oncol 30 B:278–280 Brinck U, Gunawan B, Schulten HJ et al, 2001, Clear cell odontogenic carcinoma with pulmonary metastases resembling pulmonary meningothelial-like nodules. Virchows Arch 438:412– 417 Hansen LS, Eversole LR, Green TL et al, 1985, Clear cell odontogenic tumour: a new histologic variant with aggressive potential. Head Neck Surg 8:115–123 Hadj Said M, Ordioni U, Benat G et al., 2017, Clear cell odontogenic carcinoma. A review. J Stomatol OralMaxillofac Surg Ordioni U, Benat G, Hadj Said M, et al., 2017, Clear cell odontogenic carcinoma, diagnostic difficulties. A case report. J Stomatol Oral Maxillofac Surg Datar UV, Kamat MS, Kanitkar SS et al, 2017, Clear cell odontogenic carcinoma: a rare case report with emphasis on differential diagnosis. J Cancer Res Ther 13(2):374–377 Muramatsu T, Hashimoto S, 1996, Inoue Tet al. Clear cell odontogenic carcinoma in the mandible:histochemical and immunohistochemical observations with a review of the literature. J Oral Pathol Med 25(9):516–521 Swain N, Dhariwal R, Ray JG et al, 2013, Clear cell odontogenic carcinoma of maxilla: a case report and mini review. J Oral Maxillofac Pathol 17:89–94 Chaine A, Pitak-Arnnop P, Dhanuthai K et al, 2009, An asymptomatic radiolucent lesion of the maxilla. Clear cellodontogenic carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 107:452–457 Prakash AR, Sairam V, Srinivas Reddy P et al, 2015, Clear cell odontogenic carcinoma-a rare case report. J Maxillofac Oral Surg 14(Suppl 1):60–63 Martinez Martinez M, Mosqueda-Taylor A, Carlos R et al, 2014, Malignant odontogenic tumors: amulticentric Latin American study of 25 cases. Oral Dis 20:380–385 Loyola AM, Cardoso SV, de Faria PR et al, 2015, Clear cell odontogenic carcinoma: report of 7 new cases and systematic review of the current knowledge. Oral Surg Oral Med Oral Pathol Oral Radiol 120(4):483–496 Chera BS, Villaret DB, Orlando CA et al, 2008, Clear cell odontogenic carcinoma of the maxilla: a case report and literature review. Am J Otolaryngol 29(4):284–290 Carinci F, Volinia S, Rubini C et al, 2003, Genetic profile of clear cell odontogenic carcinoma. J Craniofac Surg 14:356–362 Bilodeau EA, Weinreb I, Antonescu CR et al, 2013, Clear cell odontogenic carcinomas show EWSR1 rearrangements: a novel finding and a biological link to salivary clear cell carcinomas. Am J Surg Pathol 37(7):1001–1005 Ginat DT, Villaflor V, Cipriani NA et al, 2016, Oral cavity clear cell odontogenic carcinoma. Head Neck Pathol 10(2):217–220 Zhang J, Liu L, Pan J et al, 2011, Clear cell odontogenic carcinoma: report of 6 cases and review of the literature. Med Oncol 28(Suppl 1):S626–S633 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1559 EP 1564 DI 10.1007/s12253-019-00741-x PG 6 ER PT J AU Sirak, I Pohankova, D Ferko, A Hovorkova, E Rozkos, T Vosmik, M Hodek, M Paluska, P Buka, D Grepl, J Petera, J AF Sirak, Igor Pohankova, Denisa Ferko, Alexander Hovorkova, Eva Rozkos, Tomas Vosmik, Milan Hodek, Miroslav Paluska, Petr Buka, David Grepl, Jakub Petera, Jiri TI The Time Between Chemoradiation and Surgery for Rectal Carcinoma Negatively Influences Mesorectal Excision Quality SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Rectal carcinoma; Neoadjuvant chemoradiation; Mesorectal excision ID Rectal carcinoma; Neoadjuvant chemoradiation; Mesorectal excision AB Total mesorectal excision quality (TMEq) is a prognostic factor associated with local recurrence in rectal adenocarcinoma. Neoadjuvant chemoradiotherapy (NCRT) reduces the risk of tumor recurrence, but may compromise TMEq. The time between NCRT and surgery (TTS) and how it influences TMEq and tumor control were evaluated. In prospective registry, 236 patients after NCRT and TME were analyzed. NCRT involved radiotherapy with 45 Gy to the pelvis, plus tumor boost dose 5.4 Gy with concurrent 5-fluorouracil infusion. NCRT was followed by TME after 9 weeks on average (median 9.4 ± SD 2.5). TMEq was parametrically analyzed by standard three-grade system. With median follow-up of 47.5 months, 3-year overall survival (OS) was 83.8%, disease-free survival (DFS) was 77.7%, and 6.4% was the rate of local recurrence (LR). TTS was not associated with OS, DFS, or LR. TMEq was found to be associated with LR in univariate analysis, but not in multivariate, where pathological tumor stage and resection margins remained dominant predictors. TMEq was negatively influenced by inferior location of the tumor, longer TTS, higher tumor and nodal stage, presence of tumor perforation, perineural invasion, and close/positive resection margins. Nonetheless, TTS remained a strong predictor of TMEq in multivariate analyses. TTS was proven to be an independent predictor of TMEq. With longer TTS, fewer complete TME with intact mesorectal plane were observed. However, TTS was not associated with survival deterioration or tumor recurrence. These were negatively influenced by other factors interfering with TMEq, especially by pathological tumor stage and resection margins. C1 [Sirak, Igor] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, Sokolska 581, 500 05 Hradec Kralove, Czech Republic. [Pohankova, Denisa] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, Sokolska 581, 500 05 Hradec Kralove, Czech Republic. [Ferko, Alexander] Comenius University Bratislava, Jessenius Faculty in Martin, University Hospital Martin, Department of Surgery and Transplant CentreBratislava, Slovakia. [Hovorkova, Eva] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, The Fingerland Department of PathologyHradec Kralove, Czech Republic. [Rozkos, Tomas] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, The Fingerland Department of PathologyHradec Kralove, Czech Republic. [Vosmik, Milan] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, Sokolska 581, 500 05 Hradec Kralove, Czech Republic. [Hodek, Miroslav] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, Sokolska 581, 500 05 Hradec Kralove, Czech Republic. [Paluska, Petr] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, Sokolska 581, 500 05 Hradec Kralove, Czech Republic. [Buka, David] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, Sokolska 581, 500 05 Hradec Kralove, Czech Republic. [Grepl, Jakub] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, Sokolska 581, 500 05 Hradec Kralove, Czech Republic. [Petera, Jiri] Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, Sokolska 581, 500 05 Hradec Kralove, Czech Republic. RP Vosmik, M (reprint author), Charles University, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Department of Oncology and Radiotherapy, 500 05 Hradec Kralove, Czech Republic. EM milan.vosmik@fnhk.cz CR Parfitt JR, Driman DK, 2007, The total mesorectal excision specimen for rectal cancer: a review of its pathological assessment. J Clin Pathol 60(8):849–855 Heald RJ, Ryall RD, 1986, Recurrence and survival after total mesorectal excision for rectal cancer. Lancet 1(8496):1479–1482 Kapiteijn E, Putter H, van de Velde CJ, Cooperative investigators of the Dutch ColoRectal Cancer Group, 2002, Impact of the introduction and training of total mesorectal excision on recurrence and survival in rectal cancer in the Netherlands. Br J Surg 89(9): 1142–1149 van Gijn W, Marijnen CA, Nagtegaal ID et al, 2011, Preoperative radiotherapy combined with totalmesorectal excision for resectable rectal cancer: 12-year follow-up of the multicentre, randomised controlled TME trial. Lancet Oncol 12(6):575–582 Bosset JF, Calais G, Mineur L et al, 2014, Fluorouracil-based adjuvant chemotherapy after preoperative chemoradiotherapy in rectal cancer: long-term results of the EORTC 22921 randomised study. Lancet Oncol 15(2):184–190 Sauer R, Becker H, HohenbergerWet al, 2004, Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med 351(17):1731–1740 Sauer R, Liersch T, Merkel S et al, 2012, Preoperative versus postoperative chemoradiotherapy for locally advanced rectal cancer: results of the German CAO/ARO/AIO-94 randomized phase III trial after a median follow-up of 11 years. J Clin Oncol 30(16): 1926–1933 Nagtegaal ID, van de Velde CJ, van der Worp E et al, 2002, Macroscopic evaluation of rectal cancer resection specimen: clinical significance of the pathologist in quality control. J Clin Oncol 20(7):1729–1734 Leonard D, Penninckx F, Fieuws S et al, 2010, PROCARE, a multidisciplinary Belgian project on Cancer of the rectum. Factors predicting the quality of total mesorectal excision for rectal cancer. Ann Surg 252(6):982–988 Baik SH, Kim NK, Lee KY et al, 2008, Factors influencing pathologic results after total mesorectal excision for rectal cancer: analysis of consecutive 100 cases. Ann Surg Oncol 15(3):721–728 Deng H, Chen H, Zhao L et al, 2015, Quality of laparoscopic total mesorectal excision: results from a single institution in China. Hepatogastroenterology 62(138):264–267 Garlipp B, Ptok H, Schmidt U et al, 2012, Factors influencing the quality of total mesorectal excision. Br J Surg 99(5):714–720 Bosch SL, Nagtegaal ID, 2012, The importance of the Pathologist's role in assessment of the quality of the Mesorectum. Curr Colorectal Cancer Rep 8(2):90–98 Roh MS, Colangelo LH, O'Connell MJ et al, 2009, Preoperative multimodality therapy improves disease-free survival in patients with carcinoma of the rectum: NSABP R-03. J Clin Oncol 27: 5124–5130 Sautter-Bihl ML, Hohenberger W, Fietkau R et al, 2013, Rectal cancer: when is the local recurrence risk low enough to refrain from the aim to prevent it? Strahlenther Onkol 189(2):105–110 Bujko K, Nowacki MP, Nasierowska-Guttmejer A et al, 2006, Long-term results of a randomized trial comparing preoperative short-course radiotherapy with preoperative conventionally fractionated chemoradiation for rectal cancer. Br J Surg 93(10):1215– 1223 Hernando-Requejo O, Lopez M, Cubillo A et al, 2014, Complete pathological responses in locally advanced rectal cancer after preoperative IMRT and integrated-boost chemoradiation. Strahlenther Onkol 190(6):515–520 Wong RK, Tandan V, De Silva S, FigueredoA, 2007, Pre-operative radiotherapy and curative surgery for the management of localized rectal carcinoma. Cochrane Database Syst Rev 18(2):CD002102 Foster JD, Jones EL, Falk S et al, 2013, Timing of surgery after long-course neoadjuvant chemoradiotherapy for rectal cancer: a systematic review of the literature. Dis Colon Rectum 56(7):921– 930 Sun Z, Adam MA, Kim J et al, 2016, Optimal timing to surgery after neoadjuvant Chemoradiotherapy for locally advanced rectal Cancer. J Am Coll Surg 222(4):367–374 Pellizzon ACA, 2019, Pre-operative radiotherapy to improve local control and survival in rectal cancer optimal time intervals between radiation and surgery. Pract Rep Oncol Radiother 24(1):1–2 Petersen C, 2013, Optimal timing of surgery for rectal cancer after neoadjuvant chemoradiotherapy: which patients would benefit from waiting? Strahlenther Onkol 189(10):904–906 Sloothaak DA, Geijsen DE, van Leersum NJ et al, 2013, Optimal time interval between neoadjuvant chemoradiotherapy and surgery for rectal cancer. Br J Surg 100(7):933–939 Lefevre JH, Mineur L, Kotti S et al, 2016, Effect of interval, 7 or 11 weeks, between neoadjuvant Radiochemotherapy and surgery on complete pathologic response in rectal Cancer: a multicenter, randomized, controlled trial, GRECCAR-6). J Clin Oncol 34(31): 3773–3778 Buka D, Dvorak J, Sitorova Vet al, 2017, The changes of tumour vascular endothelial growth factor expression after neoadjuvant chemoradiation in patients with rectal adenocarcinoma. Contemp Oncol 21(1):48–53 Hodek M, Sirak I, Ferko A et al, 2016, Neoadjuvant chemoradiotherapy of rectal carcinoma : baseline hematologic parameters influencing outcomes. Strahlenther Onkol 192(9):632–640 Dvorak J, Sitorova V, Ryska A et al, 2012, Prognostic significance of changes of tumor epidermal growth factor receptor expression after neoadjuvant chemoradiation in patients with rectal adenocarcinoma. Strahlenther Onkol 188(9):833–838 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1565 EP 1572 DI 10.1007/s12253-019-00742-w PG 8 ER PT J AU Raju K, L Haragannavar, CV Patil, Sh Rao, SR Nagaraj, T Augustine, D Venkatesiah, SS Nambiar, Sh AF Raju K, Lizbeth Haragannavar, C Vanishri Patil, Shankargouda Rao, S Roopa Nagaraj, Tejavathi Augustine, Dominic Venkatesiah, S Sowmya Nambiar, Shwetha TI Expression of hTERT in Oral Submucous Fibrosis and Oral Squamous Cell Carcinoma – an Immunohistochemical Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Biomarkers; Carcinogenesis; Immunohistochemistry; Oral submucous fibrosis; Squamous cell carcinoma; Telomerase ID Biomarkers; Carcinogenesis; Immunohistochemistry; Oral submucous fibrosis; Squamous cell carcinoma; Telomerase AB Human telomerase reverse transcriptase enzyme, the catalytic subunit of telomerase are seen to be frequently reactivated in cancers including Oral squamous cell carcinoma (OSCC). Increased hTERT expression have been seen in potentially malignant conditions including Oral submucous fibrosis (OSMF). The aim of the study was to evaluate the expression levels in OSMF, OSCC in the background of OSMF and OSCC using immunohistochemistry and also to correlate hTERT expression with clinicopathologic parameters. A total of 50 histopathologically diagnosed cases of 20 OSMF, 20 OSCC wherein 5 were OSCC in the background of OSMF and 10 Normal oral mucosae were retrieved from the departmental archives and subjected to immunohistochemical analysis of hTERT. The expression of hTERT increased from normal, OSMF, to OSCC with statistically significant differences in mean labelling score (LS). We also found a shift in cellular localization of stain where, normal mucosal tissues showed a nuclear stain unlike OSMF, where combined nuclear and cytoplasmic staining as noted. The tumor cells in OSCC showed predominant cytoplasmic staining. There was no correlation between hTERT expression and clinicopathological parameters of OSMF. However, a significant increase of hTERT expression was seen with increasing histological grading of OSCC. These results suggest the role of hTERT in the early event of malignant transformation of OSMF. Telomerase could be used as a potent diagnostic marker to identify high-risk group of OSMF. C1 [Raju K, Lizbeth] Ramaiah University of Applied Sciences, Faculty of Dental Sciences, Department of Oral Pathology and Microbiology, M S R Nagar, 560054 Bangalore, Karnataka, India. [Haragannavar, C Vanishri] Ramaiah University of Applied Sciences, Faculty of Dental Sciences, Department of Oral Pathology and Microbiology, M S R Nagar, 560054 Bangalore, Karnataka, India. [Patil, Shankargouda] Jazan University, College of Dentistry, Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral PathologyJazan, Saudi Arabia. [Rao, S Roopa] Ramaiah University of Applied Sciences, Faculty of Dental Sciences, Department of Oral Pathology and Microbiology, M S R Nagar, 560054 Bangalore, Karnataka, India. [Nagaraj, Tejavathi] M.S. Ramaiah Dental College & Hospital, Department of Oral Medicine, Diagnosis and Radiology, Cholanagar, R.T. Nagar post, 560032 Bangalore, Karnataka, India. [Augustine, Dominic] Ramaiah University of Applied Sciences, Faculty of Dental Sciences, Department of Oral Pathology and Microbiology, M S R Nagar, 560054 Bangalore, Karnataka, India. [Venkatesiah, S Sowmya] Ramaiah University of Applied Sciences, Faculty of Dental Sciences, Department of Oral Pathology and Microbiology, M S R Nagar, 560054 Bangalore, Karnataka, India. [Nambiar, Shwetha] Ramaiah University of Applied Sciences, Faculty of Dental Sciences, Department of Oral Pathology and Microbiology, M S R Nagar, 560054 Bangalore, Karnataka, India. RP Patil, Sh (reprint author), Jazan University, College of Dentistry, Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral Pathology, Jazan, Saudi Arabia. EM dr.ravipatil@gmail.com CR Warnakulasuriya S, 2009, Global epidemiology of oral and oropharyngeal cancer. Oral Oncol 45:309–316. , DOI 10.1016/j. oraloncology.2008.06.002 Johnson NW, Amarasinghe HK, 2011, Epidemiology and aetiology of head and neck cancers. In: Head and neck cancer. Springer New York, New York, pp 1–40 Wanninayake Mudiyanselage Tilakaratne RPE, 2013, Oral submucous fibrosis: review on mechanisms of pathogenesis and malignant transformation. J Carcinog Mutagen. , DOI 10. 4172/2157-2518.S5-002 Hsue S-S, Wang W-C, Chen C-H, Lin CC, Chen YK, Lin LM, 2007, Malignant transformation in 1458 patients with potentially malignant oral mucosal disorders: a follow-up study based in a Taiwanese hospital. J Oral Pathol Med 36:25–29. , DOI 10.1111/j.1600-0714.2006.00491.x Vinay K, Abbas AK, 2010, Robbins & cotran pathologic basis of disease, 9th edn. Saunders Elsevier, Philadelphia BhagvanMore C, Gupta S, Joshi J, Verma SN, 2012, Classification system for oral submucous fibrosis. J Indian Acad OralMed Radiol 24:24–29. , DOI 10.5005/jp-journals-10011-1254 Bhargava A, Saigal S, Chalishazar M, 2010, Systems in oral squamous cell carcinoma : a review. J Int Oral Heal 2:1–10 Chen H-H, Yu C-H,Wang J-T, Liu BY,Wang YP, Sun A, Tsai TC, Chiang CP, 2007, Expression of human telomerase reverse transcriptase, hTERT, protein is significantly associated with the progression, recurrence and prognosis of oral squamous cell carcinoma in Taiwan. Oral Oncol 43:122–129. , DOI 10.1016/j. oraloncology.2006.01.011 Luzar B, Poljak M, Marin IJ, Eberlinc A, Klopcic U, Gale N, 2004, Human telomerase catalytic subunit gene re-expression is an early event in oral carcinogenesis. Histopathology 45:13–19. https://doi. org/10.1111/j.1365-2559.2004.01892.x Sharpless NE, DePinho RA, 2004, Telomeres, stem cells, senescence, and cancer. J Clin Invest 113:160–168. , DOI 10. 1172/JCI20761 Ito H, Kyo S, Kanaya T, Takakura M, Inoue M, Namiki M, 1998, Expression of human telomerase subunits and correlation with telomerase activity in urothelial cancer. Clin Cancer Res 4:1603–1608 Kim NW, Piatyszek MA, Prowse KR, Harley C, West M, Ho P, Coviello G, Wright W, Weinrich S, Shay J, 1994, Specific association of human telomerase activity with immortal cells and cancer. Science 266:2011–2015 Kapanadze B, Morris E, Smith E, Trojanowska M, 2010, Establishment and characterization of scleroderma fibroblast clonal cell lines by introduction of the hTERT gene. J Cell Mol Med 14: 1156–1165. , DOI 10.1111/j.1582-4934.2009.00773.x Wieser M, Stadler G, Jennings P, Streubel B, Pfaller W, Ambros P, Riedl C, Katinger H, Grillari J, Grillari-Voglauer R, 2008, hTERT alone immortalizes epithelial cells of renal proximal tubuleswithout changing their functional characteristics. Am J Physiol Renal Physiol 295:F1365–F1375. , DOI 10.1152/ajprenal. 90405.2008 Shay JW, Bacchetti S, 1997, A survey of telomerase activity in human cancer. Eur J Cancer 33:787–791. , DOI 10.1016/ S0959-8049(97)00062-2 Zhao T, HU F, Qiao B et al, 2015, Telomerase reverse transcriptase potentially promotes the progression of oral squamous cell carcinoma through induction of epithelial-mesenchymal transition. Int J Oncol 46:2205–2215. , DOI 10.3892/ijo.2015.2927 Astuti AK, Prasetyo I, Yunita JN et al, 2011, Profile of human TERTand P73 in oral squamous cell carcinoma cell lines compared to normal human oral mucosa: a preliminary study. J Dent Indones 18:10–16. , DOI 10.14693/jdi.v18i1.55 Rai A, Naikmasur VG, Sattur A, 2016, Quantification of telomerase activity in normal oral mucosal tissue and oral squamous cell carcinoma. Indian J Med Paediatr Oncol 37:183–188. , DOI 10.4103/0971-5851.190350 Miyazaki Y, Yoshida N, Nozaki T, Inoue H, Kikuchi K, Kusama K, 2015, Telomerase activity in the occurrence and progression of oral squamous cell carcinoma. J Oral Sci 57:295–303. , DOI 10.2334/josnusd.57.295 Abrahao AC, Bonelli BV, Nunes FD, Dias EP, Cabral MG, 2011, Immunohistochemical expression of p53, p16 and hTERT in oral squamous cell carcinoma and potentially malignant disorders. Braz Oral Res 25:34–41 Palani J, Lakshminarayanan V, Kannan R, 2011, Immunohistochemical detection of human telomerase reverse transcriptase in oral cancer and pre-cancer. Indian J Dent Res 22:362– 363. , DOI 10.4103/0970-9290.84281 Kannan S, Tahara H,YokozakiH et al, 1997, Telomerase activity in premalignant and malignant lesions of human oral mucosa. Cancer Epidemiol Biomark Prev 6:413–420 Pannone G, DeMaria S, Zamparese R et al, 2007, Prognostic value of human telomerase reverse transcriptase gene expression in oral carcinogenesis. Int J Oncol 30:1349–1357 Weinhold N, Jacobsen A, Schultz N, Sander C, Lee W, 2014, Genome-wide analysis of noncoding regulatory mutations in cancer. Nat Genet 46:1160–1165. , DOI 10.1038/ng.3101 Chang K-P, Wang C-I, Pickering CR, Huang Y, Tsai CN, Tsang NM, Kao HK, Cheng MH, Myers JN, 2017, Prevalence of promoter mutations in the TERT gene in oral cavity squamous cell carcinoma. Head Neck 39:1131–1137. , DOI 10.1002/hed.24728 Vinothkumar V, Arunkumar G, Revathidevi S, Arun K, Manikandan M, Rao AKDM, Rajkumar KS, Ajay C, Rajaraman R, Ramani R,Murugan AK,Munirajan AK, 2016, TERT promoter hot spot mutations are frequent in Indian cervical and oral squamous cell carcinomas. Tumor Biol 37:7907–7913. , DOI 10.1007/s13277-015-4694-2 Qu Y, Dang S, Wu K, Shao Y, Yang Q, Ji M, Shi B, Hou P, 2014, TERT promoter mutations predict worse survival in laryngeal cancer patients. Int J Cancer 135:1008–1010. , DOI 10.1002/ ijc.28728 Yin L, Hubbard AK, Giardina C, 2000, NF-κB regulates transcription of the mouse telomerase catalytic subunit. J Biol Chem 275: 36671–36675. , DOI 10.1074/jbc.M007378200 ZhangY, Toh L, Lau P,WangX, 2012, Human telomerase reverse transcriptase, hTERT , is a novel target of the Wnt/β- catenin pathway in human cancer. J Biol Chem 287:32494– 32511. , DOI 10.1074/jbc.M112.368282 Greenberg RA, O’Hagan RC, Deng H et al, 1999, Telomerase reverse transcriptase gene is a direct target of c-Myc but is not functionally equivalent in cellular transformation. Oncogene 18: 1219–1226. , DOI 10.1038/sj.onc.1202669 Fabricius E-M, Kruse-Boitschenko U, Khoury R et al, 2009, Immunohistochemical determination of the appropriate antihTERT antibodies for in situ detection of telomerase activity in frozen sections of head and neck squamous cell carcinomas and tumor margin tissues. Int J Oncol 34:1257–1279. , DOI 10.1038/sj.onc.1203890 Hoffmann D, Brunnemann KD, Prokopczyk B, Djordjevic MV, 1994, Tobacco-specific N-nitrosamines and Areca-derived N-nitrosamines: chemistry, biochemistry, carcinogenicity, and relevance to humans. J Toxicol Environ Health 41:1–52. , DOI 10. 1080/15287399409531825 Xue J, Yang S, Seng S, 2014, Mechanisms of Cancer induction by tobacco-specific NNK and NNN. Cancers, Basel, 6:1138–1156. , DOI 10.3390/cancers6021138 Sharan RN, Mehrotra R, Choudhury Y, Asotra K, 2012, Association of betel nut with carcinogenesis: revisit with a clinical perspective. PLoS One 7:e42759. , DOI 10.1371/journal. pone.0042759 Reed L, Arlt VM, Phillips DH, 2018, The role of cytochrome P450 enzymes in carcinogen activation and detoxication: an in vivo-in vitro paradox. Carcinogenesis 39:851–859. , DOI 10.1093/ carcin/bgy058 Shervington A, Mohammed K, Patel R, Lea R, 2007, Identification of a novel co-transcription of P450/1A1 with telomerase in A549. Gene 388:110–116. , DOI 10.1016/j.gene.2006.10.010 Lee H-T, Bose A, Lee C-Y, Opresko PL, Myong S, 2017, Molecular mechanisms by which oxidativeDNA damage promotes telomerase activity. Nucleic Acids Res 45:11752–11765. https:// doi.org/10.1093/nar/gkx789 Gao Y, Ling T, Yin X et al, 2007, Effects of arecoline and nicotine on the expression of hTERT in oral keratinocytes. Zhonghua Kou Qiang Yi Xue Za Zhi 42:26–30 Kanojia D, Vaidya MM, 2006, 4-Nitroquinoline-1-oxide induced experimental oral carcinogenesis. Oral Oncol 42:655–667. https:// doi.org/10.1016/j.oraloncology.2005.10.013 Sumida T, Hamakawa H, Sogawa K, Bao Y, Zen H, Sugita A, Nezu K, AbeY, TaniokaH, UedaN(1999, Telomerase activation and cell proliferation during 7,12-dimethylbenz[a]anthracene-induced hamster cheek pouch carcinogenesis. Mol Carcinog 25:164–168 Kim H, Christensen R, Park N et al, 2001, Elevated expression of hTERT is associated with dysplastic cell transformation during human oral carcinogenesis in situ. Clin Cancer Res 7:3079–3086 Raghunandan BN, Sanjai K, Kumaraswamy J et al, 2016, Expression of human telomerase reverse transcriptase protein in oral epithelial dysplasia and oral squamous cell carcinoma: An immunohistochemical study. J Oral Maxillofac Pathol 20:96–101. , DOI 10.4103/0973-029X.180953 Mutirangura A, Supiyaphun P, Trirekapan S et al, 1996, Telomerase activity in oral leukoplakia and head and neck squamous cell carcinoma. Cancer Res 56:3530–3533 Lin S-C, Lu S-Y, Lee S-Y, Lin CY, Chen CH, Chang KW, 2005, Areca, betel, nut extract activates mitogen-activated protein kinasesand NF-κB in oral keratinocytes. Int J Cancer 116:526– 535. , DOI 10.1002/ijc.21104 Srinivasan M, Jewell SD, 2001, Quantitative estimation of PCNA, c-myc, EGFR and TGF-alpha in oral submucous fibrosis–an immunohistochemical study. Oral Oncol 37:461–467 Armanios M, 2012, Telomerase and idiopathic pulmonary fibrosis. Mutat Res 730:52–58. , DOI 10.1016/j.mrfmmm.2011.10.013 Yanjia H, Xinchun J, 2007, The role of epithelial–mesenchymal transition in oral squamous cell carcinoma and oral submucous fibrosis. Clin Chim Acta 383:51–56. , DOI 10.1016/j.cca. 2007.04.014 Boscolo-Rizzo P, Da Mosto MC, Rampazzo E et al, 2016, Telomeres and telomerase in head and neck squamous cell carcinoma: from pathogenesis to clinical implications. Cancer Metastasis Rev 35:457–474. , DOI 10.1007/s10555-016-9633-1 Park Y-J, KimEK, Bae JY, Moon S, Kim J, 2016, Human telomerase reverse transcriptase, hTERT, promotes cancer invasion by modulating cathepsin D via early growth response, EGR)-1. Cancer Lett 370:222–231. , DOI 10.1016/j.canlet.2015.10.021 Lee BK, Diebel E, Neukam FWet al, 2001, Diagnostic and prognostic relevance of expression of human telomerase subunits in oral cancer. Int J Oncol 19:1063–1068 Boscolo-Rizzo P, Rampazzo E, Perissinotto E, Piano MA, Giunco S, Baboci L, Spinato G, Spinato R, Tirelli G, da Mosto MC, del Mistro A, de Rossi A, 2015, Telomere shortening in mucosa surrounding the tumor: biosensor of field cancerization and prognostic marker of mucosal failure in head and neck squamous cell carcinoma. Oral Oncol 51:500–507. , DOI 10.1016/j. oraloncology.2015.02.100 Dey A, Chakrabarti K, 2018, Current perspectives of telomerase structure and function in eukaryotes with emerging views on telomerase in human parasites. Int JMol Sci 19:333. , DOI 10. 3390/ijms19020333 Haendeler J, Hoffmann J, Brandes RP, Zeiher AM, Dimmeler S, 2003, Hydrogen peroxide triggers nuclear export of telomerase reverse transcriptase via Src kinase family-dependent phosphorylation of tyrosine 707. Mol Cell Biol 23:4598–4610. , DOI 10.1128/MCB.23.13.4598-4610.2003 Shakunthala GK, Annigeri RG, Arunkumar S, 2015, Role of oxidative stress in the pathogenesis of oral submucous fibrosis: A preliminary prospective study. Contemp Clin Dent 6:S172–S174. , DOI 10.4103/0976-237X.166823 Gupta S, ReddyMVR, Harinath BC, 2004, Role of oxidative stress and antioxidants in aetiopathogenesis and management of oral submucous fibrosis. Indian J Clin Biochem 19:138–141. , DOI 10.1007/BF02872409 Akiyama M, Hideshima T, Hayashi T, Tai YT, Mitsiades CS, Mitsiades N, Chauhan D, Richardson P, Munshi NC, Anderson KC, 2003, Nuclear factor-kappaB p65 mediates tumor necrosis factor alpha-induced nuclear translocation of telomerase reverse transcriptase protein. Cancer Res 63:18–21 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1573 EP 1582 DI 10.1007/s12253-019-00700-6 PG 10 ER PT J AU Liu, Q Zhang, X Tang, H Liu, J Fu, Ch Sun, M Zhao, L Wei, M Yu, Z Wang, P AF Liu, Qianqian Zhang, Xiling Tang, Haichao Liu, Jinwei Fu, Chen Sun, Mingli Zhao, Lin Wei, Minjie Yu, Zhaojin Wang, Ping TI Bioinformatics Analysis Suggests the Combined Expression of AURKB and KIF18B Being an Important Event in the Development of Clear Cell Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE AURKB; KIF18B; Clear cell renal cell carcinoma; Differentially expressed genes; Combined expression ID AURKB; KIF18B; Clear cell renal cell carcinoma; Differentially expressed genes; Combined expression AB Clear cell renal cell carcinoma (ccRCC) is the most common type of renal cell carcinoma with high metastatic rate and high mortality rate, needing to find potential therapeutic targets and develop new therapy methods. The bioinformatics analysis was used in this study to find the targets. Firstly, the expression profile of ccRCC obtained from The Cancer Genome Atlas (TCGA) database and GSE53757 dataset were used to identify the significant up-regulated genes. IL20RB, AURKB and KIF18B with the top efficiency of capable of diagnosis ccRCC from para cancer tissue, were over-expressed in ccRCC samples, and expressed increasedly with the development of ccRCC. There was the closest correlation between AURKB and KIF18B in these three overexpressed genes. AURKB (high) or KIF18B (high) were all significantly correlated with higher T, N, M stage, G grade and shorter overall survival (OS) of ccRCC patients. Furthermore, the ccRCC patients with AURKB (high) + KIF18B (high) showed worse clinical characteristics and prognosis. Multivariate COX regression analysis indicated AURKB (high) and KIF18B (high) were all the independent prognostic risk factor without considering the interaction of AURKB and KIF18B. Moreover, considering the combination of each other, only AURKB (high) + KIF18B (high) expression was an independent prognostic risk factor for ccRCC patients, but not other situations. Collectively, AURKB was closely associated with KIF18B, and the combined expression of AURKB and KIF18B may be of great significance in ccRCC. C1 [Liu, Qianqian] China Medical University, School of Pharmacy, Department of Pharmacology, 110122 Shenyang, Liaoning, China. [Zhang, Xiling] China Medical University, The Fourth Affiliated Hospital, Department of Urology, 110000 Shenyang, Liaoning, China. [Tang, Haichao] China Medical University, School of Pharmacy, Department of Pharmacology, 110122 Shenyang, Liaoning, China. [Liu, Jinwei] China Medical University, School of Pharmacy, Department of Pharmacology, 110122 Shenyang, Liaoning, China. [Fu, Chen] China Medical University, School of Pharmacy, Department of Pharmacology, 110122 Shenyang, Liaoning, China. [Sun, Mingli] China Medical University, School of Pharmacy, Department of Pharmacology, 110122 Shenyang, Liaoning, China. [Zhao, Lin] China Medical University, School of Pharmacy, Department of Pharmacology, 110122 Shenyang, Liaoning, China. [Wei, Minjie] China Medical University, School of Pharmacy, Department of Pharmacology, 110122 Shenyang, Liaoning, China. [Yu, Zhaojin] China Medical University, School of Pharmacy, Department of Pharmacology, 110122 Shenyang, Liaoning, China. [Wang, Ping] China Medical University, The Fourth Affiliated Hospital, Department of Urology, 110000 Shenyang, Liaoning, China. RP Yu, Z (reprint author), China Medical University, School of Pharmacy, Department of Pharmacology, 110122 Shenyang, China. EM yuzhaojincmu@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6):394–424 Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L et al, 2017, Renal cell carcinoma. Nat Rev Dis Primers 3(17009):9 Hsieh JJ, Le V, Cao D, Cheng EH, Creighton CJ, 2018, Genomic classifications of renal cell carcinoma: a critical step towards the future application of personalized kidney cancer care with panomics precision. J Pathol 244(5):525–537 Li QK, Pavlovich CP, Zhang H, Kinsinger CR, Chan DW, 2019, Challenges and opportunities in the proteomic characterization of clear cell renal cell carcinoma, ccRCC): a critical step towards the personalized care of renal cancers. Semin Cancer Biol 55:8–15 Capitanio U,Montorsi F, 2016, Renal cancer. Lancet 387:894–906 Keefe SM, Nathanson KL, Rathmell WK, 2013, The molecular biology of renal cell carcinoma. Semin Oncol 40(4):421–428 FanY, Ma X, Li H, GaoY, HuangQ, ZhangY, Bao X, duQ, Luo G, Liu K, Meng Q, Zhao C, Zhang X, 2018, miR-122 promotes metastasis of clear-cell renal cell carcinoma by downregulating dicer. Int J Cancer 142(3):547–560 Cancer Genome Atlas Research N, Weinstein JN, Collisson EA, Mills GB, Shaw KR et al, 2013, The cancer genome atlas Pan- Cancer analysis project. Nat Genet 45(10):1113–1120 Barrett T,Wilhite SE, Ledoux P, Evangelista C, Kim IF et al, 2013, NCBI GEO: archive for functional genomics datasets-update. Nucleic Acids Res 41:D991–D995 Wang Z, Yang B, Zhang M, Guo W, Wu Z et al, 2018, lncRNA epigenetic landscape analysis identifies EPIC1 as an oncogenic lncRNA that interacts with MYC and promotes cell-cycle progression in cancer. Cancer Cell 33(4):706–720 Li B,WangW, Li Z, Chen Z, Zhi X, Xu J, Li Q,Wang L, Huang X, Wang L, Wei S, Sun G, Zhang X, He Z, Zhang L, Zhang D, Xu H, el-Rifai W, Xu Z, 2017, MicroRNA-148a-3p enhances cisplatin cytotoxicity in gastric cancer through mitochondrial fission induction and cyto-protective autophagy suppression. Cancer Lett 410: 212–227 Chen L, Yuan L, Qian K, Qian G, Zhu Y,Wu CL, Dan HC, Xiao Y, Wang X, 2018, Identification of biomarkers associated with pathological stage and prognosis of clear cell renal cell carcinoma by coexpression network analysis. Front Physiol 9:399 Song J, Peng J, Zhu C, Bai G, Liu Y, Zhu J, Liu J, 2018, Identification and validation of two novel prognostic lncRNAs in kidney renal clear cell carcinoma. Cell Physiol Biochem 48(6): 2549–2562 Atkins MB, Tannir NM, 2018, Current and emerging therapies for first-line treatment of metastatic clear cell renal cell carcinoma. Cancer Treat Rev 70:127–137 Shuch B, Amin A, Armstrong AJ, Eble JN, Ficarra V, Lopez- Beltran A, Martignoni G, Rini BI, Kutikov A, 2015, Understanding pathologic variants of renal cell carcinoma: distilling therapeutic opportunities from biologic complexity. Eur Urol 67(1):85–97 Yan M, Wang C, He B, Yang M, Tong M et al, 2016, Aurora-a kinase: a potent oncogene and target for cancer therapy. Med Res Rev 36(6):1036–1079 Al-Khafaji AS, Davies MP, Risk JM, Marcus MW, Koffa M et al, 2017, Aurora B expression modulates paclitaxel response in nonsmall cell lung cancer. Br J Cancer 116(5):592–599 Enjoji M, Iida S, Sugita H, Ishikawa T, Uetake H et al, 2009, BubR1 and AURKB overexpression are associated with a favorable prognosis in gastric cancer. Mol Med Rep 2(4):589–596 Lin ZZ, Jeng YM, Hu FC, Pan HW, Tsao HW, Lai PL, Lee PH, Cheng AL, Hsu HC, 2010, Significance of Aurora B overexpression in hepatocellular carcinoma. Aurora B overexpression in HCC. BMC Cancer 10:461 ZhangY JC, Li H, Lv F, Li X et al, 2015, Elevated Aurora B expression contributes to chemoresistance and poor prognosis in breast cancer. Int J Clin Exp Pathol 8(1):751–757 Chen YJ, ChenCM, Twu NF, YenMS, Lai CR,Wu HH,Wang PH, Yuan CC, 2009, Overexpression of Aurora B is associated with poor prognosis in epithelial ovarian cancer patients. Virchows Arch 455(5):431–440 Lens SM, Voest EE, Medema RH, 2010, Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nat Rev Cancer 10(12):825–841 Tang A, Gao K, Chu L, Zhang R, Yang J et al, 2017, Aurora kinases: novel therapy targets in cancers. Oncotarget 8(14): 23937–23954 LokW, Klein RQ, SaifMW(2010, Aurora kinase inhibitors as anticancer therapy. Anti-Cancer Drugs 21(4):339–350 SehdevV, Peng D, Soutto M,WashingtonMK, Revetta F, Ecsedy J, Zaika A, Rau TT, Schneider-Stock R, Belkhiri A, el-RifaiW(2012, The aurora kinase A inhibitor MLN8237 enhances cisplatininduced cell death in esophageal adenocarcinoma cells. Mol Cancer Ther 11(3):763–774 Cheung CH, Sarvagalla S, Lee JY, Huang YC, Coumar MS, 2014, Aurora kinase inhibitor patents and agents in clinical testing: an update, 2011-2013). Expert Opin Ther Pat 24(9): 1021–1038 Schwartz GK, Carvajal RD, Midgley R, Rodig SJ, Stockman PK, Ataman O, Wilson D, Das S, Shapiro GI, 2013, Phase I study of barasertib, AZD1152), a selective inhibitor of Aurora B kinase, in patients with advanced solid tumors. Investig New Drugs 31(2): 370–380 Yu Y, Feng YM, 2010, The role of kinesin family proteins in tumorigenesis and progression: potential biomarkers and molecular targets for cancer therapy. Cancer 116(22):5150– 5160 Lee YM, Kim E, Park M, Moon E, Ahn SM, Kim W, Hwang KB, Kim YK, Choi W, Kim W, 2010, Cell cycle-regulated expression and subcellular localization of a kinesin-8 member human KIF18B. Gene 466(1–2):16–25 Wu Y, Wang A, Zhu B, Huang J, Lu E, 2018, KIF18B promotes tumor progression through activating theWnt/β-catenin pathway in cervical cancer. Onco Targets Ther 11:1707–1720 Itzel T, Scholz P, Maass T, Krupp M, Marquardt JU, Strand S, Becker D, Staib F, Binder H, Roessler S, Wang XW, Thorgeirsson S, Muller M, Galle PR, Teufel A, 2015, Translating bioinformatics in oncology: guilt-by-profiling analysis and identification of KIF18B and CDCA3 as novel driver genes in carcinogenesis. Bioinformatics 31(2):216–224 TanenbaumME,Macurek L, van der Vaart B, GalliM, Akhmanova A, Medema RH, 2011, A complex of Kif18b andMCAK promotes microtubule depolymerization and is negatively regulated by Aurora kinases. Curr Biol 21(16):1356–1365 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1583 EP 1594 DI 10.1007/s12253-019-00740-y PG 12 ER PT J AU Chai, YSz Peng, R Zhang, R Zhou, L Pillay, N Tay, HK Badrick, T Li, J Horan, PM AF Chai, Yee Sze Peng, Rongxue Zhang, Rui Zhou, Li Pillay, Nalishia Tay, Hong Kwang Badrick, Tony Li, Jinming Horan, P Martin TI External Quality Assurance of Current Technology for the Testing of Cancer-Associated Circulating Free DNA Variants SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Circulating free DNA (cfDNA); Next-generation sequencing; Digital PCR; External quality assurance; Liquid biopsy ID Circulating free DNA (cfDNA); Next-generation sequencing; Digital PCR; External quality assurance; Liquid biopsy AB Liquid biopsy testing is rapidly emerging as a diagnostic means of identifying circulating free DNA (cfDNA) disease-associated variants. However, the reporting of cfDNA variants remains inconsistent due in part to the application of multiple testing pipelines which raise uncertainty about current cfDNA detection efficiency. External quality assurance (EQA) programs are required to monitor, evaluate and help improve laboratory performance for cfDNA variant detection and in clinical interpretation. This study therefore evaluated the performance of diagnostic laboratories currently performing cfDNA testing in China, Australia and New Zealand. A total of 89 laboratories participated in this EQA program. Reference testing material comprised of cfDNA manufactured to contain six different genotypes in four different genes (EGFR, KRAS, BRAF, NRAS). The predicted genotypic variant allelic frequencies ranged between 0.5% - 2.5%. Proficiency testing used a z-score on the laboratory consensus allelic frequency data to compare laboratory performance for the detection of the different genotypes. Allelic frequency genotyping data were received from 88 of the 89 laboratories. Next generation sequencing and digital PCR testing platforms were primarily used by participants in this pilot EQA. The average consensus data for each cfDNA genotype identified allelic frequencies ranging between 0.39% - 4.4%. Z-score proficiency testing found that >92% of clinical laboratories were concordant for detecting the cfDNA variants. The data from this pilot study suggest that current cfDNA testing platforms can detect cfDNA allelic frequency variants from 0.39% and above with high levels of confidence. In addition, these data highlight the importance of laboratories enrolling on EQA programs so that proficiency in cfDNA diagnostic testing can be determined and potential sources of error identified and addressed. C1 [Chai, Yee Sze] St Leonard’s, RCPAQAP Molecular GeneticsSydney, Australia. [Peng, Rongxue] National Center of Gerontology, Beijing Hospital, National Center for Clinical LaboratoriesBeijing, China. [Zhang, Rui] National Center of Gerontology, Beijing Hospital, National Center for Clinical LaboratoriesBeijing, China. [Zhou, Li] National Center of Gerontology, Beijing Hospital, National Center for Clinical LaboratoriesBeijing, China. [Pillay, Nalishia] St Leonard’s, RCPAQAP Molecular GeneticsSydney, Australia. [Tay, Hong Kwang] St Leonard’s, RCPAQAP Molecular GeneticsSydney, Australia. [Badrick, Tony] St Leonard’s, RCPAQAP Molecular GeneticsSydney, Australia. [Li, Jinming] National Center of Gerontology, Beijing Hospital, National Center for Clinical LaboratoriesBeijing, China. [Horan, P Martin] St Leonard’s, RCPAQAP Molecular GeneticsSydney, Australia. RP Horan, PM (reprint author), St Leonard’s, RCPAQAP Molecular Genetics, Sydney, Australia. EM martin.horan@rcpaqap.com.au CR Kamat AA, Bischoff FZ, Dang D, Baldwin MF, Han LY, Lin YG, Merritt WM, Landen CN Jr, Lu C, Gershenson DM, Simpson JL, Sood AK, 2006, Circulating cell-free DNA: a novel biomarker for response to therapy in ovarian carcinoma. Cancer Biol Ther 5: 1369–1374 Warton K, Samimi G, 2015, Methylation of cell-free circulating DNA in the diagnosis of cancer. Front Mol Biosci 2:13 Shinozaki M, O'Day SJ, Kitago M, Amersi F, Kuo C, Kim J,Wang HJ, Hoon DS, 2007)Utility of circulating B-RAF DNAmutation in serum for monitoring melanoma patients receiving biochemotherapy. Clin Cancer Res 13:2068–2074 Sonnenblick A, Ponde N, Piccart M, 2016, Metastatic breast cancer: the odyssey of personalization. Mol Oncol 10:1147–1159 Tomasetti M, Amati M, Neuzil J, Santarelli L, 2017, Circulating epigenetic biomarkers in lung malignancies: from early diagnosis to therapy. Lung Cancer 107:65–72 Vendrell JA, Mau-Them FT, Beganton B, Godreuil S, Coopman P, Solassol J, 2017, Circulating cell free tumor DNA detection as a routine tool for lung cancer patient management. Int J Mol Sci. , DOI 10.3390/ijms18020264 Ishii H, Azuma K, Sakai K, Kawahara A, Yamada K, Tokito T, Okamoto I, Nishio K, Hoshino T, 2015, Digital PCR analysis of plasma cell-free DNA for non-invasive detection of drug resistance mechanisms in EGFR mutant NSCLC: correlation with paired tumor samples. Oncotarget 6:30850–30858 Momtaz P, Pentsova E, Abdel-Wahab O, Diamond E, Hyman D, Merghoub T, You D, Gasmi B, Viale A, Chapman PB, 2016, Quantification of tumor-derived cell free DNA(cfDNA, by digital PCR, DigPCR, in cerebrospinal fluid of patients with BRAFV600 mutated malignancies. Oncotarget 7:85430–85436 ZhangY, Xu Y, ZhongW, Zhao J, ChenM, Zhang L, Li L,WangM, 2017b, Total DNA input is a crucial determinant of the sensitivity of plasma cell-free DNA EGFR mutation detection using droplet digital PCR. Oncotarget. 8:5861–5873 Thierry AR, Pastor B, Jiang ZQ, Katsiampoura AD, Parseghian C, Loree JM, Overman MJ, Sanchez C, Messaoudi SE, Ychou M, Kopetz S, 2017, Circulating DNA demonstrates convergent evolution and common resistance mechanisms during treatment of colorectal cancer. Clin Cancer Res 23:4578–4591 Fan G, Zhang K, Ding J, Li J, 2017a, Prognostic value of EGFR and KRAS in circulating tumor DNA in patients with advanced non-small cell lung cancer: a systematic review and meta-analysis. Oncotarget 8:33922–33932 Fan G, Zhang K, Yang X, Ding J,Wang Z, Li J, 2017b, Prognostic value of circulating tumor DNA in patients with colon cancer: systematic review. PLoS One 12:e0171991 Basnet S, Zhang ZY, Liao WQ, Li SH, Li PS, Ge HY, 2016, The prognostic value of circulating cell-free DNA in colorectal Cancer: a meta-analysis. J Cancer 7:1105–1113 Normanno N,MaielloMR, Chicchinelli N, Iannaccone A, Esposito C, De Cecio R, D'alessio A, De Luca A, 2017, Targeting the EGFR T790M mutation in non-small-cell lung cancer. Expert Opin Ther Targets 21:159–165 Ma M, Zhu H, Zhang C, Sun X, Gao X, Chen G, 2015, "Liquid biopsy"-ctDNA detection with great potential and challenges. Ann Transl Med 3:235 Iwama E, Sakai K, Azuma K, Harada D, Nosaki K, Hotta K, Nishio M, Kurata T, Fukuhara T, Akamatsu H, Goto K, Shimose T, Kishimoto J, Nakanishi Y, Nishio K, Okamoto I, 2018, Exploration of resistance mechanisms for epidermal growth factor receptor-tyrosine kinase inhibitors based on plasma analysis by digital PCR and next-generation sequencing. Cancer Sci 109:3921– 3933 Bordi P, Del Re M, Danesi R, Tiseo M, 2015, Circulating DNA in diagnosis and monitoring EGFR gene mutations in advanced nonsmall cell lung cancer. Transl Lung Cancer Res 4:584–597 Del Re M, Tiseo M, Bordi P, D'Incecco A, Camerini A, Petrini I, LucchesiM, Inno A, Spada D, Vasile E, Citi V, Malpeli G, Testa E, Gori S, Falcone A, Amoroso D, Chella A, Cappuzzo F, Ardizzoni A, Scarpa A, Danesi R, 2017, Contribution of KRAS mutations and c.2369C>T, p.T790M, EGFR to acquired resistance to EGFRTKIs in EGFR mutant NSCLC: a study on circulating tumor DNA. Oncotarget 8:13611–13619 Diaz LA Jr, Bardelli A, 2014, Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol 32:579–586 Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A, 2013, Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 10:472–484 Esposito A, Criscitiello C, Trapani D, Curigliano G, 2017, The emerging role of "liquid biopsies," circulating tumor cells, and circulating cell-free tumor DNA in lung cancer diagnosis and identification of resistance mutations. Curr Oncol Rep. , DOI 10. 1007/s11912-017-0564-y Siravegna G, Marsoni S, Siena S, Bardelli A, 2017, Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol 14:531–548 Aggarwal C, Thompson JC, Black TA et al, 2018, Clinical implications of plasma-based genotyping with the delivery of personalized therapy in metastatic non–small cell lung cancer. JAMA Oncol. , DOI 10.1001/jamaoncol.2018.4305 Rolfo C, Castiglia M, Hong D, Alessandro R, Mertens I, Baggerman G, Zwaenepoel K, Gil-Bazo I, Passiglia F, Carreca AP, Taverna S, Vento R, Santini D, Peeters M, Russo A, Pauwels P, 2014, Liquid biopsies in lung cancer: the new ambrosia of researchers. Biochim Biophys Acta 1846:539–546 Haselmann V, Ahmad-Nejad P, Geilenkeuser WJ, Duda A, Gabor M, Eichner R, Patton S, Neumaier M, 2018, Results of the first external quality assessment scheme, EQA, for isolation and analysis of circulating tumour DNA, ctDNA). Clin Chem Lab Med 56: 220–228 Keppens C, Dequeker EMC, Patton SJ, Normanno N, Fenizia F, Butler R, Cheetham M, Fairley JA,Williams H, Hall JA, Schuuring E, Deans ZC, IQN Path ASBL, 2018, International pilot external quality assessment scheme for analysis and reporting of circulating tumour DNA. BMC Cancer 18:804 Torga G, Pienta KJ, 2018, Patient-paired sample congruence between 2 commercial liquid biopsy tests. JAMA Oncol 4:868–870 Kuderer NM, Burton KA, Blau S, Rose AL, Parker S, Lyman GH, Blau CA, 2017, Comparison of 2 commercially available nextgeneration sequencing platforms in oncology. JAMA Oncol 3: 996–998 Roy S, LaFramboiseWA, Nikiforov YE, Nikiforova MN, Routbort MJ, Pfeifer J, Nagarajan R, Carter AB, Pantanowitz L, 2016, Nextgeneration sequencing informatics: challenges and strategies for implementation in a clinical environment. Arch Pathol Lab Med 140:958–975 Samorodnitsky E, Jewell BM, Hagopian R, Miya J, Wing MR, Lyon E, Damodaran S, Bhatt D, Reeser JW, Datta J, Roychowdhury S, 2015, Evaluation of hybridization capture versus amplicon-basedmethods for whole-exome sequencing. HumMutat 36:903–914 Goodwin S, McPherson JD, McCombie WR, 2016, Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet 17:333–351 Deans ZC, Butler R, Cheetham M, Dequeker EMC, Fairley JA, Fenizia F, Hall JA, Keppens C, Normanno N, Schuuring E, Patton SJ, 2019, IQN path ASBL report from the first European cfDNA consensus meeting: expert opinion on the minimal requirements for clinical ctDNA testing. Virchows Arch. , DOI 10.1007/s00428-019-02571-3 Cook L, Starr K, Boonyaratanakornkit J, Hayden R, Caliendo AM, 2018, Does size matter? Comparison of extraction yield for different-sized DNA fragments by seven different routine and four new circulating cell-free extraction methods. J Clin Microbiol. , DOI 10.1128/JCM.01061-18 Lee RJ, Gremel G, Marshall A, Myers KA, Fisher N, Dunn JA, Dhomen N, Corrie PG,Middleton MR, Lorigan P, Marais R, 2018, Circulating tumor DNA predicts survival in patients with resected high-risk stage II/III melanoma. Ann Oncol 29:490–496 Zhang R, Peng R, Li Z, Gao P, Jia S, Yang X, Ding J, Han Y, Xie J, Li J, 2017a, Synthetic circulating cell-free DNA as quality control materials for somatic mutation detection in liquid biopsy for cancer. Clin Chem 63:1465–1475 Auliac JB, Bayle S,Vergnenegre A, Le Caer H, Falchero L, Gervais R, Doubre H, Vinas F, Marin B, Chouaid C, 2018, Patients with non-small-cell lung cancer harbouring a BRAF mutation: a multicentre study exploring clinical characteristics, management, and outcomes in a real-life setting: EXPLORE GFPC 02-14. Curr Oncol 25:e398–e402 Tian Y, Zhao J, Ren P,Wang B, Zhao C, Shi C,Wei B, Ma J, Guo Y, 2018, Different subtypes of EGFR exon19 mutation can affect prognosis of patients with non-small cell lung adenocarcinoma. PLoS One 13:e0201682 Byeon S, Kim Y, Lim SW, Cho JH, Park S, Lee J, Sun JM, Choi YL, Lee SH, Ahn JS, Park K, AhnMJ, 2019, Clinical outcomes of EGFR exon 20 insertion mutations in advanced non-small cell lung Cancer in Korea. Cancer Res Treat 51:623–631 Mehrotra M, Singh RR, Loghavi S, Duose DY, Barkoh BA, Behrens C, Patel KP, Routbort MJ, Kopetz S, Broaddus RR, Medeiros LJ, Wistuba II, Luthra R, 2017, Detection of somatic mutations in cell-free DNA in plasma and correlation with overall survival in patients with solid tumors. Oncotarget 9:10259–10271 Martignetti JA, Pandya D, Nagarsheth N, Chen Y, Camacho O, Tomita S, Brodman M, Ascher-Walsh C, Kolev V, Cohen S, Harkins T, Schadt E, Reva B, Sebra R, Dottino P, 2018, Detection of endometrial precancer by a targeted gynecologic cancer liquid biopsy. Cold Spring HarbMol Case Stud. , DOI 10.1101/mcs.a003269 Di Nunno V, Gatto L, SantoniM, Cimadamore A, Lopez-Beltran A, Cheng L, Scarpelli M, Montironi R, Massari F, 2018, Recent advances in liquid biopsy in patients with castration resistant prostate cancer. Front Oncol 8:397 Qi ZH, Xu HX, Zhang SR, Xu JZ, Li S, Gao HL, JinW,WangWQ, Wu CT, Ni QX, Yu XJ, Liu L, 2018, The significance of liquid biopsy in pancreatic Cancer. J Cancer 9:3417–3426 Balaji SA, Shanmugam A, Chougule A, Sridharan S, Prabhash K, Arya A, Chaubey A, Hariharan A, Kolekar P, Sen M, Ravichandran A, Katragadda S, Sankaran S, Bhargava S, Kulkarni P, Rao S, Sunkavalli C, Banavali S, Joshi A, Noronha V, Dutt A, Bahadur U, Hariharan R, Veeramachaneni V, Gupta V, 2018, Analysis of solid tumormutation profiles in liquid biopsy. CancerMed 7:5439– 5447 Guibert N, Tsukada H, Hwang DH, Chambers E, Cibas ES, Bale T, Supplee J, Ulrich B, Sholl LM, Paweletz CP, Oxnard GR, 2018, Liquid biopsy of fine-needle aspiration supernatant for lung cancer genotyping. Lung Cancer 122:72–75 Garcia J, Forestier J, Dusserre E, Wozny AS, Geiguer F, Merle P, Tissot C, Ferraro-Peyret C, Jones FS, Edelstein DL, Cheynet V, Bardel C, Vilchez G, Xu Z, Bringuier PP, Barritault M, Brengle- Pesce K, Guillet M, Chauvenet M, Manship B, Brevet M, Rodriguez-Lafrasse C, Hervieu V, Couraud S, Walter T, Payen L, 2018, Cross-platform comparison for the detection of RAS mutations in cfDNA, ddPCR Biorad detection assay, BEAMing assay, and NGS strategy). Oncotarget 9:21122–21131 Adalsteinsson VA, Ha G, Freeman SS, Choudhury et al, 2017, Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun 8:1324 Dominguez-Vigil IG, Moreno-Martinez AK, Wang JY, Roehrl MHA, Barrera-Saldana HA, 2017, The dawn of the liquid biopsy in the fight against cancer. Oncotarget 9:2912–2922 Rothwell DG, Ayub M, Cook N et al, 2019, Utility of ctDNA to support patient selection for early phase clinical trials: the TARGET study. Nat Med. , DOI 10.1038/s41591-019-0380-z Wang T,Wang D, Zhang Y, Zhang J, Sun X,Wu Y,Wang S, Zhang Y, Xu L, Kong Q, Gao Y,Wu Y, Liu F, Liu S, ZhangY, Lei T, Liu H, 2018, Dynasore-induced potent ubiquitylation of the exon 19 deletion mutant of epidermal growth factor receptor suppresses cell growth and migration in non-small cell lung cancer. Int J Biochem Cell Biol 105:1–12 van Veggel B, de Langen AJ, Hashemi SMS, Monkhorst K, Heideman DAM, Thunnissen E, Smit EF, 2018, Afatinib and cetuximab in four patients with EGFR exon 20 insertion-positive advanced NSCLC. J Thorac Oncol 13:1222–1226 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1595 EP 1603 DI 10.1007/s12253-019-00744-8 PG 9 ER PT J AU Ke, D Wang, Q Ke, Sh Zou, L Wang, Q AF Ke, Dong Wang, Qiushuang Ke, Shaobo Zou, Li Wang, Qi TI Long-Non Coding RNA SNHG16 Supports Colon Cancer Cell Growth by Modulating miR-302a-3p/AKT Axis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SNHG16; miR-302a-3p; AKT; Colon cancer; Cell growth ID SNHG16; miR-302a-3p; AKT; Colon cancer; Cell growth AB Small nucleolar RNA host gene 16 (SNHG16) is reported to be involved in the tumorigenesis of various kinds of tumors. SNHG16 expression was reported to be upregulated in colon cancer, however, the underlying mechanism of how SNHG16 affects the colon cancer development remains poorly elucidated. In our study, with the aim to identify the role of SNHG16 on colon cell proliferation, SNHG16 was overexpressed or knocked down in vitro, respectively. SNHG16 overexpression accelerated colon cancer cell growth, while cell growth ability was impaired in SNHG16 silencing cells. Furthermore, the starBase database predicted that miR-302a-3p was the target gene of SNHG16, which was supported by dual luciferase assay. The effect of promoting cell proliferation ability induced by SNHG16 overexpression could be partly reversed by co-transfection of miR- 302a-3p mimic. Application of the miRanda database indicated that AKT may be modulated by SNHG16, further evidenced by western blot and quantitative PCR assays. AKT overexpression could partly reverse the attenuated colon cancer cell growth caused by miR-302a-3p mimic transfection. Meanwhile, the combination of miR-302a-3p inhibitor and shAKT achieved the parallel result. In conclusion, our study revealed the SNHG16/miR-302a-3p/AKT axis might play a crucial role in colon cancer cell proliferation, thus participating in the process of colon cancer development. C1 [Ke, Dong] Renmin Hospital of Wuhan University, Department of Gastrointestinal Surgery, 430060 Wuhan, Hubei, China. [Wang, Qiushuang] Renmin Hospital of Wuhan University, Department of Gastrointestinal Surgery, 430060 Wuhan, Hubei, China. [Ke, Shaobo] Renmin Hospital of Wuhan University, Center of Oncology, 430060 Wuhan, Hubei, China. [Zou, Li] Renmin Hospital of Wuhan University, Department of Gastrointestinal Surgery, 430060 Wuhan, Hubei, China. [Wang, Qi] Renmin Hospital of Wuhan University, Department of Gastrointestinal Surgery, 430060 Wuhan, Hubei, China. RP Wang, Q (reprint author), Renmin Hospital of Wuhan University, Department of Gastrointestinal Surgery, 430060 Wuhan, China. EM qiw@whu.edu.cn CR Global Burden of Disease Cancer C, Fitzmaurice C, Allen C, Barber RM, Barregard L, Bhutta ZA, Brenner H, Dicker DJ, Chimed-Orchir O, Dandona R, Dandona L, Fleming T, Forouzanfar MH, Hancock J, Hay RJ, Hunter-Merrill R, Huynh C, Hosgood HD, Johnson CO, Jonas JB, Khubchandani J, Kumar GA, Kutz M, Lan Q, Larson HJ, Liang X, Lim SS, Lopez AD, MacIntyre MF, Marczak L, Marquez N, Mokdad AH, Pinho C, Pourmalek F, Salomon JA, Sanabria JR, Sandar L, Sartorius B, Schwartz SM, Shackelford KA, Shibuya K, Stanaway J, Steiner C, Sun J, Takahashi K, Vollset SE, Vos T, Wagner JA, Wang H, Westerman R, Zeeb H, Zoeckler L, Abd-Allah F, Ahmed MB, Alabed S, Alam NK, Aldhahri SF, Alem G, Alemayohu MA, Ali R, Al-Raddadi R, Amare A, Amoako Y, Artaman A, Asayesh H, Atnafu N, Awasthi A, Saleem HB, Barac A, Bedi N, Bensenor I, Berhane A, Bernabe E, Betsu B, Binagwaho A, Boneya D, Campos-Nonato I, Castaneda-Orjuela C, Catala-Lopez F, Chiang P, Chibueze C, Chitheer A, Choi J-Y, Cowie B, Damtew S, das Neves J, Dey S, Dharmaratne S, Dhillon P, Ding E, Driscoll T, Ekwueme D, Endries AY, Farvid M, Farzadfar F, Fernandes J, Fischer F, G/Hiwot TT, Gebru A, Gopalani S, Hailu A, Horino M, Horita N, Husseini A, Huybrechts I, Inoue M, Islami F, Jakovljevic M, James S, Javanbakht M, Jee SH, Kasaeian A, Kedir MS, Khader YS, Khang Y-H, Kim D, Leigh J, Linn S, Lunevicius R, HMA ER, Malekzadeh R, Malta DC, Marcenes W, Markos D, Melaku YA, Meles KG, Mendoza W, Mengiste DT, Meretoja TJ, Miller TR, Mohammad KA, Mohammadi A, Mohammed S, Moradi-Lakeh M, Nagel G, Nand D, Le Nguyen Q, Nolte S, Ogbo FA, Oladimeji KE, Oren E, Pa M, Park E-K, Pereira DM, Plass D, Qorbani M, Radfar A, Rafay A, Rahman M, Rana SM, Soreide K, Satpathy M, Sawhney M, Sepanlou SG, Shaikh MA, She J, Shiue I, Shore HR, Shrime MG, So S, Soneji S, Stathopoulou V, Stroumpoulis K, Sufiyan MB, Sykes BL, Tabares- Seisdedos R, Tadese F, Tedla BA, Tessema GA, Thakur JS, Tran BX, Ukwaja KN, Uzochukwu BSC, Vlassov VV, Weiderpass E, Wubshet Terefe M, Yebyo HG, Yimam HH, Yonemoto N, Younis MZ, Yu C, Zaidi Z, ZakiMES, Zenebe ZM, Murray CJL, Naghavi M, 2017, Global, regional, and National Cancer Incidence, mortality, years of life lost, years lived with disability, and disabilityadjusted life-years for 32 Cancer groups, 1990 to 2015: A systematic analysis for the global burden of disease study. JAMA Oncol 3(4):524–548. , DOI 10.1001/jamaoncol.2016.5688 Gellad ZF, Provenzale D, 2010, Colorectal cancer: national and international perspective on the burden of disease and public health impact. Gastroenterology 138(6):2177–2190. , DOI 10. 1053/j.gastro.2010.01.056 Hutter CM, Chang-Claude J, Slattery ML, Pflugeisen BM, Lin Y, Duggan D, Nan H, Lemire M, Rangrej J, Figueiredo JC, Jiao S, Harrison TA, Liu Y, Chen LS, Stelling DL, Warnick GS, Hoffmeister M, Kury S, Fuchs CS, Giovannucci E, Hazra A, Kraft P, Hunter DJ, Gallinger S, Zanke BW, Brenner H, Frank B, Ma J, Ulrich CM,White E, Newcomb PA, Kooperberg C, LaCroix AZ, Prentice RL, Jackson RD, Schoen RE, Chanock SJ, Berndt SI, Hayes RB, Caan BJ, Potter JD, Hsu L, Bezieau S, Chan AT, Hudson TJ, Peters U, 2012, Characterization of geneenvironment interactions for colorectal cancer susceptibility loci. Cancer Res 72(8):2036–2044. , DOI 10.1158/0008-5472. CAN-11-4067 Jeon J, Du M, Schoen RE, Hoffmeister M, Newcomb PA, Berndt SI, Caan B, Campbell PT, Chan AT, Chang-Claude J, Giles GG, Gong J, Harrison TA, Huyghe JR, Jacobs EJ, Li L, Lin Y, Le Marchand L, Potter JD, Qu C, Bien SA, Zubair N, Macinnis RJ, Buchanan DD, Hopper JL, Cao Y, Nishihara R, Rennert G, Slattery ML, Thomas DC, Woods MO, Prentice RL, Gruber SB, Zheng Y, Brenner H, Hayes RB, White E, Peters U, Hsu L, 2018, Determining risk of colorectal Cancer and starting age of screening based on lifestyle, environmental, and genetic factors. Gastroenterology 154(8):2152–2164.e2119. , DOI 10. 1053/j.gastro.2018.02.021 Wapinski O, Chang HY, 2011, Long noncoding RNAs and human disease. Trends Cell Biol 21(6):354–361. , DOI 10.1016/j. tcb.2011.04.001 CabiliMN, Trapnell C, Goff L, KoziolM, Tazon-Vega B, Regev A, Rinn JL, 2011, Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 25(18):1915–1927. , DOI 10.1101/gad. 17446611 Mattick JS, Rinn JL, 2015, Discovery and annotation of long noncoding RNAs. Nat Struct Mol Biol 22:5. , DOI 10.1038/ nsmb.2942 Schmitt AM, Chang HY, 2016, Long noncoding RNAs in Cancer pathways. Cancer Cell 29(4):452–463. , DOI 10.1016/j. ccell.2016.03.010 Khaitan D, Dinger ME, Mazar J, Crawford J, Smith MA, Mattick JS, Perera RJ, 2011, The melanoma-upregulated long noncoding RNA SPRY4-IT1 modulates apoptosis and invasion. Cancer Res 71(11):3852–3862. , DOI 10.1158/0008-5472.CAN-10- 4460 Liu B, Sun L, Liu Q, Gong C, Yao Y, Lv X, Lin L, Yao H, Su F, Li D, Zeng M, Song E, 2015, A cytoplasmic NF-κB interacting long noncoding RNA blocks IκB phosphorylation and suppresses breast Cancer metastasis. Cancer Cell 27(3):370–381. , DOI 10. 1016/j.ccell.2015.02.004 Zhao W, Li W, Dai W, Huang N, Qiu J, 2018, LINK-A promotes cell proliferation through the regulation of aerobic glycolysis in non-small-cell lung cancer. OncoTargets Ther 11:6071–6080. , DOI 10.2147/OTT.S171216 Ma M, Xu H, Liu G, Wu J, Li C, Wang X, Zhang S, Xu H, Ju S, Cheng W, Dai L, Wei Y, Tian Y, Fu X, 2019, MITA1, a novel energy stress-inducible lncRNA, promotes hepatocellular carcinoma metastasis. Hepatology. , DOI 10.1002/hep.30602 YuM, OhiraM, LiY, Niizuma H, OoML, ZhuY, Ozaki T, Isogai E, Nakamura Y, Koda T, Oba S, Yu B, Nakagawara A, 2009, High expression of ncRAN, a novel non-coding RNA mapped to chromosome 17q25.1, is associated with poor prognosis in neuroblastoma. Int J Oncol 34(4):931–938. , DOI 10.3892/ ijo_00000219 Zhu H, Zeng Y, Zhou C-C, Ye W, 2018, SNHG16/miR-216-5p/ ZEB1 signal pathway contributes to the tumorigenesis of cervical cancer cells. Arch Biochem Biophys 637:1–8. , DOI 10. 1016/j.abb.2017.11.003 Lu YF, Cai XL, Li ZZ, Lv J, Xiang Y, Chen JJ, ChenWJ, SunWY, Liu XM, Chen JB, 2018, LncRNA SNHG16 functions as an oncogene by sponging MiR-4518 and up-regulating PRMT5 expression in glioma. Cell Physiol Biochem 45(5):1975–1985. , DOI 10.1159/000487974 Cao X, Xu J, Yue D, 2018, LncRNA-SNHG16 predicts poor prognosis and promotes tumor proliferation through epigenetically silencing p21 in bladder cancer. Cancer Gene Ther 25(1):10–17. , DOI 10.1038/s41417-017-0006-x Zhao W, Fu H, Zhang S, Sun S, Liu Y, 2018, LncRNA SNHG16 drives proliferation, migration, and invasion of hemangioma endothelial cell through modulation of miR-520d-3p/STAT3 axis. Cancer Med 7(7):3311–3320. , DOI 10.1002/cam4.1562 Cai C, Huo Q,Wang X, Chen B, Yang Q, 2017, SNHG16 contributes to breast cancer cell migration by competitively binding miR- 98 with E2F5. Biochem Biophys Res Commun 485(2):272–278. , DOI 10.1016/j.bbrc.2017.02.094 Xu F, Zha G, Wu Y, Cai W, Ao J, 2018, Overexpressing lncRNA SNHG16 inhibited HCC proliferation and chemoresistance by functionally sponging hsa-miR-93. OncoTargets Ther 11:8855– 8863. , DOI 10.2147/OTT.S182005 Gibb EA, Warren RL, Wilson GW, Brown SD, Robertson GA, Morin GB, Holt RA, 2015, Activation of an endogenous retrovirus-associated long non-coding RNA in human adenocarcinoma. Genome Med 7(1):22–22. , DOI 10.1186/s13073- 015-0142-6 Christensen LL, True K, Hamilton MP, Nielsen MM, Damas ND, Damgaard CK, Ongen H, Dermitzakis E, Bramsen JB, Pedersen JS, Lund AH, Vang S, Stribolt K, Madsen MR, Laurberg S, McGuire SE, Orntoft TF, Andersen CL, 2016, SNHG16 is regulated by the Wnt pathway in colorectal cancer and affects genes involved in lipid metabolism. Mol Oncol 10(8):1266–1282. , DOI 10. 1016/j.molonc.2016.06.003 Wang C, Yao B, Xu M, Zheng X, 2016, RIP1 upregulation promoted tumor progression by activating AKT/Bcl-2/BAX signaling and predicted poor postsurgical prognosis in HCC. Tumor Biol 37(11):15305–15313. , DOI 10.1007/s13277-016-5342-1 C-c Z, Chen C, Z-q X, J-k Z, B-c O, Sun J, Zheng M-h, Y-p Z, A-g L, 2018, CCR6 promotes tumor angiogenesis via the AKT/NF-κB/ VEGF pathway in colorectal cancer. Biochim Biophys Acta, BBA, - Mol Basis Dis 1864(2):387–397. , DOI 10.1016/j.bbadis. 2017.10.033 Shao J, ZhuW, Ding Y, Zhu H, Jing X, Yu H, Lu M, Qiao Y,Wang X, Ai H, Wang X, 2019, Phosphorylation of LIFR promotes prostate cancer progression by activating the AKT pathway. Cancer Lett 451:110–121. , DOI 10.1016/j.canlet.2019.02.042 Ahmed D, Eide PW, Eilertsen IA, Danielsen SA, Eknaes M, Hektoen M, Lind GE, Lothe RA, 2013, Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis 2(9):e71 Jasperson KW, Tuohy TM, Neklason DW, Burt RW, 2010, Hereditary and familial Colon Cancer. Gastroenterology 138(6): 2044–2058. , DOI 10.1053/j.gastro.2010.01.054 Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A, Jemal A, 2017, Colorectal cancer statistics, 2017. CA Cancer J Clin 67(3):177–193. , DOI 10.3322/caac.21395 Song M, BodeAM, Dong Z, LeeM-H, 2019, AKTas a therapeutic target for Cancer. Cancer Res 79(6):1019. , DOI 10.1158/ 0008-5472.CAN-18-2738 Li Y, Yang Q, Guan H, Shi B, Ji M, Hou P, 2018, ZNF677 suppresses Akt phosphorylation and tumorigenesis in thyroid Cancer. Cancer Res 78(18):5216. , DOI 10.1158/0008-5472.CAN- 18-0003 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1605 EP 1613 DI 10.1007/s12253-019-00743-9 PG 9 ER PT J AU Yang, P Wang, Q Liu, A Zhu, J Feng, J AF Yang, Peipei Wang, Qian Liu, Aihua Zhu, Jun Feng, Jinzhou TI ALKBH5 Holds Prognostic Values and Inhibits the Metastasis of Colon Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ALKBH5; Colon cancer; Invasion; Metastasis ID ALKBH5; Colon cancer; Invasion; Metastasis AB N6-methyladenosine (m6A) demethylase ALKBH5 is best known for modulating transcript modification in plenty of human malignancies, but its role in the progression of colon cancer is not well understood. In the present study, we identified the tumor repressive role of ALKBH5 in colon cancer. ALKBH5 was downregulated in human colon cancer tissues, where its decreased expression significantly correlated with distant metastasis and American Joint Committee on Cancer (AJCC) stage. ALKBH5 was also an independent prognostic indicator of overall survival and disease-free survival in colon cancer patients. Furthermore, functional studies established that overexpression of ALKBH5 inhibited colon cancer cells invasion in vitro and metastasis in vivo. These results indicated that ALKBH5 significantly inhibits tumor progression and serves as a potential therapeutic target for colon cancer. C1 [Yang, Peipei] The First Affiliated Hospital of Anhui University of Science and Technology, Department of General Surgery, No. 203, Huaibin road, 232000 Huainan, Anhui Province, China. [Wang, Qian] Shandong First Medical University & Shangdong Academy of Medical Sciences, Taian City Central Hospital, Department of Central Laboratory, 29 Longtan Road, Taishan District, 271000 Taian, Shandong Province, China. [Liu, Aihua] Shandong First Medical University & Shangdong Academy of Medical Sciences, Taian City Central Hospital, Department of Central Laboratory, 29 Longtan Road, Taishan District, 271000 Taian, Shandong Province, China. [Zhu, Jun] Hang Zhou Di’an Medical Laboratory, 329 Jinpeng Road, Xihu District, 310030 Hangzhou, Zhejiang Province, China. [Feng, Jinzhou] The First Affiliated Hospital of Chongqing Medical University, Department of Neurology, 1 Youyi Road, Yuzhong District, 400016 Chongqing, China. RP Feng, J (reprint author), The First Affiliated Hospital of Chongqing Medical University, Department of Neurology, 400016 Chongqing, China. EM fengjinzhou@hotmail.com CR Brenner H, KloorM, Pox CP, 2014, Colorectal cancer. Lancet 383: 1490–1502 Brody H, 2015, Colorectal cancer. Nature 521:S1 Naxerova K, Reiter JG, Brachtel E, Lennerz JK, van de Wetering M, Rowan A, Cai T, Clevers H, Swanton C, Nowak MA, Elledge SJ, Jain RK, 2017, Origins of lymphatic and distant metastases in human colorectal cancer. Science 357:55–60 Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR, 2012, Comprehensive analysis of mRNA methylation reveals enrichment in 3' UTRs and near stop codons. Cell 149:1635–1646 Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, Jia G, Yu M, Lu Z, Deng X, Dai Q, Chen W, He C, 2014, A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol 10:93–95 Barbieri I, Tzelepis K, Pandolfini L, Shi J, Millan-Zambrano G, Robson SC, Aspris D, Migliori V, Bannister AJ, Han N, de Braekeleer E, Ponstingl H, Hendrick A, Vakoc CR, Vassiliou GS, Kouzarides T, 2017, Promoter-boundMETTL3 maintains myeloid leukaemia by m(6)A-dependent translation control. Nature 552: 126–131 Zhao X, Yang Y, Sun BF, ShiY,Yang X, XiaoW, Hao YJ, PingXL, Chen YS, Wang WJ, Jin KX, Wang X, Huang CM, Fu Y, Ge XM, Song SH, Jeong HS, Yanagisawa H, Niu Y, Jia GF, Wu W, Tong WM, Okamoto A, He C, Danielsen JMR, Wang XJ, Yang YG, 2014, FTO-dependent demethylation of N6-methyladenosine regulates mRNA splicing and is required for adipogenesis. Cell Res 24:1403–1419 Dixit D, Xie Q, Rich JN, Zhao JC, 2017, Messenger RNA methylation regulates glioblastoma tumorigenesis. Cancer Cell 31: 474–475 Zhong X, Yu J, Frazier K et al, 2018, Circadian clock regulation of hepatic lipid metabolism by modulation of m(6)A mRNA methylation. Cell Rep 25(1816–1828):e1814 Zhao BS, He C, 2017, "gamete on" for m(6)A: YTHDF2 exerts essential functions in female fertility. Mol Cell 67:903–905 Geula S, Moshitch-Moshkovitz S, Dominissini D, Mansour AAF, Kol N, Salmon-Divon M, Hershkovitz V, Peer E, Mor N, Manor YS, Ben-Haim MS, Eyal E, Yunger S, Pinto Y, Jaitin DA, Viukov S, Rais Y, Krupalnik V, Chomsky E, Zerbib M, Maza I, Rechavi Y, Massarwa R, Hanna S, Amit I, Levanon EY, Amariglio N, Stern-Ginossar N, Novershtern N, Rechavi G, Hanna JH, 2015, Stem cells. m6A mRNA methylation facilitates resolution of naive pluripotency toward differentiation. Science 347:1002–1006 Li HB, Tong J, Zhu S, Batista PJ, Duffy EE, Zhao J, Bailis W, Cao G, Kroehling L, Chen Y, Wang G, Broughton JP, Chen YG, Kluger Y, Simon MD, Chang HY, Yin Z, Flavell RA, 2017, m(6)A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature 548:338–342 He Y, Hu H, Wang Y, Yuan H, Lu Z, Wu P, Liu D, Tian L, Yin J, Jiang K, Miao Y, 2018, ALKBH5 inhibits pancreatic Cancer motility by decreasing long non-coding RNA KCNK15-AS1 methylation. Cellular physiology and biochemistry : international journal of experimental Cell Physiol Biochem 48:838–846 Zhang S, Zhao BS, Zhou A et al, 2017, M(6)a demethylase ALKBH5maintains Tumorigenicity of glioblastoma stem-like cells by sustaining FOXM1 expression and cell proliferation program. Cancer Cell 31:591–606 e596 Zhang C, Samanta D, Lu H, Bullen JW, Zhang H, Chen I, He X, Semenza GL, 2016, Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m(6)A-demethylation of NANOG mRNA. Proc Natl Acad Sci U S A 113: E2047–E2056 Yue B, Cai D, Liu C, Fang C, Yan D, 2016, Linc00152 functions as a competing endogenous RNA to confer oxaliplatin resistance and holds prognostic values in colon cancer. Mol Ther : the journal of the American Society of Gene Therapy 24:2064–2077 Yue B, Liu C, Sun H, Liu M, Song C, Cui R, Qiu S, Zhong M, 2018, A positive feed-forward loop between LncRNACYTOR and Wnt/beta-catenin signaling promotes metastasis of colon cancer. Mol Ther: the journal of the American Society of Gene Therapy 26:1287–1298 Fan J, Yan D, Teng M, Tang H, Zhou C, Wang X, Li D, Qiu G, Peng Z, 2011, Digital transcript profile analysis with aRNA-LongSAGE validates FERMT1 as a potential novel prognostic marker for colon cancer. Clin Cancer Res: an official journal of the American Association for Cancer Research 17:2908–2918 Jiang H, Deng R, Yang X, Shang J, Lu S, Zhao Y, Song K, Liu X, Zhang Q, Chen Y, Chinn YE, Wu G, Li J, Chen G, Yu J, Zhang J, 2017, Peptidomimetic inhibitors of APC-Asef interaction block colorectal cancer migration. Nat Chem Biol 13:994–1001 Buhrmann C, Shayan P, Goel A, Shakibaei M, 2017, Resveratrol regulates colorectal Cancer cell invasion by modulation of focal adhesion molecules. Nutrients 9 Overman MJ, Mcdermott R, Leach JL et al, 2017, Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer, CheckMate 142): an open-label,multicentre, phase 2 study. Lancet Oncol 18:1182–1191 Li Y, Du Y, Liang X et al, 2018, EGFR-targeted liposomal nanohybrid cerasomes: theranostic function and immune checkpoint inhibition in a mouse model of colorectal cancer. Nanoscale 10:16738–16749 Izumi D, Ishimoto T, Miyake K, Eto T, Arima K, Kiyozumi Y, Uchihara T, Kurashige J, Iwatsuki M, Baba Y, Sakamoto Y, Miyamoto Y, Yoshida N, Watanabe M, Goel A, Tan P, Baba H, 2017, Colorectal Cancer stem cells acquire Chemoresistance through the upregulation of F-box/WD repeat-containing protein 7 and the consequent degradation of c-Myc. Stem Cells 35:2027–2036 Lian H, Wang QH, Zhu CB, Ma J, Jin WL, 2018, Deciphering the Epitranscriptome in Cancer. Trends in Cancer 4:207–221 Guo J, Wu Y, Du J et al, 2018, Deregulation of UBE2C-mediated autophagy repression aggravates NSCLC progression. Oncogenesis 7:49 Zhang C, Zhi WI, Lu H, Samanta D, Chen I, Gabrielson E, Semenza GL, 2016, Hypoxia-inducible factors regulate pluripotency factor expression by ZNF217- and ALKBH5- mediated modulation of RNA methylation in breast cancer cells. Oncotarget 7:64527–64542 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1615 EP 1623 DI 10.1007/s12253-019-00737-7 PG 9 ER PT J AU Spachmann, JP Azzolina, V Weber, F Evert, M Eckstein, M Denzinger, S Burger, M Otto, W Breyer, J AF Spachmann, J Philipp Azzolina, Vanessa Weber, Florian Evert, Matthias Eckstein, Markus Denzinger, Stefan Burger, Maximilian Otto, Wolfgang Breyer, Johannes TI Loss of CHEK2 Predicts Progression in Stage pT1 Non-Muscle-Invasive Bladder Cancer (NMIBC) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-muscle-invasive bladder cancer; NMIBC; CHEK2; Smoking status; Progression ID Non-muscle-invasive bladder cancer; NMIBC; CHEK2; Smoking status; Progression AB Downregulation of checkpoint protein kinase 2 (CHEK2), which is involved in DNA repair, is associated with poorer outcome in various tumors. Little is known about the role of CHEK2 in urothelial carcinoma of the bladder (UCB). In the present study, we investigated the prognostic impact of CHEK2 protein expression in stage pT1 UCB. This retrospective, single-center analysis was carried out in a cohort of patients initially diagnosed with a pT1 UCB between 2007 and 2015. Immunohistochemical (IHC) staining of CHEK2 was performed. CHEK2 expression was correlated with recurrence-free survival (RFS), progression-free survival (PFS), and cancer-specific survival (CSS) using Kaplan-Meier analysis and multivariable Cox regression analysis. The analysis included 126 patients (86%male, median age 71 years). Loss of immunohistochemical protein expression of CHEK2 (<10%) was associated with significantly worse PFS (p = 0.041). Likewise, CHEK2 loss identified a subgroup of patients with worse PFS in the high-risk groups with concomitant CIS (p = 0.044), multifocal tumors (p < 0.001) and tumor grading G3 according to WHO1973 (p = 0.009). Multivariable Cox regression analysis revealed both loss of CHEK2 expression (HR: 4.18, 95%-CI: 1.35–12.93; p = 0.013) and multifocal tumors (HR: 4.53, 95%-CI:1.29–15.92; p = 0.018) as the only predictive factors for progression. Loss of IHC expression of CHEK2 in pT1 UCB is an independent predictor for progression to muscle-invasive disease and is also associated with worse PFS. This could help to identify high-risk patients who would benefit from early cystectomy. C1 [Spachmann, J Philipp] University of Regensburg, Caritas St. Josef Medical Center, Department of Urology, Landshuter Str. 65, 93053 Regensburg, Germany. [Azzolina, Vanessa] University of Regensburg, Caritas St. Josef Medical Center, Department of Urology, Landshuter Str. 65, 93053 Regensburg, Germany. [Weber, Florian] University of Regensburg, Institute of PathologyRegensburg, Germany. [Evert, Matthias] University of Regensburg, Institute of PathologyRegensburg, Germany. [Eckstein, Markus] Friedrich-Alexander-Universitat Erlangen-Nurnberg, University Hospital Erlangen, Institute of PathologyErlangen, Germany. [Denzinger, Stefan] University of Regensburg, Caritas St. Josef Medical Center, Department of Urology, Landshuter Str. 65, 93053 Regensburg, Germany. [Burger, Maximilian] University of Regensburg, Caritas St. Josef Medical Center, Department of Urology, Landshuter Str. 65, 93053 Regensburg, Germany. [Otto, Wolfgang] University of Regensburg, Caritas St. Josef Medical Center, Department of Urology, Landshuter Str. 65, 93053 Regensburg, Germany. [Breyer, Johannes] University of Regensburg, Caritas St. Josef Medical Center, Department of Urology, Landshuter Str. 65, 93053 Regensburg, Germany. RP Spachmann, JP (reprint author), University of Regensburg, Caritas St. Josef Medical Center, Department of Urology, 93053 Regensburg, Germany. EM philipp.spachmann@ukr.de CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6):394–424 Babjuk M, Bohle A, Burger M, Capoun O, Cohen D, Comperat EM, Hernandez V, Kaasinen E, Palou J, RoupretM, van Rhijn BW, Shariat SF, Soukup V, Sylvester RJ, Zigeuner R, 2017, EAU guidelines on non-muscle-invasive urothelial carcinoma of the bladder: update 2016. Eur Urol 71(3):447–461 Sylvester RJ, van der Meijden AP, Oosterlinck W, Witjes JA, Bouffioux C, Denis L, Newling DW, Kurth K, 2006, Predicting recurrence and progression in individual patients with stage ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials. Eur Urol 49:466–475 discussion 475–477 Denzinger S, Fritsche HM, Otto W, Blana A, Wieland WF, Burger M, 2008, Early versus deferred cystectomy for initial high-risk pT1G3 urothelial carcinoma of the bladder: do risk factors define feasibility of bladdersparing approach? Eur Urol 53:146–152 Heeke AL, Pishvaian MJ, Lynce F, Xiu J, Brody JR, Chen WJ, Baker TM, Marshall JL, Isaacs C, 2018, Prevalence of homologous recombination-related gene mutations across multiple Cancer types. JCO Precis Oncol 2018:1–13. , DOI 10.1200/PO. 17.00286. Epub 2018 Jul 23 Bartek J, Lukas J, 2003, Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell 3(5):421–429 Hirao A, Kong YY, Matsuoka S, Wakeham A, Ruland J, Yoshida H, Liu D, Elledge SJ, Mak TW, 2000, DNA damage-induced activation of p53 by the checkpoint kinase Chk2. Science 287(5459): 1824–1827 Suchy J, Cybulski C, Wokolorczyk D, Oszurek O, Gorski B, Debniak T, Jakubowska A, Gronwald J, Huzarski T, Byrski T, Dziuba I, Gogacz M, Wisniowski R, Wandzel P, Banaszkiewicz Z, Kurzawski G, Kladny J, Narod SA, Lubinski J, 2010, CHEK2 mutations and HNPCC-related colorectal cancer. Int J Cancer 126(12):3005–3009. , DOI 10.1002/ijc.25003 Havranek O, Spacek M, Hubacek P, Mocikova H, Markova J, Trneny M, Kleibl Z, 2011, Alterations of CHEK2 forkheadassociated domain increase the risk of Hodgkin lymphoma. Neoplasma 58(5):392–395 Havranek O, Kleiblova P, Hojny J, Lhota F, Soucek P, Trneny M, Kleibl Z, 2015, Association of Germline CHEK2 gene variants with risk and prognosis of non-Hodgkin lymphoma. PLoS One 10(10):e0140819. , DOI 10.1371/journal.pone. 0140819eCollection 2015 Wang Y, Dai B, Ye D, 2015, CHEK2 mutation and risk of prostate cancer: a systematic review and meta-analysis. Int J Clin Exp Med 8(9):15708–15715 eCollection 2015 Zlowocka E, Cybulski C, Gorski B, Debniak T, Slojewski M, Wokolorczyk D, Serrano-Fernandez P, Matyjasik J, van de Wetering T, Sikorski A, Scott RJ, Lubinski J, 2008, Germline mutations in the CHEK2 kinase gene are associated with an increased risk of bladder cancer. Int J Cancer 122(3):583–586 Lee HE, Han N, Kim MA, Lee HS, Yang HK, Lee BL, Kim WH, 2014, DNA damage response-related proteins in gastric cancer: ATM, Chk2 and p53 expression and their prognostic value. Pathobiology 81(1):25–35. , DOI 10.1159/000351072 Epub 2013 Aug 21 Ow GS, Ivshina AV, Fuentes G, Kuznetsov VA, 2014, Identification of two poorly prognosed ovarian carcinoma subtypes associated with CHEK2 germ-line mutation and non-CHEK2 somatic mutation gene signatures. Cell Cycle 13(14):2262–2280. , DOI 10.4161/cc.29271 Epub 2014 May 30 Slojewski M, Zlowocka E, Cybulski C, Gorski B, Debniak T, Wokolorczyk D, Matyjasik J, Sikorski A, Lubinski J, 2008, CHEK2 germline mutations correlate with recurrence rate in patients with superficial bladder cancer. Ann Acad Med Stetin 54(3): 115–121 Shahin O, Thalmann GN, Rentsch C, Mazzucchelli L, Studer UE, 2003, A retrospective analysis of 153 patients treated with or without intravesical bacillus Calmette-Guerin for primary stage T1 grade 3 bladder cancer: recurrence, progression and survival. J Urol 169(1):96–100 Aziz A, May M, Burger M, Palisaar RJ, Trinh QD, Fritsche HM, Rink M, Chun F, Martini T, Bolenz C, Mayr R, Pycha A, Nuhn P, Stief C, Novotny V,Wirth M, Seitz C, Noldus J, Gilfrich C, Shariat SF, Brookman-May S, Bastian PJ, Denzinger S, Gierth M, Roghmann F, 2014, PROMETRICS 2011 research group. Prediction of 90-day mortality after radical cystectomy for bladder cancer in a prospective European multicenter cohort. Eur Urol 66(1):156–163 GeY,WangY, ShaoW, Jin J,Du M, Ma G, Chu H,Wang M, Zhang Z, 2016, Rare variants in BRCA2 and CHEK2 are associated with the risk of urinary tract cancers. Sci Rep 6:33542. , DOI 10. 1038/srep33542 Muranen TA, Blomqvist C, Dork T, Jakubowska A, Heikkila P, Fagerholm R, Greco D, Aittomaki K, Bojesen SE, Shah M, Dunning AM, Rhenius V, Hall P, Czene K, Brand JS, Darabi H, Chang-Claude J, Rudolph A, Nordestgaard BG, Couch FJ, Hart SN, Figueroa J, Garcia-Closas M, Fasching PA, Beckmann MW, Li J, Liu J, Andrulis IL, Winqvist R, Pylkas K, Mannermaa A, Kataja V, Lindblom A, Margolin S, Lubinski J, Dubrowinskaja N, Bolla MK, Dennis J, Michailidou K,Wang Q, Easton DF, Pharoah PD, Schmidt MK, Nevanlinna H, 2016, Patient survival and tumor characteristics associated with CHEK2:p.I157T - findings from the Breast Cancer Association Consortium. Breast Cancer Res 18(1):98 Kilpivaara O, Bartkova J, Eerola H, Syrjakoski K, Vahteristo P, Lukas J, Blomqvist C, Holli K, Heikkila P, Sauter G, Kallioniemi OP, Bartek J, Nevanlinna H, 2005, Correlation of CHEK2 protein expression and c.1100delC mutation status with tumor characteristics among unselected breast cancer patients. Int J Cancer 113(4): 575–580 de Bock GH, Schutte M, Krol-Warmerdam EM, Seynaeve C, Blom J, Brekelmans CT,Meijers-Heijboer H, van Asperen CJ, Cornelisse CJ, Devilee P, Tollenaar RA, Klijn JG, 2004, Tumour characteristics and prognosis of breast cancer patients carrying the germline CHEK2*1100delC variant. J Med Genet 41(10):731–735 Ribeiro-Silva A, Moutinho MA, Moura HB, Vale FR, Zucoloto S, 2006, Expression of checkpoint kinase 2 in breast carcinomas: correlation with key regulators of tumor cell proliferation, angiogenesis, and survival. Histol Histopathol 21(4):373–382. https:// doi.org/10.14670/HH-21.373 Kriege M, Hollestelle A, Jager A, Huijts PE, Berns EM, Sieuwerts AM, Meijer-van Gelder ME, Collee JM, Devilee P, Hooning MJ, Martens JW, Seynaeve C, 2014, Survival and contralateral breast cancer in CHEK2 1100delC breast cancer patients: impact of adjuvant chemotherapy. Br J Cancer 111(5):1004–1013. , DOI 10.1038/bjc.2014.306 Epub 2014 Jun 10 Teodorczyk U, Cybulski C, Wokolorczyk D, Jakubowska A, Starzynska T, Lawniczak M, Domagala P, Ferenc K, Marlicz K, Banaszkiewicz Z,Wisniowski R, Narod SA, Lubinski J, 2013, The risk of gastric cancer in carriers of CHEK2 mutations. Familial Cancer 12(3):473–478. , DOI 10.1007/s10689-012-9599- 2 Ghelli Luserna Di Rora A, Iacobucci I, Imbrogno E, Papayannidis C, Derenzini E, Ferrari A, Guadagnuolo V, Robustelli V, Parisi S, Sartor C, Abbenante MC, Paolini S, Martinelli G, 2016, Prexasertib, a Chk1/Chk2 inhibitor, increases the effectiveness of conventional therapy in B−/T- cell progenitor acute lymphoblastic leukemia. Oncotarget 7(33):53377–53391. , DOI 10. 18632/oncotarget.10535 Zeng L, Beggs RR, Cooper TS, Weaver AN, Yang ES, 2017, Combining Chk1/2 inhibition with Cetuximab and radiation enhances In Vitro and In VivoCytotoxicity in head and neck squamous cell carcinoma.Mol Cancer Ther 16(4):591–600. , DOI 10. 1158/1535-7163.MCT-16-0352 Epub 2017 Jan 30 IsonoM, HoffmannMJ, PinkerneilM, Sato A,Michaelis M, Cinatl J Jr, Niegisch G, Schulz WA, 2017, Checkpoint kinase inhibitor AZD7762 strongly sensitises urothelial carcinoma cells to gemcitabine. J Exp Clin Cancer Res 36(1):1. , DOI 10. 1186/s13046-016-0473-1 Zhao H, Albino AP, Jorgensen E, Traganos F, Darzynkiewicz Z, 2009, DNAdamage response induced by tobacco smoke in normal human bronchial epithelial and A549 pulmonary adenocarcinoma cells assessed by laser scanning cytometry. Cytometry A 75(10): 840–847. , DOI 10.1002/cyto.a.20778 Breyer J, Denzinger S, Hartmann A, Otto W, 2016, Downregulation of checkpoint protein kinase 2 in the Urothelium of healthy male tobacco smokers. Urol Int 97(4):480–481 Epub 2016 Jun 2 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1625 EP 1632 DI 10.1007/s12253-019-00745-7 PG 8 ER PT J AU Pal, I Szilagyi, B Nagy, B Pal, T Hodosi, K Illes, Varoczy, L AF Pal, Ildiko Szilagyi, Bernadett Nagy, Bela Pal, Tibor Hodosi, Katalin Illes, Arpad Varoczy, Laszlo TI The Impact of Beta-Catenin and glutathione-S-transferase Gene Polymorphisms on the Treatment Results and Survival of Multiple Myeloma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Multiple myeloma; Cereblon; ß-catenin; Glutathione-S-transferase; Polymorphism; Personalized treatment ID Multiple myeloma; Cereblon; ß-catenin; Glutathione-S-transferase; Polymorphism; Personalized treatment AB Multiple myeloma (MM) is an incurable disease, however, novel therapeutic agents has significantly improved its prognosis. In this study we analyzed if polymorphisms in the genes of β-catenin and glutathione-S-transferase have affected the clinical course, treatment response and progression-free survival (PFS) of MM patients. Ninety-seven MM patients were involved who were administered immunomodulatory drug (Imid) or alkylating agent-based therapy. β-catenin (CTNNB1, rs4135385 A > G, rs4533622 A > C) and glutathione-S-transferase (GSTP1 105, GSTP1 114) gene polymorphisms were analyzed by Light SNiP assays. The distribution of CTNNB1 (rs4135385) AA, AG and GG genotypes were 48.4%, 47.4% and 4,1%, respectively. Patients with AA genotype were older than those who carried G allele (64.5 vs. 61.0 years of age, p < 0.05). Response to Imidbased therapies (p < 0.05) and PFS (p = 0.032) were significantly more favourable in the AA homozygous group. The other polymorphism(rs4533622) of β-catenin gene did not markedly influence these clinical parameters, althoughMMwas diagnosed at significantly younger age in subjects with CC genotype compared to AG/AA combined genotypes (59.1 vs. 65.7 years, p = 0.015). When GSTP1 polymorphisms were investigated, no such significant associations were observed. Our results demonstrate that the polymorphism of β-catenin gene (rs4135385) may be an independent predictive factor in MM. C1 [Pal, Ildiko] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Szilagyi, Bernadett] University of Debrecen, Faculty of Medicine, Department of Laboratory MedicineDebrecen, Hungary. [Nagy, Bela] University of Debrecen, Faculty of Medicine, Department of Laboratory MedicineDebrecen, Hungary. [Pal, Tibor] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Hodosi, Katalin] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Varoczy, Laszlo] University of Debrecen, Faculty of Medicine, Department of Hematology, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. RP Varoczy, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, H-4032 Debrecen, Hungary. EM varoczy@internal.med.unideb.hu CR Jaak Ph J, Klaus S, Andreas Vet al, 2018, Preventive, predictive, and personalized medicine for effective and affordable cancer care. EPMA Journal 9:113–123 Raza S, Safyan RA, Rosenbaum E, Bowman AS, Lentzsch S, 2017, Optimizing current and emerging therapies in multiple myeloma: a guide for the haematologist. Ther Adv Hematol 8:55–70 van Hengel J, Nollet F, Berx G, van Roy N, Speleman F, van Roy F, 1995, Assignment of the human beta-catenin gene, CTNNB1, to 3p22-p21.3 by fluorescence in situ hybridization. Cytogenet Cell Genet 70:68–70 Nollet F, Berx G, Molemans F, van Roy F, 1996, Genomic organization of the human beta-catenin gene, CTNNB1). Genomics. 32: 413–424 Lin Y, Huang X, He X et al, 2015, A novel effect of thalidomide and its analogs: supression of cereblon ubiquitination enhances ubiquitin ligase function. FASEB J 9:4829–4839 Chang XB, Stewart AK, 2011, What is the functional role of thalidomid binding protein cereblon? Int J Biochem Mol Biol 2: 287–294 Qiang YW, Rudikoff S, 2009)Wnt signaling pathways in multiple myeloma. In: Georgiev M, Bachev E, eds, Multiple myeloma: symptoms, diagnosis and treatment. Nova Science Publisher, New York, pp 51–75 Tian J, He H, Lei G, 2014)Wnt/ß-catenin pathway in bone cancers. Tumour Biol 35:9439–9445 Bjorklund C, Ma W, Wang ZQ et al, 2011, Evidence of a role for activation of Wnt/ß-catenin signaling in the resistance of plasma cells to lenalidomide. J Biol Chem 286:11009–11020 Butrym A, Rybka J, Lacina P et al, 2015, Polymorphisms within beta-catenin encoding gene affect multiple myeloma development and treatment. Leuk Res 39:1462–1466 Huang SY, LinCW, Lin HH, Yao M, Tang JL,Wu SJ, Chen YC, Lu HY, Hou HA, Chen CY, Chou WC, Tsay W, Chou SJ, Tien HF, 2014, Expression of cereblon protein assessed by immunohistochemical staining in myeloma cells is associated with superior response of thalidomide- and lenalidomide-based treatment, but not bortezomib-based treatment, in patients with multiple myeloma. Ann Hematol 93:1371–1380 Srivastava SK, Watkins SC, Schuetz E, Singh SV, 2002, Role of glutathione conjugate efflux in cellular protection against benzo, a, pyrene-7,8-diol-9,10-epoxide-induced DNA damage. Mol Carcinog 33:156–162 Taningher M, Malacarne D, Izzotti A, Ugolini D, Parodi S, 1999, Drug metabolism polymorphisms as modulators of cancer susceptibility. Mutat Res 436:227–261 Johansson AS, Stenberg G, Widersten M, Mannervik B, 1998, Structure-activity relationships and thermal stability of human glutathione transferase P1-1 governed by the H-site residue 105. J Mol Biol 278:687–698 Dasgupta RK, Adamson PJ, Davies FE et al, 2003, Polymorphic variation in GSTP1 modulates outcome following therapy for multiple myeloma. Blood. 102:2345–2350 Chang X, Xu Q, Hou Yet al, 2017, Mouse monoclonal antibodies generated from full length human cereblon: detection of cereblon protein in patients with multiple myeloma. Int J Mol Sci 18: 1999, , DOI 10.3390/ijms18091999. Gandhi AK, Mendy D,Waldman M, Chen G, Rychak E, Miller K, Gaidarova S, Ren Y,WangM, Breider M, Carmel G,Mahmoudi A, Jackson P, Abbasian M, Cathers BE, Schafer PH, Daniel TO, Lopez-Girona A, Thakurta A, Chopra R, 2014, Measuring cereblon as a biomarker of response or resistance to lenalidomide and pomalidomide requires use of standardized reagents and understanding of gene complexity. Br J Haematol 164:233–244 Wang S, Tian Y, Wu D, Zhu H, Luo D, Gong W, Zhou Y, Zhou J, Zhang Z, 2012, Genetic variation of CTNNB1 gene is associated with susceptibility and prognosis of gastric cancer in a Chinese population. Mutagenesis. 27:623–630 Alanazi MS, Parine NR, Shaik JP, Alabdulkarim HA, Ajaj SA, Khan Z, 2013, Association of single nucleotide polymorphisms inWnt signaling pathway genes with breast cancer in saudi patients. PLoS One 8:e59555 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1633 EP 1638 DI 10.1007/s12253-019-00747-5 PG 6 ER PT J AU Yao, Y Pang, T Cheng, Y Yong, W Kang, H Zhao, Y Wang, S Hu, X AF Yao, Yunhong Pang, Tianyun Cheng, Ying Yong, Weiwei Kang, Haixian Zhao, Yi Wang, Sen Hu, Xinrong TI Positive Correlative over-Expression between eIF4E and Snail in Nasopharyngeal Carcinoma Promotes its Metastasis and Resistance to Cisplatin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE eIF4E; Snail; NPC; EMT; Invasion; Metastasis ID eIF4E; Snail; NPC; EMT; Invasion; Metastasis AB EIF4E is the rate-limiting factor in the mRNA translation of specific set of oncogenes. Snail is the core transcription factor of epithelial-mesenchymal transition (EMT), a key step of cancer metastasis. The connection between the two oncoproteins has not been well established in the human cancer tissues and in nasopharyngeal carcinoma (NPC). Here we showed that the positive correlative over-expression was seen between eIF4E and Snail in NPC tissues, and the expression was significantly higher in the metastatic NPC than in the un-metastatic NPC. In NPC cells, eIF4E knockdown significantly reduced Snail mRNA and protein levels, increased the mRNA level of E-cad (a direct downstream gene of Snail and a negative EMT marker), attenuated the invasive ability of the cells, and sensitized the cells to cisplatin in invasion. In contrast, enforced the expression of eIF4E significantly increased Snail mRNA and protein levels, and promoted the invasive ability in NPC cells. Under the condition of the high eIF4E expression, Snail knockdown significantly increased E-cad mRNA level and weaken the invasive ability of NPC cells. Finally, eIF4E directly bound Snail mRNA for translation initiation displayed by the RIP assay. Therefore, the results firstly suggested that eIF4E enhanced the Snail expression in both transcription and translation manner in human cancer tissues and targeting the eIF4E/Snail axis might intervene with the EMT and metastasis of NPC. This finding provided a new clue for further understanding the metastatic mechanism of human cancers and for preventing and treating NPC metastasis. C1 [Yao, Yunhong] Guangdong Medical University, Cancer Research Institute, Department of Pathology, 523808 Dongguan, China. [Pang, Tianyun] Guangdong Medical University, Department of Obstetrics and Gynecology, 523808 Dongguan, China. [Cheng, Ying] Guangdong Medical University, Cancer Research Institute, Department of Pathology, 523808 Dongguan, China. [Yong, Weiwei] Guangdong Medical University, Cancer Research Institute, Department of Pathology, 523808 Dongguan, China. [Kang, Haixian] Guangdong Medical University, Cancer Research Institute, Department of Pathology, 523808 Dongguan, China. [Zhao, Yi] Guangdong Medical University, Cancer Research Institute, Department of Pathology, 523808 Dongguan, China. [Wang, Sen] Guangdong Medical University, Cancer Research Institute, Department of Pathology, 523808 Dongguan, China. [Hu, Xinrong] Guangdong Medical University, Cancer Research Institute, Department of Pathology, 523808 Dongguan, China. RP Hu, X (reprint author), Guangdong Medical University, Cancer Research Institute, Department of Pathology, 523808 Dongguan, China. EM huxinrong@163.com;404752528@qq.com CR Shair KHY, Reddy A, Cooper VS, 2018, New insights from elucidating the role of LMP1 in nasopharyngeal carcinoma. Cancers, Basel, 10(4) Zheng W, Zong J, Huang C, Chen J, Wu J, Chen C, Lin S, Pan J, 2016, Multimodality treatment may improve the survival rate of patients with metastatic nasopharyngeal carcinoma with good performance status. PLoS One 11(1):e0146771 Mittal V, 2018, Epithelial mesenchymal transition in tumor metastasis. Annu Rev Pathol 13:395–412 Zuo LL, Zhang J, Liu LZ, Zhou Q, Du SJ, Xin SY, Ning ZP, Yang J, Yu HB,YueWX,Wang J, Zhu FX, LiGY, Lu JH, 2017, Cadherin 6 is activated by Epstein-Barr virus LMP1 to mediate EMT and metastasis as an interplay node of multiple pathways in nasopharyngeal carcinoma. Oncogenesis 6(12):402 Brzozowa M, Michalski M, Wyrobiec G, Piecuch A, Dittfeld A, Harabin-Slowinska M, Boron D, Wojnicz R, 2015, The role of Snail1 transcription factor in colorectal cancer progression and metastasis. Contemp Oncol, Pozn, 19(4):265–270 Yu H, Shen Y, Hong J, Xia Q, Zhou F, Liu X, 2015, The contribution of TGF-β in epithelial-mesenchymal transition, EMT): downregulation of E-cadherin via snail. Neoplasma 62(1):1–15 Ren X, Yang X, Cheng B, Chen X, Zhang T, He Q, Li B, Li Y, Tang X, Wen X, Zhong Q, Kang T, Zeng M, Liu N, Ma J, 2017, HOPX hypermethylation promotes metastasis via activating snail transcription in nasopharyngeal carcinoma. Nat Commun 8:14053 Horikawa T, Yoshizaki T, Kondo S, Furukawa M, Kaizaki Y, Pagano JS, 2011, Epstein-Barr virus latent membrane protein 1 induces snail and epithelial-mesenchymal transition in metastatic nasopharyngeal carcinoma. Br J Cancer 104(7):1160–1167 Gingras AC, Raught B, Sonenberg N, 1999, EIF4 initiation factors: effectors of mRNA recruitment to ribosomes and regulators of translation. Annu Rev Biochem 68:913–963 Ernst BP, Mikstas C, Stover T, Stauber R, Strieth S, 2018, Association of eIF4E and SPARC expression with lymphangiogenesis and lymph node metastasis in hypopharyngeal cancer. Anticancer Res 38(2):699–706 Oblinger JL, Burns SS, Huang J, Pan L, Ren Y, Shen R, Kinghorn AD,Welling DB, Chang LS, 2018, Overexpression of eIF4F components in meningiomas and suppression of meningioma cell growth by inhibiting translation initiation. Exp Neurol 299(Pt B): 299–307 Liang S, Guo R, Zhang Z, Liu D, Xu H, Xu Z, Wang X, Yang L, 2013, Upregulation of the eIF4E signaling pathway contributes to the progression of gastric cancer, and targeting eIF4E by perifosine inhibits cell growth. Oncol Rep 29(6):2422–2430 Wang S, Pang TY, Gao M, Kang HX, Ding WB, Sun XW, Zhao Y, ZhuW, Tang XD, Yao YH, Hu XR, 2013, HPV E6 induces eIF4E transcription to promote the proliferation and migration of cervical cancer. FEBS Lett 587(6):690–697 Wu MY, Liu YX, Di XQ, Hu XR, 2013, EIF4E over-expresses and enhances cell proliferation and cell cycle progression in nasopharyngeal carcinoma. Med Oncol 30:400 Zhao Y, Pang TY, Wang Y, Wang S, Kang HX, Ding WB, Yong WW, Bie YH, Cheng XG, Zeng C, Yao YH, Li Q, Hu XR, 2014, LMP1 stimulates the transcription of eIF4E to promote the proliferation, migration and invasion of human nasopharyngeal carcinoma. FEBS J 281(13):3004–3018 Wang W, Wen Q, Luo J, Chu S, Chen L, Xu L, Zang H, Alnemah MM, Li J, Zhou J, Fan S, 2017, Suppression of β-catenin nuclear translocation by CGP57380 decelerates poor progression and potentiates radiation-induced apoptosis in nasopharyngeal carcinoma. Theranostics 7(7):2134–2149 Zheng J, Li J, Xu L, Xie G, Wen Q, Luo J, Li D, Huang D, Fan S, 2014, PhosphorylatedMnk1 and eIF4E are associated with lymph node metastasis and poor prognosis of nasopharyngeal carcinoma. PLoS One 9(2):e89220 Zheng J, Zhang C, Zhang Y, Pei J, Fan S, 2017, Over-expressions of p-eIF4E and p-4EBP1 are associated with poor prognosis in nasopharyngeal carcinoma. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 33(4):531–535 Zhang P, Wu SK, Wang Y, Fan ZX, Li CR, Feng M, Xu P, Wang WD, Lang JY, 2015, p53,MDM2, eIF4E and EGFR expression in nasopharyngeal carcinoma and their correlation with clinicopathological characteristics and prognosis: a retrospective study. Oncol Lett 9(1):113–118 Robichaud N, Del Rincon SV, Huor B, Alain T, Petruccelli LA, Hearnden J, Goncalves C, Grotegut S, Spruck CH, Furic L, Larsson O, Muller WJ, Miller WH, Sonenberg N, 2015, Phosphorylation of eIF4E promotes EMT and metastasis via translational control of SNAIL and MMP-3. Oncogene 34(16):2032– 2042 Smith KA, Zhou B, Avdulov S, Benyumov A, Peterson M, Liu Y, Okon A, Hergert P, Braziunas J, Wagner CR, Borok Z, Bitterman PB, 2015, Transforming growth factor-β1 induced epithelial mesenchymal transition is blocked by a chemical antagonist of translation factor eIF4E. Sci Rep 5:18233 Liu M, Peng P,Wang J,Wang L, Duan F, Jia D, Ruan Y, 2015, Gu J.RACK1-mediated translation control promotes liver fibrogenesis. Biochem Biophys Res Commun 463(3):255–261 Kwegyir-Afful AK, Bruno RD, Purushottamachar P, Murigi FN, Njar VC, 2016, Galeterone and VNPT55 disrupt Mnk-eIF4E to inhibit prostate cancer cell migration and invasion. FEBS J 283(21):3898–3918 Ramamurthy VP, Ramalingam S, Gediya LK, Njar VCO, 2018, The retinamide VNLG-152 inhibits f-AR/AR-V7 and MNKeIF4E signaling pathways to suppress EMT and castrationresistant prostate cancer xenograft growth. FEBS J 285(6):1051– 1063 Tajirika T, Tokumaru Y, Taniguchi K, Sugito N, Matsuhashi N, Futamura M, Yanagihara K, Akao Y, Yoshida K. DEAD-box protein RNA-helicase DDX6 regulates the expression of HER2 and FGFR2 at the post-transcriptional step in gastric cancer cells. Int J Mol Sci. 2018; 19(7). pii: E2005 Decarlo L, Mestel C, Barcellos-Hoff MH, Schneider RJ, 2015, Eukaryotic translation initiation factor 4E is a feed-forward translational coactivator of transforming growth factor β early protransforming events in breast epithelial cells. Mol Cell Biol 35(15):2597–2609 Dodd KM, Yang J, Shen MH, Sampson JR, Tee AR, 2015, mTORC1 drives HIF-1α and VEGF-A signalling via multiple mechanisms involving 4E-BP1, S6K1 and STAT3. Oncogene 34(17):2239–2250 Herdy B, Jaramillo M, Svitkin YV, Rosenfeld AB, Kobayashi M, Walsh D, Alain T, Sean P, Robichaud N, Topisirovic I, Furic L, Dowling RJO, Sylvestre A, Rong L, Colina R, Costa-Mattioli M, Fritz JH, Olivier M, Brown E, Mohr I, Sonenberg N, 2012, Translational control of the activation of transcription factor NF- κB and production of type I interferon by phosphorylation of the translation factor eIF4E. Nat Immunol 13(6):543–550 Mali AV, Joshi AA, Hegde MV, Kadam SS, 2018, Enterolactone modulates the ERK/NF-κB/snail signaling pathway in triplenegative breast cancer cell line MDA-MB-231 to revert the TGF- β-induced epithelial-mesenchymal transition. Cancer Biol Med 15(2):137–156 Jeong BY, Cho KH, Jeong KJ, Park YY, Kim JM, Rha SY, Park CG, Mills GB, Cheong JH, Lee HY, 2018, Rab25 augments cancer cell invasiveness through a β1 integrin/EGFR/VEGF-A/snail signaling axis and expression of fascin. Exp Mol Med 50(1):e435 Lin X, Sun R, Zhao X, Zhu D, Zhao X, Gu Q, Dong X, Zhang D, Zhang Y, Li Y, Sun B, 2017, C-myc overexpression drives melanoma metastasis by promoting vasculogenic mimicry via c-myc/ snail/Bax signaling. J Mol Med, Berl, 95(1):53–67 Zhai D, Cui C, Xie L, Cai L, Yu J, 2018, Sterol regulatory elementbinding protein 1 cooperates with c-Myc to promote epithelialmesenchymal transition in colorectal cancer. Oncol Lett 15(4): 5959–5965 Song R, Song H, Liang Y, Yin D, Zhang H, Zheng T,Wang J, Lu Z, Song X, Pei T, Qin Y, Li Y, Xie C, Sun B, Shi H, Li S, Meng X, Yang G, Pan S, Zhu J, Qi S, Jiang H, Zhang Z, Liu L, 2014, Reciprocal activation between ATPase inhibitory factor 1 and NF- κB drives hepatocellular carcinoma angiogenesis and metastasis. Hepatology 60(5):1659–1673 Wanami LS, Chen HY, Peiro S, 2008, Garcia de Herreros a, Bachelder RE. Vascular endothelial growth factor-a stimulates snail expression in breast tumor cells: implications for tumor progression. Exp Cell Res 314(13):2448–2453 Moon H, Ju HL, Chung SI, Cho KJ, Eun JW, Nam SW, Han KH, Calvisi DF, Ro SW, 2017, Transforming growth factor-β promotes liver tumorigenesis in mice via up-regulation of snail. Gastroenterology 153(5):1378–1391 Evdokimova V, Tognon C, Ng T, Ruzanov P, Melnyk N, Fink D, Sorokin A, Ovchinnikov LP, Davicioni E, Triche TJ, Sorensen PH, 2009, Translational activation of Snail1 and other developmentally regulated transcription factors by YB-1 promotes an epithelialmesenchymal transition. Cancer Cell 15:402–415 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1639 EP 1649 DI 10.1007/s12253-019-00733-x PG 11 ER PT J AU Bozdag, Z Tutar, E Dizibuyuk, FO Bakir, K AF Bozdag, Zehra Tutar, Ediz Dizibuyuk, Faruk Omer Bakir, Kemal TI Monoclonal Caveolin 1 Expression in the Differential Diagnosis of Malignant Pleural Mesothelioma and Pulmonary Adenocarcinoma: Is it Useful? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Caveolin 1; Mesothelioma; Pulmonary adenocarcinoma ID Caveolin 1; Mesothelioma; Pulmonary adenocarcinoma AB In this study we aim to demonstrate the value of monoclonal Caveolin 1 expression in distinguishing between malignant pleural mesothelioma and pulmonary adenocarcinoma. Total of 129 cases, consisting of 68 cases of malignant pleural mesothelioma (51 epitheloid, 12 biphasic, and 5 sarcomatoid type) and 61 cases of pulmonary adenocarcinoma were examined and stained with monoclonal Caveolin-1. Caveolin 1 expression with a membranous and /or cytoplasmic pattern was detected only in 32.35% (n:22/68) of malignant pleural mesothelioma and 6.5% (n:4/61) of pulmonary adenocarcinoma cases. This finding suggests that the choice of poly/monoclonal antibody for Caveolin 1 in the differential diagnosis of malignant pleural mesothelioma and pulmonary adenocarcinoma is important. C1 [Bozdag, Zehra] Gaziantep University, Faculty of Medicine, Department of PathologyGaziantep, Turkey. [Tutar, Ediz] Gaziantep University, Faculty of Medicine, Department of PathologyGaziantep, Turkey. [Dizibuyuk, Faruk Omer] Gaziantep University, Faculty of Medicine, Department of PathologyGaziantep, Turkey. [Bakir, Kemal] Gaziantep University, Faculty of Medicine, Department of PathologyGaziantep, Turkey. RP Bozdag, Z (reprint author), Gaziantep University, Faculty of Medicine, Department of Pathology, Gaziantep, Turkey. CR Bibby AC, TsimS,Kanellakis N, Ball H, Talbot DC, Blyth KGet al, 2016, Malignant pleural mesothelioma: an update on investigation, diagnosis and treatment. Eur Respir Rev 25:472–486 Christian Geltner C, Errhalt P, Baumgartner B, Ambrosch G, Machan B, 2016, Eckmayr J,; Austrian mesothelioma interest group, AMIG). Management of malignant pleural mesothelioma - part 1: epidemiology, diagnosis, and staging : consensus of the Austrian Mesothelioma Interest Group, AMIG). Wien Klin Wochenschr 128(17–18):611–617 Beckett P, Edwards J, Fennell D, Hubbard R, Woolhouse I, Peake MD, 2015, Demographics, management and survival of patients with malignant pleural mesothelioma in the National Lung Cancer Audit in England and Wales. Lung Cancer 88:344–348 Betta PG, Magnani C, Bensi T, Trincheri NF, Orecchia S, 2012, Immunohistochemistry and molecular diagnostics of pleural malignant mesothelioma. Arch Pathol Lab Med 136:253–261 Creaney J, Robinson BWS, Geltner C, Errhalt P, Baumgartner B, Ambrosch G, Austrian Mesothelioma Interest Group, AMIG,, 2017, Malignant Mesothelioma. biomarkers: from discovery to use in clinical practice for diagnosis, monitoring, screening, and treatment. Chest 152:143–149 Su XY, Li GD, Liu WP, Xie B, Jiang YH, 2011, Cytological differential diagnosis among adenocarcinoma, epithelial mesothelioma, and reactive mesothelial cells in serous effusions by immunocytochemistry. Diagn Cytopathol 39:900–908 Gueugnon F, Leclercq S, Blanquart C, Sagan C, Cellerin L, Padieu M, Perigaud C, Scherpereel A, GregoireM, 2011, Identification of novel markers for the diagnosis ofmalignant pleural mesothelioma. Am J Pathol 178:1033–1042 Churg A, Sheffield BS, Galateau-Salle F, 2016, New markers for separating benign from malignant Mesothelial proliferations are we there yet? Arch Pathol Lab Med 140:318–321 Fu P, Chen F, Pan Q, Zhao X, Zhao C, Cho WC et al, 2017, The different functions and clinical significances of caveolin-1 in human adenocarcinoma and squamous cell carcinoma. Onco Targets Ther 10:819–835 Amatya VJ, Takeshima Y, Kohno H, Kushitani K, Yamada T, Morimoto C, Inai K, 2009, Caveolin- 1 is a novel immunohistochemical marker to differentiate epithelioid mesothelioma from lung adenocarcinoma. Histopathology 55:10–19 Astoul P, Roca E, GalateauSalle F, Scherpereel A, 2012)Malignant pleural mesothelioma: from the bench to the bedside. Respiration 83:481–493 Husain AN, Colby T, Ordonez NG et al, 2012, Guidelines for pathologic diagnosis of malignant mesothelioma. Arch Pathol Lab Med 137 Dinu M, Ciurea RN, Stefan M, Georgescu AC, 2012, The role of immunohistochemistry in the diagnosis of neoplastic pleural effusions. Rom J Morphol Embryol 53:817–820 Panou V, Vyberg M, Weinreich UM, Meristoudis C, Falkmer UG, Roe OD, 2015, The established and future biomarkers of malignant pleural mesothelioma. Cancer Treat Rev 41:486–495 Richter G, Heidersdorf H, Hirschfeld D, Krebbel F, 2016 Mar 4, Positive TTF-1 expression in malignant mesothelioma: a case report. Am J Case Rep 17:133–136 Chaouche-Mazouni S, Scherpereel A, Zaamoum R, Mihalache A, Amir ZC, Lebaili N, Delaire B, Gosset P, 2015, Claudin 3, 4, and 15 expression in solid tumors of lung adenocarcinoma versus malignant pleural mesothelioma. Ann Diagn Pathol 19(4):193–197 Carbone M, Shimizu D, Napolitano A, Tanji M, Pass HI, Yang H, Pastorino S, 2016, Positive nuclear BAP1 immunostaining helps differentiate non-small cell lung carcinomas from malignant mesothelioma. Oncotarget 7:59314–59321 Sahin N, Akatli AN, Celik MR, Ulutas H, Samdanci ET, Colak C, 2017, The role of CD90 in the differential diagnosis of pleural malignant mesothelioma, pulmonary carcinoma and comparison with Calretinin. Pathol Oncol Res 23:487–491 Ucer O, Dagli AF, Kilicarslan A, Artas G, 2013, Value of Glut-1 and Kocmarkers in the differential diagnosis of reactivemesothelial hyperplasia, malignant mesothelioma and pulmonary adenocarcinoma. Turk Patoloji Derg 29:94–100 Amatya VJ, Kushitani K, Mawas AS, Miyata Y, Okada M, Kishimoto T et al, 2017, MUC4, a novel immunohistochemical marker identified by gene expression profiling, differentiates pleural sarcomatoid mesothelioma from lung sarcomatoid carcinoma. Mod Pathol 30:672–681 Ren R, Yin P, Zhang Y, Zhou J, Zhou Y, Xu R et al, 2016, Diagnostic value of fibulin-3 for malignant pleural mesothelioma: a systematic review and meta-analysis. Oncotarget 7:84851–84859 Jia Y, Wang N, Wang J et al, 2014, Down-regulation of stromal caveolin-1 expression in esophageal squamous cell carcinoma: a potent predictor of lymph node metastases, early tumor recurrence, and poor prognosis. Ann Surg Oncol 21:329–336 Zhan P, Shen XK, Qian Q et al, 2012, Expression of caveolin-1 is correlated with disease stage and survival in lung adenocarcinomas. Oncol Rep 27:1072–1078 Ma X, Liu L, Nie W, Li Y, Zhang B, Zhang J, Zhou R, 2013, Prognostic role of caveolin in breast cancer: a meta-analysis. Breast 22:462–469 Tang Y, Zeng X, He F, Liao Y, Qian N, ToiM, 2012, Caveolin-1 is related to invasion, survival, and poor prognosis in hepatocellular cancer. MedOncol 29:977–984 Nam KH, Lee BL, Park JH, Kim J, Han N, Lee HE, Kim MA, Lee HS, Kim WH, 2013, Caveolin 1 expression correlates with poor prognosis and focal adhesion kinase expression in gastric cancer. Pathobiology 80:87–94 Suzuoki M, Miyamoto M, Kato K, Hiraoka K, Oshikiri T, Nakakubo Y, Fukunaga A, Shichinohe T, Shinohara T, Itoh T, Kondo S, Katoh H, 2002, Impact of caveolin-1 expression on prognosis of pancreatic ductal adenocarcinoma. Br J Cancer 87: 1140–1144 Zhao Z, Han FH, Yang SB, Hua LX,Wu JH, ZhanWH, 2015, Loss of stromal caveolin-1 expression in colorectal cancer predicts poor survival. World J Gastroenterol 21:1140–1147 Joo HJ, Oh DK, Kim YS, Lee KB, Kim SJ, 2004, Increased expression of caveolin-1 and microvessel density correlates with metastasis and poor prognosis in clear cell renal cell carcinoma. BJU Int 93:291–296 Karam JA, Lotan Y, Roehrborn CG, Ashfaq R, KarakiewiczPI SSF, 2007, Caveolin-1 overexpression is associated with aggressive prostate cancer recurrence. Prostate 67:614–622 von Ruhland CJ, Campbell L, Gumbleton M, Jasani B, Newman GR, 2004, Immunolocalization of caveolin-1 in rat and human mesothelium. J Histochem Cytochem 52:1415–1425 Thapa B, Walkiewicz M, Murone C, Asadi K, Deb S, Barnett S, Knight S, Mitchell P, Liew D, Watkins DN, John T, 2016, Calretinin but not caveolin-1 correlates with tumour histology and survival in malignant mesothelioma. Pathology 48:660–665 Righi L, Cavallo MC, Gatti G, Monica V, Rapa I, Busso S et al, 2014, Tumor/stromal caveolin-1 expression patterns in pleuralmesothelioma define a subgroup of the epithelial histotype with poorer prognosis. Am J Clin Pathol 141:816–827 Attanoos RL, Griffin A, Gibbs AR, 2003, The use of immunohistochemistry distinguishing reactive from neoplastic mesothelium. A novel use for desmin and comparative evaluation with epithelial membrane antigen, p53, platelet-derived growth factor-receptor, Pglycoprotein and Bcl-2. Histopathology 43:231–238 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1651 EP 1656 DI 10.1007/s12253-019-00751-9 PG 6 ER PT J AU Bozdag, Z Tutar, E Dizibuyuk, FO Bakir, K AF Bozdag, Zehra Tutar, Ediz Dizibuyuk, Faruk Omer Bakir, Kemal TI Correction to: Monoclonal Caveolin 1 Expression in the Differential Diagnosis of Malignant Pleural Mesothelioma and Pulmonary Adenocarcinoma: Is it Useful? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Correction to: Pathology & Oncology Research https://doi.org/10.1007/s12253-019-00751-9 The original version of this article unfortunately contained an error in Fig. 1. Cav-1 expression in MPM and PA cases failed to show the histopathological details in Fig. 1 due to technical problem. The figure with the proper sharpness and clarity is shown in the next page. C1 [Bozdag, Zehra] Gaziantep University, Faculty of Medicine, Department of PathologyGaziantep, Turkey. [Tutar, Ediz] Gaziantep University, Faculty of Medicine, Department of PathologyGaziantep, Turkey. [Dizibuyuk, Faruk Omer] Gaziantep University, Faculty of Medicine, Department of PathologyGaziantep, Turkey. [Bakir, Kemal] Gaziantep University, Faculty of Medicine, Department of PathologyGaziantep, Turkey. RP Bozdag, Z (reprint author), Gaziantep University, Faculty of Medicine, Department of Pathology, Gaziantep, Turkey. EM zbozdagmd@gmail.com NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1657 EP 1658 DI 10.1007/s12253-020-00793-4 PG 2 ER PT J AU Kato, A Kato, K Miyazawa, H Kobayashi, H Noguchi, N Kawashiri, Sh AF Kato, Aki Kato, Koroku Miyazawa, Hiroki Kobayashi, Hisano Noguchi, Natsuyo Kawashiri, Shuichi TI Focal Adhesion Kinase (FAK) Overexpression and Phosphorylation in Oral Squamous Cell Carcinoma and their Clinicopathological Significance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Focal adhesion kinase; FAK; FAKpY397; p16; Oral squamous cell carcinoma; Prognosis ID Focal adhesion kinase; FAK; FAKpY397; p16; Oral squamous cell carcinoma; Prognosis AB Focal adhesion kinase (FAK) is involved in progression of various cancers, and FAK overexpression has been associated with cancer invasion and metastasis. However, the involvement of FAK expression in the clinicopathological malignancy of oral squamous cell carcinoma (OSCC) remains unknown. In addition, there is no consensus regarding the role of p16 expression in OSCC. In this study, the immunohistochemically measured expression of FAK, phosphorylated FAK (FAKpY397) and p16 expressions and their associations with clinicopathological features and 5-year survival rates were examined in surgical samples from 70 patients with primary OSCC. FAK and FAKpY397 were expressed at high levels in 42 cases (60.0%) and 34 cases (48.6%), respectively, and 9 cases (12.9%) were positive for p16. FAK expression was significantly correlated with local recurrence, subsequent metastasis, and the mode of invasion. FAKpY397 expression was significantly correlated with both N classification and the mode of invasion. p16 expression was significantly correlated with clinical stage only. Patients having high expression of FAK, FAKpY397, or both showed significantly worse prognosis, but p16 expression showed no significant relation to prognosis. The results suggested that overexpression and phosphorylation of FAK in OSCC may affect cancer progression, such as local invasion and lymph node metastasis, and thereby contribute to life prognosis. C1 [Kato, Aki] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Kato, Koroku] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Miyazawa, Hiroki] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Kobayashi, Hisano] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Noguchi, Natsuyo] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Kawashiri, Shuichi] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. RP Kato, K (reprint author), Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 920-8641 Kanazawa, Japan. EM k-koroku@oral.m.kanazawa-u.ac.jp CR Simard EP, Torre LA, Jemal A, 2014, International trends in head and neck cancer incidence rates: differences by country, sex and anatomic site. Oral Oncol 50:387–403 Schaller MD, 2010, Cellular functions of FAK kinases: insight into molecular mechanisms and novel functions. J Cell Sci 123:1007–1013 McLean GW, Carragher NO, Avizienyte E, Evans J, Brunton VG, Frame MC, 2005, The role of focal-adhesion kinase in cancer - a new therapeutic opportunity. Nat Rev Cancer 5:505–515 Lee BY, Timpson P, Horvath LG, Daly RJ, 2015, FAK signaling in human cancer as a target for therapeutics. Pharmacol Ther 146:132–149 Owens LV, Xu L, Dent GA, Yang X, Sturge GC, Craven RJ, Cance WG, 1996, Focal adhesion kinase as a marker of invasive potential in differentiated human thyroid cancer. Ann Surg Oncol 3:100–105 Tremblay L, Hauck W, Aprikian AG, Begin LR, Chapdelaine A, Chevalier S, 1996, Focal adhesion kinase, pp125FAK, expression, activation and association with paxillin and p50CSK in human metastatic prostate carcinoma. Int J Cancer 68:164–171 McCormack SJ, Brazinski SE,Moore JL Jr,Werness BA, Goldstein DJ, 1997, Activation of the focal adhesion kinase signal transduction pathway in cervical carcinoma cell lines and human genital epithelial cells immortalized with human papillomavirus type 18. Oncogene 15:265–274 Judson PL, He X, Cance WG, Van Le L, 1999, Overexpression of focal adhesion kinase, a protein tyrosine kinase, in ovarian carcinoma. Cancer 86:1551–1556 Lark AL, Livasy CA, Calvo B, Caskey L, Moore DT, Yang X, Cance WG, 2003, Overexpression of focal adhesion kinase in primary colorectal carcinomas and colorectal liver metastases: immunohistochemistry and real-time PCR analyses. Clin Cancer Res 9: 215–222 Owens LV, Xu L, Craven RJ, Dent GA, Weiner TM, Kornberg L, Liu ET, Cance WG, 1995, Overexpression of the focal adhesion kinase, p125FAK, in invasive human tumors. Cancer Res 55:2752–2755 Recher C, Ysebaert L, Beyne-Rauzy O, Mansat-De Mas V, Ruidavets JB, Cariven P, Demur C, Payrastre B, Laurent G, Racaud-Sultan C, 2004, Expression of focal adhesion kinase in acute myeloid leukemia is associated with enhanced blast migration, increased cellularity, and poor prognosis. Cancer Res 64: 3191–3197 Schlaepfer DD, Mitra SK, Ilic D, 2004, Control of motile and invasive cell phenotypes by focal adhesion kinase. Biochim Biophys Acta 1692:77–102 Ang KK,Harris J,Wheeler R,Weber R, Rosenthal DI, Nguyen-Tan PF, Westra WH, Chung CH, Jordan RC, Lu C, Kim H, Axelrod R, Silverman CC, Redmond KP, Gillison ML, 2010, Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 363:24–35 Allen CT, Lewis JS Jr, El-Mofty SK, Haughey BH, Nussenbaum B, 2010, Human papillomavirus and oropharynx cancer: biology, detection and clinical implications. Laryngoscope 120:1756–1772 Marur S, D'Souza G, Westra WH, Forastiere AA, 2010, HPVassociated head and neck cancer: a virus-related cancer epidemic. Lancet Oncol 11:781–789 Wang MB, Liu IY, Gornbein JA, Nguyen CT, 2015, HPV-positive oropharyngeal carcinoma: a systematic review of treatment and prognosis. Otolaryngol Head Neck Surg 153:758–769 In, 2009, International Union against Cancer, LH Sobin, MK Gospodrowicz, CH Wittekind, eds.). TNM Classification of Malignant Tumours. 7th Ed. Wiley-Liss, New York, NY Yamamoto E, Kohama G, Sunakawa H, Iwai M, Hiratsuka H, 1983, Mode of invasion, bleomycin sensitivity, and clinical course in squamous cell carcinoma of the oral cavity. Cancer 51:2175–2180 Theocharis S, Klijanienko J, Giaginis C, Alexandrou P, Patsouris E, Sastre-Garau X, 2012, FAK and Src expression in mobile tongue squamous cell carcinoma: associations with clinicopathological parameters and patients survival. J Cancer Res Clin Oncol 138:1369–1377 Heiduschka G, Grah A, Oberndorfer F, Kadletz L, Altorjai G, Kornek G,Wrba F, Thurnher D, Selzer E, 2015, Improved survival in HPV/p16-positive oropharyngeal cancer patients treated with postoperative radiotherapy. Strahlenther Onkol 191:209–216 Yamamoto E, 1994, Role of chemotherapy for oral cancer from the viewpoint of mode of invasion. Asian J Oral Maxillofac Surg 6: 113–121 Kurokawa H, Yamashita Y, Takeda S, Zhang M, Fukuyama H, Takahashi T, 2002, Risk factors for late cervical lymph node metastases in patients with stage I or II carcinoma of the tongue. Head Neck 24:731–736 Pereira CH, Morais MO, Martins AF, Soares MQ, Alencar Rde C, Batista AC, Leles CR, Mendonca EF, 2016, Expression of adhesion proteins, E-cadherin and β-catenin, and cell proliferation, Ki- 67, at the invasive tumor front in conventional oral squamous cell and basaloid squamous cell carcinomas. Arch Oral Biol 61:8–15 Sen S, Carnelio S, 2016, Expression of epithelial cell adhesion molecule, EpCAM, in oral squamous cell carcinoma. Histopathology 68:897–904 Richardson A, Parsons T, 1996, A mechanism for regulation of the adhesion-associated proteintyrosine kinase pp125FAK. Nature 380: 538–540 Webb DJ, Donais K, Whitmore LA, Thomas SM, Turner CE, Parsons JT, Horwitz AF, 2004, FAK-Src signalling through paxillin, ERK and MLCK regulates adhesion disassembly. Nat Cell Biol 6:154–161 Avizienyte E, Wyke AW, Jones RJ, McLean GW, Westhoff MA, Brunton VG, Frame MC, 2002, Src-induced de-regulation of Ecadherin in colon cancer cells requires integrin signalling. Nat Cell Biol 4:632–638 Eliceiri BP, Puente XS, Hood JD, Stupack DG, Schlaepfer DD, Huang XZ, Sheppard D, Cheresh DA, 2002, Src-mediated coupling of focal adhesion kinase to integrin alpha(v)beta5 in vascular endothelial growth factor signaling. J Cell Biol 157:149–160 Aronsohn MS, Brown HM, Hauptman G, Kornberg LJ, 2003, Expression of focal adhesion kinase and phosphorylated focal adhesion kinase in squamous cell carcinoma of the larynx. Laryngoscope 113:1944–1948 Moon HS, Park WI, Choi EA, Chung HW, Kim SC, 2003, The expression and tyrosine phosphorylation of E-cadherin/catenin adhesion complex, and focal adhesion kinase in invasive cervical carcinomas. Int J Gynecol Cancer 13:640–646 Brunton VG,Avizienyte E, FinchamVJ, Serrels B, MetcalfCA3rd, Sawyer TK, Frame MC, 2005, Identification of Src-specific phosphorylation site on focal adhesion kinase: dissection of the role of Src SH2 and catalytic functions and their consequences for tumor cell behavior. Cancer Res 65:1335–1342 Kornberg LJ, 1998, Focal adhesion kinase expression in oral cancers 20:634–9 Kornberg LJ, 1998, Focal adhesion kinase and its potential involvement in tumor invasion and metastasis. Head Neck 20:745–752 Grisaru-Granovsky S, Salah Z, Maoz M, Pruss D, Beller U, Bar- Shavit R, 2005, Differential expression of protease activated receptor 1, Par1, and pY397FAK in benign and malignant human ovarian tissue samples. Int J Cancer 113:372–378 Chiu YW, Liou LY, Chen PT, Huang CM, Luo FJ, Hsu YK, Yuan TC, 2016, Tyrosine 397 phosphorylation is critical for FAKpromoted Rac1 activation and invasive properties in oral squamous cell carcinoma cells. Lab Investig 96:296–306 Han X, Xue L, Zhou L, Gong L, Zhu S, Yao L,Wang S, Lan M, Li Y, ZhangW(2013, The role of PTPN13 in invasion and metastasis of lung squamous cell carcinoma. Exp Mol Pathol 95:270–275 Aust S, Auer K, Bachmayr-Heyda A, Denkert C, Sehouli J, Braicu I,Mahner S, Lambrechts S,Vergote I, GrimmC, Horvat R, Castillo- Tong DC, Zeillinger R, Pils D, 2014, Ambivalent role of pFAKY397 in serous ovarian cancer–a study of the OVCAD consortium. Mol Cancer 13:67 Lai IR, Chu PY, Lin HS, Liou JY, Jan YJ, Lee JC, Shen TL, 2010, Phosphorylation of focal adhesion kinase at Tyr397 in gastric carcinomas and its clinical significance. Am J Pathol 177:1629–1637 Albasri A, Fadhil W, Scholefield JH, Durrant LG, Ilyas M, 2014, Nuclear expression of phosphorylated focal adhesion kinase is associated with poor prognosis in human colorectal cancer. Anticancer Res 34:3969–3974 Ding L, Sun X, You Y, Liu N, Fu Z, 2010, Expression of focal adhesion kinase and phosphorylated focal adhesion kinase in human gliomas is associated with unfavorable overall survival. Transl Res 156:45–52 Duxbury MS, Ito H, Benoit E, Zinner MJ, Ashley SW, Whang EE, 2003, RNA interference targeting focal adhesion kinase enhances pancreatic adenocarcinoma gemcitabine chemosensitivity. Biochem Biophys Res Commun 311:786–792 Sonoda Y, Matsumoto Y, Funakoshi M, Yamamoto D, Hanks SK, Kasahara T, 2000, Anti-apoptotic role of focal adhesion kinase, FAK). Induction of inhibitor-of-apoptosis proteins and apoptosis suppression by the overexpression of FAKin a human leukemic cell line, HL-60. J Biol Chem 275:16309–16315 Munoz N, Bosch FX, Castellsague X, Diaz M, de Sanjose S, Hammouda D, Shah KV, Meijer CJ, 2004, Against which human papillomavirus types shall we vaccinate and screen? The international perspective. Int J Cancer 111:278–285 Khleif SN, DeGregori J, Yee CL, Otterson GA, Kaye FJ, Nevins JR, Howley PM, 1996, Inhibition of cyclin D-CDK4/CDK 6 activity is associated with an E2F-mediated induction of cyclin kinase inhibitor activity. Proc Natl Acad Sci U S A 93:424–429 Tong X, Howley PM, 1997, The bovine papillomavirus E6 oncoprotein interacts with paxillin and disrupts the actin cytoskeleton. Proc Natl Acad Sci U S A 94:4412–4417 Shintani S, Mihara M, Nakahara Y, Kiyota A, Yoshihama Y, Ueyama Y, Matsumura T, 2000, Infrequent alternations of RB pathway(Rb-p16INK4A-cyclinD1, in adenoid cystic carcinoma of salivary glands. Anticancer Res 20:2169–2175 Takeuchi H, Ozawa S, Ando N, Shih CH, Koyanagi K, Ueda M, Kitajima M, 1997, Altered p16/MTS1/CDKN2 and cyclin D1/PRAD-1 gene expression is associated with the prognosis of squamous cell carcinoma of the esophagus. Clin Cancer Res 3:2229–2236 Namazie A, Alavi S, Olopade OI, Pauletti G, Aghamohammadi N, Aghamohammadi M, Gornbein JA, Calcaterra TC, Slamon DJ, Wang MB, Srivatsan ES, 2002, Cyclin D1 amplification and p16(MTS1/CDK4I)deletion correlate with poor prognosis in head and neck tumors. Laryngoscope 112:472–481 Zafereo ME, Xu L, Dahlstrom KR, Viamonte CA, El-Naggar AK, Wei Q, Li G, Sturgis EM, 2016, Squamous cell carcinoma of the oral cavity often overexpresses p16 but is rarely driven by human papillomavirus. Oral Oncol 56:47–53 Kouketsu A, Sato I, Abe S, Oikawa M, Shimizu Y, Takahashi T, Kumamoto H, 2016, Detection of human papillomavirus infection in oral squamous cell carcinoma: a cohort study of Japanese patients. J Oral Pathol Med 45:565–572 Gotz C, Drecoll E, Straub M, Bissinger O, Wolff KD, Kolk A, 2016, Impact of HPV infection on oral squamous cell carcinoma. Oncotarget 7:76704–76712 Ohara T, Kawashiri S, TanakaA, Noguchi N, Kitahara H, Okamune A, Kato K, Hase T, Nakaya H, Yoshizawa K, 2009, Integrin expression levels correlate with invasion, metastasis and prognosis of oral squamous cell carcinoma. Pathol Oncol Res 15:429–436 Raguse JD, Gath HJ, Bier J, Riess H, Oettle H, 2004, Cilengitide, EMD 121974, arrests the growth of a heavily pretreated highly vascularised head and neck tumour. Oral Oncol 40:228–230 Brown NF, Williams M, Arkenau HT, Fleming RA, Tolson J, Yan L, Zhang J, Swartz L, Singh R, Auger KR, Lenox L, Cox D, Lewis Y, Plisson C, Searle G, Saleem A, Blagden S, Mulholland P, 2018, A study of the focal adhesion kinase inhibitor GSK2256098 in patients with recurrent glioblastoma with evaluation of tumor penetration of [11C]GSK2256098. Neuro-Oncology 20:1634–1642 Jeong K, Murphy JM, Rodriguez YAR, Kim JS, Ahn EE, Lim SS, 2019, FAK inhibition reduces metastasis of α4 integrin-expressing melanoma to lymph nodes by targeting lymphatic VCAM-1 expression. Biochem Biophys Res Commun 509:1034–1040 Hu C, Chen X,Wen J, Gong L, Liu Z,Wang J, Liang J, Hu F, Zhou Q, Wei L, Shen Y, Zhang W, 2017, Antitumor effect of focal adhesion kinase inhibitor PF562271 against human osteosarcoma in vitro and in vivo. Cancer Sci 108:1347–1356 Shimizu T, Fukuoka K, Takeda M, Iwasa T, Yoshida T, Horobin J, Keegan M, Vaickus L, Chavan A, Padval M, Nakagawa K, 2016, A first-in-Asian phase 1 study to evaluate safety, pharmacokinetics and clinical activity of VS-6063, a focal adhesion kinase, FAK, inhibitor in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol 77:997–1003 Kurio N, Shimo T, Fukazawa T, Okui T, Hassan NM, Honami T, Horikiri Y, Hatakeyama S, Takaoka M, Naomoto Y, Sasaki A, 2012, Anti-tumor effect of a novel FAK inhibitor TAE226 against human oral squamous cell carcinoma. Oral Oncol 48:1159–1170 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1659 EP 1667 DI 10.1007/s12253-019-00732-y PG 9 ER PT J AU Jing, W Peng, R Zhu, M Lv, Sh Jiang, Sh Ma, J Ming, L AF Jing, Wei Peng, Ruoyu Zhu, Man Lv, Shaogang Jiang, Shitao Ma, Junfen Ming, Liang TI Differential Expression and Diagnostic Significance of Pre-Albumin, Fibrinogen Combined with D-Dimer in AFP-Negative Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pre-albumin; Fibrinogen; D-Dimer; AFP-negative hepatocellular carcinoma; Diagnosis ID Pre-albumin; Fibrinogen; D-Dimer; AFP-negative hepatocellular carcinoma; Diagnosis AB Hepatocellular carcinoma (HCC) is one of the most malignant cancers with high morbidity and mortality. Nowadays, AFP-negative hepatocellular carcinoma (AFP-NHCC) has been found in many HCC patients and AFP analysis can’t be used to screen HCC in these cases. In this study, we have examined the expression patterns of pre-albumin (PA), fibrinogen, D-Dimer and their clinical significance in AFP-NHCC. We recruited 214 AFP-NHCC patients and 210 controls in the study. PA, fibrinogen and D-Dimer levels were detected by turbidimetry, clauss and immunoturbidimetry methods, respectively. Serum PA levels were significantly lower in AFP-NHCC (84.5 ± 24.7 mg/L) than that in the controls (240.6 ± 59.4 mg/L, P < 0.05). For plasma fibrinogen levels, there was no difference between the controls (2.9 ± 0.7 g/L) and AFP-NHCC (2.5 ± 0.7 g/L). Compared with AFP-NHCC (0.8 ± 0.2 mg/L), plasma D-Dimer levels were significantly lower in controls (0.1 ± 0.0 mg/L, P < 0.05). The levels of PA, fibrinogen and D-Dimer were significantly correlated with differentiation (P < 0.01), and the PA and D-Dimer values were correlated with TNM stage (P < 0.05). Moreover, PA levels were correlated with tumor size (P = 0.034). Receiver operating characteristic curve (ROC) analyses elaborated that combination of PA, fibrinogen and D-Dimer possessed a higher sensitivity (93.4%) for differentiating AFP-NHCC from the controls, but the diagnostic specificity was reduced due to the combination of fibrinogen. After adjusting for all significant outcome predictors of the univariate logistic regression analysis, low levels of PA and high levels of D-Dimer were remained independent unfavorable outcome predictors (P < 0.05). Our data suggested that the expression levels of PA, fibrinogen and D-Dimer played critical roles in AFP-NHCC tumorigenesis. Moreover, PA and D-Dimer might be considered as potential diagnostic indicators in AFP-NHCC. C1 [Jing, Wei] the First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Department of Clinical Laboratory, 450000 Zhengzhou, China. [Peng, Ruoyu] the First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Department of Clinical Laboratory, 450000 Zhengzhou, China. [Zhu, Man] Huazhong University of Science and Technology, Tongji Medical College, the Central Hospital of Wuhan, Department of Medical Laboratory, 430000 Wuhan, China. [Lv, Shaogang] the First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Department of Clinical Laboratory, 450000 Zhengzhou, China. [Jiang, Shitao] the First Affiliated Hospital of Zhengzhou University, Henan Medical Key Laboratory of Molecular Imaging, 450000 Zhengzhou, China. [Ma, Junfen] the First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Department of Clinical Laboratory, 450000 Zhengzhou, China. [Ming, Liang] the First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Department of Clinical Laboratory, 450000 Zhengzhou, China. RP Jing, W (reprint author), the First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Department of Clinical Laboratory, 450000 Zhengzhou, China. EM xjw2009@yeah.net CR CorwinMT, LeeAY, Fananapazir G, LoehfelmTW, Sarkar S, Sirlin CB, 2018, Nonstandardized terminology to describe focal liver lesions in patients at risk for hepatocellular carcinoma: implications regarding clinical communication. AJR Am J Roentgenol 210(1): 85–90. , DOI 10.2214/ajr.17.18416 Nadinskaia MY, Kodzoeva KB, Ulyanova KA, Volkova AS, Rogacheva SI, Dekhanov AS, Strelkova DA, Ivashkin VT, 2019, Risk factors associated with portal vein thrombosis in liver cirrhosis: a case-control study. Terapevticheskii arkhiv 91(2):73–81. , DOI 10.26442/00403660.2019.02.000153 Monfardini L, Orsi F, Caserta R, Sallemi C, Della Vigna P, Bonomo G, Varano G, Solbiati L,Mauri G, 2018, Ultrasound and cone beam CT fusion for liver ablation: technical note. International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group. 35(1):500–504. , DOI 10.1080/02656736.2018.1509237 Wang M, Devarajan K, Singal AG, Marrero JA, Dai J, Feng Z, Rinaudo JA, Srivastava S, Evans A, Hann HW, Lai Y, Yang H, Block TM, Mehta A, 2016, The Doylestown algorithm: a test to improve the performance of AFP in the detection of hepatocellular carcinoma. Cancer Prev Res, Phila, 9(2):172–179. , DOI 10.1158/1940-6207.capr-15-0186 Chen ZZ, Huang L, Wu YH, Zhai WJ, Zhu PP, Gao YF, 2016, LncSox4 promotes the self-renewal of liver tumour-initiating cells through Stat3-mediated Sox4 expression. Nat Commun 7:12598. , DOI 10.1038/ncomms12598 Arbab K, Majid H, Jafri L, Akram B, Raheem A, Jamil B, Hashmi M, Khan AH, 2019, Assessing nutritional status of critically ill patients using serum Prealbumin levels. J Ayub Med Coll, Abbottabad : JAMC 31(2):178–181 Wen X, Yao M, Lu Y, Chen J, Zhou J, Chen X, Zhang Y, Lu W, Qian X, Zhao J, Zhang L, Ding S, Lu F, 2018, Integration of Prealbumin into child-Pugh classification improves prognosis predicting accuracy in HCC patients considering curative surgery. J Clin Transl Hepatol 6(4):377–384. , DOI 10.14218/jcth. 2018.00004 Hou C, Jiang F, Ma H, ZhuQ,Wang Z, Zhao B,Xue T, Tan S,Yang R, Qian Y, Luo X, Zhao M, Xu X, Chen L, Li J, 2019, Prognostic role of preoperative platelet, fibrinogen, and D-dimer levels in patients with non-small cell lung cancer: a multicenter prospective study. Thoracic Cancer 10(2):304–311. , DOI 10.1111/ 1759-7714.12956 Cai HX, Li XQ, Wang SF, 2018, Prognostic value of fibrinogen and D-dimer-fibrinogen ratio in resectable gastrointestinal stromal tumors. World J Gastroenterol 24(44):5046–5056. , DOI 3748/wjg.v24.i44.5046 10. Fan S, Zhao G, An G, 2019, High pretreatment plasma D-dimer levels are associated with shorter overall survival in patients with small cell lung cancer. J Int Med Res 47(1):215–224. https://doi. org/10.1177/0300060518799869 Darweesh SK, Gad AA, 2019, Percutaneous microwave ablation for HCV-related hepatocellular carcinoma: efficacy, safety, and survival. Turk J Gastroenterol 30(5):445–453. , DOI 10.5152/ tjg.2019.17191 Yamada R, Bassaco B, Bracewell S, Gillen K, Kocher M, Collins H, Anderson MB, Guimaraes M, 2019, Long-term follow-up after conventional transarterial chemoembolization, c-TACE, with mitomycin for hepatocellular carcinoma, HCC). J Gastrointest Oncol 10(2):348–353. , DOI 10.21037/jgo.2019.01.01 Fung J, Cheung KS,Wong DK,Mak LY, ToWP, SetoWK, Lai CL, Yuen MF, 2018, Long-term outcomes and predictive scores for hepatocellular carcinoma and hepatitis B surface antigen seroclearance after hepatitis B e-antigen seroclearance. Hepatology, Baltimore, Md, 68(2):462–472. , DOI 10. 1002/hep.29874 Fan L, Chi C, Guo S,Wang Y, CaiW, Shao X, Xu F, Pan J, Zhu Y, Shangguan X, Xin Z, Hu J, Qian H, Xie S,Wang R, Zhou L, Dong B, Xue W, 2017, Serum pre-albumin predicts the clinical outcome in metastatic castration-resistant prostate Cancer patients treated with Abiraterone. J Cancer 8(17):3448–3455. , DOI 10. 7150/jca.21134 Braverman LE, Vagenakis A, Downs P, Foster AE, Sterling K, Ingbar SH, 1973, Effects of replacement doses of sodium Lthyroxine on the peripheral metabolism of thyroxine and triiodothyronine in man. J Clin Invest 52(5):1010–1017. , DOI 10.1172/jci107265 Yang F, Wei L, Huo X, Ding Y, Zhou X, Liu D, 2018, Effects of early postoperative enteral nutrition versus usual care on serum albumin, prealbumin, transferrin, time to first flatus and postoperative hospital stay for patients with colorectal cancer: a systematic review and meta-analysis. Contemp Nurse 54(6): 561–577. , DOI 10.1080/10376178.2018.1513809 Jia RR, Zhong JH, Huo RR, Su QB, Xiang X, Zhao FL, Qin ZB, Chen JH, Liao YY, Ma L, Xiang BD, Zhang CY, Li LQ, 2019, Correlation between serum prealbumin and prognosis of patients with hepatocellular carcinoma after hepatectomy. J Surg Oncol 119(6):794–800. , DOI 10.1002/jso.25378 Tennent GA, Brennan SO, Stangou AJ, O'Grady J, Hawkins PN, Pepys MB, 2007, Human plasma fibrinogen is synthesized in the liver. Blood 109(5):1971–1974. , DOI 10.1182/blood- 2006-08-040956 Mosesson MW, 2005, Fibrinogen and fibrin structure and functions. J Thromb Haemost 3(8):1894–1904. , DOI 10. 1111/j.1538-7836.2005.01365.x Arigami T, Okumura H, Matsumoto M, Uchikado Y, Uenosono Y, Kita Y, Owaki T, Mori S, Kurahara H, Kijima Y, Ishigami S, Natsugoe S, 2015, Analysis of the fibrinogen and neutrophillymphocyte ratio in esophageal squamous cell carcinoma: a promising blood marker of tumor progression and prognosis. Medicine 94(42):e1702. , DOI 10.1097/md.0000000000001702 Son HJ, Park JW, Chang HJ, Kim DY, Kim BC, Kim SY, Park SC, Choi HS, Oh JH, 2013, Preoperative plasma hyperfibrinogenemia is predictive of poor prognosis in patients with nonmetastatic colon cancer. Ann Surg Oncol 20(9):2908–2913. , DOI 10.1245/ s10434-013-2968-8 Garcia MG, Bayo J, Bolontrade MF, Sganga L, Malvicini M, Alaniz L, Aquino JB, Fiore E, Rizzo MM, RodriguezA, Lorenti A, Andriani O, Podhajcer O, Mazzolini G, 2011, Hepatocellular carcinoma cells and their fibrotic microenvironment modulate bone marrow-derived mesenchymal stromal cell migration in vitro and in vivo. Mol Pharm 8(5):1538–1548. , DOI 10.1021/mp200137c Zheng S, Shen J, Jiao Y, Liu Y, Zhang C, Wei M, Hao S, Zeng X, 2009, Platelets and fibrinogen facilitate each other in protecting tumor cells from natural killer cytotoxicity. Cancer Sci 100(5):859– 865. , DOI 10.1111/j.1349-7006.2009.01115.x Ridker PM, Howard CP, Walter V, Everett B, Libby P, Hensen J, Thuren T, 2012, Effects of interleukin-1beta inhibition with canakinumab on hemoglobin A1c, lipids, C-reactive protein, interleukin- 6, and fibrinogen: a phase IIb randomized, placebocontrolled trial. Circulation 126(23):2739–2748. , DOI 10.1161/circulationaha.112.122556 Adam SS, Key NS, Greenberg CS, 2009, D-dimer antigen: current concepts and future prospects. Blood 113(13):2878–2887. https:// doi.org/10.1182/blood-2008-06-165845 Man YN, Wang YN, Hao J, Liu X, Liu C, Zhu C, Wu XZ, 2015, Pretreatment plasma D-dimer, fibrinogen, and platelet levels significantly impact prognosis in patients with epithelial ovarian cancer independently of venous thromboembolism. Int J Gynecol Cancer 25(1):24–32. , DOI 10.1097/igc.0000000000000303 Batschauer AP, Figueiredo CP, Bueno EC, RibeiroMA, Dusse LM, Fernandes AP, Gomes KB, Carvalho MG, 2010, D-dimer as a possible prognostic marker of operable hormone receptor-negative breast cancer. Ann Oncol 21(6):1267–1272. , DOI 10. 1093/annonc/mdp474 Yamamoto M, Yoshinaga K, Matsuyama A, Iwasa T, Osoegawa A, Tsujita E, Yamashita Y, Tsutsui S, Ishida T, 2012, Plasma D-dimer level as a mortality predictor in patients with advanced or recurrent colorectal cancer. Oncology 83(1):10–15. , DOI 10.1159/ 000338329 Fukumoto K, Taniguchi T, Usami N, Kawaguchi K, Fukui T, Ishiguro F, Nakamura S, Yokoi K, 2015, Preoperative plasma Ddimer level is an independent prognostic factor in patients with completely resected non-small cell lung cancer. Surg Today 45(1): 63–67. , DOI 10.1007/s00595-014-0894-4 Liu P, Zhu Y, Liu L, 2015, Elevated pretreatment plasma D-dimer levels and platelet counts predict poor prognosis in pancreatic adenocarcinoma. Onco Targets Ther 8:1335–1340. , DOI 10. 2147/ott.s82329 Buller HR, van Doormaal FF, van Sluis GL, Kamphuisen PW, 2007, Cancer and thrombosis: from molecular mechanisms to clinical presentations. J Thromb Haemost 5(Suppl 1):246–254. , DOI 10.1111/j.1538-7836.2007.02497.x NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1669 EP 1676 DI 10.1007/s12253-019-00752-8 PG 8 ER PT J AU Xin, BCh Wu, QSh Jin, S Luo, AH Sun, DG Wang, F AF Xin, Bing-Chang Wu, Qi-Shan Jin, Song Luo, Ai-Hua Sun, De-Gang Wang, Fang TI Berberine Promotes Osteogenic Differentiation of Human Dental Pulp Stem Cells Through Activating EGFR-MAPK-Runx2 Pathways SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Berberine; Dental pulp stem cells; Osteogenic differentiation; EGFR; MAPK ID Berberine; Dental pulp stem cells; Osteogenic differentiation; EGFR; MAPK AB Similar to the mesenchymal stem cells (MSCs), dental pulp stem cells (DPSCs) also have pluripotent differentiation characteristic and may be more ideal for tissue regeneration, especially in tooth regeneration engineering. However, bacterial infection may be a powerful obstacle. Berberine (BBR), known with antibacterial effects, was recently found to play functions in bone formation through promoting osteogenic differentiation from pluripotent stem cells. However, whether BBR also function in DPSCs osteogenic differentiation has not yet been reported. Primary DPSCs were isolated from dental pulp tissues extracted from human impacted mandibular third molars, and identified by flow cytometry for cell surface antigen molecules. A dexamethasone osteogenic medium was used to induce DPSCs osteogenic differentiation. BBR (1 μM and 5 μM) was pre-added to into medium, and then cell proliferation, spheroid formation and osteogenic differentiation capacities of DPSCs were analyzed, as well as the underlying molecules modulation mechanism. Flow cytometry identified that CD44, CD90, CD81 and CD105 positively expressed in the isolated hDPSCs, with CD34 and CD45 negatively expressed. BBR enhanced the cell proliferation of hDPSCs in a dose-dependent pattern, and promoted dexamethasone-induced osteogenic differentiation via enhancing Runx2 transcription factor activity followed by upregulating osteogenesis markers expression, whereas the adipogenic differentiation of hDPSCs was suppressed dramatically by BBR. The EGFR and MAPK pathways were activated by BBR, and inhibitors for these pathways significantly suppressed the osteogenic differentiation promotion of BBR. These results have revealed a novel mechanism that berberine might promote hDPSCs osteogenic differentiation through activating EGFR-MAPK-Runx2 signaling pathways. C1 [Xin, Bing-Chang] Qingdao Stomatological Hospital, Department of Cariology and Endodontology, 266001 Qingdao, China. [Wu, Qi-Shan] Tai’an City Central Hospital, Department of Stomatology, 271000 Taian, China. [Jin, Song] Tai’an City Central Hospital, Department of Stomatology, 271000 Taian, China. [Luo, Ai-Hua] Guizhou Provincial People’s Hospital, Department of Stomatology, 550002 Guiyang, China. [Sun, De-Gang] Qingdao Stomatological Hospital, Department of Cariology and Endodontology, 266001 Qingdao, China. [Wang, Fang] Qingdao Stomatological Hospital, Department of Pharmacy, No.17, Dexian Road, Shinan District, 266001 Qingdao, China. RP Wang, F (reprint author), Qingdao Stomatological Hospital, Department of Pharmacy, 266001 Qingdao, China. EM wangfang6525@163.com CR Gronthos S, Mankani M, Brahim J, Robey PG, Shi S, 2000, Postnatal human dental pulp stem cells, Dpscs, in vitro and in vivo. Proc Natl Acad Sci U S A 97:13625–13630 Valverde Y, Narayanan R, Alapati SB, Chmilewsky F, Huang CC, Ravindran S, Chung SH, 2018, Poly(adenosine phosphate ribose, polymerase 1 inhibition enhances brain-derived neurotrophic factor secretion in dental pulp stem cell-derived Odontoblastlike cells. J Endod 44:1121–1125 Iohara K, Nakashima M, Ito M, Ishikawa M, Nakasima A, Akamine A, 2004, Dentin regeneration by dental pulp stem cell therapy with recombinant human bone morphogenetic protein 2. J Dent Res 83:590–595 Itoh Y, Sasaki JI, Hashimoto M, Katata C, Hayashi M, Imazato S, 2018, Pulp regeneration by 3-dimensional dental pulp stem cell constructs. J Dent Res 97:1137–1143 El-Backly RM, Massoud AG, El-BadryAM, Sherif RA,MareiMK, 2008, Regeneration of dentine/pulp-like tissue using a dental pulp stem cell/poly(lactic-co-glycolic, acid scaffold construct in New Zealand white rabbits. Aust Endod J 34:52–67 de Souza PV, Alves FB, Costa Ayub CL, de Miranda Soares MA, Gomes JR, 2013, Human immature dental pulp stem cells, Hidpscs), their application to cell therapy and bioengineering: an analysis by systematic revision of the last decade of literature. Anat Rec, Hoboken, 296:1923–1928 Chopra RN, Dikshit BB, Chowhan JS, 1932, Berberine and Berberine-containing plants in pharmacology and therapeutics. Ind Med Gaz 67:194–197 Habtemariam S, 2016, Berberine and inflammatory bowel disease: a concise review. Pharmacol Res 113:592–599 Xie Q, Johnson BR, Wenckus CS, Fayad MI, Wu CD, 2012, Efficacy of Berberine, an antimicrobial plant alkaloid, as an endodontic Irrigant against a mixed-culture biofilm in an in vitro tooth model. J Endod 38:1114–1117 Wei P, Jiao L, Qin LP, Yan F, Han T, Zhang QY, 2009, Effects of Berberine on differentiation and bone resorption of osteoclasts derived from rat bone marrow cells. Zhong Xi Yi Jie He Xue Bao 7: 342–348 Lee HW, Suh JH, Kim HN, Kim AY, Park SY, Shin CS, Choi JY, Kim JB, 2008, Berberine promotes osteoblast differentiation by Runx2 activation with P38Mapk. J Bone Miner Res 23:1227–1237 Tao K, Xiao D, Weng J, Xiong A, Kang B, Zeng H, 2016, Berberine promotes bone marrow-derived mesenchymal stem cells osteogenic differentiation via canonicalWnt/Beta-catenin signaling pathway. Toxicol Lett 240:68–80 Liu J, Zhao X, Pei D, Sun G, Li Y, Zhu C, Qiang C, Sun J, Shi J, Dong Y, Gou J, Wang S, Li A, 2018, The promotion function of Berberine for osteogenic differentiation of human periodontal ligament stem cells via Erk-Fos pathway mediated by Egfr. Sci Rep 8:2848 Chang Y, 1959, Effectiveness of Berberine in bacillary dysentery. Zhonghua Nei Ke Za Zhi 7:741–743 Dong H, Zhao Y, Zhao L, Lu F, 2013, The effects of Berberine on blood lipids: a systemic review and meta-analysis of randomized controlled trials. Planta Med 79:437–446 Ma X, Chen Z,Wang L,Wang G,Wang Z, Dong X,Wen B, Zhang Z, 2018, The pathogenesis of diabetes mellitus by oxidative stress and inflammation: its inhibition by Berberine. Front Pharmacol 9: 782 Pang B, Zhao LH, Zhou Q, Zhao TY, Wang H, Gu CJ, Tong XL, 2015, Application of Berberine on treating type 2 diabetes mellitus. Int J Endocrinol 2015:905749 Wu X, Li X, Dang Z, Jia Y, 2018, Berberine demonstrates antiinflammatory properties in helicobacter pylori-infected mice with chronic gastritis by attenuating the Th17 response triggered by the B cell-activating factor. J Cell Biochem 119:5373–5381 Chuang TY,Wu HL,Min J, DiamondM, Azziz R, ChenYH(2017, Berberine regulates the protein expression of multiple tumorigenesis-related genes in hepatocellular carcinoma cell lines. Cancer Cell Int 17:59 Tsang CM, Cheung YC, Lui VW, Yip YL, Zhang G, Lin VW, Cheung KC, Feng Y, Tsao SW, 2013, Berberine suppresses Tumorigenicity and growth of nasopharyngeal carcinoma cells by inhibiting Stat3 activation induced by tumor associated fibroblasts. BMC Cancer 13:619 Zhou Y, Tao H, Li Y, Deng M, He B, Xia S, Zhang C, Liu S, 2016, Berberine promotes proliferation of sodium nitroprussidestimulated rat chondrocytes and osteoarthritic rat cartilage via Wnt/Beta-catenin pathway. Eur J Pharmacol 789:109–118 Feng G, Zhang J, Feng X, Wu S, Huang D, Hu J, Zhu S, Song D, 2016, Runx2modified dental pulp stemcells, Dpscs, enhance new bone formation during rapid distraction osteogenesis, do). Differentiation 92:195–203 Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G, 1997, Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell 89:747–754 Artigas N, Urena C, Rodriguez-Carballo E, Rosa JL, Ventura F, 2014, Mitogen-activated protein kinase, Mapk)-regulated interactions between Osterix and Runx2 are critical for the transcriptional osteogenic program. J Biol Chem 289:27105–27117 Johnson GL, Lapadat R, 2002, Mitogen-activated protein kinase pathways mediated by Erk, Jnk, and P38 protein kinases. Science 298:1911–1912 Lee KS, Hong SH, Bae SC, 2002, Both the Smad and P38 Mapk pathways play a crucial role in Runx2 expression following induction by transforming growth factor-Beta and Bone morphogenetic protein. Oncogene 21:7156–7163 Kanno T, Takahashi T, Tsujisawa T, Ariyoshi W, Nishihara T, 2007, Mechanical stress-mediated Runx2 activation is dependent on Ras/Erk1/2 Mapk signaling in osteoblasts. J Cell Biochem 101: 1266–1277 Ge C, Xiao G, Jiang D, Yang Q, Hatch NE, Roca H, Franceschi RT, 2009, Identification and functional characterization of Erk/Mapk phosphorylation sites in the Runx2 transcription factor. J Biol Chem 284:32533–32543 Huang RL, Yuan Y, Tu J, Zou GM, Li Q, 2014, Opposing Tnfalpha/ Il-1beta- and bmp-2-activatedMapk signaling pathways converge on Runx2 to regulate bmp-2-induced osteoblastic differentiation. Cell Death Dis 5:e1187 Huang YF, Lin JJ, Lin CH, Su Y, Hung SC, 2012, C-Jun N-terminal kinase 1 negatively regulates osteoblastic differentiation induced by Bmp2 via phosphorylation of Runx2 at Ser104. J Bone Miner Res 27:1093–1105 Fujii Y, Kawase-Koga Y, Hojo H, Yano F, SatoM, Chung UI, Ohba S, Chikazu D, 2018, Bone regeneration by human dental pulp stem cells using a Helioxanthin derivative and cell-sheet technology. Stem Cell Res Ther 9:24 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1677 EP 1685 DI 10.1007/s12253-019-00746-6 PG 9 ER PT J AU Ni, YH Zhang, Xx Lu, Zy Huang, XF Wang, ZY Yang, Y Dong, Ych Jing, Y Song, Y Hou, YY Hua, Zch Hu, QG AF Ni, Yan-Hong Zhang, Xiao-xin Lu, Zhan-yi Huang, Xiao-Feng Wang, Zhi-Yong Yang, Yan Dong, Ying-chun Jing, Yue Song, Yuxian Hou, Ya-Yi Hua, Zi-chun Hu, Qin-Gang TI Tumor-Infiltrating CD1a+ DCs and CD8+/FoxP3+ Ratios Served as Predictors for Clinical Outcomes in Tongue Squamous Cell Carcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Tongue squamous cell carcinoma; Tumor infiltrating immune cell; Clinical outcome; Diagnostic value ID Tongue squamous cell carcinoma; Tumor infiltrating immune cell; Clinical outcome; Diagnostic value AB Tumor-infiltrating immune cells engage in an extensive crosstalk with tumors and act as two-edged swords by inhibiting or promoting cancer growth. Therefore, identifying the density and prognostic values of tumor-infiltrating immune cells will provide valuable tips for cancer treatments. In this study, we identified the density of tumor inflammatory infiltrates and the number of tumor-infiltrating immune cells, including CD3+, CD4+, CD8+, FoxP3+ T cells and CD1a+ dendritic cells (DCs) in 153 tongue squamous cell carcinomas (TSCC). High inflammatory cell infiltration was associated with better overall survival (OS) and disease-free survival (DFS). Moreover, the number of CD3+, CD4+, FoxP3+ and CD1a+ cells were associated with tumor differentiation (P<0.001) and the number of FoxP3+, CD1a+ cells and CD8+/FoxP3+ ratios were also associated with tumor stage (P<0.01, P<0.01, P<0.05). In addition, patients with higher CD1a+ DCs had better OS and increased CD8+/FoxP3+ ratios were associated with improved OS and DFS (P = 0.037; P = 0.047; P = 0.033). In conclusion, our results indicated that tumorinfiltrating CD1a+ DCs and CD8+/FoxP3+ ratios were associated with favorable clinical outcomes but not independent prognostic factors for TSCC patients. C1 [Ni, Yan-Hong] Medical School, Nanjing University, Nanjing Stomatological Hospital, Central LaboratoryNanjing, China. [Zhang, Xiao-xin] Medical School, Nanjing University, Nanjing Stomatological Hospital, Central LaboratoryNanjing, China. [Lu, Zhan-yi] Medical School of Nanjing University, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial SurgeryNanjing, China. [Huang, Xiao-Feng] Medical School, Nanjing University, Nanjing Stomatological Hospital, Central LaboratoryNanjing, China. [Wang, Zhi-Yong] Medical School of Nanjing University, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial SurgeryNanjing, China. [Yang, Yan] Medical School, Nanjing University, Nanjing Stomatological Hospital, Central LaboratoryNanjing, China. [Dong, Ying-chun] Medical School, Nanjing University, Nanjing Stomatological Hospital, Central LaboratoryNanjing, China. [Jing, Yue] Medical School, Nanjing University, Nanjing Stomatological Hospital, Central LaboratoryNanjing, China. [Song, Yuxian] Medical School, Nanjing University, Nanjing Stomatological Hospital, Central LaboratoryNanjing, China. [Hou, Ya-Yi] Medical School, Nanjing University, Nanjing Stomatological Hospital, Central LaboratoryNanjing, China. [Hua, Zi-chun] Medical School, Nanjing University, Nanjing Stomatological Hospital, Central LaboratoryNanjing, China. [Hu, Qin-Gang] Medical School, Nanjing University, Nanjing Stomatological Hospital, Central LaboratoryNanjing, China. RP Hu, QG (reprint author), Medical School, Nanjing University, Nanjing Stomatological Hospital, Central Laboratory, Nanjing, China. CR Bello IO, Soini Y, Salo T, 2010, Prognostic evaluation of oral tongue cancer: means, markers and perspectives, I). Oral Oncol 46(9):630–635. , DOI 10.1016/j.oraloncology.2010.06. 006 Schreiber RD, Old LJ, Smyth MJ, 2011, Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science, New York, NY, 331(6024):1565–1570. , DOI 10. 1126/science.1203486 Mao Y, Qu Q, Chen X, Huang O, Wu J, Shen K, 2016, The prognostic value of tumor-infiltrating lymphocytes in breast Cancer: a systematic review and meta-analysis. PLoS One 11(4):e0152500. , DOI 10.1371/journal.pone.0152500 Hwang WT, Adams SF, Tahirovic E, Hagemann IS, Coukos G, 2012, Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol 124(2):192–198. , DOI 10.1016/j.ygyno.2011.09.039 Fang J, Li X, Ma D, Liu X, Chen Y, Wang Y, Lui VWY, Xia J, Cheng B,Wang Z, 2017, Prognostic significance of tumor infiltrating immune cells in oral squamous cell carcinoma. BMC Cancer 17(1):375. , DOI 10.1186/s12885-017-3317-2 Geng Y, Shao Y, HeW, HuW, Xu Y, Chen J,Wu C, Jiang J, 2015, Prognostic role of tumor-infiltrating lymphocytes in lung Cancer: a meta-analysis. Cell Physiol Biochem 37(4):1560–1571. https://doi. org/10.1159/000438523 Barnes TA, Amir E, 2017)HYPE or HOPE: the prognostic value of infiltrating immune cells in cancer. Br J Cancer 117(4):451–460. , DOI 10.1038/bjc.2017.220 Liotta F, Gacci M, Frosali F, Querci V, Vittori G, Lapini A, Santarlasci V, Serni S, Cosmi L, Maggi L, Angeli R, Mazzinghi B, Romagnani P, Maggi E, Carini M, Romagnani S, Annunziato F, 2011, Frequency of regulatory T cells in peripheral blood and in tumour-infiltrating lymphocytes correlates with poor prognosis in renal cell carcinoma. BJU Int 107(9):1500–1506. , DOI 10.1111/j.1464-410X.2010.09555.x Ugel S, De Sanctis F, Mandruzzato S, Bronte V, 2015, Tumorinduced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest 125(9):3365– 3376. , DOI 10.1172/JCI80006 Wallis SP, Stafford ND, Greenman J, 2015, Clinical relevance of immune parameters in the tumor microenvironment of head and neck cancers. Head Neck 37(3):449–459. , DOI 10.1002/ hed.23736 Park K, Cho KJ, Lee M, Yoon DH, Kim SB, 2013, Importance of FOXP3 in prognosis and its relationship with p16 in tonsillar squamous cell carcinoma. Anticancer Res 33(12):5667–5673 WolfGT, ChepehaDB, Bellile E,Nguyen A, ThomasD,McHugh J University of Michigan H, neck SP, 2015, tumor infiltrating lymphocytes, TIL, and prognosis in oral cavity squamous carcinoma: a preliminary study. Oral Oncol 51(1):90–95. , DOI 10. 1016/j.oraloncology.2014.09.006 de Ruiter EJ, Ooft ML, Devriese LA, Willems SM, 2017, The prognostic role of tumor infiltrating T-lymphocytes in squamous cell carcinoma of the head and neck: a systematic review and meta- analysis. Oncoimmunology 6(11):e1356148. , DOI 10. 1080/2162402X.2017.1356148 Nguyen N, Bellile E, Thomas D, McHugh J, Rozek L, Virani S, Peterson L, Carey TE, Walline H, Moyer J, Spector M, Perim D, Prince M, McLean S, Bradford CR, Taylor JM, Wolf GT, Head NSPI, 2016, Tumor infiltrating lymphocytes and survival in patients with head and neck squamous cell carcinoma. Head Neck 38(7):1074–1084. , DOI 10.1002/hed.24406 Wansom D, Light E, Thomas D, Worden F, Prince M, Urba S, Chepeha D, Kumar B, Cordell K, Eisbruch A, Taylor J, Moyer J, Bradford C, D'Silva N, Carey T, McHugh J, Wolf G, Program UMHNS, 2012, Infiltrating lymphocytes and human papillomavirus-16–associated oropharyngeal cancer. Laryngoscope 122(1):121–127. , DOI 10.1002/lary.22133 Sun DS, Zhao MQ, Xia M, Li L, Jiang YH, 2012, The correlation between tumor-infiltrating Foxp3+ regulatory T cells and cyclooxygenase-2 expression and their association with recurrence in resected head and neck cancers. Med Oncol 29(2):707–713. , DOI 10.1007/s12032-011-9903-2 Matsutani S, ShibutaniM,Maeda K, Nagahara H, Fukuoka T, Iseki Y, Kashiwagi S, Tanaka H, Hirakawa K, Ohira M, 2018, Tumorinfiltrating immune cells in H&E-stained sections of colorectal Cancer tissue as a reasonable immunological biomarker. Anticancer Res 38(12):6721–6727. , DOI 10.21873/ anticanres.13041 Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehner FL, Penault-Llorca F, Perez EA, Thompson EA, SymmansWF, Richardson AL, Brock J, Criscitiello C, Bailey H, Ignatiadis M, Floris G, Sparano J, Kos Z, Nielsen T, Rimm DL, Allison KH, Reis-Filho JS, Loibl S, Sotiriou C, Viale G, Badve S, Adams S, Willard-Gallo K, Loi S, International TWG, 2015, The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: recommendations by an international TILs working group 2014. Ann Oncol 26, 2):259–271., DOI , DOI 10.1093/annonc/mdu450 Kushwah R, Hu J, 2011, Role of dendritic cells in the induction of regulatory T cells. Cell Biosci 1(1):20. , DOI 10.1186/ 2045-3701-1-20 Ng JH, Iyer NG, Tan MH, Edgren G, 2017, Changing epidemiology of oral squamous cell carcinoma of the tongue: a global study. Head Neck 39(2):297–304. , DOI 10.1002/hed.24589 Almangush A, Bello IO, Coletta RD, Makitie AA, Makinen LK, Kauppila JH, Pukkila M, Hagstrom J, Laranne J, Soini Y, Kosma VM, Koivunen P, Kelner N, Kowalski LP, Grenman R, Leivo I, Laara E, Salo T, 2015, For early-stage oral tongue cancer, depth of invasion andworst pattern of invasion are the strongest pathological predictors for locoregional recurrence andmortality.Virchows Arch 467(1):39–46. , DOI 10.1007/s00428-015-1758-z Sopka DM, Li T, LangoMN,Mehra R, Liu JC, Burtness B, Flieder DB, Ridge JA, Galloway TJ, 2013, Dysplasia at the margin? Investigating the case for subsequent therapy in 'low-risk' squamous cell carcinoma of the oral tongue. Oral Oncol 49(11):1083–1087. , DOI 10.1016/j.oraloncology.2013.08.001 Lundqvist L, Stenlund H, Laurell G, Nylander K, 2012, The importance of stromal inflammation in squamous cell carcinoma of the tongue. J Oral Pathol Med 41(5):379–383. , DOI 10. 1111/j.1600-0714.2011.01107.x Chatzistamou I, Rodriguez J, Jouffroy T, Girod A, Point D, Sklavounou A, Kittas C, Sastre-Garau X, Klijanienko J, 2010, Prognostic significance of tumor shape and stromal chronic inflammatory infiltration in squamous cell carcinomas of the oral tongue. J Oral Pathol Med 39(9):667–671. , DOI 10.1111/j.1600- 0714.2010.00911.x Rodrigues PC, Miguel MC, Bagordakis E, Fonseca FP, de Aquino SN, Santos-Silva AR, Lopes MA, Graner E, Salo T, Kowalski LP, Coletta RD, 2014, Clinicopathological prognostic factors of oral tongue squamous cell carcinoma: a retrospective study of 202 cases. Int J Oral Maxillofac Surg 43(7):795–801. , DOI 10.1016/j.ijom.2014.01.014 Almangush A, Bello IO, Keski-Santti H, Makinen LK, Kauppila JH, Pukkila M, Hagstrom J, Laranne J, Tommola S, Nieminen O, Soini Y, Kosma VM, Koivunen P, Grenman R, Leivo I, Salo T, 2014, Depth of invasion, tumor budding, and worst pattern of invasion: prognostic indicators in early-stage oral tongue cancer. Head Neck 36(6):811–818. , DOI 10.1002/hed.23380 Gomes JO, de Vasconcelos Carvalho M, Fonseca FP, Gondak RO, LopesMA, Vargas PA, 2016, CD1a+ and CD83+ Langerhans cells are reduced in lower lip squamous cell carcinoma. J Oral Pathol Med 45(6):433–439. , DOI 10.1111/jop.12389 Yilmaz T, Gedikoglu G, Celik A, Onerci M, Turan E, 2005, Prognostic significance of Langerhans cell infiltration in cancer of the larynx. Otolaryngol Head Neck Surg 132(2):309–316. https:// doi.org/10.1016/j.otohns.2004.04.018 JardimJF, Gondak R, GalvisMM, Pinto CAL, Kowalski LP, 2018, A decreased peritumoral CD1a+ cell number predicts a worse prognosis in oral squamous cell carcinoma. Histopathology 72(6):905– 913. , DOI 10.1111/his.13415 Balermpas P, Michel Y, Wagenblast J, Seitz O, Weiss C, Rodel F, Rodel C, Fokas E, 2014, Tumour-infiltrating lymphocytes predict response to definitive chemoradiotherapy in head and neck cancer. Br J Cancer 110(2):501–509. , DOI 10.1038/bjc.2013.640 Badoual C, Hans S, Rodriguez J, Peyrard S, Klein C,Agueznay Nel H, Mosseri V, Laccourreye O, Bruneval P, Fridman WH, Brasnu DF, Tartour E, 2006, Prognostic value of tumor-infiltrating CD4+ T-cell subpopulations in head and neck cancers. Clin Cancer Res 12(2):465–472. , DOI 10.1158/1078-0432.CCR-05-1886 Watanabe Y, Katou F, Ohtani H, Nakayama T, Yoshie O, Hashimoto K, 2010, Tumor-infiltrating lymphocytes, particularly the balance between CD8(+, T cells and CCR4(+, regulatory T cells, affect the survival of patients with oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 109(5): 744–752. , DOI 10.1016/j.tripleo.2009.12.015 Chen WY, Wu CT, Wang CW, Lan KH, Liang HK, Huang BS, Chang YL, Kuo SH, Cheng AL, 2018, Prognostic significance of tumor-infiltrating lymphocytes in patients with operable tongue cancer. Radiat Oncol 13(1):157. , DOI 10.1186/s13014- 018-1099-6 Nasman A, Romanitan M, Nordfors C, Grun N, Johansson H, Hammarstedt L, Marklund L, Munck-Wikland E, Dalianis T, Ramqvist T, 2012, Tumor infiltrating CD8+ and Foxp3+ lymphocytes correlate to clinical outcome and human papillomavirus, HPV, status in tonsillar cancer. PLoS One 7(6):e38711. https:// doi.org/10.1371/journal.pone.0038711 Gooden MJ, de Bock GH, Leffers N, Daemen T, Nijman HW, 2011, The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br J Cancer 105(1):93–103. , DOI 10.1038/bjc.2011.189 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1687 EP 1695 DI 10.1007/s12253-019-00701-5 PG 9 ER PT J AU Wang, Y Jiang, W Li, C Xiong, X Guo, H Tian, Q Li, X AF Wang, Yun Jiang, Wangjie Li, Cunjiang Xiong, Xuanxuan Guo, Hao Tian, Qingzhong Li, Xiangcheng TI Autophagy Suppression Accelerates Apoptosis Induced by Norcantharidin in Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Autophagy; Norcantharidin; Cholangiocarcinoma; Apoptosis ID Autophagy; Norcantharidin; Cholangiocarcinoma; Apoptosis AB Norcantharidin is a cantharidin demethylated analog with antitumor effects in many tumors, including cholangiocarcinoma. Autophagy suppression is known to increase chemosensitivity in cholangiocarcinoma. This study aimed to determine whether autophagy suppression accelerates apoptosis induced by norcantharidin in human cholangiocarcinoma cells. The human cholangiocarcinoma cell line QBC939 was incubated in RPMI 1640 medium with or without norcantharidin. Autophagy was induced using HBSS media with Ca2+ and Mg2+ supported by 10mMHEPES or suppressed by treatment with 3-MA or transfection with siRNA against Atg5. The comparison was drawn between these conditions in mitochondrial membrane potential disturbance, the levels of reactive oxygen species (ROS), apoptotic proteins, and apoptosis. Cholangiocarcinoma cell apoptosis was accelerated by norcantharidin. Autophagy suppression up-regulated norcantharidin’s pro-apoptotic effect, but autophagy induction weakened it. As apoptosis was accelerated, ROS production was up-regulated. Bax protein expression, cytochrome c levels and localization, mitochondrial membrane disturbance, and the levels of caspase-9, caspase-3, and cleaved PARP were higher when autophagy was suppressed, and all of those were down-regulated when autophagy was induced. To sum up, it was found that norcantharidin induced cholangiocarcinoma cell death, and autophagy suppression enhanced the pro-apoptotic action of norcantharidin, which appears to involve the mitochondrial apoptosis pathway activation and ROS generation. C1 [Wang, Yun] The First Affiliated Hospital of Nanjing Medical University, Department of Liver Transplantation Center, 300 Guang Zhou Road, 210029 Nanjing, Jiangsu, China. [Jiang, Wangjie] The First Affiliated Hospital of Nanjing Medical University, Department of Liver Transplantation Center, 300 Guang Zhou Road, 210029 Nanjing, Jiangsu, China. [Li, Cunjiang] The Affiliated Hospital of the Southeast University Medical School (Xu zhou), Xuzhou City Central Hospital, Department of Thoracic Surgery, 199 Jiefang South Road, 221009 Xuzhou, Jiangsu, China. [Xiong, Xuanxuan] The Affiliated Hospital of the Southeast University Medical School (Xu zhou), Xuzhou City Central Hospital, Department of Gastroenterology 2, 199 Jiefang South Road, 221009 Xuzhou, Jiangsu, China. [Guo, Hao] The Tumor Research Institute of the Southeast University (Xu zhou), Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xu zhou), Department of Oncological Surgery, 199 Jiefang South Road, 221009 Xuzhou, Jiangsu, China. [Tian, Qingzhong] The Tumor Research Institute of the Southeast University (Xu zhou), Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xu zhou), Department of Oncological Surgery, 199 Jiefang South Road, 221009 Xuzhou, Jiangsu, China. [Li, Xiangcheng] The First Affiliated Hospital of Nanjing Medical University, Department of Liver Transplantation Center, 300 Guang Zhou Road, 210029 Nanjing, Jiangsu, China. RP Li, X (reprint author), The First Affiliated Hospital of Nanjing Medical University, Department of Liver Transplantation Center, 210029 Nanjing, China. EM drxcli@njmu.edu.cn CR Walden D, Kunnimalaiyaan S, Sokolowski K, Clark TG, Kunnimalaiyaan M, 2017, Antiproliferative and apoptotic effects of xanthohumol in cholangiocarcinoma. Oncotarget 8:88069– 88078 Guan L, Zhang L, Gong Z, Hou X, Xu Y, Feng X,Wang H, You H, 2016, FoxO3 inactivation promotes human cholangiocarcinoma tumorigenesis and chemoresistance through Keap1-Nrf2 signaling. Hepatology 63:1914–1927 Zhang H, Yang T,WuM, Shen F, 2016, Intrahepatic cholangiocarcinoma: epidemiology, risk factors, diagnosis and surgical management. Cancer Lett 379:198–205 Hu MH, Chen LJ, Chen YL, TsaiMS, Shiau CW, Chao TI, Liu CY, Kao JH, Chen KF, 2017, Targeting SHP-1-STAT3 signaling: a promising therapeutic approach for the treatment of cholangiocarcinoma. Oncotarget 8:65077–65089 Roskoski R Jr, 2017, ROS1 protein-tyrosine kinase inhibitors in the treatment of ROS1 fusion protein-driven non-small cell lung cancers. Pharmacol Res 121:202–212 ThaneeM, LoilomeW, Techasen A, Sugihara E, Okazaki S, Abe S, Ueda S, Masuko T, Namwat N, Khuntikeo N, Titapun A, Pairojkul C, Saya H, Yongvanit P, 2016, CD44 variant-dependent redox status regulation in liver fluke-associated cholangiocarcinoma: a target for cholangiocarcinoma treatment. Cancer Sci 107:991–1000 Wang X, Yu S, Jia Q, Chen L, Zhong J, Pan Y, Shen P, Shen Y, Wang S, Wei Z, Cao Y, Lu Y, 2017, NiaoDuQing granules relieve chronic kidney disease symptoms by decreasing renal fibrosis and anemia. Oncotarget 8:55920–55937 Fu H,Wu R, LiY, Zhang L, TangX, Tu J, ZhouW,Wang J, Shou Q, 2016, Safflower yellow prevents pulmonary metastasis of breast Cancer by inhibiting tumor cell Invadopodia. Am J Chin Med 44: 1491–1506 XiaoW,Wu K, YinM, Han S, Ding Y, Qiao A, Lu G, Deng B, Bo P, GongW(2015)Wogonin inhibits tumor-derived regulatory molecules by suppressing STAT3 signaling to promote tumor immunity. J Immunother 38:167–184 Chang C, Zhu YQ, Mei JJ, Liu SQ, Luo J, 2010, Involvement of mitochondrial pathway in NCTD-induced cytotoxicity in human hepG2 cells. J Exp Clin Cancer Res 29:145 HanW,Wang S, Liang R,Wang L, ChenM, Li H,Wang Y, 2013, Non-ionic surfactant vesicles simultaneously enhance antitumor activity and reduce the toxicity of cantharidin. Int J Nanomedicine 8: 2187–2196 Yeh CB, Su CJ, Hwang JM, Chou MC, 2010, Therapeutic effects of cantharidin analogues without bridging ether oxygen on human hepatocellular carcinoma cells. Eur J Med Chem 45:3981–3985 Sakahira H, Enari M, Nagata S, 2015, Corrigendum: cleavage of CAD inhibitor in CAD activation and DNA degradation during apoptosis. Nature 526:728 Rautou PE, Mansouri A, Lebrec D, Durand F, Valla D, Moreau R, 2010, Autophagy in liver diseases. J Hepatol 53:1123–1134 Moretti J, Roy S, Bozec D,Martinez J, Chapman JR, Ueberheide B et al, 2017, STING senses microbial viability to orchestrate stressmediated autophagy of the endoplasmic reticulum. Cell 171 e13: 809–823 Xiong X, Wu M, Zhang H, Li J, Lu B, Guo Y et al, 2015, Atg5 SiRNA inhibits autophagy and enhances norcantharidin-induced apoptosis in hepatocellular carcinoma. Int J Oncol 47:1321–1328 Hou YJ, Dong LW, Tan YX, Yang GZ, Pan YF, Li Z, Tang L,Wang M, Wang Q, Wang HY, 2011, Inhibition of active autophagy induces apoptosis and increases chemosensitivity in cholangiocarcinoma. Lab Investig 91:1146–1157 Settembre C, Di Malta C, Polito VA, Garcia ArencibiaM, Vetrini F, Erdin S et al, 2011, TFEB links autophagy to lysosomal biogenesis. Science 332:1429–1433 Carchman EH, Rao J, Loughran PA, Rosengart MR, Zuckerbraun BS, 2011, Heme oxygenase-1-mediated autophagy protects against hepatocyte cell death an hepatic injury from infection/sepsis in mice. Hepatology 53:2053–2062 Lewis JS, Meeke K, Osipo C, Ross EA, Kidawi N, Li T, Bell E, Chandel NS, Jordan VC, 2005, Intrinsic mechanism of estradiolinduced apoptosis in breast cancer cells resistant to estrogen deprivation. J Natl Cancer Inst 97:1746–1175 MavrosMN, Economopoulos KP, Alexiou VG, Pawlik TM, 2014, Treatment and prognosis for patients with intrahepatic cholangiocarcinoma: systematic review and meta-analysis. JAMA Surg 149: 565–574 Lau SH, Lau WY, 2012, Current therapy of hilar cholangiocarcinoma. Hepatobiliary Pancreat Dis Int 11:12–17 Estrabaud E, De Muynck S, Asselah T, 2011, Activation of unfolded protein response and autophagy during HCV infection modulates innate immune response. J Hepatol 55:1150–1153 Tai H, Wang Z, Gong H, Han X, Zhou J, Wang XN et al, 2017, Autophagy impairment with lysosomal andmitochondrial dysfunction is an important characteristic of oxidative stress-induced senescence. Autophagy 13:99–113 Toshima T, Shirabe K, Fukuhara T, Ikegami T, Yoshizumi T, Soejima Y, Ikeda T, Okano S, Maehara Y, 2014, Suppression of autophagy during liver regeneration impairs energy charge and hepatocyte senescence in mice. Hepatology 60:290–300 Whitehead NP, 2016, Enhanced autophagy as a potential mechanism for the improved physiological function by simvastatin in muscular dystrophy. Autophagy 12:705–706 Xie SQ, Zhang YH, Li Q, Xu FH, Miao JW, Zhao J, Wang CJ, 2012, 3-nitro-naphthalimide and nitrogen mustard conjugate NNM-25 induces hepatocellular carcinoma apoptosis via PARP-1/ p53 pathway. Apoptosis 17:725–734 Qi F, Inagaki Y, Gao B, Cui X, Xu H, Kokudo N et al, 2012, Bufalin and cinobufagin induce apoptosis of human hepatocellular carcinoma cells via Fas- and mitochondria-mediated pathways. Cancer Sci 102:951–958 Shen B, He PJ, Shao CL, 2013, Norcantharidin induced DU145 cell apoptosis through ROS-mediated mitochondrial dysfunction and energy depletion. PLoS One 8:e84610 Zhao L, Yang G, Bai H, ZhangM, Mou D, 2017, NCTD promotes Birinapant-mediated anticancer activity in breast cancer cells by downregulation of c-FLIP. Oncotarget 8:26886–26895 Fang EF, Scheibye-Knudsen M, Brace LE, Kassahun H, SenGupta T, Nilsen H, Mitchell JR, Croteau DL, Bohr VA, 2014, Defective mitophagy inXPAvia PARP-1 hyperactivation and NAD(+)/SIRT1 reduction. Cell 157:882–896 Choi BM, Chen XY, Gao SS, Zhu R, Kim BR, 2011, Antiapoptotic effect of phloretin on cisplatin-induced apoptosis in HEI-OC1 auditory cells. Pharmacol Rep 63:708–716 Prenek L, Boldizsar F, Kugyelka R, Ugor E, Berta G, Nemeth P, Berki T, 2017, The regulation of the mitochondrial apoptotic pathway by glucocorticoid receptor in collaboration with Bcl-2 family proteins in developing T cells. Apoptosis 22:239–253 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1697 EP 1707 DI 10.1007/s12253-019-00719-9 PG 11 ER PT J AU Kong, J Chen, X Wang, J Li, J Xu, F Gao, Sh Yu, H Qian, B AF Kong, Jinyu Chen, Xiaojie Wang, Jian Li, Jingxin Xu, Fangxiu Gao, Shegan Yu, Herbert Qian, Biyun TI Genetic Polymorphisms in the Vitamin D Pathway and Non-small Cell Lung Cancer Survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; Vitamin D pathway; Single nucleotide polymorphism; Genetic susceptibility; Prognosis ID Non-small cell lung cancer; Vitamin D pathway; Single nucleotide polymorphism; Genetic susceptibility; Prognosis AB Various genetic polymorphisms have been linked to lung cancer susceptibility and survival outcomes. Vitamin D (VD) regulates cell proliferation and differentiation, inhibits tumor growth and induces apoptosis. Observations from several previous studies including our own suggest that genetic polymorphisms in the VD pathway may be associated with lung cancer risk. The aim of this study is to assess if genetic polymorphisms in the VD pathway are associated with the prognosis of non-small cell lung cancer (NSCLC). Nine single nucleotide polymorphisms (SNPs) in five genes in the VD pathway were genotyped with the TaqMan assays in 542 patients with primary NSCLC, and the relationships between these SNPs and overall survival were evaluated. We found that SNP rs10741657 in the CYP2R1 gene was associated with the prognosis of NSCLC, especially in elderly patients and not being treated with chemotherapy. Some of the VD pathway-related genetic polymorphisms may influence the prognosis of NSCLC. More research is needed to further confirm the finding and test if VD supplements can be used for NSCLC treatment. C1 [Kong, Jinyu] The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Cancer Institute, 471003 Luoyang, China. [Chen, Xiaojie] Henan University of Science and Technology, The First Affiliated Hospital, and College of Clinical Medicine, 471003 Luoyang, Henan, China. [Wang, Jian] Henan University of Science and Technology, The First Affiliated Hospital, and College of Clinical Medicine, Department of Image Diagnoses, 471003 Luoyang, China. [Li, Jingxin] Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Prevention and Therapy, 300060 Tianjin, China. [Xu, Fangxiu] Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Prevention and Therapy, 300060 Tianjin, China. [Gao, Shegan] The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Cancer Institute, 471003 Luoyang, China. [Yu, Herbert] University of Hawaii Cancer Center, Epidemiology Program, 701 Ilalo Street, 96813 Honolulu, HI, USA. [Qian, Biyun] Shanghai Jiao Tong University School of Medicine, Shanghai Tongren Hospital and Faculty of Public Health, Hongqiao International Institute of Medicine, 227 South Chongqing Road, 200025 Shanghai, China. RP Qian, B (reprint author), Shanghai Jiao Tong University School of Medicine, Shanghai Tongren Hospital and Faculty of Public Health, Hongqiao International Institute of Medicine, 200025 Shanghai, China. EM qianbiyun@shsmu.edu.cn CR Spiro SG, Silvestri GA, 2005, One hundred years of lung cancer. Am J Respir Crit Care Med 172:523–529. , DOI 10.1164/ rccm.200504-531OE Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D, 2011, Global cancer statistics. CA Cancer J Clin 61:69–90. https://doi. org/10.3322/caac.20107 Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics,2013. CA Cancer J Clin 63:11–30. , DOI 10.3322/caac.21166 Borczuk AC, Gorenstein L, Walter KL, Assaad AA, Wang L, Powell CA, 2003, Non-small cell lung cancer molecular signatures recapitulate lung developmental pathways. Am J Pathol 163:1949– 1960. , DOI 10.1016/S0002-9440(10)63553-5 Nakagawa K, Kawaura A, Kato S, Takeda E, Okano T, 2005, 1 alpha,25-Dihydroxyvitamin D(3, is a preventive factor in the metastasis of lung cancer. Carcinogenesis 26:429–440. , DOI 10.1093/carcin/bgh332 Bouillon R, Eelen G, Verlinden L, Mathieu C, Carmeliet G, Verstuyf A, 2006, Vitamin D and cancer. J Steroid Biochem Mol Biol 102:156–162. , DOI 10.1016/j.jsbmb.2006.09.014 Ordonez-Moran P, Larriba MJ, Pendas-Franco N, Aguilera O, Gonzalez-Sancho JM, Munoz A, 2005, Vitamin D and cancer: an update of in vitro and in vivo data. Front Biosci 10:2723–2749 ZhouW, Heist RS, Liu G et al, 2006, Polymorphisms of vitamin D receptor and survival in early-stage non-small cell lung cancer patients. Cancer Epidemiol Biomark Prev 15:2239–2245. https://doi. org/10.1158/1055-9965.EPI-06-0023 AliMM, VaidyaV, 2007, Vitamin D and cancer. J Cancer Res Ther 3:225–230. , DOI 10.4103/0973-1482.87006 Kong J, Xu F, Qu J,Wang Y, GaoM, Yu H, Qian B, 2015, Genetic polymorphisms in the vitamin D pathway in relation to lung cancer risk and survival. Oncotarget 6:2573–2582. , DOI 10. 18632/oncotarget.2951 Dong J, Hu Z, Wu C et al, 2012, Association analyses identify multiple new lung cancer susceptibility loci and their interactions with smoking in the Chinese population. Nat Genet 44:895–899. , DOI 10.1038/ng.2351 Ramos-Lopez E, Bruck P, Jansen T, Herwig J, Badenhoop K, 2007, CYP2R1, vitamin D 25-hydroxylase, gene is associated with susceptibility to type 1 diabetes and vitamin D levels in Germans. Diabetes Metab Res Rev 23:631–636. , DOI 10.1002/ dmrr.719 Slater NA, Rager ML, Havrda DE, Harralson AF, 2017, Genetic variation in CYP2R1 and GC genes associated with V itamin D deficiency status. J Pharm Pract 30:31–36. , DOI 10. 1177/0897190015585876 Wang TJ, Zhang F, Richards JB et al, 2010, Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet 376:180–188. , DOI 10.1016/S0140- 6736(10)60588-0 Bu FX, Armas L, Lappe J, Zhou Y, Gao G, Wang HW, Recker R, Zhao LJ, 2010, Comprehensive association analysis of nine candidate genes with serum 25-hydroxy vitamin D levels among healthy Caucasian subjects. Hum Genet 128:549–556. , DOI 10. 1007/s00439-010-0881-9 Lasky-Su J, Lange N, Brehm JM et al, 2012, Genome-wide association analysis of circulating vitamin D levels in children with asthma. Hum Genet 131:1495–1505. , DOI 10.1007/ s00439-012-1185-z Anic GM, Thompson RC, Nabors LB et al, 2012, An exploratory analysis of common genetic variants in the vitamin D pathway including genome-wide associated variants in relation to glioma risk and outcome. Cancer Causes Control 23:1443–1449. https:// doi.org/10.1007/s10552-012-0018-7 Anderson LN, Cotterchio M, Knight JA, Borgida A, Gallinger S, Cleary SP, 2013, Genetic variants in vitamin d pathway genes and risk of pancreas cancer; results from a population-based case-control study in Ontario, Canada. PLoS One 8:e66768. , DOI 10.1371/journal.pone.0066768 Cheng JB, Levine MA, Bell NH, Mangelsdorf DJ, Russell DW, 2004, Genetic evidence that the human CYP2R1 enzyme is a key vitamin D 25-hydroxylase. Proc Natl Acad Sci U S A 101: 7711–7715. , DOI 10.1073/pnas.0402490101 Nissen J, Rasmussen LB, Ravn-Haren G et al, 2014, Common variants in CYP2R1 and GC genes predict vitamin D concentrations in healthy Danish children and adults. PLoS One 9:e89907. , DOI 10.1371/journal.pone.0089907 Mohr SB, Garland CF, Gorham ED, Grant WB, Garland FC, 2008, Could ultraviolet B irradiance and vitamin D be associated with lower incidence rates of lung cancer? J Epidemiol Community Health 62:69–74. , DOI 10.1136/jech.2006.052571 Porojnicu AC, Robsahm TE, Dahlback A, Berg JP, Christiani D, Bruland OS, Moan J, 2007, Seasonal and geographical variations in lung cancer prognosis in Norway. Does vitamin D from the sun play a role? Lung Cancer 55:263–270. , DOI 10.1016/j. lungcan.2006.11.013 Lim HS, Roychoudhuri R, Peto J, Schwartz G, Baade P, Moller H, 2006, Cancer survival is dependent on season of diagnosis and sunlight exposure. Int J Cancer 119:1530–1536. , DOI 10. 1002/ijc.22052 Zhou W, Suk R, Liu G et al, 2005, Vitamin D is associated with improved survival in early-stage non-small cell lung cancer patients. Cancer Epidemiol Biomark Prev 14:2303–2309. https://doi. org/10.1158/1055-9965.EPI-05-0335 Zhou W, Heist RS, Liu G et al, 2007, Circulating 25- hydroxyvitamin D levels predict survival in early-stage nonsmall- cell lung cancer patients. J Clin Oncol 25:479–485. https:// doi.org/10.1200/JCO.2006.07.5358 Tretli S, Schwartz GG, Torjesen PA, Robsahm TE, 2012, Serum levels of 25-hydroxyvitamin D and survival in Norwegian patients with cancer of breast, colon, lung, and lymphoma: a populationbased study. Cancer Causes Control 23:363–370. , DOI 10. 1007/s10552-011-9885-6 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1709 EP 1715 DI 10.1007/s12253-019-00702-4 PG 7 ER PT J AU Cserni, D Zombori, T Stajer, A Rimovszki, A Cserni, G Barath, Z AF Cserni, Dorottya Zombori, Tamas Stajer, Anette Rimovszki, Annamaria Cserni, Gabor Barath, Zoltan TI Immunohistochemical Characterization of Reactive Epithelial Changes in Odontogenic Keratocysts SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Odontogenic keratocyst; Immunohistochemistry; bcl2; Cytokeratin 10; Cytokeratin 17; Cytokeratin 19 ID Odontogenic keratocyst; Immunohistochemistry; bcl2; Cytokeratin 10; Cytokeratin 17; Cytokeratin 19 AB Odontogenic keratocysts (OKCs) have a diagnostic thin epithelial lining characterised by a linear epithelial connective tissue interface generally lacking inflammatory changes, basal palisading of the nuclei and a wavy parakeratotic layer on the surface. This typical epithelium may convert to a thicker non-keratinizing one with rete pegs and a relatively flat surface after operative decompression. The aim was to characterize this type of epithelial change by immunohistochemistry for bcl2, keratin17, 10 and 19. Eleven out of 33 archived OKCs demonstrated an altered epithelium related to previous biopsy, decompressing drainage or inflammation. The typical basal bcl2 staining was lost in 10/11 cases; transepithelial CK17 was lost or markedly reduced in 9/11 cases. CK10 displayed a segmental upper layer staining in OKCs, and its loss or partial loss in the altered epithelium did not differ from negative areas of OKCs. CK19 displayed various staining patterns in the altered epithelium from lost to maintained in a patchy transepithelial distribution, the latter of which did not differ from the typical OKC staining pattern. Three of four nonkeratinizing epithelial linings with basal palisading displayed immunostaining reminiscent of typical OKC epithelium. The lack of a typical epithelium is not sufficient to exclude the diagnosis of OKC if the sampling is not generous (e.g. biopsy), and the presence of non-keratinizing epithelium with basal palisading and an immunophenotype characteristic of OKC (basal bcl2, patchy or diffuse CK17 and upper layer CK10 positivity) may be consistent with the OKC diagnosis even in the absence of typical epithelial lining. C1 [Cserni, Dorottya] University of Szeged, Faculty of Dentistry, Department of Prosthodontics, Tisza Lajos krt 64-66, H-6720 Szeged, Hungary. [Zombori, Tamas] University of Szeged, Department of Pathology, Allomas u. 1, H-6725 Szeged, Hungary. [Stajer, Anette] University of Szeged, Faculty of Dentistry, Department of Prosthodontics, Tisza Lajos krt 64-66, H-6720 Szeged, Hungary. [Rimovszki, Annamaria] Bacs-Kiskun County Hospital, Department of Pathology, Nyiri ut 38, H-6000 Kecskemet, Hungary. [Cserni, Gabor] University of Szeged, Department of Pathology, Allomas u. 1, H-6725 Szeged, Hungary. [Barath, Zoltan] University of Szeged, Faculty of Dentistry, Department of Prosthodontics, Tisza Lajos krt 64-66, H-6720 Szeged, Hungary. RP Cserni, G (reprint author), University of Szeged, Department of Pathology, H-6725 Szeged, Hungary. EM cserni@freemail.hu CR Pogrel MA, 2015, Keratocystic odontogenic tumour, KCOT, - an odyssey. Int J Oral Maxillofac Surg 44:1565–1568 Gorlin RJ, Goltz RW, 1960, Multiple nevoid basal-cell epithelioma, jaw cysts and bifid rib. A syndrome. N Engl J Med 262:908–912 El-Naggar AK, Chan JKC, Grandis JR, Takata T, Slootweg PJ, 2017, WHO classification of head and neck Tumours, 4th edn. International Agency for Research on Cancer, Lyon Barnes L, Eveson JW, Reichart P, Sidransky D, 2005, WHO classification of tumors: pathology and genetics of head and neck tumours, 3rd edn. International Agency for Research on Cancer, Lyon Diniz MG, Gomes CC, de Sousa SF, Xavier GM, Gomez RS, 2017, Oncogenic signalling pathways in benign odontogenic cysts and tumours. Oral Oncol 72:165–173 Zhang L, Chen XM, Sun ZJ, Bian Z, Fan MW, Chen Z, 2006, Epithelial expression of SHH signaling pathway in odontogenic tumors. Oral Oncol 42:398–408 Zhang L, Sun ZJ, Chen XM, Chen Z, 2010, Immunohistochemical expression of SHH, PTC, SMO and GLI1 in glandular odontogenic cysts and dentigerous cysts. Oral Dis 16:818–822 Speight PM, Takata T, 2017, New tumour entities in the 4th edition of the World Health Organisation classification of head and neck tumours: odontogenic and maxillofacial bone tumours. Virchows Arch 472:331–339 Booms P, Harth M, Sader R, Ghanaati S, 2015, Vismodegib hedgehog-signaling inhibition and treatment of basal cell carcinomas as well as keratocystic odontogenic tumors in Gorlin syndrome. Ann Maxillofac Surg 5:14–19 Menon S, 2015, Keratocystic odontogenic tumours: etiology, pathogenesis and treatment revisited. J Maxillofac Oral Surg 14:541–547 Sharif FNJ, Oliver R, Sweet C, Sharif MO, 2015, Interventions for the treatment of keratocystic odontogenic tumours. Cochrane Database Syst Rev, 11):CD008464 Vered M, Peleg O, Taicher S, BuchnerA(2009, The immunoprofile of odontogenic keratocyst, keratotic odontogenic tumor, that includes expression of PTCH, SMO, GLI-1 and bcl2 is similar to ameloblastoma but different from odontogenic cysts. J Oral Pathol Med 38:597–604 Regl G, Kasper M, Schnidar H, Eichberger T, Neill GW, Philpott MP, Esterbauer H, Hauser-Kronberger C, Frischauf AM, Aberger F, 2004, Activation of the BCL2 promoter in response to hedgehog/ GLI signal transduction is predominantly mediated by GLI2. Cancer Res 64:7724–7731 Aragaki T, Michi Y, Katsube K, Uzawa N, Okada N, Akashi T, Amagasa T, Yamaguchi A, Sakamoto K, 2010, Comprehensive keratin profiling reveals different histopathogenesis of keratocystic odontogenic tumor and orthokeratinized odontogenic cyst. Hum Pathol 41:1718–1725 Tsuji K,Wato M, Hayashi T, Yasuda N,Matsishita T, Ito T, Gamoh S, Yoshida H, Tanaka A, Morita S, 2014, The expression of cytokeratin in keratocystic odontogenic tumor, orthokeratinized odontogenic cyst, dentigerous cyst, radicular cyst and dermoid cyst. Med Mol Morphol 47:156–161 Kamath KP, VidyaM, 2015, Cytokeratin 19 expression patterns of dentigerous cysts and odontogenic keratocysts. Ann Med Health Sci Res 5:119–123 Marker P, BrondumN, Clausen PP, BastianHL, 1996, Treatment of large odontogenic keratocysts by decompression and later cystectomy: a long-term follow-up and a histologic study of 23 cases. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 82: 122–131 Oh JS, You JS, Kim SG, 2018, Clinical and histomorphometric evaluation of decompression followed by enucleation in the treatment of odontogenic keratocyst. J Dent Sci 13:329–333 August M, Faquin WC, Troulis M, Kaban LB, 2000, Differentiation of odontogenic keratocysts from nonkeratinizing cysts by use of fine-needle aspiration biopsy and cytokeratin-10 staining. J Oral Maxillofac Surg 58:935–940 August M, Faquin WC, Troulis M, Kaban LB, 2003, Dedifferentiation of odontogenic keratocyst epithelium after decompression. J Oral Maxillofac Surg 61:678–683 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1717 EP 1724 DI 10.1007/s12253-019-00749-3 PG 8 ER PT J AU Li, J Duan, H Xuan, F Zhao, E Huang, M AF Li, Jia Duan, Haoyan Xuan, Fan Zhao, Erhu Huang, Mengying TI CDGSH Iron Sulfur Domain 2 Deficiency Inhibits Cell Proliferation and Induces Cell Differentiation of Neuroblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CDGSH iron sulfur domain 2; Neuroblastoma; Cell proliferation; Tumorigenesis; Differentiation ID CDGSH iron sulfur domain 2; Neuroblastoma; Cell proliferation; Tumorigenesis; Differentiation AB CDGSH iron sulfur domain 2 (CISD2) is reported to be highly expressed in several cancers, but the role of it in neuroblastoma has not been identified yet. Here, for the first time, we show that CISD2 is involved in neuroblastoma tumorigenesis and regulates neuroblastoma cell proliferation and differentiation. We found that high CISD2 expression correlated significantly with poor outcome of neuroblastoma patients, as well as advanced neuroblastoma tumor stages. Knockdown of CISD2 greatly repressed neuroblastoma cell proliferation and tumorigenesis both in vitro and in vivo. Further investigation showed that CISD2 deficiency resulted in cell cycle arrest in G1 phase and induced cell differentiation of neuroblastoma. Several Cyclins and Cyclin-Dependent Kinases (CDKs) were down-regulated by CISD2 knockdown, indicating that CISD2 probably regulates cell cycle through those genes. Together, we provide evidence that CISD2 is an indicator for neuroblastoma patients prognosis and is indispensable for neuroblastoma cell proliferation and tumorigenesis; CISD2 deficiency can induce neuroblastoma cell cycle arrest and differentiation. These findings suggest that CISD2 could work as a novel and potential therapeutic target for neuroblastoma treatment. C1 [Li, Jia] Southwest University, State Key Laboratory of Silkworm Genome Biology, 400716 Chongqing, China. [Duan, Haoyan] Southwest University, State Key Laboratory of Silkworm Genome Biology, 400716 Chongqing, China. [Xuan, Fan] Southwest University, State Key Laboratory of Silkworm Genome Biology, 400716 Chongqing, China. [Zhao, Erhu] Southwest University, State Key Laboratory of Silkworm Genome Biology, 400716 Chongqing, China. [Huang, Mengying] Southwest University, State Key Laboratory of Silkworm Genome Biology, 400716 Chongqing, China. RP Huang, M (reprint author), Southwest University, State Key Laboratory of Silkworm Genome Biology, 400716 Chongqing, China. EM mengyingh90@gmail.com CR Brodeur GM, 2003, Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer 3(3):203–216 Takeuchi LA, Hachitanda Y,WoodsWG, TuchmanM, Lemieux B, Brisson L, Bernstein M, Brossard J, Leclerc JM, Byrne TD et al, 1995, Screening for neuroblastoma in North America. Preliminary results of a pathology review from the Quebec project. Cancer 76(11):2363–2371 Anderson DJ, 1997, Cellular and molecular biology of neural crest cell lineage determination. Trend Genet: TIG 13(7):276–280 Nishi M, Miyake H, Takeda T, Yonemori H, Hanai J, Kikuchi Y, Takasugi N, 1994, A trial to discriminate spontaneous regression from non-regression cases during mass screening for neuroblastoma. Jpn J Clin Oncol 24(5):247–251 Negroni A, Scarpa S, Romeo A, Ferrari S,Modesti A, Raschella G, 1991, Decrease of proliferation rate and induction of differentiation by a Mycn antisense DNA oligomer in a human Neuroblastoma cell-line. Cell Growth Differ 2(10):511–518 Manohar CF, Salwen HR, Furtado MR, Cohn SL, 1996, Upregulation of HOXC6, HOXD1, and HOXD8 homeobox gene expression in human neuroblastoma cells following chemical induction of differentiation. Tumour Biol 17(1):34–47 Zhang X, Hamada J, Nishimoto A, Takahashi Y, Murai T, Tada M, Moriuchi T, 2007, HOXC6 and HOXC11 increase transcription of S100beta gene in BrdU-induced in vitro differentiation of GOTO neuroblastoma cells into Schwannian cells. J Cell Mol Med 11(2): 299–306 Conlan AR, Axelrod HL, Cohen AE, Abresch EC, Zuris J, Yee D, Nechushtai R, Jennings PA, Paddock ML, 2009, Crystal structure of Miner1: the redox-active 2Fe-2S protein causative in Wolfram syndrome 2. J Mol Biol 392(1):143–153 PaddockML,Wiley SE, Axelrod HL, Cohen AE, RoyM, Abresch EC, Capraro D, Murphy AN, Nechushtai R, Dixon JE, Jennings PA, 2007, MitoNEET is a uniquely folded 2Fe 2S outer mitochondrial membrane protein stabilized by pioglitazone. Proc Natl Acad Sci U S A 104(36):14342–14347 Chen YF, Kao CH, Kirby R, Tsai TF, 2009, Cisd2 mediates mitochondrial integrity and life span in mammals. Autophagy 5(7): 1043–1045 Wu CY, Chen YF, Wang CH, Kao CH, Zhuang HW, Chen CC, Chen LK, Kirby R, Wei YH, Tsai SF, Tsai TF, 2012, A persistent level of Cisd2 extends healthy lifespan and delays aging in mice. Hum Mol Genet 21(18):3956–3968 Liu L, Xia M, Wang J, Zhang W, Zhang Y, He M, 2014, CISD2 expression is a novel marker correlating with pelvic lymph node metastasis and prognosis in patients with early-stage cervical cancer. Med Oncol 31(9):183 Chen B, Shen S, Wu J, Hua Y, Kuang M, Li S, Peng B, 2015, CISD2 associated with proliferation indicates negative prognosis in patients with hepatocellular carcinoma. Int J Clin Exp Pathol 8(10):13725–13738 Yang L, Hong S, Wang Y, He Z, Liang S, Chen H, He S, Wu S, Song L, Chen Y, 2016, A novel prognostic score model incorporatingCDGSH iron sulfur domain2, CISD2, predicts risk of disease progression in laryngeal squamous cell carcinoma. Oncotarget 7(16):22720–22732 Sohn YS, Tamir S, Song L, Michaeli D, Matouk I, Conlan AR, Harir Y, Holt SH, Shulaev V, Paddock ML, Hochberg A, Cabanchick IZ, Onuchic JN, Jennings PA, Nechushtai R, Mittler R, 2013, NAF-1 and mitoNEET are central to human breast cancer proliferation by maintaining mitochondrial homeostasis and promoting tumor growth. Proc Natl Acad Sci U S A 110(36):14676– 14681 Brodeur GM, Pritchard J, Berthold F, Carlsen NL, Castel V, Castelberry RP, De Bernardi B, Evans AE, Favrot M, Hedborg F et al, 1994, Revisions of the international criteria for neuroblastoma diagnosis, staging and response to treatment. Prog Clin Biol Res 385:363–369 SchmidtML, Lal A, Seeger RC,Maris JM, Shimada H, O'LearyM, Gerbing RB, Matthay KK, 2005, Favorable prognosis for patients 12 to 18 months of age with stage 4 nonamplified MYCN neuroblastoma: a Children's Cancer group study. J Clin Oncol 23(27): 6474–6480 Fredlund E, Ringner M, Maris JM, Pahlman S, 2008, High Myc pathway activity and low stage of neuronal differentiation associate with poor outcome in neuroblastoma. Proc Natl Acad Sci U S A 105(37):14094–14099 Mao L, Ding J, Zha YH, Yang LQ,McCarthy BA, KingW, Cui HJ, Ding HF, 2011, HOXC9 links cell-cycle exit and neuronal differentiation and is a prognostic marker in Neuroblastoma. Cancer Res 71(12):4314–4324 Sihag RK, Jaffe H, Nixon RA, Rong XH, 1999, Serine-23 is a major protein kinase a phosphorylation site on the amino-terminal head domain of the middle molecular mass subunit of neurofilament proteins. J Neurochem 72(2):491–499 Chang L, Goldman RD, 2004, Intermediate filaments mediate cytoskeletal crosstalk. Nature reviews. Mol Cell Biol 5(8):601–613 Eriksson KS, Zhang SW, Lin L, Lariviere RC, Julien JP, Mignot E, 2008, The type III neurofilament peripherin is expressed in the tuberomammillary neurons of the mouse. BMC Neurosci 9 23. Kageyama R, Ishibashi M, Takebayashi K, Tomita K, 1997, bHLH transcription factors and mammalian neuronal differentiation. Int J Biochem Cell Biol 29(12):1389–1399 24. Tamir S, Zuris JA, Agranat L, Lipper CH, Conlan AR,Michaeli D, Harir Y, Paddock ML, Mittler R, Cabantchik ZI, Jennings PA, Nechushtai R, 2013, Nutrient-deprivation autophagy factor-1, NAF-1): biochemical properties of a novel cellular target for antidiabetic drugs. PLoS One 8(5):e61202 25. Huang MY, Xuan F, Liu W, Cui HJ, 2017, MINA controls proliferation and tumorigenesis of glioblastoma by epigenetically regulating cyclins and CDKs via H3K9me3 demethylation. Oncogene 36(3):387–396 26. Murray AW, 1992, Creative blocks - cell-cycle checkpoints and feedback controls. Nature 359(6396):599–604 27. Evans AE, D'Angio GJ, Randolph J, 1971, A proposed staging for children with neuroblastoma. Children's cancer study group a. Cancer 27(2):374–378 28. Haas D, Ablin AR, Miller C, Zoger S, Matthay KK, 1988, Complete pathologic maturation and regression of stage IVS neuroblastoma without treatment. Cancer 62(4):818–825 29. Westermark UK, Wilhelm M, Frenzel A, Henriksson MA, 2011, The MYCN oncogene and differentiation in neuroblastoma. Semin Cancer Biol 21(4):256–266 30. Xuan F, Huang M, Liu W, Ding H, Yang L, Cui H, 2016, Homeobox C9 suppresses Beclin1-mediated autophagy in glioblastoma by directly inhibiting the transcription of death-associated protein kinase 1. Neuro-Oncology 18(6):819–829 31. Yang L, Huang M, Tan J, Hou J, He J,Wang F, Cui H, Yi L, 2017, Transcriptional co-activator with PDZ-binding motif overexpression promotes cell proliferation and transcriptional co-activator with PDZ-binding motif deficiency induces cell cycle arrest in neuroblastoma. Oncol Lett 13(6):4295–4301 32. Xuan F, Huang M, Zhao E, Cui H, 2018)MINA53 deficiency leads to glioblastoma cell apoptosis via inducing DNA replication stress and diminishingDNA damage response. Cell Death Dis 9(11):1062 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1725 EP 1733 DI 10.1007/s12253-019-00753-7 PG 9 ER PT J AU Krishnamurthy, K Sriganeshan, V AF Krishnamurthy, Kritika Sriganeshan, Vathany TI Pancreatic Intraepithelial Neoplasia (PanIN) as a Morphologic Marker of Pancreatobiliary Type of Ampullary Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pancreatic intraepithelial neoplasia (pan-IN); Ampullary adenocarcinoma and its sub-types; Pancreatic adenocarcinoma; CDX2 ID Pancreatic intraepithelial neoplasia (pan-IN); Ampullary adenocarcinoma and its sub-types; Pancreatic adenocarcinoma; CDX2 AB The classification of ampullary adenocarcinoma into intestinal and pancreatobiliary sub-types has been found to be important in predicting prognosis and determining therapeutic strategy. Due to considerable inter-observer variability in sub-typing based solely on morphology, higher frequency of poorly differentiated cancers and low incidence of the disease, the histomorphologic classification of ampullary adenocarcinoma remains one of the grey zones in surgical pathology. Pan-IN is a well recognized precursor to pancreatic adenocarcinoma. Three studies have shown concurrent Pan-IN in patients with ampullary carcinoma, but their association with the two sub-types has not yet been reported. Fourteen cases of surgical resection for ampullary adenocarcinoma were retrieved from the archives. The cases were classified into two groups based on the presence or absence of concomitant Pan-IN. All the cases were stained for CK7, CK 20, Villin and CDX 2 and were classified as intestinal or pancreatobiliary types based on the staining pattern. All the 10 cases with Pan-IN stained negative for CDX2 and were classified as pancreatobiliary type (p = 0.01).Of the cases without Pan-IN, 3 were classified as intestinal sub-type based on morphology and CDX2 positivity and 1 was classified as pancreatobiliary type. Concomitant Pan-IN was present in 91% of pancreatobiliary type of ampullary adenocarcinoma. The grade of Pan-IN did not influence the grade or stage of the adenocarcinoma (p > 0.05). The co-occurrence of Pan-IN in a high percentage of the pancreatobiliary sub-type and its complete absence in the intestinal sub-type may serve as a strong differentiator between the two sub-types. C1 [Krishnamurthy, Kritika] Mount Sinai Medical Center, Arkadi M Rywlin Department of Pathology and Laboratory Medicine, 4300Alton road, Suite 2400, 33140 Miami Beach, FL, USA. [Sriganeshan, Vathany] Mount Sinai Medical Center, Arkadi M Rywlin Department of Pathology and Laboratory Medicine, 4300Alton road, Suite 2400, 33140 Miami Beach, FL, USA. RP Krishnamurthy, K (reprint author), Mount Sinai Medical Center, Arkadi M Rywlin Department of Pathology and Laboratory Medicine, 33140 Miami Beach, USA. EM kritikakrishnamurthy@yahoo.com CR Albores-Saavedra J, Schwartz AM, Batich K, Henson DE, 2009, Cancers of the ampulla of vater: demographics, morphology, and survival based on 5,625 cases from the SEER program. J Surg Oncol 100:598–605 Benhamiche AM, Jouve JL, Manfredi S, Prost P, Isambert N et al, 2000, Cancer of the ampulla of Vater: results of a 20-year population- based study. Eur J Gastroenterol Hepatol 12:75–79 Winter JM, Cameron JL, Olino K, Herman JM, Jong MC et al, 2010, Clinicopathologic analysis of ampullary neoplasms in 450 patients: implications for surgical strategy and long-term prognosis. J Gastrointest Surg 14:379–387 Kimura W, Futakawa N, Yamagata S, Wada Y, Kuroda A, Muto T, Esaki Y, 1994, Different clinicopathologic findings in two histologic types of carcinoma of papilla ofVater. Jpn J Cancer Res 85(2): 161–166 Bronsert P, Kohler I,Werner M, Makowiec F, Kuesters S, Hoeppner J, Hopt UT, Keck T, Bausch D, Wellner UF, 2013, Intestinal-type of differentiation predicts favourable overall survival: confirmatory clinicopathological analysis of 198 periampullary adenocarcinomas of pancreatic, biliary, ampullary and duodenal origin. BMC Cancer 13:428 Ruemmele P, Dietmaier W, Terracciano L, Tornillo L, Bataille F et al, 2009, Histopathologic features and microsatellite instability of cancers of the papilla of Vater and their precursor lesions. Am J Surg Pathol 33:691–704 Albores-Saavedra J, Henson DE, Klimstra DS, 2000, Malignant tumors of the ampulla. In:Tumors of the gallbladder extrahepatic bile ducts and ampulla of vater Armed Forces Institute of Pathology. Washington D.C, 259–316 Westgaard A, Tafjord S, Farstad IN, Cvancarova M, Eide TJ, Mathisen O, Clausen OPF, Gladhaug IP, 2008, Pancreatobiliary versus intestinal histologic type of differentiation is an independent prognostic factor in resected periampullary adenocarcinoma. BMCCancer 8:170 Rosa VDL, Peria FM, Brunaldi MO, 2017, Carcinoma of ampulla of Vater: carcinogenesis and Immunophenotypic evaluation. J Clin Epigenet 3(2):25 Ang DC, Shia J, Tang LH, 2014, The utility of immunohistochemistry in subtyping adenocarcinoma of the ampulla of Vater. Am J Surg Pathol 38:1371–1379 Chang DK, Jamieson NB, Johns AL, 2013, Histomolecular phenotypes and outcome in adenocarcinoma of the ampulla of Vater. J Clin Oncol 31:1348–1356 Liu TH, Chen J, Zeng XJ, 1983, Histogenesis of pancreatic head and ampullary region carcinoma. Chin Med J 96:167–174 Agoff SN, Crispin DA, Bronner MP, Dail DH, Hawes SE et al, 2001, Neoplasms of the ampulla of Vater with concurrent pancreatic intraductal neoplasia: a histological and molecular study. Mod Pathol 14:139–143 Cattel RB, Pyrtek LJ, 1949, An appraisal of pancreatoduodenal resection: a follow-up study of 61 cases. Ann Surg 129:840–848 Kimura W, Futakawa N, Zhao B, 2004, Neoplastic diseases of the papilla of Vater. J Hepato-Biliary-Pancreat Surg 11:223–231 Zhou H, Schaefer N, Wolff M, Fischer H, 2004, Carcinoma of the ampulla of Vater: comparative histologic/immunohistochemical classification and follow-up. Am J Surg Pathol 28:875–882 Fischer HP, Zhou H, 2000, Pathologie der Papilla Vateri. In: Doerr W, Seifert G, Uehlinger E, eds, Spezielle Pathologische Anatomie: Pathologie der Leber und der Gallenwege, 2nd edn. Springer, Berlin Heidelberg New York Tokyo, pp 1219–1257 Sessa F, Furlan D, Zampatti C, Carnevali I, Franzi F, Capella C, 2007, Prognostic factors for ampullary adenocarcinomas: tumor stage, tumor histology, tumor location, immunohistochemistry and microsatellite instability. Virchows Arch 451:649–657 Schueneman A, Wang H, Soifer HS, Schnabel CA, Wolff RA, Varadhachary GR, Overman MJ, 2014, Molecular classification of ampullary adenocarcinoma: comparative prognostic performance of the 92-gene assay versus histomolecular analysis. J Clin Oncol 32(15):4141–4141 Perkins G, Svrcek M, Bouchet-Doumenq C, Voron T, Colussi O, Debove C, et al., 2019, Can we classify ampullary tumours better? Clinical, pathological and molecularfeatures. Results of an AGEO study. Br J Cancer [Epub ahead of print] Jeong BK, Sung YN, Kim SJ, An S, Park H, Hwang HS, Kang HJ, Lee JH, Song KB, Kim KP, Hwang DW, Lee SS, Kim SC, Hong SM, 2019, High-grade precursor lesions can be used as surrogate markers to identify the epicenter of periampullary carcinomas.Hum Pathol 84:92–104 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1735 EP 1739 DI 10.1007/s12253-019-00754-6 PG 5 ER PT J AU Zhang, X Bu, R Liu, Z Wu, B Bai, S AF Zhang, Xuanyu Bu, Renge Liu, Zeqi Wu, Bin Bai, Song TI Development and Validation of a Model for Predicting Intravesical Recurrence in Organ-confined Upper Urinary Tract Urothelial Carcinoma Patients after Radical Nephroureterectomy: a Retrospective Study in One Center with Long-term Follow-up SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Intravesical recurrence; Upper urinary tract urothelial carcinoma; Radical nephroureterectomy; Retrospective study ID Intravesical recurrence; Upper urinary tract urothelial carcinoma; Radical nephroureterectomy; Retrospective study AB Although radical nephroureterectomy is the standard treatment method for upper urinary tract urothelial carcinoma, it is associated with a high risk of intravesical recurrence. There are no models for predicting IVR after RNU in patients with organ-confined UTUC. Therefore, we developed and validated a model for postoperative prediction of IVR after RNU. The development cohort consisted of 416 patients who underwent RNU with bladder cuff excision at our center between 1 January 2007 and 31December 2015. Patient clinicopathologic data were recorded. Multivariate Cox proportional hazard ratio regression was used to build a predictive model with regression coefficients, backward step-wise selection was applied, and the likelihood ratio test with Akaike’s information criterion was used as the stopping rule. An independent cohort consisting of 152 consecutive patients from 1 January 2016 and 31 December 2017 was used for validation. The performance of this predictive model was assessed with respect to discrimination, calibration, and clinical usefulness. The predictors in this model included tumor stage, tumor diameter, tumor location, and tumor grade. In the validation cohort, the model showed good discrimination, with a concordance index of 0.689 (95% CI, 0.629 to 0.748) and good calibration. Decision curve analysis demonstrated that the model was also clinically useful. This study presents a good model that may facilitate individualized postoperative prediction of IVR after RNU in patients with organ-confined UTUC, and thus, may help improve postoperative strategies and facilitate treatment outcomes. C1 [Zhang, Xuanyu] ShengJing Hospital of China Medical University, Department of Urology, 36 Sanhao Street, 110004 Shenyang, Liaoning, China. [Bu, Renge] ShengJing Hospital of China Medical University, Department of Urology, 36 Sanhao Street, 110004 Shenyang, Liaoning, China. [Liu, Zeqi] ShengJing Hospital of China Medical University, Department of Urology, 36 Sanhao Street, 110004 Shenyang, Liaoning, China. [Wu, Bin] ShengJing Hospital of China Medical University, Department of Urology, 36 Sanhao Street, 110004 Shenyang, Liaoning, China. [Bai, Song] ShengJing Hospital of China Medical University, Department of Urology, 36 Sanhao Street, 110004 Shenyang, Liaoning, China. RP Bai, S (reprint author), ShengJing Hospital of China Medical University, Department of Urology, 110004 Shenyang, China. EM baisongcmu@163.com CR Munoz JJ, Ellison LM, 2000, Upper tract urothelial neoplasms: incidence and survival during the last 2 decades. J Urol 164(5): 1523–1525 Roupret M, Colin P, Yates DR, 2014, A new proposal to risk stratify urothelial carcinomas of the upper urinary tract, UTUCs, in a predefinitive treatment setting: low-risk versus high-risk UTUCs. Eur Urol 66(2):181–183 Soria F, Shariat SF, Lerner SP, Fritsche HM, Rink M, Kassouf W, Spiess PE, Lotan Y, Ye D, Fernandez MI, Kikuchi E, Chade DC, Babjuk M, Grollman AP, Thalmann GN, 2017, Epidemiology, diagnosis, preoperative evaluation and prognostic assessment of upper-tract urothelial carcinoma, UTUC). World J Urol 35(3): 379–387 Abouassaly R, Alibhai SM, Shah N, Timilshina N, Fleshner N, Finelli A, 2010, Troubling outcomes from population-level analysis of surgery for upper tract urothelial carcinoma. Urology 76(4): 895–901 Jeldres C, Sun M, Isbarn H, Lughezzani G, Budaus L, Alasker A, Shariat SF, Lattouf JB,WidmerH, PharandD,Arjane P,Graefen M, Montorsi F, Perrotte P, Karakiewicz PI, 2010, A population-based assessment of perioperative mortality after nephroureterectomy for upper-tract urothelial carcinoma. Urology 75(2):315–320 Lughezzani G, Burger M, Margulis V, Matin SF, Novara G, Roupret M, Shariat SF, Wood CG, Zigeuner R, 2012, Prognostic factors in upper urinary tract urothelial carcinomas: a comprehensive review of the current literature. Eur Urol 62(1):100–114 Xylinas E, Kluth L, Passoni N, Trinh QD, Rieken M, Lee RK, Fajkovic H, Novara G, Margulis V, Raman JD, Lotan Y, Roupret M, Aziz A, Fritsche HM, Weizer A, Martinez-Salamanca JI, Matsumoto K, Seitz C, Remzi M, Walton T, Karakiewicz PI, Montorsi F, Zerbib M, Scherr DS, Shariat SF, UTUC Collaboration, 2014, Prediction of intravesical recurrence after radical nephroureterectomy: development of a clinical decisionmaking tool. Eur Urol 65(3):650–658 Geneva, 1997, Guidelines for controlling and monitoring the tobacco epidemic. World Health Organization, Geneva HoangML, Chen CH, Sidorenko VS, He J, Dickman KG, Yun BH, Moriya M, Niknafs N, Douville C, Karchin R, Turesky RJ, Pu YS, Vogelstein B, Papadopoulos N, Grollman AP, Kinzler KW, Rosenquist TA, 2013, Mutational signature of aristolochic acid exposure as revealed by whole-exome sequencing. Sci Transl Med 5(197): 197ra102 Colin P, Ouzzane A, Pignot G, Ravier E, Crouzet S, Ariane MM, Audouin M, Neuzillet Y, Albouy B, Hurel S, Saint F, Guillotreau J, Guy L, Bigot P, De La Taille A, Arroua F, Marchand C, Matte A, Fais PO, Roupret M, French Collaborative National Database on U.U.T.-U.C, 2012, Comparison of oncological outcomes after segmental ureterectomy or radical nephroureterectomy in urothelial carcinomas of the upper urinary tract: results from a large French multicentre study. BJU Int 110(8):1134–1141 Siegel RL, Miller KD, Jemal A, 2017, Cancer Statistics, 2017. CA Cancer J Clin 67(1):7–30 Lughezzani G, Jeldres C, Isbarn H, Sun M, Shariat SF, Alasker A, PharandD,Widmer H, Arjane P, Graefen M, Montorsi F, Perrotte P, Karakiewicz PI, 2009, Nephroureterectomy and segmental ureterectomy in the treatment of invasive upper tract urothelial carcinoma: a population-based study of 2299 patients. Eur J Cancer 45(18):3291–3297 von der Maase H, Sengelov L, Roberts JT, Ricci S, Dogliotti L, Oliver T, MooreMJ, Zimmermann A, ArningM(2005, Long-term survival results of a randomized trial comparing gemcitabine plus cisplatin, withmethotrexate, vinblastine, doxorubicin, plus cisplatin in patients with bladder cancer. J Clin Oncol 23(21):4602–4608 Collins GS, Reitsma JB, Altman DG, Moons KG, 2015, Transparent reporting of a multivariable prediction model for individual prognosis or diagnosis, TRIPOD): the TRIPOD statement. BMJ 350:g7594 Sauerbrei W, Boulesteix AL, Binder H, 2011, Stability investigations of multivariable regression models derived from low- and high-dimensional data. J Biopharm Stat 21(6):1206–1231 Raman JD, Ng CK, Boorjian SA, Vaughan ED Jr, Sosa RE, Scherr DS, 2005, Bladder cancer after managing upper urinary tract transitional cell carcinoma: predictive factors and pathology. BJU Int 96(7):1031–1035 Zigeuner RE, Hutterer G, Chromecki T, Rehak P, Langner C, 2006, Bladder tumour development after urothelial carcinoma of the upper urinary tract is related to primary tumour location. BJU Int 98(6):1181–1186 Novara G, De Marco V, Dalpiaz O, Gottardo F, Bouygues V, Galfano A, Martignoni G, Patard JJ, Artibani W, Ficarra V, 2008, Independent predictors of metachronous bladder transitional cell carcinoma, TCC, after nephroureterectomy for TCC of the upper urinary tract. BJU Int 101(11):1368–1374 Seisen T, Granger B, Colin P, Leon P, Utard G, Renard-Penna R, Comperat E,Mozer P, Cussenot O, Shariat SF, RoupretM(2015, A Systematic Review andMeta-analysis of Clinicopathologic Factors Linked to Int r a v e s i c a l Recurrence Af t e r Radical Nephroureterectomy to Treat Upper Tract Urothelial Carcinoma. Eur Urol 67(6):1122–1133 Terakawa T, Miyake H, Muramaki M, Takenaka A, Hara I, Fujisawa M, 2008, Risk factors for intravesical recurrence after surgical management of transitional cell carcinoma of the upper urinary tract. Urology 71(1):123–127 Hisataki T, Miyao N, Masumori N, Takahashi A, Sasai M, Yanase M, Itoh N, Tsukamoto T, 2000, Risk factors for the development of bladder cancer after upper tract urothelial cancer. Urology 55(5): 663–667 Roupret M, Babjuk M, Comperat E, Zigeuner R, Sylvester RJ, Burger M, Cowan NC, Bohle A, Van Rhijn BW, Kaasinen E, Palou J, Shariat SF, 2015, European Association of Urology Guidelines on Upper Urinary Tract Urothelial Cell Carcinoma: 2015 Update. Eur Urol 68(5):868–879 Espiritu PN, Sverrisson EF, Sexton WJ, Pow-Sang JM, Poch MA, Dhillon J, Spiess PE, 2014, Effect of tumor size on recurrence-free survival of upper tract urothelial carcinoma following surgical resection. Urol Oncol 32(5):619–624 Cho YH, Seo YH, Chung SJ, Hwang I, Yu HS, Kim SO, Jung SI, Kang TW, Kwon DD, Park K, Hwang JE, Heo SH, Kim GS, Hwang EC, 2014, Predictors of intravesical recurrence after radical nephroureterectomy for upper urinary tract urothelial carcinoma: an inflammation-based prognostic score. Korean J Urol 55(7):453–459 Narukawa T, Hara T, Arai E, Komiyama M, Kawahara T, Kanai Y, Fujimoto H, 2015, Tumour multifocality and grade predict intravesical recurrence after nephroureterectomy in patients with upper urinary tract urothelial carcinoma without a history of bladder cancer. Jpn J Clin Oncol 45(5):488–493 Harris AL, Neal DE, 1992, Bladder cancer–field versus clonal origin. N Engl J Med 326(11):759–761 Hinotsu S, Akaza H, Ohashi Y, Kotake T, 1999, Intravesical chemotherapy formaximumprophylaxis of newearly phase superficial bladder carcinoma treated by transurethral resection: a combined analysis of trials by the Japanese Urological Cancer Research Group using smoothed hazard function. Cancer 86(9):1818–1826 Elawdy MM, Osman Y, Taha DE, Zahran MH, El-Halwagy S, Garba ME, Harraz AM, 2017, Risk factors and prognosis of intravesical recurrence after surgical management of upper tract urothelial carcinoma: A 30-year single centre experience. Arab J Urol 15(3):216–222 Li WM, Wu WJ, Li CC, Ke HL, Wei YC, Yeh HC, Chou YH, Huang CH, Huang CN, 2013, The effect of tumor location on prognosis in patients with primary ureteral urothelial carcinoma. Urol Oncol 31(8):1670–1675 Novara G, De Marco V, Gottardo F, Dalpiaz O, Bouygues V, Galfano A, Martignoni G, Patard JJ, Artibani W, Ficarra V, 2007, Independent predictors of cancer-specific survival in transitional cell carcinoma of the upper urinary tract: multi-institutional dataset from 3 European centers. Cancer 110(8):1715–1722 Boorjian S, Ugras S, Mongan NP, Gudas LJ, You X, Tickoo SK, Scherr DS, 2004, Androgen receptor expression is inversely correlated with pathologic tumor stage in bladder cancer. Urology 64(2): 383–388 Shen SS, Smith CL, Hsieh JT, Yu J, Kim IY, Jian W, Sonpavde G, Ayala GE, Younes M, Lerner SP, 2006, Expression of estrogen receptors-alpha and -beta in bladder cancer cell lines and human bladder tumor tissue. Cancer 106(12):2610–2616 Kluth LA, Rieken M, Xylinas E, Kent M, Rink M, Roupret M, Sharifi N, Jamzadeh A, Kassouf W, Kaushik D, Boorjian SA, Roghmann F, Noldus J, Masson-Lecomte A, Vordos D, Ikeda M, Matsumoto K, Hagiwara M, Kikuchi E, Fradet Y, Izawa J, Rendon R, Fairey A, Lotan Y, Bachmann A, ZerbibM, FischM, Scherr DS, Vickers A, Shariat SF, 2014, Gender-specific differences in clinicopathologic outcomes following radical cystectomy: an international multi-institutional study of more than 8000 patients. Eur Urol 66(5):913–919 Pignot G, Colin P, ZerbibM, Audenet F, SoulieM, Hurel S, Delage F, Irani J, Descazeaud A, Droupy S, Rozet F, Phe V, Ruffion A, Long JA, Crouzet S, Houlgatte A, Bigot P, Guy L, Fais PO, Roupret M French Collaborative National Database on UUT-UC, 2014, Influence of previous or synchronous bladder cancer on oncologic outcomes after radical nephroureterectomy for upper urinary tract urothelial carcinoma. Urol Oncol 32(1):23 e1-e8 Akdogan B, Dogan HS, Eskicorapci SY, Sahin A, Erkan I, Ozen H, 2006, Prognostic significance of bladder tumor history and tumor location in upper tract transitional cell carcinoma. J Urol 176(1):48–52 Sung HH, Jeon HG, Han DH, Jeong BC, Seo SI, Lee HM, Choi HY, Jeon SS, 2015, Diagnostic Ureterorenoscopy Is Associated with Increased Intravesical Recurrence following Radical Nephroureterectomy in Upper Tract Urothelial Carcinoma. PLoS One 10(11):e0139976 Fradet V, Mauermann J, Kassouf W, Rendon R, Jacobsen N, Fairey A, Izawa J, Kapoor A, Black P, Tanguay S, Chin J, So A, Lattouf JB, Bell D, Saad F, Sheyegan B, Drachenberg D, Cagiannos I, Lacombe L, 2014, Risk factors for bladder cancer recurrence after nephroureterectomy for upper tract urothelial tumors: results from the Canadian Upper Tract Collaboration. Urol Oncol 32(6):839–845 HagiwaraM, Kikuchi E, Tanaka N, Matsumoto K, Ide H, Miyajima A, Masuda T, Nakamura S, OyaM(2013, Impact of smoking status on bladder tumor recurrence after radical nephroureterectomy for upper tract urothelial carcinoma. J Urol 189(6):2062–2068 Xylinas E, Kluth LA, Rieken M, Lee RK, ElghouayelM, Ficarra V, Margulis V, Lotan Y, Roupret M, Martinez-Salamanca JI, Matsumoto K, Seitz C, Karakiewicz PI, Zerbib M, Scherr DS, Shariat SF, UTUC Collaboration, 2014, Impact of smoking status and cumulative exposure on intravesical recurrence of upper tract urothelial carcinoma after radical nephroureterectomy. BJU Int 114(1):56–61 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1741 EP 1748 DI 10.1007/s12253-019-00748-4 PG 8 ER PT J AU Andras, Cs Bodoki, L Nagy-Vincze, M Griger, Z Csiki, E Danko, K AF Andras, Csilla Bodoki, Levente Nagy-Vincze, Melinda Griger, Zoltan Csiki, Emese Danko, Katalin TI Retrospective Analysis of Cancer-Associated Myositis Patients over the Past 3 Decades in a Hungarian Myositis Cohort SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer-associated myositis (CAM); Dermatomyositis (DM); Polymyositis (PM); Cancer; Muscle weakness ID Cancer-associated myositis (CAM); Dermatomyositis (DM); Polymyositis (PM); Cancer; Muscle weakness AB Association between cancer and myositis has been extensively reported and malignancy is a potentially life-threating complication in myositis. In this retrospective study authors give an overview of Hungarian cancer-associated myositis (CAM) patients treated at a single centre managing 450 myositis patients. All patients were diagnosed according to Bohan and Peter. Statistical analysis of disease onset, age, sex, muscle, skin and extramuscular symptoms, muscle enzymes, presence of antibodies, treatment and prognosis was performed. 43 patients could be considered as having CAM. 83.72% had cancer within one year of diagnosis of myositis. Most common localizations were ductal carcinoma of breast and adenocarcinoma of lung. Significant differences were observed between CAM and the non-CAM control patients: DM:PM ratio was 2.31:1 vs. 0.87:1, respectively (p = 0.029), age at diagnosis was 56.60 ± 12.79 vs. 38.88 ± 10.88 years, respectively (p < 0.001). Tumour-treatment was the following: surgical removal in 55.81%, chemotherapy in 51.1%, radiotherapy in 39.53%, hormone treatment in 18.6%, combination therapy in 51.16% of patients. Muscle enzyme levels of patients undergoing surgery were significantly reduced after intervention. 36 patients died (83.72%); 25 DM(83.33%) and 11 PM patients (84.62%); 5 years survival was 15.4% for PM and 27.5% for DM. This study demonstrates that DM, distal muscle weakness, asymmetric Raynaud’s phenomenon, older age, ANA-negativity are risk factors for developing malignancy and polymyositis patients have less chance of long-lasting survival. It is very important to think about cancer and follow every single myositis patient in the clinical routine because survival rate of CAM is very poor. C1 [Andras, Csilla] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Bodoki, Levente] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Nagy-Vincze, Melinda] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Griger, Zoltan] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Csiki, Emese] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. [Danko, Katalin] University of Debrecen, Faculty of Medicine, Department of Internal MedicineDebrecen, Hungary. RP Bodoki, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Internal Medicine, Debrecen, Hungary. EM bodoki.levente@gmail.com CR Mastaglia FL, Phillips BA, 2002, Idiopathic inflammatory myopathies: epidemiology, classification, and diagnostic criteria. Rheum Dis Clin N Am 28:723–741 Sun C, Lee JH, Yang YH, Yu HH, Wang LC, Lin YT et al, 2014, Juvenile Dermatomyositis: a 20-year retrospective analysis of treatment and clinical outcomes. Pediatr Neonatol S1875-9572:00093 Luo YB, Mastaglia FL, 2014, Dermatomyositis, polymyositis and immune-mediated necrotising myopathies. Biochim Biophys Acta S0925-4439:00162–00168 Catalan M, Selva-O’Callaghan A, Grau JM, 2014, Diagnosis and classification of sporadic inclusion body myositis, sIBM). Autoimmun Rev 13:363–366 Neri R, Barsotti S, Iacopetti V, Tripoli A, d’Ascanio A, Tavoni AG et al, 2014, Clinically amyopathic dermatomyositis: analysis of a monocentric cohort. J Clin Neuromuscul Dis 15:157–160 Danko K, Ponyi A, Molnar AP, Andras C, 2009, Constantin T. Paraneoplastic myopathy Curr Opin Rheumatol 21:594–598 Prieto-Gonzalez S, Grau JM, 2014, Diagnosis and classification of granulomatous myositis. Autoimmun Rev 13:372–374 Selva-O'Callaghan A, Trallero-Araguas E, Grau JM, 2014, Eosinophilic myositis: an updated review. Autoimmun Rev 13: 375–378 Yan J,Wu P, 2014, Idiopathic orbitalmyositis. J Craniofac Surg 25: 884–887 Michaelides SA, Passalidou E, Bablekos GD, Aza E, Goulas G, Chorti M et al, 2014, Cavitating lung lesion as a manifestation of inflammatory tumor, pseudotumor, of the lung: a case report and literature review. Am J Case Rep 15:258–265 Vajsar J, Jay V, 1996, Babyn P. Infantile myositis presenting in the neonatal period 18:415–419 Stertz G, 1916, Polymyositis. Berl Klin Wochenschr 53:489 Bohan A, Peter JB, 1975, Polymyositis and dermatomyositis, first of two parts). N Engl J Med 292:344–347 Chow WH, Gridley G, Mellemkjaer L et al, 1995, Cancer risk following polymyositis and dermatomyositis: a nationwide cohort study in Denmark. Cancer Causes Control 6:9–13 Zahr ZA, Baer AN, 2011)Malignancy in myositis. Curr Rheumatol Rep 13:208–215 Yang H, Peng Q, Yin L, Li S, Shi J, Zhang Y et al, 2017, Identification of multiple cancer-associated myositis-specific autoantibodies in idiopathic inflammatory myopathies: a large longitudinal cohort study. Arthritis Res Ther 19:259 HidaA,Yamashita T, Hosono Y, InoueM, Kaida K, KadoyaMet al, 2016, Anti-TIF1-γ antibody and cancer-associated myositis: a clinicohistopathologic study. Neurology 87:299–308 Trallero-Araguas E, Labrador-Horrillo M, Selva-O'Callaghan A, Martinez MA, Martinez-Gomez X, Palou E et al, 2010, Cancerassociated myositis and anti-p155 autoantibody in a series of 85 patients with idiopathic inflammatory myopathy. Medicine, Baltimore, 89:47–52 Lundberg IE, Tjarnlund A, Bottai M, Werth VP, Pilkington C, Visser M et al, 2017, European league against rheumatism/ American College of Rheumatology classification criteria for adult and juvenile idiopathic inflammatory myopathies and their major subgroups. Ann Rheum Dis 76:1955–1964 Leclair V, Lundberg IE, 2018, New myositis classification criteriawhat we have learned since Bohan and Peter. Curr Rheumatol Rep 20:18 Tieu J, Lundberg IE, Limaye V, 2016, Idiopathic inflammatory myositis. Best Pract Res Clin Rheumatol 30:149–168 Andras C, Ponyi A, Constantin T, Csiki Z, Szekanecz E, Szodoray P et al, 2008, Dermatomyositis and polymyositis associated with malignancy: a 21-year retrospective study. J Rheumatol 35:438– 444 Neri R, Barsotti S, Iacopetti V, IacopettiG, Pepe P, d'Ascanio Aet al, 2014, Cancer-associated myositis: a 35-year retrospective study of a monocentric cohort. Rheumatol Int 34:565–569 Qiang JK, KimWB, Baibergenova A, Alhusayen R, 2017, Risk of malignancy in Dermatomyositis and Polymyositis. J Cutan Med Surg 21:131–136 Liu Y, Xu L, Wu H, Zhao N, Tang Y, Li X et al, 2018, Characteristics and predictors of malignancy in dermatomyositis: analysis of 239 patients from northern China. Oncol Lett 16:5960– 5968 Menyhart O, Fekete JT, Gyorffy B, 2018, Demographic shift disproportionately increases cancer burden in an aging nation: current and expected incidence and mortality in Hungary up to 2030. Clin Epidemiol 10:1093–1108 Flynn M, Ottaway Z, Kaur J, Waters J, 2018, Three differently timed presentations of dermatomyositis associated with advanced ovarian cancer. BMJ Case Rep pii: bcr-2017-222627 Gkegkes ID, Minis EE, Iavazzo C, 2018, Dermatomyositis and colorectal cancer: a systematic review. Ir J Med Sci 187:615–620 Lim CH, TsengCW, Lin CT, HuangWN, Chen YH, ChenYMet al, 2018, The clinical application of tumormarkers in the screening of malignancies and interstitial lung disease of dermatomyositis/ polymyositis patients: a retrospective study. SAGE Open Med 6: 2050312118781895 Kumar S, Mahajan BB, Kaur S, Singh A, 2014, Paraneoplastic dermatomyositis with carcinoma cervix: a rare clinical association. Case Rep Dermatol Med 2014:836246 Piovesan DM, da Silva VD, Reichel CL, Bau P, Hoefel Filho JR, Staub HL, 2010, Neuroendocrine pancreatic tumor and dermatomyositis. Pancreas 39:684 Lalla SC, Aldridge RD, Tidman MJ, 2001, Carcinoma of the penis presenting with dermatomyositis. Clin Exp Dermatol 26:558 Mooney CJ, Dunphy EJ, Stone B, McNeel DG, 2006, Identification of autoantibodies elicited in a patient with prostate cancer presenting as dermatomyositis. Int J Urol 13:211–217 Sellami K, Mseddi M, Snoussi M, Gharbi H, Frikha F, Salah RB et al, 2018)Malignancy in a retrospective cohort of 17 patients with Dermatomyositis or Polymyositis in southern Tunisia. Rom J Intern Med 56:243–249 Wang J, Guo G, Chen G,Wu B, Lu L, Bao L, 2013, Meta-analysis of the association of dermatomyositis and polymyositis with cancer. Br J Dermatol 169:838–847 Hoesly PM, Sluzevich JC, Jambusaria-Pahlajani A, Lesser ER, Heckman MG, Abril A, 2018, Association of antinuclear antibody status with clinical features and malignancy risk in adult-onset dermatomyositis. J Am Acad Dermatol Sutic A, Gracanin G, Morovic-Vergles J, 2014, Raynaud's phenomenon - first sign of malignancy: case report. Acta Med Croatica 68: 295–298 Zhang L, Wu G, Gao D, Liu G, Pan L, Ni L et al, 2016, Factors associated with interstitial lung disease in patients with Polymyositis and Dermatomyositis: a systematic review and meta- analysis. PLoS One 11:e0155381 Lu X, Yang H, Shu X, Chen F, Zhang Y, Zhang S et al, 2014, Factors predicting malignancy in patients with polymyositis and dermatomyostis: a systematic review and meta-analysis. PLoS One 9:e94128. , DOI 10.1371/journal.pone.0094128 Selva-O'Callaghan A, Martinez-Gomez X, Trallero-Araguas E, Pinal-Fernandez I, 2018, The diagnostic work-up of cancerassociated myositis. Curr Opin Rheumatol 30:630–636 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1749 EP 1755 DI 10.1007/s12253-019-00756-4 PG 7 ER PT J AU Guo, JL Tang, T Li, JH Yang, YH Zhang, L Quan, Y AF Guo, Jun-Liang Tang, Tian Li, Jin-Hong Yang, Yi-Hong Zhang, Long Quan, Yi TI LncRNA HEIH Enhances Paclitaxel-Tolerance of Endometrial Cancer Cells via Activation of MAPK Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial cancer; HEIH; Paclitaxel resistance; MAPK signaling pathway ID Endometrial cancer; HEIH; Paclitaxel resistance; MAPK signaling pathway AB This study aimed to investigate the function of lncRNA HEIH on promoting endometrial cancer cells’ tolerance of paclitaxel (PTX). LncRNA HEIH expression was measured by QRT-PCR in endometrial cancer tissues, human healthy tissues and cell lines. The PTX-resistant endometrial cancer cells (Ishikawa-RE and HHUA-RE) were intermittently exposed to increase concentrations of PTX and were constructed as evidenced by MTT assay. Besides, the specific siRNA of HEIH (siHEIH) and pcDNA3.1-HEIH plasmid transfection were utilized to alter the expression of HEIH in the cells and investigate the effects of HEIH on resistance to PTX in endometrial cancer cells. Moreover, MTT, colony formation and apoptosis analysis were taken advantage to evaluate cell viability and proliferation when treated with PTX. Then, differential genes in PTX-resistant and HEIH knock-down PTX-resistant endometrial cancer cells were screened out by microarray analysis. Finally, gene-set enrichment analysis was used to predict the promising signaling pathway of HEIH and western blotting analysis were performed to verify the relevant genes expression of MAPK signaling pathway. LncRNA HEIH, the dysregulation of which involved in production of drug-resistance, was overexpressed in PTX-resistant endometrial cancer cells. Up-regulating HEIH would activate MAPK pathway, promote chemo-resistance of endometrial cancer cells and enhance cell proliferation and viability, whereas silencing HEIH depressed the MAPK signaling pathway, contributed to restoring chemo-sensitivity to PTX and repressed cell physiological process. Down-regulating lncRNA HEIH expression reversed the PTX-resistance of endometrial cancer cells through MAPK signaling pathway. C1 [Guo, Jun-Liang] West China Second University Hospital, Sichuan University, West China Institute of Women and Children’s Health, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Laboratory of Molecular Translational Medicine, No.20 Section 3, Renmin South Road, 610041 Chengdu, Sichuan, China. [Tang, Tian] West China Second University Hospital, Sichuan University, West China Institute of Women and Children’s Health, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Laboratory of Molecular Translational Medicine, No.20 Section 3, Renmin South Road, 610041 Chengdu, Sichuan, China. [Li, Jin-Hong] West China Second University Hospital, Sichuan University, West China Institute of Women and Children’s Health, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Laboratory of Molecular Translational Medicine, No.20 Section 3, Renmin South Road, 610041 Chengdu, Sichuan, China. [Yang, Yi-Hong] West China Second University Hospital, Sichuan University, West China Institute of Women and Children’s Health, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Laboratory of Molecular Translational Medicine, No.20 Section 3, Renmin South Road, 610041 Chengdu, Sichuan, China. [Zhang, Long] West China Second University Hospital, Sichuan University, West China Institute of Women and Children’s Health, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Laboratory of Molecular Translational Medicine, No.20 Section 3, Renmin South Road, 610041 Chengdu, Sichuan, China. [Quan, Yi] West China Second University Hospital, Sichuan University, West China Institute of Women and Children’s Health, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Laboratory of Molecular Translational Medicine, No.20 Section 3, Renmin South Road, 610041 Chengdu, Sichuan, China. RP Quan, Y (reprint author), West China Second University Hospital, Sichuan University, West China Institute of Women and Children’s Health, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Laboratory of Molecular Translational Medicine, 610041 Chengdu, China. EM quanyi516@163.com CR Dong L, Zhou Q, Zhang Z, Zhu Y, Duan T, Feng Y, 2012, Metformin sensitizes endometrial cancer cells to chemotherapy by repressing glyoxalase I expression. J Obstet Gynaecol Res 38(8): 1077–1085. , DOI 10.1111/j.1447-0756.2011.01839.x Kharma B, Baba T, Mandai M, Matsumura N, Murphy SK, Kang HS, Yamanoi K, Hamanishi J, Yamaguchi K, Yoshioka Y, Konishi I, 2013, Utilization of genomic signatures to identify high-efficacy candidate drugs for chemorefractory endometrial cancers. Int J Cancer 133(9):2234–2244. , DOI 10.1002/ijc.28220 Viswanathan AN, Moughan J, Miller BE, Xiao Y, Jhingran A, Portelance L, Bosch WR, Matulonis UA, Horowitz NS, Mannel RS, Souhami L, Erickson BA, Winter KA, Small W Jr, Gaffney DK, 2015, NRG oncology/RTOG 0921: a phase 2 study of postoperative intensity-modulated radiotherapy with concurrent cisplatin and bevacizumab followed by carboplatin and paclitaxel for patients with endometrial cancer. Cancer 121(13):2156–2163. , DOI 10.1002/cncr.29337 Galaal K, Al MoundhriM, Bryant A, Lopes AD, Lawrie TA, 2014, Adjuvant chemotherapy for advanced endometrial cancer. Cochrane Database Syst Rev 5:CD010681. , DOI 10. 1002/14651858.CD010681.pub2 Reyes HD, Miecznikowski J, Gonzalez-Bosquet J, Devor EJ, Zhang Y, Thiel KW, Samuelson MI, McDonald M, Stephan JM, Hanjani P, Guntupalli S, Tewari KS, Backes F, Ramirez N, Fleming GF, Filiaci V, Birrer MJ, Leslie KK, 2017, High stathmin expression is a marker for poor clinical outcome in endometrial cancer: an NRG oncology group/gynecologic oncology group study. Gynecol Oncol 146(2):247–253. , DOI 10.1016/j.ygyno.2017.05.017 Altundag O, Dursun P, Ayhan A, 2010, Emerging drugs in endometrial cancers. Expert Opin Emerg Drugs 15(4):557– 568. , DOI 10.1517/14728214.2010.517521 Jiang SJ, Zhang S, Mu XY, Li W, Wang Y, 2008, Effects of trichostatin a and paclitaxel on apoptosis and microtubule stabilization in endometrial carcinoma cells: an in vitro research. Zhonghua Yi Xue Za Zhi 88(34):2427–2431 Kuittinen T, Rovio P, Staff S, Luukkaala T, Kallioniemi A, Grenman S, Laurila M, Maenpaa J, 2017, Paclitaxel, carboplatin and 1,25-D3 inhibit proliferation of endometrial cancer cells in vitro. Anticancer Res 37(12):6575–6581. , DOI 10. 21873/anticanres.12114 Liz J, Esteller M, 2016, lncRNAs and microRNAs with a role in cancer development. Biochim Biophys Acta 1859(1):169–176. , DOI 10.1016/j.bbagrm.2015.06.015 Philippen LE, Dirkx E, da Costa-Martins PA, De Windt LJ, 2015, Non-coding RNA in control of gene regulatory programs in cardiac development and disease. J Mol Cell Cardiol 89(Pt A):51–58. , DOI 10.1016/j.yjmcc.2015.03.014 Bawa P, Zackaria S, VermaM, Gupta S, Srivatsan R, Chaudhary B, Srinivasan S, 2015, Integrative analysis of Normal long intergenic non-coding RNAs in prostate Cancer. PLoS One 10(5):e0122143. , DOI 10.1371/journal.pone.0122143 Szafranski K, Abraham KJ,Mekhail K, 2015, Non-coding RNA in neural function, disease, and aging. Front Genet 6:87. https://doi. org/10.3389/fgene.2015.00087 Malek E, Kim BG, Driscoll JJ, 2016, Identification of long noncoding RNAs deregulated in multiple myeloma cells resistant to proteasome inhibitors. Genes, Basel, 7(10). , DOI 10. 3390/genes7100084 Liu S, Zou B, Tian T, Luo X, Mao B, Zhang X, Lei H, 2018, Overexpression of the lncRNA FER1L4 inhibits paclitaxel tolerance of ovarian cancer cells via the regulation of theMAPK signaling pathway. J Cell Biochem. , DOI 10.1002/jcb.28032 Zhao H, Xing G, Wang Y, Luo Z, Liu G, Meng H, 2017, Long noncoding RNA HEIH promotes melanoma cell proliferation, migration and invasion via inhibition of miR-200b/a/429. Biosci Rep 37(3). , DOI 10.1042/BSR20170682 Yang F, Zhang L, Huo XS, Yuan JH, Xu D, Yuan SX, Zhu N, Zhou WP, Yang GS, Wang YZ, Shang JL, Gao CF, Zhang FR, Wang F, Sun SH, 2011, Long noncoding RNA high expression in hepatocellular carcinoma facilitates tumor growth through enhancer of zeste homolog 2 in humans. Hepatology 54(5):1679–1689. , DOI 10.1002/hep.24563 He Y,Meng XM, Huang C,Wu BM, Zhang L, Lv XW, Li J, 2014, Long noncoding RNAs: novel insights into hepatocelluar carcinoma. Cancer Lett 344(1):20–27. , DOI 10.1016/j.canlet. 2013.10.021 Haque SU, Niu L, Kuhnell D, Hendershot J, Biesiada J, Niu W, HaganMC, Kelsey KT, Casper KA,Wise-Draper TM, Medvedovic M, Langevin SM, 2018, Differential expression and prognostic value of long non-coding RNA in HPV-negative head and neck squamous cell carcinoma. Head Neck 40(7): 1555–1564. , DOI 10.1002/hed.25136 Cui C, Zhai D, Cai L, Duan Q, Xie L, Yu J, 2018, Long noncoding RNA HEIH promotes colorectal cancer tumorigenesis via counteracting miR-939Mediated transcriptional repression of BclxL. Cancer Res Treat 50(3):992–1008. , DOI 10.4143/crt. 2017.226 Zhang Y, Li Z, Zhang Y, Zhong Q, Chen Q, Zhang L, 2015, Molecular mechanism of HEIH and HULC in the proliferation and invasion of hepatoma cells. Int J Clin Exp Med 8(8):12956–12962 Tanaka T, Toujima S, Tanaka J, 2012, Differential sensitivity to paclitaxel-induced apoptosis and growth suppression in paclitaxelresistant cell lines established from HEC-1 human endometrial adenocarcinoma cells. Int J Oncol 41(5):1837–1844. , DOI 10.3892/ijo.2012.1600 Li L, Shou H, Wang Q, Liu S, 2019, Investigation of the potential theranostic role of KDM5B/miR-29c signaling axis in paclitaxel resistant endometrial carcinoma. Gene. , DOI 10.1016/j. gene.2018.12.076 Liu Z, SunM, Lu K, Liu J, ZhangM,WuW, DeW,Wang Z,Wang R, 2013, The long noncoding RNA HOTAIR contributes to cisplatin resistance of human lung adenocarcinoma cells via downregualtion of p21(WAF1/CIP1, expression. PLoS One 8(10):e77293. , DOI 10.1371/journal.pone.0077293 Wang Q, Zhang W, Hao S, 2017, LncRNA CCAT1 modulates the sensitivity of paclitaxel in nasopharynx cancers cells via miR-181a/ CPEB2 axis. Cell Cycle 16(8):795–801. , DOI 10.1080/ 15384101.2017.1301334 Gaundar SS, Bendall LJ, 2010, The potential and limitations of p38MAPK as a drug target for the treatment of hematological malignancies. Curr Drug Targets 11(7):823–833 Noel JK, Crean S, Claflin JE, Ranganathan G, Linz H, Lahn M, 2008, Systematic review to establish the safety profiles for direct and indirect inhibitors of p38 mitogen-activated protein kinases for treatment of cancer. A systematic review of the literature. Med Oncol 25(3):323–330. , DOI 10.1007/s12032-008-9039-1 Bai L, Mao R, Wang J, Ding L, Jiang S, Gao C, Kang H, Chen X, Sun X, Xu J, 2015, ERK1/2 promoted proliferation and inhibited apoptosis of human cervical cancer cells and regulated the expression of c-Fos and c-Jun proteins. Med Oncol 32(3):57. https://doi. org/10.1007/s12032-015-0490-5 Liu C, Ding L, Bai L, Chen X, Kang H, Hou L, Wang J, 2017, Folate receptor alpha is associated with cervical carcinogenesis and regulates cervical cancer cells growth by activating ERK1/2/c- Fos/c-Jun. Biochem Biophys Res Commun 491(4):1083– 1091. , DOI 10.1016/j.bbrc.2017.08.015 McGivern N, El-Helali A, Mullan P, McNeish IA, Paul Harkin D, Kennedy RD, McCabe N, 2018, Activation of MAPK signalling results in resistance to saracatinib, AZD0530, in ovarian cancer. Oncotarget 9(4):4722–4736. , DOI 10.18632/oncotarget. 23524 Wang P, Chen D, Ma H, Li Y, 2017, LncRNA SNHG12 contributes to multidrug resistance through activating the MAPK/slug pathway by sponging miR-181a in non-small cell lung cancer. Oncotarget 8(48):84086–84101. , DOI 10.18632/ oncotarget.20475 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1757 EP 1766 DI 10.1007/s12253-019-00718-w PG 10 ER PT J AU Somoracz, Kuthi, L Micsik, T Jenei, A Hajdu, A Vrabely, B Raso, E Sapi, Z Bajory, Z Kulka, J Ivanyi, B AF Somoracz, Aron Kuthi, Levente Micsik, Tamas Jenei, Alex Hajdu, Adrienn Vrabely, Brigitta Raso, Erzsebet Sapi, Zoltan Bajory, Zoltan Kulka, Janina Ivanyi, Bela TI Renal Cell Carcinoma with Clear Cell Papillary Features: Perspectives of a Differential Diagnosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Clear cell carcinoma; Clear cell papillary carcinoma; Cytokeratin 7-positivity; Differential diagnosis; VHL gene ID Clear cell carcinoma; Clear cell papillary carcinoma; Cytokeratin 7-positivity; Differential diagnosis; VHL gene AB Thirty-one cases of low-grade renal cell carcinoma (RCC) with clear cells and tubulopapillary/papillary architecture were analyzed retrospectively with immunohistochemical and genetic markers to gain more experience with the differential diagnosis of such cases. All samples coexpressed CK7 and CA9; the TFE3 or TFEB reactions were negative; the CD10 and the AMACR stainings were negative in 27 cases and 30 cases, respectively. The FISH assays for papillary RCC, available in 27 cases, and deletion of chromosome 3p, available in 29 cases, gave negative results. The results for 3p deletion, VHL gene mutation or VHL gene promoter region hypermethylation testing, along with the diffuse CD10-positivity in 2 cases confirmed 21 cases as clear cell papillary RCC (CCPRCC; CK7+, CA9+; no 3p loss, no VHL abnormality) and 10 cases as clear cell RCC (CCRCC; CK7+, CA9+; no 3p loss, VHL abnormality mutation/hypermethylation present). In CCPRCCs, the representative growth pattern was branching tubulo-acinar, commonly accompanied by cyst formation. The linear nuclear arrangement or cup-shaped staining of CA9 did not necessarily indicate CCPRCC, and the absence of these did not exclude the diagnosis of CCPPRC. One tumor infiltrated the renal sinus; the others exhibited pT1 stage; and metastatic outcome was not recorded. The CCRCC cases were in pT1 stage; 6 exhibited cup-shaped staining of CA9, and 1 displayed lymph node metastasis at the time of surgery. Distant metastatic disease was not observed. In summary, the VHL abnormalities distinguished the subset of CCRCC with diffuse CK7- positivity and no 3p loss from cases of CCPRCC. C1 [Somoracz, Aron] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Kuthi, Levente] University of Szeged, Department of PathologySzeged, Hungary. [Micsik, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Jenei, Alex] University of Szeged, Department of PathologySzeged, Hungary. [Hajdu, Adrienn] University of Szeged, Department of PathologySzeged, Hungary. [Vrabely, Brigitta] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Raso, Erzsebet] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Bajory, Zoltan] University of Szeged, Department of UrologySzeged, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Ivanyi, Bela] University of Szeged, Department of PathologySzeged, Hungary. RP Somoracz, (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM somoracz14@gmail.com CR Srigley JR, Delahunt B, Eble JN et al, 2013, The International Society of Urological Pathology, ISUP, Vancouver Classification of Renal Neoplasia. Am J Surg Pathol 37:1469–1489 Moch H, Humphrey PA, Ulbright TM, Reuter VE, 2016, WHO Classification of Tumours of the Urinary System and Male Genital Organs. International Agency for Research on Cancer Tickoo SK, dePeralta-Venturina MN, Harik LR et al, 2006, Spectrum of epithelial neoplasms in end-stage renal disease: an experience from 66 tumor-bearing kidneys with emphasis on histologic patterns distinct from those in sporadic adult renal neoplasia. Am J Surg Pathol 30:141–153 Gobbo S, Eble JN, GrignonDJ et al, 2008, Clear cell papillary renal cell carcinoma: a distinct histopathologic and molecular genetic entity. Am J Surg Pathol 32:1239–1245 Aydin H, Chen L, Cheng L et al, 2010, Clear cell tubulopapillary renal cell carcinoma: a study of 36 distinctive low-grade epithelial tumors of the kidney. Am J Surg Pathol 34:1608–1621 Rohan SM, Xiao Y, Liang Yet al, 2011, Clear-cell papillary renal cell carcinoma: molecular and immunohistochemical analysis with emphasis on the von Hippel-Lindau gene and hypoxia-inducible factor pathway-related proteins. Mod Pathol 24:1207–1220 Ross H, Martignoni G, Argani P, 2012, Renal cell carcinoma with clear cell and papillary features. Arch Pathol LabMed 136:391–399 Michal M, Hes O, Havlicek F, 2000, Benign renal angiomyoadenomatous tumor: a previously unreported renal tumor. Ann Diagn Pathol 4:311–315 Michal M, Hes O, Kuroda N et al, 2009, Difference between RAT and clear cell papillary renal cell carcinoma/clear renal cell carcinoma. Virchows Arch. , DOI 10.1007/s00428-009-0788-9 Deml KF, Schildhaus HU, Comperat E et al, 2015, Clear cell papillary renal cell carcinoma and renal angiomyoadenomatous tumor: two variants of a morphologic, immunohistochemical, and genetic distinct entity of renal cell carcinoma. Am J Surg Pathol 39:889– 901 Tickoo SK, Reuter VE, 2011, Differential diagnosis of renal tumors with papillary architecture. Adv Anat Pathol 18:120–132 Kuroda N, Ohe C, Kawakami F et al, 2014, Clear cell papillary renal cell carcinoma: a review. Int J Clin Exp Pathol 7:7312–7318 Gobbo S, Eble JN, Maclennan GT et al, 2008, Renal cell carcinomas with papillary architecture and clear cell components: the utility of immunohistochemical and cytogenetical analyses in differential diagnosis. Am J Surg Pathol 32:1780–1786 Diolombi ML, Cheng L, Argani P et al, 2015, Do Clear Cell Papillary Renal Cell Carcinomas Have Malignant Potential? Am J Surg Pathol 39:1621–1634 Fallon KB, Palmer CA, Roth KA et al, 2004, Prognostic value of 1p, 19q, 9p, 10q, and EGFR-FISH analyses in recurrent oligodendrogliomas. J Neuropathol Exp Neurol 63:314–322 Akanni OE, Ferrari M, 2006, Sequencing of Von Hippel-Lindau, VHL, Gene from Genomic DNA for Mutation Detection in Italian Patients. EJIFCC 17:12–16 Herman JG, Graff JR, Myohanen S et al, 1996, Methylationspecific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 93:9821–9826 Hakimi AA, Tickoo SK, Jacobsen A et al, 2015, TCEB1-mutated renal cell carcinoma: a distinct genomic and morphological subtype. Mod Pathol 28:845–853 Lan TT, Keller-Ramey J, Fitzpatrick C et al, 2016, Unclassified renal cell carcinoma with tubulopapillary architecture, clear cell phenotype, and chromosome 8 monosomy: a new kid on the block. Virchows Arch 469:81–91 Canzonieri V, Volpe R, Gloghini A et al, 1993, Mixed renal tumor with carcinomatous and fibroleiomyomatous components, associated with angiomyolipoma in the same kidney. Pathol Res Pract 189:951–956 Martignoni G, Brunelli M, Segala D et al, 2014, Renal cell carcinoma with smooth muscle stroma lacks chromosome 3p and VHL alterations. Mod Pathol 27:765–774 Petersson F, Branzovsky J, Martinek P et al, 2014, The leiomyomatous stroma in renal cell carcinomas is polyclonal and not part of the neoplastic process. Virchows Arch 465:89–96 Kuhn E, De Anda J, Manoni S et al, 2006, Renal cell carcinoma associated with prominent angioleiomyoma-like proliferation: Report of 5 cases and review of the literature. Am J Surg Pathol 30:1372–1381 Shannon BA, Cohen RJ, Segal A et al, 2009, Clear cell renal cell carcinoma with smooth muscle stroma. Hum Pathol 40:425–429 Petersson F, Martinek P, Vanecek T et al, 2018, Renal Cell Carcinoma With Leiomyomatous Stroma: A Group of Tumors With Indistinguishable Histopathologic Features, But 2 Distinct Genetic Profiles: Next-Generation Sequencing Analysis of 6 Cases Negative for Aberrations Related to the VHL gene. Appl Immunohistochem Mol Morphol 26:192–197 Williamson SR, Zhang S, Eble JN et al, 2013, Clear cell papillary renal cell carcinoma–like tumors in patients with von Hippel- Lindau disease are unrelated to sporadic clear cell papillary renal cell carcinoma. Am J Surg Pathol 37:1131–1139 Rao P, Monzon F, Jonasch E et al, 2014, Clear cell papillary renal cell carcinoma in patients with von Hippel-Lindau syndrome– clinicopathological features and comparative genomic analysis of 3 cases. Hum Pathol 45:1966–1972 Hes O, Comperat EM, Rioux-LeclercqN(2016, Clear cell papillary renal cell carcinoma, renal angiomyoadenomatous tumor, and renal cell carcinoma with leiomyomatous stroma relationship of 3 types of renal tumors: a review. Ann Diagn Pathol 21:59–64 Williamson SR, Eble JN, Cheng L et al, 2013, Clear cell papillary renal cell carcinoma: differential diagnosis and extended immunohistochemical profile. Mod Pathol 26:697–708 Zhou H, Zheng S, Truong LD et al, 2014, Clear cell papillary renal cell carcinoma is the fourth most common histologic type of renal cell carcinoma in 290 consecutive nephrectomies for renal cell carcinoma. Hum Pathol 45:59–64 Dhakal HP, McKenney JK, Khor LYet al, 2016, Renal Neoplasms With Overlapping Features of Clear Cell Renal Cell Carcinoma and Clear Cell Papillary Renal Cell Carcinoma: A Clinicopathologic Study of 37 Cases From a Single Institution. Am J Surg Pathol 40:141–154 Williamson SR, Gupta NS, Eble JN et al, 2015, Clear Cell Renal Cell Carcinoma With Borderline Features of Clear Cell Papillary Renal Ce l l Ca r c i n o m a : Combined Morphol o g i c , Immunohistochemical, and Cytogenetic Analysis. Am J Surg Pathol 39:1502–1510 AronM, Chang E, Herrera L et al, 2015, Clear Cell-Papillary Renal Cell Carcinoma of the Kidney Not Associated With End-stage Renal Disease Clinicopathologic Correlation With Expanded Immunophenotypic and Molecular Characterization of a Large Cohort With Emphasis on Relationship With Renal Angiomyoadenomatous Tumor. Am J Surg Pathol 39:873–888 Alexiev BA, Drachenberg CB, 2014, Clear cell papillary renal cell carcinoma: Incidence, morphological features, immunohistochemical profile, and biologic behavior: A single institution study. Pathol Res and Pract 210:234–241 Petersson F, Grossmann P, Hora M et al, 2013, Renal cell carcinoma with areas mimicking renal angiomyoadenomatous tumor/clear cell papillary renal cell carcinoma. Hum Pathol 44:1412–1420 Dhakal P, Giri S, Siwakoti K et al, 2017, Renal Cancer in Recipients of Kidney Transplant. Rare Tumors 9:6550 Adam J, Couturier J, Molinie V et al, 2011, Clear-cell papillary renal cell carcinoma: 24 cases of a distinct low-grade renal tumour and a comparative genomic hybridization array study of seven cases. Histopathology 58:1064–1071 Massari F, Ciccarese C, Hes O et al, 2018, The Tumor Entity Denominated "clear cell-papillary renal cell carcinoma" According to the WHO 2016 new Classification, have the Clinical Characters of a Renal Cell Adenoma as does Harbor a Benign Outcome. Pathol Oncol Res 24:447–456 Mai KT, Kohler DM, Belanger EC et al, 2008, Sporadic clear cell renal cell carcinoma with diffuse cytokeratin 7 immunoreactivity. Pathology 40:481–486 Mertz KD, Demichelis F, Sboner A et al, 2008, Association of cytokeratin 7 and 19 expression with genomic stability and favorable prognosis in clear cell renal cell cancer. Int J Cancer 123:569– 576 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1767 EP 1776 DI 10.1007/s12253-019-00757-3 PG 10 ER PT J AU Steppert, C Krugmann, J Sterlacci, W AF Steppert, Claus Krugmann, Jens Sterlacci, William TI Simultaneous endocrine expression and loss of melanoma markers in malignant melanoma metastases, a retrospective analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Neuroendocrine expression; Melanoma metastases; Loss of melanoma markers ID Neuroendocrine expression; Melanoma metastases; Loss of melanoma markers AB Malignant melanoma metastases are chameleons of histopathology. In 4 primary malignant melanomas and 20 melanoma metastases expression of S-100, HMB-45 and melan-A as melanoma markers and CD56, synaptophysin and chromogranin-A as neuroendocrine markers was retrospectively analyzed. While all primary tumors expressed all 3 melanoma markers 7/20 of melanoma metastases had lost at least one melanoma marker, one had lost all three markers. Conversely about half of the samples stained for CD56, only 6/20metastases were negative for all 3 neuroendocrine markers. None expressed chromogranin-A. Partial loss of melanoma markers and expression of neuroendocrine markers seems not to be infrequent. In patients with a history of malignant melanoma and suspected metastases, losing melanoma markers while expressing neuroendocrine markers is a potential diagnostic pitfall. Therefore all 3 melanoma markers should be performed as well as chromogranin-A staining. In doubt, metastases of the melanoma should be assumed. C1 [Steppert, Claus] Klinikum Bayreuth, Department of Pulmonology and Thoracic Oncology, Preuschwitzer Str. 101Bayreuth, D-95445, Germany. [Krugmann, Jens] Klinikum Bayreuth, Department of Pathology, Preuschwitzer Str. 101Bayreuth, D-95445, Germany. [Sterlacci, William] Klinikum Bayreuth, Department of Pathology, Preuschwitzer Str. 101Bayreuth, D-95445, Germany. RP Steppert, C (reprint author), Klinikum Bayreuth, Department of Pulmonology and Thoracic Oncology, Bayreuth, Germany. EM claus.steppert@klinikum-bayreuth.de CR Shinohara MM, Deubner H, Argenyi ZB., 2009, S100, HMB-45, and Melan-A negative primary melanoma. Dermatol Online J. Sep 15;15(9):7 Agaimy A, Specht K, Stoehr R, Lorey T, Markl B, Niedobitek G, Straub M, Hager T, Reis AC, Schilling B, Schneider-Stock R, Hartmann A, Mentzel T, 2016, Metastatic Malignant Melanoma With Complete Loss of Differentiation Markers, Undifferentiated/ Dedifferentiated Melanoma): Analysis of 14 Patients Emphasizing Phenotypic Plasticity and the Value of Molecular Testing as Surrogate Diagnostic Marker. Am J Surg Pathol 40:181–191 Ilardi G, Caroppo D, Varricchio S, Vita G, Di Lorenzo P, Insabato L, De Rosa G,MascoloM(2015, Analmelanoma with neuroendocrine differentiation: report of a case. Int J Surg Pathol 23:329–332 Kacerovska D, Michal M, Sosna B, Cempirkova D, Ambrus M, Richtr P, Danis D, Zelger B, Kazakov DV, 2009, Carcinoid-like pattern in melanoma: report of 4 cases. Am J Dermatopathol 31: 542–550 Eyden B, Pandit D, Banerjee SS, 2005, Malignant melanoma with neuroendocrine differentiation: clinical, histological, immunohistochemical and ultrastructural features of three cases. Histopathology 47:402–409 Mahmood MN, Lee MW, Linden MD, Nathanson SD, Hornyak TJ, Zarbo RJ, 2002, Diagnostic value of HMB-45 and anti-Melan A staining of sentinel lymph nodes with isolated positive cells. Mod Pathol 15:1288–1293 Ali TH, Pisanti S, Ciaglia E, Mortarini R, Anichini A, Garofalo C, Tallerico R, Santinami M, Gulletta E, Ietto C, Galgani M, Matarese G, Bifulco M, Ferrone S, Colucci F, Moretta A, Karre K, Carbone E, 2014, Enrichment of CD56(dim)KIR + CD57 + highly cytotoxic NK cells in tumour-infiltrated lymph nodes of melanoma patients. Nat Commun 5:5639. , DOI 10.1038/ncomms6639 Romano RC, Carter JM, Folpe AL, 2015, Aberrant intermediate filament and synaptophysin expression is a frequent event in malignant melanoma: an immunohistochemical study of 73 cases. Mod Pathol 28:1033–1042 Guinee DG, Fishback NF, Koss MN, Abbondanzo SL, Travis WD, 1994, The spectrum of immunohistochemical staining of small-cell lung carcinoma in specimens from transbronchial and open-lung biopsies. Am J Clin Pathol 102:406–414 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1777 EP 1779 DI 10.1007/s12253-019-00761-7 PG 3 ER PT J AU Rivera-Franco, MM Leon-Rodriguez, E Torres-Ruiz, JJ Gomez-Martin, D Angles-Cano, E Sevilla-Gonzalez, dlLM AF Rivera-Franco, M Monica Leon-Rodriguez, Eucario Torres-Ruiz, J Jose Gomez-Martin, Diana Angles-Cano, Eduardo Sevilla-Gonzalez, de la Luz Maria TI Neutrophil Extracellular Traps Associate with Clinical Stages in Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Neutrophil extracellular traps; Breast Cancer; Metastasis ID Neutrophil extracellular traps; Breast Cancer; Metastasis AB Recently, neutrophil extracellular traps (NETs), three-dimensional structures formed of neutrophil enzymes such as neutrophil elastase (NE) and nuclear components (DNA), have been associated with progression in different types of cancer. However, data remain scarce in breast cancer. Thus, the aim of this study was to associate NETs with clinical stages of breast cancer. A prospective analysis was performed in 45 plasma samples of female patients with newly diagnosed breast cancer. NE-DNA complexes were evaluated by ELISA. Optical density was dichotomized at the median for comparisons (low and high levels of NE-DNA). The most frequent clinical stage was localized (n = 28, 62%) followed by regional (n = 13, 29%) and distant (n = 4, 9%). Higher levels of NE-DNA complexes were observed in regional and distant stages compared to localized disease (68% vs 32%, p = 0.034). No differences were observed when comparing other clinical characteristics between both groups. We demonstrated that the levels of NETs increase in proportion to the stage of the disease, observing higher levels of NE-DNA complexes in regional and metastatic disease, which coincides with the proposed mechanism by which cancer progression and metastasis might result from the formation of NETs. C1 [Rivera-Franco, M Monica] Instituto Politecnico Nacional, Escuela Superior de Medicina, Section of Graduate and Research Studies, Plan de San Luis y Diaz Miron s/n, Casco de Santo Tomas, Miguel Hidalgo, 11340 Mexico City, Mexico. [Leon-Rodriguez, Eucario] Department of Hematology and OncologyMexico City, Mexico. [Torres-Ruiz, J Jose] Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Department of Hematology and Oncology, Vasco de Quiroga 15, Belisario Dominguez Seccion XVI, Tlalpan, 14080 Mexico City, Mexico. [Gomez-Martin, Diana] Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prieto 3000, 64710 Nuevo Leon, Mexico. [Angles-Cano, Eduardo] Faculte de Pharmacie de Paris, Inserm UMR_S1140 Innovative Therapies in Haemostasis, 4 Avenue de l’Observatoire, 75006 Paris, France. [Sevilla-Gonzalez, de la Luz Maria] Instituto Politecnico Nacional, Escuela Superior de Medicina, Section of Graduate and Research Studies, Plan de San Luis y Diaz Miron s/n, Casco de Santo Tomas, Miguel Hidalgo, 11340 Mexico City, Mexico. RP Rivera-Franco, MM (reprint author), Instituto Politecnico Nacional, Escuela Superior de Medicina, Section of Graduate and Research Studies, 11340 Mexico City, Mexico. EM mm.riverafranco@gmail.com CR Siegel RL, Miller KD, Jemal A, 2019, Cancer statistics, 2019. CA Cancer J Clin 69:7 http://gco.iarc.fr/today/data/factsheets/populations/900-world-factsheets. pdf, Accessed on June 18, 2019) Noone AM, Howlader N, Krapcho M, et al, Eds). SEER Cancer Statistics Review, 1975-2015, National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/csr/1975_2015/, based on November 2017 SEER data submission, posted to the SEER web site, April 2018 Knaul FM, Nigenda G, Lozano R et al, 2009, Cancer de mama en Mexico: una prioridad apremiante. Salud Publica Mex 51:S335– S344 Amin MB, Edge S, Greene F et al, eds,, 2017, AJCC Cancer Staging Manual, 8th edition). American Joint Commission on Cancer, Springer International Publishing Brinkmann V, Reichard U, Goosmann C et al, 2004, Neutrophil extracellular traps kill bacteria. Science. 303:1532–1535 Berger-Achituv S, Brinkmann V, Abed UA et al, 2013, A proposed role for neutrophil extracellular traps in cancer immunoediting. Front Immunol 4:48 Erpenbeck L, SchonNeutrophil MP, 2017, Neutrophil extracellular traps: protagonists of cancer progression? Oncogene. 36(18):2483– 2490 Park J,Wysocki RW,Amoozgar Z, et al., 2016, Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci Transl Med 8:361ra138 Cools-Lartigue J, Spicer J, McDonald B et al, 2013, Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest 123(8):3446–3458 Tohme S, Yazdani HO, Al-Khafaji AB et al, 2016, Neutrophil Extracellular Traps Promote the Development and Progression of Liver Metastases after Surgical Stress. Cancer Res 76(6):1367– 1380 Najmeh S, Cools-Lartigue J, Rayes RF, Gowing S, Vourtzoumis P, 2017, Neutrophil extracellular traps sequester circulating tumor cells via β1-integrin mediated interactions. Int J Cancer 140(10): 2321–2330 Albrengues J, ShieldsMA, Ng D, Park CG, Ambrico A, Poindexter ME, et al., 2018, Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science. 361(6409). pii: eaao4227 Richardson JJR, Hendrickse CW, Gao-Smith F, ThickettDR, 2015, Neutrophil extracellular trap formation is increased in patients with colorectal cancer in-vivo. Gut 64:A530–A531 Demers M, Wong SL, Martinod K et al, 2016, Priming of neutrophils toward NETosis promotes tumor growth. Oncoimmunology. 5(5):e1134073 van der Windt DJ, Sud V, Zhang H et al, 2018, Neutrophil extracellular traps promote inflammation and development of hepatocellular carcinoma in nonalcoholic steatohepatitis. Hepatology. 68(4): 1347–1360 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1781 EP 1785 DI 10.1007/s12253-019-00763-5 PG 5 ER PT J AU Mohamed, EZAF El, ZIK Toni, DMN AF Mohamed, El-Zahraa Ammar Fatma El, Zahraa Ibrahim Khalil Toni, D M Nisreen TI Caveolin-1 Expression Together with VEGF can be a Predictor for Lung Metastasis and Poor Prognosis in Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Caveolin-1; Vascular endothelial growth factor; Osteosarcoma; Immunohistochemistry ID Caveolin-1; Vascular endothelial growth factor; Osteosarcoma; Immunohistochemistry AB Caveolin-1, the major protein component of caveolae, plays vital functions in tumorigenesis and metastasis. Previous evidence demonstrated the positive role of Caveolin-1 in the regulation of endothelial cell differentiation and the involvement of Caveolin- 1 in vascular endothelial growth factor (VEGF) mediated angiogenesis. The correlation of Caveolin-1 expression and angiogenesis is not yet elucidated in osteosarcoma. This study aimed to investigate the expression levels of Caveolin-1 and VEGF in osteosarcoma and their associations with clinicopathological data. This study included 66 formalin-fixed and paraffin embedded osteosarcoma tissue samples. The expression levels of Caveolin-1 and VEGF were assessed by immunohistochemistry. Then associations with clinicopathological variables and the correlation between both markers were evaluated statistically. We also investigated the expression of Caveolin-1 and VEGF values in gene microarrays of osteosarcoma patients and cell lines by using GEO data sets on https://www.ncbi.nlm.nih.gov. Caveolin-1 and VEGF were expressed in 19.6% and 77.3%, respectively. Caveolin-1 expression was associated positively with osteoblastic histological subtype (P < 0.0001). VEGF expression showed positive association with patient age, histological grade and clinical stage (P = 0.031, P = 0.024 and P < 0.001; respectively).An inverse correlation between Caveolin-1 and VEGF expressions in osteosarcoma was found (r = 0.2 P = 0.04). In silico analysis of Caveolin-1 and VEGF expression supported our results. Our results suggest that Caveolin-1 may act as a tumor suppressor in osteosarcoma. Down-regulation of Caveolin-1 can be used as an indicator for poor prognosis in osteosarcoma patients. Meanwhile, overexpression of VEGF is a predictor of pulmonary metastasis and poor prognosis. C1 [Mohamed, El-Zahraa Ammar Fatma] Minia University, Faculty of Medicine, Pathology DepartmentMinia, Egypt. [El, Zahraa Ibrahim Khalil] Minia University, Faculty of Medicine, Pathology DepartmentMinia, Egypt. [Toni, D M Nisreen] Minia University, Faculty of Medicine, Pathology DepartmentMinia, Egypt. RP El, ZIK (reprint author), Minia University, Faculty of Medicine, Pathology Department, Minia, Egypt. EM zahraa333eg@gmail.com CR Isakoff MS, Bielack SS, Meltzer P, Gorlick R, 2015, Osteosarcoma: Current treatment and a collaborative pathway to success. J Clin Oncol 33:3029–3035. , DOI 10.1200/JCO.2014.59.4895 KleinMJ, Siegal GP, 2006)Osteosarcoma: anatomic and histologic variants. Am J Clin Pathol 125(4):555–581. , DOI 10. 1309/UC6K-QHLD-9LV2-KENN Rubin P, Williams JP, Devesa SS, Travis LB, Constine LS, 2010, Cancer genesis across the age spectrum: associations with tissue development, maintenance, and senescence. Semin Radiat Oncol 20:3–11. , DOI 10.1016/j.semradonc.2009.08.001 Fletcher CD, Bridge J, Hogendoorn PC, Mertens F, eds,, 2013, The World Health Organization Classification of Tumors of Soft Tissue and Bone. IARC, Lyon Zalupski MM, Rankin C, Ryan JR, Lucas DR, Muler J, Lanier KS, Budd GT, Biermann JS, Meyers FJ, Antman K, 2004, Adjuvant therapy of osteosarcoma–a phase II trial: SouthwestOncology Group study 9139. Cancer 100(4):818–825. , DOI 10.1002/cncr.20021 Wada T, Isu K, Takeda N, Usui M, Ishii S, Yamawaki S, 1996, A preliminary report of neoadjuvant chemotherapy NSH-7 study in osteosarcoma: preoperative salvage chemotherapy based on clinical tumor response and the use of granulocyte colony-stimulating factor. Oncology 53:221–227. , DOI 10.1159/000227564 Xie Y, Huang J, Wu M, Zhou Y, 2018, Expression of CD133 protein in osteosarcoma and its relationship with the clinicopathological features and prognosis. J Cancer Res Ther 14:892–895. , DOI 10.4103/jcrt.JCRT_461_17 Xu M, Xie Y, ShengW, Miao J, Yang J, 2015, Adenovirus-mediated ING4 Gene Transfer in Osteosarcoma Suppresses Tumor Growth via Induction of Apoptosis and Inhibition of Tumor Angiogenesis. Technology in Cancer Research and Treatment 14:369–378. , DOI 10.1177/1533034614500424 Kaya M, Wada T, Akatsuka T, Kawaguchi S, Nagoya S, Shindoh M, Higashino F, Mezawa F, Okada F, Ishii S, 2000, Vascular endothelial growth factor expression in untreated osteosarcoma is predictive of pulmonary metastasis and poor prognosis. Clin Cancer Res 6(2):572–577 DuBois S, Demetri G, 2007, Markers of angiogenesis and clinical features in patientswith sarcoma. Cancer 109(5):813–981. , DOI 10.1002/cncr.22455 Dvorak HF, Sioussat TM, Brown LF, Berse B, Nagy JA, Sotrel A, Manseau EJ, Van de Water L, Senger DR, 1991, Distribution of vascular permeability factor, vascular endothelial growth factor, in tumors: concentration in tumor blood vessels. J Exp Med 174(5): 1275–1278 Cohen AW, Hnasko R, Schubert W, Lisanti MP, 2004, Role of caveolae and caveolins in health and disease. Physiol Rev 84: 1341–1379. , DOI 10.1152/physrev.00046.2003 Schwencke C, Braun-Dullaeus RC, Wunderlich C, Strasser RH, 2006, Caveolae and caveolin in transmembrane signaling: Implications for human disease. Cardiovasc Res 70(1):42–49. , DOI 10.1016/j.cardiores.2005.11.029 Stan RV, 2005, Structure of caveolae. Journal of Biochimica et Biophysica Acta 1746:334–348. , DOI 10.1016/j.bbamcr. 2005.08.008 Smart EJ, Graf GA, McNiven MA, Sessa WC, Engelman JA, Scherer PE, Okamoto T, Lisanti MP, 1999, Caveolins, liquidordered domains, and signal transduction. Mol Cell Biol 19: 7289–7304 Solomon KR, Danciu TE, Adolphson LD, Hecht LE, Hauschka PV, 2000, Caveolin-enriched membrane signaling complexes in human and murine osteoblasts. J Bone Miner Res 15:2380–2390. , DOI 10.1359/jbmr.2000.15.12.2380 Solomon KR, Adolphson LD, Wank DA, McHugh KP, Hauschka PV, 2000, Caveolae in human and murine osteoblasts. J Bone Miner Res 15:2391–2401. , DOI 10.1359/jbmr.2000.15.12.2391 Wiechen K, Sers C, Agoulnik A, Arlt K, Dietel M, Schlag PM, Schneider U, 2001, Down-regulation of caveolin-1, a candidate tumor suppressor gene, in sarcomas. Am J Pathol 158:833–839. , DOI 10.1016/S0002-9440(10)64031-X Belanger MM, Roussel E, Couet J, 2004, Caveolin-1 is downregulated in human lung carcinoma and acts as a candidate tumor suppressor gene. Chest 125(5 suppl):106S. , DOI 10.1378/ chest.125.5_suppl.106S Wiechen K, Diatchenko L, Agoulnik A, Scharff KM, Schober H, Arlt K, Zhumabayeva B, Siebert PD, Dietel M, Schafer R, Sers C, 2001, Caveolin-1 is down-regulated in human ovarian carcinoma and acts as a candidate tumor suppressor gene. Am J Pathol 159: 1635–1643. , DOI 10.1016/S0002-9440(10)63010-6 Kato K, Hida Y, Miyamoto M, Hashida H, Shinohara T, Itoh T, Okushiba S, Kondo S, Katoh H, 2002, Overexpression of caveolin-1 in esophageal squamous cell carcinoma correlates with lymph node metastasis and pathologic stage. Cancer 94:929–933. , DOI 10.1002/cncr.10329 YangG, Truong LD,Wheeler TM, Thompson TC, 1999, Caveolin- 1 expression in clinically confined human prostate cancer: a novel prognostic marker. Cancer Res 59:5719–5723 Cantiani L, Manara MC, Zucchini C, De Sanctis P, Zuntini M, Valvassori L, Serra M, Olivero M, Di Renzo MF, Colombo MP, Picci P, Scotlandi K, 2007, Caveolin-1 Reduces Osteosarcoma Metastases by Inhibiting c-Src Activity and Met Signaling. Cancer Res 67(16):7675–7685. , DOI 10.1158/0008- 5472.CAN-06-4697 Carver LA, Schnitzer JE, 2003, Caveolae: mining little caves for new cancer targets. Nat Rev Cancer 3:571–581. , DOI 10. 1038/nrc1146 Krajewska WM, Maslowska I, 2004, Caveolins: structure and function in signal transduction. Cell Mol Biol Lett 9:195–220 Williams TM, LisantiMP, 2005, Caveolin-1 in oncogenic transformation, cancer, andmetastasis.AmJ Physiol Cell Physiol 288:494– 506. , DOI 10.1152/ajpcell.00458.2004 Liu J, Wang XB, Park DS, Lisanti MP, 2002, Caveolin-1 expression enhances endothelial capillary tubule formation. J Biol Chem 277:10661–10668. , DOI 10.1074/jbc.M110354200 Deurs B, Roepstorff K, Hommelgaard AM, Sandvig K, 2003, Caveolae: anchored, multifunctional platforms in the lipid ocean. Trends Cell Biol 13:92–100. , DOI 10.1016/S0962- 8924(02)00039-9 Razani B, Woodman SE, Lisanti MP, 2002, Caveolae: from cell biology to animal physiology. Pharmacol Rev 54:431–467 Feron O, Kelly RA, 2001, The caveolar paradox: suppressing, inducing, and terminating eNOS signaling. Circ Res 88:129–131 Chen J, Braet F, Brodsky S, Weinstein T, Romanov V, Noiri E, Goligorsky MS, 2002, VEGF-induced mobilization of caveolae and increase in permeability of endothelial cells. Am J Phys 282(5):C1053–C1063. , DOI 10.1152/ajpcell.00292.2001 Shi L, Chen XM, Wang L, Zhang L, Chen Z, 2007, Expression of Caveolin-1 in Mucoepidermoid Carcinoma of the Salivary Glands: Correlation with Vascular Endothelial Growth Factor, Microvessel Density, and Clinical Outcome. CANCER 109(8):1523–1531. , DOI 10.1002/cncr.22573 Tahir SA, Park S, Thompson TC, 2009, Caveolin-1 regulates VEGF-stimulated angiogenic activities in prostate cancer and endothelial cells. Cancer Biol Ther 8(23):2286–2296 Gamallo C, Palacios J, Moreno G, Calvo deMora J, Suarez A, Armas A, 1999, Beta-catenin expression pattern in stage I and II ovarian carcinomas: relationship with beta-catenin gene mutations, clinicopathological features, and clinical outcome.AmJ Pathol 155:527–536 Galbiati F, Volonte D, Engelman JA et al, 1998, Targeted downregulation of caveolin-1 is sufficient to drive cell transformation and hyperactivate the p42/44 MAP kinase cascade. EMBO J 17:6633–6648 Koleske AJ, Baltimore D, LisantiMP, 1995, Reduction of caveolin and caveolae in oncogenically transformed cells. Proc Natl Acad Sci U S A 92:1381–1385 Sloan EK, Stanley KL, Anderson RL, 2004, Caveolin-1 inhibits breast cancer growth and metastasis. Oncogene 23:7893–7897 Hu YC, Lam KY, Law S et al, 2001, Profiling of differentially expressed cancer-related genes in esophageal squamous cell carcinoma, ESCC, using human cancer cDNAarrays: overexpression of oncogeneMETcorrelates with tumor differentiation in ESCC. Clin Cancer Res 7:3519–3525 Fong A, Garcia E, Gwynn L et al, 2003, Expression of caveolin-1 and caveolin-2 in urothelial carcinoma of the urinary bladder correlates with tumor grade and squamous differentiation. Am J Clin Pathol 120:93–100 Li L,YangG, Ebara S et al, 2001, Caveolin-1mediates testosterone stimulate survival/clonal growth and promotes metastatic activities in prostate cancer cells. Cancer Res 61:4386–4392 Tirado OM, Mateo-Lozano S, Villar J, Dettin LE, Llort A, Gallego S, Ban J, Kovar H, Notario V, 2006, Caveolin-1, CAV1, is a target of EWS/FLI-1 and a key determinant of the oncogenic phenotype and tumorigenicity of Ewing’s sarcoma cells. Cancer Res 66:9937– 9947. , DOI 10.1158/0008-5472.CAN-06-0927 Lee H, Volonte D, Galbiati F, Lyengar P, Lublin MD, Bregman DB, Wilson MT, Campos-Gonzalez R, Bouzahzah B, Pestell RG, Scherer PE, Lisanti MP, 2000, Constitutive and growth factor-regulated phosphorylation of caveolin-1 occurs at the same site, Tyr-14, in vivo: identification of a c-Src/ Cav-1/Grb7 signaling cassette. Mol Endocrinol 14:1750–1775. , DOI 10.1210/mend.14.11.0553 Oda Y, Yamamoto H, Tamiya S, Matsuda S, Tanaka K, Yokoyama R, Iwamoto Y, Tsuneyoshi M, 2006, CXCR4 and VEGF expression in the primary site and the metastatic site of human osteosarcoma: analysis within a group of patients, all of whom developed lung metastasis. Mod Pathol 19(5):738–745. , DOI 10. 1038/modpathol.3800587 Bajpai J, Sharma M, Sreenivas V, Kumar R, Gamnagatti S, Khan SA, Rastogi S, Malhotra A, Bakhshi S, 2009, VEGF expression as a prognostic marker in osteosarcoma. Pediatr Blood Cancer 53(6): 1035–1039. , DOI 10.1002/pbc.22178 Lammli J, Fan M, Rosenthal HG, Patni M, Rinehart E, Vergara G, Ablah E, Wooley PH, Lucas G, Yang SY, 2012, Expression of Vascular Endothelial Growth Factor correlates with the advance of clinical osteosarcoma. Int Orthop 36(11):2307– 2313. , DOI 10.1007/s00264-012-1629-z Lugowska I,WozniakW, Klepacka T, Michalak E, Szamotulska K, 2011, A prognostic evaluation of vascular endothelial growth factor in children and young adults with osteosarcoma. Pediatr Blood Cancer 57(1):63–68. , DOI 10.1002/pbc.23021 Becker RG, Galia CR, Morini S, Viana CR, 2013, Immunohistochemical expression of VEGF and her-2 proteins in osteosarcoma biopsies. Acta Ortopedica Brasileira 21(4):233–238. , DOI 10.1590/S1413-78522013000400010 Baptista AM, Camargo AF, Filippi RZ, Oliveira CR, Azevedo Neto RS, Camargo OP, 2014, Correlation between the expression of vegf and Survival in osteosarcoma. Acta Ortopedica Brasileira 22(5):250–255. , DOI 10.1590/1413-78522014220500978 Zhao J, Zhang Z, Zhao N,Ma BA, Fan QU, 2015, VEGF silencing inhibits human osteosarcoma angiogenesis and promotes cell apoptosis via PI3K/AKT signaling pathway. Cell Biochem Biophys 73(2):519–525. , DOI 10.1007/s12013-015-0692-7 Mizobuchi H, Garcia-Castellano JM, Philip S, Healey JH, Gorlick R, 2008, Hypoxia markers in human osteosarcoma: an exploratory study. Clin Orthop Relat Res 466(9):2052–2059. , DOI 10. 1007/s11999-008-0328-y Zhou Q, Zhu Y, Deng Z, Long H, Zhang S, Chen X, 2011, VEGF and EMMPRIN expression correlates with survival of patients with osteosarcoma. Surg Oncol 20(1):13–19. , DOI 10.1016/j. suronc.2009.09.002 Hassan SE, Bekarev M, Kim MY, Lin J, Piperdi S, Gorlick R, Geller DS, 2012, Cell surface receptor expression patterns in osteosarcoma. Cancer 118:740–749. , DOI 10.1002/cncr.26339 Chen D, Zhang YJ, Zhu KW, Wang WC, 2013, A systematic review of vascular endothelial growth factor expression as a biomarker of prognosis in patients with osteosarcoma. Tumour Biol 34(3): 1895–1899. , DOI 10.1007/s13277-013-0733-z Ohba T, Cates JM, Cole HA, Slosky DA, Haro H, Ando T, Schwartz HS, Schoenecker JG, 2014, Autocrine VEGF/VEGFR1 signaling in a subpopulation of cells associates with aggressive osteosarcoma. Mol Cancer Res 12:1100–1111. , DOI 10. 1158/1541-7786.MCR-14-0037 Yu XW, Wu TY, Yi X, Ren WP, Zhou ZB, Sun YQ, Zhang CQ, 2014, Prognostic significance of VEGF expression in osteosarcoma: a meta-analysis. Tumour Biol 35:155–160. , DOI 10. 1007/s13277-013-1019-1 Daft PG, Yang Y, Napierala D, ZayzafoonM(2015, The Growth and Aggressive Behavior of Human Osteosarcoma Is Regulated by a CaMKII-Controlled Autocrine VEGF Signaling Mechanism. PLoS One 10(4):1–20. e0121568. , DOI 10.1371/journal.Pone Tang Y, Zeng X, He F, Liao Y, Qian N, ToiM, 2012, Caveolin-1 is related to invasion, survival, and poor prognosis in hepatocellular cancer.Med Oncol 29(2):977–984. , DOI 10.1007/s12032- 011-9900-5 Hoffman R, 2004, Do the signalling proteins for angiogenesis exist as a modular complex? The case for the angosome. Med Hypotheses 63:675–680 Wu T, Zhang B, Ye F, Xiao Z, 2013, A potential role for caveolin-1 in VEGF-induced fibronectin upregulation in mesangial cells: involvement of VEGFR2 and Src. Am J Physiol Ren Physiol 304: F820–F830. , DOI 10.1152/ajprenal.00294.2012 Liu J, Razani B, Tang S, Terman BI, Ware JA, Lisanti MP, 1999, Angiogenesis activators and inhibitors differentially regulate caveolin-1 expression and caveolae formation in vascular endothelial cells. Angiogenesis inhibitors block vascular endothelial growth factor-induced downregulation of caveolin-1. J Biol Chem 274: 15781–15785. , DOI 10.1074/jbc.274.22.15781 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1787 EP 1795 DI 10.1007/s12253-019-00755-5 PG 9 ER PT J AU Sarnyai, F Szekerczes, T Csala, M Sumegi, B Szarka, A Schaff, Zs Mandl, J AF Sarnyai, Farkas Szekerczes, Timea Csala, Miklos Sumegi, Balazs Szarka, Andras Schaff, Zsuzsa Mandl, Jozsef TI BGP-15 Protects Mitochondria in Acute, Acetaminophen Overdose Induced Liver Injury SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Mitochondria; Acetaminophen; Liver injury; BGP-15; Reduced glutathione; Jun-kinase ID Mitochondria; Acetaminophen; Liver injury; BGP-15; Reduced glutathione; Jun-kinase AB Acetaminophen (APAP) induced hepatotoxicity involves activation of c-Jun amino-terminal kinase (JNK), mitochondrial damage and ER stress. BGP-15, a hydroximic acid derivative, has been reported to have hepatoprotective effects in APAP overdose induced liver damage. Effect of BGP-15 was further investigated on mitochondria in APAP-overdose induced acute liver injury in mice. We found that BGP-15 efficiently preserved mitochondrial morphology, and it caused a marked decrease in the number of damaged mitochondria. Attenuation of mitochondrial damage by BGP-15 is supported by immunohistochemistry as the TOMM20 label and the co-localized autophagy markers detected in the livers of APAP-treated mice were markedly reduced upon BGP-15 administration. This effect, along with the observed prevention of JNK activation likely contribute to the mitochondrial protective action of BGP-15. C1 [Sarnyai, Farkas] Semmelweis University, Department of Medical Chemistry, Molecular Biology and PathobiochemistryBudapest, Hungary. [Szekerczes, Timea] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Csala, Miklos] Semmelweis University, Department of Medical Chemistry, Molecular Biology and PathobiochemistryBudapest, Hungary. [Sumegi, Balazs] University of Pecs, Institute of Biochemistry and Medical ChemistryPecs, Hungary. [Szarka, Andras] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food Science, Laboratory of Biochemistry and Molecular BiologyBudapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi ut 93, H-1091 Budapest, Hungary. [Mandl, Jozsef] Semmelweis University, Department of Medical Chemistry, Molecular Biology and PathobiochemistryBudapest, Hungary. RP Schaff, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM schaff.zsuzsa@med.semmelweis-univ.hu CR Moynihan R, 2002, FDA fails to reduce accessibility of paracetamol despite 450 deaths a year - Confidential documents from the US Food and Drug Administration suggest that the agency has avoided a debate on tough new measures to reduce overdoses from painkillers-to avoid offending the pharmaceutical industry. Br Med J 325:678–678. , DOI 10.1136/bmj.325.7366.678 Lancaster EM, Hiatt JR, Zarrinpar A, 2015, Acetaminophen hepatotoxicity: an updated review. Arch Toxicol 89:193–199. https:// doi.org/10.1007/s00204-014-1432-2 Jaeschke H, 2015, Acetaminophen: dose-dependent drug hepatotoxicity and acute liver failure in patients. Dig Dis 33:464–471. , DOI 10.1159/000374090 Nagy G, Kardon T, Wunderlich L, Szarka A, Kiss A, Schaff Z, Banhegyi G, Mandl J, 2007, Acetaminophen induces ER dependent signaling in mouse liver. Arch Biochem Biophys 459:273– 279. , DOI 10.1016/j.abb.2006.11.021 Nagy G, Szarka A, Lotz G, Doczi J,Wunderlich L, Kiss A, Jemnitz K, Veres Z, Banhegyi G, Schaff Z, Sumegi B,Mandl J, 2010, BGP- 15 inhibits caspase-independent programmed cell death in acetaminophen-induced liver injury. Toxicol Appl Pharmacol 243: 96–103. , DOI 10.1016/j.taap.2009.11.017 Lorincz T, Jemnitz K, Kardon T, Mandl J, Szarka A, 2015, Ferroptosis is involved in acetaminophen induced cell death. Pathol Oncol Res 21: 1115–1121. , DOI 10.1007/s12253-015-9946-3 Qiu Y, Benet LZ, Burlingame AL, 1998, Identification of the hepatic protein targets of reactive metabolites of acetaminophen in vivo in mice using two-dimensional gel electrophoresis and mass spectrometry. J Biol Chem 273:17940–17953 McGill MR, Williams CD, Xie Y, Ramachandran A, Jaeschke H, 2012, Acetaminophen-induced liver injury in rats and mice: comparison of protein adducts, mitochondrial dysfunction, and oxidative stress in the mechanism of toxicity. Toxicol Appl Pharmacol 264: 387–394. , DOI 10.1016/j.taap.2012.08.015 Manyike PT, Kharasch ED, Kalhorn TF, Slattery JT, 2000, Contribution of CYP2E1 and CYP3A to acetaminophen reactive metabolite formation. Clin Pharmacol Ther 67:275–282. https:// doi.org/10.1067/mcp.2000.104736 TirmensteinMA, Nelson SD(1989, Subcellular binding and effects on calcium homeostasis produced by acetaminophen and a nonhepatotoxic regioisomer, 3′-hydroxyacetanilide, in mouse liver. J Biol Chem 264:9814–9819 McGill MR, Jaeschke H, 2013, Metabolism and disposition of acetaminophen: recent advances in relation to hepatotoxicity and diagnosis. Pharm Res 30:2174–2187. , DOI 10.1007/ s11095-013-1007-6 Meyers LL, Beierschmitt WP, Khairallah EA, Cohen SD, 1988, Acetaminophen-induced inhibition of hepatic mitochondrial respiration in mice. Toxicol Appl Pharmacol 93:378–387 Jaeschke H, 1990, Glutathione disulfide formation and oxidant stress during acetaminophen-induced hepatotoxicity in mice in vivo: the protective effect of allopurinol. J Pharmacol Exp Ther 255:935–941 Cover C, Mansouri A, Knight TR, BajtML, Lemasters JJ, Pessayre D, Jaeschke H, 2005, Peroxynitrite-induced mitochondrial and endonuclease-mediated nuclear DNA damage in acetaminophen hepatotoxicity. J Pharmacol Exp Ther 315:879–887. https://doi. org/10.1124/jpet.105.088898 Seki E, Brenner DA, KarinM(2012, A liver full of JNK: signaling in regulation of cell function and disease pathogenesis, and clinical approaches. Gastroenterology 143:307–320. , DOI 10. 1053/j.gastro.2012.06.004 Green DR, Galluzzi L, Kroemer G, 2014, Cell biology. Metabolic control of cell death. Science 345:1250256. , DOI 10.1126/ science.1250256 Bajt ML, Cover C, Lemasters JJ, Jaeschke H, 2006, Nuclear translocation of endonuclease G and apoptosis-inducing factor during acetaminophen-induced liver cell injury. Toxicol Sci 94:217–225. , DOI 10.1093/toxsci/kfl077 Chung J, Nguyen AK, Henstridge DC, Holmes AG, Chan MH, Mesa JL, Lancaster GI, Southgate RJ, Bruce CR, Duffy SJ, Horvath I, Mestril R, Watt MJ, Hooper PL, Kingwell BA, Vigh L, Hevener A, Febbraio MA, 2008, HSP72 protects against obesityinduced insulin resistance. Proc Natl Acad Sci U S A 105:1739– 1744. , DOI 10.1073/pnas.0705799105 Henstridge DC, Bruce CR, Drew BG, Tory K, Kolonics A, Estevez E, Chung J, Watson N, Gardner T, Lee-Young RS, Connor T, Watt MJ, Carpenter K, Hargreaves M, McGee SL, Hevener AL, Febbraio MA, 2014, Activating HSP72 in rodent skeletal muscle increases mitochondrial number and oxidative capacity and decreases insulin resistance. Diabetes 63:1881–1894. , DOI 10.2337/db13-0967 Gehrig SM, van der Poel C, Sayer TA, Schertzer JD, Henstridge DC, Church JE, Lamon S, Russell AP, Davies KE, Febbraio MA, Lynch GS, 2012, Hsp72 preserves muscle function and slows progression of severe muscular dystrophy. Nature 484:394–398. , DOI 10.1038/nature10980 Sapra G, Tham YK, Cemerlang N, Matsumoto A, Kiriazis H, Bernardo BC, Henstridge DC, Ooi JY, Pretorius L, Boey EJ, Lim L, Sadoshima J, Meikle PJ, Mellet NA, Woodcock EA, Marasco S, Ueyama T, Du XJ, Febbraio MA, McMullen JR, 2014, The small-molecule BGP-15 protects against heart failure and atrial fibrillation in mice. Nat Commun 5:5705. , DOI 10.1038/ncomms6705 Wu LL, Russell DL, Wong SL, Chen M, Tsai TS, St John JC, Norman RJ, Febbraio MA, Carroll J, Robker RL, 2015, Mitochondrial dysfunction in oocytes of obese mothers: transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors. Development 142:681–691. https://doi. org/10.1242/dev.114850 Szabo A, Sumegi K, Fekete K, Hocsak E, Debreceni B, Setalo G Jr, Kovacs K, Deres L, Kengyel A, Kovacs D, Mandl J, Nyitrai M, Febbraio MA, Gallyas F Jr, Sumegi B, 2018, Activation of mitochondrial fusion provides a new treatment for mitochondria-related diseases. Biochem Pharmacol 150:86–96. , DOI 10.1016/j. bcp.2018.01.038 Farkas B,MagyarlakiM, Csete B,Nemeth J, RabloczkyG, Bernath S, Literati Nagy P, Sumegi B, 2002, Reduction of acute photodamage in skin by topical application of a novel PARP inhibitor. Biochem Pharmacol 63:921–932 Literati-Nagy B, Kulcsar E, Literati-Nagy Z, Buday B, Peterfai E, Horvath T, Tory K, Kolonics A, Fleming A, Mandl J, Koranyi L, 2009, Improvement of insulin sensitivity by a novel drug, BGP-15, in insulin-resistant patients: a proof of concept randomized doubleblind clinical trial.HormMetab Res 41:374–380. , DOI 10. 1055/s-0028-1128142 Vigh L, Literati PN, Horvath I, Torok Z, Balogh G,GlatzA, Kovacs E, Boros I, Ferdinandy P, Farkas B, Jaszlits L, Jednakovits A, Koranyi L, Maresca B, 1997, Bimoclomol: a nontoxic, hydroxylamine derivative with stress protein-inducing activity and cytoprotective effects. Nat Med 3:1150–4.27 Han D, Dara L, Win S, Than TA, Yuan L, Abbasi SQ, Liu ZX, Kaplowitz N, 2013, Regulation of drug-induced liver injury by signal transduction pathways: critical role of mitochondria. Trends Pharmacol Sci 34:243–253. , DOI 10.1016/j.tips.2013.01.009 Hanawa N, ShinoharaM, Saberi B, GaardeWA, Han D, Kaplowitz N, 2008, Role of JNK translocation to mitochondria leading to inhibition of mitochondria bioenergetics in acetaminopheninduced liver injury. J Biol Chem 283:13565–13577. https://doi. org/10.1074/jbc.M708916200 Kon K, Kim JS, Jaeschke H, Lemasters JJ, 2004, Mitochondrial permeability transition in acetaminophen-induced necrosis and apoptosis of cultured mouse hepatocytes. Hepatology 40:1170–1179. , DOI 10.1002/hep.20437 Gujral JS, Knight TR, Farhood A, Bajt ML, Jaeschke H, 2002, Mode of cell death after acetaminophen overdose inmice: apoptosis or oncotic necrosis? Toxicol Sci 67:322–328 Czaja MJ, Ding WX, Donohue TM Jr, Friedman SL, Kim JS, Komatsu M, Lemasters JJ, Lemoine A, Lin JD, Ou JH, Perlmutter DH, Randall G, Ray RB, Tsung A, Yin XM, 2013, Functions of autophagy in normal and diseased liver. Autophagy. 9(8):1131–1158. , DOI 10.4161/auto.25063 Kardon T, Nagy G, Csala M,Kiss A, Schaff Z,Nagy PL,Wunderlich L, Banhegyi G, Mandl J, 2006, Influence of BGP-15, a nicotinic Amidoxime derivative, on the vascularization and growth of murine Hepatoma Xenografts. Anticancer Res 26:1023–1028 Du K, Williams CD, McGill MR, Jaeschke H, 2014, Lower susceptibility of female mice to acetaminophen hepatotoxicity: role of mitochondrial glutathione, oxidant stress and c-Jun N-terminal kinase. Toxicol Appl Pharmacol 281:58–66. , DOI 10.1016/ j.taap.2014.09.002 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1797 EP 1803 DI 10.1007/s12253-019-00721-1 PG 7 ER PT J AU Zengin, M AF Zengin, Mehmet TI Local Inflammatory Response Can Predict Clinical Outcome in Patients with Curatively Resected Stage-IIB Colon Cancer: An Advanced Methodological Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Local inflammatory response; tumour biomarkers; colon cancers; stage -IIB ID Local inflammatory response; tumour biomarkers; colon cancers; stage -IIB AB Purpose: Although local inflammatory response (LIR) is a reliable survival marker in colon cancers (CCs), there is no consensus on its use in daily practice. We investigated the prognostic value of LIR in a highly homogeneous population with a well-designed methodology. Methods: Eighty stage-IIB CC patients operated between 2002 and 2012 were included in the study. Standardization was investigated for extra-biopsy evaluation methods (magnification, staining, and counting). Model A was used for intra-biopsy evaluation methods (block, section, and focus). So, this study makes important contributions to the standardization of pathological evaluations. Results: In method 1, the following analyzes showed more successful results for LIR: relationship with prognostic factors [tumour deposits (p=0.017), Crohn’s-like reaction (p=0.019), advanced grade, (p=0.012), positive surgical margin (p=0.019), perineural invasion (p=0.025), mismatch repair proteins-proficiency (p=0.031)], reproducibility of the study (Kappa=0.49–0.73, Intra-class correlation=0.442–0.724), and correlation of estimates (r=0.704). The cut-off value was also quite useful (area of under ROC=0.820 [0.694-0.920]). In univariate analysis, low LIR was related to poor overall survival (OS; p<0.001) and poor relapse-free survival (RFS, p=0.001) . Multivariate analysis confirmed that low LIR is an independent poor survival marker for OS (Hazard Ratio [HR]=1.32 [1.08-1.61, p=0.005) and RFS (HR=1.50 [1.22-1.85], p<0.001). Conclusions: Our results showed that low LIR had an independent prognostic significance in stage -IIB CCs.We also recommend using model A and method 1 for successful results and standardization. C1 [Zengin, Mehmet] Kirikkale University, Department of PathologyKirikkale, Turkey. RP Zengin, M (reprint author), Kirikkale University, Department of Pathology, Kirikkale, Turkey. EM mz1379@hotmail.com CR Karim S, Brennan K, Nanji S et al, 2017). Association between prognosis and tumor laterality in early-stage colon cancer. JAMA Oncol 2017; 3: 1386-92 Cunningham D, Atkin W, Lenz HJ et al, 2010, Colorectal cancer. Lancet 375:1030–1047 Quasar Collaborative Group, Gray R, Barnwell J et al, 2007, Adjuvant chemotherapy versus observation in patients with colorectal cancer: a randomised study. Lancet 370:2020–2029 Benson AB 3rd, Schrag D, Somerfield MR et al, 2004, American Society of Clinical Oncology recommendations on adjuvant chemotherapy for stage II colon cancer. J Clin Oncol 22:3408–3419 Colotta F, Allavena P, Sica A et al, 2009). Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 30:1073–1081 Roxburgh CS, McMillan DC, 2010, Role of systemic inflammatory response in predicting survival in patients with primary operable cancer. Future Oncol 6:149–163 Mei Z, Liu Y, Liu C et al, 2014, Tumour-infiltrating inflammation and prognosis in colorectal cancer: systematic review and metaanalysis. Br J Cancer 110:1595–1605 Leitch EF, Chakrabarti M, Crozier JE et al, 2007, Comparison of the prognostic value of selected markers of the systemic inflammatory response in patients with colorectal cancer. Br J Cancer 97: 1266–1270 Menon AG, Janssen-van Rhijn CM, Morreau H et al, 2004, Immune system and prognosis in colorectal cancer: a detailed immunohistochemical analysis. Lab Invest 84:493–501 Ishizuka M, Nagata H, Takagi K et al, 2009, Influence of inflammation-based prognostic score on mortality of patients undergoing chemotherapy for far advanced or recurrent unresectable colorectal cancer. Ann Surg 250:268–272 Dong ZY, Wu SP, Liao RQ et al, 2016, Potential biomarker for checkpoint blockade immunotherapy and treatment strategy. Tumour Biol 37:4251–4261 Zengin M, 2019, Prognostic role of Tumor-infiltrating T lymphocytes in stage IIA, T3N0, colon cancer: A broad methodological study in a fairly homogeneous population. Annals of Diagnostic Pathology 41:69–78 McShane LM, Altman DG, Sauerbrei W et al, 2005, REporting recommendations for tumour MARKer prognostic studies, REMARK). Br J Cancer 93:387–391 Sobin LH, Compton CC, 2010). TNM seventh edition: what's new, what's changed: communication from the International Union Against Cancer and the American Joint Committee on Cancer. Cancer 116: 5336-9. Kamarudin AN, Cox T, Kolamunnage-Dona R, 2017, Timedependent ROC curve analysis in medical research: current methods and applications. BMC Med Res Methodol 17:53 McGraw KO, Wong SP, 1996, Forming inferences about some intraclass correlation coefficients. Psychol Methods 1:30–46 Landis JR, Koch GG, 1977, The measurement of observer agreement for categorical data. Biometrics 33:159–174 Nosho K, Baba Y, Tanaka N et al, 2010, Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review. J Pathol 222:350–366 Galon J, Costes A, Sanchez-Cabo F et al, 2006, Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313:1960–1964 Ropponen KM, Eskelinen MJ, Lipponen PK et al, 1997, Prognostic value of tumour-infiltrating lymphocytes, TILs, in colorectal cancer. J Pathol 182:318–324 Pages F, Berger A, Camus M et al, 2005, Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med 353:2654–2666 Laghi L, Bianchi P, Miranda E et al, 2009, CD3+ cells at the invasive margin of deeply invading, pT3-T4, colorectal cancer and risk of post-surgical metastasis: a longitudinal study. Lancet Oncol 10:877–884 DahlinAM, HenrikssonML,VanGuelpen B et al, 2011, Colorectal cancer prognosis depends on T-cell infiltration and molecular characteristics of the tumor. Mod Pathol 24:671–682 Foxtrot CG, 2012, Feasibility of preoperative chemotherapy for locally advanced, operable colon cancer: the pilot phase of a randomised controlled trial. Lancet Oncol 13:1152–1160 Prall F, Duhrkop T,Weirich Vet al, 2004, Prognostic role of CD8+ tumor-infiltrating lymphocytes in stage III colorectal cancer with and without microsatellite instability. Hum Pathol 35:808–816 Klintrup K,Makinen JM, Kauppila S et al, 2005, Inflammation and prognosis in colorectal cancer. Eur J Cancer 41:2645–2654 Nagtegaal ID,Marijnen CA, Kranenbarg EK et al, 2001, Local and distant recurrences in rectal cancer patients are predicted by the nonspecific immune response; specific immune response has only a systemic effect: a histopathological and immunohistochemical study. BMC Cancer 1:7 Berntsson J, Nodin B, Eberhard J et al, 2016, Prognostic impact of tumour-infiltrating B cells and plasma cells in colorectal cancer. Int J Cancer. 139(5):1129–1139 Guidoboni M, Gafa R, Viel A et al, 2001, Microsatellite instability and high content of activated cytotoxic lymphocytes identify colon cancer patients with a favorable prognosis. Am J Pathol 159:297– 304 Turksma AW, Coupe VM, Shamier MC et al, 2016, Extent and location of tumor-infiltrating lymphocytes in microsatellite-stable colon cancer predict outcome to adjuvant active specific immunotherapy. Clin Cancer Res 22:346–356 Galon J, Pages F, Marincola FM et al, 2012, Cancer classification using the Immunoscore: a worldwide task force. J Transl Med 10: 205 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1805 EP 1816 DI 10.1007/s12253-019-00758-2 PG 12 ER PT J AU Usman, M Ilyas, A Hashim, Z Shamshad, Z AF Usman, Muhammad Ilyas, Amber Hashim, Zehra Shamshad, Zarina TI Identification of GIMAP7 and Rabl3 as Putative Biomarkers for Oral Squamous Cell Carcinoma Through Comparative Proteomic Approach SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Serum biomarkers; OSCC; GIMAP7; Rabl3; Proteomics ID Serum biomarkers; OSCC; GIMAP7; Rabl3; Proteomics AB Oral squamous cell carcinoma (OSCC) accounts for more than 90% of all oral cancers and has been listed as sixth most common human cancer. Due to late diagnosis and insufficient therapeutic response among patients, the survival rate remains very low accentuating the importance of early diagnostic markers. The study aimed to identify differentially expressed proteins in search for putative serum biomarkers and drug targets. Serum samples (n = 45) were depleted and resolved on two dimensional gel electrophoresis. Among differentially expressed proteins, two were identified using MALDI-TOF mass spectrometry. Gene expression levels of identified proteins were quantified in malignant and normal tissue using RT-qPCR. To validate serum Rabl3 expression, sandwich ELISA was performed. Proteomics analysis revealed two proteins which were found to be associated with oral cancer. The expression of GIMAP7 was found to be down regulated in serum of patients suffering from oral cancer while the expression of Rabl3 was found to be up-regulated. Gene expression analysis in malignant tissue and adjacent normal tissue revealed the same pattern. Quantitative ELISA was used to validate expression of Rabl3 in serum from oral cancer patients and healthy subjects which demonstrated significant up-regulation in cancer patients. Findings in current study demonstrate differential expression of novel putative biomarkers GIMAP7 and Rabl3 in oral cancer which suggests their potential role in oral cancer pathology and can be considered as predictive biomarkers. C1 [Usman, Muhammad] University of Karachi, National Center for Proteomics, 75270 Karachi, Pakistan. [Ilyas, Amber] University of Karachi, National Center for Proteomics, 75270 Karachi, Pakistan. [Hashim, Zehra] University of Karachi, National Center for Proteomics, 75270 Karachi, Pakistan. [Shamshad, Zarina] University of Karachi, National Center for Proteomics, 75270 Karachi, Pakistan. RP Shamshad, Z (reprint author), University of Karachi, National Center for Proteomics, 75270 Karachi, Pakistan. EM szarina@uok.edu.pk CR Leemans CR, Braakhuis BJ, Brakenhoff RH, 2011, The molecular biology of head and neck cancer. Nat Rev Cancer 11(1):9–22. , DOI 10.1038/nrc2982 Arantes LMRB, de Carvalho AC, Melendez ME, Carvalho AL, Goloni-Bertollo EM, 2014, Methylation as a biomarker for head and neck cancer. Oral Oncol 50(6):587–592. , DOI 10. 1016/j.oraloncology.2014.02.015 Tanaka T, TanakaM, Tanaka T, 2011, Oral carcinogenesis and oral cancer chemoprevention: a review. Pathol Res Int 2011:431246– 431246. , DOI 10.4061/2011/431246 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6):394–424. , DOI 10.3322/ caac.21492 Ali NS, Khuwaja AK, Ali T, Hameed R, 2009, Smokeless tobacco use among adult patients who visited family practice clinics in Karachi, Pakistan. J Oral Pathol Med 38(5):416–421 Johnson N, 2001, Tobacco use and oral cancer: a global perspective. J Dent Educ 65(4):328–339 Khan Z, Tonnies J, Muller S, 2014, Smokeless tobacco and oral cancer in South Asia: a systematic review with meta-analysis. J Cancer Epidemiol 2014:394696–394696. , DOI 10.1155/ 2014/3946960 Gigliotti J, Madathil S, Makhoul N, 2019, Delays in oral cavity cancer. Int J Oral Maxillofac Surg. , DOI 10.1016/j.ijom. 2019.02.015 Barbosa EB, Vidotto A, Polachini GM, Henrique T, Marqui AB, Tajara EH, 2012, Proteomics: methodologies and applications to the study of human diseases. Rev Assoc Med Bras 58(3):366–375 Turhani D, Krapfenbauer K, Thurnher D, Langen H, Fountoulakis M, 2006, Identification of differentially expressed, tumorassociated proteins in oral squamous cell carcinoma by proteomic analysis. Electrophoresis 27(7):1417–1423. , DOI 10. 1002/elps.200500510 Malik UU, Zarina S, Pennington SR, 2016, Oral squamous cell carcinoma: key clinical questions, biomarker discovery, and the role of proteomics. Arch Oral Biol 63:53–65. , DOI 10.1016/j. archoralbio.2015.11.017 Blatt S, KrugerM, Ziebart T, Sagheb K, Schiegnitz E, Goetze E,Al- Nawas B, Pabst AM(2017, Biomarkers in diagnosis and therapy of oral squamous cell carcinoma: a review of the literature. J Craniomaxillofac Surg 45(5):722–730. , DOI 10.1016/j. jcms.2017.01.033 Beretta L, 2007, Proteomics from the clinical perspective: many hopes and much debate. Nat Methods 4(10):785–786. https://doi. org/10.1038/nmeth1007-785 Fernandez-Olavarria A, Mosquera-Perez R, Diaz-Sanchez R-M, Serrera-Figallo M-A, Gutierrez-Perez J-L, Torres-Lagares D, 2016, The role of serum biomarkers in the diagnosis and prognosis of oral cancer: a systematic review. J Clin Exp Dent 8(2):e184– e193. , DOI 10.4317/jced.52736 Ilyas A, Hashim Z, Zarina S, 2015, Effects of 5'-azacytidine and alendronate on a hepatocellular carcinoma cell line: a proteomics perspective. Mol Cell Biochem 405(1-2):53–61. , DOI 10. 1007/s11010-015-2395-1 Musharraf SG, Hashmi N, ChoudharyMI, Rizvi N, Usman A, Atta ur R, 2012, Comparison of plasma from healthy nonsmokers, smokers, and lung cancer patients: pattern-based differentiation profiling of low molecular weight proteins and peptides by magnetic bead technology withMALDI-TOFMS. Biomarkers 17(3):223– 230. , DOI 10.3109/1354750x.2012.657245 Bhurgri Y, 2005, Cancer of the oral cavity - trends in Karachi South, 1995-2002). Asian Pac J Cancer Prev 6(1):22–26 Chen Y, Azman SN, Kerishnan JP, Zain RB, Chen YN,Wong YL, Gopinath SC, 2014, Identification of host-immune response protein candidates in the sera of human oral squamous cell carcinoma patients. PLoS One 9(10):e109012. , DOI 10.1371/journal. pone.0109012 Nitta T, Nasreen M, Seike T, Goji A, Ohigashi I, Miyazaki T, Ohta T, Kanno M, Takahama Y, 2006, IAN family critically regulates survival and development of T lymphocytes. PLoS Biol 4(4):e103. , DOI 10.1371/journal.pbio.0040103 Poirier GM, Anderson G, Huvar A, Wagaman PC, Shuttleworth J, Jenkinson E, Jackson MR, Peterson PA, Erlander MG, 1999, Immune-associated nucleotide-1, IAN-1, is a thymic selection marker and defines a novel gene family conserved in plants. J Immunol 163(9):4960–4969 Pandarpurkar M, Wilson-Fritch L, Corvera S, Markholst H, Hornum L, Greiner DL, Mordes JP, Rossini AA, Bortell R, 2003, Ian4 is required formitochondrial integrity and Tcell survival. Proc Natl Acad Sci U S A 100(18):10382–10387. , DOI 10. 1073/pnas.1832170100 Schnell S, Demolliere C, van den Berk P, Jacobs H, 2006, Gimap4 accelerates T-cell death. Blood 108(2):591–599. , DOI 10. 1182/blood-2005-11-4616 Shiao YM, Chang YH, Liu YM, Li JC, Su JS, Liu KJ, Liu YF, Lin MW, Tsai SF, 2008, Dysregulation of GIMAP genes in non-small cell lung cancer. Lung Cancer 62(3):287–294. , DOI 10. 1016/j.lungcan.2008.03.021 Huang Z, ZhangW, Gao C, Ji B, Chi X, ZhengW,WangHL, 2016, Dysregulation of GTPase IMAP family members in hepatocellular cancer. Mol Med Rep 14(5):4119–4123. , DOI 10.3892/ mmr.2016.5764 Colicelli J, 2004, Human RAS superfamily proteins and related GTPases. Sci STKE 2004(250):Re13. , DOI 10.1126/ stke.2502004re13 Agarwal R, Jurisica I, Mills GB, Cheng KW, 2009, The emerging role of the RAB25 small GTPase in cancer. Traffic, Copenhagen, Denmark, 10(11):1561–1568. , DOI 10.1111/j.1600-0854. 2009.00969.x Li Q,Wang L, Zeng L, Zhang Y, Li K, Jin P, Su B,Wang L, 2010, Evaluation of the novel gene Rabl3 in the regulation of proliferation and motility in human cancer cells. Oncol Rep 24(2):433–440 Zhang W, Sun J, Luo J, 2016, High expression of Rab-like 3, Rabl3, is associated with poor survival of patients with nonsmall cell lung cancer via repression of MAPK8/9/10-Mediated autophagy. Med Sci Monit 22:1582–1588. , DOI 10. 12659/MSM.898632 Pan Y, Liu Z, Feng Z, Hui D, Huang X, Tong D, Jin Y, 2017, The overexpression of Rabl3 is associated with pathogenesis and clinicopathologic variables in hepatocellular carcinoma. Tumour Biol 39(4):1010428317696230. , DOI 10.1177/ 1010428317696230 An J, Liu Z, Liang Q, Pan Y, Li H, Wang R, Jin Y, 2017, Overexpression of Rabl3 and Cullin7 is associated with pathogenesis and poor prognosis in hepatocellular carcinoma. Hum Pathol 67:146–151. , DOI 10.1016/j.humpath.2017.07.008 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1817 EP 1822 DI 10.1007/s12253-019-00775-1 PG 6 ER PT J AU Eldin, EM Makboul, R Behnsawy, MH Abdelkawi, FI Moeen, MA Faddan, AA Gadelmoula, M Shahat, AA Abdel-Aziz, AM Hafez, MM Hameed, D AF Eldin, Emad Mohamed Makboul, Rania Behnsawy, M Hosny Abdelkawi, F Islam Moeen, M Ahmed Faddan, Abou Amr Gadelmoula, Mohamed Shahat, A Ahmed Abdel-Aziz, Atef Mohamed Hafez, M Moemen Hameed, A. Diaa TI Comparative Application of Different Substaging Techniques for Non-Muscle Invasive Urothelial Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Substaging; Non-muscle invasive; Urothelial carcinoma; Urinary bladder ID Substaging; Non-muscle invasive; Urothelial carcinoma; Urinary bladder AB To evaluate the diagnostic performance and clinical significance of 4 systems of substaging cases with non-muscle invasive urothelial bladder carcinoma. In addition 4 cutoff measures were evaluated for prediction of muscularis-mucosa invasion. Four substaging systems were applied to 57 NMIBC cases to assess which of these reported methods correlates best with recurrence and progression. On univariate regression analysis patients having tumor size more than 3 cm, solid tumor architecture, high grade, substage B, substage T1e, substage ROL 2 and Tumor depth more than 1 mm were associated with higher recurrence. On multivariate analysis all the four substaging systems, tumor size, grade and tumor type had significant prognostic value for recurrence. Regarding progression only the metric substaging method was associated with tumor progression (p = 0.04). However, on univariate and multivariate regression analysis none of the substaging systems showed prognostic significance and only solid tumor architecture and CIS had significant prognostic value for tumor progression. The ROC curve analysis showed that 1 mm depth of invasion had the best accuracy for detection of muscularis-mucosa invasion (80.2%). Using 1 mm cutoff in measuring the depth and 0.5 mm for the diameter of infiltration may provide clinically relevant information to guide a more personalized therapy for NMIBC. Inclusion of both measures in addition to other histopathologic variables may aid in the development of a scoring system. C1 [Eldin, Emad Mohamed] Assiut University, Urology and Nephrology HospitalAsyut, Egypt. [Makboul, Rania] Asyut University, Faculty of Medicine, Pathology DepartmentAssiut, Egypt. [Behnsawy, M Hosny] Assiut University, Urology and Nephrology HospitalAsyut, Egypt. [Abdelkawi, F Islam] Assiut University, Urology and Nephrology HospitalAsyut, Egypt. [Moeen, M Ahmed] Assiut University, Urology and Nephrology HospitalAsyut, Egypt. [Faddan, Abou Amr] Assiut University, Urology and Nephrology HospitalAsyut, Egypt. [Gadelmoula, Mohamed] Assiut University, Urology and Nephrology HospitalAsyut, Egypt. [Shahat, A Ahmed] Assiut University, Urology and Nephrology HospitalAsyut, Egypt. [Abdel-Aziz, Atef Mohamed] Assiut University, Urology and Nephrology HospitalAsyut, Egypt. [Hafez, M Moemen] Asyut University, Faculty of Medicine, Pathology DepartmentAssiut, Egypt. [Hameed, A. Diaa] Assiut University, Urology and Nephrology HospitalAsyut, Egypt. RP Abdelkawi, FI (reprint author), Assiut University, Urology and Nephrology Hospital, Asyut, Egypt. EM islamfaa@yahoo.com CR Ploeg M, Aben KK, Kiemeney LA, 2009, The present and future burden of urinary bladder cancer in the world. World J Urol 27(3): 289–293 Hu Z, Mudaliar K, Quek ML, Paner GP, Barkan GA, 2014, Measuring the dimension of invasive component in pT1 urothelial carcinoma in transurethral resection specimens can predict time to recurrence. Ann Diagn Pathol 18(2):49–52 van de Putte EE F, OttoW, Hartmann A, Bertz S,Mayr R, Brundl J, Breyer J, Manach Q, Comperat EM, Boormans JL et al, 2018, Metric substage according to micro and extensive lamina propria invasion improves prognostics in T1 bladder cancer. Urol Oncol 36(8):361e367–361e313 Patriarca C, Hurle R, Moschini M, Freschi M, Colombo P, Colecchia M, Ferrari L, Guazzoni G, Conti A, Conti G et al, 2016, Usefulness of pT1 substaging in papillary urothelial bladder carcinoma. Diagn Pathol 11:6 Magers MJ, Lopez-Beltran A, Montironi R, Williamson SR, Kaimakliotis HZ, Cheng L, 2019, Staging of bladder cancer. Histopathology 74(1):112–134 Otto W, van Rhijn BW, Breyer J, Bertz S, Eckstein M, Mayr R, Lausenmeyer EM, Denzinger S, Burger M, Hartmann A, 2018, Infiltrative lamina propria invasion pattern as an independent predictor for cancer-specific and overall survival of instillation treatment-naive stage T1 high-grade urothelial bladder cancer. Int J Urol 25(5):442–449 Cheng L, Weaver AL, Neumann RM, Scherer BG, Bostwick DG, 1999, Substaging of T1 bladder carcinoma based on the depth of invasion asmeasured by micrometer: a new proposal. Cancer 86(6): 1035–1043 Chang WC, Chang YH, Pan CC, 2012, Prognostic significance in substaging ofT1 urinary bladder urothelial carcinoma on transurethral resection. Am J Surg Pathol 36(3):454–461 van der Aa MN, van Leenders GJ, Steyerberg EW, van Rhijn BW, Jobsis AC, Zwarthoff EC, van der Kwast TH, 2005, A new system for substaging pT1 papillary bladder cancer: a prognostic evaluation. Human Pathol 36(9):981–986 van Rhijn BW, van der Kwast TH, Alkhateeb SS, Fleshner NE, van Leenders GJ, Bostrom PJ, van der Aa MN, Kakiashvili DM, Bangma CH, Jewett MA et al, 2012, A new and highly prognostic system to discern T1 bladder cancer substage. Eur Urol 61(2):378– 384 van den Bosch S, AlfredWitjes J, 2011, Long-term cancer-specific survival in patients with high-risk, non-muscle-invasive bladder cancer and tumour progression: a systematic review. Eur Urol 60(3):493–500 LeivoMZ, Sahoo D, Hamilton Z, Mirsadraei L, Shabaik A, Parsons JK, Kader AK, Derweesh I, Kane C, Hansel DE, 2018, Analysis of T1 bladder Cancer on biopsy and transurethral resection specimens: comparison and ranking of T1 quantification approaches to predict progression to Muscularis Propria invasion. Am J Surg Pathol 42(1):e1–e10 Brimo F,Wu C, Zeizafoun N, Tanguay S, Aprikian A, Mansure JJ, Kassouf W, 2013, Prognostic factors in T1 bladder urothelial carcinoma: the value of recording millimetric depth of invasion, diameter of invasive carcinoma, and muscularis mucosa invasion. Human Pathol 44(1):95–102 Paner GP,Montironi R, AminMB, 2017, Challenges in pathologic staging of bladder Cancer: proposals for fresh approaches of assessing pathologic stage in light of recent studies and observations pertaining to bladder Histoanatomic variances. Adv Anat Pathol 24(3):113–127 Kondylis FI, Demirci S, Ladaga L, Kolm P, Schellhammer PF, 2000, Outcomes after intravesical bacillus Calmette-Guerin are not affected by substaging of high grade T1 transitional cell carcinoma. J Urol 163(4):1120–1123 van de Putte EE F, BehrendtMA, Pigot GL, van der Kwast TH, van Rhijn BW, 2015, Prognostic significance of substage and WHO classification systems in T1 urothelial carcinoma of the bladder. Curr Opin Urol 25(5):427–435 Dem V, Cerruto MA, D'Elia C, Brunelli M, Otte O, Minja A, Luchini C, Novella G, Cavalleri S, Martignoni G et al, 2014, Prognostic role of substaging in T1G3 transitional cell carcinoma of the urinary bladder. Mol Clin Oncol 2(4):575–580 Nishiyama N, Kitamura H, Maeda T, Takahashi S, Masumori N, Hasegawa T, Tsukamoto T, 2013, Clinicopathological analysis of patients with non-muscle-invasive bladder cancer: prognostic value and clinical reliability of the 2004WHO classification system. Jpn J Clin Oncol 43(11):1124–1131 Bertz S, Denzinger S, Otto W, Wieland WF, Stoehr R, Hofstaedter F, Hartmann A, 2011, Substaging by estimating the size of invasive tumour can improve risk stratification in pT1 urothelial bladder cancer-evaluation of a large hospital-based single-Centre series. Histopathology 59(4):722–732 Orsola A, Werner L, de Torres I, Martin-Doyle W, Raventos CX, Lozano F, Mullane SA, Leow JJ, Barletta JA, Bellmunt J et al, 2015, Reexamining treatment of high-grade T1 bladder cancer according to depth of lamina propria invasion: a prospective trial of 200 patients. Br J Cancer 112(3):468–474 Martin-DoyleW, Leow JJ, Orsola A, Chang SL, Bellmunt J, 2015, Improving selection criteria for early cystectomy in high-grade t1 bladder cancer: a meta-analysis of 15,215 patients. J Clin Oncol 33(6):643–650 Gontero P, Sylvester R, Pisano F, Joniau S,Vander Eeckt K, Serretta V, Larre S, Di Stasi S, Van Rhijn B, Witjes AJ et al, 2015, Prognostic factors and risk groups in T1G3 non-muscle-invasive bladder cancer patients initially treated with Bacillus Calmette- Guerin: results of a retrospective multicenter study of 2451 patients. Eur Urol 67(1):74–82 Andius P, Johansson SL, Holmang S, 2007, Prognostic factors in stage T1 bladder cancer: tumor pattern, solid or papillary, and vascular invasion more important than depth of invasion. Urology 70(4):758–762 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1823 EP 1831 DI 10.1007/s12253-019-00767-1 PG 9 ER PT J AU Gao, J Ma, PX Deng, ShF Jiang, L Jia, DW Li, M AF Gao, Jian Ma, Peng Xiao Deng, Sheng Fu Jiang, Lin Jia, Dong Wei Li, Ming TI Associations of the BRAF V600E Mutation and PAQR3 Protein Expression with Papillary Thyroid Carcinoma Clinicopathological Features SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BRAF gene; PAQR3; Thyroid cancer; Papillary thyroid microcarcinoma; Lymph node metastasis (LNM); Extrathyroidal extension ID BRAF gene; PAQR3; Thyroid cancer; Papillary thyroid microcarcinoma; Lymph node metastasis (LNM); Extrathyroidal extension AB The BRAFV600E mutation is the most prevalent genetic event in patients with papillary thyroid cancer (PTC). However, no study has investigated the expression of PAQR3 in papillary thyroid tissues in relation to the BRAFV600E mutation and the clinicopathological features of PTC patients. Furthermore, the potential associations of the BRAFV600E mutation, PAQR3 expression and clinicopathological parameters in the cancerous tissues of PTC patients have not been investigated. This study was conducted on 60 patients with PTC who were treated surgically at our institution from 2017 to 2018. PCR was used to amplify DNA by the amplification refractorymutation system (ARMS) method to detect BRAFV600E gene mutations. In addition, immunohistochemical techniques were utilized to assess PAQR3 expression in tumor tissue sections. The BRAFV600E mutation was associated with lymph node metastasis (LNM, p < 0.05) but not with other clinicopathological features. Low PAQR3 expression was associated with extrathyroidal extension and LNM (χ2 = 7.143, p = 0.009; χ2 = 6.459, p = 0.014, respectively). Furthermore, a statistically significant association was observed between chronic lymphocytic thyroiditis and LNM (χ2 = 5.275, p = 0.0250). A linear relationship between the BRAFV600E mutation and PAQR3 protein expression has not been identified. These factors may be independent risk factors of extrathyroidal extension and LNM in PTC and be used to indicate the invasiveness of PTC tumors. Higher quality, multivariate analyses based on larger samples from around the world are urgently needed to further validate and revise our findings in the future. C1 [Gao, Jian] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Division of Thyroid and Breast Surgery, Department of General Surgery, 230036 Hefei, Anhui, China. [Ma, Peng Xiao] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Division of Thyroid and Breast Surgery, Department of General Surgery, 230036 Hefei, Anhui, China. [Deng, Sheng Fu] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Division of Thyroid and Breast Surgery, Department of General Surgery, 230036 Hefei, Anhui, China. [Jiang, Lin] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Division of Thyroid and Breast Surgery, Department of General Surgery, 230036 Hefei, Anhui, China. [Jia, Dong Wei] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of Hepatic Surgery, 230001 Hefei, Anhui, China. [Li, Ming] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of Pathology, 230001 Hefei, Anhui, China. RP Gao, J (reprint author), University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Division of Thyroid and Breast Surgery, Department of General Surgery, 230036 Hefei, China. EM gaojianahslyy@sina.com CR Cvejic D, Selemetjev S, Savin S, Paunovic I, Tatic S, 2009, Changes in the balance between proliferation and apoptosis during the progression of malignancy in thyroid tumours. Eur J Histochem 53:65–71 Kimura ET, Nikiforova MN, Zhu Z, Knauf JA, Nikiforov YE, Fagin JA, 2003, High prevalence of braf mutations in thyroid cancer genetic evidence for constitutive activation of the ret/ptc-rasbraf signaling pathway in papillary thyroid carcinoma. Cancer Res 63:1454–1457 Gu LQ, Li FY, Zhao L, Liu Y, Zhao YJ, 2009, Braf v600e mutation and x-linked inhibitor of apoptosis expression in papillary thyroid carcinoma. Thyroid 19:347–354 Mathur A, Moses W, Rahbari R, Khanafshar E, Duh Q, Clark O et al, 2011, Higher rate of braf mutation in papillary thyroid cancer over time. Cancer 117:4390–4395 Xing M, 2005, Braf mutation in thyroid cancer. Endocr Relat Cancer 12:245–262 Whipple CA, Boni A, Fisher JL, Hampton TH, ErnstoffMS, 2016, The mitogen-activated protein kinase pathway plays a critical role in regulating immunological properties of BRAF mutant cutaneous melanoma cells. Melanoma Res 26:223–235 Ji H, Wang Z, Perera SA et al, 2007, Mutations in BRAF and KRAS converge on activation of the mitogen-activated protein kinase pathway in lung cancer mouse models. Cancer Res 67:4933– 4939 Tang YT, Hu T, ArterburnM, Boyle B, Bright JM, Emtage PC et al, 2005, PAQR proteins: a novel membrane receptor family defined by an ancient7-transmembrane pass motif. J Mol Evol 61:372–380 Feng L, Xie X, Ding Q, Luo X, He J, Fan F et al, 2007, Spatial regulation of Raf kinase signaling by RKTG. Proc Natl Acad Sci 104:14348–14353 Fan F, Feng L, He J,Wang X, Jiang X, Zhang Yet al, 2008, RKTG sequesters B-Raf to the Golgi apparatus and inhibits the proliferation and tumorigenicity of human malignant melanoma cells. Carcinogenesis 29:1157–1163 Bai G, Chu J, EliM, Bao Y,Wen H, 2017, PAQR3 overexpression suppresses the aggressive phenotype of esophageal squamous cell carcinoma cells via inhibition of ERK signaling. Biomed Pharmacother 94:813–819 Ling ZQ, GuoW, Lu XX, Zhu X, Hong LL,Wang Z et al, 2014, A golgi-specific protein paqr3 is closely associated with the progression, metastasis and prognosis of human gastric cancers. Ann Oncol 25:1363–1372 Ma Z, Wang Y, Piao T, Li Z, Zhang H, Liu Z, Liu J, 2015, The tumor suppressor role of PAQR3 in osteosarcoma. Tumor Biol 36: 3319–3324 Chen J, Wang F, Xu J, He Z, Lu Y, Wang Z, 2016, The role of PAQR3 gene promoter hypermethylation in breast cancer and prognosis. Oncol Rep 36:1612–1618 Wu H, Zhang W, Ding Q, Peng G, Zou Z, Liu T et al, 2014, Identification of PAQR3 as a new candidate tumor suppressor in hepatocellular carcinoma. Oncol Rep 32:2687–2695 Bai G, Yang M, Zheng C, Zhang L, Eli M, 2018, Suppressor PAQR3 associated with the clinical significance and prognosis in esophageal squamous cell carcinoma. Oncol Lett 15:5703–5711 Sun J, Zhang J, Lu J, Gao J, Liang Z, 2016, BRAF V600E and TERT promoter mutations in papillary thyroid carcinoma in Chinese patients. PLoS One 11:e0153319 Guo L, Ma YQ, Yao Y et al, 2019, Role of ultrasonographic features and quantified BRAFV600E mutation in lymph node metastasis in Chinese patients with papillary thyroid carcinoma. Sci Rep 9:75 Li Y, Nakamura M, Kakudo K, 2009, Targeting of the BRAF gene in papillary thyroid carcinoma, review). Oncol Rep 22:671–681 Nasirden A, Saito T, Fukumura Y, Hara K, Akaike K, Kurisaki- Arakawa A et al, 2016, In Japanese patients with papillary thyroid carcinoma, TERTpromoter mutation is associated with poor prognosis, in contrast toBRAFV600E mutation. Virchows Arch 469: 687–696 Agnieszka C, Monika K, Dagmara R, Jolanta K, Michal J, Ewa S et al, 2015, The risk of relapse in papillary thyroid Cancer, PTC, in the context of BRAFV600E mutation status and other prognostic factors. PLOS ONE 10:e0132821 Choi SY, Park HS, Kang MK, Lee DK, Lee KD, Lee HS et al, 2013, The relationship between the BRAFV600E mutation in papillary thyroid microcarcinoma and clinicopathologic factors.World J Surg Oncol 11:1–5 Liu Z, Lv T, Xie C, Di Z, 2018, BRAFV600E gene mutation is associated with bilateral malignancy of papillary thyroid Cancer. Am J Med Sci 356:130–134 Qu HJ, Qu XY, Hu Z, Lin Y, Wang JR, Zheng CF et al, 2017, The synergic effect of BRAFV600E mutation and multifocality on central lymph node metastasis in unilateral papillary thyroid carcinoma. Endocr J 65:113–120 Chakraborty A, Narkar A, Mukhopadhyaya R, Kane S, D’Cruz A, Rajan MGR, 2012, BRAF V600E mutation in papillary thyroid carcinoma: significant association with node metastases and extra thyroidal invasion. Endocr Pathol 23:83–93 Kang SH, Pyo JY,Yang SWet al, 2013, Detection of BRAFV600E mutation with thyroid tissue using pyrosequencing. Am J Clin Pathol 139:759–764 Ahn D, Park JS, Sohn JH, Kim JH, Park SK, Seo AN et al, 2012, BRAF, V600E, mutation does not serve as a prognostic factor in Korean patients with papillary thyroid carcinoma. Auris Nasus Larynx 39:198–203 Kurt B, Yalcin S, Alagoz, Engin et al, 2012, The relationship of the BRAF, V600E, mutation and the established prognostic factors in papillary thyroid carcinomas. Endocr Pathol 23:135–140 Givens DJ, Buchmann LO, Agarwal AM, Grimmer JF, Hunt JP, 2014, BRAF V600E does not predict aggressive features of pediatric papillary thyroid carcinoma. Laryngoscope 124(9):E389– E393 Kim SJ, Lee KE, Myong JP, Park JH, Youn YK, 2011, BRAF V600E mutation is associated with tumor aggres- siveness in papillary thyroid cancer. World J Surg 36:310–317 Song JY, Sun SR, Dong F, Huang T, Wu B, Zhou J, 2018, Predictive value of BRAF V600E mutation for lymph node metastasis in papillary thyroid Cancer: a meta-analysis. CurrMed Sci 38: 785–797 Kurtulmus N, Ertas B, Saglican Y, Kaya H, DurenM(2016, BRAF V600E mutation: has it a role in cervical lymph node metastasis of papillary thyroid Cancer? Eur Thyroid J 5:195–200 Tang KT, Lee CH, 2010, BRAF mutation in papillary thyroid carcinoma: pathogenic role and clinical implications. J Chin Med Assoc 73:113–128 Li RH, Zhang AM, Li S, Li TY, Wang LJ, Zhang HR et al, 2016, PAQR3 gene expression and its methylation level in colorectal cancer tissues. Oncol Lett 12:1773–1778 Zhao C, Li Y, Chen G et al, 2017, Overexpression of miR-15b-5p promotes gastric cancer metastasis by regulating PAQR3. Oncol Rep 38:352–358 Guo W, You X, Xu D, Zhang Y, Wang Z, Man K et al, 2016, PAQR3 enhances Twist1 degradation to suppress epithelialmesenchymal transition and metastasis of gastric cancer cells. Carcinogenesis 37:397–407 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1833 EP 1841 DI 10.1007/s12253-019-00779-x PG 9 ER PT J AU Oka, S Ono, K Nohgawa, M AF Oka, Satoko Ono, Kazuo Nohgawa, Masaharu TI Clinical Effect of CD25 on the Prognosis of Diffuse Large B Cell Lymphoma with Secondary Central Nervous System Relapse SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Diffuse large B cell lymphoma; Central nervous system relapse; CD25; Rituximab ID Diffuse large B cell lymphoma; Central nervous system relapse; CD25; Rituximab AB In the present study, we investigated the effects of immunophenotyping on prognosis of diffuse large B cell lymphoma (DLBCL) with central nervous system (CNS) relapse treated with rituximab-CHOP (R-CHOP). CNS relapse occurred in 9.5% of DLBCL patients. At the diagnosis of DLBCL, CD25 was detected in 14.3% of cases. CD25 positivity correlated with an advanced stage, higher R-IPI, higher CNS-IPI, the presence of B symptoms, the presence of extranodal involvement >1, and bone involvement. Moreover CNS relapse was more frequently observed in patients with CD25+ than in those with CD25-. The univariate analysis showed that an advanced stage, high-risk R-IPI, high-risk CNS-IPI, bone involvement, and CD25+ were associated with shorter overall survival (OS). The multivariate analysis confirmed that CD25+ and high-risk CNS-IPI were independent adverse prognostic factors for shorter OS. A Kaplan-Meier analysis revealed the potential of CD25+ as a prognostic factor in patients with CNS relapse and that it correlated with shorter survival. The present results showed that the expression of CD25 in DLBCL patients with CNS relapse was associated with the patient prognosis independent other prognostic factors. The establishment of a treatment strategy for CNS relapse patients with CD25+ DLBCL cells is needed to improve poor outcomes. C1 [Oka, Satoko] Japanese Red Cross Society Wakayama Medical Center, Division of HematologyWakayama, Japan. [Ono, Kazuo] Japanese Red Cross Society Wakayama Medical Center, Division of PathologyWakayama, Japan. [Nohgawa, Masaharu] Japanese Red Cross Society Wakayama Medical Center, Division of HematologyWakayama, Japan. RP Oka, S (reprint author), Japanese Red Cross Society Wakayama Medical Center, Division of Hematology, Wakayama, Japan. EM okas@jasmine.ocn.ne.jp CR Boehme V, Zeynalova S, Kloess M, Loeffler M, Kaiser U, Pfreundschuh M, Schmitz N, German High-Grade Non- Hodgkin's Lymphoma Study Group, DSHNHL,, 2007, Incidence and risk factors of central nervous system recurrence in aggressive lymphoma–a survey of 1693 patients treated in protocols of the German high-grade non-Hodgkin's lymphoma study group, DSHNHL). Ann Oncol 18:149–157 Tomita N, Yokoyama M, Yamamoto W, Watanabe R, Shimazu Y, Masaki Y, Tsunoda S, Hashimoto C, Murayama K, Yano T, Okamoto R, Kikuchi A, Tamura K, Sato K, Sunami K, Shibayama H, Takimoto R, Ohshima R, Hatta Y, Moriuchi Y, Kinoshita T, Yamamoto M, Numata A, Ishigatsubo Y, Takeuchi K, 2012, Central nervous system event in patients with diffuse large B-cell lymphoma in the rituximab era. Cancer Sci 103:245–251 Aviles A, Jesus Nambo M, Neri N, 2013, Central nervous system prophylaxis in patients with aggressive diffuse large B cell lymphoma: an analysis of 3,258 patients in a single center. Med Oncol 30: 520 Chihara D, Oki Y, Matsuo K, Onoda H, Taji H, Yamamoto K, Morishima Y, 2011, Incidence and risk factors for central nervous system relapse in patients with diffuse large B-cell lymphoma: analyses with competing risk regression model. Leuk Lymphoma 52:2270–2275 Tai WM, Chung J, Tang PL, Koo YX, Hou X, Tay KW, Quek R, Tao M, Lim ST, 2011, Central nervous system, CNS, relapse in diffuse large B cell lymphoma, DLBCL): pre- and post-rituximab. Ann Hematol 90:809–818 Schmitz N, Zeynalova S, Nickelsen M, Kansara R, Villa D, Sehn LH, Glass B, Scott DW, Gascoyne RD, Connors JM, Ziepert M, Pfreundschuh M, Loeffler M, Savage KJ, 2016, CNS international prognostic index: a risk model for CNS relapse in patients with diffuse large B-cell lymphoma treated with R-CHOP. J Clin Oncol 34:3150–3156 Chin CK, Cheah CY, 2017, How I treat patients with aggressive lymphoma at high risk of CNS relapse. Blood. 130:867–874 HuW,Wang X, Yang R, Bi L, Xie Y, Zhang Z, Lu H,Wu L, 2017, Expression of CD56 is a risk factor for acute lymphocytic leukemia with central nervous system involvement in adults. Hematology. 22:81–87 Alinari L, Gru A, Quinion C, Huang Y, Lozanski A, Lozanski G, Poston J, Venkataraman G, Oak E, Kreisel F, Park SI, Matthews S, Abramson JS, Iris Lim H, Martin P, Cohen JB, Evens A, al- Mansour Z, Singavi A, Fenske TS, Blum KA, 2016, De novo CD5+ diffuse large B-cell lymphoma: adverse outcomes with and without stem cell transplantation in a large, multicenter, rituximab treated cohort. Am J Hematol 91:395–399 Flynn MJ, Hartley JA, 2017, The emerging role of anti-CD25 directed therapies as both immune modulators and targeted agents in cancer. Br J Haematol 179:20–35 Nakase K, Kita K, Anazawa H, Hoshino K, Shirakawa S, Tanaka I, Tsudo M, 1994, Induction of interleukin-2 receptor alpha chain expression of immature acute myelocytic leukemia cells. Leuk Res 18:269–274 Nakase K, Kita K, Nasu K, Ueda T, Tanaka I, Shirakawa S, Tsudo M, 1994, Differential expression of interleukin-2 receptors, alpha and beta chain, in mature lymphoid neoplasms. Am J Hematol 46: 179–183 Tesch H, Gunther A, Abts H et al, 1993, Expression of interleukin- 2R alpha and interleukin-2R beta in Hodgkin's disease. Am J Pathol 142:1714–1720 Waldmann TA, 2007, Anti-Tac, daclizumab, Zenapax, in the treatment of leukemia, autoimmune diseases, and in the prevention of allograft rejection: a 25-year personal odyssey. J Clin Immunol 27: 1–18 Hashimoto Y, Yokohama A, Saitoh A, Nakahashi H, Toyama K, Mitsui T, Koiso H, Saitoh T, Handa H, Uchiumi H, Jinbo T, Murayama K, Matsumoto M, Sawamura M, Karasawa M, Murakami H, Hirato J, Nojima Y, Kojima M, Tsukamoto N, 2013, Prognostic importance of the soluble form of IL-2 receptorα, sIL-2Rα, and its relationship with surface expression of IL-2Rα, CD25, of lymphoma cells in diffuse large B-cell lymphoma treated with CHOP-like regimen with or without rituximab: a retrospective analysis of 338 cases. J Clin Exp Hematop 53(3): 197–205 Yoshida N, Oda M, Kuroda Yet al, 2013, Clinical significance of sIL-2R levels in B-cell lymphomas. PLoS One 8:e78730 Geng H, Brennan S, Milne TA et al, 2012, Integrative epigenomic analysis identifies biomarkers and therapeutic targets in adult Bacute lymphoblastic leukemia. Cancer Discov 2:1004–1023 Lee JW, Chen Z, Geng H et al, 2015, CD25(IL2RA, orchestrates negative feedback control and stabilizes oncogenic signaling strength in acute lymphoblastic leukemia. Blood 126:1434 Fujiwara S, Muroi K, Hirata Y, Sato K, Matsuyama T, Ohmine K, Suzuki T, Ozaki K, Mori M, Nagai T, Tanaka A, Ozawa K, 2013, Clinical features of de novo CD25(+, diffuse large B-cell lymphoma. Hematology. 18:14–19 Fujiwara S,Muroi K, Tatara R, Matsuyama T, Ohmine K, Suzuki T, Mori M, Nagai T, Tanaka A, Ozawa K, 2014, Clinical features of de novo CD25-positive follicular lymphoma. Leuk Lymphoma 55: 307–313 Ibrahim S, Keating M, Do KA et al, 2001, CD38 expression as an important prognostic factor in B-cell chronic lymphocytic leukemia. Blood. 98:181–186 Tanimoto T, Kusumi E, Hosoda K et al, 2012, CNS prophylaxis in diffuse large B-cell lymphoma. Lancet. 379:1485–1486 Oki Y, Noorani M, Lin P et al, 2014, Double hit lymphoma: the MD Anderson Cancer Center clinical experience. Br J Haematol 166:891–901 Petrich AM, Gandhi M, Jovanovic B, Castillo JJ, Rajguru S, Yang DT, Shah KA, Whyman JD, Lansigan F, Hernandez-Ilizaliturri FJ, Lee LX, Barta SK, Melinamani S, Karmali R, Adeimy C, Smith S, Dalal N, Nabhan C, Peace D, Vose J, Evens AM, Shah N, Fenske TS, Zelenetz AD, Landsburg DJ, Howlett C, Mato A, Jaglal M, Chavez JC, Tsai JP, Reddy N, Li S, Handler C, Flowers CR, Cohen JB, Blum KA, Song K, Sun HL, Press O, Cassaday R, Jaso J, Medeiros LJ, Sohani AR, Abramson JS, 2014, Impact of induction regimen and stem cell transplantation on outcomes in double-hit lymphoma: a multicenter retrospective analysis. Blood. 124:2354–2361 Feugier P, Virion JM, Tilly H, Haioun C, Marit G, Macro M, Bordessoule D, Recher C, Blanc M, Molina T, Lederlin P, Coiffier B, 2004, Incidence and risk factors for central nervous system occurrence in elderly patients with diffuse large-B-cell lymphoma: influence of rituximab. Ann Oncol 15:129–133 Villa D, Connors JM, Shenkier TN, Gascoyne RD, Sehn LH, Savage KJ, 2010, Incidence and risk factors for central nervous system relapse in patients with diffuse large B-cell lymphoma: the impact of the addition of rituximab to CHOP chemotherapy. Ann Oncol 21:1046–1052 YamamotoW, Tomita N,Watanabe R, Hattori Y, Nakajima Y, Hyo R, Hashimoto C, Motomura S, Ishigatsubo Y, 2010, Central nervous system involvement in diffuse large B-cell lymphoma. Eur J Haematol 85:6–10 Shahab N, Doll DC, 1999, Testicular lymphoma. Semin Oncol 26: 259–269 Zucca E, Conconi A,Mughal TI, Sarris AH, Seymour JF, Vitolo U, Klasa R, Ozsahin M, Mead GM, Gianni MA, Cortelazzo S, Ferreri AJ, Ambrosetti A,MartelliM, Thieblemont C, Moreno HG, Pinotti G, MartinelliG,Mozzana R,Grisanti S, ProvencioM, Balzarotti M, Laveder F, Oltean G, Callea V, Roy P, Cavalli F, Gospodarowicz MK, International Extranodal Lymphoma Study Group, 2003, Patterns of outcome and prognostic factors in primary large-cell lymphoma of the testis in a survey by the international Extranodal lymphoma study group. J Clin Oncol 21:20–27 Chua SL, Seymour JF, Streater J, Wolf MM, Januszewicz EH, Prince HM, 2002, Intrathecal chemotherapy alone is inadequate central nervous system prophylaxis in patients with intermediategrade non-Hodgkin's lymphoma. Leuk Lymphoma 43:1783–1788 Kumar A, Vanderplas A, LaCasce AS, RodriguezMA, Crosby AL, Lepisto E, Czuczman MS, Nademanee A, Niland J, Gordon LI, Millenson M, Zelenetz AD, Friedberg JW, Abel GA, 2012, Lack of benefit of central nervous system prophylaxis for diffuse large Bcell lymphoma in the rituximab era: findings from a large national database. Cancer. 118:2944–2951 Phillips EH, Kirkwood AA, Lawrie A et al, 2016, Low rates of CNS relapse in high risk DLBCL patients treated with R-CODOXM and R-IVAC: results from a phase 2UK NCRI/Bloodwise trial. Blood 128:1855 Grommes C, Tang SS, Wolfe J, Kaley TJ, Daras M, Pentsova EI, Piotrowski AF, Stone J, Lin A, Nolan CP, Manne M, Codega P, Campos C, Viale A, Thomas AA, Berger MF, Hatzoglou V, Reiner AS, Panageas KS, DeAngelis L, Mellinghoff IK, 2019, Phase 1b trial of an ibrutinib-based combination therapy in recurrent/ refractory CNS lymphoma. Blood. 133:436–445 Rubenstein JL, Geng H, Fraser EJ, Formaker P, Chen L, Sharma J, Killea P, Choi K, Ventura J, Kurhanewicz J, Lowell C, Hwang J, Treseler P, Sneed PK, Li J, Wang X, Chen N, Gangoiti J, Munster PN, Damato B, 2018, Phase 1 investigation of lenalidomide/ rituximab plus outcomes of lenalidomide maintenance in relapsed CNS lymphoma. Blood Adv 2:1595–1607 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1843 EP 1850 DI 10.1007/s12253-019-00778-y PG 8 ER PT J AU Gao, Y Yang, M Wei, L Liang, X Wu, F Huang, Y Yang, T AF Gao, Ying Yang, Ming Wei, Liuliu Liang, Xiaofang Wu, Fang Huang, Yalan Yang, Tao TI miR-34a-5p Inhibits Cell Proliferation, Migration and Invasion Through Targeting JAG1/Notch1 Pathway in HPV-Infected Human Epidermal Keratinocytes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE miR-34a-5p; Jagged 1 (JAG1); Notch1 pathway; Human papillomavirus (HPV); Condyloma acuminate (CA) ID miR-34a-5p; Jagged 1 (JAG1); Notch1 pathway; Human papillomavirus (HPV); Condyloma acuminate (CA) AB Condyloma acuminate (CA) is a communicable disease caused by human papillomavirus (HPV). This study aimed to study the targeting relationship between miR-34a-5p and Jagged 1 (JAG1), as well as its regulatory effect in HPV-infected cells. Human keratinocyte HaCaT cells were infected with HPV16E6, and CA tissues were collected. The expression level of miR-34a-5p and JAG1 were detected in CA tissues and HPV-HaCaT cells. Cell proliferation, migration and invasion were respectively measured using 3-(4, 5)-dimethylthiahiazo-(−z-y1)-3, 5-diphenytetrazoliumromide (MTT), cell wound healing and Transwell assay. The potential binding sites of miR-34a-5p and JAG1 were predicted by website TargetScan, and confirmed using dual luciferase reporter gene assay. The proteins of Notch1 pathway-related were assessed using western blotting. The results showed that miR- 34a-5p expression was decreased, and JAG1 expression was increased in CA tissues and HPV-HaCaT cells. Cell proliferation, migration and invasion were decreased when miR-34a-5p over-expression and JAG1 knock-down in HPV-HaCaT cells. Furthermore, miR-34a-5p had a targeting effect on JAG1. The expression level of Notch1, NICD, Hes1 and Hey1 were increased when miR-34a-5p knock-down.miR-34a-5p could inhibit cell development, and regulate the activity of Notch1 pathway through targeting JAG1 expression in HPV-infected keratinocytes. C1 [Gao, Ying] Huazhong University of Science and Technology, Tongji Medical College, The Central hospital of Wuhan, Department of Dermatology, 430014 Wuhan, Hubei Province, China. [Yang, Ming] Huazhong University of Science and Technology, Tongji Medical College, The Central hospital of Wuhan, Department of Dermatology, 430014 Wuhan, Hubei Province, China. [Wei, Liuliu] Ganzhou People’s Hospital, Department of Gastroenterology, 341000 Ganzhou, Jiangxi Province, China. [Liang, Xiaofang] Huazhong University of Science and Technology, Tongji Medical College, The Central hospital of Wuhan, Department of Dermatology, 430014 Wuhan, Hubei Province, China. [Wu, Fang] Huazhong University of Science and Technology, Tongji Medical College, The Central hospital of Wuhan, Department of Dermatology, 430014 Wuhan, Hubei Province, China. [Huang, Yalan] Huazhong University of Science and Technology, Tongji Medical College, The Central hospital of Wuhan, Department of Dermatology, 430014 Wuhan, Hubei Province, China. [Yang, Tao] the First Affiliated Hospital of Gannan Medical University, Department of Dermatology, 341000 Ganzhou, Jiangxi Province, China. RP Yang, T (reprint author), the First Affiliated Hospital of Gannan Medical University, Department of Dermatology, 341000 Ganzhou, China. EM TaoYangset@163.com CR DinleyiciM, SaracogluN, ErenM,Kilic O, Ciftci E, Dinleyici EC, Sag C, Kara A, 2015, Giant Condyloma acuminate due to human papillomavirus type 16 in an infant successfully treated with topical Imiquimod therapy. Dermatol Rep 7(3). , DOI 10.4081/dr. 2015.6134 Mate JL, Ariza A, Roca X, Lopez D, Ferrandiz C, Perez-Piteira J, Navas-Palacios JJ, 1998, Expression patterns of cyclins D1 and E in condyloma acuminatum in comparison with psoriatic proliferative lesions. J Pathol 184(1):83–88. , DOI 10.1002/(sici, 1096-9896(199801)184:1<83::aid-path963>3.0.co;2-b Bu ZY, Yu XH, Wu LM, Zhong JB, Yang P, Chen J, 2017, Normalization of regulatory T cells, serum TGF-beta1, and LTN after 5-aminolevulinic acid-photodynamic therapy in patients with condyloma acuminate. Exp Ther Med 13(6):3327–3332. https:// doi.org/10.3892/etm.2017.4418 Du J, Cheng Q, Zhang Z,Wu JF, Li F, Chen SY,Wang YL, Lu XN, Xu JH, 2017, 5-Aminolevulinic acid photodynamic therapy stimulates local immunity in patients with condylomata acuminata via activation of T lymphocytes. Eur Rev Med Pharmacol Sci 21(5): 1125–1135 Barnes DM, Dublin EA, Fisher CJ, Levison DA, Millis RR, 1993, Immunohistochemical detection of p53 protein in mammary carcinoma: an important new independent indicator of prognosis? Hum Pathol 24(5):469–476 Kontomanolis EN, Koukouli Z, Liberis A, Stanulov G, Achouhan H, 2016, MiRNAs: regulators of human disease. Eur J Gynaecol Oncol 37(6):759–765 Dafa-Berger A, Kuzmina A, Fassler M, Yitzhak-Asraf H, Shemer- Avni Y, Taube R, 2012, Modulation of hepatitis C virus release by the interferon-induced protein BST-2/tetherin. Virology 428(2):98– 111. , DOI 10.1016/j.virol.2012.03.011 Nair V, Zavolan M, 2006, Virus-encoded microRNAs: novel regulators of gene expression. Trends Microbiol 14(4):169–175. , DOI 10.1016/j.tim.2006.02.007 Bertolus C, Goudot P, Gessain A, Berthet N, 2012, Clinical relevance of systematic human papillomavirus, HPV, diagnosis in oral squamous cell carcinoma. Infect Agents Cancer 7(1):13. https://doi. org/10.1186/1750-9378-7-13 Vokes EE, Agrawal N, Seiwert TY, 2015, HPV-associated head and neck cancer. J Natl Cancer Inst 107(12):djv344. https://doi. org/10.1093/jnci/djv344 Lajer CB, Garnaes E, Friis-Hansen L, Norrild B, Therkildsen MH, Glud M, Rossing M, Lajer H, Svane D, Skotte L, Specht L, Buchwald C, Nielsen FC, 2012, The role of miRNAs in human papilloma virus, HPV)-associated cancers: bridging between HPVrelated head and neck cancer and cervical cancer. Br J Cancer 106(9):1526–1534. , DOI 10.1038/bjc.2012.109 Wu W, Lin Z, Zhuang Z, Liang X, 2009, Expression profile of mammalian microRNAs in endometrioid adenocarcinoma. Eur J Cancer Prev 18(1):50–55. , DOI 10.1097/CEJ. 0b013e328305a07a McKenna DJ, Patel D, McCance DJ, 2014, miR-24 and miR-205 expression is dependent on HPV onco-protein expression in keratinocytes. Virology 448:210–216. , DOI 10.1016/j. virol.2013.10.014 Marthaler AM, Podgorska M, Feld P, Fingerle A, Knerr-Rupp K, Grasser F, Smola H, Roemer K, Ebert E, Kim YJ, Bohle RM, Muller CSL, Reichrath J, Vogt T, Malejczyk M,Majewski S, Smola S, 2017, Identification of C/EBPalpha as a novel target of the HPV8 E6 protein regulating miR-203 in human keratinocytes. PLoS Pathog 13(6): e1006406. , DOI 10.1371/journal.ppat.1006406 Wang X, Meyers C, Guo M, Zheng ZM, 2011, Upregulation of p18Ink4c expression by oncogenic HPVE6 via p53-miR-34a pathway. Int J Cancer 129(6):1362–1372. , DOI 10.1002/ijc.25800 Aqeilan RI, Calin GA, Croce CM(2010, miR-15a and miR-16-1 in cancer: discovery, function and future perspectives. Cell Death Differ 17(2):215–220. , DOI 10.1038/cdd.2009.69 Au Yeung CL, Tsang TY, Yau PL, Kwok TT, 2011, Human papillomavirus type 16 E6 induces cervical cancer cell migration through the p53/microRNA-23b/urokinase-type plasminogen activator pathway. Oncogene 30(21):2401–2410. , DOI 10. 1038/onc.2010.613 Pu Y, Zhao F, Li Y, Cui M, Wang H, Meng X, Cai S, 2017, The miR-34a-5p promotes the multi-chemoresistance of osteosarcoma via repression of the AGTR1 gene. BMC Cancer 17(1):45. https:// doi.org/10.1186/s12885-016-3002-x MacFarlane LA, Murphy PR, 2010, MicroRNA: biogenesis, function and role in Cancer. Curr Genom11(7):537–561. , DOI 10.2174/138920210793175895 Pang RT, Leung CO, Ye TM, Liu W, Chiu PC, Lam KK, Lee KF, Yeung WS, 2010, MicroRNA-34a suppresses invasion through downregulation of Notch1 and Jagged1 in cervical carcinoma and choriocarcinoma cells. Carcinogenesis 31(6):1037–1044. https:// doi.org/10.1093/carcin/bgq066 Bolha L, Dusanic D, Narat M, Oven I, 2012, Comparison of methods for relative quantification of gene expression using realtime PCR. Acta Agric Slovenica 100(2). , DOI 10.2478/ v10014-012-0018-z Darwish T, Freij M, Khadra M, Saleh H, 2016, Type specific prevalence of human papillomavirus among women with abnormal cytology in Jordan. Int J Gynecol Cancer 26:58–58 Ciotti M, Marzano V, Giuliani L, Nuccetelli M, D'Aguanno S, Azzimonti B, Bernardini S, Perno CF, Urbani A, Favalli C, Federici G, 2009, Proteomic investigation in A549 lung cell line stably infected by HPV16E6/E7 oncogenes. Respir Int Rev Thor Dis 77(4):427–439. , DOI 10.1159/000176209 WangX,WangHK,McCoy JP, Banerjee NS, Rader JS, Broker TR, Meyers C, Chow LT, Zheng ZM, 2009, Oncogenic HPV infection interrupts the expression of tumor-suppressive miR-34a through viral oncoprotein E6. RNA, New York, NY, 15(4):637–647. , DOI 10.1261/rna.1442309 Antonini D, RussoMT,De Rosa L,Gorrese M, DelVecchio L,Missero C, 2010, Transcriptional repression of miR-34 family contributes to p63-mediated cell cycle progression in epidermal cells. J Investig Dermatol 130(5):1249–1257. , DOI 10.1038/jid.2009.438 Kumar B, Yadav A, Lang J, Teknos TN, Kumar P, 2012, Dysregulation of microRNA-34a expression in head and neck squamous cell carcinoma promotes tumor growth and tumor angiogenesis. PLoS One 7(5):e37601. , DOI 10.1371/journal.pone.0037601 Ji X, Wang Z, Geamanu A, Goja A, Sarkar FH, Gupta SV, 2012, Delta-tocotrienol suppresses Notch-1 pathway by upregulating miR-34a in nonsmall cell lung cancer cells. Int J Cancer 131(11): 2668–2677. , DOI 10.1002/ijc.27549 Li B, Hu Y, Ye F, Li Y, Lv W, Xie X, 2010, Reduced miR-34a expression in normal cervical tissues and cervical lesions with highrisk human papillomavirus infection. Int J Gynecol Cancer 20(4): 597–604. , DOI 10.1111/IGC.0b013e3181d63170 Hashimi ST, Fulcher JA, ChangMH, Gov L,Wang S, Lee B, 2009, MicroRNA profiling identifies miR-34a and miR-21 and their target genes JAG1 andWNT1 in the coordinate regulation of dendritic cell differentiation. Blood 114(2):404–414. , DOI 10.1182/ blood-2008-09-179150 Reedijk M, Odorcic S, Chang L, Zhang H, Miller N, McCready DR, Lockwood G, Egan SE, 2005, High-level coexpression of JAG1 and NOTCH1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res 65(18):8530– 8537. , DOI 10.1158/0008-5472.can-05-1069 Simon DP, Giordano TJ, Hammer GD, 2012, Upregulated JAG1 enhances cell proliferation in adrenocortical carcinoma. Clin Cancer Res 18(9):2452–2464. , DOI 10.1158/1078-0432.ccr-11-2371 Ota T, Takekoshi S, Takagi T, Kitatani K, Toriumi K, Kojima T, Kato M, Ikoma N, Mabuchi T, Ozawa A, 2014, Notch signaling may be involved in the abnormal differentiation of epidermal keratinocytes in psoriasis. Acta Histochem Cytochem 47(4):175– 183. , DOI 10.1267/ahc.14027 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1851 EP 1859 DI 10.1007/s12253-019-00782-2 PG 9 ER PT J AU Kocsis, A Karsko, L Kurgyis, Zs Besenyi, Zs Pavics, L Dosa-Racz, Kis, E Baltas, E Ocsai, H Varga, E Bende, B Varga, A Mohos, G Korom, I Varga, J Kemeny, L Nemeth, BI Olah, J AF Kocsis, Adrienn Karsko, L Kurgyis, Zsuzsanna Besenyi, Zsuzsanna Pavics, Laszlo Dosa-Racz, Eva Kis, Eniko Baltas, Eszter Ocsai, Henriette Varga, Erika Bende, Balazs Varga, Attila Mohos, Gabor Korom, Irma Varga, Jozsef Kemeny, Lajos Nemeth, Balazs Istvan Olah, Judit TI Is it Necessary to Perform Sentinel Lymph Node Biopsy in Thin Melanoma? A Retrospective Single Center Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Melanoma; Sentinel lymph node biopsy; Mitotic rate; Regression; Stage ID Melanoma; Sentinel lymph node biopsy; Mitotic rate; Regression; Stage AB Sentinel lymph node biopsy (SLNB) is a standard procedure for regional lymph node staging and still has the most important prognostic value for the outcome of patients with thin melanoma. In addition to ulceration, SLNB had to be considered even for a single mitotic figure in thin (<1mm)melanoma according to AJCC7th guideline, therefore, a retrospective review was conducted involving 403 pT1 melanoma patients. Among them, 152 patients suffered from pT1b ulcerated or mitotic rate ≥ 1/ mm2 melanomas according to the AJCC7th staging system. SLNB was performed in 78 cases, of which nine (11.5%) showed SLN positivity. From them, interestingly, we found a relatively high positive sentinel rate (6/78–8%) in the case of thin primary melanomas ˂0.8 mm. Moreover, the presence of regression increased the probability of sentinel positivity by 5.796 fold. After reassessing pT stage based on the new AJCC8th, 37 pT1b cases were reordered into pT1a category. There was no significant relation between other characteristics examined (age, gender, Breslow, Clark level, and mitosis index) and sentinel node positivity. Based on our data, we suggest that mitotic rate alone is not a sufficiently powerful predictor of SLN status in thin melanomas. If strict histopathological definition criteria are applied, regression might be an additional adverse feature that aids in identifying T1 patients most likely to be SLN-positive. After reassessing of pT1b cases according to AJCC8th regression proved to be independent prognostic factor on sentinel lymph node positivity. Our results propose that sentinel lymph node biopsy might also be considered at patients with regressive thin (˂0.8 mm) melanomas. C1 [Kocsis, Adrienn] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Karsko, L] University of Szeged, Department of Nuclear MedicineSzeged, Hungary. [Kurgyis, Zsuzsanna] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Besenyi, Zsuzsanna] University of Szeged, Department of Nuclear MedicineSzeged, Hungary. [Pavics, Laszlo] University of Szeged, Department of Nuclear MedicineSzeged, Hungary. [Dosa-Racz, Eva] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Kis, Eniko] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Baltas, Eszter] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Ocsai, Henriette] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Varga, Erika] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Bende, Balazs] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Varga, Attila] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Mohos, Gabor] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Korom, Irma] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Varga, Jozsef] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Kemeny, Lajos] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Nemeth, Balazs Istvan] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Olah, Judit] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. RP Nemeth, BI (reprint author), University of Szeged, Department of Dermatology and Allergology, Szeged, Hungary. EM nemethistvanbalazs@gmail.com;nemeth.istvan.balazs@med.uszeged.hu CR Miller AJ, Mihm MC Jr, 2006, Melanoma. N Engl J Med 355:51– 65 Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63:11–30 Han D, Zager JS, Shyr Y, Chen H, Berry LD et al, 2013, Clinicopathologic predictors of sentinel lymph node metastasis in thin melanoma. J Clin Oncol 31:4387–4393 Balch CM, Gershenwald JE, Soong SJ, Thompson JF, Atkins MB et al, 2009, Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 27:6199–6206 Morton DL, Thompson JF, Cochran AJ, Mozzillo N, Nieweg OE et al, 2014, Final trial report of sentinel-node biopsy versus nodal observation in melanoma. N Engl J Med 370:599–609 Sladden M, Zagarella S, Popescu C, Bigby M, 2015, No survival benefit for patients with melanoma undergoing sentinel lymph node biopsy: critical appraisal of the multicenter selective lymphadenectomy trial-I final report. Br J Dermatol 172:566–571 Hieken TJ, Grotz TE, Comfere NI, Inselman JW, Habermann EB, 2015, The effect of the AJCC 7th edition change in T1 melanoma substaging on national utilization and outcomes of sentinel lymph node biopsy for thin melanoma. Melanoma Res 25:157–163 Hinz T, Ahmadzadehfar H, Wierzbicki A, Holler T, Wenzel J et al, 2012, Prognostic value of sentinel lymph node biopsy in 121 lowrisk melanomas, tumour thickness <1.00 mm, on the basis of a long-term follow-up. Eur J Nucl Med Mol Imaging 39:581–588 Mitteldorf C, Bertsch HP, Jung K, Thoms KM, Schon MP et al, 2014, Sentinel node biopsy improves prognostic stratification in patients with thin, pT1, melanomas and an additional risk factor. Ann Surg Oncol 21:2252–2258 Ranieri JM, Wagner JD, Wenck S, Johnson CS, Coleman JJ III, 2006, The prognostic importance of sentinel lymph node biopsy in thin melanoma. Ann Surg Oncol 13:927–932 Wright BE, Scheri RP, Ye X, Faries MB, Turner RR et al, 2008, Importance of sentinel lymph node biopsy in patients with thin melanoma. Arch Surg 143:892–899 Joyce KM,McInerney NM, Joyce CW, Jones DM, Hussey AJ et al, 2015, A review of sentinel lymph node biopsy for thin melanoma. Ir J Med Sci 184:119–123 Gershenwald JE, Scolyer RA, Hess KR, Sondak VK, LongGVet al, 2017, Melanoma staging: evidence-based changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J Clin 67:472–492 Morton DL, Cochran AJ, Thompson JF, Elashoff R, Essner R et al, 2005, Sentinel node biopsy for early-stage melanoma: accuracy and morbidity in MSLT-I, an international multicenter trial. Ann Surg 242:302–311 Murali R, Haydu LE, Quinn MJ, Saw RP, Shannon K et al, 2012, Sentinel lymph node biopsy in patients with thin primary cutaneous melanoma. Ann Surg 255:128–133 Sondak VK, Taylor JM, Sabel MS, Wang Y, Lowe L et al, 2004, Mitotic rate and younger age are predictors of sentinel lymph node positivity: lessons learned from the generation of a probabilistic model. Ann Surg Oncol 11:247–258 Bleicher RJ, Essner R, Foshag LJ, Wanek LA, Morton DL, 2003, Role of sentinel lymphadenectomy in thin invasive cutaneous melanomas. J Clin Oncol 21:1326–1331 Morris KT, Busam KJ, Bero S, Patel A, Brady MS, 2008, Primary cutaneous melanoma with regression does not require a lower threshold for sentinel lymph node biopsy. Ann Surg Oncol 15: 316–322 Kretschmer L, Starz H, Thoms KM, Satzger I, Volker B et al, 2011, Age as a key factor influencing metastasizing patterns and disease-specific survival after sentinel lymph node biopsy for cutaneous melanoma. Int J Cancer 129:1435–1442 Balch CM, Soong SJ,Gershenwald JE, Thompson JF, Coit DGet al, 2013, Age as a prognostic factor in patients with localized melanoma and regional metastases. Ann Surg Oncol 20:3961–3968 Han D, Yu D, Zhao X, Marzban SS, Messina JL et al, 2012, Sentinel node biopsy is indicated for thin melanomas >/=0.76 mm. Ann Surg Oncol 19:3335–3342 Bagaria SP, Ray PS, Joseph RW, Heckman MG, Rawal B et al, 2013, Ultrathin primary is a marker for worse prognosis in lymph node-positive cutaneous melanoma. Cancer 119:1860–1867 Cecchi R, Buralli L, Innocenti S, De Gaudio C, 2007, Sentinel lymph node biopsy in patients with thin melanomas. J Dermatol 34:512–515 Balch CM, Buzaid AC, Soong SJ, Atkins MB, Cascinelli N et al, 2001, Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 19:3635– 3648 Rousseau DL Jr, Ross MI, Johnson MM, Prieto VG, Lee JE et al, 2003, Revised American Joint Committee on Cancer staging criteria accurately predict sentinel lymph node positivity in clinically node-negative melanoma patients. Ann Surg Oncol 10:569–574 Yonick DV, Ballo RM, Kahn E, Dahiya M, Yao K et al, 2011, Predictors of positive sentinel lymph node in thin melanoma. Am J Surg 201:324–327 Oliveira Filho RS, Ferreira LM, Biasi LJ, EnokiharaMM, PaivaGR et al, 2003, Vertical growth phase and positive sentinel node in thin melanoma. Braz J Med Biol Res 36:347–350 Kesmodel SB, Karakousis GC, Botbyl JD, Canter RJ, Lewis RT et al, 2005, Mitotic rate as a predictor of sentinel lymph node positivity in patients with thin melanomas. Ann Surg Oncol 12: 449–458 Wong SL, Brady MS, Busam KJ, Coit DG, 2006, Results of sentinel lymph node biopsy in patients with thin melanoma. Ann Surg Oncol 13:302–309 Olah J, Gyulai R, Korom I, Varga E, Dobozy A, 2003, Tumour regression predicts higher risk of sentinel node involvement in thin cutaneous melanomas. Br J Dermatol 149:662–663 Markovic SN, Erickson LA, Rao RD,Weenig RH, Pockaj BA et al, 2007, Malignant melanoma in the 21st century, part 2: staging, prognosis, and treatment. Mayo Clin Proc 82:490–513 Abbas O, Miller DD, Bhawan J, 2014, Cutaneous malignant melanoma: update on diagnostic and prognostic biomarkers. Am J Dermatopathol 36:363–379 Cooper C, Wayne JD, Damstetter EM, Martini M, Gordon J et al, 2013, A 10-year, single-institution analysis of clinicopathologic features and sentinel lymph node biopsy in thin melanomas. J Am Acad Dermatol 69:693–699 Guitart J, Lowe L, Piepkorn M, Prieto VG, RabkinMS et al, 2002, Histological characteristics of metastasizing thin melanomas: a case-control study of 43 cases. Arch Dermatol 138:603–608 Blessing K, McLaren KM, McLean A, Davidson P, 1990, Thin malignant melanomas, less than 1.5 mm, with metastasis: a histological study and survival analysis. Histopathology 17:389–395 Brogelli L, Reali UM, Moretti S, Urso C, 1992, The prognostic significance of histologic regression in cutaneous melanoma. Melanoma Res 2:87–91 Botella-Estrada R, Traves V, Requena C,Guillen-Barona C,Nagore E, 2014, Correlation of histologic regression in primary melanoma with sentinel node status. JAMA Dermatol 150:828–835 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1861 EP 1868 DI 10.1007/s12253-019-00769-z PG 8 ER PT J AU Zhang, Sh He, J Tang, M Sun, H AF Zhang, Shouru He, Jingping Tang, Maocai Sun, Hao TI Prdx2 Upregulation Promotes the Growth and Survival of Gastric Cancer Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Peroxiredoxin2; Gastric cancer; Carcinogenesis ID Peroxiredoxin2; Gastric cancer; Carcinogenesis AB Peroxiredoxins (Prdxs) play important roles in cell proliferation, differentiation, and the mediation of intracellular signalling pathways. Prdx2 is an important member of the peroxiredoxin family and is upregulated in many cancers. Until now, the biological functions of Prdx2 in gastric cancer have not been completely understood, and the underlying mechanisms remain elusive. The aim of this study was to identify the role of Prdx2 on the growth of gastric cancer cells and the underlying mechanisms. We demonstrated that Prdx2 was highly expressed in gastric cancer tissues and cell lines and that the overexpression of Prdx2 correlated with the progression of gastric cancer. Further, Prdx2 was silenced with a specific, lentiviral vector-mediated shRNA, and this suppressed the proliferation of gastric cancer cells and promoted the apoptosis of gastric cancer cells. Finally, the knockdown of Prdx2 contributed to the attenuated gastric cancer growth in BALB/c nude mice. In conclusion, these findings demonstrate that Prdx2 may participate in the carcinogenesis and development of gastric cancer. C1 [Zhang, Shouru] Chongqing University, Cancer Hospital, Department of Gastrointestinal Surgery, 400030 Chongqing, China. [He, Jingping] Chongqing University, Cancer Hospital, Department of Gastrointestinal Surgery, 400030 Chongqing, China. [Tang, Maocai] Chongqing University, Cancer Hospital, Department of Gastrointestinal Surgery, 400030 Chongqing, China. [Sun, Hao] Chongqing University, Cancer Hospital, Department of Gastrointestinal Surgery, 400030 Chongqing, China. RP Sun, H (reprint author), Chongqing University, Cancer Hospital, Department of Gastrointestinal Surgery, 400030 Chongqing, China. EM sunhao68@sina.com CR Siegel R, Ma J, Zou Z, Jemal A, 2014, Cancer statistics, 2014 [J]. CA Cancer J Clin 64(1):9–29 Hu B, El Hajj N, Sittler S et al, 2012, Gastric cancer: classification, histology and application of molecular pathology [J]. J Gastrointest Oncol 3(3):251–261 Gotoda T, Kusano C, Moriyasu F, 2014, Future perspective of gastric cancer endotherapy [J]. Ann Transl Med 2(3):25 Roessner A, Kuester D, Malfertheiner P et al, 2008, Oxidative stress in ulcerative colitis-associated carcinogenesis [J]. Pathol Res Pract 204(7):511–524 Devasagayam TP, Tilak JC, Boloor KK et al, 2004, Free radicals and antioxidants in human health: current status and future prospects [J]. J Assoc Physicians India 52:794–804 Schieber M, Chandel NS, 2014, ROS function in redox signaling and oxidative stress [J]. Curr Biol 24(10):R453–r462 Poole LB, 2011, Overview of Peroxiredoxins in oxidant defense and redox regulation [J]. Chapter, Unit7.9) Memon AA, Chang JW, Oh BR, Yoo YJ, 2005, Identification of differentially expressed proteins during human urinary bladder cancer progression [J]. Cancer Detect Prev 29(3):249–255 Jarvela S, Rantala I, Rodriguez A et al, 2010, Specific expression profile and prognostic significance of peroxiredoxins in grade II-IV astrocytic brain tumors [J]. BMC Cancer 10:104 Karihtala P, Mantyniemi A, KangSWet al, 2003, Peroxiredoxins in breast carcinoma [J]. Clin Cancer Res 9(9):3418–3424 Wu XY, Fu ZX, Wang XH, 2010, Peroxiredoxins in colorectal neoplasms [J]. Histol Histopathol 25(10):1297–1303 Lee KW, Lee DJ, Lee JY, Kang DH, Kwon J, Kang SW, 2011, Peroxiredoxin II restrains DNA damage-induced death in cancer cells by positively regulating JNK-dependent DNA repair [J]. J Biol Chem 286(10):8394–8404 Park SH, Chung YM, Lee YS, Kim HJ, Kim JS, Chae HZ, Yoo YD, 2000, Antisense of human peroxiredoxin II enhances radiationinduced cell death [J]. Clin Cancer Res 6(12):4915–4920 Parmigiani RB, Xu WS, Venta-Perez G, Erdjument-Bromage H, Yaneva M, Tempst P, Marks PA, 2008, HDAC6 is a specific deacetylase of peroxiredoxins and is involved in redox regulation [J]. Proc Natl Acad Sci U S A 105(28):9633–9638 LuW, Fu Z,Wang H, Feng J,Wei J, Guo J, 2014, Peroxiredoxin 2 is upregulated in colorectal cancer and contributes to colorectal cancer cells' survival by protecting cells from oxidative stress [J]. Mol Cell Biochem 387(1–2):261–270 LuW, Fu Z,Wang H, Feng J,Wei J, Guo J, 2014, Peroxiredoxin 2 knockdown by RNA interference inhibits the growth of colorectal cancer cells by downregulating Wnt/beta-catenin signaling [J]. Cancer Lett 343(2):190–199 Wang S, Chen Z, Zhu S et al, 2019, PRDX2 protects against oxidative stress induced by H. pylori and promotes resistance to cisplatin in gastric cancer [J]. Redox Biol 28:101319 Yang SF, Ma R, Pan LL, Cao J, Sheng N, 2018, RKIP and peroxiredoxin 2 expression predicts the proliferative potential of gastric cancer stem cells [J]. Oncol Lett 15(3):3173–3177 Hanschmann EM, Godoy Je R, Berndt C et al, 2013, Thioredoxins, Glutaredoxins, and Peroxiredoxins—molecular mechanisms and health significance: from cofactors to antioxidants to redox signaling [J]. Antioxid Redox Signal 19(13):1539–1605 Lomnytska MI, Becker S, Bodin I, Olsson A, Hellman K, Hellstrom AC, Mints M, Hellman U, Auer G, Andersson S, 2011, Differential expression of ANXA6, HSP27, PRDX2, NCF2, and TPM4 during uterine cervix carcinogenesis: diagnostic and prognostic value [J]. Br J Cancer 104(1):110–119 Yue HY, Cao J, Cui JF, Dai Z, Su JJ, Duan XX, Yang C, Yue HF, Li Y, Liu YK, 2007, The expression of peroxiredoxin II in hepatocellular carcinoma and its significance [J]. Zhonghua Gan Zang Bing Za Zhi 15(5):366–369 Rhee SG, Woo HA, 2011, Multiple functions of peroxiredoxins: peroxidases, sensors and regulators of the intracellular messenger H(2)O(2), and protein chaperones [J]. Antioxid Redox Signal 15(3):781–794 Olahova M, Veal EA, 2015, A peroxiredoxin, PRDX-2, is required for insulin secretion and insulin/IIS-dependent regulation of stress resistance and longevity [J]. Aging Cell 14(4):558–568 Neumann CA, Fang Q, 2007, Are peroxiredoxins tumor suppressors? [J]. Curr Opin Pharmacol 7(4):375–380 Hirahashi M, Koga Y, Kumagai R, Aishima S, Taguchi K, Oda Y, 2014, Induced nitric oxide synthetase and peroxiredoxin expression in intramucosal poorly differentiated gastric cancer of young patients [J]. Pathol Int 64(4):155–163 Hu X,Weng Z, Chu CT, Zhang L, Cao G, Gao Y, Signore A, Zhu J, Hastings T, Greenamyre JT, Chen J, 2011, Peroxiredoxin-2 protects against 6-hydroxydopamine-induced dopaminergic neurodegeneration via attenuation of the ASK1 signaling cascade [J]. J Neurosci 31(1):247–261 Ishii T, Warabi E, Yanagawa T, 2012, Novel roles of peroxiredoxins in inflammation, cancer and innate immunity [J]. J Clin Biochem Nutr 50(2):91–105 Yang C, Fu Z, 2014, Liposomal delivery and polyethylene glycolliposomal oxaliplatin for the treatment of colorectal cancer, review, [J]. Biomed Rep 2(3):335–339 Walters J, Pop C, Scott FL et al, 2009, A constitutively active and uninhibitable caspase-3 zymogen efficiently induces apoptosis [J]. Biochem J 424(Pt 3):335–345 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1869 EP 1877 DI 10.1007/s12253-019-00783-1 PG 9 ER PT J AU Tripodi, APA Randelovic, I Biri-Kovacs, B Szeder, B Mezo, G Tovari, J AF Tripodi, Angelo Pierluigi Andrea Randelovic, Ivan Biri-Kovacs, Beata Szeder, Balint Mezo, Gabor Tovari, Jozsef TI In Vivo Tumor Growth Inhibition and Antiangiogenic Effect of Cyclic NGR Peptide-Daunorubicin Conjugates Developed for Targeted Drug Delivery SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Targeted tumor therapy; NGR peptides; Tumor growth inhibition; Antiangiogenic effect; CD13; Metastasis ID Targeted tumor therapy; NGR peptides; Tumor growth inhibition; Antiangiogenic effect; CD13; Metastasis AB Among various homing devices, peptides containing the NGR tripeptide sequence represent a promising approach to selectively recognize CD13 receptor isoforms on the surface of tumor cells. They have been successfully used for the delivery of various chemotherapeutic drugs to tumor vessels. Here, we report on the murine plasma stability, in vitro and in vivo antitumor activity of our recently described bioconjugates containing daunorubicin as payload. Furthermore, CD13 expression of KS Kaposi’s Sarcoma cell line and HT-29 human colon carcinoma cell line was investigated. Flow cytometry studies confirm the fast cellular uptake resulting in the rapid delivery of the active metabolite Dau = Aoa-Gly-OH to tumor cells. The increased in vitro antitumor effect might be explained by the faster rearrangement from NGR to isoDGR in case of conjugate 2 (Dau = Aoa- GFLGK(c[NleNGRE]-GG)-NH2) in comparison with conjugate 1 (Dau = Aoa-GFLGK(c[KNGRE]-GG)-NH2). Nevertheless, results indicated that both conjugates showed significant effect on inhibition of proliferation in the primary tumor and also on blood vessel formation making them a potential candidate for targeting angiogenesis processes in tumors where CD13 and integrins are involved. C1 [Tripodi, Angelo Pierluigi Andrea] Eotvos Lorand University, Hungarian Academy of Sciences, MTA-ELTE Research Group of Peptide ChemistryBudapest, Hungary. [Randelovic, Ivan] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Biri-Kovacs, Beata] Eotvos Lorand University, Hungarian Academy of Sciences, MTA-ELTE Research Group of Peptide ChemistryBudapest, Hungary. [Szeder, Balint] Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute of EnzymologyBudapest, Hungary. [Mezo, Gabor] Eotvos Lorand University, Hungarian Academy of Sciences, MTA-ELTE Research Group of Peptide ChemistryBudapest, Hungary. [Tovari, Jozsef] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. RP Tovari, J (reprint author), National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, Budapest, Hungary. EM tozsi@oncol.hu CR Corti A, Pastorino F, Curnis F et al, 2012, Targeted drug delivery and penetration into solid tumors. Med Res Rev 32(5):1078–1091 CortiA, Curnis F, 2011, Tumor vasculature targeting throughNGR peptide-based drug delivery systems. Curr Pharm Biotechnol 12(8): 1128–1134 Schuster S, Biri-Kovacs B, Szeder B et al, 2018, Synthesis and in vitro biochemical evaluation of oxime bond-linked daunorubicin-GnRH-III conjugates developed for targeted drug delivery. Beilstein J Org Chem 14:756–771 Thundimadathil J, 2012, Cancer treatment using peptides: current therapies and future prospects. J Amino Acids 2012:967347 Reff ME, Hariharan K, Braslawsky G, 2002, Future of monoclonal antibodies in the treatment of hematologic malignancies. Cancer Control J Moffitt Cancer Cent 9(2):152–166 Qiu X-Q, Wang H, Cai B, Wang L-L, Yue S-T, 2007, Small antibody mimetics comprising two complementarity-determining regions and a framework region for tumor targeting. Nat Biotechnol 25(8):921–929 Schuster S, Biri-Kovacs B, Szeder B et al, 2018, Enhanced in vitro antitumor activity of GnRH-III-Daunorubicin bioconjugates influenced by sequence modification. Pharmaceutics 10(4) Saiki I, Fujii H, Yoneda J et al, 1993, Role of aminopeptidase N, CD13, in tumor-cell invasion and extracellularmatrix degradation. Int J Cancer 54(1):137–143 Hynes RO, 2002, Integrins: bidirectional, allosteric signaling machines. Cell 110(6):673–687 Corti A, Curnis F, Arap W, Pasqualini R, 2008, The neovasculature homing motifNGR:more thanmeets the eye. Blood 112(7):2628–2635 Arap W, Pasqualini R, Ruoslahti E, 1998, Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model. Science 279(5349):377–380 Wang RE, Niu Y, Wu H, Amin MN, Cai J, 2011, Development of NGR peptide-based agents for tumor imaging.AmJ NuclMed Mol Imaging 1(1):36–46 Folkman J, 1995)Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med 1(1):27–31 Hanahan D, Folkman J, 1996, Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86(3):353–364 Kerbel R, Folkman J, 2002, Clinical translation of angiogenesis inhibitors. Nat Rev Cancer 2(10):727–739 Seidi K, Jahanban-Esfahlan R,Monhemi H et al, 2018, NGR, Asn- Gly-Arg)-targeted delivery of coagulase to tumor vasculature arrests cancer cell growth. Oncogene 37(29):3967–3980 Razak K, Newland AC, 1992, The significance of aminopeptidases and haematopoietic cell differentiation. Blood Rev 6(4):243–250 Luan Y, Xu W, 2007, The structure and main functions of aminopeptidase N. Curr Med Chem 14(6):639–647 RiemannD, KehlenA, Langner J, 1999, CD13–not just amarker in leukemia typing. Immunol Today 20(2):83–88 Dixon J, Kaklamanis L, Turley H et al, 1994, Expression of aminopeptidase-n, CD 13, in normal tissues and malignant neoplasms of epithelial and lymphoid origin. J Clin Pathol 47(1):43–47 Mina-Osorio P, 2008, The moonlighting enzyme CD13: old and new functions to target. Trends Mol Med 14(8):361–371 Bhagwat SV, Lahdenranta J, Giordano R et al, 2001, CD13/APN is activated by angiogenic signals and is essential for capillary tube formation. Blood 97(3):652–659 Menrad A, Speicher D, Wacker J, Herlyn M, 1993, Biochemical and functional characterization of aminopeptidase N expressed by human melanoma cells. Cancer Res 53(6):1450–1455 Rangel R, Sun Y, Guzman-Rojas L et al, 2007, Impaired angiogenesis in aminopeptidase N-null mice. Proc Natl Acad Sci U S A 104(11):4588–4593 Fukasawa K, Fujii H, Saitoh Y et al, 2006, Aminopeptidase N, APN/CD13, is selectively expressed in vascular endothelial cells and plays multiple roles in angiogenesis. Cancer Lett 243(1):135– 143 Hashida H, Takabayashi A, Kanai M et al, 2002, Aminopeptidase N is involved in cell motility and angiogenesis: its clinical significance in human colon cancer. Gastroenterology 122(2):376–386 Taylor A, 1993, Aminopeptidases: structure and function FASEB J Off Publ Fed Am Soc Exp Biol 7(2):290–298 Chen H, Kinzer CA, Paul WE, 1996, p161, a murine membrane protein expressed on mast cells and some macrophages, is mouse CD13/aminopeptidase N. J Immunol Baltim Md 157(6):2593– 2600 Drexler HG, 1987, Classification of acute myeloid leukemias–a comparison of FAB and immunophenotyping. Leukemia 1(10): 697–705 von Wallbrunn A, Waldeck J, Holtke C et al, 2008, In vivo optical imaging of CD13/APN-expression in tumor xenografts. J Biomed Opt 13(1):011007 Mate G, Kertesz I, Enyedi KN et al, 2015, In vivo imaging of aminopeptidase N, CD13, receptors in experimental renal tumors using the novel radiotracer, 68)Ga-NOTA-c(NGR). Eur J Pharm Sci Off J Eur Fed Pharm Sci 69:61–71 Yokoyama Y, Ramakrishnan S, 2005, Addition of an aminopeptidase N-binding sequence to human endostatin improves inhibition of ovarian carcinoma growth. Cancer 104(2):321–331 Buehler A, van Zandvoort MAMJ, Stelt BJ et al, 2006, cNGR: a novel homing sequence for CD13/APN targeted molecular imaging of murine cardiac angiogenesis in vivo. Arterioscler Thromb Vasc Biol 26(12):2681–2687 Zarovni N, Monaco L, Corti A, 2004, Inhibition of tumor growth by intramuscular injection of cDNA encoding tumor necrosis factor alpha coupled to NGR and RGD tumor-homing peptides. Hum Gene Ther 15(4):373–382 Bieker R, Kessler T, Schwoppe C et al, 2009, Infarction of tumor vessels by NGR-peptide-directed targeting of tissue factor: experimental results and first-in-man experience. Blood 113(20):5019– 5027 Oostendorp M, Douma K, Hackeng TM et al, 2008, Quantitative molecular magnetic resonance imaging of tumor angiogenesis using cNGR-labeled paramagnetic quantum dots. Cancer Res 68(18):7676–7683 Enyedi KN, Toth S, Szakacs G,Mezo G, 2017, NGR-peptide-drug conjugates with dual targeting properties. PLoS One 12(6): e0178632 Curnis F, Sacchi A, Borgna L et al, 2000, Enhancement of tumor necrosis factor alpha antitumor immunotherapeutic properties by targeted delivery to aminopeptidase N, CD13). Nat Biotechnol 18(11):1185–1190 Corti A, 2004, Strategies for improving the anti-neoplastic activity of TNF by tumor targeting. Methods Mol Med 98:247–264 Ellerby HM, Arap W, Ellerby LM et al, 1999, Anti-cancer activity of targeted pro-apoptotic peptides. Nat Med 5(9):1032–1038 Corti A, Curnis F, Rossoni G, Marcucci F, Gregorc V, 2013, Peptide-mediated targeting of cytokines to tumor vasculature: the NGR-hTNF example. BioDrugs Clin Immunother Biopharm Gene Ther 27(6):591–603 Pastorino F, Brignole C, Di Paolo D et al, 2006, Targeting liposomal chemotherapy via both tumor cell-specific and tumor vasculature-specific ligands potentiates therapeutic efficacy. Cancer Res 66(20):10073–10082 Mukhopadhyay S, Barnes CM, Haskel A et al, 2008, Conjugated platinum(IV)-peptide complexes for targeting angiogenic tumor vasculature. Bioconjug Chem 19(1):39–49 Ndinguri MW, Solipuram R, Gambrell RP, Aggarwal S, Hammer RP, 2009, Peptide targeting of platinum anti-cancer drugs. Bioconjug Chem 20(10):1869–1878 Zhang Z, Hatta H, Ito T, Nishimoto S, 2005, Synthesis and photochemical properties of photoactivated antitumor prodrugs releasing 5-fluorouracil. Org Biomol Chem 3(4):592–596 Kirikoshi R, Manabe N, Takahashi O, 2017, Succinimide formation from an NGR-containing cyclic peptide: computational evidence for catalytic roles of phosphate buffer and the arginine side chain. Int J Mol Sci 18(2) Violand BN, SchlittlerMR, Toren PC, Siegel NR, 1990, Formation of isoaspartate 99 in bovine and porcine somatotropins. J Protein Chem 9(1):109–117 Meinwald YC, Stimson ER, Scheraga HA, 1986, Deamidation of the asparaginyl-glycyl sequence. Int J Pept Protein Res 28(1):79–84 Geiger T, Clarke S, 1987, Deamidation, isomerization, and racemization at asparaginyl and aspartyl residues in peptides. Succinimide-linked reactions that contribute to protein degradation. J Biol Chem 262(2):785–794 Robinson NE, 2002, Protein deamidation. Proc Natl Acad Sci U S A 99(8):5283–5288 Negussie AH, Miller JL, Reddy G et al, 2010, Synthesis and in vitro evaluation of cyclic NGR peptide targeted thermally sensitive liposome. J Control Release Off J Control Release Soc 143(2): 265–273 Enyedi KN, Czajlik A,KnappK et al, 2015, Development of cyclic NGR peptides with thioether linkage: structure and dynamics determining deamidation and bioactivity. J Med Chem 58(4):1806– 1817 Curnis F, Cattaneo A, Longhi R et al, 2010, Critical role of flanking residues in NGR-to-isoDGR transition and CD13/integrin receptor switching. J Biol Chem 285(12):9114–9123 Spitaleri A, Mari S, Curnis F et al, 2008, Structural basis for the interaction of isoDGR with the RGD-binding site of alphavbeta3 integrin. J Biol Chem 283(28):19757–19768 MingozziM, Dal Corso A,MarchiniMet al, 2013, Cyclic isoDGR peptidomimetics as low-nanomolar αvβ3 integrin ligands. Chem Weinh Bergstr Ger 19(11):3563–3567 Frank AO, Otto E, Mas-Moruno C et al, 2010, Conformational control of integrin-subtype selectivity in isoDGR peptide motifs: a biological switch. Angew Chem Int Ed Engl 49(48):9278–9281 Tripodi AAP, Toth S, Enyedi KN et al, 2018, Development of novel cyclic NGR peptide-daunomycin conjugates with dual targeting property. Beilstein J Org Chem 14:911–918 Goodman SL, Grote HJ, Wilm C, 2012, Matched rabbit monoclonal antibodies against αv-series integrins reveal a novel αvβ3- LIBS epitope, and permit routine staining of archival paraffin samples of human tumors. Biol Open 1(4):329–340 Simonart T, Hermans P, Schandene L, Van Vooren JP, 2000, Phenotypic characteristics of Kaposi’s sarcoma tumour cells derived from patch-, plaque- and nodular-stage lesions: analysis of cell cultures isolated from AIDS and non-AIDS patients and review of the literature. Br J Dermatol 143(3):557–563 Ensoli B, Sgadari C, Barillari G et al, 2001, Biology of Kaposi’s sarcoma. Eur J Cancer Oxf Engl 37(10):1251–1269 Cancian L, Hansen A, Boshoff C, 2013, Cellular origin of Kaposi’s sarcoma and Kaposi’s sarcoma-associated herpesvirus-induced cell reprogramming. Trends Cell Biol 23(9):421–432 Albini A, Paglieri I, Orengo G et al, 1997, The beta-core fragment of human chorionic gonadotrophin inhibits growth of Kaposi’s sarcoma-derived cells and a new immortalized Kaposi’s sarcoma cell line. AIDS 11: 713-721. AIDS Lond Engl 11:713–721 Schreiner C, Bauer J, Margolis M, Juliano RL, 1991, Expression and role of integrins in adhesion of human colonic carcinoma cells to extracellular matrix components. Clin Exp Metastasis 9(2):163– 178 Garrigues HJ, Rubinchikova YE, Dipersio CM, Rose TM, 2008, Integrin alphaVbeta3 binds to the RGD motif of glycoprotein B of Kaposi’s sarcoma-associated herpesvirus and functions as an RGDdependent entry receptor. J Virol 82(3):1570–1580 Ensoli B, Sturzl M, 1998, Kaposi’s sarcoma: a result of the interplay among inflammatory cytokines, angiogenic factors and viral agents. Cytokine Growth Factor Rev 9(1):63–83 Chandran B, 2010, Early Events in Kaposi’s Sarcoma-Associated Herpesvirus Infection of Target Cells J Virol 84(5):2188–2199 Browning PJ, Sechler JM, KaplanMet al, 1994, Identification and culture of Kaposi’s sarcoma-like spindle cells from the peripheral blood of human immunodeficiency virus-1-infected individuals and normal controls. Blood 84(8):2711–2720 Burvenich I, Schoonooghe S, Vervoort L et al, 2008, Monoclonal antibody 14C5 targets integrin alphavbeta5. Mol Cancer Ther 7(12):3771–3779 Spitaleri A, Ghitti M, Mari S et al, 2011, Use of metadynamics in the design of isoDGR-based αvβ3 antagonists to fine-tune the conformational ensemble. Angew. Chem. Int. Ed Engl. 50(8):1832– 1836 Orban E, Mezo G, Schlage P et al, 2011, In vitro degradation and antitumor activity of oxime bond-linked daunorubicin-GnRH-III bioconjugates and DNA-binding properties of daunorubicinamino acid metabolites. Amino Acids 41(2):469–483 Scholzen T, Gerdes J, 2000, The Ki-67 protein: from the known and the unknown. J Cell Physiol 182(3):311–322 Folkman J, 1985, Tumor angiogenesis. Adv Cancer Res 43:175– 203 Kuntzman R,Mark LC, Brand LC et al, 1966)Metabolismof drugs and carcinogens by human liver enzymes. J Pharmacol Exp Ther 152(1):151–156 Mittal VK, Bhullar JS, Jayant K, 2015, Animal models of human colorectal cancer: current status, uses and limitations. World J Gastroenterol 21(41):11854–11861 Kapuvari B, Hegedus R, Schulcz A et al, 2016, Improved in vivo antitumor effect of a daunorubicin - GnRH-III bioconjugate modified by apoptosis inducing agent butyric acid on colorectal carcinoma bearing mice. Investig New Drugs 34(4):416–423 Kiss K, Biri-Kovacs B, Szabo R et al, 2019, Sequencemodification of heptapeptide selected by phage display as homing device for HT- 29 colon cancer cells to improve the anti-tumour activity of drug delivery systems. Eur J Med Chem 176:105–116 Randelovic I, Schuster S, Kapuvari B et al, 2019, Improved in vivo anti-tumor and anti-metastatic effect of GnRH-III-Daunorubicin analogs on colorectal and breast carcinoma bearingmice. Int JMol Sci 20(19):4763 Liao H-W, Hung M-C, 2017, Intracaecal Orthotopic colorectal Cancer xenograft mouse model. Bio-Protoc 7(11) Parasuraman S, 2011, Toxicological screening J Pharmacol Pharmacother 2(2):74–79 Hirst DG, Denekamp J, 1979, Tumour cell proliferation in relation to the vasculature. Cell Tissue Kinet 12(1):31–42 Dewan MZ, Terunuma H, Ahmed S et al, 2005, Natural killer cells in breast cancer cell growth and metastasis in SCID mice. Biomed Pharmacother Biomedecine Pharmacother 59(Suppl 2):S375–S379 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1879 EP 1892 DI 10.1007/s12253-019-00773-3 PG 14 ER PT J AU Sun, L Wang, Y Shi, J Zhu, W Wang, X AF Sun, Le Wang, Yusheng Shi, Jinfeng Zhu, Wei Wang, Xin TI Association of Plasma Epstein-Barr Virus LMP1 and EBER1 with Circulating Tumor Cells and the Metastasis of Nasopharyngeal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Epstein-Barr virus; LMP1; BART; EBER1; Metastasis; Nasopharyngeal carcinoma ID Epstein-Barr virus; LMP1; BART; EBER1; Metastasis; Nasopharyngeal carcinoma AB Epstein-Barr virus (EBV) has been widely recognized to contribute to the development of nasopharyngeal carcinoma (NPC). The present study was to explore the association of plasma Epstein-Barr Virus LMP1 and EBER1 with circulating tumor cells (CTCs) and the metastasis of nasopharyngeal carcinoma. In the present study, we quantified the plasma levels of EBV DNA/RNAs, such as LMP1, LMP2, BART and EBER1 with real-time quantitative PCR, and CTCs with a CellSpotter Analyzer in NPC patients, with or without metastasis. Then the correlation of each biomarker with other biomarkers and tumor metastasis was analyzed. Our data indicated that the plasma levels of EBV LMP1, BART, EBER1, along with CTCs were significantly higher in metastatic NPC patients than in non-metastatic patients. Plasma LMP1 DNA and EBER1 discriminate metastatic NPC patients from nonmetastatic patients, correlate with tumor stage and node stage for metastatic NPC patients. In summary, there were significantly higher plasma levels of Epstein-Barr Virus DNAs / RNAs in nasopharyngeal carcinoma patients. LMP1 DNA and EBER1 RNA correlated with the metastasis of nasopharyngeal carcinoma. C1 [Sun, Le] First Hospital of Jilin University, Department of Otolaryngology, Head & Neck Surgery, 71#, Xinmin Street, 130021 Changchun, China. [Wang, Yusheng] First Hospital of Jilin University, Department of Otolaryngology, Head & Neck Surgery, 71#, Xinmin Street, 130021 Changchun, China. [Shi, Jinfeng] First Hospital of Jilin University, Department of Otolaryngology, Head & Neck Surgery, 71#, Xinmin Street, 130021 Changchun, China. [Zhu, Wei] First Hospital of Jilin University, Department of Otolaryngology, Head & Neck Surgery, 71#, Xinmin Street, 130021 Changchun, China. [Wang, Xin] First Hospital of Jilin University, Department of Otolaryngology, Head & Neck Surgery, 71#, Xinmin Street, 130021 Changchun, China. RP Sun, L (reprint author), First Hospital of Jilin University, Department of Otolaryngology, Head & Neck Surgery, 130021 Changchun, China. CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65:87–108. , DOI 10.3322/caac.21262 Chua M, Wee J, Hui EP, Chan A, 2016, Nasopharyngeal carcinoma. Lancet 387:1012–1024. , DOI 10.1016/S0140- 6736(15)00055-0 Smatti MK, Al-Sadeq DW, Ali NH, Pintus G, Abou-Saleh H, Nasrallah GK, 2018, Epstein-Barr virus epidemiology, serology, and genetic variability of LMP-1 Oncogene among healthy population: an update. Front Oncol 8:211. , DOI 10.3389/fonc. 2018.00211 Pathmanathan R, Prasad U, Sadler R, Flynn K, Raab-Traub N, 1995, Clonal proliferations of cells infected with Epstein-Barr virus in preinvasive lesions related to nasopharyngeal carcinoma. N Engl J Med 333:693–698. , DOI 10.1056/ NEJM199509143331103 Liebowitz D, 1994, Nasopharyngeal carcinoma: the Epstein-Barr virus association. Semin Oncol 21:376–381 Tsao SW, Tsang CM, To KF, Lo KW, 2015, The role of Epstein- Barr virus in epithelial malignancies. J Pathol 235:323–333. https:// doi.org/10.1002/path.4448 Zou X, You R, Liu H et al, 2017, Establishment and validation of M1 stage subdivisions for de novo metastatic nasopharyngeal carcinoma to better predict prognosis and guide treatment. Eur J Cancer 77:117–126. , DOI 10.1016/j.ejca.2017.02.029 Taylor GS, Blackbourn DJ, 2011, Infectious agents in human cancers: lessons in immunity and immunomodulation from gammaherpesviruses EBV and KSHV. Cancer Lett 305:263–278. , DOI 10.1016/j.canlet.2010.08.019 Young LS, Arrand JR, Murray PG, 2007, EBV gene expression and regulation. In: Arvin A, Campadelli-Fiume G, Mocarski E, Moore PS, Roizman B, Whitley R, Yamanishi K, eds, Human Herpesviruses: Biology, Therapy, and Immunoprophylaxis, Chapter 27. Cambridge University Press, Cambridge Lin JC, Wang WY, Chen KYet al, 2004, Quantification of plasma Epstein-Barr virus DNA in patients with advanced nasopharyngeal carcinoma. N Engl J Med 350:2461–2470. , DOI 10.1056/ NEJMoa032260 Chan AT, Lo YM, Zee B et al, 2002, Plasma Epstein-Barr virus DNA and residual disease after radiotherapy for undifferentiated nasopharyngeal carcinoma. J Natl Cancer Inst 94:1614–1619 Chan A, Hui EP, Ngan R et al, 2018, Analysis of plasma Epstein- Barr virus DNA in nasopharyngeal Cancer after Chemoradiation to identify high-risk patients for adjuvant chemotherapy: a randomized controlled trial. J Clin Oncol: O2018777847. , DOI 10.1200/JCO.2018.77.7847 Lin JC, Wang WY, Liang WM et al, 2007, Long-term prognostic effects of plasma epstein-barr virus DNA by minor groove binderprobe real-time quantitative PCR on nasopharyngeal carcinoma patients receiving concurrent chemoradiotherapy. Int J Radiat Oncol Biol Phys 68:1342–1348. , DOI 10.1016/j.ijrobp. 2007.02.012 Pegtel DM, Middeldorp J, Thorley-Lawson DA, 2004, Epstein- Barr virus infection in ex vivo tonsil epithelial cell cultures of asymptomatic carriers. J Virol 78:12613–12624. , DOI 10.1128/JVI.78.22.12613-12624.2004 Hudnall SD, Ge Y, Wei L, Yang NP, Wang HQ, Chen T, 2005, Distribution and phenotype of Epstein-Barr virus-infected cells in human pharyngeal tonsils.Mod Pathol 18:519–527. , DOI 10.1038/modpathol.3800369 Shannon-Lowe C, Adland E, Bell AI, Delecluse HJ, RickinsonAB, Rowe M, 2009, Features distinguishing Epstein-Barr virus infections of epithelial cells and B cells: viral genome expression, genome maintenance, and genome amplification. J Virol 83:7749– 7760. , DOI 10.1128/JVI.00108-09 Tao Q, Swinnen LJ, Yang J, Srivastava G, Robertson KD, Ambinder RF, 1999, Methylation status of the Epstein-Barr virus major latent promoter C in iatrogenic B cell lymphoproliferative disease. Application of PCR-based analysis. AmJ Pathol 155:619– 625. , DOI 10.1016/S0002-9440(10)65157-7 Tong JH, Tsang RK, Lo KWet al, 2002, Quantitative Epstein-Barr virus DNA analysis and detection of gene promoter hypermethylation in nasopharyngeal, NP, brushing samples from patients with NP carcinoma. Clin Cancer Res 8:2612–2619 Cosmopoulos K, Pegtel M, Hawkins J et al, 2009, Comprehensive profiling of Epstein-Barr virus microRNAs in nasopharyngeal carcinoma. J Virol 83:2357–2367. https://doi. org/10.1128/JVI.02104-08 Brooks L, Yao QY, Rickinson AB, Young LS, 1992, Epstein-Barr virus latent gene transcription in nasopharyngeal carcinoma cells: coexpression of EBNA1, LMP1, and LMP2 transcripts. J Virol 66: 2689–2697 Prayongrat A, Chakkabat C, Kannarunimit D, Hansasuta P, Lertbutsayanukul C, 2017, Prevalence and significance of plasma Epstein-Barr virus DNA level in nasopharyngeal carcinoma. J Radiat Res 58:509–516. , DOI 10.1093/jrr/rrw128 Lertbutsayanukul C, Kannarunimit D, Prayongrat A, Chakkabat C, Kitpanit S, Hansasuta P, 2018, Prognostic value of plasma EBV DNA for nasopharyngeal Cancer patients during treatment with intensity-modulated radiation therapy and concurrent chemotherapy. Radiol Oncol 52:195–203. , DOI 10.2478/ raon-2018-0016 Kaye KM, Izumi KM, Mosialos G, Kieff E, 1995, The Epstein- Barr virus LMP1 cytoplasmic carboxy terminus is essential for Blymphocyte transformation; fibroblast cocultivation complements a critical function within the terminal 155 residues. J Virol 69: 675–683 Huen DS, Henderson SA, Croom-Carter D, Rowe M, 1995, The Epstein-Barr virus latent membrane protein-1, LMP1, mediates activation of NF-kappa B and cell surface phenotype via two effector regions in its carboxy-terminal cytoplasmic domain. Oncogene 10:549–560 Turunen A, Rautava J, Grenman R, Syrjanen K, Syrjanen S, 2017, Epstein-Barr virus, EBV)-encoded small RNAs, EBERs, associated with poor prognosis of head and neck carcinomas. Oncotarget 8: 27328–27338. , DOI 10.18632/oncotarget.16033 Chang Y, Cheng SD, Tsai CH, 2002, Chromosomal integration of Epstein-Barr virus genomes in nasopharyngeal carcinoma cells. Head Neck 24:143–150 Kaschka-Dierich C, Adams A, Lindahl T et al, 1976, Intracellular forms of Epstein-Barr virus DNA in human tumour cells in vivo. Nature 260:302–306 Pantel K, Brakenhoff RH, Brandt B, 2008, Detection, clinical relevance and specific biological properties of disseminating tumour cells. Nat Rev Cancer 8:329–340. , DOI 10.1038/nrc2375 Lianidou ES, Markou A, 2011, Circulating tumor cells in breast cancer: detection systems, molecular characterization, and future challenges. Clin Chem 57:1242–1255. , DOI 10.1373/ clinchem.2011.165068 Stevens SJ, Brink AA,Middeldorp JM(2005, Profiling of Epstein- Barr virus latent RNA expression in clinical specimens by genespecific multiprimed cDNA synthesis and PCR. Methods Mol Biol 292:27–38 Pavlovitch JH, Didierjean L, Rizk M, Balsan S, Saurat JH, 1983, Skin calcium-binding protein: distribution in other tissues. Am J Phys 244: C50–C57. , DOI 10.1152/ajpcell.1983.244.1.C50 Ramayanti O, Juwana H, Verkuijlen SA et al, 2017, Epstein-Barr virus mRNA profiles and viral DNA methylation status in nasopharyngeal brushings fromnasopharyngeal carcinoma patients reflect tumor origin. Int J Cancer 140:149–162. , DOI 10.1002/ijc.30418 Xu YJ, Zhou R, Zong JF et al, 2019, Epstein-Barr virus-coded miRBART13 promotes nasopharyngeal carcinoma cell growth and metastasis via targeting of the NKIRAS2/NF-kappaB pathway. Cancer Lett 447:33–40. , DOI 10.1016/j.canlet.2019.01.022 Zuo LL, Zhang J, Liu LZ et al, 2017, Cadherin 6 is activated by Epstein-Barr virus LMP1 to mediate EMT and metastasis as an interplay node of multiple pathways in nasopharyngeal carcinoma. Oncogenesis 6:402. , DOI 10.1038/s41389-017-0005-7 Huang D, Song SJ,Wu ZZ et al, 2017, Epstein-Barr virus-induced VEGF and GM-CSF drive nasopharyngeal carcinoma metastasis via recruitment and activation of macrophages. Cancer Res 77: 3591–3604. , DOI 10.1158/0008-5472.CAN-16-2706 Xie XQ, Luo Y, Ma XL et al, 2019, Clinical significance of circulating tumor cells and their expression of cyclooxygenase-2 in patients with nasopharyngeal carcinoma. Eur Rev Med Pharmacol Sci 23:6951–6961. , DOI 10.26355/eurrev_201908_18735 Lee SW, Chen YW, Kuan EC, LanMY(2019, Dual-function nanostructured platform for isolation of nasopharyngeal carcinoma circulating tumor cells and EBV DNA detection. Biosens Bioelectron 142:111509. , DOI 10.1016/j.bios.2019.111509 You R, Liu YP, LinMet al, 2019, Relationship of circulating tumor cells and Epstein-Barr virus DNA to progression-free survival and overall survival in metastatic nasopharyngeal carcinoma patients. Int J Cancer 145:2873–2883. , DOI 10.1002/ijc.32380 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1893 EP 1901 DI 10.1007/s12253-019-00777-z PG 9 ER PT J AU Yu, Ch Chen, J Ma, J Zang, L Dong, F Sun, J Zheng, M AF Yu, Chaoran Chen, Jie Ma, Junjun Zang, Lu Dong, Feng Sun, Jing Zheng, Minhua TI Identification of Key Genes and Signaling Pathways Associated with the Progression of Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Differentially expressed genes; Gene ontology; KEGG pathway; Protein-protein interaction network; Gastric cancer; Gene set enrichment analysis ID Differentially expressed genes; Gene ontology; KEGG pathway; Protein-protein interaction network; Gastric cancer; Gene set enrichment analysis AB Genomic features have been gradually regarded as part of the fundamentals to the clinical diagnosis and treatment for gastric cancer. However, the molecular alterations taking place during the progression of gastric cancer remain unclear. Therefore, identification of potential key genes and pathways in the gastric cancer progression is crucial to clinical practices. The gene expression profile, GSE103236, was retrieved for the identification of the differentially expressed genes (DEGs), followed by gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichments, gene set enrichment analysis (GSEA) and the protein-protein interaction (PPI) networks. Multiple bioinformatics platforms were employed for expression and prognostic analysis. Fresh frozen gastric cancer tissues were used for external validation. A total of 161 DEGs were identified from GSE103236. The PPI network-derived hub genes included collagen type I alpha 1 chain (COL1A1), tissue inhibitor of the metalloproteinases (TIMP1), Secreted Phosphoprotein 1 (SPP1), somatostatin (SST), neuropeptide Y (NPY), biglycan (BGN), matrix metallopeptidase 3 (MMP3), apolipoprotein E (APOE), ATPase H+/K+ transporting alpha subunit (ATP4A), lysyl oxidase (LOX). SPP1 (log rank p = 0.0048, HR = 1.39 [1.1–1.75]) and MMP3 (log rank p < 0.0001, HR = 1.77 [1.44–2.19]) were significantly associated with poor overall survival. Stage-specifically, both COL1A1 and BGN were correlated with significant in stage III and IV gastric cancer cases. LOX showed significant correlation with prognosis in stage I and stage II gastric cancer cases. Furthermore, cg00583003 of SPP1 and cg16466334 of MMP3 exhibited highly methylation level and significant prognostic values (SPP1: HR = 1.625, p = 0.013; MMP3: HR = 0.647, p = 0.011). Hub genes signature displayed a favorable prognostic value (p value = 5.227e-05). APOE demonstrated the highest correlation with CD8+ T cells, neutrophils, and dendritic cells whereas BGN had the highest correlation with macrophages. This study systematically explored the key genes and pathways involved in PGC and AGC, providing insights into therapeutic individualized management. C1 [Yu, Chaoran] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Gastrointestinal Surgery, 200025 Shanghai, China. [Chen, Jie] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Gastrointestinal Surgery, 200025 Shanghai, China. [Ma, Junjun] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Gastrointestinal Surgery, 200025 Shanghai, China. [Zang, Lu] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Gastrointestinal Surgery, 200025 Shanghai, China. [Dong, Feng] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Gastrointestinal Surgery, 200025 Shanghai, China. [Sun, Jing] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Gastrointestinal Surgery, 200025 Shanghai, China. [Zheng, Minhua] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Gastrointestinal Surgery, 200025 Shanghai, China. RP Yu, Ch (reprint author), Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Gastrointestinal Surgery, 200025 Shanghai, China. EM chaoran_yu@sjtu.edu.cn CR Siegel RL, Miller KD, Jemal A, 2016, Cancer statistics, 2016[J]. CA Cancer J Clin 66(1):7–30 Chen W, Zheng R, Baade P D, et al., 2015, Cancer statistics in China[J] Ajani JA, Bentrem DJ, Besh S, D'Amico TA, Das P, Denlinger C, Fakih MG, Fuchs CS, Gerdes H, Glasgow RE, Hayman JA, Hofstetter WL, Ilson DH, Keswani RN, Kleinberg LR, Korn WM, Lockhart AC, Meredith K, Mulcahy MF, Orringer MB, Posey JA, Sasson AR, Scott WJ, Strong VE, Varghese TK Jr, Warren G, Washington MK, Willett C, Wright CD, McMillian N, Sundar H, National Comprehensive Cancer Network, 2013, Gastric cancer, version 2.2013[J]. J Natl Compr Cancer Netw 11(5):531–546 Edge SB, Compton CC, 2010, The American joint committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM[J]. Ann Surg Oncol 17(6):1471–1474 Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC,Wong SS, Liu J, Yue YG, Wang J, Yu K, Ye XS, Do IG, Liu S, Gong L, Fu J, Jin JG, ChoiMG, Sohn TS, Lee JH, Bae JM, Kim ST, Park SH, Sohn I, Jung SH, Tan P, Chen R, Hardwick J, Kang WK, Ayers M, Hongyue D, Reinhard C, Loboda A, Kim S, Aggarwal A, 2015, Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes[J]. Nat Med 21(5):449–456 Cancer Genome Atlas Research Network, 2014, Comprehensive molecular characterization of gastric adenocarcinoma[J]. Nature 513(7517):202–209 Chivu EM, Necula LG, Dragu D et al, 2010, Identification of potential biomarkers for early and advanced gastric adenocarcinoma detection[J]. Hepato-Gastroenterol 57(104):1453 Edgar R, Domrachev M, Lash AE, 2002, Gene expression omnibus: NCBI gene expression and hybridization array data repository[J]. Nucleic Acids Res 30(1):207–210 Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, Yefanov A, Lee H, Zhang N, Robertson CL, Serova N, Davis S, Soboleva A, 2013, NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res 41(Database issue):D991-5 Davis S, Meltzer PS, 2007, GEOquery: a bridge between the gene expression omnibus, GEO, and BioConductor[J]. Bioinformatics 23(14):1846–1847 Pathan M, Keerthikumar S, Ang CS, Gangoda L, Quek CY, Williamson NA, Mouradov D, Sieber OM, Simpson RJ, Salim A, Bacic A, 2015, FunRich: an open access standalone functional enrichment and interaction network analysis tool. Proteomics 15(15):2597–2601 Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G, 2000, Gene ontology: tool for the unification of biology[J]. Nat Genet 25(1):25–29 Kanehisa M, Goto S, 2000, KEGG: Kyoto encyclopedia of genes and genomes[J]. Nucleic Acids Res 28(1):27–30 Huang DW, Sherman BT, Lempicki RA, 2009, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources[J]. Nat Protoc 4(1):44–57 Subramanian A, Tamayo P, Mootha VK et al, 2005, Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles[J]. Proc Natl Acad Sci 102(43): 15545–15550 Szklarczyk D, Franceschini A, Wyder S et al, 2014, STRING v10: protein–protein interaction networks, integrated over the tree of life[J]. Nucleic Acids Res 43(D1):D447–D452 Tang Z, Li C, Kang B, et al., 2017, GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses[J]. Nucleic Acids Res Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, Antipin Y, Reva B, Goldberg AP, Sander C, Schultz N, 2012, The cBio Cancer genomics portal: an open platform for exploring multidimensional Cancer genomics data. Cancer Discov 2(5):401–404 Gao J et al, 2013, Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Sci Signal 6(269):pl1–pl1 Cho JY, Lim JY, Cheong JH, Park YY, Yoon SL, KimSM, KimSB, Kim H, Hong SW, Park YN, Noh SH, Park ES, Chu IS, HongWK, Ajani JA, Lee JS, 2011, Gene expression signature-based prognostic risk score in gastric cancer. Clin Cancer Res 17(7):1850–1857 Cui J, Chen Y, Chou WC, Sun L, Chen L, Suo J, Ni Z, Zhang M, Kong X, Hoffman LL, Kang J, Su Y, Olman V, Johnson D, Tench DW, Amster IJ, Orlando R, Puett D, Li F, Xu Y, 2011, An integrated transcriptomic and computational analysis for biomarker identification in gastric cancer. Nucleic Acids Res 39(4):1197–1207 Uhlen M, Oksvold P, Fagerberg L, Lundberg E, Jonasson K, Forsberg M, Zwahlen M, Kampf C, Wester K, Hober S, Wernerus H, Bjorling L, Ponten F, 2010, Towards a knowledge-based human protein atlas. Nat Biotechnol 28(12):1248–1250 Spandidos A, Wang X, Wang H, Seed B, 2010, PrimerBank: a resource of human and mouse PCR primer pairs for gene expression detection and quantification. Nucleic Acids Res 38:D792– D799 Lanczky A, Nagy A, Bottai G, Munkacsy G, Paladini L, Szabo A, Santarpia L, Gyorffy B, 2016, miRpower: a web-tool to validate survival-associated miRNAs utilizing expression data from 2,178 breast cancer patients. Breast Cancer Res Treat 160(3):439–446 Modhukur V, Iljasenko T, Metsalu T, Lokk K, Laisk-Podar T, Vilo J, 2018Mar)MethSurv: a web tool to perform multivariable survival analysis using DNA methylation data. Epigenomics. 10(3):277– 288 Aguirre-Gamboa R, Gomez-Rueda H, Martinez-Ledesma E, Martinez-Torteya A, Chacolla-Huaringa R, Rodriguez-Barrientos A, Tamez-Pena JG, Trevino V, 2013, SurvExpress: an online biomarker validation tool and database for cancer gene expression data using survival analysis. PLoS One 8(9):e74250 Li T, Fan J,Wang B, Traugh N, Chen Q, Liu JS, Li B, Liu XS, 2017 Nov 1, TIMER: a web server for comprehensive analysis of tumorinfiltrating immune cells. Cancer Res 77(21):e108–e110 Hwang SH, Park DJ, Jee YS, Kim MC, Kim HH, Lee HJ, Yang HK, Lee KU, 2009, Actual 3-year survival after laparoscopyassisted gastrectomy for gastric cancer[J]. Arch Surg 144(6):559– 564 HuY, YingM,Huang C,Wei H, Jiang Z, Peng X,Hu J, du X,Wang B, Lin F, Xu J, Dong G, Mou T, Li G, Chinese Laparoscopic Gastrointestinal Surgery Study, CLASS, Group, 2014, Oncologic outcomes of laparoscopy-assisted gastrectomy for advanced gastric cancer: a large-scale multicenter retrospective cohort study from China[J]. Surg Endosc 28(7):2048–2056 Rugge M, Fassan M, Graham D Y, 2015, Epidemiology of gastric cancer[M]//gastric Cancer. Springer International Publishing, 23– 34 Strong VE, Song KY, Park CH, Jacks LM, GonenM, ShahM, Coit DG, Brennan MF, 2010, Comparison of gastric cancer survival following R0 resection in the United States and Korea using an internationally validated nomogram[J]. Ann Surg 251(4):640–646 Coussens LM, Werb Z, 2002, Inflammation and cancer[J]. Nature 420(6917):860–867 Liaw L, Birk DE, Ballas CB,Whitsitt JS, Davidson JM, Hogan BL, 1998, Altered wound healing in mice lacking a functional osteopontin gene, spp1)[J]. J Clin Investig 101(7):1468–1478 Spiegel S, Milstien S, 2003, Sphingosine-1-phosphate: an enigmatic signalling lipid[J]. Nat Rev Mol Cell Biol 4(5):397–407 Junnila S, Kokkola A, Mizuguchi T, Hirata K, Karjalainen- Lindsberg ML, Puolakkainen P, Monni O, 2010, Gene expression analysis identifies over-expression of CXCL1, SPARC, SPP1, and SULF1 in gastric cancer[J]. Genes Chromosom Cancer 49(1):28– 39 Zhuo C, Li X, Zhuang H, Tian S, Cui H, Jiang R, Liu C, Tao R, Lin X, 2016, Elevated THBS2, COL1A2, and SPP1 expression levels as predictors of gastric Cancer prognosis[J]. Cell Physiol Biochem 40(6):1316–1324 SternlichtMD, Lochter A, Sympson CJ, Huey B, Rougier JP, Gray JW, Pinkel D, Bissell MJ, Werb Z, 1999, The stromal proteinase MMP3/stromelysin-1 promotes mammary carcinogenesis[J]. Cell 98(2):137–146 Kubota E, Imamura H, Kubota T et al, 1997, Interleukin 1β and stromelysin, MMP3, activity of synovial fluid as possible markers of osteoarthritis in the temporomandibular joint[J]. J Oral Maxillofac Surg 55(1):20–27 Stamenkovic I, 2003, Extracellular matrix remodelling: the role of matrix metalloproteinases[J]. J Pathol 200(4):448–464 Yeh YC, Sheu BS, Cheng HC, Wang YL, Yang HB, Wu JJ, 2010, Elevated serum matrix metalloproteinase-3 and-7 in H. pylorirelated gastric cancer can be biomarkers correlating with a poor survival[J]. Dig Dis Sci 55(6):1649–1657 Xu J,YaoY, Ren S et al, 2016)Matrix metalloproteinase expression and molecular interaction network analysis in gastric cancer[J]. Oncol Lett 12(4):2403–2408 Fernandez-Capetillo O, Chen HT, Celeste A, Ward I, Romanienko PJ,Morales JC, Naka K, Xia Z, Camerini-Otero RD,Motoyama N, Carpenter PB, Bonner WM, Chen J, Nussenzweig A, 2002, DNA damage-induced G2–M checkpoint activation by histone H2AX and 53BP1[J]. Nat Cell Biol 4(12):993–997 Sun C, Yuan Q,Wu D et al, 2017, Identification of core genes and outcome in gastric cancer using bioinformatics analysis[J]. Oncotarget 8(41):70271 Guo L, Song C,Wang P, Dai L, Zhang J,Wang K, 2015, A systems biology approach to detect key pathways and interaction networks in gastric cancer on the basis of microarray analysis[J]. Mol Med Rep 12(5):7139–7145 Rong L, HuangW, Tian S, Chi X, Zhao P, Liu F, 2018, COL1A2 is a novel biomarker to improve clinical prediction in human gastric Cancer: integrating bioinformatics and meta-analysis[J]. Pathol Oncol Res 24(1):129–134 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1903 EP 1919 DI 10.1007/s12253-019-00781-3 PG 17 ER PT J AU Jahangiri, R Mosaffa, F EmamiRazavi, A Gharib, M Jamialahmadi, K AF Jahangiri, Rosa Mosaffa, Fatemeh EmamiRazavi, Amirnader Gharib, Masoumeh Jamialahmadi, Khadijeh TI Increased Expression of Gankyrin and Stemness Factor Oct-4 are Associated with Unfavorable Clinical Outcomes and Poor Benefit of Tamoxifen in Breast Carcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast Cancer; Tamoxifen Resistance; Gankyrin; Oct-4 ID Breast Cancer; Tamoxifen Resistance; Gankyrin; Oct-4 AB Tamoxifen is the most important treatment component in estrogen receptor positive (ER+) breast carcinoma patients. Tamoxifen resistance incidence presents an important obstacle in clinical treatment. Mechanisms underlying tamoxifen refractory are not completely understood. Although elevated expression of Gankyrin (P28GANK) and stem cell markers Nanog, Oct-4 and Sox-2 have been reported in breast carcinoma, their role in tamoxifen resistance progression has not been explored. In the present study, P28GANK and stem cell markers Nanog, Oct-4 and Sox-2 expression were evaluated using quantitative RT-PCR and immunohistochemical technology in 72 breast carcinoma patients who received tamoxifen as adjuvant anti-hormone treatment. Expression data were correlated with the clinical outcome and survival of patients. Data analysis showed that P28GANK, Oct-4 and Sox-2 transcripts were significantly overexpressed in tamoxifen resistance patients. Immunohistochemical staining indicated that protein expression of P28GANK and Oct-4 were also significantly higher in tamoxifen resistance patients. We have shown a positive correlation between mRNA and protein expression of P28GANK, Oct-4 and Sox-2. Multivariate logistic regression analysis indicated that P28GANK (P = 0.002) and Oct-4 (P = 0.013) overexpression could be negative independent factors of disease outcome. Additionally, in the whole study group, multivariate Cox regression analysis revealed that high expression of P28GANK and Oct-4 remained significant and unfavorable predictive factors for patients’ survival. These findings suggest that Gankyrin and Oct-4 overexpression could promote tamoxifen refractory in breast cancer patients. More studies are warranted to clarify the predictive role of these potential biomarkers for patients who don’t benefit from tamoxifen treatment and their possible application as prognostic markers in ER+ tamoxifen-treated breast carcinoma patients. C1 [Jahangiri, Rosa] Mashhad University of Medical Sciences, School of Medicine, Department of Medical Biotechnology and NanotechnologyMashhad, Iran. [Mosaffa, Fatemeh] Mashhad University of Medical Sciences, Pharmaceutical Technology Institute, Biotechnology Research CenterMashhad, Iran. [EmamiRazavi, Amirnader] Tehran University of Medical Sciences, Cancer Institute of IranTehran, Iran. [Gharib, Masoumeh] Mashhad University of Medical Sciences, Faculty of Medicine, Department of PathologyMashhad, Iran. [Jamialahmadi, Khadijeh] Mashhad University of Medical Sciences, Pharmaceutical Technology Institute, Biotechnology Research CenterMashhad, Iran. RP Jamialahmadi, K (reprint author), Mashhad University of Medical Sciences, Pharmaceutical Technology Institute, Biotechnology Research Center, Mashhad, Iran. EM jamialahmadikh@mums.ac.ir CR Johansson HJ, Sanchez BC, Mundt F, Forshed J, Kovacs A, Panizza E, Hultin-Rosenberg L, Lundgren B, Martens U, Mathe G, Yakhini Z, Helou K, Krawiec K, Kanter L, Hjerpe A, Stal O, Linderholm BK, Lehtio J, 2013, Retinoic acid receptor alpha is associated with tamoxifen resistance in breast cancer. Nat Commun 4:2175. , DOI 10.1038/ncomms3175 Hayes EL, Lewis-Wambi JS, 2015, Mechanisms of endocrine resistance in breast cancer: an overview of the proposed roles of noncoding RNA. Breast cancer research : BCR 17:40. https://doi. org/10.1186/s13058-015-0542-y Szostakowska M, Trebinska-Stryjewska A, Grzybowska EA, Fabisiewicz A, 2019, Resistance to endocrine therapy in breast cancer: molecular mechanisms and future goals. 173, 3):489-497. , DOI 10.1007/s10549-018-5023-4 Kumar SM, Liu S, Lu H, Zhang H, Zhang PJ, Gimotty PA, Guerra M, Guo W, Xu X, 2012, Acquired cancer stem cell phenotypes through Oct4-mediated dedifferentiation. Oncogene 31(47):4898– 4911 http://www.nature.com/onc/journal/v31/n47/suppinfo/ onc2011656s1.html Yuan J, Liu M, Yang L, Tu G, Zhu Q, Chen M, Cheng H, Luo H, Fu W, Li Z, Yang G, 2015, Acquisition of epithelialmesenchymal transition phenotype in the tamoxifen-resistant breast cancer cell: a new role for G protein-coupled estrogen receptor in mediating tamoxifen resistance through cancerassociated fibroblast-derived fibronectin and β1-integrin signaling pathway in tumor cells. Breast Cancer Res 17(1):69. https:// doi.org/10.1186/s13058-015-0579-y Maynard PV, Blamey RW, Elston CW, Haybittle JL, Griffiths K, 1978, estrogen receptor assay in primary breast cancer and early recurrence of the disease. Cancer Research 38, 11 Part 2):4292- 4295 Simoes BM, Piva M, Iriondo O, Comaills V, Lopez-Ruiz JA, Zabalza I, Mieza JA, Acinas O, Vivanco MM, 2011, Effects of estrogen on the proportion of stem cells in the breast. Breast Cancer Res Treat 129(1):23–35. , DOI 10.1007/s10549- 010-1169-4 Higashitsuji H, Itoh K, Nagao T, Dawson S, Nonoguchi K, Kido T, Mayer RJ, Arii S, Fujita J, 2000, Reduced stability of retinoblastoma protein by gankyrin, an oncogenic ankyrin-repeat protein overexpressed in hepatomas. Nat Med 6(1):96–99. , DOI 10.1038/71600 Li H, Zhang J, Zhen C, Yang B, Feng L, 2018, Gankyrin as a potential target for tumor therapy: evidence and perspectives. Am J Transl Res 10(7):1949–1960 Higashitsuji H, Higashitsuji H, Itoh K, Sakurai T, Nagao T, Sumitomo H, Masuda T, Dawson S, Shimada Y, Mayer RJ, Fujita J, 2005, The oncoprotein gankyrin binds to MDM2/HDM2, enhancing ubiquitylation and degradation of p53. Cancer Cell 8(1): 75–87. , DOI 10.1016/j.ccr.2005.06.006 Mandlekar S, Kong AN, 2001, Mechanisms of tamoxifen-induced apoptosis. Apoptosis: An International Journal on Programmed Cell Death 6(6):469–477 Gelmann EP, 1996, Tamoxifen induction of apoptosis in estrogen receptor-negative cancers: new tricks for an old dog? J Natl Cancer Inst 88(5):224–226 Kim YH, Kim JH, Choi YW, Lim SK, Yim H, Kang SY, Chung YS, Lee GY, Park TJ, 2013, Gankyrin is frequently overexpressed in breast cancer and is associated with ErbB2 expression. Exp Mol Pathol 94(2):360–365. , DOI 10.1016/j.yexmp.2012.12. 002 Mine H, Sakurai T, Kashida H, Matsui S, Nishida N, Nagai T, Hagiwara S, Watanabe T, Kudo M, 2013, Association of gankyrin and stemness factor expression in human colorectal cancer. Dig Dis Sci 58(8):2337–2344. , DOI 10.1007/s10620-013-2627-8 Jahangiri R, Mosaffa F, Emami Razavi A, Teimoori-Toolabi L, Jamialahmadi K, 2018, Altered DNA methyltransferases promoter methylation and mRNA expression are associated with tamoxifen response in breast tumors. J Cell Physiol 233(9):7305–7319. , DOI 10.1002/jcp.26562 Wang H, Yang B, Geng T, Li B, Dai P, Chen C, 2015, Tissuespecific selection of optimal reference genes for expression analysis of anti-cancer drug-related genes in tumor samples using quantitative real-time RT-PCR. ExpMol Pathol 98(3):375–381. https://doi. org/10.1016/j.yexmp.2014.10.014 Wang W-P, Sun Y, Lu Q, Zhao J-B, Wang X-J, Chen Z, Ni Y-F, Wang J-Z, Han Y, Zhang Z-P, Yan X-L, Li X-F, 2016, Gankyrin promotes epithelial-mesenchymal transition and metastasis in NSCLC through forming a closed circle with IL-6/ STAT3 and TGF-β/SMAD3 signaling pathway. Oncotarget 8(4):5909–5923. , DOI 10.18632/oncotarget.13947 Mosaffa F, Kalalinia F, Lage H, Afshari JT, Behravan J, 2012, Proinflammatory cytokines interleukin-1 beta, interleukin 6, and tumor necrosis factor-alpha alter the expression and function ofABCG2 in cervix and gastric cancer cells.Mol Cell Biochem 363(1):385–393. , DOI 10.1007/s11010-011-1191-9 Jahangiri R, Mosaffa F, Gharib M, Emami Razavi AN, Abdirad A, Jamialahmadi K, 2018, PAX2 expression is correlated with better survival in tamoxifen-treated breast carcinoma patients. Tissue Cell 52:135–142. , DOI 10.1016/j.tice.2018.05.005 Amaya CN, Bryan BA, 2015, Enrichment of the embryonic stem cell reprogramming factors Oct4, Nanog,Myc, and Sox2 in benign andmalignant vascular tumors. BMC Clin Pathol 15:18. https://doi. org/10.1186/s12907-015-0018-0 Zeng YC, Sun D, LiWH, Zhao J, Xin Y, 2017, Gankyrin promotes the proliferation of gastric cancer and is associated with chemosensitivity. Tumour Biology: the Journal of the International Society for Oncodevelopmental Biology and Medicine 39(6):1010428317704820. , DOI 10.1177/ 1010428317704820 Tanic N, Milovanovic Z, Tanic N, Dzodic R, Juranic Z, Susnjar S, Plesinac-Karapandzic V, Tatic S, Dramicanin T, Davidovic R, Dimitrijevic B, 2012, The impact of PTEN tumor suppressor gene on acquiring resistance to tamoxifen treatment in breast cancer patients. Cancer Biology & Therapy 13(12):1165–1174. https://doi. org/10.4161/cbt.21346 Thrane S, Pedersen AM, Thomsen MBH, Kirkegaard T, Rasmussen BB, Duun-Henriksen AK, Lankholm AV, Bak M, Lykkesfeldt AE, Yde CW, 2015, A kinase inhibitor screen identifies Mcl-1 and Aurora kinase A as novel treatment targets in antiestrogen-resistant breast cancer cells. Oncogene 34(32):4199– 4210. , DOI 10.1038/onc.2014.351 Zamani P, Matbou Riahi M, Momtazi-Borojeni AA, Jamialahmadi K, 2018, Gankyrin: a novel promising therapeutic target for hepatocellular carcinoma. Artificial Cells, Nanomedicine, and Biotechnology 46(7):1301–1313. , DOI 10.1080/ 21691401.2017.1388250 Taheri M, Jamialahmadi K, Kalalinia F, 2017, Unexpected Lower Expression of Oncoprotein Gankyrin in Drug Resistant ABCG2 Overexpressing Breast Cancer Cell Lines. Asian Pac J Cancer Prev 18(12):3413–3418. , DOI 10.22034/apjcp.2017.18. 12.3413 Chen X, ZhangM, Liu LX, 2009, The overexpression of multidrug resistance-associated proteins and gankyrin contribute to arsenic trioxide resistance in liver and gastric cancer cells. Oncol Rep 22(1):73–80 Qian YW, ChenY, YangW, Fu J, Cao J, Ren YB, Zhu JJ, Su B, Luo T, Zhao XF, Dai RY, Li JJ, Sun W, Wu MC, Feng GS, Wang HY, 2012, p28(GANK, prevents degradation of Oct4 and promotes expansion of tumor-initiating cells in hepatocarcinogenesis. Gastroenterology 142(7):1547–1558. e1514. , DOI 10. 1053/j.gastro.2012.02.042 Sun J, Xu K, Qiu Y, Gao H, Xu J, Tang Q, Yin P, 2017, Bufalin reverses acquired drug resistance by inhibiting stemness in colorectal cancer cells. Oncol Rep 38(3):1420–1430. , DOI 10. 3892/or.2017.5826 Rodriguez D, Ramkairsingh M, Lin X, Kapoor A,Major P, Tang D, 2019, The Central Contributions of Breast Cancer Stem Cells in Developing Resistance to Endocrine Therapy in Estrogen Receptor, ER)-Positive Breast Cancer. Cancers 11(7). , DOI 10. 3390/cancers11071028 Arif K, Hussain I, Rea C, El-Sheemy M, 2015, The role of Nanog expression in tamoxifen-resistant breast cancer cells. OncoTargets and Therapy 8:1327–1334. , DOI 10.2147/OTT.S67835 Leung EY, Askarian-AmiriME, Sarkar D, Ferraro-Peyret C, Joseph WR, Finlay GJ, Baguley BC, 2017, Endocrine Therapy of Estrogen Receptor-Positive Breast Cancer Cells: Early Differential Effects on Stem Cell Markers. Front Oncol 7:184. , DOI 10.3389/ fonc.2017.00184 Zhong B, Lin Y, Lai Y, Zheng F, Zheng X, Huang R, YangW, Chen Z, 2016, Relationship of Oct-4 to malignant stage: a meta-analysis based on 502 positive/high Oct-4 cases and 522 negative/low casefree controls. Oncotarget 7(2):2143–2152. , DOI 10. 18632/oncotarget.5737 Zhao X, Liu F, Zhang Y, Li P, 2016, Prognostic and clinicopathological significance of Gankyrin overexpression in cancers: evidence from a meta-analysis. Onco Targets Ther 9:1961–1968. , DOI 10.2147/OTT.S101687 Yang F, Zhang J, Yang H, 2018, OCT4, SOX2, and NANOG positive expression correlates with poor differentiation, advanced disease stages, and worse overall survival in HER2(+, breast cancer patients. Onco Targets Ther 11:7873–7881. , DOI 10. 2147/ott.s173522 Wilbertz T,Wagner P, Petersen K, Stiedl AC, Scheble VJ, Maier S, ReischlM,Mikut R, Altorki NK, Moch H, Fend F, Staebler A, Bass AJ, MeyersonM, Rubin MA, Soltermann A, Lengerke C, Perner S, 2011, SOX2 gene amplification and protein overexpression are associated with better outcome in squamous cell lung cancer. Modern pathology: an Official Journal of the United States and Canadian Academy of Pathology, Inc 24(7):944–953. https://doi. org/10.1038/modpathol.2011.49 Yu B, Cai H, Xu Z, Xu T, Zou Q, GuM(2016, Expressions of stem cell transcription factors Nanog and Oct4 in renal cell carcinoma tissues and clinical significance. Artificial Cells, Nanomedicine, and Biotechnology 44(8):1818–1823. , DOI 10.3109/ 21691401.2015.1105238 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1921 EP 1934 DI 10.1007/s12253-019-00766-2 PG 14 ER PT J AU Dong, R Liu, J Sun, W Ping, W AF Dong, Ruolan Liu, Jiawei Sun, Wei Ping, Wei TI Comprehensive Analysis of Aberrantly Expressed Profiles of lncRNAs and miRNAs with Associated ceRNA Network in Lung Adenocarcinoma and Lung Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung adenocarcinoma (LUAD); Lung squamous cell carcinoma (LUSC); lncRNA; ceRNA; miRNA ID Lung adenocarcinoma (LUAD); Lung squamous cell carcinoma (LUSC); lncRNA; ceRNA; miRNA AB Lung cancer (LC) continues to be the leading cause of cancer-related deaths worldwide and the prognosis remains poor worldwide. At present, the long non-coding RNAs (lncRNAs) was considered as a part of competing endogenous RNA (ceRNA) network act as natural microRNA (miRNA) sponges to regulate protein-coding gene expression. However, functional roles of lncRNA-mediated ceRNAs in LC are insufficiently understood. To classify the specific mechanism of lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), we comprehensively compared the expression profiles of mRNAs, lncRNAs and miRNAs obtained from 509 LUAD, 473 LUSC tissues and 49 adjacent non-cancerous lung tissues, based on The Cancer Genome Atlas (TCGA). After screening for differently expressed (DE) mRNAs,DEmiRNAs, DElncRNAs and weighted gene co-expression network analysis (WGCNA) (|log2FC| > 2.0 and an adjusted p value <0.05), a total of 4478 DEmRNAs, 526 DElncRNAs and 75 DEmiRNAs in LUAD, while 6237 DEmRNAs, 843 DElncRNAs and 117 DEmiRNAs in LUSC were discovered. Interaction (PPI) network analysis was performed to identify 656 nodes and 2987 edges (minimum required interaction score > 0.9), as well as 8 different protein-protein interactions. Gene ontology (GO) analysis mainly associated with cell proliferation. KEGG pathway enrichment analyses most partly associated with metabolism pathway and cytokine-cytokine receptor interaction. Finally, the dysregulated lncRNA-miRNA-ceRNA network was constructed based on correlation analyses and a total of 62 dysregulated lncRNAs, 28 DEmRNAs and 18 DEmiRNAs were involved. The most significant lncRNAs included DElncRNAs, LINC00641 and AC004947.2, miRNAs included miR-6860, miR-1285-3p, miR-767-3p and miR-7974, mRNAs included MAP3K3, FGD3 and ATP1B2. Then we analyzed and described the potential characteristics of biological function and pathological roles of the LUAD and LUSC ceRNA co-regulatory network. Our findings revealed ceRNA network will be beneficial for promoting the understanding of lncRNA-mediated ceRNA regulatory mechanisms in the pathogenesis of LUAD and LUSC. C1 [Dong, Ruolan] Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Institute of Integrated Traditional Chinese and Western Medicine, 430030 Wuhan, Hubei, China. [Liu, Jiawei] Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Department of Thoracic Surgery, 430030 Wuhan, Hubei, China. [Sun, Wei] Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Department of Thoracic Surgery, 430030 Wuhan, Hubei, China. [Ping, Wei] Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Department of Thoracic Surgery, 430030 Wuhan, Hubei, China. RP Ping, W (reprint author), Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Department of Thoracic Surgery, 430030 Wuhan, China. EM 247046170@qq.com CR Siegel RL, Miller KD, Jemal A, 2017, Cancer statistics, 2017. CA Cancer J Clin 67(1):7–30. , DOI 10.3322/caac.21387 Goldstraw P, BallD, Jett JR, Le Chevalier T, Lim E, NicholsonAG, Shepherd FA, 2011)Non-small-cell lung cancer. Lancet 378(9804): 1727–1740. , DOI 10.1016/s0140-6736(10)62101-0 Ding L, Getz G,Wheeler DA, Mardis ER,McLellanMD, Cibulskis K, Sougnez C, Greulich H, Muzny DM, Morgan MB, Fulton L, Fulton RS, Zhang Q,WendlMC, Lawrence MS, Larson DE, Chen K, Dooling DJ, Sabo A, Hawes AC, Shen H, Jhangiani SN, Lewis LR, Hall O, Zhu Y, Mathew T, Ren Y, Yao J, Scherer SE, Clerc K, Metcalf GA, Ng B, Milosavljevic A,Gonzalez-GarayML, Osborne JR, Meyer R, Shi X, Tang Y, Koboldt DC, Lin L, Abbott R, Miner TL, Pohl C, Fewell G, Haipek C, Schmidt H, Dunford-Shore BH, Kraja A, Crosby SD, Sawyer CS, Vickery T, Sander S, Robinson J, WincklerW, Baldwin J, Chirieac LR, Dutt A, Fennell T, Hanna M, Johnson BE, Onofrio RC, Thomas RK, Tonon G, Weir BA, Zhao X, Ziaugra L, Zody MC, Giordano T, Orringer MB, Roth JA, Spitz MR, Wistuba II, Ozenberger B, Good PJ, Chang AC, Beer DG, Watson MA, Ladanyi M, Broderick S, Yoshizawa A, Travis WD, Pao W, Province MA, Weinstock GM, Varmus HE, Gabriel SB, Lander ES, Gibbs RA, Meyerson M, Wilson RK, 2008, Somatic mutations affect key pathways in lung adenocarcinoma. Nature 455:1069–1075. , DOI 10.1038/nature07423. https:// www.nature.com/articles/nature07423#supplementary-information Wistuba II, Lam S, Behrens C, Virmani AK, Fong KM, LeRiche J, Samet JM, Srivastava S, Minna JD, Gazdar AF, 1997, Molecular damage in the bronchial epithelium of current and former smokers. J Natl Cancer Inst 89(18):1366–1373 Felekkis K, Touvana E, Stefanou C, Deltas C, 2010, microRNAs: a newly described class of encoded molecules that play a role in health and disease. Hippokratia 14(4):236–240 Ponting CP, Oliver PL, Reik W, 2009, Evolution and functions of long noncoding RNAs. Cell 136(4):629–641. , DOI 10. 1016/j.cell.2009.02.006 Dykes IM, Emanueli C, 2017, Transcriptional and posttranscriptional gene regulation by Long non-coding RNA. Genomics, proteomics & bioinformatics 15(3):177–186. https:// doi.org/10.1016/j.gpb.2016.12.005 Prensner JR, Chinnaiyan AM(2011, The emergence of lncRNAs in cancer biology. Cancer discovery 1(5):391–407. , DOI 10. 1158/2159-8290.Cd-11-0209 Rinn JL, Chang HY, 2012, Genome regulation by long noncoding RNAs. Annu Rev Biochem 81:145–166. , DOI 10.1146/ annurev-biochem-051410-092902 Kristensen LS, Hansen TB, Veno MT, Kjems J, 2018, Circular RNAs in cancer: opportunities and challenges in the field. Oncogene 37(5):555–565. , DOI 10.1038/onc.2017.361 Song YX, Sun JX, Zhao JH, Yang YC, Shi JX,Wu ZH, Chen XW, Gao P,Miao ZF,Wang ZN, 2017, Non-coding RNAs participate in the regulatory network of CLDN4 via ceRNA mediated miRNA evasion. Nat Commun 8(1):289. , DOI 10.1038/s41467- 017-00304-1 Wang Y, Liu Z, Yao B, Li Q, Wang L,Wang C, Dou C, Xu M, Liu Q, Tu KJMC, 2017, Long non-coding RNA CASC2 suppresses epithelial-mesenchymal transition of hepatocellular carcinoma cells through CASC2/miR-367/FBXW7 axis. Mol Cancer 16(1):123 Anders S, Huber W, 2010, Differential expression analysis for sequence count data. Genome Biol 11(10):R106. , DOI 10.1186/gb-2010-11-10-r106 Franceschini A, Szklarczyk D, Frankild S, KuhnM, Simonovic M, Roth A, Lin J,Minguez P, Bork P, von Mering C, Jensen LJ, 2013, STRING v9.1: protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res 41(Database issue): D808–D815. , DOI 10.1093/nar/gks1094 Zhao L, Ni X, Zhao L, Zhang Y, Jin D, YinW, Wang D, ZhangW, 2018, MiroRNA-188 acts as tumor suppressor in non-small-cell lung Cancer by targeting MAP3K3. Mol Pharm 15(4):1682–1689. , DOI 10.1021/acs.molpharmaceut.8b00071 Wei J, Dong Y, Lin T, Pang L, Yan R, Liang W, Jiang J, Gang C, Wen JZ, Yuan XJHP, 2016)MAP3K3 overexpression is associated with poor survival in ovarian carcinoma. Hum Pathol 50:162–169 He Y,Wang L, LiuW, Zhong J, Bai S,Wang Z, Thomas DG, Lin J, Reddy RM, Ramnath N, Carrott PW, Lynch WR, Orringer MB, Chang AC, BeerDG, ChenG, 2015)MAP3K3 expression in tumor cells and tumor-infiltrating lymphocytes is correlated with favorable patient survival in lung cancer. Sci Rep 5:11471–11413. , DOI 10.1038/srep11471 Ou YT, Cheng WY, Zheng T, Maurer MA, Anastassiou DJCEBP, 2014, Breast cancer prognostic biomarker using attractor metagenes and the FGD3-SUSD3 metagene. Cancer Epidemiol Biomark Prev 23(12):2850–2856 Garcia-Closas MJIJC, 2010, Common genetic variation in TP53 and its flanking genes, WDR79 and ATP1B2, and susceptibility to breast cancer. Int J Cancer 121(11):2532–2538 Vijayvergia N, Shah PC, Denlinger CS, 2015, Survivorship in nonsmall cell lung Cancer: challenges faced and steps forward. J Natl Compr Cancer Netw 13(9):1151–1161 Yang Y-R, Zang S-Z, Zhong C-L, Li Y-X, Zhao S-S, X-JJIjoc F, 2014, Increased expression of the lncRNA PVT1 promotes tumorigenesis in non-small cell lung cancer. Int J Clin Exp Pathol 7(10): 6929–6935 Wang C, Han C, Zhang Y, Liu F, 2018, LncRNA PVT1 regulate expression of HIF1alpha via functioning as ceRNA for miR199a5p in nonsmall cell lung cancer under hypoxia. Mol Med Rep 17(1): 1105–1110. , DOI 10.3892/mmr.2017.7962 Lu K-h, Li W, X-h L, Sun M, M-l Z,Wu W-q, Xie W-p, Y-yJBc H, 2013, Long non-coding RNA MEG3 inhibits NSCLC cells proliferation and induces apoptosis by affecting p53 expression. BMC Cancer 13(1):461 NieW, Ge HJ, Yang XQ, Sun X, Huang H, Tao X, ChenWS, Li BJ, 2016, LncRNA-UCA1 exerts oncogenic functions in non-small cell lung cancer by targeting miR-193a-3p. Cancer Lett 371(1): 99–106 Li Z, Hong S, Liu Z, 2018, LncRNA LINC00641 predicts prognosis and inhibits bladder cancer progression through miR-197-3p/ KLF10/PTEN/PI3K/AKT cascade. Biochem Biophys Res Commun 503(3):1825–1829. , DOI 10.1016/j.bbrc.2018. 07.120 WangXB,Wang H, Long HQ, Li DY, ZhengX, 2018, LINC00641 regulates autophagy and intervertebral disc degeneration by acting as a competitive endogenous RNA of miR-153-3p under nutrition deprivation stress. J Cell Physiol. , DOI 10.1002/jcp.27466 Lorenzen JM, Schauerte C,Kolling M, Hubner A, KnappM, Haller H, Thum T, 2015, Long Noncoding RNAs in Urine Are Detectable and May Enable Early Detection of Acute T Cell–Mediated Rejection of Renal Allografts. Clin Chem. , DOI 10.1373/ clinchem.2015.243600 Shi X, Tan H, Le X, Xian H, Li X, Huang K, Luo VY, Liu Y,Wu Z, Mo H, Chen AM, Liang Y, Zhang J, 2018, An expression signature model to predict lung adenocarcinoma-specific survival. Cancer Manag Res 10:3717 ZhouM, Zhang Z, Zhao H, Bao S, Sun J, 2018, A novel lncRNAfocus expression signature for survival prediction in endometrial carcinoma. BMC Cancer 18(1):39 Liu J, Yan J, Zhou C, Ma Q, Jin Q, Yang Z, 2015, miR-1285-3p acts as a potential tumor suppressor miRNA via downregulating JUN expression in hepatocellular carcinoma. Tumour Biol 36(1): 219–225. , DOI 10.1007/s13277-014-2622-5 Li H, Fan J, Yin Z, Wang F, Chen C, Wang DWJO, 2016, Identification of cardiac-related circulating microRNA profile in human chronic heart failure. Oncotarget 7(1):33–45 Keller A, Leidinger P, Gislefoss R, Haugen A, Langseth H, Staehler P, Lenhof H-P, Meese E, 2011, Stable serum miRNA profiles as potential tool for non-invasive lung cancer diagnosis. RNA Biol 8(3):506–516 KirschnerMB, ChengYY, Badrian B, Kao SC, Creaney J, Edelman JJ, Armstrong NJ, Vallely MP, Musk AW, Robinson BW, McCaughan BC, Klebe S, Mutsaers SE, van Zandwijk N, Reid G, 2012, Increased circulating miR-625-3p: a potential biomarker for patients with malignant pleural mesothelioma. J Thorac Oncol 7(7): 1184–1191 Kreth S, Limbeck E, Hinske LC, Schutz SV, Thon N, Hoefig K, Egensperger R,KrethFW(2013, In human glioblastomas transcript elongation by alternative polyadenylation andmiRNA targeting is a potent mechanism of MGMT silencing. Acta Neuropathol 125(5): 671–681 H-s LIU, Y-y MA, H-sJT XIAO, 2012, The diagnostic value of serum microRNAs including miR-129-3p, miR-767-3p and miR- 877* for colorectal cancer. Tumor 32(1):42–48 Cheng WY, Ou Yang TH, Anastassiou D, 2013, Development of a prognostic model for breast cancer survival in an open challenge environment. Sci Transl Med 5(181):181ra150. , DOI 10. 1126/scitranslmed.3005974 Gao D, Cazares LH, Fish EN, 2017, CCL5-CCR5 interactions modulate metabolic events during tumor onset to promote tumorigenesis. BMC Cancer 17(1):834. , DOI 10.1186/s12885- 017-3817-0 Li X, Li B, Ran P,Wang L, 2018, Identification of ceRNA network based on a RNA-seq shows prognostic lncRNA biomarkers in human lung adenocarcinoma. Oncol Lett 16(5):5697–5708. https:// doi.org/10.3892/ol.2018.9336 Ning P,Wu Z, Hu A, Li X, He J, Gong X, Xia Y, Shang Y, Bian H, 2018, Integrated genomic analyses of lung squamous cell carcinoma for identification of a possible competitive endogenous RNA network by means of TCGA datasets. PeerJ 6:e4254. https://doi. org/10.7717/peerj.4254 Sui J, Li YH, Zhang YQ, Li CY, Shen X, Yao WZ, Peng H, Hong WW, Yin LH, Pu YP, Liang GY, 2016, Integrated analysis of long non-coding RNAassociated ceRNA network reveals potential lncRNA biomarkers in human lung adenocarcinoma. Int J Oncol 49(5):2023–2036. , DOI 10.3892/ijo.2016.3716 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1935 EP 1945 DI 10.1007/s12253-019-00780-4 PG 11 ER PT J AU El-Ashmawy, EN Khedr, GE El-Bahrawy, AH Helmy, NN AF El-Ashmawy, E Nahla Khedr, G Eman El-Bahrawy, A Hoda Helmy, N Nada TI Modulatory Effect of Silymarin on Apoptosis in Testosterone -Induced Benign Prostatic Hyperplasia in Rats SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Silymarin; Benign prostatic hyperplasia; Survivin; Bax; Bcl-2; p27/kip1 ID Silymarin; Benign prostatic hyperplasia; Survivin; Bax; Bcl-2; p27/kip1 AB Benign prostatic hyperplasia (BPH) is considered a normal part of the aging process in men, and is characterized by an imbalance between cell proliferation and apoptosis. Our study aimed to investigate the potential protective role of silymarin (SIL) against testosterone-induced BPH in rats and to elucidate the molecular mechanisms underlying SIL pro-apoptotic and anti-proliferative effects. Forty adult male Wistar rats were divided equally into four groups: control group, BPH group (3 mg/kg testosterone propionate, s.c. for 14 days, SIL group (50 mg/kg SIL, orally, once daily concomitantly with 3mg/kg testosterone propionate s.c.) and inhibitor group (50 mg/kg SIL orally concomitantly with 3 mg/kg testosterone, s.c. and 0.5 mg/rat Z-VAD-FMK, i.p.). Silymarin induced caspase-dependent apoptosis in BPH as SIL significantly reduced prostatic Bcl-2 protein and increased Bax protein concentration. Also, SIL down-regulated survivin (Inhibitor of apoptosis protein (IAPs) gene expression in rat prostate assisting mainly caspase-dependent pathway. Silymarin significantly decreased cytochrome-c cytosolic concentration and increased caspase 3 activity compared to BPH group. Silymarin significantly increased the content of p27/kip1 (Cyclin dependent kinase inhibitor (CDKIs) promoting cell cycle arrest. The histological features of BPH such as hypertrophy, papillary projections formation, improved in SIL group. Silymarin showed a significant anti-proliferative and pro-apoptotic role in BPH and accordingly it could be effectively and safely used as a treatment tool in cases of BPH or prostatic disorders. C1 [El-Ashmawy, E Nahla] Tanta University, Faculty of Pharmacy, Department of Biochemistry, 31527 Tanta, El-Gharbia, Egypt. [Khedr, G Eman] Tanta University, Faculty of Pharmacy, Department of Biochemistry, 31527 Tanta, El-Gharbia, Egypt. [El-Bahrawy, A Hoda] Tanta University, Faculty of Pharmacy, Department of Biochemistry, 31527 Tanta, El-Gharbia, Egypt. [Helmy, N Nada] Tanta University, Faculty of Pharmacy, Department of Biochemistry, 31527 Tanta, El-Gharbia, Egypt. RP Helmy, NN (reprint author), Tanta University, Faculty of Pharmacy, Department of Biochemistry, 31527 Tanta, Egypt. EM nada.helmi@pharm.tanta.edu.eg CR Gupta K, Yezdani M, Sotelo T, Aragon-Ching JB, 2015, A synopsis of drugs currently in preclinical and early clinical development for the treatment of benign prostatic hyperplasia. Expert Opin Investig Drugs 24:1059–1073 Na R, Helfand BT, Chen H, Conran CA, Crawford SE, Hayward SW, Tammela TLJ, Hoffman-Bolton J, Zheng SL, Walsh PC, Schleutker J, Platz EA, Isaacs WB, Xu J, 2017, A genetic variant near GATA3 implicated in inherited susceptibility and etiology of benign prostatic hyperplasia, BPH, and lower urinary tract symptoms, LUTS). Prostate 77:1213–1220 Nicholson TM, Ricke WA, 2011, Androgens and estrogens in benign prostatic hyperplasia, past, present and future. Differentiation 82:184–199 Schauer IG, Rowley DR, 2011, The functional role of reactive stroma in benign prostatic hyperplasia. Differentiation 82:200–210 Li F, Pascal LE, Zhou J, ZhouY,Wang K, Parwani AV, Dhir R,Guo P, 2018, BCL-2 and BCL-XL expression are down-regulated in benign prostate hyperplasia nodules and not affected by finasteride and / or celecoxib. Ncbi.Nlm.Nih.Gov, 6):1–10 Poreba M, Strozyk A, Salvesen GS, Drag M, 2013, Caspase substrates and inhibitors. Cold Spring Harb Perspect Biol 5:8 Minutoli L, Rinaldi M, Marini H, Irrera N, Crea G, Lorenzini C, Puzzolo D, Valenti A, Pisani A, Adamo EB, Altavilla D, Squadrito F, Micali A, 2016, Apoptotic pathways linked to endocrine system as potential therapeutic targets for benign prostatic hyperplasia. Int J Mol Sci 17(8):1311 Minutoli L, Altavilla D, Marini H, Rinaldi M, Irrera N, Pizzino G, Bitto A, Arena S, Cimino S, Squadrito F, Russo GI, Morgia G, 2014, Inhibitors of apoptosis proteins in experimental benign prostatic hyperplasia: effects of serenoa repens, selenium and lycopene. J Biomed Sci 21(1):p19 Lin J, Zhou J, XuW, Zhong X, Hong Z, Peng J, 2013, Qianliening capsule treats benign prostatic hyperplasia via suppression of the EGF/STAT3 signaling pathway. Exp Ther Med 5:1293–1300 Mastron JK, Siveen KS, Sethi G, Bishayee A, 2014, Silymarin and hepatocellular carcinoma: A systematic, comprehensive, and critical review. Anti-Cancer Drugs 26:475–486 Devi KP, Malar DS, Braidy N, Nabavi SM, Nabavi SF, 2017, A mini review on the chemistry and Neuroprotective effects of Silymarin. Curr Drug Targets 18(13):1529–1536 Vaid M, Singh T, Prasad R, Katiyar SK, 2015, Silymarin inhibits melanoma cell growth both in vitro and in vivo by targeting cell cycle regulators, angiogenic biomarkers and induction of apoptosis. Mol Carcinog 54:1328–1339 Surai P, 2015, Silymarin as a natural antioxidant: an overview of the current evidence and perspectives. Antioxidants 4:204–247 Fan L, Ma Y, Liu Y, Zheng D, Huang G, 2014, Silymarin induces cell cycle arrest and apoptosis in ovarian cancer cells. Eur J Pharmacol 743:79–88 Cordaro M, Impellizzeri D, Siracusa R, Gugliandolo E, Fusco R, Inferrera A, Esposito E, Di Paola R, Cuzzocre S, 2017, Effects of a co-micronized composite containing palmitoylethanolamide and polydatin in an experimental model of benign prostatic hyperplasia. Toxicol Appl Pharmacol 329:231–240 Atawia RT, Tadros MG, Khalifa AE, Mosli HA, Abdel-Naim AB, 2013, Role of the phytoestrogenic, pro-apoptotic and antioxidative properties of silymarin in inhibiting experimental benign prostatic hyperplasia in rats. Toxicol Lett 219:160–169 Yasuda T, Takeyama Y, Ueda T, Shinzeki M, Kishi S, Sawa H, Nakajima T, Kuroda Y, 2007, Protective effect of Caspase inhibitor on intestinal integrity in experimental severe acute pancreatitis. J Surg Res 138:300–307 Flecknell P, 2016, Laboratory Animal Anaesthesia. 4th edition. Academic Press:, Appendix 3, 265-268 Pfaffl MW, 2004, Quantification strategies in real-time PCR. In: Bustin SA, ed, A-Z of quantification PCR. International University Line, La Jolla, pp 87–112 Lin X, Wu M, Liu B, Wang J, Guan G, Ma A, Zhang Y, 2015, Candesartan ameliorates acutemyocardial infarction in rats through inducible nitric oxide synthase, nuclear factor-B, monocyte chemoattractant protein-1, activator protein-1 and restoration of heat shock protein 72. Mol Med Rep 12(6):8193–8200 Bancroft JD, Layton C, Bancroft JD, 2013, The hematoxylin and eosin, connective and mesenchymal tissues with their stains. Bancroft’s theory and practice of histological techniques, 7th edn. Churchill Livingstone, Philadelphia, pp 173–212 Field A, 2009, Discovering statistics using SPSS Shoieb SM, Esmat A, Khalifa AE, Abdel-NaimAB, 2018, Chrysin attenuates testosterone-induced benign prostate hyperplasia in rats. Food Chem Toxicol 111:650–659 Mosli HH, Esmat A, Atawia RT, Shoieb SM, Mosli HA, Abdel- Naim AB, 2015, Metformin attenuates testosterone-induced prostatic hyperplasia in rats: a pharmacological perspective. Sci Rep 5: 15639 Atawia RT, Mosli HH, TadrosMG, Khalifa AE,Mosli HA, Abdel- Naim AB, 2014, Modulatory effect of silymarin on inflammatory mediators in experimentally induced benign prostatic hyperplasia: Emphasis on PTEN,HIF-1α, and NF-κB, Naunyn. Schmiedebergs. Arch Pharmacol 387:1131–1140 Delbridge ARD, Strasser A, 2015, The BCL-2 protein family, BH3-mimetics and cancer therapy. Cell Death Differ 22:1071– 1080 Czabotar PE, Lessene G, Strasser A, Adams JM, 2014, Control of apoptosis by the BCL - 2 protein family: implications for physiology and therapy. Nat Publ Gr 15:49–63 Zhang X, Bi L, Ye Y, Chen J, 2014, Formononetin Induces Apoptosis in PC-3 Prostate Cancer Cells Through Enhancing the Bax/Bcl-2 Ratios and Regulating the p38/Akt Pathway. Nutr Cancer 66:656–661 Brentnall M, Rodriguez-Menocal L, De Guevara R, Cepero E, Boise LH, 2013, Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol 14:p32 Altavilla D, Minutoli L, Polito F, Irrera N, Arena S, Magno C, Rinaldi M, Burnett BP, Squadrito F, Bitto A, 2012, Effects of flavocoxid, a dual inhibitor of COX and 5-lipoxygenase enzymes, on benign prostatic hyperplasia. Br J Pharmacol 167:95–108 Sayed RH, Saad MA, El-Sahar AE, 2016, Dapoxetine attenuates testosterone-induced prostatic hyperplasia in rats by the regulation of inflammatory and apoptotic proteins. Toxicol Appl Pharmacol 311:52–60 Ma D, Yang B, Zhou Z, Pan D, Zhang X, 2014, Correlation between molecular biomarkers and risk factors for the clinical progression of benign prostatic hyperplasia using tissue microarray immunostaining. Chin Med J 127:4031–4035 Li R, Yu J, Wang C, 2017, Silibinin promotes the apoptosis of gastric cancer BGC823 cells through caspase pathway. J BUON 22:1148–1153 Cho BO, So Y, Jin CH, Byun MW, Il Seo K, Ko K, Chun MS, Jeong IY, 2014, Induction of apoptosis by 2,3-dehydrosilybin via a caspase-dependent pathway in human HeLa cells. Biosci Biotechnol Biochem 78:255–262 Saleem M, Qadir MI, Perveen N, Ahmad B, Saleem U, Irshad T, 2013, Inhibitors of apoptotic proteins: New targets for anticancer therapy. Chem Biol Drug Des 82:243–251 Rodriguez-Berriguete G, Fraile B, de Bethencourt FR, Prieto- Folgado A, Bartolome N, Nunez C, Prati B, Martinez-Onsurbe P, Olmedilla G, Paniagua R, Royuela M, 2010, Role of IAPs in prostate cancer progression: Immunohistochemical study in normal and pathological, benign hyperplastic, prostatic intraepithelial neoplasia and cancer, human prostate. BMC Cancer 10(1):p18 Kanwar JR, Kamalapuram SK, Kanwar RK, 2013, Survivin Signaling in Clinical Oncology: A Multifaceted Dragon. Med Res Rev 33:765–789 Li D, Fu J, Du M, Zhang H, Li L, Cen J, Li W, Chen X, Lin Y, Conway EM, Pikarsky E, Wang H, Pan G, Ji Y, Wang H-Y, Hui L, 2016, Hepatocellular carcinoma repression by TNFα-mediated synergistic lethal effect of mitosis defect-induced senescence and cell death sensitization. Hepatology 64:1105–1120 Morgia G, Micali A, Rinaldi M, Irrera N, Marini H, Puzzolo D, Pisani A, Privitera S, Russo GI, Cimino S, Ieni A, Trichilo V, Altavilla D, Squadrito F, Minutoli L, 2017, Survivin and NAIP in human benign prostatic hyperplasia: Protective role of the association of Serenoa repens, lycopene and selenium from the randomized clinical study. Int J Mol Sci 18:680 Feng W, Cai D, Zhang B, Lou G, Zou X, 2015, Combination of HDAC inhibitor TSA and silibinin induces cell cycle arrest and apoptosis by targeting survivin and cyclinB1/Cdk1 in pancreatic cancer cells. Biomed Pharmacother 74:257–264 Lee JJ, Lee JH, Cho WK, Han JH, Ma JY, 2016, Herbal composition of Cinnamomum cassia, Pinus densiflora, Curcuma longa and Glycyrrhiza glabra prevents atherosclerosis by upregulating p27, Kip1, expression. BMC Complement Altern Med 16:253 Jahanafrooz Z, Motameh N, Bakhshandeh B, 2016, Comparative evaluation of silibinin effects on cell cycling and apoptosis in human breast cancer MCF-7 and T47D cell lines. J Waocp Org 17: 2661–2665 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1947 EP 1956 DI 10.1007/s12253-019-00764-4 PG 10 ER PT J AU Baranyi, M Rittler, D Molnar, E Shirasawa, S Jalsovszky, I Varga, KI Hegedus, L Nemeth, A Dank, M Aigner, C Tovari, J Timar, J Hegedus, B Garay, T AF Baranyi, Marcell Rittler, Dominika Molnar, Eszter Shirasawa, Senji Jalsovszky, Istvan Varga, Karoly Imre Hegedus, Luca Nemeth, Afrodite Dank, Magdolna Aigner, Clemens Tovari, Jozsef Timar, Jozsef Hegedus, Balazs Garay, Tamas TI Next Generation Lipophilic Bisphosphonate Shows Antitumor Effect in Colorectal Cancer In Vitro and In Vivo SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lipophilic bisphosphonate; Zoledronic acid; Colorectal cancer; In vitro; In vivo ID Lipophilic bisphosphonate; Zoledronic acid; Colorectal cancer; In vitro; In vivo AB Bisphosphonates, despite proven antitumor effect in vitro in many tumor types, are currently used only for treatment of osteoporosis and bone metastasis. Colorectal cancer is the third most commonly diagnosed type of cancer and lacks targeted therapy for RAS or RAF mutation carrying cases. A new lipophilic bisphosphonate showed promising results in lung cancer models, but their effect on colorectal cancer cells was not investigated excessively. Antitumor effects and impact on RAS-related signalization of zoledronic acid (ZA) and a lipophilic bisphosphonate (BPH1222) were investigated on 7 human colorectal cancer cell lines in vitro and in vivo. Furthermore, mutant KRAS dependent effect of prenylation inhibition was investigated using isogeneic cell lines. Both bisphosphonates reduced cell viability in vitro in a dose-dependent manner. Both compounds changed cell cycle distribution similarly by increasing the proportion of cells either in the S or in the subG1 phase or both. However, BPH1222 exerted higher inhibitory effect on spheroid growth than ZA. Interestingly, we found profound alterations in phosphorylation level of Erk and S6 proteins upon ZA or BPH1222 treatment. Furthermore, investigation of a mutant KRAS isogeneic model system suggests that the drugs interfere also with the mutant KRAS proteins. In vivo experiments with KRAS mutant xenograft model also revealed growth inhibitory potential of bisphosphonate treatment. Our results show that lipophilic bisphosphonates might extend the therapeutic spectrum of bisphosphonate drugs and could be considered as additional treatment approaches in colorectal cancer. C1 [Baranyi, Marcell] Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. [Rittler, Dominika] Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. [Molnar, Eszter] Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. [Shirasawa, Senji] Fukuoka University, Faculty of Medicine, Department of Cell BiologyFukuoka, Japan. [Jalsovszky, Istvan] Eotvos Lorand University, Faculty of Science, Institute of Chemistry, Department of Organic Chemistry, H-1117 Budapest, Hungary. [Varga, Karoly Imre] Eotvos Lorand University, Faculty of Science, Institute of Chemistry, Department of Organic Chemistry, H-1117 Budapest, Hungary. [Hegedus, Luca] University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic Surgery, D-45239 Essen, Germany. [Nemeth, Afrodite] Semmelweis University, Department of Oncology, H-1091 Budapest, Hungary. [Dank, Magdolna] Semmelweis University, Department of Oncology, H-1091 Budapest, Hungary. [Aigner, Clemens] University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic Surgery, D-45239 Essen, Germany. [Tovari, Jozsef] National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, H-1122 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. [Hegedus, Balazs] Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. [Garay, Tamas] Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. RP Hegedus, B (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM balazs.hegedues@rlk.uk-essen.de CR Mcguire S, 2016, World Cancer Report 2014. Geneva, Switzerland: World Health Organization, International Agency for Research on Cancer, WHO Press, 2015. Adv Nutr 7(2):418– 419. , DOI 10.3945/an.116.012211 Fearon ER, 2011, Molecular genetics of colorectal cancer. Annu Rev Pathol 6:479–507. , DOI 10.1146/annurev-pathol- 011110-130235 Lau KS, Haigis KM, 2009, Non-redundancy within the RAS oncogene family: insights into mutational disparities in cancer. Mol Cells 28(4):315–320. , DOI 10.1007/s10059-009-0143-7 Wood LD, Parsons DW, Jones S, Lin J, Sjoblom T, Leary RJ, Shen D, Boca SM, Barber T, Ptak J, Silliman N, Szabo S, Dezso Z, Ustyanksky V, Nikolskaya T, Nikolsky Y, Karchin R, Wilson PA, Kaminker JS, Zhang Z, Croshaw R, Willis J, Dawson D, Shipitsin M, Willson JK, Sukumar S, Polyak K, Park BH, Pethiyagoda CL, Pant PV, Ballinger DG, Sparks AB, Hartigan J, Smith DR, Suh E, Papadopoulos N, Buckhaults P, Markowitz SD, Parmigiani G, Kinzler KW, Velculescu VE, Vogelstein B, 2007, The genomic landscapes of human breast and colorectal cancers. Science 318(5853):1108–1113. , DOI 10.1126/science.1145720 Baldus SE, Schaefer KL, Engers R, Hartleb D, Stoecklein NH, Gabbert HE, 2010, Prevalence and heterogeneity of KRAS, BRAF, and PIK3CA mutations in primary colorectal adenocarcinomas and their corresponding metastases. Clin Cancer Res 16(3):790–799. , DOI 10.1158/1078-0432.CCR-09- 2446 De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, Kalogeras KT, Kotoula V, Papamichael D, Laurent- Puig P, Penault-Llorca F, Rougier P, Vincenzi B, Santini D, Tonini G, Cappuzzo F, Frattini M, Molinari F, Saletti P, De Dosso S, Martini M, Bardelli A, Siena S, Sartore-Bianchi A, Tabernero J, Macarulla T, Di Fiore F, Gangloff AO, Ciardiello F, Pfeiffer P, Qvortrup C, Hansen TP, Van Cutsem E, Piessevaux H, Lambrechts D, Delorenzi M, Tejpar S, 2010, Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol 11(8):753–762. , DOI 10.1016/S1470-2045(10, 70130-3 Cox AD, Fesik SW, Kimmelman AC, Luo J, Der CJ, 2014, Drugging the undruggable RAS: Mission possible? Nat Rev Drug Discov 13(11):828–851. , DOI 10.1038/ nrd4389 Vaughn CP, Zobell SD, Furtado LV, Baker CL, Samowitz WS, 2011, Frequency of KRAS, BRAF, and NRAS mutations in colorectal cancer. Genes Chromosomes Cancer 50(5):307–312. https:// doi.org/10.1002/gcc.20854 Timar J, Hegedus B, Raso E, 2010, KRAS mutation testing of colorectal cancer for anti-EGFR therapy: dogmas versus evidence. Curr Cancer Drug Targets 10(8):813–823. , DOI 10.2174/ 156800910793357989 Cohen R, Cervera P, Svrcek M, Pellat A, Dreyer C, de Gramont A, Andre T, 2017, BRAF-mutated colorectal cancer: what is the optimal strategy for treatment? Curr Treat Options in Oncol 18(2):9. , DOI 10.1007/s11864-017-0453-5 Sundar R, Hong DS, Kopetz S, Yap TA, 2017, Targeting BRAFmutant colorectal cancer: progress in combination strategies. Cancer Discov 7(6):558–560. , DOI 10.1158/2159-8290. CD-17-0087 Kopetz S, Desai J, Chan E, Hecht JR, O'Dwyer PJ,Maru D, Morris V, Janku F, Dasari A, ChungW, Issa JP, Gibbs P, James B, Powis G, Nolop KB, Bhattacharya S, Saltz L, 2015, Phase II pilot study of Vemurafenib in patients with metastatic BRAF-mutated colorectal cancer. J Clin Oncol 33(34):4032–4038. , DOI 10.1200/ JCO.2015.63.2497 van Beek E, Pieterman E, Cohen L, Lowik C, Papapoulos S, 1999, Farnesyl pyrophosphate synthase is the molecular target of nitrogen-containing bisphosphonates. Biochem Biophys Res Commun 264(1):108–111. , DOI 10.1006/bbrc.1999.1499 Wang M, Casey PJ, 2016, Protein prenylation: unique fats make their mark on biology. Nat Rev Mol Cell Biol 17(2):110–122. , DOI 10.1038/nrm.2015.11 Forsea AM, Muller C, Riebeling C, Orfanos CE, Geilen CC, 2004, Nitrogen-containing bisphosphonates inhibit cell cycle progression in human melanoma cells. Br J Cancer 91(4):803–810. https://doi. org/10.1038/sj.bjc.6602052 Garay T, Kenessey I, Molnar E, Juhasz E, Reti A, Laszlo V, Rozsas A, Dobos J, Dome B, Berger W, Klepetko W, Tovari J, Timar J, Hegedus B, 2015, Prenylation inhibition-induced cell death in melanoma: reduced sensitivity in BRAF mutant/PTEN wild-type melanoma cells. PLoS One 10(2):e0117021. , DOI 10.1371/ journal.pone.0117021 Fragni M, Bonini SA, Bettinsoli P, Bodei S, Generali D, Bottini A, Spano PF, Memo M, Sigala S, 2016, The miR-21/PTEN/Akt signaling pathway is involved in the anti-tumoral effects of zoledronic acid in human breast cancer cell lines. Naunyn Schmiedeberg's Arch Pharmacol 389(5):529–538. , DOI 10.1007/s00210- 016-1224-8 Ibrahim T, Mercatali L, Sacanna E, Tesei A, Carloni S, Ulivi P, Liverani C, Fabbri F, Zanoni M, Zoli W, Amadori D, 2012, Inhibition of breast cancer cell proliferation in repeated and nonrepeated treatment with zoledronic acid. Cancer Cell Int 12(1):48. , DOI 10.1186/1475-2867-12-48 Mansouri M, Mirzaei SA, Lage H, Mousavi SS, Elahian F, 2014, The cell cycle arrest and the anti-invasive effects of nitrogencontaining bisphosphonates are not mediated by DBF4 in breast cancer cells. Biochimie 99:71–76. , DOI 10.1016/j.biochi. 2013.11.010 Senaratne SG, Pirianov G, Mansi JL, Arnett TR, Colston KW, 2000, Bisphosphonates induce apoptosis in human breast cancer cell lines. Br J Cancer 82(8):1459–1468. , DOI 10.1054/ bjoc.1999.1131 Stachnik A,Yuen T, Iqbal J, SgobbaM, Gupta Y, Lu P, Colaianni G, Ji Y, Zhu LL, Kim SM, Li J, Liu P, Izadmehr S, Sangodkar J, Scherer T, Mujtaba S, Galsky M, Gomez J, Epstein S, Buettner C, Bian Z, Zallone A, Aggarwal AK, Haider S, NewMI, Sun L, Narla G, ZaidiM(2014, Repurposing of bisphosphonates for the prevention and therapy of nonsmall cell lung and breast cancer. Proc Natl Acad Sci U S A 111(50):17995–18000. , DOI 10.1073/ pnas.1421422111 Di Salvatore M, Orlandi A, Bagala C, Quirino M, Cassano A, Astone A, Barone C, 2011, Anti-tumour and anti-angiogenetic effects of zoledronic acid on human non-small-cell lung cancer cell line. Cell Prolif 44(2):139–146. , DOI 10.1111/j.1365- 2184.2011.00745.x Tassone P, Tagliaferri P, Viscomi C, Palmieri C, Caraglia M, D'Alessandro A, Galea E, Goel A, Abbruzzese A, Boland CR, Venuta S, 2003, Zoledronic acid induces antiproliferative and apoptotic effects in human pancreatic cancer cells in vitro. Br J Cancer 88(12):1971–1978. , DOI 10.1038/sj.bjc.6600986 Tamura T, Shomori K, Nakabayashi M, Fujii N, Ryoke K, Ito H, 2011, Zoledronic acid, a third-generation bisphosphonate, inhibits cellular growth and induces apoptosis in oral carcinoma cell lines. Oncol Rep 25(4):1139–1143. , DOI 10.3892/or.2011.1152 Michailidou M, Brown HK, Lefley DV, Evans A, Cross SS, Coleman RE, Brown NJ, Holen I, 2010, Microvascular endothelial cell responses in vitro and in vivo: modulation by zoledronic acid and paclitaxel? J Vasc Res 47(6):481–493. , DOI 10.1159/ 000313876 Gao X, Jiang B, Zou S, Zhang T, Qi X, Jin L, Ge X, Tang SC, Hua D, Chen W, 2015, Zoledronate can promote apoptosis and inhibit the proliferation of colorectal cancer cells. Tumour Biology : the Journal of the International Society for Oncodevelopmental Biology and Medicine 36(7):5315–5322. , DOI 10.1007/ s13277-015-3192-x Sewing L, Steinberg F, Schmidt H, Goke R, 2008, The bisphosphonate zoledronic acid inhibits the growth of HCT-116 colon carcinoma cells and induces tumor cell apoptosis. Apoptosis 13(6): 782–789. , DOI 10.1007/s10495-008-0211-z Kuroda J, Kimura S, Segawa H, Sato K,Matsumoto S, Nogawa M, Yuasa T, Kobayashi Y, Yoshikawa T, Ottmann OG, Maekawa T, 2004, p53-independent anti-tumor effects of the nitrogencontaining bisphosphonate zoledronic acid. Cancer Sci 95(2): 186–192. , DOI 10.1111/j.1349-7006.2004.tb03202.x Kato J, Futamura M, Kanematsu M, Gaowa S, Mori R, Tanahashi T, Matsuhashi N, Yoshida K, 2016, Combination therapy with zoledronic acid and cetuximab effectively suppresses growth of colorectal cancer cells regardless of KRAS status. Int J Cancer 138(6): 1516–1527. , DOI 10.1002/ijc.29881 Bonovas S, Nikolopoulos G, Bagos P, 2013, Bisphosphonate use and risk of colorectal cancer: a systematic review and meta-analysis. Br J Clin Pharmacol 76(3):329–337. , DOI 10.1111/ bcp.12135 Rennert G, Pinchev M, Rennert HS, Gruber SB, 2011, Use of bisphosphonates and reduced risk of colorectal Cancer. J Clin Oncol 29(9):1146–1150. , DOI 10.1200/JCO.2010.33.7485 Thosani N, Thosani SN, Kumar S, Nugent Z, Jimenez C, Singh H, Guha S, 2013, Reduced risk of colorectal cancer with use of oral bisphosphonates: a systematic review and meta-analysis. J Clin Oncol 31(5):623–630. , DOI 10.1200/JCO.2012.42.9530 Yang G, Hu H, Zeng R, Huang J, 2013, Oral bisphosphonates and the risk of colorectal cancer: a meta-analysis. J Clin Gastroenterol 47(9):741–748. , DOI 10.1097/MCG.0b013e31829e446b Morgan GJ, Davies FE, Gregory WM, Cocks K, Bell SE, Szubert AJ, Navarro-Coy N, Drayson MT, Owen RG, Feyler S, Ashcroft AJ, Ross F, Byrne J, Roddie H, Rudin C, Cook G, Jackson GH, Child JA, 2010, First-line treatment with zoledronic acid as compared with clodronic acid in multiple myeloma, MRC myeloma IX): a randomised controlled trial. Lancet 376(9757):1989–1999. , DOI 10.1016/S0140-6736(10)62051-X James ND, SydesMR, ClarkeNW,Mason MD, Dearnaley DP, Spears MR, Ritchie AWS, Parker CC, Russell JM, Attard G, de Bono J, Cross W, Jones RJ, Thalmann G, Amos C, Matheson D, Millman R, Alzouebi M, Beesley S, Birtle AJ, Brock S, Cathomas R, Chakraborti P, Chowdhury S, Cook A, Elliott T, Gale J, Gibbs S, Graham JD, Hetherington J, Hughes R, Laing R, McKinna F, McLaren DB, O'Sullivan JM, Parikh O, Peedell C, Protheroe A, Robinson AJ, Srihari N, Srinivasan R, Staffurth J, Sundar S, Tolan S, Tsang D, Wagstaff J, Parmar MKB, 2016, Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer, STAMPEDE): survival results from an adaptive, multiarm,multistage, platform randomised controlled trial. Lancet 387(10024):1163–1177. , DOI 10.1016/S0140-6736(15)01037-5 Coleman R, Cameron D, Dodwell D, Bell R, Wilson C, Rathbone E,KeaneM,Gil M, Burkinshaw R,Grieve R, Barrett-Lee P, Ritchie D, Liversedge V, Hinsley S, Marshall H, 2014, Adjuvant zoledronic acid in patients with early breast cancer: final efficacy analysis of the AZURE, BIG 01/04, randomised open-label phase 3 trial. Lancet Oncol 15(9):997–1006. , DOI 10.1016/S1470- 2045(14)70302-X Rakel A, Boucher A, Ste-Marie LG, 2011, Role of zoledronic acid in the prevention and treatment of osteoporosis. Clin Interv Aging 6:89–99. , DOI 10.2147/CIA.S7282 Weiss HM, Pfaar U, Schweitzer A, Wiegand H, Skerjanec A, Schran H, 2008, Biodistribution and plasma protein binding of zoledronic acid. Drug Metab Dispos 36(10):2043–2049. https:// doi.org/10.1124/dmd.108.021071 Xia Y, Liu YL, Xie Y, ZhuW, Guerra F, Shen S, Yeddula N, Fischer W, Low W, Zhou X, Zhang Y, Oldfield E, Verma IM, 2014, A combination therapy for KRAS-driven lung adenocarcinomas using lipophilic bisphosphonates and rapamycin. Sci Transl Med 6(263):263ra161. , DOI 10.1126/scitranslmed.3010382 Zhang Y, Cao R, Yin F, Hudock MP, Guo RT, Krysiak K, Mukherjee S, Gao YG, Robinson H, Song Y, No JH, Bergan K, Leon A, Cass L, Goddard A, Chang TK, Lin FY, Van Beek E, Papapoulos S, Wang AH, Kubo T, Ochi M, Mukkamala D, Oldfield E, 2009, Lipophilic bisphosphonates as dual farnesyl/ geranylgeranyl diphosphate synthase inhibitors: an X-ray and NMR investigation. J Am Chem Soc 131(14):5153–5162. https:// doi.org/10.1021/ja808285e Nath S, Devi GR, 2016, Three-dimensional culture systems in cancer research: focus on tumor spheroid model. Pharmacol Ther 163: 94–108. , DOI 10.1016/j.pharmthera.2016.03.013 Hack S, Worlein B, Hofner G, Pabel J, Wanner KT, 2011, Development of imidazole alkanoic acids as mGAT3 selective GABA uptake inhibitors. Eur J Med Chem 46(5):1483–1498. , DOI 10.1016/j.ejmech.2011.01.042 ZhangY, ZhuW, Liu YL,Wang H,Wang K, Li K, No JH,Ayong L, Gulati A, Pang R, Freitas-Junior L, Morita CT, Old-Field E, 2013, Chemo-immunotherapeutic anti-malarials targeting isoprenoid biosynthesis. ACS Med Chem Lett 4(4):423–427. , DOI 10. 1021/ml4000436 Shirasawa S, Furuse M, Yokoyama N, Sasazuki T, 1993, Altered growth of human colon cancer cell lines disrupted at activated Kiras. Science 260(5104):85–88 Mouradov D, Sloggett C, Jorissen RN, Love CG, Li S, Burgess AW, Arango D, Strausberg RL, Buchanan D, Wormald S, O'Connor L, Wilding JL, Bicknell D, Tomlinson IP, Bodmer WF, Mariadason JM, Sieber OM, 2014, Colorectal cancer cell lines are representative models of the main molecular subtypes of primary cancer. Cancer Res 74(12):3238–3247. , DOI 10.1158/ 0008-5472.CAN-14-0013 Klijn C, Durinck S, Stawiski EW, Haverty PM, Jiang Z, Liu H, Degenhardt J, Mayba O, Gnad F, Liu J, Pau G, Reeder J, Cao Y, Mukhyala K, Selvaraj SK, Yu M, Zynda GJ, Brauer MJ, Wu TD, Gentleman RC, Manning G, Yauch RL, Bourgon R, Stokoe D, Modrusan Z, Neve RM, de Sauvage FJ, Settleman J, Seshagiri S, Zhang Z, 2015, A comprehensive transcriptional portrait of human cancer cell lines. Nat Biotechnol 33(3):306–312. , DOI 10. 1038/nbt.3080 Hegedus L, Padanyi R, Molnar J, Paszty K, Varga K, Kenessey I, Sarkozy E, Wolf M, Grusch M, Hegyi Z, Homolya L, Aigner C, Garay T, Hegedus B, Timar J, Kallay E, Enyedi A, 2017, Histone Deacetylase inhibitor treatment increases the expression of the plasma membrane Ca2+ pump PMCA4b and inhibits the migration of melanoma cells independent of ERK. Front Oncol 7:95. https://doi. org/10.3389/fonc.2017.00095 Ivanov DP, Parker TL, Walker DA, Alexander C, Ashford MB, Gellert PR, Garnett MC, 2014, Multiplexing spheroid volume, resazurin and acid phosphatase viability assays for highthroughput screening of tumour spheroids and stem cell neurospheres. PLoS One 9(8):e103817. , DOI 10.1371/ journal.pone.0103817 Kenessey I, Koi K, Horvath O, Cserepes M, Molnar D, Izsak V, Dobos J, Hegedus B, Tovari J, Timar J, 2016, KRAS-mutation status dependent effect of zoledronic acid in human non-small cell cancer preclinical models. Oncotarget 7(48):79503–79514. https:// doi.org/10.18632/oncotarget.12806 Kelsey I, Manning BD, 2013, mTORC1 status dictates tumor response to targeted therapeutics. Sci Signal 6(294):pe31. https://doi. org/10.1126/scisignal.2004632 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1957 EP 1969 DI 10.1007/s12253-019-00789-9 PG 13 ER PT J AU Rojas, EEI Segura-Azuara, dlN Cabrera, VLM AF Rojas, Elisa Erana Irma Segura-Azuara, de los Angeles Nancy Cabrera, Vanessa Lopez Mildred TI Breaking the Curse: Opening Students’ Eyes to Pathology and Oncology Research SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Educational innovation; Educational strategy; Academic medicine; Research ID Educational innovation; Educational strategy; Academic medicine; Research AB Research is a particularly underexplored professional activity for physicians, leaving many medical students compelled to pursue a clinical career. However, the XXI century requires that physicians innovate and perform research that fuels their practice; students should have early exposure to research to explore it as a career path. It should be encouraged in the undergraduate program by having students take part in case reports, short communication presentations, and research seminars. As part of an educational strategy, students worked with faculty members to deliver a gynecologic oncology pathology case report as a poster for the Oncology Conference of Medical Students. We used a quantitative approach with a descriptive and cross-sectional design to assess the effect of poster presentations on developing student’s research skills. The sample comprised 118 medical students enrolled in the Pathology courses that presented a total of 23 posters. The judges who assessed had a medical specialty in Pathology, Radio-oncology, and Gynecologic Oncology. The results show that students exceeded expectations on the use of language, as it was both formal and technical; and they used relevant bibliographic support and references. However, students performed less well in the clinical case summary. The judges found that although it was coherent and chronologically ordered, they did not include all the relevant laboratory tests and analyses, nor a full description of the diagnosis. This educational strategy has proven to be valuable in promoting Pathology and Oncology Research in students; it allowed participants to adopt a systematic approach and methodologies to document, analyze, and share knowledge. C1 [Rojas, Elisa Erana Irma] Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prierto 3000 Pte col. Los Doctores, 64710 Monterrey, Mexico. [Segura-Azuara, de los Angeles Nancy] Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prierto 3000 Pte col. Los Doctores, 64710 Monterrey, Mexico. [Cabrera, Vanessa Lopez Mildred] Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prierto 3000 Pte col. Los Doctores, 64710 Monterrey, Mexico. RP Cabrera, VLM (reprint author), Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, 64710 Monterrey, Mexico. EM mildredlopez@tec.mx CR Aguilar-Vargas E, Aviles-Ibarra OJ, Mendez N, 2017, Elaboracion de un articulo de reporte de caso clinico o de revision, en la materia demetodologia de la investigacion enmedicina. An Fac Med 78(1): 55–59. , DOI 10.15381/anales.v78i1.13022 Castro CA, 2014, La investigacion formativa en estudiantes de pregrado de medicina: Analisis de la propuesta pedagogica para la formacion de investigadores [Dissertation]. Universidad Sergio Arboleda, Bogota, CO, pp 1–150 Patil SS, Hasamnis AA, 2018, The role and scope of research in undergraduate medical curriculum. J Clin Diagnostic Res 12(10): LI01–LI02 Wayment HA, Dickson KL, 2008, Increasing student participation in undergraduate research benefits students, faculty, and department. Teach Psychol 35(3):194–197 Mayta-Tristan P, Cartagena-Klein R, Pereyra-Elias R, Portillo A, Rodriguez-Morales AJ, 2013, Apreciacion de estudiantes de Medicina latinoamericanos sobre la capacitacion universitaria en investigacion cientifica. Rev Med Chil 141(6):716–722 Buch AC, Iqbal MB, 2016, Poster presentation at medical conferences: points to ponder. Med J Dr DY Patil Univ 9(2):224 Mayta-Tristan P, Mezones-Holguin E, Carbajal-Gonzalez D, Pereyra-Elias, Montenegro-Idrogo JJ, Mejia C, Munoz S et al, 2015, Motivaciones-Paraestudiar-Medicina. iMedPub J 11(3):1–7 Amgad M, TsuiMMK, Liptrott SJ, Shash E, 2015)Medical student research: an integratedmixed-methods systematic review and metaanalysis. PLoS One [Internet] 10(6):1–32. Available from. https:// doi.org/10.1371/journal.pone.0127470 Arbelaez Lopez R, Hernandez Baron NO, Perez Angulo MI, Angulo MIP, 2007, El desarrollo de la competencia investigadora en los estudiantes de pregrado. Revista Docencia Universitaria 7(1):176 Magriet Dhaese SA, Van de Caveye I, Vanden BP, Bogaert S, De Maeseneer J, 2015, Student participation: to the benefit of both the student and the faculty. Educ Heal Chang Learn Pract 28(1):79–82 Goodhand JR, Giles CL, Wahed M, Irving PM, Langmead L, Rampton DS, 2011, Poster presentations at medical conferences: an effective way of disseminating research? Clin Med, Lond, 11(2): 138–141 Salzl G, Golder S, Timmer A, Marienhagen J, Scholmerich J, Grossmann J, 2008 Feb, Poster exhibitions at national conferences: education or farce? Dtsch Arztebl Int 105(5):78–83 Ilic D, Rowe N, 2013, What is the evidence that poster presentations are effective in promoting knowledge transfer? A state of the art review. Health Inf Libr J 30(1):4–12 TecLabs, editor. Memorias CIIE 2017. Integracion del reporte de caso en pregrado: diseno de instrumento de evaluacion de carteles. Proceedings of the 4 Congreso Internacional de Innovacion Educativa; 2017 Dic 11–13; Monterrey. Monterrey: Tecnologico de Monterrey: 2017 TecLabs, editor. Memorias CIIE 2017. Impulso de Publicacion de Reportes de Caso en Congreso. Proceedings of the 4 Congreso Internacional de Innovacion Educativa; 2017 Dic 11–13; Monterrey. Monterrey: Tecnologico de Monterrey: 2017 McMillan JH, Schumacher S, 2005, Investigacion educativa: Una introduccion conceptual. Pearson Addison Wesley, Madrid NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1971 EP 1974 DI 10.1007/s12253-019-00728-8 PG 4 ER PT J AU Malgulwar, BP Nambirajan, A Singh, M Suri, V Sarkar, Ch Sharma, ChM AF Malgulwar, Benny Prit Nambirajan, Aruna Singh, Manmohan Suri, Vaishali Sarkar, Chitra Sharma, Chand Mehar TI Expression and Clinical Significance of Translation Regulatory Long Non-Coding RNA 1 (TRERNA1) in Ependymomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ependymoma; TRERNA1; lncRNA; Epithelial to mesenchymal transition ID Ependymoma; TRERNA1; lncRNA; Epithelial to mesenchymal transition AB Long noncoding RNAs (lncRNA) have emerged as vital molecules governing epithelial-to-mesenchymal transition (EMT) in cancers. Translation regulatory RNA 1 (TRERNA1) is one such lncRNA known to enhance the transcriptional activity of the EMT-transcription factor, Snail. We have previously demonstrated differential upregulation of EMT-transcription factors and cadherin switching across various clinico-pathologic-molecular subclasses of ependymomas (EPN). With an aim to analyze the correlation between the expression of TRERNA1 in EPNs, we performed gene expression analysis for TRERNA1 on 75 Grade II/ III EPNs and correlated with tumor site, C11orf95-RELA fusions, age, MIB-1 proliferative indices, and outcome wherever available. Upregulation of gene expression levels of TRERNA1 was seen in intracranial EPNs, with highest expression levels in pediatric posterior fossa EPNs. High TRERNA1 expression was found associated with higher proliferative indices (p = 0.034) and shorter progression free survival (p = 0.002). Our study, for the first time, demonstrates an association between TRERNA1 expressions and pediatric posterior fossa EPNs. Further in-vivo and in-vitro studies are required to confirm these findings and evaluate TRERNA1 as a novel biomarker and potential therapeutic target in childhood PF-EPNs. C1 [Malgulwar, Benny Prit] All India Institute of Medical Sciences, Department of Pathology, Ansari Nagar, 110029 New Delhi, India. [Nambirajan, Aruna] All India Institute of Medical Sciences, Department of Pathology, Ansari Nagar, 110029 New Delhi, India. [Singh, Manmohan] All India Institute of Medical Sciences, Department of Neurosurgery, 110029 New Delhi, India. [Suri, Vaishali] All India Institute of Medical Sciences, Department of Pathology, Ansari Nagar, 110029 New Delhi, India. [Sarkar, Chitra] All India Institute of Medical Sciences, Department of Pathology, Ansari Nagar, 110029 New Delhi, India. [Sharma, Chand Mehar] All India Institute of Medical Sciences, Department of Pathology, Ansari Nagar, 110029 New Delhi, India. RP Sharma, ChM (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM sharmamehar@yahoo.co.in CR Kilday JP, Rahman R, Dyer S, Ridley L, Lowe J, Coyle B, Grundy R, 2009, Pediatric ependymoma: biological perspectives. Mol Cancer Res 7(6):765–786 Ramaswamy V, Hielscher T, Mack SC, Lassaletta A, Lin T, Pajtler KW, Jones DTW, Luu B, Cavalli FMG, Aldape K, Remke M, Mynarek M, Rutkowski S, Gururangan S, McLendon RE, Lipp ES, Dunham C, Hukin J, Eisenstat DD, Fulton D, van Landeghem FKH, Santi M, van Veelen MLC, van Meir EG, Osuka S, Fan X, Muraszko KM, Tirapelli DPC, Oba-Shinjo SM, Marie SKN, Carlotti CG, Lee JY, Rao AAN, Giannini C, Faria CC, Nunes S, Mora J, Hamilton RL, Hauser P, Jabado N, Petrecca K, Jung S, Massimi L, Zollo M, Cinalli G, Bognar L, Klekner A, Hortobagyi T, Leary S, Ermoian RP, Olson JM, Leonard JR, Gardner C, Grajkowska WA, Chambless LB, Cain J, Eberhart CG, Ahsan S, Massimino M, Giangaspero F, Buttarelli FR, Packer RJ, Emery L, Yong WH, Soto H, Liau LM, Everson R, Grossbach A, Shalaby T, Grotzer M, Karajannis MA, Zagzag D, Wheeler H, von Hoff K, Alonso MM, Tunon T, Schuller U, Zitterbart K, Sterba J, Chan JA, Guzman M, Elbabaa SK, Colman H, Dhall G, Fisher PG, Fouladi M, Gajjar A, Goldman S, Hwang E, Kool M, Ladha H, Vera- Bolanos E, Wani K, Lieberman F, Mikkelsen T, Omuro AM, Pollack IF, Prados M, Robins HI, Soffietti R, Wu J, Metellus P, Tabori U, Bartels U, Bouffet E, Hawkins CE, Rutka JT, Dirks P, Pfister SM, Merchant TE, Gilbert MR, Armstrong TS, Korshunov A, Ellison DW, Taylor MD, 2016, Therapeutic impact of Cytoreductive surgery and irradiation of posterior Fossa Ependymoma in the molecular era: a retrospective multicohort analysis. J Clin Oncol 34:2468–2477 Zamecnik J, SnuderlM, Eckschlager T, Chanova M, Hladikova M, Tichy M, Kodet R, 2003, Paediatric intracranial ependymomas: prognostic relevance of histological, immunohistochemical, and flow cytometric factors. Mod Pathol 16:980–991 Gupta RK, Sharma MC, Suri V, Kakkar A, Singh M, Sarkar C, 2014, Study of chromosome 9q gain, notch pathway regulators and Tenascin-C in ependymomas. J Neuro-Oncol 116:267–274 Nambirajan A, Sharma MC, Gupta RK, Suri V, Singh M, Sarkar C, 2014, Study of stem cell marker nestin and its correlation with vascular endothelial growth factor and microvascular density in ependymomas. Neuropathol Appl Neurobiol 40:714–725 Rajeshwari M, Sharma MC, Kakkar A, Nambirajan A, Suri V, Sarkar C, Singh M, Saran RK, Gupta RK, 2016, Evaluation of chromosome 1q gain in intracranial ependymomas. J Neuro- Oncol 127:271–278 Pajtler KW, Witt H, Sill M, Jones DT, Hovestadt V, Kratochwil F, Wani K, Tatevossian R, Punchihewa C, Johann P, Reimand J, Warnatz HJ, Ryzhova M, Mack S, Ramaswamy V, Capper D, Schweizer L, Sieber L, Wittmann A, Huang Z, van Sluis P, Volckmann R, Koster J, Versteeg R, Fults D, Toledano H, Avigad S, Hoffman LM, Donson AM, Foreman N, Hewer E, Zitterbart K, Gilbert M, Armstrong TS, Gupta N, Allen JC, Karajannis MA, Zagzag D, Hasselblatt M, Kulozik AE, Witt O, Collins VP, von Hoff K, Rutkowski S, Pietsch T, Bader G, Yaspo ML, von Deimling A, Lichter P, Taylor MD, Gilbertson R, Ellison DW, Aldape K, Korshunov A, Kool M, Pfister SM, 2015, Molecular classification of ependymal tumors across all CNS compartments, histopathological grades, and age groups. Cancer Cell 27:728–743 Mack SC,Witt H, Piro RM, Gu L, Zuyderduyn S, Stutz AM,Wang X, GalloM, Garzia L, Zayne K, Zhang X, Ramaswamy V, Jager N, Jones DTW, Sill M, Pugh TJ, Ryzhova M, Wani KM, Shih DJH, Head R, Remke M, Bailey SD, Zichner T, Faria CC, Barszczyk M, Stark S, Seker-Cin H, Hutter S, Johann P, Bender S, Hovestadt V, Tzaridis T, Dubuc AM, Northcott PA, Peacock J, Bertrand KC, Agnihotri S, Cavalli FMG, Clarke I, Nethery-Brokx K, Creasy CL, Verma SK, Koster J, Wu X, Yao Y, Milde T, Sin-Chan P, Zuccaro J, Lau L, Pereira S, Castelo-Branco P, Hirst M, Marra MA, Roberts SS, Fults D, Massimi L, Cho YJ, van Meter T, Grajkowska W, Lach B, Kulozik AE, von Deimling A, Witt O, Scherer SW, Fan X, Muraszko KM, Kool M, Pomeroy SL, Gupta N, Phillips J, Huang A, Tabori U, Hawkins C, Malkin D, Kongkham PN, Weiss WA, Jabado N, Rutka JT, Bouffet E, Korbel JO, Lupien M, Aldape KD, Bader GD, Eils R, Lichter P, Dirks PB, Pfister SM, Korshunov A, Taylor MD, 2014, Epigenomic alterations define lethal CIMP-positive ependymomas of infancy. Nature 506:445–450 Malgulwar PB, Nambirajan A, Pathak P, Faruq M, Rajeshwari M, Singh M, Suri V, Sarkar C, Sharma MC, 2018, C11orf95-RELA fusions and upregulated NF-KB signaling characterise a subset of aggressive supratentorial ependymomas that express L1CAMand nestin. J Neuro-Oncol 138:29–39 PajtlerKW,Mack SC, Ramaswamy V, Smith CA,Witt H, Smith A, Hansford JR, von Hoff K, Wright KD, Hwang E, Frappaz D, Kanemura Y, Massimino M, Faure-Conter C, Modena P, Tabori U, Warren KE, Holland EC, Ichimura K, Giangaspero F, Castel D, von Deimling A, Kool M, Dirks PB, Grundy RG, Foreman NK, Gajjar A, Korshunov A, Finlay J, Gilbertson RJ, Ellison DW, Aldape KD, Merchant TE, Bouffet E, Pfister SM, Taylor MD, 2017, The current consensus on the clinical management of intracranial ependymoma and its distinct molecular variants. Acta Neuropathol 133:5–12 Malgulwar PB, Sharma V, Tomar AS, Verma C, Nambirajan A, Singh M, Suri V, Sarkar C, Sharma MC, 2018, Transcriptional co-expression regulatory network analysis for snail and slug identifies IL1R1, an inflammatory cytokine receptor, to be preferentially expressed in ST-EPN-RELA and PF-EPN-A molecular subgroups of intracranial ependymomas. Oncotarget 9:35480–35492 Thiery JP, Acloque H, Huang RY et al, 2009, Epithelialmesenchymal transitions in development and disease. Cell 139: 871–890 Kahlert UD, Nikkhah G, Maciaczyk J, 2013, Epithelial-tomesenchymal(− like, transition as a relevant molecular event in malignant gliomas. Cancer Lett 331:131–138 Malgulwar PB, Nambirajan A, Pathak P, Rajeshwari M, Suri V, Sarkar C, Singh M, Sharma MC, 2018, Epithelial-tomesenchymal transition related transcription factors areupregulated in ependymomas and correlate with a poor prognosis. Hum Pathol 82:149–157 Malgulwar PB, Pathak P, Desiraju BK, Nambirajan A, Sarkar C, Suri V, Singh M, Sharma MC, 2016, EPN-13 Epithelial-tomesenchymal transitions in childhood ependymomas mechanistically links oncogenic C11orf95-RELA fusion driven activation of SNAI1/Snail. Neuro-Oncology 18(Suppl 3):iii33. , DOI 10.1093/neuonc/now070.12 Mattick JS, 2009, The genetic signatures of noncoding RNAs. PLoS Genet 5:e1000459 Orom UA, Derrien T, Beringer M, Gumireddy K, Gardini A, Bussotti G, Lai F, Zytnicki M, Notredame C, Huang Q, Guigo R, Shiekhattar R, 2010, Long noncoding RNAs with enhancer-like function in human cells. Cell 143:46–58 Lin X, Qiu J, Hua K, 2018, Long non-coding RNAs as emerging regulators of epithelial to mesenchymal transition in gynecologic cancers. Biosci Trends 12:342–353 Zhang XQ, Leung GK, 2014, Long non-coding RNAs in glioma: functional roles and clinical perspectives. Neurochem Int 77:78–85 Zeng J, Du T, Song Y, Gao Y, Li F, Wu R, Chen Y, Li W, Zhou H, Yang Y, Pei Z, 2017, Knockdown of long noncoding RNA CCAT2 inhibits cellular proliferation, invasion, and epithelial-mesenchymal transition in glioma cells. Oncol Res 25:913–921 Rothschild G, Basu U, 2017, Lingering questions about enhancer RNA and enhancer transcription-coupled genomic instability. Trends Genet 33:143–154 Liu F, 2017, Enhancer-derived RNA: a primer. Genomics Proteomics Bioinformatics 15:196–200 Wu H, Hu Y, Liu X, Song W, Gong P, Zhang K, Chen Z, Zhou M, Shen X, Qian Y, Fan H, 2017, LncRNATRERNA1 function as an enhancer of SNAI1 promotes gastric Cancer metastasis by regulating epithelial-mesenchymal transition. Mol Ther Nucleic Acids 8: 291–299 EllisonDW, McLendon R,Wiestler OD et al, 2016, Ependymoma. In: Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, eds, WHO classification of tumours of the central nervous system, revised, 4th edn. IARC, Lyon, pp 106–112 Puget S, Grill J, Valent A, Bieche I, Dantas-Barbosa C, Kauffmann A, Dessen P, Lacroix L, Geoerger B, Job B, Dirven C, Varlet P, Peyre M, Dirks PB, Sainte-Rose C, Vassal G, 2009, Candidate genes on chromosome 9q33-34 involved in the progression of childhood EPNEPNs. J Clin Oncol 27:1884–1892 Griesinger AM,Witt DA, Grob ST, GeorgioWestover SR, Donson AM, Sanford B,Mulcahy Levy JM,Wong R, Moreira DC, DeSisto JA, Balakrishnan I, Hoffman LM, Handler MH, Jones KL, Vibhakar R, Venkataraman S, Foreman NK, 2017, NF-κB upregulation through epigenetic silencing of LDOC1 drives tumor biology and specific immunophenotype in Group A EPN. Neuro- Oncology 19:1350–1360 Rogers HA, Mayne C, Chapman RJ, Kilday JP, Coyle B, Grundy RG, 2013, PI3K pathway activation provides a novel therapeutic target for pediatric EPN and is an independent marker of progression-free survival. Clin Cancer Res 19:6450–6460 Parker M, Mohankumar KM, Punchihewa C, Weinlich R, Dalton JD, Li Y, Lee R, Tatevossian RG, Phoenix TN, Thiruvenkatam R, White E, Tang B, Orisme W, Gupta K, Rusch M, Chen X, Li Y, Nagahawhatte P, Hedlund E, Finkelstein D, Wu G, Shurtleff S, Easton J, Boggs K, Yergeau D, Vadodaria B, Mulder HL, Becksfort J, Gupta P, Huether R, Ma J, Song G, Gajjar A, Merchant T, Boop F, Smith AA, Ding L, Lu C, Ochoa K, Zhao D, Fulton RS, Fulton LL, Mardis ER, Wilson RK, Downing JR, Green DR, Zhang J, Ellison DW, Gilbertson RJ, 2014, C11orf95- RELA fusions drive oncogenic NF-κB signalling in EPN. Nature 506:451–455 Xue J, Liao L, Yin F, Kuang H, Zhou X,Wang Y, 2018, LncRNA AB073614 induces epithelial- mesenchymal transition of colorectal cancer cells via regulating the JAK/STAT3 pathway. Cancer Biomark 21:849–858 Shi G, Li H, Gao F, Tan Q, 2018, lncRNA H19 predicts poor prognosis in patients with melanoma and regulates cell growth, invasion, migration and epithelial-mesenchymal transition in melanoma cells. Onco Targets Ther 11:3583–3595 Gupta RA, Shah N,WangKC, KimJ, Horlings HM,Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL,Wang Y, Brzoska P, Kong B, Li R, West RB, van de Vijver MJ, Sukumar S, Chang HY, 2010, Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464:1071–1076 Zheng P, Li H, Xu P et al, 2017, High lncRNAHULC expression is associated with poor prognosis and promotes tumor progression by regulating epithelial-mesenchymal transition in prostate cancer. Arch Med Sci 14:679–686 ChenM, Xia Z, Chen C, HuW, Yuan Y, 2018, LncRNAMALAT1 promotes epithelial-to-mesenchymal transition of esophageal cancer through Ezh2-Notch1 signaling pathway. Anti-Cancer Drugs 29:767–773 Terashima M, Ishimura A, Wanna-Udom S, Suzuki T. MEG8 long noncoding RNA contributes to epigenetic progression of the epithelial- mesenchymal transition of lung and pancreatic cancer cells. J Biol Chem. 2018;293:18016-18030. Liang WC, Ren JL, Wong CW, Chan SO, Waye MM, Fu WM, Zhang JF, 2018, LncRNA-NEF antagonized epithelial to mesenchymal transition and cancer metastasis via cis-regulating FOXA2 and inactivating Wnt/β-catenin signaling. Oncogene 37:1445– 1456 Wu W, Chen F, Cui X, Yang L, Chen J, Zhao J, Huang D, Liu J, Yang L, Zeng J, Zeng Z, Pan Y, Su F, Cai J, Ying Z, Zhao Q, Song E, Su S, 2018, LncRNA NKILA suppresses TGF-β-induced epithelial-mesenchymal transition by blocking NF-κB signaling inbreast cancer. Int J Cancer 143:2213–2224 Gumireddy K, Li A, Yan J, Setoyama T, Johannes GJ, Orom UA, Tchou J, Liu Q, Zhang L, Speicher DW, Calin GA, HuangQ, 2013, Identification of a long non-coding RNA-associated RNP complex regulating metastasis at the translational step. EMBO J 32:2672– 2684 Miller CR, Ruppert AS, Fobare S et al, 2017, The long noncoding RNA, treRNA, decreases DNA damage and is associated with poor response to chemotherapy in chronic lymphocytic leukemia. Oncotarget 8:25942–25954 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1975 EP 1981 DI 10.1007/s12253-019-00736-8 PG 7 ER PT J AU Son, JH An, HCh Yoo, JN Lee, HS AF Son, Ji Hyun An, Hyeok Chang Yoo, Jin Nam Lee, Hyung Sug TI Tight Junction-Related CLDN5 and CLDN6 Genes, and Gap Junction-Related GJB6 and GJB7 Genes Are Somatically Mutated in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CLDN5; CLDN6; GJB6; GJB7; Mutation; Cancer ID CLDN5; CLDN6; GJB6; GJB7; Mutation; Cancer AB Tight junction and gap junction are major cell junctions that play important roles in cellular communication and structural integrity. Alterations of these junctions are known to be involved in cancer pathogenesis. Claudins and connexins are major tight and gap junction proteins, but genetic alterations of these genes have not been reported in gastric (GC) and colorectal cancers (CRC) with microsatellite instability (MSI). Claudin genes CLDN5 and CKDN6, and connexin genes GJB6 and GJB7 have mononucleotide repeats in the coding sequences that might be mutation targets in the cancers with MSI. We analyzed 79 GCs and 145 CRCs, and found CLDN5 frameshift mutations in 3 (3%) CRCs and 1 (2.9%) GC, CLDN6 frameshift mutations in 6 (6%)CRCs, GJB6 frameshift mutations in 5 (5%) CRCs and GJB7 frameshift mutation in one CRC (1%) with high MSI (MSIH). We also analyzed intratumoral heterogeneity (ITH) of the frameshift mutations in 16 CRCs and found that CLDN6 and GJB6 frameshift mutations showed regional ITH in 2 (12.5%) and 2 (12.5%) cases, respectively. Our results show that CLDN5, CLDN6, GJB6 and GJB7 genes harbor not only frameshift mutations but also mutational ITH, which together may be features of GC and CRC with MSI-H. Based on the roles of cellular junctions in cancers, frameshift mutations of tight junction and gap junction genes might contribute to tumorigenesis by altering their functions in GC and CRC. C1 [Son, Ji Hyun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [An, Hyeok Chang] The Catholic University of Korea, College of Medicine, Department of General SurgerySeoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Trosko JE, Ruch RJ, 1998, Cell-cell communication in carcinogenesis. Front Biosci 3:d208–236 Holder JW, Elmore E, Barrett JC, 1993, Gap junction function and cancer. Cancer Res 53:3475–3485 Martin TA,MasonMD, Jiang WG(2011, Tight junctions in cancer metastasis. Front Biosci 16:898–936 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Murphy K, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, Eshleman JR, 2006, Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn 8: 305–311 Choi EJ, Kim MS, Song SY, Yoo NJ, Lee SH, 2017, Intratumoral heterogeneity of frameshift mutations inMECOMGene is frequent in colorectal cancers with high microsatellite instability. Pathol Oncol Res 23:145–149 Bujko M, Kober P, Mikula M, Ligaj M, Ostrowski J, Siedlecki JA, 2015, Expression changes of cell-cell adhesion-related genes in colorectal tumors. Oncol Lett 9:2463–2470 Gao F, LiM,Xiang R, Zhou X, Zhu L, Zhai Y, 2019, Expression of CLDN6 in tissues of gastric cancer patients: association with clinical pathology and prognosis. Oncol Lett 17:4621–4625 Sirnes S, Honne H, Ahmed D, Danielsen SA, Rognum TO,Meling GI, Leithe E, Rivedal E, Lothe RA, Lind GE, 2011, DNA methylation analyses of the connexin gene family reveal silencing ofGJC1, Connexin45, by promoter hypermethylation in colorectal cancer. Epigenetics 6:602–609 Sentani K, Oue N, Sakamoto N, Anami K, Naito Y, Aoyagi K, Sasaki H, YasuiW(2010, Upregulation of connexin 30 in intestinal phenotype gastric cancer and its reduction during tumor progression. Pathobiology 77:241–248 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1983 EP 1987 DI 10.1007/s12253-020-00806-2 PG 5 ER PT J AU Handra-Luca, A AF Handra-Luca, Adriana TI Nuclear Cytoplasmic Inclusions in Lung Adenocarcinoma: Relevance of Immunohistochemistry SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Handra-Luca, Adriana] Universite Paris Nord Sorbonne Cite, APHP GHU Avicenne, Service d’Anatomie Pathologique, 125 rue Stalingrad, 93009 Bobigny, France. RP Handra-Luca, A (reprint author), Universite Paris Nord Sorbonne Cite, APHP GHU Avicenne, Service d’Anatomie Pathologique, 93009 Bobigny, France. EM adriana.handra-luca@aphp.fr;adriana.handra-luca@hotmail.com;adriana.handra-luca@univ-paris13.fr CR Kobayashi S, Saio M, Fukuda T et al, 2019, Image analysis of the nuclear characteristics of emerin protein and the correlation with nuclear grooves and intranuclear cytoplasmic inclusions in lung adenocarcinoma. Oncol Rep 41:133 142 Rosai J, 2011, Rosai and Ackman’s Surgical Pathology. 10th edition expert consult. Mosby Elsevier, Philadelphia Torikata C, Ishiwata K, 1977, Intranuclear tubular structures observed in the cells of an alveolar cell carcinoma of the lung. Cancer 40:1194 1201 Gyorkey F, Gyorkey P, Sinkovics JG, 1980, Origin and significance of intranuclear tubular inclusions in type II pulmonary alveolar epithelial cells of patients with bleomycin and busulfan toxicity. Ultrastruct Pathol 1:211 221 Singh G, Katyal SL, Torikata C, 1982, Carcinoma of type II pneumocytes. PAS staining as a screening test for nuclear inclusions of surfactant specific apoprotein. Cancer 50:946 948 Mizutani Y, Nakajima T, Morinaga S et al, 1988, Immunohistochemical localization of pulmonary surfactant apoproteins in various lung tumors. Special reference to nonmucus producing lung adenocarcinomas. Cancer 61:532 537 Knuhtsen H, Hiebert E, Purcifull DE, 1974, Partial purification and some properties of tobacco etch virus induced intranuclear inclusions. Virology 61:200 209 Schwertheim S,Westerwick D, Jastrow H, Theurer S, Schaefer CM, Kalsch J, Mollmann D, Schlattjan M, Wedemeyer H, Schmid KW, Baba HA, 2019, Intranuclear inclusions in hepatocellular carcinoma contain autophagy-associated proteins and correlate with prolonged survival. J Pathol Clin Res 5:164 176 Aida S, Shimazaki H, Sato K et al, 2001, Prognostic significance of frequent acidophilic nuclear inclusions in adenocarcinoma of the lung with immunohistochemical and ultrastructural studies. Cancer 91:1896 1904 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1989 EP 1990 DI 10.1007/s12253-019-00714-0 PG 2 ER PT J AU Wang, B Tao, L Huang, Y AF Wang, Bolin Tao, Lin Huang, Yan TI Comment on “The Significance of Long Non-coding RNA HULC in Predicting Prognosis and Metastasis of Cancers: a Meta-Analysis” SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Wang, Bolin] Weifang School of MedicineWeifang, Shandong, China. [Tao, Lin] Weifang School of MedicineWeifang, Shandong, China. [Huang, Yan] Affiliated Hospital of Weifang Medical University, Department of OncologyWeifang, Shandong, China. RP Huang, Y (reprint author), Affiliated Hospital of Weifang Medical University, Department of Oncology, Weifang, China. EM Yanhuangdr@163.com CR Ding Y, Sun C, Li J, Hu L, Li M, Liu J, Pu L, Xiong S, 2019, The significance of long non-coding RNA HULC in predicting prognosis and metastasis of cancers: a meta-analysis. Pathol Oncol Res 25: 311–318 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1991 EP 1992 DI 10.1007/s12253-019-00717-x PG 2 ER PT J AU Oka, S Ono, K Nohgawa, M AF Oka, Satoko Ono, Kazuo Nohgawa, Masaharu TI Successful Retreatment with Elotuzumab for Multiple Myeloma with Extramedullary Relapse while Being Treated with Lenalidomide and Dexamethasone SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Oka, Satoko] Japanese Red Cross Society Wakayama Medical Center, Division of HematologyWakayama, Japan. [Ono, Kazuo] Japanese Red Cross Society Wakayama Medical Center, Division of PathologyWakayama, Japan. [Nohgawa, Masaharu] Japanese Red Cross Society Wakayama Medical Center, Division of HematologyWakayama, Japan. RP Oka, S (reprint author), Japanese Red Cross Society Wakayama Medical Center, Division of Hematology, Wakayama, Japan. EM okas@jasmine.ocn.ne.jp CR Blade J, CibeiraMT, Rosinol L, de Larrea CF, 2009, Extramedullary Myeloma Blood 114:SCI–SC5 Descamps G, Pellat-Deceunynck C, Szpak Y, Bataille R, Robillard N, Amiot M, 2004, The magnitude of Akt/ phosphatidylinositol 3'-kinase proliferating signaling is related to CD45 expression in human myeloma cells. J Immunol 173: 4953–4959 Guo H, Cruz-Munoz ME, Wu N, Robbins M, Veillette A, 2015, Immune cell inhibition by SLAMF7 is mediated by a mechanism requiring src kinases, CD45, and SHIP-1 that is defective in multiple myeloma cells. Mol Cell Biol 35:41–51 Kumar SK, Callander NS, Alsina M, et al, 2018, NCCN Guidelines Insights; Multiple Myeloma, Version 3.2018. J Natl Compr Canc Netw 16:11–20 DimopoulosMA, Oriol A, Nahi H, San-Miguel J, BahlisNJ, Usmani SZ, Rabin N, Orlowski RZ, Komarnicki M, Suzuki K, Plesner T, Yoon SS, Ben Yehuda D, Richardson PG, Goldschmidt H, Reece D, Lisby S, Khokhar NZ, O’Rourke L, Chiu C, Qin X, Guckert M, Ahmadi T, Moreau P, 2016, Daratumumab, Lenalidomide, and Dexamethasone for Multiple Myeloma. N Engl J Med 375:1319– 1331 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1993 EP 1995 DI 10.1007/s12253-019-00725-x PG 3 ER PT J AU Hu, HL Zeng, DD Zang, JL Chen, Z AF Hu, Hao-Liang Zeng, Dan-Dan Zang, Jing-Lei Chen, Zhe TI The Pan-Cancer Atlas: a New Chapter in Cancer Molecular Targeting Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Hu, Hao-Liang] Hunan Normal University, College of Life Sciences, 410006 Changsha, China. [Zeng, Dan-Dan] Hunan Normal University, College of Life Sciences, 410006 Changsha, China. [Zang, Jing-Lei] Hunan Normal University, College of Life Sciences, 410006 Changsha, China. [Chen, Zhe] Hunan Institute of Traffic Engineering, Nursing College, 421001 Hengyang, China. RP Zang, JL (reprint author), Hunan Normal University, College of Life Sciences, 410006 Changsha, China. EM jlzang@126.com CR Sticz T, Molnar A, Danko T et al, 2018, The effects of different mTOR inhibitors in EGFR inhibitor resistant Colon carcinoma cells. Pathol. Oncol. Res: POR. , DOI 10.1007/s12253- 018-0434-4 Evans M, O'Sullivan B, Hughes F et al, 2018, The Clinicopathological and molecular associations of PD-L1 expression in non-small cell lung Cancer: analysis of a series of 10,005 cases tested with the 22C3 assay. Pathol. Oncol. Res: POR. https:// doi.org/10.1007/s12253-018-0469-6 Cancer Genome Atlas Research N, Analysis Working Group, Genome Sequencing Center et al, 2017, Integrated genomic characterization of oesophageal carcinoma. Nature 541(7636):169– 175. , DOI 10.1038/nature20805 Hoadley KA, Yau C, Hinoue T et al, 2018, Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of Cancer. Cell 173(2):291–304 e296. , DOI 10. 1016/j.cell.2018.03.022 Ding L, Bailey MH, Porta-Pardo E et al, 2018, Perspective on oncogenic processes at the end of the beginning of Cancer genomics. Cell 173(2):305–320 e310. , DOI 10.1016/j.cell. 2018.03.033 Sanchez-Vega F, Mina M, Armenia J et al, 2018, Oncogenic signaling pathways in the Cancer Genome Atlas. Cell 173(2):321–337 e310. , DOI 10.1016/j.cell.2018.03.035 Liu Y, Sethi NS, Hinoue T et al, 2018, Comparative molecular analysis of gastrointestinal adenocarcinomas. Cancer Cell 33(4): 721–735 e728. , DOI 10.1016/j.ccell.2018.03.010 Berger AC, Korkut A, Kanchi RS et al, 2018, A comprehensive pan-Cancer molecular study of gynecologic and breast cancers. Cancer Cell 33(4):690–705 e699. , DOI 10.1016/j.ccell. 2018.03.014 Campbell JD, Yau C, Bowlby R et al, 2018, Genomic, pathway network, and immunologic features distinguishing squamous carcinomas. Cell Rep 23(1):194–212 e196. , DOI 10.1016/j. celrep.2018.03.063 Ricketts CJ, De Cubas AA, Fan H et al, 2018, The Cancer Genome Atlas comprehensive molecular characterization of renal cell carcinoma. Cell Rep 23(1):313–326 e315. , DOI 10.1016/j. celrep.2018.03.075 Taylor AM, Shih J, Ha G et al, 2018, Genomic and functional approaches to understanding Cancer aneuploidy. Cancer Cell 33(4):676–689 e673. , DOI 10.1016/j.ccell.2018.03.007 Jayasinghe RG, Cao S, Gao Q et al, 2018, Systematic analysis of splice-site-creating mutations in Cancer. Cell Rep 23(1):270–281 e273. , DOI 10.1016/j.celrep.2018.03.052 Seiler M, Peng S, Agrawal AA et al, 2018, Somatic mutational landscape of splicing factor genes and their functional consequences across 33 Cancer types. Cell Rep 23(1):282–296 e284. , DOI 10.1016/j.celrep.2018.01.088 Huang KL, Mashl RJ, Wu Y et al, 2018, Pathogenic germline variants in 10,389 adult cancers. Cell 173(2):355–370 e314. , DOI 10.1016/j.cell.2018.03.039 Cilloni D, Saglio G, 2012, Molecular pathways: BCR-ABL. Clinical Cancer Research : an official journal of the American Association for Cancer Research 18(4):930–937. , DOI 10.1158/1078-0432.CCR-10-1613 Gao Q, Liang WW, Foltz SM et al, 2018, Driver fusions and their implications in the development and treatment of human cancers. Cell Rep 23(1):227–238 e223. , DOI 10.1016/j.celrep. 2018.03.050 Cancer Genome Atlas N, 2015, Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 517(7536):576–582. , DOI 10.1038/nature14129 Laroche-Clary A, Chaire V, Algeo MP et al, 2017, Combined targeting of MDM2 and CDK4 is synergistic in dedifferentiated liposarcomas. J Hematol Oncol 10(1):123. , DOI 10. 1186/s13045-017-0482-3 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 1997 EP 1999 DI 10.1007/s12253-019-00709-x PG 3 ER PT J AU Gupta, AA Shekatkar, M Raj, Th Kheur, S AF Gupta, A Archana Shekatkar, Madhura Raj, A. Thirumal Kheur, Supriya TI Potential Role of Magnesium in Cancer Initiation and Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Gupta, A Archana] Dr. D. Y. Patil Dental College and Hospital, Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Vidyapeeth, 411041 Pune, Maharashtra, India. [Shekatkar, Madhura] Dr. D. Y. Patil Dental College and Hospital, Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Vidyapeeth, 411041 Pune, Maharashtra, India. [Raj, A. Thirumal] Sri Venkateswara Dental College and Hospital, Department of Oral Pathology and MicrobiologyChennai, India. [Kheur, Supriya] Dr. D. Y. Patil Dental College and Hospital, Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Vidyapeeth, 411041 Pune, Maharashtra, India. RP Gupta, AA (reprint author), Dr. D. Y. Patil Dental College and Hospital, Department of Oral Pathology and Microbiology, 411041 Pune, India. EM archanaanshumangupta@gmail.com CR Grober U, Schmidt J, Kisters K, 2015, Magnesium in prevention and therapy. Nutrients 7(9):8199–8226 Hosthor SS, Mahesh P, Priya SM, Sharada P, Jyotsna M, Chitra S, 2014, Quantitative analysis of serum levels of trace elements in patients with oral submucous fibrosis and oral squamous cell carcinoma: a randomized cross – sectional study. J Oral MaxillfacPathol 18:46–51 Mendes PMV, Bezerra DLC, Santos LRD, Santos LDO, Melo SRDS, Morais JBS et al, 2018, Magnesium in breast Cancer: what is its influence on the progression of this disease? Biol Trace Elem Res 184(2):334–339 Bertinato J, Xiao CW, Ratnayake WM et al, 2015, Lower serum magnesium concentration is associated with diabetes, insulin resistance, and obesity in south Asian and white Canadian women but not men. Food Nutr Res 59(1):25974 Abdelgawad IA, El-Mously RH, Saber MM, Mansour OA, Shouman SA, 2015, Significance of serum levels of vitamin D and some related minerals in breast cancer patients. Int J Clin Exp Pathol 8(4):4074–4082 Anastassopoulou J, Theophanides T, 2015, Magnesium-DNA interactions and the possible relation ofmagnesiumto carcinogenesis. Irradiation and free radicals. Crit Rev Oncol Hematol 42(1):79–91 Karki K, Pande D, Negi R, Khanna S, Khanna RS, Khanna HD, 2015, Association between biomarkers of oxidative stress, trace elements, and cell proliferation index in patients with benign and malignant breast diseases. J Environ Pathol Toxicol Oncol 34(1):1– 10 Karki K, Pande D, Negi R, Khanna S, Khanna RS, Khanna HD, 2015, Correlation of serum toll like receptor 9 and trace elements with lipid peroxidation in the patients of breast diseases. J Trace Elem Med Biol 30:11–16 Castiglioni S,Maier JA, 2011, Magnesium and cancer : adangerous liason. Magnes Res 24(3):92–100 Yu E, Ahn YS, Jang SJ, KimMJ, Yoon HS, Gong G, Choi J, 2007, Overexpression of the wip1 gene abrogates the p38 MAPK/p53/ Wip1 pathway and silences p16 expression in human breast cancers. Breast Cancer Res Treat 101(3):269–278 Bagulkar BB, Chaudhary M, Gawande M, Patil S, Gadbail A, Bagulkar S, 2013, Colorimetric determination of magnesium in blood and saliva in Oral squamous cell carcinoma and potentially malignant disorders by titan yellowmethod. J Orofac Res 3(4):240– 244 Aziz NZ, Arathi K, Prasad BG, Desai D, Shetty SJ, Shahid M, 2018, Evaluation of magnesium levels in blood and saliva of oral squamous cell carcinoma and potentially malignant disorders by xylidyl blue method. J Oral Maxillofac Pathol 22(1):147–162 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 2001 EP 2002 DI 10.1007/s12253-019-00750-w PG 2 ER PT J AU Son, JH Choi, JE Yoo, JN Lee, HS AF Son, Ji Hyun Choi, Ji Eun Yoo, Jin Nam Lee, Hyung Sug TI Mutation and Expression of a Candidate Tumor Suppressor Gene EPB41L3 in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE EPB41L3; Mutation; Expression; Colon cancer; Gastric cancer ID EPB41L3; Mutation; Expression; Colon cancer; Gastric cancer AB Erythrocyte Membrane Protein Band 4.1 Like 3 (EPB41L3) is candidate tumor suppressor gene (TSG) in various cancers. EPB41L3 downregulation has been identified in many solid cancers including gastric (GC) and colorectal cancers (CRCs), but somatic inactivating mutation along with protein expression in cancers are largely unexplored. The aim of our study was to find whether EPB41L3 gene was mutated and expressionally altered in GC and CRC. EPB41L3 gene has a mononucleotide repeat in the coding sequence that could be mutated in cancers with high microsatellite instability (MSI-H). We analyzed 79 GCs and 124 CRCs, and found that only one CRC with MSI-H (1.3%) harbored the frameshift mutation within the repeat. In immunohistochemistry, loss of EPB41L3 expression was identified in 49%of GCs and 42%of CRCs. Our data may indicate EPB41L3 that loss of expression but not frameshift mutation may play a role in GC and CRC development by inhibiting TSG functions of EPB41L3. C1 [Son, Ji Hyun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Wang Z, Zhang J, Ye M, Zhu M, Zhang B, Roy M, Liu J, An X, 2014, Tumor suppressor role of protein 4.1B/DAL-1. CellMol Life Sci 71:4815–4830 Wang H, Xu M, Cui X, Liu Y, Zhang Y, Sui Y, Wang D, Peng L, Wang D, Yu J, 2016, Aberrant expression of the candidate tumor suppressor gene DAL-1 due to hypermethylation in gastric cancer. Sci Rep 6:21755 Ohno N, Terada N, Murata S, Yamakawa H, Newsham IF, Katoh R, OharaO,Ohno S, 2004, Immunolocalization of protein 4.1B/DAL-1 during neoplastic transformation of mouse and human intestinal epithelium. Histochem Cell Biol 122:579–586 Xue F, An C, Chen L, Liu G, Ren F, Guo X, Sun H,Mei L, Sun X, Li J, Tang Y, An X, Zheng P, 2019, 4.1B suppresses cancer cell proliferation by binding to EGFR P13 region of intracellular juxtamembrane segment. Cell Commun Signal 17:115 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Je EM, Kim MR, Min KO, Yoo NJ, Lee SH, 2012, Mutational analysis of MED12 exon 2 in uterine leiomyoma and other common tumors. Int J Cancer 131:E1044–E1047 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 2003 EP 2005 DI 10.1007/s12253-019-00787-x PG 3 ER PT J AU Mo, YH Choi, JE Yoo, JN Lee, HS AF Mo, Yoon Ha Choi, Ji Eun Yoo, Jin Nam Lee, Hyung Sug TI Mutational alterations of TDRD 1, 4 and 9 genes in colorectal cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Mo, Yoon Ha] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Jiang Y, Liu L, Shan W, Yang ZQ, 2016, An integrated genomic analysis of Tudor domain-containing proteins identifies PHD finger protein 20-like 1, PHF20L1, as a candidate oncogene in breast cancer. Mol Oncol 10:292–302 GuijoM, Ceballos-Chavez M, Gomez-Marin E, Basurto-Cayuela L, Reyes JC, 2017, Expression of TDRD9 in a subset of lung carcinomas by CpG island hypomethylation protects from DNA damage. Oncotarget 9:9618–9631 Boormans JL, Korsten H, Ziel-van der Made AJ, van Leenders GJ, de Vos CV, Jenster G, Trapman J, 2013, Identification of TDRD1 as a direct target gene of ERG in primary prostate cancer. Int J Cancer 133:335–345 Yoon H, Lee H, Kim HJ, You KT, Park YN, Kim H, Kim H, 2011, Tudor domain-containing protein 4 as a potential cancer/testis antigen in liver cancer. Tohoku J Exp Med 224:41–46 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 McGranahan N, Swanton C, 2015, Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 27: 15–26 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 2007 EP 2008 DI 10.1007/s12253-020-00798-z PG 2 ER PT J AU Mehemmai, Ch Cherbal, F Hamdi, Y Guedioura, A Benbrahim, W Bakour, R Abdelhak, S AF Mehemmai, Chiraz Cherbal, Farid Hamdi, Yosr Guedioura, Abdelmoumene Benbrahim, Wassila Bakour, Rabah Abdelhak, Sonia TI Correction to: BRCA1 and BRCA2 Germline Mutation Analysis in Hereditary Breast/Ovarian Cancer Families from the Aures Region (Eastern Algeria): First Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Page 1, under Abstract section, lines 11–12 should be rewritten as: Interestingly, we also detected a BRCA1 exon 15 deletion in two unrelated young female TNBC patients with strong family history of breast/ovarian cancer. C1 [Mehemmai, Chiraz] USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, El Alia, Bab Ezzouar, 16111 Algiers, Algeria. [Cherbal, Farid] USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, El Alia, Bab Ezzouar, 16111 Algiers, Algeria. [Hamdi, Yosr] University of Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Biomedical Genomics and Oncogenetics (LRTI, IPT 05)Tunis, Tunisia. [Guedioura, Abdelmoumene] USTHB, Faculty of Biological Sciences, LOBEMAlgiers, Algeria. [Benbrahim, Wassila] Anti-cancer center of BatnaBatna, Algeria. [Bakour, Rabah] USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, El Alia, Bab Ezzouar, 16111 Algiers, Algeria. [Abdelhak, Sonia] University of Tunis El Manar, Institut Pasteur de Tunis, Laboratory of Biomedical Genomics and Oncogenetics (LRTI, IPT 05)Tunis, Tunisia. RP Cherbal, F (reprint author), USTHB, Faculty of Biological Sciences, Laboratory of Molecular and Cellular Biology, Unit of Genetics, 16111 Algiers, Algeria. EM farid.cherbal@gmail.com NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 2009 EP 2010 DI 10.1007/s12253-019-00586-4 PG 2 ER PT J AU Kim, U Kim, CY Lee, MJ Oh, H Ryu, B Kim, J Park, JH AF Kim, Ukjin Kim, C-Yoon Lee, Min Ji Oh, Hanseul Ryu, Bokyeong Kim, Jin Park, Jae-Hak TI Correction to: Phloretin Inhibits the Human Prostate Cancer Cells through the Generation of Reactive Oxygen Species SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Correction to: Pathology & Oncology Research https://doi.org/10.1007/s12253-019-00643-y The original version of this article unfortunately contained an error in Figs. 1, 5 and 6. The asterisks and bars indicating statistical significance were missing in the figures. The corrected Figs. 1, 5 and 6 are shown below. The original article has been corrected. C1 [Kim, Ukjin] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. [Kim, C-Yoon] Konkuk University, School of Medicine, Department of Stem Cell Biology, 05029 Seoul, South Korea. [Lee, Min Ji] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. [Oh, Hanseul] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. [Ryu, Bokyeong] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. [Kim, Jin] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. [Park, Jae-Hak] Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. RP Park, JH (reprint author), Seoul National University, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, 08826 Seoul, South Korea. EM pjhak@snu.ac.kr NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 2011 EP 2012 DI 10.1007/s12253-019-00643-y PG 2 ER PT J AU Mohamed, EZAF El, ZIK Toni, DMN AF Mohamed, El-Zahraa Ammar Fatma El, Zahraa Ibrahim Khalil Toni, D M Nisreen TI Correction to: Caveolin-1 Expression Together with VEGF can be a Predictor for Lung Metastasis and Poor Prognosis in Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Correction to: Pathology & Oncology Research https://doi.org/10.1007/s12253-019-00755-5 The original version of this article unfortunately contained an error. The Tables 1 and 2 were missing in the published paper. The missing Tables 1 and 2 are shown in the next page. The original article has been corrected. C1 [Mohamed, El-Zahraa Ammar Fatma] Minia University, Faculty of Medicine, Pathology DepartmentMinia, Egypt. [El, Zahraa Ibrahim Khalil] Minia University, Faculty of Medicine, Pathology DepartmentMinia, Egypt. [Toni, D M Nisreen] Minia University, Faculty of Medicine, Pathology DepartmentMinia, Egypt. RP El, ZIK (reprint author), Minia University, Faculty of Medicine, Pathology Department, Minia, Egypt. EM zahraa333eg@gmail.com NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 2013 EP 2014 DI 10.1007/s12253-019-00790-2 PG 2 ER PT J AU Zheng, J Xu, T Feng, Ch Zhang, Y AF Zheng, Jing Xu, Tingting Feng, Chen Zhang, Ying TI Correction to: MiRNA-195-5p Functions as a Tumor Suppressor and a Predictive of Poor Prognosis in Non-small Cell Lung Cancer by Directly Targeting CIAPIN1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Correction to: Pathology & Oncology Research(2019)25:1181–1190 https://doi.org/10.1007/s12253-018-0552-z The original version of this article unfortunately contained an error in the affiliation section. The affiliation should have been Department of Respiratory Medicine, Taizhou Municiple Hospital, Zhejiang, China. The corrected affiliation is shown below. C1 [Zheng, Jing] Taizhou Municiple Hospital, Department of Respiratory MedicineZhejiang, China. [Xu, Tingting] Taizhou Municiple Hospital, Department of Respiratory MedicineZhejiang, China. [Feng, Chen] Taizhou Municiple Hospital, Department of Respiratory MedicineZhejiang, China. [Zhang, Ying] Taizhou Municiple Hospital, Department of Respiratory MedicineZhejiang, China. RP Xu, T (reprint author), Taizhou Municiple Hospital, Department of Respiratory Medicine, Zhejiang, China. EM Xu_tingting09@126.com NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2020 VL 26 IS 3 BP 2015 EP 2015 DI 10.1007/s12253-020-00794-3 PG 1 ER PT J AU Lee, EJ Zhu, Z Bai, Q Brady, JT Xiao, H Wakefield, RM Fang, Y AF Lee, E Jacob Zhu, Ziwen Bai, Qian Brady, J Tucker Xiao, Huaping Wakefield, R Mark Fang, Yujiang TI The Role of Interleukin-9 in Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Interleukin 9; Cancer; Inflammation ID Interleukin 9; Cancer; Inflammation AB Interluekin-9 (IL-9) is produced predominantly by helper T cells such as Th2 and Th9 cells. It normally functions through the activation of a JAK/STAT pathway and plays a critical role in immunity and the pathogenesis of cancer. In cancer, it yields different responses depending on the cancer cell line involved. This review is a summary of what is known about the involvement of IL-9 in various cancer cell lines as well as its role in immunity with a focus on allergic responses. C1 [Lee, E Jacob] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, Iowa, USA. [Zhu, Ziwen] University of Missouri, School of Medicine, Department of SurgeryColumbia, MO, USA. [Bai, Qian] University of Missouri, School of Medicine, Department of SurgeryColumbia, MO, USA. [Brady, J Tucker] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, Iowa, USA. [Xiao, Huaping] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, Iowa, USA. [Wakefield, R Mark] University of Missouri, School of Medicine, Department of SurgeryColumbia, MO, USA. [Fang, Yujiang] Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, Iowa, USA. RP Fang, Y (reprint author), Des Moines University, College of Osteopathic Medicine, Department of Microbiology, Immunology & Pathology, 50312 Des Moines, USA. EM yujiang.fang@dmu.edu CR Lavorgna A,Matsuoka M, Harhaj EW(2014, A critical role for IL- 17RB signaling in HTLV-1 tax-induced NF-KB activation and T cell transformation. PLoS Pathog 10(10):1–16 Visekruna A, Ritter J, Scholz T, Campos L, Guralnik A, Poncette L, Raifer H, Hagner S, Garn H, Staudt V, Bopp T, Reuter S, Taube C, Loser K, HuberM(2013, Tc9 cells, a new subset of CD8(+, Tcells, support Th2-mediated airway inflammation. Eur J Immunol 43: 606–618 Leech MD, Grencis RK, 2006, Induction of enhanced immunity to intestinal nematodes using IL-9- producing dendritic cells. J Immunol 176:2505–2511 Turner JE,Morrison PJ,Wilhelm C,Wilson M, Ahlfors H, Renauld JC, Panzer U, Helmby H, Stockinger B, 2013, IL-9-mediated survival of type 2 innate lymphoid cells promotes damage control in helminth-induced lung inflammation. J Exp Med 210:2951–2965 Hoelzinger DB, Dominguez AL, Cohen PA, Gendler SJ, 2014, Inhibition of adaptive immunity by IL-9 can be disrupted to achieve rapid T cell sensitization and rejection of progressive tumor challenges. Cancer Res 74(23):6845–6855 Nowak EC, Weaver CT, Turner H, Begum-Haque S, Becher B, Schreiner B, Coyle AJ, Kasper LH, Noelle RJ, 2009, IL-9 as a mediator of Th17-driven inflammatory disease. J Exp Med 206: 1653–1660 Elyaman W, Bradshaw EM, Uyttenhove C, Dardalhon V, Awasthi A, Imitola J, Bettelli E, Oukka M, van Snick J, Renauld JC, Kuchroo VK, Khoury SJ, 2009, IL-9 induces differentiation of TH17 cells and enhances function of FoxP3+ natural regulatory T cells. Proc Natl Acad Sci U S A 106:12885–12890 Veldhoen M, Uyttenhove C, van Snick J, HelmbyH,Westendorf A, Buer J, Martin B, Wilhelm C, Stockinger B, 2008, Transforming growth factor-β ‘reprograms’ the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset. Nature Immunol. 9:1341–1346 Lauwerys BR, Garot N, Renauld JC, Houssiau FA, 2000, Cytokine production and killer activity of NK/ T-NK cells derived with IL-2, IL-15, or the combination of IL-12 and IL-18. J Immunol 165: 1847–1853 Jones TG, Hallgren J, Humbles A, Burwell T, Finkelman FD, Alcaide P, Austen KF, Gurish MF, 2009, Antigen-induced increases in pulmonary mast cell progenitor numbers depend on IL- 9 and CD1d-restricted NKT cells. J Immunol 183:5251–5260 Schmitt E et al, 1991, IL-1 serves as a secondary signal for IL-9 expression. J Immunol 147:3848–3854 Schmitt E et al, 1994, IL-9 production of naive CD4+ T cells depends on IL-2, is synergistically enhanced by a combination of TGF-beta and IL-4, and is inhibited by IFN-gamma. J Immunol 153:3989–3996 Chang H-C et al, 2005, PU.1 expression delineates heterogeneity in primary Th2 cells. Immunity 22:693–703 Schmitt E, Klein M, Bopp T, 2014, Th9 cells, new players in adaptive immunity. Trends Immunol 35(2):61–68 Palmer MT, Lee YK, Maynard CL, Oliver JR, Bikle DD, Jetten AM, Weaver CT, 2011, Lineage-specific effects of 1,25-dihydroxyvitamin D(3, on the development of effector CD4 Tcells. J Biol Chem 286:997–1004 Chang HC et al, 2010, The transcription factor PU.1 is required for the development of IL-9-producing T-cells and allergic inflammation. Nature Immunol 11:527–534 Angkasekwinai P, Chang SH, Thapa M,Watarai H, Dong C, 2010, Regulation of IL-9 expression by IL-25 signaling. Nature Immunol 11:250–256 Ahyi AN, Chang HC, Dent AL, Nutt SL, Kaplan MH, 2009, IFN regulatory factor 4 regulates the expression of a subset of Th2 cytokines. J Immunol 183:1598–1606 Strahl BD, Allis CD, 2000, The language of covalent histone modifications. Nature 403:41–45 Kouzarides T, 2000, Acetylation: a regulatory modification to rival phosphorylation? EMBO J 19:1176–1179 Mak KS, Funnell AP, Pearson RC, Crossley M, 2011, PU.1 and Haematopoietic cell fate: dosage matters. Int. J. Cell biol:808524 Asao H, Okuyama C, Kumaki S, Ishii N, Tsuchiya S, Foster D, Sugamura K, 2001, Cutting edge: the common gamma-chain is an indispensable subunit of the IL-21 receptor complex. J Immunol 167:1–5 Demoulin JB, Renauld JC, 1998, "Signalling by cytokines interacting with the interleukin-2 receptor gamma chain", cytokines cell. Mol Ther 4:243–256 Demoulin JB, Uyttenhove C, Van Roost E, de Lestre B, Donckers D, Van Snick J, Renauld JC, 1996, "a single tyrosine of the interleukin-9, IL-9, receptor is required for STAT activation, antiapoptotic activity, and growth regulation by IL-9", Mol cell. Biol. 16:4710–4716 Renauld JC, Druez C, Kermouni A, Houssiau F, Uyttenhove C, Van Roost E, Van Snick J, 1992, Expression cloning of the murine and human interleukin 9 receptor cDNAs. Proc Natl Acad SciU S A 89: 5690–5694 Zhu YX, Sun HB, Tsang ML, McMahel J, Grigsby S, Yin T, Yang YC, 1997, Critical cytoplasmic domains of human interleukin-9 receptor alpha chain in interleukin-9-mediated cell proliferation and signal transduction. J Biol Chem 272:21334–21340 Zhang J, Wang WD, Geng QR, Wang L, Chen XQ, Liu CC, Lv Y, 2014, Serum levels of interleukin-9 correlate with negative prognostic factors in extranodal NK/T-cell lymphoma. PLoS One 9: e94637 Lv X, Feng L, Fang X, Jiang Y,Wang X, 2013, Overexpression of IL-9 receptor in diffuse large B-cell lymphoma. Int J Clin Exp Pathol 6:911–916 Hamre H, Zeller B, Kanellopoulos A, Ruud E, Fossa SD, Loge JH, Aukrust P, Halvorsen B, Mollnes TE, Kiserud CE, 2013, Serum cytokines and chronic fatigue in adults surviving after childhood leukemia and lymphoma. Brain Behav Immun 30:80–87 Kobayashi H, Kumai T, Hayashi S, Matsuda Y, Aoki N, Sato K, Kimura S, Celis E, 2012, A naturally processed HLA-DR-bound peptide from the IL-9 receptor alpha of HTLV-1-transformed Tcells serves as a T helper epitope. Cancer Immunol Immunother 61: 2215–2225 Fischer M, Bijman M, Molin D, Cormont F, Uyttenhove C, van Snick J, SundstromC, Enblad G, NilssonG, 2003, Increased serum levels of interleukin-9 correlate to negative prognostic factors in Hodgkin's lymphoma. Leukemia 17:2513–2516 Huang Y, Cao Y, Zhang S et al, 2015, Association between low levels of interleukin-9 and colon cancer progression. Experimental and Therapeutic Medicine 10:942–946 Ye ZJ, Zhou Q, YinW, Yuan ML, YangWB, Xiong XZ, Zhang JC, Shi HZ, 2012, Differentiation and immune regulation of IL-9 producing CD4+ T cells in malignant pleural effusion. Am J Respir Crit Care Med 186:1168–1179 Kelleher K, Bean K, Clark SC, Leung WY, Yang-Feng TL, Chen JW, Lin PF, Luo W, Yang YC, 1991, Human interleukin-9: genomic sequence, chromosomal location, and sequences essential for its expression in human T-cell leukemia virus, HTLV)-I-transformed human T cells. Blood 77:1436–1441 Chen N, Lv X, Li P, Lu K,Wang X, 2014, Role of high expression of IL-9 in prognosis of CLL. Int J Clin Exp Pathol 7(2):716–721 Hsieh TH, Hsu CY, Tsai CF et al, 2016, A novel cell penetrating peptide suppresses breast tumorigenesis by inbiting beta-catenin/ LEF-1 signaling. Sci Rep:1–12 Eller K,Wolf D, Huber JM,MetzM, Mayer G,McKenzie AN et al IL-9 production by regulatory T cells recruits mast cells that are essential for regulatory T cell-induced immune suppression. J Immunol 186:83–91 Lu LF, Lind EF, Gondek DC, Bennett KA, Gleeson MW, Pino- Lagos K, Scott ZA, Coyle AJ, Reed JL, van Snick J, Strom TB, Zheng XX, Noelle RJ, 2006)Mast cells are essential intermediaries in regulatory T-cell tolerance. Nature. 442:997–1002 Yang Z, Zhang B, Li D, Lv M, Huang C, Shen GX et al Mast cells mobilize myeloid-derived suppressor cells and Treg cells in tumor microenvironment via IL-17 pathway in murine hepatocarcinoma model. PLoS ONE 5:e8922 Zimancevic-Simonovic S, Mihaljevic O, Majsotrovic I et al, 2015, Cytokine production in patients with papillary thyroid cancer and associated autoimmune Hashimoto thyroiditis. Cancer Immunol Immunother 64:1011–1019 Qiu L, Lai R, Lin Q, Lau E, Thomazy DM, Calame D, Ford RJ, Kwak LW, Kirken RA, Amin HM, 2006, Autocrine release of interleukin-9 promotes Jak3-dependent survival of ALK+ anaplastic large-cell lymphoma cells. Blood 108(7):2407–2415 Koo GC, Tan SY, Tang T, Poon SL, Allen GE, Tan L, Chong SC, OngWS, Tay K, TaoM, Quek R, Loong S, YeohKW, Yap SP, Lee KA, Lim LC, Tan D, Goh C, Cutcutache I, Yu W, Young Ng CC, Rajasegaran V, Heng HL, Gan A, Ong CK, Rozen S, Tan P, Teh BT, Lim ST, 2012, Janus kinase 3-activating mutations identified in natural killer/T-cell lymphoma. Cancer Discov 2:591–597 Bouchekioua A, Scourzic L, de Wever O, Zhang Y, Cervera P, Aline-Fardin A, Mercher T, Gaulard P, Nyga R, Jeziorowska D, Douay L, Vainchenker W, Louache F, Gespach C, Solary E, Coppo P, 2014, JAK3 deregulation by activatingmutations confers invasive growth advantage in extranodal nasal-type natural killer cell lymphoma. Leukemia 28:338–348 Poiesz BJ, Ruscetti FW, Mier JW,Woods AM, Gallo RC, 1980, Tcell lines established from human T-lymphocytic neoplasias by direct response to T-cell growth factor. Proc Natl Acad Sci U S A 77: 6815–6819 Chen N, Lu K, Li P, Lv X,Wang X, 2014, Overexpression of IL-9 induced by STAT6 activation promotes the pathogenesis of chronic lymphocytic leukemia. Int J Clin Exp Pathol 7:2319–2323 Vieyra-Garcia PA, Wei T, Naym DG et al, 2016, STAT3/5 dependent IL-9 overexpression contributes to neoplastic cell survival in mycosis fungoides. Clin Cancer Res:1–32 Merz H, Houssiau FA, Orscheschek K, Renauld JC, Fliedner A, Herin M, Noel H, Kadin M, Mueller-Hermelink HK, Van Snick J et al, 1991, Interleukin-9 expression in human malignant lymphomas: unique association with Hodgkin’s disease and large cell anaplastic lymphoma. Blood. 78:1311–1317 Rawstron AC, Bottcher S, Letestu R, Villamor N, Fazi C, Kartsios H, de Tute RM, Shingles J, Ritgen M,Moreno C, Lin K, Pettitt AR, KnebaM,Montserrat E, Cymbalista F, Hallek M, Hillmen P, Ghia P, 2013, Improving efficiency and sensitivity: European research initiative in CLL, ERIC, update on the international harmonised approach for flow cytometric residual disease monitoring in CLL. Leukemia 27:142–149 Nagato T, Kobayashi H, Kishibe K, Takahara M, Ogino T, Ishii H, Oikawa K, Aoki N, Sato K, Kimura S, Shimizu N, Tateno M, Harabuchi Y, 2005, Expression of interleukin-9 in nasal natural killer/T-cell lymphoma cell lines and patients. Clin Cancer Res 11:8250–8257 Fang Y, Chen X, Bai Q, Qin C, Mohamud AO, Zhu Z, Ball TW, Ruth CM, Newcomer DR, Herrick EJ, Nicholl MB, 2015, IL-9 inhibits HTB-72 melanoma cell growth through upregulation of p21 and TRAIL. J Surg Oncol 111:969–974 Kim IK, Kim BS, Koh CH, Seok JW, Park JS, Shin KS, Bae EA, Lee GE, Jeon H, Cho J, Jung Y, Han D, Kwon BS, Lee HY, Chung Y, Kang CY, 2015, Glucocorticoid-induced tumor necrosis factor receptor-related protein co-stimulation facilitates tumor regression by inducing IL-9-proudcing helper T cells. Nat Med 21(9):1010– 1017 LuY, Hong S, Li H, Park J, Hong B,Wang L, ZhengY, Liu Z, Xu J, He J, Yang J, Qian J, Yi Q, 2012, Th9 Cells Promote Antitumor immune responses in vivo. J Clin Investig 122(11):4160–4171 Lu Y, Wang Q, Yi Q, 2014, Anticancer Tc9 cells. Oncolmmunology 1-1:1–3 Lu Y, Hong B, Li H, 2014, Tumor specific IL-9-producing CD8+ Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy of cancer cells. PNAS 111(6):2265–2270 Nakatsukasa H, Zhang D, Maruyama T, Chen H, Cui K, Ishikawa M, Deng L, Zanvit P, Tu E, JinW, Abbatiello B, Goldberg N, Chen Q, Sun L, Zhao K, Chen WJ, 2015, The DNA binding inhibitor Id3 regulates IL-9 production in CD4+ T cells. Nat Immunol 16(10): 1077–1084 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2017 EP 2022 DI 10.1007/s12253-019-00665-6 PG 6 ER PT J AU Qin, J Wen, B Liang, Y Yu, W Li, H AF Qin, Jingchun Wen, Bin Liang, Yuqi Yu, Weitao Li, Huixuan TI Histone Modifications and their Role in Colorectal Cancer (Review) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Histone modification; Colorectal cancer; Histone acetylation; Histone methylation; Histone phosphorylation ID Histone modification; Colorectal cancer; Histone acetylation; Histone methylation; Histone phosphorylation AB The development of colorectal cancer is a complex and multistep process mediated by a variety of factors including the dysregulation of genetic and epigenetic under the influence of microenvironment. It is evident that epigenetics that affects gene activity and expression has been recognized as a critical role in the carcinogenesis. Aside from DNA methylation, miRNA level, and genomic imprinting, histone modification is increasingly recognized as an essential mechanism underlying the occurrence and development of colorectal cancer. Aberrant regulation of histone modification like acetylation, methylation and phosphorylation levels on specific residues is implicated in a wide spectrum of cancers, including colorectal cancer. In addition, as this process is reversible and accompanied by a plethora of deregulated enzymes, inhibiting those histone-modifying enzymes activity and regulating its level has been thought of as a potential path for tumor therapy. This review provides insight into the basic information of histone modification and its application in the colorectal cancer treatment, thereby offering new potential targets for treatment of colorectal cancer. C1 [Qin, Jingchun] Guangzhou University of Chinese Medicine, Institute of Spleen and Stomach, 510000 Guangzhou, China. [Wen, Bin] Guangzhou University of Chinese Medicine, Institute of Spleen and Stomach, 510000 Guangzhou, China. [Liang, Yuqi] Guangzhou University of Chinese Medicine, Institute of Spleen and Stomach, 510000 Guangzhou, China. [Yu, Weitao] Lianyungang Affiliated Hospital of Nanjing University of Traditional Chinese MedicineNanjing, China. [Li, Huixuan] Guangzhou University of Chinese Medicine, Institute of Spleen and Stomach, 510000 Guangzhou, China. RP Wen, B (reprint author), Guangzhou University of Chinese Medicine, Institute of Spleen and Stomach, 510000 Guangzhou, China. EM wenbin@gzucm.edu.cn CR Okugawa Y, Grady WM, Goel A, 2015, Epigenetic alterations in colorectal cancer: emerging biomarkers. Gastroenterology 149(5): 1204–1225.e12 Gieni RS, Hendzel MJ, 2009, Polycomb group protein gene silencing, non-coding RNA, stem cells, and cancer. Biochem Cell Biol 87(5):711–746 Lawrence M, Daujat S, Schneider R, 2016, Lateral thinking: how histone modifications regulate gene expression. Trends Genet 32(1):42–56 Werner RJ, Kelly AD, Issa JJ, 2017, Epigenetics and precision oncology. Cancer J 23(5):262–269 Yang WY, Gu JL, Zhen TM, 2014, Recent advances of histone modification in gastric cancer. J Cancer Res Ther 10(Suppl):240–245 Nowacka-Zawisza M, Wisnik E, 2017, DNA methylation and histone modifications as epigenetic regulation in prostate cancer. Oncol Rep 38(5):2587–2596 Mehta A, Dobersch S, Romero-Olmedo AJ, Barreto G, 2015, Epigenetics in lung cancer diagnosis and therapy. Cancer Metastasis Rev 34(2):229–241 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JWW, Comber H, Forman D, Bray F, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49(6):1374–1403 De Majo F, Calore M, 2018, Chromatin remodelling and epigenetic state regulation by non-coding RNAs in the diseased heart. Non-coding RNA Research 3(1):20–28 Coppede F, 2014, Genetic and epigenetic biomarkers for diagnosis, prognosis and treatment of colorectal cancer. World J Gastroenterol 20(4):943–947 Jenuwein T, Allis CD, 2001, Translating the histone code. Science 293(5532):1074–1080 Rothbart SB, Strahl BD, 2014, Interpreting the language of histone and DNA modifications. Biochimica et Biophysica Acta, BBA, - Gene Regulatory Mechanisms 1839(8):627–643 Fu LN, Tan J, Chen YX, Fang JY, 2018, Genetic variants in the histone methylation and acetylation pathway and their risks in eight types of cancers. J Dig Dis 19(2):102–111 Azieva AM, Sheinov AA, Galkin FA, Georgieva SG, Soshnikova NV, 2018, Stability of chromatin remodeling complex subunits is determined by their phosphorylation status. Dokl Biochem Biophys 479(1):66–68 Stral BD, Allis CD, 2000, The language of covalent histone modifications. Nature 403(6):41–45 Gurard-Levin ZA, Almouzni G, 2014, Histone modifications and a choice of variant: a language that helps the genome express itself. F1000Prime Rep 6:76–86 Seligson DB, Horvath S, McBrian MA, Mah V, Yu H, Tze S, Wang Q, Chia D, Goodglick L, Kurdistani SK, 2009, Global levels of histone modifications predict prognosis in different cancers. Am J Pathol 174(5):1619–1628 O'Hagan HM, 2014, Chromatin modifications during repair of environmental exposure-induced DNA damage: a potential mechanism for stable epigenetic alterations. Environ Mol Mutagen 55(3):278–291 Berndsen CE, Denu JM, 2008, Catalysis and substrate selection by histone/protein lysine acetyltransferases. Curr Opin Struct Biol 18(6):682–689 de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB, 2003, Histone deacetylases, HDACs): characterization of the classical HDAC family. Biochem J 370(Pt 3):737–749 Mehrotra S, Galdieri L, Zhang T, Zhang M, Pemberton LF, Vancura A, 2014, Histone hypoacetylation-activated genes are repressed by acetyl-CoA- and chromatin-mediated mechanism. Biochimica et Biophysica Acta, BBA, - Gene Regulatory Mechanisms 1839(9):751–763 Graff J, Tsai L, 2013, Histone acetylation: molecular mnemonics on the chromatin. Nat Rev Neurosci 14(2):97–111 Wei S, Li C, Yin Z, Wen J, Meng H, Xue L, Wang J, 2018, Histone methylation in DNA repair and clinical practice: new findings during the past 5-years. J Cancer 9(12):2072–2081 Di Lorenzo A, Bedford MT, 2011, Histone arginine methylation. FEBS Lett 585(13):2024–2031 Wood K, Tellier M, Murphy S, 2018, DOT1L and H3K79 methylation in transcription and genomic stability. Biomolecules 8(1):11 Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA, Shi Y, 2004, Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119(7):941–953 Berger SL, 2007, The complex language of chromatin regulation during transcription. Nature 447(7143):407–412 Dawson MA, Foster SD, Bannister AJ, Robson SC, Hannah R, Wang X, Xhemalce B, Wood AD, Green AR, Gottgens B et al, 2012, Three distinct patterns of histone H3Y41 phosphorylation mark active genes. Cell Rep 2(3):470–477 Brehove M, Wang T, North J, Luo Y, Dreher SJ, Shimko JC, Ottesen JJ, Luger K, Poirier MG, 2015, Histone core phosphorylation regulates DNA accessibility. J Biol Chem 290(37):22612– 22621 Hurd PJ, Bannister AJ, Halls K, Dawson MA, Vermeulen M, Olsen JV, Ismail H, Somers J, Mann M, Owen-Hughes T et al, 2009, Phosphorylation of histone H3 Thr-45 is linked to apoptosis. J Biol Chem 284(24):16575–16583 Darieva Z, Webber A, Warwood S, Sharrocks AD, 2015, Protein kinase C coordinates histone H3 phosphorylation and acetylation. Elife 4:e09886 Trevino LS, Wang Q, Walker CL, 2015, Phosphorylation of epigenetic Breaders, writers and erasers^: implications for developmental reprogramming and the epigenetic basis for health and disease. Prog Biophys Mol Biol 118(1–2):8–13 Dawson MA, Bannister AJ, Gottgens B, Foster SD, Bartke T, Green AR, Kouzarides T, 2009, JAK2 phosphorylates histone H3Y41 and excludes HP1α from chromatin. Nature 461(7265): 819–822 Wang F, Dai J, Daum JR, Niedzialkowska E, Banerjee B, Stukenberg PT, Gorbsky GJ, Higgins JMG, 2010, Histone H3 Thr-3 phosphorylation by Haspin positions Aurora B at centromeres in mitosis. Science 330(6001):231–235 Kawashima SA, Yamagishi Y, Honda T, Ishiguro K, Watanabe Y, 2010, Phosphorylation of H2A by Bub1 prevents chromosomal instability through localizing shugoshin. Science 327(5962):172–177 Alaskhar Alhamwe B, Khalaila R,Wolf J, von Bulow V, Harb H, Alhamdan F, Hii CS, Prescott SL, Ferrante A, Renz H et al, 2018, Histone modifications and their role in epigenetics of atopy and allergic diseases. Allergy, Asthma Clin Immunol 14(1) He H, Hu Z, Xiao H, Zhou F, Yang B, 2018, The tale of histone modifications and its role in multiple sclerosis. Human Genomics 12(1) Biswas S, Rao CM, 2017, Epigenetics in cancer: fundamentals and beyond. Pharmacol Ther 173:118–134 Elsheikh SE, Green AR, Rakha EA, Powe DG, Ahmed RA, Collins HM, Soria D, Garibaldi JM, Paish CE, Ammar AA et al, 2009, Global histone modifications in breast Cancer correlate with tumor phenotypes, prognostic factors, and patient outcome. Cancer Res 69(9):3802–3809 Liu Y, Wang D, Chen S, Zhao L, Sun F, 2012, Oncogene Ras/ phosphatidylinositol 3-kinase signaling targets histone H3 acetylation at lysine 56. J Biol Chem 287(49):41469–41480 Hashimoto T, Yamakawa M, Kimura S, Usuba O, Toyono M, 2013, Expression of acetylated and Dimethylated histone H3 in colorectal cancer. Dig Surg 30(3):249–258 Karczmarski J, Rubel T, Paziewska A, Mikula M, BujkoM, Kober P, Dadlez M, Ostrowski J, 2014, Histone H3 lysine 27 acetylation is altered in colon cancer. Clin Proteomics 11(1):24 Sun W, Zhou X, Zheng J, Lu M, Nie J, Yang X, Zheng Z, 2011, Histone acetyltransferases and deacetylases: molecular and clinical implications to gastrointestinal carcinogenesis: figure 1. Acta Biochim Biophys Sin 44(1):80–91 Bardhan K, Paschall AV, Yang D, Chen MR, Simon PS, Bhutia YD,Martin PM, ThangarajuM, Browning DD, Ganapathy Vet al, 2015, IFN induces DNAmethylation-silenced GPR109A expression via pSTAT1/p300 and H3K18 acetylation in colon cancer. Cancer Immunol Res 3(7):795–805 Ashktorab H, Belgrave K, Hosseinkhah F, Brim H, Nouraie M, Takkikto M, Hewitt S, Lee EL, Dashwood RH, Smoot D, 2009, Global histone H4 acetylation and HDAC2 expression in Colon adenoma and carcinoma. Dig Dis Sci 54(10):2109–2117 Tamagawa H, Oshima T, Shiozawa M, Morinaga S, Nakamura Y, Yoshihara M, Sakuma Y, Kameda Y, Akaike M, Masuda M et al, 2012, The global histone modification pattern correlates with overall survival in metachronous liver metastasis of colorectal cancer. Oncol Rep 27(3):637 ZhangY,Wang S, KangW, Liu C, DongY, Ren F,Wang Y, Zhang J,Wang G, To KF et al, 2018, CREPT facilitates colorectal cancer growth through inducing Wnt/β-catenin pathway by enhancing p300-mediated β-catenin acetylation. Oncogene 37(26):3485– 3500 Gezer U, Ustek D, Yoruker EE, Cakiris A, Abaci N, Leszinski G, Dalay N, Holdenrieder S, 2013, Characterization of H3K9me3- and H4K20me3-associated circulating nucleosomal DNA by high-throughput sequencing in colorectal cancer. Tumor Biol 34(1):329–336 Tamagawa H, Oshima T, Numata M, Yamamoto N, Shiozawa M, Morinaga S, Nakamura Y, Yoshihara M, Sakuma Y, Kameda Y et al, 2013, Global histone modification of H3K27 correlates with the outcomes in patients with metachronous liver metastasis of colorectal cancer. Eur J Surg Oncol 39(6):655–661 Benard A, Goossens-Beumer IJ, van Hoesel AQ, de Graaf W, Horati H, Putter H, ECM Z, van de Velde CJH, Kuppen PJK, 2014, Histone trimethylation at H3K4, H3K9 and H4K20 correlates with patient survival and tumor recurrence in early-stage colon cancer. BMC Cancer 14(1):531 Yokoyama Y, Hieda M, Nishioka Y, Matsumoto A, Higashi S, Kimura H, Yamamoto H, Mori M, Matsuura S, Matsuura N, 2013, Cancer-associated upregulation of histone H3 lysine 9 trimethylation promotes cell motilityin vitro and drives tumor formation in vivo. Cancer Sci 104(7):889–895 Chen T, Li J, Xu M, Zhao Q, Hou Y, Yao L, Zhong Y, Chou P, Zhang W, Zhou P et al, 2017, PKCε phosphorylates MIIP and promotes colorectal cancer metastasis through inhibition of RelA deacetylation. Nat Commun:8(1) Yu D, Li Z, Gan M, Zhang H, Yin X, Tang S, Wan L, Tian Y, Zhang S, Zhu Yet al, 2015, Decreased expression of dual specificity phosphatase 22 in colorectal cancer and its potential prognostic relevance for stage IV CRC patients. Tumor Biol 36(11): 8531–8535 Fraga MF, Ballestar E, Villar-Garea A, Boix-Chornet M, Espada J, Schotta G, Bonaldi T,Haydon C, Ropero S, Petrie Ket al, 2005, Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet 37(4):391–400 Benard A, Goossens-Beumer IJ, van Hoesel AQ, Horati H, de Graaf W, Putter H, Zeestraten ECM, Liefers G, van de Velde CJH, Kuppen PJK, 2015, Nuclear expression of histone deacetylases and their histone modifications predicts clinical outcome in colorectal cancer. Histopathology 66(2):270–282 Cao L, Zhu L, Yang J, Su J,Ni J,Du Y, Liu D,WangY,Wang F, Jin J et al, 2014, Correlation of low expression of hMOF with clinicopathological features of colorectal carcinoma, gastric cancer and renal cell carcinoma. Int J Oncol 44(4):1207–1214 Su J, Wang F, Cai Y, Jin J, 2016, The functional analysis of histone acetyltransferase MOF in tumorigenesis. Int J Mol Sci 17(1):99 Yang H, Salz T, Zajac-Kaye M, Liao D, Huang S, Qiu Y, 2014, Overexpression of histone deacetylases in cancer cells is controlled by interplay of transcription factors and epigenetic modulators. FASEB J 28(10):4265–4279 Lutz L, Fitzner IC, Ahrens T, Geissler AL, Makowiec F, Hopt UT, Bogatyreva L, Hauschke D, Werner M, Lassmann S, 2016, Histone modifiers and marks define heterogeneous groups of colorectal carcinomas and affect responses to HDAC inhibitors in vitro. Am J Cancer Res 6(3):664–676 Cao LL, Yue Z, Liu L, Pei L, Yin Y, Qin L, Zhao J, Liu H, Wang H, Jia M, 2017, The expression of histone deacetylase HDAC1 correlates with the progression and prognosis of gastrointestinal malignancy. Oncotarget 8(24):39241–39253 Ye P, Xing H, Lou F, Wang K, Pan Q, Zhou X, Gong L, Li D, 2016, Histone deacetylase 2 regulates doxorubicin, Dox, sensitivity of colorectal cancer cells by targeting ABCB1 transcription. Cancer Chemother Pharmacol 77(3):613–621 Nemati M, Ajami N, Estiar MA, Rezapour S, Ravanbakhsh Gavgani R, Hashemzadeh S, Samadi Kafil H, Sakhinia E, 2018, Deregulated expression of HDAC3 in colorectal cancer and its clinical significance. Adv Clin Exp Med 27(3):305–311 Nakazawa T, Kondo T, Ma D, Niu D, Mochizuki K, Kawasaki T, Yamane T, Iino H, Fujii H, Katoh R, 2012, Global histone modification of histone H3 in colorectal cancer and its precursor lesions. Hum Pathol 43(6):834–842 Benard A, Goossens-Beumer IJ, van Hoesel AQ, Horati H, Putter H, Zeestraten EC, van de Velde CJ, Kuppen PJ, 2014, Prognostic value of polycomb proteins EZH2, BMI1 and SUZ12 and histone modification H3K27me3 in colorectal cancer. PLoS One 9(9):e108265 Kryczek I, Lin Y, Nagarsheth N, Peng D, Zhao L, Zhao E, Vatan L, Szeliga W, Dou Y, Owens S et al, 2014, IL-22(+)CD4(+, T cells promote colorectal cancer stemness via STAT3 transcription factor activation and induction of the methyltransferase DOT1L. Immunity 40(5):772–784 Mu Z, Li H, Fernandez SV, Alpaugh KR, Zhang R, Cristofanilli M, 2013, EZH2 knockdown suppresses the growth and invasion of human inflammatory breast cancer cells. J Exp Clin Cancer Res 32:70 Kornblihtt AR, 2017, Epigenetics at the base of alternative splicing changes that promote colorectal cancer. J Clin Invest 127(9): 3281–3283 Qin J, Zeng Z, Luo T, Li Q, Hao Y, Chen L, 2018, Clinicopathological significance of G9A expression in colorectal carcinoma. Oncol Lett 15(6):8611–8619 Wang C, 2010, EZH2 and STAT6 expression profiles are correlated with colorectal cancer stage and prognosis. World J Gastroenterol 16(19):2421–2427 Chen Z, Yang P, Li W, He F, Wei J, Zhang T, Zhong J, Chen H, Cao J, 2018, Expression of EZH2 is associated with poor outcome in colorectal cancer. Oncol Lett 15(3):2953–2961 Tokunaga R, Sakamoto Y, Nakagawa S, Miyake K, Izumi D, Kosumi K, Taki K, Higashi T, Imamura Y, Ishimoto T et al, 2016, The prognostic significance of histone lysine demethylase JMJD3/KDM6B in colorectal cancer. Ann Surg Oncol 23(2):678–685 Ding J, Zhang Z, Xia Y, Liao G, Pan Y, Liu S, Zhang Y, Yan Z, 2013, LSD1-mediated epigenetic modification contributes to proliferation and metastasis of colon cancer. Br J Cancer 109(4):994–1003 Cordeiro MH, Smith RJ, Saurin AT, 2018, A fine balancing act: a delicate kinase-phosphatase equilibrium that protects against chromosomal instability and cancer. Int J Biochem Cell Biol 96:148–156 Lee YC, Yin TC, Chen YT, Chai CY,Wang JY, Liu MC, Lin YC, Kan JY(2015)High expression of phospho-H2AX predicts a poor prognosis in colorectal cancer. Anticancer Res 35(4):2447–2453 Xiao J, Duan Q, Wang Z, Yan W, Sun H, Xue P, Fan X, Zeng X, Chen J, Shao C et al, 2016, Phosphorylation of TOPK at Y74, Y272 by Src increases the stability of TOPK and promotes tumorigenesis of colon. Oncotarget 7(17):24483 Song Y, Wu F, Wu J, 2016, Targeting histone methylation for cancer therapy: enzymes, inhibitors, biological activity and perspectives. J Hematol Oncol 9(1) Hsu H, Liu Y, Tseng K, Yang T, Yeh C, You J, Hung H, Chen S, Chen H, 2015, CBB1003, a lysine-specific demethylase 1 inhibitor, suppresses colorectal cancer cells growth through downregulation of leucine-rich repeat-containing G-protein-coupled receptor 5 expression. J Cancer Res Clin Oncol 141(1):11–21 Takawa M, Masuda K, Kunizaki M, Daigo Y, Takagi K, Iwai Y, Cho H, Toyokawa G, Yamane Y, Maejima K et al, 2011, Validation of the histone methyltransferase EZH2 as a therapeutic target for various types of human cancer and as a prognostic marker. Cancer Sci 102(7):1298–1305 Ying L, Yan F, Williams BR, Xu P, Li X, Zhao Y, Hu Y, Wang Y, Xu D, Dai J, 2018,, −)-Epigallocatechin-3-gallate and EZH2 inhibitor GSK343 have similar inhibitory effects andmechanisms of action on colorectal cancer cells. Clin Exp Pharmacol Physiol 45(1):58–67 Hsieh YY, Lo HL, Yang PM, 2016, EZH2 inhibitors transcriptionally upregulate cytotoxic autophagy and cytoprotective unfolded protein response in human colorectal cancer cells. Am J Cancer Res 6(8):1661–1680 Paschall AV, Yang D, Lu C, Choi JH, Li X, Liu F, Figueroa M, Oberlies NH, Pearce C, Bollag WB et al, 2015, H3K9 Trimethylation silences Fas expression to confer Colon carcinoma immune escape and 5-fluorouracil Chemoresistance. J Immunol 195(4):1868–1882 Kim MS, Cho HI, Yoon HJ, Ahn Y, Park EJ, Jin YH, Jang YK, 2018, JIB-04, a small molecule histone demethylase inhibitor, selectively targets colorectal cancer stem cells by inhibiting the Wnt/β-catenin signaling pathway. Sci Rep 8(1) Sonnemann J, Marx C, Becker S, Wittig S, Palani CD, Kramer OH, Beck JF, 2014, p53-dependent and p53-independent anticancer effects of different histone deacetylase inhibitors. Br J Cancer 110(3):656–667 Gargalionis AN, Piperi C, Adamopoulos C, Papavassiliou AG, 2012, Histone modifications as a pathogenic mechanism of colorectal tumorigenesis. Int J Biochem Cell Biol 44(8):1276–1289 Liu K, Wang L, Hsu S, 2018, Modification of epigenetic histone acetylation in hepatocellular carcinoma. Cancers 10(1):8 Iyer SP, Foss FF, 2015, Romidepsin for the treatment of peripheral T-cell lymphoma. Oncologist 20(9):1084–1091 Foss F, Advani R, Duvic M, Hymes KB, Intragumtornchai T, Lekhakula A, Shpilberg O, Lerner A, Belt RJ, Jacobsen ED et al, 2015, A phase II trial of Belinostat, PXD101, in patients with relapsed or refractory peripheral or cutaneous T-cell lymphoma. Br J Haematol 168(6):811–819 Xiong H, Du W, Zhang Y, Hong J, Su W, Tang J, Wang Y, Lu R, Fang J, 2012, Trichostatin A, a histone deacetylase inhibitor, suppresses JAK2/STAT3 signaling via inducing the promoterassociated histone acetylation of SOCS1 and SOCS3 in human colorectal cancer cells. Mol Carcinog 51(2):174–184 Jin J, Tsao T, Sun P, Yu C, Tzao C, 2012, SAHA inhibits the growth of Colon tumors by decreasing histone deacetylase and the expression of cyclin D1 and Survivin. Pathol Oncol Res 18(3): 713–720 Humphreys KJ, Cobiac L, Le Leu RK, Van der Hoek MB, Michael MZ, 2013, Histone deacetylase inhibition in colorectal cancer cells reveals competing roles for members of the oncogenic miR-17-92 cluster. Mol Carcinog 52(6):459–474 Strey CW, Schamell L, Oppermann E, Haferkamp A, Bechstein WO, Blaheta RA, 2011, Valproate inhibits colon cancer growth through cell cyclemodification in vivo and in vitro. Exp Ther Med 2(2):301–307 Wang TY, Chai YR, Jia YL, Gao JH, Peng XJ, Han HF, 2016, Crosstalk among the proteome, lysine phosphorylation, and acetylation in romidepsin-treated colon cancer cells. Oncotarget 7(33): 53471–53501 Mizutani H, Hiraku Y, Tada-Oikawa S, Murata M, Ikemura K, Iwamoto T, Kagawa Y, Okuda M, Kawanishi S, 2010, Romidepsin, FK228), a potent histone deacetylase inhibitor, induces apoptosis through the generation of hydrogen peroxide. Cancer Sci 101(10):2214–2219 Li Q, Ding C, Meng T, Lu W, Liu W, Hao H, Cao L, 2017, Butyrate suppresses motility of colorectal cancer cells via deactivating Akt/ERK signaling in histone deacetylase dependent manner. J Pharmacol Sci 135(4):148–155 Bracker TU, Sommer A, Fichtner I, Faus H, Haendler B, Hess- Stumpp H, 2009, Efficacy of MS-275, a selective inhibitor of class I histone deacetylases, in human colon cancer models. Int J Oncol 35(4):909–920 Zhu S, Denman CJ, Cobanoglu ZS, Kiany S, Lau CC, Gottschalk SM, Hughes DPM, Kleinerman ES, Lee DA, 2015, The narrow- Spectrum HDAC inhibitor Entinostat enhances NKG2D expression without NK cell toxicity, leading to enhanced recognition of cancer cells. Pharm Res 32(3):779–792 Anantharaju PG, Reddy BD, Padukudru MA, Kumari Chitturi CM, Vimalambike MG, Madhunapantula SV, 2017, Naturally occurring benzoic acid derivatives retard cancer cell growth by inhibiting histone deacetylases, HDAC). Cancer Biol Ther 18(7):492–504 Beck HC, Petersen J, Nielsen SJ, Morsczeck C, Jensen PB, Sehested M, Grauslund M, 2010, Proteomic profiling of human colon cancer cells treated with the histone deacetylase inhibitor belinostat. Electrophoresis 31(16):2714–2721 Maschauer S, Gahr S, Gandesiri M, Tripal P, Schneider-Stock R, Kuwert T, Ocker M, Prante O, 2016, In vivo monitoring of the anti-angiogenic therapeutic effect of the pan-deacetylase inhibitor panobinostat by small animal PET in a mouse model of gastrointestinal cancers. Nucl Med Biol 43(1):27–34 LaBonte MJ, Wilson PM, Fazzone W, Groshen S, Lenz HJ, Ladner RD, 2009, DNA microarray profiling of genes differentially regulated by the histone deacetylase inhibitors vorinostat and LBH589 in colon cancer cell lines. BMC Med Genet 2:67 Iwamoto M, Friedman EJ, Sandhu P, Agrawal NGB, Rubin EH, Wagner JA, 2013, Clinical pharmacology profile of vorinostat, a histone deacetylase inhibitor. Cancer Chemother Pharmacol 72(3): 493–508 Hu Q, Baeg GH, 2017, Role of epigenome in tumorigenesis and drug resistance. Food Chem Toxicol 109(Pt 1):663–668 Papavassiliou KA, Papavassiliou AG, 2013, Histone deacetylases inhibitors: conjugation to other anti-tumour pharmacophores provides novel tools for cancer treatment. Expert Opin Investig Drugs 23(3):291–294 Won H, Ryu H, Shin D, Yeon S, Lee DH, Kwon SH, 2018, A452, an HDAC6-selective inhibitor, synergistically enhances the anticancer activity of chemotherapeutic agents in colorectal cancer cells. Mol Carcinog 57:1383–1395 Katona BW, Liu Y, Ma A, Jin J, Hua X, 2014, EZH2 inhibition enhances the efficacy of an EGFR inhibitor in suppressing colon cancer cells. Cancer Biol Ther 15(12):1677–1687 Pitts TM, Morrow M, Kaufman SA, Tentler JJ, Eckhardt SG, 2009, Vorinostat and bortezomib exert synergistic antiproliferative and proapoptotic effects in colon cancer cell models. Mol Cancer Ther 8(2):342–349 Encarnacao JC, Pires AS, Amaral RA, Goncalves TJ, Laranjo M, Casalta-Lopes JE, Goncalves AC, Sarmento-Ribeiro AB, Abrantes AM, Botelho MF, 2018, Butyrate, a dietary fiber derivative that improves irinotecan effect in colon cancer cells. J Nutr Biochem 56:183–192 Azad NS, El-Khoueiry A, Yin J, Oberg AL, Flynn P, Adkins D, Sharma A, Weisenberger DJ, Brown T, Medvari P et al, 2017, Combination epigenetic therapy in metastatic colorectal cancer, mCRC, with subcutaneous 5-azacitidine and entinostat: a phase 2 consortium/stand up 2 cancer study. Oncotarget 8(21):35326 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2023 EP 2033 DI 10.1007/s12253-019-00663-8 PG 11 ER PT J AU Kraboth, Z Kalman, B AF Kraboth, Zoltan Kalman, Bernadette TI Longitudinal Characteristics of Glioblastoma in Genome-Wide Studies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Glioblastoma; Sequential samples; Molecular evolution; Genomics; Transcriptomics; Epigenomics ID Glioblastoma; Sequential samples; Molecular evolution; Genomics; Transcriptomics; Epigenomics AB Glioblastoma is one of the deadliest tumors with barely over one-year median survival despite intensive efforts in defining its molecular characteristics and searching for innovative treatment strategies. While major progress has been made in cataloging cross-sectional genomic, transcriptomic and epigenomic features of the tumor, and inferring its main molecular pathways and niches for potential targeted intervention, we still do not have sufficient knowledge concerning evolutionary patterns and dynamics of molecular changes or the treatment-induced effects affecting glioblastoma biology. In this review, we summarize the results of recent longitudinal genomic, transcriptomic and epigenomic studies that brought us closer to a better understanding of this lethal disease. Evidence suggests that neuronal / glioma stem cells with accumulating mutations initiate glioblastoma development and recurrence, but the hypothetical models describing the courses that lead to established tumors have not been fully proven. Moving from the histopathological phenotype to the results of high resolution OMICS studies, we try to synthesize the currently available information from sequential glioblastoma analyses in order to highlight its multifaceted features and heterogeneity, as well as the expected complexity of potential treatment strategies that might once succeed. C1 [Kraboth, Zoltan] University of Pecs, Department of Neurology, 12. Szigeti street, 7624 Pecs, Hungary. [Kalman, Bernadette] University of Pecs, Department of Neurology, 12. Szigeti street, 7624 Pecs, Hungary. RP Kalman, B (reprint author), University of Pecs, Department of Neurology, 7624 Pecs, Hungary. EM Kalman.bernadett@markusovszky.hu;Bernadette.kalman@pte.hu CR Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P, 2007, The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114(2):97–109. , DOI 10.1007/s00401-007- 78-6 2. Salcman M, Ebert PS, 1991, In vitro response of human glioblastoma and canine glioma cells to hyperthermia, radiation, and chemotherapy. Neurosurgery 29(4):526–531. , DOI 10.1227/ 00006123-199110000-00007 Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ et al, 2005, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl JMed 352(10):987– 996. , DOI 10.1056/NEJMoa043330 Kim H, Zheng S, Amini SS, Virk SM, Mikkelsen T, Brat DJ et al, 2015, Whole-genome and multisector exome sequencing of primary and post-treatment glioblastoma reveals patterns of tumor evolution. Genome Res 25(3):316–327 WellerM, Cloughesy T, Perry JR,WickW(2012, Standards of care for treatment of recurrent glioblastoma—are we there yet? Neuro- Oncol 15(1):4–27. , DOI 10.1093/neuonc/nos273 Stupp R, Taillibert S, Kanner A, ReadW, Steinberg DM, Lhermitte B, Toms S, Idbaih A, AhluwaliaMS, Fink K, di Meco F, Lieberman F, Zhu JJ, Stragliotto G, Tran DD, Brem S, Hottinger AF, Kirson ED, Lavy-Shahaf G, Weinberg U, Kim CY, Paek SH, Nicholas G, Bruna J, Hirte H,Weller M, Palti Y, HegiME, Ram Z, 2017, Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. JAMA 318(23):2306–2316. , DOI 10.1001/jama.2017.18718 Martinez R, Rohde V, Schackert G, 2010, Different molecular patterns in glioblastoma multiforme subtypes upon recurrence. J Neuro-Oncol 96(3):321–329. , DOI 10.1007/s11060-009- 9967-4 Network CGAR, 2008, Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455(7216):1061–1068. , DOI 10.1038/nature07385 Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP, Alexe G, Lawrence M, O'Kelly M, Tamayo P,Weir BA, Gabriel S,WincklerW, Gupta S, Jakkula L, Feiler HS, Hodgson JG, James CD, Sarkaria JN, Brennan C, Kahn A, Spellman PT, Wilson RK, Speed TP, Gray JW, Meyerson M, Getz G, Perou CM, Hayes DN, Cancer Genome Atlas Research Network, 2010, Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17(1):98–110. , DOI 10.1016/j.ccr.2009.12. 020 Wang Q, Hu B, Hu X,Kim H, SquatritoM, Scarpace L, deCarvalho AC, Lyu S, Li P, Li Y, Barthel F, Cho HJ, Lin YH, Satani N, Martinez-Ledesma E, Zheng S, Chang E, Gabriel Sauve CE, Olar A, Lan ZD, Finocchiaro G, Phillips JJ, Berger MS, Gabrusiewicz KR,Wang G, Eskilsson E, Hu J,Mikkelsen T, DePinho RA, Muller F, Heimberger AB, Sulman EP, Nam DH, Verhaak RGW, 2017, Tumor evolution of glioma-intrinsic gene expression subtypes associates with immunological changes in the microenvironment. Cancer Cell 32(1):42–56. , DOI 10.1016/j.ccell.2017.12. 012 Noushmehr H, Weisenberger DJ, Diefes K, Phillips HS, Pujara K, Berman BP, Pan F, Pelloski CE, Sulman EP, Bhat KP, Verhaak RG, Hoadley KA, Hayes DN, Perou CM, Schmidt HK, Ding L,Wilson RK, van den Berg D, Shen H, Bengtsson H, Neuvial P, Cope LM, Buckley J, Herman JG, Baylin SB, Laird PW, Aldape K, Cancer Genome Atlas Research Network, 2010, Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17(5):510–522. , DOI 10.1016/j.ccr. 2010.03.017 Nagy A, Garzuly F, Padanyi G, Szucs I, Feldmann A, Murnyak B, Hortobagyi T, Kalman B, 2019, Molecular subgroups of glioblastoma–an assessment by immunohistochemical markers. Pathol Oncol Res 25(1):21–31. , DOI 10.1007/s12253- 017-0311-6 Ueki K, Ono Y, Henson JW, Efird JT, von Deimling A, Louis DN, 1996, CDKN2/p16 or RB alterations occur in the majority of glioblastomas and are inversely correlated. Cancer Res 56(1):150–153 SzerlipNJ, Pedraza A, Chakravarty D, AzimM,McGuire J, FangY et al, 2012). Intratumoral heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma defines subpopulations with distinct growth factor response. Proc Natl Acad Sci U.S.A 109(8), 3041–3046. , DOI 10.1073/pnas. 1114033109 Frattini V, Trifonov V, Chan JM, Castano A, Lia M, Abate F, Keir ST, Ji AX, Zoppoli P, Niola F, Danussi C, Dolgalev I, Porrati P, Pellegatta S, Heguy A, Gupta G, Pisapia DJ, Canoll P, Bruce JN, McLendon RE, Yan H, Aldape K, Finocchiaro G, Mikkelsen T, Prive GG, Bigner DD, Lasorella A, Rabadan R, Iavarone A, 2013, The integrated landscape of driver genomic alterations in glioblastoma. Nat Genet 45(10):1141–1149. , DOI 10. 1038/ng.2734 Johnson BE, Mazor T, Hong C, Barnes M, Aihara K, McLean CY et al, 2014, Mutational analysis reveals the origin and therapydriven evolution of recurrent glioma. Science 343(6167):189–193. , DOI 10.1126/science.1239947 van den Bent MJ, Gao Y, Kerkhof M, Kros JM, Gorlia T, van Zwieten K, Prince J, van Duinen S, Sillevis Smitt PA, Taphoorn M, French PJ, 2015, Changes in the EGFR amplification and EGFRvIII expression between paired primary and recurrent glioblastomas. Neuro-Oncol 17(7):935–941. , DOI 10.1093/ neuonc/nov013 Neilsen BK, Sleightholm R, McComb R, Ramkissoon SH, Ross JS, Corona RJ, Miller VA, Cooke M, Aizenberg MR, 2018, Comprehensive genetic alteration profiling in primary and recurrent glioblastoma. J Neuro-Oncol 142:1–8. , DOI 10.1007/ s11060-018-03070-2 Wang J, Cazzato E, Ladewig E, Frattini V, Rosenbloom DI, Zairis S et al, 2016, Clonal evolution of glioblastoma under therapy. Nat Genet 48(7):768–776. , DOI 10.1038/ng.3590 Kim J, Lee IH, Cho HJ, Park CK, Jung YS, Kim Y, Nam SH, Kim BS, JohnsonMD, KongDS, Seol HJ, Lee JI, Joo KM,Yoon Y, Park WY, Lee J, Park PJ, Nam DH, 2015, Spatiotemporal evolution of the primary glioblastoma genome. Cancer Cell 28(3):318–328. , DOI 10.1016/j.ccell.2015.07.013 Muscat AM, Wong NC, Drummond KJ, Algar EM, Khasraw M, Verhaak R et al, 2018, The evolutionary pattern of mutations in glioblastoma reveals therapy-mediated selection. Oncotarget 9(8): 7844. , DOI 10.18632/oncotarget.23541 Hegi ME, Diserens AC, Gorlia T, Hamou MF, De Tribolet N, Weller M et al, 2005, MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 352(10):997–1003. , DOI 10.1056/NEJMoa043331 Nagarajan RP, Zhang B, Bell RJ, Johnson BE, Olshen AB, SundaramVet al, 2014, Recurrent epimutations activate gene body promoters in primary glioblastoma. Genome Res 24(5):761–774 http://www.genome.org/cgi/doi/10.1101/gr.164707.113 de Souza CF, Sabedot TS, Malta TM, Stetson L, Morozova O, Sokolov A, Laird PW, Wiznerowicz M, Iavarone A, Snyder J, deCarvalho A, Sanborn Z, McDonald KL, Friedman WA, Tirapelli D, Poisson L, Mikkelsen T, Carlotti CG Jr, Kalkanis S, Zenklusen J, Salama SR, Barnholtz-Sloan JS, Noushmehr H, 2018, A distinct DNA methylation shift in a subset of glioma CpG island methylator phenotypes during tumor recurrence. Cell Rep 23(2): 637–651. , DOI 10.1016/j.celrep.2018.03.107 Klughammer J, Kiesel B, Roetzer T, Fortelny N, Nemc A, Nenning KH, Furtner J, Sheffield NC, Datlinger P, Peter N, Nowosielski M, Augustin M, Mischkulnig M, Strobel T, Alpar D, Erguner B, Senekowitsch M, Moser P, Freyschlag CF, Kerschbaumer J, Thome C, Grams AE, Stockhammer G, Kitzwoegerer M, Oberndorfer S, Marhold F, Weis S, Trenkler J, Buchroithner J, Pichler J, Haybaeck J, Krassnig S, Mahdy Ali K, von Campe G, Payer F, Sherif C, Preiser J, Hauser T, Winkler PA, Kleindienst W, Wurtz F, Brandner-Kokalj T, Stultschnig M, Schweiger S, Dieckmann K, Preusser M, Langs G, Baumann B, Knosp E, Widhalm G, Marosi C, Hainfellner JA, Woehrer A, Bock C, 2018, The DNA methylation landscape of glioblastoma disease progression shows extensive heterogeneity in time and space. Nat Med 24(10):1611–1624. , DOI 10.1038/s41591-018- 0156-x Ferreira WAS, Pinheiro DDR, Costa Junior CAD, Rodrigues- Antunes S, Araujo MD, Leao Barros MB et al, 2016, An update on the epigenetics of glioblastomas. Epigenomics 8(9):1289–1305. , DOI 10.2217/epi-2016-0040 Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, Cahill DP, Nahed BV, Curry WT, Martuza RL, Louis DN, Rozenblatt-Rosen O, Suva ML, Regev A, Bernstein BE, 2014, Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344(6190):1396– 1401. , DOI 10.1126/science.1254257 Gill BJ, Pisapia DJ, Malone HR, Goldstein H, Lei L, Sonabend A et al, 2014, MRI-localized biopsies reveal subtype-specific differences in molecular and cellular composition at the margins of glioblastoma. Proc Natl Acad Sci U.S.A 111(34):12550–12555. https:// doi.org/10.1073/pnas.1405839111 Tompa M, Kalovits F, Nagy A, Kalman B, 2018, Contribution of the Wnt pathway to defining biology of glioblastoma. NeuroMolecular Med 20:1–15. , DOI 10.1007/s12017- 018-8514-x Birbrair A, Sattiraju A, Zhu D, Zulato G, Batista I, Nguyen VT, Messi ML, Solingapuram Sai KK,Marini FC, Delbono O, Mintz A, 2017, Novel peripherally derived neural-like stem cells as therapeutic carriers for treating glioblastomas. Stem Cells Transl Med 6(2):471–481. , DOI 10.5966/sctm.2016-0007 Orzan F, De Bacco F, Crisafulli G, Pellegatta S, Mussolin B, Siravegna G et al, 2017, Genetic evolution of glioblastoma stemlike cells fromprimary to recurrent tumor. Stem Cells 35(11):2218– 2228. , DOI 10.1002/stem.2703 Marucci G, Fabbri PV, Morandi L, De Biase D, Di Oto E, Tallini G et al, 2015, Pathological spectrum in recurrences of glioblastoma multiforme. Pathologica 107(1):1–8 Stark AM, Witzel P, Strege RJ, Hugo HH, Mehdorn HM, 2003, p53, mdm2, EGFR, and msh2 expression in paired initial and recurrent glioblastoma multiforme. J Neurol Neurosurg Psychiatry 74(6):779–783. , DOI 10.1136/jnnp.74.6.779 Stark AM, Doukas A, Hugo HH,Mehdorn HM(2010, The expression of mismatch repair proteins MLH1, MSH2 and MSH6 correlates with the Ki67 proliferation index and survival in patients with recurrent glioblastoma. Neurol Res 32(8):816–820. , DOI 10.1179/016164110X12645013515052 Shinsato Y, Furukawa T, Yunoue S, Yonezawa H, Minami K, Nishizawa Y et al, 2013, Reduction of MLH1 and PMS2 confers temozolomide resistance and is associated with recurrence of glioblastoma. Oncotarget 4(12):2261. , DOI 10.18632/ oncotarget.1302 Horgan RP, Kenny LC, 2011, ‘Omic’technologies: genomics, transcriptomics, proteomics and metabolomics. Obstet Gynecol 13(3): 189–195. , DOI 10.1576/toag.13.3.189.27672 Nagarajan RP, Costello JF, 2009, June, Epigenetic mechanisms in glioblastoma multiforme. In Seminars in cancer biology, Vol. 19, no. 3, pp. 188-197). Academic press. , DOI 10.1016/j. semcancer.2009.02.005 Clark SJ, Harrison J, Frommer M, 1995, CpNpG methylation in mammalian cells. Nat Genet 10(1):20–27. , DOI 10.1038/ ng0595-20 Okano M, Xie S, Li E, 1998, Cloning and characterization of a family of novel mammalian DNA, cytosine-5, methyltransferases. Nat Genet 19(3):219–220. , DOI 10.1038/890 Li E, Beard C, Jaenisch R, 1993, Role for DNA methylation in genomic imprinting. Nature 366(6453):362–365. , DOI 10.1038/366362a0 Turner BM, 2005, Reading signals on the nucleosome with a new nomenclature for modified histones. Nat Struct Mol Biol 12(2): 110–112. , DOI 10.1038/nsmb0205-110 Garzon R, Calin GA, Croce CM, 2009, MicroRNAs in cancer. Annu Rev Med 60:167–179. , DOI 10.1146/annurev.med. 59.053006.104707 Sottoriva A, Spiteri I, Piccirillo SG, Touloumis A, Collins VP, Marioni JC et al, 2013, Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc Natl Acad Sci U.S.A 110(10):4009–4014. , DOI 10.1073/pnas. 1219747110 Offer H, Erez N, Zurer I, Tang X, Milyavsky M, Goldfinger N, Rotter V, 2002, The onset of p53-dependent DNA repair or apoptosis is determined by the level of accumulated damaged DNA. Carcinogenesis 23(6):1025–1032. , DOI 10.1093/ carcin/23.6.1025 Kojima K, Konopleva M, Samudio IJ, Shikami M, Cabreira- Hansen M, McQueen T, Ruvolo V, Tsao T, Zeng Z, Vassilev LT, Andreeff M, 2005, MDM2 antagonists induce p53-dependent apoptosis in AML: implications for leukemia therapy. Blood 106(9): 3150–3159. , DOI 10.1182/blood-2005-02-0553 Lynch M, 2010, Rate, molecular spectrum, and consequences of human mutation. Proc Natl Acad Sci U.S.A 107(3):961–968. , DOI 10.1073/pnas.0912629107 Llaguno SA, Sun D, Pedraza AM, Vera E, Wang Z, Burns DK, Parada LF, 2019, Cell-of-origin susceptibility to glioblastoma formation declines with neural lineage restriction. Nat Neurosci 1: 545–555. , DOI 10.1038/s41593-018-0333-8 Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, Hau P, Brandes AA, Gijtenbeek J, Marosi C, Vecht CJ, Mokhtari K, Wesseling P, Villa S, Eisenhauer E, Gorlia T, Weller M, Lacombe D, Cairncross JG, Mirimanoff RO, European Organisation for Research and Treatment of Cancer Brain Tumour and Radiation Oncology Groups., National Cancer Institute of Canada Clinical Trials Group, 2009, Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet 10:459–466. , DOI 10. 1016/S1470-2045(09)70025-7 Clarke J, Butowski N, Chang S, 2010, Recent advances in therapy for glioblastoma. Arch Neurol 67(3):279–283. , DOI 10. 1001/archneurol.2010.5 Hou LC, Veeravagu A, Hsu AR, Victor CK, 2006, Recurrent glioblastoma multiforme: a review of natural history and management options. Neurosurg Focus 20(4):E3. , DOI 10.3171/foc. 2006.20.4.2 Nickel GC, Barnholtz-Sloan J, Gould MP, McMahon S, Cohen A, Adams MD, Guda K, Cohen M, Sloan AE, LaFramboise T, 2012, Characterizing mutational heterogeneity in a glioblastoma patient with double recurrence. PLoS One 7(4):e35262. , DOI 10. 1371/journal.pone.0035262 Andor N, Harness JV, Mueller S, Mewes HW et al, 2013, EXPANDS: expanding ploidy and allele frequency on nested subpopulations. Bioinformatics 30(1):50–60. , DOI 10.1093/ bioinformatics/btt622 Erson-Omay EZ, Henegariu O, Omay SB, Harmanci AS, Youngblood MW, Mishra-Gorur K, Li J, Ozduman K, Carrion- Grant G, Clark VE, Caglar C, Bakircioglu M, Pamir MN, Tabar V, Vortmeyer AO, Bilguvar K, Yasuno K, DeAngelis LM, Baehring JM, Moliterno J, Gunel M, 2017, Longitudinal analysis of treatment-induced genomic alterations in gliomas. Genome Med 9(1):12. , DOI 10.1186/s13073-017-0401-9 Forment JV, Kaidi A, Jackson SP, 2012, Chromothripsis and cancer: causes and consequences of chromosome shattering. Nat Rev Cancer 12(10):663–670. , DOI 10.1038/nrc3352 L'abbate A, Macchia G, D'addabbo P, Lonoce A, Tolomeo D, Trombetta D et al, 2014, Genomic organization and evolution of double minutes/homogeneously staining regions with MYC amplification in human cancer. Nucleic Acids Res 42(14):9131–9145. , DOI 10.1093/nar/gku590 Nathanson DA, Gini B, Mottahedeh J, Visnyei K, Koga T, Gomez G, Eskin A, Hwang K,Wang J,Masui K, Paucar A, Yang H, Ohashi M, Zhu S, Wykosky J, Reed R, Nelson SF, Cloughesy TF, James CD, Rao PN, Kornblum HI, Heath JR, Cavenee WK, Furnari FB, Mischel PS, 2014, Targeted therapy resistancemediated by dynamic regulation of extrachromosomal mutant EGFR DNA. Science 343(6166):72–76. , DOI 10.1126/science.1241328 Le DT, Uram JN,Wang H, Bartlett BR, Kemberling H, Eyring AD et al, 2015, PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 372(26):2509–2520. , DOI 10.1056/ NEJMoa1500596 Cloughesy TF, Mochizuki AY, Orpilla JR, Hugo W, Lee AH, Davidson TB, Wang AC, Ellingson BM, Rytlewski JA, Sanders CM, Kawaguchi ES, du L, Li G, Yong WH, Gaffey SC, Cohen AL, Mellinghoff IK, Lee EQ, Reardon DA, O’Brien BJ, Butowski NA, Nghiemphu PL, Clarke JL, Arrillaga-Romany IC, Colman H, Kaley TJ, de Groot JF, Liau LM, Wen PY, Prins RM, 2019, Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med 1:477–486. , DOI 10.1038/s41591- 018-0337-7 Zhao J, Chen AX, Gartrell RD, Silverman AM, Aparicio L, Chu T, Bordbar D, Shan D, Samanamud J, Mahajan A, Filip I, Orenbuch R, Goetz M, Yamaguchi JT, Cloney M, Horbinski C, Lukas RV, Raizer J, Rae AI, Yuan J, Canoll P, Bruce JN, Saenger YM, Sims P, Iwamoto FM, Sonabend AM, Rabadan R, 2019, Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat Med 1:462–469. , DOI 10.1038/s41591- 019-0349-y Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R, Lopez-Janeiro A, Porciuncula A, Idoate MA, Inoges S, de Andrea C, Lopez-Diaz de Cerio A, Tejada S, Berraondo P, Villarroel-Espindola F, Choi J, Gurpide A, Giraldez M, Goicoechea I, Gallego Perez-Larraya J, Sanmamed MF, Perez-Gracia JL, Melero I, 2019, Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat Med 1:470–476. , DOI 10. 1038/s41591-018-0339-5 Campos B, Olsen LR, Urup T, Poulsen HS, 2016, A comprehensive profile of recurrent glioblastoma. Oncogene 35(45):5819– 5825. , DOI 10.1038/onc.2016.85 Li R, Chen X, You Y, Wang X, Liu Y, Hu Q, Yan W, 2015, Comprehensive portrait of recurrent glioblastoma multiforme in molecular and clinical characteristics. Oncotarget 6(31):30968. , DOI 10.18632/oncotarget.5038 Kim H, Verhaak RG, 2015, Transcriptional mimicry by tumorassociated stroma. Nat Genet 47(4):307–309. , DOI 10. 1038/ng.3255 Murat A, Migliavacca E, Gorlia T, Lambiv WL, Shay T, Hamou MF, de Tribolet N, Regli L, Wick W, Kouwenhoven MCM, Hainfellner JA, Heppner FL, Dietrich PY, Zimmer Y, Cairncross JG, Janzer RC, Domany E, Delorenzi M, Stupp R, Hegi ME, 2008, Stem cell–related “self-renewal” signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma. J Clin Oncol 26(18): 3015–3024. , DOI 10.1200/JCO.2007.15.7164 Chen X, Wen Q, Stucky A, Zeng Y, Gao S, Loudon WG, Ho HW, KabeerMH, Li SC, Zhang X, Zhong JF, 2018, Relapse pathway of glioblastoma revealed by single-cell molecular analysis. Carcinogenesis 39(7):931–936. , DOI 10.1093/carcin/ bgy052 Lee HK, Finniss S, Cazacu S, Xiang C, PoissonLM, Blumberg PM et al, 2015, RasGRP3 regulates the migration of glioma cells via interaction with Arp3. Oncotarget 6:1850–1864. , DOI 10. 18632/oncotarget.2575 Laird PW, 2003, Early detection: the power and the promise of DNA methylation markers. Nat Rev Cancer 3(4):253–266. https:// doi.org/10.1038/nrc1045 BrennanCW,Verhaak RG,McKenna A, Campos B, Noushmehr H, Salama SR et al, 2013, The somatic genomic landscape of glioblastoma. Cell 155(2):462–477. , DOI 10.1016/j.cell.2013.09. 034 Capper D, Jones DT, Sill M, Hovestadt V, Schrimpf D, Sturm D et al, 2018, DNA methylation-based classification of central nervous system tumours. Nature 555(7697):469–474. https://doi. org/10.1038/nature26000 Ceccarelli M, Barthel FP, Malta TM, Sabedot TS, Salama SR, Murray BA, Morozova O, Newton Y, Radenbaugh A, Pagnotta SM, Anjum S, Wang J, Manyam G, Zoppoli P, Ling S, Rao AA, Grifford M, Cherniack AD, Zhang H, Poisson L, Carlotti Jr CG, Tirapelli DP C, Rao A, Mikkelsen T, Lau CC, Yung WKA, Rabadan R, Huse J, Brat DJ, Lehman NL, Barnholtz-Sloan JS, Zheng S, Hess K, Rao G, Meyerson M, Beroukhim R, Cooper L, Akbani R, Wrensch M, Haussler D, Aldape KD, Laird PW, Gutmann DH, Noushmehr H, Iavarone A, Verhaak RGW, Anjum S, Arachchi H, Auman JT, Balasundaram M, Balu S, Barnett G, Baylin S, Bell S, Benz C, Bir N, Black KL, Bodenheimer T, Boice L, Bootwalla MS, Bowen J, Bristow CA, Butterfield YSN, Chen QR, Chin L, Cho J, Chuah E, Chudamani S, Coetzee SG, Cohen ML, Colman H, Couce M, D’Angelo F, Davidsen T, Davis A, Demchok JA, Devine K, Ding L, Duell R, Elder JB, Eschbacher JM, Fehrenbach A, Ferguson M, Frazer S, Fuller G, Fulop J, Gabriel SB, Garofano L, Gastier-Foster JM, Gehlenborg N, Gerken M, Getz G, Giannini C, Gibson WJ, Hadjipanayis A, Hayes DN, Heiman DI, Hermes B, Hilty J, Hoadley KA, Hoyle AP, Huang M, Jefferys SR, Jones CD, Jones SJM, Ju Z, Kastl A, Kendler A, Kim J, Kucherlapati R, Lai PH, Lawrence MS, Lee S, LeraasKM, Lichtenberg TM, Lin P, Liu Y, Liu J, Ljubimova JY, Lu Y,Ma Y,Maglinte DT, Mahadeshwar HS, MarraMA,McGraw M, McPherson C, Meng S, Mieczkowski PA, Miller CR, Mills GB, Moore RA, Mose LE, Mungall AJ, Naresh R, Naska T, Neder L, Noble MS, Noss A, O’Neill BP, Ostrom QT, Palmer C, Pantazi A, Parfenov M, Park PJ, Parker JS, Perou CM, Pierson CR, Pihl T, Protopopov A, Radenbaugh A,s Ramirez NC, Rathmell WK, Ren X, Roach J, RobertsonAG, Saksena G, Schein JE, Schumacher SE, Seidman J, Senecal K, Seth S, Shen H, Shi Y, Shih J, Shimmel K, Sicotte H, Sifri S, Silva T, Simons JV, Singh R, Skelly T, Sloan AE, Sofia HJ, Soloway MG, Song X, Sougnez C, Souza C, Staugaitis SM, Sun H, Sun C, Tan D, Tang J, Tang Y, Thorne L, Trevisan FA, Triche T, van den Berg DJ, Veluvolu U, Voet D, Wan Y, Wang Z, Warnick R, Weinstein JN, Weisenberger DJ, Wilkerson MD, Williams F, Wise L, Wolinsky Y, Wu J, Xu AW, Yang L, Yang L, Zack TI, Zenklusen JC, Zhang J, Zhang W, Zhang J, Zmuda E, 2016). Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell 164(3), 550– 563. , DOI 10.1016/j.cell.2015.12.028 Sturm D, Witt H, Hovestadt V, Khuong-Quang DA, Jones DT, Konermann C et al, 2012, Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell 22(4):425–437. , DOI 10.1016/j.ccr. 2012.08.024 McCord M, Mukouyama YS, Gilbert MR, Jackson S, 2017, Targeting WNT signaling for multifaceted glioblastoma therapy. Front Cell Neurosci 11:318. , DOI 10.3389/fncel.2017. 00318 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2035 EP 2047 DI 10.1007/s12253-019-00705-1 PG 13 ER PT J AU Raffone, A Travaglino, A Santoro, A Esposito, I Angelico, G Spadola, S Zannoni, FG AF Raffone, Antonio Travaglino, Antonio Santoro, Angela Esposito, Italia Angelico, Giuseppe Spadola, Saveria Zannoni, Franco Gian TI Accuracy of One-Step Nucleic Acid Amplification in Detecting Lymph Node Metastases in Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE OSNA; Sentinel lymph node; Endometrial cancer; Intraoperative; Ultrastaging; Tailored medicine ID OSNA; Sentinel lymph node; Endometrial cancer; Intraoperative; Ultrastaging; Tailored medicine AB One-step nucleic acid amplification (OSNA) is used to intraoperatively detect sentinel lymph node metastases in breast cancer. OSNA has also been proposed in endometrial cancer, but evidence in this regard is unclear to define the diagnostic accuracy of OSNA in detecting lymph node metastases in endometrial cancer. A systematic review and meta-analysis was performed by searching 8 electronic databases from their inception to March 2019 for studies testing the diagnostic accuracy of OSNA in detecting sentinel lymph node metastasis in endometrial cancer. Pathologic ultrastaging was the reference standard. Sensitivity, specificity, positive and negative likelihood ratio (LR+ and LR-), diagnostic odds ratio (DOR) and area under the curve (AUC) on SROC curve were calculated. Four studies with 237 patients and 691 lymph nodes were included. OSNA showed sensitivity = 0.88, specificity = 0.93, LR + =17.95, LR- = 0.15, DOR = 191.23 and high diagnostic accuracy (AUC = 0.959). OSNA appears as a highly accurate tool for intraoperative assessment of sentinel lymph node in endometrial cancer. C1 [Raffone, Antonio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Travaglino, Antonio] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology UnitNaples, Italy. [Santoro, Angela] Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic, Pathology Unit, Department of Woman and Child HealthRome, Italy. [Esposito, Italia] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Angelico, Giuseppe] Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic, Pathology Unit, Department of Woman and Child HealthRome, Italy. [Spadola, Saveria] Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic, Pathology Unit, Department of Woman and Child HealthRome, Italy. [Zannoni, Franco Gian] Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic, Pathology Unit, Department of Woman and Child HealthRome, Italy. RP Raffone, A (reprint author), University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, 80131 Naples, Italy. EM anton.raffone@gmail.com CR Globocan, Northern America, WHO Europe Region, EURO). Estimated cancer incidence, all ages: female. 2012. Accessed January 23, 2017 Travaglino A, Raffone A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2018, PTEN as a predictive marker of response to conservative treatment in endometrial hyperplasia and early endometrial cancer. A systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol 231:104–110 Travaglino A, RaffoneA, Saccone G, de Luca C,Mollo A, Mascolo M, de Placido G, Insabato L, Zullo F, 2019, Immunohistochemical nuclear expression of β-catenin as a surrogate of CTNNB1 exon 3 mutation in endometrial Cancer. Am J Clin Pathol 151(5):529–538 Travaglino A, Raffone A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2019, Immunohistochemical predictive markers of response to conservative treatment of endometrial hyperplasia and early endometrial cancer: a systematic review. Acta Obstet Gynecol Scand. , DOI 10.1111/aogs.13587 [Epub ahead of print] Giampaolino P, Di Spiezio SA, Mollo A et al, 2019, Hysteroscopic Endometrial Focal Resection followed by Levonorgestrel Intrauterine Device Insertion as a Fertility-Sparing Treatment of Atypical Endometrial Hyperplasia and Early Endometrial Cancer: A Retrospective Study. J Minim Invasive Gynecol 26(4):648–656 Creasman W, 2009, Revised FIGO staging for carcinoma of the endometrium. Int J Gynaecol Obstet 105(2):109–109 Raffone A, Travaglino A, Saccone G, Mollo A, de Placido G, Insabato L, Zullo F, 2019, Should progesterone and estrogens receptors be assessed for predicting the response to conservative treatment of endometrial hyperplasia and cancer? A systematic review and meta-analysis. Acta Obstet Gynecol Scand 98:976–987. https:// doi.org/10.1111/aogs.13586. [Epub ahead of print] Lewin SN,Wright JD, 2011, Comparative performance of the 2009 international federation of gynecology and obstetrics’ staging system for uterine corpus cancer. Obstet Gynecol 117(5):1226 National Comprehensive Cancer Network, NCCN). NCCN Clinical Practice Guidelines in Oncology. NCCN Guidelines Version 2.2018 Uterine Neoplasms Colombo N, Creutzberg C, Amant F, Bosse T, Gonzalez-Martin A, Ledermann J, Marth C, Nout R, Querleu D, Mirza MR, Sessa C, ESMO-ESGO-ESTRO Endometrial Consensus Conference Working Group, 2016, ESMO-ESGO-ESTRO consensus conference on endometrial Cancer: diagnosis, treatment and followup. Int J Gynecol Cancer 26(1):2–30 Kitchener H, Swart AM, Qian Q, Amos C, Parmar MK, 2009, Efficacy of systematic pelvic lymphadenectomy in endometrial cancer, MRC ASTEC trial): a randomised study. Lancet, London, England, 373(9658):125–136 Chambers LM, Vargas R, Michener CM, 2019, Sentinel lymph node mapping in endometrial and cervical cancer: a survey of practices and attitudes in gynecologic oncologists. J Gynecol Oncol 30(3):e35 Lin H, Ding Z, Kota VG, Zhang X, Zhou J, 2017, Sentinel lymph node mapping in endometrial cancer: a systematic review and metaanalysis. Oncotarget. 8(28):46601–46610 Bodurtha Smith AJ, Fader AN, Tanner EJ, 2017, Sentinel lymph node assessment in endometrial cancer: a systematic review and meta-analysis. Am J Obstet Gynecol 216(5):459–476.e10 Ruscito I, Gasparri ML, Braicu EI, Bellati F, Raio L, Sehouli J, Mueller MD, Panici PB, Papadia A, 2016, Sentinel node mapping in cervical and endometrial Cancer: Indocyanine green versus other conventional dyes-a meta-analysis. Ann Surg Oncol 23(11):3749– 3756 Ansari M, Rad MA, Hassanzadeh M, Gholami H, Yousefi Z, Dabbagh VR, Sadeghi R, 2013, Sentinel node biopsy in endometrial cancer: systematic review and meta-analysis of the literature. Eur J Gynaecol Oncol 34(5):387–401 How JA, O'Farrell P, Amajoud Z, Lau S, Salvador S, How E, Gotlieb WH, 2018, Sentinel lymph node mapping in endometrial cancer: a systematic review and meta-analysis. Minerva Ginecol 70(2):194–214 Darai E, Dubernard G, Bats AS, Heitz D, Mathevet P, Marret H, Querleu D, Golfier F, Leblanc E, Rouzier R, Ballester M, 2015, Sentinel node biopsy for themanagement of early stage endometrial cancer: long-term results of the SENTI-ENDO study. Gynecol Oncol 136(1):54–59 Bezu C, Coutant C, Ballester M, Feron JG, Rouzier R, Uzan S, Darai E, 2010, Ultrastaging of lymph node in uterine cancers. J Exp Clin Cancer Res 29:5 Torne A, Puig-Tintore LM, 2004, The use of sentinel lymph nodes in gynaecological malignancies. Curr Opin Obstet Gynecol 16(1): 57–64 Holloway RW, Abu-Rustum NR, Backes FJ, Boggess JF, Gotlieb WH, Jeffrey Lowery W, Rossi EC, Tanner EJ, Wolsky RJ, 2017, Sentinel lymph node mapping and staging in endometrial Cancer: a Society of Gynecologic Oncology Literature Review with consensus recommendations. Gynecol Oncol 146(2):405–415 Kostun J, Pesta M, Slama J, Slunecko R, Vlasak P, Bouda J, Novotny Z, Topolcan O, Kucera R, Kulda V, Houfkova K, Berezovskiy D, Bartakova A, Presl J, 2019, One-step nucleic acid amplification vs ultrastaging in the detection of sentinel lymph node metastasis in endometrial cancer patients. J Surg Oncol 119(3): 361–369 Slama J, Dundr P, Dusek L, Cibula D, 2013, High false negative rate of frozen section examination of sentinel lymph nodes in patients with cervical cancer. Gynecol Oncol 129(2):384–388 Zhou M, Wang X, Jiang L, Chen X, Bao X, Chen X, 2018, The diagnostic value of one step nucleic acid amplification, OSNA, in differentiating lymph node metastasis of tumors: a systematic review and meta-analysis. Int J Surg 56:49–56 Shi F, Liang Z, Zhang Q, Wang C, Liu X, 2018, The performance of one-step nucleic acid amplification assay for intraoperative detection of sentinel lymph node macrometastasis in breast cancer: an updated meta-analysis. Breast. 39:39–45 Shi F, Zhang Q, Liang Z, ZhangM, Liu X, 2017, One-step nucleic acid amplification assay is an accurate technique for sentinel lymph node biopsy of breast cancer patients: a meta-analysis. Br J Cancer 117(8):1185–1191 Nagai T, Niikura H, Okamoto S, Nakabayashi K, Matoda M, Utsunomiya H, Nagase S, Watanabe M, Takeshima N, Yaegashi N, 2015, A new diagnostic method for rapid detection of lymph node metastases using a one-step nucleic acid amplification, OSNA, assay in endometrial cancer. Ann Surg Oncol 22(3):980– 986 Raffone A, Travaglino A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2019, Endometrial hyperplasia and progression to cancer: which classification system stratifies the risk better? A systematic review and meta-analysis. Arch Gynecol Obstet 299(5): 1233–1242 Raffone A, Travaglino A, Saccone G, Viggiani M, Giampaolino P, Insabato L, Mollo A, de Placido G, Zullo F, 2019, PTEN expression in endometrial hyperplasia and risk of cancer: a systematic review and meta-analysis. Arch Gynecol Obstet 299(6):1511–1524 Travaglino A, Raffone A, Saccone G, Mollo A, de Placido G, Mascolo M, Insabato L, Zullo F, 2019, Complexity of glandular architecture should be reconsidered in the classification and management of endometrial hyperplasia. APMIS. 127(6):427–434 Raffone A, Travaglino A, Saccone G, Alviggi C, Mascolo M, de Placido G, Insabato L, Mollo A, Zullo F, 2019, Management of women with atypical polypoid adenomyoma of the uterus: a quantitative systematic review. Acta Obstet Gynecol Scand 98(7):842– 855 Moher D, Shamseer L, Clarke M et al, 2015, Preferred reporting items for systematic review and meta-analysis protocols, PRISMAP, 2015 statement. Syst Rev 4:1 Sotiriadis A, Papatheodorou SI, Martins WP, 2016, Synthesizing evidence from diagnostic accuracy tests: the SEDATE guideline. Ultrasound Obstet Gynecol 47(3):386–395 Travaglino A, Raffone A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2018, Loss of B-cell lymphoma 2 immunohistochemical expression in endometrial hyperplasia: a specificmarker of precancer and novel indication for treatment: a systematic review and meta-analysis. Acta Obstet Gynecol Scand 97(12):1415–1426 Travaglino A, Raffone A, Saccone G, Mollo A, de Placido G, Insabato L, Zullo F, 2019, Endometrial hyperplasia and risk of coexistent cancer: WHO vs EIN criteria. Histopathology 74(5): 676–687 Raffone A, Travaglino A, Saccone G, Mascolo M, Insabato L, Mollo A, de Placido G, Zullo F, 2019, PAX2 in endometrial carcinogenesis and in differential diagnosis of endometrial hyperplasia. A systematic review andmeta-analysis of diagnostic accuracy. Acta Obstet Gynecol Scand 98(3):287–299 Raffone A, Travaglino A, Saccone G, CampaninoMR,Mollo A, de Placido G, Insabato L, Zullo F, 2019, Loss of PTEN expression as diagnostic marker of endometrial precancer: a systematic review and meta-analysis. Acta Obstet Gynecol Scand 98(3):275–286 Travaglino A, Raffone A, Saccone G, Mascolo M, Pignatiello S, Mollo A, de Placido G, Insabato L, Zullo F, 2019, PTEN immunohistochemistry in endometrial hyperplasia: which are the optimal criteria for the diagnosis of precancer? APMIS 127(4):161–169 Whiting PF, Rutjes AW, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, Leeflang MM, Sterne JA, Bossuyt PM, QUADAS-2 Group, 2011)QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med 155(8):529–536 Raffone A, Travaglino A, Saccone G, diMaio A,Mollo A, Mascolo M, de Rosa R, de Placido G, Insabato L, Zullo F, 2019, Diabetes mellitus and responsiveness of endometrial hyperplasia and early endometrial cancer to conservative treatment. Gynecol Endocrinol 5:1–6. , DOI 10.1080/09513590.2019.1624716 [Epub ahead of print] Raffone A, Travaglino A, Saccone G, D’Alessandro P, Arduino B, Mascolo M, de Placido G, Insabato L, Zullo F, 2019, Diabetes mellitus is associated with occult Cancer in endometrial hyperplasia. Pathol Oncol Res. , DOI 10.1007/s12253- 019-00684-3 [Epub ahead of print] Review Lopez-Ruiz ME, Diestro MD, Yebenes L, Berjon A, Diaz de la Noval B, Mendiola M, de Santiago J, Hardisson D, 2016, Onestep nucleic acid amplification, OSNA, for the detection of sentinel lymph node metastasis in endometrial cancer. Gynecol Oncol 143(1):54–59 Fanfani F, Monterossi G, Ghizzoni V, Rossi ED, Dinoi G, Inzani F, Fagotti A, Gueli Alletti S, Scarpellini F, Nero C, Santoro A, Scambia G, ZannoniGF, 2018, One-step nucleic acid amplification, OSNA): a fastmolecular test based on CK19mRNA concentration for assessment of lymph-nodes metastases in early stage endometrial cancer. PLoS One 13(4):e0195877 Fishman A, Klein A, Zemer R, Zimlichman S, Bernheim J, Cohen I, Altaras MM, 2000, Detection of micrometastasis by cytokeratin- 20, reverse transcription polymerase chain reaction, in lymph nodes of patients with endometrial cancer. Gynecol Oncol 77(3):399–404 Togami S, Kawamura T, Fukuda M, Yanazume S, Kamio M, Kobayashi H, 2019, Quantitative RT-PCR assay for detecting lymph node metastasis in endometrial Cancer: a preliminary study. Oncology 96(4):179–182 BallesterM, Naoura I, Chereau E, Seror J, Bats AS, Bricou A, Darai E, 2013, Sentinel node biopsy upstages patients with presumed low- and intermediate-risk endometrial cancer: results of a multicenter study. Ann Surg Oncol 20(2):407–412 Rozenholc A, Samouelian V, Warkus T et al, 2019, Green versus blue: Randomized controlled trial comparing indocyanine green with methylene blue for sentinel lymph node detection in endometrial cancer. Gynecol Oncol. , DOI 10.1016/j.ygyno.2019. 03.103 pii: S0090–8258(19)30296–3. [Epub ahead of print] Rossi EC, 2019, Current state of sentinel lymph nodes for women with endometrial cancer. Int J Gynecol Cancer 29(3):613–621 Liu LC, Lang JE, Lu Y, Roe D, Hwang SE, Ewing CA, Esserman LJ, Morita E, Treseler P, Leong SP, 2011, Intraoperative frozen section analysis of sentinel lymph nodes in breast cancer patients: a meta-analysis and single-institution experience. Cancer. 117(2): 250–258 Silveberg SG, 2007, The endometrium. Arch Pathol LabMed 131: 372–382 Tamaki Y, 2017, One-step nucleic acid amplification, OSNA): where do we go with it? Int J Clin Oncol 22(1):3–10 Americal Joint Committee on Cancer, AJCC, Cancer Staging Atlas. NY: Springer, 2017. 8th edition Raia-Barjat T, Trombert B, Khaddage A, Douchet C, Seffert P, Peoc’h M, Falk AT, Magne N, Chauleur C, 2014, OSNA, onestep nucleic acid amplification, sentinel lymph node intraoperative molecular analysis in breast cancer: a cost-benefit analysis. Med Oncol 31(12):322 Guillen-Paredes MP, Carrasco-Gonzalez L, Chaves-Benito A, Campillo-Soto A, Carrillo A, Aguayo-Albasini JL, 2011, Onestep nucleic acid amplification, OSNA, assay for sentinel lymph node metastases as an alternative to conventional postoperative histology in breast cancer: a cost-benefit analysis. Cir Esp 89(7): 456–462 Bogani G,Mariani A, Paolini B et al, 2019, Low-volume disease in endometrial cancer: The role of micrometastasis and isolated tumor cells. Gynecol Oncol. , DOI 10.1016/j.ygyno.2019.02.027 pii: S0090–8258(19)30137–4. [Epub ahead of print] Plante M, Stanleigh J, Renaud MC, Sebastianelli A, Grondin K, Gregoire J, 2017, Isolated tumor cells identified by sentinel lymph node mapping in endometrial cancer: does adjuvant treatment matter? Gynecol Oncol 146(2):240–246 St Clair CM, Eriksson AG, Ducie JA et al, 2016, Low-volume lymph node metastasis discovered during sentinel lymph node mapping for endometrial carcinoma. Ann Surg Oncol 23(5): 1653–1659 Tvedskov TF, 2012, Staging of women with breast cancer after introduction of sentinel node guided axillary dissection. Dan Med J 59(7):B4475 Raffone A, Travaglino A, Saccone G, Cieri M, Mascolo M, Mollo A, Insabato L, Zullo F, 2019, Diagnostic and prognostic value of ARID1A in endometrial hyperplasia: a novel marker of occult cancer. APMIS 127(9):597–606 Travaglino A, Raffone A, Saccone G, Mascolo M, D'Alessandro P, Arduino B,Mollo A, Insabato L, Zullo F, 2019, Nuclear expression of β‐catenin in endometrial hyperplasia as marker of premalignancy. APMIs. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2049 EP 2056 DI 10.1007/s12253-019-00727-9 PG 8 ER PT J AU Gargallo, P Yanez, Y Juan, A Segura, V Balaguer, J Torres, B Oltra, S Castel, V Canete, A AF Gargallo, Pablo Yanez, Yania Juan, Antonio Segura, Vanessa Balaguer, Julia Torres, Barbara Oltra, Silves Castel, Victoria Canete, Adela TI Review: Ewing Sarcoma Predisposition SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Ewing sarcoma; Cancer predisposition; Genetic susceptibility; Polymorphism ID Ewing sarcoma; Cancer predisposition; Genetic susceptibility; Polymorphism AB Ewing sarcoma is a rare tumor developed in bone and soft tissues of children and teenagers. This entity is biologically led by a chromosomal translocation, typically including EWS and FLI1 genes. Little is known about Ewing sarcoma predisposition, although the role of environmental factors, ethnicity and certain polymorphisms on Ewing sarcoma susceptibility has been studied during the last few years. Its prevalence among cancer predisposition syndromes has also been thoroughly examined. This review summarizes the available evidence on predisposing factors involved in Ewing sarcoma susceptibility. On the basis of these data, an integrated approach of the most influential factors on Ewing sarcoma predisposition is proposed. C1 [Gargallo, Pablo] La Fe Hospital, Clinical and Translational Oncology Research Group, Av. Fernando Abril Martorell 106, 46026 Valencia, Spain. [Yanez, Yania] La Fe Hospital, Clinical and Translational Oncology Research Group, Av. Fernando Abril Martorell 106, 46026 Valencia, Spain. [Juan, Antonio] La Fe Hospital, Pediatric Oncology and Hematology UnitValencia, Spain. [Segura, Vanessa] La Fe Hospital, Clinical and Translational Oncology Research Group, Av. Fernando Abril Martorell 106, 46026 Valencia, Spain. [Balaguer, Julia] La Fe Hospital, Pediatric Oncology and Hematology UnitValencia, Spain. [Torres, Barbara] La Fe Hospital, Pediatric Oncology and Hematology UnitValencia, Spain. [Oltra, Silves] La Fe Hospital, Genetics UnitValencia, Spain. [Castel, Victoria] La Fe Hospital, Pediatric Oncology and Hematology UnitValencia, Spain. [Canete, Adela] La Fe Hospital, Pediatric Oncology and Hematology UnitValencia, Spain. RP Gargallo, P (reprint author), La Fe Hospital, Clinical and Translational Oncology Research Group, 46026 Valencia, Spain. EM gargallo_pabtat@gva.es CR Horowitz M, Malawer M, Woo S, et al. Ewing's Sarcoma Family of Tumors: Ewing's Sarcoma of Bone and Soft Tissue and the Peripheral Primitive Neuroectodermal Tumors. Pizzo, PA.; Poplack, DG., editors. Principles and Practice of Pediatric Oncology. Philadelphia: Lippincott-Raven Publishers; 1997. p. 831–863 Young IL, Percy CL, Asire AI, 1981, Surveillance, epidemiology, and end results: incidence and mortality data, 1973-1977. Nat I Cancer Inst Monogr 57:149 Worch J, Cyrus J, Goldsby R, Matthay KK, Neuhaus J, DuBois SG, 2011, Racial differences in the incidence of mesenchymal tumors associated with EWSR1 translocation. Cancer Epidemiol Biomark Prev 20(3):449–453 Nakata K, Ito Y,MagadiW, Bonaventure A, Stiller CA, Katanoda K et al, 2018, Childhood cancer incidence and survival in Japan and England: a population-based study, 1993-2010). Cancer Sci 109(2):422–434 Polednak AP, 1985, Primary bone cancer incidence in black and white residents of New York State. Cancer, Phila, 55:2883–2888 Glass AG, Fraumeni JF, 1970, Epidemiology of bone cancer in children. J Natl Cancer Inst 44(1):187–199 Fraumeni JF, Glass AG, 1970, Rarity of Ewing’s sarcoma among U.S. Negro children. Lancet 1:366–367 Jensen RD, Drake RM, 1970, Rarity of Ewing’s tumour in negroes. Lancet. 1:777 Linden G, Dunn IE, 1970, Ewing’s sarcoma in negroes. Lancet 1: 1171 EddingtonGM, Bohrer SP,Middlemass IH, 1970, Ewing’s sarcoma in negroes. Lancet. 1:1171–1172 Oyemade GA, Abioye AA, 1982, Primary malignant tumors of bone: incidence in Ibadan, Nigeria. I NatI Med Assoc 74:65–68 Kramer S, Meadows AT, Jarrett P, Evans AE, 1983, Incidence of childhood cancer: experience of a decade in a population-based registry. I NatI Cancer Inst 10:49–55 Li FP, Tu JT, Liu FS, Shiang EL, 1980, Rarity of Ewing’s sarcoma in China. Lancet. 1:1255 Savita S, Stephen L, 2011, Promiscuous partnerships in Ewing’s sarcoma. Cancer Genet. 204(7):351–365 Hancock JD, Lessnick SL, 2008, A transcriptional profiling metaanalysis reveals a core EWS-FLI gene expression signature. Cell Cycle 7:250–256 Sankar S, Bell R, Stephens B, Zhuo R, Sharma S, Bearss DJ et al, 2013, Mechanism and relevance of EWS/FLI-mediated transcriptional repression in Ewing sarcoma. Oncogene. 32:5089– 5100 Lessnick SL, LadanyiM(2012)Molecular pathogenesis of Ewing sarcoma: new therapeutic and transcriptional targets. Annu Rev Pathol 7:145–159 Takigami I, Ohno T, Kitade Y, Hara A, Nagano A, Kawai G et al, 2011, Synthetic siRNA targeting the breakpoint of EWS/Fli-1 inhibits growth of Ewing sarcoma xenografts in a mouse model. Int J Cancer 128:216–226 Maksimenko A, Malvy C, 2005, Oncogene-targeted antisense oligonucleotides for the treatment of Ewing sarcoma. Expert OpinTher Targets 9:825–830 Mateo-Lozano S, Gokhale PC, Soldatenkov VA, Dritschilo A, TiradoOM, Notario V, 2006, Combined transcriptional and translational targeting of EWS/FLI-1 in Ewing's sarcoma. Clin Cancer Res 12:6781–6790 Stoll G, Surdez D, Tirode F, Laud K, Barillot E, Zinovyev A et al, 2013, Systems biology of Ewing sarcoma: a network model of EWS-FLI1 effect on proliferation and apoptosis. Nucleic Acids Res 41(19):8853–8871 Riggi N, Knoechel B, Shawn M, Rheinbay E, Boulay G, Suva M et al, 2014, EWS-FLI1 utilizes divergent chromatin remodeling mechanisms to directly activate or repress enhancer elements in Ewing sarcoma. Cancer Cell 26(5):668–681 Bilke S, Schwentner R, Yang F, Kauer M, Jug G,Walker RL et al, 2013, Oncogenic ETS fusions deregulate E2F3 target genes in Ewing sarcoma and prostate cancer. Genome Res 23:1797–1809 Gangwal K, Close D, Enriquez CA, Hill CP, Lessnick SL, 2010, Emergent properties of EWS/FLI regulation via GGAA microsatellites in Ewing's sarcoma. Genes Cancer 1:177–187 Gangwal K, Sankar S, Hollenhorst PC, Kinsey M, Haroldsen SC, Shah AA et al, 2008, Microsatellites as EWS/FLI response elements in Ewing's sarcoma. Proc Natl Acad Sci U S A 105:10149– 10154 Guillon N, Tirode F, Boeva V, Zynovyev A, Barillot E, Delattre O, 2009, The oncogenic EWS-FLI1 protein binds in vivo GGAA microsatellite sequences with potential transcriptional activation function. PLoS One 4:e4932 PatelM, Simon JM, IglesiaMD,Wu SB,McFaddenAW, Lieb JD, Davis IJ, 2012, Tumor-specific retargeting of an oncogenic transcription factor chimera results in dysregulation of chromatin and transcription. Genome Res 22:259–270 Roberts P, Burchill SA, Brownhill S, Cullinane CJ, Johnston C, Griffiths MJ et al, 2008, Ploidy and karyotype complexity are powerful prognostic indicators in the Ewing s sarcoma family of tumors: a study by the United Kingdom cancer cytogenetics and the children s cancer and leukaemia group. Genes Chromosomes Cancer 47:207–220 Mackintosh C, Ordonez JL, Garcia-Dominguez DJ, Sevillano V, Llombart-Bosch A, Szuhai K et al, 2012, 1q gain and CDT2 overexpression underlie an aggressive and highly proliferative form of Ewing sarcoma. Oncogene. 31:1287–1298 Hattinger CM, Potschger U, TarkkanenM, Squire J, Zielenska M, Kiuru-Kuhlefelt S et al, 2002, Prognostic impact of chromosomal aberrations in Ewing tumours. Br J Cancer 86:1763–1769 Kan Z, Jaiswal BS, Stinson J, Janakiraman V, Bhatt D, Stern HM et al, 2010, Diverse somatic mutation patterns and pathway alterations in human cancers. Nature. 466:869–873 Brohl AS, Solomon DA, ChangW,Wang J, Song Y, Sindiri S et al, 2014, The genomic landscape of the Ewing sarcoma family of tumors reveals recurrent STAG2 mutation. PLoS Genet 10(7): e1004475 Winn DM, Li FP, Robison LL,Mulvihill JJ, Daigle AE, Fraumeni JF, 1992, A case-control study of the etiology of Ewing's sarcoma. Cancer Epidemiol Biomark Prev 1(7):525–532 Abbott D, Randall RL, Schiffman J, Lessnick S, Cannon-Albright LA. A population-based survey of excess cancers observed in Ewing's sarcoma and in their first-, second-, and third-degree relatives. Cancer Res. 2015; 75(15 Suppl): Abstract nr 2748. https:// doi.org/10.1158/1538-7445 Riggi N, Cironi L, Provero P, Suva ML, Kaloulis K et al, 2005, Development of Ewing’s sarcoma from primary bone marrowderived mesenchymal progenitor cells. Cancer Res 65:11459– 11468 Riggi N, Suva ML, De Vito C, Provero P, Stehle JC et al, 2010, EWS-FLI-1 modulates miRNA145 and SOX2 expression to initiate mesenchymal stem cell eprogramming toward Ewing sarcoma cancer stem cells. Genes Dev 24:916–932 Tirode F, Laud-Duval K, Prieur A, Delorme B, Charbord P et al, 2007, Mesenchymal stem cell features of Ewing tumors. Cancer Cell 11:421–429 Toomey EC, Schiffman JD, Lessnick SL, 2010, Recent advances in the molecular pathogenesis of Ewing’s sarcoma. Oncogene. 29: 4504–4516 Von Levetzow C, JiangX,Gwye Y, von Levetzow G, Hung L et al, 2011, Modeling initiation of Ewing sarcoma in human neural crest cells. PLoS One 6:e19305 Ross KA, Smyth NA, Murawski Kennedy JG, 2013, The biology of Ewing sarcoma. ISRN Oncol 759725 Amaral AT, Manara MC, Berghuis D, Ordonez JL, Biscuola M, Lopez-Garcia MA et al, 2014, Characterization of human Mesenchymal stem cells from Ewing sarcoma patients. Pathogenetic Implications. Plos One 9:e85814 Johnson JJ, Chen W, Hudson W, Yao Q, Taylor M et al, 2003, Prenatal and postnatal myeloid cells demonstrate stepwise progression in the pathogenesis of MLL fusion gene leukemia. Blood. 101:3229–3235 Kovar H, Amatruda J, Brunet E, Burdach S, Cidre-Aranaz F, de Alava E et al, 2016, The second European interdisciplinary Ewing sarcoma research summit–a joint effort to deconstructing the multiple layers of a complex disease. Oncotarget. 7(8):8613–8624 Brodeur GM, Nichols KE, Plon SE, Schiffman JD, Malkin D, 2017, Pediatric Cancer predisposition and surveillance: an overview, and a tribute to Alfred G. Knudson Jr. Clin Cancer Res 23:1–5 Greer MC, Voss SD, States LJ, 2017, Pediatric Cancer predisposition imaging: focus on whole-body MRI. Clin Cancer Res 23:6–13 Porter CC et al, 2017, Recommendations for surveillance for children with leukemia-predisposing conditions. Clin Cancer Res 23: 14–22 Walsh MF et al, 2017, Recommendations for childhood Cancer screening and surveillance in DNA repair disorders. Clin Cancer Res 23:23–31 Tabori U et al, 2017, Clinical management and tumor surveillance recommendations of inheritedmismatch repair deficiency in childhood. Clin Cancer Res 23:32–37 Evans DGR et al, 2017a, Cancer and central nervous system tumor surveillance in pediatric Neurofibromatosis 1. Clin Cancer Res 23:46–53 Evans DGR et al, 2017b, Cancer and central nervous system tumor surveillance in pediatric Neurofibromatosis 2 and related disorders. Clin Cancer Res 23:54–61 FoulkesWD et al, 2017, Cancer surveillance in Gorlin syndrome and Rhabdoid tumor predisposition syndrome. Clin Cancer Res 23:62–67 Rednam SP et al, 2017, Von Hippel-Lindau and hereditary pheochromocytoma/Paraganglioma syndromes: clinical features, genetics, and surveillance recommendations in childhood. Clin Cancer Res 23:68–75 Schultz KAP et al, 2017, PTEN, DICER1, FH, and their associated tumor susceptibility syndromes: clinical features, genetics, and surveillance recommendations in childhood. Clin Cancer Res 23:76–82 Villani A et al, 2017, Recommendations for Cancer surveillance in individuals with RASopathies and other rare genetic conditions with increased Cancer risk. Clin Cancer Res 23:83–90 Druker H et al, 2017, Genetic counselor recommendations for Cancer predisposition evaluation and surveillance in the pediatric oncology patient. Clin Cancer Res 23:91–97 Kamihara J et al, 2017, Retinoblastoma and Neuroblastoma Predisposition and Surveillance. Clin Cancer Res 23:98–106 Achatz MI et al, 2017, Cancer screening recommendations and clinical Management of Inherited Gastrointestinal Cancer Syndromes in childhood. Clin Cancer Res 23:107–114 Kalish JMet al, 2017, Surveillance recommendations for children with overgrowth syndromes and predisposition to Wilms tumors and Hepatoblastoma. Clin Cancer Res 23:115–122 Wasserman JD et al, 2017, Multiple endocrine Neoplasia and Hyperparathyroid-jaw tumor syndromes: clinical features, genetics, and surveillance recommendations in childhood. Clin Cancer Res 23:123–132 Etzold A, Schroder JC, Bartsch O, Zechner U, Galetzka D, 2015, Further evidence for pathogenicity of the TP53 tetramerization domain mutation p.Arg342Pro in Li- Fraumeni syndrome. Fam Cancer. 14(1):161–165 Macedo GS, Araujo Vieira I, Brandalize AP, Giacomazzi J, Inez Palmero E, Volc S, 2016, Rare germline variant, rs78378222, in the TP53 3' UTR: evidence for a new mechanism of cancer predisposition in Li-Fraumeni syndrome. Cancer Genet 209(3):97– 106 Calvete O, Martinez P, Garcia-Pavia P, Benitez-Buelga C, Paumard-Hernandez B, Fernandez V et al, 2015, A mutation in the POT1 gene is responsable for cardiac angiosarcoma in TP53- negative Li–Fraumeni-like families. Nat Commun 6:8383 Calvete O, Garcia-Pavia P, Dominguez F, Bougeard G, Kunze K, Braeuninger A et al, 2017, The wide spectrum of POT1 gene variants correlates with multiple cancer types. Eur J Hum Genet 25(11):1278–1281 Siddiqui R, Onel K, Facio F, Nafa K, Diaz LR, Kauff N, 2005, The TP53 mutational spectrum and frequency of CHEK2*1100delC in Li-Fraumeni-like kindreds. Familial Cancer 4(2):177–181 Varley J, 2003, TP53, hChk2, and the Li-Fraumeni syndrome. Methods Mol Biol 222:117–129 Vahteristo P, Tamminen A, Karvinen P, Eerola H, Eklund C, Altonen LA et al, 2001, p53, CHK2, and CHK1 Genes in Finnish Families with Li-Fraumeni Syndrome: Further Evidence of CHK2 in Inherited Cancer Predisposition. Cancer Res 61(15): 5718–5722 Manoukian S, Peissel B, Frigerio S, Lecis D, Bartkova J, 2011, RoversiG, et al. two new CHEK2 germline variants detected in breast cancer/sarcoma families negative for BRCA1, BRCA2, and TP53 gene mutations. Breast Cancer Res Treat 130(1):207–215 Ruijs MW, Broeks A, Menko FH, Ausems MG, Wagner A, Oldenburg R et al, 2009, The contribution of CHEK2 to the TP53-negative Li-Fraumeni phenotype. Hered Cancer Clin Pract 7(1):4 Li FP, Fraumeni JF Jr, 1969, Soft-tissue sarcomas, breast cancer, and other neoplasms. A familial syndrome? Ann Intern Med 71(4):747–752 Mai PL, Best AF, Peters JA, De Castro RM, Khincha PP, Loud JT et al, 2016, Risks of first and subsequent cancers among TP53 mutation-carriers in the NCI LFS cohort. Cancer. 122(23):3673– 3681 Birch JM,Alston RD,McNally RJ, EvansDG, KelseyAM, Harris M et al, 2001, Relative frequency and morphology of cancers in carriers of germline TP53 mutations. Oncogene. 20(34):4621– 4628 Mody RJ,Wu YM, Lonigro RJ, Cao X, Roychowdhury S, Vats P et al, 2015, Integrative clinical sequencing in themanagement of refractory or relapsed cancer in youth. JAMA 314:913–925 HarrisMH, DuBois SG, Glade Bender JL, Kim A, Crompton BD, Parker E et al, 2016)Multicenter feasibility study oftumor molecular profiling to informtherapeutic decisions in advanced pediatric solid tumors: the individualized Cancer therapy, iCat, study. JAMA Oncol 2:608–615 Oberg JA, Glade Bender JL, Sulis ML, Pendrick D, Sireci AN, Hsiao SJ et al, 2016, Implementation of next generation sequencing into pediatric hematology-oncology practice: moving beyond actionable alterations. Genome Med 8:133 Parsons DW, Roy A, Yang Y, Wang T, Scollon S, Bergstrom K et al, 2016, Diagnostic yield of clinical tumor and germlinewholeexome sequencing for children with solid tumors. JAMA Oncol. 2:616–624 Worst BC, van Tilburg CM, Balasubramanian GP, Fiesel P, Witt R, Freitag A et al, 2016, Next-generation personalised medicine for high-risk paediatric cancer patients – the INFORMpilot study. Eur J Cancer 65:91–101 Harttrampf AC, Lacroix L, Deloger M, Deschamps F, Puget S, Auger N et al, 2017, MOlecular screening for CancerTreatment optimization, MOSCATO-01, in pediatric patients: a single institutional prospective molecular stratification trial. Clin Cancer Res 23:6101–6112 ChangW, Brohl AS, Patidara R, Sindiria S, Shern JF,Wei JS et al, 2016, Multi-dimensional Omics for precision therapy of children and adolescent Young adults with relapsed and refractory Cancer: report from pediatric oncology branch. NCI Clin Cancer Res 22(15):3810–3820 Pincez T, Clement N, Lapouble E, Pierron G, Kamal M, Bieche I et al, 2017, Feasibility and clinical integration of molecular profiling for target identification in pediatric solid tumors. Pediatr Blood Cancer 64:e26365 Zhang J,Walsh MF, Wu G, Edmonson MN, Gruber TA, Easton J et al, 2015, Germline mutations in predisposition genes in pediatric Cancer. N Engl J Med 373(24):2336–2346 Brohl AS, Patidar R, Turner CE,Wen X, Song YK,Wei JS, 2017, Frequent inactivating germline mutations in DNA repair genes in patients with Ewing sarcoma Germline mutations in Ewing sarcoma. Genet Med 19(8):955–958 Lerman D, Monument M, McIlvaine E, Liu X, Huang D, Monovich L et al, 2015, TumoralTP53 and/or CDKN2A alterations are not reliable prognostic biomarkers in patients with localized Ewing sarcoma: a report from the Children’s oncology group. Pediatr Blood Cancer 62(5):759–765 Id Said B, Kim H, Tran J, Novokmet A, Malkin D, 2016, Supertransactivation TP53 variant in the Germline of a family with Li- Fraumeni syndrome. Hum Mutat 37(9):889–892 Marcel V, Palmero EI, Falagan-Lotsch P, Martel-Planche G, Ashton-Prolla P, Olivier M, 2009, TP53 PIN3 and MDM2 SNP309 polymorphisms as genetic modifiers in the Li-Fraumeni syndrome: impact on age at first diagnosis. J Med Genet 46(11): 766–772 Sagne C, Marcel V, Bota M, Martel-Planche G, Nobrega A, Palmero EI et al, 2014, Age at cancer onset in germline TP53 mutation carriers: association with polymorphisms in predicted G-quadruplex structures. Carcinogenesis. 35(4):807–815 Bougeard G, Baert-Desurmont S, Tournier I, Vasseur S,Martin C, Brugieres L, 2006, Impact of the MDM2 SNP309 and p53 Arg72Pro polymorphism on age of tumour onset in Li-Fraumeni syndrome. J Med Genet 43(6):531–533 Ponti F, Corsini S, Gnoli M, Pedrini E, Mordenti M, Sangiorgi L, 2016, Evaluation of TP53 Pro72Arg and MDM2 SNP 285- SNP309 polymorphisms in an Italian cohort of LFS suggestive patients lacking identifiable TP53 germline mutations. Familial Cancer 15(4):635–643 Wu CC, Krahe R, Lozano G, Zhang B, Wilson CD, Jo EJ et al, 2011, Joint effects of germ-line p53 mutation, MDM2 SNP309, and gender on Cancer risk in family studies of Li-Fraumeni syndrome. Hum Genet 129(6):663–673 Ruijs MW, Schmidt MK, Nevanlinna H, Tommiska J, Aittomaki K, Pruntel R, 2007, The single-nucleotide polymorphism 309 in the MDM2 gene contributes to the Li-Fraumeni syndrome and related phenotypes. Eur J HumGenet 15(1):110–114 Macedo GS, Vieira IA, Vianna FSL, Alemar B, Giacomazzi J, Brandalize APC et al, 2018, P53 signaling pathway polymorphisms, cancer risk and tumor phenotype in TP53 R337H mutation carriers. Familial Cancer 17(2):269–274 Renaux-Petel M, Sesboue R, Baert-Desurmont S, Vasseur S, Fourneaux S, Bessenay E, et al. The MDM2 285G–309G haplotype is associated with an earlier age of tumour onset in patients with Li-Fraumeni syndrome. Fam Cancer. 2014;, 1):127–30 Thurow HS, Hartwig FP, Alho CS, Silva DS, Roesler R, Abujamra AL et al, 2013, Wing sarcoma: influence of TP53 Arg72Pro and MDM2 T309G SNPs. Mol Biol Rep 40(8):4929– 4934 Kleinerman RA, TuckerMA, Abramson DH, Seddon JM, Tarone RE, Fraumeni JF Jr, 2007, Risk of soft tissue sarcomas by individual subtype in survivors of hereditary retinoblastoma. J Natl Cancer Inst 99(1):24–31 MacCarthy A, Bayne AM, Brownbill PA, Bunch KJ, Diggens NL, Draper GJ et al, 2013, Second and subsequent tumours among 1927 retinoblastoma patients diagnosed in Britain 1951–2004. Br J Cancer 108:2455–2463 Cunniff C, Bassetti JA, Ellis NA, 2017, Bloom's syndrome: clinical Spectrum,molecular pathogenesis, and Cancer predisposition. Mol Syndromol 8(1):4–23 Laitman Y, Boker-Keinan L, Berkenstadt M, Liphsitz I, Weissglas-Volkov D, Ries-Levavi L et al, 2016, The risk for developing cancer in Israeli ATM, BLM, and FANCC heterozygous mutation carriers. Cancer Genet. 209(3):70–74 Dong H, Nebert DW, Bruford EA, Thompson DC, Joenje H, Vasiliou V, 2015, Update of the human and mouse Fanconi anemia genes. Hum Genomics 9:32 Malric A, Defachelles AS, Leblanc T, Lescoeur B, Lacour B, Peuchmaur M, 2015, Fanconi anemia and solid malignancies in childhood: a national retrospective study. Pediatr Blood Cancer 62(3):463–470 Wimmer K, Rosenbaum T, Messiaen L, 2017, Connections between constitutional mismatch repair deficiency syndrome and neurofibromatosis type 1. Clin Genet 91(4):507–519 Bakry D, Aronson M, Durno C, Rimawi H, Farah R, Alharbi QK et al, 2014, Genetic and clinical determinants of constitutional mismatch repair deficiency syndrome: report from the constitutional mismatch repair deficiency consortium. Eur J Cancer 50(5):987–996 Lavoine N, Colas C, Muleris M, Bodo S, Duval A, Entz-Werle N, 2015, Constitutional mismatch repair deficiency syndrome: clinical description in a French cohort. J Med Genet 52(11):770–778 Kawai K, Iwashita T, Murakami H, Hiraiwa N, Yoshiki A, Kusakabe M et al, 2000, Tissue-specific carcinogenesis in transgenic mice expressing the RET proto-oncogene with a multiple endocrine neoplasia type 2A mutation. Cancer Res 60(18):5254– 5260 Martinelli M, Parra A, Scapoli L, De Sanctis P, Chiadini V, Hattinger C et al, 2016, CD99 polymorphisms significantly influence the probability to develop Ewing sarcoma in earlier age and patient disease progression. Oncotarget. 7(47):77958–77967 Silva DS, Sawitzki FR, De Toni EC, Graebin P, Picanco JB, Abujamra AL, de Farias CB, Roesler R, Brunetto AL, Alho CS, 2012, Ewing's sarcoma: analysis of single nucleotide polymorphism in the EWS gene. Gene. 509(2):263–266 Wang J, Zhou Y, Feng D, Yang H, Li F, Cao Q, Wang A, Xing F, 2012, CD86 +1057G/a polymorphism and susceptibility to Ewing's sarcoma: a case-control study. DNA Cell Biol 31(4): 537–540 Zhang C, Hou WH, Ding XX,Wang X, Zhao H, Han XW, Wang WJ, 2016, Association of Cytotoxic T-lymphocyte Antigen-4 polymorphisms with malignant bone tumor risk: a meta-analysis. Asian Pac J Cancer Prev 17(8):3785–3791 Postel-Vinay S, VeronAS, Tirode F, Pierron G, Reynaud S, Kovar H et al, 2012, Common variants near TARDBP and EGR2 are associated with susceptibility to Ewing sarcoma. Nat Genet 44(3): 323–327 Grunewald TG, Delattre O, 2015, Cooperation between somatic mutations and germline susceptibility variants in tumorigenesis - a dangerous liaison. Mol Cell Oncol 3(3):e1086853 Machiela MJ, Grunewald TGP, Surdez D, Reynaud S, Mirabeau O, Karlins E et al, 2018, Genome-wide association study identifies multiple new loci associated with Ewing sarcoma susceptibility. Nat Commun 9(1):3184 Monument MJ, Johnson KM, Grossmann AH, Schiffman JD, Randall RL, Lessnick SL, 2012, Microsatellites with macroinfluence in Ewing sarcoma. Genes, Basel, 3(3):444–460 Monument MJ, Johnson KM, McIlvaine E, Abegglen L, Watkins WS, Jorde LB et al, 2014, Clinical and biochemical function of polymorphic NR0B1 GGAA-microsatellites in Ewing sarcoma: a report from the Children's oncology group. PLoS One 9(8): e104378 Kolomietz E, Meyn MS, Pandita A, Squire JA, 2002, The role of Alu repeat clusters as mediators of recurrent chromosomal aberrations in tumors. Genes Chromosomes and Cancer 35(2):97–112 Zucman-Rossi J, BatzerMA, StonekingM, Delattre O, Thomas G, 1997, Interethnic polymorphism of EWS intron 6: genome plasticity mediated by Alu retroposition and recombination. Hum Genet 99(3):357–363 Randall RL, Lessnick SL, Jones KB, Gouw LG, Cummings JE, Cannon-Albright L, 2010, Is there a predisposition gene for Ewing's sarcoma? J Oncol 2010:397632 Holly EA, Aston DA, Ahn DK, Kristiansen JJ, 1992, Ewing’s bone sarcoma, paternal occupational exposure, and other factors. Am J Epidemiol 135(2):122–129 Valery PC, Mc Whirter W, Sleigh A, Williams G, Bain C, 2002, Farm exposures, parental occupation, and risk of Ewing’s sarcoma in Australia: a national case-control study. Cancer Causes Control 13(3):263–270 Valery PC,McWhirterW, Sleigh A,Williams G, Bain C, 2003, A national case-control study of Ewing’s sarcoma family of tumours in Australia. Int J Cancer 105(6):825–830 Valery PC, Williams G, Sleigh A, Holly EA, Kreiger N, Bain C, 2005, Parental occupation and Ewing’s sarcoma: pooled and meta- analysis. Int J Cancer 115(5):799–806 Kovar H, Jug G, Aryee DN, Zoubek A, Ambros P, Gruber B et al, 1997, Among genes involved in the RB dependent cell cycle regulatory cascade, the p16 tumor suppressor gene is fre quently lost in the Ewing family of tumors. Oncogene. 15:2225–2232 Cortessis VK, Thomas DC, Levine AJ, Breton CV, Mack TM, Siegmund KD et al, 2012, Environmental epigenetics: prospects for studying epigenetic mediation of exposure–response relationships. Hum Genet 131:1565–1589 Hanson MA, Skinner MK. Developmental origins of epigenetic transgenerational inheritance. Environ Epigenet. 2016;2(1) Nilsson E, SkinnerM, 2015, Environmentally induced epigenetic Transgenerational inheritance of disease susceptibility. Transl Res 165(1):12–17 Adkins RM, Thomas F, Tylavsky FA, Krushkal J, 2011, Parental ages and levels ofDNAmethylation in the newborn are correlated. BMC Med Genet 12:47 Joo JE, Dowty JG, Milne RL, Wong EM, Dugue PA, English D et al, 2018, Heritable DNA methylation marks associated with susceptibility to breast cancer. Nat Commun 9(1):867 Haque MM, Nilsson EE, Holder LB, Skinner MK, 2016, Genomic clustering of differential DNA methylated regions, epimutations, associated with the epigenetic transgenerational inheritance of disease and phenotypic variation. BMC Genomics 17:418 Johnson KJ, Carozza SE, Chow EJ et al, 2009, Parental age and risk of childhood cancer. Epidemiology. 20(4):475–483 Krepischi AC, Capelli LP, Silva AG, de Araujo ES, Pearson PL, Heck B et al, 2014, Large germline copy number variations as predisposing factor in childhood neoplasms. Future Oncol 10(9): 1627–1633 Hingorani P, Janeway K, Crompton BD, Kadoch C, Mackall CL, Khan J et al, 2016, Current state of pediatric sarcoma biology and opportunities for future discovery: a report fromthe sarcoma translational research workshop. Cancer Genet. 209(5):182–194 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2057 EP 2066 DI 10.1007/s12253-019-00765-3 PG 10 ER PT J AU Travaglino, A Raffone, A Gencarelli, A Mollo, A Guida, M Insabato, L Santoro, A Zannoni, FG Zull, F AF Travaglino, Antonio Raffone, Antonio Gencarelli, Annarita Mollo, Antonio Guida, Maurizio Insabato, Luigi Santoro, Angela Zannoni, Franco Gian Zull, Fulvio TI TCGA Classification of Endometrial Cancer: the Place of Carcinosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Cancer; Endometrium; Prognosis; Risk assessment; PROMISE; Treatment ID Cancer; Endometrium; Prognosis; Risk assessment; PROMISE; Treatment AB In 2013, The Cancer Genome Atlas (TCGA) Research Network found four novel prognostic subgroups of endometrial carcinoma: POLE/ultramutated (POLE), microsatellite-instable/hypermutated (MSI), copy-number-low/TP53-wild-type (CNL), and copy-number-highTP53-mutant (CNH). However, poor is known regarding uncommon histotypes of endometrial cancer. We aimed to assess the genetic profile of uterine carcinosarcoma (UCS) on the light of these findings. A systematic review and metaanalysis was performed through electronic databases searching (up to July 2019). All studies assessing UCS series for the TCGA classification were included. For each TCGA subgroup, pooled prevalence on the total UCS number was calculated. Four studies with 231 patients were included. Pooled prevalence of the TCGA subgroups were: 5.3% for the POLE subgroup, 7.3% for the MSI subgroup, 73.9% for the CNH subgroup, 13.5% for the CNL subgroup. The CNH subgroup predominates in UCS, while subgroups with high mutational load (POLE and MSI) are less common. UCS appears as a preferential evolution of CNH carcinomas. C1 [Travaglino, Antonio] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology UnitNaples, Italy. [Raffone, Antonio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Gencarelli, Annarita] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology UnitNaples, Italy. [Mollo, Antonio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Guida, Maurizio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Insabato, Luigi] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology UnitNaples, Italy. [Santoro, Angela] Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Department of Woman and Child HealthRome, Italy. [Zannoni, Franco Gian] Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Department of Woman and Child HealthRome, Italy. [Zull, Fulvio] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. RP Raffone, A (reprint author), University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Gynecology and Obstetrics Unit, 80131 Naples, Italy. EM anton.raffone@gmail.com CR Siegel RL, Miller KD, Jemal A, 2015, Cancer statistics, 2015. CA Cancer J Clin 65(1):5–29 Raffone A, TravaglinoA,Mascolo M, Carbone L,Guida M, Insabato L, Zullo F, 2019, TCGA molecular groups of endometrial cancer: pooled data about prognosis. Gynecol Oncol 155(2):374–383 Travaglino A, Raffone A, Saccone G, de Luca C, Mollo A, Mascolo M, de Placido G, Insabato L, Zullo F, 2019, Immunohistochemical nuclear expression of β-catenin as a surrogate of CTNNB1 exon 3 mutation in endometrial Cancer. Am J Clin Pathol 151(5):529–538 Raffone A, Travaglino A, Santoro A, Esposito I, Angelico G, Spadola S, Zannoni GF, 2019, Accuracy of one-step nucleic acid amplification in detecting lymph node metastases in endometrial cancer. Pathol Oncol Res. , DOI 10.1007/s12253-019-00727-9 Travaglino A, Raffone A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2019, Immunohistochemical predictive markers of response to conservative treatment of endometrial hyperplasia and early endometrial cancer: a systematic review. Acta Obstet Gynecol Scand 98:1086–1099. , DOI 10.1111/aogs.13587 Abu-Rustum NR, Yashar CM, Bean S, et al. NCCN Clinical Practice Guidelines in Oncology, NCCN Guidelines®, – Uterine neoplasms. Version 3.2019 – February 11, 2019 Hoang LN, McConechy MK, Kobel M et al, 2013, Histotypegenotype correlation in 36 high-grade endometrial carcinomas. Am J Surg Pathol 37:1421–1432 Gilks CB, Oliva E, Soslow RA, 2013, Poor interobserver reproducibility in the diagnosis of high-grade endometrial carcinoma. Am J Surg Pathol 37:874–881 Cancer Genome Atlas Research Network et al, 2013, Integrated genomic characterization of endometrial carcinoma. Nature 497(7447):67–73 Talhouk A, McConechy MK, Leung S et al, 2015, A clinically applicable molecular-based classification for endometrial cancers. Br J Cancer 113(2):299–310 Talhouk A,McConechyMK, Leung S et al, 2017, Confirmation of ProMisE: a simple, genomics-based clinical classifier for endometrial cancer. Cancer 123(5):802–813 Kommoss S, McConechy MK, Kommoss F et al, 2018, Final validation of the ProMisE molecular classifier for endometrial carcinoma in a large population-based case series. AnnOncol 29(5): 1180–1188 Stelloo E, Nout RA, Osse EM, Jurgenliemk-Schulz IJ, Jobsen JJ, Lutgens LC, van der Steen-Banasik EM, Nijman HW, Putter H, Bosse T, Creutzberg CL, Smit VTHBM, 2016, Improved risk assessment by integrating molecular and clinicopathological factors in early-stage endometrial cancer-combined analysis of the PORTEC cohorts. Clin Cancer Res 22(16):4215–4224 Bosse T, Nout RA, McAlpine JN et al, 2018)Molecular classification of grade 3 endometrioid endometrial cancers identifies distinct prognostic subgroups. Am J Surg Pathol 42(5):561–568 Cosgrove CM, Tritchler DL, Cohn DE et al, 2018, An NRG oncology/GOG study of molecular classification for risk prediction in endometrioid endometrial cancer. GynecolOncol 148(1):174– 180 Leskela S, Perez-Mies B, Rosa-Rosa JM, Cristobal E, Biscuola M, Palacios-Berraquero ML, Ong S, Matias-Guiu Guia X, Palacios J, 2019, Molecular basis of tumor heterogeneity in endometrial carcinosarcoma. Cancers, Basel, 11(7):964 McAlpine J, Leon-Castillo A, Bosse T, 2018, The rise of a novel classification system for endometrial carcinoma; integration of molecular subclasses. J Pathol 244(5):538–549 Raffone A, Travaglino A, Saccone G, Alviggi C, Mascolo M, De Placido G, Insabato L, Mollo A, Zullo F, 2019, Management of women with atypical polypoid adenomyoma of the uterus: a quantitative systematic review. Acta Obstet Gynecol Scand 98(7):842– 855 Raffone A, Travaglino A, Saccone G et al, 2019, Should progesterone and estrogens receptors be assessed for predicting the response to conservative treatment of endometrial hyperplasia and cancer? A systematic review and meta-analysis. Acta Obstet Gynecol Scand 98(8):976–987 Lionetti R, De Luca M, Travaglino A, Raffone A, Insabato L, Saccone G, Mascolo M, D’armiento M, Zullo F, Corcione F, 2019, Treatments and overall survival in patients with Krukenberg tumor. Arch Gynecol Obstet 300(1):15–23 Moher D, Shamseer L, Clarke M et al, 2015, Preferred reporting items for systematic review and meta-analysis protocols, PRISMAP, 2015 statement. Syst Rev 4:1 Whiting PF, Rutjes AW, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, Leeflang MM, Sterne JA, Bossuyt PM, QUADAS-2 Group, 2011, QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med 155(8):529– 536 Raffone A, Travaglino A, Saccone G, Viggiani M, Giampaolino P, Insabato L, Mollo A, de Placido G, Zullo F, 2019, PTEN expression in endometrial hyperplasia and risk of cancer: a systematic review and meta-analysis. Arch Gynecol Obstet 299(6):1511–1524 Travaglino A, Raffone A, Saccone G, D’Alessandro P, Arduino B, de Placido G, Mascolo M, Insabato L, Zullo F, 2019, Significant risk of occult cancer in complex non-atypical endometrial hyperplasia. Arch Gynecol Obstet 300(5):1147–1154 Travaglino A, Raffone A, Saccone G, Mascolo M, Pignatiello S, Mollo A, de Placido G, Insabato L, Zullo F, 2019, PTEN immunohistochemistry in endometrial hyperplasia: which are the optimal criteria for the diagnosis of precancer? APMIS 127(4):161–169 Travaglino A, Raffone A, Mascolo M, Guida M, Insabato L, Zannoni GF, Zullo F, 2019, Clear cell endometrial carcinoma and the TCGA classification. Histopathology. 76:336–338. , DOI 10.1111/his.13976 Raffone A, Travaglino A, Saccone G, Insabato L, Mollo A, de Placido G, Zullo F, 2019, Endometrial hyperplasia and progression to cancer: which classification system stratifies the risk better? A systematic review and meta-analysis. Arch Gynecol Obstet 299(5): 1233–1242 Raffone A, Travaglino A, Saccone G, D’Alessandro P, Arduino B, Mascolo M, de Placido G, Insabato L, Zullo F, 2019, Diabetes mellitus is associated with occult Cancer in endometrial hyperplasia. Pathol Oncol Res. , DOI 10.1007/s12253-019-00684-3 Travaglino A, Raffone A, Saccone G, Mollo A, de Placido G, Insabato L, Zullo F, 2019, Endometrial hyperplasia and risk of coexistent cancer: WHO vs EIN criteria. Histopathology 74(5): 676–687 Raffone A, Travaglino A, Saccone G, di Maio A, Mollo A, Mascolo M, de Rosa R, de Placido G, Insabato L, Zullo F, 2019, Diabetes mellitus and responsiveness of endometrial hyperplasia and early endometrial cancer to conservative treatment. Gynecol Endocrinol 35(11):932–937 Raffone A, Travaglino A, Saccone G, Cieri M, Mascolo M, Mollo A, Insabato L, Zullo F, 2019, Diagnostic and prognostic value of ARID1A in endometrial hyperplasia: a novel marker of occult cancer. APMIS. 127:597–606. , DOI 10.1111/apm.12977 Travaglino A, Raffone A, Saccone G et al, 2019, Congruence between 1994 WHO classification of endometrial hyperplasia and endometrial intraepithelial neoplasia system. Am J Clin Pathol 153(1):40–48 Travaglino A, Raffone A, Saccone G, Fuggi M, de Placido G, Mascolo M, Mollo A, Insabato L, Zullo F, 2019, Immunophenotype of atypical polypoid adenomyoma of the uterus: diagnostic value and insight on pathogenesis. Appl Immunohistochem Mol Morphol. , DOI 10.1097/PAI. 0000000000000780 Travaglino A, Raffone A, Saccone G,MascoloM, D'Alessandro P, Arduino B,Mollo A, Insabato L, Zullo F, 2019, Nuclear expression of β-catenin in endometrial hyperplasia as marker of premalignancy. APMIS. 127:699–709. , DOI 10.1111/ apm.12988 Hoang LN, Ali RH, Lau S, Gilks CB, Lee CH, 2014, Immunohistochemical survey of mismatch repair protein expression in uterine sarcomas and carcinosarcomas. Int J Gynecol Pathol 33(5):483–491 Hembree TN, Teer JK, Hakam A, Chiappori AA, 2016, Genetic investigation of uterine carcinosarcoma: case report and cohort analysis. Cancer Control 23(1):61–66 Zhao S, Bellone S, Lopez S, Thakral D, Schwab C, English DP, Black J, Cocco E, Choi J, Zammataro L, Predolini F, Bonazzoli E, BiM, Buza N, Hui P,Wong S, Abu-KhalafM, Ravaggi A, Bignotti E, Bandiera E, Romani C, Todeschini P, Tassi R, Zanotti L, Odicino F, Pecorelli S, Donzelli C, Ardighieri L, Facchetti F, Falchetti M, Silasi DA, Ratner E, Azodi M, Schwartz PE, Mane S, Angioli R, Terranova C, Quick CM, Edraki B, Bilguvar K, Lee M, Choi M, Stiegler AL, Boggon TJ, Schlessinger J, Lifton RP, Santin AD, 2016, Mutational landscape of uterine and ovarian carcinosarcomas implicates histone genes in epithelial-mesenchymal transition. Proc Natl Acad Sci U S A 113(43):12238–12243 Chen X, Arend R, Hamele-Bena D, Tergas AI, Hawver M, Tong GX, Wright TC, Wright JD, 2017, Uterine Carcinosarcomas: clinical, Histopathologic and Immunohistochemical characteristics. Int J Gynecol Pathol 36(5):412–419 Jones NL, Xiu J, Chatterjee-Paer S, Buckley de Meritens A, Burke WM, Tergas AI, Wright JD, Hou JY, 2017, Distinct molecular landscapes between endometrioid and nonendometrioid uterine carcinomas. Int J Cancer 140(6):1396–1404 Lu X, Zhang L, Zhao H, Chen C, Wang Y, Liu S, Lin X,Wang Y, Zhang Q, Lu T, Yan F, 2019)Molecular classification and subtypespecific drug sensitivity research of uterine carcinosarcoma under multi-omics framework. Cancer Biol Ther 20(2):227–235 Ashley CW, Da Cruz PA, Kumar R et al, 2019, Analysis of mutational signatures in primary and metastatic endometrial cancer reveals distinct patterns of DNA repair defects and shifts during tumor progression. Gynecol Oncol 152(1):11–19 Franceschi T, Durieux E, Morel AP, de Saint Hilaire P, Ray- Coquard I, Puisieux A, Devouassoux-Shisheboran M, 2019, Role of epithelial-mesenchymal transition factors in the histogenesis of uterine carcinomas. Virchows Arch 475(1):85–94 Liu Y, Weber Z, San Lucas FA, Deshpande A, Jakubek YA, Sulaiman R, Fagerness M, Flier N, Sulaiman J, Davis CM, Fowler J, Starks D, Rojas-Espaillat L, Lazar AJ, Davies GE, Ehli EA, Scheet P, 2018, Assessing inter-component heterogeneity of biphasic uterine carcinosarcomas. Gynecol Oncol 151(2):243–249 McConechy MK, Hoang LN, Chui MH et al, 2015, In-depth molecular profiling of the biphasic components of uterine carcinosarcomas. J Pathol Clin Res 1(3):173–185 Cherniack AD, Shen H, Walter V, Stewart C, Murray BA, Bowlby R, Hu X, Ling S, Soslow RA, Broaddus RR, Zuna RE, Robertson G, Laird PW, Kucherlapati R, MillsGB, Weinstein JN, Zhang J,Akbani R, Levine DA, Akbani R, Ally A,Auman JT, BalasundaramM, Balu S, Baylin SB, Beroukhim R, Bodenheimer T, Bogomolniy F, Boice L, Bootwalla MS, Bowen J, Bowlby R, Broaddus R, Brooks D, Carlsen R, Cherniack AD, Cho J, Chuah E, Chudamani S, Cibulskis K, Cline M, Dao F, David M, Demchok JA, Dhalla N, Dowdy S, Felau I, Ferguson ML, Frazer S, Frick J, Gabriel S, Gastier-Foster JM, Gehlenborg N, Gerken M, Getz G, Gupta M, Haussler D, Hayes DN, Heiman DI, Hess J, Hoadley KA, Hoffmann R, Holt RA, Hoyle AP, Hu X, Huang M, Hutter CM, Jefferys SR, Jones SJM, Jones CD, Kanchi RS, Kandoth C, Kasaian K, Kerr S, Kim J, Lai PH, Laird PW, Lander E, Lawrence MS, Lee D, Leraas KM, Leshchiner I, Levine DA, Lichtenberg TM, Lin P, Ling S, Liu J, Liu W, Liu Y, Lolla L, Lu Y, Ma Y, Maglinte DT, Marra MA, Mayo M, Meng S, Meyerson M, Mieczkowski PA, Mills GB, Moore RA, Mose LE, Mungall AJ, Mungall K, Murray BA, Naresh R, Noble MS, Olvera N, Parker JS, Perou CM, Perou AH, Pihl T, Radenbaugh AJ, Ramirez NC, RathmellWK, Roach J, Robertson AG, Sadeghi S, Saksena G, Salvesen HB, Schein JE, Schumacher SE, Shen H, Sheth M, Shi Y, Shih J, Simons JV, Sipahimalani P, Skelly T, Sofia HJ, Soloway MG, Soslow RA, Sougnez C, Stewart C, Sun C, Tam A, Tan D, Tarnuzzer R, Thiessen N, Thorne LB, Tse K, Tseng J, van den Berg DJ, Veluvolu U, Verhaak RGW, Voet D, von Bismarck A, Walter V, Wan Y, Wang Z, Wang C, Weinstein JN, Weisenberger DJ, Wilkerson MD, Winterhoff B, Wise L, Wong T, Wu Y, Yang L, Zenklusen JC, Zhang J(J), Zhang H, ZhangW, Zhu JC, Zmuda E, Zuna RE, 2017, Integrated molecular characterization of uterine Carcinosarcoma. Cancer Cell 31(3):411–423 Le Gallo M, Rudd ML, Urick ME et al, 2018, The FOXA2 transcription factor is frequently somatically mutated in uterine carcinosarcomas and carcinomas. Cancer 124(1):65–73 Gotoh O, Sugiyama Y, Takazawa Y, Kato K, Tanaka N, Omatsu K, Takeshima N, Nomura H, Hasegawa K, Fujiwara K, Taki M, Matsumura N,Noda T,Mori S, 2019, Clinically relevant molecular subtypes and genomic alteration-independent differentiation in gynecologic carcinosarcoma. Nat Commun 10(1):4965 Prueksaritanond N, Chantape W, 2016, Comparative survival outcomes of uterine papillary serous carcinoma, clear cell carcinoma, grade 3 endometrioid adenocarcinoma, and carcinosarcoma of endometrial cancer in Rajavithi Hospital. J Med Assoc Thail 99(Suppl 2):S75–S83 Zhu J, Wen H, Bi R, Wu X, 2016, Clinicopathological characteristics, treatment and outcomes in uterine carcinosarcoma and grade 3 endometrial cancer patients: a comparative study. J Gynecol Oncol 27(2):e18 Zhang C, Hu W, Jia N, Li Q, Hua K, Tao X, Wang L, Feng W, 2015, Uterine carcinosarcoma and high-risk endometrial carcinomas: a clinicopathological comparison. Int J Gynecol Cancer 25(4): 629–636 Altrabulsi B, Malpica A, Deavers MT, Bodurka DC, Broaddus R, Silva EG, 2005, Undifferentiated carcinoma of the endometrium. Am J Surg Pathol 29(10):1316–1321 Taskin OC, Onder S, Topuz S et al, 2017, A selected immunohistochemical panel aids in differential diagnosis and prognostic stratification of subtypes of high-grade endometrial carcinoma: a clinicopathologic and immunohistochemical study at a single institution. Appl Immunohistochem Mol Morphol 25(10):696–702 Stewart CJ, Crook ML, 2015, SWI/SNF complex deficiency and mismatch repair protein expression in undifferentiated and dedifferentiated endometrial carcinoma. Pathology 47(5):439–445 Coatham M, Li X, Karnezis AN, Hoang LN, Tessier-Cloutier B, Meng B, Soslow RA, Blake Gilks C, Huntsman DG, Stewart CJR, Postovit LM, Kobel M, Lee CH, 2016, Concurrent ARID1A and ARID1B inactivation in endometrial and ovarian dedifferentiated carcinomas. Mod Pathol 29(12):1586–1593 Ramalingam P, Croce S, McCluggage WG, 2017, Loss of expression of SMARCA4, BRG1), SMARCA2, BRM, and SMARCB1, INI1, in undifferentiated carcinoma of the endometrium is not uncommon and is not always associated with rhabdoid morphology. Histopathology 70(3):359–366 Rosa-Rosa JM, Leskela S, Cristobal-Lana E, Santon A, Lopez- Garcia MA, Munoz G, Perez-Mies B, Biscuola M, Prat J, Esther OE, Soslow RA, Matias-Guiu X, Palacios J, 2016, Molecular genetic heterogeneity in undifferentiated endometrial carcinomas. Mod Pathol 29(11):1390–1398 Espinosa I, Lee CH, D'Angelo E, Palacios J, Prat J, 2017, Undifferentiated and dedifferentiated endometrial carcinomas with POLE exonuclease domain mutations have a favorable prognosis. Am J Surg Pathol 41(8):1121–1128 Kobel M, Hoang LN, Tessier-Cloutier B, Meng B, Soslow RA, Stewart CJR, Lee CH, 2018, Undifferentiated endometrial carcinomas show frequent loss of Core switch/sucrose nonfermentable complex proteins. Am J Surg Pathol 42(1):76–83 Murali R, Davidson B, Fadare O, Carlson JA, Crum CP, Gilks CB, Irving JA,Malpica A, Matias-Guiu X,McCluggageWG, Mittal K, Oliva E, Parkash V, Rutgers JKL, Staats PN, Stewart CJR, Tornos C, Soslow RA, 2019, High-grade endometrial carcinomas: morphologic and Immunohistochemical features, diagnostic challenges and recommendations. Int J Gynecol Pathol 38(Suppl 1):S40–S63 Stelloo E, Bosse T, Nout RA, MacKay HJ, Church DN, Nijman HW, Leary A, Edmondson RJ, Powell ME, Crosbie EJ, Kitchener HC, Mileshkin L, Pollock PM, Smit VT, Creutzberg CL, 2015, Refining prognosis and identifying targetable pathways for highrisk endometrial cancer; a TransPORTEC initiative. Mod Pathol 28(6):836–844 Meng B, Hoang LN, McIntyre JB et al, 2014, POLE exonuclease domain mutation predicts long progression-free survival in grade 3 endometrioid carcinoma of the endometrium. Gynecol Oncol 134(1):15–19 Hacking S, Jin C, Komforti M, Liang S, Nasim M, 2019, MMR deficient undifferentiated/dedifferentiated endometrial carcinomas showing significant programmed death ligand-1 expression, sp 142, with potential therapeutic implications. Pathol Res Pract 22: 152552. , DOI 10.1016/j.prp.2019.152552 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2067 EP 2073 DI 10.1007/s12253-020-00829-9 PG 7 ER PT J AU Travaglino, A Pace, M Varricchio, S Russo, D Pugliese, N Severino, A Picardi, M Pane, F Insabato, L Staibano, S Mascolo, M AF Travaglino, Antonio Pace, Mirella Varricchio, Silvia Russo, Daniela Pugliese, Novella Severino, Alessandro Picardi, Marco Pane, Fabrizio Insabato, Luigi Staibano, Stefania Mascolo, Massimo TI Involvement of Helicobacter Pylori in Ocular Adnexa Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Orbit; Ocular adnexa; Lymphoma; Helicobacter pylori; Antibiotic; Extranodal ID Orbit; Ocular adnexa; Lymphoma; Helicobacter pylori; Antibiotic; Extranodal AB Helicobacter pylori has been proposed as a possible etiologic factor of ocular adnexa lymphoma (OAL), although with conflicting results. To assess the involvement of H. pylori in OAL, as (1) H. pylori DNA positivity on OAL specimens, and (2) prevalence of H. pylori gastric infection in patients with OAL. A systematic review of studies assessing H. pylori in patients with OAL was conducted by searching electronic databases from their inception toMay 2019. Pooled positivity for H. pylori in OAL specimens detected by polymerase chain reaction, and pooled prevalence of H. pylori gastric infection,were calculatedwith 95% confidence interval (CI). Eleven studies with 308 patients were included. Pooled positivity for H. pylori was 16.8% in all OALs and 22.7% in MALT OAL, with high heterogeneity among studies. Pooled prevalence of H. pylori gastric infection in patients with OAL was 34.7%, with low statistical heterogeneity. In conclusion, H. pylori seems to be involved in a subset of OAL, but the heterogeneity found needs to be investigated in further studies. The prevalence of H. pylori gastric infection in patients with OAL does not seem to differ from that of the general population. C1 [Travaglino, Antonio] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Pace, Mirella] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Varricchio, Silvia] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Russo, Daniela] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Pugliese, Novella] University of Naples “Federico II”, Department of Clinical Medicine and Surgery, Hematology SectionNaples, Italy. [Severino, Alessandro] University of Naples “Federico II”, Department of Clinical Medicine and Surgery, Hematology SectionNaples, Italy. [Picardi, Marco] University of Naples “Federico II”, Department of Clinical Medicine and Surgery, Hematology SectionNaples, Italy. [Pane, Fabrizio] University of Naples “Federico II”, Department of Clinical Medicine and Surgery, Hematology SectionNaples, Italy. [Insabato, Luigi] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Staibano, Stefania] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. [Mascolo, Massimo] University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, Via Sergio Pansini, 5, 80131 Naples, Italy. RP Mascolo, M (reprint author), University of Naples Federico II, School of Medicine, Department of Advanced Biomedical Sciences, Anatomic Pathology Unit, 80131 Naples, Italy. EM mmascol@gmail.com CR SassoneM, PonzoniM, Ferreri AJ, 2017, Ocular adnexal marginal zone lymphoma: Clinical presentation, pathogenesis, diagnosis, prognosis, and treatment. Best Pract Res Clin Haematol 30(1–2): 118–130 Olsen TG, Heegaard S, 2019, Orbital lymphoma. Surv Ophthalmol 64(1):45–66 Kalogeropoulos D, Papoudou-Bai A, Kanavaros P, Kalogeropoulos C, 2018, Ocular adnexal marginal zone lymphoma of mucosa-associated lymphoid tissue. Clin Exp Med 18(2):151–163 Ferreri AJ, Govi S, Ponzoni M, 2013, Marginal zone lymphomas and infectious agents. Semin Cancer Biol 23(6):431–440 Chan CC, Smith JA, Shen DF, Ursea R, LeHoang P, Grossniklaus HE, 2004, Helicobacter pylori, H. pylori, molecular signature in conjunctival mucosa-associated lymphoid tissue, MALT, lymphoma. Histol Histopathol 19(4):1219–1226 Chan CC, Shen D, Mochizuki M, Gonzales JA, Yuen HK, Guex- Crosier Y, Lehoang P, 2006, Detection of helicobacter pylori and chlamydia pneumoniae genes in primary orbital lymphoma. Trans Am Ophthalmol Soc 104:62–70 Ferreri AJ, Ponzoni M, Viale E et al, 2006, Association between helicobacter pylori infection and MALT-type lymphoma of the ocular adnexa: clinical and therapeutic implications. Hematol Oncol 24(1):33–37 Grunberger B, HauffW, Lukas J, Wohrer S, Zielinski CC, Streubel B, Chott A, Raderer M, 2006, 'Blind' antibiotic treatment targeting chlamydia is not effective in patients withMALT lymphoma of the ocular adnexa. Ann Oncol 17(3):484–487 Goebel N, Serr A, Mittelviefhaus H, Reinhard T, Bogdan C, Auw- Haedrich C, 2007, Chlamydia psittaci, helicobacter pylori and ocular adnexal lymphoma-is there an association? The German experience. Leuk Res 31(10):1450–1452 Sjo NC, Foegh P, Juhl BR, Nilsson HO, Prause JU, Ralfkiaer E, Wadstrom T, Heegaard S, 2007, Role of helicobacter pylori in conjunctival mucosa-associated lymphoid tissue lymphoma. Ophthalmology. 114(1):182–186 Gruenberger B, Woehrer S, Troch M, Hauff W, Lukas J, Streubel B, Muellauer L, Chott A, RadererM(2008, Assessment of the role of hepatitis C, helicobacter pylori and autoimmunity in MALT lymphoma of the ocular adnexa in 45 Austrian patients. Acta Oncol 47(3):355–359 Lee SB, Yang JW, Kim CS, 2008, The association between conjunctival MALT lymphoma and helicobacter pylori. Br J Ophthalmol 92(4):534–536 Decaudin D, Ferroni A, Vincent-Salomon A, Beldjord K, Validire P, de Cremoux P, Validire P, Plancher C, Mathiot C, Macintyre E, Asselain B, Girodet J, Mal F, Brousse N, Beretti JL, Dendale R, Lumbroso-le Rouic L, Hermine O, LecuitM(2010, Ocular adnexal lymphoma and helicobacter pylori gastric infection. Am J Hematol 85(9):645–649 Cai JP, Cheng JW,Ma XY, Li YZ, Li Y, Huang X,Wei RL, 2012, Lack of association of conjunctival MALT lymphoma with Chlamydiae or helicobacter pylori in a cohort of Chinese patients. Med Sci Monit 18(2):BR84–BR88 Ferreri AJ, Govi S, Pasini E et al, 2012, Chlamydophila psittaci eradication with doxycycline as first-line targeted therapy for ocular adnexae lymphoma: final results of an international phase II trial. J Clin Oncol 30(24):2988–2994 Ferreri AJ, Ponzoni M, Guidoboni M et al, 2005, Regression of ocular adnexal lymphoma after Chlamydia psittaci-eradicating antibiotic therapy. J Clin Oncol 23(22):5067–5073 Ferreri AJ, Ponzoni M, Guidoboni M et al, 2006, Bacteriaeradicating therapy with doxycycline in ocular adnexal MALT lymphoma: a multicenter prospective trial. J Natl Cancer Inst 98(19):1375–1382 Zelenetz AD, Gordon LI, Abramson JS, et al. NCCN Clinical Practice Guidelines in Oncology, NCCN Guidelines®, – B-Cell Lymphomas. Version 4.2019 – May 6, 2019 Lionetti R, De Luca M, Travaglino A et al, 2019, Treatments and overall survival in patients with Krukenberg tumor. Arch Gynecol Obstet 300:15–23. , DOI 10.1007/s00404-019-05167-z Raffone A, Travaglino A, Saccone G, Cieri M, Mascolo M, Mollo A, Insabato L, Zullo F, 2019, Diagnostic and prognostic value of ARID1A in endometrial hyperplasia: a novel marker of occult cancer. APMIS. 127:597–606. , DOI 10.1111/apm.12977 Raffone A, Travaglino A, Saccone G, D’Alessandro P, Arduino B, Mascolo M, de Placido G, Insabato L, Zullo F, 2019, Diabetes mellitus is associated with occult Cancer in endometrial hyperplasia. Pathol Oncol Res 26:1377–1384. , DOI 10.1007/ s12253-019-00684-3 Moher D, Shamseer L, Clarke M et al, 2015, Preferred reporting items for systematic review and meta-analysis protocols, PRISMAP, 2015 statement. Syst Rev 4:1 Whiting PF, RutjesAW, WestwoodME et al, 2011, QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med 155(8):529–536 Raffone A, Travaglino A, Saccone G, Alviggi C, Mascolo M, de Placido G, Insabato L, Mollo A, Zullo F, 2019, Management of women with atypical polypoid adenomyoma of the uterus: a quantitative systematic review. Acta Obstet Gynecol Scand 98:842–855. , DOI 10.1111/aogs.13553 Travaglino A, Raffone A, Saccone G, de Luca C, Mollo A, Mascolo M, de Placido G, Insabato L, Zullo F, 2019, Immunohistochemical nuclear expression of β-catenin as a surrogate of CTNNB1 exon 3 mutation in endometrial Cancer. Am J Clin Pathol 151(5):529–538. , DOI 10.1093/ajcp/aqy178 Travaglino A, Raffone A, Saccone G, Mascolo M, Pignatiello S, Mollo A, de Placido G, Insabato L, Zullo F, 2019, PTEN immunohistochemistry in endometrial hyperplasia: which are the optimal criteria for the diagnosis of precancer? APMIS. 127(4):161–169 Raffone A, Travaglino A, Saccone G, Mascolo M, Insabato L, Mollo A, de Placido G, Zullo F, 2019, PAX2 in endometrial carcinogenesis and in differential diagnosis of endometrial hyperplasia. A systematic review and meta-analysis of diagnostic accuracy. Acta Obstet Gynecol Scand 98(3):287–299 Travaglino A, Raffone A, Saccone G, Mollo A, de Placido G, Mascolo M, Insabato L, Zullo F, 2019, Complexity of glandular architecture should be reconsidered in the classification and management of endometrial hyperplasia. APMIS. 127:427–434. https:// doi.org/10.1111/apm.12945 Raffone A, Travaglino A, Saccone G, di Maio A, Mollo A, Mascolo M, de Rosa R, de Placido G, Insabato L, Zullo F, 2019, Diabetes mellitus and responsiveness of endometrial hyperplasia and early endometrial cancer to conservative treatment. Gynecol Endocrinol 5:1–6. , DOI 10.1080/09513590.2019. 1624716 Qin Y, Greiner A, Trunk MJ, Schmausser B, Ott MM, Muller- Hermelink HK, 1995, Somatic hypermutationin low-grade mucosa- associated lymphoid tissue-type B-cell lymphoma. Blood 86: 3528–3534 Parsonnet J, Isaacson PG, 2004, Bacterial infection and MALT lymphoma. N Engl J Med 350:213–215 Parsonnet J, Hansen S, Rodriguez L, Gelb AB,Warnke RA, Jellum E, Orentreich N, Vogelman JH, Friedman GD, 1994, Helicobacter pylori infection and gastric lymphoma. N Engl J Med 330:1267– 1271 Yamasaki R, Yokota K, Okada H, Hayashi S, Mizuno M, Yoshino T, Hirai Y, Saitou D, Akagi T, Oguma K, 2004, Immune response in helicobacter pylori-induced low-grade gastric-mucosa-associated lymphoid tissue, MALT, lymphoma. J Med Microbiol 53:21–29 Travaglino A, Pace M, Varricchio S, Insabato L, Giordano C, Picardi M, Pane F, Staibano S, Mascolo M, 2020, Hashimoto thyroiditis in primary thyroid non-Hodgkin lymphoma. Am J Clin Pathol 153(2):156–164 Travaglino A, Pace M, Varricchio S, Insabato L, Picardi M, Severino A, Pane F, Staibano S, Mascolo M, 2020, Clinical features associated with high pathological grade in primary thyroid lymphoma. Pathol Res Pract 216(3):152819 Travaglino A, Giordano C, Pace M, Varricchio S, Picardi M, Pane F, Staibano S, Mascolo M, 2020, Sjogren syndrome in primary salivary gland lymphoma. Am J Clin Pathol 153(6):719–724 Travaglino A, Pace M, Varricchio S, Della Pepa R, Iuliano A, Picardi M, Pane F, Staibano S, Mascolo M, 2020, Prevalence of chlamydia psittaci, chlamydia pneumoniae, and chlamydia trachomatis determined by molecular testing in ocular adnexa lymphoma specimens. Am J Clin Pathol 153(4):427–434 Travaglino A, Varricchio S, Pace M, Iuliano A, Picardi M, Tranfa F, Staibano S, Mascolo M, 2020, Hepatitis C virus in MALTlymphoma of the ocular adnexa. Pathol Res Pract 216(4):152864 Gasbarrini A, Franceschi F, 1999, Autoimmune diseases and helicobacter pylori infection. Biomed Pharmacother 53:223–226 Ferreri AJ, Guidoboni M, Ponzoni M et al, 2004, Evidence for an association between chlamydia psittaci and ocular adnexal lymphomas. J Natl Cancer Inst 96(8):586–594 Ferreri AJ, Dolcetti R, Dognini GP et al, 2008, Chlamydophila psittaci is viable and infectious in the conjunctiva and peripheral blood of patients with ocular adnexal lymphoma: results of a singlecenter prospective case-control study. Int J Cancer 123(5):1089– 1093 Daibata M, Nemoto Y, Togitani K, Fukushima A, Ueno H, Ouchi K, Fukushi H, Imai S, Taguchi H, 2006, Absence of chlamydia psittaci in ocular adnexal lymphoma from Japanese patients. Br J Haematol 132(5):651–652 Liu YC, Ohyashiki JH, Ito Y, Iwaya KI, Serizawa H, Mukai K, Goto H, Usui M, Ohyashiki K, 2006, Chlamydia psittaci in ocular adnexal lymphoma: Japanese experience. Leuk Res 30(12):1587– 1589 Yakushijin Y, Kodama T, Takaoka I, Tanimoto K, Bessho H, Sakai I, Hato T, Hasegawa H, Yasukawa M, 2007, Absence of chlamydial infection in Japanese patients with ocular adnexal lymphoma of mucosa-associated lymphoid tissue. Int J Hematol 85(3):223–230 Usui Y, Rao NA, Takase H, Tsubota K, Umazume K, Diaz-Aguilar D, Kezuka T, Mochizuki M, Goto H, Sugita S, 2016, Comprehensive polymerase chain reaction assay for detection of pathogenic DNA in lymphoproliferative disorders of the ocular adnexa. Sci Rep 6:36621 Yoo C, Ryu MH, Huh J, Park JH, Kang HJ, Ahn HS, Lee Y, Kim MJ, Lee H, Kim TW, Chang HM, Lee JL, Kang YK, 2007, Chlamydia psittaci infection and clinicopathologic analysis of ocular adnexal lymphomas in Korea. Am J Hematol 82(9):821–823 Choung HK, Kim YA, Lee MJ, Kim N, Khwarg SI, 2012, Multigene methylation analysis of ocular adnexal MALT lymphoma and their relationship to Chlamydophila psittaci infection and clinical characteristics in South Korea. Invest Ophthalmol Vis Sci 53(4):1928–1935 Lee MJ,Min BJ, Choung HK, Kim N, Kim YA, Khwarg SI, 2014, Genome-wide DNA methylation profiles according to Chlamydophila psittaci infection and the response to doxycycline treatment in ocular adnexal lymphoma. Mol Vis 20:1037–1047 Kim TM, KimKH, Lee MJ, JeonYK, Lee SH, KimDW, KimCW, Kim IH, Khwarg SI, Heo DS, 2010, First-line therapy with doxycycline in ocular adnexal mucosa-associated lymphoid tissue lymphoma: a retrospective analysis of clinical predictors. Cancer Sci 101(5):1199–1203 Hooi JKY, Lai WY, Ng WK, Suen MMY, Underwood FE, Tanyingoh D, Malfertheiner P, Graham DY, Wong VWS, Wu JCY, Chan FKL, Sung JJY, Kaplan GG, Ng SC, 2017, Global prevalence of helicobacter pylori infection: systematic review and meta-analysis. Gastroenterology. 153(2):420–429 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2075 EP 2081 DI 10.1007/s12253-020-00848-6 PG 7 ER PT J AU Patai, BB Peterfy, N Szakacs, N Sapi, Z Hetthessy, RJ AF Patai, Bettina Bernadett Peterfy, Nora Szakacs, Noemi Sapi, Zoltan Hetthessy, Reka Judit TI Papillary Endothelial Hyperplasia (Masson Tumor) of the Hand. Surgical and Pathological Consideration from Seven Cases Using New Vascular Markers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE Hand; Soft tissue lesion; Vascular tumor; Vascular malformation; Intravascular papillary endothelial hyperplasia; IPEH; Masson tumor ID Hand; Soft tissue lesion; Vascular tumor; Vascular malformation; Intravascular papillary endothelial hyperplasia; IPEH; Masson tumor AB Although papillary endothelial hyperplasia may occur at almost any site, one of the most common sites is the hand. It is generally regarded as a reactive vascular proliferation i.e. exuberant form of organizing thrombus. Diagnosis of Masson tumor can be challenging due to its close clinical, radiological and even histopathological resemblance to angiosarcoma. We present seven cases ofMasson tumor of the hand; wanting to reveal its nature using new vascular markers and discuss the treatment options and expected outcomes, present clinical and radiological features that may aid diagnosis and also offer treatment plans. A multicenter retrospective study was performed between January 2014 and November 2019. Immunohistochemical stains of Glut1, WT1, ERG, CD31 and alpha smooth muscle actin (ASMA) were performed on each cases. We found seven cases during the examined period. 4 out of 7 cases were women. All lesions occurred in the hands. 3 out of 7 cases appeared in a previously present vascular malformation. All cases were treated with surgical excision and the diagnosis of papillary endothelial hyperplasia was made by histology. Pre-operative testing (radiograph/MRI/US/fine needle aspiration biopsy) did not suggest the diagnosis of Masson tumor; however, aspiration cytology could rule out malignancy. The proliferative endothelial cells proved to be Glut1 negative and WT1 positive and the accompanying pericytic cells were ASMA positive in all cases. Though Masson tumor is a rare vascular lesion in the hand among other vascular tumors, it should be considered in the differential diagnostics even in the case of previously existing vascular malformation.WT1 positivity of the endothelial cells and the accompanying pericytic cells raises the question whether the initially reactive endothelial proliferation may transform into a true benign vascular tumor. C1 [Patai, Bettina Bernadett] Military Health Centre, Department of TraumatologyBudapest, Hungary. [Peterfy, Nora] Saint John’s Hospital, Department of Traumatology and Hand surgeryBudapest, Hungary. [Szakacs, Noemi] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Hetthessy, Reka Judit] Semmelweis University, Department of OrthopedicsBudapest, Hungary. RP Hetthessy, RJ (reprint author), Semmelweis University, Department of Orthopedics, Budapest, Hungary. EM doktorno@kezklinika.hu CR Acharya G, Merritt WH, Theogaraj SD, 1980, Hemangioendotheliomas of the hand: case reports. J Hand Surg [Am] 5:181–182 Al Dhaybi R, Powell J, McCuaig C, Kokta V.(2010, Differentiation of vascular tumors from vascular malformations by expression of Wilms tumor 1 gene: evaluation of 126 cases. J Am Acad Dermatol. Dec;63(6):1052–7 Angela Sofia Cesar Faustino, 2019, Vascular malformations and tumors. Semmelweis University Medicine Faculty Semmelweis Orthopeadics Department, A therapeutical approach Buetow DC, Kransdorf MJ, Moser RP Jr, Jelink JS, Berrey BH, 1990,, 1990, radiological appearance of intra-muscular haemangioma with emphasis on MR imaging. AJR 154:563–567 Caroline Leclercq.(2014, Vascular tumors of the hand from 10th congress of the Asia-Pacific Federation of Societies of surgery fo the hand and the 6th congress of Asia-Pacific Federation of Societies of hand therapists Kuala Lumpur, Malaysia 2–4 October 2014 Clifford PD, Temple HT, JordaM,Marecos E, 2004, Intravascular papillary endothelial hyperplasia, Masson’s tumor, presenting as a triceps mass. Skelet Radiol 33:421–425 Cohen A, Maly A, Azaz B, 2009, Intravascular papillary endothelial hyperplasia of the lower lip: surgical approach and review of the literature. Gerodontology. 26(4):305–308 Corni V, Lucchina S, Fusetti S, 2014, Is Masson’s tumor only a slow-growing benign neoplasm? A case report. Hand Surg 19(3): 445–447 Darcy Fernandes, 1 Daphine Caxias Travassos, 1 Tulio Morandin Ferrisse, 1 Elaine Maria Sgavioli Massucato, 1Claudia Maria Navarro, 1 Mirian Aparecida Onofre, 1 Jorge Esquiche Leon, 2 and Andreia Bufalino 1 , *, 2016, Oral Intravascular Papillary Endothelial Hyperplasia Associated with an Organizing Thrombus: Case Report and Immunohistochemical Analysis Case Rep Pathol. 2016: 1908767 Espinosa A, Gonzalez J, Garcia-Navas F, 2017, Intravascular pappilary endothelial hyperplasia at foot level: a case report and literature review. J Foot Ankle Surg 56(1):72–74 Finn MC, Glowacki J, Mulliken JB, 1983, Congenital vascular lesions: clinical application of a new classification. J Pediatr Surg 18:894–900 Hashimoto H, Daimaru Y, Enjoji M, 1983, Intravascular papillary endothelial hyperplasia: a clinicopathologic study of 91 cases.AmJ Dermatopathol 5:539–545 Holly, B.P., et al.,, 2017, Preoperative epoxy embolization facilitates the safe and effective resection of venous malformations in the hand and forearm. Hand, New York, N.Y.),. 12(4): p. 335–341 Leclercq, C.,, 2010, Vascular tumors of the hand. BMC proceedings, 2015. 9(Suppl 3) Lee SJ et al, 2009, Imaging findings of intravascular papillary endothelial hyperplasia presenting in extremities: correlation with pathological findings. Skelet Radiol 39(8):783–789 Levere SM, Barsky SH, Meals RA, 1994, Intravascular papillary endothelial hyperplasia: a neoplastic ‘actor’ representing an exaggerated attempt at recanalization mediated by basic fibroblast growth factor. J Hand Surg Am 19:559–564 Liron Pantanowitz* and Wayne H Duke., 2008, Intravascular lesions of the hand Department of Pathology, Baystate Medical Center, Tufts University School of Medicine, Springfield, MA, USA Lysyy O, Schwartz I, Kolander Y, Strauss S, 2011, Sonographic features of intravascular papillary endothelial hyperplasia, Masson’s tumor, in the forearm. J Clin Ultrasound 39:301–303 Masson P, 1923, Hemangioendotheliome vegetant intravasculaire. Bull Soc Anat 93:517–523 Peterson MS, Baron RL, Rankin SC, 2000, Hepatic angiosarcoma: findings on multiphasic contrast-enhanced helical CT do not mimic hepatic hemangioma. AJR Am J Roentgenol 175:165–170 Requena L, Sangueza OP, 1997, Cutaneous vascular proliferations. Part II Hyperplasias and benign neoplasms J Am Acad Dermatol 37:887–919 SalyerWR, Salyer DC, 1975, Intravascular angiomatosis: development and distinction from angiosarcoma. Cancer. 36(3):995–1001 Samadi K, Salazar GM., 2019, Role of imaging in the diagnosis of vascular malformations. Cardiovasc Diagn Ther. Aug;9(Suppl 1): S143-S151 Soliman YS, Khachemoune A, 2018, Infantile hemangiomas: our current understanding and treatment options. Dermatol Online J Sep 15:24(9) Stefanos Boukovalas,MD*, Rachel Dillard, BS†, Suimin Qiu,MD, PhD‡,Eric L Cole,MD*., 2017, Intravascular papillary endothelial hyperplasia, Masson’s tumor): diagnosis the plastic surgeon should be aware of. Plast Reconstr Surg Glob Open;5:e1122 Suh JS, Hwang G, Hahn SB, 1994, Soft-tissue hemangi-omas:MR manifestations in 23 patients. Skelet Radiol 23:621–625 Tedla M, BezovaM, Biro C, Tedlova E, Eng CY, Zelenik K, 2014, Intravascular papillary endothe-lial hyperplasia of larynx: case report and literature review of all head and neck cases. Otolaryngol Pol 68:200–203 Teo Soleymani, MD,* Michael Abrouk, BS,† and Kristen M. Kelly, MD† Dermatol Surg., 2017 May, An Analysis of Laser Therapy for the Treatment of Nonmelanoma Skin Cancer; 43(5): 615–624 Timar J, Meszaros L, Orosz Z, Albini A, Raso E, 2005 Jul, WT1 expression in angiogenic tumours of the skin. Histopathology. 47(1):67–73 Vito Pesce,1 Davide Bizzoca,1 Angela Notarnicola,1 Andrea Piazzolla,1 Giovanni Vicenti, 1 Antonietta Cimmino,2Francesco Fortarezza,2 Giuseppe Maccagnano, 1 Giuseppe Solarino,1 and Biagio Moretti1, 2018, An intravascular papillary endothelial hyperplasia of the hand radiologically mimicking a hemangiopericytoma: A case report and literature review SAGE Open Med Case Rep.; 6: 2050313X17752851 Vjay Shankar,M.D., 2003-2018, Soft tissue benign vascular tumors, Intravascular papillary endothelial hyperplasia, pathologyOutlines.com,Inc Walker EA, Salesky JS, Fenton ME, Murphey MD, 2011, Magnetic resonance imaging of malignant soft tissue neoplasms in the adult. Radiol Clin N Am 49:1219–1234 Walker EA, Song AJ, Murphey MD, 2010, Magnetic resonance imaging of soft-tissue masses. Semin Roentgenol 45:277–297 Young RJ, Brown NJ, Reed MW, Hughes D, Woll PJ, 2010, Angiosarcoma. Lancet Oncol 11:983–991 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2083 EP 2090 DI 10.1007/s12253-020-00838-8 PG 8 ER PT J AU Molla, S Hembram, ChK Chatterjee, S Nayak, D Sethy, Ch Pradhan, R Kundu, NCh AF Molla, Sefinew Hembram, Chandra Krushna Chatterjee, Subhajit Nayak, Deepika Sethy, Chinmayee Pradhan, Rajalaxmi Kundu, Nath Chanakya TI PARP inhibitor Olaparib Enhances the Apoptotic Potentiality of Curcumin by Increasing the DNA Damage in Oral Cancer Cells through Inhibition of BER Cascade SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral cancer; Olaparib; Curcumin; PARP inhibitor; PARylation ID Oral cancer; Olaparib; Curcumin; PARP inhibitor; PARylation AB Although Olaparib (Ola, a PARP-inhibitor), in combination with other chemotherapeutic agents, was clinically approved to treat prostate cancer, but cytotoxicity, off-target effects of DNA damaging agents limit its applications in clinic. To improve the anticancer activity and to study the detailed mechanism of anti-cancer action, here we have used bioactive compound curcumin (Cur) in combination with Ola. Incubation of Ola in Cur pre-treated cells synergistically increased the death of oral cancer cells at much lower concentrations than individual optimum dose and inhibited the topoisomerase activity. Short exposure of Cur caused DNA damage in cells, but more increased DNA damage was noticed when Ola has incubated in Cur pre-treated cells. This combination did not alter the major components of homologous recombination (HR) and non-homologous end-joining (NHEJ) pathways but significantly altered both short patch (SP) and long patch (LP) base excision repair (BER) components in cancer cells. Significant reduction in relative luciferase activity, expression of BER components and PARylation after Cur and Ola treatment confirmed this combination inhibit the BER activity in cells. Reduction of PARylation, decreased expression of BER components, decreased tumor volume and induction of apoptosis were also noticed in Cur + Ola treated Xenograft mice model. The combination treatment of Cur and Ola also helped in recovering the body weight of tumor-bearing mice. Thus, Cur + Ola combination increased the oral cancer cells death by not only causing the DNA damage but also blocking the induction of BER activity. C1 [Molla, Sefinew] Deemed to be University, KIIT School of Biotechnology, Kalinga Institute of Industrial Technology, Cancer Biology Division, Campus-11, Patia, 751024 Bhubaneswar, Odisha, India. [Hembram, Chandra Krushna] Deemed to be University, KIIT School of Biotechnology, Kalinga Institute of Industrial Technology, Cancer Biology Division, Campus-11, Patia, 751024 Bhubaneswar, Odisha, India. [Chatterjee, Subhajit] Deemed to be University, KIIT School of Biotechnology, Kalinga Institute of Industrial Technology, Cancer Biology Division, Campus-11, Patia, 751024 Bhubaneswar, Odisha, India. [Nayak, Deepika] Deemed to be University, KIIT School of Biotechnology, Kalinga Institute of Industrial Technology, Cancer Biology Division, Campus-11, Patia, 751024 Bhubaneswar, Odisha, India. [Sethy, Chinmayee] Deemed to be University, KIIT School of Biotechnology, Kalinga Institute of Industrial Technology, Cancer Biology Division, Campus-11, Patia, 751024 Bhubaneswar, Odisha, India. [Pradhan, Rajalaxmi] Deemed to be University, KIIT School of Biotechnology, Kalinga Institute of Industrial Technology, Cancer Biology Division, Campus-11, Patia, 751024 Bhubaneswar, Odisha, India. [Kundu, Nath Chanakya] Deemed to be University, KIIT School of Biotechnology, Kalinga Institute of Industrial Technology, Cancer Biology Division, Campus-11, Patia, 751024 Bhubaneswar, Odisha, India. RP Kundu, NCh (reprint author), Deemed to be University, KIIT School of Biotechnology, Kalinga Institute of Industrial Technology, Cancer Biology Division, 751024 Bhubaneswar, India. EM cnkundu@gmail.com;cnkundu@kiitbiotech.ac.in CR Naik PP, Das DN, Panda PK, Mukhopadhyay S, Sinha N, Praharaj PP, Agarwal R, Bhutia SK, 2016, Implications of cancer stem cells in developing therapeutic resistance in oral cancer. Oral Oncol 62: 122–135 Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics. CA Cancer J Clin 63(11):30 Dufour R, Daumar P, Mounetou E, 2015, BCRP and P-gp relay overexpression in triple negative basal-like breast cancer cell line: a prospective role in resistance to Olaparib. Sci Rep 5:12670 Gelbard A, Garnett CT, Abrams SI, Patel V, Gutkind JS, Palena C, Tsang KY, Schlom J, Hodge JW, 2006, Combination chemotherapy and radiation of human squamous cell carcinoma of the head and neck augments CTL-mediated lysis. Clin Cancer Res 12:1897– 1905 Nandakumar DN, Nagaraj VA, Vathsala PG, Rangarajan P, Padmanaban G, 2006, Curcumin-artemisinin combination therapy for malaria. Antimicrob Agents Chemother 50:1859–1860 Gupta SC, Patchva S, Koh W, Aggarwal BB, 2012, Discovery of curcumin, a component of golden spice, and its miraculous biological activities. Clin Exp Pharmacol Physiol 39:283–299 Satoskar RR, Shah SJ, Shenoy SG, 1986, Evaluation of antiinflammatory property of curcumin, diferuloyl methane, in patients with postoperative inflammation. Int J Clin Pharmacol Ther Toxicol 24(12):651–654 Shao ZM, Shen ZZ, Liu CH, Sartippour MR, Go VL, Heber D, Nguyen M, 2002, Curcumin exerts multiple suppressive effects on human breast carcinoma cells. Int J Cancer 9:234–240 Park CH, Hahm ER, Park S, Kim HK, Yang CH, 2005, The inhibitory mechanism of curcumin and its derivative against beta-catenin/ Tcf signaling. FEBS Lett 579(13):2965–2971 Kunnumakkara AB, Anand P, Aggarwal BB, 2008, Curcumin inhibits proliferation, invasion, angiogenesis and metastasis of different cancers through interaction with multiple cell signaling proteins. Cancer Lett 269:199–225 Shang HS, Chang CH, Chou YR, Yeh MY, Au MK, Lu HF, Chu YL, Chou HM, Chou HC, Shih YL, Chung JG, 2016, Curcumin causes DNA damage and affects associated protein expression in HeLa human cervical cancer cells. Oncol Rep 36:2207–2215 Duvoix A, Blasius R, Delhalle S, Schnekenburger M, Morceau F, Henry E, Dicato M, Diederich M, 2005, Chemopreventive and therapeutic effects of curcumin. Cancer Lett 223:181–190 Ting CY, Wang HE, Yu CC, Liu HC, Liu YC, Chiang IT, 2015, Curcumin triggers DNA damage and inhibits expression of DNA repair proteins in human lung cancer cells. Anticancer Res 35: 3867–3873 Zhao Q,Guan J, Qin Y, Ren P, Zhang Z, Lv J, Sun S, Zhang C, Mao W, 2018, Curcumin sensitizes lymphoma cells to DNA damage agents through regulating Rad51-dependent homologous recombination. Biomed Pharmacother 97:115–119 Ogiwara H, Ui A, Shiotani B, Zou L, Yasui A, Kohno T, 2013, Curcumin suppresses multiple DNA damage response pathways and has potency as a sensitizer to a PARP inhibitor. Carcinogenesis 34:2486–2497 Kumar A, Bora U, 2013, Interactions of curcumin and its derivatives with nucleic acids and their implications. Mini-Rev Med Chem 13:256–264 Caiola E, Salles D, Frapolli R, Lupi M, Rotella G, Ronchi A, Garassino MC, Mattschas N, Colavecchio S, Broggini M, Wiesmuller L, 2015, Base excision repair-mediated resistance to cisplatin in KRAS, G12C, mutant NSCLC cells. Oncotarget 6: 30072 Underhill C, ToulmondeM, Bonnefoi H, 2011, A review of PARP inhibitors: from bench to bedside. Ann Oncol 22:268–279 Memisoglu A, Samson L, 2000, Base excision repair in yeast and mammals. Mutat Res 451:39–51 Schreiber V, Ame JC, Dolle P, Schultz I, Rinaldi B, Fraulob V, Menissier-de Murcia J, de Murcia G, 2002, Poly, ADP-ribose, polymerase-2, PARP-2, is required for efficient base excision DNA repair in association with PARP-1 and XRCC1. J Biol Chem 277:23028–23036 Almeida KH, Sobol RW, 2007, A unified view of base excision repair: lesion-dependent protein complexes regulated by posttranslational modification. DNA Repair, Amst, 6:695–711 Kundu CN, Balusu R, Jaiswal AS, Gairola CG, Narayan S, 2007, Cigarette smoke condensate-induced level of adenomatous polyposis coli blocks long-patch base excision repair in breast epithelial cells. Oncogene 26:1428–1438 Lopez-Lazaro M, Willmore E, Jobson A, Gilroy KL, Curtis H, Padget K, Austin CA, 2007, Curcumin induces high levels of topoisomerase I- and II-DNA complexes in K562 leukemia cells. J Nat Prod 70(12):1884–1888 Donawho CK, Luo Y, Luo Y, Penning TD, Bauch JL, Bouska JJ, Bontcheva-DiazVD, Cox BF, DeWeese TL,Dillehay LE, Ferguson DC, 2007, ABT-888, an orally active poly, ADP-ribose, polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models. Clin Cancer Res 13:2728–2737 Narod SA, 2010, BRCA mutations in the management of breast cancer. Nat Rev Clin Oncol 7:702–707 Evans T, Matulonis U, 2017, PARP inhibitors in ovarian cancer: evidence, experience and clinical potential. Ther AdvMed Oncol 9: 253–267 Siddharth S, Nayak D, Nayak A, Das S, Kundu CN, 2016, ABT- 888 and Quinacrine induced apoptosis in metastatic breast cancer stem cells by inhibiting base excision repair via adenomatous polyposis coli. DNA Repair, Amst, 45:44–55 Mohapatra P, Satapathy SR, Siddharth S, Das D, Nayak A, Kundu CN(2015, Resveratrol and curcumin synergistically induce apoptosis in cigarette smoke condensate transformed breast epithelial cells through a p21Waf1/Cip1mediated inhibition of Hh-Gli signaling. Int J Biochem Cell Biol 66:75–84 Preet R, Mohapatra P, Das D, Satapathy SR, Choudhuri T, Wyatt MD, Kundu CN, 2013, Lycopene synergistically enhances Quinacrine action to inhibit Wnt-TCF signaling in breast cancer cells through APC. Carcinogenesis 34:277–286 Mohapatra P, Preet R, Das D, Satapathy SR, Siddharth S, Choudhuri T, Wyatt MD, Kundu CN, 2014, The contribution of heavy metals in cigarette smoke condensate to malignant transformation of breast epithelial cells and in vivo initiation of neoplasia through induction of a PI3K-AKT-NFκB cascade. Toxicol Appl Pharmacol 274:168–179 Das S, Tripathi N, Siddharth S, Nayak A, Nayak D, Sethy C, Bharatam PV, Kundu CN, 2017, Etoposide and doxorubicin enhance the sensitivity of triple negative breast cancers through modulation of TRAIL-DR5 axis. Apoptosis 22:1205–1224 Wang JC, 2002, Cellular roles of DNA topoisomerases: a molecular perspective. Nat Rev Mol Cell Biol 3:430 Wang L, Eastmond DA, 2002, Catalytic inhibitors of topoisomerase II are DNA-damaging agents: induction of chromosomal damage by merbarone and ICRF-187. Environ Mol Mutagen 39:348– 356 Martin-Cordero C, Lopez-Lazaro M, Galvez M, Ayuso MJ, 2003, Curcumin as a DNA topoisomerase II poison. J Enzyme Inhibition Med Chem 18:505–509 Nitiss JL, 2009, Targeting DNA topoisomerase II in cancer chemotherapy. Nat Rev Cancer 9:338 Prasad R, Horton JK, Dai DP, Wilson SH, 2019, Repair pathway for PARP-1 DNA-protein crosslinks. DNA Repair 73:71–77 Wielgos M, Yang ES, 2013, Discussion of PARP inhibitors in cancer therapy. Pharm Pat Anal 2:755–766 Snyder RD, ArnoneMR, 2002, Putative identification of functional interactions between DNA intercalating agents and topoisomerase II using the V79 in vitro micronucleus assay. Mutat Res 503(1-2): 21–35 Prasad CB, Prasad SB, Yadav SS, Pandey LK, Singh S, Pradhan S, Narayan G, 2017, Olaparib modulates DNA repair efficiency, sensitizes cervical cancer cells to cisplatin and exhibits anti-metastatic property. Sci Rep 7:12876 Andrabi SA, Kim NS, Yu SW,Wang H, Koh DW, Sasaki M, Klaus JA, Otsuka T, Zhang Z, Koehler RC, Hurn PD, 2006, Poly, ADPribose,, PAR, polymer is a death signal. Proc Natl Acad Sci 103: 18308–18313 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2091 EP 2103 DI 10.1007/s12253-019-00768-0 PG 13 ER PT J AU Kato, K Miyazawa, H Kobayashi, H Noguchi, N Lambert, D Kawashiri, Sh AF Kato, Koroku Miyazawa, Hiroki Kobayashi, Hisano Noguchi, Natsuyo Lambert, Daniel Kawashiri, Shuichi TI Caveolin-1 Expression at Metastatic Lymph Nodes Predicts Unfavorable Outcome in Patients with Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Caveolin-1; p16; Oral squamous cell carcinoma; Metastatic lymph node; Prognosis ID Caveolin-1; p16; Oral squamous cell carcinoma; Metastatic lymph node; Prognosis AB We evaluated the clinical and prognostic value of the protein expression of caveolin-1 (CAV1) and p16 at the primary site and metastatic lymph nodes of oral squamous cell carcinoma (OSCC). Primary site specimens from 80 OSCC cases were randomly selected and lymph node specimens from 15 preserved metastatic lymph nodes from among those patients were selected for examination. We evaluated the CAV1 and p16 expression at both the primary site and metastatic lymph nodes, and analyzed the patients’ clinicopathological data in relation to CAV1 and p16 expression. Our analysis revealed significant positive correlations between CAV1 expression at the primary site and pathological metastasis, cell differentiation, and mode of invasion (p = 0.019, p = 0.002, p = 0.015, respectively), but p16 expression was not associated with any clinicopathological factors. Patients with high CAV1 expression at the primary sites showed significantly worse prognoses than those with low or negative CAV1 expression (p = 0.002), and multivariate analysis showed that the T-classification and CAV1 expression were independent OSCC prognostic factors. CAV1 expression was also present in the metastatic lymph nodes of the OSCC cases with particularly poor differentiation and high invasive grade, and patients with CAV1-positive metastatic lymph nodes showed significantly worse prognoses than those with CAV1-negative metastatic lymph nodes (p = 0.018). CAV1 may activate metastaticity and the invasive capacity of OSCC cells. CAV1 expression, particularly at metastatic lymph nodes, predicts a worse outcome for OSCC, suggesting that CAV1 could be used as a prognostic marker for OSCC. C1 [Kato, Koroku] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Miyazawa, Hiroki] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Kobayashi, Hisano] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Noguchi, Natsuyo] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. [Lambert, Daniel] University of Sheffield, School of Clinical Dentistry, Unit of Oral and Maxillofacial PathologySheffield, UK. [Kawashiri, Shuichi] Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 13-1 Takara-machi, 920-8641 Kanazawa, Japan. RP Kato, K (reprint author), Kanazawa University, Graduate School of Medical Science, Department of Oral and Maxillofacial Surgery, 920-8641 Kanazawa, Japan. EM k-koroku@oral.m.kanazawa-u.ac.jp CR Parkin DM, Bray F, Ferlay J, Pisani P, 2005, Global cancer statistics, 2002. CA Cancer J Clin 55:74–108 Vokes EE, Weichselbaum RR, Lippman SM, Hong WK, 1993, Head and neck cancer. N Engl J Med 328:184–194 Forastiere A, KochW, Trotti A, Sidransky D, 2001, Head and neck cancer. N Engl J Med 345:1890–1900 Yamamoto E, Miyakawa A, Kohama G, 1984, Mode of invasion and lymph node metastasis in squamous cell carcinoma of oral cavity. Head Neck 6:939–947 Jarvinen AK, Autio R, Kilpinen S, SaarelaM, Leivo I, Grenman R, Makitie AA, Monni O, 2008, High-resolution copy number and gene expression microarray analyses of head and neck squamous cell carcinoma cell lines of tongue and larynx. Genes Chromosom Cancer 47:500–509 Nakayama S, Sasaki A, Mese H, Alcalde RE, Matsumura T, 1998- 1999, Establishment of high and low metastasis cell lines derived from a human tongue squamous cell carcinoma. Invasion Metastasis 18:219–228 Wiechen K, Diatchenko L, Agoulnik A, Scharff KM, Schober H, Arlt K, Zhumabayeva B, Siebert PD, Dietel M, Schafer R, Sers C, 2001, Caveolin-1 is down-regulated in human ovarian carcinoma and acts as a candidate tumor suppressor gene. Am J Pathol 159: 1635–1643 Kato K, Hida Y, Miyamoto M, Hashida H, Shinohara T, Itoh T, Okushiba S, Kondo S, KatohH, 2002)Overexpression of caveolin- 1 in esophageal squamous cell carcinoma correlates with lymph node metastasis and pathologic stage. Cancer 94:929–933 Karam JA, Lotan Y, Roehrborn CG, Ashfaq R, Karakiewicz PI, Shariat SF, 2007, Caveolin-1 overexpression is associated with aggressive prostate cancer recurrence. Prostate 67:614–622 Ang KK,Harris J,Wheeler R,Weber R, Rosenthal DI, Nguyen-Tan PF, Westra WH, Chung CH, Jordan RC, Lu C, Kim H, Axelrod R, Silverman CC, Redmond KP, Gillison ML, 2010, Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 363:24–35 Allen CT, Lewis JS Jr, El-Mofty SK, Haughey BH, Nussenbaum B, 2010, Human papillomavirus and oropharynx cancer: biology, detection and clinical implications. Laryngoscope 120:1756–1772 Marur S, D'Souza G, Westra WH, Forastiere AA, 2010, HPVassociated head and neck cancer: a virus-related cancer epidemic. Lancet Oncol 11:781–789 Wang MB, Liu IY, Gornbein JA, Nguyen CT, 2015, HPV-positive Oropharyngeal carcinoma: a systematic review of treatment and prognosis. Otolaryngol Head Neck Surg 153:758–769 In, 2009, International Union against Cancer, LH Sobin, MK Gospodrowicz, CH Wittekind, eds.). TNM Classification of Malignant Tumours. 7th Ed. Wiley-Liss, New York, NY Yamamoto E, Kohama G, Sunagawa H, Iwai M, Hiratsuka H, 1983)Mode of invasion, bleomycin sensitivity, and clinical course in squamous cell carcinoma of the oral cavity. Cancer 51:2175– 2180 Heiduschka G, Grah A, Oberndorfer F, Kadletz L, Altorjai G, Kornek G,Wrba F, Thurnher D, Selzer E, 2015, Improved survival in HPV/p16-positive oropharyngeal cancer patients treated with postoperative radiotherapy. Strahlenther Onkol 191:209–216 Glenney JR Jr, Zokas L, 1989, Novel tyrosine kinase substrates from Rous sarcoma virus-transformed cells are present in the membrane skeleton. J Cell Biol 108:2401–2408 Rothberg KG, Heuser JE, Donzell WC, Ying YS, Glenney JR, Anderson RG, 1992, Caveolin, a protein component of caveolae membrane coats. Cell 68:673–682 Lu Z, Ghosh S, Wang Z, Hunter T, 2003, Downregulation of caveolin-1 function by EGF leads to the loss of E-cadherin, increased transcriptional activity of beta-catenin, and enhanced tumor cell invasion. Cancer Cell 4:499–515 Burgermeister E, Xing X, Rocken C, Juhasz M, Chen J, Hiber M, Mair K, Shatz M, Liscovitch M, Schmid RM, Ebert MP, 2007, Differential expression and function of caveolin-1 in human gastric cancer progression. Cancer Res 67:8519–8526 Li S, Couet J, LisantiMP, 1996, Src tyrosine kinases, Gα subunits, and H-Ras share a common membrane-anchored scaffolding protein, caveolin. Caveolin binding negatively regulates the autoactivation of Src tyrosine kinases. J Biol Chem 271:29182–29190 Couet J, Sargiacomo M, LisantiMP, 1997, Interaction of a receptor tyrosine kinase, EGF-R, with caveolins. Caveolin binding negatively regulates tyrosine and serine/threonine kinase activities. J Biol Chem 272:30429–30438 Razani B, Zhang XL, Bitzer M, von Gersdorff G, Bottinger EP, LisantiMP, 2001, Caveolin-1 regulates transforming growth factor, TGF)-beta/SMAD signaling through an interaction with the TGFbeta type I receptor. J Biol Chem 276:6727–6738 TorresVA, Tapia JC, Rodriguez DA, ParragaM, Lisboa P,Montoya M, Leyton L, Quest AF, 2006, Caveolin-1 controls cell proliferation and cell death by suppressing expression of the inhibitor of apoptosis protein survivin. J Cell Sci 119(Pt 9):1812–1823 Goetz JG, Lajoie P, Wiseman SM, Nabi IR, 2008, Caveolin-1 in tumor progression: the good, the bad and the ugly. Cancer Metastasis Rev 27:715–735 Senetta R, Stella G, Pozzi E, Sturli N, Massi D, Cassoni P, 2013, Caveolin-1 as a promoter of tumour spreading: when, how, where and why. J Cell Mol Med 17:325–336 Fu P, Chen F, Pan Q, Zhao X, Zhao C, ChoWC, Chen H(2017, The different functions and clinical significances of caveolin-1 in human adenocarcinoma and squamous cell carcinoma. Onco Targets Ther 10:819–835 Ho CC, Huang PH, Huang HY, Chen YH, Yang PC, Hsu SM, 2002, Up-regulated caveolin-1 accentuates the metastasis capability of lung adenocarcinoma by inducing filopodia formation. Am J Pathol 161:1647–1656 Chen HL, Fan LF, Gao J, Ouyang JP, ZhangYX, 2011, Differential expression and function of the caveolin-1 gene in non-small cell lung carcinoma. Oncol Rep 25:359–366 Cassoni P, Daniele L, Maldi E, Righi L, Tavaglione V, Novello S, Volante M, Scagliotti GV, Papotti M, 2009, Caveolin-1 expression in lung carcinoma varies according to tumour histotype and is acquired de novo in brain metastases. Histopathology 55:20–27 Ando T, Ishiguro H, Kimura M, Mitsui A, Mori Y, Sugito N, Tomoda K, Mori R, Harada K, Katada T, Ogawa R, Fujii Y, Kuwabara Y, 2007, The overexpression of caveolin-1 and caveolin-2 correlates with a poor prognosis and tumor progression in esophageal squamous cell carcinoma. Oncol Rep 18:601–609 Liu Z, Yu J, Wu R, Tang S, Cai X, Guo G, Chen S, 2017, Rho/ ROCK pathway regulates migration and invasion of esophageal squamous cell carcinoma by regulating Caveolin-1. Med Sci Monit 23:6174–6185 Diaz-Valdivia N, Bravo D, Huerta H, Henriquez S, Gabler F, Vega M, Romero C, Calderon C, Owen GI, Leyton L, Quest AF, 2015, Enhanced caveolin-1 expression increases migration, anchorageindependent growth and invasion of endometrial adenocarcinoma cells. BMC Cancer 15:463 Yamaguchi H, Takeo Y,Yoshida S, Kouchi Z, NakamuraY, Fukami K, 2009, Lipid rafts and caveolin-1 are required for invadopodia formation and extracellular matrix degradation by human breast cancer cells. Cancer Res 69:8594–8602 Wang R, Li Z, Guo H, Shi W, Xin Y, Chang W, Huang T, 2014, Caveolin 1 knockdown inhibits the proliferation, migration and invasion of human breast cancer BT474 cells. Mol Med Rep 9: 1723–1728 Auzair LB, Vincent-Chong VK, Ghani WM, Kallarakkal TG, Ramanathan A, Lee CE, Rahman ZA, Ismail SM, Abraham MT, Zain RB, 2016, Caveolin 1, Cav-1, and actin-related protein 2/3 complex, subunit 1B, ARPC1B, expressions as prognostic indicators for oral squamous cell carcinoma, OSCC). Eur Arch Otorhinolaryngol 273:1885–1893 Ketteler J, Klein D, 2018, Caveolin-1, cancer and therapy resistance. Int J Cancer 143:2092–2104 Wang Z, Wang N, Li W, Liu P, Chen Q, Situ H, Zhong S, Guo L, Lin Y, Shen J, Chen J, 2014, Caveolin-1 mediates chemoresistance in breast cancer stem cells via β-catenin/ABCG2 signaling pathway. Carcinogenesis 35:2346–2356 Sun S, Hong SA, Won HS, Yoo SH, Lee HH, Kim O, Ko YH, 2017, Prognostic value of metastatic Tumoral Caveolin-1 expression in patients with resected gastric Cancer. Gastroenterol Res Pract 2017:5905173 Williams TM, Medina F, Badano I, Hazan RB, Hutchinson J, Muller WJ, Chopra NG, Scherer PE, Pestell RG, Lisanti MP, 2004, Caveolin-1 gene disruption promotes mammary tumorigenesis and dramatically enhances lung metastasis in vivo. Role of Cav-1 in cell invasiveness and matrix metalloproteinase, MMP-2/ 9, secretion. J Biol Chem 279:51630–51646 Ang KK,Harris J,Wheeler R,Weber R, Rosenthal DI, Nguyen-Tan PF, Westra WH, Chung CH, Jordan RC, Lu C, Kim H, Axelrod R, Silverman CC, Redmond KP, Gillison ML, 2010, Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl JMed 363:24–35 Namazie A, Alavi S, Olopade OI, Pauletti G, Aghamohammadi N, Aghamohammadi M, Gornbein JA, Calcaterra TC, Slamon DJ, Wang MB, Srivatsan ES, 2002, Cyclin D1 amplification and p16(MTS1/CDK4I)deletion correlate with poor prognosis in head and neck tumors. Laryngoscope 112:472–481 Zafereo ME, Xu L, Dahlstrom KR, Viamonte CA, El-Naggar AK, Wei Q, Li G, Sturgis EM, 2016, Squamous cell carcinoma of the oral cavity often overexpresses p16 but is rarely driven by human papillomavirus. Oral Oncol 56:47–53 Kouketsu A, Sato I, Abe S, Oikawa M, Shimizu Y, Takahashi T, Kumamoto H, 2016, Detection of human papillomavirus infection in oral squamous cell carcinoma: a cohort study of Japanese patients. J Oral Pathol Med 45:565–572 Gotz C, Drecoll E, Straub M, Bissinger O, Wolff KD, Kolk A, 2016, Impact of HPV infection on oral squamous cell carcinoma. Oncotarget 7:76704–76712 Nopmaneepaisarn T, Tangjaturonrasme N, Rawangban W, Vinayanuwattikun C, Keelawat S, Bychkov A, 2019, Low prevalence of p16-positive HPV-related head-neck cancers in Thailand: tertiary referral center experience. BMC Cancer 19:1050 Sieczkarski SB, Whittaker GR, 2005, Viral entry. Curr Top Microbiol Immunol 285:1–23 Smith JL, Campos SK, Ozbun MA, 2007, Human papillomavirus type 31 uses a caveolin 1- and dynamin 2-mediated entry pathway for infection of human keratinocytes. J Virol 81:9922–9931 Smith JL, Campos SK, Wandinger-Ness A, Ozbun MA, 2008, Caveolin-1-dependent infectious entry of human papillomavirus type 31 in human keratinocytes proceeds to the endosomal pathway for pH-dependent uncoating. J Virol 82:9505–9512 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2105 EP 2113 DI 10.1007/s12253-019-00791-1 PG 9 ER PT J AU Oikawa, M Tanaka, T Narita, T Noro, D Iwamura, H Tobisawa, Y Yoneyama, T Kodama, H Hashimoto, Y Koie, T Ohyama, Ch AF Oikawa, Masaaki Tanaka, Toshikazu Narita, Takuma Noro, Daisuke Iwamura, Hiromichi Tobisawa, Yuki Yoneyama, Tohru Kodama, Hirotake Hashimoto, Yasuhiro Koie, Takuya Ohyama, Chikara TI Impact of the Proportion of Biopsy Positive Core in Predicting Biochemical Recurrence in Patients with Pathological Pt2 and Negative Resection Margin Status after Radical Prostatectomy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Organ-confined prostate cancer; Negative surgical margin; Radical prostatectomy; Biochemical recurrence; Proportion of biopsy positive core; Biopsy Gleason score ID Organ-confined prostate cancer; Negative surgical margin; Radical prostatectomy; Biochemical recurrence; Proportion of biopsy positive core; Biopsy Gleason score AB This study aimed to determine the prognostic factors associated with biochemical recurrence (BCR) after radical prostatectomy (RP) in patients with pathological T2 (pT2) prostate cancer (PCa) and negative resection margin (RM) status at a single institution. In this retrospective study, we examined 386 patients who were diagnosed with pT2 PCa with negative RM after RP. The length of the tumor was provided for each biopsy core and the overall percentage of PCa was calculated by a pathologist at our institution. We estimated the BCR-free survival (BRFS) in these patients. Univariate and multivariate analyses were performed using the Cox proportional hazard model to determine the risk factors of BCR. The median age of the participants was 68 years, and their initial prostate-specific antigen level was 6.55 ng/ml. The median follow-up period was 85.7 months. The 5-year BRFS rate of the participants was 89.0%. The 5-year BRFS rates were 89.8% in patients with a biopsy Gleason score of 6, 90.4% in those with 7, and 64.1% in those with ≥8 (P = 0.007). The BRFS rate was 93.3% in patients who had a biopsy positive core ≤20% and 82.0% in those who had ≥21% (P = 0.001). Based on the multivariate analysis, the proportion of biopsy positive core was significantly associated with BCR. The proportion of biopsy positive core may predict preoperative covariates in patients with pT2 PCa and negative RM status after RP. C1 [Oikawa, Masaaki] Hakodate Municipal Hospital, Department of Urology, 041-8680 Hakodate, Japan. [Tanaka, Toshikazu] Aomori Prefectural Central Hospital, Department of Urology, 030-8553 Aomori, Japan. [Narita, Takuma] Hirosaki National Hospital, Department of Urology, 036-8545 Hirosaki, Japan. [Noro, Daisuke] Mutsu General Hospital, Department of Urology, 035-8601 Mutsu, Japan. [Iwamura, Hiromichi] Oyokyo Kidney Research Institute, Department of Urology, 038-0003 Aomori, Japan. [Tobisawa, Yuki] Hirosaki University Graduate School of Medicine, Department of Urology, 036-8562 Hirosaki, Japan. [Yoneyama, Tohru] Hirosaki University Graduate School of Medicine, Department of Urology, 036-8562 Hirosaki, Japan. [Kodama, Hirotake] Tsugaru General Hospital, Department of Urology, 037-0074 Goshogawara, Japan. [Hashimoto, Yasuhiro] Hirosaki University Graduate School of Medicine, Department of Urology, 036-8562 Hirosaki, Japan. [Koie, Takuya] Gifu University Graduate School of Medicine, Department of Urology, Yanagito 1-1, 501-1194 Gifu, Japan. [Ohyama, Chikara] Hirosaki University Graduate School of Medicine, Department of Urology, 036-8562 Hirosaki, Japan. RP Koie, T (reprint author), Gifu University Graduate School of Medicine, Department of Urology, 501-1194 Gifu, Japan. EM goodwin@gifui-u.ac.jp CR Global Burden of Disease Cancer Collaboration, Fitzmaurice C, Dicker D, Pain A et al, 2015, The global burden of Cancer 2013. JAMA Oncol 1:505–527 Projected Cancer Statistics, 2018. Cancer Information Services. https://ganjoho.jp/en/public/statistics/short_pred.html Accessed 15 September 2018 Gettman MT, Blute ML, 2010, Radical prostatectomy: does surgical technique influence margin control? Urol Oncol 28:219–225 Rampersaud EN, Sun L, Moul JW et al, 2008, Percent tumor involvement and risk of biochemical progression after radical prostatectomy. J Urol 180:571–576 Ploussard G, Agamy MA, Alenda O et al, 2011, Impact of positive surgical margins on prostate-specific antigen failure after radical prostatectomy in adjuvant treatment-naive patients. BJU Int 107: 1748–1754 Chang SS, CooksonMS, 2006, Impact of positive surgicalmargins after radical prostatectomy. Urology 68:249–252 Zhang LJ, Wu B, Zha ZL et al, 2018, Perineural invasion as an independent predictor of biochemical recurrence in prostate cancer following radical prostatectomy or radiotherapy: a systematic review and meta-analysis. BMC Urol 18:5 Mitsuzuka K, Narita S, Koie T et al, 2015, Lymphovascular invasion is significantly associated with biochemical relapse after radical prostatectomy even in patients with pT2N0 negative resection margin. Prostate Cancer Prostatic Dis 18:25–30 Srougi V, Bessa J Jr, Baghdadi M et al, 2017, Surgical method influences specimen margins and biochemical recurrence during radical prostatectomy for high-risk prostate cancer: a systematic review and meta-analysis. World J Urol 35:1481–1488 Hernandez DJ, Nielsen ME, Han M et al, 2008, Natural history of pathologically organ-confined, pT2), Gleason score 6 or less, prostate cancer after radical prostatectomy. Urology 72:172–176 Budaus L, Isbarn H, Eichelberg C et al, 2010, Biochemical recurrence after radical prostatectomy: multiplicative interaction between surgical margin status and pathological stage. J Urol 184:1341–1346 Hamano I, Hatakeyama S, Yoneyama T et al, 2016, Safety of heparin bridging therapy for transrectal ultrasound-guided prostate biopsy in patients requiring temporary discontinuation of antithrombotic agents. Springerplus 5:1917 Weinreb JC, Barentsz JO, Choyke PL et al, 2016)PI-RADS prostate imaging - reporting and data system: 2015, version 2. Eur Urol 69: 16–40 Koie T, Yamamoto H, Hatakeyama S et al, 2011, Minimum incision endoscopic radical prostatectomy: clinical and oncological outcomes at a single institute. Eur J Surg Oncol 37:805–810 Fujita N, Koie T, Hashimoto Y et al, 2018, Neoadjuvant chemohormonal therapy followed by robot-assisted and minimum incision endoscopic radical prostatectomy in patients with high-risk prostate cancer: comparison of perioperative and oncological outcomes at single institution. Int Urol Nephrol 50:1999–2005 Narita T, Koie T, Ookubo T et al, 2017, The impact of extended lymph node dissection versus neoadjuvant therapy with limited lymph node dissection on biochemical recurrence in high-risk prostate cancer patients treated with radical prostatectomy: a multiinstitutional analysis. Med Oncol 34:1 Epstein JI, Allsbrook WC Jr, Amin MB et al, 2005, The 2005 International Society of Urological Pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol 29:1228–1242 American Joint Committee on Cancer, 2010, Urinary bladder. In: Edge SB, Byrd DR, Compton CC et al, ed, AJCC Cancer staging manual, 7th Edition. Springer, New York, pp457–468 Isbarn H,WannerM, Salomon G et al, 2010)Long-term data on the survival of patients with prostate cancer treated with radical prostatectomy in the prostate-specific antigen era. BJU Int 106:37–43 Kotb AF, Elabbady AA, 2011, Prognostic factors for the development of biochemical recurrence after radical prostatectomy. Prostate Cancer 2011:485189 Karakiewicz PI, Eastham JA, Graefen M et al, 2005, Prognostic impact of positive surgical margins in surgically treated prostate cancer: multi-institutional assessment of 5831 patients. Urology 66:1245–1250 Galiabovitch E, Hovens CM, Peters JS et al, 2017, Routinely reported 'equivocal' lymphovascular invasion in prostatectomy specimens is associated with adverse outcomes. BJU Int 119:567–572 Lee JT, Lee S, Yun CJ et al, 2010, Prediction of perineural invasion and its prognostic value in patients with prostate cancer. Korean J Urol 51:745–751 Yu HH, Song DY, Tsai YYet al, 2007, Perineural invasion affects biochemical recurrence-free survival in patients with prostate cancer treated with definitive external beam radiotherapy. Urology 70:111–116 Ishida M, Nakashima J, Hashiguchi A et al, 2009, Are predictive models for cancer volume clinically useful in localized prostate cancer? Int J Urol 16:936–940 Tan CH, Wei W, Johnson V, Kundra V, 2012)Diffusion-weighted MRI in the detection of prostate cancer: meta-analysis. AJR Am J Roentgenol 199:822–829 Barentsz JO, Richenberg J, Clements R et al, 2012, European Society of Urogenital Radiology. ESUR prostate MR guidelines 2012. Eur Radiol 22:746–757 Berney DM, Beltran L, Sandu H et al, 2019, The percentage of high-grade prostatic adenocarcinoma in prostate biopsies significantly improves on grade groups in the prediction of prostate cancer death. Histopathology. 75:589–597 Guedes LB, Almutairi F, Haffner MC et al, 2017, Analytic, preanalytic, and clinical validation of p53 IHC for detection of TP53 missense mutation in prostate cancer. Clin Cancer Res 23: 4693–4703 Yoneyama T, Tobisawa Y, Kaneko T et al, 2019, Clinical significance of the LacdiNAc-glycosylated prostate-specific antigen assay for prostate cancer detection. Cancer Sci 110:2573–2589 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2115 EP 2121 DI 10.1007/s12253-019-00762-6 PG 7 ER PT J AU Kuthi, L Somoracz, Micsik, T Jenei, A Hajdu, A Sejben, I Imre, D Posfai, B Koczian, K Semjen, D Bajory, Z Kulka, J Ivanyi, B AF Kuthi, Levente Somoracz, Aron Micsik, Tamas Jenei, Alex Hajdu, Adrienn Sejben, Istvan Imre, Daniel Posfai, Boglarka Koczian, Katalin Semjen, David Bajory, Zoltan Kulka, Janina Ivanyi, Bela TI Clinicopathological Findings on 28 Cases with XP11.2 Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Translocation renal cell carcinoma; Xp11.2; Immunohistochemistry; TFE3 gene; Fluorescence in situ hybridization (FISH) ID Translocation renal cell carcinoma; Xp11.2; Immunohistochemistry; TFE3 gene; Fluorescence in situ hybridization (FISH) AB Xp11.2 translocation carcinoma is a distinct subtype of renal cell carcinoma characterized by translocations involving the TFE3 gene. Our study included the morphological, immunohistochemical and clinicopathological examination of 28 Xp11.2 RCCs. The immunophenotype has been assessed by using CA9, CK7, CD10, AMACR, MelanA, HMB45, Cathepsin K and TFE3 immunostainings. The diagnosis was confirmed by TFE3 break-apart FISH in 25 cases. The ages of 13 male and 15 female patients, without underlying renal disease or having undergone chemotherapy ranged from 8 to 72. The mean size of the tumors was 78.5mm. Forty-three percent of patients were diagnosed in the pT3/pT4 stage with distant metastasis in 6 cases. Histological appearance was branching-papillary composed of clear cells with voluminous cytoplasmin 13 and variable in 15 cases, including one tumor with anaplastic carcinoma and another with rhabdoid morphology. Three tumors were labeled with CA9, while CK7 was negative in all cases. Diffuse CD10 reaction was observed in 17 tumors and diffuse AMACR positivity was described in 14 tumors. The expression of melanocytic markers and Cathepsin K were seen only in 7 and 6 cases, respectively. TFE3 immunohistochemistry displayed a positive reaction in 26/28 samples. TFE3 rearrangement was detected in all the analyzed cases (25/ 25), including one with the loss of the entire labeled break-point region. The follow-up time ranged from 2 to 300 months, with 7 cancer-related deaths. In summary, Xp11.2 carcinoma is an uncommon form of renal cell carcinoma with a variable histomorphology and rather aggressive clinical course. C1 [Kuthi, Levente] University of Szeged, Department of Pathology, 1 Allomas Street, H-6725 Szeged, Hungary. [Somoracz, Aron] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Micsik, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Jenei, Alex] University of Szeged, Department of Pathology, 1 Allomas Street, H-6725 Szeged, Hungary. [Hajdu, Adrienn] University of Szeged, Department of Pathology, 1 Allomas Street, H-6725 Szeged, Hungary. [Sejben, Istvan] Bacs-Kiskun County Hospital, Department of PathologyKecskemet, Hungary. [Imre, Daniel] Hetenyi Geza County Hospital, Department of PathologySzolnok, Hungary. [Posfai, Boglarka] University of Szeged, Department of OncotherapySzeged, Hungary. [Koczian, Katalin] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Semjen, David] University of Pecs, Department of PathologyPecs, Hungary. [Bajory, Zoltan] University of Szeged, Department of UrologySzeged, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Ivanyi, Bela] University of Szeged, Department of Pathology, 1 Allomas Street, H-6725 Szeged, Hungary. RP Kuthi, L (reprint author), University of Szeged, Department of Pathology, H-6725 Szeged, Hungary. EM kuthi.levente@med.u-szeged.hu CR Argani P, Faria M, 2005, Translocation carcinomas of the kidney. Clin Lab Med 25:363–378 Argani P, Antonescu CR, Illei PB et al, 2001, Primary renal neoplasms with the ASPL-TFE3 gene fusion of alveolar soft part sarcoma: a distinctive tumor entity previously included among renal cell carcinomas of children and adolescents. Am J Pathol 159:179– 192 Sidhar SK, Clark J, Gill S et al, 1996, The t(X;1)(p11.2;q21.2, translocation in papillary renal cell carcinoma fuses a novel gene PRCC to the TFE3 transcription factor gene. Hum Mol Genet 5: 1333–1338 Clark J, Lu YJ, Sidhar SK et al, 1997, Fusion of splicing factor genes PSF and NonO, p54nrb, to the TFE3 gene in papillary renal cell carcinoma. Oncogene 15:2233–2239 Argani P, Lui MY, Couturier J et al, 2003, A novel CLTC-TFE3 gene fusion in pediatric renal adenocarcinoma with t(X;17)(p11.2;q23). Oncogene 22:5374–5378 Argani P, Olgac S, Tickoo SK et al, 2007, Xp11 translocation renal cell carcinoma in adults: expanded clinical, pathologic and genetic spectrum. Am J Surg Pathol 31:1149–1160 ZhongM, De Angelo P, Osborne L et al, 2012, Translocation renal cell carcinomas in adults: a single-institution experience. Am J Surg Pathol 36:654–662 Qu Y, Gu C, Wang H et al., 2016, Diagnosis of adults Xp11.2 translocation renal cell carcinoma by immunohistochemistry and FISH assays: clinicopathological data from ethnic Chinese population. Sci rep 6:2167 Armah HB, Parwani AV, 2010, Xp11.2 translocation renal cell carcinoma. Arch Pathol Lab Med 134:124–129 Argani P, Cheville J, Ladanyi M, 2016, MiT family translocation renal cell carcinomas. In: Humphrey PA, Ulbright TM, Reuter VE, eds, Moch H. Pathology and Genetics of Tumours of the Urinary System and Male Genital Organs. IARCC Press Lyon, WHO Classification of Tumours, pp 33–34 Argani P, Hicks J, De Marzo AM et al, 2010, Xp11 translocation renal cell carcinoma, RCC): extended Immunohistochemical profile emphasizing novel RCC markers. Am J Surg Pathol 34:1295– 1303 Martignoni G, Gobbo S, Camparo P et al, 2011, Differential expression of cathepsin K in neoplasms harboring TFE3 gene fusions. Mod Pathol 24:1313–1319 Rao Q, Williamson SR, Zhang S et al, 2013, TFE3 break-apart FISH has a higher sensitivity for Xp11.2 translocation-associated renal cell carcinoma compared with TFE3 or cathepsin K immunohistochemical staining alone: expanding themorphologic spectrum. Am J Surg Pathol 37:804–815 Camparo P, Vasiliu V, Molinie V et al, 2008, Renal translocation carcinomas: clinicopathologic, immunohistochemical, and gene expression profiling analysis of 31 cases with a review of the literature. Am J Surg Pathol 32:656–670 Argani P, Lae M, Ballard ET et al, 2006, Translocation carcinomas of the kidney after chemotherapy in childhood. J Clin Oncol 24: 1529–1534 Malouf GG, Camparo P, Oudard S et al, 2010, Targeted agents in metastatic Xp11 translocation/TFE3 gene fusion renal cell carcinoma, RCC): a report from the juvenile RCC network. Ann Oncol 21: 1834–1838 Choueiri TK, Lim ZD, Hirsch MS et al, 2010, Vascular endothelial growth factor-targeted therapy for the treatment of adult metastatic Xp11.2 translocation renal cell carcinoma. Cancer 116:5219–5225 Argani P, Antonescu CR, Couturier J et al, 2002, PRCC-TFE3 renal carcinomas: morphologic, immunohistochemical, ultrastructural, and molecular analysis of an entity associated with the t(X;1)(p11.2;q21). Am J Surg Pathol 26:1553–1566 Kuthi L, Jenei A, Hajdu A et al, 2017, Prognostic factors for renal cell carcinoma subtypes diagnosed according to the 2016 WHO renal tumor classification: a study involving 928 patients. Pathol Oncol Res 23:689–698 Delahunt B,McKenney JK, Lohse CMet al, 2013)A novel grading system for clear cell renal cell carcinoma incorporating tumor necrosis. Am J Surg Pathol 37:311–322 Argani P, Ladanyi M, 2005, Translocation carcinomas of the kidney. Clin Lab Med 25:363–378 KomaiY, Fujiwara M, Fujii Yet al, 2009, Adult Xp11 translocation renal cell carcinoma diagnosed by cytogenetics and immunohistochemistry. Clin Cancer Res 15:1170–1176 Ellis CL, Eble JN, Subhawong AP et al, 2014, Clinical heterogeneity of Xp11 translocation renal cell carcinoma: impact of fusion subtype, age, and stage. Mod Pathol 27:875–886 Argani P, Lal P, Hutchinson B et al, 2003, Aberrant nuclear immunoreactivity for TFE3 in neoplasms with TFE3 gene fusions: a sensitive and specific immunohistochemical assay. Am J Surg Pathol 27:750–761 Yang B, Duan H, Cao Wet al, 2019, Xp11 translocation renal cell carcinoma and clear cell renal cell carcinoma with TFE3 strong positive immunostaining: morphology, immunohistochemistry, and FISH analysis. Mod Pathol 32:1521–1535 Argani P, Aulmann S, Illei PB et al, 2010, A distinctive subset of PEComas harbors TFE3 gene fusions. Am J Surg Pathol 34:1395– 1406 Martignoni G, Pea M, Gobbo S et al, 2009, Cathepsin-K immunoreactivity distinguishes MiTF/TFE family renal translocation carcinomas from other renal carcinomas. Mod Pathol 22:1016–1022 Green WM, Yonescu R, Morsberger L et al, 2013, Utilization of a TFE3 break-apart FISH assay in a renal tumor consultation service. Am J Surg Pathol 37:1150–1163 Pei J, Cooper H, Flieder DB et al, 2019, NEAT1-TFE3 and KAT6A-TFE3 renal cell carcinomas, new members of MiT family translocation renal cell carcinoma. Mod Pathol 32:710–716 Mosquera JM, Dal Cin P, Mertz KD et al, 2011, Validation of a TFE3 break-apart FISH assay for Xp11.2 translocation renal cell carcinomas. Diagn Mol Pathol 20:129–137 Argani P, Aulmann S, Karanjawala Z et al, 2009, Melanotic Xp11 translocation renal cancers: a distinctive neoplasmwith overlapping features of PEComa, carcinoma, and melanoma. Am J Surg Pathol 33:609–619 Papp G, Nagy D, Sapi Z, 2017, Unusual signal patterns of breakapart FISH probes used in the Diagnosis of Soft Tissue Sarcomas. Pathol Oncol Res 23:863–871 34. Kato I, Furuya M, Baba M et al, 2019, RBM10-TFE3 renal cell carcinoma characterised by paracentric inversion with consistent closely split signals in break-apart fluorescence in-situ hybridisation: study of 10 cases and a literature review. Histopathology 75:254–265 35. Argani P, LadanyiM(2003, Distinctive neoplasms characterised by specific chromosomal translocations comprise a significant proportion of the paediatric renal carcinomas. Pathology 35:492–498 36. Dal Cin P, Stas M, Sciot R et al, 1998, Translocation, X;1, reveals metastasis 31 years after renal cell carcinoma. Cancer Genet Cytogenet 101:58–61 37. Perot C, Bougaran J, Boccon-Gibod L et al, 1999, Two new cases of papillary renal cell carcinoma with t(X;1)(p11;q21, in females. Cancer Genet Cytogenet 110:54–56 38. Rais-Bahrami S, Drabick JJ, De Marzo AM et al, 2007, Xp11 translocation renal cell carcinoma: delayed but massive and lethal metastases of a chemotherapy-associated secondary malignancy. Urology 70:178e3–178e6 39. Mangel L, Biro K, Battyani I et al, 2015, A case study on the potential angiogenic effect of human chorionic gonadotropin hormone in rapid progression and spontaneous regression ofmetastatic renal cell carcinoma during pregnancy and after surgical abortion. BMC Cancer 15:1013 40. Patard JJ, Leray E, Rioux-Leclercq N et al, 2005, Prognostic value of histologic subtypes in renal cell carcinoma: a multicenter experience. J Clin Oncol 23:2763–2771 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2123 EP 2133 DI 10.1007/s12253-019-00792-0 PG 11 ER PT J AU Zhu, H You, Y Shen, Z Shi, L AF Zhu, Haifeng You, Yongping Shen, Zhouming Shi, Lei TI EGFRvIII-CAR-T Cells with PD-1 Knockout Have Improved Anti-Glioma Activity SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioblastoma multiform; EGFRvIII; CAR-T; PD-1; PD-L1 ID Glioblastoma multiform; EGFRvIII; CAR-T; PD-1; PD-L1 AB Glioblastoma multiforme (GBM) is the most malignant form of the brain tumors. EGFR variant III (EGFRvIII) is expressed in about 30% of GBM specimens, but not expressed in normal brain tissues. Therefore, EGFRvIII protein offers an ideal CAR-T therapeutic target for EGFRvIII-positive GBM patients. PD-L1 is expressed in a variety of cancer cells, including GBM. Tumor-associated PD-L1 can bind to PD-1 on T cells and promote apoptosis of T cells, thus suppressing the anti-cancer immune response. In our current studies, PD-1WT EGFRvIII-CAR-T cells and PD-1KD EGFRvIII-CAR-T cells were generated. Cytokine production and lytic activity of these two CAR-T cells against to PD-L1WT EGFRvIII+ U373 cells or PD-L1KO EGFRvIII+ U373 cells were evaluated. The results showed that PD-1KD EGFRvIII-CAR-T cells and PD-1WT EGFRvIII-CAR-T cells showed same levels of interferon-γ (IFN-γ) and interleukin-2 (IL-2) production as well as cytolytic activity against PD-L1KO EGFRvIII+ U373 cells; however, PD-1KD EGFRvIII-CAR-T cells exhibited higher levels of IFN-γ and IL-2 production as well as cytolytic activity against PD-L1+ EGFRvIII+ U373 cells than that of PD-1WT EGFRvIII-CAR-T cells. PD-1KD EGFRvIII-CAR-T cells also exhibited higher anti-glioma activity and longer survival in mice in vivo than that of PD-1WT EGFRvIII-CAR-T cells. Taken together, our findings indicate that PD-1 knockout enhances lytic activity of EGFRvIII-CAR-T cells against PD-L1+ EGFRvIII+ GBM cells. These might provide a new insight into strategy of GBM CAR-T cell therapy. C1 [Zhu, Haifeng] Nanjing University, School of Medicine, Jinling Hospital, Department of Neurosurgery, 210029 Nanjing, China. [You, Yongping] The First Affiliated Hospital of Nanjing Medical University, Department of Neurosurgery, 210029 Nanjing, China. [Shen, Zhouming] Funing People’s Hospital, Department of Neurosurgery, 224400 Funing, China. [Shi, Lei] Affiliated Kunshan Hospital of Jiangsu University, Department of Neurosurgery, 215300 Suzhou, China. RP Shi, L (reprint author), Affiliated Kunshan Hospital of Jiangsu University, Department of Neurosurgery, 215300 Suzhou, China. EM sl1012002322@126.com CR Schwartzbaum JA, Fisher JL, Aldape KD, Wrensch M, 2006, Epidemiology and molecular pathology of glioma. Nat Clin Pract Neurol 2(9): 494-503; quiz 1 p following 516 Oike T, Suzuki Y, Sugawara K, Shirai K, Noda SE, Tamaki T, Nagaishi M, Yokoo H, Nakazato Y, Nakano T, 2013, Radiotherapy plus concomitant adjuvant temozolomide for glioblastoma: Japanese mono-institutional results. PLoS One 8(11): e78943 GilbertMR,Wang M, Aldape KD, Stupp R, HegiME, Jaeckle KA, Armstrong TS, Wefel JS, Won M, Blumenthal DT, Mahajan A, Schultz CJ, Erridge S, Baumert B, Hopkins KI, Tzuk-Shina T, Brown PD, Chakravarti A, Curran WJ Jr, Mehta MP, 2013, Dose-dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trial. J Clin Oncol 31(32):4085–4091 Ekstrand AJ, Longo N, Hamid ML, Olson JJ, Liu L, Collins VP, James CD, 1994, Functional characterization of an EGF receptor with a truncated extracellular domain expressed in glioblastomas with EGFR gene amplification. Oncogene 9(8):2313–2320 Padfield E, Ellis HP, Kurian KM, 2015, Current therapeutic advances targeting EGFR and EGFRvIII in Glioblastoma. Front Oncol 5:5 Thorne AH, Zanca C, Furnari F, 2016, Epidermal growth factor receptor targeting and challenges in glioblastoma. Neuro-Oncology 18(7):914–918 Johnson LA, Scholler J,Ohkuri T, Kosaka A, Patel PR, McGettigan SE, Nace AK, Dentchev T, Thekkat P, Loew A, Boesteanu AC, Cogdill AP, Chen T, Fraietta JA, Kloss CC, Posey AD Jr, Engels B, Singh R, Ezell T, Idamakanti N, Ramones MH, Li N, Zhou L, Plesa G, Seykora JT, Okada H, June CH, Brogdon JL, Maus MV, 2015, Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor Tcells for glioblastoma. Sci Transl Med 7(275):275ra22 Krenciute G, Krebs S, Torres D, Wu MF, Liu H, Dotti G, Li XN, Lesniak MS, Balyasnikova IV, Gottschalk S, 2016, Characterization and functional analysis of scFv-based chimeric antigen receptors to redirect T cells to IL13Ralpha2-positive Glioma. Mol Ther 24(2):354–363 O'Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K,Morrissette JJD,Martinez-Lage M, Brem S,Maloney E, Shen A, Isaacs R, Mohan S, Plesa G, Lacey SF, Navenot JM, Zheng Z, Levine BL, Okada H, June CH, Brogdon JL, Maus MV, 2017, A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med 9(399):eaaa0984 Nishikori M, Takaori-Kondo A, 2017, PD-1 blockade therapy in lymphoid malignancies. Rinsho Ketsueki 58(10):2043–2049 Lipson EJ, Lilo MT, Ogurtsova A, Esandrio J, Xu H, Brothers P, Schollenberger M, SharfmanWH, Taube JM, 2017, Basal cell carcinoma: PD-L1/PD-1 checkpoint expression and tumor regression after PD-1 blockade. J Immunother Cancer 5:23 Serganova I, Moroz E, Cohen I, Moroz M, Mane M, Zurita J, Shenker L, Ponomarev V, Blasberg R, 2017, Enhancement of PSMA-directed CAR adoptive immunotherapy by PD-1/PD-L1 blockade. Mol Ther Oncolytics 4:41–54 Zak KM, Kitel R, Przetocka S, Golik P, Guzik K, Musielak B, Domling A, Dubin G, Holak TA, 2015, Structure of the complex of human programmed death 1, PD-1, and its ligand PD-L1. Structure 23(12):2341–2348 Thompson ED, Zahurak M, Murphy A, Cornish T, Cuka N, Abdelfatah E, Yang S, Duncan M, Ahuja N, Taube JM, Anders RA, Kelly RJ, 2017, Patterns of PD-L1 expression and CD8 T cell infiltration in gastric adenocarcinomas and associated immune stroma. Gut 66(5):794–801 MinchomA, Thavasu P, Ahmad Z, Stewart A,GeorgiouA,O'Brien MER, Popat S, Bhosle J, Yap TA, de Bono J, Banerji U, 2017, A study of PD-L1 expression in KRAS mutant non-small cell lung cancer cell lines exposed to relevant targeted treatments. PLoS One 12(10):e0186106 Kim HM, Lee J, Koo JS, 2017, Clinicopathological and prognostic significance of programmed death ligand-1 expression in breast cancer: a meta-analysis. BMC Cancer 17(1):690 Nduom EK, Wei J, Yaghi NK, Huang N, Kong LY, Gabrusiewicz K, Ling X, Zhou S, Ivan C, Chen JQ, Burks JK, Fuller GN, Calin GA, Conrad CA, Creasy C, Ritthipichai K, Radvanyi L, Heimberger AB, 2016, PD-L1 expression and prognostic impact in glioblastoma. Neuro-Oncology 18(2):195–205 Sengupta S, Katz SC, Sengupta S, Sampath P, 2018, Glycogen synthase kinase 3 inhibition lowers PD-1 expression, promotes long-term survival and memory generation in antigen-specific CAR-T cells. Cancer Lett 433:131–139 Li Y, QiuW, Zhang L, Fung J, Lin F, 2016, Painting factor H onto mesenchymal stem cells protects the cells from complement- and neutrophil-mediated damage. Biomaterials 102:209–219 Watkins MP, Bartlett NL, 2018, CD19-targeted immunotherapies for treatment of patients with non-Hodgkin B-cell lymphomas. Expert Opin Investig Drugs 27(7):601–611 Young PA, Yamada RE, Trinh KR, Vasuthasawat A, De Oliveira S, Yamada DH,Morrison SL, Timmerman JM(2018, Activity of anti- CD19 chimeric antigen receptor T cells against B cell lymphoma is enhanced by antibody-targeted interferon-alpha. J Interf Cytokine Res 38(6):239–254 Rabinovich GA, Gabrilovich D, Sotomayor EM, 2007, Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol 25:267–296 Heimberger AB, Hlatky R, Suki D, Yang D,Weinberg J, GilbertM, Sawaya R, Aldape K, 2005, Prognostic effect of epidermal growth factor receptor and EGFRvIII in glioblastoma multiforme patients. Clin Cancer Res 11(4):1462–1466 Sahin A, Sanchez C, Bullain S, Waterman P, Weissleder R, Carter BS, 2018, Development of third generation anti-EGFRvIII chimeric T cells and EGFRvIII-expressing artificial antigen presenting cells for adoptive cell therapy for glioma. PLoS One 13(7): e0199414 Miao H, Choi BD, Suryadevara CM, Sanchez-Perez L, Yang S, De Leon G, Sayour EJ, McLendon R, Herndon JE 2nd, Healy P, Archer GE, Bigner DD, Johnson LA, Sampson JH, 2014, EGFRvIII-specific chimeric antigen receptor T cells migrate to and kill tumor deposits infiltrating the brain parenchyma in an invasive xenograft model of glioblastoma. PLoS One 9(4):e94281 Thoma C, 2016, Prostate cancer: PD-L1 expression is common and indicates poor prognosis. Nat Rev Urol 13(1):5 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2135 EP 2141 DI 10.1007/s12253-019-00759-1 PG 7 ER PT J AU Szekerczes, T Gogl, A Illyes, I Mandl, J Borka, K Kiss, A Schaff, Zs Lendvai, G Werling, K AF Szekerczes, Timea Gogl, Aliz Illyes, Ildiko Mandl, Jozsef Borka, Katalin Kiss, Andras Schaff, Zsuzsa Lendvai, Gabor Werling, Klara TI Autophagy, Mitophagy and MicroRNA Expression in Chronic Hepatitis C and Autoimmune Hepatitis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Chronic hepatitis; autophagy; mitophagy; microRNA; immunohistochemistry ID Chronic hepatitis; autophagy; mitophagy; microRNA; immunohistochemistry AB Although the role of autophagy has been implicated in several forms of chronic hepatitis, it is still not fully understood. Active autophagy eliminates damaged molecules and organelles (such as mitochondria) by lysosomal degradation. In the present study, we aimed to examine and compare autophagy activity in chronic hepatitis C (CHC) and autoimmune hepatitis (AIH) by detecting the expression of autophagy (LC3 and p62) and mitochondrium-related (TOMM20) proteins, as well as the levels of selected microRNAs (miR-101, -155, -204 and − 224) known to be involved in the regulation of autophagy. In addition, the expression levels were related to pathohistological parameters. Liver biopsy samples, including 45 CHC and 18 AIH cases, were immunohistochemically stained for LC3, p62 and TOMM20 and the expression of miRNAs was determined using real-time PCR. We found elevated LC3 and p62 in AIH samples as compared with CHC ones, indicating an activated autophagy that is impaired in AIH as no degradation of p62 seemed to occur. Moreover, p62 showed strong correlation with necroinflammatory grades in the AIH group. The observed elevated levels of TOMM20 and p62 suggest a less efficient elimination of damaged mitochondria in AIH as opposed to CHC, in which autophagy seems to have a more active function. The level of miR-101 was increased in case of CHC as compared with AIH, however, miR-155, -204 and 224 resulted in no expressional. Furthermore, miR-224 level correlated with steatosis and miR-155 expression with fibrosis stage in CHC. In conclusion, dissimilar autophagic activity was observed in CHC and AIH, suggesting a close association between impaired autophagy and severity of necroinflammation. This impairment may not be regulated by the analyzed miRNAs. Nevertheless, miR-224 and − 155 seem to be associated with CHC progression. C1 [Szekerczes, Timea] Semmelweis University, 2nd Department of Pathology, Ulloi 93, 1091 Budapest, Hungary. [Gogl, Aliz] Semmelweis University, 2nd Department of Pathology, Ulloi 93, 1091 Budapest, Hungary. [Illyes, Ildiko] Semmelweis University, 2nd Department of Pathology, Ulloi 93, 1091 Budapest, Hungary. [Mandl, Jozsef] Semmelweis University, Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, 1094 Budapest, Hungary. [Borka, Katalin] Semmelweis University, 2nd Department of Pathology, Ulloi 93, 1091 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi 93, 1091 Budapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi 93, 1091 Budapest, Hungary. [Lendvai, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi 93, 1091 Budapest, Hungary. [Werling, Klara] Semmelweis University, 2nd Department of Internal Medicine, 1088 Budapest, Hungary. RP Lendvai, G (reprint author), Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. EM lendvai.gabor@med.semmelweis-univ.hu CR JefferiesM, Rauff B et al, 2018, Update on global epidemiology of viral hepatitis and preventive strategies.World J Clin Cases 6:589– 599. , DOI 10.12998/wjcc.v6.i13.589 Rios-Ocampo WA, Navas MC et al, 2019, The cellular stress response in hepatitis C virus infection: a balancing act to promote viral persistence and host cell survival. Virus Res 263:1–8. https:// doi.org/10.1016/j.virusres.2018.12.013 Vescovo T, Refolo G et al, 2014, Autophagy in HCV infection: keeping fat and inflammation at bay. Biomed Res Int 2014: 265353. , DOI 10.1155/2014/265353 Ke P-Y, 2018, The multifaceted roles of autophagy in flavivirushost interactions. Int J Mol Sci 19:3940. , DOI 10.3390/ ijms19123940 Ke PY, 2019, Diverse functions of autophagy in liver physiology and liver diseases. Int J Mol Sci 20:300. , DOI 10.3390/ ijms20020300 Jassey A, Liu C-H et al, 2019, Hepatitis C virus non-structural protein 5A, NS5A, disrupts mitochondrial dynamics and induces mitophagy. Cells 8:290. , DOI 10.3390/cells8040290 Rautou P-E,Mansouri A et al, 2010, Autophagy in liver diseases. J Hepatol 53:1123–1134. , DOI 10.1016/j.jhep.2010.07.006 Chan ST, Ou JHJ, 2017, Hepatitis C virus-induced autophagy and host innate immune response. Viruses 9:E224. , DOI 10. 3390/v9080224 Golbabapour S, Bagheri-Lankarani K et al, 2019, Autoimmune hepatitis and stellate cells; an insight into the role of autophagy. Curr Med Chem - In Press , DOI 10.2174/0929867326666190402120231 Czaja MJ, Ding Wet al, 2013, Functions of autophagy in normal and diseased liver. Autophagy 9:1131–1158. , DOI 10. 4161/auto.25063 Galluzzi L, Pietrocola F et al, 2015, Autophagy in malignant transformation and cancer progression. EMBO J 34:856–880. https:// doi.org/10.15252/embj.201490784 Eid N, Ito Yet al, 2016, Triggering of Parkin mitochondrial translocation in mitophagy: implications for liver diseases. Front Pharmacol 7:100. , DOI 10.3389/fphar.2016.00100 Wang L, Ou J et al, 2015, Hepatitis C virus and autophagy. Biol Chem 396:1215–1222. , DOI 10.1515/hsz-2015-0172 Ke P-Y, Chen SS-L, 2011, Activation of the unfolded protein response and autophagy after hepatitis C virus infection suppresses innate antiviral immunity in vitro. J Clin Invest 121:37–56. https:// doi.org/10.1172/JCI41474DS1 Dash S, Chava S et al, 2016, Hepatitis C virus infection induces autophagy as a prosurvival mechanism to alleviate hepatic ERstress response. Viruses 8:E150. , DOI 10.3390/v8050150 Christen U, Hintermann E, 2016, Immunopathogenic mechanisms of autoimmune hepatitis: how much do we know from animal models? Int J Mol Sci 17:E2007. , DOI 10.3390/ijms17122007 Sridhar S,BotbolYet al, 2012, Autophagy and disease: always two sides to a problem. J Pathol 226:255–273. , DOI 10.1038/jid.2014.371 Ott M, Norberg E et al, 2007, The mitochondrial TOM complex is required for tBid/Bax-induced cytochrome C release. J Biol Chem 282:27633–27639. , DOI 10.1074/jbc.M703155200 Sarnyai F, Szekerczes T et al, 2019, BGP-15 protects mitochondria in acute, acetaminophen overdose induced liver injury. Pathol Oncol Res - In Press Martinez-Outschoorn UE, Curry JM et al, 2013, Oncogenes and inflammation rewire host energy metabolism in the tumor microenvironment RAS and NFkB target stromal MCT4. Cell Cycle 12: 2580–2597. , DOI 10.4161/cc.25510 BabutaM, Furi I et al, 2019, Dysregulated autophagy and lysosome function are linked to exosome production by micro-RNA 155 in alcoholic liver disease. Hepatology - In Press. , DOI 10. 1002/hep.30766 Kim MK, Kim SG, 2014, Autophagy and microRNA dysregulation in liver diseases. Arch Pharm Res 37:1097–1116. https://doi. org/10.1007/s12272-014-0439-9 Frankel LB,Wen J et al, 2011)MicroRNA-101 is a potent inhibitor of autophagy. EMBO J 30:4628–4641. , DOI 10.1038/ emboj.2011.331 Li X, Liu J, 2019, LncRNA NEAT1 promotes autophagy via regulating miR-204/ATG3 and enhanced cell resistance to sorafenib in hepatocellular carcinoma. J Cell Physiol - In Press. , DOI 10.1002/jcp.29230 Lan S,Wu SYet al, 2014, Autophagy suppresses tumorigenesis of hepatitis B virus-associated hepatocellular carcinoma through degradation of microRNA-224. Hepatology 59:505–517. https://doi. org/10.1002/hep.26659 Ishak K, Baptista A et al, 1995, Histological grading and staging of chronic hepatitis. J Hepatoly 22:696–699. , DOI 10.1016/ 0168-8278(95)80226-6 Poynard T, Bedossa P et al, 1997, Natural history of liver fibrosis progression in patients with chronic hepatitis C. Lancet 349:825– 832. , DOI 10.1016/s0140-6736(96)07642-8 Kleiner DE,Makhlouf HR, 2016)Histology ofNAFLD and NASH in adults and children. Clin Liver Dis 20:293–312. , DOI 10.1016/j.cld.2015.10.011.Histology Doleshal M, Magotra AA et al, 2008, Evaluation and validation of total RNA extraction methods for microRNA expression analyses in formalin-fixed, paraffin-embedded tissues. JMol Diagnostics 10: 203–211. , DOI 10.2353/jmoldx.2008.070153 Andersen CL, Jensen JL et al, 2004, Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res 64:5245– 5250. , DOI 10.1158/0008-5472.CAN-04-0496 Klionsky DJ, Abdalla FC et al, 2012, Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 8: 445–544. , DOI 10.4161/auto.19496 Islam A, Sooro MA et al, 2018, Autophagic regulation of p62 is critical for cancer therapy. Int J Mol Sci 19:E1405. , DOI 10.3390/ijms19051405 Zhang N, Li XL et al, 2019, Impaired mitophagy triggers NLRP3 inflammasome activation during the progression from nonalcoholic fatty liver to nonalcoholic steatohepatitis. Lab Investig 99:749–763. , DOI 10.1038/s41374-018-0177-6 Panigrahi R, Chandra PK et al, 2015, Persistent hepatitis C virus infection impairs ribavirin antiviral activity through clathrinmediated trafficking of equilibrative nucleoside transporter 1. J Virol 89:626–642. , DOI 10.1128/jvi.02492-14 He G, Gunther C et al, 2017, PGAM5-mediated programmed necrosis of hepatocytes drives acute liver injury. Gut 66:716–723. , DOI 10.1136/gutjnl-2015-311247 Zhang X, Wu WKK et al, 2017, C-X-C motif chemokine 10 impairs autophagy and autolysosome formation in non-alcoholic steatohepatitis. Theranostics 7:2822–2836. , DOI 10. 7150/thno.19068 Ni H-M, Chao X et al, 2019, Dual roles of mammalian target of rapamycin in regulating liver injury and tumorigenesis in autophagy-defective mouse liver. Hepatology - In Press https:// doi.org/10.1002/hep.30770 Kim S, Syed GH et al, 2014, Hepatitis C virus triggers mitochondrial fission and attenuates apoptosis to promote viral persistence. Proc Natl Acad Sci USA 111:6413–6418. , DOI 10.1073/ pnas.1321114111 Vescovo T, Romagnoli A et al, 2012, Autophagy protects cells from HCV-induced defects in lipid metabolism. Gastroenterology 142:644–653. , DOI 10.1053/j.gastro.2011.11.033 Sasaki R, Sur S et al, 2019, Repression of microRNA-30e by hepatitis C virus enhances fatty acid synthesis. Hepatol Commun 3: 943–953. , DOI 10.1002/hep4.1362 Song HU, Du C et al, 2019, MicroRNA-101 inhibits autophagy to alleviate liver ischemia / reperfusion injury via regulating the mTOR signaling pathway. Int J Mol Med 43:1331–1342. https:// doi.org/10.3892/ijmm.2019.4077 Shakera O, Alhelfa M et al, 2017, miRNA-101-1 and miRNA-221 expressions and their polymorphisms as biomarkers for early diagnosis of hepatocellular carcinoma. Infect Genet Evol 51:173–181. , DOI 10.1016/j.meegid.2017.03.030 Lei Y, Wang Q, lan et al, 2019, MicroRNA-101 suppresses liver fibrosis by downregulating PI3K/Akt/mTOR signaling pathway. Clin Res Hepatol Gastroenterol 43:575–584. , DOI 10. 1016/j.clinre.2019.02.003 Lendvai G, Jarmay K et al, 2014, Elevated miR-33a and miR-224 in steatotic chronic hepatitis C liver biopsies.World J Gastroenterol 20:15343–15350. , DOI 10.3748/wjg.v20.i41.15343 Gelley F, Zadori G et al, 2014, MicroRNA profile before and after antiviral therapy in liver transplant recipients for hepatitis C virus cirrhosis. J Gastroenterol Hepatol 29:121–127. , DOI 10. 1111/jgh.12362 Gyugos M, Lendvai G et al, 2014, microRNA expression might predict prognosis of epithelial hepatoblastoma. Virhows Arch 464: 419–427. , DOI 10.1007/s00428-014-1549-y Smedile A, Bugianesi E, 2005, Steatosis and hepatocellular carcinoma risk. Eur Rev Med Pharmacol Sci 9:291–295 Bala S, Csak T et al, 2016, The pro-inflammatory effects of miR- 155 promote liver fibrosis and alcohol-induced steatohepatitis. J Hepatol 64:1378–1387. , DOI 10.1016/j.jhep.2016.01.035 Kaluzna EM, 2014, MicroRNA-155 and microRNA-196b: promising biomarkers in hepatitis C virus infection? Rev Med Virol 24: 169–185. , DOI 10.1002/rmv.1785 Blaya D, Aguilar-Bravo B et al, 2018, Expression of microRNA- 155 in inflammatory cells modulates liver injury. Hepatology 68: 691–706. , DOI 10.1002/hep.29833 Tiniakos DG, Brain JG et al, 2015, Role of histopathology in autoimmune hepatitis. Dig Dis 33:53–64. , DOI 10.1159/000440747 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2143 EP 2151 DI 10.1007/s12253-020-00799-y PG 9 ER PT J AU Lu, MY Wang, WCh Hou, TCh Kuo, ChY Lai, YCh AF Lu, Meng-Yao Wang, Wen-Chung Hou, Tai-Cheng Kuo, Chen-Yun Lai, Yen-Chein TI Methylation Statuses of H19DMR and KvDMR at WT2 in Wilms Tumors in Taiwan SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Multiplex ligation-dependent probe amplification; Nephroblastoma; Paternal uniparental disomy; Wilms tumor ID Multiplex ligation-dependent probe amplification; Nephroblastoma; Paternal uniparental disomy; Wilms tumor AB Wilms tumor is the most common pediatric renal malignancy. Several genetic loci have been shown to be associated with its formation. Genetic or epigenetic aberrations at WT1 and WT2 loci have been implicated in the etiology of the majority of sporadic Wilms tumors. In our previous study, most Wilms tumors tested negative for both constitutional mutations and somatic mutations in the WT1 gene. Thus, WT2 may play an important role in these tumors. In the present study, we analyzed the methylation statuses of WT2 at 11p15 using methylation sensitive multiplex ligation-dependent probe amplification in six Wilms tumors. Paternal uniparental disomy at WT2 was observed in two Wilms tumors with epithelial components due to hypermethylation at H19DMR and hypomethylation at KvDMR. Our findings highlight the benefits of testing for 11p15 epigenetic abnormalities to identify Wilms tumors with epithelial components. C1 [Lu, Meng-Yao] National Taiwan University Hospital, Department of PediatricsTaipei, Taiwan, Republic of China. [Wang, Wen-Chung] Jen-Ai Hospital, Department of Obstetrics and GynecologyTaichung, Taiwan, Republic of China. [Hou, Tai-Cheng] Jen-Ai Hospital, Department of PathologyTaichung, Taiwan, Republic of China. [Kuo, Chen-Yun] Jen-Ai Hospital, Department of PathologyTaichung, Taiwan, Republic of China. [Lai, Yen-Chein] Chung Shan Medical University, Department of Medical Laboratory and Biotechnology, No.110, Sec. 1, Chien Kuo N. Road, 402 Taichung, Taiwan, Republic of China. RP Lai, YCh (reprint author), Chung Shan Medical University, Department of Medical Laboratory and Biotechnology, 402 Taichung, Taiwan, Republic of China. EM yenchein@csmu.edu.tw CR Brown KW, Malik KT, 2001, The molecular biology of Wilms tumour. Expert Rev Mol Med 2001:1–16 Pastore G, Znaor A, Spreafico F, Graf N, Pritchard-Jones K, Steliarova-Foucher E, 2006, Malignant renal tumours incidence and survival in European children, 1978-1997): report from the automated childhood Cancer information system project. Eur J Cancer 42:2103–2114 Steliarova-Foucher E, Colombet M, Ries LAG, Moreno F, Dolya A, Bray F et al, 2017, International incidence of childhood cancer, 2001-10: a population-based registry study. Lancet Oncol 18:719– 731 Treger TD, Chowdhury T, Pritchard-Jones K, Behjati S et al, 2019, The genetic changes of Wilms tumour. Nat Rev Nephrol 15:240– 251 PDQ Pediatric Treatment Editorial Board, 2002)Wilms Tumor and Other Childhood Kidney Tumors Treatment, PDQ®): Health Professional Version. PDQ Cancer Information Summaries. Bethesda, MD): National Cancer Institute, US, 2002–2020 Gadd S, Huff V, Walz AL, Ooms A, Armstrong AE, Gerhard DS et al, 2017, A Children's oncology group and TARGET initiative exploring the genetic landscape of Wilms tumor. Nat Genet 49: 1487–1494 Mahamdallie S, Yost S, Poyastro-Pearson E, Holt E, Zachariou A, Seal S et al, 2019, Identification of new Wilms tumour predisposition genes: an exome sequencing study. Lancet Child Adolesc Health 3:322–331 Call KM, Glaser T, Ito CY, Buckler AJ, Pelletier J, Haber DA et al, 1990, Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms' tumor locus. Cell 60: 509–520 Karnik P, Chen P, Paris M, Yeger H, Williams BR, 1998, Loss of heterozygosity at chromosome 11p15 in Wilms tumors: identification of two independent regions. Oncogene 17:237–240 Al-Hussain T, Ali A, Akhtar M, 2014, Wilms tumor: an update. Adv Anat Pathol 21:166–173 Cardoso LC, Tenorio Castano JA, Pereira HS, Lima MA, Dos Santos AC, de Faria PS et al, 2012, Constitutional and somatic methylation status of DMRH19 and KvDMR in Wilms tumor patients. GenetMol Biol 35:714–724 Dekel B, Metsuyanim S, Schmidt-Ott KM, Fridman E, Jacob- Hirsch J, Simon A et al, 2006, Multiple imprinted and stemness genes provide a link between normal and tumor progenitor cells of the developing human kidney. Cancer Res 66:6040–6049 Scott RH, Douglas J, Baskcomb L, Huxter N, Barker K, Hanks S et al, 2008, Constitutional 11p15 abnormalities, including heritable imprinting center mutations, cause nonsyndromic Wilms tumor. Nat Genet 40:1329–1334 Lu MY, Wang WC, Lin CW, Chang A, Lai YC, 2014, Identification of a constitutional mutation in the WT1 gene in Taiwanese patients with Wilms tumor. Adv Biosci Biotechnol 5: 230–234 Chiang MR, Kuo CW, Wang WC, Hou TC, Kuo CY, Lu MYet al, 2019, Correlations between histological and array comparative genomic hybridization characterizations of Wilms tumor. Pathol Oncol Res 25:1199–1206 WangWC, Lai YC, 2016, Genetic analysis results of mature cystic teratomas of the ovary in Taiwan disagree with the previous origin theory of this tumor. Hum Pathol 52:128–135 Dao D,Walsh CP,Yuan L,GorelovD, Feng L,Hensle Tet al, 1999, Multipoint analysis of human chromosome 11p15/mouse distal chromosome 7: inclusion of H19/IGF2 in the minimalWT2 region, gene specificity of H19 silencing in Wilms' tumorigenesis and methylation hyper-dependence of H19 imprinting. Hum Mol Genet 8:1337–1352 Mancini-DiNardo D, Steele SJ, IngramRS, Tilghman SM, 2003, A differentially methylated region within the gene Kcnq1 functions as an imprinted promoter and silencer. Hum Mol Genet 12:283–294 Hubertus J, Lacher M, Rottenkolber M, Muller-Hocker J, Berger M, Stehr M et al, 2011, Altered expression of imprinted genes in Wilms tumors. Oncol Rep 25:817–823 Scott RH, Murray A, Baskcomb L, Turnbull C, Loveday C, Al- Saadi R et al, 2012, Stratification of Wilms tumor by genetic and epigenetic analysis. Oncotarget 3:327–335 VerschuurAC,VujanicGM,Van TinterenH, JonesKP, de Kraker J, Sandstedt B, 2010, Stromal and epithelial predominant Wilms tumours have an excellent outcome: the SIOP 93 01 experience. Pediatr Blood Cancer 55:233–238 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2153 EP 2159 DI 10.1007/s12253-020-00802-6 PG 7 ER PT J AU Yu, D Ma, Y Feng, Ch Ma, Z Guo, J Chen, H He, T Guo, J Sun, X Qin, Q Sun, X Ma, J AF Yu, Dingyue Ma, Yuanyuan Feng, Chen Ma, Zhiyu Guo, Jiayou Chen, Hui He, Tianli Guo, Jiayi Sun, Xingbang Qin, Qin Sun, Xinchen Ma, Jianxin TI PBX1 Increases the Radiosensitivity of Oesophageal Squamous Cancer by Targeting of STAT3 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oesophageal squamous cancer; PBX1; Radiosensitivity; STAT3 ID Oesophageal squamous cancer; PBX1; Radiosensitivity; STAT3 AB The radioresistance of oesophageal squamous cell carcinoma (OSCC) is a critical factor leading to a poor prognosis among patients. The expression of PBX1 is abnormally high in a broad range of human tissues, and this gene plays a key role in tumour proliferation. This research intended to explore the radiosensitization of OSCC by silencing PBX1. The OSCC cell lines KYSE450 and KYSE150 were subjected to PBX1 silencing and/or irradiation (IR). Cell proliferation, colony formation, and apoptosis were tested to evaluate the radiosensitization ability of PBX1 silencing. The levels of STAT3 and p-STAT3 in the OSCC cells were tested by Western blotting. Furthermore, KYSE150 cells with or without PBX1 silencing were xenografted into nude mice with or without radiation exposure. Concomitant PBX1 silencing and IR can obviously suppress growth and enhance radiosensitivity in OSCC cells and xenografts. Moreover, the downregulation of PBX1 inhibits the expression of STAT3 and p- STAT3. The downregulation of PBX1may increase radiosensitivity in OSCC cells and xenografts via the PBX1/STAT3 pathway. Our findings demonstrate that PBX1 may be a potential target for promoting the effect of radiation therapy in OSCC patients. C1 [Yu, Dingyue] The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, Department of radiotherapy, 57 Zhonghua West Road, 222042 Lian Yungang, Jiangsu Province, China. [Ma, Yuanyuan] The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, Department of radiotherapy, 57 Zhonghua West Road, 222042 Lian Yungang, Jiangsu Province, China. [Feng, Chen] First Hospital of Bengbu Medical College, Oncology Department, 233000 Bengbu, AnHui, China. [Ma, Zhiyu] The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, Department of radiotherapy, 57 Zhonghua West Road, 222042 Lian Yungang, Jiangsu Province, China. [Guo, Jiayou] The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, Department of radiotherapy, 57 Zhonghua West Road, 222042 Lian Yungang, Jiangsu Province, China. [Chen, Hui] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, Jiangsu, China. [He, Tianli] Changxing People’s Hospital, Department of Respiratory, 313000 Huzhou, Zhejiang, China. [Guo, Jiayi] The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, Department of radiotherapy, 57 Zhonghua West Road, 222042 Lian Yungang, Jiangsu Province, China. [Sun, Xingbang] The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, Department of radiotherapy, 57 Zhonghua West Road, 222042 Lian Yungang, Jiangsu Province, China. [Qin, Qin] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, Jiangsu, China. [Sun, Xinchen] The First Affiliated Hospital of Nanjing Medical University, Department of Radiotherapy, 210000 Nanjing, Jiangsu, China. [Ma, Jianxin] The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, Department of radiotherapy, 57 Zhonghua West Road, 222042 Lian Yungang, Jiangsu Province, China. RP Ma, J (reprint author), The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, Department of radiotherapy, 222042 Lian Yungang, China. EM mjx3416@163.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, RebeloM et al, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in globocan 2012. Int J Cancer 136(5):E359–E386 Kamangar F, Dores GM, Anderson WF, 2006, Patterns of cancer incidence,mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol 24(14):2137–2150 Tirumani H, Rosenthal MH, Tirumani SH, Shinagare AB, Krajewski KM, Ramaiya NH, 2015, Esophageal carcinoma: current concepts in the role of imaging in staging and management. Can Assoc Radiol J 66(2):130–139 Ohashi S, Miyamoto S’i, Kikuchi O, Goto T, Amanuma Y, MutoM, 2015, Recent advances from basic and clinical studies of esophageal squamous cell carcinoma. Gastroenterology 149(7):1700– 1715 Ding Y-Q, Qin Q, Yang Y, Xin-Chen S, Yang X, Zhu H-C et al, 2016, Improved sensitization effect of sunitinib in cancer cells of the esophagus under hypoxic microenviroment. Oncol Lett 12(6): 4671–4676 Song YP, Ma JB, Hu LK, Zhou W, Chen EC, Zhang W, 2011, Phase i/ii study of hypofractioned radiation with threedimensional conformal radiotherapy for clinical t3-4n0-1m0 stage esophageal carcinoma. Technol Cancer Res Treat 10(1):25 Nourse J, Mellentin JD, Galili N,Wilkinson J, Stanbridge E, Smith SD et al, 1990, Chromosomal translocation t(1;19, results in synthesis of a homeobox fusion mrna that codes for a potential chimeric transcription factor. Cell 60(4):535–545 KampsMP, Look AT, Baltimore D, 1991, The human t(1;19, translocation in pre-b all produces multiple nuclear e2a-pbx1 fusion proteins with differing transforming potentials. Genes Dev 5(3): 358–368 Hunger SP, Galili N, Carroll AJ, Crist WM, Link MP, Cleary ML, 1991, The t(1;19)(q23;p13, results in consistent fusion of e2a and pbx1 coding sequences in acute lymphoblastic leukemias. Blood 77(4):687–693 Izraeli S, Kovar H, Gadner H, Lion T, 1992, Unexpected heterogeneity in e2a/pbx1 fusion messenger rna detected by the polymerase chain reaction in pediatric patients with acute lymphoblastic leukemia. Blood 80(6):1413 Numata S, Kato K, Horibe K, 1993, New e2a/pbx1 fusion transcript in a patient with t(1;19)(q23;p13, acute lymphoblastic leukemia. Leukemia 7(9):1441 Fernandez LC, ErricoMC, Bottero L, PenkovD, ResnatiM, Blasi F et al, 2008, Oncogenic hoxb7 requires tale cofactors and is inactivated by a dominant-negative pbx1 mutant in a cell-specific manner. Cancer Lett 266(2):0–155 Ficara F, Murphy MJ, Lin M, Cleary ML, 2008, Pbx1 regulates self-renewal of long-term hematopoietic stem cells by maintaining their quiescence. Cell Stem Cell 2(5):484–496 Schnabel CA, Godin RE, Cleary ML, 2003, Pbx1 regulates nephrogenesis and ureteric branching in the developing kidney. Dev Biol 254(2):262–276 Schnabel CA, Selleri L, Cleary ML, 2010, Pbx1 is essential for adrenal development and urogenital differentiation. Genesis 37(3): 123–130 Park JT, Shih IM,Wang TL, 2008, Identification of pbx1, a potential oncogene, as a notch3 target gene in ovarian cancer. Cancer Res 68(21):8852–8860 Wang J, Shidfar A, Ivancic D, Ranjan M, Liu L, Choi MR et al, 2017, Overexpression of lipid metabolism genes and pbx1 in the contralateral breasts of women with estrogen receptor-negative breast cancer. Int J Cancer 140(11):2484 Magnani L, Ballantyne EB, Zhang X, Lupien M, 2011, Pbx1 genomic pioneer function drives erα signaling underlying progression in breast cancer. PLoS Genet 7(11):e1002368 Zhang LX, Xu L, Zhang CH, Lu YH, Ji TH, Ling LJ, 2017, Uc.38 induces breast cancer cell apoptosis via pbx1. Am J Cancer Res 7(12):2438–2451 Mo ML, Chen Z, Zhou HM, Li H, Hirata T, Jablons DM et al, 2013, Detection of e2a-pbx1 fusion transcripts in human nonsmall- cell lung cancer. J Exp Clin Cancer Res 32(1):29 Li W, Huang K, Guo H, Cui G, Zhao S, 2014, Inhibition of nonsmall- cell lung cancer cell proliferation by pbx1. Chin J Cancer Res 26(5):573–578 Jung JG, Shih IM, Park JT, Gerry E, Kim TH & Ayhan A, et al, 2016, Ovarian cancer chemoresistance relies on the the stem cell reprogramming factor pbx1. Cancer Res, 76(21) Li u DB, Gu ZD, Cao XZ, Liu H, Li JY, 2005, Immunocytochemical detection of hoxd9 and pbx1 homeodomain protein expression in chinese esophageal squamous cell carcinomas. World J Gastroenterol 11(10):1562–1566 Stepkowski SM, Chen W, Ross JA, Nagy ZS, Kirken RA, 2008, Stat3: an important regulator of multiple cytokine functions. Transplantation 85(10):1372–1377 Fukuda A, Wang S, Morris, John et al, 2011, Stat3 and mmp7 contribute to pancreatic ductal adenocarcinoma initiation and progression. Cancer Cell 19(4):456–469 Zhang Z, Duan Q, Zhao H, Liu T, Wu H, Shen Q et al, 2016, Gemcitabine treatment promotes pancreatic cancer stemness through the nox/ros/nf-κb/stat3 signaling cascade. Cancer Lett 382(1):53–63 Wang M, Meng B, Liu Y, Yu J, Chen Q & Liu Y, 2017, Mir-124 inhibits growth and enhances radiation-induced apoptosis in nonsmall cell lung cancer by inhibiting stat3. Cell Physiol Biochem, 2017-2028 Zhang Q, Zhang C, He J, Guo Q, Hu D, Yang X et al, 2015, Stat3 inhibitor stattic enhances radiosensitivity in esophageal squamous cell carcinoma. Tumor Biol 36(3):2135–2142 Wei X, Yu L & Li Y, 2018, Pbx1 promotes the cell proliferation via jak2/stat3 signaling in clear cell renal carcinoma. Biochem Biophys Res Commun Zhang C, Yang X, Zhang Q, Guo Q, He J, Qin Q et al, 2014, Stat3 inhibitor nsc74859 radiosensitizes esophageal cancer via the downregulation of hif-1α. Tumour Biol 35(10):9793–9799 Monica K, Galili N, Nourse J, Saltman D, Cleary ML, 1991, Pbx2 and pbx3, new homeobox genes with extensive homology to the human proto-oncogene pbx1. Mol Cell Biol 11(12):6149–6157 Qiu Y, Song B, Zhao G, Deng B, Makino T, Tomita Yet al, 2010, Expression level of pre b cell leukemia homeobox 2 correlates with poor prognosis of gastric adenocarcinoma and esophageal squamous cell carcinoma. Int J Oncol 36(3):651–663 Wang T, Niu G, KortylewskiM, Burdelya L, Shain K, Zhang S et al, 2004, Regulation of the innate and adaptive immune responses by stat-3 signaling in tumor cells. Nat Med 10(1):48 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2161 EP 2168 DI 10.1007/s12253-020-00803-5 PG 8 ER PT J AU Tang, X He, Q Sun, G Qu, H Liu, J Gao, L Shi, J Ye, J Liang, Y AF Tang, Xiaolong He, Qingsi Sun, Guorui Qu, Hui Liu, Jia Gao, Lei Shi, Jingbo Ye, Jianhong Liang, Yahang TI Total Tumor Volume Should be Considered as an Important Prognostic Factor for Synchronous Multiple Gastric Cancer Patients with Curative Gastrectomy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Tumor volume; Gastrectomy; Prognosis ID Gastric cancer; Tumor volume; Gastrectomy; Prognosis AB Synchronous multiple gastric cancer (SMGC) was a special type of gastric cancer with relatively low incidence. This article was designed to demonstrate that the total tumor volume (TTV) should be treated as an important prognostic factor in SMGC patients with curative gastrectomy. This study retrospective analyzed 140 SMGC patients who received curative gastrectomy between December 2004 and December 2014 in our hospital. Clinicopathological features, preoperative evaluation, surgical treatment, and outcome parameters were reviewed and analyzed. This study applied univariate and multivariate analyses to identify the most significant prognostic factors. In the univariate analysis, the TTV, pTTVNM, pN stage, pT of main tumor were all significant prognostic factors in SMGC patients (all P < 0.05). In the multivariate analysis, pN stage, TTV and pTTVNM were confirmed to be independent prognostic factors (all P < 0.05). In the comparison of survival analysis, the pTTVNM stage system (P < 0.05) was superior to the pTNM stage system (P > 0.05) in SMGC patients. In conclusion, the TTV should be considered as an independent prognostic factor in overall survival in SMGC patients who received curative gastrectomy. The pTTVNM stage should be recommended as a suitable staging system for SMGC patients. C1 [Tang, Xiaolong] Qilu Hospital of Shandong University, Department of General Surgery, No.107, West of Wenhua Street, Lixia District, 250012 Jinan, China. [He, Qingsi] Qilu Hospital of Shandong University, Department of General Surgery, No.107, West of Wenhua Street, Lixia District, 250012 Jinan, China. [Sun, Guorui] Qilu Hospital of Shandong University, Department of General Surgery, No.107, West of Wenhua Street, Lixia District, 250012 Jinan, China. [Qu, Hui] Qilu Hospital of Shandong University, Department of General Surgery, No.107, West of Wenhua Street, Lixia District, 250012 Jinan, China. [Liu, Jia] Qilu Hospital of Shandong University, Department of Health Management Center, 250012 Jinan, China. [Gao, Lei] Qilu Hospital of Shandong University, 250011 Jinan, China. [Shi, Jingbo] Qilu Hospital of Shandong University, 250011 Jinan, China. [Ye, Jianhong] Qilu Hospital of Shandong University, 250011 Jinan, China. [Liang, Yahang] Qilu Hospital of Shandong University, 250011 Jinan, China. RP Sun, G (reprint author), Qilu Hospital of Shandong University, Department of General Surgery, 250012 Jinan, China. EM doctorsungr@163.com CR Isobe T, Hashimoto K, Kizaki J, Murakami N, Aoyagi K, Koufuji K, Akagi Y, Shirouzu K, 2013, Characteristics and prognosis of synchronous multiple early gastric cancer. World J Gastroenterol 19(41):7154–7159 Nozaki I, Hato S, Kobatake T, Ohta K, Kubo Y, Nishimura R, Kurita A, 2014, Incidence of metachronous gastric cancer in the remnant stomach after synchronous multiple cancer surgery. Gastric Cancer 17(1):61–66 Borie F, Plaisant N, Millat B, Hay JM, Fagniez PL, De Saxce B, French Associations for Surgical Research, 2003, Treatment and prognosis of early multiple gastric cancer. Eur J Surg Oncol 29(6): 511–514 Deng J, Liang H, Sun D, Wang D, Pan Y, 2010, Suitability of 7th UICC N Stage for Predicting the Overall Survival of Gastric Cancer Patients After Curative Resection in China. Ann Surg Oncol 17(5): 1259–1266 Pak K, Cheon GJ, Nam HY, Kim SJ, Kang KW, Chung JK, Kim EE, Lee DS, 2014, Prognostic value of metabolic tumor volume and total lesion glycolysis in head and neck cancer: a systematic review and meta-analysis. J Nucl Med 55(6):884–890 EomBW, Lee JH, Choi IJ, KookMC, NamBH, RyuKW, KimYW, 2012, Pretreatment risk factors for multiple gastric cancer and missed lesions. J Surg Oncol 105(8):813–817 Fujita T, Gotohda N, Takahashi S, Nakagohri T, Konishi M, Kinoshita T, 2009, Clinical and histopathological features of remnant gastric cancers, after gastrectomy for synchronous multiple gastric cancers. J Surg Oncol 100(6):466–471 Zu H, Wang F, Ma Y, Xue Y, 2013, Stage-stratified analysis of prognostic significance of tumor size in patients with gastric cancer. PLoS One 8(1):e54502 Zhang J, Zhou Y, Jiang K, Shen Z, Ye Y,Wang S, 2014, Evaluation of the seventh AJCC TNM staging system for gastric cancer: a meta-analysis of cohort studies. Tumour Biol 35(9):8525–8532 Zhao LY, Zhang WH, Chen XZ, Yang K, Chen XL, Liu K, Zhang B, Chen ZX, Chen JP, Zhou ZG, Hu JK, 2015, Prognostic Significance of Tumor Size in 2405 Patients With Gastric Cancer: A Retrospective Cohort Study. Med, Baltim, 94(50):e2288 Li H, Chen XL, Li JR, Li ZL, Chen TW, Pu H, Yin LL, Xu GH, Li ZW, Reng J, Zhou P, Cheng ZZ, Cao Y, 2016, Tumor volume of resectable gastric adenocarcinoma on multidetector computed tomography: association with N categories. Clinics 71(4):199–204 Yoon H, Lee DH, 2014, New approaches to gastric cancer staging: beyond endoscopic ultrasound, computed tomography and positron emission tomography. World J Gastroenterol 20(38):13783–13790 Neri E,Guidi E, Pancrazi F, Castagna M, Castelluccio E, Balestri R, Buccianti P,Masi L, Falcone A,Manfredi B, Faggioni L, Bartolozzi C, 2015, MRI tumor volume reduction rate vs tumor regression grade in the pre-operative re-staging of locally advanced rectal cancer after chemo- radiotherapy. Eur J Radiol 84(12):2438–2443 LiMX, Zhao H, Bi XY, Li ZY, Huang Z, Han Y, Zhou JG, Zhao JJ, Zhang YF, WeiWQ, Zhao DB, Cai JQ, 2016, Total tumor volume predicts survival following liver resection in patients with hepatocellular carcinoma. Tumour Biol 37(7):9301–9310 Voss B,Wilop S, Jonas S, El-Komy MH, Schaller J, von Felbert V, Megahed M, 2014, Tumor volume as a prognostic factor in resectable malignant melanoma. Dermatology 228(1):66–70 Yu Y, Guan H, Xing LG, Xiang YB, 2015, Role of gross tumor volume in the prognosis of non-small cell lung cancer treated with 3D conformal radiotherapy: a meta-analysis. Clin Ther 37(10): 2256–2266 Choi SM, Choi DK, Kim TH, Jeong BC, Seo SI, Jeon SS, Lee HM, Choi HY, Jeon HG, 2015, A comparison of radiologic tumor volume and pathologic tumor volume in renal cell carcinoma, RCC). PLoS One 10(3):e0122019 Jiang N, Deng JY, Ding XW, Liu Y, Liang H, 2015, Tumor volume as a prognostic factor was superior to the seventh edition of the pT classification in resectable gastric cancer. Eur J Surg Oncol 41(3): 315–322 Lee JA, Yang D, Yoon WS, Park YJ, Kim CY, Moon HY, Lee SI, 2014, Tumor volume reduction assessed by planning computed tomography in patients with rectal cancer during preoperative chemoradiation: impact of residual tumor volume on the prediction of pathologic tumor regression. Tumori 100(2):158–162 Lu J,HuangCM, Zheng CH, Li P, Xie JW,Wang JB, Lin JX, 2013, Consideration of tumor size improves the accuracy of TNMpredictions in patients with gastric cancer after curative gastrectomy. Surg Oncol 22(3):167–171 Dicken BJ, Saunders LD, Jhangri GS, de Gara C, Cass C, Andrews S, Hamilton SM, 2004, Gastric cancer: establishing predictors of biologic behavior with use of population-based data. Ann Surg Oncol 11(6):629–635 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2169 EP 2175 DI 10.1007/s12253-020-00804-4 PG 7 ER PT J AU Xu, Y Zhang, L Wang, Q Zheng, M AF Xu, Yuting Zhang, Lin Wang, Qingling Zheng, Maojin TI Comparison of Different Colorectal Cancer With Liver Metastases Models Using Six Colorectal Cancer Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer cell lines; Liver metastases; Orthotopic model; Ectopic model ID Colorectal cancer cell lines; Liver metastases; Orthotopic model; Ectopic model AB At present, modeling methods of colorectal cancer with liver metastases have significant limitations. Here, we established orthotopic and ectopic hepatic metastases models using six colorectal cancer cell lines to choose an ideal animal model for studying colorectal cancer growth and liver metastases. Luciferin-expressing six colorectal cancer cell lines were used to induce animal models of colorectal cancer with liver metastases by intra-splenic injection or implantation of tumor tissue in the caecum. Tumors growth and metastatic events were observed by bioluminescence imaging. In orthotopic transplantation group, six cell lines all had taken rates of 100%for orthotopic tumors but showed variations in rates of growth. HCT-116 cell developed the 50% liver metastases. However, the ectopic transplantation group achieved higher liver metastatic rate, with the highest frequencies for HCT116 cell (90%) and SW620 cell (77.8%). Furthermore, the time to develop liver metastases and survival rates of bearing tumor mice were shorter than orthotopic transplantation group. Additionally, six colorectal cancer cell lines resulted in more lymph node metastases in orthotopic transplantation group, whereas produced widespread peritoneal seeding in ectopic transplantation group. Bioluminescence imaging and pathological findings confirmed the growth and metastatic characteristics of tumors. Two animal models of colorectal cancer using six cell lines showed highly variations in rates of growth, survival rates of bearing-tumor mice and frequencies of metastases. The study provides useful information for the establishment of clinically relevant colorectal cancer with liver metastases animal models. C1 [Xu, Yuting] Xuzhou Medical University, Department of Pathology, 221004 Xuzhou, Jiangsu, China. [Zhang, Lin] Xuzhou Medical University, Department of Pathology, 221004 Xuzhou, Jiangsu, China. [Wang, Qingling] Xuzhou Medical University, Department of Pathology, 221004 Xuzhou, Jiangsu, China. [Zheng, Maojin] Xuzhou Medical University, Department of Pathology, 221004 Xuzhou, Jiangsu, China. RP Xu, Y (reprint author), Xuzhou Medical University, Department of Pathology, 221004 Xuzhou, China. EM xyt325@xzhmu.edu.cn CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, 2015, Global cancer statistics, 2012. CA Cancer J Clin 65(2):87– 108. , DOI 10.3322/caac.21262 Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F, 2017, Global patterns and trends in colorectal cancer incidence and mortality. Gut 66(4):683–691. , DOI 10.1136/ gutjnl-2015-310912 Siegel R, Desantis C, Jemal A, 2014, Colorectal cancer statistics, 2014. CA Cancer J Clin 64(2):104–117. , DOI 10.3322/ caac.21220 O’Connell JB, MaggardMA, Ko CY, 2004, Colon cancer survival rates with the new American Joint Committee on Cancer sixth edition staging. J Natl Cancer Inst 96(19):1420–1425. https://doi. org/10.1093/jnci/djh275 Cespedes MV, Espina C, Garcia-Cabezas MA, Trias M, Boluda A, Gomez del Pulgar MT, Sancho FJ, Nistal M, Lacal JC, Mangues R, 2007, Orthotopic microinjection of human colon cancer cells in nude mice induces tumor foci in all clinically relevant metastatic sites. Am J Pathol 170(3):1077–1085. , DOI 10.2353/ ajpath.2007.060773 Kazuhiro ISHIZU, Naohide SUNOSE, Kanami YAMAZAKI, Takashi TSURUO, Sotaro SADAHIRO, Hiroyasu MAKUUCHI, Takao YAMORI, 2007, Development and Characterization of a Model of Liver Metastasis Using Human Colon Cancer HCT-116 Cells. Biol Pharm Bull 30:1779–1783. , DOI 10.1248/bpb. 30.1779 Cho YB, Hong HK, Choi YL, Oh E, Joo KM, Jin J, Nam DH, Ko YH, LeeWY, 2014, Colorectal cancer patient-derived xenografted tumors maintain characteristic features of the original tumors. J Surg Res 187(2):502–509. , DOI 10.1016/j.jss.2013.11. 010 Sasaki H, Miura K, Horii A, Kaneko N, Fujibuchi W, Kiseleva L, Gu Z, Murata Y, Karasawa H, Mizoi T, Kobayashi T, Kinouchi M, Ohnuma S, Yazaki N, UnnoM, Sasaki I, 2008, Orthotopic implantation mouse model and cDNA microarray analysis indicates several genes potentially involved in lymph node metastasis of colorectal cancer. Cancer Sci 99(4):711–719. , DOI 10.1111/j. 1349-7006.2008.00725.x Bedard PL, Hansen AR, Ratain MJ, Siu LL, 2013, Tumour heterogeneity in the clinic. Nature 501(7467):355–364. , DOI 10. 1038/nature12627 Lee WY, Hong HK, Ham SK, Kim CI, CHO YB, 2014, Comparison of Colorectal Cancer in differentially established liver metastasis models. Anticancer research 34(7):3321–3328 Roque-Lima B, Roque C, BegnamiMD, Peresi P, Lima ENP,Mello CAL, Coimbra FJ, Chojniak R, Goss Santos T, 2018)Development of patient-derived orthotopic xenografts from metastatic colorectal cancer in nude mice. J Drug Target:1–7. , DOI 10.1080/ 1061186X.2018.1509983 Nie S, Zhou J, Bai F, Jiang B, Chen J, Zhou J, 2014, Role of endothelin A receptor in colon cancer metastasis: in vitro and in vivo evidence. Mol Carcinog 53 Suppl 1:E85–E91. https://doi. org/10.1002/mc.22036 Yang J, Shikata N, Mizuoka H, Tsuburaa A, 1996, Colon carcinogenesis in shrews by intrarectal infusion of N-methyl-Nnitrosourea. Cancer letters 110:105–112. , DOI 10.1016/ s0304-3835(96)04468-0 O’Rourke KP, Loizou E, Livshits G, Schatoff EM, Baslan T, Manchado E, Simon J, Romesser PB, Leach B, Han T, Pauli C, Beltran H, Rubin MA, Dow LE, Lowe SW, 2017, Transplantation of engineered organoids enables rapid generation of metastatic mouse models of colorectal cancer. Nat Biotechnol 35(6):577– 582. , DOI 10.1038/nbt.3837 Sloan EK, Priceman SJ, Cox BF, Yu S, Pimentel MA, Tangkanangnukul V, Arevalo JM, Morizono K, Karanikolas BD, Wu L, SoodAK, Cole SW(2010, The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res 70(18):7042–7052. , DOI 10.1158/0008-5472.CAN-10- 0522 Thalheimer A, Korb D, Bonicke L, Wiegering A, Muhling B, Schneider M, Koch S, Riedel SS, Germer CT, Beilhack A, Brandlein S, Otto C, 2013, Noninvasive visualization of tumor growth in a human colorectal liver metastases xenograft model using bioluminescence in vivo imaging. J Surg Res 185(1):143– 151. , DOI 10.1016/j.jss.2013.03.024 Terracina KP, Aoyagi T, Huang WC, Nagahashi M, Yamada A, Aoki K, Takabe K, 2015, Development of a metastatic murine colon cancer model. J Surg Res 199(1):106–114. , DOI 10.1016/j.jss.2015.04.030 Trainer DL, Kline T, McCabe FL, Faucette LF, Feild J, Chaikin M, Anzano M, Rieman D, Hoffstein S, Li DJ, 1988, Biological characterization and oncogene expression in human colorectal carcinoma cell lines. Int J Cancer 41:287–296. , DOI 10.1002/ijc. 2910410221 ZengQ, Lei F, Chang Y, Gao Z,WangY, Gao Q, Niu P, Li Q, 2019, An oncogenic gene, SNRPA1, regulates PIK3R1, VEGFC, MKI67, CDK1 and other genes in colorectal cancer. Biomed Pharmacother. 117: , DOI 10.1016/j.biopha.2019.109076 Mittal VK, Bhullar JS, Jayant K, 2015, Animal models of human colorectal cancer: Current status, uses and limitations. World J Gastroenterol 21(41):11854–11861. , DOI 10.3748/wjg. v21.i41.11854 Zigmond E, Halpern Z, Elinav E, Brazowski E, Jung S, Varol C, 2011, Utilization of murine colonoscopy for orthotopic implantation of colorectal cancer. PLoS One 6(12):e28858. , DOI 10.1371/journal.pone.0028858 Golovko D, Kedrin D, Yilmaz OH, Roper J, 2015, Colorectal cancer models for novel drug discovery. Expert Opin Drug Discov 10(11):1217–1229. , DOI 10.1517/17460441.2015. 1079618 Hackl C, Man S, Francia G, Milsom C, Xu P, Kerbel RS, 2013, Metronomic oral topotecan prolongs survival and reduces liver metastasis in improved preclinical orthotopic and adjuvant therapy colon cancer models. Gut 62(2):259–271. , DOI 10.1136/ gutjnl-2011-301585 Hite N, Klinger A, Hellmers L,Maresh GA,Miller PE, Zhang X, Li L, Margolin DA, 2018, An Optimal Orthotopic Mouse Model for Human Colorectal Cancer Primary Tumor Growth and Spontaneous Metastasis. Dis Colon Rectum 61(6):698–705. , DOI 10.1097/DCR.0000000000001096 Valastyan S, Weinberg RA, 2011, Tumor metastasis: Molecular insights and evolving paradigms. Cell 147:275–292. https://doi. org/10.1016/j.cell.2011.09.024 Flatmark K, Maelandsmo GM, Martinsen M, Rasmussen H, Fodstad O, 2004, Twelve colorectal cancer cell lines exhibit highly variable growth and metastatic capacities in an orthotopic model in nude mice. Eur J Cancer 40(10):1593–1598. , DOI 10. 1016/j.ejca.2004.02.023 Rajput A, Dominguez San Martin I, Rose R, Beko A, Levea C, Sharratt E, Mazurchuk R, Hoffman RM, Brattain MG, Wang J, 2008, Characterization of HCT116 human colon cancer cells in an orthotopic model. J Surg Res 147(2):276–281. , DOI 10.1016/j.jss.2007.04.021 Beauchemin N, 2011, The colorectal tumor microenvironment: the next decade. Cancer Microenviron 4(2):181–185. , DOI 10.1007/s12307-011-0074-7 Qin JZ, Upadhyay V, Prabhakar B, Maker AV, 2013, Shedding LIGHT TNFSF14 on the tumor microenvironment of colorectal cancer liver metastases. Journal of translational medicine 11:1–10. , DOI 10.1186/1479-5876-11-70 Bocuk D, Wolff A, Krause P, Salinas G, Bleckmann A, Hackl C, Beissbarth T, Koenig S, 2017, The adaptation of colorectal cancer cells when forming metastases in the liver: expression of associated genes and pathways in a mouse model. BMC Cancer 17:1–15 Lu Y, Zhao X, Li K, Luo G, Nie Y, Shi Y, Zhou Y, Ren G, Feng B, Liu Z, Pan Y, Li T, GuoX,Wu K,Miranda-VizueteA,Wang X, Fan D, 2013, Thioredoxin-like protein 2 is overexpressed in colon cancer and promotes cancer cell metastasis by interaction with ran. Antioxid Redox Signal 19(9):899–911. , DOI 10.1089/ ars.2012.4736 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2177 EP 2183 DI 10.1007/s12253-020-00805-3 PG 7 ER PT J AU Guo, J Cui, Z Zheng, Y Li, X Chen, Y AF Guo, Junying Cui, Zhaolei Zheng, Yuhong Li, Xiaoli Chen, Yan TI Comparison of Epstein-Barr Virus Serological Tools for the Screening and Risk Assessment of Nasopharyngeal Carcinoma: a Large Population-based Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Nasopharyngeal carcinoma; Epstein-Barr virus; VCA-IgA; EA-IgA; Rta-IgG; Serological screening; Diagnostic efficiency; ROC curve analysis ID Nasopharyngeal carcinoma; Epstein-Barr virus; VCA-IgA; EA-IgA; Rta-IgG; Serological screening; Diagnostic efficiency; ROC curve analysis AB Epstein-Barr virus (EBV)-based serologic antibody testing has been found to be a feasible alternative for nasopharyngeal carcinoma (NPC) screening in endemic areas. The purpose of this study was to evaluate the performance of ELISA based on VCA IgA antibody, EA-IgA and Rta-IgG antibody specific to EBV in the diagnosis of NPC. A total of 2155 untreated NPC patients and 6957 healthy volunteers without nasopharyngeal disorder were recruited, and all subjects received EBV VCA-IgA, EA-IgA and Rta-IgG antibody tests simultaneously. The diagnostic efficiency of three testing alone or in combination for the diagnosis of NPC was evaluated. The prevalence of IgA antibody against EBV-VCA, IgA antibody against EBV-EA and IgG antibody against EBV-Rta was 89.9%, 46.6% and 63.2%. The sensitivity, specificity, positive predictive value, negative predictive value and Youden index were 89.88%, 89.65%, 73.18%, 96.63% and 0.79 for the EBV VCA-IgA antibody test, 46.59%, 96.89%, 82.5%, 85.42%and 0.43 for the EA-IgA antibody test, and 63.25%, 94.87%, 79.48%, 89.29%and 0.58 for the Rta-IgG antibody test in the diagnosis of NPC, and ROC curve analysis revealed the greatest diagnostic efficiency for EBV VCA-IgA antibody test and the lowest efficiency for EBV EA-IgA antibody test in the diagnosis of NPC. In addition, the simultaneous triple positivity of VCA-IgA, EA-IgA and Rta-IgG antibodies specific to EBV indicated the highest risk of NPC, and the simultaneous triple negativity of the three types of anti-EBV antibodies suggested the lowest risk of NPC. Our data demonstrate that EBV VCA-IgA antibody test shows a higher diagnostic efficiency than EA-IgA and Rta-IgG antibody tests for the screening of NPC, and triple positivity of is a better biomarker for the diagnosis of NPC. C1 [Guo, Junying] Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fujian Provincial Key Laboratory of Tumor Biotherapy, No. 420 Fuma Road, 350014 Fuzhou, Fujian Province, China. [Cui, Zhaolei] Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fujian Provincial Key Laboratory of Tumor Biotherapy, No. 420 Fuma Road, 350014 Fuzhou, Fujian Province, China. [Zheng, Yuhong] Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fujian Provincial Key Laboratory of Tumor Biotherapy, No. 420 Fuma Road, 350014 Fuzhou, Fujian Province, China. [Li, Xiaoli] Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fujian Provincial Key Laboratory of Tumor Biotherapy, No. 420 Fuma Road, 350014 Fuzhou, Fujian Province, China. [Chen, Yan] Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fujian Provincial Key Laboratory of Tumor Biotherapy, No. 420 Fuma Road, 350014 Fuzhou, Fujian Province, China. RP Chen, Y (reprint author), Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fujian Provincial Key Laboratory of Tumor Biotherapy, 350014 Fuzhou, China. EM 19895758592@163.com CR Chua MLK, Wee JTS, Hui EP, Chan ATC, 2016, Nasopharyngeal carcinoma. Lancet 387:1012–1024 Tang LL, Chen WQ, Xue WQ, He YQ, Zheng RS, Zeng YX, Jia WH, 2016, Global trends in incidence and mortality of nasopharyngeal carcinoma. Cancer Lett 374:22–30 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68:394–424 Fu ZT, Guo XL, Zhang SW, Zeng HM, Sun KX, Chen WQ, He J, 2018, Incidence and mortality of nasopharyngeal carcinoma in China, 2014. Zhonghua Zhong Liu Za Zhi 40:566–571 Petersson F, 2015, Nasopharyngeal carcinoma: a review. Semin Diagn Pathol 32:54–73 Kamran SC, Riaz N, Lee N, 2015, Nasopharyngeal carcinoma. Surg Oncol Clin N Am 24:547–561 Tsao SW, Tsang CM, Lo KW, 2017, Epstein-Barr virus infection and nasopharyngeal carcinoma. Philos Trans R Soc Lond B Biol Sci 372:20160270 Tsang CM, Tsao SW, 2015, The role of Epstein-Barr virus infection in the pathogenesis of nasopharyngeal carcinoma. Virol Sin 30: 107–121 Lee AW, Ma BB, Ng WT, Chan AT, 2015, Management of nasopharyngeal carcinoma: current practice and future perspective. J Clin Oncol 33:3356–3364 Tabuchi K, Nakayama M, Nishimura B, Hayashi K, Hara A, 2011, Early detection of nasopharyngeal carcinoma. Int J Otolaryngol. 2011:638058 Hildesheim A, 2013, Epstein-Barr virus-based screening for the early detection of nasopharyngeal carcinoma: a new frontier. Am J Epidemiol 177:251–253 Ng MH, Chan KH, Ng SP, Zong YS, 2006, Epstein-Barr virus serology in early detection and screening of nasopharyngeal carcinoma. Ai Zheng 25:250–256 Cao SM, Liu Z, JiaWH, Huang QH, Liu Q, Guo X, Huang TB, Ye W, Hong MH, 2011, Fluctuations of epstein-barr virus serological antibodies and risk for nasopharyngeal carcinoma: a prospective screening study with a 20-year follow-up. PLoS One 6:e19100 Lee AWM, Ng WT, Chan LK, Chan OSH, Hung WM, Chan CC, Cheng PTC, Sze H, Lam TS, Yau TK, 2012, The strength/ weakness of the AJCC/UICC staging system, 7th edition, for nasopharyngeal cancer and suggestions for future improvement. Oral Oncol 48:1007–1013 Chan AT, Gregoire V, Lefebvre JL, Licitra L, Hui EP, Leung SF, Felip E, EHNS–ESMO–ESTRO Guidelines Working Group, 2012, Nasopharyngeal cancer: EHNS-ESMO-ESTRO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 23:vii83–vii85 Gurtsevitch VE, Senyuta NB, Ignatova AV, Lomaya MV, Kondratova VN, Pavlovskaya AI, Dushenkina TE, Maximovich DM, Smirnova KV, Mudunov AM, Lichtenstein AV, 2017, Epstein-Barr virus biomarkers for nasopharyngeal carcinoma in non-endemic regions. J Gen Virol 98:2118–2127 Anghel I, Anghel AG, Dumitru M, Soreanu CC, 2012, Nasopharyngeal carcinoma – analysis of risk factors and immunological markers. Chirurgia, Bucur, 107:640–645 Wong MM, Lye MS, Cheng HM, Sam CK, 2005, Epstein-Barr virus serology in the diagnosis of nasopharyngeal carcinoma. Asian Pac J Allergy Immunol 23:65–67 Chen Y, Xin X, Cui Z, Zheng Y, Guo J, Chen Y, Lin Y, Su G, 2016, Diagnostic value of serum Epstein-Barr virus capsid antigen-IgA for nasopharyngeal carcinoma: a meta-analysis based on 21 Studies. Clin Lab 62:1155–1166 Li Y, Wang K, Yin SK, Zheng HL, Min DL, 2016, Expression of Epstein-Barr virus antibodies EA-IgG, Rta-IgG, and VCA-IgA in nasopharyngeal carcinoma and their use in a combined diagnostic assay. Genet Mol Res 15:gmr7368 Cai YL, Li J, Lu AY, Zheng YM, Zhong WM, Wang W, Gao JQ, Zeng H, Cheng JR, Tang MZ, 2014, Diagnostic significance of combined detection of Epstein-Barr virus antibodies, VCA/IgA, EA/IgA, Rta/IgG and EBNA1/IgA for nasopharyngeal carcinoma. Asian Pac J Cancer Prev 15:2001–2006 Xia C, Zhu K, Zheng G, 2015, Expression of EBV antibody EAIgA, Rta-IgGand VCA-IgAand SA in serumand the implication of combined assay in nasopharyngeal carcinoma diagnosis. Int J Clin Exp Pathol 8:16104–16110 Sun P, Chen C, Cheng YK, Zeng ZJ, Chen XL, Liu LZ, Gu MF, 2014, Serologic biomarkers of Epstein-Barr virus correlate with TNM classification according to the seventh edition of the UICC/ AJCC staging system for nasopharyngeal carcinoma. Eur Arch Otorhinolaryngol 271:2545–2554 Sun R, Wang X, Li X, 2015, Correlation Analysis of Nasopharyngeal Carcinoma TNM Staging with Serum EA IgA and VCA IgA in EBV and VEGF-C and -D. Med Sci Monit 21: 2105–2109 Luo YL, Ou GP, Chi PD, Liang YN, Liu YH, Huang MY, 2009, Combined determination of Epstein-Barr virus-related antibodies and antigens for diagnosis of nasopharyngeal carcinoma. Ai Zheng 28:76–78 Li S, Deng Y, Li X, Chen QP, Liao XC, Qin X, 2010, Diagnostic value of Epstein-Barr virus capsid antigen-IgA in nasopharyngeal carcinoma: a meta-analysis. Chin Med J, Engl, 123:1201–1205 Shao JY, Li YH, Gao HY,Wu QL, Cui NJ, Zhang L, Cheng G, Hu LF, Ernberg I, Zeng YX(2004, Comparison of plasma Epstein-Barr virus, EBV, DNA levels and serum EBV immunoglobulin A/virus capsid antigen antibody titers in patients with nasopharyngeal carcinoma. Cancer 100:1162–1170 Song C, Yang S, 2013, A meta-analysis on the EBV DNA and VCA-IgA in diagnosis of Nasopharyngeal Carcinoma. Pak J Med Sci 29:885–890 Leung SF, Tam JS, Chan AT, Zee B, Chan LY, Huang DP, Van Hasselt A, Johnson PJ, Lo YM, 2004, Improved accuracy of detection of nasopharyngeal carcinoma by combined application of circulating Epstein-Barr virusDNAand anti-Epstein-Barr viral capsid antigen IgA antibody. Clin Chem 50:339–345 Chan KH, Gu YL, Ng F, Ng PS, Seto WH, Sham JS, Chua D,Wei W, Chen YL, Luk W, Zong YS, Ng MH, 2003, EBV specific antibody-based and DNA-based assays in serologic diagnosis of nasopharyngeal carcinoma. Int J Cancer 105:706–709 Zhao FP, Liu X, Zhong ZM, Lu J, Yu BL, Zeng FY, Chen XM, Chen HH, Peng XH, Wang F, Peng Y, Li XP, 2014, Positivity of both plasma Epstein-Barr virus DNA and serum Epstein-Barr virus capsid specific immunoglobulin A is a better prognostic biomarker for nasopharyngeal carcinoma. BBA Clin 2:88–93 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2185 EP 2190 DI 10.1007/s12253-020-00808-0 PG 6 ER PT J AU Malusecka, E Chmielik, E Suwinski, R Giglok, M Lange, D Rutkowski, T Mazurek, MA AF Malusecka, Ewa Chmielik, Ewa Suwinski, Rafal Giglok, Monika Lange, Dariusz Rutkowski, Tomasz Mazurek, M Agnieszka TI Significance of HPV16 Viral Load Testing in Anal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HPV; HPV16 viral load; p16INK4A; p53 ID HPV; HPV16 viral load; p16INK4A; p53 AB Human papilloma virus (HPV) is highly frequent among patients with anal squamous cell carcinoma, but the viral load (VL) differs between patients. This study aimed to compare the rate of HPV positivity, HPV16VL, p16INK4A and p53 expression between treatment naive and recurrent anal cancer patients. HPV was genotyped via AmpliSens® HPV HCR-genotype-titre-FRT kit. HPV16 VL was determined via quantitative polymerase chain reaction-based in-house test. p16INK4A and p53 expression was tested via immunohistochemistry. The cohort comprised 13 treatment-naive and 17 recurrent anal SCC patients. High-risk HPV was detected in 87% of cases, and HPV16 (73%) was the predominant genotype. The rate of HPV positivity was higher among women and nonsmokers, and majority of HPV-positive cases were also p16INK4A-positive. All p53-negative tumors were HPV16-positive. The most predominant p53 staining pattern in the HPV-positive group was scattered type, whereas it was diffuse type in the HPV-negative group. The HPV16 VL was higher in the treatment-naive group. Further, in the treatment-naive group, cases with scattered staining pattern of p53 had higher HPV16 VL than cases with diffuse staining pattern. The opposite result was noted in the recurrent cancer group. Moreover, p16-positive cases with scattered p53 staining pattern in the treatment naive group had higher HPV16 VL than their counterparts in the recurrent cancer group. In conclusion, the HPV VL, as is the association between VL and p16INK4A /p53, is in an inversed trend in treatment naive and recurrent cancer patients, highlighting the importance of HPV VL measurement in anal SCC. C1 [Malusecka, Ewa] Maria Sklodowska-Curie National Research Institute of Oncology, Center for Translational Research and Molecular Biology of Cancer, Gliwice Branch, 44-102 Gliwice, Poland. [Chmielik, Ewa] Maria Sklodowska-Curie National Research Institute of Oncology, Tumor Pathology DepartmentGliwice, Poland. [Suwinski, Rafal] Maria Sklodowska-Curie National Research Institute of Oncology, II Radiotherapy and Chemotherapy Clinic and Teaching HospitalGliwice, Poland. [Giglok, Monika] Maria Sklodowska-Curie National Research Institute of Oncology, II Radiotherapy and Chemotherapy Clinic and Teaching HospitalGliwice, Poland. [Lange, Dariusz] Maria Sklodowska-Curie National Research Institute of Oncology, Tumor Pathology DepartmentGliwice, Poland. [Rutkowski, Tomasz] Maria Sklodowska-Curie National Research Institute of Oncology, I Radiation and Clinical Oncology DepartmentGliwice, Poland. [Mazurek, M Agnieszka] Maria Sklodowska-Curie National Research Institute of Oncology, Center for Translational Research and Molecular Biology of Cancer, Gliwice Branch, 44-102 Gliwice, Poland. RP Malusecka, E (reprint author), Maria Sklodowska-Curie National Research Institute of Oncology, Center for Translational Research and Molecular Biology of Cancer, 44-102 Gliwice, Poland. EM Ewa.Malusecka@io.gliwice.pl CR Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA, eds). SEER Cancer Statistics Review, 1975–2016, National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/ csr/1975_2016/, based on November 2018 SEER data submission, posted to the SEER web site, April 2019 Litwin TR, Clarke MA, Dean M, Wentzensen N, 2017, Somatic host cell alterations in HPV carcinogenesis. Viruses 9(8). https:// doi.org/10.3390/v9080206 Review Ang KK, Sturgis EM, 2012, Human papillomavirus as a marker of the natural history and response to therapy of head and neck squamous cell carcinoma. Semin Radiat Oncol 22(2):128–142. https:// doi.org/10.1016/j.semradonc.2011.12.004 Review Wright AA, Howitt BE, Myers AP, Dahlberg SE, Palescandolo E, Van Hummelen P, MacConaill LE, Shoni M, Wagle N, Jones RT, Quick CM, Laury A, Katz IT, Hahn WC, Matulonis UA, Hirsch MS, 2013, Oncogenic mutations in cervical cancer: genomic differences between adenocarcinomas and squamous cell carcinomas of the cervix. Cancer 119(21):3776–3783. , DOI 10.1002/ cncr.28288 Chung CH, Guthrie VB, Masica DL, Tokheim C, Kang H, Richmon J, Agrawal N, Fakhry C, Quon H, Subramaniam RM, Zuo Z, Seiwert T, Chalmers ZR, Frampton GM, Ali SM, Yelensky R, Stephens PJ, Miller VA, Karchin R, Bishop JA, 2015, Genomic alterations in head and neck squamous cell carcinoma determined by cancer gene-targeted sequencing. Ann Oncol 26(6):1216–1223. , DOI 10.1093/annonc/mdv109 Chung JH, Sanford E, Johnson A, Klempner SJ, Schrock AB, Palma NA, Erlich RL, Frampton GM, Chalmers ZR, Vergilio J, Rubinson DA, Sun JX, Chmielecki J, Yelensky R, Suh JH, Lipson D, George TJ Jr, Elvin JA, Stephens PJ, Miller VA, Ross JS, Ali SM, 2016, Comprehensive genomic profiling of anal squamous cell carcinoma reveals distinct genomically defined classes. Ann Oncol 27(7):1336–1341. , DOI 10.1093/annonc/ mdw152 Cacheux W, Tsantoulis P, Briaux A, Vacher S, Mariani P, Richard- Molard M, Buecher B, Richon S, Jeannot E, Lazartigues J, Rouleau E,Mariani O, El AlamE, Cros J, Roman-Roman S,Mitry E, Girard E, Dangles-Marie V, Lievre A, Bieche I, 2018, Array comparative genomic hybridization identifies high level of PI3K/Akt/mTOR pathway alterations in anal cancer recurrences. Cancer Med. , DOI 10.1002/cam4.1533 Mouw KW, Cleary JM, Reardon B, Pike J, Braunstein LZ, Kim J, Amin-Mansour A, Miao D, Damish A, Chin J, Ott PA, Fuchs CS, Martin NE, Getz G, Carter S, Mamon HJ, Hornick JL, Van Allen EM, D'Andrea AD, 2017, Genomic evolution after Chemoradiotherapy in anal squamous cell carcinoma. Clin Cancer Res 23(12):3214–3222. , DOI 10.1158/1078-0432.CCR- 16-2017 Goon PK, Stanley MA, Ebmeyer J, Steinstrasser L, Upile T, Jerjes W, Bernal-Sprekelsen M, Gorner M, Sudhoff HH, 2009, HPV & head and neck cancer: a descriptive update. Head Neck Oncol 1:36. , DOI 10.1186/1758-3284-1-36 Review Prigge ES, Arbyn M, von Knebel Doeberitz M, Reuschenbach M, 2017, Diagnostic accuracy of p16(INK4a, immunohistochemistry in oropharyngeal squamous cell carcinomas: a systematic review and meta-analysis. Int J Cancer 140(5):1186–1198. , DOI 10.1002/ijc.30516 Review Singhi AD, Westra WH, 2010, Comparison of human papillomavirus in situ hybridization and p16 immunohistochemistry in the detection of human papillomavirus-associated head and neck cancer based on a prospective clinical experience. Cancer 116(9):2166– 2173. , DOI 10.1002/cncr.25033 AndoK, Oki E, Saeki H,Yan Z, Tsuda Y, Hidaka G,Kasagi Y, Otsu H, Kawano H, Kitao H, Morita M, Maehara Y, 2015, Discrimination of p53 immunohistochemistry-positive tumors by its staining pattern in gastric cancer. Cancer Med 4(1):75–83. , DOI 10.1002/cam4.346 Kaserer K, Schmaus J, Bethge U, Migschitz B, Fasching S, Walch A, Herbst F, Teleky B, Wrba F, 2000, Staining patterns of p53 immunohistochemistry and their biological significance in colorectal cancer. J Pathol Mar 190(4):450–456 Chang MS, Oh S, Jung EJ, Park JH, Jeon HW, Lee TS, Kim JH, Choi E, Byeon SJ, Park IA, 2014, High-risk human papillomavirus load and biomarkers in cervical intraepithelial neoplasia and cancer. APMIS 122(5):427–436. , DOI 10.1111/apm.12163 Rodel F,Wieland U, Fraunholz I, Kitz J, Rave-Frank M,Wolff HA, Weiss C, Wirtz R, Balermpas P, Fokas E, Rodel C, 2015, Human papillomavirus DNA load and p16INK4a expression predict for local control in patients with anal squamous cell carcinoma treated with chemoradiotherapy. Int J Cancer 136(2):278–288. https://doi. org/10.1002/ijc.28979 Biesaga B, Mucha-Malecka A, Janecka-Widla A,Kolodziej-Rzepa M, Szostek S, Slonina D, Kowalczyk A, Halaszka K, Przewoznik M, 2018, Differences in the prognosis of HPV16-positive patients with squamous cell carcinoma of head and neck according to viral load and expression of P16. J Cancer Res Clin Oncol 144(1):63–73. , DOI 10.1007/s00432-017-2531-2 Morris BJ, 2005, Cervical human papillomavirus screening by PCR: advantages of targeting the E6/E7 region. Clin Chem Lab Med 43(11):1171–1177 Review MazurekAM, Rutkowski T, SnieturaM, PiglowskiW, Suwinski R, Skladowski K, 2019, Detection of circulating HPV16 DNA as a biomarker in the blood of patients with human papillomaviruspositive oropharyngeal squamous cell carcinoma. Head Neck 41(3):632–641. , DOI 10.1002/hed.25368 Cocuzza CE,MartinelliM, Sina F, Piana A, Sotgiu G, Dell'Anna T, Musumeci R, 2017, Human papillomavirusDNA detection in plasma and cervical samples of women with a recent history of low grade or precancerous cervical dysplasia. PLoS One 12(11): e0188592. , DOI 10.1371/journal.pone.0188592 eCollection 2017 Kim J, Kim BK, Jeon DS, Lee CH, Roh JW, Kim JY, Park SY, 2019, Type-specific viral load and physical state of HPV type 16, 18, and 58 as diagnostic biomarkers for high-grade squamous intraepithelial lesions or cervical Cancer. Cancer Res Treat. https:// doi.org/10.4143/crt.2019.152 Lillsunde Larsson G, KaliffM, Sorbe B, Helenius G, Karlsson MG, 2018, HPV16 viral characteristics in primary, recurrent and metastatic vulvar carcinoma. Papillomavirus Res 6:63–69. https://doi. org/10.1016/j.pvr.2018.10.008 Hu Z, Liu L, Zhang W, Liu H, Li J, Jiang L, Zeng K, 2018, Dynamics of HPV viral loads reflect the treatment effect of photodynamic therapy in genital warts. Photodiagn Photodyn Ther 21: 86–90. , DOI 10.1016/j.pdpdt.2017.11.005 Badaracco G, Savarese A, Micheli A, Rizzo C, Paolini F, CarosiM, Cutillo G, Vizza E, Arcangeli G, Venuti A, 2010, Persistence of HPVafter radio-chemotherapy in locally advanced cervical cancer. Oncol Rep 23(4):1093–1099 Castellsague X, Munoz N, 2003, Chapter 3: cofactors in human papillomavirus carcinogenesis–role of parity, oral contraceptives, and tobacco smoking. J Natl Cancer Inst Monogr, 31):20–28 Review Ding L, Liu C, Zhou Q, Feng M, Wang J, 2019, Association of estradiol and HPV/HPV16 infection with the occurrence of cervical squamous cell carcinoma. Oncol Lett. , DOI 10.3892/ol. 2019.10005 Auborn KJ, Woodworth C, DiPaolo JA, Bradlow HL, 1991, The interaction between HPVinfection and estrogen metabolism in cervical carcinogenesis. Int J Cancer 49(6):867–869 Newfield L, Bradlow HL, Sepkovic DW, Auborn K, 1998, Estrogen metabolism and the malignant potential of human papillomavirus immortalized keratinocytes. Proc Soc Exp Biol Med 217(3):322–326 Bandyopadhyay S, Chatterjee R, 2006, HPV viral load determination during pregnancy as a possible cervical cancer risk. J Exp Clin Cancer Res 25(1):29–38 Wei L, Griego AM, Chu M, Ozbun MA, 2014, Tobacco exposure results in increased E6 and E7 oncogene expression, DNA damage and mutation rates in cells maintaining episomal human papillomavirus 16 genomes. Carcinogenesis 35(10):2373–2381. https://doi. org/10.1093/carcin/bgu156 Schiffman MH, Haley NJ, Felton JS, Andrews AW, Kaslow RA, LancasterWD, Kurman RJ, Brinton LA, LannomLB, Hoffmann D, 1987, Biochemical epidemiology of cervical neoplasia: measuring cigarette smoke constituents in the cervix. Cancer Res 47(14): 3886–3888 ParkNH, Gujuluva CN, Baek JH, Cherrick HM, Shin KH, Min BM, 1995, Combined oral carcinogenicity of HPV-16 and benzo(a)pyrene: an in vitro multistep carcinogenesis model. Oncogene 10(11):2145–2153 Xi LF, Koutsky LA, Castle PE, Edelstein ZR, Meyers C, Ho J, Schiffman M, 2009, Relationship between cigarette smoking and human papilloma virus types 16 and 18 DNA load. Cancer Epidemiol Biomark Prev 18(12):3490–3496. , DOI 10. 1158/1055-9965.EPI-09-0763 Meulendijks D, Tomasoa NB, Dewit L, Smits PH, Bakker R, van Velthuysen ML, Rosenberg EH, Beijnen JH, Schellens JH, Cats A, 2015, HPV-negative squamous cell carcinoma of the anal canal is unresponsive to standard treatment and frequently carries disruptive mutations in TP53. Br J Cancer 112(8):1358–1366. , DOI 10.1038/bjc.2015.20 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2191 EP 2199 DI 10.1007/s12253-020-00801-7 PG 9 ER PT J AU Geczi, L Bodoky, Gy Rokszin, Gy Fabian, I Torday, L AF Geczi, Lajos Bodoky, Gyorgy Rokszin, Gyorgy Fabian, Ibolya Torday, Laszlo TI Survival Benefits of Second-line Axitinib Versus Everolimus After First Line Sunitinib Treatment in Metastatic Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Metastatic renal cell carcinoma; Axitinib; Sequential therapy; VEGF inhibitors; Overall survival ID Metastatic renal cell carcinoma; Axitinib; Sequential therapy; VEGF inhibitors; Overall survival AB Background: Targeted therapies significantly improve clinical outcomes among patients with metastatic renal cell carcinoma (mRCC). Several new agents have been approved for first- and second-line use. However, there is a lack of compelling evidence comparing sequencing strategies, and available comparative data regarding the real-world effectiveness of different therapeutic sequences are limited. Materials and Methods: We identified mRCC patients who initiated targeted therapy between January 1, 2008 and May 31, 2017 from the National Health Insurance Fund (NHIF) database of Hungary. Overall survival (OS) and duration of first-line treatment (DFT) were obtained for patients receiving sunitinib-everolimus, sunitinib-axitinib, or pazopanib-everolimus treatment sequences. OS of sunitinib-everolimus and sunitinib-axitinib sequences was also determined for patients having better or worse response to sunitinib first-line therapy. Results: Median OS was significantly longer among patients treated with sunitinib-axitinib compared to those receiving sunitinib-everolimus. Median DFT was also significantly longer in the sunitinib-axitinib vs. sunitinib-everolimus group. Sunitinib-axitinib was associated with significantly longer median OS compared to sunitinib-everolimus in patients with better response to first-line sunitinib in the pooled sunitinib population. In patients with worse response to sunitinib, sunitinib-axitinib was associated with a trend towards greater OS compared to sunitinib-everolimus, but the difference did not reach statistical significance. Conclusions: In this nationwide database analysis, mRCC patients treated with the sunitinib-axitinib sequence had significantly longer OS compared to those receiving sunitinib-everolimus therapy. The OS benefits of second-line axitinib were consistent among patients with better response to sunitinib defined by DFT values. C1 [Geczi, Lajos] National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology Department, Rath Gyorgy u. 7–9, 1122 Budapest, Hungary. [Bodoky, Gyorgy] Egyesitett Szent Istvan es Szent Laszlo Korhaz, Onkologiai Osztaly, Albert Florian ut 5, 1097 Budapest, Hungary. [Rokszin, Gyorgy] RxTarget Ltd, Bacso Nandor ut 10, 5000 Szolnok, Hungary. [Fabian, Ibolya] RxTarget Ltd, Bacso Nandor ut 10, 5000 Szolnok, Hungary. [Torday, Laszlo] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, 6720 Szeged, Hungary. RP Geczi, L (reprint author), National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology Department, 1122 Budapest, Hungary. EM gelajos@oncol.hu CR Ferrara N, Gerber HP, LeCouter J, 2003, The biology of VEGF and its receptors. Nat Med 9:669–676 Carmeliet P, 2005, VEGF as a key mediator of angiogenesis in cancer. Oncology 69(suppl 3):4–10 Albiges L, Salem M, Rini B et al, 2011, Vascular endothelial growth factor-targeted therapies in advanced renal cell carcinoma. Hematol Oncol Clin North Am 25:813–833 Hutson TE, 2011, Targeted therapies for the treatment of metastatic renal cell carcinoma: clinical evidence. Oncologist 16(suppl 2):14– 22 Molina AM, Motzer RJ, 2011, Clinical practice guidelines for the treatment of metastatic renal cell carcinoma: today and tomorrow. Oncologist 16(suppl 2):45–50 Mendel DB, Laird AD,XinX et al, 2003, In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res 9:327–337 Motzer RJ, Mazumdar M, Bacik J et al, 1999, Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma. J Clin Oncol 17:2530–2540 Escudier B, Porta C, Schmidinger M et al, 2016, Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 27(suppl 5):v58–v68 Motzer RJ, Jonasch E, Agarwal N et al, 2017, Kidney Cancer, Version 2.2017, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 15:804–834 Powles T, Albiges L, Staehler M et al, 2017, Updated European Association of Urology Guidelines Recommendations for the Treatment of First-line Metastatic Clear Cell Renal Cancer. Eur Urol. , DOI 10.1016/j.eururo.2017.11.016 Rini BI, Atkins MB, 2009, Resistance to targeted therapy in renalcell carcinoma. Lancet Oncol 10:992–1000 Joosten SC, Hamming L, Soetekouw PM, 2015, Resistance to sunitinib in renal cell carcinoma: From molecular mechanisms to predictive markers and future perspectives. Biochim Biophys Acta 1855:1–16 Butz H, Ding Q, Nofech-Mozes R et al, 2017, Elucidating mechanisms of sunitinib resistance in renal cancer: an integrated pathological-molecular analysis. Oncotarget 9:4661–4674 Rini BI, Escudier B, Tomczak P et al, 2011, Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma, AXIS): a randomised phase 3 trial. Lancet 378:1931–1939 Motzer RJ, Escudier B, Oudard S et al, 2008, Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372:449–456 Sternberg CN, Davis ID, Mardiak J et al, 2010, Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 28:1061–1068 Motzer RJ, Hutson TE, Tomczak P et al, 2009, Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J Clin Oncol 27:3584– 3590 Gore ME, Szczylik C, Porta C et al, 2015, Final results from the large sunitinib global expanded-access trial in metastatic renal cell carcinoma. Br J Cancer 113:12–19 Noize P, Grelaud A, Bay JO et al, 2017, Real-life patterns of use, safety and effectiveness of sunitinib in first-line therapy of metastatic renal cell carcinoma: the SANTORIN cohort study. Pharmacoepidemiol Drug Saf 26:1561–1569 Koutsoukos K, Bamias A, Tzannis K et al, 2017, Real-world experience of everolimus as second-line treatment in metastatic renal cell cancer after failure of pazopanib. Onco Targets Ther 10:4885– 4893 Guida A, Albiges L, Derosa L et al, 2017, Everolimus Versus Axitinib as Second-line Therapy in Metastatic Renal Cell Carcinoma: Experience From Institut Gustave Roussy. Clin Genitourin Cancer 15:e1081–e1088 Iqbal S, BadreldinW, Sundar S et al, 2016, Comparison of Axitinib and Everolimus in the Second Line Treatment of Metastatic Renal Cell Carcinoma Following Previous Treatment with a Tyrosine Kinase Inhibitor: a Multicentre Retrospective Analysis. Clin Onc 28:e9–e16 Basappa NS, Bjarnason G, Hansen G et al, 2018, Comparing outcomes of second line axitinib or everolimus in metastatic renal cell carcinoma patients: Results from the Canadian Kidney Cancer information system. Presented at the 73rd Canadian Urological Association Annual Meeting, Halifax, Nova Scotia, Canada. Poster UP-10.1 Gill DM, Agarwal N, 2018, Rapidly Changing Treatment Landscape of mRCC: What Is the Optimal Sequencing of Agents? https://gucasym.org/daily-news/rapidly-changingtreatment- landscape-mrcc-what-optimal-sequencing-agents. Accessed 14 November 2018 Choueiri TK, Halabi S, Sanford BL et al, 2017, Cabozantinib Vesrus Sunitinib As Initial Targeted Therapy fot Patients With Metastatic Renal Cell Carcinoma of Poor or Intermediate Risk: The Alliance A031203 CABOSUN Trial. J Clin Oncol 35:591–598 Motzer RJ, Penkov K, Haanen J et al, 2019, Avelumab plus Axitinib versus Sunitinb for Advanced Renal-Cell Carcinoma. N Engl J Med 380:1103–1115 Rini BI, Plimack ER, Stus V, 2019, Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med 380:1116–1127 Rini BI, Powles T, Atkins MB et al, 2019, Atezolizumab plus bevacizumab versus sunitinib in patients with previously untreated metastatic renal cell carcinoma, IMmotion151): a multicentre, open-label, phase 3, randomised controlled trial. Lancet 393: 2404–2415 Lalani AA, Li H, Heng DYC et al, 2017, First-line sunitinib or pazopanib in metastatic renal cell carcinoma: The Canadian experience. Can Urol Assoc J 11:112–117 Climent MA, Munoz-Langa J, Basterretxea-Badiola L et al, 2018, Systematic review and survival meta-analysis of real world evidence on first-line pazopanib for metastatic renal cell carcinoma. Crit Rev Oncol Hematol 121:45–50 Motzer RJ, Hutson TE, Cella D et al, 2013, Pazopanib versus sunitinib in metastatic renal-cell carcinoma. N Engl J Med 369: 722–731 Haaland B, Chopra A, Acharyya S et al, 2014, Comparative effectiveness of approved first-line anti-angiogenic and molecularly targeted therapeutic agents in the treatment of good and intermediate risk metastatic clear cell renal cell carcinoma. BMC Cancer 14: 592 Choueiri TK, Escudier B, Powles T et al, 2015, Cabozantinib versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med 373:1814–1823 Motzer RJ, Hutson TE, Glen H et al, 2015, Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial. Lancet Oncol 16:1473–1482 Motzer RJ, Escudier B, McDermott DF et al, 2015, Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med 373:1803–1813 Atkins MB, Clark JI, Quinn DI, 2017, Immune checkpoint inhibitors in advanced renal cell carcinoma: experience to date and future directions. Ann Oncol 28:1484–1494 Heng DY, Signorovitch J, Swallow E et al, 2014, Comparative Effectiveness of Second-Line Targeted Therapies for Metastatic Renal Cell Carcinoma: A Systematic Review and Meta-Analysis of Real-World Observational Studies. PLoS One 9:e114264 MacLean E, Cisar L,Mehle K et al, 2016, Real-World Axitinib Use in the United States: A Retrospective Study Using Linked Datasets. J Manag Care Spec Pharm 22:723–732u D’Aniello C, Vitale MG, Farnesi A et al, 2016, Axitinib after Sunitinib in Metastatic Renal Cancer: Preliminary Results from Italian “Real-World” SAX Study. Front Pharmacol 7:331 Matias M, Le Teuff G, Albiges L et al, 2017, Real world prospective experience of axitinib in metastatic renal cell carcinoma in a large comprehensive cancer centre. Eur J Cancer 79:185–192 Motzer RJ, Tannir NM, McDermott DF et al, 2018, Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N Engl J Med 375:1277–1290 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2201 EP 2207 DI 10.1007/s12253-020-00809-z PG 7 ER PT J AU Galamb, O Kalmar, A Sebestyen, A Danko, T Kriston, Cs Furi, I Hollosi, P Csabai, I Wichmann, B Krenacs, T Bartak, KB Nagy, BZs Zsigrai, S Barna, G Tulassay, Zs Igaz, P Molnar, B AF Galamb, Orsolya Kalmar, Alexandra Sebestyen, Anna Danko, Titanilla Kriston, Csilla Furi, Istvan Hollosi, Peter Csabai, Istvan Wichmann, Barnabas Krenacs, Tibor Bartak, Kinga Barbara Nagy, Brigitta Zsofia Zsigrai, Sara Barna, Gabor Tulassay, Zsolt Igaz, Peter Molnar, Bela TI Promoter Hypomethylation and Increased Expression of the Long Non-coding RNA LINC00152 Support Colorectal Carcinogenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE LINC00152; CYTOR; Long non-codingRNA; Colorectal cancer; Colorectal adenoma ID LINC00152; CYTOR; Long non-codingRNA; Colorectal cancer; Colorectal adenoma AB Up-regulation of the long non-coding RNA LINC00152 can contribute to cancer development, proliferation and invasion, including colorectal cancer, however, its mechanism of action in colorectal carcinogenesis and progression is only insufficiently understood. In this work we correlated LINC00152 expression with promoter DNA methylation changes in colorectal tissues along the normal-adenoma-carcinoma sequence and studied the effects of LINC00152 silencing on the cell cycle regulation and on the whole transcriptome in colon carcinoma cells using cell and molecular biology techniques. LINC00152 was significantly up-regulated in adenoma and colorectal cancer (p < 0.001) compared to normal samples, which was confirmed by real-time PCR and in situ hybridization. LINC00152 promoter hypomethylation detected in colorectal cancer (p < 0.01) was strongly correlated with increased LINC00152 expression (r=-0.90). Silencing of LINC00152 significantly suppressed cell growth, induced apoptosis and decreased cyclin D1 expression (p < 0.05). Whole transcriptome analysis of LINC00152-silenced cells revealed significant down-regulation of oncogenic and metastasis promoting genes (e.g. YES proto-oncogene 1, PORCN porcupine Oacyltransferase), and up-regulation of tumour suppressor genes (e.g. DKK1 dickkopfWNTsignalling pathway inhibitor 1, PERP p53 apoptosis effector) (adjusted p < 0.05). Pathway analysis confirmed the LINC00152-related activation of oncogenic molecular pathways including those driven by PI3K/Akt, Ras, WNT, TP53, Notch and ErbB. Our results suggest that promoter hypomethylation related overexpression of LINC00152 can contribute to the pathogenesis of colorectal cancer by facilitating cell progression through the up-regulation of several oncogenic and metastasis promoting pathway elements. C1 [Galamb, Orsolya] Semmelweis University, Szentkiralyi str 46, 1088 Budapest, Hungary. [Kalmar, Alexandra] Semmelweis University, Szentkiralyi str 46, 1088 Budapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Danko, Titanilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kriston, Csilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Furi, Istvan] Semmelweis University, Szentkiralyi str 46, 1088 Budapest, Hungary. [Hollosi, Peter] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Csabai, Istvan] Eotvos Lorand University, Department of Physics of Complex SystemsBudapest, Hungary. [Wichmann, Barnabas] Semmelweis University, Szentkiralyi str 46, 1088 Budapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Bartak, Kinga Barbara] Semmelweis University, Szentkiralyi str 46, 1088 Budapest, Hungary. [Nagy, Brigitta Zsofia] Semmelweis University, Szentkiralyi str 46, 1088 Budapest, Hungary. [Zsigrai, Sara] Semmelweis University, Szentkiralyi str 46, 1088 Budapest, Hungary. [Barna, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Tulassay, Zsolt] Hungarian Academy of Sciences, Molecular Medicine Research GroupBudapest, Hungary. [Igaz, Peter] Semmelweis University, Szentkiralyi str 46, 1088 Budapest, Hungary. [Molnar, Bela] Semmelweis University, Szentkiralyi str 46, 1088 Budapest, Hungary. RP Galamb, O (reprint author), Semmelweis University, 1088 Budapest, Hungary. EM udvardyne_galamb.orsolya@semmelweis-univ.hu CR Ferlay J, SoerjomataramI, Dikshit R, Eser S, Mathers C, RebeloM, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386. https://doi. org/10.1002/ijc.29210 PancioneM, Remo A, Colantuoni V, 2012, Genetic and epigenetic events generate multiple pathways in colorectal cancer progression. Patholog Res Int 2012:509348. , DOI 10.1155/2012/ 509348 Kornienko AE, Guenzl PM, Barlow DP, Pauler FM, 2013, Gene regulation by the act of long non-coding RNA transcription. BMC Biol 11:59. , DOI 10.1186/1741-7007-11-59 Gutschner T, Diederichs S, 2012, The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol 9(6):703–719. https:// doi.org/10.4161/rna.20481 Ma Y, YangY,Wang F, MoyerMP,Wei Q, Zhang P, Yang Z, LiuW, Zhang H, Chen N, Wang H, Wang H, Qin H, 2016, Long noncoding RNA CCAL regulates colorectal cancer progression by activating Wnt/beta-catenin signalling pathway via suppression of activator protein 2alpha. Gut 65(9):1494–1504. , DOI 10. 1136/gutjnl-2014-308392 Xia T, Liao Q, Jiang X, Shao Y, Xiao B, Xi Y, Guo J, 2014, Long noncoding RNA associated-competing endogenous RNAs in gastric cancer. Sci Rep 4:6088. , DOI 10.1038/srep06088 CaoWJ,Wu HL, He BS, Zhang YS, Zhang ZY, 2013, Analysis of long non-coding RNA expression profiles in gastric cancer.World J Gastroenterol 19(23):3658–3664. , DOI 10.3748/wjg.v19. i23.3658 ChenWM, HuangMD, Sun DP, Kong R, Xu TP, Xia R, Zhang EB, Shu YQ, 2016, Long intergenic non-coding RNA 00152 promotes tumor cell cycle progression by binding to EZH2 and repressing p15 and p21 in gastric cancer. Oncotarget 7(9):9773–9787. https:// doi.org/10.18632/oncotarget.6949 Ji J, Tang J, Deng L, Xie Y, Jiang R, Li G, Sun B, 2015, LINC00152 promotes proliferation in hepatocellular carcinoma by targeting EpCAM via the mTOR signaling pathway. Oncotarget 6(40):42813–42824. , DOI 10.18632/ oncotarget.5970 Pang Q, Ge J, Shao Y, SunW, Song H, Xia T, Xiao B, Guo J, 2014, Increased expression of long intergenic non-coding RNA LINC00152 in gastric cancer and its clinical significance. Tumour Biol 35(6):5441–5447. , DOI 10.1007/s13277-014-1709-3 Zhao J, Liu Y, Zhang W, Zhou Z, Wu J, Cui P, Zhang Y, Huang G, 2015, Long non-coding RNA Linc00152 is involved in cell cycle arrest, apoptosis, epithelial to mesenchymal transition, cell migration and invasion in gastric cancer. Cell Cycle 14(19):3112–3123. , DOI 10.1080/15384101.2015.1078034 Li Q, Shao Y, Zhang X, Zheng T, Miao M, Qin L, Wang B, Ye G, Xiao B, Guo J, 2015, Plasma long noncoding RNA protected by exosomes as a potential stable biomarker for gastric cancer. Tumour Biol 36(3):2007–2012. , DOI 10.1007/s13277-014-2807-y Li J,Wang X, Tang J, Jiang R, ZhangW, Ji J, Sun B, 2015, HULC and Linc00152 Act as Novel Biomarkers in Predicting Diagnosis of Hepatocellular Carcinoma. Cell Physiol Biochem 37(2):687–696. , DOI 10.1159/000430387 Neumann O, KesselmeierM, Geffers R, Pellegrino R, Radlwimmer B, Hoffmann K, Ehemann V, Schemmer P, Schirmacher P, Lorenzo Bermejo J, Longerich T, 2012)Methylome analysis and integrative profiling of human HCCs identify novel protumorigenic factors. Hepatology 56(5):1817–1827. , DOI 10.1002/hep.25870 Zhou J, Zhi X, Wang L, Wang W, Li Z, Tang J, Wang J, Zhang Q, Xu Z, 2015, Linc00152 promotes proliferation in gastric cancer through the EGFR-dependent pathway. J Exp Clin Cancer Res 34:135. , DOI 10.1186/s13046-015-0250-6 Bian Z, Zhang J, Li M, Feng Y, Yao S, SongM, Qi X, Fei B, Yin Y, Hua D, Huang Z, 2017, Long non-coding RNA LINC00152 promotes cell proliferation, metastasis, and confers 5-FU resistance in colorectal cancer by inhibiting miR-139-5p. Oncogenesis 6(11): 395. , DOI 10.1038/s41389-017-0008-4 Yue B, Cai D, Liu C, Fang C, Yan D, 2016, Linc00152 Functions as a Competing Endogenous RNA to Confer Oxaliplatin Resistance and Holds Prognostic Values in Colon Cancer. Mol Ther 24(12): 2064–2077. , DOI 10.1038/mt.2016.180 Qiu JJ, Yan JB, 2015, Long non-coding RNA LINC01296 is a potential prognostic biomarker in patients with colorectal cancer. Tumour Biol 36(9):7175–7183. , DOI 10.1007/s13277- 015-3448-5 Galamb O, Gyorffy B, Sipos F, Spisak S, Nemeth AM, Miheller P, Tulassay Z, Dinya E,Molnar B, 2008, Inflammation, adenoma and cancer: objective classification of colon biopsy specimens with gene expression signature. Dis Markers 25(1):1–16 Galamb O, Sipos F, Solymosi N, Spisak S, Krenacs T, Toth K, Tulassay Z, Molnar B, 2008, Diagnostic mRNA expression patterns of inflamed, benign, and malignant colorectal biopsy specimen and their correlation with peripheral blood results. Cancer Epidemiol Biomarkers Prev 17(10):2835–2845. , DOI 10. 1158/1055-9965.EPI-08-0231 Galamb O, Wichmann B, Sipos F, Spisak S, Krenacs T, Toth K, Leiszter K, Kalmar A, Tulassay Z, Molnar B, 2012, Dysplasiacarcinoma transition specific transcripts in colonic biopsy samples. PLoS One 7(11):e48547. , DOI 10.1371/journal.pone. 0048547 Kalmar A, Nagy ZB, Galamb O, Csabai I, Bodor A,Wichmann B, Valcz G, Bartak BK, Tulassay Z, Igaz P, Molnar B, 2019, Genomewide expression profiling in colorectal cancer focusing on lncRNAs in the adenoma-carcinoma transition. BMC Cancer 19(1):1059. , DOI 10.1186/s12885-019-6180-5 Galamb O, Kalmar A, Peterfia B, Csabai I, Bodor A, Ribli D, Krenacs T, Patai AV, Wichmann B, Bartak BK, Toth K, Valcz G, Spisak S, Tulassay Z,Molnar B, 2016, Aberrant DNA methylation of WNT pathway genes in the development and progression of CIMP-negative colorectal cancer. Epigenetics 11(8):588–602. , DOI 10.1080/15592294.2016.1190894 Luo Y, Wong CJ, Kaz AM, Dzieciatkowski S, Carter KT, Morris SM, Wang J, Willis JE, Makar KW, Ulrich CM, Lutterbaugh JD, Shrubsole MJ, Zheng W, Markowitz SD, Grady WM, 2014, Differences in DNA methylation signatures reveal multiple pathways of progression from adenoma to colorectal cancer. Gastroenterology 147(2):418-429 e418. , DOI 10.1053/j. gastro.2014.04.039 Cancer Genome Atlas N, 2012, Comprehensive molecular characterization of human colon and rectal cancer. Nature 487(7407):330– 337. , DOI 10.1038/nature11252 Anderson CM, Zhang B, Miller M, Butko E, Wu X, Laver T, Kernag C, Kim J, Luo Y, Lamparski H, Park E, Su N, Ma XJ, 2016, Fully Automated RNAscope In Situ Hybridization Assays for Formalin-Fixed Paraffin-Embedded Cells and Tissues. J Cell Biochem 117(10):2201–2208. , DOI 10.1002/jcb.25606 Jackson AL, Linsley PS, 2010, Recognizing and avoiding siRNA off-target effects for target identification and therapeutic application. Nat Rev Drug Discov 9(1):57–67. , DOI 10.1038/ nrd3010 Darzynkiewicz Z, Bruno S, Del Bino G, Gorczyca W, Hotz MA, Lassota P, Traganos F, 1992, Features of apoptotic cells measured by flow cytometry. Cytometry 13(8):795–808. , DOI 10. 1002/cyto.990130802 Galamb O, Spisak S, Sipos F, Toth K, Solymosi N, Wichmann B, Krenacs T, Valcz G, Tulassay Z, Molnar B, 2010, Reversal of gene expression changes in the colorectal normal-adenoma pathway by NS398 selective COX2 inhibitor. Br J Cancer 102(4):765–773. , DOI 10.1038/sj.bjc.6605515 Cai Q, Wang ZQ, Wang SH, Li C, Zhu ZG, Quan ZW, Zhang WJ, 2016, Upregulation of long non-coding RNA LINC00152 by SP1 contributes to gallbladder cancer cell growth and tumor metastasis via PI3K/AKT pathway. Am J Transl Res 8(10):4068–4081 Wang Y, Liu J, Bai H, Dang Y, Lv P, Wu S, 2017, Long intergenic non-coding RNA 00152 promotes renal cell carcinoma progression by epigenetically suppressing P16 and negatively regulates miR- 205. Am J Cancer Res 7(2):312–322 Deng X, Zhao XF, Liang XQ, Chen R, Pan YF, Liang J, 2017, Linc00152 promotes cancer progression in hepatitis B virusassociated hepatocellular carcinoma. Biomed Pharmacother 90: 100–108. , DOI 10.1016/j.biopha.2017.03.031 Hu HB, Jie HY, Zheng XX, 2016, Three Circulating LncRNA Predict Early Progress of Esophageal Squamous Cell Carcinoma. Cell Physiol Biochem 40(1–2):117–125. , DOI 10.1159/ 000452529 Yu X, Lin Y, Sui W, Zou Y, Lv Z, 2017, Analysis of distinct long noncoding RNA transcriptional fingerprints in pancreatic ductal adenocarcinoma. Cancer Med 6(3):673–680. , DOI 10. 1002/cam4.1027 Wu Y, Tan C,WengWW, Deng Y, Zhang QY, Yang XQ, Gan HL, Wang T, Zhang PP, XuMD,WangYQ,Wang CF, 2016, Long noncoding RNA Linc00152 is a positive prognostic factor for and demonstrates malignant biological behavior in clear cell renal cell carcinoma. Am J Cancer Res 6(2):285–299 Yu J, Liu Y, Guo C, Zhang S, Gong Z, Tang Y, Yang L, He Y, Lian Y, Li X, Deng H, Liao Q, Li X, Li Y, Li G, Zeng Z, XiongW, Yang X, 2017, Upregulated long non-coding RNA LINC00152 expression is associated with progression and poor prognosis of tongue squamous cell carcinoma. J Cancer 8(4):523–530. , DOI 10.7150/jca.17510 Matsuyama T, Ishikawa T, MogushiK, Yoshida T, Iida S, Uetake H, Mizushima H, Tanaka H, Sugihara K, 2010, MUC12 mRNA expression is an independent marker of prognosis in stage II and stage III colorectal cancer. Int J Cancer 127(10):2292–2299. https://doi. org/10.1002/ijc.25256 Hong Y, Downey T, Eu KW, Koh PK, Cheah PY, 2010, A “metastasis- prone” signature for early-stage mismatch-repair proficient sporadic colorectal cancer patients and its implications for possible therapeutics. Clin Exp Metastasis 27(2):83–90. , DOI 10. 1007/s10585-010-9305-4 Zhang YH, Fu J, Zhang ZJ, Ge CC, Yi Y, 2016, LncRNALINC00152 down-regulated by miR-376c-3p restricts viability and promotes apoptosis of colorectal cancer cells. Am J Transl Res 8(12):5286–5297 Luo J, Qu J, Wu DK, Lu ZL, Sun YS, Qu Q, 2017, Long noncoding RNAs: a rising biotarget in colorectal cancer. Oncotarget 8(13):22187–22202. , DOI 10.18632/oncotarget.14728 Nishizawa Y, Konno M, Asai A, Koseki J, Kawamoto K, Miyoshi N, Takahashi H, Nishida N, Haraguchi N, Sakai D, Kudo T, Hata T, Matsuda C,Mizushima T, Satoh T, Doki Y,Mori M, Ishii H, 2018, Hypoxia stimulates the cytoplasmic localization of oncogenic long noncoding RNA LINC00152 in colorectal cancer. Int J Oncol 52(2):453–460. , DOI 10.3892/ijo.2017.4218 Yue B, Liu C, Sun H, Liu M, Song C, Cui R, Qiu S, Zhong M, 2018, A Positive Feed-Forward Loop between LncRNA-CYTOR and Wnt/beta-Catenin Signaling Promotes Metastasis of Colon Cancer. Mol Ther 26(5):1287–1298. , DOI 10.1016/j. ymthe.2018.02.024 Marisa L, de Reynies A, Duval A, Selves J, Gaub MP, Vescovo L, Etienne-Grimaldi MC, Schiappa R, Guenot D, Ayadi M, Kirzin S, Chazal M, Flejou JF, Benchimol D, Berger A, Lagarde A, Pencreach E, Piard F, Elias D, Parc Y, Olschwang S, Milano G, Laurent-Puig P, Boige V, 2013, Gene expression classification of colon cancer into molecular subtypes: characterization, validation, and prognostic value. PLoS Med 10(5):e1001453. , DOI 10.1371/journal.pmed.1001453 Ernst J,Kheradpour P, Mikkelsen TS, ShoreshN,Ward LD, Epstein CB, Zhang X, Wang L, Issner R, Coyne M, Ku M, Durham T, Kellis M, Bernstein BE, 2011, Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473(7345):43–49. , DOI 10.1038/nature09906 Han H, Cortez CC, Yang X, Nichols PW, Jones PA, Liang G, 2011, DNA methylation directly silences genes with non-CpG island promoters and establishes a nucleosome occupied promoter. HumMol Genet 20(22):4299–4310. , DOI 10.1093/hmg/ddr356 Oster B, Linnet L, Christensen LL, Thorsen K, Ongen H, Dermitzakis ET, Sandoval J, Moran S, Esteller M, Hansen TF, Lamy P, Laurberg S, Orntoft TF, Andersen CL, 2013, Non-CpG island promoter hypomethylation and miR-149 regulate the expression of SRPX2 in colorectal cancer. Int J Cancer 132(10):2303– 2315. , DOI 10.1002/ijc.27921 Marx A, Kahan T, Simon I, 2013, Integrative analysis of methylome and transcriptome reveals the importance of unmethylated CpGs in non-CpG island gene activation. Biomed Res Int 2013:785731. , DOI 10.1155/2013/785731 Yu Y, Yang J, Li Q, Xu B, Lian Y, Miao L, 2017, LINC00152: A pivotal oncogenic long non-coding RNA in human cancers. Cell Prolif 50, 4). , DOI 10.1111/cpr.12349 Zhang Y, Xiang C,Wang Y, Duan Y, Liu C, Jin Y, Zhang Y, 2017, lncRNA LINC00152 knockdown had effects to suppress biological activity of lung cancer via EGFR/PI3K/AKT pathway. Biomed Pharmacother 94:644–651. , DOI 10.1016/j.biopha.2017. 07.120 Rosenbluh J, Nijhawan D, CoxAG, Li X,Neal JT, Schafer EJ, Zack TI,Wang X, Tsherniak A, Schinzel AC, Shao DD, Schumacher SE, Weir BA, Vazquez F, Cowley GS, Root DE, Mesirov JP, Beroukhim R, Kuo CJ, Goessling W, Hahn WC, 2012, beta- Catenin-driven cancers require a YAP1 transcriptional complex for survival and tumorigenesis. Cell 151(7):1457–1473. https:// doi.org/10.1016/j.cell.2012.11.026 Madan B, Ke Z, Harmston N, Ho SY, Frois AO, Alam J, Jeyaraj DA, Pendharkar V, Ghosh K, Virshup IH, Manoharan V, Ong EH, Sangthongpitag K, Hill J, Petretto E, Keller TH, LeeMA,Matter A, Virshup DM, 2016, Wnt addiction of genetically defined cancers reversed by PORCN inhibition. Oncogene 35(17):2197–2207. , DOI 10.1038/onc.2015.280 Beaudry VG, Jiang D, Dusek RL, Park EJ, Knezevich S, Ridd K, Vogel H, Bastian BC, Attardi LD, 2010, Loss of the p53/p63 regulated desmosomal protein Perp promotes tumorigenesis. PLoS Genet 6(10):e1001168. , DOI 10.1371/journal.pgen. 1001168 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2209 EP 2223 DI 10.1007/s12253-020-00800-8 PG 15 ER PT J AU Beer, A Taghizadeh, H Schiefer, AI Puhr, H Karner, A Jomrich, G Schoppmann, S Kain, R Preusser, M Ilhan-Mutlu, A AF Beer, Andrea Taghizadeh, Hossein Schiefer, Ana-Iris Puhr, C. Hannah Karner, K. Alexander Jomrich, Gerd Schoppmann, F. Sebastian Kain, Renate Preusser, Matthias Ilhan-Mutlu, Aysegul TI PD-L1 and HER2 Expression in Gastroesophageal Cancer: a Matched Case Control Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastroesophageal tumor; Esophageal tumor; Gastroesophageal junction tumor; Gastric tumor; PD-L1; HER2; Immunotherapy; TPS; CPS ID Gastroesophageal tumor; Esophageal tumor; Gastroesophageal junction tumor; Gastric tumor; PD-L1; HER2; Immunotherapy; TPS; CPS AB Immunotherapy with check-point inhibitors serves as a promising treatment strategy in patients with upper gastrointestinal (GI) tumors. Human epidermal growth factor receptor 2 (HER2) is the only identified therapeutic target in upper GI tumors, whose potential interaction with programmed death-ligand 1 (PD-L1) is unknown. The aim of this study was the investigation of PD-L1 and HER2 in upper GI tumors.We retrospectively identified patients with HER2 positive gastroesophageal cancers and matched them with a HER2 negative group. We investigated the tumor specimens for HER2 status and PD-L1 expression, with the following assessments being performed: i) staining of tumor cells in terms of tumor proportion score (TPS), ii) staining for tumorassociated immune cells (TAIs), iii) interface pattern and iv) combined positive score (CPS). Both HER2 positive and negative group consisted of 59 patients. Expression of PD-L1 in TAIs and interface pattern were associated with a favorable outcome (p = 0.02, HR = 0.8; p = 0.04, HR = 0.39; respectively) in patients with localized disease, whereas TPS was associated with an unfavorable outcome in patients with advanced tumor (p = 0.02, HR = 1.4). These effects were HER2 independent. PD-L1 expression in its different assessment is equally observed in HER2 positive and negative patients. Future studies will show whether dual inhibition of HER2 and PD-L1 improves survival of this selected patient population. C1 [Beer, Andrea] Medical University of Vienna, Department of PathologyVienna, Austria. [Taghizadeh, Hossein] Medical University of Vienna, Comprehensive Cancer Center Vienna, Upper GI Tumors UnitVienna, Austria. [Schiefer, Ana-Iris] Medical University of Vienna, Department of PathologyVienna, Austria. [Puhr, C. Hannah] Medical University of Vienna, Comprehensive Cancer Center Vienna, Upper GI Tumors UnitVienna, Austria. [Karner, K. Alexander] Medical University of Vienna, Comprehensive Cancer Center Vienna, Upper GI Tumors UnitVienna, Austria. [Jomrich, Gerd] Medical University of Vienna, Comprehensive Cancer Center Vienna, Upper GI Tumors UnitVienna, Austria. [Schoppmann, F. Sebastian] Medical University of Vienna, Comprehensive Cancer Center Vienna, Upper GI Tumors UnitVienna, Austria. [Kain, Renate] Medical University of Vienna, Department of PathologyVienna, Austria. [Preusser, Matthias] Medical University of Vienna, Comprehensive Cancer Center Vienna, Upper GI Tumors UnitVienna, Austria. [Ilhan-Mutlu, Aysegul] Medical University of Vienna, Comprehensive Cancer Center Vienna, Upper GI Tumors UnitVienna, Austria. RP Ilhan-Mutlu, A (reprint author), Medical University of Vienna, Comprehensive Cancer Center Vienna, Upper GI Tumors Unit, Vienna, Austria. EM aysegul.ilhan@meduniwien.ac.at CR Smyth EC, Verheij M, Allum W, Cunningham D, Cervantes A, Arnold D, Committee EG, 2016, Gastric cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 27(suppl 5):v38–v49. , DOI 10.1093/annonc/ mdw350 Cancer Genome Atlas Research N, 2014, Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513(7517): 202–209. , DOI 10.1038/nature13480 Cho J, Lee J, Bang H, Kim ST, Park SH, An JY, Choi MG, Lee JH, Sohn TS, Bae JM,KangWK, KimS,KimKM(2017, Programmed cell death-ligand 1 expression predicts survival in patients with gastric carcinoma with microsatellite instability. Oncotarget 8, 8): 13320-13328. , DOI 10.18632/oncotarget.14519 Polom K,Marano L,Marrelli D, De Luca R, Roviello G, Savelli V, Tan P, Roviello F, 2017, Meta-analysis of microsatellite instability in relation to clinicopathological characteristics and overall survival in gastric cancer. Br J Surg 105:159–167. , DOI 10.1002/ bjs.10663 Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M, 2012, Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366(26):2443–2454. https://doi. org/10.1056/NEJMoa1200690 Muro K, Chung HC, Shankaran V, Geva R, Catenacci D, Gupta S, Eder JP, Golan T, Le DT, Burtness B,McRee AJ, Lin CC, Pathiraja K, Lunceford J, Emancipator K, Juco J, KoshijiM, Bang YJ, 2016, Pembrolizumab for patients with PD-L1-positive advanced gastric cancer, KEYNOTE-012): a multicentre, open-label, phase 1b trial. Lancet Oncol 17(6):717–726. , DOI 10.1016/S1470- 2045(16)00175-3 Fuchs CS, Doi T, Jang RW, Muro K, Satoh T, MachadoM, SunW, Jalal SI, Shah MA, Metges JP, Garrido M, Golan T, Mandala M, Wainberg ZA, Catenacci DV, Ohtsu A, Shitara K, Geva R, Bleeker J, Ko AH, Ku G, Philip P, Enzinger PC, Bang YJ, Levitan D,Wang J, Rosales M, Dalal RP, Yoon HH, 2018, Safety and efficacy of Pembrolizumab Monotherapy in patients with previously treated advanced gastric and Gastroesophageal junction Cancer: phase 2 clinical KEYNOTE-059 trial. JAMA Oncol 4(5):e180013. https:// doi.org/10.1001/jamaoncol.2018.0013 Kang YK, Boku N, Satoh T, RyuMH, Chao Y, Kato K, Chung HC, Chen JS, Muro K, Kang WK, Yeh KH, Yoshikawa T, Oh SC, Bai LY, Tamura T, Lee KW, Hamamoto Y, Kim JG, Chin K, Oh DY, Minashi K, Cho JY, Tsuda M, Chen LT, 2017, Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens, ONO-4538-12, ATTRACTION-2): a randomised, double- blind, placebo-controlled, phase 3 trial. Lancet 390(10111): 2461–2471. , DOI 10.1016/S0140-6736(17)31827-5 Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T, Aprile G, Kulikov E, Hill J, Lehle M, Ruschoff J, Kang YK, To GATI, 2010, Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer, ToGA): a phase 3, openlabel, randomised controlled trial. Lancet 376(9742):687–697. , DOI 10.1016/S0140-6736(10)61121-X Jorgensen JT, 2010, Targeted HER2 treatment in advanced gastric cancer. Oncology 78(1):26–33. , DOI 10.1159/000288295 Van Cutsem E, Bang YJ, Feng-Yi F, Xu JM, Lee KW, Jiao SC, Chong JL, Lopez-Sanchez RI, Price T, Gladkov O, Stoss O, Hill J, Ng V, Lehle M, Thomas M, Kiermaier A, Ruschoff J, 2015, HER2 screening data from ToGA: targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer 18(3):476–484. https:// doi.org/10.1007/s10120-014-0402-y Gravalos C, Jimeno A, 2008, HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol 19(9):1523– 1529. , DOI 10.1093/annonc/mdn169 Hofmann M, Stoss O, Shi D, Buttner R, van de Vijver M, Kim W, Ochiai A, Ruschoff J, Henkel T, 2008, Assessment of a HER2 scoring system for gastric cancer: results from a validation study. Histopathology 52(7):797–805. , DOI 10.1111/j.1365- 2559.2008.03028.x Kim KC, Koh YW, Chang HM, Kim TH, Yook JH, Kim BS, Jang SJ, Park YS, 2011, Evaluation of HER2 protein expression in gastric carcinomas: comparative analysis of 1,414 cases of wholetissue sections and 595 cases of tissue microarrays. Ann Surg Oncol 18(10):2833–2840. , DOI 10.1245/s10434-011- 1695-2 Shitara K, Yatabe Y, Matsuo K, Sugano M, Kondo C, Takahari D, Ura T, Tajika M, Ito S, Muro K, 2013, Prognosis of patients with advanced gastric cancer byHER2 status and trastuzumab treatment. Gastric cancer : official journal of the International Gastric Cancer Association and the Japanese Gastric Cancer Association 16(2): 261–267. , DOI 10.1007/s10120-012-0179-9 Gu J, Zheng L, Wang Y, Zhu M, Wang Q, Li X, 2014, Prognostic significance of HER2 expression based on trastuzumab for gastric cancer, ToGA, criteria in gastric cancer: an updated meta-analysis. Tumour Biol 35(6):5315–5321. , DOI 10.1007/s13277- 014-1693-7 Ilhan-Mutlu A, Taghizadeh H, Beer A, Dolak W, Ba-Ssalamah A, Schoppmann SF, Hejna M, Birner P, PreusserM(2018, Correlation of trastuzumab-based treatment with clinical characteristics and prognosis in HER2-positive gastric and gastroesophageal junction cancer: A retrospective single center analysis. Cancer biology & therapy 19(3):169–174. , DOI 10.1080/15384047.2017. 1414759 Hecht JR, Bang YJ, Qin SK, Chung HC, Xu JM, Park JO, Jeziorski K, Shparyk Y, Hoff PM, Sobrero A, Salman P, Li J, Protsenko SA, Wainberg ZA, Buyse M, Afenjar K, Houe V, Garcia A, Kaneko T, Huang Y, Khan-Wasti S, Santillana S, Press MF, Slamon D, 2016, Lapatinib in combination with Capecitabine plus Oxaliplatin in human epidermal growth factor receptor 2-positive advanced or metastatic gastric, esophageal, or Gastroesophageal adenocarcinoma: TRIO-013/LOGiC–A randomized phase III trial. J Clin Oncol 34(5):443–451. , DOI 10.1200/JCO.2015.62.6598 J. Tabernero PMH, L. Shen, A. Ohtsu, M.A. Shah, K. Cheng, C. Song, H. Wu, J. Eng-Wong, Y. Kang, 2017, Pertuzumab, P, + trastuzumab, H, + chemotherapy, CT, for HER2-positive metastatic gastric or gastro-oesophageal junction cancer, mGC/GEJC). Annals of Oncology 28, suppl_5):209–268 Satoh T, Xu RH, Chung HC, Sun GP, Doi T, Xu JM, Tsuji A, Omuro Y, Li J, Wang JW, Miwa H, Qin SK, Chung IJ, Yeh KH, Feng JF, Mukaiyama A, Kobayashi M, Ohtsu A, Bang YJ, 2014, Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN–a randomized, phase III study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 32(19):2039–2049. , DOI 10.1200/JCO.2013. 53.6136 Thuss-Patience PC, Shah MA, Ohtsu A, Van Cutsem E, Ajani JA, Castro H, Mansoor W, Chung HC, Bodoky G, Shitara K, Phillips GDL, van der Horst T, Harle-Yge ML, Althaus BL, Kang YK, 2017, Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma, GATSBY): an international randomised, open-label, adaptive, phase 2/3 study. Lancet Oncol 18(5):640–653. , DOI 10.1016/S1470-2045(17, 30111-0 Lauren P, 1965, The two histological Main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a Histo-clinical classification. Acta Pathol Microbiol Scand 64: 31–49 Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S,MooneyM, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J, 2009, New response evaluation criteria in solid tumours: revised RECIST guideline, version 1.1). European journal of cancer, Oxford, England : 1990, 45(2):228–247. , DOI 10.1016/j.ejca. 2008.10.026 Hofheinz R, Hegewisch-Becker S, Thuss-Patience PC, Kunzmann V, Fuchs M, Graeven U, Homann N, Heinemann V, Pohl M, Tannapfel A, Al-Batran SE, 2014, HER-FLOT: Trastuzumab in combination with FLOTas perioperative treatment for patients with HER2-positive locally advanced esophagogastric adenocarcinoma: Aphase II trial of theAIOgastric Cancer study group. JCOabstract. , DOI 10.1200/jco.2014.32.15_suppl.4073 journal of clinical oncology 32, no. 15_suppl, May 20 2014, 4073-4073 Pham T, Roth S, Kong J, Guerra G, Narasimhan V, Pereira L, Desai J, Heriot A, Ramsay R, 2018, An update on immunotherapy for solid tumors: A review. Ann Surg Oncol 25:3404–3412. https://doi. org/10.1245/s10434-018-6658-4 Oki E, Okano S, Saeki H, Umemoto Y, Teraishi K, Nakaji Y, Ando K, Zaitsu Y, Yamashita N, Sugiyama M, Nakashima Y, Ohgaki K, Oda Y,Maehara Y, 2017, Protein expression of programmed death 1 ligand 1 and HER2 in gastric carcinoma. Oncology 93(6):387– 394. , DOI 10.1159/000479231 Wang L, Zhang Q, Ni S, Tan C, Cai X, Huang D, Sheng W, 2018, Programmed death-ligand 1 expression in gastric cancer: correlation with mismatch repair deficiency and HER2-negative status. Cancer Med 7(6):2612–2620. , DOI 10.1002/cam4.1502 Cheng A-L QS, Ikeda M et al, 2019, IMbrave150: efficacy and safety results from a Ph 3 study evaluating Atezolizumab, atezo, + Bevacizumab, bev, vs Sorafenib, sor, as first treatment, tx, for patients, pts, with Unresectable hepatocellular carcinoma, HCC). ESMO Asia 2019:LBA3 Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, Alekseev B, Soulieres D, Melichar B, Vynnychenko I, Kryzhanivska A, Bondarenko I, Azevedo SJ, Borchiellini D, Szczylik C, Markus M, McDermott RS, Bedke J, Tartas S, Chang YH, Tamada S, Shou Q, Perini RF, ChenM, Atkins MB, Powles T, Investigators K, 2019, Pembrolizumab plus Axitinib versus Sunitinib for advanced renal-cell carcinoma. N Engl J Med 380(12):1116–1127. , DOI 10.1056/NEJMoa1816714 Y.Y. Janjigian SM, J.F. Chou, A.R. Gabler, M.Z. Simmons, P. Momtaz, M. Shcherba, G.Y. Ku, E. Won, F. Sanchez-Vega, H. Gerdes, D.P. Kelsen, D.H. Ilson, D. Solit, N. Schultz, P.M. Shah, M. Capanu, J.F. Hechtman, M. Lamendola-Essel, 2019, First-line pembrolizumab, p), trastuzumab, t), capecitabine, c, and oxaliplatin, o, in her2-positive metastatic esophagogastric adenocarcinoMA. ESMO 2019 Annals of Oncology, 2019, 30, suppl_5): v253-v324. , DOI 10.1093/annonc/mdz247 Chang H, Jung WY, Kang Y, Lee H, Kim A, Kim HK, Shin BK, Kim BH, 2016, Programmed death-ligand 1 expression in gastric adenocarcinoma is a poor prognostic factor in a high CD8+ tumor infiltrating lymphocytes group. Oncotarget 7, 49):80426-80434. , DOI 10.18632/oncotarget.12603 Dai C, Geng R,Wang C,Wong A, Qing M, Hu J, Sun Y, Lo AW, Li J, 2016, Concordance of immune checkpoints within tumor immune contexture and their prognostic significance in gastric cancer. Mol Oncol 10(10):1551–1558. , DOI 10.1016/j. molonc.2016.09.004 Kollmann D, Ignatova D, Jedamzik J, Chang YT, Jomrich G, Baierl A, Kazakov D,MichalM, French LE, HoetzeneckerW, Schatton T, Asari R, PreusserM,GnantM, Guenova E, Schoppmann SF, 2018, PD-L1 expression is an independent predictor of favorable outcome in patients with localized esophageal adenocarcinoma. Oncoimmunology 7(6):e1435226. , DOI 10.1080/ 2162402X.2018.1435226 Gu L, Chen M, Guo D, Zhu H, Zhang W, Pan J, Zhong X, Li X, Qian H, Wang X, 2017, PD-L1 and gastric cancer prognosis: A systematic review and meta-analysis. PLoS One 12(8):e0182692. , DOI 10.1371/journal.pone.0182692 Kim ST, Cristescu R, Bass AJ, Kim KM, Odegaard JI, Kim K, Liu XQ, Sher X, Jung H, Lee M, Lee S, Park SH, Park JO, Park YS, Lim HY, Lee H, ChoiM, Talasaz A, Kang PS, Cheng J, Loboda A, Lee J, KangWK, 2018, Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med 24:1449–1458. , DOI 10.1038/s41591-018- 0101-z Janjigian YY, Sanchez-Vega F, Jonsson P, Chatila WK, Hechtman JF, Ku GY, Riches JC, Tuvy Y, Kundra R, Bouvier N, Vakiani E, Gao J, Heins ZJ, Gross BE, Kelsen DP, Zhang L, Strong VE, Schattner M, Gerdes H, Coit DG, Bains M, Stadler ZK, Rusch VW, Jones DR, Molena D, Shia J, Robson ME, Capanu M, Middha S, Zehir A, Hyman DM, Scaltriti M, Ladanyi M, Rosen N, Ilson DH, Berger MF, Tang L, Taylor BS, Solit DB, Schultz N, 2018, Genetic predictors of response to systemic therapy in Esophagogastric Cancer. Cancer Discov 8(1):49–58. https://doi. org/10.1158/2159-8290.CD-17-0787 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2225 EP 2235 DI 10.1007/s12253-020-00814-2 PG 11 ER PT J AU Qadir, J Majid, S Khan, M Rashid, F Wani, DM Din, I Bashir, H AF Qadir, Jasiya Majid, Sabhiya Khan, S. Mosin Rashid, Fouzia Wani, Din Mumtaz Din, Inshah Bashir, Haamid TI AT-rich Interaction Domain 1A Gene Variations: Genetic Associations and Susceptibility to Gastric Cancer Risk SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ARID1A; Gastric cancer; Pro912Thr; Gln944Lys; Gln920Ter; Kashmiri Population ID ARID1A; Gastric cancer; Pro912Thr; Gln944Lys; Gln920Ter; Kashmiri Population AB AT-rich interaction domain containing protein 1A (ARID1A), has recently emerged as a novel class of gene which acts as a potent tumor suppressor in numerous types of cancers such asGastric, Breast, Ovarian, Colorectal, Lung cancers. ARID1A is involved in the regulation of various cellular processes such as proliferation, differentiation and DNA repair, yet its association with the susceptibility of cancer remains unknown. Here, we aimed to analyse the association of the ARID1A variants (Pro912Thr, Gln944Lys and Gln920Ter) with the risk of Gastric cancer (GC) in Kashmiri population. The study included 103 confirmed cases of GC and 163 normal controls. The genotypes were studied using Polymerase Chain Reaction. Different bioinformatic predictive tools were also used to analyse the possible effect of these SNP’s on the resultant protein. The Pro912Thr and Gln920Ter variants of ARID1A showed significant difference in genotypic and allelic frequencies between the GC cases and controls (P < 0.05), whereas, the data did not reveal any correlation between Gln944Lys variant and Gastric cancer risk. Both Pro912Thr and Gln920Ter SNP’s follow “Dominant mode of inheritance”. In Silico analysis predicted that amino acid substitution of Pro912Thr SNP decreases the protein stability thus changing the functional properties of resultant protein, so backing the possibility of damaging effect of this SNP. Our study suggests that Pro912Thr and Gln920Ter SNP’s of ARD1A gene are associated with increased risk of GC in Kashmiri population. C1 [Qadir, Jasiya] Government Medical College Srinagar and Associated Hospitals, Department of Biochemistry and Research Centre University of Kashmir, 190010 Srinagar, India. [Majid, Sabhiya] Government Medical College Srinagar and Associated Hospitals, Department of Biochemistry and Research Centre University of Kashmir, 190010 Srinagar, India. [Khan, S. Mosin] Government Medical College Srinagar and Associated Hospitals, Department of Biochemistry and Research Centre University of Kashmir, 190010 Srinagar, India. [Rashid, Fouzia] University of Kashmir, Department of Clinical Biochemistry, 190006 Srinagar, India. [Wani, Din Mumtaz] Government Medical College Srinagar and Associated Hospitals, Department of Surgery, 190010 Srinagar, India. [Din, Inshah] Government Medical College Srinagar and Associated Hospitals, Department of Biochemistry and Research Centre University of Kashmir, 190010 Srinagar, India. [Bashir, Haamid] Government Medical College Srinagar and Associated Hospitals, Department of Biochemistry and Research Centre University of Kashmir, 190010 Srinagar, India. RP Majid, S (reprint author), Government Medical College Srinagar and Associated Hospitals, Department of Biochemistry and Research Centre University of Kashmir, 190010 Srinagar, India. EM sabuumajid@gmail.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and Mortality worldwide for 36 cancers in 185 countries. CA: A Cancer. Journal for Clinicians 68:394–424 QurieshiMA,Khan SM,MasoodiMA, Qurieshi U, AinQ, JanYQ, Haq I, Sheikh ZA, 2016, Epidemiology of cancers in Kashmir, India: an analysis of hospital data. Adv Prev Med 02:1–6 Iqbal QM, Ganai AM, Bhat GM, Fazili AB, 2016, Pattern and magnitude of various cancers registered at regional centre of a tertiary care institute in North India. Int J Community Med Public Health 3:1672–1680 Wang D, Chen Y, Pan K, Wang W, Chen S, Chen J, Zhao J, Lv L, Pan Q, Li Y, Wang Q, Huang L, Ke M, He J, Xia J, 2012, Decreased expression of the ARID1A gene is associated with poor prognosis in primary gastric cancer. PLoS One 7:e40364 Wani I, Parray F, Wani RA, Naqash SH, Wani KA, Malik AA, Choudri NA, Wani MA, Khan NA, Sheikh TA, 2013, Noon chai and gastric cancer. Int J Case Rep Images 4(3):138–142 Wu R, Wang T, Shih L, 2014, The emerging roles of ARID1A in tumor suppression. Cancer Biology therapy 15:655–664 Guan B, Mao TL, Panuganti PK, Kuhn E, Kurman RJ, Maeda D, Chen E, Jeng YM,Wang TL, Shih IM, 2011, Mutation and loss of expression of ARID1A in uterine low-grade endometrioid carcinoma. Am J Surg Pathol 35:625–632 Wu JN, Roberts CWM, 2013, ARID1A mutations in cancer: Another epigenetic tumor suppressor? Cancer Discov 3:35–43 Huang HN, LinMC, HuangWC, Chiang YC, Kuo KT, 2014, Loss of ARID1A expression and its relationship with PI3K-Akt pathway alterations and ZNF217 amplification in ovarian clear cell carcinoma. Mod Pathol 27:983–990 Mamo A, Cavallone L, Tuzmen S, Chabot C, Ferrario C, Hassan S, Edgren H, Kallioniemi O, Aleynikova O, Przybytkowski E, Malcolm K, Mousses S, Tonin PN, Basik M, 2012, An integrated genomic approach identifies ARID1A as a candidate tumorsuppressor gene in breast cancer. Oncogene 31:2090–2100 Samartzis EP, Noske A, Dedes KJ, Fink D, Imesch P, 2013, ARID1A Mutations and PI3K/AKT Pathway Alterations in Endometriosis and Endometriosis-Associated Ovarian Carcinomas. Int J Mol Sci 14:18824–18849 Jones S, Meng L, Parsons W, Zhang X, Wesseling J, Kristel P, Schmidt MK, Markowitz S, Yan H, Bigner D, Hruban RH, Eshleman JR, Iacobuzio-Donahue CA, Goggins M, Maitra A, Malek SN, Powell S, Vogelstein B, Kinzler KW, Velculescu VE, PapadopoulosN(2012, Somaticmutations in the chromatin remodeling gene ARID1A occur in several tumor types. Hum Mutat 33: 100–103 Wiegand KC, Lee AF, Al-Agha OM, Chow C, Kalloger SE, Scott DW, Steidl C, Wiseman SM, Gascoyne RD, Gilks B, Huntsman DG, 2011, Loss of BAF250a, ARID1A, is frequent in high-grade endometrial carcinomas. J Pathol 224:328–333 Cajuso T, Hanninen UA, Kondelin J, Gylfe AE, Tanskanen T, Katainen R, Pitkanen E, Ristolainen H, Kaasinen E, Taipale M, Taipale J, Bohm J, Renkonen-Sinisalo L, Mecklin J, Jarvinen H, Tuupanen S, Kilpivaara O, Vahteristo P, 2014, Exome sequencing reveals frequent inactivating mutations in ARID1A, ARID1B, ARID2, and ARID4A in microsatellite unstable colorectal cancer. Int J Cancer 135:611–623 Deng N, Zhou H, FanYuan YH, 2017, Single nucleotide polymorphisms and cancer susceptibility. Oncotarget 8:110635–110649 Chu D,Wai L, 2019, Nonsynonymous, synonymous and nonsense mutations in human cancer-related genes undergo stronger purifying selection than expectation. BMC Cancer 19:359–365 Sanger F, Nicklen S, Coulson AR, 1977, DNA sequencing with chainterminating inhibitors. Proc Natl Acad Sci U S A 74:5463–5467 Fadare O, Renshaw IL, Liang SX, 2012, Does the loss of ARID1A, baf-250a, expression in endometrial clear cell carcinomas have any clinicopathologic significance? A pilot assessment. J Cancer 3:129–136 Yang L, Wei S, Zhao R, Wu Y, Qiu H, Xiong H, 2016, Loss of ARID1A expression predicts poor survival prognosis in gastric cancer: a systematic meta-analysis from 14 studies. Sci Rep 6: 2891–2899 Wang K, Kan J, Yuen ST, Shi ST, Chu KM, Law S, Chan TL, Kan Z, Chan ASY, TsuiWY, Lee SP, Ho SL, Chan AKW, Cheng GHW, Robert PC, Rejto PA, Gibson NW, Pocalyko DJ, Moa MXJ, Leung SY, 2011, Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat Genet 43: 1219–1212 Database of Single Nucleotide Polymorphism, dbSNP). Bethesda, MD): National Center for Biotechnology Information, National Library of Medicine., dbSNP Build ID: http://www.ncbi.nim.nih. gov/snp/?term=rs753300592 ) Venselaar H, Te Beek TA, Kuipers RK, Hekkelman ML, Vriend G, 2010, Protein structure analysis of mutations causing inheritable diseases: an e-Science approach with life scientist friendly interfaces. BMC Bioinformatics 11:548–554 Capriotti E, Fariselli P, Calabrese R, Casadio R, 2005, Predicting protein stability changes from sequences using support vector machines. Bioinformatics 21(Suppl 2):54–58 Cheng J, Randall A, Baldi P, 2005, Prediction of protein stability changes for single-site mutations using support vector machines. Proteins 62:1125–1132 Osier MV, Pakstis AJ, Goldman D, Edenberg HJ, Kidd JR, Kidd KK, 2002, A proline–threonine substitution in codon 351 of ADH1C is common in Native Americans. Clin Exp Res 26:1759– 1763 Karimi P, Islami F, Anandasabapathy S, Freedmen ND, Kamangar F, 2014, Gastric Cancer: Descriptive epidemiology, risk factors, screening and prevention. Cancer Epidemiol Biomarkers Prev 23: 700–713 Lee SR, Kim HO, Yoo CH, 2012, Impact of chronologic age in the elderly with gastric cancer. J Korean Surg Soc 82:211–218 Pak LM, Wang J, 2006, The appropriate treatment for elderly gastric cancer patients. Art Surg 1:4–9 Bernini M, Barbi S, Roviello F, Scarpa A, Moore P, Pedrazzani C, Beghelli S, Marrelli D, Manzoni GD, 2006, Family history of gastric cancer: a correlation between epidemiologic findings and clinical data. Gastric Cancer 9:9–13 Tavana Z, Khalili A, Namazi G, Ebrahimi A, Davoodi S, Alborizi S, Roozmeh S, 2018, Prevalence of common polymorphisms of AT-rich interaction domain 1A and endothelial nitric oxide synthase in patients with endometriosis compared to control group. J Endometriosis Pelvic Pain Disord 10:26–31 Bidou L, Bugaud O, Belakhov V, Baasov T, Namy O, 2017, Characterization of new-generation aminoglycoside promoting premature termination codon readthrough in cancer cells. RNA Biol 14:378–388 Ueno D, Matsumoto H, Kubota H, Higashida M, Akiyama T, Shiotani A, Hirai T, 2017, Prognostic factors for gastrectomy in elderly patients with gastric cancer. World J Surg Onc 15:59–65 Neuhaus H, 2017, Early gastric cancer in super-agers: to treator not to treat? Gastrointest Endosc 85:973–975 Lin Y, Ueda J, Kikuchi S, Totsuka Y, Wei WQ, Qiao YL, Inoue M, 2011, Comparative epidemiology of gastric cancer between Japan and China. World J Gastroenterol 17:4421–4428 Wang Q, 2016, Cancer predisposition genes: molecular mechanisms and clinical impact on personalized cancer care: examples of Lynch and HBOC syndromes. Acta Pharmacol Sin 37:143–149 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2237 EP 2246 DI 10.1007/s12253-020-00815-1 PG 10 ER PT J AU Watanabe, T Sato, K Kono, T Yamagishi, Y Kumazawa, F Miyamoto, M Takano, M Tsuda, H AF Watanabe, Takanori Sato, Kimiya Kono, Takako Yamagishi, Yoji Kumazawa, Fumihisa Miyamoto, Morikazu Takano, Masashi Tsuda, Hitoshi TI Aquaporin 3 Expression in Endometrioid Carcinoma of the Uterine Body Correlated With Early Stage and Lower Grade SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Aquaporin; water channel; uterus; endometrioid carcinoma; immunohistochemistry ID Aquaporin; water channel; uterus; endometrioid carcinoma; immunohistochemistry AB Aquaporins (AQPs) are a family of transmembrane water channel proteins distributed in various human tissues. Recent studies revealed that AQPs play important roles in cancer biology. Few studies have documented the relationship between the prognosis, stage, and histological grade of uterine endometrioid carcinoma, with AQP expression. Hence, the present study aimed to investigate this relationship between uterine endometrioid carcinoma and AQP expression.We retrospectively reviewed records of the patients who underwent surgery for uterine body cancer between 1990 and 2010 at the National Defense Medical College Hospital, Saitama, Japan. In 241 cases of endometrioid carcinoma, we immunohistochemically examined the expression of AQP 1, 2, 3, 4, and 5, and their relationship with clinicopathological parameters and the patients’ prognosis. We investigated the relationship between the clinicopathological parameters and AQP3 expression, and found that as the FIGO stage and histological grade progressed, the percentage of AQP3 expression tends to decrease. Furthermore, we analyzed progression-free survival/ overall survival (PFS/OS) using the log-rank test, and found that the AQP3-positive group had a better prognosis than AQP3- negative group (PFS: P < 0.001, OS: P = 0.002, respectively). Using Cox’s univariate proportional hazard model, we revealed that AQP3 had a low hazard ratio. However, according to Cox’s multivariate proportional hazard model, AQP3 was not an independent prognostic factor. Among the endometrioid carcinoma patients, the AQP3-positive group was associated with early stage and lower grade compared to the AQP3-negative group. Therefore, AQP3 has the potential to serve as a predictor of prognosis, although further investigation is necessary to elucidate the biologicalmechanismofAQP3 in endometrioid carcinoma. C1 [Watanabe, Takanori] National Defense Medical College, Department of Basic Pathology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Sato, Kimiya] National Defense Medical College, Department of Basic Pathology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Kono, Takako] National Defense Medical College, Department of Basic Pathology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Yamagishi, Yoji] National Defense Medical College, Department of Basic Pathology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Kumazawa, Fumihisa] National Defense Medical College, Department of Basic Pathology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. [Miyamoto, Morikazu] National Defense Medical College, Department of Obstetrics and GynecologyTokorozawa, Saitama, Japan. [Takano, Masashi] National Defense Medical College, Department of Obstetrics and GynecologyTokorozawa, Saitama, Japan. [Tsuda, Hitoshi] National Defense Medical College, Department of Basic Pathology, 3-2 Namiki, 359-8513 Tokorozawa, Saitama, Japan. RP Sato, K (reprint author), National Defense Medical College, Department of Basic Pathology, 359-8513 Tokorozawa, Japan. EM kimiya@ndmc.ac.jp CR Hori M, Matsuda T, Shibata A et al, 2015, Cancer incidence and incidence rates in Japan in 2009: a study of 32 population-based cancer registries for the Monitoring of Cancer Incidence in Japan, MCIJ, project. Jpn J Clin Oncol 45:884–891 Verkman AS, 2005, More than just water channels: unexpected cellular roles of aquaporins. J Cell Sci 118:3225–3232 King LS, Kozono D, Agre P, 2004, From structure to disease: the evolving tale of aquaporin biology. Nat Rev Mol Cell Biol 5:687– 698 Wang J, Feng L, Zhu Z et al, 2015, Aquaporins as diagnostic and therapeutic targets in cancer: how far we are? J Transl Med 13:96. , DOI 10.1186/s12967-015-0439-7 Verkman AS, Hara-Chikuma M, Papadopoulos MC, 2008, Aquaporins–new players in cancer biology. J Mol Med, Berl, 86: 523–529 Zhang D, Tan YJ, Qu F et al, 2012, Functions of water channels in male and female reproductive systems. Mol Aspects Med 33:676– 690 Papadopoulos MC, Saadoun S, 2015, Key roles of aquaporins in tumor biology. Biochim Biophys Acta 1848:2576–2583 Pan H, Sun CC, Zhou CYet al, 2008, Expression of aquaporin-1 in normal, hyperplasic, and carcinomatous endometria. Int J Gynecol Obstet 101:239–244 Marlar S, Jensen HH, Login FH et al, 2017, Aquaporin-3 in Cancer. Int J Mol Sci 18. , DOI 10.3390/ijms18102106 Liu YL, Matsuzaki T, Nakazawa T, et al, 2007, Expression of aquaporin 3, AQP3, in normal and neoplastic lung tissues. Hum Pathol 38:171–178 Rubenwolf PC, Denzinger S, Otto W, 2012, Aquaporin 3 protein expression in transitional cell carcinoma: a potential marker with regard to tumour progression and prognosis? Eur Urol 61:627–628 Rubenwolf PC, Otto W, Denzinger S et al, 2014, Expression of aquaporin water channels in human urothelial carcinoma: correlation of AQP3 expression with tumour grade and stage.World J Urol 32:991–997 Huang YT, Zhou J, Shi S et al, 2015, Identification of estrogen response element in aquaporin-3 gene that mediates estrogeninduced cell migration and invasion in estrogen receptor-positive breast cancer. Sci Rep 5:12484. , DOI 10.1038/srep12484 Chen J, Wang T, Zhou YC et al, 2014, Aquaporin 3 promotes epithelial-mesenchymal transition in gastric cancer. J Exp Clin Cancer Res 33:38. , DOI 10.1186/1756-9966-33-38 Sato K, Miyamoto M, Takano M et al, 2018, Different prognostic implications of aquaporin-1 and aquaporin-5 expression among different histological types of ovarian carcinoma. Pathol Oncol. , DOI 10.1007/s12253-018-0456-y, in press) Ji C, Cao C, Lu S et al, 2008, Curcumin attenuates EGF-induced AQP3 up-regulation and cell migration in human ovarian cancer cells. Cancer Chemother Pharmacol 62:857–865 He R, Han W, Hu Yet al, 2019, AQP2 is regulated by estradiol in human endometrium and is associated with spheroid attachment in vitro. Mol Med Rep 20:1306–1312 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2247 EP 2253 DI 10.1007/s12253-020-00813-3 PG 7 ER PT J AU Karaszi, K Vigh, R Mathe, M Fullar, A Olah, L Fule, T Papp, Z Kovalszky, I AF Karaszi, Katalin Vigh, Renata Mathe, Miklos Fullar, Alexandra Olah, Laszlone Fule, Tibor Papp, Zoltan Kovalszky, Ilona TI Aberrant Expression of Syndecan-1 in Cervical Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Syndecan-1; Cervical cancer; Survival analysis; Cancer-associated fibroblasts; Extracellularmatrix remodeling ID Syndecan-1; Cervical cancer; Survival analysis; Cancer-associated fibroblasts; Extracellularmatrix remodeling AB Syndecan-1, is a transmembrane heparan/chondroitin sulfate proteoglycan necessary for cell-cell and cell-matrix interactions. Its decreased level on the cell surface correlates with poor prognosis in several tumor types. Aberrant stromal localization of syndecan- 1 is also considered an unfavorable prognostic factor in various human malignancies. In the presented work the question was addressed if changes in syndecan-1 expression are related to the prognosis of cervical cancer. Immunohistochemistry for syndecan- 1 extracellular domain was performed on surgical specimens of primary cervical cancer. To follow the communication between tumor cells and stromal fibroblasts, their mono-and co-cultures were studied, detecting the expression of syndecan-1, smooth muscle actin, vimentin, and desmin. Immunohistochemistry of tumorous specimens revealed that while cell surface syndecan-1 expression was reduced on cancer cells, it appeared on the surface of tumor-associated fibroblasts. Until year 7, the cohort with high cell surface syndecan-1 expression had significantly longer survival. No difference in the same time-period could be detected when stromal syndecan-1 expression was analyzed. In vitro analysis revealed, that tumor cells can induce syndecan-1 expression on fibroblast, and fibroblasts showed that fibroblast-like cells are built by two cell types: (a) syndecan-1 positive, cytokeratin negative real fibroblasts, and (b) syndecan-1 and cytokeratin positive epithelial-mesenchymal transformed tumor cells. Syndecan-1 on the surface of cancer cells appears to be a positive prognostic marker. Although syndecan-1 positive fibroblasts promote tumor cell proliferation in vitro, we failed to detect their cancer promoting effect in vivo. C1 [Karaszi, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Vigh, Renata] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Mathe, Miklos] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Fullar, Alexandra] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Olah, Laszlone] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Fule, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. [Papp, Zoltan] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, H-1082 Budapest, Hungary. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, H-1085 Budapest, Hungary. RP Kovalszky, I (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, H-1085 Budapest, Hungary. EM kovalelastic@gmail.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Cancer J Clin 68(6):394–424. , DOI 10.3322/caac.21492 LaVigne AW, Triedman SA, Randall TC, Trimble EL, Viswanathan AN(2017, Cervical cancer in low andmiddle income countries: Addressing barriers to radiotherapy delivery. Gynecol Oncol Rep 22:16–20. , DOI 10.1016/j.gore.2017.08.004 Witsch E, Sela M, Yarden Y, 2010, Roles for growth factors in cancer progression. Physiology, Bethesda Md, 25(2):85–101. , DOI 10.1152/physiol.00045.2009 Erdogan B, Webb DJ, 2017, Cancer-associated fibroblasts modulate growth factor signaling and extracellular matrix remodeling to regulate tumor metastasis. Biochem Soc Trans 45(1):229–236. , DOI 10.1042/bst20160387 Liotta LA, 1986, Tumor invasion and metastases–role of the extracellular matrix: Rhoads Memorial Award lecture. Cancer Res 46(1):1–7 Liotta LA, 2016, Adhere, degrade, and move: the three-step model of invasion. Cancer Res 76(11):3115–3117. , DOI 10. 1158/0008-5472.Can-16-1297 Bernfield M, Kokenyesi R, Kato M, Hinkes MT, Spring J, Gallo RL, Lose EJ, 1992, Biology of the syndecans: a family of transmembrane heparan sulfate proteoglycans. Annu Rev Cell Biol 8: 365–393. , DOI 10.1146/annurev.cb.08.110192.002053 Kovalszky I, Nagy P, Szende B, Lapis K, Szalay F, Jeney A, Schaff Z, 1998, Experimental and human liver fibrogenesis. Scand J Gastroenterol Suppl 228:51–55 Rapraeger AC, 2000, Syndecan-regulated receptor signaling. J Cell Biol 149(5):995–998. , DOI 10.1083/jcb.149.5.995 Woods A, Couchman JR, 1998, Syndecans: synergistic activators of cell adhesion. Trends Cell Biol 8(5):189–192 Beauvais DM, Rapraeger AC, 2003, Syndecan-1-mediated cell spreading requires signaling by alphavbeta3 integrins in human breast carcinoma cells. Exp Cell Res 286(2):219–232. https://doi. org/10.1016/s0014-4827(03)00126-5 Dobra K, Nurminen M, Hjerpe A, 2003, Growth factors regulate the expression profile of their syndecan co-receptors and the differentiation of mesothelioma cells. Anticancer Res 23(3b):2435–2444 Jayson GC, Vives C, Paraskeva C, Schofield K, Coutts J, Fleetwood A, Gallagher JT, 1999, Coordinated modulation of the fibroblast growth factor dual receptor mechanism during transformation from human colon adenoma to carcinoma. Int J Cancer 82(2):298–304 Larrain J, Carey DJ, Brandan E, 1998, Syndecan-1 expression inhibits myoblast differentiation through a basic fibroblast growth factor-dependent mechanism. J Biol Chem 273(48):32288–32296. , DOI 10.1074/jbc.273.48.32288 Mundhenke C, Meyer K, Drew S, Friedl A, 2002, Heparan sulfate proteoglycans as regulators of fibroblast growth factor-2 receptor binding in breast carcinomas. Am J Pathol 160(1):185–194. https:// doi.org/10.1016/s0002-9440(10)64362-3 Soukka T, Pohjola J, Inki P, Happonen RP, 2000, Reduction of syndecan-1 expression is associatedwith dysplastic oral epithelium. J Oral Pathol Med 29(7):308–313 Timar J, Lapis K, Dudas J, Sebestyen A, Kopper L, Kovalszky I, 2002, Proteoglycans and tumor progression: Janus-faced molecules with contradictory functions in cancer. Sem Cancer Biol 12(3):173–186. , DOI 10.1016/s1044-579x(02)00021-4 Sobel G, Szabo I, Paska C, Kiss A, Kovalszky I, Kadar A, Paulin F, Schaff Z, 2005, Changes of cell adhesion and extracellular matrix, ECM, components in cervical intraepithelial neoplasia. Pathol Oncol Res 11(1):26–31 Kurokawa H,Matsumoto S, Murata T, Yamashita Y, Tomoyose T, ZhangM, Fukuyama H, Takahashi T, 2003, Immunohistochemical study of syndecan-1 down-regulation and the expression of p53 protein or Ki-67 antigen in oral leukoplakia with or without epithelial dysplasia. J Oral Pathol Med 32(9):513–521 Inki P, Stenback F, Grenman S, Jalkanen M, 1994, Immunohistochemical localization of syndecan-1 in normal and pathological human uterine cervix. J Pathol 172(4):349–355. , DOI 10.1002/path.1711720410 Wiksten JP, Lundin J, Nordling S, Lundin M, Kokkola A, von Boguslawski K, Haglund C, 2001, Epithelial and stromal syndecan-1 expression as predictor of outcome in patients with gastric cancer. Int J Cancer 95(1):1–6 Rintala M, Inki P, Klemi P, Jalkanen M, Grenman S, 1999, Association of syndecan-1 with tumor grade and histology in primary invasive cervical carcinoma. Gynecol Oncol 75(3):372–378., DOI , DOI 10.1006/gyno.1999.5595 Nakanishi K, Yoshioka N, Oka K, Hakura A, 1999, Reduction of syndecan-1 mRNA in cervical-carcinoma cells is involved with the 3’ untranslated region. Int J Cancer 80(4):527–532 Numa F, Hirabayashi K, Kawasaki K, Sakaguchi Y, Sugino N, Suehiro Y, Suminami Y, Hirakawa H, Umayahara K, Nawata S, Ogata H, Kato H, 2002, Syndecan-1 expression in cancer of the uterine cervix: association with lymph node metastasis. Int J Oncol 20(1):39–43 Kopper L, Sebestyen A, 2000, Syndecans and the lymphoid system. Leuk Lymphoma 38(3–4):271–281. , DOI 10.3109/ 10428190009087018 Larrain J, Cizmeci-Smith G, Troncoso V, Stahl RC, Carey DJ, Brandan E, 1997, Syndecan-1 expression is down-regulated during myoblast terminal differentiation. Modulation by growth factors and retinoic acid. J Biol Chem 272(29):18418–18424. https://doi. org/10.1074/jbc.272.29.18418 Mennerich D, Vogel A, Klaman I, Dahl E, Lichtner RB, Rosenthal A, Pohlenz HD, Thierauch KH, Sommer A, 2004, Shift of syndecan-1 expression from epithelial to stromal cells during progression of solid tumours. Eur J Cancer, Oxford, England: 1990, 40(9):1373–1382. , DOI 10.1016/j.ejca.2004.01.038 Stanley MJ, StanleyMW, Sanderson RD, Zera R, 1999, Syndecan- 1 expression is induced in the stroma of infiltrating breast carcinoma. Am J Clin Pathol 112(3):377–383. , DOI 10.1093/ ajcp/112.3.377 Mukunyadzi P, Liu K, Hanna EY, Suen JY, Fan CY, 2003, Induced expression of syndecan-1 in the stroma of head and neck squamous cell carcinoma. Mod Pathol 16(8):796–801. https://doi. org/10.1097/01.Mp.0000081731.17549.53 LeivonenM, Lundin J,Nordling S, von Boguslawski K,Haglund C, 2004, Prognostic value of syndecan-1 expression in breast cancer. Oncology 67(1):11–18. , DOI 10.1159/000080280 Ito Y, Yoshida H, Nakano K, Takamura Y, Miya A, Kobayashi K, Yokozawa T, Matsuzuka F, Matsuura N, Kuma K, Miyauchi A, 2003, Syndecan-1 expression in thyroid carcinoma: stromal expression followed by epithelial expression is significantly correlated with dedifferentiation. Histopathology 43(2):157–164 MatheM, Suba Z, Nemeth Z, Tatrai P, Fule T, Borgulya G, Barabas J, Kovalszky I, 2006, Stromal syndecan-1 expression is an adverse prognostic factor in oral carcinomas. Oral Oncol 42(5):493–500. , DOI 10.1016/j.oraloncology.2005.10.003 Maeda T, Alexander CM, Friedl A, 2004, Induction of syndecan-1 expression in stromal fibroblasts promotes proliferation of human breast cancer cells. Cancer Res 64(2):612–621 Szabo S, Xu Y, Romero R, Fule T, Karaszi K, Bhatti G, Varkonyi T, Varkonyi I, Krenacs T, Dong Z, Tarca AL, Chaiworapongsa T, Hassan SS, Papp Z, Kovalszky I, Than NG, 2013, Changes of placental syndecan-1 expression in preeclampsia and HELLP syndrome. Virchows Arch 463(3):445–458. , DOI 10.1007/ s00428-013-1426-0 Fule T, Csapo Z, MatheM, Tatrai P, Laszlo V, Papp Z, Kovalszky I, 2006, Prognostic significance of high-risk HPV status in advanced cervical cancers and pelvic lymph nodes. Gynecol Oncol 100(3): 570–578. , DOI 10.1016/j.ygyno.2005.09.019 Hazelbag S, Fleuren GJ, Baelde JJ, Schuuring E,KenterGG, Gorter A, 2001, Cytokine profile of cervical cancer cells. Gynecol Oncol 83(2):235–243. , DOI 10.1006/gyno.2001.6378 Baghy K, Dezso K, Laszlo V, Fullar A, Peterfia B, Paku S, Nagy P, Schaff Z, Iozzo RV, Kovalszky I, 2011, Ablation of the decorin gene enhances experimental hepatic fibrosis and impairs hepatic healing in mice. Lab Invest 91(3):439–451. , DOI 10. 1038/labinvest.2010.172 Fullar A, Dudas J, Olah L, Hollosi P, Papp Z, Sobel G, Karaszi K, Paku S, Baghy K, Kovalszky I, 2015, Remodeling of extracellular matrix by normal and tumor-associated fibroblasts promotes cervical cancer progression. BMC Cancer 15:256. , DOI 10. 1186/s12885-015-1272-3 Maeda T, Desouky J, Friedl A, 2006, Syndecan-1 expression by stromal fibroblasts promotes breast carcinoma growth in vivo and stimulates tumor angiogenesis. Oncogene 25(9):1408–1412. , DOI 10.1038/sj.onc.1209168 Yang N,Mosher R, Seo S, Beebe D, Friedl A, 2011, Syndecan-1 in breast cancer stroma fibroblasts regulates extracellular matrix fiber organization and carcinoma cell motility. Am J Pathol 178(1):325– 335. , DOI 10.1016/j.ajpath.2010.11.039 Boutin EL, Sanderson RD, Bernfield M, Cunha GR, 1991, Epithelial-mesenchymal interactions in uterus and vagina alter the expression of the cell surface proteoglycan, syndecan. Dev Biol 148(1):63–74. , DOI 10.1016/0012-1606(91)90317-v SamoszukM, Tan J, Chorn G, 2005, Clonogenic growth of human breast cancer cells co-cultured in direct contact with serumactivated fibroblasts. Breast Cancer Res 7(3):R274–R283. https:// doi.org/10.1186/bcr995 Mise I, Vucic M, 2018, Comparison of Syndecan-1 Immunohistochemical Expression in Lobular and Ductal Breast Carcinoma with Nodal Metastases. Anal Cell Pathol, Amsterdam, 2018:9432375. , DOI 10.1155/2018/ 9432375 Hashimoto Y, Skacel M, Adams JC, 2008, Association of loss of epithelial syndecan-1 with stage and local metastasis of colorectal adenocarcinomas: An immunohistochemical study of clinically annotated tumors. BMC Cancer 8(1):185. , DOI 10.1186/ 1471-2407-8-185 Contreras HR, Ledezma RA, Vergara J, Cifuentes F, Barra C, Cabello P, Gallegos I, Morales B, Huidobro C, Castellon EA, 2010, The expression of syndecan-1 and – 2 is associated with Gleason score and epithelial-mesenchymal transition markers, Ecadherin and beta-catenin, in prostate cancer. Urol Oncol 28(5): 534–540. , DOI 10.1016/j.urolonc.2009.03.018 Szatmari T, Otvos R, Hjerpe A, Dobra K, 2015, Syndecan-1 in Cancer: Implications for Cell Signaling, Differentiation, and Prognostication. Dis Markers 2015:796052. , DOI 10. 1155/2015/796052 Kim YI, Lee A, Lee BH, Kim SY, 2011, Prognostic significance of syndecan-1 expression in cervical cancers. J Gynecol Oncol 22(3): 161–167. , DOI 10.3802/jgo.2011.22.3.161 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2255 EP 2264 DI 10.1007/s12253-020-00816-0 PG 10 ER PT J AU Viola, L Londero, PA Bertozzi, S Orsaria, M Marzinotto, S Antoniazzi, F Renda, V Cinel, J Fruscalzo, A Lelle, JR Mariuzzi, L AF Viola, Luigi Londero, P Ambrogio Bertozzi, Serena Orsaria, Maria Marzinotto, Stefania Antoniazzi, Fulvio Renda, Valentina Cinel, Jacqueline Fruscalzo, Arrigo Lelle, J Ralph Mariuzzi, Laura TI Prognostic Role of Kruppel-Like Factors 5, 9, and 11 in Endometrial Endometrioid Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Kruppel-like factors; KLF; KLF5; KLF11; KLF9; Endometrioid endometrial cancer; Endometrial cancer ID Kruppel-like factors; KLF; KLF5; KLF11; KLF9; Endometrioid endometrial cancer; Endometrial cancer AB Background and Objective: Kruppel-like factors (KLFs) are transcription factors with the ability to mediate cross-talk with signaling pathways involved in cell proliferation control, apoptosis, migration, and differentiation. They also appear to influence steroid hormone signaling through transcriptional networks involving steroid hormone receptors and members of the nuclear receptor family of transcription factors. Our study aims to evaluate the potential prognostic role of KLF5, KLF9, and KLF11 in endometrial cancer, and their correlation with hormonal receptor status and cellular proliferation. Materials and Methods: Retrospective observational study on cases of endometrioid endometrial adenocarcinoma collected in the period January 2000–December 2011 at the University of Udine. Formalin-fixed, paraffin-embedded tissue samples were all submitted to tissue microarray immunohistochemical study. A survival analysis was performed. Results: One hundred forty seven patients were included in the study with a mean age at surgery of 65.6 years (±10.2). 80.3% of endometrial malignancies were classified as stage FIGO I (118/147). Radiation therapy and chemotherapy were administered in 62.3% (91/146) and 6.2% (9/145) of patients respectively. Five-year overall survival and disease-free survival resulted 85.4% (95% CI, 79.8–91.4%) and 79.4% (95% CI, 73.0–86.4%) respectively. A high Ki-67, cytoplasmatic KLF5 (HR 4.72, CI.95 1.61–13.89, p < 0.05), and nuclear KLF11 (HR 3.04, CI.95 0.99–9.36, p = 0.053) scores correlated with a shorter overall survival. In addition, a high nuclear KLF11 (HR 2.59, CI.95 1.13–5.95, p < 0.05) score correlated with a shorter disease-free survival. Conclusions: In patients affected by endometrioid endometrial carcinoma, higher staining levels of KLF5 and KLF11 correlated with a poorer prognosis. However, further studies are required in order to better clarify the role of KLFs in the natural history of endometrial cancer. C1 [Viola, Luigi] University of Campania “Luigi Vanvitelli”, Radiology Department, 80138 Naples, Italy. [Londero, P Ambrogio] University of Udine, DAME, Academic Hospital of Udine, Clinic of Obstetrics and Gynecology, Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy. [Bertozzi, Serena] Ennergi Research (Non-Profit Organization), 33050 Lestizza, Italy. [Orsaria, Maria] University of Udine, DAME, Academic Hospital of Udine, Institute of Pathology, 33100 Udine, Italy. [Marzinotto, Stefania] University of Udine, DAME, Academic Hospital of Udine, Institute of Pathology, 33100 Udine, Italy. [Antoniazzi, Fulvio] University of Udine, DAME, Academic Hospital of Udine, Institute of Pathology, 33100 Udine, Italy. [Renda, Valentina] University of Udine, DAME, Academic Hospital of Udine, Institute of Pathology, 33100 Udine, Italy. [Cinel, Jacqueline] University of Udine, Academic Hospital of Udine, Clinic of Surgery, 33100 Udine, Italy. [Fruscalzo, Arrigo] Christophorus-Kliniken, Clinic of Obstetrics and Gynecology, 48653 Coesfeld, Germany. [Lelle, J Ralph] University Hospital of Munster, Clinic of Obstetrics and Gynecology, Albert-Schweitzer-Campus 1, Gebaude: A1, 48149 Munster, Germany. [Mariuzzi, Laura] University of Udine, DAME, Academic Hospital of Udine, Institute of Pathology, 33100 Udine, Italy. RP Londero, PA (reprint author), University of Udine, DAME, Academic Hospital of Udine, Clinic of Obstetrics and Gynecology, 33100 Udine, Italy. EM ambrogio.londero@gmail.com CR Allen NE, Key TJ, Dossus L, Rinaldi S, Cust A, Lukanova A, Peeters PH, Onland-Moret NC, Lahmann PH, Berrino F, Panico S, Larranaga N, Pera G, Tormo MJ, Sanchez MJ, Ramon Quiros J, Ardanaz E, Tjonneland A, Olsen A, Chang-Claude J, Linseisen J, Schulz M, Boeing H, Lundin E, Palli D, Overvad K, Clavel- Chapelon F, Boutron-Ruault MC, Bingham S, Khaw KT, Bas Bueno-de-Mesquita H, Trichopoulou A, Trichopoulos D, Naska A, Tumino R, Riboli E, Kaaks R, 2008, Endogenous sex hormones and endometrial cancer risk in women in the European prospective investigation into cancer and nutrition, EPIC). Endocr Relat Cancer 15:485–497 Lacey JV, Yang H, Gaudet MM, Dunning A, Lissowska J, Sherman ME et al, 2011, Endometrial cancer and genetic variation in PTEN, PIK3CA,AKT1,MLH1, andMSH2 within a populationbased case-control study. Gynecol Oncol 120:167–173 Simmen RCM, Pabona JMP, Velarde MC, Simmons C, Rahal O, Simmen FA, 2010, The emerging role of Kruppel-like factors in endocrine-responsive cancers of female reproductive tissues. J Endocrinol 204:223–231 Simmen RCM, Zhang XL, Michel FJ, Min SH, Zhao G, Simmen FA, 2002, Molecular markers of endometrial epithelial cell mitogenesis mediated by the Sp/Kruppel-like factor BTEB1. DNA Cell Biol 21:115–128 Yoon HS, Chen X, Yang VW, 2003, Kruppel-like factor 4 mediates p53-dependent G1/S cell cycle arrest in response to DNA damage. J Biol Chem 278:2101–2105 Rowland BD, Bernards R, Peeper DS, 2005, The KLF4 tumour suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene. Nat Cell Biol 7:1074–1082 Wang Z, Spittau B, Behrendt M, Peters B, Krieglstein K, 2007, Human TIEG2/KLF11 induces oligodendroglial cell death by downregulation of Bcl-XL expression. J Neural Transm, Vienna, 114:867–875 Simmen FA, Su Y, Xiao R, Zeng Z, Simmen RCM, 2008, The Kruppel-like factor 9, KLF9, network in HEC-1-A endometrial carcinoma cells suggests the carcinogenic potential of dys-regulated KLF9 expression. Reprod Biol Endocrinol 6:41 Mutter GL, Baak JP, Fitzgerald JT, Gray R, Neuberg D, Kust GA et al, 2001, Global expression changes of constitutive and hormonally regulated genes during endometrial neoplastic transformation. Gynecol Oncol 83:177–185 Wang G, Li X, Tian W, Wang Y, Wu D, Sun Z, Zhao E, 2015, PromoterDNAmethylation is associated with KLF11 expression in epithelial ovarian cancer. Genes Chromosomes Cancer 54:453–462 Buck A, Buchholz M,Wagner M, Adler G, Gress T, Ellenrieder V, 2006, The tumor suppressor KLF11 mediates a novel mechanism in transforming growth factor beta-induced growth inhibition that is inactivated in pancreatic cancer. Mol Cancer Res 4:861–872 Londero AP, Orsaria M, Tell G, Marzinotto S, Capodicasa V, Poletto M, Vascotto C, Sacco C, Mariuzzi L, 2014, Expression and prognostic significance of APE1/Ref-1 and NPM1 proteins in high-grade ovarian serous cancer. Am J Clin Pathol 141:404–414 Velarde MC, Geng Y, Eason RR, Simmen FA, Simmen RCM, 2005, Null mutation of Kruppel-like factor9/basic transcription element binding protein-1 alters peri-implantation uterine development in mice. Biol Reprod 73:472–481 Zhang D, Zhang XL, Michel FJ, Blum JL, Simmen FA, Simmen RCM, 2002, Direct interaction of the Kruppel-like family, KLF, member, BTEB1, and PR mediates progesterone-responsive gene expression in endometrial epithelial cells. Endocrinology 143:62– 73 Pabona JMP, Velarde MC, Zeng Z, Simmen FA, Simmen RCM, 2009, Nuclear receptor co-regulator Kruppel-like factor 9 and prohibitin 2 expression in estrogen-induced epithelial cell proliferation in the mouse uterus. J Endocrinol 200:63–73 Velarde MC, Zeng Z, McQuown JR, Simmen FA, Simmen RCM, 2007, Kruppel-like factor 9 is a negative regulator of liganddependent estrogen receptor alpha signaling in Ishikawa endometrial adenocarcinoma cells. Mol Endocrinol 21:2988–3001 Pearson R, Fleetwood J, Eaton S, Crossley M, Bao S, 2008, Kruppel-like transcription factors: a functional family. Int J Biochem Cell Biol 40:1996–2001 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2265 EP 2272 DI 10.1007/s12253-020-00817-z PG 8 ER PT J AU Tanaka, T Miyamoto, Sh Terada, Sh Kogata, Y Fujiwara, S Tanaka, Y Taniguchi, K Komura, K Yamamoto, K Yamada, T Ohmichi, M AF Tanaka, Tomohito Miyamoto, Shunsuke Terada, Shinichi Kogata, Yuhei Fujiwara, Satoe Tanaka, Yoshimichi Taniguchi, Kohei Komura, Kazumasa Yamamoto, Kazuhiro Yamada, Takashi Ohmichi, Masahide TI The Diagnostic Accuracy of an Intraoperative Frozen Section Analysis and Imprint Cytology of Sentinel Node Biopsy Specimens from Patients with Uterine Cervical and Endometrial Cancer: a Retrospective Observational Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial cancer; Cervical cancer; Sentinel node biopsy; Frozen section; Imprint cytology ID Endometrial cancer; Cervical cancer; Sentinel node biopsy; Frozen section; Imprint cytology AB Sentinel node biopsy (SNB) may be a decision-making tool for function preservation surgery, including radical trachelectomy and ovary preservation in the treatment of cervical and endometrial cancer. The intraoperative diagnosis is important for guiding treatment decisions for patients with these conditions. Three hundred seventy-one patients with cervical and endometrial cancer received SNB with an intraoperative frozen section analysis and imprint cytology. The sentinel node was cut in half, parallel to the longest axis, to obtain the maximum section area. After performing imprint cytology, one half was used to create a frozen section. The specimen was cut at 2-mm intervals into 5-μm-thick sections, which were subjected to hematoxylin and eosin staining. The diagnostic accuracy of intraoperative frozen section analyses and imprint cytology was compared to the final pathological diagnosis. Among 951 detected sentinel nodes, 51 nodes were found to be positive in the final pathological diagnosis. The sensitivity of a frozen section analysis, imprint cytology and the combination of the two modalities was 76.5%, 72.6%, and 92.2%, respectively. The specificity of a frozen section analysis and imprint cytology was 100%. The negative predictive value of a frozen section analysis and imprint cytology was 98.7% and 98.5%, respectively. In these settings, the accuracy of the frozen section analysis and imprint cytology in the evaluation of SNB specimens was considered acceptable; however, the sensitivity of the combined approach was higher in comparison to when a frozen section analysis or imprint cytology was performed alone. C1 [Tanaka, Tomohito] Osaka Medical College, Department of Obstetrics and Gynecology, 2-7, Daigaku-machi, 569-8686 Takatsuki, Osaka, Japan. [Miyamoto, Shunsuke] Osaka Medical College, Department of Obstetrics and Gynecology, 2-7, Daigaku-machi, 569-8686 Takatsuki, Osaka, Japan. [Terada, Shinichi] Osaka Medical College, Department of Obstetrics and Gynecology, 2-7, Daigaku-machi, 569-8686 Takatsuki, Osaka, Japan. [Kogata, Yuhei] Osaka Medical College, Department of Obstetrics and Gynecology, 2-7, Daigaku-machi, 569-8686 Takatsuki, Osaka, Japan. [Fujiwara, Satoe] Osaka Medical College, Department of Obstetrics and Gynecology, 2-7, Daigaku-machi, 569-8686 Takatsuki, Osaka, Japan. [Tanaka, Yoshimichi] Osaka Medical College, Department of Obstetrics and Gynecology, 2-7, Daigaku-machi, 569-8686 Takatsuki, Osaka, Japan. [Taniguchi, Kohei] Osaka Medical College, Translational Research ProgramTakatsuki, Osaka, Japan. [Komura, Kazumasa] Osaka Medical College, Translational Research ProgramTakatsuki, Osaka, Japan. [Yamamoto, Kazuhiro] Osaka Medical College, RadiologyTakatsuki, Osaka, Japan. [Yamada, Takashi] Osaka Medical College, PathologyTakatsuki, Osaka, Japan. [Ohmichi, Masahide] Osaka Medical College, Department of Obstetrics and Gynecology, 2-7, Daigaku-machi, 569-8686 Takatsuki, Osaka, Japan. RP Tanaka, T (reprint author), Osaka Medical College, Department of Obstetrics and Gynecology, 569-8686 Takatsuki, Japan. EM gyn123@osaka-med.ac.jp CR Tanaka T, Terai Y, Fujiwara S, Tanaka Y, Sasaki H, Tsunetoh S, Yamamoto K, Yamada T, Ohmichi M, 2017, The detection of sentinel lymph nodes in laparoscopic surgery can eliminate systemic lymphadenectomy for patients with early stage endometrial cancer. Int J Clin Oncol 23:305–313. , DOI 10.1007/s10147- 017-1196-9 Tanaka T, Terai Y, Ashihara K, Tsunetoh S, Akagi H, Yamada T, Ohmichi M, 2017, The detection of sentinel lymph nodes in laparoscopic surgery for uterine cervical cancer using 99m-technetiumtin colloid, indocyanine green, and blue dye. J Gynecol Oncol 28(2):e13. , DOI 10.3802/jgo.2017.28.e13 Tanaka T, Sasaki S, Tsuchihashi H, Terai Y, Yamamoto K, Yamada T, OhmichiM(2018)Which is better for predicting pelvic lymph node metastases in patients with cervical cancer: Fluorodeoxyglucose-positron emission tomography/computed tomography or a sentinel node biopsy? A retrospective observational study. Medicine 97(16):e0410. , DOI 10.1097/md. 0000000000010410 Tanaka T, Terai Y, Yamamoto K, Yamada T, Ohmichi M, 2018, The diagnostic accuracy of fluorodeoxyglucose-positron emission tomography/computed tomography and sentinel node biopsy in the prediction of pelvic lymph node metastasis in patients with endometrial cancer: a retrospective observational study. Medicine 97(38):e12522. , DOI 10.1097/md.0000000000012522 Abu-Rustum NR, Alektiar K, Iasonos A, Lev G, Sonoda Y, Aghajanian C, Chi DS, Barakat RR, 2006, The incidence of symptomatic lower-extremity lymphedema following treatment of uterine corpus malignancies: a 12-year experience at Memorial Sloan-Kettering Cancer Center. Gynecol Oncol 103(2):714–718. https:// doi.org/10.1016/j.ygyno.2006.03.055 Eiriksson LR, Covens A, 2012, Sentinel lymph node mapping in cervical cancer: the future? BJOG : an international journal of obstetrics and gynaecology 119(2):129–133. , DOI 10.1111/ j.1471-0528.2011.03134.x van Deurzen CH, de Boer M, Monninkhof EM, Bult P, van der Wall E, Tjan-Heijnen VC, van Diest PJ, 2008, Non-sentinel lymph node metastases associated with isolated breast cancer cells in the sentinel node. J Natl Cancer Inst 100(22):1574–1580. https://doi. org/10.1093/jnci/djn343 Perez-Sanchez VM, Vela-Chavez TA, Villarreal-Colin P, Bargallo- Rocha E, Ramirez-Ugalde MT, Munoz-Gonzalez D, Zeichner- Gancz I, 2010, Intraoperative touch imprint cytology of sentinel lymph nodes in breast cancer: experience at a tertiary care center in Mexico. Med Oncol 27(2):233–236. , DOI 10.1007/ s12032-009-9197-9 Contractor K, Gohel M, Al-Salami E, Kaur K, Aqel N, Nigar E, Burke M, Singhal H, 2009, Intra-operative imprint cytology for assessing the sentinel node in breast cancer: results of its routine use over 8 years. Eur J Surg Oncol 35(1):16–20. , DOI 10. 1016/j.ejso.2008.04.005 Tew K, Irwig L, Matthews A, Crowe P, Macaskill P, 2005, Metaanalysis of sentinel node imprint cytology in breast cancer. Br J Surg 92(9):1068–1080. , DOI 10.1002/bjs.5139 Motomura K, Nagumo S, Komoike Y, Koyama H, Inaji H, 2007, Intraoperative imprint cytology for the diagnosis of sentinel node metastases in breast cancer. Breast cancer, Tokyo, Japan, 14(4): 350–353 Sonoda K, Yahata H, Okugawa K, Kaneki E, Ohgami T, Yasunaga M, Baba S,OdaY,Honda H,Kato K(2018, Value of intraoperative cytological and pathological sentinel lymph node diagnosis in fertility-sparing Trachelectomy for early-stage cervical Cancer. Oncology 94(2):92–98. , DOI 10.1159/000484049 Barranger E, Cortez A, Uzan S, Callard P, Darai E, 2004, Value of intraoperative imprint cytology of sentinel nodes in patients with cervical cancer. Gynecol Oncol 94(1):175–180. , DOI 10. 1016/j.ygyno.2004.04.015 Darai E, Lavoue V, Rouzier R, Coutant C, Barranger E, Bats AS, 2007, Contribution of the sentinel node procedure to tailoring the radicality of hysterectomy for cervical cancer. Gynecol Oncol 106(1):251–256. , DOI 10.1016/j.ygyno.2007.03.034 Martinez A, Mery E, Filleron T, Boileau L, Ferron G, Querleu D, 2013, Accuracy of intraoperative pathological examination of SLN in cervical cancer. Gynecol Oncol 130(3):525–529. https:// doi.org/10.1016/j.ygyno.2013.01.023 Fader AN, Edwards RP, Cost M, Kanbour-Shakir A, Kelley JL, Schwartz B, Sukumvanich P, Comerci J, Sumkin J, Elishaev E, Rohan LC, 2008, Sentinel lymph node biopsy in early-stage cervical cancer: utility of intraoperative versus postoperative assessment. Gynecol Oncol 111(1):13–17. , DOI 10.1016/j. ygyno.2008.06.009 Ballester M, Dubernard G, Bats AS, Heitz D, Mathevet P, Marret H, Querleu D, Golfier F, Leblanc E, Rouzier R, Darai E, 2012, Comparison of diagnostic accuracy of frozen section with imprint cytology for intraoperative examination of sentinel lymph node in early-stage endometrial cancer: results of Senti-Endo study. Ann Surg Oncol 19(11):3515–3521. , DOI 10.1245/s10434- 012-2390-7 Roy M, Bouchard-Fortier G, Popa I, Gregoire J, Renaud MC, Tetu B, PlanteM(2011, Value of sentinel node mapping in cancer of the cervix. Gynecol Oncol 122(2):269–274. , DOI 10.1016/j. ygyno.2011.04.002 Slama J, Dundr P, Dusek L, Cibula D, 2013, High false negative rate of frozen section examination of sentinel lymph nodes in patients with cervical cancer. Gynecol Oncol 129(2):384–388. https:// doi.org/10.1016/j.ygyno.2013.02.001 Niikura H, Okamoto S, Otsuki T, Yoshinaga K, Utsunomiya H, Nagase S, Takano T, Ito K, Watanabe M, Yaegashi N, 2012, Prospective study of sentinel lymph node biopsy without further pelvic lymphadenectomy in patients with sentinel lymph nodenegative cervical cancer. Int J Gynecol Cancer 22(7):1244–1250. , DOI 10.1097/IGC.0b013e318263f06a Gortzak-Uzan L, Jimenez W, Nofech-Mozes S, Ismiil N, Khalifa MA, Dube V, Rosen B, Murphy J, Laframboise S, Covens A, 2010, Sentinel lymph node biopsy vs. pelvic lymphadenectomy in early stage cervical cancer: is it time to change the gold standard? Gynecol Oncol 116(1):28–32. , DOI 10.1016/j.ygyno. 2009.10.049 Shiino S, Yoshida M, Jimbo K, Asaga S, Takayama S, Maeshima A, Tsuda H, Kinoshita T, Hiraoka N, 2019, Two rare cases of endosalpingiosis in the axillary sentinel lymph nodes: evaluation of immunohistochemical staining and one-step nucleic acid amplification, OSNA, assay in patients with breast cancer. Virchows Archiv : an international journal of pathology 474(5):633–638. , DOI 10.1007/s00428-019-02521-z Tohya T, Nakamura M, Fukumatsu Y, Katabuchi H, Matsuura K, Itoh M, Okamura H, 1991, Endosalpingosis in the pelvic peritoneum and pelvic lymph nodes. Nihon Sanka Fujinka Gakkai zasshi 43(7):756–762 Horn LC, Bilek K, 1995, Frequency and histogenesis of pelvic retroperitoneal lymph node inclusions of the female genital tract. An immunohistochemical study of 34 cases. Pathol Res Pract 191(10):991–996. , DOI 10.1016/s0344-0338(11)80597-2 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2273 EP 2279 DI 10.1007/s12253-020-00822-2 PG 7 ER PT J AU Benati, M Montagnana, M Danese, E Mazzon, M Paviati, E Garzon, S Lagana, SA Casarin, J Giudici, S Raffaelli, R Ghezzi, F Franchi, M Lippi, G AF Benati, Marco Montagnana, Martina Danese, Elisa Mazzon, Martina Paviati, Elisa Garzon, Simone Lagana, Simone Antonio Casarin, Jvan Giudici, Silvia Raffaelli, Ricciarda Ghezzi, Fabio Franchi, Massimo Lippi, Giuseppe TI Aberrant Telomere Length in Circulating Cell-Free DNA as Possible Blood Biomarker with High Diagnostic Performance in Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial cancer; Endometrial hyperplasia; cfDNA; Telomere length; Diagnosis; Biomarkers ID Endometrial cancer; Endometrial hyperplasia; cfDNA; Telomere length; Diagnosis; Biomarkers AB To investigate the diagnostic performance of relative telomere length (RTL) in cell-free DNA (cfDNA) for endometrioid endometrial cancer (EC). We measured RTL in cfDNA of 40 EC patients (65 ± 12 years) and 31 healthy controls (HC) (63 ± 13 years), excluding in both groups other oncologic and severe non-oncologic diseases to limit confounders. Circulating cfDNA was extracted from serum using the QIAamp DNA Blood Mini kit (Qiagen, Hilden, Germany). After the quantitative real-time polymerase chain reaction, telomere repeat copy number to single-gene copy number ratio was calculated. RTL in cfDNA was found to be significantly lower in EC patients than inHC (p < 0.0001). The diagnostic performance of cfDNA RTLwas estimated with receiver operating characteristics (ROC) curve analysis, which showed a diagnostic accuracy for EC of 0.87 (95% CI: 0.79– 0.95, p < 0.0001). The cutoff cfDNA RTL value of 2.505 (T/S copy ratio) reported a sensitivity of 80.0%(95%CI: 64.35–90.95) and a specificity of 80.65%(95%CI: 62.53–92.55). Significant differences of RTL among EC stages or grades (p = 0.85 and p = 0.89, respectively) were not observed. Our results suggest that cfDNA RTL analysis may be a diagnostic tool for EC detection since the early stage, whilst its diagnostic performance seems unsatisfactory for cancer progression, staging, and grading. However, further studies are needed to confirm these preliminary findings. In particular, future investigations should focus on high-risk patients (such as those with atypical endometrial hyperplasia) that may benefit from this tool, because TL shortening is not specific for EC and is influenced by other oncologic and non-oncologic diseases. C1 [Benati, Marco] University of Verona, Section of Clinical BiochemistryVerona, Italy. [Montagnana, Martina] University of Verona, Section of Clinical BiochemistryVerona, Italy. [Danese, Elisa] University of Verona, Section of Clinical BiochemistryVerona, Italy. [Mazzon, Martina] AOUI Verona, Laboratory of Clinical Chemistry and HematologyVerona, Italy. [Paviati, Elisa] University of Verona, Section of Clinical BiochemistryVerona, Italy. [Garzon, Simone] University of Insubria, “Filippo Del Ponte” Hospital, Department of Obstetrics and Gynecology, Piazza Biroldi 1, 21100 Varese, Italy. [Lagana, Simone Antonio] University of Insubria, “Filippo Del Ponte” Hospital, Department of Obstetrics and Gynecology, Piazza Biroldi 1, 21100 Varese, Italy. [Casarin, Jvan] University of Insubria, “Filippo Del Ponte” Hospital, Department of Obstetrics and Gynecology, Piazza Biroldi 1, 21100 Varese, Italy. [Giudici, Silvia] University of Verona, AOUI Verona, Department of Obstetrics and GynecologyVerona, Italy. [Raffaelli, Ricciarda] University of Verona, AOUI Verona, Department of Obstetrics and GynecologyVerona, Italy. [Ghezzi, Fabio] University of Insubria, “Filippo Del Ponte” Hospital, Department of Obstetrics and Gynecology, Piazza Biroldi 1, 21100 Varese, Italy. [Franchi, Massimo] University of Verona, AOUI Verona, Department of Obstetrics and GynecologyVerona, Italy. [Lippi, Giuseppe] University of Verona, Section of Clinical BiochemistryVerona, Italy. RP Garzon, S (reprint author), University of Insubria, “Filippo Del Ponte” Hospital, Department of Obstetrics and Gynecology, 21100 Varese, Italy. EM simone.garzon@yahoo.it CR Colombo N, Creutzberg C, Amant F, Bosse T, Gonzalez-Martin A, Ledermann J, Marth C, Nout R, Querleu D, Mirza MR, Sessa C, ESMO-ESGO-ESTRO Endometrial Consensus Conference Working Group, 2016, ESMO-ESGO-ESTRO consensus conference on endometrial cancer: diagnosis, treatment and follow-up. Int J Gynecol Cancer 26:2–30. , DOI 10.1097/IGC. 0000000000000609 Yeramian A, Moreno-Bueno G, Dolcet X, Catasus L, Abal M, Colas E, Reventos J, Palacios J, Prat J, Matias-Guiu X, 2013, Endometrial carcinoma: molecular alterations involved in tumor development and progression. Oncogene 32:403–413. https://doi. org/10.1038/onc.2012.76 Montagnana M, Lippi G, Ruzzenente O, Bresciani V, Danese E, Scevarolli S, Salvagno GL, Giudici S, FranchiM, Guidi GC, 2009, The utility of serum human epididymis protein 4, HE4, in patients with a pelvic mass. J Clin Lab Anal 23:331–335. , DOI 10. 1002/jcla.20340 Montagnana M, Benati M, Danese E, Giudici S, Perfranceschi M, Ruzzenenete O, Salvagno GL, Bassi A, Gelati M, Paviati E, Guidi GC, Franchi M, Lippi G, 2017, AberrantMicroRNA expression in patients with endometrial cancer. Int J Gynecol Cancer 27:459– 466. , DOI 10.1097/IGC.0000000000000913 Benati M, Montagnana M, Danese E, Paviati E, Giudici S, Franchi M, Lippi G, 2017, Evaluation of mir-203 expression levels and DNA promoter methylation status in serum of patients with endometrial cancer. Clin Lab 63:1675–1681. , DOI 10.7754/ Clin.Lab.2017.170421 Benati M, Montagnana M, Danese E, Paviati E, Giudici S, Ruzzenente O, FranchiM, Lippi G, 2018, The clinical significance of DJ-1 and HE4 in patients with endometrial cancer. J Clin Lab Anal 32:e22223. , DOI 10.1002/jcla.22223 Casarin J, Bogani G, Serati M, Pinelli C, Lagana AS, Garzon S, Raspagliesi F, Ghezzi F, 2019, Presence of glandular cells at the preoperative cervical cytology and local recurrence in endometrial cancer. Int J Gynecol Pathol. , DOI 10.1097/PGP. 0000000000000642 Blackburn EH, 1991, Structure and function of telomeres. Nature 350:569–573. , DOI 10.1038/350569a0 Moyzis RK, Buckingham JM, Cram LS, Dani M, Deaven LL, JonesMD,Meyne J, Ratliff RL,Wu JR, 1988, A highly conserved repetitiveDNA sequence,, TTAGGG, n, present at the telomeres of human chromosomes. Proc Natl Acad Sci U S A 85:6622–6626. , DOI 10.1073/pnas.85.18.6622 McClintock B, 1941, The stability of broken ends of chromosomes in Zea Mays. Genetics 26:234–282 Blasco MA, 2005, Telomeres and human disease: ageing, cancer and beyond. Nat Rev Genet 6:611–622. , DOI 10.1038/ nrg1656 Lo AW, Sabatier L, Fouladi B, Pottier G, Ricoul M, Mumane JP, 2002, DNA amplification by breakage/fusion/bridge cycles initiated by spontaneous telomere loss in a human cancer cell line. Neoplasia 4:531–538. , DOI 10.1038/sj.neo.7900267 Maser RS, 2002, Connecting chromosomes, crisis, and cancer. Science 297:565–569. , DOI 10.1126/science.297.5581. 565 Bunting SF,Nussenzweig A, 2013, End-joining, translocations and cancer. Nat Rev Cancer 13:443–454. , DOI 10.1038/ nrc3537 Counter CM,HirteHW, Bacchetti S,Harley CB, 1994, Telomerase activity in human ovarian carcinoma. Proc Natl Acad Sci U S A 91: 2900–2904. , DOI 10.1073/pnas.91.8.2900 Hastie ND, Dempster M, Dunlop MG, Thompson AM, Green DK, Allshire RC, 1990, Telomere reduction in human colorectal carcinoma and with ageing. Nature 346:866–868. , DOI 10. 1038/346866a0 Sieglova Z, Zilovcova S, Cermak J, Ríhova H, Brezinova D, Dvorakova R, Markova M, Maaloufova J, Sajdova J, Brezinova J, Zemanova Z,Michalova K, 2004, Dynamics of telomere erosion and its association with genome instability in myelodysplastic syndromes, MDS, and acute myelogenous leukemia arising from MDS: a marker of disease prognosis? Leuk Res 28:1013–1021. , DOI 10.1016/j.leukres.2003.11.020 McGrath M, Wong JY, Michaud D, Hunter DJ, De Vivo I, 2007, Telomere length, cigarette smoking, and bladder cancer risk in men and women. Cancer Epidemiol Biomark Prev 16:815–819. https:// doi.org/10.1158/1055-9965.EPI-06-0961 Shao L,Wood CG, ZhangD, Tannir NM, Matin S, Dinney CP,Wu X, 2007, Telomere dysfunction in peripheral lymphocytes as a potential predisposition factor for renal cancer. J Urol 178:1492– 1496. , DOI 10.1016/j.juro.2007.05.112 Wu X, Amos CI, Zhu Y, Zhao H, Grossman BH, Shay JW, Luo S, Hong WK, Spitz MR, 2003, Telomere dysfunction: a potential cancer predisposition factor. J Natl Cancer Inst 95:1211–1218. , DOI 10.1093/jnci/djg011 Jang JS, Choi YY, Lee WK, Choi JE, Cha SI, Kim YJ, Kim CH, Kam S, Jung TH, Park JY, 2008, Telomere length and the risk of lung cancer. Cancer Sci 99:1385–1389. , DOI 10.1111/j. 1349-7006.2008.00831.x Willeit P, Willeit J, Mayr A, Weger S, Oberhollenzer F, Brandstatter A, Kronenberg F, Kiechl S, 2010, Telomere length and risk of incident cancer and cancer mortality. JAMA304:69–75. , DOI 10.1001/jama.2010.897 Weischer M, Nordestgaard BG, Cawthon RM, Freiberg JJ, Tybjaerg-Hansen A, Bojesen SE, 2013, Short telomere length, cancer survival, and cancer risk in 47102 individuals. J Natl Cancer Inst 105:459–468. , DOI 10.1093/jnci/djt016 Rode L, Nordestgaard BG, Bojesen SE, 2015, Peripheral blood leukocyte telomere length and mortality among 64 637 individuals from the general population. J Natl Cancer Inst 107:1–8. https://doi. org/10.1093/jnci/djv074 Wan S, Hann HW, Myers RE, Fu X, Hann RS, Kim SH, Tang H, Xing J, Yang H, 2012, Telomere length in circulating serum DNA as a novel non-invasive biomarker for cirrhosis: a nested casecontrol analysis. Liver Int 32:1233–1241. , DOI 10.1111/ j.1478-3231.2012.02801.x Wu X, Tanaka H, 2015, Aberrant reduction of telomere repetitive sequences in plasma cell-free DNA for early breast cancer detection. Oncotarget 6:29795–29807. , DOI 10.18632/ oncotarget.5083 Wan S, Hann HW, Ye Z, Hann RS, Lai Y, Wang C, Li L, Myers RE, Li B, Xing J, Yang H, 2017, Prospective and longitudinal evaluations of telomere length of circulating DNA as a risk predictor of hepatocellular carcinoma in HBV patients. Carcinogenesis 38:439–446. , DOI 10.1093/carcin/bgx021 FranchiM, Garzon S, Zorzato PC, Lagana AS, Casarin J, Locantore L, Raffaelli R, Ghezzi F, 2019, PET-CT scan in the preoperative workup of early stage intermediate- and high-risk endometrial cancer. Minim Invasive Ther Allied Technol. , DOI 10.1080/ 13645706.2019.1624576 Creasman W, 2009, Revised FIGO staging for carcinoma of the endometrium. Int J Gynecol Obstet 105:109–109. , DOI 10.1016/j.ijgo.2009.02.010 Cawthon RM, 2002, Telomere measurement by quantitative PCR. Nucleic Acids Res 30:e47–e51 Montagnana M, Lippi G, 2017, Cancer diagnostics: current concepts and future perspectives. Ann Transl Med 5:268–271. https:// doi.org/10.21037/atm.2017.06.20 Cerne D, Bajalo JL, 2014, Cell-free nucleic acids as a non-invasive route for investigating atherosclerosis. Curr Pharm Des 20:5004– 5009. , DOI 10.2174/1381612819666131206110317 Siravegna G, Marsoni S, Siena S, Bardelli A, 2017, Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol 14:531–548. , DOI 10.1038/nrclinonc.2017.14 Swarup V, Rajeswari MR, 2007, Circulating, cell-free, nucleic acids - a promising, non-invasive tool for early detection of several human diseases. FEBS Lett 581:795–799. , DOI 10.1016/j. febslet.2007.01.051 Danese E, Montagnana M, Fava C, Guidi G, 2014, Circulating nucleic acids and hemostasis: biological basis behind their relationship and technical issues in assessment. Semin Thromb Hemost 40: 766–773 , DOI 10.1055/s-0034-1387923 Schwarzenbach H, Muller V, Stahmann N, Pantel K, 2004, Detection and characterization of circulating microsatellite-DNA in blood of patients with breast cancer. Ann N Y Acad Sci 1022: 25–32. , DOI 10.1196/annals.1318.005 Schwarzenbach H, Eichelser C, Kropidlowski J, Janni W, Rack B, Pantel K, 2012, Loss of heterozygosity at tumor suppressor genes detectable on fractionated circulating cell-free tumor DNA as indicator of breast cancer progression. Clin Cancer Res 18:5719–5730. , DOI 10.1158/1078-0432.CCR-12-0142 Phallen J, Sausen M, Adleff V, Leal A, Hruban C, White J, Anagnostou V, Fiksel J, Cristiano S, Papp E, Speir S, Reinert T, Orntoft MW, Woodward BD, Murphy D, Parpart-Li S, Riley D, Nesselbush M, Sengamalay N, Georgiadis A, Li QK,MadsenMR, Mortensen FV, Huiskens J, Punt C, van Grieken N, Fijneman R, Meijer G, Husain H, Scharpf RB, Diaz Jr LA, Jones S, Angiuoli S, Orntoft T, Nielsen HJ, Andersen CL, Velculescu VE, 2017, Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl med 9:eaan2415. , DOI 10.1126/scitranslmed. aan2415 Medford AJ, Gillani RN, Park BH, 2018, Detection of cancer DNA in early stage and metastatic breast cancer patients. Methods Mol Biol 1768:209–227. , DOI 10.1007/978-1-4939-7778-9_ 13 Schwarzenbach H, Hoon DS, Pantel K, 2011, Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer 11:426– 437. , DOI 10.1038/nrc3066 Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R, 2001, DNA fragments in the blood plasma of cancer patients: Quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 61:1659–1665 Heitzer E, Ulz P, Geigl JB, 2015, Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem 61:112–123. , DOI 10.1373/clinchem.2014.222679 Perakis S, Auer M, Belic J, Heitzer E, 2017, Advances in circulating tumor DNA analysis. Adv Clin Chem 80:73–153. https://doi. org/10.1016/bs.acc.2016.11.005 Ulz P, Heitzer E, Geigl JB, SpeicherMR, 2017, Patient monitoring through liquid biopsies using circulating tumor DNA. Int J Cancer 141:887–896. , DOI 10.1002/ijc.30759 Marzese DM, Hirose H, Hoon DS, 2013, Diagnostic and prognostic value of circulating tumor-related DNA in cancer patients. Expert Rev Mol Diagn 13:827–844. , DOI 10.1586/ 14737159.2013.845088 Chan KC, Jiang P, Zheng YW, Liao GJ, Sun H, Wong J, Siu SS, Chan WC, Chan SL, Chan AT, Lai PB, Chiu RW, Lo YM, 2013, Cancer genome scanning in plasma: detection of tumor-associated copy number aberrations, single-nucleotide variants, and Tumoral heterogeneity by massively parallel sequencing. Clin Chem 59: 211–224. , DOI 10.1373/clinchem.2012.196014 The Cancer Genome Atlas Research Network, 2013, Integrated genomic characterization of endometrial carcinoma. Nature 497: 67–73 , DOI 10.1038/nature12113 MacKay HJ, Levine DA, Bae-Jump VL, Bell DW, McAlpine JN, Santin A, Fleming GF, Mutch DG, Nephew KP, Wentzensen N, Goodfellow PJ, Dorigo O, Nijman HW, Broaddus R, Kohn EC, 2017, Moving forward with actionable therapeutic targets and opportunities in endometrial cancer: NCI clinical trials planning meeting report on identifying key genes and molecular pathways for targeted endometrial cancer trials. Oncotarget 8:84579–84594 , DOI 10.18632/oncotarget.19961 Martin-Ruiz CM, Baird D, Roger L, Boukamp P, Krunic D, Cawthon R, Dokter MM, van der Harst P, Bekaert S, de Meyer T, Roos G, Svenson U, Codd V, Samani NJ, McGlynn L, Shiels PG, Pooley KA, Dunning AM, Cooper R, Wong A, Kingston A, von Zglinicki T, 2015, Reproducibility of telomere length assessment: an international collaborative study. Int J Epidemiol 44: 1673–1683. , DOI 10.1093/ije/dyu191 Cicchillitti L, Corrado G, De Angeli M, Mancini E, Baiocco E, Patrizi L, Zampa A, Merola R, Martayan A, Conti L, Piaggio G, Vizza E, 2017, Circulating cell-free DNA content as blood based biomarker in endometrial cancer. Oncotarget 8:115230–115243. , DOI 10.18632/oncotarget.23247 Vizza E, Corrado G, De Angeli M, Carosi M, Mancini E, Baiocco E, Chiofalo B, Patrizi L, Zampa A, Piaggio G, Cicchillitti L, 2018, Serum DNA integrity index as a potential molecular biomarker in endometrial cancer. J Exp Clin Cancer Res 37:16–24. https://doi. org/10.1186/s13046-018-0688-4 Harley CB, 1991, Telomere loss: mitotic clock or genetic time bomb? Mutat Res 256:271–282. , DOI 10.1016/0921- 8734(91)90018-7 Rizvi S, Raza ST, Mahdi F, 2014, Telomere length variations in aging and age-related diseases. Curr Aging Sci 7:161–167 https:// doi.org/10.2174/1874609808666150122153151 Goya RG, Console GM, Spinelli OM, Carino MH, Riccillo F, Corrons FJ, 2003, Glucocorticoid-induced apoptosis in lymphoid organs is associated with a delayed increase in circulating deoxyribonucleic acid. Apoptosis 8:171–177. , DOI 10.1023/A: 1022922726418 Perez LM, Amaral MA, Mundstock E, Barbe-Tuana FM, Guma FTCR, Jones MH, Machado DC, Sarria EE, Marques e Marques M, Preto LT, Epifanio M, Meinem Garbin JG, Mattiello R, 2017, Effects of diet on telomere length: systematic review and metaanalysis. Public Health Genomics 20:286–292. , DOI 10. 1159/000486586 Cherepanova AV, Tamkovich SN, Bryzgunova OE, Vlassov VV, Laktionov PP, 2008, Deoxyribonuclease activity and circulating DNA concentration in blood plasma of patients with prostate tumors. Ann N Y Acad Sci 1137:218–221. , DOI 10.1196/ annals.1448.016 Astuti Y,Wardhana A,Watkins J, WulaningsihW(2017, Cigarette smoking and telomere length: a systematic review of 84 studies and meta-analysis. Environ Res 158:480–489. , DOI 10.1016/j. envres.2017.06.038 Haghiac M, Vora NL, Basu S, Johnson KL, Presley L, Bianchi DW, deMouzon SH, 2012, Increased death of adipose cells, a path to release cell free DNA into systemic circulation of obese women. Obesity, Silver Spring)Md 20:2213–2219. , DOI 10.1038/ oby.2012.138 Needham BL, Adler N, Gregorich S, Rehkopf D, Lin J, Blackburn EH, Epel ES, 2013, socioeconomic status, health behavior, and leukocyte telomere length in the National Health and nutrition examination survey, 1999–2002. Soc SciMed 85:1–8. , DOI 10.1016/j.socscimed.2013.02.023 Lu W, Zhang Y, Liu D, Songyang Z, Wan M, 2013, Telomeresstructure, function, and regulation. Exp Cell Res 319:133–141 , DOI 10.1016/j.yexcr.2012.09.005 Haycock PC, Heydon EE, Kaptoge S, Butterworth AS, Thompson A, Willeit P, 2014, Leucocyte telomere length and risk of cardiovascular disease: systematic review and meta-analysis. BMJ 349: g4227. , DOI 10.1136/bmj.g4227 Pavanello S, Angelici L, Hoxha M, Cantone L, Campisi M, Tirelli AS, Vigna L, Pesatori AC, Bollati V, 2018, Sterol 27-hydroxylase polymorphism significantly associates with shorter telomere, higher cardiovascular and Type-2 diabetes risk in obese subjects. Front Endocrinol 9:309. , DOI 10.3389/fendo.2018.00309 Russo P, Prinzi G, Proietti S, Lamonaca P, Frustaci A, Boccia S, Amore R, Lorenzi M, onder G, Marzetti E, Valdiglesias V, Guadagni F, Valente MG, Cascio GL, Fraietta S, Ducci G, Bonassi S, 2018, Shorter telomere length in schizophrenia: evidence from a real-world population and meta-analysis of most recent literature. Schizophr Res 202:37–45. , DOI 10.1016/j. schres.2018.07.015 Needham BL, Mezuk B, Bareis N, Lin J, Blackburn EH, Epel ES, 2015, Depression, anxiety and telomere length in young adults: evidence from the National Health and nutrition examination survey. Mol Psychiatry 20:520–528. , DOI 10.1038/mp.2014. 89 Yaffe K, Lindquist K, Kluse M, Cawthon R, Harris T, Hsueh WC, Simonsick EM, Kuller L, Li R, Ayonayon HN, Rubin SM, Cummings SR, 2011, Telomere length and cognitive function in community-dwelling elders: findings from the health ABC study. Neurobiol Aging 32:2055–2060. , DOI 10.1016/j. neurobiolaging.2009.12.006 Sun B, Wang Y, Kota K, Shi Y, Motlak S, Makambi K, Loffredo CA, Shields PG, Yang Q, Harris CC, Zheng YL, 2015, Telomere length variation: a potential new telomere biomarker for lung cancer risk. Lung Cancer 88:297–303. , DOI 10.1016/j.lungcan. 2015.03.011 Latifovic L, Peacock SD, Massey TE, King WD, 2016, The influence of alcohol consumption, cigarette smoking, and physical activity on leukocyte telomere length. Cancer Epidemiol Biomarkers Prev 25:374–380. , DOI 10.1158/1055-9965.EPI-14-1364 Shammas MA, 2011, Telomeres, lifestyle, cancer, and aging. Curr Opin Clin Nutr Metab Care 14:28–34. , DOI 10.1097/ MCO.0b013e32834121b1 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2281 EP 2289 DI 10.1007/s12253-020-00819-x PG 9 ER PT J AU Chai, YB Yip, KW Dusa, N Mohtarrudin, N Seow, FH AF Chai, Yean Boon Yip, Kien Wai Dusa, Noraini Mohtarrudin, Norhafizah Seow, Fong Heng TI Loss of Interleukin-17RA Expression is Associated with Tumour Progression in Colorectal Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE IL-17; IL-17RA; Colorectal carcinoma; Prognostic biomarker; Tumour progression ID IL-17; IL-17RA; Colorectal carcinoma; Prognostic biomarker; Tumour progression AB Interleukin-17 (IL-17) is a pro-inflammatory cytokine found in various cancers. Current evidence indicates that IL-17 plays a vital role in tumour initiation and progression in colorectal carcinoma (CRC) via binding with its receptor, IL-17RA. However, the association between clinicopathological features and presence of IL-17 and IL-17RA protein in primary CRC tissues remains unclear. This study also investigates the difference between the presence of IL-17 and IL-17RA in the paired tumour tissues versus adjacent normal tissues. The presence of IL-17RA and IL-17 protein in primary CRC tissues was determined by immunohistochemistry. Associations between clinicopathological features and IL-17RA and IL-17 immunoreactivity, were analyzed by χ2 tests. We found that both IL-17RA (p = 0.001) and IL-17 (p = 0.025) in tumour cells of primary CRC tissues was significantly lower as compared to adjacent normal tissue. Positive immunoreactivity for IL-17RA and IL-17 were detected in 51.0% and 16.8% of tumour tissues, respectively. Furthermore, negative immunoreactivity of IL-17R was significantly associated with advanced stage according to TNM classifier (p = 0.027), high grade of tumour (p = 0.019), increased depth of tumour invasion (p = 0.023) and vascular invasion (p = 0.039). Positive IL-17 immunoreactivity was associated with advanced stage (p = 0.008) and lymph node metastasis (p = 0.008). Thus, this study suggests that the loss of IL-17RA expression occurs as tumour progresses and this may predict the aggressiveness of tumour whilst expression of IL-17 promotes tumour progression and lymph node metastasis. Thus, loss of IL-17RA could be a useful prognostic biomarker for tumour progression in CRC patients. C1 [Chai, Yean Boon] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, Immunology Laboratory, 43400 Serdang, Selangor, Malaysia. [Yip, Kien Wai] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, Immunology Laboratory, 43400 Serdang, Selangor, Malaysia. [Dusa, Noraini] Hospital Kuala Lumpur, Department of PathologyKuala Lumpur, Malaysia. [Mohtarrudin, Norhafizah] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, 43400 Serdang, Selangor, Malaysia. [Seow, Fong Heng] Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, Immunology Laboratory, 43400 Serdang, Selangor, Malaysia. RP Seow, FH (reprint author), Universiti Putra Malaysia, Faculty of Medicine and Health Sciences, Department of Pathology, Immunology Laboratory, 43400 Serdang, Malaysia. EM shf@upm.edu.my CR Erreni M, Mantovani A, Allavena P, 2011, Tumor-associated Macrophages, TAM, and inflammation in colorectal cancer. Cancer Microenviron 4(2):141–154. , DOI 10.1007/ s12307-010-0052-5 Pernot S, Terme M, Voron T, Colussi O, Marcheteau E, Tartour E, Taieb J, 2014, Colorectal cancer and immunity: what we know and perspectives.World J Gastroenterol 20(14):3738–3750. https://doi. org/10.3748/wjg.v20.i14.3738 Sadanandam A, Lyssiotis CA, Homicsko K, Collisson EA, Gibb WJ,Wullschleger S,Ostos LC, LannonWA, Grotzinger C, Del Rio M, Lhermitte B, Olshen AB, Wiedenmann B, Cantley LC, Gray JW, Hanahan D, 2013, A colorectal cancer classification system that associates cellular phenotype and responses to therapy. Nat Med 19(5):619–625. , DOI 10.1038/nm.3175 Misiakos EP, Karidis NP, Kouraklis G, 2011, Current treatment for colorectal liver metastases. World J Gastroenterol 17(36):4067– 4075. , DOI 10.3748/wjg.v17.i36.4067 JinW, Dong C, 2013, IL-17 cytokines in immunity and inflammation. Emerg Microbes Infect. 2. , DOI 10.1038/emi.2013. 58 Punt S, Langenhoff JM, Putter H, Fleuren GJ, Gorter A, Jordanova ES, 2015, The correlations between IL-17 vs. Th17 cells and cancer patient survival: a systematic review. Oncoimmunology 4(2):1– 10. , DOI 10.4161/2162402X.2014.984547 Wu D, Wu P, Huang Q, Liu Y, Ye J, Huang J, 2013, Interleukin- 17: a promoter in colorectal cancer progression. Clin Dev Immunol. 2013:436307. , DOI 10.1155/2013/436307 Liu J, Duan Y, Cheng X, Chen X, Xie W, Long H, Lin Z, Zhu B, 2011, IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma. Biochem Biophys Res Commun 407(2):348– 354. , DOI 10.1016/j.bbrc.2011.03.021 Yu JJ, Gaffen SL, 2008, Interleukin-17: a novel inflammatory cytokine that bridges innate and adaptive immunity. Front Biosci 13(13):170–177. , DOI 10.2741/2667 Gu C,Wu L, Li X, 2013, Cytokine IL-17 family: cytokines, receptors and signaling. Cytokine 64(2):477–485. , DOI 10. 1016/j.cyto.2013.07.022 Song X, Qian Y, 2013, IL-17 family cytokines mediated signaling in the pathogenesis of inflammatory diseases. Cell Signal 25(12): 2335–2347. , DOI 10.1016/j.cellsig.2013.07.021 Fabre J, Giustiniani J, Garbar C, Antonicelli F, Merrouche Y, Bensussan A, Bagot M, Al-Dacak R, 2016, Targeting the tumor microenvironment: The protumor effects of IL-17 related to cancer type. Int J Mol Sci 17(9). , DOI 10.3390/ijms17091433 Doroudchi M, Pishe ZG, Malekzadeh M, Golmoghaddam H, Taghipour M, Ghaderi A, 2013, Elevated Serum IL-17A but not IL-6 in glioma versus meningioma and schwannoma. Asian Pac J Cancer Prev 14(9):5225–5230. , DOI 10.7314/apjcp.2013. 9.5225 14. Pan B, Shen J, Cao J, Zhou Y, Shang L, Jin S, Cao S, Che D, Liu F, Yu Y, 2015, Interleukin-17 promotes angiogenesis by stimulating VEGF production of cancer cells via the STAT3/GIV signaling pathway in non-small-cell lung cancer. Sci Rep. 2015. https://doi. org/10.1038/srep16053 Cui G, Yuan A, Goll R, Florholmen J, 2012, IL-17A in the tumor microenvironment of the human colorectal adenoma-carcinoma sequence. Scand J Gastroenterol 47(11):1304–1312. , DOI 10.3109/00365521.2012.725089 Steiner GE, NewmanME, Paikl D, Stix U,Memaran-Dagda N, Lee C, Marberger MJ, 2003, Expression and function of proinflammatory interleukin IL-17 and IL-17 receptor in normal, benign hyperplastic, and malignant prostate. Prostate 56(3):171–182. , DOI 10.1002/pros.10238 Murugaiyan G, Saha B, 2009, Protumor vs antitumor functions of IL-17. J Immunol 183(7):4169–4175. , DOI 10.4049/ jimmunol.0901017 Yang JZ, Li Y, Sun LX, Fang J, Kong LJ, Zhang JQ, 2014, Interleukin-17 receptor expression on vascular endothelial cells of masses of skeletal extramedullary disease in myeloma patients. Pathol Res Pract 210(9):586–590. , DOI 10.1016/j.prp. 2014.04.018 Ibrahim S, Girault A, OhresserM, Lereclus E, Paintaud G, Lecomte T, Raoul W, 2018, Monoclonal antibodies targeting the IL-17/IL- 17RA axis: an opportunity to improve the efficiency of anti-VEGF therapy in fighting metastatic colorectal cancer? Clin Colorectal Cancer 17(1):e109–e113. , DOI 10.1016/j.clcc.2017.10. 003 Razi S, BaradaranNoveiry B, Keshavarz-Fathi M, Rezaei N, 2019, IL-17 and colorectal cancer: From carcinogenesis to treatment. Cytokine 116:7–12. , DOI 10.1016/j.cyto.2018.12.021 Yang B, Kang H, Fung A, Zhao H,Wang T,Ma D, 2014, The role of interleukin 17 in tumour proliferation, angiogenesis, and metastasis. Mediators Inflamm. 2014:. , DOI 10.1155/2014/ 623759 Amicarella F, Muraro MG, Hirt C, Cremonesi E, Padovan E, Mele V, GovernaV, Han J, HuberX,Droeser RA, ZuberM, AdaminaM, Bolli M, Rosso R, Lugli A, Zlobec I, Terracciano L, Tornillo L, Zajac P, Eppenberger-Castori S, Trapani F, Oertli D, Iezzi G.(2017). Dual role of tumour-infiltrating T helper 17 cells in human colorectal cancer. 66:692–704. , DOI 10.1136/gutjnl- 2015-310016 Wu Z, He D, Zhao S, Wang H, 2019, IL-17A/IL-17RA promotes invasion and activates MMP-2 and MMP-9 expression via p38 MAPK signaling pathway in non-small cell lung cancer. 455(1– 2):195–206. , DOI 10.1007/s11010-018-3483-9 Ernst M, Putoczki T, 2014, IL-17 Cuts to the Chase in Colon Cancer. Immunity 41(6):880–882. , DOI 10.1016/j. immuni.2014.12.004 Moseley TA, Haudenschild DR, Rose L, Reddi AH, 2003, Interleukin-17 family and IL-17 receptors. Cytokine Growth Factor Rev 14(2):155–174. , DOI 10.1016/S1359- 6101(03)00002-9 Liu Y, Zhao X, Sun X, Li Y, Wang Z, Jiang J, Han H, Shen W, Corrigan CJ, Sun Y, 2015, Expression of IL-17A, E, and F and their receptors in human prostatic cancer: comparison with benign prostatic hyperplasia. Prostate 75(16):1844–1856. , DOI 10.1002/pros.23058 Li YX, Zhang L, Simayi D, Zhang N, Tao L, Yang L, 2015, Human papilloma virus infection correlates with inflammatory Stat3 signaling activity and IL-17 level in patients with colorectal cancer. 10(2):e0118391. , DOI 10.1371/journal.pone. 0118391 Jiang Y, Li P, Yang S, Hao Y, Yu P, 2015, Increased chemokine receptor IL-17RA expression is associated with poor survival in gastric cancer patients. 8(6):7002–7008 Wang M, Wang L, Ren T, Xu L, Wen Z, 2013, IL-17A/IL-17RA interaction promoted metastasis of osteosarcoma cells. Cancer Biol Ther 14(2):155–163. , DOI 10.4161/cbt.22955 Yan C, HuangWY, Boudreau J, Mayavannan A, Cheng Z,Wang J, 2019, IL-17R deletion predicts high-grade colorectal cancer and poor clinical outcomes. Int J Cancer 145(2):548–558. https://doi. org/10.1002/ijc.32122 Fujii T, Sutoh T, Morita H, Yajima R, Yamaguchi S, Tsutsumi S, Asao T, Kuwano H, 2014, Vascular invasion, but not lymphatic invasion, of the primary tumor is a strong prognostic factor in patients with colorectal cancer. Anticancer Res 34(6):3147–3152 Xie Z, Qu Y, Leng Y, Sun W, Ma S, Wei J, Hu J, Zhang X, 2015, Human colon carcinogenesis is associated with increased interleukin-17-driven inflammatory responses. Drug Des Devel Ther 9:1679–1689. , DOI 10.2147/DDDT.S79431 Wu D, Wu P, Huang Q, Liu Y, Ye J, Huang J, 2013, Interleukin- 17: a promoter in colorectal cancer progression. Clin Dev Immunol. 2013:436307. , DOI 10.1155/2013/436307 Tseng JY, Yang CY, Liang SC, Liu RS, Yang SH, Lin JK, Chen YM, Wu YC, Jiang JK, Lin CH, 2014, Interleukin-17A modulates circulating tumor cells in tumor draining vein of colorectal cancers and affects metastases. Clin Cancer Res 20(11):2885–2897. https:// doi.org/10.1158/1078-0432.CCR-13-2162 Lin Y, Xu J, Su H, ZhongW,Yuan Y, Yu Z, FangY, Zhou H, Li C, Huang K, 2015, Interleukin-17 is a favorable prognostic marker for colorectal cancer. Clin Transl Oncol 17(1):50–56. , DOI 10.1007/s12094-014-1197-3 Qiu L, He D, Fan X, Li Z, Liao C, Zhu Y, Wang H, 2011, The expression of interleukin, IL)-17 and IL-17 receptor andMMP-9 in human pituitary adenomas. Pituitary 14(3):266–275. https://doi. org/10.1007/s11102-011-0292-5 Rizzo A, Pallone F, Monteleone G, Fantini MC, 2011, Intestinal inflammation and colorectal cancer: a double-edged sword? World J Gastroenterol. 14;17(26):3092 – 100. , DOI 10.3748/wjg. v17.i26.3092 ChowMT,Moller A, SmythMJ, 2012, Inflammation and immune surveillance in cancer. Semin Cancer Biol 22(1):23–32. https://doi. org/10.1016/j.semcancer.2011.12.004 Riabov V, Gudima A, Wang N, Mickley A, Orekhov A, Kzhyshkowska J, 2014, Role of tumor associated macrophages in tumor angiogenesis and lymphangiogenesis. Front Physiol 5: 75. , DOI 10.3389/fphys.2014.00075 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2291 EP 2298 DI 10.1007/s12253-020-00820-4 PG 8 ER PT J AU Kakimoto, S Miyamoto, M Einama, T Matsuura, H Iwahashi, H Ishibashi, H Sakamoto, T Hada, T Takano, M AF Kakimoto, Soichiro Miyamoto, Morikazu Einama, Takahiro Matsuura, Hiroko Iwahashi, Hideki Ishibashi, Hiroki Sakamoto, Takahiro Hada, Taira Takano, Masashi TI Co-Expression of Mesothelin and CA125 Is Associated with the Poor Prognosis of Endometrial Serous Carcinoma and Mixed Carcinomas Including Serous Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrial carcinoma; Serous carcinoma; Mixed carcinoma; Mesothelin; CA125; Prognosis ID Endometrial carcinoma; Serous carcinoma; Mixed carcinoma; Mesothelin; CA125; Prognosis AB The aim of this study was to investigate the association between the clinicopathologic factors and either expression or coexpression of mesothelin and cancer antigen (CA) 125 in endometrial serous carcinoma and mixed carcinomas including serous carcinoma. Between 1990 and 2017, patients with endometrial serous carcinoma and mixed carcinoma including serous carcinoma treated by total hysterectomy and bilateral salpingo-oophorectomy at our hospital were identified. The association between either expression or co-expression of mesothelin and CA125 was evaluated by immunochemical analysis and the clinicopathological features were retrospectively examined. Among the 40 patients included, 19, 31, and 18 patients exhibited single positive mesothelin, single positive CA125, and positive co-expression, respectively. The expression of mesothelin and CA125 was observed to be positively associated (p = 0.021). There was no significant association of age and FIGO stage with individual mesothelin or CA125 expression or their co-expression. Overall survival (OS), but not progression-free survivals (PFS), of only mesothelin-positive patientswas worse (p = 0.024). Hence, OS and PFS of patients with positive co-expression were worse (PFS: p = 0.043, OS: p = 0.012). In multivariate analysis, single mesothelin expression and single CA125 expression did not lead to worse prognosis. However, positive co-expression was the worst prognostic factor for OS (hazard ratio: 3.32, p = 0.039). Coexpression of mesothelin and CA125 may accurately predict OS in endometrial serous carcinoma and mixed carcinomas including serous carcinoma. Further studies should examine this relationship. C1 [Kakimoto, Soichiro] National Defense Medical College, Department of Obstetrics and Gynecology, 359-8513 Tokorozawa, Saitama, Japan. [Miyamoto, Morikazu] National Defense Medical College, Department of Obstetrics and Gynecology, 359-8513 Tokorozawa, Saitama, Japan. [Einama, Takahiro] National DefenseMedical College Hospital, Department of Surgery, 359-8513 Tokorozawa, Saitama, Japan. [Matsuura, Hiroko] National Defense Medical College, Department of Obstetrics and Gynecology, 359-8513 Tokorozawa, Saitama, Japan. [Iwahashi, Hideki] National Defense Medical College, Department of Obstetrics and Gynecology, 359-8513 Tokorozawa, Saitama, Japan. [Ishibashi, Hiroki] National Defense Medical College, Department of Obstetrics and Gynecology, 359-8513 Tokorozawa, Saitama, Japan. [Sakamoto, Takahiro] National Defense Medical College, Department of Obstetrics and Gynecology, 359-8513 Tokorozawa, Saitama, Japan. [Hada, Taira] National Defense Medical College, Department of Obstetrics and Gynecology, 359-8513 Tokorozawa, Saitama, Japan. [Takano, Masashi] National Defense Medical College, Department of Obstetrics and Gynecology, 359-8513 Tokorozawa, Saitama, Japan. RP Miyamoto, M (reprint author), National Defense Medical College, Department of Obstetrics and Gynecology, 359-8513 Tokorozawa, Japan. EM mmiyamoto@ndmc.ac.jp CR Wartko P, Sherman ME, Yang HP, Felix AS, Brinton LA, Trabert B, 2013, Recent changes in endometrial cancer trends among menopausal age US women. Cancer Epidemiol 37:374–377 Bokhman JV, Chepick OF, Volkova AT, Vishnevsky AS, 1981, Adjuvant hormone therapy of primary endometrial carcinoma with oxyprogesterone caproate. Gynecol Oncol 11:371–378 Wright JD, Medel NB, Sehouli J, Fujiwara K, Herzog TJ, 2012, Contemporary management of endometrial cancer. Lancet 379: 1352–1360 Tashiro H, Isacson C, Levine R, Kurman RJ, Cho KR, Hedrick L, 1997, p53 genemutations are common in uterine serous carcinoma and occur early in their pathogenesis. Am J Pathol 150:177–185 Hendrickson M, Ross J, Eifel P, Martinez A, Kempson R, 1982, Uterine papillary serous carcinoma: a highly malignant form of endometrial carcinoma. Am J Surg Pathol 6:93–108 FaderAN, Starks D, Gehrig PA, Secord AA, Frasure HE,O’Malley DM, Tuller ER, Rose PG, Havrilesky LJ, Moore KN, Huh WK, Axtell AE, Kelley JL, Zanotti KM, 2009, UPSC consortium. An updated clinicopathologic study of early-stage uterine papillary serous carcinoma, UPSC). Gynecol Oncol 115:244–248 Faratian D, Stillie A, Busby-Earle RM, Cowie VJ, Monaghan H, 2006, A review of the pathology and management of uterine papillary serous carcinoma and correlation with outcome. Int J Gynecol Cancer 16:972–978 Miyamoto M, Takano M, Tsuda H, Soyama H, Aoyama T, Ishibashi H, Kato K, Iwahashi H, Matuura H, Yoshikawa T, Suzuki A, Hirata J, Furuya K, 2017, Small foci of serous component as a predictor of recurrence and prognosis for stage IA endometrial carcinomas. Oncology. 93:29–35 Chang K, Pastan I,WillinghamMC, 1992, Isolation and characterization of a monoclonal antibody, K1, reactive with ovarian cancers and normal mesothelium. Int J Cancer 50:373–381 Chang K, Pastan I, 1996, Molecular cloning of mesothelin, a differentiation antigen present on mesothelium, mesotheliomas, and ovarian cancers. Proc Natl Acad Sci U S A 93:136–140 Morello A, Sadelain M, Adusumilli P, 2016, Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov 6:133–146 Dainty LA, Risinger JI, Morrison C, Chandramouli GVR, Bidus MA, Zahn C, Rose GS, Fowler J, Berchuck A, Maxwell GL, 2007, Overexpression of folate binding protein and mesothelin are associated with uterine serous carcinoma. Gynecol Oncol 105:563–570 Kachala SS, Bograd AJ, Villena-Vargas J, Suzuki K, Servais EL, Kadota K, Chou J, Sima CS, Vertes E, Rusch VW, Travis WD, Sadelain M, Adusumilli PS, 2014, Mesothelin overexpression is a marker of tumor aggressiveness and is associated with reduced recurrence-free and overall survival in early stage lung adenocarcinoma. Clin Cancer Res 20:1020–1028 Einama T, Kawamata F, Kamachi H, Nishihara H, Homma S, Matsuzawa F, Mizukami T, Konishi Y, Tahara M, Kamiyama T, Hino O, Taketomi A, Todo S, 2016, Clinical impacts ofmesothelin expression in gastrointestinal carcinomas. World J Gastrointest Pathphysiol 7:218–222 Ordonez NG, 2003, Application of mesothelin immunostaining in tumor diagnosis. Am J Surg Pathol 27:1418–1428 Kawamata F, Homma S, Kamachi H, Einama T, Kato Y, Tsuda M, Tanaka S, Maeda M, Kajino K, Hino O, Takahashi N, Kamiyama T, Nishihara H, Taketomi A, Todo S, 2014, C-ERC/mesothelin provokes lymphatc invasion of colorectal adenocarcinoma. J Gastroenterol 49:81–92 Shiraishi T, Shinto E, Mochizuki S, Tsuda H, Kajiwara Y, Okamoto K, Einama T, Hase K, Ueno H, 2019, Mesothelin expression has prognostic value in stage II/III colorectal cancer. Virchows Arch 474:297–307 Nahm CB, Turchini J, Jamieson N, Moon E, Sioson L, Itchins M, Arena J, Colvin E, Howell VM, Pavlakis N, Clarke S, Samra JS, Gill AJ, Mittal A, 2019, Biomarker panel predicts survival after resection in pancreatic ductal adenocarcinoma: a multiinstitutional cohort study. Eur J Surg Oncol 45:218–224 Thomas A, Chen Y, Steinberg SM, Luo J, Pack S, Abdullaev Z, Alewine C, Rajan A, Giaccone G, Pastan I, MiettinenM, Hassan R, 2015, High mesothelin expression in advanced lung adenocarcinoma is associated with KRAS mutations and a poor prognosis. Oncotarget 6:11694–11703 Li YR, Xian RR, Ziober A, Conejo-Garcia J, Perales-Puchalt A, June CH, Zhang PJ, Tchou J, 2014)Mesothelin expression is associated with poor outcomes in breast cancer. Breast Cancer Res Treat 147:675–684 O’Brien TJ, Beard JB, Underwood LJ, Shigemasa K, 2002, The CA 125 gene: a newly discovered extension of the glycosylated Nterminal domain doubles the size of this extracellular superstructure. Tumour Biol 23:154–169 Sl T, James TQ, Sanford CS, 1992, Distribution of CA125 in adenocarcinomas: an immunohistochemical study of 481 cases. Am J Clin Pathol 98:175–179 Streppel MM, Vincent A, Mukherjee R, Campbell NR, Chen S, Konstantopoulos K, Goggins MG, Seuningen IV, Maitra A, Montgomery EA, 2012, Mucin 16, cancer antigen 125, expression in human tissues and cell lines and correlation with clinical outcome in adenocarcinomas of the pancreas, esophagus, stomach, and colon. Hum Pathol 43:1755–1763 Rosen DG, Wang L, Atkinson JN, Yu Y, Lu KH, Diamandis EP, Hellstrom I, Mok SC, Liu J, Bast RC, 2005, Potential markers that complement expression of CA125 in epithelial ovarian cancer. Gynecol Oncol 99:267–277 de la Cuesta R, Maestro ML, Solana J, Vidart JA, Escudero M, Iglesias E, Valor R, 1999, Tissue quantification of CA125 in epithelial ovarian cancer. Int J Biol Markers 14:106–114 Ginath S, Menczer J, Fintsi Y, Ben-Shem E, Glezerman M, Avinoach I, 2002, Tissue and serum CA125 expression in endometrial cancr. Int J Gynecol Cancer 12:372–375 Hogdall EVS, Christensen L, Kjaer SK, Blaakaer J, Kjaerbye- Thygesen A, Gayther S, Jacobs IJ, Hogdall CK, 2007, CA125 expression pattern, prognosis and correlation with serum CA125 in ovarian tumor patients from the Danish “MALOVA” ovarian cancer study. Gynecol Oncol 104:508–515 Gubbeles JA, Belisle J, Onda M, Rancoourt C, Migneault M, Ho M, Bera TK, Connor J, Sathyanarayana BK, Lee B, Pastan I, Patankar MS, 2006, Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol Cancer 5:50 Einama T, Kamachi H, Nishihara H, Homma S, Kannno H, Takahashi K, Sasaki A, Tahara M, Okada K, Murakawa S, Kamiyama T, Matsuno Y, Ozaki M, Todo S, 2011, Coexpression of mesothelin and CA125 correlates with unfavorable patient outcome in pancreatic ductal adenocarcinoma. Pancreas 40: 1276–1282 Rump A, Morikawa Y, Tanaka M, Minami S, Umesaki N, TakeuchiM, Miyajima A, 2004, Binding of ovarian cancer antigen CA125/MUC16 tomesothelinmediates cell adhesion. J Biol Chem 279:9190–9198 Einama T, Homma S, Kamachi H, Kawamata F, Takehashi K, Takahashi N, Taniguchi M, Kamiyama T, Furukawa H, Matsuno Y, Tanaka S, Nishihara H, Taketomi A, Todo S, 2012, Luminal membrane expression of mesothelin is a prominent poor prognostic factor for gastric cancer. Br J Cancer 107:137–142 Einama T, Kamachi H, NishiharaH, Homma S, Kanno H, Ishikawa M, Kawamata F, Konishi Y, SatoM, TaharaM, Okada K, Muraoka S, Kamiyama T, Taketomi A, Matsuno Y, Furukawa H, Todo S, 2015, Importance of luminal membrane mesothelin expression in intraductal papillary mucinous neoplasms. Oncol Lett 9:1583–1589 Slomovites BM, Burke TW, Eifel PJ, Ramondetta LM, Silva EG, Jhingran A, Oh JC, Atkinson EN, Broaddus RR, Gershenson DM, LuKH(2003, Uterine papillary serous caricinoma, UPSC): a single institution review of 129 cases. Gynecol Oncol 91:463–469 He X,Wang L, Riedel H,Wang K, Yang Y, Dinu CZ, Rojanasakul Y, 2017, Mesothelin promotes epithelial-to-mesenchymal transition and tumorigenicity of human lung cancer and mesothelioma cells. Mol Cancer 16:63 Bhadwaj U, Marin-Muller C, Li M, Chen C, Yao Q, 2011, Mesothelin confers pancreatic cancer cell resistance to TNFalpha- induced apoptosis through Akt/P13K/NF-kappaB activation and IL-6/Mcl-1 overexpression. Mol Cancer 10:106 Hilliard TS, 2018, The impact of mesothelin in the ovarian cancer tumor microenvironment. Cancers 10:277 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2299 EP 2306 DI 10.1007/s12253-020-00823-1 PG 8 ER PT J AU Meszaros, N Smanyko, V Major, T Stelczer, G Janvary, ZsL Kovacs, E Baheri, M Zaka, Z Pukancsik, D Takacsi-Nagy, Z Polgar, Cs AF Meszaros, Norbert Smanyko, Viktor Major, Tibor Stelczer, Gabor Janvary, Zsolt Levente Kovacs, Eszter Baheri, Maria Zaka, Zoltan Pukancsik, David Takacsi-Nagy, Zoltan Polgar, Csaba TI Implementation of Stereotactic Accelerated Partial Breast Irradiation Using Cyber-Knife – Technical Considerations and Early Experiences of a Phase II Clinical Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; APBI; SBRT; Cyber-knife; Phase II ID Breast cancer; APBI; SBRT; Cyber-knife; Phase II AB To report the implementation, dosimetric results of and early experiences with stereotactic accelerated partial breast irradiation (SAPBI) following breast conserving surgery (BCS) for postmenopausal low-risk St I-II invasive breast cancer (IBC) patients. Between November 2018 and August 2019, 27 patients were registered in our phase II prospective study. SAPBI was performed with Cyber-Knife (CK) M6 machine, in 4 daily fractions of 6.25 Gy to a total dose of 25 Gy. Respiratory movements were followed with implanted gold markers and Synchrony system. Corrections for patient displacement and respiratory movement during treatment were performed with the robotic arm. Early side effects, cosmetic results, and dosimetric parameters were assessed. The average volume of the surgical cavity, clinical target volume (CTV), and planning target volume (PTV_EVAL) were 8.1 cm3 (range: 1.75–27.3 cm3), 55.3 cm3 (range: 26.2–103.5 cm3), and 75.7 cm3 (range: 40–135.4 cm3), respectively. The mean value of the PTV_eval/whole breast volume ratio was 0.09 (range: 0.04–0.19). No grade 2 or worst acute side-effect was detected. Grade 1 (G1) erythema occurred in 6 (22.2%) patients, while G1 oedema was reported by 3 (11.1%) cases. G1 pain was observed in 1 (3.4%) patient. Cosmetic result were excellent in 17 (62.9%) and good in 10 (37.1%) patients. SAPBI with CK is a suitable and practicable technique for the delivery of APBI after BCS for low-risk, St. I-II. IBC. Our early findings are encouraging, CK-SAPBI performed with four daily fractions is convenient and perfectly tolerated by the patients. C1 [Meszaros, Norbert] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Smanyko, Viktor] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Major, Tibor] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Stelczer, Gabor] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Janvary, Zsolt Levente] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Kovacs, Eszter] National Institute of Oncology, Center of RadiologyBudapest, Hungary. [Baheri, Maria] National Institute of Oncology, Center of RadiologyBudapest, Hungary. [Zaka, Zoltan] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Pukancsik, David] National Institute of Oncology, Department of SurgeryBudapest, Hungary. [Takacsi-Nagy, Zoltan] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. [Polgar, Csaba] National Institute of Oncology, Center of Radiotherapy, Rath Gyorgy u. 7-9, H-1122 Budapest, Hungary. RP Meszaros, N (reprint author), National Institute of Oncology, Center of Radiotherapy, H-1122 Budapest, Hungary. EM meszarosnorbert@oncol.hu CR Fischer B, Anderson S, Bryant J et al, 2002, Twenty-year follow up of a randomized trial comparing total mastectomy, lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 347:1233–1241 Veronesi U, Cascinelli N, Mariani L, Greco M, Saccozzi R, Luini A, Aguilar M, Marubini E, 2002, Twenty-year follow-up of a randomized study comparing breast-conserving surgery with radical mastectomy for early breast cancer. N Engl J Med 347:1227–1232 Darby S, McGale P, Correa C et al, 2011, Effect of radiotherapy after breast-conserving surgery on 10-year recurrence and 15-year breast cancer death: meta-analysis of individual patient data for 10, 801 women in 17 randomised trials. Lancet 378:1707–1716 Holland R, Veling SH, Mravunac M et al, 1985, Histologic multifocality of tis, T1-2 breast carcinomas. Implications for clinical trials of breast-conserving surgery. Cancer 56(5):979–990 Faverly D, Holland R, Burgers L, 1992, An original stereomicroscopic analysis of the mammary glandular tree. Virchows Arch A Pathol Anat Histopathol 421(2):115–119 Veronesi U, Cascinelli N, Mariani L, Greco M, Saccozzi R, Luini A, AguilarM,Marubini E, 2002, Twenty-year follow-up of a randomized study comparing breast-conserving surgery with radical mastectomy for early breast cancer. N Engl J Med 347(16):1227–1232 Nattinger AB, Hoffmann RG, Kneusel RT, Schapira MM, 2000, Relation between appropriateness of primary therapy for earlystage breast carcinoma and increased use of breast-conserving surgery. Lancet 356:1148–1153 Magee B, Swindell R, Harris M, Banerjee SS, 1996, Prognostic factors for breast recurrence after conservative breast surgery and radiotherapy: results of a randomised trial. Radiother Oncol 39: 223–227 Fentiman IS, Poole C, Tong D et al, 1996, Inadequacy of iridium implant as sole radiation treatment for operable breast cancer. Eur J Cancer 32A:608–611 Strnad V, Ott OJ, Hildebrandt G, Kauer-Dorner D, Knauerhase H, Major T, Lyczek J, Guinot JL, Dunst J, Gutierrez Miguelez C, Slampa P, Allgauer M, Lossl K, Polat B, Kovacs G, Fischedick AR, Wendt TG, Fietkau R, Hindemith M, Resch A, Kulik A, Arribas L, Niehoff P, Guedea F, Schlamann A, Potter R, Gall C, MalzerM, UterW, Polgar C, Groupe Europeen de Curietherapie of European Society for Radiotherapy and Oncology, GEC-ESTRO,, 2016, Accelerated partial breast irradiation, APBI, using sole interstitial brachytherapy versus whole breast irradiation with boost after breast conserving surgery for low risk invasive and is situ carcinoma of the female breast: 5-year results of a randomised phase 3 non-inferiority trial. Lancet 387:229–238 Polgar C, Ott OJ, Hildebrandt G, Kauer-Dorner D, Knauerhase H, Major T, Lyczek J, Guinot JL, Dunst J, Miguelez CG, Slampa P, AllgauerM, Lossl K, Polat B, Kovacs G, FischedickAR, Fietkau R, Resch A, Kulik A, Arribas L, Niehoff P, Guedea F, Schlamann A, Potter R, Gall C, Uter W, Strnad V, Groupe Europeen de Curietherapie of European Society for Radiotherapy and Oncology, GEC-ESTRO,, 2017, Late side-effects and cosmetic results of accelerated partial breast irradiation with interstitial brachytherapy versus whole-breast irradiation after breastconserving surgery for low-risk invasive and in-situ carcinoma of the female breast: 5-year results of a randomised, controlled, phase 3 trial. Lancet Oncol 18:259–268 Schafer R, Strnad V, Polgar C, Uter W, Hildebrandt G, Ott OJ, Kauer-Dorner D, Knauerhase H, Major T, Lyczek J, Guinot JL, Dunst J, Miguelez CG, Slampa P, Allgauer M, Lossl K, Kovacs G, Fischedick AR, Fietkau R, Resch A,Kulik A,Arribas L,Niehoff P, Guedea F, Schlamann A, Gall C, Polat B,Wendt TG, Hindemith M, Potter R,MalzerM(2018, Quality-of-life results for accelerated partial breast irradiation with interstitial brachytherapy versus whole-breast irradiation in early breast cancer after breastconserving surgery, GEC-ESTRO): 5-year results of a randomised, phase 3 trial. Lancet Oncol 19:834–844 Formenti SC, Hsu H, Fenton-Kerimian M, Roses D, Guth A, Jozsef G, Goldberg JD, DeWyngaert JK, 2012, Prone accelerated partial breast irradiation after breast-conserving surgery: five-year results of 100 patients. Int J Radiat Oncol Biol Phys 84:606–611 Shah C, Wilkinson JB, Lanni T, Jawad M, Wobb J, Fowler A, Wallace M, Chen P, Grills IS, Vicini F, 2013, Five-year outcomes and toxicities using 3-dimensional conformal external beam radiation therapy to deliver accelerated partial breast irradiation. Clin Breast Cancer 13:206–211 Vicini F,Winter K,Wong J, PassH, Rabinovitch R, Chafe S,Arthur D, Petersen I,White J,McCormick B, 2010, Initial efficacy results of RTOG 0319: three-dimensional conformal radiation therapy, 3D-CRT, confined to the region of the lumpectomy cavity for stage I/II breast carcinoma. Int J Radiat Oncol Biol Phys 77: 1120–1127 Pashtan IM, Recht A, Ancukiewicz M et al, 2012, External beam accelerated partial-breast irradiation using 32 Gy in 8 twice-daily fractions: 5-year results of a prospective study. Int J Radiat Oncol Biol Phys 84:e-271-7 Lei RY, Leonard CE, Howell CE et al, 2013, Four-year clinical update from a prospective trial of accelerated breast intensity-modulated radiotherapy, APBIMRT). Breast Cancer Res Treat 140: 119–133 Rodriguez N, Sanz X, Dengra J, Foro P,Membrive I, Reig A, Quera J, Fernandez-Velilla E, Pera O, Lio J, Lozano J, Algara M, 2013, Five-year outcomes, cosmesis, and toxicity with 3-dimensional conformal external beam radiation therapy to deliver accelerated partial breast irradiation. Int J Radiat Oncol Biol Phys 87:1051– 1057 Mozsa E, Polgar Cs, Frohlich G et al, 2012, Akceleralt parcialis konformalis kulso emlobesugarzas emlomegtartomutet utan – fazis II prospektv klinikai vizsgalat elozetes eredmenyei. Magy Onkol 56:235–241 Mozsa E, Meszaros N, Major T, Frohlich G, Stelczer G, Sulyok Z, Fodor J, Polgar C, 2014, Accelerated partial breast irradiation with external beam three-dimensional conformal radiotherapy: 5-year results of a prospective phase II clinical study. Strahlenther Onkol 190:444–450 Olivotto IA,Whelan TJ, Parpia S, Kim DH, Berrang T, Truong PT, Kong I, Cochrane B, Nichol A, Roy I, Germain I, Akra M, Reed M, Fyles A, Trotter T, Perera F, Beckham W, Levine MN, Julian JA, 2013, Interim cosmetic and toxicity results from RAPID: a randomized trial of accelerated partial breast irradiation using threedimensional conformal external beam radiation therapy. J Clin Oncol 31:4038–4045 Meszaros N, Major T, Stelczer G, Zaka Z, Mozsa E, Pukancsik D, Takacsi-Nagy Z, Fodor J, Polgar C, 2017, Implementation of image- guided, intensity modulated accelerated partial breast irradiation – three-year results of a phase II clinical study. Strahlenther Onkol Jan 193(1):70–79 Meszaros N, Major T, Stelczer G et al, 2015, Gyorsitott, reszleges kulso emlobesugarzas kepvezerelt, intenzitasmodulalt radioterapiaval emlomegtarto mutet utan – Fazis II klinikai vizsgalat elozetes eredmenyei. Magyar Onkologia 59(2):111–118 Vicini F, Arthur D, Polgar C et al, 2003, Defining the efficacy of accelerated partial breast irradiation: the importance of proper patient selection, adequate quality assurance and common sense. Int J Radiat Oncol Biol Phys 57:1210–1213 Cs P, Van Limbergen E, Potter R et al, 2010, Patient selection for accelerated partial breast irradiation, APBI, after breast-conserving surgery: Recommendations of the Groupe Europeen de Curietherapie-European Society for Therapeutic Radiology and Oncology, GEC-ESTRO, Breast Cancer Working Group based on clinical evidence, 2009). Radiother Oncol 94:264–273 Zhang B, Zhu F, Ma X, Tian Y, Cao D, Luo S, Xuan Y, Liu L,Wei Y, 2014)Matched-pair comparisons of stereotactic body radiotherapy, SBRT, versus surgery for the treatment of early stage nonsmall cell lung cancer: a systematic review and meta-analysis. Radiother Oncol 112(2):250–255 Jorgo K, Agoston P, Janvary L et al, 2019, Stereotactic body radiation therapy with CyberKnife accelerator for low- and intermediate risk prostate cancer. Magy Onkol 63(1):52–59 Bhattacharya IS, Woolf DK, Hughes RJ et al, 2015, Stereotactic body radiotherapy, SBRT, in the management of extracranial oligometastatic, OM, disease. Br J Radiol 88(1048):20140712 Wowra B, Muacevic A, Tonn JC et al, 2012, CyberKnife radiosurgery for brain metastases. Prog Neurol Surg 25:201–209 Obayomi-Davies O, Kole TP, Oppong B et al, 2016, Stereotactic accelerated partial breast irradiation for early-stage breast cancer: rationale, feasibility and early experiences using the CyberKnife radiosurgerydelivery platform. Front Oncol 6:129 Vermeulen S, Cotrutz C, Morris A et al, 2011, Accelerated Partial Breast Irradiation: Using the CyberKnife as the Radiation Delivery Platform in the Treatment of Early Breast Cancer. Front Oncol 1:43 Lozza L, Fariselli L, Sandri M et al, 2018, Partial breast irradiation with CyberKnife after breast conserving surgery: a pilot study in early breast cancer. Radiat Oncol 13(1):49 Xu Q, Chen Y, Grimm J et al, 2012, Dosimetric investigation of accelerated partial breast irradiation, APBI, using CyberKnife.Med Phys 39(11):6621–6628 Zhen X, Zhao B, Wang Z et al, 2017, Comprehensive target geometric errors and margin assessment in stereotactic partial breast irradiation. Radiat Oncol 12(1):151 RahimiA, ThomasK, SpanglerAet al, 2017, Preliminary results of a phase 1 dose-escalation trial for early-stage breast Cancer using 5- fraction stereotactic body radiation therapy for partial-breast irradiation. Int J Radiat Oncol Biol Phys 98(1):196–205.e2 Vermeulen SS, Haas JA et al, 2014, CyberKnife stereotactic body radiotherapy and CyberKnife accelerated partial breast irradiation for the treatment of early breast cancer. Transl Cancer Res 3(4): 295–302 Hepel JT, TokitaM,MacAusland SG et al, 2009, Toxicity of threedimensional conformal radiotherapy for accelerated partial breast irradiation. Int J Radiat Oncol Biol Phys 75:1290–1296 Seiler S, Rahimi A, Choudhery S, GarwoodD, Spangler A, Cherian S, Goudreau S, 2016, Ultrasound-guided placement of gold Fiducial markers for stereotactic partial-breast irradiation. AJR Am J Roentgenol 207(3):685–688 Major T, Gutierrez C, Guix B, van Limbergen E, Strnad V, Polgar C, Breast Cancer Working Group of GEC-ESTRO, 2016, Recommendations from GEC ESTRO Breast Cancer Working Group, II): Target definition and target delineation for accelerated or boost partial breast irradiation using multicatheter interstitial brachytherapy after breast conserving open cavity surgery. Radiother Oncol 118:199–204 Cox JD, Stetz J, Pajak TF et al, 1995, Toxicity criteria of the radiation therapy oncology group, RTOG, and the European Organization for Research and Treatment of Cancer, EORTC). Int J Radiat Oncol Biol Phys 31:1341–1346 Vicini F, Cecchini R,White J et al, 2019, Long-term primary results of accelerated partial breast irradiation after breast-conserving surgery for early-stage breast cancer: a randomised, phase 3, equivalence trial. Lancet 394(10215):2155–2164 Whelan T, Julian J, Levine M et al, 2019, External beam accelerated partial breast irradiation versus whole breast irradiation after breast conserving surgery in women with ductal carcinoma in situ and node-negative breast cancer, RAPID): a randomised controlled trial. Lancet 394(10215):2165–2172 Livi L,Meattini I,Marrazo L et al, 2015, Accelerated partial breast irradiation using intensity-modulated radiotherapy versus whole breast irradiation: 5-year survival analysis of a phase 3 randomised controlled trial. Eur J Cancer 51:451–463 Peterson D, Truong PT, Parpia S, Olivotto IA, Berrang T, Kim DH, Kong I, Germain I, Nichol A, Akra M, Roy I, Reed M, Fyles A, Trotter T, Perera F, Balkwill S, Lavertu S, Elliott E, Julian JA, Levine MN, Whelan TJ, 2015, Predictors of adverse cosmetic outcome in the RAPID trial: an exploratory analysis. Int J Radiat Oncol Biol Phys 91:968–976 Jagsi R, Ben-David MA, Moran JM, Marsh RB, Griffith KA, Hayman JA, Pierce LJ, 2010, Unacceptable cosmesis in a protocol investigating intensity-modulated radiotherapy with active breathing control for accelerated partial-breast irradiation. Int J Radiat Oncol Biol Phys 76:71–78 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2307 EP 2313 DI 10.1007/s12253-020-00821-3 PG 7 ER PT J AU Kengkarn, S Petmitr, S Boonyuen, U Reamtong, O Poomsawat, S Sanguansin, S AF Kengkarn, Sudaporn Petmitr, Songsak Boonyuen, Usa Reamtong, Onrapak Poomsawat, Sopee Sanguansin, Sirima TI Identification of Novel Candidate Biomarkers for Oral Squamous Cell Carcinoma Based on Whole Gene Expression Profiling SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral squamous cell carcinoma; Gene expression profile; Biomarker; PI3; KRT17 ID Oral squamous cell carcinoma; Gene expression profile; Biomarker; PI3; KRT17 AB This study aimed to determine the whole gene expression profiles and to ascertain potential biomarkers for 22 oral squamous cell carcinoma (OSCC) among Thai patients using the Illumina Human HT-12, V4.0 Expression BeadChip array. Result indicated 2,724 differential expressed genes composed of 1,560 up-regulated and 1,164 down-regulated genes (unpaired t-test, p-value <0.05; fold change ≥2.0 and ≤2.0). The top 9 up-regulated genes were validated in 39 OSCC cases using TaqMan real-time quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) assay. Among these, the up-regulation of peptidase inhibitor 3 (PI3) and keratin 17 (KRT17) genes was harbored in all 39 OSCC patients (100%). Likewise, statistical analysis indicated that gene expression in 8 selective genes including keratin 16 (KRT16), keratin 14 (KRT14), keratinocyte differentiation-associated protein (KRTDAP), keratin 6B (KRT6B), PI3, S100 calcium binding protein A7 (S100A7), stratifin (SFN) and keratin 5 (KRT5) was significantly associated with well differentiated OSCC (p-value <0.05). Moreover, high level of KRT17 protein was significantly associated with well differentiated OSCC compared to moderately OSCC (p-value = 0.041). Notably, using nested-PCR analysis indicated all OSCC cases in this study were HPV-free. Especially, KRTDAP, PI3, SFN mRNA expression were first reported among patients with OSCC. Conclusion, the whole transcript expression study and TaqMan real-time qRT-PCR assay were relevant regarding the increase in gene expression in OSCC. In addition, the upregulation of PI3 and KRT17 might constitute potential candidate molecular biomarkers to diagnose patients with OSCC. C1 [Kengkarn, Sudaporn] Mahidol University, Faculty of Tropical Medicine, Department of Molecular Tropical Medicine & GeneticsBangkok, Thailand. [Petmitr, Songsak] Mahidol University, Faculty of Tropical Medicine, Department of Molecular Tropical Medicine & GeneticsBangkok, Thailand. [Boonyuen, Usa] Mahidol University, Faculty of Tropical Medicine, Department of Molecular Tropical Medicine & GeneticsBangkok, Thailand. [Reamtong, Onrapak] Mahidol University, Faculty of Tropical Medicine, Department of Molecular Tropical Medicine & GeneticsBangkok, Thailand. [Poomsawat, Sopee] Mahidol University, Faculty of Dentistry, Department of Oral and Maxillofacial PathologyBangkok, Thailand. [Sanguansin, Sirima] Mahidol University, Faculty of Dentistry, Department of Oral BiologyBangkok, Thailand. RP Sanguansin, S (reprint author), Mahidol University, Faculty of Dentistry, Department of Oral Biology, Bangkok, Thailand. EM sirima.san@mahidol.ac.th CR Ferlay J, SoerjomataramI, Dikshit R, Eser S, Mathers C, RebeloM, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386. https://doi. org/10.1002/ijc.29210 Brinkman BMN, Wong DT, 2006, Disease mechanism and biomarkers of oral squamous cell carcinoma. Curr Opin Oncol 18(3): 228–233. , DOI 10.1097/01.cco.0000219250.15041 National Cancer Institute, 2015, SEER Cancer Statistics Review, 1975–2012 http://seer.cancer.gov/statfacts/html/oralcav.html. Accessed 8 March 2016 Mendez E, Cheng C, Farwell DG, Ricks S, Agoff SN, Futran ND, Weymuller EA Jr, Maronian NC, Zhao LP, Chen C, 2002, Transcriptional expression profiles of oral squamous cell carcinomas. Cancer 95(7):1482–1494. , DOI 10.1002/cncr.10875 Tomioka H,Morita K, Hasegawa S, Omura K, 2006, Gene expression analysis by cDNA microarray in oral squamous cell carcinoma. J Oral Pathol Med 35(4):206–211. , DOI 10.1111/j. 1600-0714.2006.00410x Wang YH, Tian X, Liu OS, Fang XD, Quan HZ, Xie S, Gao S, Tang ZG, 2014, Gene profiling analysis for patients with oral verrucous carcinoma and oral squamous cell carcinoma. Int J Clin Exp Med 7(7):1845–1852 Chakrabarti S, Multani S, Dabholkar J, Saranath D, 2015, Whole genome expression profiling in chewing-tobacco-associated oral cancers: a pilot study. Med Oncol 32(3):60. , DOI 10. 1007/s12032-015-0483-4 Amin MB, Edge S, Greene F et al, 2017, AJCC Cancer Staging Manual, 8th edn. Springer, New York Khovidhunkit SP, Buajeeb W, Sanguansin S, Poomsawat S, Weerapradist W, 2008, Detection of Human Papillomavirus in Oral Squamous Cell Carcinoma, Leukoplakia and Lichen Planus in Thai Patients. Asian Pac J Cancer Prev 9(4):771–775 Nopmaneepaisarn T, Tangjaturonrasme N, Rawangban W, Vinayanuwattikun C, Keelawat S, Bychkov A, 2019, Low Prevalence of p16-positive HPV-related Head-Neck Cancers in Thailand: Tertiary Referral Center Experience. BMC Cancer 19(1):1050. , DOI 10.1186/s12885-019-6266-0 Bhattacharjee A, RichardsWG, Staunton J, Li C, Monti S, Vasa P, Ladd C, Beheshti J, Bueno R, GilletteM, Loda M,Weber G,Mark EJ, Lander ES, Wong W, Johnson BE, Golub TR, Sugarbaker DJ, Meyerson M, 2001, Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. Proc Natl Acad Sci USA 98(24):13790–13795. https://doi. org/10.1073/pnas.191502998 Welsh JB, Zarrinkar PP, Sapinoso LM, Kern SG, Behling CA, Monk BJ, Lockhart DJ, Burger RA, Hampton GM, 2001, Analysis of gene expression profiles in normal and neoplastic ovarian tissue samples identifies candidate molecular markers of epithelial ovarian cancer. Proc Natl Acad Sci USA 98(3):1176–1181. , DOI 10.1073/pnas.98.3.1176 Subrungruang I, Thawornkuno C, Chawalitchewinkoon-Petmitr P, Pairojkul C, Wongkham S, Petmitr S, 2013, Gene expression profiling of intrahepatic cholangiocarcinoma. Asian Pac J Cancer Prev 14(1):557–563. , DOI 10.7314/Apjcp.2013.14.1.557 Arnutt i P, Kotepui M, Asanprakit W, Punyarit P, Chavalitshewinkoon-Petmitr P, Harnroongroj T, Petmitr S, 2013, Determination of whole transcription profiles and specific pathways in invasive ductal breast carcinoma. Int J Clin Exp Patho 6(6):1112–1120 Sakamoto K, Aragaki T, Morita K, Kawachi H, Kayamori K, Nakanishi S, Omura K, Miki Y, Okada N, Katsube K, Takizawa T, Yamaguchi A, 2011, Down-regulation of keratin 4 and keratin 13 expression in oral squamous cell carcinoma and epithelial dysplasia: a clue for histopathogenesis. Histopathol 58(4):531–542. , DOI 10.1111/j.1365-2559.2011.03759.x Hunt KK, Wingate H, Yokota T, Liu Y, Mills GB, Zhang F, Fang B, Su CH, Zhang M,Yi M, KeyomarsiK, 2013, Elafin, an inhibitor of elastase, is a prognostic indicator in breast cancer. Breast Cancer Res 15(1):R3. , DOI 10.1186/bcr3374 Labidi-Galy SI, Clauss A, Ng V, Duraisamy S, Elias KM, Piao HY, Bilal E, Davidowitz RA, Lu Y, Badalian-Very G, Gyorffy B, Kang UB, Ficarro S, Ganesan S, Mills GB, Marto JA, Drapkin R, 2015, Elafin drives poor outcome in high-grade serous ovarian cancers and basal-like breast tumors. Oncogene 34(3):373–383. https://doi. org/10.1038/onc.2013.562 Westin U, Nystrom M, Ljungcrantz I, Eriksson B, Ohlsson K, 2002, The presence of elafin, SLPI, IL1-RA and STNFalpha RI in head and neck squamous cell carcinomas and their relation to the degree of tumour differentiation. Mediators Inflamm 11(1):7–12. , DOI 10.1080/09629350210304 Tsuchida S, Bonkobara M, McMillan JR, Akiyama M, Yudate T, Aragane Y, Tezuka T, Shimizu H, Cruz PD, Ariizumi K, 2004, Characterization of Kdap, a protein secreted by keratinocytes. J Invest Dermatol 122(5):1225–1234. , DOI 10.1111/j. 0022-202X.2004.22511x Thibodeau BJ, Geddes TJ, Fortier LE, Ahmed S, Pruetz BL,Wobb J, Chen P, Wilson GD, Akervall JA, 2015, Gene Expression Characterization of HPV Positive Head and Neck Cancer to Predict Response to Chemoradiation. Head Neck Pathol 9(3): 345–353. , DOI 10.1007/s12105-014-0597-6 Probstmeier R, Kraus D, Wenghoefer M, Winter J, 2019, S100 Proteins as Biomarkers in Risk Estimations for Malignant Transformation in Oral Lesions. Methods Mol Biol 1929:763 – 71. , DOI 10.1007/978-1-4939-9030-6_48 Kesting MR, Sudhoff H, Hasler RJ, Nieberler M, Pautke C, Wolff KD, Wagenpfeil S, Al-Benna S, Jacobsen F, Steinstraesser L, 2009, Psoriasin, S100A7, up-regulation in oral squamous cell carcinoma and its relation to clinicopathologic features. Oral Oncol 45(8):731–6. , DOI 10.1016/j.oraloncology.2008.11.012 Kong F, Li Y, Hu E,Wang R, Wang J, Liu J, Zhang J, He D, Xiao X, 2016, The Characteristic of S100A7 Induction by the Hippo- YAP Pathway in Cervical and Glossopharyngeal Squamous Cell Carcinoma. PLoS One 11(12):e0167080. , DOI 10.1371/ journal.pone.0167080 Chen J, He QY, Yuen AP, Chiu JF, 2004, Proteomics of buccal squamous cell carcinoma: the involvement of multiple pathways in tumorigenesis. Proteomics 4(8):2465–2475. , DOI 10. 1002/pmic.200300762 Ren HZ, Pan GQ,Wang JS,Wen JF,Wang KS, Luo GQ, Shan XZ, 2010, Reduced stratifin expression can serve as an independent prognostic factor for poor survival in patients with esophageal squamous cell carcinoma. Dig Dis Sci 55(9):2552–2560. , DOI 10.1007/s10620-009-1065-0 Mirza S, Sharma G, Parshad R, Srivastava A, Gupta SD, Ralhan R, 2010, Clinical significance of Stratifin, ERalpha and PR promoter methylation in tumor and serum DNA in Indian breast cancer patients. Clin Biochem 43(4–5):380–386. , DOI 10.1016/j. clinbiochem.2009.11.016 Husni RE, Shiba-Ishii A, Nakagawa T, Dai T, Kim Y, Hong J, Sakashita S, Sakamoto N, Sato Y, Noguchi M, 2019, DNA hypomethylation-related overexpression of SFN, GORASP2 and ZYG11A is a novel prognostic biomarker for early stage lung adenocarcinoma. Oncotarget 10(17):1625–1636. , DOI 10. 18632/oncotarget.26676 Toyoshima T, Koch F, Kaemmerer P, Vairaktaris E, Al-Nawas B, Wagner W, 2009, Expression of cytokeratin 17 mRNA in oral squamous cell carcinoma cells obtained by brush biopsy: preliminary results. J Oral Pathol Med 38(6):530–534. , DOI 10. 1111/j.1600-0714.2009.00748x Kitamura R, Toyoshima T, Tanaka H, Kawano S, Kiyosue T, Matsubara R, Goto Y, Hirano M, Oobu K, Nakamura S, 2012, Association of cytokeratin 17 expression with differentiation in oral squamous cell carcinoma. J Cancer Res Clin 138(8):1299–1310. , DOI 10.1007/s00432-012-1202-6 Liu J, Liu L, Cao L, Wen Q, 2018, Keratin 17 promote lung adenocarcinoma progression by enhancing cell proliferation and invasion. Med Sci Monit 24:4782–4790. , DOI 10.12659/ MSM.909350 Wang YF, Lang HY, Yuan J,Wang J,Wang R, Zhang XH, Zhang J, Zhao T, Li YR, Liu JY, Zeng LH, Guo GZ, 2013, Overexpression of keratin 17 is associated with poor prognosis in epithelial ovarian cancer. Tumour Biol 34(3):1685–1689. https:// doi.org/10.1007/s13277-013-0703-5 Ricciardelli C, Lokman NA, Pyragius CE, Ween MP, Macpherson AM, Ruszkiewicz A, Hoffmann, Oehler MK, 2017, Keratin 5 overexpression is associated with serous ovarian cancer recurrence and chemotherapy resistance. Oncotarget 8(11):17819–17832. , DOI 10.18632/oncotarget.14867 Eckstein M,Wirtz MR, Gross-Weege M, Breyer J, Otto W, Stoehr R, Sikic D, Keck B, Eidt S, Burger M, Bolenz C, Nitschke K, Porubsky S, Hartmann A, Erben P, 2018, mRNA –expression of KRT5 andKRT10 defines distinct prognostic subgroups of muscleinvasive urothelial bladder cancer correlating with histological variants. Int J Mol Sci 19(11):3396. , DOI 10.3390/ ijms19113396 Liu YR, Yin PN, Silvers CR, Lee YF, 2019, Enhanced metastatic potential in the MB49 urothelial carcinoma model. Sci Rep 9(1): 7425. , DOI 10.1038/s41598-019-43641-5 Ohkura S, Kondoh N, Hada A, Arai M, Yamazaki Y, Sindoh M, Takahashi M, Matsumoto I, Yamamoto M, 2005, Differential expression of the keratin-4, -13, -14, -17 and transglutaminase 3 genes during the development of oral squamous cell carcinoma from leukoplakia. Oral Oncol 41(6):607–613. , DOI 10.1016/j. oraloncology.2005.01.011 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2315 EP 2325 DI 10.1007/s12253-020-00828-w PG 11 ER PT J AU Cheng, Z Shu, H Cui, Y Zhang, Q Zhao, B Pan, D Chao, Q Wang, D AF Cheng, Zhe Shu, Hansheng Cui, Ying Zhang, Qiujian Zhao, Biao Pan, Didi Chao, Qing Wang, Dawei TI MiR-424-5p Inhibits Proliferation, Invasion and Promotes Apoptosis and Predicts Good Prognosis in Glioma by Directly Targeting BFAR SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Glioma; Bifunctional apoptosis regulator; Prognosis; MicroRNA ID Glioma; Bifunctional apoptosis regulator; Prognosis; MicroRNA AB The biological function of miRNA (miR)-424-5p in glioma has not been clarified. This study was to explore the roles of miR- 424-5p/Bifunctional apoptosis regulator (BFAR) axis in glioma. Ninety-six pairs of human glioma tissues and their adjacent noncancer tissues were collected. The levels of BFAR and miR-424-5p were detected by quantitative polymerase chain reaction (qPCR) in glioma tissues and cell lines. Moreover, the biological roles of miR-424-5p and BFAR in glioma cells were assessed. We found a miR-424-5p binding site in the 3’UTR of BFAR by using TargetScan 7.2 online database. ThemiR-424-5p level was dramatically decreased in glioma tissues and cell lines, and the BFAR expression was significantly increased. The BFAR expression was negatively related to the miR-424-5p level in glioma tissues. Compared to patients with high miR-424-5p levels in glioma tissues, patients with low miR-424-5p levels had significantly lower survival rate (χ2 = 13.728 and P < 0.001). Compared to patients with high BFAR levels in glioma tissues, patients with low BFAR levels had significantly higher survival rate (χ2 = 5.516 and P = 0.027). Furthermore, up-regulation of miR-424-5p obviously restrained glioma cells proliferation and invasion, and promoted apoptosis. Besides, knockdown of BFAR also could markedly inhibit the proliferation and invasion, and promote apoptosis. Finally, overexpression of BFAR in glioma cells partially reversed the inhibited effects of miR-424-5p mimic. Knockdown of miR-424-5p restrained glioma cell apoptosis and promoted invasion and proliferation via regulation of BFAR. C1 [Cheng, Zhe] The Second Affiliated Hospital of Bengbu Medical College, Department of Neurosurgery, No. 220 Hongye Road, West of Longzi LakeBengbu, Anhui Province, China. [Shu, Hansheng] The Second Affiliated Hospital of Bengbu Medical College, Department of Neurosurgery, No. 220 Hongye Road, West of Longzi LakeBengbu, Anhui Province, China. [Cui, Ying] The Second Affiliated Hospital of Bengbu Medical College, Department of Neurosurgery, No. 220 Hongye Road, West of Longzi LakeBengbu, Anhui Province, China. [Zhang, Qiujian] The Second Affiliated Hospital of Bengbu Medical College, Department of Neurosurgery, No. 220 Hongye Road, West of Longzi LakeBengbu, Anhui Province, China. [Zhao, Biao] The Second Affiliated Hospital of Bengbu Medical College, Department of Neurosurgery, No. 220 Hongye Road, West of Longzi LakeBengbu, Anhui Province, China. [Pan, Didi] The Second Affiliated Hospital of Bengbu Medical College, Department of Neurosurgery, No. 220 Hongye Road, West of Longzi LakeBengbu, Anhui Province, China. [Chao, Qing] The Second Affiliated Hospital of Bengbu Medical College, Department of Neurosurgery, No. 220 Hongye Road, West of Longzi LakeBengbu, Anhui Province, China. [Wang, Dawei] The Second Affiliated Hospital of Bengbu Medical College, Department of Neurosurgery, No. 220 Hongye Road, West of Longzi LakeBengbu, Anhui Province, China. RP Wang, D (reprint author), The Second Affiliated Hospital of Bengbu Medical College, Department of Neurosurgery, Bengbu, China. EM yu_geng16@yeah.net CR Chen J, Wu X, Xing Z, Ma C, Xiong W, Zhu X, He X, 2018, FOXG1 expression is elevated in Glioma and inhibits Glioma cell apoptosis. J Cancer 9:778–783 Jansen E, Hamisch C, Ruess D, Heiland DH, Goldbrunner R, Ruge MI, Schnell O, Grau SJ, 2019, Observation after surgery for low grade glioma: long-term outcome in the light of the 2016 WHO classification. J Neuro-Oncol 145:501–507 Chapman CH, Hara JH, Molinaro AM, Clarke JL, Oberheim Bush NA, Taylor JW, Butowski NA, Chang SM, Fogh SE, Sneed PK, Nakamura JL, Raleigh DR, Braunstein SE, 2019, Reirradiation of recurrent high-grade glioma and development of prognostic scores for progression and survival. Neurooncol Pract 6:364–374 Gorbea C, Mosbruger T, Cazalla D, 2017, A viral Sm-class RNA base-pairs with mRNAs and recruits microRNAs to inhibit apoptosis. Nature 550:275–279 Khan AQ, Ahmed EI, Elareer NR, Junejo K, Steinhoff M, Uddin S, 2019, Role of miRNA-regulated Cancer stem cells in the pathogenesis of human malignancies. Cells 8:840 Qin WJ, Wang WP, Wang XB, Zhang XT, Du JD, 2019, MiR- 1290 targets CCNG2 to promote the metastasis of oral squamous cell carcinoma. Eur Rev Med Pharmacol Sci 23:10332–10342 Buonfiglioli A, Efe IE, Guneykaya D, Ivanov A, Huang Y, Orlowski E, Kruger C, Deisz RA, Markovic D, Fluh C, Newman AG, Schneider UC, Beule D, Wolf SA, Dzaye O, Gutmann DH, Semtner M, Kettenmann H, Lehnardt S, 2019, Let-7 MicroRNAs regulate microglial function and suppress Glioma growth through toll-like receptor 7. Cell Rep 29:3460–3471 Guo X, Zhang M, Li Q, Zhao J, Wang B, Wang J, Mao P, Song J, 2020, Evaluation of genetic variants in MIR3142HG in susceptibility to and prognosis of Glioma. Am J Clin Oncol 43:1–8 Wu H, Liu L, Zhu JM, 2019)MiR-93-5p inhibited proliferation and metastasis of glioma cells by targeting MMP2. Eur Rev Med Pharmacol Sci 23:9517–9524 Xu CH, Xiao LM, Zeng EM, Chen LK, Zheng SY, Li DH, Liu Y, 2019, MicroRNA-181 inhibits the proliferation, drug sensitivity and invasion of human glioma cells by targeting Selenoprotein K, SELK). Am J Transl Res 11:6632–6640 Hou L, Luo P, Ma Y, Jia C, Yu F, Lv Z, Wu C, Fu D, 2017, MicroRNA-125a-3p downregulation correlates with tumorigenesis and poor prognosis in patients with non-small cell lung cancer. Oncol Lett 14:4441–4448 Shao N,Wang L, Xue L,Wang R, Lan Q, 2015, Plasma miR-454- 3p as a potential prognostic indicator in human glioma. Neurol Sci 36:309–313 Wang G, Li Y, Li J, Zhang D, Luo C, Zhang B, Sun X, 2019, microRNA-199a-5p suppresses glioma progression by inhibiting MAGT1. J Cell Biochem 120:15248–15254 Kaminski A, Gupta KH, Goldufsky JW, Lee HW, Gupta V, Shafikhani SH, 2018, Pseudomonas aeruginosa ExoS induces intrinsic apoptosis in target host cells in amanner that is dependent on its GAP domain activity. Sci Rep 1:14047 Bianchini M, Levy E, Zucchini C, Pinski V, Macagno C, De Sanctis P, Valvassori L, Carinci P, Mordoh J, 2006, Comparative study of gene expression by cDNA microarray in human colorectal cancer tissues and normal mucosa. Int J Oncol 29:83–94 Barbagallo I, Parenti R, Zappala A, Vanella L, Tibullo D, Pepe F, Onni T, Li Volti G, 2015, Combined inhibition of Hsp90 and heme oxygenase-1 induces apoptosis and endoplasmic reticulum stress in melanoma. Acta Histochem 117:705–711 Lee B, Galli S, Tsokos M, 2007, Sensitive Ewing sarcoma and neuroblastoma cell lines have increased levels of BAD expression and decreased levels of BAR expression compared to resistant cell lines. Cancer Lett 247:110–114 Litak J, Mazurek M, Grochowski C, Kamieniak P, Rolinski J, 2019, PD-L1/PD-1 Axis in Glioblastoma Multiforme. Int J Mol Sci 20:5347 McCarthy BJ, Shibui S, Kayama T, Miyaoka E, Narita Y, Murakami M, Matsuda A, Matsuda T, Sobue T, Palis BE, Dolecek TA, Kruchko C, Engelhard HH, Villano JL, 2012, Primary CNS germ cell tumors in Japan and the United States: an analysis of 4 tumor registries. Neuro-Oncology 14:1194–1200 Johnson DR, O'Neill BP, 2012, Glioblastoma survival in the United States before and during the temozolomide era. J Neuro- Oncol 107:359–364 Yin F, Zhang JN, Wang SW, Zhou CH, Zhao MM, Fan WH, Fan M, Liu S, 2015, MiR-125a-3p regulates glioma apoptosis and invasion by regulating Nrg1. PLoS One 10:e0116759 Wu J, Yang B, Zhang Y, Feng X, He B, Xie H, Zhou L, Wu J, Zheng S, 2019, miR-424-5p represses the metastasis and invasion of intrahepatic cholangiocarcinoma by targeting ARK5. Int J Biol Sci 15:1591–1599 Sahami-FardMH, Kheirandish S, SheikhhaMH, 2019, Expression levels of miR-143-3p and −424-5p in colorectal cancer and their clinical significance. Cancer Biomark 24:291–297 Wang S, Zhang JH,Wang H, Yang L, Hong S, Yu B, Guo JC, Liu J, Zhu YB, 2019, A novel multidimensional signature predicts prognosis in hepatocellular carcinoma patients. J Cell Physiol 234: 11610–11619 Cha SY, Choi YH, Hwang S, Jeong JY, An HJ, 2017, Clinical impact of microRNAs associated with Cancer stem cells as a prognostic factor in ovarian carcinoma. J Cancer 8:3538–3547 Song ZB, Yang HP, Xu AQ, Zhan ZM, Song Y, Li ZY, 2020, Connective tissue growth factor as an unfavorable prognosticmarker promotes the proliferation, migration, and invasion of gliomas. Chin Med J 6:670–678 Zhang HY, Zhang BW, Zhang ZB, Deng QJ, 2020, Circular RNA TTBK2 regulates cell proliferation, invasion and ferroptosis via miR-761/ITGB8 axis in glioma. Eur Rev Med Pharmacol Sci 24: 2585–2600 Fan R, Wang H, Zhang L, Ma T, Tian Y, Li H, 2020, Nanocrystallized Oleanolic acid better inhibits proliferation,migration and invasion in intracranial Glioma via Caspase-3 pathway. J Cancer 11:1949–1958 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2327 EP 2335 DI 10.1007/s12253-020-00831-1 PG 9 ER PT J AU Bartu, M Hojny, J Hajkova, N Michalkova, R Krkavcova, E Simon, K Fryba, V Struzinska, I Nemejcova, K Dundr, P AF Bartu, Michaela Hojny, Jan Hajkova, Nikola Michalkova, Romana Krkavcova, Eva Simon, Karol Fryba, Vladimir Struzinska, Ivana Nemejcova, Kristyna Dundr, Pavel TI Expression, Epigenetic, and Genetic Changes of HNF1B in Colorectal Lesions: an Analysis of 145 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon; Adenoma; HNF-1-beta; Immunohistochemistry; Mutation analysis; Methylation ID Colon; Adenoma; HNF-1-beta; Immunohistochemistry; Mutation analysis; Methylation AB Hepatocyte nuclear factor 1 beta (HNF1B) is transcription factor which plays a crucial role in the regulation of the development of several organs, but also seems to be implicated in the development of certain tumours, especially the subset of clear cell carcinomas of the ovary and kidney. Depending on the type of the tumour, HNF1B may act as either a tumour suppressor or an oncogene, although the exact mechanism by which HNF1B participates in the process of cancerogenesis is unknown. Using immunohistochemical approach and methylation and mutation analysis, we have investigated the expression, epigenetic, and genetic changes of HNF1B on 40 cases of colorectal adenomas and 105 cases of colorectal carcinomas. The expression of HNF1B was correlated with the benign or malignant behaviour of the lesion, given that carcinomas showed significantly lower levels of expression compared to adenomas. In carcinomas, lower levels of HNF1B expression were associated with recurrence and shortened disease-free survival. Themutation analysis revealed three somaticmutations (two frameshift and one nonsense) in the carcinoma sample set. Promoter methylation was detected in three carcinomas. These results suggest that in colorectal cancer, HNF1B may play a part in the pathogenesis and act in a tumour suppressive fashion. C1 [Bartu, Michaela] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Hojny, Jan] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Hajkova, Nikola] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Michalkova, Romana] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Krkavcova, Eva] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Simon, Karol] Charles University, 1st Faculty of Medicine and General Teaching Hospital, 12800 Prague, Czech Republic. [Fryba, Vladimir] Charles University and General University Hospital, First Faculty of Medicine, 1st Department of Surgery - Department of Abdominal, Thoracic Surgery and Traumatology, 12808 Prague, Czech Republic. [Struzinska, Ivana] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Nemejcova, Kristyna] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. [Dundr, Pavel] Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, Studnickova 2, 12800 Prague, Czech Republic. RP Dundr, P (reprint author), Charles University, 1st Faculty of Medicine and General Teaching Hospital, Department of Pathology, 12800 Prague, Czech Republic. EM Pavel.Dundr@vfn.cz CR Liu Y, Kanyomse Q, Xie Y, 2019, Tumor-suppressive activity of Hnf1beta in Wilms’ tumor. Biosci Biotechnol Biochem 83(11): 2008–2015. , DOI 10.1080/09168451.2019.1611409 Yu DD, Guo SW, Jing YY, Dong YL,Wei LX, 2015, A review on hepatocyte nuclear factor-1beta and tumor. Cell Biosci 5:58. https:// doi.org/10.1186/s13578-015-0049-3 Carithers LJ, Ardlie K, Barcus M, Branton PA, Britton A, Buia SA, Compton CC, DS DL, Peter-Demchok J, Gelfand ET, Guan P, Korzeniewski GE, Lockhart NC, Rabiner CA, Rao AK, Robinson KL, Roche NV, Sawyer SJ, Segre AV, Shive CE, Smith AM, Sobin LH, Undale AH, Valentino KM, Vaught J, Young TR, Moore HM, Consortium GT, 2015, A novel approach to high-quality postmortem tissue procurement: the GTEx project. Biopreserv Biobank 13(5):311–319. , DOI 10.1089/bio.2015.0032 Pontoglio M, 2000, Hepatocyte nuclear factor 1, a transcription factor at the crossroads of glucose homeostasis. J Am Soc Nephrol 11(Suppl 16):S140–S143 Suzuki E, Kajita S, Takahashi H,Matsumoto T, Tsuruta T, Saegusa M, 2015, Transcriptional upregulation of HNF-1beta by NFkappaB in ovarian clear cell carcinoma modulates susceptibility to apoptosis through alteration in bcl-2 expression. Lab Investig 95(8): 962–972. , DOI 10.1038/labinvest.2015.73 Tsuchiya A, Sakamoto M, Yasuda J, Chuma M, Ohta T, Ohki M, Yasugi T, Taketani Y, Hirohashi S, 2003, Expression profiling in ovarian clear cell carcinoma: identification of hepatocyte nuclear factor-1 beta as a molecular marker and a possible molecular target for therapy of ovarian clear cell carcinoma. Am J Pathol 163(6): 2503–2512 Adalat S,Woolf AS, Johnstone KA,Wirsing A, Harries LW, Long DA, Hennekam RC, Ledermann SE, Rees L, van't Hoff W, Marks SD, Trompeter RS, Tullus K, Winyard PJ, Cansick J, Mushtaq I, Dhillon HK, Bingham C, Edghill EL, Shroff R, Stanescu H, Ryffel GU, Ellard S, Bockenhauer D, 2009, HNF1B mutations associate with hypomagnesemia and renal magnesium wasting. J Am Soc Nephrol 20(5):1123–1131. , DOI 10.1681/ASN. 2008060633 Anik A, Catli G, Abaci A, Bober E, 2015, Maturity-onset diabetes of the young, MODY): an update. J Pediatr Endocrinol Metab 28(3–4):251–263. , DOI 10.1515/jpem-2014-0384 Debiais-Delpech C, Godet J, Pedretti N, Bernard FX, Irani J, Cathelineau X, Cussenot O, Fromont G, 2014, Expression patterns of candidate susceptibility genes HNF1beta and CtBP2 in prostate cancer: association with tumor progression. Urol Oncol 32(4):426– 432. , DOI 10.1016/j.urolonc.2013.09.006 Elliott KS, Zeggini E, McCarthy MI, Gudmundsson J, Sulem P, Stacey SN, Thorlacius S, Amundadottir L, Gronberg H, Xu J, Gaborieau V, Eeles RA, Neal DE, Donovan JL, Hamdy FC, Muir K, Hwang SJ, Spitz MR, Zanke B, Carvajal-Carmona L, Brown KM, Australian Melanoma Family Study I, Hayward NK, Macgregor S, Tomlinson IP, Lemire M, Amos CI, Murabito JM, Isaacs WB, Easton DF, Brennan P, PanScan C, Barkardottir RB, Gudbjartsson DF, Rafnar T, Hunter DJ, Chanock SJ, Stefansson K, Ioannidis JP, 2010, Evaluation of association of HNF1B variants with diverse cancers: collaborative analysis of data from 19 genome-wide association studies. PLoS One 5(5):e10858. https:// doi.org/10.1371/journal.pone.0010858 Glinsky GV, Glinskii AB, Stephenson AJ, Hoffman RM, Gerald WL, 2004, Gene expression profiling predicts clinical outcome of prostate cancer. J Clin Invest 113(6):913–923. , DOI 10. 1172/JCI20032 Janky R, Binda MM, Allemeersch J, Van den Broeck A, Govaere O, Swinnen JV, Roskams T, Aerts S, Topal B, 2016, Prognostic relevance ofmolecular subtypes and master regulators in pancreatic ductal adenocarcinoma. BMC Cancer 16:632. , DOI 10. 1186/s12885-016-2540-6 Kondratyeva LG, Chernov IP, Zinovyeva MV, Kopantzev EP, Sverdlov ED, 2017, Expression of master regulatory genes of embryonic development in pancreatic tumors. Dokl Biochem Biophys 475(1):250–252. , DOI 10.1134/S1607672917040020 Spurdle AB, Thompson DJ, Ahmed S, Ferguson K, Healey CS, O'Mara T, Walker LC, Montgomery SB, Dermitzakis ET, Australian National Endometrial Cancer Study G, Fahey P, Montgomery GW, Webb PM, Fasching PA, Beckmann MW, Ekici AB, Hein A, Lambrechts D, Coenegrachts L, Vergote I, Amant F, Salvesen HB, Trovik J, Njolstad TS, Helland H, Scott RJ, Ashton K, Proietto T, Otton G, National Study of Endometrial Cancer Genetics G, Tomlinson I, GormanM, Howarth K, Hodgson S, Garcia-Closas M, Wentzensen N, Yang H, Chanock S, Hall P, Czene K, Liu J, Li J, Shu XO, Zheng W, Long J, Xiang YB, Shah M, Morrison J, Michailidou K, Pharoah PD, Dunning AM, Easton DF, 2011, Genome-wide association study identifies a common variant associated with risk of endometrial cancer. Nat Genet 43(5):451–454. , DOI 10.1038/ng.812 Yamamoto S, Tsuda H, Aida S, Shimazaki H, Tamai S, Matsubara O, 2007, Immunohistochemical detection of hepatocyte nuclear factor 1beta in ovarian and endometrial clear-cell adenocarcinomas and nonneoplastic endometrium. Hum Pathol 38(7):1074–1080. , DOI 10.1016/j.humpath.2006.12.018 Rebouissou S, Vasiliu V, Thomas C, Bellanne-Chantelot C, Bui H, Chretien Y, Timsit J, Rosty C, Laurent-Puig P, Chauveau D, Zucman-Rossi J, 2005, Germline hepatocyte nuclear factor 1alpha and 1beta mutations in renal cell carcinomas. Hum Mol Genet 14(5):603–614. , DOI 10.1093/hmg/ddi057 Painter JN, O'Mara TA, Batra J, Cheng T, Lose FA, Dennis J, Michailidou K, Tyrer JP, Ahmed S, Ferguson K, Healey CS, Kaufmann S, Hillman KM, Walpole C, Moya L, Pollock P, Jones A, HowarthK,Martin L,GormanM,Hodgson S, National Study of Endometrial Cancer Genetics G, Consortium C, De Polanco MM, Sans M, Carracedo A, Castellvi-Bel S, Rojas-Martinez A, Santos E, Teixeira MR, Carvajal-Carmona L, Shu XO, Long J, Zheng W, Xiang YB, Australian National Endometrial Cancer Study G, Montgomery GW, Webb PM, Scott RJ, McEvoy M, Attia J, Holliday E, Martin NG, Nyholt DR, Henders AK, Fasching PA, Hein A, Beckmann MW, Renner SP, Dork T, Hillemanns P, Durst M, Runnebaum I, Lambrechts D, Coenegrachts L, Schrauwen S, Amant F, Winterhoff B, Dowdy SC, Goode EL, Teoman A, Salvesen HB, Trovik J, Njolstad TS, Werner HM, Ashton K, Proietto T, Otton G, Tzortzatos G, Mints M, Tham E, Rendocas HP, Czene K, Liu J, Li J, Hopper JL, Southey MC, Australian Ovarian Cancer S, Ekici AB, Ruebner M, Johnson N, Peto J, Burwinkel B, Marme F, Brenner H, Dieffenbach AK, Meindl A, Brauch H, Network G, Lindblom A, Depreeuw J, Moisse M, Chang-Claude J, Rudolph A, Couch FJ, Olson JE, Giles GG, Bruinsma F, Cunningham JM, Fridley BL, Borresen-Dale AL, Kristensen VN, Cox A, Swerdlow AJ, Orr N, Bolla MK, Wang Q, Weber RP, Chen Z, Shah M, French JD, Pharoah PD, Dunning AM, Tomlinson I, Easton DF, Edwards SL, Thompson DJ, Spurdle AB, 2015, Fine-mapping of the HNF1B multicancer locus identifies candidate variants that mediate endometrial cancer risk. Hum Mol Genet 24(5):1478–1492. , DOI 10.1093/ hmg/ddu552 Setiawan VW, Haessler J, Schumacher F, Cote ML, Deelman E, FesinmeyerMD, Henderson BE, Jackson RD, Vockler JS,Wilkens LR, Yasmeen S, Haiman CA, Peters U, Le Marchand L, Kooperberg C, 2012, HNF1B and endometrial cancer risk: results from the PAGE study. PLoS One 7(1):e30390. , DOI 10. 1371/journal.pone.0030390 Berndt SI, Sampson J, YeagerM, Jacobs KB,Wang Z, Hutchinson A, Chung C, Orr N, Wacholder S, Chatterjee N, Yu K, Kraft P, Feigelson HS, Thun MJ, Diver WR, Albanes D, Virtamo J, Weinstein S, Schumacher FR, Cancel-Tassin G, Cussenot O, Valeri A, Andriole GL, Crawford ED, Haiman C, Henderson B, Kolonel L, Le Marchand L, Siddiq A, Riboli E, Travis RC, Kaaks R, IsaacsW, Isaacs S,Wiley KE,Gronberg H, Wiklund F, Stattin P, Xu J, Zheng SL, Sun J, Vatten LJ, Hveem K, Njolstad I, Gerhard DS, TuckerM, Hayes RB, Hoover RN, Fraumeni JF Jr, Hunter DJ, Thomas G, Chanock SJ, 2011, Large-scale fine mapping of the HNF1B locus and prostate cancer risk. Hum Mol Genet 20(16): 3322–3329. , DOI 10.1093/hmg/ddr213 Hindorff LA, Gillanders EM,Manolio TA, 2011, Genetic architecture of cancer and other complex diseases: lessons learned and future directions. Carcinogenesis 32(7):945–954. , DOI 10.1093/carcin/bgr056 Ross-Adams H, Ball S, Lawrenson K, Halim S, Russell R,Wells C, Strand SH, Orntoft TF, LarsonM, Armasu S,Massie CE, Asim M, Mortensen MM, Borre M, Woodfine K, Warren AY, Lamb AD, Kay J, Whitaker H, Ramos-Montoya A, Murrell A, Sorensen KD, Fridley BL, Goode EL, Gayther SA, Masters J, Neal DE, Mills IG, 2016, HNF1B variants associate with promoter methylation and regulate gene networks activated in prostate and ovarian cancer. Oncotarget 7(46):74734–74746. , DOI 10.18632/ oncotarget.12543 Matsui A, Fujimoto J, Ishikawa K, Ito E, Goshima N,Watanabe S, Semba K, 2016, Hepatocyte nuclear factor 1 beta induces transformation and epithelial-to-mesenchymal transition. FEBS Lett 590(8):1211–1221. , DOI 10.1002/1873-3468.12147 Bosman FT, World Health Organization., International Agency for Research on Cancer, 2010, WHO classification of tumours of the digestive system. World Health Organization classification of tumours, vol 3, 4th edn. International Agency for Research on Cancer, Lyon Specht E, Kaemmerer D, Sanger J, Wirtz RM, Schulz S, Lupp A, 2015, Comparison of immunoreactive score, HER2/neu score and H score for the immunohistochemical evaluation of somatostatin receptors in bronchopulmonary neuroendocrine neoplasms. Histopathology 67(3):368–377. , DOI 10.1111/his.12662 Gregova M, Hojny J,Nemejcova K, BartuM,MaraM, Boudova B, Laco J, Krbal L, Ticha I, Dundr P, 2019, Leiomyoma with bizarre nuclei: a study of 108 cases focusing on clinicopathological features, morphology, and fumarate hydratase alterations. Pathol Oncol Res. , DOI 10.1007/s12253-019-00739-5 Ticha I, Hojny J, Michalkova R, Kodet O, Krkavcova E, Hajkova N, Nemejcova K, Bartu M, Jaksa R, Dura M, Kanwal M, Martinikova AS, Macurek L, Zemankova P, Kleibl Z, Dundr P, 2019, A comprehensive evaluation of pathogenic mutations in primary cutaneous melanomas, including the identification of novel loss-of-function variants. Sci Rep 9(1):17050. , DOI 10. 1038/s41598-019-53636-x Ticha I, Hojny J, Michalkova R, Kodet O, Krkavcova E, Hajkova N, Nemejcova K, Bartu M, Jaksa R, Dura M, Kanwal M, Martinikova AS, L. Macurek, Zemankova P, Kleibl Z, Dundr P, 2019, A comprehensive evaluation of pathogenic mutations in primary cutaneous melanomas, including the identification of novel loss-of-function variants. Sci Rep Accepted for publication Suraweera N, Duval A, Reperant M, Vaury C, Furlan D, Leroy K, Seruca R, Iacopetta B, Hamelin R, 2002, Evaluation of tumor microsatellite instability using five quasimonomorphic mononucleotide repeats and pentaplex PCR. Gastroenterology 123(6):1804– 1811. , DOI 10.1053/gast.2002.37070 Shen H, Fridley BL, Song H, Lawrenson K, Cunningham JM, Ramus SJ, Cicek MS, Tyrer J, Stram D, Larson MC, Kobel M, Consortium P, Ziogas A, Zheng W, Yang HP, Wu AH, Wozniak EL, Woo YL, Winterhoff B, Wik E, Whittemore AS, Wentzensen N,Weber RP, Vitonis AF, Vincent D, Vierkant RA, Vergote I, Van Den Berg D, Van AltenaAM, Tworoger SS, Thompson PJ, Tessier DC, Terry KL, Teo SH, Templeman C, Stram DO, Southey MC, Sieh W, Siddiqui N, Shvetsov YB, Shu XO, Shridhar V, Wang- Gohrke S, Severi G, Schwaab I, Salvesen HB, Rzepecka IK, Runnebaum IB, Rossing MA, Rodriguez-Rodriguez L, Risch HA, Renner SP, Poole EM, Pike MC, Phelan CM, Pelttari LM, Pejovic T, Paul J, Orlow I, Omar SZ, Olson SH, Odunsi K, Nickels S, Nevanlinna H, Ness RB, Narod SA, Nakanishi T, Moysich KB, Monteiro AN, Moes-Sosnowska J, Modugno F, Menon U, McLaughlin JR, McGuire V, Matsuo K, Adenan NA, Massuger LF, Lurie G, Lundvall L, Lubinski J, Lissowska J, Levine DA, Leminen A, Lee AW, Le ND, Lambrechts S, Lambrechts D, Kupryjanczyk J, Krakstad C, Konecny GE, Kjaer SK, Kiemeney LA, Kelemen LE, Keeney GL, Karlan BY, Karevan R, Kalli KR, Kajiyama H, Ji BT, Jensen A, Jakubowska A, Iversen E, Hosono S, Hogdall CK, Hogdall E, Hoatlin M, Hillemanns P, Heitz F, Hein R, Harter P, Halle MK, Hall P, Gronwald J, Gore M, Goodman MT, Giles GG, Gentry-Maharaj A, Garcia-Closas M, Flanagan JM, Fasching PA, Ekici AB, Edwards R, Eccles D, Easton DF, Durst M, du Bois A, Dork T, Doherty JA, Despierre E, Dansonka- Mieszkowska A, Cybulski C, Cramer DW, Cook LS, Chen X, Charbonneau B, Chang-Claude J, Campbell I, Butzow R, Bunker CH, Brueggmann D, Brown R, Brooks-Wilson A, Brinton LA, Bogdanova N, Block MS, Benjamin E, Beesley J, Beckmann MW, Bandera EV, Baglietto L, Bacot F, Armasu SM, Antonenkova N, Anton-Culver H, Aben KK, Liang D, Wu X, Lu K, Hildebrandt MA, Australian Ovarian Cancer Study G, Australian Cancer S, Schildkraut JM, Sellers TA, Huntsman D, Berchuck A, Chenevix-Trench G, Gayther SA, Pharoah PD, Laird PW, Goode EL, Pearce CL, 2013, Epigenetic analysis leads to identification of HNF1B as a subtype-specific susceptibility gene for ovarian cancer. Nat Commun 4:1628. , DOI 10.1038/ ncomms2629 Wojdacz TK, Dobrovic A, Hansen LL, 2008, Methylationsensitive high-resolution melting. Nat Protoc 3(12):1903–1908. , DOI 10.1038/nprot.2008.191 Lau HH, Ng NHJ, Loo LSW, Jasmen JB, Teo AKK, 2018, The molecular functions of hepatocyte nuclear factors - in and beyond the liver. J Hepatol 68(5):1033–1048. , DOI 10.1016/j. jhep.2017.11.026 Rezanejad H, Ouziel-Yahalom L, Keyzer CA, Sullivan BA, Hollister-Lock J, Li WC, Guo L, Deng S, Lei J, Markmann J, Bonner-Weir S, 2018, Heterogeneity of SOX9 and HNF1beta in pancreatic ducts is dynamic. Stem Cell Reports 10(3):725–738. , DOI 10.1016/j.stemcr.2018.01.028 Senkel S, Lucas B, Klein-Hitpass L, Ryffel GU, 2005, Identification of target genes of the transcription factor HNF1beta and HNF1alpha in a human embryonic kidney cell line. Biochim Biophys Acta 1731(3):179–190. , DOI 10.1016/j.bbaexp. 2005.10.003 Kobel M, Kalloger SE, Carrick J, Huntsman D, Asad H, Oliva E, Ewanowich CA, Soslow RA, Gilks CB, 2009, A limited panel of immunomarkers can reliably distinguish between clear cell and high-grade serous carcinoma of the ovary. Am J Surg Pathol 33(1):14–21. , DOI 10.1097/PAS.0b013e3181788546 LebrunG,Vasiliu V, Bellanne-Chantelot C, Bensman A,Ulinski T, Chretien Y, Grunfeld JP, 2005, Cystic kidney disease, chromophobe renal cell carcinoma and TCF2, HNF1 beta, mutations. Nat Clin Pract Nephrol 1(2):115–119. , DOI 10.1038/ ncpneph0054 Nemejcova K, Cibula D, Dundr P, 2015, Expression of HNF-1beta in cervical carcinomas: an immunohistochemical study of 155 cases. Diagn Pathol 10:8. , DOI 10.1186/s13000-015- 0245-9 Bartu M, Dundr P, Nemejcova K, Ticha I, Hojny H, Hajkova N, 2018, The role of HNF1B in tumorigenesis of solid Tumours: a review of current knowledge. Folia Biol, Praha, 64(3):71–83 Harries LW, Perry JR, McCullagh P, Crundwell M, 2010, Alterations in LMTK2, MSMB and HNF1B gene expression are associated with the development of prostate cancer. BMC Cancer 10:315. , DOI 10.1186/1471-2407-10-315 Kim HJ, Bae JS, Lee J, Chang IH, Kim KD, Shin HD, Han JH, Lee SY, Kim W, Myung SC, 2011, HNF1B polymorphism associated with development of prostate cancer in Korean patients. Urology 78(4):969 e961–969 e966. , DOI 10.1016/j.urology.2011. 06.045 Mandato VD, Farnetti E, Torricelli F, Abrate M, Casali B, Ciarlini G, Pirillo D, Gelli MC, Nicoli D, Grassi M, LAS GB, Palomba S, 2015, HNF1B polymorphisminfluences the prognosis of endometrial cancer patients: a cohort study. BMC Cancer 15:229. https:// doi.org/10.1186/s12885-015-1246-5 Kountourakis P, Souglakos J, Gouvas N, Androulakis N, Athanasiadis A, Boukovinas I, Christodoulou C, Chrysou E, Dervenis C, Emmanouilidis C, Georgiou P, Karachaliou N, Katopodi O, Makatsoris T, Papakostas P, Pentheroudakis G, Pilpilidis I, Sgouros J, Tekkis P, Triantopoulou C, Tzardi M, Vassiliou V, Vini L, Xynogalos S, Xynos E, Ziras N, Papamichael D, 2016, Adjuvant chemotherapy for colon cancer: a consensus statement of the Hellenic and Cypriot Colorectal Cancer Study Group by the HeSMO. Ann Gastroenterol 29(1): 18–23 Silva TD, Vidigal VM, Felipe AV, DEL JM, Neto RA, Saad SS, Forones NM, 2013, DNA methylation as an epigenetic biomarker in colorectal cancer. Oncol Lett 6(6):1687–1692. , DOI 10. 3892/ol.2013.1606 Terasawa K, Toyota M, Sagae S, Ogi K, Suzuki H, Sonoda T, Akino K, Maruyama R, Nishikawa N, Imai K, Shinomura Y, Saito T, Tokino T, 2006, Epigenetic inactivation of TCF2 in ovarian cancer and various cancer cell lines. Br J Cancer 94(6):914–921. , DOI 10.1038/sj.bjc.6602984 Suaud L, Joseph B, Formstecher P, Laine B, 1997, mRNA expression of HNF-4 isoforms and of HNF-1alpha/HNF-1beta variants and differentiation of human cell lines that mimic highly specialized phenotypes of intestinal epithelium. Biochem Biophys Res Commun 235(3):820–825. , DOI 10.1006/bbrc.1997.6888 Yang R, Kerschner JL, Harris A, 2016, Hepatocyte nuclear factor 1 coordinatesmultiple processes in amodel of intestinal epithelial cell function. Biochim Biophys Acta 1859(4):591–598. , DOI 10.1016/j.bbagrm.2016.02.005 D'Angelo A, Bluteau O, Garcia-Gonzalez MA, Gresh L, Doyen A, Garbay S, Robine S, PontoglioM(2010, Hepatocyte nuclear factor 1alpha and beta control terminal differentiation and cell fate commitment in the gut epithelium. Development 137(9):1573–1582. , DOI 10.1242/dev.044420 Amano Y, Mandai M, Yamaguchi K, Matsumura N, Kharma B, Baba T, Abiko K, Hamanishi J, Yoshioka Y, Konishi I, 2015, Metabolic alterations caused by HNF1beta expression in ovarian clear cell carcinoma contribute to cell survival. Oncotarget 6(28): 26002–26017. , DOI 10.18632/oncotarget.4692 Rougemont AL, Tille JC, 2018, Role of HNF1beta in the differential diagnosis of yolk sac tumor from other germ cell tumors. Hum Pathol 81:26–36. , DOI 10.1016/j.humpath.2018.04.025 Zheng J, Liu X, Xue Y, Gong W,Ma J, Xi Z, Que Z, Liu Y, 2017, TTBK2 circular RNA promotes glioma malignancy by regulating miR-217/HNF1beta/Derlin-1 pathway. J Hematol Oncol 10(1):52. , DOI 10.1186/s13045-017-0422-2 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2337 EP 2350 DI 10.1007/s12253-020-00830-2 PG 14 ER PT J AU Khatib, G Gulec, KU Guzel, BA Bagir, E Paydas, S Vardar, AM AF Khatib, Ghanim Gulec, Kucukgoz Umran Guzel, Baris Ahmet Bagir, Emine Paydas, Semra Vardar, Ali Mehmet TI Prognosis Trend of Grade 2 Endometrioid Endometrial Carcinoma: Toward Grade 1 or 3? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endometrioid endometrial carcinoma; FIGOgrade; Oncologic outcomes; Interobserver reproducibility ID Endometrioid endometrial carcinoma; FIGOgrade; Oncologic outcomes; Interobserver reproducibility AB Although the prognostic significance of grade in endometrial cancer is well known, grade 2 cases have not been evaluated separately in most of the previous studies. In this study, we aim to investigate whether the oncologic outcomes of grade 2 endometrioid endometrial carcinomas trend towards grade 1 or 3 tumors. Patients’ records and pathological reports were reviewed retrospectively and eligible patients with endometrioid endometrial carcinoma were determined and distributed into 3 groups according to their 1988 International Federation of Gynecology and Obstetrics (FIGO) grade. Groups’ characteristics and oncologic outcomes were compared. Differences between grades were tested with z-test and adjusted by Bonferroni method. Kaplan–Meier method was performed for the survival analysis. In total, 776 patients of endometrioid endometrial carcinoma were included in this study. Mean follow-up time was 52 ± 14 months. Patients’ mean age was 56.3 ± 10.8 years. Even though grade 2 endometrioid endometrial carcinomas were different from both grade 1 and 3 in terms of the pathological features, survival analyses demonstrated that their oncologic outcomes trended towards grade 1. The grade was determined as an independent prognostic factor for overall survival (OS). The interobserver reproducibility will be improved among pathologists by combining FIGO grade 1 and 2 endometrioid endometrial carcinomas, while prognosis prediction is not likely to be affected. C1 [Khatib, Ghanim] Cukurova University, Faculty of Medicine, Department of Obstetrics and Gynecology, 01330 Adana, Turkey. [Gulec, Kucukgoz Umran] Cukurova University, Faculty of Medicine, Department of Obstetrics and Gynecology, 01330 Adana, Turkey. [Guzel, Baris Ahmet] Cukurova University, Faculty of Medicine, Department of Obstetrics and Gynecology, 01330 Adana, Turkey. [Bagir, Emine] Cukurova University, School of Medicine, Pathology Department, 01330 Adana, Turkey. [Paydas, Semra] Cukurova University, Faculty of Medicine, Department of Medical Oncology, 01330 Adana, Turkey. [Vardar, Ali Mehmet] Cukurova University, Faculty of Medicine, Department of Obstetrics and Gynecology, 01330 Adana, Turkey. RP Khatib, G (reprint author), Cukurova University, Faculty of Medicine, Department of Obstetrics and Gynecology, 01330 Adana, Turkey. EM ghanim.khatib@gmail.com CR Clarke BA, Gilks CB, 2010, Endometrial carcinoma: controversies in histopathological assessment of grade and tumour cell type. J Clin Pathol 63:410–415 Tanaka K, Kobayashi Y, Sugiyama J et al, 2017, Histologic grade and peritoneal cytology as prognostic factors in type 1 endometrial cancer. Int J Clin Oncol 22:533–540 Alkushi A, Abdul-Rahman ZH, Lim P et al, 2005, Description of a novel system for grading of endometrial carcinoma and comparison with existing grading systems. Am J Surg Pathol 29:295–304 Scholten AN, Creutzberg CL, Noordijk EM, SmitVT, 2002, Longterm outcome in endometrial carcinoma favors a two- instead of a three-tiered grading system. Int J Radiat Oncol Biol Phys 52:1067– 1074 Bilgin T, Ozuysal S, Ozan H, 2005, A comparison of three histological grading systems in endometrial cancer. Arch Gynecol Obstet 272:23–25 Colombo N, Creutzberg C, Aman t Fet al., 2016, ESMO-ESGOESTRO consensus conference on endometrial Cancer: diagnosis, treatment and follow-up. Int J Gynecol Cancer 26:2–30 Jones HW 3rd, 1999, The importance of grading in endometrial cancer. Gynecol Oncol 74:1–2 Creasman WT, Odicino F, Maisonneuve P et al, 2006, Carcinoma of the corpus uteri. FIGO 26th annual report on the results of treatment in gynecological Cancer. Int J Gynaecol Obstet 95(Suppl 1): S105–S143 Binder PS, Mutch DG, 2014, Update on prognostic markers for endometrial cancer. Womens Health, Lond, 10:277–288 Lax SF, Kurman RJ, Pizer ES et al, 2000, A binary architectural grading systemfor uterine endometrial endometrioid carcinoma has superior reproducibility compared with FIGO grading and identifies subsets of advance-stage tumors with favorable and unfavorable prognosis. Am J Surg Pathol 24:1201–1208 Scholten AN, Smit VT, Beerman H et al, 2004, Prognostic significance and interobserver variability of histologic grading systems for endometrial carcinoma. Cancer 100:764–772 Creutzberg CL, van PuttenWL, Koper PC et al, 2000, Surgery and postoperative radiotherapy versus surgery alone for patients with stage-1 endometrial carcinoma: multicentre randomised trial. PORTEC study group. Post operative radiation therapy in endometrial carcinoma. Lancet 355:1404–1411 Taylor RR, Zeller J, Lieberman RW, O'Connor DM, 1999, An analysis of two versus three grades for endometrial carcinoma. Gynecol Oncol 74:3–6 Gemer O, Uriev L, VoldarskyMet al, 2009, The reproducibility of histological parameters employed in the novel binary grading systems of endometrial cancer. Eur J Surg Oncol 35:247–251 Kapucuoglu N, Bulbul D, Tulunay G, Temel MA, 2008, Reproducibility of grading systems for endometrial endometrioid carcinoma and their relationwith pathologic prognostic parameters. Int J Gynecol Cancer 18:790–796 Zaino RJ, Silverberg SG, Norris HJ, Bundy BN, Morrow CP, Okagaki T, 1994, The prognostic value of nuclear versus architectural grading in endometrial adenocarcinoma: a gynecologic oncology group study. Int J Gynecol Pathol 13:29–36 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2351 EP 2356 DI 10.1007/s12253-020-00836-w PG 6 ER PT J AU Kravchick, S Cherniavsky, E Peled, R Cytron, Sh Verhovsky, G AF Kravchick, Sergey Cherniavsky, Eugenia Peled, Ronit Cytron, Shmuel Verhovsky, Guy TI Power Doppler Sonography (PDS) and Modified TRUS Systematic Biopsies – Can this Combination Adequately ReplaceMultiparametric Prostate Magnetic Resonance Imaging (mp-MRI) in Candidates for Re Biopsies Who cannot Undergo mp-MRI SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Targeted biopsy; Diagnostic; Imaging ID Prostate cancer; Targeted biopsy; Diagnostic; Imaging AB The MRI targeted biopsy (MRI-TBx) may increase the detection rate of clinically significant cancer (csPCa) in candidates for rebiopsy. However, there will be several patients in whom MRI is contraindicated. In this retrospective study we assessed the ability of combination of PDS guided biopsies (PDS-TBx) and modified SBx to substitute MRI-TBx. 154 men with persistently elevated PSA were referred for re-biopsy. Our protocol included a combination of MRI-TBx, DPS-TBx and modified SBx with additional biopsies from anterior lateral horns and anterior aspects of apex. MRI findings were defined as suspicious lesions (MRI-SL) and highly suspicious lesions (MRI-HL), based on PIRADS scale. In 40 patients csPCa was detected.While,MRI diagnosed csPCa in 36 patients (23%, n-36/154): 25% and 92% of biopsies targeted to theMRI- SL and MRI-HSL confirmed csPCa. Thirty-eight PDS hypervascular areas were found, while csPCa was diagnosed in 84% of these lesions, or in 28 patients (18%, n-28/154). SBx detected csPCa in 34 cores or in 21 patients (13%, n – 21/154). SBx missed cancers in the in the anterior aspect of middle gland. Combination of PDSTBx + SBx detected csPCa in 35 (88% of csPCa) patients. Strongest predictors for the csPCa presence were MRI-HSL, PDS’ lesions and biopsies from anterior aspect that included apex, mid gland and anterior lateral horns (p < 0.001 and p-0.008, respectively). The combination of PDS-TBx + SBx may miss 15% of csPCa detected by MRI. However, it can detect additional 10%of csPCa that were missed by MRI. To improve the accuracy of this combination, the anterior aspect of middle gland should be also included in the modified SBx. These changes in combination can make it helpful in candidates for re-biopsy who cannot undergo MRI. C1 [Kravchick, Sergey] Upstate Medical University, Department of UrologySyracuse, NY, USA. [Cherniavsky, Eugenia] Barzilay Medical Center, Department of RadiologyAshkelon, Israel. [Peled, Ronit] Ben Gurion University, Epidemiology and StatisticsBeer-Sheva, Israel. [Cytron, Shmuel] Barzialy Medical Center, Urology DepartmentAshkelon, Israel. [Verhovsky, Guy] Hasharon Hospital, Rabin Medical Center, Petah-Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Department of UrologyTel Aviv, Israel. RP Verhovsky, G (reprint author), Hasharon Hospital, Rabin Medical Center, Petah-Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Department of Urology, Tel Aviv, Israel. EM guy.verchovsky@gmail.com CR Welch HG, Fisher ES, Gottlieb DJ, Barry MJ, 2007, Detection of prostate cancer via biopsy in the Medicare–SEER population during the PSA era. J Natl Cancer Inst 99:1395–1400 Moore CM, Robertson NL, Arsanious N, Middleton T, Villers A, Klotz L, Taneja SS, Emberton M, 2013, Image-guided prostate biopsy using magnetic resonance imaging-derived targets: a systematic review. Eur Urol 63:125–140 Sonn GA, Chang E, Natarajan S et al, 2014, Value of targeted prostate biopsy using magnetic resonance-ultrasound fusion in men with prior negative biopsy and elevated prostate-specific antigen. Eur Urol 65:809–815 Wysock JS, Rosenkrantz AB, Huang WC et al, 2013, A prospective, blinded comparison of magnetic resonance, MR, imagingultrasound fusion and visual estimation in the performance of MR-targeted prostate biopsy: the PROFUS trial. Eur Urol 66: 343–351 Salami SS, Ben-Levi E, YaskivO, Ryniker L, Turkbey B,Kavoussi LR, Villani R, Rastinehad AR, 2015, In patients with a previous negative prostate biopsy and a suspicious lesion on magnetic resonance imaging, is a 12-core biopsy still necessary in addition to a targeted biopsy? BJU Int 115:562–570 Schoots IG, Roobol MJ, Nieboer D, Chris H, Bangma CH, Steyerberg EW, Hunink MG, 2015, Magnetic resonance imaging–targeted biopsy may enhance the diagnostic accuracy of significant prostate cancer detection compared to standard transrectal ultrasound-guided biopsy: a systematic review and meta- analysis. Eur Urol 68:438–450 Le JD, Stephenson S, Brugger M et al, 2014, MRI-ultrasound fusion biopsy for prediction of final prostate pathology. J Urol 192: 1367–1373 Mathur S, O'MalleyME,Ghai S, Jhaveri K, Sreeharsha B,Margolis M, Zhong L, Maan H, Toi A, 2019, Correlation of 3T multiparametric prostate MRI using prostate imaging reporting and data system, PIRADS, version 2 with biopsy as reference standard. Abdom Radiol, NY, 44(1):252–258 Puech P, Rouviere O, Renard-Penna R et al, 2013, Prostate cancer diagnosis: multiparametric MR-targeted biopsy with cognitive and transrectal US-MR fusion guidance versus systematic biopsyprospective multicenter study. Radiology 268:461–469 Dewey M, Schink T, Dewey CF, 2007, Frequency of referral of patients with safety-related contraindications tomagnetic resonance imaging. Eur J Radiol 63(1):124–127 Chow GV, Nazarian S, 2014, MRI for patients with cardiac implantable electrical devices. Cardiol Clin 32:299–304 Bastide C, Lechevallier E, Eghazarian C, Ortega JC, Coulange C, 2003, Tolerance of pain during transrectal ultrasound-guided biopsy of the prostate: risk factors. Prostate Cancer Prostatic Dis 6: 239–241 Kravchick S, Yoffe B, Cytron S, 2007, Modified perianal/ pericapsular anesthesia for transrectal biopsy of prostate in patients with anal rectal problems. Urology 69(1):139–141 Gold SA, Hale GR, Bloom JB, Smith CP, Rayn KN, Valera V, Wood BJ, Choyke PL, Turkbey B, Pinto PA, 2019, Follow-up of negative MRI-targeted prostate biopsies: when are we missing cancer? World J Urol 37(2):235–241 Takahashi S, Yamada Y, Homma Y, Horie S, Hosaka Y, Kitamura T, 2002, Power Doppler ultrasonography-directed prostate biopsy in men with elevated serum PSA levels: an evaluation of the clinical utility and limitations. Urology. 60(2):248–252 Eisenberg ML, Cowan JE, Carroll PR, Shinohara K, 2010, The adjunctive use of power Doppler imaging in the preoperative assessment of prostate cancer. BJU Int 105:1237–1241 Remzi M, DobrovitsM, Reissigl A, Ravery V,WaldertM,Wiunig C, Fong YK, Djavan B, European Society for Oncological Urology, ESOU,, 2004, Can power Doppler enhanced transrectal ultrasound guided biopsy improve prostate cancer detection on first and repeat prostate biopsy? Eur Urol 46(4):451–456 Wright JL, EllisWJ, 2006, Improved prostate cancer detection with anterior apical prostate biopsies. Urol Oncol 24:492–495 DjavanB ZAR, Remzi M, Ghawidel K, Basharkhah A, Schulman CC et al, 2000, Optimal predictors of prostate cancer in repeat prostate biopsy: a prospective study of 1051 men. J Urol 163: 1144–1148 Kim M, Choi SK, Park M, Shim M, Song C, Jeong IG, Hong JH, Kim CS, Ahn H, 2016, Characteristics of anterior located prostate cancer and usefulness of multiparametric magnetic resonance imaging for diagnosis. J Urol 196(2):367–373 Takashima R, Egawa S, Kuwao S, Baba S, 2002, Anterior distribution of Stage T1c nonpalpable tumors in radical prostatectomy specimens. Urology 59(5):692–697 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2357 EP 2361 DI 10.1007/s12253-020-00824-0 PG 5 ER PT J AU Guleria, P Kumar, S Malik, SP Jain, D AF Guleria, Prerna Kumar, Sunil Malik, Singh Prabhat Jain, Deepali TI PD-L1 Expression in Small Cell and Large Cell Neuroendocrine Carcinomas of Lung: an Immunohistochemical Study with Review of Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pulmonary high grade neuroendocrine tumors; PD-L1; SP263; Immunotherapy; Small cell lung carcinoma ID Pulmonary high grade neuroendocrine tumors; PD-L1; SP263; Immunotherapy; Small cell lung carcinoma AB High-grade neuroendocrine tumors (HGNET) have distinctive tumor biology/behaviour. Newer modalities of treatment (immunotherapy) for them have been included in recent NCCN guidelines. Detection of programmed death receptor-ligand 1 (PD-L1) expression by immunohistochemistry have made easy identification of patients eligible for immunotherapy. We aimed to ascertain expression of PD-L1 on small cell and large cell neuroendocrine carcinomas of lung and review existing literature. Eighty-five cases of HGNET lung (primary/metastatic), were retrieved and reviewed. Immunostaining for PD-L1 using clone SP263 was done. Any amount/intensity of membranous staining of > = 1% tumor cells was cut-off for positivity. Previously published studies using Google and/Pubmed search engines were reviewed. Of 85 cases, 70 were small-cell lung cancer (SCLC), 11 large-cell neuroendocrine carcinoma (LCNEC) and 4 combined SCLC. Median age was 46.5 years with male preponderance. No PD-L1 expression was seen in 91.6% cases. The 7 positive cases were 4 LCNEC, 2 SCLC and 1 combined SCLC. The percentage positivity varied from 1–100%; lower percentage positivity was seen in SCLC. PD-L1 expression on immune cells was seen in 31.3%cases. Sixteen studies evaluating 1992 NETwere found; E1L3N PD-L1 clone was commonly used clone. PDL1 positivity was associated with better prognosis in most studies. There are only a few studies available in literature related to PDL1 expression in high grade neuroendocrine carcinomas of lung. In general, PD-L1 positivity is highly variable and seen in lower percentage of these tumors. With the recent approval of immunotherapy, biomarkers other than PD-L1 should also be investigated in these tumors. C1 [Guleria, Prerna] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. [Kumar, Sunil] All India Institute of Medical Sciences, Dr BRA Institute-Rotary Cancer Hospital, Department of Surgical Oncology, 110029 New Delhi, India. [Malik, Singh Prabhat] All India Institute of Medical Sciences, Dr BRA Institute-Rotary Cancer Hospital, Department of Medical Oncology, 110029 New Delhi, India. [Jain, Deepali] All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. RP Jain, D (reprint author), All India Institute of Medical Sciences, Department of Pathology, 110029 New Delhi, India. EM deepalijain76@gmail.com CR Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JH, Beasley MB, Chirieac LR, Dacic S, Duhig E, Flieder DB, Geisinger K The 2015 World health organization classification of lung tumors: impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol 2015:1243–1260 Horn L, Reck M, Spigel DR. The future of immunotherapy in the treatment of small cell lung cancer. Oncologist 2016:910–921 National Comprehensive Cancer Network. Small cell Lung cancer, Version 1.2019, https://www.nccn.org/professionals/physician_ gls/pdf/sclc.pdf., Accessed 22 Apr 2019) Tang J, Yu JX, Hubbard-Lucey VM, Neftelinov ST, Hodge JP, Lin Y. Trial watch: the clinical trial landscape for PD1/PDL1 immune checkpoint inhibitors. Nat Rev Drug Discov 2018:854–855 Ventana Medical Systems, 2016, VENTANA PD-L1, SP263, Assay staining of non-small cell lung cancer. [interpretation manual] 1:1–42 Inamura K. Update on immunohistochemistry for the diagnosis of lung cancer. Cancers, Basel, 2018:1–15 Tsao MS, Kerr KM, Dacic S, Yatabe Y, Hirsch FR, 2017, IASLC atlas of PD-L1 immunohistochemistry testing in lung cancer. Editorial Rx Press, North Fort Myers Schultheis AM, Scheel AH, Ozretic L, George J, Thomas RK, Hagemann T, Zander T, Wolf J, Buettner R. PD-L1 expression in small cell neuroendocrine carcinomas. Eur J Cancer 2015:421–426 George J, Fernandez-Cuesta L, Walter V, Hayes N. Comparative analysis of small cell lung cancer and other pulmonary neuroendocrine tumors. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16–20; New Orleans, LA. Philadelphia, PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 122 Takada K, Toyokawa G, Okamoto T, Akamine T, Takamori S, Katsura M, Fujishita T, Shoji F, Oda Y, Maehara Y. An immunohistochemical analysis of PD-L1 protein expression in surgically resected small cell lung cancer using different antibodies and criteria. Anticancer Res 2016:3409–3412 Inamura K, Yokouchi Y, KobayashiM, Ninomiya H, Sakakibara R, Nishio M, Okumura S, Ishikawa Y. Relationship of tumor PD-L1, CD274, expression with lower mortality in lung high-grade neuroendocrine tumor. Cancer Med 2017:2347–2356 Tsuruoka K, Horinouchi H, Goto Y, Kanda S, Fujiwara Y, Nokihara H, Yamamoto N, Asakura K, Nakagawa K, Sakurai H, Watanabe SI. PD-L1 expression in neuroendocrine tumors of the lung. Lung Cancer 2017:115–120 Ohtaki Y, Kaira K, Atsumi J, Nagashima T, Kawashima O, Ibe T, Kamiyoshihara M, Onozato R, Fujita A, Yazawa T, Sugano M. Prognostic significance of PD-L1 expression and tumor infiltrating lymphocytes in large cell neuroendocrine carcinoma of lung. Am J Transl Res 2018:3243–3253 George J, Saito M, Tsuta K, Iwakawa R, Shiraishi K, Scheel AH, Uchida S, Watanabe SI, Nishikawa R, Noguchi M, Peifer M. Genomic amplification of CD274, PD-L1, in small-cell lung cancer. Clin Cancer Res 2017:1220–1226 Yasuda Y, Ozasa H, Kim YH. PD-L1 Expression in Small Cell Lung Cancer. J Thorac Oncol 2018:e40–e41 Kasajima A, Ishikawa Y, Iwata A, Steiger K, Oka N, Ishida H, Sakurada A, Suzuki H, Kameya T, Konukiewitz B, Kloppel G. Inflammation and PD-L1 expression in pulmonary neuroendocrine tumors. Endocr Relat Cancer 2018:339–350 Yoshimura A, Yamada T, Miyagawa-Hayashino A, Sonobe Y, Imabayashi T, Yamada T, Okada S, Shimamoto T, Chihara Y, Iwasaku M, Kaneko Y. Comparing three different anti-PD-L1 antibodies for immunohistochemical evaluation of small cell lung cancer. Lung Cancer 2019:108–112 Yu H, Batenchuk C, Badzio A, Boyle TA, Czapiewski P, Chan DC, Lu X, Gao D, Ellison K, Kowalewski AA, Rivard CJ. PD-L1 Expression by two complementary diagnostic assays and mRNA in situ hybridization in small cell lung cancer. J Thorac Oncol 2017:110–120 Fan Y,Ma K,Wang C, Ning J, Hu Y, Dong D, Dong X, Geng Q, Li E, Wu Y. Prognostic value of PD-L1 and PD-1 expression in pulmonary neuroendocrine tumors. Onco Targets Ther 2016:6075– 6082 Kim HS, Lee JH, Nam SJ, Ock CY, Moon JW, Yoo CW, Lee GK, Han JY. Association of PD-L1 Expression with tumor-infiltrating immune cells and mutation burden in high-grade neuroendocrine carcinoma of the lung. J Thorac Oncol 2018:636–648 Eichhorn F, Harms A,Warth A, Muley T,Winter H, Eichhorn ME. PD-L1 expression in large cell neuroendocrine carcinoma of the lung. Lung Cancer 2018:76–82 Wang H, Li Z, Dong B, SunW, Yang X, Liu R, Zhou L, Huang X, Jia L, Lin D. Prognostic significance of PD-L1 expression and CD8 + T cell infiltration in pulmonary neuroendocrine tumors. Diagn Pathol 2018:30 Ishii H, Azuma K, Kawahara A, Yamada K, Imamura Y, Tokito T, Kinoshita T, Kage M, Hoshino T. Significance of programmed cell death-ligand 1 expression and its association with survival in patients with small cell lung cancer. J Thorac Oncol 2015:426–430 Miao L, LuY,Xu Y, Zhang G, Huang Z, Gong L, FanY. PD-L1 and c-MET expression and survival in patients with small cell lung cancer. Oncotarget 2017:53978–53988 Hendifar AE, Marchevsky AM, Tuli R. Neuroendocrine tumors of the lung: current challenges and advances in the diagnosis and management of well-differentiated disease. J Thorac Oncol 2017: 425–436 Menon S, Shin S, Dy G. Advances in cancer immunotherapy in solid tumors. Cancers, Basel, 2016:1–21 WangM, Zhao J, Zhang L,Wei F, Lian Y,Wu Y, Gong Z, Zhang S, Zhou J, Cao K, Li X. Role of tumor microenvironment in tumorigenesis. J Cancer 2017:761–773 Teixido C, Vilarino N, Reyes R, Reguart N. PD-L1 expression testing in non-small cell lung cancer. Ther Adv Med Oncol 2018: 175883591876349 Derks JL, Leblay N, Lantuejoul S, Dingemans AMC, Speel EJM, Fernandez-Cuesta L. New insights into the molecular characteristics of pulmonary carcinoids and large cell neuroendocrine carcinomas, and the impact on their clinical management. J Thorac Oncol 2018:752–766 Maddison P, Newsom-Davis J,Mills KR, Souhami RL. Favourable prognosis in Lambert-Eaton myasthenic syndrome and small-cell lung carcinoma. Lancet 1999:117–118 Wang W, Hodkinson P, McLaren F, Mackean MJ, Williams L, Howie SE, Wallace WA, Sethi T. Histologic assessment of tumorassociated CD45 + cell numbers is an independent predictor of prognosis in small cell lung cancer. Chest 2013:146–151 Scorer P, Scott M, Lawson N, Ratcliffe MJ, Barker C, Rebelatto MC,Walker J. Consistency of tumor and immune cell programmed cell death ligand-1 expression within and between tumor blocks using the VENTANA SP263 assay. Diagn Pathol 2018:47 Durvalumab, Imfinzi). https://www.fda.gov/drugs/resourcesinformation- approved-drugs/durvalumab-imfinzi. Accessed 04 Feb 2020 Williams GH, NicholsonAG, Snead DR, Thunnissen E, Lantuejoul S, Cane P, Kerr KM, Loddo M, Scott ML, Scorer PW, Barker C. Inter-observer Reliability of Programmed Cell Death Ligand-1 Scoring Using the VENTANA PD-L1, SP263, Assay in Non- Small Cell Lung Cancer. J Thorac Oncol 2020:550–555 Zhao X, Kallakury B, Chahine JJ, Hartmann D, Zhang Y, Chen Y, Zhang H, Zhang B, Wang C, Giaccone G. Surgical resection of SCLC: prognostic factors and the tumor microenvironment. J Thorac Oncol 2019:914–923 Ye Y, Zhou L, Xie X, Jiang G, Xie H, Zheng S. Interaction of B7- H1 on intrahepatic cholangiocarcinoma cells with PD-1 on tumorinfiltrating Tcells as amechanismof immune evasion. J Surg Oncol 2009:500–504 Antonia SJ, Lopez-Martin JA, Bendell J, Ott PA, TaylorM, Eder JP, Jager D, PietanzaMC, Le DT, de Braud F,MorseMA, Ascierto PA, Horn L, Amin A, Pillai RN, Evans J, Chau I, Bono P, Atmaca A, Sharma P, Harbison CT, Lin CS, Christensen O, Calvo E. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer, CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol 2016:883–895 da Silva A, Bowden M, Zhang S, Masugi Y, Thorner AR, Herbert ZT, ZhouCW, Brais L, Chan JA, Hodi FS, Rodig S, Ogino S,Kulke MH. Characterization of the neuroendocrine tumor immune microenvironment. Pancreas 2018:1123–1129 Schats KA, Van Vre EA, Boeckx C, De Bie M, Schrijvers DM, Neyns B, De Meester I, Kockx MM. Optimal evaluation of programmed death ligand-1 on tumor cells versus immune cells requires different detection methods. Arch Pathol Lab Med 2018: 982–991 Ott PA, Elez E, Hiret S, Kim DW, Morosky A, Saraf S, Piperdi B, Mehnert JM. Pembrolizumab in patients with extensive-stage small-cell lung cancer: results from the phase Ib KEYNOTE-028 study. J Clin Oncol 2017:3823–3829 Pacheco J, Bunn PA. Advancements in small-cell lung cancer: the changing landscape following IMpower-133. Clin Lung Cancer 2019:148–160 Yang S, Zhang Z, Wang Q. Emerging therapies for small cell lung cancer. J Hematol Oncol 2019:47 Verma V, Sharma G, Singh A. Immunotherapy in extensive small cell lung cancer. Exp Hematol Oncol 2019:5 Shah MH, GoldnerWS, Halfdanarson TR, Bergsland E, Berlin JD, Halperin D, Chan J, Kulke MH, Benson AB, Blaszkowsky LS, Eads J. NCCN guidelines insights: neuroendocrine and adrenal tumors, version 2.2018. J Natl Compr Canc Netw 2018:693–702 Pavan A, Attili I, Pasello G, Guarneri V, Conte PF, Bonanno L. Immunotherapy in small-cell lung cancer: from molecular promises to clinical challenges. J Immunother Cancer 2019:205 TsaoM, Kerr K,Yatabe Y, Hirsch FR. PD-L1 Immunohistochemistry comparability study in real-life, clinical samples: Results of Blueprint Phase 2 Project. J Thorac Oncol 2018:1302–1311 Hendry S, Byrne DJ, Wright GM, Young RJ, Sturrock S, Cooper WA, Fox SB. Comparison of four PD-L1 immunohistochemical assays in lung cancer. J Thorac Oncol 2018:367–376 AbsengerG, Terzic J, Bezan A. ASCO update: lung cancer. Memo - Mag Eur Med Oncol 2017:224–227 Diggs LP, Hsueh EC. Utility of PD-L1 immunohistochemistry assays for predicting PD-1/PD-L1 inhibitor response. Biomark Res 2017:12 Wang VE, Urisman A, Albacker L, Ali S, Miller V, Aggarwal R, Jablons D, 2017, Checkpoint inhibitor is active against large cell neuroendocrine carcinoma with high tumor mutation burden. J Immunother Cancer 5:75 Zimmerman S, Das A,Wang S, Julian R, Gandhi L, Wolf J. 2017– 2018 scientific advances in thoracic oncology: small cell lung cancer. J Thorac Oncol 2019:768–783 Li Q, Yuan D, Ma C, Liu Y,Ma L, Lv T, Song Y. A new hope: the immunotherapy in small cell lung cancer. Neoplasma 2016:342–350 Eerola AK, Soini Y, Paakko P, PerssonM, Bremnes RM, Busund L-T. A high number of tumor-infiltrating lymphocytes are associated with a small tumor size, low tumor stage, and a favorable prognosis in operated small cell lung carcinoma. Clin Cancer Res 2000:1875–1881 Vallonthaiel AG, Malik PS, Singh V, Kumar V, Kumar S, Sharma MC, Mathur S, Arava S, Guleria R, Jain D. Clinicopathologic correlation of programmed death ligand-1 expression in non-small cell lung carcinomas: a report from India. Ann Diagn Pathol 2017:56– 61 Guleria P, Husain N, Shukla S, Kumar S, Parshad R, Jain D. PD-L1 immuno-expression assay in thymomas: study of 84 cases and review of literature. Ann Diagn Pathol 2018:135–141 Wang C, Hahn E, Slodkowska E, Eskander A, Enepekides D, Higgins K, Vesprini D, Liu SK, Downes MR, Xu B. Reproducibility of PD-L1 immunohistochemistry interpretation across various types of genitourinary and head/neck carcinomas, antibody clones, and tissue types. Hum Pathol 2018:131–139 Li C, Huang C, Mok TS, ZhuangW, Xu H, Miao Q, Fan X, ZhuW, Huang Y, Lin X, Jiang K. Comparison of 22C3 PD-L1 expression between surgically resected specimens and paired tissue microarrays in non–small cell lung cancer. J Thorac Oncol 2017:1536– 1543 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2363 EP 2370 DI 10.1007/s12253-020-00832-0 PG 8 ER PT J AU Ye, L Ardakani, MN Thomas, C Spilsbury, K Leslie, C Amanuel, B Millward, M AF Ye, Linda Ardakani, Mesbah Nima Thomas, Carla Spilsbury, Katrina Leslie, Connull Amanuel, Benhur Millward, Michael TI Detection of Low-level EGFR c.2369 C > T (p.Thr790Met) Resistance Mutation in Pre-treatment Non-small Cell Lung Carcinomas Harboring Activating EGFR Mutations and Correlation with Clinical Outcomes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Activating EGFR mutation; T790M mutation; Digital PCR; Tyrosine kinase inhibitor; Mutation allele frequency ID Activating EGFR mutation; T790M mutation; Digital PCR; Tyrosine kinase inhibitor; Mutation allele frequency AB Increasing evidence points to the presence of low-level de novo T790Mmutations in patients with non-small cell lung carcinoma (NSCLC) harboring activating EGFR mutations. We utilized digital PCR (dPCR), a highly sensitive gene mutation detection method, to detect pre-treatment T790M mutations in NSCLC tumor samples and correlated the T790M status with clinical features and patient outcomes. DNA extracted from pre-treatment NSCLC tumor tissue with known activating EGFR mutations, diagnosed between October 2010 and May 2017 at PathWest laboratory, was used to perform targeted dPCR for quantitative detection of T790M mutations. T790M was detected in 42 of 109 pre-treatment samples (38.5%). Median variant allele frequency was 0.14%(range 0.02–28.5%). Overall response rate to first generation EGFR tyrosine kinase inhibitors (TKI) was 67% regardless of T790Mstatus. The median progression free survival was 10.7 (IQR 5.6–19.9) versus 6.7 (IQR 3.5–20.8) months in T790Mnegative and positive patients respectively. T790M positivity correlated with increased rate of early disease progression. It also correlated with increased mortality (HR 3.1 95%CI 1.2–8.1, p = 0.022) in patients who did not respond to TKI treatment. We detected a significant rate of low-level pre-treatment T790M mutations in NSCLC using highly sensitive dPCR. Low-level pre-treatment T790M did not impact treatment response rate or overall survival, but was associated with increased rate of early progression on TKI therapy. C1 [Ye, Linda] Sir Charles Gairdner Hospital, Department of Medical OncologyPerth, Western Australia, Australia. [Ardakani, Mesbah Nima] The University of Western Australia, School of Pathology and Laboratory MedicineCrawley, Western Australia, Australia. [Thomas, Carla] The University of Western Australia, School of Pathology and Laboratory MedicineCrawley, Western Australia, Australia. [Spilsbury, Katrina] The University of Notre Dame Australia, Institute for Health ResearchPerth, Western Australia, Australia. [Leslie, Connull] The University of Western Australia, School of Pathology and Laboratory MedicineCrawley, Western Australia, Australia. [Amanuel, Benhur] The University of Western Australia, School of Pathology and Laboratory MedicineCrawley, Western Australia, Australia. [Millward, Michael] Sir Charles Gairdner Hospital, Department of Medical OncologyPerth, Western Australia, Australia. RP Ardakani, MN (reprint author), The University of Western Australia, School of Pathology and Laboratory Medicine, Crawley, Australia. CR Kobayashi S, Boggon TJ, Dayaram T et al, 2005, EGFR mutation and resistance of non–small-cell lung cancer to gefitinib. N Engl J Med 352:786–792 Pao W, Miller VA, Politi KA et al, 2005, Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain, resistance to EGFR kinase inhibitors). PLoS Med 2:e73 Mitsudomi T, Yatabe Y, 2007, Mutations of the epidermal growth factor receptor gene and related genes as determinants of epidermal growth factor receptor tyrosine kinase inhibitors sensitivity in lung cancer, vol 98. Blackwell Publishing Asia, Melbourne, pp 1817– 1824 Sequist LV, Waltman BA, Dias-Santagata D et al, 2011)Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med 3:75ra26-75ra26 Cross DAE, Ashton SE, Ghiorghiu S et al, 2014, AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov 4:1046–1061 Mok TS, Wu Y-L, Ahn M-J et al, 2017, Osimertinib or platinum– pemetrexed in EGFR T790M–positive lung cancer. N Engl J Med 376:629–640 Ramalingam SS, Ohe JEG Y, Cho BC, Vansteenkiste J, Zhou C, Reungwetwattana T, Cheng Y, Chewaskulyong B, Shah R, Lee KH, Cheema P, Tiseo M, John T, Lin MC, Imamura F, Hodge R, Rukazenkov Y, Soria J, 2019, Planchard. Osimertinib vs comparator EGFR-TKI as first-line treatment for EGFRm advanced NSCLC, FLAURA): Final overall survival analysis. Ann Oncol 30:v851–v934 Inukai M, Toyooka S, Ito S et al, 2006, Presence of epidermal growth factor receptor gene T790M mutation as a minor clone in non-small cell lung cancer. Cancer Res 66:7854–7858 Su K-Y, Chen H-Y, Li K-C et al, 2012, Pretreatment epidermal growth factor receptor, EGFR, T790M mutation predicts shorter EGFR tyrosine kinase inhibitor response duration in patients with non-small-cell lung cancer. J Clin Oncol 30:433–440 Costa C, Molina MA, Drozdowskyj A et al, 2014, The impact of EGFR T790M mutations and BIM mRNA expression on outcome in patients with EGFR-mutant NSCLC treated with erlotinib or chemotherapy in the randomized phase III EURTAC trial. Clin Cancer Res 20:2001 Fujita Y, Suda K, Kimura H et al, 2012, Highly sensitive detection of EGFR T790M mutation using colony hybridization predicts favorable prognosis of patients with lung cancer harboring activating EGFR mutation. J Thorac Oncol 7:1640–1644 Iwama E, Takayama K, Harada T et al, 2015, Highly sensitive and quantitative evaluation of the EGFR T790M mutation by nanofluidic digital PCR. Oncotarget 6:20466–20473 Li H, Hu H, Wang R et al, 2014, Primary concomitant EGFR T790M mutation predicted worse prognosis in non-small cell lung cancer patients. OncoTargets Ther 7:513 Maheswaran S, Sequist LV, Nagrath S et al, 2008, Detection of mutations in EGFR in circulating lung-cancer cells. N Engl J Med 359:366–377 Rosell R, Molina M, Costa C et al, 2011, Pretreatment EGFR T790M mutation and BRCA1 mRNA expression in Erlotinibtreated advanced non-small-cell lung cancer patients with EGFR mutations. Clin Cancer Res 17:1160–1168 Watanabe M, Kawaguchi T, Isa S-I et al, 2015, Ultra-sensitive detection of the pretreatment EGFR T790M mutation in nonsmall cell lung cancer patients with an EGFR-activating mutation using droplet digital PCR. Clin Cancer Res 21:3552–3560 Yu HA, Arcila ME, Hellmann MD et al, 2014, Poor response to erlotinib in patients with tumors containing baseline EGFR T790M mutations found by routine clinical molecular testing. Ann Oncol 25:423–428 Zhang B, Xu C-W, Shao Y et al, 2015, Comparison of droplet digital PCR and conventional quantitative PCR for measuring EGFR gene mutation. Exp Ther Med 9:1383–1388 Armbruster DA, Pry T, 2008, Limit of blank, limit of detection and limit of quantitation. Clin Biochem Rev 29(Suppl 1):S49 Royston P, Parmar MKB, 2002, Flexible parametric proportionalhazards and proportional‐odds models for censored survival data, with application to prognostic modelling and estimation of treatment effects. Stat Med 21:2175–2197 Zhang B, Xu J, Zhang X et al, 2018, Coexistence of sensitive and resistant epidermal growth factor receptor, EGFR, mutations in pretreatment non-small cell lung cancer, NSCLC, patients: First or third generation tyrosine kinase inhibitors, TKIs)? Lung Cancer 117:27–31 Wu J-Y, Yu C-J, Chang Y-C et al, 2011, Effectiveness of tyrosine kinase inhibitors on “uncommon” epidermal growth factor receptor mutations of unknown clinical significance in non-small cell lung cancer. Clin Cancer Res 17:3812–3821 Oh JE, An CH, Yoo NJ et al, 2011, Detection of low-level EGFR T790M mutation in lung cancer tissues.(Report). APMIS 119:403 Mok TS, Wu Y-L, Thongprasert S et al, 2009, Gefitinib or carboplatin–paclitaxel in pulmonary adenocarcinoma. N Engl J Med 361:947–957 Lee Y, Lee GK, Lee YS et al, 2014, Clinical outcome according to the level of preexisting epidermal growth factor receptor T790M mutation in patients with lung cancer harboring sensitive epidermal growth factor receptor mutations. Cancer 120:2090–2098 Zhao J, Feng H-H, Zhao J-Y et al, 2016, A sensitive and practical method to detect the T790M mutation in the epidermal growth factor receptor. Oncol Lett 11:2573–2579 Tatematsu T, Okuda K, Suzuki A et al, 2017, The detectability of the pretreatment EGFR T790M mutations in lung adenocarcinoma using CAST-PCR and digital PCR. J Thorac Dis 9:2397–2403 Prediletto I, Vasile E, Bruno R et al., 2018, 1423PPre-treatment EGFR-T790M subclones in lung adenocarcinoma harboring activating mutation of EGFR: a positive prognostic factor for survival? Ann Oncol 29 Vallee A, Le Loupp A-G, Denis MG, 2014, Efficiency of the Therascreen[R] RGQ PCR kit for the detection of EGFR mutations in non-small cell lung carcinomas. Clin Chim Acta 429:8 Liang H, Pan Z, Wang W et al, 2018, The alteration of T790M between 19 del and L858R in NSCLC in the course of EGFR-TKIs therapy: a literature-based pooled analysis. J Thorac Dis 10:2311 Chen L-Y,Molina-Vila MA, Ruan S-Y et al, 2016, Coexistence of EGFR T790M mutation and common activating mutations in pretreatment non-small cell lung cancer: a systematic review andmetaanalysis. Lung Cancer 94:46–53 Ma G, Zhang J, Yin L et al, 2017, The prognostic role of pretreatment epidermal growth factor receptor T790M mutation in advanced non-small cell lung cancer patients treated with EGFR tyrosine kinase inhibitors. Oncotarget 8:50941–50948 Ding D, Yu Y, Li Z et al, 2014, The predictive role of pretreatment epidermal growth factor receptor T790M mutation on the progression-free survival of tyrosine-kinase inhibitor-treated nonsmall cell lung cancer patients: a meta-analysis. OncoTargets Ther 7:387–393 Liu Y, Sun L,Xiong Z-C et al, 2017)Meta-analysis of the impact of de novo and acquired EGFR T790M mutations on the prognosis of patients with non-small cell lung cancer receiving EGFR-TKIs. OncoTargets Ther 10:2267–2279 Vendrell J,Mazieres J, Senal R et al. Ultra-sensitive EGFR T790M detection as an independent prognostic marker for lung cancer patients harboring EGFR del19 mutations and treated with firstgeneration TKIs. Clin Cancer Res 2019:clincanres.2683.2018 Bell DW, Gore I, Okimoto RA et al, 2005, Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR. Nat Genet 37:1315–1316 Sequist LV, Martins RG, Spigel D et al, 2008, First-Line Gefitinib in Patients With Advanced Non–Small-Cell Lung Cancer Harboring Somatic EGFR Mutations. J Clin Oncol 26:2442–2449 Shih J-Y, Gow C-H, Yang P-C, 2005, EGFR Mutation Conferring Primary Resistance to Gefitinib in Non–Small-Cell Lung Cancer. N Engl J Med 353:207–208 Tokumo M, Toyooka S, Ichihara S et al, 2006, Double mutation and gene copy number of EGFR in gefitinib refractory non-smallcell lung cancer. Lung Cancer 53:117–121 Yang JCH, Sequist LV, Geater SL et al, 2015, Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: a combined post-hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol 16:830–838 Chmielecki J, Foo J, Oxnard GR et al, 2011, Optimization of dosing for EGFR-mutant non-small cell lung cancer with evolutionary cancer modeling. Sci Transl Med 3:90ra59 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2371 EP 2379 DI 10.1007/s12253-020-00833-z PG 9 ER PT J AU Gibo, T Yamada, Shi Kawamoto, M Uehara, T Kurita, H AF Gibo, Takahiko Yamada, Shin-ichi Kawamoto, Makiko Uehara, Takeshi Kurita, Hiroshi TI Immunohistochemical Investigation of Predictive Biomarkers for Mandibular Bone Invasion in Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Oral squamous cell carcinoma; Mandible; Bone invasion; ɑ-SMA; Predictive marker ID Oral squamous cell carcinoma; Mandible; Bone invasion; ɑ-SMA; Predictive marker AB The accurate preoperative determination of the extent of mandibular resection remains a challenge for the surgeons. The purpose of the present study was to immunohistochemically investigate predictive markers for histological bone invasion of oral squamous cell carcinoma (OSCC). The medical records of primary OSCC patients with mandibular bone contact in preoperative computed tomography scans between January 2003 and December 2017 were retrospectively reviewed and an immunohistochemical investigation was performed. Forty-five OSCC patients with mandibular bone contact radiographically were included in this study. Histopathologically, infiltrative bone invasion was observed in 19 patients (42.2%) and compressive bone invasion in 15 (33.3%). A correlation was noted between the histological pattern of bone invasion and mode of invasion (chi-squared test, p < 0.05). At the tumor surface, a correlation was observed between the expression of IL-6 and bone invasion (theWilcoxon test, p < 0.05), although the expressionwas soweak. At the bone contact area, the expression of both ɑ-SMAand OPG correlated with infiltrative bone invasion (ɑ-SMA; the Wilcoxon test, p < 0.05, OPG; p < 0.05). These results suggest that predictive markers for aggressive (infiltrative) bone invasion in OSCC patients with a higher mode of invasion are the expression of ɑ-SMA and OPG. C1 [Gibo, Takahiko] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. [Yamada, Shin-ichi] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. [Kawamoto, Makiko] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. [Uehara, Takeshi] Shinshu University School of Medicine, Department of Laboratory Medicine, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. [Kurita, Hiroshi] Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 3-1-1, Asahi, 390-8621 Matsumoto, Japan. RP Yamada, Shi (reprint author), Shinshu University, School of Medicine, Department of Dentistry and Oral Surgery, 390-8621 Matsumoto, Japan. EM yshinshin@shinshu-u.ac.jp CR Johnson NW, Jayasekara P, Amarasinghe AA, 2011, Squamous cell carcinoma and precursor lesions of the oral cavity: epidemiology and aetiology. Periodontol 57:19–37 Yamada SI, Kurita H, Tomioka T, Ohta R, Yoshimura N, Nishimaki F, Koyama Y, Kondo E, Kamata T, 2017, Healthy life expectancy of oral squamous cell carcinoma patients aged 75years and older. Oral Oncol 64:22–26 Brown JS, Lowe D, Kalavrezos N, D'Souza J, Magennis P, Woolgar J, 2002, Patterns of invasion and routes of tumor entry into the mandible by oral squamous cell carcinoma. Head Neck 24: 370–383 Chen YL, Kuo SW, Fang KH, Hao SP, 2011, Prognostic impact of marginal mandibulectomy in the presence of superficial bone invasion and the nononcologic outcome. Head Neck 33:708–713 Vaassen LAA, Speel EM, Kessler PAWH(2017, Bone invasion by oral squamous cell carcinoma: molecular alterations leading to osteoclastogenesis – a review of literature. J Craniomaxillofac Surg 45:1464–1471 Du W, Fang Q, Wu Y, Wu J, Zhang X, 2019, Oncologic outcome of marginal mandibulectomy in squamous cell carcinoma of the lower gingiva. BMC Cancer 19:775. , DOI 10.1186/ s12885-019-5999-0 Uribe S, Rojas LA, Rosas CF, 2013, Accuracy of imagingmethods for detection of bone tissue invasion in patients with oral squamous cell carcinoma. Dentomaxillofac Radiol 42:20120346 Totsuka Y, Usui Y, Tei K, Fukuda H, Shindo M, Lizuka T, Amemiya A, 1991, Mandibular involvement by squamous cell carcinoma of the lower alveolus: analysis and comparative study of histologic and radiologic features. Head Neck 13:40–50 De Wever O, Demetter P, Mareel M, Bracke M, 2008, Stromal myofibroblasts are drivers of invasive cancer growth. Int J Cancer 123:2229–2238 Michalek J, Brychtova S, Pink R, Dvorak Z, 2019, Prognostic and predictive markers for perineural and bone invasion of oral squamous cell carcinoma. Biomed PapMed Fac Univ Palacky Olomouc Czech Repub 163:302–308. , DOI 10.5507/bp.2019.032 Sobin LH, Gospodarowicz MK, Wittekind C, eds,, 2009, TNM classification of malignant tumours, 7th edn. Wiley-Blackwell, Chichester Yamamoto E, Miyakawa A, Kohama G, 1984, Mode of invasion and lymph node metastasis in squamous cell carcinoma of the oral cavity. Head Neck Surg 6:938–947 Hirsch FR, Varella-Garcia M, Bunn PA Jr, Di Maria MV, Veve R, Bremmes RM, Baron AE, Zeng C, FranklinWA, 2003, Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol 21:3798–37807 Nomura T, Shibahara T, Cui NH, Noma H, 2005, Patterns of mandibular invasion by gingival squamous cell carcinoma. J Oral Maxillofac Surg 63:1489–1493 Elmusrati AA, Pilborough AE, Khurram SA, Lambert DW, 2017, Cancer-associated fibroblasts promote bone invasion in oral squamous cell carcinoma. Br J Cancer 117:867–875 Ebrahimi A, Murali R, Gao K, Elliott MS, Clark JR, 2011, The prognostic and staging implications of bone invasion in oral squamous cell carcinoma. Cancer 117:4460–4467 Fried D, Mullins B, Weissler M, Shores C, Zanation A, Hackman T, ShockleyW, Hayes N, Chera BS, 2014, Prognostic significance of bone invasion for oral cavity squamous cell carcinoma considered T1/T2 by American joint committee on cancer size criteria. Head Neck 36:776–781 Radisky DC, Kenny PA, Bissell MJ, 2007, Fibrosis and cancer: do myofibroblasts come also from epithelial cells via EMT? J Cell Biochem 101:830–839 Saadi A, Shannon NB, Lao-Sirieix P, O'Donovan M, Walker E, Clemons NJ, Hardwick JS, Zhang C, Das M, Save V, Novelli M, Balkwill F, Fitzgerald RC, 2010, Stromal genes discriminate preinvasive from invasive disease, predict outcome, and highlight inflammatory pathways in digestive cancers. Proc Natl Acad Sci U S A 107:2177–2182 VeredM, Dayan D, Yahalom R, Dobriyan A, Barshack I, Bello IO, Kantola S, Salo T, 2010, Cancer-associated fibroblasts and epithelial-mesenchymal transition in metastatic oral tongue squamous cell carcinoma. Int J Cancer 127:1356–1362 Marsh D, Suchak K, Moutasim KA, Vallath S, Hopper C, Jerjes W, Upile T, Kalavrezos N, Violette SM, Weinreb PH, Chester KA, Chana JS, Marshall JF, Hart IR, Hackshaw AK, Piper K, Thomas GJ, 2011, Stromal features are predictive of disease mortality in oral cancer patients. J Pathol 223:470–481 Ishikuro M, Sakamoto K, Kayamori K, Akashi T, Kanda H, Izumo T, Yamaguchi A, 2008, Significance of the fibrous stroma in bone invasion by human gingival squamous cell carcinomas. Bone 43: 621–627 Quan J, Du Q, Hou Y, Wang Z, Zhang J, 2017, Utilization of Ecadherin by monocytes from tumour cells plays key roles in the progression of bone invasion by oral squamous cell carcinoma. Oncol Rep 38:850–858. , DOI 10.3892/or.2017.5749 Tada T, Jimi E, Okamoto M, Ozeki S, Okabe K, 2005, Oral squamous cell carcinoma cells induce osteoclast differentiation by suppression of osteoprotegerin expression in osteoblasts. Int J Cancer 116:253–262 Russmueller G, Moser D, Wurger T, Wrba F, Christopoulos P, Kostakis G, Seemann R, Stadler V, Wimmer G, Kornek G, Psyrri A, FilipitsM, Perisanidis C, 2015, Upregulation of osteoprotegerin expression correlates with bone invasion and predicts poor clinical outcome in oral cancer. Oral Oncol 51:247–253 Quan J, Elhousiny M, Johnson NW, Gao J, 2013, Transforming growth factor-β1 treatment of oral cancer induces epithelialmesenchymal transition and promotes bone invasion via enhanced activity of osteoclasts. Clin Exp Metastasis 30:659–670 Ozaki-Honda Y, Seki S, Fujiwara M, Matsuura M, Fujita S, Ikeda H, Umeda M, Ayuse T, Ikeda T, 2017, Prognostic prediction of Oral squamous cell carcinoma by E-cadherin and N-cadherin expression in overall cells in tumor nests or tumor cells at the invasive front. Cancer Microenviron 10:87–94 Kessenbrock K, Plaks V, Werb Z, 2010, Matrix metalloproteinases: regulators of the tumor microenvironment. Cell 141:52–67 Qiao B, Johnson NW, Gao J, 2010, Epithelial-mesenchymal transition in oral squamous cell carcinoma triggered by transforming growth factor-beta1 is snail family-dependent and correlates with matrix metalloproteinase-2 and -9 expressions. Int J Oncol 37:663– 668 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2381 EP 2389 DI 10.1007/s12253-020-00826-y PG 9 ER PT J AU Pal, D Szilagyi, B Berczeli, M Szalay, ICs Sardy, B Olah, Z Szekely, T Racz, G Banga, P Czinege, Zs Sotonyi, P AF Pal, Daniel Szilagyi, Brigitta Berczeli, Marton Szalay, Imre Csaba Sardy, Balazs Olah, Zoltan Szekely, Tamas Racz, Gergely Banga, Peter Czinege, Zsofia Sotonyi, Peter TI Ruptured Aortic Aneurysm and Dissection Related Death: an Autopsy Database Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Acute aortic syndrome; Aortic dissection; Aortic aneurysm; Autopsy; Rupture; Bl ID Acute aortic syndrome; Aortic dissection; Aortic aneurysm; Autopsy; Rupture; Bl AB Acute aortic catastrophes (AAC), mainly ruptured aneurysms and dissections, lead all other vascular conditions in morbidity and mortality, even if intervention occurs. The aim of our study was to give a descriptive overview of the demographic and pathological characteristics of AAC. Between 1994 and 2013, 80,469 autopsies were performed at Semmelweis University hospitals in Budapest. After collecting the autopsy reports we were able to create the AAC database upon which we conducted our analysis. We found 567 cases of AAC. The cause of death in 120 of them was classified as a non-ruptured aorta with malperfusion or distal embolization. Of the remaining 447 cases, in 305 the cause of death was a ruptured aortic aneurysm (rAA), and in 142 it was a ruptured aortic dissection (rAD). The distribution of rAA cases was 34.4% thoracal, 4.3% thoracoabdominal, and 61.3% abdominal. We found female dominance where the rAA was thoracal. In rAD cases, 84% were Stanford A and 16% Stanford B type. In both groups we found different pathological distributions. In the prehospital group, the number of thoracal ruptures was considerable. 88% of the patients with Stanford A dissection died in the prehospital or perioperative period. The most progressive AACs were ruptures of intrapericardial aneurysms and Stanford A dissections., however survival rate can be elevated by using rapid imaging examination and immediate surgical intervention. We want to highlight that our study contains such gender differences, which are worth to be taken into consideration. C1 [Pal, Daniel] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor u. 68, 1122 Budapest, Hungary. [Szilagyi, Brigitta] Budapest University of Technology and Economics, Institute of Mathematics, Department of GeometryBudapest, Hungary. [Berczeli, Marton] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor u. 68, 1122 Budapest, Hungary. [Szalay, Imre Csaba] Semmelweis University, Department of Cardiovascular SurgeryBudapest, Hungary. [Sardy, Balazs] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor u. 68, 1122 Budapest, Hungary. [Olah, Zoltan] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor u. 68, 1122 Budapest, Hungary. [Szekely, Tamas] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Racz, Gergely] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Banga, Peter] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor u. 68, 1122 Budapest, Hungary. [Czinege, Zsofia] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor u. 68, 1122 Budapest, Hungary. [Sotonyi, Peter] Semmelweis University, Department of Cardiovascular Surgery, Varosmajor u. 68, 1122 Budapest, Hungary. RP Sotonyi, P (reprint author), Semmelweis University, Department of Cardiovascular Surgery, 1122 Budapest, Hungary. EM sotonyi@hotmail.com CR Deaths in reference year, in Hungary, 1995–2015,, 2016, Hungarian Central Statistical Office Clouse WD et al, 1998, Improved prognosis of thoracic aortic aneurysms: a population-based study. JAMA 280(22):1926–1929 Upchurch GRJ, 2014, Thoracic and Thoracoabdominal Aortic Aneurysms: Evaluation and Decision Making. Rutherford’s Vascular Surgery, Edition E, Cronenwett JL, Johnston KW, eds), Elsevier Saunders, Philadelphia, pp 2084–2101 Coady MA et al, 1997, What is the appropriate size criterion for resection of thoracic aortic aneurysms? J Thorac Cardiovasc Surg 113(3):476–491; discussion 489–491 Davies RR et al, 2006, Novel measurement of relative aortic size predicts rupture of thoracic aortic aneurysms. Ann Thorac Surg 81(1):169–177 Kuivaniemi H, Elmore JR, 2012, Opportunities in abdominal aortic aneurysm research: epidemiology, genetics, and pathophysiology. Ann Vasc Surg 26(6):862–870 Holt PJE, Thompson MM, 2014, Abdominal aortic aneurysms: evaluation and decision making. Rutherford’s Vascular Surgery, Edition E, Cronenwett JL, Johnston KW, eds), Elsevier Saunders, Philadelphia, pp 1999–2023 Wilmink AB, Quick CR, 1998, Epidemiology and potential for prevention of abdominal aortic aneurysm. Br J Surg 85(2):155–162 Robertson L, Nandhra S, 2017, Laparoscopic surgery for elective abdominal aortic aneurysm repair. Cochrane Database Syst Rev 5: CD012302 Choksy SA, Wilmink AB, Quick CR, 1999, Ruptured abdominal aortic aneurysm in the Huntingdon district: a 10-year experience. Ann R Coll Surg Engl 81(1):27–31 Schlosser FJ et al, 2010, Mortality after elective abdominal aortic aneurysm repair. Ann Surg 251(1):158–164 Conrad MF, Cambria RP, 2014, Aortic dissection. Rutherford’s Vascular Surgery, Edition E, Cronenwett JL, Johnston KW, eds), Elsevier Saunders, Philadelphia, pp 2169–2188 Meszaros I et al, 2000, Epidemiology and clinicopathology of aortic dissection. Chest 117(5):1271–1278 Hagan PG et al, 2000, The International Registry of Acute Aortic Dissection, IRAD): new insights into an old disease. JAMA283(7): 897–903 Januzzi JL et al, 2004, Characterizing the young patient with aortic dissection: results from the International Registry of Aortic Dissection, IRAD). J Am Coll Cardiol 43(4):665–669 Mehta RH et al, 2002, Predicting death in patients with acute type a aortic dissection. Circulation 105(2):200–206 Cambria RP et al, 1988, Vascular complications associated with spontaneous aortic dissection. J Vasc Surg 7(2):199–209 Khan IA, Nair CK, 2002, Clinical, diagnostic, and management perspectives of aortic dissection. Chest 122(1):311–328 Distribution of Hungary’s population, 2nd November 2017, Hungarian Central Statistical Office Brindley P, Stembridge VA, 1956, Aneurysms of the aorta; a clinicopathologic study of 369 necropsy cases. Am J Pathol 32(1):67– 82 Olsson C et al, 2002, Thoracic aortic aneurysm and dissection: increasing prevalence and improved outcomes reported in a nationwide population-based study of more than 14,000 cases from 1987 to 2002. Circulation 114(24):2611–2618 Global regional, 2016, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990– 2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet 388(10053):1545–1602 Lauterbach SR et al, 2001, Contemporary management of aortic branch compromise resulting from acute aortic dissection. J Vasc Surg 33(6):1185–1192 Grimm JC et al, 2016, Differential outcomes of type A dissection with malperfusion according to affected organ system. Ann Cardiothorac Surg 5(3):202–208 KoppMS, Rethelyi J, 2004)Where psychology meets physiology: chronic stress and premature mortality - the Central-Eastern European health paradox. Brain Res Bull 62(5):351–367 Young R, Ostertag H, 1987, Incidence, etiology and risk of rupture of aortic aneurysm. An autopsy study. Dtsch Med Wochenschr 112(33):1253–1256 Hoornweg LL et al, 2008, Meta analysis on mortality of ruptured abdominal aortic aneurysms. Eur J Vasc Endovasc Surg 35(5):558– 570 Makrygiannis G et al, 2014, Sex differences in abdominal aortic aneurysm: the role of sex hormones. Ann Vasc Surg 28(8):1946– 1958 Villard C et al, 2011, Reproductive history in women with abdominal aortic aneurysms. J Vasc Surg 54(2):341–345 Villard C et al, 2017, Differential expression of sex hormone receptors in abdominal aortic aneurysms. Maturitas 96:39–44 Reimerink JJ et al, 2013, Systematic review and meta-analysis of population-based mortality from ruptured abdominal aortic aneurysm. Br J Surg 100(11):1405–1413 Johansson G, Markstrom U, Swedenborg J, 1995, Ruptured thoracic aortic aneurysms: a study of incidence and mortality rates. J Vasc Surg 21(6):985–988 Melvinsdottir IH et al, 2016, The incidence and mortality of acute thoracic aortic dissection: results from a whole nation study. Eur J Cardiothorac Surg 50(6):1111–1117 Axelsson C et al, 2015, A description of the prehospital phase of aortic dissection in terms of early suspicion and treatment. Prehosp Disaster Med 30(2):155–162 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2391 EP 2399 DI 10.1007/s12253-020-00835-x PG 9 ER PT J AU Kontsek, E Pesti, A Bjornstedt, M Uveges, Szabo, E Garay, T Gordon, P Gergely, Sz Kiss, A AF Kontsek, Endre Pesti, Adrian Bjornstedt, Mikael Uveges, T. Szabo, Edina Garay, Tamas Gordon, Peter Gergely, Szilveszter Kiss, Andras TI Mid-Infrared Imaging Is Able to Characterize and Separate Cancer Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cancer; FT-IR; Fingerprint region; Transflectance ID Cancer; FT-IR; Fingerprint region; Transflectance AB Malignancies are still responsible for a large share of lethalities. Macroscopical evaluation of the surgical resection margins is uncertain. Big data based imaging approaches have emerged in the recent decade (mass spectrometry, two-photon microscopy, infrared and Raman spectroscopy). Indocianine green labelled MS is the most common approach, however, label free midinfrared imaging is more promising for future practical application. We aimed to identify and separate different transformed (A-375, HT-29) and non-transformed (CCD986SK) cell lines by a label-free infrared spectroscopy method. Our approach applied a novel set-up for label-free mid-infrared range classificationmethod. Transflection spectroscopywas used on aluminium coated glass slides. Both whole range spectra (4000–648 cm−1) and hypersensitive fingerprint regions (1800–648 cm−1) were tested on the imaged areas of cell lines fixed in ethanol. Non-cell spectra were possible to be excluded based on mean transmission values being above 90%. Feasibility of a mean transmission based spectra filtering method with principal component analysis and linear discriminant analysis was shown to separate cell lines representing different tissue types. Fingerprint region resulted the best separation of cell lines spectra with accuracy of 99.84% at 70–75 mean transmittance range. Our approach in vitro was able to separate unique cell lines representing different tissues of origin. Proper data handling and spectra processing are key steps to achieve the adaptation of this dye-free technique for intraoperative surgery. Further studies are urgently needed to test this novel, marker-free approach. C1 [Kontsek, Endre] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Pesti, Adrian] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Bjornstedt, Mikael] Karolinska University, Department of PathologyStockholm, Sweden. [Uveges, T.] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food ScienceBudapest, Hungary. [Szabo, Edina] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food ScienceBudapest, Hungary. [Garay, Tamas] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Gordon, Peter] Budapest University of Technology and Economics, Department of Electronics TechnologyBudapest, Hungary. [Gergely, Szilveszter] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food ScienceBudapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Kontsek, E (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. CR Dolgin E, 2018, Bringing down the cost of cancer treatment. Nature 555(7695):S26–S29. , DOI 10.1038/d41586-018- 483-3 2. Prades J, Remue E, van Hoof E, Borras JM, 2015, Is it worth reorganising cancer services on the basis of multidisciplinary teams, MDTs)? A systematic review of the objectives and organisation of MDTs and their impact on patient outcomes. Health Policy 119(4): 464–474. , DOI 10.1016/j.healthpol.2014.09.006 Orosco RK, Tapia VJ, Califano JA, Clary B, Cohen EEW, Kane C, Lippman SM,Messer K, Molinolo A,Murphy JD, Pang J, Sacco A, Tringale KR,Wallace A, Nguyen QT, 2018, Positive surgical margins in the 10 Most common solid cancers. Sci rep-Uk 8. Doi: ARTN 5686 10.1038/s41598–018–23403-5 Jones EA, Simon D, Karancsi T, Balog J, Pringle SD, Takats Z, 2019, Matrix assisted rapid evaporative ionization mass spectrometry. Anal Chem 91(15):9784–9791. , DOI 10.1021/acs. analchem.9b01441 Bunaciu AA, Hoang VD, Aboul-Enein HY, 2017, Vibrational micro-spectroscopy of human tissues analysis: review. Crit Rev Anal Chem 47(3):194–203. , DOI 10.1080/10408347. 2016.1253454 Haque A, Faizi MS, Rather JA, Khan MS, 2017, Next generation NIR fluorophores for tumor imaging and fluorescence-guided surgery: a review. Bioorg Med Chem 25(7):2017–2034. https://doi. org/10.1016/j.bmc.2017.02.061 Wolthuis R, Travo A, Nicolet C, Neuville A, GaubMP, Guenot D, Ly E,ManfaitM, Jeannesson P, Piott O, 2008, IR spectral imaging for histopathological characterization of Xenografted human colon carcinomas. Anal Chem 80(22):8461–8469. , DOI 10. 1021/ac801191x Silva JSF, Silva JYR, de Sa GF, Araujo SS, Filho MAG, Ronconi CM, Santos TC, Junior SA, 2018, Multifunctional system polyaniline-decorated ZIF-8 nanoparticles as a new chemo-Photothermal platform for cancer therapy. ACS Omega 3(9): 12147–12157. , DOI 10.1021/acsomega.8b01067 Oh Y, Quan YH, Choi Y, Kim CK, Kim H, Kim HK, Kim BM, 2013, Intraoperative combined color and fluorescent images-based sentinel node mapping in the porcine lung: comparison of indocyanine green with or without albumin premixing. J Thorac Cardiov Sur 146(6):1509–1515. , DOI 10.1016/j.jtcvs.2013.02.044 Ishizawa T, Fukushima N, Shibahara J,Masuda K, Tamura S, Aoki T, Hasegawa K, Beck Y, Fukayama M, Kokudo N, 2009, Realtime identification of liver cancers by using Indocyanine green fluorescent imaging. Cancer 115(11):2491–2504. , DOI 10.1002/cncr.24291 Balan V, Mihai CT, Cojocaru FD, Uritu CM, Dodi G, Botezat D, Gardikiotis I, 2019, Vibrational spectroscopy fingerprinting in medicine: from molecular to clinical practice. Materials 12, 18). Doi: ARTN 2884 10.3390/ma12182884. Bogomolov A, Belikova V, Zabarylo UJ, Bibikova O, Usenov I, Sakharova T, Krause H, Minet O, Feliksberger E, Artyushenko V, 2017, Synergy effect of combining fluorescence and mid infrared fiber spectroscopy for kidney tumor diagnostics. Sensors, Basel, 17(11). , DOI 10.3390/s17112548 Sojka L, Tang Z, Furniss D, Sakr H, Fang Y, Beres-Pawlik E, Benson TM, Seddon AB, Sujecki S, 2017, Mid-infrared emission in Tb3+−doped selenide glass fiber. J Opt Soc Am B 34(3):A70– A79. , DOI 10.1364/Josab.34.000a70 Nallala J, Lloyd GR, Hermes M, Shepherd N, Stone N, 2017, Enhanced spectral histology in the colon using high-magnification benchtop FTIR imaging. Vib Spectrosc 91:83–91. , DOI 10.1016/j.vibspec.2016.08.013 Joro R, Laaperi AL, Dastidar P, Soimakallio S, Kuukasjarvi T, Toivonen T, Saaristo R, Jarvenpaa R, 2008, Imaging of breast cancer with mid- and long-wave infrared camera. J Med Eng Technol 32(3):189–197. h t t p s : / / d o i . o r g / 1 0 . 1 0 8 0 / 03091900701234358 Paraskevaidi M, Morais CLM, Lima KMG, Ashton KM, Stringfellow HF, Martin-Hirsch PL, Martin FL, 2018, Potential of mid-infrared spectroscopy as a non-invasive diagnostic test in urine for endometrial or ovarian cancer. Analyst 143(13):3156– 3163. , DOI 10.1039/c8an00027a Zohdi V, Whelan DR,Wood BR, Pearson JT, Bambery KR, Black MJ, 2015, Importance of tissue preparation methods in FTIR micro-spectroscopical analysis of biological tissues: 'traps for new users'. PLoS One 10(2):e0116491. , DOI 10.1371/journal. pone.0116491 Sreedhar H, Varma VK, Nguyen PL, Davidson B, Akkina S, Guzman G, Setty S, Kajdacsy-Balla A, Walsh MJ, 2015, Highdefinition Fourier transform infrared, FT-IR, spectroscopic imaging of human tissue sections towards improving pathology. J Vis Exp 95:52332. , DOI 10.3791/52332 Gaydou V, Polette M, Gobinet C, Kileztky C, Angiboust JF, ManfaitM, Birernbaut P, PiotO(2016, Vibrational analysis of lung tumor cell lines: implementation of an invasiveness scale based on the cell infrared signatures. Anal Chem 88(17):8459–8467. https:// doi.org/10.1021/acs.analchem.6b00590 Grosserueschkamp F, Kallenbach-Thieltges A, Behrens T, Bruning T, Altmayer M, Stamatis G, Theegarten D, Gerwert K, 2015, Marker-free automated histopathological annotation of lung tumour subtypes by FTIR imaging. Analyst 140(7):2114–2120. https://doi. org/10.1039/c4an01978d Petersen D, Mavarani L, Niedieker D, Freier E, Tannapfel A, Kotting C, Gerwert K, El-Mashtoly SF, 2017, Virtual staining of colon cancer tissue by label-free Raman micro-spectroscopy. Analyst 142(8):1207–1215. , DOI 10.1039/c6an02072k Perez-Guaita D, Kuligowski J, Quintas G, Garrigues S, de la GuardiaM(2013, Atmospheric compensation in Fourier transform infrared, FT-IR, spectra of clinical samples. Appl Spectrosc 67(11): 1339–1342. , DOI 10.1366/13-07159 Hoult RA, 2003, Angle of rotation sensor having a rotating annular magnet and two ferritic stator halves United States Patent Hastie T, Tibshirani R, Friedman JH, 2009, The elements of statistical learning : data mining, inference, and prediction. Springer series in statistics,, 2nd edn. Springer, New York, NY Landis JR, Koch GG, 1977, The measurement of observer agreement for categorical data. Biometrics 33(1):159–174 Jansen-Winkeln B, Holfert N, Kohler H,Moulla Y, Takoh JP, Rabe SM, Mehdorn M, Barberio M, Chalopin C, Neumuth T, Gockel I, 2019, Determination of the transection margin during colorectal resection with hyperspectral imaging, HSI). Int J Color Dis 34(4): 731–739. , DOI 10.1007/s00384-019-03250-0 ChanKLA, Kazarian SG(2016)Attenuated total reflection Fouriertransform infrared, ATR-FTIR, imaging of tissues and live cells. Chem Soc Rev 45(7):1850–1864. , DOI 10.1039/ c5cs00515a Smialowski P, Frishman D, Kramer S, 2010, Pitfalls of supervised feature selection. Bioinformatics 26(3):440–443. , DOI 10. 1093/bioinformatics/btp621 Hendry S, Salgado R, Gevaert T, Russell PA, John T, Thapa B, Christie M, van de Vijver K, Estrada MV, Gonzalez-Ericsson PI, Sanders M, Solomon B, Solinas C, Van den Eynden G, Allory Y, Preusser M, Hainfellner J, Pruneri G, Vingiani A, Demaria S, Symmans F, Nuciforo P, Comerma L, Thompson EA, Lakhani S, Kim SR, Schnitt S, Colpaert C, Sotiriou C, Scherer SJ, Ignatiadis M, Badve S, Pierce RH, Viale G, Sirtaine N, Penault-Llorca F, Sugie T, Fineberg S, Paik S, Srinivasan A, Richardson A, Wang Y, Chmielik E, Brock J, Johnson DB, Balko J, Wienert S, Bossuyt V, Michiels S, Ternes N, Burchardi N, Luen SJ, Savas P,Klauschen F,Watson PH, Nelson BH, Criscitiello C, O'Toole S, Larsimont D, deWind R, Curigliano G, Andre F, Lacroix-Triki M, van de Vijver M, Rojo F, Floris G, Bedri S, Sparano J, Rimm D, Nielsen T, Kos Z, Hewitt S, Singh B, Farshid G, Loibl S, Allison KH, Tung N, Adams S, Willard-Gallo K, Horlings HM, Gandhi L, Moreira A, Hirsch F, Dieci MV, Urbanowicz M, Brcic I, Korski K, Gaire F, Koeppen H, Lo A, Giltnane J, Rebelatto MC, Steele KE, Zha J, Emancipator K, Juco JW, Denkert C, Reis-Filho J, Loi S, Fox SB, 2017, Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the international Immunooncology biomarkers working group: part 1: assessing the host immune response, TILs in invasive breast carcinoma and ductal carcinoma in situ, metastatic tumor deposits and areas for further research. Adv Anat Pathol 24(5):235–251. , DOI 10.1097/PAP.0000000000000162 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2401 EP 2407 DI 10.1007/s12253-020-00825-z PG 7 ER PT J AU Kotarac, N Dobrijevic, Z Matijasevic, S Savic-Pavicevic, D Brajuskovic, G AF Kotarac, Nevena Dobrijevic, Zorana Matijasevic, Suzana Savic-Pavicevic, Dusanka Brajuskovic, Goran TI Association of KLK3, VAMP8 and MDM4 Genetic Variants within microRNA Binding Sites with Prostate Cancer: Evidence from Serbian Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE miRSNPs; rs1058205; rs1010; rs4245739; Prostate cancer ID miRSNPs; rs1058205; rs1010; rs4245739; Prostate cancer AB A growing number of studies have suggested that genetic variants affecting the micro-RNA- binding mechanisms (miRSNPs) constitute a promising novel class of biomarkers for prostate cancer (PCa) biology. Among the most extensively studied miRSNPs in the context of cancer is the variation rs4245739 in the MDM4 gene, while a recent large-scale analysis revealed significant differences in genotype distributions between aggressive and non-aggressive disease for rs1058205 in KLK3 and rs1010 in VAMP8. In this study, we examined a total of 1083 subjects for these three variants using Taqman® SNP Genotyping Assays. Three hundred and fifty-five samples of peripheral blood were obtained from patients with PCa and 358 samples from patients with benign prostatic hyperplasia (BPH). The control group consisted of 370 healthy volunteers. Comparisons of genotype distributions among PCa and BPH patients, as well as between PCa patients and healthy controls, yielded no evidence of association between the analyzed genetic variants and the risk of developing PCa. However, all three tested genetic variants have shown the association with the parameters of PCa progression. For KLK3 variant rs1058205, minor allele C was found to associate with the lower serum PSA score in PCa patients (PSA > 20 ng/ml vs. PSA < 10 ng/ml comparison, Prec = 0.038; ORrec = 0.20, 95%CI 0.04–1.05). The obtained results point out the potential relevance of the tested genetic variants for the disease aggressiveness assessment. C1 [Kotarac, Nevena] University in Belgrade, Medical Faculty, Institute for Human GeneticsBelgrade, Serbia. [Dobrijevic, Zorana] University in Belgrade, Medical Faculty, Institute for Human GeneticsBelgrade, Serbia. [Matijasevic, Suzana] University in Belgrade, Medical Faculty, Institute for Human GeneticsBelgrade, Serbia. [Savic-Pavicevic, Dusanka] University in Belgrade, Medical Faculty, Institute for Human GeneticsBelgrade, Serbia. [Brajuskovic, Goran] University in Belgrade, Medical Faculty, Institute for Human GeneticsBelgrade, Serbia. RP Brajuskovic, G (reprint author), University in Belgrade, Medical Faculty, Institute for Human Genetics, Belgrade, Serbia. EM brajuskovic@bio.bg.ac.rs CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6):394–424. , DOI 10.3322/caac. 21492 Eeles R, Ni Raghallaigh H, 2018, Men with a susceptibility to prostate cancer and the role of genetic based screening. Translat Androl Urol 7(1):61–69. , DOI 10.21037/tau.2017.12.30 Schumacher FR, Al Olama AA, Berndt SI, Benlloch S, Ahmed M, Saunders EJ, Dadaev T, Leongamornlert D, Anokian E, Cieza- Borrella C, Goh C, Brook MN, Sheng X, Fachal L, Dennis J, Tyrer J, Muir K, Lophatananon A, Stevens VL, Gapstur SM, Carter BD, Tangen CM, Goodman PJ, Thompson IM Jr, Batra J, Chambers S, Moya L, Clements J, Horvath L, TilleyW, Risbridger GP, Gronberg H, Aly M, Nordstrom T, Pharoah P, Pashayan N, Schleutker J, Tammela TLJ, Sipeky C, Auvinen A, Albanes D, Weinstein S, Wolk A, Hakansson N, West CML, Dunning AM, Burnet N, Mucci LA, Giovannucci E, Andriole GL, Cussenot O, Cancel-Tassin G, Koutros S, Beane Freeman LE, Sorensen KD, Orntoft TF, Borre M, Maehle L, Grindedal EM, Neal DE, Donovan JL, Hamdy FC, Martin RM, Travis RC, Key TJ, Hamilton RJ, Fleshner NE, Finelli A, Ingles SA, Stern MC, Rosenstein BS, Kerns SL, Ostrer H, Lu YJ, Zhang HW, Feng N, Mao X, Guo X, Wang G, Sun Z, Giles GG, SoutheyMC,MacInnis RJ, FitzGerald LM, Kibel AS, Drake BF, Vega A, Gomez- Caamano A, Szulkin R, Eklund M, Kogevinas M, Llorca J, Castano-Vinyals G, Penney KL, Stampfer M, Park JY, Sellers TA, Lin HY, Stanford JL, Cybulski C, Wokolorczyk D, Lubinski J, Ostrander EA, Geybels MS, Nordestgaard BG, Nielsen SF, Weischer M, Bisbjerg R, Roder MA, Iversen P, Brenner H, Cuk K, Holleczek B, Maier C, Luedeke M, Schnoeller T, Kim J, Logothetis CJ, John EM, Teixeira MR, Paulo P, Cardoso M, Neuhausen SL, Steele L, Ding YC, De Ruyck K, De Meerleer G, Ost P, Razack A, Lim J, Teo SH, Lin DW, Newcomb LF, Lessel D, Gamulin M, Kulis T, Kaneva R, Usmani N, Singhal S, Slavov C, Mitev V, Parliament M, Claessens F, Joniau S, Van den Broeck T, Larkin S, Townsend PA, Aukim-Hastie C, Gago-Dominguez M, Castelao JE, Martinez ME, Roobol MJ, Jenster G, van Schaik RHN, Menegaux F, Truong T, Koudou YA, Xu J, Khaw KT, Cannon-Albright L, Pandha H, Michael A, Thibodeau SN, McDonnell SK, Schaid DJ, Lindstrom S, Turman C, Ma J, Hunter DJ, Riboli E, Siddiq A, Canzian F, Kolonel LN, Le Marchand L, Hoover RN, Machiela MJ, Cui Z, Kraft P, Amos CI, Conti DV, Easton DF, Wiklund F, Chanock SJ, Henderson BE, Kote-Jarai Z, Haiman CA, Eeles RA, Profile S, Australian Prostate Cancer B, Study I, Canary PI, Breast, Prostate Cancer Cohort C, Consortium P, Cancer of the Prostate in S, Prostate Cancer Genome-wide Association Study of Uncommon Susceptibility L, Genetic A, Mechanisms in Oncology /Elucidating Loci Involved in Prostate Cancer Susceptibility C, 2018, Association analyses of more than 140,000 men identify 63 new prostate cancer susceptibility loci. Nat Genet 50(7):928– 936. , DOI 10.1038/s41588-018-0142-8 Buniello A, MacArthur JAL, Cerezo M, Harris LW, Hayhurst J, Malangone C, McMahon A, Morales J, Mountjoy E, Sollis E, Suveges D, Vrousgou O, Whetzel PL, Amode R, Guillen JA, Riat HS, Trevanion SJ, Hall P, Junkins H, Flicek P, Burdett T, Hindorff LA, Cunningham F, Parkinson H, 2019, The NHGRI-EBI GWAS catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res 47(D1): D1005–D1012. , DOI 10.1093/nar/gky1120 Wilk G, Braun R, 2018, regQTLs: single nucleotide polymorphisms that modulate microRNA regulation of gene expression in tumors. PLoS Genet 14(12):e1007837. , DOI 10.1371/ journal.pgen.1007837 Vishnoi A, Rani S, 2017, MiRNA biogenesis and regulation of diseases: an overview. Methods Mol Biol 1509:1–10. https://doi. org/10.1007/978-1-4939-6524-3_1 Salzman DW, Weidhaas JB, 2013, SNPing cancer in the bud: microRNA andmicroRNA-target site polymorphisms as diagnostic and prognostic biomarkers in cancer. Pharmacol Ther 137(1):55– 63. , DOI 10.1016/j.pharmthera.2012.08.016 CipolliniM, Landi S, Gemignani F, 2014, MicroRNA binding site polymorphisms as biomarkers in cancer management and research. Pharmacogen Personal Med 7:173–191. , DOI 10.2147/ PGPM.S61693 Yuan Y, Weidhaas JB, 2019, Functional microRNA binding site variants. Mol Oncol 13(1):4–8. , DOI 10.1002/1878-0261. 12421 Kotarac N, Dobrijevic Z, Matijasevic S, Savic-Pavicevic D, Brajuskovic G, 2019, Analysis of association of potentially functional genetic variants within genes encoding miR-34b/c, miR-378 and miR-143/145 with prostate cancer in Serbian population. EXCLI J 18:515–529. , DOI 10.17179/excli2019-1257 Nikolic Z, Savic Pavicevic D, Vucic N, Cidilko S, Filipovic N, Cerovic S, Vukotic V, Romac S, Brajuskovic G, 2015, Assessment of association between genetic variants in microRNA genes hsa-miR-499, hsa-miR-196a2 and hsa-miR-27a and prostate cancer risk in Serbian population. Exp Mol Pathol 99(1):145–150. , DOI 10.1016/j.yexmp.2015.06.009 Nikolic ZZ, Savic Pavicevic DL, Vukotic VD, Tomovic SM, Cerovic SJ, Filipovic N, Romac SP, Brajuskovic GN, 2014, Association between genetic variant in hsa-miR-146a gene and prostate cancer progression: evidence from Serbian population. Cancer Causes Cont: CCC 25(11):1571–1575. , DOI 10. 1007/s10552-014-0452-9 Hughes L, Ruth K, Rebbeck TR, Giri VN, 2013, Genetic variation in IL-16 miRNA target site and time to prostate cancer diagnosis in African-American men. Prostate Cancer Prostatic Dis 16(4):308– 314. , DOI 10.1038/pcan.2013.36 Liu J, Huang J, He Y, Liu J, Liao B, Liao G, 2014, Genetic variants in the integrin gene predicted microRNA-binding sites were associated with the risk of prostate cancer. Mol Carcinog 53(4):280– 285. , DOI 10.1002/mc.21973 Stegeman S, Amankwah E, Klein K, O'Mara TA, Kim D, Lin HY, Permuth-Wey J, Sellers TA, Srinivasan S, Eeles R, Easton D, Kote- Jarai Z,Amin Al Olama A, Benlloch S,Muir K, GilesGG,Wiklund F, Gronberg H, Haiman CA, Schleutker J, Nordestgaard BG, Travis RC, Neal D, Pharoah P, Khaw KT, Stanford JL, Blot WJ, Thibodeau S, Maier C, Kibel AS, Cybulski C, Cannon-Albright L, Brenner H, Kaneva R, Teixeira MR, Consortium P, Australian Prostate Cancer B, Spurdle AB, Clements JA, Park JY, Batra J, 2015, A large-scale analysis of genetic variants within putative miRNA binding sites in Prostate Cancer. Cancer Discov 5(4): 368–379. , DOI 10.1158/2159-8290.CD-14-1057 Stegeman S, Moya L, Selth LA, Spurdle AB, Clements JA, Batra J, 2015, A genetic variant of MDM4 influences regulation by multiplemicroRNAs in prostate cancer. Endocr Relat Cancer 22(2):265– 276. , DOI 10.1530/ERC-15-0013 Hong SK, 2014, Kallikreins as biomarkers for prostate cancer. Biomed Res Int 2014:526341–526310. , DOI 10.1155/ 2014/526341 Bensen JT, Xu Z, Smith GJ, Mohler JL, Fontham ET, Taylor JA, 2013, Genetic polymorphism and prostate cancer aggressiveness: a case-only study of 1,536 GWAS and candidate SNPs in African- Americans and European-Americans. Prostate 73(1):11–22. https:// doi.org/10.1002/pros.22532 Savblom C, Hallden C, Cronin AM, Sall T, Savage C, Vertosick EA, Klein RJ, Giwercman A, Lilja H, 2014, Genetic variation in KLK2 and KLK3 is associated with concentrations of hK2 and PSA in serum and seminal plasma in young men. Clin Chem 60(3):490–499. , DOI 10.1373/clinchem.2013.211219 Chen C, Xin Z, 2017, Single-nucleotide polymorphism rs1058205 of KLK3 is associated with the risk of prostate cancer: a casecontrol study of Han Chinese men in Northeast China. Medicine 96(10):e6280. , DOI 10.1097/MD.0000000000006280 Penney KL, Schumacher FR, Kraft P, Mucci LA, Sesso HD, Ma J, Niu Y, Cheong JK, Hunter DJ, Stampfer MJ, Hsu SI, 2011, Association of KLK3, PSA, genetic variants with prostate cancer risk and PSA levels. Carcinogenesis 32(6):853–859. , DOI 10.1093/carcin/bgr050 Wang CC, Shi H, Guo K, Ng CP, Li J, Gan BQ, Chien Liew H, Leinonen J, Rajaniemi H, Zhou ZH, Zeng Q, Hong W, 2007, VAMP8/endobrevin as a general vesicular SNARE for regulated exocytosis of the exocrine system. Mol Biol Cell 18(3):1056–1063. , DOI 10.1091/mbc.e06-10-0974 Zhu D, Zhang Y, Lam PP, Dolai S, Liu Y, Cai EP, Choi D, Schroer SA, Kang Y, Allister EM, Qin T, Wheeler MB, Wang CC, Hong WJ, Woo M, Gaisano HY, 2012, Dual role of VAMP8 in regulating insulin exocytosis and islet beta cell growth. Cell Metab 16(2):238–249. , DOI 10.1016/j.cmet.2012.07.001 Gansmo LB, Bjornslett M, Halle MK, Salvesen HB, Dorum A, Birkeland E, Hveem K, Romundstad P, Vatten L, Lonning PE, Knappskog S, 2016, The MDM4 SNP34091, rs4245739, Callele is associated with increased risk of ovarian-but not endometrial cancer. Tumour Biol 37(8):10697–10702. , DOI 10. 1007/s13277-016-4940-2 Gao F, Xiong X, Pan W, Yang X, Zhou C, Yuan Q, Zhou L, Yang M(2015, A regulatory MDM4 genetic variant locating in the binding sequence of multiple MicroRNAs contributes to susceptibility of small cell lung Cancer. PLoS One 10(8):e0135647. https://doi. org/10.1371/journal.pone.0135647 Garcia-Closas M, Couch FJ, Lindstrom S, Michailidou K, Schmidt MK, Brook MN, Orr N, Rhie SK, Riboli E, Feigelson HS, Le Marchand L, Buring JE, Eccles D, Miron P, Fasching PA, Brauch H, Chang-Claude J, Carpenter J, Godwin AK, Nevanlinna H, Giles GG, Cox A, Hopper JL, Bolla MK, Wang Q, Dennis J, Dicks E, Howat WJ, Schoof N, Bojesen SE, Lambrechts D, Broeks A, Andrulis IL, Guenel P, Burwinkel B, Sawyer EJ, Hollestelle A, Fletcher O, Winqvist R, Brenner H, Mannermaa A, Hamann U, Meindl A, Lindblom A, Zheng W, Devillee P, Goldberg MS, Lubinski J, Kristensen V, Swerdlow A, Anton-Culver H, Dork T, Muir K, Matsuo K, Wu AH, Radice P, Teo SH, Shu XO, Blot W, Kang D, Hartman M, Sangrajrang S, Shen CY, Southey MC, Park DJ, Hammet F, Stone J, Veer LJ, Rutgers EJ, Lophatananon A, Stewart-Brown S, Siriwanarangsan P, Peto J, Schrauder MG, Ekici AB, Beckmann MW, Dos Santos SI, Johnson N, Warren H, Tomlinson I, Kerin MJ, Miller N, Marme F, Schneeweiss A, Sohn C, Truong T, Laurent-Puig P, Kerbrat P, Nordestgaard BG, Nielsen SF, Flyger H,Milne RL, Perez JI, Menendez P, Muller H, Arndt V, Stegmaier C, Lichtner P, Lochmann M, Justenhoven C, Ko YD, Gene EI, breast CN, Muranen TA, Aittomaki K, Blomqvist C, Greco D, Heikkinen T, Ito H, Iwata H, Yatabe Y, Antonenkova NN, Margolin S, Kataja V, Kosma VM, Hartikainen JM, Balleine R, kConFab I, Tseng CC, Berg DV, Stram DO, Neven P, Dieudonne AS, Leunen K, Rudolph A, Nickels S, Flesch-Janys D, Peterlongo P, Peissel B, Bernard L, Olson JE, Wang X, Stevens K, Severi G, Baglietto L, MClean C, Coetzee GA, Feng Y, Henderson BE, Schumacher F, Bogdanova NV, Labreche F, Dumont M, Yip CH, Taib NA, Cheng CY, Shrubsole M, Long J, Pylkas K, Jukkola-Vuorinen A, Kauppila S, Knight JA, Glendon G, Mulligan AM, Tollenaar RA, Seynaeve CM, Kriege M, Hooning MJ, van den Ouweland AM, van Deurzen CH, LuW, Gao YT, Cai H, Balasubramanian SP, Cross SS, ReedMW, Signorello L, Cai Q, ShahM, Miao H, Chan CW, Chia KS, Jakubowska A, Jaworska K, Durda K, Hsiung CN, Wu PE, Yu JC, Ashworth A, Jones M, Tessier DC, Gonzalez-Neira A, Pita G, Alonso MR, Vincent D, Bacot F, Ambrosone CB, Bandera EV, John EM, Chen GK, Hu JJ, Rodriguez-Gil JL, Bernstein L, Press MF, Ziegler RG, Millikan RM, Deming-Halverson SL, Nyante S, Ingles SA, Waisfisz Q, Tsimiklis H, Makalic E, Schmidt D, Bui M, Gibson L, Muller- Myhsok B, Schmutzler RK, Hein R, Dahmen N, Beckmann L, Aaltonen K, Czene K, Irwanto A, Liu J, Turnbull C, Familial Breast Cancer S, Rahman N, Meijers-Heijboer H, Uitterlinden AG, Rivadeneira F, Australian Breast Cancer Tissue Bank I, Olswold C, Slager S, Pilarski R, Ademuyiwa F, Konstantopoulou I, Martin NG, MontgomeryGW, Slamon DJ, Rauh C, LuxMP, Jud SM, Bruning T,Weaver J, Sharma P, Pathak H, TapperW, Gerty S, Durcan L, Trichopoulos D, Tumino R, Peeters PH, Kaaks R, Campa D, Canzian F, Weiderpass E, Johansson M, Khaw KT, Travis R, Clavel-Chapelon F, Kolonel LN, Chen C, Beck A, Hankinson SE, Berg CD, Hoover RN, Lissowska J, Figueroa JD, Chasman DI, Gaudet MM, Diver WR, Willett WC, Hunter DJ, Simard J, Benitez J, Dunning AM, Sherman ME, Chenevix- Trench G, Chanock SJ, Hall P, Pharoah PD, Vachon C, Easton DF, Haiman CA, Kraft P, 2013, Genome-wide association studies identify four ER negative-specific breast cancer risk loci. Nat Genet 45(4):392–398, 398e391-392. , DOI 10.1038/ng.2561 Zhou L, Zhang X, Li Z, Zhou C, Li M, Tang X, Lu C, Li H, Yuan Q, Yang M, 2013, Association of a genetic variation in a miR-191 binding site in MDM4 with risk of esophageal squamous cell carcinoma. PLoS One 8(5):e64331. , DOI 10.1371/journal. pone.0064331 Xu C, Zhu J, FuW, Liang Z, Song S, Zhao Y, Lyu L, Zhang A, He J, Duan P, 2016, MDM4 rs4245739 A > C polymorphism correlates with reduced overall cancer risk in a meta-analysis of 69477 subjects. Oncotarget 7(44):71718–71726. , DOI 10.18632/ oncotarget.12326 Gansmo LB, Romundstad P, Birkeland E, Hveem K, Vatten L, Knappskog S, Lonning PE, 2015, MDM4 SNP34091, rs4245739, and its effect on breast-, colon-, lung-, and prostate cancer risk. Cancer Med 4(12):1901–1907. , DOI 10. 1002/cam4.555 Heidenreich A, Bastian PJ, Bellmunt J, Bolla M, Joniau S, van der Kwast T, Mason M, Matveev V, Wiegel T, Zattoni F, Mottet N, European Association of U, 2014, EAU guidelines on prostate cancer. part 1: Screening, diagnosis, and local treatment with curative intent-update 2013. Eur Urol 65, 1):124–137., DOI https://doi. org/10.1016/j.eururo.2013.09.046 Sole X, Guino E, Valls J, Iniesta R, Moreno V, 2006, SNPStats: a web tool for the analysis of association studies. Bioinformatics 22(15):1928–1929. , DOI 10.1093/bioinformatics/btl268 Preskill C,Weidhaas JB, 2013, SNPs inmicroRNAbinding sites as prognostic and predictive cancer biomarkers. Crit Rev Oncog 18(4):327–340. , DOI 10.1615/critrevoncog.2013007254 Moszynska A, GebertM, Collawn JF, Bartoszewski R, 2017, SNPs in microRNA target sites and their potential role in human disease. Open Biol 7(4):170019. , DOI 10.1098/rsob.170019 Pelletier C, Weidhaas JB, 2010, MicroRNA binding site polymorphisms as biomarkers of cancer risk. Expert Rev Mol Diagn 10(6): 817–829. , DOI 10.1586/erm.10.59 Yu Z, Li Z, JolicoeurN, Zhang L, Fortin Y,Wang E, WuM, Shen SH, 2007, Aberrant allele frequencies of the SNPs located in microRNA target sites are potentially associated with human cancers. Nucleic Acids Res 35(13):4535–4541. , DOI 10. 1093/nar/gkm480 Ding WH, Ren KW, Yue C, Zou JG, Zuo L, Zhang LF, Bai Y, Okada A, Yasui T, Mi YY, 2018, Association between three genetic variants in kallikrein 3 and prostate cancer risk. Biosci Rep 38(6). , DOI 10.1042/BSR20181151 Zong H, Wang CC, Vaitheesvaran B, Kurland IJ, Hong W, Pessin JE, 2011, Enhanced energy expenditure, glucose utilization, and insulin sensitivity in VAMP8 null mice. Diabetes 60(1):30–38. , DOI 10.2337/db10-0231 Kote-Jarai Z, Olama AA, Giles GG, Severi G, Schleutker J, Weischer M, Campa D, Riboli E, Key T, Gronberg H, Hunter DJ, Kraft P, Thun MJ, Ingles S, Chanock S, Albanes D, Hayes RB, Neal DE, Hamdy FC, Donovan JL, Pharoah P, Schumacher F, Henderson BE, Stanford JL, Ostrander EA, Sorensen KD, Dork T, Andriole G, Dickinson JL, Cybulski C, Lubinski J, Spurdle A, Clements JA, Chambers S, Aitken J, Gardiner RA, Thibodeau SN, Schaid D, John EM, Maier C, Vogel W, Cooney KA, Park JY, Cannon-Albright L, Brenner H, Habuchi T, Zhang HW, Lu YJ, Kaneva R, Muir K, Benlloch S, Leongamornlert DA, Saunders EJ, Tymrakiewicz M, Mahmud N, Guy M, O'Brien LT, Wilkinson RA, Hall AL, Sawyer EJ, Dadaev T, Morrison J, Dearnaley DP, Horwich A, Huddart RA, Khoo VS, Parker CC, Van As N, Woodhouse CJ, Thompson A, Christmas T, Ogden C, Cooper CS, Lophatonanon A, Southey MC, Hopper JL, English DR, Wahlfors T, Tammela TL, Klarskov P, Nordestgaard BG, Roder MA, Tybjaerg-Hansen A, Bojesen SE, Travis R, Canzian F,KaaksR,Wiklund F,AlyM, Lindstrom S, DiverWR, Gapstur S, Stern MC, Corral R, Virtamo J, Cox A, Haiman CA, Le Marchand L, Fitzgerald L, Kolb S,Kwon EM,KaryadiDM,Orntoft TF, Borre M, Meyer A, Serth J, Yeager M, Berndt SI,Marthick JR, Patterson B, Wokolorczyk D, Batra J, Lose F, McDonnell SK, Joshi AD, Shahabi A, Rinckleb AE, Ray A, Sellers TA, Lin HY, Stephenson RA, Farnham J, Muller H, Rothenbacher D, Tsuchiya N, Narita S, Cao GW, Slavov C, Mitev V, Easton DF, Eeles RA, Oncology UKGPCSCBAoUSSo, Uk ProtecT Study Collaborators TAPCB, Consortium P, 2011, Seven prostate cancer susceptibility loci identified by a multi-stage genome-wide association study. Nat Genet 43(8):785–791. , DOI 10.1038/ng.882 Li Q, Lozano G, 2013, Molecular pathways: targeting Mdm2 and Mdm4 in cancer therapy. Clin Cancer Res 19(1):34–41. https://doi. org/10.1158/1078-0432.CCR-12-0053 Karni-Schmidt O, LokshinM, Prives C, 2016, The roles ofMDM2 and MDMX in Cancer. Annu Rev Pathol 11:617–644. https://doi. org/10.1146/annurev-pathol-012414-040349 Wynendaele J, Bohnke A, Leucci E, Nielsen SJ, Lambertz I, Hammer S, Sbrzesny N, Kubitza D, Wolf A, Gradhand E, Balschun K, Braicu I, Sehouli J, Darb-Esfahani S, Denkert C, Thomssen C, Hauptmann S, Lund A, Marine JC, Bartel F, 2010, An illegitimate microRNA target site within the 3' UTR of MDM4 affects ovarian cancer progression and chemosensitivity. Cancer Res 70(23):9641–9649. , DOI 10.1158/0008-5472.CAN- 10-0527 Eeles RA, Olama AA, Benlloch S, Saunders EJ, Leongamornlert DA, Tymrakiewicz M, Ghoussaini M, Luccarini C, Dennis J, Jugurnauth-Little S, Dadaev T, Neal DE, Hamdy FC, Donovan JL, Muir K, Giles GG, Severi G, Wiklund F, Gronberg H, Haiman CA, Schumacher F, Henderson BE, Le Marchand L, Lindstrom S, Kraft P, Hunter DJ, Gapstur S, Chanock SJ, Berndt SI, Albanes D, Andriole G, Schleutker J, Weischer M, Canzian F, Riboli E, Key TJ, Travis RC, CampaD, Ingles SA, John EM, Hayes RB, Pharoah PD, Pashayan N, Khaw KT, Stanford JL, Ostrander EA, Signorello LB, Thibodeau SN, Schaid D, Maier C, Vogel W, Kibel AS, Cybulski C, Lubinski J, Cannon-Albright L, Brenner H, Park JY, Kaneva R, Batra J, Spurdle AB, Clements JA, Teixeira MR, Dicks E, Lee A, Dunning AM, Baynes C, Conroy D, Maranian MJ, Ahmed S, Govindasami K, Guy M, Wilkinson RA, Sawyer EJ, Morgan A, Dearnaley DP, Horwich A, Huddart RA, Khoo VS, Parker CC, Van As NJ, Woodhouse CJ, Thompson A, Dudderidge T, Ogden C, Cooper CS, Lophatananon A, Cox A, Southey MC, Hopper JL, English DR, Aly M, Adolfsson J, Xu J, Zheng SL, YeagerM, Kaaks R, DiverWR, GaudetMM, SternMC, Corral R, Joshi AD, Shahabi A,Wahlfors T, Tammela TL, Auvinen A, Virtamo J, Klarskov P, Nordestgaard BG, Roder MA, Nielsen SF, Bojesen SE, Siddiq A, Fitzgerald LM, Kolb S, Kwon EM, Karyadi DM, Blot WJ, Zheng W, Cai Q, McDonnell SK, Rinckleb AE, Drake B, Colditz G, Wokolorczyk D, Stephenson RA, Teerlink C, Muller H, Rothenbacher D, Sellers TA, Lin HY, Slavov C,MitevV, Lose F, Srinivasan S,Maia S, Paulo P, Lange E, Cooney KA, Antoniou AC, Vincent D, Bacot F, Tessier DC, Initiative CO-CRUG-E, Australian Prostate Cancer B, Oncology UKGPCSCBAoUSSo, Collaborators UKPS, Consortium P, Kote- Jarai Z, Easton DF, 2013, Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array. Nat Genet 45(4):385–391, 391e381-382. , DOI 10.1038/ng. 2560 Fan C, Wei J, Yuan C, Wang X, Jiang C, Zhou C, Yang M, 2014, The functional TP53 rs1042522 and MDM4 rs4245739 genetic variants contribute to non-Hodgkin lymphoma risk. PLoS One 9(9):e107047. , DOI 10.1371/journal.pone.0107047 Hashemi M, Sanaei S, Hashemi SM, Eskandari E, Bahari G, 2018, Association of Single Nucleotide Polymorphisms of the MDM4 Gene with the susceptibility to breast Cancer in a southeast Iranian population sample. Clin Breast Cancer 18(5):e883–e891. , DOI 10.1016/j.clbc.2018.01.003 Mohammad Khanlou Z, Pouladi N, Hosseinpour Feizi M, Pedram N, 2017, Lack of associations of the MDM4 rs4245739 polymorphism with risk of thyroid Cancer among Iranian-Azeri patients: a case-control Study. Asian Pac J Cancer Prevent: APJCP 18(4): 1133–1138. , DOI 10.22034/APJCP.2017.18.4.1133 Pedram N, Pouladi N, Feizi MA, Montazeri V, Sakhinia E, Estiar MA, 2016, Analysis of the association betweenMDM4 rs4245739 single nucleotide polymorphism and breast Cancer susceptibility. Clin Lab 62(7):1303–1308. , DOI 10.7754/Clin.Lab.2016. 151128 Zhai Y, Dai Z, He H, Gao F, Yang L, Dong Y, Lu J, 2016, A PRISMA-compliant meta-analysis of MDM4 genetic variants and cancer susceptibility. Oncotarget 7(45):73935–73944. https://doi. org/10.18632/oncotarget.12558 Purrington KS, Slager S, Eccles D, Yannoukakos D, Fasching PA, Miron P, Carpenter J, Chang-Claude J, Martin NG, Montgomery GW, Kristensen V, Anton-Culver H, Goodfellow P, Tapper WJ, Rafiq S, Gerty SM, Durcan L, Konstantopoulou I, Fostira F, Vratimos A, Apostolou P, Konstanta I, Kotoula V, Lakis S, Dimopoulos MA, Skarlos D, Pectasides D, Fountzilas G, Beckmann MW, Hein A, Ruebner M, Ekici AB, Hartmann A, Schulz-Wendtland R, Renner SP, Janni W, Rack B, Scholz C, Neugebauer J, Andergassen U, Lux MP, Haeberle L, Clarke C, Pathmanathan N, Rudolph A, Flesch-Janys D, Nickels S, Olson JE, Ingle JN, Olswold C, Slettedahl S, Eckel-Passow JE, Anderson SK, Visscher DW, Cafourek VL, Sicotte H, Prodduturi N, Weiderpass E, Bernstein L, Ziogas A, Ivanovich J, Giles GG, Baglietto L, SoutheyM, KosmaVM, Fischer HP, Network G, Reed MW, Cross SS, Deming-Halverson S, Shrubsole M, Cai Q, Shu XO, Daly M, Weaver J, Ross E, Klemp J, Sharma P, Torres D, Rudiger T, Wolfing H, Ulmer HU, Forsti A, Khoury T, Kumar S, Pilarski R, Shapiro CL, Greco D, Heikkila P, Aittomaki K, Blomqvist C, Irwanto A, Liu J, Pankratz VS, Wang X, Severi G, Mannermaa A, Easton D, Hall P, Brauch H, Cox A, Zheng W, Godwin AK, Hamann U, Ambrosone C, Toland AE, Nevanlinna H, Vachon CM, Couch FJ, 2014, Genome-wide association study identifies 25 known breast cancer susceptibility loci as risk factors for triple-negative breast cancer. Carcinogenesis 35(5):1012–1019. , DOI 10.1093/carcin/bgt404 Stevens KN, Vachon CM, Couch FJ, 2013, Genetic susceptibility to triple-negative breast cancer. Cancer Res 73(7):2025–2030. , DOI 10.1158/0008-5472.CAN-12-1699 Liu J, Tang X, Li M, Lu C, Shi J, Zhou L, Yuan Q, YangM(2013, Functional MDM4 rs4245739 genetic variant, alone and in combination with P53 Arg72Pro polymorphism, contributes to breast cancer susceptibility. Breast Cancer Res Treat 140(1):151–157. , DOI 10.1007/s10549-013-2615-x NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2409 EP 2423 DI 10.1007/s12253-020-00839-7 PG 15 ER PT J TI A Three-microRNA Panel in Serum: Serving as a Potential Diagnostic Biomarker for Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Renal cell carcinoma; SerummiRNA; Biomarker; Non-invasive ID Renal cell carcinoma; SerummiRNA; Biomarker; Non-invasive AB Purpose: Renal cell carcinoma (RCC) accounts for about 120,000 death each year. Although surgery is a routine treatment, RCC could be fatal if not diagnosed at an early stage. This study aims to search for suitable serum biomarkers and construct a miRNA panel with high diagnostic sensitivity or specificity. Methods: Totally 146 RCC patients and 150 normal control were involved in this three-stage study. Serum expression levels of 30 miRNAs selected from literature were tested by reverse transcription quantitative PCR (RT-qPCR) in the screening stage, the testing stage, and the validation stage. The diagnostic efficiency of miRNAs was evaluated by receiver operating characteristic (ROC) curve and area under curve (AUC) analysis. A panel with the highest diagnostic efficiency was constructed by backward stepwise logistic regression analysis. Additionally, bioinformatics analysis was used to investigate potential biological functions and mechanisms of candidate miRNAs. Results: MiR-224-5p, miR-34b-3p, miR-129-2-3p and miR-182-5p with low to moderate diagnostic ability (AUC = 0.692, 0.778, 0.687 and 0.745, respectively) were selected as candidate miRNAs after the three-stage study. The final diagnostic panel was consisted by miR-224-5p, miR-34b-3p and miR-182-5p with AUC = 0.855. No significance has been found between these four miRNAs and tumor location, Fuhrman Grade and AJCC clinical stages of RCC. Bioinformatic analysis suggested that the three-miRNAs panel may participate in tumorigenesis of RCC by targeting CORO1C. Conclusions: The three-miRNA panel in serum could serve as a non-invasive diagnostic biomarker of RCC. CR Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM, 2010, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 127(12):2893–2917. https://doi. org/10.1002/ijc.25516 LinehanWM, Schmidt LS, Crooks DR,Wei D, Srinivasan R, Lang M, Ricketts CJ, 2019, The metabolic basis of kidney cancer. Cancer Discov 9(8):1006–1021. , DOI 10.1158/2159- 8290.cd-18-1354 Gago-Dominguez M, Yuan JM, Castelao JE, Ross RK, Yu MC, 2001, Family history and risk of renal cell carcinoma. Cancer Epidemiol Biomark Prev 10(9):1001–1004 Shen C, Beroukhim R, Schumacher SE, Zhou J, Chang M, Signoretti S, Kaelin WG, 2011, Genetic and functional studies implicate HIF1 as a 14q kidney cancer suppressor gene. Cancer Discov 1(3):222–235. , DOI 10.1158/2159-8290.cd-11- 0098 Srinivasan R, Ricketts CJ, Sourbier C, Linehan WM, 2015, New strategies in renal cell carcinoma: Targeting the genetic and metabolic basis of disease. Clin Cancer Res 21(1):10–17. , DOI 10.1158/1078-0432.ccr-13-2993 Sato Y, Yoshizato T, Shiraishi Y, Maekawa S, Okuno Y, Kamura T, Shimamura T, Sato-Otsubo A, Nagae G, Suzuki H, Nagata Y, Yoshida K, Kon A, Suzuki Y, Chiba K, Tanaka H, Niida A, Fujimoto A, Tsunoda T, Morikawa T, Maeda D, Kume H, Sugano S, Fukayama M, Aburatani H, Sanada M, Miyano S, Homma Y, Ogawa S, 2013, Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet 45(8):860–867. https:// doi.org/10.1038/ng.2699 Neal CS, Michael MZ, Rawlings LH, Van der Hoek MB, Gleadle JM, 2010, The VHL-dependent regulation of microRNAs in renal cancer. BMC Med 8:64. , DOI 10.1186/1741-7015-8-64 Baranwal S, Alahari SK, 2010, miRNA control of tumor cell invasion andmetastasis. Int J Cancer 126(6):1283–1290. , DOI 10.1002/ijc.25014 He YH, Chen C, Shi Z, 2018, The biological roles and clinical implications of microRNAs in clear cell renal cell carcinoma. J Cell Physiol 233(6):4458–4465. , DOI 10.1002/jcp.26347 Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A, Lin DW, Urban N, Drescher CW, Knudsen BS, Stirewalt DL, Gentleman R, Vessella RL, Nelson PS, Martin DB, Tewari M, 2008, Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A 105(30): 10513–10518. , DOI 10.1073/pnas.0804549105 Krazinski BE, Kiewisz J, Sliwinska-Jewsiewicka A, Kowalczyk AE, Grzegrzolka J, Godlewski J, Kwiatkowski P, Dziegiel P, Kmiec Z, 2019, Altered expression of DDR1 in clear cell renal cell carcinoma correlates with miR-199a/b-5p and patients’ outcome. Cancer Genomics Proteomics 16(3):179–193. , DOI 10. 21873/cgp.20124 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T), Method. Methods 25(4):402–408. , DOI 10.1006/ meth.2001.1262 Feng H, Ge F, Du L, Zhang Z, Liu D, 2019, MiR-34b-3p represses cell proliferation, cell cycle progression and cell apoptosis in nonsmall- cell lung cancer, NSCLC, by targeting CDK4. J Cell Mol Med 23(8):5282–5291. , DOI 10.1111/jcmm.14404 Zhu XL, Ren LF,Wang HP, Bai ZT, Zhang L,MengWB, Zhu KX, Ding FH, Miao L, Yan J, Wang YP, Liu YQ, Zhou WC, Li X, 2019, Plasma microRNAs as potential new biomarkers for early detection of early gastric cancer. World J Gastroenterol 25(13): 1580–1591. , DOI 10.3748/wjg.v25.i13.1580 Roman-Canal B, Moiola CP, Gatius S, Bonnin S, Ruiz-Miro M, Gonzalez E, Gonzalez-Tallada X, Llordella I, Hernandez I, Porcel JM, Gil-Moreno A, Falcon-Perez JM, Ponomarenko J,Matias-Guiu X, Colas E, 2019, EV-associated miRNAs from peritoneal lavage are a source of biomarkers in endometrial cancer. Cancers, Basel, 11(6). , DOI 10.3390/cancers11060839 Muinelo-Romay L, Casas-Arozamena C, Abal M, 2018, Liquid biopsy in endometrial cancer: New opportunities for personalized oncology. Int J Mol Sci 19(8). , DOI 10.3390/ ijms19082311 Chanudet E, Wozniak MB, Bouaoun L, Byrnes G, Mukeriya A, Zaridze D, Brennan P, Muller DC, Scelo G, 2017, Large-scale genome-wide screening of circulating microRNAs in clear cell renal cell carcinoma reveals specific signatures in late-stage disease. Int J Cancer 141(9):1730–1740. , DOI 10.1002/ijc.30845 Wang C, Hu J, Lu M, Gu H, Zhou X, Chen X, Zen K, Zhang CY, Zhang T, Ge J, Wang J, Zhang C, 2015, A panel of five serum miRNAs as a potential diagnostic tool for early-stage renal cell carcinoma. Sci Rep 5:7610. , DOI 10.1038/srep07610 QiY, WangL,Wang K, PengZ, MaY, Zheng Z, ShangD,XuW, Zheng J, 2019, New mechanistic insights of clear cell renal cell carcinoma fromintegratedmiRNAandmRNA expression profiling studies. Biomed Pharmacother 111:821–834. , DOI 10. 1016/j.biopha.2018.12.099 Jing ZF, Bi JB, Li Z, Liu X, Li J, Zhu Y, Zhang XT, Zhang Z, Li Z, Kong CZ, 2019, Inhibition of miR-34a-5p can rescue disruption of the p53-DAPK axis to suppress progression of clear cell renal cell carcinoma. Mol Oncol 13(10):2079–2097. , DOI 10.1002/ 1878-0261.12545 Vogt M, Munding J, Gruner M, Liffers ST, Verdoodt B, Hauk J, Steinstraesser L, Tannapfel A, Hermeking H, 2011, Frequent concomitant inactivation of miR-34a and miR-34b/c by CpG methylation in colorectal, pancreatic, mammary, ovarian, urothelial, and renal cell carcinomas and soft tissue sarcomas. Virchows Arch 458(3):313–322. , DOI 10.1007/s00428-010-1030-5 Shiomi E, Sugai T, Ishida K, Osakabe M, Tsuyukubo T, Kato Y, Takata R, Obara W, 2019, Analysis of expression patterns of MicroRNAs that are closely associated with renal carcinogenesis. Front Oncol 9:431. , DOI 10.3389/fonc.2019.00431 Xu X, Wu J, Li S, Hu Z, Xu X, Zhu Y, Liang Z, Wang X, Lin Y, Mao Y, Chen H, Luo J, Liu B, Zheng X, Xie L, 2014, Downregulation of microRNA-182-5p contributes to renal cell carcinoma proliferation via activating the AKT/FOXO3a signaling pathway. Mol Cancer 13:109. , DOI 10.1186/1476-4598- 13-109 Cao MQ, You AB, Zhu XD, Zhang W, Zhang YY, Zhang SZ, Zhang KW, Cai H, Shi WK, Li XL, Li KS, Gao DM, Ma DN, Ye BG,Wang CH, Qin CD, Sun HC, Zhang T, Tang ZY, 2018, miR- 182-5p promotes hepatocellular carcinoma progression by repressing FOXO3a. J Hematol Oncol 11(1):12. , DOI 10.1186/s13045-018-0555-y Kulkarni P, Dasgupta P, Bhat NS, Shahryari V, Shiina M, Hashimoto Y, Majid S, Deng G, Saini S, Tabatabai ZL, Yamamura S, Tanaka Y, Dahiya R, 2018, Elevated miR-182-5p associates with renal cancer cell mitotic arrest through diminished MALAT-1 expression.Mol Cancer Res 16(11):1750–1760. https:// doi.org/10.1158/1541-7786.MCR-17-0762 Cheng Y, Li Z, Xie J, Wang P, Zhu J, Li Y, Wang Y, 2018, MiRNA-224-5p inhibits autophagy in breast cancer cells via targeting Smad4. Biochem Biophys Res Commun 506(4):793– 798. , DOI 10.1016/j.bbrc.2018.10.150 Jiang Y, Zhang H, Li W, Yan Y, Yao X, Gu W, 2020, FOXM1- Activated LINC01094 promotes clear cell renal cell carcinoma development via MicroRNA 224-5p/CHSY1. Mol Cell Biol 40(3). , DOI 10.1128/MCB.00357-19 White NM, Bao TT, Grigull J, Youssef YM, Girgis A, Diamandis M, Fatoohi E,MetiasM, Honey RJ, Stewart R, Pace KT, Bjarnason GA, Yousef GM, 2011, miRNA profiling for clear cell renal cell carcinoma: biomarker discovery and identification of potential controls and consequences of miRNA dysregulation. J Urol 186(3): 1077–1083. , DOI 10.1016/j.juro.2011.04.110 Fan L, Wei Y, Ding X, Li B, 2019, Coronin3 promotes nasopharyngeal carcinoma migration and invasion by induction of epithelial-to-mesenchymal transition. Onco Targets Ther 12: 9585–9598. , DOI 10.2147/OTT.S215674 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2425 EP 2434 DI 10.1007/s12253-020-00842-y PG 10 ER PT J TI In Vivo Detection of CTC and CTC Plakoglobin Status Helps Predict Prognosis in Patients with Metastatic Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Circulating tumour cells; CellCollector; Metastatic breast cancer; Plakoglobin; Progression-free survival; Overall survival ID Circulating tumour cells; CellCollector; Metastatic breast cancer; Plakoglobin; Progression-free survival; Overall survival AB This study aims to detect the prognostic value of circulating tumour cell (CTC) in patients with metastatic breast cancer. In this study, 38 patients with metastatic breast cancer were enrolled. The in vivo CellCollector® method was used to detect the number of CTC in patients. Single CTC and CTC clusters were counted, and the expression of plakoglobin was also analysed. At baseline, 73.7% (28/38) of the patients were positive for ≥ 1 CTC (range, 1–14 cells). No CTC-like events were observed in the control group. Among the CTC-positive patients, 21.4% (6/28) of patients had CTC clusters, and 42.9% (12/28) of patients had plakoglobin-positive CTC. After chemotherapy, 48.6% (17/35) of the patients were positive for ≥ 1 CTC (range, 1–3 cells), of which 3 patients had CTC clusters, and 35.3% (6/17) had plakoglobin-positive CTC. Additionally, we found that the number of CTC clusters in plakoglobin-positive patients was much greater than that in plakoglobin-negative patients, and the number of CTC was associated with the number of sites of metastases. We also found that patients with ≥ 3 CTC at baseline had shorter progression-free survival (PFS) and overall survival (OS), and pre-chemotherapy CTC detection was associated with PFS (P = 0.0001) and OS (P = 0.0091). CTC plakoglobin expression was associated with PFS (P = 0.02) but not OS (P = 0.22). CTC collected by the in vivo CellCollector method in Chinese patients with metastatic breast cancer have prognostic significance. CTC plakoglobin expression may be associated with CTC clusters, and more in-depth studies are needed. CR Siegel RL, Miller KD, Jemal A, 2018, Cancer statistics, 2018. CA Cancer J Clin 68(1):7–30. , DOI 10.3322/caac.21442 DeSantis CE, Lin CC, Mariotto AB, Siegel RL, Stein KD, Kramer JL, Alteri R, Robbins AS, Jemal A, 2014, Cancer treatment and survivorship statistics, 2014. CA Cancer J Clin 64(4):252–271. , DOI 10.3322/caac.21235 Zhang L, Riethdorf S, Wu G, Wang T, Yang K, Peng G, Liu J, Pantel K, 2012, Meta-analysis of the prognostic value of circulating tumor cells in breast cancer. Clin Cancer Res 18(20):5701–5710. , DOI 10.1158/1078-0432.CCR-12-1587 Coumans FA, Siesling S, Terstappen LW, 2013, Detection of cancer before distant metastasis. BMC Cancer 13:283. , DOI 10.1186/1471-2407-13-283 Meng S, Tripathy D, Frenkel EP, Shete S, Naftalis EZ, Huth JF, Beitsch PD, Leitch M, Hoover S, Euhus D, Haley B, Morrison L, Fleming TP, Herlyn D, Terstappen LW, Fehm T, Tucker TF, Lane N, Wang J, Uhr JW, 2004, Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res 10(24):8152–8162. , DOI 10.1158/1078-0432.CCR-04-1110 Deryugina EI, Kiosses WB, 2017, Intratumoral cancer cell intravasation can occur independent of invasion into the adjacent stroma. Cell Rep 19(3):601–616. , DOI 10.1016/j.celrep.2017.03. 064 Chambers AF, Groom AC, MacDonald IC, 2002, Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer 2(8): 563–572. , DOI 10.1038/nrc865 Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: the next generation. Cell 144(5):646–674. , DOI 10.1016/j.cell. 2011.02.013 Wang H, Stoecklein NH, Lin PP, Gires O, 2017, Circulating and disseminated tumor cells: diagnostic tools and therapeutic targets in motion. Oncotarget 8(1):1884–1912. , DOI 10.18632/ oncotarget.12242 Jiang ZF, CristofanilliM, Shao ZM, Tong ZS, SongEW,Wang XJ, Liao N, Hu XC, Liu Y, Wang Y, Zeng L, Zhang M, 2013, Circulating tumor cells predict progression-free and overall survival in Chinese patients with metastatic breast cancer, HER2-positive or triple-negative, CBCSG004): a multicenter, double-blind, prospective trial. Ann Oncol 24(11):2766–2772. , DOI 10.1093/ annonc/mdt246 Giordano A, Giuliano M, De Laurentiis M, Arpino G, Jackson S, Handy BC, Ueno NT, Andreopoulou E, Alvarez RH, Valero V, De Placido S, Hortobagyi GN, Reuben JM, Cristofanilli M, 2012, Circulating tumor cells in immunohistochemical subtypes of metastatic breast cancer: lack of prediction in HER2-positive disease treated with targeted therapy. Ann Oncol 23(5):1144–1150. , DOI 10.1093/annonc/mdr434 Liu XR, Shao B, Peng JX, Li HP, Yang YL, Kong WY, Song GH, Jiang HF, Liang X, Yan Y, 2017, Identification of high independent prognostic value of nanotechnology based circulating tumor cell enumeration in first-line chemotherapy for metastatic breast cancer patients. Breast 32:119–125. , DOI 10.1016/j. breast.2017.01.007 Iwata H, Masuda N, Yamamoto D, Sagara Y, Sato N, Yamamoto Y, SaitoM, Fujita T, Oura S,Watanabe J, Tsukabe M, Horiguchi K, Hattori S, Matsuura Y, Kuroi K, 2017, Circulating tumor cells as a prognosticmarker for efficacy in the randomized phase III JO21095 trial in Japanese patients with HER2-negative metastatic breast cancer. Breast Cancer Res Treat 162(3):501–510. , DOI 10. 1007/s10549-017-4138-3 Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, Washington MK, Gershenwald JE, Compton CC, Hess KR, Sullivan DC, Jessup JM, Brierley JD, Gaspar LE, Schilsky RL, Balch CM, Winchester DP, Asare EA, Madera M, Gress DM, Meyer LR, 2017, AJCC cancer staging manual, 8th edn. Springer, Boston Alix-Panabieres C, Pantel K, 2014, Challenges in circulating tumour cell research. Nat Rev Cancer 14(9):623–631. , DOI 10.1038/nrc3820 Saucedo-Zeni N, Mewes S, Niestroj R, Gasiorowski L, Murawa D, Nowaczyk P, Tomasi T,Weber E, Dworacki G,Morgenthaler NG, Jansen H, Propping C, Sterzynska K, Dyszkiewicz W, Zabel M, Kiechle M, Reuning U, Schmitt M, Lucke K, 2012, A novel method for the in vivo isolation of circulating tumor cells fromperipheral blood of cancer patients using a functionalized and structured medical wire. Int J Oncol 41(4):1241–1250. , DOI 10.3892/ijo. 2012.1557 Li JB, Geng CZ, YanM,Wang YS, Ouyang QC, YinYM, Wu LN, He J, Jiang ZF, 2017, Circulating tumor cells in patients with breast tumors were detected by a novel device: a multicenter, clinical trial in China. Zhonghua Yi Xue Za Zhi 97(24):1857–1861. https://doi. org/10.3760/cma.j.issn.0376-2491.2017.24.003 Aceto N, Bardia A, Miyamoto DT, Donaldson MC, Wittner BS, Spencer JA, Yu M, Pely A, Engstrom A, Zhu H, Brannigan BW, Kapur R, Stott SL, Shioda T, Ramaswamy S, Ting DT, Lin CP, Toner M, Haber DA, Maheswaran S, 2014, Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 158(5):1110–1122. , DOI 10.1016/j.cell.2014.07.013 Lu L, Zeng H, Gu X,MaW(2015, Circulating tumor cell clustersassociated gene plakoglobin and breast cancer survival. Breast Cancer Res Treat 151(3):491–500. , DOI 10.1007/ s10549-015-3416-1 Aktary Z, Alaee M, Pasdar M, 2017, Beyond cell-cell adhesion: Plakoglobin and the regulation of tumorigenesis and metastasis. Oncotarget 8(19):32270–32291. , DOI 10.18632/ oncotarget.15650 Heller G, McCormack R, Kheoh T, Molina A, Smith MR, Dreicer R, Saad F, de Wit R, Aftab DT, Hirmand M, Limon A, Fizazi K, Fleisher M, de Bono JS, Scher HI, 2018, Circulating tumor cell number as a response measure of prolonged survival for metastatic castrationresistant prostate cancer: A comparison with prostate-specific antigen across five randomized phase III clinical trials. J Clin Oncol 36(6):572– 580. , DOI 10.1200/JCO.2017.75.2998 Bidard FC, Mathiot C, Delaloge S, Brain E, Giachetti S, de Cremoux P, Marty M, Pierga JY, 2010, Single circulating tumor cell detection and overall survival in nonmetastatic breast cancer. Ann Oncol 21(4):729–733. , DOI 10.1093/annonc/ mdp391 Riethdorf S,Muller V, Loibl S, Nekljudova V,Weber K, Huober J, Fehm T, Schrader I, Hilfrich J, Holms F, Tesch H, Schem C, von Minckwitz G, Untch M, Pantel K, 2017, Prognostic impact of circulating tumor cells for breast cancer patients treated in the neoadjuvant “Geparquattro” trial. Clin Cancer Res 23(18):5384–5393. , DOI 10.1158/1078-0432.CCR-17-0255 Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, Reuben JM,DoyleGV,Allard WJ, Terstappen LW, Hayes DF, 2004, Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl JMed 351(8):781–791. https://doi. org/10.1056/NEJMoa040766 Smerage JB, Barlow WE, Hortobagyi GN, Winer EP, Leyland- Jones B, Srkalovic G, Tejwani S, Schott AF, O’Rourke MA, Lew DL, Doyle GV, Gralow JR, Livingston RB, Hayes DF, 2014, Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J Clin Oncol 32(31):3483–3489. , DOI 10.1200/JCO.2014.56.2561 Gkountela S, Castro-Giner F, Szczerba BM, Vetter M, Landin J, Scherrer R, Krol I, Scheidmann MC, Beisel C, Stirnimann CU, Kurzeder C, Heinzelmann-Schwarz V, Rochlitz C, Weber WP, Aceto N, 2019, Circulating tumor cell clustering shapes DNA methylation to enable metastasis seeding. Cell 176(1–2):98–112 e114. , DOI 10.1016/j.cell.2018.11.046 Rodrigues P, Vanharanta S, 2019, Circulating tumor cells: Come together, right now, over metastasis. Cancer Discov 9(1):22–24. , DOI 10.1158/2159-8290.CD-18-1285 Gorges TM, Penkalla N, Schalk T, Joosse SA, Riethdorf S, Tucholski J, Lucke K, Wikman H, Jackson S, Brychta N, von Ahsen O, Schumann C, Krahn T, Pantel K, 2016, Enumeration and molecular characterization of tumor cells in lung cancer patients using a novel in vivo device for capturing circulating tumor cells. Clin Cancer Res 22(9):2197–2206. , DOI 10.1158/ 1078-0432.CCR-15-1416 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2435 EP 2442 DI 10.1007/s12253-020-00847-7 PG 8 ER PT J AU Sejben, A Nyari, T Zombori, T Cserni, G AF Sejben, Anita Nyari, Tibor Zombori, Tamas Cserni, Gabor TI Comparison of Nottingham Prognostic Index, PREDICT and PrognosTILs in Triple Negative Breast Cancer –a Retrospective Cohort Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Triple negative breast cancer; Nottingham Prognostic Index; Predict; Tumor infiltrating lymphocytes; PrognosTILs; Prognosis ID Triple negative breast cancer; Nottingham Prognostic Index; Predict; Tumor infiltrating lymphocytes; PrognosTILs; Prognosis AB Triple-negative breast cancer (TNBC) represents a heterogenous subtype of breast cancer with generally poor prognosis. The prediction of its prognosis remains essential to clinicians in their therapeutical decision-making process. The aimof our study was to compare the validity of three multivariable analysis derived prognostic systems, the Nottingham Prognostic Index (NPI), PREDICT and PrognosTILs (a prognosticator including tumor infiltrating lymphocytes, TILs) in a series of TNBCs. Patients operated on with TNBC at the Department of Surgery, Bacs-Kiskun County Teaching Hospital, Kecskemet between 2005 and 2016 were included. Clinical and pathological parameters and follow-up data were collected from medical charts. TILs were assessed retrospectively, following international recommendations. Estimated survivals of PrognosTILs, PREDICT and NPI were recorded and compared with real outcomes. Altogether 136 patients were included in this retrospective study. In univariate Cox analysis, type of surgery, pT, pN, stage, NPI and type of adjuvant therapy were the significant prognostic variables. The multivariate Cox-regression strengthened that NPI is an independent predictor of overall and disease-free survivals in TNBCs. The NPI, PREDICT and PrognosTILs could be compared directly only in a ROC curve analysis: the sensitivities and specificities of these predicting systems are rather similar with area under the curve values falling between 0.7 and 0.8, and NPI having the highest values. Our findings reflect the diverse prognosis of TNBC and highlight the difficulties of predicting its outcome. None of the three multivariable prognosticators is inferior to the others, the NPI can reliably be used for TNBCs. C1 [Sejben, Anita] University of Szeged, Department of Pathology, Allomas u. 1, 6725 Szeged, Hungary. [Nyari, Tibor] University of Szeged, Department of Medical InformaticsSzeged, Hungary. [Zombori, Tamas] University of Szeged, Department of Pathology, Allomas u. 1, 6725 Szeged, Hungary. [Cserni, Gabor] University of Szeged, Department of Pathology, Allomas u. 1, 6725 Szeged, Hungary. RP Sejben, A (reprint author), University of Szeged, Department of Pathology, 6725 Szeged, Hungary. EM sejben.anita@gmail.com CR Aysola K, Desai A,Welch C, Xu J, Qin Y, Reddy V, Matthews R, Owens C, Okoli J, Beech DJ, Piyathilake CJ, Reddy SP, Rao VN, 2013, Triple negative breast cancer – an overview. Hereditary Genet Suppl 2:001. , DOI 10.4172/2161-1041.S2-001 WHO Classification of Tumours Editorial Board, ed,, 2019)WHO classification of tumours, 5th edition – breast tumours. International Agency for Research on Cancer, Lyon Siddharth S, Sharma D, 2018, Racial disparity and triple-negative breast cancer in african-american women: a multifaceted affair between obesity, biology, and socioeconomic determinants. Cancers, Basel, 10:514. , DOI 10.3390/cancers10120514 Anders C, Carey LA, 2008, Understanding and treating triplenegative breast cancer. Oncology 22:1233–1243 Al-Mahmood S, Sapiezynski J, Garbuzenko OB, Minko T, 2018, Metastatic and triple-negative breast cancer: challenges and treatment options. Drug Deliv Transl Res 8:1483–1507. , DOI 10.1007/s13346-018-0551-3 Park JH, Ahn JH, Kim SB, 2018, How shall we treat early triplenegative breast cancer, TNBC): from the current standard to upcoming immuno-molecular strategies. ESMO Open 3(3):e000357. , DOI 10.1136/esmoopen-2018-000357 Wahba HA, El-Hadaad HA, 2015, Current approaches in treatment of triple-negative breast cancer. Cancer Biol Med 12:106–116. , DOI 10.7497/j.issn.2095-3941.2015.0030 He MY, Rancoule C, Rehailia-Blanchard A, Espenel S, Trone JC, Bernichon E, Guillaume E, Vallard A, Magne N, 2018, Radiotherapy in triple-negative breast cancer: current situation and upcoming strategies. Crit Rev Oncol Hematol 131:96–101. , DOI 10.1016/j.critrevonc.2018.09.004 Lebert JM, Lester R, Powell E, Seal M, McCarthy J, 2018, Advances in the systemic treatment of triple-negative breast cancer. Curr Oncol 25:S142–S150. , DOI 10.3747/co.25.3954 Fong Y, Evans J, Brook D, Kenkre J, Jarvis P, Gower-Thomas K, 2015, The Nottingham prognostic index: five- and ten-year data for all-cause survival within a screened population. Ann R Coll Surg Engl 97:137–139. , DOI 10.1308/003588414X14055925060514 Ovcaricek T, Frkovic SG, Matos E, Mozina B, Bostnar S, 2011, Triple negative breast cancer – prognostic factors and survival. Radiol Oncol 45:46–52. , DOI 10.2478/v10019-010-0054-4 Urru SAM, Gallus S, Bosetti C,Moi T,Medda R, Sollai E, Murgia A, Sanges F, Pira G, Manca A, Palmas D, Floris M, Asunis AM, Atzori F, Carru C, D’Incalci M, Ghiani M, Marras V, Onnis D, Santona MC, Sarobba G, Valle E, Canu L, Cossu S, Bulfone A, Rocca PC, De Miglio MR, Orru S, 2018, Clinical and pathological factors influencing survival in a large cohort of triple-negative breast cancer patients. BMC Cancer 18:56. , DOI 10. 1186/s12885-017-3969-y Asaga S, Kinoshita T, Hojo T, Suzuki J, Jimbo K, Tsuda H, 2013, Prognostic factors for triple-negative breast cancer patients receiving preoperative systemic chemotherapy. Clin Breast Cancer 13: 40–46. , DOI 10.1016/j.clbc.2012.09.013 Haybittle JL, Blamey RW, Elston CW, Johnson J, Doyle PJ, Campbell FC, Nicholson RI, Griffiths K, 1982, A prognostic index in primary breast cancer. Br J Cancer 45:361–366. , DOI 10.1038/bjc.1982.62 Galea MH, Blamey RW, Elston CE, Ellis IO, 1992, The Nottingham prognostic index in primary breast cancer. Breast Cancer Res Treat 22:207–219. , DOI 10.1007/ BF01840834 Lee AH, Ellis IO, 2008, The Nottingham prognostic index for invasive carcinoma of the breast. Pathol Oncol Res 14:113–115. , DOI 10.1007/s12253-008-9067-3 Gray E, Marti J, Brewster DH, Wyatt JC, Hall PS, SATURNE Advisory Group, 2018, Independent validation of the PREDICT breast cancer prognosis prediction tool in 45,789 patients using Scottish Cancer Registry data. Br J Cancer 119:808–814. https:// doi.org/10.1038/s41416-018-0256-x Wishart GC, Azzato EM, Greenberg DC, Rashbass J, Kearins O, Lawrence G, Caldas C, Pharoah PD, 2010, PREDICT: a new UK prognostic model that predicts survival following surgery for invasive breast cancer. Breast Cancer Res 12:R1. , DOI 10. 1186/bcr2464 https://breast.predict.nhs.uk/predict_v2.0.html. Accessed 28 May 2020 Candido Dos Reis FJ, Wishart GC, Dicks EM, Greenberg D, Rashbass J, Schmidt MK, van den Broek AJ, Ellis IO, Green A, Rakha E, Maishman T, Eccles DM, Pharoah PDP, 2017, An updated PREDICT breast cancer prognostication and treatment benefit prediction model with independent validation. Breast Cancer Res 19:58. , DOI 10.1186/s13058-017-0852-3 Maishman T, Copson E, Stanton L, Gerty S, Dicks E, Durcan L, Wishart GC, Pharoah P, POSH Steering Group; Eccles D, 2015, An evaluation of the prognostic model PREDICT using the POSH cohort of women aged ⩽40 years at breast cancer diagnosis. Br J Cancer 112:983–991. , DOI 10.1038/bjc.2015.57 Garcia-Teijido P, Cabal ML, Fernandez IP, Perez YF, 2016, Tumor-infiltrating lymphocytes in triple-negative breast cancer: the future of immune targeting. Clin Med Insights Oncol 10:31– 39. , DOI 10.4137/CMO.S34540 Disis ML, Stanton SE, 2015, Triple-negative breast cancer: immune modulation as the new treatment paradigm. Am Soc Clin Oncol Educ Book 2015:25–30. , DOI 10.14694/EdBook_ AM.2015.35.e25 Loi S, Drubay D, Adams S, Pruneri G, Francis PA, Lacroix-Triki M, Joensuu H, Dieci MV, Badve S, Demaria S, Gray R, Munzone E, Lemonnier J, Sotiriou C, Piccart MJ, Kellokumpu-Lehtinen PL, Vingiani A, Gray K, Andre F, Denkert C, Salgado R, Michiels S, 2019, Tumor-infiltrating lymphocytes and prognosis: a pooled individual patient analysis of early-stage triple-negative breast cancers. J Clin Oncol 37:559–569. , DOI 10.1200/JCO.18. 01010 https://cesp-proxy2.vjf.inserm.fr/shiny/prognosTILs/. Accessed 28 May 2020 Hendry S, Salgado R, Gevaert T, Russell PA, John T, Thapa B, Christie M, van de Vijver K, Estrada MV, Gonzalez-Ericsson PI, Sanders M, Solomon B, Solinas C, Van den Eynden GGGM, Allory Y, Preusser M, Hainfellner J, Pruneri G, Vingiani A, Demaria S, Symmans F, Nuciforo P, Comerma L, Thompson EA, Lakhani S, Kim SR, Schnitt S, Colpaert C, Sotiriou C, Scherer SJ, Ignatiadis M, Badve S, Pierce RH, Viale G, Sirtaine N, Penault- Llorca F, Sugie T, Fineberg S, Paik S, Srinivasan A, Richardson A, Wang Y, Chmielik E, Brock J, Johnson DB, Balko J, Wienert S, Bossuyt V, Michiels S, Ternes N, Burchardi N, Luen SJ, Savas P, Klauschen F, Watson PH, Nelson BH, Criscitiello C, O'Toole S, Larsimont D, deWind R, Curigliano G, Andre F, Lacroix-Triki M, van de Vijver M, Rojo F, Floris G, Bedri S, Sparano J, Rimm D, Nielsen T, Kos Z, Hewitt S, Singh B, Farshid G, Loibl S, Allison KH, Tung N, Adams S,Willard-Gallo K, Horlings HM, Gandhi L, Moreira A, Hirsch F, Dieci MV, UrbanowiczM, Brcic I, Korski K, Gaire F, Koeppen H, Lo A, Giltnane J, Rebelatto MC, Steele KE, Zha J, Emancipator K, Juco JW, Denkert C, Reis-Filho J, Loi S, Fox SB, 2017, Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the International Immunooncology Biomarkers Working Group: part 1: assessing the host immune response, TILs in invasive breast carcinoma and ductal carcinoma in situ,metastatic tumor deposits and areas for further research. Adv Anat Pathol 24:235–251. , DOI 10.1097/PAP. 0000000000000162 http://virtuelle-mikroskopie.de/TIL-training/. Accessed 28 May 2020 Denkert C, Wienert S, Poterie A, Loibl S, Budczies J, Badve S, Bago-Horvath Z, Bane A, Bedri S, Brock J, Chmielik E, Christgen M, Colpaert C, Demaria S, Van den Eynden G, Floris G, Fox SB, Gao D, Ingold Heppner B, Kim SR, Kos Z, Kreipe HH, Lakhani SR, Penault-Llorca F, Pruneri G, Radosevic-Robin N, Rimm DL, Schnitt SJ, Sinn BV, Sinn P, Sirtaine N, O'Toole SA, Viale G, Van de Vijver K, deWind R, vonMinckwitz G, Klauschen F, Untch M, Fasching PA, Reimer T, Willard-Gallo K, Michiels S, Loi S, Salgado R, 2016, Standardized evaluation of tumor-infiltrating lymphocytes in breast cancer: results of the ring studies of the international Immuno-oncology biomarker working group. Mod Pathol 29:1155–1164. , DOI 10.1038/modpathol.2016.109 Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehner FL, Penault-Llorca F, Perez EA, Thompson EA, SymmansWF, Richardson AL, Brock J, Criscitiello C, Bailey H, Ignatiadis M, Floris G, Sparano J, Kos Z, Nielsen T, Rimm DL, Allison KH, Reis-Filho JS, Loibl S, Sotiriou C, Viale G, Badve S, Adams S, Willard-Gallo K, Loi S, International TILs Working Group 2014, 2015, The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: Recommendations by an International TILs Working Group 2014. Ann Oncol 26:259–271. , DOI 10.1093/annonc/ mdu450 Amin MB, Edge SB, Greene FL, Schilsky RL, Gaspar LE, Washington MK, Sullivan DC, Brookland RK, Brierley JD, Balch CM, Compton CC, Hess KR, Gershenwald JE, Jessup JM, Byrd DR,Winchester DP, Madera M, Asare EA, Madera M, Gress DM, Meyer LR, eds,, 2017, AJCC Cancer staging manual, 8th edn. Berlin, Springer Nature Foschini MP, Asioli S, Foreid S, Cserni G, Ellis IO, Eusebi V, Rosai J, 2017, Solid papillary breast carcinomas resembling the tall cell variant of papillary thyroid neoplasms: a unique invasive tumor with indolent behavior. Am J Surg Pathol 41:887–895. https://doi. org/10.1097/PAS.0000000000000853 Horowitz DP, Sharma CS, Connolly E, Gidea-Addeo D, Deutsch I, 2012, Secretory carcinoma of the breast: results from the survival, epidemiology and end results database. Breast 21:350–353. https:// doi.org/10.1016/j.breast.2012.02.013 Kulkarni N, Pezzi CM, Greif JM, Suzanne Klimberg V, Bailey L, Korourian S, Zuraek M Rare breast cancer: 933 adenoid cystic carcinomas from the National Cancer Data Base. Ann Surg Oncol 20:2236–2241. , DOI 10.1245/s10434-013-2911-z Gao GX, Wang ZH, Qu X, Zhang ZT, 2020, Prognostic value of tumor-infiltrating lymphocytes in patients with triple-negative breast cancer: a systematic review and meta-analysis. BMC Cancer 20:179. , DOI 10.1186/s12885-020-6668-z Todd JH, Dowle C,WilliamsMR, ElstonCW, Ellis IO, Hinton CP, Blamey RW, Haybittle JL, 1987, Confirmation of a prognostic index in primary breast cancer. Br J Cancer 56:489–492. https:// doi.org/10.1038/bjc.1987.230 Balslev I, Axelsson CK, Zedeler K, Rasmussen BB, Carstensen B, Mouridsen HT, 1994, The Nottingham prognostic index applied to 9,149 patients from the studies of the Danish breast Cancer cooperative group, DBCG). Breast Cancer Res Treat 32:281–290. , DOI 10.1007/BF00666005 Sundquist M, Thorstenson S, Brudin L, Nordenskjold B, 1999, Applying the Nottingham prognostic index to a swedish breast cancer population. South east Swedish breast Cancer study group. Breast Cancer Res Treat 53:1–8. , DOI 10.1023/a: 1006052115874 D'Eredita G, Giardina C, Martellotta M, Natale T, Ferrarese F, 2001, Prognostic factors in breast cancer: the predictive value of the Nottingham prognostic index in patients with a long-term follow- up that were treated in a single institution. Eur J Cancer 37: 591–596. , DOI 10.1016/s0959-8049(00)00435-4 Blamey RW, Ellis IO, Pinder SE, Lee AH,Macmillan RD,Morgan DA, Robertson JF,MitchellMJ, Ball GR, Haybittle JL, Elston CW, 2007, Survival of invasive breast cancer according to the Nottingham prognostic index in cases diagnosed in 1990-1999. Eur J Cancer 43:1548–1555. , DOI 10.1016/j.ejca.2007. 01.016 Aguirre U, Garcia-Gutierrez S, Romero A, Domingo L, Castells X, Sala M, CAMISS Study Group, 2019, External validation of the PREDICT tool in Spanish women with breast cancer participating in population-based screening programmes. J Eval Clin Pract 25: 873–880. , DOI 10.1111/jep.13084 Albergaria A, Ricardo S, Milanezi F, Carneiro V, Amendoeira I, Vieira D, Cameselle-Teijeiro J, Schmitt F, 2011, Nottingham prognostic index in triple-negative breast cancer: a reliable prognostic tool? BMC Cancer 11:299. , DOI 10.1186/1471-2407-11- 299 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2443 EP 2450 DI 10.1007/s12253-020-00846-8 PG 8 ER PT J AU Zombori, T Sejben, A Tiszlavicz, L Cserni, G Palfoldi, R Csada, E Furak, J AF Zombori, Tamas Sejben, Anita Tiszlavicz, Laszlo Cserni, Gabor Palfoldi, Regina Csada, Edit Furak, Jozsef TI Architectural Grade Combined With Spread Through Air Spaces (STAS) Predicts Recurrence and is Suitable for Stratifying Patients Who Might Be Eligible for Lung Sparing Surgery for Stage I Adenocarcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Lung adenocarcinoma; Spread through airspaces (STAS); Architectural grade; Lung sparing surgery; Sublobar resection ID Lung adenocarcinoma; Spread through airspaces (STAS); Architectural grade; Lung sparing surgery; Sublobar resection AB The spread through air spaces (STAS) has a main role in local recurrence of stage I lung adenocarcinomas (LAs), therefore its presence might question sublobar resection as a therapeutic option. The aimof our study was to evaluate the distribution of STAS in stage I LAs, to stratify patients according to local recurrence and to identify a group of patients who might be suitable for sublobar surgery. Patients resected with LA were included. The presence of STAS was recorded on hematoxylin eosin stained slides and clinicopathological data were obtained from medical charts. Overall survival (OS) and disease-free survival (DFS) were registered. Statistical methods included Kruskal-Wallis tests, Kaplan-Meier analyses, log-rank tests and Cox-regressions. 292 patients were included. STAS was identified in 38.7% and 95.7% of micropapillary carcinomas showed STAS. Significant correlation was found between STAS and high-grade patterns. Significant differences were found between OS and DFS estimates of STAS0 and STAS1 cases (5-y-OS: 80.0%vs. 68.4%; 5-y-DFS: 71.1%vs. 57.1%). The presence of STAS was associated with unfavorable prognosis in low and intermediate architectural grades, but not in high-grade. Multivariate analysis revealed that architectural grade (HR(OS):2.09; HR(DFS):1.52) and STAS (HR(OS):1.51; HR(DFS):1.48) were independent prognostic markers in stage I LA. Architectural grade combined with STAS was superior to other prognostic grades. The combination of architectural grade and STAS proved to be a prognostic factor that is superior to previously introduced grading systems. Patients having low and intermediate grade LAs without STAS might be eligible for sublobar resection. C1 [Zombori, Tamas] University of Szeged, Department of Pathology, Allomas u. 1., H6725 Szeged, Hungary. [Sejben, Anita] University of Szeged, Department of Pathology, Allomas u. 1., H6725 Szeged, Hungary. [Tiszlavicz, Laszlo] University of Szeged, Department of Pathology, Allomas u. 1., H6725 Szeged, Hungary. [Cserni, Gabor] University of Szeged, Department of Pathology, Allomas u. 1., H6725 Szeged, Hungary. [Palfoldi, Regina] Csongrad County Hospital of Chest Diseases, Alkotmany u. 36., H6772 Deszk, Hungary. [Csada, Edit] Csongrad County Hospital of Chest Diseases, Alkotmany u. 36., H6772 Deszk, Hungary. [Furak, Jozsef] University of Szeged, Department of Surgery, Semmelweis u. 8., H6720 Szeged, Hungary. RP Zombori, T (reprint author), University of Szeged, Department of Pathology, H6725 Szeged, Hungary. EM zomtam@gmail.com CR Siegel RL, Miller KD, Jemal A, 2019, Cancer statistics, 2019. CA Cancer J Clin 69:7–34. , DOI 10.3322/caac.21551 Toyokawa G, Yamada Y, Tagawa T, Oda Y, 2018, Significance of spread through air spaces in early-stage lung adenocarcinomas undergoing limited resection. Thorac Cancer 9:1255–1261. https:// doi.org/10.1111/1759-7714.12828 Ginsberg RJ, Rubinstein LV, 1995, Randomized trial of lobectomy versus limited resection for T1 N0 non-small cell lung cancer. Lung Cancer Study Group. Ann Thorac Surg 60:615–622 Aokage K, Yoshida J, Hishida T, Tsuboi M, Saji H, Okada M, Suzuki K, Watanabe S, Asamura H, 2017, Limited resection for early-stage non-small cell lung cancer as function-preserving radical surgery: a review. Jpn J Clin Oncol 47:7–11. , DOI 10. 1093/jjco/hyw148 Suzuki K, Watanabe S, Wakabayashi M, Moriya Y, Yoshino I, Tsuboi M, Mitsudomi T, Asamura H, 2017, A nonrandomized confirmatory phase III study of sublobar surgical resection for peripheral ground glass opacity dominant lung cancer defined with thoracic thin-section computed tomography, JCOG0804/ WJOG4507L). J Clin Oncol 35: Abstract 8561 Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, BeasleyMB, Chirieac LR,Dacic S, Duhig E, FliederDB,Geisinger K, Hirsch FR, Ishikawa Y, Kerr KM, NoguchiM, Pelosi G, Powell CA, Tsao MS, Wistuba I, WHO Panel, 2015, The 2015 World Health Organization classification of lung tumors: Impact of genetic, clinical and radiologic advances since the 2004 Classification. J Thorac Oncol 10:1243–1260. , DOI 10.1097/JTO. 0000000000000630 Kadota K, Nitadori J, Sima CS, Ujiie H, Rizk NP, Jones DR, Adusumilli PS, TravisWD, 2015, Tumor spread through air spaces is an important pattern of invasion and impacts the frequency and location of recurrences after limited resection for small stage I lung adenocarcinomas. J Thorac Oncol 10:806–814. , DOI 10. 1097/JTO.0000000000000486 Warth A, 2017, Spread through air spaces, STAS): a comprehensive update. Transl Lung Cancer Res 6:501–507. , DOI 10. 21037/tlcr.2017.06.08 TravisWD, Brambilla E, Burke AP, Marx A, NicholsonAG, 2015, WHO classification of tumours of the lung, pleura, thymus and heart. Fourth edition. International Agency for Research on Cancer, Lyon Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, Washington MK, Gershenwald JE, Compton CC, Hess KR, Sullivan DC, Jessup JM, Brierley JD, Gaspar LE, Schilsky RL, Balch CM, Winchester DP, Asare EA, Madera M, Gress DM, Meyer LR, 2018, AJCC Cancer Staging Manual. Vol xvii. 8th ed. Springer International Publishing, Cham Shih AR,Mino-KenudsonM(2020, Updates on spread through air spaces, STAS, in lung cancer. Histopathology. , DOI 10. 1111/his.14062 Shiono S, Yanagawa N, 2016, Spread through air spaces is a predictive factor of recurrence and a prognostic factor in stage I lung adenocarcinoma. Interact Cardiovasc Thorac Surg 23:567–572. , DOI 10.1093/icvts/ivw211 Uruga H, Fujii T, Fujimori S, Kohno T, Kishi K, 2017, Semiquantitative assessment of tumor spread through air spaces, STAS, in early-stage lung adenocarcinomas. J Thorac Oncol 12: 1046–1051. , DOI 10.1016/j.jtho.2017.03.019 Kadota K, Kushida Y, Katsuki N, Ishikawa R, Ibuki E, Motoyama M, Nii K, Yokomise H, Bandoh S, Haba R, 2017, Tumor spread through air spaces is an independent predictor of recurrence-free survival in patients with resected lung squamous cell carcinoma. Am J Surg Pathol 41:1077–1086. , DOI 10.1097/PAS. 0000000000000872 Lu S, Tan KS, Kadota K, Eguchi T, Bains S, Rekhtman N, Adusumilli PS, Travis WD, 2017, Spread through air spaces, STAS, is an independent predictor of recurrence and lung cancer-specific death in squamous cell carcinoma. J Thorac Oncol 12:223–234. , DOI 10.1016/j.jtho.2016.09.129 Morimoto J, Nakajima T, Suzuki H, Nagato K, Iwata T, Yoshida S, Fukuyo M, Ota S, Nakatani Y, Yoshino I, 2016, Impact of free tumor clusters on prognosis after resection of pulmonary adenocarcinoma. J Thorac Cardiovasc Surg 152:64–72.e1. , DOI 10.1016/j.jtcvs.2016.03.088 Shiono S, EndoM, Suzuki K, Yarimizu K, Hayasaka K, Yanagawa N, 2018, Spread through air spaces is a prognostic factor in sublobar resection of non-small cell lung Cancer. Ann Thorac Surg 106:354–360. , DOI 10.1016/j.athoracsur.2018.02. 076 Zombori T, Furak J, Nyari T, Cserni G, Tiszlavicz L, 2018, Evaluation of grading systems in stage I lung adenocarcinomas: a retrospective cohort study. J Clin Pathol 71:135–140. https://doi. org/10.1136/jclinpath-2016-204302 Dai C, Xie H, Su H, She Y, Zhu E, Fan Z, Zhou F, Ren Y, Xie D, Zheng H, Kadeer X, Chen D, Zhang L, Jiang G, Wu C, Chen C, 2017, Tumor spread through air spaces affects the recurrence and overall survival in patients with lung adenocarcinoma > 2 to 3 cm. J Thorac Oncol 12:1052–1060. , DOI 10.1016/j.jtho.2017. 03.020 Onozato ML, Kovach AE, Yeap BY,Morales-Oyarvide V, Klepeis VE, Tammireddy S, Heist RS, Mark EJ, Dias-Santagata D, Iafrate AJ, Yagi Y, Mino-Kenudson M, 2013, Tumor islands in resected early-stage lung adenocarcinomas are associated with unique clinicopathologic and molecular characteristics and worse prognosis. Am J Surg Pathol 37:287–294. , DOI 10.1097/PAS. 0b013e31826885fb Thunnissen E, Blaauwgeers HJ, de Cuba EM, Yick CY, Flieder DB, 2016, Ex vivo artifacts and histopathologic pitfalls in the lung. Arch Pathol Lab Med 140:212–220. , DOI 10.5858/arpa. 2015-0292-OA Blaauwgeers H, Flieder D,Warth A, Harms A,Monkhorst K, Witte B, Thunnissen E, 2017, A prospective study of loose tissue fragments in non-small cell lung cancer resection specimens: An alternative view to “spread through air spaces”. Am J Surg Pathol 41: 1226–1230. , DOI 10.1097/PAS.0000000000000889 KadotaK,Kushida Y, Kagawa S, Ishikawa R, Ibuki E, Inoue K, Go T, Yokomise H, Ishii T, Kadowaki N, Haba R, 2019, Limited resection is associated with a higher risk of locoregional recurrence than lobectomy in stage I lung adenocarcinoma with tumor spread through air spaces. Am J Surg Pathol 43:1033–1041. https://doi. org/10.1097/PAS.0000000000001285 Eguchi T, KamedaK, Lu S, BottMJ, Tan KS, Montecalvo J, Chang JC, Rekhtman N, Jones DR, Travis WD, Adusumilli PS, 2019, Lobectomy is associated with better outcomes than sublobar resection in spread through air spaces, STAS)-positive T1 lung adenocarcinoma: A Propensity score-matched analysis. J Thorac Oncol 14:87–98. , DOI 10.1016/j.jtho.2018.09.005 Warth A, Muley T, Kossakowski CA, Goeppert B, Schirmacher P, Dienemann H, Weichert W, 2015, Prognostic impact of intraalveolar tumor spread in pulmonary adenocarcinoma. Am J Surg Pat h o l 39:793–801 . ht t ps : / / doi . o r g /10. 1097/PAS . 0000000000000409 Lee JS, Kim EK, Kim M, Shim HS, 2018, Genetic and clinicopathologic characteristics of lung adenocarcinoma with tumor spread through air spaces. Lung Cancer 123:121–126. , DOI 10. 1016/j.lungcan.2018.07.020 Kadota K, Yeh YC, Sima CS, Rusch VW, Moreira AL, Adusumilli PS, Travis WD, 2014, The cribriform pattern identifies a subset of acinar predominant tumors with poor prognosis in patients with stage I lung adenocarcinoma: a conceptual proposal to classify cribriform predominant tumors as a distinct histologic subtype. Mod Pathol 27:690–700. , DOI 10.1038/modpathol.2013.188 KadotaK,Kushida Y, Kagawa S, Ishikawa R, Ibuki E, Inoue K, Go T, Yokomise H, Ishii T, Kadowaki N, Haba R, 2019, Cribriform subtype is an independent predictor of recurrence and survival after adjustment for the eighth edition of TNMstaging system in patients with resected lung adenocarcinoma. J Thorac Oncol 14:245–254. , DOI 10.1016/j.jtho.2018.09.028 Yanagawa N, Shiono S, Abiko M, Katahira M, Osakabe M, Ogata SY, 2016, The clinical impact of solid and micropapillary patterns in resected lung adenocarcinoma. J Thorac Oncol 11:1976–1983. , DOI 10.1016/j.jtho.2016.06.014 Cha MJ, Lee HY, Lee KS, Jeong JY, Han J, Shim YM, Hwang HS, 2014, Micropapillary and solid subtypes of invasive lung adenocarcinoma: clinical predictors of histopathology and outcome. J Thorac Cardiovasc Surg 147:921–928.e2. , DOI 10.1016/ j.jtcvs.2013.09.045 Hung JJ, Yeh YC, JengWJ,Wu KJ, Huang BS,Wu YC, Chou TY, HsuWH, 2014, Predictive value of the international association for the study of lung cancer/American Thoracic Society/European Respiratory Society classification of lung adenocarcinoma in tumor recurrence and patient survival. J Clin Oncol 32:2357–2364. https:// doi.org/10.1200/JCO.2013.50.1049 Zombori T, Nyari T, Tiszlavicz L, Palfoldi R, Csada E, Geczi T, Ottlakan A, Pecsy B, Cserni G, Furak J, 2018, The more the micropapillary pattern in stage I lung adenocarcinoma, the worse the prognosis-a retrospective study on digitalized slides. Virchows Arch 472:949–958. , DOI 10.1007/s00428-018-2337-x Medina MA, Onken AM, de Margerie-Mellon C, Heidinger BH, Chen Y, Bankier AA, VanderLaan PA, 2020, Preoperative bronchial cytology for the assessment of tumor spread through air spaces in lung adenocarcinoma resection specimens. Cancer Cytopathol 128:278–286. , DOI 10.1002/cncy.22243 Yeh YC, Nitadori J, Kadota K, Yoshizawa A, Rekhtman N, Moreira AL, Sima CS, Rusch VW, Adusumilli PS, Travis WD, 2015, Using frozen section to identify histological patterns in stage I lung adenocarcinoma of ≤ 3 cm: accuracy and interobserver agreement. Histopathology 66:922–938. , DOI 10.1111/ his.12468 Morimoto J, Nakajima T, Suzuki H, Nagato K, Iwata T, Yoshida S, Fukuyo M, Ota S, Nakatani Y, Yoshino I, 2016, Impact of free tumor clusters on prognosis after resection of pulmonary adenocarcinoma. J Thorac Cardiovasc Surg 152:64–72.e1. , DOI 10.1016/j.jtcvs.2016.03.088 Postmus PE, Kerr KM, Oudkerk M, Senan S, Waller DA, Vansteenkiste J, Escriu C, Peters S, ESMO Guidelines Committee, 2017, Early and locally advanced non-small-cell lung cancer, NSCLC): ESMOClinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 28:iv1–iv21. , DOI 10.1093/annonc/mdx222 http://ftsz.pte.hu/docs/protokollok/TUDOHorgorak.pdf. Accessed 30 Apr 2020 Tsubokawa N, Mimae T,MiyataY, Sasada S,Yoshiya T, Kushitani K, Takeshima Y, Murakami S, Yokose T, Ito H, Nakayama H, Okada M, 2016, Prognostic significance of vascular invasion in intermediate-grade subtype of lung adenocarcinoma. Jpn J Clin Oncol 46:1015–1021. , DOI 10.1093/jjco/hyw113 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2451 EP 2458 DI 10.1007/s12253-020-00855-7 PG 8 ER PT J AU Muraro, E Vaccher, E Furlan, C Fratta, E Fanetti, G Fae’, AD Martorelli, D Cangemi, M Polesel, J Navarria, F Gobitti, C Comaro, E Scaini, Ch Pratesi, Ch Zanussi, S Lupato, V Grando, G Giacomarra, V Sulfaro, S Barzan, L Dolcetti, R Steffan, A Canzonieri, V Franchin, G AF Muraro, Elena Vaccher, Emanuela Furlan, Carlo Fratta, Elisabetta Fanetti, Giuseppe Fae’, Antonia Damiana Martorelli, Debora Cangemi, Michela Polesel, Jerry Navarria, Federico Gobitti, Carlo Comaro, Elisa Scaini, Chiara Pratesi, Chiara Zanussi, Stefania Lupato, Valentina Grando, Giuseppe Giacomarra, Vittorio Sulfaro, Sandro Barzan, Luigi Dolcetti, Riccardo Steffan, Agostino Canzonieri, Vincenzo Franchin, Giovanni TI Predictive Value of CD8 Expression and FoxP3 Methylation in Nasopharyngeal Carcinoma Patients Treated with Chemoradiotherapy in a Non-endemic Area SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Nasopharyngeal carcinoma; Immunosuppression; EBV-specific immunity; Chemoradiotherapy; CD8; FoxP3 ID Nasopharyngeal carcinoma; Immunosuppression; EBV-specific immunity; Chemoradiotherapy; CD8; FoxP3 AB Undifferentiated Nasopharyngeal Carcinoma (UNPC) is associated with Epstein-Barr Virus (EBV) and characterized by an abundant immune infiltrate potentially influencing the prognosis. Thus, we retrospectively assessed the significance of immunosuppression in the UNPC microenvironment as prognostic biomarker of treatment failure in a non-endemic area, and monitored the variation of systemic EBV-specific immunity before and after chemoradiotherapy (CRT). DNA and RNA were extracted from diagnostic biopsies obtained by tumor and adjacent mucosa from 63 consecutive EBV+ UNPC patients who underwent radical CRT. Among these patients 11 relapsed within 2 years. The expression of the EBV-derived UNPC-specific BARF1 gene and several immune-related genes was monitored through quantitative RT-PCR and methylation-specific PCR analyses. Peripheral T cell responses against EBV and BARF1 were measured in 14 patients (7 relapses) through IFN-γ ELISPOT assay. We found significantly higher expression levels of BARF1, CD8, IFN-γ, IDO, PD-L1, and PD-1 in UNPC samples compared to healthy tissues. CD8 expression was significantly reduced in both tumor and healthy tissues in UNPC patients who relapsed within two years. We observed a hypomethylated FOXP3 intron 1 exclusively in relapsed UNPC patients. Finally, we noticed a significant decrease in EBV- and BARF1-specific T-cells after CRT only in relapsing patients. Our data suggest that a high level of immunosuppression (low CD8, hypomethylated FoxP3) in UNPC microenvironment may predict treatment failure and may allow an early identification of patients who could benefit from the addition of immune modulating strategies to improve first line CRT. C1 [Muraro, Elena] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers UnitAviano, PN, Italy. [Vaccher, Emanuela] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department ofMedicalOncologyAviano, PN, Italy. [Furlan, Carlo] General Hospital “San Martino”, Department of Radiation OncologyBelluno, Italy. [Fratta, Elisabetta] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers UnitAviano, PN, Italy. [Fanetti, Giuseppe] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Radiation OncologyAviano, PN, Italy. [Fae’, Antonia Damiana] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers UnitAviano, PN, Italy. [Martorelli, Debora] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers UnitAviano, PN, Italy. [Cangemi, Michela] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers UnitAviano, PN, Italy. [Polesel, Jerry] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Unit of Cancer EpidemiologyAviano, PN, Italy. [Navarria, Federico] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Radiation OncologyAviano, PN, Italy. [Gobitti, Carlo] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Radiation OncologyAviano, PN, Italy. [Comaro, Elisa] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers UnitAviano, PN, Italy. [Scaini, Chiara] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers UnitAviano, PN, Italy. [Pratesi, Chiara] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers UnitAviano, PN, Italy. [Zanussi, Stefania] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers UnitAviano, PN, Italy. [Lupato, Valentina] General Hospital “S. Maria degli Angeli”, Unit of OtolaryngologyPordenone, Italy. [Grando, Giuseppe] General Hospital “S. Maria degli Angeli”, Unit of OtolaryngologyPordenone, Italy. [Giacomarra, Vittorio] General Hospital “S. Maria degli Angeli”, Unit of OtolaryngologyPordenone, Italy. [Sulfaro, Sandro] General Hospital “S. Maria degli Angeli”, Division of PathologyPordenone, Italy. [Barzan, Luigi] Centro di Riferimento Oncologico di Aviano (CRO) IRCCSAviano, PN, Italy. [Dolcetti, Riccardo] University of Queensland Diamantina Institute, Translational Research InstituteBrisbane, Australia. [Steffan, Agostino] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers UnitAviano, PN, Italy. [Canzonieri, Vincenzo] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Pathology UnitAviano, PN, Italy. [Franchin, Giovanni] Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Radiation OncologyAviano, PN, Italy. RP Muraro, E (reprint author), Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Department of Translational Research, Immunopathology and Cancer Biomarkers Unit, Aviano, Italy. EM elemuraro@gmail.com CR Yu MC, Yuan JM, 2002, Epidemiology of nasopharyngeal carcinoma. Semin Cancer Biol 12:421–429 Chan JKC, Bray F, McCarron P, Foo W, Lee AWM, Yip T et al, 2005, Nasopharyngeal carcinoma. In: Barnes L, Eveson JW, Reichart P et al, eds, WHO classification of tumours. Pathology and genetics. Head and neck tumours. IARC Press, Lyon, pp 85–97 Chua MLK,Wee JTS, Hui EP, Chan ATC, 2016, Nasopharyngeal carcinoma. Lancet 387:1012–1024 Sun X, Su S, Chen C, Han F, Zhao C, Xiao W, Deng X, Huang S, Lin C, Lu T, 2014, Long-term outcomes of intensity-modulated radiotherapy for 868 patients with nasopharyngeal carcinoma: an analysis of survival and treatment toxicities. Radiother Oncol 110: 398–403 Lo KW, To KF, Huang DP, 2004, Focus on nasopharyngeal carcinoma. Cancer Cell 5:423–428 Merlo A, Turrini R, Dolcetti R, Martorelli D, Muraro E, Comoli P, Rosato A, 2010, The interplay between Epstein-Barr virus and the immune system: a rationale for adoptive cell therapy of EBVrelated disorders. Haematologica 95:1769–1777 Decaussin G, Sbih-Lammali F, de Turenne-Tessier M, Bouguermouh A, Ooka T, 2000, Expression of BARF1 gene encoded by Epstein-Barr virus in nasopharyngeal carcinoma biopsies. Cancer Res 60:5584–5588 Stevens SJ, Verkuijlen SA, Hariwiyanto B, Harijadi PDK, Fachiroh J et al, 2006, Noninvasive diagnosis of nasopharyngeal carcinoma: nasopharyngeal brushings reveal high Epstein-Barr virusDNAload and carcinoma-specific viral BARF1 mRNA. Int J Cancer 119: 608–614 Ramayanti O, Juwana H, Verkuijlen SA, Adham M, Pegtel MD, Greijer AE et al, 2017, Epstein-Barr virus mRNA profiles and viral DNA methylation status in nasopharyngeal brushings from nasopharyngeal carcinoma patients reflect tumor origin. Int J Cancer 140:149–162 Martorelli D, Houali K, Caggiari L, Vaccher E, Barzan L, Franchin G, Gloghini A, Pavan A, da Ponte A, Tedeschi RM, de Re V, Carbone A, Ooka T, de Paoli P, Dolcetti R, 2008, Spontaneous T cell responses to Epstein-Barr virus-encoded BARF1 protein and derived peptides in patients with nasopharyngeal carcinoma: bases for improved immunotherapy. Int J Cancer 123:1100–1107 Fae DA,Martorelli D,MastorciK,Muraro E, Dal Col J, Franchin G et al, 2016, Broadening specificity and enhancing cytotoxicity of adoptive T cells for nasopharyngeal carcinoma immunotherapy. Cancer Immunol Res 4(5):431–440 Chen QY, Guo SY, Tang LQ, Lu TY, Chen BL, Zhong QY, Zou MS, Tang QN, Chen WH, Guo SS, Liu LT, Li Y, Guo L, Mo HY, Sun R, Luo DH, Zhao C, Cao KJ, Qian CN, Guo X, ZengMS, Mai HQ, 2018, Combination of tumor volume and Epstein-Barr virus DNA improved prognostic stratification of stage II nasopharyngeal carcinoma in the IMRT era: a large-scale cohort study. Cancer Res Treat Jul 50(3):861–871 BortolinMT, Pratesi C, Dolcetti R, Bidoli E, Vaccher E, Zanussi S, Tedeschi R, de Paoli P, 2006, Clinical value of Epstein-Barr virus DNA levels in peripheral blood samples of Italian patients with undifferentiated carcinoma of nasopharyngeal type. Cancer Lett 233:247–254 Shanmugaratnam K, Chan SH, de-The G, Goh JE, Khor TH, Simons MJ et al, 1979, Histopathology of nasopharyngeal carcinoma: correlations with epidemiology, survival rates and other biological characteristics. Cancer 44:1029–1044 Zhang YL, Li J, Mo HY, Qiu F, Zheng LM, Qian CN, Zeng YX, 2010, Different subsets of tumor infiltrating lymphocytes correlate with NPC progression in different ways. Mol Cancer 9:4 Lee JJ, Kao KC, Chiu YL, Jung CJ, Liu CJ, Cheng SJ, Chang YL, Ko JY, Chia JS, 2017, Enrichment of human CCR6(+, regulatory T cells with superior suppressive activity in oral cancer. J Immunol 199:467–476 Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D, Parmentier N et al, 2003, Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med 9:1269–1274 Ben-Haj-Ayed A, Moussa A, Ghedira R, Gabbouj S, Miled S, Bouzid N, Tebra-Mrad S, Bouaouina N, Chouchane L, Zakhama A, Hassen E, 2016, Prognostic value of indoleamine 2,3- dioxygenase activity and expression in nasopharyngeal carcinoma. Immunol Lett 169:23–32 Pardoll DM, 2012, The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12:252–264 Zhang J, Fang W, Qin T, Yang Y, Hong S, Liang W, Ma Y, Zhao H, Huang Y, Xue C, Huang P, Hu Z, Zhao Y, Zhang L, 2015, Coexpression of PD-1 and PD-L1 predicts poor outcome in nasopharyngeal carcinoma. Med Oncol 32:86 Zhou Y, Shi D, Miao J, Wu H, Chen J, Zhou X, Hu D, Zhao C, Deng W, Xie C, 2017, PD-L1 predicts poor prognosis for nasopharyngeal carcinoma irrespective of PD-1 and EBV-DNA load. Sci Rep 7:43627 Lee VH, Lo AW, Leung CY, ShekWH, Kwong DL, Lam KO et al, 2016, Correlation of PD-L1 expression of tumor cells with survival outcomes after radical intensity-modulated radiation therapy for non-metastatic nasopharyngeal carcinoma. PLoS One 11:e0157969 ChanOS,Kowanetz M, NgWT,Koeppen H, Chan LK,YeungRM et al, 2017, Characterization of PD-L1 expression and immune cell infiltration in nasopharyngeal cancer. Oral Oncol 67:52–60 Hsu MC, Hsiao JR, Chang KC, Wu YH, Su IJ, Jin YT, Chang Y, 2010, Increase of programmed death-1-expressing intratumoral CD8 T cells predicts a poor prognosis for nasopharyngeal carcinoma. Mod Pathol 23:1393–1403 Zhou Y, Miao J,Wu H, Tang H, Kuang J, Zhou X, Peng Y, Hu D, Shi D, Deng W, Cao X, Zhao C, Xie C, 2017, PD-1 and PD-L1 expression in 132 recurrent nasopharyngeal carcinoma: the correlation with anemia and outcomes. Oncotarget 8:51210–51223 Topalian SL, Taube JM, Anders RA, Pardoll DM, 2016, Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 16:275–287 Pilones KA, Vanpouille-Box C, Demaria S, 2015, Combination of radiotherapy and immune checkpoint inhibitors. Semin Radiat Oncol 25:28–33 Gameiro SR, JammehML,WattenbergMM, Tsang KY, Ferrone S, Hodge JW, 2014, Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget 5:403–416 Franchin G, Vaccher E, Talamini R, Gobitti C,Minatel E, Politi D, Sartor G, Trovo MG, Barzan L, 2002, Nasopharyngeal cancer WHOtype II-III:monoinstitutional retrospective analysis with standard and accelerated hyperfractionated radiation therapy. Oral Oncol 38:137–144 Franchin G, Vaccher E, Talamini R, Politi D, Gobitti C, Minatel E, LLeshi A, Sartor G,MascarinM, Rumeileh IA, TrovoMG, Barzan L, 2011, Intensity-modulated radiotherapy, IMRT)/Tomotherapy following neoadjuvant chemotherapy in stage IIB-IVA/B undifferentiated nasopharyngeal carcinomas, UCNT): a mono-institutional experience. Oral Oncol 47:905–909 Muls N, Dang HA, Sindic CJ, van Pesch V, 2014, Fingolimod increases CD39-expressing regulatory T cells in multiple sclerosis patients. PLoS One 9:e113025 Ooft ML, van Ipenburg JA, BrauniusWW, Zuur CI, Koljenovic S, Willems SM, 2017, Prognostic role of tumor infiltrating lymphocytes in EBV positive and EBV negative nasopharyngeal carcinoma. Oral Oncol 71:16–25 Zhang L, MacIsaac KD, Zhou T, Huang PY, Xin C, Dobson JR et al, 2017, Genomic analysis of nasopharyngeal carcinoma reveals TME-based subtypes. Mol Cancer Res 15:1722–1732 Lau KM, Cheng SH, Lo KW, Lee SA, Woo JK, van Hasselt CA et al, 2007, Increase in circulating Foxp3+CD4+CD25(high, regulatory T cells in nasopharyngeal carcinoma patients. Br J Cancer 96:617–622 Furlan C, Polesel J, Barzan L, Franchin G, Sulfaro S, Romeo S, Colizzi F, Rizzo A, Baggio V, Giacomarra V, Dei Tos AP, Boscolo-Rizzo P, Vaccher E, Dolcetti R, Sigalotti L, Fratta E, 2017, Prognostic significance of LINE-1 hypomethylation in oropharyngeal squamous cell carcinoma. Clin Epigenetics 9:58 ZhaoW,Mo Y,Wang S,Midorikawa K,Ma N, Hiraku Y, Oikawa S, Huang G, Zhang Z, Murata M, Takeuchi K, 2017, Quantitation of DNA methylation in Epstein-Barr virus-associated nasopharyngeal carcinoma by bisulfite amplicon sequencing. BMC Cancer 17: 489 Seto E, Yang L, Middeldorp J, Sheen TS, Chen JY, Fukayama M, Eizuru Y, Ooka T, Takada K, 2005, Epstein-Barr virus, EBV)- encoded BARF1 gene is expressed in nasopharyngeal carcinoma and EBV-associated gastric carcinoma tissues in the absence of lytic gene expression. J Med Virol 76:82–88 Cosmopoulos K, Pegtel M, Hawkins J, Moffett H, Novina C, Middeldorp J, Thorley-Lawson DA, 2009, Comprehensive profiling of Epstein-Barr virus microRNAs in nasopharyngeal carcinoma. J Virol 83:2357–2367 Takada K, 2012, Role of EBER and BARF1 in nasopharyngeal carcinoma, NPC, tumorigenesis. Semin Cancer Biol 22:162–165 Liu P, Xie BL, Cai SH, He YW, Zhang G, Yi YM, du J, 2009, Expression of indoleamine 2,3-dioxygenase in nasopharyngeal carcinoma impairs the cytolytic function of peripheral blood lymphocytes. BMC Cancer 9:416 Lee SJ, Jang BC, Lee SW, Yang YI, Suh SI, Park YM, Oh S, Shin JG, Yao S, Chen L, Choi IH, 2006, Interferon regulatory factor-1 is prerequisite to the constitutive expression and IFN-gamma-induced upregulation of B7-H1, CD274). FEBS Lett 580:755–762 Hsu C, Lee SH, Ejadi S, Even C, Cohen RB, Le Tourneau C et al, 2017, Safety and antitumor activity of Pembrolizumab in patients with programmed death-ligand 1-positive nasopharyngeal carcinoma: results of the KEYNOTE-028 study. J Clin Oncol 35(36): 4050–4056 FangW, Zhang J, Hong S, Zhan J, Chen N, Qin T, Tang Y, Zhang Y, Kang S, Zhou T, Wu X, LiangW, Hu Z,Ma Y, Zhao Y, Tian Y, YangY, Xue C, Yan Y, Hou X,Huang P,HuangY, Zhao H, Zhang L, 2014, EBV-driven LMP1 and IFN-gamma up-regulate PD-L1 in nasopharyngeal carcinoma: implications for oncotargeted therapy. Oncotarget 5:12189–12202 Zhu Q, Cai MY, Chen CL, Hu H, Lin HX, Li M, Weng DS, Zhao JJ, Guo L, Xia JC, 2017, Tumor cells PD-L1 expression as a favorable prognosis factor in nasopharyngeal carcinoma patients with pre-existing intratumor-infiltrating lymphocytes.Oncoimmunology 6:e1312240 Ma BBY, Lim WT, Goh BC, Hui EP, Lo KW, Pettinger A et al, 2018, Antitumor activity of Nivolumab in recurrent and metastatic nasopharyngeal carcinoma: an international, multicenter study of the Mayo Clinic phase 2 consortium, NCI-9742). J Clin Oncol 36(14):1412–1418 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2459 EP 2467 DI 10.1007/s12253-020-00859-3 PG 9 ER PT J AU Rossat, S Perrier, H Lefevre, M Louvet, Ch Berre, LN Chamois, J Dorel, M Vacque, D Guillaudeau, A Genet, D Maillet, E Triby, S Sabourin, JCh AF Rossat, Stephane Perrier, Herve Lefevre, Marine Louvet, Christophe Berre, Le Nathalie Chamois, Jerome Dorel, Maryline Vacque, Daniel Guillaudeau, Angelique Genet, Dominique Maillet, Evelyne Triby, Simon Sabourin, Jean-Christophe TI Drastic Reduction of Turnaround Time After Implementation of a Fully Automated Assay for RAS-BRAF Mutations in Colorectal Cancer: A Pilot Prospective Study in Real-life Conditions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Metastatic colorectal cancer; Mutation analysis; KRAS mutation; BRAF mutation ID Metastatic colorectal cancer; Mutation analysis; KRAS mutation; BRAF mutation AB In some situations, there is a need for rapid mutation tests for guiding clinical decisions and starting targeted therapies with minimal delays. In this study we evaluated the turnaround time before and after the implementation of a fully automated multiplex assay for KRAS and NRAS/BRAF mutation tests (Idylla™ platform, Biocartis) in metastatic colorectal cancer. The objective of this project was to compare the turnaround times in 2017–2018 with the fully automated multiplex assay to the 2016 results with previous methods. Centers with a number of tests for metastatic colorectal cancer > 100 yearly and a usual turnaround time ≥ 3 weeks for mutation detection were selected. Results of 505 KRAS tests and 369 NRAS/BRAF tests were transmitted by 10 centers. The mean turnaround time from test prescription to reception of results was reduced from 25.8 days in 2016 to 4.5 days in 2017–2018. In conclusion, this pilot project shows that the Idylla™platform for testing KRAS and NRAS/BRAF mutations allows an optimized turnaround time from test prescription to reception of results. C1 [Rossat, Stephane] Medipath, Aix-MarseilleEguilles, France. [Perrier, Herve] Saint-Joseph HospitalMarseille, France. [Lefevre, Marine] Institute MontsourisParis, France. [Louvet, Christophe] Institute MontsourisParis, France. [Berre, Le Nathalie] Ouest PathologieRennes, France. [Chamois, Jerome] Private Hospital St GregoireSaint-Gregoire, France. [Dorel, Maryline] Alizes Pathology CenterBaie-Mahault, Guadeloupe, France. [Vacque, Daniel] Les Eaux Claires ClinicBaie-Mahault, Guadeloupe, France. [Guillaudeau, Angelique] Limoges PolyclinicLimoges, France. [Genet, Dominique] Chenieux ClinicLimoges, France. [Maillet, Evelyne] Pathologie Center MAILLET EvelyneSte Clotilde, La Reunion, France. [Triby, Simon] AMGEN FranceBoulogne-Billancourt, France. [Sabourin, Jean-Christophe] Rouen University HospitalRouen, France. RP Sabourin, JCh (reprint author), Rouen University Hospital, Rouen, France. EM sabourinjc@gmail.com CR Kuipers EJ, Grady WM, Lieberman D, Seufferlein T, Sung JJ, Boelens PG, van de Velde CJ, Watanabe T, 2015, Colorectal cancer. Nat Rev Dis Primers 1:15065 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D, Bray F, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403 Hocking CM, Price TJ, 2014, Panitumumab in the management of patients with KRAS wild-type metastatic colorectal cancer. Therap Adv Gastroenterol 7:20–37 Prenen H, Tejpar S, Van Cutsem E, 2010, New strategies for treatment of KRAS mutant metastatic colorectal cancer. Clin Cancer Res 16:2921–2926 Siena S, Sartore-Bianchi A, Di Nicolantonio F, Balfour J, Bardelli A, 2009, Biomarkers predicting clinical outcome of epidermal growth factor receptor-targeted therapy inmetastatic colorectal cancer. J Natl Cancer Inst 101:1308–1324 Er TK, Chen CC, Bujanda L, Herreros-Villanueva M, 2014, Current approaches for predicting a lack of response to anti- EGFR therapy in KRAS wild-type patients. Biomed Res Int 2014:591867 Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, Juan T, Sikorski R, Suggs S, Radinsky R, Patterson SD, Chang DD, 2008, Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 26:1626– 1634 Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ, Tu D, Tebbutt NC, Simes RJ, Chalchal H, Shapiro JD, Robitaille S, Price TJ, Shepherd L, Au HJ, Langer C, Moore MJ, Zalcberg JR, 2008, K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 359:1757–1765 Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, Rivera F, Kocakova I, Ruff P, Blasinska-Morawiec M, Smakal M, Canon JL, Rother M, Williams R, Rong A, Wiezorek J, Sidhu R, Patterson SD, 2013, Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med 369:1023–1034 Hecht JR, Douillard JY, Schwartzberg L, Grothey A, Kopetz S, Rong A, Oliner KS, Sidhu R, 2015, Extended RAS analysis for anti-epidermal growth factor therapy in patients with metastatic colorectal cancer. Cancer Treat Rev 41:653–659 Sorich MJ, Wiese MD, Rowland A, Kichenadasse G, McKinnon RA, Karapetis CS, 2015, Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: ameta-analysis of randomized, controlled trials. Ann Oncol 26: 13–21 Zenonos K, Kyprianou K, 2013, RAS signaling pathways, mutations and their role in colorectal cancer.World J Gastrointest Oncol 5:97–101 Malumbres M, Barbacid M, 2003, RAS oncogenes: the first 30 years. Nat Rev Cancer 3:459–465 Fearon ER, 2011, Molecular genetics of colorectal cancer. Annu Rev Pathol 6:479–507 Rajagopalan H, Bardelli A, Lengauer C, Kinzler KW, Vogelstein B, Velculescu VE, 2002, Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature 418:934 Oikonomou E, Koustas E, GoulielmakiM, Pintzas A, 2014, BRAF vs RAS oncogenes: are mutations of the same pathway equal? Differential signalling and therapeutic implications. Oncotarget 5: 11752–11777 Van Cutsem E, Cervantes A, Nordlinger B, Arnold D, Group EGW, 2014, Metastatic colorectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 25(Suppl 3):iii1–i9 Ciardiello F, Tejpar S, Normanno N, Mercadante D, Teague T, Wohlschlegel B, Van Cutsem E, 2011, Uptake of KRAS mutation testing in patients with metastatic colorectal cancer in Europe, Latin America and Asia. Target Oncol 6:133–145 Institut National du Cancer. Acces aux tests moleculaires EGFR, RAS et BRAF. Resultats d’une enquete dans 5 regions francaises. Janvier 2016 de Biase D, de Luca C, Gragnano G, Visani M, Bellevicine C, Malapelle U, Tallini G, Troncone G, 2016, Fully automated PCR detection of KRAS mutations on pancreatic endoscopic ultrasound fine-needle aspirates. J Clin Pathol Solassol J, Vendrell J, Markl B, Haas C, Bellosillo B, Montagut C, Smith M, O’Sullivan B, D’Haene N, Le Mercier M, Grauslund M, Melchior LC, Burt E, Cotter F, Stieber D, Schmitt FL, Motta V, Lauricella C, Colling R, Soilleux E, FassanM,Mescoli C, Collin C, Pages JC, Sillekens P, 2016, Multi-Center Evaluation of the Fully Automated PCR-Based Idylla KRAS Mutation Assay for Rapid KRAS Mutation Status Determination on Formalin-Fixed Paraffin-Embedded Tissue of Human Colorectal Cancer. PLoS One 11:e0163444 Johnston L, Power M, Sloan P, Long A, Silmon A, Chaffey B, Lisgo AJ, Little L, Vercauteren E, Steiniche T, Meyer T, Simpson J, 2018, Clinical performance evaluation of the Idylla NRASBRAF mutation test on retrospectively collected formalin-fixed paraffin-embedded colorectal cancer tissue. J Clin Pathol 71:336– 343 Scott RJ, Fox SB, Desai J, Grieu F, Amanuel B, Garrett K, Harraway J, Cheetham G, Pattle N, Haddad A, Byron K, Rudzki B, Waring P, Iacopetta B, 2014, KRAS mutation testing of metastatic colorectal cancer in Australia: where are we at? Asia Pac J Clin Oncol 10:261–265 INCa, Bonnes pratiques pour la recherche a visee theranostique demutations somatiques dans les tumeurs solides. Aout 2010, https://www.e-cancer.fr/content/download/63175/568709/file/ OUTTHERANOS10.pdf INCa. Plan Cancer 2014–2019 https://www.e-cancer.fr/Expertiseset- publications/Catalogue-des-publications/Plan-Cancer-2014- 2019 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2469 EP 2473 DI 10.1007/s12253-020-00818-y PG 5 ER PT J AU Kuronya, Zs Szonyi, DM Nagyivanyi, K Gyergyay, F Geczi, L Budai, B Martin, T Ladanyi, A Kiss, E Biro, K AF Kuronya, Zsofia Szonyi, Daniel Mihaly Nagyivanyi, Krisztian Gyergyay, Fruzsina Geczi, Lajos Budai, Barna Martin, Tamas Ladanyi, Andrea Kiss, Edina Biro, Krisztina TI Predictive Markers of First Line Pazopanib Treatment in Kidney Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Kidney cancer; Pazopanib; Livermetastasis; Side effects; Efficacy ID Kidney cancer; Pazopanib; Livermetastasis; Side effects; Efficacy AB Real-world evidence from clinical practices is fundamental for understanding the efficacy and tolerability of medicinal products. Patients with renal cell cancer were studied to gain data not represented by analyses conducted on highly selected patients participating in clinical trials. Our goal was to retrospectively collect data from patients with advanced renal tumours treated with pazopanib (PZ) to investigate the efficacy, frequency of side effects, and searching for predictive markers. Eighty-one patients who had received PZ therapy as first-line treatment were retrospectively evaluated. Overall survival (OS), progression-free survival (PFS) were assessed as endpoints. Median PFS and OS were 11.8 months (95% CI: 8.8–22.4); and 30.2 months (95% CI: 20.3–41.7) respectively. Severe side effects were only encountered in 11 (14%) patients. The presence of liver metastasis shortened the median PFS (5.5 vs. 14.8 months, p = 0.003). Median PFS for patients with or without side effects was 25.6 vs. 7.3 months, respectively (p = 0.0001). Patients younger than 65 years had a median OS of 41.7 months vs. 25.2 months for those over 65 years of age (p = 0.008). According to our results absence of liver metastases, younger age (<65 years) and presence of side effects proved to be independent predictive markers of better PFS and OS. C1 [Kuronya, Zsofia] National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology Department, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Szonyi, Daniel Mihaly] Semmelweis UniversityBudapest, Hungary. [Nagyivanyi, Krisztian] National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology Department, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Gyergyay, Fruzsina] National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology Department, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Geczi, Lajos] National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology Department, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Budai, Barna] Orszagos Onkologiai Intezet, Molekularis Genetika OsztalyBudapest, Hungary. [Martin, Tamas] National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology Department, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Ladanyi, Andrea] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Kiss, Edina] Honved Korhaz, Onkologiai OsztalyBudapest, Hungary. [Biro, Krisztina] National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology Department, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. RP Kuronya, Zs (reprint author), National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology Department, 1122 Budapest, Hungary. CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68:394–424 National Comprehensive Cancer Network NCCN clinical practice guidelines in oncology, 2019 Mihaly Z, Sztupinszki Z, Surowiak P, Gyorffy B, 2012, A comprehensive overview of targeted therapy in metastatic renal cell carcinoma. Curr Cancer Drug Targets 12:857–872 Sloan B, Scheinfeld NS, 2008, Pazopanib, a VEGF receptor tyrosine kinase inhibitor for cancer therapy. Curr Opin Investig Drugs 9:1324–1335 Sternberg CN, Davis ID,Mardiak J, Szczylik C, Lee E,Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A, Zarba JJ, Chen M, McCann L, Pandite L, Roychowdhury DF, Hawkins RE, 2010, Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 28:1061–1068 Sternberg CN, Hawkins RE, Wagstaff J, Salman P, Mardiak J, Barrios CH, Zarba JJ, Gladkov OA, Lee E, Szczylik C, McCann L, Rubin SD, Chen M, Davis ID, 2013, A randomised, doubleblind phase III study of pazopanib in patients with advanced and/ or metastatic renal cell carcinoma: final overall survival results and safety update. Eur J Cancer 49:1287–1296 Administration FaD, 2009, NDA Approval Nieto M, Borregaard J, Ersboll J, ten Bosch GJA, van Zwieten- Boot B, Abadie E, Schellens JHM, Pignatti F, 2011, The European medicines agency review of pazopanib for the treatment of advanced renal cell carcinoma: summary of the scientific assessment of the Committee forMedicinal Products for human use. Clin Cancer Res 17:6608–6614 Motzer RJ, Hutson TE, Cella D, Reeves J, Hawkins R, Guo J, Nathan P, Staehler M, de Souza P, Merchan JR, Boleti E, Fife K, Jin J, Jones R, Uemura H, De Giorgi U, Harmenberg U, Wang J, Sternberg CN, Deen K, McCann L, Hackshaw MD, Crescenzo R, Pandite LN, Choueiri TK, 2013, Pazopanib versus sunitinib in metastatic renal-cell carcinoma. N Engl J Med 369:722–731 Motzer RJ, Hutson TE, McCann L, Deen K, Choueiri TK, 2014, Overall survival in renal-cell carcinoma with pazopanib versus sunitinib. N Engl J Med 370:1769–1770 Escudier BPC, Bono P, 2014, Randomized, controlled, doubleblind, cross-over trial assessing treatment preference for pazopanib versus sunitinib in patients with metastatic renal cell carcinoma: PISCES study. J Clin Oncol 32:1412–1418 Motzer RJ, Mazumdar M, Bacik J, BergW, Amsterdam A, Ferrara J, 1999, Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma. J Clin Oncol 17:2530–2540 Woldrich JM,Mallin K, Ritchey J, Carroll PR, Kane CJ, 2008, Sex differences in renal cell cancer presentation and survival: an analysis of the National Cancer Database, 1993–2004. J Urol 179:1709– 1713 Maraz A, Bodrogi I, Csejtei A, Dank M, Geczi L, Kuronya Z, Mangel L, Petranyi A, Szucs M, Bodoky G, 2013, First Hungarian experience with pazopanib therapy for patients with metastatic renal cancer. Magy Onkol 57:173–176 Schmidinger M, Bamias A, Procopio G, Hawkins R, Sanchez AR, Vazquez S, Srihari N, Kalofonos H, Bono P, Pisal CB, Hirschberg Y, Dezzani L, Ahmad Q, Jonasch E, PRINCIPAL Study Group, 2019, Prospective observational study of pazopanib in patients with advanced renal cell carcinoma, PRINCIPAL study). Oncologist 24:491–497 Nazha S, Tanguay S, Kapoor A, Jewett M, Kollmansberger C, Wood L, Bjarnason G, Heng D, Soulieres D, Reaume N, Basappa N, Levesque E, Dragomir A, 2018, Use of targeted therapy in patients with metastatic renal cell carcinoma: clinical and economic impact in a Canadian real-life setting. Curr Oncol 25:e576–e584 Kim MS, Chung HS, Hwang EC, Jung SI, Kwon DD, Hwang JE, BaeWK, Park JY, Jeong CW, Kwak C, Song C, Seo SI, Byun SS, Hong SH, Chung J, 2018, Efficacy of first-line targeted therapy in real-world Korean patients with metastatic renal cell carcinoma: focus on sunitinib and pazopanib. J Korean Med Sci 33:e325 Nagyivanyi K, Budai B, Biro K, Gyergyay F, Noszek L, Kuronya Z, Nemeth H, Nagy P, Geczi L, 2016, Synergistic survival: a new phenomenon connected to adverse events of first line sunitinib treatment in advanced renal cell carcinoma. Clin Genitourin Cancer 14:314–322 Maraz A, Cserhati A, Uhercsak G, Szilagyi E, Varga Z, Kahan Z, 2014, Therapeutic significance of sunitinib-induced "off-target" side effects. Magy Onkol 58:167–172 Di Fiore F, Rigal O, Menager C, Michel P, Pfister C, 2011, Severe clinical toxicities are correlated with survival in patients with advanced renal cell carcinoma treated with sunitinib and sorafenib. Br J Cancer 105:1811–1813 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2475 EP 2481 DI 10.1007/s12253-020-00853-9 PG 7 ER PT J AU Tas, F Erturk, K AF Tas, Faruk Erturk, Kayhan TI De Novo and Nevus-Associated Melanomas: Different Histopathologic Characteristics but Similar Survival Rates SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Melanoma; de novo; Nevus-associated; Survival ID Melanoma; de novo; Nevus-associated; Survival AB The clinical significances of de novo and nevus-associated melanomas are controversial. In this study, we investigated the correlations of these forms of melanomas in respect to their pathological and clinical features and patient outcomes. The data of 660 pathologically confirmed Turkish-Caucasian melanoma patients, whose tumors were either associated with a pre-existing melanocytic nevus or not, were analyzed retrospectively. They were treated and followed up at a single tertiary referral center. A total of 440 de novo (66.7%) and 220 nevus-associated melanomas (33.3%) were enrolled into the study. The median age of the patients was 51 years. The patients consisted of 341 men (51.7%) and 319 women (48.3%). There were significant correlations between de novo melanomas and advanced age (p = 0.003), tumor thickness greater than 2mm(p = 0.0001), ulceration (p = 0.01) and high mitotic rate (p = 0.03). On the other hand, nevus-associated melanomas were found significantly associated with histological regression (p = 0.03) and BRAFV600E mutation (p = 0.003). Most of the nevus-associated melanomas were found on trunk and head/neck, whereas extremities were more frequently inflicted by de novo melanomas (p = 0.0001). Furthermore, none of other variables, such as sex, histopathology, lymph node involvement and presence of metastasis, showed statistically significant difference between de novo and nevus-associated melanoma patients (p > 0.05). The 5-year DFS rates were 62.4% and 72.7% for de novo melanoma and for nevus-associated melanoma patients, respectively (p = 0.1). The 5-year OS rate were 72.1% and 76.4% for de novo melanoma and nevus-associated melanoma patients, respectively (p = 0.2). In conclusion, even though de novo melanomas are more significantly correlated with aggressive histopathologic variables, such as tumor depth, ulceration and high mitotic rate, the survival rates of de novo and nevus-associated melanomas are similar. C1 [Tas, Faruk] Istanbul Medical Faculty, Istanbul University, Oncology Institute, Capa, 34390 Istanbul, Turkey. [Erturk, Kayhan] Koc University, Department of Medical OncologyIstanbul, Turkey. RP Tas, F (reprint author), Istanbul Medical Faculty, Istanbul University, Oncology Institute, 34390 Istanbul, Turkey. EM faruktas2002@yahoo.com CR Siegel RL, Miller KD, Jemal A, 2020, Cancer statistics, 2020. CA Cancer J Clin 70:7–30 Cymerman RM, Shao Y, Wang K, Zhang Y, Murzaku EC, Penn LA et al, 2016, De novo vs nevus-associated melanomas: differences in associations with prognostic indicators and survival. J Natl Cancer Inst 108(10):djw121 Haenssle HA, Mograby N, Ngassa A, Buhl T, Emmert S, Schon MP et al, 2016, Association of patient risk factors and frequency of nevus-associated cutaneous melanomas. JAMA Dermatol 152: 291–298 Shitara D, Nascimento MM, Puig S, Yamada S, Enokihara M, Michalany N et al, 2014, Nevus-associated melanomas: clinicopathologic features. Am J Clin Pathol 142:485–491 Pampena R, Kyrgidis A, Lallas A, Moscarella E, Argenziano G, Longo C et al, 2017, A meta-analysis of nevus-associated melanoma: prevalence and practical implications. J AmAcad Dermatol 77: 938–945 Lin WM, Luo S, Muzikansky A, Lobo AZC, Tanabe KK, Sober AJ, Cosimi AB, Tsao H, Duncan LM, 2015, Outcome of patients with de novo versus nevus-associated melanoma. J Am Acad Dermatol 72:54–58 Purdue MP, From L, Armstrong BK, Kricker A, Gallagher RP, McLauhglin JR et al, 2005, Etiologic and other factors predicting nevus-associated cutaneous malignant melanoma. Cancer Epidemiol Biomark Prev 14:2015–2022 Weatherhead SC, Haniffa M, Lawrence CM, 2007, Melanomas arising from naevi and de novo melanomas—does origin matter? Br J Dermatol 156:72–76 Garcia-Cruz A, Florez A, de la Torre-Fraga C, Cruces PM, 2009, Observational cross-sectional study comparing Breslow thickness of melanoma arising from naevi and melanoma de novo. Br J Dermatol 161:700–702 Kaddu S, Smolle J, Zenahlik P, Hofmann-Wellenhof R, Kerl H, 2002, Melanoma with benign melanocytic naevus components: reappraisal of clinicopathological features and prognosis. Melanoma Res 12:271–278 Cochran AJ, 1969, Histology and prognosis in malignant melanoma. J Pathol 97:459–468 Rhodes AR, Sober AJ, Day CL, Melski JW, Harris TJ, Mihm MC Jr et al, 1982, The malignant potential of small congenital nevocellular nevi. An estimate of association based on a histologic study of 234 primary cutaneous melanomas. J Am Acad Dermatol 6:230–241 Friedman RJ, Rigel DS, Kopf AW, Lieblich L, Lew R, Harris MN, Roses DF, Gumport SL, Ragaz A, Waldo E, Levine J, Levenstein M, Koenig R, Bart RS, Trau H, 1983, Favorable prognosis for malignant melanomas associated with acquired melanocytic nevi. Arch Dermatol 119:455–462 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2483 EP 2487 DI 10.1007/s12253-020-00858-4 PG 5 ER PT J AU Marta, NG Velho, IP Bonadio, R Nardo, M Faraj, Sh Manoel Carlos, dAS Muniz, D Bastos, AD Dzik, C AF Marta, Nader Guilherme Velho, Isaacsson Pedro Bonadio, R. C. Renata Nardo, Mirella Faraj, F. Sheila Manoel Carlos, L. de Azevedo Souza Muniz, Q. B. David Bastos, Assed Diogo Dzik, Carlos TI Prognostic Value of Systemic Inflammatory Biomarkers in Patients with Metastatic Renal Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Renal cell carcinoma; Pazopanib; Sunitinib; Neutrophil-to-lymphocyte ratio; Platelet-to-lymphocyte ratio ID Renal cell carcinoma; Pazopanib; Sunitinib; Neutrophil-to-lymphocyte ratio; Platelet-to-lymphocyte ratio AB Metastatic renal cell carcinoma (mRCC) encompasses a heterogeneous group of neoplasms with distinct clinical behavior and prognoses. As a result of the increasing number of therapeutic options in the metastatic setting, it is crucial to improve prognostic stratification ability. We aimed to evaluate the prognostic value of neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and combination platelet count and neutrophil lymphocyte ratio (COP-NLR) in patients with mRCC. We evaluated a cohort of mRCC patients treated with first-line pazopanib or sunitinib. Levels of NLR, PLR and COP-NLR were measured prior to systemic treatment and evaluated as prognostic predictors. Primary endpoint was overall survival (OS). Data from 276 patients were included, of which 54.7%received first-line pazopanib and 45.3%, sunitinib. Memorial Sloan-Kettering Cancer Center risk classification was intermediate and poor in 50% and 42.6% of patients, respectively. High NLR (> 3.5) was associated with inferior OS (median 9.6 vs 17.8 months, P < 0.001). A high PLR (> 200) was associated with inferior OS (median 10.3 vs 17 months, P = 0.002). The median OS in the COP-NLR 1, 2 and 3 groups were 19.0 months (95% CI 15.3–26.0), 13.1 months (95% CI 9.8–17.0) and 7.4 months (95% CI 3.6–11.9), respectively (P < 0.001). In the multivariate analysis, high NLR and high COP-NLR were associated with inferior OS. Both high NLR and high COP-NLR were associated with poorer OS in our cohort of patients with mRCC treated with first-line pazopanib or sunitinib. C1 [Marta, Nader Guilherme] Instituto do Cancer do Estado de Sao Paulo, Av. Dr. Arnaldo, 251, 01246-000 Sao Paulo, SP, Brazil. [Velho, Isaacsson Pedro] Johns Hopkins University, Johns Hopkins Sidney Kimmel Cancer CenterBaltimore, MD, USA. [Bonadio, R. C. Renata] Instituto do Cancer do Estado de Sao Paulo, Av. Dr. Arnaldo, 251, 01246-000 Sao Paulo, SP, Brazil. [Nardo, Mirella] Instituto do Cancer do Estado de Sao Paulo, Av. Dr. Arnaldo, 251, 01246-000 Sao Paulo, SP, Brazil. [Faraj, F. Sheila] Instituto do Cancer do Estado de Sao Paulo, Av. Dr. Arnaldo, 251, 01246-000 Sao Paulo, SP, Brazil. [Manoel Carlos, L. de Azevedo Souza] Instituto do Cancer do Estado de Sao Paulo, Av. Dr. Arnaldo, 251, 01246-000 Sao Paulo, SP, Brazil. [Muniz, Q. B. David] Instituto do Cancer do Estado de Sao Paulo, Av. Dr. Arnaldo, 251, 01246-000 Sao Paulo, SP, Brazil. [Bastos, Assed Diogo] Instituto do Cancer do Estado de Sao Paulo, Av. Dr. Arnaldo, 251, 01246-000 Sao Paulo, SP, Brazil. [Dzik, Carlos] Instituto do Cancer do Estado de Sao Paulo, Av. Dr. Arnaldo, 251, 01246-000 Sao Paulo, SP, Brazil. RP Marta, NG (reprint author), Instituto do Cancer do Estado de Sao Paulo, 01246-000 Sao Paulo, Brazil. EM guilherme.marta@usp.br CR Siegel RL, Miller KD, Jemal A, 2018, Cancer statistics, 2018. CA Cancer J Clin 68:7–30. , DOI 10.3322/caac.21442 Howlader N, Noone AM, Krapcho M, Miller D, Bishop K, Altekruse SF, Kosary CL, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ CK, 2016, SEER Cancer statistics review, 1975–2013, National Cancer Institute. Bethesda, MD,. In: SEER web site. https://seer.cancer.gov/archive/csr/1975_2013/. Accessed 3 Apr 2018 Kidney and Renal Pelvis Cancer - Cancer Stat Facts. https://seer. cancer.gov/statfacts/html/kidrp.html. Accessed 1May 2019 Lalani A-KA, Xie W,Martini DJ et al, 2018, Change in neutrophilto- lymphocyte ratio, NLR, in response to immune checkpoint blockade for metastatic renal cell carcinoma. J Immunother cancer 6:5. , DOI 10.1186/s40425-018-0315-0 Motzer RJ, Mazumdar M, Bacik J et al, 1999, Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma. J Clin Oncol 17:2530–2530. , DOI 10.1200/JCO. 1999.17.8.2530 Heng DYC, Xie W, Regan MM et al, 2009, Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol 27:5794–5799. , DOI 10.1200/JCO.2008.21.4809 Amin MB, Edge SB, American Joint Committee on Cancer, 2017, AJCC cancer staging manual, 8th ed. Springer, Switzerland Delahunt B, Cheville JC, Martignoni G et al, 2013, The International Society of Urological Pathology, ISUP, grading system for renal cell carcinoma and other prognostic parameters. AmJ Surg Pathol 37:1490–1504. , DOI 10.1097/PAS. 0b013e318299f0fb Wei S, Al-Saleem T, 2017, The pathology and molecular genetics of Sarcomatoid renal cell carcinoma: a mini-review. J Kidney Cancer VHL 4:19–23. , DOI 10.15586/jkcvhl.2017.70 Network TCGAR, 2013, Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 499:43–49. https:// doi.org/10.1038/nature12222 Gerlinger M, Rowan AJ, Horswell S et al, 2012, Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366:883–892. , DOI 10.1056/ NEJMoa1113205 Mantovani A, Allavena P, Sica A, Balkwill F, 2008, Cancer-related inflammation. Nature 454:436–444. , DOI 10.1038/ nature07205 Watt DG, Proctor MJ, Park JH, et al, 2015, The neutrophil-platelet score, NPS, predicts survival in primary operable colorectal Cancer and a variety of common cancers. , DOI 10.1371/journal. pone.0142159 Tsujino T, Komura K,Matsunaga T et al, 2017, Preoperative measurement of the modified Glasgow prognostic score predicts patient survival in non-metastatic renal cell carcinoma prior to nephrectomy. Ann Surg Oncol 24:2787–2793. , DOI 10.1245/ s10434-017-5948-6 Boissier R, Campagna J, Branger N et al, 2017, The prognostic value of the neutrophil-lymphocyte ratio in renal oncology: a review. Urol Oncol Semin Orig Investig 35:135–141. , DOI 10.1016/J.UROLONC.2017.01.016 Semeniuk-Wojtas A, Lubas A, Stec R et al, 2018, Neutrophil-tolymphocyte ratio, platelet-to-lymphocyte ratio, and C-reactive protein as new and simple prognostic factors in patients withmetastatic renal cell Cancer treated with tyrosine kinase inhibitors: a systemic review and meta-analysis. Clin Genitourin Cancer. , DOI 10.1016/J.CLGC.2018.01.010 Tsujino T, Komura K, Ichihashi A et al, 2017, The combination of preoperative platelet count and neutrophil lymphocyte ratio as a prognostic indicator in localized renal cell carcinoma. Oncotarget 8:110311–110325. , DOI 10.18632/oncotarget.22688 Hu H, Yao X, Xie X et al, 2017, Prognostic value of preoperative NLR, dNLR, PLR and CRP in surgical renal cell carcinoma patients. World J Urol 35:261–270. , DOI 10.1007/s00345- 016-1864-9 Bilen MA, Dutcher GMA, Liu Y et al, 2018, Association between pretreatment neutrophil-to-lymphocyte ratio and outcome of patients with metastatic renal cell carcinoma treated with nivolumab. Clin Genitourin Cancer. , DOI 10.1016/J.CLGC.2017.12. 015 Tanaka N,Mizuno R, Yasumizu Y et al, 2017, Prognostic value of neutrophil-to-lymphocyte ratio in patients with metastatic renal cell carcinoma treated with first-line and subsequent second-line targeted therapy: a proposal of the modified-IMDC risk model. Urol Oncol Semin Orig Investig 35:39.e19–39.e28. https://doi. org/10.1016/J.UROLONC.2016.10.001 Chrom P, Stec R, Bodnar L, Szczylik C, 2018, Incorporating neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in place of neutrophil count and platelet count improves prognostic accuracy of the international metastatic renal cell carcinoma database consortium model. Cancer Res Treat 50:103–110. https://doi. org/10.4143/crt.2017.033 Koo AS, Armstrong C, Bochner B et al, 1992, Interleukin-6 and renal cell cancer: production, regulation, and growth effects. Cancer Immunol Immunother 35:97–105 Negrier S, Perol D, Menetrier-Caux C et al, 2004, Interleukin-6, interleukin-10, and vascular endothelial growth factor in metastatic renal cell carcinoma: prognostic value of interleukin-6 - from the Groupe Francais d’Immunotherapie. J Clin Oncol 22:2371–2378. , DOI 10.1200/JCO.2004.06.121 Menetrier-Caux C, Montmain G, Dieu MC et al, 1998, Inhibition of the differentiation of dendritic cells fromCD34(+, progenitors by tumor cells: role of interleukin-6 and macrophage colonystimulating factor. Blood 92:4778–4791 Motzer RJ, Tannir NM, McDermott DF et al, 2018, Nivolumab plus Ipilimumab versus Sunitinib in advanced renal-cell carcinoma. N Engl J Med 378:1277–1290. , DOI 10.1056/ NEJMoa1712126 Sreeramkumar V, Adrover JM, Ballesteros I, et al, 2014, Neutrophils scan for activated platelets to initiate inflammation. Science, 80- , 346:1234–1238. , DOI 10.1126/science. 1256478 Tsimafeyeu I, 2017, Management of non–clear cell renal cell carcinoma: current approaches. Urol Oncol Semin Orig Investig 35:5– 13. , DOI 10.1016/j.urolonc.2016.07.011 Buti S, BersanelliM,Maines F et al, 2017, First-line PAzopanib in NOn–clear-cell renal cArcinoMA: the Italian retrospective multicenter PANORAMA study. Clin Genitourin Cancer 15:e609–e614. , DOI 10.1016/j.clgc.2016.12.024 De Giorgi U, Procopio G, Giannarelli D et al, 2019, Association of Systemic Inflammation Index and Body Mass Index with survival in patients with renal cell Cancer treated with Nivolumab. Clin Cancer Res 25:3839–3846. , DOI 10.1158/1078-0432. CCR-18-3661 Bilen MA, Martini DJ, Liu Y et al, 2019, The prognostic and predictive impact of inflammatory biomarkers in patients who have advanced-stage cancer treated with immunotherapy. Cancer 125: 127–134. , DOI 10.1002/cncr.31778 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2489 EP 2497 DI 10.1007/s12253-020-00840-0 PG 9 ER PT J AU Wojtukiewicz, M Mysliwiec, M Sierko, E Sobierska, M Kruszewska, J Lipska, A Radziwon, P Tucker, S Honn, K AF Wojtukiewicz, Z. Marek Mysliwiec, Marta Sierko, Ewa Sobierska, Monika Kruszewska, Joanna Lipska, Alina Radziwon, Piotr Tucker, C. Stephanie Honn, V. Kenneth TI Elevated Microparticles, Thrombin-antithrombin and VEGF Levels in Colorectal Cancer Patients Undergoing Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Endothelium-derived microparticles; Blood coagulation; Angiogenesis; Colorectal cancer ID Endothelium-derived microparticles; Blood coagulation; Angiogenesis; Colorectal cancer AB Hypercoagulable state and neoangiogenesis are common phenomena associated with malignancy. Cancer patients have increased levels of circulating endothelium-derived microparticles (EMPs), which have been hypothesized to be involved in numerous pathophysiological processes. Hemostasis and angiogenesis are also activated in colorectal cancer (CRC) patients. The study aimed to investigate potential influence of chemotherapy on EMPs, thrombin anti-thrombin complex (TAT) and vascular endothelial growth factor (VEGF) levels in CRC patients undergoing chemotherapy. The study group consisted of 18 CRC patients: 8 stage III colon cancer (CC) and 10 stage IV rectal cancer (RC) patients. EMPs, TAT and VEGF levels were assessed before chemotherapy and after the third course. Results were compared with 10 healthy subjects. EMP concentration was measured by flow cytometry, while TAT and VEGF concentrations were assayed employing ELISA. Compared to the control group, CC and RC patients had significantly higher levels of tissue factor (TF)-bearing and non-TF-bearing EMPs before and after three courses of chemotherapy. VEGF concentrations in CRC patients were higher than in the control groups and increased following chemotherapy. TAT levels were elevated in CRC patients before chemotherapy compared to healthy subjects and significantly increased after the third course of chemotherapy. No significant correlation was found either betweenEMP and TAT levels, or between EMP concentrations and VEGF levels in the study group. CRC patients have increased EMPs, and TAT as well as VEGF levels tend to increase during chemotherapy. C1 [Wojtukiewicz, Z. Marek] Medical University of Bialystok, Department of Oncology, 12 Ogrodowa St, 15-027 Bialystok, Poland. [Mysliwiec, Marta] Medical University of Bialystok, Department of Oncology, 12 Ogrodowa St, 15-027 Bialystok, Poland. [Sierko, Ewa] Medical University of Bialystok, Department of Oncology, 12 Ogrodowa St, 15-027 Bialystok, Poland. [Sobierska, Monika] Medical University of Bialystok, Department of Oncology, 12 Ogrodowa St, 15-027 Bialystok, Poland. [Kruszewska, Joanna] Medical University of Bialystok, Department of Oncology, 12 Ogrodowa St, 15-027 Bialystok, Poland. [Lipska, Alina] Regional Centre for Transfusion MedicineBialystok, Poland. [Radziwon, Piotr] Regional Centre for Transfusion MedicineBialystok, Poland. [Tucker, C. Stephanie] Wayne State University, Department of Pharmacology, 48202 Detroit, MI, USA. [Honn, V. Kenneth] Wayne State University, Department of Pharmacology, 48202 Detroit, MI, USA. RP Wojtukiewicz, M (reprint author), Medical University of Bialystok, Department of Oncology, 15-027 Bialystok, Poland. EM mzwojtukiewicz@gmail.com CR Trousseau A, 1865, Phlegmasia alba dolens. CliniqueMedicale de l′Hotel-Dieu de Paris: The Sydenham Society, pp 94–96 Khorana AA, Francis CW, Culakova E, Fisher RI, Kuderer NM, Lyman GH, 2006, Thromboembolism in hospitalized neutropenic cancer patients. J Clin Oncol 24:484–490 Sallah S, Wan JY, Nguyen NP, 2002, Venous thrombosis in patients with solid tumors: determination of frequency and characteristics. Thromb Haemost 87:575–579 Sorensen HT, Mellemkjaer L, Olsen JH, Baron JA, 2000, Prognosis of cancers associated with venous thromboembolism. N Engl J Med 21:343: 1846–1850 Khorana AA, Francis CW, Culakova E, Kuderer NM, Lyman GH, 2007, Thromboembolism is a leading cause of death in cancer patients receiving outpatient chemotherapy. J Thromb Haemost 5: 632–634 Lee AY, LevineMN(2003, Venous thromboembolismand cancer: risks and outcomes. Circulation 17(23 Suppl 1):I17–I21 107( Rees PA, Clouston HW, Duff S, Kirwan CC, 2018, Colorectal cancer and thrombosis. Int J Colorectal Dis 33:105–108 Heit JA, Silverstein MD, Mohr DN, Petterson TM, O’Fallon WM, Melton LJ 3rd, 2000, Risk factors for deep vein thrombosis and pulmonary embolism: a population-based case-control study. Arch Intern Med 27;160:809–815 Folkman J, 1971, Tumor angiogenesis: therapeutic implications. N Engl J Med 285:1182–1186 Wojtukiewicz MZ, Sierko E, Klement P, Rak J, 2001, The Hemostatic System and Angiogenesis in Malignancy. Neoplasia 3:371–384 Abdulkadir SA, Carvalhal GF, Kaleem Z et al, 2000, Tissue factor expression and angiogenesis in human prostate carcinoma. Hum Pathol 31:443–447 Folkman J, 1996, Tumor angiogenesis and tissue factor. Nat Med 2:167–168 Wojtukiewicz MZ, Sierko E, Rak J, 2004, Contribution of the Hemostatic System to Angiogenesis in Cancer. Semin Thromb Hemost 30:5–20 Yan SF, Zou YS,GaoY, Zhai C, MackmanN, Lee SL,Milbrandt J, Pinsky D, Kisiel W, Stern D, 1998, Tissue factor transcription driven by Egr-1 is a critical mechanism of murine pulmonary fibrin deposition in hypoxia. Proc Natl Acad Sci USA 95:8298–8303 Shweiki D, Itin A, Soffer D, Keshet E, 1992, Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature 359:843–845 Flak B,Wawrzyniec K, Kwiatek S, Kawczyk-Krupka A, Czuba Z, Sieron-Stoltny K, Sieron A, 2013, Vascular endothelial growth factor, VEGF, as a marker for cancer progression - a review. Acta Bio-Optica et Informatica Medica Inzynieria Biomedyczna 19:4: 205–209 Sierko E, Sokol M, Wojtukiewicz MZ, 2015, Mikropecherzyki pochodzenia srodblonkowego, EMP, rola w fizjologii i patologii. Postepy Hig Med Dosw 69:925–932 Nomura S, Niki M, Nisizawa T, Tamaki T, Shimizu M, 2015, Microparticles as Biomarkers of Blood Coagulation in Cancer. Biomark Cancer 7:51–6 Morel O, Toti F, Hugel B et al, 2006, Procoagulant microparticles: disrupting the vascular homeostasis equation? Arteriosclerosis. Thromb Vasc Biol 26:2594–2604 Markiewicz M, Richard E, Marks N, Ludwicka-Bradley A, 2013, Impact of endothelial microparticles on coagulation, inflammation, and angiogenesis in age-related vascular diseases. J Aging Res 2013:734509. , DOI 10.1155/2013/734509 Amabile N, Heiss C, RealWM,Minasi P,McGlothlin D, Rame EJ, Grossman W, De Marco T, Yeghiazarians Y, 2008, Circulating endothelial microparticle levels predict hemodynamic severity of pulmonary hypertension. Am J Respir Crit Care Med 177:1268– 1275 Jimenez JJ, JyW,Mauro LM, Soderland C, Horstman LL, Ahn YS, 2003, Endothelial cells release phenotypically and quantitatively distinct microparticles in activation and apoptosis. Thromb Res 109:175–180 Silva J, Garcia V, Rodriguez M, Compte M, Cisneros E, Veguillas P, Garcia JM, Dominguez G, Campos-Martin Y, Cuevas J, Pena C, Herrera M, Diaz R, Mohammed N, Bonilla F, 2012, Analysis of exosome release and its prognostic value in human colorectal cancer. Genes Chromosomes Cancer 51:409–418 Stec M, Baj-KrzyworzekaM, Baran J,Weglarczyk K, ZembalaM, Barbasz J, Szczepanik A, Zembala M, 2015, Isolation and characterization of circulating micro(nano)vesicles in the plasma of colorectal cancer patients and their interactions with tumor cells. Oncol Rep 34:2768–2775 Goon PKY, Lip GYH, Boos CJ, Stonelake PS, Blann AD, 2006, Circulating endothelial cells, endothelial progenitor cells, and endothelial microparticles in cancer. Neoplasia 8:79–88 Pabinger I, Thaler J, Ay C, 2013, Biomarkers for prediction of venous thromboembolism in cancer. Blood 122:2011–2018 Wojtukiewicz MZ, Sierko E, Kisiel W, 2007, The role of hemostatic system inhibitors in malignancy. Semin Thromb Hemost 33: 621–642 Wojtukiewicz MZ, Hempel D, Sierko E, Tucker SC, Honn KV, 2016, Thrombin-unique coagulation system protein with multifaceted impacts on cancer and metastasis. Cancer Metastasis Rev 35: 213–233 Sierko E, Wojtukiewicz MZ, Zimnoch L, 2010, Expression of TFPI in breast cancer and colon cancer. Thromb Haemost 103: 198–204 Woj t ukiewicz MZ, Sierko E, Zimnoch L, 2003, Immunohistochemical localization of TFPI-2 in human tumor tissue. Thromb Haemost 90:140–146 Wojtukiewicz MZ, Hempel D, Sierko E, Tucker ST, Honn KV, 2015, Protease-activated receptors, PARs, – biology and role in cancer invasion and metastasis. Cancer Metast Rev 34:775–796 Wojtukiewicz MZ, Hempel D, Sierko E, Tucker SC, Honn KV, 2019, Endothelial Protein C Receptor, EPCR), Protease Activated Receptor-1, PAR-1, and their interplay in cancer growth and metastatic dissemination. Review. Cancers, Basel, 8:11. , DOI 10.3390/cancers11010051 Falanga A, MarchettiM(2012, Anticancer treatment and thrombosis. Thromb Res Mar 129:353–359 Haddad TC, Greeno EW, 2006, Chemotherapy-induced thrombosis. Thromb Res 118:555–568 Seng S, Liu Z, Chiu SK, Proverbs-Singh T, Sonpavde G, Choueiri TK, Tsao CK, Yu M, Hahn NM, OhWK, GalskyMD, 2012, Risk of venous thromboembolism in patients with cancer treated with Cisplatin: a systematic review and meta-analysis. J Clin Oncol 10;30:4416–4426 Swystun LL, Mukherjee S, Liaw PC, 2011, Breast cancer chemotherapy induces the release of cell-free DNA, a novel procoagulant stimulus. J Thromb Haemost 9:2313–2321 Hugel B, Martinez MC, Kunzelmann C, Freyssinet JM, 2005, Membrane microparticles: two sides of the coin. Physiology, Bethesda, 20:22–27 Morel O, Toti F, Hugel B, Freyssinet JM(2004, Cellular microparticles: a disseminated storage pool of bioactive vascular effectors. Curr Opin Hematol 11:156–164 VanWijk MJ, VanBavel E, Sturk A, Nieuwland R, 2003, Microparticles in cardiovascular diseases. Cardiovasc Res 59: 277–287 Hron G, Kollars M, Weber H, Sagaster V, Quehenberger P, Eichinger S, Kyrle PA, Weltermann A, 2007, Tissue factorpositive microparticles: cellular origin and association with coagulation activation in patients with colorectal cancer. Thromb Haemost 97:119–123 Khorana AA, Francis CW, Menzies KE, Wang JG, Hyrien O, Hathcock J, Mackman N, Taubman MB, 2008, Plasma tissue factor may be predictive of venous thromboembolism in pancreatic cancer. J Thromb Haemost 6:1983–1985 Thaler J, Ay C,Mackman N, Bertina RM, Kaider A, Marosi C, Key NS, Barcel DA, Scheithauer W, Kornek G, Zielinski C, Pabinger I, 2012, Microparticle-associated tissue factor activity, venous thromboembolism and mortality in pancreatic, gastric, colorectal and brain cancer patients. J Thromb Haemost 10:1363–1370 Wang M, Fu Y, Xu L, Xiao L, Yue Y, Liu S, Huang Q, Li S, Li Y, 2018, Diagnostic value of platelet-derived microparticles in pulmonary thromboembolism: A population-based study. Exp Ther Med 16:3099–3106 Tesselaar ME, Romijn FP, Van Der Linden IK, Prins FA, Bertina RM, Osanto S, 2007, Microparticle-associated tissue factor activity: a link between cancer and thrombosis? J Thromb Haemost 5: 520–527 Enjeti AK, Lincz LF, SeldonM(2007, Detection andmeasurement of microparticles: an evolving research tool for vascular biology. Semin Thromb Hemost 33:771–779 Chirinos JA, Heresi GA, Velasquez H, Jy W, Jimenez JJ, Ahn E, Horstman LL, Soriano AO, Zambrano JP, Ahn YS, 2005, Elevation of endothelial microparticles, platelets, and leukocyte activation in patients with venous thromboembolism. J Am Coll Cardiol 45:1467–1471 Ye R, Ye C, Huang Y, Liu L, Wang S, 2012, Circulating tissue factor positive microparticles in patients with acute recurrent deep venous thrombosis. Thromb Res 130:253–258 Deguchi K, Noguchi M, Yuwasaki E, Endou T, Deguchi A, Wada H, Murashima S,Nishikawa M, Shirakawa S, TanakaK,Kusagawa M, 1991, Dynamic fluctuations in blood of thrombin/antithrombin III complex, TAT). Am J Hematol 38:86–89 Haas SL, Jesnowski R, SteinerM, Hummel F, Ringel J, Burstein C, Nizze H, Liebe S, Lohr JM, 2006, Expression of tissue factor in pancreatic adenocarcinoma is associated with activation of coagulation. World J Gastroenterol 12:4843–4849 van Duijnhoven EM, Lustermans FA, van Wersch JW, 1993, Evaluation of the coagulation/fibrinolysis balance in patients with colorectal cancer. Haemostasis 23:168–172 Tesselaar ME, Romijn FP, van der Linden IK, Bertina RM, Osanto S, 2009, Microparticle-associated tissue factor activity in cancer patients with and without thrombosis. J Thromb Haemost 7: 1421–1423 Lechner D, Weltermann A, 2008, Chemotherapy-induced thrombosis: a role for microparticles and tissue factor? Semin Thromb Hemost 34:199–203 Ferrara N, 2001, Role of vascular endothelial growth factor in regulation of physiological angiogenesis. Am J Physiol Cell Physiol 280:C1358–C1366 Sakurai T, Kudo M, 2011, Signaling pathways governing tumor angiogenesis. Oncology 81(Suppl 1):24–29 Volm M, Koomagi R, Mattern J, 1999, PD-ECGF, bFGF, and VEGF expression in non-small cell lung carcinomas and their association with lymph node metastasis. Anticancer Res 19:651–655 Seo HY, Park JM, Park KH, Kim SJ, Oh SC, Kim BS, Kim YH, Kim JS, 2010, Prognostic significance of serum vascular endothelial growth factor per platelet count in unresectable advanced gastric cancer patients. Jpn J Clin Oncol 40:1147–1153 Ferroni P, Spila A, Martini F, D’Alessandro R, Mariotti S, Del Monte G, Graziano P, Buonomo O, Guadagni F, Roselli M, 2005, Prognostic value of vascular endothelial growth factor tumor tissue content of colorectal cancer. Oncology 69:145–153 ChinKF,Greenman J, Gardiner E, Kumar H, Topping K,Monson J, 2000, Pre-operative serum vascular endothelial growth factor can select patients for adjuvant treatment after curative resection in colorectal cancer. Br J Cancer 83:1425–1431 Ng T, Phey XY, Yeo HL, Shwe M, Gan YX, Ng R, Ho HK, Chan A, 2018, Impact of Adjuvant Anthracycline-Based and Taxane- Based Chemotherapy on Plasma VEGF Levels and Cognitive Function in Breast Cancer Patients: A Longitudinal Study. Clin Breast Cancer 18:e927–e937 Neves KB, Rios FJ, Jones R, Evans TRJ, Montezano AC, Touyz RM, 2019, Microparticles from vascular endothelial growth factor pathway inhibitor-treated cancer patients mediate endothelial cell injury. Cardiovasc Res 115:978–988 Giusti I,Delle Monache S,Di FrancescoM, Sanita P,D’Ascenzo S, Gravina GL, Festuccia C, Dolo V, 2016, From glioblastoma to endothelial cells through extracellular vesicles: messages for angiogenesis. Tumour Biol 37:12743–12753 Kim CW, Lee HM, Lee TH, Kang C, Kleinman HK, Gho YS, 2002, Extracellular membrane vesicles from tumor cells promote angiogenesis via sphingomyelin. Cancer Res 62:6312–6317 KosakaN,YoshiokaY, FujitaY,Ochiya T, 2016)Versatile roles of extracellular vesicles in cancer. J Clin Invest 126:1163–1172 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2499 EP 2507 DI 10.1007/s12253-020-00854-8 PG 9 ER PT J AU Marquet, B Bressenot, MA Fichel, C Bouland, N Barbe, C Bouche, O Kianmanesh, R Diebold, MD Boulagnon-Rombi, C AF Marquet, Benjamin Bressenot, Marchal Aude Fichel, Caroline Bouland, Nicole Barbe, Coralie Bouche, Olivier Kianmanesh, Reza Diebold, Marie-Daniele Boulagnon-Rombi, Camille TI Expression of the Serrated Markers Annexin A10 or Gremlin1 in Colonic Adenocarcinomas: Morphology and Prognostic Values SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal neoplasms; AnnexinA10; Gremlin1; Prognosis; Serrated adenocarcinoma ID Colorectal neoplasms; AnnexinA10; Gremlin1; Prognosis; Serrated adenocarcinoma AB Describe clinical, histological and molecular charatcteristics and prognosis values of the serrated candidate markers AnnexinA10 and Gremlin1 in colon adenocarcinomas. Immunohistochemical expression of AnnexinA10 and Gremlin1 was evaluated on 346 colonic adenocarcinomas. Clinicopathological, molecular features and prognostic characteristics were then evaluated. A total of 40 colonic adenocarcinomas expressed AnnexinA10 (11.6%) and, 115 expressed Gremlin1 (40.4%). AnnexinA10 expression was significantly associated, on univariate analyses, with female gender (p = 0.03), right tumor location (p < 0.001), differentiation grade 3 (p < 0.001), serrated adenocarcinoma subtype (p < 0.001), serrated (p < 0.001), medullary (p = 0.005), and mucinous component (p = 0.004), cytoplasmic eosinophilia (p < 0.001), discernible nuclei (p = 0.001), preserved polarity (p < 0.001), lymphatic invasion (p = 0.01), BRAFV600E mutation (p < 0.001), MSI-H status (p < 0.001) and CIMP-H status (p = 0.019). Multivariate analyses revealed that mucinous component (p = 0.002), lymphatic invasion (p = 0.02) and BRAFV600E mutation (p < 0.001) were independently associated with AnnexinA10 expression. In addition, AnnexinA10 was an indicator of poorer overall survival (OS) in UICC stage IV adenocarcinomas (p = 0.01) only. Gremlin1 expression was neither associated with serrated adenocarcinoma subtype (p = 0.51) nor with AnnexinA10 expression (p = 0,31), but was significantly associated, in univariate analysis with male gender (p = 0.002), younger age (p = 0.002), left tumor location (p=0.04), andMSS status (p = 0.03). Gremlin1 expression was associated with better OS only in UICC stage III colon adenocarcinomas (p = 0.006). Colon adenocarcinomas expressing AnnexinA10 have distinct clinico-pathological and molecular features. AnnexinA10 expression is an indicator of poorer OS in UICC stage IV patients. Gremlin1 expression is not associated with serrated adenocarcinomas subtype. Its expression was associated with better OS in UICC Stage III patients. C1 [Marquet, Benjamin] Academic Hospital, Department of Biopathology, rue du General Koenig, 51100 Reims, France. [Bressenot, Marchal Aude] Academic Hospital, Department of Biopathology, rue du General Koenig, 51100 Reims, France. [Fichel, Caroline] Medicine University, Department of PathologyReims, France. [Bouland, Nicole] Medicine University, Department of PathologyReims, France. [Barbe, Coralie] Academic Hospital, Clinical Research UnitReims, France. [Bouche, Olivier] Academic Hospital, Gatroenterology and Digestive Oncology DepartmentReims, France. [Kianmanesh, Reza] Academic Hospital, Digestive Surgery DepartmentReims, France. [Diebold, Marie-Daniele] Academic Hospital, Department of Biopathology, rue du General Koenig, 51100 Reims, France. [Boulagnon-Rombi, Camille] Academic Hospital, Department of Biopathology, rue du General Koenig, 51100 Reims, France. RP Marquet, B (reprint author), Academic Hospital, Department of Biopathology, 51100 Reims, France. EM marquetbenjamin@orange.fr CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68:394–424. , DOI 10.3322/caac.21492 Makinen MJ, 2007, Colorectal serrated adenocarcinoma. Histopathology 50:131–150. , DOI 10.1111/j.1365-2559. 2006.02548.x Makinen MJ, 2014, Serrated polyps and colorectal cancer risk. Colorectal Cancer 3:77–91. , DOI 10.2217/crc.13.84 Nagtegaal ID, Odze RD, Klimstra D et al, 2019, WHO classification of tumours of the digestive system. In: WHO classification of tumours editorial board, 5th edn. Lyon, pp 157-192 Tuppurainen K, Makinen JM, Junttila O, Liakka A, Kyllonen AP, Tuominen H, Karttunen TJ, Makinen MJ, 2005, Morphology and microsatellite instability in sporadic serrated and non-serrated colorectal cancer. J Pathol 207:285–294. , DOI 10.1002/path. 1850 Sajanti SA, Vayrynen JP, Sirnio P, Klintrup K,Makela J, Tuomisto A, Makinen MJ, 2015, Annexin A10 is a marker for the serrated pathway of colorectal carcinoma. Virchows Arch 466:5–12. https:// doi.org/10.1007/s00428-014-1683-6 Mussunoor S, Murray G, 2008, The role of annexins in tumour development and progression. J Pathol 216:131–140. https://doi. org/10.1002/path.2400 Munksgaard PP, Mansilla F, Brems Eskildsen A-S, Fristrup N, Birkenkamp-Demtroder K, Ulhoi BP, Borre M, Agerbaek M, Hermann GG, Orntoft TF, Dyrskjot L, 2011, Low ANXA10 expression is associated with disease aggressiveness in bladder cancer. Br J Cancer 105:1379–1387. , DOI 10.1038/bjc.2011. 404 Kim J, Kim MA, Jee CD, Jung EJ, Kim WH, 2009, Reduced expression and homozygous deletion of annexin A10 in gastric carcinoma. Int J Cancer 125:1842–1850. , DOI 10.1002/ ijc.24541 Liu S-H, Lin C-Y, Peng S-Y, Jeng YM, Pan HW, Lai PL, Liu CL, Hsu HC, 2002, Down-regulation of Annexin A10 in hepatocellular carcinoma is associated with vascular invasion, early recurrence, and poor prognosis in synergy with p53 mutation. Am J Pathol 160:1831–1837. , DOI 10.1016/S0002-9440(10)61129-7 Shimizu T, Kasamatsu A, Yamamoto A, Koike K, Ishige S, Takatori H, Sakamoto Y, Ogawara K, Shiiba M, Tanzawa H, Uzawa K, 2012, Annexin A10 in human oral cancer: biomarker for tumoral growth via G1/S transition by targeting MAPK signaling pathways. PLoS One 7:e45510. , DOI 10.1371/journal. pone.0045510 Bae JM, 2015, Annexin A10 expression in colorectal cancers with emphasis on the serrated neoplasia pathway. World J Gastroenterol 21:9749–9757. , DOI 10.3748/wjg.v21.i33.9749 Kim JH, Rhee Y-Y, Kim K-J, Cho NY, Lee HS, Kang GH, 2014, Annexin A10 expression correlates with serrated pathway features in colorectal carcinoma with microsatellite instability. APMIS 122: 1187–1195. , DOI 10.1111/apm.12284 Tsai J-H, Lin Y-L, ChengY-C, Chen CC, Lin LI, Tseng LH, Cheng ML, Liau JY, JengYM(2015, Aberrant expression of annexin A10 is closely related to gastric phenotype in serrated pathway to colorectal carcinoma. Mod Pathol 28:268–278. , DOI 10.1038/ modpathol.2014.96 Pelli A, Vayrynen JP, Klintrup K, Makela J, Makinen MJ, Tuomisto A, Karttunen TJ, 2016, Gremlin1 expression associates with serrated pathway and favourable prognosis in colorectal cancer. Histopathology 69:831–838. , DOI 10.1111/his.13006 YamasakiY, Ishigami S,Arigami T, Kita Y,Uchikado Y, Kurahara H, Kijima Y, Maemura K, Natsugoe S, 2018, Expression of gremlin1 in gastric cancer and its clinical significance. Med Oncol 35(3): 30. , DOI 10.1007/s12032-017-1073-4 Sobin LH, Gospodarowicz MK,Wittekind C, 2010, TNM classification of malignant tumours. In: Wiley-Blackwell, 7th edn. Chichester, West Sussex, UK, pp 309 Wang LM, Kevans D, Mulcahy H, O Sullivan J, Fennelly D, Hyland J, O Donoghue D, Sheahan K, 2009, Tumor budding is a strong and reproducible prognostic marker in T3N0 colorectal cancer. Am J Surg Pathol 33:134–141. , DOI 10.1097/PAS. 0b013e318184cd55 Boulagnon C, Dudez O, Beaudoux O, Dalstein V, Kianmanesh R, Bouche O, Diebold MD, 2016, BRAFV600E gene mutation in colonic adenocarcinomas. Immunohistochemical detection using tissue microarray and clinicopathologic characteristics: an 86 case series.Appl ImmunohistochemMolMorphol 24:88–96. https://doi. org/10.1097/PAI.0000000000000157 Boulagnon-Rombi C, Schneider C, Leandri C, Jeanne A, Grybek V, Bressenot AM, Barbe C,Marquet B, Nasri S, Coquelet C, Fichel C, Bouland N, Bonnomet A, Kianmanesh R, Lebre AS, Bouche O, DieboldMD, Bellon G, Dedieu S, 2018, LRP1 expression in colon cancer predicts clinical outcome. Oncotarget 9:8849–8869. https:// doi.org/10.18632/oncotarget.24225 Jass JR, Smith M(1992, Sialic acid and epithelial differentiation in colorectal polyps and cancer. A morphological, mucin and lectin histochemical study. Pathology 24:233–242. , DOI 10. 3109/00313029209068874 Garcia-Solano J, Perez-Guillermo M, Conesa-Zamora P, Acosta- Ortega J, Trujillo-Santos J, Cerezuela-Fuentes P, Makinen MJ, 2010, Clinicopathologic study of 85 colorectal serrated adenocarcinomas: further insights into the full recognition of a new subset of colorectal carcinoma. Hum Pathol 41:1359–1368. , DOI 10.1016/j.humpath.2010.04.002 Shida Y, Fujimori T, Tanaka H, Fujimori Y, Kimura R, Ueda H, Ichikawa K, Tomita S, Nagata H, Kubota K, Tsubaki M, Kato H, Yao T, Sugai T, Sugihara K, Ohkura Y, Imura J, 2012, Clinicopathological features of serrated adenocarcinoma defined by Makinen in dukes’ B colorectal carcinoma. Pathobiology 79: 169–174. , DOI 10.1159/000334837 Makinen MJ, George SM, Jernvall P et al, 2001, Colorectal carcinoma associated with serrated adenoma - prevalence, histological features, and prognosis. J Pathol 193:286–294. , DOI 10. 1002/1096-9896(2000)9999:9999<::AID-PATH800>3.0.CO;2-2 Lee C-T, Huang Y-C, Hung L-Y, Chow NH, Su PF, Ho CL, Tsai HW, Chen YL, Lin SC, Lin BW, Lin PC, Lee JC, 2017, Serrated adenocarcinoma morphology in colorectal mucinous adenocarcinoma is associated with improved patient survival. Oncotarget 8: 35165–35175. , DOI 10.18632/oncotarget.16815 Stefanius K, Ylitalo L, Tuomisto A, Kuivila R, Kantola T, Sirnio P, Karttunen TJ, Makinen MJ, 2011, Frequent mutations of KRAS in addition to BRAF in colorectal serrated adenocarcinoma: KRAS and BRAF in serrated adenocarcinoma. Histopathology 58:679– 692. , DOI 10.1111/j.1365-2559.2011.03821.x Conesa-Zamora P, Garcia-Solano J, Garcia-Garcia F, Turpin MC, Trujillo-Santos J, Torres-Moreno D, Oviedo-Ramirez I, Carbonell- Munoz R, Munoz-Delgado E, Rodriguez-Braun E, Conesa A, Perez-Guillermo M, 2013, Expression profiling shows differential molecular pathways and provides potential new diagnostic biomarkers for colorectal serrated adenocarcinoma. Int J Cancer 132: 297–307. , DOI 10.1002/ijc.27674 Kim JH, Bae JM, Cho N-Y, Kang GH, 2016, Distinct features between MLH1-methylated and unmethylated colorectal carcinomas with the CpG island methylator phenotype: implications in the serrated neoplasia pathway. Oncotarget 7:14095–14111. , DOI 10.18632/oncotarget.7374 Garcia-Solano J, Conesa-Zamora P, Carbonell P, Trujillo-Santos J, Torres-Moreno D D, Pagan-Gomez I, Rodriguez-Braun E, Perez- Guillermo M, 2012, Colorectal serrated adenocarcinoma shows a different profile of oncogenemutations,MSI status andDNA repair protein expression compared to conventional and sporadic MSI-H carcinomas. Int J Cancer 131:1790–1799. , DOI 10.1002/ ijc.27454 Gonzalo DH, Lai KK, Shadrach B, Goldblum JR, Bennett AE, Downs-Kelly E, Liu X, Henricks W, Patil DT, Carver P, Na J, Gopalan B, Rybicki L, Pai RK, 2013, Gene expression profiling of serrated polyps identifies annexin A10 as a marker of a sessile serrated adenoma/polyp. J Pathol 230:420–429. , DOI 10. 1002/path.4200 De Sousa EMelo F,Wang X, JansenMet al, 2013, Poor-prognosis colon cancer is defined by a molecularly distinct subtype and develops from serrated precursor lesions. Nat Med 19:614–618. , DOI 10.1038/nm.3174 Rad R, Cadinanos J, Rad L, Varela I, Strong A, Kriegl L, Constantino-Casas F, Eser S, Hieber M, Seidler B, Price S, Fraga MF, Calvanese V, Hoffman G, Ponstingl H, Schneider G, Yusa K, Grove C, Schmid RM, Wang W, Vassiliou G, Kirchner T, McDermott U, Liu P, Saur D, Bradley A, 2013, A genetic progression model of BrafV600E-induced intestinal tumorigenesis reveals targets for therapeutic intervention. Cancer Cell 24:15–29. https:// doi.org/10.1016/j.ccr.2013.05.014 Guinney J, Dienstmann R, Wang X, de Reynies A, Schlicker A, Soneson C, Marisa L, Roepman P, Nyamundanda G, Angelino P, Bot BM, Morris JS, Simon IM, Gerster S, Fessler E, de Sousa E Melo F, Missiaglia E, Ramay H, Barras D, Homicsko K, Maru D, Manyam GC, Broom B, Boige V, Perez-Villamil B, Laderas T, Salazar R, Gray JW, Hanahan D, Tabernero J, Bernards R, Friend SH, Laurent-Puig P, Medema JP, Sadanandam A, Wessels L, DelorenziM, Kopetz S, Vermeulen L, Tejpar S, 2015, The consensus molecular subtypes of colorectal cancer. Nat Med 21:1350– 1356. , DOI 10.1038/nm.3967 Dalerba P, Sahoo D, Paik S, Guo X, Yothers G, Song N, Wilcox- Fogel N, Forgo E, Rajendran PS, Miranda SP, Hisamori S, Hutchison J, Kalisky T, Qian D, Wolmark N, Fisher GA, van de Rijn M, Clarke MF, 2016, CDX2 as a prognostic biomarker in stage II and stage III colon cancer. N Engl J Med 374:211–222. , DOI 10.1056/NEJMoa1506597 Tomasello G, Barni S, Turati L, Ghidini M, Pezzica E, Passalacqua R, Petrelli F, 2018, Association of CDX2 expression with survival in early colorectal cancer: a systematic review and meta-analysis. Clin Colorectal Cancer 17:97–103. , DOI 10.1016/j.clcc. 2018.02.001 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2509 EP 2521 DI 10.1007/s12253-020-00857-5 PG 13 ER PT J AU Hegedus, L Rittler, D Garay, T Stockhammer, P Kovacs, I Dome, B Theurer, S Hager, Th Herold, Th Kalbourtzis, S Bankfalvi, Schmid, K Fuhrer, D Aigner, C Hegedus, B AF Hegedus, Luca Rittler, Dominika Garay, Tamas Stockhammer, Paul Kovacs, Ildiko Dome, Balazs Theurer, Sarah Hager, Thomas Herold, Thomas Kalbourtzis, Stavros Bankfalvi, Agnes Schmid, W. Kurt Fuhrer, Dagmar Aigner, Clemens Hegedus, Balazs TI HDAC Inhibition Induces PD-L1 Expression in a Novel Anaplastic Thyroid Cancer Cell Line SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Anaplastic thyroid cancer; BRAF mutation; TERT promoter mutation; HDAC inhibition; Pleural effusion ID Anaplastic thyroid cancer; BRAF mutation; TERT promoter mutation; HDAC inhibition; Pleural effusion AB While papillary thyroid cancer (PTC) has largely favorable prognosis, anaplastic thyroid cancer (ATC) is a rare but extremely aggressive malignancy with grim clinical outcome. Even though new therapeutic options are emerging for ATC, additional preclinical models and novel combinations are needed for specific subsets of patients. We established a novel cell line (PF49) from the malignant pleural effusion of a 68-year-old male patient with ATC that rapidly transformed from a BRAF and TERT promoter mutant PTC. PF49 cells demonstrated a robust migratory activity in vitro and strong invasive capacity in vivo in a pleural carcinosis model. Combined BRAF andMEK inhibition decreased the proliferation and migration of PF49 cells, however could not induce cell death. Importantly, HDAC inhibitor treatment with SAHA or valproic acid induced cell cycle arrest and strongly increased PD-L1 expression of the tumor cells. Induction of PD-L1 expression was also present when paclitaxelcisplatin chemotherapeutic treatment was combined with HDAC inhibitor treatment. Increased PD-L1 expression after HDAC inhibition was recapitulated in an international ATC cell model. Our data suggest that HDAC inhibition alone or in combination with standard chemotherapy may potentiate anaplastic thyroid cancer cells for immunotherapy. C1 [Hegedus, Luca] University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Rittler, Dominika] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Garay, Tamas] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Stockhammer, Paul] University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Kovacs, Ildiko] National Koranyi Institute of PulmonologyBudapest, Hungary. [Dome, Balazs] Medical University of Vienna, Department of Thoracic SurgeryVienna, Austria. [Theurer, Sarah] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Hager, Thomas] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Herold, Thomas] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Kalbourtzis, Stavros] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Bankfalvi, Agnes] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Schmid, W. Kurt] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Fuhrer, Dagmar] University Duisburg-Essen, University Clinic Essen, Department of EndocrinologyEssen, Germany. [Aigner, Clemens] University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Hegedus, Balazs] University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. RP Hegedus, B (reprint author), University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic Surgery, Essen, Germany. EM balazs.hegedues@rlk.uk-essen.de CR Ragazzi M, Ciarrocchi A, Sancisi V, Gandolfi G, Bisagni A, Piana S, 2014, Update on anaplastic thyroid carcinoma: morphological, molecular, and genetic features of the most aggressive thyroid cancer. Int J Endocrinol 2014:790834–790813. , DOI 10. 1155/2014/790834 Dal Maso L, Tavilla A, Pacini F, Serraino D, BAC v D, Chirlaque MD, Capocaccia R, Larranaga N, ColonnaM, Agius D, Ardanaz E, Rubio-Casadevall J, Kowalska A, Virdone S,Mallone S,Amash H, De Angelis R, Group E-W, 2017, Survival of 86,690 patients with thyroid cancer: a population-based study in 29 European countries from EUROCARE-5. Eur J Cancer 77:140–152. , DOI 10. 1016/j.ejca.2017.02.023 Pozdeyev N, Gay LM, Sokol ES, Hartmaier R, Deaver KE, Davis S, French JD, Borre PV, LaBarbera DV, Tan AC, Schweppe RE, Fishbein L, Ross JS, Haugen BR, Bowles DW, 2018, Genetic analysis of 779 advanced differentiated and anaplastic thyroid cancers. Clin Cancer Res 24:3059–3068. , DOI 10.1158/1078- 32.CCR-18-0373 4. Park JH, Kwon HJ, Park CS, Hong S, 2014, Anaplastic transformation of papillary thyroid carcinoma in a young man: a case study with Immunohistochemical and BRAF analysis. Korean J Pathol 48(3):234–240. , DOI 10.4132/KoreanJPathol.2014.48.3. 234 Abe T, SuzukiM, Shimizu K, ShinagawaN, Oizumi S,Matsuno Y, Miyazaki M, Tanino M, Tanaka S, NishimuraM(2014, Anaplastic transformation of papillary thyroid carcinoma in multiple lung metastases presenting with a malignant pleural effusion: a case report. J Med Case Rep 8:460. , DOI 10.1186/1752-1947-8-460 Chen H, Luthra R, Routbort MJ, Patel KP, Cabanillas ME, Broaddus RR,WilliamsMD, 2018)Molecular profile of advanced thyroid carcinomas by next-generation sequencing: characterizing tumors beyond diagnosis for targeted therapy.Mol Cancer Ther 17: 1575–1584. , DOI 10.1158/1535-7163.MCT-17-0871 Bonhomme B, Godbert Y, Perot G, Al Ghuzlan A, Bardet S, Belleannee G, Criniere L, Do Cao C, Fouilloux G, Guyetant S, Kelly A, Leboulleux S, Buffet C, Leteurtre E, Michels JJ, Tissier F, ToubertME, Wassef M, Pinard C, Hostein I, Soubeyran I, 2017, Molecular pathology of anaplastic thyroid carcinomas: a retrospective study of 144 cases. Thyroid 27(5):682–692. , DOI 10. 1089/thy.2016.0254 Tiedje V, Ting S, Herold T, Synoracki S, Latteyer S, Moeller LC, Zwanziger D, Stuschke M, Fuehrer D, Schmid KW, 2017, NGS based identification of mutational hotspots for targeted therapy in anaplastic thyroid carcinoma. Oncotarget 8((26)):42613–42620. , DOI 10.18632/oncotarget.17300 Vinagre J,Almeida A, Populo H, Batista R, Lyra J, Pinto V, Coelho R, Celestino R, Prazeres H, Lima L, Melo M, da Rocha AG, Preto A, Castro P, Castro L, Pardal F, Lopes JM, Santos LL, Reis RM, Cameselle-Teijeiro J, Sobrinho-Simoes M, Lima J, Maximo V, Soares P, 2013, Frequency of TERT promoter mutations in human cancers. Nat Commun 4:2185. , DOI 10.1038/ ncomms3185 Keutgen XM, Sadowski SM, Kebebew E, 2015, Management of anaplastic thyroid cancer. Gland Surg 4(1):44–51. , DOI 10.3978/j.issn.2227-684X.2014.12.02 Molinaro E, Romei C, Biagini A, Sabini E, Agate L, Mazzeo S, Materazzi G, Sellari-Franceschini S, Ribechini A, Torregrossa L, Basolo F, Vitti P, Elisei R, 2017, Anaplastic thyroid carcinoma: from clinicopathology to genetics and advanced therapies. Nat Rev Endocrinol 13(11):644–660. , DOI 10.1038/nrendo.2017. 76 Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM, Soria JC, Wen PY, Zielinski C, Cabanillas ME, Urbanowitz G, Mookerjee B, Wang D, Rangwala F, Keam B, 2018, Dabrafenib and Trametinib treatment in patients with locally advanced or metastatic BRAF V600-mutant anaplastic thyroid Cancer. J Clin Oncol 36(1):7–13. , DOI 10.1200/JCO.2017.73.6785 Chintakuntlawar AV, Rumilla KM, Smith CY, Jenkins SM, Foote RL, Kasperbauer JL, Morris JC, Ryder M, Alsidawi S, Hilger C, Bible KC, 2017, Expression of PD-1 and PD-L1 in anaplastic thyroid Cancer patients treated with multimodal therapy: results from a retrospective study. J Clin Endocrinol Metab 102(6):1943–1950. , DOI 10.1210/jc.2016-3756 Kollipara R, Schneider B, Radovich M, Babu S, Kiel PJ, 2017, Exceptional response with immunotherapy in a patient with anaplastic thyroid Cancer. Oncologist 22(10):1149–1151. https://doi. org/10.1634/theoncologist.2017-0096 Cantara S, Bertelli E, Occhini R, Regoli M, Brilli L, Pacini F, Castagna MG, Toti P, 2019, Blockade of the programmed death ligand 1, PD-L1, as potential therapy for anaplastic thyroid cancer. Endocrine 64(1):122–129. , DOI 10.1007/s12020-019- 01865-5 Gunda V, Gigliotti B, Ndishabandi D, Ashry T,McCarthyM, Zhou Z, Amin S, Freeman GJ, Alessandrini A, Parangi S, 2018, Combinations of BRAF inhibitor and anti-PD-1/PD-L1 antibody improve survival and tumour immunity in an immunocompetent model of orthotopic murine anaplastic thyroid cancer. Br J Cancer 119(10):1223–1232. , DOI 10.1038/s41416-018-0296-2 Cabanillas ME, Ferrarotto R, Garden AS, Ahmed S, Busaidy NL, Dadu R, Williams MD, Skinner H, Gunn GB, Grosu H, Iyer P, Hofmann MC, Zafereo M, 2018, Neoadjuvant BRAF- and immune-directed therapy for anaplastic thyroid carcinoma. Thyroid 28(7):945–951. , DOI 10.1089/thy.2018.0060 Iyer PC, Dadu R, Gule-Monroe M, Busaidy NL, Ferrarotto R, Habra MA, Zafereo M, Williams MD, Gunn GB, Grosu H, Skinner HD, Sturgis EM, Gross N, Cabanillas ME, 2018, Salvage pembrolizumab added to kinase inhibitor therapy for the treatment of anaplastic thyroid carcinoma. J Immunother Cancer 6(1):68. , DOI 10.1186/s40425-018-0378-y Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R, 2007, FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 12(10):1247– 1252. , DOI 10.1634/theoncologist.12-10-1247 Grabarska A, Luszczki JJ, Nowosadzka E, Gumbarewicz E, Jeleniewicz W, Dmoszynska-Graniczka M, Kowalczuk K, Kupisz K, Polberg K, Stepulak A, 2017, Histone Deacetylase inhibitor SAHA as potential targeted therapy agent for larynx Cancer cells. J Cancer 8(1):19–28. , DOI 10.7150/jca.16655 Yang B, Yu D, Liu J, Yang K, Wu G, Liu H, 2015, Antitumor activity of SAHA, a novel histone deacetylase inhibitor, against murine B cell lymphoma A20 cells in vitro and in vivo. Tumour Biol 36(7):5051–5061. , DOI 10.1007/s13277-015-3156-1 Bubna AK, 2015, Vorinostat-An overview. Vorinostat-An Overview Indian J Dermatol 60(4):419. , DOI 10.4103/ 0019-5154.160511 Richon VM, 2006, Cancer biology: mechanism of antitumour action of vorinostat, suberoylanilide hydroxamic acid), a novel histone deacetylase inhibitor. Br J Cancer 95(Suppl 1):S2–S6. https:// doi.org/10.1038/sj.bjc.6603463 Duenas-Gonzalez A, Candelaria M, Perez-Plascencia C, Perez- Cardenas E, de la Cruz-Hernandez E, Herrera LA, 2008, Valproic acid as epigenetic cancer drug: preclinical, clinical and transcriptional effects on solid tumors. Cancer Treat Rev 34(3): 206–222. , DOI 10.1016/j.ctrv.2007.11.003 Atmaca A, Al-Batran SE, Maurer A, Neumann A, Heinzel T, Hentsch B, Schwarz SE, Hovelmann S, Gottlicher M, Knuth A, Jager E, 2007, Valproic acid, VPA, in patients with refractory advanced cancer: a dose escalating phase I clinical trial. Br J Cancer 97(2):177–182. , DOI 10.1038/sj.bjc.6603851 Brodie SA, Brandes JC, 2014, Could valproic acid be an effective anticancer agent? The evidence so far. Expert Rev Anticanc 14(10): 1097–1100. , DOI 10.1586/14737140.2014.940329 Catalano MG, Fortunati N, Pugliese M, Poli R, Bosco O, Mastrocola R, Aragno M, Boccuzzi G, 2006, Valproic acid, a histone deacetylase inhibitor, enhances sensitivity to doxorubicin in anaplastic thyroid cancer cells. J Endocrinol 191(2):465–472. , DOI 10.1677/joe.1.06970 Noguchi H, Yamashita H, Murakami T, Hirai K, Noguchi Y, Maruta J, Yokoi T, Noguchi S, 2009, Successful treatment of anaplastic thyroid carcinoma with a combination of oral valproic acid, chemotherapy, radiation and surgery. Endocr J 56(2):245–249 Catalano MG, Pugliese M, Gallo M, Brignardello E, Milla P, Orlandi F, Limone PP, Arvat E, Boccuzzi G, Piovesan A, 2016, Valproic acid, a histone Deacetylase inhibitor, in combination with paclitaxel for anaplastic thyroid Cancer: results of a multicenter randomized controlled phase II/III trial. Int J Endocrinol 2016: 2930414–2930418. , DOI 10.1155/2016/2930414 Catalano MG, Pugliese M, Gargantini E, Grange C, Bussolati B, Asioli S, Bosco O, Poli R, Compagnone A, Bandino A, Mainini F, Fortunati N, Boccuzzi G, 2012, Cytotoxic activity of the histone deacetylase inhibitor panobinostat, LBH589, in anaplastic thyroid cancer in vitro and in vivo. Int J Cancer 130(3):694–704. https://doi. org/10.1002/ijc.26057 Lin CL, Tsai ML, Lin CY, Hsu KW, Hsieh WS, Chi WM, Huang LC, Lee CH, 2019, HDAC1 and HDAC2 double knockout triggers cell apoptosis in advanced thyroid Cancer. Int J Mol Sci 20(2). , DOI 10.3390/ijms20020454 Besic N, Gazic B, 2013, Sites of metastases of anaplastic thyroid carcinoma: autopsy findings in 45 cases from a single institution. Thyroid 23(6):709–713. , DOI 10.1089/thy.2012.0252 Maggisano V, Celano M, Lombardo GE, Lepore SM, Sponziello M, Rosignolo F, Verrienti A, Baldan F, Puxeddu E, Durante C, Filetti S, Damante G, Russo D, Bulotta S, 2017, Silencing of hTERT blocks growth and migration of anaplastic thyroid cancer cells. Mol Cell Endocrinol 448:34–40. , DOI 10.1016/j. mce.2017.03.007 Zhang L, ZhangY,Mehta A, BoufraqechM, Davis S,Wang J, Tian Z, Yu Z, Boxer MB, Kiefer JA, Copland JA, Smallridge RC, Li Z, Shen M, Kebebew E, 2015, Dual inhibition of HDAC and EGFR signaling with CUDC-101 induces potent suppression of tumor growth and metastasis in anaplastic thyroid cancer. Oncotarget 6((11)):9073–9085. , DOI 10.18632/oncotarget.3268 Kim S, Schiff BA, Yigitbasi OG, Doan D, Jasser SA, Bekele BN, Mandal M, Myers JN, 2005, Targeted molecular therapy of anaplastic thyroid carcinoma with AEE788. Mol Cancer Ther 4(4): 632–640. , DOI 10.1158/1535-7163.MCT-04-0293 Garay T, Juhasz E, Molnar E, Eisenbauer M, Czirok A, Dekan B, Laszlo V, Hoda MA, Dome B, Timar J, Klepetko W, Berger W, Hegedus B, 2013, Cellmigration or cytokinesis and proliferation?– revisiting the "go or grow" hypothesis in cancer cells in vitro. Exp Cell Res 319(20):3094–3103. , DOI 10.1016/j.yexcr.2013. 08.018 Chou TC, 2010, Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res 70(2):440– 446. , DOI 10.1158/0008-5472.CAN-09-1947 Hegedus L, Garay T, Molnar E, Varga K, Bilecz A, Torok S, Padanyi R, Paszty K, Wolf M, Grusch M, Kallay E, Dome B, Berger W, Hegedus B, Enyedi A, 2017, The plasma membrane Ca(2+, pump PMCA4b inhibits the migratory and metastatic activity of BRAF mutant melanoma cells. Int J Cancer 140(12):2758– 2770. , DOI 10.1002/ijc.30503 Long GV, Flaherty KT, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, Garbe C, Jouary T, Hauschild A, Chiarion- Sileni V, Lebbe C, Mandala M, Millward M, Arance A, Bondarenko I, Haanen J, Hansson J, Utikal J, Ferraresi V, Mohr P, Probachai V, Schadendorf D, Nathan P, Robert C, Ribas A, Davies MA, Lane SR, Legos JJ, Mookerjee B, Grob JJ, 2017, Dabrafenib plus trametinib versus dabrafenib monotherapy in patients with metastatic BRAF V600E/K-mutant melanoma: longterm survival and safety analysis of a phase 3 study. Ann Oncol 28(7):1631–1639. , DOI 10.1093/annonc/mdx176 Kurebayashi J, Otsuki T, Tanaka K, Yamamoto Y, Moriya T, Sonoo H, 2003, Medroxyprogesterone acetate decreases secretion of interleukin-6 and parathyroid hormone-related protein in a new anaplastic thyroid cancer cell line, KTC-2. Thyroid 13(3):249–258. , DOI 10.1089/105072503321582042 Pilli T, Prasad KV, Jayarama S, Pacini F, Prabhakar BS, 2009, Potential utility and limitations of thyroid cancer cell lines as models for studying thyroid cancer. Thyroid 19(12):1333–1342. , DOI 10.1089/thy.2009.0195 Oishi N, Kondo T, Ebina A, Sato Y, Akaishi J, Hino R, Yamamoto N, Mochizuki K, Nakazawa T, Yokomichi H, Ito K, Ishikawa Y, Katoh R, 2017, Molecular alterations of coexisting thyroid papillary carcinoma and anaplastic carcinoma: identification of TERT mutation as an independent risk factor for transformation. Mod Pathol 30(11):1527–1537. , DOI 10.1038/modpathol. 2017.75 LamKY, LuiMC, Lo CY(2001, Cytokeratin expression profiles in thyroid carcinomas. Eur J Surg Oncol 27(7):631–635. https://doi. org/10.1053/ejso.2001.1203 Nonaka D, Tang Y, Chiriboga L, Rivera M, Ghossein R, 2008, Diagnostic utility of thyroid transcription factors Pax8 and TTF-2, FoxE1, in thyroid epithelial neoplasms. Mod Pathol 21(2):192– 200. , DOI 10.1038/modpathol.3801002 SaiseletM, Floor S, TarabichiM, Dom G, Hebrant A, van Staveren WC, Maenhaut C, 2012, Thyroid cancer cell lines: an overview. Front Endocrinol, Lausanne, 3:133. , DOI 10.3389/fendo. 2012.00133 Antonello ZA, Nucera C, 2014, Orthotopic mouse models for the preclinical and translational study of targeted therapies against metastatic human thyroid carcinoma with BRAF(V600E, or wild-type BRAF. Oncogene 33(47):5397–5404. , DOI 10.1038/onc. 2013.544 McFadden DG, Vernon A, Santiago PM, Martinez-McFaline R, Bhutkar A, Crowley DM, McMahon M, Sadow PM, Jacks T, 2014, p53 constrains progression to anaplastic thyroid carcinoma in a Braf-mutant mouse model of papillary thyroid cancer. Proc Natl Acad Sci U S A 111(16):E1600–E1609. , DOI 10. 1073/pnas.1404357111 Nucera C, Porrello A, Antonello ZA, Mekel M, Nehs MA, Giordano TJ, Gerald D, Benjamin LE, Priolo C, Puxeddu E, Finn S, Jarzab B, Hodin RA, Pontecorvi A, Nose V, Lawler J, Parangi S, 2010, B-Raf(V600E, and thrombospondin-1 promote thyroid cancer progression. Proc Natl Acad Sci U S A 107(23):10649–10654. , DOI 10.1073/pnas.1004934107 NehsMA, Nucera C, Nagarkatti SS, SadowPM,Morales-GarciaD, Hodin RA, Parangi S, 2012, Late intervention with anti- BRAF(V600E, therapy induces tumor regression in an orthotopic mouse model of human anaplastic thyroid cancer. Endocrinology 153(2):985–994. , DOI 10.1210/en.2011-1519 Bu R, Siraj AK, Divya SP,KongY, Parvathareddy SK, Al-Rasheed M, Al-Obaisi KAS, Victoria IG, Al-Sobhi SS, Al-Dawish M, Al- Dayel F, Al-Kuraya KS, 2018, Telomerase reverse transcriptase mutations are independent predictor of disease-free survival in middle eastern papillary thyroid cancer. Int J Cancer 142(10):2028– 2039. , DOI 10.1002/ijc.31225 Bullock M, Ren Y, O'Neill C, Gill A, Aniss A, Sywak M, Sidhu S, Delbridge L, Learoyd D, de Vathaire F, Robinson BG, Clifton- Bligh RJ, 2016, TERT promoter mutations are a major indicator of recurrence and death due to papillary thyroid carcinomas. Clin Endocrinol 85(2):283–290. , DOI 10.1111/cen.12999 Chiu HW, Yeh YL, Wang YC, Huang WJ, Chen YA, Chiou YS, Ho SY, Lin P, Wang YJ, 2013, Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, enhances radiosensitivity and suppresses lung metastasis in breast cancer in vitro and in vivo. PLoS One 8(10):e76340. , DOI 10.1371/journal.pone. 0076340 Hegedus L, Padanyi R, Molnar J, Paszty K, Varga K, Kenessey I, Sarkozy E, Wolf M, Grusch M, Hegyi Z, Homolya L, Aigner C, Garay T, Hegedus B, Timar J, Kallay E, Enyedi A, 2017, Histone Deacetylase inhibitor treatment increases the expression of the plasma membrane Ca(2+, pump PMCA4b and inhibits the migration of melanoma cells independent of ERK. Front Oncol 7:95. https://doi. org/10.3389/fonc.2017.00095 Park J, Thomas S, Munster PN, 2015, Epigenetic modulation with histone deacetylase inhibitors in combination with immunotherapy. Epigenomics 7(4):641–652. , DOI 10.2217/epi.15.16 Terranova-Barberio M, Thomas S, Ali N, Pawlowska N, Park J, Krings G, RosenblumMD, Budillon A,Munster PN, 2017, HDAC inhibition potentiates immunotherapy in triple negative breast cancer. Oncotarget 8((69)):114156–114172. , DOI 10.18632/ oncotarget.23169 Woods DM, Sodre AL, Villagra A, Sarnaik A, Sotomayor EM, Weber J, 2015, HDAC inhibition Upregulates PD-1 ligands in melanoma and augments immunotherapy with PD-1 blockade. Cancer Immunol Res 3(12):1375–1385. , DOI 10.1158/ 2326-6066.CIR-15-0077-T Booth L, Roberts JL, Poklepovic A, Kirkwood J, Dent P, 2017, HDAC inhibitors enhance the immunotherapy response of melanoma cells. Oncotarget 8, 47):83155-83170. , DOI 10.18632/ oncotarget.17950 Bastman JJ, Serracino HS, Zhu Y, Koenig MR, Mateescu V, Sams SB, Davies KD, Raeburn CD, McIntyre RC Jr, Haugen BR, French JD, 2016, Tumor-infiltrating T cells and the PD-1 checkpoint pathway in advanced differentiated and anaplastic thyroid Cancer. J Clin Endocrinol Metab 101(7):2863–2873. , DOI 10. 1210/jc.2015-4227 Goodman AM, Piccioni D, Kato S, Boichard A, Wang HY, Frampton G, Lippman SM, Connelly C, Fabrizio D, Miller V, Sicklick JK, Kurzrock R, 2018, Prevalence of PDL1 amplification and preliminary response to immune checkpoint blockade in solid tumors. JAMA Oncol 4(9):1237–1244. , DOI 10.1001/ jamaoncol.2018.1701 MaM, Lin B,WangM, Liang X, Su L, Okose O, LvW, Li J, 2020, Immunotherapy in anaplastic thyroid cancer. Am J Transl Res 12(3):974–988 Brauner E, Gunda V, Vanden Borre P, Zurakowski D, Kim YS, Dennett KV, Amin S, Freeman GJ, Parangi S, 2016, Combining BRAF inhibitor and anti PD-L1 antibody dramatically improves tumor regression and anti tumor immunity in an immunocompetent murine model of anaplastic thyroid cancer. Oncotarget 7((13)): 17194–17211. , DOI 10.18632/oncotarget.7839 Cabanillas M, Busaidy N, Dadu R, Ferrarotto R, Gross N, Gule- Monroe M, Lu C,GrosuH,WilliamsM, ZafereoM(2019)OR27-6 combination Vemurafenib, BRAF inhibitor)/Cobimetinib, MEK inhibitor)/Atezolizumab, anti-PDL1 inhibitor, in BRAF-V600E mutated anaplastic thyroid Cancer, ATC): initial safety and feasibility. Journal of the Endocrine Society 3((Supplement_1)). https:// doi.org/10.1210/js.2019-OR27-6 Perri F, Lorenzo GD, Scarpati GD, Buonerba C, 2011, Anaplastic thyroid carcinoma: a comprehensive review of current and future therapeutic options. World J Clin Oncol 2(3):150–157. https://doi. org/10.5306/wjco.v2.i3.150 Voigt W, Kegel T, Weiss M, Mueller T, Simon H, Schmoll HJ, 2005, Potential activity of paclitaxel, vinorelbine and gemcitabine in anaplastic thyroid carcinoma. J Cancer Res Clin Oncol 131(9): 585–590. , DOI 10.1007/s00432-005-0673-0 Catalano MG, Poli R, Pugliese M, Fortunati N, Boccuzzi G, 2007, Valproic acid enhances tubulin acetylation and apoptotic activity of paclitaxel on anaplastic thyroid cancer cell lines. Endocr Relat Cancer 14(3):839–845. , DOI 10.1677/ERC-07-0096 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2523 EP 2535 DI 10.1007/s12253-020-00834-y PG 13 ER PT J AU Tabuso, M Christian, M Kimani, P Gopalakrishnan, K Arasaradnam, R AF Tabuso, Maria Christian, Mark Kimani, K. Peter Gopalakrishnan, Kishore Arasaradnam, Ramesh TI KRAS Status is Associated with Metabolic Parameters in Metastatic Colorectal Cancer According to Primary Tumour Location SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; KRAS; Metabolic syndrome; Tumour location; Lipids ID Colorectal cancer; KRAS; Metabolic syndrome; Tumour location; Lipids AB Colorectal cancer (CRC) is characterized by complex interplay between macroenvironmental factors and tumour microenvironment, leading to variable outcomes in CRC patients. To date, there is still a need to identify macroenvironment/microenvironment factors that could define subgroup of patients that would benefit from specific anti-cancer treatment in order to improve patient selection for individualized targeted-based therapy. Aim of this study was to evaluate associations between metabolic parameters and KRAS status in metastatic CRC (mCRC) according to a new tumour site classification. Retrospective data were extracted from a total of 201 patients diagnosed with mCRC between 2012 and 2017 extracted from an established CRC database at our tertiary institute. Clinical-pathological data, including age, gender, BMI, hypertension, diabetes, pre-CRC diagnosis serum lipid levels and KRAS status were recorded. Categorical characteristics were compared using chi-squared test. Continuous characteristics were compared using Mann-Whitney U test. Log rank test was used to compare hazards for survival. In all comparisons, a two-sided P value <0.05 was considered statistically significant. Out of 201 patients, 170 patients with complete serum lipid profile were included in the analysis. In recto-sigmoid cancers there was a statistically significant association between high cholesterol:high-density lipoprotein (chol:HDL) ratio and KRAS mutation (OR 2.69, 95% CI 1.1–6.4, p = 0,02). In non recto-sigmoid cancers, high cholesterol was associated with KRAS WT (OR 0.39, CI 0.15–0.97, p = 0.04). In 22 patients with KRAS mutated recto-sigmoid cancer stage IV at diagnosis normal chol:HDL ratio was associated with a trend to better survival (p = 0.06).High chol:HDL ratiowas significantly associated with KRAS mutatedmetastatic recto-sigmoid cancers. A subgroup of mCRC patients with KRAS mutated recto-sigmoid cancer may benefit from optimal lipid lowering treatment. C1 [Tabuso, Maria] University Hospital Coventry and Warwickshire, Department of Gastroenterology, Clifford Bridge Road, CV2 2DX Coventry, UK. [Christian, Mark] Nottingham Trent University, School of Science and Technology, NG11 8NS Nottingham, UK. [Kimani, K. Peter] University of Warwick, Warwick Medical School, CV4 7AL Coventry, UK. [Gopalakrishnan, Kishore] University Hospitals of Coventry and Warwickshire NHS Trust, Department of Pathology, Clifford Bridge Road, CV2 2DX Coventry, UK. [Arasaradnam, Ramesh] University Hospital Coventry and Warwickshire, Department of Gastroenterology, Clifford Bridge Road, CV2 2DX Coventry, UK. RP Tabuso, M (reprint author), University Hospital Coventry and Warwickshire, Department of Gastroenterology, CV2 2DX Coventry, UK. EM Maria.Tabuso@uhcw.nhs.uk CR Patel A, Tripathi G, Gopalakrishnan K, Williams N, Arasaradnam RP, 2015, Field cancerisation in colorectal cancer: a new frontier or pastures past? World J Gastroenterol 21(13):3763–3772 Parekh N, Chandran U, Bandera EV, 2012, Obesity in cancer survival. Annu Rev Nutr 32:311–342 Esposito K, Chiodini P, Capuano A, Bellastella G, Maiorino MI, Rafaniello C, Panagiotakos DB, Giugliano D, 2013, Colorectal cancer association with metabolic syndrome and its components: a systematic review with meta-analysis. Endocrine. 44(3):634–647 Tabuso M, Homer-Vanniasinkam S, Adya R, Arasaradnam RP, 2017, Role of tissue microenvironment resident adipocytes in colon cancer. World J Gastroenterol 23(32):5829–5835 van Kruijsdijk RC, van derWall E, Visseren FL, 2009)Obesity and cancer: the role of dysfunctional adipose tissue. Cancer Epidemiol Biomark Prev 18(10):2569–2578 Siegel R, Ward E, Brawley O, Jemal A, 2011, Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 61(4):212–236 Shah MA, Renfro LA, Allegra CJ, Andre T, de Gramont A, Schmoll HJ et al, 2016, Impact of patient factors on recurrence risk and time dependency of Oxaliplatin benefit in patients with Colon Cancer: analysis from modern-era adjuvant studies in the adjuvant Colon Cancer end points, ACCENT, database. J Clin Oncol 34(8): 843–853 Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, Tebbutt NC, Simes RJ, Chalchal H, Shapiro JD, Robitaille S, Price TJ, Shepherd L, Au HJ, Langer C, Moore MJ, Zalcberg JR, 2008, K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 359(17):1757–1765 Guinney J, Dienstmann R, Wang X, de Reynies A, Schlicker A, Soneson C et al, 2015, The consensus molecular subtypes of colorectal cancer. Nat Med 21(11):1350–1356 Arnold D, Lueza B, Douillard JY, Peeters M, Lenz HJ, Venook A, Heinemann V, van Cutsem E, Pignon JP, Tabernero J, CervantesA, Ciardiello F, 2017, Prognostic and predictive value of primary tumour side in patients with RAS wild-type metastatic colorectal cancer treated with chemotherapy and EGFR directed antibodies in six randomized trials. Ann Oncol 28(8):1713–1729 Wang XY, Zhang R,Wang Z, Geng Y, Lin J,Ma K, Zuo JL, Lu L, Zhang JB, Zhu WW, Chen JH, 2019, Meta-analysis of the association between primary tumour location and prognosis after surgical resection of colorectal liver metastases. Br J Surg 106(13):1747– 1760 Charlton ME, Kahl AR, Greenbaum AA, Karlitz JJ, Lin C, Lynch CF, Chen VW, 2017, KRAS testing, tumor location, and survival in patients with stage IV colorectal Cancer: SEER 2010-2013. J Natl Compr Cancer Netw 15(12):1484–1493 Ogino S, Chan AT, Fuchs CS, Giovannucci E, 2011, Molecular pathological epidemiology of colorectal neoplasia: an emerging transdisciplinary and interdisciplinary field. Gut. 60(3):397–411 Brandstedt J, Wangefjord S, Nodin B, Eberhard J, Sundstrom M, Manjer J et al, 2014, Associations of anthropometric factors with KRAS and BRAF mutation status of primary colorectal cancer in men and women: a cohort study. PLoS One 9(2):e98964 Slattery ML, Curtin K, Wolff RK, Herrick JS, Caan BJ, Samowitz W, 2010, Diet, physical activity, and body size associations with rectal tumor mutations and epigenetic changes. Cancer Causes Control 21(8):1237–1245 Yilmaz A,Mohamed N, Patterson KA, Tang Y, Shilo K, Villalona- Calero MA, Davis M, Zhou XP, Frankel W, Otterson G, Zhao W, 2014, Clinical and metabolic parameters in non-small cell lung carcinoma and colorectal cancer patients with and without KRAS mutations. Int J Environ Res Public Health 11(9):8645–8660 Slattery ML, Anderson K, Curtin K, Ma K, Schaffer D, Edwards S et al, 2001, Lifestyle factors and Ki-ras mutations in colon cancer tumors. Mutat Res 483(1–2):73–81 Myte R, Gylling B, Haggstrom J, Haggstrom C, Zingmark C, Lofgren Burstrom A et al, 2019, Metabolic factors and the risk of colorectal cancer byKRAS and BRAFmutation status. Int J Cancer 145:327–337 Yamauchi M, Morikawa T, Kuchiba A, Imamura Y, Qian ZR, Nishihara R, Liao X, Waldron L, Hoshida Y, Huttenhower C, Chan AT, Giovannucci E, Fuchs C, Ogino S, 2012, Assessment of colorectal cancer molecular features along bowel subsites challenges the conception of distinct dichotomy of proximal versus distal colorectum. Gut. 61(6):847–854 Loree JM, Pereira AAL, Lam M, Willauer AN, Raghav K, Dasari A, Morris VK, Advani S,Menter DG, Eng C, Shaw K, Broaddus R, Routbort MJ, Liu Y, Morris JS, Luthra R, Meric-Bernstam F, Overman MJ, Maru D, Kopetz S, 2018, Classifying colorectal Cancer by tumor location rather than sidedness highlights a continuum in mutation profiles and consensus molecular subtypes. Clin Cancer Res 24(5):1062–1072 Edge SB, Compton CC, 2010, The American joint committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol 17(6):1471–1474 Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet. 2004;363(9403):157–63 Alberti KG, Zimmet P, Shaw J, 2006, Metabolic syndrome–a new world-wide definition. A consensus statement from the international diabetes federation. Diabet Med 23(5):469–480 Kwak MS, Cha JM, Cho YH, Kim SH, Yoon JY, Jeon JW, Shin HP, Joo KR, Lee JI, 2018, Clinical predictors for KRAS codon 13 mutations in patients with colorectal Cancer. J Clin Gastroenterol 52(5):431–436 Gray RT, Loughrey MB, Bankhead P, Cardwell CR, McQuaid S, O'Neill RF, Arthur K, Bingham V,McGready C, Gavin AT, James JA, Hamilton PW, Salto-Tellez M, Murray LJ, Coleman HG, 2017, Statin use, candidate mevalonate pathway biomarkers, and colon cancer survival in a population-based cohort study. Br J Cancer 116(12):1652–1659 Voorneveld PW, Reimers MS, Bastiaannet E, Jacobs RJ, van Eijk R, Zanders MMJ et al, 2017, Statin use after diagnosis of colon cancer and patient survival. Gastroenterology 153(2):470–9.e4 Ng K, Ogino S, Meyerhardt JA, Chan JA, Chan AT, Niedzwiecki D, Hollis D, Saltz LB, Mayer RJ, Benson AB, Schaefer PL, Whittom R, Hantel A, Goldberg RM, Bertagnolli MM, Venook AP, Fuchs CS, 2011, Relationship between statin use and colon cancer recurrence and survival: results from CALGB 89803. J Natl Cancer Inst 103(20):1540–1551 Gray RT, Coleman HG, Hughes C, Murray LJ, Cardwell CR, 2016, Statin use and survival in colorectal cancer: results from a population-based cohort study and an updated systematic review and meta-analysis. Cancer Epidemiol 45:71–81 Li Y, He X, Ding Y, Chen H, Sun L, 2019, Statin uses and mortality in colorectal cancer patients: an updated systematic review and meta-analysis. Cancer Med 8(6):3305–3313 Mace AG, Gantt GA, Skacel M, Pai R, Hammel JP, Kalady MF, 2013, Statin therapy is associated with improved pathologic response to neoadjuvant chemoradiation in rectal cancer. Dis Colon Rectum 56(11):1217–1227 Armstrong D, Raissouni S, Price Hiller J, Mercer J, Powell E, MacLean A, Jiang M, Doll C, Goodwin R, Batuyong E, Zhou K, Monzon JG, Tang PA, Heng DY, Cheung WY, Vickers MM, 2015, Predictors of pathologic complete response after Neoadjuvant treatment for rectal Cancer: a multicenter study. Clin Colorectal Cancer 14(4):291–295 Katzke VA, Sookthai D, Johnson T, Kuhn T, Kaaks R, 2017, Blood lipids and lipoproteins in relation to incidence and mortality risks for CVD and cancer in the prospective EPIC-Heidelberg cohort. BMC Med 15(1):218 Eichholzer M, Stahelin HB, Gutzwiller F, Ludin E, Bernasconi F, 2000, Association of low plasma cholesterol with mortality for cancer at various sites in men: 17-y follow-up of the prospective Basel study. Am J Clin Nutr 71(2):569–574 Yang Y,Mauldin PD, Ebeling M, Hulsey TC, Liu B, Thomas MB, Camp ER, Esnaola NF, 2013, Effect of metabolic syndrome and its components on recurrence and survival in colon cancer patients. Cancer. 119(8):1512–1520 Winawer SJ, Flehinger BJ, Buchalter J, Herbert E, ShikeM, 1990, Declining serum cholesterol levels prior to diagnosis of colon cancer. A time-trend, case-control study. JAMA 263(15):2083–2085 Lee J, Lee I, Han B, Park JO, Jang J, Park C, Kang WK, 2011, Effect of simvastatin on cetuximab resistance in human colorectal cancer with KRAS mutations. J Natl Cancer Inst 103(8):674–688 Kodach LL, Jacobs RJ, Voorneveld PW, Wildenberg ME, Verspaget HW, van Wezel T, Morreau H, Hommes DW, Peppelenbosch MP, van den Brink GR, Hardwick JCH, 2011, Statins augment the chemosensitivity of colorectal cancer cells inducing epigenetic reprogramming and reducing colorectal cancer cell 'stemness' via the bone morphogenetic protein pathway. Gut. 60(11):1544–1553 Gabitova L, Restifo D, Gorin A,Manocha K, Handorf E, Yang DH, Cai KQ, Klein-Szanto AJ, Cunningham D, Kratz LE, Herman GE, Golemis EA, Astsaturov I, 2015, Endogenous sterol metabolites regulate growth of EGFR/KRAS-dependent tumors via LXR. Cell Rep 12(11):1927–1938 Sukhanova A, Gorin A, Serebriiskii IG, Gabitova L, Zheng H, Restifo D, Egleston BL, Cunningham D, Bagnyukova T, Liu H, Nikonova A, Adams GP, Zhou Y, Yang DH,Mehra R, Burtness B, Cai KQ, Klein-Szanto A, Kratz LE, Kelley RI, Weiner LM, Herman GE, Golemis EA, Astsaturov I, 2013, Targeting C4- demethylating genes in the cholesterol pathway sensitizes cancer cells to EGF receptor inhibitors via increased EGF receptor degradation. Cancer Discov 3(1):96–111 Krens LL, Simkens LH, Baas JM, Koomen ER, Gelderblom H, Punt CJ et al, 2014, Statin use is not associated with improved progression free survival in cetuximab treated KRAS mutant metastatic colorectal cancer patients: results from the CAIRO2 study. PLoS One 9(11):e112201 Gouw AM, Eberlin LS, Margulis K, Sullivan DK, Toal GG, Tong L, Zare RN, Felsher DW, 2017, Oncogene KRAS activates fatty acid synthase, resulting in specific ERK and lipid signatures associated with lung adenocarcinoma. Proc Natl Acad Sci U S A 114(17):4300–4305 Padanad MS, Konstantinidou G, Venkateswaran N, Melegari M, Rindhe S,MitscheM, Yang C, Batten K, Huffman KE, Liu J, Tang X, Rodriguez-Canales J,Kalhor N, Shay JW, Minna JD, McDonald J, Wistuba II, DeBerardinis RJ, Scaglioni PP, 2016, Fatty acid oxidation mediated by acyl-CoA Synthetase long chain 3 is required for mutant KRAS lung tumorigenesis. Cell Rep 16(6): 1614–1628 Cox AD, Der CJ, 1997, Farnesyltransferase inhibitors and cancer treatment: targeting simply Ras? Biochim Biophys Acta 1333(1): F51–F71 Pecot CV, Wu SY, Bellister S, Filant J, Rupaimoole R, Hisamatsu T, Bhattacharya R, Maharaj A, Azam S, Rodriguez-Aguayo C, Nagaraja AS, Morelli MP, Gharpure KM, Waugh TA, Gonzalez- Villasana V, Zand B, Dalton HJ, Kopetz S, Lopez-Berestein G, Ellis LM, Sood AK, 2014, Therapeutic silencing of KRAS using systemically delivered siRNAs. Mol Cancer Ther 13(12):2876– 2885 Plowman SJ, Muncke C, Parton RG, Hancock JF, 2005, H-ras, Kras, and inner plasma membrane raft proteins operate in nanoclusters with differential dependence on the actin cytoskeleton. Proc Natl Acad Sci U S A 102(43):15500–15505 Zhou Y, Liang H, Rodkey T, Ariotti N, Parton RG, Hancock JF, 2014, Signal integration by lipid-mediated spatial cross talk between Ras nanoclusters. Mol Cell Biol 34(5):862–876 Prior IA,Hancock JF, 2001, Compartmentalization of Ras proteins. J Cell Sci 114(Pt 9):1603–1608 Zhang XF, Settleman J, Kyriakis JM, Takeuchi-Suzuki E, Elledge SJ, Marshall MS, Bruder JT, Rapp UR, Avruch J, 1993, Normal and oncogenic p21ras proteins bind to the amino-terminal regulatory domain of c-Raf-1. Nature. 364(6435):308–313 Rodriguez-Viciana P, Warne PH, Dhand R, Vanhaesebroeck B, Gout I, Fry MJ, Waterfield MD, Downward J, 1994, Phosphatidylinositol-3-OH kinase as a direct target of Ras. Nature. 370(6490):527–532 Pavlova NN, Thompson CB, 2016, The emerging hallmarks of Cancer metabolism. Cell Metab 23(1):27–47 DeBerardinis RJ, Chandel NS, 2016, Fundamentals of cancer metabolism. Sci Adv 2(5):e1600200 Kerr EM, Gaude E, Turrell FK, Frezza C, Martins CP, 2016, Mutant Kras copy number defines metabolic reprogramming and therapeutic susceptibilities. Nature. 531(7592):110–113 Kimmelman AC, 2015, Metabolic dependencies in RAS-driven cancers. Clin Cancer Res 21(8):1828–1834 Charitou T, Srihari S, Lynn MA, Jarboui MA, Fasterius E, Moldovan M, Shirasawa S, Tsunoda T, Ueffing M, Xie J, Xin J, Wang X, Proud CG, Boldt K, al-Khalili Szigyarto C, Kolch W, Lynn DJ, 2019, Transcriptional and metabolic rewiring of colorectal cancer cells expressing the oncogenic KRAS(G13D, mutation. Br J Cancer 121(1):37–50 Pupo E, Avanzato D, Middonti E, Bussolino F, Lanzetti L, 2019, KRAS-drivenmetabolic rewiring reveals novel actionable targets in Cancer. Front Oncol 9:848 Tape CJ, Ling S, Dimitriadi M, McMahon KM, Worboys JD, Leong HS et al, 2016, Oncogenic KRAS regulates tumor cell signaling via stromal reciprocation. Cell 165(4):910–920 Mayers JR, Torrence ME, Danai LV, Papagiannakopoulos T, Davidson SM, Bauer MR, Lau AN, Ji BW, Dixit PD, Hosios AM, Muir A, Chin CR, Freinkman E, Jacks T, Wolpin BM, Vitkup D, Vander Heiden MG, 2016, Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science. 353(6304):1161–1165 Weeber F, van de Wetering M, Hoogstraat M, Dijkstra KK, Krijgsman O, Kuilman T, Gadellaa-van Hooijdonk CGM, van der Velden DL, Peeper DS, Cuppen EPJG, Vries RG, Clevers H, Voest EE, 2015, Preserved genetic diversity in organoids cultured from biopsies of human colorectal cancer metastases. Proc Natl Acad Sci U S A 112(43):13308–13311 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2537 EP 2548 DI 10.1007/s12253-020-00850-y PG 12 ER PT J AU Molnar, Sz Beke, L Mehes, G Poka, R AF Molnar, Szabolcs Beke, Livia Mehes, Gabor Poka, Robert TI The Prognostic Value of PARP Expression in High-Grade Epithelial Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Ovarian cancer; PARP expression; Gynaecological oncology; Platinum-based chemotherapy; Progression-free survival ID Ovarian cancer; PARP expression; Gynaecological oncology; Platinum-based chemotherapy; Progression-free survival AB In an attempt to clarify the prognostic relevance of poly (ADP-ribose) polymerase (PARP) expression, we analysed the clinical data of 86 high-grade epithelial ovarian cancer (EOC) cases in which PARP immunohistochemistry results were available. Immunostaining to highlight PARP protein expression was performed using a Leica Bond MAX Immunostainer (Leica Microsystems, Wetzlar, Germany). We applied a rabbit polyclonal anti-PARP antibody (ab6079 330, Abcam, Cambridge, UK) for the specific reaction. The intensity and distribution of immunostaining were assessed by light microscopy (Leica DM2500 microscope, DFC 420 camera, and Leica Application Suite V3 software; Leica) and evaluated with a four-grade (0– 3+) system. The median progression-free survival (PFS) was generated for each semiquantitative group of PARP expression among chemotherapy-naive cases at the time of PARP immunohistochemistry. Eighty-six cases were chemotherapy-naive at the time of PARP immunohistochemistry, and 41 cases showed no PARP expression. Forty-five cases showed intermediate or high PARP expression. The median PFS among patients in the PARP-negative group was 16 months (interquartile range; IQR 10.7– 35.9 months), and the median PFS of patients in the PARP-positive group was 12 months (IQR 6.1–21.8 months). The difference was significant according to the log-rank test (p = 0.01). The median overall survival (OS) of patients in the PARP-negative group was 65 months (IQR 43.6–110.8 months), and the median OS of patients in the PARP-positive group was 52 months (IQR 36.9– 66.7 months). The difference was significant according to the log-rank test (p = 0.028). Multiple comparisons confirmed that PARP expression results in a significant difference in PFS and OS achieved by first-line Taxol-carboplatin chemotherapy. The lack of PARP expression assessed by immunohistochemistry may predict improved PFS in ovarian cancer patients after adjuvant platinum-based chemotherapy. C1 [Molnar, Szabolcs] University Medical School of Debrecen, Department of Gynecology and Obstetrics, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Beke, Livia] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of Pathology, Nagyerdei krt. 98, 4032 Debrecen, Hungary. [Poka, Robert] University Medical School of Debrecen, Department of Gynecology and Obstetrics, Nagyerdei krt. 98, 4032 Debrecen, Hungary. RP Molnar, Sz (reprint author), University Medical School of Debrecen, Department of Gynecology and Obstetrics, 4032 Debrecen, Hungary. EM szmolnar.md@gmail.com CR Ataseven B, GrimmC,Harter P, Prader S, Traut A, Heitz F, du Bois A, 2014, Prognostic value of lymph node ratio in patients with advanced epithelial ovarian cancer. Gynecol Oncol 135:435–440 Hacker NF, Rao A, 2016, Surgery for advanced epithelial ovarian cancer. Best Pract Res Clin Obstet Gynaecol 1–17 Wright AA, Bohlke K, Armstrong DK, Bookman MA, ClibyWA, Coleman RL, Dizon DS, Kash JJ,Meyer LA,Moore KN, Olawaiye AB, Oldham J, Salani R, Sparacio D, Tew WP, Vergote I, Edelson MI, 2016, Neoadjuvant chemotherapy for newly diagnosed, advanced ovarian cancer: Society of Gynecologic Oncology and American Society of clinical oncology clinical practice guideline. Gynecol Oncol 143:3–15 Sammartino P, Biacchi D, Di Giorgio A, 2012, Letters to the editor how to improve cytoreductive surgery for advanced ovarian cancer and talk about it in a common language. Gynecol Oncol 127:433– 436 Finch A, Shaw P, Rosen B,Murphy J, Narod SA, Colgan TJ, 2006, Clinical and pathologic findings of prophylactic salpingo- oophorectomies in 159 BRCA1 and BRCA2 carriers. GynecolOncol 100: 58–60 Marquard AM, Eklund AC, Joshi T, Krzystanek M, Favero F, Wang ZC, Richardson AL, Silver DP, Szallasi Z, Birkbak NJ, 2015, Pan-cancer analysis of genomic scar signatures associated with homologous recombination deficiency suggests novel indications for existing cancer drugs. Biomark Res 3:9 Wiggans AJ, Cass GKS, Bryant A et al., 2015, Poly(ADP-ribose, polymerase, PARP, inhibitors for the treatment of ovarian cancer, Review). Cochrane Libr, 5) Zhang F, Shi J, Bian Ch YX, 2015, Poly(ADP-ribose)mediates the BRCA2-dependent early DNA damage response. Cell Rep 13:678– 689 Bi FF, Li D, Yang Q, 2013, Promoter hypomethylation, especially around the E26 transformation-specific motif, and increased expression of poly, ADP-ribose, polymerase 1 in BRCA-mutated serous ovarian cancer. BMC Cancer 13:90 Marques M, BeauchampMC, Fleury H, Laskov I, Qiang S, Pelmus M, Provencher D, Mes-Masson AM, Gotlieb WH, Witcher M, 2015, Chemotherapy reduces PARP1 in cancers of the ovary: implications for future clinical trials involving PARP inhibitors. BMC Med 13:217 Godoy H, Mhawech-Fauceglia P, Beck A,Miller A, Lele S, Odunsi K, 2011, Expression of poly, adenosine Diphosphate-ribose, polymerase and p53 in epithelial ovarian Cancer and their role in prognosis and disease outcome. Int J Gynecol Pathol 30:139–144 Barnett JC, Bean SM, Nakayama JM, Kondoh E, Murphy SK, Berchuck A, 2010, High poly, adenosine Diphosphate-ribose, polymerase expression and poor survival in advanced-stage serous ovarian Cancer. Obstet Gynecol 115:49–54 Gan A, Green AR, Nolan CC, Martin S, Deen S, 2013, Poly, adenosine diphosphate-ribose, polymerase expression in BRCAproficient ovarian high-grade serous carcinoma; association with patient survival. Hum Pathol 44:1638–1647 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2549 EP 2555 DI 10.1007/s12253-020-00856-6 PG 7 ER PT J AU Koroknai, V Patel, V Szasz, I Adany, R Balazs, M AF Koroknai, Viktoria Patel, Vikas Szasz, Istvan Adany, Roza Balazs, Margit TI Gene Expression Signature of BRAF Inhibitor Resistant Melanoma Spheroids SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE 2D and 3D cell culture; Spheroids; Gene expression and BRAF inhibitor resistance; Melanoma cell lines ID 2D and 3D cell culture; Spheroids; Gene expression and BRAF inhibitor resistance; Melanoma cell lines AB In vitro cell cultures are frequently used to define the molecular background of drug resistance. The majority of currently available data have been obtained from 2D in vitro cultures, however, 3D cell culture systems (spheroids) are more likely to behave similarly to in vivo conditions. Our major aim was to compare the gene expression signature of 2D and 3D cultured BRAFV600E mutant melanoma cell lines. We successfully developed BRAF-drug resistant cell lines from paired primary/ metastatic melanoma cell lines in both 2D and 3D in vitro cultures. Using Affymetrix Human Gene 1.0 ST arrays, we determined the gene expression pattern of all cell lines. Our analysis revealed 1049 genes (562 upregulated and 487 downregulated) that were differentially expressed between drug-sensitive cells grown under different cell cultures. Pathway analysis showed that the differently expressed genes were mainly associated with the cell cycle, p53, and other cancer-related pathways. The number of upregulated genes (72 genes) was remarkably fewer when comparing the resistant adherent cells to cells that grow in 3D, and were associated with cell adhesion molecules and IGF1R signalling. Only 1% of the upregulated and 5.6% of the downregulated genes were commonly altered between the sensitive and the resistant spheroids. Interestingly, we found several genes (BNIP3, RING1 and ABHD4) with inverse expression signature between sensitive and resistant spheroids, which are involved in anoikis resistance and cell cycle regulation. In summary, our study highlights gene expression alterations that might help to understand the development of acquired resistance in melanoma cells in tumour tissue. C1 [Koroknai, Viktoria] Debreceni Egyetem, Nepegeszsegugyi Iskola, Kassai St 26/B, H-4028 Debrecen, Hungary. [Patel, Vikas] Debreceni Egyetem, Nepegeszsegugyi Iskola, Kassai St 26/B, H-4028 Debrecen, Hungary. [Szasz, Istvan] Debreceni Egyetem, Nepegeszsegugyi Iskola, Kassai St 26/B, H-4028 Debrecen, Hungary. [Adany, Roza] Debreceni Egyetem, Nepegeszsegugyi Iskola, Kassai St 26/B, H-4028 Debrecen, Hungary. [Balazs, Margit] Debreceni Egyetem, Nepegeszsegugyi Iskola, Kassai St 26/B, H-4028 Debrecen, Hungary. RP Balazs, M (reprint author), Debreceni Egyetem, Nepegeszsegugyi Iskola, H-4028 Debrecen, Hungary. EM balazs.margit@sph.unideb.hu CR Bandarchi B, Jabbari CA, Vedadi A, Navab R, 2013, Molecular biology of normal melanocytes and melanoma cells. J Clin Pathol 66(8):644–648 Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix- Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JWC, Leung SY, Yuen ST,Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA, 2002, Mutations of the BRAF gene in human cancer. Nature 417(6892):949–954 Seghers AC, Wilgenhof S, Lebbe C, Neyns B, 2012, Successful rechallenge in two patients with BRAF-V600-mutant melanoma who experienced previous progression during treatment with a selective BRAF inhibitor. Melanoma Res 22(6):466–472 Aasen SN, Parajuli H, Hoang T, Feng Z, Stokke K,Wang J, Roy K, Bjerkvig R, Knappskog S, Thorsen F, 2019, Effective treatment of metastatic melanoma by combining MAPK and PI3K signaling pathway inhibitors. Int J Mol Sci 20(17):4235 Wang J, Huang SK, Marzese DM, Hsu SC, Kawas NP, Chong KK, Long GV,Menzies AM, Scolyer RA, Izraely S, Sagi-Assif O,Witz IP, Hoon DSB, 2015, Epigenetic changes of EGFR have an important role in BRAF inhibitor-resistant cutaneousmelanomas. J Invest Dermatol 135(2):532–541 Molnar E, Garay T, Donia M, Baranyi M, Rittler D, Berger W, Timar J, Grusch M, Hegedus B, 2019, Long-term Vemurafenib exposure induced alterations of cell phenotypes in melanoma: increased cell migration and its association with EGFR expression. Int J Mol Sci 20(18):4484 Kenessey I, Kramer Z, Istvan L, Cserepes MT, Garay T, Hegedus B, Dobos J, Timar J, Tovari J, 2018, Inhibition of epidermal growth factor receptor improves antitumor efficacy of vemurafenib in BRAF-mutant human melanoma in preclinical model. Melanoma Res 28(6):536–546 Timar J, Vizkeleti L, Doma V, Barbai T, Raso E, 2016, Genetic progression of malignant melanoma. Cancer Metastasis Rev 35(1): 93–107 Van Allen EM, Wagle N, Sucker A et al, 2014, The genetic landscape of clinical resistance to RAF inhibition in metastatic melanoma. Cancer Discov 4(1):94–109 Kakadia S, Yarlagadda N, Awad R, Kundranda M, Niu J, Naraev B, Mina L, Dragovich T, Gimbel M, Mahmoud F, 2018, Mechanisms of resistance to BRAF and MEK inhibitors and clinical update of US Food and Drug Administration-approved targeted therapy in advanced melanoma. Onco Targets Ther 11:7095–7107 Kapalczynska M, Kolenda T, Przybyla W, Zajaczkowska M, Teresiak A, Filas V, Ibbs M, Blizniak R, Luczewski L, Lamperska K, 2018, 2D and 3D cell cultures - a comparison of different types of cancer cell cultures. ArchMed Sci 14(4):910–919 Antoni D, Burckel H, Josset E, Noel G, 2015, Three-dimensional cell culture: a breakthrough in vivo. Int J Mol Sci 16(3):5517–5527 Birgersdotter A, Sandberg R, Ernberg I, 2005, Gene expression perturbation in vitro-a growing case for three-dimensional, 3D, culture systems. Semin Cancer Biol 15(5):405–412 Fontoura JC, Viezzer C, dos Santos FG, Ligabue RA, Weinlich R, Puga RD, Antonow D, Severino P, Bonorino C, 2020, Comparison of 2D and 3D cell culture models for cell growth, gene expression and drug resistance. Mater Sci Eng C 107:110264 Ravi M, 2017, Applications of three-dimensional cell cultures in the early stages of drug discovery, focusing on gene expressions, drug metabolism, and susceptibility. Crit Rev Eukaryot Gene Expr 27(1):53–62 Souza AG, Silva IBB, Campos-Fernandez E, Barcelos LS, Souza JB, Marangoni K, Goulart LR, Alonso-Goulart V, 2018, Comparative assay of 2D and 3D cell culture models: proliferation, gene expression and anticancer drug response. Curr Pharm Des 24(15):1689–1694 Castiaux AD, Spence DM, Martin RS, 2019, Review of 3D cell culture with analysis in microfluidic systems. Anal Methods 11(33):4220–4232 Ghosh S, Spagnoli GC, Martin I, Ploegert S, Demougin P, Heberer M, Reschner A, 2005, Three-dimensional culture of melanoma cells profoundly affects gene expression profile: a high density oligonucleotide array study. J Cell Physiol 204(2):522–531 Duval K, Grover H, Han L-H, Mou Y, Pegoraro AF, Fredberg J, Chen Z, 2017, Modeling physiological events in 2D vs. 3D cell culture. Physiology 32(4):266–277 Furney SJ, Higgins DG, Ouzounis CA, Lopez-Bigas N, 2006, Structural and functional properties of genes involved in human cancer. BMC Genomics 7(1):3 Kumar HR, Zhong X, Hoelz DJ, Rescorla FJ, Hickey RJ, Malkas LH, Sandoval JA, 2008, Three-dimensional neuroblastoma cell culture: proteomic analysis between monolayer and multicellular tumor spheroids. Pediatr Surg Int 24(11):1229–1234 Tung YC, Hsiao AY, Allen SG, Torisawa YS, Ho M, Takayama S, 2011, High-throughput 3D spheroid culture and drug testing using a 384 hanging drop array. Analyst 136(3):473–478 Brancato V, Oliveira JM, Correlo VM, Reis RL, Kundu SC, 2020, Could 3D models of cancer enhance drug screening? Biomaterials 232:119744 Szasz I, Koroknai V, Kiss T, Vizkeleti L, Adany R, Balazs M, 2019, Molecular alterations associated with acquired resistance to BRAFV600E targeted therapy in melanoma cells. Melanoma Res 29(4):390–400 Vizkeleti L, Kiss T, Koroknai V, Ecsedi S, Papp O, Szasz I, Adany R, Balazs M, 2017, Altered integrin expression patterns shown by microarray in human cutaneous melanoma. Melanoma Res 27(3): 180–188 Livak KJ, Schmittgen TD, 2001, Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method. Methods 25(4):402–408 Gencoglu MF, Barney LE, Hall CL, Brooks EA, Schwartz AD, Corbett DC, Stevens KR, Peyton SR, 2018, Comparative study of multicellular tumor spheroid formation methods and implications for drug screening. ACS Biomater Sci Eng 4(2):410–420 Lee MA, Bergdorf KN, Phifer CJ, Jones CY, Byon SY, Sawyer LM, Bauer JA, Weiss VL, 2019, Novel three dimensional cultures provide insights into thyroid cancer behavior. Endocr Relat Cancer 27(2):111–121 Ryan SL, Baird AM, Vaz G, Urquhart AJ, Senge M, Richard DJ, O'Byrne KJ, Davies AM, 2016, Drug discovery approaches utilizing three-dimensional cell culture. Assay Drug Dev Technol 14(1): 19–28 Breslin S, O'Driscoll L, 2016, The relevance of using 3D cell cultures, in addition to 2D monolayer cultures, when evaluating breast cancer drug sensitivity and resistance. Oncotarget 7(29):45745– 45756 Ryabaya O, Prokofieva A, Akasov R, Khochenkov D, Emelyanova M, Burov S, Markvicheva E, Inshakov A, Stepanova E, 2019, Metformin increases antitumor activity of MEK inhibitor binimetinib in 2D and 3D models of human metastatic melanoma cells. Biomed Pharmacother 109:2548–2560 Ahn JH, Hwang SH, Cho HS, Lee M, 2019, Differential gene expression common to acquired and intrinsic resistance to BRAF inhibitor revealed by RNA-seq analysis. Biomol Ther 27(3):302– 310 Qiu T, Wang H, Wang Y, Zhang Y, Hui Q, Tao K, 2015, Identification of genes associated with melanoma metastasis. Kaohsiung J Med Sci 31(11):553–561 Lim SY, Yuzhalin AE, Gordon-Weeks AN, Muschel RJ, 2016, Targeting the CCL2-CCR2 signaling axis in cancer metastasis. Oncotarget 7(19):28697–28710 Schmit TL, Zhong W, Setaluri V, Spiegelman VS, Ahmad N, 2009, Targeted depletion of polo-like kinase, Plk, 1 through lentiviral shrna or a small-molecule inhibitor causes mitotic catastrophe and induction of apoptosis in human melanoma cells. J Invest Dermatol 129(12):2843–2853 KazanietzMG, CalocaMJ, 2017, The Rac GTPase in Cancer: from old concepts to new paradigms. Cancer Res 77(20):5445–5451 Eckert RL, Adhikary G, Young CA, Jans R, Crish JF, XuW, Rorke EA, 2013, AP1 transcription factors in epidermal differentiation and skin Cancer. J Skin Cancer 2013:537028 Bhandari A, Xia E, Zhou Y, Guan Y, Xiang J, Kong L, Wang Y, Yang F, Wang O, Zhang X, 2018, ITGA7 functions as a tumor suppressor and regulates migration and invasion in breast cancer. Cancer Manag Res 10:969–976 Argast GM, Croy CH, Couts KL, Zhang Z, Litman E, Chan DC, Ahn NG, 2009, Plexin B1 is repressed by oncogenic B-Raf signaling and functions as a tumor suppressor in melanoma cells. Oncogene 28(30):2697–2709 Denk AE, Braig S, Schubert T, Boserhof AK, 2011, Slit3 inhibits activator protein 1-mediated migration of malignant melanoma cells. Int J Mol Med 28(5):721–726 Dave B, Granados-Principal S, Zhu R, Benz S, Rabizadeh S, Soon- Shiong P, Yu KD, Shao Z, Li X, Gilcrease M, Lai Z, Chen Y, Huang THM, Shen H, Liu X, Ferrari M, Zhan M, Wong STC, Kumaraswami M, Mittal V, Chen X, Gross SS, Chang JC, 2014, Targeting RPL39 and MLF2 reduces tumor initiation and metastasis in breast cancer by inhibiting nitric oxide synthase signaling. Proc Natl Acad Sci U S A 111(24):8838–8843 Cheng WC, Tsui YC, Ragusa S, Koelzer VH, Mina M, Franco F, Laubli H, Tschumi B, Speiser D, Romero P, Zippelius A, Petrova TV, Mertz K, Ciriello G, Ho PC, 2019, Uncoupling protein 2 reprograms the tumor microenvironment to support the anti-tumor immune cycle. Nat Immunol 20(2):206–217 Wang Y, Hu S, Gabisi AM, Er JAV, Pope A, Burstein G, Schardon CL, Cardounel AJ, Ekmekcioglu S, Fast W, 2014, Developing an irreversible inhibitor of human DDAH-1, an enzyme upregulated in melanoma. ChemMedChem 9(4):792–797 Arimura Y, Ikura M, Fujita R, Noda M, Kobayashi W, Horikoshi N, Sun J, Shi L, Kusakabe M, Harata M, Ohkawa Y, Tashiro S, Kimura H, Ikura T, Kurumizaka H, 2018, Cancer-associated mutations of histones H2B, H3.1 and H2A.Z.1 affect the structure and stability of the nucleosome. Nucleic Acids Res 46(19):10007– 10018 Xin H, Lei M, Zhang Z, Li J, Zhang H, Luo X, Wang A, Deng F, 2018, Suppression of IGF1R in melanoma cells by an adenovirusmediated one-step knockdown system. Mol Ther - Nucleic Acids 12:861–871 Eriksson J, Le Joncour V, Nummela P, Jahkola T, Virolainen S, Laakkonen P, Saksela O, Holtta E, 2016, Gene expression analyses of primary melanomas reveal CTHRC1 as an important player in melanoma progression. Oncotarget 7:15065–15092 Igci YZ, Bozgeyik E, Borazan E, Pala E, Suner A, Ulasli M, Gurses SA, Yumrutas O, Balik AA, Igci M, 2015, Expression profiling of SCN8A and NDUFC2 genes in colorectal carcinoma. Exp Oncol 37(1):77–80 Huang JM, Hornyak TJ, 2015, Polycomb group proteins - epigenetic repressors with emerging roles in melanocytes and melanoma. Pigment Cell Melanoma Res 28(3):330–339 Chen J, Peng C, Lei L, Zhang J, Zeng W, Chen X, 2017, Nuclear envelope-distributed CD147 interacts with and inhibits the transcriptional function of RING1 and promotes melanoma cell motility. PLoS One 12(8):e0183689 Simpson CD, Hurren R, Kasimer D, MacLean N, Eberhard Y, Ketela T, Moffat J, Schimmer AD, 2012, A genome wide shRNA screen identifies α/β hydrolase domain containing 4, ABHD4, as a novel regulator of anoikis resistance. Apoptosis 17(7):666–678 Paoli P, Giannoni E, Chiarugi P, 2013, Anoikis molecular pathways and its role in cancer progression. Biochim Biophys Acta 1833(12):3481–3498 Kruiswijk F, Hasenfuss SC, Sivapatham R, Baar MP, Putavet D, Naipal KAT, Van Den Broek NJF, KruitW, Van Der Spek PJ, Van Gent DC, Brenkman AB, Campisi J, Burgering BMT, Hoeijmakers JHJ, De Keizer PLJ, 2016, Targeted inhibition of metastatic melanoma through interference with Pin1-FOXM1 signaling. Oncogene 35(17):2166–2177 Abourbih DA, Di Cesare S, Orellana ME, Antecka E, Martins C, Petruccelli LA, Burnier MN, 2010, Lysyl oxidase expression and inhibition in uveal melanoma. Melanoma Res 20(2):97–106 Romano E, Rufo N, Korf H, Mathieu C, Garg AD, Agostinis P, 2018, BNIP3modulates the interface between B16-F10melanoma cells and immune cells. Oncotarget 9:17631–17644 Maes H, Van Eygen S, Krysko DV, Vandenabeele P, Nys K, Rillaerts K, Garg AD, Verfaillie T, Agostinis P, 2014, BNIP3 supports melanoma cell migration and vasculogenic mimicry by orchestrating the actin cytoskeleton. Cell Death Dis 5(3):e1127 Sarkaria I, O-Charoenrat P, Talbot SG, Reddy PG, Ngai I, Maghami E, Patel KN, Lee B, Yonekawa Y, Dudas M, Kaufman A, Ryan R, Ghossein R, Rao PH, Stoffel A, Ramanathan Y, Singh B, 2006, Squamous cell carcinoma related oncogene/DCUN1D1 is highly conserved and activated by amplification in squamous cell carcinomas. Cancer Res 66(19):9437–9444 Sarogni P, Palumbo O, Servadio A, Astigiano S, D'Alessio B, Gatti V, Cukrov D, Baldari S, Pallotta MM, Aretini P, Dell'Orletta F, Soddu S, Carella M, Toietta G, Barbieri O, Fontanini G, Musio A, 2019, Overexpression of the cohesin-core subunit SMC1A contributes to colorectal cancer development. J Exp Clin Cancer Res 38(1):108 Imoto I, Yuki Y, Sonoda I, Ito T, Shimada Y, ImamuraM, Inazawa J, 2003, Identification of ZASC1 encoding a Kruppel-like zinc finger protein as a novel target for 3q26 amplification in esophageal squamous cell carcinomas. Cancer Res 63(18):5691–5696 Gervais FG, Singaraja R, Xanthoudakis S, Gutekunst CA, Leavitt BR,Metzler M, Hackam AS, Tam J, Vaillancourt JP, Houtzager V, Rasper DM, Roy S, Hayden MR, Nicholson DW, 2002, Recruitment and activation of caspase-8 by the Huntingtininteracting protein Hip-1 and a novel partner Hippi. Nat Cell Biol 4(2):95–105 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2557 EP 2566 DI 10.1007/s12253-020-00837-9 PG 10 ER PT J AU Aralica, G Sarec Ivelj, M Pacic, A Bakovic, J Milkovic Perisa, M Kristic, A Konjevoda, P AF Aralica, Gorana Sarec Ivelj, Martina Pacic, Arijana Bakovic, Josip Milkovic Perisa, Marija Kristic, Anteja Konjevoda, Pasko TI Prognostic Significance of Lacunarity in Preoperative Biopsy of Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; Preoperative biopsy; Intratumoral budding; Lacunarity; Prognosis; Recursive partitioning; Image analysis ID Colorectal cancer; Preoperative biopsy; Intratumoral budding; Lacunarity; Prognosis; Recursive partitioning; Image analysis AB The quantity and quality of preoperative material in colorectal cancer is often limiting factor in determination of risk factors and therapy planning. The most important negative prognostic factors are intravascular and perineural invasion, as well as tumor budding. Usually, the only parameter available in preoperative biopsy is tumor budding. However, the growing body of evidence suggests that cancer differentiation based on the poorly differentiated clusters has better prognostic value. The limiting factor in applying of these new parameters is reproducible, simple, cheap and fast method of their determination. In this paper we investigated the prognostic value of lacunarity, determined in preoperative biopsy. Lacunarity is a measure of spatial heterogeneity (inhomogeneity) in an image. It quantifies how objects fill the space, and enables analysis of gaps distribution, homogeneity of gaps, and presence of structures. It was shown that lacunarity and the total number of buds could be combined in a model which clearly divides colorectal cancer patients in low, medium and high risk subgroups. The paper also points out that the quantitative numerical methods are superior to semiquantitative methods, and that individual methods should be combined using algorithms to obtain a more accurate prediction. Because the study described is designed as a pilot study, verification is needed on a larger sample of patients from independent researchers. C1 [Aralica, Gorana] University of Zagreb, School of MedicineZagreb, Croatia. [Sarec Ivelj, Martina] University of Zagreb, School of MedicineZagreb, Croatia. [Pacic, Arijana] University Hospital DubravaZagreb, Croatia. [Bakovic, Josip] University Hospital DubravaZagreb, Croatia. [Milkovic Perisa, Marija] University of Zagreb, School of MedicineZagreb, Croatia. [Kristic, Anteja] University of Zagreb, School of MedicineZagreb, Croatia. [Konjevoda, Pasko] Rudjer Boskovic InstituteZagreb, Croatia. RP Aralica, G (reprint author), University of Zagreb, School of Medicine, Zagreb, Croatia. EM garalica@kbd.hr CR Jass JR, Morson BC, 1987, Reporting colorectal cancer. J Clin Pathol 40:1016–1023 Morodomi T, Isomoto H, Shirouzu K, Kakegawa K, Irie K, Morimatsu M(1989, An index estimating the probability of lymph node metastasis in rectal cancers. Cancer 63:539–543 Hase K, Shatney C, Johnson D et al, 1993, Prognostic value of tumor "budding" in patients with colorectal cancer. Dis Colon Rectum 36:627–635. , DOI 10.1007/bf02238588 Prall F, 2007, Tumor budding incolorectal carcinoma. Histopathology 50:151–162. , DOI 10.1111/j.1365-2559. 2006.02551.x Dawson H, Lugli A, 2015, Molecular and pathogenetic aspects of tumor budding in colorectal cancer. Front Med, Lausanne, 2:11. , DOI 10.3389/fmed.2015.00011 Zlobec I, Lugli A, 2010, Epithelial mesenchymal transition and tumor budding in aggressive colorectal cancer: tumor budding as oncotarget. Oncotarget 1:651–661. , DOI 10.18632/ oncotarget.199 Lugli A, Karamitopolou E, Zlobec I, 2012, Tumor budding: a promising parameter in colorectal cancer. Br J Cancer 106:1713– 1717. , DOI 10.1038/bjc.2012.127 Puppa G, Senore C, Sheahan K et al, 2012, Diagnostic reproducibility of tumour budding in colorectal cancer: a multicentre, multinational study using virtual microscopy. Histopathology 61:562– 575. , DOI 10.1111/j.1365-2559.2012.04270.x Wang LM, Kevans D, Mulcahy H et al, 2009, Tumor budding is a strong and reproducible prognostic marker in T3N0 colorectal cancer. Am J Surg Pathol 33:134–141. , DOI 10.1097/PAS. 0b013e318184cd55 Ueno H, Murphy J, Jass JR et al, 2002, Tumour 'budding' as an index to estimate the potential of aggressiveness in rectal cancer. Histopathology 40(2):127–132. , DOI 10.1046/j.1365- 2559.2002.01324.x Nakamura T, Mitomi H, Kikuchi S et al, 2005, Evaluation of the usefulness of tumor budding on the prediction of metastasis to the lung and liver after curative excision of colorectal cancer. Hepatogastroenterology 52:1432–1435 Ueno H, Kajiwara Y, Shimazaki H et al, 2012, New criteria for histologic grading of colorectal cancer. Am J Surg Pathol 36:193– 201. , DOI 10.1097/PAS.0b013e318235edee Horcic M, Koelzer VH, Karamitopoulou E et al, 2012, Tumor budding score based on 10 high-power fields is a promising basis for a standardized prognostic scoring system in stage II colorectal cancer. Hum Pathol 44:697–705. , DOI 10.1016/j. humpath.2012.07.026 Koelzer VH, Zlobec I, Berger MD et al, 2015, Tumor budding in colorectal cancer revisited: results of a multicenter interobserver study. Virchows Arch 466:485–493. , DOI 10.1007/ s00428-015-1740-9 Koelzer VH, Zlobec I, Lugli A, 2016, Tumor budding in colorectal cancer–ready for diagnostic practice? Hum Pathol 47:4–19. https:// doi.org/10.1016/j.humpath.2015.08.007 van Wyk HC, Park J, Roxburgh C et al, 2015, The role of tumour budding in predicting survival in patients with primary operable colorectal cancer: a systematic review. Cancer Treat Rev 41:151– 159,. , DOI 10.1016/j.ctrv.2014.12.007 Okamura T, Shimada Y, Nogami H et al, 2016, Tumor budding detection by immunohistochemical staining is not superior tohematoxylin and eosin staining for predicting lymph node metastasis in pT1 colorectal cancer. Dis Colon Rectum 59:396–402. , DOI 10.1097/DCR.0000000000000567 Eriksen AC, Andersen JB, Lindebjerg J et al, 2018, Does heterogeneity matter in the estimation of tumour budding and tumour stroma ratio in colon cancer? Diagn Pathol 13:20. , DOI 10.1186/s13000-018-0697-9 Karamitopoulou E, Zlobec I, Kolzer V et al, 2013, Proposal for a 10-high-power-fields scoring method for the assessment of tumor budding in colorectal cancer. Mod Pathol 26:295–301. https://doi. org/10.1038/modpathol.2012.155 Martin B, Schafer E, Jakubowicz E et al, 2018, Level of interobserver variability estimation as a valuable tool: assessment of tumour budding in colon cancer. Histopathology 73:864–868. , DOI 10.1111/his.13698 Martin B, Schafer E, Jakubowicz E et al, 2018, Interobserver variability in the H&E-based assessment of tumor budding in pT3/4 colon cancer: does it affect the prognostic relevance. Virchows Arch 473:189–197. , DOI 10.1007/s00428-018-2341-1 Lugli A, Kirsch R, Ajioka Y et al, 2017, Recommendations for reporting tumor budding in colorectal cancer based on the International Tumor Budding Consensus Conference, ITBCC, 2016. Mod Pathol 30:1299–1311. , DOI 10.1038/ modpathol.2017.46 Rogers AC, Gibbons D, Hanly AM et al, 2013, Prognostic significance of tumor budding in rectal cancer biopsies before neoadjuvant therapy. Mod Pathol 27:156–162. , DOI 10.1038/ modpathol.2013.124 Jessup JM, Goldberg RM, Asare EA et al, 2018, Colon and rectum. In: Edge SB, Greene LF, Schilsky RI, eds, AJCC cancer staging manual, 8th edn. Springer, Berlin, pp 251–294 Betge J, Langner C, 2011, Vascular invasion, perineural invasion, and tumour budding: predictors of outcome in colorectal cancer. Acta Gastroenterol Bel:516–529 Beaton C, Twine CP, Williams GL, Radcliffe AG, 2013, Systematic review and meta-analysis of histopathological factors influencing the risk of lymph node metastasis in early colorectal cancer. Colorectal Dis 15:788–797. , DOI 10.1111/codi. 12129 Poeschl EM, Pollheimer MJ, Kornprat P et al, 2010, Perineural invasion: correlation with aggressive phenotype and independent prognostic variable in both colon and rectum cancer. J Clin Oncol 28:e358–e362. , DOI 10.1200/JCO.2009.27.3581 Ueno H, Shirouzu K, Eishi Yet al, 2013, Characterization of perineural invasion as a component of colorectal cancer staging. Am J Surg Pathol 37:1542–1549. , DOI 10.1097/PAS. 0b013e318297ef6e Zlobec I, HadrichM, Dawson H et al, 2014, Intratumoural budding, ITB, in preoperative biopsies predicts the presence of lymph node and distant metastases in colon and rectal cancer patients. Br J Cancer 110:1008–1013. , DOI 10.1038/bjc.2013.797 Koelzer VH, Lugli A, Dawson H et al, 2014, CD8/CD45RO T-cell infiltration in endoscopic biopsies of colorectal cancer predicts nodal metastasis and survival. J Transl Med 12:81. , DOI 10. 1186/1479-5876-12-81 Giger OT, Comtesse SC, Lugli A et al, 2012, Intra-tumoral budding in preoperative biopsy specimens predicts lymph node and distant metastasis in patients with colorectal cancer. Mod Pathol 25:1048– 1053. , DOI 10.1038/modpathol.2012.56 Barresi V, Bonetti LR, Ieni A et al, 2014, Histologic grading based on counting poorly differentiated clusters in preoperative biopsy predicts nodal involvement and pTNM stage in colorectal cancer patients. Hum Pathol 45:268–275,. , DOI 10.1016/j. humpath.2013.07.046 Barresi V, Reggiani Bonetti L, Ieni A et al, 2016, Histologic prognostic markers in stage IIA colorectal cancer: a comparative study. Scand J Gastroenterol 51:314–320. , DOI 10.3109/ 00365521.2015.1084646 Konishi T, Shimada Y, Lee LH et al, 2018, Poorly differentiated clusters predict colon cancer recurrence: an In-depth comparative analysis of invasive-front prognostic markers.AmJ Surg Pathol 42: 705–714,. , DOI 10.1097/PAS.0000000000001059 Karperien A, Charles Sturt University, Australia/Canada, FracLac for ImageJ, V. 2.5, Available at: https://www.researchgate.net/ publication/258341589_FracLac_for_ImageJ. Accessed 19 Dec 2019 Valous NA, Xiong W, Halama N et al, 2018, Multilacunarity as a spatial multiscale multi-mass morphometric of change in the mesoarchitecture of plant parenchyma tissue. Chaos 28:093110. https:// doi.org/10.1063/1.5047021 Mambetsariev I,Mirzapoiazova T, Lennon F et al, 2019, Small cell lung cancer therapeutic responses through fractal measurements: from radiology to mitochondrial biology. J Clin Med 8:1038. , DOI 10.3390/jcm8071038 Waliszewski P, 2017, The quantitative criteria based on the fractal dimensions, entropy, and lacunarity for the spatial distribution of cancer cell nuclei enable identification of low or high aggressive prostate carcinomas. Front Physiol 7:E34. , DOI 10.3389/ fphys.2016.00034 Cencini M, Cecconi F, Vulpiani A, 2010, Chaos: From Simple Models to Complex Systems. World Scientific, Hackensack Lennon FE, Cianci GC, Kanteti R et al, 2016, Unique fractal evaluation and therapeutic implications of mitochondrialmorphology in malignantmesothelioma. Sci Rep 6:24578. , DOI 10.1038/ srep24578 Esgiar AN, Naguib RMN, Sharif BC et al, 2002, Fractal analysis in the detection of colonic cancer images. IEEE Trans Inf Technol Biomed 6:54–58 Broeke J, Perez JMM, Pascau J, 2015, Image Processing with ImageJ. Packt Publishing, Birmingham R Core Team, 2018, R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna. https://www.R-project.org/ Rowe P, 2016, Essential Statistics for the Pharmaceutical sciences. Wiley, Chichester Therneau T, Atkinson B, 2018, rpart: Recursive Partitioning and Regression Trees. R package version 4.1–13. https://CRAN.Rproject. org/package=rpart Ulm K, Kriner M, Eberle S, Reck M, Hessler S, 2006, Statistical methods to identify predictive factors. In: Crowley J, Ankerst D, eds, Handbook of statistics in clinical oncology, 2nd edn. Chapman & Hall/CRC, Boca Raton, pp 335–345 Schumacher M, Hollander N, Schwarzer G, Sauerbrei W, 2006, Prognostic factor studies. In: Crowley J, Ankerst D, eds, Handbook of statistics in clinical oncology, 2nd edn. Chapman & Hall/CRC, Boca Raton, pp 289–333 Therneau T, 2015, A Package for Survival Analysis in S. version 2.38. https://CRAN.R-project.org/package=survival>. Swets M, Kuppen PJK, Blok EJ et al, 2018, Are pathologicalhighriskfeaturesinlocallyadvancedrectalcancer a usefulselectiontool for adjuvantchemotherapy? Eur J Cancer 89:1–8. , DOI 10. 1016/j.ejca.2017.11.006 Park JH, van Wyk H, Roxburgh CSD et al, 2017, Tumorinvasiveness, thelocalandsystemic environment andthebasisof staging system in colorectalcancer. British J Cancer 116:1444–1450. , DOI 10.1038/bjc.2017.108 Huang Y, Lou X, Zhu Yet al, 2019, Local environment in biopsy better predict the pathologicalresponse to neoadjuvant chemoradiotherapy in rectal cancer. Biosci Rep 39:BSR20190003. https://doi. org/10.1042/BSR20190003 Huh JW, Lee WY, Shin JK, Park YA, Cho YB, Kim HC, Yun SH, 2019, A novelhistologicgrading system based on lymphovascularinvasion, perineuralinvasion, and tumor buddingincolorectalcancer. J Cancer Res Clin Oncol 145:471– 477. , DOI 10.1007/s00432-018-2804 Wai Kwan Lee V, Chan KF.(2018, Tumor budding and poorlydifferentiated cluster in prognostication in Stage II Colon Cancer. Pathology - Research Practice 214 , DOI 10.1016/j.prp. 2017.12.019 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2567 EP 2576 DI 10.1007/s12253-020-00851-x PG 10 ER PT J AU Niu, D Li, L Yu, Y Zang, W Li, Z Zhou, L Jia, L Rao, G Gao, L Cheng, G Ji, K Lin, D AF Niu, Dongfeng Li, Lei Yu, Yang Zang, Wanchun Li, Zhongwu Zhou, Lixin Jia, Ling Rao, Guanhua Gao, Lianju Cheng, Gang Ji, Ke Lin, Dongmei TI Evaluation of Next Generation Sequencing for Detecting HER2 Copy Number in Breast and Gastric Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Next generation sequencing; HER2 amplification; Breast cancer; Gastric cancer; FISH/IHC ID Next generation sequencing; HER2 amplification; Breast cancer; Gastric cancer; FISH/IHC AB Amplicon-based next generation sequencing (NGS) approaches have been preferentially adopted by the clinical laboratories on the basis of a short turnaround time (TAT) and small DNA input needs. However, little work has been done to assess the amplicon-based NGS methods for copy number variation (CNV) detection in comparison with current standard methods like immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH). The correlation between NGS based CNV detection and the later standard methods has remained unexplored. We developed an amplicon-based panel to detect human epidermal receptor growth factor (HER2) amplification in formalin-fixed paraffin-embedded (FFPE) tumor tissue samples from 280 breast cancer and 50 gastric cancer patients. Assessment by IHC and FISH was conducted in parallel, and descriptive statistics were used to assess the concordance. The copy number detected by NGS was correlated with either the average HER2 copy number (signals/cell) (r = 0.844; p < 0.001) or the HER2/CEP17 ratio (r = 0.815; p < 0.001). We determined a cut-off value for NGS to categorize HER2 amplification status by using 151 HER2 non-amplified FFPE samples. In breast cancer patients, the cut-off value was 2.910, with 95.35%, 98.67% and 97.29% sensitivity, specificity and concordance, respectively. However, this cut-off value displayed low sensitivity in gastric cancer patients (64.71%), and the following macrodissection procedure was not effective for increasing sensitivity (57.14%). Evaluation of HER2 copy number with NGS in our study was comparable with IHC and FISH in breast cancer patients, but concordance in gastric cancer was only moderate. The greater discordance in gastric cancer may reflect the underlying biological mechanisms, and further study is warranted. NGS-based HER2 assessment may decrease the equivocal HER2 determinations in breast cancer patients assessed by FISH/IHC. C1 [Niu, Dongfeng] Peking University Cancer Hospital & Institute, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of PathologyBeijing, China. [Li, Lei] Beijing Novogene Bioinformatics Technology Co., LtdBeijing, China. [Yu, Yang] Beijing Novogene Bioinformatics Technology Co., LtdBeijing, China. [Zang, Wanchun] Beijing Novogene Bioinformatics Technology Co., LtdBeijing, China. [Li, Zhongwu] Peking University Cancer Hospital & Institute, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of PathologyBeijing, China. [Zhou, Lixin] Peking University Cancer Hospital & Institute, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of PathologyBeijing, China. [Jia, Ling] Peking University Cancer Hospital & Institute, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of PathologyBeijing, China. [Rao, Guanhua] Beijing Novogene Bioinformatics Technology Co., LtdBeijing, China. [Gao, Lianju] Beijing Novogene Bioinformatics Technology Co., LtdBeijing, China. [Cheng, Gang] Beijing Novogene Bioinformatics Technology Co., LtdBeijing, China. [Ji, Ke] Peking University Cancer Hospital & Institute, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal SurgeryBeijing, China. [Lin, Dongmei] Peking University Cancer Hospital & Institute, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of PathologyBeijing, China. RP Lin, D (reprint author), Peking University Cancer Hospital & Institute, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Beijing, China. EM Dongm_lin@126.com CR Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ Jr, Wu YL, Paz-Ares L, 2017, Lung cancer: current therapies and new targeted treatments. Lancet 389(10066):299–311. , DOI 10.1016/S0140-6736(16)30958-8 Rehm HL, Bale SJ, Bayrak-Toydemir P, Berg JS, Brown KK, Deignan JL, Friez MJ, Funke BH, Hegde MR, Lyon E, Working Group of the American College of Medical G, Genomics Laboratory Quality Assurance C, 2013, ACMG clinical laboratory standards for next-generation sequencing. Genet Med 15(9):733– 747. , DOI 10.1038/gim.2013.92 Hamblin A, Wordsworth S, Fermont JM, Page S, Kaur K, Camps C, Kaisaki P, Gupta A, Talbot D,MiddletonM, Henderson S, Cutts A, Vavoulis DV, Housby N, Tomlinson I, Taylor JC, Schuh A, 2017, Clinical applicability and cost of a 46-gene panel for genomic analysis of solid tumours: retrospective validation and prospective audit in the UK National Health Service. PLoS Med 14(2): e1002230. , DOI 10.1371/journal.pmed.1002230 Pfarr N, Penzel R, Endris V, Lier C, Flechtenmacher C, Volckmar AL, Kirchner M, Budczies J, Leichsenring J, Herpel E, Noske A, Weichert W, Schneeweiss A, Schirmacher P, Sinn HP, Stenzinger A, 2017, Targeted next-generation sequencing enables reliable detection of HER2, ERBB2, status in breast cancer and provides ancillary information of clinical relevance. Gene Chromosome Cancer 56(4):255–265. , DOI 10.1002/gcc.22431 Yamamoto T, Shimojima K, Ondo Y, Imai K, Chong PF, Kira R, Amemiya M, Saito A, Okamoto N, 2016, Challenges in detecting genomic copy number aberrations using next-generation sequencing data and the eXome hidden Markov model: a clinical exomefirst diagnostic approach. Hum Genome Var 3:16025. https://doi. org/10.1038/hgv.2016.25 Ellingford JM, Campbell C, Barton S, Bhaskar S, Gupta S, Taylor RL, Sergouniotis PI, Horn B, Lamb JA, Michaelides M, Webster AR, Newman WG, Panda B, Ramsden SC, Black GC, 2017, Validation of copy number variation analysis for next-generation sequencing diagnostics. Eur J Hum Genet 25(6):719–724. https:// doi.org/10.1038/ejhg.2017.42 Grasso C, Butler T, Rhodes K, Quist M, Neff TL, Moore S, Tomlins SA, Reinig E, Beadling C, Andersen M, Corless CL, 2015, Assessing copy number alterations in targeted, ampliconbased next-generation sequencing data. J Mol Diagn 17(1):53–63. , DOI 10.1016/j.jmoldx.2014.09.008 Van Cutsem E, Bang YJ, Feng-Yi F, Xu JM, Lee KW, Jiao SC, Chong JL, Lopez-Sanchez RI, Price T, Gladkov O, Stoss O, Hill J, Ng V, LehleM, ThomasM, Kiermaier A, Ruschoff J, 2015, HER2 screening data from ToGA: targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer 18(3):476–484. https:// doi.org/10.1007/s10120-014-0402-y OwensMA, Horten BC, Da Silva MM, 2004, HER2 amplification ratios by fluorescence in situ hybridization and correlation with immunohistochemistry in a cohort of 6556 breast cancer tissues. Clin Breast Cancer 5(1):63–69. , DOI 10.3816/cbc.2004. n.011 Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, Bilous M, Ellis IO, Fitzgibbons P, HannaW, Jenkins RB, Press MF, Spears PA, Vance GH, Viale G, McShane LM, Dowsett M, 2018, Human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/ College of American Pathologists Clinical Practice Guideline Focused Update. J Clin Oncol 36(20):2105–2122. , DOI 10.1200/JCO.2018.77.8738 Chakravarty D, Gao J, Phillips SM, Kundra R, Zhang H, Wang J, Rudolph JE, Yaeger R, Soumerai T, Nissan MH, Chang MT, Chandarlapaty S, Traina TA, Paik PK, Ho AL, Hantash FM, Grupe A, Baxi SS, Callahan MK, Snyder A, Chi P, Danila D, Gounder M, Harding JJ, Hellmann MD, Iyer G, Janjigian Y, Kaley T, Levine DA, Lowery M, Omuro A, Postow MA, Rathkopf D, Shoushtari AN, Shukla N, Voss M, Paraiso E, Zehir A, Berger MF, Taylor BS, Saltz LB, Riely GJ, Ladanyi M, Hyman DM, Baselga J, Sabbatini P, Solit DB, Schultz N, 2017, OncoKB: a precision oncology Knowledge Base. JCO precision oncology 2017. , DOI 10.1200/PO.17.00011 Bartley AN, Washington MK, Colasacco C, Ventura CB, Ismaila N, Benson AB 3rd, Carrato A, Gulley ML, Jain D, Kakar S, Mackay HJ, Streutker C, Tang L, Troxell M, Ajani JA, 2017, HER2 Testing and Clinical Decision Making in Gastroesophageal Adenocarcinoma: Guideline From the College of American Pathologists, American Society for Clinical Pathology, and the American Society of Clinical Oncology. J Clin Oncol 35(4):446– 464. , DOI 10.1200/JCO.2016.69.4836 Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JM, Bilous M, Fitzgibbons P, Hanna W, Jenkins RB, Mangu PB, Paik S, Perez EA, Press MF, Spears PA, Vance GH, Viale G, Hayes DF, American Society of Clinical O, College of American P, 2014, Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. Arch Pathol Lab Med 138(2):241–256. , DOI 10.5858/arpa.2013-0953-SA Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, Schnall-Levin M,White J, Sanford EM, An P, Sun J, Juhn F, Brennan K, Iwanik K, Maillet A, Buell J, White E, Zhao M, Balasubramanian S, Terzic S, Richards T, Banning V, Garcia L, Mahoney K, Zwirko Z, Donahue A, Beltran H, Mosquera JM, Rubin MA, Dogan S, Hedvat CV, Berger MF, Pusztai L, Lechner M, Boshoff C, Jarosz M, Vietz C, Parker A, Miller VA, Ross JS, Curran J, Cronin MT, Stephens PJ, Lipson D, Yelensky R, 2013, Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol 31(11):1023–1031. , DOI 10.1038/nbt.2696 Fehm T, Muller V, Aktas B, Janni W, Schneeweiss A, Stickeler E, Lattrich C, Lohberg CR, Solomayer E, Rack B, Riethdorf S, Klein C, Schindlbeck C, Brocker K, Kasimir-Bauer S, Wallwiener D, Pantel K, 2010, HER2 status of circulating tumor cells in patients with metastatic breast cancer: a prospective,multicenter trial. Breast Cancer Res Treat 124(2):403–412. , DOI 10.1007/s10549- 010-1163-x Gao J, Wang H, Zang W, Li B, Rao G, Li L, Yu Y, Li Z, Dong B, Lu Z, Jiang Z, Shen L, 2017, Circulating tumor DNA functions as an alternative for tissue to overcome tumor heterogeneity in advanced gastric cancer. Cancer Sci 108(9):1881–1887. https://doi. org/10.1111/cas.13314 Mishima Y, Matsusaka S, Chin K, Mikuniya M, Minowa S, Takayama T, Shibata H, Kuniyoshi R, Ogura M, Terui Y, Mizunuma N, Hatake K, 2017, Detection of HER2 amplification in circulating tumor cells ofHER2-negative gastric Cancer patients. Target Oncol 12(3):341–351. , DOI 10.1007/s11523-017- 0493-6 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2577 EP 2585 DI 10.1007/s12253-020-00844-w PG 9 ER PT J AU Choi, S Park, S Cho, AY Park, ChK Ha, YS AF Choi, Sangjoon Park, Sujin Cho, Ah Yoon Park, Cheol-Keun Ha, Yun Sang TI Clinical Significance of Trk Receptor Expression as a New Therapeutic Target in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hepatocellular carcinoma; Prognosis; NTRK; Trk; Target ID Hepatocellular carcinoma; Prognosis; NTRK; Trk; Target AB Oncogenic fusion of the tropomyosin receptor kinase (Trk) receptor family encoded by the NTRK gene has been found in several carcinomas. About ten targeted therapies have been developed and clinical trials are in progress. However, the results of studies on expression of the Trk receptor in HCC have not yet been published. Immunohistochemical staining was performed using anti- TrkA+B+C antibody (ab181560, Abcam) in 288 curatively resected primary HCC samples, and the correlation between Trk expression and NTRK copy number was assessed. Targeted next generation sequencing was performed in cases with Trk overexpression to detect NTRK fusion genes. Overexpression of Trk protein was observed in 21 (7.3%) of 288 cases. The Trk overexpression group showed a trend toward shorter recurrence-free survival (RFS) (p = 0.092) and overall survival (OS) (p = 0.079) than the low expression group, with frequent multicentric occurrence. Differences in RFS and OS were statistically significant in specific sub-populations including AJCC T1 stage HCCs, tumors less than 5 cm, patients without cirrhosis, tumors without vascular invasion, or Edmondson grades I and II. Trk expression was also an independent prognostic factor in both RFS and OS. Trk expression was not associated with copy number of each NTRK gene, and NTRK fusion was not detected in HCCs with Trk overexpression. Trk expression might play an important role in the development and progression of HCC, and emerging target therapy against the Trk protein could be applicable in patients with Trk-overexpressing HCC. C1 [Choi, Sangjoon] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of Pathology, 81 Irwon-ro, Gangnam-gu, 06351 Seoul, South Korea. [Park, Sujin] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of Pathology, 81 Irwon-ro, Gangnam-gu, 06351 Seoul, South Korea. [Cho, Ah Yoon] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of Pathology, 81 Irwon-ro, Gangnam-gu, 06351 Seoul, South Korea. [Park, Cheol-Keun] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of Pathology, 81 Irwon-ro, Gangnam-gu, 06351 Seoul, South Korea. [Ha, Yun Sang] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of Pathology, 81 Irwon-ro, Gangnam-gu, 06351 Seoul, South Korea. RP Ha, YS (reprint author), Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of Pathology, 06351 Seoul, South Korea. EM sangyun.ha@skku.edu CR AmatuA, Sartore-Bianchi A, Siena S, 2016, NTRKgene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open 1:e000023 Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, Washington MK, Gershenwald JE, Compton CC, Hess KR, Sullivan DC, Jessup JM, Brierley JD, Gaspar LE, Schilsky RL, Balch CM, Cancer AJCo, 2017, AJCC cancer staging manual, 8th edn. Springer International Publishing, Berlin Bruix J, Qin S,Merle P, Granito A, Huang Y-H, Bodoky G, Pracht M, Yokosuka O, Rosmorduc O, Breder V, Gerolami R, Masi G, Ross PJ, Song T, Bronowicki J-P, Ollivier-Hourmand I, Kudo M, Cheng A-L, Llovet JM, Finn RS, LeBerre M-A, Baumhauer A, Meinhardt G, Han G, 2017, Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment, RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 389:56–66 Camp RL, Dolled-Filhart M, Rimm DL, 2004, X-tile: a new bioinformatics tool for biomarker assessment and outcome-based cutpoint optimization. Clin Cancer Res 10:7252–7259 Cho YA, Chung JM, Ryu H, Kim EK, Cho BC, Yoon SO, 2019, Investigating Trk protein expression between Oropharyngeal and non-oropharyngeal squamous cell carcinoma: clinical implications and possible roles of human papillomavirus infection. Cancer Res Treat 51:1052–1063 Choi S, Chu J, Kim B, Ha SY, Kim ST, Lee J, Kang WK, Han H, Sohn I, Kim KM, 2019, Tumor heterogeneity index to detect human epidermal growth factor receptor 2 amplification by nextgeneration sequencing: a direct comparison study with immunohistochemistry. J Mol Diagn 21:612–622 Drilon A, Laetsch TW, Kummar S, DuBois SG, Lassen UN, Demetri GD, Nathenson M, Doebele RC, Farago AF, Pappo AS, Turpin B, Dowlati A, Brose MS, Mascarenhas L, Federman N, Ber l in J, El-Deiry WS, Baik C, Deeken J, Boni V, Nagasubramanian R, Taylor M, Rudzinski ER, Meric-Bernstam F, Sohal DPS, Ma PC, Raez LE, Hechtman JF, Benayed R, Ladanyi M, Tuch BB, Ebata K, Cruickshank S, Ku NC, Cox MC, Hawkins DS, Hong DS, Hyman DM, 2018, Efficacy of Larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med 378:731–739 Drilon A, Siena S, Ou SI, Patel M, Ahn MJ, Lee J, Bauer TM, Farago AF, Wheler JJ, Liu SV, Doebele R, Giannetta L, Cerea G, Marrapese G, Schirru M, Amatu A, Bencardino K, Palmeri L, Sartore-Bianchi A, Vanzulli A, Cresta S, Damian S, Duca M, Ardini E, Li G, Christiansen J, Kowalski K, Johnson AD, Patel R, Luo D, Chow-Maneval E, Hornby Z, Multani PS, Shaw AT, De Braud FG, 2017, Safety and antitumor activity of the multitargeted pan-TRK, ROS1, and ALK inhibitor Entrectinib: combined results from two phase I trials, ALKA-372-001 and STARTRK-1). Cancer Discov 7:400–409 Edmondson HA, Steiner PE, 1954, Primary carcinoma of the liver: a study of 100 cases among 48,900 necropsies. Cancer 7:462–503 El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, Kim T-Y, Choo S-P, Trojan J, Welling TH, Meyer T, Kang Y-K, YeoW, Chopra A, Anderson J, dela Cruz C, Lang L, Neely J, Tang H, Dastani HB, Melero I, 2017, Nivolumab in patients with advanced hepatocellular carcinoma, CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 389:2492–2502 Hechtman JF, Benayed R, Hyman DM, Drilon A, Zehir A, Frosina D, Arcila ME, Dogan S, Klimstra DS, Ladanyi M, Jungbluth AA, 2017, Pan-Trk immunohistochemistry is an efficient and reliable screen for the detection of NTRK fusions. Am J Surg Pathol 41: 1547–1551 Hyeon J, Ahn S, Park CK, 2013, CHD1L is a marker for poor prognosis of hepatocellular carcinoma after surgical resection. Korean Journal of Pathology 47:9–15 Imamura H, Matsuyama Y, Tanaka E, Ohkubo T, Hasegawa K, Miyagawa S, Sugawara Y, Minagawa M, Takayama T, Kawasaki S, Makuuchi M, 2003, Risk factors contributing to early and late phase intrahepatic recurrence of hepatocellular carcinoma after hepatectomy. J Hepatol 38:200–207 Kheder ES, Hong DS, 2018, Emerging targeted therapy for tumors with NTRK fusion proteins. Clin Cancer Res 24:5807–5814 KudoM, Finn RS, Qin S, Han K-H, Ikeda K, Piscaglia F, Baron A, Park J-W, Han G, Jassem J, Blanc JF, Vogel A, Komov D, Evans TRJ, Lopez C, Dutcus C, Guo M, Saito K, Kraljevic S, Tamai T, Ren M, Cheng A-L, 2018, Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet 391:1163–1173 Kumada T, Nakano S, Takeda I, Sugiyama K, Osada T, Kiriyama S, Sone Y, Toyoda H, Shimada S, Takahashi M, Sassa T, 1997, Patterns of recurrence after initial treatment in patients with small hepatocellular carcinoma. Hepatology 25:87–92 Llovet JM, Bru C, Bruix J, 1999, Prognosis of hepatocellular carcinoma: the BCLC staging classification. Semin Liver Dis 19:329– 338 Ma D, Wang Z, Yang L, Mu X, Wang Y, Zhao X, Li J, Lin D, 2016, Responses to crizotinib in patients with ALK-positive lung adenocarcinoma who tested immunohistochemistry, IHC)-positive and fluorescence in situ hybridization, FISH)-negative. Oncotarget 7:64410–64420 Okamura R, Boichard A, Kato S, Sicklick JK, Bazhenova L, Kurzrock R, 2018, Analysis of NTRK Alterations in Pan-Cancer Adult and Pediatric Malignancies: Implications for NTRKTargeted Therapeutics. JCO Precis Oncol 2018 Remmele W, Stegner HE, 1987, Recommendation for uniform definition of an immunoreactive score, IRS, for immunohistochemical estrogen receptor detection, ER-ICA, in breast cancer tissue. Pathologe 8:138–140 Rudzinski ER, Lockwood CM, Stohr BA, Vargas SO, Sheridan R, Black JO, Rajaram V, Laetsch TW, Davis JL, 2018, Pan-Trk immunohistochemistry identifies NTRK rearrangements in pediatric Mesenchymal tumors. Am J Surg Pathol 42:927–935 Sclabas GM, Fujioka S, Schmidt C, Li Z, Frederick WA, YangW, Yokoi K, Evans DB, Abbruzzese JL, Hess KR, ZhangW, Fidler IJ, Chiao PJ, 2005, Overexpression of tropomysin-related kinase B in metastatic human pancreatic cancer cells. Clin Cancer Res 11:440– 449 Shan C, Wei J, Hou R, Wu B, Yang Z, Wang L, Lei D, Yang X, 2016, Schwann cells promote EMT and the Schwann-like differentiation of salivary adenoid cystic carcinoma cells via the BDNF/ TrkB axis. Oncol Rep 35:427–435 Sun JM, Choi YL, Won JK, Hirsch FR, Ahn JS, Ahn MJ, Park K, 2012, A dramatic response to crizotinib in a non-small-cell lung cancer patient with IHC-positive and FISH-negativeALK. J Thorac Oncol 7:e36–e38 Vaishnavi A, Le AT, Doebele RC, 2015, TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov 5:25–34 Wang K, Lim HY, Shi S, Lee J, Deng S, Xie T, Zhu Z, Wang Y, Pocalyko D, Yang WJ, Rejto PA, Mao M, Park CK, Xu J, 2013, Genomic landscape of copy number aberrations enables the identification of oncogenic drivers in hepatocellular carcinoma. Hepatology 58:706–717 Xu CW, Wang WX, Chen YP, Chen Y, Liu W, Zhong LH, Chen FF, ZhuangW, Song ZB, Chen XH, Huang YJ, Guan YF, Yi X, Lv TF, Zhu WF, Lu JP, Wang XJ, Shi Y, Lin XD, Chen G, Song Y, 2018, Simultaneous VENTANA IHC and RT-PCR testing of ALK status in Chinese non-small cell lung cancer patients and response to crizotinib. J Transl Med 16:93 Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D, Verslype C, Zagonel V, Fartoux L, Vogel A, Sarker D, Verset G, Chan SL, Knox J, Daniele B, Webber AL, Ebbinghaus SW, Ma J, Siegel AB, Cheng A-L, Kudo M, Alistar A, Asselah J, Blanc J-F, Borbath I, Cannon T, Chung K, Cohn A, Cosgrove DP, Damjanov N, Gupta M, Karino Y, Karwal M, Kaubisch A, Kelley R, Van Laethem J-L, Larson T, Lee J, Li D, Manhas A, Manji GA, Numata K, Parsons B, Paulson AS, Pinto C, Ramirez R, Ratnam S, Rizell M, Rosmorduc O, Sada Y, Sasaki Y, Stal PI, Strasser S, Trojan J, Vaccaro G, Van Vlierberghe H, Weiss A, Weiss K-H, Yamashita T, 2018, Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib, KEYNOTE-224): a non-randomised, open-label phase 2 trial. The Lancet Oncology 19:940–952 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2587 EP 2595 DI 10.1007/s12253-020-00871-7 PG 9 ER PT J TI Novel CXCR4 Inhibitor CPZ1344 Inhibits the Proliferation, Migration and Angiogenesis of Glioblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CXCR4; CPZ1344; Migration; Glioblastoma ID CXCR4; CPZ1344; Migration; Glioblastoma AB Glioblastoma (GBM) are life-threatening tumors with a poor prognosis and low cure rates. GBMs are malignant brain tumors that develop from astrocytes. Most GBMs are not inherited and occur sporadically. GBM recurrence after standard treatment has led to the assessment of agents targeting the CXCR4 chemokine receptor as alternative drug target for much needed GBM therapeutics. In present study, a novel CXCR4 inhibitor modified with a picolinamide scaffold (CPZ1344) was designed and synthesized. Its anti-GBM function was then evaluated. Our results showed that CPZ1344 reduced the growth of GBM cells in a concentration dependent manner. The anti-GBM activity of CPZ1344 was due to alteration in GBM-cell morphology and apoptotic induction in GBM cells. CPZ1344 inhibited the migration and angiogenesis of U87 cells, led to cell cycle arrest in the G1 phase and inhibited CXCR4 signaling. These findings demonstrate the anticancer effects of CPZ1344 and its potential as a novel anti-GBM therapeutic. CR Omuro A,DeAngelisLM(2013)Glioblastoma and othermalignant gliomas: a clinical review. Jama 310, 17): 1842–1850 Delgado-Lopez PD, Corrales-Garcia EM, 2016, Survival in glioblastoma: a review on the impact of treatment modalities. Clin Transl Oncol 18, 11): 1062–1071 Fianco G, Contadini C, Ferri A, Cirotti C, Stagni V, Barila D, 2018, Caspase-8: A Novel Target to Overcome Resistance to Chemotherapy in Glioblastoma. Int J Mol Sci 19, 12): 3798 Perry JR, Laperriere N, O’Callaghan CJ, Brandes AA, Menten J, Phillips C, et al, 2017, Short-Course Radiation plus Temozolomide in Elderly Patients with Glioblastoma. N Engl J Med 376, 11): 1027–1037 Osuka S, Van Meir EG, 2017, Overcoming therapeutic resistance in glioblastoma: the way forward. J Clin Investig 127, 2): 415–426 Campos B, Olsen LR, Urup T, Poulsen HS, 2016, A comprehensive profile of recurrent glioblastoma. Oncogene 35, 45): 5819– 5825 Richardson PJ, 2016, CXCR4 and Glioblastoma. Anticancer Agents Med Chem 16, 1): 59–74 Stevenson CB, Ehtesham M, McMillan KM, Valadez JG, Edgeworth ML, Price RR, et al, 2008, CXCR4 expression is elevated in glioblastoma multiforme and correlates with an increase in intensity and extent of peritumoral T2-weighted magnetic resonance imaging signal abnormalities. Neurosurgery 63, 3): 560-9; discussion 569–570. Terasaki M, Sugita Y, Arakawa F, Okada Y, Ohshima K, Shigemori M, 2011, CXCL12/CXCR4 signaling in malignant brain tumors: a potential pharmacological therapeutic target. Brain Tumor Pathol 28, 2): 89–97 Ehtesham M, Mapara KY, Stevenson CB, Thompson RC, 2009, CXCR4 mediates the proliferation of glioblastoma progenitor cells. Cancer Letters 274, 2): 305–312 Gravina GL, Mancini A, Colapietro A, Vitale F, Vetuschi A, Pompili S, et al, 2017, The novel CXCR4 antagonist, PRX177561, reduces tumor cell proliferation and accelerates cancer stem cell differentiation in glioblastoma preclinical models. Tumour Biol 39, 6): 1010428317695528 Rios A, Hsu SH, Blanco A, Buryanek J, Day AL, McGuire MF, et al, 2016, Durable response of glioblastoma to adjuvant therapy consisting of temozolomide and a weekly dose of AMD3100, plerixafor), a CXCR4 inhibitor, together with lapatinib, metformin and niacinamide. Oncoscience 3, 5–6): 156–163 De Clercq E, 2019, Mozobil(R,, Plerixafor, AMD3100), 10 years after its approval by the US Food and Drug Administration. Antivir Chem Chemother 27: 2040206619829382 Hendrix CW, Collier AC, Lederman MM, Schols D, Pollard RB, Brown S, et al, 2004, Safety, pharmacokinetics, and antiviral activity of AMD3100, a selective CXCR4 receptor inhibitor, in HIV-1 infection. J Acquir Immune Defic Syndr 37, 2): 1253–1262 Liang Z, Zhan W, Zhu A, Yoon Y, Lin S, Sasaki M, et al, 2012, Development of a unique small molecule modulator of CXCR4. PloS one 7, 4): e34038 Liu Y, Zhou J, Pan JA, Mabiala P, Guo D, 2014, A novel approach to block HIV-1 coreceptor CXCR4 in non-toxic manner. Mol Biotechnol 56, 10): 890–902 Zhan W, Liang Z, Zhu A, Kurtkaya S, Shim H, Snyder JP et al, 2007, Discovery of small molecule CXCR4 antagonists. J Med Chem 50(23):5655–5664 Rubin JB, Kung AL, Klein RS, Chan JA, Sun Y, Schmidt K, et al, 2003, A small-molecule antagonist of CXCR4 inhibits intracranial growth of primary brain tumors. Proc Natl Acad Sci USA 100, 23): 13513–13518 Weathers SP, de Groot J, 2015, VEGF Manipulation in Glioblastoma. Oncology, Williston Park, 29, 10): 720–727 Barone A, Sengupta R, Warrington NM, Smith E, Wen PY, Brekken RA, et al, 2014, Combined VEGF and CXCR4 antagonism targets the GBM stem cell population and synergistically improves survival in an intracranial mouse model of glioblastoma. Oncotarget 5, 20): 9811–9822 Yamaguchi K, Sudo H, Imai K, 2019, Vascular endothelial growth factor signaling in VE-cadherin expression and tube-like formation by rheumatoid arthritic synovial fibroblast-like cells. Biochem Biophys Res Commun 508, 2): 405–409 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2597 EP 2604 DI 10.1007/s12253-020-00827-x PG 8 ER PT J AU Soenens, Ch Dekuyper, P De Coster, G Van Damme, N Van Eycken, E Quackels, Th Roumeguere, Th Van Cleynenbreugel, B Joniau, S Ameye, F AF Soenens, Charlotte Dekuyper, Peter De Coster, Greet Van Damme, Nancy Van Eycken, Elizabeth Quackels, Thierry Roumeguere, Thierry Van Cleynenbreugel, Ben Joniau, Steven Ameye, Filip TI Concordance Between Biopsy and Radical Prostatectomy Gleason Scores: Evaluation of Determinants in a Large-Scale Study of Patients Undergoing RARP in Belgium SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Prostate cancer; Gleason score; Concordance; Upgrading; Center size ID Prostate cancer; Gleason score; Concordance; Upgrading; Center size AB To determine whether Gleason scores were concordant between prostate biopsies (bGS) and the definitive resection specimen (pGS) excised with robot-assisted radical prostatectomy (RARP); to identify clinical and pathological factors that might predict upgrading; and to evaluate how upgrading affected outcome. Between 2009 and 2016, 25 Belgian centers participated in collecting prospective data for patients that underwent RARP. We analyzed the concordance rate between the bGS and the pGS in 8021 patients with kappa statistics, and we compared concordance rates from different centers. We assessed the effect of several clinical and pathological factors on the concordance rate with logistic regression analysis. The concordance rate for the entire population was 62.9%. Upgrading from bGS to pGS occurred in 27.3% of patients. The number of biopsies was significantly associated with concordance. Older age (>60 y), a higher clinical T stage (≥cT2), a higher PSA value at the time of biopsy (>10 ng/ml), and more time between the biopsy and the radical prostatectomy were significantly associated with a higher risk of upgrading. Positive margins and PSA relapse occurred more frequently in upgraded patients. Center size did not significantly affect the concordance rate (p = 0.40).This prospective, nationwide analysis demonstrated a Gleason score concordance rate of 62.9%. Upgrading was most frequently observed in the non-concordant group. We identified clinical and pathological factors associated with (non)-concordance. Upgrading was associated with a worse oncological outcome. Center volume was not associated with pathological accuracy. C1 [Soenens, Charlotte] AZ Maria Middelares, Department of UrologyGhent, Belgium. [Dekuyper, Peter] AZ Maria Middelares, Department of UrologyGhent, Belgium. [De Coster, Greet] Belgian Cancer RegistryBrussels, Belgium. [Van Damme, Nancy] Belgian Cancer RegistryBrussels, Belgium. [Van Eycken, Elizabeth] Belgian Cancer RegistryBrussels, Belgium. [Quackels, Thierry] Erasmus Hospital, Department of UrologyBrussels, Belgium. [Roumeguere, Thierry] Erasmus Hospital, Department of UrologyBrussels, Belgium. [Van Cleynenbreugel, Ben] University Hospital of Leuven, Department of UrologyLeuven, Belgium. [Joniau, Steven] University Hospital of Leuven, Department of UrologyLeuven, Belgium. [Ameye, Filip] AZ Maria Middelares, Department of UrologyGhent, Belgium. RP Soenens, Ch (reprint author), AZ Maria Middelares, Department of Urology, Ghent, Belgium. EM charlotte.soenens@azmmsj.be CR Gleason DF, 1966, Classification of prostate carcinomas. Cancer Chemother 50:125–128 Epstein JI, Allsbrook WC Jr, Amin MB, Egevad LL, 2005, The 2005 International Society of Urological Pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol 29(9):1228–1242 Epstein JI, Egevad L, Amin MB, Delahunt B, Srigley JR, Humphrey PA, 2014, The 2014 International Society of Urological Pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma: definition of grading patterns and proposal for a new grading system. Am J Surg Pathol 40(2):244– 252 Kvale R,Moller B,Wahiqvist R et al, 2008, Concordance between Gleason score of needle biopsies and radical prostatectomy specimens: a population-based study. BJUI 103:1647–1654 Rapiti E, Schaffar R, Iselin C et al, 2013, Importance and determinants of Gleason score undergrading on biopsy sample of prostate cancer in a population-based study. MBC Urol 13:19 Muntener M, Epstein JI, Henandez DJ et al, 2008, Prognostic significance of Gleason score discrepancies between needle biopsy and radical prostatectomy. Eur Urol 53:767–776 King CR, 2000, Patterns of prostate cancer biopsy grading: trends and clinical implications. Int J Cancer 90:305–311 Isariyawongse BK, Sun L, Banez L et al, 2008, Significant discrepancies between diagnostic and pathologic Gleason sums in prostate cancer: the predictive role of age and prostate-specific antigen. Urology 72:882–886 Joniau S, Spyrantis M, Birganti A et al, 2018, Gleason score 6 prostate cancer is not always harmless. Eur Urol Suppl 17(2): e242–e243 Albissini S, Joniau S, Quackels T et al, 2017, Current trends in patient enrolment for robotic-assisted laparoscopic prostatectomy in Belgium. Cancer 123(21):4139–4146 European association of urology: guideline on clinical diagnosis of prostate. https://uroweb.org/guideline/prostate-cancer/ McHugh, 2012, Interrater reliability: the kappa statistic. Biochem Med 22(3):276–282 Wong AT, AgarwalM, Navo EB, Schwartz D, Schreiber D, 2017, Concordance of gleason score on biopsy and after prostatectomy: a SEER databyse analysis. J Clin Oncol 33:50 Evans SM, Bandarage VP, Kronborg C, Earnest A, Millar J, Clouston D, 2016, Gleason group concordance between biopsy and radical prostatectomy specimen. A cohort study from prostate Cancer outcome registry – Victoria. Prostate Int 4:145–151 Cumming JA, Ritchie AWS, Goodman CM, McIntyre MA, ChisholmGF, 1990, De-differentiation with time in prostate cancer and the influence of treatment on the course of the disease. BJI 65: 271–274 Fossati N, RossiMS, Cucchiara V et al, 2017, Evaluating the effect of time from prostate cancer diagnosis to radical prostatectomy on cancer control: Can surgery be postponed safely? Urol Oncol 35(4): 150.e9–150.15 Gupta N, Bivalacqua TJ, Han M, Gorin MA, Challacombe BJ, Partin AW,MamawalaMK, 2019, Evaluating the impact of length of time from diagnosis to surgery in patients with unfavourable intermediate-risk to very-high-risk clinically localised prostate cancer. BJU Int 124:268–274 Meunier ME, Neuzillet Y, Radulescu C, Cherbonnier C, Herve JM, RouanneM,Molinie V, Lebret T, 2018, Does the delay from prostate biopsy to radical prostatectomy influence the risk of biochemical recurrence? Prog Urol 28(10):475–481 Wallis CJD, Novara G, Marandino L, Bex A, Kamat AM, Karnes RJ, Morgan TM, Mottet N, Gillessen S, Bossi A, Roupret M, Powles T, Necchi A, Catto JWF, Klaassen Z, 2020, Risks from deferring treatment for genitourinary cancers: a collaborative review to aid triage and management during the COVID-19 pandemic. Eur Urol 78:29–42 Ahmed HU, Bosaily AE, Brown LC et al, 2017, Diagnostic accuracy of multi-parametric MRI and TRUS biopsy in prostate cancer, PROMIS): a paired validating confirmatory study. Lancet 389: 815–822 Kasivisvanathan V, Rannikko AS, Borghi M, Panebianco V, Mynderse LA, Vaarala MH, Briganti A, Budaus L, Hellawell G, Hindley RG, Roobol MJ, Eggener S, Ghei M, Villers A, Bladou F, Villeirs GM, Virdi J, Boxler S, Robert G, Singh PB, VenderinkW, Hadaschik BA, Ruffion A, Hu JC, Margolis D, Crouzet S, Klotz L, Taneja SS, Pinto P, Gill I, Allen C,Giganti F, Freeman A,Morris S, Punwani S,Williams NR, Brew-Graves C, Deeks J, Takwoingi Y, Emberton M, Moore CM, PRECISION Study Group Collaborators, 2018, MRI-targeted or standard biopsy for prostate-Cancer diagnosis. N Engl J Med 378(19):1767–1777 Van Hove A, Savoie PH, Maurin C et al, 2014, Comparison of image-guided targeted biopsies versus systematic randomized biopsies in the detection of prostate cancer: a systematic literature review of well-designed studies. World J Urol 32:847–858 Kayano PP, Carneiro K, Castilho TML et al, 2018, Comparison of Gleason upgrading rates in transrectal ultrasound systematic random biopsies versus US-MRI fusion biopsies for prostate cancer. Int Braz J Urol 44(6):1106–1113 Truesdale MD, Cheetham PJ, Turk AT et al, 2010, Gleason score concordance on biopsy-confirmed prostate cancer: is pathological re-evaluation necessary prior to radical prostatectomy? BJU Int 107:749–754 Allsbrook WC, Mangold KA, Johnson MH, Lane RB, Lane CG, Epstein JI, 2001, Interobserver reproducibility of Gleason grading of prostatic carcinoma: general pathologist. Hum Pathol 32(1):81– 88 Corcoran NM, Hong MK, Casey RG et al, 2011, Upgrading in Gleason score between prostate biopsies and pathology following radical prostatectomy significantly impacts upon the risk of biochemical recurrence. BJU Int 108:202–210 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2605 EP 2612 DI 10.1007/s12253-020-00860-w PG 8 ER PT J AU Cserni, D Zombori, T Voros, A Stajer, A Rimovszki, A Daru, K Barath, Z Cserni, G AF Cserni, Dorottya Zombori, Tamas Voros, Andras Stajer, Anette Rimovszki, Annamaria Daru, Krisztian Barath, Zoltan Cserni, Gabor TI A Clinicopathological Approach to Odontogenic Cysts: the Role of Cytokeratin 17 and bcl2 Immunohistochemistry in Identifying Odontogenic Keratocysts SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Odontogenic keratocyst; Cytokeratin 17; B cell lymphoma – 2 (bcl2); Dentigerous cyst; Radicular cyst ID Odontogenic keratocyst; Cytokeratin 17; B cell lymphoma – 2 (bcl2); Dentigerous cyst; Radicular cyst AB Odontogenic keratocysts (OKCs) are developmental cysts of the jaws that require proper diagnosis due to their potential for local aggressive growth and recurrences. OKCs have a typical parakeratotic epithelium demonstrating transepithelial cytokeratin 17 (CK17) and basal bcl2 staining on immunohistochemistry (IHC), which distinguishes them from other common jaw cysts. Secondary to inflammation, the epithelial lining may be altered and loses the typical IHC phenotype. The aim of the present study was to analyse a series of consecutive jaw cysts for their expression of CK17 and bcl2 and assess how these IHC stains may help in their diagnosis. All cysts were retrospectively assessed for available clinical, radiological and pathological findings and diagnoses were revised whenever needed. 85 cysts from 72 patients were collected from two departments. The series had 21 OKCs, the remaining non-OKCs included radicular/residual, dentigerous, paradental, lateral periodontal, botryoid odontogenic cysts. OKCs with typical epithelium showed the typical IHC phenotype, which was generally lost in inflammation-associated altered epithelium. Contrarily to earlier descriptions, a wide variety of CK17 positivity was seen in the majority of non-OKCs, including focal transepithelial staining. Basal bcl2 staining was also seen in 16 non-OKCs. These stainings were never as strong in intensity as seen in OKCs. One case was histopathologically identified as OKC due to focally maintained IHC profile. CK17 and bcl2 IHCmay help in the diagnosis of OKCs, but must be interpreted with caution and is not a yes or no tool in the diagnostic puzzle. C1 [Cserni, Dorottya] University of Szeged, Faculty of Dentistry, Department of Prosthodontics, Tisza Lajos krt 64-66, H-6720 Szeged, Hungary. [Zombori, Tamas] University of Szeged, Department of Pathology, H-6725 Szeged, Hungary. [Voros, Andras] University of Szeged, Department of Pathology, H-6725 Szeged, Hungary. [Stajer, Anette] University of Szeged, Faculty of Dentistry, Department of Prosthodontics, Tisza Lajos krt 64-66, H-6720 Szeged, Hungary. [Rimovszki, Annamaria] Bacs-Kiskun County Hospital, Department of Pathology, H-6000 Kecskemet, Hungary. [Daru, Krisztian] University of Szeged, Department of Pathology, H-6725 Szeged, Hungary. [Barath, Zoltan] University of Szeged, Faculty of Dentistry, Department of Prosthodontics, Tisza Lajos krt 64-66, H-6720 Szeged, Hungary. [Cserni, Gabor] University of Szeged, Department of Pathology, H-6725 Szeged, Hungary. RP Cserni, G (reprint author), University of Szeged, Department of Pathology, H-6725 Szeged, Hungary. EM cserni@freemail.hu CR El-Naggar AK, Chan JKC, Grandis JR, Takata T, Slootweg PJ, 2017, WHO classification of head and neck Tumours, 4th edn. International Agency for Research on Cancer, Lyon Garcia CC, Sempere FV, Diago MP, Bowen EM, 2007, The postendodontic periapical lesion: histologic and etiopathogenic aspects. Med Oral Patol Oral Cir Bucal 12:E585–E590 Barnes L, Eveson JW, Reichart P, Sidransky D, 2005, WHO classification of tumors: pathology and genetics of head and neck tumours, 3rd edn. International Agency for Research on Cancer, Lyon Gorlin RJ, Goltz RW, 1960, Multiple nevoid basal-cell epithelioma, jaw cysts and bifid rib. A syndrome. N Engl J Med 262:908– 912. , DOI 10.1056/NEJM196005052621803 Menon S, 2015, Keratocystic odontogenic tumours: etiology, pathogenesis and treatment revisited. J Maxillofac Oral Surg 14:541– 547. , DOI 10.1007/s12663-014-0734-5 Speight PM, Takata T, 2017, New tumour entities in the 4th edition of the World Health Organisation classification of head and neck tumours: odontogenic and maxillofacial bone tumours. Virchows Arch 472:331–339. , DOI 10.1007/s00428-017-2182-3 Aragaki T, Michi Y, Katsube K, Uzawa N, Okada N, Akashi T, Amagasa T, Yamaguchi A, Sakamoto K, 2010, Comprehensive keratin profiling reveals different histopathogenesis of keratocystic odontogenic tumor and orthokeratinized odontogenic cyst. Hum Pathol 41:1718–1725. , DOI 10.1016/j.humpath.2010.05.007 Tsuji K,WatoM, Hayashi T, Yasuda N, Matsishita T, Ito T, Gamoh S, Yoshida H, Tanaka A, Morita S, 2014, The expression of cytokeratin in keratocystic odontogenic tumor, orthokeratinized odontogenic cyst, dentigerous cyst, radicular cyst and dermoid cyst. Med Mol Morphol 47:156–161. , DOI 10.1007/s00795-013-0058-4 Cserni D, Zombori T, Stajer A, Rimovszki A, Cserni G, Barath Z, 2020, Immunohistochemical characterization of reactive epithelial changes in odontogenic keratocysts. Pathol Oncol Res 26:1717– 1724. , DOI 10.1007/s12253-019-00749-3 Slusarenko da Silva Y, da Graca Naclerio-Homem M, 2020, A systematic review on the expression of bcl-2 in the nonsyndromic odontogenic keratocyst: should it be considered a cyst or a tumor? Oral Maxillofac Surg E-pub ahead of print, 2020 Jun 3. https://doi. org/10.1007/s10006-020-00856-5 Meara JG, Pilch BZ, Shah SS, CunninghamMJ, 2000, Cytokeratin expression in the odontogenic keratocyst. J Oral Maxillofac Surg 58:862–865. , DOI 10.1053/joms.2000.8205 Pogrel MA, Jordan RC, 2004, Marsupialization as a definitive treatment for the odontogenic keratocyst. J Oral Maxillofac Surg 62:651–656. , DOI 10.1016/j.joms.2003.08.029 Stoll C, Stollenwerk C, Riediger D, Mittermayer C, Alfer J, 2005, Cytokeratin expression patterns for distinction of odontogenic keratocysts from dentigerous and radicular cysts. J Oral Pathol Med 34:558–564. , DOI 10.1111/j.1600-0714.2005.00352.x Tsuneki M, Yamazaki M, Cheng J, Maruyama S, Kobayashi T, Saku T, 2010, Combined immunohistochemistry for the differential diagnosis of cystic jaw lesions: its practical use in surgical pathology. Histopathology 57:806–813. , DOI 10.1111/j. 1365-2559.2010.03712.x Hoshino M, Inoue H, Kikuchi K, Miyazaki Y, Yoshino A, Hara H, Terui T, Kusama K, Sakashita H, 2015, Comparative study of cytokeratin and langerin expression in keratinized cystic lesions of the oral and maxillofacial regions. J Oral Sci 57:287–294. https:// doi.org/10.2334/josnusd.57.287 Zivkovic ND,Mihailovic DS, Kostic MS, Cvetanovic AS,Mijovic ZZ, Milentijevic MV, Dencic TV, 2017, Markers of proliferation and cytokeratins in the differential diagnosis of jaw cysts. Ear Nose Throat J 96:376–383 Piattelli A, Fioroni M, Rubini C, 1998, Differentiation of odontogenic keratocysts from other odontogenic cysts by the expression of bcl-2 immunoreactivity. Oral Oncol 34:404–407. , DOI 10.1016/s1368-8375(98)00026-8 Martins CA, Rivero ERC, DuflothRM, Figueiredo CP, Vieira DSC, 2011, Immunohistochemical detection of factors related to cellular proliferation and apoptosis in radicular and dentigerous cysts. J Endod 37:36–39. , DOI 10.1016/j.joen.2010.09.010 Redman RS, Paal E, Chauhan S, Avers R, Bayley N, 2017, Botryoid odontogenic cyst. Exploration of proliferative activity, apoptosis and expression of TP53 and BCL2 compared to the histologically identical lateral periodontal and gingival cysts. Biotech Histochem 92:569–576. , DOI 10.1080/10520295.2017.1367231 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2613 EP 2620 DI 10.1007/s12253-020-00866-4 PG 8 ER PT J TI The Ratio of ssDNA to dsDNA in Circulating Cell-Free DNA Extract is a Stable Indicator for Diagnosis of Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cfDNA; ssDNA to dsDNA ratio; Gastric cancer; Tumor biomarker; Non-invasive ID cfDNA; ssDNA to dsDNA ratio; Gastric cancer; Tumor biomarker; Non-invasive AB Due to the different mechanisms of cell-free DNA production, the single-stranded DNA to double-stranded DNA ratio in blood maybe different between healthy individuals and gastric cancer (GC) patients.We aimed to explore the potential application of this ratio in GC diagnosis. The plasma cell-free DNA extracts from 118 healthy individuals and 106 GC patients were prepared. The levels of singlestranded DNA or double-stranded DNA in plasma, and the single-stranded DNA to double-stranded DNA ratio on the diagnostic efficiency for GC were assessed with ROC curve. The relationships between this ratio and the clinical characteristics of GC patients were analyzed. The ratios in 63 GC patients before and after surgery were compared. In healthy individuals, the single-stranded DNA to double-stranded DNA ratio was not affected by factors including age, gender and BMI, and subjected to normal distribution (P = 0.1090). GC patients had a lower value of this ratio than healthy individuals (P < 0.0001). Considering this ratio as a GC diagnostic indicator, the area under ROC curve (AUC) was 0.923[95% confidence interval (CI):0.880–0.955]. This ratio in unresectable GC was obviously lower than that in resectable GC (P = 0.0045). There was a rank correlation between this ratio and GC TNM staging (rho = −0.266, P = 0.0058), but it had no correlation with tumor size (r = 0.14, P = 0.145). Additionally, this ratio was not affected by hemolysis and repeated freeze-thaw of blood samples, and was significantly elevated after surgery(P < 0.0001). The single-stranded DNA to double-stranded DNA ratio in plasma is a stable non-invasive indicator for GC diagnosis. CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6):394–424. , DOI 10.3322/caac. 21492 Herszenyi L, Tulassay Z, 2010, Epidemiology of gastrointestinal and liver tumors. Eur Rev Med Pharmacol Sci 14(4):249–258 Kim YI, Kim YW, Choi IJ, Kim CG, Lee JY, Cho SJ, Eom BW, Yoon HM, Ryu KW, Kook MC, 2015, Long-term survival after endoscopic resection versus surgery in early gastric cancers. Endoscopy 47(4):293–301. , DOI 10.1055/s-0034- 1391284 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J, 2016, Cancer statistics in China, 2015. CA Cancer J Clin 66(2):115–132. , DOI 10.3322/caac.21338 Zeng H, Zheng R, Guo Y, Zhang S, Zou X, Wang N, Zhang L, Tang J, Chen J,Wei K, Huang S, Wang J, Yu L, Zhao D, Song G, Chen J, Shen Y, Yang X, Gu X, Jin F, Li Q, Li Y, Ge H, Zhu F, Dong J, Guo G,WuM, Du L, Sun X,He Y, ColemanMP, Baade P, Chen W, Yu XQ, 2015, Cancer survival in China, 2003-2005: a population-based study. Int J Cancer 136(8):1921–1930. https:// doi.org/10.1002/ijc.29227 Ronellenfitsch U, Schwarzbach M, Hofheinz R, Kienle P, Kieser M, Slanger TE, Burmeister B, Kelsen D, Niedzwiecki D, Schuhmacher C, Urba S, van de Velde C, Walsh TN, Ychou M, JensenK(2013, Preoperative chemo, radio, therapy versus primary surgery for gastroesophageal adenocarcinoma: systematic review with meta-analysis combining individual patient and aggregate data. Eur J Cancer 49(15):3149–3158. , DOI 10.1016/j.ejca. 2013.05.029 Alsina M,Miquel JM, DiezM, Castro S, Tabernero J, 2019, How I treat gastric adenocarcinoma. ESMO Open 4(Suppl 2):e000521. , DOI 10.1136/esmoopen-2019-000521 Skrypkina I, Tsyba L, Onyshchenko K, Morderer D, Kashparova O, Nikolaienko O, Panasenko G, Vozianov S, Romanenko A, Rynditch A, 2016, Concentration and methylation of cell-free DNA from blood plasma as diagnostic markers of renal Cancer. DisMarkers 2016:3693096. , DOI 10.1155/2016/3693096 Burnham P, Kim MS, Agbor-Enoh S, Luikart H, Valantine HA, Khush KK, De Vlaminck I, 2016, Single-stranded DNA library preparation uncovers the origin and diversity of ultrashort cellfree DNA in plasma. Sci Rep 6:27859. , DOI 10.1038/ srep27859 Bettegowda C, SausenM, Leary RJ, Kinde I,Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, Antonarakis ES, Azad NS, Bardelli A, Brem H, Cameron JL, Lee CC, Fecher LA, Gallia GL, Gibbs P, Le D, Giuntoli RL, Goggins M, Hogarty MD, Holdhoff M, Hong SM, Jiao Y, Juhl HH, Kim JJ, Siravegna G, Laheru DA, Lauricella C, Lim M, Lipson EJ, Marie SK, Netto GJ, Oliner KS, Olivi A, Olsson L, Riggins GJ, Sartore-Bianchi A, Schmidt K, Shih LM,Oba-Shinjo SM, Siena S, Theodorescu D, Tie J, Harkins TT, Veronese S,Wang TL,Weingart JD,Wolfgang CL, Wood LD, Xing D, Hruban RH, Wu J, Allen PJ, Schmidt CM, Choti MA, Velculescu VE, Kinzler KW, Vogelstein B, Papadopoulos N, Diaz LA Jr, 2014, Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 6(224):224ra224. , DOI 10.1126/scitranslmed.3007094 Siravegna G, Marsoni S, Siena S, Bardelli A, 2017, Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol 14(9):531–548. , DOI 10.1038/nrclinonc.2017.14 Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T, Piskorz AM, Parkinson C, Chin SF, Kingsbury Z, Wong AS, Marass F, Humphray S, Hadfield J, Bentley D, Chin TM, Brenton JD, Caldas C, Rosenfeld N, 2013, Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497(7447):108–112. , DOI 10.1038/nature12065 Wu HC, Yang HI, Wang Q, Chen CJ, Santella RM, 2017, Plasma DNA methylation marker and hepatocellular carcinoma risk prediction model for the general population. Carcinogenesis 38(10): 1021–1028. , DOI 10.1093/carcin/bgx078 Liao W, Mao Y, Ge P, Yang H, Xu H, Lu X, Sang X, Zhong S, 2015, Value of quantitative and qualitative analyses of circulating cell-free DNA as diagnostic tools for hepatocellular carcinoma: a meta-analysis. Medicine, Baltimore, 94(14):e722. , DOI 10.1097/MD.0000000000000722 Volckmar AL, Sultmann H, Riediger A, Fioretos T, Schirmacher P, Endris V, Stenzinger A, Dietz S, 2018, A field guide for cancer diagnostics using cell-free DNA: from principles to practice and clinical applications. Genes Chromosom Cancer 57(3):123–139. , DOI 10.1002/gcc.22517 Wei L, Wu W, Han L, YuW, Du Y, 2018, A quantitative analysis of the potential biomarkers of non-small cell lung cancer by circulating cell-free DNA. Oncol Lett 16(4):4353–4360. , DOI 10.3892/ol.2018.9198 Ponti G, Maccaferri M, Manfredini M, Kaleci S, Mandrioli M, Pellacani G, Ozben T, Depenni R, Bianchi G, Pirola GM, Tomasi A, 2018, The value of fluorimetry, Qubit, and spectrophotometry, NanoDrop, in the quantification of cell-free DNA, cfDNA, in malignant melanoma and prostate cancer patients. Clin Chim Acta 479:14–19. , DOI 10.1016/j.cca.2018.01.007 Lee TH, Montalvo L, Chrebtow V, Busch MP, 2001, Quantitation of genomic DNA in plasma and serum samples: higher concentrations of genomic DNA found in serum than in plasma. Transfusion 41(2):276–282. , DOI 10.1046/j.1537-2995.2001. 41020276.x Macheret M, Halazonetis TD, 2015, DNA replication stress as a hallmark of cancer. Annu Rev Pathol 10:425–448. , DOI 10.1146/annurev-pathol-012414-040424 Branzei D, Foiani M, 2010, Maintaining genome stability at the replication fork. Nat Rev Mol Cell Biol 11(3):208–219. https://doi. org/10.1038/nrm2852 Zeman MK, Cimprich KA, 2014, Causes and consequences of replication stress. Nat Cell Biol 16(1):2–9. , DOI 10. 1038/ncb2897 Gaillard H, Garcia-Muse T, Aguilera A, 2015, Replication stress and cancer. Nat Rev Cancer 15(5):276–289. , DOI 10. 1038/nrc3916 Thierry AR, ElMessaoudi S, Gahan PB, Anker P, StrounM(2016, Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev 35(3):347–376. , DOI 10.1007/ s10555-016-9629-x Snyder MW, Kircher M, Hill AJ, Daza RM, Shendure J, 2016, Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell 164(1–2):57–68. , DOI 10. 1016/j.cell.2015.11.050 Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R, 2001, DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 61(4):1659–1665 Zhang X,Wu Z, Shen Q, Li R, Jiang X,Wu J, Li D,Wang D, Zou C, Zhong Y, ChengX(2019, Clinical significance of cell-freeDNA concentration and integrity in serum of gastric cancer patients before and after surgery. Cell Mol Biol, Noisy-le-grand, 65(7):111– 117 Park JL, Kim HJ, Choi BY, Lee HC, Jang HR, Song KS, Noh SM, Kim SY, Han DS, Kim YS, 2012, Quantitative analysis of cell-free DNA in the plasma of gastric cancer patients. Oncol Lett 3(4):921– 926. , DOI 10.3892/ol.2012.592 You Y, Teng W, Wang J, Ma G, Ma A, Wang J, Liu P, 2018, Hypertension and physical activity in middle-aged and older adults in China. Sci Rep 8(1):16098. , DOI 10.1038/s41598-018- 34617-y Steevens J, Schouten LJ, Goldbohm RA, van den Brandt PA, 2010, Alcohol consumption, cigarette smoking and risk of subtypes of oesophageal and gastric cancer: a prospective cohort study. Gut 59(1):39–48. , DOI 10.1136/gut.2009.191080 Sorber L, ZwaenepoelK, Deschoolmeester V, Roeyen G, Lardon F, Rolfo C, Pauwels P, 2017, A comparison of cell-free DNA isolation kits: isolation and quantification of cell-free DNA in plasma. J Mol Diagn 19(1):162–168. , DOI 10.1016/j.jmoldx.2016. 09.009 Sato A, Nakashima C, Abe T, Kato J, Hirai M, Nakamura T, Komiya K, Kimura S, Sueoka E, Sueoka-Aragane N, 2018, Investigation of appropriate pre-analytical procedure for circulating free DNA from liquid biopsy. Oncotarget 9(61):31904–31914. , DOI 10.18632/oncotarget.25881 Lippi G, Salvagno GL, Montagnana M, Brocco G, Guidi GC, 2006, Influence of hemolysis on routine clinical chemistry testing. Clin Chem Lab Med 44(3):311–316. , DOI 10.1515/ CCLM.2006.054 Chang Y, Tolani B, Nie X, Zhi X, Hu M, He B, 2017, Review of the clinical applications and technological advances of circulating tumor DNA in cancer monitoring. Ther Clin RiskManag 13:1363– 1374. , DOI 10.2147/TCRM.S141991 Mathurin P, Cadranel JF, Bouraya D, Bronstein JA, Collot G, Devergie B, Poynard T, Opolon P, 1996)Marked increase in serum CA 19-9 level in patients with alcoholic cirrhosis: report of four cases. Eur J Gastroenterol Hepatol 8(11):1129–1131. https://doi. org/10.1097/00042737-199611000-00019 Shang X, Song C, Du X, Shao H, Xu D,Wang X, 2017, The serum levels of tumor marker CA19-9, CEA, CA72-4, and NSE in type 2 diabetes without malignancy and the relations to the metabolic control. Saudi Med J 38(2):204–208. , DOI 10.15537/smj. 2017.2.15649 Zali H, Rezaei-Tavirani M, Azodi M, 2011, Gastric cancer: prevention, risk factors and treatment. Gastroenterol Hepatol Bed Bench 4(4):175–185 Swets JA, 1988, Measuring the accuracy of diagnostic systems. Science 240(4857):1285–1293. , DOI 10.1126/science. 3287615 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2621 EP 2632 DI 10.1007/s12253-020-00869-1 PG 12 ER PT J AU Szekerczes, T Galamb, Varga, N Benczik, M Kocsis, A Schlachter, K Kiss, A Acs, N Schaff, Zs Jeney, Cs Lendvai, G Sobel, G AF Szekerczes, Timea Galamb, Adam Varga, Norbert Benczik, Marta Kocsis, Adrienn Schlachter, Krisztina Kiss, Andras Acs, Nandor Schaff, Zsuzsa Jeney, Csaba Lendvai, Gabor Sobel, Gabor TI Increased miR-20b Level in High Grade Cervical Intraepithelial Neoplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Cervical cancer; Cervical intraepithelial neoplasia (CIN); microRNA; Human papilloma virus ID Cervical cancer; Cervical intraepithelial neoplasia (CIN); microRNA; Human papilloma virus AB Cervical cancer is a common malignant tumor worldwide ranking fourth in incidence and mortality among females, which was reduced significantly by cytology screening and human papilloma virus (HPV) DNA testing. The specificity of cytology is high; however, the sensitivity is low, in contrast to the HPV DNA testing. Despite the success of these measures, new biomarkers are still considered to aim increasing sensitivity and specificity of screening and diagnosis. Significant alterations in microRNA (miRNA) expression have been detected in several cancers with variable consistency. To investigate the stratification role of miRNAs between normal epithelium and cervical intraepithelial neoplasia (CIN2–3), we screened the expression of 667 miRNAs to identify significant markers (n = 10), out of them 9 miRNAs were applied in the study (miR-20b, −24, −26a, −29b, −99a, −100, −147, −212, −515-3p) along with RNU48 and U6 as the references. To benchmark the miRNAs, 22 paired (tumor-free and tumor tissue pairs) laser microdissection-obtained cervical formalin fixed, paraffin embedded tissue samples were assayed. The expression of miR-20b was 2.4 times higher in CIN2–3 samples as compared to normal tissues (p < 0.0001). In the HPV16-positive subsets of the samples (n = 13), miR-20b showed 2.9-times elevation (p < 0.001), whereas miR-515 was 1.15-times downregulated (p < 0.05) in CIN2–3 as compared to normal tissue. These results suggest the potential value of miR- 20b as a statification biomarker in order to differentiate neoplastic and non-tumorous cases. C1 [Szekerczes, Timea] Semmelweis University, 2nd Department of PathologyBudapest, Hungary, Hungary. [Galamb, Adam] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Ulloi 78/A, 1082 Budapest, Hungary, Hungary. [Varga, Norbert] Oxford University, Nuffield Department of Clinical Neurosciences, Sleep & Circadian Neuroscience InstituteOxford, UK, UK. [Benczik, Marta] Synlab Hungary Ltd.Budapest, Hungary, Hungary. [Kocsis, Adrienn] NEUMANN Diagnostics Ltd.Budapest, Hungary, Hungary. [Schlachter, Krisztina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of PathologyBudapest, Hungary, Hungary. [Acs, Nandor] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Ulloi 78/A, 1082 Budapest, Hungary, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of PathologyBudapest, Hungary, Hungary. [Jeney, Csaba] Albert-Ludwigs University, Department of Microsystems EngineeringFreiburg, Germany, Germany. [Lendvai, Gabor] Semmelweis University, 2nd Department of PathologyBudapest, Hungary, Hungary. [Sobel, Gabor] Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, Ulloi 78/A, 1082 Budapest, Hungary, Hungary. RP Sobel, G (reprint author), Semmelweis Medical University, Ist Department of Gynaecology and Obstetrics, 1082 Budapest, Hungary. EM sobelg@gmail.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68:394–424. , DOI 10.3322/caac.21492 Safaeian M, ShermanME, 2013, From papanicolaou to papillomaviruses: evolving challenges in cervical cancer screening in the era of human papillomavirus vaccination. J Natl Cancer Inst 105:1524– 1526. , DOI 10.1093/jnci/djt267 Laengsri V, Kerdpin U, Plabplueng C, Treeratanapiboon L, Nuchnoi P, 2018, Cervical Cancer markers: epigenetics and microRNAs. Lab Med 49:97–111. , DOI 10.1093/ labmed/lmx080 Solomon D, Davey D, Kurman R, Moriarty A, O'Connor D, Prey M, Raab S, Sherman M, Wilbur D, Wright T Jr, Young N, Forum Group Members, Bethesda 2001 Workshop, 2002, The 2001 Bethesda system: terminology for reporting results of cervical cytology. JAMA 287:2114–2119 Crosbie E, Einstein M et al, 2013, Human papillomavirus and cervical cancer. Lancet 382:889–899. , DOI 10.1016/ S0140-6736(13)60022-7 Ronco G, Dillner J, Elfstrom KM, Tunesi S, Snijders PJF, Arbyn M, Kitchener H, Segnan N, Gilham C, Giorgi-Rossi P, Berkhof J, Peto J, Meijer CJLM, 2014, Efficacy of HPV-based screening for prevention of invasive cervical cancer: follow-up of four European randomised controlled trials. Lancet 383:524–532. , DOI 10.1016/S0140-6736(13)62218-7 Isidean SD, Franco EL, 2014, Embracing a new era in cervical cancer screening. Lancet 383:493–494. , DOI 10.1016/ S0140-6736(13)62028-0 ArbynM, Ronco G, Anttila A,Meijer CJLM, Poljak M, Ogilvie G, Koliopoulos G, Naucler P, Sankaranarayanan R, Peto J, 2012, Evidence regarding human papillomavirus testing in secondary prevention of cervical cancer. Vaccine 30:F88–F99. , DOI 10. 1016/j.vaccine.2012.06.095 Tornesello ML, Buonaguro L, Giorgi-Rossi P, Buonaguro FM, 2013, Viral and cellular biomarkers in the diagnosis of cervical intraepithelial neoplasia and cancer. Biomed Res Int 2013:519619– 519610. , DOI 10.1155/2013/519619 Nicolas I, Saco A, Barnadas E, Marimon L, Rakislova N, Fuste P, Rovirosa A, Gaba L, Bunesch L, Gil-Ibanez B, Pahisa J, Diaz- Feijoo B, Torne A, Ordi J, del Pino M, 2020, Prognostic implications of genotyping and p16 immunostaining in HPV-positive tumors of the uterine cervix.Mod Pathol 33:128–137. , DOI 10.1038/s41379-019-0360-3 Galamb A, Benczik M, Zinner B, Vigh E, Baghy K, Jeney C, Kiss A, Lendvai G, Sobel G, 2015, Dysregulation of microRNA expression in human cervical preneoplastic and neoplastic lesions. Pathol Oncol Res Oncol Ogy Res 21:503–508 Pereira PM, Marques JP, Soares AR, Carreto L, Santos MAS, 2010, Microrna expression variability in human cervical tissues. PLoS One 5:e11780. , DOI 10.1371/journal.pone. 0011780 Gocze K, Gombos K et al, 2013, Unique microRNA expression profiles in cervical cancer. Anticancer Res 33:2561–2568 Gocze K, Gombos K, Kovacs K, Juhasz K, Gocze P, Kiss I, 2015, MicroRNA expressions in HPV-induced cervical dysplasia and cancer. Anticancer Res 35:523–530 Pedroza-Torres A, Fernandez-Retana J, Peralta-Zaragoza O, Jacobo-Herrera N, Cantu de Leon D, Cerna-Cortes JF, Lopez- Camarillo C, Perez-Plasencia C, 2016, A microRNA expression signature for clinical response in locally advanced cervical cancer. Gynecol Oncol 142:557–565. , DOI 10.1016/j.ygyno. 2016.07.093 Liu SS, Chan KKL, Chu DKH, Wei TN, Lau LSK, Ngu SF, Chu MMY, Tse KY, Ip PPC, Ng EKO, Cheung ANY, Ngan HYS, 2018, Oncogenic microRNA signature for early diagnosis of cervical intraepithelial neoplasia and cancer. Mol Oncol 12:2009– 2022. , DOI 10.1002/1878-0261.12383 Pardini B, De Maria D et al, 2018, MicroRNAs as markers of progression in cervical cancer: a systematic review. BMC Cancer 18:1–17. , DOI 10.1186/s12885-018-4590-4 Kawai S, Fujii T, Kukimoto I, Yamada H, Yamamoto N, Kuroda M, Otani S, Ichikawa R, Nishio E, Torii Y, Iwata A, 2018, Identification of miRNAs in cervical mucus as a novel diagnostic marker for cervical neoplasia. Sci Rep 8:1–11. , DOI 10. 1038/s41598-018-25310-1 Wang S, Chen X, 2018, Identification of potential biomarkers in cervical cancer with combined public mRNA and miRNA expression microarray data analysis. Oncol Lett 16:5200–5208. https:// doi.org/10.3892/ol.2018.9323 Kocsis A, Takacs T, Jeney C, Schaff Z, Koiss R, Jaray B, Sobel G, Pap K, Szekely I, Ferenci T, Lai HC, Nyiri M, Benczik M, 2017, Performance of a new HPV and biomarker assay in the management of hrHPV positive women: subanalysis of the ongoing multicenter TRACE clinical trial, n > 6,000, to evaluate POU4F3 methylation as a potential biomarker of cervical precancer and cancer. Int J Cancer 140:1119–1133. , DOI 10.1002/ijc.30534 Andersen CL, Jensen JL, Orntoft TF, 2004, Normalization of realtime quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and Colon Cancer data sets. Cancer Res 64:5245–5250. , DOI 10.1158/0008-5472.CAN-04-0496 Yang Y, Liu Y, Li G, Li L, Geng P, SongH, 2018)MicroRNA-214 suppresses the growth of cervical cancer cells by targeting EZH2. Oncol Lett 16:5679–5686. , DOI 10.3892/ol.2018.9363 Chen R, Shi YH et al, 2018, Systematic prediction of target genes and pathways in cervical cancer from microRNA expression data. Oncol Lett 15:9994–10000. , DOI 10.3892/ol.2018.8566 Dweep H, Sticht C, Pandey P, Gretz N, 2011, miRWalk–database: prediction of possible miRNA binding sites by “walking” the genes of three genomes. J Biomed Inform 44:839–847. , DOI 10. 1016/j.jbi.2011.05.002 Iorio MV, Croce CM, 2012, Causes and consequences of MicroRNA dysregulation. Cancer J, United States, 18:215–222. , DOI 10.1097/PPO.0b013e318250c001 Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles G, Clark NR, Ma’ayan A, 2013, Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14:128. , DOI 10.1186/1471-2105-14-128 Gonzalez-Quintana V, Palma-Berre L et al, 2016, MicroRNAs are involved in cervical cancer development, progression, clinical outcome and improvement treatment response, review). Oncol Rep 35: 3–12. , DOI 10.3892/or.2015.4369 Kori M, Arga KY, 2018, Potential biomarkers and therapeutic targets in cervical cancer: insights from the meta-analysis of transcriptomics data within network biomedicine perspective. PLoS One 13:1–27. , DOI 10.1371/journal.pone.0200717 Cheung TH, Man KNM, Yu MY, Yim SF, Siu NSS, Lo KWK, Doran G, Wong RRY, Wang VW, Smith DI, Worley Jr MJ, Berkowitz RS, Chung TKH, Wong YF, 2012, Dysregulated microRNAs in the pathogenesis and progression of cervical neoplasm. Cell Cycle 11:2876–2884. , DOI 10.4161/cc.21278 Xu H, Li W, Luo S, Yuan J, Hao L, 2019, Adipose derived stem cells promote tumor metastasis in breast Cancer cells by stem cell factor inhibition of miR20b. Cell Signal 62:109350. , DOI 10.1016/j.cellsig.2019.109350 Cheng Y, Geng L, Zhao L, Zuo P, Wang J, 2017, Human papillomavirus E6-regulated microRNA-20b promotes invasion in cervical cancer by targeting tissue inhibitor of metalloproteinase 2. Mol Med Rep 16:5464–5470. , DOI 10.3892/mmr.2017.7231 He Y, Lin J, Ding Y, Liu G, Luo Y, Huang M, Xu C, Kim TK, Etheridge A, Lin M, Kong D, Wang K, 2016, A systematic study on dysregulated microRNAs in cervical cancer development. Int J Cancer 138:1312–1327. , DOI 10.1002/ijc.29618 Wang X., Tang S., le S.Y., Lu R., Rader J.S., Meyers C., Zheng Z.M., 2008, Aberrant expression of oncogenic and tumorsuppressive microRNAs in cervical cancer is required for cancer cell growth PLoS One 3 , DOI 10.1371/journal.pone. 0002557 Li Y, Zhang Z, Xiao Z, Lin Y, Luo T, Zhou Q, Zhang X, 2017, Chemotherapy-mediated miR-29b expression inhibits the invasion and angiogenesis of cervical cancer. Oncotarget 8:14655–14665. , DOI 10.18632/oncotarget.14738 Servin-Gonzalez LS, Granados-Lopez AJ et al, 2015, Families of microRNAs expressed in clusters regulate cell signaling in cervical cancer. Int J Mol Sci 16:12773–12790. , DOI 10.3390/ ijms160612773 Zhang Y, Zhang H et al, 2016, MicroRNA-147 suppresses proliferation, invasion and migration through the AKT/mTOR signaling pathway in breast cancer. Oncol Lett 11:405–410. , DOI 10.3892/ol.2015.3842 Han X, Li X, Zhao H, Zhou D, Sun B, Liu A, Zhang J, Cui Z, Ma X, Yuan L, 2019, Serum miR-515-3p, a potential new RNA biomarker, is involved in gastric carcinoma. J Cell Biochem 120:15834– 15843. , DOI 10.1002/jcb.28854 Virtanen E., Pietila T., Nieminen P., Qian K., Auvinen E., 2016, Low expression levels of putative HPV encoded microRNAs in cervical samples. Springerplus 5 , DOI 10.1186/s40064- 016-3524-3 Wang X,Wang HK, Li Y, HafnerM, Banerjee NS, Tang S, Briskin D, Meyers C, Chow LT, Xie X, Tuschl T, Zheng ZM, 2014, MicroRNAs are biomarkers of oncogenic human papillomavirus infections. Proc Natl Acad Sci U S A 111:4262–4267. https://doi. org/10.1073/pnas.1401430111 Liu Z, Mao L,Wang L, Zhang H, Hu X, 2020, MiR-218 functions as a tumor suppressor gene in cervical cancer. Mol Med Rep 21: 209–219. , DOI 10.3892/mmr.2019.10809 Nan P, Niu Y, Wang X, Li Q, 2019, MiR-29a function as tumor suppressor in cervical cancer by targeting sirt1 and predict patient prognosis. Onco Targets Ther 12:6917–6925. , DOI 10. 2147/OTT.S218043 Jiang W, Pan J-J, Deng YH, Liang MR, Yao LH, 2017, Downregulated serum microRNA-101 is associated with aggressive progression and poor prognosis of cervical cancer. J Gynecol Oncol 28:e75. , DOI 10.3802/jgo.2017.28.e75 Su Y, Xiong J, Hu J, Wei X, Zhang X, Rao L, 2016, MicroRNA- 140-5p targets insulin like growth factor 2 mRNA binding protein 1, IGF2BP1, to suppress cervical cancer growth and metastasis. Oncotarget 7:68397–68411. , DOI 10.18632/ ONCOTARGET.11722 Varghese VK, Shukla V, Jishnu PV, Kabekkodu SP, Pandey D, Sharan K, Satyamoorthy K, 2019, Characterizing methylation regulated miRNA in carcinoma of the human uterine cervix. Life Sci 232:116668. , DOI 10.1016/j.lfs.2019.116668 Berti F, Salviano-Silva A et al, 2019, From squamous intraepithelial lesions to cervical cancer: circulating microRNAs as potential biomarkers in cervical carcinogenesis. Biochim Biophys Acta Rev Cancer 1872:188306. , DOI 10.1016/ j.bbcan.2019.08.001 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2633 EP 2640 DI 10.1007/s12253-020-00852-w PG 8 ER PT J AU del Arco, DC Munoz, EL Manchado, BR Garcia, PA Medina, OL Roldan, ME Fernandez, SG Soledad, GGdlH Acenero, MJF AF del Arco, Diaz Cristina Munoz, Estrada Lourdes Manchado, Barderas Rodrigo Garcia, Pelaez Alberto Medina, Ortega Luis Roldan, Molina Elena Fernandez, Solis Guillermo Soledad, Garcia Gomez de las Heras Acenero, Maria Jesus Fernandez TI Prognostic Role of Aryl Hydrocarbon Receptor Interacting Protein (AIP) Immunohistochemical Expression in Patients with Resected Gastric Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Gastric cancer; Prognosis; Aryl hydrocarbon receptor interacting protein; AIP; Immunohistochemistry ID Gastric cancer; Prognosis; Aryl hydrocarbon receptor interacting protein; AIP; Immunohistochemistry AB Aryl hydrocarbon receptor (AHR) interacting protein (AIP) is a chaperone which binds to inactive AHR in the cell cytoplasm. AHR is best known for mediating the toxicity of halogenated aromatics, but it has also been linked to carcinogenesis and tumor progression in several tumor types. Our aims are to assess the features of AIP immunohistochemical (IHC) staining and to evaluate its possible role as a prognostic marker in gastric cancer (GC). Retrospective study of 147 cases of resected GC. Clinicopathological features were collected, tissue microarrays were constructed for AIP IHC and statistical analysis were performed. AIP staining was observed in 50.3% of tumors. All AIP-positive cases exhibited cytoplasmic or membranous staining, variably associated with nuclear co-staining. 93.2% of AIP-positive tumors showed AIP immunoreactivity in 100% of cells. Staining intensity was mild, moderate and intense in 33.8%, 13.5%and 52.7%of cases. Tumors were stratified according to AIP staining intensity into low expression (no or mild AIP immunoreactivity) and high expression (moderate or intense AIP immunoreactivity). 36.6% of our cases showed high AIP expression. High AIP expression was significantly and independently correlated to tumor progression and cancer death. Tumors with high AIP expression showed lower survival and higher progression rates. AIP expression might be useful for determining GC prognosis. More studies are needed to clarify the role of AHR pathway in GC, AIP expression and its potential use as a surrogate marker for selecting patients for AHR modulation therapy. C1 [del Arco, Diaz Cristina] Autonomous University of MadridMadrid, Spain. [Munoz, Estrada Lourdes] Rey Juan Carlos UniversityMadrid, Spain. [Manchado, Barderas Rodrigo] Carlos III Health Institute (ISCIII)Madrid, Spain. [Garcia, Pelaez Alberto] Autonomous University of Madrid, University Hospital La PazMadrid, Spain. [Medina, Ortega Luis] Autonomous University of MadridMadrid, Spain. [Roldan, Molina Elena] Hospital Clinico San CarlosMadrid, Spain. [Fernandez, Solis Guillermo] Carlos III Health Institute (ISCIII)Madrid, Spain. [Soledad, Garcia Gomez de las Heras] Rey Juan Carlos UniversityMadrid, Spain. [Acenero, Maria Jesus Fernandez] Autonomous University of MadridMadrid, Spain. RP del Arco, DC (reprint author), Autonomous University of Madrid, Madrid, Spain. EM crisdelarco@gmail.com CR Camargo MC, Figueiredo C, Machado JC, 2019, Review: gastric malignancies: basic aspects. Helicobacter 24:e12642 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A et al, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68:394–424 Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F, 2014, Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomark Prev 23: 700–713 Sitarz R, Skierucha M, Mielko J, Offerhaus GJA, MAciejewski R, Polkowski WP, 2018, Gastric cancer: epidemiology, prevention, classification, and treatment. Cancer Manag Res 10:239 Wittekind C, 2015, The development of the TNM classification of gastric cancer. Pathol Li Y, Wang D, Zhao B, Wang W, Yuan S, Huang C et al, 2012, Poor prognosis of gastric adenocarcinoma with decreased expression of AHRR. PLoS One 7:e43555 Apicella M, Corso S, Giordano S, 2017, Targeted therapies for gastric cancer: failures and hopes from clinical trials. Oncotarget 8:57654–57669 Salati M, Orsi G, Smyth E, Beretta G, De Vita F, Di BartolomeoM et al, 2019, Gastric cancer: translating novels concepts into clinical practice. Cancer Treat Rev 79:101889 Boyiadzis MM, Kirkwood JM, Marshall JL, Pritchard CC, Azad NS, Gulley JL, 2018, Significance and implications of FDA approval of pembrolizumab for biomarker-defined disease. J Immunother Cancer 6:35 Yelamanchi SD, Solanki HS, Radhakrishnan A, Balakrishnan L, Advani J, Raja R et al, 2016, Signaling network map of the aryl hydrocarbon receptor. J Cell Commun Signal 10:341 Murray IA, Patterson AD, Perdew GH, 2014, Aryl hydrocarbon receptor ligands in cancer: friend and foe. Nat Rev Cancer 14:801– 814 h Safe S, Lee S-O, Jin U-H, 2013, Role of the aryl hydrocarbon receptor in carcinogenesis and potential as a drug target. Toxicol Sci 135:1–16 Poland A, Glover E, 1973, Studies on the mechanism of toxicity of the chlorinated dibenzo-p-dioxins. Environ Health Perspect 5:245– 251 Coumoul X, Fernandez-Salguero PM, 2007, The aryl hydrocarbon receptor, more than a xenobiotic-interacting protein. FEBS Lett 581:3608–3615 Feng S, Cao Z, Wang X, 2013, Role of aryl hydrocarbon receptor in cancer. Biochim Biophys Acta - Rev Cancer 1836:197–210 Kolluri SK, Jin U-H, Safe S, 2017, Role of the aryl hydrocarbon receptor in carcinogenesis and potential as an anti-cancer drug target. Arch Toxicol 91:2497–2513 Peng T-L, Chen J, Mao W, Liu X, Tao Y, Chen L-Z et al, 2009, Potential therapeutic significance of increased expression of aryl hydrocarbon receptor in human gastric cancer. World J Gastroenterol 15:1719 Zhu R, Gao C, Wang L, Zhang G, ZhangW, Zhang Z et al, 2018, Involvement of aryl hydrocarbon receptor and aryl hydrocarbon receptor repressor in Helicobacter Pylori -related gastric pathogenesis. J Cancer 9:2757–2764 Andersson P, McGuire J, Rubio C, Gradin K, Whitelaw ML, Pettersson S et al, 2002, A constitutively active dioxin/aryl hydrocarbon receptor induces stomach tumors. Proc Natl Acad Sci 99: 9990–9995. , DOI 10.1073/pnas.152706299 Peng T-L, Chen J,MaoW, Song X, Chen M-H, 2009, Aryl hydrocarbon receptor pathway activation enhances gastric cancer cell invasiveness likely through a c-Jun-dependent induction of matrix metalloproteinase-9. BMC Cell Biol 10:27 Gasiewicz TA, Henry EC, Collins LL, 2008, Expression and activity of aryl hydrocarbon receptors in development and Cancer. Crit Rev Eukaryot Gene Expr 18:279–321 RichterCA, Tillitt DE,HanninkM(2001, Regulation of subcellular localization of the aryl hydrocarbon receptor, AhR). Arch Biochem Biophys 389:207–217 Novikov O,Wang Z, Stanford EA, Parks AJ, Ramirez-Cardenas A, Landesman E et al, 2016, An aryl hydrocarbon receptor-mediated amplification loop that enforces cellmigration in ER − /PR − /Her2 − human breast cancer cells. Mol Pharmacol 90:674–688 Ma J-X, Zhang K-L, Liu X,Ma Y-L, Pei L-N, Zhu Y-F et al, 2006, Concurrent expression of aryl hydrocarbon receptor and CYP1A1 but not CYP1A1 MspI polymorphism is correlated with gastric cancers raised in Dalian, China. Cancer Lett 240:253–260 Yin X-F, Chen J, Mao W, Wang Y-H, Chen M-H, 2013, Downregulation of aryl hydrocarbon receptor expression decreases gastric cancer cell growth and invasion. Oncol Rep 30:364–370 Wei Y, Zhao L, HeWet al, 2016, Benzo[a]pyrene promotes gastric cancer cell proliferation and metastasis likely through the aryl hydrocarbon receptor and ERK-dependent induction ofMMP9 and cmyc. Int J Oncol 49:2055–2063 Yin X-F, Chen J, Mao W, Wang Y-H, Chen M-H, 2012, A selective aryl hydrocarbon receptor modulator 3,3′-Diindolylmethane inhibits gastric cancer cell growth. J Exp Clin Cancer Res 31:46 Lai D-W, Liu S-H, Karlsson AI, Lee W-J, Wang K-B, Chen Y-C et al, 2014, The novel aryl hydrocarbon receptor inhibitor biseugenol inhibits gastric tumor growth and peritoneal dissemination. Oncotarget 5. , DOI 10.18632/oncotarget.2307 Tsay JJ, Tchou-Wong K-M, Greenberg AK, Pass H, Rom WN, 2013, Aryl hydrocarbon receptor and lung Cancer. Anticancer Res 33:1247–1256 Jaffrain-Rea M-L, Angelini M, Gargano D, Tichomirowa MA, Daly AF, Vanbellinghen J-F et al, 2009, Expression of aryl hydrocarbon receptor, AHR, and AHR-interacting protein in pituitary adenomas: pathological and clinical implications. Endocr Relat Cancer 16:1029–1043. , DOI 10.1677/ERC-09-0094 Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A et al, 2015, Tissue-based map of the human proteome. Science 347:1260419–1260419 Narasimhan S, Stanford Zulick E, Novikov O, Parks A, Schlezinger J, Wang Z et al, 2018, Towards resolving the pro- and anti-tumor effects of the aryl hydrocarbon receptor. Int J Mol Sci 19:1388 YeM, Zhang Y, Gao H, Xu Y, Jing P,Wu J et al, 2018, Activation of the aryl hydrocarbon receptor leads to resistance to EGFR TKIs in non–small cell lung Cancer by activating Src-mediated bypass signaling. Clin Cancer Res 24:1227–1239 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2641 EP 2650 DI 10.1007/s12253-020-00863-7 PG 10 ER PT J AU Dobi, Darazs, B Fodor, E Cserhati, A Egyud, Zs Maraz, A Laszlo, Sz Dodd, L Reisz, Z Barzo, P Olah, J Hideghety, K AF Dobi, Agnes Darazs, Barbara Fodor, Emese Cserhati, Adrienne Egyud, Zsofia Maraz, Aniko Laszlo, Szilvia Dodd, Leopold Reisz, Zita Barzo, Pal Olah, Judit Hideghety, Katalin TI Low Fraction Size Re-irradiation for Large Volume Recurrence of Glial Tumours SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Re-irradiation; Multiformglioblastoma; Glioma ID Re-irradiation; Multiformglioblastoma; Glioma AB The aim of the present study was to evaluate the efficacy of re-irradiation (re-RT) in patients with advanced local relapses of glial tumours and to define the factors influencing the result of the hyper-fractionated external beam therapy on progression after primary management. We have analysed the data of 55 patients with brain tumours (GBM: 28) on progression, who were reirradiated between January 2007 and December 2018. The mean volume of the recurrent tumour was 118 cm3, and the mean planning target volume (PTV) was 316 cm3, to which 32 Gy was delivered in 20 fractions at least 7.7 months after the first radiotherapy, using 3D conformal radiotherapy (CRT) or intensity modulated radiotherapy (IMRT). The median overall survival (mOS) from the re-RT was 8.4 months, and the 6-month and the 12-month OS rate was 64% and 31%, respectively. The most important factors by univariate analysis, which significantly improved the outcome of re-RT were the longer time interval between the diagnosis and second radiotherapy (p = 0.029), the lower histology grade (p = 0.034), volume of the recurrent tumour (p = 0.006) and Karnofsky performance status (KPS) (p = 0.009) at the re-irradiation. Our low fraction size re-irradiation ≥ 8 months after the first radiotherapy proved to be safe and beneficial for patients with large volume recurrent glial tumours. C1 [Dobi, Agnes] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Darazs, Barbara] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Fodor, Emese] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Cserhati, Adrienne] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Egyud, Zsofia] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Maraz, Aniko] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Laszlo, Szilvia] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Dodd, Leopold] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Reisz, Zita] University of Szeged, Department of Pathology, Allomas utca 1, H-6725 Szeged, Hungary. [Barzo, Pal] University of Szeged, Department of Neurosurgery, Semmelweis utca 6, H-6725 Szeged, Hungary. [Olah, Judit] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. [Hideghety, Katalin] University of Szeged, Department of Oncotherapy, Koranyi fasor 12, H-6720 Szeged, Hungary. RP Dobi, (reprint author), University of Szeged, Department of Oncotherapy, H-6720 Szeged, Hungary. CR Stupp R, Tonn J-C, BradaM, Pentheroudakis G, 2010, High-grade malignant glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annal of Oncology 21:v190–v193. , DOI 10.1093/annonc/mdq187 Dolocek TA, Propp JM, StroupNE et al, 2012, CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005–2009. Neuro Oncol 14(Suppl 5):1–49 Wem PY, Kesari S, 2008, Malignant gliomas in adults. New England Med 359:492–507 Sherriff J, Tamangani J, Senthil L, Cruickshank G, Spooner D, Jones B et al, 2013, Patterns of relapse in glioblastoma multiforme following concomitant chemoradiotherapy with temozolomide. Br J Radiol 86:20120414 Pichlmeier U, Bink A, Schackert G et al, 2008, Resection and survival in glioblastoma multiforme: an RTOG recursive partitioning analysis of ALA study patients. Neuro Oncology 10: 1025–1034 Studio R, Hegi ME,Mason WP et al, 2009, Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5 year analysis of the EORTC-NCIC trial. Lancet Oncol 10:459–466 Dirks P, Bernstein M, Muller PJ, Tucker WS, 1993, The value of re-operation for recurrent glioblastoma. Can J Surg 36:271–275 Barbagallo GM, Jenkinson MD, Brodbelt AR, 2008, ”Recurrent” glioblastoma multiforme, when should we reoperate? Br J Neurosurg 22:452–455 Perry JR et al, 2010, Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. Clin Oncol 28(12):2051–2057 Meryem Aktan M, Koc, Gul Kanyilmaz, 2015, Survival following re-irradiation using intensity-modulated radiaton therapy with temozolomide in selected patients with recurrent high grade gliomas. Ann Transl Med 3(20):304. , DOI 10.3978/j.issn.2305- 5839.2015.11.29 Wang 13X, Guo G, Guan H, Yu Y, Lu J, Jinming Yu, 2019, Challenges and potential of PD-1/PD-L1checkpoint blockade immunotherapy for glioblastoma. J Exp Clin Cancer Res 38:87. , DOI 10.1186/s13046-019-1085-3 Phuphanich S,Wheeler CJ, Rudnick JD,Mazer M,Wang H, Nuno MAet al, 2013, Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunol Immunother 62:125–135 Liau LM, Ashkan K, Tran DD, Campian JL, Trusheim JE, Cobbs CS et al, 2018, First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J Transl Med 16:142 Jain KK, 2018, Critical overview of targeted therapies for glioblastoma. Front Oncol 8:419 Taunk NK, Moraes FY, Escorcia FE, Mendez LC, Beal K, Marta GN, 2016, External beam re-irradiation, combination chemoradiotherapy, and particle therapy for the treatment of recurrent glioblastoma. Expert Rev Anticancer Ther 16(3):347–358 Amichetti M, Amelio D, 2011, A Review of the role of the Reirradiation in recurrent high-grade glioma, HGG). Cancers, Basel, 3:4061–4089 Kazmi F, Soon YY, Leong Y, KohWY, Vellayappan B, 2018, Reirradiation for recurrent glioblastoma, GBM, a systematic review and meta-analyis. Journal of Neuro-Oncology 142(1):79–90 Minniti G, Scaringi C, De Sanctis V, Lanzetta G, Falco T, Di Stefano D et al, 2013, Hypofractionated stereotactic radiotherapy and continuous low- dose temozolomide in patients with recurrent or progressive malignant gliomas. J Neurooncol 111:187–194 Grosu AL, Weber WA, Franz M, Stark S, Piert M, Thamm R et al, 2005, Re-irradiation of recurrent high- grade gliomas using amino acid PET, SPECT)/CT/MRI image fusion to determine gross tumor volume for stereotactic fractionated radiotherapy. Int J Radiat Oncol Biol Phys 63:511–519.24 Ernst-Stecken A, Ganslandt O, Lambrecht U, Sauer R, Grabenbauer G, 2007, Survival and quality of life after hypofractionated stereotactic radiotherapy for recurrent malignant glioma. J Neurooncol 81:287–294.25 Wuthrick EJ, Curran WJ, Camphausen K, Lin A, Glass J, Evans J et al, 2014, A pilot study of hypofractionated stereotactic radiation therapy and sunitinib in previously irradiated patients with recurrent high- grade glioma. Int J Radiat Oncol Biol Phys 90:369–375.26 Patel M, Siddiqui F, Jin J-Y, Mikkelsen T, Rosenblum M, Movsas B et al, 2009, Salvage re-irradiation for recurrent glioblastoma with radiosurgery: radiographic response and improved survival. J Neurooncol 92:185–191 Bratengeier K, Oechsner M, Gainey M, Flentje M, 2009, Remarks on reporting and recording consistent with the ICRU reference dose. Radiat Oncol 4:44. , DOI 10.1186/1748-717X-4-44 Lin NU, Lee EQ et al, 2015, Response assessment criteria for brain metastases: proposal from the RANO group. Lancet Oncology 16: e270–e278 Straube C, Elpula G, Gempt J, Gerhardt J, Bette S, Zimmer C, Schmidt-Graf F, Meyer B, Combs SE, 2017, Re-irradiation after gross total resection of recurrent glioblastoma. Strahlenther Onkol. , DOI 10.1007/s00066-017-1161-6 Fokas E, Wacker U, GrossMW, Henzel M, Encheva E, Engenhart- Cabillic R, 2009, Hypofractionated stereotactic re-irradiation of recurrent glioblastomas. Strahlenther Onkol 185:235–240. https:// doi.org/10.1007/s00066-009-1753-x Niyazi M, Siefert A, Schwarz SB et al, 2011, Therapeutic options for recurrent malignant glioma. Radiother Oncol 98:1–14 Amelio D, Amichetti M, 2012, Radiation therapy for the treatment of recurrent glioblastoma: an overview. Cancers, Basel, 4:257–280 Redmond KJ, Mehta M, 2015, Stereotactic radiosurgery for glioblastoma. Cureus 7:e413 Fogh SE, Andrews DW, Glass J et al, 2010, Hypofractionated stereotactic radiation therapy: an effective therapy for recurrent high-grade gliomas. J Clin Oncol 28:3048–3053 Chatzikonstantinou G, Zamboglou N, Archavlis E, Strouthos I, Zoga E, Milickovic N, Hilaris B, Baltas D, Rodel C, Tselis N., 2018, CT-guided interstitial HDR-brachytherapy for recurrent glioblastoma multiforme: a 20-year single-institute experience Strahlenther Onkol. 194(12):1171–1179. , DOI 10.1007/ s00066-018-1358-3 Combs SE, Niyazi M, Adeberg S, Bougatf N, Kaul D et al, 2018, Re-irradiation of recurrent gliomas: pooled analysis and validation of an established prognostic score—report of the Radiation Oncology Group, ROG, of the German Cancer Consortium, DKTK). Cancer Med 7(5): 1742–1749 Krauze AV, Peters C, Cheng J et al, 2017, Re-irradiation for recurrent glioma- the NCI experience in tumor control, OAR toxicity and proposal of a novel prognostic scoring system. Radiat Oncol Lond Engl 12(1):191. , DOI 10.1186/s13014-017-0930-9 Zwirner K, Paulsen F, Schittenhelm J et al, 2017, Prognostic parameters and outcome after re-irradiation for progressive glioblastoma. Acta Neurol Scand 136:239–245 Klobukowski L, Falkov A, Chelimo C et al, 2018, A retrospective review of re-irradiating patients’ recurrent high-grade gliomas. Clin Oncol, R Coll Radiol, 30:563–570 Post CCB, Kramer MCA, Smid EJ et al, 2019, Patterns of reirradiation for recurrent gliomas and validation of a prognostic score. Radiother Oncol 130:156–163 ShiW, Scannell BryanM,GilbertMR,MehtaMP, Blumenthal DT, Brown PD, Valeinis E, Hopkins K, Souhami L, Andrews DW, Tzuk-Shina T, Howard SP, Youssef EF, Lessard N, Dignam JJ, Werner-Wasik M, 2017, Investigating the effect of reirradiation or systemic therapy in patients with glioblastoma after tumor progression: a secondary analysis of NRG oncology/radiation therapy oncology group trial 0525. Int J Radiat Oncol Biol Phys 100:38–44. , DOI 10.1016/j.ijrobp.2017.08.038 Combs SE, Edler L, Rausch R,Welzel T, WickW, Debus J, 2013, Generation and validation of a prognostic score to predict outcome after re-irradiation of recurrent glioma. Acta Oncol 52:147–152. , DOI 10.3109/0284186X.2012.692882 Kessel KA, Hesse J, Straube C, Zimmer C, Schmidt-Graf F, Schlegel J, Meyer B, Combs SE, 2017, Modification and optimization of an established prognostic score after re-irradiation of recurrent glioma. PLoS ONE 12:e0180457. , DOI 10.1371/ journal.pone.0180457 Kim IH, 2019, Appraisal of re-irradiation for the recurrent glioblastoma in the era of MGMT promotor methylation. Radiat Oncol J 37(1):1–12. , DOI 10.3857/roj.2019.00171T Tunthanathip T, Ratanalert S, Sae-heng S, Oearsakul T, Sakaruncchai I, Kaewborisutsakul A, Chotsampancharoen T, Intusoma U, Kitkhuandee A, Vaniyapong T, 2020, Prognostic Factors and Nomogram Predicting Survival in Diffuse Astrocytoma. J Neurosci Rural Pract 11(1):135–143. https://doi. org/10.1055/s-0039-3403446 Guyotat J, Signorelli F, Frappaz D,Madarassy G, Ricci AC, Bret P, 2000, Is re-operation for recurrence of glioblastoma justified? Oncol Rep 7:899–904 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2651 EP 2658 DI 10.1007/s12253-020-00868-2 PG 8 ER PT J AU Hou, R Liu, Y Su, Y Shu, Z AF Hou, Ruizhi Liu, Yan Su, Yanzhuo Shu, Zhenbo TI Overexpression of Long Non-Coding RNA FGF14-AS2 Inhibits Colorectal Cancer Proliferation Via the RERG/Ras/ERK Signaling by Sponging microRNA-1288-3p SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE FGF14-AS2; RERG; Colorectal cancer ID FGF14-AS2; RERG; Colorectal cancer AB Colorectal cancer remains one of most common cancer types with poor prognosis globally. Recent years, numerous studies depicted pivotal roles of lncRNAs in colorectal cancer progression. This study aimed to investigate the role of FGF14-AS2 in colorectal cancer development. FGF14-AS2 was found as a significantly downregulated lncRNA in TCGA dataset. Via RTqPCR, we confirmed the downregulation of FGF14-AS2 in collected colorectal carcinoma samples. Transfection of plasmid containing full length of FGF14-AS2 repressed cell proliferation and induced elevation of cell apoptosis in colorectal cancer cells. In addition, FGF14-AS2 overexpression inactivated MAPK/ERK signaling in cells. Bioinformatic analysis and subsequent cellbased assays showed that FGF14-AS2 sponging miR-1288-3p, an oncogenic miRNA in colorectal cancer. RERG, the regulator of Ras/ERK pathway, was predicted and verified as target gene of miR-1288. Via downregulation of miR-1288, FGF14-AS2 elevated RERG expression in colorectal cancer cells. Rescue assays indicated that FGF14-AS2 relied on regulation of RERG to control cell proliferation and apoptosis in colorectal cancer. Taken together, the current study demonstrated FGF14-AS2 as a regulator of colorectal cancer development via downregulation of miR-1288-3p and inactivation of Ras/ERK signaling. C1 [Hou, Ruizhi] Jilin University, the Third Hospital of Jilin University, Department of Gastrointestinal Colorectal Surgery, 130033 Changchun, Jilin, China. [Liu, Yan] Jilin University, the Third Hospital of Jilin University, Department of Ultrasound, 130033 Changchun, Jilin, China. [Su, Yanzhuo] Jilin University, the Third Hospital of Jilin University, Department of Gastrointestinal Colorectal Surgery, 130033 Changchun, Jilin, China. [Shu, Zhenbo] Jilin University, the Third Hospital of Jilin University, Department of Gastrointestinal Colorectal Surgery, 130033 Changchun, Jilin, China. RP Shu, Z (reprint author), Jilin University, the Third Hospital of Jilin University, Department of Gastrointestinal Colorectal Surgery, 130033 Changchun, China. EM shuzhenbobo@outlook.com CR Siegel RL, Miller KD, Jemal A, 2019, Cancer statistics, 2019. CA Cancer J Clin 69:7–34 Rosello S, Papaccio F, Roda D, Tarazona N, Cervantes A, 2018, The role of chemotherapy in localized and locally advanced rectal cancer: a systematic revision. Cancer Treat Rev 63:156–171 Mitra SA, Mitra AP, Triche TJ, 2012, A central role for long noncoding RNA in cancer. Front Genet 3:17 Fan Q, Liu B, 2018, Comprehensive analysis of a long noncoding RNA-associated competing endogenous RNA network in colorectal cancer. Onco Targets Ther 11:2453–2466 Wei L, Wang X, Lv L, Zheng Y, Zhang N, Yang M, 2019, The emerging role of noncoding RNAs in colorectal cancer chemoresistance. Cell Oncol, Dordr, 42:757–768 Xu T, Wu K, Zhang L, et al., 2019, Long non-coding RNA LINC00858 exerts a tumor-promoting role in colon cancer via HNF4alpha and WNK2 regulation. Cell Oncol, Dordr) Bhan A,Mandal SS, 2014, Long noncoding RNAs: emerging stars in gene regulation, epigenetics and human disease. ChemMedChem. 9:1932–1956 Xu Y, Zhang X, Hu X, ZhouW, Zhang P, Zhang J, Yang S, Liu Y, 2018, The effects of lncRNA MALAT1 on proliferation, invasion and migration in colorectal cancer through regulating SOX9. Mol Med 24:52 Yang F, LiuYH,Dong SY,Ma RM, Bhandari A, ZhangXH, Wang OC, 2016, A novel long non-coding RNA FGF14-AS2 is correlated with progression and prognosis in breast cancer. Biochem Biophys Res Commun 470:479–483 Yang Y, Xun N, Wu JG, 2019, Long non-coding RNA FGF14- AS2 represses proliferation, migration, invasion, and induces apoptosis in breast cancer by sponging miR-205-5p. Eur Rev Med Pharmacol Sci 23:6971–6982 BurottoM, Chiou VL, Lee JM, Kohn EC, 2014, The MAPK pathway across different malignancies: a new perspective. Cancer. 120: 3446–3456 Sun QY, Ding LW, Johnson K, Zhou S, Tyner JW, Yang H, Doan NB, Said JW, Xiao JF, LohXY, Ran XB,VenkatachalamN, Lao Z, Chen Y, Xu L, Fan LF, Chien W, Lin DC, Koeffler HP, 2019, SOX7 regulates MAPK/ERK-BIM mediated apoptosis in cancer cells. Oncogene. 38:6196–6210 Pan H, Wang Y, Na K, Wang Y, Wang L, Li Z, Guo C, Guo D, Wang X, 2019, Autophagic flux disruption contributes to Ganoderma lucidum polysaccharide-induced apoptosis in human colorectal cancer cells via MAPK/ERK activation. Cell Death Dis 10:456 Zhang X, Ma L, Qi J, Shan H, Yu W, Gu Y, 2015)MAPK/ERK signaling pathway-inducedhyper-O-GlcNAcylation enhances cancer malignancy. Mol Cell Biochem 410:101–110 Finlin BS, Gau CL, Murphy GA, Shao H, Kimel T, Seitz RS, Chiu YF, Botstein D, Brown PO, der CJ, Tamanoi F, Andres DA, Perou CM, 2001, RERG is a novel ras-related, estrogen-regulated and growth-inhibitory gene in breast cancer. J Biol Chem 276:42259– 42267 Habashy HO, Powe DG, Glaab E, Ball G, Spiteri I, Krasnogor N, Garibaldi JM, Rakha EA, Green AR, Caldas C, Ellis IO, 2011, RERG, Ras-like, oestrogen-regulated, growth-inhibitor, expression in breast cancer: a marker of ER-positive luminal-like subtype. Breast Cancer Res Treat 128:315–326 Wang AG, Fang W, Han YH, Cho SM, Choi JY, Lee KH, Kim WH, Kim JM, Park MG, Yu DY, Kim NS, Lee DS, 2006, Expression of the RERG gene is gender-dependent in hepatocellular carcinoma and regulated by histone deacetyltransferases. J Korean Med Sci 21:891–896 Xiong Y, HuangH, Chen S,Dai H, Zhang L, 2019, ERK5regulated RERG expression promotes cancer progression in prostatic carcinoma. Oncol Rep 41:1160–1168 Yang R, Chen B, Pfutze K, Buch S, Steinke V, Holinski-Feder E, Stocker S, von Schonfels W, Becker T, Schackert HK, Royer- Pokora B, Kloor M, Schmiegel WH, Buttner R, Engel C, Lascorz Puertolas J, Forsti A, Kunkel N, Bugert P, Schreiber S, Krawczak M, Schafmayer C, Propping P, Hampe J, Hemminki K, Burwinkel B, 2014)Genome-wide analysis associates familial colorectal cancer with increases in copy number variations and a rare structural variation at 12p12.3. Carcinogenesis. 35:315–323 Chen Y, Yu X, Xu Y, Shen H, 2017, Identification of dysregulated lncRNAs profiling andmetastasis-associated lncRNAs in colorectal cancer by genome-wide analysis. Cancer Med 6:2321–2330 Berg M, Soreide K, 2012, EGFR and downstream genetic alterations in KRAS/BRAF and PI3K/AKT pathways in colorectal cancer: implications for targeted therapy. Discov Med 14:207–214 Berg M, Danielsen SA, Ahlquist T, Merok MA, Agesen TH, Vatn MH, Mala T, Sjo OH, Bakka A, Moberg I, Fetveit T, Mathisen O, Husby A, Sandvik O, Nesbakken A, Thiis-Evensen E, Lothe RA, 2010, DNA sequence profiles of the colorectal cancer critical gene set KRAS-BRAF-PIK3CA-PTEN-TP53 related to age at disease onset. PLoS One 5:e13978 Wang Q,Yang L,Hu X, JiangY, Hu Y, Liu Z, Liu J,Wen T,Ma Y, An G, Feng G, 2017, Upregulated NNT-AS1, a long noncoding RNA, contributes to proliferation andmigration of colorectal cancer cells in vitro and in vivo. Oncotarget. 8:3441–3453 Sang B, Zhang YY, Guo ST, Kong LF, Cheng Q, Liu GZ, Thorne RF, Zhang XD, Jin L, WuM(2018, Dual functions for OVAAL in initiation of RAF/MEK/ERK prosurvival signals and evasion of p27-mediated cellular senescence. Proc Natl Acad Sci U S A 115: E11661–E11E70 ZhaoW,Ma N,Wang S,MoY, Zhang Z, HuangG,MidorikawaK, Hiraku Y, Oikawa S, Murata M, Takeuchi K, 2017, RERG suppresses cell proliferation,migration and angiogenesis through ERK/ NF-kappaB signaling pathway in nasopharyngeal carcinoma. J Exp Clin Cancer Res 36:88 Ho JY, Hsu RJ, Liu JM, Chen SC, Liao GS, Gao HW, Yu CP, 2017)MicroRNA-382-5p aggravates breast cancer progression by regulating the RERG/Ras/ERK signaling axis. Oncotarget. 8: 22443–22459 Zhang Y, Ren S, Yuan F et al, 2018)miR-135 promotes proliferation and stemness of oesophageal squamous cell carcinoma by targeting RERG. Artif Cells Nanomed Biotechnol 46:1210–1219 Gopalan V, Pillai S, Ebrahimi F, Salajegheh A, Lam TC, le TK, Langsford N, Ho YH, Smith RA, Lam AKY, 2014, Regulation of microRNA-1288 in colorectal cancer: altered expression and its clinicopathological significance. Mol Carcinog 53(Suppl 1):E36– E44 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2659 EP 2667 DI 10.1007/s12253-020-00862-8 PG 9 ER PT J AU Lendvai, G Szekerczes, T Kontsek, E Selvam, A Szakos, A Schaff, Zs Bjornstedt, M Kiss, A AF Lendvai, Gabor Szekerczes, Timea Kontsek, Endre Selvam, Arun Szakos, Attila Schaff, Zsuzsa Bjornstedt, Mikael Kiss, Andras TI The Effect of Methylselenocysteine and Sodium Selenite Treatment on microRNA Expression in Liver Cancer Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Methylselenocysteine; Sodium selenite; microRNA expression; Hepatocellular carcinoma; Cholangiocarcinoma; IC50 value ID Methylselenocysteine; Sodium selenite; microRNA expression; Hepatocellular carcinoma; Cholangiocarcinoma; IC50 value AB The unique character of selenium compounds, including sodium selenite and Se-methylselenocysteine (MSC), is that they exert cytotoxic effects on neoplastic cells, providing a great potential for treating cancer cells being highly resistant to cytostatic drugs. However, selenium treatment may affect microRNA (miRNA) expression as the pattern of circulating miRNAs changed in a placebo-controlled selenium supplement study. This necessitates exploring possible changes in the expression profiles of miRNAs. For this, miRNAs being critical for liver function were selected and their expression was measured in hepatocellular carcinoma (HLE and HLF) and cholangiocarcinoma cell lines (TFK-1 and HuH-28) using individual TaqMan MicroRNA Assays following selenite or MSC treatments. For establishing tolerable concentrations, IC50 values were determined by performing SRB proliferation assays. The results revealed much lower IC50 values for selenite (from 2.7 to 11.3 μM) compared to MSC (from 79.5 to 322.6 μM). The treatments resulted in cell line-dependent miRNA expression patterns, with all miRNAs found to show fold change differences; however, only a few of these changes were statistically different in treated cells compared to untreated cells below IC50. Namely, miR-199a in HLF, miR-143 in TFK-1 upon MSC treatment,miR-210 in HLF and TFK-1, miR-22, -24, -122, −143 in HLF upon selenite treatment. Fold change differences revealed that miR-122 with both selenium compounds, miR-199a with MSC and miR-22 with selenite were affected. The miRNAs showing minimal alterations included miR-125b and miR-194. In conclusion, our results revealed moderately altered miRNA expression in the cell lines (less alterations following MSC treatment), being miR-122, −199a the most affected and miR-125b, -194 the least altered miRNAs upon selenium treatment. C1 [Lendvai, Gabor] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Szekerczes, Timea] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Kontsek, Endre] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Selvam, Arun] Karolinska University, Department of Pathology, SE-14186 Stockholm, Sweden. [Szakos, Attila] Karolinska University, Department of Pathology, SE-14186 Stockholm, Sweden. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. [Bjornstedt, Mikael] Karolinska University, Department of Pathology, SE-14186 Stockholm, Sweden. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, Ulloi 93, H-1091 Budapest, Hungary. RP Kiss, A (reprint author), Semmelweis University, 2nd Department of Pathology, H-1091 Budapest, Hungary. EM kiss.andras@med.semmelweis-univ.hu CR Clark LC, Combs GF Jr, Turnbull BW, Slate EH, Chalker DK, Chow J, Davis LS, Glover RA, Graham GF, Gross EG, Krongrad A, Lesher JL Jr, Park HK, Sanders BB Jr, Smith CL, Taylor JR, 1996, Effects of selenium supplementation for cancer prevention in patients with carcinoma of the skin. A randomized controlled trial Nutritional Prevention of Cancer Study Group JAMA 276: 1957–1963 Misra S, Boylan M, Selvam A, Spallholz JE, Bjornstedt M, 2015, Redox-active selenium compounds–from toxicity and cell death to cancer treatment. Nutrients 7:3536–3556. , DOI 10.3390/ nu7053536 Selenius M, Rundlof AK, Olm E, Fernandes AP, Bjornstedt M, 2010, Selenium and the selenoprotein thioredoxin reductase in the prevention, treatment and diagnostics of cancer. Antioxid Redox Signal 12:867–880. , DOI 10.1089/ars.2009.2884 TarzeA, DauplaisM,Grigoras I, LazardM, Ha-Duong NT, Barbier F, Blanquet S, Plateau P, 2007, Extracellular production of hydrogen selenide accounts for thiol-assisted toxicity of selenite against Saccharomyces cerevisiae. J Biol Chem 282:8759–8767. https:// doi.org/10.1074/jbc.M610078200 Moghadaszadeh B, Beggs AH, 2006, Selenoproteins and their impact on human health through diverse physiological pathways. Physiology, Bethesda, 21:307–315. , DOI 10.1152/ physiol.00021.2006 Ganther HE, 1968, Selenotrisulfides. Formation by the reaction of thiols with selenious acid Biochemistry 7:2898–2905. https://doi. org/10.1021/bi00848a029 Weekley CM, Harris HH, 2013, Which form is that? The importance of selenium speciation and metabolism in the prevention and treatment of disease. Chem Soc Rev 42:8870–8894. , DOI 10.1039/c3cs60272a Brodin O, Eksborg S, Wallenberg M, Asker-Hagelberg C, Larsen EH, Mohlkert D, Lenneby-Helleday C, Jacobsson H, Linder S, Misra S, Bjornstedt M, 2015, Pharmacokinetics and toxicity of sodium selenite in the treatment of patients with carcinoma in a phase I clinical trial: the SECAR study. Nutrients 7:4978–4994. , DOI 10.3390/nu7064978 Evans SO, Jacobson GM, Goodman HJB, Bird S, Jameson MB, 2019, Comparative safety and pharmacokinetic evaluation of three Oral selenium compounds in Cancer patients. Biol Trace Elem Res 189:395–404. , DOI 10.1007/s12011-018-1501-0 Marshall JR, Burk RF, Payne Ondracek R, Hill KE, Perloff M, Davis W, Pili R, George S, Bergan R, 2017, Selenomethionine and methyl selenocysteine: multiple-dose pharmacokinetics in selenium-replete men. Oncotarget 8:26312–26322. , DOI 10.18632/oncotarget.15460 Marshall JR, Ip C, Romano K, Fetterly G, Fakih M, Jovanovic B, PerloffM, Crowell J, DavisW, French-Christy R, Dew A, Coomes M, Bergan R, 2011)Methyl selenocysteine: single-dose pharmacokinetics in men. Cancer Prev Res, Phila, 4:1938–1944. https://doi. org/10.1158/1940-6207.CAPR-10-0259 Lee JI, Nian H, Cooper AJ, Sinha R, Dai J, BissonWH, Dashwood RH, Pinto JT, 2009, Alpha-keto acid metabolites of naturally occurring organoselenium compounds as inhibitors of histone deacetylase in human prostate cancer cells. Cancer Prev Res, Phila, 2:683–693. , DOI 10.1158/1940-6207.CAPR-09- 0047 Pinto JT, Lee JI, Sinha R, MacEwan ME, Cooper AJ, 2011, Chemopreventive mechanisms of alpha-keto acid metabolites of naturally occurring organoselenium compounds. Amino Acids 41:29–41. , DOI 10.1007/s00726-010-0578-3 Rooseboom M, Vermeulen NP, van Hemert N, Commandeur JN, 2001, Bioactivation of chemopreventive selenocysteine seconjugates and related amino acids by amino acid oxidases novel route of metabolism of selenoamino acids. Chem Res Toxicol 14: 996–1005. , DOI 10.1021/tx000265r Stevens JL, Robbins JD, Byrd RA, 1986, A purified cysteine conjugate beta-lyase from rat kidney cytosol. Requirement for an alpha-keto acid or an amino acid oxidase for activity and identity with soluble glutamine transaminase K. J Biol Chem 261:15529– 15537 Gabel-Jensen C, Lunoe K, Gammelgaard B, 2010, Formation of methylselenol, dimethylselenide and dimethyldiselenide in in vitro metabolismmodels determined by headspaceGC-MS.Metallomics 2:167–173. , DOI 10.1039/b914255j Vadhanavikit S, Ip C, Ganther HE, 1993, Metabolites of sodium selenite and methylated selenium compounds administered at cancer chemoprevention levels in the rat. Xenobiotica 23:731–745. , DOI 10.3109/00498259309166780 Zeng H, Wu M, Botnen JH, 2009, Methylselenol, a selenium metabolite, induces cell cycle arrest in G1 phase and apoptosis via the extracellular-regulated kinase 1/2 pathway and other cancer signaling genes. J Nutr 139:1613–1618. , DOI 10.3945/jn.109. 110320 Nian H, Bisson WH, Dashwood WM, Pinto JT, Dashwood RH, 2009, Alpha-keto acid metabolites of organoselenium compounds inhibit histone deacetylase activity in human colon cancer cells. Carcinogenesis 30:1416–1423. , DOI 10.1093/carcin/ bgp147 Lee RC, Feinbaum RL, Ambros V, 1993, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75:843–854. , DOI 10.1016/ 0092-8674(93)90529-y Wightman B, Ha I, Ruvkun G, 1993, Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 75:855–862. , DOI 10.1016/ 0092-8674(93)90530-4 Tsai WC, Hsu SD, Hsu CS, Lai TC, Chen SJ, Shen R, Huang Y, Chen HC, Lee CH, Tsai TF, Hsu MT, Wu JC, Huang HD, Shiao MS, Hsiao M, Tsou AP, 2012)MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. J Clin Invest 122: 2884–2897. , DOI 10.1172/JCI63455 Russano M, Napolitano A, Ribelli G, Iuliani M, Simonetti S, Citarella F, Pantano F, Dell'Aquila E, Anesi C, Silvestris N, Argentiero A, Solimando A, Vincenzi B, Tonini G, Santini D, 2020, Liquid biopsy and tumor heterogeneity in metastatic solid tumors: the potentiality of blood samples. J Exp Clin Cancer Res 39:95. , DOI 10.1186/s13046-020-01601-2 Peng C, Ye Y, Wang Z, Guan L, Bao S, Li B, Li W, 2019, Circulating microRNAs for the diagnosis of hepatocellular carcinoma. Dig Liver Dis 51:621–631. , DOI 10.1016/j.dld.2018. 12.011 Alehagen U, Johansson P, Aaseth J, Alexander J, Wagsater D, 2017, Significant changes in circulating microRNA by dietary supplementation of selenium and coenzyme Q10 in healthy elderly males. A subgroup analysis of a prospective randomized doubleblind placebo-controlled trial among elderly Swedish citizens. PLoS one 12:e0174880. , DOI 10.1371/journal.pone. 0174880 Siegel R, Naishadham D, Jemal A, 2013, Cancer statistics, 2013. CA Cancer J Clin 63:11–30. , DOI 10.3322/caac.21166 Chen XM, 2009, MicroRNA signatures in liver diseases. World J Gastroenterol 15:1665–1672. , DOI 10.3748/wjg.15.1665 Tili E, Michaille JJ, Croce CM, 2013, MicroRNAs play a central role in molecular dysfunctions linking inflammation with cancer. Immunol Rev 253:167–184. , DOI 10.1111/imr.12050 Ma D, Tao X, Gao F, Fan C, Wu D, 2012, miR-224 functions as an onco-miRNA in hepatocellular carcinoma cells by activating AKT signaling. Oncol Lett 4:483–488. , DOI 10.3892/ol.2012. 742 Girard M, Jacquemin E,Munnich A, Lyonnet S, Henrion-Caude A, 2008, miR-122, a paradigmfor the role of microRNAs in the liver. J Hepatol 48:648–656. , DOI 10.1016/j.jhep.2008.01.019 Amr KS, Elmawgoud Atia HA, ElazeemElbnhawy RA, EzzatWM, 2017, Early diagnostic evaluation of miR-122 and miR-224 as biomarkers for hepatocellular carcinoma. Genes Dis 4:215–221. , DOI 10.1016/j.gendis.2017.10.003 Soliman B, Salem A, Ghazy M, Abu-Shahba N, El Hefnawi M, 2018, Bioinformatics functional analysis of let-7a, miR-34a, and miR-199a/b reveals novel insights into immune system pathways and cancer hallmarks for hepatocellular carcinoma. Tumour Biol 40:1010428318773675. h t t p s : / / d o i . o r g / 1 0 . 1 1 7 7 / 1010428318773675 Waly AA, El-Ekiaby N, Assal RA, AbdelrahmanMM, Hosny KA, El Tayebi HM, Esmat G, Breuhahn K, Abdelaziz AI, 2018, Methylation in MIRLET7A3 gene induces the expression of IGFII and itsmRNA binding proteins IGF2BP-2 and 3 in hepatocellular carcinoma. Front Physiol 9:1918. , DOI 10.3389/fphys. 2018.01918 Wang J, Li Y, Ding M, Zhang H, Xu X, Tang J, 2017, Molecular mechanisms and clinical applications of miR-22 in regulating malignant progression in human cancer, review). Int J Oncol 50:345– 355. , DOI 10.3892/ijo.2016.3811 Wang Y, Zeng G, Jiang Y, 2020, The emerging roles of miR-125b in cancers. Cancer Manag Res 12:1079–1088. , DOI 10. 2147/CMAR.S232388 Xue F, Yin J, Xu L, Wang B, 2017, MicroRNA-143 inhibits tumorigenesis in hepatocellular carcinoma by downregulating GATA6. Exp Ther Med 13:2667–2674. , DOI 10.3892/ etm.2017.4348 Kang H, Heo S, Shin JJ, Ji E, Tak H, Ahn S, Lee KJ, Lee EK, Kim W(2019, A miR-194/PTBP1/CCND3 axis regulates tumor growth in human hepatocellular carcinoma. J Pathol 249:395–408. https:// doi.org/10.1002/path.5325 Wang Q, Ye B, Wang P, Yao F, Zhang C, Yu G, 2019, Overview of microRNA-199a regulation in Cancer. Cancer Manag Res 11: 10327–10335. , DOI 10.2147/CMAR.S231971 Zhang T, Yang Z, Kusumanchi P, Han S, Liangpunsakul S, 2020, Critical role of microRNA-21 in the pathogenesis of liver diseases. Front Med, Lausanne, 7:7. , DOI 10.3389/fmed.2020. 00007 Chen L, Luo L, Chen W, Xu HX, Chen F, Chen LZ, Zeng WT, Chen JS, Huang XH, 2016, MicroRNA-24 increases hepatocellular carcinoma cell metastasis and invasion by targeting p53: miR-24 targeted p53. Biomed Pharmacother 84:1113–1118. , DOI 10.1016/j.biopha.2016.10.051 Ehrlich L, Hall C, Venter J, Dostal D, Bernuzzi F, Invernizzi P, Meng F, Trzeciakowski JP, Zhou T, Standeford H, Alpini G, Lairmore TC, Glaser S, 2017, miR-24 inhibition increases Menin expression and decreases Cholangiocarcinoma proliferation. Am J Pathol 187:570–580. , DOI 10.1016/j.ajpath.2016.10.021 Ji J, RongY, Luo CL, Li S, Jiang X,Weng H, Chen H, ZhangWW, Xie W, Wang FB, 2018, Up-regulation of hsa-miR-210 promotes venous metastasis and predicts poor prognosis in hepatocellular carcinoma. Front Oncol 8:569. , DOI 10.3389/fonc.2018. 00569 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2669 EP 2681 DI 10.1007/s12253-020-00870-8 PG 13 ER PT J AU Nemeth, Zs Wijker, W Lengyel, Zs Hitre, E Borbely, K AF Nemeth, Zsuzsanna Wijker, Wouter Lengyel, Zsolt Hitre, Erika Borbely, Katalin TI Metabolic Parameters as Predictors for Progression Free and Overall Survival of Patients with Metastatic Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Standardized added metabolic activity; FDG-PET/CT; Interim; Metastatic colorectal cancer; Overall survival; Progression free survival; Response ID Standardized added metabolic activity; FDG-PET/CT; Interim; Metastatic colorectal cancer; Overall survival; Progression free survival; Response AB We tested the prognostic relevance of metabolic parameters and their relative changes in patients with metastatic colorectal cancer (mCRC) treated with monoclonal antibody and chemotherapy. SUVmax (standardized uptake volume), SAM (standardized added metabolic activity) and TLG (total lesion glycolysis) are assessed with 18Ffluorodeoxyglucosepositron emission tomography and computed tomography (FDG-PET/CT) to evaluate total metabolic activity of malignant processes. Our purpose was to investigate the change of glucose metabolism in relation to PFS (progression free survival) and OS (overall survival). Fifty-three patients with mCRC with at least one measurable liver metastasis were included in this prospective, multi-center, early exploratory study. All patients were treated with firstline chemotherapy and targeted therapy. Metabolic parameters, like SUVmax, SAM, normalized SAM (NSAM) and TLG were assessed by FDG-PET/CT, carried out at baseline (scan-1) and after two therapeutic cycle (scan-2). Our results suggested neither SUVmax nor TLG have such prognostic value as NSAM in liver metastases of colorectal cancer. The parameters after the two cycles of chemotherapy proved to be better predictors of the clinical outcome. NSAM after two cycles of treatment has a statistically significant predictive value on OS, while SAM was predictive to the PFS. The follow up normalized SAM after 2 cycles of first line oncotherapy was demonstrated to be useful as prognostic biomarkers for OS in metastatic colorectal cancer. We should introduce this measurement in metastatic colorectal cancer if there is at least one metastasis in the liver. C1 [Nemeth, Zsuzsanna] Orszagos Onkologiai Intezet, „B” Belgyogyaszati-Onkologiai es Klinikai Farmakologiai Osztaly, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Wijker, Wouter] Auxiliis Pharma Ltd, Bokor utca 17, H-1037 Budapest, Hungary. [Lengyel, Zsolt] Pozitron Diagnosztika Kft, Hunyadi ut 9-11, 1117 Budapest, Hungary. [Hitre, Erika] Orszagos Onkologiai Intezet, „B” Belgyogyaszati-Onkologiai es Klinikai Farmakologiai Osztaly, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. [Borbely, Katalin] National Institute of Oncology, PET/CT Outpatient Department, Rath Gyorgy utca 7-9, 1122 Budapest, Hungary. RP Nemeth, Zs (reprint author), Orszagos Onkologiai Intezet, „B” Belgyogyaszati-Onkologiai es Klinikai Farmakologiai Osztaly, 1122 Budapest, Hungary. EM zsnemethzsuzsanna@gmail.com CR Biasco G, Derenzini E, Ercolani G, Ravaioli M, Pantaleo MA, Brandi G, 2006, Treatment of hepatic metastases from colorectal cancer: many doubts, some certainties. Cancer Treat Rev 32:214– 228. , DOI 10.1016/j.ctrv.2005.12.011 Sheth KR, Clary BM, 2005, Management of hepatic metastases from colorectal cancer. Clin Colon Rectal Surg 18:215–223. , DOI 10.1055/s-2005-916282 Adam R, De Gramont A, Figureas J, Guthrie A, Kokudo N, Kunstlinger F, Loyer E, Poston G, Rougier P, Rubbia-Brandt L, Sobrero A, Tabernero J, Teh C, Van Cutsem E, Vauthey JN of the EGOSLIM, Expert Group on OncoSurgery management of LIver Metastases, group, 2012, The oncosurgery approach to managing liver metastases from colorectal cancer: a multidisciplinary international consensus. Oncologist 17:1225–1239. , DOI 10. 1634/theoncologist.2012-0121 Bystrom P, Berglund A, Garske U, Jacobsson H, Sundin A, Nygren P, Frodin JE, Glimelius B, 2009, Early prediction of response to first-line chemotherapy by sequential [18F]-2-fluoro-2-deoxy-Dglucose positron emission tomography in patients with advanced colorectal cancer. Ann Oncol 20:1057–1061. , DOI 10. 1093/annonc/mdn744 Votrubova J, Belohlavek O, Jaruskova M, Oliverius M, Lohynska R, Trskova K, Sedlackova E, Lipska L, Stahalova V, 2006, The role of FDG-PET/CT in the detection of recurrent colorectal cancer. Eur J NuclMedMol Imaging 33:779–784. , DOI 10.1007/ s00259-006-0072-z Geus-Oei LF, Vriens D, van Laarhoven HW, van der Graaf WT, Oyen WJ, 2009, Monitoring and predicting response to therapy with 18F-FDG PET in colorectal Cancer: a systematic review. J Nucl Med 1:43S–54S. , DOI 10.2967/jnumed.108.057224 Lovinfosse P, Koopmansch B, Lambert F, Jodogne S, Kustermans G, Hatt M, Visvikis D, Seidel L, Polus M, Albert A, Delvenne P, Hustinx R, 2016,, 18)F-FDG PET/CT imaging in rectal cancer: relationship with the RAS mutational status. Br J Radiol 89(1063):20160212. , DOI 10.1259/bjr.20160212 Krikelis D, Skoura E, Kotoula V, Rondogianni P, Pianou N, Samartzis A, Xanthakis I, Fountzilas G, Datseris IE, 2014, Lack of association between KRAS mutations and 18F-FDG PET/CT in caucasian metastatic colorectal cancer patients. Anticancer Res 34: 2571–2579 Kawada K, Nakamoto Y, Kawada M, Hida K, Matsumoto T, Murakami T, Hasegawa S, Togashi K, Sakai Y, 2012, Relationship between 18F-Fluorodeoxyglucose accumulation and KRAS/BRAF mutation in colorectal cancer. Clin Can Res 18: 1696–1703. , DOI 10.1158/1078-0432.CCR-11-1909 Van Custem E, Cervantes A, Nordlinger B, Arnold D, ESMO Guidelines Working Group, 2014, Metastatic colorectal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow- up. Ann Oncol 22:229–237. , DOI 10.1093/annonc/ mdu260 Mertens J,De Bruyne S,VanDamme N, Smeets P, CeelenW, Troisi R, Laurent S, Geboes K, Peeters M, Goethals I, Van de Wiele C, 2013, Standardized added metabolic activity, SAM, IN 18F-FDG PETassessment of treatment response in colorectal livermetastases. Eur J Nucl Med Mol Imaging 40:1214–1222. , DOI 10. 1007/s00259-013-2421-z Young H, Baum R, Cremerius U, Herholz K, Hoekstra O, Lammertsma AA, Pruim J, Price P, 1999, Measurement of clinical and subclinical tumour response using, 18F)-fluorodeoxyglucose and positron emission tomography: review and 1999 EORTC recommendations. European Organization for Research and Treatment of Cancer, EORTC, PET study group. Eur J Cancer 35:1773–1782. , DOI 10.1016/s0959-8049(99)00229-4 Dempke W, Heinemann V, 2010, Ras mutational status is a biomarker for resistance to EGFR inhibitors in colorectal carcinoma. Anticancer Res 30:4673–4678 Gerber DE, 2008, Targeted therapies: a new generation of cancer treatment. Am Fam Physician 77:311–319 Buvat I, Necib H, Garcia C,Wagner A, Vanderlinden B, Emonts P, Hendlisz A, Flamen P, 2012, Lesion-based detection of early chemosensitivity using serial static FDG PET/CT in metastatic colorectal cancer. Eur J Nucl Med Mol Imaging 39:1628–1634. , DOI 10.1007/s00259-012-2172-2 Maisonobe JA, Garcia CA, Necib H, Vanderlinden B, Hendlisz A, Flamen P, Buvat I, 2013, Comparison of PET metabolic indices for the early assessment of tumour response in metastatic colorectal cancer patients treated by polychemotherapy. Eur J Nucl Med Mol Imaging 40:166–174. , DOI 10.1007/s00259-012- 2274-x Engelmann BE, Loft A, Kjaer A, Nielsen HJ, Gerds TA, Benzon EV, Brunner N, Christensen IJ, Hansson SH, Hollander NH, Kristensen MH, Lofgren J, Markova E, Sloth C, Hojgaard L, 2014, Positron emission tomography/computed tomography and biomarkers for early treatment response evaluation in metastatic colon cancer. Oncologist 19:164–172. , DOI 10.1634/ theoncologist.2013-0229 Kim DH, Kim SH, Im SA, Han SW, Goo JM, Willmann JK, Lee ES, Eo JS, Paeng JC, Han JK, Choi BI, 2012, Intermodality comparison between 3D perfusion CT and 18F-FDG PET/CT imaging for predicting early tumor response in patient with liver metastasis after chemotherapy: preliminary results of a prospective study. Eur J Radiol 81:3542–3550. , DOI 10.1016/j.ejrad.2012.02.012 Liu FY, Yen TC, Wang JY, Yang TS, 2015, Early prediction by 18F-FDG PET/CT for progression-free survival and overall survival in patients with metastatic colorectal cancer receiving third-line cetuximab-based therapy. Clin Nucl Med 40:200–205. https://doi. org/10.1097/RLU.0000000000000693 Hendlisz A, Golfinopoulos V, Garcia C, Covas A, Emonts P, Ameye L, Paesmans M, Deleporte A, Machiels G, Toussaint E, Vanderlinden B, Awada A, Piccart M, Flamen P, 2012, Serial FDG-PET/CT for early outcome prediction in patients with metastatic colorectal cancer undergoing chemotherapy. Ann Oncol 23: 1687–1693. , DOI 10.1093/annonc/mdr554 Ulanja MB, Rishi M, Beutler BD, 2019, Colon cancer sidedness, presentation, and survival at different stages. Journal of Oncology 2019:4315032. , DOI 10.1155/2019/4315032 Maus MKH, Hanna DL, Stephens C, Grimminger PP, 2013, Gene expression profiles and tumor locations in colorectal cancer, left vs. right vs. rectum). J Clin Oncol 31(15_suppl):3527–3527. https:// doi.org/10.1093/jnci/dju427 Brule SY, Jonker DJ, Karapetis CS, O’Callaghan CJ, 2015, Location of colon cancer, right-sided versus left-sided, as a prognostic factor and a predictor of benefit from cetuximab in NCIC CO.17. Eur J Cancer 51(11):1405–1414. , DOI 10.1016/j. ejca.2015.03.015 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2683 EP 2691 DI 10.1007/s12253-020-00865-5 PG 9 ER PT J AU Barragan-Perez, JE Altamirano-Vergara, EC Alvarez-Amado, ED Garcia-Beristain, CJ Chico-Ponce-de-Leon, F Gonzalez-Carranza, V Juarez-Villegas, L Murata, Ch AF Barragan-Perez, Javier Eduardo Altamirano-Vergara, Enrique Carlos Alvarez-Amado, Eduardo Daniel Garcia-Beristain, Carlos Juan Chico-Ponce-de-Leon, Fernando Gonzalez-Carranza, Vicente Juarez-Villegas, Luis Murata, Chiharu TI The Role of Time as a Prognostic Factor in Pediatric Brain Tumors: a Multivariate Survival Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Symptomatic interval; Delayed diagnosis; CNS tumors; Survival outcome ID Symptomatic interval; Delayed diagnosis; CNS tumors; Survival outcome AB There is no evidence that prolonged pre diagnostic symptomatic intervals (PSI) increases the risk of death in pediatric brain tumors. When investigating the role of time previous research had not controlled for confounding variables or measured the pretreatment interval (PTI). We use the term global delay interval (GDI) to describe the sum of PSI and PTI. The aimof this researchwas to evaluate whether there was a decrease in the probability of survival in children with brain tumors due to a prolonged PSI, PTI and GDI, using a multivariate survival analysis. We retrospective review 127 clinical records labeled with the diagnosis of CNS tumors attended at a specialized pediatric center inMexico City from January 2008 toDecember 2012. Patients with PSI and GDI diagnosed between 3 and 6 months showed statistical lower probability of surviving that those with intervals <3 months even when adjusting for age, sex, localization and tumor grade.When stratified for the place of residency and adjusted for sex, age, localization, grade of tumor, type of surgery and coadjuvant therapy, a GDI between 3 and 6 months showed to be a risk factor for the overall survival of brain tumors compared with an interval < 3 months. When analyzing the interaction, high grade tumors are at more risk of dying when GDI was between 3 and 6 months compared to <3 months. Prolonged PSI and GDI showed to be a potential prognostic factor for survival in CNS tumors, especially in high grade tumors. Future prospective research should measure the PSI, PTI and GDI and adjust for covariates in order to properly infer the effect of time in pediatric brain tumors. C1 [Barragan-Perez, Javier Eduardo] Hospital Infantil de Mexico Federico Gomez, Pediatric Neurology DepartmentMexico City, Mexico. [Altamirano-Vergara, Enrique Carlos] Hospital Infantil de Mexico Federico Gomez, Pediatric Neurology DepartmentMexico City, Mexico. [Alvarez-Amado, Eduardo Daniel] Hospital Infantil de Mexico Federico Gomez, Pediatric Neurology DepartmentMexico City, Mexico. [Garcia-Beristain, Carlos Juan] Hospital Infantil de Mexico Federico Gomez, Pediatric Neurology DepartmentMexico City, Mexico. [Chico-Ponce-de-Leon, Fernando] Hospital Infantil de Mexico Federico Gomez, Pediatric Neurosurgery DepartmentMexico City, Mexico. [Gonzalez-Carranza, Vicente] Hospital Infantil de Mexico Federico Gomez, Pediatric Neurosurgery DepartmentMexico City, Mexico. [Juarez-Villegas, Luis] Hospital Infantil de Mexico Federico Gomez, Pediatric Oncology DepartmentMexico City, Mexico. [Murata, Chiharu] Instituto Nacional de Pediatria, Research Methodology DepartmentMexico City, Mexico. RP Alvarez-Amado, ED (reprint author), Hospital Infantil de Mexico Federico Gomez, Pediatric Neurology Department, Mexico City, Mexico. EM alvarezamado@hotmail.com CR Wells EM, Packer RJ, 2015, Pediatric brain tumors. Continuum21: 373–396 Siegel RL, Miller KD, Jemal A, 2019, Cancer statistics, 2019. CA Cancer J Clin 69:7–34 Siegel RL,Miller KD, Jemal A, 2016, Cancer statistics. CA Cancer J Clin 66:7–30 Wilne S, Collier J, Kennedy C, Koller K, Grundy R, Walker D, 2007, Presentation of childhood CNS tumours: a systematic review and meta-analysis. Lancet Oncol 8:685–695 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68:394–424 Dang-Tan T, Franco EL, 2007, Diagnosis delays in childhood cancer. Cancer. 110:703–713 Crawford JR, Santi MR, Vezina G, Myseros JS, Keating RF, LaFond DA et al, 2007, CNS germ cell tumor, CNSGCT, of childhood: presentation and delayed diagnosis. Neurology. 68:1668– 1673 Patel V, McNinch NL, Rush S, 2019, Diagnostic delay and morbidity of central nervous system tumors in children and young adults: a pediatric hospital experience. J Neuro-Oncol 143:297–304 Shay V, Fattal-Valevski A, Beni-Adani L, Constantini S, 2012, Diagnostic delay of pediatric brain tumors in Israel: a retrospective risk factor analysis. Childs Nerv Syst 28:93–100 Sethi RV, Marino R, Niemierko A, Tarbell NJ, Yock TI, MacDonald SM, 2013, Delayed diagnosis in children with intracranial germ cell tumors. J Pediatr 163:1448–1453 Kukal K, Dobrovoljac M, Boltshauser E, Ammann RA, Grotzer MA, 2009, Does diagnostic delay result in decreased survival in paediatric brain tumours? Eur J Pediatr 168:303–310 Flores LE, Williams DL, Bell BA, O’Brien M, Ragab AH, 1986, Delay in the diagnosis of pediatric brain tumors. Am J Dis Child 140:684–686 Handayani K, Sitaresmi MN, Supriyadi E, Widjajanto PH, Susilawati D, Njuguna F, van de Ven PM, Kaspers GJL, Mostert S, 2016, Delays in diagnosis and treatment of childhood cancer in Indonesia. Pediatr Blood Cancer 63:2189–2196 Dobrovoljac M, Hengartner H, Boltshauser E, Grotzer M, 2002, Delay in the diagnosis of paediatric brain tumours. Eur J Pediatr 161:663–667 Molineus A, Boxberger N, Redlich A, Vorwerk P, 2013, Time to diagnosis of brain tumors in children: a single-Centre experience. Pediatr Int 55:305–309 Azizi AA, Heßler K, Leiss U, Grylli C, Chocholous M, Peyrl A, Gojo J, Slavc I, 2017, From symptom to diagnosis—the Prediagnostic symptomatic interval of pediatric central nervous system tumors in Austria. Pediatr Neurol 76:27–36 Mehta V, Chapman A,McNeely PD,Walling S, HowesWJ, 2002, Latency between symptom onset and diagnosis of pediatric brain tumors: an eastern Canadian geographic study. Neurosurgery. 51: 365–373 Stocco C, Pilotto C, Passone E, Nocerino A, Tosolini R, Pusiol A, Cogo P, 2017, Presentation and symptom interval in children with central nervous system tumors. A single-center experience. Childs Nerv Syst 33:2109–2116 Wilne S, Collier J, Kennedy C, Jenkins A, Grout J, Mackie S, Koller K, Grundy R,Walker D, 2012, Progression from first symptom to diagnosis in childhood brain tumours. Eur J Pediatr 171:87– 93 Reulecke BC, Erker CG, Fiedler BJ, Niederstadt T-U, Kurlemann G, 2008, Brain tumors in children: initial symptoms and their influence on the time span between symptom onset and diagnosis. J Child Neurol 23:178–183 Elhassan MMA, Mohamedani AA, Osman HHM, Yousif NO, Elhaj NM, Qaddoumi I, 2019, Patterns, treatments, and outcomes of pediatric central nervous system tumors in Sudan: a single institution experience. Childs Nerv Syst 35:437–444 Fukuoka K, Yanagisawa T, Suzuki T, Shirahata M, Adachi J-I, Mishima K, Fujimaki T, Matsutani M, Nishikawa R, 2014, Duration between onset and diagnosis in central nervous system tumors: impact on prognosis and functional outcome. Pediatr Int 56:829–833 Walker D,Wilne S, Grundy R, Kennedy C, Neil DA et al, 2016, A new clinical guideline from the Royal College of Paediatrics and Child Health with a national awareness campaign accelerates brain tumor diagnosis in UK children - “headSmart: Be Brain Tumour Aware”. Neuro-Oncology 18:445–454 Abdelmabood S, Kandil S, Megahed A, Fouda A, 2017, Delays in diagnosis and treatment among children with cancer: Egyptian perspective. East Mediterr Health J 23:422–429 Njuguna F, MartijnH, Langat S,Musimbi J,Muliro H, Skiles J, Vik T, Sitaresmi MN, van de Ven PM, Kaspers GJL, Mostert S, 2016, Factors influencing time to diagnosis and treatment among pediatric oncology patients in Kenya. Pediatr Hematol Oncol 33:186–199 INEGI. Encuesta Intercensal 2015. In: Instituto Nacional de Estadistica y Geografia [Internet]. 14 Mar 2015 [cited 28 Jan 2020]. Available: https://www.inegi.org.mx/programas/ intercensal/2015/ CONAPO. 31.4 por ciento de la poblacion en Mexico son ninas, ninos y adolescentes, de 0 a 17 anos. In: Consejo Nacional de Poblacion [Internet]. 30 April, 2019 [cited 28 Jan 2020]. Available: https://www.gob.mx/conapo/prensa/31-4-por-cientode- la-poblacion-en-mexico-son-ninas-ninos-y-adolescentes-de-0- a-17-anos-198293?idiom=es INEGI. Por actividad economica. In: Instituto Nacional de Estadistica y Geografia [Internet]. 2018 [cited 28 Jan 2020]. Available: https://www.inegi.org.mx/temas/pib/ CONEVAL. Medicion de Pobreza 2008-2018, Estados Unidos Mexicanos. In: Consejo Nacional de Evaluacion de la Politica de Desarrollo Social [Internet]. 2018 [cited 28 Jan 2020]. Available: https://www.coneval.org.mx/Medicion/PublishingImages/ Pobreza_2018/Serie_2008-2018.jpg Santos-Preciado JI, 2008, El Hospital Infantil de Mexico y su Boletin Medico: un hito en la historia de la pediatria mexicana. Bol Med Hosp Infant Mex 65:81–82 de Leon Castro-Sierra Eduardo FC-P, Perez Pena-DiazcontiMet al, 2006, Bol Med Hosp Infant Mex 63:367–381 Ostrom QT, Gittleman H, Truitt G, Boscia A, Kruchko C, Barnholtz-Sloan JS, 2018, CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2011–2015. Neuro Oncol 20:iv1–iv86 Rivas-Vilela S, Rubio-Casadevall J, Fabrega-Ribas A, Joly-Torta C, Vilardell L, Marcos-Gragera R, 2019, Incidence and survival of central nervous system tumors in childhood and adolescence in Girona, Spain, 1990–2013: national and international comparisons. Clin Transl Oncol 21:1177–1185. , DOI 10.1007/s12094- 019-02043-9 Desandes E, Guissou S, Chastagner P, Lacour B, 2014, Incidence and survival of children with central nervous system primitive tumors in the French National Registry of childhood solid tumors. Neuro-Oncology 16:975–983 Farinotti M, FerrariniM, Solari A, Filippini G, 1998, Incidence and survival of childhood CNS tumours in the region of Lombardy. Italy Brain 121(Pt 8):1429–1436 Magnani C, Aareleid T, Viscomi S, Pastore G, Berrino F, Oberaigner W et al, 2001, Variation in survival of children with central nervous system, CNS, malignancies diagnosed in Europe between 1978 and 1992: the EUROCARE study. Eur J Cancer 37: 711–721 Robertson PL, Zeltzer PM, Boyett JM, Rorke LB, Allen JC, Geyer JR, Stanley P, Li H, Albright AL, McGuire-Cullen P, Finlay JL, Stevens KR, Milstein JM, Packer RJ, Wisoff J, _ _ Survival and prognostic factors following radiation therapy and chemotherapy for ependymomas in children: a report of the Children’s Cancer group. J Neurosurg 1998;88: 695–703 de Araujo OL, da Trindade KM, Trompieri NM, Fontenele JB, Felix FHC, 2011, Analysis of survival and prognostic factors of pediatric patients with brain tumor. J Pediatr 87:425–432 Maria GVM, Francis ERN, Nelson UL, Gutierrez E, Gimon AV, Barboza D et al, 2013, Tumores cerebrales pediatricos experiencia de 10 anos. Revista Venezolana de Oncologia 25:85–97 Zareifar S, Rowshani F, Haghpanah S, Bordbar M, 2018, Fiveyear survival rate of children with central nervous system tumors in shiraz, Iran. Iranian Journal of Pediatric Hematology and Oncology 8:1–11 Mehrvar A, Faranoush M, Hedayati Asl AA, Tashvighi M, Fazeli MA, Qaddoumi I, Mehrvar N, Sobuti B, Jafarpour A, Ravan parsa R, Zangooei R, Alebouyeh M, Vossough P, 2014, Childhood central nervous system tumors at MAHAK’s pediatric Cancer treatment and research center, MPCTRC), Tehran. Iran Childs Nerv Syst 30:491–496 KAR, 2011, 810, Epidemiology and survival of childhood primary central nervous system malignancies in Iran: Results from a single center. Pediatr Blood Cancer 57 Grob ST, Levy JMM, 2017, Improving diagnostic and therapeutic outcomes in pediatric brain tumors. Mol Diagn Ther 22:25–39. , DOI 10.1007/s40291-017-0299-3 Pogorzala M, Styczynski J, Wysocki M, 2014, Survival and prognostic factors in children with brain tumors: long-term follow-up single center study in Poland. Anticancer Res 34:323–326 Segal D, Karajannis MA, 2016, Pediatric brain tumors: an update. Curr Probl Pediatr Adolesc Health Care 46:242–250 Rey-Casserly C, Diver T, 2019, Late effects of pediatric brain tumors. Curr Opin Pediatr 31:789–796 Chen J, Mullen CA, 2017, Patterns of diagnosis and misdiagnosis in pediatric Cancer and relationship to survival. J Pediatr Hematol Oncol 39:e110–e115 Qaddoumi I, Merchant TE, Boop FA, Gajjar A, 2019, Diagnostic delay in children with central nervous system tumors and the need to improve education. J Neuro-Oncol 145:591–592 Ferrari A, Lo Vullo S, Giardiello D, Veneroni L, Magni C, Clerici CA, Chiaravalli S, Casanova M, Luksch R, Terenziani M, Spreafico F, Meazza C, Catania S, Schiavello E, Biassoni V, Podda M, Bergamaschi L, Puma N, Massimino M, Mariani L, 2016, The sooner the better? How symptom interval correlates with outcome in children and adolescents with solid tumors: regression tree analysis of the findings of a prospective study. Pediatr Blood Cancer 63:479–485 Gerber NU, von Hoff K, von Bueren AO, Treulieb W, Deinlein F, Benesch M, Zwiener I, Soerensen N,Warmuth-Metz M, Pietsch T, Mittler U, Kuehl J, Kortmann RD, Grotzer MA, Rutkowski S, 2012, A long duration of the prediagnostic symptomatic interval is not associated with an unfavourable prognosis in childhood medulloblastoma. Eur J Cancer 48:2028–2036 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2693 EP 2701 DI 10.1007/s12253-020-00875-3 PG 9 ER PT J AU Kabalak, AP Kizilgoz, D Kavurgaci, S Demirci, YN Yilmaz, Ak, G Metintas, S Metintas, M Demirag, F Yilmaz, AF Kabalak, Akin Pinar Kizilgoz, Derya Kavurgaci, Suna Demirci, Yilmaz Nilgun Yilmaz, Senay Ak, Guntulu Metintas, Selma Metintas, Muzaffer Demirag, Funda Yilmaz, Ulku TI The Clinical Impact of Re-biopsies in Lung Adenocarcinoma: a Retrospective Multicenter Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Non-small cell lung cancer; Re-biopsy; Targeted therapy ID Non-small cell lung cancer; Re-biopsy; Targeted therapy AB If a patient’s cancer progresses while undergoing targeted therapy, a re-biopsy is notmandatory. But when evaluating the benefits and risks on a case-by-case basis (transformation to small cell, assessing for a clinical trial), physicians should inform patients about the possible need for a re-biopsy (5). This was a retrospective and multicentre study. A total of 644 patients with lung adenocarcinoma were reviewed, 625 of whom were ruled eligible. From them, 399 were found to show disease progression, and 126 re-biopsies were performed. Progression status, re-biopsy sites, success of obtaining adequate tissue, molecular patterns after re-biopsy and subsequent treatments were analysed. Survival differences among patients with disease progression were then examined according to re-biopsy status. Overall, 625 patients with adenocarcinoma and a median age of 61.4 were evaluated. Initial tyrosine kinase inhibitor (TKI) usage numbered 37 patients (5.9%). Progression was diagnosed in 399 (63.8%) patients, out of which 26 (31.6%) underwent re-biopsies. The successful number of re-biopsies was 103 (81.7%). No complications were observed after any of the biopsy procedures. Subsequent treatments were changed in 15 patients (11.9%), who began new TKI treatments. Poor performance status was the most common reason for not performing a biopsy (n = 65; 23.8%), followed by the physician’s decision (n = 40; 14.6%). Re-biopsies can demonstrate the new characteristics of a tumour and can detect the activation of pre-existing clones, making possible new treatment opportunities for patients. According to the performance status of the patient and the availability of the progressive lesion, we should increase the rate of re-biopsies before the decision to follow up with the best supportive care. C1 [Kabalak, Akin Pinar] Ataturk Chest Disease and Thoracic Surgery Teaching and Research Hospital, Department of Chest Disease, 06290 Ankara, Turkey. [Kizilgoz, Derya] Ataturk Chest Disease and Thoracic Surgery Teaching and Research Hospital, Department of Chest Disease, 06290 Ankara, Turkey. [Kavurgaci, Suna] Ataturk Chest Disease and Thoracic Surgery Teaching and Research Hospital, Department of Chest Disease, 06290 Ankara, Turkey. [Demirci, Yilmaz Nilgun] Gazi University School of Medicine, Department of Chest DiseaseAnkara, Turkey. [Yilmaz, Senay] Eskisehir Osmangazi University, Lung and Pleural Cancers Research and Clinical CenterEskisehir, Turkey. [Ak, Guntulu] Eskisehir Osmangazi University, Lung and Pleural Cancers Research and Clinical CenterEskisehir, Turkey. [Metintas, Selma] Eskisehir Osmangazi University, Lung and Pleural Cancers Research and Clinical CenterEskisehir, Turkey. [Metintas, Muzaffer] Eskisehir Osmangazi University, Lung and Pleural Cancers Research and Clinical CenterEskisehir, Turkey. [Demirag, Funda] Ankara Ataturk Training and Research Hospital, 1st Pathology ClinicAnkara, Turkey. [Yilmaz, Ulku] Ataturk Chest Disease and Thoracic Surgery Teaching and Research Hospital, Department of Chest Disease, 06290 Ankara, Turkey. RP Kabalak, AP (reprint author), Ataturk Chest Disease and Thoracic Surgery Teaching and Research Hospital, Department of Chest Disease, 06290 Ankara, Turkey. EM pinarakinn@yahoo.com CR Ettinger DS, Aisner DL, Wood DE, Akerley W, Bauman J, Chang JY et al, 2018 Jul, NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 5.2018. J Natl Compr Canc Netw 16(7): 807–821 Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH et al, 2018 Mar, Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. Arch Pathol Lab Med 142(3):321–346 HuangD,KimDW, Kotsakis A,Deng S, Lira P,Ho SNet al, 2013, Multiplexed deep sequencing analysis of ALK kinase domain identifies resistance mutations in relapsed patients following crizotinib treatment. Genomics 102(3):157–162 Vavala T, Novello S, 2018 Aug)Alectinib in the treatment of ALKpositive non-small cell lung cancer: an update on its properties, efficacy, safety and place in therapy. Ther Adv Med Oncol 3:10: 1758835918789364 Besse B, Adjei A, Baas P, Meldgaard P, Nicolson M, Paz-Ares L et al, 2014, 2nd ESMO Consensus Conference on Lung Cancer: non-small-cell lung cancer first-line/second and further lines of treatment in advanced disease. Ann Oncol 25(8):1475-84 Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L et al, 2000, New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 92: 205–216 Thunnissen E, 2012 Dec, Pulmonary adenocarcinoma histology. Transl Lung Cancer Res 1(4):276–279 Rossi G, Pelosi G, Graziano P, BarbareschiM, Papotti M(2009, A reevaluation of the clinical significance of histological subtyping of non–small-cell lung carcinoma: Diagnostic algorithms in the era of personalized treatments. Int J Surg Pathol 17(3):206–18 Kawamura T, Kenmotsu H, Taira T, Omori S, Nakashima K, Wakuda K et al, 2016 Jul, Rebiopsy for patients with non-smallcell lung cancer after epidermal growth factor receptor-tyrosine kinase inhibitor failure. Cancer Sci 107(7):1001–1005 Nosaki K, Inamasu E, Shimamatsu S, Yoshida T, Toyokawa G, Hirai F et al. Re-biopsy in advanced non-small cell lung cancer patients after the development of 3rd generation EGFR-TKI and new targeted therapies. , DOI 10.1200/jco.2015.33.15_ suppl.e19081 Yoon HJ, Lee HY, Lee KS, Choi YL, Ahn MJ, Park K et al, 2012 Dec, Repeat biopsy for mutational analysis of non-small cell lung cancers resistant to previous chemotherapy: adequacy and complications. Radiology 265(3):939–948 Thunnissen E, Kerr KM, Herth FJV, Lantuejoul S, Papotti M, Rintoul RC et al, 2012, The challenge of NSCLC diagnosis and predictive analysis on small samples. Practical approach of a working group. Lung Cancer 76:1–18 Yatabe Y, 2010, EGFR mutations and the terminal respiratory unit. Cancer Metastasis Rev 29:23–36 Jekunen AP, 2015, Role of rebiopsy in relapsed non-small cell lung cancer for directing oncology treatments. J Oncol 2015:809835 Hata A, Katakami N, Nanjo S, Okuda C, Kaji R, Imai Y. Rebiopsy of Histological Samples in Pretreated Non-small Cell Lung Cancer: Comparison Among Rebiopsy Procedures. In Vivo. 2017 31(3): 475–479 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2703 EP 2708 DI 10.1007/s12253-020-00876-2 PG 6 ER PT J AU Bernreuther, Ch Daghigh, F Moller, K Hube-Magg, C Lennartz, M Lutz, F Rico, DS Fraune, Ch Dum, D Luebke, A Eichenauer, T Moller-Koop, Ch Schlomm, Th Wittmer, C Huland, H Heinzer, H Graefen, M Haese, A Burandt, E Tsourlakis, ChM Clauditz, T Hoflmayer, D Izbicki, J Simon, R Sauter, G Minner, S Steurer, S Meiners, J AF Bernreuther, Christian Daghigh, Ferdous Moller, Katharina Hube-Magg, Claudia Lennartz, Maximilian Lutz, Florian Rico, Dwertmann Sebastian Fraune, Christoph Dum, David Luebke, M. Andreas Eichenauer, Till Moller-Koop, Christina Schlomm, Thorsten Wittmer, Corinna Huland, Hartwig Heinzer, Hans Graefen, Markus Haese, Alexander Burandt, Eike Tsourlakis, Christina Maria Clauditz, S. Till Hoflmayer, Doris Izbicki, R. Jakob Simon, Ronald Sauter, Guido Minner, Sarah Steurer, Stefan Meiners, Jan TI Secreted Frizzled-Related Protein 4 (SFRP4) Is an Independent Prognostic Marker in Prostate Cancers Lacking TMPRSS2: ERG Fusions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SFRP4; Prognosis; Prostate cancer; TMA ID SFRP4; Prognosis; Prostate cancer; TMA AB Secreted frizzled-related protein 4 (SFRP4) controlsWNT signaling and is thought to play a role for tumor aggressiveness. Here, we analyzed a tissue microarray containing 11,152 prostate cancers with pathological, clinical and molecular data by immunohistochemistry. SFRP4 expression was higher in cancer than in non-neoplastic acinar cells. SFRP4 staining was seen in 64.9%of tumors and classified as weak in 33.2%, moderate in 23.9% and strong in 7.8% of cancers. SFRP4 overexpression was linked to advanced tumor stage, high classical/quantitative Gleason grade (p < 0.0001 each), lymph node metastasis (p = 0.0002), and a positive surgical margin (p = 0.0017). SFRP4 positivity was markedly more frequent in ERG positive (77.4%) than in ERG negative cancers (57.4% p < 0.0001). Subset analyses in 2725 cancers with and 3592 cancers without TMPRSS2:ERG fusion revealed that associations with tumor phenotype and patient outcome were largely driven by the subset of ERG negative tumors. In a multivariate analysis including various postoperative and prognostic clinico-pathological features, SFRP4 protein expression emerged as an independent prognostic parameter in ERG negative cancers. SFRP4 immunostaining was significantly linked with 10 of 11 previously analyzed chromosomal deletions (p < 0.05 each). In conclusion, high SFRP4 immunostaining is associated with poor prognosis and genomic instability in ERG negative prostate cancers. C1 [Bernreuther, Christian] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Daghigh, Ferdous] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Moller, Katharina] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Hube-Magg, Claudia] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Lennartz, Maximilian] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Lutz, Florian] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Rico, Dwertmann Sebastian] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Fraune, Christoph] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Dum, David] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Luebke, M. Andreas] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Eichenauer, Till] University Medical Center Hamburg-Eppendorf, Department of UrologyHamburg, Germany. [Moller-Koop, Christina] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Schlomm, Thorsten] Charite-Universitatsmedizin Berlin, Department of UrologyBerlin, Germany. [Wittmer, Corinna] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Huland, Hartwig] Martini-Clinic, Prostate Cancer Center Hamburg-EppendorfHamburg, Germany. [Heinzer, Hans] Martini-Clinic, Prostate Cancer Center Hamburg-EppendorfHamburg, Germany. [Graefen, Markus] Martini-Clinic, Prostate Cancer Center Hamburg-EppendorfHamburg, Germany. [Haese, Alexander] Martini-Clinic, Prostate Cancer Center Hamburg-EppendorfHamburg, Germany. [Burandt, Eike] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Tsourlakis, Christina Maria] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Clauditz, S. Till] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Hoflmayer, Doris] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Izbicki, R. Jakob] University Medical Center Hamburg-Eppendorf, General, Visceral and Thoracic Surgery DepartmentHamburg, Germany. [Simon, Ronald] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Sauter, Guido] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Minner, Sarah] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Steurer, Stefan] University Medical Center Hamburg Eppendorf, Pathology, Martinistr, 52, 20246 Hamburg, Germany. [Meiners, Jan] University Medical Center Hamburg-Eppendorf, General, Visceral and Thoracic Surgery DepartmentHamburg, Germany. RP Simon, R (reprint author), University Medical Center Hamburg Eppendorf, Pathology, 20246 Hamburg, Germany. EM R.Simon@uke.de CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6):394–424. , DOI 10.3322/caac. 21492 Melkonyan HS, Chang WC, Shapiro JP, Mahadevappa M, Fitzpatrick PA, Kiefer MC, Tomei LD, Umansky SR, 1997, SARPs: a family of secreted apoptosis-related proteins. Proc Natl Acad Sci U S A 94(25):13636–13641 Chien AJ, Conrad WH, Moon RT, 2009, A Wnt survival guide: from flies to human disease. J Invest Dermatol 129(7):1614–1627. , DOI 10.1038/jid.2008.445 Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson A, Kampf C, Sjostedt E, Asplund A, Olsson I, Edlund K, Lundberg E, Navani S, Szigyarto CA, Odeberg J, Djureinovic D, Takanen JO, Hober S, Alm T, Edqvist PH, Berling H, Tegel H,Mulder J, Rockberg J, Nilsson P, Schwenk JM, Hamsten M, von Feilitzen K, Forsberg M, Persson L, Johansson F, Zwahlen M, von Heijne G, Nielsen J, Ponten F, 2015, Proteomics. Tissue-based map of the human proteome. Science 347(6220):1260419. , DOI 10.1126/science. 1260419 Fox SA, Richards AK, Kusumah I, Perumal V, Bolitho EM, Mutsaers SE, Dharmarajan AM, 2013, Expression profile and function ofWnt signaling mechanisms in malignant mesothelioma cells. Biochem Biophys Res Commun 440(1):82–87. https://doi. org/10.1016/j.bbrc.2013.09.025 Jacob F, Ukegjini K, Nixdorf S, Ford CE, Olivier J, Caduff R, Scurry JP, Guertler R, Hornung D, Mueller R, Fink DA, Hacker NF, Heinzelmann-Schwarz VA, 2012, Loss of secreted frizzledrelated protein 4 correlates with an aggressive phenotype and predicts poor outcome in ovarian cancer patients. PLoS One 7(2): e31885. , DOI 10.1371/journal.pone.0031885 Ford CE, Jary E, Ma SS, Nixdorf S, Heinzelmann-Schwarz VA, Ward RL, 2013, TheWnt gatekeeper SFRP4 modulates EMT, cell migration and downstreamWnt signalling in serous ovarian cancer cells. PLoS One 8(1):e54362. , DOI 10.1371/journal.pone. 0054362 Kamposioras K, Konstantara A, Kotoula V, Lakis S, Kouvatseas G, Akriviadis E, Vrettou E, Dionysopoulos D, Krikelis D, Papadopoulou K, Charalambous E, Chrisafi S, Konstantaras C, Fountzilas G, 2013, The prognostic significance of WNT pathway in surgically-treated colorectal cancer: beta-catenin expression predicts for disease-free survival. Anticancer Res 33(10):4573–4584 Huang D, Yu B, Deng Y, Sheng W, Peng Z, Qin W, Du X, 2010, SFRP4 was overexpressed in colorectal carcinoma. J Cancer Res Clin Oncol 136(3):395–401. , DOI 10.1007/s00432-009- 0669-2 Abu-Jawdeh G, Comella N, Tomita Y, Brown LF, Tognazzi K, Sokol SY, Kocher O, 1999, Differential expression of frpHE: a novel human stromal protein of the secreted frizzled gene family, during the endometrial cycle and malignancy. Lab Investig 79(4): 439–447 Chung MT, Sytwu HK, Yan MD, Shih YL, Chang CC, Yu MH, Chu TY, Lai HC, Lin YW, 2009, Promoter methylation of SFRPs gene family in cervical cancer. Gynecol Oncol 112(2):301–306. , DOI 10.1016/j.ygyno.2008.10.004 Urakami S, Shiina H, Enokida H, Kawakami T, Kawamoto K, Hirata H, Tanaka Y, Kikuno N, Nakagawa M, Igawa M, Dahiya R, 2006, Combination analysis of hypermethylatedWnt-antagonist family genes as a novel epigenetic biomarker panel for bladder cancer detection. Clin Cancer Res 12(7 Pt 1):2109–2116. https:// doi.org/10.1158/1078-0432.CCR-05-2468 Bu XM, Zhao CH, Zhang N, Gao F, Lin S, Dai XW, 2008, Hypermethylation and aberrant expression of secreted frizzledrelated protein genes in pancreatic cancer. World J Gastroenterol 14(21):3421–3424 Surana R, Sikka S, CaiW, Shin EM,Warrier SR, Tan HJ, Arfuso F, Fox SA, Dharmarajan AM, Kumar AP, 2014, Secreted frizzled related proteins: implications in cancers. Biochim Biophys Acta 1845(1):53–65. , DOI 10.1016/j.bbcan.2013.11.004 Pohl S, Scott R, Arfuso F, Perumal V, Dharmarajan A, 2015, Secreted frizzled-related protein 4 and its implications in cancer and apoptosis. Tumour Biol 36(1):143–152. , DOI 10. 1007/s13277-014-2956-z Horvath LG, Henshall SM, Kench JG, Saunders DN, Lee CS, Golovsky D, Brenner PC, O'Neill GF, Kooner R, Stricker PD, Grygiel JJ, Sutherland RL, 2004, Membranous expression of secreted frizzled-related protein 4 predicts for good prognosis in localized prostate cancer and inhibits PC3 cellular proliferation in vitro. Clin Cancer Res 10(2):615–625 Horvath LG, Lelliott JE, Kench JG, Lee CS, Williams ED, Saunders DN, Grygiel JJ, Sutherland RL, Henshall SM, 2007, Secreted frizzled-related protein 4 inhibits proliferation and metastatic potential in prostate cancer. Prostate 67(10):1081–1090. , DOI 10.1002/pros.20607 MortensenMM, Hoyer S, Lynnerup AS, Orntoft TF, Sorensen KD, BorreM, Dyrskjot L, 2015, Expression profiling of prostate cancer tissue delineates genes associated with recurrence after prostatectomy. Sci Rep 5:16018. , DOI 10.1038/srep16018 Sandsmark E, Andersen MK, Bofin AM, Bertilsson H, Drablos F, Bathen TF, RyeMB, TessemMB, 2017, SFRP4 gene expression is increased in aggressive prostate cancer. Sci Rep 7(1):14276. https:// doi.org/10.1038/s41598-017-14622-3 Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, Kaushik P, Cerami E, Reva B, Antipin Y,Mitsiades N, Landers T, Dolgalev I, Major JE, Wilson M, Socci ND, Lash AE, Heguy A, Eastham JA, Scher HI, Reuter VE, Scardino PT, Sander C, Sawyers CL, Gerald WL, 2010, Integrative genomic profiling of human prostate cancer. Cancer Cell 18(1):11–22. , DOI 10.1016/j.ccr.2010.05.026 Wang Y, Xia XQ, Jia Z, Sawyers A, Yao H, Wang-Rodriquez J, Mercola D,McClellandM(2010, In silico estimates of tissue components in surgical samples based on expression profiling data. Cancer Res 70(16):6448–6455. , DOI 10.1158/0008- 5472.CAN-10-0021 Jia Z, Wang Y, SawyersA, Yao H, Rahmatpanah F, Xia XQ,Xu Q, Pio R, Turan T, Koziol JA, Goodison S, Carpenter P, Wang- Rodriguez J, Simoneau A, Meyskens F, Sutton M, Lernhardt W, Beach T, Monforte J, McClelland M, Mercola D, 2011, Diagnosis of prostate cancer using differentially expressed genes in stroma. Cancer Res 71(7):2476–2487. , DOI 10.1158/0008-5472. CAN-10-2585 Chen X, Xu S, McClelland M, Rahmatpanah F, Sawyers A, Jia Z, Mercola D, 2012, An accurate prostate cancer prognosticator using a seven-gene signature plus Gleason score and taking cell type heterogeneity into account. PLoS One 7(9):e45178. https://doi. org/10.1371/journal.pone.0045178 Erho N, Crisan A, Vergara IA, Mitra AP, Ghadessi M, Buerki C, Bergstralh EJ, Kollmeyer T, Fink S, Haddad Z, Zimmermann B, Sierocinski T, Ballman KV, Triche TJ, Black PC, Karnes RJ, Klee G, Davicioni E, Jenkins RB, 2013, Discovery and validation of a prostate cancer genomic classifier that predicts early metastasis following radical prostatectomy. PLoS One 8(6):e66855. https://doi. org/10.1371/journal.pone.0066855 Klein EA, CooperbergMR, Magi-Galluzzi C, Simko JP, Falzarano SM, Maddala T, Chan JM, Li J, Cowan JE, Tsiatis AC, Cherbavaz DB, Pelham RJ, Tenggara-Hunter I, Baehner FL, Knezevic D, Febbo PG, Shak S, Kattan MW, Lee M, Carroll PR, 2014, A 17- gene assay to predict prostate cancer aggressiveness in the context of Gleason grade heterogeneity, tumor multifocality, and biopsy undersampling. Eur Urol 66(3):550–560. , DOI 10.1016/j. eururo.2014.05.004 Schlomm T, Iwers L, Kirstein P, Jessen B, Kollermann J,Minner S, Passow-Drolet A, Mirlacher M, Milde-Langosch K, Graefen M, Haese A, Steuber T, Simon R, Huland H, Sauter G, Erbersdobler A, 2008, Clinical significance of p53 alterations in surgically treated prostate cancers. Mod Pathol 21(11):1371–1378. , DOI 10.1038/modpathol.2008.104 Sauter G, Steurer S, Clauditz TS, Krech T, Wittmer C, Lutz F, Lennartz M, Janssen T, Hakimi N, Simon R, von Petersdorff- Campen M, Jacobsen F, von Loga K, Wilczak W, Minner S, Tsourlakis MC, Chirico V, Haese A, Heinzer H, Beyer B, Graefen M, Michl U, Salomon G, Steuber T, Budaus LH, Hekeler E, Malsy-Mink J, Kutzera S, Fraune C, Gobel C, Huland H, Schlomm T, 2016, Clinical utility of quantitative Gleason grading in prostate biopsies and prostatectomy specimens. Eur Urol 69(4): 592–598. , DOI 10.1016/j.eururo.2015.10.029 Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP, 1998, Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4(7):844–847 Tennstedt P, Koster P, Bruchmann A, Mirlacher M, Haese A, Steuber T, Sauter G, Huland H, Graefen M, Schlomm T, Minner S, Simon R, 2012, The impact of the number of cores on tissue microarray studies investigating prostate cancer biomarkers. Int J Oncol 40(1):261–268. , DOI 10.3892/ijo.2011.1216 Minner S, Enodien M, Sirma H, Luebke AM, Krohn A,Mayer PS, Simon R, Tennstedt P, Muller J, Scholz L, Brase JC, Liu AY, Schluter H, Pantel K, Schumacher U, Bokemeyer C, Steuber T, Graefen M, Sauter G, Schlomm T, 2011, ERG status is unrelated to PSA recurrence in radically operated prostate cancer in the absence of antihormonal therapy. Clin Cancer Res 17(18):5878–5888. , DOI 10.1158/1078-0432.CCR-11-1251 Minner S,Wittmer C, GraefenM, Salomon G, Steuber T, Haese A, Huland H, Bokemeyer C, Yekebas E, Dierlamm J, Balabanov S, Kilic E, Wilczak W, Simon R, Sauter G, Schlomm T, 2011, High level PSMA expression is associated with early PSA recurrence in surgically treated prostate cancer. Prostate 71(3):281–288. https:// doi.org/10.1002/pros.21241 Krohn A, Seidel A, Burkhardt L, Bachmann F,MaderM, Grupp K, Eichenauer T, Becker A, AdamM, GraefenM, Huland H, Kurtz S, Steurer S, Tsourlakis MC, Minner S, Michl U, Schlomm T, Sauter G, Simon R, Sirma H, 2013, Recurrent deletion of 3p13 targets multiple tumour suppressor genes and defines a distinct subgroup of aggressive ERG fusion-positive prostate cancers. J Pathol 231(1):130–141. , DOI 10.1002/path.4223 Burkhardt L, Fuchs S, Krohn A, Masser S, Mader M, Kluth M, Bachmann F, Huland H, Steuber T, Graefen M, Schlomm T, Minner S, Sauter G, Sirma H, Simon R, 2013, CHD1 is a 5q21 tumor suppressor required for ERG rearrangement in prostate cancer. Cancer Res 73(9):2795–2805. , DOI 10.1158/ 0008-5472.CAN-12-1342 Kluth M, Hesse J, Heinl A, Krohn A, Steurer S, Sirma H, Simon R, Mayer PS, Schumacher U, Grupp K, Izbicki JR, Pantel K,Dikomey E, Korbel JO, Plass C, Sauter G, Schlomm T, Minner S, 2013, Genomic deletion of MAP3K7 at 6q12-22 is associated with early PSA recurrence in prostate cancer and absence of TMPRSS2:ERG fusions. Mod Pathol 26:975–983. , DOI 10.1038/ modpathol.2012.236 Kluth M, Amschler NN, Galal R, Moller-Koop C, Barrow P, Tsourlakis MC, Jacobsen F, Hinsch A, Wittmer C, Steurer S, Krech T, Buscheck F, Clauditz TS, Beyer B, Wilczak W, Graefen M, Huland H, Minner S, Schlomm T, Sauter G, Simon R, 2017, Deletion of 8p is an independent prognostic parameter in prostate cancer. Oncotarget 8(1):379–392. , DOI 10.18632/ oncotarget.13425 Krohn A, Diedler T, Burkhardt L, Mayer PS, De Silva C, Meyer- Kornblum M, Kotschau D, Tennstedt P, Huang J, Gerhauser C, Mader M, Kurtz S, Sirma H, Saad F, Steuber T, Graefen M, Plass C, Sauter G, Simon R, Minner S, Schlomm T, 2012, Genomic deletion of PTEN is associated with tumor progression and early PSA recurrence in ERG fusion-positive and fusion-negative prostate cancer. Am J Pathol 181(2):401–412. , DOI 10.1016/j. ajpath.2012.04.026 Kluth M, Ahrary R, Hube-Magg C, AhmedM, Volta H, Schwemin C, Steurer S,Wittmer C,WilczakW, Burandt E, Krech T, AdamM, Michl U, Heinzer H, Salomon G, Graefen M, Koop C, Minner S, Simon R, Sauter G, Schlomm T, 2015, Genomic deletion of chromosome 12p is an independent prognostic marker in prostate cancer. Oncotarget 6(29):27966–27979. , DOI 10.18632/ oncotarget.4626 Weischenfeldt J, Simon R, Feuerbach L, Schlangen K,Weichenhan D, Minner S, Wuttig D, Warnatz HJ, Stehr H, Rausch T, Jager N, Gu L, Bogatyrova O, Stutz AM, Claus R, Eils J, Eils R, Gerhauser C, Huang PH, Hutter B, Kabbe R, Lawerenz C, Radomski S, Bartholomae CC, Falth M, Gade S, Schmidt M, Amschler N, Hass T, Galal R, Gjoni J, Kuner R, Baer C, Masser S, von Kalle C, Zichner T, Benes V, Raeder B, MaderM, Amstislavskiy V, Avci M, Lehrach H, Parkhomchuk D, Sultan M, Burkhardt L, Graefen M, Huland H, Kluth M, Krohn A, Sirma H, Stumm L, Steurer S, Grupp K, Sultmann H, Sauter G, Plass C, Brors B, Yaspo ML, Korbel JO, Schlomm T, 2013, Integrative genomic analyses reveal an androgen-driven somatic alteration landscape in early-onset prostate cancer. Cancer Cell 23(2):159–170. , DOI 10. 1016/j.ccr.2013.01.002 Kluth M, Scherzai S, Buschek F, Fraune C, Moller K, Hoflmayer D, Minner S, Gobel C, Moller-Koop C, Hinsch A, Neubauer E, Tsourlakis MC, Sauter G, Heinzer H, Graefen M, Wilczak W, Luebke AM, Burandt E, Steurer S, Schlomm T, Simon R, 2018, 13q deletion is linked to an adverse phenotype and poor prognosis in prostate cancer. Genes, chromosomes & cancer 57(10):504–512. , DOI 10.1002/gcc.22645 Kluth M, Jung S, Habib O, Eshagzaiy M, Heinl A, Amschler N, Masser S, Mader M, Runte F, Barow P, Frogh S, Omari J, Moller- Koop C, Hube-Magg C, Weischenfeldt J, Korbel J, Steurer S, Krech T, Huland H, Graefen M, Minner S, Sauter G, Schlomm T, Simon R, 2017, Deletion lengthening at chromosomes 6q and 16q targets multiple tumor suppressor genes and is associated with an increasingly poor prognosis in prostate cancer. Oncotarget 8(65): 108923–108935. , DOI 10.18632/oncotarget.22408 Kluth M, Harasimowicz S, Burkhardt L, Grupp K, Krohn A, Prien K,Gjoni J,Hass T,Galal R,GraefenM, Haese A, Simon R,Huhne- Simon J, Koop C, Korbel J, Weischenfeld J, Huland H, Sauter G, Quaas A, Wilczak W, Tsourlakis MC, Minner S, Schlomm T, 2014, Clinical significance of different types of p53 gene alteration in surgically treated prostate cancer. Int J Cancer 135(6):1369– 1380. , DOI 10.1002/ijc.28784 Kluth M, Graunke M, Moller-Koop C, Hube-Magg C, Minner S, Michl U, Graefen M, Huland H, Pompe R, Jacobsen F, Hinsch A, Wittmer C, Lebok P, Steurer S, Buscheck F, Clauditz T, Wilczak W, Sauter G, Schlomm T, Simon R, 2016, Deletion of 18q is a strong and independent prognostic feature in prostate cancer. Oncotarget 7(52):86339–86349. , DOI 10.18632/ oncotarget.13404 Grupp K, Roettger L, Kluth M, Hube-Magg C, Simon R, Lebok P, Minner S, TsourlakisMC, Koop C, Graefen M, Adam M, Haese A, Wittmer C, SauterG,WilczakW, Huland H, SchlommT, Steurer S, Krech T, 2015, Expression of DNA ligase IV is linked to poor prognosis and characterizes a subset of prostate cancers harboring TMPRSS2:ERG fusion and PTEN deletion. Oncol Rep 34(3): 1211–1220. , DOI 10.3892/or.2015.4080 Wissmann C,Wild PJ, Kaiser S, Roepcke S, Stoehr R,Woenckhaus M, Kristiansen G, Hsieh JC, Hofstaedter F, Hartmann A, Knuechel R, Rosenthal A, Pilarsky C, 2003, WIF1, a component of the Wnt pathway, is down-regulated in prostate, breast, lung, and bladder cancer. J Pathol 201(2):204–212. , DOI 10.1002/path.1449 Perez C, Castillo M, Alemany L, Tous S, Klaustermeier J, de Sanjose S, Velasco J, 2014, Evaluation of p16(INK4a, overexpression in a large series of cervical carcinomas: concordance with SPF10-LiPA25 PCR. Int J Gynecol Pathol 33(1):74–82. https:// doi.org/10.1097/PGP.0b013e3182774546 Ross J, Busch J, Mintz E, Ng D, Stanley A, Brafman D, Sutton VR, Van den Veyver I, Willert K, 2014, A rare human syndrome provides genetic evidence that WNT signaling is required for reprogramming of fibroblasts to induced pluripotent stem cells. Cell Rep 9(5):1770–1780. , DOI 10.1016/j.celrep.2014. 10.049 Aoki K, Aoki M, Sugai M, Harada N, Miyoshi H, Tsukamoto T, Mizoshita T, Tatematsu M, Seno H, Chiba T, OshimaM,Hsieh CL, Taketo MM, 2007, Chromosomal instability by beta-catenin/TCF transcription in APC or beta-catenin mutant cells. Oncogene 26(24):3511–3520. , DOI 10.1038/sj.onc.1210141 Fodde R, Smits R, Clevers H, 2001, APC, signal transduction and genetic instability in colorectal cancer. Nat Rev Cancer 1(1):55–67. , DOI 10.1038/35094067 Hadjihannas MV, Behrens J, 2006, CIN by WNT: growth pathways, mitotic control and chromosomal instability in cancer. Cell Cycle 5(18):2077–2081. , DOI 10.4161/cc.5.18.3282 Uren A, Reichsman F, Anest V, Taylor WG, Muraiso K, Bottaro DP, Cumberledge S, Rubin JS, 2000, Secreted frizzled-related protein- 1 binds directly to wingless and is a biphasic modulator of Wnt signaling. J Biol Chem 275(6):4374–4382. , DOI 10.1074/ jbc.275.6.4374 Brase JC, Johannes M, Mannsperger H, Falth M, Metzger J, Kacprzyk LA, Andrasiuk T, Gade S, Meister M, Sirma H, Sauter G, Simon R, SchlommT, Beissbarth T, Korf U, Kuner R, Sultmann H, 2011, TMPRSS2-ERG -specific transcriptional modulation is associated with prostate cancer biomarkers and TGF-beta signaling. BMC Cancer 11:507. , DOI 10.1186/1471-2407-11-507 Tomlins SA, Laxman B, Varambally S, Cao X, Yu J, Helgeson BE, Cao Q, Prensner JR, Rubin MA, Shah RB, Mehra R, Chinnaiyan AM, 2008, Role of the TMPRSS2-ERG gene fusion in prostate cancer. Neoplasia 10(2):177–188 Kunderfranco P,Mello-Grand M, Cangemi R, Pellini S, Mensah A, Albertini V, Malek A, Chiorino G, Catapano CV, Carbone GM, 2010, ETS transcription factors control transcription of EZH2 and epigenetic silencing of the tumor suppressor gene Nkx3.1 in prostate cancer. PLoS One 5(5):e10547. , DOI 10.1371/ journal.pone.0010547 Yuting L. JQ, 2018, Sa1621 - Ezh2 Regulates Sfrp4 Expression without Affecting the Hypermethylation of Sfrp4 in Colorectal Cancer Cell Line. American Gastrointestinal Association 154, 6). , DOI 10.1016/S0016-5085(18)31445-8 Wu L, Zhao JC, Kim J, Jin HJ, Wang CY, Yu J, 2013, ERG is a critical regulator of Wnt/LEF1 signaling in prostate cancer. Cancer Res 73(19):6068–6079. , DOI 10.1158/0008-5472.CAN- 13-0882 Chakravarthi B, Chandrashekar DS, Hodigere Balasubramanya SA, Robinson AD, Carskadon S, Rao U, Gordetsky J, Manne U, Netto GJ, Sudarshan S, Palanisamy N, Varambally S, 2018, Wnt receptor frizzled 8 is a target of ERG in prostate cancer. Prostate 78(16):1311–1320. , DOI 10.1002/pros.23704 Gupta S, Iljin K, Sara H, Mpindi JP, Mirtti T, Vainio P, Rantala J, Alanen K, Nees M, Kallioniemi O, 2010, FZD4 as a mediator of ERG oncogene-induced WNT signaling and epithelial-tomesenchymal transition in human prostate cancer cells. Cancer Res 70(17):6735–6745. , DOI 10.1158/0008-5472.CAN- 10-0244 He Y, Hooker E, Yu EJ, Wu H, Cunha GR, Sun Z, 2018, An indispensable role of androgen receptor in Wnt responsive cells during prostate development, maturation, and regeneration. Stem Cells 36(6):891–902. , DOI 10.1002/stem.2806 Burdelski C, Bujupi E, Tsourlakis MC, Hube-Magg C, Kluth M, Melling N, Lebok P, Minner S, Koop C, Graefen M, Heinzer H, Wittmer C, Sauter G,WilczakW, Simon R, Schlomm T, Steurer S, Krech T, 2015, Loss of SOX9 expression is associated with PSA recurrence in ERG-positive and PTEN deleted prostate cancers. PLoS One 10(6):e0128525. , DOI 10.1371/journal.pone. 0128525 Grupp K, Hohne TS, Prien K, Hube-Magg C, Tsourlakis MC, Sirma H, Pham T, Heinzer H, Graefen M, Michl U, Simon R, Wilczak W, Izbicki J, Sauter G, Minner S, Schlomm T, Steurer S, 2013, Reduced CD147 expression is linked to ERG fusionpositive prostate cancers but lacks substantial impact on PSA recurrence in patients treated by radical prostatectomy. Exp Mol Pathol 95(2):227–234. , DOI 10.1016/j.yexmp.2013.08. 002 Burdelski C, Dieckmann T, Heumann A, Hube-Magg C, Kluth M, Beyer B, Steuber T, Pompe R, Graefen M, Simon R, Minner S, Tsourlakis MC, Koop C, Izbicki J, Sauter G, Krech T, Schlomm T, Wilczak W, Lebok P, 2016, p16 upregulation is linked to poor prognosis in ERG negative prostate cancer. Tumour Biol 37(9): 12655–12663. , DOI 10.1007/s13277-016-5167-y Grupp K, Boumesli R, TsourlakisMC, Koop C,Wilczak W, Adam M, Sauter G, Simon R, Izbicki JR, GraefenM, Huland H, Steurer S, Schlomm T, Minner S, Quaas A, 2014, The prognostic impact of high Nijmegen breakage syndrome, NBS1, gene expression in ERG-negative prostate cancers lacking PTEN deletion is driven by KPNA2 expression. Int J Cancer 135(6):1399–1407. https:// doi.org/10.1002/ijc.28778 Heumann A, Kaya O, Burdelski C, Hube-Magg C, Kluth M, Lang DS, Simon R, Beyer B, Thederan I, Sauter G, Izbicki JR, Luebke AM, Hinsch A, Jacobsen F,Wittmer C, Buscheck F, Hoflmayer D, Minner S, Tsourlakis MC, Schlomm T, Wilczak W, 2017, Up regulation and nuclear translocation of Y-box binding protein 1, YB-1, is linked to poor prognosis in ERG-negative prostate cancer. Sci Rep 7(1):2056. , DOI 10.1038/s41598-017-02279- x Burdelski C, Kleinhans S, Kluth M, Hube-Magg C, Minner S, Koop C, Graefen M, Heinzer H, Tsourlakis MC, Wilczak W, Marx A, Sauter G, Wittmer C, Huland H, Simon R, Schlomm T, Steurer S, 2016, Reduced AZGP1 expression is an independent predictor of early PSA recurrence and associated with ERGfusion positive and PTEN deleted prostate cancers. Int J Cancer 138(5):1199–1206. , DOI 10.1002/ijc.29860 Melling N, Thomsen E, Tsourlakis MC, Kluth M, Hube-Magg C, Minner S, Koop C, Graefen M, Heinzer H, Wittmer C, Sauter G, Wilczak W, Huland H, Simon R, Schlomm T, Steurer S, Krech T, 2015, Overexpression of enhancer of zeste homolog 2, EZH2, characterizes an aggressive subset of prostate cancers and predicts patient prognosis independently from pre- and postoperatively assessed clinicopathological parameters. Carcinogenesis 36(11): 1333–1340. , DOI 10.1093/carcin/bgv137 Burdelski C, Reiswich V, Hube-Magg C, KluthM, Minner S,Koop C, Graefen M, Heinzer H, Tsourlakis MC, Wittmer C, Huland H, Simon R, Schlomm T, Sauter G, Steurer S, 2015, Cytoplasmic accumulation of Sequestosome 1, p62, is a predictor of biochemical recurrence, rapid tumor cell proliferation, and genomic instability in prostate Cancer. Clin Cancer Res 21(15):3471–3479. https://doi. org/10.1158/1078-0432.CCR-14-0620 Eichenauer T, Hussein M, Hube-Magg C, Kluth M, Buscheck F, Hoflmayer D, TsourlakisMC, Steurer S, Clauditz TS, Luebke AM, Burandt E, Wilczak W, Hinsch A, Dum D, Beyer B, Steuber T, Huland H, GraefenM, Simon R, Sauter G, Melling N, Schlomm T, Minner S, 2019, A nuclear shift of GSK3beta protein is an independent prognostic factor in prostate cancer. Oncotarget 10(18): 1729–1744. , DOI 10.18632/oncotarget.26739 Heinrich MC, Gobel C, Kluth M, Bernreuther C, Sauer C, Schroeder C, Moller-Koop C, Hube-Magg C, Lebok P, Burandt E, Sauter G, Simon R, Huland H, Graefen M, Heinzer H, Schlomm T, Heumann A, 2018, PSCA expression is associated with favorable tumor features and reduced PSA recurrence in operated prostate cancer. BMC Cancer 18(1):612. , DOI 10. 1186/s12885-018-4547-7 Berinstein NL, McNamara M, Nguyen A, Egan J,Wolf GT, 2018, Increased immune infiltration and chemokine receptor expression in head and neck epithelial tumors after neoadjuvant immunotherapy with the IRX-2 regimen. Oncoimmunology 7(5):e1423173. , DOI 10.1080/2162402X.2017.1423173 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2709 EP 2722 DI 10.1007/s12253-020-00861-9 PG 14 ER PT J AU Uzelac, B Krivokuca, A Brankovic-Magic, M Magic, Z Susnjar, S Milovanovic, Z Supic, G AF Uzelac, Bojana Krivokuca, Ana Brankovic-Magic, Mirjana Magic, Zvonko Susnjar, Snezana Milovanovic, Zorka Supic, Gordana TI Expression of SIRT1, SIRT3 and SIRT6 Genes for Predicting Survival in Triple-Negative and Hormone Receptor-Positive Subtypes of Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Sirtuins; Gene expression; Triple-negative breast Cancer; Hormone receptor-positive breast Cancer; Survival ID Sirtuins; Gene expression; Triple-negative breast Cancer; Hormone receptor-positive breast Cancer; Survival AB Triple-negative breast cancer (TNBC) is characterized by aggressive phenotype and a poorer prognosis compared to the estrogen and progesterone receptor positive, Her2 negative (ER + PR + Her2-) breast cancer. Increasing evidence suggests that sirtuins, a family of histone deacetylases, could have an important role in aggressiveness of TNBC’s. The current study evaluated the potential clinical relevance of SIRT1, SIRT3 and SIRT6 gene expressions in two prognostically distinctive subtypes of breast cancer, the most aggressive TNBC and the least aggressive ER + PR + Her2- tumors. Total RNAs were isolated from 48 TNBC and 63 ER + PR + Her2- tumor samples. Relative gene expression was determined by SYBR Green RT-PCR and delta-delta Ct method, normalized to GAPDH.Mean gene expression of both SIRT1 and SIRT3 was significantly lower in the TNBC compared to ER + PR + Her2- tumors (p = 0.0001). Low SIRT1 and SIRT6 expressions associatedwith worse overall survival in ER + PR + Her2- patients (p = 0.039, p = 0.006, respectively), while TNBC patients with high SIRT1 tend to have a poor prognosis (p = 0.057). In contrast, high expression of SIRT3 in TNBC patients associated with higher histological grade (p = 0.027) and worse overall survival (p = 0.039). The Cox regression analysis revealed that low SIRT1 expression could be an independent prognostic marker of poor survival in ER + PR + Her2- breast cancers (HR = 11.765, 95% CI:1.234–100, p = 0.033). Observed differential expression of SIRT1, SIRT3 and SIRT6 genes in TNBC and ER + PR + Her2- subtypes, with opposite effects on patients’ survival, suggests context-dependent mechanisms underlying aggressiveness of breast cancer. Further investigations are necessary to evaluate sirtuins as potential biomarkers and therapeutic targets in breast cancer. C1 [Uzelac, Bojana] Military Medical Academy, Institute for Medical Research, Crnotravska 17, 11 000 Belgrade, Serbia. [Krivokuca, Ana] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. [Brankovic-Magic, Mirjana] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. [Magic, Zvonko] Military Medical Academy, Institute for Medical Research, Crnotravska 17, 11 000 Belgrade, Serbia. [Susnjar, Snezana] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. [Milovanovic, Zorka] Institute of Oncology and Radiology of Serbia, Pasterova 14, 11 000 Belgrade, Serbia. [Supic, Gordana] Military Medical Academy, Institute for Medical Research, Crnotravska 17, 11 000 Belgrade, Serbia. RP Supic, G (reprint author), Military Medical Academy, Institute for Medical Research, 11 000 Belgrade, Serbia. EM gogasupic@gmail.com CR Fallahpour S, Navaneelan T, De P, Borgo A, 2017, Breast cancer survival by molecular subtype: a population-based analysis of cancer registry data. CMAJ Open 5(3):E734–E739 CarafaV, Altucci L, NebbiosoA, 2019, Dual tumor suppressor and tumor promoter action of Sirtuins in determining malignant phenotype. Front Pharmacol 10:38 JinM-S, Hyun CL, Park IA, Kim JY, Chung YR, Im S-A, Lee KH, Moon HG, Ryu HS, 2016, SIRT1 induces tumor invasion by targeting epithelial mesenchymal transition-related pathway and is a prognostic marker in triple negative breast cancer. Tumor Biol 37(4):4743–4753 Yeung F, Hoberg JE, Ramsey CS, KellerMD, Jones DR, Frye RA, MayoMW(2004, Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J 23(12): 2369–2380 Yi YW, Kang HJ, Kim HJ, Kong Y, Brown ML, Bae I, 2013, Targeting mutant p53 by a SIRT1 activator YK-3-237 inhibits the proliferation of triple-negative breast Cancer cells. Oncotarget 4(7): 984–994 Wang C, Yang W, Dong F, Guo Y, Tan J, Ruan S, Huang T, 2017, The prognostic role of Sirt1 expression in solid malignancies: a meta-analysis. Oncotarget 8(39):66343–66351 Bosch-Presegue L, Vaquero A, 2011, The dual role of sirtuins in cancer. Genes Cancer 2(6):648–662 Cao Y-W, Li W-Q, Wan G-X, Li Y-X, Du X-M, Li Y-C et al, 2014, Correlation and prognostic value of SIRT1 and Notch1 signaling in breast cancer. J Exp Clin Cancer Res CR 33:97 Chung YR, Kim H, Park SY, Park IA, Jang JJ, Choe J-Y, Jung YY, Im SA, Moon HG, Lee KH, Suh KJ, Kim TY, Noh DY, Han W, Ryu HS, 2015, Distinctive role of SIRT1 expression on tumor invasion and metastasis in breast cancer by molecular subtype. Hum Pathol 46(7):1027–1035 Jeong SM, Haigis MC, 2015, Sirtuins in Cancer: a balancing act between genome stability and metabolism. Mol Cells 38(9):750– 758 Ansari A, Rahman MS, Saha SK, Saikot FK, Deep A, Kim K, 2017, Function of the SIRT3 mitochondrial deacetylase in cellular physiology, cancer, and neurodegenerative disease. Aging Cell 16(1):4–16 He S, He C, Yuan H, Xiong S, Xiao Z, Chen L, 2014, The SIRT 3 expression profile is associated with pathological and clinical outcomes in human breast cancer patients. Cell Physiol Biochem Int J Exp Cell Physiol Biochem Pharmacol 34(6):2061–2069 Huang N, Liu Z, Zhu J, Cui Z, Li Y, Yu Y, Sun F, Pan Q, Yang Q, 2017, Sirtuin 6 plays an oncogenic role and induces cell autophagy in esophageal cancer cells. Tumor Biol 39(6):1010428317708532 KhongkowM, Olmos Y, Gong C, Gomes AR,Monteiro LJ, Yague E, Cavaco TB, Khongkow P, Man EPS, Laohasinnarong S, Koo CY, Harada-Shoji N, Tsang JWH, Coombes RC, Schwer B, Khoo US, Lam EWF, 2013, SIRT6 modulates paclitaxel and epirubicin resistance and survival in breast cancer. Carcinogenesis 34(7): 1476–1486 Deng C-X, 2009, SIRT1, is it a tumor promoter or tumor suppressor? Int J Biol Sci 5(2):147–152 Rifai K, Judes G, Idrissou M, Daures M, Bignon Y-J, Penault- Llorca F, Bernard-GallonD, 2017, Dual SIRT1 expression patterns strongly suggests its bivalent role in human breast cancer. Oncotarget 8(67):110922–110930 Lee J-J, Lee HJ, Son B-H, Kim S-B, Ahn J-H, Ahn SD, Cho EY, Gong G, 2016, Expression of FOXM1 and related proteins in breast cancer molecular subtypes. Int J Exp Pathol 97(2):170–177 Zhang S, Chen P, Huang Z, Hu X, Chen M, Hu S, Hu Y, Cai T, 2015, Sirt7 promotes gastric cancer growth and inhibits apoptosis by epigenetically inhibiting miR-34a. Sci Rep 5:9787. https://doi. org/10.1038/srep09787 Hajian-Tilaki K, 2013, Receiver operating characteristic, ROC, curve analysis for medical diagnostic test evaluation. Casp J Intern Med 4(2):627–635 Kim H, Kwon YM, Kim JS, Han J, Shim YM, Park J, Kim DH, 2006, Elevated mRNA levels of DNA methyltransferase-1 as an independent prognostic factor in primary nonsmall cell lung cancer. Cancer 107(5):1042–1049 Chung SY, Jung YY, Park IA, Kim H, Chung YR, Kim JY, Park SY, Im SA, Lee KH,Moon HG, Noh DY, HanW, Lee C, Kim TY, Ryu HS, 2016, Oncogenic role of SIRT1 associated with tumor invasion, lymph node metastasis, and poor disease-free survival in triple negative breast cancer. Clin Exp Metastasis 33(2):179–185 Desouki MM, Doubinskaia I, Gius D, Abdulkadir SA, 2014, Decreasedmitochondrial SIRT3 expression is a potentialmolecular biomarker associated with poor outcome in breast cancer. Hum Pathol 45(5):1071–1077 Wilking MJ, Ahmad N, 2015, The role of SIRT1 in Cancer. Am J Pathol 185(1):26–28 Simic P,Williams EO, Bell EL, Gong JJ, Bonkowski M, Guarente L, 2013, SIRT1 suppresses the epithelial-to-Mesenchymal transition in Cancer metastasis and organ fibrosis. Cell Rep 3(4):1175– 1186. , DOI 10.1016/j.celrep.2013.03.019 Yuan J, Minter-Dykhouse K, Lou Z, 2009, A c-Myc–SIRT1 feedback loop regulates cell growth and transformation. J Cell Biol 185(2):203–211 Elangovan S, Ramachandran S, Venkatesan N, Ananth S, Gnana- Prakasam JP, Martin PM, Browning DD, Schoenlein PV, Prasad PD, Ganapathy V, Thangaraju M, 2011, SIRT1 is essential for oncogenic signaling by estrogen/estrogen receptor α in breast cancer. Cancer Res 71(21):6654–6664 Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, Pandita TK et al, 2001, hSIR2(SIRT1, functions as an NAD-dependent p53 deacetylase. Cell 107(2):149–159 Wu M,WeiW, Xiao X, Guo J, Xie X, Li L, Kong Y, Lv N, Jia W, Zhang Y, Xie X, 2012, Expression of SIRT1 is associated with lymph node metastasis and poor prognosis in both operable triplenegative and non-triple-negative breast cancer. Med Oncol 29(5): 3240–3249 Santolla MF, Avino S, Pellegrino M, De Francesco EM, De Marco P, Lappano R et al, 2015, SIRT1 is involved in oncogenic signaling mediated by GPER in breast cancer. Cell Death Dis 6:e1834 Torrens-Mas M, Pons DG, Sastre-Serra J, Oliver J, Roca P, 2017, SIRT3 silencing sensitizes breast Cancer cells to cytotoxic treatments through an increment in ROS production. J Cell Biochem 118(2):397–406 Bae JS, Park S-H, Jamiyandorj U, Kim KM, Noh SJ, Kim JR, Park HJ, Kwon KS, Jung SH, Park HS, Park BH, Lee H, Moon WS, Sylvester KG, Jang KY, 2016, CK2α/CSNK2A1 phosphorylates SIRT6 and is involved in the progression of breast carcinoma and predicts shorter survival of diagnosed patients. Am J Pathol 186(12):3297–3315 Igci M, Kalender ME, Borazan E, Bozgeyik I, Bayraktar R, Bozgeyik E, Camci C, Arslan A, 2016, High-throughput screening of Sirtuin family of genes in breast cancer. Gene. 586(1):123–128 Wang D, Li C, Zhang X, 2014, The promoter methylation status and mRNA expression levels of CTCF and SIRT6 in sporadic breast cancer. DNA Cell Biol 33(9):581–590 Ioris RM, Galie M, Ramadori G, Anderson JG, Charollais A, Konstantinidou G, Brenachot X, Aras E, Goga A, Ceglia N, Sebastian C, Martinvalet D, Mostoslavsky R, Baldi P, Coppari R, 2017, SIRT6 suppresses Cancer stem-like capacity in tumors with PI3K activation independently of its Deacetylase activity. Cell Rep 18(8):1858–1868 Thirumurthi U, Shen J, Xia W, LaBaff AM, Wei Y, Li C-W, et al., 2014, MDM2-mediated degradation of SIRT6 phosphorylated by AKT1 promotes tumorigenesis and trastuzumab resistance in breast cancer. Sci signal. 7(336):ra71 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2723 EP 2731 DI 10.1007/s12253-020-00873-5 PG 9 ER PT J AU Ozkan, SH Ugurlu, UM Yumuk, FP Kaya, H AF Ozkan, Sahin Hulya Ugurlu, Umit Mustafa Yumuk, Fulden Perran Kaya, Handan TI Prognostic Role of Immune Markers in Triple Negative Breast Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Triple negative breast cancer; Tumor infiltrating lymphocytes; Tumor associated macrophages; PD-L1; Fork head box protein 3; Immunohistochemistry ID Triple negative breast cancer; Tumor infiltrating lymphocytes; Tumor associated macrophages; PD-L1; Fork head box protein 3; Immunohistochemistry AB Tumor immune microenvironment (TIME) is a significant prognostic parameter for triple negative breast carcinomas (TNBC) due to being a target for immunotherapeutic agents and its essential role during the cancer immunoediting process. In this study, CD8, FOXP3, CD163, PD-L1/SP142 and PD-L1/SP263 antibodies were examined in a sample of 51 TNBC cases. Patients who received neoadjuvant therapy were excluded. CD8, FOXP3 and CD163 antibodies were evaluated separately in intratumoral area (ITA) and tumor stroma (TS). PD-L1 status was also examined in tumor cells (TC) and immune cells (IC) using both SP142 and SP263 antibodies. In multivariate Cox regressions, the only antibody that was found to be significantly associated with survival was SP142. SP142-positivity in TC and IC was related to increased overall survival. Higher CD163 expression in ITA and SP263-positivity in IC were associated with younger age. Lymphatic/angioinvasion was more frequent in cases with negative/ low CD8 and FOXP3 expressions. Moreover, metastatic axillary lymph node(s) was associated with negative/low FOXP3 expression in TS. CD8, FOXP3, CD163, SP142 and SP263 expressions were positively correlated with each other, except a mild discordance caused by CD163 in ITA. Although PD-L1 status with both SP142 and SP263 antibodies were concordant in the majority of cases, 33.3% and 13.7% of the cases showed SP142-negative/SP263-positive pattern in TC and IC respectively. In conclusion, we suggest that composition, density and localization of the immune cells and the check point molecules are important prognostic parameters in TNBC. Immunohistochemistry can be used as an accessible and less expensive tool to demonstrate TIME. C1 [Ozkan, Sahin Hulya] Marmara University, School of Medicine, Department of PathologyIstanbul, Turkey. [Ugurlu, Umit Mustafa] Marmara University, School of Medicine, Department of General SurgeryIstanbul, Turkey. [Yumuk, Fulden Perran] Marmara University School of Medicine, Department of Medical OncologyIstanbul, Turkey. [Kaya, Handan] Marmara University, School of Medicine, Department of PathologyIstanbul, Turkey. RP Ozkan, SH (reprint author), Marmara University, School of Medicine, Department of Pathology, Istanbul, Turkey. EM drhulyasahin@gmail.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68(6):394–424. , DOI 10.3322/caac. 21492 Lehmann BD, Pietenpol JA, 2014, Identification and use of biomarkers in treatment strategies for triple-negative breast cancer subtypes. J Pathol 232(2):142–150. , DOI 10.1002/path.4280 Sporikova Z, Koudelakova V, Trojanec R, Hajduch M, 2018, Genetic markers in triple-negative breast Cancer. Clinical breast cancer 18(5):e841–e850. , DOI 10.1016/j.clbc.2018.07. 023 Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, Lickley LA, Rawlinson E, Sun P, Narod SA, 2007, Triplenegative breast cancer: clinical features and patterns of recurrence. Clinical cancer research : an official journal of the American Association for Cancer Research 13, 15 Pt 1):4429–4434., DOI , DOI 10.1158/1078-0432.CCR-06-3045 Chacon RD, Costanzo MV, 2010, Triple-negative breast cancer. Breast cancer research : BCR 12 Suppl 2:S3., DOI , DOI 10. 1186/bcr2574 Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V, 2007, Descriptive analysis of estrogen receptor, ER)-negative, progesterone receptor, PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a populationbased study from the California cancer registry. Cancer 109(9): 1721–1728. , DOI 10.1002/cncr.22618 Carey LA, Perou CM, Livasy CA, Dressler LG, Cowan D, Conway K, Karaca G, Troester MA, Tse CK, Edmiston S, Deming SL, Geradts J, Cheang MC, Nielsen TO, Moorman PG, Earp HS, Millikan RC, 2006, Race, breast cancer subtypes, and survival in the Carolina breast Cancer study. Jama 295(21):2492–2502. https:// doi.org/10.1001/jama.295.21.2492 Hubalek M, Czech T, Muller H, 2017, Biological subtypes of triple-negative breast Cancer. Breast care 12(1):8–14. https://doi. org/10.1159/000455820 Badve S, Dabbs DJ, Schnitt SJ, Baehner FL, Decker T, Eusebi V, Fox SB, Ichihara S, Jacquemier J, Lakhani SR, Palacios J, Rakha EA, Richardson AL, Schmitt FC, Tan PH, Tse GM, Weigelt B, Ellis IO, Reis-Filho JS, 2011, Basal-like and triple-negative breast cancers: a critical review with an emphasis on the implications for pathologists and oncologists.Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 24(2):157–167. , DOI 10.1038/modpathol.2010.200 Mittal D, Gubin MM, Schreiber RD, Smyth MJ, 2014, New insights into cancer immunoediting and its three component phases– elimination, equilibriumand escape. Curr Opin Immunol 27:16–25. , DOI 10.1016/j.coi.2014.01.004 Schreiber RD, Old LJ, Smyth MJ, 2011, Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 331(6024):1565–1570. , DOI 10.1126/science. 1203486 Dunn GP, Old LJ, Schreiber RD, 2004, The immunobiology of cancer immunosurveillance and immunoediting. Immunity 21(2): 137–148. , DOI 10.1016/j.immuni.2004.07.017 Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehner FL, Penault-Llorca F, Perez EA, Thompson EA, SymmansWF, Richardson AL, Brock J, Criscitiello C, Bailey H, Ignatiadis M, Floris G, Sparano J, Kos Z, Nielsen T, Rimm DL, Allison KH, Reis-Filho JS, Loibl S, Sotiriou C, Viale G, Badve S, Adams S, Willard-Gallo K, Loi S, International TWG(2015, The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: recommendations by an international TILs working group 2014. Annals of oncology : official journal of the European Society for Medical Oncology 26(2):259– 271. , DOI 10.1093/annonc/mdu450 Castaneda CA, Mittendorf E, Casavilca S, Wu Y, Castillo M, Arboleda P, Nunez T, Guerra H, Barrionuevo C, Dolores-Cerna K, Belmar-Lopez C, Abugattas J, Calderon G, De La Cruz M, Cotrina M, Dunstan J, Gomez HL, Vidaurre T, 2016, Tumor infiltrating lymphocytes in triple negative breast cancer receiving neoadjuvant chemotherapy. World journal of clinical oncology 7(5): 387–394. , DOI 10.5306/wjco.v7.i5.387 Ladoire S, Martin F, Ghiringhelli F, 2011, Prognostic role of FOXP3+ regulatory T cells infiltrating human carcinomas: the paradox of colorectal cancer. Cancer immunology, immunotherapy : CII 60(7):909–918. , DOI 10.1007/s00262-011-1046-y Adams TA, Vail PJ, Ruiz A,MollaeeM,McCue PA, Knudsen ES, Witkiewicz AK, 2018, Composite analysis of immunological and metabolic markers defines novel subtypes of triple negative breast cancer. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 31(2):288–298. https:// doi.org/10.1038/modpathol.2017.126 Caldwell C Jr, Johnson CE, Balaji VN, Balaji GA, Hammer RD, Kannan R, 2017, Identification and validation of a PD-L1 binding peptide for determination of PDL1 expression in tumors. Sci Rep 7(1):13682. , DOI 10.1038/s41598-017-10946-2 Francisco LM, Sage PT, Sharpe AH, 2010, The PD-1 pathway in tolerance and autoimmunity. Immunol Rev 236:219–242. https:// doi.org/10.1111/j.1600-065X.2010.00923.x Beckers RK, Selinger CI, Vilain R, Madore J, Wilmott JS, Harvey K, Holliday A, Cooper CL, Robbins E, Gillett D, Kennedy CW, Gluch L, Carmalt H, Mak C,Warrier S, Gee HE, Chan C, McLean A, Walker E, McNeil CM, Beith JM, Swarbrick A, Scolyer RA, O'Toole SA(2016, Programmed death ligand 1 expression in triplenegative breast cancer is associated with tumour-infiltrating lymphocytes and improved outcome. Histopathology 69(1):25–34. , DOI 10.1111/his.12904 Keir ME, Butte MJ, Freeman GJ, Sharpe AH, 2008, PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 26:677– 704. , DOI 10.1146/annurev.immunol.26.021607.090331 Atezolizumab Combo Approved for PD-L1-positive TNBC, 2019). Cancer discovery 9, 5):OF2., DOI , DOI 10.1158/2159- 8290.CD-NB2019-038 Schmid P, Rugo HS, Adams S, Schneeweiss A, Barrios CH, Iwata H, Dieras V, Henschel V, Molinero L, Chui SY, Maiya V, Husain A, Winer EP, Loi S, Emens LA, Investigators IM, 2020, Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer, IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet Oncology 21(1):44–59. , DOI 10.1016/S1470- 2045(19)30689-8 Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, Fitzgibbons PL, Francis G, Goldstein NS, Hayes M, Hicks DG, Lester S, Love R, Mangu PB, McShane L, Miller K, Osborne CK, Paik S, Perlmutter J, Rhodes A, Sasano H, Schwartz JN, Sweep FC, Taube S, Torlakovic EE, Valenstein P, Viale G, Visscher D, Wheeler T, Williams RB, Wittliff JL, Wolff AC, 2010, American Society of Clinical Oncology/college of American pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 28(16):2784–2795. , DOI 10.1200/JCO.2009.25.6529 Hida AI, Sagara Y, Yotsumoto D, Kanemitsu S, Kawano J, Baba S, Rai Y, Oshiro Y, Aogi K, Sagara Y, Ohi Y, 2016, Prognostic and predictive impacts of tumor-infiltrating lymphocytes differ between triple-negative and HER2-positive breast cancers treated with standard systemic therapies. Breast Cancer Res Treat 158(1):1–9. , DOI 10.1007/s10549-016-3848-2 Hida AI, Ohi Y, 2015, Evaluation of tumor-infiltrating lymphocytes in breast cancer; proposal of a simpler method. Annals of oncology : official journal of the European Society for Medical Oncology 26(11):2351. , DOI 10.1093/annonc/mdv363 Ohtani H, Mori-Shiraishi K, Nakajima M, Ueki H, 2015, Defining lymphocyte-predominant breast cancer by the proportion of lymphocyte-rich stroma and its significance in routine histopathological diagnosis. Pathol Int 65(12):644–651. , DOI 10. 1111/pin.12355 Herrero-Vicent C, Guerrero A, Gavila J, Gozalbo F, Hernandez A, Sandiego S, Algarra MA, Calatrava A, Guillem-Porta V, Ruiz- Simon A, 2017, Predictive and prognostic impact of tumourinfiltrating lymphocytes in triple-negative breast cancer treated with neoadjuvant chemotherapy. Ecancermedicalscience 11:759. https:// doi.org/10.3332/ecancer.2017.759 O'Loughlin M, Andreu X, Bianchi S, Chemielik E, Cordoba A, Cserni G, Figueiredo P, Floris G, Foschini MP, Heikkila P, Kulka J, Liepniece-Karele I, Regitnig P, Reiner A, Ryska A, Sapino A, Shalaby A, Stovgaard ES, Quinn C, Walsh EM, Zolota V, Glynn SA, Callagy G, 2018, Reproducibility and predictive value of scoring stromal tumour infiltrating lymphocytes in triple-negative breast cancer: a multi-institutional study. Breast Cancer Res Treat 171(1):1–9. , DOI 10.1007/s10549-018-4825-8 Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, Budczies J, Huober J, Klauschen F, Furlanetto J, Schmitt WD, Blohmer JU, Karn T, Pfitzner BM, Kummel S, Engels K, Schneeweiss A, Hartmann A, Noske A, Fasching PA, Jackisch C, van Mackelenbergh M, Sinn P, Schem C, Hanusch C, Untch M, Loibl S, 2018, Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. The Lancet Oncology 19(1):40–50. , DOI 10.1016/S1470- 2045(17)30904-X Stanton SE, Disis ML, 2016, Clinical significance of tumorinfiltrating lymphocytes in breast cancer. Journal for immunotherapy of cancer 4:59. , DOI 10.1186/s40425-016-0165-6 Tiainen S, Tumelius R, Rilla K, Hamalainen K, Tammi M, Tammi R, Kosma VM, Oikari S, Auvinen P, 2015, High numbers of macrophages, especially M2-like, CD163-positive), correlate with hyaluronan accumulation and poor outcome in breast cancer. Histopathology 66(6):873–883. , DOI 10.1111/his.12607 Yang M, Li Z, Ren M, Li S, Zhang L, Zhang X, Liu F, 2018, Stromal infiltration of tumor-associated macrophages conferring poor prognosis of patients with basal-like breast carcinoma. J Cancer 9(13):2308–2316. , DOI 10.7150/jca.25155 Engebraaten O, Vollan HKM, Borresen-Dale AL, 2013, Triplenegative breast cancer and the need for new therapeutic targets. Am J Pathol 183(4):1064–1074. , DOI 10.1016/j.ajpath. 2013.05.033 Plasilova ML, Hayse B, Killelea BK, Horowitz NR, Chagpar AB, Lannin DR, 2016, Features of triple-negative breast cancer: analysis of 38,813 cases from the national cancer database. Medicine 95(35):e4614. , DOI 10.1097/MD.0000000000004614 Burstein MD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SA, Savage MI, Osborne CK, Hilsenbeck SG, Chang JC, Mills GB, Lau CC, Brown PH, 2015, Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clinical cancer research : an official journal of the American Association for Cancer Research 21(7):1688–1698. , DOI 10.1158/1078-0432.CCR-14-0432 Kim S, Moon BI, Lim W, Park S, Cho MS, Sung SH, 2018, Feasibility of classification of triple negative breast Cancer by Immunohistochemical surrogate markers. Clinical breast cancer 18(5):e1123–e1132. , DOI 10.1016/j.clbc.2018.03.012 Le Du F, Eckhardt BL, Lim B, Litton JK, Moulder S, Meric- Bernstam F, Gonzalez-Angulo AM, Ueno NT, 2015, Is the future of personalized therapy in triple-negative breast cancer based on molecular subtype? Oncotarget 6, 15):12890-12908. Doi:https:// doi.org/10.18632/oncotarget.3849 Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, Pietenpol JA, 2011, Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 121(7):2750–2767. https://doi. org/10.1172/JCI45014 Yam C, Mani SA, Moulder SL, 2017, Targeting the molecular subtypes of triple negative breast Cancer: understanding the diversity to Progress the field. Oncologist 22(9):1086–1093. https://doi. org/10.1634/theoncologist.2017-0095 Perou CM, 2011, Molecular stratification of triple-negative breast cancers. Oncologist 16(Suppl 1):61–70. , DOI 10.1634/ theoncologist.2011-S1-61 Lehmann BD, Jovanovic B, Chen X, Estrada MV, Johnson KN, Shyr Y, Moses HL, Sanders ME, Pietenpol JA, 2016, Refinement of triple-negative breast Cancer molecular subtypes: implications for Neoadjuvant chemotherapy selection. PLoS One 11(6): e0157368. , DOI 10.1371/journal.pone.0157368 Leon-Ferre RA, Polley MY, Liu H, Gilbert JA, Cafourek V, Hillman DW, Elkhanany A, Akinhanmi M, Lilyquist J, Thomas A, Negron V, Boughey JC, Liu MC, Ingle JN, Kalari KR, Couch FJ, Visscher DW, Goetz MP, 2018, Impact of histopathology, tumor-infiltrating lymphocytes, and adjuvant chemotherapy on prognosis of triple-negative breast cancer. Breast Cancer Res Treat 167(1):89–99. , DOI 10.1007/s10549-017-4499-7 Loi S, Drubay D, Adams S, Pruneri G, Francis PA, Lacroix-Triki M, Joensuu H, Dieci MV, Badve S, Demaria S, Gray R, Munzone E, Lemonnier J, Sotiriou C, Piccart MJ, Kellokumpu-Lehtinen PL, Vingiani A, Gray K, Andre F, Denkert C, Salgado R, Michiels S, 2019, Tumor-infiltrating lymphocytes and prognosis: a pooled individual patient analysis of early-stage triple-negative breast cancers. Journal of clinical oncology : official journal of the American Society of Clinical Oncology:JCO1801010., DOI , DOI 10. 1200/JCO.18.01010 Matsumoto H, Thike AA, Li H, Yeong J, Koo SL, Dent RA, Tan PH, Iqbal J, 2016, Increased CD4 and CD8-positive T cell infiltrate signifies good prognosis in a subset of triple-negative breast cancer. Breast Cancer Res Treat 156(2):237–247. , DOI 10.1007/ s10549-016-3743-x Baker K, Lachapelle J, Zlobec I, Bismar TA, Terracciano L, Foulkes WD, 2011, Prognostic significance of CD8+ T lymphocytes in breast cancer depends upon both oestrogen receptor status and histological grade. Histopathology 58(7):1107–1116. https:// doi.org/10.1111/j.1365-2559.2011.03846.x Liu S, Lachapelle J, Leung S, Gao D, Foulkes WD, Nielsen TO, 2012, CD8+ lymphocyte infiltration is an independent favorable prognostic indicator in basal-like breast cancer. Breast cancer research : BCR 14(2):R48. , DOI 10.1186/bcr3148 Mao Y, Qu Q, Chen X, Huang O, Wu J, Shen K, 2016, The prognostic value of tumor-infiltrating lymphocytes in breast Cancer: a systematic review and meta-analysis. PLoS One 11(4): e0152500. , DOI 10.1371/journal.pone.0152500 Chen Z, Chen X, Zhou E, Chen G, Qian K,Wu X,Miao X, Tang Z, 2014, Intratumoral CD8(+, cytotoxic lymphocyte is a favorable prognostic marker in node-negative breast cancer. PLoS One 9(4): e95475. , DOI 10.1371/journal.pone.0095475 Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH, Ellis IO, Green AR, 2011, Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 29(15):1949–1955. , DOI 10.1200/JCO.2010. 30.5037 Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL, Banham AH, 2006, Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 24(34):5373–5380. , DOI 10.1200/JCO.2006.05.9584 Miyashita M, Sasano H, Tamaki K, Hirakawa H, Takahashi Y, Nakagawa S, Watanabe G, Tada H, Suzuki A, Ohuchi N, Ishida T, 2015, Prognostic significance of tumor-infiltrating CD8+ and FOXP3+ lymphocytes in residual tumors and alterations in these parameters after neoadjuvant chemotherapy in triple-negative breast cancer: a retrospective multicenter study. Breast cancer research : BCR 17:124. , DOI 10.1186/s13058-015-0632-x TakenakaM, Seki N, Toh U,Hattori S,Kawahara A,Yamaguchi T, Koura K, Takahashi R, Otsuka H, Takahashi H, Iwakuma N, Nakagawa S, Fujii T, Sasada T, Yamaguchi R, Yano H, Shirouzu K, Kage M, 2013, FOXP3 expression in tumor cells and tumorinfiltrating lymphocytes is associated with breast cancer prognosis. Molecular and clinical oncology 1(4):625–632. , DOI 10. 3892/mco.2013.107 Lee S, Cho EY, Park YH, Ahn JS, Im YH, 2013, Prognostic impact of FOXP3 expression in triple-negative breast cancer. Acta Oncol 52(1):73–81. , DOI 10.3109/0284186X.2012.731520 Papaioannou E, Sakellakis M, Melachrinou M, Tzoracoleftherakis E, Kalofonos H, Kourea E, 2019, A standardized evaluation method for FOXP3+ Tregs and CD8+ T-cells in breast carcinoma: association with breast carcinoma subtypes, stage and prognosis. Anticancer research 39, 3):1217-1232. Doi:, DOI 10. 21873/anticanres.13232 Zhang L,Wang XI, Ding J, Sun Q, Zhang S, 2019, The predictive and prognostic value of Foxp3+/CD25+ regulatory T cells and PDL1 expression in triple negative breast cancer. Ann Diagn Pathol 40:143–151. , DOI 10.1016/j.anndiagpath.2019.04.004 Ali HR, Provenzano E, Dawson SJ, Blows FM, Liu B, Shah M, Earl HM, Poole CJ, Hiller L, Dunn JA, Bowden SJ, Twelves C, Bartlett JM, Mahmoud SM, Rakha E, Ellis IO, Liu S, Gao D, Nielsen TO, Pharoah PD, Caldas C, 2014, Association between CD8+ T-cell infiltration and breast cancer survival in 12,439 patients. Annals of oncology : official journal of the European Society for Medical Oncology 25(8):1536–1543. , DOI 10.1093/ annonc/mdu191 Mahmoud SM, Paish EC, Powe DG,Macmillan RD, Lee AH, Ellis IO, Green AR, 2011, An evaluation of the clinical significance of FOXP3+ infiltrating cells in human breast cancer. Breast Cancer Res Treat 127(1):99–108. , DOI 10.1007/s10549-010- 0987-8 Ladoire S, Arnould L, Mignot G, Coudert B, Rebe C, Chalmin F, Vincent J, Bruchard M, Chauffert B, Martin F, Fumoleau P, Ghiringhelli F, 2011, Presence of Foxp3 expression in tumor cells predicts better survival in HER2-overexpressing breast cancer patients treated with neoadjuvant chemotherapy. Breast Cancer Res Treat 125(1):65–72. , DOI 10.1007/s10549-010-0831-1 Merlo A, Casalini P, Carcangiu ML, Malventano C, Triulzi T, Menard S, Tagliabue E, Balsari A, 2009, FOXP3 expression and overall survival in breast cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 27(11):1746–1752. , DOI 10.1200/JCO.2008.17.9036 DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N, Coussens LM(2009, CD4(+, T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 16(2):91–102. https://doi. org/10.1016/j.ccr.2009.06.018 Su S, Liu Q, Chen J, Chen J, Chen F,He C, HuangD,WuW, Lin L, Huang W, Zhang J, Cui X, Zheng F, Li H, Yao H, Su F, Song E, 2014, A positive feedback loop between mesenchymal-like cancer cells and macrophages is essential to breast cancer metastasis. Cancer Cell 25(5):605–620. , DOI 10.1016/j.ccr.2014.03. 021 Medrek C, Ponten F, Jirstrom K, Leandersson K, 2012, The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients. BMC Cancer 12:306. , DOI 10.1186/1471-2407-12-306 Zhao X, Qu J, Sun Y,Wang J, Liu X,Wang F, Zhang H,Wang W, Ma X, Gao X, Zhang S, 2017, Prognostic significance of tumorassociated macrophages in breast cancer: a meta-analysis of the literature. Oncotarget 8, 18):30576-30586. Doi:, DOI 10. 18632/oncotarget.15736 Bellucci R, Martin A, Bommarito D, Wang K, Hansen SH, Freeman GJ, Ritz J, 2015, Interferon-gamma-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression. Oncoimmunology 4(6):e1008824. , DOI 10.1080/2162402X.2015.1008824 Sanmamed MF, Chen L, 2014, Inducible expression of B7-H1, PD-L1, and its selective role in tumor site immune modulation. Cancer J 20(4):256–261. , DOI 10.1097/PPO. 0000000000000061 Botti G, Collina F, Scognamiglio G, Rao F, Peluso V, De Cecio R, Piezzo M, Landi G, De Laurentiis M, Cantile M, Di Bonito M, 2017, Programmed death ligand 1, PD-L1, tumor expression is associated with a better prognosis and diabetic disease in triple negative breast Cancer patients. Int J Mol Sci 18(2). https://doi. org/10.3390/ijms18020459 Brockhoff G, Seitz S, Weber F, Zeman F, Klinkhammer-Schalke M, Ortmann O, Wege AK, 2018, The presence of PD-1 positive tumor infiltrating lymphocytes in triple negative breast cancers is associated with a favorable outcome of disease. Oncotarget 9, 5): 6201-6212. Doi:, DOI 10.18632/oncotarget.23717 Sabatier R, Finetti P, Mamessier E, Adelaide J, Chaffanet M, Ali HR, Viens P, Caldas C, Birnbaum D, Bertucci F, 2015, Prognostic and predictive value of PDL1 expression in breast cancer. Oncotarget 6, 7):5449-5464. Doi:, DOI 10.18632/ oncotarget.3216 Uhercik M, Sanders AJ, Owen S, Davies EL, Sharma AK, Jiang WG, Mokbel K, 2017, Clinical significance of PD1 and PDL1 in human breast Cancer. Anticancer research 37, 8):4249-4254. Doi: , DOI 10.21873/anticanres.11817 Wang ZQ, Milne K, Derocher H, Webb JR, Nelson BH, Watson PH, 2017, PD-L1 and intratumoral immune response in breast cancer. Oncotarget 8, 31):51641-51651. Doi:, DOI 10.18632/ oncotarget.18305 Wu Z, Zhang L, Peng J, Xu S, Zhou L, Lin Y, Wang Y, Lu J, Yin W, Lu J, 2019, Predictive and prognostic value of PDL1 protein expression in breast cancer patients in neoadjuvant setting. Cancer biology & therapy 20(6):941–947. , DOI 10.1080/ 15384047.2019.1583533 Li Z, Dong P, Ren M, Song Y, Qian X, Yang Y, Li S, Zhang X, Liu F, 2016, PD-L1 expression is associated with tumor FOXP3(+, regulatory T-cell infiltration of breast Cancer and poor prognosis of patient. J Cancer 7(7):784–793. , DOI 10.7150/jca. 14549 Muenst S, Schaerli AR, Gao F, Daster S, Trella E, Droeser RA, MuraroMG, Zajac P, Zanetti R, GillandersWE,WeberWP, Soysal SD, 2014, Expression of programmed death ligand 1, PD-L1, is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat 146(1):15–24. , DOI 10.1007/s10549- 014-2988-5 Jiang C, Cao S, Li N, Jiang L, Sun T, 2019, PD-1 and PD-L1 correlated gene expression profiles and their association with clinical outcomes of breast cancer. Cancer Cell Int 19:233. https://doi. org/10.1186/s12935-019-0955-2 Gaule P, Smithy JW, TokiM, Rehman J, Patell-Socha F, Cougot D, Collin P, Morrill P, Neumeister V, Rimm DL, 2017, A quantitative comparison of antibodies to programmed cell death 1 ligand 1. JAMA oncology 3(2):256–259. , DOI 10.1001/ jamaoncol.2016.3015 Zajac M, Scott M, Ratcliffe M, Scorer P, Barker C, Al-Masri H, RebelattoMC,Walker J, 2019, Concordance among four commercially available, validated programmed cell death ligand-1 assays in urothelial carcinoma. Diagn Pathol 14(1):99. , DOI 10. 1186/s13000-019-0873-6 Buttner R, Gosney JR, Skov BG, Adam J, Motoi N, Bloom KJ, Dietel M, Longshore JW, Lopez-Rios F, Penault-Llorca F, Viale G, Wotherspoon AC, Kerr KM, Tsao MS, 2017, Programmed deathligand 1 immunohistochemistry testing: a review of analytical assays and clinical implementation in non-small-cell lung Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 35(34):3867–3876. , DOI 10.1200/JCO.2017.74.7642 Karnik T, Kimler BF, Fan F, Tawfik O, 2018, PD-L1 in breast cancer: comparative analysis of 3 different antibodies. Hum Pathol 72:28–34. , DOI 10.1016/j.humpath.2017.08.010 Ghebeh H, Barhoush E, Tulbah A, Elkum N, Al-Tweigeri T, Dermime S, 2008, FOXP3+ Tregs and B7-H1+/PD-1+ T lymphocytes co-infiltrate the tumor tissues of high-risk breast cancer patients: implication for immunotherapy. BMC Cancer 8:57. https:// doi.org/10.1186/1471-2407-8-57 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2733 EP 2745 DI 10.1007/s12253-020-00874-4 PG 13 ER PT J AU Sejben, A Koszo, R Kahan, Zs Cserni, G Zombori, T AF Sejben, Anita Koszo, Renata Kahan, Zsuzsanna Cserni, Gabor Zombori, Tamas TI Examination of Tumor Regression Grading Systems in Breast Cancer Patients Who Received Neoadjuvant Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; Neoadjuvant therapy; Regression pattern; Grading systems; Residual cancer burden ID Breast cancer; Neoadjuvant therapy; Regression pattern; Grading systems; Residual cancer burden AB Neoadjuvant therapy is a common formof treatment in locally advanced breast cancer (LABC) patients. Besides some guidelines for grading regression, a standardized general scheme is not yet available. The aim of our study was to compare the prognostic impact of different regression grading systems, namely the TR/NR, Chevallier, Sataloff, Denkert-Sinn, Miller-Payne, NSABPB18, Residual Disease in Breast and Nodes and Residual Cancer Burden (RCB) on disease-free (DFS) and overall survival (OS). Data of 746 breast cancer patients treated in neoadjuvant setting between 1999 and 2019 have been included. The different regression grades and follow-up data were collected from medical charts. Statistical analysis included the Kaplan-Meier method, log-rank test and multivariate Cox regression. The average patient age was 55 years. The DFS and OS estimates of patients with complete pathological regression and residual in situ carcinoma have been significantly more favorable than those having partial regression or no signs of regression (pDFS<0.001, pOS < 0.001). Significant differences were found between DFS estimates of classes with partial regression and without regression defined by RCB. Concerning DFS estimates, the RCB classification (p = 0.019), while regarding OS data the y-stage (p = 0.011) and the nodal status (ypN; p = 0.045) were significant prognosticators by multivariate Cox regression. Regression grading systems help the evaluation of regression in LABC patients treated with neoadjuvant therapy. Of the several grading systems compared, the RCB classification makes the best distinction between the outcomes of the different classes, therefore we recommend the inclusion of RCB into the histopathological findings. C1 [Sejben, Anita] University of Szeged, Department of Pathology, Allomas u. 1, 6725 Szeged, Hungary. [Koszo, Renata] University of Szeged, Department of OncotherapySzeged, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of OncotherapySzeged, Hungary. [Cserni, Gabor] University of Szeged, Department of Pathology, Allomas u. 1, 6725 Szeged, Hungary. [Zombori, Tamas] University of Szeged, Department of Pathology, Allomas u. 1, 6725 Szeged, Hungary. RP Sejben, A (reprint author), University of Szeged, Department of Pathology, 6725 Szeged, Hungary. EM sejben.anita@gmail.com CR Giordano SH, 2003, Update on locally advanced breast cancer. Oncologist 8:521–530. , DOI 10.1634/theoncologist.8-6-521 Rustogi A, Budrukkar A, Dinshaw K, Jalali R, 2005, Management of locally advanced breast cancer: evolution and current practice. J Can Res Ther 1:21–30. , DOI 10.4103/0973-1482.16086 Thompson AM,Moulder-Thompson SL, 2012, Neoadjuvant treatment of breast cancer. Ann Oncol 23:231–236. , DOI 10. 1093/annonc/mds324 Dani M, McDonnell J, Karp S, Jaffe V, 2007, Do breast cancer tumours downsize as well as downgrade with neoadjuvant chemotherapy? Breast Cancer Res 9:SP3. , DOI 10.1186/bcr1709 Corben AD, Abi-Raad R, Popa I, Chy T, Macklin EA, Koerner FC, Taghian AG, Brachtel EF, 2013, Pathologic response and longterm follow-up in breast cancer patients treated with neoadjuvant chemotherapy. A comparison between classifications and their practical application. Arch Pathol Lab Med 137:1074–1082. , DOI 10.5858/arpa.2012-0290-OA Kaufmann M, von Minckwitz G, Mamounas EP, Cameron D, Carey LA, Cristofanilli M, Denkert C, Eiermann W, Gnant M, Harris JR, Karn T, Liedtke C, Mauri D, Rouzier R, Ruckhaeberle E, Semiglazov V, Symmans WF, Tutt A, Pusztai L, 2012, Recommendations from an international consensus conference on the current status and future of neoadjuvant systemic therapy in primary breast cancer. Ann Surg Oncol 19:1508–1516. https:// doi.org/10.1245/s10434-011-2108-2 Philipovskiy A, Corral J,Dwivedi KA, Heydarian R,Gaur S, 2019, Efficacy of neoadjuvant versus adjuvant chemotherapy in Hispanic/ Latino, H/L, women with local or locally advanced triple-negative breast cancer, TNBC). In Vivo 33:1227–1234. , DOI 10. 21873/invivo.11594 Coates AS,Winer EP, Goldhirsch A, Gelber RD, GnantM, Piccart- Gebhart M, Thurlimann B, Senn HJ, Panel Members, 2015, Tailoring therapies – improving the management of early breast cancer: St Gallen international expert consensus on the primary therapy of early breast cancer. Ann Oncol 26:1533–1546. https:// doi.org/10.1093/annonc/mdv221 Senkus E, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rutgers E, Zackrisson S, Cardoso F, Guidelines Committee ESMO, 2015, Primary breast cancer: ESMO clinical practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 26:8–30. https:// doi.org/10.1093/annonc/mdv298 Provenzano E, Bossuyt V, Viale G, Cameron D, Badve S, Denkert C, MacGrogan G, Penault-Llorca F, Boughey J, Curigliano G, Dixon JM, Esserman L, Fastner G, Kuehn T, Peintinger F, von Minckwitz G, White J, Yang W, Symmans WF, 2015, Residual disease characterization working Group of the Breast International Group-North American Breast Cancer Group Collaboration. Standardization of pathologic evaluation and reporting of postneoadjuvant specimens in clinical trials of breast cancer: recommendations from an international working group. Mod Pathol 28:1185–1201. , DOI 10.1038/modpathol.2015.74 Cserni G, Kulka J, Francz M, Jaray B, Kalman E, Kovacs I, Krenacs T, Udvarhelyi N, Vass L, 2016, Pathological diagnosis, work-up and reporting of breast cancer. Recommendations of the 3rd Hungarian consensus conference on breast Cancer. Magy Onkol 60:209–228 von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, Gerber B, EiermannW,Hilfrich J,Huober J, Jackisch C, Kaufmann M, Konecny GE, Denkert C, Nekljudova V, Mehta K, Loibl S, 2012, Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol 30:1796–1804. , DOI 10.1200/JCO.2011.38.8595 U.S. Food and Drug Administration, 2014, Guidance for Industry: Pathological complete response in neoadjuvant treatment of highrisk early-stage breast cancer: use as an endpoint to support accelerated approval. http://www.fda.gov/downloads/Drugs/ GuidanceComplianceRegulatoryInformation/Guidances/ UCM305501.pdf Accessed 19 Jan 2020 Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, Washington MK, Gershenwald JE, Compton CC, Hess KR, Sullivan DC, Jessup JM, Brierly JD, Gaspar LE, Schilsky RL, Balch CM, Winchester DP, Madera M, Asare EA, 2017, AJCC Cancer staging manual, 8th edn. Springer, Chicago Sethi D, Sen R, Parshad S, Khetarpal S, Garg M, Sen J, 2012, Histopathologic changes following neoadjuvant chemotherapy in various malignancies. Int J Appl Basic Med Res 2:111–116. , DOI 10.4103/2229-516X.106353 Park CK, JungWH, Koo JS, 2016, Pathologic evaluation of breast cancer after neoadjuvant therapy. J Pathol TranslMed 50:173–180. , DOI 10.4132/jptm.2016.02.02 Mamounas EP, Anderson SJ, Dignam JJ, Bear HD, Julian TB, Geyer CE Jr, Taghian A, Wickerham DL, Wolmark N, 2012, Predictors of locoregional recurrence after neoadjuvant chemotherapy: results from combined analysis of National Surgical Adjuvant Breast and bowel project B-18 and B-27. J Clin Oncol 30:3960– 3966. , DOI 10.1200/JCO.2011.40.8369 Chevallier B, Chollet P,Merrouche Y, Roche H, Fumoleau P, Kerbrat P, Genot JY, Fargeot P,Olivier JP, Fizames C et al, 1995, Lenograstim prevents morbidity from intensive induction chemotherapy in the treatment of inflammatory breast cancer. J Clin Oncol 13:1564–1571. , DOI 10.1200/JCO.1995.13.7.1564 Sataloff DM, Mason BA, Prestipino AJ, Seinige UL, Lieber CP, Baloch Z, 1995, Pathologic response to induction chemotherapy in locally advanced carcinoma of the breast: a determinant of outcome. J Am Coll Surg 180:297–306 Ogston KN, Miller ID, Payne S, Hutcheon AW, Sarkar TK, Smith I, Schofield A, Heys SD, 2003, A new histological grading system to assess response of breast cancers to primary chemotherapy: prognostic significance and survival. Breast 12:320–327. , DOI 10.1016/s0960-9776(03)00106-1 Denkert C, Schickling O, von Minckwitz G, 2006, Preoperative chemotherapy in breast cancer and the development of new predictive markers. Verh Dtsch Ges Pathol 90:114–123 Symmans WF, Peintinger F, Hatzis C, Rajan R, Kuerer H, Valero V, Assad L, Poniecka A, Hennessy B, Green M, Buzdar AU, Singletary SE, Hortobagyi GN, Pusztai L, 2007, Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol 28:4414–4422. 10.1200/JCO.2007.10.6823 Wells CA, Amendoeira I, Bellocq JP, Bianchi S, Boecker W, Borisch B, Rasmussen B, Callagy GM, Chmielik E, Cordoba A, Cserni G, Decker T, DeGaetano J, Drijkoningen M, Ellis IO, Faverly DR, Foschini MP, Frkovic-Grazio S, Grabau D, Heikkila P, Iacovou E, Jacquemier J, Kaya H, Kulka J, Lacerda M, Liepniece-Karele I, Martinez-Penuela J, Quinn CM, Rank F, Regitnig P, Reiner A, Sapino A, Tot T, Van Diest P, Varga Z, Wesseling J, Zolota V, Zozaya-Alvarez E, 2013, S2: pathology update. Quality assurance guidelines for pathology. In: European guidelines for quality assurance in breast cancer screening and diagnosis. Fourth edition, Supplements. Perry N, Broeders M, de Wolf C, Tornberg S, Holland R, von Karsa L, eds.). European Commission, Office for Official Publications of the European Union, Luxembourg, pp. 73–120 Chollet P, Abrial C, Durando X, Thivat E, Tacca O, Mouret- Reynier MA, Leheurteur M, Kwiatkowski F, Dauplat J, Penault- Llorca F, 2008, A new prognostic classification after primary chemotherapy for breast cancer: residual disease in breast and nodes, RDBN). Cancer J 14:128–132. , DOI 10.1097/PPO. 0b013e31816bdea2 Fowler AM, Mankoff DA, Joe BN, 2017, Imaging neoadjuvant therapy response in breast cancer. Radiology 285:358-375. https:// doi.org/10.1148/radiol.2017170180, 285, 358, 375 Royal College of Pathologists of Australasia, 2012, Invasive breast cancer structured reporting protocol. https://www.rcpa.edu.au/ getattachment/7b70b3e5-5dca-403f-893e-638815f487b1/Protocolinvasive- breast-cancer.aspx. Accessed 22 Nov 2019 Lambein K, Van deVijver K, FaverlyD, Colpaert C, 2011, Belgian guidelines for laboratory handling and pathology reporting of breast carcinoma after neoadjuvant therapy. Belg J Clin Oncol 5:144–153. , DOI 10.1111/j.1365-2559.2006.02419.x Arbeitsgemeinschaft Gynakologische Onkologie Studiengruppe, 2018, Diagnosis and treatment of patients with primary and metastatic breast cancer. https://www.ago-online.de/fileadmin/ downloads/leitlinien/mamma/2018-03/EN/Gesamt_PDF_ Englisch/Updated_Guidelines_2018.pdf. NHS Cancer Screening Programmes jointly with The Royal College of Pathologists, 2005, Pathology reporting of breast disease. https://www.cmcanceralliance.nhs.uk/application/files/3615/ 4815/5660/Guidelines_for_NHSBSP58_January_2005_ Reviewed_CNG_June_2010.pdf Accessed 18 Jan 2020 Integraal Kankercentrum Nederland, 2012, Beoordeling na neoadjuvante chemo- of endocriene therapie. http://www. oncoline.nl/breastcancer Accessed 26 June 2018 Lester SC, Bose S, Chen YY, Connolly JL, de Baca ME, Fitzgibbons PL, Hayes DF, Hill KA, Kleer C, O’Malley FP, Page DL, Smith BL, Tan LK,Weaver DL, Winer E, Simpson JF, 2012, Protocol for the examination of specimens from patients with invasive carcinoma of the breast. http://www.cap.org/ShowProperty? nodePath=/UCMCon/Contribution%20Folders/WebContent/pdf/ cp-breast-invasive-16protocol-3300.pdf Accessed 22 Nov 2019 SymmansWF,Wei C, Gould R, Yu X, Zhang Y, Liu M, Walls A, Bousamra A, Ramineni M, Sinn B, Hunt K, Buchholz TA, Valero V, Buzdar AU, Yang W, Brewster AM, Moulder S, Pusztai L, Hatzis C, Hortobagyi GN, 2017, Long-term prognostic risk after neoadjuvant chemotherapy associated with residual cancer burden and breast cancer subtype. J Clin Oncol 35:1049–1060. https://doi. org/10.1200/JCO.2015.63.1010 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2747 EP 2754 DI 10.1007/s12253-020-00867-3 PG 8 ER PT J AU Zengin, M Benek, S AF Zengin, Mehmet Benek, Suat TI The Proportion of Tumour-Stroma in Metastatic Lymph Nodes is An Accurately Prognostic Indicator of Poor Survival for Advanced-Stage Colon Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colon cancers; Proportion of tumour-stroma; Pathology; Prognostic markers; Stage III-IV ID Colon cancers; Proportion of tumour-stroma; Pathology; Prognostic markers; Stage III-IV AB The importance of tumour microenvironment in tumour behaviour has now become clearer. This study aimed to determine the prognostic effect of the proportion of tumour-stroma (PTS) in metastatic lymph nodes of advanced-stage colon cancers (CCs). We investigated PTS in positive lymph nodes of stage III-IV CC patients who underwent surgical treatment between 2004 and 2014. We used a standard approach in methodology. PTS was significantly associated with prognostic factors in the metastatic lymph nodes (perineural invasion [p = 0.031], lymphatic invasion [p = 0.032], invasive margin [p = 0.043], advanced pT [p = 0.020], and margin involvement [p = 0.034]). In addition, the correlations between PTS estimates (R = 0.704 to 0.617, p < 0.001), the reproducibility of the research (Κappa = 0.72–0.68) and the usefulness of the cut-off value (ROC: 50.33%; AUC = 0.752 [0.667–0.857]) were successful. In univariate analysis, 5-year survival was poor for RFS (p < 0.001), OS (p = 0.001) and LR (p = 0.013) in high PTS patients. Multivariate analysis confirmed that high PTS was an independent worse parameter for RFS (HR = 1.32, 95% CI: 1.17–2.55, p = 0.001) and OS (HR = 1.37, 95% CI: 1.25–1 - 2.56, p = 0.009). In this study, we showed that high PTS inmetastatic lymph nodes was a successful prognosticmarker for advanced-stage CCs. Also, the standard approach we used for the methodology was successful. C1 [Zengin, Mehmet] Kirikkale University, Department of PathologyKirikkale, Turkey. [Benek, Suat] Beylikduzu State Hospital, Department of General SurgeryIstanbul, Turkey. RP Zengin, M (reprint author), Kirikkale University, Department of Pathology, Kirikkale, Turkey. EM mz1379@hotmail.com CR Deijen CL, Vasmel JE, de Lange-de Klerk ESM et al, 2017, Tenyear outcomes of a randomised trial of laparoscopic versus open surgery for colon cancer. Surg Endosc 31:2607–2615 Bertelsen CA, Neuenschwander AU, Jansen JE et al, 2015, Disease-free survival after complete mesocolic excision compared with conventional colon cancer surgery: a retrospective, population-based study. Lancet Oncol 16:161–168 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J et al, 2013, Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403 Gray R, Barnwell J, McConkey C et al, 2007, Adjuvant chemotherapy versus observation in patients with colorectal cancer: a randomised study. Lancet. 370:2020–2029 Benson AB, Schrag D, Somerfield MR et al, 2004, American Society of Clinical Oncology recommendations on adjuvant chemotherapy for stage II colon cancer. J Clin Oncol 22:3408–3419 Langley RR, Fidler IJ, 2011, The seed and soil hypothesis revisited–the role of tumour-stroma interactions in metastasis to different organs. Int J Cancer 128:2527–2535 De Wever O, Mareel M, 2003, Role of tissue stroma in cancer cell invasion. J Pathol 200:429–447 Quail DF, Joyce JA, 2013, Microenvironmental regulation of tumour progression and metastasis. Nat Med 19:1423–1437 Pietras K, Ostman A, 2010, Hallmarks of cancer: interactions with the tumour stroma. Exp Cell Res 316:1324–1331 MeskerWE, Junggeburt JM, Szuhai K et al, 2007, The carcinomastromal ratio of colon carcinoma is an independent factor for survival compared to lymph node status and tumour stage. Cell Oncol 29:387–398 Park JH, Richards CH, McMillan DC, 2014, The relationship between tumour stroma percentage, the tumour microenvironment and survival in patients with primary operable colorectal cancer. Ann Oncol 25:644–651 Huijbers A, Tollenaar RA, 2013, V Pelt GW, et al. the proportion of tumour-stroma as a strong prognosticator for stage II and III colon cancer patients: validation in the VICTOR trial. Ann Oncol 24:179–185 Mesker WE, Liefers GJ, Junggeburt JM et al, 2009, Presence of a high amount of stroma and downregulation of SMAD4 predict for worse survival for stage I-II colon cancer patients. Cell Oncol 31: 169–178 ZenginM(2019, Prognostic role of tumor-infiltrating T lymphocytes in stage IIA, T3N0, colon cancer: a broad methodological study in a fairly homogeneous population. Ann Diagn Pathol 41:69–78 Zengin M, 2020, Local inflammatory response can predict clinical outcome in patients with curatively resected stage-IIB Colon Cancer: an advanced methodological study. Pathol Oncol Res 26: 1805-1816 McShane LM, Altman DG, Sauerbrei W et al, 2005, REporting recommendations for tumour MARKer prognostic studies, REMARK). Br J Cancer 93:387–391 Sobin LH, Compton CC, 2010, TNMseventh edition: what’s new, what’s changed: communication from the International Union against Cancer and the American joint committee on Cancer. Cancer. 116(22):5336–5339 Eriksen AC, Sorensen FB, Lindebjerg J et al, 2018, The prognostic value of tumour stroma ratio and tumour budding in stage II colon cancer. A nationwide population-based study. Int J Color Dis 33(8): 1115–1124 Salgado R, Denkert C, Demaria S et al, 2015, The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: recommendations by an international TILs working group 2014. Ann Oncol 26:259–271 McGraw KO, 1996, Forming inferences about some intraclass correlation coefficient. Psychol Methods 1:30–46 Landis JR, Koch GG, 1977, The measurement of observer agreement for categorical data. Biometrics. 33:159–174 West NP, Dattani M, McShane P et al, 2020, The proportion of tumour cells is an independent predictor for survival in colorectal cancer patients. Br J Cancer 102(10):1519–1523 Hutchins GGA, Treanor D, Wright A et al, 2017, Intra-tumoural stromal morphometry predicts disease recurrence but no a response to 5-fluorouracil—results from the QUASAR trial of colorectal cancer. Histopathology 3:391–404 Hynes SO, Coleman HG, Kelly PJ et al, 2017, Back to the future: routine morphological assessment of the tumour microenvironment is prognostic in stage II/III colon cancer in a large population-based study. Histopathology. 71:12–26 van PeltGW, Hansen TB, Esther B et al, 2016, Stroma-high lymph node involvement predicts poor survival more accurately for patients with stage III colon cancer. J Med Surg Pathol 1:116 Moserle L, Casanovas O, 2013, Anti-angiogenesis and metastasis: a tumour and stromal cell alliance. J Intern Med 273:128–137 Koukourakis MI, Giatromanolaki A, Harris AL et al, 2006, Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: a metabolic survival role for tumor-associated stroma. Cancer Res 66:632–637 Guthrie GJ, Roxburgh CS, Richards CH et al, 2013, Circulating IL- 6 concentrations link tumour necrosis and systemic and local inflammatory responses in patients undergoing resection for colorectal cancer. Br J Cancer 109:131–137 Ueno H, Jones AM, Wilkinson KH et al, 2004, Histological categorisation of fibrotic cancer stroma in advanced rectal cancer. Gut. 53:581–586 Lieubeau B, HeymannMF, Henry F et al, 1999, Immunomodulatory effects of tumor-associated fibroblasts in colorectal-tumor development. Int J Cancer 81:629–636 HemmingsC, 2013, Is carcinoma amesenchymal disease? The role of the stromal microenvironment in carcinogenesis. Pathology, Phila, 45:371–381 Tsujino T, Seshimo I, Yamamoto H et al, 2007, Stromal myofibroblasts predict disease recurrence for colorectal cancer. Clin Cancer Res 13:2082–2090 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2755 EP 2764 DI 10.1007/s12253-020-00877-1 PG 10 ER PT J AU Csernus, B Timar, B Fulop, Zs Matolcsy, A AF Csernus, Balazs Timar, Botond Fulop, Zsolt Matolcsy, Andras TI Grade I, II and III Follicular Lymphomas Express Ig VH Genes with Different Patterns of Somatic Mutation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Follicular lymphoma|; Immunoglobulin gene; Somaticmutation; Clonal evolution ID Follicular lymphoma|; Immunoglobulin gene; Somaticmutation; Clonal evolution AB Follicular lymphoma (FL) is an indolent, B-cell, non-Hodgkin’s lymphoma with varying cytological appearance and clinical behavior. The genetic hallmark of FL is the t(14;18) translocation, and as a germinal center derived entity it is also characterized by somatic hypermutation of the immunoglobulin heavy chain (IgH) gene. In an attempt to correlate thismolecular signature with the cytological grading of FL, we have analyzed the IgH variable (IgVH), regions in all cytological grades of FL. Four FL cases showing t(14;18) translocation were classified into grade I-III categories according to the current WHO guidelines. The IgVH gene segments were PCR-amplified, sequenced, and compared to their respective germline IgVH sequences. The neoplastic cells of grade I and II FLs revealed clonally related, but highly divergent IgVH gene sequences indicating the ongoing nature of somatic hypermutation. Grade III FL also showed extensive presence of somatic hypermutation, but these mutations were not associated with intraclonal divergence. Thus, these results suggest that grade I-II and grade III FL may represent different biological entities. The presence of ongoing somatic hypermutation of IgVH sequences in grade I and II FLs is compatible with direct follicular origin of these tumor cells, contrasting the homogenous, stable clones of grade III FL resembling a post-follicular stage of B-cell development. Our findings demonstrate that contrary to the three tiered cytological grading, molecular features of IgH genes classify FL into two distinct subcategories. These studies also suggest that with progression FL gains post-follicular– like molecular features and becomes independent of the germinal center microenvironment. C1 [Csernus, Balazs] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Timar, Botond] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Fulop, Zsolt] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. [Matolcsy, Andras] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Ulloi ut 26, 1085 Budapest, Hungary. RP Matolcsy, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. EM matolcsy.andras@med.semmelweis-univ.hu CR Harris NL, 1992, Ferry JA, 1992, follicular lymphoma and related disorders, germinal center lymphomas). In: Knowles DM, ed, Neoplastic Hematopathology. Williams &Wilkins, Baltimore, p 645 Harris NL, Jaffe ES, Stein H, Banks PM, Chan JKC, Cleary ML, Delsol G, Wolf-Peeters CD, Falini B, Gatter KC, Grogan TM, Isaacson PG, Knowles DM, Mason DY, Muller-Hermelink HK, Pileri SA, Piris MA, Ralfkiaer E, Warnke RA, 1994, A revised European-American classification of lymphoid neoplasms: a proposal from the international lymphoma study group. Blood 84: 1361–1392 Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R et al, 2016, The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 127(20):2375–2390 Jaffe ES, RaffeldM, Mederios LJ, 1993, Histopathologic subtypes of indolent lymphomas: caricatures of the mature B-cell system. Semin Oncol 20(suppl 5):3 Martin AR, Weisenburger DD, Chan WC, Ruby EI, Anderson JR, Vose JM, Bierman PJ, Bast MA, Daley DT, Armitage JO, 1995, Prognostic value of cellular proliferation and histologic grade in follicular lymphoma. Blood 85:3671–3678 Oviatt DL, Vousar JB, Collins RD, Flexner JM, Stein RS, 1984, Malignant lymphomas of follicular center cell origin in humans. V. Incidence, clinical features, and prognostic implications of transformation of small cleaved cell nodular lymphoma. Cancer 53:1109 Shustik J, QuinnM, Connors JM, Gascoyne RD, Skinnider B, Sehn L, 2008, Follicular non-Hodgkin lymphoma grade 3a and 3b subtypes exhibit similar clinical behaviour and treatment outcome. Blood 112(11):3777 ErsbOll J, Schultz HB, Perdersen-Bjergaard J, Nissen NJ, 1989, Follicular low-grade non-Hodgkin lymphomas: long-term outcome with or without tumor progression. Eur J Haematol 42:155 Sundaram S,Moore E, Ayyappan S, Covut F, Tomlinson B, Creger R,Malek E, Metheny L, CooperBW, Lazarus HM, Lima M, Caimi P, 2017, Effect of histologic grade on clinical outcomes of follicular lymphoma: prolonged progression free survival of grade 3 follicular lymphoma in the rituximab era. Blood 130(Supplement 1):2775 Salmon SE, Seligmann M, 1974, B-cell neoplasia in man. Lancet.;2(7891):1230–1233. , DOI 10.1016/s0140- 6736(74)90748-x SeifertM, Scholtysik R, Kuppers R, 2019, Origin and pathogenesis of B cell lymphomas. Methods Mol Biol 1956:1–33. https://doi. org/10.1007/978-1-4939-9151-8_1 Kluin PM, 2013, Origin and migration of follicular lymphoma cells. Haematologica. 98(9):1331–1333. , DOI 10.3324/ haematol.2013.091546 Carbone A, Roulland S, Gloghini A, Younes A, von Keudell G, Lopez-Guillermo A, Fitzgibbon J, 2019, Follicular lymphoma. Nat Rev Dis Primers 5:83. , DOI 10.1038/s41572-019-0132-x Zelenetz AD, Chen TT, Levy R, 1992, Clonal expansion in follicular lymphoma occurs subsequent to antigenic selection. J Exp Med 176:1137–1148 Bahler DW, Levy R, 1992, Clonal evolution of a follicular lymphoma: evidence for antigen selection. Proc Natl Acad Sci U S A 89:6770–6774 Deane M, Norton JD, 1990, Immunoglobulin heavy chain variable region family usage is independent of tumor cell phenotype in human B lineage leukemias. Eur J Immunol 20:2209–2217 Sanz I, Kelly P, Williams C, Scholl S, Tucker P, Capra JD, 1989, The smaller human VH gene families display remarkably little polymorphism. EMBO J 8:3741–3748 Tomlinson IM, Walter G, Marks JD, Llewelyn MB, Winter G, 1992, The repertoire of human germline VH sequences reveals about fifty groups of VH segments with different hypervariable loops. J Mol Biol 227:776–798 Chen PP, 1990, Structural analyses of human developmentally regulated VH3 genes. Scand J Immunol 31:257–267 Baer R, Forster A, Lavenir I, Rabbitts TH, 1988, Immunoglobulin VH genes are transcribed by T cells in association with a new 5′ exon. J Exp Med 167:2011–2016 Chang B, Casali P, 1994, The CDR1 sequences of a major proportion of human germline Ig VH genes are inherently susceptible to amino acid replacement. Immunol Today 15:367–373 Okosun J, Bodor C, Wang J, Araf S, Yang CY, Pan C, Boller S, Cittaro D, Bozek M, Iqbal S, Matthews J, Wrench D, Marzec J, Tawana K, Popov N, O'Riain C, O'Shea D, Carlotti E, Davies A, Lawrie CH, Matolcsy A, Calaminici M, Norton A, Byers RJ, Mein C, Stupka E, Lister TA, Lenz G, Montoto S, Gribben JG, Fan Y, Grosschedl R, Chelala C, Fitzgibbon J, 2014, Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma.Nat Genet 46:176–181 GreenMR, Kihira S, Liu CL, 2015, Et al., 2015)mutations in early follicular lymphoma progenitors are associated with suppressed antigen presentation. Proc Natl Acad Sci 112:1116–1125 Lackraj T, Goswami R, Kridel R, 2018,, 2018, pathogenesis of follicular lymphoma. Best Pract Res Clin Haematol 31:2–14 Randall C, Fedoriw Y, 2020, Pathology and diagnosis of follicular lymphoma and related entities. Pathology 52(1):30–39. https://doi. org/10.1016/j.pathol.2019.09.010 Pasqualucci L, Khiabanian H, Fangazio M, Vasishtha M, Messina M,HolmesAB,Ouillette P, Trifonov V, RossiD, Tabbo F, Ponzoni M, Chadburn A, Murty VV, Bhagat G, Gaidano G, Inghirami G, Malek SN, Rabadan R, Dalla-Favera R, 2014,, 2014, genetics of follicular lymphoma transformation. Cell Rep 6:130–140 Bouska A, ZhangW, Gong Q, Iqbal J, Scuto A, Vose J, Ludvigsen M, Fu K, Weisenburger DD, Greiner TC, Gascoyne RD, Rosenwald A, Ott G, Campo E, Rimsza LM, Delabie J, Jaffe ES, Braziel RM, Connors JM, Wu CI, Staudt LM, D‘Amore F, McKeithan TW, Chan WC, 2017, Combined copy number and mutation analysis identifies oncogenic pathways associated with transformation of follicular lymphoma. Leukemia. 31:83–91 Wartenberg M, Vasil P, Meyer zum Bueschenfelde C, Ott G, Rosenwald A, Fend F, Kremer M, 2013, Somatic Hypermutation analysis in follicular lymphoma provides evidence suggesting bidirectional cell migration between lymph node and bone marrow during disease progression and relapse. Haematologica 98(9): 1433–1441. , DOI 10.3324/haematol.2012.074252 Bognar A, Csernus B, Bodor C, Reiniger L, Szepesi A, Toth E, Kopper L, Matolcsy A, 2005, Clonal selection in the bone marrow involvement of follicular lymphoma. Leukemia. 19(9):1656–1662. , DOI 10.1038/sj.leu.2403844 Schroeder HW, Dighiero G, 1994, The pathogenesis of chronic lymphocytic leukemia: analysis of the antibody repertoire. Immunol Today 15:288–294 Dunn-Walters D, Thiede C, Alpen B, Spencer J, 2001, Somatic hypermutation and B-cell lymphoma. Philos Trans R Soc Lond B Biol Sci 356(1405):73–82. , DOI 10.1098/rstb.2000.0751 Zuckerman NS,McCann KJ, Ottensmeier CH, BarakM, Shahaf G, Edelman H, Dunn-Walters D,Abraham RS, Stevenson FK,Mehr R, 2010, Ig gene diversification and selection in follicular lymphoma, diffuse large B cell lymphoma and primary central nervous system lymphoma revealed by lineage tree and mutation analyses. Int Immunol 22(11):875–887. , DOI 10.1093/intimm/dxq44 Kuppers R, Schneider M, Hansmann ML, 2019, Laser-based microdissection of single cells from tissue sections and PCR analysis of rearranged immunoglobulin genes from isolated normal and malignant human B cells. In: Kuppers R, ed, Lymphoma. Methods in molecular biology, 1956. Humana Press, New York Kridel R, Chan FC, Mottok A et al, 2016, Histological transformation and progression in follicular lymphoma: a clonal evolution study. PLoS Med 13:e1002197. , DOI 10.1371/journal. pmed.1002197 Horn H, Kohler C, Witzig R, Kreuz M, Leich E, Klapper W, Hummel M, Loeffler M, Trumper L, Spang R, Rosenwald A, Ott G, for the Molecular Mechanisms in Malignant Lymphomas, MMML, Network Project, 2018, Gene expression profiling reveals a close relationship between follicular lymphoma grade 3A and 3B, but distinct profiles of follicular lymphoma grade 1 and 2. Haematologica. 103:1182–1190 GreenMR,Gentles AJ,Nair RV, Irish JM, Kihira S, Liu CL, Kela I, Hopmans ES, Myklebust JH, Ji H, Plevritis SK, Levy R, Alizadeh AA, 2013, Hierarchy in somatic mutations arising during genomic evolution and progression of follicular lymphoma., Blood, vol. 121, 9, pg. 1604–1611 Carlotti E, Wrench D, Matthews J, Iqbal S, Davies A, Norton A, Hart J, Lai R, Montoto S, Gribben JG, Lister TA, Fitzgibbon J, 2009, Transformation of follicular lymphoma to diffuse large Bcell lymphoma may occur by divergent evolution from a common progenitor cell or by direct evolution from the follicular lymphoma clone. Blood, vol 113(15):3553–3557 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2765 EP 2772 DI 10.1007/s12253-020-00843-x PG 8 ER PT J AU Nagy, N Reis, H Hadaschik, B Niedworok, Ch Modos, O Szendroi, A Biro, K Hager, Th Herold, Th Ablat, J Black, P Okon, K Tolkach, Y Csizmarik, A Olah, Cs Keresztes, D Bremmer, F Gaisa, N Kriegsmann, J Kovalszky, I Kiss, A Timar, J Szasz, AM Rink, M Fisch, M Nyirady, P Szarvas, T AF Nagy, Nikolett Reis, Henning Hadaschik, Boris Niedworok, Christian Modos, Orsolya Szendroi, Attila Biro, Krisztina Hager, Thomas Herold, Thomas Ablat, Jason Black, C. Peter Okon, Krzysztof Tolkach, Yuri Csizmarik, Anita Olah, Csilla Keresztes, David Bremmer, Felix Gaisa, T. Nadine Kriegsmann, Joerg Kovalszky, Ilona Kiss, Andras Timar, Jozsef Szasz, Attila Marcell Rink, Michael Fisch, Margit Nyirady, Peter Szarvas, Tibor TI Prevalence of APC and PTEN Alterations in Urachal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Urachal cancer; Colorectal cancer; Molecular genetics; Mutation; APC; ß-catenin ID Urachal cancer; Colorectal cancer; Molecular genetics; Mutation; APC; ß-catenin AB Urachal carcinoma (UrC) is a rare tumor with remarkable histological and molecular similarities to colorectal cancer (CRC). Adenomatous polyposis coli (APC) is the most frequently affected gene in CRC, but the prevalence and significance of its alterations in UrC is poorly understood. In addition, loss of phosphatase and tensin homologue (PTEN) was shown to be associated with therapy resistance in CRC. Our primary aim was to assess specific genetic alterations including APC and PTEN in a large series of UrC samples in order to identify clinically significant genomic alterations. We analyzed a total of 40 UrC cases. Targeted 5-gene (APC, PTEN, DICER1, PRKAR1A, TSHR, WRN) panel sequencing was performed on the Illumina MiSeq platform(n = 34). In addition, ß-catenin (n = 38) and PTEN (n = 30) expressionswere assessed by immunohistochemistry. APC and PTEN genes were affected in 15% (5/34) and 6% (2/34) of cases. Two of five APC alterations (p.Y1075*, p.K1199*) were truncating pathogenic mutations. One of the two PTEN variants was a pathogenic frameshift insertion (p.C211fs). In 29% (11/38) of samples, at least some weak nuclear ß-catenin immunostaining was detected and PTEN loss was observed in 20% (6/30) of samples. The low prevalence of APC mutations in UrC represents a characteristic difference to CRC. Based on APC and ß-catenin results, the Wnt pathway seems to be rarely affected in UrC. Considering the formerly described involvement of PTEN protein loss in anti-EGFR therapy-resistance its immunohistochemical testing may have therapeutic relevance. C1 [Nagy, Nikolett] Semmelweis University, Department of Urology, 1082 Budapest, Hungary. [Reis, Henning] University of Duisburg-Essen, University Hospital of Essen, Institute of Pathology, 45147 Essen, Germany. [Hadaschik, Boris] University of Duisburg-Essen, Department of Urology, 45147 Essen, Germany. [Niedworok, Christian] University of Duisburg-Essen, Department of Urology, 45147 Essen, Germany. [Modos, Orsolya] Semmelweis University, Department of Urology, 1082 Budapest, Hungary. [Szendroi, Attila] Semmelweis University, Department of Urology, 1082 Budapest, Hungary. [Biro, Krisztina] National Institute of Oncology, 1122 Budapest, Hungary. [Hager, Thomas] University of Duisburg-Essen, University Hospital of Essen, Institute of Pathology, 45147 Essen, Germany. [Herold, Thomas] University of Duisburg-Essen, University Hospital of Essen, Institute of Pathology, 45147 Essen, Germany. [Ablat, Jason] University of British Columbia, Vancouver Prostate Centre & Department of Urologic Science, V6H 3Z6 Vancouver, Canada. [Black, C. Peter] University of British Columbia, Vancouver Prostate Centre & Department of Urologic Science, V6H 3Z6 Vancouver, Canada. [Okon, Krzysztof] Jagiellonian University, Department of Pathomorphology, 30252 Cracow, Poland. [Tolkach, Yuri] University of Bonn, Institute of Pathology, 53113 Bonn, Germany. [Csizmarik, Anita] Semmelweis University, Department of Urology, 1082 Budapest, Hungary. [Olah, Csilla] Semmelweis University, Department of Urology, 1082 Budapest, Hungary. [Keresztes, David] Semmelweis University, Department of Urology, 1082 Budapest, Hungary. [Bremmer, Felix] University of Gottingen, Institute of Pathology, 37073 Gottingen, Germany. [Gaisa, T. Nadine] Medical Faculty of the Technical University of Aachen, Department of Pathology, 52074 Aachen, Germany. [Kriegsmann, Joerg] Center for Histology, Cytology and Molecular Diagnostics Trier, 54296 Trier, Germany. [Kovalszky, Ilona] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, 1085 Budapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of Pathology, 1091 Budapest, Hungary. [Szasz, Attila Marcell] Semmelweis University, Department of Oncology, 1083 Budapest, Hungary. [Rink, Michael] University Medical Center Hamburg-Eppendorf, Department of Urology, 20246 Hamburg, Germany. [Fisch, Margit] University Medical Center Hamburg-Eppendorf, Department of Urology, 20246 Hamburg, Germany. [Nyirady, Peter] Semmelweis University, Department of Urology, 1082 Budapest, Hungary. [Szarvas, Tibor] Semmelweis University, Department of Urology, 1082 Budapest, Hungary. RP Szarvas, T (reprint author), Semmelweis University, Department of Urology, 1082 Budapest, Hungary. EM sztibusz@gmail.com CR Schubert GE, Pavkovic MB, Bethke-Bedurftig BA, 1982, Tubular urachal remnants in adult bladders. J Urol 127(1):40–42 Amin MB, Smith SC, Eble JN, Rao P, Choi WWL, Tamboli P, Young RH, 2014, Glandular neoplasms of the urachus: a report of 55 cases emphasizing mucinous cystic tumors with proposed classification. Am J Surg Pathol 38(8):1033–1045. , DOI 10. 1097/PAS.0000000000000250 Kumar N, Khosla D, Kumar R,Mandal AK, Saikia UN, Kapoor R, Singh SK, Sharma SC, 2014, Urachal carcinoma: clinicopathological features, treatment and outcome. J Cancer Res Ther 10(3):571– 574. , DOI 10.4103/0973-1482.137955 Reis H, Krafft U, Niedworok C, Modos O, Herold T, Behrendt M, al-Ahmadie H, Hadaschik B, Nyirady P, Szarvas T, 2018, Biomarkers in Urachal Cancer and Adenocarcinomas in the Bladder: A Comprehensive Review Supplemented by Own Data. Dis Markers:7308168. , DOI 10.1155/2018/7308168 Szarvas T, Modos O, Niedworok C et al, 2016, Clinical, prognostic, and therapeutic aspects of urachal carcinoma-a comprehensive review with meta-analysis of 1,010 cases. Urol Oncol 34(9):388– 398. , DOI 10.1016/j.urolonc.2016.04.012 Riva G, Mian C, Luchini C, Girolami I, Ghimenton C, Cima L, Novelli L, Hanspeter E, Mazzoleni G, Schwienbacher C, Pycha S, D’Elia C, Trenti E, Pycha A,Martignoni G, Hes O, Eccher A, Nesi G, Brunelli M, 2019, Urachal carcinoma: from gross specimen to morphologic, immunohistochemical, and molecular analysis. Virchows Arch 474(1):13–20. , DOI 10.1007/s00428- 018-2467-1 Reis H, van der Vos KE, Niedworok C, Herold T, Modos O, Szendroi A, Hager T, Ingenwerth M, Vis DJ, Behrendt MA, de Jong J, van der Heijden MS, Peyronnet B, Mathieu R, Wiesweg M, Ablat J, Okon K, Tolkach Y, Keresztes D, Nagy N, Bremmer F, Gaisa NT, Chlosta P, Kriegsmann J, Kovalszky I, Timar J, Kristiansen G, Radzun HJ, Knuchel R, Schuler M, Black PC, Rubben H, Hadaschik BA, Schmid KW, van Rhijn BWG, Nyirady P, Szarvas T, 2018, Pathogenic and targetable genetic alterations in 70 urachal adenocarcinomas. Int J Cancer 143: 1764–1773. , DOI 10.1002/ijc.31547 Modos O, Reis H, Niedworok C et al, 2016, Mutations of KRAS, NRAS, BRAF, EGFR, and PIK3CA genes in urachal carcinoma: Occurence and prognostic significance. Oncotarget 7(26):39293– 39301. , DOI 10.18632/oncotarget.9828 Kardos J, Wobker SE,WoodsME, NielsenME, Smith AB,Wallen EM, Pruthi RS, Hayward MC, McGinty KA, Grilley-Olson JE, Patel NM, Weck KE, Black P, Parker JS, Milowsky MI, Hayes DN, Kim WY, 2017, Comprehensive molecular characterization of Urachal adenocarcinoma reveals commonalities with colorectal Cancer, including a Hypermutable phenotype. JCO Prec Oncol 1(1):1–12. , DOI 10.1200/po.17.00027 Lee S, Lee J, Sim SH, Lee Y, Moon KC, Lee C, Park WY, Kim NKD, Lee SH, Lee H, 2017, Comprehensive somatic genome alterations of urachal carcinoma. JMed Genet 54(8):572–578. https:// doi.org/10.1136/jmedgenet-2016-104390 Hang JF, Pan CC, 2017, Absence of GNAS and BRAF mutations but presence of KRAS mutation in urachal adenocarcinoma. Pathology. 49(3):316–317. , DOI 10.1016/j.pathol.2016. 11.017 Sirintrapun SJ,WardM,Woo J, CimicA(2014, High-stage urachal adenocarcinoma can be associated with microsatellite instability and KRAS mutations. Hum Pathol 45(2):327–330. , DOI 10.1016/j.humpath.2013.09.008 Loh KP, Mondo E, Hansen EA, Sievert L, Fung C, Sahasrabudhe DM, Guancial E, 2016, Targeted therapy based on tumor genomic analyses in metastatic Urachal carcinoma. Clin Genitourin Cancer 14(4):e449–e452. , DOI 10.1016/j.clgc.2016.03.013 Collazo-Lorduy A, Castillo-Martin M, Wang L, Patel V, Iyer G, Jordan E, al-Ahmadie H, Leonard I, Oh WK, Zhu J, McBride RB, Cordon-Cardo C, Solit DB, Sfakianos JP, Galsky MD, 2016, Urachal Carcinoma Shares Genomic Alterations with Colorectal Carcinoma and May Respond to Epidermal Growth Factor Inhibition. Eur Urol 70(5):771–775. , DOI 10.1016/j. eururo.2016.04.037 Cha S, Lee J, Shin JY, Kim JY, Sim SH, Keam B, Kim TM, Kim DW, Heo DS, Lee SH, Kim JI, 2016, Clinical application of genomic profiling to find druggable targets for adolescent and young adult, AYA, cancer patients with metastasis. BMC Cancer 16: 170. , DOI 10.1186/s12885-016-2209-1 Singh H, Liu Y, Xiao X et al, 2016, Whole exome sequencing of urachal adenocarcinoma reveals recurrent NF1 mutations. Oncotarget 7(20):29211–29215. , DOI 10.18632/ oncotarget.8640 Cancer Genome Atlas N, 2012, Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487(7407): 330–337. , DOI 10.1038/nature11252 Cornejo KM, Paner GP, Tomaszewicz K et al, 2016, Mutational profile using next generation sequencing may aid in distinguishing Urachal adenocarcinoma from bladder adenocarcinoma nature modern pathology, 105th annual meeting of the United States and Canadian academy of pathology, USCAP), 2015, Seattle. Washington. 29:528–556. , DOI 10.1038/modpathol. 2016.25 Roy S, Smith MA, Cieply KM, Acquafondata MB, Parwani AV, 2012, Primary bladder adenocarcinoma versus metastatic colorectal adenocarcinoma: a persisting diagnostic challenge. Diagn Pathol 7:151. , DOI 10.1186/1746-1596-7-151 Aoki K, Taketo MM, 2007, Adenomatous polyposis coli, APC): a multi-functional tumor suppressor gene. J Cell Sci 120(19):3327– 3335. , DOI 10.1242/jcs.03485 Yazdani Y, Farazmandfar T, Azadeh H, Zekavatian Z, 2016, The prognostic effect of PTEN expression status in colorectal cancer development and evaluation of factors affecting it: miR-21 and promoter methylation. J Biomed Sci 23:9. , DOI 10.1186/ s12929-016-0228-5 FrattiniM, Saletti P, Romagnani E,Martin V,Molinari F, Ghisletta M, Camponovo A, Etienne LL, Cavalli F, Mazzucchelli L, 2007, PTEN loss of expression predicts cetuximab efficacy in metastatic colorectal cancer patients. Br J Cancer 97(8):1139–1145. https:// doi.org/10.1038/sj.bjc.6604009 Sheldon CA, Clayman RV, Gonzalez R, Williams RD, Fraley EE, 1984, Malignant urachal lesions. J Urol 131(1):1–8 Toubaji A, Jordan EJ, Desai N et al, 2016, Genomic Alterations in Primary Bladder Adenocarcinoma and Urachal Adenocarcinoma. Nature Modern Pathology, 106th Annual Meeting of the United States and Canadian Academy of Pathology, USCAP), 2016, San Antonio, Texas. 30:539. , DOI 10.1038/modpathol.2016. 265 Pires-Luis A, Martinek P, Filipovic J et al, 2018, Primary adenocarcinoma of the urinary bladder: next-generation sequencing, NGS, of non-urachal enteric-type adenocarcinomas, Urachal adenocarcinomas, mucinous adenocarcinomas, and colonic Metaplasias/adenomas. Drugs Future 43:359. , DOI 10. 1358/dof.2018.043.05.2808557 Lee B, Jordan E, Won H, Bagrodia A, Desai N, Bajorin D, Rosenberg J, Bochner B, Kim W, Berger M, Solit D, al-Ahmadie H, Iyer G, 2016, Mutational landscape of primary bladder and Urachal adenocarcinoma. J Urol 195(4S):e1133–e11e4. https:// doi.org/10.1016/j.juro.2016.02.2431 Wong SZ, Lo E, Lee K et al, 2004, Prognostic and diagnostic significance of ß-catenin nuclear Immunostaining in colorectal Cancer. Clin Cancer Res 10(4):1401–1408. , DOI 10. 1158/1078-0432.CCR-0157-03 Alomar SY, Mansour L, Abuderman A, Alkhuriji A, Arafah M, Alwasel S, Harrath AH, Almutairi M, Trayhyrn P, Dar JA, 2016, β-Catenin accumulation and S33F mutation of CTNNB1 gene in colorectal cancer in Saudi Arabia. Pol J Pathol 67(2):156–162. , DOI 10.5114/pjp.2016.61452 Perrone F, Lampis A, Orsenigo M, di BartolomeoM, GevorgyanA, Losa M, FrattiniM, Riva C, Andreola S, Bajetta E, Bertario L, Leo E, Pierotti MA, Pilotti S, 2009, PI3KCA/PTEN deregulation contributes to impaired responses to cetuximab in metastatic colorectal cancer patients. Ann Oncol 20(1):84–90. , DOI 10.1093/ annonc/mdn541 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2773 EP 2781 DI 10.1007/s12253-020-00872-6 PG 9 ER PT J AU Major, T Gindele, R Szabo, Zs Kis, Zs Bora, L Joni, N Bardossy, P Racz, T Bereczky, Zs AF Major, Tamas Gindele, Reka Szabo, Zsuzsanna Kis, Zsuzsanna Bora, Laszlo Joni, Natalia Bardossy, Peter Racz, Tamas Bereczky, Zsuzsanna TI The Stratified Population Screening of Hereditary Hemorrhagic Telangiectasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hereditary hemorrhagic telangiectasia; Prevalence; Stratified screening; ACVRL1; ENG; Founder effect ID Hereditary hemorrhagic telangiectasia; Prevalence; Stratified screening; ACVRL1; ENG; Founder effect AB Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant multisystemic vascular disease with a worldwide prevalence of 1:5000–1:10000. We introduce our algorithm for the stratified population screening of HHT. Probands are selected from the consecutive hospital database review for HHT (I7800) and recurrent epistaxis (R0400) and the review of patient records referred by family practitioners. A proportion of probands might be de novo diagnosed with HHT in the 10-year study period. The checkup of probands consists of physical examination, arteriovenous malformation exploration and genetic testing (ACVRL1 and ENG sequence analysis). The family screening of HHT consists of physical examination and screening for the family-specific mutation of each at-risk individual, and furthermore, arteriovenous malformation exploration in individuals with suspected/definite HHT and/or carrying the mutation. Twenty-five definite HHT patients were explored: 7 of them by the I7800 review, 1 by the R0400 review, 3 were de novo diagnosed, and the remaining 14 were explored by the systematic family screening. Considering the 20 patients alive at the end of the study period and the unavailable 5 potential HHT patients and 12 at-risk family members, the HHT prevalence is estimated to be 1:6090–1:11267 in our study area, implying our algorithm’s effectivity in the stratified population screening of HHT. C1 [Major, Tamas] B-A-Z County Central Hospital and University Teaching Hospital, Department of Otolaryngology and Head and Neck Surgery, Szentpeteri kapu 72-76, H-3526 Miskolc, Hungary. [Gindele, Reka] University of Debrecen, Faculty of Medicine, Department of Laboratory Medicine, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Szabo, Zsuzsanna] University of Debrecen, Faculty of Medicine, Department of Laboratory Medicine, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. [Kis, Zsuzsanna] Ferenc Markhot County Hospital, Department of Radiology, Szechenyi u. 27-29, H-3300 Eger, Hungary. [Bora, Laszlo] Szent Lazar County Hospital, Department of Radiology, Fuleki ut 54-56, H-3100 Salgotarjan, Hungary. [Joni, Natalia] Ferenc Markhot County Hospital, Department of Internal Medicine, Szechenyi u. 27-29, H-3300 Eger, Hungary. [Bardossy, Peter] Hungarian Heraldry and Genealogical Society, Becsi Kapu ter 2-4, H-1014 Budapest, Hungary. [Racz, Tamas] Ferenc Markhot County Hospital, Department of Otorhinolaryngology, Szechenyi u. 27-29, H-3300 Eger, Hungary. [Bereczky, Zsuzsanna] University of Debrecen, Faculty of Medicine, Department of Laboratory Medicine, Nagyerdei krt. 98, H-4032 Debrecen, Hungary. RP Major, T (reprint author), B-A-Z County Central Hospital and University Teaching Hospital, Department of Otolaryngology and Head and Neck Surgery, H-3526 Miskolc, Hungary. EM majordoki2@gmail.com CR Shovlin CL, Guttmacher AE, Buscarini E, Faughnan ME, Hyland RH, Westermann CJ, Kjeldsen AD, Plauchu H, 2000, Diagnostic criteria for hereditary hemorrhagic telangiectasia, Rendu-Osler-weber syndrome). Am J Med Genet 91:66–67 Sharathkumar AA, Shapiro A, 2008, Hereditary haemorrhagic telangiectasia. Haemophilia 14:1269–1280. , DOI 10.1111/j. 1365-2516.2008.01774.x University of Utah, Department of Pathology. HHT Mutation Database. http://arup.utah.edu/database/HHT/. Accessed December 3rd, 2018 Faughnan ME, Palda VA, Garcia-Tsao G, Geisthoff UW, McDonald J, Proctor DD, Spears J, Brown DH, Buscarini E, Chesnutt MS, Cottin V, Ganguly A, Gossage JR, Guttmacher AE, Hyland RH, Kennedy SJ, Korzenik J, Mager JJ, Ozanne AP, Piccirillo JF, Picus D, Plauchu H, Porteous ME, Pyeritz RE, Ross DA, Sabba C, Swanson K, Terry P, Wallace MC, Westermann CJ, White RI, Young LH, Zarrabeitia R, 2011, International guidelines for the diagnosis and management of hereditary haemorrhagic telangiectasia. J Med Genet 48:73–87. , DOI 10.1136/jmg. 2009.069013 Bayrak-Toydemir P, McDonald J,Markewitz B, Lewin S,Miller F, Chou LS, Gedge F, Tang W, Coon H, Mao R, 2006, Genotypephenotype correlation in hereditary hemorrhagic telangiectasia: mutations and manifestations. Am J Med Genet 140: 463–470. , DOI 10.1002/ajmg.a.31101 Lesca G, Genin E, Blachier C, Olivieri C, Coulet F, Brunet G, Dupuis-Girod S, Buscarini E, Soubrier F, Calender A, Danesino C, Giraud S, Plauchu H, 2008, Hereditary hemorrhagic telangiectasia: evidence for regional founder effects of ACVRL1 mutations in French and Italian patients. Eur J Hum Genet 16:742–749. , DOI 10.1038/ejhg.2008.3 Brusgaard K, Kjeldsen AD, Poulsen L, Moss H, Vase P, Rasmussen K, Kruse TA, HorderM(2004)Mutations in endoglin and in activin receptor-like kinase 1 among Danish patients with hereditary haemorrhagic telangiectasia. Clin Genet 66:556–561. , DOI 10.1111/j.1399-0004.2004.00341.x Heimdal K, Dalhus B, Rodningen OK, Kroken M, Eiklid K, Dheyauldeen S, Roysland T, Andersen R, Kulseth MA, 2015, Mutation analysis in Norwegian families with hereditary hemorrhagic telangiectasia: founder mutations in ACVRL1. Clin Genet 89:182–186. , DOI 10.1111/cge.12612 Major T, Gindele R, Szabo Z, Alef T, Thiele B, Bora L, Kis Z, Bardossy P, Racz T, Havacs I, Bereczky Z, 2016, Evidence for the founder effect of a novel ACVRL1 splice-site mutation in Hungarian hereditary hemorrhagic telangiectasia families. Clin Genet 90:466–467 Torring PM, Brusgaard K, Ousager LB, Andersen PE, Kjeldsen AD, 2014, National mutation study among Danish patients with hereditary haemorrhagic telangiectasia. Clin Genet 86:123–133. , DOI 10.1111/cge.12269, , DOI 10.1111/cge.12806 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2783 EP 2788 DI 10.1007/s12253-019-00602-7 PG 6 ER PT J AU Sangam, K Kumar, Y Walker Minz, R Varma, N Varma, S Anand, Sh AF Sangam, Kumar Kumar, Yashwant Walker Minz, Ranjana Varma, Neelam Varma, Subhash Anand, Shashi TI Patients with Plasma Cell Disorders Have High EBV DNA in Peripheral Blood than the General Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Epstein - Barr virus; Plasma cell disorders; Quantitative real time PCR; Immunocompetent ID Epstein - Barr virus; Plasma cell disorders; Quantitative real time PCR; Immunocompetent AB Epstein-Barr virus (EBV) is involved in the development of a wide range of B cell lympho-proliferative disorders. Its association with plasma cell disorders (PCD) however is not clear, especially in immunocompetent patients. To explore any relationship, 39 patients of suspected PCD with positive M-band on electrophoresis and 50 healthy controls were enrolled. EBV DNA in peripheral blood was quantified using quantitative Real Time Polymerase Chain Reaction (qPCR). Of 39 patients, 15 (38.5%) had EBV DNA compared to 8/50 (16%) controls (p = 0.0008). The mean viral copy number was found to be significantly high in patients compared to controls (1.8 × 105; range = 2.6 × 103–7.6 × 105 copies/ml and 1.7 × 104; range = 7.0 × 102–6.1 × 104 copies/ml respectively; p = 0.003). This is the first study, which characterizes the frequency of EBV in circulation in patients of PCD. The significance of increased prevalence of circulating EBV and a higher viral load in our immunocompetent patients however, needs further evaluation. C1 [Sangam, Kumar] Post Graduate Institute of Medical Education & Research, Department of ImmunopathologyChandigarh, India. [Kumar, Yashwant] Post Graduate Institute of Medical Education & Research, Department of ImmunopathologyChandigarh, India. [Walker Minz, Ranjana] Post Graduate Institute of Medical Education & Research, Department of ImmunopathologyChandigarh, India. [Varma, Neelam] Post Graduate Institute of Medical Education & Research, Department of ImmunopathologyChandigarh, India. [Varma, Subhash] Post Graduate Institute of Medical Education & Research, Department of Internal MedicineChandigarh, India. [Anand, Shashi] Post Graduate Institute of Medical Education & Research, Department of ImmunopathologyChandigarh, India. RP Kumar, Y (reprint author), Post Graduate Institute of Medical Education & Research, Department of Immunopathology, Chandigarh, India. EM dryashwant@ymail.com CR Babel N, Schwarzmann F, Pruss A, Volk HD, Reinke P, 2004, Monoclonal gammopathy of undetermined significance, MGUS, is associated with an increased frequency of Epstein-Barr virus, EBV, latently infected B lymphocytes in long-term renal transplant patients. Transplant Proc 36:2679–2682 Giordano M, Santangelo L, Scarasciulli ML, Calvario A, Miragliotta G, Giordano P, Cecinati V, 2014, Monoclonal gammopathy of undetermined significance in pediatric kidney transplant: a possible role of Epstein-Barr virus. Pediatr Transplant 18:42–46 Hsieh AT, Ho CL, Chen YC et al, 2002, Epstein-Barr virus and lymphoid hematological disorders. ZonghuaYi Xue Za Zhi, Taipei, 65:119–123 Chang ST, Liao YL, Lu CL, Chuang SS, Li CY, 2007, Plasmablastic cytomorphologic features in plasma cell neoplasms in immunocompetent patients are significantly associated with EBV. Am J Clin Pathol 128(2):339–344 Nael A, Wu WW, Siddiqi I et al Epstein - Barr virus association with. Plasma Cell Neoplasms Histol Histopath 2018Nov 19:18066. , DOI 10.14670/HH-18-066. [Epub ahead of print] Lin JC,Wan gWY, Chen KYet al, 2004, Quantification of plasma Epstein-Barr virus DNA in patients with advanced nasopharyngeal carcinoma. N Engl J Med 350:2461–2470 Zhang Y, Zhao H, He X, Zheng S, Wang T, Yan D, Sun J, Lu X, Wen J, Lau WY, 2012, Effects of Epstein-Barr virus infection on the development of multiple myeloma after liver transplantation. Sci China Life Sci 55:735–743 Thorley-Lawson DA, Babcock GJ, 1999, A model for persistent infection with Epstein-Barr virus: the stealth virus of human B cells. Life Sci 65(14):1433–1453 Sasaki S, Hashimoto K, Nakatsuka S, Hasegawa M, Nakano T, Nagata S, Kanakura Y, Hayashi N, 2011, Plasmablastic extramedullary plasmacytoma associated with Epstein-Barr virus arising in an immunocompetent patient with multiple myeloma. Intern Med 50:2615–2620 Chen H, Hutt-Fletcher L, Cao L, Hayward SD, 2003, A positive autoregulatory loop of lmp1 expression and stat activation in epithelial cells latently infected with Epstein-Barr virus. J Virol 77:4139–4148 Laichalk LL, Thorley-Lawson DA, 2005, Terminal differentiation into plasma cells initiates the replicative cycle of Epstein-Barr virus in vivo. J Virol 79:1296–1307 SadeghianMH, Ayatollahi H, Keramati MR(2011, The association of Epstein-Barr virus infection with multiple myeloma. Ind J Pathol Microbiol 54:720–724 Tomita Y, Ohsawa M, Hashimoto M, 1998, Plasmacytoma of the gastrointestinal tract in Korea: higher incidence than in Japan and Epstein-Barr virus association. Oncology 55(1):27–32 Hoebeeck J, Speleman F, Vandesompele J, 2007, Real-time quantitative PCR as an alternative to southern blot or fluorescence in situ hybridization for detection of gene copy number changes. Methods Mol Biol 353:205–226 Lunemann JD, Tackenberg B, Stein A,Wandinger KP, Oertel WH, Wagner HJ, Munz C, Meisel H, Sommer N, Zipp F, 2010, Dysregulated Epstein-Barr virus infection in patients with CIDP. J Neuroimmunol 218(1–2):107–111 Chang YT, Liu HN, Wong CK, Chow KC, Chen KY, 1997, Detection of Epstein-Barr virus in primary cutaneous amyloidosis. Br J Dermatol 136(6):823–826 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2789 EP 2794 DI 10.1007/s12253-019-00640-1 PG 6 ER PT J AU Nassereddine, H Sannier, A Brosseau, S Rodier, JM Khalil, A Msika, S Danel, C Couvelard, A Theou-Anton, N Cazes, A AF Nassereddine, Hussein Sannier, Aurelie Brosseau, Solenn Rodier, Jean-Michel Khalil, Antoine Msika, Simon Danel, Claire Couvelard, Anne Theou-Anton, Nathalie Cazes, Aurelie TI Clinicopathological and Molecular Study of Peritoneal Carcinomatosis Associated with Non-Small Cell Lung Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Peritoneal carcinomatosis; Non-small cell lung carcinoma; NSCLC; Massively-parallel sequencing; Molecular pathology ID Peritoneal carcinomatosis; Non-small cell lung carcinoma; NSCLC; Massively-parallel sequencing; Molecular pathology AB To retrospectively characterize the molecular features of Non-Small Cell Lung Carcinomas (NSCLC) with peritoneal carcinomatosis (PC), clinicopathological data of 12 patients diagnosed with NSCLC and PC between 2007 and 2016 were collected. Immunohistochemistry and Next Generation Sequencing (NGS) were performed on cases with available material. PC was the initial presentation of NSCLC in 17% of the cases. Overall, patients with PC displayed a poor median survival of 12 weeks. Histology was adenocarcinoma in 11 cases. 37.5% of cases showed PD-L1 immunostaining positivity (50% cut-off). ALK and ROS1 immunostainings were negative. Using NGS, we identified 17 molecular alterations in 9 genes (TP53, KRAS, STK11, BRAF, EGFR, DDR2, ERBB4, SMAD4, CTNNB1) in 88.9% of adenocarcinomas. To the best of our knowledge, 5 of these variants are not referenced in the literature. In conclusion, PC might be the initial presentation of NSCLC. Molecular profiling of our cases did not find any effective targetable alteration, except from high PD-L1 expression. C1 [Nassereddine, Hussein] Assistance Publique – Hopitaux de Paris, Hopital Bichat-Claude Bernard, departement de pathologieParis, France. [Sannier, Aurelie] Assistance Publique – Hopitaux de Paris, Hopital Bichat-Claude Bernard, departement de pathologieParis, France. [Brosseau, Solenn] University of Paris 5Paris, France. [Rodier, Jean-Michel] APHP, Hopital Bichat-Claude Bernard, service d’OncologieParis, France. [Khalil, Antoine] University of Paris 5Paris, France. [Msika, Simon] University of Paris 5Paris, France. [Danel, Claire] Assistance Publique – Hopitaux de Paris, Hopital Bichat-Claude Bernard, departement de pathologieParis, France. [Couvelard, Anne] Assistance Publique – Hopitaux de Paris, Hopital Bichat-Claude Bernard, departement de pathologieParis, France. [Theou-Anton, Nathalie] APHP, Hopital Bichat-Claude Bernard, laboratoire de GenetiqueParis, France. [Cazes, Aurelie] Assistance Publique – Hopitaux de Paris, Hopital Bichat-Claude Bernard, departement de pathologieParis, France. RP Nassereddine, H (reprint author), Assistance Publique – Hopitaux de Paris, Hopital Bichat-Claude Bernard, departement de pathologie, Paris, France. EM hussein.nassereddine@hotmail.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F, 2015, Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136(5):E359–E386 Satoh H, Ishikawa H, Yamashita YT, Kurishima K, Ohtsuka M, Sekizawa K, 2001, Peritoneal carcinomatosis in lung cancer patients. Oncol Rep 8(6):1305–1307 Patil T, Aisner DL, Noonan SA, Bunn PA, Purcell WT, Carr LL, Camidge DR, Doebele RC, 2016, Malignant pleural disease is highly associated with subsequent peritoneal metastasis in patients with stage IV non-small cell lung cancer independent of oncogene status. Lung Cancer 96:27–32 Sereno M, Rodriguez-Esteban I, Gomez-Raposo C, Merino M, Lopez-Gomez M, Zambrana F et al, 2013, Lung cancer and peritoneal carcinomatosis. Oncol Lett 6(3):705–708 Su H-T, Tsai C-M, Perng R-P, 2008, Peritoneal carcinomatosis in lung cancer. Respirol 13(3):465–467 McNeill PM, Wagman LD, Neifeld JP, 1987, Small bowel metastases from primary carcinoma of the lung. Cancer 59(8):1486–1489 Okami J, Taniguchi K, Higashiyama M, Maeda J, Oda K, Orita N, Koizumi K, Kodama K, Kato K, 2007, Prognostic factors for gefitinib-treated postoperative recurrence in non-small cell lung cancer. Oncology 72(3–4):234–242 Klughammer B, BruggerW, Cappuzzo F, Ciuleanu T,Mok T, Reck M, Tan EH, Delmar P, Klingelschmitt G, Yin AY, Spleiss O,Wu L, Shames DS, 2016, Examining treatment outcomeswith Erlotinib in patients with advanced non-small cell lung cancer whose tumors harbor uncommon EGFR mutations. J Thorac Oncol 11(4):545– 555 Shajani-Yi Z, de Abreu FB, Peterson JD, Tsongalis GJ, 2018, Frequency of somatic TP53 mutations in combination with known pathogenicmutations in colon adenocarcinoma, non-small cell lung carcinoma, and gliomas as identified by next-generation sequencing. Neoplasia 20(3):256–262 Marte B, 2013, Tumour heterogeneity. Nature 501:327 Bhattacharya S, SocinskiMA, Burns TF, 2015, KRAS mutant lung cancer: progress thus far on an elusive therapeutic target. Clin Transl Med 4(1):35 Del Re M, Rofi E, Restante G, Crucitta S, Arrigoni E, Fogli S et al, 2017, Implications of KRAS mutations in acquired resistance to treatment in NSCLC. Oncotarget 9(5):6630–6643 Ding L, Getz G,Wheeler DA, Mardis ER,McLellanMD, Cibulskis K et al, 2008, Somatic mutations affect key pathways in lung adenocarcinoma. Nature 455(7216):1069–1075 Pecuchet N, Laurent-Puig P, Mansuet-Lupo A, Legras A, Alifano M, Pallier K et al, 2017, Different prognostic impact of STK11 mutations in non-squamous non-small-cell lung cancer. Oncotarget 8(14):23831–23840 Shinozaki E, Yoshino T, Yamazaki K, Muro K, Yamaguchi K, Nishina T et al, 2017, Clinical significance of BRAF non-V600E mutations on the therapeutic effects of anti-EGFR monoclonal antibody treatment in patients with pretreated metastatic colorectal cancer: the Biomarker Research for anti-EGFR monoclonal Antibodies by Comprehensive Cancer genomics, BREAC, study. Br J Cancer 117(10):1450–1458 Carter J, Tseng L-H, Zheng G, Dudley J, Illei P, Gocke CD, Eshleman JR, Lin MT, 2015, Non-p.V600E BRAF mutations are common using amore sensitive and broad detection tool.AmJ Clin Pathol 144(4):620–628 Heidorn SJ, Milagre C,Whittaker S, Nourry A, Niculescu-Duvas I, Dhomen N, Hussain J, Reis-Filho JS, Springer CJ, Pritchard C, Marais R, 2010, Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell 140(2):209–221 Cancer Genome Atlas Research Network, 2014, Comprehensive molecular profiling of lung adenocarcinoma. Nature 511(7511): 543–550 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2795 EP 2800 DI 10.1007/s12253-019-00713-1 PG 6 ER PT J AU Burian, A Lujber, L Gerlinger, I Jarai, T Orosz, Turiak, L Acs, A Hegedus, Z Konigne Peter, A Tornoczki, T Gombos, K Mark, L AF Burian, Andras Lujber, Laszlo Gerlinger, Imre Jarai, Tamas Orosz, Eva Turiak, Lilla Acs, Andras Hegedus, Zoltan Konigne Peter, Aniko Tornoczki, Tamas Gombos, Katalin Mark, Laszlo TI Label-Free Semiquantitative Liquid Chromatography-Tandem Mass Spectrometry Proteomics Analysis of Laryngeal/Hypopharyngeal Squamous Cell Carcinoma on Formalin-Fixed, Paraffin-Embedded Tissue Samples - a Pilot Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Biomarker discovery; Laryngeal cancer; LC/MS; Squamous cell carcinoma; Proteomics ID Biomarker discovery; Laryngeal cancer; LC/MS; Squamous cell carcinoma; Proteomics AB Squamous cell carcinoma (SCC) of the head and neck region is the sixth most frequent malignancy with high mortality rate. Due to its poor prognosis it is considered a growing public health problem worldwide inspite of existing treatment modalities. Thus, early diagnosis of new diseases and recurrences is emerging on one hand, but on the other hand troublesome in the lack of reliable tumor markers in this field. The rapid development of proteomics has opened new perspectives in tumor marker discovery. Liquid chromatography/mass spectrometry (LC/MS) as the gold standard in proteomics enables the semi-quantitative analysis of proteins within various tissues. Abundance differences between tumor and normal tissue also can be interpreted as tumor specific changes. The aimof this study was to identify potential tumor markers of laryngeal/hypopharyngeal SCC by revealing abundance changes between cancerous and the surrounding phenotypically healthy tissue. After separating the phenotypically cancerous and healthy parts of formalin-fixed paraffin-embedded tissues, each sample underwent protein recovery process and tryptic digestion for label-free semi-quantitative LC/MS analysis. Eight proteins showed significantly higher abundance in tumor including tenascin, transmembrane emp24 domain-containing protein 2, cytoplasmic dynein light chain 1, coactosin-like protein, small proline-rich protein 2D, nucleolin, U5 small nuclear RNP 200-kDa helicase and fatty aldehyde dehydrogenase. Desmoglein-1 and keratin type I cytoskeletal 9 were down-regulated in tumor. Using Ingenuity Pathway Analysis we mapped the signaling pathways these proteins play role in regarding other tumors. Based on these findings these proteins may serve as promising biomarkers in the fight against laryngeal/hypopharyngeal SCCs. C1 [Burian, Andras] PTE KK, Ful-Orr-Gegeszeti es Fej-Nyaksebeszeti Klinika, Munkacsy M Str 2, H-7621 Pecs, Hungary. [Lujber, Laszlo] PTE KK, Ful-Orr-Gegeszeti es Fej-Nyaksebeszeti Klinika, Munkacsy M Str 2, H-7621 Pecs, Hungary. [Gerlinger, Imre] PTE KK, Ful-Orr-Gegeszeti es Fej-Nyaksebeszeti Klinika, Munkacsy M Str 2, H-7621 Pecs, Hungary. [Jarai, Tamas] Tolna County Balassa Janos Hospital, Beri Balogh Adam Str 5-7, H-7100 Szekszard, Hungary. [Orosz, Eva] PTE KK, Ful-Orr-Gegeszeti es Fej-Nyaksebeszeti Klinika, Munkacsy M Str 2, H-7621 Pecs, Hungary. [Turiak, Lilla] Hungarian Academy of Sciences, Research Centre for Natural Sciences, MS Proteomics Research Group, Magyar tudosok Blvd 2, H-1117 Budapest, Hungary. [Acs, Andras] Hungarian Academy of Sciences, Research Centre for Natural Sciences, MS Proteomics Research Group, Magyar tudosok Blvd 2, H-1117 Budapest, Hungary. [Hegedus, Zoltan] Hungarian Academy of Sciences, Biological Research Centre, Institute of Biophysics, Temesvari Blvd 62, H-6726 Szeged, Hungary. [Konigne Peter, Aniko] PTE KK, Bioanalitikai Intezet, Honved Str 1, H-7624 Pecs, Hungary. [Tornoczki, Tamas] University of Pecs, Department of Pathology, Szigeti Str 12, H-7624 Pecs, Hungary. [Gombos, Katalin] University of Pecs, Department of Laboratory Medicine, Ifjusag Str 13, H-7624 Pecs, Hungary. [Mark, Laszlo] University of Pecs, Institute of Biochemistry and Medical Chemistry, Szigeti Str 12, H-7624 Pecs, Hungary. RP Mark, L (reprint author), University of Pecs, Institute of Biochemistry and Medical Chemistry, H-7624 Pecs, Hungary. EM laszlo.mark@aok.pte.hu CR Marur S, Forastiere AA, 2008, Head and neck cancer: changing epidemiology, diagnosis, and treatment. Mayo Clin Proc 83:489– 501 Hashibe M, Brennan P, Chuang SC, Boccia S, Castellsague X, Chen C, Curado MP, Dal Maso L, Daudt AW, Fabianova E, Fernandez L, Wunsch-Filho V, Franceschi S, Hayes RB, Herrero R, Kelsey K, Koifman S, la Vecchia C, Lazarus P, Levi F, Lence JJ, Mates D,Matos E, Menezes A,McCleanMD,Muscat J, Eluf-Neto J, Olshan AF, PurdueM, Rudnai P, Schwartz SM, Smith E, Sturgis EM, Szeszenia-Dabrowska N, Talamini R, Wei Q, Winn DM, Shangina O, Pilarska A, Zhang ZF, Ferro G, Berthiller J, Boffetta P, 2009, Interaction between tobacco and alcohol use and the risk of head and neck Cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. Cancer Epidemiol Biomark Prev 18:541–550 Argiris A, Karamouzis MV, Raben D, Ferris RL, 2008, Head and neck cancer. Lancet 371:1695–1709 Erich K, Reinle K, Muller T, Munteanu B, Sammour DA, Hinsenkamp I, Gutting T, Burgermeister E, Findeisen P, Ebert MP, Krijgsveld J, Hopf C, 2019, Spatial distribution of endogenous tissue protease activity in gastric carcinoma mapped by MALDI mass spectrometry imaging. Mol Cell Proteomics 18: 151–161 Hanahan D, Coussens LM, 2012, Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21:309–322 Paulo JA, Lee LS, Banks PA, Steen H, Conwell DL, 2012, Proteomic analysis of formalin-fixed paraffin-embedded pancreatic tissue using liquid chromatography tandem mass spectrometry. Pancreas 41:175–185 Carnielli CM, Macedo CCS, De Rossi T, Granato DC, Rivera C, Domingues RR et al, 2018, Combining discovery and targeted proteomics reveals a prognostic signature in oral cancer. Nat Commun 9:3598 Li L, Zhang Z,Wang C, Miao L, Zhang J, Wang J, Jiao B, Zhao S, 2014, Quantitative proteomics approach to screening of potential diagnostic and therapeutic targets for laryngeal carcinoma. PLoS One 9:e90181 Metz B, Kersten GF, Hoogerhout P, Brugghe HF, Timmermans HA, de Jong A et al, 2004, Identification of formaldehydeinduced modifications in proteins: reactions with model peptides. J Biol Chem 279:6235–6243 Fowler CB, O'Leary TJ, Mason JT et al, 2013, Toward improving the proteomic analysis of formalin-fixed, paraffin-embedded tissue. Expert Rev Proteomics 10:389–400 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2801 EP 2807 DI 10.1007/s12253-020-00849-5 PG 7 ER PT J AU Son, JH Mo, YH Choi, JE Yoo, JN Lee, HS AF Son, Ji Hyun Mo, Yoon Ha Choi, Ji Eun Yoo, Jin Nam Lee, Hyung Sug TI Promoter Mutation Analysis of Long-Non-coding RNA RMRP Gene in Solid Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Son, Ji Hyun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Mo, Yoon Ha] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Rheinbay E, Parasuraman P, Grimsby J, Tiao G, Engreitz JM, Kim J, Lawrence MS, Taylor-Weiner A, Rodriguez-Cuevas S, Rosenberg M, Hess J, Stewart C, Maruvka YE, Stojanov P, Cortes ML, Seepo S, Cibulskis C, Tracy A, Pugh TJ, Lee J, Zheng Z, Ellisen LW, Iafrate AJ, Boehm JS, Gabriel SB, Meyerson M, Golub TR, Baselga J, Hidalgo-Miranda A, Shioda T, Bernards A, Lander ES, Getz G, 2017, Recurrent and functional regulatory mutations in breast cancer. Nature 547:55–60 Huarte M, 2015, The emerging role of lncRNAs in cancer. Nat Med 21:1253–1261 Hermanns P, Bertuch AA, Bertin TK, Dawson B, Schmitt ME, Shaw C, Zabel B, Lee B, 2005, Consequences of mutations in the noncoding RMRP RNA in cartilage-hair hypoplasia. Hum Mol Genet 14:3723–3740 Xiao X, Gu Y,Wang G, Chen S, 2019, c-Myc, RMRP, andmiR-34a- 5p form a positive-feedback loop to regulate cell proliferation and apoptosis in multiple myeloma. Int J Biol Macromol 122:526–537 Shao Y, Ye M, Li Q, SunW, Ye G, Zhang X, Yang Y, Xiao B, Guo J, 2016, LncRNA-RMRP promotes carcinogenesis by acting as a miR-206 sponge and is used as a novel biomarker for gastric cancer. Oncotarget 7:37812–37824 Yoo NJ, Kim HR, Kim YR, An CH, Lee SH, 2012, Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology 60:943–952 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2809 EP 2810 DI 10.1007/s12253-019-00723-z PG 2 ER PT J AU Son, JH Choi, JE Yoo, JN Lee, HS AF Son, Ji Hyun Choi, Ji Eun Yoo, Jin Nam Lee, Hyung Sug TI Intratumoral heterogeneity of FLCN somatic mutations in gastric and colorectal cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Son, Ji Hyun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Hartman TR, Nicolas E, Klein-Szanto A, Al-Saleem T, Cash TP, Simon MC, Henske EP, 2009, The role of the Birt-Hogg-Dube protein in mTOR activation and renal tumorigenesis. Oncogene 28: 1594–1604 Khoo SK, Giraud S, Kahnoski K, Chen J, Motorna O, Nickolov R, Binet O, Lambert D, Friedel J, Levy R, Ferlicot S, Wolkenstein P, Hammel P, Bergerheim U, Hedblad MA, Bradley M, Teh BT, Nordenskjold M, Richard S, 2002, Clinical and genetic studies of Birt-Hogg-Dube syndrome. J Med Genet 39:906–912 Kahnoski K, Khoo SK, Nassif NT, Chen J, Lobo GP, Segelov E, Teh BT, 2003, Alterations of the Birt-Hogg-Dube gene, BHD, in sporadic colorectal tumours. J Med Genet 40:511–515 Shin JH, Shin YK, Ku JL, Jeong SY, Hong SH, Park SY, Kim WH, Park JG, 2003, Mutations of the Birt-Hogg-Dube, BHD, gene in sporadic colorectal carcinomas and colorectal carcinoma cell lines with microsatellite instability. J Med Genet 40:364–367 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Choi EJ, Kim MS, Song SY, Yoo NJ, Lee SH, 2017, Intratumoral Heterogeneity of Frameshift Mutations in MECOM Gene is Frequent in Colorectal Cancers with High Microsatellite Instability. Pathol Oncol Res 23:145–149 McGranahan N, Swanton C, 2015, Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 27: 15–26 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2811 EP 2812 DI 10.1007/s12253-019-00774-2 PG 2 ER PT J AU Son, JH Choi, JE Yoo, JN Lee, HS AF Son, Ji Hyun Choi, Ji Eun Yoo, Jin Nam Lee, Hyung Sug TI Somatic Mutations and Intratumoral Heterogeneity of Cancer-Related Genes NLK, YY1 and PA2G4 in Gastric and Colorectal Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE NLK; YY1; PG2G4; Mutation; Cancer; Microsatellite instability ID NLK; YY1; PG2G4; Mutation; Cancer; Microsatellite instability AB Many genes act as both tumor suppressor gene (TSG) and proto-oncogene depending on cellular context and cancer type. Nemo-like kinase (NLK) encoding a serine/threonine kinase, Yin Yang 1 (YY1) encoding a zinc-finger transcription factor and PA2G4 encoding an ErbB3 binding protein have both of these two opposing functions. In the present study, we analyzed NLK, YY1 and PA2G4 frameshift mutations in sporadic GC and CRC with high microsatellite instability (MSI-H). Also, regional intratumoral heterogeneity (ITH) of frameshift mutations of these genes was analyzed in CRCs. We found frameshift mutations of NLK, YY1 and PA2G4 in CRC and GC with MSI-H (17/132: 12.9%), but not in those with MSS (0/90). Two (12.5%), one (6.3%) and one (6.3%) CRC (s) of the 16 CRCs exhibited ITH of NLK, YY1 and PA2G4 mutations among the 4–7 regions, suggesting that ITH of the frameshift mutations might be frequent in the CRCs. These results suggest that frameshift mutations of NLK, YY1 and PA2G4 along with the ITH might contribute to MSI-H cancer pathogenesis. C1 [Son, Ji Hyun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Choi, Ji Eun] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of Pathology, 505 Banpo-dong, Socho-gu, 137-701 Seoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, 137-701 Seoul, South Korea. EM suhulee@catholic.ac.kr CR Shen L, Shi Q, Wang W, 2018, Double agents: genes with both oncogenic and tumor-suppressor functions. Oncogenesis 7:25 Eskelinen EL, 2011, The dual role of autophagy in cancer. Curr Opin Pharmacol 11:294–300 Li SZ, Zeng F, Li J, Shu QP, Zhang HH, Xu J, Ren JW, Zhang XD, SongXM, Du RL, 2018, Nemo-like kinase, NLK, primes colorectal cancer progression by releasing the E2F1 complex from HDAC1. Cancer Lett 431:43–53 Hays E, Bonavida B, 2019, YY1 regulates cancer cell immune resistance by modulating PD-L1 expression. Drug Resist Updat 43: 10–28 Nguyen DQ, Hoang DH, Nguyen Vo TT, Huynh V, Ghoda L, Marcucci G, Nguyen LXT, 2018, The role of ErbB3 binding protein 1 in cancer: friend or foe? J Cell Physiol 233:9110–9120 Imai K, Yamamoto H, 2008, Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29:673–680 Choi EJ, Kim MS, Song SY, Yoo NJ, Lee SH, 2017, Intratumoral heterogeneity of Frameshift mutations in MECOM gene is frequent in colorectal cancers with high microsatellite instability. Pathol Oncol Res 23:145–149 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2813 EP 2815 DI 10.1007/s12253-019-00785-z PG 3 ER PT J AU Harsha, SB Manikandan, R Sreerag, S Dorairajan, NL Rajesh, GN Kar, R AF Harsha, Sri Bokka Manikandan, Ramanitharan Sreerag, K. Sreenivasan Dorairajan, Narayanan Lalgudi Rajesh, Ganesh Nachiappa Kar, Rakhi TI Eosinophilic Leukemoid Reaction with Eosinophilic Tumor Tissue Infiltration as an Extermely Poor Prognostic Factor in Urinary Bladder Cancer- a Known Entity Revisited SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE Bladder cancer; Leukemoid reaction; Eosinophilia; Hematuria ID Bladder cancer; Leukemoid reaction; Eosinophilia; Hematuria AB Eosinophilia can be a manifestation of a variety of causes such as infections, allergic reactions and autoimmune processes. Also, it is described in various solid malignancies in the presence of tumour eosinophilic infiltration. We report a patient of high-grade urinary bladder cancer with eosinophilic leukemoid reaction and tumour histopathology demonstrated diffuse infiltration of eosinophils. Though the entity is described to carry a good prognosis in literature, our experience is totally different as the patient deteriorated rapidly in a matter of days, was deemed inoperable in view of worsening performance status and was referred for palliative management. C1 [Harsha, Sri Bokka] JIPMER, Department of Urology and Renal TransplantationPuducherry, India. [Manikandan, Ramanitharan] JIPMER, Department of Urology and Renal TransplantationPuducherry, India. [Sreerag, K. Sreenivasan] JIPMER, Department of Urology and Renal TransplantationPuducherry, India. [Dorairajan, Narayanan Lalgudi] JIPMER, Department of Urology and Renal TransplantationPuducherry, India. [Rajesh, Ganesh Nachiappa] JIPMER, Department of PathologyPuducherry, India. [Kar, Rakhi] JIPMER, Department of PathologyPuducherry, India. RP Harsha, SB (reprint author), JIPMER, Department of Urology and Renal Transplantation, Puducherry, India. EM bokkaharsha7@gmail.com CR Lowe D, Jorizzo J, Hutt MS, 1981 Dec, Tumour-associated eosinophilia: a review. J Clin Pathol 34(12):1343–1348 Flamm J, 1992 Aug, Tumor-associated tissue inflammatory reaction and eosinophilia in primary superficial bladder cancer. Urology. 40(2):180–185 Lowe D, Fletcher CD, Gower RL, 1984 May, Tumour-associated eosinophilia in the bladder. J Clin Pathol 37(5):500–502 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2817 EP 2819 DI 10.1007/s12253-020-00807-1 PG 3 ER PT J AU Bareeqa, BS Ahmed, IS Samar, SS Humayun, HS AF Bareeqa, Beenish Syeda Ahmed, Ijlal Syed Samar, Sana Syeda Humayun, Hasham Syed TI A Letter to Editor Regarding the Article “Identification of Differentially Expressed Proteins from Smokeless Tobacco Addicted Patients Suffering from Oral Squamous Cell Carcinoma” SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Bareeqa, Beenish Syeda] Jinnah Medical and Dental College, 22-23 Shaheed-e-Millat RoadKarachi, Pakistan. [Ahmed, Ijlal Syed] Liaquat National Medical College and HospitalKarachi, Pakistan. [Samar, Sana Syeda] Jinnah Sindh Medical UniversityKarachi, Pakistan. [Humayun, Hasham Syed] Jinnah Medical and Dental CollegeKarachi, Pakistan. RP Bareeqa, BS (reprint author), Jinnah Medical and Dental College, Karachi, Pakistan. EM syedabeenishbareeqa@gmail.com CR Malik, U. U., Siddiqui, I. A., Ilyas, A., Hashim, Z., Staunton, L., Kwasnik, A., . . . Zarina, S., 2019). Identification of differentially expressed proteins from smokeless tobacco addicted patients suffering from Oral squamous cell carcinoma. Pathol Oncol Res, 1–9 Rivera C, 2015, Essentials of oral cancer. Int J Clin Exp Pathol 8(9): 11884–11894 Ebbert JO, Patten CA, Schroeder DR, 2006, The Fagerstrom test for nicotine dependence-smokeless tobacco, FTND-ST). Addict Behav 31(9):1716–1721 Mushtaq N, Huque R, Beebe LA, Shah S, Siddiqi K, 2019, Evaluation of tobacco dependence measures in south Asian smokeless tobacco users. Drug Alcohol Depend 203:66–71 Hoppin JA, Tolbert PE, Taylor JA, Schroeder JC, Holly EA, 2002, Potential for selection bias with tumor tissue retrieval in molecular epidemiology studies. Ann Epidemiol 12(1):1–6 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2821 EP 2822 DI 10.1007/s12253-020-00812-4 PG 2 ER PT J AU Tsiambas, E Papanikolaou, V Chrysovergis, A Mastronikolis, N Ragos, V Kavantzas, N Lazaris, A Kyrodimos, E AF Tsiambas, Evangelos Papanikolaou, Vasileios Chrysovergis, Aristeidis Mastronikolis, Nicholas Ragos, Vasileios Kavantzas, Nikolaos Lazaris, C. Andreas Kyrodimos, Efthymios TI Coronavirus in Hematologic Malignancies: Targeting Molecules Beyond the Angiotensin-Converting Enzyme 2 (ACE2) Wall in COVID-19 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Tsiambas, Evangelos] 401 General Army Hospital, Department of Pathology-CytologyAthens, Greece. [Papanikolaou, Vasileios] National and Kapodistrian, University of Athens, Hippocration Hospital, 1st ENT DepartmentAthens, Greece. [Chrysovergis, Aristeidis] National and Kapodistrian, University of Athens, Hippocration Hospital, 1st ENT DepartmentAthens, Greece. [Mastronikolis, Nicholas] University of Patras, Medical School, Department of Otorhinolaryngology, Head and Neck SurgeryPatras, Greece. [Ragos, Vasileios] University of Ioannina, School of Health Sciences, Department of Maxillofacial and Department of MedicineIoannina, Greece. [Kavantzas, Nikolaos] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, First Department of Pathology, 17 Patriarchou Grigoriou E Street, Ag. Paraskevi, 153 41 Athens, Greece. [Lazaris, C. Andreas] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, First Department of Pathology, 17 Patriarchou Grigoriou E Street, Ag. Paraskevi, 153 41 Athens, Greece. [Kyrodimos, Efthymios] National and Kapodistrian, University of Athens, Hippocration Hospital, 1st ENT DepartmentAthens, Greece. RP Tsiambas, E (reprint author), 401 General Army Hospital, Department of Pathology-Cytology, Athens, Greece. EM tsiambasecyto@yahoo.gr CR Fehr AR, Perlman S, 2015, Coronaviruses: an overview of their replication and pathogenesis. Methods Mol Biol 1282:1–23 Wu A, Peng Y, Huang B et al, 2020, Genome Composition and Divergence of the Novel Coronavirus, 2019-nCoV, Originating in China. Cell Host Microb 27(3):325–328 Li F, 2016, Structure, function, and evolution of coronavirus spike proteins. Ann Rev Virol 3:237–261 Song Z, Xu Y, Bao L et al, 2019, From SARS to MERS, thrusting coronaviruses into the spotlight. Viruses 11:E59–E63 Ge XY, Li JL, Yang XL, Chmura AA, Zhu G, Epstein JH, Mazet JK, Hu B, ZhangW, Peng C, Zhang YJ, Luo CM, Tan B,Wang N, Zhu Y, Crameri G, Zhang SY, Wang LF, Daszak P, Shi ZL, 2013, Isolation and characterization of a bat SARS-like coronavirus that uses the ACE2 receptor. Nature 503:535–538 Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D, 2020, Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 180:1–12 Coutard B, Valle C, de Lamballerie X, Canard B, Seidah NG, Decroly E, 2020, The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoVof the same clade. Antiviral Res 176:104742–247 Hallenberger S, Bosch V, Angliker H, Shaw E, Klenk HD, Garten W(1992, Inhibition of furin-mediated cleavage activation of HIV-1 glycoprotein gp 160. Nature 360(6402):358–361 Ogimi C, Waghmare AA, Kuypers JM et al, 2017, Clinical Significance of Human Coronavirus in Bronchoalveolar Lavage Samples From Hematopoietic Cell Transplant Recipients and Patients With Hematologic Malignancies 64(11):1532–1539 Hakki M, Rattray RM, Press RD, 2015, The clinical impact of coronavirus infection in patients with hematologic malignancies and hematopoietic stem cell transplant recipients. J Clin Virol 68:1–5 Joshi S, Wollenzien H, Leclerc E, Jarajapu YP, 2019, Hypoxic regulation of angiotensin- converting enzyme 2 and Mas receptor in human CD34+ cells. J Cell Physiol 234(11):20420– 20431 Hu W, Yen YT, Singh S, Kao CL, Wu-Hsieh BA, 2012, SARSCoV regulates immune function-related gene expression in human monocytic cells. Viral Immunol 25(4):277–288 Fu J, Zhang J, Gong Y, Testa CL, Klein-Szanto AJ, 2015, Regulation of HIF-1 alpha by the proprotein convertases furin and PC7 in human squamous carcinoma cells. Mol Carcino 54(9):698–706 Braun E, Hotter D, Koepke L et al, 2019, Guanylate-Binding Proteins 2 and 5 Exert Broad Antiviral Activity by Inhibiting Furin-Mediated Processing of Viral Envelope Proteins. Cell Rep 27(7):2092–2104 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2823 EP 2825 DI 10.1007/s12253-020-00810-6 PG 3 ER PT J AU Yoo, HK Choi, T Lee, HL Song, JM Chung, IB AF Yoo, Han Koo Choi, Taesoo Lee, Hyung-Lae Song, Jeong Min Chung, I Benjamin TI Aggressive Paraganglioma of the Urinary Bladder with Local Recurrence and Pelvic Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE Paraganglioma; Urinary bladder; Neoplasm metastasis ID Paraganglioma; Urinary bladder; Neoplasm metastasis AB Many pheochromocytoma and extra-adrenal paraganglioma are benign, but some are malignant. Pheochromocytoma of the Adrenal gland Scaled Score analyzed the histological characteristics of the tumor. Tumors with a Pheochromocytoma of the Adrenal gland Scaled Score of 4 or higher have a higher risk of recurrence. This pattern is thought to be applicable to paraganglioma as well, and to future patient follow-up efforts. We report a recurrent and metastatic paraganglioma of the urinary bladder. C1 [Yoo, Han Koo] Stanford University Medical Center, Department of UrologyStanford, CA, USA. [Choi, Taesoo] Kyung Hee University, School of Medicine, Department of Urology, 892, Dongnam-ro, Gangdong-gu, 05278 Seoul, South Korea. [Lee, Hyung-Lae] Kyung Hee University, School of Medicine, Department of Urology, 892, Dongnam-ro, Gangdong-gu, 05278 Seoul, South Korea. [Song, Jeong Min] Kyung Hee University, College of Medicine, Department of Pathology, 892, Dongnam-ro, Gangdong-gu, 05278 Seoul, South Korea. [Chung, I Benjamin] Stanford University Medical Center, Department of UrologyStanford, CA, USA. RP Yoo, HK (reprint author), Stanford University Medical Center, Department of Urology, Stanford, USA. EM yookoohan@khu.ac.kr CR Pacak K, Wimalawansa SJ, 2015, Pheochromocytoma and paraganglioma. Endocr Pract: Off J Am Coll Endocrinol Am Assoc Clin Endocrinologists 21(4):406–412. , DOI 10. 4158/EP14481.RA Lenders JW, Pacak K, Walther MM, Linehan WM, Mannelli M, Friberg P, Keiser HR, Goldstein DS, Eisenhofer G, 2002, Biochemical diagnosis of pheochromocytoma: which test is best? Jama 287(11):1427–1434. , DOI 10.1001/jama.287.11. 1427 Walz MK, Alesina PF, Wenger FA, Deligiannis A, Szuczik E, Petersenn S, Ommer A, Groeben H, Peitgen K, Janssen OE, Philipp T, Neumann HP, Schmid KW, Mann K, 2006, Posterior retroperitoneoscopic adrenalectomy–results of 560 procedures in 520 patients. Surgery 140(6):943–948; discussion 948-950. https:// doi.org/10.1016/j.surg.2006.07.039 Gonias S, Goldsby R, Matthay KK, Hawkins R, Price D, Huberty J, Damon L, Linker C, Sznewajs A, Shiboski S, Fitzgerald P, 2009, Phase II study of high-dose [131I]metaiodobenzylguanidine therapy for patients with metastatic pheochromocytoma and paraganglioma. J Clin Oncol: Off J Am Soc Clin Oncol 27(25):4162–4168. https:// doi.org/10.1200/JCO.2008.21.3496 Thompson LD, 2002, Pheochromocytoma of the adrenal gland scaled score, PASS, to separate benign from malignant neoplasms: a clinicopathologic and immunophenotypic study of 100 cases. AmJ Surg Pathol 26(5):551–566. , DOI 10.1097/00000478- 200205000-00002 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2827 EP 2829 DI 10.1007/s12253-020-00841-z PG 3 ER PT J AU Hatzl, S Posch, F Schulz, E Gornicec, M Deutsch, A Beham-Schmid, Ch Pichler, M Greinix, H Sill, H Zebisch, A Neumeister, P Prochazka, K AF Hatzl, Stefan Posch, Florian Schulz, Eduard Gornicec, Maximilian Deutsch, Alexander Beham-Schmid, Christine Pichler, Martin Greinix, Hildegard Sill, Heinz Zebisch, Armin Neumeister, Peter Prochazka, T. Katharina TI The Role of Immunohistochemical Overexpression of p53 as Adverse Prognostic Factor in Primary Testicular Diffuse Large B Cell Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Hatzl, Stefan] Medical University of Graz, Department of Internal Medicine, Division of Hematology, Auenbruggerplatz 38D, A-8036 Graz, Austria. [Posch, Florian] Medical University of Graz, Department of Internal Medicine, Division of OncologyGraz, Austria. [Schulz, Eduard] Medical University of Graz, Department of Internal Medicine, Division of Hematology, Auenbruggerplatz 38D, A-8036 Graz, Austria. [Gornicec, Maximilian] Medical University of Graz, Department of Internal Medicine, Division of Hematology, Auenbruggerplatz 38D, A-8036 Graz, Austria. [Deutsch, Alexander] Medical University of Graz, Department of Internal Medicine, Division of Hematology, Auenbruggerplatz 38D, A-8036 Graz, Austria. [Beham-Schmid, Christine] Medical University of Graz, Department of PathologyGraz, Austria. [Pichler, Martin] Medical University of Graz, Department of Internal Medicine, Division of OncologyGraz, Austria. [Greinix, Hildegard] Medical University of Graz, Department of Internal Medicine, Division of Hematology, Auenbruggerplatz 38D, A-8036 Graz, Austria. [Sill, Heinz] Medical University of Graz, Department of Internal Medicine, Division of Hematology, Auenbruggerplatz 38D, A-8036 Graz, Austria. [Zebisch, Armin] Medical University of Graz, Department of Internal Medicine, Division of Hematology, Auenbruggerplatz 38D, A-8036 Graz, Austria. [Neumeister, Peter] Medical University of Graz, Department of Internal Medicine, Division of Hematology, Auenbruggerplatz 38D, A-8036 Graz, Austria. [Prochazka, T. Katharina] Medical University of Graz, Department of Internal Medicine, Division of Hematology, Auenbruggerplatz 38D, A-8036 Graz, Austria. RP Hatzl, S (reprint author), Medical University of Graz, Department of Internal Medicine, Division of Hematology, A-8036 Graz, Austria. EM stefan.hatzl@medunigraz.at CR Sukswai N, Lyapichev K, Khoury JD, Medeiros LJ, 2020, Diffuse large B-cell lymphoma variants: an update. Pathology. 52:53–67 Magnoli F, Bernasconi B,Vivian L, Proserpio I, Pinotti G, Campiotti L, Mazzucchelli L, Sessa F, Tibiletti MG, Uccella S, 2018, Primary extranodal diffuse large B-cell lymphomas: many sites, many entities? Clinico-pathological, immunohistochemical and cytogenetic study of 106 cases. Cancer Genet 228-229:28–40 Peroja P, Pedersen M, Mantere T, Norgaard P, Peltonen J, Haapasaari KM, Bohm J, Jantunen E, Turpeenniemi-Hujanen T, Rapakko K, Karihtala P, Soini Y, Vasala K, Kuittinen O, 2018, Mutation of TP53, translocation analysis and immunohistochemical expression of MYC, BCL-2 and BCL-6 in patients with DLBCL treated with R-CHOP. Sci. Rep. 8:14814. , DOI 10.1038/ s41598-018-33230-3 Hatzl S, Posch F, Deutsch A, Beham-Schmid C, Stoger H, Greinix H, Pichler M, Neumeister P, Prochazka KT, 2020, Immunohistochemistry for c-myc and bcl-2 overexpression improves risk stratification in primary central nervous system lymphoma. Hematol Oncol Wang XJ, Jeffrey ML, Bueso-Ramos CE, Tang G, Wang S, Oki Y, Desai P, Khoury JD, Miranda RN, Tang Z, Reddy N, Li S, 2017, P53 expression correlates with poorer survival and augments the negative prognostic effect of MYC rearrangement, expression or concurrent MYC/BCL2 expression in diffuse large B-cell lymphoma. Mod Pathol 30:194–203 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2831 EP 2833 DI 10.1007/s12253-020-00864-6 PG 3 ER PT J AU Zhou, Sh Zhang, D Li, J Zhao, J Wang, G Zhang, Y Bai, Y Chen, D Wu, H AF Zhou, Shaoqiang Zhang, Ding Li, Junjie Zhao, Jing Wang, Guoqiang Zhang, Yuzi Bai, Yuezong Chen, Dedian Wu, Hao TI Landscape of RAS Variations in 17,993 Pan-cancer Patients Identified by Next-generation Sequencing SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE Carcinoma; Genetics; RAS gene; Next-generation sequencing ID Carcinoma; Genetics; RAS gene; Next-generation sequencing AB RAS family genes (HRAS, KRAS and NRAS) were frequently observed in several tumors. The expression of constitutively active RAS proteins mediated by RAS variations promote the development of tumors. KRAS is an important prognostic and drug resistance biomarker. It would also be a promising drug target. Several trials which evaluating the efficacy of RAS G12C inhibitor in solid tumors are initiated. Herein, we analyzed the alterations status of KRAS/NRAS/HRAS across diverse solid tumors. The sing nucleotide variants (SNV) and copy number variants (CNV) data of 17993 Chinese patients from 22 types of cancer were obtained in our database. Genomic profiling of DNA was performed through a next-generation sequencing on tissue. Only the pathogenic mutations and likely pathogenic mutations in clinical significance were rolled into our analysis. Among 17993 pancancer patients, the total RAS variants frequency was 22.58%. KRAS was the most frequently altered, followed by NRAS and HRAS. For the SNV, KRAS were most commonly found in pancreas cancer, intestine cancer and colorectal cancer. Further analysis among KRAS SNV patients showed that the mutation frequency of KRAS G12C, G12D, G12R, and G12V was 1.81%, 6.81%, 0.69% and 4.25%, respectively. A total of 21 in 22 types of solid tumors had KRAS G12C/D/R/V pathogenic or likely pathogenic mutation, which occurred most frequently in colorectal cancer, pancreas cancer and lung cancer. Our results suggested that a variety of solid tumors may harbor KRAS G12C/D/R/V mutation. These patients may benefit from KRAS inhibitors. C1 [Zhou, Shaoqiang] The Third Affiliated Hospital ofKunming Medical University (Yunnan Tumor Hospital), Galactophore Department, No.518 Kunzhou Road, 650118 Kunming, Yunnan Province, China. [Zhang, Ding] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Li, Junjie] The Central Hospital of Zibo, Department of AnesthesiologyZibo, China. [Zhao, Jing] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Wang, Guoqiang] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Zhang, Yuzi] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Bai, Yuezong] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Chen, Dedian] The Third Affiliated Hospital ofKunming Medical University (Yunnan Tumor Hospital), Galactophore Department, No.518 Kunzhou Road, 650118 Kunming, Yunnan Province, China. [Wu, Hao] Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, NO.300 Guangzhou Road, 210029 Nanjing, Jiangsu Province, China. RP Wu, H (reprint author), Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, 210029 Nanjing, China. EM whdactor@163.com CR Vetter IR, Wittinghofer A, 2001, The guanine nucleotide-binding switch in three dimensions. Science 294(5545):1299–1304 Wan XB,Wang AQ, Cao J et al, 2019, Relationships among KRAS mutation status, expression of RAS pathway signaling molecules, and clinicopathological features and prognosis of patients with colorectal cancer. World J Gastroenterol 25(7):808–823 Lartigue JD, 2019, New strategies generate optimism for targeting “Undruggable” KRAS. Oncol Live 20:19 Chaft JE, Litvak A, Arcila ME et al, 2014, Phase II study of the GI- 4000 KRAS vaccine after curative therapy in patients with stage I-III lung adenocarcinoma harboring a KRAS G12C, G12D, or G12V mutation. Clin Lung Cancer 15(6):405–410 Shepherd FA, Domerg C, Hainaut P et al, 2013, Pooled analysis of the prognostic and predictive effects of KRAS mutation status and KRAS mutation subtype in early-stage resected non-small-cell lung cancer in four trials of adjuvant chemotherapy. J Clin Oncol 31(17): 2173–2181 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2835 EP 2837 DI 10.1007/s12253-020-00845-9 PG 3 ER PT J AU Tsiambas, E Chrysovergis, A Papanikolaou, V Mastronikolis, N Ragos, V Kavantzas, N Lazaris, A Patsouris, E Riziotis, Ch Paschopoulos, M Kyrodimos, E AF Tsiambas, Evangelos Chrysovergis, Aristeidis Papanikolaou, Vasileios Mastronikolis, Nicholas Ragos, Vasileios Kavantzas, Nikolaos Lazaris, C. Andreas Patsouris, Efstratios Riziotis, Christos Paschopoulos, Minas Kyrodimos, Efthymios TI Chromosome X riddle in SARS-CoV-2 (COVID-19) - related lung pathology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter C1 [Tsiambas, Evangelos] VA Hospital (NIMTS), Department of CytologyAthens, Greece. [Chrysovergis, Aristeidis] National and Kapodistrian, University of Athens, Hippocration Hospital, 1st ENT DepartmentAthens, Greece. [Papanikolaou, Vasileios] National and Kapodistrian, University of Athens, Hippocration Hospital, 1st ENT DepartmentAthens, Greece. [Mastronikolis, Nicholas] University of Patras, Medical School, Department of Otorhinolaryngology, Head and Neck SurgeryPatras, Greece. [Ragos, Vasileios] University of Ioannina, School of Health Sciences, Department of Maxillofacial and Department of MedicineIoannina, Greece. [Kavantzas, Nikolaos] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, First Department of PathologyAthens, Greece. [Lazaris, C. Andreas] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, First Department of PathologyAthens, Greece. [Patsouris, Efstratios] National and Kapodistrian University of Athens, Medical School, Laiko General Hospital, First Department of PathologyAthens, Greece. [Riziotis, Christos] National Hellenic Research Foundation, Theoretical and Physical Chemistry Institute, Photonics for Nanoapplications LaboratoryAthens, Greece. [Paschopoulos, Minas] Ioannina University Hospital, Department of Obstetrics and GynecologyIoannina, Greece. [Kyrodimos, Efthymios] National and Kapodistrian, University of Athens, Hippocration Hospital, 1st ENT DepartmentAthens, Greece. RP Tsiambas, E (reprint author), VA Hospital (NIMTS), Department of Cytology, Athens, Greece. EM tsiambasecyto@yahoo.gr CR Wu A, Peng Y, Huang B et al, 2020, Genome Composition and Divergence of the Novel Coronavirus, 2019-nCoV, Originating in China. Cell Host Microb 27(3):325–328 Lukassen S, Chua RL, Trefzer T et al, 2020, SARS-CoV-2 Receptor ACE2 and TMPRSS2 Are Primarily Expressed in Bronchial Transient Secretory Cells. EMBO J e105114–118 Coutard B, Valle C, de Lamballerie X, Canard B, Seidah NG, Decroly E, 2020, The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antiviral Res 176:104742–104747 Tsiambas E, Papanikolaou V, Chrysovergis A et al, 2020, Coronavirus in hematologic malignancies: targeting molecules beyond the angiotensin-converting enzyme 2, ACE2, wall. Pathol Oncol Res 1–3 Buja LM,Wolf DA, Zhao B et al, 2020, The emerging spectrum of cardiopulmonary pathology of the coronavirus disease 2019, COVID-19): Report of 3 autopsies from Houston, Texas, and review of autopsy findings from other United States cities. Cardiovasc Pathol 48:107233–107243 Lewnard JA, Liu VX, Jackson ML et al, 2020, Incidence, clinical outcomes, and transmission dynamics of severe coronavirus disease 2019 in California andWashington: prospective cohort study. BMJ 369:m1923–1933 Yamada S, Utsunomiya M, Inoue K et al, 2004, Genome-wide Scan for Japanese Familial Intracranial Aneurysms: Linkage to Several Chromosomal Regions. Circulation 110:3727–3733 Zhong J, Yan Z, Liu D et al, 2006, Association of Angiotensin- Converting Enzyme 2 Gene A/G Polymorphism and Elevated Blood Pressure in Chinese Patients With Metabolic Syndrome. J Lab Clin Med 147(2):91–95 Conti P, Younes A, 2020, Coronavirus COV-19/SARS-CoV-2 Affects Women Less Than Men: Clinical Response to Viral Infection. J Biol Regul Homeost Agents 34(2) Li G, He X, Zhang L et al, 2020, Assessing ACE2 Expression Patterns in Lung Tissues in the Pathogenesis of COVID-19. J Autoimmun:102463–49 Sawalha AH, Zhao M, Coit P, Lu Q, 2020, Epigenetic Dysregulation of ACE2 and Interferon-Regulated Genes Might Suggest Increased COVID-19 Susceptibility and Severity in Lupus Patients. Clin Immunol 215:108410–108415 Yamaguchi M, Osuka S, Shoji M, Weitzmann MN, Murata T, 2017, Survival of lung cancer patients is prolonged with higher regucalcin gene expression: suppressed proliferation of lung adenocarcinoma A549 cells in vitro. Mol Cell Biochem 430(1–2):37– 46 Wei X, Yu H, Zhao P, Xie L, Li L, Zhang J, 2019, Serum regucalcin is a useful indicator of liver injury severity in patients with hepatitis B virus-related liver diseases. Braz J Med Biol Res 52(10):e8845–e8850 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2020 VL 26 IS 4 BP 2839 EP 2841 DI 10.1007/s12253-020-00878-0 PG 3 ER PT J AU Shen, H Liao, K Wu, W Li, G Chen, Sh Nan, N Yu, H Wu, H AF Shen, Hua Liao, Kai Wu, Weili Li, Gongyu Chen, Shijin Nan, Nan Yu, Hongbo Wu, Hongfei TI Case Report: Mucinous Adenocarcinoma Arising From Congenital Ejaculatory Duct Cyst SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE mucinous adenocarcinoma; ejaculatory duct; ejaculatory duct cyst; ejaculatory duct tumor; ejaculatory duct adenocarcinoma ID mucinous adenocarcinoma; ejaculatory duct; ejaculatory duct cyst; ejaculatory duct tumor; ejaculatory duct adenocarcinoma AB Herein we present a previously unreported rare case of mucinous adenocarcinoma arising from a congenital ejaculatory duct cyst. Radiographic and endoscopic examinations revealed the tumor occurred in a cyst running through the prostate. Initially, the immunohistochemical pathology results showed that it was a metastatic mucinous adenocarcinoma, but no other primary lesions were clinically evidenced. Based on the embryonic development process of the male urogenital tract, the malformation of the patient’s ejaculatory duct, and the pathological examination of the resected specimen, we considered the tumor to be a primary mucinous adenocarcinoma which originating from the hypoplastic ejaculatory duct. The tumor may have developed from the foci of intestinal metaplasia from cloacal remnants during embryonic development. C1 [Shen, Hua] The Affiliated BenQ Hospital of Nanjing Medical University, BenQ Medical Center, Department of UrologyNanjing, China. [Liao, Kai] The Affiliated BenQ Hospital of Nanjing Medical University, BenQ Medical Center, Department of UrologyNanjing, China. [Wu, Weili] The Affiliated BenQ Hospital of Nanjing Medical University, BenQ Medical Center, Department of UrologyNanjing, China. [Li, Gongyu] The Affiliated BenQ Hospital of Nanjing Medical University, BenQ Medical Center, Department of UrologyNanjing, China. [Chen, Shijin] The Affiliated BenQ Hospital of Nanjing Medical University, BenQ Medical Center, Department of PathologyNanjing, China. [Nan, Nan] The Affiliated BenQ Hospital of Nanjing Medical University, BenQ Medical Center, Department of RadiologyNanjing, China. [Yu, Hongbo] The Affiliated BenQ Hospital of Nanjing Medical University, BenQ Medical Center, Department of UrologyNanjing, China. [Wu, Hongfei] The Affiliated BenQ Hospital of Nanjing Medical University, BenQ Medical Center, Department of UrologyNanjing, China. RP Shen, H (reprint author), The Affiliated BenQ Hospital of Nanjing Medical University, BenQ Medical Center, Department of Urology, Nanjing, China. EM shnjmu@hotmail.com CR Bohman, K. D., and Osunkoya, A. O., 2012). Mucin-producing tumors and tumor-like lesions involving the prostate: a comprehensive review. Adv. Anat. Pathol. 19, 6), 374–387., DOI 10.1097/PAP.0b013e318271a361 Fan, K., and Johnson, D. F., 1985). Adenomatoid tumor of ejaculatory duct. Urology 25, 653–654., DOI 10.1016/0090-4295(85)90307-3 Hamilton, S. R., and Aaltonen, L. A., 2000). World health organization classification of tumours, Lyon, France: IARC Press). Kang, H., O’Connell, J. B., Maggard, M. A., Sack, J., and Ko, C. Y., 2005). A 10- year outcomes evaluation of mucinous and signet-ring cell carcinoma of the colon and rectum. Dis. Colon Rectum 48, 1161., DOI 10.1007/s10350-004-0932-1 Kruepunga, N., Hikspoors, J. P. J. M.,Mekonen, H. K., Mommen, G.M. C., Meemon, K., Weerachatyanukul, W., et al., 2018). The development of the cloaca in the human embryo. J. Anat. 233, 6), 724–739., DOI 10.1111/joa. 12882 Morson, B. C., and Sobin, L. H., 1976). “Histological typing of intestinal tumours,” in International histological classification of tumours. No. 15. Geneva: World Health Organization, 69. Ormsby, A. H., Haskell, R., Jones, D., and Goldblum, J. R., 2000). Primary seminal vesicle carcinoma: an immunohistochemical analysis of four cases. Mod. Pathol. 13, 1), 46–51., DOI 10.1038/modpathol.3880008 Osunkoya, A. O., Nielsen, M. E., and Epstein, J. I., 2008). Prognosis of mucinous adenocarcinoma of the prostate treated by radical prostatectomy: a study of 47 cases. Am. J. Surg. Pathol. 32, 3), 468–472., DOI 10.1097/PAS. 0b013e3181589f72 Sanchez-Salazar, A. J., Basler, J. W., and Nicolas, M. M., 2010). Intraductal carcinoma of the prostate in the ejaculatory duct. Int. J. Surg. Pathol. 18, 298–299., DOI 10.1177/1066896910364534 Seibel, J. L., Prasad, S., Weiss, R. E., Bancila, E., and Epstein, J. I., 2002). Villous adenoma of the urinary tract: a lesion frequently associated with malignancy. Hum. Pathol. 33, 2), 236–241., DOI 10.1053/hupa.2002.31293 Wang, J., and Manucha, V., 2016). Villous adenoma of the urinary bladder: a brief review of the literature. Arch. Pathol. Lab Med. 140, 1), 91–93., DOI 10. 5858/arpa.2014-0198-RS 12 Wang, S., Chen, S. W., Liu, W., Zhu, H. J., Jiang, H., Wang, Y. B., et al., 2007). Laparoscopic excision of extraprostatic ejaculatory duct cyst. Andrologia 39, 81–86., DOI 10.1111/j.1439-0272.2007.00768.x 13. Wein, A. J., 2012). Campbell walsh-urology. 10th Edn. Philadelphia, PA: Saunders Elsevier, 1012–1013. 14. Werner, D., Wilkinson, E. J., Ripley, D.and Yachnis, A., 2004). Primary adenocarcinoma of the vagina withmucinous-entericdifferentiation: a report of two cases with associated vaginal adenosis without history of diethylstilbestrol exposure. J. Low. Genit. Tract Dis. 8, 1), 38–42., DOI 10. 1097/00128360-200401000-00009 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 528050 EP 528054 DI 10.3389/pore.2021.528050 PG 5 ER PT J AU Bi, Y Zhang, J Zeng, D Chen, L Ye, W Yang, Q Ling, Y AF Bi, Yanzhi Zhang, Junling Zeng, Dongxiang Chen, Lili Ye, Wei Yang, Quanliang Ling, Yang TI Cholinesterase is Associated With Prognosis and Response to Chemotherapy in Advanced Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cholinesterase; prognosis; gastric cancer; chemotherapy; progression-free survival; overall survival ID cholinesterase; prognosis; gastric cancer; chemotherapy; progression-free survival; overall survival AB Background: Cholinesterase (CHE) is a routine serum biomarker in gastric cancer (GC). However, little research has been done on its clinical value in advanced GC. In addition, it is not clear whether it can be used as biomarker for the response and prognosis of advanced GC patients. Methods: Between Jan. 2013 and Dec. 2016, a total of 150 patients with advanced GC treated with first-line chemotherapy were admitted to Changzhou Tumor Hospital Affiliated to Soochow University. We retrospectively identified serum CHE level on the day before chemotherapy and at the end of chemotherapy and abstracted clinicopathologic features and treatment outcomes. Univariate and multivariate survival analyses were performed to assess the relationship between serum CHE levels and progression-free survival (PFS) and overall survival (OS). Results: A total of 150 advanced GC patients were included and divided into serum level ≥5,000 IU/L and serum level <5,000 IU/L. CHE level lower than 5,000 IU/L was associated with poorer PFS (HR, 1.60; 95% CI, 1.141–2.243; p=0.006), poorer OS (HR, 1.76; 95% CI, 1.228–2.515; p=0.002) and trend of poorer response (HR, 0.56; 95% CI, 0.272–1.129; p=0.104). In univariate and multivariate logistic regression analysis, only liver metastasis and PS score were significantly associated with objective response (p < 0.05). The medium PFS was 8.0 months in patients with post-treatment CHE increased vs. 3.8 months in patients with CHE decreased after chemotherapy (HR, 1.82; 95% CI 1.28–2.57; p=0.0002). The medium OS was 13.1 months in patients with increased posttreatment CHE vs. 8.1 months in patients with decreased post-treatment CHE (HR, 1.87; 95% CI 1.29–2.71; p=0.0002). Conclusion: Advanced GC with CHE levels below 5,000 IU/L was significantly associated with poor PFS and OS. The results suggested that CHE analysis before chemotherapy was a promising prognostic marker for advanced GC. C1 [Bi, Yanzhi] Changzhou Tumor Hospital Affiliated to Soochow University, Department of OncologyChangzhou, China. [Zhang, Junling] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Zeng, Dongxiang] Changzhou Tumor Hospital Affiliated to Soochow University, Department of OncologyChangzhou, China. [Chen, Lili] The Suqian Affiliated Hospital of Xuzhou Medicine University, Department of HematologySuqian, China. [Ye, Wei] Changzhou Tumor Hospital Affiliated to Soochow University, Department of OncologyChangzhou, China. [Yang, Quanliang] Changzhou Tumor Hospital Affiliated to Soochow University, Department of OncologyChangzhou, China. [Ling, Yang] Changzhou Tumor Hospital Affiliated to Soochow University, Department of OncologyChangzhou, China. RP Ling, Y (reprint author), Changzhou Tumor Hospital Affiliated to Soochow University, Department of Oncology, Changzhou, China. EM medilyn@vip.126.com CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A Global cancer statistics,, 2012). CA Cancer J Clin, 2015, 65:87–108., DOI 10.3322/ caac.21262 Wang F-H, Shen L, Li J, Zhou Z-W, Liang H, Zhang X-T, et al. The Chinese Society of Clinical Oncology, CSCO): clinical guidelines for the diagnosis and treatment of gastric cancer. Cancer Commun, 2019, 39:10., DOI 10.1186/s40880- 019-0349-9 Feng R-M, Zong Y-N, Cao S-M, Xu R-H Current cancer situation in China: good or bad news from the 2018 Global Cancer Statistics?. Cancer Commun, 2019, 39:22., DOI 10.1186/s40880-019-0368-6 KoizumiW, Narahara H, Hara T, Takagane A, Akiya T, Takagi M, et al. S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer, SPIRITS trial): a phase III trial. Lancet Oncol, 2008, 9:215–21., DOI 10.1016/ S1470-2045(08)70035-4 Mohri Y, Tanaka K, Ohi M, Yokoe T, Miki C, Kusunoki M Prognostic significance of host- and tumor-related factors in patients with gastric cancer. World J Surg, 2010, 34:285–90., DOI 10.1007/s00268-009-0302-1 Wang Y, Wang H, Chen H-z. AChE inhibition-based multi-target-directed ligands, a novel pharmacological approach for the symptomatic and diseasemodifying therapy of alzheimer’s disease. Cn, 2016, 14:364–75., DOI 10.2174/ 1570159x14666160119094820 Ford MD Acute poisoning. In: L Goldman D Ausiello, editors. Cecil medicine volume chap 111. 23rd ed. Philadelphia, Pa: Saunders Elsevier, 2007). Battisti V, Bagatini MD, Maders LDK, Chiesa J, Santos KF, Goncalves JF, et al. Cholinesterase activities and biochemical determinations in patients with prostate cancer: influence of Gleason score, treatment and bone metastasis. Biomed Pharmacother, 2012, 66:249–55., DOI 10.1016/j.biopha.2011.11.005 Bradamante V, Smigovec E, Bukovi´c D, Geber J, Matani´c D Plasma cholinesterase activity in patients with uterine cervical cancer during radiotherapy. Coll Antropol, 2000, 24:373–(80.) Mitsunaga S, Kinoshita T, Hasebe T, Nakagohri T, Konishi M, Takahashi S, et al. Low serum level of cholinesterase at recurrence of pancreatic cancer is a poor prognostic factor and relates to systemic disorder and nerve plexus invasion. Pancreas, 2008, 36:241–8., DOI 10.1097/MPA. 0b013e31815b6b2b Maltoni M, Caraceni A, Brunelli C, Broeckaert B, Christakis N, Eychmueller S, et al. Prognostic factors in advanced cancer patients: evidence-based clinical recommendations-A study by the steering committee of the European association for palliative care. Jco, 2005, 23:6240–8., DOI 10.1200/JCO.2005.06.866 Yasui W, Oue N, Aung PP, Matsumura S, Shutoh M, Nakayama H Molecularpathological prognostic factors of gastric cancer: a review. Gastric Cancer, 2005, 8:86–94., DOI 10.1007/s10120-005-0320-0 Yamashita K, Hosoda K, Ema A, WatanabeMLymph node ratio as a novel and simple prognostic factor in advanced gastric cancer. Eur J Surg Oncol, Ejso,, 2016, 42:1253–60., DOI 10.1016/j.ejso.2016.03.001 Zhou Y, Li XB Endoscopic prediction of tumor margin and invasive depth in early gastric cancer. J Dig Dis, 2015, 16:303–10.doi:10.1111/1751-2980.12251 Sagi Y, Weinstock M, Youdim MBH Attenuation of MPTP-induced dopaminergic neurotoxicity by TV3326, a cholinesterase-monoamine oxidase inhibitor. J Neurochem, 2003, 86:290–7., DOI 10.1046/j.1471-4159. 2003.01801.x Gu S-Z, Zhao XH, Quan P, Li SB, Pan BR Alterations of serum cholinesterase in patients with gastric cancer. Wjg, 2005, 11:4604–6., DOI 10.3748/wjg.v11.i29.4604 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 580800 EP 580807 DI 10.3389/pore.2021.580800 PG 8 ER PT J AU Kiss, T Jambor, K Koroknai, V Szasz, I Bardos, H Mokanszki, A Adany, R Balazs, M AF Kiss, Timea Jambor, Krisztina Koroknai, Viktoria Szasz, Istvan Bardos, Helga Mokanszki, Attila Adany, Roza Balazs, Margit TI Silencing Osteopontin Expression Inhibits Proliferation, Invasion and Induce Altered Protein Expression in Melanoma Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE osteopontin expression; osteopontin knockdown; protein expression profile; melanoma progression; invasion; proliferation ID osteopontin expression; osteopontin knockdown; protein expression profile; melanoma progression; invasion; proliferation AB Osteopontin (OPN) is a multifunctional phosphoprotein that is expressed in different types of cancers, including melanoma. OPN overexpression is associated with tumor progression and metastasis formation; however, the role of OPN in cell invasion and metastasis formation is not completely understood. In this study we aimed to define OPN expression in melanoma tissues and cell lines and investigate the effect of OPN expression on cell proliferation and invasion after inhibiting OPN expression with small interfering RNA (siRNA). OPN gene expression was determined by qRT-PCR, while protein expression was examined using a Proteome Profiler Oncology Array. siRNA-mediated OPN knockdown led to decreased OPN expression in melanoma cell lines, which was associated with decreased cell proliferation and invasion. Proteome profile analysis revealed significantly different protein expression between the original and transfected cell lines. The altered expression of the differently expressed proteins was validated at the mRNA level. Furthermore, OPN-specific siRNA was able to reduce OPN expression and inhibit the invasiveness of melanoma cells. Our results revealed for the first time that silencing the OPN gene influences proliferation and invasion of melanoma cells by effecting EGFR, tenascin C, survivin, galectin-3 and enolase 2 expression. To predict proteinprotein interactions along with putative pathways we used STRING analysis for the differentially expressed proteins. These proteins formed multiple clusters, including extracellular matrix organization, regulation of angiogenesis, cell death and cell migration, PI3K-Akt, MAPK and focal adhesion signaling pathways. Taken together these data suggest that OPN might be an ideal target for drug development and therapies. C1 [Kiss, Timea] Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai TanszekDebrecen, Hungary. [Jambor, Krisztina] Debreceni Egyetem, Egeszsegtudomanyok Doktori IskolaDebrecen, Hungary. [Koroknai, Viktoria] University of Debrecen, MTA-DE Public Health Research GroupDebrecen, Hungary. [Szasz, Istvan] University of Debrecen, MTA-DE Public Health Research GroupDebrecen, Hungary. [Bardos, Helga] Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai TanszekDebrecen, Hungary. [Mokanszki, Attila] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Adany, Roza] Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai TanszekDebrecen, Hungary. [Balazs, Margit] Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai TanszekDebrecen, Hungary. RP Balazs, M (reprint author), Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai Tanszek, Debrecen, Hungary. EM balazs.margit@med.unideb.hu CR Schadendorf, D., van Akkooi, A. C. J., Berking, C., Griewank, K. G., Gutzmer, R., Hauschild, A., et al., 2018). Melanoma. Lancet. 392, 10151), 971–984., DOI 10. 1016/S0140-6736(18)31559-9 Turner,N., Ware,O., and Bosenberg,M., 2018).Genetics ofmetastasis: melanoma and other cancers.Clin. Exp. Metastasis. 35, 5-6), 379–391., DOI 10.1007/s10585-018-9893-y Ferguson, P.M., Long, G. V., Scolyer, R. A., and Thompson, J. F., 2018). Impact of genomics on the surgical management of melanoma. Br. J. Surg. 105, 2), e31–e47., DOI 10.1002/bjs.10751 Malissen, N., and Grob, J. J., 2018). Metastatic melanoma: recent therapeutic progress and future perspectives. Drugs. 78, 12), 1197–1209., DOI 10.1007/ s40265-018-0945-z Guarneri,C.,Bevelacqua, V., Polesel, J., Falzone, L.,Cannavo, P. S., Spandidos,D.A., et al., 2017).NFkappaB inhibition is associated withOPN/MMP9 downregulation in cutaneous melanoma. Oncol. Rep. 37, 2), 737–746., DOI 10.3892/or.2017.5362 Wei, R., Wong, J. P. C., and Kwok, H. F., 2017). Osteopontin–a promising biomarker for cancer therapy. J. Canc. 8, 12), 2173–2183., DOI 10.7150/jca.20480 Icer, M. A., and Gezmen-Karadag,M., 2018). The multiple functions and mechanisms of osteopontin. Clin. Biochem. 59, 17–24., DOI 10.1016/j.clinbiochem.2018.07.003 Zhao, H., Chen, Q., Alam, A., Cui, J., Suen, K. C., Soo, A. P., et al., 2018). The role of osteopontin in the progression of solid organ tumour. Cell Death Dis. 9, 3), 356., DOI 10.1038/s41419-018-0391-6 Wai, P. Y., and Kuo, P. C., 2008). Osteopontin: regulation in tumor metastasis. Canc. Metastasis Rev. 27, 1), 103–118., DOI 10.1007/s10555-007-9104-9 Weber, G. F., Lett, G. S., and Haubein, N. C., 2010). Osteopontin is a marker for cancer aggressiveness and patient survival. Br. J. Canc. 103, 6), 861–869., DOI 10.1038/sj.bjc.6605834 Rakosy, Z., Ecsedi, S., Toth, R., Vizkeleti, L., Hernandez-Vargas, H., Lazar, V., et al., 2013). Integrative genomics identifies gene signature associated with melanoma ulceration. PloS One. 8, 1), e54958., DOI 10.1371/journal.pone.0054958 Yin, M., Soikkeli, J., Jahkola, T., Virolainen, S., Saksela, O., and Holtta, E., 2014). Osteopontin promotes the invasive growth of melanoma cells by activating integrin alphavbeta3 and down-regulating tetraspanin CD9. Am. J. Pathol. 184, 3), 842–858., DOI 10.1016/j.ajpath.2013.11.020 Damsky, W. E., Rosenbaum, L. E., and Bosenberg, M., 2010). Decoding melanoma metastasis. Cancers. 3, 1), 126–163., DOI 10.3390/cancers3010126 Valastyan, S., andWeinberg, R. A., 2011). Tumor metastasis: molecular insights and evolving paradigms. Cell. 147, 2), 275–292., DOI 10.1016/j.cell.2011.09.024 Lambert, A. W., Pattabiraman,D. R., and Weinberg, R.A., 2017). Emerging biological principles of metastasis. Cell. 168, 4), 670–691., DOI 10.1016/j.cell.2016.11.037 Ahmed, M., Sottnik, J. L., Dancik, G. M., Sahu, D., Hansel, D. E., Theodorescu, D., et al., 2016). An osteopontin/CD44 Axis in RhoGDI2-mediated metastasis suppression. Canc. Cell. 30, 3), 432–443., DOI 10.1016/j.ccell.2016.08.002 Shirasaki, T., Honda, M., Yamashita, T., Nio, K., Shimakami, T., Shimizu, R., et al., 2018). The osteopontin-CD44 axis in hepatic cancer stem cells regulates IFN signaling andHCVreplication. Sci. Rep. 8, 1), 13143., DOI 10.1038/s41598-018-31421-6 Zhang, H., Guo, M., Chen, J. H., Wang, Z., Du, X. F., Liu, P. X., et al., 2014). Osteopontin knockdown inhibits alphav,beta3 integrin-induced cell migration and invasion and promotes apoptosis of breast cancer cells by inducing autophagy and inactivating the PI3K/Akt/mTOR pathway. Cell. Physiol. Biochem. 33, 4), 991–1002., DOI 10.1159/000358670 Maeda, N., Ohashi, T., Chagan-Yasutan, H., Hattori, T., Takahashi, Y., Harigae, H., et al., 2015). Osteopontin-integrin interaction as a novel molecular target for antibody-mediated immunotherapy in adult T-cell leukemia. Retrovirology. 12, 99., DOI 10.1186/s12977-015-0225-x Shurin, M. R., 2018). Osteopontin controls immunosuppression in the tumor microenvironment. J. Clin. Invest. 128, 12), 5209–5212., DOI 10.1172/JCI124918 Zhao, B., Sun, T., Meng, F., Qu, A., Li, C., Shen, H., et al., 2011). Osteopontin as a potential biomarker of proliferation and invasiveness for lung cancer. J. Canc. Res. Clin. Oncol. 137, 7), 1061–1070., DOI 10.1007/s00432-010-0968-7 Bandopadhyay, M., Bulbule, A., Butti, R., Chakraborty, G., Ghorpade, P., Ghosh, P., et al., 2014). Osteopontin as a therapeutic target for cancer. Expert Opin. Ther. Targets. 18, 8), 883–895., DOI 10.1517/14728222.2014.925447 Cho,W. Y., Hong, S. H., Singh, B., Islam,M. A., Lee, S., Lee, A. Y., et al., 2015). Suppression of tumor growth in lung cancer xenograft model mice by poly(sorbitol-co-PEI)-mediated delivery of osteopontin siRNA. Eur. J. Pharm. Biopharm. 94, 450–462., DOI 10.1016/j.ejpb.2015.06.017 Subraman, V., Thiyagarajan,M., Malathi, N., and Rajan, S. T., 2015).OPN -Revisited. J. Clin. Diagn. Res. 9, 6), ZE10–ZE13., DOI 10.7860/JCDR/2015/12872.6111 Ben-David-Naim, M., Dagan, A., Grad, E., Aizik, G., Nordling-David, M. M., Morss Clyne, A., et al., 2019). Targeted siRNA nanoparticles for mammary carcinoma therapy. Cancers. 11, 4), 442., DOI 10.3390/cancers11040442 Livak, K. J., and Schmittgen, T. D., 2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T), method. Methods. 25, 4), 402–408., DOI 10.1006/meth.2001.1262 Tuzmen, S., Kiefer, J., and Mousses, S., 2007). Validation of short interfering RNA knockdowns by quantitative real-time PCR. Methods Mol. Biol. 353, 177–203., DOI 10.1385/1-59745-229-7:177 Caffrey, D. R., Zhao, J., Song, Z., Schaffer, M. E., Haney, S. A., Subramanian, R. R., et al., 2011). siRNA off-target effects can be reduced at concentrations that match their individual potency. PloS One. 6, 7), e21503., DOI 10.1371/journal.pone. 0021503 Koroknai, V., Ecsedi, S., Vizkeleti, L., Kiss, T., Szasz, I., Lukacs, A., et al., 2016). Genomic profiling of invasive melanoma cell lines by array comparative genomic hybridization. Melanoma Res. 26, 2), 100–107., DOI 10.1097/CMR. 0000000000000227 Szklarczyk, D., Gable, A. L., Lyon,D., Junge, A.,Wyder, S., Huerta-Cepas, J., et al., 2019). STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47, D1), D607–D613., DOI 10.1093/nar/gky1131 Shevde, L. A., and Samant,R. S., 2014). Role of osteopontin in the pathophysiology of cancer. Matrix Biol. 37, 131–141., DOI 10.1016/j.matbio.2014.03.001 Han, X., Wang, W., He, J., Jiang, L., and Li, X., 2019). Osteopontin as a biomarker for osteosarcoma therapy and prognosis. Oncol Lett. 17, 3), 2592–2598., DOI 10.3892/ol.2019.9905 Maier, T., Laubender, R. P., Sturm, R. A., Klingenstein, A., Korting, H. C., Ruzicka, T., et al., 2012). Osteopontin expression in plasma of melanoma patients and in melanocytic tumours. J. Eur. Acad. Dermatol. Venereol. 26, 9), 1084–1091., DOI 10.1111/j.1468-3083.2011.04210.x Karagiannis, P., Fittall, M., and Karagiannis, S. N., 2014). Evaluating biomarkers in melanoma. Front Oncol. 4, 383., DOI 10.3389/fonc.2014.00383 Mansoori, B., Sandoghchian Shotorbani, S., and Baradaran, B., 2014). RNA interference and its role in cancer therapy. Adv. Pharmaceut. Bull. 4, 4), 313–321., DOI 10.5681/apb.2014.046 Maduri, S., 2015). Applicability of RNA interference in cancer therapy: current status. Indian J. Canc. 52, 1), 11–21., DOI 10.4103/0019-509X.175598 Elazar, V., Adwan, H., Bauerle, T., Rohekar, K., Golomb, G., and Berger, M. R., 2010). Sustained delivery and efficacy of polymeric nanoparticles containing osteopontin and bone sialoprotein antisenses in rats with breast cancer bone metastasis. Int. J. Canc. 126, 7), 1749–1760., DOI 10.1002/ijc.24890 Reufsteck, C., Lifshitz-Shovali, R., Zepp, M., Bauerle, T., Kubler, D., Golomb, G., et al., 2012). Silencing of skeletal metastasis-associated genes impairs migration of breast cancer cells and reduces osteolytic bone lesions. Clin. Exp. Metastasis. 29, 5), 441–456., DOI 10.1007/s10585-012-9462-8 Wu, X. L., Lin, K. J., Bai, A. P., Wang, W. X.,Meng, X. K., Su, X. L., et al., 2014). Osteopontin knockdown suppresses the growth and angiogenesis of colon cancer cells. World J. Gastroenterol. 20, 30), 10440–10448., DOI 10.3748/wjg.v20.i30.10440 Xu, S. T.,Guo, C.,Ding, X., Fan,W. J., Zhang, F.H., Xu,W. L., et al., 2015). Role of osteopontin in the regulation of human bladder cancer proliferation and migration in T24 cells. Mol. Med. Rep. 11, 5), 3701–3707., DOI 10.3892/mmr.2015.3202 Zhou, Y., Dai, D. L., Martinka, M., Su, M., Zhang, Y., Campos, E. I., et al., 2005). Osteopontin expression correlates with melanoma invasion. J. Invest. Dermatol. 124, 5), 1044–1052., DOI 10.1111/j.0022-202X.2005.23680.x Ji, Z., Kumar, R., Taylor, M., Rajadurai, A., Marzuka-Alcala, A., Chen, Y. E., et al., 2013). Vemurafenib synergizes with nutlin-3 to deplete survivin and suppresses melanoma viability and tumor growth. Clin. Canc. Res. 19, 16), 4383–4391., DOI 10.1158/1078-0432.CCR-13-0074 Dong, H., Qian, D.,Wang, Y., Meng, L., Chen, D., Ji, X., et al., 2015). Survivin expression and serum levels in pancreatic cancer. World J. Surg. Oncol. 13, 189., DOI 10.1186/s12957-015-0605-7 Chen, X., Duan, N., Zhang, C., and Zhang, W., 2016). Survivin and tumorigenesis: molecular mechanisms and therapeutic strategies. J. Canc. 7, 3), 314–323., DOI 10.7150/jca.13332 Liu, J., Liu, Q., Wan, Y., Zhao, Z., Yu, H., Luo, H., et al., 2014). Osteopontin promotes the progression of gastric cancer through theNF-kappaB pathway regulated by the MAPK and PI3K. Int. J. Oncol. 45, 1), 282–290., DOI 10.3892/ijo.2014.2393 Grahovac, J., Becker, D., and Wells, A., 2013). Melanoma cell invasiveness is promoted at least in part by the epidermal growth factor-like repeats of tenascin- C. J. Invest. Dermatol. 133, 1), 210–220., DOI 10.1038/jid.2012.263 Matusan-Ilijas, K., Damante, G., Fabbro, D., Dordevic, G., Hadzisejdic, I., Grahovac, M., et al., 2013). EGFR expression is linked to osteopontin and NfkappaB signaling in clear cell renal cell carcinoma. Clin. Transl. Oncol. 15, 1), 65–71., DOI 10.1007/s12094-012-0889-9 Lee, S. H., Park, J. W., Woo, S. H., Go, D. M., Kwon, H. J., Jang, J. J., et al., 2016). Suppression of osteopontin inhibits chemically induced hepatic carcinogenesis by induction of apoptosis in mice. Oncotarget. 7, 52), 87219–87231., DOI 10.18632/oncotarget.13529 Ahmed, H., and AlSadek, D. M., 2015). Galectin-3 as a potential target to prevent cancer metastasis. Clin. Med. Insights Oncol. 9, 113–121., DOI 10.4137/ CMO.S29462 Li, Y. S., Li, X. T., Yu, L. G., Wang, L., Shi, Z. Y., and Guo, X. L., 2020). Roles of galectin-3 in metabolic disorders and tumor cell metabolism. Int. J. Biol. Macromol. 142, 463–473., DOI 10.1016/j.ijbiomac.2019.09.118 Sanchis-Gomar, F., Santos-Lozano, A., Pareja-Galeano, H., Garatachea, N., Alis, R., Fiuza-Luces, C., et al., 2016). Galectin-3, osteopontin and successful aging. Clin. Chem. Lab. Med. 54, 5), 873–877., DOI 10.1515/cclm-2015-0821 Li, X., Chen, Q., Yin, D., Shi, S., Yu, L., Zhou, S., et al., 2017). Novel role of semaphorin 3A in the growth and progression of hepatocellular carcinoma. Oncol. Rep. 37, 6), 3313–3320., DOI 10.3892/or.2017.5616 Vizin, T., and Kos, J., 2015). Gamma-enolase: awell-known tumourmarker,with a less-known role in cancer. Radiol. Oncol. 49, 3), 217–226., DOI 10.1515/raon-2015- 0035 Lavoie, H., Li, J. J., Thevakumaran, N., Therrien, M., and Sicheri, F., 2014). Dimerization-induced allostery in protein kinase regulation. Trends Biochem. Sci. 39, 10), 475–486., DOI 10.1016/j.tibs.2014.08.004 Nangia-Makker, P., Hogan, V., and Raz, A., 2018). Galectin-3 and cancer stemness. Glycobiology. 28, 4), 172–181., DOI 10.1093/glycob/cwy001 Ibrahim, T.M., Ernst, C., Lange,A., Hennig, S., and Boeckler, F. M., 2019). Smallmolecule intervention at the dimerization interface of survivin by novel rigidized scaffolds. Drug Des. Dev. Ther. 13, 4247–4263., DOI 10.2147/DDDT.S224561 Yue, B., 2014). Biology of the extracellular matrix: an overview. J. Glaucoma. 23, 8 Suppl. 1), S20–S23., DOI 10.1097/IJG.0000000000000108 Tuck, A. B., Hota, C.,Wilson, S. M., and Chambers, A. F., 2003). Osteopontininduced migration of human mammary epithelial cells involves activation of EGF receptor and multiple signal transduction pathways. Oncogene. 22, 8), 1198–1205., DOI 10.1038/sj.onc.1206209 Jones, P. L., Crack, J., and Rabinovitch, M., 1997). Regulation of tenascin-C, a vascular smooth muscle cell survival factor that interacts with the alpha v beta 3 integrin to promote epidermal growth factor receptor phosphorylation and growth. J. Cell Biol. 139, 1), 279–293., DOI 10.1083/jcb.139.1.279 Hayashi, C., Rittling, S., Hayata, T., Amagasa, T., Denhardt, D., Ezura, Y., et al., 2007). Serum osteopontin, an enhancer of tumor metastasis to bone, promotes B16 melanoma cell migration. J. Cell. Biochem. 101, 4), 979–986., DOI 10.1002/jcb.21298 Eberwein, P., Laird, D., Schulz, S., Reinhard, T., Steinberg, T., and Tomakidi, P., 2015). Modulation of focal adhesion constituents and their down-stream events by EGF: on the cross-talk of integrins and growth factor receptors. Biochim. Biophys. Acta. 1853, 10 Pt A), 2183–2198., DOI 10.1016/j.bbamcr. 2015.06.004 Huang, R. H., Quan, Y. J., Chen, J. H.,Wang, T. F., Xu, M., Ye, M., et al., 2017). Osteopontin promotes cell migration and invasion, and inhibits apoptosis and autophagy in colorectal cancer by activating the p38 MAPK signaling pathway. Cell. Physiol. Biochem. 41, 5), 1851–1864., DOI 10. 1159/000471933 Eke, I., and Cordes, N., 2015). Focal adhesion signaling and therapy resistance in cancer. Semin. Canc. Biol. 31, 65–75., DOI 10.1016/j. semcancer.2014.07.009 Sun, Y., Liu, W. Z., Liu, T., Feng, X., Yang, N., and Zhou, H. F., 2015). Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J. Recept. Signal Transduct. Res. 35, 6), 600–604., DOI 10.3109/10799893.2015.1030412 Savoia, P., Fava, P., Casoni, F., and Cremona, O., 2019). Targeting the ERK signaling pathway in melanoma. Int. J. Mol. Sci. 20, 6), 1483., DOI 10.3390/ ijms20061483 Zhang, X. Y., and Zhang, P. Y., 2016). Genetics and epigenetics of melanoma. Oncol Lett. 12, 5), 3041–3044., DOI 10.3892/ol.2016.5093 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 581395 EP 581404 DI 10.3389/pore.2021.581395 PG 10 ER PT J AU Gao, Y Gan, K Liu, K Xu, B Chen, M AF Gao, Yue Gan, Kai Liu, Kuangzheng Xu, Bin Chen, Ming TI SP1 Expression and the Clinicopathological Features of Tumors: A Meta-Analysis and Bioinformatics Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE specificity protein 1; cancer; clinicopathological characteristics; prognosis; biomarker ID specificity protein 1; cancer; clinicopathological characteristics; prognosis; biomarker AB Objective: Specificity protein 1 (SP1) plays a vital role to promote carcinogenesis in a variety of tumors, and its up-regulated expression is reported to be a hinter of poor prognosis of patients. We conducted this meta-analysis to elucidate the clinical significance and prognostic value of SP1 in malignant tumors. Methods: PubMed and Cochrane Library were searched for studies published between January 1, 2000 and June 1, 2020. The combined odds ratios (ORs) and hazard ratios (HRs) with 95% confidence intervals (95%CIs)were used to investigate the correlation of SP1 with clinical behaviors and prognosis in patients with solid tumors. UALCAN was used to conduct bioinformatics analysis. Results: A total of 24 documents involving 2,739 patients were enrolled in our review. The random-effectmodel was used to perform this analysis due to the high level of heterogeneity. SP1 low expression was not conducive to lymph nodemetastasis (OR=0.42; 95% CI: 0.28-0.64; p < 0.05), progression of TNM stage (OR=0.34; 95% CI: 0.20-0.57; p < 0.05) and tumor infiltration (OR=0.33; 95% CI: 0.18-0.60; p < 0.05). Elevated SP1 expression was connected with shorter survival time of patients with hepatocellular carcinoma, pancreatic cancer, gastric cancer and esophageal cancer (HR=1.95; 95% CI: 1.16-3.28; p < 0.05). According to UALCAN database, breast cancer, ovarian cancer, colon cancer and lung adenocarcinoma display an elevated SP1 expression in comparison with normal tissues. Kaplan-Meier survival plots indicate SP1mRNA level has negative effects on prognosis of liver hepatocellular carcinoma and brain lower grade glioma. Conclusion: SP1 was associated with lymph node metastasis, TNM stage and depth of invasion, and indicated poor clinical outcome, which brought new insights on the potential candidacy of SP1 in clinical usage. C1 [Gao, Yue] Medical School of Southeast University Nanjing, Institute of Urology, Surgical Research CenterJiangsu, China. [Gan, Kai] Medical School of Southeast University Nanjing, Institute of Urology, Surgical Research CenterJiangsu, China. [Liu, Kuangzheng] Medical School of Southeast University Nanjing, Institute of Urology, Surgical Research CenterJiangsu, China. [Xu, Bin] Affiliated Zhongda Hospital of Southeast University, Department of UrologyJiangsu, China. [Chen, Ming] Affiliated Zhongda Hospital of Southeast University, Department of UrologyJiangsu, China. RP Chen, M (reprint author), Affiliated Zhongda Hospital of Southeast University, Department of Urology, Jiangsu, China. EM mingchenseu@126.com CR Beishline, K., and Azizkhan-Clifford, J., 2015). Sp1 and the ‘hallmarks of cancer’. FEBS J. 282, 2), 224–258., DOI 10.1111/febs.13148 Maor, S., Mayer, D., Yarden, R. I., Lee, A. V., Sarfstein, R., Werner, H., et al., 2006). Estrogen receptor regulates insulin-like growth factor-I receptor gene expression in breast tumor cells: involvement of transcription factor Sp1. J. Endocrinol. 191, 3), 605–612., DOI 10.1677/joe.1.07016 Santra, M., Santra, S., Zhang, J., and Chopp, M., 2008). Ectopic decorin expression up-regulates VEGF expression in mouse cerebral endothelial cells via activation of the transcription factors Sp1, HIF1alpha, and Stat3. J. Neurochem. 105, 2), 324–337., DOI 10.1111/j.1471-4159.2007.05134.x Hung, W. C., Tseng, W. L., Shiea, J., and Chang, H. C., 2010). Skp2 overexpression increases the expression of MMP-2 and MMP-9 and invasion of lung cancer cells. Canc. Lett. 288, 2), 156–161., DOI 10.1016/j. canlet.2009.06.032 Chang, S., Sun, L., and Feng, G., 2019). SP1-mediated long noncoding RNA POU3F3 accelerates the cervical cancer through miR-127-5p/FOXD1. Biomed. Pharmacother. 117, 109133., DOI 10.1016/j.biopha.2019.109133 Zhang, B., Song, L., Cai, J., Li, L., Xu, H., Li, M., et al., 2019). The LIM protein Ajuba/SP1 complex forms a feed forward loop to induce SP1 target genes and promote pancreatic cancer cell proliferation. J. Exp. Clin. Canc. Res. 38, 1), 205., DOI 10.1186/s13046-019-1203-2 Chandrashekar, D. S., Bashel, B., Balasubramanya, S. A. H., Creighton, C. J., Ponce-Rodriguez, I., Chakravarthi, B. V. S. K., et al., 2017). UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 19, 8), 649–658., DOI 10.1016/j.neo.2017.05.002 Wang, L., Wei, D., Huang, S., Peng, Z., Le, X., Wu, T. T., et al., 2003). Transcription factor Sp1 expression is a significant predictor of survival in human gastric cancer. Clin. Canc. Res. 9, 17), 6371–6380. Yao, J. C., Wang, L., Wei, D., Gong, W., Hassan, M., Wu, T. T., et al., 2004). Association between expression of transcription factor Sp1 and increased vascular endothelial growth factor expression, advanced stage, and poor survival in patients with resected gastric cancer. Clin. Canc. Res. 10, 12 Pt 1), 4109–4117., DOI 10.1158/1078-0432.CCR-03-0628 Zhang, J., Zhu, Z. G., Ji, J., Yuan, F., Yu, Y. Y., Liu, B. Y., et al., 2005). Transcription factor Sp1 expression in gastric cancer and its relationship to long-term prognosis. World J. Gastroenterol. 11, 15), 2213–2217., DOI 10.3748/ wjg.v11.i15.2213 Wang, X. B., Peng, W. Q., Yi, Z. J., Zhu, S. L., and Gan, Q. H., 2007). [Expression and prognostic value of transcriptional factor sp1 in breast cancer]. Ai Zheng. 26, 9), 996–1000. Guan, H., Cai, J., Zhang, N., Wu, J., Yuan, J., Li, J., et al., 2012). Sp1 is upregulated in human glioma, promotes MMP-2-mediated cell invasion and predicts poor clinical outcome. Int. J. Canc. 130, 3), 593–601., DOI 10.1002/ijc. 26049 Lee, H. S., Park, C. K., Oh, E., Erkin, O. C., Jung, H. S., Cho, M. H., et al., 2013). Low SP1 expression differentially affects intestinal-type compared with diffuse-type gastric adenocarcinoma. PloS One. 8, 2), e55522., DOI 10.1371/ journal.pone.0055522 Wang, F., Ma, Y. L., Zhang, P., Shen, T. Y., Shi, C. Z., Yang, Y. Z., et al., 2013). SP1 mediates the link between methylation of the tumour suppressor miR-149 and outcome in colorectal cancer. J. Pathol. 229, 1), 12–24., DOI 10.1002/path.4078 Dong, Q., Cai, N., Tao, T., Zhang, R., Yan, W., Li, R., et al., 2014). An axis involving SNAI1, microRNA-128 and SP1 modulates glioma progression. PloS One. 9, 6), e98651., DOI 10.1371/journal.pone.0098651 Li, L., Gao, P., Li, Y., Shen, Y., Xie, J., Sun, D., et al., 2014). JMJD2A-dependent silencing of Sp1 in advanced breast cancer promotes metastasis by downregulation of DIRAS3. Breast Canc. Res. Treat. 147, 3), 487–500., DOI 10.1007/s10549-014-3083-7 Zhang, J. P., Zhang, H.,Wang, H. B., Li, Y. X., Liu, G. H., Xing, S., et al., 2014). Down-regulation of Sp1 suppresses cell proliferation, clonogenicity and the expressions of stem cell markers in nasopharyngeal carcinoma. J. Transl. Med. 12, 222., DOI 10.1186/s12967-014-0222-1 Jiang, W., Jin, Z., Zhou, F., Cui, J., Wang, L., and Wang, L., 2015). High coexpression of Sp1 and HER-2 is correlated with poor prognosis of gastric cancer patients. Surg. Oncol. 24, 3), 220–225., DOI 10.1016/j.suronc.2015.05.004 Wang, J., Kang, M., Qin, Y. T., Wei, Z. X., Xiao, J. J., and Wang, R. S., 2015). Sp1 is over-expressed in nasopharyngeal cancer and is a poor prognostic indicator for patients receiving radiotherapy. Int. J. Clin. Exp. Pathol. 8, 6), 6936–6943. Hang, J., Hu, H., Huang, J., Han, T., Zhuo, M., Zhou, Y., et al., 2016). Sp1 and COX2 expression is positively correlated with a poor prognosis in pancreatic ductal adenocarcinoma. Oncotarget. 7, 19), 28207–28217., DOI 10.18632/ oncotarget.8593 Hu, J., Hu, H., Hang, J. J., Yang, H. Y., Wang, Z. Y., Wang, L., et al., 2016). Simultaneous high expression of PLD1 and Sp1 predicts a poor prognosis for pancreatic ductal adenocarcinoma patients. Oncotarget. 7, 48), 78557–78565., DOI 10.18632/oncotarget.12447 Liu, L., Ji, P., Qu, N., Pu, W. L., Jiang, D. W., Liu, W. Y., et al., 2016). The impact of high co-expression of Sp1 and HIF1α on prognosis of patients with hepatocellular cancer. Oncol. Lett. 12, 1), 504–512., DOI 10.3892/ol.2016.4634 Hu, H., Wu, L. L., Han, T., Zhuo, M., Lei, W., Cui, J. J., et al., 2017). Correlated high expression of FXR and Sp1 in cancer cells confers a poor prognosis for pancreatic cancer: a study based on TCGA and tissuemicroarray. Oncotarget. 8, 20), 33265–33275., DOI 10.18632/oncotarget.16633 Qian, Y., Yao, W., Yang, T., Yang, Y., Liu, Y., Shen, Q., et al., 2017). aPKC- ι/P-Sp1/Snail signaling induces epithelial-mesenchymal transition and immunosuppression in cholangiocarcinoma. Hepatology. 66, 4), 1165–1182., DOI 10.1002/hep.29296 Wang, X. X., Liao, Y., Hong, L., Zeng, Z., Yuan, T. B., Xia, X., et al., 2017). Tissue microarray staining reveals PLD1 and Sp1 have a collaborative, protumoral effect in patients with osteosarcomas. Oncotarget. 8, 43), 74340–74347., DOI 10.18632/oncotarget.20605 Chen, Y. T., Tsai, H. P., Wu, C. C., Chen, C. Y., Chai, C. Y., and Kwan, A. L., 2019). High-level Sp1 is associated with proliferation, invasion, and poor prognosis in astrocytoma. Pathol. Oncol. Res. 25, 3), 1003–1013., DOI 10.1007/ s12253-018-0422-8 Gu, L., Sang, M., Li, J., Liu, F., Wu, Y., Liu, S., et al., 2019). Expression and prognostic significance of MAGE-A11 and transcription factors, SP1,TFCP2 and ZEB1, in ESCC tissues. Pathol. Res. Pract. 215, 7), 152446., DOI 10.1016/j. prp.2019.152446 Kim, I. K., Lee, Y. S., Kim, H. S., Dong, S. M., Park, J. S., and Yoon, D. S., 2019). Specific protein 1(SP1, regulates the epithelial-mesenchymal transition via lysyl oxidase-like 2(LOXL2, in pancreatic ductal adenocarcinoma. Sci. Rep. 9, 1), 5933., DOI 10.1038/s41598-019-42501-6 Hu, X., Lin, J., Jiang, M., He, X., Wang, K., Wang, W., et al., 2020). HIF-1α promotes the metastasis of esophageal squamous cell carcinoma by targeting SP1. J. Canc. 11, 1), 229–240., DOI 10.7150/jca.35537 Li, J., Peng,W., Yang, P., Chen, R., Gu, Q., Qian,W., et al., 2020). MicroRNA- 1224-5p inhibits metastasis and epithelial-mesenchymal transition in colorectal cancer by targeting SP1-mediated NF-κB signaling pathways. Front. Oncol. 10, 294., DOI 10.3389/fonc.2020.00294 Bajpai, R., and Nagaraju, G. P., 2017). Specificity protein 1: its role in colorectal cancer progression and metastasis. Crit. Rev. Oncol. Hematol. 113, 1–7., DOI 10. 1016/j.critrevonc.2017.02.024 Liu, X. B.,Wang, J., Li, K., and Fan, X. N., 2019). Sp1 promotes cell migration and invasion in oral squamous cell carcinoma by upregulating Annexin A2 transcription. Mol. Cell. Probes. 46, 101417., DOI 10.1016/j.mcp.2019.06.007 Yu, Y., Peng, K., Li, H., Zhuang, R., Wang, Y., Li, W., et al., 2018). SP1 upregulated FoxO3a promotes tumor progression in colorectal cancer. Oncol. Rep. 39, 5), 2235–2242., DOI 10.3892/or.2018.6323 Huang, Z., Huang, L., Shen, S., Li, J., Lu, H., Mo, W., et al., 2015). Sp1 cooperates with Sp3 to upregulate MALAT1 expression in human hepatocellular carcinoma. Oncol. Rep. 34, 5), 2403–2412., DOI 10.3892/or. 2015.4259 Mir, R., Sharma, A., Pradhan, S. J., and Galande, S., 2018). Regulation of transcription factor SP1 by the β-catenin destruction complex modulates Wnt response. Mol. Cell Biol. 38, 22), e00188-18., DOI 10.1128/MCB.00188-18 Monteleone, E., Orecchia, V., Corrieri, P., Schiavone, D., Avalle, L., Moiso, E., et al., 2019). SP1 and STAT3 functionally synergize to induce the RhoU small GTPase and a subclass of non-canonical WNT responsive genes correlating with poor prognosis in breast cancer. Cancers. 11, 1), 101., DOI 10.3390/ cancers11010101 Kwon, Y. J., Baek, H. S., Ye, D. J., Shin, S., Kim, D., and Chun, Y. J., 2016). CYP1B1 enhances cell proliferation and metastasis through induction of EMT and activation of wnt/β-catenin signaling via Sp1 upregulation. PloS One. 11, 3), e0151598., DOI 10.1371/journal.pone.0151598 Sun, Y., Xu, K., He, M., Fan, G., and Lu, H., 2018). Overexpression of glypican 5, GPC5, inhibits prostate cancer cell proliferation and invasion via suppressing sp1-mediated EMT and activation of wnt/β-catenin signaling. Oncol. Res. 26, 4), 565–572., DOI 10.3727/096504017X15044461944385 Cheang, M. C., Treaba, D. O., Speers, C. H., Olivotto, I. A., Bajdik, C. D., Chia, S. K., et al., 2006). Immunohistochemical detection using the new rabbit monoclonal antibody SP1 of estrogen receptor in breast cancer is superior to mouse monoclonal antibody 1D5 in predicting survival. J. Clin. Oncol. 24, 36), 5637–5644., DOI 10.1200/JCO.2005.05.4155 Welsh, A. W., Harigopal, M., Wimberly, H., Prasad, M., and Rimm, D. L., 2013). Quantitative analysis of estrogen receptor expression shows SP1 antibody is more sensitive than 1D5. Appl. Immunohistochem. Mol. Morphol. 21, 2), 139–147., DOI 10.1097/PAI.0b013e31825d73b2 Bae, Y. K., Gong, G., Kang, J., Lee, A., Cho, E. Y., Lee, J. S., et al., 2012). Hormone receptor expression in invasive breast cancer among Korean women and comparison of 3 antiestrogen receptor antibodies: a multi-institutional retrospective study using tissue microarrays. Am. J. Surg. Pathol. 36, 12), 1817–1825., DOI 10.1097/PAS.0b013e318267b012 Kozlowicz-Gudzinska, I., Pietraszek, A., and Kaminska, J., 1989). Initial concentration of the neoplastic biological markers AFP, alpha fetoprotein), HCG, chorionic gonadotropin), and SP1, specific pregnancy protein, in separate types and stages of clinically advanced lung cancer]. Nowotwory. 39, 3-4), 162–165. Ordonez, N. G., 1989). The immunohistochemical diagnosis of mesothelioma. Differentiation of mesothelioma and lung adenocarcinoma. Am. J. Surg. Pathol. 13, 4), 276–291. Bedolla, R. G., Gong, J., Prihoda, T. J., Yeh, I. T., Thompson, I. M., Ghosh, R., et al., 2012). Predictive value of Sp1/Sp3/FLIP signature for prostate cancer recurrence. PloS One. 7, 9), e44917., DOI 10.1371/journal.pone.0044917 Zhao, Q., Cai, W., Zhang, X., Tian, S., Zhang, J., Li, H., et al., 2017). RYBP expression is regulated by KLF4 and Sp1 and is related to hepatocellular carcinoma prognosis. J. Biol. Chem. 292, 6), 2143–2158., DOI 10.1074/jbc.M116. 770727 Ren, Y., Zhang, H., and Jiang, P., 2018). MicroRNA-382 inhibits cell growth and migration in colorectal cancer by targeting SP1. Biol. Res. 51, 1), 51., DOI 10. 1186/s40659-018-0200-9 Wang, Z. Q., Cai, Q., Hu, L., He, C. Y., Li, J. F., Quan, Z. W., et al., 2017). Long noncoding RNA UCA1 induced by SP1 promotes cell proliferation via recruiting EZH2 and activating AKT pathway in gastric cancer. Cell Death Dis. 8, 6), e2839., DOI 10.1038/cddis.2017.143 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 581998 EP 582009 DI 10.3389/pore.2021.581998 PG 12 ER PT J AU Xin, W Zhao, Ch Jiang, L Pei, D Zhao, L Zhang, Ch AF Xin, Wei Zhao, Chaoran Jiang, Longyang Pei, Dongmei Zhao, Lin Zhang, Chengpu TI Identification of a Novel Epithelial–Mesenchymal Transition Gene Signature Predicting Survival in Patients With HNSCC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE epithelial-mesenchymal transition; HNSCC; mRNAs; Prognostic; survival ID epithelial-mesenchymal transition; HNSCC; mRNAs; Prognostic; survival AB Head and neck squamous cell cancer (HNSCC) is one of the most common types of cancer worldwide. There have beenmany reports suggesting that biomarkers explored via database mining plays a critical role in predicting HNSCC prognosis. However, a single biomarker for prognostic analysis is not adequate. Additionally, there is growing evidence indicating that gene signature could be a better choice for HNSCC prognosis. We performed a comprehensive analysis of mRNA expression profiles using clinical information of HNSCC patients from The Cancer Genome Atlas (TCGA). Gene Set Enrichment Analysis (GSEA) was performed, and we found that a set of genes involved in epithelial mesenchymal transition (EMT) contributed to HNSCC. Cox proportional regressionmodel was used to identify a fourgene (WIPF1, PPIB, BASP1, PLOD2) signature that were significantly associated with overall survival (OS), and all the four genes were significantly upregulated in tumor tissues. We successfully classified the patients with HNSCC into high-risk and low-risk groups, where in high-risk indicated poorer patient prognosis, indicating that this gene signature might be a novel potential biomarker for the prognosis of HNSCC. The prognostic ability of the gene signature was further validated in an independent cohort fromthe Gene Expression Omnibus (GEO) database. In conclusion, we identified a four-EMT-based gene signature which provides the potentiality to serve as novel independent biomarkers for predicting survival in HNSCC patients, as well as a new possibility for individualized treatment of HNSCC. C1 [Xin, Wei] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Zhao, Chaoran] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Jiang, Longyang] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Pei, Dongmei] China Medical University, Shengjing Hospital, Department of Family MedicineShenyang, China. [Zhao, Lin] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Zhang, Chengpu] China Medical University, Shengjing Hospital, Department of Family MedicineShenyang, China. RP Zhang, Ch (reprint author), China Medical University, Shengjing Hospital, Department of Family Medicine, Shenyang, China. EM zhangcp@sj-hospital.org CR Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA: A Cancer J Clinicians, 2016, 66(1):7–30., DOI 10.3322/caac.21332 Garcia-Carracedo D, Villaronga MA, Alvarez-Teijeiro S, Hermida-Prado F, Santamaria I, Allonca E, et al. Impact of PI3K/AKT/mTOR pathway activation on the prognosis of patients with head and neck squamous cell carcinomas. Oncotarget, 2016, 7(20):29780–93., DOI 10.18632/oncotarget.8957 Leemans CR, Braakhuis BJM, Brakenhoff RH. The molecular biology of head and neck cancer. Nat Rev Cancer, 2011, 11(1):9–22., DOI 10.1038/nrc2982 Patel SG, Shah JP. TNM staging of cancers of the head and neck: striving for uniformity among diversity. CA: A Cancer J Clinicians, 2005, 55(4):242–58., DOI 10.3322/canjclin.55.4.242 Wyss A, Hashibe M, Chuang S-C, Lee Y-CA, Zhang Z-F, Yu G-P, et al. Cigarette, cigar, and pipe smoking and the risk of head and neck cancers: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. Am J Epidemiol, 2013, 178(5):679–90., DOI 10.1093/aje/ kwt029 Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tˆan PF, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med, 2010, 363(1):24–35., DOI 10.1056/nejmoa0912217 Xiang Y, Li F, Wang L, Zheng A, Zuo J, Li M, et al. Decreased calpain 6 expression is associated with tumorigenesis and poor prognosis in HNSCC. Oncol Lett, 2017, 13(4):2237–43., DOI 10.3892/ol.2017.5687 Wang Y, Liu Y, Liu H, Zhang Q, Song H, Tang J, et al. FcGBP was upregulated by HPV infection and correlated to longer survival time of HNSCC patients. Oncotarget, 2017, 8(49):86503–14., DOI 10.18632/oncotarget.21220 Chen Y-L, Ge G-J, Qi C, Wang H, Wang H-L, Li L-Y, et al. A five-gene signature may predict sunitinib sensitivity and serve as prognostic biomarkers for renal cell carcinoma. J Cel Physiol, 2018, 233(10):6649–60., DOI 10.1002/jcp.26441 Cheng W, Ren X, Zhang C, Cai J, Liu Y, Han S, et al. Bioinformatic profiling identifies an immune-related risk signature for glioblastoma. Neurology, 2016, 86(24):2226–34., DOI 10.1212/wnl.0000000000002770 Yeung KT, Yang J. Epithelial-mesenchymal transition in tumor metastasis. Mol Oncol, 2017, 11(1):28–39., DOI 10.1002/1878-0261.12017 Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, Gillespie S, et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell, 2017, 171(7):1611–24. e24., DOI 10.1016/j.cell.2017. 10.044 Puram SV, Parikh AS, Tirosh I. Single cell RNA-seq highlights a role for a partial EMT in head and neck cancer. Mol Cell Oncol, 2018, 5(3):e1448244., DOI 10.1080/23723556.2018.1448244 Thomas MA, Yang L, Carter BJ, Klaper RD. Gene set enrichment analysis of microarray data from Pimephales promelas, Rafinesque), a non-mammalian model organism. BMC Genomics, 2011, 12:66., DOI 10.1186/1471-2164-12-66 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci, 2005, 102(43):15545–50., DOI 10.1073/pnas.0506580102 Cheng W, Li M, Cai J, Wang K, Zhang C, Bao Z, et al. HDAC4, a prognostic and chromosomal instability marker, refines the predictive value of MGMT promoter methylation. J Neurooncol, 2015, 122(2):303–12., DOI 10.1007/ s11060-014-1709-6 Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc, 2009, 4(1): 44–57., DOI 10.1038/nprot.2008.211 Guo W, Chen X, Zhu L, Wang Q. A six-mRNA signature model for the prognosis of head and neck squamous cell carcinoma. Oncotarget, 2017, 8(55): 94528–38., DOI 10.18632/oncotarget.21786 Li L, Zhang L, Zhang Y, Zhou F. Increased expression of LncRNA BANCR is associated with clinical progression and poor prognosis in gastric cancer. Biomed Pharmacother, 2015, 72:109–12., DOI 10.1016/j.biopha.2015. 04.007 Shan H, Yang Y, Zhu X, Han X, Zhang P, Zhang X. FAM83H-AS1 is associated with clinical progression and modulates cell proliferation, migration, and invasion in bladder cancer. J Cel Biochem, 2018, 120(3):4687–93., DOI 10.1002/ jcb.27758 Ng SWK, Mitchell A, Kennedy JA, Chen WC, McLeod J, Ibrahimova N, et al. A 17-gene stemness score for rapid determination of risk in acute leukaemia. Nature, 2016, 540(7633):433–7., DOI 10.1038/nature20598 Bao Z-S, Li M-Y, Wang J-Y, Zhang C-B, Wang H-J, Yan W, et al. Prognostic value of a nine-gene signature in glioma patients based on mRNA expression profiling. CNS Neurosci Ther, 2014, 20(2):112–8., DOI 10.1111/cns.12171 Niyazi M, Pitea A, MittelbronnM, Steinbach J, Sticht C, Zehentmayr F, et al. A 4-miRNA signature predicts the therapeutic outcome of glioblastoma. Oncotarget, 2016, 7(29):45764–75., DOI 10.18632/oncotarget.9945 ChengW, Ren X, Cai J, Zhang C, Li M, Wang K, et al. A five-miRNA signature with prognostic and predictive value for MGMT promoter-methylated glioblastoma patients. Oncotarget, 2015, 6(30):29285–95., DOI 10.18632/ oncotarget.4978 Chen P-f., Wang F, Zhang Z-x., Nie J-y., Liu L, Feng J-r., et al. A novel genepair signature for relapse-free survival prediction in colon cancer. Cmar, 2018, 10:4145–53., DOI 10.2147/cmar.s176260 Peng P-L, Zhou X-Y, Yi G-D, Chen P-F, Wang F, Dong W-G. Identification of a novel gene pairs signature in the prognosis of gastric cancer. Cancer Med, 2018, 7(2):344–50., DOI 10.1002/cam4.1303 Yan H, Xin S, Ma J, Wang H, Zhang H, Liu J. A three microRNA-based prognostic signature for small cell lung cancer overall survival. J Cel Biochem, 2018, 120(5):8723–30., DOI 10.1002/jcb.28159 Brabletz T, Jung A, Reu S, Porzner M, Hlubek F, Kunz-Schughart LA, et al. Variable -catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci, 2001, 98(18):10356–61., DOI 10.1073/pnas.171610498 Baek T-H, Kang D-W, Kim J-H, Son H-J. Gland attenuation, a novel morphological feature of colorectal cancer: evidence for an epithelialmesenchymal transition. Ann Coloproctol, 2018, 34(4):187–96., DOI 10.3393/ ac.2017.12.02 Thiery JP. Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cel Biol, 2003, 15(6):740–6., DOI 10.1016/j.ceb.2003. 10.006 Brabletz T, Jung A, Spaderna S, Hlubek F, Kirchner T. Migrating cancer stem cells - an integrated concept of malignant tumour progression. Nat Rev Cancer, 2005, 5(9):744–9., DOI 10.1038/nrc1694 Hollier BG, Evans K, Mani SA. The epithelial-to-mesenchymal transition and cancer stem cells: a coalition against cancer therapies. J Mammary Gland Biol Neoplasia, 2009, 14(1):29–43., DOI 10.1007/s10911-009-9110-3 Blick T, Widodo E, Hugo H, Waltham M, Lenburg ME, Neve RM, et al. Epithelial mesenchymal transition traits in human breast cancer cell lines. Clin Exp Metastasis, 2008, 25(6):629–42., DOI 10.1007/s10585-008-9170-6 Wu D, Cheng J, Sun G, Wu S, Li M, Gao Z, et al. p70S6K promotes IL-6- induced epithelial-mesenchymal transition and metastasis of head and neck squamous cell carcinoma. Oncotarget, 2016, 7(24):36539–50., DOI 10.18632/ oncotarget.9282 Ock C-Y, Kim S, Keam B, KimM, Kim TM, Kim J-H, et al. PD-L1 expression is associated with epithelial-mesenchymal transition in head and neck squamous cell carcinoma. Oncotarget, 2016, 7(13):15901–14., DOI 10.18632/oncotarget. 7431 Zhang W, Li J, Wu Y, Ge H, Song Y, Wang D, et al. TEAD4 overexpression promotes epithelial-mesenchymal transition and associates with aggressiveness and adverse prognosis in head neck squamous cell carcinoma. Cancer Cel Int, 2018, 18:178., DOI 10.1186/s12935-018-0675-z Obata Y, Yamamoto K, Miyazaki M, Shimotohno K, Kohno S, Matsuyama T. Role of cyclophilin B in activation of interferon regulatory factor-3. J Biol Chem, 2005, 280(18):18355–60., DOI 10.1074/jbc.m501684200 Rycyzyn MA, Clevenger CV. The intranuclear prolactin/cyclophilin B complex as a transcriptional inducer. Proc Natl Acad Sci, 2002, 99(10):6790–5., DOI 10. 1073/pnas.092160699 Bukrinsky MI. Cyclophilins: unexpected messengers in intercellular communications. Trends Immunol, 2002, 23(7):323–5., DOI 10.1016/s1471- 4906(02)02237-8 Choi TG, Nguyen MN, Kim J, Jo YH, Jang M, Nguyen NNY, et al. Cyclophilin B induces chemoresistance by degrading wild-type p53 via interaction with MDM2 in colorectal cancer. J Pathol, 2018, 246(1):115–26., DOI 10.1002/path. 5107 Meng DQ, Li PL, Xie M. Expression and role of cyclophilin B in stomach cancer. Genet Mol Res, 2015, 14(2):5346–54., DOI 10.4238/2015.may.22.5 Kim Y, JangM, Lim S, Won H, Yoon K-S, Park J-H, et al. Role of cyclophilin B in tumorigenesis and cisplatin resistance in hepatocellular carcinoma in humans. Hepatology, 2011, 54(5):1661–78., DOI 10.1002/hep.24539 Ray P, Rialon-Guevara KL, Veras E, Sullenger BA, White RR. Comparing human pancreatic cell secretomes by in vitro aptamer selection identifies cyclophilin B as a candidate pancreatic cancer biomarker. J Clin Invest, 2012, 122(5):1734–41., DOI 10.1172/jci62385 Toska E, Shandilya J, Goodfellow SJ, Medler KF, Roberts SGE. Prohibitin is required for transcriptional repression by the WT1-BASP1 complex. Oncogene, 2014, 33(43):5100–8., DOI 10.1038/onc.2013.447 Carpenter B, Hill KJ, Charalambous M, Wagner KJ, Lahiri D, James DI, et al. BASP1 is a transcriptional cosuppressor for the Wilms’ tumor suppressor protein WT1. Mcb, 2004, 24(2):537–49., DOI 10.1128/mcb.24.2.537-549.2004 Sugiyama H. WT1, Wilms’ tumor gene 1): biology and cancer immunotherapy. Jpn J Clin Oncol, 2010, 40(5):377–87., DOI 10.1093/jjco/hyp194 Qi X-W, Zhang F, Yang X-H, Fan L-J, Zhang Y, Liang Y, et al. High Wilms’ tumor 1 mRNA expression correlates with basal-like and ERBB2 molecular subtypes and poor prognosis of breast cancer. Oncol Rep, 2012, 28(4):1231–6., DOI 10.3892/or.2012.1906 Oji Y, Miyoshi Y, Koga S, Nakano Y, Ando A, Nakatsuka S-i., et al. Overexpression of the Wilms’ tumor gene WT1 in primary thyroid cancer. Cancer Sci, 2003, 94(7):606–11., DOI 10.1111/j.1349-7006.2003.tb01490.x Guo R-S, Yu Y, Chen J, Chen Y-Y, Shen N, QiuM. Restoration of brain acid soluble protein 1 inhibits proliferation and migration of thyroid cancer cells. Chin Med J, Engl,, 2016, 129(12):1439–46., DOI 10.4103/0366-6999. 183434 Wu C, Wang Y, Xia Y, He S,Wang Z, Chen Y, et al. Wilms’ tumor 1 enhances Cisplatin-resistance of advanced NSCLC. FEBS Lett, 2014, 588(24):4566–72., DOI 10.1016/j.febslet.2014.10.026 Li X, Ottosson S, Wang S, Jernberg E, Boldrup L, Gu X, et al. Wilms’ tumor gene 1 regulates p63 and promotes cell proliferation in squamous cell carcinoma of the head and neck. BMC Cancer, 2015, 15:342., DOI 10.1186/ s12885-015-1356-0 Yamauchi M, Sricholpech M. Lysine post-translational modifications of collagen. Essays Biochem, 2012, 52:113–33., DOI 10.1042/bse0520113 Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG, Keely PJ. Collagen reorganization at the tumor-stromal interface facilitates local invasion. BMC Med, 2006, 4(1):38., DOI 10.1186/1741-7015-4-38 Chen Y, Guo H, Terajima M, Banerjee P, Liu X, Yu J, et al. Lysyl hydroxylase 2 is secreted by tumor cells and can modify collagen in the extracellular space. J Biol Chem, 2016, 291(50):25799–808., DOI 10.1074/ jbc.m116.759803 Miyamoto K, Seki N, Matsushita R, Yonemori M, Yoshino H, Nakagawa M, et al. Tumour-suppressive miRNA-26a-5p and miR-26b-5p inhibit cell aggressiveness by regulating PLOD2 in bladder cancer. Br J Cancer, 2016, 115(3):354–63., DOI 10.1038/bjc.2016.179 Kurozumi A, Kato M, Goto Y, Matsushita R, Nishikawa R, Okato A, et al. Regulation of the collagen cross-linking enzymes LOXL2 and PLOD2 by tumor-suppressive microRNA-26a/b in renal cell carcinoma. Int J Oncol, 2016, 48(5):1837–46., DOI 10.3892/ijo.2016.3440 Reis PP, Waldron L, Goswami RS, Xu W, Xuan Y, Perez-Ordonez B, et al. mRNA transcript quantification in archival samples using multiplexed, colorcoded probes. BMC Biotechnol, 2011, 11:46., DOI 10.1186/1472-6750-11-46 Du H, Pang M, Hou X, Yuan S, Sun L. PLOD2 in cancer research. Biomed Pharmacother, 2017, 90:670–6., DOI 10.1016/j.biopha.2017.04.023 Escoll M, Gargini R, Cuadrado A, Anton IM, Wandosell F. Mutant p53 oncogenic functions in cancer stem cells are regulated by WIP through YAP/ TAZ. Oncogene, 2017, 36(25):3515–27., DOI 10.1038/onc.2016.518 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 585192 EP 585205 DI 10.3389/pore.2021.585192 PG 14 ER PT J AU Wang, X Liu, Sh Cao, H Li, X Rong, Y Liu, G Du, H Shen, H AF Wang, Xiaopai Liu, Shousheng Cao, Huijiao Li, Xiubo Rong, Yuming Liu, Guorong Du, Hong Shen, Hong TI Increasing Embryonic Morphogen Nodal Expression Suggests Malignant Transformation in Colorectal Lesions and as a Potential Marker for CMS4 Subtype of Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Nodal; TGF-β; colorectal cancer; CMS-4; tumor-stroma percentage ID Nodal; TGF-β; colorectal cancer; CMS-4; tumor-stroma percentage AB Nodal, an embryonic morphogen in TGF-β family, is related with tumorigenicity and progression in various tumors including colorectal cancer (CRC). However, the difference of Nodal expression between CRC and colorectal polyps has not yet been investigated. Besides, whether Nodal can be used as a marker for consensus molecular subtype classification-4 (CMS4) of CRC is also worth studying. We analyzed Nodal expression in patients of CRC (161), high-grade intraepithelial neoplasia (HGIN, 28) and five types of colorectal polyps (116). The Nodal expression difference among groups and the association between Nodal expression and clinicopathological features were analyzed. Two categories logistic regression model was used to predict the odds ratio (OR) of risk factors for high tumor-stroma percentage (TSP), and ROC curve was used to assess the diagnostic value of Nodal in predicting high TSP in CRC. We found that Nodal expression was significantly elevated in CRC and HGIN (p < 0.0001). The increased expression of Nodal was related with high TSP, mismatch repair-proficient (pMMR) status, lymph node metastasis and advanced AJCC stage (p < 0.05). Besides, Nodal expression was the only risk factor for high TSP (OR=6.94; p < 0.001), and ROC curve demonstrated that Nodal expression was able to efficiently distinguish high and low TSP. In conclusion, different expression of Nodal between CRC/HGIN and benign lesions is suggestive of a promoting role for Nodal in colorectal tumor progression. Besides, Nodal might also be used as a potential marker for CMS4 subtype of CRC. C1 [Wang, Xiaopai] Southern Medical University, College of Basic Medicine, Department of PathologyGuangzhou, China. [Liu, Shousheng] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of General MedicineGuangzhou, China. [Cao, Huijiao] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of General MedicineGuangzhou, China. [Li, Xiubo] South China University of Technology, School of Medicine, Guangzhou First People’s Hospital, Department of PathologyGuangzhou, China. [Rong, Yuming] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of General MedicineGuangzhou, China. [Liu, Guorong] South China University of Technology, School of Medicine, Guangzhou First People’s Hospital, Department of PathologyGuangzhou, China. [Du, Hong] South China University of Technology, School of Medicine, Guangzhou First People’s Hospital, Department of PathologyGuangzhou, China. [Shen, Hong] Southern Medical University, College of Basic Medicine, Department of PathologyGuangzhou, China. RP Shen, H (reprint author), Southern Medical University, College of Basic Medicine, Department of Pathology, Guangzhou, China. EM shenhong2010168@163.com CR Vogelstein, B, Papadopoulos, N, Velculescu, VE, Zhou, S, Diaz, LJ, and Kinzler, KW. Cancer genome landscapes. Science, 2013, 339:1546–58., DOI 10.1126/ science.1235122 Vogelstein, B, Fearon, ER, Hamilton, SR, Kern, SE, Preisinger, AC, Leppert, M, et al. Genetic alterations during colorectal-tumor development. N Engl J Med, 1988, 319:525–32., DOI 10.1056/NEJM198809013190901 Topczewska, JM, Postovit, LM, Margaryan, NV, Sam, A, Hess, AR, Wheaton, WW, et al. Embryonic and tumorigenic pathways converge via Nodal signaling: role in melanoma aggressiveness. Nat Med, 2006, 12:925–32., DOI 10.1038/ nm1448 Postovit, LM, Margaryan, NV, Seftor, EA, Kirschmann, DA, Lipavsky, A, Wheaton, WW, et al. Human embryonic stem cell microenvironment suppresses the tumorigenic phenotype of aggressive cancer cells. Proc Natl Acad Sci USA, 2008, 105:4329–34., DOI 10.1073/pnas.0800467105 Lawrence, MG, Margaryan, NV, Loessner, D, Collins, A, Kerr, KM, Turner, M, et al. Reactivation of embryonic nodal signaling is associated with tumor progression and promotes the growth of prostate cancer cells. Prostate, 2011, 71:1198–209., DOI 10.1002/pros.21335 Strizzi, L, Hardy, KM, Margaryan, NV, Hillman, DW, Seftor, EA, Chen, B, et al. Potential for the embryonic morphogen Nodal as a prognostic and predictive biomarker in breast cancer. Breast Cancer Res, 2012, 14:R75., DOI 10.1186/ bcr3185 Strizzi, L, Postovit, LM, Margaryan, NV, Lipavsky, A, Gadiot, J, Blank, C, et al. Nodal as a biomarker for melanoma progression and a new therapeutic target for clinical intervention. Expert Rev Dermatol, 2009, 4:67–78., DOI 10.1586/ 17469872.4.1.67 Gong, Y, Guo, Y, Hai, Y, Yang, H, Liu, Y, Yang, S, et al. Nodal promotes the selfrenewal of human colon cancer stem cells via an autocrine manner through Smad2/3 signaling pathway. Biomed Res Int, 2014, 2014:364134., DOI 10.1155/ 2014/364134 Li, H, Jiang, Y, Pei, F, Li, L, Yan, B, Geng, X, et al. Aldehyde Dehydragenase 1 and nodal as significant prognostic markers in colorectal cancer. Pathol Oncol Res, 2016, 22:121–7., DOI 10.1007/s12253-015-9984-x Guinney, J, Dienstmann, R, Wang, X, de Reynies, A, Schlicker, A, Soneson, C, et al. The consensus molecular subtypes of colorectal cancer. Nat Med, 2015, 21:1350–6., DOI 10.1038/nm.3967 Komor, MA, Bosch, LJ, Bounova, G, Bolijn, AS, Delis-van, DP, Rausch, C, et al. Consensus molecular subtype classification of colorectal adenomas. J Pathol, 2018, 246:266–76., DOI 10.1002/path.5129 Inamura, K. Colorectal cancers: an update on their molecular pathology. Cancers, 2018, 10:26., DOI 10.3390/cancers10010026 Roepman, P, Schlicker, A, Tabernero, J, Majewski, I, Tian, S, Moreno, V., et al. Colorectal cancer intrinsic subtypes predict chemotherapy benefit, deficient mismatch repair and epithelial-to-mesenchymal transition. Int J Cancer, 2014, 134:552–62., DOI 10.1002/ijc.28387 Roseweir, AK, McMillan, DC, Horgan, PG, Edwards, J. Colorectal cancer subtypes: translation to routine clinical pathology. Cancer Treat Rev, 2017, 57: 1–7., DOI 10.1016/j.ctrv.2017.04.006 Yu, L, Harms, PW, Pouryazdanparast, P, Kim, DS, Ma, L, Fullen, DR. Expression of the embryonic morphogen Nodal in cutaneous melanocytic lesions. Mod Pathol, 2010, 23:1209–14., DOI 10.1038/ modpathol.2010.101 Chen, MH, Yip, GW, Tse, GM, Moriya, T, Lui, PC, Zin, ML, et al. Expression of basal keratins and vimentin in breast cancers of young women correlates with adverse pathologic parameters. Mod Pathol, 2008, 21:1183–91., DOI 10.1038/ modpathol.2008.90 Mesker, WE, Junggeburt, JM, Szuhai, K, de Heer, P, Morreau, H, Tanke, HJ, et al. The carcinoma-stromal ratio of colon carcinoma is an independent factor for survival compared to lymph node status and tumor stage. Cell Oncol, 2007, 29:387–98., DOI 10.1155/2007/175276 Park, JH, Richards, CH, McMillan, DC, Horgan, PG, Roxburgh, CS. The relationship between tumour stroma percentage, the tumour microenvironment and survival in patients with primary operable colorectal cancer. Ann Oncol, 2014, 25:644–51., DOI 10.1093/annonc/mdt593 Quail,DF, Zhang, G, Findlay, SD,Hess,DA, Postovit, LM. Nodal promotes invasive phenotypes via a mitogen-activated protein kinase-dependent pathway. Oncogene, 2014, 33:461–73., DOI 10.1038/onc.2012.608 Quail, DF, Zhang, G, Walsh, LA, Siegers, GM, Dieters-Castator, DZ, Findlay, SD, et al. Embryonic morphogen nodal promotes breast cancer growth and progression. PLoS One, 2012, 7:e48237., DOI 10.1371/journal. pone.0048237 Quail, DF, Walsh, LA, Zhang, G, Findlay, SD, Moreno, J, Fung, L, et al. Embryonic protein nodal promotes breast cancer vascularization. Cancer Res, 2012, 72:3851–63., DOI 10.1158/0008-5472.CAN-11-3951 Kirsammer, G, Strizzi, L, Margaryan, NV, Gilgur, A,Hyser, M, Atkinson, J, et al. Nodal signaling promotes a tumorigenic phenotype in human breast cancer. Semin Cancer Biol, 2014, 29:40–50., DOI 10.1016/j.semcancer.2014.07.007 Li, Z, Zhang, J, Zhou, J, Lu, L, Wang, H, Zhang, G, et al. Nodal facilitates differentiation of fibroblasts to cancer-associated fibroblasts that support tumor growth in melanoma and colorectal cancer. Cells, 2019, 8:538., DOI 10.3390/cells8060538 Ubink, I, van Eden, WJ, Snaebjornsson, P, Kok, N, van Kuik, J, van Grevenstein, W, et al. Histopathological and molecular classification of colorectal cancer and corresponding peritoneal metastases. Br J Surg, 2018, 105:e204–11., DOI 10.1002/bjs.10788 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 587029 EP 587035 DI 10.3389/pore.2021.587029 PG 7 ER PT J AU Qian, C Yang, Y Lan, T Wang, Y Yao, J AF Qian, Cuijuan Yang, Yisheng Lan, Tianchen Wang, Yichao Yao, Jun TI Hsa_circ_0043265 Restrains Cell Proliferation, Migration and Invasion of Tongue Squamous Cell Carcinoma via Targeting the miR-1243/SALL1 Axis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE circ_0043265; miR-1243; SALL1; circRNAs; tongue squamous cell carcinoma ID circ_0043265; miR-1243; SALL1; circRNAs; tongue squamous cell carcinoma AB Increasing evidence has displayed critical roles of circular RNAs (circRNAs) in tongue squamous cell carcinoma (TSCC). Hsa_circ_0043265 (circ_0043265) has been identified as a tumor suppressor in various tumors. Nevertheless, the critical roles of circ_0043265 in the initiation and progression of TSCC are yet to be fully elucidated. In our study, RNA and protein expressions were detected via qRT-PCR and Western blot. Cell proliferation, migration and invasion were evaluated via CCK-8 and transwell assays. The interactions between circ_0043265, miR-1243 and SALL1 were analyzed via bioinformatics analyses, RNA pull-down and luciferase assays, respectively. The current study demonstrated that circ_0043265 expression was downmodulated in TSCC tissues and cell lines (SCC25, SCC15, SCC9 and Cal27). Functionally, circ_0043265 overexpression led to an attenuation of cell proliferation, migration and invasion of SCC25 and Cal27 cells. Mechanistically, circ_0043265 acted as a competing endogenous RNA (ceRNA) via competitively sponging miR-1243, and restoration of miR-1243 rescued the inhibitory effects of circ_0043265 on cell proliferation, migration and invasion of SCC25 and Cal27 cells. Finally, it was observed that spalt like transcription factor 1 (SALL1), a potential target of miR-1243, was positively modulated via circ_0043265 in SCC25 and Cal27 cells, and SALL1 knockdown reversed the inhibitory effects of circ_0043265 on SCC25 and Cal27 cells. Collectively, the current study demonstrated that circ_0043265 was downmodulated in TSCC and was identified as a ceRNA that restrained the cell proliferation, migration and invasion of SCC25 and Cal27 cells via modulating the miR-1243/SALL1 axis. C1 [Qian, Cuijuan] Taizhou University, School of Medicine, Institute of TumorTaizhou, China. [Yang, Yisheng] Taizhou University, School of Medicine, Institute of TumorTaizhou, China. [Lan, Tianchen] Taizhou University, School of Medicine, Institute of TumorTaizhou, China. [Wang, Yichao] Taizhou Municipal Hospital, Department of Clinical LaboratoryTaizhou, China. [Yao, Jun] Taizhou University, School of Medicine, Institute of TumorTaizhou, China. RP Yao, J (reprint author), Taizhou University, School of Medicine, Institute of Tumor, Taizhou, China. EM yaojuntzu@yeah.net CR Ng, J.H., Iyer, N. G., Tan, M. H., and Edgren, G., 2017). Changing epidemiology of oral squamous cell carcinoma of the tongue: a global study. Head Neck. 39, 297–304., DOI 10.1002/hed.24589 Kim, Y. J., and Kim, J. H., 2020). Increasing incidence and improving survival of oral tongue squamous cell carcinoma. Sci. Rep. 10, 7877., DOI 10.1038/s41598-020-64748-0 Almangush, A., Heikkinen, I.,Makitie, A. A., Coletta, R. D., Laara, E., Leivo, I., et al., 2017). Prognostic biomarkers for oral tongue squamous cell carcinoma: a systematic review and meta-analysis. Br. J. Canc. 117, 856–866., DOI 10.1038/bjc.2017.244 Ebermann, C., Schnarr, T., and Muller, S., 2020). Recent advances in understanding circular RNAs. F1000Res. 9, F1000., DOI 10.12688/f1000research.25060.1 Lin, Z., Long, F., Zhao, M., Zhang, X., and Yang, M., 2020). The role of circular RNAs in hematological malignancies. Genomics. 112, 30385–30392., DOI 10. 1016/j.ygeno.2020.06.051 Liu, J., Zhang, X., Yan, M., and Li, H., 2020). Emerging role of circular RNAs in cancer. Front Oncol. 10, 663., DOI 10.3389/fonc.2020.00663 Hu, Y. T., Li, X. X., and Zeng, L. W., 2019). Circ_0001742 promotes tongue squamous cell carcinoma progression via miR-431-5p/ATF3 axis. Eur. Rev. Med. Pharmacol. Sci. 23, 10300–10312., DOI 10.26355/eurrev_201912_19668 Qiu, X., Ke, X., Ma, H., Han, L., Chen, Q., Zhang, S., et al., 2019). Profiling and bioinformatics analyses reveal differential expression of circular RNA in tongue cancer revealed by high-throughput sequencing. J. Cell. Biochem. 120, 4102–4112., DOI 10.1002/jcb.27695 Shao, B., and He, L., 2019). Hsa_circ_0001742 promotes tongue squamous cell carcinoma progression via modulating miR-634 expression. Biochem. Biophys. Res. Commun. 513, 135–140., DOI 10.1016/j.bbrc.2019.03.122 Wei, T., Ye, P., Yu, G. Y., and Zhang, Z. Y., 2020). Circular RNA expression profiling identifies specific circular RNAs in tongue squamous cell carcinoma. Mol. Med. Rep. 21:1727–1738., DOI 10.3892/mmr.2020.10980 Yao, Y., Bi, L., and Zhang, C., 2020). Circular RNA_0001742 has potential to predict advanced tumor stage and poor survival profiles in tongue squamous cell carcinoma management. J. Clin. Lab. Anal. 34 e23330., DOI 10.1002/jcla.23330 Ren, T., Liu, C., Hou, J., and Shan, F., 2020). Hsa_circ_0043265 suppresses proliferation, metastasis, EMT and promotes apoptosis in non-small cell lung cancer through miR-25-3p/FOXP2 pathway. OncoTargets Ther. 13, 3867–3880., DOI 10.2147/OTT.S235231 Ma, C., Wang, F., Han, B., Zhong, X., Si, F., Ye, J., et al., 2018). SALL1 functions as a tumor suppressor in breast cancer by regulating cancer cell senescence and metastasis through the NuRD complex. Mol. Canc. 17, 78., DOI 10.1186/s12943-018-0824-y Mu, X., Mou, K. H., Ge, R., Han, D., Zhou, Y., andWang, L. J., 2019). Linc00961 inhibits the proliferation and invasion of skin melanoma by targeting the miR‑367/PTEN axis. Int. J. Oncol. 55, 708–720., DOI 10.3892/ijo.2019.4848 Chen, T.,Yang, Z., Liu,C., Wang, L., Yang, J., Chen, L., et al., 2019). Circ_0078767 suppresses non-small-cell lung cancer by protecting RASSF1A expression via sponging miR-330-3p. Cell Prolif. 52, e12548., DOI 10.1111/cpr.12548 Zhang,X. Z., Liu,H., andChen, S. R., 2020).Mechanisms of long non-CodingRNAs in cancers and their dynamic regulations. Cancers 12, 1245., DOI 10.3390/cancers12051245 Weng, W., Zhang, Z., Huang, W., Xu, X., Wu, B., Ye, T., et al., 2020). Identification of a competing endogenous RNA network associated with prognosis of pancreatic adenocarcinoma. Canc. Cell Int. 20, 231., DOI 10. 1186/s12935-020-01243-6 Zhang, S., Ma, H., Zhang, D., Xie, S., Wang, W., Li, Q., et al., 2018). LncRNA KCNQ1OT1 regulates proliferation and cisplatin resistance in tongue cancer via miR-211-5p mediated Ezrin/Fak/Src signaling. Cell Death Dis. 9, 742., DOI 10.1038/s41419-018-0793-5 He, S., Wang, X., Zhang, J., Zhou, F., Li, L., and Han, X., 2020). TRG-AS1 is a potent driver of oncogenicity of tongue squamous cell carcinoma through microRNA-543/Yes-associated protein 1 axis regulation. Cell Cycle. 19, 1969–1982., DOI 10.1080/15384101.2020.1786622 Liang, Z. Z., Guo, C., Zou, M. M.,Meng, P., and Zhang, T. T., 2020). circRNAmiRNA- mRNA regulatory network in human lung cancer: an update. Canc. Cell Int. 20, 173., DOI 10.1186/s12935-020-01245-4 Ren, S., Lin, P., Wang, J., Yu, H., Lv, T., Sun, L., et al., 2020). Circular RNAs: promising molecular biomarkers of human aging-related diseases via functioning as an miRNA sponge. Mol. Ther. Methods Clin. Dev. 18, 215–229., DOI 10.1016/j.omtm.2020.05.027 Chen, X., Jiang, J., Zhao, Y., Wang, X., Zhang, C., Zhuan, L., et al., 2020). Circular RNA circNTRK2 facilitates the progression of esophageal squamous cell carcinoma through up-regulating NRIP1 expression via miR-140-3p. J. Exp. Clin. Canc. Res. 39, 133., DOI 10.1186/s13046-020-01640-9 Pei, X., Zhang, Y.,Wang, X., Xue, B., Sun, M., and Li, H., 2020). Circular RNA circ-ZEB1 acts as an oncogene in triple negative breast cancer via sponging miR-448. Int. J. Biochem. Cell Biol. 126, 105798., DOI 10.1016/j.biocel.2020. 105798 Zhao, F., Lv, J., Gan, H., Li, Y., Wang, R., Zhang, H., et al., 2015). MiRNA profile of osteosarcoma with CD117 and stro-1 expression: miR-1247 functions as an onco-miRNA by targeting MAP3K9. Int. J. Clin. Exp. Pathol. 8, 1451–1458. Hiramoto, H., Muramatsu, T., Ichikawa, D., Tanimoto, K., Yasukawa, S., Otsuji, E., et al., 2017). miR-509-5p and miR-1243 increase the sensitivity to gemcitabine by inhibiting epithelial-mesenchymal transition in pancreatic cancer. Sci. Rep. 7, 4002., DOI 10.1038/s41598-017-04191-w Ali Syeda, Z., Langden, S. S. S., Munkhzul, C., Lee, M., and Song, S. J., 2020). Regulatory mechanism of MicroRNA expression in cancer. Int. J. Mol. Sci. 21, 1723., DOI 10.3390/ijms21051723 Chi, D., Zhang, W., Jia, Y., Cong, D., and Hu, S., 2019). Spalt-like transcription factor 1, SALL1, gene expression inhibits cell proliferation and cell migration of human glioma cells through the Wnt/β-Catenin signaling pathway. Med. Sci. Monit. Basic Res. 25, 128–138., DOI 10.12659/MSMBR.915067 Li, Z., Zhao, S., Wang, H., Zhang, B., and Zhang, P., 2019). miR-4286 promotes prostate cancer progression via targeting the expression of SALL1. J. Gene Med. e3127, 2019 [Epub ahead of print]., DOI 10.1002/jgm.3127 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 587130 EP 587141 DI 10.3389/pore.2021.587130 PG 12 ER PT J AU Zhang, P Feng, J Wu, X Chu, W Zhang, Y Li, P AF Zhang, Peng Feng, Jing Wu, Xue Chu, Weike Zhang, Yilian Li, Ping TI Bioinformatics Analysis of Candidate Genes and Pathways Related to Hepatocellular Carcinoma in China: A Study Based on Public Databases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE China; hepatocellular carcinoma; differentially expressed genes; hub genes; cell cycle; bioinformatics analysis ID China; hepatocellular carcinoma; differentially expressed genes; hub genes; cell cycle; bioinformatics analysis AB Background and Objective: Hepatocellular carcinoma (HCC) is a highly aggressive malignant tumor of the digestive system worldwide. Chronic hepatitis B virus (HBV) infection and aflatoxin exposure are predominant causes of HCC in China, whereas hepatitis C virus (HCV) infection and alcohol intake are likely the main risk factors in other countries. It is an unmet need to recognize the underlying molecular mechanisms of HCC in China. Methods: In this study, microarray datasets (GSE84005, GSE84402, GSE101685, and GSE115018) derived from Gene Expression Omnibus (GEO) database were analyzed to obtain the common differentially expressed genes (DEGs) by R software. Moreover, the gene ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed by using Database for Annotation, Visualization and Integrated Discovery (DAVID). Furthermore, the protein-protein interaction (PPI) network was constructed, and hub genes were identified by the Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape, respectively. The hub genes were verified using Gene Expression Profiling Interactive Analysis (GEPIA), UALCAN, and Kaplan-Meier Plotter online databases were performed on the TCGA HCC dataset. Moreover, the Human Protein Atlas (HPA) database was used to verify candidate genes’ protein expression levels. Results: A total of 293 common DEGs were screened, including 103 up-regulated genes and 190 down-regulated genes. Moreover, GO analysis implied that common DEGs were mainly involved in the oxidation-reduction process, cytosol, and protein binding. KEGG pathway enrichment analysis presented that common DEGs were mainly enriched in metabolic pathways, complement and coagulation cascades, cell cycle, p53 signaling pathway, and tryptophan metabolism. In the PPI network, three subnetworks with high scores were detected using the Molecular Complex Detection (MCODE) plugin. The top 10 hub genes identified were CDK1, CCNB1, AURKA, CCNA2, KIF11, BUB1B, TOP2A, TPX2, HMMR and CDC45. The other public databases confirmed that high expression of the aforementioned genes related to poor overall survival among patients with HCC. Conclusion: This study primarily identified candidate genes and pathways involved in the underlying mechanisms of Chinese HCC, which is supposed to provide new targets for the diagnosis and treatment of HCC in China. C1 [Zhang, Peng] Tianjin Medical University, School of GraduatesTianjin, China. [Feng, Jing] Tianjin Medical University, School of GraduatesTianjin, China. [Wu, Xue] Tianjin Medical University, School of GraduatesTianjin, China. [Chu, Weike] Tianjin Medical University, School of GraduatesTianjin, China. [Zhang, Yilian] Tianjin Medical University, School of GraduatesTianjin, China. [Li, Ping] Tianjin Second People’s Hospital, Department of HepatologyTianjin, China. RP Li, P (reprint author), Tianjin Second People’s Hospital, Department of Hepatology, Tianjin, China. EM tjlplxg@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018). 68(6):394–424., DOI 10. 3322/caac.21492 Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, et al. Hepatocellular carcinoma. Nat Rev Dis Primers, 2016). 2:16018., DOI 10.1038/ nrdp.2016.18 Zhang BH, Yang BH, Tang ZY. Randomized controlled trial of screening for hepatocellular carcinoma. J Cancer Res Clin Oncol, 2004). 130(7):417–422., DOI 10.1007/s00432-004-0552-0 Guo X, Wu J, Wei F, Ouyang Y, Li Q, Liu K, et al. Trends in hepatitis B virus resistance to nucleoside/nucleotide analogues in North China from 2009-2016: a retrospective study. Int J Antimicrob Agents, 2018). 52(2):201–209., DOI 10. 1016/j.ijantimicag.2018.04.002 Estes C, Anstee QM, Arias-Loste MT, Bantel H, Bellentani S, Caballeria J, et al. Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016-2030. J Hepatol, 2018). 69(4):896–904., DOI 10.1016/j.jhep.2018. 05.036 Gershon D. DNA microarrays: more than gene expression. Nature, 2005). 437(7062):1195–1198., DOI 10.1038/4371195a Rung J, Brazma A, Reuse of public genome-wide gene expression data. Nat Rev Genet, 2013). 14(2):89–99., DOI 10.1038/nrg3394 Qin G, Tu X, Li H, Cao P, Chen X, Song J, et al. Long Noncoding RNA p53- stabilizing and activating RNA promotes p53 signaling by inhibiting heterogeneous nuclear ribonucleoprotein K deSUMOylation and suppresses hepatocellular carcinoma. Hepatology, 2020). 71(1):112–129., DOI 10.1002/hep.30793 Wang H, Huo X, Yang XR, He J, Cheng L, Wang N, et al. STAT3-mediated upregulation of lncRNA HOXD-AS1 as a ceRNA facilitates liver cancer metastasis by regulating SOX4. Mol Cancer, 2017). 16(1):136., DOI 10.1186/ s12943-017-0680-1 Shi J, Ye G, Zhao G, Wang X, Ye C, Thammavong K, et al. Coordinative control of G2/M phase of the cell cycle by non-coding RNAs in hepatocellular carcinoma. PeerJ, 2018). 6, e5787., DOI 10.7717/peerj.5787 Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc, 2009). 4(1):44–57., DOI 10.1038/nprot.2008.211 Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res, 2019). 47(D1):D607–D613., DOI 10.1093/nar/gky1131 Shannon P,Markiel A, OzierO, BaligaNS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res, 2003). 13(11):2498–2504., DOI 10.1101/gr.1239303 Bader GD, Hogue CW. An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics, 2003). 4:2., DOI 10.1186/1471-2105-4-2 Chin CH, Chen SH, Wu HH, Ho CW, Ko MT, Lin CY. CytoHubba: identifying hub objects and sub-networks from complex interactome. BMC Syst Biol, 2014). 8 Suppl 4(Suppl 4):S11., DOI 10.1186/1752-0509-8-s4-s11 Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res, 2017). 45(W1):W98–W102., DOI 10.1093/nar/gkx247 Uhlen M, Zhang C, Lee S, Sjostedt E, Fagerberg L, Bidkhori G, et al. A pathology atlas of the human cancer transcriptome. Science, 2017). 357(6352): eaan2507., DOI 10.1126/science.aan2507 Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce- Rodriguez I, Chakravarthi BVSK, et al. UALCAN: a Portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia, 2017). 19(8):649–658., DOI 10.1016/j.neo.2017.05.002 Nagy A, Lanczky A, Menyhart O, Gy}orffy B. Validation of miRNA prognostic power in hepatocellular carcinoma using expression data of independent datasets. Sci Rep, 2018). 8(1):9227., DOI 10.1038/s41598-018-27521-y Ding Z, Ericksen RE, Escande-Beillard N, Lee QY, Loh A, Denil S, et al. Metabolic pathway analyses identify proline biosynthesis pathway as a promoter of liver tumorigenesis. J Hepatol, 2020). 72(4):725–735., DOI 10. 1016/j.jhep.2019.10.026 Gao Q, Zhu H, Dong L, Shi W, Chen R, Song Z, et al. Integrated proteogenomic characterization of HBV-related hepatocellular carcinoma. Cell, 2019). 179(2): 561–577., DOI 10.1016/j.cell.2019.08.052 Meng Z, Wu J, Liu X, Zhou W, Ni M, Liu S, et al. Identification of potential hub genes associated with the pathogenesis and prognosis of hepatocellular carcinoma via integrated bioinformatics analysis. J Int Med Res, 2020). 48(7):300060520910019., DOI 10.1177/0300060520910019 Ji Y, Yin Y, ZhangW. Integrated bioinformatic analysis identifies networks and promising biomarkers for hepatitis B virus-related hepatocellular carcinoma. Int J Genomics, 2020). 2020:2061024., DOI 10.1155/2020/2061024 Zhang L, Huang Y, Ling J, Zhuo W, Yu Z, Shao M, et al. Screening and function analysis of hub genes and pathways in hepatocellular carcinoma via bioinformatics approaches. Cancer Biomark, 2018). 22(3):511–521., DOI 10.3233/cbm-171160 Roskoski R, Jr. Cyclin-dependent protein kinase inhibitors including palbociclib as anticancer drugs. Pharmacol Res, 2016). 107:249–275., DOI 10. 1016/j.phrs.2016.03.012 Zhang X,Wang L, Yan Y Identification of potential key genes and pathways in hepatitis B virus-associated hepatocellular carcinoma by bioinformatics analyses. Oncol Lett, 2020). 19(5):3477–3486., DOI 10.3892/ol.2020.11470 Sabbaghi M, Gil-Gomez G, Guardia C, Servitja S, Arpi O, Garcia-Alonso S, et al. Defective cyclin B1 induction in Trastuzumab-emtansine, T-DM1, acquired resistance in HER2-positive breast cancer. Clin Cancer Res, 2017). 23(22):7006–7019., DOI 10.1158/1078-0432.Ccr-17-0696 Ding L, Yang L, He Y, Zhu B, Ren F, Fan X, et al. CREPT/RPRD1B associates with aurora B to regulate cyclin B1 expression for accelerating the G2/M transition in gastric cancer. Cell Death Dis, 2018). 9(12):1172., DOI 10.1038/ s41419-018-1211-8 Wu CX, Wang XQ, Chok SH, Man K, Tsang SHY, Chan ACY, et al. Blocking CDK1/PDK1/beta-Catenin signaling by CDK1 inhibitor RO3306 increased the efficacy of sorafenib treatment by targeting cancer stem cells in a preclinical model of hepatocellular carcinoma. Theranostics, 2018). 8(14):3737–3750., DOI 10.7150/thno.25487 Li L., Huang K., Zhao H., Chen B., Ye Q., Yue J. CDK1-PLK1/SGOL2/ANLN pathway mediating abnormal cell division in cell cycle may be a critical process in hepatocellular carcinoma. Cell Cycle, 2020). 19(10):1236–1252., DOI 10.1080/ 15384101.2020.1749471 ChengP, Li Y, Yang L,WenY, ShiW,MaoY, et al. HepatitisBvirusXprotein(HBx, induces G2/M arrest and apoptosis through sustained activation of cyclin B1-CDK1 kinase. Oncol Rep, 2009). 22(5):1101–1107., DOI 10.3892/or_00000542 Dong S., Huang F., Zhang H., Chen Q. Overexpression of BUB1B, CCNA2, CDC20, and CDK1 in tumor tissues predicts poor survival in pancreatic ductal adenocarcinoma. Biosci Rep, 2019). 39(2):BSR20182306., DOI 10.1042/bsr20182306 Teng L, Wang K, Liu Y, Ma Y, Chen W, Bi L. Based on integrated bioinformatics analysis identification of biomarkers in hepatocellular carcinoma patients from different regions. Biomed Res Int, 2019). 2019: 1742341., DOI 10.1155/2019/1742341 Wang J, Chenivesse X, Henglein B, Brechot C. Hepatitis B virus integration in a cyclin A gene in a hepatocellular carcinoma. Nature, 1990). 343(6258): 555–557., DOI 10.1038/343555a0 Tatsuno K, Midorikawa Y, Takayama T, Yamamoto S, Nagae G, MoriyamaM, et al. Impact of AAV2 and hepatitis B virus integration into genome on development of hepatocellular carcinoma in patients with prior hepatitis B virus infection. Clin Cancer Res, 2019). 25(20):6217–6227., DOI 10.1158/1078- 0432.Ccr-18-4041 Bayard Q, Meunier L, Peneau C, Renault V, Shinde J, Nault JC, et al. Cyclin A2/ E1 activation defines a hepatocellular carcinoma subclass with a rearrangement signature of replication stress. Nat Commun, 2018). 9(1): 5235., DOI 10.1038/s41467-018-07552-9 Yang WX, Pan YY, You CG CDK1, CCNB1, CDC20, BUB1, MAD2L1, MCM3, BUB1B, MCM2, and RFC4 may be potential therapeutic targets for hepatocellular carcinoma using integrated bioinformatic analysis. Biomed Res Int, 2019). 2019:1245072., DOI 10.1155/2019/1245072 Broderick R, Nasheuer HP Regulation of Cdc45 in the cell cycle and after DNA damage. Biochem Soc Trans, 2009). 37(Pt 4):926–930., DOI 10.1042/bst0370926 Li JN, Feng CJ, Lu YJ, Li HJ, Tu Z, Liao GQ, et al. mRNA expression of the DNA replication-initiation proteins in epithelial dysplasia and squamous cell carcinoma of the tongue. BMC Cancer, 2008). 8:395., DOI 10.1186/1471-2407-8-395 Xiang XH, Yang L, Zhang X, Ma XH, Miao RC, Gu JX, et al. Seven-senescenceassociated gene signature predicts overall survival for Asian patients with hepatocellular carcinoma. World J Gastroenterol, 2019). 25(14):1715–1728., DOI 10.3748/wjg.v25.i14.1715 Xie W, Wang B, Wang X, Hou D, Su H, Huang H. Nine hub genes related to the prognosis ofHBV-positive hepatocellular carcinoma identified by protein interaction analysis. Ann Transl Med, 2020). 8(7):478., DOI 10.21037/atm.2020.03.94 Yan M, Wang C, He B, Yang M, Tong M, Long Z, et al. Aurora-A kinase: a potent oncogene and target for cancer therapy. Med Res Rev, 2016). 36(6): 1036–1079., DOI 10.1002/med.21399 Chen C, Song G, Xiang J, Zhang H, Zhao S, Zhan Y. AURKA promotes cancer metastasis by regulating epithelial-mesenchymal transition and cancer stem cell properties in hepatocellular carcinoma. Biochem Biophys Res Commun, 2017). 486(2):514–520., DOI 10.1016/j.bbrc.2017.03.075 Shen ZT, Chen Y, Huang GC, Zhu XX, Wang R, Chen LB. Aurora-a confers radioresistance in human hepatocellular carcinoma by activating NF-κB signaling pathway. BMC Cancer, 2019). 19(1):1075., DOI 10.1186/s12885-019-6312-y Asbaghi Y, Thompson LL, Lichtensztejn Z, McManus KJ. KIF11 silencing and inhibition induces chromosome instability that may contribute to cancer. Genes Chromosomes Cancer, 2017). 56(9):668–680., DOI 10.1002/gcc.22471 Chen J, Li S, Zhou S, Cao S, Lou Y, Shen H, et al. Kinesin superfamily protein expression and its association with progression and prognosis in hepatocellular carcinoma. J Cancer Res Ther, 2017). 13(4):651–659., DOI 10.4103/jcrt.JCRT_491_17 Shan S, Chen W, Jia JD. Transcriptome analysis revealed a highly connected gene module associated with cirrhosis to hepatocellular carcinoma development. Front Genet, 2019). 10:305., DOI 10.3389/fgene.2019.00305 Chen Z, Chen J, Huang X, Wu Y, Huang K, Xu W, et al. Identification of potential key genes for hepatitis B virus-associated hepatocellular carcinoma by bioinformatics analysis. J Comput Biol, 2019). 26(5):485–494., DOI 10.1089/ cmb.2018.0244 Neumayer G, Belzil C, Gruss OJ, NguyenMDTPX2: of spindle assembly, DNA damage response, and cancer. Cell Mol Life Sci, 2014). 71(16):3027–3047., DOI 10.1007/s00018-014-1582-7 Yang W, Wan H, Shan R, Wen W, Li J, Luo D, et al. The clinical significance and prognostic value of Xenopus kinesin-like protein 2 expressions in human tumors: a systematic review and meta-analysis. J Cell Physiol, 2019). 234: 14991., DOI 10.1002/jcp.28343 Liu Q, Tu K, Zhang H, Zheng X, Yao Y, Liu Q. TPX2 as a novel prognostic biomarker for hepatocellular carcinoma. Hepatol Res, 2015). 45(8):906–918., DOI 10.1111/hepr.12428 He Z, Mei L, Connell M, Maxwell CA. Hyaluronan mediated motility receptor, HMMR, encodes an evolutionarily conserved homeostasis, mitosis, and meiosis regulator rather than a hyaluronan receptor. Cells, 2020). 9(4):819., DOI 10.3390/cells9040819 Shen S, Kong J, Qiu Y, Yang X, Wang W, Yan L. Identification of core genes and outcomes in hepatocellular carcinoma by bioinformatics analysis. J Cell Biochem, 2019). 120(6):10069–10081., DOI 10.1002/jcb.28290 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 588532 EP 588545 DI 10.3389/pore.2021.588532 PG 14 ER PT J AU Huang, R Severson, E Haberberger, J Duncan, D Hemmerich, A Edgerly, C Ferguson, Ly Frampton, G Owens, C Williams, E Elvin, J Vergilio, JA Killian, KJ Lin, D Morley, S McEwan, D Holmes, O Danziger, N Cohen, M Sathyan, P McGregor, K Reddy, P Venstrom, J Anhorn, R Alexander, B Brown, Ch Ross, J Ramkissoon, Sh AF Huang, S. P. Richard Severson, Eric Haberberger, James Duncan, L. Daniel Hemmerich, Amanda Edgerly, Claire Ferguson, Lynn N. Frampton, Garrett Owens, Clarence Williams, Erik Elvin, Julia Vergilio, Jo-Anne Killian, Keith Jonathan Lin, Douglas Morley, Samantha McEwan, Deborah Holmes, Oliver Danziger, Natalie Cohen, B. Michael Sathyan, Pratheesh McGregor, Kimberly Reddy, Prasanth Venstrom, Jeffrey Anhorn, Rachel Alexander, Brian Brown, Charlotte Ross, S. Jeffrey Ramkissoon, H. Shakti TI Landscape of Biomarkers in Non-small Cell Lung Cancer Using Comprehensive Genomic Profiling and PD-L1 Immunohistochemistry SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarkers; non-small cell lung cancer; comprehensive genomic profiling; PD-L1; immunohistochemistry ID biomarkers; non-small cell lung cancer; comprehensive genomic profiling; PD-L1; immunohistochemistry AB Comprehensive genomic profiling (CGP) and immunohistochemistry (IHC) are important biomarker tools used for patients with non-small cell lung cancer (NSCLC) given the expanding number of standard-of-care therapies that require companion diagnostic testing. We examined 9450 NSCLC real-world patient samples that underwent both CGP and programmed death-ligand 1 (PD-L1) IHC to understand the biomarker landscape in this patient cohort. By assessing National Comprehensive Cancer Network (NCCN)-recommended biomarkers including genomic alterations, tumor mutational burden (≥10 mutations/Mb cut-off), and PD-L1 expression (Tumor Proportion Score (TPS) ≥ 50% cut-off), we show that CGP + PD-L1 IHC yielded potentially actionable results for 70.5% of the 9,450 patients with NSCLC. Among the remaining 29.5% (2,789/9,450) of patients, 86.7% (2,419/2,789) were potentially eligible for another biomarker-associated therapy and/or clinical trial based on their genomic profile. In addition, in the PD-L1TPS≥50% disease subset, BRAF mutations, MET mutations, MET amplifications, and KRAS mutations were significantly enriched; and in the PDL1TPS< 50%, EGFR mutations, ERBB2 mutations, STK11 mutations, and KEAP1 mutations were enriched. These findings highlight the improved clinical utility of combining CGP with IHC to expand the biomarker-guided therapeutic options available for patients with NSCLC, relative to single biomarker testing alone. C1 [Huang, S. P. Richard] Foundation Medicine, Inc.Morrisville, NC, USA. [Severson, Eric] Foundation Medicine, Inc.Morrisville, NC, USA. [Haberberger, James] Foundation Medicine, Inc.Morrisville, NC, USA. [Duncan, L. Daniel] Foundation Medicine, Inc.Morrisville, NC, USA. [Hemmerich, Amanda] Foundation Medicine, Inc.Morrisville, NC, USA. [Edgerly, Claire] Foundation Medicine, Inc.Morrisville, NC, USA. [Ferguson, Lynn N.] Foundation Medicine, Inc.Morrisville, NC, USA. [Frampton, Garrett] Foundation Medicine, Inc.Cambridge, MA, USA. [Owens, Clarence] Foundation Medicine, Inc.Morrisville, NC, USA. [Williams, Erik] Foundation Medicine, Inc.Cambridge, MA, USA. [Elvin, Julia] Foundation Medicine, Inc.Cambridge, MA, USA. [Vergilio, Jo-Anne] Foundation Medicine, Inc.Cambridge, MA, USA. [Killian, Keith Jonathan] Foundation Medicine, Inc.Cambridge, MA, USA. [Lin, Douglas] Foundation Medicine, Inc.Cambridge, MA, USA. [Morley, Samantha] Foundation Medicine, Inc.Cambridge, MA, USA. [McEwan, Deborah] Foundation Medicine, Inc.Cambridge, MA, USA. [Holmes, Oliver] Foundation Medicine, Inc.Cambridge, MA, USA. [Danziger, Natalie] Foundation Medicine, Inc.Cambridge, MA, USA. [Cohen, B. Michael] Wake Forest School of Medicine, Wake Forest Comprehensive Cancer CenterWinston-Salem, NC, USA. [Sathyan, Pratheesh] Foundation Medicine, Inc.Cambridge, MA, USA. [McGregor, Kimberly] Foundation Medicine, Inc.Cambridge, MA, USA. [Reddy, Prasanth] Foundation Medicine, Inc.Cambridge, MA, USA. [Venstrom, Jeffrey] Foundation Medicine, Inc.Cambridge, MA, USA. [Anhorn, Rachel] Foundation Medicine, Inc.Cambridge, MA, USA. [Alexander, Brian] Foundation Medicine, Inc.Cambridge, MA, USA. [Brown, Charlotte] Foundation Medicine, Inc.Morrisville, NC, USA. [Ross, S. Jeffrey] Foundation Medicine, Inc.Morrisville, NC, USA. [Ramkissoon, H. Shakti] Foundation Medicine, Inc.Morrisville, NC, USA. RP Huang, R (reprint author), Foundation Medicine, Inc., Morrisville, USA. EM rhuang@foundationmedicine.com CR Society, AC. Cancer Facts and figures. Atlanta, GA: American Cancer Society Inc., 2019). FaD Administration. List of cleared or approved companion diagnostic devices, in vitro and imaging tools). White Oak, MD: FaD Administration, 2019). NCCN, NCCN Clinical practice guidelines in Oncology, non-small cell lung cancer. Version 5. Plymouth Meeting, PA; NCCN, 2019). Eberhard, DA, Johnson, BE, Amler, LC, Goddard, AD, Heldens, SL, Herbst, RS, et al. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol, 2005, 23(25), 5900–9., DOI 10.1200/JCO.2005.02.857 Canon, J, Rex, K, Saiki, AY, Mohr, C, Cooke, K, Bagal, D, et al. The clinical KRAS(G12C, inhibitor AMG 510 drives anti-tumour immunity. Nature, 2019, 575(7781), 217–23., DOI 10.1038/s41586-019-1694-1 FDA. FDA expands pembrolizumab indication for first-line treatment of NSCLC, TPS ≥ 1%). Silver Spring, MD. FDA, 2019). FDA. FDA grants accelerated approval to pembrolizumab for first tissue/site agnostic indication. Silver Spring, MD. FDA, 2020). FDA. FDA approves pembrolizumab for adults and children with TMB-H solid tumors. Silver Spring, MD. FDA, 2020). Scheerens, H,Malong, A, Bassett, K, Boyd, Z,Gupta,V,Harris, J, et al. Current status of companion and complementary diagnostics: strategic considerations for development and launch. Clin Transl Sci, 2017, 10(2), 84–92., DOI 10.1111/cts.12455 Skoulidis, F, Goldberg, ME, Greenawalt, DM, Hellmann, MD, Awad, MM, Gainor, JF, et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov, 2018, 8(7), 822–35., DOI 10.1158/2159-8290.CD-18-0099 Arbour, KC, Jordan, E, Kim, HR, Dienstag, J, Yu, HA, Sanchez-Vega, F, et al. Effects of co-occurring genomic alterations on outcomes in patients with KRAS-mutant non-small cell lung cancer. Clin Cancer Res, 2018, 24(2), 334–40., DOI 10.1158/1078-0432.ccr-17-1841 Yu, S, Liu, D, Shen, B, Shi, M, and Feng, J. Immunotherapy strategy of EGFR mutant lung cancer. Am J Cancer Res, 2018, 8(10), 2106–15. DAKO.PD-L1 IHC22C3 pharmDx interpretation manual—NSCLC, 2019). Available from: https://www.agilent.com/cs/library/usermanuals/public/29158_pd-l1-ihc-22C3- pharmdx-nsclc-interpretation-manual.pdf, Accessed September 09,, 2019). Frampton, GM, Fichtenholtz, A, Otto, GA,Wang, K, Downing, SR, He, J, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol, 2013, 31, 11), 1023–31., DOI 10.1038/nbt.2696 Chalmers, ZR,Connelly,CF,Fabrizio,D, Gay, L,Ali, SM,Ennis,R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med, 2017, 9, 1), 34., DOI 10.1186/s13073-017-0424-2 Trabucco, SE, Gowen, K, Maund, SL, Sanford, E, Fabrizio, DA, Hall, MJ, et al. A novel next-generation sequencing approach to detecting microsatellite instability and pan-tumor characterization of 1000 microsatellite instabilityhigh cases in 67,000 patient samples. J Mol Diagn, 2019, 21, 6), 1053–66., DOI 10.1016/j.jmoldx.2019.06.011 Goeman, JJ, and Solari, A. Multiple hypothesis testing in genomics. Stat Med., 2014, 33, 11), 1946–78., DOI 10.1002/sim.6082 Liu, C, Zheng, S, Jin, R, Wang, X, Wang, F, Zang, R, et al. The superior efficacy of anti-PD-1/PD-L1 immunotherapy in KRAS-mutant non-small cell lung cancer that correlates with an inflammatory phenotype and increased immunogenicity. Cancer Lett, 2020, 470, 95–105., DOI 10.1016/j.canlet.2019. 10.027 Guisier, F, Dubos-Arvis, C, Vinas, F,Doubre, H, Ricordel, C, Ropert, S, et al. Efficacy and safety of anti-PD-1 immunotherapy in patients with advanced NSCLC with BRAF, HER2, or MET mutations or RET translocation: GFPC 01-2018. J Thorac Oncol, 2020, 15, 4), 628–36., DOI 10.1016/j.jtho.2019. 12.129 Zhang, YL, Yuan, JQ, Wang, KF, Fu, XH, Han, XR, Threapleton, D, et al. The prevalence of EGFR mutation in patients with non-small cell lung cancer: a systematic review and meta-analysis. Oncotarget, 2016, 7, 48), 78985–93., DOI 10.18632/oncotarget.12587 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 592997 EP 593002 DI d10.3389/pore.2021.592997 PG 6 ER PT J AU Cai, Shz Xiong, Qw Zhao, Ln Ji, Yt Luo, Zx Ma, Zr AF Cai, Shi-zhong Xiong, Qian-wei Zhao, Li-na Ji, Yi-ting Luo, Zheng-xiang Ma, Zhou-rui TI β-Elemene Triggers ROS-dependent Apoptosis in Glioblastoma Cells Through Suppressing STAT3 Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE β-elemene; glioblastoma; apoptosis; stat3; ROS ID β-elemene; glioblastoma; apoptosis; stat3; ROS AB Glioblastoma is one of the most aggressive primary brain tumors with few treatment strategies. β-Elemene is a sesquiterpene known to have broad spectrum antitumor activity against various cancers. However, the signaling pathways involved in β-elemene induced apoptosis of glioblastoma cells remains poorly understood. In this study, we reported that β-elemene exhibited antiproliferative activity on U87 and SHG-44 cells, and induced cell death through induction of apoptosis. Incubation of these cells with β-elemene led to the activation of caspase-3 and generation of reactive oxygen species (ROS). Western blot assay showed that β-elemene suppressed phosphorylation of STAT3, and subsequently down-regulated the activation of p-JAK2 and p-Src. Moreover, pre-incubation of cells with ROS inhibitor N-acetyl-L-cysteine (NAC) significantly reversed β-elemene-mediated apoptosis effect and down-regulation of JAK2/Src-STAT3 signaling pathway. Overall, our findings implied that generation of ROS and suppression of STAT3 signaling pathway is critical for the apoptotic activity of β-elemene in glioblastoma cells. C1 [Cai, Shi-zhong] Children’s Hospital of Soochow University, Department of Child and Adolescent HealthcareSuzhou, China. [Xiong, Qian-wei] Children’s Hospital of Soochow University, Department of SurgerySuzhou, China. [Zhao, Li-na] The First Affiliated Hospital of Soochow University, Key Laboratory of Clinical Immunology of Jiangsu ProvinceSuzhou, China. [Ji, Yi-ting] Children’s Hospital of Soochow University, Department of Child and Adolescent HealthcareSuzhou, China. [Luo, Zheng-xiang] Nanjing Brain Hospital Affiliated to Nanjing Medical University, Department of NeurosurgeryNanjing, China. [Ma, Zhou-rui] Children’s Hospital of Soochow University, Department of SurgerySuzhou, China. RP Ma, Zr (reprint author), Children’s Hospital of Soochow University, Department of Surgery, Suzhou, China. EM chngrey@suda.edu.cn CR Preusser M, de Ribaupierre S, Wohrer A, Erridge SC, Hegi M, Weller M, et al. Current concepts and management of glioblastoma. Ann Neurol., 2011). 70(1): 9–21., DOI 10.1002/ana.22425 Hombach-Klonisch S, Mehrpour M, Shojaei S, Harlos C, Pitz M, Hamai A, et al. Glioblastoma and chemoresistance to alkylating agents: involvement of apoptosis, autophagy, and unfolded protein response. Pharmacology Ther, 2018). 184:13–41., DOI 10.1016/j.pharmthera.2017.10.017 Osuka S, Van Meir EG Overcoming therapeutic resistance in glioblastoma: the way forward. J Clin Invest, 2017). 127(2):415–26., DOI 10.1172/jci89587 Geraldo LHM, Garcia C, da Fonseca ACC, Dubois LGF, de Sampaio e Spohr TCL, Matias D, et al. Glioblastoma therapy in the age of molecular medicine. Trends Cancer, 2019). 5(1):46–65., DOI 10.1016/j.trecan.2018.11.002 McGranahan T, Therkelsen KE, Ahmad S, Nagpal S Current state of immunotherapy for treatment of glioblastoma. Curr Treat Options Oncol, 2019). 20(3):24., DOI 10.1007/s11864-019-0619-4 Liu Y, Li C, Lin J STAT3 as a therapeutic target for glioblastoma. Acamc, 2010). 10(7):512–9., DOI 10.2174/187152010793498636 Furtek SL, Backos DS, Matheson CJ, Reigan P Strategies and approaches of targeting STAT3 for cancer treatment. ACS Chem. Biol., 2016). 11(2):308–18., DOI 10.1021/acschembio.5b00945 Kang S-H, Yu MO, Park K-J, Chi S-G, Park D-H, Chung Y-G Activated STAT3 regulates hypoxia-induced angiogenesis and cell migration in human glioblastoma. Neurosurgery, 2010). 67(5):1386–95; discussion 1395., DOI 10. 1227/neu.0b013e3181f1c0cd Jackson C, Ruzevick J, Amin AG, Lim M Potential role for STAT3 inhibitors in glioblastoma. Neurosurg Clin North America, 2012). 23(3):379–89., DOI 10.1016/ j.nec.2012.04.002 Zhai B, Zhang N, Han X, Li Q, Zhang M, Chen X, et al. Molecular targets of β-elemene, a herbal extract used in traditional Chinese medicine, and its potential role in cancer therapy: a review. Biomed Pharmacother, 2019). 114: 108812., DOI 10.1016/j.biopha.2019.108812 Qureshi MZ, Attar R, Romero MA, Sabitaliyevich UY, Nurmurzayevich SB, Ozturk O, et al. Regulation of signaling pathways by β-elemene in cancer progression and metastasis. J Cel Biochem, 2019). 120(8):12091–100., DOI 10. 1002/jcb.28624 Guan C, Liu W, Yue Y, Jin H, Wang X, Wang XJ Inhibitory effect of β-elemene on human breast cancer cells. Int J Clin Exp Pathol, 2014). 7(7):3948–56. Yu X, Xu M, Li N, Li Z, Li H, Shao S, et al. β-elemene inhibits tumor-promoting effect of M2 macrophages in lung cancer. Biochem Biophysical Res Commun, 2017). 490(2):514–20., DOI 10.1016/j.bbrc.2017.06.071 Wu XS, Xie T, Lin J, Fan HZ, Huang-Fu HJ, Ni LF, et al. An investigation of the ability of elemene to pass through the blood-brain barrier and its effect on brain carcinomas. J Pharm Pharmacol, 2009). 61(12):1653–6., DOI 10.1211/jpp/ 61.12.0010 Chen J, Wang T, Xu S, Lin A, Yao H, Xie W, et al. Novel hybrids of natural β -elemene bearing isopropanolamine moieties: synthesis, enhanced anticancer profile, and improved aqueous solubility. Fitoterapia, 2017). 120:117–25., DOI 10.1016/j.fitote.2017.05.002 Boulton S, Anderson A, Swalwell H, Henderson JR,Manning P, Birch-Machin MA Implications of using the fluorescent probes, dihydrorhodamine 123 and 2’,7’-dichlorodihydrofluorecein diacetate, for the detection of UVA-induced reactive oxygen species. Free Radic Res, 2011). 45(2):139–46., DOI 10.3109/ 10715762.2010.517751 SlatteryML, Lundgreen A, Kadlubar SA, Bondurant KL, Wolff RK JAK/STAT/ SOCS-signaling pathway and colon and rectal cancer. Mol. Carcinog., 2013). 52(2):155–66., DOI 10.1002/mc.21841 Mitsuyama K, Matsumoto S, Masuda J, Yamasakii H, Kuwaki K, Takedatsu H, et al. Therapeutic strategies for targeting the IL-6/STAT3 cytokine signaling pathway in inflammatory bowel disease. Anticancer Res, 2007). 27(6A):3749–56. Shabaninejad Z, Pourhanifeh MH, Movahedpour A, Mottaghi R, Nickdasti A, Mortezapour E, et al. Therapeutic potentials of curcumin in the treatment of glioblstoma. Eur J Med Chem, 2020). 188:112040., DOI 10.1016/j.ejmech.2020. 112040 Chen J, Wang T, Xu S, Zhang P, Lin A, Wu L, et al. Discovery of novel antitumor nitric oxide-donating β -elemene hybrids through inhibiting the PI3K/Akt pathway. Eur J Med Chem, 2017). 135:414–23., DOI 10.1016/j.ejmech. 2017.04.045 Tavana E, Mollazadeh H, Mohtashami E, Modaresi SMS, Hosseini A, Sabri H, et al. Quercetin: A promising phytochemical for the treatment of glioblastoma multiforme. Biofactors, 2020). 46:356–66., DOI 10.1002/biof.1605 Liu Y, Fan C, Pu L, Wei C, Jin H, Teng Y, et al. Phloretin induces cell cycle arrest and apoptosis of human glioblastoma cells through the generation of reactive oxygen species. J Neurooncol, 2016). 128(2):217–23., DOI 10.1007/s11060-016-2107-z Zhu Y, Hu J, Shen F, Shen H, Liu W, Zhang J The cytotoxic effect of β-elemene against malignant glioma is enhanced by base-excision repair inhibitor methoxyamine. J Neurooncol, 2013). 113(3):375–84., DOI 10.1007/s11060- 013-1136-0 Li CL, Chang L, Guo L, Zhao D, Liu HB, Wang QS, et al. β-elemene induces caspase-dependent apoptosis in human glioma cells in vitro through the upregulation of Bax and Fas/FasL and downregulation of Bcl-2. Asian Pac J Cancer Prev, 2014). 15(23):10407–12., DOI 10.7314/apjcp.2014.15.23.10407 Zhang H, Xu F, Xie T, Jin H, Shi L β-elemene induces glioma cell apoptosis by downregulating survivin and its interaction with hepatitis B X-interacting protein. Oncol Rep, 2012). 28(6):2083–90., DOI 10.3892/or.2012.2022 Simon H-U, Haj-Yehia A, Levi-Schaffer F Role of reactive oxygen species, ROS, in apoptosis induction. Apoptosis, 2000). 5(5):415–8., DOI 10.1023/a: 1009616228304 Yan X, Liang F, Li D, Zheng J Ouabain elicits human glioblastoma cells apoptosis by generating reactive oxygen species in ERK-p66SHC-dependent pathway. Mol Cel Biochem, 2015). 398(1–2):95–104., DOI 10.1007/s11010-014- 2208-y Balic JJ, Garama DJ, Saad MI, Yu L, West AC, West AJ, et al. Serinephosphorylated STAT3 promotes tumorigenesis via modulation of RNA polymerase transcriptional activity. Cancer Res, 2019). 79(20):5272–87., DOI 10.1158/0008-5472.can-19-0974 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 594299 EP 594306 DI 10.3389/pore.2021.594299 PG 8 ER PT J TI Upregulation of Fatty Acid Transporters is Associated With Tumor Progression in Non-Muscle-Invasive Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bladder cancer; Fatty acid; FATP4; CD36; ACSL1 ID bladder cancer; Fatty acid; FATP4; CD36; ACSL1 AB As patients with non-muscle-invasive bladder cancer (NMIBC) show a high degree of heterogeneity in tumor recurrence or progression, many clinicians demand a detailed risk stratification. Although modified fatty acid metabolism in cancer cells is reported to reflect malignant phenotypes such as metastasis, the impact of fatty acid transporters on NMIBC has never been investigated. This study examined the clinicopathologic implications of fatty acid transporters such as fatty acid transport protein 4 (FATP4), cluster of differentiation 36/fatty acid translocase (CD36/FAT), and long chain acyl CoA synthetase 1 (ACSL1) in 286 NMIBC cases. This study revealed that FATP4, CD36, and ACSL1 were overexpressed in 123 (43.0%), 43 (15.0%), and 35 (12.2%) NMIBC cases, respectively. High FATP4 in tumor cells was associated with high grade (p = 0.004) and high stage (p = 0.039). High CD36 was related to high grade (p < 0.001), high stage (p = 0.002), and non-papillary growth type (p = 0.004). High ACSL1 showed an association with high grade (p < 0.001), high stage (p = 0.01), non-papillary growth type (p = 0.002), and metastasis (p = 0.033). High FATP4 was an independent factor predicting short overall survival (OS) (hazard ratio = 3.32; 95% confidence interval, 1.07–10.31; p = 0.038). In conclusion, upregulation of FATP4, CD36, and ACSL1 might promote the NMIBC progression and could be exploited in clinical risk stratification and targeted therapy. CR Knowles MA Molecular subtypes of bladder cancer: jekyll and Hyde or chalk and cheese?. Carcinogenesis, 2006). 27(3):361–73., DOI 10.1093/carcin/bgi310 Du X, Wang Q-R, Chan E, MerchantM, Liu J, French D, et al. FGFR3 stimulates stearoyl CoA desaturase 1 activity to promote bladder tumor growth. Cancer Res, 2012). 72(22):5843–55., DOI 10.1158/0008-5472.can-12-1329 Chen Y, Li P Fatty acid metabolism and cancer development. Sci Bull, 2016). 61(19):1473–9., DOI 10.1007/s11434-016-1129-4 Fermor BF, Masters JR, Wood CB, Miller J, Apostolov K, Habib NA Fatty acid composition of normal and malignant cells and cytotoxicity of stearic, oleic and sterculic acids in vitro. Eur J Cancer, 1992). 28A(6-7):1143–7., DOI 10.1016/0959- 8049(92)90475-h Miryaghoubzadeh J, Darabi M, Madaen K, Shaaker M, Mehdizadeh A, Hajihosseini R Tissue fatty acid composition in human urothelial carcinoma. Br J Biomed Sci, 2013). 70(1):1–5., DOI 10.1080/09674845.2013.11669921 Sugino T, Baba K, Hoshi N, Aikawa K, Yamaguchi O, Suzuki T Overexpression of fatty acid synthase in human urinary bladder cancer and combined expression of the synthase and Ki-67 as a predictor of prognosis of cancer patients. Med Mol Morphol, 2011). 44(3):146–50., DOI 10.1007/s00795-010- 0517-0 Kuo C-Y, Ann DK When fats commit crimes: fatty acid metabolism, cancer stemness and therapeutic resistance. Cancer Commun, 2018). 38(1):47., DOI 10. 1186/s40880-018-0317-9 Schneider H, Staudacher S, Poppelreuther M, StremmelW, Ehehalt R, Fullekrug J Protein mediated fatty acid uptake: synergy between CD36/FAT-facilitated transport and acyl-CoA synthetase-driven metabolism. Arch Biochem Biophys, 2014). 546:8–18., DOI 10.1016/j.abb.2014.01.025 Li Z, Kang Y Lipid metabolism fuels cancer’s spread. Cel Metab, 2017). 25(2): 228–30., DOI 10.1016/j.cmet.2017.01.016 Pascual G, Avgustinova A, Mejetta S, Martin M, Castellanos A, Attolini CS-O, et al. Targetingmetastasis-initiating cells through the fatty acid receptor CD36. Nature, 2017). 541(7635):41–5., DOI 10.1038/nature20791 Kim YS, Jung J, Jeong H, Lee JH, Oh HE, Lee ES, et al. High membranous expression of fatty acid transport protein 4 is associated with tumorigenesis and tumor progression in clear cell renal cell carcinoma. Dis Markers, 2019). 2019:5702026., DOI 10.1155/2019/5702026 Yen M-C, Chou S-K, Kan J-Y, Kuo P-L, Hou M-F, Hsu Y-L Solute carrier family 27 member 4, SLC27A4, enhances cell growth, migration, and invasion in breast cancer cells. Ijms, 2018). 19(11):3434., DOI 10.3390/ijms19113434 Chen WC, Wang CY, Hung YH, Weng TY, Yen MC, Lai MD Systematic analysis of gene expression alterations and clinical outcomes for long-chain acyl-coenzyme A synthetase family in cancer. PLoS One, 2016). 11(5): e0155660., DOI 10.1371/journal.pone.0155660 Amin MB, Edge SB, Greene FL, Byrd DR, Brookland RK, Washington MK, et al. AJCC cancer staging manual. 8th ed. Springer Nature, 2017). Moch H, Humphrey PA, Ulbright TM, Reuter VE. WHO classification of tumours of the urinary system and male genital organs. 4th ed. Lyon: IARC, 2016). Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, et al. Fatty acid uptake and lipid storage induced by HIF-1α contribute to cell growth and survival after hypoxia-reoxygenation. Cel Rep, 2014). 9(1):349–65., DOI 10. 1016/j.celrep.2014.08.056 Jiang L, Xiao L, Sugiura H, Huang X, Ali A, Kuro-o M, et al. Metabolic reprogramming during TGFβ1-induced epithelial-to-mesenchymal transition. Oncogene, 2015). 34(30):3908–16., DOI 10.1038/onc.2014.321 Bonen A, Chabowski A, Luiken JJFP, Glatz JFC Mechanisms and regulation of protein-mediated cellular fatty acid uptake: molecular, biochemical, and physiological evidence. Physiology, 2007). 22:15–28., DOI 10.1152/ physiologyonline.2007.22.1.15 Kazantzis M, Stahl A Fatty acid transport proteins, implications in physiology and disease. Biochim Biophys Acta, Bba, - Mol Cel Biol Lipids, 2012). 1821(5): 852–7., DOI 10.1016/j.bbalip.2011.09.010 Milger K, Herrmann T, Becker C, Gotthardt D, Zickwolf J, Ehehalt R, et al. Cellular uptake of fatty acids driven by the ER-localized acyl-CoA synthetase FATP4. J Cel Sci, 2006). 119(Pt 22):4678–88., DOI 10.1242/jcs.03280 Stahl A, Hirsch DJ, Gimeno RE, Punreddy S, Ge P, Watson N, et al. Identification of the major intestinal fatty acid transport protein. Mol Cel, 1999). 4(3):299–308., DOI 10.1016/s1097-2765(00)80332-9 Blackburn C, Guan B, Brown J, Cullis C, Condon SM, Jenkins TJ, et al. Identification and characterization of 4-aryl-3,4-dihydropyrimidin-2(1H)- ones as inhibitors of the fatty acid transporter FATP4. Bioorg Med Chem Lett, 2006). 16(13):3504–9., DOI 10.1016/j.bmcl.2006.03.102 Baillie AGS, Coburn CT, Abumrad NA Reversible binding of long-chain fatty acids to purified FAT, the adipose CD36 homolog. J Membr Biol, 1996). 153(1):75–81., DOI 10.1007/s002329900111 Watkins PA, Maiguel D, Jia Z, Pevsner J Evidence for 26 distinct acylcoenzyme A synthetase genes in the human genome. J Lipid Res, 2007). 48(12):2736–50., DOI 10.1194/jlr.m700378-jlr200 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 594705 EP 594710 DI 10.3389/pore.2021.594705 PG 6 ER PT J AU Wang, Y Ji, N Wang, J Cao, J Li, D Zhang, Y Zhang, L AF Wang, Yi Ji, Nan Wang, Junmei Cao, Jingli Li, Deling Zhang, Yang Zhang, Liwei TI SCG3 Protein Expression in Glioma Associates With less Malignancy and Favorable Clinical Outcomes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SCG3; glioma; clinicopathological features; prognosis ID SCG3; glioma; clinicopathological features; prognosis AB Introduction: Secretogranin III (SCG3) physiologically participates in neurotransmitter storage/transport and is widely expressed in neuroendocrine tumors. However, there is no report on SCG3 protein expression in gliomas. Methods: The method of immunohistochemical staining on a glioma tissue microarray was utilized to detect SCG3 protein expression and investigate the correlations of its expression with clinicopathological and genetic features in gliomas. The RNA-seq data of SCG3 in The Cancer Genome Atlas database was exploited to explore these correlations at the transcriptional level. Results: There were 57.5% (130/226) glioma cases having SCG3 cytoplasmic staining in the tissue microarray. SCG3 expression inversely correlated with malignancy grade at both transcriptional and protein levels. The highest level was observed in oligodendroglial tumors, especially in oligodendrogliomas (ODs) with IDH-mutation/1p19q-codeletion. The lowest SCG3 expression was observed in glioblastomas (GBMs), especially in the mesenchymal subtype. Nearly a half of GBM cases (44.4%, 64/144) had any discernible SCG3 staining, and were defined as SCG3-positive by the microarray study. SCG3-positive GBM cases exhibited improved overall survival as compared with the SCG3-negative cases (29.3 vs. 14.5 months; Hazard ratio, 0.364; 95% CI, 0.216–0.612; p < 0.001). A multivariate Cox regression analysis also revealed SCG3 positivity as an independent favorable prognosticator in GBM patients. Conclusion: SCG3 protein expression inversely correlates with glioma malignancy and predicts favorable outcomes in GBM patients. C1 [Wang, Yi] Capital Medical University, Beijing Tiantan Hospital, Department of Neurosurgery / China National Clinical Research Center for Neurological DiseasesBeijing, China. [Ji, Nan] Capital Medical University, Beijing Tiantan Hospital, Department of Neurosurgery / China National Clinical Research Center for Neurological DiseasesBeijing, China. [Wang, Junmei] Capital Medical University, Beijing Neurosurgical Institute, Department of PathologyBeijing, China. [Cao, Jingli] Capital Medical University, Beijing Tiantan Hospital, Beijing Key Laboratory of Translational Medicine for Cerebrovascular DiseaseBeijing, China. [Li, Deling] Capital Medical University, Beijing Tiantan Hospital, Department of Neurosurgery / China National Clinical Research Center for Neurological DiseasesBeijing, China. [Zhang, Yang] Capital Medical University, Beijing Tiantan Hospital, Department of Neurosurgery / China National Clinical Research Center for Neurological DiseasesBeijing, China. [Zhang, Liwei] Capital Medical University, Beijing Tiantan Hospital, Department of Neurosurgery / China National Clinical Research Center for Neurological DiseasesBeijing, China. RP Zhang, Y (reprint author), Capital Medical University, Beijing Tiantan Hospital, Department of Neurosurgery / China National Clinical Research Center for Neurological Diseases, Beijing, China. EM zhangyang8025@163.com CR Reardon, DA, and Wen, PY. Glioma in 2014: unravelling tumour heterogeneityimplications for therapy. Nat Rev Clin Oncol, 2015, 12(2):69–70., DOI 10.1038/ nrclinonc.2014.223 Aubry, M, de Tayrac, M, Etcheverry, A, Clavreul, A, Saikali, S, Menei, P, et al. From the core to beyond the margin: a genomic picture of glioblastoma intratumor heterogeneity. Oncotarget, 2015, 6(14):12094–109., DOI 10.18632/ oncotarget.3297 Yan, H, Parsons, DW, Jin, G, McLendon, R, Rasheed, BA, Yuan, W, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med, 2009, 360(8):765–73., DOI 10. 1056/NEJMoa0808710 Killela, PJ, Pirozzi, CJ, Healy, P, Reitman, ZJ, Lipp, E, Rasheed, BA, et al. Mutations in IDH1, IDH2, and in the TERT promoter define clinically distinct subgroups of adult malignant gliomas. Oncotarget, 2014, 5(6):1515–25., DOI 10. 18632/oncotarget.1765 Hegi, ME, Diserens, AC, Gorlia, T, Hamou,MF, de Tribolet, N, Weller,M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med, 2005, 352(10):997–1003., DOI 10.1056/ NEJMoa043331 Verhaak, RG, Hoadley, KA, Purdom, E, Wang, V, Qi, Y, Wilkerson, MD, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell, 2010, 17(1):98–110., DOI 10.1016/j.ccr.2009.12.020 Maslehaty, H, Cordovi, S, and Hefti, M. Symptomatic spinal metastases of intracranial glioblastoma: clinical characteristics and pathomechanism relating to GFAP expression. J NeuroOncol, 2011, 101(2):329–33., DOI 10.1007/s11060- 010-0257-y Stark, AM, Doukas, A, Hugo, HH, and Mehdorn, HM. The expression of mismatch repair proteins MLH1, MSH2 and MSH6 correlates with the Ki67 proliferation index and survival in patients with recurrent glioblastoma. Neurol Res, 2010, 32(8):816–20., DOI 10.1179/016164110X12645013515052 Li, F, Tian, X, Zhou, Y, Zhu, L, Wang, B, Ding, M, et al. Dysregulated expression of secretogranin III is involved in neurotoxin-induced dopaminergic neuron apoptosis. J Neurosci Res, 2012, 90(12):2237–46., DOI 10.1002/jnr.23121 Taupenot, L, Harper, KL, and O’Connor, DT. The chromograninsecretogranin family. N Engl J Med, 2003, 348(12):1134–49., DOI 10.1056/ NEJMra021405 Hosaka, M, and Watanabe, T. Secretogranin III: a bridge between core hormone aggregates and the secretory granule membrane. Endocr J, 2010, 57(4):275–86., DOI 10.1507/endocrj.k10e-038 Prasad, P, Yanagihara, AA, Small-Howard, AL, Turner, H, and Stokes, AJ. Secretogranin III directs secretory vesicle biogenesis in mast cells in a manner dependent upon interaction with chromogranin A. J Immunol, 2008, 181(7): 5024–34., DOI 10.4049/jimmunol.181.7.5024 Hotta, K, Hosaka, M, Tanabe, A, and Takeuchi, T. Secretogranin II binds to secretogranin III and forms secretory granules with orexin, neuropeptide Y, and POMC. J Endocrinol, 2009, 202(1):111–21., DOI 10.1677/JOE-08-0531 Teyssier, JR, Ragot, S, Chauvet-Gelinier, JC, Trojak, B, and Bonin, B. Activation of a ΔFOSB dependent gene expression pattern in the dorsolateral prefrontal cortex of patients with major depressive disorder. J Affect Disord, 2011, 133(1–2):174–8., DOI 10.1016/j.jad.2011.04.021 LeBlanc, ME, Wang, W, Chen, X, Caberoy, NB, Guo, F, Shen, C, et al. Secretogranin III as a disease-associated ligand for antiangiogenic therapy of diabetic retinopathy. J Exp Med, 2017, 214(4):1029–47., DOI 10.1084/jem. 20161802 He, C, Huang, R, Du, F, Zheng, F, Wei, L, and Wu, J. LDL oxidation by THP-1 monocytes: implication of HNP-1, SgIII and DMT-1. Clin Chim Acta, 2009, 402(1–2):102–6., DOI 10.1016/j.cca.2008.12.025 Martyniuk, CJ, Xiong, H, Crump, K, Chiu, S, Sardana, R, Nadler, A, et al. Gene expression profiling in the neuroendocrine brain of male goldfish, Carassius auratus, exposed to 17alpha-ethinylestradiol. Physiol Genomics, 2006, 27(3): 328–36., DOI 10.1152/physiolgenomics.00090.2006 Portela-Gomes, GM, Grimelius, L, and Stridsberg, M. Secretogranin III in human neuroendocrine tumours: a comparative immunohistochemical study with chromogranins A and B and secretogranin II. Regul Pept, 2010, 165(1): 30–5., DOI 10.1016/j.regpep.2010.06.002 Jongsma, J, Oomen, MH, Noordzij, MA, VanWeerden, WM, Martens, GJ, van der Kwast, TH, et al. Androgen deprivation of the PC-310 [correction of prohormone convertase-310] human prostate cancer model system induces neuroendocrine differentiation. Cancer Res, 2000, 60(3):741–8. Wang, Y, Cui, M, Cai, X, Sun, B, Liu, F, Zhang, X, et al. The oncoprotein HBXIP up-regulates SCG3 through modulating E2F1 and miR-509-3p in hepatoma cells. Cancer Lett, 2014, 352(2):169–78., DOI 10.1016/j.canlet.2014. 05.007 Moss, AC, Jacobson, GM, Walker, LE, Blake, NW, Marshall, E, and Coulson, JM. SCG3 transcript in peripheral blood is a prognostic biomarker for RESTdeficient small cell lung cancer. Clin Cancer Res, 2009, 15(1):274–83., DOI 10. 1158/1078-0432.CCR-08-1163 Phillips, HS, Kharbanda, S, Chen, R, Forrest, WF, Soriano, RH, Wu, TD, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell, 2006, 9(3):157–73., DOI 10.1016/j.ccr.2006.02.019 Shirahata, M, Oba, S, Iwao-Koizumi, K, Saito, S, Ueno, N, Oda, M, et al. Using gene expression profiling to identify a prognostic molecular spectrum in gliomas. Cancer Sci, 2009, 100(1):165–72., DOI 10.1111/j.1349-7006.2008. 01002.x Wong, KK, Rostomily, R, and Wong, STC. Prognostic gene discovery in glioblastoma patients using deep learning. Cancers, 2019, 11(1)., DOI 10.3390/ cancers11010053 Louis, DN, Perry, A, Reifenberger, G, von Deimling, A, Figarella-Branger, D, Cavenee, WK, et al. The 2016 world health organization classification of tumors of the central nervous system: a summary. Acta Neuropathol, 2016, 131(6):803–20., DOI 10.1007/s00401-016-1545-1 Paco, S, Pozas, E, and Aguado, F. Secretogranin III is an astrocyte granin that is overexpressed in reactive glia. Cereb Cortex, 2010, 20(6):1386–97., DOI 10.1093/ cercor/bhp202 Berezovsky, AD, Poisson, LM, Cherba, D, Webb, CP, Transou, AD, Lemke, NW, et al. Sox2 promotes malignancy in glioblastoma by regulating plasticity and astrocytic differentiation. Neoplasia, 2014, 16(3):193–25., DOI 10.1016/j. neo.2014.03.006 Shirahata,M, Iwao-Koizumi, K, Saito, S, Ueno, N, Oda, M, Hashimoto, N, et al. Gene expression-based molecular diagnostic system for malignant gliomas is superior to histological diagnosis. Clin Cancer Res, 2007, 13(24):7341–56., DOI 10.1158/1078-0432.CCR-06-2789 Eckel-Passow, JE, Lachance, DH, Molinaro, AM, Walsh, KM, Decker, PA, Sicotte, H, et al. Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors. N Engl J Med, 2015, 372(26):2499–508., DOI 10.1056/ NEJMoa1407279 Kim,YW,Koul,D,Kim,SH, Lucio-Eterovic,AK, Freire,PR, Yao, J, et al. Identification of prognostic gene signatures of glioblastoma: a study based on TCGA data analysis. Neuro-oncology, 2013, 15(7):829–39., DOI 10.1093/neuonc/not024 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 594931 EP 594939 DI 10.3389/pore.2021.594931 PG 9 ER PT J AU Wang, L Li, X Zhao, L Jiang, L Song, X Qi, A Chen, T Ju, M Hu, B Wei, M He, M Zhao, L AF Wang, Lin Li, Xueping Zhao, Lan Jiang, Longyang Song, Xinyue Qi, Aoshuang Chen, Ting Ju, Mingyi Hu, Baohui Wei, Minjie He, Miao Zhao, Lin TI Identification of DNA-Repair-Related Five-Gene Signature to Predict Prognosis in Patients with Esophageal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognostic biomarkers; DNA repair; overall survival; esophageal cancer; small molecular drugs; targeted therapy ID prognostic biomarkers; DNA repair; overall survival; esophageal cancer; small molecular drugs; targeted therapy AB Esophageal cancer (ESCA) is a leading cause of cancer-related mortality, with poor prognosis worldwide. DNA damage repair is one of the hallmarks of cancer. Loss of genomic integrity owing to inactivation of DNA repair genes can increase the risk of cancer progression and lead to poor prognosis. We aimed to identify a novel gene signature related to DNA repair to predict the prognosis of ESCA patients. Based on gene expression profiles of ESCA patients from The Cancer Genome Atlas and gene set enrichment analysis, 102 genes related to DNA repair were identified as candidates. After stepwise Cox regression analysis, we established a five-gene prognostic model comprising DGCR8, POM121, TAF9, UPF3B, and BCAP31. Kaplan-Meier survival analysis confirmed a strong correlation between the prognostic model and survival. Moreover, we verified the clinical value of the prognostic signature under the influence of different clinical parameters. We found that small-molecule drugs (trametinib, selumetinib, and refametinib) could help to improve patient survival. In summary, our study provides a novel and promising prognostic signature based on DNA-repair-related genes to predict survival of patients with ESCA. Systematic data mining provides a theoretical basis for further exploring the molecular pathogenesis of ESCA and identifying therapeutic targets. C1 [Wang, Lin] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Li, Xueping] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Zhao, Lan] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Jiang, Longyang] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Song, Xinyue] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Qi, Aoshuang] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Chen, Ting] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Ju, Mingyi] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Hu, Baohui] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Wei, Minjie] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [He, Miao] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Zhao, Lin] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. RP He, M (reprint author), China Medical University, School of Pharmacy, Department of Pharmacology, Shenyang, China. EM hemiao_cmu@126.com CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer, 2015). 136(5):E359–E386., DOI 10.1002/ijc. 29210 Domper AM, Ferrandez AA, Lanas AA. Esophageal cancer: risk factors, screening and endoscopic treatment in Western and Eastern countries. World J Gastroenterol, 2015). 21(26):7933–7943., DOI 10.3748/wjg.v21.i26.7933 Lagergren J, Smyth E, Cunningham D, Lagergren P. Oesophageal cancer. The Lancet, 2017). 390(10110):2383–2396., DOI 10.1016/S0140-6736(17)31462-9 Januszewicz W, Fitzgerald RC. Early detection and therapeutics. Mol Oncol, 2019). 13(3):599–613., DOI 10.1002/1878-0261.12458 Nishizuka SS, Mills GB. New era of integrated cancer biomarker discovery using reverse-phase protein arrays. Drug Metab Pharmacokinet, 2016). 31(1):35–45., DOI 10.1016/j.dmpk.2015.11.009 Haisley KR, Hart CM, Kaempf AJ, Dash NR, Dolan JP, Hunter JG. Specific tumor characteristics predict upstaging in early-stage esophageal cancer. Ann Surg Oncol, 2019). 26(2):514–522., DOI 10.1245/s10434-018-6804-z Pearl LH, Schierz AC, Ward SE, Al-Lazikani B, Pearl FMG. Therapeutic opportunities within the DNA damage response. Nat Rev Cancer, 2015). 15(3):166–180., DOI 10.1038/nrc3891 Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature, 2008). 455(7216):1069–1075., DOI 10.1038/nature07423 Zhou Y, Chu L, Wang Q, Dai W, Zhang X, Chen J, et al. CD59 is a potential biomarker of esophageal squamous cell carcinoma radioresistance by affecting DNA repair. Cell Death Dis, 2018). 9(9):887., DOI 10.1038/s41419-018-0895-0 Yang Q, Lin W, Liu Z, Zhu J, Huang N, Cui Z, et al. RAP80 is an independent prognosis biomarker for the outcome of patients with esophageal squamous cell carcinoma. Cel Death Dis, 2018). 9(2):146., DOI 10.1038/s41419-017-0177-2 Kuo I-Y, Huang Y-L, Lin C-Y, Lin C-H, Chang W-L, Lai W-W, et al. SOX17 overexpression sensitizes chemoradiation response in esophageal cancer by transcriptional down-regulation of DNA repair and damage response genes. J Biomed Sci, 2019). 26(1):20., DOI 10.1186/s12929-019-0510-4 Deng M, Bragelmann J, Schultze JL, Perner S. Web-TCGA: an online platform for integrated analysis of molecular cancer data sets. BMC Bioinformatics, 2016). 17(1):72., DOI 10.1186/s12859-016-0917-9 Weinstein JN, Collisson EA, Collisson EA, Mills GB, Shaw KRM, Ozenberger BA, et al. The cancer genome Atlas pan-cancer analysis project. Nat Genet, 2013). 45(10):1113–1120., DOI 10.1038/ng.2764 Bakhoum MF, Esmaeli B. Molecular characteristics of uveal melanoma: insights from the cancer genome Atlas, TCGA, project. Cancers, 2019). 11(8):1061., DOI 10.3390/cancers11081061 He W, Chen L, Yuan K, Zhou Q, Peng L, Han Y. Gene set enrichment analysis and meta-analysis to identify six key genes regulating and controlling the prognosis of esophageal squamous cell carcinoma. J Thorac Dis, 2018). 10(10): 5714–5726., DOI 10.21037/jtd.2018.09.55 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci, 2005). 102(43):15545–15550., DOI 10.1073/pnas.0506580102 Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun, 2019). 10(1):1523., DOI 10.1038/s41467-019-09234-6 Franz M, Rodriguez H, Lopes C, Zuberi K, Montojo J, Bader GD, et al. GeneMANIA update 2018. Nucleic Acids Res, 2018). 46(W1):W60–W64., DOI 10.1093/nar/gky311 Liu C-J, Hu F-F, Xia M-X, Han L, Zhang Q, Guo A-Y. GSCALite: a web server for gene set cancer analysis. Bioinformatics, 2018). 34(21):3771–3772., DOI 10. 1093/bioinformatics/bty411 Calses PC, Dhillon KK, Tucker N, Chi Y, Huang J-w., Kawasumi M, et al. DGCR8 mediates repair of UV-induced DNA damage independently of RNA processing. Cel Rep, 2017). 19(1):162–174., DOI 10.1016/j.celrep.2017.03.021 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA: A Cancer J Clinicians, 2015). 65(2):87–108., DOI 10.3322/ caac.21262 Ma W, Zhang C-Q, Dang C-X, Cai H-Y, Li H-l., Miao G-Y, et al. Upregulated long-non-coding RNA DLEU2 exon 9 expression was an independent indicator of unfavorable overall survival in patients with esophageal adenocarcinoma. Biomed Pharmacother, 2019). 113:108655., DOI 10.1016/j. biopha.2019.108655 Dong G, Mao Q, Yu D, Zhang Y, Qiu M, Dong G, et al. Integrative analysis of copy number and transcriptional expression profiles in esophageal cancer to identify a novel driver gene for therapy. Sci Rep, 2017). 7(1):42060., DOI 10. 1038/srep42060 Zhu X, Luo X, Feng G, Huang H, He Y, Ma W, et al. CENPE expression is associated with its DNA methylation status in esophageal adenocarcinoma and independently predicts unfavorable overall survival. PLoS One, 2019). 14(2): e0207341., DOI 10.1371/journal.pone.0207341 Mao Y, Fu Z, Zhang Y, Dong L, Zhang Y, Zhang Q, et al. A seven-lncRNA signature predicts overall survival in esophageal squamous cell carcinoma. Sci Rep, 2018). 8(1):8823., DOI 10.1038/s41598-018-27307-2 Dong Z, Zhang H, Zhan T, Xu S. Integrated analysis of differentially expressed genes in esophageal squamous cell carcinoma using bioinformatics. Neoplasma, 2018). 65(4):523–531., DOI 10.4149/neo_2018_170708N470 Men CD, Liu QN, Ren Q. A prognostic 11 genes expression model for ovarian cancer. J Cel Biochem., 2018). 119(2):1971–1978., DOI 10.1002/jcb.26358 Li J, Wang W, Xia P, Wan L, Zhang L, Yu L, et al. Identification of a fivelncRNA signature for predicting the risk of tumor recurrence in patients with breast cancer. Int J Cancer, 2018). 143(9):2150–2160., DOI 10.1002/ijc.31573 Chen L, Lu D, Sun K, Xu Y, Hu P, Li X, et al. Identification of biomarkers associated with diagnosis and prognosis of colorectal cancer patients based on integrated bioinformatics analysis. Gene, 2019). 692:119–125., DOI 10.1016/j. gene.2019.01.001 Sancar A, Lindsey-Boltz LA, Unsal-Kacmaz K, Linn S. Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annu Rev Biochem, 2004). 73:39–85., DOI 10.1146/annurev.biochem.73.011303.073723 Rocha C, Silva M, Quinet A, Cabral-Neto J, Menck C. DNA repair pathways and cisplatin resistance: an intimate relationship. Clinics, 2018). 73(Suppl. 1): e478s., DOI 10.6061/clinics/2018/e478s Swahari V, Nakamura A, Baran-Gale J, Garcia I, Crowther AJ, Sons R, et al. Essential function of dicer in resolving DNA damage in the rapidly dividing cells of the developing and malignant cerebellum. Cel Rep, 2016). 14(2): 216–224., DOI 10.1016/j.celrep.2015.12.037 Wen J, Lv Z, Ding H, Fang X, Sun M. Association of miRNA biosynthesis genes DROSHA and DGCR8 polymorphisms with cancer susceptibility: a systematic review and meta-analysis. Biosci Rep, 2018). 38(3):BSR20180072., DOI 10.1042/ BSR20180072 Rodriguez-Bravo V, Pippa R, Song W-M, Carceles-Cordon M, Dominguez- Andres A, Fujiwara N, et al. Nuclear pores promote lethal prostate cancer by increasing pom121-driven E2F1, MYC, and AR nuclear import. Cell, 2018). 174(5):1200–1215.e20., DOI 10.1016/j.cell.2018.07.015 Guo J, Liu X, Wu C, Hu J, Peng K, Wu L, et al. The transmembrane nucleoporin Pom121 ensures efficient HIV-1 pre-integration complex nuclear import. Virology, 2018). 521:169–174., DOI 10.1016/j.virol.2018. 06.008 Saint M, Sawhney S, Sinha I, Singh RP, Dahiya R, Thakur A, et al. The TAF9 C-terminal conserved region domain is required for SAGA and TFIID promoter occupancy to promote transcriptional activation. Mol Cell Biol, 2014). 34(9):1547–1563., DOI 10.1128/mcb.01060-13 Yoon JW, Lamm M, Iannaccone S, Higashiyama N, Leong KF, Iannaccone P, et al. p53 modulates the activity of the GLI1 oncogene through interactions with the shared coactivator TAF9. DNA Repair, 2015). 34:9–17., DOI 10.1016/j.dnarep.2015. 06.006 Huang L, Low A, Damle SS, Keenan MM, Kuntz S, Murray SF, et al. Antisense suppression of the nonsense mediated decay factor Upf3b as a potential treatment for diseases caused by nonsense mutations. Genome Biol, 2018). 19(1):4., DOI 10.1186/s13059-017-1386-9 Elsemman IE, Mardinoglu A, Shoaie S, Soliman TH, Nielsen J. Systems biology analysis of hepatitis C virus infection reveals the role of copy number increases in regions of chromosome 1q in hepatocellular carcinoma metabolism. Mol Biosyst, 2016). 12(5):1496–1506., DOI 10. 1039/c5mb00827a Benevolenskaya EV, Islam ABMMK, Ahsan H, Kibriya MG, Jasmine F, Wolff B, et al. DNA methylation and hormone receptor status in breast cancer. Clin Epigenet, 2016). 8(1):17., DOI 10.1186/s13148-016-0184-7 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 596899 EP 596911 DI 10.3389/pore.2021.596899 PG 13 ER PT J AU Chalela, R Gonzalez-Garcia, GJ Khilzi, K Curull, V Sanchez-Font, A Longaron, R Rodrigo-Calvo, TM Martin-Ontiyuelo, C Gea, J Bellosillo, B AF Chalela, Roberto Gonzalez-Garcia, Gregorio Jose Khilzi, Karys Curull, Victor Sanchez-Font, Albert Longaron, Raquel Rodrigo-Calvo, Teresa Maria Martin-Ontiyuelo, Clara Gea, Joaquim Bellosillo, Beatriz TI EGFR and KRAS Mutations in Lung Parenchyma of Subjects With EGFR/ KRAS Wild-Type Lung Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE adenocacinoma lung; driver mutation; EGFR–epidermal growth factor receptor; KRAS; Prognosis ID adenocacinoma lung; driver mutation; EGFR–epidermal growth factor receptor; KRAS; Prognosis AB The acquisition of driver mutations in non-tumoral cells appears to be very important during the carcinogenesis of adenocarcinoma (ADC). Recent studies suggest that cancer-related mutations may not necessarily be present only in malignant cells, but also in histologically “healthy cells”. Objective: to demonstrate the presence of EGFR or KRAS mutations in non-tumoral lung cells in subjects with ADC and negative mutational status. Results: mutations in EGFR or KRAS oncogenes were identified in the normal lung in 9.7% of the subjects. Exon 21 substitution L858R in EGFR was detected in two cases while the exon 19 deletion E746-A750 in the EGFR, the G12C and G12D substitutions in the KRAS were detected once. One patient presented three different mutations in the normal lung parenchyma (EGFR_L858R, KRAS_G12C and KRAS_G12D). The negative-mutation status of the tumor and the mutations detected in the “normal lung” were confirmed using highly sensitive and specific TaqMan PCR (CAST-PCR). No differences were found in terms of progression, progression-free survival or overall survival during the 18 months follow-up. Conclusions: These results confirm the presence of driver mutations in the histologically normal lung parenchyma cells in the absence of mutations coexisting with the primary tumor. C1 [Chalela, Roberto] Hospital delMar Medical Research Institute (IMIM)Barcelona, Spain. [Gonzalez-Garcia, Gregorio Jose] Hospital delMar Medical Research Institute (IMIM)Barcelona, Spain. [Khilzi, Karys] Hospital del Mar PSMAR, Respiratory Medicine DepartmentBarcelona, Spain. [Curull, Victor] Hospital delMar Medical Research Institute (IMIM)Barcelona, Spain. [Sanchez-Font, Albert] Hospital delMar Medical Research Institute (IMIM)Barcelona, Spain. [Longaron, Raquel] Hospital del Mar PSMAR, Pathology DepartmentBarcelona, Spain. [Rodrigo-Calvo, Teresa Maria] Hospital del Mar PSMAR, Pathology DepartmentBarcelona, Spain. [Martin-Ontiyuelo, Clara] Hospital delMar Medical Research Institute (IMIM)Barcelona, Spain. [Gea, Joaquim] Hospital delMar Medical Research Institute (IMIM)Barcelona, Spain. [Bellosillo, Beatriz] Hospital delMar Medical Research Institute (IMIM)Barcelona, Spain. RP Chalela, R (reprint author), Hospital delMar Medical Research Institute (IMIM), Barcelona, Spain. EM rchalela@psmar.cat CR Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA: A Cancer J Clinicians, 2016). 66:7–30., DOI 10.3322/caac.21332 American Cancer Society. Cancer facts & figures 2016. Atlanta: American Cancer Society, 2016). Goldstraw P, Chansky K, Crowley J, Rami-Porta R, Asamura H, Eberhardt WEE, et al. The IASLC lung cancer staging project: proposals for revision of the TNMstage groupings in the forthcoming, eighth, edition of the TNM Classification for lung cancer. J Thorac Oncol, 2016). 11:39–51., DOI 10.1016/j.jtho.2015.09.009 Nakagawa T, Okumura N, Ohata K, Igai H, Matsuoka T, Kameyama K. Postrecurrence survival in patients with stage I non-small cell lung cancer. Eur J Cardio-Thoracic Surg, 2008). 34:499–504., DOI 10.1016/j.ejcts.2008.05.016 Martini N, Bains MS, Burt ME, Zakowski MF, McCormack P, Rusch VW, et al. Incidence of local recurrence and second primary tumors in resected stage I lung cancer. J Thorac Cardiovasc Surg, 1995). 109:120–9., DOI 10.1016/s0022-5223(95)70427-2 Chalela R, Curull V, Enriquez C, Pijuan L, Bellosillo B, Gea J. Lung adenocarcinoma: from molecular basis to genome-guided therapy and immunotherapy. J Thorac Dis, 2017). 9:2142–58., DOI 10.21037/jtd.2017.06.20 Stratton MR, Campbell PJ, Futreal PA. The cancer genome. Nature, 2009). 458(7239):719–24., DOI 10.1038/nature07943 Anglesio MS, Papadopoulos N, Ayhan A, Nazeran TM, Noe M, Horlings HM, et al. Cancer-associated mutations in endometriosis without cancer. N Engl J Med, 2017). 376(19):1835–48., DOI 10.1056/nejmoa1614814 Nikolaev SI, Vetiska S, Bonilla X, Boudreau E, Jauhiainen S, Rezai Jahromi B, et al. Somatic activating KRAS mutations in arteriovenous malformations of the brain. N Engl J Med, 2018). 378(3):250–61., DOI 10. 1056/nejmoa1709449 ChalelaR, BellosilloB, Curull V, LongaronR, Pascual-Guardia S,Badenes-BonetD, et al. EGFR and KRAS mutations in the non-tumoral lung. Prognosis in patients with adenocarcinoma. Jcm, 2019). 8(4):529., DOI 10.3390/jcm8040529 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 598292 EP 598297 DI 10.3389/pore.2021.598292 PG 6 ER PT J AU Chen, M Nie, Z Cao, H Gao, Y Wen, X Zhang, Ch Zhang, Sh AF Chen, Mei Nie, Zhenyu Cao, Hui Gao, Yuanhui Wen, Xiaohong Zhang, Chong Zhang, Shufang TI Rac3 Expression and its Clinicopathological Significance in Patients With Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Rac3; bladder cancer; TCGA; GSEA; prognose; clinicopathological significance ID Rac3; bladder cancer; TCGA; GSEA; prognose; clinicopathological significance AB Background: Ras-related C3 botulinum toxin substrate 3 (Rac3) is overexpressed in malignancies and promotes tumor progression. However, the correlations between Rac3 expression and the clinicopathological characteristics and prognoses of patients with bladder cancer (BC) remain unclear. Methods: Data from The Cancer Genome Atlas (TCGA) were used to analyze Rac3 expression in BC and normal bladder tissues and validated using the Oncomine database, quantitative real-time PCR (qRT-PCR) and western blot. The Kaplan-Meier method was used to analyze the relationship between Rac3 expression and the prognosis of patients with BC. Cox univariate and multivariate analyses of BC patients overall survival (OS) were performed. Signaling pathways that potentially mediate Rac3 activity in BC were then analyzed by gene set enrichment analysis (GSEA). Results: The Rac3 expression in BC tissues was significantly higher than that in normal bladder tissues. Rac3 expression was significantly correlated with grade and stage. Overexpression of Rac3 was associated with a poor prognosis. GSEA showed that the cell cycle, DNA replication, p53 signaling pathway and mismatch repair were differentially enriched in the high Rac3 expression phenotype. The qRT-PCR and western blot results confirmed that the Rac3 expression in BC tissues was higher than that in normal bladder tissues. Conclusion: Rac3 is highly expressed in BC, which is related to the advanced clinicopathological variables and adverse prognosis of patients with BC. These results provide a new therapeutic target for BC. C1 [Chen, Mei] Central South University, Affiliated Hai Kou Hospital, Xiangya Medical College, Central LaboratoryHaikou, China. [Nie, Zhenyu] Central South University, Affiliated Hai Kou Hospital, Xiangya Medical College, Central LaboratoryHaikou, China. [Cao, Hui] Central South University, Affiliated Hai Kou Hospital, Xiangya Medical College, Central LaboratoryHaikou, China. [Gao, Yuanhui] Central South University, Affiliated Hai Kou Hospital, Xiangya Medical College, Central LaboratoryHaikou, China. [Wen, Xiaohong] Central South University, Affiliated Hai Kou Hospital, Xiangya Medical College, Central LaboratoryHaikou, China. [Zhang, Chong] Central South University, Affiliated Haikou Hospital of Xiangya Medical College, UrologyHaikou, China. [Zhang, Shufang] Central South University, Affiliated Hai Kou Hospital, Xiangya Medical College, Central LaboratoryHaikou, China. RP Zhang, Ch (reprint author), Central South University, Affiliated Haikou Hospital of Xiangya Medical College, Urology, Haikou, China. EM 763954234@qq.com CR Ferlay J, Shin H-R, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer, 2010). 127(12):2893–917., DOI 10.1002/ijc.25516 Samaratunga H, Makarov DV, Epstein JI. Comparison of WHO/ISUP and WHO classification of noninvasive papillary urothelial neoplasms for risk of progression. Urology, 2002). 60(2):315–9., DOI 10.1016/s0090-4295(02)01705-3 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA: A Cancer J Clinicians, 2017). 67(1):7–30., DOI 10.3322/caac.21387 Kamat AM, Hahn NM, Efstathiou JA, Lerner SP, Malmstrom P-U, Choi W, et al. Bladder cancer. The Lancet, 2016). 388(10061):2796–810., DOI 10.1016/ s0140-6736(16)30512-8 Johnson DC, Greene PS, Nielsen ME. Surgical advances in bladder cancer. Urol Clin North America, 2015). 42(2):235–52., DOI 10.1016/j.ucl.2015.01.005 Chavan S, Bray F, Lortet-Tieulent J, Goodman M, Jemal A. International variations in bladder cancer incidence and mortality. Eur Urol, 2014). 66(1): 59–73., DOI 10.1016/j.eururo.2013.10.001 Bruchbacher A, Soria F, Hassler M, Shariat SF, D’Andrea D. Tissue biomarkers in nonmuscle-invasive bladder cancer. Curr Opin Urol, 2018). 28(6):584–90., DOI 10.1097/mou.0000000000000546 Van Aelst L, D’Souza-Schorey C. Rho GTPases and signaling networks. Genes Develop, 1997). 11(18):2295–322., DOI 10.1101/gad.11.18.2295 Fritz G, Just I, Kaina B. Rho GTPases are over-expressed in human tumors. Int J Cancer, 1999). 81(5):682–7., DOI 10.1002/(sici)1097-0215(19990531)81:5<682:: aid-ijc2>3.0.co;2-b Onesto C, Shutes A, Picard V, Schweighoffer F, Der CJ. Characterization of EHT 1864, a novel small molecule inhibitor of Rac family small GTPases. Methods Enzymol, 2008). 439:111–29., DOI 10.1016/s0076-6879(07, 00409-0 Haataja L, Groffen J, Heisterkamp N. Characterization of RAC3, a novel member of the Rho family. J Biol Chem, 1997). 272(33):20384–8., DOI 10.1074/ jbc.272.33.20384 Engers R, Ziegler S, Mueller M, Walter A, Willers R, Gabbert HE. Prognostic relevance of increased Rac GTPase expression in prostate carcinomas. Endocr Relat Cancer, 2007). 14(2):245–56., DOI 10.1677/erc-06-0036 Hwang S-L, Chang J-H, Cheng T-S, Sy W-D, Lieu A-S, Lin C-L, et al. Expression of Rac3 in human brain tumors. J Clin Neurosci, 2005). 12(5): 571–4., DOI 10.1016/j.jocn.2004.08.013 Lee J-S, Leem S-H, Lee S-Y, Kim S-C, Park E-S, Kim S-B, et al. Expression signature of E2F1 and its associated genes predict superficial to invasive progression of bladder tumors. Jco, 2010). 28(16):2660–7., DOI 10.1200/jco. 2009.25.0977 Sanchez-Carbayo M, Socci ND, Lozano J, Saint F, Cordon-Cardo C. Defining molecular profiles of poor outcome in patients with invasive bladder cancer using oligonucleotide microarrays. JCO, 2006). 24(5):778–89., DOI 10.1200/jco. 2005.03.2375 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci, 2005). 102(43):15545–50., DOI 10.1073/pnas.0506580102 Dong S, Zhao J, Wei J, Bowser RK, Khoo A, Liu Z, et al. F-box protein complex FBXL19 regulates TGFβ1-induced E-cadherin down-regulation by mediating Rac3 ubiquitination and degradation. Mol Cancer, 2014). 13:76., DOI 10.1186/ 1476-4598-13-76 Chan AY, Coniglio SJ, Chuang Y-Y, Michaelson D, Knaus UG, Philips MR, et al. Roles of the Rac1 and Rac3 GTPases in human tumor cell invasion. Oncogene, 2005). 24(53):7821–9., DOI 10.1038/sj.onc.1208909 Baugher PJ, Krishnamoorthy L, Price JE, Dharmawardhane SF. Rac1 and Rac3 isoform activation is involved in the invasive and metastatic phenotype of human breast cancer cells. Breast Cancer Res, 2005). 7(6):R965–R974., DOI 10. 1186/bcr1329 Mira J-P, Benard V, Groffen J, Sanders LC, Knaus UG. Endogenous, hyperactive Rac3 controls proliferation of breast cancer cells by a p21- activated kinase-dependent pathway. Proc Natl Acad Sci, 2000). 97(1): 185–9., DOI 10.1073/pnas.97.1.185 Gest C, Joimel U, Huang L, Pritchard LL, Petit A, Dulong C, et al. Rac3 induces a molecular pathway triggering breast cancer cell aggressiveness: differences in MDA-MB-231 and MCF-7 breast cancer cell lines. BMC Cancer, 2013). 13:63., DOI 10.1186/1471-2407-13-63 Pan Y, Bi F, Liu N, Xue Y, Yao X, Zheng Y, et al. Expression of seven main Rho family members in gastric carcinoma. Biochem Biophysical Res Commun, 2004). 315(3):686–91., DOI 10.1016/j.bbrc.2004.01.108 Qiu HD, Hu XR, He C, Yu BB, Li YQ, Li JN. Identification and validation of an individualized prognostic signature of bladder cancer based on seven immune related genes. Front Genet, 2020). 11:12., DOI 10.3389/fgene.2020.00012 Liu L, Hu J, Wang Y, Sun T, Zhou X, Li X, et al. Establishment of a novel risk score model by comprehensively analyzing the immunogen database of bladder cancer to indicate clinical significance and predict prognosis. Aging, 2020). 12(12):11967–89., DOI 10.18632/aging.103364 Na L, Bai Y, Sun Y, Wang Z, Wang W, Yuan L, et al. Identification of 9-core immune-related genes in bladder urothelial carcinoma prognosis. Front Oncol, 2020). 10:1142., DOI 10.3389/fonc.2020.01142 Tang Y, Hu Y, Wang J, Zeng Z. A novel risk score based on a combined signature of 10 immune system genes to predict bladder cancer prognosis. Int Immunopharmacology, 2020). 87:106851., DOI 10.1016/j.intimp.2020.106851 Cheng CY, Song DK, Wu YD, Liu BQ. RAC3 promotes proliferation, migration and invasion via PYCR1/JAK/STAT signaling in bladder cancer. Front Mol biosciences, 2020). 7:218., DOI 10.3389/fmolb.2020.00218 Zhang C, Liu T, Wang G, Wang H, Che X, Gao X, et al. Rac3 regulates cell invasion, migration and EMT in lung adenocarcinoma through p38 MAPK pathway. J Cancer, 2017). 8(13):2511–22., DOI 10.7150/jca.18161 Wang L, Fu D, Qiu Y, Xing X, Xu F, Han C, et al. Genome-wide screening and identification of long noncoding RNAs and their interaction with protein coding RNAs in bladder urothelial cell carcinoma. Cancer Lett, 2014). 349(1): 77–86., DOI 10.1016/j.canlet.2014.03.033 Mootha VK, Lindgren CM, Eriksson K-F, Subramanian A, Sihag S, Lehar J, et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet, 2003). 34(3): 267–73., DOI 10.1038/ng1180 Cantini L, Isella C, Petti C, Picco G, Chiola S, Ficarra E, et al. MicroRNAmRNA interactions underlying colorectal cancer molecular subtypes. Nat Commun, 2015). 6:8878., DOI 10.1038/ncomms9878 Menzl I, Zhang T, Berger-Becvar A, Grausenburger R, Heller G, Prchal- Murphy M, et al. A kinase-independent role for CDK8 in BCR-ABL1(+, leukemia. Nat Commun, 2019). 10(1):4741., DOI 10.1038/s41467-019-12656-x Lee K-S, Kim S-W, Lee H-S. Orostachys japonicus induce p53-dependent cell cycle arrest through the MAPK signaling pathway in OVCAR-3 human ovarian cancer cells. Food Sci Nutr, 2018). 6(8):2395–401., DOI 10.1002/ fsn3.836 Liu T-Q, Wang G-B, Li Z-J, Tong X-D, Liu H-X. Silencing of Rac3 inhibits proliferation and induces apoptosis of human lung cancer cells. Asian Pac J Cancer Prev, 2015). 16(7):3061–5., DOI 10.7314/apjcp.2015.16.7.3061 Wang G, Wang H, Zhang C, Liu T, Li Q, Lin X, et al. Rac3 regulates cell proliferation through cell cycle pathway and predicts prognosis in lung adenocarcinoma. Tumor Biol, 2016). 37(9):12597–607., DOI 10.1007/s13277- 016-5126-7 Zhu Y, Dai B, Zhang H, Shi G, Shen Y, Ye D. Long non-coding RNA LOC572558 inhibits bladder cancer cell proliferation and tumor growth by regulating the AKT-MDM2-p53 signaling axis. Cancer Lett, 2016). 380(2): 369–74., DOI 10.1016/j.canlet.2016.04.030 Zhang ZF, Zhang HR, Zhang QY, Lai SY, Feng YZ, Zhou Y, et al. High expression of TMEM40 is associated with the malignant behavior and tumorigenesis in bladder cancer. J Transl Med, 2018). 16(1):9., DOI 10.1186/ s12967-017-1377-3 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 598460 EP 598468 DI 10.3389/pore.2021.598460 PG 9 ER PT J AU Kato, KM Yoshida, H Tanase, Y Uno, M Ishikawa, M Kato, T AF Kato, Kobayashi Mayumi Yoshida, Hiroshi Tanase, Yasuhito Uno, Masaya Ishikawa, Mitsuya Kato, Tomoyasu TI Loss of ARID1A Expression as a Favorable Prognostic Factor in Early-Stage Grade 3 Endometrioid Endometrial Carcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE endometrioid endometrial carcinoma; grade 3; prognosis; ARID1A; p53; mismatch repair ID endometrioid endometrial carcinoma; grade 3; prognosis; ARID1A; p53; mismatch repair AB Introduction: High-risk patients with grade 3 endometrioid endometrial carcinoma (G3EEC) who require adjuvant therapy have not been clearly identified. Therefore, the current study aimed to investigate the prognostic impact of ARID1A, p53, and mismatch repair (MMR) protein expressions, previously reported as prognosticators in some gynecological cancers, in patients with early-stage G3EEC. Methods: A total of 67 patients with pathologically confirmed early-stage G3EEC diagnosed between 1997 and 2020 were identified; none received adjuvant chemotherapy. The recurrence-free survival (RFS) and overall survival (OS) were estimated using the Kaplan-Meier method and compared with a log-rank test. The protein expressions of ARID1A, p53, and MMR were examined via immunohistochemistry, and the associations between these biomarkers and clinical outcomes were evaluated. Results: Recurrence was observed in 9 (13%) of the 67 patients with early stage G3EEC. The respective 5-years RFS and OS rates were 87.7% and 93.7%, and 68.6%and 85.7%, respectively for stages I and II. Multivariate analysis showed significantly longer RFS among patients with ARID1A loss (hazard ratio = 8.7; 95% CI, 1.09–69.6, p = 0.04). No significant differences were observed in RFS and OS of patients according to p53 and MMR expression status. Conclusion: ARID1A expression status was a prognosticator for patients with early stage G3EEC without adjuvant therapy, whereas p53 and MMR expression status showed no impact on survival outcomes. ARID1A may become a useful biomarker for stratification of adjuvant treatment for early stage G3EEC patients. C1 [Kato, Kobayashi Mayumi] National Cancer Center Hospital, Department of GynecologyTokyo, Japan. [Yoshida, Hiroshi] National Cancer Center Hospital, Department of Diagnostic PathologyTokyo, Japan. [Tanase, Yasuhito] National Cancer Center Hospital, Department of GynecologyTokyo, Japan. [Uno, Masaya] National Cancer Center Hospital, Department of GynecologyTokyo, Japan. [Ishikawa, Mitsuya] National Cancer Center Hospital, Department of GynecologyTokyo, Japan. [Kato, Tomoyasu] National Cancer Center Hospital, Department of GynecologyTokyo, Japan. RP Kato, KM (reprint author), National Cancer Center Hospital, Department of Gynecology, Tokyo, Japan. EM maykobay@ncc.go.jp CR Siegel RL,Miller KD, Jemal A. Cancer statistics, 2019. CA A Cancer J Clin, 2019, 69(1):7–34., DOI 10.3322/caac.21551 McGunigal M, Liu J, Kalir T, Chadha M, Gupta V. Survival differences among uterine papillary serous, clear cell and grade 3 endometrioid adenocarcinoma endometrial cancers: a national cancer database analysis. Int J Gynecol Cancer, 2017, 27(1):85–92., DOI 10.1097/IGC.0000000000000844 Voss MA, Ganesan R, Ludeman L, McCarthy K, Gornall R, Schaller G, et al. Should grade 3 endometrioid endometrial carcinoma be considered a type 2 cancer-a clinical and pathological evaluation. Gynecol Oncol, 2012, 124(1): 15–20., DOI 10.1016/j.ygyno.2011.07.030 Creasman WT, Kohler MF, Odicino F, Maisonneuve P, Boyle P. Prognosis of papillary serous, clear cell, and grade 3 stage I carcinoma of the endometrium. Gynecol Oncol, 2004, 95(3):593–6., DOI 10.1016/j.ygyno.2004.08.019 Alektiar KM, McKee A, Lin O, Venkatraman E, Zelefsky MJ, McKee B, et al. Is there a difference in outcome between stage I-II endometrial cancer of papillary serous/clear cell and endometrioid FIGO Grade 3 cancer? Int J Radiat Oncology*Biology*Physics, 2002, 54(1):79–85., DOI 10.1016/s0360-3016(02)02913-9 Cirisano FD, Jr, Robboy SJ, Dodge RK, Bentley RC, Krigman HR, Synan IS, et al. The outcome of stage I-II clinically and surgically staged papillary serous and clear cell endometrial cancers when compared with endometrioid carcinoma. Gynecol Oncol, 2000, 77(1):55–65., DOI 10.1006/ gyno.2000.5737 Soslow RA, Bissonnette JP,Wilton A, Ferguson SE, Alektiar KM, Duska LR, et al. Clinicopathologic analysis of 187 high-grade endometrial carcinomas of different histologic subtypes: similar outcomes belie distinctive biologic differences. Am J Surg Pathol, 2007, 31(7):979–87., DOI 10.1097/PAS.0b013e31802ee494 Alvarez T, Miller E, Duska L, Oliva E. Molecular profile of grade 3 endometrioid endometrial carcinoma. Am J Surg Pathol, 2012, 36(5):753–61., DOI 10.1097/ PAS.0b013e318247b7bb Hamilton CA, Cheung MK, Osann K, Chen L, Teng NN, Longacre TA, et al. Uterine papillary serous and clear cell carcinomas predict for poorer survival compared to grade 3 endometrioid corpus cancers. Br J Cancer, 2006, 94(5): 642–6., DOI 10.1038/sj.bjc.6603012 Alkushi A, Kobel M, Kalloger SE, Gilks CB. High-grade endometrial carcinoma: serous and grade 3 endometrioid carcinomas have different immunophenotypes and outcomes. Int J Gynecol Pathol, 2010, 29(4): 343–50., DOI 10.1097/PGP.0b013e3181cd6552 Boruta DM, 2nd, Gehrig PA, Groben PA, Bae-Jump V, Boggess JF, Fowler WC, Jr, et al. Uterine serous and grade 3 endometrioid carcinomas. Cancer, 2004, 101(10):2214–21., DOI 10.1002/cncr.20645 Saso S, Chatterjee J, Georgiou E, Ditri AM, Smith JR, Ghaem-Maghami S. Endometrial cancer. BMJ, 2011, 343:d3954., DOI 10.1136/bmj.d3954 Heckl M, Schmoeckel E, Hertlein L, Rottmann M, Jeschke U, Mayr D. The ARID1A, p53 and ss-Catenin statuses are strong prognosticators in clear cell and endometrioid carcinoma of the ovary and the endometrium. PLoS One, 2018, 13(2):e0192881., DOI 10.1371/journal.pone.0192881 Pasanen A, Loukovaara M, Butzow R. Clinicopathological significance of deficient DNA mismatch repair and MLH1 promoter methylation in endometrioid endometrial carcinoma. Mod Pathol, 2020, 33:1443., DOI 10. 1038/s41379-020-0501-8 Shen J, Ju Z, Zhao W, Wang L, Peng Y, Ge Z, et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat Med, 2018, 24(5):556–62., DOI 10.1038/s41591-018-0012-z Zhang Z-M, Xiao S, Sun G-Y, Liu Y-P, Zhang F-P, Yang H-F, et al. The clinicopathologic significance of the loss of BAF250a, ARID1A, expression in endometrial carcinoma. Int J Gynecol Cancer, 2014, 24(3):534–40., DOI 10. 1097/IGC.0000000000000092 Susumu N, Sagae S, Udagawa Y, Niwa K, Kuramoto H, Satoh S, et al. Randomized phase III trial of pelvic radiotherapy versus cisplatin-based combined chemotherapy in patients with intermediate- and high-risk endometrial cancer: a Japanese Gynecologic Oncology Group study. Gynecol Oncol, 2008, 108(1):226–33., DOI 10.1016/j.ygyno.2007.09.029 Hall G, Clarkson A, Shi A, Langford E, Leung H, Eckstein RP, et al. Immunohistochemistry for PMS2 and MSH6 alone can replace a four antibody panel for mismatch repair deficiency screening in colorectal adenocarcinoma. Pathology, 2010, 42(5):409–13., DOI 10.3109/00313025. 2010.493871 Rasool N, Fader AN, Seamon L, Neubauer NL, Shahin FA, Alexander HA, et al. Stage I, grade 3 endometrioid adenocarcinoma of the endometrium: an analysis of clinical outcomes and patterns of recurrence. Gynecol Oncol, 2010, 116(1):10–4., DOI 10.1016/j.ygyno.2009.10.043 Thomas S, Hussein Y, Bandyopadhyay S, Cote M, Hassan O, Abdulfatah E, et al. Interobserver variability in the diagnosis of uterine high-grade endometrioid carcinoma. Arch Pathol Lab Med, 2016, 140(8):836–43., DOI 10.5858/arpa.2015-0220-OA Wang X, Nagl NG, Wilsker D, Van Scoy M, Pacchione S, Yaciuk P, et al. Two related ARID family proteins are alternative subunits of human SWI/SNF complexes. Biochem J, 2004, 383(Pt 2):319–25., DOI 10.1042/BJ20040524 Cancer Genome Atlas Research N, Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated genomic characterization of endometrial carcinoma. Nature, 2013, 497(7447):67–73., DOI 10.1038/nature12113 McConechy MK, Ding J, Cheang MC, Wiegand KC, Senz J, Tone AA, et al. Use of mutation profiles to refine the classification of endometrial carcinomas. J Pathol, 2012, 228(1):20–30., DOI 10.1002/path.4056 Wiegand KC, Lee AF, Al-Agha OM, Chow C, Kalloger SE, Scott DW, et al. Loss of BAF250a, ARID1A, is frequent in high-grade endometrial carcinomas. J Pathol, 2011, 224(3):328–33., DOI 10.1002/path.2911 Mao T-L, Ardighieri L, Ayhan A, Kuo K-T, Wu C-H, Wang T-L, et al. Loss of ARID1A expression correlates with stages of tumor progression in uterine endometrioid carcinoma. Am J Surg Pathol, 2013, 37(9):1342–8., DOI 10.1097/ PAS.0b013e3182889dc3 Werner HMJ, Berg A, Wik E, Birkeland E, Krakstad C, Kusonmano K, et al. ARID1A loss is prevalent in endometrial hyperplasia with atypia and lowgrade endometrioid carcinomas. Mod Pathol, 2013, 26(3):428–34., DOI 10. 1038/modpathol.2012.174 Jiang T, Chen X, Su C, Ren S, Zhou C. Pan-cancer analysis of ARID1A alterations as biomarkers for immunotherapy outcomes. J Cancer, 2020, 11(4): 776–80., DOI 10.7150/jca.41296 Kurz L, Miklyaeva A, Skowron MA, Overbeck N, Poschmann G, Becker T, et al. ARID1A regulates transcription and the epigenetic landscape via POLE and DMAP1 while ARID1A deficiency or pharmacological inhibition sensitizes germ cell tumor cells to ATR inhibition. Cancers, 2020, 12(4): 905., DOI 10.3390/cancers12040905 Lee YH, Heo J-h., Kim TH, Kang H, Kim G, Kim J, et al. Significance of cell cycle regulatory proteins as malignant and prognostic biomarkers in ovarian epithelial tumors. Int J Gynecol Pathol, 2011, 33(3):205–17., DOI 10.1097/PGP. 0b013e3182063e71 Bosse T, Nout RA, McAlpine JN, McConechy MK, Britton H, Hussein YR, et al. Molecular classification of grade 3 endometrioid endometrial cancers identifies distinct prognostic subgroups. Am J Surg Pathol, 2018, 42(5):561–8., DOI 10.1097/pas.0000000000001020 Karamurzin Y, Rutgers JKL. DNA mismatch repair deficiency in endometrial carcinoma. Int J Gynecol Pathol, 2009, 28(3):239–55., DOI 10.1097/PGP. 0b013e31818d8fe6 Garg K, Shih K, Barakat R, Zhou Q, Iasonos A, Soslow RA. Endometrial carcinomas in women aged 40 years and younger: tumors associated with loss of DNA mismatch repair proteins comprise a distinct clinicopathologic subset. Am J Surg Pathol, 2009, 33(12):1869–77., DOI 10.1097/PAS.0b013e3181bc9866 Shih KK, Garg K, Levine DA, Kauff ND, Abu-Rustum NR, Soslow RA, et al. Clinicopathologic significance of DNA mismatch repair protein defects and endometrial cancer in women 40years of age and younger. Gynecol Oncol, 2011, 123(1):88–94., DOI 10.1016/j.ygyno.2011.06.005 Grzankowski KS, Shimizu DM, Kimata C, Black M, Terada KY. Clinical and pathologic features of young endometrial cancer patients with loss of mismatch repair expression. Gynecol Oncol, 2012, 126(3):408–12., DOI 10. 1016/j.ygyno.2012.05.019 Black D, Soslow RA, Levine DA, Tornos C, Chen SC, Hummer AJ, et al. Clinicopathologic significance of defective DNA mismatch repair in endometrial carcinoma. JCO, 2006, 24(11):1745–53., DOI 10.1200/JCO.2005. 04.1574 Zighelboim I, Goodfellow PJ, Gao F, Gibb RK, Powell MA, Rader JS, et al. Microsatellite instability and epigenetic inactivation of MLH1 and outcome of patients with endometrial carcinomas of the endometrioid type. JCO, 2007, 25(15):2042–8., DOI 10.1200/JCO.2006.08.2107 Diaz-Padilla I, Romero N, Amir E, Matias-Guiu X, Vilar E, Muggia F, et al. Mismatch repair status and clinical outcome in endometrial cancer: a systematic review and meta-analysis. Crit Rev Oncology/Hematology, 2013, 88(1):154–67., DOI 10.1016/j.critrevonc.2013.03.002 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 598550 EP 598558 DI 10.3389/pore.2021.598550 PG 9 ER PT J AU Huang, X Xu, J Wu, Y Sheng, L Li, Y Zha, B Sun, T Yang, J Zang, Sh Liu, J AF Huang, Xinmei Xu, Jiong Wu, Yueyue Sheng, Li Li, Yue Zha, Bingbing Sun, Tiange Yang, Ju Zang, Shufei Liu, Jun TI Alterations in CD8+ Tregs, CD56+ Natural Killer Cells and IL-10 Are Associated With Invasiveness of Nonfunctioning Pituitary Adenomas (NFPAs) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE invasive nonfunctioning pituitary adenomas; immune tolerance; CD8+ tregs; natural killer cells; IL-10 ID invasive nonfunctioning pituitary adenomas; immune tolerance; CD8+ tregs; natural killer cells; IL-10 AB Invasive nonfunctioning pituitary adenomas (NFPAs) grow rapidly and the mechanisms are unclear. Among many complex mechanisms, the role of immunity in the development of NFPAs has not been fully explored. Here, we analyzed the clinical features 146 NFPA patients who underwent trans-sphenoidal surgery or craniotomy and examined the effects of immune tolerance in invasiveness of NFPA patients using fluorescence-activated cell sorting and immunohistochemical methods. We found patients with invasive NFPAs had more visual deficits and defective fields, higher tumor size, and lower white blood cell count compared with patients with noninvasive NFPAs. Additionally, compared with patients with noninvasive NFPAs, patients with invasive NFPAs had conspicuously lower CD3−CD56+ natural killer (NK) cells and significantly higher levels of CD3+CD8+CD28-T cells (CD8+ Tregs) and interleukin-10 (IL-10) in peripheral blood. Moreover, patients with invasive NFPAs had lower infiltrated CD56+ cells, less infiltrated CD28+ cells, and significantly greater IL-10 expression. These results demonstrated that low CD56+ cells infiltration and CD28+ cells infiltration, as well as high IL-10 expression in pituitary tumor tissues, were related with increased invasiveness of NFPAs. Levels of CD3−CD56+ NK cells, CD8+ Tregs and IL-10 in the peripheral blood could be feasible diagnostic markers for invasive NFPAs. C1 [Huang, Xinmei] Fudan University, The Fifth People’s Hospital of Shanghai, Department of EndocrinologyShanghai, China. [Xu, Jiong] Fudan University, The Fifth People’s Hospital of Shanghai, Department of EndocrinologyShanghai, China. [Wu, Yueyue] Fudan University, The Fifth People’s Hospital of Shanghai, Department of EndocrinologyShanghai, China. [Sheng, Li] Fudan University, The Fifth People’s Hospital of Shanghai, Department of EndocrinologyShanghai, China. [Li, Yue] Fudan University, The Fifth People’s Hospital of Shanghai, Department of EndocrinologyShanghai, China. [Zha, Bingbing] Fudan University, The Fifth People’s Hospital of Shanghai, Department of EndocrinologyShanghai, China. [Sun, Tiange] Fudan University, The Fifth People’s Hospital of Shanghai, Department of EndocrinologyShanghai, China. [Yang, Ju] Fudan University, The Fifth People’s Hospital of Shanghai, Department of PathologyShanghai, China. [Zang, Shufei] Fudan University, The Fifth People’s Hospital of Shanghai, Department of EndocrinologyShanghai, China. [Liu, Jun] Fudan University, The Fifth People’s Hospital of Shanghai, Department of EndocrinologyShanghai, China. RP Zang, Sh (reprint author), Fudan University, The Fifth People’s Hospital of Shanghai, Department of Endocrinology, Shanghai, China. EM zangshufei@sina.com CR Loyo-Varela, M, Herrada-Pineda, T, Revilla-Pacheco, F, and Manrique- Guzman, S. Pituitary tumor surgery: review of 3004 cases. World Neurosurg, 2013, 79, 2), 331–6., DOI 10.1016/j.wneu.2012.06.024 Bronstein, MD, and Melmed, S. Pituitary tumorigenesis. Arq Bras Endocrinol Metabol, 2005, 49, 5), 615–25., DOI 10.1590/s0004-27302005000500003 Syro, LV, Rotondo, F, Ramirez, A, Di Ieva, A, Sav, MA, Restrepo, LM, et al. Progress in the diagnosis and classification of pituitary adenomas. Front Endocrinol, Lausanne,, 2015, 6, 97., DOI 10.3389/fendo.2015. 00097 Chatzellis, E, Alexandraki, KI, AndroulakisII, and Kaltsas, G. Aggressive pituitary tumors. Neuroendocrinology, 2015, 101, 2), 87–104., DOI 10.1159/ 000371806 Dekkers, OM, Hammer, S, de Keizer, RJ, Roelfsema, F, Schutte, PJ, Smit, JW, et al. The natural course of non-functioning pituitary macroadenomas. Eur J Endocrinol, 2007, 156, 2), 217–24., DOI 10.1530/eje.1.02334 Melmed, S. Pathogenesis of pituitary tumors. Nat Rev Endocrinol, 2011, 7, 5), 257–66., DOI 10.1038/nrendo.2011.40 Charles, NA, Holland, EC, Gilbertson, R, Glass, R, and Kettenmann, H. The brain tumor microenvironment. Glia, 2012, 60, 3), 502–14., DOI 10.1002/glia. 21264 Reiman, JM, Kmieciak, M, Manjili, MH, and Knutson, KL. Tumor immunoediting and immunosculpting pathways to cancer progression. Semin Cancer Biol, 2007, 17, 4), 275–87., DOI 10.1016/j.semcancer.2007. 06.009 Singh, NJ, and Schwartz, RH. Primer: mechanisms of immunologic tolerance. Nat Clin Pract Rheumatol, 2006, 2, 1), 44–52., DOI 10.1038/ncprheum0049 Dunn, GP, Dunn, IF, and Curry, WT. Focus on TILs: prognostic significance of tumor infiltrating lymphocytes in human glioma. Cancer Immun, 2007, 7, 12. Lampson, LA. Brain tumor immunotherapy: an immunologist’s perspective. J Neurooncol, 2003, 64, 1-2), 3–11., DOI 10.1007/BF02700015 Jacobs, JF, Idema, AJ, Bol, KF, Nierkens, S, Grauer, OM, Wesseling, P, et al. Regulatory T cells and the PD-L1/PD-1 pathway mediate immune suppression in malignant human brain tumors. Neuro-oncology, 2009, 11, 4), 394–402., DOI 10.1215/15228517-2008-104 Heshmati, HM, Kujas, M, Casanova, S, Wollan, PC, Racadot, J, Van Effenterre, R, et al. Prevalence of lymphocytic infiltrate in 1400 pituitary adenomas. Endocr J, 1998, 45, 3), 357–61., DOI 10.1507/endocrj.45.357 Rossi, ML, Jones, NR, Esiri, MM, Havas, L, al Izzi, M, and Coakham, HB. Mononuclear cell infiltrate and HLA-Dr expression in 28 pituitary adenomas. Tumori, 1990, 76, 6), 543–7., DOI 10.1177/030089169007600605 Ma, L, Li, G, Su, Y, He, Q, Zhang, C, and Zhang, J. The soluble major histocompatibility complex class I-related chain A protein reduced NKG2D expression on natural killer and T cells from patients with prolactinoma and non-secreting pituitary adenoma. J Clin Neurosci, 2010, 17, 2), 241–7., DOI 10. 1016/j.jocn.2009.05.023 Lu, JQ, Adam, B, Jack, AS, Lam, A, Broad, RW, and Chik, CL. Immune cell infiltrates in pituitary adenomas: more macrophages in larger adenomas and more T cells in growth hormone adenomas. Endocr Pathol, 2015, 26, 3), 263–72., DOI 10.1007/s12022-015-9383-6 Wei, J, Gabrusiewicz, K, and Heimberger, A. The controversial role of microglia in malignant gliomas. Clin Dev Immunol, 2013, 2013, 285246., DOI 10.1155/2013/285246 Zamarron, BF, and Chen, W. Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci., 2011, 7, 5), 651–8., DOI 10. 7150/ijbs.7.651 Butz, H, Liko, I, Czirjak, S, Igaz, P, Korbonits, M, Racz, K, et al. MicroRNA profile indicates downregulation of the TGFβ pathway in sporadic nonfunctioning pituitary adenomas. Pituitary, 2011, 14, 2), 112–24., DOI 10. 1007/s11102-010-0268-x Meij, BP, Lopes, MB, Ellegala, DB, Alden, TD, and Laws, ER, Jr. The long-term significance of microscopic dural invasion in 354 patients with pituitary adenomas treated with transsphenoidal surgery. J Neurosurg, 2002, 96, 2), 195–208., DOI 10.3171/jns.2002.96.2.0195 Erreni, M, Mantovani, A, and Allavena, P. Tumor-associated macrophages, TAM, and inflammation in colorectal cancer. Cancer Microenviron, 2011, 4, 2), 141–54., DOI 10.1007/s12307-010-0052-5 Sato, E, Olson, SH, Ahn, J, Bundy, B, Nishikawa, H, Qian, F, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/ regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA, 2005, 102, 51), 18538–43., DOI 10.1073/pnas. 0509182102 Melero, I, Rouzaut, A, Motz, GT, and Coukos, G. T-cell and NK-cell infiltration into solid tumors: a key limiting factor for efficacious cancer immunotherapy. Cancer Discov, 2014, 4, 5), 522–6., DOI 10.1158/2159-8290. CD-13-0985 Lohr, J, Ratliff, T, Huppertz, A, Ge, Y, Dictus, C, Ahmadi, R, et al. Effector T-cell infiltration positively impacts survival of glioblastoma patients and is impaired by tumor-derived TGF-β. Clin Cancer Res., 2011, 17, 13), 4296–308., DOI 10.1158/1078-0432.CCR-10-2557 Orhan, A, Vogelsang, RP, Andersen, MB, Madsen, MT, Holmich, ER, Raskov, H, et al. The prognostic value of tumour-infiltrating lymphocytes in pancreatic cancer: a systematic review and meta-analysis. Eur J Cancer, 2020, 132, 71–84., DOI 10.1016/j.ejca.2020.03.013 Wang, P, Wang, T, Yang, Y, Yu, C, Liu, N, and Yan, C. Detection of programmed death ligand 1 protein and CD8+ lymphocyte infiltration in plurihormonal pituitary adenomas. Medicine, 2017, 96, 49), e9056., DOI 10. 1097/MD.0000000000009056 Alsaab, HO, Sau, S, Alzhrani, R, Tatiparti, K, Bhise, K, Kashaw, SK, et al. PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol, 2017, 8, 561., DOI 10.3389/fphar.2017.00561 Hazrati, SM, Aghazadeh, J, Mohtarami, F, Abouzari, M, and Rashidi, A. Immunotherapy of prolactinoma with a T helper 1 activator adjuvant and autoantigens: a case report. Neuroimmunomodulation, 2006, 13, 4), 205–8., DOI 10.1159/000100405 Vivier, E, Tomasello, E, Baratin, M, Walzer, T, and Ugolini, S. Functions of natural killer cells. Nat Immunol, 2008, 9, 5), 503–10., DOI 10.1038/ni1582 Halama, N, Braun,M, Kahlert, C, Spille, A, Quack, C, Rahbari, N, et al. Natural killer cells are scarce in colorectal carcinoma tissue despite high levels of chemokines and cytokines. Clin Cancer Res, 2011, 17, 4), 678–89., DOI 10.1158/ 1078-0432.CCR-10-2173 Cantoni, C, Huergo-Zapico, L, Parodi, M, Pedrazzi, M, Mingari, MC, Moretta, A, et al. NK cells, tumor cell transition, and tumor progression in solid malignancies: new hints for NK-based immunotherapy? J Immunol Res, 2016, 2016, 4684268., DOI 10.1155/2016/4684268 Levy, EM, Roberti, MP, and Mordoh, J. Natural killer cells in human cancer: from biological functions to clinical applications. J Biomed Biotechnol, 2011, 2011, 676198., DOI 10.1155/2011/676198 Wennerberg, E, Kremer, V, Childs, R, and Lundqvist, A. CXCL10-induced migration of adoptively transferred human natural killer cells toward solid tumors causes regression of tumor growth in vivo. Cancer Immunol Immunother, 2015, 64, 2), 225–35., DOI 10.1007/s00262-014-1629-5 Wang, HY, and Wang, RF. Regulatory T cells and cancer. Curr Opin Immunol, 2007, 19, 2), 217–23., DOI 10.1016/j.coi.2007.02.004 Rodi, M, Dimisianos, N, de Lastic, AL, Sakellaraki, P, Deraos, G, Matsoukas, J, et al. Regulatory cell populations in relapsing-remitting multiple sclerosis, RRMS, patients: effect of disease activity and treatment regimens. Int JMol Sci, 2016, 17, 9)., DOI 10.3390/ijms17091398 Chen, C, Chen, D, Zhang, Y, Chen, Z, Zhu, W, Zhang, B, et al. Changes of CD4+CD25+FOXP3+ and CD8+CD28− regulatory T cells in non-small cell lung cancer patients undergoing surgery. Int Immunopharmacol, 2014, 18, 2), 255–61., DOI 10.1016/j.intimp.2013.12.004 Karagoz, B, Bilgi, O, Gumus, M, Erikci, AA, Sayan, O, Turken, O, et al. CD8+CD28− cells and CD4+CD25+ regulatory T cells in the peripheral blood of advanced stage lung cancer patients. Med Oncol, 2010, 27, 1), 29–33., DOI 10. 1007/s12032-008-9165-9 Chen, C, Chen, Z, Chen, D, Zhang, B, Wang, Z, and Le, H. Suppressive effects of gemcitabine plus cisplatin chemotherapy on regulatory T cells in nonsmall-cell lung cancer. J Int Med Res, 2015, 43, 2), 180–7., DOI 10.1177/0300060514561504 Wu, M, Chen, X, Lou, J, Zhang, S, Zhang, X, Huang, L, et al. Changes in regulatory T cells in patients with ovarian cancer undergoing surgery: preliminary results. Int Immunopharmacol, 2017, 47, 244–50., DOI 10.1016/j.intimp.2017.04.004 Tanikawa, T, Wilke, CM, Kryczek, I, Chen, GY, Kao, J, Nunez, G, et al. Interleukin-10 ablation promotes tumor development, growth, and metastasis. Cancer Res, 2012, 72, 2), 420–9., DOI 10.1158/0008-5472.CAN-10-4627 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 598887 EP 598895 DI 10.3389/pore.2021.598887 PG 9 ER PT J TI Autophagy Plays a Role in the CUL4A-Related Poor Prognosis of Intrahepatic Cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE autophagy; CUL4A; intrahepatic cholangiocarcinoma; prognosis; LC3II ID autophagy; CUL4A; intrahepatic cholangiocarcinoma; prognosis; LC3II AB CUL4A regulate the termination of autophagy in a physical process. However, the relationship between CUL4A and autophagy in cancer is unclear. We retrospectively investigated 99 intrahepatic cholangiocarcinoma (iCCA) cases. Whole sections were used for immunohistochemical analysis for p62, and LC3B expression. Q-score was defined as the sum of the labeling intensity and proportion. The cut-off point for immunoreactivity was set. CUL4A was overexpressed in cell lines and autophagy reflux was compared after manipulation. The iCCA cases with CUL4A overexpression had significantly higher prevalence of intact activated autophagy (42.4 vs. 15.2%; p = 0.003), which was significantly associated with advance tumor stage (34.1%vs. 15.4%; p = 0.032), less extensive necrosis (8.3 vs. 49.3%; p < 0.001), and shortened disease-free survival (mean, 19.6 vs. 65.5months, p = 0.015). In vitro, iCCA cells with CUL4A overexpression significantly increased LC3II level as compared to the cells under basal condition. Although both cell types showed intact autophagy with increased LC3II expression after bafilomycin A1 treatment, the accumulation of LC3II was higher in CUL4A-overexpressing cells. CUL4A overexpression increased the proliferation of cells as compared with control cells. After treatment with bafilomycin A1, proliferation was inhibited in both cell types, but the effects were more prominent in the cells overexpressing CUL4A. CUL4A promotes autophagy, and exhibits significantly higher autophagic flux which affects the proliferation of iCCA cells; these effects correlated with advance tumor stage and poor prognosis. Thus, targeting autophagy may be potentially therapeutic in iCCA. CR Sharma, P., and Nag, A., 2014). CUL4A ubiquitin ligase: a promising drug target for cancer and other human diseases. Open Biol. 4, 130217., DOI 10.1098/rsob. 130217 Nag, A., Bagchi, S., and Raychaudhuri, P., 2004). Cul4A physically associates with MDM2 and participates in the proteolysis of p53. Cancer Res. 64, 8152–8155., DOI 10.1158/0008-5472.CAN-04-2598 Chen, L. C., Manjeshwar, S., Lu, Y., Moore, D., Ljung, B. M., Kuo, W. L., et al., 1998). The human homologue for the Caenorhabditis elegans cul-4 gene is amplified and overexpressed in primary breast cancers. Cancer Res. 58, 3677–3683. Yasui, K., Arii, S., Zhao, C., Imoto, I., Ueda, M., Nagai, H., et al., 2002). TFDP1, CUL4A, and CDC16 identified as targets for amplification at 13q34 in hepatocellular carcinomas. Hepatology 35, 1476–1484., DOI 10.1053/jhep.2002. 33683 Hung, M. S., Mao, J. H., Xu, Z., Yang, C. T., Yu, J. S., Harvard, C., et al., 2011). Cul4A is an oncogene in malignant pleural mesothelioma. J. Cell Mol. Med. 15, 350–358., DOI 10.1111/j.1582-4934.2009.00971.x Ren, S., Xu, C., Cui, Z., Yu, Y., Xu, W., Wang, F., et al., 2012). Oncogenic CUL4A determines the response to thalidomide treatment in prostate cancer. J. Mol. Med. 90, 1121–1132., DOI 10.1007/s00109-012-0885-0 Melchor, L., Saucedo-Cuevas, L. P., Munoz-Repeto, I., Rodriguez-Pinilla, S. M., Honrado, E., Campoverde, A., et al., 2009). Comprehensive characterization of the DNA amplification at 13q34 in human breast cancer reveals TFDP1 and CUL4A as likely candidate target genes. Breast Cancer Res. 11, R86., DOI 10.1186/ bcr2456 Wang, Y., Wen, M., Kwon, Y., Xu, Y., Liu, Y., Zhang, P., et al., 2014). CUL4A induces epithelial-mesenchymal transition and promotes cancer metastasis by regulating ZEB1 expression. Cancer Res. 74, 520–531., DOI 10.1158/0008-5472. CAN-13-2182 Liu, T. T., You, H. L., Weng, S. W., Wei, Y. C., Eng, H. L., and Huang, W. T., 2015). Recurrent amplification at 13q34 targets at CUL4A, IRS2, and TFDP1 as an independent adverse prognosticator in intrahepatic cholangiocarcinoma. Plos One. 10, e0145388., DOI 10.1371/journal.pone.0145388 Huang, G., Liu, T. T., Weng, S. W., You, H. L., Wei, Y. C., Chen, C. H., et al., 2017). CUL4A overexpression as an independent adverse prognosticator in intrahepatic cholangiocarcinoma. BMC Cancer. 17, 395., DOI 10.1186/s12885- 017-3389-z Singh, S. S., Vats, S., Chia, A. Y., Tan, T. Z., Deng, S., Ong, M. S., et al., 2018). Dual role of autophagy in hallmarks of cancer. Oncogene 37, 1142–1158., DOI 10.1038/s41388-017-0046-6 Choi, A. M., Ryter, S. W., and Levine, B., 2013). Autophagy in human health and disease. N. Engl. J. Med. 368, 651–662., DOI 10.1056/NEJMc1303158 Rabinowitz, J. D., and White, E., 2010). Autophagy and metabolism. Science 330, 1344–1348., DOI 10.1126/science.1193497 Antonioli, M., Albiero, F., Nazio, F., Vescovo, T., Perdomo, A. B., Corazzari, M., et al., 2014). AMBRA1 interplay with cullin E3 ubiquitin ligases regulates autophagy dynamics. Dev. Cell 31, 734–746., DOI 10.1016/j.devcel.2014.11.013 Schlafli, A. M., Berezowska, S., Adams, O., Langer, R., and Tschan, M. P., 2015). Reliable LC3 and p62 autophagy marker detection in formalin fixed paraffin embedded human tissue by immunohistochemistry. Eur. J. Histochem. 59, 2481., DOI 10.4081/ejh.2015.2481 Falasca, L., Torino, F., Marconi, M., Costantini, M., Pompeo, V., Sentinelli, S., et al., 2015). AMBRA1 and SQSTM1 expression pattern in prostate cancer. Apoptosis 20, 1577–1586., DOI 10.1007/s10495-015-1176-3 Liu, J. L., Chen, F. F., Lung, J., Lo, C. H., Lee, F. H., Lu, Y. C., et al., 2014). Prognostic significance of p62/SQSTM1 subcellular localization and LC3B in oral squamous cell carcinoma. Br. J. Cancer 111, 944–954., DOI 10.1038/bjc. 2014.355 Niklaus, M., Adams, O., Berezowska, S., Zlobec, I., Graber, F., Slotta- Huspenina, J., et al., 2017). Expression analysis of LC3B and p62 indicates intact activated autophagy is associated with an unfavorable prognosis in colon cancer. Oncotarget 8, 54604–54615., DOI 10.18632/oncotarget.17554 Li, J., Yang, B., Zhou, Q., Wu, Y., Shang, D., Guo, Y., et al., 2013). Autophagy promotes hepatocellular carcinoma cell invasion through activation of epithelial-mesenchymal transition. Carcinogenesis 34, 1343–1351., DOI 10. 1093/carcin/bgt063 Kim, Y. H., Baek, S. H., Kim, E. K., Ha, J. M., Jin, S. Y., Lee, H. S., et al., 2016). Uncoordinated 51-like kinase 2 signaling pathway regulates epithelialmesenchymal transition in A549 lung cancer cells. FEBS Lett. 590, 1365–1374., DOI 10.1002/1873-3468.12172 Qu, X., Yu, J., Bhagat, G., Furuya, N., Hibshoosh, H., Troxel, A., et al., 2003). Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Invest. 112, 1809–20., DOI 10.1172/JCI20039 Yue, Z., Jin, S., Yang, C., Levine, A. J., and Heintz, N., 2003). Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc. Natl. Acad. Sci. USA. 100, 15077–15082., DOI 10.1073/pnas.2436255100 Lazova, R., Camp, R. L., Klump, V., Siddiqui, S. F., Amaravadi, R. K., and Pawelek, J. M., 2012). Punctate LC3B expression is a common feature of solid tumors and associated with proliferation, metastasis, and poor outcome. Clin. Cancer Res. 18, 370–379., DOI 10.1158/1078-0432.CCR-11- 1282 Peng, Y. F., Shi, Y. H., Ding, Z. B., Ke, A. W., Gu, C. Y., Hui, B., et al., 2013). Autophagy inhibition suppresses pulmonary metastasis of HCC in mice via impairing anoikis resistance and colonization of HCC cells. Autophagy 9, 2056–2068., DOI 10.4161/auto.26398 Peng, Y. F., Shi, Y. H., Shen, Y. H., Ding, Z. B., Ke, A. W., Zhou, J, et al., 2013). Promoting colonization in metastatic HCC cells by modulation of autophagy. PloS One 8, e74407., DOI 10.1371/journal.pone.0074407 Mowers, E. E., Sharifi, M. N., and Macleod, K. F., 2017). Autophagy in cancer metastasis. Oncogene 36, 1619–1630., DOI 10.1038/onc.2016.333 Han, X., Fang, Z., Wang, H., Jiao, R., Zhou, J., and Fang, N., 2016). CUL4A functions as an oncogene in ovarian cancer and is directly regulated by miR- 494. Biochem. Biophys. Res. Commun. 480, 675–681., DOI 10.1016/j.bbrc.2016. 10.114 Yu, R., Cai, L., Chi, Y., Ding, X., and Wu, X., 2018). miR377 targets CUL4A and regulates metastatic capability in ovarian cancer. Int. J. Mol. Med. 41, 3147–3156., DOI 10.3892/ijmm.2018.3540 Hung, M. S., Chen, I. C., You, L., Jablons, D. M., Li, Y. C., Mao, J. H., et al., 2016). Knockdown of cullin 4A inhibits growth and increases chemosensitivity in lung cancer cells. J. Cell Mol. Med. 20, 1295–1306., DOI 10.1111/jcmm.12811 Hung, M. S., Chen, I. C., You, L., Jablons, D. M., Li, Y. C., Mao, J. H., et al., 2015). Knockdown of Cul4A increases chemosensitivity to gemcitabine through upregulation of TGFBI in lung cancer cells. Oncol. Rep. 34, 3187–3195., DOI 10.3892/or.2015.4324 Sui, X., Chen, R., Wang, Z., Huang, Z., Kong, N., Zhang, M., et al., 2013). Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis. 4, e838., DOI 10.1038/cddis.2013.350 Copeland, R. A., Solomon, M. E., and Richon, V. M., 2009). Protein methyltransferases as a target class for drug discovery. Nat. Rev. Drug Discov. 8, 724–732., DOI 10.1038/nrd2974 Hsieh, Y. Y., Lo, H. L., and Yang, P.M., 2016). EZH2 inhibitors transcriptionally upregulate cytotoxic autophagy and cytoprotective unfolded protein response in human colorectal cancer cells. Am. J. Cancer Res. 6, 1661–1680. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 602714 EP 602721 DI 10.3389/pore.2021.602714 PG 8 ER PT J AU Shen, GJ Shen, J Teng, YR Wang, BL Zhao, HW Wang, ChQ AF Shen, Guo Jian Shen, Jun Teng, Yue Rong Wang, Bo Lin Zhao, He Wen Wang, Chuan Qin TI High GP73 Expression Correlates with Poor Response to Neoadjuvant Chemotherapy and Survival in Gastric Cancer: A Tissue Microarray Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE GP73; gastric cancer; prognosis; neoadjuvant chemotherapy; pathological response ID GP73; gastric cancer; prognosis; neoadjuvant chemotherapy; pathological response AB Golgi protein 73 (GP73) is a type II Golgi transmembrane protein which is overexpressed in several cancers, however, its role in gastric cancer is still unclear. The aim of this study is to investigate if high GP73 expression is associated with pathological tumor response to neoadjuvant chemotherapy and prognosis for patients with gastric cancer. A total of 348 patients with gastric cancer, who had undergone surgery between 1999 and 2011 were retrospectively reviewed, GP73 expression was examined in tumor tissues using tissue microarray and the correlations between its expression and pathological response to neoadjuvant chemotherapy as well as patients prognosis were analyzed. We found that GP73 expression was not associated with clinicopathologic features including tumor size, differentiation and TNM stage. High expression of GP73 was associated with less pathological tumor response to neoadjuvant chemotherapy and poor survival in gastric cancer, multivariate analysis showed GP73 expression was an independent predictive factor for pathological response to neoadjuvant chemotherapy and for prognosis in patients with gastric cancer. Our results suggest that GP73 expression correlates with the effect of neoadjuvant chemotherapy and is a promising biomarker to identify patients with poor prognosis. C1 [Shen, Guo Jian] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical OncologyHangzhou, China. [Shen, Jun] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical OncologyHangzhou, China. [Teng, Yue Rong] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical OncologyHangzhou, China. [Wang, Bo Lin] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical OncologyHangzhou, China. [Zhao, He Wen] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical OncologyHangzhou, China. [Wang, Chuan Qin] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical OncologyHangzhou, China. RP Wang, ChQ (reprint author), Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Surgical Oncology, Hangzhou, China. EM wqctiger@zju.edu.cn CR Jemal A, Siegel R, Xu J, Ward E Cancer statistics, 2010. CA: A Cancer J Clinicians, 2010). 60(5):277–300., DOI 10.3322/caac.20073 ChenW, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA: A Cancer J Clinicians, 2016). 66(2):115–32., DOI 10.3322/caac.21338 Paoletti X, Paoletti X, Oba K, Burzykowski T, Michiels S, Ohashi Y, et al. Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis. JAMA, 2010). 303(17):1729–37., DOI 10.1001/jama.2010.534 Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJH, Nicolson M, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med, 2006). 355(1):11–20., DOI 10.1056/nejmoa055531 Giralt J, Eraso A, Armengol M, Rossello J, Majo J, Ares C, et al. Epidermal growth factor receptor is a predictor of tumor response in locally advanced rectal cancer patients treated with preoperative radiotherapy. Int J Radiat Oncol Biol Phys, 2002). 54(5):1460–5., DOI 10.1016/s0360-3016(02)03752-5 Kladney RD, Bulla GA, Guo L, Mason AL, Tollefson AE, Simon DJ, et al. GP73, a novel Golgi-localized protein upregulated by viral infection. Gene, 2000). 249(1-2):53–65., DOI 10.1016/s0378-1119(00)00136-0 Yang J, Li J, Dai W, Wang F, Shen M, Chen K, et al. Golgi protein 73 as a biomarker for hepatocellular carcinoma: a diagnostic meta-analysis. Exp Ther Med, 2015). 9(4):1413–20., DOI 10.3892/etm.2015.2231 Liang R, Liu Z, Piao X, ZuoM, Zhang J, Liu Z, et al. Research progress on GP73 in malignant tumors. OncoTargets Ther, 2018). 11:7417–21., DOI 10.2147/ott.s181239 Chen L-G, Wang H-J, Yao H-b., Guan T-P, Wu F, He X-J, et al. GP73 is downregulated in gastric cancer and associated with tumor differentiation. World J Surg Onc, 2013). 11(1):132., DOI 10.1186/1477-7819-11-132 Liu G, Zhang Y, He F, Li J, Wei X, Li Y, et al. Expression of GOLPH2 is associated with the progression of and poor prognosis in gastric cancer. Oncol Rep, 2014). 32(5):2077–85., DOI 10.3892/or.2014.3404 Mandard A-M, Dalibard F, Mandard J-C, Marnay J, Henry-Amar M, Petiot J-F, et al. Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Clinicopathol Correl Cancer, 1994). 73(11):2680–6., DOI 10. 1002/1097-0142(19940601)73:11<2680::aid-cncr2820731105>3.0.co;2-c Wang Q, Liu X, Zhou J, Huang Y, Zhang S, Shen J, et al. Ribonucleotide reductase large subunit M1 predicts poor survival due to modulation of proliferative and invasive ability of gastric cancer. PLoS One, 2013). 8(7): e70191., DOI 10.1371/journal.pone.0070191 Zhou Y, Li L,Hu L, Peng T Golgi phosphoprotein 2, GOLPH2/GP73/GOLM1, interacts with secretory clusterin. Mol Biol Rep, 2011). 38(3):1457–62., DOI 10. 1007/s11033-010-0251-7 Ai N, Liu W, Li Z-G, Ji H, Li B, Yang G High expression of GP73 in primary hepatocellular carcinoma and its function in the assessment of transcatheter arterial chemoembolization. Oncol Lett, 2017). 14(4):3953–8., DOI 10.3892/ol. 2017.6697 Bao YX, Yang Y, Zhao HR, Mao R, Xiao L, Zhang YF, et al. Clinical significance and diagnostic value of Golgi-protein 73 in patients with early-stage primary hepatocellular carcinoma. Diagn Pathol, 2013). 35(7):505–8., DOI 10.1186/ 1746-1596-8-197 Zhang S, Ge W, Zou G, Yu L, Zhu Y, Li Q, et al. MiR-382 targets GOLM1 to inhibit metastasis of hepatocellular carcinoma and its down-regulation predicts a poor survival. Am J Cancer Res, 2018). 8(1):120–31., DOI 10. 18632/oncotarget.13037 Jiang K, Li W, Shang S, Sun L, Guo K, Zhang S, et al. Aberrant expression of Golgi protein 73 is indicative of a poor outcome in hepatocellular carcinoma. Oncol Rep, 2016). 35(4):2141–50., DOI 10.3892/or.2016.4601 Sun Y, Yang H, Mao Y, Xu H, Zhang J, Li G, et al. Increased Golgi protein 73 expression in hepatocellular carcinoma tissue correlates with tumor aggression but not survival. J Gastroenterol Hepatol, 2011). 26(7):1207–12., DOI 10.1111/j. 1440-1746.2011.06733.x Wang LB, Teng RY, Jiang ZN, Hu WX, Dong MJ, Yuan XM, et al. Clinicopathologic variables predicting tumor response to neoadjuvant chemotherapy in patients with locally advanced gastric cancer. J Surg Oncol, 2012). 105(3):293–6., DOI 10.1002/jso.22085 Ye J-Z, Yan S-m., Yuan C-l., Wu H-n., Zhang J-y., Liu Z-H, et al. GP73 level determines chemotherapeutic resistance in human hepatocellular carcinoma cells. J Cancer, 2018). 9(2):415–23., DOI 10.7150/jca.19185 Zhou ZP, Wang LP, Hong ZS, Qiu CZ, Wang MZ, Chen ZX, et al. Silencing GOLPH3 gene expression reverses resistance to cisplatin in HT29 colon cancer cells via multiple signaling pathways. Int J Oncol, 2018). 53(3):1183–92., DOI 10. 3892/ijo.2018.4471 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 603838 EP 603843 DI 10.3389/pore.2021.603838 PG 6 ER PT J TI The Clinicopathological Significance of YAP/TAZ Expression in Hepatocellular Carcinoma with Relation to Hypoxia and Stemness SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE hepatocellular carcinoma; Hippo pathway; hypoxia; stemness; immunohistochemistry ID hepatocellular carcinoma; Hippo pathway; hypoxia; stemness; immunohistochemistry AB Background/Aims: Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) activation has been implicated in hepatocarcinogenesis and hepatic progenitor cell differentiation, and hypoxia has been shown to induce nuclear translocation of YAP in cancer cells. Here, we aimed to investigate the relationship between hypoxia, YAP and TAZ expression and stemness-related marker expression in human hepatocellular carcinomas (HCCs) and its clinical implications. Methods: Immunohistochemical stains were performed on tissue microarrays from 305 surgically resected HCCs, and the expression status of YAP and TAZ were correlated with CAIX, stemness markers (K19, EpCAM) and epithelial-mesenchymal transition (EMT)-related markers (uPAR, ezrin). The clinicopathological significance of YAP/TAZ expression was analyzed with relation to CAIX expression status. Results: YAP and TAZ expression were seen in 13.4 and 4.3% of HCCs, respectively. YAP/TAZ-positive HCCs frequently demonstrated higher serum AFP levels, microvascular invasion, advanced tumor stage, increased proliferative activity and expression of stemness- and EMT-related markers, CAIX, p53 and Smad2/3 (p < 0.05, all). Interestingly, YAP/TAZ-positivity was associated with microvascular invasion, higher serum AFP levels, stemness and EMT-related marker expression only in tumors expressing CAIX (p < 0.05, all), while these associations were not seen in CAIXnegative HCCs. Conclusions: YAP/TAZ expression is associated with vascular invasion, stemness and EMT in HCCs with hypoxia marker expression. The effect of Hippo signaling pathway deregulation in HCC may depend on the presence or absence of a hypoxic microenvironment, and hypoxia marker expression status should be taken into account when considering the use of YAP/TAZ as markers of aggressive biologic behavior in HCC. CR Kim, H, Choi, GH, Na, DC, Ahn, EY, Kim, GI, Lee, JE, et al. Human hepatocellular carcinomas with “Stemness”-related marker expression: keratin 19 expression and a poor prognosis. Hepatology., 2011, 54(5): 1707–17., DOI 10.1002/hep.24559 Boyault, S, Rickman, DS, de Reynies, A, Balabaud, C, Rebouissou, S, Jeannot, E, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology., 2007, 45(1):42–52., DOI 10.1002/hep.21467 Calderaro, J, Couchy, G, Imbeaud, S, Amaddeo, G, Letouze, E, Blanc, JF, et al. Histological subtypes of hepatocellular carcinoma are related to gene mutations and molecular tumour classification. J Hepatol., 2017, 67(4):727–38., DOI 10. 1016/j.jhep.2017.05.014 Hoshida, Y, Nijman, SM, Kobayashi, M, Chan, JA, Brunet, JP, Chiang, DY, et al. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Res., 2009, 69(18):7385–92., DOI 10. 1158/0008-5472.CAN-09-1089 Zhao, B, Lei, QY, and Guan, KL. The Hippo-YAP pathway: new connections between regulation of organ size and cancer. Curr Opin Cell Biol., 2012, 20(6): 638–46., DOI 10.1016/j.ceb.2008.10.001 Mo, JS, Yu, FX, Gong, R, Brown, JH, and Guan, KL. Regulation of the Hippo- YAP pathway by protease-activated receptors, PARs). Genes Dev., 2012, 26(19):2138–43., DOI 10.1101/gad.197582.112 Johnson, R, and Halder, G. The two faces of Hippo: targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat Rev Drug Discov., 2014, 13(1):63–79., DOI 10.1038/nrd4161 Yu, FX, Zhao, B, and Guan, KL. Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell., 2015, 163(4):811–28., DOI 10.1016/j.cell.2015.10.044 Zhou, D, Conrad, C, Xia, F, Park, JS, Payer, B, Yin, Y, et al. Mst1 and Mst2 maintain hepatocyte quiescence and suppress hepatocellular carcinoma development through inactivation of the Yap1 oncogene. Cancer Cell., 2009, 16(5):425–38., DOI 10.1016/j.ccr.2009.09.026 Kang, W, Tong, JH, Chan, AW, Lee, TL, Lung, RW, Leung, PP, et al. Yesassociated protein 1 exhibits oncogenic property in gastric cancer and its nuclear accumulation associates with poor prognosis. Clin Cancer.(2011, Res. 17(8):2130–9., DOI 10.1158/1078-0432.CCR-10-2467 Van Haele, M, Moya, IM, Karaman, R, Rens, G, Snoeck, J, Govaere, O, et al. YAP and TAZ heterogeneity in primary liver cancer: an analysis of its prognostic and diagnostic role. Int J Mol Sci., 2019, 20(3):638., DOI 10.3390/ijms20030638 Wang, L, Shi, S, Guo, Z, Zhang, X, Han, S, Yang, A, et al. Overexpression of YAP and TAZ is an independent predictor of prognosis in colorectal cancer and related to the proliferation and metastasis of colon cancer cells. PLoS One., 2013, 8(6):e65539., DOI 10.1371/journal.pone.0065539 Wang, Y, Dong, Q, Zhang, Q, Li, Z, Wang, E, and Qiu, X. Overexpression of yes-associated protein contributes to progression and poor prognosis of nonsmall- cell lung cancer. Cancer Sci., 2010, 101(5):1279–85., DOI 10.1111/j.1349- 7006.2010.01511.x Xu, MZ, Yao, TJ, Lee, NP, Ng, IO, Chan, YT, Zender, L, et al. Yes-associated protein is an independent prognostic marker in hepatocellular carcinoma. Cancer., 2009, 115(19):4576–85., DOI 10.1002/cncr.24495 Kim,GJ, Kim,H, and Park, YN. Increased expression of Yes-associated protein 1 in hepatocellular carcinoma with stemness and combined hepatocellularcholangiocarcinoma. PLoS One., 2013, 8(9):e75449., DOI 10.1371/journal.pone. 0075449 Vaupel, P, and Mayer, A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev., 2007, 26(2):225–39., DOI 10.1007/ s10555-007-9055-1 McDonald, PC, Winum, JY, Supuran, CT, and Dedhar, S. Recent developments in targeting carbonic anhydrase IX for cancer therapeutics. Oncotarget., 2012, 3(1):84–97., DOI 10.18632/oncotarget.422 Semenza, GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer., 2003, 3(10):721–32., DOI 10.1038/nrc1187nrc1187 Rhee, H, Nahm, JH, Kim, H, Choi, GH, Yoo, JE, Lee, HS, et al. Poor outcome of hepatocellular carcinoma with stemness marker under hypoxia: resistance to transarterial chemoembolization. Mod Pathol., 2016, 29(9):1038–49., DOI 10. 1038/modpathol.2016.111 Wei, H, Xu, Z, Liu, F, Wang, F, Wang, X, Sun, X, et al. Hypoxia induces oncogene yes-associated protein 1 nuclear translocation to promote pancreatic ductal adenocarcinoma invasion via epithelial-mesenchymal transition. Tumour Biol., 2017, 39(5):1010428317691684., DOI 10.1177/ 1010428317691684 Lee, Y, Park, H, Lee, H, Cho, JY, Yoon, YS, Choi, YR, et al. The clinicopathological and prognostic significance of the gross classification of hepatocellular carcinoma. J Pathol Transl Med., 2018, 52(2):85–92., DOI 10. 4132/jptm.2017.11.13 Kang, I, Jang, M, Lee, JG, Han, DH, Joo, DJ, Kim, KS, et al. Subclassification of microscopic vascular invasion in hepatocellular carcinoma. Ann Surg., 2020)., DOI 10.1097/SLA.0000000000003781 Yan, L, Cai, Q, and Xu, Y. Hypoxic conditions differentially regulate TAZ and YAP in cancer cells. Arch Biochem Biophys., 2014, 562:31–6., DOI 10.1016/j.abb. 2014.07.024 Oh, SH, Swiderska-Syn, M, Jewell, ML, Premont, RT, and Diehl, AM. Liver regeneration requires Yap1-TGFbeta-dependent epithelial-mesenchymal transition in hepatocytes. J Hepatol., 2018, 69(2):359–67., DOI 10.1016/j. jhep.2018.05.008 Seok, JY, Na, DC, Woo, HG, Roncalli, M, Kwon, SM, Yoo, JE, et al. A fibrous stromal component in hepatocellular carcinoma reveals a cholangiocarcinoma-like gene expression trait and epithelial-mesenchymal transition. Hepatology., 2012, 55(6):1776–86., DOI 10.1002/hep.25570 Guo, Y, Pan, Q, Zhang, J, Xu, X, Liu, X, Wang, Q, et al. Functional and clinical evidence that TAZ is a candidate oncogene in hepatocellular carcinoma. J Cell Biochem., 2015, 116(11):2465–75., DOI 10.1002/jcb.25117 Lee, K, Lee, KB, Jung, HY, Yi, NJ, Lee, KW, Suh, KS, et al. The correlation between poor prognosis and increased yes-associated protein 1 expression in keratin 19 expressing hepatocellular carcinomas and cholangiocarcinomas. BMC Cancer., 2017, 17(1):441., DOI 10.1186/ s12885-017-3431-1 Sun, PL, Kim, JE, Yoo, SB, Kim, H, Jin, Y, Jheon, S, et al. Cytoplasmic YAP expression is associated with prolonged survival in patients with lung adenocarcinomas and epidermal growth factor receptor tyrosine kinase inhibitor treatment. Ann Surg Oncol., 2014, 21(Suppl. 4):S610–8., DOI 10. 1245/s10434-014-3715-5 Rothzerg, E, Ingley, E, Mullin, B, Xue, W, Wood, D, and Xu, J. The Hippo in the room: targeting the Hippo signalling pathway for osteosarcoma therapies. J Cell Physiol., 2020, 236:1606–15., DOI 10.1002/jcp.29967 Perra, A, Kowalik, MA, Ghiso, E, Ledda-Columbano, GM, Di Tommaso, L, Angioni, MM, et al., 2014). YAP activation is an early event and a potential therapeutic target in liver cancer development. J Hepatol. 61(5):1088–96., DOI 10.1016/j.jhep.2014.06.033 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 604600 EP 604607 DI 10.3389/pore.2021.604600 PG 8 ER PT J AU Wojtukiewicz, M Mysliwiec, M Matuszewska, E Sulkowski, S Zimnoch, L Politynska, B Wojtukiewicz, A Tucker, S Honn, K AF Wojtukiewicz, Z. Marek Mysliwiec, Marta Matuszewska, Elwira Sulkowski, Stanislaw Zimnoch, Lech Politynska, Barbara Wojtukiewicz, M. Anna Tucker, C. Stephanie Honn, V. Kenneth TI Heterogeneous Expression of Proangiogenic and Coagulation Proteins in Gliomas of Different Histopathological Grade SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE angiogenesis; VEGF; bFGF; D-dimers; blood coagulation; fibrin; tissue factor; glial tumors 3 ID angiogenesis; VEGF; bFGF; D-dimers; blood coagulation; fibrin; tissue factor; glial tumors 3 AB Brain gliomas are characterized by remarkably intense invasive growth and the ability to create new blood vessels. Angiogenesis is a key process in the progression of these tumors. Coagulation and fibrinolysis factors play a role in promoting angiogenesis. The aim of the study was to evaluate the expression of proangiogenic proteins (VEGF and bFGF) and hemostatic proteins (TF, fibrinogen, fibrin, D-dimers) associated with neoplastic cells and vascular endothelial cells in brain gliomas of various degrees of malignancy. Immunohistochemical tests were performed using the ABC method with the use of mono- and polyclonal antibodies. The obtained results indicated that both neoplastic cells and vascular endothelial cells in gliomas of various degrees of malignancy are characterized by heterogeneous expression of proteins of the hemostatic system and angiogenesis markers. The strongest expression of proangiogenic factors and procoagulant factors was demonstrated in gliomas of higher-grade malignancy. C1 [Wojtukiewicz, Z. Marek] Medical University of Bialystok, Department of OncologyBialystok, Poland. [Mysliwiec, Marta] Medical University of Bialystok, Department of OncologyBialystok, Poland. [Matuszewska, Elwira] Comprehensive Cancer Center of Bialystok, Department of Clinical OncologyBialystok, Poland. [Sulkowski, Stanislaw] Medical University of Bialystok, Department of General PathomorphologyBialystok, Poland. [Zimnoch, Lech] Medical University of Bialystok, Department of General PathomorphologyBialystok, Poland. [Politynska, Barbara] Medical University of Bialystok, Department of Philosophy and Human PsychologyBialystok, Poland. [Wojtukiewicz, M. Anna] Medical University of Bialystok, Department of Philosophy and Human PsychologyBialystok, Poland. [Tucker, C. Stephanie] Wayne State University, Bioactive Lipids Research Program, Department of Pathology-School of MedicineDetroit, MI, USA. [Honn, V. Kenneth] Wayne State University, Department of ChemistryDetroit, MI, USA. RP Wojtukiewicz, M (reprint author), Medical University of Bialystok, Department of Oncology, Bialystok, Poland. EM mzwojtukiewicz@gmail.com CR Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med, 1971). 285:1182–6., DOI 10.1056/nejm197108122850711 Folkman J. What is the evidence that tumors are angiogenesis dependent? J Nat Cancer Inst, 1990). 82:4–7., DOI 10.1093/jnci/82.1.4 Wesseling P, van der Laak JAWM, Link M, Teepen HLJM, Ruiter DJ. Quantitative analysis of microvascular changes in diffuse astrocytic neoplasms with increasing grade of malignancy. Hum Pathol, 1998). 29: 352–8., DOI 10.1016/s0046-8177(98)90115-0 Leon SP, Folkerth RD, Black PM. Microvessel density is a prognostic indicator for patients with astroglial brain tumors. Cancer, 1996). 77:362–72., DOI 10.1002/, sici)1097-0142(19960115)77:2<362::aid-cncr20>3.0.co;2-z Li VW, Yu Ab C, Folkman J, Scott RM, McL Black P, Folkerth R, et al. Microvessel count and cerebrospinal fluid basic fibroblast growth factor in children with brain tumours. The Lancet, 1994). 344:82–6., DOI 10.1016/s0140- 6736(94)91280-7 Jain RK, di Tomaso E, Duda DG, Loeffler JS, Sorensen AG, Batchelor TT. Angiogenesis in brain tumours. Nat Rev Neurosci, 2007). 8:610–22., DOI 10.1038/ nrn2175 Rak J,Mitsuhashi Y, Bayko L, Filmus J, Shirasawa S, Sasazuki T, et al. Mutant ras oncogenes upregulate VEGF/VPF expression: implications for induction and inhibition of tumor angiogenesis. Cancer Res, 1995). 55:4575–80. Ahir BK, Engelhard HH, Lakka SS. Tumor development and angiogenesis in adult brain tumor: glioblastoma. Mol Neurobiol, 2020). 57:2461–78., DOI 10. 1007/s12035-020-01892-8 Johnson LD. Hypoxia: a key regulator of angiogenesis in cancer. Cancer Metast Rev, 2007). 26:281–90., DOI 10.1007/s10555-007-9066-y Dvorak HF. Angiogenesis: update 2005. J Thromb Haemost, 2005). 3:1835–42., DOI 10.1111/j.1538-7836.2005.01361.x Seghezzi G, Patel S, Ren CJ, Gualandris A, Pintucci G, Robbins ES, et al. Fibroblast growth factor-2, FGF-2, induces vascular endothelial growth factor, VEGF, expression in the endothelial cells of forming capillaries: an autocrine mechanism contributing to angiogenesis. J Cel Biol, 1998). 141:1659–73., DOI 10.1083/jcb.141.7.1659 Murakami M, Simons M. Fibroblast growth factor regulation of neovascularization. Curr Opin Hematol, 2008). 15:215–20., DOI 10.1097/ moh.0b013e3282f97d98 Batchelor TT, Reardon DA, de Groot JF, Wick W, Weller M. Antiangiogenic therapy for glioblastoma: current status and future prospects. Clin Cancer Res, 2014). 20:5612–9., DOI 10.1158/1078-0432.ccr-14-0834 McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Franklin RA, Montalto G, et al. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascade inhibitors: how mutations can result in therapy resistance and how to overcome resistance. Oncotarget, 2012). 3:1068–111., DOI 10.18632/oncotarget.659 Grover SP,Mackman N. Tissue FactorAn essential mediator of hemostasis and trigger of thrombosis. Arterioscler Thromb Vasc Biol, 2018). 38:709–25., DOI 10. 1161/atvbaha.117.309846 Ornstein DL, Meehan KR, Zacharski LR. The coagulation system as a target for the treatment of human gliomas. Semin Thromb Hemost, 2002). 28:21–7., DOI 10.1055/s-2002-20561 Versteeg HH, Schaffner F, Kerver M, Petersen HH, Ahamed J, Felding- Habermann B, et al. Inhibition of tissue factor signaling suppresses tumor growth. Blood, 2008). 111:190–9., DOI 10.1182/blood-2007-07-101048 Rao LVM. Tissue Factor as a tumor procoagulant. Cancer Metastasis Rev, 1992). 11:249–266., DOI 10.1007/bf01307181 Wojtukiewicz MZ, Sierko E, Rak J. Contribution of the hemostatic system to angiogenesis in cancer. Semin Thromb Hemost, 2004). 30:5–20., DOI 10.1055/s- 2004-822967 Wojtukiewicz MZ, Zacharski LR, Memoli VA, Kisiel W, Kudryk BJ, Rousseau SM, et al. Abnormal regulation of coagulation/fibrinolysis in small cell carcinoma of the lung. Cancer, 1990). 65:481–5., DOI 10.1002/1097- 0142(19900201)65:3<481::aid-cncr2820650318>3.0.co;2-u Wojtukiewicz MZ, Zacharski LR, Memoli VA, Kisiel W, Kudryk BJ, Rousseau SM, et al. Fibrinogen-fibrin transformation in situ in renal cell carcinoma. Anticancer Res, 1990). 10:579–82. Wojtukiewicz MZ, Zacharski LR, Memoli VA, Kisiel W, Kudryk BJ, Rousseau SM, et al. Malignant melanoma: interaction with coagulation and fibrinolysis pathways in situ. Am J Clin Pathol, 1990). 93:516–21., DOI 10.1093/ajcp/93. 4.516 Wojtukiewicz MZ, Sierko E, Zacharski LR, Zimnoch L, Kudryk B, Kisiel W. Tissue factor-dependent coagulation activation and impaired fibrinolysis in situ in gastric cancer. Semin Thromb Hemost, 2003). 29:291–300., DOI 10.1055/ s-2003-40967 Wojtukiewicz MZ, Rucinska M, Zimnoch L, Jaromin J, Piotrowski Z, Rozanska-Kudelska M, et al. Expression of prothrombin fragment 1+2 in cancer tissue as an indicator of local activation of blood coagulation. Thromb Res, 2000). 97:335–42., DOI 10.1016/s0049-3848(99)00169-3 Hsu SM, Raine L, Fanger H. Use of avidin-biotin-peroxidase complex, ABC, in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody, PAP, procedures. J Histochem Cytochem, 1981). 29:577–80., DOI 10. 1177/29.4.6166661 Wojtukiewicz MZ, Zacharski LR, Rucinska M, Zimnoch L, Jaromin J, Rozanska-Kudelska M, et al. Expression of tissue factor and tissue factor pathway inhibitor in situ in laryngeal carcinoma. Thromb Haemost, 1999). 82: 1659–62. Hirsch FR, Varella-Garcia M, Bunn PA, Jr, Di Maria MV, Veve R, Bremnes RM, et al. Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. Jco, 2003). 21:3798–807., DOI 10.1200/jco.2003.11.069 Pirker R, Pereira JR, von Pawel J, Krzakowski M, Ramlau R, Park K, et al. EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: analysis of data from the phase 3 FLEX study. Lancet Oncol, 2012). 13:33–42., DOI 10.1016/s1470- 2045(11)70318-7 Constantini V, Zacharski LR. The role of fibrin in tumor metastasis. Cancer Metast Rev, 1992). 11:283–90., DOI 10.1007/BF01307183 Devy L, Blacher S, Grignet-Debrus C, Bajou K, Masson V, Gerard RD, et al. The pro- or antiangiogenic effect of plasminogen activator inhibitor 1 is dose dependent. FASEB j., 2002). 16:147–54., DOI 10.1096/fj.01-0552com Siemann DW, Chaplin DJ, Horsman MR. Realizing the potential of vascular targeted therapy: the rationale for combining vascular disrupting agents and anti-angiogenic agents to treat cancer. Cancer Invest, 2017). 35:519–34., DOI 10. 1080/07357907.2017.1364745 Och W, Mariak Z, Smolka M, Badowski J, Koziorowski M. [Vascular endothelial growth factor expression in cerebral neoplasms]. Neurol Neurochir Pol, 2001). 35:1071–9. Plate KH, Breier G, Weich HA, Mennel HD, Risau W. Vascular endothelial growth factor and glioma angiogenesis: coordinate induction of VEGF receptors, distribution of VEGF protein and possibleIn vivo regulatory mechanisms. Int J Cancer, 1994). 59:520–9., DOI 10.1002/ijc.2910590415 Carrol RS, Zhang J, Melnick MB, Maruyama T, McL-Black P. KDR activation in astrocytic neoplasm. Cancer, 1999). 86:1335–41., DOI 10.1002/(sici)1097- 0142(19991001)86:7<1335::aid-cncr32>3.0.co;2-z Holash J, Maisonpierre PC, Comton D, Boland P, Alexander CR, Zagzag D, et al. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science, 1999). 284:1994–8., DOI 10.1126/science.284.5422.1994 Poech M, Farion R, Hiou A, Le Bas JF, Pasquier B, Remy C. Immunohistochemical study of VEGF, angiopoietin 2 and their receptors in the neovascularization following microinjection of C6 glioma cells into rat brain. Anticancer Res, 2002). 22(4):2147-51. Jubb AM, Pham TQ, Hanby AM, Frantz GD, Peale FV, Wu TD, et al. Expression of vascular endothelial growth factor, hypoxia inducible factor 1 , and carbonic anhydrase IX in human tumours. J Clin Pathol, 2004). 57: 504–12., DOI 10.1136/jcp.2003.012963 Turner HE, Harris AL, Melmed S, Wass JAH. Angiogenesis in endocrine tumors. Endocr Rev, 2003). 24:600–32., DOI 10.1210/er.2002-0008 Dabrosin C. Variability of vascular endothelial growth factor in normal human breast tissue in vivo during the menstrual cycle. J Clin Endocrinol Metab, 2003). 88:2695–8., DOI 10.1210/jc.2002-021584 Maeda K, Chung Y-s., Ogawa Y, Kang S-M, Ogawa M, Sawada T, et al. Prognostic value of vascular endothelial growth factor expression in gastric carcinoma. Cancer, 1996). 77:858–63., DOI 10.1002/(sici)1097-0142(19960301, 77:5<858::aid-cncr8>3.0.co;2-a Salven P, Heikkila P, Joensuu H. Enhanced expression of vascular endothelial growth factor in metastatic melanoma. Br J Cancer, 1997). 76:930–4., DOI 10. 1038/bjc.1997.486 Weidner N, Semple JP, Welch WR, Folkman J. Tumor angiogenesis and metastasis - correlation in invasive breast carcinoma. N Engl JMed, 1991). 324: 1–8., DOI 10.1056/nejm199101033240101 Lizarraga-Verdugo E, Avendano-Felix M, Bermudez M, Ramos-Payan R, Perez-Plasencia C, Aguilar-Medina M. Cancer stem cells and its role in angiogenesis and vasculogenic mimicry in gastrointestinal cancers. Front Oncol, 2020). 10:413., DOI 10.3389/fonc.2020.00413 Xin M, Yin-Sheng C, Fu-Rong C, Shao-Yan X, Zhong-Ping C. Glioblastomas stem cell differentiation into endothelial cells evidenced through live-cell imaging. Neuro-Oncology, 2017). 19(8):1109–18., DOI 10.1093/neuonc/nox016 Mercurio A. VEGF/Neuropilin signaling in cancer stem cells. Ijms, 2019). 20: 490., DOI 10.3390/ijms20030490 Elaimy AL, Amante JJ, Zhu LJ, Wang M, Walmsley CS, FitzGerald TJ, et al. The VEGF receptor neuropilin 2 promotes homologous recombination by stimulating YAP/TAZ mediated Rad51 expression. Proc Natl Acad Sci U S A, 2019). 9(116):14174–80., DOI 10.1073/pnas.1821194116 Ke LD, Shi YX, Im SA, Chen X, Yung WK. The relevance of cell proliferation, vascular endothelial growth factor, and basic fibroblast growth factor production to angiogenesis and tumorigenicity in human glioma cell lines. Clin Cancer Res, 2000). 6:2562–72. Landriscina M, Cassano A, Ratto C, Longo R, Ippoliti M, Palazzotti B, et al. Quantitative analysis of basic fibroblast growth factor and vascular endothelial growth factor in human colorectal cancer. Br J Cancer, 1998). 78:765–70., DOI 10.1038/bjc.1998.575 Takahashi Y, Ellis LM, Ohta T, Mai M. Angiogenesis in poorly differentiated medullary carcinoma of the stomach. Surg Today, 1998). 28:367–72., DOI 10. 1007/s005950050143 Brem S, Tsanaclis AMC, Gately S, Gross JL, Herblin WF. Immunolocalization of basic fibroblast growth factor to the micro vasculature of human brain tumors. Cancer, 1992). 70:2673–80., DOI 10.1002/1097-0142(19921201)70: 11<2673::aid-cncr2820701118>3.0.co;2-f Takahashi JA, Fukumoto M, Igarashi K, Oda Y, Kikuchi H, Hatanaka M. Correlation of basic fibroblast growth factor expression levels with the degree of malignancy and vascularity in human gliomas. J Neurosurg, 1992). 76: 792–8., DOI 10.3171/jns.1992.76.5.0792 Zagzag D, Miller DC, Sato Y, Rifkin DB, Burstein DE. Immunohistochemical localization of basic fibroblast growth factor in astrocytomas. Cancer Res, 1990). 50:7393–8. Stan AC, Nemati MN, Pietsch T, Walter GF, Dietz H. In vivo inhibition of angiogenesis and growth of the human U-87 malignant glial tumor by treatment with an antibody against basic fibroblast growth factor. J Neurosurg, 1995). 82:1044–52., DOI 10.3171/jns.1995.82.6.1044 Hamada K, Kuratsu J, Saitoh Y, Takeshima H, Nishi T, Ushio Y. Expression of tissue factor correlates with grade of malignancy in human glioma. Cancer, 1996). 77:1877–83., DOI 10.1002/(SICI)1097-0142(19960501)77:9<1877::AIDCNCR18> 3.0.CO;2-X Takano S, Tsuboi K, Tomono Y, Mitsui Y, Nose T. Tissue factor, osteopontin, alphavbeta3 integrin expression in microvasculature of gliomas associated with vascular endothelial growth factor expression. Br J Cancer, 2000). 82:1967–73., DOI 10.1054/bjoc.2000.1150 Hamada K, Kuratsu J-i., Saitoh Y, Takeshima H, Nishi T, Ushio Y. Expression of tissue factor correlates with grade of malignancy in human glioma. Cancer, 1996). 77:1877–83., DOI 10.1002/(sici)1097-0142(19960501)77:9<1877::aidcncr18> 3.0.co;2-x Morrissey JH, Neuenschwander PF, Huang Q, McCallum CD, Su B, Johnson AE. Factor VIIa-tissue factor: functional importance of protein-membrane interactions. Thromb Haemost, 1997). 78:112–6. Guan M, Jin J, Su B, Liu WW, Lu Y. Tissue factor expression and angiogenesis in human glioma. Clin Biochem, 2002). 35:321–5., DOI 10.1016/s0009-9120(02, 00312-0 Abe K, Shoji M, Chen J, Bierhaus A, Danave I, Micko C, et al. Regulation of vascular endothelial growth factor production and angiogenesis by the cytoplasmic tail of tissue factor. Proc Natl Acad Sci, 1999). 96:8663–8., DOI 10.1073/pnas.96.15.8663 Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med, 1995). 1:27–30., DOI 10.1038/nm0195-27 Zhang Y, Deng Y, Luther T, Muller M, Ziegler R,Waldherr R, et al. Tissue factor controls the balance of angiogenic and antiangiogenic properties of tumor cells in mice. J Clin Invest, 1994). 94:1320–7., DOI 10.1172/ jci117451 Bardos H, Molnar P, Csecsei G, Adany R. Fibrin deposition in primary and metastatic human brain tumours. Blood Coagul Fibrinolysis, 1996). 7:536–48. Wojtukiewicz MZ, Sierko E, Klementt P, Rak J. The hemostatic system and angiogenesis in malignancy. Neoplasia, 2001). 3:371–84., DOI 10.1038/sj.neo. 7900184 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 605017 EP 605023 DI 10.3389/pore.2021.605017 PG 7 ER PT J AU Kamseng, P Siriboonpiputtana, T Puavilai, T Chuncharunee, S Paisooksantivatana, K Chareonsirisuthigul, T Junking, M Chiraphapphaiboon, W Yenchitsomanus, Pth Rerkamnuaychoke, B AF Kamseng, Parin Siriboonpiputtana, Teerapong Puavilai, Teeraya Chuncharunee, Suporn Paisooksantivatana, Karan Chareonsirisuthigul, Takol Junking, Mutita Chiraphapphaiboon, Wannasiri Yenchitsomanus, Pa-thai Rerkamnuaychoke, Budsaba TI Targeting UCHL1 Induces Cell Cycle Arrest in High-Risk Multiple Myeloma with t(4;14) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE UCHL1; t(4; 14) multiple myeloma; cell cycle regulation; biomarker; expression profile ID UCHL1; t(4; 14) multiple myeloma; cell cycle regulation; biomarker; expression profile AB Multiple myeloma (MM) patients considered to be at high cytogenetic risk commonly fail to respond to standard treatment. A thorough understanding of the molecular mechanism of MM development is, therefore, needed. We endeavored to explore the transcriptional signature among different subgroups of newly diagnosed MM using gene chip-based expression microarray. Bone marrow samples of 15 newly diagnosed Thai MM patients were included. The chromosomal translocation t(4;14) was the most frequently identified genetic alteration in the high-risk subgroup. Cluster analysis from expression profiling demonstrated that high-riskMMhave a distinctly different expression pattern compared to standard-risk patients. The most significant differentially expressed gene was UCHL1. Functional enrichment analysis by Gene Set Enrichment Analysis, FUNRICH, and Gene Ontology Panther pathway revealed the gene sets involved in cell cycle control to be enriched in the t(4;14) high-risk group. Interestingly, among the well-established downstream targets of UCHL1, only CCND2 was significantly expressed in the t(4;14) high-risk group. Suppression of UCHL1 protein level by LDN-5744 inhibitor could arrest the cell cycle in G1 phase in cell lines. These findings shed light on the molecular mechanism of UCHL1 in t(4;14) high-risk MM and support the evidence that alteration of the UCHL1 pathway may play a role in the pathogenesis of high-risk MM. C1 [Kamseng, Parin] Mahidol University, Faculty of Medicine Ramathibodi Hospital, Department of PathologyBangkok, Thailand. [Siriboonpiputtana, Teerapong] Mahidol University, Faculty of Medicine Ramathibodi Hospital, Department of PathologyBangkok, Thailand. [Puavilai, Teeraya] Mahidol University, Faculty of Medicine Ramathibodi Hospital, Department of MedicineBangkok, Thailand. [Chuncharunee, Suporn] Mahidol University, Faculty of Medicine Ramathibodi Hospital, Department of MedicineBangkok, Thailand. [Paisooksantivatana, Karan] Mahidol University, Faculty of Medicine Ramathibodi Hospital, Department of PathologyBangkok, Thailand. [Chareonsirisuthigul, Takol] Mahidol University, Faculty of Medicine Ramathibodi Hospital, Department of PathologyBangkok, Thailand. [Junking, Mutita] Mahidol University, Faculty of Medicine Siriraj Hospital, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT)Bangkok, Thailand. [Chiraphapphaiboon, Wannasiri] Mahidol University, Faculty of Medicine Siriraj Hospital, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT)Bangkok, Thailand. [Yenchitsomanus, Pa-thai] Mahidol University, Faculty of Medicine Siriraj Hospital, Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT)Bangkok, Thailand. [Rerkamnuaychoke, Budsaba] Mahidol University, Faculty of Medicine Ramathibodi Hospital, Department of PathologyBangkok, Thailand. RP Rerkamnuaychoke, B (reprint author), Mahidol University, Faculty of Medicine Ramathibodi Hospital, Department of Pathology, Bangkok, Thailand. EM budsaba.rer@mahidol.ac.th CR Palumbo A, Anderson K. Multiple myeloma. N Engl J Med, 2011, 364(11): 1046–60., DOI 10.1056/NEJMra1011442 Committee of The Ramathibodi Hospital Cancer Center. Ramathibodi cancer report 2017. Bangkok, Thailand: Ramathibodi Comprehensive Cancer Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 2017). Bazarbachi AH, Al Hamed R, Malard F, Harousseau JL, Mohty M. Relapsed refractory multiple myeloma: a comprehensive overview. Leukemia, 2019, 33(10):2343–57., DOI 10.1038/s41375-019-0561-2 Bittner M, Meltzer P, Chen Y, Jiang Y, Seftor E, Hendrix M, et al. Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature, 2000, 406(6795):536–40., DOI 10.1038/35020115 Zhan F, Hardin J, Kordsmeier B, Bumm K, Zheng M, Tian E, et al. Global gene expression profiling of multiple myeloma, monoclonal gammopathy of undetermined significance, and normal bone marrow plasma cells. Blood, 2002, 99(5):1745–57., DOI 10.1182/blood.v99.5.1745 Meissner T, Seckinger A, Reme T, Hielscher T, Mohler T, Neben K, et al. Gene expression profiling in multiple myeloma–reporting of entities, risk, and targets in clinical routine. Clin Cancer Res, 2011, 17(23):7240–7., DOI 10.1158/1078- 0432.Ccr-11-1628 Weinhold N, Heuck CJ, Rosenthal A, Thanendrarajan S, Stein CK, Van Rhee F, et al. Clinical value of molecular subtyping multiple myeloma using gene expression profiling. Leukemia, 2016, 30(2):423–30., DOI 10.1038/leu.2015.309 Lohr JG, Stojanov P, Carter SL, Cruz-Gordillo P, Lawrence MS, Auclair D, et al. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy. Cancer Cell, 2014, 25(1):91–101., DOI 10.1016/j.ccr.2013. 12.015 Chng WJ, Chung TH, Kumar S, Usmani S, Munshi N, Avet-Loiseau H, et al. Gene signature combinations improve prognostic stratification of multiple myeloma patients. Leukemia, 2016, 30(5):1071–8., DOI 10.1038/leu.2015.341 Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple myeloma. Nat Rev Cancer, 2012, 12(5):335–48., DOI 10.1038/nrc3257 Hussain S, Bedekovics T, Chesi M, Bergsagel PL, Galardy PJ. UCHL1 is a biomarker of aggressive multiple myeloma required for disease progression. Oncotarget, 2015, 6(38):40704–18., DOI 10.18632/oncotarget.5727 Larsen CN, Price JS, Wilkinson KD. Substrate binding and catalysis by ubiquitin C-terminal Hydrolases: identification of two active site residues. Biochemistry, 1996, 35(21):6735–44., DOI 10.1021/bi960099f Setsuie R, Wada K. The functions of UCH-L1 and its relation to neurodegenerative diseases. Neurochem Int, 2007, 51(2-4):105–11., DOI 10. 1016/j.neuint.2007.05.007 Andersson FI, Werrell EF, McMorran L, Crone WJ, Das C, Hsu ST, et al. The effect of Parkinson’s-disease-associated mutations on the deubiquitinating enzyme UCH-L1. J Mol Biol, 2011, 407(2):261–72., DOI 10.1016/j.jmb.2010. 12.029 Mani A, Gelmann EP. The ubiquitin-proteasome pathway and its role in cancer. J Clin Oncol, 2005, 23(21):4776–89., DOI 10.1200/JCO.2005.05.081 Hussain S, Zhang Y, Galardy PJ. DUBs and cancer: the role of deubiquitinating enzymes as oncogenes, non-oncogenes and tumor suppressors. Cell Cycle, 2009, 8(11):1688–97., DOI 10.4161/cc.8.11.8739 Bedekovics T, Hussain S, Feldman AL, Galardy PJ. UCH-L1 is induced in germinal center B cells and identifies patients with aggressive germinal center diffuse large B-cell lymphoma. Blood, 2016, 127(12):1564–74., DOI 10.1182/ blood-2015-07-656678 Zhong J, Zhao M, Ma Y, Luo Q, Liu J, Wang J, et al. UCHL1 acts as a colorectal cancer oncogene via activation of the beta-catenin/TCF pathway through its deubiquitinating activity. Int J MolMed, 2012, 30(2):430–6., DOI 10.3892/ijmm. 2012.1012 Goto Y, Zeng L, Yeom CJ, Zhu Y, Morinibu A, Shinomiya K, et al. UCHL1 provides diagnostic and antimetastatic strategies due to its deubiquitinating effect on HIF-1alpha. Nat Commun, 2015, 6:6153., DOI 10.1038/ncomms7153 Hussain S, Bedekovics T, Liu Q, Hu W, Jeon H, Johnson SH, et al. UCH-L1 bypasses mTOR to promote protein biosynthesis and is required for MYCdriven lymphomagenesis in mice. Blood, 2018, 132(24):2564–74., DOI 10.1182/ blood-2018-05-848515 Kabuta T, Mitsui T, Takahashi M, Fujiwara Y, Kabuta C, Konya C, et al. Ubiquitin C-terminal hydrolase L1, UCH-L1, acts as a novel potentiator of cyclin-dependent kinases to enhance cell proliferation independently of its hydrolase activity. J Biol Chem, 2013, 288(18):12615–26., DOI 10.1074/jbc. M112.435701 Sonneveld P, Avet-Loiseau H, Lonial S, Usmani S, Siegel D, Anderson KC, et al. Treatment ofmultiple myeloma with high-risk cytogenetics: a consensus of the International Myeloma Working Group. Blood, 2016, 127(24):2955–62., DOI 10.1182/blood-2016-01-631200 Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res, 2018, 47(D1):D607–D613., DOI 10.1093/nar/gky1131 Gong B, Cao Z, Zheng P, Vitolo OV, Liu S, Staniszewski A, et al. Ubiquitin hydrolase uch-L1 rescues β-amyloid-induced decreases in synaptic function and contextual memory. Cell, 2006, 126(4):775–88., DOI 10.1016/j.cell.2006. 06.046 Chng WJ, Dispenzieri A, Chim CS, Fonseca R, Goldschmidt H, Lentzsch S, et al. IMWG consensus on risk stratification in multiple myeloma. Leukemia, 2014, 28(2):269–77., DOI 10.1038/leu.2013.247 Phornsarayuth P, KorKiasakul V, Puavilai T, Boonyawat K, Niparuck P, Chuncharunee S, et al. Molecular cytogenetic aberrations in Thai patients with multiple myeloma. Genomics Genet, 2016, 9:25–30., DOI 10.14456/gag. 2016.4 Zheng S, Qiao G, Min D, Zhang Z, Lin F, Yang Q, et al. Heterogeneous expression and biological function of ubiquitin carboxy-terminal hydrolase-L1 in osteosarcoma. Cancer Lett, 2015, 359(1):36–46., DOI 10.1016/j.canlet.2014. 12.001 Luo Y, He J, Yang C, Orange M, Ren X, Blair N, et al. UCH-L1 promotes invasion of breast cancer cells through activating Akt signaling pathway. J Cell Biochem, 2018, 119(1):691–700., DOI 10.1002/jcb.26232 Tan YY, Zhou HY, Wang ZQ, Chen SD. Endoplasmic reticulum stress contributes to the cell death induced by UCH-L1 inhibitor. Mol Cell Biochem, 2008, 318(1-2):109–15., DOI 10.1007/s11010-008-9862-x Hu Y, Chen W, Chen S, Huang Z. Cytogenetic abnormality in patients with multiple myeloma analyzed by fluorescent in situ hybridization. Onco Targets Ther, 2016, 9:1145–9., DOI 10.2147/OTT.S95818 Yoshida T, RiM, Fujinami H, Oshima Y, Tachita T, Marumo Y, et al. Impact of chromosomal abnormalities on the efficacy of lenalidomide plus dexamethasone treatment in patients with relapsed/refractory multiple myeloma. Int J Hematol, 2019, 110(2):228–36., DOI 10.1007/s12185-019-02669-z Sahota SS, Leo R, Hamblin TJ, Stevenson FK. Ig VH gene mutational patterns indicate different tumor cell status in human myeloma and monoclonal gammopathy of undetermined significance. Blood, 1996, 87(2):746–55., DOI 10.1182/blood.V87.2.746.bloodjournal872746 Walker BA, Leone PE, Jenner MW, Li C, Gonzalez D, Johnson DC, et al. Integration of global SNP-based mapping and expression arrays reveals key regions, mechanisms, and genes important in the pathogenesis of multiple myeloma. Blood, 2006, 108(5):1733–43., DOI 10.1182/blood- 2006-02-005496 Anderson K, Lutz C, van Delft FW, Bateman CM, Guo Y, Colman SM, et al. Genetic variegation of clonal architecture and propagating cells in leukaemia. Nature, 2011, 469(7330):356–61., DOI 10.1038/nature09650 Sun J, Wen X, Jin F, Li Y, Hu J, Sun Y. Bioinformatics analyses of differentially expressed genes associated with bisphosphonate-related osteonecrosis of the jaw in patients with multiple myeloma. OncoTargets Ther, 2015, 8:2681–8., DOI 10.2147/OTT.S88463 David CJ, Massague J. Contextual determinants of TGFβ action in development, immunity and cancer. Nat Rev Mol Cel Biol, 2018, 19(7): 419–35., DOI 10.1038/s41580-018-0007-0 Chattopadhyay S, Thomsen H, Yadav P, da Silva Filho MI, Weinhold N, Nothen MM, et al. Genome-wide interaction and pathway-based identification of key regulators in multiple myeloma. Commun Biol, 2019, 2(1):89., DOI 10. 1038/s42003-019-0329-2 Yan H, Zheng G, Qu J, Liu Y, Huang X, Zhang E, et al. Identification of key candidate genes and pathways in multiple myeloma by integrated bioinformatics analysis: YAN et al. J Cell Physiol, 2019, 234(12):23785–97., DOI 10.1002/jcp.28947 Akiyama M, Hideshima T, Shammas MA, Hayashi T, Hamasaki M, Tai YT, et al. Effects of oligonucleotide N3′→P5′ thio-phosphoramidate, GRN163, targeting telomerase RNA in human multiple myeloma cells. Cancer Res, 2003, 63(19):6187–94. Walker BA, Wardell CP, Melchor L, Hulkki S, Potter NE, Johnson DC, et al. Intraclonal heterogeneity and distinct molecular mechanisms characterize the development of t(4;14, and t(11;14, myeloma. Blood, 2012, 120(5):1077–86., DOI 10.1182/blood-2012-03-412981 Kim HJ, Kim YM, Lim S, Nam YK, Jeong J, Kim HJ, et al. Ubiquitin C-terminal hydrolase-L1 is a key regulator of tumor cell invasion and metastasis. Oncogene, 2009, 28(1):117–27., DOI 10.1038/onc.2008.364 Moore MD, Finnerty B, Gray KD, Hoda R, Liu YF, Soong L, et al. Decreased UCHL1 expression as a cytologic biomarker for aggressive behavior in pancreatic neuroendocrine tumors. Surgery, 2018, 163(1):226–31., DOI 10. 1016/j.surg.2017.04.040 Sanchez-Diaz PC, Chang JC, Moses ES, Dao T, Chen Y, Hung JY. Ubiquitin carboxyl-terminal esterase L1, UCHL1, is associated with stem-like cancer cell functions in pediatric high-grade glioma. PLoS One, 2017, 12(5):e0176879., DOI 10.1371/journal.pone.0176879 Lien HC, Wang CC, Lin CH, Lu YS, Huang CS, Hsiao LP, et al. Differential expression of ubiquitin carboxy-terminal hydrolase L1 in breast carcinoma and its biological significance. Hum Pathol, 2013, 44(9):1838–48., DOI 10.1016/ j.humpath.2013.02.006 Jin C, Yu W, Lou X, Zhou F, Han X, Zhao N, et al. UCHL1 is a putative tumor suppressor in ovarian cancer cells and contributes to cisplatin resistance. J Cancer, 2013, 4(8):662–70., DOI 10.7150/jca.6641 Hussain S, Foreman O, Perkins SL, Witzig TE, Miles RR, van Deursen J, et al. The de-ubiquitinase UCH-L1 is an oncogene that drives the development of lymphoma in vivo by deregulating PHLPP1 and Akt signaling. Leukemia, 2010, 24(9):1641–55., DOI 10.1038/leu.2010.138 Xiang T, Li L, Yin X, Yuan C, Tan C, Su X, et al. The ubiquitin peptidase UCHL1 induces G0/G1 cell cycle arrest and apoptosis through stabilizing p53 and is frequently silenced in breast cancer. PloS one, 2012, 7(1):e29783., DOI 10. 1371/journal.pone.0029783 Finnerty BM, Moore M, Verma A, Aronova A, Huang S, Edwards DP, et al. UCHL1 loss alters the cell-cycle in metastatic pancreatic neuroendocrine tumors. Endocr Relat Cancer, 2019, 26(4):411–23., DOI 10.1530/erc-18-0507 Bergsagel PL, Kuehl WM, Zhan F, Sawyer J, Barlogie B, Shaughnessy J, Jr. Cyclin D dysregulation: an early and unifying pathogenic event in multiple myeloma. Blood, 2005, 106(1):296–303., DOI 10.1182/blood-2005-01-0034 Mulligan G, Mitsiades C, Bryant B, Zhan F, Chng WJ, Roels S, et al. Gene expression profiling and correlation with outcome in clinical trials of the proteasome inhibitor bortezomib. Blood, 2007, 109(8):3177–88., DOI 10.1182/ blood-2006-09-044974 Liu Y, Lashuel HA, Choi S, Xing X, Case A, Ni J, et al. Discovery of inhibitors that elucidate the role of UCH-L1 activity in the H1299 lung cancer cell line. Chem Biol, 2003, 10(9):837–46., DOI 10.1016/j.chembiol.2003.08.010 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 606567 EP 606579 DI 10.3389/pore.2021.606567 PG 13 ER PT J AU Locsei, Z Sebestyen, K Sebestyen, Zs Feher, E Soltesz, D Musch, Z Mangel, CsL AF Locsei, Zoltan Sebestyen, Klara Sebestyen, Zsolt Feher, Eszter Soltesz, Dorottya Musch, Zoltan Mangel, Csaba Laszlo TI IMAT-IGRT Treatment with Simultaneous Integrated Boost as Dose Escalation for Patients with Cervical Cancer: A Single Institution, Prospective Pilot Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cervical cancer; SIB; IGRT; IMRT; dose escalation; toxicity 3 ID cervical cancer; SIB; IGRT; IMRT; dose escalation; toxicity 3 AB Purpose: The aim of this study was to introduce the simultaneous integrated boost (SIB) technique to assess the safety of replacement of the brachytherapy (BT) boost for ineligible patients with cervical cancer receiving radiochemotherapy (RCT). Methods: Fourteen patients were enrolled between 2015 and 2018. SIB was delivered using RapidArc technique at doses of 2.4 Gy per fraction during pelvic irradiation with 50.4/ 1.8 Gy in seven patients (to a total dose of 67.2 Gy) with limited volume disease. In 7 patients with a more advanced disease stage (>5 cm tumor, parametric invasion both sides), parametric boost therapy was added to the pelvic radiotherapy to a total dose of the macroscopic tumor of 79.2 Gy. All patients received simultaneous cisplatin-based chemotherapy for 5 cycles with a dosage of 40 mg/m2. We examined acute toxicity (CTCAE v4.1) and quality of life (EORTC QLQ30 and CX24). The tumor regression rate was evaluated with RECIST 1.1 after the first 3- to 4-months follow-up Magnetic Resonance Imaging (MRI) scan. We calculated the percentage of tumor regression rate and the local control during the follow-up period and evaluated the survival data. Results: Our patient data are presented at a median follow-up time of 24.5 months. During the treatment period, no grade 3 to 4 toxicity was observed. During the follow-up period, no late-onset toxicity was observed. The tumor regression rate at the first MRI scan was 95.31% on average. Disease free survival (DFS) during the median follow-up of 24 months was 98.6%. Conclusion: In patients with cervical cancer, the SIB technique is amenable as part of definitive RCT. Dose escalation with the SIB technique can be safely administered to cervical cancer patients during definitive RCT if BT is not feasible. However, further randomized clinical studies are needed to validate the method, so routine use of it cannot be recommended yet. C1 [Locsei, Zoltan] University of Pecs, Department of OncologyPecs, Hungary. [Sebestyen, Klara] University of Pecs, Department of OncologyPecs, Hungary. [Sebestyen, Zsolt] University of Pecs, Department of OncologyPecs, Hungary. [Feher, Eszter] Pecsi Diagnosztikai Kozpont Kft.Pecs, Hungary. [Soltesz, Dorottya] University of Pecs, Department of OncologyPecs, Hungary. [Musch, Zoltan] University of Pecs, Department of OncologyPecs, Hungary. [Mangel, Csaba Laszlo] University of Pecs, Department of OncologyPecs, Hungary. RP Locsei, Z (reprint author), University of Pecs, Department of Oncology, Pecs, Hungary. EM locsei.zoltan@pte.hu CR Kilic S, Cracchiolo B, Mahmoud O. Non-brachytherapy alternatives in cervical cancer radiotherapy: why not?. Appl Radiat Oncol, 2015). 4(4):11–7. Cibula D, Potter R, Planchamp F, Avall-Lundqvist E, Fischerova D, Haie Meder C, et al. The European society of gynaecological Oncology/European society for radiotherapy and Oncology/European society of Pathology guidelines for the management of patients with cervical cancer. Radiother Oncol, 2018). 127(3): 404–16., DOI 10.1016/j.radonc.2018.03.003 FollenM, Levenback CF, Iyer RB,Grigsby PW, Boss EA, Delpassand ES, et al. Imaging in cervical cancer. Cancer, 2003). 98:2028–38., DOI 10.1002/cncr. 11679 Rose PG, Adler LP, Rodriguez M, Faulhaber PF, Abdul-Karim FW, Miraldi F. Positron emission tomography for evaluating para-aortic nodal metastasis in locally advanced cervical cancer before surgical staging: a surgicopathologic study. J Clin Oncol, 1999). 17:41., DOI 10.1200/jco.1999.17.1.41 Narayan K, Hicks RJ, Jobling T, Bernshaw D, Mckenzie AF. A comparison of MRI and PET scanning in surgically staged loco-regionally advanced cervical cancer: potential impact on treatment. Int J Gynecol Cancer, 2001). 11:263–71., DOI 10.1046/j.1525-1438.2001.011004263.x Haie-Meder C, Potter R, Van Limbergen E, Briot E, De Brabandere M, Dimopoulos J, et al. Recommendations from Gynaecological, GYN, GECESTRO Working Groupq, I): concepts and terms in 3D image based 3D treatment planning in cervix cancer brachytherapy with emphasis on MRI assessment of GTV and CTV. Radiother Oncol, 2005). 74:235–45., DOI 10.1016/j. radonc.2004.12.015 Potter R, Georg P, Dimopoulos JCA, Grimm M, Berger D, Nesvacil N, et al. Clinical outcome of protocol based image, MRI, guided adaptive brachytherapy combined with 3D conformal radiotherapy with or without chemotherapy in patients with locally advanced cervical cancer. Radiother Oncol, 2011). 100: 116–23., DOI 10.1016/j.radonc.2011.07.012 Sturdza A, Potter R, Fokdal LU, Haie-Meder C, Potter R, Haie-Meder C, et al. Image guided brachytherapy in locally advanced cervical cancer: improved pelvic control and survival in RetroEMBRACE, a multicenter cohort study. Radiother Oncol, 2016). 120:428–33., DOI 10.1016/j.radonc.2016.03.011 Kirchheiner K, Nout RA, Tanderup K, Lindegaard JC, Westerveld H, Haie- Meder C, et al. Manifestation pattern of early-late vaginal morbidity after definitive radiation, chemo)therapy and image-guided adaptive brachytherapy for locally advanced cervical cancer: an analysis from the EMBRACE study. Int J Radiat Oncol Biol Phys, 2014). 89:88–95., DOI 10.1016/j.ijrobp.2014.01.032 Kirchheiner K, Potter R, Tanderup K, Lindegaard JC, Haie-Meder C, Petriˇc P, et al. Health-related quality of life in locally advanced cervical cancer patients after definitive chemoradiation therapy including image guided adaptive brachytherapy: an analysis from the EMBRACE study. Int J Radiat Oncol Biol Phys, 2016). 94:1088–98., DOI 10.1016/j.ijrobp.2015.12.363 Guy JB, Falk AT, Auberdiac P, Cartier L, Vallard A, Ollier E, et al. Dosimetric study of volumetric arc modulation with RapidArc and intensity-modulated radiotherapy in patients with cervical cancer and comparison with 3- dimensional conformal technique for definitive radiotherapy in patients with cervical cancer. Med Dosimetry, 2016). 41(1):9–14., DOI 10.1016/j. meddos.2015.06.002 Mazeron R, Gilmore J, Khodari W, Dumas I, Haie-Meder C. Locally advanced cervical cancer: should intensity-modulated radiotherapy replace brachytherapy. Cancer Radiother, 2011). 15(6-7):477–83., DOI 10.1016/j.canrad.2011.07.232 Guerrero M, Li XA, Ma L, Linder J, Deyoung C, Erickson B. Simultaneous integrated intensity-modulated radiotherapy boost for locally advanced gynecological cancer: radiobiological and dosimetric considerations. Int J Radiat Oncol Biol Phy, 2005). 62(3):933–9., DOI 10.1016/j.ijrobp.2004.11.040 Vandecasteele K, De Neve W, De Gersem W, Delrue L, Paelinck L, Makar A, et al. Intensity-Modulated Arc therapy with simultaneous integrated boost in the treatment of primary irresectable cervical cancer. Treatment planning, quality control, and clinical implementation. Strahlenther Onkol, 2009). 185(12):799–807., DOI 10.1007/s00066-009-1986-8 Wang X, Zhao Y, Shen Y, Shu P, Li Z, Bai S, et al. Long-term follow-up results of simultaneous integrated or late course accelerated boost with external beam radiotherapy to vaginal cuff for high risk cervical cancer patients after radical hysterectomy. BMC Cancer, 2015). 15:257., DOI 10. 1186/s12885-015-1248-3 Vandecasteele K, Makar A, Van den Broecke R, Delrue L, Denys H, Lambein K, et al. Intensity-modulated arc therapy with cisplatin as neo-adjuvant treatment for primary irresectable cervical cancer. Toxicity, tumour response and outcome. Strahlenther Onkol, 2012). 188(7):576–81., DOI 10. 1007/s00066-012-0097-0 OʼDonnell B, Shiao JC, Pezzi TA, Waheed N, Sharma S, Bonnen MD, et al. Stereotactic body radiation therapy, intensity-modulated radiation therapy, and brachytherapy boost modalities in invasive cervical cancer: a study of the national cancer data base. Int J Gynecol Cancer, 2018). 28(3):563–74., DOI 10. 1097/IGC.0000000000001200 Herrera FG, Callaway S, Delikgoz-Soykut E, Coskun M, Porta L, Meuwly JY, et al. Retrospective feasibility study of simultaneous integrated boost in cervical cancer using Tomotherapy: the impact of organ motion and tumor regression. Radiat Oncol, 2013). 8:5., DOI 10.1186/1748-717X-8-5 Morgenthaler J, Kohler C, Budach V, Sehouli J, Stromberger C, Besserer A, et al. Long-term results of robotic radiosurgery for non brachytherapy patients with cervical cancer. Strahlenther Onkol, 2020)., DOI 10.1007/s00066-020- 01685-x NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 608446 EP 608454 DI 10.3389/pore.2021.608446 PG 9 ER PT J AU Chen, H Lin, X Liu, H Huang, Ch Li, R Ai, J Wei, J Xiao, Sh AF Chen, Huoying Lin, Xiaoying Liu, Hongbo Huang, Cheng Li, Rong Ai, Jie Wei, Jiaxue Xiao, Shengjun TI HMGB1 Translocation is Associated with Tumor-Associated Myeloid Cells and Involved in the Progression of Fibroblastic Sarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE high-mobility group box 1 protein; translocation; sarcoma; tumor-associated macrophage; myeloid cell ID high-mobility group box 1 protein; translocation; sarcoma; tumor-associated macrophage; myeloid cell AB The morphological variability and genetic complexity of fibroblastic sarcoma makes its diagnosis and treatment a challenge. High-mobility group box 1 protein (HMGB1), which functions as a DNA chaperone and a prototypical damage-associated molecular pattern, plays a paradoxical role in cancer. However, the expression pattern and role of HMGB1 in fibroblastic sarcomas is ill defined. By immunostaining of 95 tissue microarray cores of fibroblastic sarcomas, HMGB1 was found to be expressed in most tumor tissues. Nuclear HMGB1 translocation to cytoplasm was observed both in tumor cells and vascular endothelial cells. A visible number of tumor-associated myeloid cells including CD68+ and CD163+ macrophages and CD33+ myeloid cells were also detected in most tumor tissues. HMGB1 translocation was not only associated with CD68, CD163, and CD33 density, but also with disease progression. These results imply that HMGB1, an important regulator of the tumor microenvironment, is associated with tumor-associated myeloid cells and involved in the progression of fibroblastic sarcomas; HMGB1 may serve as a promising prognostic biomarker and a potential therapeutic target for fibroblastic sarcoma. C1 [Chen, Huoying] Guangdong Provincial Emergency Hospital, Guangdong Second Provincial General Hospital, Prenatal Diagnosis CenterGuangzhou, China. [Lin, Xiaoying] Guangdong Provincial Emergency Hospital, Guangdong Second Provincial General Hospital, Prenatal Diagnosis CenterGuangzhou, China. [Liu, Hongbo] The Second Affiliated Hospital of Guilin Medical University, Department of Laboratory MedicineGuilin, China. [Huang, Cheng] Guangdong Provincial Emergency Hospital, Guangdong Second Provincial General Hospital, Prenatal Diagnosis CenterGuangzhou, China. [Li, Rong] Guangdong Provincial Emergency Hospital, Guangdong Second Provincial General Hospital, Prenatal Diagnosis CenterGuangzhou, China. [Ai, Jie] The Second Affiliated Hospital of Guilin Medical University, Department of Laboratory MedicineGuilin, China. [Wei, Jiaxue] Guangdong Provincial Emergency Hospital, Guangdong Second Provincial General Hospital, Prenatal Diagnosis CenterGuangzhou, China. [Xiao, Shengjun] The Second Affiliated Hospital of Guilin Medical University, Department of PathologyGuilin, China. RP Xiao, Sh (reprint author), The Second Affiliated Hospital of Guilin Medical University, Department of Pathology, Guilin, China. EM xiaoshengjun@glmc.edu.cn CR Cancer Research UK. Soft tissue sarcoma statistics, 2013): Available from: https://www.cancerresearchuk.org/health-professional/cancer-statistics/ statistics-by-cancer-type/soft-tissue-sarcoma, Accessed June 1st, 2020). Coffin CM, Alaggio R. Fibroblastic and myofibroblastic tumors in children and adolescents. Pediatr Dev Pathol, 2012, 15(1 Suppl. l):127–80., DOI 10.2350/10- 12-0944-PB.1 CDM Fletcher, JA Bridge, P Hogendoorn, F Mertens, editors. World Health Organization classification of tumours of soft tissue and bone. 4th ed. LyonBordeaux, France: IARC Press, 2013). Meyer M, Seetharam M. First-line therapy for metastatic soft tissue sarcoma. Curr Treat Options Oncol, 2019, 20(1):6., DOI 10.1007/s11864-019-0606-9 Sabolch A, FengM, Griffith K, Rzasa C, Gadzala L, Feng F, et al. Risk factors for local recurrence and metastasis in soft tissue sarcomas of the extremity. Am J Clin Oncol, 2012, 35(2):151–7., DOI 10.1097/COC.0b013e318209cd72 Chawla SP, Papai Z, Mukhametshina G, Sankhala K, Vasylyev L, Fedenko A, et al. First-line aldoxorubicin vs doxorubicin in metastatic or locally advanced unresectable soft-tissue sarcoma. JAMA Oncol, 2015, 1(9):1272–80., DOI 10. 1001/jamaoncol.2015.3101 Gupta S, Gouw L, Wright J, Chawla S, Pitt D, Wade M, et al. Phase II study of amrubicin, SM-5887), a synthetic 9-aminoanthracycline, as first line treatment in patients with metastatic or unresectable soft tissue sarcoma: durable response in myxoid liposarcoma with TLS-CHOP translocation. Invest New Drugs, 2016, 34(2):243–52., DOI 10.1007/s10637-016-0333-z Schoffski P, Chawla S,Maki RG, Italiano A, Gelderblom H, Choy E, et al. Eribulin versus dacarbazine in previously treated patients with advanced liposarcoma or leiomyosarcoma: a randomised, open-label, multicentre, phase 3 trial. The Lancet, 2016, 387(10028):1629–37., DOI 10.1016/S0140-6736(15)01283-0 Merchant MS,Wright M, Baird K, Wexler LH, Rodriguez-Galindo C, Bernstein D, et al. Phase I clinical trial of ipilimumab in pediatric patients with advanced solid tumors. Clin Cancer Res, 2016, 22(6):1364–70., DOI 10.1158/1078-0432. CCR-15-0491 Tawbi HA, Burgess M, Bolejack V, Van Tine BA, Schuetze SM, Hu J, et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma, SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol, 2017, 18(11):1493–501., DOI 10.1016/S1470-2045(17)30624-1 Campana L, Bosurgi L, Rovere-Querini P. HMGB1: a two-headed signal regulating tumor progression and immunity. Curr Opin Immunol, 2008, 20(5):518–23., DOI 10.1016/j.coi.2008.04.012 Kang R, Zhang Q, Zeh HJ, 3rd, Lotze MT, Tang D. HMGB1 in cancer: good, bad, or both? Clin Cancer Res, 2013, 19(15):4046–57., DOI 10.1158/1078-0432. CCR-13-0495 Dajon M, Iribarren K, Cremer I. Toll-like receptor stimulation in cancer: a proand anti-tumor double-edged sword. Immunobiology, 2017, 222(1):89–100., DOI 10.1016/j.imbio.2016.06.009 Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol, 2010, 28:367–88., DOI 10.1146/ annurev.immunol.021908.132603 Chiba S, Baghdadi M, Akiba H, Yoshiyama H, Kinoshita I, Dosaka-Akita H, et al. Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat Immunol, 2012, 13(9):832–42., DOI 10.1038/ni.2376 Xu J, Li J, Yu Z, Rao H,Wang S, Lan H. HMGB1 promotes HLF-1 proliferation and ECM production through activating HIF1-α-regulated aerobic glycolysis. Pulm Pharmacol Ther, 2017, 45:136–41., DOI 10.1016/j.pupt.2017.05.015 Park SY, Lee SW, Kim HY, Lee WS, Hong KW, Kim CD. HMGB1 induces angiogenesis in rheumatoid arthritis via HIF-1α activation. Eur J Immunol, 2015, 45(4):1216–27., DOI 10.1002/eji.201444908 Di Maggio S, Milano G, De Marchis F, D’Ambrosio A, Bertolotti M, Palacios BS, et al. Non-oxidizable HMGB1 induces cardiac fibroblasts migration via CXCR4 in a CXCL12-independent manner and worsens tissue remodeling after myocardial infarction. Biochim Biophys Acta, Bba, Mol Basis Dis, 2017, 1863(11):2693–704., DOI 10.1016/j.bbadis.2017.07.012 Takeuchi A, Yamamoto Y, Munesue S, Harashima A, Watanabe T, Yonekura H, et al. Low molecular weight heparin suppresses receptor for advanced glycation end products-mediated expression of malignant phenotype in human fibrosarcoma cells. Cancer Sci, 2013, 104(6):740–9., DOI 10.1111/cas.12133 Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Tolllike receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med, 2007, 13(9):1050–9., DOI 10.1038/ nm1622 Gulic T, Silva-Gomes R, Davoudian S, Sironi M, Allavena P, Mantovani A, et al. Tumor-associated myeloid cells in cancer progression. In: N Rezaei, editor. Cancer immunology. Second edn. Cham, Switzerland: Springer, 2020). p. 29–46., DOI 10.1007/978-3-030-30845-2_3 Evankovich J, Cho SW, Zhang R, Cardinal J, Dhupar R, Zhang L, et al. High mobility group box 1 release from hepatocytes during ischemia and reperfusion injury is mediated by decreased histone deacetylase activity. J Biol Chem, 2010, 285(51):39888–97., DOI 10.1074/jbc.M110.128348 Bronte V, Brandau S, Chen S-H, Colombo MP, Frey AB, Greten TF, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun, 2016, 7:12150., DOI 10.1038/ ncomms12150 Taguchi A, Blood DC, del Toro G, Canet A, Lee DC, Qu W, et al. Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature, 2000, 405(6784):354–60., DOI 10.1038/35012626 Liu L, Yang M, Kang R, Wang Z, Zhao Y, Yu Y, et al. HMGB1-induced autophagy promotes chemotherapy resistance in leukemia cells. Leukemia, 2011, 25(1):23–31., DOI 10.1038/leu.2010.225 Liang X, Romo de Vivar Chavez A, Schapiro NE, Loughran P, Thorne SH, Amoscato AA, et al. Ethyl pyruvate administration inhibits hepatic tumor growth. J Leukoc Biol, 2009, 86(3):599–607., DOI 10.1189/jlb.0908578 Tang D, Kang R, Cheh C-W, Livesey KM, Liang X, Schapiro NE, et al. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene, 2010, 29(38):5299–310., DOI 10.1038/onc.2010.261 Ohndorf U-M, Rould MA, He Q, Pabo CO, Lippard SJ. Basis for recognition of cisplatin-modified DNA by high-mobility-group proteins. Nature, 1999, 399(6737):708–12., DOI 10.1038/21460 Suren D, Arda Gokay A, Sayiner A. High mobility group box 1, HMGB1, expression in gastric adenocarcinomas. J BUON, 2018, 23(2):422–7. Dong Y-D, Cui L, Peng C-H, Cheng D-F, Han B-S, Huang F. Expression and clinical significance of HMGB1 in human liver cancer: knockdown inhibits tumor growth and metastasis in vitro and in vivo. Oncol Rep, 2013, 29(1): 87–94., DOI 10.3892/or.2012.2070 Lee HJ, Kim A, Song IH, Park IA, Yu JH, Ahn JH, et al. Cytoplasmic expression of high mobility group B1, HMGB1, is associated with tumor-infiltrating lymphocytes, TILs, in breast cancer. Pathol Int, 2016, 66(4):202–9., DOI 10.1111/pin.12393 Takeuchi T, Sakazume K, Tonooka A, Zaitsu M, Takeshima Y,Mikami K, et al. Cytosolic HMGB1 expression in human renal clear cell cancer indicates higher pathological T classifications and tumor grades. Urol J, 2013, 10(3):960–5., DOI 10.22037/uj.v10i3.1425 Lee HJ, Kim JY, Song IH, Park IA, Yu JH, Ahn J-H, et al. High mobility group B1 and N1, HMGB1 and HMGN1, are associated with tumor-infiltrating lymphocytes in HER2-positive breast cancers. Virchows Arch, 2015, 467(6): 701–9., DOI 10.1007/s00428-015-1861-1 Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers, 2014, 6(3):1670–90., DOI 10.3390/cancers6031670 Komohara Y, Fujiwara Y, Ohnishi K, Takeya M. Tumor-associated macrophages: potential therapeutic targets for anti-cancer therapy. Adv Drug Deliv Rev, 2016, 99(Pt B):180–5., DOI 10.1016/j.addr.2015.11.009 Troiano G, Caponio VCA, Adipietro I, Tepedino M, Santoro R, Laino L, et al. Prognostic significance of CD68+ and CD163+ tumor associated macrophages in head and neck squamous cell carcinoma: a systematic review and meta-analysis. Oral Oncol, 2019, 93:66–75., DOI 10.1016/j. oraloncology.2019.04.019 Li J, Xie Y, Wang X, Li F, Li S, Li M, et al. Prognostic impact of tumorassociated macrophage infiltration in esophageal cancer: a meta-analysis. Future Oncol, 2019, 15(19):2303–17., DOI 10.2217/fon-2018-0669 Dancsok AR, Gao D, Lee AF, Steigen SE, Blay J-Y, Thomas DM, et al. Tumorassociated macrophages and macrophage-related immune checkpoint expression in sarcomas. Oncoimmunology, 2020, 9(1):1747340., DOI 10.1080/ 2162402X.2020.1747340 Ostrand-Rosenberg S, Fenselau C. Myeloid-derived suppressor cells: immunesuppressive cells that impair antitumor immunity and are sculpted by their environment. J.I., 2018, 200(2):422–31., DOI 10.4049/jimmunol.1701019 Tcyganov E, Mastio J, Chen E, Gabrilovich DI. Plasticity of myeloid-derived suppressor cells in cancer. Curr Opin Immunol, 2018, 51:76–82., DOI 10.1016/j. coi.2018.03.009 Akaike H, Kono K, Sugai H, Takahashi A, Mimura K, Kawaguchi Y, et al. Expression of high mobility group box chromosomal protein-1, HMGB-1, in gastric cancer. Anticancer Res, 2007, 27(1A):449–57. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 608582 EP 608590 DI 10.3389/pore.2021.608582 PG 9 ER PT J AU Feng, D Lin, J Wang, W Yan, K Liang, H Liang, J Yu, H Ling, B AF Feng, Dingqing Lin, Jie Wang, Wenhui Yan, Keqin Liang, Haiyan Liang, Jing Yu, Huan Ling, Bin TI Wnt3a/β-Catenin/CBP Activation in the Progression of Cervical Intraepithelial Neoplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cervical intraepithelial neoplasia; wnt signaling; CBP; Piwil2; stemness; Differentiation ID cervical intraepithelial neoplasia; wnt signaling; CBP; Piwil2; stemness; Differentiation AB Piwil2 reprograms HPV-infected reserve cells in the cervix into tumor-initiated cells (TICs) and upregulates Wnt3a expression sequentially, which leads to cervical intraepithelial neoplasia (CIN) and ultimately squamous cell carcinoma (SCC). However, little is known regarding Wnt signaling in the maintenance of TIC stemness during the progression of cervical lesions. We herein investigated the expression of canonical Wnt3a signaling and related genes by microarray data set analysis and immunohistochemical (IHC) staining of samples obtained by biopsy of normal cervix, low- and high-grade CIN, and invasive SCC tissue. Array data analyzed by GEO2R showed higher expression levels of Wnt signaling and their target genes, significant upregulation of stemness-associated markers, and notably downregulated cell differentiation markers in CIN and SCC tissues compared with those in the normal cervix tissue. Further, Gene Set Enrichment Analysis (GSEA) revealed that Wnt pathway-related genes significantly enriched in SCC. IHC staining showed gradually increased immunoreactivity score of Wnt3a and CBP and notable translocation of β-catenin from the membrane to the cytoplasm and nucleus during the lesion progression. The intensity and proportion of P16, Ki67 and CK17 staining also increased with the progression of cervical lesions, whereas minimal to negative Involucrin expression was observed in CIN2/3 and SCC. Therefore, canonical Wnt signaling may contribute to the progression of CIN to SCC and may be a potential therapeutic target. C1 [Feng, Dingqing] China-Japan Friendship Hospital, Department of Obstetrics and GynecologyBeijing, China. [Lin, Jie] China-Japan Friendship Hospital, Department of PathologyBeijing, China. [Wang, Wenhui] China-Japan Friendship Hospital, Department of Obstetrics and GynecologyBeijing, China. [Yan, Keqin] China-Japan Friendship Hospital, Department of Obstetrics and GynecologyBeijing, China. [Liang, Haiyan] China-Japan Friendship Hospital, Department of Obstetrics and GynecologyBeijing, China. [Liang, Jing] China-Japan Friendship Hospital, Department of Obstetrics and GynecologyBeijing, China. [Yu, Huan] China-Japan Friendship Hospital, Department of Obstetrics and GynecologyBeijing, China. [Ling, Bin] China-Japan Friendship Hospital, Department of Obstetrics and GynecologyBeijing, China. RP Ling, B (reprint author), China-Japan Friendship Hospital, Department of Obstetrics and Gynecology, Beijing, China. EM lingbin.ling@vip.sina.com CR Chibwesha CJ, Stringer JSA. Cervical cancer as a global concern. JAMA, 2019, 322(16):1558–60., DOI 10.1001/jama.2019.16176 ChenW, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA: A Cancer J Clinicians, 2016, 66(2):115–32., DOI 10.3322/caac. 21338 Ferenczy A, Franco E. Persistent human papillomavirus infection and cervical neoplasia. Lancet Oncol, 2002, 3(1):11–6., DOI 10.1016/s1470-2045(01)00617-9 Feng D, Yan K, Zhou Y, Liang H, Liang J, Zhao W, et al. Piwil2 is reactivated by HPV oncoproteins and initiates cell reprogramming via epigenetic regulation during cervical cancer tumorigenesis. Oncotarget, 2016, 7(40):64575–88., DOI 10.18632/oncotarget.11810 Pakula H, Xiang D, Li Z. A tale of two signals: AR and WNTin development and tumorigenesis of prostate and mammary gland. Cancers, 2017, 9(2):14., DOI 10. 3390/cancers9020014 KahnM. Wnt signaling in stem cells and cancer stem cells: a tale of two coactivators. Prog Mol Biol Transl Sci, 2018, 153:209–44., DOI 10.1016/bs.pmbts.2017.11.007 Yan KS, Janda CY, Chang J, Zheng GXY, Larkin KA, Luca VC, et al. Nonequivalence of Wnt and R-spondin ligands during Lgr5+ intestinal stem-cell self-renewal. Nature, 2017, 545(7653):238–42., DOI 10.1038/nature22313 Doumpas N, Lampart F, Robinson MD, Lentini A, Nestor CE, Cantu C, et al. TCF/LEF dependent and independent transcriptional regulation of Wnt/ β-catenin target genes. EMBO J, 2019, 38(2)., DOI 10.15252/embj.201798873 Lukaszewicz AI, Nguyen C, Melendez E, Lin DP, Teo J-L, Lai KKY, et al. The mode of stem cell division is dependent on the differential interaction of β-catenin with the Kat3 coactivators CBP or p300. Cancers, 2019, 11(7):962., DOI 10.3390/cancers11070962 Rieger ME, Zhou B, Solomon N, Sunohara M, Li C, Nguyen C, et al. p300/ β-Catenin interactions regulate adult progenitor cell differentiation downstream of WNT5a/protein kinase C, PKC). J Biol Chem, 2016, 291(12):6569–82., DOI 10.1074/jbc.M115.706416 Hoffmeyer K, Raggioli A, Rudloff S, Anton R, Hierholzer A, Del Valle I, et al. Wnt/-catenin signaling regulates telomerase in stem cells and cancer cells. Science, 2012, 336(6088):1549–54., DOI 10.1126/science.1218370 Schaefer KN, Peifer M. Wnt/Beta-catenin signaling regulation and a role for biomolecular condensates. Develop Cel, 2019, 48(4):429–44., DOI 10.1016/j. devcel.2019.01.025 Martins-Neves SR, Corver WE, Paiva-Oliveira DI, van den Akker BEWM, Briaire-de-Bruijn IH, Bovee JVMG, et al. Osteosarcoma stem cells have active wnt/β-catenin and overexpress SOX2 and KLF4. J Cel Physiol, 2016, 231(4): 876–86., DOI 10.1002/jcp.25179 Trisno SL, Philo KED, McCracken KW, Cata EM, Ruiz-Torres S, Rankin SA, et al. Esophageal organoids from human pluripotent stem cells delineate Sox2 functions during esophageal specification. Cell Stem Cell, 2018, 23(4):501–15., DOI 10.1016/j.stem.2018.08.008 Yong X, Tang B, Xiao Y-F, Xie R, Qin Y, Luo G, et al. Helicobacter pylori upregulates Nanog and Oct4 via Wnt/β-catenin signaling pathway to promote cancer stem cell-like properties in human gastric cancer. Cancer Lett, 2016, 374(2):292–303., DOI 10.1016/j.canlet.2016.02.032 Ogawa K, Nishinakamura R, Iwamatsu Y, Shimosato D, Niwa H. Synergistic action of Wnt and LIF in maintaining pluripotency of mouse ES cells. Biochem Biophysical Res Commun, 2006, 343(1):159–66., DOI 10.1016/j.bbrc.2006.02.127 Zandi M, Muzaffar M, Shah SM, Kaushik R, Singh MK, Palta P, et al. WNT3A signalling pathway in buffalo, Bubalus bubalis, embryonic stem cells. Reprod Fertil Dev, 2014, 26(4):551–61., DOI 10.1071/RD13084 Nourbakhsh M, Mansoor A, Koro K, Zhang Q,Minoo P. Expression profiling reveals involvement of WNT pathway in the malignant progression of sessile serrated adenomas. Am J Pathol, 2019, 189(9):1732–43., DOI 10.1016/j.ajpath.2019.05.009 Lane TF, Leder P. Wnt-10b directs hypermorphic development and transformation in mammary glands of male and female mice. Oncogene, 1997, 15(18):2133–44., DOI 10.1038/sj.onc.1201593 Wong GT, Gavin BJ, McMahon AP. Differential transformation of mammary epithelial cells by Wnt genes. Mol Cel Biol., 1994, 14(9):6278–86., DOI 10.1128/ mcb.14.9.6278 Zhuang S-M,Wiseman RW, Soderkvist P. Frequent mutations of the Trp53, Hras1 and β-catenin, Catnb, genes in 1,3-butadiene-induced mammary adenocarcinomas in B6C3F1 mice. Oncogene, 2002, 21(36):5643–8., DOI 10.1038/sj.onc.1205649 Moser AR, Mattes EM, Dove WF, Lindstrom MJ, Haag JD, Gould MN. ApcMin, a mutation in the murine Apc gene, predisposes to mammary carcinomas and focal alveolar hyperplasias. Proc Natl Acad Sci, 1993, 90(19):8977–81., DOI 10.1073/pnas.90.19.8977 Martens JE, Arends J, Van der Linden PJ, De Boer BA, Helmerhorst TJ. Cytokeratin 17 and p63 are markers of the HPV target cell, the cervical stem cell. Anticancer Res, 2004, 24(2B):771–5. Ortiz-Sanchez E, Santiago-Lopez L, Cruz-Dominguez VB, Toledo-Guzman ME, Hernandez-Cueto D, Muniz-Hernandez S, et al. Characterization of cervical cancer stem cell-like cells: phenotyping, stemness, and human papilloma virus co-receptor expression. Oncotarget, 2016, 7(22):31943–54., DOI 10.18632/oncotarget.8218 Zhan T, Ambrosi G, Wandmacher AM, Rauscher B, Betge J, Rindtorff N, et al. MEKinhibitors activateWnt signalling and induce stem cell plasticity in colorectal cancer. Nat Commun, 2019, 10(1):2197., DOI 10.1038/s41467-019-09898-0 Liu X-F, Li X-Y, Zheng P-S, Yang W-T. DAX1 promotes cervical cancer cell growth and tumorigenicity through activation of Wnt/β-catenin pathway via GSK3β. Cell Death Dis, 2018, 9(3):339., DOI 10.1038/s41419-018-0359-6 Dai Y, Liu L, Zeng T, Liang J-Z, Song Y, Chen K, et al. Overexpression of MUC13, a poor prognostic predictor, promotes cell growth by activating Wnt signaling in hepatocellular carcinoma. Am J Pathol, 2018, 188(2):378–91., DOI 10.1016/j.ajpath.2017.10.016 Ayala-Calvillo E, Mojica-Vazquez LH, Garcia-Carranca A, Gonzalez-Maya L. Wnt/βcatenin pathway activation and silencing of the APC gene in HPVpositive human cervical cancerderived cells. Mol Med Rep, 2018, 17(1):200–8., DOI 10.3892/mmr.2017.7853 Kim B, Byun S-J, Kim YA, Kim JE, Lee BL, Kim WH, et al. Cell cycle regulators, APC/β-catenin, NF-κB and Epstein-Barr virus in gastric carcinomas. Pathology, 2010, 42(1):58–65., DOI 10.3109/00313020903356392 Uren A, Fallen S, Yuan H, Usubutun A, Kucukali T, Schlegel R, et al. Activation of the canonical Wnt pathway during genital keratinocyte transformation: a model for cervical cancer progression. Cancer Res, 2005, 65(14):6199–206., DOI 10.1158/0008-5472.CAN-05-0455 Gang EJ, Hsieh Y-T, Pham J, Zhao Y, Nguyen C, Huantes S, et al. Smallmolecule inhibition of CBP/catenin interactions eliminates drug-resistant clones in acute lymphoblastic leukemia. Oncogene, 2014, 33(17):2169–78., DOI 10.1038/onc.2013.169 Li J, Sutter C, Parker DS, Blauwkamp T, Fang M, Cadigan KM. CBP/p300 are bimodal regulators of Wnt signaling. EMBO J, 2007, 26(9):2284–94., DOI 10. 1038/sj.emboj.7601667 Bordonaro M, Lazarova DL. CREB-binding protein, p300, butyrate, and Wnt signaling in colorectal cancer. Wjg, 2015, 21(27):8238–48., DOI 10.3748/wjg. v21.i27.8238 Bordonaro M, Lazarova DL. Determination of the role of CBP- and p300- mediated Wnt signaling on colonic cells. JMIR Res Protoc, 2016, 5(2):e66., DOI 10.2196/resprot.5495 Ring A, Kim Y-M, Kahn M. Wnt/catenin signaling in adult stem cell physiology and disease. Stem Cel Rev Rep, 2014, 10(4):512–25., DOI 10.1007/s12015-014-9515-2 Malleske DT, Hayes D, Jr, Lallier SW, Hill CL, Reynolds SD. Regulation of human airway epithelial tissue stem cell differentiation by β-catenin, P300, and CBP. Stem Cells, 2018, 36(12):1905–16., DOI 10.1002/stem.2906 Jakoby M, Schnittger A. Cell cycle and differentiation. Curr Opin Plant Biol, 2004, 7(6):661–9., DOI 10.1016/j.pbi.2004.09.015 Otto T, Candido SV, Pilarz MS, Sicinska E, Bronson RT, Bowden M, et al. Cell cycle-targeting microRNAs promote differentiation by enforcing cell-cycle exit. Proc Natl Acad Sci USA, 2017, 114(40):10660–5., DOI 10.1073/pnas.1702914114 Wang Y, Zhao J, Cao C, Yan Y, Chen J, Feng F, et al. The role of E2F1-topoIIβ signaling in regulation of cell cycle exit and neuronal differentiation of human SH-SY5Y cells. Differentiation, 2018, 104:1–12., DOI 10.1016/j.diff.2018.07.002 Kahn M. Symmetric division versus asymmetric division: a tale of two coactivators. Future Med Chem, 2011, 3(14):1745–63., DOI 10.4155/fmc.11.126 Tang Y, Berlind J, Mavila N. Inhibition of CREB binding protein-beta-catenin signaling down regulates CD133 expression and activates PP2A-PTEN signaling in tumor initiating liver cancer cells. Cell Commun Signal, 2018, 16(1):9., DOI 10.1186/s12964-018-0222-5 Chan KC, Chan LS, Ip JCY, Lo C, Yip TTC, Ngan RKC, et al. Therapeutic targeting of CBP/β-catenin signaling reduces cancer stem-like population and synergistically suppresses growth of EBV-positive nasopharyngeal carcinoma cells with cisplatin. Sci Rep, 2015, 5:9979., DOI 10.1038/srep09979 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 609620 EP 609627 DI 10.3389/pore.2021.609620 PG 8 ER PT J AU Naso, J Povshedna, T Wang, G Banyi, N MacAulay, C Ionescu, D Zhou, Ch AF Naso, R. Julia Povshedna, Tetiana Wang, Gang Banyi, Norbert MacAulay, Calum Ionescu, N. Diana Zhou, Chen TI Automated PD-L1 Scoring for Non-Small Cell Lung Carcinoma Using Open-Source Software SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE pathology; PD-L1; biomarker; non-small cell lung cancer; digital pathology ID pathology; PD-L1; biomarker; non-small cell lung cancer; digital pathology AB PD-L1 expression in non-small cell lung cancer (NSCLC) is predictive of response to immunotherapy, but scoring of PD-L1 immunohistochemistry shows considerable interobserver variability. Automated methods may allow more consistent and expedient PD-L1 scoring. We aimed to assess the technical concordance of PD-L1 scores produced using free open source QuPath software with the manual scores of three pathologists. A classifier for PD-L1 scoring was trained using 30 NSCLC image patches. A separate test set of 207 image patches from 69 NSCLC resection cases was used for comparison of automated and manual scores. Automated and average manual scores showed excellent correlation (concordance correlation coeffecient = 0.925), though automated scoring resulted in significantly more 1–49% scores than manual scoring (p = 0.012). At both 1% and 50% thresholds, automated scores showed a level of concordance with our ‘gold standard’ (the average of three pathologists’ manual scores) similar to that of individual pathologists. Automated scoring showed high sensitivity (95%) but lower specificity (84%) at a 1% threshold, and excellent specificity (100%) but lower sensitivity (71%) at a 50% threshold. We conclude that our automated PD-L1 scoring system for NSCLC has an accuracy similar to that of individual pathologists. The detailed protocol we provide for free open source scoring software and our discussion of the limitations of this technology may facilitate more effective integration of automated scoring into clinical workflows. C1 [Naso, R. Julia] University of British Columbia, Department of PathologyVancouver, BC, Canada. [Povshedna, Tetiana] BC Cancer, Department of PathologyVancouver, BC, Canada. [Wang, Gang] BC Cancer, Department of PathologyVancouver, BC, Canada. [Banyi, Norbert] BC Cancer, Department of PathologyVancouver, BC, Canada. [MacAulay, Calum] British Columbia Cancer Research Center, Department of Integrative OncologyVancouver, BC, Canada. [Ionescu, N. Diana] BC Cancer, Department of PathologyVancouver, BC, Canada. [Zhou, Chen] BC Cancer, Department of PathologyVancouver, BC, Canada. RP Zhou, Ch (reprint author), BC Cancer, Department of Pathology, Vancouver, Canada. EM czhou@bccancer.bc.ca CR Gong J, Chehrazi-Raffle A, Reddi S, Salgia R Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J Immunother Cancer, 2018). 6:8., DOI 10.1186/s40425-018-0316-z Buttner R, Gosney JR, Skov BG, Adam J, Motoi N, Bloom KJ, et al. Programmed death-ligand 1 immunohistochemistry testing: a review of analytical assays and clinical implementation in non-small-cell lung cancer. JCO, 2017). 35:3867–76., DOI 10.1200/jco.2017.74.7642 Tsao MS, Kerr KM, Kockx M, Beasley M-B, Borczuk AC, Botling J, et al. PD-L1 immunohistochemistry comparability study in real-life clinical samples: results of blueprint phase 2 Project. J Thorac Oncol, 2018). 13:1302–11., DOI 10.1016/j. jtho.2018.05.013 Brunnstrom H, Johansson A, Westbom-Fremer S, Backman M, Djureinovic D, Patthey A, et al. PD-L1 immunohistochemistry in clinical diagnostics of lung cancer: inter-pathologist variability is higher than assay variability. Mod Pathol, 2017). 30:1411–21., DOI 10.1038/modpathol.2017.59 Ratcliffe MJ, Sharpe A, Midha A, Barker C, Scott M, Scorer P, et al. Agreement between programmed cell death ligand-1 diagnostic assays across multiple protein expression cutoffs in non-small cell lung cancer. Clin Cancer Res, 2017). 23:3585–91., DOI 10.1158/1078-0432.ccr-16-2375 Chang S, Park HK, Choi Y-L, Jang SJ Interobserver reproducibility of PD-L1 biomarker in non-small cell lung cancer: a multi-institutional study by 27 pathologists. J Pathol Transl Med, 2019). 53:347–53., DOI 10.4132/jptm.2019. 09.29 Koomen BM, Badrising SK, van den Heuvel MM, Willems SMComparability of PD-L1 immunohistochemistry assays for non-small-cell lung cancer: a systematic review. Histopathology, 2019). 76(6):793–802., DOI 10.1111/his.14040 Taylor CR, Jadhav AP, Gholap A, Kamble G, Huang J, Gown A, et al. A multiinstitutional study to evaluate automated whole slide scoring of immunohistochemistry for assessment of programmed death-ligand 1, PDL1, expression in non-small cell lung cancer. Appl Immunohistochem Mol Morphol, 2019). 27:263–9., DOI 10.1097/pai.0000000000000737 Kapil A, Meier A, Zuraw A, Steele KE, Rebelatto MC, Schmidt G, et al. Deep semi supervised generative learning for automated tumor proportion scoring on NSCLC tissue needle biopsies. Sci Rep, 2018). 8:17343., DOI 10.1038/s41598-018- 35501-5 Bankhead P, Fernandez JA, McArt DG, Boyle DP, Li G, Loughrey MB, et al. Integrated tumor identification and automated scoring minimizes pathologist involvement and provides new insights to key biomarkers in breast cancer. Lab Invest, 2018). 98:15–26., DOI 10.1038/labinvest.2017.131 Bankhead P, Loughrey MB, Fernandez JA, Dombrowski Y,McArt DG, Dunne PD, et al. QuPath: open source software for digital pathology image analysis. Sci Rep, 2017). 7:16878., DOI 10.1038/s41598-017-17204-5 Humphries MP, Bingham V, Abdullahi Sidi F, Craig SG, McQuaid S, James J, et al. Improving the diagnostic accuracy of the PD-L1 test with image analysis and multiplex hybridization. Cancers, 2020). 12:1114., DOI 10.3390/ cancers12051114 Naso JR, Wang G, Banyi N, Derakhshan F, Shokoohi A, Ho C, et al., 2020). Comparability of Laboratory-Developed and Commercial PD-L1 Assays in Non-Small Cell Lung carcinoma. Annals of Diagnostic Pathology 50(1):151590., DOI 10.1016/j.anndiagpath.2020.151590 Cooper WA, Russell PA, Cherian M, Duhig EE, Godbolt D, Jessup PJ, et al. Intra- and interobserver reproducibility assessment of PD-L1 biomarker in non-small cell lung cancer. Clin Cancer Res, 2017). 23:4569–77., DOI 10.1158/ 1078-0432.ccr-17-0151 Rimm DL, Han G, Taube JM, Yi ES, Bridge JA, Flieder DB, et al. A prospective, multi-institutional, pathologist-based assessment of 4 immunohistochemistry assays for PD-L1 expression in non-small cell lung cancer. JAMA Oncol, 2017). 3:1051–8., DOI 10.1001/jamaoncol.2017.0013 Humphries MP, McQuaid S, Craig SG, Bingham V, Maxwell P, Maurya M, et al. Critical appraisal of programmed death ligand 1 reflex diagnostic testing: current standards and future opportunities. J Thorac Oncol, 2019). 14:45–53., DOI 10.1016/j.jtho.2018.09.025 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 609717 EP 609725 DI 10.3389/pore.2021.609717 PG 9 ER PT J AU Dai, J Zhou, N Wu, R Du, J Miao, Sh Gong, K Yang, L Chen, W Li, X Li, Ch Wu, Y AF Dai, Juanjuan Zhou, Ning Wu, Rui Du, Jing Miao, Shuang Gong, Kaikai Yang, Lijuan Chen, Weiwei Li, Xuelin Li, Chen Wu, Yan TI LncRNA MALAT1 Regulating Lung Carcinoma Progression via the miR-491-5p/UBE2C Axis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE MALAT1; miR-491-5p; UBE2C; lung carcinoma; proliferatiom ID MALAT1; miR-491-5p; UBE2C; lung carcinoma; proliferatiom AB Long noncoding RNAs (lncRNAs) play a critical role in the development of lung carcinoma. The mechanism of MALAT1 in lung carcinoma development is not understood very well. This study aimed to investigate the role of MALAT1 in lung carcinoma progression and the mechanism underlying the role of miR-491-5p in the MALAT1 mediated regulation of UBE2C expression. The results indicated that the expression of MALAT1 was often augmented in lung carcinoma cells. Suppression of MALAT1 blocked the proliferation, invasion and migration ability of cancer cells and inhibited the expression of UBE2C. UBE2C restoration attenuated the MALAT1 knockdown-induced anti-cancer effects. Moreover, UBE2C and MALAT1 were indicated as targets of miR-491-5p and inhibition of miR-491-5p restored the MALAT1 knockdown-induced inhibition of the progression of lung carcinoma. Furthermore, MALAT1 sponged miR-491-5p to upregulate UBE2C expression, causing it to act as a competing endogenous RNA. Collectively, MALAT1 downregulation suppressed lung carcinoma progression by regulating the miR-491-5p/UBE2C axis. These results indicate that MALAT1 could be a molecular target for lung carcinoma treatment and prognosis. C1 [Dai, Juanjuan] Binzhou Medical University Hospital, Cancer Research InstituteBinzhou, China. [Zhou, Ning] Binzhou Medical University Hospital, Department of Otolaryngology Head and Neck SurgeryBinzhou, China. [Wu, Rui] Binzhou Medical University Hospital, Cancer Research InstituteBinzhou, China. [Du, Jing] Binzhou Medical University Hospital, Cancer Research InstituteBinzhou, China. [Miao, Shuang] Binzhou Medical University Hospital, Cancer Research InstituteBinzhou, China. [Gong, Kaikai] Binzhou Medical University Hospital, Cancer Research InstituteBinzhou, China. [Yang, Lijuan] Binzhou Medical University Hospital, Cancer Research InstituteBinzhou, China. [Chen, Weiwei] Binzhou Medical University Hospital, Cancer Research InstituteBinzhou, China. [Li, Xuelin] Binzhou Medical University Hospital, Cancer Research InstituteBinzhou, China. [Li, Chen] Binzhou Medical University Hospital, Institute for Metabolic and Neuropsychiatric DisordersBinzhou, China. [Wu, Yan] Binzhou Medical University Hospital, Cancer Research InstituteBinzhou, China. RP Wu, Y (reprint author), Binzhou Medical University Hospital, Cancer Research Institute, Binzhou, China. EM wuyan55@126.com CR Siegel RL,Miller KD, Jemal A. Cancer statistics, 2019. CA A Cancer J Clin, 2019, 69:7–34., DOI 10.3322/caac.21551 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA: A Cancer J Clinicians, 2016, 66:115–32., DOI 10.3322/caac. 21338 Shi X, Shao X, Zhang Y,Wu F, Tao Y. Tumor location and survival outcomes in lung adenosquamous carcinoma: a propensity score matched analysis. Med Sci Monit, 2020, 26:e922138., DOI 10.12659/msm.922138 Reck M, Heigener DF, Mok T, Soria J-C, Rabe KF. Management of non-smallcell lung cancer: recent developments. The Lancet, 2013, 382:709–19., DOI 10. 1016/s0140-6736(13)61502-0 Li X, Wu Z, Fu X, Han W. Long noncoding RNAs: insights from biological features and functions to diseases. Med Res Rev, 2013, 33:517–53., DOI 10.1002/ med.21254 Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell, 2009, 136:629–41., DOI 10.1016/j.cell.2009.02.006 Gibb EA, Brown CJ, Lam WL. The functional role of long non-coding RNA in human carcinomas. Mol Cancer, 2011, 10:38., DOI 10.1186/1476-4598-10-38 Hu X, Sood AK, Dang CV, Zhang L. The role of long noncoding RNAs in cancer: the dark matter matters. Curr Opin Genet Dev, 2018, 48:8–15., DOI 10. 1016/j.gde.2017.10.004 Sanchez Calle A, Kawamura Y, Yamamoto Y, Takeshita F, Ochiya T. Emerging roles of long non-coding RNA in cancer. Cancer Sci, 2018, 109:2093–100., DOI 10.1111/cas.13642 Cheng Y, Imanirad P, Jutooru I, Hedrick E, Jin UH, Rodrigues Hoffman A, et al. Role of metastasis-associated lung adenocarcinoma transcript-1, MALAT-1, in pancreatic cancer. Plos One, 2018, 13:e192264., DOI 10.1371/ journal.pone.0192264 Arun G, Spector DL. MALAT1 long non-coding RNA and breast cancer. RNA Biol, 2019, 16:860–3., DOI 10.1080/15476286.2019.1592072 Gutschner T, Hammerle M, Eissmann M, Hsu J, Kim Y, Hung G, et al. The noncoding RNAMALAT1 is a critical regulator of themetastasis phenotype of lung cancer cells. Cancer Res, 2013, 73:1180–9., DOI 10.1158/0008-5472.can-12-2850 Chan JJ, Tay Y. Noncoding RNA:RNA regulatory networks in cancer. Int J Mol Sci, 2018, 19., DOI 10.3390/ijms19051310 Xiang Y, Zhang Y, Tang Y, Li Q. MALAT1 modulates TGF-β1-induced endothelial-to-mesenchymal transition through downregulation of miR- 145. Cell Physiol Biochem, 2017, 42:357–72., DOI 10.1159/000477479 YiRen H, YingCong Y, Sunwu Y, Keqin L, Xiaochun T, Senrui C, et al. Long noncoding RNA MALAT1 regulates autophagy associated chemoresistance via miR-23b-3p sequestration in gastric cancer. Mol Cancer, 2017, 16:174., DOI 10.1186/s12943-017-0743-3 Hao Z, Zhang H, Cowell J. Ubiquitin-conjugating enzyme UBE2C: molecular biology, role in tumorigenesis, and potential as a biomarker. Tumor Biol, 2012, 33:723–30., DOI 10.1007/s13277-011-0291-1 van Ree JH, Jeganathan KB, Malureanu L, van Deursen JM. Overexpression of the E2 ubiquitin-conjugating enzyme UbcH10 causes chromosome missegregation and tumor formation. J Cell Biol, 2010, 188:83–100., DOI 10. 1083/jcb.200906147 Guo J, Wu Y, Du J, Yang L, Chen W, Gong K, et al. Deregulation of UBE2Cmediated autophagy repression aggravates NSCLC progression. Oncogenesis, 2018, 7:49., DOI 10.1038/s41389-018-0054-6 Guo J, Jin D, Wu Y, Yang L, Du J, Gong K, et al. RETRACTED: the miR 495- UBE2C-ABCG2/ERCC1 axis reverses cisplatin resistance by downregulating drug resistance genes in cisplatin-resistant non-small cell lung cancer cells. Ebiomedicine, 2018, 35:204–21., DOI 10.1016/j.ebiom.2018.08.001 Wu Y, Jin D, Wang X, Du J, Di W, An J, et al. UBE2C induces cisplatin resistance via ZEB1/2-dependent upregulation of ABCG2 and ERCC1 in NSCLC cells. J Oncol, 2019, 8607859., DOI 10.1155/2019/8607859 Ye Y, Rape M. Building ubiquitin chains: E2 enzymes at work. Nat Rev Mol Cel Biol, 2009, 10:755–64., DOI 10.1038/nrm2780 Dong Y, Liang G, Yuan B, Yang C, Gao R, Zhou X. MALAT1 promotes the proliferation and metastasis of osteosarcoma cells by activating the PI3K/ Akt pathway. Tumor Biol, 2015, 36:1477–86., DOI 10.1007/s13277-014- 2631-4 Li MM, Dong CX, Sun B, Lei HZ, Wang YL, Gong YB, et al. LncRNAMALAT1 promotes tumorogenesis of infantile hemangioma by competitively binding miR-424 to stimulate MEKK3/NF-κB pathway. Life Sci, 2019, 239: 116946., DOI 10.1016/j.lfs.2019.116946 Liang J, Liang L, Ouyang K, Li Z, Yi X. MALAT1 induces tongue cancer cells′ EMT and inhibits apoptosis through Wnt/beta-catenin signaling pathway. J Oral Pathol Med, 2019, 46:98–105., DOI 10.1111/jop.12466 Chen L, Feng P, Zhu X, He S, Duan J, Zhou D. Long non-coding RNA Malat1 promotes neurite outgrowth through activation of ERK/MAPK signalling pathway in N2a cells. J Cel Mol. Med., 2016, 20:2102–10., DOI 10.1111/ jcmm.12904 Ji P, Diederichs S, Wang W, Boing S, Metzger R, Schneider PM, et al. MALAT- 1, a novel noncoding RNA, and thymosin β4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene, 2003, 22:8031–41., DOI 10. 1038/sj.onc.1206928 Fernandez EJ, Villa-Roel N, Kumar S, Gu L, Salim MT, Ohh M, et al. Disturbed flow increases UBE2C, ubiquitin E2 ligase C, via loss of miR-483-3p, inducing aortic valve calcification by the pVHL, von Hippel-lindau protein, and HIF- 1alpha, Hypoxia-Inducible factor-1alpha, pathway in endothelial cells. Arterioscler Thromb Vasc Biol, 2019, 39:467–81., DOI 10.1161/ATVBAHA. 118.312233 Sun R, Liu Z, Tong D, Yang Y, Guo B,Wang X, et al. miR-491-5p, mediated by Foxi1, functions as a tumor suppressor by targeting Wnt3a/beta-catenin signaling in the development of gastric cancer. Cell Death Dis, 2017, 8: e2714., DOI 10.1038/cddis.2017.134 Huang W-C, Chan S-H, Jang T-H, Chang J-W, Ko Y-C, Yen T-C, et al. miRNA-491-5p and GIT1 serve as modulators and biomarkers for oral squamous cell carcinoma invasion and metastasis. Cancer Res, 2014, 74: 751–64., DOI 10.1158/0008-5472.can-13-1297 Xu Y, Hou R, Lu Q, Zhang Y, Chen L, Zheng Y, et al. MiR-491-5p negatively regulates cell proliferation and motility by targeting PDGFRA in prostate cancer. Am J Cancer Res, 2017, 7:2545–53. Gong F, Ren P, Zhang Y, Jiang J, Zhang H.MicroRNAs-491-5p suppresses cell proliferation and invasion by inhibiting IGF2BP1 in non-small cell lung cancer. Am J Transl Res, 2016, 8:485–95. Hui Z, Yiling C, Wenting Y, XuQun H, ChuanYi Z, Hui L. miR-491-5p functions as a tumor suppressor by targeting JMJD2B in ERα-positive breast cancer. Febs Lett, 2015, 589:812–21., DOI 10.1016/j.febslet.2015.02.014 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 610159 EP 610168 DI 10.3389/pore.2021.610159 PG 10 ER PT J AU Wu, Y Chen, S Zhang, M Liu, K Jing, J Pan, K Zhang, L Xu, B Lu, X Chen, M AF Wu, Yuqing Chen, Saisai Zhang, Minhao Liu, Kuangzheng Jing, Jibo Pan, Kehao Zhang, Lihua Xu, Bin Lu, Xiaoming Chen, Ming TI Factors Associated with Survival From Xp11.2 Translocation Renal Cell Carcinoma Diagnosis—A Systematic Review and Pooled Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE TFE3; kidney; Xp112 translocation renal cell carcinoma; survival; prognosis ID TFE3; kidney; Xp112 translocation renal cell carcinoma; survival; prognosis AB Purpose: Xp11.2 translocation renal cell carcinoma (Xp11.2 tRCC) is a rare subtype of renal cell carcinoma (RCC), characterized by translocations of Xp11.2 breakpoints, involving of the transcription factor three gene (TFE3). The aim of our study was to comprehensively characterize the clinical characteristics and outcomes, and to identify risk factors associated with OS and PFS in Xp11.2 tRCC patients. Methods: Literature search on Xp11.2 tRCC was performed using databases such as pubmed EMBASE and Web of Science. Studies were eligible if outcomes data (OS and/or PFS) were reported for patients with a histopathologically confirmed Xp11.2 tRCC. PFS and OS were evaluated using the univariable and multivariable Cox regression model. Results: There were 80 eligible publications, contributing 415 patients. In multivariable analyses, the T stage at presentation was significantly associated with PFS (HR: 3.87; 95% CI: 1.70 to 8.84; p = 0.001). The median time of PFS was 72 months. In the multivariable analyses, age at diagnosis (HR: 2.16; 95% CI: 1.03 to 4.50; p = 0.041), T stage at presentation (HR: 4.44; 95% CI: 2.16 to 9.09; p < 0.001) and metastasis status at presentation (HR: 2.67; 95% CI: 1.12 to 6.41; p = 0.027) were all associated with OS, with a median follow-up time of 198 months. Conclusion: T stage at presentation is the only factor that is associated with both PFS and OS in patients with Xp11.2 tRCC. Also, patients over 45 or with metastases are more likely to have poorer OS. C1 [Wu, Yuqing] Southeast University, School of Medicine, Surgical Research Center, Institute of UrologyNanjing, China. [Chen, Saisai] Southeast University, School of Medicine, Surgical Research Center, Institute of UrologyNanjing, China. [Zhang, Minhao] Southeast University, School of Medicine, Surgical Research Center, Institute of UrologyNanjing, China. [Liu, Kuangzheng] Southeast University, School of Medicine, Surgical Research Center, Institute of UrologyNanjing, China. [Jing, Jibo] Southeast University, School of Medicine, Surgical Research Center, Institute of UrologyNanjing, China. [Pan, Kehao] Southeast University, School of Medicine, Surgical Research Center, Institute of UrologyNanjing, China. [Zhang, Lihua] Affiliated Zhongda Hospital of Southeast University, Department of PathologyNanjing, China. [Xu, Bin] Southeast University, School of Medicine, Surgical Research Center, Institute of UrologyNanjing, China. [Lu, Xiaoming] Yancheng Third People's Hospital, Department of UrologyYancheng, China. [Chen, Ming] Affiliated Zhongda Hospital of Southeast University, Department of UrologyNanjing, China. RP Chen, M (reprint author), Affiliated Zhongda Hospital of Southeast University, Department of Urology, Nanjing, China. EM mingchenseu@126.com CR Eble JN, Sauter GInternational Agency for Research on Cancer. Pathology and genetics of Tumors of the urinary system and male genital organs. Lyon, France: IARC Press, 2004): 359. Hirobe M, Masumori N, Tanaka T, Kitamura H, Tonooka A, Hasegawa T, et al. Clinicopathological characteristics of Xp11.2 translocation renal cell carcinoma in adolescents and adults: diagnosis using immunostaining of transcription factor E3 and fluorescence in situ hybridization analysis. Int J Urol, 2016). 23(2): 140–5., DOI 10.1111/iju.13007 Argani P, Ladanyi M. Translocation carcinomas of the kidney. Clin Lab Med, 2005). 25:363–78., DOI 10.1016/j.cll.2005.01.008 Argani P, Lae M, Hutchinson B, Reuter VE, Collins MH, Perentesis J, et al. Renal carcinomas with the t(6;11)(p21;q12): clinicopathologic features and demonstration of the specific alpha-TFEB gene fusion by immunohistochemistry, RT-PCR, and DNA PCR. Am J Surg Pathol, 2005). 29(2):230–40., DOI 10.1097/01.pas.0000146007.54092.37 Pan CC, Sung MT, Huang HY, Yeh KT. High chromosomal copy number alterations in Xp11 translocation renal cell carcinomas detected by array comparative genomic hybridization are associated with aggressive behavior. Am J Surg Pathol, 2013). 37(7):1116–9., DOI 10.1097/PAS.0b013e318293d872 Moch H, Reuter VE. WHO classification of tumours of the urinary system and male genital organs. Lyon, France: International Agency for Research on Cancer, 2016): 356. Malouf GG, Camparo P, Molinie V, Dedet G, Oudard S, Schleiermacher G, et al. Transcription factor E3 and transcription factor EB renal cell carcinomas: clinical features, biological behavior and prognostic factors. J Urol, 2011). 185(1):24–9., DOI 10.1016/j.juro.2010.08.092 Rao Q, Williamson SR, Zhang S, Eble JN, Grignon DJ, Wang M, et al. TFE3 break-apart FISH has a higher sensitivity for Xp11.2 translocationassociated renal cell carcinoma compared with TFE3 or cathepsin K immunohistochemical staining alone: expanding the morphologic spectrum. Am J Surg Pathol, 2013). 37:804–15., DOI 10.1097/PAS.0b013e31827e17cb Ahluwalia P, Nair B, Kumar G. Renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusion: a rare case report with review of the literature. Case Rep Urol, 2013). 2013:810590., DOI 10.1155/2013/810590 Altinok G, Kattar MM, Mohamed A, Poulik J, Grignon D, Rabah R. Pediatric renal carcinoma associated with Xp11.2 translocations/TFE3 gene fusions and clinicopathologic associations. Pediatr Dev Pathol, 2005). 8(2):168–80., DOI 10. 1007/s10024-004-9106-3 Antic T, Taxy JB, Alikhan M, Segal J. Melanotic translocation renal cell carcinoma with a novel ARID1B-TFE3 gene fusion. Am J Surg Pathol, 2017). 41:1576–1580., DOI 10.1097/PAS.0000000000000927 Aoyagi T, Shinohara N, Kubota-Chikai K, Kuroda N, Nonomura K. Long-term survival in a patient with node-positive adult-onset Xp11.2 translocation renal cell carcinoma. Urol Int, 2011). 86(4):487–90., DOI 10.1159/000323866 Argani P, Antonescu CR, Couturier J, Fournet JC, Sciot R, Debiec-Rychter M, et al. PRCC-TFE3 renal carcinomas: morphologic, immunohistochemical, ultrastructural, and molecular analysis of an entity associated with the t(X; 1)(p11.2;q21). Am J Surg Pathol, 2002). 26(12):1553–66., DOI 10.1097/ 00000478-200212000-00003 Argani P, Olgac S, Tickoo SK, Goldfischer M, Moch H, Chan DY, et al. Xp11 translocation renal cell carcinoma in adults: expanded clinical, pathologic, and genetic spectrum. Am J Surg Pathol, 2007). 31(8):1149–60., DOI 10.1097/PAS. 0b013e318031ffff Argani P, Zhang L, Reuter VE, Tickoo SK, Antonescu CR. RBM10-TFE3 renal cell carcinoma: a potential diagnostic pitfall due to cryptic intrachromosomal Xp11.2 inversion resulting in false-negative TFE3 FISH. Am J Surg Pathol, 2017). 41:655–662., DOI 10.1097/PAS.0000000000000835 Argani P, Zhong M, Reuter VE, Fallon JT, Epstein JI, Netto GJ, et al. TFE3- Fusion variant analysis defines specific clinicopathologic associations among Xp11 translocation cancers. Am J Surg Pathol, 2020). 40:723–37., DOI 10.1097/ PAS.0000000000000631 Armah HB, Parwani AV, Surti U, Bastacky SI. Xp11.2 translocation renal cell carcinoma occurring during pregnancy with a novel translocation involving chromosome 19: a case report with review of the literature. Diagn Pathol, 2009). 4:15., DOI 10.1186/1746-1596-4-15 Arnoux V, Long JA, Fiard G, Pasquier D, Bensaadi L, Terrier N, et al. [Xp11.2 translocation renal carcinoma in adults over 50 years of age: about four cases]. Prog Urol, 2012). 22(15):932–7., DOI 10.1016/j.purol.2012.06.009 Barroca H, Castedo S, Vieira J, Teixeira M, Muller-Hocker J. Altered expression of key cell cycle regulators in renal cell carcinoma associated with Xp11.2 translocation. Pathol Res Pract, 2009). 205(7):466–72., DOI 10. 1016/j.prp.2009.01.005 Bovio IM, Allan RW, Oliai BR, Hampton T, Rush DS. Xp11.2 translocation renal carcinoma with placental metastasis: a case report. Int J Surg Pathol, 2011). 19(1):80–3., DOI 10.1177/1066896908331231 Brassesco MS, Valera ET, Bonilha TA, Scrideli CA, Carvalho de Oliveira J, Pezuk JA, et al. Secondary PSF/TFE3-associated renal cell carcinoma in a child treated for genitourinary rhabdomyosarcoma. Cancer Genet, 2011). 204(2): 108–10., DOI 10.1016/j.cancergencyto.2010.10.006 Cheng X, He J, GanW, Fan X, Yang J, Zhu B, et al. Pseudocapsule of renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusion: a clue for tumor enucleation?. Int J Clin Exp Pathol, 2015). 8(5):5403–10 . Choueiri TK, Mosquera JM, Hirsch MS. A case of adult metastatic Xp11 translocation renal cell carcinoma treated successfully with sunitinib. Clin Genitourin Cancer, 2009). 7(3):E93–4., DOI 10.3816/CGC.2009. n.031 Classe M, Malouf GG, Su X, Yao H, Thompson EJ, Doss DJ, et al. Incidence, clinicopathological features and fusion transcript landscape of translocation renal cell carcinomas. Histopathology, 2017). 70(7):1089–1097., DOI 10.1111/ his.13167 Dai C, Sheng R, Ding Y, Yang M, Hou J, Zhou J. Magnetic resonance imaging findings of renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusion in adults: a pilot study. Abdom Radiol, NY,, 2019). 44(1):209–217., DOI 10.1007/s00261-018-1703-0 Dang TT, Ziv E, Weinstein S, Meng MV, Wang Z, Coakley FV. Computed tomography and magnetic resonance imaging of adult renal cell carcinoma associated with Xp11.2 translocation. J Comput Assist Tomogr, 2012). 36(6): 669–74., DOI 10.1097/RCT.0b013e3182680182 Dey B, Badhe B, Govindarajan KK, Ramesh RA. Xp11.2 translocation renal cell carcinoma diagnosed by immunohistochemistry and cytogenetics. J Lab Physicians, 2016). 8(2):123–5., DOI 10.4103/0974-2727.180796 Driss M, Boukadi A, Charfi L, Douira W, Mrad K, Bellagha I, et al. Renal cell carcinoma associated with Xp11.2 translocation arising in a horseshoe kidney. Pathology, 2009). 41(6):587–90., DOI 10.1080/00313020903071496 El Naili R, Nicolas M, Gorena A, Policarpio-Nicolas ML. Fine-needle aspiration findings of Xp11 translocation renal cell carcinoma metastatic to a hilar lymph node. Diagn Cytopathol, 2017). 45(5):456–462., DOI 10.1002/dc. 23676 Fukuda H, Kato I, Furuya M, Tanaka R, Takagi T, Kondo T, et al. A novel partner of TFE3 in the Xp11 translocation renal cell carcinoma: clinicopathological analyses and detection of EWSR1-TFE3 fusion. Virchows Arch, 2019). 474(3):389–393., DOI 10.1007/s00428-018-2509-8 Ge L, Tian X, Ma J, Zhao G, Song Y, Zhang S, et al. Surgical treatment for Xp11.2 translocation renal cell carcinoma with venous thrombus: a STROBEcompliant study. Medicine, Baltimore,, 2019). 98(37):e17172., DOI 10.1097/ MD.0000000000017172 Gong P, Zhuang Q, Wang K, Xu R, Chen Y, Wang X, et al. Adult-onset renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusion: 3 case reports and review of literature. Medicine, Baltimore,, 2018). 97(24):e11023., DOI 10.1097/MD.0000000000011023 Gorin MA, Ball MW, Pierorazio PM, Argani P, Allaf ME. Partial nephrectomy for the treatment of translocation renal cell carcinoma. Clin Genitourin Cancer, 2015). 13(3):e199–201., DOI 10.1016/j.clgc.2014.12.008 Green WM, Yonescu R, Morsberger L, Morris K, Netto GJ, Epstein JI, et al. Utilization of a TFE3 break-apart FISH assay in a renal tumor consultation service. Am J Surg Pathol, 2013). 37(8):1150–63., DOI 10.1097/PAS. 0b013e31828a69ae Haudebourg J, Hoch B, Fabas T, Burel-Vandenbos F, Carpentier X, Amiel J, et al. A novel case of t(X;1)(p11.2;p34, in a renal cell carcinoma with TFE3 rearrangement and favorable outcome in a 57-year-old patient. Cancer Genet Cytogenet, 2010). 200(2):75–8., DOI 10.1016/j.cancergencyto.2010. 03.011 Hayes M, Peckova K, Martinek P, Hora M, Kalusova K, Straka L, et al. Molecular-genetic analysis is essential for accurate classification of renal carcinoma resembling Xp11.2 translocation carcinoma. Virchows Arch, 2015). 466(3):313–22., DOI 10.1007/s00428-014-1702-7 He J, Chen X, Gan W, Zhu B, Fan X, Guo H, et al. Renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusions: clinical experience and literature review. Future Oncol, 2015). 11(24):3243–52., DOI 10.2217/fon. 15.177 Hou MM, Hsieh JJ, Chang NJ, Huang HY, Wang HM, Chuang CK, et al. Response to sorafenib in a patient with metastatic xp11 translocation renal cell carcinoma. Clin Drug Investig, 2010). 30(11):799–804., DOI 10.2165/11537220- 000000000-00000 Hung CC, Pan CC, Lin CC, Lin AT, Chen KK, Chang YH. XP11.2 translocation renal cell carcinoma: clinical experience of Taipei Veterans General Hospital. J Chin Med Assoc, 2011). 74(11):500–4., DOI 10.1016/j. jcma.2011.09.005 Jayasinghe C, Siegler N, Leuschner I, Fleischhack G, Born M, Muller AM. Renal cell carcinoma with Xp11.2 translocation in a 7-year-old boy. Klin Padiatr, 2010). 222(3):187–9., DOI 10.1055/s-0030-1252011 Jing H, Tai Y, Xu D, Yang F, Geng M. Renal cell carcinoma associated with Xp11.2 translocations, report of a case. Urology, 2010). 76(1):156–8., DOI 10. 1016/j.urology.2009.07.1276 Jin M, Parwani A, Li Z, Wakely PE. Cytopathology of Xp11 translocation renal cell carcinoma: a report of 5 cases. J Am Soc Cytopathol, 2020). 9, 2):95–102., DOI 10.1016/j.jasc.2019.10.005 Wang W, Ding J, Li Y, Wang C, Zhou L, Zhu H, et al. Magnetic resonance imaging and computed tomography characteristics of renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusion. Plos One, 2014). 9(6): e99990., DOI 10.1371/journal.pone.0099990 Kakoki K, Miyata Y, Mochizuki Y, Iwata T, Obatake M, Abe K, et al. Long-term treatment with sequential molecular targeted therapy for Xp11.2 translocation renal cell carcinoma: a case report and review of the literature. Clin Genitourin Cancer, 2017). 15(3):e503–e506., DOI 10.1016/j.clgc.2016.12.026 Karashima T, Kuno T, Kuroda N, Satake H, Fukata S, Chikazawa M, et al. Bilateral Xp11.2 translocation renal cell carcinoma: a case report. BMC Urol, 2018). 18(1):106., DOI 10.1186/s12894-018-0419-3 Kato I, Furuya M, Baba M, Kameda Y, Yasuda M, Nishimoto K, et al. RBM10- TFE3 renal cell carcinoma characterised by paracentric inversion with consistent closely split signals in break-apart fluorescence in-situ hybridisation: study of 10 cases and a literature review. Histopathology, 2019). 75(2):254–265., DOI 10.1111/his.13866 Kmetec A, Jeruc J. Xp 11.2 translocation renal carcinoma in young adults; recently classified distinct subtype. Radiol Oncol, 2014). 48(2):197–202., DOI 10. 2478/raon-2013-0077 Koie T, Yoneyama T,Hashimoto Y, Kamimura N, Kusumi T, Kijima H, et al. An aggressive course of Xp11 translocation renal cell carcinoma in a 28- year-old man. Int J Urol, 2009). 16(3):333–5., DOI 10.1111/j.1442-2042.2008. 02231.x Komai Y, Fujiwara M, Fujii Y, Mukai H, Yonese J, Kawakami S, et al. Adult Xp11 translocation renal cell carcinoma diagnosed by cytogenetics and immunohistochemistry. Clin Cancer Res, 2009). 15(4):1170–6., DOI 10.1158/ 1078-0432.CCR-08-1183 Koo HJ, Choi HJ, Kim MH, Cho KS. Radiologic-pathologic correlation of renal cell carcinoma associated with Xp11.2 translocation. Acta Radiol, 2013). 54(7): 827–34., DOI 10.1177/0284185113484019 Kuroda N, Tamura M, Tanaka Y, Hes O,Michal M, Inoue K, et al. Adult-onset renal cell carcinoma associated with Xp11.2 translocations/TFE3 gene fusion with smooth muscle stroma and abnormal vessels. Pathol Int, 2009). 59(7): 486–91., DOI 10.1111/j.1440-1827.2009.02398.x Kuthi L, Somoracz A, Micsik T, Jenei A, Hajdu A, Sejben I, et al. Clinicopathological findings on 28 cases with XP11.2 renal cell carcinoma. Pathol Oncol Res, 2020). 26:2123–2133., DOI 10.1007/s12253-019-00792-0 Michalova K, Tretiakova M, Pivovarcikova K, Alaghehbandan R, Perez Montiel D, Ulamec M, et al. Expanding the morphologic spectrum of chromophobe renal cell carcinoma: a study of 8 cases with papillary architecture. Ann Diagn Pathol, 2020). 44:151448., DOI 10.1016/j. anndiagpath.2019.151448 Liang W, Xu S. Xp11.2 translocation renal cell carcinoma with egg-shell calcification mimicking a benign renal tumour: a case report. Oncol Lett, 2015). 10(5):3191–3194., DOI 10.3892/ol.2015.3718 Lim B, You D, Jeong IG, Kwon T, Hong S, Song C, et al. Clinicopathological features of Xp11.2 translocation renal cell carcinoma. Korean J Urol, 2015). 56(3):212–7., DOI 10.4111/kju.2015.56.3.212 Iinuma K, Kojima K, Okamoto K, Yuhara K. [A case of Xp.11.2 traslocational renal cell carcinoma diagnosed by fluorescence in Situ hybridization, FISH)]. Hinyokika kiyo, 2016). 62:411–4., DOI 10.14989/ActaUrolJap_62_8_411 Liu C, Zhang W, Song H. Nephron-sparing surgery in the treatment of pediatric renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusions. J Pediatr Surg, 2017). 52(9):1492–1495., DOI 10.1016/j. jpedsurg.2017.03.052 Liu J, Su Z, Li Y, Chen D, Ni L, Mao X, et al. Xp11.2 translocation renal cell carcinoma with multiple bone metastases: a case report. Oncol Lett, 2016). 11(3):2233–2236., DOI 10.3892/ol.2016.4211 Liu N, Wang Z, Gan W, Xiong L, Miao B, Chen X, et al. Renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusions: clinical features, treatments and prognosis. PLoS One, 2016). 11(11):e0166897., DOI 10.1371/ journal.pone.0166897 Ma J, Pan C, Yin M. Translocation renal cell carcinoma in a child previously treated for infantile fibrosarcoma. Pediatr Dev Pathol, 2018). 21(4):418–422., DOI 10.1177/1093526617707849 Malouf GG, Camparo P, Oudard S, Schleiermacher G, Theodore C, Rustine A, et al. Targeted agents in metastatic Xp11 translocation/TFE3 gene fusion renal cell carcinoma, RCC): a report from the Juvenile RCC Network. Ann Oncol, 2010). 21(9):1834–8., DOI 10.1093/annonc/mdq029 Masago T, Kobayakawa S, Ohtani Y, Taniguchi K, Naka T, Kuroda N, et al. Xp11.2 translocation renal cell carcinoma with TFE3 gene fusion in the elderly: case report and literature review. Int Cancer Conf J, 2020). 9(4), 182–186., DOI 10.1007/s13691-020-00430-6 Meyer PN, Clark JI, Flanigan RC, Picken MM. Xp11.2 translocation renal cell carcinoma with very aggressive course in five adults. Am J Clin Pathol, 2007). 128(1):70–9., DOI 10.1309/LR5G1VMXPY3G0CUK Mir MC, Trilla E, de Torres IM, Panizo A, Zlotta AR, Van Rhijn B, et al. Altered transcription factor E3 expression in unclassified adult renal cell carcinoma indicates adverse pathological features and poor outcome. BJU Int, 2011). 108(2):E71–6., DOI 10.1111/j.1464-410X.2010.09818.x Morii A, Fujiuchi Y, Nomoto K, Komiya A, Fuse H. Rapidly progressing renal cell carcinoma associated with Xp11.2 translocations: a case report. JMed Case Rep, 2012). 6:164., DOI 10.1186/1752-1947-6-164 Nishimura K, Takagi T, Toda N, Yamamoto T, Kondo T, Ishida H, et al. A case of metastatic Xp11.2 translocation renal cell carcinoma successfully managed by cytoreductive nephrectomy followed by axitinib therapy. Mol Clin Oncol, 2017). 6(3):362–364., DOI 10.3892/mco.2017.1142 Pan X, Quan J, Zhao L, Li W, Wei B, Yang S, et al. Xp11.2 translocation renal cell carcinoma with TFE3 gene fusion: a case report. Mol Clin Oncol, 2018). 8(1):83–85., DOI 10.3892/mco.2017.1497 Parikh J, Coleman T, Messias N, Brown J. Temsirolimus in the treatment of renal cell carcinoma associated with Xp11.2 translocation/TFE gene fusion proteins: a case report and review of literature. Rare Tumors, 2009). 1(2):e53., DOI 10.4081/rt.2009.e53 Pwint TP, Macaulay V, Roberts IS, Sullivan M, Protheroe A. An adult Xp11.2 translocation renal carcinoma showing response to treatment with sunitinib. Urol Oncol, 2011). 29(6):821–4., DOI 10.1016/j.urolonc.2009. 10.007 Qu Y, Gu C, Wang H, Chang K, Yang X, Zhou X, et al. Diagnosis of adults Xp11.2 translocation renal cell carcinoma by immunohistochemistry and FISH assays: clinicopathological data from ethnic Chinese population. Sci Rep, 2016). 6:21677., DOI 10.1038/srep21677 Rais-Bahrami S, Drabick JJ, De Marzo AM, Hicks J, Ho C, Caroe AE, et al. Xp11 translocation renal cell carcinoma: delayed but massive and lethal metastases of a chemotherapy-associated secondary malignancy. Urology, 2007). 70(1), 178–6., DOI 10.1016/j.urology.2007.03.037 Rao Q, Chen JY, Wang JD, Ma HH, Zhou HB, Lu ZF, et al. Renal cell carcinoma in children and young adults: clinicopathological, immunohistochemical, and VHL gene analysis of 46 cases with follow-up. Int J Surg Pathol, 2011). 19(2):170–9., DOI 10.1177/1066896909354337 Rua Fernandez OR, Escala Cornejo R, Navarro Martin M, Garcia Munoz M, Antunez Plaza P, Garcia Dominguez AR, et al. Renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene-fusion: a long response to mammalian target of rapamycin, mTOR, inhibitors. Urology, 2018). 117:41–43., DOI 10. 1016/j.urology.2018.03.032 Song HC, Sun N, ZhangWP, He L, Fu L, Huang C. Biological characteristics of pediatric renal cell carcinoma associated with Xp11.2 translocations/TFE3 gene fusions. J Pediatr Surg, 2014). 49(4):539–42., DOI 10.1016/j.jpedsurg.2013. 10.005 Su HH, Sung MT, Chiang PH, Cheng YT, Chen YT. The preliminary experiences of translocation renal cell carcinoma and literature review. Kaohsiung J Med Sci, 2014). 30(8):402–8., DOI 10.1016/j.kjms.2014. 03.003 Sudour-Bonnange H, Leroy X, Chauvet MP, Classe M, Robin PM, Leblond P. Cutaneous metastases during an aggressive course of Xp11.2 translocation renal cell carcinoma in a teenager. Pediatr Blood Cancer, 2014). 61(9): 1698–700., DOI 10.1002/pbc.25015 Sukov WR, Hodge JC, Lohse CM, Leibovich BC, Thompson RH, Pearce KE, et al. TFE3 rearrangements in adult renal cell carcinoma: clinical and pathologic features with outcome in a large series of consecutively treated patients. Am J Surg Pathol, 2012). 36(5):663–70., DOI 10.1097/PAS. 0b013e31824dd972 Taskinlar H, Avlan D, Citak C, Polat A, Nayci A. A rare cause of childhood renal cysts: Xp11.2 translocation renal cell carcinoma. Can Urol Assoc J, 2015). 9(1-2):E36–38., DOI 10.5489/cuaj.2321 Wang Y, Wang Y, Feng M, Lian X, Lei Y, Zhou H. Renal cell carcinoma associated with Xp11.2 translocation/transcription factor E3 gene fusion: an adult case report and literature review. J Int Med Res, 2020). 48: 300060520942095., DOI 10.1177/0300060520942095 Wang Z, Liu N, Gan W, Li X, Zhang G, Li D, et al. Postoperative recurrence of adult renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusion. J Int Med Res, 2017). 45(4):1287–1296., DOI 10.1177/ 0300060517711358 Xia QY, Wang XT, Zhan XM, Tan X, Chen H, Liu Y, et al. Xp11 translocation renal cell carcinomas, RCCs, with RBM10-TFE3 gene fusion demonstrating melanotic features and overlapping morphology with t(6;11, RCC: interest and diagnostic pitfall in detecting a paracentric inversion of TFE3. Am J Surg Pathol, 2017). 41(5):663–676., DOI 10.1097/PAS.0000000000000837 Xia QY, Wang Z, Chen N, Gan HL, Teng XD, Shi SS, et al. Xp11.2 translocation renal cell carcinoma with NONO-TFE3 gene fusion: morphology, prognosis, and potential pitfall in detecting TFE3 gene rearrangement. Mod Pathol, 2017). 30(3):416–426., DOI 10.1038/modpathol.2016.204 Xu L, Yang R, Wang W, Zhang Y, Gan W. Laparoscopic radiofrequency ablation-assisted enucleation of Xp11.2 translocation renal cell carcinoma: a case report. Oncol Lett, 2014). 8(3):1237–1239., DOI 10. 3892/ol.2014.2267 Zhong M, De Angelo P, Osborne L, Paniz-Mondolfi AE, Geller M, Yang Y, et al. Translocation renal cell carcinomas in adults: a single-institution experience. Am J Surg Pathol, 2012). 36(5):654–62., DOI 10.1097/PAS. 0b013e31824f24a6 Zhu G, Qiu X, Chen X, Liu G, Zhang G, Gan W, et al. Cardiopulmonary bypass-assisted surgery for the treatment of Xp11.2 translocation/TFE3 gene fusion renal cell carcinoma with a tumor thrombus within the inferior vena cava: a case report. Oncol Lett, 2015). 10(6):3532–3534., DOI 10.3892/ol.2015. 3739 Zou H, Kang X, Pang LJ, Hu W, Zhao J, Qi Y, et al. Xp11 translocation renal cell carcinoma in adults: a clinicopathological and comparative genomic hybridization study. Int J Clin Exp Pathol, 2014). 7(1):236–45., DOI 10.1016/ j.prp.2014.02.001 Wang XT, Xia QY, Ye SB, Wang X, Li R, Fang R, et al. RNA sequencing of Xp11 translocation-associated cancers reveals novel gene fusions and distinctive clinicopathologic correlations. Mod Pathol, 2018). 31;1346–1360., DOI 10.1038/s41379-018-0051-5 Huang HY, Lui MY, Ladanyi M. Nonrandom cell-cycle timing of a somatic chromosomal translocation: the t(X;17, of alveolar soft-part sarcoma occurs in G2. Genes Chromosomes Cancer, 2005). 44(2):170–6., DOI 10. 1002/gcc.20229 Argani P, Lal P, Hutchinson B, Lui MY, Reuter VE, Ladanyi M. Aberrant nuclear immunoreactivity for TFE3 in neoplasms with TFE3 gene fusions: a sensitive and specific immunohistochemical assay. Am J Surg Pathol, 2003). 27:750–61., DOI 10.1097/00000478-200306000-00005 Calio A, Segala D, Munari E, Brunelli M, Martignoni G. MiT family translocation renal cell carcinoma: from the early descriptions to the current knowledge. Cancers, 2019). 11(8):1110., DOI 10.3390/ cancers11081110 Asaki HE, Moshero G, Stanton ML, Humphreys MR. Xp11.2 translocation tumor: a rare cause of gross hematuria. JAAPA, 2014). 27(2):24–7., DOI 10. 1097/01.JAA.0000442700.87975.0b Kuroda N, Mikami S, Pan CC, Cohen RJ, Hes O, Michal M, et al. Review of renal carcinoma associated with Xp11.2 translocations/TFE3 gene fusions with focus on pathobiological aspect. Histol Histopathol, 2012). 27(2):133–40., DOI 10.14670/HH-27.133 Ellis CL, Eble JN, Subhawong AP, Martignoni G, Zhong M, Ladanyi M, et al. Clinical heterogeneity of Xp11 translocation renal cell carcinoma: impact of fusion subtype, age, and stage. Mod Pathol, 2014). 27(6):875–86., DOI 10.1038/ modpathol.2013.208 Kim SP, Campbell SC, Gill I, Lane BR, Van Poppel H, Smaldone MC, et al. Collaborative review of risk benefit trade-offs between partial and radical nephrectomy in the management of anatomically complex renal masses. Eur Urol, 2017). 72(1):64–75., DOI 10.1016/j.eururo.2016.11.038 Tan HJ,Norton EC, Ye Z, Hafez KS, Gore JL,MillerDC. Long-termsurvival following partial vs radical nephrectomy among older patients with earlystage kidney cancer. JAMA, 2012). 307(15):1629–35., DOI 10.1001/jama. 2012.475 Agizamhan S, Qu F, Liu N, Sun J, Xu W, Zhang L, et al. Preoperative neutrophil-to-lymphocyte ratio predicts the surgical outcome of Xp11.2 translocation/TFE3 renal cell carcinoma patients. BMC Urol, 2018). 18(1): 60., DOI 10.1186/s12894-018-0374-z Kakoki K, Miyata Y, Mochizuki Y, Iwata T, Obatake M, Abe K, et al. Longterm treatment with sequential molecular targeted therapy for Xp11.2 translocation renal cell carcinoma: a case report and review of the literature. Clin Genitourin Cancer, 2017). 15(3):e503–06., DOI 10.1016/j. clgc.2016.12.026 Choueiri TK, Lim ZD, Hirsch MS, Tamboli P, Jonasch E, McDermott DF, et al. Vascular endothelial growth factor-targeted therapy for the treatment of adult metastatic Xp11.2 translocation renal cell carcinoma. Cancer, 2010). 116(22): 5219–25., DOI 10.1002/cncr.25512 Kauffman EC, Lang M, Rais-Bahrami S, et al. Preclinical efficacy of dual mTORC1/2 inhibitor AZD8055 in renal cell carcinoma harboring a TFE3 gene fusion. BMC Cancer, 2019). 19(1):917., DOI 10.1186/s12885-019- 6096-0 Rua Fernandez OR, Escala Cornejo R, Navarro Martin M, Garcia Munoz M, Antunez Plaza P, Garcia Dominguez AR, et al. Renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene-fusion: a long response to mammalian target of rapamycin, mTOR, inhibitors. Urology, 2018). 117: 41–43., DOI 10.1016/j.urology.2018.03.032 Calio A, Brunelli M, Segala D, Pedron S, Remo A, Ammendola S, et al. Comprehensive analysis of 34 MiT family translocation renal cell carcinomas and review of the literature: investigating prognostic markers and therapy targets. Pathology, 2020). 52(3):297–309., DOI 10. 1016/j.pathol.2019.11.006 Chipollini J, AziziM, PeytonCC, TangDH,Dhillon J, Spiess PE. Implications of programmed death ligand-1 positivity in non-clear cell renal cell carcinoma. J Kidney Cancer VHL, 2018). 5(4):6–13., DOI 10.15586/jkcvhl.2018.107 Damayanti NP, Budka JA, Khella HWZ, Ferris MW, Ku SY, Kauffman E, et al. Therapeutic targeting of TFE3/IRS-1/PI3K/mTOR Axis in translocation renal cell carcinoma. Clin Cancer Res, 2018). 24, 23):5977–5989., DOI 10.1158/ 1078-0432.CCR-18-0269 Chang K, Qu Y, Dai B, Zhao JY, Gan H, Shi G, et al. PD-L1 expression in Xp11.2 translocation renal cell carcinoma: indicator of tumor aggressiveness. Sci Rep, 2017). 7(1):2074., DOI 10.1038/s41598-017-02005-7 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 610360 EP 610368 DI 10.3389/pore.2021.610360 PG 9 ER PT J AU Mokanszki, A Badon, SE Monus, A Toth, L Bittner, N Mehes, G AF Mokanszki, Attila Badon, Sarolta Emese Monus, Aniko Toth, Laszlo Bittner, Nora Mehes, Gabor TI Cell-free DNA From Pleural Effusion Samples: Is It Right for Molecular Testing in Lung Adenocarcinoma? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lung carcinoma; pleural effusion fluid; liquid biopsy; cytological cell block; mutation analysis ID lung carcinoma; pleural effusion fluid; liquid biopsy; cytological cell block; mutation analysis AB Pathogenic molecular features gained specific significance in therapeutic decisions in lung carcinoma in the past decade. Initial and follow up genetic testing requres appropriate amounts and quality of tumor derived DNA, but tumor sampling, especially for disease monitoring is generally limited. Further to the peripheral blood (PB), samples from pleural fluid, accumulating in diverse lung processes might serve as an alternative source for cellfree DNA (cfDNA) for genetic profiling. In our study, cfDNA isolated from the pleural effusion and from the PB, and genomic DNA (gDNA) obtained from tissue/cellular samples were analyzed and compared from altogether 65 patients with pulmonary disease, including 36 lung adenocarcinomas. The quantity of effusion cfDNA yield appeared to be significantly higher compared to that from simultaneously collected PB plasma (23.2 vs. 4.8 ng/μl, p < 0.05). Gene mutations could be safely demonstrated from the effusion cfDNA fraction obtained from adenocarcinoma patients, 3/36 EGFR, 9/36 KRAS and 1/36 BRAF gene variants were detected. In this series, 9/13 samples showed an effusion+/plasmamutational status, while only 1/13 samples presented with the opposite findings (effusion-/plasma+). gDNA analysis from sediment cell blocks from the identical effusion sample was surprisingly ineffective for lung adenocarcinoma profiling due to the low DNA yield. In conclusion, the cell free supernatant of pleural effusions appears to concentrate cancer derived cfDNA and seems to be particularly suitable for serial genotyping of pulmonary adenocarcinoma. C1 [Mokanszki, Attila] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Badon, Sarolta Emese] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Monus, Aniko] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Toth, Laszlo] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Bittner, Nora] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. RP Mokanszki, A (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, Debrecen, Hungary. EM mokanszki.attila@med.unideb.hu CR Diaz LA, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature, 2012, 486:537–40., DOI 10.1038/nature11219 Gerlinger M, Rowan AJ, Horswell S, Math M, Larkin J, Endesfelder D, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med, 2012, 366:883–92., DOI 10.1056/NEJMoa1113205 Wang Y, Carlton VEH, Karlin-Neumann G, Sapolsky R, Zhang L, Moorhead M, et al. High quality copy number and genotype data from FFPE samples using Molecular Inversion Probe, MIP, microarrays. BMC Med Genomics, 2009, 2:8., DOI 10.1186/1755-8794-2-8 Lo YMD, Chan KCA, Sun H, Chen EZ, Jiang P, Lun FMF, et al. Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus. Sci Translational Med, 2010, 2:61ra91., DOI 10.1126/scitranslmed.3001720 Ou S-HI, Nagasaka M, Zhu VW. Liquid biopsy to identify actionable genomic alterations. Am Soc Clin Oncol Educ Book, 2018, 38:978–97., DOI 10.1200/ edbk_199765 Rolfo C, Mack PC, Scagliotti GV, Baas P, Barlesi F, Bivona TG, et al. Liquid biopsy for advanced non-small cell lung cancer, nsclc): a statement paper from the iaslc. J Thorac Oncol, 2018, 13:1248–68., DOI 10.1016/j.jtho.2018.05.030 Mehes G. Liquid biopsy for predictive mutational profiling of solid cancer: the pathologist’s perspective. J Biotechnol, 2019, 297:66–70., DOI 10.1016/j.jbiotec. 2019.04.002 Stelcer E, Konkol M, Gle¸boka A, Suchorska WM. Liquid biopsy in oligometastatic prostate cancer-A biologist’s point of view. Front Oncol, 2019, 9:775., DOI 10.3389/fonc.2019.00775 Sriram KB, Relan V, Clarke BE, Duhig EE, Yang IA, Bowman RV, et al. Diagnostic molecular biomarkers for malignant pleural effusions. Future Oncol, 2011, 7:737–52., DOI 10.2217/fon.11.45 Medford A, Medford A, Maskell N. Pleural effusion. Postgrad Med J, 2005, 81: 702–10., DOI 10.1136/pgmj.2005.035352 Ong KC, Indumathi V, Poh WT, Ong YY. The diagnostic yield of pleural fluid cytology in malignant pleural effusions. Singapore Med J, 2000, 41:19–23. Renshaw AA, Dean BR, Cibas ES, Antman KH, Sugarbaker DJ. The role of cytologic evaluation of pleural fluid in the diagnosis of malignant mesothelioma. Chest, 1997, 111:106–9., DOI 10.1378/chest.111.1.106 Husain H, Nykin D, Bui N, Quan D, Gomez G, Woodward B, et al. Cell-free DNA from ascites and pleural effusions: molecular insights into genomic aberrations and disease biology. Mol Cancer Ther, 2017, 16:948–55., DOI 10. 1158/1535-7163.mct-16-0436 Rapisuwon S, Vietsch EE, Wellstein A. Circulating biomarkers to monitor cancer progression and treatment. Comput Struct Biotechnol J, 2016, 14: 211–22., DOI 10.1016/j.csbj.2016.05.004 Li YS, Jiang BY, Yang JJ, Zhang XC, Zhang Z, Ye JY, et al. Unique genetic profiles from cerebrospinal fluid cell-free DNA in leptomeningeal metastases of EGFR-mutant non-small-cell lung cancer: a new medium of liquid biopsy. Ann Oncol, 2018, 29:945–52., DOI 10.1093/annonc/mdy009 Zhou S, Xu B, Qi L, Zhu D, Liu B, Wei J. Next-generation sequencing reveals mutational accordance between cell-free DNA from plasma, malignant pleural effusion and ascites and directs targeted therapy in a gastric cancer patient. Cancer Biol Ther, 2019, 20:15–20., DOI 10.1080/15384047.2018.1504720 Hooper C, Lee YCG, Maskell NBTS Pleural Guideline Group. Investigation of a unilateral pleural effusion in adults: British thoracic society pleural disease guideline 2010. Thorax, 2010, 65(Suppl. 2):ii4-17., DOI 10.1136/thx.2010. 136978 Fenton KN, David Richardson J. Diagnosis and management of malignant pleural effusions. Am J Surg, 1995, 170:69–74., DOI 10.1016/s0002-9610(99, 80257-8 Shin S, Kim J, Kim Y, Cho S-M, Lee K-A. Assessment of real-time PCR method for detection of EGFR mutation using both supernatant and cell pellet of malignant pleural effusion samples from non-small-cell lung cancer patients. Clin Chem Lab Med, 2017, 55:1962–1969., DOI 10.1515/cclm-2016-0851 Carter J, Miller JA, Feller-Kopman D, Ettinger D, Sidransky D, Maleki Z. Molecular profiling of malignant pleural effusion in metastatic non-small-cell lung carcinoma. The effect of preanalytical FactorsThe effect of preanalytical factors. Ann Am Thorac Soc, 2017, 14:1169–76., DOI 10.1513/AnnalsATS. 201609-709OC Liu L, Shao D, Deng Q, Tang H, Wang J, Liu J, et al. Next generation sequencing-based molecular profiling of lung adenocarcinoma using pleural effusion specimens. J Thorac Dis, 2018, 10:2631–7., DOI 10.21037/jtd.2018. 04.125 Yang S-R, Lin C-Y, Stehr H, Long SR, Kong CS, Berry GJ, et al. Comprehensive genomic profiling of malignant effusions in patients with metastatic lung adenocarcinoma. J Mol Diagn, 2018, 20:184–94., DOI 10.1016/j.jmoldx.2017. 10.007 Pinzani P, Salvianti F, Zaccara S, Massi D, De Giorgi V, Pazzagli M, et al. Circulating cell-free DNA in plasma of melanoma patients: qualitative and quantitative considerations. Clinica Chim Acta, 2011, 412:2141–5., DOI 10. 1016/j.cca.2011.07.027 Wang BG, Huang HY, Chen YC, Bristow RE, Kassauei K, Cheng CC, et al. Increased plasma DNA integrity in cancer patients. Cancer Res, 2003, 63: 3966–8. Bronkhorst AJ, Wentzel JF, Aucamp J, van Dyk E, du Plessis L, Pretorius PJ. Characterization of the cell-free DNA released by cultured cancer cells. Biochim Biophys Acta, Bba, - Mol Cel Res, 2016, 1863:157–65., DOI 10. 1016/j.bbamcr.2015.10.022 Goto K, Ichinose Y, Ohe Y, Yamamoto N, Negoro S, Nishio K, et al. Epidermal growth factor receptor mutation status in circulating free DNA in serum: from IPASS, a phase III study of gefitinib or carboplatin/paclitaxel in non-small cell lung cancer. J Thorac Oncol, 2012, 7:115–21., DOI 10.1097/jto. 0b013e3182307f98 Yung TKF, Chan KCA, Mok TSK, Tong J, To K-F, Dennis Lo Y M. Singlemolecule detection of epidermal growth factor receptor mutations in plasma by microfluidics digital PCR in non-small cell lung cancer patients. Clin Cancer Res Off J Am Assoc Cancer Res, 2009, 15:2076–2084., DOI 10.1158/1078-0432. ccr-08-2622 Zhang P, Wu X, Tang M, Nie X, Li L. Detection of EGFR gene mutation status from pleural effusions and other body fluid specimens in patients with lung adenocarcinoma. Thorac Cancer, 2019, 10:2218–24., DOI 10.1111/1759-7714. 13201 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 613071 EP 613077 DI 10.3389/pore.2021.613071 PG 7 ER PT J AU Dastych, M Hubatka, F Turanek-Knotigova, P Masek, J Kroupa, R Raska, M Turanek, J Prochazka, L AF Dastych, Milan Hubatka, Frantisek Turanek-Knotigova, Pavlina Masek, Josef Kroupa, Radek Raska, Milan Turanek, Jaroslav Prochazka, Lubomir TI Overexpression of CD44v8-10 in Colon Polyps—A Possible Key to Early Diagnosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD44 isoforms; colorectal precancerosis; colon polyps; cancer markers; RNA splicing ID CD44 isoforms; colorectal precancerosis; colon polyps; cancer markers; RNA splicing AB Background and aims: The majority of colorectal cancers arise from detectable adenomatous or serrated lesions. Here we demonstrate how deregulated alternative splicing of CD44 gene in diseased colon mucosa results in downregulation of standard isoform of CD44 gene (CD44s) and upregulation of variant isoform CD44v8-10. Our aim is to show that upregulation of CD44v8-10 isoform is a possible marker of precancerous lesion in human colon. Methods: We analysed pairs of fresh biopsy specimen of large intestine in a cohort of 50 patients. We studied and compared alternative splicing profile of CD44 gene in colon polyps and adjoined healthy colon mucosa. We performed end-point and qRT PCR, western blotting, IHC staining and flow cytometry analyses. Results: We detected more than five-fold overexpression of CD44v8-10 isoform and almost twenty-fold downregulation of standard isoform CD44s in colon polyps compared to adjoined healthy tissue with p = 0.018 and p < 0.001 in a cohort of 50 patients. Our results also show that aberrant splicing of CD44 occurs in both biologically distinct subtypes of colorectal adenoma possibly in ESRP-1 specific manner. Conclusion: 92% of the colon polyp positive patients overexpressed CD44v8-10 isoform in their colon polyps while only 36% of them had positive fecal occult blood test which is currently a standard non-invasive screening technique. Impact: We believe that our results are important for further steps leading to application of CD44v8-10 isoform as a biomarker of colorectal precancerosis in non-invasive detection. Early detection of colon precancerosis means successful prevention of colorectal carcinoma. C1 [Dastych, Milan] University Hospital Brno and Faculty of Medicine Masaryk University Brno, Department of Gastroenterology and Internal MedicineBrno, Czech Republic. [Hubatka, Frantisek] Veterinary Research Institute, Department of Pharmacology and ToxicologyBrno, Czech Republic. [Turanek-Knotigova, Pavlina] Veterinary Research Institute, Department of Pharmacology and ToxicologyBrno, Czech Republic. [Masek, Josef] Veterinary Research Institute, Department of Pharmacology and ToxicologyBrno, Czech Republic. [Kroupa, Radek] University Hospital Brno and Faculty of Medicine Masaryk University Brno, Department of Gastroenterology and Internal MedicineBrno, Czech Republic. [Raska, Milan] Palacky University Olomouc and the University Hospital Olomouc, Faculty of Medicine and Dentistry, Department of ImmunologyOlomouc, Czech Republic. [Turanek, Jaroslav] Veterinary Research Institute, Department of Pharmacology and ToxicologyBrno, Czech Republic. [Prochazka, Lubomir] Veterinary Research Institute, Department of Pharmacology and ToxicologyBrno, Czech Republic. RP Turanek, J (reprint author), Veterinary Research Institute, Department of Pharmacology and Toxicology, Brno, Czech Republic. EM turanek@seznam.cz CR Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JWW, Comber H, et al.Cancer incidence andmortality patterns inEurope: estimates for 40 countries in 2012. Eur J Cancer, 2013). 49(6):1374–403., DOI 10.1016/j.ejca.2012.12.027 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin, 2012). 65(2):87–108., DOI 10.3322/caac.21262 Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, et al. Genetic alterations during colorectal-tumor development. N Engl J Med, 1988). 319(9):525–32., DOI 10.1056/nejm198809013190901 Tomasetti C, Marchionni L, Nowak MA, Parmigiani G, Vogelstein B. Only three driver gene mutations are required for the development of lung and colorectal cancers. Proc Natl Acad Sci USA, 2015). 112(1):118–23., DOI 10.1073/pnas. 1421839112 Zavoral M, Suchanek S, Majek O, Fric P, Minarikova P, Minarik M, et al. Colorectal cancer screening: 20 years of development and recent progress. World J. Gastroenterol, 2014). 20(14):3825–34., DOI 10.3748/wjg.v20.i14.3825 Prochazka L, Tesarik R, Turanek J. Regulation of alternative splicing of CD44 in cancer. Cell Signal, 2014). 26(10):2234–9., DOI 10.1016/j.cellsig.2014.07.011 Toole BP. Hyaluronan-CD44 interactions in cancer: paradoxes and possibilities. Clin Cancer Res, 2009). 15(24):7462–8., DOI 10.1158/1078-0432.ccr-09-0479 Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nat Rev Cancer, 2004). 4(7):528–39., DOI 10.1038/nrc1391 Orian-Rousseau V, Ponta H. Perspectives of CD44 targeting therapies. Arch Toxicol, 2015). 89(1):3–14., DOI 10.1007/s00204-014-1424-2 Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci, 2003). 100(7):3983–8., DOI 10.1073/pnas.0530291100 Takeo K, Kawai T, Nishida K, Masuda K, Teshima-Kondo S, Tanahashi T, et al. Oxidative stress-induced alternative splicing of transformer 2β, SFRS10, and CD44 pre-mRNAs in gastric epithelial cells. Am J Physiology-Cell Physiol, 2009). 297(2):C330–C338., DOI 10.1152/ajpcell. 00009.2009 Batsche E, Yaniv M, Muchardt C. The human SWI/SNF subunit Brm is a regulator of alternative splicing. Nat Struct Mol Biol, 2006). 13(1):22–9., DOI 10. 1038/nsmb1030 Cappellari M, Bielli P, Paronetto MP, Ciccosanti F, Fimia GM, Saarikettu J, et al. The transcriptional co-activator SND1 is a novel regulator of alternative splicing in prostate cancer cells. Oncogene, 2014). 33(29):3794–802., DOI 10. 1038/onc.2013.360 Ameyar-Zazoua M, Rachez C, Souidi M, Robin P, Fritsch L, Young R, et al. Argonaute proteins couple chromatin silencing to alternative splicing. Nat Struct Mol Biol, 2012). 19(10):998–1004., DOI 10.1038/nsmb.2373 Orian-Rousseau V, Chen L, Sleeman JP, Herrlich P, Ponta H. CD44 is required for two consecutive steps in HGF/c-Met signaling. Genes Dev, 2002). 16(23): 3074–86., DOI 10.1101/gad.242602 Brown RL, Reinke LM, Damerow MS, Perez D, Chodosh LA, Yang J, et al. CD44 splice isoform switching in human and mouse epithelium is essential for epithelial-mesenchymal transition and breast cancer progression. J Clin Invest, 2011). 121(3):1064–74., DOI 10.1172/jci44540 Rudy W, HofmannM, Schwartz-Albiez R, Zoller M, Heider KH, Ponta H, et al. The two major CD44 proteins expressed on a metastatic rat tumor cell line are derived from different splice variants: each one individually suffices to confer metastatic behavior. Cancer Res, 1993). 53(6):1262–8. Seiter S, Arch R, Reber S, Komitowski D, Hofmann M, Ponta H, et al. Prevention of tumor metastasis formation by anti-variant CD44. J Exp Med, 1993). 177(2):443–55., DOI 10.1084/jem.177.2.443 Tanabe KK, Ellis LM, Saya H. Expression of CD44R1 adhesion molecule in colon carcinomas and metastases. The Lancet, 1993). 341(8847):725–6., DOI 10. 1016/0140-6736(93)90490-8 Gunthert U, Hofmann M, Rudy W, Reber S, Zoller M, Hauβmann I, et al. A new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell, 1991). 65(1):13–24., DOI 10.1016/0092-8674(91, 90403-l Lau WM, Teng E, Chong HS, Lopez KAP, Tay AYL, Salto-Tellez M, et al. CD44v8-10 is a cancer-specificmarker for gastric cancer stem cells. Cancer Res, 2014). 74(9):2630–41., DOI 10.1158/0008-5472.can-13-2309 Jiang H, Zhao W, Shao W. Prognostic value of CD44 and CD44v6 expression in patients with non-small cell lung cancer: meta-analysis. Tumor Biol, 2014). 35(8):7383–9., DOI 10.1007/s13277-014-2150-3 Yan Y, Zuo X, Wei D. Concise review: emerging role of CD44 in cancer stem cells: a promising biomarker and therapeutic target. Stem Cell Transl Med, 2015). 4(9):1033–43., DOI 10.5966/sctm.2015-0048 Gao AC, Lou W, Dong JT, Isaacs JT. CD44 is a metastasis suppressor gene for prostatic cancer located on human chromosome 11p13. Cancer Res, 1997). 57(5):846–9. Neumayer R, Rosen HR, Reiner A, Sebesta C, Schmid A, Tuchler H, et al. CD44 expression in benign and malignant colorectal polyps. Dis Colon Rectum, 1999). 42(1):50–5., DOI 10.1007/bf02235182 Asao T, Nakamura J-i., Shitara Y, Tsutsumi S,Mochiki E, Shimura T, et al. Loss of standard type of CD44 expression in invaded area as a good indicator of lymph-node metastasis in colorectal carcinoma. Dis Colon Rectum, 2000). 43(9):1250–4. ;discussion 1254-5., DOI 10.1007/bf02237430 Miyake H, Eto H, Arakawa S, Kamidono S, Hara I. Over expression of CD44V8-10 in urinary exfoliated cells as an independent prognostic predictor in patients with urothelial cancer. J Urol, 2002). 167(3):1282–7., DOI 10.1016/s0022-5347(05)65282-2 Levine JS, Ahnen DJ. Adenomatous polyps of the colon. N Engl J Med, 2006). 355(24):2551–7., DOI 10.1056/nejmcp063038 Risio M. Reprint of: the natural history of adenomas. Best Pract Res Clin Gastroenterol, 2010). 24(4):397–406., DOI 10.1016/j.bpg.2010.08.002 Lech G, Slotwi´nski R, Slodkowski M, Krasnode˛bski IW. Colorectal cancer tumour markers and biomarkers: recent therapeutic advances. World J. Gastroenterol, 2016). 22(5):1745–55., DOI 10.3748/wjg.v22.i5.1745 Pfaffl MW. A new mathematical model for relative quantification in realtime RT-PCR. Nucleic Acids Res, 2001). 29(9):45e–45., DOI 10.1093/nar/ 29.9.e45 Prochazka L, Turanek J, Tesaˇrik R, Knotigova P, Polaˇskova P, Andrysik Z, et al. Apoptosis and inhibition of gap-junctional intercellular communication induced by LA-12, a novel hydrophobic platinum(IV, complex. Arch Biochem Biophys, 2007). 462(1):54–61., DOI 10.1016/j.abb.2007.03.021 Iida J, Clancy R, Dorchak J, Somiari RI, Somiari S, Cutler ML, et al. DNA aptamers against exon v10 of CD44 inhibit breast cancer cell migration. PLoS One, 2014). 9(2):e88712., DOI 10.1371/journal.pone.0088712 Warzecha CC, Sato TK, Nabet B, Hogenesch JB, Carstens RP. ESRP1 and ESRP2 are epithelial cell-type-specific regulators of FGFR2 splicing. Mol Cel, 2009). 33(5):591–601., DOI 10.1016/j.molcel.2009.01.025 Bartheldyova E, Effenberg R, Maˇsek J, Prochazka L, Knotigova PT, Kulich P, et al. Hyaluronic acid surface modified liposomes prepared via orthogonal aminoxy coupling: synthesis of nontoxic aminoxylipids based on symmetrically α-branched fatty acids, preparation of liposomes by microfluidic mixing, and targeting to cancer cells expressing CD44. Bioconjug Chem., 2018). 29(7):2343–56., DOI 10.1021/acs.bioconjchem. 8b00311 Wielenga VJ, Heider KH, Offerhaus GJ, Adolf GR, van den Berg FM, Ponta H, et al. Expression of CD44 variant proteins in human colorectal cancer is related to tumor progression. Cancer Res, 1993). 53(20):4754–6. Kopp R, Fichter M, Schalhorn G, Danescu J, Classen S. Frequent expression of the high molecular, 673-bp CD44v3,v8-10 variant in colorectal adenomas and carcinomas. Int J Mol Med, 2009). 24(5):677–83., DOI 10.3892/ijmm_00000279 Makinen MJ. Colorectal serrated adenocarcinoma. Histopathology, 2007). 50(1):131–50., DOI 10.1111/j.1365-2559.2006.02548.x Konishi K, Yamochi T, Makino R, Kaneko K, Yamamoto T, Nozawa H, et al. Molecular differences between sporadic serrated and conventional colorectal adenomas. Clin Cancer Res, 2004). 10(9):3082–90., DOI 10.1158/1078-0432.ccr- 03-0334 Muinuddin A, Aslahi R, Hopman WM, Paterson WG. Relationship between the number of positive fecal occult blood tests and the diagnostic yield of colonoscopy. Can J Gastroenterol, 2013). 27(2):90–4., DOI 10.1155/2013/ 612314 Wei SC, Tsao PN, Wang YT, Lin BR, Wu DC, Tsai WS, et al. Using serum placenta growth factor could improve the sensitivity of colorectal cancer screening in fecal occult blood negative population: a multicenter with independent cohort validation study. Cancer Med, 2019). 8(7):3583–91., DOI 10.1002/cam4.2216 Mashayekhi F, Aryaee H, Mirzajani E, Yasin AA, Fathi A. Soluble CD44 concentration in the serum and peritoneal fluid samples of patients with different stages of endometriosis. Arch Gynecol Obstet, 2015). 292(3):641–5., DOI 10.1007/s00404-015-3654-9 Palagani V, El Khatib M, Krech T, Manns MP, Malek NP, Plentz RR. Decrease of CD44-positive cells correlates with tumor response to chemotherapy in patients with gastrointestinal cancer. Anticancer Res, 2012). 32(5):1747–55. Seyedmajidi S, SeyedmajidiM, Foroughi R, Zahedpasha A, Zolfaghari Saravi Z, Pourbagher R, et al. Comparison of salivary and serum soluble CD44 levels between patients with oral SCC and healthy controls. Asian Pac J Cancer Prev, 2018). 19(11):3059–63., DOI 10.31557/apjcp.2018.19.11.3059 Zeilstra J, Joosten SPJ, Dokter M, Verwiel E, Spaargaren M, Pals ST. Deletion of the WNT target and cancer stem cell marker CD44 in Apc(Min/+, mice attenuates intestinal tumorigenesis. Cancer Res, 2008). 68(10):3655–61., DOI 10. 1158/0008-5472.can-07-2940 Zeilstra J, Joosten SPJ, van Andel H, Tolg C, Berns A, Snoek M, et al. Stem cell CD44v isoforms promote intestinal cancer formation in Apc(min, mice downstream of Wnt signaling. Oncogene, 2014). 33(5):665–70., DOI 10.1038/ onc.2012.611 Guo W, Frenette PS. Alternative CD44 splicing in intestinal stem cells and tumorigenesis. Oncogene, 2014). 33(5):537–8., DOI 10.1038/onc.2013.260 Zhang S, Wu CCN, Fecteau J-F, Cui B, Chen L, Zhang L, et al. Targeting chronic lymphocytic leukemia cells with a humanized monoclonal antibody specific for CD44. Proc Natl Acad Sci, 2013). 110(15):6127–32., DOI 10.1073/ pnas.1221841110 Birzele F, Voss E, Nopora A, Honold K, Heil F, Lohmann S, et al. CD44 isoform status predicts response to treatment with anti-CD44 antibody in cancer patients. Clin Cancer Res, 2015). 21(12):2753–62., DOI 10.1158/1078-0432.ccr- 14-2141 Sy MS, Guo YJ, Stamenkovic I. Distinct effects of two CD44 isoforms on tumor growth in vivo. J Exp Med, 1991). 174(4):859–66., DOI 10.1084/jem. 174.4.859 Bartolazzi A, Peach R, Aruffo A, Stamenkovic I. Interaction between CD44 and hyaluronate is directly implicated in the regulation of tumor development. J Exp Med, 1994). 180(1):53–66., DOI 10.1084/jem.180.1.53 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 614281 EP 614291 DI 10.3389/pore.2021.614281 PG 11 ER PT J AU Simon, Zs Virga, B Pinczes, L Mehes, G Miltenyi, Zs Barna, S Szabo, R Illes, AF Simon, Zsofia Virga, Balint Pinczes, Laszlo Mehes, Gabor Miltenyi, Zsofia Barna, Sandor Szabo, Roxana Illes, Arpad TI Transition Between Diffuse Large B-Cell Lymphoma and Classical Hodgkin Lymphoma– Our Histopathological and Clinical Experience With Patients With Intermediate Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gray zone lymphoma; hodgkin lymphoma; diffuse large B cell lymphoma; brentuximab vedotine; stem cell transplant ID gray zone lymphoma; hodgkin lymphoma; diffuse large B cell lymphoma; brentuximab vedotine; stem cell transplant AB Even though information about the pathophysiology and clinical features of grey-zone lymphoma, an entity intermediate between classical Hodgkin lymphoma and diffuse large B-cell lymphoma, is growing, there are still a number of unanswered questions. The disease has no easily reproducible diagnostic criteria, which makes identification challenging. Uncommon, mixed histological picture and unusual clinical presentation should raise suspicion for grey-zone lymphoma. In this retrospective analysis we present 9 gray zone lymphoma patients, who were diagnosed in our institute between 2008 and 2018. The histological diagnoses was oftentime challenging, we asked for a revision in three cases due to the unusual clinical behavior and in other three cases only the relapse of the disease proved to be grey-zone lymphoma. Based on the initial histopathological diagnoses we applied adriablastine-bleomycine-vinblastine and procarbasine or cyclophosphamide-vincristine-adriablastine and prednisolon as first line chemotherapy regime with additional rituximab in six cases and brentuximabvedotine in one patient. In six of the nine patients due to the primary refractory disease we used rituximab plus cisplatine, cytosine-arabinoside, prednisolone salvage treatment and five of these patients responded well enough to become eligible for autologous stem cell transplantation. One young male patient was refractory for various treatments and died due to the progression of his lymphoma. As a rare disease grey-zone lymphoma has no existing diagnostic criteria or guiedlines for its standard of care, which makes the everyday practice rather challenging for the clinicians, and emphasize the importance of unique decision making in every case and the repeated consultation between the pathologist and hematologist. C1 [Simon, Zsofia] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Virga, Balint] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Pinczes, Laszlo] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Miltenyi, Zsofia] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Barna, Sandor] Scanomed Orvosi Diagnosztikai Kutato es Oktato Kft.Budapest, Hungary. [Szabo, Roxana] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. RP Illes, (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM illesarpaddr@gmail.com CR Traverse-Glehen A, Pittaluga S, Gaulard P, Sorbara L, Alonso MA, Raffeld M, et al. Mediastinal gray zone lymphoma. Am J Surg Pathol, 2005). 29:1411–21., DOI 10.1097/01.pas.0000180856.74572.73 Swerdlow S, Campo E, Lee Harris N, Jaffe E, Pileri S, Stein H, et al. WHO classification of tumours of haematopoetic and lymphoid tissues. 4th ed. Lyon: WHO Press, 2008). Evens AM, Kanakry JA, Sehn LH, Kritharis A, Feldman T, Kroll A, et al. Gray zone lymphoma with features intermediate between classical Hodgkin lymphoma and diffuse large B-cell lymphoma: characteristics, outcomes, and prognostication among a large multicenter cohort. Am J Hematol, 2015). 90: 778–83., DOI 10.1002/ajh.24082 Kritharis A, Pilichowska M, Evens AM How I manage patients with grey zone lymphoma. Br J Haematol, 2016). 174:345–50., DOI 10.1111/bjh.14174 Sarkozy C, Molina T, Ghesquieres H,Michallet A-S, Dupuis J, Damotte D, et al. Mediastinal gray zone lymphoma: clinico-pathological characteristics and outcomes of 99 patients from the Lymphoma Study Association. Haematologica, 2017). 102(1):150–9., DOI 10.3324/haematol. 2016.152256 Pilichowska M, Kritharis A, Evens AM Gray zone lymphoma. Hematology/ Oncology Clin North America, 2016). 30(6):1251–60., DOI 10.1016/j.hoc.2016. .006 7. Mallipudi RM, Alqzran L, Shenoy VA, Leslie LA, Conti JA A rare case of grey zone lymphoma successfully treated with brentuximab vedotin and R-CHP chemotherapy. Case Rep Oncol Med, 2019). 1–4., DOI 10.1155/2019/ 4121234 Magyari F, Barna S, Miltenyi Z, Rajnai H, Csomor J, Udvardy M, et al. Histopathology difficulties in an adolescent lymphoma patient. Path Oncol Res, 2014). 21(1):2013–217., DOI 10.1007/s12253-014-9810-x Svoboda J, Landsburg DJ, Nasta SD, et al. Brentuximab vedotin with R-CHP chemotherapy as frontline treatment for patients with Cd30 positive primary mediastinal large B-cell, diffuse large B-cell, and grey zone lymphomas: results of a phase I/II multisite trial. Blood, 2017). 130(Suppl. 1):191., DOI 10.3324/ haematol.2019.238675 Berger GK, McBride A, LAwson S, Royball K, Yun S, Gee K, et al. Brentuximab vedotin for treatment of non-Hodgkin lymphomas: a systematic review. Crit Rev Oncology/Hematology, 2017). 109:42–50., DOI 10.1016/j.critrevonc.2016. 009 11. Takaishi K, Muto T, Mimura N, Takiguchi J, Nagao Y, Oshima-Hasegawa N, et al. Long-term complete remission following tandem autologous stem cell transplantation and consolidative radiotherapy for refractory mediastinal gray-zone lymphoma. Int J Hematol, 2018). 108:452–5., DOI 10.1007/s12185- 018-2471-x Melani C, Major A, Schowinsky J, Roschewski M, Pittaluga S, Jaffe ES, et al. PD-1 blockade in mediastinal gray-zone lymphoma. N Engl J Med, 2017). 377(1):89–91., DOI 10.1056/nejmc1704767 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 625529 EP 625535 DI 10.3389/pore.2021.625529 PG 7 ER PT J AU Mu, W Su, P Ning, Sh AF Mu, Wentao Su, Peng Ning, Shanglei TI Case Report: Incidentally Discovered a Rare Cystic Lesion of Liver: Multicystic Biliary Hamartoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE multicystic biliary hamartoma; cystic lesion of liver; laparoscopic resection; liver; immunohistochemistry ID multicystic biliary hamartoma; cystic lesion of liver; laparoscopic resection; liver; immunohistochemistry AB Multicystic biliary hamartoma (MCBH) is an extremely rare cystic lesion of the liver. A 37-year old male patient was admitted to our hospital for incidentally discovered hepatic cystic lesions on abdominal ultrasonography. Abdominal contrast-enhanced computed tomography (CT) showed a multilocular cystic lesion in the segment VI, with mild enhancement in the septae and peripheral wall within the lesion. Only alanine transaminase (ALT) and carbohydrate antigen 19–9 (CA19–9) increased slightly above normal value. Preoperative tests suggested possibility of a benign mucinous cystic neoplasm (MCN) or intraductal papillary neoplasm of the bile duct (IPNB). Laparoscopic complete resection of the lesion was performed. Histopathological examination showed numerous variably sized ductal structures surrounded by periductal glands and fibrous connective tissues containing small blood vessels and smooth muscle bundles. Immunohistochemical staining (IHC) revealed that dilated ducts were positive for cytokeratin CK19, characteristic for biliary tract. Histopathological findings confirmed diagnosis of multicystic biliary hamartoma (MCBH). No recurrence occurred during 6 months follow-up. In conclusion, MCBH should be differentiating from hepatic cystic lesion and could be resected laparoscopically safely. C1 [Mu, Wentao] Qilu Hospital of Shandong University, Department of Hepatobiliary Surgery, General SurgeryJinan, China. [Su, Peng] Shandong University, Qilu Hospital, Department of PathologyJinan, China. [Ning, Shanglei] Qilu Hospital of Shandong University, Department of Hepatobiliary Surgery, General SurgeryJinan, China. RP Ning, Sh (reprint author), Qilu Hospital of Shandong University, Department of Hepatobiliary Surgery, General Surgery, Jinan, China. EM shangleining@163.com CR Beard R. E., Yee E. U., Mortele K. J., Khwaja K., 2014). Multicystic biliary hamartoma: A report of a rare entity and a review of the literature. Int. J. Surg. Case Rep. 5, 12), 919–923., DOI 10.1016/j.ijscr.2014.10.014 Fernandez-Carrion M. J., Robles Campos R., Lopez Conesa A., Brusadin R., Parrilla Paricio P., 2015). Intrahepatic multicystic biliary hamartoma: Presentation of a case report. Cir. Esp. 93, 9), e103-5., DOI 10.1016/j.ciresp. 2014.02.015 Caroli J., Soupault R., Kossakowski J., Plocker L., Paradowska, 1958). Congenital polycystic dilation of the intrahepatic bile ducts; attempt at classification. Sem. Hop. 34, 8/2), 488–495. Heinke T, Pellacani L. B., Costa H. d. O., Fuziy R. A., Franco M., 2008). Hepatocellular carcinoma in association with bile duct hamartomas: report on 2 cases and review of the literature. Ann. Diagn. Pathol. 12, 3), 208–211., DOI 10.1016/j.anndiagpath.2006.12.003 Kai K., Takahashi T., Miyoshi A., Yasui T., Tokunaga O., Miyazaki K., 2008). Intrahepatic multicystic biliary hamartoma: report of a case. Hepatol. Res. 38, 6), 629–634., DOI 10.1111/j.1872-034x.2007.00314.x Kobayashi A., Takahashi S., Hasebe T, Konishi M, Nakagohri T, Gotohda N, et al., 2005). Solitary bile duct hamartoma of the liver. Scand. J. Gastroenterol. 40, 11), 1378–1381., DOI 10.1080/00365520510023387 Morinaga T., Imai K., Yamashita Y.-I., Yamao T., Kaida T.,Nakagawa S., et al., 2017). Multicystic biliary hamartoma with extremely elevated CA19-9: a case report. Scand. J. Gastroenterol. 52, 8), 916–919., DOI 10.1080/00365521.2017.1322140 Ogura T., Kurisu Y., Miyano A., Higuchi K., 2018). A huge rapidly-enlarging multicystic biliary hamartoma. Dig. Liver Dis. 50, 7), 723. Pitchaimuthu M., Duxbury M., 2017). Cystic lesions of the liver-A review. Curr. Probl. Surg. 54, 10), 514–542., DOI 10.1067/j.cpsurg.2017.09.001 Ryu Y., Matsui O., Zen Y., Ueda K., Abo H., Nakanuma Y., et al., 2010). Multicystic biliary hamartoma: imaging findings in four cases. Abdom Imaging 35, 5), 543–547., DOI 10.1007/s00261-009-9566-z Shi Q. S., Xing L. X., Jin L. F., Wang H., Lv X. H., Du L. F., et al., 2015). Imaging findings of bile duct hamartomas: a case report and literature review. Int. J. Clin. Exp. Med. 8, 8), 13145–13153. Song J. S., NohS. J., ChoB.H.,MoonW. S., 2013).Multicystic biliary hamartoma of the liver. Korean J. Pathol. 47, 3), 275–278., DOI 10.4132/koreanjpathol. 2013.47.3.275 Stocker J. T., Ishak K. G., 1983). Mesenchymal hamartoma of the liver: report of 30 cases and review of the literature. Pediatr. Pathol. 1, 3), 245–267., DOI 10.3109/ 15513818309040663 Sugawara T., Shindoh J., Hoshi D., Hashimoto M., 2018). Intrahepatic cholangiocarcinoma and portal hypertension developing in a patient with multicystic biliary microhamartomas. Malays. J. Pathol. 40, 3), 331–335. Tominaga T., Abo T., KinoshitaN.,Murakami T., Sato Y.,Nakanuma Y., et al., 2015). A variant of multicystic biliary hamartoma presenting as an intrahepatic cystic neoplasm. Clin. J. Gastroenterol. 8, 3), 162–166., DOI 10.1007/s12328-015-0574-y Yoh T., Okamura R., Nakayama H., Lin X., Nakamura Y., Kato T., 2014). Multicystic biliary hamartoma mimicking intrahepatic cholangiocarcinoma: Report of a case. Clin. J. Gastroenterol. 7, 5), 418–421., DOI 10.1007/s12328-014-0513-3 Zen Y., Terahata S., Miyayama S., Mitsui T., Takehara A., Miura S., et al., 2006). Multicystic biliary hamartoma. Hum. Pathol. 37, 3), 339–344., DOI 10.1016/j. humpath.2005.11.008 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 628323 EP 628327 DI 10.3389/pore.2021.628323 PG 5 ER PT J AU Deng, Y Zhao, Y Qin, J Huang, X Wu, R Zhou, C Pan, Z AF Deng, Yuxiang Zhao, Yujie Qin, Jiayi Huang, Xiaozhen Wu, Ruomei Zhou, Caixia Pan, Zhizhong TI Prognostic Value of the C-Reactive Protein/Albumin Ratio and Systemic Immune-Inflammation Index for Patients With Colorectal Liver Metastasis Undergoing Curative Resection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal neoplasms; liver metastases; prognosis; CAR; SII ID colorectal neoplasms; liver metastases; prognosis; CAR; SII AB Background: We evaluated the prognostic value of C-reactive protein/albumin (CAR) and systemic immune-inflammation index (SII), which we calculated as neutrophil × platelet/ lymphocyte) in patients with colorectal liver metastasis (CRLM) after curative resection. Methods: We retrospectively enrolled 283 consecutive patients with CRLM who underwent curative resection between 2006 and 2016. We determined the optimal cutoff values of CAR and SII using receiver operating curve (ROC) analysis. Overall survival (OS)- and recurrence-free survival (RFS)-related to CAR and SII were analyzed using the log-rank test and multivariate Cox regression methods. Results: We found that a high CAR was significantly associated with poor OS (P < 0.001) and RFS (P = 0.008) rates compared with a low CAR; a high SII was significantly associated with poor RFS (P = 0.003) rates compared with a low SII. The multivariate analysis indicated that CAR was an independent predictor of OS (hazard ratio [HR] = 2.220; 95% confidence interval [CI] = 1.387–3.550; P = 0.001) and RFS (HR = 1.494; 95% CI = 1.086–2.056; P = 0.014). The SII was an independent predictor of RFS (HR = 1.973; 95% CI = 1.230–3.162; P = 0.005) in patients with CRLM. Conclusion: We proved that CAR was an independent predictor of OS and RFS in patients with CRLM who underwent curative resection, and that the prognostic value of CAR was superior to that of SII. C1 [Deng, Yuxiang] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Zhao, Yujie] Peking University Shenzhen Hospital, Department of Radiation OncologyShenzhen, China. [Qin, Jiayi] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Huang, Xiaozhen] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Wu, Ruomei] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Zhou, Caixia] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Pan, Zhizhong] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. RP Pan, Z (reprint author), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal Surgery, Guangzhou, China. EM panzhzh@sysucc.org.cn CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Ca: A Cancer J Clinicians, 2018). 68:394–424., DOI 10.3322/caac.21492 Van Cutsem E, Nordlinger B, Cervantes A. Advanced colorectal cancer: ESMO clinical practice guidelines for treatment. Ann Oncol, 2010). 21(Suppl. 5): v93–v97., DOI 10.1093/annonc/mdq222 Kanas GP, Taylor A, Primrose JN, Langeberg WJ, Kelsh MA, Mowat FS, et al. Survival after liver resection in metastatic colorectal cancer: review and metaanalysis of prognostic factors. Clin Epidemiol, 2012). 4:283–301., DOI 10.2147/ CLEP.S34285 Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol, 2016). 27:1386–422., DOI 10.1093/ annonc/mdw235 Gallinger S, Biagi JJ, Fletcher GG, Nhan C, Ruo L, McLeod RS. Liver resection for colorectal cancer metastases. Curr Oncol, 2013). 20:255–65., DOI 10.3747/co. 20.1341 Engels B, Gevaert T, Everaert H, De Coninck P, Sermeus A, Christian N, et al. Phase II study of helical tomotherapy in the multidisciplinary treatment of oligometastatic colorectal cancer. Radiat Oncol, 2012). 7:34., DOI 10.1186/1748- 717X-7-34 Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature, 2008). 454:436–44., DOI 10.1038/nature07205 Schwegler I, von Holzen A, Gutzwiller J-P, Schlumpf R, Muhlebach S, Stanga Z. Nutritional risk is a clinical predictor of postoperative mortality and morbidity in surgery for colorectal cancer. Br J Surg, 2010). 97:92–7., DOI 10.1002/bjs.6805 Saito H, Kono Y, Murakami Y, Shishido Y, Kuroda H, Matsunaga T, et al. Prognostic significance of the preoperative ratio of C-reactive protein to albumin and neutrophil-lymphocyte ratio in gastric cancer patients. World J Surg, 2018). 42:1819–25., DOI 10.1007/s00268-017-4400-1 Chen YY, Zhang JH, Zhang W, Yang ZK, Luo RC, Kang SJ. [C-reactive protein/albumin ratio as a novel inflammation-based prognostic index for predicting outcomes of patients with colorectal cancer]. Nan Fang Yi Ke Da Xue Xue Bao, 2017). 37:622–7. Zhou Q-p., Li X-j. C-reactive protein to albumin ratio in colorectal cancer: a meta-analysis of prognostic value. Dose-Response, 2019). 17:155932581988981., DOI 10.1177/1559325819889814 Wang F, Li P, Li F-s. Prognostic role of C-reactive protein to albumin ratio in colorectal cancer. Medicine, 2019). 98:e16064,., DOI 10.1097/MD.0000000000016064 Fan Y, Xiang S, Dai Z, Zou C, Wang X, Gao Z. Prognostic significance of C-reactive protein to albumin ratio in colorectal cancer patients: a metaanalysis. Int J Colorectal Dis, 2019). 34:1105–11., DOI 10.1007/s00384-019- 03299-x Feng J-F, Chen S, Yang X. Systemic immune-inflammation index, SII, is a useful prognostic indicator for patients with squamous cell carcinoma of the esophagus. Medicine, Baltimore,, 2017). 96:e5886., DOI 10.1097/MD. 0000000000005886 Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow?. The Lancet, 2001). 357:539–45., DOI 10.1016/S0140-6736(00)04046-0 Wu Y, Zhou BP. Inflammation: a driving force speeds cancer metastasis. Cell Cycle, 2009). 8:3267–73., DOI 10.4161/cc.8.20.9699 Chan JCY, Chan DL, Diakos CI, Engel A, Pavlakis N, Gill A, et al. The lymphocyte-to-monocyte ratio is a superior predictor of overall survival in Comparison to established biomarkers of resectable colorectal cancer. Ann Surg, 2017). 265:539–46., DOI 10.1097/SLA.0000000000001743 Asegaonkar SB, Asegaonkar BN, Takalkar UV, Advani S, Thorat AP. C.-reactive protein and breast cancer: new insights from old molecule. Int J Breast Cancer, 2015). 2015:1., DOI 10.1155/2015/145647 Woo HD, Kim K, Kim J. Association between preoperative C-reactive protein level and colorectal cancer survival: a meta-analysis. Cancer Causes Control, 2015). 26:1661–70., DOI 10.1007/s10552-015-0663-8 Thomsen M, Kersten C, Sorbye H, Skovlund E, Glimelius B, Pfeiffer P, et al. Interleukin-6 and C-reactive protein as prognostic biomarkers in metastatic colorectal cancer. Oncotarget, 2016). 7:75013–22., DOI 10.18632/oncotarget. 12601 Chiang JM, Chang CJ, Jiang SF, Yeh CY, You JF, Hsieh PS, et al. Preoperative serum albumin level substantially predicts post-operative morbidity and mortality among patients with colorectal cancer who undergo elective colectomy. Eur J Cancer Care, 2017). 26:e12403., DOI 10. 1111/ecc.12403 Lambert JW, Ingham M, Gibbs BB, Given RW, Lance RS, Riggs SB. Using preoperative albumin levels as a surrogate marker for outcomes after radical cystectomy for bladder cancer. Urology, 2013). 81:587–92., DOI 10.1016/j. urology.2012.10.055 Sonnenschein C, Soto AM, Michaelson CL. Human serum albumin shares the properties of estrocolyone-I, the inhibitor of the proliferation of estrogentarget cells. J Steroid Biochem Mol Biol, 1996). 59:147–54., DOI 10.1016/s0960- 0760(96)00112-4 Shibutani M, Nagahara H, Fukuoka T, Iseki Y, Matsutani S, Wang E, et al. Prognostic significance of the C-reactive protein-to-albumin ratio in patients with metastatic colorectal cancer treated with trifluridine/thymidine phosphorylase inhibitor as later-line chemotherapy. Anticancer Res, 2019). 39:1051–7., DOI 10.21873/anticanres.13212 Sakamoto Y, Mima K, Imai K, Miyamoto Y, Tokunaga R, Akiyama T, et al. Preoperative C-reactive protein-to-albumin ratio and clinical outcomes after resection of colorectal liver metastases. Surg Oncol, 2020). 35, 243–8., DOI 10. 1016/j.suronc.2020.09.014 Hu B, Yang X-R, Xu Y, Sun Y-F, Sun C, Guo W, et al. Systemic immuneinflammation index predicts prognosis of patients after curative resection for hepatocellular carcinoma. Clin Cancer Res, 2014). 20:6212–22., DOI 10.1158/ 1078-0432.CCR-14-0442 Bambace NM, Holmes CE. The platelet contribution to cancer progression. J Thromb Haemost, 2011). 9:237–49., DOI 10.1111/j.1538-7836.2010.04131.x Ferrone C, Dranoff G. Dual roles for immunity in gastrointestinal cancers. Jco, 2010). 28:4045–51., DOI 10.1200/JCO.2010.27.9992 Zhou Z-q., Pang S, Yu X-c., Xue Q, Jiang H-y., Liang X-j., et al. Predictive values of postoperative and dynamic changes of inflammation indexes in survival of patients with resected colorectal cancer. Curr Med Sci, 2018). 38: 798–808., DOI 10.1007/s11596-018-1946-6 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 633480 EP 633489 DI 10.3389/pore.2021.633480 PG 10 ER PT J AU Stockhammer, P Okumus, Hegedus, L Rittler, D Ploenes, T Herold, Th Kalbourtzis, S Bankfalvi, Sucker, A Kimmig, R Aigner, C Hegedus, B AF Stockhammer, Paul Okumus, Ozlem Hegedus, Luca Rittler, Dominika Ploenes, Till Herold, Thomas Kalbourtzis, Stavros Bankfalvi, Agnes Sucker, Antje Kimmig, Rainer Aigner, Clemens Hegedus, Balazs TI HDAC Inhibition Induces Cell Cycle Arrest and Mesenchymal-Epithelial Transition in a Novel Pleural-Effusion Derived Uterine Carcinosarcoma Cell Line SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE uterine carcinosarcoma; targeted therapy; HDAC inhibition; epithelial-mesenchymal transition; mesenchymal-epithelial transition; ARID1A ID uterine carcinosarcoma; targeted therapy; HDAC inhibition; epithelial-mesenchymal transition; mesenchymal-epithelial transition; ARID1A AB Objective: Uterine carcinosarcoma (UCS) is a rare but highly aggressive malignancy with biphasic growth pattern. This morphology can be attributed to epithelial-mesenchymal transition (EMT) that often associates with tumor invasion and metastasis. Accordingly, we analyzed a novel patient-derived preclinical model to explore whether EMT is a potential target in UCS. Methods: A novel UCS cell line (PF338) was established from the malignant pleural effusion of a 59-year-old patient at time of disease progression. Immunohistochemistry was performed in primary and metastatic tumor lesions. Oncogenic mutations were identified by next-generation sequencing. Viability assays and cell cycle analyses were used to test in vitro sensitivity to different standard and novel treatments. E-cadherin, β-catenin and pSMAD2 expressions were measured by immunoblot. Results: Whereas immunohistochemistry of the metastatic tumor showed a predominantly sarcomatous vimentin positive tumor that has lost E-cadherin expression, PF338 cells demonstrated biphasic growth and carried mutations in KRAS, PIK3CA, PTEN and ARID1A. PF338 tumor cells were resistant to MEK- and TGF-β signaling-inhibition but sensitive to PIK3CA- and PARP-inhibition and first-line chemotherapeutics. Strikingly, histone deacetylase (HDAC) inhibition markedly reduced cell viability by inducing a dose-dependent G0/1 arrest and led to mesenchymal-epithelial transition as evidenced by morphological change and increased E-cadherin and β-catenin expression. Conclusions: Our data suggest that HDAC inhibition is effective in a novel UCS cell line by interfering with both viability and differentiation. These findings emphasize the dynamic manner of EMT/MET and epigenetics and the importance of molecular profiling to pave the way for novel therapies in UCS. C1 [Stockhammer, Paul] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Okumus, Ozlem] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Hegedus, Luca] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Rittler, Dominika] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Ploenes, Till] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Herold, Thomas] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Kalbourtzis, Stavros] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Bankfalvi, Agnes] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Sucker, Antje] University Duisburg-Essen, University Hospital Essen, Department of DermatologyEssen, Germany. [Kimmig, Rainer] University Duisburg-Essen, University Hospital Essen, Department of Gynecology and ObstetricsEssen, Germany. [Aigner, Clemens] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Hegedus, Balazs] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. RP Hegedus, B (reprint author), University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic Surgery, Essen, Germany. EM balazs.hegedues@rlk.uk-essen.de CR Silverberg SG, Major FJ, Blessing JA, Fetter B, Askin FB, Liao S-Y, et al. Carcinosarcoma, malignant mixed mesodermal tumor, of the uterus. Int J Gynecol Pathol, 1990). 9(1):1–19., DOI 10.1097/00004347-199001000-00001 Kernochan LE, Garcia RL. Carcinosarcomas, malignant mixed mullerian tumor, of the uterus: advances in elucidation of biologic and clinical characteristics. J Natl Compr Canc Netw, 2009). 7(5):550–7., DOI 10.6004/ jnccn.2009.0037 Murali R, Davidson B, Fadare O, Carlson JA, Crum CP, Gilks CB, et al. Highgrade endometrial carcinomas. Int J Gynecol Pathol, 2019). 38(Suppl. 1): S40–S63., DOI 10.1097/pgp.0000000000000491 Pang A, Carbini M, Moreira AL, Maki RG. Carcinosarcomas and related cancers: tumors caught in the act of epithelial-mesenchymal transition. Jco, 2018). 36(2):210–6., DOI 10.1200/jco.2017.74.9523 Callister M, Ramondetta LM, Jhingran A, Burke TW, Eifel PJ. Malignant mixed mullerian tumors of the uterus: analysis of patterns of failure, prognostic factors, and treatment outcome. Int J Radiat Oncology*Biology*Physics, 2004). 58(3): 786–96., DOI 10.1016/s0360-3016(03)01561-x Zhu J, Wen H, Bi R, Wu X. Clinicopathological characteristics, treatment and outcomes in uterine carcinosarcoma and grade 3 endometrial cancer patients: a comparative study. J Gynecol Oncol, 2016). 27(2):e18., DOI 10.3802/jgo.2016. 27.e18 Cherniack AD, Shen H, Walter V, Stewart C, Murray BA, Bowlby R, et al. Integrated molecular characterization of uterine carcinosarcoma. Cancer Cell, 2017). 31(3):411–23., DOI 10.1016/j.ccell.2017.02.010 Jones S, Stransky N, McCord CL, Cerami E, Lagowski J, Kelly D, et al. Genomic analyses of gynaecologic carcinosarcomas reveal frequent mutations in chromatin remodelling genes. Nat Commun, 2014). 5:5006., DOI 10.1038/ ncomms6006 McConechy MK, Hoang LN, Chui MH, Senz J, Yang W, Rozenberg N, et al. Indepth molecular profiling of the biphasic components of uterine carcinosarcomas. J Path: Clin Res, 2015). 1(3):173–85., DOI 10.1002/cjp2.18 Xu J, Lamouille S, Derynck R. TGF-β-induced epithelial to mesenchymal transition. Cell Res, 2009). 19(2):156–72., DOI 10.1038/cr.2009.5 Chiyoda T, Tsuda H, Tanaka H, Kataoka F, Nomura H, Nishimura S, et al. Expression profiles of carcinosarcoma of the uterine corpus-are these similar to carcinoma or sarcoma?. Genes Chromosom Cancer, 2012). 51(3):229–39., DOI 10.1002/gcc.20947 Wawruszak A, Kalafut J, Okon E, Czapinski J, Halasa M, Przybyszewska A, et al. Histone deacetylase inhibitors and phenotypical transformation of cancer cells. Cancers, Basel,, 2019). 11(2)., DOI 10.3390/ cancers11020148 Dwivedi SK, McMeekin SD, Slaughter K, Bhattacharya R. Role of TGF-β signaling in uterine carcinosarcoma. Oncotarget, 2015). 6(16):14646–55., DOI 10.18632/oncotarget.3711 Osakabe M, Fukagawa D, Sato C, Sugimoto R, Uesugi N, Ishida K, et al. Immunohistochemical analysis of the epithelial to mesenchymal transition in uterine carcinosarcoma. Int J Gynecol Cancer, 2019). 29(2):277–281., DOI 10. 1136/ijgc-2018-000038 Nishimura I, Ohishi Y, Oda Y, Kishimoto J, Yasunaga M, Okuma E, et al. Expression and localization of E-cadherin and β-catenin in uterine carcinosarcoma. Virchows Arch, 2011). 458(1):85–94., DOI 10.1007/s00428- 010-1002-9 Wilson MR, Reske JJ, Holladay J, Wilber GE, Rhodes M, Koeman J, et al. ARID1A and PI3-kinase pathway mutations in the endometrium drive epithelial transdifferentiation and collective invasion. Nat Commun, 2019). 10(1):3554., DOI 10.1038/s41467-019-11403-6 Cuevas IC, Sahoo SS, Kumar A, Zhang H, Westcott J, Aguilar M, et al. Fbxw7 is a driver of uterine carcinosarcoma by promoting epithelial-mesenchymal transition. Proc Natl Acad Sci USA, 2019). 116(51):25880–90., DOI 10.1073/ pnas.1911310116 Fukumoto T, Park PH,Wu S, Fatkhutdinov N, Karakashev S, Nacarelli T, et al. Repurposing pan-HDAC inhibitors for arid1a-mutated ovarian cancer. Cell Rep, 2018). 22(13):3393–400., DOI 10.1016/j.celrep.2018.03.019 Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau KW, et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature, 2012). 483(7391):570–5., DOI 10.1038/nature11005 Li Y, Seto E. HDACs and HDAC inhibitors in cancer development and therapy. Cold Spring Harb Perspect Med, 2016). 6(10):a026831., DOI 10. 1101/cshperspect.a026831 Hrzenjak A, Moinfar F, Kremser M-L, Strohmeier B, Staber PB, Zatloukal K, et al. Valproate inhibition of histone deacetylase 2 affects differentiation and decreases proliferation of endometrial stromal sarcoma cells. Mol Cancer Ther, 2006). 5(9):2203–10., DOI 10.1158/1535-7163.mct-05-0480 Hrzenjak A, Moinfar F, Kremser M-L, Strohmeier B, Petru E, Zatloukal K, et al. Histone deacetylase inhibitor vorinostat suppresses the growth of uterine sarcomas in vitro and in vivo. Mol Cancer, 2010). 9:49., DOI 10.1186/1476- 4598-9-49 Schulten H-J, Wolf-Salgo J, Grundker C, Gunawan B, Fuzesi L. Characterization of a newly established uterine carcinosarcoma cell line featuring the sarcomatous phenotype of the tumor in vitro. Int J Gynecol Cancer, 2008). 18(2):339–44., DOI 10.1111/j.1525-1438.2007.01004.x Laszlo V, Hoda MA, Garay T, Pirker C, GhanimB, Klikovits T, et al. Epigenetic down-regulation of integrin α7 increases migratory potential and confers poor prognosis in malignant pleural mesothelioma. J Pathol, 2015). 237(2):203–14., DOI 10.1002/path.4567 Mikami Y, Hata S, Kiyokawa T, Manabe T. Expression of CD10 in malignant mullerian mixed tumors and adenosarcomas: an immunohistochemical study. Mod Pathol, 2002). 15(9):923–30., DOI 10.1097/01.mp.0000026058.33869.db Bernabe R, Patrao A, Carter L, Blackhall F, Dean E. Selumetinib in the treatment of non-small-cell lung cancer. Future Oncol, 2016). 12(22): 2545–60., DOI 10.2217/fon-2016-0132 Hanson K, Robinson SD, Al-Yousuf K, Hendry AE, Sexton DW, Sherwood V, et al. The anti-rheumatic drug, leflunomide, synergizes with MEK inhibition to suppress melanoma growth. Oncotarget, 2018). 9(3):3815–29., DOI 10.18632/ oncotarget.23378 Yamada SD, Burger RA, Brewster WR, Anton D, Kohler MF, Monk BJ. Pathologic variables and adjuvant therapy as predictors of recurrence and survival for patients with surgically evaluated carcinosarcoma of the uterus. Cancer, 2000). 88(12):2782–6., DOI 10.1002/1097-0142(20000615)88:12<2782: aid-cncr17>3.0.co;2-k Wolfson A, Brady M, Rocereto T, Mannel R, Lee Y, Futoran R, et al. A gynecologic oncology group randomized phase III trial of whole abdominal irradiation, WAI, vs. cisplatin-ifosfamide and mesna, CIM, as post-surgical therapy in stage I-IV carcinosarcoma, CS, of the uterus. Gynecol Oncol, 2007). 107(2):177–85., DOI 10.1016/j.ygyno.2007.07.070 Pectasides D, Pectasides E, Papaxoinis G, Xiros N, Sykiotis C, Papachristodoulou A, et al. Combination chemotherapy with carboplatin, paclitaxel and pegylated liposomal doxorubicin for advanced or recurrent carcinosarcoma of the uterus: clinical experience of a single institution. Gynecol Oncol, 2008). 110(3):299–303., DOI 10.1016/j. ygyno.2008.05.017 Abdulfatah E, Lordello L, Khurram M, Van de Vijver K, Alosh B, Bandyopadhyay S, et al. Predictive histologic factors in carcinosarcomas of the uterus. Int J Gynecol Pathol, 2019). 38(3):205–15., DOI 10.1097/pgp. 0000000000000497 Yuan Y, Kim W-H, Han HS, Lee J-H, Park H-S, Chung J-K, et al. Establishment and characterization of cell lines derived from uterine malignant mixed mullerian tumor. Gynecol Oncol, 1997). 66(3):464–74., DOI 10.1006/gyno.1997.4802 Gorai I, Doi C, Minaguchi H. Establishment and characterization of carcinosarcoma cell line of the human uterus. Cancer, 1993). 71(3):775–86., DOI 10.1002/1097-0142(19930201)71:3<775:aid-cncr2820710321>3.0.co;2-c Consortium APG. AACR project GENIE: powering precision medicine through an international consortium. Cancer Discov, 2017). 7(8):818–31., DOI 10.1158/2159-8290.CD-17-0151 Chiba Y, Sato S, Itamochi H, Suga Y, Fukagawa T,Oumi N, et al. Establishment and characterization of a novel uterine carcinosarcoma cell line, TU-ECS-1, with mutations of TP53 and KRAS. Hum Cell, 2017). 30(2):140–8., DOI 10. 1007/s13577-016-0154-6 Miraflor AP, de Abreu FB, Peterson JD, Turner SA, Amos CI, Tsongalis GJ, et al. Somatic mutation analysis in melanoma using targeted next generation sequencing. Exp Mol Pathol, 2017). 103(2):172–7., DOI 10.1016/j.yexmp.2017. 08.006 Somarelli JA, Boss MK, Epstein JI, Armstrong AJ, Garcia-Blanco MA. Carcinosarcomas: tumors in transition?. Histol Histopathol, 2015). 30(6): 673–87., DOI 10.14670/HH-30.673 Dwivedi SKD, Rao G, Dey A, Buechel M, Zhang Y, Zhang M, et al. Targeting the TGFβ pathway in uterine carcinosarcoma. Cell Stress, 2020). 4(11):252–60., DOI 10.15698/cst2020.11.234 Ossovskaya V, Koo IC, Kaldjian EP, Alvares C, Sherman BM. Upregulation of poly, ADP-Ribose, polymerase-1, PARP1, in triple-negative breast cancer and other primary human tumor types. Genes Cancer, 2010). 1(8):812–21., DOI 10. 1177/1947601910383418 Bitler BG, Wu S, Park PH, Hai Y, Aird KM, Wang Y, et al. ARID1A-mutated ovarian cancers depend on HDAC6 activity. Nat Cell Biol, 2017). 19(8): 962–73., DOI 10.1038/ncb3582 Wells A, Yates C, Shepard CR. E-cadherin as an indicator of mesenchymal to epithelial reverting transitions during the metastatic seeding of disseminated carcinomas. Clin Exp Metastasis, 2008). 25(6):621–8., DOI 10.1007/s10585-008- 9167-1 Yao D, Dai C, Peng S. Mechanism of the mesenchymal-epithelial transition and its relationship with metastatic tumor formation. Mol Cancer Res, 2011). 9(12):1608–20., DOI 10.1158/1541-7786.mcr-10-0568 Hernandez-Vargas H, Palacios J, Moreno-Bueno G. Telling cells how to die: docetaxel therapy in cancer cell lines. Cell Cycle, 2007). 6(7):780–3., DOI 10. 4161/cc.6.7.4050 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2021 VL 27 IS 1 BP 636088 EP 636097 DI 10.3389/pore.2021.636088 PG 10 ER PT J AU Ma, J Du, R Huang, Y Zhong, W Gui, H Mao, Ch Song, X Lu, J AF Ma, Jin Du, Rao Huang, Yan Zhong, Wen Gui, Huan Mao, Chenmei Song, Xiudao Lu, Jun TI Expression, Prognosis and Gene Regulation Network of NFAT Transcription Factors in Non-Small Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE non-small cell lung cancer; NFATs; prognosis; bioinformatics analysis; gene expression ID non-small cell lung cancer; NFATs; prognosis; bioinformatics analysis; gene expression AB Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide. The nuclear factor of activated T cells (NFAT) family is implicated in tumorigenesis and progression in various types of cancer. However, little is known about their expression patterns, distinct prognostic values, and potential regulatory networks in NSCLC. In this study, we comprehensively analyzed the distinct expression and prognostic value of NFATs in NSCLC through various large databases, including the Oncomine, UCSC Xena Browser, UALCAN databases, Kaplan–Meier Plotter, cBioPortal, and Enrichr. In lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), NFAT1/2/4/5 mRNA expression levels were significantly decreased and NFAT3 mRNA expression level was significantly increased. The cBioPortal database analysis showed that the mRNA dysregulation was one of the single most important factors for NFAT alteration in LUAD and LUSC and that both LUAD and LUSC cases with the alterations in the mRNA expression of NFATs had significantly better overall survival (OS). High expression levels of NFAT1/2/4/5 were significantly associated with better OS in LUAD, whereas high NFAT3 expression led to a worse OS. Overexpression of NFAT1/2 predicted better OS in LUSC, whereas high NFAT5 expression led to a worse OS. The networks for NFATs and the 50 most frequently altered neighbor genes in LUAD and LUSC were also constructed. NFATs and genes significantly associated with NFAT mRNA expression in LUAD and LUSC were significantly enriched in the cGMP-dependent protein kinase and Wnt signaling pathways. These results showed that the NFAT family members displayed varying degrees of abnormal expressions, suggesting that NFATs may be therapeutic targets for patients with NSCLC. Aberrant expression of NFATs was found to be associated with OS in the patients with NSCLC; among NFATs, NFAT3/4 may be new biomarkers for the prognosis of LUAD. However, further studies are required to validate our findings. C1 [Ma, Jin] Children’s Hospital of Soochow University, Department of PharmacySuzhou, China. [Du, Rao] Children’s Hospital of Soochow University, Department of PharmacySuzhou, China. [Huang, Yan] Children’s Hospital of Soochow University, Department of PharmacySuzhou, China. [Zhong, Wen] Children’s Hospital of Soochow University, Department of PharmacySuzhou, China. [Gui, Huan] Children’s Hospital of Soochow University, Department of PharmacySuzhou, China. [Mao, Chenmei] Children’s Hospital of Soochow University, Department of PharmacySuzhou, China. [Song, Xiudao] Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Clinical Pharmaceutical Laboratory of Traditional Chinese MedicineSuzhou, China. [Lu, Jun] Children’s Hospital of Soochow University, Department of HaematologySuzhou, China. RP Song, X (reprint author), Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Clinical Pharmaceutical Laboratory of Traditional Chinese Medicine, Suzhou, China. EM songxiudao0109@163.com CR Shaw J, Utz P, Durand D, Toole J, Emmel E, Crabtree G Identification of a putative regulator of early T cell activation genes. Science, 1988, 241(4862): 202–5., DOI 10.1126/science.3260404 Macian F NFAT proteins: key regulators of T-cell development and function. Nat Rev Immunol, 2005, 5(6):472–84., DOI 10.1038/nri1632 Mancini M, Toker A NFAT proteins: emerging roles in cancer progression. Nat Rev Cancer, 2009, 9(11):810–20., DOI 10.1038/nrc2735 Lee J-U, Kim L-K, Choi J-M Revisiting the concept of targeting NFAT to control T cell immunity and autoimmune diseases. Front Immunol, 2018, 9:2747., DOI 10.3389/fimmu.2018.02747 Mognol GP, Carneiro FRG, Robbs BK, Faget DV, Viola JPB Cell cycle and apoptosis regulation by NFAT transcription factors: new roles for an old player. Cell Death Dis, 2016, 7(4):e2199., DOI 10.1038/cddis.2016.97 Jauliac S, Lopez-Rodriguez C, Shaw LM, Brown LF, Rao A, Toker A The role of NFAT transcription factors in integrin-mediated carcinoma invasion. Nat Cel Biol, 2002, 4(7):540–4., DOI 10.1038/ncb816 Xu S, Shu P, Zou S, Shen X, Qu Y, Zhang Y, et al. NFAT c1 is a tumor suppressor in hepatocellular carcinoma and induces tumor cell apoptosis by activating the FasL-mediated extrinsic signaling pathway. Cancer Med, 2018, 7(9):4701–17., DOI 10.1002/cam4.1716 Qin X, Wang Y, Li J, Xiao Y, Liu Z NFAT5 inhibits invasion and promotes apoptosis in hepatocellular carcinoma associated with osmolality. neo, 2017, 64(04):502–10., DOI 10.4149/neo_2017_403 Quang CT, Leboucher S, Passaro D, Fuhrmann L,Nourieh M, Vincent-Salomon A, et al. The calcineurin/NFAT pathway is activated in diagnostic breast cancer cases and is essential to survival and metastasis of mammary cancer cells. Cel Death Dis, 2015, 6(2):e1658., DOI 10.1038/cddis.2015.14 Remo A, Simeone I, Pancione M, Parcesepe P, Finetti P, Cerulo L, et al. Systems biology analysis reveals NFAT5 as a novel biomarker and master regulator of inflammatory breast cancer. J Transl Med, 2015, 13(1):138., DOI 10. 1186/s12967-015-0492-2 Duque J, Fresno M, Iniguez MA Expression and function of the nuclear factor of activated T cells in colon carcinoma cells. J Biol Chem, 2005, 280(10): 8686–93., DOI 10.1074/jbc.m413076200 Ding W, Tong Y, Zhang X, Pan M, Chen S Study of arsenic sulfide in solid tumor cells reveals regulation of nuclear factors of activated T-cells by PML and p53. Sci Rep, 2016, 6(1):19793., DOI 10.1038/srep19793 Daniel C, Gerlach K, Vath M, Neurath MF, Weigmann B Nuclear factor of activated T cells-A transcription factor family as critical regulator in lung and colon cancer. Int J Cancer, 2014, 134(8):1767–75., DOI 10.1002/ijc.28329 Guo K, Jin F NFAT5 promotes proliferation and migration of lung adenocarcinoma cells in part through regulating AQP5 expression. Biochem Biophysical Res Commun, 2015, 465(3):644–9., DOI 10.1016/j.bbrc.2015.08.078 Heim L, Friedrich J, Engelhardt M, Trufa DI, Geppert CI, Rieker RJ, et al. NFATc1 promotes anti-tumoral effector functions and memory CD8+ T cell differentiation during non-small cell lung cancer development. Cancer Res, 2018, 78:canres.3297.2017–3633., DOI 10.1158/0008-5472.CAN-17-3297 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J Clinicians, 2018, 68(6): 394–424., DOI 10.3322/caac.21492 Yang P, Allen MS, Aubry MC, Wampfler JA,Marks RS, Edell ES, et al. Clinical features of 5,628 primary lung cancer patients. Chest, 2005, 128:452–62., DOI 10.1378/chest.128.1.452 Pikor LA, Ramnarine VR, Lam S, LamWL Genetic alterations defining NSCLC subtypes and their therapeutic implications. Lung Cancer, 2013, 82(2):179–89., DOI 10.1016/j.lungcan.2013.07.025 Spira A, Ettinger DS Multidisciplinary management of lung cancer. N Engl J Med, 2004, 350(4):379–92., DOI 10.1056/NEJMra035536 Shou J, Jing J, Xie J, You L, Jing Z, Yao J, et al. Nuclear factor of activated T cells in cancer development and treatment. Cancer Lett, 2015, 361(2):174–84., DOI 10.1016/j.canlet.2015.03.005 Lu H, Huang C Transcription factor NFAT, its role in cancer development, and as a potential target for chemoprevention. Ccdt, 2007, 7(4):343–53., DOI 10. 1016/j.canlet.2015.03.00510.2174/156800907780809750 Gopinath S, Vanamala SK, Gujrati M, Klopfenstein JD, Dinh DH, Rao JS Doxorubicin-mediated apoptosis in glioma cells requires NFAT3. Cell. Mol. Life Sci., 2009, 66(24):3967–78., DOI 10.1007/s00018-009- 0157-5 Lee H, Chouinard L, Bonin M, Michel RN NFATc3 deficiency may contribute to the development of mammary gland adenocarcinoma in aging female mice. Mol Carcinog, 2005, 44(3):219–22., DOI 10.1002/mc. 20136 Kuper C, Beck FX, Neuhofer W NFAT5-mediated expression of S100A4 contributes to proliferation and migration of renal carcinoma cells. Front Physiol, 2014, 5:293., DOI 10.3389/fphys.2014.00293 Chen Z-L, Zhao S-H, Wang Z, Qiu B, Li B-Z, Zhou F, et al. Expression and unique functions of four nuclear factor of activated T cells isoforms in nonsmall cell lung cancer. Chin J Cancer, 2011, 30(1):62–8., DOI 10.5732/cjc.010. 10156 Maxeiner JH, Karwot R, Sauer K, Scholtes P, Boross I, Koslowski M, et al. A key regulatory role of the transcription factor NFATc2 in bronchial adenocarcinoma via CD8+ T lymphocytes. Cancer Res, 2009, 69(7): 3069–76., DOI 10.1158/0008-5472.can-08-1678 Hou J, Aerts J, den Hamer B, van Ijcken W, den Bakker M, Riegman P, et al. Gene expression-based classification of non-small cell lung carcinomas and survival prediction. PLoS One, 2010, 5(4):e10312., DOI 10.1371/journal.pone. 0010312 Bhattacharjee A, Richards WG, Staunton J, Li C, Monti S, Vasa P, et al. Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. Proc Natl Acad Sci, 2001, 98(24):13790–5., DOI 10.1073/pnas.191502998 Beer DG, Kardia SL, Huang CC, Giordano TJ, Levin AM, Misek DE, et al. Gene-expression profiles predict survival of patients with lung adenocarcinoma. Nat Med, 2012, 8:816–24., DOI 10.1038/cddis.2016.9710. 1038/nm733 Garber ME, Troyanskaya OG, Schluens K, Petersen S, Thaesler Z, Pacyna- Gengelbach M, et al. Diversity of gene expression in adenocarcinoma of the lung. Proc Natl Acad Sci, 2001, 98:13784–9., DOI 10.1038/nm73310.1073/pnas. 241500798 Zhang X, Zhang Z, Cheng J, Li M, Wang W, Xu W, et al. Transcription factor NFAT1 activates the mdm2 oncogene independent of p53. J Biol Chem, 2012, 287(36):30468–76., DOI 10.1074/jbc.M112.373738 Chen X, Cao Q, Liao R,Wu X, Xun S, Huang J, et al. Loss of ABAT-mediated GABAergic system promotes basal-like breast cancer progression by activating Ca2+-NFAT1 Axis. Theranostics, 2019, 9(1):34–47., DOI 10. 7150/thno.29407 Kim G-C, Kwon H-K, Lee C-G, Verma R, Rudra D, Kim T, et al. Upregulation of Ets1 expression by NFATc2 and NFKB1/RELA promotes breast cancer cell invasiveness. Oncogenesis, 2018, 7(11):91., DOI 10.1038/ s41389-018-0101-3 Teixeira LK, Carrossini N, Secca C, Kroll JE, DaCunha DC, Faget DV, et al. NFAT1 transcription factor regulates cell cycle progression and cyclin E expression in B lymphocytes. Cell Cycle, 2016, 15(17):2346–59., DOI 10. 1080/15384101.2016.1203485 Marafiot T, Pozzobon M, Hansmann M-L, Ventura R, Pileri SA, Roberton H, et al. The NFATc1 transcription factor is widely expressed in white cells and translocates from the cytoplasm to the nucleus in a subset of human lymphomas. Br J Haematol, 2005, 128(3):333–42., DOI 10.1111/j.1365-2141. 2004.05313.x Buchholz M, Schatz A, Wagner M, Michl P, Linhart T, Adler G, et al. Overexpression of c-myc in pancreatic cancer caused by ectopic activation of NFATc1 and the Ca2+/calcineurin signaling pathway. EMBO J, 2006, 25(15):3714–24., DOI 10.1038/sj.emboj.7601246 Wang S, Kang X, Cao S, Cheng H, Wang D, Geng J Calcineurin/NFATc1 pathway contributes to cell proliferation in hepatocellular carcinoma. Dig Dis Sci, 2012, 57(12):3184–8., DOI 10.1007/s10620-012-2255-8 Akimzhanov A, Krenacs L, Schlegel T, Klein-Hessling S, Bagdi E, Stelkovics E, et al. Epigenetic changes and suppression of the nuclear factor of activated T cell 1, NFATC1, promoter in human lymphomas with defects in immunoreceptor signaling. Am J Pathol, 2008, 172(1):215–24., DOI 10.2353/ ajpath.2008.070294 Muller DJ,Wirths S, Fuchs AR, Marklin M, Heitmann JS, Sturm M, et al. Loss of NFAT2 expression results in the acceleration of clonal evolution in chronic lymphocytic leukemia. J Leukoc Biol, 2019, 105(3):531–8., DOI 10.1002/JLB. 2AB0218-076RR Ma SSQ, Srivastava S, Llamosas E, Hawkins NJ, Hesson LB, Ward RL, et al. ROR2 is epigenetically inactivated in the early stages of colorectal neoplasia and is associated with proliferation and migration. BMC Cancer, 2016, 16(1): 508., DOI 10.1186/s12885-016-2576-7 Wu X, Nguyen B-C, Dziunycz P, Chang S, Brooks Y, Lefort K, et al. Opposing roles for calcineurin and ATF3 in squamous skin cancer. Nature, 2010, 465(7296):368–72., DOI 10.1038/nature08996 Zhang Y, Liao JM, Zeng SX, Lu H p53 downregulates Down syndromeassociated DYRK1A through miR-1246. EMBO Rep, 2011, 12(8):811–7., DOI 10.1038/embor.2011.98 Im J-Y, Lee K-W, Won K-J, Kim B-K, Ban HS, Yoon S-H, et al. DNA damage-induced apoptosis suppressor, DDIAS), a novel target of NFATc1, is associated with cisplatin resistance in lung cancer. Biochim Biophys Acta, Bba, - Mol Cel Res, 2016, 1863(1):40–9., DOI 10. 1016/j.bbamcr.2015.10.011 Zhang H, Xie X, Zhu X, Zhu J, Hao C, Lu Q, et al. Stimulatory cross-talk between NFAT3 and estrogen receptor in breast cancer cells. J Biol Chem, 2005, 280(52):43188–97., DOI 10.1074/jbc.M506598200 Xiao T, Zhu JJ, Huang S, Peng C, He S, Du J, et al. Phosphorylation of NFAT3 by CDK3 induces cell transformation and promotes tumor growth in skin cancer. Oncogene, 2017, 36(20):2835–45., DOI 10.1038/onc. 2016.434 Hessmann E, Zhang J-S, Chen N-M, Hasselluhn M, Liou G-Y, Storz P, et al. NFATc4 RegulatesSox9Gene expression in acinar cell plasticity and pancreatic cancer initiation. Stem Cell Int, 2016, 2016:1., DOI 10.1155/ 2016/5272498 Zhang X, Kang T, Zhang L, Tong Y, Ding W, Chen S NFATc3 mediates the sensitivity of gastric cancer cells to arsenic sulfide. Oncotarget, 2017, 8(32): 52735–45., DOI 10.18632/oncotarget.17175 Pintarelli G, Dassano A, Cotroneo CE, Galvan A, Noci S, Piazza R, et al. Readthrough transcripts in normal human lung parenchyma are down-regulated in lung adenocarcinoma. Oncotarget, 2016, 7(19):27889–98., DOI 10.18632/ oncotarget.8556 Meng X, Li Z, Zhou S, Xiao S, Yu P miR-194 suppresses high glucose-induced non-small cell lung cancer cell progression by targeting NFAT5. Thorac Cancer, 2019, 10(5):1051–9., DOI 10.1111/1759-7714.13038 Rahman NIA, Abdul Murad NA, Mollah MM, Jamal R, Harun R NFIX as a master regulator for lung cancer progression. Front Pharmacol, 2017, 8:540., DOI 10.3389/fphar.2017.00540 Mijatovic T, Mathieu V, Gaussin J-F, De Neve N, Ribaucour F, Van Quaquebeke E, et al. Cardenolide-induced lysosomal membrane permeabilization demonstrates therapeutic benefits in experimental human non-small cell lung cancers. Neoplasia, 2006, 8(5):402–12., DOI 10.1593/neo. 05850 Cho HJ, Yun H-J, Yang HC, Kim SJ, Kang SK, Che C, et al. Prognostic significance of nuclear factor of activated T-cells 5 expression in non-small cell lung cancer patients who underwent surgical resection. J Surg Res, 2018, 226: 40–7., DOI 10.1016/j.jss.2017.12.036 Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, et al. ONCOMINE: a cancer microarray database and integrated data-mining platform. Neoplasia, 2004, 6(1):1–6., DOI 10.1016/s1476-5586(04)80047-2 Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce- Rodriguez I, Chakravarthi BVSK, et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia, 2017, 19(8): 649–58., DOI 10.1016/j.neo.2017.05.002 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data: figure 1. Cancer Discov, 2012, 2(5):401–4., DOI 10.1158/ 2159-8290.CD-12-0095 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signaling, 2013, 6(269):pl1., DOI 10.1126/scisignal. 2004088 Gy}orffy B, Surowiak P, Budczies J, Lanczky A Online survival analysis software to assess the prognostic value of biomarkers using transcriptomic data in nonsmall- cell lung cancer. PLoS One, 2013, 8(12):e82241., DOI 10.1371/journal. pone.0082241 Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res, 2016, 44(W1):W90–W97., DOI 10.1093/nar/ gkw377 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 529240 EP 529256 DI 10.3389/pore.2021.529240 PG 17 ER PT J AU Shen, W Shan, B Liang, Sh Zhang, J Yu, Y Zhang, Y Wang, G Bai, Y Qian, B Lu, J Jiang, Z AF Shen, Wenbin Shan, Boer Liang, Shanhui Zhang, Junling Yu, Yangyang Zhang, Yuzi Wang, Guoqiang Bai, Yuezong Qian, Bing Lu, Jin Jiang, Zhi TI Hybrid Capture-based Genomic Profiling of Circulating Tumor DNA From Patients With Advanced Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE circulating tumor DNA; liquid biopsy; ovarian cancer; genomic profiling; genomic alterations ID circulating tumor DNA; liquid biopsy; ovarian cancer; genomic profiling; genomic alterations AB Objective: We conducted this study to characterize somatic genomic alterations in circulating tumor DNA (ctDNA) from patients with ovarian cancer and compare GAs detected in ctDNA with tissue databases. Methods: Hybrid capture-next generation sequencing genomic profiling of 150 genes was performed on ctDNA from 138 patients with ovarian cancer with 1,500× sequencing depth. The GAs detected in ctDNA were compared with those in our ovarian cancer tissue database (N=488) and the Cancer Genome Atlas (TCGA) database (N=489). Results: 115 patients (83%) had at least 1 GA detected in ctDNA. The most frequently altered genes detected in ctDNA were TP53 (72%), KRAS (11%), LRP1B (10%), ZNF703 (9%) and NF1 (8%). Comparative analysis with our tissue database showed similar frequencies of GAs per gene, although PIK3CA and KRAS mutations were more frequent in tissue and ctDNA, respectively (p < 0.05). Gene amplification and rearrangement were more frequent in ctDNA samples. The mutation frequency of homologous recombination repair associated-genes, VEGF signal/angiogenesis pathways, RAS pathways, NOTCH pathways and MSI-H ratio was not statistically different either in ctDNA or in tissue database. However, the mutation frequency of AKT, PIK3CA, PTEN and STK11 in PI3K/AKT/mTOR pathway was significantly lower than that in tissue samples (p < 0.05). Conclusions: Our results suggest that genomic profiling of ctDNA could detect somatic GAs in a significant subset of patients with ovarian cancer. Hybrid capture-NGS based on liquid biopsy has the potential capability to serve as a substitute to tissue biopsy and further studies are warranted. C1 [Shen, Wenbin] Fudan University Shanghai Cancer Center, Department of OncologyShanghai, China. [Shan, Boer] Fudan University Shanghai Cancer Center, Department of OncologyShanghai, China. [Liang, Shanhui] Fudan University Shanghai Cancer Center, Department of OncologyShanghai, China. [Zhang, Junling] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Yu, Yangyang] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Zhang, Yuzi] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Wang, Guoqiang] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Bai, Yuezong] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Qian, Bing] Jiangsu Cancer Hospital, the Affiliated Hospital of Nanjing Medical University, Departments of Gynecomatous SurgeryNanjing, China. [Lu, Jin] Jiangsu Cancer Hospital, the Affiliated Hospital of Nanjing Medical University, Departments of Gynecomatous SurgeryNanjing, China. [Jiang, Zhi] Jiangsu Cancer Hospital, the Affiliated Hospital of Nanjing Medical University, Departments of Gynecomatous SurgeryNanjing, China. RP Jiang, Z (reprint author), Jiangsu Cancer Hospital, the Affiliated Hospital of Nanjing Medical University, Departments of Gynecomatous Surgery, Nanjing, China. EM margrate71@163.com CR Matulonis UA, Sood AK, Fallowfield L, Howitt BE, Sehouli J, Karlan BY Ovarian cancer. Nat Rev Dis Primers, 2016). 2:16061., DOI 10.1038/nrdp.2016.61 National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: ovarian cancer, version 1.2020). Available from: https://www.nccn. org/professionals/physician_gls/default.aspx. Kondrashova O, Nguyen M, Shield-Artin K, Tinker AV, Teng NNH, Harrell MI, et al. Secondary somaticmutations restoring RAD51C and RAD51D associated with acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma. Cancer Discov, 2017). 7(9):984–98., DOI 10.1158/2159-8290.cd-17-0419 Yeung TL, Leung CS, Yip KP, Yeung CLA, Wong STC, Mok SC Cellular and molecular processes in ovarian cancer metastasis. a review in the theme: cell and molecular processes in cancer metastasis. Am J Physiol Cell Physiol, 2015). 309(7):444–56., DOI 10.1152/ajpcell.00188.2015 Wang Z, Duan J, Cai S, Han M, Dong H, Zhao J, et al. Assessment of blood tumor mutational burden as a potential biomarker for immunotherapy in patients with non-small cell lung cancer with use of a next-generation sequencing cancer gene panel. JAMA Oncol, 2019). 5(5):696–702., DOI 10. 1001/jamaoncol.2018.7098 Yang N, Li Y, Liu Z, Qin H, Du D, Cao X, et al. The characteristics of ctDNA reveal the high complexity in matching the corresponding tumor tissues. BMC Cancer, 2018). 18(1):319., DOI 10.1186/s12885-018-4199-7 Li H, Durbin R Fast and accurate long-read alignment with burrows-wheeler transform. Bioinformatics, 2010). 26:589–95., DOI 10.1093/bioinformatics/ btp698 Cibulskis K, Lawrence MS, Carter SL, Sivachenko A, Jaffe D, Sougnez C, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol, 2013). 31:213–9., DOI 10. 1038/nbt.2514 Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res, 2012). 22:568–76., DOI 10.1101/gr.129684.111 Ye K, Schulz MH, Long Q, Apweiler R, Ning Z Pindel: a pattern growth approach to detect break points of large deletions and medium sized insertions from paired-end short reads. Bioinformatics, 2009). 25:2865–71., DOI 10.1093/ bioinformatics/btp394 Xi R, Lee S, Xia Y, Kim T-M, Park PJ Copy number analysis of wholegenome data using BIC-seq2 and its application to detection of cancer susceptibility variants. Nucleic Acids Res, 2016). 44:6274–86., DOI 10.1093/ nar/gkw491 Chabon JJ, Simmons AD, Lovejoy AF, Esfahani MS, Newman AM, Haringsma HJ, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun, 2016). 7:11815., DOI 10.1038/ncomms11815 Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol, 2011). 29:24–6., DOI 10.1038/ nbt.1754 The Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature, 2011). 474(7353):609–15., DOI 10.1038/ nature10166 Garziera M, Roncato R, Montico M, De Mattia E, Gagno S, Poletto E, et al. New challenges in tumor mutation heterogeneity in advanced ovarian cancer by a targeted next-generation sequencing, ngs, approach. Cells, 2019). 8(6):584., DOI 10.3390/cells8060584 Macintyre G, Goranova TE, Silva DD, Ennis D, Piskorz AM, Eldridge M, et al. Copy-number signatures and mutational processes in ovarian carcinoma. Nat Genet, 2017). 50(9):1262–70., DOI 10.1038/s41588-018-0179-8 The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature, 2017). 487(7407):330–7., DOI 10.1038/nature11252 Santillan A, Kim YW, Zahurak ML, Gardner GJ, Giuntoli RL, Shih IM, et al. Differences of chemoresistance assay between invasive micropapillary/lowgrade serous ovarian carcinoma and high-grade serous ovarian carcinoma. Int J Gynecol Cancer, 2007). 17(3):601–6., DOI 10.1111/j.1525-1438.2007.00820.x NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 581534 EP 581539 DI 10.3389/pore.2021.581534 PG 6 ER PT J AU Zheng, L Kang, L Cheng, Y Cao, J Liu, L Xu, H Gao, L AF Zheng, Lei Kang, Liying Cheng, Yan Cao, Junli Liu, Lijie Xu, Hongmei Gao, Liming TI Tumor Inhibitory Effect of Long Non-coding RNA LOC100505817 on Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE long non-coding RNA LOC100505817; wnt/β-catenin signaling pathway; gastric cancer; epithelialmesenchymal transition; proliferation; invasion; migration; apoptosis ID long non-coding RNA LOC100505817; wnt/β-catenin signaling pathway; gastric cancer; epithelialmesenchymal transition; proliferation; invasion; migration; apoptosis AB Gastric cancer (GC) is one of the major malignancies worldwide. Emerging evidence has revealed the potential involvement of long noncoding RNA (lncRNA) in human genetic disorders and cancer, but the role of LOC100505817 remains unknown. Thus, in this study, we isolated tissues from GC patients to characterize the functional importance of LOC100505817 in GC tumorigenesis. We also proposed a hypothesis that the regulation of Wnt/β-catenin pathway by LOC100505817 was regulated by miR-20a-mediated WT1. After the collection of cancer tissues and adjacent tissues were obtained fromGC patients, expression of LOC100505817, Wnt/β-catenin pathway- and EMT-related genes was quantified. Ectopic expression and knockdown experiments were applied in order to investigate the protective role of LOC100505817 in the progression of GC. Subsequently, cell viability, flow cytometry for apoptosis and cell cycle were detected via CCK-8, while migration and invasion were determined using scratch test and Transwell assay respectively. Then interactions among LOC100505817, miR-20a and WT1 were explored by dual luciferase reporter gene assay, RNA pull down assay and RNA binding protein immunoprecipitation (RIP) assay. The results found poor expression LOC100505817 was poorly expressed in GC cells and tissues. Overexpressed LOC100505817 resulted in the significant reduction of cell proliferation, migration and invasion as well as the expression of Wnt2b, β-catenin, CyclinD1, N-cadherin, Vimentin and snail, while increased cell apoptosis along with the expression of E-cadherin. Wnt/ β-catenin pathway and EMT in GC cells were suppressed by LOC100505817 through miR-20a-inhibted WT1. In summary, our results provided evidence suggesting that LOC100505817 inhibits GC through LOC100505817-mediated inhibition of Wnt/ β-catenin pathway, that leads to the overall restraining of GC cell proliferation, migration and invasion through miR-20a-reduced WT1. C1 [Zheng, Lei] The First Hospital of Qinhuangdao, Department of OncologyQinhuangdao, China. [Kang, Liying] Wuqing People Hospital, Department of OncologyTianjin, China. [Cheng, Yan] The First Hospital of Qinhuangdao, Disinfection Supply RoomQinhuangdao, China. [Cao, Junli] The First Hospital of Qinhuangdao, Department of OncologyQinhuangdao, China. [Liu, Lijie] The First Hospital of Qinhuangdao, Department of OncologyQinhuangdao, China. [Xu, Hongmei] The First Hospital of Qinhuangdao, Department of OncologyQinhuangdao, China. [Gao, Liming] The First Hospital of Qinhuangdao, Department of OncologyQinhuangdao, China. RP Gao, L (reprint author), The First Hospital of Qinhuangdao, Department of Oncology, Qinhuangdao, China. EM tshgaoliming@126.com CR Van Cutsem E, Sagaert X, Topal B, Haustermans K, Prenen H. Gastric cancer. The Lancet, 2016). 388(10060):2654–64., DOI 10.1016/s0140-6736(16)30354-3 Peng Y, Zhang X, Ma Q, Yan R, Qin Y, Zhao Y, et al. MiRNA-194 activates the Wnt/β-catenin signaling pathway in gastric cancer by targeting the negative Wnt regulator, SUFU. Cancer Lett, 2017). 385:117–27., DOI 10.1016/j.canlet. 2016.10.035 Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarkers Prev, 2014). 23(5):700–13., DOI 10.1158/1055-9965.epi- 13-1057 Sui W, Shi Z, Xue W, Ou M, Zhu Y, Chen J, et al. Circular RNA and gene expression profiles in gastric cancer based on microarray chip technology. Oncol Rep, 2017). 37(3):1804–14., DOI 10.3892/or.2017.5415 Ding J, Li D, Gong M, Wang J, Huang X, Wu T, et al. Expression and clinical significance of the long non-coding RNA PVT1 in human gastric cancer. Ott, 2014). 7:1625–30., DOI 10.2147/ott.s68854 Lin X-C, Zhu Y, Chen W-B, Lin L-W, Chen D-H, Huang J-R, et al. Integrated analysis of long non-coding RNAs and mRNA expression profiles reveals the potential role of lncRNAs in gastric cancer pathogenesis. Int J Oncol, 2014). 45(2):619–28., DOI 10.3892/ijo.2014.2431 Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet, 2014). 15(1):7–21., DOI 10.1038/nrg3606 Mitra SA, Mitra AP, Triche TJ. A central role for long non-coding RNA in cancer. Front Genet, 2012). 3:17., DOI 10.3389/fgene.2012.00017 Kogo R, Shimamura T, Mimori K, Kawahara K, Imoto S, Sudo T, et al. Long noncoding RNA HOTAIR regulates polycomb-dependent chromatin modification and is associated with poor prognosis in colorectal cancers. Cancer Res, 2011). 71(20):6320–6., DOI 10.1158/0008-5472.can-11-1021 Lai M-c., Yang Z, Zhou L, Zhu Q-q., Xie H-y., Zhang F, et al. Long non-coding RNA MALAT-1 overexpression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med Oncol, 2012). 29(3):1810–6., DOI 10. 1007/s12032-011-0004-z Matouk IJ, Mezan S, Mizrahi A, Ohana P, Abu-Lail R, Fellig Y, et al. The oncofetal H19 RNA connection: hypoxia, p53 and cancer. Biochim Biophys Acta, Bba, - Mol Cel Res, 2010). 1803(4):443–51., DOI 10.1016/j.bbamcr.2010. 01.010 Li C, Liang G, Yao W, Sui J, Shen X, Zhang Y, et al. Differential expression profiles of long non-coding RNAs reveal potential biomarkers for identification of human gastric cancer. Oncol Rep, 2016). 35(3):1529–40., DOI 10.3892/or.2015.4531 Zhao L, Guo H, Zhou B, Feng J, Li Y, Han T, et al. Long non-coding RNA SNHG5 suppresses gastric cancer progression by trapping MTA2 in the cytosol. Oncogene, 2016). 35(44):5770–80., DOI 10.1038/onc.2016.110 Tajima Y, Murakami T, Saito T, Hiromoto T, Akazawa Y, Sasahara N, et al. Distinct involvement of the sonic hedgehog signaling pathway in gastric adenocarcinoma of fundic gland type and conventional gastric adenocarcinoma. Digestion, 2017). 96(2):81–91., DOI 10.1159/000478999 Yuan Z, Yu X, Ni B, Chen D, Yang Z, Huang J, et al. Overexpression of long non-coding RNA-CTD903 inhibits colorectal cancer invasion and migration by repressing Wnt/β-catenin signaling and predicts favorable prognosis. Int J Oncol, 2016). 48(6):2675–85., DOI 10.3892/ijo.2016.3447 Zhao H, Hou W, Tao J, Zhao Y, Wan G, Ma C, et al. Upregulation of lncRNA HNF1A-AS1 promotes cell proliferation and metastasis in osteosarcoma through activation of the Wnt/β-catenin signaling pathway. Am J Transl Res, 2016). 8(8):3503–12. Peng Z, Wang CX, Fang EH, Wang GB, Tong Q. Role of epithelialmesenchymal transition in gastric cancer initiation and progression. Wjg, 2014). 20:5403–10., DOI 10.3748/wjg.v20.i18.5403 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res, 2015). 43(7):e47., DOI 10.1093/nar/gkv007 Wen S-L, Zhang F, Feng S. Decreased copy number of mitochondrial DNA: a potential diagnostic criterion for gastric cancer. Oncol Lett, 2013). 6(4): 1098–102., DOI 10.3892/ol.2013.1492 Yao X-M, Tang J-H, Zhu H, Jing Y. High expression of lncrna casc15 is a risk factor for gastric cancer prognosis and promote the proliferation of gastric cancer. Eur Rev Med Pharmacol ences, 2017). 21(24):5661. Scagliotti G, Kang JH, Smith D, Rosenberg R, Park K, Kim S-W, et al. Phase II evaluation of LY2603618, a first-generation CHK1 inhibitor, in combination with pemetrexed in patients with advanced or metastatic non-small cell lung cancer. Invest New Drugs, 2016). 34(5):625–35., DOI 10.1007/s10637-016- 0368-1 Goss AM, Tian Y, Tsukiyama T, Cohen ED, Zhou D, Lu MM, et al. Wnt2/2b and β-catenin signaling are necessary and sufficient to specify lung progenitors in the foregut. Develop Cel, 2009). 17(2):290–8., DOI 10.1016/j.devcel.2009. 06.005 Dozier C, Mazzolini L, Cenac C, Froment C, Burlet-Schiltz O, Besson A, et al. CyclinD-CDK4/6 complexes phosphorylate CDC25A and regulate its stability. Oncogene, 2017). 36(26):3781–8., DOI 10.1038/onc.2016.506 Geissler AL, Geissler M, Kottmann D, Lutz L, Fichter CD, Fritsch R, et al. ATM mutations and E-cadherin expression define sensitivity to EGFR-targeted therapy in colorectal cancer. Oncotarget, 2017). 8(10):17164–90., DOI 10. 18632/oncotarget.15211 Xu J, Lu MX, Cui YD, Du YZ. Selection and evaluation of reference genes for expression analysis using qRT-PCR in chilo suppressalis, Lepidoptera: Pyralidae). J Econ Entomol, 2017). 110(2):683–91., DOI 10.1093/jee/tow297 Emons G, Spitzner M, Reineke S, Moller J, Auslander N, Kramer F, et al. Chemoradiotherapy resistance in colorectal cancer cells is mediated by wnt/ β-catenin signaling. Mol Cancer Res, 2017). 15(11):1481–90., DOI 10.1158/ 1541-7786.mcr-17-0205 Pasechnikov V, Chukov S, Fedorov E, Kikuste I, Leja M. Gastric cancer: prevention, screening and early diagnosis. Wjg, 2014). 20(38):13842–62., DOI 10.3748/wjg.v20.i38.13842 Harada K, Mizrak Kaya D, Shimodaira Y, Ajani JA. Global chemotherapy development for gastric cancer. Gastric Cancer, 2017). 20(Suppl. 1):92–101., DOI 10.1007/s10120-016-0655-8 Song H, Sun W, Ye G, Ding X, Liu Z, Zhang S, et al. Long non-coding RNA expression profile in human gastric cancer and its clinical significances. J Translational Med, 2013). 11:225., DOI 10.1186/1479-5876-11-225 Gibb EA, Brown CJ, Lam WL. The functional role of long non-coding RNA in human carcinomas. Mol Cancer, 2011). 10:38., DOI 10.1186/1476-4598-10-38 Zeng S, Xie X, Xiao Y-F, Tang B, Hu C-J, Wang S-M, et al. Long noncoding RNA LINC00675 enhances phosphorylation of vimentin on Ser83 to suppress gastric cancer progression. Cancer Lett, 2018). 412:179–87., DOI 10.1016/j. canlet.2017.10.026 Saito T, Kurashige J, Nambara S, Komatsu H, Hirata H, Ueda M, et al. A long non-coding RNA activated by transforming growth factor-beta is an independent prognostic marker of gastric cancer. Ann Surg Oncol, 2015). 22(Suppl. 3):S915–922., DOI 10.1245/s10434-015-4554-8 Li Y, Wang K, Wei Y, Yao Q, Zhang Q, Qu H, et al. lncRNA-MIAT regulates cell biological behaviors in gastric cancer through a mechanism involving the miR-29a-3p/HDAC4 axis. Oncol Rep, 2017). 38(6):3465–72., DOI 10.3892/or. 2017.6020 Deng W, Wang J, Zhang J, Cai J, Bai Z, Zhang Z. TET2 regulates LncRNAANRIL expression and inhibits the growth of human gastric cancer cells. IUBMB Life, 2016). 68(5):355–64., DOI 10.1002/iub.1490 Wang Y, Liu X, Zhang H, Sun L, Zhou Y, Jin H, et al. Hypoxia-inducible lncRNA-ak058003 promotes gastric cancer metastasis by targeting γ-synuclein. Neoplasia, 2014). 16(12):1094–106., DOI 10.1016/j.neo.2014. 10.008 Ma P, Xu T, Huang M, Shu Y. Increased expression of LncRNA PANDAR predicts a poor prognosis in gastric cancer. Biomed Pharmacother, 2016). 78: 172–6., DOI 10.1016/j.biopha.2016.01.025 Sun M, Xia R, Jin F, Xu T, Liu Z, De W, et al. Downregulated long noncoding RNA MEG3 is associated with poor prognosis and promotes cell proliferation in gastric cancer. Tumor Biol, 2014). 35(2):1065–73., DOI 10.1007/s13277-013- 1142-z Zhao J, Liu Y, Zhang W, Zhou Z, Wu J, Cui P, et al. Long non-coding RNA Linc00152 is involved in cell cycle arrest, apoptosis, epithelial to mesenchymal transition, cell migration and invasion in gastric cancer. Cell Cycle, 2015). 14(19):3112–23., DOI 10.1080/15384101.2015.1078034 Xu Z-Y, Yu Q-M, Du Y-A, Yang L-T, Dong R-Z, Huang L, et al. Knockdown of long non-coding RNA HOTAIR suppresses tumor invasion and reverses epithelial-mesenchymal transition in gastric cancer. Int J Biol Sci, 2013). 9(6):587–97., DOI 10.7150/ijbs.6339 Chiurillo MA. Role of the Wnt/β-catenin pathway in gastric cancer: an indepth literature review. Wjem, 2015). 5(2):84–102., DOI 10.5493/wjem.v5.i2.84 Hoffmeyer K, Raggioli A, Rudloff S, Anton R, Hierholzer A, Del Valle I, et al. Wnt/-catenin signaling regulates telomerase in stem cells and cancer cells. Science, 2012). 336(6088):1549–54., DOI 10.1126/science.1218370 Yao H, Ashihara E, Maekawa T. Targeting the Wnt/β-catenin signaling pathway in human cancers. Expert Opin Ther Targets, 2011). 15(7):873–87., DOI 10.1517/14728222.2011.577418 Wang X, Lu X, Geng Z, Yang G, Shi Y. LncRNA PTCSC3/miR-574-5p governs cell proliferation andmigration of papillary thyroid carcinoma via wnt/β-catenin signaling. J Cel Biochem., 2017). 118(12):4745–52., DOI 10.1002/jcb.26142 He S, Zhao Y,Wang X, Deng Y, Wan Z, Yao S, et al. Up-regulation of long noncoding RNA SNHG20 promotes ovarian cancer progression via Wnt/betacatenin signaling. Biosci Rep, 2018). 38(1)., DOI 10.1042/bsr20170681 Liang S, Zhang S, Wang P, Yang C, Shang C, Yang J, et al. LncRNA, TUG1 regulates the oral squamous cell carcinoma progression possibly via interacting with Wnt/β-catenin signaling. Gene, 2017). 608:49–57., DOI 10.1016/j.gene. 2017.01.024 Li K, Dan Z. [Research progress of Wnt/β-catenin signaling pathway in prevention and treatment of gastric cancer]. Nan Fang Yi Ke Da Xue Xue Bao, 2014). 34(12):1852–6. Wu Y, Ginther C, Kim J, Mosher N, Chung S, Slamon D, et al. Expression of Wnt3 activates wnt/β-catenin pathway and promotes EMT-like phenotype in trastuzumab-resistant HER2-overexpressing breast cancer cells. Mol Cancer Res, 2012). 10(12):1597–606., DOI 10.1158/1541-7786.mcr-12-0155-t Shi S, Li Q, Cao Q, Diao Y, Zhang Y, Yue L, et al. EMT transcription factors are involved in the altered cell adhesion under simulated microgravity effect or overloading by regulation of E-cadherin. Int J Mol Sci, 2020). 21(4):1349., DOI 10.3390/ijms21041349 Dhamija S, Diederichs S. From junk to master regulators of invasion: lncRNA functions in migration, EMT and metastasis. Int J Cancer, 2016). 139(2): 269–80., DOI 10.1002/ijc.30039 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 581542 EP 581553 DI 10.3389/pore.2021.581542 PG 12 ER PT J AU Zhang, D Dai, J Pan, Y Wang, X Qiao, J Sasano, H Zhao, B McNamara, K Guan, X Liu, L Zhang, Y Chan, M Cao, Sh Liu, M Song, S Wang, L AF Zhang, Dongmei Dai, Jiali Pan, Yu Wang, Xiuli Qiao, Juanjuan Sasano, Hironobu Zhao, Baoshan McNamara, M. Keely Guan, Xue Liu, Lili Zhang, Yanzhi Chan, S. M. Monica Cao, Shuwen Liu, Ming Song, Sihang Wang, Lin TI Overexpression of PELP1 in Lung Adenocarcinoma Promoted E2 Induced Proliferation, Migration and Invasion of the Tumor Cells and Predicted a Worse Outcome of the Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE estrogen; proline-; glutamic acid-; leucine-rich protein 1; lung adenocarcinoma; tumor progression ID estrogen; proline-; glutamic acid-; leucine-rich protein 1; lung adenocarcinoma; tumor progression AB The expression of Proline-, glutamic acid-, and leucine-rich protein 1 (PELP1) has been reported to be dysregulated in non-small cell lung carcinoma, especially in lung adenocarcinoma (LUAD). Therefore, we aimed to investigate the functional and prognostic roles of PELP1 in LUAD in this study. We first immunolocalized PELP1 in 76 cases of LUAD and 17 non-pathological or tumorous lung (NTL) tissue specimens and correlated the findings with the clinicopathological parameters of the patients. We then performed in vitro analysis including MTT, flow cytometry, wound healing, and transwell assays in order to further explore the biological roles of PELP1 in 17-β-estradiol (E2) induced cell proliferation, migration, and invasion of LUAD cells. We subsequently evaluated the prognostic significance of PELP1 in LUAD patients using the online survival analysis tool Kaplan-Meier Plotter. The status of PELP1 immunoreactivity in LUAD was significantly higher than that in the NTL tissues and significantly positively correlated with less differentiated features of carcinoma cells, positive lymph node metastasis, higher clinical stage as well as the status of ERα, ERβ, and PCNA. In vitro study did reveal that E2 promoted cell proliferation and migration and elevated PELP1 protein level in PELP1-high A549 and H1975 cells but not in PELP1-low H-1299 cells. Knock down of PELP1 significantly attenuated E2 induced cell proliferation, colony formation, cell cycle progress as well as migration and invasion of A549 and H1975 cells. Kaplan-Meier Plotter revealed that LUAD cases harboring higher PELP1 expression had significantly shorter overall survival. In summary, PELP1 played a pivotal role in the estrogen-induced aggressive transformation of LUAD and could represent adverse clinical outcome of the LUAD patients. C1 [Zhang, Dongmei] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [Dai, Jiali] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [Pan, Yu] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [Wang, Xiuli] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [Qiao, Juanjuan] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [Sasano, Hironobu] Tohoku Medical and Pharmaceutical University, Faculty of Medicine, Division of PathologySendai, Japan. [Zhao, Baoshan] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [McNamara, M. Keely] Tohoku Medical and Pharmaceutical University, Faculty of Medicine, Division of PathologySendai, Japan. [Guan, Xue] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [Liu, Lili] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [Zhang, Yanzhi] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [Chan, S. M. Monica] Tohoku Medical and Pharmaceutical University, Faculty of Medicine, Division of PathologySendai, Japan. [Cao, Shuwen] Daqing Oilfield General Hospital, Department of PathologyDaqing, China. [Liu, Ming] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [Song, Sihang] Harbin Medical University-Daqing, Department of PathologyDaqing, China. [Wang, Lin] Harbin Medical University-Daqing, Department of PathologyDaqing, China. RP Wang, L (reprint author), Harbin Medical University-Daqing, Department of Pathology, Daqing, China. CR Henley SJ, Ward EM, Scott S, Ma J, Anderson RN, Firth AU, et al. Annual report to the nation on the status of cancer, part I: National cancer statistics. Cancer, 2020, 126(10):2225–49., DOI 10.1002/cncr.32802 Hammoud Z, Tan B, Badve S, Bigsby RM Estrogen promotes tumor progression in a genetically defined mouse model of lung adenocarcinoma. Endocr Relat Cancer, 2008, 15(2):475–83., DOI 10.1677/ERC-08-0002 Chen W, Xin B, Pang H, Han L, Shen W, Zhao Z, et al. Downregulation of estrogen receptor β inhibits lung adenocarcinoma cell growth. Oncol Rep, 2019, 41(5):2967–74., DOI 10.3892/or.2019.7044 Ivanova MM, Mazhawidza W, Dougherty SM, Klinge CM Sex differences in estrogen receptor subcellular location and activity in lung adenocarcinoma cells. Am J Respir Cel Mol Biol, 2010, 42(3):320–30., DOI 10.1165/rcmb.2009-0059OC Stabile LP, Davis AL, Gubish CT, Hopkins TM, Luketich JD, Christie N, et al. Human non-small cell lung tumors and cells derived from normal lung express both estrogen receptor alpha and beta and show biological responses to estrogen. Cancer Res, 2002, 62(7):2141–50. Klinge CM, Radde BN, Imbert-Fernandez Y, Teng Y, Ivanova MM, Abner SM, et al. Targeting the intracellular MUC1 C-terminal domain inhibits proliferation and estrogen receptor transcriptional activity in lung adenocarcinoma cells. Mol Cancer Ther, 2011, 10(11):2062–71., DOI 10.1158/ 1535-7163.MCT-11-0381 Monica V, Longo M, Felice B, Scagliotti GV, Papotti M, Novello S Role of hormone receptor expression in patients with advanced-stage lung cancer treated with chemotherapy. Clin Lung Cancer, 2012, 13(6):416–23., DOI 10. 1016/j.cllc.2012.03.006 Berardi R, Morgese F, Santinelli A, Onofri A, Biscotti T, Brunelli A, et al. Hormonal receptors in lung adenocarcinoma: expression and difference in outcome by sex. Oncotarget, 2016, 7(50):82648–57., DOI 10.18632/oncotarget. 12244 Hsu LH, Liu KJ, Tsai MF, Wu CR, Feng AC, Chu NM, et al. Estrogen adversely affects the prognosis of patients with lung adenocarcinoma. Cancer Sci, 2015, 106(1):51–9., DOI 10.1111/cas.12558 Smith CL, O’Malley BW Coregulator function: a key to understanding tissue specificity of selective receptor modulators. Endocr Rev, 2004, 25(1):45–71., DOI 10.1210/er.2003-0023 Murakami S, Nagari A, Kraus WL Dynamic assembly and activation of estrogen receptor α enhancers through coregulator switching. Genes Dev, 2017, 31(15):1535–48., DOI 10.1101/gad.302182.117 Manavathi B, Kumar R Steering estrogen signals from the plasma membrane to the nucleus: two sides of the coin. J Cel Physiol., 2006, 207(3):594–604., DOI 10.1002/jcp.20551 Acconcia F, Kumar R Signaling regulation of genomic and nongenomic functions of estrogen receptors. Cancer Lett, 2006, 238(1):1–14., DOI 10. 1016/j.canlet.2005.06.018 He L-R, Zhao H-Y, Li B-K, Zhang L-J, Liu M-Z, Kung H-F, et al. Overexpression of AIB1 negatively affects survival of surgically resected non-small-cell lung cancer patients. Ann Oncol, 2010, 21(8):1675–81., DOI 10.1093/annonc/mdp592 Wang H, Zhang D,WuW, Zhang J, Guo D, Wang Q, et al. Overexpression and gender-specific differences of SRC-3, SRC-3/AIB1, immunoreactivity in human non-small cell lung cancer: an in vivo study. J Histochem Cytochem, 2010, 58(12):1121–7., DOI 10.1369/jhc.2010.956979 Sareddy GR, Vadlamudi RK PELP1: structure, biological function and clinical significance. Gene, 2016, 585(1):128–34., DOI 10.1016/j.gene.2016.03.017 Girard BJ, Daniel AR, Lange CA, Ostrander JH PELP1: a review of PELP1 interactions, signaling, and biology. Mol Cell Endocrinol, 2014, 382(1):642–51., DOI 10.1016/j.mce.2013.07.031 Balasenthil S, Vadlamudi RK Functional interactions between the estrogen receptor coactivator PELP1/MNAR and retinoblastoma protein. J Biol Chem, 2003, 278(24):22119–27., DOI 10.1074/jbc.M212822200 Roy SS, Gonugunta VK, Bandyopadhyay A, Rao MK, Goodall GJ, Sun L-Z, et al. Significance of PELP1/HDAC2/miR-200 regulatory network in EMT and metastasis of breast cancer. Oncogene, 2014, 33(28):3707–16., DOI 10.1038/onc. 2013.332 Wan J, Li X PELP1/MNAR suppression inhibits proliferation and metastasis of endometrial carcinoma cells. Oncol Rep, 2012, 28(6):2035–42., DOI 10.3892/or. 2012.2038 Habashy HO, Powe DG, Rakha EA, Ball G, Macmillan RD, Green AR, et al. The prognostic significance of PELP1 expression in invasive breast cancer with emphasis on the ER-positive luminal-like subtype. Breast Cancer Res Treat, 2010, 120(3):603–12., DOI 10.1007/s10549-009-0419-9 Vadlamudi RK, Manavathi B, Balasenthil S, Nair SS, Yang Z, Sahin AA, et al. Functional implications of altered subcellular localization of PELP1 in breast cancer cells. Cancer Res, 2005, 65(17):7724–32., DOI 10.1158/0008-5472.CAN-05-0614 Vadlamudi RK, Balasenthil S, Broaddus RR, Gustafsson J-A, Kumar R Deregulation of estrogen receptor coactivator proline-, glutamic acid-, and leucine-rich protein-1/modulator of nongenomic activity of estrogen receptor in human endometrial tumors. J Clin Endocrinol Metab, 2004, 89(12):6130–8., DOI 10.1210/jc.2004-0909 Aust S, Horak P, Pils D, Pils S, Grimm C, Horvat R, et al. The prognostic value of estrogen receptor beta and proline-, glutamic acid- and leucine-rich protein 1, PELP1, expression in ovarian cancer. BMC cancer, 2013, 13(1):115., DOI 10. 1186/1471-2407-13-115 Grivas PD, Tzelepi V, Sotiropoulou-Bonikou G, Kefalopoulou Z, Papavassiliou AG, Kalofonos H Expression of ERα, ERβ and Co-regulator PELP1/MNAR in colorectal cancer: prognostic significance and clinicopathologic correlations. Anal Cell Pathol, 2009, 31(3):235–47., DOI 10.3233/CLO-2009-046710.1155/ 2009/697376 Ma C, Miao C, Wang C, Song F, Luo M PELP1 is a novel oncogene in gastric tumorigenesis and negatively regulated by miR-15 family microRNAs. Cbm, 2019, 26(1):1–9., DOI 10.3233/CBM-182279 Vadlamudi RK, Balasenthil S, Sahin AA, Kies M, Weber RS, Kumar R, et al. Novel estrogen receptor coactivator PELP1/MNAR gene and ERβ expression in salivary duct adenocarcinoma: potential therapeutic targets. Hum Pathol, 2005, 36(6):670–5., DOI 10.1016/j.humpath.2005.03.016 Marquez-Garban DC, Chen H-W, Fishbein MC, Goodglick L, Pietras RJ Estrogen receptor signaling pathways in human non-small cell lung cancer. Steroids, 2007, 72(2):135–43., DOI 10.1016/j.steroids.2006.11.019 Slowikowski BK, Galecki B, Dyszkiewicz W, Jagodzinski PP Increased expression of proline-, glutamic acid- and leucine-rich protein PELP1 in non-small cell lung cancer. Biomed. Pharmacother, 2015, 73:97–101., DOI 10.1016/j.biopha.2015.05.015 Ohshiro K, Rayala SK, Kondo S, Gaur A, Vadlamudi RK, El-Naggar AK, et al. Identifying the estrogen receptor coactivator PELP1 in autophagosomes. Cancer Res, 2007, 67(17):8164–71., DOI 10.1158/0008-5472.CAN-07-0038 Xu Y, Li Z, Jiang P,Wu G, Chen K, Zhang X, et al. The co-expression of MMP- 9 and Tenascin-C is significantly associated with the progression and prognosis of pancreatic cancer. Diagn Pathol, 2015, 10(1):211., DOI 10.1186/ s13000-015-0445-3 Grossi F, Loprevite M, Chiaramondia M, Ceppa P, Pera C, Ratto GB, et al. Prognostic significance of K-ras, p53, bcl-2, PCNA, CD34 in radically resected non-small cell lung cancers. Eur J Cancer, 2003, 39(9):1242–50., DOI 10.1016/ s0959-8049(03)00232-6 Wang X, Tsang JYS, Lee MA, Ni Y-B, Tong JH, Chan S-K, et al. The clinical value of PELP1 for breast cancer: a comparison with multiple cancers and analysis in breast cancer subtypes. Cancer Res Treat, 2019, 51(2):706–17., DOI 10.4143/crt.2018.316 Mishra SK, Balasenthil S, Nguyen D, Vadlamudi RK Cloning and functional characterization of PELP1/MNAR promoter. Gene, 2004, 330:115–22., DOI 10. 1016/j.gene.2004.01.011 Nair BC, Nair SS, Chakravarty D, Challa R, Manavathi B, Yew PR, et al. Cyclindependent kinase-mediated phosphorylation plays a critical role in the oncogenic functions of PELP1. Cancer Res, 2010, 70(18):7166–75., DOI 10. 1158/0008-5472.CAN-10-0628 Chakravarty D, Nair SS, Santhamma B, Nair BC, Wang L, Bandyopadhyay A, et al. Extranuclear functions of ER impact invasive migration andmetastasis by breast cancer cells. Cancer Res, 2010, 70(10):4092–101., DOI 10.1158/0008- 5472.CAN-09-3834 Dang DN, Raj G, Sarode V, Molberg KH, Vadlamudi RK, Peng Y Significantly increased PELP1 protein expression in primary and metastatic triple-negative breast carcinoma: comparison with GATA3 expression and PELP1’s potential role in triple-negative breast carcinoma. Hum Pathol, 2015, 46(12):1829–35., DOI 10.1016/j.humpath.2015.07.023 Roy S, Chakravarty D, Cortez V, De Mukhopadhyay K, Bandyopadhyay A, Ahn J-M, et al. Significance of PELP1 in ER-negative breast cancer metastasis. Mol Cancer Res, 2012, 10(1):25–33., DOI 10.1158/1541-7786. MCR-11-0456 Xu R, Shen H, Guo R, Sun J, Gao W, Shu Y Combine therapy of gefitinib and fulvestrant enhances antitumor effects on NSCLC cell lines with acquired resistance to gefitinib. Biomed Pharmacother, 2012, 66(5):384–9., DOI 10.1016/ j.biopha.2012.02.004 Zhu H, Cheng H, Ren Y, Liu ZG, Zhang YF, De Luo B Synergistic inhibitory effects by the combination of gefitinib and genistein on NSCLC with acquired drug-resistance in vitro and in vivo. Mol Biol Rep, 2012, 39(4):4971–9., DOI 10. 1007/s11033-011-1293-1 Ko J-C, Chiu H-C, Syu J-J, Jian Y-J, Chen C-Y, Jian Y-T, et al. Tamoxifen enhances erlotinib-induced cytotoxicity through down-regulating AKTmediated thymidine phosphorylase expression in human non-small-cell lung cancer cells. Biochem Pharmacol, 2014, 88(1):119–27., DOI 10.1016/j. bcp.2014.01.010 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 582443 EP 582453 DI 10.3389/pore.2021.582443 PG 11 ER PT J AU Yang, X Miao, S Mao, X Xiu, Ch Sun, J Pei, R Jia, Sh AF Yang, Xianguang Miao, Susheng Mao, Xionghui Xiu, Cheng Sun, Ji Pei, Rong Jia, Shenshan TI LncRNA LINC-PINT Inhibits Malignant Behaviors of Laryngeal Squamous Cell Carcinoma Cells via Inhibiting ZEB1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE laryngeal squamous cell carcinoma; ZEB1; migration; invasion; EZH2; LINC-PINT ID laryngeal squamous cell carcinoma; ZEB1; migration; invasion; EZH2; LINC-PINT AB Objective: Laryngeal squamous cell carcinoma (LSCC) belongs to head and neck squamous cell carcinoma (HNSCC), with dismal prognosis. Here, this study aims to disclose the role of LINC-PINT in cancer development, which may contribute to improving the clinical outcomes of LSCC treatment. Methods: LINC-PINT expression in LSCC tissues and in TU-177 and Hep-2 cells was quantified, and subsequently, the association between LINC-PINT and LSCC malignancies was analyzed. pcDNA3.1-LINC-PINT or pcDNA3.1-EZH2 was introduced into Hep-2 and TU-177 cells. qRT-PCR and Western blot analyses examined the levels of proteins related to the AKT/mTOR pathway and their phosphorylated proteins in Hep-2 and TU-177 cells. The viability as well as migration and invasion abilities of Hep-2 and TU-177 cells were determined. Also, the distribution of LINC-PINT in Hep-2 cells was investigated as well as the interplay between LINC-PINT and EZH2. The downstream genes that might interact with EZH2 were screened. Results: LINC-PINT expression was inhibited in LSCC tissues and in Hep-2 and TU-177 cells, whose downregulation was associated with unsatisfactory prognosis. LINC-PINT overexpression suppressed the proliferative, migratory and invasive capacities of Hep-2 and TU-177 cells. LINC-PINT, mainly expressing in nuclei, could enrich EZH2 to silence ZEB1. In Hep-2 and TU-177 cells, the inhibition of LINC-PINT or overexpression of ZEB1 could enhance cell proliferation, migration and invasion. The phosphorylated levels of proteins related to the AKT/mTOR pathway were declined in cells with LINC-PINT overexpression, and the levels of these phosphorylated proteins were increased in cells with LINC-PINT inhibition. Conclusion: LINC-PINT enriches EZH2 to silence ZEB1 and thus inhibits the proliferative, migratory, and invasive capacities of Hep-2 and TU-177 cells. In addition, LINC-PINT might exert its biological function through the AKT/mTOR pathway. C1 [Yang, Xianguang] Harbin Medical University Cancer Hospital, Department of Head and Neck SurgeryHarbin, China. [Miao, Susheng] Harbin Medical University Cancer Hospital, Department of Head and Neck SurgeryHarbin, China. [Mao, Xionghui] Harbin Medical University Cancer Hospital, Department of Head and Neck SurgeryHarbin, China. [Xiu, Cheng] Harbin Medical University Cancer Hospital, Department of Head and Neck SurgeryHarbin, China. [Sun, Ji] Harbin Medical University Cancer Hospital, Department of Head and Neck SurgeryHarbin, China. [Pei, Rong] Harbin Medical University Cancer Hospital, Department of Head and Neck SurgeryHarbin, China. [Jia, Shenshan] Harbin Medical University Cancer Hospital, Department of Head and Neck SurgeryHarbin, China. RP Jia, Sh (reprint author), Harbin Medical University Cancer Hospital, Department of Head and Neck Surgery, Harbin, China. CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer, 2015). 136(5):E359–86., DOI 10.1002/ijc.29210 Li X, Xu F, Meng Q, Gong N, Teng Z, Xu R, et al. Long noncoding RNA DLEU2 predicts a poor prognosis and enhances malignant properties in laryngeal squamous cell carcinoma through the miR-30c-5p/PIK3CD/Akt axis. Cell Death Dis, 2020). 11(6):472., DOI 10.1038/s41419-020-2581-2 Department of Veterans Affairs Laryngeal Cancer Study GroupWolf GT, Fisher SG, Hong WK, Hillman R, Spaulding M, et al. Induction chemotherapy plus radiation compared with surgery plus radiation in patients with advanced laryngeal cancer. N Engl J Med, 1991). 324(24):1685–90., DOI 10.1056/NEJM199106133242402 Rosenthal DI, Mohamed ASR, Weber RS, Garden AS, Sevak PR, Kies MS, et al. Long-term outcomes after surgical or nonsurgical initial therapy for patients with T4 squamous cell carcinoma of the larynx: a 3-decade survey. Cancer, 2015). 121(10):1608–19., DOI 10.1002/cncr.29241 Forastiere AA, Zhang Q, Weber RS, Maor MH, Goepfert H, Pajak TF, et al. Long-term results of RTOG 91-11: a comparison of three nonsurgical treatment strategies to preserve the larynx in patients with locally advanced larynx cancer. Jco, 2013). 31(7):845–52., DOI 10.1200/jco.2012.43.6097 Da CM, Cheng ZY, Gong CY, Nan W, Zhou KS, Zhao GH, et al. Role of HAND2-AS1 in human tumors. Clin Chim Acta, 2020). 511:189–97., DOI 10. 1016/j.cca.2020.10.020 Qin H, Xu J, Gong L, Jiang B, Zhao W The long noncoding RNA ST7-AS1 promotes laryngeal squamous cell carcinoma by stabilizing CARM1. Biochem Biophys Res Commun, 2019). 512(1):34–40., DOI 10.1016/j.bbrc.2019.02.057 Xiao D, Cui X,Wang X Long noncoding RNA XIST increases the aggressiveness of laryngeal squamous cell carcinoma by regulating miR-124-3p/EZH2. Exp Cell Res, 2019). 381(2):172–8., DOI 10.1016/j.yexcr.2019.04.034 Zheng X, Zhao K, Liu T, Liu L, Zhou C, Xu M Long noncoding RNA PVT1 promotes laryngeal squamous cell carcinoma development by acting as a molecular sponge to regulate miR-519d-3p. J Cell Biochem, 2019). 120(3): 3911–21., DOI 10.1002/jcb.27673 Yuan Z, Xiu C, Liu D, Zhou G, Yang H, Pei R, et al. Long noncoding RNA LINC-PINT regulates laryngeal carcinoma cell stemness and chemoresistance through miR-425-5p/PTCH1/SHH axis. J Cell Physiol, 2019). 234(12): 23111–22., DOI 10.1002/jcp.28874 Rong H, Chen B, Ma K, Wei X, Peng J, Zhu J Downregulation of lncRNA LINC-PINT participates in the recurrence of esophageal squamous cell carcinoma possibly by interacting miRNA-2. Cancer Biother Radiopharm, 2020). [Epub ahead of print]., DOI 10.1089/cbr.2019.3167 Comet I, Riising EM, Leblanc B, Helin K Maintaining cell identity: PRC2- mediated regulation of transcription and cancer. Nat Rev Cancer, 2016). 16(12):803–10., DOI 10.1038/nrc.2016.83 Di Croce L, Helin K Transcriptional regulation by Polycomb group proteins. Nat Struct Mol Biol, 2013). 20(10):1147–55., DOI 10.1038/nsmb.2669 Kim KH, Roberts CWM Targeting EZH2 in cancer. Nat Med, 2016). 22(2): 128–34., DOI 10.1038/nm.4036 Xu Y,Wang H, Li F, Heindl LM, He X, Yu J, et al. Long non-coding RNA LINCPINT suppresses cell proliferation and migration ofmelanoma via recruiting EZH2. Front Cell Dev Biol, 2019). 7:350., DOI 10.3389/fcell.2019.00350 Agarwal S, BehringM, Kim H-G, Bajpai P, Chakravarthi BVSK, Gupta N, et al. Targeting P4HA1 with a small molecule inhibitor in a colorectal cancer PDX model. Transl Oncol, 2020). 13(4):100754., DOI 10.1016/j.tranon.2020.100754 Chen H, Hou G, Yang J, Chen W, Guo L, Mao Q, et al. SOX9-activated PXNAS1 promotes the tumorigenesis of glioblastoma by EZH2-mediated methylation of DKK1. J Cell Mol Med, 2020). 24(11):6070–82., DOI 10.1111/ jcmm.15189 Chen M, Zheng Y, Xie J, Zhen E, Zhou X Integrative profiling analysis identifies the oncogenic long noncoding RNA DUXAP8 in oral cancer. Anticancer Drugs, 2020). 31(8):792–8., DOI 10.1097/CAD.0000000000000936 Izawa K, Shirakura K, Kakiuchi K, Funahashi N, Maekawa N, Hino N, et al. PRC2 components maintain DNA hypermethylation of the upstream promoter and regulate Robo4 expression in endothelial cells. Biol Pharm Bull, 2020). 43(4):742–6., DOI 10.1248/bpb.b19-01014 Li Y, Li H, Zhou L EZH2-mediated H3K27me3 inhibits ACE2 expression. Biochem Biophys Res Commun, 2020). 526(4):947–52., DOI 10.1016/j.bbrc.2020. 04.010 Wu L, Gong Y, Yan T, Zhang H LINP1 promotes the progression of cervical cancer by scaffolding EZH2, LSD1, and DNMT1 to inhibit the expression of KLF2 and PRSS8. Biochem Cell Biol, 2020). 98(5):591–9., DOI 10.1139/bcb- 2019-0446 Wu Q, Xiang S, Ma J, Hui P, Wang T, Meng W, et al. Long non-coding RNA CASC 15 regulates gastric cancer cell proliferation, migration and epithelial mesenchymal transition by targeting CDKN 1A and ZEB 1. Mol Oncol, 2018). 12(6):799–813., DOI 10.1002/1878-0261.12187 XuW, Yan Z,Hu F,Wei W, Yang C, Sun Z Long non-coding RNAGAS5 accelerates oxidative stress in melanoma cells by rescuing EZH2-mediated CDKN1C downregulation. Cancer Cell Int, 2020). 20:116., DOI 10.1186/s12935-020-01167-1 Kishikawa T, Otsuka M, Yoshikawa T, Ohno M, Ijichi H, Koike K Satellite RNAs promote pancreatic oncogenic processes via the dysfunction of YBX1. Nat Commun, 2016). 7:13006., DOI 10.1038/ncomms13006 Shen Z, Yuan J, Tong Q, Hao W, Deng H, Li Q, et al. Long non-coding RNA AC023794.4-201 exerts a tumor-suppressive function in laryngeal squamous cell cancer and may serve as a potential prognostic biomarker. Oncol Lett, 2020). 20(1):774–84., DOI 10.3892/ol.2020.11595 Wang X, Liu L, Zhao W, Li Q, Wang G, Li H LncRNA SNHG16 promotes the progression of laryngeal squamous cell carcinoma by mediating miR-877-5p/ FOXP4 Axis. Ott, 2020). 13:4569–79., DOI 10.2147/ott.s250752 Hong L, Wang H, Wang J, Wei S, Zhang F, Han J, et al. LncRNA PTCSC3 inhibits tumor growth and cancer cell stemness in gastric cancer by interacting with lncRNA linc-pint. Cmar, 2019). 11:10393–9., DOI 10.2147/cmar.s231369 Huang Q, Zhang D, Diao Q, Lin M lncRNA LINC-PINT is downregulated in melanoma and regulates cell proliferation by downregulating lncRNA BANCR. Oncol Lett, 2019). 18(3):2917–22., DOI 10.3892/ol.2019.10631 Zhang L, Chen J, Wang L, Chen L, Du Z, Zhu L, et al. Linc-PINT acted as a tumor suppressor by sponging miR-543 and miR-576-5p in esophageal cancer. J Cell Biochem, 2019). 120(12):19345–57., DOI 10.1002/jcb.28699 Wang S, Jiang W, Zhang X, Lu Z, Geng Q, Wang W, et al. LINC-PINT alleviates lung cancer progression via sponging miR-543 and inducing PTEN. Cancer Med, 2020). 9(6):1999–2009., DOI 10.1002/cam4.2822 Margueron R, Reinberg D The Polycomb complex PRC2 and its mark in life. Nature, 2011). 469(7330):343–9., DOI 10.1038/nature09784 Marin-Bejar O, Mas AM, Gonzalez J, Martinez D, Athie A, Morales X, et al. The human lncRNA LINC-PINT inhibits tumor cell invasion through a highly conserved sequence element. Genome Biol, 2017). 18(1):202., DOI 10.1186/ s13059-017-1331-y Duan J, Ma X, Shi J, Xuan Y, Wang H, Li P, et al. Long noncoding RNA LINCPINT promotes proliferation through EZH2 and predicts poor prognosis in clear cell renal cell carcinoma. Ott, 2019). 12:4729–40., DOI 10.2147/ott.s202938 Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, et al. Long noncoding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature, 2010). 464(7291):1071–6., DOI 10.1038/nature08975 Yang J, Tian B, Sun H, Garofalo RP, Brasier AR Epigenetic silencing of IRF1 dysregulates type III interferon responses to respiratory virus infection in epithelial to mesenchymal transition. Nat Microbiol, 2017). 2:17086., DOI 10. 1038/nmicrobiol.2017.86 Wang H, Qian J, Xia X, Ye B Long non-coding RNA OIP5-AS1 serves as an oncogene in laryngeal squamous cell carcinoma by regulating miR-204-5p/ ZEB1 axis. Naunyn Schmiedebergs Arch Pharmacol, 2020). 393(11):2177–84., DOI 10.1007/s00210-020-01811-7 Zhang J, Lu C, Zhang J, Kang J, Cao C, Li M Involvement of ZEB1 and E-cadherin in the invasion of lung squamous cell carcinoma. Mol Biol Rep, 2013). 40(2):949–56., DOI 10.1007/s11033-012-2136-4 Zhao R, Tian L, Zhao B, Sun Y, Cao J, Chen K, et al. FADS1 promotes the progression of laryngeal squamous cell carcinoma through activating AKT/ mTOR signaling. Cell Death Dis, 2020). 11(4):272., DOI 10.1038/s41419-020- 2457-5 Gonzalez ME, DuPrie ML, Krueger H, Merajver SD, Ventura AC, Toy KA, et al. Histone methyltransferase EZH2 induces Akt-dependent genomic instability and BRCA1 inhibition in breast cancer. Cancer Res, 2011). 71(6):2360–70., DOI 10.1158/0008-5472.can-10-1933 Liu Y, Lu C, Fan L, Wang J, Li T, Liu Z, et al. MiR-199a-5p targets ZEB1 to inhibit the epithelial-mesenchymal transition of ovarian ectopic endometrial stromal cells via PI3K/Akt/mTOR signal pathway in vitro and in vivo. Reprod Sci, 2020). 27(1):110–8., DOI 10.1007/s43032-019-00016-5 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 584466 EP 584476 DI 10.3389/pore.2021.584466 PG 11 ER PT J AU Aslam, N Abusharieh, E Abuarqoub, D Alhattab, D Jafar, H Alshaer, W Masad, R Awidi, A AF Aslam, Nazneen Abusharieh, Elham Abuarqoub, Duaa Alhattab, Dana Jafar, Hanan Alshaer, Walhan Masad, J. Razan Awidi, S. Abdalla TI An In Vitro Comparison of Anti-Tumoral Potential of Wharton’s Jelly and Bone Marrow Mesenchymal Stem Cells Exhibited by Cell Cycle Arrest in Glioma Cells (U87MG) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mesenchymal stem cells; glioblastoma; secretome; WJ-MSCs; BM-MSC; cell lysate MSC-condition media ID mesenchymal stem cells; glioblastoma; secretome; WJ-MSCs; BM-MSC; cell lysate MSC-condition media AB The therapeutic potential ofmesenchymal stem cells (MSCs) for variousmalignancies is currently under investigation due to their unique properties. However,many discrepancies regarding their anti-tumoral or pro-tumoral properties have raised uncertainty about their application for anticancer therapies. To investigate, if the anti-tumoral or pro-tumoral properties are subjective to the type of MSCs under different experimental conditions we set out these experiments. Three treatments namely cell lysates (CL), serum-free conditioned media and FBS conditioned media (FBSCM) from each of Wharton’s Jelly MSCs and BoneMarrow-MSCs were applied to evaluate the anti-tumoral or pro-tumoral effect on the glioma cells (U87MG). The functional analysis included; Morphological evaluation, proliferation and migration potential, cell cycle analysis, and apoptosis for glioma cells. The fibroblast cell line was added to investigate the stimulatory or inhibitory effect of treatments on the proliferation of the normal cell. We found that cell lysates induced a generalized inhibitory effect on the proliferation of the glioma cells and the fibroblasts from both types of MSCs. Similarly, both types of conditioned media from two types of MSCs exerted the same inhibitory effect on the proliferation of the glioma cells. However, the effect of two types of conditioned media on the proliferation of fibroblasts was stimulatory from BMMSCs and variable from WJ-MSCs. Moreover, all three treatments exerted a likewise inhibitory effect on themigration potential of the glioma cells. Furthermore,we found that the cell cycle was arrested significantly at the G1 phase after treating cells with conditionedmedia whichmay have led to inhibit the proliferative and migratory abilities of the glioma cells (U87MG).We conclude that cell extracts of MSCs in the form of secretome can induce specific anti-tumoral properties in serum-free conditions for the glioma cells particularly theWJ-MSCs and the effect is mediated by the cell cycle arrest at the G1 phase. C1 [Aslam, Nazneen] The University of Jordan, Cell Therapy CenterAmman, Jordan. [Abusharieh, Elham] The University of Jordan, Cell Therapy CenterAmman, Jordan. [Abuarqoub, Duaa] The University of Jordan, Cell Therapy CenterAmman, Jordan. [Alhattab, Dana] The University of Jordan, Cell Therapy CenterAmman, Jordan. [Jafar, Hanan] The University of Jordan, Cell Therapy CenterAmman, Jordan. [Alshaer, Walhan] The University of Jordan, Cell Therapy CenterAmman, Jordan. [Masad, J. Razan] The University of Jordan, Cell Therapy CenterAmman, Jordan. [Awidi, S. Abdalla] The University of Jordan, Cell Therapy CenterAmman, Jordan. RP Awidi, A (reprint author), The University of Jordan, Cell Therapy Center, Amman, Jordan. EM aabbadi@ju.edu.jo CR Brat DJ, Scheithauer BW, Fuller GN, Tihan T Newly codified glial neoplasms of the 2007 WHO classification of tumours of the central nervous system: angiocentric glioma, pilomyxoid astrocytoma and pituicytoma. Brain Pathol, 2007). 17(3):319–24., DOI 10.1111/j.1750-3639.2007.00082.x Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol, 2009). 10(5):459–66., DOI 10.1016/s1470-2045(09)70025-7 Gauthaman K, Fong C-Y, Arularasu S, Subramanian A, Biswas A, Choolani M, et al. HumanWharton’s Jelly stem cell conditioned medium and cell-free lysate inhibit human osteosarcoma and mammary carcinoma cell growth in vitro and in xenograft mice. J Cel Biochem, 2013). 114(2):366–77., DOI 10.1002/jcb.24367 Chamberlain G, Fox J, Ashton B, Middleton J Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem cells, 2007). 25, 11), 2739–49., DOI 10.1634/ stemcells.2007-0197 Miranda CO, Teixeira CA, Liz MA, Sousa VF, Franquinho F, Forte G, et al. Systemic delivery of bone marrow-derived mesenchymal stromal cells diminishes neuropathology in a mouse model of krabbe’s disease. Stem Cells, 2011). 29(11):1738–51., DOI 10.1002/stem.724 Kidd S, Spaeth E, Dembinski JL, Dietrich M, Watson K, Klopp A, et al. Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging. Stem cells, 2009). 27(10):2614–23., DOI 10.1002/stem.187 Kidd S, Caldwell L, Dietrich M, Samudio I, Spaeth EL, Watson K, et al. Mesenchymal stromal cells alone or expressing interferon-β suppress pancreatic tumors in vivo, an effect countered by anti-inflammatory treatment. Cytotherapy, 2010). 12(5):615–25., DOI 10.3109/14653241003631815 Loebinger MR, Kyrtatos PG, Turmaine M, Price AN, Pankhurst Q, Lythgoe MF, et al. Magnetic resonance imaging of mesenchymal stem cells homing to pulmonary metastases using biocompatible magnetic nanoparticles. Cancer Res, 2009). 69(23):8862–67., DOI 10.1158/0008-5472.can-09-1912 Sasportas LS, Kasmieh R, Wakimoto H, Hingtgen S, van de Water JAJM, Mohapatra G, et al. Assessment of therapeutic efficacy and fate of engineered human mesenchymal stem cells for cancer therapy. Proc Natl Acad Sci, 2009). 106(12):4822–27., DOI 10.1073/pnas.0806647106 Yang B, Wu X, Mao Y, Bao W, Gao L, Zhou P, et al. Dual-targeted antitumor effects against brainstem glioma by intravenous delivery of tumor necrosis factor-related, apoptosis-inducing, ligand-engineered human mesenchymal stem cells. Neurosurgery, 2009). 65(3):610–24., DOI 10.1227/01.neu. 0000350227.61132.a7 Roorda BD, ter Elst A, Kamps WA, de Bont ESJM Bone marrow-derived cells and tumor growth: contribution of bone marrow-derived cells to tumor micro-environments with special focus on mesenchymal stem cells. Crit Rev oncology/hematology, 2009). 69(3):187–98., DOI 10.1016/j. critrevonc.2008.06.004 Choumerianou DM, Dimitriou H, Perdikogianni C, Martimianaki G, Riminucci M, Kalmanti M Study of oncogenic transformation in ex vivo expanded mesenchymal cells, from paediatric bone marrow. Cel Prolif, 2008). 41(6):909–22., DOI 10.1111/j.1365-2184.2008.00559.x Zhang L, Xiang J, Li G The uncertain role of unmodified mesenchymal stem cells in tumor progression: what master switch?. Stem Cel Res Ther, 2013). 4(2):22., DOI 10.1186/scrt170 Bartosh TJ, Ullah M, Zeitouni S, Beaver J, Prockop DJ Cancer cells enter dormancy after cannibalizing mesenchymal stem/stromal cells, MSCs). Proc Natl Acad Sci USA, 2016). 113(42):E6447–E6456., DOI 10.1073/pnas. 1612290113 Feng B, Chen L Review of mesenchymal stem cells and tumors: executioner or coconspirator?. Cancer Biother Radiopharm, 2009). 24(6):717–21., DOI 10. 1089/cbr.2009.0652 Maffini MV, Soto AM, Calabro JM, Ucci AA, Sonnenschein C The stroma as a crucial target in rat mammary gland carcinogenesis. J Cel Sci, 2004). 117(8): 1495–502., DOI 10.1242/jcs.01000 Dezawa M, Kanno H, Hoshino M, Cho H, Matsumoto N, Itokazu Y, et al. Specific induction of neuronal cells from bone marrow stromal cells and application for autologous transplantation. J Clin Invest, 2004). 113(12): 1701–10., DOI 10.1172/jci200420935 Wislet-Gendebien S, Hans G, Leprince P, Rigo JM, Moonen G, Rogister B Plasticity of cultured mesenchymal stem cells: switch from nestin-positive to excitable neuron-like phenotype. Stem cells, 2005). 23(3):392–402., DOI 10. 1634/stemcells.2004-0149 Keilhoff G, Stang F, Goihl A, Wolf G, Fansa H Transdifferentiated mesenchymal stem cells as alternative therapy in supporting nerve regeneration and myelination. Cell Mol Neurobiol, 2006). 26(7-8):1233–50., DOI 10.1007/s10571-006-9029-9 Horwitz E, Dominici M How do mesenchymal stromal cells exert their therapeutic benefit?. Cytotherapy, 2008). 10, 8), 771–4., DOI 10.1080/ 14653240802618085 Rosario CM, Yandava BD, Kosaras B, Zurakowski D, Sidman RL, Snyder EY Differentiation of engrafted multipotent neural progenitors towards replacement of missing granule neurons in meander tail cerebellum may help determine the locus of mutant gene action. Development, 1997). 124(21): 4213–(24.) Li T-S, Takahashi M, Ohshima M, Qin S-L, Kubo M, Muramatsu K, et al. Myocardial repair achieved by the intramyocardial implantation of adult cardiomyocytes in combination with bone marrow cells. Cel Transpl, 2008). 17(6):695–703., DOI 10.3727/096368908786092702 Yang D, Wang W, Li L, Peng Y, Chen P, Huang H, et al. The relative contribution of paracine effect versus direct differentiation on adipose-derived stem cell transplantation mediated cardiac repair. PloS one, 2013). 8(3): e59020., DOI 10.1371/journal.pone.0059020 Kim HO, Choi S-M, Kim H-S Mesenchymal stem cell-derived secretome and microvesicles as a cell-free therapeutics for neurodegenerative disorders. Tissue Eng Regen Med, 2013). 10(3):93–101., DOI 10.1007/s13770-013-0010-7 Ramdasi S, Sarang S, Viswanathan C Potential of mesenchymal stem cell based application in cancer. Int J Hematol Oncol Stem Cel Res, 2015). 9(2):95 Bhang SH, Lee S, Shin J-Y, Lee T-J, Jang H-K, Kim B-S Efficacious and clinically relevant conditioned medium of human adipose-derived stem cells for therapeutic angiogenesis. Mol Ther, 2014). 22(4):862–72., DOI 10.1038/mt. 2013.301 Ho JCY, LaiW-H, LiM-F, Au K-W, Yip M-C, Wong NLY, et al. Reversal of endothelial progenitor cell dysfunction in patients with type 2 diabetes using a conditioned medium of human embryonic stem cell-derived endothelial cells. Diabetes Metab Res Rev, 2012). 28(5):462–73., DOI 10. 1002/dmrr.2304 Di Santo S, Yang Z, von Ballmoos MW, Voelzmann J, Diehm N, Baumgartner I, et al. Novel cell-free strategy for therapeutic angiogenesis: in vitro generated conditioned medium can replace progenitor cell transplantation. PloS one, 2009). 4(5):e5643., DOI 10.1371/journal.pone.0005643 Mirabella T, Cilli M, Carlone S, Cancedda R, Gentili C Amniotic liquid derived stem cells as reservoir of secreted angiogenic factors capable of stimulating neo-arteriogenesis in an ischemic model. Biomaterials, 2011). 32(15):3689–99., DOI 10.1016/j.biomaterials.2011.01.071 Kim J, Lee JH, Yeo SM, Chung HM, Chae J-I Stem cell recruitment factors secreted from cord blood-derived stem cells that are not secreted from mature endothelial cells enhance wound healing. In Vitro Cell.Dev.Biol.-Animal, 2014). 50(2):146–54., DOI 10.1007/s11626-013-9687-0 Lee MJ, Kim J, Lee KI, Shin JM, Chae JI, Chung HM Enhancement of wound healing by secretory factors of endothelial precursor cells derived from human embryonic stem cells. Cytotherapy, 2011). 13(2):165–78., DOI 10.3109/ 14653249.2010.512632 Parekkadan B, Van Poll D, Suganuma K, Carter EA, Berthiaume F, Tilles AW, et al. Mesenchymal stem cell-derived molecules reverse fulminant hepatic failure. PloS one, 2007). 2(9):e941., DOI 10.1371/journal.pone.0000941 Cantinieaux D, Quertainmont R, Blacher S, Rossi L, Wanet T, Noel A, et al. Conditioned medium from bone marrow-derived mesenchymal stem cells improves recovery after spinal cord injury in rats: an original strategy to avoid cell transplantation. PloS one, 2013). 8(8):e69515., DOI 10.1371/journal.pone.0069515 Rafiee MR, Malekzadeh Shafaroudi A, Rohban S, Khayatzadeh H, Kalhor HR, Mowla SJ Enrichment of a rare subpopulation of miR-302-expressing glioma cells by serum deprivation. Cell J, 2015). 16(4):494-505., DOI 10.22074/cellj. 2015.495 Zhao W, Ren G, Zhang L, Zhang Z, Liu J, Kuang P, et al. Efficacy of mesenchymal stem cells derived from human adipose tissue in inhibition of hepatocellular carcinoma cells in vitro. Cancer Biother Radiopharm, 2012). 27(9):606–13., DOI 10.1089/cbr.2011.1150 Ryu H, Oh J-E, Rhee K-J, Baik SK, Kim J, Kang SJ, et al. Adipose tissue-derived mesenchymal stem cells cultured at high density express IFN-β and suppress the growth of MCF-7 human breast cancer cells. Cancer Lett, 2014). 352(2): 220–7., DOI 10.1016/j.canlet.2014.06.018 Takahara K, Ii M, Inamoto T, Komura K, Ibuki N, Minami K, et al. Adiposederived stromal cells inhibit prostate cancer cell proliferation inducing apoptosis. Biochem biophysical Res Commun, 2014). 446(4):1102–7., DOI 10. 1016/j.bbrc.2014.03.080 Ahn JO, Coh YR, Lee HW, Shin IS, Kang SK, Youn HY Human adipose tissuederived mesenchymal stem cells inhibit melanoma growth in vitro and in vivo. Anticancer Res, 2015). 35(1):159–(68.) Bohrnsen F, Fricke M, Sander C, Leha A, Schliephake H, Kramer FJ Interactions of human MSC with head and neck squamous cell carcinoma cell line PCI-13 reduce markers of epithelia-mesenchymal transition. Clin Oral Invest, 2015). 19(5):1121–28., DOI 10.1007/s00784-014-1338-7 Secchiero P, Zorzet S, Tripodo C, Corallini F, Melloni E, Caruso L, et al. Human bone marrow mesenchymal stem cells display anti-cancer activity in SCID mice bearing disseminated non-Hodgkin’s lymphoma xenografts. PloS one, 2010). 5(6):e11140., DOI 10.1371/journal.pone.0011140 Wieder H, Beer AJ, Holzapfel K, Henninger M, Maurer T, Schwarzenboeck S, et al. 11C-choline PET/CT and whole-body MRI including diffusion-weighted imaging for patients with recurrent prostate cancer. Oncotarget, 2017). 8(39): 66516., DOI 10.18632/oncotarget.16227 Mehrabadi S, Sadr SS, Hoseini M Stem cell conditioned medium as a novel treatment for neuroinflamation diseases. Int J Med Invest, 2019). 8(3):1–(12.). Dahbour S, Jamali F, Alhattab D, Al-Radaideh A, Ababneh O, Al-Ryalat N, et al. Mesenchymal stem cells and conditioned media in the treatment of multiple sclerosis patients: clinical, ophthalmological and radiological assessments of safety and efficacy. CNS Neurosci Ther, 2017). 23(11): 866–74., DOI 10.1111/cns.12759 Gauthaman K, Fong C-Y, Suganya C-A, Subramanian A, Biswas A, Choolani M, et al. Extra-embryonic human Wharton’s jelly stem cells do not induce tumorigenesis, unlike human embryonic stem cells. Reprod Biomed Online, 2012). 24(2):235–46., DOI 10.1016/j.rbmo.2011.10.007 Bajetto A, Pattarozzi A, Corsaro A, Barbieri F, Daga A, Bosio A, et al. Different effects of human umbilical cord mesenchymal stem cells on glioblastoma stem cells by direct cell interaction or via released soluble factors. Front Cell Neurosci, 2017). 11:312., DOI 10.3389/fncel.2017.00312 Pavon LF, Marti LC, Sibov TT, Malheiros SM, Brandt RA, Cavalheiro S, et al. In vitro analysis of neurospheres derived from glioblastoma primary culture: a novel methodology paradigm. Front Neurol, 2014). 4:214., DOI 10.3389/fneur. 2013.00214 Wei X, Yang X, Han Z-p., Qu F-f., Shao L, Shi Y-f. Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol Sin, 2013). 34(6):747., DOI 10.1038/ aps.2013.50 Prockop DJ, Oh JY Medical therapies with adult stem/progenitor cells, MSCs): a backward journey from dramatic results in vivo to the cellular and molecular explanations. J Cel Biochem, 2012). 113(5):1460–1469., DOI 10.1002/jcb.24046 Russell KC, Phinney DG, Lacey MR, Barrilleaux BL, Meyertholen KE, O’Connor KC . In Vitro high-capacity assay to quantify the clonal heterogeneity in trilineage potential of mesenchymal stem cells reveals a complex hierarchy of lineage commitment. Stem cells, 2010). 28(4):788–98., DOI 10.1002/stem.312 Baglio SR, Pegtel DM, Baldini N Mesenchymal stem cell secreted vesicles provide novel opportunities in, stem, cell-free therapy. Front Physiol, 2012). 3: 359., DOI 10.3389/fphys.2012.00359 Yu JM, Jun ES, Bae YC, Jung JS Mesenchymal stem cells derived from human adipose tissues favor tumor cell growth in vivo. Stem Cell Dev, 2008). 17(3): 463–74., DOI 10.1089/scd.2007.0181 Studeny M,Marini FC, Champlin RE, Zompetta C, Fidler IJ, Andreeff M Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors. Cancer Res, 2002). 62(13):3603–(8.). Mishra PJ, Mishra PJ, Humeniuk R, Medina DJ, Alexe G, Mesirov JP, et al. Carcinoma-Associated fibroblast-like differentiation of human mesenchymal stem cells. Cancer Res, 2008). 68(11):4331–9., DOI 10.1158/0008-5472.can-08- 0943 Spaeth EL, Dembinski JL, Sasser AK, Watson K, Klopp A, Hall B, et al. Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PloS one, 2009). 4(4):e4992., DOI 10.1371/journal.pone.0004992 Shinagawa K, Kitadai Y, Tanaka M, Sumida T, Kodama M, Higashi Y, et al. Mesenchymal stem cells enhance growth and metastasis of colon cancer. Int J Cancer, 2010). 127(10):2323–33., DOI 10.1002/ijc.25440 Gleisner MA, Pereda C, Tittarelli A, Navarrete M, Fuentes C, Avalos I, et al. A heat-shocked melanoma cell lysate vaccine enhances tumor infiltration by prototypic effector T cells inhibiting tumor growth. J Immunother Cancer, 2020). 8(2):e000999., DOI 10.1136/jitc-2020-000999 Granada AE, Jimenez A, Stewart-Ornstein J, Bluthgen N, Reber S, Jambhekar A, et al. The effects of proliferation status and cell cycle phase on the responses of single cells to chemotherapy. MBoC, 2020). 31(8):845–57., DOI 10.1091/mbc. e19-09-0515 Healy L, Hunt C, Young L, Stacey G The UK Stem Cell Bank: its role as a public research resource centre providing access to well-characterised seed stocks of human stem cell lines. Adv Drug Deliv Rev, 2005). 57(13):1981–8., DOI 10.1016/ j.addr.2005.07.019 Spees JL, Gregory CA, Singh H, Tucker HA, Peister A, Lynch PJ, et al. Internalized antigens must be removed to prepare hypoimmunogenic mesenchymal stem cells for cell and gene therapy. Mol Ther, 2004). 9(5): 747–56., DOI 10.1016/j.ymthe.2004.02.012 Kunz-Schughart LA, Knuechel R Tumor-associated fibroblasts, part II): functional impact on tumor tissue. Histol Histopathol, 2002). 17, 623., DOI 10.14670/HH-17.623 Yamashita M, Ogawa T, Zhang X, Hanamura N, Kashikura Y, Takamura M, et al. Role of stromal myofibroblasts in invasive breast cancer: stromal expression of alpha-smooth muscle actin correlates with worse clinical outcome. Breast cancer, 2012). 19(2):170–6., DOI 10.1007/s12282-010-0234-5 Akimoto K, Kimura K, Nagano M, Takano S, To’a Salazar G, Yamashita T, et al. Umbilical cord blood-derived mesenchymal stem cells inhibit, but adipose tissuederived mesenchymal stem cells promote, glioblastoma multiforme proliferation. Stem Cell Dev, 2012). 22(9):1370–86., DOI 10.1089/scd.2012.0486 Kim D-W, Staples M, Shinozuka K, Pantcheva P, Kang S-D, Borlongan C Wharton’s jelly-derived mesenchymal stem cells: phenotypic characterization and optimizing their therapeutic potential for clinical applications. Int. J. Mol. Sci, 2013). 14(6):11692–712., DOI 10.3390/ijms140611692 Rachakatla RS, Marini F, Weiss ML, Tamura M, Troyer D Development of human umbilical cord matrix stem cell-based gene therapy for experimental lung tumors. Cancer Gene Ther, 2007). 14(10):828., DOI 10.1038/sj.cgt.7701077 Chao K-C, Yang H-T, Chen M-W Human umbilical cord mesenchymal stem cells suppress breast cancer tumourigenesis through direct cell-cell contact and internalization. J Cell Mol Med, 2012). 16(8):1803–15., DOI 10.1111/j.1582- 4934.2011.01459.x Ma Y, Hao X, Zhang S, Zhang J The in vitro and in vivo effects of human umbilical cord mesenchymal stem cells on the growth of breast cancer cells. Breast Cancer Res Treat, 2012). 133(2):473–85., DOI 10.1007/s10549-011- 1774-x Fan C-G, Zhang Q-j., Zhou J-r. Therapeutic potentials of mesenchymal stem cells derived from human umbilical cord. Stem Cel Rev Rep, 2011). 7(1): 195–207., DOI 10.1007/s12015-010-9168-8 Pevsner-Fischer M, Levin S, Zipori D The origins of mesenchymal stromal cell heterogeneity. Stem Cel Rev Rep, 2011). 7(3):560–8., DOI 10.1007/s12015-011- 9229-7 Eckschlager T, Plch J, Stiborova M, Hrabeta J Histone deacetylase inhibitors as anticancer drugs. Int. J. Mol. Sci, 2017). 18, 7):1414., DOI 10.3390/ ijms18071414 Santamaria D, Barriere C, Cerqueira A, Hunt S, Tardy C, Newton K, et al. Cdk1 is sufficient to drive the mammalian cell cycle. Nature, 2007). 448:811–5., DOI 10.1038/nature06046 Gerard C, Goldbeter A The balance between cell cycle arrest and cell proliferation: control by the extracellular matrix and by contact inhibition. Interf Focus, 2014). 4(3):20130075., DOI 10.1098/rsfs.2013.0075 Bruschini S, Ciliberto G, Mancini R The emerging role of cancer cell plasticity and cell-cycle quiescence in immune escape. Cell Death Dis, 2020). 11:471., DOI 10.1038/s41419-020-2669-8 Zamarin D, Holmgaard RB, Subudhi SK, Park JS, Mansour M, Palese P, et al. Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy. Sci translational Med, 2014). 6(226):226ra32., DOI 10.1126/scitranslmed.3008095 Kelly JJP, Stechishin O, Chojnacki A, Lun X, Sun B, Senger DL, et al. Proliferation of human glioblastoma stem cells occurs independently of exogenous mitogens. Stem cells, 2009). 27(8):1722–33., DOI 10.1002/stem.98 Tavaluc RT, Hart LS, Dicker DT, El-Deiry WS Effects of low confluency, serum starvation and hypoxia on the side population of cancer cell lines. Cell cycle, 2007). 6(20):2554–62., DOI 10.4161/cc.6.20.4911 Lopez-Lazaro M, Pastor N, Azrak SS, Ayuso MJ, Austin CA, Cortes F Digitoxin inhibits the growth of cancer cell lines at concentrations commonly found in cardiac patients. J Nat Prod, 2005). 68(11):1642–5., DOI 10.1021/np050226l NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 584710 EP 584726 DI 10.3389/pore.2021.584710 PG 17 ER PT J AU Choi, IS AF Choi, Il Sun TI Effects of Gallotannin-Enriched Extract of Galla Rhois on the Activation of Apoptosis, Cell Cycle Arrest, and Inhibition of Migration Ability in LLC1 Cells and LLC1 Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE galla rhois; gallotanin; apoptosis; migration; NF-kB signaling pathway; lung carcinoma ID galla rhois; gallotanin; apoptosis; migration; NF-kB signaling pathway; lung carcinoma AB Gallotannin (GT) and GT-enriched extracts derived from various sources are reported to have anti-tumor activity in esophageal, colon and prostate tumors, although their antitumor effects have not been determined in lung carcinomas. To investigate the anti-tumor activity of GT-enriched extract of galla rhois (GEGR) against lung carcinomas, alterations in the cytotoxicity, apoptosis activation, cell cycle progression, migration ability, tumor growth, histopathological structure, and the regulation of signaling pathways were analyzed in Lewis lung carcinoma (LLC1) cells and LLC1 tumor bearing C57BL/6NKorl mice, after exposure to GEGR. A high concentration of GT (69%) and DPPH scavenging activity (IC50=7.922 μg/ml) was obtained in GEGR. GEGR treatment exerted strong cytotoxicity, cell cycle arrest at the G2/M phase and subsequent activation of apoptosis, as well as inhibitory effects on the MAPK pathway and PI3K/AKT mediated cell migration in LLC1 cells. In the in vivo syngeneic model, exposure to GEGR resulted in suppressed growth of the LLC1 tumors, as well as inhibition of NF-κB signaling and their inflammatory cytokines. Taken together, our results provide novel evidence that exposure to GEGR induces activation of apoptosis, cell cycle arrest, and inhibition of cell migration via suppression of the MAPK, NF-κB and PI3K/AKT signaling pathways in LLC1 cells and the LLC1 syngeneic model. C1 [Choi, Il Sun] Research Institute of National Cancer Center, Goyang, Division of Convergence TechnologyGoyang, South Korea. CR Cai Y, Luo Q, Sun M, and Corke H. Antioxidant activity and phenolic compounds of 112 traditional Chinese medicinal plants associated with anticancer. Life Sci, 2004, 74:2157–84., DOI 10.1016/j.lfs.2003.09.047 Chen G-H, Lin Y-L, HsuW-L, Hsieh S-K, and Tzen JTC. Significant elevation of antiviral activity of strictinin from Pu’er tea after thermal degradation to ellagic acid and gallic acid. J Food Drug Anal, 2015, 23:116–23., DOI 10.1016/j. jfda.2014.07.007 Yen G-C, Duh P-D, and Tsai H-L. Antioxidant and pro-oxidant properties of ascorbic acid and gallic acid. Food Chem, 2002, 79:307–13., DOI 10.1016/s0308- 8146(02)00145-0 Jagan S, Ramakrishnan G, Anandakumar P, Kamaraj S, and Devaki T. Antiproliferative potential of gallic acid against diethylnitrosamine-induced rat hepatocellular carcinoma. Mol Cel Biochem, 2008, 319:51–9., DOI 10.1007/ s11010-008-9876-4 Kim S-H, Jun C-D, Suk K, Choi B-J, Lim H, Park S, et al. Gallic acid inhibits histamine release and pro-inflammatory cytokine production in mast cells. Toxicol Sci, 2006, 91:123–31., DOI 10.1093/toxsci/kfj063 Al-Halabi R, Bou Chedid M, Abou Merhi R, El-Hajj H, Zahr H, Schneider-Stock R, et al. Gallotannin inhibits NFĸB signaling and growth of human colon cancer xenografts. Cancer Biol Ther, 2011, 12:59–68., DOI 10.4161/cbt.12.1.15715 Zhao T, Sun Q, del Rincon SV, Lovato A, Marques M, and Witcher M. Gallotannin imposes S phase arrest in breast cancer cells and suppresses the growth of triple-negative tumors in vivo. PLoS One, 2014, 9:e92853., DOI 10. 1371/journal.pone.0092853 Uruena C, Mancipe J, Hernandez J, Castaneda D, Pombo L, Gomez A, et al. Gallotannin-rich Caesalpinia spinosa fraction decreases the primary tumor and factors associated with poor prognosis in a murine breast cancer model. BMC Complement Altern Med, 2013, 13:74., DOI 10.1186/1472- 6882-13-74 Chen H-M, Wu Y-C, Chia Y-C, Chang F-R, Hsu H-K, Hsieh Y-C, et al. Gallic acid, a major component of Toona sinensis leaf extracts, contains a ROSmediated anti-cancer activity in human prostate cancer cells. Cancer Lett, 2009, 286:161–71., DOI 10.1016/j.canlet.2009.05.040 Faried A, Kurnia D, Faried LS, Usman N,Miyazaki T, Kato H, et al. Anticancer effects of gallic acid isolated from Indonesian herbal medicine, Phaleria macrocarpa, Scheff., Boerl, on human cancer cell lines. Int J Oncol, 2007, 30:605–13., DOI 10.3892/ijo.30.3.605 Ho H-H, Chang C-S, Ho W-C, Liao S-Y, Wu C-H, and Wang C-J. Antimetastasis effects of gallic acid on gastric cancer cells involves inhibition of NF- κB activity and downregulation of PI3K/AKT/small GTPase signals. Food Chem Toxicol, 2010, 48:2508–16., DOI 10.1016/j.fct.2010.06.024 Kuo C-L, Lai K-C, Ma Y-S, Weng S-W, Lin J-P, and Chung J-G. Gallic acid inhibits migration and invasion of SCC-4 human oral cancer cells through actions of NF-κB, Ras and matrix metalloproteinase-2 and -9. Oncol Rep, 2014, 32:355–61., DOI 10.3892/or.2014.3209 Zhao B, and Hu M. Gallic acid reduces cell viability, proliferation, invasion and angiogenesis in human cervical cancer cells. Oncol Lett, 2013, 6:1749–55., DOI 10.3892/ol.2013.1632 Ji B-C, Hsu W-H, Yang J-S, Hsia T-C, Lu C-C, Chiang J-H, et al. Gallic acid induces apoptosis via caspase-3 and mitochondrion-dependent pathways in vitro and suppresses lung xenograft tumor growth in vivo. J Agric Food Chem, 2009, 57:7596–604., DOI 10.1021/jf901308p Go J, Kim JE, Koh EK, Song SH, Kang HG, Lee YH, et al. Hepatoprotective effect of gallotannin-enriched extract isolated from gall on hydrogen peroxideinduced cytotoxicity in HepG2 cells. Pharmacogn Mag, 2017, 13:S294–S300., DOI 10.4103/pm.pm_424_15 Lee SM, Lee DW, Park JD, and Kim JI. Study on formation and development of Schlechtendalia chinensis gall in Rhus javanica. Korean JAppl Entomol, 1997, 36:83–7. Ren Z, Zhu B, Wang D, Ma E, Su D, and Zhong Y. Comparative population structure of Chinese sumac aphid Schlechtendalia chinensis and its primary host-plant Rhus chinensis. Genetica, 2008, 132:103–12., DOI 10.1007/s10709- 007-9153-6 Kim JE, Go J, Koh EK, Song SH, Sung JE, Lee HA, et al. Gallotannin-enriched extract isolated from galla rhois may be a functional candidate with laxative effects for treatment of loperamide-induced constipation of SD rats. PLoS One, 2016, 11:e0161144., DOI 10.1371/journal.pone.0161144 Mun JG, Kee JY, Han YH, Lee S, Park SH, Jeon HD, et al. Galla Rhois water extract inhibits lung metastasis by inducing AMPK-mediated apoptosis and suppressing metastatic properties of colorectal cancer cells. Oncol Rep, 2019, 41:202–12., DOI 10.3892/or.2018.6812 Go J, Kim J, Koh E, Song S, Sung J, Lee H, et al. Protective effect of gallotanninenriched extract isolated from galla rhois against CCl4-induced hepatotoxicity in ICR mice. Nutrients, 2016, 8:107., DOI 10.3390/nu8030107 Kwon OJ, Bae J-S, Lee H, Hwang J-Y, Lee E-W, Ito H, et al. Pancreatic lipase inhibitory gallotannins from Galla Rhois with inhibitory effects on adipocyte differentiation in 3T3-L1 cells. Molecules, 2013, 18:10629–38., DOI 10.3390/ molecules180910629 Lee K, Kim J, Lee BJ, Park JW, Leem KH, and Bu Y. Protective effects of Galla Rhois, the excrescence produced by the sumac aphid, Schlechtendalia chinensis, on transient focal cerebral ischemia in the rat. J Insect Sci, 2012, 12:10., DOI 10.1673/031.012.0110 Choi JG, Mun SH, Chahar HS, Bharaj P, Kang OH, Kim SG, et al. Methyl gallate from Galla rhois successfully controls clinical isolates of Salmonella infection in both in vitro and in vivo systems. PLoS One, 2014, 9:e102697., DOI 10.1371/journal.pone.0102697 Yim N-H, Gu MJ, Hwang Y-H, Cho W-K, and Ma JY. Water extract of Galla Rhois with steaming process enhances apoptotic cell death in human colon cancer cells. Integr Med Res, 2016, 5:284–92., DOI 10.1016/j.imr. 2016.10.001 Lee Y-H, Hwang E-K, Baek Y-M, and Kim H-D. Deodorizing function and antibacterial activity of fabrics dyed with gallnut, Galla Chinensis, extract. Textile Res J, 2015, 85:1045–54., DOI 10.1177/0040517514559580 Wang H-L, Chang J-C, Fang L-W, Hsu H-F, Lee L-C, Yang J-F, et al. Bulnesia sarmientoi supercritical fluid extract exhibits necroptotic effects and antimetastatic activity on lung cancer cells. Molecules, 2018, 23:3304., DOI 10.3390/ molecules23123304 Livak KJ, and Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods, 2001, 25: 402–408., DOI 10.1006/meth.2001.1262 da Silva Faria MC, Santos NAGd., Carvalho Rodrigues MA, Rodrigues JL, Barbosa Junior F, and Santos ACd. Effect of diabetes on biodistribution, nephrotoxicity and antitumor activity of cisplatin in mice. Chem Biol Interact, 2015, 229:119–31., DOI 10.1016/j.cbi.2015.01.027 Wu H-T, Lu F-H, Su Y-C, Ou H-Y, Hung H-C, Wu J-S, et al. In vivo and in vitro anti-tumor effects of fungal extracts. Molecules, 2014, 19:2546–56., DOI 10.3390/molecules19022546 Go J, Kim J-E, Koh E-K, Song S-H, Seung J-E, Park C-K, et al. Hepatotoxicity and nephrotoxicity of gallotannin-enriched extract isolated from Galla Rhois in ICR mice. Lab Anim Res, 2015, 31: 101–10., DOI 10.5625/lar.2015.31.3.101 Wang J, Zhao Z, Shen S, Zhang C, Guo S, Lu Y, et al. Selective depletion of tumor neovasculature by microbubble destruction with appropriate ultrasound pressure. Int J Cancer, 2015, 137:2478–91., DOI 10.1002/ijc.29597 Koyuncu I. Evaluation of anticancer, antioxidant activity and phenolic compounds of Artemisia absinthium L. extract. Cel Mol Biol, Noisy-legrand,, 2018, 64:25–34., DOI 10.14715/cmb/2018.64.3.5 Miyata Y, Shida Y, Hakariya T, and Sakai H. Anti-cancer effects of green tea polyphenols against prostate cancer. Molecules, 2019, 24:193., DOI 10.3390/ molecules24010193 Suh S-S,Hong J-M, KimEJ, Jung SW, KimS-M, KimJE, et al.Anti-inflammation and anti-cancer activity of ethanol extract of antarctic freshwater microalga, micractinium sp. Int J Med Sci, 2018, 15:929–36., DOI 10.7150/ijms.26410 Qin Y, Conley AP, Grimm EA, and Roszik J. A tool for discovering drug sensitivity and gene expression associations in cancer cells. PLoS One, 2017, 12:e0176763., DOI 10.1371/journal.pone.0176763 Baig S, Seevasant I, Mohamad J, Mukheem A, Huri HZ, and Kamarul T. Potential of apoptotic pathway-targeted cancer therapeutic research: where do we stand?. Cell Death Dis, 2016, 7:e2058., DOI 10.1038/cddis.2015.275 Adams JM. Ways of dying: multiple pathways to apoptosis. Genes Dev, 2003, 17:2481–95., DOI 10.1101/gad.1126903 Vitagliano O, Addeo R, D’Angelo V, Indolfi C, Indolfi P, and Casale F. The Bcl-2/Bax and Ras/Raf/MEK/ERK signaling pathways: implications in pediatric leukemia pathogenesis and new prospects for therapeutic approaches. Expert Rev Hematol, 2013, 6(5):587–97., DOI 10.1586/ 17474086.2013.827415 Dhanalakshmi S, Singh RP, Agarwal C, and Agarwal R. Silibinin inhibits constitutive and TNFα-induced activation of NF-κB and sensitizes human prostate carcinoma DU145 cells to TNFα-induced apoptosis. Oncogene, 2002, 21:1759–67., DOI 10.1038/sj.onc.1205240 Ghosh S, and Hayden MS. New regulators of NF-κB in inflammation. Nat Rev Immunol, 2008, 8:837–48., DOI 10.1038/nri2423 Prasad S, Ravindran J, and Aggarwal BB. NF-κB and cancer: how intimate is this relationship. Mol Cel Biochem, 2010, 336:25–37., DOI 10.1007/s11010-009- 0267-2 Jandial R. Metastatic cancer: clinical and biological perspectives. Austin: Landes Bioscience, 2013) TraceyAM, Lin Y, Andrew JS, Jane L, and WenGJ. Cancer invasion andmetastasis: molecular and cellular perspective. Austin: Landes Bioscience, 2013) Folkman J, and Shing Y. Angiogenesis. J Biol Chem, 1992, 267:10931–4., DOI 10.1016/s0021-9258(19)49853-0 Martin TA, and Jiang WG. Loss of tight junction barrier function and its role in cancer metastasis. Biochim Biophys Acta, 2009, 1788:872–91., DOI 10.1016/j. bbamem.2008.11.005 Bravo-Cordero JJ, Hodgson L, and Condeelis J. Directed cell invasion and migration during metastasis. Curr Opin Cel Biol, 2012, 24:277–83., DOI 10. 1016/j.ceb.2011.12.004 Kim D, Kim S, Koh H, Yoon SO, Chung AS, Cho KS, et al. Akt/PKB promotes cancer cell invasion via increased motility and metalloproteinase production. FASEB J, 2001, 15:1953–62., DOI 10.1096/fj.01-0198com Yang SX, Polley E, and Lipkowitz S. New insights on PI3K/AKT pathway alterations and clinical outcomes in breast cancer. Cancer Treat Rev, 2016, 45: 87–96., DOI 10.1016/j.ctrv.2016.03.004 Zhang L, Wang J-N, Tang J-M, Kong X, Yang J-Y, Zheng F, et al. VEGF is essential for the growth and migration of human hepatocellular carcinoma cells. Mol Biol Rep, 2012, 39:5085–93., DOI 10.1007/s11033-011-1304-2 Webb AH, Gao BT, Goldsmith ZK, Irvine AS, Saleh N, Lee RP, et al. Inhibition of MMP-2 and MMP-9 decreases cellular migration, and angiogenesis in in vitro models of retinoblastoma. BMC Cancer, 2017, 17:434., DOI 10.1186/ s12885-017-3418-y Malinda KM, Ponce L, Kleinman HK, Shackelton LM, and Millis AJT. Gp38k, a protein synthesized by vascular smooth muscle cells, stimulates directional migration of human umbilical vein endothelial cells. Exp Cel Res, 1999, 250: 168–73., DOI 10.1006/excr.1999.4511 Nishikawa KC, and Millis AJT. gp38k, CHI3L1, is a novel adhesion and migration factor for vascular cells. Exp Cel Res, 2003, 287:79–87., DOI 10.1016/ s0014-4827(03)00069-7 Recklies AD, White C, and Ling H. The chitinase 3-like protein human cartilage glycoprotein 39, HC-gp39, stimulates proliferation of human connective-tissue cells and activates both extracellular signal-regulated kinase- and protein kinase B-mediated signalling pathways. Biochem J, 2002, 365:119–26., DOI 10.1042/bj20020075 Ku BM, Lee YK, Ryu J, Jeong JY, Choi J, Eun KM, et al. CHI3L1, YKL-40, is expressed in human gliomas and regulates the invasion, growth and survival of glioma cells. Int J Cancer, 2011, 128:1316–26., DOI 10.1002/ ijc.25466 Newman DJ. Natural products as leads to potential drugs: an old process or the new hope for drug discovery?. J Med Chem, 2008, 51:2589–99., DOI 10.1021/ jm0704090 Efange SMN. Natural products: a continuing source of inspiration for the medical chemist: advances in phytomedicine. Amsterdam: Elsevier Science, 2002) Cragg GM, and Newman DJ. Drug discovery and development from natural products: the way forward. 11th NAPRECA Symposium Book of Proceedings, Antananarivo: Natural Products Research Network for Eastern and Central Africa, NAPRECA,, 2005) NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 588084 EP 588101 DI 10.3389/pore.2021.588084 PG 18 ER PT J AU Sato, N Ise, K Hata, Sh Yamashita, Sh Ito, A Sasano, H Nakamura, Y AF Sato, Naomi Ise, Kazue Hata, Shuko Yamashita, Shinichi Ito, Akihiro Sasano, Hironobu Nakamura, Yasuhiro TI Clinicopathological Significance of Estrogen Receptor β and Estrogen Synthesizing/Metabolizing Enzymes in Urothelial Carcinoma of Urinary Bladder SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ERβ; urothelial bladder carcinoma; steroid metabolism; steroid sulfatase; aromatase; estrogen sulfotransferase 5 ID ERβ; urothelial bladder carcinoma; steroid metabolism; steroid sulfatase; aromatase; estrogen sulfotransferase 5 AB Sex-specific differences in the incidence of urinary bladder carcinomas are well known, and the possible involvement of sex steroids has been proposed. We previously reported the association of the loss of androgen receptors and androgen-producing enzymes with tumor progression of urinary bladder cancer patients. Clinically, the selective estrogen receptor modulators (SERMs) were reported to suppress the progression of these tumors but the status of estrogen receptors (ERs) has not been well studied in patients with bladder urinary cancer. Moreover, not only ERs but also estrogen-related enzymes, such as aromatase, steroid sulfatase (STS), and estrogen sulfotransferase (EST), have been reported in the biological/clinical behavior of various hormone-dependent carcinomas but not studied in urinary bladder carcinoma. Therefore, in this study, we immunolocalized ERs as well as estrogen metabolizing enzymes in urinary bladder carcinoma and performed immunoblotting and cell proliferation assays using the bladder urothelial carcinoma cell line, T24. The results revealed that the loss of STS and aromatase was significantly correlated with advanced stages of the carcinoma. In vitro studies also revealed that T24 cell proliferation rates were significantly ameliorated after treatment with estradiol or diarylpropionitrile (DPN). EST and aromatase were also significantly correlated with the nuclear grade of the carcinoma. The results of our present study, for the first time, demonstrated that biologically active estrogens that bind to ERs could suppress tumor progression and the inactive ones could promote its progression and the potential clinical utility of SERM treatment in selective patients with urinary bladder carcinoma. C1 [Sato, Naomi] Sendai City Hospital, Division of PathologySendai, Japan. [Ise, Kazue] Tohoku Medical and Pharmaceutical University, Faculty of Medicine, Division of PathologySendai, Japan. [Hata, Shuko] Tohoku Medical and Pharmaceutical University, Faculty of Medicine, Division of PathologySendai, Japan. [Yamashita, Shinichi] Tohoku University Graduate School of Medicine, Department of UrologySendai, Japan. [Ito, Akihiro] Tohoku University Graduate School of Medicine, Department of UrologySendai, Japan. [Sasano, Hironobu] Tohoku Medical and Pharmaceutical University, Faculty of Medicine, Division of PathologySendai, Japan. [Nakamura, Yasuhiro] Tohoku Medical and Pharmaceutical University, Faculty of Medicine, Division of PathologySendai, Japan. RP Nakamura, Y (reprint author), Tohoku Medical and Pharmaceutical University, Faculty of Medicine, Division of Pathology, Sendai, Japan. EM yasu-naka@tohoku-mpu.ac.jp CR Burger M, Catto JWF, Dalbagni G, Grossman HB, Herr H, Karakiewicz P, et al. Epidemiology and risk factors of urothelial bladder cancer. Eur Urol, 2013). 63(2):234–41., DOI 10.1016/j.eururo.2012.07.033 Madeb R,MessingEMGender, racial and age differences in bladder cancer incidence and mortality. Urol Oncol, 2004). 22(2):86–92., DOI 10.1016/s1078-1439(03)00139-x Tuygun C, Kankaya D, Imamoglu A, Sertcelik A, Zengin K, Oktay M, et al. Sexspecific hormone receptors in urothelial carcinomas of the human urinary bladder: a comparative analysis of clinicopathological features and survival outcomes according to receptor expression. Urol Oncol, 2011). 29(1):43–51., DOI 10.1016/j.urolonc.2009.01.033 Boorjian S, Ugras S, Mongan NP, Gudas LJ, You X, Tickoo SK, et al. Androgen receptor expression is inversely correlated with pathologic tumor stage in bladder cancer. Urology, 2004). 64(2):383–8., DOI 10.1016/j.urology.2004.03.025 McGrath M, Michaud DS, De Vivo I Hormonal and reproductive factors and the risk of bladder cancer in women. Am J Epidemiol, 2005). 163(3):236–44., DOI 10.1093/aje/kwj028 Suzuki T, Miki Y, Nakamura Y, Ito K, Sasano H Steroid sulfatase and estrogen sulfotransferase in human carcinomas. Mol Cell Endocrinol, 2011). 340(2): 148–53., DOI 10.1016/j.mce.2010.11.001 Begam AJ, Jubie S, Nanjan MJ Estrogen receptor agonists/antagonists in breast cancer therapy: a critical review. Bioorg Chem, 2017). 71:257–74., DOI 10.1016/j. bioorg.2017.02.011 Ghosh DHuman sulfatases: a structural perspective to catalysis. Cell Mol Life Sci, 2007). 64(15):2013–22., DOI 10.1007/s00018-007-7175-y Evans T, Rowlands M, Law M, Coombes R Intratumoral oestrone sulphatase activity as a prognostic marker in human breast carcinoma. Br J Cancer, 1994). 69:555–61., DOI 10.1038/bjc.1994.101 Utsunomiya H, Ito K, Suzuki T, Kitamura T, Kaneko C, Nakata T, et al. Steroid sulfatase and estrogen sulfotransferase in human endometrial carcinoma. Clin Cancer Res, 2004). 10(17):5850–6., DOI 10.1158/1078-0432.ccr-04-0040 Okuda T, Saito H, Sekizawa A, Shimizu Y, Akamatsu T, Kushima M, et al. Steroid sulfatase expression in ovarian clear cell adenocarcinoma: immunohistochemical study. Gynecol Oncol, 2001). 82(3):427–34., DOI 10. 1006/gyno.2001.6322 Nakamura Y, Suzuki T, Fukuda T, Ito A, Endo M, Moriya T, et al. Steroid sulfatase and estrogen sulfotransferase in human prostate cancer. Prostate, 2006). 66(9):1005–12., DOI 10.1002/pros.20426 Sato R, Suzuki T, Katayose Y, Miura K, Shiiba K, Tateno H, et al. Steroid sulfatase and estrogen sulfotransferase in colon carcinoma: regulators of intratumoral estrogen concentrations and potent prognostic factors. Cancer Res, 2009). 69(3):914–22., DOI 10.1158/0008-5472.can-08-0906 Utsumi T, Yoshimura N, Maruta M, Takeuchi S, Ando J, Maeda K, et al. Significance of steroid sulfatase expression in human breast cancer. Breast Cancer, 1999). 6:298–300., DOI 10.1007/bf02966443 Chura JC, Blomquist CH, Ryu HS, Argenta PA Estrone sulfatase activity in patients with advanced ovarian cancer. Gynecol Oncol, 2009). 112(1):205–9., DOI 10.1016/j.ygyno.2008.08.037 Miki Y, Nakata T, Suzuki T, Darnel AD, Moriya T, Kaneko C, et al. Systemic distribution of steroid sulfatase and estrogen sulfotransferase in human adult and fetal tissues. J Clin Endocrinol Metab, 2002). 87(12):5760–8., DOI 10.1210/ jc.2002-020670 Suzuki T, Miki Y, Nakata T, Shiotsu Y, Akinaga S, Inoue K, et al. Steroid sulfatase and estrogen sulfotransferase in normal human tissue and breast carcinoma. J Steroid Biochem Mol Biol, 2003). 86(3–5):449–54., DOI 10.1016/ s0960-0760(03)00356-x Noronha RFX, Rao BR Sex hormone receptors in localized and advanced transitional cell carcinoma of urinary tract in humans. Urology, 1986). 28(5): 401–3., DOI 10.1016/0090-4295(86)90073-7 Shen SS, Smith CL, Hsieh J-T, Yu J, Kim IY, Jian W, et al. Expression of estrogen receptors-α and -β in bladder cancer cell lines and human bladder tumor tissue. Cancer, 2006). 106(12):2610–6., DOI 10.1002/cncr.21945 Sonpavde G, Okuno N, Weiss H, Yu J, Shen SS, Younes M, et al. Efficacy of selective estrogen receptor modulators in nude mice bearing human transitional cell carcinoma. Urology, 2007). 69(6):1221–6., DOI 10.1016/j. urology.2007.02.041 Miki Y, Suzuki T, Tazawa C, Yamaguchi Y, Kitada K, Honma S, et al. Aromatase localization in human breast cancer tissues: possible interactions between intratumoral stromal and parenchymal cells. Cancer Res, 2007). 67(8):3945–54., DOI 10.1158/0008-5472.can-06-3105 Aasmundstad TA, Haugen OA, Johannesen E, Hoe AL, Kvinnsland S Oestrogen receptor analysis: correlation between enzyme immunoassay and immunohistochemical methods. J Clin Pathol, 1992). 45:125–9., DOI 10.1136/ jcp.45.2.125 Kaufmann O, Baume H, Dietel M Detection of oestrogen receptors in noninvasive and invasive transitional cell carcinomas of the urinary bladder using both conventional immunohistochemistry and the tyramide staining amplification, TSA, technique. J Pathol, 1998). 186(2):165–8., DOI 10.1002/, sici)1096-9896(1998100)186:2<165::aid-path155>3.0.co;2-y Miyamoto H, Yao JL, Chaux A, Zheng Y, Hsu I, Izumi K, et al. Expression of androgen and oestrogen receptors and its prognostic significance in urothelial neoplasm of the urinary bladder. BJU Int, 2012). 109(11):1716–26., DOI 10. 1111/j.1464-410x.2011.10706.x Nam JK, Park SW, Lee SD, Chung MK Prognostic value of sex-hormone receptor expression in non-muscle-invasive bladder cancer. Yonsei Med J, 2014). 55(5):1214., DOI 10.3349/ymj.2014.55.5.1214 Larocca LM, Giustacchini M, Maggiano N, Ranelletti FO, Piantelli M, Alcini E, et al. Growth-inhibitory effect of quercetin and presence of type II estrogen binding sites in primary human transitional cell carcinomas. J Urol, 1994). 152(3):1029–33., DOI 10.1016/s0022-5347(17)32649-6 Kontos S, Kominea A, Melachrinou M, Balampani E, Sotiropoulou-Bonikou G Inverse expression of estrogen receptor-β and nuclear factor-κB in urinary bladder carcinogenesis. Int J Urol, 2010). 17(9):801–9., DOI 10.1111/j.1442- 2042.2010.02603.x Han B, Cui D, Jing Y, Hong Y, Xia S Estrogen receptor β, ERβ, is a novel prognostic marker of recurrence survival in non-muscle-invasive bladder cancer potentially by inhibiting cadherin switch. World J Urol, 2012). 30(11):861–7., DOI 10.1007/s00345-011-0819-4 Mashhadi R, Pourmand G, Kosari F, Mehrsai A, SalemS,PourmandMR, et al.Role of steroid hormone receptors in formation and progression of bladder carcinoma: a case-control study. Urol J, 2014). 11(06):1968–73., DOI 10.22037/uj.v11i06.2617 Hsieh T-S Tsong-chang tsai, ann-lii cheng, chang-yao hsieh, ih-jen su, and ming-kuen Lai.Tamoxifen enhances the chemosensitivity of bladder carcinoma cells. J Urol, 1995). 154(2):601–5., DOI 10.1097/00005392-199508000-00078 Kim HT, Kim BC, Kim IY, Mamura M, Seong DH, Jang J-J, et al. Raloxifene, a mixed estrogen agonist/antagonist, induces apoptosis through cleavage of BAD in TSU-PR1 human cancer cells. J Biol Chem, 2002). 277(36): 32510–5., DOI 10.1074/jbc.m202852200 Hoffman KL, Lerner SP, Smith CL Raloxifene inhibits growth of RT4 urothelial carcinoma cells via estrogen receptor-dependent induction of apoptosis and inhibition of proliferation. Horm Cancer, 2012). 4(1):24–35., DOI 10.1007/ s12672-012-0123-9 Kiemeney LALM, Coebergh JWW, Koper NP, Van Der Heijden LH, Pauwels RPE, Schapers RFM, et al. Bladder cancer incidence and survival in the southeastern part of The Netherlands, 1975-1989. Eur J Cancer, 1994). 30(8): 1134–7., DOI 10.1016/0959-8049(94)90472-3 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 589649 EP 589655 DI 10.3389/pore.2021.589649 PG 7 ER PT J AU Yuan, W Yan, J Liu, H Li, L Wu, B Guo, C Zhang, M AF Yuan, Wei Yan, Jiaqin Liu, Hongtao Li, Ling Wu, BoWen Guo, Can Zhang, Mingzhi TI Identification of Prognostic Related Genes of Tumor Microenvironment Derived From Esophageal Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal cancer; ESTIMATE score; stromal score; immune score; the tumor microenvironment ID Esophageal cancer; ESTIMATE score; stromal score; immune score; the tumor microenvironment AB Background and Objective: Esophageal cancer (ESCA) is a commonly occurring cancer worldwide with poor survival and limited therapeutic options. Due to the lack of biomarkers that facilitate early detection, its treatment remains a great challenge. This study aims at identifying the tumor microenvironment (TME)-related genes, which might affect prognosis and accelerate clinical treatment for ESCA patients. Methods: We integrated the expression profiles from ESCA patients in The Cancer Genome Atlas. Then, we determined the stromal and immune scores of each sample using the R package. The Gene Expression Omnibus database was used to validate the expression profile of the key genes. Results: Tumor mutational burden showed a significant difference between the groups of ESCA patients with high and low ESTIMATE scores. We identified 859 intersection genes among patients with different immune and stromal scores. Moreover, gene ontology analysis demonstrated that these 859 intersection genes were closely related to adaptive immune response and regulation of lymphocyte activation. Kyoto Encyclopedia of Genes and Genomes showed the enrichment of cytokine-cytokine receptor interaction and chemokine signaling pathway in the TME. Furthermore, the protein–protein interaction network consisted of 175 nodes. We selected 35 hub genes, including ITGAM, CXCL10, CCR2, CCR5, and CCR1. Of these, 23 intersection genes predicted the overall survival rate. C1QA and FCER1G correlated with overall survival of the ESCA patients in the two databases. Conclusion: We identified a set of stromal and immune score-related prognostic differentially expressed genes that could influence the complexity of the TME. C1QA and FCER1G were identified and validated with respect to their role in the progression of ESCA. C1 [Yuan, Wei] First Affiliated Hospital of Zhengzhou University, Department of OncologyZhengzhou, China. [Yan, Jiaqin] First Affiliated Hospital of Zhengzhou University, Department of OncologyZhengzhou, China. [Liu, Hongtao] Zhengzhou University, College of Life SciencesZhengzhou, China. [Li, Ling] First Affiliated Hospital of Zhengzhou University, Department of OncologyZhengzhou, China. [Wu, BoWen] First Affiliated Hospital of Zhengzhou University, Department of OncologyZhengzhou, China. [Guo, Can] First Affiliated Hospital of Zhengzhou University, Department of OncologyZhengzhou, China. [Zhang, Mingzhi] First Affiliated Hospital of Zhengzhou University, Department of OncologyZhengzhou, China. RP Zhang, M (reprint author), First Affiliated Hospital of Zhengzhou University, Department of Oncology, Zhengzhou, China. EM mingzhi_zhang1@163.com CR Alsop BR, Sharma P. Esophageal cancer. Gastroenterol Clin North America, 2016, 45(3):399–412., DOI 10.1016/j.gtc.2016.04.001 Arnal MJD, Ferrandez AA, Arbeloa A L. Esophageal cancer: risk factors, screening and endoscopic treatment in Western and Eastern countries. Wjg, 2015, 21(26):7933–43., DOI 10.3748/wjg.v21.i26.7933 Huang F-L, Yu S-J. Esophageal cancer: risk factors, genetic association, and treatment. Asian J Surg, 2018, 41(3):210–5., DOI 10.1016/j.asjsur.2016.10.005 Short MW, Burgers KG, Fry VT. Esophageal cancer. Am Fam Physician, 2017, 95(1):22–8. Huang T-X, Fu L. The immune landscape of esophageal cancer. Cancer Commun, 2019, 39(1):79., DOI 10.1186/s40880-019-0427-z Alsina M, Moehler M, Lorenzen S. Immunotherapy of esophageal cancer: current status, many trials and innovative strategies. Oncol Res Treat, 2018, 41(5):266–71., DOI 10.1159/000488120 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA: a Cancer J clinicians, 2017, 67(1):7–30., DOI 10.3322/caac.21387 Lu Y-f.ew, Yu J-r., Yang Z, Zhu G-x., Gao P, Wang H, et al. Promoter hypomethylation mediated upregulation of MicroRNA-10b-3p targets FOXO3 to promote the progression of esophageal squamous cell carcinoma, ESCC). J Exp Clin Cancer Res, 2018, 37(1):301., DOI 10.1186/s13046-018- 66-1 9. McAllister SS, Weinberg RA. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cel Biol, 2014, 16(8):717–27., DOI 10.1038/ncb3015 Yin Q, Han T, Fang B, Zhang G, Zhang C, Roberts ER, et al. K27-linked ubiquitination of BRAF by ITCH engages cytokine response to maintain MEKERK signaling. Nat Commun, 2019, 10(1):1870., DOI 10.1038/s41467-019- 09844-0 Tanaka K, Miyata H, Sugimura K, Fukuda S, Kanemura T, Yamashita K, et al. miR-27 is associated with chemoresistance in esophageal cancer through transformation of normal fibroblasts to cancer-associated fibroblasts. Carcin, 2015, 36(8):894–903., DOI 10.1093/carcin/bgv067 Karakasheva TA, Lin EW, Tang Q, Qiao E, Waldron TJ, Soni M, et al. IL-6 mediates cross-talk between tumor cells and activated fibroblasts in the tumor microenvironment. Cancer Res, 2018, 78(17):4957–70., DOI 10.1158/0008- 5472.Can-17-2268 Kato T, Noma K, Ohara T, Kashima H, Katsura Y, Sato H, et al. Cancerassociated fibroblasts affect intratumoral CD8+ and FoxP3+ T cells via IL6 in the tumor microenvironment. Clin Cancer Res, 2018, 24(19):4820–33., DOI 10. 1158/1078-0432.Ccr-18-0205 Kashima H, Noma K, Ohara T, Kato T, Katsura Y, Komoto S, et al. Cancerassociated fibroblasts, CAFs, promote the lymph node metastasis of esophageal squamous cell carcinoma. Int J Cancer, 2019, 144(4):828–40., DOI 10.1002/ijc.31953 Lin EW, Karakasheva TA, Hicks PD, Bass AJ, Rustgi AK. The tumor microenvironment in esophageal cancer. Oncogene, 2016, 35(41):5337–49., DOI 10.1038/onc.2016.34 Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, KimH, Torres-Garcia W, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun, 2013, 4:2612., DOI 10.1038/ncomms3612 Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther, 2017, 16(11): 2598–608., DOI 10.1158/1535-7163.Mct-17-0386 Mayakonda A, Lin D-C, Assenov Y, Plass C, Koeffler HP. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res, 2018, 28(11):1747–56., DOI 10.1101/gr.239244.118 Yu G, Wang L-G, Han Y, He Q-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS: A J Integr Biol, 2012, 16(5): 284–7., DOI 10.1089/omi.2011.0118 Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res, 2019, 47(D1):D607–d613., DOI 10.1093/nar/gky1131 Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol, 2019, 30(1):44–56., DOI 10.1093/ annonc/mdy495 Wang X, Li M. Correlate tumor mutation burden with immune signatures in human cancers. BMC Immunol, 2019, 20(1):4., DOI 10.1186/s12865-018- 0285-5 Taube JM, Galon J, Sholl LM, Rodig SJ, Cottrell TR, Giraldo NA, et al. Implications of the tumor immune microenvironment for staging and therapeutics. Mod Pathol, 2018, 31(2):214–34., DOI 10.1038/modpathol. 2017.156 Mao M, Yu Q, Huang R, Lu Y, Wang Z, Liao L. Stromal score as a prognostic factor in primary gastric cancer and close association with tumor immune microenvironment. Cancer Med, 2020, 9:4980., DOI 10.1002/cam4.2801 Pan X-B, Lu Y, Huang J-L, Long Y, Yao D-S. Prognostic genes in the tumor microenvironment in cervical squamous cell carcinoma. Aging, 2019, 11(22): 10154–66., DOI 10.18632/aging.102429 Sellar GC, Blake DJ, Reid KB. Characterization and organization of the genes encoding the A-, B- and C-chains of human complement subcomponent C1q. The complete derived amino acid sequence of human C1q. Biochem J, 1991, 274(Pt 2Pt 2):481–90., DOI 10.1042/bj2740481 Botto M, Dell’ Agnola C, Bygrave AE, Thompson EM, Cook HT, Petry F, et al. Homozygous C1q deficiency causes glomerulonephritis associated with multiple apoptotic bodies. Nat Genet, 1998, 19(1):56–9., DOI 10.1038/ ng0598-56 Racila E, Racila DM, Ritchie JM, Taylor C, Dahle C, Weiner GJ. The pattern of clinical breast cancer metastasis correlates with a single nucleotide polymorphism in the C1qA component of complement. Immunogenetics, 2006, 58(1):1–8., DOI 10.1007/s00251-005-0077-y Winslow S, Leandersson K, Edsjo A, Larsson C. Prognostic stromal gene signatures in breast cancer. Breast Cancer Res, 2015, 17(1):23., DOI 10.1186/ s13058-015-0530-2 Bulla R, Tripodo C, Rami D, Ling GS, Agostinis C, Guarnotta C, et al. C1q acts in the tumour microenvironment as a cancer-promoting factor independently of complement activation. Nat Commun, 2016, 7:10346., DOI 10.1038/ ncomms10346 Duggan SP, Garry C, Behan FM, Phipps S, Kudo H, Kirca M, et al. siRNA library screening identifies a druggable immune-signature driving esophageal adenocarcinoma cell growth. Cell Mol Gastroenterol Hepatol, 2018, 5(4): 569–90., DOI 10.1016/j.jcmgh.2018.01.012 Blank U, Jouvin M-H, Guerin-Marchand C, Kinet J-P. Le recepteur de forte affinite pour l’IgE : lecons d’une analyse structurale. Med Sci, Paris,, 2003, 19(1):63–9., DOI 10.1051/medsci/200319163 Chen L, Yuan L, Wang Y, Wang G, Zhu Y, Cao R, et al. Co-expression network analysis identified FCER1G in association with progression and prognosis in human clear cell renal cell carcinoma. Int J Biol Sci, 2017, 13(11):1361–72., DOI 10.7150/ijbs.21657 Guo T, Ma H, Zhou Y. Bioinformatics analysis of microarray data to identify the candidate biomarkers of lung adenocarcinoma. PeerJ, 2019, 7:e7313., DOI 10.7717/peerj.7313 Zhao X, Hu D, Li J, Zhao G, Tang W, Cheng H. Database mining of genes of prognostic value for the prostate adenocarcinomamicroenvironment using the cancer gene Atlas. Biomed Research International, 2020, 2020:1., DOI 10.1155/2020/5019793 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 589662 EP 589672 DI 10.3389/pore.2021.589662 PG 11 ER PT J AU Zhang, X Zhang, J Liu, Y Li, J Tan, J Song, Z AF Zhang, Xi Zhang, Junjun Liu, Yang Li, Jie Tan, Juan Song, Zewen TI Bcl-2 Associated Athanogene 2 (BAG2) is Associated With Progression and Prognosis of Hepatocellular Carcinoma: A Bioinformatics-Based Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE BAG2; hepatocellar carcinoma; ribosome biogenesis; invasion; apoptosis ID BAG2; hepatocellar carcinoma; ribosome biogenesis; invasion; apoptosis AB Background: Bcl-2 associated athanogene2 (BAG2) is reported to act as an oncogene or a tumor-suppressor in tumors in a context-dependent way; however, its function in hepatocellular carcinoma (HCC) remains unclear. Methods: Immunohistochemistry (IHC) staining, cell counting kit-8 (CCK-8) assay, apoptotic assay, cell invasion assay and a set of bioinformatics tools were integrated to analyze the role of BAG2 in hepatocellular carcinoma. Results: BAG2 was significantly up-regulated in HCC. Prognostic analysis indicated that HCC patients with high expression of BAG2 had significantly shorter overall survival, progression free survival and disease specific survival. Besides, silencing BAG2 in HCC cells impaired cell proliferation, facilitated apoptosis and repressed invasion of the cells. Bioinformatics analysis showed that BAG2 might regulate ribosome biogenesis in HCC. Conclusion: This study revealed that the up-regulated BAG2 in HCC was associated with a worse prognosis and might favor the progression of the disease. C1 [Zhang, Xi] The Third Xiangya Hospital of Central South University, Department of OncologyChangsha, China. [Zhang, Junjun] The Third Xiangya Hospital of Central South University, Department of OncologyChangsha, China. [Liu, Yang] Central South University, Third Xiangya Hospital, Department of PathologyChangsha, China. [Li, Jie] Hunan University of Chinese Medicine, Department of Information Science and EngineeringChangsha, China. [Tan, Juan] Central South University, Third Xiangya Hospital, Department of PathologyChangsha, China. [Song, Zewen] The Third Xiangya Hospital of Central South University, Department of OncologyChangsha, China. RP Song, Z (reprint author), The Third Xiangya Hospital of Central South University, Department of Oncology, Changsha, China. CR Pelletier J, Thomas G, Volarevi´c S. Ribosome biogenesis in cancer: new players and therapeutic avenues. Nat Rev Cancer, 2018, 18(1):51–63., DOI 10.1038/nrc. 2017.104 PenzoM,Montanaro L, Trere D, DerenziniM. The ribosome biogenesis-cancer connection. Cells, 2019, 8(1):55., DOI 10.3390/cells8010055 Prakash V, Carson BB, Feenstra JM, Dass RA, Sekyrova P, Hoshino A, et al. Ribosome biogenesis during cell cycle arrest fuels EMT in development and disease. Nat Commun, 2019, 10(1):2110., DOI 10.1038/s41467-019-10100-8 Khot A, Brajanovski N, Cameron DP, Hein N, Maclachlan KH, Sanij E, et al. First-in-Human RNA polymerase I transcription inhibitor CX-5461 in patients with advanced hematologic cancers: results of a phase I dose-escalation study. Cancer Discov, 2019, 9(8):1036–49., DOI 10.1158/2159-8290.cd-18-1455 Mariotto E, Viola G, Zanon C, Aveic S. A BAG’s life: every connection matters in cancer. Pharmacol Ther, 2020, 209:107498., DOI 10.1016/j.pharmthera.2020. 107498 Lee J-H, Takahashi T, Yasuhara N, Inazawa J, Kamada S, Tsujimoto Y. Bis, a Bcl-2-binding protein that synergizes with Bcl-2 in preventing cell death. Oncogene, 1999, 18(46):6183–90., DOI 10.1038/sj.onc.1203043 Sun L, Chen G, Sun A, Wang Z, Huang H, Gao Z, et al. BAG2 promotes proliferation and metastasis of gastric cancer via ERK1/2 signaling and partially regulated by miR186. Front Oncol, 2020, 10:31., DOI 10.3389/fonc.2020.00031 Yue X, Zhao Y, Liu J, Zhang C, Yu H, Wang J, et al. BAG2 promotes tumorigenesis through enhancing mutant p53 protein levels and function. Elife, 2015, 4:e08401., DOI 10.7554/elife.08401 Yang K-M, Bae E, Ahn SG, Pang K, Park Y, Park J, et al. Co-chaperone BAG2 determines the pro-oncogenic role of cathepsin B in triple-negative breast cancer cells. Cel Rep, 2017, 21(10):2952–64., DOI 10.1016/j.celrep.2017.11.026 Liu Y-S, Wei B. Over-expression of Bcl2-associated athanogene 2 in oral cancer promotes cellular proliferation and is associated with poor prognosis. Arch Oral Biol, 2019, 102:164–70., DOI 10.1016/j.archoralbio.2019.04.015 Wang H-Q, Zhang H-Y, Hao F-J, Meng X, Guan Y, Du Z-X. Induction of BAG2 protein during proteasome inhibitor-induced apoptosis in thyroid carcinoma cells. Br J Pharmacol, 2008, 155(5):655–60., DOI 10.1038/bjp. 2008.302 Song Z, Yu Z, Chen L, Zhou Z, Zou Q, Liu Y. MicroRNA-1181 supports the growth of hepatocellular carcinoma by repressing AXIN1. Biomed Pharmacother, 2019, 119:109397., DOI 10.1016/j.biopha.2019.109397 Song Z, Fusco J, Zimmerman R, Fischbach S, Chen C, Ricks DM, et al. Epidermal growth factor receptor signaling regulates β cell proliferation in adult mice. J Biol Chem, 2016, 291(43):22630–7., DOI 10.1074/jbc.m116. 747840 Huang J-Z, Chen M, Chen D, Gao X-C, Zhu S, Huang H, et al. A peptide encoded by a putative lncRNA HOXB-AS3 suppresses colon cancer growth. Mol Cel, 2017, 68(1):171–84., DOI 10.1016/j.molcel.2017.09.015 Yu G, Wang L-G, Han Y, He Q-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS: A J Integr Biol, 2012, 16(5): 284–7., DOI 10.1089/omi.2011.0118 Camp RL, Dolled-Filhart M, Rimm DL. X-Tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res, 2004, 10(21):7252–9., DOI 10.1158/1078-0432.ccr-04-0713 Adachi K, Soeta-Saneyoshi C, Sagara H, Iwakura Y. Crucial role of Bysl in mammalian preimplantation development as an integral factor for 40S ribosome biogenesis. Mcb, 2007, 27(6):2202–14., DOI 10.1128/mcb.01908-06 Morello LG, Coltri PP, Quaresma AJC., Simabuco FM, Silva TCL, Singh G, et al. The human nucleolar protein FTSJ3 associates with NIP7 and functions in pre-rRNA processing. PLoS One, 2011, 6(12):e29174., DOI 10.1371/journal. pone.0029174 Zhang J, Zhang Y-L, Zhao L-W, Guo J-X, Yu J-L, Ji S-Y, et al. Mammalian nucleolar protein DCAF13 is essential for ovarian follicle maintenance and oocyte growth by mediating rRNA processing. Cell Death Differ, 2019, 26(7): 1251–66., DOI 10.1038/s41418-018-0203-7 Holzel M, Rohrmoser M, Schlee M, Grimm T, HarasimT,Malamoussi A, et al. Mammalian WDR12 is a novel member of the Pes1-Bop1 complex and is required for ribosome biogenesis and cell proliferation. J Cel Biol, 2005, 170(3):367–78., DOI 10.1083/jcb.200501141 Talkish J, Campbell IW, Sahasranaman A, Jakovljevic J, Woolford JL. Ribosome assembly factors Pwp1 and Nop12 are important for folding of 5.8S rRNA during ribosome biogenesis in Saccharomyces cerevisiae. Mol Cell Biol, 2014, 34(10):1863–77., DOI 10.1128/mcb.01322-13 Pan W-A, Tsai H-Y, Wang S-C, Hsiao M, Wu P-Y, Tsai M-D. The RNA recognition motif of NIFK is required for rRNA maturation during cell cycle progression. RNA Biol, 2015, 12(3):255–67., DOI 10.1080/15476286.2015.1017221 Asano N, Kato K, Nakamura A, Komoda K, Tanaka I, Yao M. Structural and functional analysis of the Rpf2-Rrs1 complex in ribosome biogenesis. Nucleic Acids Res, 2015, 43(9):4746–57., DOI 10.1093/nar/gkv305 Assoun S, Brosseau S, Steinmetz C, Gounant V, Zalcman G. Bevacizumab in advanced lung cancer: state of the art. Future Oncol, 2017, 13(28):2515–35., DOI 10.2217/fon-2017-0302 Knispel S, Zimmer L, Kanaki T, Ugurel S, Schadendorf D, Livingstone E. The safety and efficacy of dabrafenib and trametinib for the treatment of melanoma. Expert Opin Drug Saf, 2018, 17(1):73–87., DOI 10.1080/ 14740338.2018.1390562 Fornasier G, Francescon S, Baldo P. An update of efficacy and safety of cetuximab in metastatic colorectal cancer: a narrative review. Adv Ther, 2018, 35(10):1497–509., DOI 10.1007/s12325-018-0791-0 Shields S, Conroy E, O’Grady T, McGoldrick A, Connor K, Ward MP, et al. BAG3 promotes tumour cell proliferation by regulating EGFR signal transduction pathways in triple negative breast cancer. Oncotarget, 2018, 9(21):15673–90., DOI 10.18632/oncotarget.24590 Lv J, Zhang F, Zhai C, Wang G, Qu Y. Bag-1 silence sensitizes non-small cell lung cancer cells to cisplatin through multiple gene pathways. Ott, 2019, 12: 8977–89., DOI 10.2147/ott.s218182 Kilbas PO, Akcay IM, Doganay GD, Arisan ED. Bag-1 silencing enhanced chemotherapeutic drug-induced apoptosis in MCF-7 breast cancer cells affecting PI3K/Akt/mTOR and MAPK signaling pathways. Mol Biol Rep, 2019, 46(1):847–60., DOI 10.1007/s11033-018-4540-x Kizilboga T, Baskale EA, Yildiz J, Akcay IM, Zemheri E, Can ND, et al. Bag-1 stimulates bad phosphorylation through activation of Akt and Raf kinases to mediate cell survival in breast cancer. BMC Cancer, 2019, 19(1):1254., DOI 10. 1186/s12885-019-6477-4 Bruchmann A, Roller C, Walther TV, Schafer G, Lehmusvaara S, Visakorpi T, et al. Bcl-2 associated athanogene 5, Bag5, is overexpressed in prostate cancer and inhibits ER-stress induced apoptosis. BMC Cancer, 2013, 13:96., DOI 10.1186/1471-2407-13-96 Ni W, Chen B, Zhou G, Lu C, Xiao M, Guan C, et al. Overexpressed nuclear BAG-1 in human hepatocellular carcinoma is associated with poor prognosis and resistance to doxorubicin. J Cel Biochem., 2013, 114(9):2120–30., DOI 10. 1002/jcb.24560 Kong D-H, Li S, Du Z-X, Liu C, Liu B-Q, Li C, et al. BAG3 elevation inhibits cell proliferation via direct interaction with G6PD in hepatocellular carcinomas. Oncotarget, 2016, 7(1):700–11., DOI 10.18632/oncotarget.6396 Xiao H, Cheng S, Tong R, Lv Z, Ding C, Du C, et al. BAG3 regulates epithelialmesenchymal transition and angiogenesis in human hepatocellular carcinoma. Lab Invest, 2014, 94(3):252–61., DOI 10.1038/labinvest.2013.151 Catez F, Dalla Venezia N, Marcel V, Zorbas C, Lafontaine DLJ, Diaz J-J. Ribosome biogenesis: an emerging druggable pathway for cancer therapeutics. Biochem Pharmacol, 2019, 159:74–81., DOI 10.1016/j.bcp.2018.11.014 Derenzini M, Montanaro L, Trere D. Ribosome biogenesis and cancer. Acta Histochem, 2017, 119(3):190–7., DOI 10.1016/j.acthis.2017.01.009 Sharifi S, Bierhoff H. Regulation of RNA polymerase I transcription in development, disease, and aging. Annu Rev Biochem, 2018, 87:51–73., DOI 10.1146/annurev-biochem-062917-012612 Ahuja R, Kumar V. Stimulation of Pol III-dependent 5S rRNA and U6 snRNA gene expression by AP-1 transcription factors. FEBS J, 2017, 284(13):2066–77., DOI 10.1111/febs.14104 LiaoP,Zeng SX, ZhouX, ChenT, Zhou F, CaoB, et al. Mutant p53 gains its function via c-myc activation upon CDK4 phosphorylation at serine 249 and consequent PIN1 binding. Mol Cel, 2017, 68(6):1134–46., DOI 10.1016/j.molcel.2017.11.006 Calderaro J, Couchy G, Imbeaud S, Amaddeo G, Letouze E, Blanc J-F, et al. Histological subtypes of hepatocellular carcinoma are related to gene mutations and molecular tumour classification. J Hepatol, 2017, 67(4): 727–38., DOI 10.1016/j.jhep.2017.05.014 Li Q, Liu X, Jin K, Lu M, Zhang C, Du X, et al. NAT10 is upregulated in hepatocellular carcinoma and enhances mutant p53 activity. BMC Cancer, 2017, 17(1):605., DOI 10.1186/s12885-017-3570-4 Piazzi M, Bavelloni A, Gallo A, Faenza I, Blalock WL. Signal transduction in ribosome biogenesis: a recipe to avoid disaster. Int J Mol Sci, 2019, 20(11): 2718., DOI 10.3390/ijms20112718 Qin L, Guo J, Zheng Q, ZhangH. BAG2 structure, function and involvement in disease. Cell Mol Biol Lett, 2016, 21:18., DOI 10.1186/s11658-016-0020-2 Komarova EY, Afanasyeva EA, Bulatova MM, Cheetham ME, Margulis BA, Guzhova IV. Downstream caspases are novel targets for the antiapoptotic activity of the molecular chaperone hsp70. Cell Stress Chaper, 2004, 9(3): 265–75., DOI 10.1379/csc-27r1.1 Sverchinsky DV, Nikotina AD, Komarova EY, Mikhaylova ER, Aksenov ND, Lazarev VF, et al. Etoposide-induced apoptosis in cancer cells can be reinforced by an uncoupled link between Hsp70 and caspase-3. Int J Mol Sci, 2018, 19(9): 2519., DOI 10.3390/ijms19092519 Kroemer G. Heat shock protein 70 neutralizes apoptosis-inducing factor. The Scientific World JOURNAL, 2001, 1:590–2., DOI 10.1100/tsw.2001.322 Gotoh T, Terada K, Oyadomari S, Mori M. hsp70-DnaJ chaperone pair prevents nitric oxide- and CHOP-induced apoptosis by inhibiting translocation of Bax to mitochondria. Cel Death Differ, 2004, 11(4): 390–402., DOI 10.1038/sj.cdd.4401369 Saleh A, Srinivasula SM, Balkir L, Robbins PD, Alnemri ES. Negative regulation of the apaf-1 apoptosome by Hsp70. Nat Cel Biol, 2000, 2(8): 476–83., DOI 10.1038/35019510 Bruckheimer EM, Cho SH, Sarkiss M, Herrmann J, McDonnell TJ. The Bcl-2 gene family and apoptosis. Adv Biochem Eng Biotechnol, 1998, 62:75–105., DOI 10.1007/bfb0102306 Nadler Y, Camp RL, Giltnane JM, Moeder C, Rimm DL, Kluger HM, et al. Expression patterns and prognostic value of Bag-1 and Bcl-2 in breast cancer. Breast Cancer Res, 2008, 10(2):R35., DOI 10.1186/bcr1998 Xue C, Wang K, Jiang X, Gu C, Yu G, Zhong Y, et al. The down-regulation of SUZ12 accelerates the migration and invasion of liver cancer cells via activating ERK1/2 pathway. J Cancer, 2019, 10(6):1375–84., DOI 10.7150/jca. 29932 Gao X, Shan W, Liu X, Zhang J, Zheng J, Yao H. JNK1/2 and ERK1/2 provides vital clues about tumor recurrence and survival in hepatocellular carcinoma patients. Future Oncol, 2018, 14(24):2471–81., DOI 10.2217/ fon-2018-0171 Rhee H, Kim H-Y, Choi J-H, Woo HG, Yoo JE, Nahm JH, et al. Keratin 19 expression in hepatocellular carcinoma is regulated by fibroblast-derived HGF via a MET-ERK1/2-AP1 and SP1 axis. Cancer Res, 2018, 78(7):1619–31., DOI 10.1158/0008-5472.can-17-0988 Xu Z-Z, Xiu P, Lv J-W, Wang F-H, Dong X-F, Liu F, et al. Integrin αvβ3 is required for cathepsin B-induced hepatocellular carcinoma progression. Mol Med Rep, 2015, 11(5):3499–504., DOI 10.3892/mmr.2014.3140 Varshosaz J, Fard MM, Mirian M, Hassanzadeh F. Targeted nanoparticles for Co-delivery of 5-FU and nitroxoline, a cathepsin B inhibitor, in HepG2 cells of hepatocellular carcinoma. Anticancer Agents Med Chem, 2020, 20(3):346–358., DOI 10.2174/1871520619666190930124746 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 594649 EP 594661 DI 10.3389/pore.2021.594649 PG 13 ER PT J AU Du, H Luo, F Shi, M Che, J Zhu, L Li, H Hang, J AF Du, Hailei Luo, Fangxiu Shi, Minmin Che, Jiaming Zhu, Lianggang Li, Hecheng Hang, Junbiao TI Beclin-1 is a Promising Prognostic Biomarker in a Specific Esophageal Squamous Cell Carcinoma Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE esophageal squamous cell carcinoma (ESCC); autophagy; apoptosis; beclin-1; Prognosis ID esophageal squamous cell carcinoma (ESCC); autophagy; apoptosis; beclin-1; Prognosis AB The effects of autophagy and apoptosis in the prognostic assessment and treatment of Esophageal squamous cell carcinoma (ESCC) remain to be elucidated. Here, we conducted a retrospective study on the histopathology of ESCC, investigated the expression of Beclin-1 and Bcl-2 proteins (both autophagy- and apoptosis-related) in esophageal cancer tissue, and analyzed the significance of these proteins for the prognosis of ESCC. In the present study, the expression level of Beclin-1 in ESCC was significantly lower than that in adjacent tissues (p < 0.01), whereas the expression level of Bcl-2 showed the opposite pattern (p < 0.01). Furthermore, low expression of Beclin-1 was associated with more advanced ESCC stages and lymph node metastasis. However, high expression of Bcl-2 was associated with more advanced ESCC stages, deeper tumor invasion, and lymph node metastasis. Moreover, the relationship between Bcl-2 expression and OS was not significant (p > 0.05), whereas Beclin-1 expression was significantly associated with OS (p < 0.05). Subgroup analysis showed that Beclin-1 expression was significantly associated with OS in the high-Bcl-2-expression group but not in the low-Bcl-2-expression group. Importantly, Beclin-1 upregulation or downregulation significantly upregulated or downregulated invasion, respectively, in EC9706 cells in combination with high expression but not low expression of Bcl-2. These findings reveal that differences in autophagy and apoptotic states and their activities may promote malignant tumor differentiation, which could lead to a more aggressive esophageal squamous cell phenotype and a worse survival prognosis. Here, Beclin-1 was shown to be a promising prognostic biomarker and therapeutic target for patients with ESCC in the high-Bcl-2-expression population. C1 [Du, Hailei] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Thoracic SurgeryShanghai, China. [Luo, Fangxiu] Shanghai Jiao Tong University School ofMedicine, Ruijin Hospital North, Department of PathologyShanghai, China. [Shi, Minmin] Institute of Digestive SurgeryShanghai, China. [Che, Jiaming] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Thoracic SurgeryShanghai, China. [Zhu, Lianggang] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Thoracic SurgeryShanghai, China. [Li, Hecheng] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Thoracic SurgeryShanghai, China. [Hang, Junbiao] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Thoracic SurgeryShanghai, China. RP Hang, J (reprint author), Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of Thoracic Surgery, Shanghai, China. EM drhangjb@hotmail.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J Clin, 2018). 68(6):394–424., DOI 10. 3322/caac.21492 Siegel RL, Miller KD, Jemal A Cancer statistics, 2016. CA: A Cancer J Clin, 2016). 66(1):7–30., DOI 10.3322/caac.21332 Torre LA, Siegel RL, Ward EM, Jemal A Global cancer incidence and mortality rates and trends-an update. Cancer Epidemiol Biomarkers Prev, 2016). 25(1): 16–27., DOI 10.1158/1055-9965.epi-15-0578 Yu L, Chen Y, Tooze SA Autophagy pathway: cellular and molecular mechanisms. Autophagy, 2018). 14(2):207–15., DOI 10.1080/15548627.2017. 1378838 Saha S, Panigrahi DP, Patil S, Bhutia SK Autophagy in health and disease: a comprehensive review. Biomed Pharmacother, 2018). 104:485–95., DOI 10.1016/ j.biopha.2018.05.007 Li Q, Liu Y, Sun M Autophagy and alzheimer’s disease. Cell Mol Neurobiol, 2017). 37(3):377–88., DOI 10.1007/s10571-016-0386-8 Mialet-Perez J, Vindis C Autophagy in health and disease: focus on the cardiovascular system. Essays Biochem, 2017). 61(6):721–32., DOI 10.1042/ ebc20170022 Hall TM, Tetreault M-P, Hamilton KE, Whelan KA Autophagy as a cytoprotective mechanism in esophageal squamous cell carcinoma. Curr Opin Pharmacol, 2018). 41:12–9., DOI 10.1016/j.coph.2018.04.003 Hamurcu Z, Delibas¸i N, Gecene S, S¸ener EF, Donmez-Altuntas¸ H, Ozkul Y, et al. Targeting LC3 and Beclin-1 autophagy genes suppresses proliferation, survival, migration and invasion by inhibition of Cyclin-D1 and uPAR/Integrin β1/Src signaling in triple negative breast cancer cells. J Cancer Res Clin Oncol, 2018). 144(3):415–30., DOI 10.1007/s00432-017-2557-5 Huang F, Wang B-R, Wang Y-G Role of autophagy in tumorigenesis, metastasis, targeted therapy and drug resistance of hepatocellular carcinoma. World J Gastroenterol, 2018). 24(41):4643–51., DOI 10.3748/wjg. v24.i41.4643 Shen H, Yin L, Deng G, Guo C, Han Y, Li Y, et al. Knockdown of Beclin-1 impairs epithelial-mesenchymal transition of colon cancer cells. J Cel Biochem, 2018). 119(8):7022–31., DOI 10.1002/jcb.26912 Sohn EJ, Park HT Natural agents mediated autophagic signal networks in cancer. Cancer Cel Int, 2017). 17:110., DOI 10.1186/ s12935-017-0486-7 Zhang XF, Yang Y, Zhang J, Cao W Microvesicle-containing miRNA-153-3p induces the apoptosis of proximal tubular epithelial cells and participates in renal interstitial fibrosis. Eur Rev Med Pharmacol Sci, 2019). 23(22):10065–71., DOI 10.26355/eurrev_201911_19574 Zeng H, Zheng R, Zhang S, Zuo T, Xia C, Zou X, et al. Esophageal cancer statistics in China, 2011: estimates based on 177 cancer registries. Thorac Cancer, 2016). 7(2):232–7., DOI 10.1111/1759-7714.12322 Malhotra GK, Yanala U, Ravipati A, Follet M, Vijayakumar M, Are C Global trends in esophageal cancer. J Surg Oncol, 2017). 115(5):564–79., DOI 10.1002/jso.24592 Pakzad R, Mohammadian-Hafshejani A, Khosravi B, Soltani S, Pakzad I, Mohammadian M, et al. The incidence and mortality of esophageal cancer and their relationship to development in Asia. Ann Transl Med, 2016). 4(2):29., DOI 10.3978/j.issn.2305- 5839.2016.01.11 Xu H-D, Qin Z-H Beclin 1, bcl-2 and autophagy. Adv Exp Med Biol, 2019). 1206:109–26., DOI 10.1007/978-981-15-0602-4_5 Feng F, Zhang M, Yang C, Heng X, Wu X The dual roles of autophagy in gliomagenesis and clinical therapy strategies based on autophagic regulation mechanisms. Biomed Pharmacother, 2019). 120:109441., DOI 10.1016/j.biopha. 2019.109441 Kasprowska-Li´skiewicz D The cell on the edge of life and death: crosstalk between autophagy and apoptosis. Postepy Hig Med Dosw, Online,, 2017). 71(0):825–41., DOI 10.5604/01.3001.0010.4672 Du H, Che J, Shi M, Zhu L, Hang JB, Chen Z, et al. Beclin 1 expression is associated with the occurrence and development of esophageal squamous cell carcinoma. Oncol Lett, 2017). 14(6):6823–8., DOI 10. 3892/ol.2017.7015 Farkas R, Pozsgai E, Bellyei S, Cseke L, Szigeti A, Vereczkei A, et al. Correlation between tumor-associated proteins and response to neoadjuvant treatment in patients with advanced squamous-cell esophageal cancer. Anticancer Res, 2011). 31(5):1769–75. Badrinath N, Yoo SY Mitochondria in cancer: in the aspects of tumorigenesis and targeted therapy. Carcinogenesis, 2018). 39(12): 1419–30., DOI 10.1093/carcin/bgy148 Hasanain M, Sahai R, Pandey P, et al. Microtubule disrupting agent-mediated inhibition of cancer cell growth is associated with blockade of autophagic flux and simultaneous induction of apoptosis. Cell Prolif, 2020). 53(4):e12749., DOI 10.1111/cpr.12749 Geng X, Ren Y, Wang F, Tian D, Yao X, Zhang Y, et al. Harmines inhibit cancer cell growth through coordinated activation of apoptosis and inhibition of autophagy. Biochem Biophysical Res Commun, 2018). 498(1):99–104., DOI 10.1016/j.bbrc.2018.02.205 Athamneh K, Alneyadi A, Alsamri H, Alrashedi A, Palakott A, El-Tarabily KA, et al. Origanum majorana essential oil triggers p38 MAPK-mediated protective autophagy, apoptosis, and caspase-dependent cleavage of P70S6K in colorectal cancer cells. Biomolecules, 2020). 10(3):412., DOI 10.3390/biom10030412 Law BYK, Michelangeli F, Qu YQ, Xu SW, Han Y, Mok SWF, et al. Neferine induces autophagy-dependent cell death in apoptosis-resistant cancers via ryanodine receptor and Ca(2+)-dependent mechanism. Sci Rep, 2019). 9(1): 20034., DOI 10.1038/s41598-019-56675-6 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 594724 EP 594732 DI 10.3389/pore.2021.594724 PG 9 ER PT J AU Meder, Ly Florin, A Ozretic, L Nill, M Koker, M Meemboor, S Radtke, F Diehl, L Ullrich, R Odenthal, M Buttner, R Heukamp, L AF Meder, Lydia Florin, Alexandra Ozretic, Luka Nill, Marieke Koker, Mirjam Meemboor, Sonja Radtke, Freddy Diehl, Linda Ullrich, T. Roland Odenthal, Margarete Buttner, Reinhard Heukamp, C. Lukas TI Notch1 Deficiency Induces Tumor Cell Accumulation Inside the Bronchiolar Lumen and Increases TAZ Expression in an Autochthonous KrasLSL-G12V Driven Lung Cancer Mouse Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mouse model; lung cancer; KRAS; NOTCH1; TAZ ID mouse model; lung cancer; KRAS; NOTCH1; TAZ AB Purpose: Abrogation of Notch signaling, which is pivotal for lung development and pulmonary epithelial cell fate decisions was shown to be involved in the aggressiveness and the differentiation of lung carcinomas. Additionally, the transcription factors YAP and TAZ which are involved in the Hippo pathway, were recently shown to be tightly linked with Notch signaling and to regulate the cell fate in epidermal stem cells. Thus, we aim to elucidate the effects of conditional Notch1 deficiency on carcinogenesis and TAZ expression in lung cancer. Methods: We investigated the effect of conditional Cre-recombinase mediated Notch1 knock-out on lung cancer cells in vivo using an autochthonous mouse model of lung adenocarcinomas driven by KrasLSL-G12V and comprehensive immunohistochemical analysis. In addition, we analyzed clinical samples and human lung cancer cell lines for TAZ expression and supported our findings by publicly available data from The Cancer Genome Atlas (TCGA). Results: In mice, we found induction of papillary adenocarcinomas and protrusions of tumor cells from the bronchiolar lining upon Notch1 deficiency. Moreover, the mutated Kras driven lung tumors with deleted Notch1 showed increased TAZ expression and focal nuclear translocation which was frequently observed in human pulmonary adenocarcinomas and squamous cell carcinomas of the lung, but not in small cell lung carcinomas. In addition, we used data from TCGA to show that putative inactivating NOTCH1 mutations co-occur with KRAS mutations and genomic amplifications in lung adenocarcinomas. Conclusion: Our in vivo study provides evidence that Notch1 deficiency in mutated Kras driven lung carcinomas contributes to lung carcinogenesis in a subgroup of patients by increasing TAZ expression who might benefit from TAZ signaling blockade. C1 [Meder, Lydia] University Hospital Cologne, Department I of Internal MedicineCologne, Germany. [Florin, Alexandra] University of Cologne Medical School, Institute of PathologyCologne, Germany. [Ozretic, Luka] Royal Free Hospital, Department of Cellular PathologyLondon, UK. [Nill, Marieke] University Hospital Cologne, Department I of Internal MedicineCologne, Germany. [Koker, Mirjam] University Hospital Cologne, Department I of Internal MedicineCologne, Germany. [Meemboor, Sonja] University of Cologne Medical School, Institute of PathologyCologne, Germany. [Radtke, Freddy] Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Federale de LausanneLausanne, Switzerland. [Diehl, Linda] University Medical Center Hamburg-Eppendorf, Institute of Experimental Immunology and HepatologyHamburg, Germany. [Ullrich, T. Roland] University Hospital Cologne, Department I of Internal MedicineCologne, Germany. [Odenthal, Margarete] University of Cologne, Center for Molecular Medicine CologneCologne, Germany. [Buttner, Reinhard] University of Cologne, Center for Molecular Medicine CologneCologne, Germany. [Heukamp, C. Lukas] Institute for Hematopathology HamburgHamburg, Germany. RP Meder, Ly (reprint author), University Hospital Cologne, Department I of Internal Medicine, Cologne, Germany. EM lydia.meder@uk-koeln.de CR Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin, 2018, 68(1):7–30., DOI 10.3322/caac.21442 Konig K, Peifer M, Fassunke J, Ihle MA, Kunstlinger H, Heydt C, et al. Implementation of amplicon parallel sequencing leads to improvement of diagnosis and therapy of lung cancer patients. J Thorac Oncol, 2015, 10(7): 1049–57., DOI 10.1097/jto.0000000000000570 Travis WD. Advances in neuroendocrine lung tumors. Ann Oncol, 2010, 21(Suppl. 7):vii65–71., DOI 10.1093/annonc/mdq380 Hou S, Zhou S, Qin Z, Yang L, Han X, Yao S, et al. Evidence, mechanism, and clinical relevance of the transdifferentiation from lung adenocarcinoma to squamous cell carcinoma. Am J Pathol, 2017, 187(5):954–62., DOI 10.1016/j. ajpath.2017.01.009 Cooke DT, Nguyen DV, Yang Y, Chen SL, Yu C, Calhoun RF. Survival comparison of adenosquamous, squamous cell, and adenocarcinoma of the lung after lobectomy. Ann Thorac Surg, 2010, 90(3):943–8., DOI 10.1016/j. athoracsur.2010.05.025 Sutherland KD, Song J-Y, Kwon MC, Proost N, Zevenhoven J, Berns A. Multiple cells-of-origin of mutant K-Ras-induced mouse lung adenocarcinoma. Proc Natl Acad Sci USA, 2014, 111(13):4952–7., DOI 10.1073/pnas.1319963111 Mainardi S, Mijimolle N, Francoz S, Vicente-Duenas C, Sanchez-Garcia I, Barbacid M. Identification of cancer initiating cells in K-Ras driven lung adenocarcinoma. Proc Natl Acad Sci, 2014, 111(1):255–60., DOI 10.1073/pnas. 1320383110 Wang NJ, Sanborn Z, Arnett KL, Bayston LJ, Liao W, Proby CM, et al. Loss-offunction mutations in Notch receptors in cutaneous and lung squamous cell carcinoma. Proc Natl Acad Sci, 2011, 108(43):17761–6., DOI 10.1073/pnas. 1114669108 Tsao P-N, Matsuoka C, Wei S-C, Sato A, Sato S, Hasegawa K, et al. Epithelial Notch signaling regulates lung alveolar morphogenesis and airway epithelial integrity. Proc Natl Acad Sci USA, 2016, 113(29):8242–7., DOI 10.1073/pnas. 1511236113 Totaro A, Castellan M, Battilana G, Zanconato F, Azzolin L, Giulitti S, et al. YAP/TAZ link cell mechanics to Notch signalling to control epidermal stem cell fate. Nat Commun, 2017, 8(1):15206., DOI 10.1038/ ncomms15206 Zhou Z, Hao Y, Liu N, Raptis L, Tsao M-S, Yang X. TAZ is a novel oncogene in non-small cell lung cancer. Oncogene, 2011, 30(18):2181–6., DOI 10.1038/onc. 2010.606 Xie M, Zhang L, He C-S, Hou J-H, Lin S-X, Hu Z-H, et al. Prognostic significance of TAZ expression in resected non-small cell lung cancer. J Thorac Oncol, 2012, 7(5):799–807., DOI 10.1097/jto.0b013e318248240b Konig K, Meder L, Kroger C, Diehl L, Florin A, Rommerscheidt-Fuss U, et al. Loss of the keratin cytoskeleton is not sufficient to induce epithelial mesenchymal transition in a novel KRAS driven sporadic lung cancer mouse model. PLoS One, 2013, 8(3):e57996., DOI 10.1371/journal.pone. 0057996 DuPage M, Dooley AL, Jacks T. Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase. Nat Protoc, 2009, 4(7): 1064–72., DOI 10.1038/nprot.2009.95 Radtke F, Wilson A, Stark G, Bauer M, van Meerwijk J, MacDonald HR, et al. Deficient T cell fate specification in mice with an induced inactivation of Notch1. Immunity, 1999, 10(5):547–58., DOI 10.1016/s1074-7613(00, 80054-0 Nikitin AY, Alcaraz A, Anver MR, Bronson RT, Cardiff RD, Dixon D, et al. Classification of proliferative pulmonary lesions of the mouse. Cancer Res, 2004, 64(7):2307–16., DOI 10.1158/0008-5472.can-03-3376 Vici P, Mottolese M, Pizzuti L, Barba M, Sperati F, Terrenato I, et al. TheHippo transducer TAZ as a biomarker of pathological complete response in HER2- positive breast cancer patients treated with trastuzumab-based neoadjuvant therapy. Oncotarget, 2014, 5(20):9619–25., DOI 10.18632/oncotarget.2449 Meder L, Konig K, Ozreti´c L, Schultheis AM, Ueckeroth F, Ade CP, et al. NOTCH , ASCL1 , p53 and RB alterations define an alternative pathway driving neuroendocrine and small cell lung carcinomas. Int J Cancer, 2016, 138(4):927–38., DOI 10.1002/ijc.29835 Yoo AS, Sun AX, Li L, Shcheglovitov A, Portmann T, Li Y, et al. MicroRNAmediated conversion of human fibroblasts to neurons. Nature, 2011, 476(7359):228–31., DOI 10.1038/nature10323 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data: figure 1. Cancer Discov, 2012, 2(5):401–4., DOI 10.1158/ 2159-8290.cd-12-0095 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signaling, 2013, 6(269):pl1., DOI 10.1126/scisignal.2004088 Cancer Genome Atlas Research N. Comprehensive molecular profiling of lung adenocarcinoma. Nature, 2014, 511(7511):543–50., DOI 10.1038/nature13385 Cancer Genome Atlas Research N. Comprehensive genomic characterization of squamous cell lung cancers. Nature, 2012, 489(7417):519–25., DOI 10.1038/ nature11404 Skibinski A, Breindel JL, Prat A, Galvan P, Smith E, Rolfs A, et al. The Hippo transducer TAZ interacts with the SWI/SNF complex to regulate breast epithelial lineage commitment. Cel Rep, 2014, 6(6):1059–72., DOI 10.1016/j. celrep.2014.02.038 Jackson EL, Willis N, Mercer K, Bronson RT, Crowley D, Montoya R, et al. Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Dev, 2001, 15(24):3243–8., DOI 10.1101/gad.943001 Wang S, Hao J, Qian C, Wang H. Tumor spread through air spaces is a survival predictor in non-small-cell lung cancer. Clin Lung Cancer, 2019, 20(5): e584–e591., DOI 10.1016/j.cllc.2019.05.012 Travis WD, Brambilla E, Noguchi M, Nicholson AG, Geisinger KR, Yatabe Y, et al. International association for the study of lung cancer/american thoracic society/european respiratory society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol, 2011, 6(2):244–85., DOI 10.1097/JTO.0b013e318206a221 Prall F. Tumour budding in colorectal carcinoma. Histopathology, 2007, 50(1): 151–62., DOI 10.1111/j.1365-2559.2006.02551.x Aoyagi Y, Yokose T, Minami Y, Ochiai A, Iijima T, Morishita Y, et al. Accumulation of losses of heterozygosity and multistep carcinogenesis in pulmonary adenocarcinoma. Cancer Res, 2001, 61(21):7950–4. Davies EJ, Marsh Durban V, Meniel V, Williams GT, Clarke AR. PTEN loss and KRAS activation leads to the formation of serrated adenomas and metastatic carcinoma in the mouse intestine. J Pathol, 2014, 233(1):27–38., DOI 10.1002/path.4312 Sakai E, Nakayama M, Oshima H, Kouyama Y, Niida A, Fujii S, et al. Combined mutation of apc, Kras, and Tgfbr2 effectively drives metastasis of intestinal cancer. Cancer Res, 2018, 78(5):1334–46., DOI 10.1158/0008-5472. can-17-3303 Lim KJ, Brandt WD, Heth JA, Muraszko KM, Fan X, Bar EE, et al. Lateral inhibition of Notch signaling in neoplastic cells. Oncotarget, 2015, 6(3): 1666–77., DOI 10.18632/oncotarget.2762 Dupont S, Morsut L, Aragona M, Enzo E, Giulitti S, Cordenonsi M, et al. Role of YAP/TAZ in mechanotransduction. Nature, 2011, 474(7350):179–83., DOI 10.1038/nature10137 Dupont S. Regulation of YAP/TAZ activity by mechanical cues: an experimental overview. Methods Mol Biol, 2019, 1893:183–202., DOI 10. 1007/978-1-4939-8910-2_15 Xu X, Huang L, Futtner C, Schwab B, Rampersad RR, Lu Y, et al. The cell of origin and subtype of K-Ras-induced lung tumors are modified by Notch and Sox2. Genes Dev, 2014, 28(17):1929–39., DOI 10.1101/gad.243717.114 Pobbati AV, Hong W. A combat with the YAP/TAZ-TEAD oncoproteins for cancer therapy. Theranostics, 2020, 10(8):3622–35., DOI 10.7150/thno.40889 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 596522 EP 596533 DI 10.3389/pore.2021.596522 PG 12 ER PT J AU Cruz-Rico, G Aviles-Salas, A Popa-Navarro, X Lara-Mejia, L Catalan, R Sanchez-Reyes, R Lopez-Sanchez, D Cabrera-Miranda, L Maldonado-Martinez, AH Samtani-Bassarmal, S Arrieta, O AF Cruz-Rico, Graciela Aviles-Salas, Alejandro Popa-Navarro, Xitlally Lara-Mejia, Luis Catalan, Rodrigo Sanchez-Reyes, Roberto Lopez-Sanchez, Dennis Cabrera-Miranda, Luis Maldonado-Martinez, Aquiles Hector Samtani-Bassarmal, Suraj Arrieta, Oscar TI Association of Lung Adenocarcinoma Subtypes According to the IASLC/ ATS/ERS Classification and Programmed Cell Death Ligand 1 (PD-L1) Expression in Tumor Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunotherapy; NSCLC; lung adenocarcinoma; programmed-death receptor ligand 1 (PD-L1); immunohistochemistry; Tumor-infiltrating lymphocytes (TILs) 4 ID immunotherapy; NSCLC; lung adenocarcinoma; programmed-death receptor ligand 1 (PD-L1); immunohistochemistry; Tumor-infiltrating lymphocytes (TILs) 4 AB Background: Programmed cell death-ligand 1 (PD-L1) protein expression is one of the most extensively studied biomarkers in patients with non-small cell lung cancer (NSCLC). However, there is scarce information regarding its association with distinct adenocarcinoma subtypes. This study evaluated the frequency of PD-L1 expression according to the IASLC/ATS/ERS classification and other relevant histological and clinical features. Patients and Methods: PD-L1 expression was assessed by immunohistochemistry (IHC). According to its positivity in tumor cells membrane, we stratified patients in three different tumor proportions score (TPS) cut-off points: a) <1% (negative), b) between 1 and 49%, and c) ≥50%; afterward, we analyzed the association among PD-L1 expression and lung adenocarcinoma (LADC) predominant subtypes, as well as other clinical features. As an exploratory outcome we evaluated if a PD-L1 TPS score ≥15% was useful as a biomarker for determining survival. Results: A total of 240 patients were included to our final analysis. Median age at diagnosis was 65 years (range 23–94 years). A PD-L1 TPS ≥1% was observed in 52.5% of the entire cohort; regarding specific predominant histological patterns, a PDL1 TPS ≥1 was documented in 31.2% of patients with predominant-lepidic pattern, 46.2% of patients with predominant-acinar pattern, 42.8% of patients with a predominantpapillary pattern, and 68.7% of patients with predominant-solid pattern (p  0.002). On the other hand, proportion of tumors with PD-L1 TPS ≥50% was not significantly different among adenocarcinoma subtypes. At the univariate survival analysis, a PD-L1 TPS cut-off value of ≥15% was associated with a worse PFS and OS. Conclusion: According to IASLC/ATS/ERS lung adenocarcinoma classification, the predominant-solid pattern is associated with a higher proportion of PD-L1 positive samples, no subtype was identified to be associated with a high (≥50%) TPS PD-L1. C1 [Cruz-Rico, Graciela] Instituto Nacional de Cancerologia, Thoracic Oncology UnitMexico City, Mexico. [Aviles-Salas, Alejandro] Instituto Nacional de Cancerologia, Thoracic Oncology UnitMexico City, Mexico. [Popa-Navarro, Xitlally] Instituto Nacional de Cancerologia, Thoracic Oncology UnitMexico City, Mexico. [Lara-Mejia, Luis] Instituto Nacional de Cancerologia, Thoracic Oncology UnitMexico City, Mexico. [Catalan, Rodrigo] Instituto Nacional de Cancerologia, Thoracic Oncology UnitMexico City, Mexico. [Sanchez-Reyes, Roberto] Instituto Nacional de Cancerologia, Thoracic Oncology UnitMexico City, Mexico. [Lopez-Sanchez, Dennis] Instituto Nacional de Cancerologia, Thoracic Oncology UnitMexico City, Mexico. [Cabrera-Miranda, Luis] Instituto Nacional de Cancerologia, Thoracic Oncology UnitMexico City, Mexico. [Maldonado-Martinez, Aquiles Hector] Instituto Nacional de Cancerologia, Thoracic Oncology UnitMexico City, Mexico. [Samtani-Bassarmal, Suraj] Clinica Bradford Hill, Medical Oncology ServiceSantiago, Chile. [Arrieta, Oscar] Instituto Nacional de Cancerologia, Thoracic Oncology UnitMexico City, Mexico. RP Arrieta, O (reprint author), Instituto Nacional de Cancerologia, Thoracic Oncology Unit, Mexico City, Mexico. CR Siegel RL,Miller KD, Jemal A. Cancer statistics, 2019. CA A Cancer J Clin, 2019, 69(1):7–34., DOI 10.3322/caac.21551 Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med, 2012, 366(26):2443–54., DOI 10.1056/nejmoa1200690 Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumorassociated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med, 2002, 8(8):793–800., DOI 10.1038/nm730 Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer, 2012, 12(4):252–64., DOI 10.1038/nrc3239 Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med, 2015, 373(17):1627–39., DOI 10.1056/nejmoa1507643 Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Cs}oszi T, Fulop A, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med, 2016, 375(19):1823–33., DOI 10.1056/nejmoa1606774 Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther, 2015, 14(4):847–56., DOI 10.1158/1535- 7163.mct-14-0983 Helissey C, Champiat S, Soria J-C. Immune checkpoint inhibitors in advanced nonsmall cell lung cancer. Curr Opin Oncol, 2015, 27(2):108–17., DOI 10.1097/ cco.0000000000000167 Arrieta O, Barron F, Ramirez-Tirado LA, Zatarain-Barron ZL, Cardona AF, Diaz-Garcia D, et al. Efficacy and safety of pembrolizumab plus docetaxel vs docetaxel alone in patients with previously treated advanced non-small cell lung cancer: the PROLUNG phase 2 randomized clinical trial. JAMA Oncol, 2020, 6(6):1–9., DOI 10.1001/jamaoncol.2020.0409 Herbst RS, Baas P, Kim D-W, Felip E, Perez-Gracia JL, Han J-Y, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer, KEYNOTE-010): a randomised controlled trial. The Lancet, 2016, 387(10027):1540–50., DOI 10.1016/s0140- 6736(15)01281-7 Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, et al. Atezolizumab versus docetaxel for patients with previously treated nonsmall- cell lung cancer, POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. The Lancet, 2016, 387(10030):1837–46., DOI 10. 1016/s0140-6736(16)00587-0 Hirsch FR,McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, et al. PD-L1 immunohistochemistry assays for lung cancer: results from phase 1 of the Blueprint PD-L1 IHC assay comparison Project. J Thorac Oncol, 2017, 12(2):208–22., DOI 10.1016/j.jtho.2016.11.2228 Tsao MS, Kerr KM, Kockx M, Beasley M-B, Borczuk AC, Botling J, et al. PD-L1 immunohistochemistry comparability study in real-life clinical samples: results of Blueprint phase 2 Project. J Thorac Oncol, 2018, 13(9):1302–11., DOI 10.1016/j.jtho.2018.05.013 Koh J, Go H, Keam B, Kim M-Y, Nam SJ, Kim TM, et al. Clinicopathologic analysis of programmed cell death-1 and programmed cell death-ligand 1 and 2 expressions in pulmonary adenocarcinoma: comparison with histology and driver oncogenic alteration status. Mod Pathol, 2015, 28(9):1154–66., DOI 10. 1038/modpathol.2015.63 Zugazagoitia J, Enguita AB, Nunez JA, Iglesias L, Ponce S. The new IASLC/ ATS/ERS lung adenocarcinoma classification from a clinical perspective: current concepts and future prospects. J Thorac Dis, 2014, 6(Suppl. 5): S526–36., DOI 10.3978/j.issn.2072-1439.2014.01.27 Yoshizawa A, Motoi N, Riely GJ, Sima CS, Gerald WL, Kris MG, et al. Impact of proposed IASLC/ATS/ERS classification of lung adenocarcinoma: prognostic subgroups and implications for further revision of staging based on analysis of 514 stage I cases. Mod Pathol, 2011, 24(5):653–64., DOI 10.1038/ modpathol.2010.232 Yu H, Boyle TA, Zhou C, Rimm DL, Hirsch FR. PD-L1 expression in lung cancer. J Thorac Oncol, 2016, 11(7):964–75., DOI 10.1016/j.jtho.2016. 04.014 Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Cs}oszi T, Fulop A, et al. Updated analysis of KEYNOTE-024: pembrolizumab versus platinum-based chemotherapy for advanced non-small-cell lung cancer with PD-L1 tumor proportion score of 50% or greater. J Clin Oncol, 2019, 37(7):537–46., DOI 10. 1200/JCO.18.00149 Rizvi NA, Cho BC, Reinmuth N, Lee KH, Luft A, Ahn M-J, et al. Durvalumab with or without tremelimumab vs standard chemotherapy in first-line treatment of metastatic non-small cell lung cancer. JAMA Oncol, 2020, 6(5):661–74., DOI 10.1001/jamaoncol.2020.0237 Kerr KM, Tsao M-S, Nicholson AG, Yatabe Y, WistubaII, Hirsch FR. Programmed death-ligand 1 immunohistochemistry in lung cancer: in what state is this art? J Thorac Oncol, 2015, 10(7):985–9., DOI 10.1097/jto. 0000000000000526 Igarashi T, Teramoto K, Ishida M, Hanaoka J, Daigo Y. Scoring of PD-L1 expression intensity on pulmonary adenocarcinomas and the correlations with clinicopathological factors. ESMO Open, 2016, 1(4):e000083., DOI 10.1136/ esmoopen-2016-000083 Takada K, Okamoto T, Shoji F, Shimokawa M, Akamine T, Takamori S, et al. Clinical significance of PD-L1 protein expression in surgically resected primary lung adenocarcinoma. J Thorac Oncol, 2016, 11(11):1879–90., DOI 10.1016/j.jtho.2016.06.006 Mori S, Motoi N, Ninomiya H, Matsuura Y, Nakao M, Mun M, et al. High expression of programmed cell death 1 ligand 1 in lung adenocarcinoma is a poor prognostic factor particularly in smokers and wild-type epidermal growth-factor receptor cases. Pathol Int, 2017, 67(1):37–44., DOI 10.1111/ pin.12489 Calles A, Liao X, Sholl LM, Rodig SJ, Freeman GJ, Butaney M, et al. Expression of PD-1 and its ligands, PD-L1 and PD-L2, in smokers and never smokers with KRAS-mutant lung cancer. J Thorac Oncol, 2015, 10(12):1726–35., DOI 10. 1097/jto.0000000000000687 Hodge G, Nairn J, Holmes M, Reynolds PN, Hodge S Increased intracellular T helper 1 proinflammatory cytokine production in peripheral blood, bronchoalveolar lavage and intraepithelial T cells of COPD subjects. Clin Exp Immunol, 2007, 150(1):22–9., DOI 10.1111/j.1365-2249.2007.03451.x Batenchuk C, Albitar M, Zerba K, Sudarsanam S, Chizhevsky V, Jin C, et al. A real-world, comparative study of FDA-approved diagnostic assays PD-L1 IHC 28-8 and 22C3 in lung cancer and other malignancies. J Clin Pathol, 2018, 71(12):1078–83., DOI 10.1136/jclinpath-2018-205362 Teng MWL, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res, 2015, 75(11):2139–45., DOI 10.1158/0008- 5472.can-15-0255 Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med, 2018, 378(22):2078–92., DOI 10.1056/ nejmoa1801005 Arrieta O, Cardona AF, Federico Bramuglia G, Gallo A, Campos-Parra AD, Serrano S, et al. Genotyping non-small cell lung cancer, NSCLC, in Latin America. J Thorac Oncol, 2011, 6(11):1955–9., DOI 10.1097/jto.0b013e31822f655f Arrieta O, Cardona AF, Martin C, Mas-Lopez L, Corrales-Rodriguez L, Bramuglia G, et al. Updated frequency of EGFR and KRAS mutations in NonSmall-cell lung cancer in Latin America: the Latin-American consortium for the investigation of lung cancer, CLICaP). J Thorac Oncol, 2015, 10(5): 838–43., DOI 10.1097/jto.0000000000000481 Arrieta O, Ramirez-Tirado L-A, Baez-Saldana R, Pena-Curiel O, Soca-Chafre G, Macedo-Perez E-O. Different mutation profiles and clinical characteristics among Hispanic patients with non-small cell lung cancer could explain the “Hispanic paradox”. Lung Cancer, 2015, 90(2):161–6., DOI 10.1016/j.lungcan. 2015.08.010 Yoneshima Y, Ijichi K, Anai S, Ota K, Otsubo K, Iwama E, et al. PD-L1 expression in lung adenocarcinoma harboring EGFR mutations or ALK rearrangements. Lung Cancer, 2018, 118:36–40., DOI 10.1016/j.lungcan.2018. 01.024 Chen N, Fang W, Zhan J,Hong S, Tang Y, Kang S, et al. Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: implication for optional immune targeted therapy for NSCLC patients with EGFR mutation. J Thorac Oncol, 2015, 10(6):910–23., DOI 10.1097/jto. 0000000000000500 Matsumoto Y, Sawa K, Fukui M, Oyanagi J, IzumiM, Ogawa K, et al. Impact of tumor microenvironment on the efficacy of epidermal growth factor receptortyrosine kinase inhibitors in patients with EGFR -mutant non-small cell lung cancer. Cancer Sci, 2019, 110(10):3244–54., DOI 10.1111/cas.14156 Azuma K, Ota K, Kawahara A, Hattori S, Iwama E, Harada T, et al. Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann Oncol, 2014, 25(10):1935–40., DOI 10.1093/annonc/mdu242 Tang Y, Fang W, Zhang Y, Hong S, Kang S, Yan Y, et al. The association between PD-L1 and EGFR status and the prognostic value of PD-L1 in advanced non-small cell lung cancer patients treated with EGFR-TKIs. Oncotarget, 2015, 6(16):14209–19., DOI 10.18632/oncotarget.3694 Takada K, Toyokawa G, Tagawa T, Kohashi K, Shimokawa M, Akamine T, et al. PD-L1 expression according to the EGFR status in primary lung adenocarcinoma. Lung Cancer, 2018, 116:1–6., DOI 10.1016/j.lungcan.2017. 12.003 Chan AWH, Tong JHM, Kwan JSH, Chow C, Chung LY, Chau SL, et al. Assessment of programmed cell death ligand-1 expression by 4 diagnostic assays and its clinicopathological correlation in a large cohort of surgical resected non-small cell lung carcinoma. Mod Pathol, 2018, 31(9):1381–90., DOI 10.1038/s41379-018-0053-3 Rangachari D, VanderLaan PA, Shea M, Le X, Huberman MS, Kobayashi SS, et al. Correlation between classic driver oncogene mutations in EGFR, ALK, or ROS1 and 22C3-PD-L1 ≥50% expression in lung adenocarcinoma. J Thorac Oncol, 2017, 12(5):878–83., DOI 10.1016/j.jtho.2016.12.026 Ota K, Azuma K, Kawahara A, Hattori S, Iwama E, Tanizaki J, et al. Induction of PD-L1 expression by the EML4-ALK oncoprotein and downstream signaling pathways in non-small cell lung cancer. Clin Cancer Res, 2015, 21(17):4014–21., DOI 10.1158/1078-0432.ccr-15-0016 Marzec M, Zhang Q, Goradia A, Raghunath PN, Liu X, Paessler M, et al. Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274, PD-L1, B7-H1). Proc Natl Acad Sci, 2008, 105(52):20852–7., DOI 10.1073/pnas.0810958105 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 597499 EP 597507 DI 10.3389/pore.2021.597499 PG 9 ER PT J AU Zhang, R Huang, M Wang, H Wu, Sh Yao, J Ge, Y Lu, Y Hu, Q AF Zhang, Ruohao Huang, Miao Wang, Hong Wu, Shengming Yao, Jiali Ge, Yingying Lu, Yufei Hu, Qiping TI Identification of Potential Biomarkers From Hepatocellular Carcinoma With MT1 Deletion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE hepatocellular carcinoma; copy number variation; metallothionein; hub gene; deletion; screening; biomarker ID hepatocellular carcinoma; copy number variation; metallothionein; hub gene; deletion; screening; biomarker AB Background: Hepatocellular carcinoma (HCC) is one of the deadliest cancers worldwide. Metallothioneins (MTs) are metal-binding proteins involved in multiple biological processes such as metal homeostasis and detoxification, as well as in oncogenesis. Copy number variation (CNV) plays a vital role in pathogenesis and carcinogenesis. Nevertheless, there is no study on the role of MT1 CNV in HCC. Methods: Array-based Comparative Genomic Hybridization (aCGH) analysis was performed to obtain the CNV data of 79 Guangxi HCC patients. The prognostic effect of MT1-deletion was analyzed by univariate and multivariate Cox regression analysis. The differentially expressed genes (DEGs) were screened based on The Gene Expression Omnibus database (GEO) and the Liver Hepatocellular Carcinoma of The Cancer Genome Atlas (TCGA-LIHC). Then function and pathway enrichment analysis, protein-protein interaction (PPI) and hub gene selection were applied on the DEGs. Lastly, the hub genes were validated by immunohistochemistry, tissue expression and prognostic analysis. Results: The MT1-deletion was demonstrated to affect the prognosis of HCC and can act as an independent prognostic factor. 147 common DEGs were screened. The most significant cluster of DEGs identified by Molecular Complex Detection (MCODE) indicated that the expression of four MT1s were down-regulated. MT1X and other five hub genes (TTK, BUB1, CYP3A4, NR1I2, CYP8B1) were associated with the prognosis of HCC. TTK, could affect the prognosis of HCC with MT1-deletion and non-deletion. NR1I2, CYP8B1, and BUB1 were associated with the prognosis of HCC with MT1-deletion. Conclusions: In the current study, we demonstrated that MT1-deletion can be an independent prognostic factor in HCC. We identified TTK, BUB1, NR1I2, CYP8B1 by processing microarray data, for the first time revealed the underlying function of MT1 deletion in HCC, MT1-deletion may influence the gene expression in HCC, which may be the potential biomarkers for HCC with MT1 deletion. C1 [Zhang, Ruohao] Guangxi Medical University, School of Pre-clinical Medicine, Department of Cell Biology and GeneticsNanning, China. [Huang, Miao] Affiliated Tumor Hospital of Guangxi Medical University, Radiology DepartmentNanning, China. [Wang, Hong] Guangxi Medical University, School of Pre-clinical Medicine, Department of Cell Biology and GeneticsNanning, China. [Wu, Shengming] Affiliated Tumor Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Yao, Jiali] Guangxi Medical University, School of Pre-clinical Medicine, Department of Cell Biology and GeneticsNanning, China. [Ge, Yingying] Guangxi Medical University, School of Pre-clinical Medicine, Department of Histology and EmbryologyNanning, China. [Lu, Yufei] Guangxi Medical University, School of Pre-clinical Medicine, Department of Cell Biology and GeneticsNanning, China. [Hu, Qiping] Guangxi Medical University, School of Pre-clinical Medicine, Department of Cell Biology and GeneticsNanning, China. RP Hu, Q (reprint author), Guangxi Medical University, School of Pre-clinical Medicine, Department of Cell Biology and Genetics, Nanning, China. EM huqiping@gxmu.edu.cn CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer J clinicians, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Shlien A., Malkin D. Copy number variations and cancer. Genome Med, 2009, 1(6):62., DOI 10.1186/gm62 Cao P, Yang A, Wang R, Xia X, Zhai Y, Li Y, et al. Germline duplication of SNORA18L5 increases risk for HBV-related hepatocellular carcinoma by altering localization of ribosomal proteins and decreasing levels of p53. Gastroenterology, 2018, 155(2):542–56., DOI 10.1053/j.gastro.2018.04.020 Stranger BE, Forrest MS, Dunning M, Ingle CE, Beazley C, Thorne N, et al. Relative impact of nucleotide and copy number variation on gene expression phenotypes. Science, 2007, 315(5813):848–53., DOI 10.1126/science.1136678 Zhang C, Cao P, Yang A, Xia X, Li Y, Shi M, et al. Downregulation of ZC3H14 driven by chromosome 14q31 deletion promotes hepatocellular carcinoma progression by activating integrin signaling. Carcinogenesis, 2019, 40(3): 474–86., DOI 10.1093/carcin/bgy146 Vasak M. Advances in metallothionein structure and functions. J Trace Elem Med Biol, 2005, 19(1):13–7., DOI 10.1016/j.jtemb.2005.03.003 Takahashi S. Positive and negative regulators of the metallothionein gene, review). Mol Med Rep, 2015, 12(1):795–9., DOI 10.3892/mmr.2015.3459 Si M., Lang J. The roles of metallothioneins in carcinogenesis. J Hematol Oncol, 2018, 11(1):107., DOI 10.1186/s13045-018-0645-x Grommisch B, DiAdamo AJ, Xu ZY, Pan XH, Ma DQ, Xie JS, et al. Biobanking of residual specimens from diagnostic genetic laboratories: standard operating procedures, ethical and legal considerations, and research applications. North Am J Med Sci, 2013, 6(4):200–7., DOI 10. 7156/najms.2013.0604200 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res, 2015, 43(7):e47., DOI 10.1093/nar/gkv007 Chen H. Boutros PCJBB VennDiagram: a package for the generation of highlycustomizable venn and euler diagrams in R. 12. BMC Bioinformatics, 2011, 12: 35., DOI 10.1186/1471-2105-12-35 Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res, 2019, 47(D1):D607–d613., DOI 10.1093/nar/gky1131 Shannon P,Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res, 2003, 13(11):2498–504., DOI 10.1101/gr.1239303 Chin C-H, Chen S-H, Wu H-H, Ho C-W, Ko M-T, Lin C-Y cytoHubba: identifying hub objects and sub-networks from complex interactome. BMC Syst Biol, 2014, 8(Suppl 4):S11., DOI 10.1186/1752-0509-8-s4-s11 Dennis G, Jr., Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, et al. DAVID: database for annotation, visualization, and integrated discovery. Genome Biol, 2003, 4(5):P3., DOI 10.1186/gb-2003-4-5-p3 Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res, 2017, 45(W1):W98–w102., DOI 10.1093/nar/gkx247 Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Tissue-based map of the human proteome. Science, 2015, 347(6220): 1260419., DOI 10.1126/science.1260419 Lapenna S, Giordano A. Cell cycle kinases as therapeutic targets for cancer. Nat Rev Drug Discov, 2009, 8(7):547–66., DOI 10.1038/nrd2907 Otto T, Sicinski P. Cell cycle proteins as promising targets in cancer therapy. Nat Rev Cancer, 2017, 17(2):93–115., DOI 10.1038/nrc.2016.138 Levine AJ. p53: 800 million years of evolution and 40 years of discovery. Nat Rev Cancer, 2020, 20(8):471–80., DOI 10.1038/s41568-020-0262-1 Zarrinpar A. Metabolic pathway inhibition in liver cancer. Slas Technology: Translating Life Sci Innovation, 2017, 22(3):237–44., DOI 10.1177/2472630317698683 Wang XT, Chen CW, Zheng XM, Wang B, Zhang SX, Yao MH, et al. Expression and prognostic significance of melatonin receptor MT1 in patients with gastric adenocarcinoma. neo, 2020, 67(2):415–20., DOI 10. 4149/neo_2019_190220N141 Jagielski L, Jelen M, Kobierzycki C, Jagielska G, Blok R. Increase of nuclear expression of metallothionein I/II in neoplastic transformation of the endometrium. Ginekol Pol, 2015, 86(3):182–7., DOI 10.17772/gp/2060 Wu Y-J, Ko B-S, Liang S-M, Lu Y-J, Jan Y-J, Jiang S-S, et al. ZNF479 downregulates metallothionein-1 expression by regulating ASH2L and DNMT1 in hepatocellular carcinoma. Cell Death Dis, 2019, 10(6):408., DOI 10.1038/s41419-019-1651-9 Wang Y, Wang G, Tan X, Ke K, Zhao B, Cheng N, et al. MT1G serves as a tumor suppressor in hepatocellular carcinoma by interacting with p53. Oncogenesis, 2019, 8(12):67., DOI 10.1038/s41389-019-0176-5 Lu DD, Chen YC, Zhang XR, Cao XR, Jiang HY, Yao L. The relationship between metallothionein-1F, MT1F, gene and hepatocellular carcinoma. Yale J Biol Med, 2003, 76(2):55–62. Liu Z, Ye Q, Wu L, Gao F, Xie H, Zhou L, et al. Metallothionein 1 family profiling identifies MT1X as a tumor suppressor involved in the progression and metastastatic capacity of hepatocellular carcinoma. Mol carcinogenesis, 2018, 57(11):1435–44., DOI 10.1002/mc.22846 Musacchio A. Spindle assembly checkpoint: the third decade. Phil Trans R Soc B, 2011, 366(1584):3595–604., DOI 10.1098/rstb.2011.0072 Xie Y,Wang A, Lin J, Wu L, Zhang H, Yang X, et al. Mps1/TTK: a novel target and biomarker for cancer. J Drug Target, 2017, 25(2):112–8., DOI 10.1080/ 1061186x.2016.1258568 Daniel J, Coulter J, Woo J-H, Wilsbach K, Gabrielson E. High levels of the Mps1 checkpoint protein are protective of aneuploidy in breast cancer cells. Proc Natl Acad Sci, 2011, 108(13):5384–9., DOI 10.1073/pnas.1007645108 Maire V, Baldeyron C, Richardson M, Tesson B, Vincent-Salomon A, Gravier E, et al. TTK/hMPS1 is an attractive therapeutic target for triple-negative breast cancer. PloS one, 2013, 8(5):e63712., DOI 10.1371/journal.pone.0063712 Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA: a Cancer J clinicians, 2011, 61(2):69–90., DOI 10.3322/caac.20107 Manic G, Corradi F, Sistigu A, Siteni S, Vitale I. Molecular regulation of the spindle assembly checkpoint by kinases and phosphatases. Int Rev CelMol Biol, 2017, 328:105–61., DOI 10.1016/bs.ircmb.2016.08.004 Bolanos-Garcia VM, Blundell TL. BUB1 and BUBR1: multifaceted kinases of the cell cycle. Trends Biochemical Sciences, 2011, 36(3):141–50., DOI 10.1016/j. tibs.2010.08.004 Alzahrani AM, Rajendran P. The multifarious link between cytochrome P450s and cancer. Oxidative Med Cell longevity, 2020, 2020:1., DOI 10.1155/2020/3028387 Sandstrom R, Knutson TW, Knutson L, Jansson B, Lennernas H. The effect of ketoconazole on the jejunal permeability and CYP3A metabolism of, R/S)- verapamil in humans. Br J Clin Pharmacol, 1999, 48(2):180–9., DOI 10.1046/j. 1365-2125.1999.00999.x Murray GI, Patimalla S, Stewart KN, Miller ID, Heys SD. Profiling the expression of cytochrome P450 in breast cancer. Histopathology, 2010, 57(2):202–11., DOI 10.1111/j.1365-2559.2010.03606.x Fujimura T, Takahashi S, Urano T, Kumagai J, Murata T, Takayama K, et al. Expression of cytochrome P450 3A4 and its clinical significance in human prostate cancer. Urology, 2009, 74(2):391–7., DOI 10.1016/j.urology.2009.02.033 Ashida R, Okamura Y, Ohshima K, Kakuda Y, Uesaka K, Sugiura T, et al. Gene is a novel biomarker for predicting a poor prognosis in hepatocellular carcinoma. Cancer Genomics Proteomics, 2017, 14(6):445–53., DOI 10.21873/ cgp.20054 Yu T, Wang X, Zhu G, Han C, Su H, Liao X, et al. The prognostic value of differentially expressed CYP3A subfamily members for hepatocellular carcinoma. Cmar, 2018, Vol. 10:1713–26., DOI 10. 2147/cmar.S159425 Feng F, Jiang Q, Cao S, Cao Y, Li R, Shen L, et al. Pregnane X receptor mediates sorafenib resistance in advanced hepatocellular carcinoma. Biochim Biophys Acta, Bba)—Gen Subjects, 2018, 1862(4):1017–30., DOI 10.1016/j.bbagen.2018. 01.011 Harmsen S, Meijerman I, Maas-Bakker RF, Beijnen JH, Schellens JHM. PXRmediated P-glycoprotein induction by small molecule tyrosine kinase inhibitors. Eur J Pharm Sci, 2013, 48(4-5):644–9., DOI 10.1016/j.ejps.2012. 12.019 Jiang H, Chen K, He J, Pan F, Li J, Chen J, et al. Association of pregnane X receptor with multidrug resistance-related protein 3 and its role in human colon cancer chemoresistance. J Gastrointest Surg, 2009, 13(10):1831–8., DOI 10.1007/s11605-009-0964-x Gupta D, Venkatesh M, Wang H, Kim S, Sinz M, Goldberg GL, et al. Expanding the roles for pregnane X receptor in cancer: proliferation and drug resistance in ovarian cancer. Clin Cancer Res, 2008, 14(17):5332–40., DOI 10.1158/1078-0432.Ccr-08-1033 Chen Y, Tang Y, Wang M-T, Zeng S, Nie D. Human pregnane X receptor and resistance to chemotherapy in prostate cancer. Cancer Res, 2007, 67(21): 10361–7., DOI 10.1158/0008-5472.Can-06-4758 Yoshinari K. Role of nuclear receptors PXR and CAR in xenobiotic-induced hepatocyte proliferation and chemical carcinogenesis. Biol Pharm Bull, 2019, 42(8):1243–52., DOI 10.1248/bpb.b19-00267 Chen H, Shen ZY, Xu W, Fan TY, Li J, Lu YF, et al. Expression of P450 and nuclear receptors in normal and end-stage Chinese livers. Wjg, 2014, 20(26): 8681–90., DOI 10.3748/wjg.v20.i26.8681 Zhang M, Chiang JYL. Transcriptional regulation of the human sterol 12α- hydroxylase gene, CYP8B1). J Biol Chem, 2001, 276(45):41690–9., DOI 10. 1074/jbc.M105117200 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 597527 EP 597539 DI 10.3389/pore.2021.597527 PG 13 ER PT J AU Tamasi, L Horvath, K Kiss, Z Bogos, K Ostoros, Gy Muller, V Urban, L Bittner, N Sarosi, V Vastag, A Polanyi, Z Nagy-Erdei, Zs Daniel, A Nagy, B Rokszin, Gy Abonyi-Toth, Zs Moldvay, J Voko, Z Galffy, G AF Tamasi, Lilla Horvath, Krisztian Kiss, Zoltan Bogos, Krisztina Ostoros, Gyula Muller, Veronika Urban, Laszlo Bittner, Nora Sarosi, Veronika Vastag, Aladar Polanyi, Zoltan Nagy-Erdei, Zsofia Daniel, Andrea Nagy, Balazs Rokszin, Gyorgy Abonyi-Toth, Zsolt Moldvay, Judit Voko, Zoltan Galffy, Gabriella TI Age and Gender Specific Lung Cancer Incidence and Mortality in Hungary: Trends from 2011 Through 2016 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE epidemiology; lung cancer; incidence; mortality; age and gender ID epidemiology; lung cancer; incidence; mortality; age and gender AB Objective: No assessment was conducted describing the age and gender specific epidemiology of lung cancer (LC) prior to 2018 in Hungary, thus the objective of this study was to appraise the detailed epidemiology of lung cancer (ICD-10 C34) in Hungary based on a retrospective analysis of the National Health Insurance Fund database. Methods: This longitudinal study included patients aged ≥20 years with LC diagnosis (ICD-10 C34) between January 1, 2011 and December 31, 2016. Patients with different cancer-related codes 6 months before or 12 months after LC diagnosis or having any cancer treatment other than lung cancer protocols were excluded. Results: Lung cancer incidence and mortality increased with age, peaking in the 70–79 age group (375.0/100,000 person-years) among males, while at 60–69 age group for females (148.1/100,000 person-years). The male-to-female incidence rate ratio reached 2.46–3.01 (p < 0.0001) among the 70–79 age group. We found 2–11% decrease in male incidence rate at most age groups, while a significant 1–3% increase was observed in older females (>60) annually during the study period. Conclusion: This nationwide epidemiology study demonstrated that LC incidence and mortality in Hungary decreased in younger male and female population, however we found significant increase of incidence in older female population, similar to international trends. Incidence rates peaked in younger age-groups compared to Western countries, most likely due to higher smoking prevalence in these cohorts, while lower age LC incidence could be attributed to higher competing cardiovascular risk resulting in earlier mortality in smoking population. C1 [Tamasi, Lilla] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Horvath, Krisztian] Eotvos Lorand University, Department of Health Policy and Health EconomicsBudapest, Hungary. [Kiss, Zoltan] MSD Pharma Hungary Kft.Budapest, Hungary. [Bogos, Krisztina] National Koranyi Institute for TB and Pulmonology, Department of PulmonologyBudapest, Hungary. [Ostoros, Gyula] National Koranyi Institute for TB and Pulmonology, Department of PulmonologyBudapest, Hungary. [Muller, Veronika] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Urban, Laszlo] Matrahaza Healthcare Center and University Teaching HospitalMatrahaza, Hungary. [Bittner, Nora] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Sarosi, Veronika] University of Pecs, Faculty of MedicinePecs, Hungary. [Vastag, Aladar] MSD Pharma Hungary Kft.Budapest, Hungary. [Polanyi, Zoltan] MSD Pharma Hungary Kft.Budapest, Hungary. [Nagy-Erdei, Zsofia] MSD Pharma Hungary Kft.Budapest, Hungary. [Daniel, Andrea] MSD Pharma Hungary Kft.Budapest, Hungary. [Nagy, Balazs] Eotvos Lorand University, Department of Health Policy and Health EconomicsBudapest, Hungary. [Rokszin, Gyorgy] RxTarget LtdSzolnok, Hungary. [Abonyi-Toth, Zsolt] RxTarget LtdSzolnok, Hungary. [Moldvay, Judit] National Koranyi Institute of Pulmonology, Department of Tumor BiologyBudapest, Hungary. [Voko, Zoltan] Eotvos Lorand University, Department of Health Policy and Health EconomicsBudapest, Hungary. [Galffy, Gabriella] Semmelweis University, Department of PulmonologyBudapest, Hungary. RP Horvath, K (reprint author), Eotvos Lorand University, Department of Health Policy and Health Economics, Budapest, Hungary. EM horvath.krisztian@tatk.elte.hu CR Ferlay, J., Soerjomataram, I., and Dikshit, R., 2015). Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int. J. Canc. 136, 5), E359–E386., DOI 10.1002/ijc.29210 Alberg, A. J., and Samet, J. M., 2003). Epidemiology of lung cancer*. Chest 123, 1), 21S–49S. ISSN 0012-3692. Cancer Research UK, 2019). Data are fromCancer Research UK portal, referenced per instruction on the homepage. Available at: https://www.cancerresearchuk.org/ health-professional/cancer-statistics/statistics-by-cancer-type/lung-cancer#heading- Three, Accessed September 24, 2019). Patricia de, G., and Munden, R. F., 2012). Lung cancer epidemiology, risk factors, and prevention. Radiol. Clin. 50, 5), 863–876., DOI 10.1016/j.rcl.2012.06.006 Cancer Research UK database, 2019). Data were provided by the Cancer Research UK database. Available at: https://www.cancerresearchuk.org/health-professional/ cancer-statistics/statistics-by-cancer-type/lung-cancer/incidence#ref, Accessed October 6, 2019). Akhtar-Danesh, N., and Finley, C., 2015). Temporal trends in the incidence and relative survival of non-small cell lung cancer in Canada: a population-based study. Lung Canc. 90, 1), 8–14., DOI 10.1016/j.lungcan.2015.07.004 Torre, L. A., Siegel, R. L., and Jemal, A., 2016). Lung cancer statistics. Adv. Exp. Med. Biol. 893, 1–19., DOI 10.1007/978-3-319-24223-1_1 Malvezzi, M., Bertuccio, P., and Rosso, T., 2015). European cancer mortality predictions for the year 2015: does lung cancer have the highest death rate in EU women? Ann. Oncol. 26, 4), 779–786., DOI 10. 1093/annonc/mdv001 Paggi, M. G., Vona, R., Abbruzzese, C., and Malorni,W., 2010). Gender-related disparities in non-small cell lung cancer. Cancer Lett. 298, 1), 1–8., DOI 10.1016/j. canlet.2010.08.009 Oberli, L. S., Valeri, F., and Korol, D., 2016). 31 years of lung cancer in the canton of Zurich, Switzerland: incidence trends by sex, histology and laterality. Swiss Med. Wkly. 146, w14327., DOI 10.4414/smw.2016.14327 eCollection 2016 Bogos, K., Kiss, Z., Galffy, G., Tamasi, L., Ostoros, G., Muller, V., et al., 2019). Revising incidence and mortality of lung cancer in Central Europe: an epidemiology review from Hungary. Front. Oncol. 9, 1051., DOI 10.3389/ fonc.2019.01051 Statinfo, K. S. H., 2019). http://statinfo.ksh.hu/Statinfo/index.jsp, Accessed November 24, 2019). European Commission, 2013). Revision of the European standard population—report of Eurostat’s task force. Luxembourg: publications office of the European union. Available at: https://ec.europa.eu/eurostat/documents/ 3859598/5926869/KS-RA-13-028-EN.PDF/e713fa79-1add-44e8-b23d- 5e8fa09b3f8f, Accessed March 9, 2019). Jemal, A., Thun, M. J., and Ries, L. A., 2008). Annual report to the nation on the status of cancer, 1975-2005, featuring trends in lung cancer, tobacco use, and tobacco control. J. Natl. Canc. Inst. 100, 1672–1694., DOI 10.1093/jnci/ djn389 McGuire, S., 2013). World cancer report 2014. Geneva, Switzerland: World Health Organization, international agency for research on cancer,WHOPress, 2015. Adv. Nutr. 7, 418–419., DOI 10.3945/an.116.012211 Youlden, D. R., Cramb, S. M., and Baade, P. D., 2008). The International Epidemiology of Lung Cancer: geographical distribution and secular trends. J. Thorac. Oncol. 3, 819–831., DOI 10.1097/JTO.0b013e31818020eb Siegel, R. L., Miller, K. D., and Jemal, A., 2018). Cancer statistics. CA Canc. J. Clin. 68, 7–30., DOI 10.3322/caac.21442 Hakulinen, T., Engholm, G., and Gislum, M., 2010). Trends in the survival of patients diagnosed with cancers in the respiratory system in the Nordic countries 1964–2003 followed up to the end of 2006. Acta Oncol. 49, 5), 608–623., DOI 10.3109/02841860903575281 Fu, J. B., Kau, T. Y., and Severson, R. K., 2005). Lung cancer in women: analysis of the national surveillance, epidemiology, and end results database. Chest 127, 3), 768–777., DOI 10.1378/chest.127.3.768 Carole, A. R., and Aoife, M., 2013). McErlean. Epidemiology of lung cancer. Semin. Intervent. Radiol. 30, 93–98., DOI 10.1055/s-0033-1342949 Tombor, I., Paksi, B., and Urban, B., 2011). Epidemiology of smoking in the Hungarian population, based on national representative data. Clin. Exp. Med. J. 5, 1), 27–37., DOI 10.1556/CEMEd.4.2010.28817 Joo, T., Voko, Z., and Bodrogi, J., 2017). Anti-smoking actions between 2010–2014. IME. XVI, 10), 11–17. Thomas, A., Chen, Y., and Yu, T., 2015). Trends and Characteristics of young non-small cell lung cancer patients in the United States. Front. Oncol. 5, 113., DOI 10.3389/fonc.2015.00113 eCollection 2015 DelaCruz,C. S.,Tanoue, L. T.,andMatthay, R. A. Lung cancer: epidemiology, etiology, and prevention. Clin. Chest Med. 32, 4), 605–644., DOI 10.1016/j.ccm.2011.09.001 Rivera, M. P., 2013). Lung cancer in women: differences in epidemiology, biology, histology, and treatment outcomes. Semin. Respir. Crit. Care Med. 34, 6), 792–801., DOI 10.1055/s-0033-1358550 KSH, 2009). Europai Lakossagi Egeszsegfelmeres Statisztikai Tukor IV evfolyam, 50. Szam 2010. http://www.ksh.hu/docs/hun/xftp/gyor/jel/ jel310021.pdf, Accessed November 28, 2019), 1–7. Cselko, Z., and Kovacs, G., 2013). Smoking habits in Hungary: analysis based on surveys of the past decade. Hungarian. Orv Hetil. 154, 37), 1454–1468., DOI 10.1556/OH.2013.29694 Kovacs, G., Barsai, A., and Szilasi, M., 2012). A dohanyzas, mint a tudorakos betegek tulelesenek prognosztikai tenyezoje [Smoking: a prognostic factor of lung cancer survival]. Magy. Onkol. 56, 3), 187–191. Taioli, E., andWynder, E. L., 1994). Re: Endocrine factors and adenocarcinoma of the lung in women. J. Natl. Canc. Inst. 86, 11), 869–870., DOI 10.1093/jnci/86.11.869 European Environment Agency, 2014). Air pollution fact sheet, Hungary. Available at: https://www.eea.europa.eu/themes/air/air-pollution-countryfact- sheets-2014/hungary-air-pollutant-emissions-country-factsheet+&cd 1&hlhu&ctclnk&glqa, Accessed September 20, 2020). Aareleid, T., Zimmermann, M. L., and Baburin, A., 2017). Divergent trends in lung cancer incidence by gender, age and histological type in Estonia: a nationwide population-based study. BMC Canc. 17, 1), 596., DOI 10.1186/ s12885-017-3605-x Thakkar, J. P., McCarthy, B. J., and Villano, J. L., 2014). Age-specific cancer incidence rates increase through the oldest age groups. Am. J. Med. Sci. 348, 1), 65–70., DOI 10.1097/MAJ.0000000000000281 Timmis, A., Townsend, N., and Gale, C., 2018). European society of cardiology: cardiovascular disease statistics 2017. Eur. Heart J. 39, 7), 508–579., DOI 10.1093/eurheartj/ehx628 Taghizadeh, N., Vonk, J. M., and Boezen,H.M., 2016). Lifetime smoking history and cause-specificmortality in a cohort study with 43 years of follow-up. PLoS One. 11, 4), e0153310., DOI 10.1371/journal.pone.0153310 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 598862 EP 598870 DI 10.3389/pore.2021.598862 PG 9 ER PT J AU Peng, L Zhang, Z Zhao, D Zhao, J Mao, F Sun, Q AF Peng, Li Zhang, Zhen Zhao, Dachun Zhao, Jialin Mao, Feng Sun, Qiang TI Discordance in ER, PR, HER2, and Ki-67 Expression Between Primary and Recurrent/Metastatic Lesions in Patients with Primary Early Stage Breast Cancer and the Clinical Significance: Retrospective Analysis of 75 Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomolecular status; discordance; recurrence; metastatic disease; PFS; breast cancer; surrogate subtypes ID biomolecular status; discordance; recurrence; metastatic disease; PFS; breast cancer; surrogate subtypes AB Background: The objective was to explore the discordance in the expression of the estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), and Ki-67 between primary and recurrent/metastatic lesions in patients with early stage breast cancer as well as the prognostic impact. Method: Patients with early-stage primary breast cancer and confirmed recurrence/ metastasis at Peking Union Medical College Hospital between January 2005 and August 2018 were screened. The details of discordance in each parameter between primary and recurrent/metastatic lesions and progression were recorded. Regression and survival analysis were applied to determine the association and clinical impact of the discordance. Results: We evaluated 75 patients. The discordance rate of ER, PR, HER2, and Ki-67 expression was 9.3, 14.7, 14.7, and 21.5%, respectively. Additionally, 66.7, 11.8, 14.3, and 0% of patients with Luminal A, Luminal B, HER2, and triple-negative primary tumors presented with a different subtype for the recurrent/metastatic tumors, respectively. No statistical difference in progression-free survival was observed according to the subtype of the recurrent or metastatic breast cancer (p > 0.05). Among 69 patients for whom treatment was adjusted after recurrence or metastasis, 66 patients remained recurrence-free during the follow-up period. Conclusion: For patients with early-stage breast cancer, the ER, PR, HER2, and Ki-67 expression profile for recurrent/metastatic tumors does not always match that of the primary tumor. After adjusting treatment according to the receptor expression in recurrent/ metastatic lesions, most patients remained progression-free during the follow-up period. C1 [Peng, Li] Peking Union Medical College Hospital, Department of Breast SurgeryBeijing, China. [Zhang, Zhen] Peking Union Medical College Hospital, Department of Breast SurgeryBeijing, China. [Zhao, Dachun] Peking Union Medical College Hospital, Department of PathologyBeijing, China. [Zhao, Jialin] Peking Union Medical College Hospital, Department of Breast SurgeryBeijing, China. [Mao, Feng] Peking Union Medical College Hospital, Department of Breast SurgeryBeijing, China. [Sun, Qiang] Peking Union Medical College Hospital, Department of Breast SurgeryBeijing, China. RP Mao, F (reprint author), Peking Union Medical College Hospital, Department of Breast Surgery, Beijing, China. EM mmc8941@me.com CR Sancho-Garnier H, Colonna M. Epidemiologie des cancers du sein. La Presse Medicale, 2019, 48(10):1076–84., DOI 10.1016/j.lpm.2019.09.022 Zhang ML, Peng P, Wu CX, Gong YM, Zhang SW, Chen WQ, et al. [Report of breast cancer incidence and mortality in China registry regions, 2008-2012]. Zhonghua Zhong Liu Za Zhi, 2019, 41(4):315–20., DOI 10.3760/cma.j.issn.0253- 3766.2019.04.013 Street W. Cancer facts & Figures 2019. Atlanta: Am Cancer Soc, 2018). 76. Akram M, Iqbal M, Daniyal M, Khan AU. Awareness and current knowledge of breast cancer. Biol Res, 2017, 50(1):33., DOI 10.1186/s40659-017-0140-9 Lambert AW, Pattabiraman DR, Weinberg RA. Emerging biological principles of metastasis. Cell, 2017, 168(4):670–91., DOI 10.1016/j.cell.2016.11.037 Ginsburg O, Yip CH, Brooks A, Cabanes A, Caleffi M, Dunstan Yataco JA, et al. Breast cancer early detection: a phased approach to implementation. Cancer, 2020, 126(S10):2379–93., DOI 10.1002/cncr.32887 Redig AJ,McAllister SS. Breast cancer as a systemic disease: a view of metastasis. J Intern Med, 2013, 274(2):113–26., DOI 10.1111/joim.12084 Anand AS, Velayudhan ST. Discordance of estrogen & progesterone receptors after neoadjuvant chemotherapy in breast cancer- an Indian study. Indian J Surg Oncol, 2016, 7(3):316–9., DOI 10.1007/s13193-016-0515-3 Ilgun S, Sarsenov D, Erdogan Z, Ordu C, Celebi F, Nur Pilanci K, et al. Receptor discordance rate and its effects on survival in primary and recurrent breast cancer patients. J Buon, 2016, 21(6):1425–32 PMID: 28039703. Kutomi G, Mizuguchi T, Satomi F, Maeda H, Shima H, Kimura Y, et al. Current status of the prognostic molecular biomarkers in breast cancer: a systematic review. Oncol Lett, 2017, 13(3):1491–8., DOI 10.3892/ol.2017.5609 Watkins EJ. Overview of breast cancer. Jaapa, 2019, 32(10):13–7., DOI 10.1097/ 01.JAA.0000580524.95733.3d Dieci MV, Barbieri E, Piacentini F, Ficarra G, Bettelli S, Dominici M, et al. Discordance in receptor status between primary and recurrent breast cancer has a prognostic impact: a single-institution analysis. Ann Oncol, 2013, 24(1): 101–8., DOI 10.1093/annonc/mds248 Berghuis AMS, van Deurzen CHM, Koffijberg H, Terstappen LWMM, Sleijfer S, IJzerman MJ. Real-world data on discordance between estrogen, progesterone, and HER2 receptor expression on diagnostic tumor biopsy versus tumor resection material. Breast Cancer Res Treat, 2019, 175(2): 451–8., DOI 10.1007/s10549-019-05141-y Matsumoto A, Jinno H, Murata T, Seki T, Takahashi M, Hayashida T, et al. Prognostic implications of receptor discordance between primary and recurrent breast cancer. Int J Clin Oncol, 2015, 20(4):701–8., DOI 10.1007/ s10147-014-0759-2 Yeung C, Hilton J, Clemons M, Mazzarello S, Hutton B, Haggar F, et al. Estrogen, progesterone, and HER2/neu receptor discordance between primary and metastatic breast tumours-a review. Cancer Metastasis Rev, 2016, 35(3): 427–37., DOI 10.1007/s10555-016-9631-3 Yao Z-x., Lu L-j., Wang R-j., Jin L-b., Liu S-c., Li H-y., et al. Discordance and clinical significance of ER, PR, and HER2 status between primary breast cancer and synchronous axillary lymph node metastasis. Med Oncol, 2014, 31(1):798., DOI 10.1007/s12032-013-0798-y Curigliano G, Burstein HJ, Winer EP, Gnant M, Dubsky P, Loibl S, et al. Deescalating and escalating treatments for early-stage breast cancer: the st. Gallen international expert consensus conference on the primary therapy of early breast cancer 2017. Ann Oncol, 2017, 28(8):1700–12., DOI 10.1093/annonc/ mdx308 Bond-Bero S. Filling the gap for early-stage breast cancer follow-up: an overview for primary care providers. J Midwifery Women’s Health, 2016, 61(2):166–76., DOI 10.1111/jmwh.12439 Giuliano AE, Edge SB, Hortobagyi GN. Eighth edition of the AJCC cancer staging manual: breast cancer. Ann Surg Oncol, 2018, 25(7):1783–5., DOI 10. 1245/s10434-018-6486-6 Hammond MEH, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer, unabridged version). Arch Pathol Lab Med, 2010, 134(7):e48–e72., DOI 10.5858/134.7.e48 Wolff AC, Hammond MEH, Hicks DG, Dowsett M, McShane LM, Allison KH, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. Jco, 2013, 31(31):3997–4013., DOI 10.1200/jco.2013.50.9984 Zhao J, Wu R, Au A, Marquez A, Yu Y, Shi Z. Determination of HER2 gene amplification by chromogenic in situ hybridization, CISH, in archival breast carcinoma. Mod Pathol, 2002, 15(6):657–65., DOI 10.1038/modpathol.3880582 Kobayashi T, Iwaya K, Moriya T, Yamasaki T, Tsuda H, Yamamoto J, et al. A simple immunohistochemical panel comprising 2 conventional markers, Ki67 and p53, is a powerful tool for predicting patient outcome in luminal-type breast cancer. BMC Clin Pathol, 2013, 13:5., DOI 10.1186/1472-6890-13-5 Fulawka L, Halon A. Ki-67 evaluation in breast cancer: the daily diagnostic practice. Indian J Pathol Microbiol, 2017, 60(2):177–84., DOI 10.4103/ijpm. Ijpm_732_15 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, et al. Personalizing the treatment of women with early breast cancer: highlights of the st gallen international expert consensus on the primary therapy of early breast cancer 2013. Ann Oncol, 2013, 24(9): 2206–23., DOI 10.1093/annonc/mdt303 Hess LM, Brnabic A, Mason O, Lee P, Barker S. Relationship between progression-free survival and overall survival in randomized clinical trials of targeted and biologic agents in Oncology. J Cancer, 2019, 10(16):3717–27., DOI 10.7150/jca.32205 Aurilio G, Disalvatore D, Pruneri G, Bagnardi V, Viale G, Curigliano G, et al. A meta-analysis of oestrogen receptor, progesterone receptor and human epidermal growth factor receptor 2 discordance between primary breast cancer and metastases. Eur J Cancer, 2014, 50(2):277–89., DOI 10.1016/j. ejca.2013.10.004 Medeiros B, Allan AL. Molecular mechanisms of breast cancer metastasis to the lung: clinical and experimental perspectives. Ijms, 2019, 20(9):2272., DOI 10.3390/ijms20092272 Alzubi MA, Turner TH, Olex AL, Sohal SS, Tobin NP, Recio SG, et al. Separation of breast cancer and organ microenvironment transcriptomes in metastases. Breast Cancer Res, 2019, 21(1):36., DOI 10.1186/s13058-019- 1123-2 Ng CKY, Bidard F-C, Piscuoglio S, Geyer FC, Lim RS, de Bruijn I, et al. Genetic heterogeneity in therapy-naive synchronous primary breast cancers and their metastases. Clin Cancer Res, 2017, 23(15):4402–15., DOI 10.1158/1078-0432. Ccr-16-3115 Nishimura R, Osako T, Okumura Y, Tashima R, Toyozumi Y, Arima N. Changes in the ER, PgR, HER2, p53 and Ki-67 biological markers between primary and recurrent breast cancer: discordance rates and prognosis. World J Surg Onc, 2011, 9:131., DOI 10.1186/1477-7819-9-131 Yi ZB, Yu P, Zhang S, Wang WN, Han YQ, Ouyang QC, et al. Profile and outcome of receptor conversion in breast cancer metastases: a nation-wide multicenter epidemiological study. Int J Cancer, 2020, 148:692., DOI 10.1002/ ijc.33227 Rossi S, Basso M, Strippoli A, Dadduzio V, Cerchiaro E, Barile R, et al. Hormone receptor status and HER2 expression in primary breast cancer compared with synchronous axillary metastases or recurrent metastatic disease. Clin Breast Cancer, 2015, 15(5):307–12., DOI 10.1016/j.clbc.2015. 03.010 Jensen JD, Knoop A, Ewertz M, Laenkholm A-V ER, HER2, and TOP2A expression in primary tumor, synchronous axillary nodes, and asynchronous metastases in breast cancer. Breast Cancer Res Treat, 2012, 132(2):511–21., DOI 10.1007/s10549-011-1610-3 Liu J, Deng H, Jia W, Zeng Y, Rao N, Li S, et al. Comparison of ER/PR and HER2 statuses in primary and paired liver metastatic sites of breast carcinoma in patients with or without treatment. J Cancer Res Clin Oncol, 2012, 138(5): 837–42., DOI 10.1007/s00432-012-1150-1 Strien L, Leidenius M, von Smitten K, Heikkila P. Concordance between HER- 2 and steroid hormone receptor expression between primary breast cancer, sentinel node metastases, and isolated tumor cells. Pathol - Res Pract, 2010, 206(4):253–8., DOI 10.1016/j.prp.2009.12.006 Erdem GU, Altundag K, Ozdemir NY, Sahin S, Demirci NS, Karatas F, et al. Comparative study of receptor discordance between primary and corresponding metastatic lesions in breast cancer. J Buon, 2017a, 22(2): 365–76. Arslan C, Sari E, Aksoy S, Altundag K. Variation in hormone receptor and HER-2 status between primary and metastatic breast cancer: review of the literature. Expert Opin Ther Targets, 2011, 15(1):21–30., DOI 10.1517/ 14656566.2011.537260 Fragomeni SM, Sciallis A, Jeruss JS. Molecular subtypes and local-regional control of breast cancer. Surg Oncol Clin North America, 2018, 27(1):95–120., DOI 10.1016/j.soc.2017.08.005 Wang G-S, Zhu H, Bi S-J. Pathological features and prognosis of different molecular subtypes of breast cancer. Mol Med Rep, 2012, 6(4):779–82., DOI 10. 3892/mmr.2012.981 Yeo SK, Guan J-L Breast cancer: multiple subtypes within a tumor?. Trends Cancer, 2017, 3(11):753–60., DOI 10.1016/j.trecan.2017.09.001 Ellsworth RE, Blackburn HL, Shriver CD, Soon-Shiong P, Ellsworth DL. Molecular heterogeneity in breast cancer: state of the science and implications for patient care. Semin Cel Dev Biol, 2017, 64:65–72., DOI 10.1016/ j.semcdb.2016.08.025 Beca F, Polyak K. Intratumor heterogeneity in breast cancer. Adv Exp Med Biol, 2016, 882:169–89., DOI 10.1007/978-3-319-22909-6_7 Kuukasjarvi T, Karhu R, Tanner M, Kahkonen M, Schaffer A, Nupponen N, et al. Genetic heterogeneity and clonal evolution underlying development of asynchronous metastasis in human breast cancer. Cancer Res, 1997, 57(8):1597–604. Manjili MH, Kmieciak M. Does HER-2/neu antigen loss in metastatic breast tumors occur under immune pressure?. Int J Cancer, 2008, 123(6):1476–7., DOI 10.1002/ijc.23663 Rasbridge S, Gillett C, Seymour A-M, Patel K, Richards M, Rubens R, et al. The effects of chemotherapy on morphology, cellular proliferation, apoptosis and oncoprotein expression in primary breast carcinoma. Br J Cancer, 1994, 70(2): 335–41., DOI 10.1038/bjc.1994.303 Cardoso F, Costa A, Senkus E, AaproM, Andre F, Barrios CH, et al. Correction to: 3rd ESO-ESMO international consensus guidelines for advanced breast cancer, ABC 3). Ann Oncol, 2017, 28(12):3111., DOI 10.1093/annonc/mdx036 Cardoso F, Senkus E, Costa A, Papadopoulos E, Aapro M, Andre F, et al. 4th ESO-ESMO international consensus guidelines for advanced breast cancer, ABC 4). Ann Oncol, 2018, 29(8):1634–57., DOI 10.1093/annonc/mdy192 Pusztai L, Viale G, Kelly CM, Hudis CA. Estrogen and HER-2 receptor discordance between primary breast cancer and metastasis. The Oncologist, 2010, 15(11):1164–8., DOI 10.1634/theoncologist.2010-0059 Seyfried TN, Huysentruyt LC. On the origin of cancer metastasis. Crit Rev Oncog, 2013, 18(1-2):43–73., DOI 10.1615/critrevoncog.v18.i1-2.40 Moller EK, Kumar P, Voet T, Peterson A, Van Loo P, Mathiesen RR, et al. Next-generation sequencing of disseminated tumor cells. Front Oncol, 2013, 3: 320., DOI 10.3389/fonc.2013.00320 Sperduto PW, Mesko S, Li J, Cagney D, Aizer A, Lin NU, et al. Estrogen/ progesterone receptor and HER2 discordance between primary tumor and brain metastases in breast cancer and its effect on treatment and survival. Neuro Oncol, 2020, 22(9):1359–67., DOI 10.1093/neuonc/ noaa025 Fehm T, Muller V, Aktas B, Janni W, Schneeweiss A, Stickeler E, et al. HER2 status of circulating tumor cells in patients with metastatic breast cancer: a prospective, multicenter trial. Breast Cancer Res Treat, 2010, 124(2):403–12., DOI 10.1007/s10549-010-1163-x Sieuwerts AM, Mostert B, Bolt-de Vries J, Peeters D, de Jongh FE, Stouthard JML, et al. mRNA and microRNA expression profiles in circulating tumor cells and primary tumors of metastatic breast cancer patients. Clin Cancer Res, 2011, 17(11):3600–18., DOI 10.1158/1078-0432.Ccr-11-0255 Prat A, Fan C, Fernandez A, Hoadley KA, Martinello R, Vidal M, et al. Response and survival of breast cancer intrinsic subtypes following multiagent neoadjuvant chemotherapy. BMC Med, 2015, 13:303., DOI 10.1186/ s12916-015-0540-z Zardavas D, Irrthum A, Swanton C, Piccart M. Clinical management of breast cancer heterogeneity. Nat Rev Clin Oncol, 2015, 12(7):381–94., DOI 10.1038/ nrclinonc.2015.73 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 599894 EP 599906 DI 10.3389/pore.2021.599894 PG 13 ER PT J AU Hakim, AS Gabal, HAH AF Hakim, Adel Sarah Gabal, Hassan Abou Hoda TI Diagnostic Utility of BAP1, EZH2 and Survivin in Differentiating Pleural Epithelioid Mesothelioma and Reactive Mesothelial Hyperplasia: Immunohistochemical Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mesothelioma; reactive mesothelial hyperplasia; BAP1; EZH2; survivin; immunohistochemistry ID mesothelioma; reactive mesothelial hyperplasia; BAP1; EZH2; survivin; immunohistochemistry AB Background: Epithelioid mesothelioma (EM) is the commonest subtype of malignant pleural mesothelioma. Its histopathological discrimination from reactive mesothelial hyperplasia (RMH) could be challenging. Thus, an immunohistochemical panel is mandatory for better discrimination. BAP1 is a newly identified diagnostic marker whose loss is specific to malignant mesothelioma. EZH2 overexpression is reported in different cancers, but its relation to BAP1 in malignant mesothelioma has not been fully understood. Survivin expression is said to be significantly higher in EM than in nonneoplastic pleural tissue, but its diagnostic utility as an immunohistochemical marker has not been thoroughly investigated in this field. To the best of our knowledge, no previous studies have been conducted to assess the diagnostic accuracy of the combined use of these three nuclear markers (BAP1, EZH2 and Survivin) in discriminating pleural EM from RMH. Methods: This retrospective study includes two groups: 81 cases of pleural EM and 67 cases of RMH, retrieved from the archives of Pathology Department of Ain Shams University Hospitals and Ain-Shams University Specialized Hospital during the period from January 2016 to December 2019. An immunohistochemical study was performed using BAP1, EZH2 and Survivin antibodies. Results: There were highly statistically significant relations between study groups as regards the studied markers (p = 0.001 for each). The specificity was 100% for all combinations of immunohistochemical markers. Sensitivity of any combination of the immunohistochemical markers used in this study was found to be higher than the sensitivity of any of these markers used individually. The combination of all three markers showed the highest diagnostic accuracy (95.9%) and the highest sensitivity (92.6%). However, the combination of Survivin and EZH2 yielded the same diagnostic accuracy and sensitivity. Conclusion: Adding EZH2, Survivin and BAP1 to the diagnostic IHC panel for differentiating pleural EM and RMH could enhance diagnostic sensitivity. Moreover, Survivin is a potentially promising marker in this context, especially when combined with EZH2. C1 [Hakim, Adel Sarah] Ain Shams Faculty of Medicine, Department of PathologyCairo, Egypt. [Gabal, Hassan Abou Hoda] Ain Shams Faculty of Medicine, Department of PathologyCairo, Egypt. RP Hakim, AS (reprint author), Ain Shams Faculty of Medicine, Department of Pathology, Cairo, Egypt. EM sarahadel2003@yahoo.com CR Churg A, Sheffield BS, Galateau-Salle F. New markers for separating benign from malignant mesothelial proliferations: are we there yet? Arch Pathol Lab Med, 2016). 140(4):318–21., DOI 10.5858/arpa.2015-0240-SA Testa JR, CheungM, Pei J, Below JE, Tan Y, Sementino E., et al. Germline BAP1 mutations predispose to malignant mesothelioma. Nat Genet, 2011). 43(10): 1022–5., DOI 10.1038/ng.912 Bott M, BrevetM, Taylor BS, Shimizu S, Ito T,Wang L., et al. The nuclear deubiquitinase BAP1 is commonly inactivated by somatic mutations and 3p21.1 losses in malignant pleural mesothelioma. Nat Genet, 2011). 43(7):668–72., DOI 10.1038/ng.855 Shinozaki-Ushiku A, Ushiku T, Morita S, Anraku M, Nakajima J, Fukayama M. Diagnostic utility of BAP1 and EZH2 expression in malignant mesothelioma. Histopathology, 2017). 70:722–33., DOI 10.1111/his.13123 LaFave LM, Beguelin W, Koche R, Teater M, Spitzer B, Chramiec A., et al. Loss of BAP1 function leads to EZH2-dependent transformation. Nat Med, 2015). 21(11):1344–9., DOI 10.1038/nm.3947 Kim KH, Roberts CW. Targeting EZH2 in cancer. Nat Med, 2016). 22(2): 128–34., DOI 10.1038/nm.4036 Yamaguchi H, Hung MC. Regulation and role of EZH2 in cancer. Cancer Res Treat, 2014). 46(3):209–22., DOI 10.4143/crt.2014.46.3.209 Chen X, Duan N, Zhang C, Zhang W. Survivin and tumorigenesis: molecular mechanisms and therapeutic strategies. J Cancer, 2016). 7:314–23., DOI 10.7150/ jca.13332 Kushitani K, Amatya VJ, Mawas AS, Miyata Y, Okada M, Takeshima Y. Use of anti-noxa antibody for differential diagnosis between epithelioid mesothelioma and reactive mesothelial hyperplasia. Pathobiology, 2016). 83(1):33–40., DOI 10. 1159/000442092 Nicholson AG, Sauter JL, Nowak AK, Kindler HL, Gill RR, Remy-Jardin M., et al. EURACAN/IASLC proposals for updating the histologic classification of pleural mesothelioma: towards a more multidisciplinary approach. J Thorac Oncol, 2020). 15(1):29–49., DOI 10.1016/j.jtho.2019.08.2506 Husain AN, Colby TV, Ordonez NG, Allen TC, Attanoos RL, Beasley MB., et al. Guidelines for pathologic diagnosis of malignant mesothelioma 2017 update of the consensus statement from the international mesothelioma interest group. Arch Pathol Lab Med, 2018). 142(1):89–108., DOI 10.5858/arpa. 2017-0124-RA Hsu SM, Raine L, Fanger H. Use of avidin-biotin-peroxidase complex, ABC, in immunoperoxidase techniques: a comparison between ABC and unlabeled antibody, PAP, procedures. J Histochem Cytochem, 1981). 29:577–80., DOI 10. 1177/29.4.6166661 13 Vassallo J, Al Saati T, Gascoyne RD, Welsh K, Reed JC, Brousset P., et al. Immunoexpression of Survivin in non-neoplastic lymphoid tissues and malignant lymphomas using a new monoclonal antibody reactive on paraffin sections. J Hematop, 2010). 3:3–9., DOI 10.1007/s12308-009-0054-8 14. Farquhar N, Thornton S, Coupland SE, Coulson JM, Sacco JJ, Krishna Y., et al. Patterns of BAP1 protein expression provide insights into prognostic significance and the biology of uveal melanoma. J Pathol Clin Res, 2018). 4:26–38., DOI 10.1002/cjp2.86 15. Yoshimura M, Kinoshita Y, Hamasaki M, Matsumoto S, Hida T, Oda Y., et al. Highly expressed EZH2 in combination with BAP1 and MTAP loss, as detected by immunohistochemistry, is useful for differentiating malignant pleural mesothelioma from reactive mesothelial hyperplasia. Lung Cancer, 2019). 130:187–93., DOI 10.1016/j.lungcan.2019.02.004 16. Kushitani K, Amatya VJ, Mawas AS, Suzuki R, Miyata Y, Okada M., et al. Utility of survivin, BAP1, and ki-67 immunohistochemistry in distinguishing epithelioid mesothelioma from reactive mesothelial hyperplasia. Oncol Lett, 2018). 15:3540–7., DOI 10.3892/ol.2018.7765 17. Rossini M, Rizzo P, Bononi I, Clementz A, Ferrari R, Martini F., et al. New perspectives on diagnosis and therapy of malignant pleural mesothelioma. Front Oncol, 2018). 8:91., DOI 10.3389/fonc.2018.00091 18. Hida T, HamasakiM,Matsumoto S, Sato A, Tsujimura T, Kawahara K., et al. BAP1 immunohistochemistry and p16 FISH results in combination provide higher confidence in malignant pleural mesothelioma diagnosis: ROC analysis of the two tests. Pathol Int, 2016). 66(10):563–70., DOI 10.1111/ pin.12453 19. Hwang HC, Sheffield BS, Rodriguez S, Thompson K, Tse CH, Gown AM., et al. Utility of BAP1 immunohistochemistry and p16, CDKN2A, FISH in the diagnosis of malignant mesothelioma in effusion cytology specimens. Am J Surg Pathol, 2016). 40(1):120–6., DOI 10.1097/PAS. 0000000000000529 20. Cigognetti M, Lonardi S, Fisogni S, Balzarini P, Pellegrini V, Tironi A., et al. BAP1, BRCA1-associated protein 1, is a highly specific marker for differentiating mesothelioma from reactive mesothelial proliferations. Mod Pathol, 2015). 28(8):1043–57., DOI 10.1038/modpathol.2015.65 21. Ang PP, Tan GC, Karim N, Wong YP Diagnostic value of the EZH2 immunomarker in malignant effusion cytology. Acta Cytol, 2020). 64: 248–55., DOI 10.1159/000501406 22. Meerang M, Berard K, Friess M, Bitanihirwe BK, Soltermann A, Vrugt B., et al. Low merlin expression and high survivin labeling index are indicators for poor prognosis in patients with malignant pleural mesothelioma. Mol Oncol, 2016). 10(8):1255–65., DOI 10.1016/j.molonc.2016.06.005 23. Hmeljak J, Erˇculj N, Dolˇzan V, Piˇzem J, Kern I, Kovaˇc V., et al. Is survivin expression prognostic or predictive in malignant pleural mesothelioma? Virchows Arch, 2013). 462(3):315–21., DOI 10.1007/s00428-013-1373-9 24. Bilecz A, Stockhammer P, Theegarten D, Kern I, Jakopovic M, Samarzija M., et al. Comparative analysis of prognostic histopathologic parameters in subtypes of epithelioid pleural mesothelioma. Histopathology, 2020). 77(1): 55–66., DOI 10.1111/his.14105 25. Chapel DB, Husain AN, Krausz T. Immunohistochemical evaluation of nuclear 5-hydroxymethylcytosine, 5-hmC, accurately distinguishes malignant pleural mesothelioma from benign mesothelial proliferations. Mod Pathol, 2019). 32(3):376–86., DOI 10.1038/s41379-018-0159-7 26. Sandeck HP, Roe OD, Kjaerheim K, Willen H, Larsson E. Re-evaluation of histological diagnoses of malignant mesothelioma by immunohistochemistry. Diagn Pathol, 2010). 5:47., DOI 10.1186/1746-1596-5-47 27. Lin F, Chen Z. Standardization of diagnostic immunohistochemistry: literature review and geisinger experience. Arch Pathol Lab Med, 2014). 138(12): 1564–77., DOI 10.5858/arpa.2014-0074-RA 28. Chui J, Singh A, Gill AJ Loss of BAP1 expression is very rare in breast carcinoma. Pathology, 2017). 49(5):557–60., DOI 10.1016/j.pathol.2017.03.008 29. Tayao M, Andrici J, Farzin M, Clarkson A, Sioson L, Watson N., et al. Loss of BAP1 expression is very rare in pancreatic ductal adenocarcinoma. PLoS One, 2016). 11:e0150338., DOI 10.1371/journal.pone.0150338 30. Andrici J, Parkhill TR, Jung J, Wardell KL, Verdonk B, Singh A., et al. Loss of expression of BAP1 is very rare in non-small cell lung carcinoma. Pathology, 2016). 48(4):336–40., DOI 10.1016/j.pathol.2016.03.005 31. Chui J, Verdonk B, Gill AJ. Loss of BAP1 expression occurs rarely to never in colorectal adenocarcinoma. Appl Immunohistochem Mol Morphol, 2018). 26(9):e91–e92., DOI 10.1097/PAI.0000000000000506 32. Behrens C, Solis LM, Lin H, Yuan P, Tang X, Kadara H., et al. EZH2 protein expression associates with the early pathogenesis, tumor progression, and prognosis of non-small cell lung carcinoma. Clin Cancer Res, 2013). 19(23): 6556–65., DOI 10.1158/1078-0432.CCR-12-3946 33. Han T, Jiao F, Hu H, Yuan C,Wang L, Jin ZL., et al. EZH2 promotes cell migration and invasion but not alters cell proliferation by suppressing E-cadherin, partly through association with MALAT-1 in pancreatic cancer. Oncotarget, 2016). 7(10):11194–207., DOI 10.18632/oncotarget. 7156 34. Abdel Raouf SM, Ibrahim TR, Abdelaziz LA, Farid MI, Mohamed SY. Prognostic value of TWIST1 and EZH2 expression in colon cancer. J Gastrointest Cancer, 2019). 52:90–98., DOI 10.1007/s12029-019-00344-4 35. Jang SH, Lee JE, Oh MH, Lee JH, Cho HD, Kim KJ., et al. High EZH2 protein expression is associated with poor overall survival in patients with luminal A breast cancer. J Breast Cancer, 2016). 19(1):53–60., DOI 10.4048/jbc.2016.19. 1.53 36. Chu JS, Shew JY,Huang CS. Immunohistochemical analysis of survivin expression in primary breast cancers. J Formos Med Assoc, 2004). 103(12):925–31. 37. Satoh K, Kaneko K, Hirota M, Masamune A, Satoh A, Shimosegawa T. Expression of survivin is correlated with cancer cell apoptosis and is involved in the development of human pancreatic duct cell tumors. Cancer, 2001). 92(2):271–8., DOI 10.1002/1097-0142(20010715)92:2<271::aidcncr1319> 3.0.co;2-0 38. Atikcan S, Unsal E, Demirag F, Koksal D, Yilmaz A. Correlation between survivin expression and prognosis in non-small cell lung cancer. Respir Med, 2006). 100(12):2220–6., DOI 10.1016/j.rmed.2006.02.031 39. Jakubowska K, Pryczynicz A, Dymicka-Piekarska V, Famulski W, Guzi´nska- Ustymowicz K. Immunohistochemical expression and serum level of survivin protein in colorectal cancer patients. Oncol Lett, 2016). 12:3591–7., DOI 10. 3892/ol.2016.5075 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 600073 EP 600082 DI 10.3389/pore.2021.600073 PG 10 ER PT J AU Qi, A Ju, M Liu, Y Bi, J Wei, Q He, M Wei, M Zhao, L AF Qi, Aoshuang Ju, Mingyi Liu, Yinfeng Bi, Jia Wei, Qian He, Miao Wei, Minjie Zhao, Lin TI Development of a Novel Prognostic Signature Based on Antigen Processing and Presentation in Patients with Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE antigen processing and presentation; breast cancer; cell biology; prognostic; survival ID antigen processing and presentation; breast cancer; cell biology; prognostic; survival AB Background: Complex antigen processing and presentation processes are involved in the development and progression of breast cancer (BC). A single biomarker is unlikely to adequately reflect the complex interplay between immune cells and cancer; however, there have been few attempts to find a robust antigen processing and presentation-related signature to predict the survival outcome of BC patients with respect to tumor immunology. Therefore, we aimed to develop an accurate gene signature based on immune-related genes for prognosis prediction of BC. Methods: Information on BC patients was obtained from The Cancer Genome Atlas. Gene set enrichment analysis was used to confirm the gene set related to antigen processing and presentation that contributed to BC. Cox proportional regression, multivariate Cox regression, and stratified analysis were used to identify the prognostic power of the gene signature. Differentially expressed mRNAs between high- and low-risk groups were determined by KEGG analysis. Results: A three-gene signature comprising HSPA5 (heat shock protein family A member 5), PSME2 (proteasome activator subunit 2), and HLA-F (major histocompatibility complex, class I, F) was significantly associated with OS. HSPA5 and PSME2 were protective (hazard ratio (HR) < 1), and HLA-F was risky (HR > 1). Risk score, estrogen receptor (ER), progesterone receptor (PR) and PD-L1 were independent prognostic indicators. KIT and ACACB may have important roles in the mechanismby which the gene signature regulates prognosis of BC. Conclusion: The proposed three-gene signature is a promising biomarker for estimating survival outcomes in BC patients. C1 [Qi, Aoshuang] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Ju, Mingyi] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Liu, Yinfeng] The First Hospital of Qinhuangdao, Department of Breast SurgeryQinhuangdao, China. [Bi, Jia] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Wei, Qian] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [He, Miao] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Wei, Minjie] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. [Zhao, Lin] China Medical University, School of Pharmacy, Department of PharmacologyShenyang, China. RP Zhao, L (reprint author), China Medical University, School of Pharmacy, Department of Pharmacology, Shenyang, China. EM lzhao@cmu.edu.cn CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J Clinicians, 2018). 68(6):394–424., DOI 10.3322/caac.21492 Untch M, Konecny GE, Paepke S, von Minckwitz G Current and future role of neoadjuvant therapy for breast cancer. The Breast, 2014). 23(5):526–37., DOI 10. 1016/j.breast.2014.06.004 Gemignani ML, Armstrong DK Breast cancer. Gynecol Oncol, 2014). 132(2): 264–7., DOI 10.1016/j.ygyno.2014.01.022 Castaneda SA, Strasser J Updates in the treatment of breast cancer with radiotherapy. Surg Oncol Clin North America, 2017). 26(3):371–82., DOI 10. 1016/j.soc.2017.01.013 Harbeck N, Sotlar K, Wuerstlein R, Doisneau-Sixou S Molecular and protein markers for clinical decision making in breast cancer: today and tomorrow. Cancer Treat Rev, 2014). 40(3):434–44., DOI 10.1016/j.ctrv.2013.09.014 Blackley EF, Loi S Targeting immune pathways in breast cancer: review of the prognostic utility of TILs in early stage triple negative breast cancer, TNBC). The Breast, 2019). 48(Suppl. 1):S44–S48., DOI 10.1016/S0960-9776(19)31122-1 Sugie T Immunotherapy for metastatic breast cancerHammerl D, Smid M, Timmermans AM, Sleijfer S, Martens JWM, Debets R. Breast cancer genomics and immuno-oncological markers to guide immune therapies. Chin Clin Oncol.Semin Cancer Biol, 2018). 7(3Pt 2):28178–28., DOI 10.21037/cco.2018.05.05 9. de la Cruz-Merino L, Chiesa M, Caballero R, Rojo F, Palazon N, Carrasco FH, et al. Breast cancer immunology and immunotherapy. Int Rev Cel Mol Biol, 2017). 331:1–53., DOI 10.1016/bs.ircmb.2016.09.008 10. Kasahara M, Flajnik MF, Takahama Y Biology, evolution, and history of antigen processing and presentation: immunogenetics special issue 2019. Immunogenetics, 2019). 71(3):137–9., DOI 10.1007/s00251-019-01107-y 11. Cresswell P A personal retrospective on the mechanisms of antigen processing. Immunogenetics, 2019). 71(3):141–60., DOI 10.1007/s00251-018-01098-2 12. Miller MA, Ganesan APV, Luckashenak N, Mendonca M, Eisenlohr LC Endogenous antigen processing drives the primary CD4+ T cell response to influenza. Nat Med, 2015). 21(10):1216–22., DOI 10.1038/nm.3958 13. Wan X, Unanue ER Antigen recognition in autoimmune diabetes: a novel pathway underlying disease initiation. Precis Clin Med, 2018). 1(3):102–10., DOI 10.1093/pcmedi/pby015 14. Reeves E, James E Antigen processing and immune regulation in the response to tumours. Immunology, 2017). 150(1):16–24., DOI 10.1111/imm.12675 15. Schuijs MJ, Hammad H, Lambrecht BN Professional and ’amateur’ antigenpresenting cells in type 2 immunity. Trends Immunol, 2019). 40(1):22–34., DOI 10.1016/j.it.2018.11.001 16. Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A, et al. CD4+ follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest, 2013). 123(7):2873–92., DOI 10.1172/JCI67428 17. Curtis C, Shah SP, Shah SP, Chin S-F, Turashvili G, Rueda OM, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature, 2012). 486(7403):346–52., DOI 10.1038/nature10983 18. Carbognin L, Miglietta F, Paris I, Dieci MV Prognostic and predictive implications of PTEN in breast cancer: unfulfilled promises but intriguing perspectives. Cancers, 2019). 11(9):1401., DOI 10.3390/cancers11091401 19. Li Y, Jiao Y, Luo Z, Li Y, Liu Y High peroxidasin-like expression is a potential and independent prognostic biomarker in breast cancer. Medicine, 2019). 98(44):e17703., DOI 10.1097/MD.0000000000017703 20. Xu W, Wang Y, Wang Y, Lv S, Xu X, Dong X Screening of differentially expressed genes and identification of NUF2 as a prognostic marker in breast cancer. Int J Mol Med, 2019). 15(4):1567–77., DOI 10.4049/jimmunol.1300080 21. Chen W-x., Yang L-g., Xu L-y., Cheng L, Qian Q, Sun L, et al. Bioinformatics analysis revealing prognostic significance of RRM2 gene in breast cancer. Biosci Rep, 2019). 39(4)., DOI 10.1042/BSR20182062 22. Dai J-b., Zhu B, Lin W-j., Gao H-y., Dai H, Zheng L, et al. Identification of prognostic significance of BIRC5 in breast cancer using integrative bioinformatics analysis. Biosci Rep, 2020). 40(2)., DOI 10.1042/BSR20193678 23. Cheng L, Huang Y-Z, Chen W-X, Shi L, Li Z, Zhang X, et al. Cell division cycle proteinising prognostic biomarker of breast cancer. Biosci Rep, 2020). 40(5)., DOI 10.1042/BSR20191227 24. Cheng W, Ren X, Zhang C, Cai J, Liu Y, Han S, et al. Bioinformatic profiling identifies an immune-related risk signature for glioblastoma. Neurology, 2016). 86:2226., DOI 10.1212/WNL.0000000000002770 25. Chen Y-L, Ge G-J, Qi C, Wang H, Wang H-L, Li L-Y, et al. A five-gene signature may predict sunitinib sensitivity and serve as prognostic biomarkers for renal cell carcinoma. J Cel Physiol, 2018). 233(10):6649–60., DOI 10.1002/ jcp.26441 26. Guo W, Chen X, Zhu L, Wang Q A six-mRNA signature model for the prognosis of head and neck squamous cell carcinoma. Oncotarget, 2017). 8(55):94528–38., DOI 10.18632/oncotarget.21786 27. Silsirivanit A Glycosylation markers in cancer. Adv Clin Chem, 2019). 89: 189–213., DOI 10.1016/bs.acc.2018.12.005 28. Gunawardene A, Dennett E, Larsen P Prognostic value of multiple cytokine analysis in colorectal cancer: a systematic review. J Gastrointest Oncol, 2018). 10(1):134–43., DOI 10.21037/jgo.2018.07.11 29. Di Gioia D, Blankenburg I, Nagel D, Heinemann V, Stieber P Tumor markers in the early detection of tumor recurrence in breast cancer patients: CA 125, CYFRA 21-1, HER2 shed antigen, LDH and CRP in combination with CEA and CA 15-3. Clinica Chim Acta, 2016). 461:1–7., DOI 10.1016/j.cca.2016. 07.014 30. Huang X, Li X, Xie X, Ye F, Chen B, Song C, et al. High expressions of LDHA and AMPK as prognostic biomarkers for breast cancer. The Breast, 2016). 30: 39–46., DOI 10.1016/j.breast.2016.08.014 31. Al-Maghrabi J, Al-Sakkaf K, Qureshi IA, Butt NS, Damnhory L, Elshal M, et al. AMPK expression patterns are significantly associated with poor prognosis in breast cancer patients. Ann Diagn Pathol, 2017). 29:62–7., DOI 10.1016/j. anndiagpath.2017.05.012 32. Vesely MD, Schreiber RD Cancer immunoediting: antigens, mechanisms, and implications to cancer immunotherapy. Ann N.Y Acad Sci, 2013). 1284(1):1–5., DOI 10.1111/nyas.12105 33. Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ Natural innate and adaptive immunity to cancer. Annu Rev Immunol, 2011). 29:235–71., DOI 10.1146/annurev-immunol-031210-101324 34. Duwell P, Heidegger S, Kobold S Innate immune stimulation in cancer therapy. Hematology/Oncology Clin North America, 2019). 33(2):215–31., DOI 10.1016/j.hoc.2018.12.002 35. Mirza AH, Thomas G, Ottensmeier CH, King EV Importance of the immune system in head and neck cancer. Head Neck, 2019). 41(8):2789–800., DOI 10. 1002/hed.25716 36. Accolla RS, Lombardo L, Abdallah R, Raval G, Forlani G, Tosi G Boosting the MHC class II-restricted tumor antigen presentation to CD4+ T helper cells: a critical issue for triggering protective immunity and re-orienting the tumor microenvironment toward an anti-tumor state. Front Oncol, 2014). 4:32., DOI 10.3389/fonc.2014.00032 37. de Visser KE, Eichten A, Coussens LM Paradoxical roles of the immune system during cancer development. Nat Rev Cancer, 2006). 6(1):24–37., DOI 10.1038/ nrc1782 38. Raulet DH Interplay of natural killer cells and their receptors with the adaptive immune response. Nat Immunol, 2004). 5(10):996–1002., DOI 10.1038/ni1114 39. Hamerman JA, Ogasawara K, Lanier LL NK cells in innate immunity. Curr Opin Immunol, 2005). 17(1):29–35., DOI 10.1016/j.coi.2004.11.001 40. Agrawal S, Reemtsma K, Bagiella E, Oluwole SF, Braunstein NS Role of TAP-1 and/or TAP-2 antigen presentation defects in tumorigenicity of mouse melanoma. Cell Immunol, 2004). 228(2):130–7., DOI 10.1016/j.cellimm.2004. 04.006 41. Geng J, Zaitouna AJ, Raghavan M Selected HLA-B allotypes are resistant to inhibition or deficiency of the transporter associated with antigen processing, TAP). Plos Pathog, 2018). 14(7):e1007171., DOI 10.1371/journal.ppat.1007171 42. Lee AS Glucose-regulated proteins in cancer: molecular mechanisms and therapeutic potential. Nat Rev Cancer, 2014). 14(4):263–76., DOI 10.1038/ nrc3701 43. Respondek D, Voss M, Kuhlewindt I, Klingel K, Kruger E, Beling A PA28 modulates antigen processing and viral replication during coxsackievirus B3 infection. PLoS One, 2017). 12(3):e0173259., DOI 10.1371/journal.pone. 0173259 44. Huang Q, Huang Q, Lin W, Lin J, Lin X Potential roles for PA28β in gastric adenocarcinoma development and diagnosis. J Cancer Res Clin Oncol, 2010). 136(8):1275–82., DOI 10.1007/s00432-010-0778-y 45. Zheng D-L, Huang Q-L, Zhou F, Huang Q-J, Lin J-Y, Lin X PA28β regulates cell invasion of gastric cancer via modulating the expression of chloride intracellular channel 1. J Cel Biochem., 2011). 113(5):a–n., DOI 10.1002/jcb. 24022 46. Lin A, Zhang X, Ruan Y-Y, Wang Q, Zhou W-J, Yan W-H HLA-F expression is a prognostic factor in patients with non-small-cell lung cancer. Lung Cancer, 2011). 74(3):504–9., DOI 10.1016/j.lungcan.2011.04.006 47. Goodridge JP, Lee N, Burian A, Pyo C-W, Tykodi SS,Warren EH, et al. HLA-F and MHC-I open conformers cooperate in a MHC-I antigen cross-presentation pathway. J.I., 2013). 191(4):1567–77., DOI 10.4049/jimmunol.1300080 48. Lunemann S, Schobel A, Kah J, Fittje P, Holzemer A, Langeneckert AE, et al. Interactions between KIR3DS1 and HLA-F activate natural killer cells to control HCV replication in cell culture. Gastroenterology, 2018). 155(5): 1366–71., DOI 10.1053/j.gastro.2018.07.019 49. Shien T, Iwata H Adjuvant and neoadjuvant therapy for breast cancer. Jpn J Clin Oncol, 2020). 50(3):225–9., DOI 10.1093/jjco/hyz213 50. Litton JK, Scoggins ME, Hess KR, Adrada BE, Murthy RK, Damodaran S, et al. Neoadjuvant talazoparib for patients with operable breast cancer with a germline BRCA pathogenic variant. Jco, 2020). 38(5):388., DOI 10.1200/JCO. 19.01304 51. Rossi L,McCartney A, Risi E, De Santo I, Migliaccio I, Malorni L, et al. Cyclindependent kinase 4/6 inhibitors in neoadjuvant endocrine therapy of hormone receptor-positive breast cancer. Clin Breast Cancer, 2019). 19(6):392–8., DOI 10. 1016/j.clbc.2019.05.019 52. Cerbelli B, Botticelli A, Pisano A, Pernazza A, Campagna D, De Luca A, et al. CD73 expression and pathologic response to neoadjuvant chemotherapy in triple negative breast cancer. Virchows Arch, 2019). 476(4):569–76., DOI 10. 1007/s00428-019-02722-6 53. Zamarron BF, Chen W Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci, 2011). 7(5):651–8., DOI 10.7150/ ijbs.7.651 54. Gobert M, Treilleux I, Bendriss-Vermare N, Bachelot T, Goddard-Leon S, Arfi V, et al. Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome. Cancer Res, 2009). 69(5):2000–9., DOI 10.1158/ 0008-5472.CAN-08-2360 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 600727 EP 600737 DI 10.3389/pore.2021.600727 PG 11 ER PT J AU Zhou, W Chen, Y Luo, R Li, Z Jiang, G Ou, X AF Zhou, Wanbang Chen, Yiyang Luo, Ruixing Li, Zifan Jiang, Guanwei Ou, Xi TI Identification of Biomarkers Related to Immune Cell Infiltration in Hepatocellular Carcinoma Using Gene Co-Expression Network SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE hepatocellular carcinoma; CIBERSORT; weighted gene co-expression network analysis; bioinformatics; the cancer genome atlas ID hepatocellular carcinoma; CIBERSORT; weighted gene co-expression network analysis; bioinformatics; the cancer genome atlas AB Hepatocellular carcinoma (HCC) is a common cancer with poor prognosis. Due to the lack of effective biomarkers and its complex immune microenvironment, the effects of current HCC therapies are not ideal. In this study, we used the GSE57957 microarray data from Gene Expression Omnibus database to construct a co-expression network. The weighted gene co-expression network analysis and CIBERSORT algorithm, which quantifies cellular composition of immune cells, were used to identify modules related to immune cells. Four hub genes (EFTUD2, GAPDH, NOP56, PA2G4) were identified by co-expression network and protein-protein interactions network analysis. We examined these genes in TCGA database, and found that the four hub genes were highly expressed in tumor tissues in multiple HCC groups, and the expression levels were significantly correlated with patient survival time, pathological stage and tumor progression. On the other hand, methylation analysis showed that the up-regulation of EFTUD2, GAPDH, NOP56 might be due to the hypomethylation status of their promoters. Next, we investigated the correlations between the expression levels of four hub genes and tumor immune infiltration using Tumor Immune Estimation Resource (TIMER). Gene set variation analysis suggested that the four hub genes were associated with numerous pathways that affect tumor progression or immune microenvironment.Overall, our results showed that the four hub genes were closely related to tumor prognosis, and may serve as targets for treatment and diagnosis of HCC. In addition, the associations between these genes and immune infiltration enhanced our understanding of tumor immune environment and provided new directions for the development of drugs and the monitoring of tumor immune status. C1 [Zhou, Wanbang] Peking University Shenzhen HospitalShenzhen, China. [Chen, Yiyang] Peking University Shenzhen HospitalShenzhen, China. [Luo, Ruixing] Peking University Shenzhen HospitalShenzhen, China. [Li, Zifan] Peking University Shenzhen HospitalShenzhen, China. [Jiang, Guanwei] Peking University Shenzhen HospitalShenzhen, China. [Ou, Xi] Peking University Shenzhen HospitalShenzhen, China. RP Ou, X (reprint author), Peking University Shenzhen Hospital, Shenzhen, China. EM bdszyyox@163.com CR Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin, 2019). 69(1):7–34., DOI 10.3322/caac.21551 McGlynn KA, Petrick JL, London WT. Global epidemiology of hepatocellular carcinoma. Clin Liver Dis, 2015). 19(2):223–38., DOI 10.1016/j.cld.2015.01.001 Reghupaty SC, Sarkar D. Current status of gene therapy in hepatocellular carcinoma. Cancers, Basel,, 2019). 11(9):265., DOI 10.3390/cancers11091265 Khalil DN, Smith EL, Brentjens RJ,Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol, 2016). 13(5):273–90., DOI 10.1038/nrclinonc.2016.25 El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, et al. Nivolumab in patients with advanced hepatocellular carcinoma, CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet, 2017). 389(10088):2492–502., DOI 10.1016/s0140-6736(17)31046-2 Sia D, Jiao Y, Martinez-Quetglas I, Kuchuk O, Villacorta-Martin C, Castro de Moura M, et al. Identification of an immune-specific class of hepatocellular carcinoma, based on molecular features. Gastroenterology, 2017). 153(3): 812–26., DOI 10.1053/j.gastro.2017.06.007 Johnston MP, Khakoo SI. Immunotherapy for hepatocellular carcinoma: current and future. Wjg, 2019). 25(24):2977–89., DOI 10.3748/wjg.v25.i24.2977 Wang D, Liu J, Liu S, Li W. Identification of crucial genes associated with immune cell infiltration in hepatocellular carcinoma by weighted gene Coexpression network analysis. Front Genet, 2020). 11:342., DOI 10.3389/fgene. 2020.00342 Song ZY, Chao F, Zhuo Z, Ma Z, Li W, Chen G. Identification of hub genes in prostate cancer using robust rank aggregation and weighted gene coexpression network analysis. Aging, 2019). 11(13):4736–56., DOI 10. 18632/aging.102087 Zhou L, Du Y, Kong L, Zhang X, Chen Q. Identification of molecular target genes and key pathways in hepatocellular carcinoma by bioinformatics analysis. Onco Targets Ther, 2018). 11:1861–9., DOI 10.2147/ott.s156737 Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics, 2008). 9:559., DOI 10.1186/1471-2105- 9-559 Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods, 2015). 12(5):453–7., DOI 10.1038/nmeth.3337 Zhou Y, Zhou B, Pache L, Chang M, Khodabakshi AH, Tanaseichuk O, et al. Metascape provides a biologist-oriented resource for the analysis of systemslevel datasets. Nat Commun, 2019). 10(1):1523., DOI 10.1038/s41467-019- 09234-6 Szklarczyk D, Franceschini A,Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res, 2015). 43(Database issue):D447–D452., DOI 10. 1093/nar/gku1003 Tomczak K, Czerwi´nska PM, Wiznerowicz M. The Cancer Genome Atlas, TCGA): an immeasurable source of knowledge. Contemp Oncol, Pozn,, 2015). 19(1A):A68–77., DOI 10.5114/wo.2014.47136 Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: a web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res, 2017). 77(21):e108–e110., DOI 10.1158/0008-5472.can-17-0307 Hanzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics, 2013). 14:7., DOI 10.1186/ 1471-2105-14-7 Carbonell P, Faulon J-L. Molecular signatures-based prediction of enzyme promiscuity. Bioinformatics, 2010). 26(16):2012–9., DOI 10.1093/ bioinformatics/btq317 Vogel A, Cervantes A, Chau I, Daniele B, Llovet JM, Meyer T, et al. Correction to: "hepatocellular carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up". Ann Oncol, 2019). 30(5):871–3., DOI 10.1093/ annonc/mdy510 European Association for the Study of the Liver. Corrigendum to "EASL clinical practice guidelines: management of hepatocellular carcinoma" [J hepatol 69, 2018, 182–236]. J Hepatol, 2019). 70(4):817., DOI 10.1016/j.jhep. 2019.01.020 Hilmi M, Neuzillet C, Calderaro J, Lafdi F, Pawlotsky JM, Rousseau B. Angiogenesis and immune checkpoint inhibitors as therapies for hepatocellular carcinoma: current knowledge and future research directions. J Immunother Cancer, 2019). 7(1):333., DOI 10.1186/s40425-019- 0824-5 Hu X, BaoM, Huang J, Zhou L, Zheng S. Identification and validation of novel biomarkers for diagnosis and prognosis of hepatocellular carcinoma. Front Oncol, 2020). 10:541479., DOI 10.3389/fonc.2020.541479 Zhu C, Xiao F, Hong J, Wang K, Liu X, Cai D, et al. EFTUD2 is a novel innate immune regulator restricting hepatitis C virus infection through the RIG-I/ MDA5 pathway. J Virol, 2015). 89(13):6608–18., DOI 10.1128/jvi.00364-15 Lv Z, Wang Z, Luo L, Chen Y, Han G, Wang R, et al. Spliceosome protein Eftud2 promotes colitis-associated tumorigenesis bymodulating inflammatory response of macrophage. Mucosal Immunol, 2019). 12(5):1164–73., DOI 10. 1038/s41385-019-0184-y Chi Q, Geng X, Xu K, Wang C, Zhao H. Potential targets and molecular mechanism of miR-331-3p in hepatocellular carcinoma identified by weighted gene coexpression network analysis. Biosci Rep, 2020). 40(6)., DOI 10.1042/ bsr20200124 Scarpa ES, Fabrizio G, Di Girolamo M. A role of intracellular mono-ADPribosylation in cancer biology. FEBS J, 2013). 280(15):3551–62., DOI 10.1111/ febs.12290 Chen RW, Saunders PA, Wei H, Li Z, Seth P, Chuang DM. Involvement of glyceraldehyde-3-phosphate dehydrogenase, GAPDH, and p53 in neuronal apoptosis: evidence that GAPDH is upregulated by p53. J Neurosci, 1999). 19(21):9654–62., DOI 10.1523/jneurosci.19-21-09654.1999 Sirover MA. Pleiotropic effects of moonlighting glyceraldehyde-3-phosphate dehydrogenase, GAPDH, in cancer progression, invasiveness, and metastases. Cancer Metastasis Rev, 2018). 37(4):665–76., DOI 10.1007/s10555-018-9764-7 Gong Y, Zou B, Peng S, Li P, Zhu G, Chen J, et al. Nuclear GAPDH is vital for hypoxia-induced hepatic stellate cell apoptosis and is indicative of aggressive hepatocellular carcinoma behavior. Cancer Manag Res, 2019). 11:4947–56., DOI 10.2147/cmar.s202268 Deng F, Chen D, Wei X, Lu S, Luo X, He J, et al. Development and validation of a prognostic classifier based on HIF-1 signaling for hepatocellular carcinoma. Aging, 2020). 12(4):3431–50., DOI 10.18632/aging.102820 Stepanov GA, Filippova JA, Komissarov AB, Kuligina EV, Richter VA, Semenov DV. Regulatory role of small nucleolar RNAs in human diseases. Biomed Res Int, 2015). 2015:206849., DOI 10.1155/2015/206849 Ussowicz M, Marcel V, Nguyen Van Long F, Kazanowska B, Diaz JJ, Wolowiec D. Analysis of the rRNA methylation complex components in pediatric B-cell precursor acute lymphoblastic leukemia: a pilot study. Adv Clin Exp Med, 2020). 29(1):107–13., DOI 10.17219/acem/112608 Giridhar KV, Sosa CP, Hillman DW, Sanhueza C, Dalpiaz CL, Costello BA, et al. Whole blood mRNA expression-based prognosis of metastatic renal cell carcinoma. Int J Mol Sci, 2017). 18(11):2326., DOI 10.3390/ ijms18112326 Miao X, Tang Q, Miao X, Wu Y, Qian J, ZhaoW, et al. ErbB3 binding protein 1, EBP1, participates in the regulation of intestinal inflammation via mediating Akt signaling pathway. Mol Immunol, 2015). 67(2 Pt B):540–51., DOI 10.1016/j. molimm.2015.07.032 Koach J, Holien JK, Massudi H, Carter DR, Ciampa OC, Herath M, et al. Drugging MYCN oncogenic signaling through the MYCN-PA2G4 binding interface. Cancer Res, 2019). 79(21):5652–67., DOI 10.1158/0008-5472.can-19- 1112 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 601693 EP 601704 DI 10.3389/pore.2021.601693 PG 12 ER PT J AU Yuan, J Kihara, T Kimura, N Hashikura, Y Ohkouchi, M Isozaki, K Takahashi, T Nishida, T Ito, A Hirota, S AF Yuan, Jiayin Kihara, Takako Kimura, Neinei Hashikura, Yuka Ohkouchi, Mizuka Isozaki, Koji Takahashi, Tsuyoshi Nishida, Toshirou Ito, Akihiko Hirota, Seiichi TI Differential Expression of CADM1 in Gastrointestinal Stromal Tumors of Different Sites and with Different Gene Abnormalities SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gastrointestinal stromal tumor; GIST; CADM1; gastric GIST; duodenal/jejuno-ileal GIST; mutation type ID gastrointestinal stromal tumor; GIST; CADM1; gastric GIST; duodenal/jejuno-ileal GIST; mutation type AB Gastrointestinal stromal tumor (GIST), the most common mesenchymal tumor of the human gastrointestinal tract, differentiating toward the interstitial cell of Cajal (ICC), arises predominantly in the stomach and small intestine. Small intestinal GISTs appear to have worse prognosis than gastric GISTs. In a pilot study of a cDNA expression chip using several GISTs, we found that Cell Adhesion Molecule 1 (CADM1), which could contribute to tumor growth and infiltration, is expressed more strongly in small intestinal GISTs than gastric GISTs. In the present study, we examined CADM1 expression in GISTs of different sites and with different gene abnormalities using a large number of gastric and small intestinal GISTs. First, immunoblotting confirmed significantly higher CADM1 expression in small intestinal GISTs with exon 11 c-kit mutation than gastric GISTs with exon 11 c-kit mutation. Real-time PCR also revealed that small intestinal GISTs with exon 11 c-kit mutation showed significantly higher CADM1 mRNA than gastric GISTs with exon 11 c-kit mutation. Although most small intestinal GISTs showed high CADM1 mRNA expression regardless of gene abnormality types, different CADM1 expression was detected between gastric GISTs with c-kit mutation and those with PDGFRA mutation. Immunohistochemistry showed that many small intestinal GISTs were CADM1-positive but most gastric GISTs CADM1-negative or -indefinite. In the normal gastric and small intestinal walls, immunoreactivity of CADM1 was detected only in nerves, but neither in gastric ICCs nor small intestinal ICCs, indicating that the high CADM1expression in small intestinal GISTs might be acquired during tumorigenesis. Different CADM1 expression between gastric and small intestinal GISTs might be related to different prognoses between them. Further functional experiments are needed to elucidate the role of CADM1 on GIST biology, and there is a possibility that targeting therapy against CADM1 has a preventive effect for tumor spreading in small intestinal GISTs. C1 [Yuan, Jiayin] Hyogo College of Medicine, Department of Surgical PathologyNishinomiya, Japan. [Kihara, Takako] Hyogo College of Medicine, Department of Surgical PathologyNishinomiya, Japan. [Kimura, Neinei] Hyogo College of Medicine, Department of Surgical PathologyNishinomiya, Japan. [Hashikura, Yuka] Hyogo College of Medicine, Department of Surgical PathologyNishinomiya, Japan. [Ohkouchi, Mizuka] Hyogo College of Medicine, Department of Surgical PathologyNishinomiya, Japan. [Isozaki, Koji] Hyogo College of Medicine, Department of Surgical PathologyNishinomiya, Japan. [Takahashi, Tsuyoshi] Osaka University Graduate School of Medicine, Departtment of Gastroenterological SurgerySuita, Japan. [Nishida, Toshirou] Osaka Hospital, Japan Community Healthcare Organization (JCHO)Osaka, Japan. [Ito, Akihiko] Kindai University, Faculty of Medicine, Department of PathologyOsaka, Japan. [Hirota, Seiichi] Hyogo College of Medicine, Department of Surgical PathologyNishinomiya, Japan. RP Hirota, S (reprint author), Hyogo College of Medicine, Department of Surgical Pathology, Nishinomiya, Japan. EM hiros@hyo-med.ac.jp CR Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Ishiguro S, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science, 1998, 279(5350):577–80., DOI 10.1126/science.279.5350.577 Heinrich MC, Corless CL, Duensing A, McGreevey L, Chen CJ, Joseph N, et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science, 2003, 299(5607):708–10., DOI 10.1126/science.1079666 Martin-Broto J, Rubio L, Alemany R, Lopez-Guerrero JA. Clinical implications of KIT and PDGFRA genotyping in GIST. Clin Transl Oncol, 2010, 12(10): 670–6., DOI 10.1007/s12094-010-0576-7 Antonescu CR, Sommer G, Sarran L, Tschernyavsky SJ, Riedel E, Woodruff JM, et al. Association of KIT exon 9 mutations with nongastric primary site and aggressive behavior: KIT mutation analysis and clinical correlates of 120 gastrointestinal stromal tumors. Clin Cancer Res, 2003, 9(9):3329–37. NanniniM, Biasco G, Astolfi A, Pantaleo MA. An overview on molecular biology of KIT/PDGFRA wild type, WT, gastrointestinal stromal tumours, GIST). J Med Genet, 2013, 50(10):653–61., DOI 10.1136/jmedgenet-2013-101695 Pantaleo MA, Astolfi A, Astolfi A, Urbini M, Nannini M, Paterini P, et al. Analysis of all subunits, SDHA, SDHB, SDHC, SDHD, of the succinate dehydrogenase complex in KIT/PDGFRA wild-type GIST. Eur J Hum Genet, 2014, 22(1):32–9., DOI 10.1038/ejhg.2013.80 Kinoshita K, Hirota S, Isozaki K, Ohashi A, Nishida T, Kitamura Y, et al. Absence of c-kit gene mutations in gastrointestinal stromal tumours from neurofibromatosis type 1 patients. J Pathol, 2004, 202(1):80–5., DOI 10.1002/ path.1487 Agaram NP, Wong GC, Guo T, Maki RG, Singer S, Dematteo RP, et al. Novel V600E BRAF mutations in imatinib-naive and imatinib-resistant gastrointestinal stromal tumors. Genes Chromosom Cancer, 2008, 47(10): 853–9., DOI 10.1002/gcc.20589 Miettinen M, Lasota J. Gastrointestinal stromal tumors - definition, clinical, histological, immunohistochemical, and molecular genetic features and differential diagnosis. Virchows Archiv, 2001, 438(1):1–12., DOI 10.1007/ s004280000338 Joensuu H. Risk stratification of patients diagnosed with gastrointestinal stromal tumor. Hum Pathol, 2008, 39(10):1411–9., DOI 10.1016/j.humpath. 2008.06.025 Miettinen M, Lasota J. Gastrointestinal stromal tumors: pathology and prognosis at different sites. Semin Diagn Pathol, 2006, 23(2):70–83., DOI 10. 1053/j.semdp.2006.09.001 Emory TS, Sobin LH, Lukes L, Lee DH, O’Leary TJ. Prognosis of gastrointestinal smooth-muscle, stromal, tumors. Am J Surg Pathol, 1999, 23(1):82–7., DOI 10.1097/00000478-199901000-00009 Yang Z, Wang F, Liu S, Guan W. Comparative clinical features and short-term outcomes of gastric and small intestinal gastrointestinal stromal tumours: a retrospective study. Sci Rep, 2019, 9(1):10033., DOI 10.1038/s41598-019-46520-1 Antonescu CR, Viale A, Sarran L, Tschernyavsky SJ, Gonen M, Segal NH, et al. Gene expression in gastrointestinal stromal tumors is distinguished by KIT genotype and anatomic site. Clin Cancer Res, 2004, 10(10):3282–90., DOI 10. 1158/1078-0432.CCR-03-0715 Ichikawa H, Yoshida A, Kanda T, Kosugi Si., Ishikawa T, Hanyu T, et al. Prognostic significance of promyelocytic leukemia expression in gastrointestinal stromal tumor; integrated proteomic and transcriptomic analysis. Cancer Sci, 2015, 106(1):115–24., DOI 10.1111/cas.12565 Biederer T, Sara Y, Mozhayeva M, Atasoy D, Liu XR, Kavalali ET, et al., 2002). SynCAM, a synaptic adhesion molecule that drives synapse assembly. Science 297, 5586):1525–31., DOI 10.1126/science.1072356 Ito A, Oonuma J. Direct interaction between nerves andmast cells mediated by the SgIGSF/SynCAM adhesion molecule. J Pharmacol Sci, 2006, 102(1):1–5., DOI 10.1254/jphs.cpj06014x Biederer T. Bioinformatic characterization of the SynCAM family of immunoglobulin-like domain-containing adhesion molecules. Genomics, 2006, 87(1):139–50., DOI 10.1016/j.ygeno.2005.08.017 Ito A, Hagiyama M, Oonuma J. Nerve-mast cell and smooth muscle-mast cell interaction mediated by cell adhesion molecule-1, CADM1. J Smooth Muscle Res, 2008, 44(2):83–93., DOI 10.1540/jsmr.44.83 Moiseeva EP, Roach KM, Leyland ML, Bradding P. CADM1 is a key receptor mediating human mast cell adhesion to human lung fibroblasts and airway smooth muscle cells. PLoS One, 2013, 8(4):e61579., DOI 10.1371/journal.pone. 0061579 Moiseeva EP, Straatman KR, Leyland ML, Bradding P. CADM1 controls actin cytoskeleton assembly and regulates extracellular matrix adhesion in human mast cells. PLoS One, 2014, 9(1):e85980, DOI 10.1371/journal.pone. 0085980 Wakayama T, Iseki S. Role of the spermatogenic-Sertoli cell interaction through cell adhesion molecule-1, CADM1, in spermatogenesis. Anat Sci Int, 2009, 84(3):112–21., DOI 10.1007/s12565-009-0034-1 Koma YI, Furuno T, Hagiyama M, Hamaguchi K, Nakanishi M, Masuda M, et al. Cell adhesion molecule 1 is a novel pancreatic-islet cell adhesion molecule that mediates nerve-islet cell interactions. Gastroenterology, 2008, 134(5): 1544–54., DOI 10.1053/j.gastro.2008.01.081 Kuramochi M, Fukuhara H, Nobukuni T, Kanbe T, Maruyama T, Ghosh HP, et al., 2001). TSLC1 is a tumor-suppressor gene in human non-small-cell lung cancer. Nat Genet 27, 4):427–30., DOI 10.1038/86934 Fukami T, Fukuhara H, Kuramochi M, Maruyama T, Isogai K, Sakamoto M, et al. Promoter methylation of theTSLC1 gene in advanced lung tumors and various cancer cell lines. Int J Cancer, 2003, 107(1):53–9., DOI 10.1002/ijc.11348 Honda T, Tamura G, Waki T, Jin Z, Sato K, Motoyama T, et al. Hypermethylation of theTSLC1Gene promoter in primary gastric cancers and gastric cancer cell lines. Jpn J Cancer Res, 2002, 93(8):857–60., DOI 10.1111/ j.1349-7006.2002.tb01329.x Allinen M, Peri L, Kujala S, Lahti-Domenici J, Outila K, Karppinen S-M, et al. Analysis of 11q21-24 loss of heterozygosity candidate target genes in breast cancer: indications of TSLC1 promoter hypermethylation. Genes Chromosom Cancer, 2002, 34(4):384–9., DOI 10.1002/gcc.10079 Takahashi Y, Iwai M, Kawai T, Arakawa A, Ito T, Sakurai-Yageta M, et al. Aberrant expression of tumor suppressors CADM1 and 4.1B in invasive lesions of primary breast cancer. Breast Cancer, 2012, 19(3):242–52., DOI 10. 1007/s12282-011-0272-7 Lung HL, Leung Cheung AK, Xie D, Cheng Y, Kwong FM, Murakami Y, et al. TSLC1 is a tumor suppressor gene associated with metastasis in nasopharyngeal carcinoma. Cancer Res, 2006, 66(19):9385–92., DOI 10.1158/ 0008-5472.CAN-06-0590 Liu D, Feng X, Wu X, Li Z, Wang W, Tao Y, et al. Tumor suppressor in lung cancer 1, TSLC1), a novel tumor suppressor gene, is implicated in the regulation of proliferation, invasion, cell cycle, apoptosis, and tumorigenicity in cutaneous squamous cell carcinoma. Tumor Biol, 2013, 34(6):3773–83., DOI 10.1007/s13277-013-0961-2 Zeng D, Wu X, Zheng J, Zhuang Y, Chen J, Hong C, et al. Loss of CADM1/ TSLC1 expression is associated with poor clinical outcome in patients with esophageal squamous cell carcinoma. Gastroenterol Res Pract, 2016, 2016:1., DOI 10.1155/2016/6947623 Sasaki H, Nishikata I, Shiraga T, Akamatsu E, Fukami T, Hidaka T, et al. Overexpression of a cell adhesion molecule, TSLC1, as a possible molecular marker for acute-type adult T-cell leukemia. Blood, 2005, 105(3):1204–13., DOI 10.1182/blood-2004-03-1222 Dewan MZ, Takamatsu N, Hidaka T, Hatakeyama K, Nakahata S, Fujisawa J-i., et al. Critical role for TSLC1 expression in the growth and organ infiltration of adult T-cell leukemia cells in vivo. Jvi, 2008, 82(23):11958–63., DOI 10.1128/JVI.01149-08 Ito A, Hagiyama M, Mimura T, Matsumoto M, Wakayama T, Iseki S, et al. Expression of cell adhesion molecule 1 in malignant pleural mesothelioma as a cause of efficient adhesion and growth on mesothelium. Lab Invest, 2008, 88(5):504–14., DOI 10.1038/labinvest.2008.15 Kimura R, Yoneshige A, Hagiyama M, Otani T, Inoue T, Shiraishi N, et al. Expression of cell adhesion molecule 1 in gastric neck and base glandular cells: possible involvement in peritoneal dissemination of signet ring cells. Life Sci, 2018, 213:206–13., DOI 10.1016/j.lfs.2018.10.017 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 602008 EP 602017 DI 10.3389/pore.2021.602008 PG 10 ER PT J AU Mao, L Zhao, W Li, X Zhang, Sh Zhou, Ch Zhou, D Ou, X Xu, Y Tang, Y Ou, X Hu, Ch Ding, X Luo, P Yu, Sh AF Mao, Linlin Zhao, Weiwei Li, Xiaoxia Zhang, Shangfei Zhou, Changhong Zhou, Danyan Ou, Xiaohua Xu, Yanyan Tang, Yuanxiao Ou, Xiaoyong Hu, Changming Ding, Xiangdong Luo, Pifu Yu, Shihui TI Mutation Spectrum of EGFR From 21,324 Chinese Patients With Non-Small Cell Lung Cancer (NSCLC) Successfully Tested by Multiple Methods in a CAP-Accredited Laboratory SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE non-small cell lung cancer; EGFR mutation testing; Sanger sequencing; real-time PCR; next-generation sequencing ID non-small cell lung cancer; EGFR mutation testing; Sanger sequencing; real-time PCR; next-generation sequencing AB Genotyping epidermal growth factor receptor (EGFR) gene in patients with advanced nonsmall cell lung cancers (NSCLC) is essential for identifying those patients who may benefit from targeted therapies. Systemically evaluating EGFR mutation detection rates of different methods currently used in clinical setting will provide valuable information to clinicians and laboratory scientists who take care of NSCLC patients. This study retrospectively reviewed the EGFR data obtained in our laboratory in last 10 years. A total of 21,324 NSCLC cases successfully underwent EGFR genotyping for clinical therapeutic purpose, including 5,244 cases tested by Sanger sequencing, 13,329 cases tested by real-time PCR, and 2,751 tested by next-generation sequencing (NGS). The average EGFR mutation rate was 45.1%, with 40.3% identified by Sanger sequencing, 46.5% by real-time PCR and 47.5% by NGS. Of these cases with EGFR mutations identified, 93.3% of them harbored a single EGFR mutation (92.1% with 19del or L858R, and 7.9% with uncommon mutations) and 6.7% harbored complex EGFR mutations. Of the 72 distinct EGFR variants identified in this study, 15 of them (single or complex EGFR mutations) were newly identified in NSCLC. For these cases with EGFR mutations tested by NGS, 65.3% of them also carried tumor-related variants in some non-EGFR genes and about one third of them were considered candidates of targeted drugs. NGS method showed advantages over Sanger sequencing and real-time PCR not only by providing the highest mutation detection rate of EGFR but also by identifying actionable non-EGFR mutations with targeted drugs in clinical setting. C1 [Mao, Linlin] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Zhao, Weiwei] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Li, Xiaoxia] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Zhang, Shangfei] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Zhou, Changhong] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Zhou, Danyan] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Ou, Xiaohua] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Xu, Yanyan] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Tang, Yuanxiao] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Ou, Xiaoyong] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Hu, Changming] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. [Ding, Xiangdong] KingMed Diagnostics, Department of PathologyGuangzhou, China. [Luo, Pifu] KingMed Diagnostics, Department of PathologyGuangzhou, China. [Yu, Shihui] KingMed Diagnostics, Clinical Genome CenterGuangzhou, China. RP Yu, Sh (reprint author), KingMed Diagnostics, Clinical Genome Center, Guangzhou, China. CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018, 68:394–424., DOI 10.3322/ caac.21492 Siegel RL,Miller KD, Jemal A. Cancer statistics, 2019. CA A Cancer J Clin, 2019, 69:7–34., DOI 10.3322/caac.21551 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin, 2016, 66:115–32., DOI 10.3322/caac.21338 Gridelli C, Rossi A, Carbone DP, Guarize J, Karachaliou N, Mok T, et al. Nonsmall- cell lung cancer. Nat Rev Dis Primers, 2015, 1:15009., DOI 10.1038/nrdp. 2015.9 Kumarakulasinghe NB, van Zanwijk N, Soo RA. Molecular targeted therapy in the treatment of advanced stage non-small cell lung cancer, NSCLC). Respirology, 2015, 20:370–8., DOI 10.1111/resp.12490 Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med, 2009, 361: 947–57., DOI 10.1056/nejmoa0810699 Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor, WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol, 2010, 11:121–8., DOI 10.1016/s1470-2045(09)70364-x Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer, OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol, 2010, 12: 735–42., DOI 10.1016/S1470-2045(11)70184-X Castellanos E, Feld E, Horn L Driven by mutations: the predictive value of mutation subtype in EGFR -mutated non-small cell lung cancer. J Thorac Oncol, 2017, 12:612–23., DOI 10.1016/j.jtho.2016.12.014 Dearden S, Stevens J, Wu YL, Blowers D. Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology, mutMap). Ann Oncol, 2013, 24:2371–6., DOI 10.1093/annonc/mdt205 National Comprehensive Cancer Network. NCCN clinical practice guidelines in Oncology, NCCN Guidelines®, non-small cell lung cancer, 2020). Available from: https://www.nccn.org/professionals/physician_gls/default.aspx#nscl/, Accessed 7 August 2020). Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, et al. Correction to: “metastatic non-small cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up”. Ann Oncol, 2019, 30: 863–70., DOI 10.1093/annonc/mdy474 National Health Commission of the People’s Republic of China. Chinese guidelines for diagnosis and treatment of primary lung cancer 2018, English version). Chin J Cancer Res, 2019, 31:1–28., DOI 10.21147/j.issn.1000-9604. 2019.01.01 Kalemkerian GP, Narula N, Kennedy EB, Biermann WA, Donington J, Leighl NB, et al. Molecular testing guideline for the selection of patients with lung cancer for treatment with targeted tyrosine kinase inhibitors: American society of clinical oncology endorsement of the college of American pathologists/ international association for the study of lung cancer/association for molecular pathology clinical practice guideline update. J Clin Oncol, 2018, 36:911–9., DOI 10.1200/jco.2017.76.7293 Bai H,Wang Z, Wang Y, Zhuo M, Zhou Q, Duan J, et al. Detection and clinical significance of intratumoral EGFR mutational heterogeneity in Chinese patients with advanced non-small cell lung cancer. PLoS One, 2013, 8: e54170., DOI 10.1371/journal.pone.0054170 Cheng Y,Wang Y, Zhao J, Liu Y, Gao H, Ma K, et al. Real-world EGFR testing in patients with stage IIIB/IV non-small-cell lung cancer in North China: a multicenter, non-interventional study. Thorac Cancer, 2018, 9:1461–9., DOI 10. 1111/1759-7714.12859 Li MM, Datto M, Duncavage EJ, Kulkarni S, Lindeman NI, Roy S, et al. Standards and guidelines for the interpretation and reporting of sequence variants in cancer. J Mol Diagn, 2017, 19:4–23., DOI 10.1016/j.jmoldx.2016. 10.002 Wu JY, Yu CJ, Chang YC, Yang CH, Shih JY, Yang PC. Effectiveness of tyrosine kinase inhibitors on “uncommon” epidermal growth factor receptor mutations of unknown clinical significance in non-small cell lung cancer. Clin Cancer Res, 2011, 17:3812–21., DOI 10.1158/1078-0432. ccr-10-3408 Tu HY, Ke EE, Yang JJ, Sun YL, Yan HH, Zheng MY, et al. A comprehensive review of uncommon EGFR mutations in patients with non-small cell lung cancer. Lung Cancer, 2017, 114:96–102., DOI 10.1016/j.lungcan.2017.11.005 Massarelli E, Johnson FM, Erickson HS, Wistuba II, Papadimitrakopoulou V Uncommon epidermal growth factor receptor mutations in non-small cell lung cancer and their mechanisms of EGFR tyrosine kinase inhibitors sensitivity and resistance. Lung Cancer, 2013, 80:235–41., DOI 10.1016/j. lungcan.2013.01.018 Liu Y, Xu F,Wang Y, Wu Q, Wang B, Yao Y, et al. Mutations in exon 8 of TP53 are associated with shorter survival in patients with advanced lung cancer. Oncol Lett, 2019, 18:3159–69., DOI 10.3892/ol.2019.10625 Xiao Y, Yin C, Wang Y, Lv H, Wang W, Huang Y, et al. FBXW 7 deletion contributes to lung tumor development and confers resistance to gefitinib therapy. Mol Oncol, 2018, 12:883–95., DOI 10.1002/1878-0261.12200 Labbe C, Cabanero M, Korpanty GJ, Tomasini P, Doherty MK, Mascaux C., et al. Prognostic and predictive effects of TP53 co-mutation in patients with EGFR -mutated non-small cell lung cancer, NSCLC). Lung Cancer, 2017, 111: 23–9., DOI 10.1016/j.lungcan.2017.06.014 Skoulidis F, Heymach JV Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat Rev Cancer, 2019, 19:495–509., DOI 10. 1038/s41568-019-0179-8 Tsukumo Y, Naito M, Suzuki T Influence of EGFR-activating mutations on sensitivity to tyrosine kinase inhibitors in a KRAS mutant non-small cell lung cancer cell line. PLoS One, 2020, 15:e0229712., DOI 10.1371/journal.pone. 0229712 Yang J-J, Zhang XC, Su J, Xu CR, Zhou Q, Tian HX, et al. Lung cancers with concomitant EGFR mutations and ALK rearrangements: diverse responses to EGFR-TKI and crizotinib in relation to diverse receptors phosphorylation. Clin Cancer Res, 2014, 20:1383–92., DOI 10.1158/1078-0432.ccr-13-0699 Lee CK, Wu Y-L, Ding PN, Lord SJ, Inoue A, Zhou C, et al. Impact of specific epidermal growth factor receptor, EGFR, mutations and clinical characteristics on outcomes after treatment with EGFR tyrosine kinase inhibitors versus chemotherapy in EGFR-mutant lung cancer: a metaanalysis. J Clin Oncol, 2015, 33:1958–65., DOI 10.1200/jco.2014.58.1736 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 602726 EP 602733 DI 10.3389/pore.2021.602726 PG 8 ER PT J AU Bogos, K Kiss, Z Tamasi, L Ostoros, Gy Muller, V Urban, L Bittner, N Sarosi, V Vastag, A Polanyi, Z Nagy-Erdei, Zs Daniel, A Voko, Z Nagy, B Horvath, K Rokszin, Gy Abonyi-Toth, Zs Barcza, Zs Galffy, G Moldvay, J AF Bogos, Krisztina Kiss, Zoltan Tamasi, Lilla Ostoros, Gyula Muller, Veronika Urban, Laszlo Bittner, Nora Sarosi, Veronika Vastag, Aladar Polanyi, Zoltan Nagy-Erdei, Zsofia Daniel, Andrea Voko, Zoltan Nagy, Balazs Horvath, Krisztian Rokszin, Gyorgy Abonyi-Toth, Zsolt Barcza, Zsofia Galffy, Gabriella Moldvay, Judit TI Improvement in Lung Cancer Survival: 6-Year Trends of Overall Survival at Hungarian Patients Diagnosed in 2011–2016 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lung cancer; long-term survival; mortality; Hungary; survival ID lung cancer; long-term survival; mortality; Hungary; survival AB Objective: Lung cancer is one of the most common cancers worldwide and its survival is still poor. The objective of our study was to estimate long-term survival of Hungarian lung cancer patients at first time based on a nationwide review of the National Health Insurance Fund database. Methods: Our retrospective, longitudinal study included patients aged ≥20 years who were diagnosed with lung cancer (ICD-10 C34) between January 1, 2011 and December 31, 2016. Survival rates were evaluated by year of diagnosis, patient gender and age, and morphology of lung cancer. Results: 41,854 newly diagnosed lung cancer patients were recorded. Mean age at diagnosis varied between 64.7 and 65.9 years during study period. One- and 5-year overall survival rates for the total population were 42.2 and 17.9%, respectively. Survival was statistically associated with gender, age and type of lung cancer. Female patients (n = 16,362) had 23% better survival (HR: 0.77, 95% confidence interval (CI): 0.75–0.79; p < 0.001) than males (n = 25,492). The highest survival rates were found in the 20–49 age cohort (5Y = 31.3%) and if the cancer type was adenocarcinoma (5Y = 20.5%). We measured 5.3% improvement (9.2% adjusted) in lung cancer survival comparing the period 2015–2016 to 2011–2012 (HR: 0.95 95% CI: 0.92–0.97; p = 0.003), the highest at females <60 year (0.86 (adjusted HR was 0.79), interaction analysis was significant for age and histology types. Conclusion: Our study provided long-term Lung cancer survival data in Hungary for the first time. We found a 5.3% improvement in 5-year survival in 4 years. Women and young patients had better survival. Survival rates were comparable to–and at the higher end of–rates registered in other East-Central European countries (7.7%–15.7%). C1 [Bogos, Krisztina] National Koranyi Institute of PulmonologyBudapest, Hungary. [Kiss, Zoltan] MSD Pharma Hungary Kft.Budapest, Hungary. [Tamasi, Lilla] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Ostoros, Gyula] National Koranyi Institute of PulmonologyBudapest, Hungary. [Muller, Veronika] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Urban, Laszlo] Matrahaza Healthcare Center and University Teaching HospitalMatrahaza, Hungary. [Bittner, Nora] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Sarosi, Veronika] University of Pecs, Faculty of MedicinePecs, Hungary. [Vastag, Aladar] MSD Pharma Hungary Kft.Budapest, Hungary. [Polanyi, Zoltan] MSD Pharma Hungary Kft.Budapest, Hungary. [Nagy-Erdei, Zsofia] MSD Pharma Hungary Kft.Budapest, Hungary. [Daniel, Andrea] MSD Pharma Hungary Kft.Budapest, Hungary. [Voko, Zoltan] Eotvos Lorand UniversityBudapest, Hungary. [Nagy, Balazs] Eotvos Lorand UniversityBudapest, Hungary. [Horvath, Krisztian] Eotvos Lorand UniversityBudapest, Hungary. [Rokszin, Gyorgy] RxTarget LtdSzolnok, Hungary. [Abonyi-Toth, Zsolt] RxTarget LtdSzolnok, Hungary. [Barcza, Zsofia] Syntesia LtdBudapest, Hungary. [Galffy, Gabriella] County Hospital of PulmonologyTorokbalint, Hungary. [Moldvay, Judit] National Koranyi Institute of Pulmonology, Semmelweis University, Ist Department of PulmonologyBudapest, Hungary. RP Bogos, K (reprint author), National Koranyi Institute of Pulmonology, Budapest, Hungary. EM bogos@koranyi.hu CR Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature, 2008). 455:1069–75., DOI 10.1038/nature07423 Ferlay J, Colombet M, Soerjomataram I, Dyba T, Randi G, Bettio M, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries and 25 major cancers in 2018. Eur J Cancer, 2018). 103:356–87., DOI 10.1016/j.ejca.2018.07.005 Hakulinen T, Engholm G, Gislum M, Storm HH, Klint A, Tryggvadottir L, et al. Trends in the survival of patients diagnosed with cancers in the respiratory system in the Nordic countries 1964-2003 followed up to the end of 2006. Acta Oncologica, 2010). 49(5):608–23., DOI 10.3109/02841860903575281 Torre LA, Siegel RL, Jemal A Lung cancer statistics. Adv Exp Med Biol, 2016). 893:1–19., DOI 10.1007/978-3-319-24223-1_1 Jemal A, Siegel R, Xu J, Ward E Cancer statistics, 2010. CA Cancer J Clin, 2010). 60(5):277–300., DOI 10.3322/caac.20073 Heuvers ME, Hegmans JP, Stricker BH, Aerts JG Improving lung cancer survival; time to move on. BMC Pulm Med, 2012). 12:77., DOI 10.1186/1471-2466-12-77 Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Nikˇsi´c M, et al. Global surveillance of trends in cancer survival 2000-14, CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet, 2018). 391(10125):1023–75., DOI 10.1016/S0140-6736(17)33326-3 American Cancer Society. Cancer facts & figures 2018. Atlanta: American Cancer Society, 2018). Woodard GA, Jones KD, Jablons DM Lung cancer staging and prognosis. Cancer Treat Res, 2016). 170:47–75., DOI 10.1007/978-3-319-40389-2_3 Lu T, Yang X, Huang Y, Zhao M, Li M, Ma K, et al. Trends in the incidence, treatment, and survival of patients with lung cancer in the last four decades. Cmar, 2019). 11:943–53., DOI 10.2147/cmar.s187317 BogosK,Kiss Z, GalffyG, Tamasi L,Ostoros G,Muller V, et al. Revising incidence and mortality of lung cancer in central Europe: an epidemiology review from Hungary. Front Oncol, 2019). 9:1051., DOI 10.3389/fonc.2019.01051 Tan KS, Eguchi T, Adusumilli PS Reporting net survival in populations: a sensitivity analysis in lung cancer demonstrates the differential implications of reporting relative survival and cause-specific survival. Clep, 2019). 11:781–92. eCollection 2019., DOI 10.2147/CLEP.S210894 ArnoldM,RutherfordMJ, BardotA, Ferlay J,AnderssonTM,MyklebustTA, et al. Progress in cancer survival, mortality, and incidence in seven high-income countries 1995-2014, ICBP SURVMARK-2): a population-based study. Lancet Oncol, 2019). 20(11):1493–505., DOI 10.1016/S1470-2045(19)30456-5 Jakobsen E, Rasmussen TR, Green A Mortality and survival of lung cancer in Denmark: results from the Danish lung cancer group 2000-2012. Acta Oncologica, 2016). 55(Suppl. 2):2–9., DOI 10.3109/0284186x.2016.1150608 Rossi S, Baili P, Capocaccia R, Caldora M, Carrani E, Minicozzi P, et al. The EUROCARE-5 study on cancer survival in Europe 1999-2007: database, quality checks and statistical analysis methods. Eur J Cancer, 2015). 51(15): 2104–19., DOI 10.1016/j.ejca.2015.08.001 Cancer Research UK, 2019). https://www.cancerresearchuk.org/healthprofessional/ cancer-statistics/statistics-by-cancer-type/lung-cancer/survival#heading- Zero, Accessed January 24, 2020). ArnoldM, Rutherford MJ, Bardot A, Ferlay J, Andersson TM-L,Myklebust TA, et al. Progress in cancer survival, mortality, and incidence in seven high-income countries 1995-2014, ICBP SURVMARK-2): a population-based study. Lancet Oncol, 2019). 20(11):1493–505., DOI 10.1016/s1470-2045(19)30456-5 National Cancer Institute. Surveillance, epidemiology and end result program. Bethesda:, SEER database): cancer stat facts: lung and bronchus cancer, 2020). https://seer.cancer.gov/statfacts/html/lungb.html. Last update, Accessed October 31, 2020). Wakelee HA, Wang W, Schiller JH, Langer CJ, Sandler AB, Belani CP, et al. Survival differences by sex for patients with advanced non-small cell lung cancer on eastern cooperative oncology group trial 1594. J Thorac Oncol, 2006). 1(5):441–6., DOI 10.1016/s1556-0864(15)31609-9 Radzikowska E, Glaz P, Roszkowski K Lung cancer in women: age, smoking, histology, performance status, stage, initial treatment and survival. Populationbased study of 20 561 cases. Ann Oncol, 2002). 13:1087–93., DOI 10.1093/ annonc/mdf187 Visbal AL, Williams BA, Nichols FC, Marks RS, Jett JR, Aubry M-C, et al. Gender differences in non-small-cell lung cancer survival: an analysis of 4,618 patients diagnosed between 1997 and 2002. Ann Thorac Surg, 2004). 78: 209–15., DOI 10.1016/j.athoracsur.2003.11.021 Rivera M Lung cancer in women: differences in epidemiology, biology, histology, and treatment outcomes. Semin Respir Crit Care Med, 2013). 34(6):792–801., DOI 10.1055/s-0033-1358550 Devesa SS, Bray F, Vizcaino AP, Parkin DM International lung cancer trends by histologic type: male:female differences diminishing and adenocarcinoma rates rising. Int J Cancer, 2005). 117(2):294–9., DOI 10.1002/ijc.21183 Meza R, Meernik C, Jeon J, Cote ML Lung cancer incidence trends by gender, race and histology in the United States, 1973-2010. PLoS One, 2015). 10(3): e0121323., DOI 10.1371/journal.pone.0121323 Welcker K Genderspezifische Unterschiede des Lungenkarzinoms. Zentralbl Chir, 2015). 140(3):260–5., DOI 10.1055/s-0034-1396231 Ramalingam S,DinanMA, Crawford J Survival comparison in patientswith stage IV lung cancer in academic versus community centers in the United States. J Thorac Oncol, 2018). 13(12):1842–50., DOI 10.1016/j.jtho.2018.09.007 Kinoshita FL, Ito Y, Morishima T, Miyashiro I, Nakayama T Sex differences in lung cancer survival: long-term trends using population-based cancer registry data in Osaka, Japan. Jpn J Clin Oncol, 2017). 47(9):863–9., DOI 10.1093/jjco/ hyx094 Fu JB, Kau TY, Severson RK, Kalemkerian GP Lung cancer in women. Chest, 2005). 127(3):768–77., DOI 10.1378/chest.127.3.768 Cerfolio RJ, Bryant AS, Scott E, SharmaM, Robert F, Spencer SA, et al. Women with pathologic stage I, II, and III non-small cell lung cancer have better survival than men. Chest, 2006). 130(6):1796–802., DOI 10.1378/chest. 130.6.1796 Velinovic M, Jankovic R, Jovanovic D, Skodric Trifunovic V, Gavrilovic D, Stojsic J, et al. Tumor characteristics, expressions of ERCC1, Bax, p53, IGF1R, Bcl2, Bcl2/Bax and prognostic factors for overall survival in patients with lung carcinoid. J BUON, 2019). 24(1):256–66. Walters S, Maringe C, Coleman MP, Peake MD, Butler J, Young N, et al. Lung cancer survival and stage at diagnosis in Australia, Canada, Denmark, Norway, Sweden and the UK: a population-based study, 2004-2007. Thorax, 2013). 68(6):551–64., DOI 10.1136/thoraxjnl-2012-202297 Duma NN, Santana-Davila JR, Molina JR Non-small cell lung cancer: epidemiology, screening, diagnosis, and treatment. Mayo Clinic Proc, 2019). 94(8):1623–40., DOI 10.1016/j.mayocp.2019.01.013 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 603937 EP 603945 DI 10.3389/pore.2021.603937 PG 9 ER PT J TI Single Cell Gene Transcriptome Analysis of Ovarian Mature Teratomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE teratoma; expression profile; single cell; ovarian tumor; transcriptome ID teratoma; expression profile; single cell; ovarian tumor; transcriptome AB Teratoma is a type of germ cell tumor that originates from totipotential germ cells that are present in gonads, which can differentiate into any of the cell types found in adult tissues. Ovarian teratomas are usually mature cystic teratomas (OMCTs, also known as dermoid cysts). Chromosome studies in OMCTs show that the chromosomes are uniformly homozygous with karyotype of 46, XX, indicating that they may be parthenogenic tumors that arise from a single ovum after thefirst meiotic division. However, the tissues in OMCTs have been known to be morphologically and immunophenotypically identical to the orthotopic tissues. Currently, expression profiles of tissue components in OMCTs are not known. To identify whether OMCT tissues are expressionally similar to or different from the orthotopic tissues, we adopted single-cell RNA-sequencing (scRNAseq), and analyzed transcriptomes of individual cells in heterogenous tissues of two OMCTs. We found that transcriptome profiles of the OMCTs at single cell level were not significantly different from those of normal cells in orthotopic locations. The present data suggest that parthenogeneticlly altered OMCTs may not alter expression profiles of inrivirual tissue components in OMCTs. CR Peterson WF, Prevost EC, Edmunds FT, Hundley JM, Jr, Morris FK. Benign cystic teratomas of the ovary. Am J Obstet Gynecol, 1955). 70:368–82., DOI 10. 1016/s0002-9378(16)37681-5 Ayhan A, Bukulmez O, Genc C, Karamursel BS, Ayhan A. Mature cystic teratomas of the ovary: case series from one institution over 34 years. Eur J Obstet Gynecol Reprod Biol, 2000). 88:153–7., DOI 10.1016/s0301-2115(99, 00141-4 Linder D, McCaw BK, Hecht F. Parthenogenic origin of benign ovarian teratomas. N Engl J Med, 1975). 292:63–6., DOI 10.1056/nejm197501092920202 Parrington JM,West LF, Povey S. The origin of ovarian teratomas. J Med Genet, 1984). 21:4–12., DOI 10.1136/jmg.21.1.4 Surti U, Hoffner L, Chakravarti A, Ferrell RE. Genetics and biology of human ovarian teratomas. I. Cytogenetic analysis and mechanism of origin. Am J Hum Genet, 1990). 47:635–43. Kato N, Kamataki A, Kurotaki H. Methylation profiles of imprinted genes are distinct between mature ovarian teratoma, complete hydatidiform mole, and extragonadalmature teratoma. Mod Pathol, 34, 502, 2021)., DOI 10.1038/s41379- 020-00668-8 Ding Y, Gu X-Y, Xu F, Shi X-Y, Yang D-Z, Zhong J, et al. MicroRNA expression profiling of mature ovarian teratomas. Oncol Lett, 2012). 3:35–8., DOI 10.3892/ol. 2011.438 Simboeck E, Di Croce L HDAC1, a novel marker for benign teratomas. EMBO J, 2010). 29:3893–5., DOI 10.1038/emboj.2010.281 Wagner M, Yoshihara M, Douagi I, Damdimopoulos A, Panula S, Petropoulos S, et al. Single-cell analysis of human ovarian cortex identifies distinct cell populations but no oogonial stem cells. Nat Commun, 2020). 11:1147., DOI 10. 1038/s41467-020-14936-3 Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM, III, et al. Comprehensive integration of single-cell data. Cell, 2019). 177:1888–902., DOI 10.1016/j.cell.2019.05.031 Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The molecular signatures database hallmark gene set collection. Cell Syst, 2015). 1:417–25., DOI 10.1016/j.cels.2015.12.004 Aran D, Looney AP, Liu L, Wu E, Fong V, Hsu A, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol, 2019). 20:163–72., DOI 10.1038/s41590-018- 0276-y Chu PG, Weiss LM. Keratin expression in human tissues and neoplasms. Histopathology, 2002). 40:403–39., DOI 10.1046/j.1365-2559.2002. 01387.x Cheng JB, Sedgewick AJ, Finnegan AI, Harirchian P, Lee J, Kwon S, et al. Transcriptional programming of normal and inflamed human epidermis at single-cell resolution. Cell Rep, 2018). 25:871–83., DOI 10.1016/j.celrep.2018. 09.006 The ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature, 2012). 489:57–74., DOI 10.1038/ nature11247 Martens JHA, Stunnenberg HG. BLUEPRINT: mapping human blood cell epigenomes. Haematologica, 2013). 98:1487–9., DOI 10.3324/haematol.2013. 094243 Mabbott NA, Baillie J, Brown H, Freeman TC, Hume DA. An expression atlas of human primary cells: inference of gene function from coexpression networks. BMC Genomics, 2013). 14:632., DOI 10.1186/ 1471-2164-14-632 Swindell WR, Johnston A, Voorhees JJ, Elder JT, Gudjonsson JE. Dissecting the psoriasis transcriptome: inflammatory- and cytokine-driven gene expression in lesions from 163 patients. BMC Genomics, 2013). 14:527., DOI 10.1186/1471-2164-14-527 Izar B, Tirosh I, Stover EH, Wakiro I, Cuoco MS, Alter I, et al. A single-cell landscape of high-grade serous ovarian cancer. Nat Med, 2020). 26:1271–9., DOI 10.1038/s41591-020-0926-0 Rozenblatt-Rosen O, Regev A, Oberdoerffer P, Nawy T, Hupalowska A, Rood JE, et al. Human tumor atlas network, 2020, the human tumor atlas network: charting tumor transitions across space and time at single-cell resolution. Cell 181:236–49., DOI 10.1016/j.cell.2020.03.053 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 604228 EP 604232 DI 10.3389/pore.2021.604228 PG 5 ER PT J AU Zhou, W Wang, Y Gao, H Jia, Y Xu, Y Wan, X Zhang, Z Yu, H Yan, Sh AF Zhou, Weiyu Wang, Yujing Gao, Hongmei Jia, Ying Xu, Yuanxin Wan, Xiaojing Zhang, Zhiying Yu, Haiqiao Yan, Shuang TI Identification of Key Genes Involved in Pancreatic Ductal Adenocarcinoma with Diabetes Mellitus Based on Gene Expression Profiling Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE diabetes mellitus; pancreatic ductal adenocarcinoma; meta-analysis; support vector machine; survival analysis ID diabetes mellitus; pancreatic ductal adenocarcinoma; meta-analysis; support vector machine; survival analysis AB This study aimed to identify key genes involved in the progression of diabetic pancreatic ductal adenocarcinoma (PDAC). Two gene expression datasets (GSE74629 and GSE15932) were obtained from Gene Expression Omnibus. Then, differentially expressed genes (DEGs) between diabetic PDAC and non-diabetic PDAC were identified, followed by a functional analysis. Subsequently, gene modules related to DM were extracted by weighed gene co-expression network analysis. The protein-protein interaction (PPI) network for genes in significant modules was constructed and functional analyses were also performed. After that, the optimal feature genes were screened by support vector machine (SVM) recursive feature elimination and SVM classification model was built. Finally, survival analysis was conducted to identify prognostic genes. The correlations between prognostic genes and other clinical factors were also analyzed. Totally, 1546 DEGs with consistent change tendencies were identified and functional analyses showed they were strongly correlated with metabolic pathways. Furthermore, there were two significant gene modules, in which RPS27A and UBA52 were key genes. Functional analysis of genes in two gene modules revealed that these genes primarily participated in oxidative phosphorylation pathway. Additionally, 21 feature genes were closely related with diabetic PDAC and the corresponding SVM classifier markedly distinguished diabetic PDAC from non-diabetic PDAC patients. Finally, decreased KIF22 and PYGL levels had good survival outcomes for PDAC. Four genes (RPS27A, UBA52, KIF22 and PYGL) might be involved in the pathogenesis of diabetic PDAC. Furthermore, KIF22 and PYGL acted as prognostic biomarkers for diabetic PDAC. C1 [Zhou, Weiyu] The Fourth Affiliated Hospital of Harbin Medical University, Department of EndocrinologyHarbin, China. [Wang, Yujing] The Fourth Affiliated Hospital of Harbin Medical University, Department of EndocrinologyHarbin, China. [Gao, Hongmei] The Fourth Affiliated Hospital of Harbin Medical University, Department of NeurologyHarbin, China. [Jia, Ying] The First Affiliated Hospital of Harbin Medical University, Department of PathologyHarbin, China. [Xu, Yuanxin] The Fourth Affiliated Hospital of Harbin Medical University, Department of EndocrinologyHarbin, China. [Wan, Xiaojing] The Fourth Affiliated Hospital of Harbin Medical University, Department of EndocrinologyHarbin, China. [Zhang, Zhiying] The Fourth Affiliated Hospital of Harbin Medical University, Department of EndocrinologyHarbin, China. [Yu, Haiqiao] The Fourth Affiliated Hospital of Harbin Medical University, Department of EndocrinologyHarbin, China. [Yan, Shuang] The Fourth Affiliated Hospital of Harbin Medical University, Department of EndocrinologyHarbin, China. RP Yan, Sh (reprint author), The Fourth Affiliated Hospital of Harbin Medical University, Department of Endocrinology, Harbin, China. EM qingmei0724@163.com CR Iozzo RV. The family of the small leucine-rich proteoglycans: key regulators of matrix assembly and cellular growth. Crit Rev Biochem Mol Biol, 1997). 32, 2): 141–74., DOI 10.3109/10409239709108551 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin, 2017). 67, 1):7–30., DOI 10.3322/caac.21387 Hine RJ, Srivastava S, Milner JA, Ross SA. Nutritional links to plausible mechanisms underlying pancreatic cancer: a conference report. Pancreas, 2003). 27, 4):356–66., DOI 10.1097/00006676-200311000-00014 Andersen DK, Andren-Sandberg A, Duell EJ, Goggins M, Korc M, Petersen GM, et al. Pancreatitis-diabetes-pancreatic cancer. Pancreas, 2013). 42, 8): 1227–37., DOI 10.1097/mpa.0b013e3182a9ad9d Maisonneuve P, Lowenfels AB. Risk factors for pancreatic cancer: a summary review of meta-analytical studies. Int J Epidemiol, 2015). 44, 1):186–98., DOI 10. 1093/ije/dyu240 Huxley R, Ansary-Moghaddam A, Berrington de Gonzalez A, Barzi F, Woodward M. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer, 2005). 92, 11):2076–83., DOI 10.1038/sj.bjc.6602619 Kleeff J, Costello E, Jackson R, Halloran C, Greenhalf W, Ghaneh P, et al. The impact of diabetes mellitus on survival following resection and adjuvant chemotherapy for pancreatic cancer. Br J Cancer, 2016). 115, 7):887–94., DOI 10.1038/bjc.2016.277 Lee S, Hwang HK, Kang CM, Lee WJ. Adverse oncologic impact of newonset diabetes mellitus on recurrence in resected pancreatic ductal adenocarcinoma: a comparison with long-standing and non-diabetes mellitus patients. Pancreas, 2018). 47, 7):816–22., DOI 10.1097/mpa. 0000000000001099 Gao W, Zhou Y, Li Q, Zhou Q, Tan L, Song Y, et al. Analysis of global gene expression profiles suggests a role of acute inflammation in type 3C diabetes mellitus caused by pancreatic ductal adenocarcinoma. Diabetologia, 2015). 58, 4):835–44., DOI 10.1007/s00125-014-3481-8 Sun Y, He W, Luo M, Zhou Y, Chang G, Ren W, et al. Role of transgelin-2 in diabetes-associated pancreatic ductal adenocarcinoma. Oncotarget, 2017). 8, 30):49592–604., DOI 10.18632/oncotarget.17519 Boursi B, Finkelman B, Giantonio BJ, Haynes K, Rustgi AK, Rhim AD, Mamtani R, et al. A clinical prediction model to assess risk for pancreatic cancer among patients with new-onset diabetes. Gastroenterology, 2017). 152, 4):840–50., DOI 10.1053/j.gastro.2016.11.046 Huang H, Dong X, Kang MX, Xu B, Chen Y, Zhang B, et al. Novel blood biomarkers of pancreatic cancer-associated diabetes mellitus identified by peripheral blood-based gene expression profiles. Am J Gastroenterol, 2010). 105, 7):1661–9., DOI 10.1038/ajg.2010.32 Kang M, Qin W, Buya M, Dong X, Zheng W, Lu W, et al. VNN1, a potential biomarker for pancreatic cancer-associated new-onset diabetes, aggravates paraneoplastic islet dysfunction by increasing oxidative stress. Cancer Lett, 2016). 373, 2):241–50., DOI 10.1016/j.canlet.2015.12.031 Barrett T, Troup DB, Wilhite SE, Ledoux P, Rudnev D, Evangelista C, et al. NCBI GEO: mining tens of millions of expression profiles--database and tools update. Nucleic Acids Res, 2007). 35:D760–D765., DOI 10.1093/nar/gkl887 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res, 2015). 43, 7):e47., DOI 10.1093/nar/gkv007 Parrish RS, Spencer HJ. Effect of normalization on significance testing for oligonucleotide microarrays. J Biopharm Stat, 2004). 14, 3):575–89., DOI 10. 1081/bip-200025650 Chang LC, Lin HM, Sibille E, Tseng GC. Meta-analysis methods for combining multiple expression profiles: comparisons, statistical characterization and an application guideline. BMC Bioinformatics, 2013). 14:368., DOI 10.1186/1471- 2105-14-368 Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID Bioinformatics Resources. Nat Protoc, 2009). 4, 1):44–57., DOI 10.1038/nprot.2008.211 Huang DW, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res, 2009). 37, 1):1–13., DOI 10.1093/nar/gkn923 Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics, 2008). 9:559., DOI 10.1186/1471-2105- 9-559 Szklarczyk D, Franceschini A,Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res, 2015). 43:D447–D452., DOI 10.1093/nar/gku1003 Shannon P, Ozier O, Baliga NS,Wang JT, Ramage D, Amin N, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res, 2003). 13, 11):2498–504., DOI 10.1101/gr.1239303 Lu X, Yang Y, Wu F, Gao M, Xu Y, Zhang Y, Yao Y, et al. Discriminative analysis of schizophrenia using support vector machine and recursive feature elimination on structural MRI images. Medicine, Baltimore,, 2016). 95, 30): e397310., DOI 10.1097/md.0000000000003973 Deist TM, Dankers FJ, Valdes G, Wijsman R, Hsu IC, Oberije C, Lustberg T, et al. Machine learning algorithms for outcome prediction in, chemo, radiotherapy: an empirical comparison of classifiers. Med Phys, 2018). 45, 7):3449–59., DOI 10.1002/mp.12967 Wang Q, Liu X. Screening of feature genes in distinguishing different types of breast cancer using support vector machine. Onco Targets Ther, 2015). 8: 2311–7., DOI 10.2147/OTT.S85271 Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez JC, Muller M., 2011). pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics 12:77., DOI 10.1186/1471-2105-12-77 Wang P, Wang Y, Hang B, Zou X, Mao JH. A novel gene expression-based prognostic scoring system to predict survival in gastric cancer. Oncotarget, 2016). 7, 34):55343–51., DOI 10.18632/oncotarget.10533 Weinberg SE, Chandel NS. Targeting mitochondria metabolism for cancer therapy. Nat Chem Biol, 2015). 11, 1):9–15., DOI 10.1038/nchembio.1712 Moreno-Sanchez R, Sara Rodriguez-Enriquez S, Marin-Hernandez A, Saavedra E. Energy metabolism in tumor cells. FEBS J, 2007). 274: 1393–418., DOI 10.1111/j.1742-4658.2007.05686.x Rademaker G, Costanza B, Anania S, Agirman F, Maloujahmoum N, Di Valentin E, Goval JJ, et al. Myoferlin contributes to themetastatic phenotype of pancreatic cancer cells by enhancing their migratory capacity through the control of oxidative phosphorylation. Cancers, 2019). 11, 6): 853., DOI 10.3390/ cancers11060853 Ashton TM, McKenna WG, Kunz-Schughart LA, Higgins GS. Oxidative phosphorylation as an emerging target in cancer therapy. Clin Cancer Res, 2018). 24, 11):2482–90., DOI 10.1158/1078-0432.ccr-17-3070 Viale A, Pettazzoni P, Lyssiotis CA, Ying H, Sanchez N, Marchesini M, Carugo A, et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature, 2014). 514, 7524):628–32., DOI 10.1038/ nature13611 Zhou C, Sun H, Zheng C, Gao J, Fu Q, Hu N, Shao X, et al. Oncogenic HSP60 regulates mitochondrial oxidative phosphorylation to support Erk1/2 activation during pancreatic cancer cell growth. Cell Death Dis, 2018). 9, 2):161., DOI 10.1038/s41419-017-0196-z Wada J, Nakatsuka AMitochondrial dynamics and mitochondrial dysfunction in diabetes. Acta Med Okayama, 2016). 70, 3):151–8., DOI 10.18926/AMO/ 54413 Haythorne E, Rohm M, van de Bunt M, Brereton MF, Tarasov AI, Blacker TS, et al. Diabetes causes marked inhibition of mitochondrial metabolism in pancreatic β-cells. Nat Commun, 2019). 10, 1):2474., DOI 10.1038/s41467- 019-10189-x Wong JM, Mafune K, Yow H, Rivers EN, Ravikumar TS, Steele GD, Jr, Chen LB, 1993). Ubiquitin-ribosomal protein S27a gene overexpressed in human colorectal carcinoma is an early growth response gene. Cancer Res 53, 8): 1916–20. Kanayama H TK, Tanaka K, Aki M, Kagawa S, Miyaji H, Satoh M, Okada F, et al. Changes in expressions of proteasome and ubiquitin genes in human renal cancer cells. Cancer Res, 1991). 51, 24):6677–85. Wang H YJ, Zhang L, Xiong Y, Chen S, Xing H, Tian Z, et al. RPS27a promotes proliferation, regulates cell cycle progression and inhibits apoptosis of leukemia cells. Biochem Biophysical Res Commun, 2014). 446, 4):1204–10., DOI 10.1016/j.bbrc.2014.03.086 Yang H, Zhang X, Xin G. Investigation of mechanisms of mesenchymal stem cells for treatment of diabetic nephropathy via construction of a miRNA-TFmRNA network. Ren Fail, 2018). 40, 1):136–45., DOI 10.1080/0886022x.2017. 1421556 Dihazi H, Muller GA, Lindner S, Meyer M, Asif AR, Oellerich M, Strutz F. Characterization of diabetic nephropathy by urinary proteomic analysis: identification of a processed ubiquitin form as a differentially excreted protein in diabetic nephropathy patients. Clin Chem, 2007). 53, 9): 1636–45., DOI 10.1373/clinchem.2007.088260 Wada J, Sun L, Kanwar YS. Discovery of genes related to diabetic nephropathy in various animal models by current techniques. Contrib Nephrol, 2011). 169: 161–74., DOI 10.1159/000313951 Tokai N F-NA, Toyoshima Y, Yonemura S, Tsukita S, Inoue J, Yamamota T. Kid, a novel kinesin-like DNA binding protein, is localized to chromosomes and the mitotic spindle. The EMBO J, 1996). 15, 3):457–67., DOI 10.1002/j. 1460-2075.1996.tb00378.x Yu Y, Wang XY, Sun L, Wang YL, Wan YF, Li XQ, Feng YM. Inhibition of KIF22 suppresses cancer cell proliferation by delaying mitotic exit through upregulating CDC25C expression. Carcinogenesis, 2014). 35, 6):1416–25., DOI 10.1093/carcin/bgu065 Zhang Z XH, Zhu S, Chen X, Yu J, Shen T, Li X, et al. High expression of KIF22/kinesin-like DNA binding protein, Kid, as a poor prognostic factor in prostate cancer patients. Med Sci Monit, 2018). 24:8190–7., DOI 10.12659/msm. 912643 Chen J CS, Situ B, Zhong J, Hu Y, Li S, Huang J, et al. Metabolic reprogramming-based characterization of circulating tumor cells in prostate cancer. J Exp Clin Cancer Res, 2018). 37, 1):127., DOI 10.1186/s13046-018- 0789-0 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 604730 EP 604739 DI 10.3389/pore.2021.604730 PG 10 ER PT J AU Moon, WS Son, JH Mo, YH Yoo, JN Lee, HS AF Moon, Won Seong Son, Ji Hyun Mo, Yoon Ha Yoo, Jin Nam Lee, Hyung Sug TI Somatic Mutation of NLRP Genes in Gastric and Colonic Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NLRP; Somatic mutation; loss of expression; cancer; colon cancer ID NLRP; Somatic mutation; loss of expression; cancer; colon cancer AB Nucleotide-binding and leucine-rich repeat protein (NLRP) genes are involved in inflammasome formation that plays a role in inflammation/host defense and cell death. Both cell death and inflammation are crucial for cancer development, but the roles of NLRPs in cancer are partially known. In this study, we analyzed mononucleotide repeats in coding sequences of NLRP1, NLRP2, NLRP4 and NLRP9, and found 1, 1, 1 and 8 frameshift mutation (s) in gastric (GC) and colonic cancers (CRC), respectively. Five of the 32 high microsatellite instability (MSI-H) GCs (15.5%) and 6 of 113 MSI-H CRCs (5.5%) exhibited the frameshift mutations. There was no NLRP frameshift mutations in microsatellite stable (MSS) GCs and CRCs. We also discovered that 2 of 16 CRCs (12.5%) harbored intratumoral heterogeneity (ITH) of the NLRP9 frameshift mutations in one or more areas. In both GC and CRC with MSI-H, NLRP9 expression in NLRP9-mutated cases was significantly lower than that in NLRP9-non-mutated cases. Our data indicate that NLRP9 is altered at multiple levels (frameshift mutation, mutational ITH and loss of expression), which together could contribute to pathogenesis of MSI-H GC and CRC. C1 [Moon, Won Seong] The Catholic University of Korea, College of Medicine, Department of PathologySeoul, South Korea. [Son, Ji Hyun] The Catholic University of Korea, College of Medicine, Department of PathologySeoul, South Korea. [Mo, Yoon Ha] The Catholic University of Korea, College of Medicine, Department of PathologySeoul, South Korea. [Yoo, Jin Nam] The Catholic University of Korea, College of Medicine, Department of PathologySeoul, South Korea. [Lee, Hyung Sug] The Catholic University of Korea, College of Medicine, Department of PathologySeoul, South Korea. RP Lee, HS (reprint author), The Catholic University of Korea, College of Medicine, Department of Pathology, Seoul, South Korea. EM suhulee@catholic.ac.kr CR Martinon F, Tschopp J. Inflammatory caspases and inflammasomes: master switches of inflammation. Cell Death Differ, 2007). 14:10–22., DOI 10.1038/sj. cdd.4402038 Karan D. Inflammasomes: emerging central players in cancer immunology and immunotherapy. Front Immunol, 2018). 9:3028., DOI 10.3389/fimmu.2018. 028 3. Place DE, Kanneganti T-D. Recent advances in inflammasome biology. Curr Opin Immunol, 2018). 50:32–8., DOI 10.1016/j.coi.2017.10.011 Chavarria-Smith J, Vance RE. The NLRP1 inflammasomes. Immunol Rev, 2015). 265:22–34., DOI 10.1111/imr.12283 Moossavi M, Parsamanesh N, Bahrami A, Atkin SL, Sahebkar A. Role of the NLRP3 inflammasome in cancer. Mol Cancer, 2018). 17:158., DOI 10.1186/ s12943-018-0900-3 Alimohammadi M, Bjorklund P, Hallgren A, Pontynen N, Szinnai G, Shikama N, et al. Autoimmune polyendocrine syndrome type 1 and NALP5, a parathyroid autoantigen. N Engl J Med, 2008). 358:1018–28., DOI 10.1056/ nejmoa0706487 Franchi L, Eigenbrod T, Munoz-Planillo R, Nunez G. The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol, 2009). 10:241–7., DOI 10.1038/ni.1703 Allen IC, TeKippe EM, Woodford R-MT, Uronis JM, Holl EK, Rogers AB, et al. The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med, 2010). 207:1045–56., DOI 10.1084/ jem.20100050 Williams TM, Leeth RA, Rothschild DE, Coutermarsh-Ott SL, McDaniel DK, Simmons AE, et al. The NLRP1 inflammasome attenuates colitis and colitisassociated tumorigenesis. J Immunol, 2015). 194:3369–80., DOI 10.4049/ jimmunol.1402098 Sand J, Fenini G, Grossi S, Hennig P, Di FilippoM, LevesqueM, et al. The NLRP1 inflammasome pathway is silenced in cutaneous squamous cell carcinoma. J Invest Dermatol, 2019). 139:1788–97., DOI 10.1016/j.jid.2019.01.025 Wu G-Q, Liao Y-J, Qin Z-Q, He L-R, Chen Y-C, Zeng Y-X, et al. PYRIN domain of NALP2 inhibits cell proliferation and tumor growth of human glioblastoma. Plasmid, 2010). 64:41–50., DOI 10.1016/j.plasmid.2010.01.003 Karki R, Man SM, Malireddi RKS, Kesavardhana S, Zhu Q, Burton AR, et al. NLRC3 is an inhibitory sensor of PI3K-mTOR pathways in cancer. Nature, 2016). 540:583–7., DOI 10.1038/nature20597 Wei Y, Huang H, Qiu Z, Li H, Tan J, Ren G, et al. NLRP1 overexpression is correlated with the tumorigenesis and proliferation of human breast tumor. Biomed Res Int, 2017). 2017:4938473., DOI 10.1155/2017/4938473 Zhai Z, Liu W, Kaur M, Luo Y, Domenico J, Samson JM, et al. NLRP1 promotes tumor growth by enhancing inflammasome activation and suppressing apoptosis in metastatic melanoma. Oncogene, 2017). 36: 3820–30., DOI 10.1038/onc.2017.26 Iyer RR, Pluciennik A, Burdett V, Modrich PL. DNA mismatch repair: functions and mechanisms. Chem Rev, 2006). 106:302–23., DOI 10.1021/ cr0404794 Imai K, Yamamoto H. Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis, 2008). 29:673–80., DOI 10.1093/carcin/bgm228 Kim T-M, Laird PW, Park PJ. The landscape of microsatellite instability in colorectal and endometrial cancer genomes. Cell, 2013). 155:858–68., DOI 10. 1016/j.cell.2013.10.015 Ionov Y, Yamamoto H, Krajewski S, Reed JC, Perucho M. Mutational inactivation of the proapoptotic gene bax confers selective advantage during tumor clonal evolution. Proc Natl Acad Sci, 2000). 97:10872–7., DOI 10.1073/pnas.190210897 Markowitz S,Wang J,Myeroff L, Parsons R, Sun L, Lutterbaugh J, et al. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science, 1995). 268:1336–8., DOI 10.1126/science.7761852 Murphy KM, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, et al. Comparison of the microsatellite instability analysis system and the Bethesda panel for the determination of microsatellite instability in colorectal cancers. J Mol Diagn, 2006). 8:305–11., DOI 10.2353/jmoldx.2006. 050092 Mo HY, An CH, Choi EJ, Yoo NJ, Lee SH. Somatic mutation and loss of expression of a candidate tumor suppressor gene TET3 in gastric and colorectal cancers. Pathol Res Pract, 2020). 216:152759., DOI 10.1016/j.prp. 2019.152759 An CH, Je EM, Yoo NJ, Lee SH. Frameshift mutations of cadherin genes DCHS2, CDH10 and CDH24 genes in gastric and colorectal cancers with high microsatellite instability. Pathol Oncol Res, 2015). 21:181–5., DOI 10.1007/ s12253-014-9804-8 Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell, 2011). 144:646–74., DOI 10.1016/j.cell.2011.02.013 Dalbies-Tran R, Papillier P, Pennetier S, Uzbekova S, Monget P. Bovinematerlike NALP9 is an oocyte marker gene. Mol Reprod Dev, 2005). 71:414–21., DOI 10.1002/mrd.20298 Zhu S, Ding S, Wang P, Wei Z, Pan W, Palm NW, et al. Nlrp9b inflammasome restricts rotavirus infection in intestinal epithelial cells. Nature, 2017). 546: 667–70., DOI 10.1038/nature22967 Poli G, Brancorsini S, Cochetti G, Barillaro F, Egidi MG, Mearini E. Mearini, Expression of inflammasome-related genes in bladder cancer and their association with cytokeratin 20 messenger RNA. Urol Oncol, 2015). 33:505., DOI 10.1016/j.urolonc.2015.07.012 Jounai N, Kobiyama K, Shiina M, Ogata K, Ishii KJ, Takeshita F. NLRP4 negatively regulates autophagic processes through an association with beclin1. J.I., 2011). 186:1646–55., DOI 10.4049/jimmunol.1001654 Ahn CH, Jeong EG, Lee JW, Kim MS, Kim SH, Kim SS, et al. Expression of beclin-1, an autophagy-related protein, in gastric and colorectal cancers. APMIS, 2007). 115:1344–9., DOI 10.1111/j.1600-0463.2007.00858.x Ramon Y, Cajal S, Sese M, Capdevila C, Aasen T, De Mattos-Arruda L, et al. Clinical implications of intratumor heterogeneity: challenges and opportunities. J Mol Med, Berlin,, 2020). 98:161–77., DOI 10.1007/s00109- 020-01874-2 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 607385 EP 607391 DI 10.3389/pore.2021.607385 PG 7 ER PT J AU Zhang, X Zhang, H Li, J Ma, X He, Z Liu, C Gao, Ch Li, H Wang, X Wu, J AF Zhang, Xiaoming Zhang, Haiyan Li, Jie Ma, Xiaoran He, Zhengguo Liu, Cun Gao, Chundi Li, Huayao Wang, Xue Wu, Jibiao TI 6-lncRNA Assessment Model for Monitoring and Prognosis of HER2-Positive Breast Cancer: Based on Transcriptome Data SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE human epidermal growth factor receptor 2-positive breast cancer; random forests; prognosis; long noncoding RNA; competing endogenous RNA ID human epidermal growth factor receptor 2-positive breast cancer; random forests; prognosis; long noncoding RNA; competing endogenous RNA AB Background: In view of the high malignancy and poor prognosis of human epidermal growth factor receptor 2 (HER2)-positive breast cancer, we analyzed the RNA expression profiles of HER2-positive breast cancer samples to identify the new prognostic biomarkers. Methods: The linear fitting method was used to identify the differentially expressed RNAs from the HER2-positive breast cancer RNA expression profiles in the Cancer Genome Atlas (TCGA). Then, a series of methods including univariate Cox, Kaplan-Meier, and random forests, were used to identify the core long non-coding RNAs (lncRNAs) with stable prognostic value for HER2-positive breast cancer. A clinical feature analysis was performed, and a competing endogenous RNA network was constructed to explore the role of these core lncRNAs in HER2-positive breast cancer. In addition, a functional analysis of differentially expressed messenger RNAs in HER-2 positive breast cancer also provided us with some enlightening insights. Results: The high expression of four core lncRNAs (AC010595.1, AC046168.1, AC069277.1, and AP000904.1) was associated with worse overall survival, while the low expression of LINC00528 and MIR762HG was associated with worse overall survival. The 6-lncRNA model has an especially good predictive power for overall survival (p < 0.0001) and 3-year survival (the area under the curve = 0.980) in HER2-positive breast cancer patients. Conclusion: This study provides a new efficient prognostic model and biomarkers of HER2-positive breast cancer. Meanwhile, it also provides a new perspective for elucidating the molecular mechanisms underlying HER2-positive breast cancer. C1 [Zhang, Xiaoming] Shandong University of Traditional Chinese Medicine, College of Chinese MedicineJinan, China. [Zhang, Haiyan] Shandong University of Traditional Chinese Medicine, College of Chinese MedicineJinan, China. [Li, Jie] Shandong University of Traditional Chinese Medicine, College of Chinese MedicineJinan, China. [Ma, Xiaoran] Shandong University of Traditional Chinese Medicine, The First Clinical Medical CollegeJinan, China. [He, Zhengguo] Columbus Technical CollegeColumbus, GA, USA. [Liu, Cun] Shandong University of Traditional Chinese Medicine, The First Clinical Medical CollegeJinan, China. [Gao, Chundi] Shandong University of Traditional Chinese Medicine, The First Clinical Medical CollegeJinan, China. [Li, Huayao] Shandong University of Traditional Chinese Medicine, College of Chinese MedicineJinan, China. [Wang, Xue] Qingdao University, College of Basic MedicineQingdao, China. [Wu, Jibiao] Shandong University of Traditional Chinese Medicine, College of Chinese MedicineJinan, China. RP Wu, J (reprint author), Shandong University of Traditional Chinese Medicine, College of Chinese Medicine, Jinan, China. EM wujibiao1963@163.com CR Wolff AC, Hammond MEH, Hicks DG, Dowsett M, McShane LM, Allison KH, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. Arch Pathol Lab Med, 2013, 138(2):241–56., DOI 10. 5858/arpa.2013-0953-sa Sun Y, Li W, Li A-J, Su H, Yue J, and Yu J. Increased systemic immuneinflammation index independently predicts poor survival for hormone receptor-negative, HER2-positive breast cancer patients. Cmar, 2019, 11: 3153., DOI 10.2147/cmar.s190335 Badache A, and Goncalves A. The ErbB2 signaling network as a target for breast cancer therapy. J Mammary Gland Biol Neoplasia, 2006, 11(1):13–25., DOI 10.1007/s10911-006-9009-1 Parise CA, and Caggiano V. Breast cancer survival defined by the ER/PR/ HER2 subtypes and a surrogate classification according to tumor grade and immunohistochemical biomarkers. J Cancer Epidemiol, 2014, 2014:1–11., DOI 10.1155/2014/469251 Cerk S, Schwarzenbacher D, Adiprasito J, Stotz M, Hutterer G, Gerger A, et al. Current status of long non-coding RNAs in human breast cancer. Ijms, 2016, 17(9):1485., DOI 10.3390/ijms17091485 Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, et al. Chromatin signature reveals over a thousand highly conserved large noncoding RNAs in mammals. Nature, 2009, 458(7235):223., DOI 10.1038/ nature07672 Hung T, and Chang HY. Long noncoding RNA in genome regulation. RNA Biol, 2010, 7(5):582–5., DOI 10.4161/rna.7.5.13216 Fatica A, and Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet, 2014, 15(1):7., DOI 10. 1038/nrg3606 Gutschner T, and Diederichs S. The hallmarks of cancer. RNA Biol, 2012, 9(6): 703–19., DOI 10.4161/rna.20481 Huarte M. The emerging role of lncRNAs in cancer. Nat Med, 2015, 21(11): 1253., DOI 10.1038/nm.3981 Lipovich L, Johnson R, and Lin C-Y. MacroRNA underdogs in a microRNA world: evolutionary, regulatory, and biomedical significance of mammalian long non-protein-coding RNA. Biochim Biophys Acta, Bba, - Gene Regul Mech, 2010, 1799(9):597–615., DOI 10.1016/j.bbagrm.2010.10.001 Wang Z, Jin Y, Ren H, Ma X, Wang B, and Wang Y. Downregulation of the long non-coding RNA TUSC7 promotes NSCLC cell proliferation and correlates with poor prognosis. Am J Transl Res, 2016, 8(2):680 Zhao J, Liu Y, Huang G, Cui P, Zhang W, and Zhang Y. Long non-coding RNAs in gastric cancer: versatile mechanisms and potential for clinical translation. Am J Cancer Res, 2015, 5(3):907–27. Bao X, Anastasov N, Wang Y, and Rosemann M. A novel epigenetic signature for overall survival prediction in patients with breast cancer. J Transl Med, 2019, 17(1):380., DOI 10.1186/s12967-019-2126-6 Zhang D, Duan Y, Cun J, and Yang Q. Identification of prognostic alternative splicing signature in breast carcinoma. Front Genet, 2019, 10:278., DOI 10.3389/ fgene.2019.00278 Leung F, Diamandis EP, and Kulasingam V. Advances in clinical chemistry, 66. Elsevier, 2014), Ovarian cancer biomarkers: current state and future implications from high-throughput technologies. p. 25–77. Gao C, Li H, Zhuang J, Zhang H, Wang K, Yang J, et al. The construction and analysis of ceRNA networks in invasive breast cancer: a study based on the Cancer Genome Atlas. Cancer Manag Res, 2019, 11:1., DOI 10.2147/CMAR. S182521 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res, 2015, 43(7):e47., DOI 10.1093/ nar/gkv007 Gentleman R, and Ihaka R. R: a language and environment for statistical computing. Computing, 2011, 1:12–21. Zhang X, Gao C, Liu L, Zhou C, Liu C, Li J, et al. DNA methylation-based diagnostic and prognostic biomarkers of nonsmoking lung adenocarcinoma patients. J Cell Biochem, 2019). 120(8):13520–30., DOI 10.1002/jcb.28627 Liu J, Li H, Sun L, Wang Z, Xing C, and Yuan Y. Aberrantly methylateddifferentially expressed genes and pathways in colorectal cancer. Cancer Cel Int, 2017, 17(1):75., DOI 10.1186/s12935-017-0444-4 Bader GD, and Hogue CW. An automated method for finding molecular complexes in large protein interaction networks. BMC bioinformatics, 2003, 4: 2., DOI 10.1186/1471-2105-4-2 Sacrdoni G, Petterlini M, and Laudanna C. Analyzing biological network parameters with CentiScaPe. Bioinformatics, Oxford, England,, 2009, 25(21): 2857–9. 10.1093/bioinformatics/btp517 Lin H, and Zelterman D. Modeling survival data: extending the Cox model. Technometrics, 2000, 44(1):85–6. Ishwaran H, Kogalur UB, and Kogalur MUB. Package ‘randomForestSRC’. Hemant Ishwaran and Udaya B. Kogalur, 2019). Available at: https://kogalur. github.io/randomForestSRC/. Heagerty PJ, and Saha P. time-dependent ROC curve estimation from censored survival data. Biometrics,, 2013).66(4):999-1011. 10.1111/j.1541- 0420.2009.01375.x Langfelder P, and Horvath S. Fast R functions for robust correlations and hierarchical clustering. J Stat Softw, 2012, 46 Wang J-D, Zhou H-S, Tu X-X, He Y, Liu Q-F, Liu Q, et al. Prediction of competing endogenous RNA coexpression network as prognostic markers in AML. Aging, 2019, 11(10):3333., DOI 10.18632/ aging.101985 Xu P, Yang J, Liu J, Yang X, Liao J, Yuan F, et al. Identification of glioblastoma gene prognosis modules based on weighted gene co-expression network analysis. BMC Med genomics, 2018, 11(1):96., DOI 10.1186/s12920-018- 0407-1 Tang Z, Li C, Kang B, Gao G, Li C, and Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res, 2017, 45(W1):W98–W102., DOI 10.1093/ nar/gkx247 Hu X, Stern HM, Ge L, O’Brien C, Haydu L, Honchell CD, et al. Genetic alterations and oncogenic pathways associated with breast cancer subtypes. Mol Cancer Res, 2009, 7(4):511–22., DOI 10.1158/1541-7786. mcr-08-0107 Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, Frederick AM, et al. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature, 2012, 486(7403):405., DOI 10.1038/nature11154 Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature, 2012, 486(7403):395., DOI 10.1038/nature10933 Stephens PJ, Tarpey PS, Tarpey PS, Davies H, Van Loo P, Greenman C, et al. The landscape of cancer genes and mutational processes in breast cancer. Nature, 2012, 486(7403):400., DOI 10.1038/nature11017 Gy}orffy B, Pongor L, Bottai G, Li X, Budczies J, Szabo A, et al. An integrative bioinformatics approach reveals coding and non-coding gene variants associated with gene expression profiles and outcome in breast cancermolecular subtypes. Br J Cancer, 2018, 118(8):1107., DOI 10.1038/s41416-018-0030-0 Zeng P, Sun S, Li R, Xiao Z-X, and Chen H. HER2 upregulates ATF4 to promote cell migration via activation of ZEB1 and downregulation of E-cadherin. Ijms, 2019, 20(9):2223., DOI 10.3390/ijms20092223 Greenshields AL, Fernando W, and Hoskin DW. The anti-malarial drug artesunate causes cell cycle arrest and apoptosis of triple-negative MDAMB- 468 and HER2-enriched SK-BR-3 breast cancer cells. Exp Mol Pathol, 2019, 107:10–22., DOI 10.1016/j.yexmp.2019.01.006 Garcia D, and Shaw RJ. AMPK: mechanisms of cellular energy sensing and restoration of metabolic balance. Mol Cel, 2017, 66(6):789–800., DOI 10.1016/j. molcel.2017.05.032 Cao W, Li J, Hao Q, Vadgama JV, Wu Y, and Wu Y. AMP-activated protein kinase: a potential therapeutic target for triple-negative breast cancer. Breast Cancer Res, 2019, 21(1):29., DOI 10.1186/s13058-019-1107-2 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 609083 EP 609095 DI 10.3389/pore.2021.609083 PG 13 ER PT J AU Behzatoglu, K AF Behzatoglu, Kemal TI Osteoclasts in Tumor Biology: Metastasis and Epithelial- Mesenchymal-Myeloid Transition SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE tumor biology; metastasis; breast carcinoma metastasis; giant cell tumor of bone; EMMT; osteoclast; epithelial-mesenchymal-myeloid transition ID tumor biology; metastasis; breast carcinoma metastasis; giant cell tumor of bone; EMMT; osteoclast; epithelial-mesenchymal-myeloid transition AB Osteoclast is a specialized cell that originates from monocytic lineage, communicates closely with osteoblasts under physiological conditions, participates in bone modeling and remodeling, contributes to calcium homeostasis and osteoimmunity. In pathological conditions, it is involved in many tumors such as giant cell bone tumor (osteoclastoma), aneurysmal bone cyst, osteosarcoma, and metastatic cancers, and it usually causes local spread and progression of the tumor, working against the host. Since osteoclasts play an active role in primary bone tumors and bone metastases, the use of anti-osteoclastic agents significantly reduces the mortality and morbidity rates of patients by preventing the progression and local spread of tumors. Osteoclasts also accompany undifferentiated carcinomas of many organs, especially pancreas, thyroid, bladder and ovary. Undifferentiated carcinomas rich in osteoclasts have osteoclastoma-like histology. In these organs, osteoclastoma-like histology may accompany epithelial carcinomas, and de novo, benign and borderline tumors. Mature and immature myeloid cells, including osteoclasts, play an active role in the tumor progression in primary and metastatic tumor microenvironment, in epithelial-mesenchymal transition (EMT), mesenchymal-epithelialtransition (MET), and cancer stem cell formation. Additionally, they are the most suitable candidates for cancer cells in cell fusion due to their evolutionary fusion capabilities. Myeloid features and markers (CD163, CD33, CD68 etc.) can be seen in metastatic cancer cells. Consequently, they provide metastatic cancer cells with motility, margination, transmigration, chemotaxis, phagocytosis, angiogenesis, matrix degradation, and resistance to chemotherapy. For these reasons, we think that the concept of Epithelial-Mesencyhmal-Myeloid-Transition (EMMT) will be more accurate than EMT for cancer cells with myeloid properties. C1 [Behzatoglu, Kemal] Acibadem University, School of Medicine, Pathology DepartmentIstanbul, Turkey. RP Behzatoglu, K (reprint author), Acibadem University, School of Medicine, Pathology Department, Istanbul, Turkey. EM kbehzatoglu@hotmail.com CR Scully OJ, Bay BH, Yip G, and Yu Y. Breast cancer metastasis. Cancer Genomics Proteomics, 2012, 9(5):311–20. Lorusso G, and Ruegg C. New insights into the mechanisms of organ-specific breast cancer metastasis. Semin Cancer Biol, 2012, 22(3):226–33., DOI 10. 1016/j.semcancer.2012.03.007 Xiao L, and Xiao Y. The autophagy in osteoimmunology: self-eating, maintance, and beyond. Front Endocrinol, Lausanne,, 2019, 10:490., DOI 10.3389/fendo.2019.00490 Madel M-B, Ibanez L, Wakkach A, de Vries TJ, Teti A, Apparaily F, et al. Immune function and diversty of osteoclasts in normal and pathological conditions. Front Immunol, 2019, 10:1408., DOI 10.3389/fimmu.2019.01408 Blin-Wakkach C, and de Vries TJ. Editorial: advances im osteoimmunology. Front Immunol, 2019, 10:2595., DOI 10.3389/fimmu.2019.02595 Mori G, D’Amelio P, Faccio R, and Brunetti G. The Interplay between the bone and the immune system. Clin Dev Immunol, 2013, 2013:720504., DOI 10. 1155/2013/720504 Feng X, and Teitelbaum SL. Osteoclasts: new insights. Bone Res, 2013, 1(1): 11–26., DOI 10.4248/BR201301003 Tsukasaki M, and Takayanagi H. Osteoimmunology: evolving concepts in bone-immune interactions in health and disease. Nat Rev Immunol, 2019, 19(10):626–42., DOI 10.1038/s41577-019-0178-8 Boullion R, and Suda T. Vitamin D: calcium and bone homeostasis during evolution. Bonekey Rep, 2014, 3:480., DOI 10.1038/bonekey. 2013.214 Weiss MC, Sousa FL,Mrnjavac N, Neukirchen S, Roettger M, Nelson-Sathi S, et al. The physiology and habitat of the last universal common ancestor. Nat Microbiol, 2016, 1(9):16116., DOI 10.1038/nmicrobiol.2016.116 Kong Y-Y, Yoshida H, Sarosi I, Tan H-L, Timms E, Capparelli C, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature, 1999, 397(6717):315–23., DOI 10. 1038/16852 Hikosaka Y, Nitta T, Ohigashi I, Yano K, Ishimaru N, Hayashi Y, et al. The cytokine RANKL produced by positively selected thymocytes fosters medullary thymic epithelial cells that express autoimmune regulator. Immunity, 2008, 29(3):438–50., DOI 10.1016/j.immuni.2008.06.018 Nagashima K, Sawa S, Nitta T, TsutsumiM, Okamura T, Penninger JM, et al. Idendification sub epithelial mesenchymal cells that induce IgA and diversity gut microbiata. Nat Immunol, 2017, 18(6):65–680., DOI 10.1038/ni.3732 Lampiasi N, Russo R, and Zito F. The alternative faces of macrophage generate osteoclasts. Biomed Res Int, 2016, 2016:9089610., DOI 10.1155/ 2016/9089610 Mansour A, Wakkach A, and Bli-Wakkach C. Emerging roles of osteoclast in the modulation of bone microenvironment and immune suppression in multipl myeloma. Front Immunol, 2017, 8:954., DOI 10.3389/fimmu.2017. 00954 Wu M-Y, Li C-J, Yiang G-T, Cheng Y-L, Tsai AP-Y, Hou Y-T, et al. Molecular regulation of bone metastasis pathogenesis. Cell Physiol Biochem, 2018, 46(4):1423–38., DOI 10.1159/000489184 Yago T, Nanke Y, Kawamoto M, Furuya T, Kobashigawa T, Kamatani N, et al. IL-23 induces human osteoclastogenesis via IL-17 in vitro, and anti-IL-23 antibody attenuates collagen-induced arthritis in rats. Arthritis Res Ther, 2019, 9(5):R96., DOI 10.1186/ar2297 Sims JE, and Smith DE. The IL-1 family: regulators of immunity. Nat Rev Immunol, 2010, 10(2):89–102., DOI 10.1038/nri2691 Kylmaoja E, Nakamura M, Turunen S, Patlaka C, Andersson G, Lehenkari P, et al. Peripheral blood monocytes show increased osteoclast differentiation potential compared to bone marrow monocytes. Heliyon, 2018, 4(9):e00780., DOI 10.1016/j.heliyon.2018.e00780 Chen Z, Buki K, Vaaraniemi J, Gu G, and Vaananen HK. The critical role of IL-34 in osteoclastogenesis. PLoS One, 2011, 6(4):e18689., DOI 10.1371/ journal.pone.0018689 Sprangers S, Schoenmaker T, Cao Y, Everts V, and de Vries TJ. Different blood-borne human osteoclast precursors respond in distinct ways to IL-17A. J Cel Physiol, 2016, 231(6):1249–60., DOI 10.1002/jcp.25220 Sherr C. Colony-stimulating factor-1 receptor. Blood, 1990, 75:1–12., DOI 10. 1182/blood.v75.1.1.bloodjournal7511 Heiland GR, Zwerina K, Baum W, Kireva T, Distler JH, Grisanti M, et al. Neutralisation of Dkk-1 protects from systemic bone loss during inflammation and reduces sclerostin expression. Ann Rheum Dis, 2010, 69(12):2152–9., DOI 10.1136/ard.2010.132852 Furukawa M, Takaishi H, Takito J, Yoda M, Sakai S, Hikata T, et al. IL-27 abrogates receptor activator of NF-κB Ligand-mediated osteoclastogenesis of human granulocyte-macrophage Colony-Forming unit cells through STAT1- Dependent inhibition of c-fos. J Immunol, 2009, 183(4):2397–406., DOI 10. 4049/jimmunol.0802091 Wu L, Luo Z, Liu Y, Jia L, Jiang Y, Du J, et al. Aspirin inhibits RANKLinduced osteoclast differentiation in dendritic cells by suppressing NF-κB and NFATc1 activation. Stem Cel Res Ther, 2019, 10(1):375., DOI 10.1186/s13287- 019-1500-x Seong S, Kim JH, and Kim N. Pro-inflammatory cytokines modulating osteoclast differentiation and function. J Rheum Dis, 2016, 23:148–53., DOI 10.4078/jrd.2016.23.3.148 D’Oronzo S, Coleman R, Brown J, and Silvestris F. Metastatic bone disease: pathogenesis and therapeutic options: up-date on bone metastasis management. J Bone Oncol, 2019, 15:004–4., DOI 10.1016/j. jbo.2018.10.004 Li H, Hong S, Qian J, Zheng Y, Yang J, and Yi Q. Cross talk between the bone and immune systems: osteoclasts function as antigen-presenting cells and activate CD4+ and CD8+ T cells. Blood, 2010, 116(2):210–7., DOI 10.1182/ blood-2009-11-255026 Maurizi A, and Rucci N. The osteoclast in bone metastasis: player and target. Cancers, 2018, 10(7):218., DOI 10.3390/cancers10070218 Coleman RE. Metastatic bone disease: Clinical features, pathophysiology and treatment strategies. Cancer Treat Rev, 2001, 27(3):165–76., DOI 10.1053/ctrv. 2000.0210 Paget S. The Distribution of secondary growths in cancer of the Breast. The Lancet, 1889, 133:571–3., DOI 10.1016/s0140-6736(00)49915-0 Ewing J. A treatise on tumours. 3RD ed.. Philadelphia: W.B. Saunders, 1928) Erler JT, Bennewith KL, Cox TR, Lang G, Bird D, Koong A, et al. Hypoxiainduced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell, 2009, 15(1):35–44., DOI 10.1016/j. ccr.2008.11.012 Kaplan RN, Psaila B, and Lyden D. Bone marrow cells in the ’pre-metastatic niche’: within bone and beyond. Cancer Metastasis Rev, 2006, 25(4):521–9., DOI 10.1007/s10555-006-9036-9 Timar J, Csuka O, Orosz Z, Jeney A, and Kopper L. Molecular pathology of tumor metastasis. Pathol Oncol Res, 2001, 7(3):217–30., DOI 10.1007/ bf03032353 Rucci N, and Teti A. Osteomimicry: how the seed grows in the soil. Calcif Tissue Int, 2018, 102(2):131–40., DOI 10.1007/s00223-017-0365-1 Fata JE, Kong Y-Y, Li J, Sasaki T, Irie-Sasaki J, Moorehead RA, et al. The osteoclast differentiation factor osteoprotegerin-ligand is essential for mammary gland development. Cell, 2000, 103(1):41–50., DOI 10.1016/ s0092-8674(00)00103-3 Cao Y, Bonizzi G, Seagroves TN, Greten FR, Johnson R, Schmidt EV, et al. IKKα Provides an Essential Link between RANK Signaling and Cyclin D1 Expression during Mammary Gland Development. Cell, 2001, 107(6): 763–75., DOI 10.1016/s0092-8674(01)00599-2 Yoneda T, Tanaka S, and Hata K. Role of RANKL/RANK in primary and secondary breast cancer. Wjo, 2013, 4(4):178–85., DOI 10.5312/wjo.v4.i4.178 Palafox M, Ferrer I, Pellegrini P, Vila S, Hernandez-Ortega S, Urruticoechea A, et al. RANK induces epithelial-mesenchymal transition and stemness in human mammary epithelial cells and promotes tumorigenesis and metastasis. Cancer Res, 2012, 72:2879–88., DOI 10.1158/0008-5472.can-12- 0044 Shemanko C, Cong Y, and Forsyth A.What Is breast in the bone?. Ijms, 2016, 17(10):1764., DOI 10.3390/ijms17101764 Mundy GR. Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer, 2002, 2:584–93., DOI 10.1038/nrc867 Mori K, Le Goff B, Charrier C, Battaglia S, Heymann D, and Redini F. DU145 human prostate cancer cells express functional receptor activator of NFκB: new insights in the prostate cancer bone metastasis process. Bone, 2007, 40(4):981–90., DOI 10.1016/j.bone.2006.11.006 Kopper L. Denosumab-A powerful RANKL inhibitor to stop lytic metastases and other bone loss actions by osteoclasts. Pathol Oncol Res, 2012, 18(4): 743–7., DOI 10.1007/s12253-012-9538-4 van Dam PA, Verhoeven Y, Trinh XB, Wouters A, Lardon F, Prenen H, et al. RANK/RANKL signaling inhibition may improve the effectiveness of checkpoint blockade in cancer treatment. Crit Rev Oncology/Hematology, 2019, 133:85–91., DOI 10.1016/j.critrevonc.2018.10.011 Schramm HM. Should EMT of cancer cells be understood as epithelialmyeloid transition?. J Cancer, 2014, 5(2):125–32., DOI 10.7150/jca.8242 Schramm HM. The epithelial-myeloid-transition, EMyeT, of cancer cells as a wrongly perceived primary inflammatory process eventually progressing to a bone remodeling malignancy: the alternative pathway for epithelialmesenchymal- transition hypothesis, EMT)?. J Cancer, 2019, 10:3798–809., DOI 10.7150/jca.31364 Cowan RW, and Singh G. Giant cell tumor of bone: a basic science perspective. Bone, 2013, 52(1):238–46., DOI 10.1016/j.bone.2012.10.002 Gupta R, Seethalakshmi V, Jambhekar NA, Prabhudesai S, Merchant N, Puri A, et al. Clinicopathologic profile of 470 giant cell tumors of bone from a cancer hospital in western India. Ann Diagn Pathol, 2008, 12:239–48., DOI 10. 1016/j.anndiagpath.2007.09.002 Unni KK, and Inwards CY. Giant Cell Tumor, Osteoclastoma). Dahlin’s Bone Tumors; General aspects and data on 10,165 Cases. New York: L˙Ippincott Willams & Wilkins, 2010). p. 225–42. Behjati S, Tarpey PS, Presneau N, Scheipl S, Pillay N, Van Loo P, et al. Distinct H3F3A and H3F3B driver mutations define chondroblastoma and giant cell tumor of bone. Nat Genet, 2013, 45:1479–82., DOI 10.1038/ng.2814 Salerno M, Avnet S, Alberghini M, Giunti A, and Baldini N. Histogenetic characterization of giant cell tumor of bone. Clin Orthop Relat Res, 2008, 466: 2081–91., DOI 10.1007/s11999-008-0327-z Atkins GJ, Haynes DR, Graves SE, Evdokiou A, Hay S, Bouralexis S, et al. Expression of osteoclast differentiation signals by stromal elements of giant cell tumors. J Bone Miner Res, 2000, 15(4):640–9., DOI 10.1359/jbmr.2000.15. 4.640 Kim Y, Nizami S, Goto H, and Lee FY. Modern interpretation of giant cell tumor of bone: predominantly osteoclastogenic stromal tumor. Clin Orthop Surg, 2012, 4(2):107–16., DOI 10.4055/cios.2012.4.2.107 Chiu Y-H, Mensah KA, Schwarz EM, Ju Y, Takahata M, Feng C, et al. Regulation of human osteoclast development by dendritic cell-specific transmembrane protein, DC-STAMP). J Bone Miner Res, 2012, 27(1): 79–92., DOI 10.1002/jbmr.531 Nakashima M, Nakayama T, Ohtsuru A, Fukada E, Niino D, Yamazumi K, et al. Expression of parathyroid hormone, PTH)-related peptide, PTHrP, and PTH/PTHrP receptor in osteoclast-like giant cells. Pathol - Res Pract, 2003, 199(2):85–92., DOI 10.1078/0344-0338-00359 Zhou Z, Li Y, Xu L, Wang X, Chen S, Yang C, et al. Biological characteristics of a novel giant cell tumor cell line derived from spine. Tumor Biol, 2016, 37(7):9681–9., DOI 10.1007/s13277-016-4867-7 Haute Autorite de Sante XGEVA, denosumab). anticorps monoclonal, 2015).Available at: www.has-sante.fr/portail/jcms/c_2046978/fr/xgevadenosumab- anticorps-monoclonal(Accessed April 27, 2018) Thomas D, Henshaw R, Skubitz K, Chawla S, Staddon A, Blay J-Y, et al. Denosumab in patients with giant-cell tumour of bone: an open-label, phase 2 study. Lancet Oncol, 2010, 11(3):275–80., DOI 10.1016/s1470-2045(10, 70010-3 Branstetter DG, Nelson SD, Manivel JC, Blay J-Y, Chawla S, Thomas DM, et al. Denosumab induces tumor reduction and bone formation in patients with giant-cell tumor of bone. Clin Cancer Res, 2012, 18(16):4415–24., DOI 10. 1158/1078-0432.ccr-12-0578 Hakozaki M, Tajino T, Yamada H, Hasegawa O, Tasaki K, Watanabe K, et al. Radiological and pathological characteristics of giant cell tumor of bone treated with denosumab. Diagn Pathol, 2014, 9(1):111., DOI 10.1186/1746- 1596-9-111 Wojcik J, Rosenberg AE, Bredella MA, Choy E, Hornicek FJ, Nielsen GP, et al. Denosumab-treated giant cell tumor of bone exhibits morphologic overlap with malignant giant cell tumor of bone. Am J Surg Pathol, 2016, 40(1): 72–80., DOI 10.1097/pas.0000000000000506 Roitman PD, Jauk F, Farfalli GL, Albergo JI, and Aponte-Tinao LA. Denosumab-treated giant cell tumor of bone. Its histologic spectrum and potential diagnostic pitfalls. Hum Pathol, 2017, 63:89–97., DOI 10.1016/j. humpath.2017.02.008 Kubota K, Sakikawa C, Katsumata M, Nakamura T, and Wakabayashi K. Platelet-derived growth factor BB secreted from osteoclasts acts as an osteoblastogenesis inhibitory factor. J Bone Miner Res, 2002, 17(2): 257–65., DOI 10.1359/jbmr.2002.17.2.257 Fu R, Gao S, Peng F, Li J, Liu H, Wang H, et al. Relationship between abnormal osteoblasts and cellular immunity in multiple myeloma. Cancer Cel Int, 2014, 14:62., DOI 10.1186/1475-2867-14-62 Wuelling M, Delling G, and Kaiser E. Differential gene expression in stromal cells of human giant cell tumor of bone. Virchows Arch, 2004, 445(6):467–76., DOI 10.1007/s00428-004-1113-2 Negishi-Koga T, Shinohara M, Komatsu N, Bito H, Kodama T, Friedel RH, et al. Suppression of bone formation by osteoclastic expression of semaphorin 4D. Nat Med, 2011, 17(11):1473–80., DOI 10.1038/nm.2489 WuellingM, Delling G, and Kaiser E. The origin of the neoplastic stromal cell in giant cell tumor of bone. Hum Pathol, 2003, 34(10):983–93. Brˇci´c I, Yamani F, Inwards CY, Sumathi V, Dodd L, Kreiger PA, et al. Giant cell tumor of bone with cartilage matrix: a clinicopathologic study of 17 Cases. Am J Surg Pathol, 2020, 44(6):748–56., DOI 10.1097/PAS.0000000000001446 Folpe AL, Morris RJ, and Weiss SW. Soft tissue giant cell tumor of low malignant potential: a proposal for the reclassification of malignant giant cell tumor of soft parts. Mod Pathol, 1999, 12(9):894–902. Calli AO, Tunakan M, Katilmis H, Kilciksiz S, and Ozturkcan S. Soft tissue giant cell tumor of low malignant potential of the neck: a case report and review of the literature. Tjpath, 2014, 30(1):73–7., DOI 10.5146/tjpath.2014. 01167 Gao H-Q, Yang YM, Zhuang Y, and Liu P. locally advanced undifferentiated carcinoma with osteoclast-like giant cells of the pancreas. Wjg, 2015, 21(2): 694–8., DOI 10.3748/wjg.v21.i2.694 Albores-Saavedra J, Grider DJ, Wu J, Henson DE, and Goodman ZD. Giant cell tumor of the extrahepatic biliary tree: a clinicopathologic study of 4 cases and comparison with anaplastic spindle and giant cell carcinoma with osteoclast-like giant cells. Am J Surg Pathol, 2006, 30(4):495–500., DOI 10. 1097/00000478-200604000-00010 Terada M, Gondo N, Sawaki M, Hattori M, Yoshimura A, Kotani H, et al. Case of giant cell tumor of the breast, clinically suspected as malignant breast tumor. Surg Case Rep, 2019, 5(1):77., DOI 10.1186/s40792-019-0635-4 Bui MM, Purohit CN, and Hakam A. Fine needle aspiration biopsy of an osteoclast-rich undifferentiated urothelial carcinoma: a cytology case report and review of the literature. Cytojournal, 2010, 7:18., DOI 10.4103/1742-6413. 70407 Fang X, Hicks DG, Hicks W, Jr, and Zhang S. Osteoclastlike giant cell tumor of the salivary gland. Ann Diagn Pathol, 2009, 13(2):114–8., DOI 10.1016/j. anndiagpath.2007.06.009 Derbel O, Zrounba P, Chassagne-Clement C, Decouvelaere AV, Orlandini F, Duplomb S, et al. An unusual giant cell tumor of the thyroid: case report and review of the literature. J Clin Endocrinol Metab, 2013, 98(1):1–6., DOI 10. 1210/jc.2011-2306 Singh R, Zaheer S, and Mandal A. Primary osteoclast-like giant cell tumor of parotid gland: a rare extraskeletal presentation with diagnostic challenges. J Oral Maxillofac Pathol, 2016, 20(2):331., DOI 10.4103/0973-029x.185903 Froboese N, Barth T, Brosch S, and Reiter R. Kasuistik: osteoklastischer riesenzelltumor der stimmlippe - eine sehr seltene Ursache fur Heiserkeit. Laryngo-Rhino-Otol, 2015, 94(4):246–7., DOI 10.1055/s-0034-1385945 Kanthan R, and Torkian B. Primary De Novo malignant giant cell tumor of kidney: a case report. BMC Urol, 2004, 4:1–7., DOI 10.1186/1471-2490-4-7 Priori SF, Schwartz LE, and Epstein JI. An expandet immunohistochemical profile os osteoclastic-rich undifferenriated carcinoma of the urinary bladder. Mod Pathol, 2018, 31(6):984–8., DOI 10.1038/s41379-018-0012-z Muraki T, Reid MD, Basturk O, Jang K-T, Bedolla G, Bagci P, et al. undifferentiated carcinoma with osteoclastic giant cells of the pancreas. Am J Surg Pathol, 2016, 40(9):1203–16., DOI 10.1097/pas.0000000000000689 Deeken-Draisey A, Yang G-Y, Gao J, and Alexiev BA. Anaplastic thyroid carcinoma: an epidemiologic, histologic, immunohistochemical, and molecular single-institution study. Hum Pathol, 2018, 82:140–8., DOI 10. 1016/j.humpath.2018.07.027 Luchini C, Pea A, Lionheart G, Mafficini A, Nottegar A, Veronese N, et al. Pancreatic undifferentiated carcinoma with osteoclast-like giant cells is genetically similar to, but clinically distinct from, conventional ductal adenocarcinoma. J Pathol, 2017, 243(2):148–54., DOI 10.1002/path.4941 Baydar D, Amin MB, and Epstein JI. Osteoclast-rich undifferentiated carcinomas of the urinary tract. Mod Pathol, 2006, 19(2):161–71., DOI 10. 1038/modpathol.3800521 Kawano H, Tanaka S, Ishii A, Cui D, Eguchi S, Hashimoto O, et al. Osteoclast-rich undifferentiated carcinoma of the urinary bladder: an immunohistochemical study. Pathol - Res Pract, 2011, 207(11):722–7., DOI 10.1016/j.prp.2011.09.003 Behzato˘glu K, Durak H, Canberk S, Aydin O, Huq GE, Oznur M, et al. Giant cell tumor-like lesion of the urinary bladder: a report of two cases and literature review; giant cell tumor or undifferentiated carcinoma?. Diagn Pathol, 2009, 4:48., DOI 10.1186/1746-1596-4-48 Behzatoglu K. Osteoclast-rich undifferentiated carcinoma of the urinary bladder: is it really an entity?. Adv Anat Pathol, 2010, 17(4):288, 2010 ., author reply 288–289), DOI 10.1097/PAP.0b013e3181e4e348 Kim HM, Kim H, and Park YN. Sarcomatoid cholangiocarcinoma with osteoclast-like giant cells associated with hepatolithiasis: a case report. Clin Mol Hepatol, 2015, 21(3):309–13., DOI 10.3350/cmh.2015.21.3.309 Oehler U, Jurs M, Kloppel G, and Helpap B. Osteoclast-like giant cell tumour of the pancreas presenting as a pseudocyst-like lesion. Virchows Archiv, 1997, 431(3):215–8., DOI 10.1007/s004280050091 McFarland M, Dina R, Fisher C, and McCluggage WG. Osteosarcoma as malignantmural nodule in ovarianmucinous neoplasms of intestinal Ttype. Int J Gynecol Pathol, 2015, 34(4):369–73., DOI 10.1097/pgp.0000000000000153 Nai GA, Amico E, Gimenez VR, and Guilmar M. Osteoclast-like giant cell tumor of the pancreas associated with mucus-secreting adenocarcinoma. Case report and discussion of the histogenesis. Pancreatology, 2005, 5(2-3): 279–84., DOI 10.1159/000085283 Manduch M, Dexter DF, Jalink DW, Vanner SJ, and Hurlbut DJ. Undifferentiated pancreatic carcinoma with osteoclast-like giant cells: report of a case with osteochondroid differentiation. Pathol - Res Pract, 2009, 205(5):353–9., DOI 10.1016/j.prp.2008.11.006 Bergmann F, Esposito I, Michalski CW, Herpel E, Friess H, and Schirmacher P. Early undifferentiated pancreatic carcinoma with osteoclastlike giant cells: direct evidence for ductal evolution. Am J Surg Pathol, 2007, 31(12):1919–25., DOI 10.1097/pas.0b013e318067bca8 Hunt JL, Tometsko M, LiVolsi VA, Swalsky P, Finkelstein SD, and Barnes EL. Molecular Evidence of Anaplastic Transformation in Coexisting Well- Differentiated and Anaplastic Carcinomas of the Thyroid. Am J Surg Pathol, 2003, 27(12):1559–64., DOI 10.1097/00000478-200312000-00009 Jung CW, Han KH, Seol H, Park S, Koh JS, Lee SS, et al. Expression of cancer stem cell markers and epithelial-mesenchymal transition-related factors in anaplastic thyroid carcinoma. Int J Clin Exp Pathol, 2015, 8(1):560–8. Gaumann A, Hansen T, Kohler HH, Kommoss F, Mann W, Maurer J, et al. The Expression of cathepsins in osteoclast-like giant cells of an anaplastic thyroid carcinoma with tracheal perforation. Pathol - Res Pract, 2001, 197(4): 257–62., DOI 10.1078/0344-0338-00044 Ahmed R, Din HU, Hashmi SN, and Muhammad I. Sarcoma-Like mural nodule in a borderline mucinous tumour of ovary. J Coll Physicians Surg Pak, 2016, 26(5):435. Greer JB, and Ferrone CR. Spectrum and classification of cystic neoplasms of the pancreas. Surg Oncol Clin North America, 2016, 25(2):339., DOI 10.1016/j. soc.2015.11.002 Joseph L, Augustine J, and Omana M. Multilocular cystadenoma of the liver with mural nodules. Indian J Pathol Microbiol, 1993, 36(3):308–10 . Bakker RFR, Stoot JHMB, Blok P, and Merkus JWS. Primary retroperitoneal mucinous cystadenoma with sarcoma-like mural nodule. Virchows Arch, 2007, 451(4):853., DOI 10.1007/s00428-007-0479-3 Demirel D, Gun I, Kucukodaci Z, Balta AZ, and Ramzy I. Primary retroperitoneal mucinous cystadenoma with a sarcoma-like mural nodule. Int J Gynecol Pathol, 2013, 32(1):15., DOI 10.1097/pgp.0b013e31825f7c41 Kurman RJ, and Shih I-M.Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer-Shifting the paradigm. Hum Pathol, 2011, 42(7): 918–31., DOI 10.1016/j.humpath.2011.03.003 Wang Y, Wu R-c., Shwartz LE, Haley L, Lin M-t., Shih I-m., et al. Clonality analysis of combined brenner and mucinous tumours of the ovary reveals their monoclonal origin. J Pathol, 2015, 237(2):146–51., DOI 10.1002/path.4572 Pawelek JM. Tumour-cell fusion as a source of myeloid traits in cancer. Lancet Oncol, 2005, 6(12):988–93., DOI 10.1016/s1470-2045(05)70466-6 Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC, et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol, 2001, 166(1): 678–89., DOI 10.4049/jimmunol.166.1.678 Elkabets M, Ribeiro VSG, Dinarello CA, Ostrand-Rosenberg S, Di Santo JP, Apte RN, et al. IL-1β regulates a novel myeloid-derived suppressor cell subset that impairs NK cell development and function. Eur J Immunol, 2010, 40(12):3347–57., DOI 10.1002/eji.201041037 Smith HA, and Kang Y. The metastasis-promoting roles of tumor-associated immune cells. J Mol Med, 2013, 91(4):411–29., DOI 10.1007/s00109-013- 1021-5 Williams ED, Gao D, Redfern A, and Thompson EW. Controversies around epithelial-mesenchymal plasticity in cancer metastasis. Nat Rev Cancer, 2019, 19(12):716–32., DOI 10.1038/s41568-019-0213-x Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature, 2007, 449(7162):557–63., DOI 10.1038/nature06188 Soundararajan R, Paranjape AN, Barsan V, Chang JT, and Mani SA. A novel embryonic plasticity gene signature that predicts metastatic competence and clinical outcome. Sci Rep, 2015, 5(5):11766., DOI 10.1038/srep11766 Chikaishi Y, Uramoto H, and Tanaka F. The EMT status in the primary tumor does not predict postoperative recurrence or disease-free survival in lung adenocarcinoma. Anticancer Res, 2011, 31(12):4451–6 . Ouzounova M, Lee E, Piranlioglu R, El Andaloussi A, Kolhe R, Demirci MF, et al. Monocytic and granulocytic myeloid derived suppressor cells differentially regulate spatiotemporal tumour plasticity during metastatic cascade. Nat Commun, 2017, 8:14979., DOI 10.1038/ncomms14979 Quail DF, and Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med, 2013, 19(11):1423–37., DOI 10.1038/ nm.3394 Oren-Suissa M, and Podbilewicz B. Cell fusion during development. Trends Cel Biol, 2007, 17(11):537–46., DOI 10.1016/j.tcb.2007.09.004 Duelli D, and Lazebnik Y. Cell-to-cell fusion as a link between Viruses and cancer. Nat Rev Cancer, 2007, 7(12):968–76., DOI 10.1038/nrc2272 Zhang L-N, Huang Y-H, and Zhao L. Fusion of macrophages promotes breast cancer cell proliferation, migration and invasion through activating epithelial-mesenchymal transition and Wnt/β-catenin signaling pathway. Arch Biochem Biophys, 2019, 676:108137., DOI 10. 1016/j.abb.2019.108137 Noubissi F, and Ogle B. Cancer cell fusion: mechanisms slowly unravel. Ijms, 2016, 17(9):1587., DOI 10.3390/ijms17091587 Andersen T, Boissy P, Sondergaard T, Kupisiewicz K, Plesner T, Rasmussen T, et al. Osteoclast nuclei of myeloma patients show chromosome translocations specific for the myeloma cell clone: a new type of cancerhost partnership?. J Pathol, 2007, 211(1):10–7., DOI 10.1002/path.2078 Ding J, Jin W, Chen C, Shao Z, and Wu J. Tumor associated macrophage × cancer cell hybrids may acquire cancer stem cell properties in breast cancer. PLoS One, 2012, 7(7):e41942., DOI 10.1371/journal.pone.0041942 Athanasou NA, Bansal M, and Forsyth R. “Giant cell tumor of bone. In: CDM Fletcher, JA Bridge, PCW Hogendoorn, and FMertens., editors. World Health Organization Classification of Tumours of Soft Tissue and Bone. 4th ed.. Lyon, France: IARC Press, 2013). p. 321–4. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 609472 EP 609484 DI 10.3389/pore.2021.609472 PG 13 ER PT J AU Bus, D Nagy, Gy Poka, R Vajda, Gy AF Bus, Dorottya Nagy, Gyongyi Poka, Robert Vajda, Gyorgy TI Clinical Impact of Preoperative Magnetic Resonance Imaging in the Evaluation of Myometrial Infiltration and Lymph-Node Metastases in Stage I Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE endometrial; MRI; Cancer; Radical hysterectomy; postoperative stage; preoperative stage ID endometrial; MRI; Cancer; Radical hysterectomy; postoperative stage; preoperative stage AB Purpose: In the developed world, endometrial cancer is one of the most common malignant gynecological cancer types. Due to the highly available diagnostic modalities and patient education, the early detection of the tumor leads to high overall survival. Methods: In this study we analyzed the reliability of preoperativeMRI findings in the staging of early stage endometrial cancer, as well as the clinical characteristics of patients underwent radical hysterectomy and the histopathologic evaluation of their tumor, with the retrospective data of radical hysterectomies performed in our hospital between 2010 and 2019. Results: The accuracy, sensitivity, specificity, negative- and positive predictive value of MRI regarding stage were 94.7, 63.3, 94.8, 83.8, and 83.8%, respectively. The accuracy, sensitivity, specificity, negative- and positive predictive value of MRI for the detection of the myometrial invasion were 69.8, 80.0, 60.8, 64.3, and 77.5%, respectively. The accuracy, sensitivity, specificity, negative- and positive predictive value of MRI for the detection of lymph node metastases were 78.1, 28.6, 82, 11.1, and 93.6%, respectively. Conclusions: Based on our results, MRI is the method of choice in terms of evaluating overall staging, as well as myometrial invasion, as its specificity and negative predictive value are relatively high. However, systematic lymphadenectomy showed improved cancer-related survival and recurrence-free survival. Our studies showed that the diagnosis of lymph node metastases is difficult with MRI modality since hyperplastic and metastatic nodes cannot easily differentiate, leading to a high percentage of false-positive results. Therefore, other imagingmodalitiesmay be used formore accurate evaluation. New findings of our study were that the role of the radiologist’s expertise in the evaluation of MR imaging plays an essential role in lowering false-negative and false-positive results. Therefore, findings evaluated by a radiologist with high-level expertise in gynecological imaging can complement the clinical findings and help substantially define the needed treatment. C1 [Bus, Dorottya] Zala County Saint Rafael Hospital, Department of Obstetrics and GynecologyZalaegerszeg, Hungary. [Nagy, Gyongyi] Zala Megyei Korhaz, RadiologiaZalaegerszeg, Hungary. [Poka, Robert] University Medical School of Debrecen, Department of Gynecology and ObstetricsDebrecen, Hungary. [Vajda, Gyorgy] University of Pecs, Faculty of Health SciencesPecs, Hungary. RP Vajda, Gy (reprint author), University of Pecs, Faculty of Health Sciences, Pecs, Hungary. EM drvagy11@gmail.com CR Ferlay J, Colombet M, Bray F. Cancer incidence in five continents, CI5plus: IARC CancerBase No. 9 [internet]. Lyon, France: International Agency for Research on Cancer, 2019). Nemzeti Rakregiszter [Hungarian National Cancer database] Available at: http://stat.nrr.hu/, Accessed July 21, 2020) American Cancer Society. Cancer facts and figures. Atlanta, GA: American Cancer Society, 2019). Rizzo S, Femia M, Buscari V, Franchi D, Garbi A, Zanagnolo V, et al. Endometrial cancer: an overview of novelties in treatment and related imaging keypoints for local staging. Cancer Imaging, 2018, 18(1):4., DOI 10. 1186/s40644-018-0180-6 KimDH, Seong SJ, KimMK, BaeHS, KimM-L,Yun BS, et al. Dilatation and curettage ismore accurate than endometrial aspiration biopsy in early-stage endometrial cancer patients treated with high dose oral progestin and levonorgestrel intrauterine system. J Gynecol Oncol, 2017, 28(1):e1., DOI 10.3802/jgo.2017.28.e1 M Otero-Garcia M, Mesa-Alvarez A, Nikolic O, Nikolic N, Blance-Lobato P, Busta-Nikolic M, et al. Role of MRI in staging and follow-up of endometrial and cervical cancer: pitfalls and mimickers. Insights Imaging, 2019, 10(1):19., DOI 10. 1186/s13244-019-0696-8 Creasman W. Revised FIGO staging for carcinoma of the endometrium. Int J Gynecol Obstet, 2009, 105(2):109., DOI 10.1016/j.ijgo.2009.02.010 Amin B, Edge S, Greene F, Byrd DR, Brookland RK, Washington MK, et al., editors. AJCC cancer staging manual. 8th ed. New York, NY: Springer, 2017). Plataniotis G, Castiglione MESMO guidelines working Group. Endometrial cancler: ESMO clinical practice guidelines for diagnosis, treatment and followup. Ann Oncol, 2010, 21(Suppl. 5):v41–v45., DOI 10.1093/annonc/mdq245 Colombo N, Creutzberg C, Amant F, Bosse T, Nout R, Seesa C, et al. ESMOESGO- ESTRO endometrial consensus conference working group. ESMOESGO- ESTRO consensus conference on endometrial cancer: diagnosis, treatment and follow-up. Ann Oncol, 2016, 27(1):16–41., DOI 10.1093/ annonc/mdv484 Yang T, Tian S, Li Y, Tian X, Wang W, Zhao J, et al. Magnetic Resonance Imaging, MRI, and three-dimensional transvaginal ultrasonography scanning for preoperative assessment of high risk in women with endometrial cancer. Med Sci Monit, 2019, 25:2024–31., DOI 10.12659/msm.915276 Haldorsen IS, Salvesen HB. Staging of endometrial carcinomas with MRI using traditional and novel MRI techniques. Clin Radiol, 2012, 67(1):2–12., DOI 10. 1016/j.crad.2011.02.018 Guo Y,Wang P, Wang P, GaoW, Li F, Yang X, et al. Myometrial invasion and overall staging of endometrial carcinoma: assessment using fusion of T2- weighted magnetic resonance imaging and diffusion-weighted magnetic resonance imaging. Onco Targets Ther, 2017, 10:5937-5943., DOI 10.2147/ OTT.S145763 Body N, Lavoue V, De Kerdaniel O, Foucher F, Henno S, Cauchois A, et al. Are preoperative histology and MRI useful for classification of endometrial cancer risk?. BMC Cancer, 2016, 16(1):498., DOI 10.1186/s12885-016-2554-0 Multinu F, Ducie JA, Zahl Eriksson A G, Schlappe BA, Cliby WA, Glaser GE, et al. Role of lymphadenectomy in endometrial cancer with nonbulky lymph node metastasis: comparison of comprehensive surgical staging and sentinel lymph node algorithm. Gynecol Oncol, 2019, 155(2):177–85., DOI 10.1016/j. ygyno.2019.09.011 Karatasli V, Cakir I, Sain H, Ayaz D, Saci M. Can preoperative magnetic resonance imaging replace intraoperative frozen sectioning in the evaluation of myometrial invasion for early-stage endometrial carcinoma? Ginekol Pol, 2019, 90(3):128–33., DOI 10.5603/GP.2019.0023 Lin G, Ng K-K, Chang C-J, Wang J-J, Ho K-C, Yen T-C, et al. Myometrial invasion in endometrial cancer: diagnostic accuracy of diffusion-weighted 3.0- T MR imaging-initial experience. Radiology, 2009, 250(3):784–92., DOI 10. 1148/radiol.2503080874 Benedetti Pacini P, Basile S, Maneschi F, Lissioi A-A, Signorelli M, Scambia G, et al. Systematic pelvic lymphadenectomy vs. no lymphadenectomy in earlystage endometrial carcinoma: a randomized clinical trial. J Natl Cancer Inst, 2008, 100(23):1707–16., DOI 10.1093/jnci/djn397 Creutzberg CL, van Putten WL, Koper PC, Lybeert ML, Jobsen JJ, Warlam- Rodenhuis CC, et al. Surgery and postoperative radiotherapy versus surgery alone for patients with stage-1 endometrial carcinoma: multicentre randomised trial. PORTEC study group. Post operative radiation therapy in endometrial CarcinomaPost operative radiation therapy in endometrial carcinoma. Lancet, 2003, 355(9213):1404–11., DOI 10.1016/s0140-6736(00, 02139-5 Kitchener H, Swart AM, Qian Q, Parmar M. Efficacy of systematic pelvic lymphadenectomy in endometrial cancer, MRC ASTEC trial): a randomised study. Lancet, 2009, 373(9658):125–36., DOI 10.1016/s0140- 6736(09)60678-4 Todo Y, Kato H, Kaneuchi M, Watari H, Takeda M, Sakuragi N. Survival effect of para-aortic lymphadenectomy in endometrial cancer, SEPAL study): a retrospective cohort analysis. The Lancet, 2010, 375:1165–72., DOI 10.1016/ s0140-6736(09)62002-x Goel G, Rajanbabu A, Sandhya CJ, Nair IR. A prospective observational study evaluating the accuracy of MRI in predicting the extent of disease in endometrial cancer. Indian J Surg Oncol, 2019, 10(1):220–4., DOI 10.1007/ s13193-018-0832-9 Tanase Y, Takahama J, Kawaguchi R, Kobayashi H. Analysis of risk factors for lymphatic metastasis in endometrial carcinoma and utility of threedimensional magnetic resonance imaging in Gynecology. World J Oncol, 2018, 9(3):74–9., DOI 10.14740/wjon1106w Huang Y-T, Huang Y-L, Ng K-K, Lin G. Current status of magnetic resonance imaging in patients with malignant uterine neoplasms: a review. Korean J Radiol, 2018, 20(1):18–33., DOI 10.3348/kjr.2018.0090 Shatat OMM, Fakhry S, Helal MH. The fusion of T2 weighted MRI and diffusion-weighted imaging in evaluating the depth of myometrial invasion in endometrial cancer. Erciyes Med J, 2019, 41(4):375–80., DOI 10.14744/etd.2019. 43815 Yildirim N, Saatli B, Kose S, Sancar C, Ulukus C, Koyuncuoglu M, et al. Predictability of myometrial, lower uterine segment and cervical invasion with 3D transvaginal ultrasonography and magnetic resonance imaging in endometrial cancer patients: a prospective cohort study. Med Ultrason, 2018, 20(3):348–54., DOI 10.11152/mu-1493 Gil RT, Cunha TM, Horta M, Alves I. The added value of diffusion-weighted imaging in the preoperative assessment of endometrial cancer. Radiol Bras, 2019, 52(4):229–36., DOI 10.1590/0100-3984.2018.0054 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 611088 EP 611095 DI 10.3389/pore.2021.611088 PG 8 ER PT J AU Franzese, O Di Francesco, A Meco, D Graziani, G Cusano, G Levati, L Riccardi, R Ruggiero, A AF Franzese, Ornella Di Francesco, M. Angela Meco, Daniela Graziani, Grazia Cusano, Gabriella Levati, Lauretta Riccardi, Riccardo Ruggiero, Antonio TI hTERT Transduction Extends the Lifespan of Primary Pediatric Low-Grade Glioma Cells While Preserving the Biological Response to NGF SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE low-grade glioma; senescence; hTERT; NGF (nerve growth factor); differentiation ID low-grade glioma; senescence; hTERT; NGF (nerve growth factor); differentiation AB The neurotrophin nerve growth factor (NGF) modulates the growth of human gliomas and is able to induce cell differentiation through the engagement of tropomyosin receptor kinase A (TrkA) receptor, although the role played in controlling glioma survival has proved controversial. Unfortunately, the slow growth rate of low-grade gliomas (LGG) has made it difficult to investigate NGF effects on these tumors in preclinical models. In fact, patientderived low-grade human astrocytoma cells duplicate only a limited number of times in culture before undergoing senescence. Nevertheless, replicative senescence can be counteracted by overexpression of hTERT, the catalytic subunit of telomerase, which potentially increases the proliferative potential of human cells without inducing cancerassociated changes. We have extended, by hTERT transduction, the proliferative in vitro potential of a human LGG cell line derived from a pediatric pilocytic astrocytoma (PA) surgical sample. Remarkably, the hTERT-transduced LGG cells showed a behavior similar to that of the parental line in terms of biological responses to NGF treatment, including molecular events associated with induction of NGF-related differentiation. Therefore, transduction of LGG cells with hTERT can provide a valid approach to increase the in vitro life-span of patient-derived astrocytoma primary cultures, characterized by a finite proliferative potential. C1 [Franzese, Ornella] University of Rome Tor Vergata, Department of Systems MedicineRome, Italy. [Di Francesco, M. Angela] Fondazione Policlinico A. Gemelli, IRCCS, Institute of Internal Medicine, Periodic Fever and Rare Diseases CenterRome, Italy. [Meco, Daniela] Fondazione Policlinico Universitario A. Gemelli, IRCCS, UOC di Oncologia PediatricaRome, Italy. [Graziani, Grazia] University of Rome Tor Vergata, Department of Systems MedicineRome, Italy. [Cusano, Gabriella] Fondazione Policlinico Universitario A. Gemelli, IRCCS, UOC di Oncologia PediatricaRome, Italy. [Levati, Lauretta] IDI-IRCCS, Molecular Oncology LaboratoryRome, Italy. [Riccardi, Riccardo] Fondazione Policlinico Universitario A. Gemelli, IRCCS, UOC di Oncologia PediatricaRome, Italy. [Ruggiero, Antonio] Fondazione Policlinico Universitario A. Gemelli, IRCCS, UOC di Oncologia PediatricaRome, Italy. RP Franzese, O (reprint author), University of Rome Tor Vergata, Department of Systems Medicine, Rome, Italy. EM franzese@uniroma2.it CR Qaddoumi I, Sultan I, Gajjar A. Outcome and prognostic features in pediatric gliomas. Cancer, 2009, 115(24):5761–5770., DOI 10.1002/cncr.24663 Rickert C, Paulus W. Epidemiology of central nervous system tumors in childhood and adolescence based on the new WHO classification. Childs Nerv Syst, 2001, 17(9):503–11., DOI 10.1007/s003810100496 Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol, 2007, 114(2):97–109., DOI 10.1007/s00401-007-0243-4 Listernick R, Charrow J, Gutmann DH. Intracranial gliomas in neurofibromatosis type 1. Am J Med Genet, 1999, 89:38–44., DOI 10.1002/, sici)1096-8628(19990326)89:1<38::aid-ajmg8>3.0.co;2-m Stokland T, Liu J-F, Ironside JW, Ellison DW, Taylor R, Robinson KJ, et al. A multivariate analysis of factors determining tumor progression in childhood low-grade glioma: a population-based cohort study, CCLG CNS9702). Neuro Oncol, 2010, 12(12):1257–68., DOI 10.1093/neuonc/noq092 Falsini B, Chiaretti A, Rizzo D, Piccardi M, Ruggiero A, Manni L, et al. Nerve growth factor improves visual loss in childhood optic gliomas: a randomized, double-blind, phase II clinical trial. Brain, 2016, 139(2):404–14., DOI 10.1093/ brain/awv366 Falsini B, Chiaretti A, Barone G, Piccardi M, Pierri F, Colosimo C, et al. Topical nerve growth factor as a visual rescue strategy in pediatric optic gliomas. Neurorehabil Neural Repair, 2011, 25(6):512–20., DOI 10.1177/ 1545968310397201 Chao MV. Nerve growth factor. In: MB Sporn AB. Roberts, editors. Peptide growth factors and their receptors II. New York, NY: Springer Publishing Co, 1991). 135–65. Frade JM, Barde Y-A. Nerve growth factor: two receptors, multiple functions. Bioessays, 1998, 20(2):137–45., DOI 10.1002/(SICI)1521-1878(199802)20: 2<137::AID-BIES6>3.0.CO;2-Q Chiaretti A, Falsini B, Aloe L, Pierri F, Fantacci C, Riccardi R. Neuroprotective role of nerve growth factor in hypoxicischemic injury. From brain to skin. Arch Ital Biol, 2011, 149(2):275–82., DOI 10.4449/aib. v149i2.1364 Liu TT, Wang H, Wang FJ, Xi YF, Chen LH. Expression of nerve growth factor and brain-derived neurotrophic factor in astrocytomas. Oncol Lett, 2018, 15(1):533–7., DOI 10.3892/ol.2017.7333 Meco D, Di Francesco AM, Melotti L, Ruggiero A, Riccardi R. Ectopic nerve growth factor prevents proliferation in glioma cells by senescence induction. J Cell Physiol, 2019, 234(5):6820–30., DOI 10.1002/jcp.27430 Segal RA, Greenberg ME. Intracellular signaling pathways activated by neuropathic factors. Annu Rev Neuro Sci, 1996, 19:463–89., DOI 10.1146/ annurev.ne.19.030196.002335 Kraemer BR, Snow JP, Vollbrecht P, Pathak A, Valentine WM, Deutch AY, et al. A role for the p75 neurotrophin receptor in axonal degeneration and apoptosis induced by oxidative stress. J Biol Chem, 2014, 289(31):21205–16., DOI 10.1074/jbc.M114.563403 Cragnolini AB, Huang Y, Gokina P, Friedman WJ. Nerve growth factor attenuates proliferation of astrocytes via the p75 neurotrophin receptor. Glia, 2009, 57(13):1386–92., DOI 10.1002/glia.20857 Aloe L, Alleva E, Bohm A, Levi-Montalcini R. Aggressive behavior induces release of nerve growth factor from mouse salivary gland into the bloodstream. Proc Natl Acad Sci U S A, 1986, 83(16):6184–7., DOI 10. 1073/pnas.83.16.6184 Kimura S, Yoshino A, Katayama Y, Watanabe T, Fukushima T. Growth control of C6 glioma in vivo by nerve growth factor. J Neurooncol, 2002, 59(3):199–205., DOI 10.1023/a:1019919019497 BrownMC, Staniszewska I, Lazarovici P, Tuszynski GP,Del Valle L,Marcinkiewicz C. Regulatory effect of nerve growth factor in α9β1 integrin-dependent progression of glioblastoma. Neuro Oncol, 2008, 10(6):968–80., DOI 10.1215/15228517-2008- 04710.1215/15228517-2008-0047 Tada K, Kochi M, Saya H, Kuratsu J-i., Shiraishi S, Kamiryo T, et al. Preliminary observations on genetic alterations in pilocytic astrocytomas associated with neurofibromatosis 1. Neuro Oncol, 2003, 5(4):228–34., DOI 10.1215/S115285170300005X Jacob K, Quang-Khuong D-A, Jones DTW, Witt H, Lambert S, Albrecht S, et al. Genetic aberrations leading to MAPK pathway activation mediate oncogene-induced senescence in sporadic pilocytic astrocytomas. Clin Cancer Res, 2011, 17(14):4650–60., DOI 10.1158/ 1078-0432.CCR-11-0127 Descamps S, Toillon R-A, Adriaenssens E, Pawlowski V, Cool SM, Nurcombe V, et al. Nerve growth factor stimulates proliferation and survival of human breast cancer cells through two distinct signaling pathways. J Biol Chem, 2001, 276(21):17864–17870., DOI 10.1074/jbc.M010499200 Stoczynska-Fidelus E, Och W, Rieske P, Bienkowski M, Banaszczyk M, Winiecka-Klimek M, et al. Spontaneous in vitro senescence of glioma cells confirmed by an antibody against IDH1R132H. Anticancer Res, 2014, 34(6): 2859–67. Courtois-Cox S, Genther Williams SM, Reczek EE, Johnson BW, McGillicuddy LT, Johannessen CM, et al. A negative feedback signaling network underlies oncogene-induced senescence. Cancer Cell, 2006, 10(6):459–72., DOI 10.1016/j. ccr.2006.10.003 Sarkisian CJ, Keister BA, Stairs DB, Boxer RB, Moody SE, Chodosh LA. Dosedependent oncogene-induced senescence in vivo and its evasion during mammary tumorigenesis. Nat Cell Biol, 2007, 9(5):493–505., DOI 10.1038/ ncb1567 Broniscer A, Baker SJ,West AN, FraserMM, Proko E, KocakM, et al. Clinical and molecular characteristics of malignant transformation of low-grade glioma in children. J Clin Oncol, 2007, 25(6):682–9., DOI 10.1200/JCO.2006.06.8213 Cheng Y, Pang JC-S, Ng H-K, Ding M, Zhang S-F, Zheng J, et al. Pilocytic astrocytomas do not show most of the genetic changes commonly seen in diffuse astrocytomas. Histopathology, 2000, 37(5):437–44., DOI 10.1046/j.1365- 2559.2000.01005.x Lin AW, Barradas M, Stone JC, van Aelst L, Serrano M, Lowe SW. Premature senescence involving p53 and p16 is activated in response to constitutiveMEK/MAPK mitogenic signaling. Genes Dev, 1998, 12(19):3008–19., DOI 10.1101/gad.12.19.3008 Harley CB. Telomerase is not an oncogene. Oncogene, 2002, 21(4):494–502., DOI 10.1038/sj.onc.1205076 Chong EY, Lam PY, Poon W-S, Ho-Kueng NG. Telomerase expression in gliomas including the nonastrocytic tumors. Hum Pathol, 1998, 29(6): 599–603., DOI 10.1016/s0046-8177(98)80009-9 Kotoula V, Cheva A, Barbanis S, Papadimitriou CS, Karkavelas G. hTERT immunopositivity patterns in the normal brain and in astrocytic tumors. Acta Neuropathol, 2006, 111:569–78., DOI 10.1007/ s00401-006-0036-1 Maes L, Van Neste L, Van Damme K, Kalala J, De Ridder L, Bekaert S, et al. Relation between telomerase activity, hTERT and telomere length for intracranial tumours. Oncol Rep, 2007, 18(6):1571–6., DOI 10.3892/or.18.6. 1571 Maes L, Kalala JPO, Cornelissen M, de Ridder L. Progression of astrocytomas and meningiomas: an evaluation in vitro. Cell Prolif, 2007, 40(1):14–23., DOI 10. 1111/j.1365-2184.2007.00415.x Sonoda Y, Ozawa T, Hirose Y, Aldape KD, McMahon M, Berger MS, et al. Formation of intracranial tumors by genetically modified human astrocytes defines four pathways critical in the development of human anaplastic astrocytoma. Cancer Res, 2001, 61(13):4956–60. Plunkett FJ, Franzese O, Belaramani LL, Fletcher JM, Gilmour KC, Sharifi R, et al. The impact of telomere erosion on memory CD8+ T cells in patients with X-linked lymphoproliferative syndrome. Mech Ageing Dev, 2005, 126(8): 855–65., DOI 10.1016/j.mad.2005.03.006 Forte G, Franzese O, Pagliari S, Pagliari F, Di Francesco AM, Cossa P, et al. Interfacing sca-1posMesenchymal stem cells with biocompatible scaffolds with different chemical composition and geometry. J Biomed Biotechnol, 2009, 2009:910610., DOI 10.1155/2009/910610 Mizuguchi H, Xu Z, Ishii-Watabe A, Uchida E, Hayakawa T. IRES-dependent second gene expression is significantly lower than cap-dependent first gene expression in a bicistronic vector. Mol Ther, 2000, 1(4):376–82., DOI 10.1006/ mthe.2000.0050 Berdun S, Rychter J, Vergara P. Effects of nerve growth factor antagonist K252a on peritoneal mast cell degranulation: implications for rat postoperative ileus. Am J Physiol Gastrointest Liver Physiol, 2015, 309(10):G801–G806., DOI 10. 1152/ajpgi.00152.2015 Ito K, Sanosaka T, Igarashi K, Ideta-OtsukaIdeta-Otsuka M, Aizawa A, Uosaki Y, et al. Identification of genes associated with the astrocyte-specific gene Gfap during astrocyte differentiation. Sci Rep, 2016, 6:23903., DOI 10.1038/srep23903 Bax DA, Little SE, Gaspar N, Perryman L, Marshall L, Viana-Pereira M, et al. Molecular and phenotypic characterisation of paediatric glioma cell lines as models for preclinical drug development. PLoS One, 2009, 4(4):e5209., DOI 10. 1371/journal.pone.0005209 Zhang X, ZhaoM, Huang A-y., Fei Z, ZhangW,Wang X-l.. The effect of cyclin D expression on cell proliferation in human gliomas. J Clin Neurosci, 2005, 12(2):166–8., DOI 10.1016/j.jocn.2004.03.036 Lavoie H, Therrien M. Regulation of RAF protein kinases in ERK signalling. Nat Rev Mol Cell Biol, 2015, 16(5):281–98., DOI 10.1038/nrm3979 Behling F, Schittenhelm J. Oncogenic BRAF alterations and their role in brain tumors. Cancers, Basel,, 2019, 11(6):794., DOI 10.3390/cancers11060794 Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature, 2002, 417(6892):949–954., DOI 10. 1038/nature00766 Rodriguez EF, Scheithauer BW, Giannini C, Rynearson A, Cen L, Hoesley B, et al. PI3K/AKT pathway alterations are associated with clinically aggressive and histologically anaplastic subsets of pilocytic astrocytoma. Acta Neuropathol, 2011, 121(3):407–420., DOI 10.1007/s00401-010-0784-9 Kim EJ, Battiste J, Nakagawa Y, Johnson JE. Ascl1, Mash1, lineage cells contribute to discrete cell populations in CNS architecture. Mol Cell Neurosci, 2008, 38(4):595–606., DOI 10.1016/j.mcn.2008.05.008 Somasundaram K, Reddy SP, Vinnakota K, Britto R, Subbarayan M, Nambiar S, et al. Upregulation of ASCL1 and inhibition of Notch signaling pathway characterize progressive astrocytoma. Oncogene, 2005, 24(47):7073–83., DOI 10.1038/sj.onc.1208865 Weng NP, Hodes RJ. The role of telomerase expression and telomere length maintenance in human and mouse. J Clin Immunol, 2000, 20(4):257–67., DOI 10.1023/a:1017223602293 Lee KM, Choi KH, Ouellette MM. Use of exogenous hTERT to immortalize primary human cells. Cytotechnology, 2004, 45(1-2):33–8., DOI 10.1007/10. 1007/s10616-004-5123-3 Tentori L, Vergati M, Muzi A, Levati L, Ruffini F, Forini O, et al. Generation of an immortalized human endothelial cell line as a model of neovascular proliferating endothelial cells to assess chemosensitivity to anticancer drugs. Int J Oncol, 2005, 27(2):525–35., DOI 10.3892/ijo.27.2.525 Davis T, Brook A, Rokicki MJ, Bagley MC, Kipling D. Evaluating the role of p38 MAPK in the accelerated cell senescence of werner syndrome fibroblasts. J Pharm, 2016, 9(2):23., DOI 10.3390/ph9020023 Lanna A, Coutavas E, Levati L, Seidel J, Rustin MH, Henson SM, et al. IFN-α inhibits telomerase in human CD8⁺ T cells by both hTERT downregulation and induction of p38 MAPK signalling. J Immunol, 2013, 191(7):3744–52., DOI 10.4049/jimmunol.1301409 Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 world health organization classification of tumors of the central nervous system: a summary. Acta Neuropathol, 2016, 131(6): 803–20., DOI 10.1007/s00401-016-1545-1 Comandini A, Naro C, Adamo R, Akbar AN, Lanna A, Bonmassar E, et al. Molecular mechanisms involved in HIV-1-tat mediated inhibition of telomerase activity in human CD4+ T lymphocytes. Mol Immunol, 2013, 54(2):181–92., DOI 10.1016/j.molimm.2012.12.003 Tacha DE, Tyrrell J, Lobo M. MASH-1 expression in high grade astrocytoma may demonstrate a potential model for prognosis that may lead to a molecular target for future therapy. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 April 16–20; New Orleans, LA [abstract], 2016). Franzese O, Barbaccia ML, Bonmassar E, Graziani G. Beneficial and detrimental effects of antiretroviral therapy on HIV-associated immunosenescence. Chemotherapy, 2018, 63(2):64–75., DOI 10.1159/ 000487534 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 612375 EP 612386 DI 10.3389/pore.2021.612375 PG 12 ER PT J AU Wang, H Yang, J AF Wang, Huai Yang, Jiankang TI Colorectal Cancer that Highly Express Both ACE2 and TMPRSS2, Suggesting Severe Symptoms to SARS-CoV-2 Infection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SARS-COV-2; cancer; ACE2; TMPRSS2; colorectum ID SARS-COV-2; cancer; ACE2; TMPRSS2; colorectum AB The epidemic of the novel, pathogenic SARS-coronavirus 2 (SARS-CoV-2) in the world pose a global health emergency. Cancer has been identified as a risk factor for the novel Coronavirus disease 2019 (COVID-19). The ACE2 and TMPRSS2 have been implicated in SARS-CoV-2 infection for mediating viral entry into the host cell. However, a systematic analysis of aberrant expression of ACE2 and TMPRSS2 was not yet reported in multiple human cancers. Here, we analyzed gene expression of ACE2 and TMPRSS2 across 31 types of tumors. Notably, overexpression of ACE2 and TMPRSS2 have been observed in colorectal cancer including colon adenocarcinoma (COAD), and rectum adenocarcinoma (READ). In addition, the colorectal tumors with upregulated gene expressing presented with decreased DNA methylation levels. DNA methylation might be one of the reasons for abnormal expression of ACE2 and TMPRSS2. Conclusively, colorectal cancer was the only cancer with the upregulated expression of ACE2 and TMPRSS2. More care of colorectal cancer patients is needed in multiple cancers affected by the COVID-19 outbreak. C1 [Wang, Huai] Dali University, School of Basic Medical Sciences, Department of Biochemistry and Molecular BiologyDali, China. [Yang, Jiankang] Dali University, School of Basic Medical Sciences, Department of Biochemistry and Molecular BiologyDali, China. RP Yang, J (reprint author), Dali University, School of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Dali, China. EM jkyang1984@126.com CR Phan T. Novel coronavirus: from discovery to clinical diagnostics. Infect Genet Evol, 2020, 79:104211., DOI 10.1016/j.meegid.2020.104211 Zhou P, Yang X-L, Wang X-G, Hu B, Zhang L, Zhang W, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature, 2020, 579:270–3., DOI 10.1038/s41586-020-2012-7 Hoffmann M, Kleine-Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S, et al. SARS-CoV-2 cell entry depends onACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell, 2020, 181:271–80., DOI 10.1016/j.cell.2020.02.052 Ziegler CGK,Allon SJ,Nyquist SK,Mbano IM,MiaoVN, Tzouanas CN, et al.HCA Lung Biological Network. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell, 2020, 181:1016–35., DOI 10.1016/j.cell.2020.04.035 LiangW, GuanW, Chen R, Wang W, Li J, Xu K, et al. Cancer patients in SARSCoV- 2 infection: a nationwide analysis in China. Lancet Oncol, 2020, 21:335–7., DOI 10.1016/s1470-2045(20)30096-6 Tang Z, Kang B, Li C, Chen T, Zhang Z. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res, 2019, 47:W556–W560., DOI 10.1093/nar/gkz430 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signaling, 2013, 6:pl1., DOI 10.1126/scisignal.2004088 Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce- Rodriguez I, Chakravarthi BVSK, et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia, 2017, 19:649–58., DOI 10.1016/j.neo.2017.05.002 Bakouny Z,Hawley JE, Choueiri TK, Peters S, Rini BI, Warner JL, et al. COVID-19 and cancer: current challenges and perspectives. Cancer Cel, 2020, 38(5):629–46. 1016/j.ccell.2020.09 10. Lamers MM, Beumer J, van der Vaart J, Knoops K, Puschhof J, Breugem TI, et al. SARS-CoV-2 productively infects human gut enterocytes. Science, 2020, 369:50–4., DOI 10.1126/science.abc1669 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 612969 EP 612972 DI 10.3389/pore.2021.612969 PG 4 ER PT J AU Weidemann, S Bohle, LJ Contreras, H Luebke, A Kluth, M Buscheck, F Hube-Magg, C Hoflmayer, D Moller, K Fraune, Ch Bernreuther, Ch Rink, M Simon, R Menz, A Hinsch, A Lebok, P Clauditz, T Sauter, G Uhlig, R Wilczak, W Steurer, S Burandt, E Krech, R Dum, D Krech, T Marx, A Minner, S AF Weidemann, Soren Bohle, Lukas Jan Contreras, Hendrina Luebke, M. Andreas Kluth, Martina Buscheck, Franziska Hube-Magg, Claudia Hoflmayer, Doris Moller, Katharina Fraune, Christoph Bernreuther, Christian Rink, Michael Simon, Ronald Menz, Anne Hinsch, Andrea Lebok, Patrick Clauditz, Till Sauter, Guido Uhlig, Ria Wilczak, Waldemar Steurer, Stefan Burandt, Eike Krech, Rainer Dum, David Krech, Till Marx, Andreas Minner, Sarah TI Napsin A Expression in Human Tumors and Normal Tissues SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE napsin A; immunohistochemistry; tissue micro array; diagnostic; human cancer types ID napsin A; immunohistochemistry; tissue micro array; diagnostic; human cancer types AB Background: Novel aspartic proteinase of the pepsin family A (Napsin A, TAO1/TAO2) is a functional aspartic proteinase which is involved in the maturation of prosurfactant protein B in type II pneumocytes and the lysosomal protein catabolism in renal cells. Napsin A is highly expressed in adenocarcinomas of the lung and is thus commonly used to affirm this diagnosis. However, studies have shown that other tumors can also express Napsin A. Methods: To comprehensively determine Napsin A expression in normal and tumor tissue, 11,957 samples from 115 different tumor types and subtypes as well as 500 samples of 76 different normal tissue types were evaluable by immunohistochemistry on tissue microarrays. Results: Napsin A expression was present in 16 different tumor types. Adenocarcinoma of the lung (85.6%), clear cell adenocarcinoma of the ovary (71.7%), clear cell adenocarcinoma of the endometrium (42.8%), papillary renal cell carcinoma (40.2%), clear cell (tubulo) papillary renal cell carcinoma (16.7%), endometrial serous carcinoma (9.3%), papillary thyroid carcinoma (9.3%) and clear cell renal cell carcinoma (8.2%) were among the tumors with the highest prevalence of Napsin A positivity. In papillary and clear cell renal cell carcinoma, reduced Napsin A expression was linked to adverse clinicpathological features (p ≤ 0.03). Conclusion: This methodical approach enabled us to identify a ranking order of tumors according to their relative prevalence of Napsin A expression. The data also show that loss of Napsin A is linked to tumor dedifferentiation in renal cell carcinomas. C1 [Weidemann, Soren] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Bohle, Lukas Jan] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Contreras, Hendrina] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Luebke, M. Andreas] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Kluth, Martina] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Buscheck, Franziska] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Hube-Magg, Claudia] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Hoflmayer, Doris] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Moller, Katharina] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Fraune, Christoph] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Bernreuther, Christian] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Rink, Michael] University Medical Center Hamburg-Eppendorf, Department of UrologyHamburg, Germany. [Simon, Ronald] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Menz, Anne] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Hinsch, Andrea] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Lebok, Patrick] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Clauditz, Till] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Sauter, Guido] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Uhlig, Ria] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Wilczak, Waldemar] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Steurer, Stefan] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Burandt, Eike] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Krech, Rainer] Clinical Center Osnabrueck, Institute of PathologyOsnabrueck, Germany. [Dum, David] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Krech, Till] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Marx, Andreas] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. [Minner, Sarah] University Medical Center Hamburg Eppendorf, PathologyHamburg, Germany. RP Minner, S (reprint author), University Medical Center Hamburg Eppendorf, Pathology, Hamburg, Germany. EM s.minner@uke.de CR Hirano T, Franzen B, Uryu K, Okuzawa K, Alaiya A, Vanky F, et al. Detection of polypeptides associated with the histopathological differentiation of primary lung carcinoma. Br J Cancer, 1995, 72(4):840–8., DOI 10.1038/bjc.1995.422 Tatnell PJ, Powell DJ, Hill J, Smith TS, Tew DG, and Kay J. Napsins: new human aspartic proteinases. FEBS Lett, 1998, 441(1):43–8., DOI 10.1016/s0014- 5793(98)01522-1 Ordonez NG. Napsin A expression in lung and kidney neoplasia. Adv Anat Pathol, 2012, 19(1):66–73., DOI 10.1097/PAP. 0b013e31823e472e Mori K, Ogawa Y, Tamura N, Ebihara K, Aoki T, Muro S, et al. Molecular cloning of a novel mouse aspartic protease-like protein that is expressed abundantly in the kidney. FEBS Lett, 1997, 401(2-3):218–22., DOI 10.1016/ s0014-5793(96)01473-1 Hirano T, Auer G, Maeda M, Hagiwara Y, Okada S, Ohira T, et al. Human tissue distribution of TA02, which is homologous with a new type of aspartic proteinase, napsin A. Jpn J Cancer Res, 2000, 91(10):1015–21., DOI 10.1111/j. 1349-7006.2000.tb00879.x Xu B, Abourbih S, Sircar K, Kassouf W, Aprikian A, Tanguay S, et al. Diagnostic and prognostic role of immunohistochemical expression of napsin-A aspartic peptidase in clear cell and papillary renal cell carcinoma. Appl Immunohistochem Mol Morphol, 2014, 22(3):206–12., DOI 10.1097/PAI. 0b013e31828ef24e Iwamoto M, Nakatani Y, Fugo K, Kishimoto T, and Kiyokawa T. Napsin A is frequently expressed in clear cell carcinoma of the ovary and endometrium. Hum Pathol, 2015, 46(7):957–62., DOI 10.1016/j.humpath. 2015.03.008 Brasch F, Ochs M, Kahne T, Guttentag S, Schauer-Vukasinovic V, Derrick M, et al. Involvement of napsin A in the C- and N-terminal processing of surfactant protein B in type-II pneumocytes of the human lung. J Biol Chem, 2003, 278(49):49006–14., DOI 10.1074/jbc.M306844200 Mori K, Shimizu H, Konno A, and Iwanaga T. Immunohistochemical localization of napsin and its potential role in protein catabolism in renal proximal tubules. Arch Histol Cytol, 2002, 65(4):359–68., DOI 10.1679/aohc. 65.359 Guan S, Wu Y, Zhang Q, and Zhou J. TGF-β1 induces CREB1-mediated miR- 1290 upregulation to antagonize lung fibrosis via Napsin A. Int J Mol Med, 2020, 46(1):141–8., DOI 10.3892/ijmm.2020.4565 Wu J, Zhang Y, Ding T, Cheng R, Gong W, Guo Y, et al. Napsin A expression in subtypes of thyroid tumors: comparison with lung adenocarcinomas. Endocr Pathol, 2020, 31(1):39–45., DOI 10.1007/s12022-019-09600-6 Surrey LF, Frank R, Zhang PJ, and Furth EE. TTF-1 and napsin-A are expressed in a subset of cholangiocarcinomas arising from the gallbladder and hepatic ducts. Am J Surg Pathol, 2014, 38(2):224–7., DOI 10.1097/PAS. 0000000000000138 Stoll LM, Johnson MW, Gabrielson E, Askin F, Clark DP, and Li QK. The utility of napsin-A in the identification of primary and metastatic lung adenocarcinoma among cytologically poorly differentiated carcinomas. Cancer Cytopathology, 2010, 118(6):441–9., DOI 10.1002/cncy.20108 Argani P, Hicks J, De Marzo AM, Albadine R, Illei PB, Ladanyi M, et al. Xp11 translocation renal cell carcinoma, RCC): extended immunohistochemical profile emphasizing novel RCC markers. Am J Surg Pathol, 2010, 34(9): 1295–303., DOI 10.1097/PAS.0b013e3181e8ce5b Velez MJ, Thomas CL, Stratton J, Bergfeld W, and Weaver J. The utility of using immunohistochemistry in the differentiation of metastatic, cutaneous clear cell renal cell carcinoma and clear cell hidradenoma. J Cutan Pathol, 2017, 44(7):612–5., DOI 10.1111/cup.12946 El-Maqsoud NMRA, Tawfiek ER, Abdelmeged A, Rahman MFA, and Moustafa AAE. The diagnostic utility of the triple markers Napsin A, TTF- 1, and PAX8 in differentiating between primary and metastatic lung carcinomas. Tumor Biol, 2016, 37(3):3123–34., DOI 10.1007/s13277-015- 3964-3 Zhu B, Rohan SM, and Lin X. Immunoexpression of napsin A in renal neoplasms. Diagn Pathol, 2015, 10:4., DOI 10.1186/s13000-015-0242-z Bishop JA, Sharma R, and Illei PB. Napsin A and thyroid transcription factor-1 expression in carcinomas of the lung, breast, pancreas, colon, kidney, thyroid, and malignant mesothelioma. Hum Pathol, 2010, 41(1):20–5., DOI 10.1016/j. humpath.2009.06.014 Kim M-Y, Go H, Koh J, Lee K, Min H-S, Kim M-A, et al. Napsin A is a useful marker for metastatic adenocarcinomas of pulmonary origin. Histopathology, 2014, 65(2):195–206., DOI 10.1111/his.12383 Ye J, Findeis-Hosey JJ, Yang Q, McMahon LA, Yao JL, Li F, et al. Combination of napsin A and TTF-1 immunohistochemistry helps in differentiating primary lung adenocarcinoma from metastatic carcinoma in the lung. Appl Immunohistochem Mol Morphol, 2011, 19(4):313–7., DOI 10.1097/PAI. 0b013e318205b059 Chevarie-Davis M, Riazalhosseini Y, Arseneault M, Aprikian A, Kassouf W, Tanguay S, et al. The morphologic and immunohistochemical spectrum of papillary renal cell carcinoma. Am J Surg Pathol, 2014, 38(7):887–94., DOI 10. 1097/PAS.0000000000000247 Turner BM, Cagle PT, Sainz IM, Fukuoka J, Shen SS, and Jagirdar J. Napsin A, a new marker for lung adenocarcinoma, is complementary and more sensitive and specific than thyroid transcription factor 1 in the differential diagnosis of primary pulmonary carcinoma: evaluation of 1674 cases by tissue microarray. Arch Pathol Lab Med, 2012, 136(2):163–71., DOI 10.5858/arpa.2011-0320-OA Chernock RD, El-Mofty SK, Becker N, and Lewis JS, Jr. Napsin A expression in anaplastic, poorly differentiated, and micropapillary pattern thyroid carcinomas. Am J Surg Pathol, 2013, 37(8):1215–22., DOI 10.1097/PAS. 0b013e318283b7b2 Kadivar M, and Boozari B. Applications and limitations of immunohistochemical expression of "Napsin-A" in distinguishing lung adenocarcinoma from adenocarcinomas of other organs. Appl Immunohistochem Mol Morphol, 2013, 21(3):191–5., DOI 10.1097/PAI. 0b013e3182612643 Leskela S, Romero I, Cristobal E, Perez-Mies B, Rosa-Rosa JM, Gutierrez- Pecharroman A, et al. The frequency and prognostic significance of the histologic type in early-stage ovarian carcinoma. Am J Surg Pathol, 2020, 44(2):149–61., DOI 10.1097/PAS.0000000000001365 Rao LKF, and Gilbertson JR. Longitudinal engagement of pathology residents. Am J Clin Pathol, 2014, 142(6):748–54., DOI 10.1309/AJCPWJ61RERWEDIK Kandalaft PL, Gown AM, and Isacson C. The lung-restricted marker napsin A is highly expressed in clear cell carcinomas of the ovary. Am J Clin Pathol, 2014, 142(6):830–6., DOI 10.1309/AJCP8WO2EOIAHSOF Li Q, Zeng X, Cheng X, Zhang J, Ji J, Wang J, et al. Diagnostic value of dual detection of hepatocyte nuclear factor 1 beta, HNF-1β, and napsin A for diagnosing ovarian clear cell carcinoma. Int J Clin Exp Pathol, 2015, 8(7): 8305–10. Rekhi B, Deodhar KK, Menon S,Maheshwari A, Bajpai J, Ghosh J, et al. Napsin A and WT 1 are useful immunohistochemical markers for differentiating clear cell carcinoma ovary from high-grade serous carcinoma. APMIS, 2018, 126(1):45–55., DOI 10.1111/apm.12784 Lim D, Ip PPC, Cheung ANY, Kiyokawa T, and Oliva E. Immunohistochemical comparison of ovarian and uterine endometrioid carcinoma, endometrioid carcinoma with clear cell change, and clear cell carcinoma. Am J Surg Pathol, 2015, 39(8):1061–9., DOI 10.1097/PAS. 0000000000000436 Fadare O, Zhao C, Khabele D, Parkash V, Quick CM, Gwin K, et al. Comparative analysis of Napsin A, alpha-methylacyl-coenzyme A racemase, AMACR, P504S), and hepatocyte nuclear factor 1 beta as diagnostic markers of ovarian clear cell carcinoma: an immunohistochemical study of 279 ovarian tumours. Pathology, 2015, 47(2):105–11., DOI 10.1097/PAT. 0000000000000223 Lee S, Piskorz AM, Le Page C, MesMasson A-M, Provencher D, Huntsman D, et al. Calibration and optimization of p53, WT1, and napsin A immunohistochemistry ancillary tests for histotyping of ovarian carcinoma. Int J Gynecol Pathol, 2016, 35(3):209–21., DOI 10.1097/PGP. 0000000000000251 RoyM, Jain D, Yadav R,Mathur SR, and Iyer VK. TTF-1 and napsin-A are not markers for biliary phenotype. Am J Surg Pathol, 2015, 39(12):1742–4., DOI 10. 1097/PAS.0000000000000551 Yamashita Y, Nagasaka T, Naiki-Ito A, Sato S, Suzuki S, Toyokuni S, et al. Napsin A is a specific marker for ovarian clear cell adenocarcinoma. Mod Pathol, 2015, 28(1):111–7., DOI 10.1038/modpathol.2014.61 Tennstedt P, Koster P, Bruchmann A, Mirlacher M, Haese A, Steuber T, et al. The impact of the number of cores on tissue microarray studies investigating prostate cancer biomarkers. Int J Oncol, 2012, 40(1):261–8., DOI 10.3892/ijo. 2011.1216 Buscheck F, Fraune C, Simon R, KluthM, Hube-Magg C, Moller-Koop C, et al. Aberrant expression of membranous carbonic anhydrase IX, CAIX, is associated with unfavorable disease course in papillary and clear cell renal cell carcinoma. Urol Oncol Semin Original Invest, 2018, 36(12):e19–531., DOI 10.1016/j.urolonc.2018.08.015 Eichenauer T, Bannenberg DC, Kluth M, Wittmer C, Buscheck F, Moller K, et al. 8p deletions in renal cell carcinoma are associated with unfavorable tumor features and poor overall survival. Urol Oncol Semin Original Invest, 2020, 38(2):e13–43., DOI 10.1016/j.urolonc.2019.09.024 Eichenauer T, Simmendinger L, Kluth M, Chirico V, Luebke AM, Hoflmayer D, et al. Chromosomal deletion of 9p21 is linked to poor patient prognosis in papillary and clear cell kidney cancer. Urol Oncol Semin Original Invest, 2020, 38(6):e1–605., DOI 10.1016/j.urolonc.2020.02.022 Eichenauer T, Simmendinger L, Fraune C, Mandelkow T, Blessin NC, Kluth M, et al. High level of EZH2 expression is linked to high density of CD8- positive T-lymphocytes and an aggressive phenotype in renal cell carcinoma. World J Urol, 2020, 39:481., DOI 10.1007/s00345-020-03200-4 Buscheck F, Fraune C, Simon R, Kluth M, Hube-Magg C, Moller-Koop C, et al. Prevalence and clinical significance of VHL mutations and 3p25 deletions in renal tumor subtypes. Oncotarget, 2020, 11(3):237–49., DOI 10.18632/ oncotarget.27428 Buscheck F, Fraune C, Kluth M, Lennartz M, Simon R, Hube-Magg C, et al. A non-diploid DNA status is linked to poor prognosis in renal cell cancer. World J Urol, 2020, 39:829., DOI 10.1007/s00345-020-03226-8 Eichenauer T, Shadanpour N, Kluth M, Gobel C, Weidemann S, Fraune C, et al. Chromosome 17p13 deletion is associated with an aggressive tumor phenotype in clear cell renal cell carcinoma. World J Surg Onc, 2020, 18(1): 128., DOI 10.1186/s12957-020-01902-y Zenali MJ, Weissferdt A, Solis LM, Ali S, Tang X, Mehran RJ, et al. An update on clinicopathological, immunohistochemical, and molecular profiles of colloid carcinoma of the lung. Hum Pathol, 2015, 46(6):836–42., DOI 10. 1016/j.humpath.2014.10.032 Dejmek A, Naucler P, Smedjeback A, Kato H, Maeda M, Yashima K, et al. Napsin A, TA02, is a useful alternative to thyroid transcription factor-1, TTF- 1, for the identification of pulmonary adenocarcinoma cells in pleural effusions. Diagn Cytopathol, 2007, 35(8):493–7., DOI 10.1002/dc.20667 Bian T, Zhao J, Feng J, Zhang Q, Qian L, Liu J, et al. Combination of cadherin- 17 and SATB homeobox 2 serves as potential optimal makers for the differential diagnosis of pulmonary enteric adenocarcinoma and metastatic colorectal adenocarcinoma. Oncotarget, 2017, 8(38):63442–52., DOI 10.18632/ oncotarget.18828 Mukhopadhyay S, and Katzenstein A-LA. Subclassification of non-small cell lung carcinomas lacking morphologic differentiation on biopsy specimens. Am J Surg Pathol, 2011, 35(1):15–25., DOI 10.1097/PAS.0b013e3182036d05 Kadota K, Nitadori J-i., Rekhtman N, Jones DR, Adusumilli PS, and Travis WD. Reevaluation and reclassification of resected lung carcinomas originally diagnosed as squamous cell carcinoma using immunohistochemical analysis. Am J Surg Pathol, 2015, 39(9):1170–80., DOI 10.1097/PAS.0000000000000439 Ezzat NE, Tahoun NS, and Ismail YM. The role of S100P and IMP3 in the cytologic diagnosis of pancreatic adenocarcinoma. J Egypt Natl Cancer Inst, 2016, 28(4):229–34., DOI 10.1016/j.jnci.2016.10.002 Aulakh KS, Chisholm CD, Smith DA, and Speights VO. TTF-1 and napsin A do not differentiate metastatic lung adenocarcinomas from primary esophageal adenocarcinomas: proposal of a novel staining panel. Arch Pathol Lab Med, 2013, 137(8):1094–8., DOI 10.5858/arpa.2012-0305-OA Ueno T, Linder S, and Elmberger G. Aspartic proteinase napsin is a useful marker for diagnosis of primary lung adenocarcinoma. Br J Cancer, 2003, 88(8):1229–33., DOI 10.1038/sj.bjc.6600879 Inamura K, Satoh Y, Okumura S, Nakagawa K, Tsuchiya E, FukayamaM, et al. Pulmonary adenocarcinomas with enteric differentiation. Am J Surg Pathol, 2005, 29(5):660–5., DOI 10.1097/01.pas.0000160438.00652.8b Hirano T, Gong Y, Yoshida K, Kato Y, Yashima K, Maeda M, et al. Usefulness of TA02, napsin A, to distinguish primary lung adenocarcinoma from metastatic lung adenocarcinoma. Lung Cancer, 2003, 41(2):155–62., DOI 10. 1016/s0169-5002(03)00194-6 Wu J, Chu PG, Jiang Z, and Lau SK. Napsin A expression in primary mucinproducing adenocarcinomas of the lung. Am J Clin Pathol, 2013, 139(2): 160–6., DOI 10.1309/AJCP62WJUAMSZCOM Rossi G, Cavazza A, Righi L, Sartori G, Bisagni A, Longo L, et al. Napsin-A, TTF-1, EGFR, and ALK status determination in lung primary and metastatic mucin-producing adenocarcinomas. Int J Surg Pathol, 2014, 22(5):401–7., DOI 10.1177/1066896914527609 Piljic Burazer M, Mladinov S, Capkun V, Kuret S, and Glavina Durdov M. The utility of thyroid transcription factor 1, TTF-1), napsin A, excision repair cross-complementing 1, ERCC1), anaplastic lymphoma kinase, alk, and the epidermal growth factor receptor, egfr, expression in small biopsy in prognosis of patients with lung adenocarcinoma - a retrograde single-center study from Croatia. Med Sci Monit, 2017, 23:489–97., DOI 10.12659/msm.899378 Ye J, Hameed O, Findeis-Hosey J, Fan L, Li F, McMahon L, et al. Diagnostic utility of PAX8, TTF-1 and napsin A for discriminating metastatic carcinoma from primary adenocarcinoma of the lung. Biotech Histochem, 2012, 87(1): 30–4., DOI 10.3109/10520295.2011.591838 Nishino M, Hoang MP, Della Pelle P, Morales-Oyarvide V, Huynh TG, Mark EJ, et al. Napsin A/p40 antibody cocktail for subtyping non-small cell lung carcinoma on cytology and small biopsy specimens. Cancer Cytopathology, 2016, 124(7):472–84., DOI 10.1002/cncy.21707 Cadioli A, Rossi G, Costantini M, Cavazza A, Migaldi M, and Colby TV. Lung cancer histologic and immunohistochemical heterogeneity in the era of molecular therapies. Am J Surg Pathol, 2014, 38(4):502–9., DOI 10.1097/ PAS.0000000000000154 Suzuki A, Shijubo N, Yamada G, Ichimiya S, Satoh M, Abe S, et al. Napsin A is useful to distinguish primary lung adenocarcinoma from adenocarcinomas of other organs. Pathol - Res Pract, 2005, 201(8-9):579–86., DOI 10.1016/j.prp. 2005.05.010 Yang X, Liu Y, Lian F, Guo L,Wen P, Liu XY, et al. Lepidic and micropapillary growth pattern and expression of Napsin A can stratify patients of stage I lung adenocarcinoma into different prognostic subgroup. Int J Clin Exp Pathol, 2014, 7(4):1459–68. Jin L, Liu Y, Wang X, and Qi X. Immunohistochemical analysis and comparison of napsin A, TTF1, SPA and CK7 expression in primary lung adenocarcinoma. Biotech Histochem, 2018, 93(5):364–72., DOI 10.1080/ 10520295.2018.1444790 Vidarsdottir H, Tran L, Nodin B, Jirstrom K, Planck M, Jonsson P, et al. Immunohistochemical profiles in primary lung cancers and epithelial pulmonary metastases. Hum Pathol, 2019, 84:221–30., DOI 10.1016/j. humpath.2018.10.009 Zhang P, Han Y-P, Huang L, Li Q, and Ma D-L. Value of napsin A and thyroid transcription factor-1 in the identification of primary lung adenocarcinoma. Oncol Lett, 2010, 1(5):899–903., DOI 10.3892/ol_00000160 Koh J, Go H, Kim M-Y, Jeon YK, Chung J-H, and Chung DH. A comprehensive immunohistochemistry algorithm for the histological subtyping of small biopsies obtained from non-small cell lung cancers. Histopathology, 2014, 65(6):868–78., DOI 10.1111/his.12507 Kriegsmann K, Cremer M, Zgorzelski C, Harms A, Muley T, Winter H, et al. Agreement of CK5/6, p40, and p63 immunoreactivity in non-small cell lung cancer. Pathology, 2019, 51(3):240–5., DOI 10.1016/j.pathol.2018.11.009 Bulutay P, Akyurek N, and Memis L. Clinicopathological and prognostic significance of the EML4-ALK translocation and IGFR1, TTF1, napsin A expression in patients with lung adenocarcinoma. Tjpath, 2020, 37(1):7–71., DOI 10.5146/tjpath.2020.01503 Kushitani K, Amatya VJ, Okada Y, Katayama Y, Mawas AS, Miyata Y, et al. Utility and pitfalls of immunohistochemistry in the differential diagnosis between epithelioid mesothelioma and poorly differentiated lung squamous cell carcinoma. Histopathology, 2017, 70(3):375–84., DOI 10.1111/his.13073 Ordonez NG. A word of caution regarding napsin A expression in squamous cell carcinomas of the lung. Am J Surg Pathol, 2012, 36(3):396–401., DOI 10. 1097/PAS.0b013e31823b13e2 Zhang Y, Zheng D, Li Y, Pan Y, Sun Y, and Chen H. Comprehensive investigation of clinicopathologic features, oncogenic driver mutations and immunohistochemical markers in peripheral lung squamous cell carcinoma. J Thorac Dis, 2017, 9(11):4434–40., DOI 10.21037/jtd.2017.10.43 Zannoni GF, Santoro A, Angelico G, Spadola S, Arciuolo D, Valente M, et al. Clear cell carcinoma of the endometrium: an immunohistochemical and molecular analysis of 45 cases. Hum Pathol, 2019, 92:10–7., DOI 10.1016/j. humpath.2019.06.005 Fadare O, Desouki MM, Gwin K, Hanley KZ, Jarboe EA, Liang SX, et al. Frequent expression of napsin A in clear cell carcinoma of the endometrium. Am J Surg Pathol, 2014, 38(2):189–96., DOI 10.1097/PAS.0000000000000085 Flanigan RC, Campbell SC, Clark JI, and Picken MM. Metastatic renal cell carcinoma. Curr Treat Options Oncol, 2003, 4(5):385–90., DOI 10.1007/s11864- 003-0039-2 Kubo T, Hirohashi Y, Fujita H, Sugita S, Kikuchi Y, Shinkawa T, et al. Occult ovarian clear-cell carcinoma diagnosed as primary adenocarcinoma of the lung: a case report of a diagnostic pitfall for clinicians and pathologists. Respir Med Case Rep, 2018, 25:306–8., DOI 10.1016/j.rmcr.2018.10.013 Jain D, Mallick SR, Singh V, Singh G, Mathur SR, and Sharma MC. Napsin A expression in anaplastic lymphoma kinase-positive diffuse large B-cell lymphoma: a diagnostic pitfall. Appl Immunohistochem Mol Morphol, 2016, 24(5):e34–e40., DOI 10.1097/PAI.0000000000000289 Ito T, Nakanishi K, and Goto H. A case of napsin A-positive metastatic lung cancer originating from the colon. Surg Case Rep, 2017, 3(1):114., DOI 10.1186/ s40792-017-0389-9 Torhorst J, Bucher C, Kononen J, Haas P, Zuber M, Kochli OR, et al. Tissue microarrays for rapid linking of molecular changes to clinical endpoints. Am J Pathol, 2001, 159(6):2249–56., DOI 10.1016/S0002- 9440(10)63075-1 Zhou L, Lv X, Yang J, Zhu Y,Wang Z, and Xu T. Napsin A is negatively associated with EMT-mediated EGFR-TKI resistance in lung cancer cells. Mol Med Rep, 2018, 18(2):1247–52., DOI 10.3892/mmr.2018. 9075 Zhou L, Lv X, Yang J, Zhu Y, Wang Z, and Xu T. Overexpression of Napsin A resensitizes drug-resistant lung cancer A549 cells to gefitinib by inhibiting EMT. Oncol Lett, 2018, 16(2):2533–8., DOI 10.3892/ol.2018. 8963 Lee JG, Kim S, and Shim HS. Napsin A is an independent prognostic factor in surgically resected adenocarcinoma of the lung. Lung Cancer, 2012, 77(1): 156–61., DOI 10.1016/j.lungcan.2012.02.013 Ma Y, Fan M, Dai L, Kang X, Liu Y, Sun Y, et al. The expression of TTF-1 and Napsin A in early-stage lung adenocarcinoma correlates with the results of surgical treatment. Tumor Biol, 2015, 36(10):8085–92., DOI 10.1007/s13277- 015-3478-z NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 613099 EP 613110 DI 10.3389/pore.2021.613099 PG 12 ER PT J AU Varga, G Toth, DA Szita, RV Csukly, Z Hardi, A Gaal-Weisinger, J Nagy, Zs Altai, E Rencsik, A Plander, M Szendrei, T Korad, K Radvanyi, G Rottek, J Deak, B Szaleczky, E Schneider, T Kohl, Z Kosztolanyi, Sz Alizadeh, H Lengyel, Zs Modok, Sz Borbenyi, Z Lovas, Sz Varoczy, L Illes, Rajnics, P Masszi, T Mikala, G AF Varga, Gergely Toth, David Andras Szita, Reka Virag Csukly, Zoltan Hardi, Apor Gaal-Weisinger, Julia Nagy, Zsolt Altai, Elvira Rencsik, Annamaria Plander, Mark Szendrei, Tamas Korad, Krisztina Radvanyi, Gaspar Rottek, Janos Deak, Beata Szaleczky, Erika Schneider, Tamas Kohl, Zoltan Kosztolanyi, Szabolcs Alizadeh, Hussain Lengyel, Zsuzsanna Modok, Szabolcs Borbenyi, Zita Lovas, Szilvia Varoczy, Laszlo Illes, Arpad Rajnics, Peter Masszi, Tamas Mikala, Gabor TI Beneficial Effect of Lenalidomide-Dexamethason Treatment in Relapsed/Refractory Multiple Myeloma Patients: Results of Real-Life Data From 11 Hungarian Centers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE myeloma; lenalidomide; relapsed; real life; treatment ID myeloma; lenalidomide; relapsed; real life; treatment AB In Hungary, the cost of lenalidomide-based therapy is covered only for relapsed multiple myeloma (MM) patients, therefore lenalidomide is typically used in the second-line either as part of a triplet with proteasome inhibitors or as a doublet. Lenalidomide-dexamethasone is a standard treatment approach for relapsed/refractory MM, and according to recent large randomized clinical trials (RCT, the standard arm of POLLUX, ASPIRE, TOURMALINE), the progression-free survival (PFS) is expected to be approximately 18 months. We surveyed ten Hungarian centers treating MM and collected data of 278 patients treated predominantly after 2016. The median age was 65 years, and patients were distributed roughly equally over the 3 international staging system groups, but patients with high risk cytogenetics were underrepresented. 15.8% of the patients reached complete response, 21.6% very good partial response, 40.6% partial response, 10.8% stable disease, and 2.5% progressed on treatment. The median PFS was unexpectedly long, 24 months, however only 9 months in those with high risk cytogenetics. We found interesting differences between centers regarding corticosteroid type (prednisolone, methylprednisolone or dexamethasone) and dosing, and also regarding the choice of anticoagulation, but the outcome of the various centers were not different. Although the higher equivalent steroid dose resulted in more complete responses, the median PFS of those having lower corticosteroid dose and methylprednisolone were not inferior compared to the ones with higher dose dexamethasone. On multivariate analysis high risk cytogenetics and the number of prior lines remained significant independent prognostic factors regarding PFS (p < 0.001 and p = 0.005). Our results show that in well-selected patients Lenalidomide-dexamethasone can be a very effective treatment with real-world results that may even outperform those reported in the recent RCTs. This real world information may be more valuable than outdated RCT data when treatment options are discussed with patients. C1 [Varga, Gergely] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Toth, David Andras] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Szita, Reka Virag] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Csukly, Zoltan] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Hardi, Apor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Gaal-Weisinger, Julia] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Nagy, Zsolt] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Altai, Elvira] Veszprem- Csolnoky Ferenc County Hospital, HematologyVeszprem, Hungary. [Rencsik, Annamaria] Veszprem- Csolnoky Ferenc County Hospital, HematologyVeszprem, Hungary. [Plander, Mark] Vas County Markusovszky HospitalSzombathely, Hungary. [Szendrei, Tamas] Vas County Markusovszky HospitalSzombathely, Hungary. [Korad, Krisztina] County Hospital of Borsod-Abauj-ZemplenMiskolc, Hungary. [Radvanyi, Gaspar] County Hospital of Borsod-Abauj-ZemplenMiskolc, Hungary. [Rottek, Janos] National Institute of OncologyBudapest, Hungary. [Deak, Beata] National Institute of OncologyBudapest, Hungary. [Szaleczky, Erika] National Institute of OncologyBudapest, Hungary. [Schneider, Tamas] National Institute of OncologyBudapest, Hungary. [Kohl, Zoltan] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Kosztolanyi, Szabolcs] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Alizadeh, Hussain] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Lengyel, Zsuzsanna] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Modok, Szabolcs] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Borbenyi, Zita] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Lovas, Szilvia] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Varoczy, Laszlo] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Rajnics, Peter] Kaposi Mor Teaching HospitalKaposvar, Hungary. [Masszi, Tamas] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Mikala, Gabor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. RP Varga, G (reprint author), Semmelweis University, Department of Internal Medicine and Haematology, Budapest, Hungary. EM vargager@gmail.com CR Dimopoulos M, Spencer A, Attal M, Prince HM, Harousseau J-L, Dmoszynska A, et al.Lenalidomide Plus Dexamethasone for Relapsed or Refractory Multiple Myeloma. N Engl J Med, 2007, 357:2123–32., DOI 10.1056/nejmoa070594 Dimopoulos MA, Chen C, Spencer A, Niesvizky R, Attal M, Stadtmauer EA, et al.Long-term Follow-Up on Overall Survival from the MM-009 and MM- 010 Phase III Trials of Lenalidomide Plus Dexamethasone in Patients with Relapsed or Refractory Multiple Myeloma. Leukemia, 2009, 23:2147–52., DOI 10.1038/leu.2009.147 Weber DM, Chen C, Niesvizky R, Wang M, Belch A, Stadtmauer EA, et al.Lenalidomide Plus Dexamethasone for Relapsed Multiple Myeloma in North America. N Engl J Med, 2007, 357:2133–42., DOI 10.1056/ nejmoa070596 Dimopoulos MA, Oriol A, Nahi H, San-Miguel J, Bahlis NJ, Usmani SZ, et al.Daratumumab, Lenalidomide, and Dexamethasone for Multiple Myeloma. N Engl J Med, 2016, 375:1319–31., DOI 10.1056/nejmoa1607751 Stewart AK, Rajkumar SV, Dimopoulos MA, Masszi T, ˇSpiˇcka I, Oriol A, et al.Carfilzomib, Lenalidomide, and Dexamethasone for Relapsed Multiple Myeloma. N Engl J Med, 2015, 372:142–52., DOI 10.1056/nejmoa1411321 Moreau P, Masszi T, Grzasko N, Bahlis NJ, Hansson M, Pour L, et al.Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma. N Engl J Med, 2016, 374:1621–34., DOI 10.1056/nejmoa1516282 Durie BGM, Harousseau JL, Harousseau J-L, Miguel JS, Blade J, Barlogie B, et al.International Uniform Response Criteria for Multiple Myeloma. Leukemia, 2006, 20:1467–73., DOI 10.1038/sj.leu.2404284 Kyle RA, and Rajkumar SVCriteria for Diagnosis, Staging, Risk Stratification and Response Assessment of Multiple Myeloma. Leukemia, 2008, 23:3–9., DOI 10.1038/leu.2008.291 Palumbo A, Avet-Loiseau H, Oliva S, Lokhorst HM, Goldschmidt H, Rosinol L, et al.Revised International Staging System for Multiple Myeloma: A Report from International Myeloma Working Group. Jco, 2015, 33:2863–9., DOI 10. 1200/jco.2015.61.2267 Knauf W, Aldaoud A, Losem C, Mittermueller J, Neise M, Niemeier B, et al.Lenalidomide Plus Dexamethasone for Patients with Relapsed or Refractory Multiple Myeloma: Final Results of a Non-interventional Study and Comparison with the Pivotal Phase 3 Clinical Trials. Leuk Res, 2018, 68: 90–7., DOI 10.1016/j.leukres.2018.03.008 Rajkumar SV, Jacobus S, Callander NS, Fonseca R, Vesole DH, Williams ME, et al.Lenalidomide Plus High-Dose Dexamethasone versus Lenalidomide Plus Low-Dose Dexamethasone as Initial Therapy for Newly Diagnosed Multiple Myeloma: an Open-Label Randomised Controlled Trial. Lancet Oncol, 2010, 11:29–37., DOI 10.1016/s1470-2045(09)70284-0 Larocca A, Salvini M, Gaidano G, Cascavilla N, Baldini L, Glietta M, et al.Sparing Steroids in Elderly Intermediate Fit Newly Diagnosed Multiple Myeloma Patients Treated with a Dose/schedule Adjusted RD-R vs. Continuous RD: Results of RV-MM-PI-0752 Phase III Randomized Study: PF586. Hema Sphere, 2019, 3(S1):244. . Abstract retrieved from EHA Library. 06/14/19; 266385., DOI 10.1097/01.HS9.0000560632.24271.d7 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 613264 EP 613269 DI 10.3389/pore.2021.613264 PG 6 ER PT J TI NOTCH Single Nucleotide Polymorphisms in the Predisposition of Breast and Colorectal Cancers in Saudi Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Notch; single nucleotide polymorphism; breast cancer; colorectal cancer; genetic screening marker ID Notch; single nucleotide polymorphism; breast cancer; colorectal cancer; genetic screening marker AB Breast cancer (BC) is a heterogeneous disease and is one of the most common malignancy affecting women worldwide while colorectal cancer (CRC) is estimated to be the third common cancer and second leading cause of cancer related death globally. Both BC and CRC involve multiple genetic and epigenetic alterations in genes belonging to various signaling pathways including NOTCH that has been implicated in the development of these cancers. We investigated four single nucleotide polymorphisms, each in genes encoding NOTCH1-4 receptors for their role in susceptibility to breast and colorectal cancers in Saudi population. In this case-control study, TaqMan genotypic analysis of rs3124591 in NOTCH1 and rs3820041 in NOTCH4 did not exhibit association with breast as well as colorectal cancers. However, a strong association of rs11249433 which is in close proximity to NOTCH2 was observed with breast cancer susceptibility especially with those having an early onset of the disease. Interestingly, the rs1043994 located in NOTCH3 showed gender preference and was found to be significantly associated with colorectal cancers in males. Validation of these findings in bigger populations of different ethnicities may prove beneficial in identifying rs11249433 and rs1043994 as genetic screening markers for early detection of breast and colorectal carcinomas, respectively. CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J Clin, 2021)., DOI 10. 3322/caac.21660 Albeshan SM, Mackey MG, Hossain SZ, Alfuraih AA, Brennan PC. Breast cancer epidemiology in Gulf Cooperation Council countries: a regional and international comparison. Clin Breast Cancer, 2018, 18(3):e381–e392., DOI 10. 1016/j.clbc.2017.07.006 Ferlay JE, Lam F, Colombet M, Mery L, Pineros M, et al. Global cancer observatory: cancer today. Lyon, France: International Agency for Research on Cancer, 2020). Available from: https://gco.iarc.fr/today, Accessed February 21, 2021). Lilyquist J, Ruddy KJ, Vachon CM, Couch FJ. Common genetic variation and breast cancer risk-past, present, and future. Cancer Epidemiol Biomarkers Prev, 2018, 27(4):380–94., DOI 10.1158/1055-9965.EPI-17-1144 Kruk J, Czerniak U. Physical activity and its relation to cancer risk: updating the evidence. Asian Pac J Cancer Prev, 2013, 14(7):3993–4003., DOI 10.7314/apjcp. 2013.14.7.3993 Tse G, Eslick GD. Cruciferous vegetables and risk of colorectal neoplasms: a systematic review and meta-analysis. Nutr Cancer, 2014, 66(1):128–39., DOI 10. 1080/01635581.2014.852686 Ruiz-Narvaez EA, Lunetta KL, Hong C-C, Haddad S, Yao S, Cheng T-YD, et al. Genetic variation in the insulin, insulin-like growth factor, growth hormone, and leptin pathways in relation to breast cancer in African-American women: the AMBER consortium. NPJ Breast Cancer, 2016, 2(1):16034., DOI 10.1038/ npjbcancer.2016.34 Fernandez-Lopez JC, Romero-Cordoba S, Rebollar-Vega R, Alfaro-Ruiz LA, Jimenez-Morales S, Beltran-Anaya F, et al. Population and breast cancer patients’ analysis reveals the diversity of genomic variation of the BRCA genes in the Mexican population. Hum Genomics, 2019, 13(1):3., DOI 10. 1186/s40246-018-0188-9 Zhang K, Civan J, Mukherjee S, Patel F, Yang H. Genetic variations in colorectal cancer risk and clinical outcome. World J Gastroenterol, 2014, 20(15):4167–77., DOI 10.3748/wjg.v20.i15.4167 Phipps AI, Passarelli MN, Chan AT, Harrison TA, Jeon J, Hutter CM, et al. Common genetic variation and survival after colorectal cancer diagnosis: a genome-wide analysis. Carcinogenesis, 2016, 37(1):87–95., DOI 10.1093/carcin/ bgv161 Savas S, Liu G. Studying genetic variations in cancer prognosis, and risk): a primer for clinicians. Oncologist, 2009, 14(7):657–66., DOI 10.1634/ theoncologist.2009-0042 Bray SJ. Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol, 2006, 7(9):678–89., DOI 10.1038/nrm2009 Parr C, Watkins G, JiangW. The possible correlation of Notch-1 and Notch-2 with clinical outcome and tumour clinicopathological parameters in human breast cancer. Int J Mol Med, 2004, 14(5):779–86., DOI 10.3892/ ijmm.14.5.779 Mikaelian I, Blades N, Churchill GA, Fancher K, Knowles BB, Eppig JT, et al. Proteotypic classification of spontaneous and transgenic mammary neoplasms. Breast Cancer Res, 2004, 6(6):R668–679., DOI 10.1186/bcr930 Zhang Y, Li B, Ji Z-Z, Zheng P-S. Notch1 regulates the growth of human colon cancers. Cancer, 2010, 116(22):5207–18., DOI 10.1002/cncr.25449 Vinson KE, George DC, Fender AW, Bertrand FE, Sigounas G. The Notch pathway in colorectal cancer. Int J Cancer, 2016, 138(8):1835–42., DOI 10.1002/ ijc.29800 Wu G, Chen Z, Li J, Ye F, Chen G, Fan Q, et al. NOTCH4 is a novel prognostic marker that correlates with colorectal cancer progression and prognosis. J Cancer, 2018, 9(13):2374–9., DOI 10.7150/jca.26359 Thomas G, Jacobs KB, Kraft P, Yeager M, Wacholder S, Cox DG, et al. A multistage genome-wide association study in breast cancer identifies two new risk alleles at 1p11.2 and 14q24.1, RAD51L1). Nat Genet, 2009, 41(5):579–84., DOI 10.1038/ng.353 Fu Y-P, Edvardsen H, Kaushiva A, Arhancet JP, Howe TM, Kohaar I, et al. NOTCH2 in breast cancer: association of SNP rs11249433 with gene expression in ER-positive breast tumors without TP53 mutations. Mol Cancer, 2010, 9:113., DOI 10.1186/1476-4598-9-113 Campa D, Kaaks R, Le Marchand L, Haiman CA, Travis RC, Berg CD, et al. Interactions between genetic variants and breast cancer risk factors in the breast and prostate cancer cohort consortium. JNCI J Nat Cancer Inst, 2011, 103(16):1252–63., DOI 10.1093/jnci/djr265 Wu S, Cai J,Wang H, Zhang H, Yang W. Association between 1p11-rs11249433 polymorphism and breast cancer susceptibility: evidence from 15 case-control studies. PLoS One, 2013, 8(8):e72526., DOI 10.1371/journal.pone.0072526 Cao YW, Wan GX, Zhao CX, Hu JM, Li L, Liang WH, et al. Notch1 single nucleotide polymorphismrs3124591 is associated with the risk of development of invasive ductal breast carcinoma in a Chinese population. Int J Clin Exp Pathol, 2014, 7(7):4286–94. Yu T, Han C, Zhu G, Liao X, Qin W, Yang C, et al. Prognostic value of Notch receptors in postsurgical patients with hepatitis B virus-related hepatocellular carcinoma. Cancer Med, 2017, 6(7):1587–600., DOI 10.1002/cam4.1077 Yagci E, Degirmenci I, Ozbayer C, Ak G, Saydam F, Metintas M. Common variants rs3815188 and rs1043994 on Notch3 gene confer susceptibility to lung cancer: a hospital-based case-control study. J Environ Pathol Toxicol Oncol, 2019, 38(1):61–8., DOI 10.1615/JEnvironPatholToxicolOncol.2018028403 Livak KJ. Allelic discrimination using fluorogenic probes and the 5’ nuclease assay. Genet Anal, 1999, 14(5-6):143–9., DOI 10.1016/s1050-3862(98)00019-9 Venkatesh V, Nataraj R, Thangaraj GS, Karthikeyan M, Gnanasekaran A, Kaginelli SB, et al. Targeting Notch signalling pathway of cancer stem cells. Stem Cel Investig., 2018, 5:5., DOI 10.21037/sci.2018.02.02 Zardawi SJ, Zardawi I, McNeil CM, Millar EKA, McLeod D, Morey AL, et al. High Notch1 protein expression is an early event in breast cancer development and is associated with the HER-2 molecular subtype. Histopathology, 2010, 56(3):286–96., DOI 10.1111/j.1365-2559.2009.03475.x Hanahan D, Weinberg RA. The hallmarks of cancer. Cell, 2000, 100(1):57–70., DOI 10.1016/s0092-8674(00)81683-9 Schmith VD, Campbell DA, Sehgal S, Anderson WH, Burns DK, Middleton LT, et al. Pharmacogenetics and disease genetics of complex diseases. Cell Mol Life Sci, CMLS,, 2003, 60(8):1636–46., DOI 10.1007/ s00018-003-2369-4 Eccles D, Tapper W. The influence of common polymorphisms on breast cancer. Cancer Treat Res, 2010, 155:15–32., DOI 10.1007/978-1-4419-6033-7_2 Talseth-Palmer BA, Scott RJ. Genetic variation and its role in malignancy. Int J Biomed Sci, 2011, 7(3):158–71. Schmidt H, Zeginigg M, Wiltgen M, Freudenberger P, Petrovic K, Cavalieri M, et al. Genetic variants of the NOTCH3 gene in the elderly and magnetic resonance imaging correlates of age-related cerebral small vessel disease. Brain, 2011, 134(Pt 11):3384–97., DOI 10.1093/brain/awr252 Shibata N, Ohnuma T, Higashi S, Higashi M, Usui C, Ohkubo T, et al. Genetic association between Notch4 polymorphisms and Alzheimer’s disease in the Japanese population. J Gerontol Ser A: Biol Sci Med Sci, 2007, 62(4):350–1., DOI 10.1093/gerona/62.4.350 Jiang Y, Shen H, Liu Xa., Dai J, Jin G, Qin Z, et al. Genetic variants at 1p11.2 and breast cancer risk: a two-stage study in Chinese women. PLoS One, 2011, 6(6):e21563., DOI 10.1371/journal.pone.0021563 Figueroa JD, Garcia-Closas M, Humphreys M, Platte R, Hopper JL, Southey MC, et al. Associations of common variants at 1p11.2 and 14q24.1, RAD51L1, with breast cancer risk and heterogeneity by tumor subtype: findings from the Breast Cancer Association Consortium. Hum Mol Genet, 2011, 20(23): 4693–706., DOI 10.1093/hmg/ddr368 Antoniou AC, Kartsonaki C, Sinilnikova OM, Soucy P, McGuffog L, Healey S, et al. Common alleles at 6q25.1 and 1p11.2 are associated with breast cancer risk for BRCA1 and BRCA2 mutation carriers. Hum Mol Genet, 2011, 20(16): 3304–21., DOI 10.1093/hmg/ddr226 Smith RE, Colangelo L, Wieand HS, Begovic M, Wolmark N. Randomized trial of adjuvant therapy in colon carcinoma: 10-year results of NSABP protocol C-01. J Nat Cancer Inst, 2004, 96(15):1128–32., DOI 10.1093/jnci/ djh220 Simon MS, Chlebowski RT, Wactawski-Wende J, Johnson KC, Muskovitz A, Kato I, et al. Estrogen plus progestin and colorectal cancer incidence and mortality. J Clin Oncol, 2012, 30(32):3983–90., DOI 10.1200/JCO.2012.42. 7732 Majek O, Gondos A, Jansen L, Emrich K, Holleczek B, Katalinic A, et al. Sex differences in colorectal cancer survival: population-based analysis of 164,996 colorectal cancer patients in Germany. PLoS One, 2013, 8(7):e68077., DOI 10. 1371/journal.pone.0068077 Lucca I, Klatte T, Fajkovic H, de Martino M, Shariat SF. Gender differences in incidence and outcomes of urothelial and kidney cancer. Nat Rev Urol, 2015, 12(10):585–92., DOI 10.1038/nrurol.2015.232 Yoshida Y, Murayama T, Sato Y, Suzuki Y, Saito H, Nomura Y. Gender differences in long-term survival after surgery for non-small cell lung cancer. Thorac Cardiovasc Surg, 2016, 64(6):507–14., DOI 10.1055/s-0035- 1558995 Press OA, Zhang W, Gordon MA, Yang D, Lurje G, Iqbal S, et al. Genderrelated survival differences associated with EGFR polymorphisms in metastatic colon cancer. Cancer Res, 2008, 68(8):3037–42., DOI 10.1158/0008-5472.CAN- 07-2718 Garufi C, Giacomini E, Torsello A, Sperduti I, Melucci E, Mottolese M, et al. Gender effects of single nucleotide polymorphisms and miRNAs targeting clock-genes in metastatic colorectal cancer patients, mCRC). Sci Rep, 2016, 6: 34006., DOI 10.1038/srep34006 Schwaag S, Evers S, Schirmacher A, Stogbauer F, Ringelstein E, Kuhlenbaumer G. Genetic variants of the NOTCH3 gene in migraine-A mutation analysis and association study. Cephalalgia, 2006, 26(2):158–61., DOI 10.1111/j.1468-2982. 2005.01007.x Li Y, Liu N, Chen H, Huang Y, Zhang W. Association of Notch3 singlenucleotide polymorphisms and lacunar infarctions in patients. Exp Ther Med, 2016, 11(1):28–32., DOI 10.3892/etm.2015.2898 Yang R, Hong H, Wang M, Ma Z. Correlation between single-nucleotide polymorphisms within miR-30a and related target genes and risk or prognosis of nephrotic syndrome. DNA Cell Biol, 2018, 37(3):233–43., DOI 10.1089/dna. 2017.4024 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 616204 EP 616214 DI 10.3389/pore.2021.616204 PG 11 ER PT J AU Nelhubel, Gy Cserepes, M Szabo, B Turk, D Karpati, A Kenessey, I Raso, E Barbai, T Hegedus, Z Laszlo, V Szokol, B Dobos, J Orfi, L Tovari, J AF Nelhubel, A. Gyorgyi Cserepes, Mihaly Szabo, Balazs Turk, Dora Karpati, Adel Kenessey, Istvan Raso, Erzsebet Barbai, Tamas Hegedus, Zita Laszlo, Viktoria Szokol, Balint Dobos, Judit Orfi, Laszlo Tovari, Jozsef TI EGFR Alterations Influence the Cetuximab Treatment Response and c-MET Tyrosine-Kinase Inhibitor Sensitivity in Experimental Head and Neck Squamous Cell Carcinomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR; c-Met; HNSCC; R521K; targeted tumor therapy ID EGFR; c-Met; HNSCC; R521K; targeted tumor therapy AB Background: Anti-EGFR antibody therapy is still one of the clinical choices in head and neck squamous cell carcinoma (HNSCC) patients, but the emergence of cetuximab resistance questioned its effectiveness and reduced its applicability. Although several possible reasons of resistance against the antibody treatment and alternative therapeutic proposals have been described (EGFR alterations, activation of other signaling pathways), there is no method to predict the effectiveness of anti-EGFR antibody treatments and to suggest novel therapeutics. Our study investigated the effect of EGFR R521K alteration on efficiency of cetuximab therapy of HNSCC cell lines and tried to find alternative therapeutic approaches against the resistant cells. Methods: After genetic characterization of HNSCC cells, we chose one wild type and one R521K+ cell line for in vitro proliferation and apoptosis tests, and in vivo animal models using different therapeutic agents. Results: Although the cetuximab treatment affected EGFR signalization in both cells, it did not alter in vitro cell proliferation or apoptosis. In vivo cetuximab therapy was also ineffective on R521K harboring tumor xenografts, while blocked the tumor growth of EGFR-wild type xenografts. Interestingly, the cetuximab-resistant R521K tumors were successfully treated with c-MET tyrosine kinase inhibitor SU11274. Conclusion: Our results suggest that HNSCC cell line expressing the R521K mutant form of EGFR does not respond well to cetuximab treatment in vitro or in vivo, but hopefully might be targeted by c-MET tyrosine kinase inhibitor treatment. C1 [Nelhubel, A. Gyorgyi] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Cserepes, Mihaly] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Szabo, Balazs] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Turk, Dora] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Karpati, Adel] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Kenessey, Istvan] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Raso, Erzsebet] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Barbai, Tamas] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Hegedus, Zita] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. [Laszlo, Viktoria] Medical University of Vienna, Department of Thoracic SurgeryVienna, Austria. [Szokol, Balint] Vichem Kft.Budapest, Hungary. [Dobos, Judit] Vichem Kft.Budapest, Hungary. [Orfi, Laszlo] Vichem Kft.Budapest, Hungary. [Tovari, Jozsef] National Institute of Oncology, Center of Diagnostic and Experimental TumorpathologyBudapest, Hungary. RP Tovari, J (reprint author), National Institute of Oncology, Center of Diagnostic and Experimental Tumorpathology, Budapest, Hungary. EM tozsi@oncol.hu CR Marur S, and Forastiere AA Head and Neck Cancer: Changing Epidemiology, Diagnosis, and Treatment. Mayo Clinic Proceedingsaprilis, 2008, 83(4): 489–501., DOI 10.4065/83.4.489 Dobrossy L Epidemiology of Head and Neck Cancer: Magnitude of the Problem. Cancer Metastasis Rev, 2005, 24(1):9–17., DOI 10.1007/s10555-005- 5044-4 Goerner M, Seiwert TY, and Sudhoff HMolecular Targeted Therapies in Head and Neck Cancer—an Update of Recent Developements -. Head Neck Oncol, 2010, 2:8., DOI 10.1186/1758-3284-2-8 Mehra R, Cohen RB, and Burtness BA The Role of Cetuximab for the Treatment of Squamous Cell Carcinoma of the Head and Neck. Clin Adv Hematol Oncoloktober, 2008, 6(10):742–50. Zimmermann M, Zouhair A, Azria D, and Ozsahin M The Epidermal Growth Factor Receptor, EGFR, in Head and Neck Cancer: its Role and Treatment Implications. Radiat Oncol, 2006, 1:11., DOI 10.1186/1748- 717x-1-11 Chung CH, Ely K, McGavran L, Varella-Garcia M, Parker J, Parker N, et al. Increased Epidermal Growth Factor Receptor Gene Copy Number Is Associated with Poor Prognosis in Head and Neck Squamous Cell Carcinomas. Jco, 2006, 24(25):4170–4176., DOI 10.1200/jco.2006.07.2587 Rogers SJ, Harrington KJ, Rhys-Evans P, O-Charoenrat P, and Eccles SA Biological Significance of C-erbB Family Oncogenes in Head and Neck Cancer. Cancer Metastasis Rev, 2005, 24(1):47–69., DOI 10.1007/s10555-005-5047-1 Ali MALS, Gunduz M, Nagatsuka H, Gunduz E, Cengiz B, Fukushima K, et al. Expression and Mutation Analysis of Epidermal Growth Factor Receptor in Head and Neck Squamous Cell Carcinoma. Cancer Sci, 2008, 99(8):1589–94., DOI 10.1111/j.1349-7006.2008.00861.x Temam S, Kawaguchi H, El-Naggar AK, Jelinek J, Tang H, Liu DD, et al. Epidermal Growth Factor Receptor Copy Number Alterations Correlate with Poor Clinical Outcome in Patients with Head and Neck Squamous Cancer. Jco, 2007, 25(16):2164–70., DOI 10.1200/jco.2006.06.6605 Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen RB, et al. Radiotherapy Plus Cetuximab for Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med, 2006, 354(6):567–78., DOI 10.1056/nejmoa053422 Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, et al. Platinum-based Chemotherapy Plus Cetuximab in Head and Neck Cancer. N Engl J Med, 2008, 359:1116., DOI 10.1056/NEJMoa0802656 Astsaturov I, Cohen R, and Harari P EGFR-targeting Monoclonal Antibodies in Head and Neck Cancer. Ccdt, 2006, 6(8):691–710.doi:10.2174/ 156800906779010191 Soulieres D, Senzer NN, Vokes EE, Hidalgo M, Agarwala SS, and Siu LL Multicenter Phase II Study of Erlotinib, an Oral Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor, in Patients with Recurrent or Metastatic Squamous Cell Cancer of the Head and Neck. Jco, 2004, 22(1):77–85.doi:10. 1200/jco.2004.06.075 Cohen EEW, Rosen F, Stadler WM, Recant W, Stenson K, Huo D, et al. Phase II Trial of ZD1839 in Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck. Jco, 2003, 21(10):1980–7., DOI 10. 1200/jco.2003.10.051 Cohen EEW, Kane MA, List MA, Brockstein BE, Mehrotra B, Huo D, et al. Phase II Trial of Gefitinib 250 Mg Daily in Patients with Recurrent And/or Metastatic Squamous Cell Carcinoma of the Head and Neck. Clin Cancer Res. 2005;11(23):8418–8424.doi:10.1158/1078-0432.ccr-05-1247 Wheeler DL, Iida M, Kruser TJ, Nechrebecki MM, Dunn EF, Armstrong EA, et al. Epidermal Growth Factor Receptor Cooperates with Src Family Kinases in Acquired Resistance to Cetuximab. Cancer Biol Ther, 2009, 8(8):696–703., DOI 10.4161/cbt.8.8.7903 Wheeler DL, Huang S, Kruser TJ, Nechrebecki MM, Armstrong EA, Benavente S, et al. Mechanisms of Acquired Resistance to Cetuximab: Role of HER, ErbB, Family Members. Oncogene, 2008, 27(28):3944–56., DOI 10.1038/onc.2008.19 Braig F, Kriegs M, Voigtlaender M, Habel B, Grob T, Biskup K, et al. Cetuximab Resistance in Head and Neck Cancer Is Mediated by EGFRK521 Polymorphism. Cancer Res, 2017, 77(5):1188–1199.doi:10.1158/0008- 5472.can-16-0754 Benavente S, Huang S, Armstrong EA, Chi A, Hsu K-T, Wheeler DL, et al. Establishment and Characterization of a Model of Acquired Resistance to Epidermal Growth Factor Receptor Targeting Agents in Human Cancer Cells. Clin Cancer Res, 2009, 15(5):1585–92., DOI 10.1158/1078-0432.ccr-08-2068 Boerner JL Role of Src Family Kinases in Acquired Resistance to EGFR Therapies in Cancer. Cancer Biol Ther, 2009, 8(8):704–6., DOI 10.4161/cbt.8.8.8183 Koppikar P, Choi S-H, Egloff AM, Cai Q, Suzuki S, FreilinoM, et al. Combined Inhibition of C-Src and Epidermal Growth Factor Receptor Abrogates Growth and Invasion of Head and Neck Squamous Cell Carcinoma. Clin Cancer Res, 2008, 14(13):4284–91., DOI 10.1158/1078-0432.ccr-07-5226 Cortesina G, Martone T, Galeazzi E, Olivero M, De Stefani A, Bussi M, et al. Staging of Head and Neck Squamous Cell Carcinoma Using theMET Oncogene Product as Marker of Tumor Cells in Lymph Node Metastases. Int J Cancer, 2000, 89(3):286–92., DOI 10.1002/1097-0215(20000520)89: 3<286::aid-ijc12>3.0.co;2-u Seiwert TY, Jagadeeswaran R, Faoro L, Janamanchi V, Nallasura V, El Dinali M, et al. The MET Receptor Tyrosine Kinase Is a Potential Novel Therapeutic Target for Head and Neck Squamous Cell Carcinoma. Cancer Res, 2009, 69(7): 3021–31., DOI 10.1158/0008-5472.can-08-2881 Sattler M, Pride YB, Ma P, Gramlich JL, Chu SC, Quinnan LA, et al. A Novel Small Molecule Met Inhibitor Induces Apoptosis in Cells Transformed by the Oncogenic TPR-MET Tyrosine Kinase. Cancer Res, 2003, 63(17):5462–9. Madoz-Gurpide J, Zazo S, Chamizo C, Casado V, Carames C, Gavin E, et al. Activation of MET Pathway Predicts Poor Outcome to Cetuximab in Patients with Recurrent or Metastatic Head and Neck Cancer. J Transl Med, 2015, 13: 282., DOI 10.1186/s12967-015-0633-7 Cohen RB Current Challenges and Clinical Investigations of Epidermal Growth Factor Receptor, EGFR)- and ErbB Family-Targeted Agents in the Treatment of Head and Neck Squamous Cell Carcinoma, HNSCC). Cancer Treat Rev, 2014, 40(4):567–77., DOI 10.1016/j.ctrv.2013.10.002 Zhao B, Wang L, Qiu H, Zhang M, Sun L, Peng P, et al. Mechanisms of Resistance to Anti-EGFR Therapy in Colorectal Cancer. Oncotarget, 2017, 8(3):3980–4000., DOI 10.18632/oncotarget.14012 Bou-Assaly W, and Mukherji S Cetuximab, Erbitux). AJNR Am J Neuroradiol, 2010, 31(4):626–7., DOI 10.3174/ajnr.a2054 Holubec L, Polivka J, Safanda M, Karas M, and Liska V The Role of Cetuximab in the Induction of Anticancer Immune Response in Colorectal Cancer Treatment. Ar, 2016, 36(9):4421–6., DOI 10.21873/anticanres.10985 Trotta AM, Ottaiano A, Romano C, Nasti G, Nappi A, De Divitiis C, et al. Prospective Evaluation of Cetuximab-Mediated Antibody-dependent Cell Cytotoxicity in Metastatic Colorectal Cancer Patients Predicts Treatment Efficacy. Cancer Immunol Res, 2016, 4(4):366–74., DOI 10.1158/2326-6066. cir-15-0184 Wang D, Qian G, Zhang H,Magliocca KR, Nannapaneni S, Amin ARMR, et al. HER3 Targeting Sensitizes HNSCC to Cetuximab by Reducing HER3 Activity and HER2/HER3 Dimerization: Evidence from Cell Line and Patient-Derived Xenograft Models. Clin Cancer Res, 2017, 23(3):677–86., DOI 10.1158/1078- 0432.ccr-16-0558 De Pauw I, Lardon F, Van den Bossche J, Baysal H, Fransen E, Deschoolmeester V, et al. Simultaneous Targeting of EGFR , HER 2, and HER 4 by Afatinib Overcomes Intrinsic and Acquired Cetuximab Resistance in Head and Neck Squamous Cell Carcinoma Cell Lines. Mol Oncol, 2018, 12(6): 830–54., DOI 10.1002/1878-0261.12197 Ather F, Hamidi H, Fejzo MS, Letrent S, Finn RS, Kabbinavar F, et al. Dacomitinib, an Irreversible Pan-ErbB Inhibitor Significantly Abrogates Growth in Head and Neck Cancer Models that Exhibit Low Response to Cetuximab. PLoS One, 2013, 8(2):e56112., DOI 10.1371/journal.pone. 0056112 Adkins D, Mehan P, Ley J, Siegel MJ, Siegel BA, Dehdashti F, et al. Pazopanib Plus Cetuximab in Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: an Open-Label, Phase 1b and Expansion Study. Lancet Oncol, 2018, 19(8):1082–93., DOI 10.1016/s1470-2045(18, 30350-4 Ouyang X, Barling A, Lesch A, Tyner JW, Choonoo G, Zheng C, et al. Induction of Anaplastic Lymphoma Kinase, ALK, as a Novel Mechanism of EGFR Inhibitor Resistance in Head and Neck Squamous Cell Carcinoma Patient-Derived Models. Cancer Biol Ther, 2018, 19(10):921–33., DOI 10.1080/ 15384047.2018.1451285 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 620256 EP 620268 DI 10.3389/pore.2021.620256 PG 13 ER PT J TI Estrogen Aggravates Tumor Growth in a Diffuse Gastric Cancer Xenograft Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE diffuse gastric cancer; SUN-16 cells; estrogen; ovariectomized mice; cancer xenograft model ID diffuse gastric cancer; SUN-16 cells; estrogen; ovariectomized mice; cancer xenograft model AB Gastric cancer has the fifth-highest incidence rate and is the third leading cause of cancerrelated deaths worldwide. The incidence of gastric cancer is higher in men than in women, but for the diffuse types of gastric cancer, the trend is opposite. Estrogen is considered the prime culprit behind these differences. Nevertheless, the action of estrogen in gastric cancers remains unclear. In this study, we investigated the effect of estrogen on diffusetype gastric cancer. Human female diffuse gastric cancer SNU-16 cells were transplanted into male and female mice to analyze the effect of endogenous estrogen on tumor growth. Furthermore, the effect of exogenous estrogen was evaluated in ovariectomized mice. Expressed genes were compared between female and male xenograft models using RNA sequencing analysis. Furthermore, human gene expression omnibus databases were utilized to examine the effect of our target genes on overall survival. SNU-16-derived tumor growth was faster in female mice than in male mice. In total RNA sequencing, interferon gamma receptor 2 (IFNGR2), IQ motif containing E (IQCE), transient receptor potential cation channel subfamily M member 4 (TRPM4), and structure-specific endonuclease subunit SLX4 (SLX4) were found. These genes could be associated with the tumor growth in female diffuse-type gastric cancer which was affected by endogenous estrogen. In an ovariectomized gastric cancer xenograft model, exogenous estrogen promoted tumor growth. Especially, our results indicated that estrogen induced G protein-coupled estrogen receptor expression in these mice. These results suggest that estrogen aggravates tumor progression in female diffuse gastric cancer. CR Rawla P, Barsouk A. Epidemiology of gastric cancer: global trends, risk factors and prevention. Prz Gastroenterol, 2019, 14(1):26–38., DOI 10.5114/pg.2018. 80001 Chia NY, Tan P.Molecular classification of gastric cancer. Ann Oncol, 2016, 27: 763–9., DOI 10.1093/annonc/mdw040 Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature, 2014, 513(7517):202–9., DOI 10.1038/nature13480 Lauren P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand, 1965, 64:31–49., DOI 10.1111/ apm.1965.64.1.31 Chandanos E, Rubio CA, Lindblad M, Jia C, Tsolakis AV, Warner M, et al. Endogenous estrogen exposure in relation to distribution of histological type and estrogen receptors in gastric adenocarcinoma. Gastric Cancer, 2008, 11: 168–74., DOI 10.1007/s10120-008-0475-6 Kim SM, Min BH, Lee J, An JY, Lee JH, Sohn TS, et al. Protective effects of female reproductive factors on lauren intestinal-type gastric adenocarcinoma. Yonsei Med J, 2018, 59(1):28–34., DOI 10.3349/ymj.2018.59.1.28 Onland-Moret NC, Kaaks R, van Noord PAH, Rinaldi S, Key T, Grobbee DE, et al. Urinary endogenous sex hormone levels and the risk of postmenopausal breast cancer. Br J Cancer, 2003, 88:1394–99., DOI 10. 1038/sj.bjc.6600890 Missmer SA, Eliassen AH, Barbieri RL, Hankinson SE. Endogenous estrogen, androgen, and progesterone concentrations and breast cancer risk among postmenopausal women. J Natl Cancer Inst, 2004, 96(24):1856–65., DOI 10. 1093/jnci/djh336 Tanaka K, Sakai H, Hashizume M, Hirohata T., 2000, Serum testosterone: estradiol ratio and the development of hepatocellular carcinoma among male cirrhotic patients. Cancer Res 60, 5106–10. Farinati F, De Maria N, Marafin C, Fagiuoli S, Della Libera G, Naccarato R. Hepatocellular carcinoma in alcoholic cirrhosis: is sex hormone imbalance a pathogenetic factor? Eur J Gastroenterol Hepatol, 1995, 7:145–50. Marquez-Garban DC, Chen HW, Goodglick L, Fishbein MC, Pietras RJ. Targeting aromatase and estrogen signaling in human non-small cell lung cancer. Ann N Y Acad Sci., 2009, 1155:194–205., DOI 10.1111/j.1749-6632. 2009.04116.x Brady CW. Liver disease in menopause. World J Gastroenterol., 2015, 21(25): 7613–20., DOI 10.3748/wjg.v21.i25.7613 Freedman ND, Chow WH, Gao YT, Shu XO, Ji BT, Yang G, et al. Menstrual and reproductive factors and gastric cancer risk in a large prospective study of women. Gut, 2007, 56(12):1671–77., DOI 10.1136/gut.2007.129411 Deli T, Orosz M, Jakab A. Hormone replacement therapy in cancer survivorsreview of the literature. Pathol Oncol Res., 2020, 26(1):63–78., DOI 10.1007/ s12253-018-00569-x Brusselaers N, Maret-Ouda J, Konings P, El-Serag HB, Lagergren J. Menopausal hormone therapy and the risk of esophageal and gastric cancer. Int J Cancer, 2017, 140(7):1693–99., DOI 10.1002/ijc.30588 Takano N, Iizuka N, Hazama S, Yoshino S, Tangoku A, Oka M. Expression of estrogen receptor-alpha and -beta mRNAs in human gastric cancer. Cancer Lett, 2002, 176(2):129–35., DOI 10.1016/s0304-3835(01)00739-x Kim HW, Kim JH, Lim BJ, Kim H, Kim H, Park JJ, et al. Sex disparity in gastric cancer: female sex is a poor prognostic factor for advanced gastric cancer. Ann Surg Oncol, 2016, 23(13):4344–51., DOI 10.1245/s10434-016-5448-0 Qin J, Liu M, Ding Q, Ji X, Hao Y,Wu X, et al. The direct effect of estrogen on cell viability and apoptosis in human gastric cancer cells. Mol Cell Biochem, 2014, 395(1-2):99–107., DOI 10.1007/s11010-014-2115-2 Zhu G, Huang Y, Wu C, Wei D, Shi Y. Activation of G-protein-coupled estrogen receptor inhibits the migration of human nonsmall cell lung cancer cells via IKK-β/NF-κB signals. DNA Cell Biol, 2016, 35(8):434–42., DOI 10. 1089/dna.2016.3235 Wei T, Chen W, Wen L, Zhang J, Zhang Q, Yang J, et al. G protein-coupled estrogen receptor deficiency accelerates liver tumorigenesis by enhancing inflammation and fibrosis. Cancer Lett, 2016, 382(2):195–202., DOI 10.1016/ j.canlet.2016.08.012 Chen ZJ, WeiW, Jiang GM, Liu H, Wei WD, Yang X, et al. Activation of GPER suppresses epithelial mesenchymal transition of triple negative breast cancer cells via NF-κB signals. Mol Oncol, 2016, 10(6):775–88., DOI 10.1016/j.molonc. 2016.01.002 Lee S, Lee SK, Jung J., 2021, Potentiating activities of chrysin in the therapeutic efficacy of 5-fluorouracil in gastric cancer cells. Oncol Lett 21(1):24., DOI 10. 3892/ol.2020.12285 Oh S, Choi K, Kim KM, Jung J. Sex-dependent effects of estrogen pellets in human liver cancer xenograft models. Toxicol Res, 2020, 36(2):109–14., DOI 10. 1007/s43188-019-00020-6 Ohta R, Takagi A, Ohmukai H, Marumo H, Ono A, Matsushima Y, et al. Ovariectomized mouse uterotrophic assay of 36 chemicals. J Toxicol Sci, 2012, 37(5):879–89., DOI 10.2131/jts.37.879 Ho-Pham LT, Nguyen ND, Nguyen TV. Quantification of the relative contribution of estrogen to bone mineral density in men and women. BMC Musculoskelet Disord, 2013, 14:366., DOI 10.1186/1471-2474-14-366 Brohawn DG, O’Brien LC, Bennett JP, Jr. RNAseq analyses identify tumor necrosis factor-mediated inflammation as a major abnormality in ALS spinal cord. PLoS One, 2016, 11(8):e0160520., DOI 10.1371/journal.pone. 0160520 Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B, Methodol,, 1995, 57: 289–300., DOI 10.1111/j.2517-6161.1995.tb02031.x Szasz AM, Lanczky A, Nagy A, Forster S, Hark K, Green JE, et al. Crossvalidation of survival associated biomarkers in gastric cancer using transcriptomic data of 1,065 patients. Oncotarget, 2016, 7:49322–33., DOI 10.18632/oncotarget.10337 Jiang X, Chen K, Fan K, Guo J. Prognostic significance of artemin in gastric cancer and its role in tumorigenesis. Transl Cancer Res TCR, 2020, 9:12–20., DOI 10.21037/tcr.2019.11.13 Lee HS, Park CK, Oh E, Erkin OC, Jung HS, Cho MH, et al. Low SP1 expression differentially affects intestinal-type compared with diffuse-type gastric adenocarcinoma. PLoS One, 2013, 8:e55522., DOI 10.1371/journal. pone.0055522 Wang X, Snoeyink J. Multiple structure alignment by optimal RMSD implies that the average structure is a consensus. Comput Syst Bioinformatics Conf, 2006, 79–87. Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol, 2005, 5(5):375–86., DOI 10.1038/nri1604 Zaidi MR. The interferon-gamma paradox in cancer. J Interferon Cytokine Res., 2019, 39(1):30–8., DOI 10.1089/jir.2018.0087 Hou L, El-Omar EM, Chen J, Grillo P, Rabkin CS, Baccarelli A, et al. Polymorphisms in Th1-type cell-mediated response genes and risk of gastric cancer. Carcinogenesis, 2007, 28(1):118–23., DOI 10.1093/carcin/bgl130 IQCE. The human protein ATLAS, version 20.1, 2021, Available from: https:// www.proteinatlas.org/ENSG00000106012-IQCE/pathology. Gao Y, Liao P. TRPM4 channel and cancer. Cancer Lett, 2019, 454:66–9., DOI 10.1016/j.canlet.2019.04.012 Kappel S, Stoklosa P, Hauert B, Ross-Kaschitza D, Borgstrom A, Baur R, et al. TRPM4 is highly expressed in human colorectal tumor buds and contributes to proliferation, cell cycle, and invasion of colorectal cancer cells. Mol Oncol, 2019, 13(11):2393–405., DOI 10.1002/1878-0261.12566 Wong KK, Hussain FA. TRPM4 is overexpressed in breast cancer associated with estrogen response and epithelial-mesenchymal transition gene sets. PLoS One, 2020, 15(6):e0233884., DOI 10.1371/ journal.pone.0233884 Sagredo AI, Sagredo EA, Cappelli C, Baez P, Andaur RE, Blanco C, et al. TRPM4 regulates Akt/GSK3-β activity and enhances β-catenin signaling and cell proliferation in prostate cancer cells. Mol Oncol, 2018, 12(2):151–65., DOI 10.1002/1878-0261.12100 Li XC, Cheng Y, Yang X, Zhou JY, Dong YY, Shen BQ, et al. Decreased expression of TRPM4 is associated with unfavorable prognosis and aggressive progression of endometrial carcinoma. Am J Transl Res., 2020, 12(7):3926–39. Landwehr R, Bogdanova NV, Antonenkova N, Meyer A, Bremer M, Park- Simon TW, et al. Mutation analysis of the SLX4/FANCP gene in hereditary breast cancer. Breast Cancer Res Treat, 2011, 130(3):1021–8., DOI 10.1007/ s10549-011-1681-1 Zhang S, Mu T, Dang S, Huang T, Duan J, Chen S, et al. Identification of SLX4 as the most frequently mutated gene in homologous recombination deficiency in Asian gastric cancer. J Clin Oncol, 2020, 38(15_Suppl.):4570., DOI 10.1200/ JCO.2020.38.15_suppl.4570 Jung J. Role of G protein-coupled estrogen receptor in cancer progression. Toxicol Res., 2019, 35(3):209–14., DOI 10.5487/TR.2019. 35.3.209 Wang X, Baloh RH, Milbrandt J, Garcia KC. Structure of artemin complexed with its receptor GFRalpha3: convergent recognition of glial cell line-derived neurotrophic factors. Structure, 2006, 14(6):1083–92., DOI 10.1016/j.str.2006. 05.010 Fielder GC, Yang TW, Razdan M, Li Y, Lu J, Perry JK, et al. The GDNF family: a role in cancer? Neoplasia, 2018, 20(1):99–117., DOI 10.1016/j.neo.2017.10.010 Takahashi M. The GDNF/RET signaling pathway and human diseases. Cytokine Growth Factor Rev, 2001, 12(4):361–73., DOI 10.1016/s1359- 6101(01)00012-0 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 622733 EP 622741 DI 10.3389/pore.2021.622733 PG 9 ER PT J AU Rossouw, S Bendou, H Blignaut, R Bell, L Rigby, J Christoffels, A AF Rossouw, C. Sophia Bendou, Hocine Blignaut, J. Renette Bell, Liam Rigby, Jonathan Christoffels, Alan TI Evaluation of Protein Purification Techniques and Effects of Storage Duration on LC-MS/MS Analysis of Archived FFPE Human CRC Tissues SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE formalin-fixed paraffin-embedded proteomics; acetone precipitation and formic acid resolubilization; detergent removal plates; SP3/HILIC-on-bead-digestion; mass spectrometry; LC-MS/MS ID formalin-fixed paraffin-embedded proteomics; acetone precipitation and formic acid resolubilization; detergent removal plates; SP3/HILIC-on-bead-digestion; mass spectrometry; LC-MS/MS AB To elucidate cancer pathogenesis and its mechanisms at the molecular level, the collecting and characterization of large individual patient tissue cohorts are required. Since most pathology institutes routinely preserve biopsy tissues by standardized methods of formalin fixation and paraffin embedment, these archived FFPE tissues are important collections of pathology material that include patient metadata, such as medical history and treatments. FFPE blocks can be stored under ambient conditions for decades, while retaining cellular morphology, due to modifications induced by formalin. However, the effect of long-term storage, at resource-limited institutions in developing countries, on extractable protein quantity/quality has not yet been investigated. In addition, the optimal sample preparation techniques required for accurate and reproducible results from label-free LC-MS/MS analysis across block ages remains unclear. This study investigated protein extraction efficiency of 1, 5, and 10-year old human colorectal carcinoma resection tissue and assessed three different gel-free protein purification methods for label-free LC-MS/MS analysis. A sample size of n = 17 patients per experimental group (with experiment power = 0.7 and α = 0.05, resulting in 70% confidence level) was selected. Data were evaluated in terms of protein concentration extracted, peptide/protein identifications, method reproducibility and efficiency, sample proteome integrity (due to storage time), as well as protein/peptide distribution according to biological processes, cellular components, and physicochemical properties. Data are available via ProteomeXchange with identifier PXD017198. The results indicate that the amount of protein extracted is significantly dependent on block age (p < 0.0001), with older blocks yielding less protein than newer blocks. Detergent removal plates were the most efficient and overall reproducible protein purification method with regard to number of peptide and protein identifications, followed by the MagReSyn® SP3/HILIC method (with on-bead enzymatic digestion), and lastly the acetone precipitation and formic acid resolubilization method. Overall, the results indicate that long-term storage of FFPE tissues (as measured by methionine oxidation) does not considerably interfere with retrospective proteomic analysis (p > 0.1). Block age mainly affects initial protein extraction yields and does not extensively impact on subsequent label-free LC-MS/MS analysis results. C1 [Rossouw, C. Sophia] University of the Western Cape, South African National Bioinformatics Institute, South African Medical Research Council Bioinformatics UnitBellville, South Africa. [Bendou, Hocine] University of the Western Cape, South African National Bioinformatics Institute, South African Medical Research Council Bioinformatics UnitBellville, South Africa. [Blignaut, J. Renette] University of the Western Cape, Department of Statistics and Population StudiesBellville, South Africa. [Bell, Liam] Centre for Proteomic and Genomic Research, ObservatoryCape Town, South Africa. [Rigby, Jonathan] National Health Laboratory Service, Tygerberg Hospital, Faculty of Health Sciences, University of Stellenbosch, Division of Anatomical Pathology, Department of PathologyCape Town, South Africa. [Christoffels, Alan] University of the Western Cape, South African National Bioinformatics Institute, South African Medical Research Council Bioinformatics UnitBellville, South Africa. RP Christoffels, A (reprint author), University of the Western Cape, South African National Bioinformatics Institute, South African Medical Research Council Bioinformatics Unit, Bellville, South Africa. EM alan@sanbi.ac.za CR Daniele L, D’Armento G, and Bussolati G. Preanalytical time interval, PATI, and fixation. In: G Stanta, editor. Guidelines for molecular analysis in archive tissues. Berlin: Springer-Verlag, 2011). p. 5–11. Avaritt NL, Shalin S, and Tackett AJ. Decoding the proteome in formalin-fixed paraffin-embedded, FFPE, tissues. J Proteomics Bioinform, 2014, 7:e25., DOI 10.4172/jpb.10000e25 Bronsert P, Weißer J, Biniossek ML, Kuehs M, Mayer B, Drendel V, et al. Impact of routinely employed procedures for tissue processing on the proteomic analysis of formalin-fixed paraffin-embedded tissue. Prot Clin Appl, 2014, 8:796–804., DOI 10.1002/prca.201300082 Gustafsson OJR, Arentz G, and Hoffmann P. Proteomic developments in the analysis of formalin-fixed tissue. Biochim Biophys Acta, Bba, - Proteins Proteomics, 2015, 1854:559–80., DOI 10.1016/j.bbapap.2014.10.003 Klockenbusch C, O’Hara JE, and Kast J. Advancing formaldehyde crosslinking towards quantitative proteomic applications. Anal Bioanal Chem, 2012, 404:1057–67., DOI 10.1007/s00216-012-6065-9 Magdeldin S, and Yamamoto T. Toward deciphering proteomes of formalinfixed paraffin-embedded, FFPE, tissues. Proteomics, 2012, 12:1045–58., DOI 10. 1002/pmic.201100550 Maes E, Valkenborg D, Mertens I, Broeckx V, Baggerman G, Sagaert X, et al. Proteomic analysis of formalin-fixed paraffin-embedded colorectal cancer tissue using tandem mass tag protein labeling. Mol Biosyst, 2013, 9: 2686–95., DOI 10.1039/c3mb70177h Tanca A, Abbondio M, Pisanu S, Pagnozzi D, Uzzau S, and Addis MF. Critical comparison of sample preparation strategies for shotgun proteomic analysis of formalin-fixed, paraffin-embedded samples: insights from liver tissue. Clin Proteomics, 2014, 11(28):1–11., DOI 10.1186/1559-0275-11-28 Shen K, Sun J, Cao X, Zhou D, and Li J. Comparison of different buffers for protein extraction from formalin-fixed and paraffin-embedded tissue specimens. PLoS ONE, 2015, 10(11):e0142650., DOI 10.1371/journal.pone. 0142650 Craven RA, Cairns DA, Zougman A, Harnden P, Selby PJ, and Banks RE. Proteomic analysis of formalin-fixed paraffin-embedded renal tissue samples by label-free MS: assessment of overall technical variability and the impact of block age. Proteomics Clin Appl, 2013, 7(3-4):273–82., DOI 10.1002/prca. 201200065 Fowler CB, O’Leary TJ, and Mason JT. Toward improving the proteomic analysis of formalin-fixed, paraffin-embedded tissue. Expert Rev Proteomics, 2013, 10(4):389–400., DOI 10.1586/14789450.2013.820531 Kessner D, Chambers M, Burke R, Agus D, and Mallick P. ProteoWizard: open source software for rapid proteomics tools development. Bioinformatics, 2008, 24:2534–6., DOI 10.1093/bioinformatics/btn323 Wolff C, Schott C, Porschewski P, Reischauer B, and Becker K-F. Successful protein extraction from over-fixed and long-term stored formalin-fixed tissues. PLoS ONE, 2011, 6(1):e16353., DOI 10.1371/journal.pone.0016353 Sprung RW, Brock JWC, Tanksley JP, LiM,Washington MK, Slebos RJC, et al. Equivalence of protein inventories obtained from formalin-fixed paraffinembedded and frozen tissue in multidimensional liquid chromatographytandem mass spectrometry shotgun proteomic analysis. Mol Cel Proteomics, 2009, 8., DOI 10.1074/mcp.m800518-mcp200 Tanca A, Pagnozzi D, Falchi G, Biosa G, Rocca S, Foddai G, et al. Impact of fixation time on GeLC-MS/MS proteomic profiling of formalin-fixed, paraffinembedded tissues. J Proteomics, 2011, 74:1015–21., DOI 10.1016/j.jprot.2011. 03.015 Piehowski PD, Petyuk VA, Sontag RL, Gritsenko MA, Weitz KK, Fillmore TL, et al. Residual tissue repositories as a resource for population-based cancer proteomic studies. Clin Proteom, 2018, 15:26., DOI 10.1186/s12014-018-9202-4 Wi´sniewski JR, Zougman A, Nagaraj N, and Mann M. Universal sample preparation method for proteome analysis. Nat Methods, 2009, 6(5):359–62., DOI 10.1038/nmeth.1322 Botelho D, Wall MJ, Vieira DB, Fitzsimmons S, Liu F, and Doucette A, 2010). Top-down and bottom-up proteomics of SDS-containing solutions following mass-based separation. J Proteome Res., 9, 2863–70., DOI 10.1021/ pr900949p Pellerin D, Gagnon H, Dube J, and Corbin F. Amicon-adapted enhanced FASP: an in-solution digestion-based alternative sample preparation method to FASP. F1000Res, 2015, 4:140–18., DOI 10.12688/f1000research.6529.1 Kachuk C, Stephen K, and Doucette A. Comparison of sodium dodecyl sulfate depletion techniques for proteome analysis by mass spectrometry. J Chromatogr A, 2015, 1418:158–66., DOI 10.1016/j.chroma.2015.09.042 Doucette AA, Vieira DB, Orton DJ, and Wall MJ. Resolubilization of precipitated intact membrane proteins with cold formic acid for analysis by mass spectrometry. J Proteome Res, 2014, 13(12):6001–12., DOI 10.1021/ pr500864a Hughes CS, Foehr S, Garfield DA, Furlong EE, Steinmetz LM, and Krijgsveld J. Ultrasensitive proteome analysis using paramagnetic bead technology. Mol Syst Biol, 2014, 10(10):1–10., DOI 10.15252/msb.20145625 Scicchitano MS, Dalmas DA, Boyce RW, Thomas HC, and Frazier KS. Protein extraction of formalin-fixed, paraffin-embedded tissue enables robust proteomic profiles by mass spectrometry. J Histochem Cytochem, 2009, 57(9):849–60., DOI 10.1369/jhc.2009.953497 Wi´sniewski JR. Proteomic sample preparation from formalin fixed and paraffin embedded tissue. J Vis Exp, 2013, 79:e50589., DOI 10.3791/50589 Hughes CS, Sorensen PH, and Morin GB. A standardized and reproducible proteomics protocol for bottom-up quantitative analysis of protein samples using SP3 and mass spectrometry. In: V Brun and Y Coute´, editors. Proteomics for biomarker discovery: methods and protocols, methods in molecular biology. New York: Springer Nature, 2019). p. 65–87., DOI 10. 1007/978-1-4939-9164-8_5 Craig R, and Beavis RC. TANDEM: matching proteins with tandem mass spectra. Bioinformatics, 2004, 20(9):1466–7., DOI 10.1093/bioinformatics/ bth092 Dorfer V, Pichler P, Stranzl T, Stadlmann J, Taus T, Winkler S, et al. MS Amanda, a universal identification algorithm optimized for high accuracy tandem mass spectra. J Proteome Res, 2014, 13:3679–84., DOI 10.1021/ pr500202e Kim S, and Pevzner PA.MS-GF+ makes progress towards a universal database search tool for proteomics. Nat Commun, 2014, 5:5277., DOI 10.1038/ ncomms6277 Vaudel M, Barsnes H, Berven FS, Sickmann A, and Martens L. SearchGUI: an open-source graphical user interface for simultaneous OMSSA and X!Tandem searches. Proteomics, 2011, 11:996–9., DOI 10.1002/pmic.201000595 Elias JE, and Gygi SP. Target-Decoy search strategy for mass spectrometrybased proteomics. Methods Mol Biol, 2010, 604:55–71., DOI 10.1007/978-1- 60761-444-9_5 Apweiler R, Bairoch A, Wu CH, Barker WC, Boeckmann B, Ferro S, et al. UniProt: the universal protein knowledgebase. Nucleic Acids Res, 2004, 32: 115D–119., DOI 10.1093/nar/gkh131 Vaudel M, Burkhart JM, Zahedi RP, Oveland E, Berven FS, Sickmann A, et al. PeptideShaker enables reanalysis of MS-derived proteomics data sets. Nat Biotechnol, 2015, 33(1):22–4., DOI 10.1038/nbt.3109 Vaudel M, Breiter D, Beck F, Rahnenfuhrer J, Martens L, and Zahedi RP. D-score: a search engine independent MD-score. Proteomics, 2013, 13(6): 1036–41., DOI 10.1002/pmic.201200408 Taus T, Kocher T, Pichler P, Paschke C, Schmidt A, Henrich C, et al. Universal and confident phosphorylation site localization using phosphoRS. J Proteome Res, 2011, 10(12):5354–62., DOI 10.1021/pr200611n Barsnes H, Vaudel M, Colaert N, Helsens K, Sickmann A, Berven FS, et al. Compomics-utilities: an open-source Java library for computational proteomics. BMC Bioinformatics, 2011, 12:70., DOI 10.1186/1471-2105-12-70 Farias SE, Kline KG, Klepacki J, and Wu CC. Quantitative improvements in peptide recovery at elevated chromatographic temperatures from microcapillary liquid Chromatography−Mass spectrometry analyses of brain using selected reaction monitoring. Anal Chem, 2010, 82(9):3435–40., DOI 10.1021/ac100359p Powell DW, Weaver CM, Jennings JL, McAfee KJ, He Y,Weil PA, et al. Cluster analysis of mass spectrometry data reveals a novel component of SAGA. Mcb, 2004, 24:7249–59., DOI 10.1128/mcb.24.16.7249-7259.2004 Vaudel M. MS2-based quantitation. In: C Bessant, editor. New developments in mass spectrometry no. 5: proteome informatics. Cambridge: The Royal Society of Chemistry, 2017). p. 155–77., DOI 10.1039/9781782626732-FP001 Wu S, and Zhu Y. ProPAS: standalone software to analyze protein properties. Bioinformation, 2012, 8(3):167–9., DOI 10.6026/97320630008167 Oliveros JC. Venny. An interactive tool for comparing lists with Venn’s diagrams, 2007). http://bioinfogp.cnb.csic.es/tools/venny/index.html, Accessed May 11, 2020). Rossouw SC, Bendou H, Blignaut RJ, Bell L, Rigby J, and Christoffels A. Evaluation of protein purification techniques and effects of storage duration on LC-MS/MS analysis of archived FFPE human CRC tissues. In: PRIDE database. PXD017198, 2020). https://www.ebi.ac.uk/pride/archive/, Accessed January 20, 2020). Deutsch EW, Csordas A, Sun Z, Jarnuczak A, Perez-Riverol Y, Ternent T, et al. The ProteomeXchange Consortium in 2017: supporting the cultural change in proteomics public data deposition. Nucleic Acids Res, 2017, 45(D1): D1100–D1106., DOI 10.1093/nar/gkw936 Perez-Riverol Y, Csordas A, Bai J, Bernal-Llinares M, Hewapathirana S, Kundu DJ, et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res, 2019, 47(D1):D442–D450., DOI 10.1093/nar/gky1106 Wi´sniewski JR, Du´s K, and Mann M. Proteomic workflow for analysis of archival formalin-fixed and paraffin-embedded clinical samples to a depth of 10 000 proteins. Proteomics Clin Appl, 2012, 7:1–9., DOI 10.1002/prca. 201200046 Baiwir T, Valladares-Ayerbes M, Haz-Conde M, Blanco M, Aparicio G, Fernandez-Puente P, et al. A novel procedure for protein extraction from formalin-fixed paraffin-embedded tissues. Proteomics, 2011, 11:2555–9., DOI 10.1002/pmic.201000809 Agilent Technologies. FFPE protein extraction solution protocol, 2009). Retrieved from https://www.chem-agilent.com/pdf/strata/400925.pdf, Accessed October 12, 2019). Lee S, Bang S, Song K, and Lee I. Differential expression in normal-adenomacarcinoma sequence suggests complex molecular carcinogenesis in colon. Oncol Rep, 2006, 16(4):747–54., DOI 10.3892/or.16.4.747 Andersen CL, Christensen LL, Thorsen K, Schepeler T, Sorensen FB, Verspaget HW, et al. Dysregulation of the transcription factors SOX4, CBFB and SMARCC1 correlates with outcome of colorectal cancer. Br J Cancer, 2009, 100(3):511–23., DOI 10.1038/sj.bjc.6604884 Mikesch J-H, Buerger H, Simon R, and Brandt B. Decay-accelerating factor, CD55): a versatile acting molecule in human malignancies. Biochim Biophys Acta, Bba, - Rev Cancer, 2006, 1766(1):42–52., DOI 10.1016/j.bbcan.2006. 04.001 Helman LJ, Gazdar AF, Park JG, Cohen PS, Cotelingam JD, and Israel MA. Chromogranin A expression in normal and malignant human tissues. J Clin Invest, 1988, 82(2):686–90., DOI 10.1172/jci113648 Bustin SA, Li S-R, and Dorudi S. Expression of the Ca2+-activated chloride channel genes CLCA1 and CLCA2 is downregulated in human colorectal cancer. DNA Cel Biol, 2001, 20(6):331–8., DOI 10.1089/ 10445490152122442 Toiyama Y, Inoue Y, Yasuda H, Saigusa S, Yokoe T, Okugawa Y, et al. DPEP1, expressed in the early stages of colon carcinogenesis, affects cancer cell invasiveness. J Gastroenterol, 2011, 46:153–63., DOI 10.1007/s00535-010- 0318-1 Yao X, Zhao G, Yang H, Hong X, Bie L, and Liu G. Overexpression of highmobility group box 1 correlates with tumor progression and poor prognosis in human colorectal carcinoma. J Cancer Res Clin Oncol, 2010, 136(5):677–84., DOI 10.1007/s00432-009-0706-1 Quesada-Calvo F, Massot C, Bertrand V, Longuespee R, Bletard N, Somja J, et al. OLFM4, KNG1 and Sec24C identified by proteomics and immunohistochemistry as potential markers of early colorectal cancer stages. Clin Proteom, 2017, 14(9)., DOI 10.1186/s12014-017-9143-3 Li A, Goto M, Horinouchi M, Tanaka S, Imai K, Kim YS, et al. Expression of MUC1 and MUC2 mucins and relationship with cell proliferative activity in human colorectal neoplasia. Pathol Int, 2001, 51(11):853–60., DOI 10.1046/j. 1440-1827.2001.01291.x Ogata S, Uehara H, Chen A, and Itzkowitz SH. Mucin gene expression in colonic tissues and cell lines. Cancer Res, 1992, 52(21):5971–8. Nozawa Y, Van Belzen N, Van der Made ACJ, Dinjens WNM, and Bosman FT. Expression of nucleophosmin/B23 in normal and neoplastic colorectal mucosa. J Pathol, 1996, 178(1):48–52., DOI 10.1002/(sici)1096-9896(199601, 178:1<48::aid-path432>3.0.co;2-y Yung BY. Oncogenic role of nucleophosmin/B23. Chang Gung Med J, 2007, 30(4):285–93. Vie N, Copois V, Bascoul-Mollevi C, Denis V, Bec N, Robert B, et al. Overexpression of phosphoserine aminotransferase PSAT1 stimulates cell growth and increases chemoresistance of colon cancer cells. Mol Cancer, 2008, 7:14., DOI 10.1186/1476-4598-7-14 Zheng H, Tsuneyama K, Cheng C, Takahashi H, Cui Z, Murai Y, et al. Maspin expression was involved in colorectal adenoma-adenocarcinoma sequence and liver metastasis of tumors. Anticancer Res, 2007, 27(1A):259–65. Powis G, Mustacich D, and Coon A. The role of the redox protein thioredoxin in cell growth and cancer. Free Radic Biol Med, 2000, 29(3–4):312–22., DOI 10. 1016/s0891-5849(00)00313-0 Fu Z, Yan K, Rosenberg A, Jin Z, Crain B, Athas G, et al. Improved protein extraction and protein identification from archival formalin-fixed paraffinembedded human aortas. Prot Clin Appl, 2013, 7:217–24., DOI 10.1002/prca. 201200064 Gamez-Pozo A, Ferrer NI, Ciruelos E, Lopez-Vacas R, Martinez FG, Espinosa E, et al. Shotgun proteomics of archival triple-negative breast cancer samples. Prot Clin Appl, 2013, 7:283–91., DOI 10.1002/prca.201200048 Wi´sniewski JR, Ostasiewicz P, and Mann M. High recovery FASP applied to the proteomic analysis of microdissected formalin fixed paraffin embedded cancer tissues retrieves known colon cancer markers. J Proteome Res, 2011, 10: 3040–9., DOI 10.1021/pr200019m Wi´sniewski JR, Du´s K, and Mann M. Proteomic workflow for analysis of archival formalin-fixed and paraffin-embedded clinical samples to a depth of 10 000 proteins. Proteomics Clin Appl, 2013, 7:225–33., DOI 10.1002/prca. 201200046 Lemaire R, Desmons A, Tabet JC, Day R, Salzet M, and Fournier I. Direct analysis and MALDI imaging of formalin-fixed, paraffin-embedded tissue sections. J Proteome Res, 2007, 6:1295–305., DOI 10.1021/ pr060549i Paine MRL, Ellis SR, Maloney D, Heeren RMA, and Verhaert PDEM. Digestion-free analysis of peptides from 30-year-old formalin-fixed, paraffin-embedded tissue by mass spectrometry imaging. Anal Chem, 2018, 90:9272–80., DOI 10.1021/acs.analchem.8b01838 Kyte J, and Doolittle RF. A simple method for displaying the hydropathic character of a protein. J Mol Biol, 1982, 157:105–32., DOI 10.1016/0022- 2836(82)90515-0 Moggridge S, Sorensen PH, Morin GB, and Hughes CS. Extending the compatibility of the SP3 paramagnetic bead processing approach for proteomics. J Proteome Res, 2018, 17(4):1730–40., DOI 10.1021/acs. jproteome.7b00913 Batth TS, Tollenaere MAX, Ruther PL, Gonzalez-Franquesa A, Prabhakar BS, Bekker-Jensen S, et al. Protein aggregation capture on microparticles enables multi-purpose proteomics sample preparation. Mol Cell Proteomics, 2018, 18(5):1027–35., DOI 10.1074/mcp.TIR118. 001270 Hughes CS, Moggridge S, Muller T, Sorensen PH, Morin GB, and Krijgsveld J. Single-pot, solid-phase-enhanced sample preparation for proteomics experiments. Nat Protoc, 2018, 14(1):1–18., DOI 10.1038/ s41596-018-0082-x Liu H, Ponniah G, Neill A, Patel R, and Andrien B. Accurate determination of protein methionine oxidation by stable isotope labeling and LC-MS analysis. Anal Chem, 2013, 85, 11705–9., DOI 10.1021/ac403072w Zang L, Carlage T, Murphy D, Frenkel R, Bryngelson P, Madsen M, et al. Residual metals cause variability in methionine oxidation measurements in protein pharmaceuticals using LC-UV/MS peptide mapping. J Chromatogr B Analyt Technol Biomed Life Sci, 2012, 895-896, 71–6., DOI 10.1016/j.jchromb. 2012.03.016 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 622855 EP 622874 DI 10.3389/pore.2021.622855 PG 20 ER PT J TI Brief Research Report Regional Difference in TRAF2 and TRAF3 Gene Mutations in Colon Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TRAF2; TRAF3; mutation; cancer; expression ID TRAF2; TRAF3; mutation; cancer; expression AB TRAF2 and TRAF3 genes of tumor necrosis factor receptor (TNF-R)-associated factor (TRAF) family are involved in diverse cell signaling, and function as both tumor suppressor gene and oncogene. Alterations of TRAF2 and TRAF3 in colon cancer (CC) along with their regional difference and microsatellite instability (MSI) are largely unknown. In the present study, we analyzed TRAF2 and TRAF3 frameshift mutations in 168 sporadic CCs (100 high MSI (MSI-H) and 68microsatellite-stable (MSS)CCs).Weidentified TRAF2 and TRAF3 frameshift mutations in 4 (4%) and 3 CCs (3%) with MSI-H, respectively, but none in 68 cases of MSS CCs. Of the 168 CCs,we analyzed the mutations inmulti-regions for 39CCs (16MSI-Hand23MSSCCs), and discovered that 12.5% (2/16) and 6.3%(1/16) ofMSI-H CCs exhibited regional difference in TRAF2 and TRAF3 mutations, respectively. In the multi-region samples of 23 MSS CCs, neither TRAF2 nor TRAF3 frameshift mutation was found. In 40% of CCs, both TRAF2 and TRAF3 expressions were increased compared to normal colon cells. Our data indicate that TRAF2 and TRAF3 frameshift mutations and their regional difference as well as altered expressions are present inMSI-H CCs, which could contribute toMSI-H cancer development. CR Bradley JR, Pober JS. Tumor necrosis factor receptor-associated factors, TRAFs). Oncogene, 2001, 20:6482–91., DOI 10.1038/sj.onc.1204788 Inoue J-I., Ishida T, Tsukamoto N, Kobayashi N, Naito A, Azuma S, et al. Tumor necrosis factor receptor-associated factor, TRAF, family: adapter proteins that mediate cytokine signaling. Exp Cell Res, 2000, 254:14–24., DOI 10.1006/excr.1999.4733 Lalani AI, Zhu S, Gokhale S, Jin J, Xie P. TRAF molecules in inflammation and inflammatory diseases. Curr Pharmacol Rep, 2018, 4:64–90., DOI 10.1007/ s40495-017-0117-y Park HH. Structure of TRAF family: current understanding of receptor recognition. Front Immunol, 2018, 9:1999., DOI 10.3389/fimmu.2018.01999 Xie P. TRAF molecules in cell signaling and in human diseases. JMS, 2013, 8:7., DOI 10.1186/1750-2187-8-7 Keats JJ, Fonseca R, Chesi M, Schop R, Baker A, Chng W-J, et al. Promiscuous mutations activate the noncanonical NF-κB pathway in multiple myeloma. Cancer Cell, 2007, 12:131–44., DOI 10.1016/j.ccr.2007.07.003 Doppler H, Liou GY, Storz P. Downregulation of TRAF2mediates NIK-induced pancreatic cancer cell proliferation and tumorigenicity. PLoS One, 2013, 8: e53676., DOI 10.1371/journal.pone.0053676 Zapata JM, Krajewska M, Krajewski S, Kitada S, Welsh K, Monks A, et al. TNFR-associated factor family protein expression in normal tissues and lymphoid malignancies. J Immunol, 2000, 165:5084–96., DOI 10.4049/ jimmunol.165.9.5084 Imai K, Yamamoto H. Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis, 2008, 29: 673–80., DOI 10.1093/carcin/bgm228 Son HJ, An CH, Yoo NJ, Lee SH. Tight junction-related CLDN5 and CLDN6 genes, and gap junction-related GJB6 and GJB7 genes are somatically mutated in gastric and colorectal cancers. Pathol Oncol Res, 2020, 26:1983–7., DOI 10. 1007/s12253-020-00806-2 Mo HY, An CH, Choi EJ, Yoo NJ, Lee SH. Somatic mutation and loss of expression of a candidate tumor suppressor gene TET3 in gastric and colorectal cancers. Pathol - Res Pract, 2020, 216:152759., DOI 10.1016/j.prp. 2019.152759 Cortes-Ciriano I, Lee S, Park WY, Kim TM, Park PJ. A molecular portrait of microsatellite instability across multiple cancers. Nat Commun, 2017, 8:15180., DOI 10.1038/ncomms15180 Takeuchi M, Rothe M, Goeddel DV. Anatomy of TRAF2, 1996, Distinct domains for nuclear factor-kappaB activation and association with tumor necrosis factor signaling proteins. J Biol Chem 271:19935–42., DOI 10.1074/jbc. 271.33.19935 Bokhari A, Jonchere V, Lagrange A, Bertrand R, Svrcek M, Marisa L, et al. Targeting nonsense-mediated mRNA decay in colorectal cancers with microsatellite instability. Oncogenesis, 2018, 7:70., DOI 10.1038/s41389-018- 0079-x Cheng G, Cleary A, Ye Z, Hong D, Lederman S, Baltimore D. Involvement of CRAF1, a relative of TRAF, in CD40 signaling. Science, 1995, 267:1494–8., DOI 10.1126/science.7533327 Chen M-B, Zhang Y, Wei M-X, Shen W, Wu X-Y, Yao C, et al. Activation of AMP-activated protein kinase, AMPK, mediates plumbagin-induced apoptosis and growth inhibition in cultured human colon cancer cells. Cell Signal, 2013, 25:1993–2002., DOI 10.1016/j.cellsig.2013.05.026 Piao J-H, Hasegawa M, Heissig B, Hattori K, Takeda K, Iwakura Y, et al. Tumor necrosis factor receptor-associated factor, TRAF, 2 controls homeostasis of the colon to prevent spontaneous development of murine inflammatory bowel disease. J Biol Chem, 2011, 286:17879–88., DOI 10.1074/ jbc.m111.221853 Allen IC, Wilson JE, Schneider M, Lich JD, Roberts RA, Arthur JC, et al. NLRP12 suppresses colon inflammation and tumorigenesis through the negative regulation of noncanonical NF-κB signaling. Immunity, 2012, 36: 742–54., DOI 10.1016/j.immuni.2012.03.012 Malcomson FC, Willis ND, McCallum I, Xie L, Lagerwaard B, Kelly S, et al. Non-digestible carbohydrates supplementation increases miR-32 expression in the healthy human colorectal epithelium: a randomized controlled trial. Mol Carcinog, 2017, 56:2104–11., DOI 10.1002/mc.22666 Hayashi K. PCR-SSCP: a simple and sensitive method for detection of mutations in the genomic DNA. PCR Methods Appl, 1991, 1:34–8., DOI 10. 1101/gr.1.1.34 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 625438 EP 625443 DI 10.3389/pore.2021.625438 PG 6 ER PT J AU Kong, J Yuan, X Wang, J Liu, Y Sun, W Gu, B Lan, Z Gao, Sh AF Kong, Jinyu Yuan, Xiang Wang, Jian Liu, Yiwen Sun, Wei Gu, Bianli Lan, Zijun Gao, Shegan TI Frequencies of Porphyromonas gingivalis Detection in Oral-Digestive Tract Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Porphyromonas gingivalis; oral squamous cell carcinoma; oesophageal squamous cell carcinoma; digestive tract cancer; prognosis ID Porphyromonas gingivalis; oral squamous cell carcinoma; oesophageal squamous cell carcinoma; digestive tract cancer; prognosis AB Mounting evidence suggests a causal relationship between specific bacterial infections and the development of certain malignancies. In this study, we examined the presence of Porphyromonas gingivalis (P. gingivalis) in oral-digestive tract tumors by immunohistochemistry (IHC) and PCR and analyzed the correlation between P. gingivalis detection and clinicopathological characteristics and prognosis of oral and esophageal carcinoma. The IHC results showed that the positive rates of P. gingivalis were 60.00, 46.00, 20.00, 6.67, and 2.86% in oral, esophagus, cardiac, stomach, and colorectal cancer tissues, respectively. Likewise, PCR results showed rates of 56.00, 42.00, 16.67, 3.33, and 2.86%, respectively. The two methods were consistent, and the kappa value was 0.806, P < 0.001. In addition, P. gingivalis expression was significantly correlated with lymph node metastasis and the clinical stages of oral and esophageal cancer (P < 0.05). The overall survival rate of the P. gingivalis undetected group (86, 50%) was significantly higher than that of the P. gingivalis detected group (57, 14%) for oral and esophageal cancer, respectively. In conclusion, the detection rate of P. gingivalis showed a decreasing trend in oral-digestive tract tumors. Detection with P. gingivalis was associated with poor prognosis for oral and esophageal cancer. C1 [Kong, Jinyu] The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Cancer InstituteLuoyang, China. [Yuan, Xiang] The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Cancer InstituteLuoyang, China. [Wang, Jian] The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Radiodiagnosis CenterLuoyang, China. [Liu, Yiwen] The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Cancer InstituteLuoyang, China. [Sun, Wei] The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Cancer InstituteLuoyang, China. [Gu, Bianli] The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Cancer InstituteLuoyang, China. [Lan, Zijun] The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Cancer InstituteLuoyang, China. [Gao, Shegan] The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Cancer InstituteLuoyang, China. RP Gao, Sh (reprint author), The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Cancer Institute, Luoyang, China. EM gsg112258@163.com CR Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA: A Cancer J Clinicians, 2015, 65:87–108., DOI 10.3322/caac. 21262 Gupta B, Johnson NW. Emerging and established global life-style risk factors for cancer of the upper aero-digestive tract. Asian Pac J Cancer Prev, 2014, 15: 5983–91., DOI 10.7314/apjcp.2014.15.15.5983 Shin H-R, Shin A, Woo H, Fox K, Walsh N, Lo Y-R, et al. Prevention of infection-related cancers in the WHO western pacific region. Jpn J Clin Oncol, 2016, 46:13–22., DOI 10.1093/jjco/hyv092 Crowe SE. Helicobacter infection, chronic inflammation, and the development of malignancy. Curr Opin Gastroenterol, 2005, 21:32–8., DOI 10.1007/978-4-431- 56068-5_47 Zhang L, Liu Y, Zheng HJ, Zhang CP. The oral microbiota may have influence on oral cancer. Front Cel Infect Microbiol, 2019, 9:476., DOI 10.3389/fcimb.2019. 00476 Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res, 2012, 22:299–306., DOI 10.1101/gr.126516.111 Ahn J, Segers S, Hayes RB. Periodontal disease, Porphyromonas gingivalis serum antibody levels and orodigestive cancer mortality. Carcinogenesis, 2012, 33: 1055–8., DOI 10.1093/carcin/bgs112 Darveau RP, Hajishengallis G, Curtis MA. Porphyromonas gingivalisas a potential community activist for disease. J Dent Res, 2012, 91:816–20., DOI 10.1177/0022034512453589 Fiorillo L, Cervino G, Laino L, D’Amico C, Mauceri R, Tozum TF, et al. Porphyromonas gingivalis, periodontal and systemic implications: a systematic review. Dent J, Basel,, 2019, 7:114., DOI 10.3390/dj7040114 Chang C, Wang H, Liu J, Pan C, Zhang D, Li X, et al. Porphyromonas gingivalis infection promoted the proliferation of oral squamous cell carcinoma cells through the miR-21/PDCD4/AP-1 negative signaling pathway. ACS Infect Dis, 2019, 5:1336–47., DOI 10.1021/acsinfecdis.9b00032 Mohammed H, Varoni EM, Cochis A, Cordaro M, Gallenzi P, Patini R, et al. Oral dysbiosis in pancreatic cancer and liver cirrhosis: a review of the literature. Biomedicines, 2018, 6., DOI 10.3390/biomedicines6040115 Katz J, Onate MD, Pauley KM, Bhattacharyya I, Cha S. Presence of Porphyromonas gingivalis in gingival squamous cell carcinoma. Int J Oral Sci, 2011, 3:209–15., DOI 10.4248/ijos11075 Yuan X, Liu Y, Kong J, Gu B, Qi Y, Wang X, et al. Different frequencies of Porphyromonas gingivalis infection in cancers of the upper digestive tract. Cancer Lett, 2017, 404:1–7., DOI 10.1016/j.canlet.2017.07.003 Gao S, Li S,Ma Z, Liang S, Shan T, Zhang M, et al. Presence of Porphyromonas gingivalis in esophagus and its association with the clinicopathological characteristics and survival in patients with esophageal cancer. Infect Agent Cancer, 2016, 11:3., DOI 10.1186/s13027-016-0049-x Gao SG, Yang JQ, Ma ZK, Yuan X, Zhao C, Wang GC, et al. Preoperative serum immunoglobulin G and A antibodies to Porphyromonas gingivalis are potential serum biomarkers for the diagnosis and prognosis of esophageal squamous cell carcinoma. BMC Cancer, 2018, 18:17., DOI 10.1186/s12885-017- 3905-1 Yuan X, Liu Y, Li G, Lan Z, Ma M, Li H, et al. Blockade of immune-checkpoint B7-H4 and lysine demethylase 5B in esophageal squamous cell carcinoma confers protective immunity against P. Gingivalis infection. Cancer Immunol Res, 2019, 7:1440–56., DOI 10.1158/2326-6066.cir-18-0709 Liang G, Wang H, Shi H, Zhu M, An J, Qi Y, et al. Porphyromonas gingivalis promotes the proliferation and migration of esophageal squamous cell carcinoma through the miR-194/GRHL3/PTEN/akt Axis. ACS Infect Dis, 2020, 6:871–81., DOI 10.1021/acsinfecdis.0c00007 Yilmaz O, Young PA, Lamont RJ, Kenny GE. Gingival epithelial cell signalling and cytoskeletal responses to Porphyromonas gingivalis invasion. Microbiology, Reading,, 2003, 149:2417–26., DOI 10.1099/mic.0.26483-0 Ashimoto A, Chen C, Bakker I, Slots J. Polymerase chain reaction detection of 8 putative periodontal pathogens in subgingival plaque of gingivitis and advanced periodontitis lesions. Oral Microbiol Immunol, 1996, 11:266–73., DOI 10.1111/j.1399-302x.1996.tb00180.x Cugini C, Klepac-Ceraj V, Rackaityte E, Riggs JE, Davey ME. Porphyromonas gingivalis: keeping the pathos out of the biont. J Oral Microbiol, 2013, 5., DOI 10. 3402/jom.v5i0.19804 Singhrao SK, Harding A, Poole S, Kesavalu L, Crean S. Porphyromonas gingivalis periodontal infection and its putative links with Alzheimer’s disease. Mediators Inflamm, 2015, 2015:137357., DOI 10.1155/2015/137357 Olsen I, Taubman MA, Singhrao SK. Porphyromonas gingivalis suppresses adaptive immunity in periodontitis, atherosclerosis, and Alzheimer’s disease. J Oral Microbiol, 2016, 8:33029., DOI 10.3402/jom.v8.33029 Hajishengallis G, Lamont RJ. Breaking bad: manipulation of the host response byPorphyromonas gingivalis. Eur J Immunol, 2014, 44:328–38., DOI 10.1002/eji. 201344202 Perera M, Al-Hebshi NN, Speicher DJ, Perera I, Johnson NW. Emerging role of bacteria in oral carcinogenesis: a review with special reference to periopathogenic bacteria. J Oral Microbiol, 2016, 8:32762., DOI 10.3402/jom.v8. 32762 Liu Y, Yuan X, Chen K, Zhou F, Yang H, Yang H, et al. Clinical significance and prognostic value of Porphyromonas gingivalis infection in lung cancer. Transl Oncol, 2021, 14:100972., DOI 10.1016/j.tranon.2020.100972 Whitmore SE, Lamont RJ. Oral bacteria and cancer. Plos Pathog, 2014, 10: e1003933., DOI 10.1371/journal.ppat.1003933 Yang CY, Yeh YM, Yu HY, Chin CY, Hsu CW, Liu H, et al. Oral microbiota community dynamics associated with oral squamous cell carcinoma staging. Front Microbiol, 2018, 9:862., DOI 10.3389/fmicb.2018.00862 Chang CR, Liu JC, Pan YP. “Correlation between Porphyromonas gingivalis and oral squamous cell carcinoma,” in Summary of the 10th national conference on periodontal disease,, 2014, Changchun, China. Lafuente Ibanez de Mendoza I, Maritxalar Mendia X, Garcia de la Fuente AM, Quindos Andres G, Aguirre Urizar JM. Role of Porphyromonas gingivalis in oral squamous cell carcinoma development: a systematic review. J Periodont Res, 2020, 55:13–22., DOI 10.1111/jre.12691 Peters BA, Wu J, Pei Z, Yang L, Purdue MP, Freedman ND, et al. Oral microbiome composition reflects prospective risk for esophageal cancers. Cancer Res, 2017, 77:6777–87., DOI 10.1158/0008-5472.can-17-1296 Gagliardi D, Makihara S, Corsi PR, De Toledo Viana A, Wiczer MVFS, Nakakubo S, et al. Microbial flora of the normal esophagus. Dis Esophagus, 1998, 11:248–50., DOI 10.1093/dote/11.4.248 Lawson RD, Coyle WJ. The noncolonic microbiome: does it really matter? Curr Gastroenterol Rep, 2010, 12:259–62., DOI 10.1007/ s11894-010-0111-6 Qi YJ, Jiao YL, Chen P, Kong JY, Gu BL, Liu K, et al. Porphyromonas gingivalis promotes progression of esophageal squamous cell cancer via TGFbetadependent Smad/YAP/TAZ signaling. Plos Biol, 2020, 18:e3000825., DOI 10. 1371/journal.pbio.3000825 Chen MF, Lu MS, Hsieh CC, Chen WC. Porphyromonas gingivalis promotes tumor progression in esophageal squamous cell carcinoma. Cel Oncol, Dordr,, 2020, 12, 34., DOI 10.1007/s13402-020-00573-x Salazar CR, Sun J, Li Y, Francois F, Corby P, Perez-Perez G, et al. Association between selected oral pathogens and gastric precancerous lesions. PLoS One, 2013, 8:e51604., DOI 10.1371/journal.pone.0051604 Brennan CA, Garrett WS. Gut microbiota, inflammation, and colorectal cancer. Annu Rev Microbiol, 2016, 70:395–411., DOI 10.1146/annurevmicro- 102215-095513 Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol, 2014, 12:661–72., DOI 10.1038/ nrmicro3344 Wu N, Yang X, Zhang R, Li J, Xiao X, Hu Y, et al. Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb Ecol, 2013, 66:462–70., DOI 10. 1007/s00248-013-0245-9 Ahn J, SinhaR,Pei Z,DominianniC,WuJ,Shi J, et al.Humangutmicrobiome andrisk for colorectal cancer. J Natl Cancer Inst, 2013, 105:1907–11., DOI 10.1093/jnci/djt300 Flemer B, Lynch DB, Brown JMR, Jeffery IB, Ryan FJ, Claesson MJ, et al. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut, 2017, 66:633–43., DOI 10.1136/gutjnl-2015-309595 Liang Q, Chiu J, Chen Y, Huang Y, Higashimori A, Fang J, et al. Fecal bacteria act as novel biomarkers for noninvasive diagnosis of colorectal cancer. Clin Cancer Res, 2017, 23:2061–70., DOI 10.1158/1078-0432.ccr-16-1599 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 628942 EP 628950 DI 10.3389/pore.2021.628942 PG 9 ER PT J AU Chung, SM Choi, JY Lee, SY Yoon, IB Ha, US AF Chung, Su Mun Choi, Jin Yeong Lee, Sub Young Yoon, Il Byung Ha, U-Syn TI How Much Reliable Is the Current Belief on Grade Group 1 Prostate Cancer? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prostatic neoplasms; prostatectomy; neoplasm grading; gleason score; neoplasm invasiveness ID prostatic neoplasms; prostatectomy; neoplasm grading; gleason score; neoplasm invasiveness AB Objective: To evaluate the clinicopathological characteristics of grade group 1 (GG1) prostate cancer in Korean populations. Methods: We retrospectively analyzed 492 consecutive radical prostatectomy specimens from our institution, which included those from 322 men with clinical GG1 and 170 with clinical GG2 tumors between years 2009 and 2018. The incidence of Gleason score (GS) upgrading, extraprostatic extension (EPE), and seminal vesicle invasion (SVI) were evaluated in patients with clinical GG1. In pathological GG1 cases, the distribution of adverse pathological features including EPE, lymphovascular invasion (LVI), perineural invasion (PNI), and biochemical recurrence (BCR) was analyzed. Results: Altogether, 78 (24.2%) out of 322 men in the clinical GG1 group demonstrated upgrading of GS, including 19 men with pathological Gleason score 4 + 3 = 7 and 6 with ≥ pathological Gleason score 4 + 4 = 8 cases. EPE was found in 37 (11.5%) and 22 (8.9%) men in clinical GG1 and pathological GG1 group, respectively. The incidence of LVI and PNI in the pathological GG1 cases was 2.8% (n = 7) and 28.6% (n = 71), respectively. BCR was observed in 4 men in pathological GG1 T2 (n = 226) and 2 men in GG1 T3 (n = 22) group. When we compared the pathological features between pathological GG1 T3 vs. GG2 T2, there was no statistical differences in the incidence of LVI and PNI between the two groups. Conclusions: Contrary to the current concept that GG1 is almost always clinically insignificant, it seems that GG1 still possess its respectable position as a group of cancer with aggressiveness. These findings should be kept in mind when deciding on treatment options for prostate cancer patients in the Asian populations. C1 [Chung, Su Mun] Catholic Kwandong University, International St. Mary’s Hospital, Department of UrologyIncheon, South Korea. [Choi, Jin Yeong] The Catholic University of Korea, College of Medicine, Seoul St. Mary’s HospitalSeoul, South Korea. [Lee, Sub Young] The Catholic University of Korea, College of Medicine, Eunpyeong St. Mary’s HospitalSeoul, South Korea. [Yoon, Il Byung] Catholic Kwandong University, International St. Mary’s Hospital, Department of UrologyIncheon, South Korea. [Ha, U-Syn] The Catholic University of Korea, College of Medicine, Seoul St. Mary’s HospitalSeoul, South Korea. RP Yoon, IB (reprint author), Catholic Kwandong University, International St. Mary’s Hospital, Department of Urology, Incheon, South Korea. EM yoonbi0948@catholic.ac.kr CR Epstein JI, Zelefsky MJ, Sjoberg DD, Nelson JB, Egevad L, and Magi-Galluzzi C. A contemporary prostate cancer grading system: a validated alternative to the Gleason score. Eur Urol, 2016, 69:428–35., DOI 10.1016/j.eururo.2015.06.046 Loeb S, Folkvaljon Y, Robinson D, Lissbrant IF, Egevad L, and Stattin P. Evaluation of the 2015 Gleason grade groups in a nationwide population-based cohort. Eur Urol, 2016, 69:1135–41., DOI 10.1016/j.eururo.2015.11.036 He J, Albertsen PC, Moore D, Rotter D, Demissie K, and Lu-Yao G. Validation of a contemporary five-tiered Gleason grade grouping using population-based data. Eur Urol, 2017, 71:760–3., DOI 10.1016/j.eururo.2016.11.031 Pompe RS, Davis-Bondarenko H, Zaffuto E, Tian Z, Shariat SF, and Leyh- Bannurah SR. Population-based validation of the 2014 ISUP Gleason grade groups in patients treated with radical prostatectomy, brachytherapy, external beam radiation, or no local Treatment. Prostate, 2017, 77:686–93., DOI 10.1002/ pros.23316 Miah S, Ahmed HU, Freeman A, and Emberton M. Does true Gleason pattern 3 merit its cancer descriptor? Nat Rev Urol, 2016, 13:541–8., DOI 10.1038/nrurol. 2016.141 Lepor H, and Donin NM. Gleason 6 prostate cancer: serious malignancy or toothless lion? Oncology, Williston Park,, 2014, 28:16–22. Hanahan D, and Weinberg RA. Hallmarks of cancer: the next generation. Cell, 2011, 144:646–74., DOI 10.1016/j.cell.2011.02.013 Nickel JC, and Speakman M. Should we really consider Gleason 6 prostate cancer? BJU Int, 2012, 109:645–6., DOI 10.1111/j.1464-410X.2011.10854.x Carter HB, Partin AW, Walsh PC, Trock BJ, Veltri RW, and Nelson WG. Gleason score 6 adenocarcinoma: should it be labeled as cancer? J Clin Oncol, 2012, 30:4294–6., DOI 10.1200/JCO.2012.44.0586 Ahmed HU, Arya M, Freeman A, and Emberton M. Do low-grade and lowvolume prostate cancers bear the hallmarks of malignancy? Lancet Oncol, 2012, 13(11):e509–517., DOI 10.1016/S1470-2045(12)70388-1 Anderson BB, Oberlin DT, Razmaria AA, Choy B, Zagaja GP, and Shalhav AL. Extraprostatic extension is extremely rare for contemporary Gleason score 6 prostate cancer. Eur Urol, 2017, 72:455–60., DOI 10.1016/j.eururo.2016.11.028 Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, and Trotti A. American Joint committee on cancer staging manual. 7th ed. New York, NY: Springer, 2010). Epstein JI, Egevad L, Amin MB, Delahunt B, Srigley JR, and Humphrey PA. The 2014 international society of urological Pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma: definition of grading patterns and proposal for a new grading system. Am J Surg Pathol, 2016, 40:244–52., DOI 10.1097/PAS.0000000000000530 Epstein JI, Feng Z, Trock BJ, and Pierorazio PM. Upgrading and downgrading of prostate cancer from biopsy to radical prostatectomy: incidence and predictive factors using the modified Gleason grading system and factoring in tertiary grades. Eur Urol, 2012, 61:1019–24., DOI 10.1016/j.eururo.2012. 01.050 Hassan O, Han M, Zhou A, Paulk A, Sun Y, and Al-Harbi A. Incidence of extraprostatic extension at radical prostatectomy with pure Gleason score 3 + 3  6, Grade Group 1, cancer: implications for whether Gleason score 6 prostate cancer should be renamed “not cancer” and for selection criteria for active surveillance. J Urol, 2018, 199:1482–7., DOI 10.1016/j.juro.2017.11.067 Mitsuzuka K, Narita S, Koie T, Kaiho Y, Tsuchiya N, and Yoneyama T. Lymphovascular invasion is significantly associated with biochemical relapse after radical prostatectomy even in patients with pT2N0 negative resection margin. Prostate Cancer Prostatic Dis, 2015, 18:25–30., DOI 10.1038/pcan. 2014.40 Park YH, Kim Y, Yu H, Choi IY, Byun SS, and Kwak C. Is lymphovascular invasion a powerful predictor for biochemical recurrence in pT3 N0 prostate cancer? Results from the K-CaP database. Sci Rep, 2016, 5(6):25419., DOI 10. 1038/srep25419 Yee DS, Shariat SF, Lowrance WT, Maschino AC, Savage CJ, and Cronin AM. Prognostic significance of lymphovascular invasion in radical prostatectomy specimens. BJU Int, 2011, 108:502–7., DOI 10.1111/j.1464- 410X.2010.09848.x Kang YJ, Kim HS, Jang WS, Kwon JK, Yoon CY, and Lee JY. Impact of lymphovascular invasion on lymph node metastasis for patients undergoing radical prostatectomy with negative resection margin. BMC Cancer, 2017, 8(17):321., DOI 10.1186/s12885-017-3307-4 Herman CM, Wilcox GE, Kattan MW, Scardino PT, and Wheeler TM. Lymphovascular invasion as a predictor of disease progression in prostate cancer. Am J Surg Pathol, 2000, 24:859–63., DOI 10.1097/00000478-200006000-00012 Zhang LJ,Wu B, Zha ZL, QuW, Zhao H, and Yuan J. Perineural invasion as an independent predictor of biochemical recurrence in prostate cancer following radical prostatectomy or radiotherapy: a systematic review and meta-analysis. BMC Urol, 2018, 18(4):5., DOI 10.1186/s12894-018-0319-6 Kang M, Oh JJ, Lee S, Hong SK, Lee SE, and Byun SS. Perineural invasion and lymphovascular invasion are associated with increased risk of biochemical recurrence in patients undergoing radical prostatectomy. Ann Surg Oncol, 2016, 23:2699–706., DOI 10.1245/s10434-016-5153-z Kraus RD, Barsky A, Ji L, Garcia Santos PM, Cheng N, and Groshen S. The perineural invasion paradox: is perineural invasion an independent prognostic indicator of biochemical recurrence risk in patients with pT2N0R0 prostate cancer? A multi-institutional study. Adv Radiat Oncol, 2018, 19(4):96–102. eCollection 2019 Jan-Mar., DOI 10.1016/j.adro.2018.09.006 Xu Y, Yang X, Si T, Yu H, ZhangW, and Li Y. Clinicopathological and prognostic factors in 106 prostate cancer patients aged ≤55 years: a single-center study in China. Med Sci Monit, 2016, 23(22):3935–42., DOI 10.12659/msm.901040 Man A, Pickles T, and Chi KN. British Columbia Cancer Agency Prostate Cohort Outcomes Initiative, 2003, Asian race and impact on outcomes after radical radiotherapy for localized prostate cancer. J Urol, 2003, 170:901–4., DOI 10.1097/01.ju.0000081423.37043.b4 Song C, Ro JY, Lee MS, Hong SJ, Chung BH, and Choi HY. Prostate cancer in Korean men exhibits poor differentiation and is adversely related to prognosis after radical prostatectomy. Urology, 2006, 68:820–4., DOI 10.1016/j.urology. 2006.04.029 Berman DM, and Epstein JI. When is prostate cancer really cancer? Urol Clin North Am, 2014, 41:339–46., DOI 10.1016/j.ucl.2014.01.006 Kulac I, Haffner MC, Yegnasubramanian S, Epstein JI, and De Marzo AM. Should Gleason 6 be labeled as cancer? Curr Opin Urol, 2015, 25:238–45., DOI 10.1097/MOU.0000000000000165 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 629489 EP 629495 DI 10.3389/pore.2021.629489 PG 7 ER PT J AU Zhou, J Zhang, Sh Sun, X Lou, Y Yu, J AF Zhou, Jingbo Zhang, Shu Sun, Xinyi Lou, Yan Yu, Jiangyi TI Hyperoside Protects HK-2 Cells Against High Glucose-Induced Apoptosis and Inflammation via the miR-499a-5p/NRIP1 Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE hyperoside; HK-2 cells; apoptosis; inflammation; molecular biology ID hyperoside; HK-2 cells; apoptosis; inflammation; molecular biology AB Hyperoside, a flavonol glycoside, is derived from plants of the genera Hypericum and Crataegus. Recent studies have indicated the anti-apoptotic and anti-inflammatory roles of hyperoside. The present study was designed to measure the effects of hyperoside on high glucose (HG)-treated HK-2 cells. HK-2 is a human papillomavirus 16 transformed cell line and can be used as a model for normal tubular cell. Cell apoptosis was examined by TUNEL assays and flow cytometry analysis. Inflammatory response was detected by Enzyme linked immunosorbent assay kits. Western blotting was applied to detect protein levels of apoptosis-related genes and inflammatory cytokines. Mechanistical assays including luciferase reporter and RNA pull down assays were applied to detect the binding relationship between molecules. We identified that hyperoside protected HK-2 cells against HG-induced apoptosis and inflammation. Moreover, miR-499a-5p was upregulated by hyperoside in a dose dependent manner. MiR-499a-5p inhibition rescued the suppressive effects of hyperoside on apoptosis and inflammation of HGtreated HK-2 cells. Furthermore, miR-499a-5p targeted NRIP1 to inhibit its mRNA expression, and further suppressed its translation. NRIP1 was downregulated by hyperoside in a dose dependent manner. Finally, rescue assays indicated that miR-499a-5p inhibition rescued the protective effects of hyperoside on apoptosis and inflammatory response of HK-2 cells by NRIP1. In conclusion, our findings revealed that hyperoside alleviates HG-induced apoptosis and inflammatory response of HK-2 cells by the miR-499a-5p/NRIP1 axis. C1 [Zhou, Jingbo] Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Department of EndocrinologyNanjing, China. [Zhang, Shu] Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Department of EndocrinologyNanjing, China. [Sun, Xinyi] Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Department of EndocrinologyNanjing, China. [Lou, Yan] Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Department of EndocrinologyNanjing, China. [Yu, Jiangyi] Jiangsu Provincial Hospital of Traditional Chinese Medicine, Department of EndocrinologyNanjing, China. RP Yu, J (reprint author), Jiangsu Provincial Hospital of Traditional Chinese Medicine, Department of Endocrinology, Nanjing, China. CR Boner G, Cooper ME. Diabetic nephropathy. Diabetes Technol Ther, 1999, 1(4): 489–96., DOI 10.1089/152091599317044 Xu Y, Wang L, He J, Bi Y, Li M, Wang T, et al. Prevalence and control of diabetes in Chinese adults. Jama, 2013, 310(9):948–59., DOI 10.1001/jama.2013.168118 Helou N, Dwyer A, Shaha M, Zanchi A Multidisciplinary management of diabetic kidney disease. JBI Database Syst Rev Implementation Rep, 2016, 14(7): 169–207., DOI 10.11124/jbisrir-2016-003011 Mesarosova L, Ochodnicky P, Leemans JC, Florquin S, Krenek P, Klimas J. High glucose induces HGF-independent activation of met receptor in human renal tubular epithelium. J Receptors Signal Transduction, 2017, 37(6):535–42., DOI 10. 1080/10799893.2017.1365902 Habib SL. Diabetes and renal tubular cell apoptosis. World J Diabetes, 2013, 4(2):27–30., DOI 10.4239/wjd.v4.i2.27 Sugiyama H, Kashihara N, Makino H, Yamasaki Y, Ota Z. Apoptosis in glomerular sclerosis. Kidney Int, 1996, 49(1):103–11., DOI 10.1038/ki.1996.14 Truong LD, Petrusevska G, Yang G, Gurpinar T, Shappell S, Lechago J, et al. Cell apoptosis and proliferation in experimental chronic obstructive uropathy. Kidney Int, 1996, 50(1):200–7., DOI 10.1038/ki.1996.303 Thomas G, Yang B, Wagner BE, Savill J, Nahas AME. Cellular apoptosis and proliferation in experimental renal fibrosis. Nephrol Dial Transpl, 1998, 13(9): 2216–26., DOI 10.1093/ndt/13.9.2216 Shimizu S, Konishi A, Kodama T, Tsujimoto Y. BH4 domain of antiapoptotic Bcl-2 family members closes voltage-dependent anion channel and inhibits apoptotic mitochondrial changes and cell death. Proc Natl Acad Sci, 2000, 97(7):3100–5., DOI 10.1073/pnas.97.7.3100 Rivero A, Mora C, Muros M, Garcia J, Herrera H, Navarro-Gonzalez JF. Pathogenic perspectives for the role of inflammation in diabetic nephropathy. Clin Sci, 2009, 116(6):479–92., DOI 10.1042/cs20080394 Navarro-Gonzalez JF, Mora-Fernandez C, de Fuentes MM, Garcia-Perez J. Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol, 2011, 7(6):327–40., DOI 10.1038/nrneph. 2011.51 Zou Y, Lu Y, Wei D. Antioxidant activity of a flavonoid-rich extract of hypericum perforatumL.in Vitro. J Agric Food Chem, 2004, 52(16):5032–9., DOI 10.1021/jf049571r Kim S-J, Um J-Y, Hong S-H, Lee J-Y. Anti-inflammatory activity of hyperoside through the suppression of nuclear factor-κb activation in mouse peritoneal macrophages. Am J Chin Med, 2011, 39(1):171–81., DOI 10.1142/ s0192415x11008737 Ku S-K, Kwak S, Kwon O-J, Bae J-S. Hyperoside inhibits high-glucose-induced vascular inflammation in vitro and in vivo. Inflammation, 2014, 37(5): 1389–400., DOI 10.1007/s10753-014-9863-8 Ku S-K, Zhou W, Lee W, Han M-S, Na M, Bae J-S. Anti-inflammatory effects of hyperoside in human endothelial cells and in mice. Inflammation, 2015, 38(2):784–99., DOI 10.1007/s10753-014-9989-8 Guo X, Zhang Y, Lu C, Qu F, Jiang X. Protective effect of hyperoside on heart failure rats via attenuating myocardial apoptosis and inducing autophagy. Biosci Biotechnol Biochem, 2020, 84(4):714–24., DOI 10.1080/09168451.2019. 1685369 Wu L, Li Q, Liu S, An X, Huang Z, Zhang B, et al. Protective effect of hyperoside against renal ischemia-reperfusion injury via modulating mitochondrial fission, oxidative stress, and apoptosis. Free Radic Res, 2019, 53(7):727–36., DOI 10.1080/10715762.2019.1623883 Cao X, Fan Q-L. LncRNA MIR503HG promotes high-glucose-induced proximal tubular cell apoptosis by targeting miR-503-5p/bcl-2 pathway. Dmso, 2020, Vol. 13:4507–17., DOI 10.2147/dmso.s277869 Li S, Zheng L, Zhang J, Liu X, Wu Z. Inhibition of ferroptosis by up-regulating Nrf2 delayed the progression of diabetic nephropathy. Free Radic Biol Med, 2020, 162:435–449., DOI 10.1016/j.freeradbiomed.2020.10.323 Salti T, Khazim K, Haddad R, Campisi-Pinto S, Bar-Sela G, Cohen I. Glucose induces IL-1α-dependent inflammation and extracellular matrix proteins expression and deposition in renal tubular epithelial cells in diabetic kidney disease. Front Immunol, 2020, 11:1270., DOI 10.3389/fimmu.2020.01270 Li J-H, Liu S, Zhou H, Qu L-H, Yang J-H. starBase v2.0: decoding miRNAceRNA, miRNA-ncRNA and protein-RNA interaction networks from largescale CLIP-Seq data. Nucl Acids Res, 2014, 42(D1):D92–D97., DOI 10.1093/nar/ gkt1248 Livak KJ, Schmittgen TD. Analysis of relative gene expression data using realtime quantitative PCR and the 2−ΔΔCT method. Methods, 2001, 25(4):402–8., DOI 10.1006/meth.2001.1262 Yang XH, Zhang BL, Zhang XM, Tong JD, Gu YH, Guo LL, et al. EGCG attenuates renal damage via reversing klotho hypermethylation in diabetic db/ db mice and HK-2 cells. Oxid Med Cel Longev, 2020, 2020:6092715., DOI 10. 1155/2020/6092715 Lv L, Zhang J, Tian F, Li X, Li D, Yu X. Arbutin protects HK-2 cells against high glucose-induced apoptosis and autophagy by up-regulating microRNA-27a. Artif Cell Nanomedicine Biotechnol, 2019, 47(1):2940–7., DOI 10.1080/ 21691401.2019.1640231 Lv N, Li C, Liu X, Qi C, Wang Z. miR-34b alleviates high glucose-induced inflammation and apoptosis in human HK-2 cells via IL-6R/JAK2/STAT3 signaling pathway. Med Sci Monit, 2019, 25:8142–51., DOI 10.12659/msm. 917128 Wu J, Liu J, Ding Y, Zhu M, Lu K, Zhou J, et al. MiR-455-3p suppresses renal fibrosis through repression of ROCK2 expression in diabetic nephropathy. Biochem Biophys Res Commun, 2018, 503(2):977–83., DOI 10.1016/j.bbrc.2018. 06.105 Wang L, Zhang N, Pan H-p., Wang Z, Cao Z-y. MiR-499-5p contributes to hepatic insulin resistance by suppressing PTEN. Cell Physiol Biochem, 2015, 36(6):2357–65., DOI 10.1159/000430198 Fluitt MB, Kumari N, Nunlee-Bland G, Nekhai S, Gambhir KK. miRNA-15a, miRNA-15b, and miRNA-499 are reduced in erythrocytes of pre-diabetic african-American adults. Jacobs J Diabetes Endocrinol, 2016, 2(1):014 Ciccacci C, Latini A, Greco C, Politi C, D’Amato C, Lauro D, et al. Association between a MIR499A polymorphism and diabetic neuropathy in type 2 diabetes. J Diabetes its Complications, 2018, 32(1):11–7., DOI 10.1016/j. jdiacomp.2017.10.011 Fawzy MS, Abu AlSel BT, Al Ageeli E, Al-Qahtani SA, Abdel-Daim MM, Toraih EA. Long non-coding RNA MALAT1 and microRNA-499a expression profiles in diabetic ESRD patients undergoing dialysis: a preliminary crosssectional analysis. Arch Physiol Biochem, 2020, 126(2):172–82., DOI 10.1080/ 13813455.2018.1499119 L’Horset F, Dauvois S, Heery DM, Cavailles V, Parker MG. RIP-140 interacts with multiple nuclear receptors by means of two distinct sites. Mol Cel Biol., 1996, 16(11):6029–36., DOI 10.1128/mcb.16.11.6029 Seth A, Steel JH, Nichol D, Pocock V, Kumaran MK, Fritah A, et al. The transcriptional corepressor RIP140 regulates oxidative metabolism in skeletal muscle. Cel Metab, 2007, 6(3):236–45., DOI 10.1016/j.cmet.2007.08.004 Xue J, Zhao H, Shang G, Zou R, Dai Z, Zhou D, et al. RIP140 is associated with subclinical inflammation in type 2 diabetic patients. Exp Clin Endocrinol Diabetes, 2013, 121(1):37–42., DOI 10.1055/s-0032-1323683 Zschiedrich I, Hardeland U, Krones-Herzig A, Berriel Diaz M, Vegiopoulos A, Muggenburg J, et al. Coactivator function of RIP140 for NFκB/RelAdependent cytokine gene expression. Blood, 2008, 112(2):264–76., DOI 10. 1182/blood-2007-11-121699 Ho P-C, Tsui Y-C, Feng X,GreavesDR,Wei L-NNF-κB-mediated degradation of the coactivator RIP140 regulates inflammatory responses and contributes to endotoxin tolerance. Nat Immunol, 2012, 13(4):379–86., DOI 10.1038/ni.2238 Ho P-C, Chang K-C, Chuang Y-S, Wei L-N. Cholesterol regulation of receptor-interacting protein 140 via microRNA-33 in inflammatory cytokine production. FASEB j., 2011, 25(5):1758–66., DOI 10.1096/fj.10-179267 Yi Z-J, Gong J-P, ZhangW. Transcriptional co-regulator RIP140: an important mediator of the inflammatory response and its associated diseases. Mol Med Rep, 2017, 16(2):994–1000., DOI 10.3892/mmr.2017.6683 Li W, Liu M, Xu Y-F, Feng Y, Che J-P, Wang G-C, et al. Combination of quercetin and hyperoside has anticancer effects on renal cancer cells through inhibition of oncogenic microRNA-27a. Oncol Rep, 2014, 31(1):117–24., DOI 10.3892/or.2013.2811 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 629829 EP 629837 DI 10.3389/pore.2021.629829 PG 9 ER PT J AU Valdivia, D Cheufou, D Fels, B Puhlvers, S Mardanzai, K Zaatar, M Weinreich, G Taube, Ch Theegarten, D Stuschke, M Schuler, M Stamatis, G Hegedus, B Aigner, C AF Valdivia, Daniel Cheufou, Danjouma Fels, Benjamin Puhlvers, Stephan Mardanzai, Khaled Zaatar, Mohamed Weinreich, Gerhard Taube, Christian Theegarten, Dirk Stuschke, Martin Schuler, Martin Stamatis, Georgios Hegedus, Balazs Aigner, Clemens TI Potential Prognostic Value of Preoperative Leukocyte Count, Lactate Dehydrogenase and C-Reactive Protein in Thymic Epithelial Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE thymoma; thymic epithelial tumor; thymectomy; LDH; CRP; C-reactive protein ID thymoma; thymic epithelial tumor; thymectomy; LDH; CRP; C-reactive protein AB Thymic epithelial tumors are the most common mediastinal tumors. Surgery is the mainstay of treatment and complete resection provides the best survival rate. However, advanced tumors often require multimodality treatment and thus we analyzed the prognostic potential of routine circulating biomarkers that might help to risk-stratify patients beyond tumor stage and histology. Preoperative values for white blood cell count (WBC), C-reactive protein (CRP) and lactate dehydrogenase (LDH) were analyzed in 220 thymic epithelial tumor patients operated between 1999 and 2018. Increased CRP levels (>1 mg/dl) were significantly more often measured in thymic carcinoma and neuroendocrine tumors when compared to thymoma. LDH serum activity was higher in thymic neuroendocrine tumors when compared to thymoma or thymic carcinoma. The median disease specific survival was significantly longer in thymoma cases than in thymic carcinoma and neuroendocrine tumors. Increased preoperative LDH level (>240 U/L) associated with shorter survival in thymus carcinoma (HR 4.76, p = 0.0299). In summary, higher CRP associated with carcinoma and neuroendocrine tumors, while LDH increased primarily in neuroendocrine tumors suggesting that biomarker analysis should be performed in a histology specific manner. Importantly, preoperative serum LDH might be a prognosticator in thymic carcinoma and may help to risk stratify surgically treated patients in multimodal treatment regimens. C1 [Valdivia, Daniel] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Cheufou, Danjouma] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Fels, Benjamin] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Puhlvers, Stephan] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Mardanzai, Khaled] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Zaatar, Mohamed] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Weinreich, Gerhard] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of PulmonologyEssen, Germany. [Taube, Christian] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of PulmonologyEssen, Germany. [Theegarten, Dirk] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Stuschke, Martin] University Duisburg-Essen, West German Cancer Center, University Hospital Essen, Department of Radiation OncologyEssen, Germany. [Schuler, Martin] University Duisburg- Essen, West German Cancer Center, University Hospital Essen, Department of Medical OncologyEssen, Germany. [Stamatis, Georgios] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Hegedus, Balazs] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Aigner, Clemens] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. RP Aigner, C (reprint author), University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic Surgery, Essen, Germany. EM clemens.aigner@rlk.uk-essen.de CR Kondo K. Therapy for thymic epithelial tumors. Gen Thorac Cardiovasc Surg, 2014, 62:468–74., DOI 10.1007/s11748-014-0420-z Berardi R, Morgese F, Garassino MC, and Cascinu S. New findings on thymic epithelial tumors: something is changing. World J Clin Oncol, 2015, 6:96–8., DOI 10.5306/wjco.v6.i5.96 Marx A, Chan JKC, Coindre J-M, Detterbeck F, Girard N, Harris NL, et al. The 2015 world health organization classification of tumors of the thymus: continuity and changes. J Thorac Oncol, 2015, 10:1383–95., DOI 10.1097/jto. 0000000000000654 Moser B, Scharitzer M, Hacker S, Ankersmit J, Matilla JR, Lang G, et al. Thymomas and thymic carcinomas: prognostic factors and multimodal management. Thorac Cardiovasc Surg, 2014, 62:153–60., DOI 10.1055/s- 0032-1322611 Strobel P, Bauer A, Puppe B, Kraushaar T, Krein A, Toyka K, et al. Tumor recurrence and survival in patients treated for thymomas and thymic squamous cell carcinomas: a retrospective analysis. J Clin Oncol, 2004, 22: 1501–9., DOI 10.1200/jco.2004.10.113 Filosso PL, Yao X, Ahmad U, Zhan Y, Huang J, Ruffini E, et al. Outcome of primary neuroendocrine tumors of the thymus: a joint analysis of the international thymic malignancy interest group and the European society of thoracic surgeons databases. J Thorac Cardiovasc Surg, 2015, 149:103–9., DOI 10.1016/j.jtcvs.2014.08.061 Ruffini E, Detterbeck F, Van Raemdonck D, Rocco G, Thomas P, Weder W, et al. Thymic carcinoma: a cohort study of patients from the European society of thoracic surgeons database. J Thorac Oncol, 2014, 9:541–8., DOI 10.1097/jto. 0000000000000128 Marulli G, Margaritora S, Lucchi M, Cardillo G, Granone P, Mussi A, et al. Surgical treatment of recurrent thymoma: is it worthwhile?. Eur J Cardiothorac Surg, 2016, 49:327–32., DOI 10.1093/ejcts/ezv086 Hamaji M, Allen MS, Cassivi SD, Nichols FC, 3rd, Wigle DA, Deschamps C, et al. The role of surgical management in recurrent thymic tumors. Ann Thorac Surg, 2012, 94:247–54. discussion 254., DOI 10.1016/j.athoracsur. 2012.02.092 Dai J, Song N, Yang Y, and Jiang G. Is it valuable and safe to perform reoperation for recurrent thymoma?: Table 1:. Interact Cardiovasc Thorac Surg, 2015, 21:526–31., DOI 10.1093/icvts/ivv144 Bae MK, Byun CS, Lee CY, Lee JG, Park IK, Kim DJ, et al. Clinical outcomes and prognosis of recurrent thymoma management. J Thorac Oncol, 2012, 7: 1304–14., DOI 10.1097/jto.0b013e3182598a91 Pepys MB, and Hirschfield GM. C-reactive protein: a critical update. J Clin Invest, 2003, 111:1805–12., DOI 10.1172/jci200318921 Janik S, Bekos C, Hacker P, Raunegger T, Ghanim B, Einwallner E, et al. Elevated CRP levels predict poor outcome and tumor recurrence in patients with thymic epithelial tumors: a pro- and retrospective analysis. Oncotarget, 2017, 8:47090–102., DOI 10.18632/oncotarget.17478 Quoix E, Purohit A, Faller-Beau M, Moreau L, Oster JP, and Pauli G. Comparative prognostic value of lactate dehydrogenase and neuron-specific enolase in small-cell lung cancer patients treated with platinum-based chemotherapy. Lung Cancer, 2000, 30:127–34., DOI 10.1016/s0169-5002(00, 00131-8 Zhou G-Q, Tang L-L, Mao Y-P, Chen L, Li W-F, Sun Y, et al. Baseline serum lactate dehydrogenase levels for patients treated with intensity-modulated radiotherapy for nasopharyngeal carcinoma: a predictor of poor prognosis and subsequent liver metastasis. Int J Radiat Oncology*Biology*Physics, 2012, 82: e359–e365., DOI 10.1016/j.ijrobp.2011.06.1967 Shamash J, Oliver RT, Gallagher CJ, Newland AC, Lister TA, Kelsey S, et al. Pre-induction LDH as a prognostic factor for outcome of high dose chemotherapy, HDCT, for germ cell tumours relapsing or refractory to conventional chemotherapy. Br J Cancer, 2000, 82:2022–3., DOI 10.1054/ bjoc.1999.1140 Liu Q, Fu X, Su X, Wang X, Zhang Y, Yang H, et al. Elevated pretreatment serum lactate dehydrogenase level predicts inferior overall survival and disease-free survival after resection of thymic carcinoma. J Thorac Dis, 2017, 9:4550–60., DOI 10.21037/jtd.2017.10.86 Yuan ZY, Gao SG, Mu JW, Xue Q, Mao YS, Wang DL, et al. Prognostic value of preoperative serum lactate dehydrogenase in thymic carcinoma. J Thorac Dis, 2016, 8:2464–72., DOI 10.21037/jtd.2016.08.56 N.C.C. Network. NCCN thymomas and thymic carcinomas, version 2.2016). Plymouth Meeting, PA: National Comprehensive Cancer Network, 2016). Bhora FY, Chen DJ, Detterbeck FC, Asamura H, Falkson C, Filosso PL, et al. The ITMIG/IASLC thymic epithelial tumors staging project: a proposed lymph node map for thymic epithelial tumors in the forthcoming 8th edition of the TNM classification of malignant tumors. J Thorac Oncol, 2014, 9:S88–S96., DOI 10.1097/jto.0000000000000293 Masaoka A, Monden Y, Nakahara K, and Tanioka T. Follow-up study of thymomas with special reference to their clinical stages. Cancer, 1981, 48: 2485–92., DOI 10.1002/1097-0142(19811201)48:11<2485::aid-cncr2820481123>3.0.co; 2-r Koga K, Matsuno Y, NoguchiM,Mukai K, Asamura H, Goya T, et al. A review of 79 thymomas: modification of staging system and reappraisal of conventional division into invasive and non-invasive thymoma. Pathol Int, 1994, 44:359–67., DOI 10.1111/j.1440-1827.1994.tb02936.x Travis WDWorld health Organization; International agency for research on Cancer; International association for the study of lung cancer; International Academy of Pathology; Pathology and genetics of tumours of the lung, pleura, thymus and heart. Lyon: IARC Press, 2004). Haigl B, Vanas V, Setinek U, Hegedus B, Gsur A, and Sutterluty-fall H. Expression of microRNA-21 in non-small cell lung cancer tissue increases with disease progression and is likely caused by growth conditional changes during malignant transformation. Int J Oncol, 2014, 44:1325–34., DOI 10.3892/ijo.2014.2272 Filosso PL, Ruffini E, Solidoro P, Roffinella M, Lausi PO, Lyberis P, et al. Neuroendocrine tumors of the thymus. J Thorac Dis, 2017, 9:S1484–S1490., DOI 10.21037/jtd.2017.10.83 Fukai I, Masaoka A, Fujii Y, Yamakawa Y, Yokoyama T, Murase T, et al. Thymic neuroendocrine tumor, thymic carcinoid): a clinicopathologic study in 15 patients. Ann Thorac Surg, 1999, 67:208–11., DOI 10.1016/s0003-4975(98, 01063-7 Oberg K, Hellman P, Ferolla P, Papotti M, and Group EGW. Neuroendocrine bronchial and thymic tumors: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol, 2012, 23(Suppl. 7): vii120–vii123., DOI 10.1093/annonc/mds267 Girard N. Neuroendocrine tumors of the thymus: the oncologist point of view. J Thorac Dis, 2017, 9:S1491–S1500., DOI 10.21037/jtd.2017.08.18 Bakhos CT, Salami AC, Kaiser LR, Petrov RV, and Abbas AE. Thymic neuroendocrine tumors and thymic carcinoma: demographics, treatment, and survival. Innovations, Phila, 15, 2020, 468–74., DOI 10.1177/1556984520949287 Sullivan JL, and Weksler B. Neuroendocrine tumors of the thymus: analysis of factors affecting survival in 254 patients. Ann Thorac Surg, 2017, 103:935–9., DOI 10.1016/j.athoracsur.2016.07.050 Veraar C, Janik S, Thanner J, Veraar C, Mouhieddine M, Schiefer AI, et al. Clinical prognostic scores for patients with thymic epithelial tumors. Sci Rep, 2019, 9:18581., DOI 10.1038/s41598-019-54906-4 Okada S, Shimomura M, Tsunezuka H, Ishihara S, Ikebe S, Furuya T, et al. High neutrophil count as a negative prognostic factor for relapse in patients with thymic epithelial tumor. Ann Surg Oncol, 2020, 27:2438–47., DOI 10.1245/ s10434-020-08228-9 Yang DH, Qian MZ, Wei MM, Li J, Yu MM, Lu XM, et al. The correlation of neutrophil-to-lymphocyte ratio with the presence and activity of myasthenia gravis. Oncotarget, 2017, 8:76099–107., DOI 10.18632/oncotarget.18546 Yuan ZY, Gao SG, Mu JW, Xue Q, Mao YS, Wang DL, et al. Prognostic value of preoperative neutrophil-lymphocyte ratio is superior to platelet-lymphocyte ratio for survival in patients who underwent complete resection of thymic carcinoma. J Thorac Dis, 2016, 8:1487–96., DOI 10.21037/jtd.2016.05.05 Janik S, Raunegger T, Hacker P, Ghanim B, Einwallner E, Mullauer L, et al. Prognostic and diagnostic impact of fibrinogen, neutrophil-to-lymphocyte ratio, and platelet-to-lymphocyte ratio on thymic epithelial tumors outcome. Oncotarget, 2018, 9:21861–75., DOI 10.18632/oncotarget.25076 Yanagiya M, Nitadori JI, Nagayama K, Anraku M, Sato M, and Nakajima J. Prognostic significance of the preoperative neutrophil-to-lymphocyte ratio for complete resection of thymoma. Surg Today, 2018, 48:422–30., DOI 10.1007/ s00595-017-1602-y Szathmary I, Selmeci L, Posch E, Szobor A, and Molnar J. Myasthenia gravis: long-term prognostic value of thymus lactate dehydrogenase isoenzyme pattern of hyperplastic thymus and thymoma. J Neurol Neurosurg Psychiatry, 1985, 48:757–61., DOI 10.1136/jnnp.48.8.757 Wu JX, Chen HQ, Shao LD, Qiu SF, Ni QY, Zheng BH, et al. Long-term follow-up and prognostic factors for advanced thymic carcinoma. Medicine, Baltimore,, 2014, 93:e324., DOI 10.1097/md. 0000000000000324 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 629993 EP 630001 DI 10.3389/pore.2021.629993 PG 9 ER PT J AU Szekely, R Suhai, IF Karlinger, K Baksa, G Szabaczki, B Barany, L Poloskei, G Racz, G Wagner, Merkely, B Ruttkay, T AF Szekely, Reka Suhai, Imre Ferenc Karlinger, Kinga Baksa, Gabor Szabaczki, Bence Barany, Laszlo Poloskei, Gergely Racz, Gergely Wagner, Odon Merkely, Bela Ruttkay, Tamas TI Human Cadaveric Artificial Lung Tumor-Mimic Training Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tumor mimic; pseudotumor; cadaver workshop; hands-on training; lung tumor ID tumor mimic; pseudotumor; cadaver workshop; hands-on training; lung tumor AB Introduction: An important phase in surgical training is gaining experience in real human anatomical situations. When a cadaver is available it may complement the various artificial practice models. However, it is often necessary to supplement the characteristics of the cadavers with a simulation of a tumor. Our objective was to develop an easy-to-create, realistic artificial tumor-mimic model for peripheral lung tumor resection practice. Methods: In our work we injected barium sulphate enriched silicone suspension into 10 isolated, non-fixed lungs of human cadavers, through the puncture of the visceral pleura. Four lesions–apical, hilar and two peripheral–were created in each of ten specimens. After fixation CT scans were obtained and analyzed. The implanted tumor-mimics were examined after anatomical preparation and slicing. Also performed CT-guided percutaneous puncture was also performed to create the lesions in situ in two lungs of human cadavers. Results: Analyzing the CT data of 10 isolated lungs, out of 40 lesions, 34 were nodular (85.0%) and in the nodular group five were spiculated (12.5%). Satellite lesions were formed in two cases (5.0%). Relevant outflow into vessels or airway occurred in five lesions (12.5%). Reaching the surface of the lung occured in 11 lesions (27.5%). The tumormimics were elastic and adhered well to the surrounding tissue. The two lesions, implanted via percutaneous puncture, both were nodular and one also showed lobulated features. Conclusion: Our artificial tumor-mimics were easy to create, varied in shape and size, and with percutaneous implantation the lesions provide a model for teaching every step of a surgical procedure. C1 [Szekely, Reka] Semmelweis University, Department of Anatomy, Histology and Embryology, Laboratory for Applied and Clinical AnatomyBudapest, Hungary. [Suhai, Imre Ferenc] Semmelweis University, Department of Cardiovascular SurgeryBudapest, Hungary. [Karlinger, Kinga] Semmelweis University, Medical Imaging CenterBudapest, Hungary. [Baksa, Gabor] Semmelweis University, Department of Anatomy, Histology and Embryology, Laboratory for Applied and Clinical AnatomyBudapest, Hungary. [Szabaczki, Bence] Semmelweis University, Department of Anatomy, Histology and Embryology, Laboratory for Applied and Clinical AnatomyBudapest, Hungary. [Barany, Laszlo] Semmelweis University, Department of Anatomy, Histology and Embryology, Laboratory for Applied and Clinical AnatomyBudapest, Hungary. [Poloskei, Gergely] Semmelweis University, Medical Imaging CenterBudapest, Hungary. [Racz, Gergely] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Wagner, Odon] Budapest University of Technology and Economics, Department of Inorganic and Analytical ChemistryBudapest, Hungary. [Merkely, Bela] Semmelweis University, Department of Cardiovascular SurgeryBudapest, Hungary. [Ruttkay, Tamas] Semmelweis University, Department of Anatomy, Histology and Embryology, Laboratory for Applied and Clinical AnatomyBudapest, Hungary. RP Racz, G (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM racz.gergely@med.semmelweisuniv.hu CR Faure JP, Breque C, Danion J, Delpech PO, Oriot D, and Richer JPSIM Life: a new surgical simulation device using a human perfused cadaver. Surg Radiol Anat, 2017, 39(2):211–7., DOI 10.1007/s00276-016-1715-9 Chai DQ, Naunton-Morgan R, and Hamdorf JFresh frozen cadaver workshops for general surgical training. ANZ J Surg, 2019, 89(11):1428–31., DOI 10.1111/ ans.15258 Greene CL, Minneti M, Sullivan ME, and Baker CJPressurized cadaver model in cardiothoracic surgical simulation. Ann Thorac Surg, 2015, 100(3):1118–20., DOI 10.1016/j.athoracsur.2015.02.134 Kovacs G, Levitan R, and Sandeski RClinical cadavers as a simulation resource for procedural learning. AEM Educ Train, 2018, 2(3):239–47., DOI 10.1002/ aet2.10103 Hornblower VDM, Yu E, Fenster A, Battista JJ, and Malthaner RA3D thoracoscopic ultrasound volume measurement validation in anex vivoandin vivoporcine model of lung tumours. Phys Med Biol, 2007, 52(1): 91–106., DOI 10.1088/0031-9155/52/1/007 Scott D, Young W, Watumull L, Lindberg G, Fleming J, Rege R, et al.Development of an in vivo tumor-mimic model for learning radiofrequency ablation. J Gastrointest Surg, 2000, 4(6):620–5., DOI 10.1016/ s1091-255x(00)80112-2 Taylor GD, Johnson DB, Hogg DC, and Cadeddu JADevelopment of a renal tumor mimic model for learning minimally invasive nephron sparing surgical techniques. J Urol, 2004, 172(1):382–5., DOI 10.1097/01.ju.0000132358. 82641.10 Nomori H, Imazu Y, Watanabe K, Ohtsuka T, Naruke T, Kobayashi T, et al.Radiofrequency ablation of pulmonary tumors and normal lung tissue in swine and rabbits. Chest, 2005, 127(3):973–7., DOI 10.1378/chest.127.3.973 Chen A, Machuzak M, Edell E, and Silvestri GAPeripheral bronchoscopy training using a human cadaveric model and simulated tumor targets. J Bronchology Interv Pulmonol, 2016, 23(1):83–6., DOI 10.1097/LBR. 0000000000000200 Wolfram F, Boltze C, Schubert H, Bischoff S, and Lesser TEffect of lung flooding and high-intensity focused ultrasound on lung tumours: an experimental study in an ex vivo human cancer model and simulated in vivo tumours in pigs. Eur J Med Res, 2014, 19:1., DOI 10.1186/2047-783X- 19-1 Eun D, Bhandari A, Boris R, Lyall K, Bhandari M, Menon M, et al. A novel technique for creating solid renal pseudotumors and renal vein-inferior vena caval pseudothrombus in a porcine and cadaveric model. J Urol, 2008, 180(4): 1510–4., DOI 10.1016/j.juro.2008.06.005 Vegesna A, Raju R, Asfari W, Korimilli A, Varia A, OrfanidisN, et al.Threedimensional US volume analysis of gastric pseudotumors in a porcine model. Gastrointest Endosc, 2006, 64(4):635–40., DOI 10.1016/j.gie.2006. 04.034 Kawai T, Kaminou T, Sugiura K, Hashimoto M, Ohuchi Y, Adachi A, et al.Creation of a tumor-mimic model using a muscle paste for radiofrequency ablation of the lung. Cardiovasc Intervent Radiol, 2009, 32(2):296–302., DOI 10.1007/s00270-008-9463-9 Kawai T, Kaminou T, Sugiura K, Hashimoto M, Ohuchi Y, Adachi A, et al.Percutaneous radiofrequency lung ablation combined with transbronchial saline injection: an experimental study in swine. Cardiovasc Intervent Radiol, 2010, 33(1):143–9., DOI 10.1007/s00270-009-9769-2 Planche O, Teriitehau C, Boudabous S, Robinson JM, Rao P, Deschamps F, et al.In vivo evaluation of lung microwave ablation in a porcine tumor mimic model. Cardiovasc Intervent Radiol, 2013, 36(1):221–8., DOI 10.1007/s00270- 012-0399-8 Hidalgo J, Belani J, Maxwell K, Lieber D, Talcott M, Baron P, et al.Development of exophytic tumor model for laparoscopic partial nephrectomy: technique and initial experience. Urology, 2005, 65(5):872–6., DOI 10.1016/j.urology.2004.12.002 Goldberg SN, Gazelle GS, Compton CC, Mueller PR, and McLoud TCRadiofrequency tissue ablation of VX2 tumor nodules in the rabbit lung. Acad Radiol, 1996, 3(11):929–35., DOI 10.1016/s1076-6332(96)80303-5 Miao Y, Ni Y, Bosmans H, Yu J, Vaninbroukx J, Dymarkowski S, et al.Radiofrequency ablation for eradication of pulmonary tumor in rabbits. J Surg Res, 2001, 99(2):265–71., DOI 10.1006/jsre.2001.6208 Gragnaniello C, Nader R, Doormaal Tv., Kamel M, Voormolen EHJ, Lasio G, et al.Skull base tumor model. Jns, 2010, 113(5):1106–11., DOI 10.3171/2010.3. JNS09513 Lee J-S, Tailor A-R, Lamki T, Zhang J, Irani S, and Ammirati MPetroclival tumor model-technical note and educational implications. Neurosurg Rev, 2016, 39(2):251–8. discussion 7–8., DOI 10.1007/s10143-015-0683-6 Zhang Y, GeH-W, Li N-C, Yu C-F, Guo H-F, Jin S-H, et al.Evaluation of threedimensional printing for laparoscopic partial nephrectomy of renal tumors: a preliminary report. World J Urol, 2016, 34(4):533–7., DOI 10.1007/s00345-015- 1530-7 Golab A, Smektala T, Kaczmarek K, Stamirowski R, Hrab M, and Slojewski MLaparoscopic partial nephrectomy supported by training involving personalized silicone replica poured in three-dimensional printed casting mold. J Laparoendoscopic Adv Surg Tech, 2017, 27(4):420–2., DOI 10.1089/ lap.2016.0596 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 630459 EP 630466 DI 10.3389/pore.2021.630459 PG 8 ER PT J AU Yang, A Li, M Fang, M AF Yang, Ai Li, Min Fang, Mingzhi TI The Research Progress of Direct KRAS G12C Mutation Inhibitors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE KRAS mutation; targeted drugs; oncogene; inhibitor; oncology ID KRAS mutation; targeted drugs; oncogene; inhibitor; oncology AB KRAS mutations have long been considered undruggable. However, a series of direct KRAS mutation inhibitors have been developed since the switch II pocket was discovered recently. This review will summarize progress in the development of direct KRAS G12C mutation inhibitors, current relevant drugs under study and challenges that need to be considered in future research. C1 [Yang, Ai] Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Department of OncologyNanjing, China. [Li, Min] Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Department of OncologyNanjing, China. [Fang, Mingzhi] Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Department of OncologyNanjing, China. RP Li, M (reprint author), Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Department of Oncology, Nanjing, China. EM doctorlimin@163.com CR Hobbs GA, Der CJ, and Rossman KL RAS isoforms and mutations in cancer at a glance. J Cel Sci, 2016, 129(7):1287–92., DOI 10.1242/jcs.182873 Ostrem JML, and Shokat KM Direct small-molecule inhibitors of KRAS: from structural insights to mechanism-based design. Nat Rev Drug Discov, 2016, 15(11):771–85., DOI 10.1038/nrd.2016.139 Prior IA, Lewis PD, and Mattos C A comprehensive survey of Ras mutations in cancer. Cancer Res, 2012, 72(10):2457–67., DOI 10.1158/0008-5472.can-11-2612 Lindsay CR, Jamal-Hanjani M, Forster M, and Blackhall F KRAS: reasons for optimism in lung cancer. Eur J Cancer, 2018, 99:20–7., DOI 10.1016/j.ejca.2018. .001 5. Lanman BA, Allen JR, Allen JG, Amegadzie AK, Ashton KS, Booker SK, et al. Discovery of a covalent inhibitor of KRASG12C, AMG 510, for the treatment of solid tumors. J Med Chem, 2020, 63(1):52–65., DOI 10.1021/acs.jmedchem. 9b01180 Becher I, Savitski MM, Savitski MF, Hopf C, Bantscheff M, and Drewes G Affinity profiling of the cellular kinome for the nucleotide cofactors ATP, ADP, and GTP. ACS Chem Biol, 2013, 8(3):599–607., DOI 10.1021/cb3005879 Burns MC, Howes JE, Sun Q, Little AJ, Camper DV, Abbott JR, et al. Highthroughput screening identifies small molecules that bind to the RAS:SOS:RAS complex and perturb RAS signaling. Anal Biochem, 2018, 548:44–52., DOI 10. 1016/j.ab.2018.01.025 Xu L-l., Li C-c., An L-y., Dai Z, Chen X-y., You Q-d., et al. Selective apoptosisinducing activity of synthetic hydrocarbon-stapled SOS1 helix with d-amino acids in H358 cancer cells expressing KRASG12C. Eur J Med Chem, 2020, 185: 111844., DOI 10.1016/j.ejmech.2019.111844 Leung ELH, Luo LX, Li Y, Liu ZQ, Li LL, Shi DF, et al. Identification of a new inhibitor of KRAS-PDEδ interaction targeting KRAS mutant nonsmall cell lung cancer. Int J Cancer, 2019, 145(5):1334–45., DOI 10.1002/ijc.32222 Ostrem JM, Peters U, Sos ML, Wells JA, and Shokat KM K-Ras(G12C, inhibitors allosterically control GTP affinity and effector interactions. Nature, 2013, 503(7477):548–51., DOI 10.1038/nature12796 Lim SM, Westover KD, Ficarro SB, Harrison RA, Choi HG, Pacold ME, et al. Therapeutic targeting of oncogenic K-Ras by a covalent catalytic site inhibitor. Angew Chem, 2014, 126(1):203–8., DOI 10.1002/ange. 201307387 Lito P, Solomon M, Li L-S, Hansen R, and Rosen N Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science, 2016, 351(6273):604–8., DOI 10.1126/science.aad6204 Patricelli MP, Janes MR, Li L-S, Hansen R, Peters U, Kessler LV, et al. Selective inhibition of oncogenic KRAS output with small molecules targeting the inactive state. Cancer Discov, 2016, 6(3):316., DOI 10.1158/2159-8290.cd-15- 1105 Shipman L Putting the brakes on KRAS-G12C nucleotide cycling. Nat Rev Drug Discov, 2016, 15(3):159., DOI 10.1038/nrd.2016.40 Janes MR, Zhang J, Li L-S, Hansen R, Peters U, Guo X, et al. Targeting KRAS mutant cancers with a covalent G12C-specific inhibitor. Cell, 2018, 172(3): 578–89., DOI 10.1016/j.cell.2018.01.006 Molina-Arcas M, Moore C, Rana S, van Maldegem F, Mugarza E, Romero- Clavijo P, et al. Development of combination therapies to maximize the impact of KRAS-G12C inhibitors in lung cancer. Sci Transl Med, 2019, 11(510): eaaw7999., DOI 10.1126/scitranslmed.aaw7999 Gentile DR, Rathinaswamy MK, Jenkins ML, Moss SM, Siempelkamp BD, Renslo AR, et al. Ras binder induces a modified switch-II pocket in GTP and GDP states. Cel Chem Biol, 2017, 24(12):1455–66., DOI 10.1016/j.chembiol.2017. 08.025 Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, et al. The clinical KRAS(G12C, inhibitor AMG 510 drives anti-tumour immunity. Nature, 2019, 575(7781):217–23., DOI 10.1038/s41586-019-1694-1 Herbst RS, and Schlessinger J Small molecule combats cancer-causing KRAS protein at last [J]. Nature 2019, 575, 7782), 294-295. Saiki AY, Gaida K, Rex K, Achanta P, Lipford JR, et al. Discovery and in vitro characterization of AMG 510–a potent and selective covalent small-molecule inhibitor of KRASG12C [J]. Cancer Res, 2017, 79(13 Suppl. ment):4484. Fakih M, O’Neil B, Price TJ, Falchook GS, Desai J, Kuo J, et al. Phase 1 study evaluating the safety, tolerability, pharmacokinetics, PK), and efficacy of AMG 510, a novel small molecule KRASG12C inhibitor, in advanced solid tumors. Jco, 2019, 37:3003., DOI 10.1200/jco.2019.37.15_suppl.3003 Govindan R, Fakih MG, Price TJ, Falchook GS, and Hong DS 446PD - phase I study of AMG 510, a novel molecule targeting KRAS G12C mutant solid tumours[J]. Ann Oncol, 2019, 30:163–4., DOI 10.1093/annonc/mdz244.008 Robert C, Karaszewska B, Schachter J, Rutkowski P, Mackiewicz A, Stroiakovski D, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med, 2015, 372(1):30–9., DOI 10.1056/nejmoa1412690 Selby MJ, Engelhardt JJ, Johnston RJ, Lu L-S, Han M, Thudium K, et al. Preclinical development of ipilimumab and nivolumab combination immunotherapy: mouse tumor models, in vitro functional studies, and cynomolgus macaque toxicology. Plos One, 2016, 11(9):e0161779., DOI 10. 1371/journal.pone.0161779 Mosely SIS, Prime JE, Sainson RCA, Koopmann J-O,Wang DYQ, Greenawalt DM, et al. Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol Res, 2017, 5(1): 29–41., DOI 10.1158/2326-6066.cir-16-0114 Hallin J, Engstrom LD, Hargis L, Calinisan A, Aranda R, Briere DM, et al. The KRASG12C inhibitor MRTX849 provides insight toward therapeutic susceptibility of KRAS-mutant cancers in mouse models and patients. Cancer Discov, 2020, 10(1):54–71., DOI 10.1158/2159-8290.cd-19-1167 Bar-Sagi D, Knelson EH, and Sequist LV. 2020. A bright future for KRAS inhibitors. Nature Cancer[J]. 1, 1). 25-27. Kessler D, Gmachl M, Mantoulidis A, Martin LJ, Zoephel A, Mayer M, et al. Drugging an undruggable pocket on KRAS. Proc Natl Acad Sci USA, 2019, 116(32):15823–9., DOI 10.1073/pnas.1904529116 Cruz-Migoni A, Canning P, Quevedo CE, Bataille CJR, Bery N, Miller A, et al. Structure-based development of new RAS-effector inhibitors from a combination of active and inactive RAS-binding compounds. Proc Natl Acad Sci USA, 2019, 116(7):2545–50., DOI 10.1073/pnas.1811360116 Quevedo CE, Cruz-Migoni A, Bery N, Ami M, Tomoyuki T, and Donna P Small molecule inhibitors of RAS-effector protein interactions derived using an intracellular antibody fragment[J]. Nat Commun, 2018, 9(1):3169., DOI 10. 1038/s41467-018-05707-2 Nagasaka M, Li Y, Sukari A, Ou S-HI, Al-Hallak MN, and Azmi AS KRAS G12C Game of Thrones, which direct KRAS inhibitor will claim the iron throne? Cancer Treat Rev, 2020, 84:101974., DOI 10.1016/j.ctrv.2020.101974 Xue JY, Zhao Y, Aronowitz J, Mai TT, Vides A, Qeriqi B, et al. Rapid nonuniform adaptation to conformation-specific KRAS(G12C, inhibition. Nature, 2020, 577(7790):421–5., DOI 10.1038/s41586-019-1884-x NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 631095 EP 631102 DI 10.3389/pore.2021.631095 PG 8 ER PT J AU Puskas, R Bikov, A Horvath, P Lazar, Zs Kunos, L Nagy, R Pinter, G Galffy, G AF Puskas, Rita Bikov, Andras Horvath, Peter Lazar, Zsofia Kunos, Laszlo Nagy, Reka Pinter, Gabriella Galffy, Gabriella TI Circulating Survivin Protein Levels in Lung Cancer Patients Treated With Platinum-Based Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE survivin; lung cancer; ELISA; disease progression; biomarker ID survivin; lung cancer; ELISA; disease progression; biomarker AB The survivin protein contributes to the development and progression of tumors. Protein expression and mRNA levels correlate with clinicopathological parameters and survival of cancer patients. Our purpose was to evaluate whether circulating survivin levels have any diagnostic or predictive value in lung cancer. 118 patients with advanced stage lung cancer participated in our study. 53 suffered from adenocarcinoma (ADC), 33 from squamous cell carcinoma (SqCC), and 32 from small cell lung cancer (SCLC). We also enrolled 21 control subjects. Blood samples were collected before and after two cycles of chemotherapy. We measured survivin concentrations with ELISA. Non-parametric tests were used for analysis. We did not find significant difference in survivin levels between patients and control subjects (17.19/0–829.74/vs. 49.13/0–165.92/pg/ml; p = 0.07). We found lower survivin concentrations in patients with SqCC (0/0–171.24/ pg/ml) than in those with ADC (24.94/0–626.46 pg/ml) and SCLC (45.51/0–829.74/pg/ ml) (ADC vs. SqCC p < 0.0001, ADC vs. SCLC p = 0.0405, SqCC vs. SCLC p < 0.0001). Survivin levels were higher in stage IV patients than in patients without distant metastases (p = 0.0061), and concentrations were progressively higher with increasing number of metastatic organ sites (p = 0.04). We observed a decrease in survivin levels in ADC patients after platinum plus pemetrexed chemotherapy (26.22/0–626.46/pg/ml before vs. 0/0–114.36/pg/ml after; p = 0.01). Neither progression-free nor overall survival correlated with survivin levels at baseline. Our data imply that survivin may be involved in the development of metastases and it might be used as a biomarker of disease progression. However, circulating survivin concentrations do not predict survival of patients with lung cancer. C1 [Puskas, Rita] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Bikov, Andras] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Horvath, Peter] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Lazar, Zsofia] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Kunos, Laszlo] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Nagy, Reka] Semmelweis UniversityBudapest, Hungary. [Pinter, Gabriella] Semmelweis UniversityBudapest, Hungary. [Galffy, Gabriella] Semmelweis University, Department of PulmonologyBudapest, Hungary. RP Puskas, R (reprint author), Semmelweis University, Department of Pulmonology, Budapest, Hungary. CR Ambrosini G, Adida C, and Altieri DC. A Novel Anti-apoptosis Gene, Survivin, Expressed in Cancer and Lymphoma. Nat Med, 1997, 3(8): 917–21., DOI 10.1038/nm0897-917 Altieri DC. Survivin - the Inconvenient IAP. Semin Cel Dev Biol, 2005, 39: 91–6., DOI 10.1016/j.semcdb.2014.12.007 Uren AG, Wong L, Pakusch M, Fowler KJ, Burrows FJ, Vaux DL, et al. Survivin and the Inner Centromere Protein INCENP Show Similar Cell-Cycle Localization and Gene Knockout Phenotype. Curr Biol, 2000, 10(21): 1319–28., DOI 10.1016/s0960-9822(00)00769-7 Mita AC, Mita MM, Nawrocki ST, and Giles FJ. Survivin: Key Regulator of Mitosis and Apoptosis and Novel Target for Cancer Therapeutics. Clin Cancer Res, 2008, 14(16):5000–5., DOI 10.1158/1078-0432.ccr-08-0746 Kanwar JR, Kamalapuram SK, and Kanwar RK. Targeting Survivin in Cancer: the Cell-Signalling Perspective. Drug Discov Today, 2011, 16(11-12):485–94., DOI 10.1016/j.drudis.2011.04.001 Tamm I, Wang Y, Sausville E, Scudiero DA, Vigna N, Oltersdorf T, et al. IAPfamily Protein Survivin Inhibits Caspase Activity and Apoptosis Induced by Fas, CD95), Bax, Caspases, and Anticancer Drugs. Cancer Res, 1998, 58(23):5315–20. Jaiswal PK, Goel A, and Mittal RD. Survivin: A Molecular Biomarker in Cancer. Indian J Med Res, 2015, 141(4):389–97., DOI 10.4103/0971-5916. 159250 McKenzie JA, Liu T, Goodson AG, and Grossman D. Survivin Enhances Motility of Melanoma Cells by Supporting Akt Activation and α5 Integrin Upregulation. Cancer Res, 2010, 70(20):7927–37., DOI 10.1158/0008-5472.can- 10-0194 Fernandez JG, Rodriguez DA, Valenzuela M, Calderon C, Urzua U, Munroe D, et al. Survivin Expression Promotes VEGF-Induced Tumor Angiogenesis via PI3K/Akt Enhanced β-catenin/Tcf-Lef Dependent Transcription. Mol Cancer, 2014, 9(13):209., DOI 10.1186/1476-4598-13-209 Duan L, Hu X, Jin Y, Liu R, and YouQ. Survivin Protein Expression Is Involved in the Progression of Non-small Cell Lung Cancer in Asians: a Meta-Analysis. BMC Cancer, 2016, 16:276., DOI 10.1186/s12885-016-2304-3 Krepela E, Dankova P, Moravcikova E, Krepelova A, Prochazka J, Cermak J, et al. Increased Expression of Inhibitor of Apoptosis Proteins, Survivin and XIAP, in Non-small Cell Lung Carcinoma. Int J Oncol, 2009, 35(6):1449–62., DOI 10.3892/ijo_00000464 Kapellos G, Polonifi K, Farmakis D, Spartalis E, Tomos P, Aessopos A, et al. Overexpression of Survivin Levels in Circulation and Tissue Samples of Lung Cancer Patients. Anticancer Res, 2013, 33(8):3475–80. Yie S-m., Lou B, Ye S-r., He X, Cao M, Xie K, et al. Clinical Significance of Detecting Survivin-Expressing Circulating Cancer Cells in Patients with Nonsmall Cell Lung Cancer. Lung Cancer, 2009, 63(2):284–90., DOI 10.1016/j. lungcan.2008.05.024 Hirano H, Maeda H, Yamaguchi T, Yokota S, Mori M, and Sakoda S. Survivin Expression in Lung Cancer: Association with Smoking, Histological Types and Pathological Stages. Oncol Lett, 2015, 10(3):1456–62., DOI 10.3892/ol.2015.3374 Porebska I, Kosacka M, Sobanska E, Wyrodek E, and Jankowska R. Comparative Expression of Apoptotic Markers in Lung Adenocarcinoma and Squamous Cell Carcinoma. Adv Exp Med Biol, 2015, 873:101–7., DOI 10.1007/5584_2015_121 Sun P-L, Jin Y, Kim H, Seo AN, Jheon S, Lee C-T, et al. Survivin Expression Is an Independent Poor Prognostic Marker in Lung Adenocarcinoma but Not in Squamous Cell Carcinoma. Virchows Arch, 2013, 463(3):427–36., DOI 10.1007/ s00428-013-1462-9 Tang X-P, Li J, Yu L-C, Chen Y-C, Shi S-B, Zhu L-R, et al. Clinical Significance of Survivin and VEGF mRNA Detection in the Cell Fraction of the Peripheral Blood in Non-small Cell Lung Cancer Patients before and after Surgery. Lung Cancer, 2013, 81(2):273–9., DOI 10.1016/j.lungcan. 2013.05.005 Du Y-J, Li J, Zhu W-F, Wu Y, Tang X-P, Wang Y, et al. Survivin mRNACirculating Tumor Cells Predict Treatment Efficacy of Chemotherapy and Survival for Advanced Non-small Cell Lung Cancer Patients. Tumor Biol, 2014, 35(5):4499–507., DOI 10.1007/s13277-013-1592-3 Fawzy A, Gaafar R, Kasem F, Ali SS, Elshafei M, and Eldeib M. Importance of Serum Levels of Angiopoietin-2 and Survivin Biomarkers in Non-small Cell Lung Cancer. J Egypt Natl Cancer Inst, 2012, 24(1):41–5., DOI 10.1016/j.jnci. 2011.12.006 Naumnik W, Nilklinska W, OssolinskaM, and Chyczewska E. Serum Levels of HMGB1, Survivin, and VEGF in Patients with Advanced Non-small Cell Lung Cancer during Chemotherapy. Folia Histochem Cytobiol, 2009, 47(4):703–9., DOI 10.2478/v10042-009-0024-0 Eberhardt WE, Mitchell A, Crowley J, Kondo H, Kim YT, Turrisi A, et al. The IASLC Lung Cancer Staging Project: Proposals for the Revision of the M Descriptors in the Forthcoming Eighth Edition of the TNM Classification of Lung Cancer. J Thorac Oncol, 2015, 10(11):1515–22., DOI 10.1097/JTO. 0000000000000673 Oh Y, Taylor S, Bekele BN, Debnam JM, Allen PK, Suki D, et al. Number of Metastatic Sites Is a Strong Predictor of Survival in Patients with Nonsmall Cell Lung Cancer with or without Brain Metastases. Cancer, 2009, 115(13):2930–8., DOI 10.1002/cncr.24333 Amini A, Li R, Liu A, Ray P, Maniyedath A, Huntzinger C, et al. Number of Metastatic Organ Sites and Survival by Molecular Profile in Lung Adenocarcinoma. Int J Radiat Oncology, 2019, 104(5):1191–2., DOI 10.1016/ j.ijrobp.2019.05.049 Wang J, Huang C, Wei X-y., Qi D-l., Gong L-q., Mu H-y., et al. Changes of Activated Circulating Endothelial Cells and Survivin in Patients with Nonsmall Cell Lung Cancer after Antiangiogenesis Therapy. Chin Med J, 2008, 121(22):2234–40., DOI 10.1097/00029330-200811020-00005 Derin D, Soydinc HO, Guney N, Tas F, Camlica H, Duranyildiz D, et al. Serum Levels of Apoptosis Biomarkers, Survivin and TNF-Alpha in Nonsmall Cell Lung Cancer. Lung Cancer, 2008, 59(2):240–5., DOI 10.1016/j.lungcan.2007.08.005 Zhang J, Li Y, Wang L, Zhang Y, Zhang Q, and Liu J. Folate Receptor Alpha Promotes Endometrial Carcinoma Cell Proliferation and Inhibits Apoptosis by Regulating the ERK Signaling Pathway. Int J Clin Exp Med, 2019, 12(7): 8791–8. Liang J, Lu T, Chen Z, Zhan C, and Wang Q. Mechanisms of Resistance to Pemetrexed in Non-small Cell Lung Cancer. Transl Lung Cancer Res, 2019, 8(6):1107–18., DOI 10.21037/tlcr.2019.10.14 Xiong Y-Q, Sun H-C, Zhu X-D, Zhang W, Zhuang P-Y, Zhang J-B, et al. Bevacizumab Enhances Chemosensitivity of Hepatocellular Carcinoma to Adriamycin Related to Inhibition of Survivin Expression. J Cancer Res Clin Oncol, 2011, 137(3):505–12., DOI 10.1007/s00432-010-0914-8 Chen P, Zhu J, Liu DY, Li HY, Xu N, and Hou M. Over-expression of Survivin and VEGF in Small-Cell Lung Cancer May Predict the Poorer Prognosis. Med Oncol, 2014, 31(1):775., DOI 10.1007/s12032-013-0775-5 Yano Y, Otsuka T, Hirano H, Uenami T, Satomi A, KuroyamaM, et al. Nuclear Survivin Expression in Small Cell Lung Cancer. Anticancer Res, 2015, 35(5): 2935–9. Dong H, Qian D, Wang Y, Meng L, Chen D, Ji X, et al. Survivin Expression and Serum Levels in Pancreatic Cancer. World J Surg Oncol, 2015, 13:189., DOI 10. 1186/s12957-015-0605-7 Guney N, Soydine HO, Derin D, Camlica H, Duranyildiz D, Yasasever V, et al. Serum and Urine Survivin Levels in Breast Cancer. Mo, 2006, 23(3):335–40., DOI 10.1385/mo:23:3:335 Tas F, Duranyildiz D, Argon A, Oguz H, Camlica H, Yasasever V, et al. Serum Bcl-2 and Survivin Levels in Melanoma. Melanoma Res, 2004, 14(6):543–6., DOI 10.1097/00008390-200412000-00017 Sohair AH, Naema Z, and Olfat H. Determination of MMP3 and Survivin as Non-invasive Circulating Markers in Patients of Urinary Bladder Cancer. Biohealth Sci Bull, 2009, 1(1):29–37. Khan S, Jutzy JMS, Aspe JR, McGregor DW, Neidigh JW, and Wall NR. Survivin Is Released from Cancer Cells via Exosomes. Apoptosis, 2011, 16(1): 1–12., DOI 10.1007/s10495-010-0534-4 Khan S, Aspe JR, Asumen MG, Almaguel F, Odumosu O, Acevedo-Martinez S, et al. Extracellular, Cell-Permeable Survivin Inhibits Apoptosis while Promoting Proliferative and Metastatic Potential. Br J Cancer, 2009, 100: 1073–86., DOI 10.1038/sj.bjc.6604978 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 631969 EP 631975 DI 10.3389/pore.2021.631969 PG 7 ER PT J AU Wang, X Liu, Y Leng, X Cao, K Sun, W Zhu, J Ma, J AF Wang, Xiaoyuan Liu, Yang Leng, Xue Cao, Kui Sun, Wentao Zhu, Jinhong Ma, Jianqun TI UBE2T Contributes to the Prognosis of Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE UBE2T; ESCC; prognosis; TCGA; immunohistochemistry ID UBE2T; ESCC; prognosis; TCGA; immunohistochemistry AB Background: The ubiquitin-conjugating enzyme E2 T (UBE2T) has been shown to contribute to several types of cancer. However, no publication has reported its implication in esophageal squamous cell cancer (ESCC). Methods: We explored several public databases, including The Cancer Genome Atlas (TCGA), Oncomine, and gene expression Omnibus (GEO). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and gene set enrichment analysis (GSEA) were adopted to explore involved signaling pathways. We used R software to develop prognostic gene signatures with the LASSO and stepwise Cox regression analysis, separately. Immunohistochemistry staining was performed to detect UBE2T in 90 ESCC patients, followed by survival analysis. We also used an R package pRRophetic to evaluate chemotherapy sensitivity for the TCGA–ESCC cohort. Results: We found significantly increased UBE2T transcript levels and DNA copy numbers in ESCC tissues. UBE2T was associated with the p53 signaling pathway, cell cycle, Fanconi anemia pathway, and DNA replication, as indicated by Go, KEGG pathway enrichment analysis. These pathways were also upregulated in ESCC. The prognostic signatures with UBE2T-associated genes could stratify ESCC patients into low- and highrisk groups with significantly different overall survival in the TCGA–ESCC cohort. We also validated the association of UBE2T with unfavorable survival in 90 ESCC patients recruited for this study. Moreover, we found that the low-risk group was significantly more sensitive to chemotherapy than the high-risk group. Conclusions: UBE2T is involved in the development of ESCC, and gene signatures derived from UBE2T-associated genes are predictive of prognosis in ESCC. C1 [Wang, Xiaoyuan] Harbin Medical University Cancer Hospital, Department of Thoracic SurgeryHarbin, China. [Liu, Yang] Harbin Medical University, Cancer Hospital, Department of Medical OncologyHarbin, China. [Leng, Xue] Harbin Medical University Cancer Hospital, Department of Thoracic SurgeryHarbin, China. [Cao, Kui] Harbin Medical University, Cancer Hospital, Department of Medical OncologyHarbin, China. [Sun, Wentao] Harbin Medical University Cancer Hospital, Department of RadiologyHarbin, China. [Zhu, Jinhong] Harbin Medical University Cancer Hospital, Department of Clinical LaboratoryHarbin, China. [Ma, Jianqun] Harbin Medical University Cancer Hospital, Department of Thoracic SurgeryHarbin, China. RP Zhu, J (reprint author), Harbin Medical University Cancer Hospital, Department of Clinical Laboratory, Harbin, China. EM jinhongzhu625@gmail.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J Clinicians, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA: A Cancer J Clinicians, 2016, 66(2):115–32., DOI 10.3322/caac. 21338 van Rossum PSN, Mohammad NH, Vleggaar FP, van Hillegersberg R. Treatment for unresectable or metastatic oesophageal cancer: current evidence and trends. Nat Rev Gastroenterol Hepatol, 2018, 15(4):235–49., DOI 10.1038/nrgastro.2017.162 Lagergren J, Smyth E, Cunningham D, Lagergren P. Oesophageal cancer. The Lancet, 2017, 390(10110):2383–96., DOI 10.1016/s0140-6736(17)31462-9 Alpi AF, Chaugule V, Walden H. Mechanism and disease association of E2- conjugating enzymes: lessons from UBE2T and UBE2L3. Biochem J, 2016, 473(20):3401–19., DOI 10.1042/BCJ20160028 Hoeller D, Hecker C-M, Dikic I. Ubiquitin and ubiquitin-like proteins in cancer pathogenesis. Nat Rev Cancer, 2006, 6(10):776–88., DOI 10.1038/nrc1994 Lim K-H, Song M-H, Baek K-H. Decision for cell fate: deubiquitinating enzymes in cell cycle checkpoint. Cell. Mol. Life Sci., 2016, 73(7):1439–55., DOI 10.1007/s00018-015-2129-2 Mattern M, Sutherland J, Kadimisetty K, Barrio R, Rodriguez MS. Using ubiquitin binders to decipher the ubiquitin code. Trends Biochem Sci, 2019, 44(7):599–615., DOI 10.1016/j.tibs.2019.01.011 Kwon YT, Ciechanover A. The ubiquitin code in the ubiquitin-proteasome system and autophagy. Trends Biochem Sci, 2017, 42(11):873–86., DOI 10.1016/j. tibs.2017.09.002 Longerich S, San Filippo J, Liu D, Sung P. FANCI binds branched DNA and is monoubiquitinated by UBE2T-FANCL. J Biol Chem, 2009, 284(35):23182–6., DOI 10.1074/jbc.C109.038075 Perez-Pena J, Corrales-Sanchez V, Amir E, Pandiella A, Ocana A. Ubiquitinconjugating enzyme E2T, UBE2T, and denticleless protein homolog, DTL, are linked to poor outcome in breast and lung cancers. Sci Rep, 2017, 7(1): 17530., DOI 10.1038/s41598-017-17836-7 Luo C, Yao Y, Yu Z, Zhou H, Guo L, Zhang J, et al. UBE2T knockdown inhibits gastric cancer progression. Oncotarget, 2017, 8(20):32639–54., DOI 10.18632/ oncotarget.15947 Liu L-p., Yang M, Peng Q-z., LiM-y., Zhang Y-s., Guo Y-h., et al. UBE2T promotes hepatocellular carcinoma cell growth via ubiquitination of p53. Biochem Biophys Res Commun, 2017, 493(1):20–7., DOI 10.1016/j.bbrc.2017.09.091 Hu W, Xiao L, Cao C, Hua S, Wu D. UBE2T promotes nasopharyngeal carcinoma cell proliferation, invasion, and metastasis by activating the AKT/ GSK3β/β-catenin pathway. Oncotarget, 2016, 7(12):15161–72., DOI 10.18632/ oncotarget.7805 Wang Y, Leng H, Chen H, Wang L, Jiang N, Huo X, et al. Knockdown of UBE2T inhibits osteosarcoma cell proliferation, migration, and invasion by suppressing the PI3K/akt signaling pathway. Oncol Res, 2016, 24(5):361–9., DOI 10.3727/096504016X14685034103310 Wen M, Kwon Y, Wang Y, Mao J-H, Wei G. Elevated expression of UBE2T exhibits oncogenic properties in human prostate cancer. Oncotarget, 2015, 6(28):25226–39., DOI 10.18632/oncotarget.4712 Su H, Hu N, Yang HH, Wang C, Takikita M, Wang Q-H, et al. Global gene expression profiling and validation in esophageal squamous cell carcinoma and its association with clinical phenotypes. Clin Cancer Res, 2011, 17(9): 2955–66., DOI 10.1158/1078-0432.ccr-10-2724 Liu Y, Wu L, Ao H, Zhao M, Leng X, Liu M, et al. Prognostic implications of autophagy-associated gene signatures in non-small cell lung cancer. Aging, 2019, 11(23):11440–62., DOI 10.18632/aging.102544 Zhu J, Liu Y, Ao H, Liu M, Zhao M, Ma J. Comprehensive analysis of the immune implication of ACK1 gene in non-small cell lung cancer. Front Oncol, 2020, 10:1132., DOI 10.3389/fonc.2020.01132 Geeleher P, Cox N, Huang RS. pRRophetic: an R package for prediction of clinical chemotherapeutic response from tumor gene expression levels. PLoS One, 2014, 9(9):e107468., DOI 10.1371/journal.pone.0107468 Geeleher P, Cox NJ, Huang R. Clinical drug response can be predicted using baseline gene expression levels and in vitro drug sensitivity in cell lines. Genome Biol, 2014, 15(3):R47., DOI 10.1186/gb-2014-15-3-r47 Hu N, Wang C, Ng D, Clifford R, Yang HH, Tang Z-Z, et al. Genomic characterization of esophageal squamous cell carcinoma from a high-risk population in China. Cancer Res, 2009, 69(14):5908–17., DOI 10.1158/0008- 5472.Can-08-4622 Hao Y, Triadafilopoulos G, Sahbaie P, Young HS, Omary MB, Lowe AW. Gene expression profiling reveals stromal genes expressed in common between Barrett’s esophagus and adenocarcinoma. Gastroenterology, 2006, 131(3): 925–33., DOI 10.1053/j.gastro.2006.04.026 Kim SM, Park Y-Y, Park ES, Cho JY, Izzo JG, Zhang D, et al. Prognostic biomarkers for esophageal adenocarcinoma identified by analysis of tumor transcriptome. PloS One, 2010, 5(11):e15074., DOI 10.1371/journal.pone. 0015074 Ueki T, Park J-H, Nishidate T, Kijima K, Hirata K, Nakamura Y, et al. Ubiquitination and downregulation of BRCA1 by ubiquitin-conjugating enzyme E2T overexpression in human breast cancer cells. Cancer Res, 2009, 69(22):8752–60., DOI 10.1158/0008-5472.CAN-09-1809 Ramaekers CHMA, van den Beucken T, Meng A, Kassam S, Thoms J, Bristow RG, et al. Hypoxia disrupts the Fanconi anemia pathway and sensitizes cells to chemotherapy through regulation of UBE2T. Radiother Oncol, 2011, 101(1): 190–7., DOI 10.1016/j.radonc.2011.05.059 Kelsall IR, Langenick J, MacKay C, Patel KJ, Alpi AF. The Fanconi anaemia components UBE2T and FANCM are functionally linked to nucleotide excision repair. PloS One, 2012, 7(5):e36970., DOI 10.1371/journal.pone. 0036970 Hao J, Xu A, Xie X, Hao J, Tian T, Gao S, et al. Elevated expression of UBE2T in lung cancer tumors and cell lines. Tumor Biol, 2008, 29(3):195–203., DOI 10. 1159/000148187 Wang Q, Wang F, Lv J, Xin J, Xie L, ZhuW, et al. Interactive online consensus survival tool for esophageal squamous cell carcinoma prognosis analysis. Oncol Lett, 2019, 18(2):1199–206., DOI 10.3892/ol.2019.10440 Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell, 2011, 144(5):646–74., DOI 10.1016/j.cell.2011.02.013 Zhang W, Zhang Y, Yang Z, Liu X, Yang P, Wang J, et al. High expression of UBE2T predicts poor prognosis and survival in multiple myeloma. Cancer Gene Ther, 2019, 26(11–12):347–55., DOI 10.1038/s41417-018-0070-x Gong YQ, Peng D, Ning XH, Yang XY, Li XS, Zhou LQ, et al. UBE2T silencing suppresses proliferation and induces cell cycle arrest and apoptosis in bladder cancer cells. Oncol Lett, 2016, 12(6):4485–92., DOI 10.3892/ol.2016.5237 Morreale FE, Testa A, Chaugule VK, Bortoluzzi A, Ciulli A, Walden H. Mind the metal: a fragment library-derived zinc impurity binds the E2 ubiquitin-conjugating enzyme Ube2T and induces structural rearrangements. J Med Chem, 2017, 60(19):8183–91., DOI 10.1021/acs. jmedchem.7b01071 Machida YJ,Machida Y, Chen Y, Gurtan AM, Kupfer GM, D’Andrea AD, et al. UBE2T is the E2 in the Fanconi anemia pathway and undergoes negative autoregulation. Mol Cel, 2006, 23(4):589–96., DOI 10.1016/j. molcel.2006.06.024 Tian X, Zhu X, Yan T, Yu C, Shen C, Hu Y, et al. Recurrence-associated gene signature optimizes recurrence-free survival prediction of colorectal cancer. Mol Oncol, 2017, 11(11):1544–60., DOI 10.1002/1878-0261.12117 Zhang J-X, Song W, Chen Z-H, Wei J-H, Liao Y-J, Lei J, et al. Prognostic and predictive value of amicroRNA signature in stage II colon cancer: amicroRNA expression analysis. Lancet Oncol, 2013, 14(13):1295–306., DOI 10.1016/s1470- 2045(13)70491-1 Chai R-C, Wu F, Wang Q-X, Zhang S, Zhang K-N, Liu Y-Q, et al. m6A RNA methylation regulators contribute to malignant progression and have clinical prognostic impact in gliomasA RNA methylation regulators contribute to malignant progression and have clinical prognostic impact in gliomas. Aging, 2019, 11(4):1204–25., DOI 10.18632/aging.101829 Liu Y, Guo X, Zhao M, Ao H, Leng X, Liu M, et al. Contributions and prognostic values of m 6 A RNA methylation regulators in non-small-cell lung cancer. J Cel Physiol, 2020, 235(9):6043–57., DOI 10.1002/jcp.29531 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 632531 EP 632544 DI 10.3389/pore.2021.632531 PG 14 ER PT J AU Uchida, Sh Kojima, T Sugino, T AF Uchida, Shiro Kojima, Takaaki Sugino, Takashi TI Clinicopathological Features, Tumor Mutational Burden, and Tumour-Infiltrating Lymphocyte Interplay in ERBB2-Mutated Breast Cancer: In Silico Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Breast cancer; ErbB2 mutation; tumor mutational burden; tumour-infiltrating lymphocytes; Bioinformactics analysis ID Breast cancer; ErbB2 mutation; tumor mutational burden; tumour-infiltrating lymphocytes; Bioinformactics analysis AB Recent evidence suggests that somatic mutations in ERBB2 activate ERBB2 signaling. These mutations occur at a frequency of approximately 3% in breast cancer (BC). ERBB2 mutations indicate poor prognosis as they are associated with recurrence and metastasis. This study aimed to evaluate the clinicopathological features, immune infiltration levels, tumor mutational burden (TMB), and tumor-infiltrating lymphocytes (TILs) in ERBB2-mutated breast cancer (ERBB2-mutated BC) using a bioinformatic approach and publicly available datasets (i.e., TCGA-BRCA and TIMER2.0). ERBB2-mutated BCs were associated with a high histological grade. ERBB2-mutated BCs comprised invasive breast carcinoma of no special type (21/35, 60%), classic invasive lobular carcinoma (12/35, 34.3%), and pleomorphic invasive lobular carcinoma (2/35, 5.7%). A Kaplan-Meier survival curve demonstrated that ERBB2-mutated BC was associated with a significantly worse prognosis compared to ERBB2 non-mutated BC (p < 0.01). Furthermore, 40% (14/35) of the patients with ERBB2-mutated BC harbored CDH1 mutations. Mutations at L755 and V777 accounted for 30.5% of these mutations in ERBB2-mutated BC, suggesting that these sites are mutational hot spots in BC, particularly in invasive lobular carcinoma. Of the ERBB2-mutated BCs, 8.6% were classified as TIL-high, whereas 77.1% were TILs-low; TMB significantly correlated with TILs (p < 0.05). CD8+ T cell infiltration levels were significantly higher in ERBB2 nonmutated BC. Among ERBB2-mutated BCs, 22.9% were classified as TMB-high, which was significantly higher than the rate in the ERBB2 non-mutated BC (p < 0.01). These findings provide evidence for a link between ERBB2 mutations and high TMB in BC. C1 [Uchida, Shiro] Kikuna Memorial Hospital, Division of Diagnostic PathologyYokohama, Japan. [Kojima, Takaaki] Nagoya University, Graduate School of Bioagricultural SciencesNagoya, Japan. [Sugino, Takashi] Shizuoka Cancer Center, Division of PathologyShizuoka, Japan. RP Uchida, Sh (reprint author), Kikuna Memorial Hospital, Division of Diagnostic Pathology, Yokohama, Japan. EM Dr.Uchida@gmail.com CR Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC, et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov, 2013, 3:224–37., DOI 10.1158/2159-8290.cd-12- 0349 Zabransky DJ, Yankaskas CL, Cochran RL, Wong HY, Croessmann S, Chu D, et al. HER2 missense mutations have distinct effects on oncogenic signaling and migration. Proc Natl Acad Sci USA, 2015, 112:E6205–E6214., DOI 10.1073/pnas. 1516853112 Petrelli F, Tomasello G, Barni S, Lonati V, Passalacqua R, Ghidini M. Clinical and pathological characterization of HER2 mutations in human breast cancer: a systematic review of the literature. Breast Cancer Res Treat, 2017, 166:339–49., DOI 10.1007/s10549-017-4419-x Ma CX, Bose R, Gao F, Freedman RA, Telli ML, Kimmick G, et al. Neratinib efficacy and circulating tumor DNA detection of HER2 mutations in HER2 nonamplified metastatic breast cancer. Clin Cancer Res, 2017, 23:5687–95., DOI 10.1158/1078-0432.ccr-17-0900 Berx G, Cleton-Jansen AM, Nollet F, De Leeuw WJ, Van de Vijver M, Cornelisse C, et al. E-cadherin is a tumour/invasion suppressor gene mutated in human lobular breast cancers. EMBO J, 1995, 14:6107–15., DOI 10.1002/j.1460-2075. 1995.tb00301.x Takeichi M. Cadherin cell adhesion receptors as a morphogenetic regulator. Science, 1991, 251:1451–5., DOI 10.1126/science.2006419 Ross JS, Wang K, Sheehan CE, Boguniewicz AB, Otto G, Downing SR, et al. Relapsed classic E-cadherin, CDH1)-mutated invasive lobular breast cancer shows a high frequency of HER2, ERBB2, gene mutations. Clin Cancer Res, 2013, 19:2668–76., DOI 10.1158/1078-0432.ccr-13-0295 Wang T, Xu Y, Sheng S, Yuan H, Ouyang T, Li J, et al. HER2somatic mutations are associated with poor survival in HER2-negative breast cancers. Cancer Sci, 2017, 108:671–7., DOI 10.1111/cas.13182 Ping Z, Siegal GP, Harada S, Eltoum I-E, Youssef M, Shen T, et al. ERBB2 mutation is associated with a worse prognosis in patients with CDH1 altered invasive lobular cancer of the breast. Oncotarget, 2016, 7:80655–63., DOI 10. 18632/oncotarget.13019 Christgen M, Bartels S, Luft A, Persing S, Henkel D, Lehmann U, et al. Activating human epidermal growth factor receptor 2, HER2, gene mutation in bone metastases from breast cancer. Virchows Arch, 2018, 473:577–82., DOI 10.1007/s00428-018-2414-1 Lien H-C, Chen Y-L, Juang Y-L, Jeng Y-M Frequent alterations of HER2 through mutation, amplification, or overexpression in pleomorphic lobular carcinoma of the breast. Breast Cancer Res Treat, 2015, 150:447–55., DOI 10. 1007/s10549-015-3336-0 Christgen M, Bartels S, Radner M, Raap M, Rieger L, Christgen H, et al. ERBB2 mutation frequency in lobular breast cancer with pleomorphic histology or high-risk characteristics by molecular expression profiling. Genes Chromosomes Cancer, 2019, 58:175–85., DOI 10.1002/gcc.22716 Deniziaut G, Tille JC, Bidard F-C, Vacher S, Schnitzler A, Chemlali W, et al. ERBB2 mutations associated with solid variant of high-grade invasive lobular breast carcinomas. Oncotarget, 2016, 7:73337–46., DOI 10.18632/oncotarget. 11819 Ding Q, Chen H, Lim B, Damodaran S, Chen W, Tripathy D, et al. HER2 somatic mutation analysis in breast cancer: correlation with clinicopathological features. Hum Pathol, 2019, 92:32–8., DOI 10.1016/j. humpath.2019.07.006 WHO Classification of Tumours Editorial Board. WHO classification of tumours. In: Breast tumours. 5th ed. Lyon: IARC, 2019). Elston CW, Ellis IO. Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology, 1991, 19:403–10., DOI 10.1111/j.1365- 2559.1991.tb00229.x Weidner N, Semple JP. Pleomorphic variant of invasive lobular carcinoma of the breast. Hum Pathol, 1992, 23:1167–71., DOI 10.1016/0046-8177(92, 90035-2 Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, et al. The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol, 2015, 26:259–71., DOI 10.1093/annonc/mdu450 Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, et al. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucl. Acids Res, 2020, 48:W509–W514., DOI 10.1093/nar/gkaa407 Schumacher TN, Kesmir C, van Buuren MM Biomarkers in cancer immunotherapy. Cancer Cell, 2015, 27:12–4., DOI 10.1016/j.ccell.2014. 12.004 Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med, 2017, 9:34., DOI 10.1186/s13073-017-0424-2 Barroso-Sousa R, Jain E, Cohen O, Kim D, Buendia-Buendia J, Winer E, et al. Prevalence and mutational determinants of high tumor mutation burden in breast cancer. Ann Oncol, 2020, 31:387–94., DOI 10.1016/j. annonc.2019.11.010 Ross JS, Wang K, Gay LM, Al-Rohil RN, Nazeer T, Sheehan CE, et al. A high frequency of activating extracellular domain ERBB2, HER2, mutation in micropapillary urothelial carcinoma. Clin Cancer Res, 2014, 20:68–75., DOI 10.1158/1078-0432.ccr-13-1992 Ross JS, Gay LM, Wang K, Ali SM, Chumsri S, Elvin JA, et al. NonamplificationERBB2genomic alterations in 5605 cases of recurrent and metastatic breast cancer: an emerging opportunity for anti-HER2 targeted therapies. Cancer, 2016, 122:2654–62., DOI 10.1002/cncr.30102 Kurozumi S, Alsaleem M, Monteiro CJ, Bhardwaj K, Joosten SEP, Fujii T, et al. Targetable ERBB2 mutation status is an independent marker of adverse prognosis in estrogen receptor positive, ERBB2 non-amplified primary lobular breast carcinoma: a retrospective in silico analysis of public datasets. Breast Cancer Res, 2020, 22:85., DOI 10.1186/s13058-020- 01324-4 Ben-Baruch NE, Bose R, Kavuri SM, Ma CX, Ellis MJ. HER2-Mutated breast cancer responds to treatment with single-agent neratinib, a second-generation HER2/EGFR tyrosine kinase inhibitor. J Natl Compr Canc Netw, 2015, 13: 1061–4., DOI 10.6004/jnccn.2015.0131 Hanker AB, Brewer MR, Sheehan JH, Koch JP, Sliwoski GR, Nagy R, et al. An acquired HER2T798I gatekeeper mutation induces resistance to neratinib in a patient with HER2 mutant-driven breast cancer. Cancer Discov, 2017, 7: 575–85., DOI 10.1158/2159-8290.cd-16-1431 Westover D, Zugazagoitia J, Cho BC, Lovly CM, Paz-Ares L. Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors. Ann Oncol, 2018, 29:i10–i19., DOI 10.1093/annonc/ mdx703 Wu Y, Xu J, Du C, Wu Y, Xia D, Lv W, et al. The predictive value of tumor mutation burden on efficacy of immune checkpoint inhibitors in cancers: a systematic review and meta-analysis. Front Oncol, 2019, 9:1161., DOI 10.3389/ fonc.2019.01161 Samstein RM, Lee C-H, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet, 2019, 51:202–6., DOI 10.1038/s41588-018- 2-8 31. FDA. FDA approves pembrolizumab for adults and children with TMB-H solid tumors, 2020). http://www.fda.gov/drugs/drug-approvals-and-databases/fdaapproves- pembrolizumab-adults-and-children-tmb-h-solid-tumors., DOI 10. 31525/fda1-ucm626720.htm Gonzalez-Ericsson PI, Stovgaard ES, Sua LF, Reisenbichler E, Kos Z, Carter JM, et al. The path to a better biomarker: application of a risk management framework for the implementation of PD-L1 and TILs as immuno-oncology biomarkers in breast cancer clinical trials and daily practice. J Pathol, 2020, 250:667–84., DOI 10.1002/path.5406 Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol, 2018, 19:40–50., DOI 10. 1016/s1470-2045(17)30904-x Stanton SE, Adams S, Disis ML. Variation in the incidence and magnitude of tumor-infiltrating lymphocytes in breast cancer subtypes. JAMA Oncol, 2016, 2:1354–60., DOI 10.1001/jamaoncol.2016.1061 Merino DM, McShane L, Butler M, Funari VA, Hellmann MD, Chaudhary R, et al. TMB standardization by alignment to reference standards: phase II of the friends of cancer research TMB harmonization project. J Clin Oncol, 2019, 37: 2631., DOI 10.1200/jco.2019.37.15_suppl.2624 Jain MS, Massoud TF. Predicting tumour mutational burden from histopathological images using multiscale deep learning. Nat Mach Intell, 2020, 2:356–62., DOI 10.1038/s42256-020-0190-5 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 633243 EP 633251 DI 10.3389/pore.2021.633243 PG 9 ER PT J AU Zhang, Y Qin, Y Xu, H Yao, Q Gao, Y Feng, Y Ren, J AF Zhang, Yan Qin, Yaping Xu, Hongen Yao, Qihui Gao, Yalan Feng, Yushu Ren, Jingli TI Case Report: A Case Report of a Histological Transformation of ALK-Rearranged Adenocarcinoma With High Expression of PD-L1 to Squamous Cell Carcinoma After Treatment With Alectinib SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE ALK-Rearranged adenocarcinoma; PD-L1; squamous cell carcinoma; alectinib; histological transformation ID ALK-Rearranged adenocarcinoma; PD-L1; squamous cell carcinoma; alectinib; histological transformation AB We report an anaplastic lymphoma kinase (ALK)-positive patient shows a poor response to the ALK inhibitor alectinib due to the high expression of programmed death-ligand 1 (PDL1). After treatment with alectinib, the pathological form changed from adenocarcinoma into squamous cell carcinoma without novel genetic changes. This case may reveal a direct relationship between ALK mutation and a high level of PD-L1 expression. C1 [Zhang, Yan] The Second Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. [Qin, Yaping] The Second Affiliated Hospital of Zhengzhou University, Department of Precision Medicine CenterZhengzhou, China. [Xu, Hongen] The Second Affiliated Hospital of Zhengzhou University, Department of Precision Medicine CenterZhengzhou, China. [Yao, Qihui] The Second Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. [Gao, Yalan] The Second Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. [Feng, Yushu] The Second Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. [Ren, Jingli] The Second Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. RP Ren, J (reprint author), The Second Affiliated Hospital of Zhengzhou University, Department of Pathology, Zhengzhou, China. EM jingliren123002@126.com CR Kwak LE, Shaw AT, Ou S-HI, Bang YJ, Janne PA, Haber DA, et al. Anaplastic lymphoma kinase inhibition in non–small-cell lung cancer. New Engl J Med, 2011). 364(6):588., DOI 10.1056/NEJMoa1006448 Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim D-W, et al. Alectinib versus crizotinib in untreated ALK-positive non-small-cell lung cancer. N Engl J Med, 2017). 377(9):829–38., DOI 10.1056/ nejmoa1704795 Yang CY, Liao WY, Ho CC, Chen KY, Tsai TH, Hsu CL, et al. Association of programmed death-ligand 1 expression with fusion variants and clinical outcomes in patients with anaplastic lymphoma kinase-positive lung adenocarcinoma receiving crizotinib. Oncologist, 2020). 25(8):702–11., DOI 10. 1634/theoncologist.2020-0088 Muller IB, de Langen AJ, Giovannetti E, Peters GJ. Anaplastic lymphoma kinase inhibition in metastatic non-small cell lung cancer: clinical impact of alectinib. Onco Targets Ther, 2017). 10:4535–41., DOI 10.2147/ott.s109493 Doebele RC, Pilling AB, Aisner DL, Kutateladze TG, Le AT, Weickhardt AJ, et al. Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin Cancer Res, 2012). 18(5): 1472–82., DOI 10.1158/1078-0432.ccr-11-2906 Lovly CM, Mcdonald NT, Chen H, Ortiz-Cuaran S, Heukamp LC, Yan Y, et al. Rationale for co-targeting IGF-1R and ALK in ALK fusion-positive lung cancer. Nat Med, 2014). 20(9):1027–34., DOI 10.1038/nm.3667 Crystal AS, Shaw AT, Sequist LV, Friboulet L, Niederst MJ, Lockerman EL, et al. Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science, 2014). 346(6216):1480–6., DOI 10.1126/ science.1254721 Hrustanovic G, Olivas V, Pazarentzos E, Tulpule A, Asthana S, Blakely CM, et al. RAS-MAPK dependence underlies a rational polytherapy strategy in EML4-ALK-positive lung cancer. Nat Med, 2015). 21(9):1038–47., DOI 10.1038/ nm.3930 Fujita S, Masago K, Katakami N, Yatabe Y. Transformation to SCLC after treatment with the ALK inhibitor alectinib. J Thorac Oncol, 2016). 11(6): e67–e72., DOI 10.1016/j.jtho.2015.12.105 Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res, 2013). 19(8):2240–7., DOI 10.1158/1078-0432.ccr-12-2246 Sequist LV, Waltman BA, Dias-Santagata D, Digumaethy S, Turke AB, Fidias P, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Translational Med, 2011). 3(75):26r–75r., DOI 10.1126/scitranslmed.3002003 Mamesaya N, Nakashima K, Naito T, Nakajima T, Endo M, Takahashi T. ALK-rearranged lung squamous cell carcinoma responding to alectinib: a case report and review of the literature. BMC Cancer, 2017). 17(1):471., DOI 10.1186/ s12885-017-3468-1 Sagawa R, Ohba T, Ito E, Isogai S. ALK-positive squamous cell carcinoma dramatically responded to alectinib. Case Rep Oncol Med, 2018). 2018:1–3., DOI 10.1155/2018/4172721 Hsu K-H, Huang Y-H, Tseng J-S, Chen K-C, Ku W-H, Su K-Y, et al. High PD-L1 expression correlates with primary resistance to EGFRTKIs in treatment naive advanced EGFR-mutant lung adenocarcinoma patients. Lung Cancer, 2019). 127:37–43., DOI 10.1016/j.lungcan.2018. 11.021 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 637745 EP 637750 DI 10.3389/pore.2021.637745 PG 6 ER PT J AU Safi, M Trapani, D Alradhi, M Shan, X Jiwei, L AF Safi, Mohammed Trapani, Dario Alradhi, Mohammed Shan, Xiu Jiwei, Liu TI No Overall Survival Difference in the Immunotherapy Era for Rare Subtypes of Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE melanoma; SEER database; immunotherapy; survival; epidimiology ID melanoma; SEER database; immunotherapy; survival; epidimiology C1 [Safi, Mohammed] The First Affiliated Hospital of Dalian Medical University, Department of OncologyDalian, China. [Trapani, Dario] European Institute of OncologyMilan, Italy. [Alradhi, Mohammed] Dalian Medical University, The Second HospitalDalian, China. [Shan, Xiu] The First Affiliated Hospital of Dalian Medical University, Department of OncologyDalian, China. [Jiwei, Liu] The First Affiliated Hospital of Dalian Medical University, Department of OncologyDalian, China. RP Safi, M (reprint author), The First Affiliated Hospital of Dalian Medical University, Department of Oncology, Dalian, China. EM Mhosafi86@gmail.com CR Uprety D, Bista A, Chennamadhavuni A, Niroula A, Jafri SIM, Smith A, et al. Survival trends among patients with metastatic melanoma in the pretargeted and the post-targeted era: a US population-based study. Melanoma Res, 2018, 28(1):56–60., DOI 10.1097/cmr.0000000000000394 RodriguesM, de Koning L, Coupland S, Jochemsen A,Marais R, SternM-H, et al. So close, yet so far: discrepancies between uveal and othermelanomas. A position paper from um cure 2020. Cancers, 2019, 11(7):1032., DOI 10.3390/cancers11071032 Alicea GM, Rebecca VW. Emerging strategies to treat rare and intractable subtypes of melanoma. Pigment Cel Melanoma Res., 2021, 34(1):44–58., DOI 10.1111/pcmr.12880 Rapisuwon S, Qin Y, Roszik J, Carapeto F, Patel S, Carvajal RD. Systemic therapy for mucosal, acral, and uveal melanoma. Cutan Melanoma, 2020, 1301–35., DOI 10.1007/978-3-030-05070-2_62 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 639004 EP 639005 DI 10.3389/pore.2021.639004 PG 2 ER PT J AU Krokker, L Szabo, B Nemeth, K Tohati, R Sarkadi, B Meszaros, K Patocs, A Butz, H AF Krokker, Lilla Szabo, Borbala Nemeth, Kinga Tohati, Rebeka Sarkadi, Balazs Meszaros, Katalin Patocs, Attila Butz, Henriett TI Three Dimensional Cell Culturing for Modeling Adrenal and Pituitary Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE 3D cell culture; adrenal tumor; adrenocortical carcinoma; pheochromocytoma; pitNET; pituitary adenoma ID 3D cell culture; adrenal tumor; adrenocortical carcinoma; pheochromocytoma; pitNET; pituitary adenoma AB In vitro monolayer conditions are not able to reproduce the complexity of solid tumors, still, there is scarce information about the 3D cell culture models of endocrine tumor types. Therefore, our aim was to develop in vitro 3D tumor models by different methodologies for adrenocortical carcinoma (H295R), pituitary neuroendocrine tumor (RC-4B/C and GH3) and pheochromocytoma (PC-12). Various methodologies were tested. Cell biological assays (cell viability, proliferation and live cell ratio) and steroid hormone production by HPLC-MS/MS method were applied to monitor cellular well-being. Cells in hanging drops and embedded in matrigel formed multicellular aggregates but they were difficult to handle and propagate for further experiments. The most widely used methods: ultra-low attachment plate (ULA) and spheroid inducing media (SFDM) were not the most viable 3D model of RC-4B/C and GH3 cells that would be suitable for further experiments. Combining spheroid generation with matrigel scaffold H295R 3D models were viable for 7 days, RC-4B/C and GH3 3D models for 7–10 days. ULA and SFDM 3D models of PC-12 cells could be used for further experiments up to 4 days. Higher steroid production in 3D models compared to conventional monolayer culture was detected. Endocrine tumor cells require extracellular matrix as scaffold for viable 3D models that can be one reason behind the lack of the usage of endocrine 3D cultures. Our models help understanding the pathogenesis of endocrine tumors and revealing potential biomarkers and therapeutic targets. They could also serve as an excellent platform for preclinical drug test screening. C1 [Krokker, Lilla] Semmelweis University, Department of Laboratory MedicineBudapest, Hungary. [Szabo, Borbala] Semmelweis University, Department of Laboratory MedicineBudapest, Hungary. [Nemeth, Kinga] Hungarian Academy of Sciences and Semmelweis University, Hereditary Endocrine Tumors Research GroupBudapest, Hungary. [Tohati, Rebeka] Semmelweis University, Department of Laboratory MedicineBudapest, Hungary. [Sarkadi, Balazs] Hungarian Academy of Sciences and Semmelweis University, Hereditary Endocrine Tumors Research GroupBudapest, Hungary. [Meszaros, Katalin] Hungarian Academy of Sciences and Semmelweis University, Hereditary Endocrine Tumors Research GroupBudapest, Hungary. [Patocs, Attila] Semmelweis University, Department of Laboratory MedicineBudapest, Hungary. [Butz, Henriett] Semmelweis University, Department of Laboratory MedicineBudapest, Hungary. RP Butz, H (reprint author), Semmelweis University, Department of Laboratory Medicine, Budapest, Hungary. EM butz.henriett@med.semmelweis-univ. hu CR Ravi M, Ramesh A, and Pattabhi A. Contributions of 3D Cell Cultures for Cancer Research. J Cel Physiol, 2017, 232:2679–97., DOI 10.1002/jcp.25664 Costa EC, Moreira AF, de Melo-Diogo D, Gaspar VM, Carvalho MP, and Correia IJ. 3D Tumor Spheroids: an Overview on the Tools and Techniques Used for Their Analysis. Biotechnol Adv, 2016, 34:1427–41., DOI 10.1016/j. biotechadv.2016.11.002 Lv D, Hu Z, Lu L, Lu H, and Xu X. Three-dimensional Cell Culture: A Powerful Tool in Tumor Research and Drug Discovery. Oncol Lett, 2017, 14:6999–7010., DOI 10.3892/ol.2017.7134 Jo Y, Choi N, Kim K, Koo H-J, Choi J, and Kim HN. Chemoresistance of Cancer Cells: Requirements of TumorMicroenvironment-Mimicking In Vitro Models in Anti-cancer Drug Development. Theranostics, 2018, 8:5259–75., DOI 10.7150/thno.29098 Nietzer S, Baur F, Sieber S, Hansmann J, Schwarz T, Stoffer C, et al. Mimicking Metastases Including Tumor Stroma: A New Technique to Generate a Three- Dimensional Colorectal Cancer Model Based on a Biological Decellularized Intestinal Scaffold. Tissue Eng C: Methods, 2016, 22:621–35., DOI 10.1089/ten. tec.2015.0557 Hallanger-Johnson JE. Systemic Therapy for Adrenocortical Carcinoma: a Review. AME Med J, 2020, 5:5., DOI 10.21037/amj.2020.01.07 Chan DL, and Singh S. Current Chemotherapy Use in Neuroendocrine Tumors. Endocrinol Metab Clin North America, 2018, 47:603–14., DOI 10. 1016/j.ecl.2018.04.006 Otoukesh S, Cooper CJ, Lou W, Mojtahedzadeh M, Nasrazadani A, Wampler M, et al. Combination Chemotherapy Regimen in a Patient with Metastatic Malignant Pheochromocytoma and Neurofibromatosis Type 1. Am J Case Rep, 2014, 15:123–7., DOI 10.12659/AJCR.890181 Sarkadi B, Meszaros K, Krencz I, Canu L, Krokker L, Zakarias S, et al. Glutaminases as a Novel Target for SDHB-Associated Pheochromocytomas/Paragangliomas. Cancers, 2020, 12:599., DOI 10.3390/ cancers12030599 D’Antongiovanni V, Martinelli S, Richter S, et al. The Microenvironment Induces Collective Migration in SDHB-Silenced Mouse Pheochromocytoma Spheroids. Endocr Relat Cancer, 2017, 24:555–64. Erndt-Marino JD, and HahnMS. Probing the Response of Human Osteoblasts Following Exposure to Sympathetic Neuron-like PC-12 Cells in a 3D Coculture Model. J Biomed Mater Res, 2017, 105:984–90., DOI 10.1002/jbm. a.35964 Jin L, Feng Z-Q, Zhu M-L, Wang T, Leach MK, and Jiang Q. A Novel Fluffy Conductive Polypyrrole Nano-Layer Coated PLLA Fibrous Scaffold for Nerve Tissue Engineering. J Biomed Nanotechnology, 2012, 8:779–85., DOI 10.1166/ jbn.2012.1443 Ozone C, and Suga H. Functional Pituitary Tissue Formation. Methods Mol Biol Clifton NJ, 2017, 1597:57–65., DOI 10.1007/978-1-4939- 6949-4_5 Tsukada T, Kouki T, Fujiwara K, Ramadhani D, Horiguchi K, Kikuchi M, et al. Reassembly of Anterior Pituitary Organization by Hanging Drop Three- Dimensional Cell Culture. Acta Histochem Cytochem, 2013, 46:121–7., DOI 10.1267/ahc.13015 Diao J, Zhang C, Zhang D, et al. Role andMechanisms of a Three-Dimensional Bioprinted Microtissue Model in Promoting Proliferation and Invasion of Growth-Hormone-Secreting Pituitary Adenoma Cells. Biofabrication, 2019, 11:025006., DOI 10.1088/1758-5090/aaf7ea Tsukada T, Fujiwara K, Horiguchi K, Azuma M, Ramadhani D, Tofrizal A, et al. Folliculostellate Cells Are Required for Laminin Release from Gonadotrophs in Rat Anterior Pituitary. Acta Histochem Cytochem, 2014, 47:239–45., DOI 10.1267/ahc.14036 Balyura M, Gelfgat E, Ehrhart-Bornstein M, Ludwig B, Gendler Z, Barkai U, et al. Transplantation of Bovine Adrenocortical Cells Encapsulated in Alginate. Proc Natl Acad Sci USA, 2015, 112:2527–32., DOI 10.1073/pnas.1500242112 Zhang F, Lin H, Cao K,Wang H, Pan J, Zhuang J, et al. Vasculogenic Mimicry Plays an Important Role in Adrenocortical Carcinoma. Int J Urol, 2016, 23: 371–7., DOI 10.1111/iju.13070 Mallet C, Feraud O, Ouengue-Mbele G, Gaillard I, Sappay N, Vittet D, et al. Differential Expression of VEGF Receptors in Adrenal Atrophy Induced by Dexamethasone: a Protective Role of ACTH. Am J Physiology- EndocrinologyMetab, 2003, 284:E156–E167., DOI 10.1152/ajpendo.00450. 2001 Gaillard I, Prugnard E, Brand C, Chambaz E, and Vilgrain I. [Interactions between Steroidogenic Cells and Capillary Endothelial Cells in the Adrenal Gland]. Pathol Biol, Paris,, 1999, 47:314–21. Meszaros K, Karvaly G, Marta Z, Magda B, T}oke J, Szucs N, et al. Diagnostic Performance of a Newly Developed Salivary Cortisol and Cortisone Measurement Using an LC-MS/MS Method with Simple and Rapid Sample Preparation. J Endocrinol Invest, 2018, 41:315–23., DOI 10.1007/s40618-017- 0743-6 Langhans SA. Three-Dimensional In Vitro Cell Culture Models in Drug Discovery and Drug Repositioning. Front Pharmacol, 2018, 9:6., DOI 10. 3389/fphar.2018.00006 Basu B, Sirohi B, and Corrie P. Systemic Therapy for Neuroendocrine Tumours of Gastroenteropancreatic Origin. Endocr Relat Cancer, 2010, 17: R75–R90., DOI 10.1677/erc-09-0108 Megerle F, Herrmann W, Schloetelburg W, Ronchi CL, Pulzer A, Quinkler M, et al. Mitotane Monotherapy in Patients with Advanced Adrenocortical Carcinoma. J Clin Endocrinol Metab, 2018, 103:1686–95., DOI 10.1210/jc. 2017-02591 Else T, Kim AC, Sabolch A, Raymond VM, Kandathil A, Caoili EM, et al. Adrenocortical Carcinoma. Endocr Rev, 2014, 35:282–326., DOI 10.1210/er. 2013-1029 Lichtenauer UD, Shapiro I, Osswald A, Meurer S, Kulle A, Reincke M, et al. Characterization of NCI-H295r Cells as an In Vitro Model of Hyperaldosteronism. Horm Metab Res, 2013, 45:124–9., DOI 10.1055/s- 0032-1323810 Dai C, Liu X, Ma W, and Wang R. The Treatment of Refractory Pituitary Adenomas. Front Endocrinol, 2019, 10:334., DOI 10.3389/fendo.2019.00334 Marques P, and Korbonits M. Genetic Aspects of Pituitary Adenomas. Endocrinol Metab Clin North America, 2017, 46:335–74., DOI 10.1016/j.ecl. 2017.01.004 Baek N, Seo OW, Lee J, Hulme J, and An SS. Real-timeMonitoring of Cisplatin Cytotoxicity on Three-Dimensional Spheroid Tumor Cells. Drug Des Devel Ther, 2016, 10:2155–65., DOI 10.2147/DDDT.S108004 Safford SD, Coleman RE, Gockerman JP, Moore J, Feldman JM, Leight GS, et al. Iodine -131 Metaiodobenzylguanidine Is an Effective Treatment for Malignant Pheochromocytoma and Paraganglioma. Surgery, 2003, 134: 956–62; discussion 962-963., DOI 10.1016/s0039-6060(03)00426-4 Jimenez C, Rohren E, Habra MA, Rich T, Jimenez P, Ayala-Ramirez M, et al. Current and Future Treatments for Malignant Pheochromocytoma and Sympathetic Paraganglioma. Curr Oncol Rep, 2013, 15:356–71., DOI 10. 1007/s11912-013-0320-x Galan SR, and Kann PH. Genetics and Molecular Pathogenesis of Pheochromocytoma and Paraganglioma. Clin Endocrinol, 2013, 78:165–75., DOI 10.1111/cen.12071 Igaz P, Igaz I, Nagy Z, Nyir}o G, Szabo PM, Falus A, et al. MicroRNAs in Adrenal Tumors: Relevance for Pathogenesis, Diagnosis, and Therapy. Cell. Mol. Life Sci., 2015, 72:417–28., DOI 10.1007/s00018-014-1752-7 Jimenez C. Treatment for Patients with Malignant Pheochromocytomas and Paragangliomas: A Perspective from the Hallmarks of Cancer. Front Endocrinol, 2018, 9:277., DOI 10.3389/fendo.2018.00277 Tischler AS, and deKrijger RR. 15 YEARS of PARAGANGLIOMA: Pathology of Pheochromocytoma and Paraganglioma. Endocr Relat Cancer, 2015, 22: T123–T133., DOI 10.1530/erc-15-0261 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 640676 EP 640685 DI 10.3389/pore.2021.640676 PG 10 ER PT J AU Lin, H Zeng, W Lei, Y Chen, D Nie, Z AF Lin, Hua Zeng, Weifeng Lei, Yuhang Chen, Desheng Nie, Zhen TI Tuftelin 1 (TUFT1) Promotes the Proliferation and Migration of Renal Cell Carcinoma via PI3K/AKT Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE renal cell carcinoma; tuftelin 1; proliferation; migration; signaling pathway ID renal cell carcinoma; tuftelin 1; proliferation; migration; signaling pathway AB Tuftelin 1 (TUFT1), a protein functioning distinctively in different tissues, is reported to be elevated in several types of cancers and the elevation of TUFT1 is correlated with unfavorable clinicopathologic characteristics and poor survival. However, the involvement of TUFT1 in renal cell carcinoma (RCC) remains unknown. In the current study, we investigated the role of TUFT1 in RCC and potential underlying mechanisms. RTPCR and Western blot analysis showed that both the mRNA and protein levels of TUFT1 were increased in primary RCC tissue and RCC cell lines. TUFT1 overexpression in RCC cells resulted in enhanced cell proliferation and migration while knockdown of TUFT1 by contrast decreased the growth and migration of the RCC cells, indicating TUFT1 expression is involved in RCC cell growth and migration. The involvement of TUFT1 in the epithelial-mesenchymal transition (EMT) of RCC cells was also determined by measuring the expression of EMT-related markers. Our data showed that TUFT1 overexpression promoted RCC cell EMT progression while knockdown of TUFT1 suppressed such process. Further signaling pathway inhibition assay revealed that TUFT1-induced RCC cell growth, migration and EMT was significantly suppressed by PI3K inhibitor, but not JNK or MEK inhibitors. In addition, TUFT1 overexpression enhanced the AKT phosphorylation, a key member of the PI3K signaling pathway, while PI3K inhibitor suppressed such process. Taken together, our study showed that TUFT1 expression was elevated in RCC and such elevation promoted the proliferation, migration and EMT of RCC cells in vitro, through PI3K/AKT signaling pathway. The findings of our current study imply that TUFT1 is involved in RCC tumorigenesis, and it may serve as a biomarker for RCC diagnosis and a potential target for RCC treatment. C1 [Lin, Hua] Xiantao First People’s Hospital Affiliated to Yangtze University, Department of UrologyXiantao, China. [Zeng, Weifeng] Xiantao First People’s Hospital Affiliated to Yangtze University, Department of UrologyXiantao, China. [Lei, Yuhang] Xiantao First People’s Hospital Affiliated to Yangtze University, Department of UrologyXiantao, China. [Chen, Desheng] Xiantao First People’s Hospital Affiliated to Yangtze University, Department of UrologyXiantao, China. [Nie, Zhen] Xiantao First People’s Hospital Affiliated to Yangtze University, Department of UrologyXiantao, China. RP Nie, Z (reprint author), Xiantao First People’s Hospital Affiliated to Yangtze University, Department of Urology, Xiantao, China. EM n_zhen@hotmail.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, et al. Renal cell carcinoma. Nat Rev Dis Primers, 2017, 3(1):17009., DOI 10.1038/ nrdp.2017.9 Van Poppel H, Da Pozzo L, Albrecht W, Matveev V, Bono A, Borkowski A, et al. A prospective, randomised EORTC intergroup phase 3 study comparing the oncologic outcome of elective nephron-sparing surgery and radical nephrectomy for low-stage renal cell carcinoma. Eur Urol, 2011, 59(4): 543–52., DOI 10.1016/j.eururo.2010.12.013 El Dib R, Touma NJ, and Kapoor A. Cryoablation vs radiofrequency ablation for the treatment of renal cell carcinoma: a meta-analysis of case series studies. BJU Int, 2012, 110(4):510–6., DOI 10.1111/j.1464-410x.2011.10885.x Ljungberg B, Albiges L, Abu-Ghanem Y, Bensalah K, Dabestani S, Fernandez- Pello S, et al. European association of urology guidelines on renal cell carcinoma: the 2019 update. Eur Urol, 2019, 75(5):799–810., DOI 10.1016/j. eururo.2019.02.011 McDermott DF, Regan MM, Clark JI, Flaherty LE, Weiss GR, Logan TF, et al. Randomized phase III trial of high-dose interleukin-2 versus subcutaneous interleukin-2 and interferon in patients with metastatic renal cell carcinoma. J Clin Oncol, 2004, 23(1):133–41., DOI 10.1200/ JCO.2005.03.206 Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med, 2007, 356(2):125–34., DOI 10.1056/nejmoa060655 Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med, 2007, 356(2):115–24., DOI 10.1056/nejmoa065044 Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma, AXIS): a randomised phase 3 trial. The Lancet, 2011, 378(9807): 1931–9., DOI 10.1016/s0140-6736(11)61613-9 Posadas EM, Limvorasak S, and Figlin RA. Targeted therapies for renal cell arcinoma. Nat Rev Nephrol, 2017, 13(8):496., DOI 10.1038/nrneph. 2017.82 Unverzagt S, Moldenhauer I, Nothacker M, Rossmeissl D, Hadjinicolaou AV, Peinemann F, et al. Immunotherapy for metastatic renal cell carcinoma. Cochrane Database Syst Rev, 2017, 5:CD011673., DOI 10.1002/14651858. CD011673.pub2 Xu JX,Maher VE, Zhang L, Tang S, Sridhara R, Ibrahim A, et al. FDA approval summary: nivolumab in advanced renal cell carcinoma after anti-angiogenic therapy and exploratory predictive biomarker analysis. Oncol, 2017, 22(3):311., DOI 10.1634/theoncologist.2016-0476 Rini BI, Battle D, Figlin RA, George DJ, Hammers H, Hutson T, et al. The society for immunotherapy of cancer consensus statement on immunotherapy for the treatment of advanced renal cell carcinoma, RCC). J Immunotherapy Cancer, 2019, 7(1):354., DOI 10.1186/s40425-019-0813-8 Network NCC. Referenced with Permission from the NCCN Clinical Practice Guidelines in Oncology, NCCN Guidelines®, for Non-small Cell Lung Cancer V1, 2019). , 2020. © National Comprehensive Cancer Network, Inc. 2019, Plymouth Meeting, PA: 2020. © National Comprehensive Cancer Network, Inc. 2019. All rights reserved, Accessed November 6). Atkins MB, Clark JI, and Quinn DI. Immune checkpoint inhibitors in advanced renal cell carcinoma: experience to date and future directions. Ann Oncol, 2017, 28(7):1484–94., DOI 10.1093/annonc/mdx151 Deutsch D, Palmon A, Dafni L, Mao Z, Leytin V, Young M, et al. Tuftelin–aspects of protein and gene structure. Eur J Oral Sci, 1998, 106(S1):315–23., DOI 10.1111/j.1600-0722.1998.tb02192.x Leiser Y, Silverstein N, Blumenfeld A, Shilo D, Haze A, Rosenfeld E, et al. The induction of tuftelin expression in PC12 cell line during hypoxia and NGFinduced differentiation. J Cell. Physiol, 2011, 226(1):165–72., DOI 10.1002/jcp. 22318 Liu W, Zhang L, Jin Z, Zhao M, Li Z, Chen G, et al. TUFT1 is expressed in breast cancer and involved in cancer cell proliferation and urvival. Oncotarget, 2017, 8(43):74962–74., DOI 10.18632/oncotarget.20472 Yu YP, He JG, Li P, Qiu NH, Wang LJ, and Feng H. TUFT1 promotes osteosarcoma cell proliferation and predicts poor prognosis in osteosarcoma atients. Open Life Sci, 2018, 13(1):396–403., DOI 10.1515/ biol-2018-0048 Dou C, Zhou Z, Xu Q, Liu Z, Zeng Y,Wang Y, et al. Hypoxia-induced TUFT1 promotes the growth and metastasis of hepatocellular carcinoma by activating the Ca2+/PI3K/AKT pathway. Oncogene, 2019, 38(8):1239–55., DOI 10.1038/ s41388-018-0505-8 Liu W, Han J, Shi S, Dai Y, and He J. TUFT1 promotes metastasis and chemoresistance in triple negative breast cancer through the TUFT1/Rab5/ Rac1 pathway. Cancer Cell Int, 2019, 19(1):242., DOI 10.1186/s12935-019- 0961-4 Hu K, Fu M, Wang J, Luo S, Barreto M, Singh R, et al. HSV-2 infection of human genital epithelial cells upregulates TLR9 expression through the SP1/ JNK signaling pathway. Front Immunol, 2020, 11(356)., DOI 10.3389/fimmu. 2020.00356 Fu M, Hu K, Hu H, Ni F, Du T, Shattock RJ, et al. Antigenicity and immunogenicity of HIV-1 gp140 with different combinations of glycan mutation and V1/V2 region or V3 crown deletion. Vaccine, 2019, 37(51): 7501–8., DOI 10.1016/j.vaccine.2019.09.073 Liang CC, Park AY, and Guan JL. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc, 2007, 2(2):329–33., DOI 10.1038/nprot.2007.30 Pijuan J, Barcelo C, Moreno DF, Maiques O, Siso P, Marti RM, et al. In vitro cell migration, invasion, and adhesion assays: from cell imaging to data analysis. Front Cell Dev Biol, 2019, 7(107)., DOI 10.3389/fcell.2019.00107 Kalluri R, andWeinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest, 2009, 119(6):1420–8., DOI 10.1172/JCI39104 Lamouille S, Xu J, and Derynck R. Molecular mechanisms of epithelialmesenchymal transition. Nat Rev Mol Cell Biol, 2014, 15(3):178–96., DOI 10. 1038/nrm3758 Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer, 2002, 2(6):442–54., DOI 10.1038/nrc822 Onder TT, Gupta PB, Mani SA, Yang J, Lander ES, and Weinberg RA. Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res, 2008, 68(10):3645–54., DOI 10.1158/0008-5472.Can- 07-2938 Hulit J, Suyama K, Chung S, Keren R, Agiostratidou G, Shan W, et al. N-cadherin signaling potentiates mammary tumor metastasis via enhanced extracellular signal-regulated kinase activation. Cancer Res, 2007, 67(7): 3106–16., DOI 10.1158/0008-5472.Can-06-3401 Wang M, Ren D, Guo W, Huang S, Wang Z, Li Q, et al. N-cadherin promotes epithelial-mesenchymal transition and cancer stem cell-like traits via ErbB signaling in prostate cancer cells. Int J Oncol, 2016, 48(2):595–606., DOI 10. 3892/ijo.2015.3270 Nieto MA. The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol, 2002, 3(3):155–66., DOI 10.1038/nrm757 Vara JAF, Casado E, de Castro J, Cejas P, Belda-Iniesta C, and Gonzalez-Baron M. PI3K/Akt signalling pathway and cancer. Cancer Treat Rev, 2004, 30(2): 193–204., DOI 10.1016/j.ctrv.2003.07.007 Bubici C, and Papa S. JNK signalling in cancer: in need of new, smarter therapeutic targets. Br J Pharmacol, 2014, 171(1):24–37., DOI 10.1111/bph. 12432 Neuzillet C, Tijeras-Raballand A, de Mestier L, Cros J, Faivre S, and Raymond E. MEK in cancer and cancer therapy. Pharmacol Ther, 2014, 141(2):160–71., DOI 10.1016/j.pharmthera.2013.10.001 Delgado S, Deutsch D, and Sire JY. Evolutionary analysis of the mammalian tuftelin sequence reveals features of functional importance. IntMol Evol, 2017, 84:214–224., DOI 10.1007/s00239-017-9789-5 Leiser Y, Blumenfeld A, Haze A, Dafni L, Taylor AL, Rosenfeld E, et al. Localization, quantification, and characterization of tuftelin in soft tissues. Anat Rec, 2007, 290(5):449–54., DOI 10.1002/ar.20512 Sliz E, Taipale M, Welling M, Skarp S, Alaraudanjoki V, Ignatius J, et al. TUFT1, a novel candidate gene for metatarsophalangeal osteoarthritis, plays a role in chondrogenesis on a calcium-related pathway. PLoS one, 2017, 12(4): e0175474., DOI 10.1371/journal.pone.0175474 Kawasaki N, Isogaya K, Dan S, Yamori T, Takano H, Yao R, et al. TUFT1 interacts with RABGAP1 and regulates mTORC1 signaling. Cell Discov, 2018, 4(1):1–16., DOI 10.1038/s41421-017-0001-2 Rini BI, Campbell SC, and Escudier B. Renal cell carcinoma. The Lancet, 2009, 373(9669):1119–32., DOI 10.1016/s0140-6736(09)60229-4 The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature, 2013, 499(7456): 43., DOI 10.1038/nature12222 Davis CF, Ricketts CJ, Wang M, Yang L, Cherniack AD, Shen H, et al. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer cell, 2014, 26(3):319–30., DOI 10.1016/j.ccr.2014.07.014 Cairns P. Renal cell carcinoma. Cbm, 2011, 9(1–6):461–73., DOI 10.3233/cbm- 2011-0176 Altomare DA, and Testa JR. Perturbations of the AKT signaling pathway in human cancer. Oncogene, 2005, 24(50):7455–64., DOI 10.1038/sj.onc.1209085 Roberts PJ, and Der CJ. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene, 2007, 26(22): 3291–310., DOI 10.1038/sj.onc.1210422 Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, et al. Oncogenic signaling pathways in the cancer genome atlas. Cell, 2018, 173(2): 321–37.e10., DOI 10.1016/j.cell.2018.03.035 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 640936 EP 640945 DI 10.3389/pore.2021.640936 PG 10 ER PT J AU Cai, M Tan, R Huang, Y Chen, X Kong, Q Guo, K Xu, M AF Cai, Min Tan, Rukeng Huang, Yunyi Chen, Xuanyi Kong, Qingci Guo, Kaixin Xu, Meng TI High Expression of Tomm34 and Its Correlations With Clinicopathology in Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE oral squamous cell carcinoma; Tomm34; Immunohistochemistry; clinicopathology; bioinformatics ID oral squamous cell carcinoma; Tomm34; Immunohistochemistry; clinicopathology; bioinformatics AB Tomm34, as a member of the outer mitochondrial membrane proteins, is evenly distributed between the cytoplasmand the outer mitochondrialmembrane. It is up-regulated in a variety of tumors and correlates with poor prognosis. This study aimed to investigate expression of Tomm34 and its correlations with clinicopathology in oral squamous cell carcinoma (OSCC). Oncomine database and UALCAN database were utilized to predict the expression and prognosis values of Tomm34 in head and neck squamous cell carcinoma (HNSCC). By immunohistochemistry, a retrospective study was performed to verify the bioinformatics results to evaluate the Tomm34 expression and clinicopathological variables in both HPVpositive OSCC and HPV-negative OSCC. Immunohistochemistry of our cohort revealed that 48 cases fulfilled the Tomm34 high expression judgment criteria, and the overall positive rate was 60% (48/80), and 27 cases fulfilled the p16 expression judgment criteria (33.75%, 27/ 80). The high expression of Tomm34 was closely related with the TNM classification of OSCC (p < 0.01) and tumor size (p < 0.01) both in HPV-negative OSCC and HPV-positive OSCC, while related with lymph node metastasis (p = 0.001) in HPV-negative OSCC and drinking history (p = 0.044) in HPV-positive OSCC. In addition, the Kaplan-Meier curves indicated that higher level of Tomm34 was correlated with poorer overall survival (OS) and disease-free survival (DFS) in HPV-negativeOSCC (OS, p = 0.046; DFS, p = 0.020) but not in HPV-positive OSCC (OS, p = 0.824; DFS, p = 0.782). In conclusion, Tomm34 is highly expressed in OSCC and may be a useful factor to provide prognostic information, especially in HPV-negative OSCC group. C1 [Cai, Min] Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Hospital of StomatologyGuangzhou, China. [Tan, Rukeng] Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Hospital of StomatologyGuangzhou, China. [Huang, Yunyi] Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Hospital of StomatologyGuangzhou, China. [Chen, Xuanyi] Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Hospital of StomatologyGuangzhou, China. [Kong, Qingci] Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Hospital of StomatologyGuangzhou, China. [Guo, Kaixin] Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Hospital of StomatologyGuangzhou, China. [Xu, Meng] Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Hospital of StomatologyGuangzhou, China. RP Xu, M (reprint author), Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Hospital of Stomatology, Guangzhou, China. EM xumeng23@mail.sysu.edu.cn CR Rahman QB, Iocca O, Kufta K, Shanti RM. Global burden of head and neck cancer. Oral Maxillofac Surg Clin North Am, 2020, 32(3):367–75., DOI 10.1016/j. coms.2020.04.002 Mourad M, Jetmore T, Jategaonkar AA, Moubayed S, Moshier E, Urken ML. Epidemiological trends of head and neck cancer in the United States: a seer population study. J Oral Maxillofac Surg, 2017, 75(12):2562–72., DOI 10.1016/j. joms.2017.05.008 Ding D, Stokes W, Eguchi M, Hararah M, Sumner W, Amini A, et al. Association between lymph node ratio and recurrence and survival outcomes in patients with oral cavity cancer. JAMA Otolaryngol Head Neck Surg, 2019, 145(1):53–61., DOI 10.1001/jamaoto.2018.2974 Dionne KR, Warnakulasuriya S, Zain RB, Cheong SC. Potentially malignant disorders of the oral cavity: current practice and future directions in the clinic and laboratory. Int J Cancer, 2015, 136(3):503–15., DOI 10.1002/ijc.28754 Zaravinos A. An updated overview of HPV-associated head and neck carcinomas. Oncotarget, 2014, 5(12):3956–69., DOI 10.18632/oncotarget.1934 Huang SH, O’Sullivan B. Overview of the 8th edition TNM classification for head and neck cancer. Curr Treat Options Oncol, 2017, 18(7):13., DOI 10.1007/ s11864-017-0484-y Mueller S. Update from the 4th edition of the world health organization of head and neck tumours: tumours of the oral cavity and mobile tongue. Head Neck Pathol, 2017, 11(1):33–40., DOI 10.1007/s12105-017-0792-3 Westra WH, Lewis JS, Jr. Update from the 4th edition of the world health organization classification of head and neck tumours: oropharynx. Head Neck Pathol, 2017, 11(1):41–7., DOI 10.1007/s12105-017-0793-2 Paver EC, Currie AM, Gupta R, Dahlstrom JE. Human papilloma virus related squamous cell carcinomas of the head and neck: diagnosis, clinical implications and detection of HPV. Pathology, 2020, 52(2):179–91., DOI 10.1016/j.pathol. 2019.10.008 Nuttall SD, Hanson BJ, Mori M, Hoogenraad NJ. hTom34: a novel translocase for the import of proteins into human mitochondria. DNA Cell Biol, 1997, 16(9):1067–74., DOI 10.1089/dna.1997.16.1067 Yang CS,Weiner H. Yeast two-hybrid screening identifies binding partners of human Tom34 that have ATPase activity and form a complex with Tom34 in the cytosol. Arch Biochem Biophys, 2002, 400(1):105–10., DOI 10.1006/abbi. 2002.2778 Chewawiwat N, YanoM, Terada K, Hoogenraad NJ, MoriM. Characterization of the novel mitochondrial protein import component, Tom34, in mammalian cells. J Biochem, 1999, 125(4):721–7., DOI 10.1093/oxfordjournals.jbchem. a022342 Faou P, Hoogenraad NJ. Tom34: a cytosolic cochaperone of the Hsp90/Hsp70 protein complex involved in mitochondrial protein import. Biochim Biophys Acta, 2012, 1823(2):348–57., DOI 10.1016/j.bbamcr.2011.12.001 Trcka F, Durech M, Man P, Hernychova L, Muller P, Vojtesek B. The assembly and intermolecular properties of the Hsp70-Tomm34-Hsp90 molecular chaperone complex. J Biol Chem, 2014, 289(14):9887–901., DOI 10.1074/jbc. M113.526046 Matsushita N, Yamamoto S, Inoue Y, Aruga A, Yamamoto M. RT-qPCR analysis of the tumor antigens TOMM34 and RNF43 in samples extracted from paraffin-embedded specimens of colorectal cancer. Oncol Lett, 2017, 14(2):2281–7., DOI 10.3892/ol.2017.6412 Toraih EA, Alrefai HG, Hussein MH, Helal GM, Khashana MS, Fawzy MS. Overexpression of heat shock protein HSP90AA1 and translocase of the outer mitochondrial membrane TOM34 in HCV-induced hepatocellular carcinoma: a pilot study. Clin Biochem, 2019, 63:10–7., DOI 10.1016/j.clinbiochem.2018. 12.001 Ahmed MAH, Ali MH, Abbas HH, Elatrash GA, Foda AAM. Expression of TOMM34 and its clinicopathological correlations in urothelial carcinoma of the bladder. Pathol Oncol Res, 2020, 26(1):411–8., DOI 10.1007/s12253-018- 0524-3 Aleskandarany MA, Negm OH, Rakha EA, Ahmed MA, Nolan CC, Ball GR, et al. TOMM34 expression in early invasive breast cancer: a biomarker associated with poor outcome. Breast Cancer Res Treat, 2012, 136(2): 419–27., DOI 10.1007/s10549-012-2249-4 Aleskandarany MA, Soria D, Green AR, Nolan C, Diez-Rodriguez M, Ellis IO, et al. Markers of progression in early-stage invasive breast cancer: a predictive immunohistochemical panel algorithm for distant recurrence risk stratification. Breast Cancer Res Treat, 2015, 151(2):325–33., DOI 10.1007/ s10549-015-3406-3 Muller P, Coates PJ, Nenutil R, Trcka F, Hrstka R, Chovanec J, et al. Tomm34 is commonly expressed in epithelial ovarian cancer and associates with tumour type and high FIGO stage. J Ovarian Res, 2019, 12(1):30., DOI 10.1186/s13048- 019-0498-0 Shimokawa T, Matsushima S, Tsunoda T, Tahara H, Nakamura Y, Furukawa Y. Identification ofTOMM34, which shows elevated expression in the majority of human colon cancers, as a novel drug target. Int J Oncol, 2006, 29(2):381–6., DOI 10.3892/ijo.29.2.381 Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce- Rodriguez I, Chakravarthi B, et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia, 2017, 19(8): 649–58., DOI 10.1016/j.neo.2017.05.002 Dard L, Blanchard W, Hubert C, Lacombe D, Rossignol R. Mitochondrial functions and rare diseases. Mol Aspects Med, 2020, 71:100842., DOI 10.1016/j. mam.2019.100842 Ryan MT, Wagner R, Pfanner N. The transport machinery for the import of preproteins across the outer mitochondrial membrane. Int J Biochem Cell Biol, 2000, 32(1):13–21., DOI 10.1016/s1357-2725(99)00114-4 Gouirand V, Guillaumond F, Vasseur S. Influence of the tumor microenvironment on cancer cells metabolic reprogramming. Front Oncol, 2018, 8:117., DOI 10.3389/fonc.2018.00117 Tsaytler PA, Krijgsveld J, Goerdayal SS, Rudiger S, Egmond MR. Novel Hsp90 partners discovered using complementary proteomic approaches. Cell Stress Chaperones, 2009, 14(6):629–38., DOI 10.1007/s12192-009-0115-z Schlegel T, Mirus O, von Haeseler A, Schleiff E. The tetratricopeptide repeats of receptors involved in protein translocation across membranes. Mol Biol Evol, 2007, 24(12):2763–74., DOI 10.1093/molbev/msm211 Calderwood SK, Khaleque MA, Sawyer DB, Ciocca DR. Heat shock proteins in cancer: chaperones of tumorigenesis. Trends Biochem Sci, 2006, 31(3):164–72., DOI 10.1016/j.tibs.2006.01.006 Rodina A, Vilenchik M, Moulick K, Aguirre J, Kim J, Chiang A, et al. Selective compounds define Hsp90 as a major inhibitor of apoptosis in small-cell lung cancer. Nat Chem Biol, 2007, 3(8):498–507., DOI 10.1038/nchembio.2007.10 Giaginis C, Daskalopoulou SS, Vgenopoulou S, Sfiniadakis I, Kouraklis G, Theocharis SE. Heat Shock Protein-27, -60 and -90 expression in gastric cancer: association with clinicopathological variables and patient survival. BMC Gastroenterol, 2009, 9:14., DOI 10.1186/1471-230X-9-14 Pick E, Kluger Y, Giltnane JM, Moeder C, Camp RL, Rimm DL, et al. High HSP90 expression is associated with decreased survival in breast cancer. Cancer Res, 2007, 67(7):2932–7., DOI 10.1158/0008-5472.CAN-06-4511 Priyanka KP, Majumdar S, Kotina S, Uppala D, Balla H. Expression of heat shock protein 70 in oral epithelial dysplasia and oral squamous cell carcinoma: an immunohistochemical study. Contemp Clin Dentistry, 2019, 10(2):185–90., DOI 10.4103/ccd.ccd_101_18 Chanthammachat P, Promwikorn W, Pruegsanusak K, Roytrakul S, Srisomsap C, Chokchaichamnankit D, et al. Comparative proteomic analysis of oral squamous cell carcinoma and adjacent non-tumour tissue from Thailand. Arch Oral Biol, 2013, 58(11):1677–85., DOI 10.1016/j.archoralbio.2013.08.002 Markopoulos AK, Deligianni E, Antoniades DZ. Heat shock protein 70 membrane expression in oral cancer: a possible new target in antineoplastic therapy? Chemotherapy, 2009, 55(4):211–4., DOI 10. 1159/000218099 Lee S-S, Tsai C-H, Ho Y-C, Chang Y-C. The upregulation of heat shock protein 70 expression in areca quid chewing-associated oral squamous cell carcinomas. Oral Oncol, 2008, 44(9):884–90., DOI 10.1016/j.oraloncology. 2007.11.004 Shevtsov M, Balogi Z, Khachatryan W, Gao H, Vigh L, Multhoff G. Membrane-associated heat shock proteins in Oncology: from basic research to new theranostic targets. Cells, 2020, 9(5)., DOI 10.3390/cells9051263 Pickering CR, Zhang JX, Yoo SY, Bengtsson L, Moorthy S, Neskey DM, et al. Integrative genomic characterization of oral squamous cell carcinoma identifies frequent somatic drivers. Cancer Discov, 2013, 3(7):770–81., DOI 10.1158/2159-8290.Cd-12-0537 Blesa JR, Prieto-Ruiz JA, Abraham BA, Harrison BL, Hegde AA, Hernandez- Yago J. NRF-1 is the major transcription factor regulating the expression of the human TOMM34 gene. Biochem Cel Biol, 2008, 86(1):46–56., DOI 10.1139/ o07-151 Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem Sci, 2014, 39(4):199–218., DOI 10.1016/j.tibs.2014.02.002 Terada K, Ueno S, Yomogida K, Imai T, Kiyonari H, Takeda N, et al. Expression of Tom34 splicing isoforms in mouse testis and knockout of Tom34 in mice. J Biochem, 2003, 133(5):625–31., DOI 10.1093/jb/ mvg080 Okuno K, Sugiura F, Hida JI, Tokoro T, Ishimaru E, Sukegawa Y, et al. Phase I clinical trial of a novel peptide vaccine in combination with UFT/LV for metastatic colorectal cancer. Exp Ther Med, 2011, 2(1):73–9., DOI 10.3892/etm. 2010.182 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 641042 EP 641051 DI 10.3389/pore.2021.641042 PG 10 ER PT J AU Peeters, M Geusens, J Van der Cruyssen, F Michaux, L de Leval, L Tousseyn, Th Vandenberghe, P Politis, C AF Peeters, Maxime Geusens, Joris Van der Cruyssen, Frederic Michaux, Lucienne de Leval, Laurence Tousseyn, Thomas Vandenberghe, Peter Politis, Constantinus TI Case Report: Spontaneous Remission of an Infraorbital Follicular B-Cell Lymphoma: Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE follicular lymphoma; cheek; spontaneous remission; head and neck neoplasms; case reports ID follicular lymphoma; cheek; spontaneous remission; head and neck neoplasms; case reports AB Non-Hodgkin lymphomas comprise a heterogeneous group of malignancies, with a wide scope of clinical, radiological and histological presentations. In this paper, a case is presented of a 59-year-old white male with an infraorbital follicular B-cell lymphoma, which appeared as a painless mass in the left cheek. The lymphoma achieved spontaneous remission five and a half months after his diagnostic incision biopsy. The literature is reviewed, focusing on this rare site of presentation and spontaneous remission. In literature, only four cases have been reported with a follicular B-cell lymphoma of the cheek or infraorbital region, and only 26 cases of spontaneous remission of an extracranial non-Hodgkin lymphoma in the head and neck region have been described. To the authors’ best knowledge, this is the first time spontaneous remission of an infraorbital follicular lymphoma could be observed. The nature of the processes inducing spontaneous remission remains obscure. It is important to recognize this phenomenon as this might prevent unnecessary treatment. C1 [Peeters, Maxime] University Hospitals Leuven, Department of Oral and Maxillofacial SurgeryLeuven, Belgium. [Geusens, Joris] University Hospitals Leuven, Department of Oral and Maxillofacial SurgeryLeuven, Belgium. [Van der Cruyssen, Frederic] University Hospitals Leuven, Department of Oral and Maxillofacial SurgeryLeuven, Belgium. [Michaux, Lucienne] University Hospitals Leuven, Department of Human GeneticsLeuven, Belgium. [de Leval, Laurence] Lausanne University Hospital (CHUV) and Lausanne University, Department of PathologyLausanne, Switzerland. [Tousseyn, Thomas] University Hospitals Leuven, Department of PathologyLeuven, Belgium. [Vandenberghe, Peter] University Hospitals Leuven, Department of HematologyLeuven, Belgium. [Politis, Constantinus] University Hospitals Leuven, Department of Oral and Maxillofacial SurgeryLeuven, Belgium. RP Peeters, M (reprint author), University Hospitals Leuven, Department of Oral and Maxillofacial Surgery, Leuven, Belgium. EM maxime.peeters.mp1@gmail.com CR Luminari S, Bellei M, Biasoli I, Federico M Follicular lymphoma. Rev Bras Hematol Hemoter, 2011, 34:54., DOI 10.5581/1516-8484.20120015 Jessy T. Immunity over inability: the spontaneous regression of cancer. J Nat Sci Biol Med, 2011, 2:43., DOI 10.4103/0976-9668.82318 Vega F, Lin P, Medeiros LJ. Extranodal lymphomas of the head and neck. Ann Diagn Pathol, 2005, 9:340., DOI 10.1016/j.anndiagpath.2005.09.020 Malaguarnera M, Giordano M, Russo C, Puzzo L, Trainiti M, Consoli AS, et al. Lymphoma of cheek: a case report. Eur Rev Med Pharmacol Sci, 2012, 16 Suppl 4(4 Suppl. l):4–7. PMID: 23090795. Gleeson MJ, Bennett MH, Cawson RA. Lymphomas of salivary glands. Cancer, 1986, 58:699., DOI 10.1002/1097-0142(19860801)58:3<699::AIDCNCR2820580317> 3.0.CO;699-E Wright DH. Pathology of extra-nodal non hodgkin lymphomas. Clin Oncol, 2012, 24:319., DOI 10.1016/j.clon.2012.02.004 Goodlad JR, MacPherson S, Jackson R, Batstone P, White J. Extranodal follicular lymphoma: a clinicopathological and genetic analysis of 15 cases arising at non-cutaneous extranodal sites. Histopathology, 2004, 44:268., DOI 10. 1111/j.1365-2559.2004.01804.x Tart RP, Mukherji SK, Avino AJ, Stringer SP, Mancuso AA. Facial lymph nodes: normal and abnormal CT appearance. Radiology, 1993, 13:35., DOI 10.1148/ radiology.188.3.8351335 PanW-R, Le Roux CM, Briggs CA. Variations in the lymphatic drainage pattern of the head and neck: further anatomic studies and clinical implications. Plast Reconstr Surg, 2011, 127:611., DOI 10.1097/PRS.0b013e3181fed511 Bou-Assaly W. The forgotten lymph nodes: review of the superficial head and neck lymphatic system. J Radiol Imaging, 2016, 19:172., DOI 10.14312/2399- 8172.2016-3 Kukreti V, Petersen P, Pintilie M, Tsang R, Crump M, Gospodarowicz M. Extranodal follicular lymphoma - a retrospective review and comparison with localized nodal follicular lymphoma. Blood, 2004, 104:1375., DOI 10.1182/ blood.V104.11.1375.1375 Buchpiguel CA. Current status of PET/CT in the diagnosis and follow up of lymphomas. Rev Bras Hematol Hemoter, 2011, 33:140., DOI 10.5581/1516- 8484.20110035 Johnson SA, Kumar A, Matasar MJ, Schoder H, Rademaker J. Imaging for staging and response assessment in lymphoma. Radiology, 2015, 276:323., DOI 10.1148/radiol.2015142088 Carbone A, Roulland S, Gloghini A, Younes A, von Keudell G, Lopez- Guillermo A, et al. Follicular lymphoma. Nat Rev Dis Primers, 2019, 5:132., DOI 10.1038/s41572-019-0132-x Costa F, Cian R, Robiony M, Zerman N, Politi M. Unilateral swelling of the cheek. J Oral Maxillofac Surg, 2008, 66:342., DOI 10.1016/j.joms.2007.04.030 Graham RM, Thomson EF, Cousin GC, Kumar S, Awasthi A. A case of facial lymphoma mimicking dental infection. Dental Update, 2009, 36:244., DOI 10. 12968/denu.2009.36.4.244 Weiler-Sagie M, Bushelev O, EpelbaumR, Dann EJ, Haim N, Avivi I, et al. 18FFDG avidity in lymphoma readdressed: a study of 766 patients. J Nucl Med, 2010, 51:25., DOI 10.2967/jnumed.109.067892 Luminari S, Biasoli I, Arcaini L, Versari A, Rusconi C, Merli F, et al. The use of FDG-PET in the initial staging of 142 patients with follicular lymphoma: a retrospective study from the FOLL05 randomized trial of the Fondazione Italiana Linfomi. Ann Oncol, 2013, 24:2108., DOI 10.1093/annonc/mdt137 Bachy E, Maurer MJ, Habermann TM, Gelas-Dore B, Maucort-Boulch D, Estell JA, et al. A simplified scoring system in de novo follicular lymphoma treated initially with immunochemotherapy. Blood, 2018, 132(1):49–58., DOI 10.1182/blood-2017-11-816405 Federico M, Bellei M, Marcheselli L, Luminari S, Lopez-Guillermo A, Vitolo U, et al. Follicular lymphoma international prognostic index 2: a new prognostic index for follicular lymphoma developed by the international follicular lymphoma prognostic factor project. Jco, 2009, 27:4555., DOI 10.1200/JCO.2008.21.3991 Potts DA, Fromm JR, Gopal AK, Cassaday RD. Spontaneous remission of an untreated, myc and BCL2 coexpressing, high-grade B-cell lymphoma: a case report and literature review. Case Rep Hematol, 2017, 2017:1., DOI 10.1155/ 2017/2676254 Papac RJ. Spontaneous regression of cancer: possible mechanisms. In Vivo, Brooklyn,, 1998, 12(6):571-8. Kumamoto M, Nakamine H, Hara T, Yokoya Y, Kawai J, Ito H, et al. Spontaneous complete regression of high grade non-Hodgkin’s lymphoma. Morphologic, immunohistochemical, and gene amplification analyses. Cancer, 1994, 74:320., DOI 10.1002/1097-0142(19941201)74:11<3023::AIDCNCR2820741120> 3.0.CO;3023-Y Rawal S, Chu F, Zhang M, Park HJ, Nattamai D, Kannan S, et al. Cross talk between follicular Th cells and tumor cells in human follicular lymphoma promotes immune evasion in the tumor microenvironment. J Immunol, 2013, 190:6681., DOI 10.4049/jimmunol.1201363 Farquhar D, Sobanko J, Williams K, Newman JG. A vanishing lymphoma in the cheek. ORL, 2014, 76:189., DOI 10.1159/000365853 Hardin C. Unexpected diagnosis of follicular lymphoma during Mohs micrographic surgery. J Am Acad Dermatol, 2018, 5:12., DOI 10.1016/j.jaad. 2018.05.1208 Dhadlie S, Strekozov B. Cutaneous extra nodal lymphoma relapse: a case report and review of literature. Int J Surg Case Rep, 2018, 51:306., DOI 10.1016/j. ijscr.2018.09.020 Thaker R, Lee KC, Peters S, Greenman D, Kings JR. Asymptomatic nodule in the right cheek in a 65-year-old female. Oral Surg Oral Med Oral Pathol Oral Radiol, 2019, 128:567., DOI 10.1016/j.oooo.2019.02.017 Burkitt DP, Kyalwazi SK. Spontaneous remission of African lymphoma. Br J Cancer, 1967, 21:14., DOI 10.1038/bjc.1967.2 Grem JL, Hafez GR, Brandenburg JH, Carbone PP. Spontaneous remission in diffuse large cell lymphoma. Cancer, 1986, 28:1097., DOI 10.1002/1097- 0142(19860515)57:10<2042::AID-CNCR2820571027>3.0.CO;2-# Poppema S, Postma L, Brinker M, De Jong B. Spontaneous regression of a small non-cleaved cell malignant lymphoma, non-Burkitt’s lymphoblastic lymphoma). Morphologic, immunohistological, and immunoglobulin gene analysis. Cancer, 1988, 62:791., DOI 10.1002/1097-0142(19880815)62:4<791:: AID-CNCR2820620425>3.0.CO;791-M Savarrio L, Gibson J, Dunlop DJ, O’Rourke N, Fitzsimons EJ. Spontaneous regression of an anaplastic large cell lymphoma in the oral cavity: first reported case and review of the literature. Oral Oncol, 1999, 35:609., DOI 10.1016/S1368- 8375(99)00034-2 Koga M, Kusukawa J, Hayabuchi N. Spontaneous regression of extranodal malignant lymphoma occurred in the gingiva. Oral Oncol, 2003, 39:323., DOI 10.1016/S1368-8375(02)00122-7 Heibel H, Knodgen R, Bredenfeld H, Wickenhauser C, Scheer M, Zoller JE. Complete spontaneous remission of an aggressive non-hodgkin’s lymphoma with primary manifestation in the oral cavity. Leuk Lymphoma, 2004, 45:171., DOI 10.1080/1042819031000139747 Sasaki K, Sugaya M, Fujita H, Takeuchi K, Torii H, Asahina A, et al. A case of primary cutaneous anaplastic large cell lymphoma with variant anaplastic lymphoma kinase translocation. Br J Dermatol, 2004, 150:1202., DOI 10.1111/j. 1365-2133.2004.05987.x Chang Y-C, Chang C-H, Liu Y-T, Tsai K-B, Liu T-C, Lin Y-N. Spontaneous regression of a large-cell lymphoma in the conjunctiva and orbit. Ophthal Plast Reconstr Surg, 2004, 20:461., DOI 10.1097/01.IOP.0000144791. 05993.64 Winhoven SM,Murugesan S, Coulson IH. Solitary CD30+ anaplastic large cell lymphoma of the eyelid showing regression. Br J Ophthalmol, 2005, 89:385., DOI 10.1136/bjo.2004.042804 Sakuma H, Okabe M, Yokoi M, Eimoto T, Inagaki H. Spontaneous regression of intraoral mucosa-associated lymphoid tissue lymphoma: molecular study of a case. Pathol Int, 2006, 56:331., DOI 10.1111/j.1440-1827.2006.01967.x Madan V, Cox NH. Primary cutaneous peripheral T-cell lymphoma, unspecified, that completely regressed after skin biopsy. Br J Dermatol, 2007, 156:785., DOI 10.1111/j.1365-2133.2007.07751.x Ogden S, Coulson IH. B-cell lymphoma mimicking rhinophyma. Clin Exp Dermatol, 2008, 33:213., DOI 10.1111/j.1365-2230.2007.02476.x Daly R-M, Healy CM, Toner ME, Flint SR. Spontaneous regression of non- Hodgkin’s lymphoma in the oral cavity after incisional biopsy. Br J Oral Maxillofac Surg, 2008, 46:223., DOI 10.1016/j.bjoms.2007.03.010 Santiago-et-Sanchez-Mateos D, Hernandez-Martin A, Colmenero I, Mediero IG, Leon A, Torrelo A. Primary cutaneous anaplastic large cell lymphoma of the nasal tip in a child. Pediatr Dermatol, 2011, 28(5):570–5., DOI 10.1111/j. 1525-1470.2010.01245.x Tamas L, Sari E, Repassy G, Szabo P, Bagdi E, Krenacs L, et al. Spontaneous remission in localized diffuse large b-cell lymphoma. Pathol Oncol Res, 2011, 17:779., DOI 10.1007/s12253-011-9379-6 Buckner TW, Dunphy C, Fedoriw YD, van Deventer HW, Foster MC, Richards KL, et al. Complete spontaneous remission of diffuse large B-cell lymphoma of the maxillary sinus after concurrent infections. Clin Lymphoma Myeloma Leuk, 2012, 12:455., DOI 10.1016/j.clml.2012.06.007 Igawa T, Sato Y, Kawai H, Kondo E, Takeuchi M, Miyata-Takata T, et al. Spontaneous regression of plasmablastic lymphoma in an elderly human immunodeficiency virus, HIV)-negative patient. Diagn Pathol, 2015, 10: 421., DOI 10.1186/s13000-015-0421-y Gonzalez Fernandez D, Valdes Pineda F, Gomez Diez S, Vivanco Allende B. Primary cutaneous CD4+ small/medium-sized T-cell lymphoma with spontaneous regression after biopsy. Actas Dermo-Sifiliograficas, English Edition,, 2015, 106:767., DOI 10.1016/j.adengl.2015.09.015 Kaibuchi N, Okamoto T, Kataoka T, Kumasaka A, Ando T. A case of spontaneous regression of lymphoma in the mandibular gingiva after biopsy. Oral Maxillofac Surg Cases, 2015, 1:33., DOI 10.1016/j.omsc.2015. 06.002 Miyagawa F, Ogawa K, Asada H. A case of CD4+/CD8+ double-positive primary cutaneous anaplastic large cell lymphoma of the lip involving spontaneous regression after biopsy. Eur J Dermatol, 2017, 27:68., DOI 10. 1684/ejd.2016.2899 Snijder J, Mihyawi N, Frolov A, Ewton A, Rivero G. Spontaneous remission in diffuse large cell lymphoma: a case report. J Med Case Rep, 2019, 13., DOI 10. 1186/s13256-018-1937-z Pan Q, Luo Y, Cao X, Zhang Y, Li F. Spontaneous regression of clinically indolent lymphomas revealed by 18F-FDG PET/CT. Clin Nucl Med, 2019, 44: 321., DOI 10.1097/RLU.0000000000002457 Flatow-Trujillo L, Win K, Jencks A, Andritsos L, Arana Yi C. Spontaneous resolution of untreated diffuse large B-cell lymphoma of maxillary bone after incisional biopsy. Clin Case Rep, 2019, 12:305., DOI 10.1002/ccr3.2408 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 642433 EP 642441 DI 10.3389/pore.2021.642433 PG 9 ER PT J AU Maros, M Balla, P Micsik, T Sapi, Z Szendroi, M Wenz, H Groden, Ch Forsyth, R Picci, P Krenacs, T AF Maros, E. Mate Balla, Peter Micsik, Tamas Sapi, Zoltan Szendroi, Miklos Wenz, Holger Groden, Christoph Forsyth, G. Ramses Picci, Piero Krenacs, Tibor TI Cell Cycle Regulatory Protein Expression in Multinucleated Giant Cells of Giant Cell Tumor of Bone: do They Proliferate? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE giant cell tumor of bone; giant cells; Ki-67; cyclin D1; p53; p21 (CDKN1A) WAF1; cyclin G1 ID giant cell tumor of bone; giant cells; Ki-67; cyclin D1; p53; p21 (CDKN1A) WAF1; cyclin G1 AB Cells of the monocyte macrophage lineage form multinucleated giant cells (GCs) by fusion, which may express some cell cycle markers. By using a comprehensive marker set, here we looked for potential replication activities in GCs, and investigated whether these have diagnostic or clinical relevance in giant cell tumor of bone (GCTB). GC rich regions of 10 primary and 10 first recurrence GCTB cases were tested using immunohistochemistry in tissue microarrays. The nuclear positivity rate of the general proliferation marker, replication licensing, G1/S-phase, S/G2/M-phase, mitosis promoter, and cyclin dependent kinase (CDK) inhibitor reactions was analyzed in GCs. Concerning Ki67, moderate SP6 reaction was seen in many GC nuclei, while B56 and Mib1 positivity was rare, but the latter could be linked to more aggressive (p = 0.012) phenotype. Regular MCM6 reaction, as opposed to uncommon MCM2, suggested an initial DNA unwinding. Early replication course in GCs was also supported by widely detecting CDK4 and cyclin E, for the first time, and confirming cyclin D1 upregulation. However, post-G1-phase markers CDK2, cyclin A, geminin, topoisomerase-2a, aurora kinase A, and phospho-histone H3 were rare or missing. These were likely silenced by upregulated CDK inhibitors p15INK4b, p16INK4a, p27KIP1, p53 through its effector p21WAF1 and possibly cyclin G1, consistent with the prevention of DNA replication. In conclusion, the upregulation of known and several novel cell cycle progression markers detected here clearly verify early replication activities in GCs, which are controlled by cell cycle arresting CDK inhibitors at G1 phase, and support the functional maturation of GCs in GCTB. C1 [Maros, E. Mate] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Balla, Peter] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Micsik, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Szendroi, Miklos] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Wenz, Holger] University of Heidelberg, Medical Faculty Mannheim, Department of NeuroradiologyMannheim, Germany. [Groden, Christoph] University of Heidelberg, Medical Faculty Mannheim, Department of NeuroradiologyMannheim, Germany. [Forsyth, G. Ramses] University Ziekenhuis, Department of Anatomic Pathology and Experimental PathologyBrussels, Belgium. [Picci, Piero] Istituti Ortopedici Rizzoli, Laboratorio di Ricerca OncologicaBologna, Italy. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Krenacs, T (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM krenacst@gmail.com CR Chambers TJMultinucleate Giant Cells. J Pathol, 1978, 126(3):125–48., DOI 10. 1002/path.1711260302 Ariizumi T, Ogose A, Kawashima H, Hotta T, Umezu H, and Endo NMultinucleation Followed by an Acytokinetic Cell Division in Myxofibrosarcoma with Giant Cell Proliferation. J Exp Clin Cancer Res, 2009, 28:44., DOI 10.1186/1756-9966-28-44 Vignery AMacrophage Fusion. J Exp Med, 2005, 202(3):337–40., DOI 10.1084/ jem.20051123 Maros ME, Schnaidt S, Balla P, Kelemen Z, Sapi Z, Szendroi M, et al.In situ cell Cycle Analysis in Giant Cell Tumor of Bone Reveals Patients with Elevated Risk of Reduced Progression-free Survival. Bone, 2019, 127:188–98., DOI 10. 1016/j.bone.2019.06.022 Kauzman A, Li SQ, Bradley G, Bell RS, Wunder JS, and Kandel RCyclin Alterations in Giant Cell Tumor of Bone. Mod Pathol, 2003, 16(3):210–8., DOI 10.1097/01.MP.0000057235.65327.40 Kandel R, Li SQ, Bell R, Wunder J, Ferguson P, Kauzman A, et al.Cyclin D1 and P21 Is Elevated in the Giant Cells of Giant Cell Tumors. J Orthop Res, 2006, 24(3):428–37., DOI 10.1002/jor.20036 Maggiani F, Forsyth R, Hogendoorn PCW, Krenacs T, and Athanasou NAThe Immunophenotype of Osteoclasts and Macrophage Polykaryons. J Clin Pathol, 2011, 64(8):701–5., DOI 10.1136/jcp.2011.090852 Behjati S, Tarpey PS, Presneau N, Scheipl S, Pillay N, Van Loo P, et al.Distinct H3F3A and H3F3B Driver Mutations Define Chondroblastoma and Giant Cell Tumor of Bone. Nat Genet, 2013, 45(12):1479–82., DOI 10.1038/ng.2814 Szendr}oi MGiant-cell Tumour of Bone, GCT). In: G Bentley, editor. European Surgical Orthopaedics and Traumatology. Berlin, Germany: Springer, 2014). p. 4037–54. Cowan RW, and Singh GGiant Cell Tumor of Bone: A Basic Science Perspective. Bone, 2013, 52(1):238–46., DOI 10.1016/j.bone.2012.10.002 Knowles HJ, and Athanasou NAAcute Hypoxia and Osteoclast Activity: a Balance between Enhanced Resorption and Increased Apoptosis. J Pathol, 2009, 218(2):256–64., DOI 10.1002/path.2534 Knowles HJ, and Athanasou NACanonical and Non-canonical Pathways of Osteoclast Formation. Histol Histopathol, 2009, 24:337., DOI 10.14670/HH- 24.337 Balla P, Moskovszky L, Sapi Z, Forsyth R, Knowles H, Athanasou NA, et al.Epidermal Growth Factor Receptor Signalling Contributes to Osteoblastic Stromal Cell Proliferation, Osteoclastogenesis and Disease Progression in Giant Cell Tumour of Bone. Histopathology, 2011, 59(3): 376–89., DOI 10.1111/j.1365-2559.2011.03948.x Balla P, Maros ME, Barna G, Antal I, Papp G, Sapi Z, et al.Prognostic Impact of Reduced Connexin43 Expression and Gap Junction Coupling of Neoplastic Stromal Cells in Giant Cell Tumor of Bone. PLoS One, 2015, 10(5):e0125316., DOI 10.1371/journal.pone.0125316 Satyanarayana A, and Kaldis PMammalian Cell-Cycle Regulation: Several Cdks, Numerous Cyclins and Diverse Compensatory Mechanisms. Oncogene, 2009, 28(33):2925–39., DOI 10.1038/onc.2009.170 Williams GH, and Stoeber KThe Cell Cycle and Cancer. J Pathol, 2012, 226(2): 352–64., DOI 10.1002/path.3022 Kimura SH, and Nojima HCyclin G1 Associates with MDM2 and Regulates Accumulation and Degradation of P53 Protein. Genes Cells, 2002, 7(8): 869–80., DOI 10.1046/j.1365-2443.2002.00564.x Gordon E, Ravicz J, Liu S, Chawla S, and Hall FCell Cycle Checkpoint Control: The Cyclin G1/Mdm2/p53 axis Emerges as a Strategic Target for Broad-Spectrum Cancer Gene Therapy - A Review of Molecular Mechanisms for Oncologists. Mol Clin Onc, 2018, 9(2):115–34., DOI 10. 3892/mco.2018.1657 Wenz H, Maros ME, Meyer M, Forster A, Haubenreisser H, Kurth S, et al.Image Quality of 3rd Generation Spiral Cranial Dual-Source CT in Combination with an Advanced Model Iterative Reconstruction Technique: A Prospective Intra-individual Comparison Study to Standard Sequential Cranial CT Using Identical Radiation Dose. PLoS One, 2015, 10(8): e0136054., DOI 10.1371/journal.pone.0136054 Knowles H, and Athanasou NHypoxia-inducible Factor Is Expressed in Giant Cell Tumour of Bone and Mediates Paracrine Effects of Hypoxia on Monocyte-Osteoclast Differentiation via Induction of VEGF. J Pathol, 2008, 215(1):56–66., DOI 10.1002/path.2319 Knowles HJ, and Athanasou NACanonical and Non-canonical Pathways of Osteoclast Formation. Histol Histopathol, 2009, 24(3):337–46., DOI 10.14670/ HH-24.337 Choi JW, Lee JH, Kim YS, and Kim YSFrequent Upregulation of Cyclin D1 and p16INK4a Expression with Low Ki-67 Scores in Multinucleated Giant Cells. Pathobiology, 2011, 78(4):233–7., DOI 10.1159/000327359 Matsubayashi S, Nakashima M, Kumagai K, Egashira M, Naruke Y, Kondo H, et al.Immunohistochemical Analyses of β-catenin and Cyclin D1 Expression in Giant Cell Tumor of Bone, GCTB): A Possible Role of Wnt Pathway in GCTB Tumorigenesis. Pathol - Res Pract, 2009, 205(9):626–33., DOI 10.1016/j.prp. 2009.02.011 Lujic N, Sopta J, Kovacevic R, Stevanovic V, and Davidovic RRecurrence of Giant Cell Tumour of Bone: Role of P53, Cyclin D1, β-catenin and Ki67. Int Orthopaedics, Sicot,, 2016, 40(11):2393–9., DOI 10.1007/s00264-016-3292-2 Sun X, and Kaufman PDKi-67: More Than a Proliferation Marker. Chromosoma, 2018, 127(2):175–86., DOI 10.1007/s00412-018-0659-8 Sales Gil R, and Vagnarelli PKi-67: More Hidden behind a ’Classic Proliferation Marker’. Trends Biochem Sci, 2018, 43(10):747–8., DOI 10. 1016/j.tibs.2018.08.004 Ismail FW, Shamsudin AM, Wan Z, Daud SM, and Samarendra MSKi-67 Immuno-Histochemistry Index in Stage III Giant Cell Tumor of the Bone. J Exp Clin Cancer Res, 2010, 29:25., DOI 10.1186/1756-9966-29-25 Miller I, Min M, Yang C, Tian C, Gookin S, Carter D, et al.Ki67 Is a Graded Rather Than a Binary Marker of Proliferation versus Quiescence. Cell Rep, 2018, 24(5):1105–12.e5., DOI 10.1016/j.celrep.2018.06.110 Vijayraghavan S, and Schwacha AThe Eukaryotic Mcm2-7 Replicative Helicase. Subcell Biochem, 2012, 62:113–34., DOI 10.1007/978-94-007-4572-8_7 Malumbres MCyclin-dependent Kinases. Genome Biol, 2014, 15(6):122., DOI 10.1186/gb4184 Pestell RGNew Roles of Cyclin D1. Am J Pathol, 2013, 183(1):3–9., DOI 10. 1016/j.ajpath.2013.03.001 Romero-Pozuelo J, Figlia G, Kaya O, Martin-Villalba A, and Teleman AACdk4 and Cdk6 Couple the Cell-Cycle Machinery to Cell Growth via mTORC1. Cell Rep, 2020, 31(2):107504., DOI 10.1016/j.celrep.2020.03.068 Soonpaa MH, Koh GY, Pajak L, Jing S, Wang H, Franklin MT, et al.Cyclin D1 Overexpression Promotes Cardiomyocyte DNA Synthesis and Multinucleation in Transgenic Mice. J Clin Invest, 1997, 99(11):2644–54., DOI 10.1172/JCI119453 Palazon LS, Davies TJ, and Gardner RLTranslational Inhibition of Cyclin B1 and Appearance of Cyclin D1 Very Early in the Differentiation of Mouse Trophoblast Giant Cells. Mol Hum Reprod, 1998, 4(11):1013–20., DOI 10.1093/molehr/4.11.1013 Georgakilas AG, Martin OA, and Bonner WMp21: A Two-Faced Genome Guardian. Trends MolMed, 2017, 23(4):310–9., DOI 10.1016/j.molmed.2017.02.001 Cheng M, Olivier P, Diehl JA, Fero M, Roussel MF, Roberts JM, et al.The p21Cip1 and p27Kip1 CDK `inhibitors’ Are Essential Activators of Cyclin D-dependent Kinases in Murine Fibroblasts. EMBO J, 1999, 18(6):1571–83., DOI 10.1093/emboj/18.6.1571 Alt JR, Cleveland JL, Hannink M, and Diehl JAPhosphorylation-dependent Regulation of Cyclin D1 Nuclear Export and Cyclin D1-dependent Cellular Transformation. Genes Dev, 2000, 14(24):3102–14., DOI 10.1101/gad.854900 Alt JR, Gladden AB, and Diehl JAp21Cip1 Promotes Cyclin D1 Nuclear Accumulation via Direct Inhibition of Nuclear Export. J Biol Chem, 2002, 277(10):8517–23., DOI 10.1074/jbc.M108867200 Hafner A, Bulyk ML, Jambhekar A, and Lahav GThe Multiple Mechanisms that Regulate P53 Activity and Cell Fate. Nat Rev Mol Cell Biol, 2019, 20(4): 199–210., DOI 10.1038/s41580-019-0110-x Yalcinkaya U, Ugras N, Kabul S, Ocakoglu G, and Bilgen MSPrognostic Value of P53 Protein Expression in Giant Cell Tumor of Bone. pjp, 2015, 4(4): 389–96., DOI 10.5114/pjp.2015.57252 Ganem NJ, and Pellman DLimiting the Proliferation of Polyploid Cells. Cell, 2007, 131(3):437–40., DOI 10.1016/j.cell.2007.10.024 Rieder CL, and Maiato HStuck in Division or Passing through. Dev Cell, 2004, 7(5):637–51., DOI 10.1016/j.devcel.2004.09.002 Wander SA, Zhao D, and Slingerland JMp27: A Barometer of Signaling Deregulation and Potential Predictor of Response to Targeted Therapies: Figure 1. Clin Cancer Res, 2011, 17(1):12–8., DOI 10.1158/1078-0432.Ccr-10- 0752 Sankar U, Patel K, Rosol TJ, and Ostrowski MCRANKL Coordinates Cell Cycle Withdrawal and Differentiation in Osteoclasts Through the Cyclin- Dependent Kinase Inhibitors p27KIP1and p21CIP1. J Bone Miner Res, 2004, 19(8):1339–48., DOI 10.1359/JBMR.040321 Lindeman JHN, Hanemaaijer R, Mulder A, Dijkstra PDS, Szuhai K, Bromme D, et al.Cathepsin K Is the Principal Protease in Giant Cell Tumor of Bone. Am J Pathol, 2004, 165(2):593–600., DOI 10.1016/S0002-9440(10)63323-8 Okamoto K, Li H, Jensen MR, Zhang T, Taya Y, Thorgeirsson SS, et al.Cyclin G Recruits PP2A to Dephosphorylate Mdm2. Mol Cell, 2002, 9(4):761–71., DOI 10.1016/s1097-2765(02)00504-x Sherr CJ, and Roberts JMCDK Inhibitors: Positive and Negative Regulators of G1-phase Progression. Genes Dev, 1999, 13(12):1501–12., DOI 10.1101/gad.13. 12.1501 Carnero A, Hudson JD, Hannon GJ, and Beach DHLoss-of-function Genetics in Mammalian Cells: the P53 Tumor Suppressor Model. Nucleic Acids Res, 2000, 28(11):2234–41., DOI 10.1093/nar/28.11.2234 Hudson JD, Shoaibi MA, Maestro R, Carnero A, Hannon GJ, and Beach DHA Proinflammatory Cytokine Inhibits P53 Tumor Suppressor Activity. J Exp Med, 1999, 190(10):1375–82., DOI 10.1084/jem.190.10.1375 Dimri GP, Hara E, and Campisi JRegulation of Two E2F-Related Genes in Presenescent and Senescent Human Fibroblasts. J Biol Chem, 1994, 269(23): 16180–6., DOI 10.1016/s0021-9258(17)33990-x Lari R, Kitchener PD, and Hamilton JAThe Proliferative Human Monocyte Subpopulation Contains Osteoclast Precursors. Arthritis Res Ther, 2009, 11(1):R23., DOI 10.1186/ar2616 Forsyth RG, De Boeck G, Baelde JJ, Taminiau AH, Uyttendaele D, Roels H, et al.CD33+ CD14− Phenotype Is Characteristic of Multinuclear Osteoclastlike Cells in Giant Cell Tumor of Bone. J Bone Mineral Res, 2009, 24(1):70–7., DOI 10.1359/jbmr.080905 Orriss IR, and Arnett TRRodent Osteoclast Cultures. Methods Mol Biol, 2012, 816:103–17., DOI 10.1007/978-1-61779-415-5_8 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 643146 EP 643156 DI 10.3389/pore.2021.643146 PG 11 ER PT J AU Zhang, Y Zhou, B Sun, J He, Q Zhao, Y AF Zhang, Ye Zhou, Bo Sun, Jingjing He, Qun Zhao, Yujie TI Knockdown of GPSM1 Inhibits the Proliferation and Promotes the Apoptosis of B-Cell Acute Lymphoblastic Leukemia Cells by Suppressing the ADCY6-RAPGEF3-JNK Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE B-cell acute lymphoblastic leukemia; GPSM1; ADCY6; RAPGEF3; JNK ID B-cell acute lymphoblastic leukemia; GPSM1; ADCY6; RAPGEF3; JNK AB B-cell acute lymphoblastic leukemia (B-ALL) is the common type of blood cancer. Although the remission rate has increased, the current treatment options for B-ALL are usually related to adverse reactions and recurrence, so it is necessary to find other treatment options. G protein signaling modulator 1 (GPSM1) is one of several factors that affect the basic activity of the G protein signaling system, but its role in B-ALL has not yet been clarified. In this study, we analyzed the expression of GPSM1 in the Oncomine database and found that the GPSM1 levels were higher in B-ALL cells than in peripheral blood mononuclear cells (PBMCs). Analyses of the Gene Expression Profiling Interactive Analysis (GEPIA) demonstrated that patients with high GPSM1 levels had shorter survival times than those with low levels. Additionally, gene set enrichment analysis (GSEA) suggested that GPSM1 was positively correlated with proliferation, G protein-coupled receptor (GPCR) ligand binding, Gαs signaling and calcium signaling pathways. In further experiments, GPSM1 was found to be highly expressed in Acute lymphoblastic leukemia (ALL) cell lines, and downregulation of GPSM1 inhibited proliferation and promoted cell cycle arrest and apoptosis in BALL-1 and Reh cells. Moreover, knockdown of GPSM1 suppressed ADCY6 and RAPGEF3 expression in BALL-1 and Reh cells. Furthermore, we reported that GPSM1 regulated JNK expression via ADCY6-RAPGEF3. The present study demonstrates that GPSM1 promotes tumor growth in BALL-1 and Reh cells by modulating ADCY6-RAPGEF3-JNK signaling. C1 [Zhang, Ye] China Medical University, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of EducationShenyang, China. [Zhou, Bo] China Medical University, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of EducationShenyang, China. [Sun, Jingjing] China Medical University, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of EducationShenyang, China. [He, Qun] China Medical University, School of Life Sciences, Department of BioinformaticsShenyang, China. [Zhao, Yujie] China Medical University, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of EducationShenyang, China. RP Zhao, Y (reprint author), China Medical University, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Shenyang, China. EM yjzhao_biocell@163.com CR Nathwani SM, Greene LM, Butini S, Campiani G, Williams DC, Samali A, et al. The pyrrolo-1,5-benzoxazepine, PBOX-15, enhances TRAIL-induced apoptosis by upregulation of DR5 and downregulation of core cell survival proteins in acute lymphoblastic leukaemia cells. Int J Oncol, 2016, 49(1):74–88., DOI 10.3892/ijo.2016.3518 Gholman RR, Felemban EH, El Meligy OA. Dental rehabilitation of a child with acute lymphocytic leukemia: a case report. Int J Clin Pediatr dentistry, 2019, 12(6):582–6., DOI 10.5005/jp-journals-10005-1664 Sriram K, Insel PA. G protein-coupled receptors as targets for approved drugs: how many targets and how many drugs? Mol Pharmacol, 2018, 93(4):251–8., DOI 10.1124/mol.117.111062 O’Hayre M, Degese MS, Gutkind JS. Novel insights into G protein and G protein-coupled receptor signaling in cancer. Curr Opin Cell Biol, 2014, 27: 126–35., DOI 10.1016/j.ceb.2014.01.005 Bond J, Domaschenz R, Roman-Trufero M, Sabbattini P, Ferreiros-Vidal I, Gerrard G, et al. Direct interaction of Ikaros and Foxp1 modulates expression of the G protein-coupled receptor G2A in B-lymphocytes and acute lymphoblastic leukemia. Oncotarget, 2016, 7(40):65923–36., DOI 10.18632/ oncotarget.11688 Maiga A, Lemieux S, Pabst C, Lavallee V-P, Bouvier M, Sauvageau G, et al. Transcriptome analysis reveals that G protein-coupled receptors are potential diagnostic markers or therapeutic targets in acute myeloid leukemia. Blood, 2015, 126(23):3855., DOI 10.1182/blood.V126.23.3855.3855 Tyndall JD, Sandilya R. GPCR agonists and antagonists in the clinic. Med Chem, 2005, 1(4):405–21., DOI 10.2174/1573406054368675 Mahadevan D, Choi J, Cooke L, Simons B, Riley C, Klinkhammer T, et al. Gene expression and serum cytokine profiling of low stage CLL identify WNT/PCP, flt-3L/flt-3 and CXCL9/CXCR3 as regulators of cell proliferation, survival and migration. Hum Genomics Proteomics, 2009, 2009:453634., DOI 10.4061/2009/ 453634 Nairismagi ML, Tan J, Lim JQ, Nagarajan S, Ng CC, Rajasegaran V, et al. JAKSTAT and G-protein-coupled receptor signaling pathways are frequently altered in epitheliotropic intestinal T-cell lymphoma. Leukemia, 2016, 30(6): 1311–9., DOI 10.1038/leu.2016.13 Oner SS, Maher EM, Gabay M, Tall GG, Blumer JB, Lanier SM. Regulation of the G-protein regulatory-Gαi signaling complex by nonreceptor guanine nucleotide exchange factors. J Biol Chem, 2013, 288(5):3003–15., DOI 10. 1074/jbc.M112.418467 Blumer JB, Lanier SM. Activators of G protein signaling exhibit broad functionality and define a distinct core signaling triad. Mol Pharmacol, 2014, 85(3):388–96., DOI 10.1124/mol.113.090068 Pattingre S, De Vries L, Bauvy C, Chantret I, Cluzeaud F, Ogier-Denis E, et al. The G-protein regulator AGS3 controls an early event during macroautophagy in human intestinal HT-29 cells. J Biol Chem, 2003, 278(23):20995–1002., DOI 10.1074/jbc.M300917200 Sanada K, Tsai LH. G protein betagamma subunits and AGS3 control spindle orientation and asymmetric cell fate of cerebral cortical progenitors. Cell, 2005, 122(1):119–31., DOI 10.1016/j.cell.2005.05.009 Vural A, Al-Khodor S, Cheung GY, Shi CS, Srinivasan L, McQuiston TJ, et al. Activator of G-protein signaling 3-induced lysosomal biogenesis limits macrophage intracellular bacterial infection. J Immunol, 1950, 196(2): 846–56., DOI 10.4049/jimmunol.1501595 Oner SS, Vural A, Lanier SM. Translocation of activator of G-protein signaling 3 to the Golgi apparatus in response to receptor activation and its effect on the trans-Golgi network. J Biol Chem, 2013, 288(33):24091–103., DOI 10.1074/jbc. M112.444505 Bowers MS, Hopf FW, Chou JK, Guillory AM, Chang SJ, Janak PH, et al. Nucleus accumbens AGS3 expression drives ethanol seeking through G betagamma. Proc Natl Acad Sci USA, 2008, 105(34):12533–8., DOI 10.1073/ pnas.0706999105 Kwon M, Pavlov TS, Nozu K, Rasmussen SA, Ilatovskaya DV, Lerch-Gaggl A, et al. G-protein signaling modulator 1 deficiency accelerates cystic disease in an orthologous mouse model of autosomal dominant polycystic kidney disease. Proc Natl Acad Sci USA, 2012, 109(52):21462–7., DOI 10.1073/pnas. 1216830110 Branham-O’Connor M, Robichaux WG, 3rd, Zhang XK, Cho H, Kehrl JH, Lanier SM, et al. Defective chemokine signal integration in leukocytes lacking activator of G protein signaling 3, AGS3). J Biol Chem, 2014, 289(15): 10738–47., DOI 10.1074/jbc.M113.515031 Rhodes DR, Kalyana-Sundaram S, Mahavisno V, Varambally R, Yu J, Briggs BB, et al. Oncomine 3.0: genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia, 2007, 9(2):166–80., DOI 10. 1593/neo.07112 Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res, 2017, 45(W1):W98–102., DOI 10.1093/nar/gkx247 Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez JC, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics, 2011, 12:77., DOI 10.1186/1471-2105-12-77 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA, 2005, 102(43):15545–50., DOI 10.1073/pnas.0506580102 HallsML, CooperDMF. Adenylyl cyclase signalling complexes–pharmacological challenges and opportunities. Pharmacol Ther, 2017, 172:171–80., DOI 10.1016/j. pharmthera.2017.01.001 Bos JL. Epac proteins: multi-purpose cAMP targets. Trends Biochem Sci, 2006, 31(12):680–6., DOI 10.1016/j.tibs.2006.10.002 Schwindinger WF, Robishaw JD. Heterotrimeric G-protein betagammadimers in growth and differentiation. Oncogene, 2001, 20(13):1653–60., DOI 10.1038/sj.onc.1204181 Hammouda MB, Ford AE, Liu Y, Zhang JY. The JNK signaling pathway in inflammatory skin disorders and cancer. Cells, 2020, 9(4)., DOI 10.3390/ cells9040857 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10. 3322/caac.21492 Shao S, Huang X, Wang Y, He S, Xu X, Zhu X, et al. A role for activator of G-protein signaling 3, AGS3, in multiple myeloma. Int J Hematol, 2014, 99(1):57–68., DOI 10.1007/s12185-013-1484-8 Adekoya TO, Smith N, Aladeniyi T, Blumer JB, Chen XL, Richardson RM. Activator of G protein signaling 3 modulates prostate tumor development and progression. Carcinogenesis, 2019, 40(12):1504–13., DOI 10.1093/carcin/ bgz076 Shi H, Ren H, Yang X, Zhu H, Yao L, Hang Q, et al. Overexpression of activator of G-protein signaling 3 decreases the proliferation of esophageal squamous cell carcinoma. Pathol Res Pract, 2015, 211(6):449–55., DOI 10.1016/j.prp.2014.12.016 Regner KR, Nozu K, Lanier SM, Blumer JB, Avner ED, Sweeney WE, Jr, et al. Loss of activator of G-protein signaling 3 impairs renal tubular regeneration following acute kidney injury in rodents. FASEB J, 2011, 25(6):1844–55., DOI 10.1096/fj.10-169797 Nadella R, Blumer JB, Jia G, Kwon M, Akbulut T, Qian F, et al. Activator of G protein signaling 3 promotes epithelial cell proliferation in PKD. J Am Soc Nephrol, 2010, 21(8):1275–80., DOI 10.1681/ASN.2009121224 Keri KC, Regner KR, Dall AT, Park F. Urinary exosomal expression of activator of G protein signaling 3 in polycystic kidney disease. BMC Res Notes, 2018, 11(1):359., DOI 10.1186/s13104-018-3467-6 Hirsch GE, Parisi MM, Martins LA, Andrade CM, Barbe-Tuana FM, Guma FT. γ-oryzanol reduces caveolin-1 and PCGEM1 expression, markers of aggressiveness in prostate cancer cell lines. Prostate, 2015, 75(8):783–97., DOI 10.1002/pros.22960 Wei W, Qi L, Feihui Z, Zhihua Y, Yunfeng W, Ting L, et al. Increased expression of AGS3 in rat brain cortex after traumatic brain injury. J Neurosci Res, 2013, 91(5):726–36., DOI 10.1002/jnr.23195 Patra C, Foster K, Corley JE, Dimri M, Brady MF. Biochemistry, cAMP. In: StatPearls [Internet]. Treasure Island, FL): StatPearls Publishing, 2020). La Marca JE, Richardson HE Two-faced: roles of JNK signalling during tumourigenesis in the Drosophila model. Front Cell Dev Biol, 2020, 8:42., DOI 10.3389/fcell.2020.00042 Leventaki V, Drakos E, Karanikou M, Psatha K, Lin P, Schlette E, et al. c-JUN N-terminal kinase, JNK, is activated and contributes to tumor cell proliferation in classical Hodgkin lymphoma. Hum Pathol, 2014, 45(3): 565–72., DOI 10.1016/j.humpath.2013.10.024 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 643376 EP 643386 DI 10.3389/pore.2021.643376 PG 11 ER PT J AU Szasz, R Telek, B Illes, AF Szasz, Robert Telek, Bela Illes, Arpad TI Fludarabine-Cyclophosphamide- Rituximab Treatment in Chronic Lymphocytic Leukemia, Focusing on Long Term Cytopenias Before and After the Era of Targeted Therapies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE treatment; chronic lymphocytic leukemia; fludarabine; cyclophosphamide; rituximab; cytopenia ID treatment; chronic lymphocytic leukemia; fludarabine; cyclophosphamide; rituximab; cytopenia AB The widespread application of fludarabine, cyclophosphamide, and rituximab combination is limited due to its toxicity, particularly the prolonged cytopenias. The study aimed to compare the prolonged cytopenias depending on fitness and report real-life data on dose reduction measures and efficacy. According to our database, 120 and 14 patients were treated with FCR between 2011 and 2015 and between 2016 and 2019. Out of the first cohort, 34 patients were treated in subsequent lines. The complete and partial remission rate after first-line treatment was 79%, 16% in the first cohort and 86%, 14% in the second cohort, respectively; and 47%, 35% after non first-line treatment. Based on today’s standards, only 37.5% of the patients were fit for FCR. The frequency of persistent cytopenia was 14%, and it was significantly associated with fitness (χ2 (1) = 6.001, p = 0.014 for all patients). The small number of FCR treated patients after 2016 shows how the availability of targeted therapies, mostly ibrutinib, in later lines changed the first-line choice. Recently, it is recommended first-line for fit patients with mutated IGHV and no TP53 aberrations. With this narrow indication, a decrease in the frequency of persistent cytopenias is predicted. C1 [Szasz, Robert] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Telek, Bela] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. RP Szasz, R (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM szaszr@med.unideb.hu CR Keating MJ, O’Brien S, Albitar M, Lerner S, Plunkett W, Giles F, et al. Early Results of a Chemoimmunotherapy Regimen of Fludarabine, Cyclophosphamide, and Rituximab as Initial Therapy for Chronic Lymphocytic Leukemia. Jco, 2005, 23(18):4079–88., DOI 10.1200/JCO.2005. 12.051 Hallek M, Fischer K, Fingerle-Rowson G, Fink A, Busch R, Mayer J, et al. Addition of Rituximab to Fludarabine and Cyclophosphamide in Patients with Chronic Lymphocytic Leukaemia: a Randomised, Open-Label, Phase 3 Trial. The Lancet, 2010, 376(9747):1164–74., DOI 10.1016/S0140-6736(10)61381-5 Fischer K, Bahlo J, Fink AM, Goede V, Herling CD, Cramer P, et al. Long-term Remissions after FCR Chemoimmunotherapy in Previously Untreated Patients with CLL: Updated Results of the CLL8 Trial. Blood, 2016, 127(2):208–15., DOI 10.1182/blood-2015-06-651125 Gozzetti A, Candi V, Fabbri A, Schiattone L, Cencini E, Lauria F, et al. Chemoimmunotherapy with Oral Low-Dose Fludarabine, Cyclophosphamide and Rituximab, Old-FCR, as Treatment for Elderly Patients with Chronic Lymphocytic Leukaemia. Leuk Res, 2014, 38(8):891–5., DOI 10.1016/j.leukres. 2014.05.016 Dartigeas C, Van Den Neste E, Berthou C, Maisonneuve H, Leprˆetre S, Dilhuydy M-S, et al. Evaluating Abbreviated Induction with Fludarabine, Cyclophosphamide, and Dose-Dense Rituximab in Elderly Patients with Chronic Lymphocytic Leukemia. Leuk Lymphoma, 2016, 57(2):328–34., DOI 10.3109/10428194.2015.1063139 Foon KA, Boyiadzis M, Land SR, Marks S, Raptis A, Pietragallo L, et al. Chemoimmunotherapy with Low-Dose Fludarabine and Cyclophosphamide and High Dose Rituximab in Previously Untreated Patients with Chronic Lymphocytic Leukemia. Jco, 2009, 27(4):498–503., DOI 10.1200/jco.2008.17. 2619 Garcia-Barchino MJ, Sarasquete ME, Panizo C, Morscio J, Martinez A, Alcoceba M, et al. Richter Transformation Driven by Epstein-Barr Virus Reactivation during Therapy-Related Immunosuppression in Chronic Lymphocytic Leukaemia. J Pathol, 2018, 245(1):61–73., DOI 10.1002/path.5060 Innocenti I, Rossi D, Trape G, Autore F, Larocca LM, Gomes V, et al. Clinical, Pathological, and Biological Characterization of Richter Syndrome Developing after Ibrutinib Treatment for Relapsed Chronic Lymphocytic Leukemia. Hematological Oncol, 2018, 36(3):600–3., DOI 10.1002/hon.2502 Tam CS, O’Brien S, Wierda W, Kantarjian H, Wen S, Do K-A, et al. Long-term Results of the Fludarabine, Cyclophosphamide, and Rituximab Regimen as Initial Therapy of Chronic Lymphocytic Leukemia. Blood, 2008, 112(4): 975–80., DOI 10.1182/blood-2008-02-140582 Thompson PA, Tam CS, O’Brien SM,Wierda WG, Stingo F, Plunkett W, et al. Fludarabine, Cyclophosphamide, and Rituximab Treatment Achieves Long- Term Disease-free Survival in IGHV-Mutated Chronic Lymphocytic Leukemia. Blood, 2016, 127(3):303–9., DOI 10.1182/blood-2015-09-667675 Strati P, Wierda W, Burger J, Ferrajoli A, Tam C, Lerner S, et al. Myelosuppression after Frontline Fludarabine, Cyclophosphamide, and Rituximab in Patients with Chronic Lymphocytic Leukemia. Cancer, 2013, 119(21):3805–11., DOI 10.1002/cncr.28318 Herishanu Y, Tadmor T, Braester A, Bairey O, Aviv A, Rahimi-Levene N, et al. Low-dose Fludarabine and Cyclophosphamide Combined with Standard Dose Rituximab, LD-FCR, Is an Effective and Safe Regimen for Elderly Untreated Patients with Chronic Lymphocytic Leukemia: The Israeli CLL Study Group Experience. Hematological Oncol, 2019, 37(2):185–92., DOI 10.1002/hon.2580 Goede V, Fischer K, Busch R, Engelke A, Eichhorst B, Wendtner CM, et al. Obinutuzumab Plus Chlorambucil in Patients with CLL and Coexisting Conditions. N Engl J Med, 2014, 370(12):1101–10., DOI 10.1056/ NEJMoa1313984 Hillmen P, Gribben JG, Follows GA, Milligan D, Sayala HA, Moreton P, et al. Rituximab Plus Chlorambucil as First-Line Treatment for Chronic Lymphocytic Leukemia: Final Analysis of an Open-Label Phase II Study. Jco, 2014, 32(12):1236–41., DOI 10.1200/JCO.2013.49.6547 Foa R, Del Giudice I, Cuneo A, Del Poeta G, Ciolli S, Di Raimondo F, et al. Chlorambucil Plus Rituximab with or without Maintenance Rituximab as First-Line Treatment for Elderly Chronic Lymphocytic Leukemia Patients. Am J Hematol, 2014, 89(5):480–6., DOI 10.1002/ajh.23668 Jain N, and O’Brien S Initial Treatment of CLL: Integrating Biology and Functional Status. Blood, 2015, 126(4):463–70., DOI 10.1182/blood-2015-04- 585067 Fischer K, Cramer P, Busch R, Bottcher S, Bahlo J, Schubert J, et al. Bendamustine in Combination with Rituximab for Previously Untreated Patients with Chronic Lymphocytic Leukemia: a Multicenter Phase II Trial of the German Chronic Lymphocytic Leukemia Study Group. Jco, 2012, 30(26):3209–16., DOI 10.1200/JCO.2011.39.2688 Fischer K, Al-Sawaf O, Bahlo J, Fink A-M, Tandon M, Dixon M, et al. Venetoclax and Obinutuzumab in Patients with CLL and Coexisting Conditions. N Engl J Med, 2019, 380(23):2225–36., DOI 10.1056/ nejmoa1815281 Tausch E, Schneider C, Robrecht S, Zhang C, Dolnik A, Bloehdorn J, et al. Prognostic and Predictive Impact of Genetic Markers in Patients with CLL Treated with Obinutuzumab and Venetoclax. Blood, 2020, 135(26):2402–12., DOI 10.1182/blood.2019004492 Barr P, Robak T, Owen CJ, Tedeschi A, Bairey O, Bartlett NL, et al. Updated Efficacy and Safety from the Phase 3 Resonate-2 Study: Ibrutinib as First-Line Treatment Option in Patients 65 Years and Older with Chronic Lymphocytic Leukemia/Small Lymphocytic Leukemia. Blood, 2016, 128(22):234., DOI 10. 1182/blood.v128.22.234.234 Burger JA, Tedeschi A, Barr PM, Robak T, Owen C, Ghia P, et al. Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia. N Engl J Med, 2015, 373(25):2425–37., DOI 10.1056/NEJMoa1509388 Byrd JC, Brown JR, O’Brien S, Barrientos JC, Kay NE, Reddy NM, et al. Ibrutinib versus Ofatumumab in Previously Treated Chronic Lymphoid Leukemia. N Engl J Med, 2014, 371(3):213–23., DOI 10.1056/NEJMoa1400376 Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G, Dohner H, et al. Guidelines for the Diagnosis and Treatment of Chronic Lymphocytic Leukemia: a Report from the International Workshop on Chronic Lymphocytic Leukemia Updating the National Cancer Institute-Working Group 1996 Guidelines. Blood, 2008, 111(12):5446–56., DOI 10.1182/blood- 2007-06-093906 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609742 EP 1609748 DI 10.3389/pore.2021.1609742 PG 7 ER PT J AU Huang, L Li, Y Du, J Li, H Lu, M Wang, Y Zhou, W Wang, W Wu, H AF Huang, Liangliang Li, Yujie Du, Jun Li, Heng Lu, Mengmeng Wang, Yuting Zhou, Wenchao Wang, Wei Wu, Haibo TI The Prognostic Value of Retraction Clefts in Chinese Invasive Breast Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; clinicopathological features; invasive breast carcinoma; retraction clefts; Chinese ID prognosis; clinicopathological features; invasive breast carcinoma; retraction clefts; Chinese AB Some studies reported the correlation between retraction clefts (RCs) and the clinicopathological features as well as prognosis in invasive breast carcinoma. However, limited number of investigations have been done and controversial results were reported. Larger population studies around the world might help to provide more accurate and comprehensive information. Thus, we examined the correlation between the extent of RCs and the clinicopathological features as well as the prognosis in 541 invasive breast carcinoma samples from Central China in this study. The statistical analyses were performed with the Pearson χ2 tests and univariate Cox proportional hazards regression assays. Compared with other studies, lower RCs occurrence rate (15.5%) was observed in Chinese breast cancer patients and opposite association between the presence of RCs and lymph nodes metastasis was identified, in which both progression free survival (PFS) and overall survival (OS) were improved with the presence of RCs in our study. Besides, despite some statistically significant associations between RCs and molecular subtypes, RCs and estrogen receptor status, the results were largely depending on the stratification methods. Generally, no convincing association was detected between the extent of RCs and the clinicopathological features or prognosis. In sum, the extent of RCs showed limited value as a prognostic predictor in invasive breast carcinoma patients from Central China. C1 [Huang, Liangliang] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of PathologyHefei, China. [Li, Yujie] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of PathologyHefei, China. [Du, Jun] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of PathologyHefei, China. [Li, Heng] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of PathologyHefei, China. [Lu, Mengmeng] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of PathologyHefei, China. [Wang, Yuting] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of PathologyHefei, China. [Zhou, Wenchao] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of PathologyHefei, China. [Wang, Wei] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of PathologyHefei, China. [Wu, Haibo] University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of PathologyHefei, China. RP Wang, W (reprint author), University of Science and Technology of China, Division of Life sciences and Medicine, The First Affiliated Hospital of USTC, Department of Pathology, Hefei, China. EM weiwang@hmfl.ac.cn CR Harbeck N, and Gnant M. Breast cancer. The Lancet, 2017, 389:1134–50., DOI 10.1016/S0140-6736(16)31891-8 Ulamec M, Dˇzombeta T, ˇ Cupi´c H, Leniˇcek T, Tomas D, and Kruˇslin B. Periacinar retraction clefting and D2-40 expression in prostatic adenocarcinoma. Pathol Oncol Res, 2012, 18:365–70., DOI 10.1007/s12253- 011-9453-0 Kruˇslin B, Tomas D, Cviko A, Cupic H, Odak L, and Belicza M. Periacinar Clefting and p63 immunostaining in prostatic intraepithelial neoplasia and prostatic carcinoma. Pathol Oncol Res, 2006, 12:205–9., DOI 10.1007/ BF02893413PAOR.2006.12.4.0205 Acs G, Dumoff KL, Solin LJ, Pasha T, Xu X, and Zhang PJ. Extensive retraction artifact correlates with lymphatic invasion and nodal metastasis and predicts poor outcome in early stage breast carcinoma. Am J Surg Pathol, 2007, 31: 129–40., DOI 10.1097/01.pas.0000213316.59176.9b Acs G, Paragh G, Chuang S-T, Laronga C, and Zhang PJ. The presence of micropapillary features and retraction artifact in core needle biopsy material predicts lymph node metastasis in breast carcinoma. Am J Surg Pathol, 2009, 33:202–10., DOI 10.1097/PAS.0b013e318185e171 Acs G, Khakpour N, Kiluk J, Lee MC, and Laronga C. The presence of extensive retraction clefts in invasive breast carcinomas correlates with lymphatic invasion and nodal metastasis and predicts poor outcome. Am J Surg Pathol, 2015, 39:325–37., DOI 10.1097/PAS.0000000000000339 Favaro WJ, Hetzl AC, Reis LO, Ferreira U, Billis A, and Cagnon VHA. Periacinar retraction clefting in nonneoplastic and neoplastic prostatic glands: artifact or molecular involvement. Pathol Oncol Res, 2012, 18: 285–92., DOI 10.1007/s12253-011-9440-5 Kruˇslin B, Tomas D, Rogatsch H, Relji´c A, Vuci´c M, Balicevi´c D, et al. Correlation of periacinar retraction clefting in needle core biopsies and corresponding prostatectomy specimens of patients with prostatic adenocarcinoma. Int J Surg Pathol, 2005, 13:67–72., DOI 10.1177/ 106689690501300109 Acs G, Paragh G, Rakosy Z, Laronga C, and Zhang PJ. The extent of retraction clefts correlates with lymphatic vessel density and VEGF-C expression and predicts nodal metastasis and poor prognosis in early-stage breast carcinoma. Mod Pathol, 2012, 25:163–77., DOI 10.1038/modpathol.2011.138 Irie J, Manucha V, Ioffe OB, and Silverberg SG. Artefact as the pathologist’s friend: peritumoral retraction in in situ and infiltrating duct carcinoma of the breast. Int J Surg Pathol, 2007, 15:53–9., DOI 10.1177/1066896906295690 Tomas D, Spaji´c B, Miloˇsevi´c M, Demirovi´c A, Maruˇsi´c Z, and Kruˇslin B. Extensive retraction artefact predicts biochemical recurrence-free survival in prostatic carcinoma. Histopathology, 2011, 58:447–54., DOI 10.1111/j.1365- 2559.2011.03769.x Mori K, Takeda M, Kodama Y, Kiyokawa H, Yasojima H, Mizutani M, et al. Tumor thickness and histological features as predictors of invasive foci within preoperatively diagnosed ductal carcinoma in situ. Hum Pathol, 2017, 64: 145–55., DOI 10.1016/j.humpath.2017.04.004 Shah TS, Kaag M, Raman JD, Chan W, Tran T, Kunchala S, et al. Clinical significance of prominent retraction clefts in invasive urothelial carcinoma. Hum Pathol, 2017, 61:90–6., DOI 10.1016/j.humpath.2016.10.021 Jain D, Tikku G, Bhadana P, Dravid C, and Grover RK. The impact of peritumoral retraction clefting & intratumoral eosinophils on overall survival in oral squamous carcinoma patients. Pathol Oncol Res, 2019, 25:183–9., DOI 10.1007/s12253-017-0328-x Deng HY, Wang XR, Yue M, Zhang LL, Li F, Wang XL, et al. [Extensive peritumoral retraction clefts and prognosis in invasive breast carcinomas of no specific type]. Zhonghua Bing Li Xue Za Zhi, 2018, 47(3):196–200., DOI 10. 3760/cma.j.issn.0529-5807.2018.03.010 Dˇzombeta T, and Kruˇslin B. High grade T1 papillary urothelial bladder cancer shows prominent peritumoral retraction clefting. Pathol Oncol Res, 2018, 24: 567–74., DOI 10.1007/s12253-017-0279-2 McKenney JK, Gilks CB, Kalloger S, and Longacre TA. Classification of extraovarian implants in patients with ovarian serous borderline tumors, tumors of low malignant potential, based on clinical outcome. Am J Surg Pathol, 2016, 40:1155–64., DOI 10.1097/PAS. 0000000000000692 Bettelheim R, Mitchell D, and Gusterson BA. Immunocytochemistry in the identification of vascular invasion in breast cancer. J Clin Pathol, 1984, 37: 364–6., DOI 10.1136/jcp.37.4.364 Saigo PE, and Rosen PP. The application of immunohistochemical stains to identify endothelial-lined channels in mammary carcinoma. Cancer, 1987, 59: 51–4., DOI 10.1002/1097-0142(19870101)59:1<51::aid-cncr2820590114>3.0.co; 2-5 Van den Eynden GG, Van der Auwera I, Van Laere SJ, Colpaert CG, van Dam P, Dirix LY, et al. Distinguishing blood and lymph vessel invasion in breast cancer: a prospective immunohistochemical study. Br J Cancer, 2006, 94: 1643–9., DOI 10.1038/sj.bjc.6603152 Hoda SA, Hoda RS, Merlin S, Shamonki J, and Rivera M. Issues relating to lymphovascular invasion in breast carcinoma. Adv Anat Pathol, 2006, 13: 308–15., DOI 10.1097/01.pap.0000213048.69564.26 Gujam FJA, Going JJ, Mohammed ZMA, Orange C, Edwards J, and McMillan DC. Immunohistochemical detection improves the prognostic value of lymphatic and blood vessel invasion in primary ductal breast cancer. BMC Cancer, 2014, 14:676., DOI 10.1186/1471-2407-14-676 Marina K, and Tanja L. Lymphatic and small blood vessel density in the tumor and peritumoral tissue in invasive breast carcinoma of no special type. Libri Oncol, 2018, 46(1):5–13., DOI 10.20471/LO.2018.46.01.0124 Giuliano AE, Edge SB, and Hortobagyi GN. Eighth Edition of the AJCC cancer staging manual: breast cancer. Ann Surg Oncol, 2018, 25(7):1783–5., DOI 10. 1245/s10434-018-6486-6 WHO Classification of Tumours Editorial Board. Breast tumours: WHO classification of tumours, medicine). 5 ed.. Lyon, France: World Health OrganizationIRAC, 2019). Galea MH, Blamey RW, Elston CE, and Ellis IO. The Nottingham prognostic index in primary breast cancer. Breast Cancer Res Tr, 1992, 22:207–19., DOI 10. 1007/BF01840834 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn H-J, et al. Strategies for subtypes-dealing with the diversity of breast cancer: highlights of the st gallen international expert consensus on the primary therapy of early breast cancer 2011. Ann Oncol, 2011, 22:1736–47., DOI 10. 1093/annonc/mdr304 Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, et al. Human epidermal growth factor receptor 2 testing in breast cancer: American society of clinical oncology/college of American pathologists clinical practice guideline focused update. Jco, 2018, 36:2105–22., DOI 10.1200/JCO. 2018.77.8738 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609743 EP 1609750 DI 10.3389/pore.2021.1609743 PG 8 ER PT J AU Au, KY Lo, ChLR AF Au, Kwan-Yung Lo, Cheuk-Lam Regina TI An Immunohistochemical Study of β-catenin Expression and Immune Cell Population in Metastatic Carcinoma to the Liver SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE liver; β-catenin; CD8; tregs; metastatic carcinoma ID liver; β-catenin; CD8; tregs; metastatic carcinoma AB The liver is the commonest site of cancer metastasis. In this study, we asked whether the immune tumor microenvironment in liver metastases was governed by the β-catenin activation status of the tumor. To this end, we analyzed CD8 and FoxP3 immunohistochemical expression against β-catenin expression status of the tumor in a cohort of 52 liver samples with metastatic carcinoma. The results showed that colorectal primary constituted the largest proportion of metastatic carcinoma showing β-catenin overexpression. Intra-tumoral CD8 count was lower and FoxP3 count was higher when compared with the non-tumoral liver parenchyma. β-catenin overexpression was associated with a lower CD8 count in the tumor region (p = 0.003). In summary, our findings are in support of an altered immune tumor microenvironment vs. the non-tumor liver tissues in the metastatic site. Suppression of CD8 count was associated with activated Wnt/β-catenin signaling in the metastatic tumor. C1 [Au, Kwan-Yung] The University of Hong Kong, Department of PathologyPok Fu Lam, Hong Kong. [Lo, Cheuk-Lam Regina] The University of Hong Kong, Department of PathologyPok Fu Lam, Hong Kong. RP Lo, ChLR (reprint author), The University of Hong Kong, Department of Pathology, Pok Fu Lam, Hong Kong. EM reginalo@pathology.hku.hk CR Hinshaw DC, and Shevde LA. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res, 2019, 79(18):4557–66., DOI 10. 1158/0008-5472.CAN-18-3962[published Online First: Epub Date] Luke JJ, Bao R, Sweis RF, Spranger S, and Gajewski TF. WNT/β-catenin Pathway Activation Correlates with Immune Exclusion across Human Cancers. Clin Cancer Res, 2019, 25(10):3074–83., DOI 10.1158/1078-0432. CCR-18-1942[published Online First: Epub Date] Galluzzi L, Spranger S, Fuchs E, and Lopez-Soto A. WNT Signaling in Cancer Immunosurveillance. Trends Cel Biol, 2019, 29(1):44–65., DOI 10.1016/j.tcb. 2018.08.005[published Online First: Epub Date] Li X, Xiang Y, Li F, Yin C, Li B, and Ke X. WNT/β-Catenin Signaling Pathway Regulating T Cell-Inflammation in the Tumor Microenvironment. Front Immunol, 2019, 10:2293., DOI 10.3389/fimmu.2019.02293[published Online First: Epub Date] White BD, Chien AJ, and Dawson DW. Dysregulation of Wnt/β-Catenin Signaling in Gastrointestinal Cancers. Gastroenterol, 2012, 142(2):219–32., DOI 10.1053/j.gastro.2011.12.001[published Online First: Epub Date] Leung CO-N, MakW-N, Kai AK-L, Chan K-S, Lee TK-W, Ng IO-L, et al.Sox9 Confers Stemness Properties in Hepatocellular Carcinoma through Frizzled-7 Mediated Wnt/β-Catenin Signaling. Oncotarget, 2016, 7(20):29371–86., DOI 10.18632/oncotarget.8835[published Online First: Epub Date] Lo RC-L, Leung CO-N, Chan KK-S, Ho DW-H, Wong C-M, Lee TK-W, et al.Cripto-1 Contributes to Stemness in Hepatocellular Carcinoma by Stabilizing Dishevelled-3 and Activating Wnt/β-Catenin Pathway. Cell Death Differ, 2018, 25(8):1426–41., DOI 10.1038/s41418-018-0059-x [published Online First: Epub Date] Bugter JM, Fenderico N, and Maurice MM. Mutations and Mechanisms of WNT Pathway Tumour Suppressors in Cancer. Nat Rev Cancer, 2020, 21: 5–21., DOI 10.1038/s41568-020-00307-z[published Online First: Epub Date] Liu Y, and Cao X. Characteristics and Significance of the Pre-metastatic Niche. Cancer Cell, 2016, 30(5):668–81., DOI 10.1016/j.ccell.2016.09.011[published Online First: Epub Date] Peinado H, Zhang H, Matei IR, Costa-Silva B, Hoshino A, Rodrigues G, et al.Pre-metastatic Niches: Organ-specific Homes for Metastases. Nat Rev Cancer, 2017, 17(5):302–17., DOI 10.1038/nrc.2017.6 [published Online First: Epub Date] Haydon RC, Deyrup A, Ishikawa A, Heck R, Jiang W, Zhou L, et al.Cytoplasmic And/or Nuclear Accumulation of the beta-catenin Protein Is a Frequent Event in Human Osteosarcoma. Int J Cancer, 2002, 102(4):338–42., DOI 10.1002/ijc.10719[published Online First: Epub Date] Gomez-Macias GS, Molinar-Flores G, Lopez-Garcia CA, Santuario-Facio S, Decanini-Arcaute H, Valero-Elizondo J, et al.Immunotyping of Tumor- Infiltrating Lymphocytes in Triple-Negative Breast Cancer and Genetic Characterization. Oncol Lett, 2020, 20(5):140., DOI 10.3892/ol.2020.12000 [published Online First: Epub Date] Thompson ED, Taube JM, Asch-Kendrick RJ, Ogurtsova A, Xu H, Sharma R, et al.PD-L1 Expression and the Immune Microenvironment in Primary Invasive Lobular Carcinomas of the Breast. Mod Pathol, 2017, 30(11): 1551–60., DOI 10.1038/modpathol.2017.79[published Online First: Epub Date] de Ridder J, de Wilt JHW, Simmer F, Overbeek L, Lemmens V, and Nagtegaal I. Incidence and Origin of Histologically Confirmed Liver Metastases: an Explorative Case-Study of 23,154 Patients. Oncotarget, 2016, 7(34):55368–76., DOI 10.18632/oncotarget.10552[published Online First: Epub Date] Ali HR, Provenzano E, Dawson S-J, Blows FM, Liu B, Shah M, et al.Association between CD8+ T-Cell Infiltration and Breast Cancer Survival in 12 439 Patients. Ann Oncol, 2014, 25(8):1536–43., DOI 10.1093/annonc/mdu191 [published Online First: Epub Date] Mahmoud SMA, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AHS, et al.Tumor-infiltrating CD8+ Lymphocytes Predict Clinical Outcome in Breast Cancer. Jco, 2011, 29(15):1949–55., DOI 10.1200/JCO.2010.30.5037 [published Online First: Epub Date] Masugi Y, Abe T, Ueno A, Fujii-Nishimura Y, Ojima H, Endo Y, et al.Characterization of Spatial Distribution of Tumor-Infiltrating CD8+ T Cells Refines Their Prognostic Utility for Pancreatic Cancer Survival. Mod Pathol, 2019, 32(10):1495–507., DOI 10.1038/s41379-019-0291-z [published Online First: Epub Date] NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609752 EP 1609757 DI 10.3389/pore.2021.1609752 PG 6 ER PT J AU Xing, AY Wang, B Li, YH Chen, X Wang, YW Liu, HT Gao, P AF Xing, Ai-Yan Wang, Bin Li, Yu-Hong Chen, Xu Wang, Ya-Wen Liu, Hai-Ting Gao, Peng TI Identification of miRNA Signature in Breast Cancer to Predict Neoadjuvant Chemotherapy Response SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; microarray; miRNA signature; neoadjuvant chemotherapy; drug resistance ID breast cancer; microarray; miRNA signature; neoadjuvant chemotherapy; drug resistance AB Chemotherapy failure causes high breast cancer recurrence and poor patient prognosis. Thus, we studied a cohort of novel biomarkers to predict chemotherapeutic response in breast cancer. In this study, miRNA expression profiling was performed on 10 breast cancer punctured specimens sensitive to chemotherapy (MP grade 4, 5) and 10 chemotherapy resistant (MP grade 1). Differentially expressed miRNAs were verified by qRT-PCR in 60 initial samples, 59 validated samples and 71 independent samples. A miRNA signature was generated using a Logistic regression model. A receiver operating characteristic (ROC) test was used to assess specificity and sensitivity of single miRNA and miRNA signature. Target genes regulated by miRNAs and their involved signaling pathways were analyzed using GO enrichment and KEGG software. MiRNAs expression were separately compared with ER, PR, HER2 immunohistochemical staining and different drugs. qRT-PCR showed that the high expression of miR-23a-3p, miR-200c-3p, miR-214-3p and the low expression of miR-451a and miR-638 were closely related to chemoresistance. According to the formula for calculating the drug resistance risk, patients in the high-risk group were more likely to develop chemotherapy resistance than the low-risk group. Bioinformatics analysis showed that 5 miRNAs and target genes are mainly involved in p53, ubiquitin-mediated proteolysis, mTOR, Wnt, cells skeletal protein regulation, cell adhesion and ErbB signaling pathways. miR-451a expression was associated with ER, HER-2 status and anthracyclines. A miRNA signature of chemotherapeutic response may be clinically valuable for improving current chemotherapy regimens of individual treatment for patients with breast cancer. C1 [Xing, Ai-Yan] Shandong Univeristy, School of Medicine, Department of PathologyJinan, China. [Wang, Bin] The First Affiliated Hospital of Shandong First Medical University, Department of SurgeryJinan, China. [Li, Yu-Hong] Liaocheng People’s Hospital, Department of PathologyLiaocheng, China. [Chen, Xu] Shandong University, Qilu Hospital, Department of PathologyJinan, China. [Wang, Ya-Wen] Shandong Univeristy, QiLu Hospital, Department of Breast SurgeryJinan, China. [Liu, Hai-Ting] Shandong Univeristy, School of Medicine, Department of PathologyJinan, China. [Gao, Peng] Shandong Univeristy, School of Medicine, Department of PathologyJinan, China. RP Gao, P (reprint author), Shandong Univeristy, School of Medicine, Department of Pathology, Jinan, China. EM gaopeng@sdu.edu.cn CR Siegel RL, Miller KD, and Jemal A Cancer statistics, 2016. CA: A Cancer J Clinicians, 2016, 66:7–30., DOI 10.3322/caac.21332 Debled M, MacGrogan G, Breton-Callu C, Ferron S, Hurtevent G, Fournier M, et al. Surgery following neoadjuvant chemotherapy for HER2-positive locally advanced breast cancer. time to reconsider the standard attitude. Eur J Cancer, 2015, 51:697–704., DOI 10.1016/j.ejca.2015.01.063 Balko JM, Cook RS, Vaught DB, Kuba MG, Miller TW, Bhola NE, et al. Profiling of residual breast cancers after neoadjuvant chemotherapy identifies DUSP4 deficiency as a mechanism of drug resistance. Nat Med, 2012, 18: 1052–9., DOI 10.1038/nm.2795 Miller TE, Ghoshal K, Ramaswamy B, Roy S, Datta J, Shapiro CL, et al. MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1. J Biol Chem, 2008, 283:29897–903., DOI 10.1074/jbc.m804612200 Pan Y-Z, Morris ME, and Yu A-M MicroRNA-328 negatively regulates the expression of breast cancer resistance protein, BCRP/ABCG2, in human cancer cells. Mol Pharmacol, 2009, 75:1374–9., DOI 10.1124/mol.108.054163 Pogribny IP, Filkowski JN, Tryndyak VP, Golubov A, Shpyleva SI, and Kovalchuk O Alterations of microRNAs and their targets are associated with acquired resistance of MCF-7 breast cancer cells to cisplatin. Int J Cancer, 2010, 127:1785–94., DOI 10.1002/ijc.25191 Duch J, Fuster D, Munoz M, Fernandez PL, Paredes P, Fontanillas M, et al. 18F-FDG PET/CT for early prediction of response to neoadjuvant chemotherapy in breast cancer. Eur J Nucl Med Mol Imaging, 2009, 36: 1551–7., DOI 10.1007/s00259-009-1116-y Gao P, Wong CC, Tung EK, Lee JM, Wong CM, and Ng IO Deregulation of microRNA expression occurs early and accumulates in early stages of HBVassociated multistep hepatocarcinogenesis. J Hepatol, 2012, 54:1177–84., DOI 10.1016/j.jhep.2010.09.023 Gnant M, Harbeck N, and Thomssen C St. Gallen 2011: summary of the consensus discussion. Breast Care, 2011, 6:136–41., DOI 10.1159/000328054 Inic Z, ZegaracM, Inic M, Markovic I, Kozomara Z, Djurisic I, et al. Difference between luminal A and luminal B subtypes according to Ki-67, tumor size, and progesterone receptor negativity providing prognostic information. Clin Med Insights Oncol, 2014, 8:107–11., DOI 10.4137/cmo.s18006 Gao P, Zhou G-Y, Zhang Q-H, Su Z-X, Zhang T-G, Xiang L, et al. Lymphangiogenesis in gastric carcinoma correlates with prognosis. J Pathol, 2009, 218:192–200., DOI 10.1002/path.2523 Wolff AC, Hammond MEH, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Jco, 2007, 26:118–45., DOI 10.1200/jco.2006.09.2775 Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. American society of clinical oncology/college of American pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. Arch Pathol Lab Med, 2010, 134:907–22., DOI 10.1043/1543-2165-134.6.907 Shen S, Sun Q, Liang Z, Cui X, Ren X, Chen H, et al. A prognostic model of triple-negative breast cancer based on miR-27b-3p and node status. PLoS One, 2014, 9:e100664., DOI 10.1371/journal.pone.0100664 Chen J, Yao D, Li Y, Chen H, He C, Ding N, et al. Serum microRNA expression levels can predict lymph node metastasis in patients with early-stage cervical squamous cell carcinoma. Int J Mol Med, 2013, 32:557–67., DOI 10.3892/ijmm. 2013.1424 Singh PK, Preus L, Hu Q, Yan L, Long MD, Morrison CD, et al. Serum microRNA expression patterns that predict early treatment failure in prostate cancer patients. Oncotarget, 2014, 5:824–40., DOI 10.18632/oncotarget.1776 Huang Z, Zhu D, Wu L, He M, Zhou X, Zhang L, et al. Six serum-based miRNAs as potential diagnostic biomarkers for gastric cancer. Cancer Epidemiol Biomarkers Prev, 2017, 26:188–96., DOI 10.1158/1055-9965.epi- 16-0607 Petriella D, De Summa S, Lacalamita R, Galetta D, Catino A, Logroscino AF, et al. miRNA profiling in serum and tissue samples to assess noninvasive biomarkers for NSCLC clinical outcome. Tumor Biol, 2016, 37:5503–13., DOI 10.1007/s13277-015-4391-1 Xi Y, Nakajima G, Gavin E, Morris CG, Kudo K, Hayashi K, et al. Systematic analysis of microRNA expression of RNA extracted from fresh frozen and formalin-fixed paraffin-embedded samples. RNA, 2007, 13:1668–74., DOI 10. 1261/rna.642907 Zhang X, Chen J, Radcliffe T, LeBrun DP, Tron VA, and Feilotter H An arraybased analysis of microRNA expression comparing matched frozen and formalin-fixed paraffin-embedded human tissue samples. J Mol Diagn, 2008, 10:513–9., DOI 10.2353/jmoldx.2008.080077 Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med, 2004, 351:2817–26., DOI 10.1056/nejmoa041588 Bergamaschi A, and Katzenellenbogen BS Tamoxifen downregulation of miR-451 increases 14-3-3ζ and promotes breast cancer cell survival and endocrine resistance. Oncogene, 2010, 31:39–47., DOI 10.1038/onc. 2011.223 Yu Z-W, Zhong L-P, Ji T, Zhang P, Chen W-T, and Zhang C-P MicroRNAs contribute to the chemoresistance of cisplatin in tongue squamous cell carcinoma lines. Oral Oncol, 2010, 46:317–22., DOI 10.1016/j.oraloncology. 2010.02.002 Wang F, Lou J-F, Cao Y, Shi X-H, Wang P, Xu J, et al. miR-638 is a new biomarker for outcome prediction of non-small cell lung cancer patients receiving chemotherapy. Exp Mol Med, 2015, 47:e162., DOI 10.1038/emm. 2015.17 Lv J, Xia K, Xu P, Sun E, Ma J, Gao S, et al. miRNA expression patterns in chemoresistant breast cancer tissues. Biomed Pharmacother, 2014, 68:935–42., DOI 10.1016/j.biopha.2014.09.011 Valicsek E, Koszo R, Dobi A, Uhercsak G, Varga Z, Vass A, et al. Cardiac surveillance findings during adjuvant and palliative trastuzumab therapy in patients with breast cancer. Anticancer Res, 2015, 35:4967–73. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609753 EP 1609763 DI 10.3389/pore.2021.1609753 PG 11 ER PT J AU Hrudka, J Prouzova, Z Mydlikova, K Jedlickova, K Holesta, M Whitley, A Havluj, L AF Hrudka, Jan Prouzova, Zuzana Mydlikova, Katarina Jedlickova, Kristina Holesta, Michal Whitley, Adam Havluj, Lukas TI FOXF1 as an Immunohistochemical Marker of Hilar Cholangiocarcinoma or Metastatic Pancreatic Ductal Adenocarcinoma. Single Institution Experience SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE intrahepatic cholangiocarcinoma; cholangiocarcinoma; Foxf1; peripheral; hilar; extrahepatic ID intrahepatic cholangiocarcinoma; cholangiocarcinoma; Foxf1; peripheral; hilar; extrahepatic AB Cholangiocarcinoma (CCA) is a liver malignancy associated with a poor prognosis. Its main subtypes are peripheral/intrahepatic and hilar/extrahepatic CCA. Several molecular, morphological and clinical similarities between hilar/extrahepatic CCA and pancreatic ductal adenocarcinoma (PDAC) have been described. FOXF1 is a transcription factor which has been described to have prognostic significance in various tumors and it is involved in the development of bile ducts. The aim of this study is to determine occurrence of nuclear expression of FOXF1 in both subtypes of CCA and metastatic PDAC and assess its potential usefulness as a diagnostic marker. Secondary aims were to investigate the use of C-reactive protein (CRP) immunohistochemistry for diagnosing intrahepatic peripheral CCA and the significance of histological features in CCA subtypes. 32 archive specimens of CCA, combined hepatocellular carcinoma-CCA (HCC-CCA) and liver metastasis of PDAC were stained by FOXF1 and CRP immunohistochemistry and evaluated to determine histological pattern. The CCAs were classified radiologically into peripheral/ intrahepatic and hilar subtype. Using Fisher exact test, we identified nuclear FOXF1 as a fairly specific (87%) but insensitive (65%) marker of hilar and extrahepatic CCA and metastatic PDAC (p = 0.005). CRP immunohistochemistry was characterized by a high sensitivity and specificity, of 79% and 88%, respectively (p = 0.001). We did not identify any histomorphological features associated with either types of CCA or metastatic PDAC. As a conclusion of novel finding, FOXF1 immunohistochemistry may be regarded as a specific but insensitive marker of hilar/extrahepatic CCA and metastatic PDAC and it may help distinguish them from peripheral CCA. C1 [Hrudka, Jan] Charles University, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, The Fingerland Department of PathologyPrague, Czech Republic. [Prouzova, Zuzana] Charles University, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, The Fingerland Department of PathologyPrague, Czech Republic. [Mydlikova, Katarina] Charles University, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, The Fingerland Department of PathologyPrague, Czech Republic. [Jedlickova, Kristina] Institute for Clinical and Experimental Medicine, Clinical and Transplant Pathology CentrePrague, Czech Republic. [Holesta, Michal] Charles University, 3rd Faculty of Medicine, University Hospital Kralovske Vinohrady, Department of RadiodiagnosticsPrague, Czech Republic. [Whitley, Adam] Charles University, 3rd Faculty of Medicine, University Hospital Kralovske Vinohrady, Department of General SurgeryPrague, Czech Republic. [Havluj, Lukas] Charles University, 3rd Faculty of Medicine, University Hospital Kralovske Vinohrady, Department of General SurgeryPrague, Czech Republic. RP Hrudka, J (reprint author), Charles University, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, The Fingerland Department of Pathology, Prague, Czech Republic. EM jan.hrudka@fnkv.cz CR Nakanuma Y, Klimstra DS, Komuta M, Zen Y. Intrahepatic cholangiocarcinoma. In: WHO Classification of Tumors Editorial Board. Digestive system tumors, WHO classification of tumors series. 5th ed. Vol. 1. Lyon, France: International Agency for Research on Cancer, 2019), p. 254–9. Roa JC, Adsay NV, Arola J, Tsui WM, Zen Y. Carcinoma of the gallbladder. In: WHO classification of tumors editorial board. Digestive system tumors, WHO classification of tumors series. Vol. 1. Lyon, France: International Agency for Research on Cancer. 5th ed., 2019), p. 283–8. Roa JC, Adsay NV, Arola J, Tsui WM, Zen Y. Carcinoma of the extrahepatic bile ducts. In: WHO Classification of Tumors Editorial Board. Digestive system tumors, WHO classification of tumors series. 5th ed. Vol. 1. Lyon, France: International Agency for Research on Cancer, 2019), p. 289–91. Sempoux C, Kakar S, Kondo F, Schirmacher P. Combined hepatocellularcholangiocarcinoma and undifferentiated primary liver carcinoma. In: WHO classification of tumors editorial board. Digestive system tumors. 5th ed. Vol. 1. Lyon, France: International Agency for Research on Cancer, 2019), p. 260–2. Guedj N, Bedossa P, Paradis V. Anatomopathologie des cholangiocarcinomes. Ann de Pathologie, 2010, 30(6):455–63., DOI 10.1016/j.annpat.2010.10.004 Gandou C, Harada K, Sato Y, Igarashi S, Sasaki M, Ikeda H, et al. Hilar cholangiocarcinoma and pancreatic ductal adenocarcinoma share similar histopathologies, immunophenotypes, and development-related molecules. Hum Pathol, 2013, 44(5):811–21., DOI 10.1016/j.humpath.2012.08.004 Nakanuma Y, Sato Y. Hilar cholangiocarcinoma is pathologically similar to pancreatic duct adenocarcinoma: suggestions of similar background and development. J Hepatob Pancreat Sci, 2014, 21(7):441–7., DOI 10.1002/jhbp.70 Lepreux S, Bioulac-Sage P, Chevet E. Differential expression of the anterior gradient protein-2 is a conserved feature during morphogenesis and carcinogenesis of the biliary tree. Liver Int, 2011, 31(3):322–8., DOI 10.1111/j. 1478-3231.2010.02438.x Yeh Y-C, Lei H-J, Chen M-H, Ho H-L, Chiu L-Y, Li C-P, et al. C-reactive protein, CRP, is a promising diagnostic immunohistochemical marker for intrahepatic cholangiocarcinoma and is associated with better prognosis. Am J Surg Pathol, 2017, 41(12):1630–41., DOI 10.1097/PAS.0000000000000957 Shahid M, Mubeen A, Tse J, Kakar S, Bateman AC, Borger D, et al. Branched chain in situ hybridization for albumin as a marker of hepatocellular differentiation. Am J Surg Pathol, 2015, 39(1):25–34., DOI 10.1097/PAS. 0000000000000343 Cardinale V, Bragazzi MC, Carpino G, Torrice A, Fraveto A, Gentile R, et al. Cholangiocarcinoma: increasing burden of classifications. Hepatob Surg Nutr, 2013, 2(5):272–80., DOI 10.3978/j.issn.2304-3881.2013.10.02 Tan JCC, Coburn NG, Baxter NN, Kiss A, Law CHL. Surgical management of intrahepatic cholangiocarcinoma - a population-based study. Ann Surg Oncol, 2008, 15(2):600–8., DOI 10.1245/s10434-007-9627-x Peterson RS, Lim L, Ye H, Zhou H, Overdier DG, Costa RH. The winged helix transcriptional activator HFH-8 is expressed in the mesoderm of the primitive streak stage of mouse embryos and its cellular derivatives. Mech Dev, 1997, 69: 53–69., DOI 10.1016/s0925-4773(97)00153-6 Kalinichenko VV, Lim L, Stolz DB, Shin B, Rausa FM, Clark J, et al. Defects in pulmonary vasculature and perinatal lung hemorrhage in mice heterozygous null for the Forkhead Box f1 transcription factor. Dev Biol, 2001, 235(2): 489–506., DOI 10.1006/dbio.2001.0322 Kalinichenko VV, Gusarova GA, Kim I-M, Shin B, Yoder HM, Clark J, et al. FOXF1 haploinsufficiency reduces Notch-2 signaling during mouse lung development. Am J Physiol Lung Cell Mol Physiol, 2004, 286(3): L521–L530., DOI 10.1152/ajplung.00212.2003 Szafranski P, Yang Y, Nelson MU, Bizzarro MJ, Morotti RA, Langston C, et al. Novel FOXF1 deep intronic deletion causes lethal lung developmental disorder, alveolar capillary dysplasia with misalignment of pulmonary veins. Hum Mutat, 2013, 34(11):1467–71., DOI 10.1002/humu.22395 Szafranski P, Dharmadhikari AV, Wambach JA, Towe CT, White FV, Grady RM, et al. Two deletions overlapping a distant FOXF1 enhancer unravel the role of lncRNALINC01081in etiology of alveolar capillary dysplasia with misalignment of pulmonary veins. Am J Med Genet, 2014, 164(8):2013–9., DOI 10.1002/ajmg.a.36606 Szafranski P, Herrera C, Proe LA, Coffman B, Kearney DL, Popek E, et al. Narrowing the FOXF1 distant enhancer region on 16q24.1 critical for ACDMPV. Clin Epigenet, 2016, 8:112., DOI 10.1186/s13148-016-0278-2 Szafranski P, Gambin T, Dharmadhikari AV, Akdemir KC, Jhangiani SN, Schuette J, et al. Pathogenetics of alveolar capillary dysplasia with misalignment of pulmonary veins. Hum Genet, 2016, 135(5):569–86., DOI 10.1007/s00439-016-1655-9 Steiner LA, Getman M, Schiralli Lester GM, Iqbal MA, Katzman P, Szafranski P, et al. Disruption of normal patterns of FOXF1 expression in a lethal disorder of lung development. J Med Genet, 2020, 57(5):296–300., DOI 10.1136/ jmedgenet-2019-106095 Kalinichenko VV, Zhou Y, Bhattacharyya D, Kim W, Shin B, Bambal K, et al. Haploinsufficiency of the mouse Forkhead Box f1 gene causes defects in gall bladder development. J Biol Chem, 2002, 277(14):12369–74., DOI 10.1074/jbc. M112162200 Lo P-K, Lee JS, Chen H, Reisman D, Berger FG, Sukumar S. Cytoplasmic mislocalization of overexpressed FOXF1 is associated with themalignancy and metastasis of colorectal adenocarcinomas. Exp Mol Pathol, 2013, 94(1):262–9., DOI 10.1016/j.yexmp.2012.10.014 Carpino G, Cardinale V, Onori P, Franchitto A, Berloco PB, Rossi M, et al. Biliary tree stem/progenitor cells in glands of extrahepatic and intraheptic bile ducts: an anatomical in situ study yielding evidence of maturational lineages. J Anat, 2012, 220:186–99., DOI 10.1111/j.1469-7580.2011.01462.x Komuta M, Govaere O, Vandecaveye V, Akiba J, Van Steenbergen W, Verslype C, et al. Histological diversity in cholangiocellular carcinoma reflects the different cholangiocyte phenotypes. Hepatology, 2012, 55:1876–88., DOI 10. 1002/hep.25595 Spence JR, Lange AW, Lin S-CJ, Kaestner KH, Lowy AM, Kim I, et al. Sox17 regulates organ lineage segregation of ventral foregut progenitor cells. Dev Cell, 2009, 17:62–74., DOI 10.1016/j.devcel.2009.05.012 Fukuda A, Kawaguchi Y, Furuyama K, Kodama S, Horiguchi M, Kuhara T, et al. Ectopic pancreas formation in Hes1-knockout mice reveals plasticity of endodermal progenitors of the gut, bile duct, and pancreas. J Clin Invest, 2006, 116(6):1484–93., DOI 10.1172/JCI27704 Wei H-J, Nickoloff JA, ChenW-H, Liu H-Y, Lo W-C, Chang Y-T, et al. FOXF1 mediates mesenchymal stem cell fusion-induced reprogramming of lung cancer cells. Oncotarget, 2014, 5:9514–29., DOI 10.18632/oncotarget.2413 Malin D, Kim I-M, Boetticher E, Kalin TV, Ramakrishna S, Meliton L, et al. Forkhead box F1 is essential for migration of mesenchymal cells and directly induces integrin-beta3 expression. Mol Cell Biol, 2007, 27:2486–98., DOI 10. 1128/MCB.01736-06 Wang S, Yan S, Zhu S, Zhao Y, Yan J, Xiao Z, et al. FOXF1 induces epithelialmesenchymal transition in colorectal cancer metastasis by transcriptionally activating SNAI1. Neoplasia, 2018, 20(10):996–1007., DOI 10.1016/j.neo.2018. 08.004 Wang S, Xiao Z, Hong Z, Jiao H, Zhu S, Zhao Y, et al. FOXF1 promotes angiogenesis and accelerates bevacizumab resistance in colorectal cancer by transcriptionally activating VEGFA. Cancer Lett, 2018, 439:78–90., DOI 10. 1016/j.canlet.2018.09.026 Saito R-A, Micke P, Paulsson J, Augsten M, Pena C, Jonsson P, et al. Forkhead box F1 regulates tumor-promoting properties of cancer-associated fibroblasts in lung cancer. Cancer Res, 2010, 70(7):2644–54., DOI 10.1158/0008-5472. CAN-09-3644 Zhao ZG, Wang DQ, Hu DF, Li YS, Liu SH. Decreased FOXF1 promotes hepatocellular carcinoma tumorigenesis, invasion, and stemness and is associated with poor clinical outcome. Onco Targets Ther, 2016, 9: 1743–52., DOI 10.2147/OTT.S95002 Lo P-K, Lee JS, Liang X, Han L, Mori T, Fackler MJ, et al. Epigenetic inactivation of the potential tumor suppressor gene FOXF1 in breast cancer. Cancer Res, 2010, 70:6047–58., DOI 10.1158/0008-5472.CAN-10-1576 Nilsson J, Helou K, Kovacs A, Bendahl P-O, Bjursell G, Ferno M, et al. Nuclear Janus-activated kinase 2/nuclear factor 1-C2 suppresses tumorigenesis and epithelial-to-mesenchymal transition by repressing Forkhead box F1. Cancer Res, 2010, 70:2020–9., DOI 10.1158/0008-5472. CAN-09-1677 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609756 EP 1609764 DI 10.3389/pore.2021.1609756 PG 9 ER PT J AU Wang, L Liu, D Wei, J Yuan, L Zhao, Sh Huang, Y Ma, J Yang, Z AF Wang, Linlin Liu, Dan Wei, Jun Yuan, Liwei Zhao, Shiyun Huang, Yani Ma, Jingwen Yang, Zhijuan TI MiR-543 Inhibits the Migration and Epithelial-To-Mesenchymal Transition of TGF-β-Treated Endometrial Stromal Cells via the MAPK and Wnt/β-Catenin Signaling Pathways SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE molecular biology; miRNA-543; MAPK1; wnt/β-catenin; endometrial stromal cell ID molecular biology; miRNA-543; MAPK1; wnt/β-catenin; endometrial stromal cell AB Intrauterine adhesion (IUA) is one of the most prevalent reproductive system diseases in females. MicroRNAs (miRNAs) are reported to be master regulators in a variety of diseases, including IUA, but the role of microRNA-543 (miR-543) in IUA remains to be elucidated. In this study, we observed that miR-543 was downregulated in transforming growth factorbeta (TGF-β)-treated endometrial stromal cells (ESCs). Functionally, we observed that miR-543 suppressed the migration, epithelial-to-mesenchymal transition (EMT), and inhibited expression of extracellular matrix (ECM) proteins in TGF-β-treated ESCs. Mechanistically, MAPK1 is targeted by miR-543 after prediction and screening. A luciferase reporter assay demonstrated that miR-543 complementarily binds with the 3′ untranslated region of mitogen-activated protein kinase 1 (MAPK1), and western blot analysis indicated that miR-543 negatively regulates MAPK1 protein levels. In addition, results from rescue assays showed that miR-543 inhibits the migration and EMT of TGF-β-treated ESCs by targeting MAPK1. In addition, we observed that miR-543 inactivates the Wnt/β-catenin signaling pathway through inhibiting the phosphorylation of MAPK1 and β-catenin. Finally, we confirmed that miR-543 represses migration, EMT and inhibits levels of ECM proteins in TGF-β-treated ESCs by targeting the Wnt/β-catenin signaling pathway. Our results demonstrated that miR-543 suppresses migration and EMT of TGF-β-treated ESCs by targeting the MAPK and Wnt/β-catenin pathways. C1 [Wang, Linlin] Ningxia Medical University, College of Clinical MedicineYinchuan, China. [Liu, Dan] Ningxia Medical University, College of Clinical MedicineYinchuan, China. [Wei, Jun] Ningxia Medical University, College of Clinical MedicineYinchuan, China. [Yuan, Liwei] Ningxia Medical University, College of Clinical MedicineYinchuan, China. [Zhao, Shiyun] Ningxia Medical University, College of Clinical MedicineYinchuan, China. [Huang, Yani] Ningxia Medical University, College of Clinical MedicineYinchuan, China. [Ma, Jingwen] General Hospital of Ningxia Medical University, Department of GynecologyYinchuan, China. [Yang, Zhijuan] General Hospital of Ningxia Medical University, Department of GynecologyYinchuan, China. RP Wei, J (reprint author), Ningxia Medical University, College of Clinical Medicine, Yinchuan, China. EM 13895418876@163.com CR Healy MW, Schexnayder B, Connell MT, Terry N, DeCherney AH, and Csokmay JM. Intrauterine Adhesion Prevention after Hysteroscopy: a Systematic Review and Meta-Analysis. Am J Obstet Gynecol, 2016, 215(3): 267–75., DOI 10.1016/j.ajog.2016.05.001 Tam WH, Lau WC, Cheung LP, Yuen PM, and Chung TK-H. Intrauterine adhesions after conservative and surgical management of spontaneous abortion. The J Am Assoc Gynecol Laparoscopists, 2002, 9(2):182–5., DOI 10. 1016/s1074-3804(05)60129-6 Ludwin A, Martins WP, and Ludwin I. Ultrasound-guided repeat intrauterine balloon dilatation for prevention of adhesions. Ultrasound Obstet Gynecol, 2019, 54:566–8., DOI 10.1002/uog.20223 Lagana AS, Garzon S, Franchi M, Casarin J, Gullo G, and Ghezzi F. Translational animal models for endometriosis research: a long and windy road. Ann Transl Med, 2018, 6(22):431., DOI 10.21037/atm.2018.08.24 Fei Z, Bin Z, Xin X, Fei H, and Yuechong C. Meta-analysis on the use of hyaluronic acid gel to prevent recurrence of intrauterine adhesion after hysteroscopic adhesiolysis. Taiwanese J Obstet Gynecol, 2019, 58(6):731–6., DOI 10.1016/j.tjog.2019.09.002 Yu D, Wong Y-M, Cheong Y, Xia E, and Li T-C. Asherman syndrome-one century later. Fertil sterility, 2008, 89(4):759–79., DOI 10.1016/j.fertnstert.2008. 02.096 Maslar IA, and Riddick DH. Prolactin production by human endometrium during the normal menstrual cycle. Am J Obstet Gynecol, 1979, 135(6):751–4., DOI 10.1016/0002-9378(79)90386-7 Irwin JC, Kirk D, King RJB, Quigley MM, and Gwatkin RBL. Hormonal regulation of human endometrial stromal cells in culture: an in vitro model for decidualization. Fertil sterility, 1989, 52(5):761–8., DOI 10.1016/s0015- 0282(16)61028-2 Zhu H-Y, Ge T-X, Pan Y-B, and Zhang S-Y. Advanced role of hippo signaling in endometrial fibrosis. Chin Med J, 2017, 130(22):2732–7., DOI 10.4103/0366- 6999.218013 Guo LP, Chen LM, Chen F, Jiang NH, and Sui L. Smad signaling coincides with epithelial-mesenchymal transition in a rat model of intrauterine adhesion. Am J Transl Res, 2019, 11(8):4726–37. Sporn MB, and Roberts AB. TGF-beta: problems and prospects. Cell Regul, 1990, 1(12):875–82., DOI 10.1091/mbc.1.12.875 Kamato D, Burch ML, Piva TJ, Rezaei HB, Rostam MA, and Xu S. Transforming growth factor-β signalling: Role and consequences of Smad linker region phosphorylation. Cell Signal, 2013, 25(10):2017–24., DOI 10.1016/ j.cellsig.2013.06.001 Bai X, Liu J, Yuan W, Liu Y, Li W, and Cao S. Therapeutic effect of human amniotic epithelial cells in rat models of intrauterine adhesions. Cel Transpl, 2020, 29:096368972090849., DOI 10.1177/0963689720908495 Song M, Cao C, Zhou Z, Yao S, Jiang P, and Wang H. HMGA2-induced epithelial-mesenchymal transition is reversed by let-7d in intrauterine adhesions. Mol Hum Reprod, 2021, 27(2)., DOI 10.1093/molehr/gaaa074 Ai Y, Chen M, Liu J, Ren L, Yan X, and Feng Y. lncRNA TUG1 promotes endometrial fibrosis and inflammation by sponging miR-590-5p to regulate Fasl in intrauterine adhesions. Int immunopharmacol., 2020, 86:106703., DOI 10.1016/j.intimp.2020.106703 Cao J, Liu D, Zhao S, Yuan L, Huang Y, and Ma J . Estrogen attenuates TGF- β1-induced EMT in intrauterine adhesion by activating Wnt/β-catenin signaling pathway. Braz J Med Biol Res, 2020, 53(8):e9794., DOI 10.1590/ 1414-431x20209794 Cao J, Huang M, Guo L, Zhu L, Hou J, and Zhang L. MicroRNA-195 rescues ApoE4-induced cognitive deficits and lysosomal defects in Alzheimer’s disease pathogenesis. Mol Psychiatry, 2020)., DOI 10.1038/s41380-020-0824-3 Drewry MD, Challa P, Kuchtey JG, Navarro I, Helwa I, and Hu Y. Differentially expressed microRNAs in the aqueous humor of patients with exfoliation glaucoma or primary open-angle glaucoma. HumMol Genet, 2018, 27(7):1263–75., DOI 10.1093/hmg/ddy040 Torella D, Iaconetti C, Tarallo R, Marino F, Giurato G, and Veneziano C. miRNA regulation of the hyperproliferative phenotype of vascular smooth muscle cells in diabetes. Diabetes, 2018, 67(12):2554–68., DOI 10.2337/db17- 1434 Zhou C-F, Liu M-J, Wang W, Wu S, Huang Y-X, and Chen G-B. miR-205-5p inhibits human endometriosis progression by targeting ANGPT2 in endometrial stromal cells. Stem Cel Res Ther, 2019, 10(1):287., DOI 10.1186/ s13287-019-1388-5 Zhang L, Liu X, Liu J, Ma X, Zhou Z, and Song Y. miR-26a promoted endometrial epithelium cells, EECs, proliferation and induced stromal cells, ESCs, apoptosis via the PTEN -PI3K/AKT pathway in dairy goats. J Cel Physiol, 2018, 233(6):4688–706., DOI 10.1002/jcp.26252 Berkhout RP, Keijser R, Repping S, Lambalk CB, Afink GB, and Mastenbroek S. High-quality human preimplantation embryos stimulate endometrial stromal cell migration via secretion of microRNA hsa-miR-320a. Hum Reprod, 2020)., DOI 10.1093/humrep/deaa149 Yu L, Zhou L, Cheng Y, Sun L, Fan J, and Liang J. MicroRNA-543 acts as an oncogene by targeting PAQR3 in hepatocellular carcinoma. Am J Cancer Res, 2014, 4(6):897–906. Wang L-H, Tsai H-C, Cheng Y-C, Lin C-Y, Huang Y-L, and Tsai C-H. CTGF promotes osteosarcoma angiogenesis by regulating miR-543/angiopoietin 2 signaling. Cancer Lett, 2017, 391:28–37., DOI 10.1016/j.canlet.2017.01.013 Bing L, Hong C, Li-Xin S, and Wei G. MicroRNA-543 suppresses endometrial cancer oncogenicity via targeting FAK and TWIST1 expression. Arch Gynecol Obstet, 2014, 290(3):533–41., DOI 10.1007/s00404-014-3219-3 Yang P, Wu Z, Ma C, Pan N, Wang Y, and Yan L. Endometrial miR-543 is downregulated during the implantation window in women with endometriosis-related infertility. Reprod Sci, 2019, 26(7):900–8., DOI 10. 1177/1933719118799199 Liu X, Duan H, Zhang H-H, Gan L, and Xu Q. Integrated data set of microRNAs and mRNAs involved in severe intrauterine adhesion. Reprod Sci, 2016, 23(10):1340–7., DOI 10.1177/1933719116638177 KrikunG,Mor G, Alvero A, Guller S, Schatz F, and Sapi E. A novel immortalized human endometrial stromal cell line with normal progestational response. Endocrinology, 2004, 145(5):2291–6., DOI 10.1210/en.2003-1606 Livak KJ, and Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods, 2001, 25(4): 402–8., DOI 10.1006/meth.2001.1262 Zhou C, Zhao X, and Duan S. The role of miR-543 in human cancerous and noncancerous diseases. J Cel Physiol, 2020, 236:15., DOI 10.1002/jcp.29860 Zhao H, Diao C, Wang X, Xie Y, Liu Y, and Gao X. MiR-543 promotes migration, invasion and epithelial-mesenchymal transition of esophageal cancer cells by targeting phospholipase A2 group IVA. Cell Physiol Biochem, 2018, 48(4):1595–604., DOI 10.1159/000492281 Du Y, Liu X-H, Zhu H-C, Wang L, Ning J-Z, and Xiao C-C. MiR-543 promotes proliferation and epithelial-mesenchymal transition in prostate cancer via targeting RKIP. Cel Physiol Biochem, 2017, 41(3):1135–46., DOI 10.1159/ 000464120 Fuentes-Mattei E, Bayraktar R, Manshouri T, Silva AM, Ivan C, and Gulei D . miR-543 regulates the epigenetic landscape of myelofibrosis by targeting TET1 and TET2. JCI insight, 2020, 5(1)., DOI 10.1172/jci.insight.121781 Zheng Y, Zhou C, Yu X-X, Wu C, Jia H-L, and Gao X-M. Osteopontin promotes metastasis of intrahepatic cholangiocarcinoma through recruiting MAPK1 and mediating Ser675 phosphorylation of β-Catenin. Cell Death Dis, 2018, 9(2):179., DOI 10.1038/s41419-017-0226-x Wang DP, Tang XZ, Liang QK, Zeng XJ, Yang JB, and Xu J. Overexpression of long noncoding RNA SLC26A4-AS1 inhibits the epithelial-mesenchymal transition via the MAPK pathway in papillary thyroid carcinoma. J Cel Physiol, 2020, 235(3):2403–13., DOI 10.1002/jcp.29145 Liu S, Huang X, Liu Y, Song D, and Xiao Y. Functional analysis of miRNAs combined with TGF-β1/Smad3 inhibitor in an intrauterine rat adhesion cell model. Mol Cel Biochem, 2020, 470(1–2):15–28., DOI 10.1007/s11010-020- 41-7 37. Lucchi NW, and Moore JM. LPS induces secretion of chemokines by human syncytiotrophoblast cells in a MAPK-dependent manner. J Reprod Immunol, 2007, 73(1):20–7., DOI 10.1016/j.jri.2006.05.005 Chen P, XuW, Luo Y, Zhang Y, He Y, and Yang S . MicroRNA 543 suppresses breast cancer cell proliferation, blocks cell cycle and induces cell apoptosis via direct targeting of ERK/MAPK. Ott, 2017, 10:1423–31., DOI 10.2147/ott. S118366 Huang P, Yan R, Zhang X, Wang L, Ke X, and Qu Y. Activating Wnt/β-catenin signaling pathway for disease therapy: challenges and opportunities. Pharmacol Ther, 2019, 196:79–90., DOI 10.1016/j.pharmthera.2018.11.008 Chen Z-Y, Du Y,Wang L, Liu X-H, Guo J, andWeng X-D. MiR-543 promotes cell proliferation and metastasis of renal cell carcinoma by targeting Dickkopf 1 through theWnt/β-catenin signaling pathway. J Cancer, 2018, 9(20):3660–8., DOI 10.7150/jca.27124 Gao RL, Chen XR, Li YN, Yan XY, Sun JG, and He QL. Upregulation of miR- 543-3p promotes growth and stem cell-like phenotype in bladder cancer by activating the Wnt/β-catenin signaling pathway. Int J Clin Exp Pathol, 2017, 10(9):9418–26. Lv Y, Zhang L,Ma J, Fei X, Xu K, and Lin J. CTHRC1 overexpression promotes ectopic endometrial stromal cell proliferation, migration and invasion via activation of theWnt/β-catenin pathway. Reprod Biomed Online, 2020, 40(1): 26–32., DOI 10.1016/j.rbmo.2019.10.001 Yi T, Liu M, Li X, Liu X, Ding Y, and He J. Benzo(a)pyrene inhibits endometrial cell apoptosis in early pregnant mice via the WNT5A pathway. J Cel Physiol, 2019, 234(7):11119–29., DOI 10.1002/jcp.27762 Zhou Q, Yan G, Ding L, Liu J, Yu X, and Kong S. EHD1 impairs decidualization by regulating the Wnt4/β-catenin signaling pathway in recurrent implantation failure. EBioMedicine, 2019, 50:343–54., DOI 10. 1016/j.ebiom.2019.10.018 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609761 EP 1609771 DI 10.3389/pore.2021.1609761 PG 11 ER PT J AU Wu, B Tang, X Ke, H Zhou, Q Zhou, Z Tang, Sh Ke, R AF Wu, Baojin Tang, Xinjie Ke, Honglin Zhou, Qiong Zhou, Zhaoping Tang, Shao Ke, Ronghu TI Gene Regulation Network of Prognostic Biomarker YAP1 in Human Cancers: An Integrated Bioinformatics Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer; prognosis; YAP1; bioinformatic; mitochondrial ID cancer; prognosis; YAP1; bioinformatic; mitochondrial AB Background: Yes-associated protein 1 (YAP1) is the main downstream effector of the Hippo signaling pathway, which is involved in tumorigenesis. This study aimed to comprehensively understand the prognostic performances of YAP1 expression and its potential mechanism in pan-cancers by mining databases. Methods: The YAP1 expression was evaluated by the Oncomine database and GEPIA tool. The clinical significance of YAP1 expression was analyzed by the UALCAN, GEPIA, and DriverDBv3 database. Then, the co-expressed genes with YAP1 were screened by the LinkedOmics, and annotated by the Metascape and DAVID database. Additionally, by the MitoMiner 4.0 v tool, the YAP1 co-expressed genes were screened to obtain the YAP1-associated mitochondrial genes that were further enriched by DAVID and analyzed by MCODE for the hub genes. Results: YAP1 was differentially expressed in human cancers. Higher YAP1 expression was significantly associated with poorer overall survival and disease-free survival in adrenocortical carcinoma (ACC), brain Lower Grade Glioma (LGG), and pancreatic adenocarcinoma (PAAD). The LinkedOmics analysis revealed 923 co-expressed genes with YAP1 in adrenocortical carcinoma, LGG and PAAD. The 923 genes mainly participated in mitochondrial functions including mitochondrial gene expression and mitochondrial respiratory chain complex I assembly. Of the 923 genes, 112 mitochondrial genes were identified by MitoMiner 4.0 v and significantly enriched in oxidative phosphorylation. The MCODE analysis identified three hub genes including CHCHD1, IDH3G and NDUFAF5. Conclusion: Our findings showed that the YAP1 overexpression could be a biomarker for poor prognosis in ACC, LGG and PAAD. Specifically, the YAP1 co-expression genes were mainly involved in the regulation of mitochondrial function especially in oxidative phosphorylation. Thus, our findings provided evidence of the carcinogenesis of YAP1 in human cancers and new insights into the mechanisms underlying the role of YAP1 in mitochondrial dysregulation. C1 [Wu, Baojin] Fudan University, Huashan Hospital, Department of Plastic SurgeryShanghai, China. [Tang, Xinjie] Fudan University, Huashan Hospital, Department of Plastic SurgeryShanghai, China. [Ke, Honglin] Fudan University, Huashan Hospital, Department of EmergencyShanghai, China. [Zhou, Qiong] Florida State University, Department of StatisticsTallahassee, FL, USA. [Zhou, Zhaoping] Fudan University, Huashan Hospital, Department of Plastic SurgeryShanghai, China. [Tang, Shao] Florida State University, Department of StatisticsTallahassee, FL, USA. [Ke, Ronghu] Fudan University, Huashan Hospital, Department of Plastic SurgeryShanghai, China. RP Ke, R (reprint author), Fudan University, Huashan Hospital, Department of Plastic Surgery, Shanghai, China. EM ronghuke@163.com CR Solanki S, Chakinala RC, Haq KF, Khan MA, Kifayat A, Linder K, et al. Inpatient burden of Gastric Cancer in the United States. Ann Transl Med, 2019, 7(23):772., DOI 10.21037/atm.2019.11.54 Miller KD, Nogueira L, Mariotto AB, Rowland JH, Yabroff KR, Alfano CM, et al. Cancer Treatment and Survivorship Statistics, 2019. CA A Cancer J Clin, 2019, 69(5):363–85., DOI 10.3322/caac.21565 Huang J, Wu S, Barrera J, Matthews K, and Pan D The Hippo Signaling Pathway Coordinately Regulates Cell Proliferation and Apoptosis by Inactivating Yorkie, the DrosophilaHomolog ofYAP. Cell, 2005, 122(3):421–34., DOI 10.1016/j.cell.2005.06.007 Liu JY, Li YH, Lin HX, Liao YJ, Mai SJ, Liu ZW, et al. Overexpression of YAP 1 Contributes to Progressive Features and Poor Prognosis of Human Urothelial Carcinoma of the Bladder. BMC Cancer, 2013, 13:349., DOI 10.1186/1471-2407-13-349 Guo L, Chen Y, Luo J, Zheng J, and Shao G YAP 1 Overexpression Is Associated with Poor Prognosis of Breast Cancer Patients and Induces Breast Cancer Cell Growth by Inhibiting PTEN. FEBS Open Bio, 2019, 9(3):437–45., DOI 10.1002/2211-5463.12597 Yu L, Gao C, Feng B, Wang L, Tian X, Wang H, et al. Distinct Prognostic Values of YAP1 in Gastric Cancer. Tumour Biol, 2017, 39(4): 1010428317695926., DOI 10.1177/1010428317695926 Eun Y-G, Lee D, Lee YC, Sohn BH, Kim EH, Yim SY, et al. Clinical Significance of YAP1 Activation in Head and Neck Squamous Cell Carcinoma. Oncotarget, 2017, 8(67):111130–43., DOI 10.18632/oncotarget.22666 Dong T, Yuan Y, Xiang X, Sang S, Shen H, Wang L, et al. High Cytoplasmic YAP1 Expression Predicts a Poor Prognosis in Patients with Colorectal Cancer. PeerJ, 2020, 8:e10397., DOI 10.7717/peerj.10397 Zhou Q, Bauden M, Andersson R, Hu D, Marko-Varga G, Xu J, et al. YAP1 Is an Independent Prognostic Marker in Pancreatic Cancer and Associated with Extracellular Matrix Remodeling. J Transl Med, 2020, 18(1):77., DOI 10.1186/ s12967-020-02254-7 Dong J, Feldmann G, Huang J, Wu S, Zhang N, Comerford SA, et al. Elucidation of a Universal Size-Control Mechanism in Drosophila and Mammals. Cell, 2007, 130(6):1120–33., DOI 10.1016/j.cell.2007.07.019 Lapi E, Di Agostino S, Donzelli S, Gal H, Domany E, Rechavi G, et al. PML, YAP, and P73 Are Components of a Proapoptotic Autoregulatory Feedback Loop. Mol Cel, 2008, 32(6):803–14., DOI 10.1016/j.molcel.2008.11.019 Guichet P-O, Masliantsev K, Tachon G, Petropoulos C, Godet J, Larrieu D, et al. Fatal Correlation between YAP1 Expression and Glioma Aggressiveness: Clinical and Molecular Evidence. J Pathol, 2018, 246(2):205–16., DOI 10.1002/path.5133 Giraud J, Molina-Castro S, Seeneevassen L, Sifre E, Izotte J, Tiffon C, et al. Verteporfin Targeting YAP1/TAZ-TEAD Transcriptional Activity Inhibits the Tumorigenic Properties of Gastric Cancer Stem Cells. Int J Cancer, 2020, 146(8):2255–67., DOI 10.1002/ijc.32667 Rhodes DR, Kalyana-Sundaram S, Mahavisno V, Varambally R, Yu J, Briggs BB, et al. Oncomine 3.0: Genes, Pathways, and Networks in a Collection of 18,000 Cancer Gene Expression Profiles. Neoplasia, 2007, 9(2):166–80., DOI 10. 1593/neo.07112 Tang Z, Li C, Kang B, Gao G, Li C, and Zhang Z GEPIA: a Web Server for Cancer and normal Gene Expression Profiling and Interactive Analyses. Nucleic Acids Res, 2017, 45(W1):W98–W102., DOI 10.1093/nar/gkx247 Wei R, Qiu H, Xu J, Mo J, Liu Y, Gui Y, et al. Expression and Prognostic Potential of GPX1 in Human Cancers Based on Data Mining. Ann Transl Med, 2020, 8(4):124., DOI 10.21037/atm.2020.02.36 Liu S-H, Shen P-C, Chen C-Y, Hsu A-N, Cho Y-C, Lai Y-L, et al. DriverDBv3: a Multi-Omics Database for Cancer Driver Gene Research. Nucleic Acids Res, 2020, 48(D1):D863–D870., DOI 10.1093/nar/gkz964 Liu Y, Yang Y, Luo Y, Wang J, Lu X, Yang Z, et al. Prognostic Potential of PRPF3 in Hepatocellular Carcinoma. Aging, 2020, 12(1):912–30., DOI 10. 18632/aging.102665 Noli L, Khorsandi SE, Pyle A, Giritharan G, Fogarty N, Capalbo A, et al. Effects of Thyroid Hormone on Mitochondria and Metabolism of Human Preimplantation Embryos. Stem Cells, 2020, 38(3):369–81., DOI 10.1002/stem.3129 Zhou Z, Wu B, Tang X, Ke R, and Zou Q Comprehensive Analysis of Fibroblast Growth Factor Receptor, FGFR, Family Genes in Breast Cancer by Integrating Online Databases and Bioinformatics. Med Sci Monit, 2020, 26: e923517., DOI 10.12659/msm.923517 Xu B-F, Liu R, Huang C-X, He B-S, Li G-Y, Sun H-S, et al. Identification of Key Genes in Ruptured Atherosclerotic Plaques by Weighted Gene Correlation Network Analysis. Sci Rep, 2020, 10(1):10847., DOI 10.1038/s41598-020-67114-2 Luo Y, Ma J, and Lu W The Significance of Mitochondrial Dysfunction in Cancer. Int J Mol Sci, 2020, 21(16):5598., DOI 10.3390/ijms21165598 Mammoto A, Muyleart M, Kadlec A, Gutterman D, and Mammoto T YAP1- TEAD1 Signaling Controls Angiogenesis and Mitochondrial Biogenesis through PGC1α. Microvasc Res, 2018, 119:73–83., DOI 10.1016/j.mvr.2018.04.003 Li L, BertramS, Kaplan J, Jia X, andWard DMThe Mitochondrial Iron Exporter GenesMMT1 and MMT2 in Yeast Are Transcriptionally Regulated by Aft1 and Yap1. J Biol Chem, 2020, 295(6):1716–26., DOI 10.1074/jbc.ra119.011154 Li H, He F, Zhao X, Zhang Y, Chu X, Hua C, et al. YAP Inhibits the Apoptosis and Migration of Human Rectal Cancer Cells via Suppression of JNK-Drp1-Mitochondrial Fission-HtrA2/Omi Pathways. Cell Physiol Biochem, 2017, 44(5):2073–89., DOI 10.1159/000485946 Ye S, Xu M, Zhu T, Chen J, Shi S, Jiang H, et al. Cytoglobin Promotes Sensitivity to Ferroptosis by Regulating p53-YAP1 axis in colon Cancer Cells. J Cel Mol Med, 2021, 25(7):3300–11., DOI 10.1111/jcmm.16400 Guo J, Kleeff J, Zhao Y, Li J, Giese T, Esposito I, et al. Yes-associated Protein, YAP65, in Relation to Smad7 Expression in Human Pancreatic Ductal Adenocarcinoma. Int J Mol Med, 2006, 17(5):761–7., DOI 10.3892/ijmm.17. 5.761 Yang Y, Zheng Y, Liu X, Ji R, Chen Z, Guo Q, et al. Comprehensive Analysis of Gene Regulation Network and Immune Signatures of Prognostic Biomarker YAP1 in Pancreatic Cancer. J Cancer, 2020, 11(23):6960–9., DOI 10.7150/jca. 49117 Orr BA, Bai H, Odia Y, Jain D, Anders RA, and Eberhart CG Yes-associated Protein 1 Is Widely Expressed in Human Brain Tumors and Promotes Glioblastoma Growth. J Neuropathol Exp Neurol, 2011, 70(7):568–77., DOI 10.1097/nen.0b013e31821ff8d8 Sang W, Xue J, Su LP, Gulinar A, Wang Q, Zhai YY, et al. Expression of YAP1 and pSTAT3-S727 and Their Prognostic Value in Glioma. J Clin Pathol, 2020, jclinpath:2020–206868., DOI 10.1136/jclinpath-2020-206868 Shen J, Cao B, Wang Y, Ma C, Zeng Z, Liu L, et al. Hippo Component YAP Promotes Focal Adhesion and Tumour Aggressiveness via Transcriptionally Activating THBS1/FAK Signalling in Breast Cancer. J Exp Clin Cancer Res, 2018, 37(1):175., DOI 10.1186/s13046-018-0850-z Azad T, Rezaei R, Surendran A, Singaravelu R, Boulton S, Dave J, et al. Hippo Signaling Pathway as a Central Mediator of Receptors Tyrosine Kinases, RTKs, in Tumorigenesis. Cancers, Basel,, 2020, 12(8)., DOI 10.3390/ cancers12082042 Azad T, Nouri K, Janse van Rensburg HJ, Maritan SM, Wu L, Hao Y, et al. A Gain-Of-Functional Screen Identifies the Hippo Pathway as a central Mediator of Receptor Tyrosine Kinases during Tumorigenesis. Oncogene, 2020, 39(2): 334–55., DOI 10.1038/s41388-019-0988-y Ashton TM, McKenna WG, Kunz-Schughart LA, and Higgins GS Oxidative Phosphorylation as an Emerging Target in Cancer Therapy. Clin Cancer Res, 2018, 24(11):2482–90., DOI 10.1158/1078-0432.ccr-17-3070 Lonardo E, Cioffi M, Sancho P, Sanchez-Ripoll Y, Trabulo SM, Dorado J, et al. Metformin Targets the Metabolic Achilles Heel of Human Pancreatic Cancer Stem Cells. PLoS One, 2013, 8(10):e76518., DOI 10.1371/journal.pone.0076518 Zhang X, Liu P, Shang Y, Kerndl H, Kumstel S, Gong P, et al. Metformin and LW6 Impairs Pancreatic Cancer Cells and Reduces Nuclear Localization of YAP1. J Cancer, 2020, 11(2):479–87., DOI 10.7150/jca.33029 White SM, Avantaggiati ML, Nemazanyy I, Di Poto C, Yang Y, Pende M, et al. YAP/TAZ Inhibition Induces Metabolic and Signaling Rewiring Resulting in Targetable Vulnerabilities in NF2-Deficient Tumor Cells. Developmental Cel, 2019, 49(3):425–43.e9., DOI 10.1016/j.devcel.2019.04.014 Deng Q, Guo T, Zhou X, Xi Y, Yang X, and Ge W Cross-Talk between Mitochondrial Fusion and the Hippo Pathway in Controlling Cell Proliferation during Drosophila Development. Genetics, 2016, 203(4):1777–88., DOI 10. 1534/genetics.115.186445 Koc EC, Cimen H, Kumcuoglu B, Abu N, Akpinar G, Haque ME, et al. Identification and Characterization of CHCHD1, AURKAIP1, and CRIF1 as New Members of the Mammalian Mitochondrial Ribosome. Front Physiol, 2013, 4:183., DOI 10.3389/fphys.2013.00183 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609768 EP 1609780 DI 10.3389/pore.2021.1609768 PG 13 ER PT J AU Bogos, K Kiss, Z Kerpel Fronius, A Temesi, G Elek, J Madurka, I Cselko, Zs Csanyi, P Abonyi-Toth, Zs Rokszin, Gy Barcza, Zs Moldvay, J AF Bogos, Krisztina Kiss, Zoltan Kerpel Fronius, Anna Temesi, Gabriella Elek, Jen}o Madurka, Ildiko Cselko, Zsuzsanna Csanyi, Peter Abonyi-Toth, Zsolt Rokszin, Gyorgy Barcza, Zsofia Moldvay, Judit TI Different Trends in Excess Mortality in a Central European Country Compared to Main European Regions in the Year of the COVID-19 Pandemic (2020): a Hungarian Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hungary; covid-19 pandemic; excess mortality; cumulative death; age-standardized mortality rate ID Hungary; covid-19 pandemic; excess mortality; cumulative death; age-standardized mortality rate AB Objective: This study examined cumulative excess mortality in European countries in the year of the Covid-19 pandemic and characterized the dynamics of the pandemic in different countries, focusing on Hungary and the Central and Eastern European region. Methods: Age-standardized cumulative excess mortality was calculated based on weekly mortality data from the EUROSTAT database, and was compared between 2020 and the 2016–2019 reference period in European countries. Results: Cumulate weekly excess mortality in Hungary was in the negative range until week 44. By week 52, it reached 9,998 excess deaths, corresponding to 7.73% cumulative excess mortality vs. 2016–2019 (p-value = 0.030 vs. 2016–2019). In Q1, only Spain and Italy reported excess mortality compared to the reference period. Significant increases in excess mortality were detected between weeks 13 and 26 in Spain, United Kingdom, Belgium, Netherland and Sweden. Romania and Portugal showed the largest increases in agestandardized cumulative excess mortality in the Q3. The majority of Central and Eastern European countries experienced an outstandingly high impact of the pandemic in Q4 in terms of excess deaths. Hungary ranked 11th in cumulative excess mortality based on the latest available data of from the EUROSTAT database. Conclusion: Hungary experienced a mortality deficit in the first half of 2020 compared to previous years, which was followed by an increase in mortality during the second wave of the COVID-19 pandemic, reaching 7.7% cumulative excess mortality by the end of 2020. The excess was lower than in neighboring countries with similar dynamics of the pandemic. C1 [Bogos, Krisztina] National Koranyi Institute of PulmonologyBudapest, Hungary. [Kiss, Zoltan] University of Pecs, 2nd Department of Medicine and Nephrological CenterPecs, Hungary. [Kerpel Fronius, Anna] National Koranyi Institute of PulmonologyBudapest, Hungary. [Temesi, Gabriella] National Koranyi Institute of PulmonologyBudapest, Hungary. [Elek, Jen}o] National Koranyi Institute of PulmonologyBudapest, Hungary. [Madurka, Ildiko] National Koranyi Institute of PulmonologyBudapest, Hungary. [Cselko, Zsuzsanna] National Koranyi Institute of PulmonologyBudapest, Hungary. [Csanyi, Peter] National Koranyi Institute of PulmonologyBudapest, Hungary. [Abonyi-Toth, Zsolt] RxTarget LtdSzolnok, Hungary. [Rokszin, Gyorgy] RxTarget LtdSzolnok, Hungary. [Barcza, Zsofia] Syntesia LtdBudapest, Hungary. [Moldvay, Judit] National Koranyi Institute of Pulmonology, Semmelweis University, Ist Department of PulmonologyBudapest, Hungary. RP Kiss, Z (reprint author), University of Pecs, 2nd Department of Medicine and Nephrological Center, Pecs, Hungary. EM dr.zoltan.kiss.privat@gmail.com CR European Centre for Disease Prevention and Control, ECDC). Rapid Risk Assessment: outbreak of acute respiratory syndrome associated with a novel coronavirus, Wuhan, China; first update – 22 January 2020. Available from: https://www.ecdc.europa.eu/en/publications-data/risk-assessmentoutbreak- acute-respiratory-syndrome-associated-novel-coronavirus, Accessed February 1, 2021). Stockholm: ECDC, 2020). European Center for Disease Prevention and Control, ECDC). Timeline of ECDC’s response to COVID-19. Available from: https://www.ecdc.europa.eu/ en/covid-19/timeline-ecdc-response, Accessed February 5, 2021). World Health Organization, WHO). Listings ofWHO’s response to COVID-19. Available from: https://www.who.int/news/item/29-06-2020-covidtimeline, Accessed February 5, 2021). World Health Organization, WHO). A timeline of WHO’s response to COVID 19 in the WHO European Region. Available from: https://apps. who.int/iris/bitstream/handle/10665/334205/70id07e-COVID19-Timeline- 200749.pdf?sequence1&isAllowedy, Accessed January 29, 2021). World Health Organization, WHO). WHO coronavirus disease, COVID-19, dashboard. Available from: https://covid19.who.int, Accessed January 26, 2021). European Centre for Disease Prevention and Control, ECDC). COVID-19. Available from: https://qap.ecdc.europa.eu/public/extensions/COVID-19/ COVID-19.html, Accessed January 25, 2021, Stockholm: ECDC. Fuller JA, Hakim A, Victory KR, Date K, Lynch M, Dahl B, et al.CDC COVID-19 Response Team. Mitigation policies and COVID-19- associated mortality - 37 European countries, january 23-june 30, 2020. MMWR Morb Mortal Wkly Rep, 2021, 70(2):58–62., DOI 10.15585/mmwr. mm7002e4 Karako K, Song P, Chen Y, Tang W, and Kokudo N. Overview of the characteristics of and responses to the threewaves of COVID-19 in Japan during 2020-2021. Biosci Trends, 2021, 15(1):1–8., DOI 10.5582/bst.2021.01019 Rampal L, and Liew BS. Malaysia’s third COVID-19 wave - a paradigm shift required. Med J Malaysia, 2021, 76(1):1–4. Available from: http://www.emjm. org/2021/v76n1/third-COVID-19-wave.pdf. Aleta A, and Moreno Y. Age differential analysis of COVID-19 second wave in Europe reveals highest incidence among young adults. Medrxiv [Preprint], 2020). Available from: https://www.medrxiv.org/content/10.1101/2020.11. 20230177v1, Accessed January 27, 2021)., DOI 10.1101/2020.11.11. 20230177 11. Checchi F, and Roberts L. Interpreting and using mortality data in humanitarian emergencies. London,UK:Humanitarian PracticeNetwork, 2005). 52 p. Available from: https://odihpn.org/resources/interpreting-and-using-mortality-data-inhumanitarian- emergencies/, Accessed February 5, 2021). European Mortality Monitoring, EuroMOMO). Substantial increased excess mortality seen in Europe. EuroMOMO Bulletin, Week 2, 2021. Available from: https://www.euromomo.eu, Accessed January 27, 2021). Eurostat. Excess mortality - statistics. Available from: https://ec.europa.eu/ eurostat/statistics-explained/index.php?titleExcess_mortality_-_statistics#Excess _mortality_in_Europe_over_January-November_2020, Accessed March 4, 2021). European Mortality Monitoring, EuroMOMO). Available from: https://www. euromomo.eu/graphs-and-maps/, Accessed February 5, 2021). Kontis V, Bennett JE, Rashid T, Parks RM, Pearson-Stuttard J, Guillot M, et al. Magnitude, demographics and dynamics of the effect of the first wave of the COVID-19 pandemic on all-cause mortality in 21 industrialized countries. Nat Med, 2020, 26(12):1919–1928., DOI 10.1038/s41591-020- 1112-0 Rocks S, and Idriss O. Did hospital capacity affect mortality during the pandemic’s first wave? Available from: https://www.health.org.uk/news-andcomment/ charts-and-infographics/did-hospital-capacity-affect-mortality -during-the-pandemic, Accessed November 7, 2020). Stoddard M, Van Egeren D, Johnson K, Rao S, Furgeson J, White DE, et al. Model-based evaluation of the impact of noncompliance with public health measures on COVID-19 disease control. Available from: , DOI 10. 1101/2020.11.29.20240440https://www.medrxiv.org/content/10.1101/2020. 11.29.20240440v1.full.pdf, Accessed February 5, 2021). Ayoubkhani D, Khunti K, Nafilyan V, Maddox T, Humberstone B, Diamond I, et al. Epidemiology of post-COVID syndrome following hospitalisation with coronavirus: a retrospective cohort study. MedRxiv Available from: https://www.medrxiv.org/content/10.1101/2021.01.15.21249885v1, Accessed February 5, 2021)., DOI 10.1101/2021.01.15.21249885 Hu J, Li C, Wang S, Li T, and Zhang H. Genetic variants are identified to increase risk of COVID-19 related mortality from UK Biobank data. Hum Genomi, 2021, 15(1):10., DOI 10.1186/s40246-021-00306-7 Hale T, Angrist N, Cameron-Blake E, Hallas L, Kira B, Majumdar S, et al.Oxford COVID-19 government response tracker. England, UK: Blavatnik School of Government, 2020). Available from: https://www.bsg. ox.ac.uk/research/research-projects/coronavirus-government-response-tracker, Accessed February 1, 2021). Hirsch C. Europe’s coronavirus lockdownmeasures compared. Published March 31, 2020. Available at: https://www.politico.eu/article/europes-coronaviruslockdown- measures-compared/, Accessed February 1, 2021). Hungarian Central Statistical Office. Road accidents involving personal injury. 2016–2020. Available from: https://www.ksh.hu/docs/eng/xstadat/xstadat_ infra/e_ods002.html, Accessed on February 2, 2021). Meier T, Grafe K, Senn F, Sur P, Stangl GI, Dawczynski C, et al. Cardiovascular mortality attributable to dietary risk factors in 51 countries in the WHO European region from 1990 to 2016: a systematic analysis of the global burden of disease study. Eur J Epidemi, 2019, 34(1):37–55., DOI 10.1007/s10654-018- 0473-x OECD/European Union. Health expenditure per capita. in: Health at a glance: Europe 2018: state of health in the EU cycle. Brussels: OECD Publishing, Paris/ European Union. Published December 14, 2018. Available from:, DOI 10.1787/ health_glance_eur-2018-29-en Katsoulis M, Gomes M, Lai AG, Henry A, Denaxas S, Lagiou P, et al. Estimating the effect of reduced attendance at emergency departments for suspected cardiac conditions on cardiac mortality during the COVID-19 pandemic. Circ Cardiovasc Qual Outcomes, 2021, 14(1):e007085., DOI 10. 1161/CIRCOUTCOMES.120.007085 Solomon MD, McNulty EJ, Rana JS, Leong TK, Lee C, Sung S-H, et al. The covid-19 pandemic and the incidence of acute myocardial infarction. N Engl J Med, 2020, 383(7):691–3., DOI 10.1056/NEJMc2015630 De Filippo O, D’Ascenzo F, Angelini F, Bocchino PP, Conrotto F, Saglietto A, et al. Reduced rate of hospital admissions for ACS during covid-19 outbreak in northern Italy. N Engl JMed, 2020, 383(1):88–89., DOI 10.1056/ NEJMc2009166 World Health Organization, WHO). How comparable is covid-19 mortality across countries? Published June 4, 2020. Available from: https://analysis. covid19healthsystem.org/index.php/2020/06/04/how-comparable-is-covid- 19-mortality-across-countries/(Accessed May 19, 2020). Spencer MR, and Ahmad F. Timeliness of death certificate data for mortality surveillance and provisional estimates. Waltham, MA: National Center for Health Statistics, 2017, Report No.: 001. Available from: https://www.cdc.gov/ nchs/data/vsrr/report001.pdf, Accessed February 5, 2021). NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609774 EP 1609782 DI 10.3389/pore.2021.1609774 PG 9 ER PT J AU Qin, J Xie, F Li, Ch Han, N Lu, H AF Qin, Jing Xie, Fajun Li, Chenghui Han, Na Lu, Hongyang TI MYCL1 Amplification and Expression of L-Myc and c-Myc in Surgically Resected Small-Cell Lung Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; small-cell lung carcinoma; C-MYC; fluorescence in situ hybridization; MYCL1 amplification; L-myc ID immunohistochemistry; small-cell lung carcinoma; C-MYC; fluorescence in situ hybridization; MYCL1 amplification; L-myc AB Purpose: The Myc family, especially C-MYC and MYCL1, has been found involved in small-cell lung carcinoma (SCLC). Identification of the frequency of C-MYC and MYCL1 expression among SCLC patients may help to identify potential targets for therapeutic intervention. Our aim was to detect MYCL1 amplification, L-Myc and c-Myc expression, and investigate clinicopathological characteristics and survival status in patients with surgically resected SCLC. Methods: MYCL1 amplification was detected using fluorescence in situ hybridization (FISH), while L-Myc and c-Myc protein expressions were determined using immunohistochemistry (IHC) in the primary tumors of 46 resected SCLC patients. Results: Among the 46 evaluated specimens, MYCL1 amplification was identified in 3/46 cases (6.5%). One of the positive cases was MYCL1 gene amplification combined with fusion. 3/46 (6.5%) was positive for L-myc protein expression, and 4/46 (8.7%) was positive for c-Myc protein expression. Conclusion: Our study firstly multidimensional explored the expression of MYCL1 amplification, L-Myc and c-Myc protein and investigated clinicopathological characteristics and survival status in patients with surgically resected SCLC, which makes a contribution to subsequent research and therapeutic strategies. C1 [Qin, Jing] Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Department of Thoracic Medical OncologyHangzhou, China. [Xie, Fajun] Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Department of Thoracic Medical OncologyHangzhou, China. [Li, Chenghui] Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Department of Thoracic Medical OncologyHangzhou, China. [Han, Na] Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Department of Thoracic Medical OncologyHangzhou, China. [Lu, Hongyang] Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Department of Thoracic Medical OncologyHangzhou, China. RP Lu, H (reprint author), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Department of Thoracic Medical Oncology, Hangzhou, China. EM luhy@zjcc.org.cn CR Siegel RL, Miller KD, Fuchs HE, and Jemal A. Cancer Statistics, 2021. CA A Cancer J Clin, 2021, 71(1):7–33., DOI 10.3322/caac.21654 ChenW, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer Statistics in China, 2015. CA: a Cancer J clinicians, 2016, 66(2):115–32., DOI 10.3322/caac.21338 Bernhardt EB, and Jalal SI. Small Cell Lung Cancer. Cancer Treat Res, 2016, 170:301–22., DOI 10.1007/978-3-319-40389-2_14 IamsWT, Porter J, and Horn L. Immunotherapeutic Approaches for Small-Cell Lung Cancer. Nat Rev Clin Oncol, 2020, 17(5):300–12., DOI 10.1038/s41571-019-0316-z Iwakawa R, Takenaka M, Kohno T, Shimada Y, Totoki Y, Shibata T, et al. Genome-wide Identification of Genes with Amplification And/or Fusion in Small Cell Lung Cancer. Genes Chromosomes Cancer, 2013, 52(9):802–16., DOI 10.1002/gcc.22076 Ireland AS, Micinski AM, Kastner DW, Guo B,Wait SJ, Spainhower KB, et al. MYC Drives Temporal Evolution of Small Cell Lung Cancer Subtypes by Reprogramming Neuroendocrine Fate. Cancer cell, 2020, 38(1):60–78., DOI 10.1016/j.ccell.2020.05.001 Hwang DH, Sun H, Rodig SJ, Hornick JL, and Sholl LM. Myc Protein Expression Correlates withMYCamplification in Small-Cell Lung Carcinoma. Histopathology, 2015, 67(1):81–9., DOI 10.1111/his.12622 Wasylishen AR, Stojanova A, Oliveri S, Rust AC, Schimmer AD, and Penn LZ. New Model Systems Provide Insights into Myc-Induced Transformation. Oncogene, 2011, 30(34):3727–34., DOI 10.1038/ onc.2011.88 Nakagawa M, Takizawa N, Narita M, Ichisaka T, and Yamanaka S. Promotion of Direct Reprogramming by Transformation-Deficient Myc. Proc Natl Acad Sci, 2010, 107(32):14152–7., DOI 10.1073/pnas.1009374107 Kc W, Satpathy AT, Rapaport AS, Briseno CG, Wu X, Albring JC, et al. L-myc Expression by Dendritic Cells Is Required for Optimal T-Cell Priming. Nature, 2014, 507(7491):243–7., DOI 10.1038/nature12967 Chen S, Tang J, Huang L, and Lin J. Expression and Prognostic Value of Mycl1 in Gastric Cancer. Biochem biophysical Res Commun, 2015, 456(4):879–83., DOI 10.1016/j.bbrc.2014.12.060 Borromeo MD, Savage TK, Kollipara RK, He M, Augustyn A, Osborne JK, et al. ASCL1 and NEUROD1 Reveal Heterogeneity in Pulmonary Neuroendocrine Tumors and Regulate Distinct Genetic Programs. Cel Rep, 2016, 16(5):1259–72., DOI 10.1016/j.celrep.2016.06.081 Xiong F, Wu C, Chang J, Yu D, Xu B, Yuan P, et al. Genetic Variation in an miRNA-1827 Binding Site in MYCL1 Alters Susceptibility to Small-Cell Lung Cancer. Cancer Res, 2011, 71(15):5175–81., DOI 10.1158/0008-5472.CAN-10-4407 Kim YH, Girard L, Giacomini CP, Wang P, Hernandez-Boussard T, Tibshirani R, et al. Combined Microarray Analysis of Small Cell Lung Cancer Reveals Altered Apoptotic Balance and Distinct Expression Signatures of MYC Family Gene Amplification. Oncogene, 2006, 25(1):130–8., DOI 10.1038/ sj.onc.1208997 Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, et al.WHO Panel. The 2015 World Health Organization Classification of Lung Tumors. J Thorac Oncol, 2015, 10(9):1243–60., DOI 10.1097/ JTO.0000000000000630 Nicholson AG, Chansky K, Crowley J, Beyruti R, Kubota K, Turrisi A, et al. Staging and Prognostic Factors Committee, Advisory Boards, and Participating Institutions, & Staging and Prognostic Factors Committee Advisory Boards and Participating InstitutionsThe International Association for the Study of Lung Cancer Lung Cancer Staging Project: Proposals for the Revision of the Clinical and Pathologic Staging of Small Cell Lung Cancer in the Forthcoming Eighth Edition of the TNMClassification for Lung Cancer. J ThoracOncol : official Publ Int Assoc Study Lung Cancer, 2016, 11(3):300–11., DOI 10.1016/j.jtho.2015.10.008 Tian X, Xu J, and Dorfman DM. Utility of Combined EZH2, P-Erk1/2, P-STAT, and MYC Expression in the Differential Diagnosis of EZH2- Positive Hodgkin Lymphomas and Related Large B-Cell Lymphomas. Am J Surg Pathol, 2019, 43(1):102–9., DOI 10.1097/PAS.0000000000001180 Kato F, Fiorentino FP, Alibes A, Perucho M, Sanchez-Cespedes M, Kohno T, et al. MYCL Is a Target of a BET Bromodomain Inhibitor, JQ1, on Growth Suppression Efficacy in Small Cell Lung Cancer Cells. Oncotarget, 2016, 7(47): 77378–88., DOI 10.18632/oncotarget.12671 Bragelmann J, Bohm S, Guthrie MR, Mollaoglu G, Oliver TG, and Sos ML. Family Matters: How MYC Family Oncogenes Impact Small Cell Lung Cancer. Cell Cycle, 2017, 16(16):1489–98., DOI 10.1080/15384101.2017.1339849 Cheng J, Park DE, Berrios C, White EA, Arora R, Yoon R, et al. Merkel Cell Polyomavirus Recruits MYCL to the EP400 Complex to Promote Oncogenesis. Plos Pathog, 2017, 13(10):e1006668e1006668., DOI 10.1371/ journal.ppat.1006668 Alves RC, Meurer RT, and Roehe AV. MYC Amplification Is Associated with Poor Survival in Small Cell Lung Cancer: a Chromogenic In Situ Hybridization Study. J Cancer Res Clin Oncol, 2014, 140(12):2021–5., DOI 10.1007/s00432- 014-1769-1 Almodovar K, Iams WT, Meador CB, Zhao Z, York S, Horn L, et al. Longitudinal Cell-free DNA Analysis in Patients with Small Cell Lung Cancer Reveals Dynamic Insights into Treatment Efficacy and Disease Relapse. J Thorac Oncol, 2018, 13(1):112–23., DOI 10.1016/ j.jtho.2017.09.1951 Kambara T, Sharp GB, Nagasaka T, Takeda M, SasamotoH, Nakagawa H, et al. Allelic Loss of a Common Microsatellite Marker MYCL1. Clin Cancer Res, 2004, 10(5):1758–63., DOI 10.1158/1078-0432.ccr-0779-3 Rudin CM, Durinck S, Stawiski EW, Poirier JT,Modrusan Z, Shames DS, et al. Comprehensive Genomic Analysis Identifies SOX2 as a Frequently Amplified Gene in Small-Cell Lung Cancer. Nat Genet, 2012, 44(10):1111–6., DOI 10.1038/ng.2405 Sos ML, Dietlein F, Peifer M, Schottle J, Balke-Want H, Muller C, et al. A Framework for Identification of Actionable Cancer Genome Dependencies in Small Cell Lung Cancer. Proc Natl Acad Sci, 2012, 109(42):17034–9., DOI 10.1073/pnas.1207310109 Melichar B, Adenis A, Lockhart AC, Bennouna J, Dees EC, Kayaleh O, et al. Safety and Activity of Alisertib, an Investigational aurora Kinase A Inhibitor, in Patients with Breast Cancer, Small-Cell Lung Cancer, Nonsmall- cell Lung Cancer, Head and Neck Squamous-Cell Carcinoma, and Gastro-Oesophageal Adenocarcinoma: a Five-Arm Phase 2 studyThe Lancet. Lancet Oncol, 2015, 16(4):395–405., DOI 10.1016/S1470-2045(15, 70051-3 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609775 EP 1609782 DI 10.3389/pore.2021.1609775 PG 8 ER PT J AU Kurdi, M Butt, ShN Baeesa, S Alghamdi, B Maghrabi, Y Bardeesi, A Saeedi, R Al-Sinani, T Alghanmi, N Bari, M Samkari, A Lary, A AF Kurdi, Maher Butt, Shafique Nadeem Baeesa, Saleh Alghamdi, Badrah Maghrabi, Yazid Bardeesi, Anas Saeedi, Rothaina Al-Sinani, Taghreed Alghanmi, Najla Bari, O. Mohammed Samkari, Alaa Lary, I. Ahmed TI The Impact of IDH1 Mutation and MGMT Promoter Methylation on Recurrence-Free Interval in Glioblastoma Patients Treated With Radiotherapy and Chemotherapeutic Agents SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE glioblastoma; chemotherapy; IDH1 mutation; MGMT promoter methylation; radiotherapy ID glioblastoma; chemotherapy; IDH1 mutation; MGMT promoter methylation; radiotherapy AB The aim of this study is to investigate the relationship between isocitrate dehydrogenase-1 (IDH1) mutation andO6-methylguanine-DNA methyltransferase (MGMT) promoter methylation with recurrence-free interval in glioblastoma patients treated with chemoradiotherapies. Clinical data were collected from 82 patients with totally resected glioblastoma and treated with adjuvant therapies from 2014 to 2019. IDH1 mutation was assessed by immunohistochemistry and MGMT promoter methylation was assessed by different sequencing methods. IDH1 mutation was present in 32 cases and 50 cases were IDH1 wildtype; 54 and 28 patients had unmethylated and methylated MGMT promoter, respectively, Of the 82 patients, 62 patients received chemoradiotherapy while 20 patients only received radiation. Approximately, 61% of patients had a tumor recurrence after 1 year, and 39% showed a recurrence before 1 year of treatment. There was no significant relationship between IDH1 mutation and MGMT promoter methylation (p-value = 0.972). Patients with IDH1 mutation and their age <50 years showed a significant difference in recurrence-free interval (p-value = 0.014). Difference in recurrence-free interval was also statistically observed in patients with unmethylated MGMT promoter and treated with chemoradiotherapies (p-value = 0.031), by which they showed a late tumor recurrence (p-value = 0.016). This revealed that IDH1 mutation and MGMT methylation are independent prognostic factors in glioblastoma. Although IDH1-mutant glioblastomas showed late tumor recurrence in patients less than 50 years old, the type of treatment modalitiesmay not show additional beneficial outcome. Patients with unmethylated MGMT and IDH1 mutation, treated with different chemoradiotherapies, showed a late tumor recurrence. C1 [Kurdi, Maher] King Abdulaziz University, Faculty of Medicine in Rabigh, Department of PathologyJeddah, Saudi Arabia. [Butt, Shafique Nadeem] King Abdulaziz University, Faculty of Medicine in Rabigh, Department of Family and Community MedicineJeddah, Saudi Arabia. [Baeesa, Saleh] King Abdulaziz University, Faculty of Medicine in Rabigh, Division of NeurosurgeryJeddah, Saudi Arabia. [Alghamdi, Badrah] King Abdulaziz University, Faculty of Medicine in Rabigh, Department of PhysiologyJeddah, Saudi Arabia. [Maghrabi, Yazid] King Faisal Specialist Hospital, Department of NeuroscienceJeddah, Saudi Arabia. [Bardeesi, Anas] King Faisal Specialist Hospital, Department of NeuroscienceJeddah, Saudi Arabia. [Saeedi, Rothaina] King Abdulaziz University, Faculty of Medicine in Rabigh, Division of NeurosurgeryJeddah, Saudi Arabia. [Al-Sinani, Taghreed] King Fahad General Hospital, Department of Surgery, Division of NeurosurgeryJeddah, Saudi Arabia. [Alghanmi, Najla] King Abdulaziz University Hospital, Department of PathologyJeddah, Saudi Arabia. [Bari, O. Mohammed] King Abdulaziz University Hospital, Department of PathologyJeddah, Saudi Arabia. [Samkari, Alaa] King Abdulaziz University, Department of PathologyJeddah, Saudi Arabia. [Lary, I. Ahmed] King Abdulaziz Medical City, Department of Surgery, Section of NeurosurgeryJeddah, Saudi Arabia. RP Kurdi, M (reprint author), King Abdulaziz University, Faculty of Medicine in Rabigh, Department of Pathology, Jeddah, Saudi Arabia. EM Ahkurdi@kau.edu.sa CR Louis DN, Ohgaki H, Wiestler OD, and Cavenee WK. World Health Organization Histiological Classification of Tumours of the Central Nervous System. France: International Agency for Research on Cancer, 2016). Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL, et al. Genetic pathways to glioblastoma. Cancer Res, 2004, 64(19):6892–9., DOI 10.1158/0008-5472.can-04-1337 Stupp R, Hegi ME, Mason WP, Van den Bent MJ, Taphoorn MJB, Janzer RC, et al., 2009, Effects of radiotherapy with concomitant and adjuvant temozolamide versus radiotherapy alone on survival in glioblastoma in a randomized phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10(5):459–66., DOI 10.1016/s1470-2045(09)70025-7 Hartmann C, Meyer J, Balss J, Capper D, Mueller W, Christians A, et al. Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas. Acta Neuropathol, 2009, 118:469–74., DOI 10.1007/s00401-009-0561-9 Capper D, WeiA¨ Yert S, Balss JR, Habel A, Meyer J, Jager D, et al. Characterization of R132H mutation-specific IDH1 antibody binding in brain tumors. Brain Pathol, 2010, 20:245–54., DOI 10.1111/j.1750-3639. 2009.00352.x Nobusawa S, Watanabe T, Kleihues P, and Ohgaki H. IDH1 mutations as molecular signature and predictive factor of secondary glioblastomas. Clin Cancer Res, 2009, 15(19):6002–7., DOI 10.1158/1078-0432.ccr-09-0715 Pegg AE. Repair of O6-alkylguanine by alkyltransferases. Mutat Research/ Reviews Mutat Res, 2000, 462:83–100., DOI 10.1016/s1383-5742(00)00017-x Ludlum DB. DNA alkylation by the haloethylnitrosoureas: nature of modifications produced and their enzymatic repair or removal. Mutat Research/Fundamental Mol Mech Mutagenesis, 1990, 233:117–26., DOI 10. 1016/0027-5107(90)90156-x Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMTGene silencing and benefit from temozolomide in glioblastoma. N Engl J Med, 2005, 352:997–1003., DOI 10.1056/nejmoa043331 Dunn J, Baborie A, Alam F, Joyce K, Moxham M, Sibson R, et al. Extent of MGMT promoter methylation correlates with outcome in glioblastomas given temozolomide and radiotherapy. Br J Cancer, 2009, 101:124–31., DOI 10.1038/ sj.bjc.6605127 Wick W, Hartmann C, Engel C, Stoffels M, Felsberg J, Stockhammer F, et al. NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. Jco, 2009, 27:5874–80., DOI 10.1200/jco.2009.23.6497 Costa BM, Caeiro C, Guimaraes I, Martinho O, Jaraquemada T, Augusto I, et al. Prognostic value of MGMT promoter methylation in glioblastoma patients treated with temozolomide-based chemoradiation: a Portuguese multicentre study. Oncol Rep, 2010, 23:1655–62., DOI 10.3892/or_00000808 Park C-K, Park S-H, Lee S-H, Kim C-Y, Kim D-W, Paek SH, et al. Methylation status of the MGMT gene promoter fails to predict the clinical outcome of glioblastoma patients treated with ACNU plus cisplatin. Neuropathology, 2009, 29:443–9., DOI 10.1111/j.1440-1789.2008.00998.x Millward CP, Brodbelt AR, Haylock B, Zakaria R, Baborie A, Crooks D, et al. The impact of MGMT methylation and IDH-1 mutation on long-term outcome for glioblastoma treated with chemoradiotherapy. Acta Neurochir, 2016, 158:1943–53., DOI 10.1007/s00701-016-2928-8 Esteller M, Hamilton SR, Burger PC, Baylin SB, and Herman JG. Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res, 1999, 59:793–7. Panagopoulos I, Gorunova L, Leske H, Niehusmann P, Johannessen LE, Staurseth J, et al. Pyrosequencing Analysis of MGMT promoter methylation in meningioma. Cancer Genomics Proteomics, 2018, 15: 379–85., DOI 10.21873/cgp.20096 Combs S, Reikan S, Wick W, Abdollah A, Deimling AV, Debus J, et al. Prognostic significance of IDH1 and MGMT in patients with glioblastoma: one step forward, and one step back. Radiat Oncol, 2011, 6: 115., DOI 10.1186/1748-717x-6-115 Alassiri AH, Alkhaibary A, Al-Sarheed S, Alsufani F, Alharbi M, Alkhani A, et al. O 6 -methylguanine-DNA methyltransferase promoter methylation and isocitrate dehydrogenase mutation as prognostic factors in a cohort of Saudi patients with glioblastoma. Ann Saudi Med, 2019, 39(6):410–6., DOI 10.5144/ 0256-4947.2019.410 Pandith AA, Qasim I, Baba SM, Koul A, Zahoor W, Afroz D, et al. Favorable role of IDH1/2 mutations aided with MGMT promoter gene methylation in the outcome of patients with malignant glioma. Future Sci OA, 2020, 7(3): FSO663., DOI 10.2144/fsoa-2020-0057 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609778 EP 1609786 DI 10.3389/pore.2021.1609778 PG 9 ER PT J AU Boer, K Kahan, Zs Landherr, L Cs}oszi, T Mahr, K Ruzsa, Horvath, Zs Budai, B Rubovszky, G AF Boer, Katalin Kahan, Zsuzsanna Landherr, Laszlo Cs}oszi, Tibor Mahr, Karoly Ruzsa, Agnes Horvath, Zsolt Budai, Barna Rubovszky, Gabor TI Pathologic Complete Response Rates After Neoadjuvant Pertuzumab and Trastuzumab with Chemotherapy in Early Stage HER2-Positive Breast Cancer - Increasing Rates of Breast Conserving Surgery: A Real-World Experience SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE early breast cancer; HER2+; neoadjuvant systemic therapy; pathological complete response; breastconserving surgery; neutrophil-to-lymphocyte ratio ID early breast cancer; HER2+; neoadjuvant systemic therapy; pathological complete response; breastconserving surgery; neutrophil-to-lymphocyte ratio AB Purpose: The neoadjuvant use of pertuzumab and trastuzumab with chemotherapy improves the pathologic complete response (pCR) in early HER2+ breast cancer. The aim of this study was to determine the pCR rate obtained with dual HER2 blockade in routine clinical practice. The secondary and tertiary objective was to investigate the impact of neoadjuvant systemic therapy (NST) on performing breast-conserving surgery and survival data. Methods: This was a multicentre, retrospective, observational study in patients with stage II and III HER2+ early breast cancer who received pertuzumab and trastuzumab-based NST. Data were collected from patients’ medical records. Results: Eighty-two patients were included in the study treated in 8 cancer centers in Hungary between March 2015 and January 2020. The study included women with a median age of 50.3 years. The majority of the patients (95%) received a sequence of anthracycline-based chemotherapy followed by docetaxel. pCR was achieved in 54% of the cases. As a result of NST a significant increase of conservative breast surgeries (33% vs. 3.6% planned, p = 0.0001) was observed. Ki67 expression and neutrophil-tolymphocyte ratio (NLR) significantly predicted pCR. None of the variables were independent predictors of DFS. Conclusion: The pCR rate achieved in our study demonstrates the reproducibility of trial data in a real-world population. The rate of breast-conserving surgery was significantly increased. C1 [Boer, Katalin] St. Margit Hospital, Clinical Oncology DepartmentBudapest, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of OncotherapySzeged, Hungary. [Landherr, Laszlo] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai KozpontBudapest, Hungary. [Cs}oszi, Tibor] Jasz-Nagykun-Szolnok Megyei Hetenyi Geza Korhaz, Klinikai OnkologiaSzolnok, Hungary. [Mahr, Karoly] Zala Megyei Szent Rafael Korhaz, OnkologiaZalaegerszeg, Hungary. [Ruzsa, Agnes] Somogy County Kaposi Mor Hospital, Department of OncologyKaposvar, Hungary. [Horvath, Zsolt] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. [Budai, Barna] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Rubovszky, Gabor] Semmelweis University, Department of OncologyBudapest, Hungary. RP Rubovszky, G (reprint author), Semmelweis University, Department of Oncology, Budapest, Hungary. EM garub@oncol.hu CR Early Breast Cancer Trialists’ Collaborative Group, EBCTCG). Long-term Outcomes for Neoadjuvant versus Adjuvant Chemotherapy in Early Breast Cancer: Meta-Analysis of Individual Patient Data from Ten Randomised Trials. Lancet Oncol, 2018, 19(1):27–39., DOI 10.1016/S1470-2045(17, 30777-5 Cain H, Macpherson IR, Beresford M, Pinder SE, Pong J, and Dixon JM. Neoadjuvant Therapy in Early Breast Cancer: Treatment Considerations and Common Debates in Practice. Clin Oncol, 2017, 29(10):642–52., DOI 10.1016/j. clon.2017.06.003 Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological Complete Response and Long-Term Clinical Benefit in Breast Cancer: the CTNeoBC Pooled Analysis. Lancet, 2014, 384(9938):164–72., DOI 10.1016/S0140-6736(13)62422-8 Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, et al. Early Breast Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol, 2019, 30(8):1194–220., DOI 10.1093/ annonc/mdz173 Burstein HJ, Curigliano G, Loibl S, Dubsky P, Gnant M, Poortmans P, et al. Estimating the Benefits of Therapy for Early-Stage Breast Cancer: the St. Gallen International Consensus Guidelines for the Primary Therapy of Early Breast Cancer 2019. Ann Oncol, 2019, 30(10):1541–57., DOI 10.1093/annonc/ mdz235 Buzdar AU, Ibrahim NK, Francis D, Booser DJ, Thomas ES, Theriault RL, et al. Significantly Higher Pathologic Complete Remission Rate after Neoadjuvant Therapy with Trastuzumab, Paclitaxel, and Epirubicin Chemotherapy: Results of a Randomized Trial in Human Epidermal Growth Factor Receptor 2- positive Operable Breast Cancer. J Clin Oncol, 2005, 23(16):3676–85., DOI 10. 1200/JCO.2005.07.032 Gianni L, EiermannW, Semiglazov V, Manikhas A, Lluch A, Tjulandin S, et al. Neoadjuvant Chemotherapy with Trastuzumab Followed by Adjuvant Trastuzumab versus Neoadjuvant Chemotherapy Alone, in Patients with HER2-Positive Locally Advanced Breast Cancer, The NOAH Trial): a Randomised Controlled Superiority Trial with a Parallel HER2-Negative Cohort. Lancet, 2010, 375(9712):377–84., DOI 10.1016/S0140-6736(09, 61964-4 Gianni L, EiermannW, Semiglazov V, Lluch A, Tjulandin S, Zambetti M, et al. Neoadjuvant and Adjuvant Trastuzumab in Patients with HER2-Positive Locally Advanced Breast Cancer, NOAH): Follow-Up of a Randomised Controlled Superiority Trial with a Parallel HER2-Negative Cohort. Lancet Oncol, 2014, 15(6):640–7., DOI 10.1016/S1470-2045(14)70080-4 Gianni L, Pienkowski T, ImY-H, Roman L, Tseng L-M, Liu M-C, et al. Efficacy and Safety of Neoadjuvant Pertuzumab and Trastuzumab in Women with Locally Advanced, Inflammatory, or Early HER2-Positive Breast Cancer, NeoSphere): a Randomised Multicentre, Open-Label, Phase 2 Trial. Lancet Oncol, 2012, 13(1):25–32., DOI 10.1016/S1470-2045(11)70336-9 Gianni L, Pienkowski T, Im Y-H, Tseng L-M, Liu M-C, Lluch A, et al. 5-year Analysis of Neoadjuvant Pertuzumab and Trastuzumab in Patients with Locally Advanced, Inflammatory, or Early-Stage HER2-Positive Breast Cancer, NeoSphere): a Multicentre, Open-Label, Phase 2 Randomised Trial. Lancet Oncol, 2016, 17(6):791–800., DOI 10.1016/S1470-2045(16, 00163-7 Blumenthal GM, Scher NS, Cortazar P, Chattopadhyay S, Tang S, Song P, et al. First FDA Approval of Dual Anti-HER2 Regimen: Pertuzumab in Combination with Trastuzumab and Docetaxel for HER2-Positive Metastatic Breast Cancer. Clin Cancer Res, 2013, 19(18):4911–6., DOI 10. 1158/1078-0432.CCR-13-1212 Horvath Z, Boer K, Dank M, Zs K, Kocsis J, Kover E, et al. [Systemic Therapy of Breast Cancer: Practice Guideline]. Magy Onkol, 2016, 60(3):241–57. Beitsch P, Whitworth P, Baron P, Rotkis MC, Mislowsky AM, Richards PD, et al. Pertuzumab/trastuzumab/CT versus Trastuzumab/CT Therapy for HER2+ Breast Cancer: Results from the Prospective Neoadjuvant Breast Registry Symphony Trial, NBRST). Ann Surg Oncol, 2017, 24:2539–46., DOI 10.1245/s10434-017-5863-x Fasching PA, Hartkopf AD, Gass P, Haberle L, Akpolat-Basci L, Hein A, et al. Efficacy of Neoadjuvant Pertuzumab in Addition to Chemotherapy and Trastuzumab in Routine Clinical Treatment of Patients with Primary Breast Cancer: a Multicentric Analysis. Breast Cancer Res Treat, 2019, 173(2):319–28., DOI 10.1007/s10549-018-5008-3 Samiei S, van Nijnatten TJA, de Munck L, Keymeulen KBMI, Simons JM, Kooreman LFS, et al. Correlation between Pathologic Complete Response in the Breast and Absence of Axillary Lymph NodeMetastases after Neoadjuvant Systemic Therapy. Ann Surg, 2020, 271(3):574–80., DOI 10.1097/SLA. 0000000000003126 Schneeweiss A, Chia S, Hickish T, Harvey V, Eniu A, Hegg R, et al. Pertuzumab Plus Trastuzumab in Combination with Standard Neoadjuvant Anthracycline- Containing and Anthracycline-free Chemotherapy Regimens in Patients with HER2-Positive Early Breast Cancer: a Randomized Phase II Cardiac Safety Study, TRYPHAENA). Ann Oncol, 2013, 24:2278–84., DOI 10.1093/annonc/ mdt182 Hurvitz SA, Martin M, Symmans WF, Jung KH, Huang C-S, Thompson AM, et al. Neoadjuvant Trastuzumab, Pertuzumab, and Chemotherapy versus Trastuzumab Emtansine Plus Pertuzumab in Patients with HER2- Positive Breast Cancer, KRISTINE): a Randomised, Open-Label, Multicentre, Phase 3 Trial. Lancet Oncol, 2018, 19:115–26., DOI 10.1016/ S1470-2045(17)30716-7 Ishii K, Morii N, and Yamashiro H. Pertuzumab in the Treatment of HER2-Positive Breast Cancer: an Evidence-Based Review of its Safety, Efficacy, and Place in Therapy. Core Evid, 2019, 14:51–70., DOI 10.2147/CE. S217848 van Ramshorst MS, van der Voort A, van Werkhoven ED, Mandjes IA, Kemper I, Dezentje VO, et al. Neoadjuvant Chemotherapy with or without Anthracyclines in the Presence of Dual HER2 Blockade for HER2-Positive Breast Cancer, TRAIN-2): a Multicentre, Open-Label, Randomised, Phase 3 Trial. Lancet Oncol, 2018, 19:1630–40., DOI 10.1016/S1470-2045(18, 30570-9 Gonzalez AF, Ambite RA, Valenti EL, Cubero RU, Gala MCA, Guisado AM, et al. Neopersur: Neoadjuvant Pertuzumab in a Real World Data Population in the South of Spain [abstract]. Aacr; Cancer Res, 2020, 80(4 Suppl. l):18–29., DOI 10.1158/1538-7445.SABCS19-P1-18-29 Chang Y-K, Co M, and Kwong A. Conversion Rate from Mastectomy to Breast Conservation after Neoadjuvant Dual Target Therapy for HER2-Positive Breast Cancer in the Asian Population. Breast Cancer, 2020, 27:456–63., DOI 10. 1007/s12282-019-01037-3 Munoz-Montano W, Cabrera-Galeana P, Alvarado-Miranda A, Villarreal- Garza C, Mohar A, Olvera A, et al. Prognostic Value of the Pretreatment Neutrophil-To-Lymphocyte Ratio in Different Phenotypes of Locally Advanced Breast Cancer during Neoadjuvant Systemic Treatment. Clin Breast Cancer, 2020, 20:307–16., DOI 10.1016/j.clbc.2019.12.011 Bae SJ, Yoon C-I, Park SE, Cha CH, Kim D, Lee J, et al. A Neutrophil to Lymphocyte Ratio Is Predictive of Response to Neoadjuvant HER2- Targeted Therapies in the Patients with HER2-Positive Breast Cancer. J. Clin. Oncol, 2019, 37(15_Suppl. l):587., DOI 10.1200/jco.2019.37. 15_suppl.587 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609785 EP 1609793 DI 10.3389/pore.2021.1609785 PG 9 ER PT J AU Park, HM Choi, EJ Kim, JR Bae, KY AF Park, Hui Min Choi, Eun Jung Kim, Jae-Ryong Bae, Kyung Young TI Immunohistochemical Expressions of Senescence-Associated Secretory Phenotype and Its Association With Immune Microenvironments and Clinicopathological Factors in Invasive Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; breast cancer; senescence; prognosis; markers ID immunohistochemistry; breast cancer; senescence; prognosis; markers AB This study was undertaken to investigate immunohistochemical expression of the senescence-associated secretory phenotype (SASP) in invasive breast cancer (IBC) tissues and to determine relationships between SASP positivity and tumor microenvironments and the clinicopathological characteristics of IBC. Immunohistochemistry for senescence markers, that is, high mobility group box-1 (HMGB1), p16, p15, and decoy receptor 2 (DCR2), was performed in tissue microarrays of 1140 IBC samples. Cases positive for at least one of these four markers were considered SASP-positive. Relations between SASP and tumor characteristics, including immune microenvironments (stromal tumor-infiltrating lymphocytes [sTILs] density and numbers of intraepithelial CD103-positive [iCD103 + ] lymphocytes) and clinical outcomes were retrospectively evaluated. HMGB1, p16, p15, or DCR2 was positive in 6.7%, 26.6%, 21.1%, and 26.5%, respectively, of the 1,140 cases. Six hundred and five (53.1%) cases were SASP positive, and SASP positivity was significantly associated with histologic grade 3, high-sTIL and iCD103 + lymphocyte counts, absence of ER or PR, and a high Ki-67 index. Although SASP did not predict breast cancer-specific survival (BCSS) or disease-free survival (DFS) in the entire cohort, SASP positivity in luminal A IBC was associated with poor BCSS and DFS. However, patients with SASP-positive TNBC showed better survival than those with SASP-negative TNBC. In multivariate analysis, SASP positivity was an independent prognostic factor in both luminal A IBC and TNBC, although the effect on prognosis was the opposite. In conclusion, SASP would be involved in the modulation of immune microenvironments and tumor progression in IBC, and its prognostic significance depends on molecular subtype. C1 [Park, Hui Min] Yeungnam University, College of Medicine, Department of PathologyDaegu, South Korea. [Choi, Eun Jung] Yeungnam University College of Medicine, Department of SurgeryDaegu, South Korea. [Kim, Jae-Ryong] Yeungnam University College of Medicine, Department of Biochemistry and Molecular BiologyDaegu, South Korea. [Bae, Kyung Young] Yeungnam University, College of Medicine, Department of PathologyDaegu, South Korea. RP Bae, KY (reprint author), Yeungnam University, College of Medicine, Department of Pathology, Daegu, South Korea. EM ykbae@ynu.ac.kr CR Childs BG, Gluscevic M, Baker DJ, Laberge R-M, Marquess D, Dananberg J, et al. Senescent Cells: an Emerging Target for Diseases of Ageing. Nat Rev Drug Discov, 2017, 16(10):718–35., DOI 10.1038/nrd.2017.116 Basisty N, Kale A, Jeon OH, Kuehnemann C, Payne T, and Rao CA. Proteomic Atlas of Senescence-Associated Secretomes for Aging Biomarker Development. Plos Biol, 2020, 18(1):e3000599., DOI 10.1371/journal.pbio.3000599 Coppe J-P, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, et al. Senescence-associated Secretory Phenotypes Reveal Cell-Nonautonomous Functions of Oncogenic RAS and the P53 Tumor Suppressor. Plos Biol, 2008, 6(12):e301–2868., DOI 10.1371/journal.pbio.0060301 Coppe J-P, Desprez P-Y, Krtolica A, and Campisi J. The Senescence-Associated Secretory Phenotype: the Dark Side of Tumor Suppression. Annu Rev Pathol Mech Dis, 2010, 5:99–118., DOI 10.1146/annurev-pathol-121808-102144 Kang T-W, Yevsa T, Woller N, Hoenicke L, Wuestefeld T, Dauch D, et al. Senescence Surveillance of Pre-malignant Hepatocytes Limits Liver Cancer Development. Nature, 2011, 479(7374):547–51., DOI 10.1038/nature10599 Krizhanovsky V, Xue W, Zender L, Yon M, Hernando E, and Lowe SW. Implications of Cellular Senescence in Tissue Damage Response, Tumor Suppression, and Stem Cell Biology. Cold Spring Harbor Symposia Quantitative Biol, 2008, 73:513–22., DOI 10.1101/sqb.2008.73.048 Di Mitri D, and Alimonti A. Non-Cell-Autonomous Regulation of Cellular Senescence in Cancer. Trends Cel Biol, 2016, 26(3):215–26., DOI 10.1016/ j.tcb.2015.10.005 Biran A, Zada L, Abou Karam P, Vadai E, Roitman L, Ovadya Y, et al. Quantitative Identification of Senescent Cells in Aging and Disease. Aging Cell, 2017, 16(4):661–71., DOI 10.1111/acel.12592 Collado M, and SerranoM. The Power and the Promise of Oncogene-Induced Senescence Markers. Nat Rev Cancer, 2006, 6(6):472–6., DOI 10.1038/nrc1884 Davalos AR, Kawahara M, Malhotra GK, Schaum N, Huang J, Ved U, et al. p53-dependent Release of Alarmin HMGB1 Is a central Mediator of Senescent Phenotypes. J Cel Biol, 2013, 201(4):613–29., DOI 10.1083/jcb.201206006 Wiley CD, Velarde MC, Lecot P, Liu S, Sarnoski EA, Freund A, et al. Mitochondrial Dysfunction Induces Senescence with a Distinct Secretory Phenotype. Cel Metab, 2016, 23(2):303–14., DOI 10.1016/j.cmet.2015.11.011 Zhang Z, Rosen DG, Yao JL, Huang J, and Liu J. Expression of p14ARF, p15INK4b, p16INK4a, and DCR2 Increases during Prostate Cancer Progression. Mod Pathol, 2006, 19(10):1339–43., DOI 10.1038/modpathol.3800655 Nobori T, Miura K, Wu DJ, Lois A, Takabayashi K, and Carson DA. Deletions of the Cyclin-dependent Kinase-4 Inhibitor Gene in Multiple Human Cancers. Nature, 1994, 368(6473):753–6., DOI 10.1038/368753a0 Stone S, Jiang P, Dayananth P, Tavtigian SV, Katcher H, Parry D, et al. Complex Structure and Regulation of the P16, MTS1, Locus. Cancer Res, 1995, 55(14):2988–94. Krishnamurthy J, Ramsey MR, Ligon KL, Torrice C, Koh A, Bonner-Weir S, et al. p16INK4a Induces an Age-dependent Decline in Islet Regenerative Potential. Nature, 2006, 443(7110):453–7., DOI 10.1038/nature05092 Gunakan E, Tohma YA, Karakas¸ LA, Akilli H, Haberal AN, and Ayhan A. Prognostic Impact of P16 and P53 Gene Expressions in Stage 1a Epithelial Ovarian Cancer. Obstet Gynecol Sci, 2020, 63(4):464–9., DOI 10.5468/ogs.19204 Kida K, Terada T, Uwa N, Omori Y, Fujii T, Tomita Y, et al. Relationship between P16 Expression and Prognosis in Patients with Oropharyngeal Cancer Undergoing Surgery. In Vivo, 2018, 32(4):927–35., DOI 10.21873/invivo.11331 Reimers N, Kasper HU, Weissenborn SJ, Stutzer H, Preuss SF, Hoffmann TK, et al. Combined Analysis of HPV-DNA, P16 and EGFR Expression to Predict Prognosis in Oropharyngeal Cancer. Int J Cancer, 2007, 120(8):1731–8., DOI 10.1002/ijc.22355 Malumbres M, Pérez De Castro I, Hernández MI, Jiménez M, Corral T, and Pellicer A. Cellular Response to Oncogenic Ras Involves Induction of the Cdk4 and Cdk6 Inhibitor P15 INK4b. Mol Cel Biol, 2000, 20(8):2915–25., DOI 10.1128/mcb.20.8.2915-2925.2000 Liu X, Yue P, Khuri FR, and Sun S-Y. Decoy Receptor 2, DcR2, Is a P53 Target Gene and Regulates Chemosensitivity. Cancer Res, 2005, 65(20):9169–75., DOI 10.1158/0008-5472.Can-05-0939 Bianchi ME. DAMPs, PAMPs and Alarmins: All We Need to Know about Danger. J Leukoc Biol, 2007, 81(1):1–5., DOI 10.1189/jlb.0306164 Volp K, Brezniceanu ML, Bosser S, Brabletz T, Kirchner T, Gottel D, et al. Increased Expression of High Mobility Group Box 1, HMGB1, Is Associated with an Elevated Level of the Antiapoptotic C-IAP2 Protein in Human colon Carcinomas. Gut, 2006, 55(2):234–42., DOI 10.1136/gut.2004.062729 Yamada S, and Maruyama I. HMGB1, a Novel Inflammatory Cytokine. Clinica Chim Acta, 2007, 375(1-2):36–42., DOI 10.1016/j.cca.2006.07.019 Jeon OH, Kim C, Laberge R-M, Demaria M, Rathod S, Vasserot AP, et al. Local Clearance of Senescent Cells Attenuates the Development of post-traumatic Osteoarthritis and Creates a Pro-regenerative Environment. Nat Med, 2017, 23(6):775–81., DOI 10.1038/nm.4324 Victorelli S, Lagnado A, Halim J, Moore W, Talbot D, Barrett K, et al. Senescent Human Melanocytes Drive Skin Ageing via Paracrine Telomere Dysfunction. Embo j, 2019, 38(23):e101982., DOI 10.15252/embj.2019101982 Brezniceanu M-L, Volp K, Bosser S, Solbach C, Lichter P, Joos S, et al. HMGB1 Inhibits Cell Death in Yeast and Mammalian Cells and Is Abundantly Expressed in Human Breast Carcinoma. FASEB j., 2003, 17(10):1295–7., DOI 10.1096/fj.02-0621fje Choi YR, Kim H, Kang HJ, Kim NG, Kim JJ, Park KS, et al. Overexpression of High Mobility Group Box 1 in Gastrointestinal Stromal Tumors with KIT Mutation. Cancer Res, 2003, 63(9):2188–93. Pare R, Soon PS, Shah A, and Lee CS. Differential Expression of Senescence Tumour Markers and its Implications on Survival Outcomes of Breast Cancer Patients. PLoS One, 2019, 14(4):e0214604., DOI 10.1371/journal.pone.0214604 Kim MC, Park MH, Kang SH, and Bae YK. NDRG3 Protein Expression Is Associated with Aggressive Biologic Phenotype and Unfavorable Outcome in Patients with Invasive Breast Cancer. Int J Clin Exp Pathol, 2019, 12(10):3886–93. Park MH, Kwon SY, Choi JE, Gong G, and Bae YK. Intratumoral CD103- positive Tumour-infiltrating Lymphocytes Are Associated with Favourable Prognosis in Patients with Triple-negative Breast Cancer. Histopathology, 2020, 77:560–9., DOI 10.1111/his.14126 Kwon HJ, Choi JE, Kang SH, Son Y, and Bae YK. Prognostic Significance of CD9 Expression Differs between Tumour Cells and Stromal Immune Cells, and Depends on the Molecular Subtype of the Invasive Breast Carcinoma. Histopathology, 2017, 70(7):1155–65., DOI 10.1111/his.13184 Hammond MEH, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for Immunohistochemical Testing of Estrogen and Progesterone Receptors in Breast Cancer. Jco, 2010, 28(16):2784–95., DOI 10.1200/jco.2009.25.6529 Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, et al. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. Jco, 2018, 36(20):2105–22., DOI 10.1200/jco.2018.77.8738 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, et al. Personalizing the Treatment of Women with Early Breast Cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol, 2013, 24(9): 2206–23., DOI 10.1093/annonc/mdt303 Jang N, Kwon HJ, Park MH, Kang SH, and Bae YK. Prognostic Value of Tumor-Infiltrating Lymphocyte Density Assessed Using a Standardized Method Based on Molecular Subtypes and Adjuvant Chemotherapy in Invasive Breast Cancer. Ann Surg Oncol, 2018, 25(4):937–46., DOI 10.1245/ s10434-017-6332-2 Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, et al. The Evaluation of Tumor-Infiltrating Lymphocytes, TILs, in Breast Cancer: Recommendations by an International TILs Working Group 2014. Ann Oncol, 2015, 26(2):259–71., DOI 10.1093/annonc/mdu450 Wang Z-Q, Milne K, Derocher H, Webb JR, Nelson BH, and Watson PH. CD103 and Intratumoral Immune Response in Breast Cancer. Clin Cancer Res, 2016, 22(24):6290–7., DOI 10.1158/1078-0432.Ccr-16-0732 Webb JR, Milne K, Watson P, Deleeuw RJ, and Nelson BH. Tumorinfiltrating Lymphocytes Expressing the Tissue Resident Memory Marker CD103 Are Associated with Increased Survival in High-Grade Serous Ovarian Cancer. Clin Cancer Res, 2014, 20(2):434–44., DOI 10.1158/1078- 0432.Ccr-13-1877 Liu Z, Wang L, Yang J, Bandyopadhyay A, Kaklamani V, Wang S, et al. Estrogen Receptor Alpha Inhibits Senescence-like Phenotype and Facilitates Transformation Induced by Oncogenic Ras in Human Mammary Epithelial Cells. Oncotarget, 2016, 7(26):39097–107., DOI 10.18632/oncotarget.9772 Kim GJ, Kim DH, Min KW, and Kim SH. Prognostic Impact of High P16/ cyclin D1 index in Breast Cancer. Int J Clin Exp Pathol, 2019, 12(6):2224–32. Kim YH, Choi YW, Lee J, Soh EY, Kim J-H, and Park TJ. Senescent Tumor Cells lead the Collective Invasion in Thyroid Cancer. Nat Commun, 2017, 8: 15208., DOI 10.1038/ncomms15208 Xie T-x., Wei D, Liu M, Gao AC, Ali-Osman F, Sawaya R, et al. Stat3 Activation Regulates the Expression of Matrix Metalloproteinase-2 and Tumor Invasion and Metastasis. Oncogene, 2004, 23(20):3550–60., DOI 10.1038/sj.onc.1207383 Waugh DJJ, and Wilson C. The Interleukin-8 Pathway in Cancer. Clin Cancer Res, 2008, 14(21):6735–41., DOI 10.1158/1078-0432.Ccr-07-4843 Carmeliet P. VEGF as a Key Mediator of Angiogenesis in Cancer. Oncology, 2005, 69(Suppl. 3):4–10., DOI 10.1159/000088478 Fisher DT, Appenheimer MM, and Evans SS. The Two Faces of IL-6 in the Tumor Microenvironment. Semin Immunol, 2014, 26(1):38–47., DOI 10.1016/ j.smim.2014.01.008 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609795 EP 1609804 DI 10.3389/pore.2021.1609795 PG 10 ER PT J AU Soldos, P Besenyi, Zs Hideghety, K Pavics, L Heged}us, Racz, L Kopper, B AF Soldos, Peter Besenyi, Zsuzsanna Hideghety, Katalin Pavics, Laszlo Heged}us, Adam Racz, Levente Kopper, Bence TI Comparison of Shear Wave Elastography and Dynamometer Test in Muscle Tissue Characterization for Potential Medical and Sport Application SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE dynamometry; muscle thickness; muscle stiffness; shear wave elastography; strength parameters ID dynamometry; muscle thickness; muscle stiffness; shear wave elastography; strength parameters AB Skeletal muscle status and its dynamic follow up are of particular importance in the management of several diseases where weight and muscle mass loss and, consequently, immobilization occurs, as in cancer and its treatment, as well as in neurodegenerative disorders. But immobilization is not the direct result of body and muscle mass loss, but rather the loss of the maximal tension capabilities of the skeletal muscle. Therefore, the development of a non-invasive and real-time method which can measure muscle tension capabilities in immobile patients is highly anticipated. Our aim was to introduce and evaluate a special ultrasound measurement technique to estimate a maximal muscle tension characteristic which can be used in medicine and also in sports diagnostics. Therefore, we determined the relationship between the results of shear wave elastography measurements and the dynamometric data of individuals. The measurements were concluded on the m. vastus lateralis. Twelve healthy elite athletes took part in our preliminary proof of principle study—five endurance (S) and seven strength (F) athletes showing unambiguously different muscle composition features, nine healthy subjects (H) without prior sports background, and four cancer patients in treatment for a stage 3 brain tumor (T). Results showed a high correlation between the maximal dynamometric isometric torque (Mmax) and mean elasticity value (E) for the non-athletes [(H + T), (r = 0.795)] and for the athletes [(S + F), (r = 0.79)]. For the athletes (S + F), the rate of tension development at contraction (RTDk) and E correlation was also determined (r = 0.84, p < 0.05). Our measurements showed significantly greater E values for the strength athletes with fast muscle fiber dominance than endurance athletes with slow muscle fiber dominance (p < 0.05). Our findings suggest that shear wave ultrasound elastography is a promising method for estimating maximal muscle tension and, also, the human skeletal muscle fiber ratio. These results warrant further investigations with a larger number of individuals, both in medicine and in sports science. C1 [Soldos, Peter] University of Physical Education, Department of KinesiologyBudapest, Hungary. [Besenyi, Zsuzsanna] University of Szeged, Department of Nuclear MedicineSzeged, Hungary. [Hideghety, Katalin] University of Szeged, Department of OncotherapySzeged, Hungary. [Pavics, Laszlo] University of Szeged, Department of Nuclear MedicineSzeged, Hungary. [Heged}us, Adam] University of Physical Education, Department of KinesiologyBudapest, Hungary. [Racz, Levente] University of Physical Education, Department of KinesiologyBudapest, Hungary. [Kopper, Bence] University of Physical Education, Department of KinesiologyBudapest, Hungary. RP Soldos, P (reprint author), University of Physical Education, Department of Kinesiology, Budapest, Hungary. EM peter.soldos@gmail.com CR Valente KP, Almeida BL, Lazzarini TR, Souza VFd., Ribeiro Td. SC, Guedes de Moraes RA, et al. Association of Adductor Pollicis Muscle Thickness and Handgrip Strength with Nutritional Status in Cancer Patients. PLoS One, 2019, 14:e0220334., DOI 10.1371/journal.pone.0220334 Barata AT, Santos C, Cravo M, Vinhas Md. C, Morais C, Carolino E, et al. Handgrip Dynamometry and Patient-Generated Subjective Global Assessment in Patients with Nonresectable Lung Cancer. Nutr Cancer, 2017, 69:1154–8., DOI 10.1080/01635581.2017.1250923 Bishop DJ, and Girard O. Determinants of Team-Sport Performance: Implications for Altitude Training by Team-Sport Athletes. Br J Sports Med, 2013, 47(1):i17–i21., DOI 10.1136/bjsports-2013-092950 Abbott A, Button C, Pepping GJ, and Collins D. Unnatural Selection: Talent Identification and Development in Sport. Nonlinear Dyn Psychol Life Sci, 2005, 9(1):61–88., DOI 10.4324/9781003049111-4 Pukkala E, Kaprio J, Koskenvuo M, Kujala U, and Sarna S. Cancer Incidence Among Finnish World Class Male Athletes. Int J Sports Med, 2000, 21(3): 216–20., DOI 10.1055/s-2000-8878 Robsahm TE, Hestvik UE, Veierod MB, Fagerlie A, Nystad W, Engebretsen L, et al. Cancer Risk in Norwegian World Class Athletes. Cancer Causes Control, 2010, 21:1711–9., DOI 10.1007/s10552-010-9600-z Wilson A, and Lichtwark G. The Anatomical Arrangement of Muscle and Tendon Enhances Limb Versatility and Locomotor Performance. Phil Trans R Soc B, 2011, 366:15701540–1553., DOI 10.1098/rstb.2010.0361 Bohannon RW. Considerations and Practical Options for Measuring Muscle Strength: A Narrative Review. Biomed Res Int, 2019, 2019:1–10., DOI 10.1155/ 2019/8194537 Knapik JJ, and Ramos MU. Isokinetic and Isometric Torque Relationships in the Human Body. Arch Phys Med Rehabil, 1980, 61(2):64–7. Baltzopoulos V, and Brodie DA. Isokinetic Dynamometry. Sports Med, 1989, 8(2):101–16., DOI 10.2165/00007256-198908020-00003 Thistle HG, Hislop HJ,Moffroid M, and Lowman EW. Isokinetic Contraction: A New Concept of Resistive Exercise. Acta Physiol Scand Suppl, 1967, 48(6): 279–82. Aagaard P, Simonsen EB, Magnusson SP, Larsson B, and Dyhre-Poulsen P. A New Concept for Isokinetic Hamstring: Quadriceps Muscle Strength Ratio. Am J Sports Med, 1998, 26(2):231–7., DOI 10.1177/03635465980260021201 Duarte JP, Valente-Dos-Santos J, Coelho-E-Silva MJ, Couto P, Costa D, Martinho D, et al. Reproducibility of Isokinetic Strength Assessment of Knee Muscle Actions in Adult Athletes: Torques and Antagonist-Agonist Ratios Derived at the Same Angle Position. PLoS One, 2018, 13:e0202261., DOI 10.1371/journal.pone.0202261 Hug F, Tucker K, Gennisson J-L, Tanter M, and Nordez A. Elastography for Muscle Biomechanics. Exerc Sport Sci Rev, 2015, 43(3):125–33., DOI 10.1249/ JES.0000000000000049 Yoshitake Y, Takai Y, Kanehisa H, and Shinohara M. Muscle Shear Modulus Measured with Ultrasound Shear-Wave Elastography across a Wide Range of Contraction Intensity. Muscle Nerve, 2014, 50:103–13., DOI 10.1002/mus.24104 Roberts D, Ageberg E, Andersson G, and Fride T. Clinical Measurements of Proprioception, Muscle Strength and Laxity in Relation to Function in the ACL-Injured Knee. Knee Surg Sports Traumatol Arthrosc, 2007, 15:19–6., DOI 10.1007/s00167-006-0128-4 Taljanovic MS, Gimber LH, Becker GW, Latt LD, Klauser AS, Melville DM, et al. Shear-Wave Elastography: Basic Physics and Musculoskeletal Applications. Radiographics, 2017, 37(3):855–70., DOI 10.1148/rg.2017160116 Creze M, Nordez A, SoubeyrandM, Rocher L, Maˆitre X, and Bellin M-F. Shear Wave Sonoelastography of Skeletal Muscle: Basic Principles, Biomechanical Concepts, Clinical Applications, and Future Perspectives. Skeletal Radiol, 2018, 47(4):457–71., DOI 10.1007/s00256-017-2843-y Brandenburg JE, Eby SF, Song P, Zhao H, Brault JS, Chen S, et al. Ultrasound Elastography: The New Frontier in Direct Measurement of Muscle Stiffness. Arch Phys Med Rehabil, 2014, 95(11):2207–19., DOI 10.1016/ j.apmr.2014.07.007 Andresen JL, Schjerling P, and Saltin B. Muscles, Genes and Athletic Performance. Sci Am, 2000, 283(3):48–55., DOI 10.1038/ scientificamerican0900-48 Methenitis S, Spengos K, Zaras N, Stasinaki A-N, Papadimas G, Karampatsos G, et al. Fiber Type Composition and Rate of Force Development in Endurance- and Resistance-Trained Individuals. J Strength Cond Res, 2019, 33(9):2388–97., DOI 10.1519/JSC.0000000000002150 Methenitis S, Terzis G, Zaras N, Stasinaki A-N, and Karandreas N. Intramuscular Fiber Conduction Velocity, Isometric Force and Explosive Performance. J Hum Kinet, 2016, 51:93–101., DOI 10.1515/hukin-2015-0174 Methenitis S, Karandreas N, Spengos K, Zaras N, Stasinaki A-N, and Terzis G. Muscle Fiber Conduction Velocity, Muscle Fiber Composition, and Power Performance. Med Sci Sports Exerc, 2016, 48(9):1761–71., DOI 10.1249/ MSS.0000000000000954 Sadoyama T, Masuda T, Miyata H, and Katsuta S. Fibre Conduction Velocity and Fibre Composition in Human Vastus Lateralis. Europ J Appl Physiol, 1988, 57(6):767–71., DOI 10.1007/BF01076001 Taljanovic MS, Gimber LH, Becker GW, Latt LD, Klauser AS, Melville DM, et al. Shear-wave Elastography: Basic Physics and Musculoskeletal Applications. Radiographics, 2017, 37(3):855–70., DOI 10.1148/rg.2017160116 Newman AB, Haggerty CL, Goodpaster B, Harris T, Kritchevsky S, Nevitt M, et al. Strength and Muscle Quality in a Well-Functioning Cohort of Older Adults: the Health, Aging and Body Composition Study. J Am Geriatr Soc, 2003, 51(3):323–30., DOI 10.1046/j.1532-5415.2003.51105.x Hasan NAKAK, Kamal HM, and Hussein ZA. Relation between Body Mass index Percentile and Muscle Strength and Endurance. Egypt J Med Hum Genet, 2016, 17(4):367–72., DOI 10.1016/j.ejmhg.2016.01.002 Jackson W, Alexander N, Schipper M, Fig L, Feng F, and Jolly S. Characterization of Changes in Total Body Composition for Patients with Head and Neck Cancer Undergoing Chemoradiotherapy Using Dual-Energy X-ray Absorptiometry. Head Neck, 2013, 36:a1356., DOI 10.1002/hed.23461 Antoun S, Birdsell L, Sawyer MB, Venner P, Escudier B, and Baracos VE. Association of Skeletal Muscle Wasting with Treatment with Sorafenib in Patients with Advanced Renal Cell Carcinoma: Results from a Placebo- Controlled Study. Jco, 2010, 28:61054–1060., DOI 10.1200/JCO.2009.24.9730 Toth MJ, Callahan DM, Miller MS, Tourville TW, Hackett SB, Couch ME, et al. Skeletal Muscle Fiber Size and Fiber Type Distribution in Human Cancer: Effects of Weight Loss and Relationship to Physical Function. Clin Nutr, 2016, 35:61359–1365., DOI 10.1016/j.clnu.2016.02.016 Mankodi A, Logigian E, Callahan L, McClainCWhite R, Henderson D, et al. Myotonic Dystrophy in Transgenic Mice Expressing an Expanded CUG Repeat. Science, 2000, 289:1769–72., DOI 10.1126/science.289.5485.1769 Lukaˇs Z, Kroupova I, Bednaˇrik J, Falk M, Fajkusova L, Sedlaˇckova J, et al. Muscular Biopsy in Myotonic Dystrophy in the Era of Molecular Genetics. Ceska a Slovanska Neurologia a Neurochirurgie, 2007, 70: 103–401.doi:10.5772/30209 Taneja KL. Localization of Trinucleotide Repeat Sequences in Myotonic Dystrophy Cells Using a Single Fluorochrome-Labeled PNA Probe. Biotechniques, 1998, 24(3):472–6., DOI 10.2144/98243rr02 Jiang H, Mankodi A, Swanson MS, Moxley RT, and Thornton CA. Myotonic Dystrophy Type 1 Is Associated with Nuclear Foci of Mutant RNA, Sequestration of Muscleblind Proteins and Deregulated Alternative Splicing in Neurons. Hum Mol Genet, 2004, 13:243079–3088., DOI 10.1093/hmg/ ddh327 Thornton CA, Griggs RC, and Moxley RT. Myotonic Dystrophy with No Trinucleotide Repeat Expansion. Ann Neurol, 1994, 35(3):269–72., DOI 10.1002/ana.410350305 Liquori CL, Ricker K, Moseley ML, Jacobsen JF, Kress W, Naylor SL, et al. Myotonic Dystrophy Type 2 Caused by a CCTG Expansion in Intron 1 of ZNF9. Science, 2001, 293:5531864–7., DOI 10.1126/science.1062125 Day JW, and Ranum LPW. RNA Pathogenesis of the Myotonic Dystrophies. Neuromuscul Disord, 2005, 15:15–6., DOI 10.1016/j.nmd.2004.09.012 Falk M, Vojtiˇskova M, Lukaˇs Z, Kroupova I, and Froster U. Simple Procedure for Automatic Detection of Unstable Alleles in the Myotonic Dystrophy and Huntington’s Disease Loci. Genet Test, 2006, 10:85–97., DOI 10.1089/ gte.2006.10.85 Vihola A, Bassez G, Meola G, Zhang S, Haapasalo H, Paetau A, et al. Histopathological Differences of Myotonic Dystrophy Type 1, DM1, and PROMM/DM2. Neurology, 2003, 60:1854–7., DOI 10.1212/ 01.wnl.0000065898.61358.09 Day JW, Ricker K, Jacobsen JF, Rasmussen LJ, Dick KA, Kress W, et al. Myotonic Dystrophy Type 2. Neurology, 2003, 60:4657–664., DOI 10.1212/ 01.wnl.0000054481.84978.f9 Hayashida I, Tanimoto Y, Takahashi Y, Kusabiraki T, and Tamaki J. Correlation between Muscle Strength and Muscle Mass, and Their Association with Walking Speed, in Community-Dwelling Elderly Japanese Individuals. PLoS One, 2014, 9:e111810., DOI 10.1371/journal.pone.0111810 Mourtzakis M, Parry S, Connolly B, and Puthucheary Z. Skeletal Muscle Ultrasound in Critical Care: a Tool in Need of Translation. Ann ATS, 2017, 14: 1495–503., DOI 10.1513/AnnalsATS.201612-967PS Lacourpaille L, Hug F, Guevel A, Pereon Y, Magot A, Hogrel J-Y, et al. Non-invasive Assessment of Muscle Stiffness in Patients with Duchenne Muscular Dystrophy. Muscle Nerve, 2015, 51(2):284–6., DOI 10.1002/ mus.24445 Carpenter EL, Lau HA, Kolodny EH, and Adler RS. Skeletal Muscle in Healthy Subjects versus Those withGNE-Related Myopathy: Evaluation with Shear- Wave US-A Pilot Study. Radiology, 2015, 277(2):546–54., DOI 10.1148/ radiol.2015142212 Du L-j., He W, Cheng L-g., Li S, Pan Y-s., and Gao J. Ultrasound Shear Wave Elastography in Assessment of Muscle Stiffness in Patients with Parkinson’s Disease: a Primary Observation. Clin Imaging, 2016, 40:61075–1080., DOI 10.1016/j.clinimag.2016.05.008 Flatres A, Aarab Y, Nougaret S, Garnier F, Larcher R, Amalric M, et al. Realtime Shear Wave Ultrasound Elastography: a New Tool for the Evaluation of Diaphragm and Limb Muscle Stiffness in Critically Ill Patients. Crit Care, 2020, 24:1., DOI 10.1186/s13054-020-2745-6 Stout NL, Baima J, Swisher A, Winters-Stone KM, and Welsh J. A Systematic Review of Exercise Systematic Reviews in the Cancer Literature. PM&R.(2005). 9. S347–S384., DOI 10.1016/j.pmrj.2017.07.074 Moore SC, Lee I-M, Weiderpass E, Campbell PT, Sampson JN, Kitahara CM, et al. Association of Leisure-Time Physical Activity with Risk of 26 Types of Cancer in 1.44 Million Adults. JAMA Intern Med, 2016, 176(6):816–25., DOI 10.1001/jamainternmed.2016.1548 Lee J, Lin JB, Wu MH, Jan YT, Chang CL, Huang CY, et al. Muscle Radiodensity Loss during Cancer Therapy Is Predictive for Poor Survival in Advanced Endometrial Cancer. J Cachexia, Sarcopenia Muscle, 2019, 10(4): 814–26., DOI 10.1002/jcsm.12440 Weinberg MS, Shachar SS, Muss HB, Deal AM, Popuri K, Yu H, et al. Beyond Sarcopenia: Characterization and Integration of Skeletal Muscle Quantity and Radiodensity in a Curable Breast Cancer Population. Breast J, 2018, 24(3): 278–84., DOI 10.1111/tbj.12952 Ruiz-Casado A, Martin-Ruiz A, Perez LM, Provencio M, Fiuza-Luces C, and Lucia A. Exercise and the Hallmarks of Cancer. Trends Cancer, 2017, 3: 423–41., DOI 10.1016/j.trecan.2017.04.007 Piraux E, Reychler G, de Noordhout LM, Forget P, Deswysen Y, and Caty G. What Are the Impact and the Optimal Design of a Physical Prehabilitation Program in Patients with Esophagogastric Cancer Awaiting Surgery? A Systematic Review. BMC Sports Sci Med Rehabil, 2021, 13:1., DOI 10.1186/ s13102-021-00260-w Campbell KL, Winters-Stone KM, Wiskemann J, May AM, Schwartz KS, et al. Exercise Guidelines for Cancer Survivors: Consensus Statement from International Multidisciplinary Roundtable. Med Sci Sports Exerc, 2019, 51: 112375–2390., DOI 10.1249/MSS.0000000000002116 Ungar N, Tsiouris A, Haussmann A,Herbolsheimer F, Wiskemann J, Steindorf K, et al. To Rest or Not to Rest-Health Care Professionals’ Attitude toward Recommending Physical Activity to Their Cancer Patients. Psycho-Oncology, 2019, 28:4784–791., DOI 10.1002/pon.5020 Rock CL, Doyle C, Demark-Wahnefried W, Meyerhardt J, Courneya KS, Schwartz AL, et al. Nutrition and Physical Activity Guidelines for Cancer Survivors. CA: A Cancer J Clinicians, 2012, 62(4):242–74., DOI 10.3322/ caac.21142 Nakano J, Hashizume K, Fukushima T, Ueno K, Matsuura E, IKio Y, et al. Effects of Aerobic and Resistance Exercises on Physical Symptoms in Cancer Patients: A Meta-Analysis. Integr Cancer Ther, 2018, 17:41048–1058., DOI 10.1177/1534735418807555 Lobelo F, Rohm Young D, Sallis R, Garber MD, Billinger SA, Duperly J, et al. Routine Assessment and Promotion of Physical Activity in Healthcare Settings: A Scientific Statement from the American Heart Association. Circulation, 2018, 137:18:e495–e522., DOI 10.1161/CIR.0000000000000559 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609798 EP 1609808 DI 10.3389/pore.2021.1609798 PG 11 ER PT J AU Sebestyen, A Kopper, L Danko, T Timar, J AF Sebestyen, Anna Kopper, Laszlo Danko, Titanilla Timar, Jozsef TI Hypoxia Signaling in Cancer: From Basics to Clinical Practice SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE cancer; hypoxia; angiogenesis; metabolism; therapy ID cancer; hypoxia; angiogenesis; metabolism; therapy AB Cancer hypoxia, recognized as one of the most important hallmarks of cancer, affects gene expression, metabolism and ultimately tumor biology-related processes. Major causes of cancer hypoxia are deficient or inappropriate vascularization and systemic hypoxia of the patient (frequently induced by anemia), leading to a unique form of genetic reprogramming by hypoxia induced transcription factors (HIF). However, constitutive activation of oncogene-driven signaling pathways may also activate hypoxia signaling independently of oxygen supply. The consequences of HIF activation in tumors are the angiogenic phenotype, a novel metabolic profile and the immunosuppressive microenvironment. Cancer hypoxia and the induced adaptation mechanisms are two of the major causes of therapy resistance. Accordingly, it seems inevitable to combine various therapeutic modalities of cancer patients by existing anti-hypoxic agents such as anti-angiogenics, anti-anemia therapies or specific signaling pathway inhibitors. It is evident that there is an unmet need in cancer patients to develop targeted therapies of hypoxia to improve efficacies of various anti-cancer therapeutic modalities. The case has been opened recently due to the approval of the first-in-class HIF2α inhibitor. C1 [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Danko, Titanilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM jtimar@gmail.com CR Muz B, de la Puente P, Azab F, and Azab AK. The Role of Hypoxia in Cancer Progression, Angiogenesis, Metastasis, and Resistance to Therapy. Hp, 2015, 3:83–92., DOI 10.2147/HP.S93413 Folkman J. Tumor Angiogenesis: Therapeutic Implications. N Engl J Med, 1971, 285:1182–6., DOI 10.1056/NEJM19711118285210810.1056/ nejm197108122850711 Heldin C-H, Rubin K, Pietras K, and Ostman A. High Interstitial Fluid Pressure - an Obstacle in Cancer Therapy. Nat Rev Cancer, 2004, 4:806–13., DOI 10.1038/nrc1456 Siemann DW. The Unique Characteristics of Tumor Vasculature and Preclinical Evidence for its Selective Disruption by Tumor-Vascular Disrupting Agents. Cancer Treat Rev, 2011, 37:63–74., DOI 10.1016/j.ctrv. 2010.05.001 Lee P, Chandel NS, and Simon MC. Cellular Adaptation to Hypoxia through Hypoxia Inducible Factors and beyond. Nat Rev Mol Cel Biol, 2020, 21: 268–83., DOI 10.1038/s41580-020-0227-y Semenza GL. HIF-1 and Tumor Progression: Pathophysiology and Therapeutics. Trends Mol Med, 2002, 8:s62–s67., DOI 10.1016/s1471- 4914(02)02317-1 Pugh CW, and Ratcliffe PJ. New Horizons in Hypoxia Signaling Pathways. Exp Cel Res, 2017, 356:116–21., DOI 10.1016/j.yexcr.2017.03.008 Ortiz-Barahona A, Villar D, Pescador N, Amigo J, and del Peso L. Genomewide Identification of Hypoxia-Inducible Factor Binding Sites and Target Genes by a Probabilistic Model Integrating Transcription-Profiling Data and In Silico Binding Site Prediction. Nucl Acid Res, 2010, 38:2332–45., DOI 10. 1093/nar/gkp1205 SEMENZA., 2021), Available at: www. gsea-msigdb.org/gsea/msigdb/cards/ SEMENZA_HIF1A_TARGETS, Accessed 09. 03, 2021). Kaplan AR, and Glazer PM. Impact of Hypoxia on DNA Repair and Genome Integrity. Mutagenesis, 2020, 35:61–8., DOI 10.1093/mutage/gez019 Morris MR, Hughes DJ, Tian YM, Ricketts CJ, Gentle D, Serrano-Fernandez P, et al. Mutation Analysis of Hypoxia-Inducible Factors HIF1α and HIF2α in Renal Carcinoma. Anticancer Res, 2009, 29:4337–43. Pang Y, Gupta G, Jha A, Yue X, Wang H, Huynh TT, et al. Nonmosaic Somatic HIF2A Mutations Associated with Late Onset Polycythemiaparaganglioma Syndrome: Newly Recognized Subclass of Polycythemiaparaganglioma Syndrome. Cancer, 2019, 125:1258–66., DOI 10.1002/cncr. 31839 Glasker S, Vergauwen E, Koch CA, Kutikov A, and Vortmeyer AO. Von Hippel-Lindau Disease: Current Challenges and Future Prospects. Ott, 2020, Vol. 13:5669–90., DOI 10.2147/OTT.S190753 Dizman N, Philip EJ, and Pal SK. Genomic Profiling in Renal Cell Carcinoma. Nat Rev Nephrol, 2020, 16:435–51., DOI 10.1038/s41581-020- 0301-x Macklin PS, Yamamoto A, Browning L, Hofer M, Adam J, and Pugh CW. Recent Advances in the Biology of Tumour Hypoxia with Relevance to Diagnostic Practice and Tissue-based Research. J Pathol, 2020, 250:593–611., DOI 10.1002/path.5402 Szendr}oi A, Szasz AM, Kardos M, T}okes AM, Idan R, Sz}ucs M, et al. Opposite Prognostic Roles of HIF1α and HIF2α Expressions in Bone Metastatic Clear Cell Renal Cell Cancer. Oncotarget, 2016, 7:42086–98., DOI 10.18632/ oncotarget.9669 Aoki M, and Fujishita T. Oncogenic Roles of the PI3K/AKT/mTOR axis. Curr Top Microbiol Immunol, 2017, 407:153–89., DOI 10.1007/ 82_2017_6 Semenza GL. The Hypoxic Tumor Microenvironment: A Driving Force for Breast Cancer Progression. Biochim Biophys Acta, Bba, - Mol Cel Res, 2016, 1863:382–91., DOI 10.1016/j.bbamcr.2015.05.036 Tatrai E, Bartal A, Gacs A, Paku S, Kenessey I, Garay T, et al. Cell Typedependent HIF1 α-mediated Effects of Hypoxia on Proliferation, Migration and Metastatic Potential of Human Tumor Cells. Oncotarget, 2017, 8: 44498–510., DOI 10.18632/oncotarget.17806 Dome B, Hendrix MJC, Paku S, Tovari J, and Timar J. Alternative Vascularization Mechanisms in Cancer. Am J Pathol, 2007, 170:1–15., DOI 10.2353/ajpath.2007.060302 Dome B, Timar J, Dobos J, Meszaros L, Raso E, Paku S, et al. Identification and Clinical Significance of Circulating Endothelial Progenitor Cells in Human Non-small Cell Lung Cancer. Cancer Res, 2006, 66:7341–7., DOI 10.1158/0008-5472.CAN-05-4654 Dome B, Paku S, Somlai B, and Timar J. Vascularization of Cutaneous Melanoma Involves Vessel Co-option and Has Clinical Significance. J Pathol, 2002, 197:355–62., DOI 10.1002/path.1124 Bridgeman VL, Vermeulen PB, Foo S, Bilecz A, Daley F, Kostaras E, et al. Vessel Co-option Is Common in Human Lung Metastases and Mediates Resistance to Anti-angiogenic Therapy in Preclinical Lung Metastasis Models. J Pathol, 2017, 241:362–74., DOI 10.1002/path.4845 Dezs}o K, Bugyik E, Papp V, Laszlo V, Dome B, Tovari J, et al. Development of Arterial Blood Supply in Experimental Liver Metastases. Am J Pathol, 2009, 175:835–43., DOI 10.2353/ajpath.2009.090095 Szabo V, Bugyik E, Dezso K, Ecker N, Nagy P, Timar J, et al. Mechanism of Tumour Vascularization in Experimental Lung Metastases. J Pathol, 2015, 235:384–96., DOI 10.1002/path.4464 Dome B, Timar J, and Paku S. A Novel Concept of Glomeruloid Body Formation in Experimental Cerebral Metastases. J Neuropathol Exp Neurol, 2003, 62:655–61., DOI 10.1093/jnen/62.6.655 Inan S, and Hayran M. Cell Signaling Pathways Related to Epithelial Mesenchymal Transition in Cancer Metastasis. Crit Rev Oncog, 2019, 24: 47–54., DOI 10.1615/CritRevOncog.2018029509 Timar J, Tovari J, Raso E, Meszaros L, Bereczky B, and Lapis K. Plateletmimicry of Cancer Cells: Epiphenomenon with Clinical Significance. Oncology, 2005, 69:185–201., DOI 10.1159/000088069 Manoitis AJ, Folberg R, Hess A, Seftor EA, Gardner LMG, Pe’er J, et al. Vascular Channel Formation by Human Melanoma Cells: Role in Vasculogenic Mimicry. Am J Pathol, 1999, 155:739–52., DOI 10.1016/ S0002-9440(10)65173-5 Timar J, and Toth J. Tumor Sinuses - Vascular Channels. Pathol Oncol Res, 2000, 6:83–6., DOI 10.1007/BF03032354 Hendrix MJC, Seftor EA, Hess AR, and Seftor REB. Vasculogenic Mimicry and Tumour-Cell Plasticity: Lessons from Melanoma. Nat Rev Cancer, 2003, 3:411–21., DOI 10.1038/nrc1092 L}orincz T, Timar J, and Szendr}oi M. Alterations of Microvascular Density in Bone Metastases of Adenocarcinomas. Pathol Oncol Res, 2004, 10:149–53., DOI 10.1007/BF03033743 Gilreath JA, Stenehjem DD, and Rodgers GM. Diagnosis and Treatment of Cancer-Related Anemia. Am J Hematol, 2014, 89:203–12., DOI 10.1002/ajh. 23628 Parks SK, Cormerais Y, and Pouyssegur J. Hypoxia and Cellular Metabolism in Tumour Pathophysiology. J Physiol, 2017, 595:2439–50., DOI 10.1113/ JP273309 Robey IF, Lien AD, Welsh SJ, Baggett BK, and Gillies RJ. Hypoxia-Inducible Factor-1α and the Glycolytic Phenotype in Tumors. Neoplasia, 2005, 7: 324–30., DOI 10.1593/neo.04430 Faubert B, Solmonson A, and DeBerardinis RJ. Metabolic Reprogramming and Cancer Progression. Science, 2020, 368:eaaw5473., DOI 10.1126/science. aaw5473 Yoshida GJ. Metabolic Reprogramming: the Emerging Concept and Associated Therapeutic Strategies. J Exp Clin Cancer Res, 2015, 34:11., DOI 10.1186/s13046-015-0221-y Danhier P, Ba´nski P, Payen VL, Grasso D, Ippolito L, Sonveaux P, et al. Cancer Metabolism in Space and Time: Beyond the Warburg Effect. Biochim Biophys Acta, Bba, - Bioenerg, 2017, 1858:556–72., DOI 10.1016/j.bbabio. 2017.02.001 Moldogazieva NT, Mokhosoev IM, and Terentiev AA. Metabolic Heterogeneity of Cancer Cells: An Interplay between HIF-1, GLUTs, and AMPK. Cancers, 2020, 12:862., DOI 10.3390/cancers12040862 Sarkadi B, Meszaros K, Krencz I, Canu L, Krokker L, Zakarias S, et al. Glutaminases as a Novel Target for SDHB-Associated Pheochromocytomas/ Paragangliomas. Cancers, 2020, 12:599., DOI 10.3390/cancers12030599 Payen VL, Zampieri LX, Porporato PE, and Sonveaux P. Pro- and Antitumor Effects of Mitochondrial Reactive Oxygen Species. Cancer Metastasis Rev, 2019, 38:189–203., DOI 10.1007/s10555-019-09789-2 Liu GY, and Sabatini DM. mTOR at the Nexus of Nutrition, Growth, Ageing and Disease. Nat Rev Mol Cel Biol, 2020, 21(4):183–203., DOI 10.1038/s41580- 019-0199-y He L, Gomes AP, Wang X, Yoon SO, Lee G, Nagiec MJ, et al. mTORC1 Promotes Metabolic Reprogramming by the Suppression of GSK3-dependent Foxk1 Phosphorylation. Mol Cel, 2018, 70(5):949–60., DOI 10.1016/j.molcel. 2018.04.024 Li W, Petrimpol M, Molle KD, Hall MN, Battegay EJ, and Humar R. Hypoxiainduced Endothelial Proliferation Requires Both mTORC1 and mTORC2. Circ Res, 2007, 100(1):79–87., DOI 10.1161/01.RES.0000253094.03023.3f Daskalaki I, Gkikas I, and Tavernarakis N. Hypoxia and Selective Autophagy in Cancer Development and Therapy. Front Cel Dev. Biol., 2018, 6:104., DOI 10.3389/fcell.2018.00104 Mutvei AP, Nagiec MJ, Hamann JC, Kim SG, Vincent CT, and Blenis J. Rap1- GTPases Control mTORC1 Activity by Coordinating Lysosome Organization with Amino Acid Availability. Nat Commun, 2020, 11(1): 1416., DOI 10.1038/s41467-020-15156-5 Parks SK, and Pouyssegur J. Targeting pH Regulating Proteins for Cancer Therapy-Progress and Limitations. Semin Cancer Biol, 2017, 43:66–73., DOI 10.1016/j.semcancer.2017.01.007 Li S, Topatana W, Juengpanich S, Cao J, Hu J, Zhang B, et al. Development of Synthetic Lethality in Cancer: Molecular and Cellular Classification. Sig Transduct Target Ther, 2020, 5(1):241., DOI 10.1038/s41392-020-00358-6 Diaz-Bulnes P, Saiz ML, Lopez-Larrea C, and Rodriguez RM. Crosstalk between Hypoxia and ER Stress Response: a Key Regulator of Macrophage Polarization. Front Immunol, 2020, 10:2951., DOI 10.3389/ fimmu.2019.02951 Vetsika E-K, Koukos A, and Kotsakis A. Myeloid-Derived Suppressor Cells: Major Figures that Shape the Immunosuppressive and Angiogenic Network in Cancer. Cells, 2019, 8:1647., DOI 10.3390/cells8121647 Taylor CT, and Colgan SP. Regulation of Immunity and Inflammation by Hypoxia in Immunological Niches. Nat Rev Immunol, 2017, 17:774–85., DOI 10.1038/nri.2017.103 Kaur S, Chang T, Singh SP, Lim L, Mannan P, Garfield SH, et al. CD47 Signaling Regulates the Immunosuppressive Activity of VEGF in T Cells. J.I., 2014, 193:3914–24., DOI 10.4049/jimmunol.1303116 Petrova V, Annicchiarico-Petruzzelli M, Melino G, and Amelio I. The Hypoxic Tumour Microenvironment. Oncogenesis, 2018, 7:10., DOI 10. 1038/s41389-017-0011-9 Reiniger L, Teglasi V, Pipek O, Rojko L, Glasz T, Vagvolgyi A, et al. Tumor Necrosis Correlates with PD-L1 and PD-1 Expression in Lung Adenocarcinoma. Acta Oncologica, 2019, 58:1087–94., DOI 10.1080/ 0284186X.2019.1598575 Kiss J, Timar J, Somlai B, Gilde K, Fej}os Z, Gaudi I, et al. Association of Microvessel Density with Infiltrating Cells in Human Cutaneous Malignant Melanoma. Pathol Oncol Res, 2007, 13:21–31., DOI 10.1007/BF02893437 Serganova I, Humm J, Ling C, and Blasberg R. Tumor Hypoxia Imaging: Fig. 1. Clin Cancer Res, 2006, 12:5260–4., DOI 10.1158/1078-0432.CCR-06- 0517 Krohn KA, Link JM, and Mason RP. Molecular Imaging of Hypoxia. J Nucl Med, 2008, 49:129S–148S., DOI 10.2967/jnumed.107.045914 Scotto KW. Transcriptional Regulation of ABC Drug Transporters. Oncogene, 2003, 22:7496–511., DOI 10.1038/sj.onc.1206950 Jin X, Gong L, Peng Y, Li L, and Liu G. Enhancer-bound Nrf2 Licenses HIF- 1α Transcription under Hypoxia to Promote Cisplatin Resistance in Hepatocellular Carcinoma Cells. Aging, 2020, 13:364–75., DOI 10.18632/ aging.202137 Belisario DC, Kopecka J, Pasino M, Akman M, De Smaele E, Donadelli M, et al. Hypoxia Dictates Metabolic Rewiring of Tumors: Implications for Chemoresistance. Cells, 2020, 9:2598., DOI 10.3390/cells9122598 Cosse J-P, and Michiels C. Tumour Hypoxia Affects the Responsiveness of Cancer Cells to Chemotherapy and Promotes Cancer Progression. Acamc, 2008, 8:790–7., DOI 10.2174/187152008785914798 Koshinani HM, Afshar S, and Najafi R. Hypoxia: a Double-Edged Sword in Cancer Therapy. Cancer Invest, 2016, 34:536–45., DOI 10.1080/07357907. 2016.1245317 Aebersold DM, Burri P, Beer KT, Laissue J, Djonov V, Greiner RH, et al. Expression of Hypoxia Inducible Factor-Alpha: a Novel Predictive, Prognostic Parameter in the Radiotherapy of Oropharyngeal Cancer. Cancer Res, 2001, 61:2911–6. Lovey J, Lukits J, Remenar E, Koronczay K, Kasler M, Nemeth G, et al. Antiangiogenic Effects of Radiotherapy but Not Initial Microvessel Density Predict Survival in Inoperable Oropharyngeal Squamous Cell Carcinoma. Strahlenther Onkol, 2006, 182:149–56., DOI 10.1007/s00066-006-1447-6 Jaszai J, and Schmidt M. Trends and Challenges in Tumor Anti-angiogenic Therapies. Cells, 2019, 8:1102., DOI 10.3390/cells8091102 Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R, et al. Bevacizumab, Avastin, in Cancer Treatment: A Review of 15 Years of Clinical Experience and Future Outlook. Cancer Treat Rev., 2020). 86. p. 102017., DOI 10.1016/j.ctrv.2020.102017 Martin JD, Seano G, and Jain RK. Normalizing Function of Tumor Vessels: Progress, Opportunities, and Challenges. Annu Rev Physiol, 2019, 81:505–34., DOI 10.1146/annurev-physiol-020518-114700 Chen Z, Wei J, Ma X, and Yu J. Efficacy of EGFR-TKIs with or without Angiogenesis Inhibitors in Advanced Non-small-cell Lung Cancer: A Systematic Review and Meta-Analysis. J Cancer, 2020, 11:686–95., DOI 10. 7150/jca.34957 Sun D-c., Shi Y, Wang Y-r., Lv Y, Yan H, Mao H, et al. KRAS Mutation and Primary Tumor Location Do Not Affect Efficacy of Bevacizumab-Containing Chemotherapy in Stagae IV Colorectal Cancer Patients. Sci Rep, 2017, 7:14368., DOI 10.1038/s41598-017- 14669-2 Ghimessy AK, Gellert A, Schlegl E, Hegedus B, Raso E, Barbai T, et al. KRAS Mutations Predict Response and Outcome in Advanced Lung Adenocarcinoma Patients Receiving First-Line Bevacizumab and Platinum-Based Chemotherapy. Cancers, 2019, 11:1514., DOI 10.3390/ cancers11101514 Haibe Y, Kreidieh M, El Hajj H, Khalifeh I, Mukherji D, Temraz S, et al. Resistance Mechanisms to Anti-angiogenic Therapies in Cancer. Front Oncol, 2020, 10:221., DOI 10.3389/fonc.2020.00221 Uribesalgo I, Hoffmann D, Zhang Y, Kavirayani A, Lazovic J, Berta J, et al. Apelin Inhibition Prevents Resistance and Metastasis Associated with Antiangiogenic Therapy. EMBO Mol Med, 2019, 11:e9266., DOI 10.15252/emmm. 201809266 Torok S, Rezeli M, Kelemen O, Vegvari A, Watanabe K, Sugihara Y, et al. Limited Tumor Tissue Drug Penetration Contributes to Primary Resistance against Angiogenesis Inhibitors. Theranostics, 2017, 7:400–12., DOI 10.7150/ thno.16767 Fukumura D, Kloepper J, Amoozgar Z, Duda DG, and Jain RK. Enhancing Cancer Immunotherapy Using Antiangiogenics: Opportunities and Challenges. Nat Rev Clin Oncol, 2018, 15:325–40., DOI 10.1038/nrclinonc. 2018.29 Rassy E, Flippot R, and Albiges L. Tyrosine Kinase Inhibitors and Immunotherapy Combinations in Renal Cell Carcinoma. Ther Adv Med Oncol, 2020, 12:175883592090750., DOI 10.1177/1758835920907504 Gubens MA, and Davies M. NCCN Guidelines Updates: New Immunotherapy Strategies for Improving Outcomes in Non-small Cell Lung Cancer. J Natl Compr Canc Netw, 2019, 17:574–8., DOI 10.6004/ jnccn.2019.5005 Hilmi M, Neuzillet C, Calderaro J, Lafdil F, Pawlotsky J-M, and Rousseau B. Angiogenesis and Immune Checkpoint Inhibitors as Therapies for Hepatocellular Carcinoma: Current Knowledge and Future Research Directions. J Immunotherapy Cancer, 2019, 7:333., DOI 10.1186/s40425- 019-0824-5 Gaspar BL, Sharma P, and Das R. Anemia in Malignancies: Pathogenetic and Diagnostic Considerations. Hematology, 2015, 20:18–25., DOI 10.1179/ 1607845414Y.0000000161 Bohlius J, Bohlke K, Castelli R, Djulbegovic B, Lustberg MB, MartinoM, et al. Management of Cancer-Associated Anemia with Erythropoiesis-Stimulating Agents: ASCO/ASH Clinical Practice Guideline Update. Jco, 2019, 37: 1336–51., DOI 10.1200/JCO.18.02142 Tovari J, Gilly R, Raso E, Paku S, Bereczky B, Varga N, et al. Recombinant Human Erythropoietin α Targets Intratumoral Blood Vessels, Improving Chemotherapy in Human Xenograft Models. Cancer Res, 2005, 65:7186–93., DOI 10.1158/0008-5472.CAN-04-2498 Lovey J, Bereczky B, Gilly R, Kenessey I, Raso E, Simon E, et al. Recombinant Human Erythropoietin Alpha Improves the Efficacy of Radiotherapy of a Human Tumor Xenograft, Affecting Tumor Cells and Microvessels. Strahlenther Onkol, 2008, 184:1–7., DOI 10.1007/s00066- 008-1745-2 Zou Z, Tao T, Li H, and Zhu X. mTOR Signaling Pathway and mTOR Inhibitors in Cancer: Progress and Challenges. Cell Biosci, 2020, 10:31., DOI 10.1186/s13578-020-00396-1 Karakhasev S, and Reginato MJ. Progress toward Overcoming Hypoxia- Induced Resistance to Solid Tumor Therapy. Cancer Management Res, 2015, 7:253–64., DOI 10.2147/CMAR.S58285 Burian Z, Ladanyi A, Barbai T, Piurko V, Garay T, Raso E, et al. Selective Inhibition of HIF1α Expression by ZnSO4 Has Antitumoral Effects in Human Melanoma. Pathol Oncol Res, 2020, 26:673–9., DOI 10.1007/ s12253-018-00573-1 Mabjeesh NJ, Escuin D, LaVallee TM, Pribluda VS, Swartz GM, Johnson MS, et al. 2ME2 Inhibits Tumor Growth and Angiogenesis by Disrupting Microtubules and Dysregulating HIF. Cancer Cell, 2003, 3:363–75., DOI 10. 1016/s1535-6108(03)00077-1 Modi S, Stopeck A, Linden H, Solit D, Chandarlapaty S, Rosen N, et al. HSP90 Inhibition Is Effective in Breast Cancer: A Phase II Trial of Tanespimycin, 17- AAG, Plus Trastuzumab in Patients with HER2-Positive Metastatic Breast Cancer Progressing on Trastuzumab. Clin Cancer Res, 2011, 17:5132–9., DOI 10.1158/1078-0432.CCR-11-0072 Kong D, Park EJ, Stephen AG, Calvani M, Cardellina JH, Monks A, et al. Echinomycin, a Small-Molecule Inhibitor of Hypoxia-Inducible Factor-1 DNA-Binding Activity. Cancer Res, 2005, 65:9047–55., DOI 10.1158/0008- 5472.CAN-05-1235 Yeo E-J, Chun Y-S, Cho Y-S, Kim J, Lee J-C, Kim M-S, et al. YC-1: A Potential Anticancer Drug Targeting Hypoxia-Inducible Factor 1. JNCI J Natl Cancer Inst, 2003, 95:516–25., DOI 10.1093/jnci/95.7.516 Courtney KD, Infante JR, Lam ET, Figlin RA, Rini BI, Brugarolas J, et al. Phase I Dose-Escalation Trial of PT2385, a First-In-Class Hypoxia-Inducible Factor-2α Antagonist in Patients with Previously Treated Advanced Clear Cell Renal Cell Carcinoma Antagonist in Patients with Previously Treated Advanced Clear Cell Renal Cell Carcinoma. Jco, 2018, 36:867–74., DOI 10. 1200/JCO.2017.74.2627 Jonasch E, Donskov F, Iliopoulos O, Rathmell WK, Narayan V, Maughan BL, et al. Phase II Study of the Oral HIF-2α Inhibitor MK-6482 for Von Hippel- Lindau Disease-Associated Renal Cell Carcinoma. Jco, 2020, 38:5003., DOI 10. 1200/JCO.2020.38.15_suppl.5003 Overgaard J. Hypoxic Modification of Radiotherapy in Squamous Cell Carcinoma of the Head and Neck - A Systematic Review and Meta- Analysis. Radiother Oncol, 2011, 100:22–32., DOI 10.1016/j.radonc.2011. 03.004 Janssen GO, Rademakers SE, Terhaard CH, Doornaert PA, Bijl HP, van der Ende P, et al. Accelerated Radiotherapy with Carbogen and Nicotinamide for Laryngeal Cancer: Results of a Phase III Randomized Trial. J Clin Oncol, 2012, 30:1777–83., DOI 10.1200/JCO.2011.35.9315 Thompson D, Yang H, Baines H, Miles E, Bolton S, West C, et al. NIMRAD - a Phase III Trial to Investigate the Use of Nimorazole Hypoxia Modification with Intensity-Modulated Radiotherapy in Head and Neck Cancer. Clin Oncol, R Coll Radiol, 2014, 26:344–7., DOI 10.1016/j.clon.2014.03.003 Barker HE, Paget JTE, Khan AA, and Harrington KJ. The Tumour Microenvironment after Radiotherapy: Mechanisms of Resistance and Recurrence. Nat Rev Cancer, 2015, 15:409–25., DOI 10.1038/nrc3958 Jimenez-Valerio G, and Casanovas O. Angiogenesis and Metabolism: Entwined for Therapy Resistance. Trends Cancer, 2017, 3:10–8., DOI 10. 1016/j.trecan.2016.11.007 Peiris-Pages M, Martinez-Outschoorn UE, Pestell RG, Sotgia F, and Lisanti MP. Cancer Stem Cell Metabolism. Breast Cancer Res, 2016, 18:55., DOI 10. 1186/s13058-016-0712-6 Liang L, Hui K, Hu C, Wen Y, Yang S, Zhu P, et al. Autophagy Inhibition Potentiates the Anti-angiogenic Property of Multikinase Inhibitor Anlotinib through JAK2/STAT3/VEGFA Signaling in Non-small Cell Lung Cancer Cells. J Exp Clin Cancer Res, 2019, 38:71., DOI 10.1186/s13046-019-1093-3 AllenMieville EP, Mieville P, Warren CM, Saghafinia S, Li L, PengM-W, et al. Metabolic Symbiosis Enables Adaptive Resistance to Anti-angiogenic Therapy that Is Dependent on mTOR Signaling. Cel Rep, 2016, 15: 1144–60., DOI 10.1016/j.celrep.2016.04.029 Vallet S, Pahernik S, Hofner T, Tosev G, Hadaschik B, Duensing S, et al. Efficacy of Targeted Treatment beyond Third-Line Therapy in Metastatic Kidney Cancer: Retrospective Analysis from a Large-Volume Cancer Center. Clin Genitourinary Cancer, 2015, 13(3):e145–e152., DOI 10.1016/j.clgc.2014. 12.012 Wick W, Platten M, Wick A, Hertenstein A, Radbruch A, Bendszus M, et al. Current Status and Future Directions of Anti-angiogenic Therapy for Gliomas. Neuro Oncol, 2016, 18(3):315–28., DOI 10.1093/neuonc/nov180 Galdy S, Cella CA, Spada F, Murgioni S, Frezza AM, Ravenda SP, et al. Systemic Therapy beyond First-Line in Advanced Gastric Cancer: An Overview of the Main Randomized Clinical Trials. Crit Rev Oncology/ Hematology, 2016, 99:1–12., DOI 10.1016/j.critrevonc.2015.09.004 Grozinsky-Glasberg S, Lines KE, Avniel-Polak S, Bountra C, and Thakker RV. Preclinical Drug Studies in MEN1-Related Neuroendocrine Neoplasms, MEN1-NENs). Endocr Relat Cancer, 2020, 27(9):R345–R355., DOI 10. 1530/ERC-20-0127 Yoshida GJ. The Harmonious Interplay of Amino Acid and Monocarboxylate Transporters Induces the Robustness of Cancer Cells. Metabolites, 2021, 11(1):27., DOI 10.3390/metabo11010027 BenejM, Svastova E, Banova R, Kopacek J, Gibadulinova A, Kery M, et al. CA IX Stabilizes Intracellular pH to Maintain Metabolic Reprogramming and Proliferation in Hypoxia. Front Oncol, 2020, 10:1462., DOI 10.3389/fonc.2020. 01462 Marrone KA, Zhou X, Forde PM, Purtell M, Brahmer JR, Hann CL, et al. A Randomized Phase II Study of Metformin Plus Paclitaxel/Carboplatin/ Bevacizumab in Patients with Chemotherapy-Naive Advanced or Metastatic Nonsquamous Non-Small Cell Lung Cancer. Oncol, 2018, 23: 859–65., DOI 10.1634/theoncologist.2017-0465 Markowska A, Sajdak S, Markowska J, and Huczy´nski A. Angiogenesis and Cancer Stem Cells: New Perspectives on Therapy of Ovarian Cancer. Eur J Med Chem, 2017, 142:87–94., DOI 10.1016/j.ejmech.2017.06.030 Pet}ovari G, Danko T, Krencz I, Hujber Z, Rajnai H, Vetlenyi E, et al. Inhibition of Metabolic Shift Can Decrease Therapy Resistance in Human High-Grade Glioma Cells. Pathol Oncol Res, 2020, 26:23–33., DOI 10.1007/ s12253-019-00677-2 Felkai L, Krencz I, Kiss DJ, Nagy N, Pet}ovari G, Danko T, et al. Characterization of mTOR Activity and Metabolic Profile in Pediatric Rhabdomyosarcoma. Cancers, 2020, 12:1947., DOI 10.3390/cancers12071947 Pet}ovari G, Danko T, T}okes AM, Vetlenyi E, Krencz I, Raffay R, et al. In situ metabolic Characterisation of Breast Cancer and its Potential Impact on Therapy. Cancers, 2020, 9:2492., DOI 10.3390/cancers12092492 Rotundo MS, Galeano T, Tassone P, and Tagliaferri P. mTOR Inhibitors, a New Era for Metastatic Luminal HER2-Negative Breast Cancer? A Systematic Review and a Meta-Analysis of Randomized Trials. Oncotarget, 2016, 7(19): 27055–66., DOI 10.18632/oncotarget.7446 Wu K, Sun XQ, Wang CQ, Gao TX, Sun P, Wang Y, et al. Metronomic Combination Chemotherapy Using Everolimus and Etoposide for the Treatment of non-Hodgkin Lymphoma. Cancer Med, 2019, 8(10): 4688–98., DOI 10.1002/cam4.2364 Hue-Fontaine L, Lemelin A, Forestier J, Raverot G, Milot L, Robinson P, et al. Metformin and Everolimus in Neuroendocrine Tumours: A Synergic Effect?. Clin Res Hepatol Gastroenterol, 2020, 44(6):954–60., DOI 10.1016/j.clinre. 2020.02.011 Ariaans G, Jalving M, Vries EGEd., and Jong Sd. Anti-tumor Effects of Everolimus and Metformin Are Complementary and Glucose-dependent in Breast Cancer Cells. BMC Cancer, 2017, 17(1):232., DOI 10.1186/s12885-017- 3230-8 Moore EC, Cash HA, Caruso AM, Uppaluri R, Hodge JW, Van Waes C, et al. Enhanced Tumor Control with Combination mTOR and PD-L1 Inhibition in Syngeneic Oral Cavity Cancers. Cancer Immunol Res, 2016, 4(7):611–20., DOI 10.1158/2326-6066.CIR-15-0252 Ramesh A, Natarajan SK, Nandi D, and Kulkarni A. Dual Inhibitors-Loaded Nanotherapeutics that Target Kinase Signaling Pathways Synergize with Immune Checkpoint Inhibitor. Cel Mol Bioeng, 2019, 12(5):357–73., DOI 10.1007/s12195-019-00576-1 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609802 EP 1609816 DI 10.3389/pore.2021.1609802 PG 15 ER PT J AU Islam, Sh Kitagawa, T Kuramitsu, Y AF Islam, Shajedul Kitagawa, Takao Kuramitsu, Yasuhiro TI Prognostic Significance of Cofilin Isoforms in Patients With Pancreatic Ductal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE biomarker; pancreatic ductal adenocarcinoma; cofilin; proteomics; kaplan-meier ID biomarker; pancreatic ductal adenocarcinoma; cofilin; proteomics; kaplan-meier C1 [Islam, Shajedul] Health Sciences University of Hokkaido, Advanced Research Promotion CenterHokkaido, Japan. [Kitagawa, Takao] Health Sciences University of Hokkaido, Advanced Research Promotion CenterHokkaido, Japan. [Kuramitsu, Yasuhiro] Health Sciences University of Hokkaido, Advanced Research Promotion CenterHokkaido, Japan. RP Kuramitsu, Y (reprint author), Health Sciences University of Hokkaido, Advanced Research Promotion Center, Hokkaido, Japan. EM climates@hoku-iryo-u.ac.jp CR Lippolis R, and De Angelis M. Proteomics and Human Diseases. J Proteomics Bioinform, 2016, 09:3., DOI 10.4172/jpb.1000391 Wang Y, Kuramitsu Y, Ueno T, Suzuki N, Yoshino S, Iizuka N, et al. Differential Expression of Up-Regulated Cofilin-1 and Down-Regulated Cofilin-2 Characteristic of Pancreatic Cancer Tissues. Oncol Rep, 2011, 26:1595–9., DOI 10.3892/or.2011.1447 Nagy A, Munkacsy G, and Gyorffy B. Pancancer Survival Analysis of Cancer Hallmark Genes. Sci Rep, 2021, 11:6047., DOI 10.1038/s41598-021-84787-5 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609821 EP 1609822 DI 10.3389/pore.2021.1609821 PG 2 ER PT J AU Kim, JEJ Kocsmar, I Buzas, MGy Szirtes, I Rusz, O Diczhazi, Cs Szijarto, A Hritz, I Schaff, Zs Kiss, A Kocsmar, Lotz, G AF Kim, Ju Ea Jewel Kocsmar, Ildiko Buzas, Miklos Gyorgy Szirtes, Ildiko Rusz, Orsolya Diczhazi, Csaba Szijarto, Attila Hritz, Istvan Schaff, Zsuzsa Kiss, Andras Kocsmar, Eva Lotz, Gabor TI Efficacy of Clarithromycin Depends on the Bacterial Density in Clarithromycin-Heteroresistant Helicobacter pylori Infections: An In Situ Detected Susceptibility and Quantitative Morphometry-Based Retrospective Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE fluorescence in situ hybridization; Helicobacter pylori; clarithromycin; antibiotic resistance; heteroresistance; bacterial density ID fluorescence in situ hybridization; Helicobacter pylori; clarithromycin; antibiotic resistance; heteroresistance; bacterial density AB The global rise in clarithromycin (Cla) resistance is considered to be the main contributor of Helicobacter pylori (Hp) eradication failures. In nearly half of the Cla-resistant Hp infections, Clasusceptible bacteria are simultaneously present with the Cla-resistant ones (Claheteroresistance). The proportion of resistant bacteria in the bacterial population (R-fraction) and its predictive role for the use of Cla-based therapies in Cla-heteroresistant infections has not yet been investigated. Our retrospective study analyzed gastric biopsy samples of 62 Hp-positive patients with Cla-heteroresistant infection. Fluorescence In Situ Hybridization technique was used to visualize the coexistence of resistant and susceptible bacteria within one tissue sample. R-fraction was quantified on multichannel microimages by digital morphometry. Resistant bacteria had a patchy distribution within the whole bacterial population causing high diversity among the investigated areas. Patients were subdivided into two major groups according to whether a Cla-based eradication attempt was conducted before or after the biopsy sampling. R-fraction was significantly lower among cases having only one previous Cla-based eradication attempt vs. those that had multiple previous eradications, including at least one Cla-containing therapy (0.41 vs. 0.89, p = 0.0308). Majority of the patients without previous eradication attempt had successful eradicationwithCla-containing regimen (59.26%), verified by a negative 13C-urea breath test or control biopsy. Multivariable model indicated that the therapeutic outcome using Cla-based regimens depended on the bacterial density rather than the R-fraction. Our study raises the potential use of Cla-containing eradication therapies in certain Cla-heteroresistant Hp infections, taking into account the possible predictive role of bacterial density. C1 [Kim, Ju Ea Jewel] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kocsmar, Ildiko] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Buzas, Miklos Gyorgy] Ferencvaros Health Centre, Department of GastroenterologyBudapest, Hungary. [Szirtes, Ildiko] Peterfy Hospital - National Institute of Traumatology, Department of PharmacyBudapest, Hungary. [Rusz, Orsolya] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Diczhazi, Csaba] Peterfy Hospital - National Institute of Traumatology, Department of PathologyBudapest, Hungary. [Szijarto, Attila] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Hritz, Istvan] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Schaff, Zsuzsa] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kocsmar, Eva] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Lotz, G (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM lotz.gabor@med.semmelweisuniv.hu CR Pounder RE, and Ng D. The Prevalence of Helicobacter pylori Infection in Different Countries. Aliment Pharmaco Ther, 1995, 9(Suppl. 2):33–9. Leung WK, and Graham DY. Clarithromycin for Helicobacter pylori Infection. Expert Opin Pharmacother, 2000, 1(3):507–14., DOI 10.1517/14656566.1.3.507 Goddard A, Jessa M, Barrett D, Shaw P, Idstrom J, Cederberg C, et al. Effect of Omeprazole on the Distribution of Metronidazole, Amoxicillin, and Clarithromycin in Human Gastric Juice. Gastroenterology, 1996, 111(2): 358–67., DOI 10.1053/gast.1996.v111.pm8690200 Bazzoli F, Zagari M, Pozzato P, Varoli O, Fossi S, Ricciardiello L, et al. Evaluation of Short-Term Low-Dose Triple Therapy for the Eradication of Helicobacter pylori by Factorial Design in a Randomized, Double-Blind, Controlled Study. Aliment Pharmaco Ther, 1998, 12(5):439–45., DOI 10.1046/j.1365-2036.1998.00330.x Lind T, Zanten SV, Unge P, Spiller R, Bayerdorffer E, O’Morain C, et al. Eradication of Helicobacter pylori Using One-Week Triple Therapies Combining Omeprazole with Two Antimicrobials: the MACH I Study. Helicobacter, 1996, 1(3):138–44., DOI 10.1111/j.1523-5378.1996.tb00027.x Lind T, Megraud F, Unge P, Bayerdorffer E, O’morain C, Spiller R, et al. The MACH2 Study: Role of Omeprazole in Eradication of Helicobacter pylori with 1-week Triple Therapies. Gastroenterology, 1999, 116(2):248–53., DOI 10.1016/ s0016-5085(99)70119-8 Graham DY, and Qureshi WA. Antibiotic-resistant H. pylori Infection and its Treatment. Curr Pharm Des, 2000, 6(15):1537–44., DOI 10.2174/ 1381612003399077 Megraud F. H Pylori Antibiotic Resistance: Prevalence, Importance, and Advances in Testing. Gut, 2004, 53(9):1374–84., DOI 10.1136/gut.2003.022111 Graham DY, and Fischbach L. Helicobacter pylori Treatment in the Era of Increasing Antibiotic Resistance. Gut, 2010, 59(8):1143–53., DOI 10.1136/ gut.2009.192757 Thung I, Aramin H, Vavinskaya V, Gupta S, Park JY, Crowe SE, et al. Review Article: the Global Emergence ofHelicobacter Pyloriantibiotic Resistance. Aliment Pharmacol Ther, 2016, 43(4):514–33., DOI 10.1111/apt.13497 El-Serag HB, Kao JY, Kanwal F, Gilger M, LoVecchio F, Moss SF, et al. Houston Consensus Conference on Testing for Helicobacter pylori Infection in the United States. Clin Gastroenterol Hepatol, 2018, 16(7):992–1002. e6., DOI 10.1016/j.cgh.2018.03.013 Malfertheiner P,Megraud F, O’MorainCA, Gisbert JP, Kuipers EJ, AxonAT, et al. Management ofHelicobacter Pyloriinfection-The Maastricht V/Florence Consensus Report. Gut, 2017, 66(1):6–30., DOI 10.1136/gutjnl- 2016-312288 Megraud F, and Lehours P. Helicobacter pylori Detection and Antimicrobial Susceptibility Testing. Clin Microbiol Rev, 2007, 20(2):280–322., DOI 10.1128/ CMR.00033-06 Monno R, Giorgio F, Carmine P, Soleo L, Cinquepalmi V, and Ierardi E. Helicobacter pylori Clarithromycin Resistance Detected by Etest and TaqMan Real-Time Polymerase Chain Reaction: a Comparative Study. APMIS, 2012, 120(9):712–7., DOI 10.1111/j.1600-0463.2012.02896.x Alarcon T, Urruzuno P, Martinez MJ, Domingo D, Llorca L, Correa A, et al. Antimicrobial Susceptibility of 6 Antimicrobial Agents in Helicobacter pylori Clinical Isolates by Using EUCAST Breakpoints Compared with Previously Used Breakpoints. Enfermedades Infecciosas y Microbiologia Clinica, English ed.,, 2017, 35(5):278–82., DOI 10.1016/j.eimc.2016.02.01010.1016/ j.eimce.2017.03.018 Matteo MJ, Granados G, Olmos M, Wonaga A, and Catalano M Helicobacter pylori Amoxicillin Heteroresistance Due to point Mutations in PBP-1A in Isogenic Isolates. J Antimicrob Chemother, 2008, 61(3):474–7., DOI 10.1093/jac/ dkm504 Kim JJ, Kim JG, and Kwon DH. Mixed-infection of Antibiotic Susceptible and Resistant Helicobacter pylori Isolates in a Single Patient and Underestimation of Antimicrobial Susceptibility Testing. Helicobacter, 2003, 8(3):202–6., DOI 10.1046/j.1523-5378.2003.00145.x Kocsmar E, Kocsmar I, Buzas GM, Szirtes I, Wacha J, Takats A, et al. Helicobacter pylori Heteroresistance to Clarithromycin in Adults-New Data by In Situ Detection and Improved Concept. Helicobacter, 2020, 25(1). e12670., DOI 10.1111/hel.12670 Trebesius K, Panthel K, Strobel S, Vogt K, Faller G, Kirchner T, et al. Rapid and Specific Detection of Helicobacter pylori Macrolide Resistance in Gastric Tissue by Fluorescent In Situ Hybridisation. Gut, 2000, 46(5):608–14., DOI 10.1136/ gut.46.5.608 Russmann H, Adler K, Haas R, Gebert B, Koletzko S, and Heesemann J. Rapid and Accurate Determination of Genotypic Clarithromycin Resistance in Cultured Helicobacter pylori by Fluorescent In Situ Hybridization. J Clin Microbiol, 2001, 39(11):4142–4., DOI 10.1128/JCM.39.11.4142-4144.2001 Rizvanov AA, Haertle T, Bogomolnaya L, and Talebi Bezmin Abadi A. Helicobacter pylori and its Antibiotic Heteroresistance: A Neglected Issue in Published Guidelines. Front Microbiol, 2019, 10:1796., DOI 10.3389/ fmicb.2019.01796 Dixon MF, Genta RM, Yardley JH, and Correa P. Classification and Grading of Gastritis. Am J Surg Pathol, 1996, 20(10):1161–81., DOI 10.1097/00000478- 199610000-00001 Samarbaf-Zadeh AR, Tajbakhsh S, Moosavian SM, Sadeghi-Zadeh M, Azmi M, Hashemi J, et al. Application of Fluorescent In Situ Hybridization, FISH, for the Detection of Helicobacter pylori. Med Sci Mont, 2006, 12(10): CR426–CR430., DOI 10.1016/0016-5085(95)23178-1 Wolfgang V. Conducting Meta-Analyses in R with the Metafor Package. J Stat Softw, 2010, 36:1–48., DOI 10.7287/peerj.preprints.27608 Higgins JPT, Thompson SG, Deeks JJ, and Altman DG. Measuring Inconsistency in Meta-Analyses. Bmj, 2003, 327(7414):557–60., DOI 10.1136/ bmj.327.7414.557 Ghotaslou R, Leylabadlo HE, and Asl YM. Prevalence of Antibiotic Resistance in Helicobacter pylori: A Recent Literature Review. Wjm, 2015, 5(3):164–74., DOI 10.5662/wjm.v5.i3.164 Bilgilier C, Stadlmann A, Makristathis A, Thannesberger J, Kastner M-T, Knoflach P, et al. Prospective Multicentre Clinical Study on Inter- and Intrapatient Genetic Variability for Antimicrobial Resistance of Helicobacter pylori. Clin Microbiol Infect, 2018, 24(3):267–72., DOI 10.1016/ j.cmi.2017.06.025 Marzio L, Cellini L, Amitrano M, Grande R, Serio M, Cappello G, et al. Helicobacter pylori Isolates from Proximal and Distal Stomach of Patients Never Treated and Already Treated Show Genetic Variability and Discordant Antibiotic Resistance. Eur J Gastroenterol Hepatol, 2011, 23(6):1–472., DOI 10.1097/MEG.0b013e328345d40f Selgrad M, Tammer I, Langner C, Bornschein J, Meißle J, Kandulski A, et al. Different Antibiotic Susceptibility between Antrum and Corpus of the Stomach, a Possible Reason for Treatment Failure ofHelicobacter Pyloriinfection. Wjg, 2014, 20(43):16245–51., DOI 10.3748/wjg.v20.i43.16245 Kao C-Y, Lee A-Y, Huang A-H, Song P-Y, Yang Y-J, Sheu S-M, et al. Heteroresistance of Helicobacter pylori from the Same Patient Prior to Antibiotic Treatment. Infect Genet Evol, 2014, 23:196–202., DOI 10.1016/ j.meegid.2014.02.009 Crowe SE. Helicobacter pylori Infection. N Engl JMed, 2019, 380(12):1158–65., DOI 10.1056/NEJMcp1710945 De Francesco V, Zullo A, Ierardi E, Giorgio F, Perna F, Hassan C, et al. Phenotypic and Genotypic Helicobacter pylori Clarithromycin Resistance and Therapeutic Outcome: Benefits and Limits. J Antimicrob Chemother, 2010, 65(2):327–32., DOI 10.1093/jac/dkp445 O’Connor A, Liou JM, Gisbert JP, and O’Morain C. Review: Treatment of Helicobacter pylori Infection 2019. Helicobacter, 2019, 24(Suppl. 1):e12640., DOI 10.1111/hel.12640 Kocsmar E, Szirtes I, Kramer Z, Szijarto A, Bene L, Buzas GM, et al. Sensitivity of Helicobacter Pyloridetection by Giemsa Staining Is Poor in Comparison with Immunohistochemistry and Fluorescent In Situ Hybridization and Strongly Depends on Inflammatory Activity. Helicobacter, 2017, 22(4): e12387., DOI 10.1111/hel.12387 BenMansour K, Fendri C, Battikh H, Garnier M, Zribi M, Jlizi A, et al. Multiple and Mixed Helicobacter pylori Infections: Comparison of Two Epidemiological Situations in Tunisia and France. Infect Genet Evol, 2016, 37:43–8., DOI 10.1016/j.meegid.2015.10.028 Graham DY, Opekun AR, and Klein PD. Clarithromycin for the Eradication of Helicobacter pylori. J Clin Gastroenterol, 1993, 16(4):292–4., DOI 10.1097/ 00004836-199306000-00004 Qasim A, and O’Morain CA. Treatment of Helicobacter Pyloriinfection and Factors Influencing Eradication. Aliment Pharmacol Ther, 2002, 16(Suppl. 1): 24–30., DOI 10.1046/j.1365-2036.2002.0160s1024.x Kocsmar E, Buzas GM, Szirtes I, Kocsmar I, Kramer Z, Szijarto A, et al. Primary and Secondary Clarithromycin Resistance in Helicobacter pylori and Mathematical Modeling of the Role of Macrolides. Nat Commun, 2021, 12(1): 2255., DOI 10.1038/s41467-021-22557-7 Liu M, and Douthwaite S. Activity of the Ketolide Telithromycin Is Refractory to Erm Monomethylation of Bacterial rRNA. Antimicrob Agents Chemother, 2002, 46(6):1629–33., DOI 10.1128/aac.46.6.1629-1633.2002 Webber MA, and Piddock LJ. The Importance of Efflux Pumps in Bacterial Antibiotic Resistance. J Antimicrob Chemother, 2003, 51(1):9–11., DOI 10.1093/ jac/dkg050 Mamelli L, Amoros JP, Pages JM, and Bolla JM. A Phenylalanine-Arginine β-naphthylamide Sensitive Multidrug Efflux Pump Involved in Intrinsic and Acquired Resistance of Campylobacter to Macrolides. Int J Antimicrob Agents, 2003, 22(3):237–41., DOI 10.1016/s0924-8579(03)00199-7 Kocazeybek B, Sakli MK, Yuksel P, Demirci M, Caliskan R, Sarp TZ, et al. Comparison of New and Classical point Mutations Associated with Clarithromycin Resistance in Helicobacter pylori Strains Isolated from Dyspeptic Patients and Their Effects on Phenotypic Clarithromycin Resistance. J Med Microbiol, 2019, 68(4):566–73., DOI 10.1099/jmm.0.000944 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2021 VL 27 IS 2 BP 1609863 EP 1609873 DI 10.3389/pore.2021.1609863 PG 11 ER PT J AU Xu, N Guo, H Li, X Zhao, Q Li, J AF Xu, Ning Guo, Hui Li, Xurui Zhao, Qian Li, Jianguo TI A Five-Genes Based Diagnostic Signature for Sepsis-Induced ARDS SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE sepsis; ARDS; GO and KEGG analysis; logistic regression model; SVM model ID sepsis; ARDS; GO and KEGG analysis; logistic regression model; SVM model AB Background: Acute respiratory distress syndrome (ARDS) is a frequent and serious complication of sepsis without specific and sensitive diagnostic signatures. Methods: The mRNA profiles, including 60 blood samples with sepsis-induced ARDS and 86 blood samples with sepsis alone, were obtained from the Gene Expression Omnibus (GEO). The differently expressed genes (DEGs) were analyzed by limma package of R language. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were carried out using the clusterProfiler package of R. Eventually, multivariate logistic regression model was established through the glm function of R, and support vector machine (SVM) model was constructed via the e1071 package of R. Results: A total of 242 DEGs in GSE32707 and 102 DEGs in GSE66890 were identified. Notably, five genes exhibited significant differences between the two datasets and were considered to be closely associated with the occurrence of ARDS induced by sepsis. Furthermore, functional enrichment analysis based on the DEGs showed there were 80 overlapped GO terms and one KEGG pathway which were significantly enriched in the two datasets. The logistic regression model and SVM model constructed could efficiently distinguish sepsis patients with or without ARDS. Conclusion: In brief, our study suggested that NKG7, SPTA1, FGL2, RGS2, and IFI27 might be potential diagnostic signatures for sepsis-induced ARDS, which contributed to the future exploration in mechanism of ARDS occurrence and development. C1 [Xu, Ning] Hebei General Hospital, Department of EmergencyShijiazhuang, China. [Guo, Hui] Hebei General Hospital, Department of EmergencyShijiazhuang, China. [Li, Xurui] Hebei General Hospital, Department of General PracticeShijiazhuang, China. [Zhao, Qian] Hebei General Hospital, Department of EmergencyShijiazhuang, China. [Li, Jianguo] Hebei General Hospital, Department of EmergencyShijiazhuang, China. RP Li, J (reprint author), Hebei General Hospital, Department of Emergency, Shijiazhuang, China. EM lijianguo285@outlook.com CR Sweeney RM, and McAuley DF. Acute Respiratory Distress Syndrome. The Lancet, 2016, 388(10058):2416–30., DOI 10.1016/s0140-6736(16)00578-x Fan E, Brodie D, and Slutsky AS. Acute Respiratory Distress Syndrome. JAMA, 2018, 319(7):698–710., DOI 10.1001/jama.2017.21907 Confalonieri M, Salton F, and Fabiano F. Acute Respiratory Distress Syndrome. Eur Respir Rev, 2017, 26(144):160116., DOI 10.1183/16000617.0116-2016 Englert JA, Bobba C, and Baron RM. Integrating Molecular Pathogenesis and Clinical Translation in Sepsis-Induced Acute Respiratory Distress Syndrome. JCI Insight, 2019, 4(2):e124061., DOI 10.1172/jci.insight.124061 Huppert L, Matthay M, and Ware L. Pathogenesis of Acute Respiratory Distress Syndrome. Semin Respir Crit Care Med, 2019, 40(1):031–9., DOI 10.1055/s-0039-1683996 Eworuke E, Major JM, and Gilbert McClain LI. National Incidence Rates for Acute Respiratory Distress Syndrome, ARDS, and ARDS Cause-specific Factors in the United States, 2006-2014). J Crit Care, 2018, 47:192–7., DOI 10.1016/j.jcrc.2018.07.002 Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, et al. Epidemiology, Patterns of Care, and Mortality for Patients with Acute Respiratory Distress Syndrome in Intensive Care Units in 50 Countries. JAMA, 2016, 315(8):788–800., DOI 10.1001/jama.2016.0291 Force ADT, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, et al. Acute Respiratory Distress Syndrome: the Berlin Definition. JAMA, 2012, 307(23):2526–33., DOI 10.1001/jama.2012.5669 Stanski NL, and Wong HR. Prognostic and Predictive Enrichment in Sepsis. Nat Rev Nephrol, 2020, 16(1):20–31., DOI 10.1038/s41581-019-0199-3 Kangelaris KN, Prakash A, Liu KD, Aouizerat B, Woodruff PG, Erle DJ, et al. Increased Expression of Neutrophil-Related Genes in Patients with Early Sepsis-Induced ARDS. Am J Physiol Lung Cell Mol Physiol, 2015, 308(11): L1102–L1113., DOI 10.1152/ajplung.00380.2014 Zhang J, Luo Y, Wang X, Zhu J, Li Q, Feng J, et al. Global Transcriptional Regulation of STAT3- and MYC-Mediated Sepsis-Induced ARDS. Ther Adv Respir Dis, 2019, 13:1753466619879840., DOI 10.1177/1753466619879840 Zhang Z, Han N, and Shen Y. S100A12 Promotes Inflammation and Cell Apoptosis in Sepsis-Induced ARDS via Activation of NLRP3 Inflammasome Signaling. Mol Immunol, 2020, 122:38–48., DOI 10.1016/j.molimm.2020.03.022 XueM, Sun Z, Shao M, Yin J, Deng Z, Zhang J, et al. Diagnostic and Prognostic Utility of Tissue Factor for Severe Sepsis and Sepsis-Induced Acute Lung Injury. J Transl Med, 2015, 13:172., DOI 10.1186/s12967-015-0518-9 Dolinay T, Kim YS, Howrylak J, Hunninghake GM, An CH, Fredenburgh L, et al. Inflammasome-regulated Cytokines Are Critical Mediators of Acute Lung Injury. Am J Respir Crit Care Med, 2012, 185(11):1225–34., DOI 10.1164/ rccm.201201-0003oc Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res, 2015, 43(7):e47., DOI 10.1093/nar/gkv007 Yu G, Wang L-G, Han Y, and He Q-Y. clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters. OMICS: A J Integr Biol, 2012, 16(5):284–7., DOI 10.1089/omi.2011.0118 Friedman J, Hastie T, and Tibshirani R. Regularization Paths for Generalized Linear Models via Coordinate Descent. J Stat Softw, 2010, 33(1):1–22., DOI 10.18637/jss.v033.i01 Sagy M, Al-Qaqaa Y, and Kim P. Definitions and Pathophysiology of Sepsis. Curr Probl Pediatr Adolesc Health Care, 2013, 43(10):260–3., DOI 10.1016/ j.cppeds.2013.10.001 Stevens JP, Law A, and Giannakoulis J. Acute Respiratory Distress Syndrome. JAMA, 2018, 319(7):732., DOI 10.1001/jama.2018.0483 Ferre S, Deng Y, Huen SC, Lu CY, Scherer PE, Igarashi P, et al. Renal Tubular Cell Spliced X-Box Binding Protein 1, Xbp1s, Has a Unique Role in Sepsis-Induced Acute Kidney Injury and Inflammation. Kidney Int, 2019, 96(6):1359–73., DOI 10.1016/ j.kint.2019.06.023 Li L, Dong L, Zhao D, Gao F, and Yan J. Classical Dendritic Cells Regulate Acute Lung Inflammation and Injury in Mice with Lipopolysaccharide- Induced Acute Respiratory Distress Syndrome. Int J Mol Med, 2019, 44(2): 617–29., DOI 10.3892/ijmm.2019.4208 Meng L, Cao H, Wan C, and Jiang L. MiR-539-5p Alleviates Sepsis-Induced Acute Lung Injury by Targeting ROCK1. Folia Histochem Cytobiol, 2019, 57(4):168–78., DOI 10.5603/FHC.a2019.0019 Sweeney TE, Azad TD, Donato M, Haynes WA, Perumal TM, Henao R, et al. Unsupervised Analysis of Transcriptomics in Bacterial Sepsis Across Multiple Datasets Reveals Three Robust Clusters. Crit Care Med, 2018, 46(6):915–25., DOI 10.1097/ccm.0000000000003084 Mori S, Jewett A, Cavalcanti M, Murakami-Mori K, Nakamura S, and Bonavida B. Differential Regulation of Human NK Cell-Associated Gene Expression Following Activation by IL-2, IFN-Alpha and PMA/ionomycin. Int J Oncol, 1998, 12(5):1165–70., DOI 10.3892/ijo.12.5.1165 Gao Q, Cui Y, Shen Y, Li Y, Gao X, Xi Y, et al. Identifying Mutually Exclusive Gene Sets with Prognostic Value and Novel Potential Driver Genes in Patients with Glioblastoma. Biomed Res Int, 2019, 2019:4860367., DOI 10.1155/2019/ 4860367 Fan C, Wang J, Mao C, Li W, Liu K, and Wang Z. The FGL2 Prothrombinase Contributes to the Pathological Process of Experimental Pulmonary Hypertension. J Appl Physiol, 1985,, 2019, 127(6):1677–87., DOI 10.1152/ japplphysiol.00396.2019 Lee H-K, Park D-W, Bae JH, Kim HJ, Shin D-G, Park J-S, et al. RGS2 Is a Negative Regulator of STAT3-Mediated Nox1 Expression. Cell Signal, 2012, 24(3):803–9., DOI 10.1016/j.cellsig.2011.11.015 Chiang K-C, Huang S-T, Wu R-C, Huang S-C, Yeh T-S, Chen M-H, et al. Interferon α-inducible Protein 27 Is an Oncogene and Highly Expressed in Cholangiocarcinoma Patients with Poor Survival. Cancer Manag Res, 2019, 11:1893–905., DOI 10.2147/ cmar.s196485 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 580801 EP 580807 DI 10.3389/pore.2021.580801 PG 7 ER PT J AU Juris, A Taylor-Gehman, A Spencer, B Schaefer, E Pameijer, C AF Juris, Aubrey Taylor-Gehman, Amanda Spencer, Brianna Schaefer, Eric Pameijer, Colette TI The Impact of Sarcopenia in Patients with Peritoneal Surface Disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE sarcopenia; muscle mass; cytoreduction; outcomes; intraperitoneal chemotherapy; peritoneal surface disease ID sarcopenia; muscle mass; cytoreduction; outcomes; intraperitoneal chemotherapy; peritoneal surface disease AB Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy is increasingly performed in patients with advanced cancer in the abdomen. This treatment prolongs survival for some patients but is known to have a substantial rate of complications. Choosing patients for this procedure can be difficult, and no clear guidelines exist. Muscle mass is a general measure of a patient’s wellness, meaning that patients with low muscle mass for their body weight tend to have more complications from treatment and overall do worse. We evaluated muscle mass prior to surgery in our Cytoreductive surgery/ hyperthermic intraperitoneal chemotherapy population to assess how many patients have low muscle mass and the impact on outcomes, such as length of hospital stay, complications and survival. We find that about 25% of our patient population has low muscle mass, and low muscle mass is associated with a higher burden of cancer and shorter survival. We were able to evaluate muscle mass in a small number of patients after surgery, expecting to find decreased muscle mass in all the patients after a complex operation and long recovery. In fact, none of the patients had low muscle mass, including those who were low prior to surgery. C1 [Juris, Aubrey] The Pennsylvania State University, College of Medicine, Department of SurgeryHershey, PA, USA. [Taylor-Gehman, Amanda] The Pennsylvania State University, College of Medicine, Department of Family and Community MedicineHershey, PA, USA. [Spencer, Brianna] The Pennsylvania State University, College of Medicine, Department of SurgeryHershey, PA, USA. [Schaefer, Eric] The Pennsylvania State University, College of Medicine, Department of Public Health SciencesHershey, PA, USA. [Pameijer, Colette] The Pennsylvania State University, College of Medicine, Department of SurgeryHershey, PA, USA. RP Pameijer, C (reprint author), The Pennsylvania State University, College of Medicine, Department of Surgery, Hershey, USA. EM cpameijer@pennstatehealth.psu.edu CR Chemama S, Bayar MA, Lanoy E, Ammari S, Stoclin A, Goere D, et al. Sarcopenia is associated with chemotherapy toxicity in patients undergoing cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis from colorectal cancer. Ann Surg Oncol, 2016). 23(12):3891–8., DOI 10.1245/s10434-016-5360-7 van Vugt JLA, Braam HJ, van Oudheusden TR, Vestering A, Bollen TL, Wiezer MJ, et al. Skeletal muscle depletion is associated with severe postoperative complications in patients undergoing cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis of colorectal cancer. Ann Surg Oncol, 2015). 22(11):3625–31., DOI 10.1245/s10434-015- 4429-z Jacquet P, Sugarbaker PH. Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis. Cancer Treat Res, 1996). 82: 359–74., DOI 10.1007/978-1-4613-1247-5_23 Lodewick TM, Arnoldussen CWKP, Lahaye MJ, van Mierlo KMC, Neumann UP, Beets-Tan RG, et al. Fast and accurate liver volumetry prior to hepatectomy. Hpb, 2016). 18(9):764–72., DOI 10.1016/j.hpb.2016.06.009 Chung E, Lee HS, Cho E-S, Park EJ, Baik SH, Lee KY, et al. Prognostic significance of sarcopenia and skeletal muscle mass change during preoperative chemoradiotherapy in locally advanced rectal cancer. Clin Nutr, 2020). 39(3):820–8., DOI 10.1016/j.clnu.2019.03.014 Cespedes Feliciano EM, Kroenke CH, Meyerhardt JA, Prado CM, Bradshaw PT, Kwan ML, et al. Association of systemic inflammation and sarcopenia with survival in nonmetastatic colorectal cancer: results from the C SCANS study. JAMA Oncol, 2017). 3(12):e172319., DOI 10.1001/jamaoncol.2017.2319 Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc, 1958). 53:457–81., DOI 10.1080/01621459.1958.10501452 Mayhew AJ, Amog K, Phillips S, Parise G, McNicholas PD, de Souza RJ, et al. The prevalence of sarcopenia in community-dwelling older adults, an exploration of differences between studies and within definitions: a systematic review and meta-analyses. Age Ageing, 2019). 48(1):48–56., DOI 10. 1093/ageing/afy106 Hanach NI, McCullough F, Avery A. The impact of dairy protein intake on muscle mass, muscle strength, and physical performance in middle-aged to older adults with or without existing sarcopenia: a systematic review and meta-analysis. Adv Nutr, 2019). 10(1):59–69., DOI 10.1093/advances/ nmy065 van Vledder MG, Levolger S, Ayez N, Verhoef C, Tran TCK, Ijzermans JNM. Body composition and outcome in patients undergoing resection of colorectal liver metastases. Br J Surg, 2012). 99(4):550–7., DOI 10.1002/bjs.7823 Lieffers JR, Bathe OF, Fassbender K, Winget M, Baracos VE. Sarcopenia is associated with postoperative infection and delayed recovery from colorectal cancer resection surgery. Br J Cancer, 2012). 107(6):931–6., DOI 10.1038/bjc. 2012.350 Gupta D, Lis CG. Pretreatment serum albumin as a predictor of cancer survival: a systematic review of the epidemiological literature. Nutr J, 2010). 9(9), 27., DOI 10.1186/1475-2891-9-69 Choi Y, Oh DY, Kim TY, Lee KH, Han SW, Im SA, et al. Skeletal muscle depletion predicts the prognosis of patients with advanced pancreatic cancer undergoing palliative chemotherapy, independent of body mass index. PLoS One, 2015). 10(10):e0139749., DOI 10.1371/journal.pone.0139749 Xiao DY, Luo S, O’Brian K, Ganti A, Riedell P, Sanfilippo KM, et al. Impact of sarcopenia on treatment tolerance in United States veterans with diffuse large B-cell lymphoma treated with CHOP-based chemotherapy. Am J Hematol, 2016). 91(10):1002–7., DOI 10.1002/ajh.24465 Pin F, Couch ME, Bonetto A. Preservation of muscle mass as a strategy to reduce the toxic effects of cancer chemotherapy on body composition. Curr Opin Support Palliat Care, 2018). 12(4):420–6., DOI 10.1097/spc. 0000000000000382 Zimmer P, Trebing S, Timmers-Trebing U, Schenk A, Paust R, Bloch W, et al. Eight-week, multimodal exercise counteracts a progress of chemotherapy-induced peripheral neuropathy and improves balance and strength in metastasized colorectal cancer patients: a randomized controlled trial. Support Care Cancer, 2018). 26(2):615–24., DOI 10.1007/ s00520-017-3875-5 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 638857 EP 638861 DI 10.3389/pore.2021.638857 PG 5 ER PT J AU Qi, W Yan, Q Lv, M Song, D Wang, X Tian, K AF Qi, Wei Yan, Qian Lv, Ming Song, Delei Wang, Xianbin Tian, Kangsong TI Prognostic Signature of Osteosarcoma Based on 14 Autophagy-Related Genes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; risk score; osteosarcoma; autophagy-related genes; nomogram ID prognosis; risk score; osteosarcoma; autophagy-related genes; nomogram AB Background: Osteosarcoma is a common malignancy of bone with inferior survival outcome. Autophagy can exert multifactorial influence on tumorigenesis and tumor progression. However, the specific function of genes related to autophagy in the prognosis of osteosarcoma patients remains unclear. Herein, we aimed to explore the association of genes related to autophagy with the survival outcome of osteosarcoma patients. Methods: The autophagy-associated genes that were related to the prognosis of osteosarcoma were optimized by LASSO Cox regression analysis. The survival of osteosarcoma patients was forecasted by multivariate Cox regression analysis. The immune infiltration status of 22 immune cell types in osteosarcoma patients with high and low risk scores was compared by using the CIBERSORT tool. Results: The risk score model constructed according to 14 autophagy-related genes (ATG4A, BAK1, BNIP3, CALCOCO2, CCL2, DAPK1, EGFR, FAS, GRID2, ITGA3, MYC, RAB33B, USP10, and WIPI1) could effectively predict the prognosis of patients with osteosarcoma. A nomogram model was established based on risk score and metastasis. Conclusion: Autophagy-related genes were identified as pivotal prognostic signatures, which could guide the clinical decision making in the treatment of osteosarcoma. C1 [Qi, Wei] Zibo Central Hospital, Department ofWest Hospital Orthopaedic TraumaZibo, China. [Yan, Qian] Zibo Central Hospital, Department of Information SectionZibo, China. [Lv, Ming] Zibo Central Hospital, Department ofWest Hospital Orthopaedic TraumaZibo, China. [Song, Delei] Zibo Central Hospital, Department ofWest Hospital Orthopaedic TraumaZibo, China. [Wang, Xianbin] Zibo Central Hospital, Department of Eastern Hospital Orthopaedic TraumaZibo, China. [Tian, Kangsong] Zibo Central Hospital, Department ofWest Hospital Orthopaedic TraumaZibo, China. RP Tian, K (reprint author), Zibo Central Hospital, Department ofWest Hospital Orthopaedic Trauma, Zibo, China. EM tiankangsong@outlook.com CR Picci P. Osteosarcoma, Osteogenic Sarcoma). Orphanet J Rare Dis, 2007, 2:6., DOI 10.1186/1750-1172-2-6 Mirabello L, Troisi RJ, and Savage SA. Osteosarcoma Incidence and Survival Rates from 1973 to 2004. Cancer, 2009, 115(7):1531–43., DOI 10.1002/ cncr.24121 Singh HB, Singh H, and Chakraborty M. Metastatic Osteosarcoma of the Maxilla. J Laryngol Otol, 1978, 92(7):619–22., DOI 10.1017/s0022215100085832 Wolfe TD, Pillai SPS, Hildreth BE, 3rd, Lanigan LG, Martin CK, Werbeck JL, et al. Effect of Zoledronic Acid and Amputation on Bone Invasion and Lung Metastasis of Canine Osteosarcoma in Nude Mice. Clin Exp Metastasis, 2011, 28(4):377–89., DOI 10.1007/s10585-011-9377-9 Smeland S, Bielack SS, Whelan J, Bernstein M, Hogendoorn P, Krailo MD, et al. Survival and Prognosis with Osteosarcoma: Outcomes in More Than 2000 Patients in the EURAMOS-1, European and American Osteosarcoma Study, Cohort. Eur J Cancer, 2019, 109:36–50., DOI 10.1016/j.ejca.2018.11.027 White E, Mehnert JM, and Chan CS. Autophagy, Metabolism, and Cancer. Clin Cancer Res, 2015, 21(22):5037–46., DOI 10.1158/1078-0432.ccr-15-0490 Dong W, and Wang R. [Effects of Resveratrol-Induced Cellular Autophagy in Control of Neurodegenerative Diseases]. Yao Xue Xue Bao, 2016, 51(1):18–22., DOI 10.16438/j.0513-4870.2015-0660 Wu S-Y, Lan S-H, Cheng D-E, Chen W-K, Shen C-H, Lee Y-R, et al. Rasrelated Tumorigenesis Is Suppressed by BNIP3-Mediated Autophagy through Inhibition of Cell Proliferation. Neoplasia, 2011, 13(12):1171–82., DOI 10.1593/ neo.11888 Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, et al. Autophagy Promotes Tumor Cell Survival and Restricts Necrosis, Inflammation, and Tumorigenesis. Cancer Cell, 2006, 10(1):51–64., DOI 10.1016/j.ccr.2006.06.001 Mathew R, Khor S, Hackett SR, Rabinowitz JD, Perlman DH, and White E. Functional Role of Autophagy-Mediated Proteome Remodeling in Cell Survival Signaling and Innate Immunity. Mol Cel, 2014, 55(6):916–30., DOI 10.1016/j.molcel.2014.07.019 Onorati AV, Dyczynski M, Ojha R, and Amaravadi RK. Targeting Autophagy in Cancer. Cancer, 2018, 124(16):3307–18., DOI 10.1002/cncr.31335 O’Farrill JS, and Gordon N. Autophagy in Osteosarcoma. Adv Exp Med Biol, 2014, 804:147–60., DOI 10.1007/978-3-319-04843-7_8 Huang J, Liu K, Yu Y, Xie M, Kang R, Vernon PJ, et al. Targeting HMGB1- Mediated Autophagy as a Novel Therapeutic Strategy for Osteosarcoma. Autophagy, 2012, 8(2):275–7., DOI 10.4161/auto.8.2.18940 Huang Y, Liu W, He B, Wang L, Zhang F, Shu H, et al. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells Promote Osteosarcoma Development by Activating Oncogenic Autophagy. J Bone Oncol, 2020, 21:100280., DOI 10.1016/j.jbo.2020.100280 Liu Y, Gu S, Li H, Wang J, Wei C, and Liu Q. SNHG16 Promotes Osteosarcoma Progression and Enhances Cisplatin Resistance by Sponging miR-16 to Upregulate ATG4B Expression. Biochem Biophysical Res Commun, 2019, 518(1):127–33., DOI 10.1016/j.bbrc.2019.08.019 Buddingh EP, KuijjerML, Duim RAJ, Burger H, Agelopoulos K,Myklebost O, et al. Tumor-infiltrating Macrophages Are Associated with Metastasis Suppression in High-Grade Osteosarcoma: a Rationale for Treatment withMacrophage Activating Agents. Clin Cancer Res, 2011, 17(8):2110–9., DOI 10.1158/1078-0432.ccr-10-2047 Paoloni M, Davis S, Lana S, Withrow S, Sangiorgi L, Picci P, et al. Canine Tumor Cross-Species Genomics Uncovers Targets Linked to Osteosarcoma Progression. BMC Genomics, 2009, 10:625., DOI 10.1186/1471-2164-10-625 Mao D, Zhang Z, Zhao X, and Dong X. Autophagy-related Genes Prognosis Signature as Potential Predictive Markers for Immunotherapy in Hepatocellular Carcinoma. PeerJ, 2020, 8:e8383., DOI 10.7717/peerj.8383 Friedman J, Hastie T, and Tibshirani R. Regularization Paths for Generalized Linear Models via Coordinate Descent. J Stat Softw, 2010, 33(1):1–22., DOI 10.18637/jss.v033.i01 Heagerty PJ, Lumley T, and Pepe MS. Time-dependent ROC Curves for Censored Survival Data and a Diagnostic Marker. Biometrics, 2000, 56(2): 337–44., DOI 10.1111/j.0006-341x.2000.00337.x Jager KJ, van Dijk PC, Zoccali C, and Dekker FW. The Analysis of Survival Data: the Kaplan-Meier Method. Kidney Int, 2008, 74(5):560–5., DOI 10.1038/ ki.2008.217 Rey D, and Neuhauser. M. Wilcoxon-signed-rank Test. Springer Berlin Heidelberg, 2014). Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust Enumeration of Cell Subsets from Tissue Expression Profiles. Nat Methods, 2015, 12(5):453–7., DOI 10.1038/nmeth.3337 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: a Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci, 2005, 102(43):15545–50., DOI 10.1073/pnas.0506580102 Fang C, Wang W, Feng X, Sun J, Zhang Y, Zeng Y, et al. Nomogram Individually Predicts the Overall Survival of Patients with Gastroenteropancreatic Neuroendocrine Neoplasms. Br J Cancer, 2017, 117(10):1544–50., DOI 10.1038/bjc.2017.315 Dong S, Huo H, Mao Y, Li X, and Dong L. A Risk Score Model for the Prediction of Osteosarcoma Metastasis. FEBS Open Bio, 2019, 9(3):519–26., DOI 10.1002/2211-5463.12592 Ingvarsen SZ, Gardsvoll H, van Putten S, Norregaard KS, Krigslund O, Meilstrup JA, et al. Tumor Cell MT1-MMP Is Dispensable for Osteosarcoma Tumor Growth, Bone Degradation and Lung Metastasis. Sci Rep, 2020, 10(1):19138., DOI 10.1038/s41598-020-75995-6 Allison DC, Carney SC, Ahlmann ER, Hendifar A, Chawla S, Fedenko A, et al. A Meta-Analysis of Osteosarcoma Outcomes in the Modern Medical Era. Sarcoma, 2012, 2012:704872., DOI 10.1155/2012/704872 Luetke A, Meyers PA, Lewis I, and Juergens H. Osteosarcoma Treatment - where Do We Stand? A State of the Art Review. Cancer Treat Rev, 2014, 40(4): 523–32., DOI 10.1016/j.ctrv.2013.11.006 Takamura A, Komatsu M, Hara T, Sakamoto A, Kishi C, Waguri S, et al. Autophagy-deficient Mice Develop Multiple Liver Tumors. Genes Develop, 2011, 25(8):795–800., DOI 10.1101/gad.2016211 Rosenfeldt MT, O’Prey J, Morton JP, Nixon C, MacKay G, Mrowinska A, et al. p53 Status Determines the Role of Autophagy in Pancreatic Tumour Development. Nature, 2013, 504(7479):296–300., DOI 10.1038/nature12865 Yang S, Wang X, Contino G, Liesa M, Sahin E, Ying H, et al. Pancreatic Cancers Require Autophagy for Tumor Growth. Genes Develop, 2011, 25(7): 717–29., DOI 10.1101/gad.2016111 Lock R, Kenific CM, Leidal AM, Salas E, and Debnath J. Autophagy-dependent Production of Secreted Factors Facilitates Oncogenic RAS-Driven Invasion. Cancer Discov, 2014, 4(4):466–79., DOI 10.1158/2159-8290.cd-13-0841 Su H, Zhu G, Rong X, Zhou Y, Jiang P, and Chen P. Upregulation of ATG4A Promotes Osteosarcoma Cell Epithelial-To-Mesenchymal Transition through the Notch Signaling Pathway. Int J Clin Exp Pathol, 2017, 10(7):7975–82. He Q, Lu Y, Hu S, Huang Q, Li S, Huang Y, et al. An Intron SNP Rs807185 in ATG4A Decreases the Risk of Lung Cancer in a Southwest Chinese Population. Eur J Cancer Prev, 2016, 25(4):255–8., DOI 10.1097/cej.0000000000000174 Liao Y-P, Chen L-Y, Huang R-L, Su P-H, Chan MWY, Chang C-C, et al. Hypomethylation Signature of Tumor-Initiating Cells Predicts Poor Prognosis of Ovarian Cancer Patients. Hum Mol Genet, 2014, 23(7):1894–906., DOI 10.1093/hmg/ddt583 Lian B, Hu X, and Shao ZM. Unveiling Novel Targets of Paclitaxel Resistance by Single Molecule Long-Read RNA Sequencing in Breast Cancer. Sci Rep, 2019, 9(1):6032., DOI 10.1038/s41598-019-42184-z Gu X-y., Wang J, Luo Y-z., Du Q, Li R-r., Shi H, et al. Down-regulation of miR- 150 Induces Cell Proliferation Inhibition and Apoptosis in Non-small-cell Lung Cancer by Targeting BAK1 In Vitro. Tumor Biol, 2014, 35(6):5287–93., DOI 10.1007/s13277-014-1688-4 Wang YD, Cai N, Wu XL, Cao HZ, Xie LL, and Zheng PS. OCT4 Promotes Tumorigenesis and Inhibits Apoptosis of Cervical Cancer Cells by miR-125b/ BAK1 Pathway. Cell Death Dis, 2013, 4:e760., DOI 10.1038/cddis.2013.272 Jin Y, and Qin X. Development of a Prognostic Signature Based on Autophagy- Related Genes for Head and Neck Squamous Cell Carcinoma. Arch Med Res, 2020, 51(8):860–7., DOI 10.1016/j.arcmed.2020.09.009 Chourasia AH, Tracy K, Frankenberger C, Boland ML, Sharifi MN, Drake LE, et al. Mitophagy Defects Arising from BNip3 Loss Promote Mammary Tumor Progression to Metastasis. EMBO Rep, 2015, 16(9):1145–63., DOI 10.15252/ embr.201540759 Giatromanolaki A, Koukourakis MI, Sowter HM, Sivridis E, Gibson S, Gatter KC, et al. BNIP3 Expression Is Linked with Hypoxia-Regulated Protein Expression and with Poor Prognosis in Non-small Cell Lung Cancer. Clin Cancer Res, 2004, 10(16):5566–71., DOI 10.1158/1078-0432.ccr-04-0076 Jiang Z, Yu F, and Li M. Upregulation of BCL2 19 kD Protein-Interacting Protein 3, BNIP3, Is Predictive of Unfavorable Prognosis in Uveal Melanoma. Med Sci Monit, 2018, 24:4711–7., DOI 10.12659/msm.907679 Ye F, Wang H, Zhang L, Zou Y, Han H, and Huang J. Baicalein Induces Human Osteosarcoma Cell Line MG-63 Apoptosis via ROS-Induced BNIP3 Expression. Tumor Biol, 2015, 36(6):4731–40., DOI 10.1007/s13277-015-3122-y Cao W, Liu N, Tang S, Bao L, Shen L, Yuan H, et al. Acetyl-Coenzyme A Acyltransferase 2 Attenuates the Apoptotic Effects of BNIP3 in Two Human Cell Lines. Biochim Biophys Acta, Bba, - Gen Subjects, 2008, 1780(6):873–80., DOI 10.1016/j.bbagen.2008.02.007 Lazarou M, Sliter DA, Kane LA, Sarraf SA, Wang C, Burman JL, et al. The Ubiquitin Kinase PINK1 Recruits Autophagy Receptors to Induce Mitophagy. Nature, 2015, 524(7565):309–14., DOI 10.1038/nature14893 Zhu FX, Wang XT, Zeng HQ, Yin ZH, and Ye ZZ. A Predicted Risk Score Based on the Expression of 16 Autophagy-Related Genes for Multiple Myeloma Survival. Oncol Lett, 2019, 18(5):5310–24., DOI 10.3892/ol.2019.10881 Deshmane SL, Kremlev S, Amini S, and Sawaya BE. Monocyte Chemoattractant Protein-1, MCP-1): an Overview. J Interferon Cytokine Res, 2009, 29(6):313–26., DOI 10.1089/jir.2008.0027 Bakst RL, Xiong H, Chen C-H, Deborde S, Lyubchik A, Zhou Y, et al. Inflammatory Monocytes Promote Perineural Invasion via CCL2-Mediated Recruitment and Cathepsin B Expression. Cancer Res, 2017, 77(22):6400–14., DOI 10.1158/0008-5472.can-17-1612 Carrero Y, Mosquera J, Callejas D, and Alvarez-Mon M. In Situ increased Chemokine Expression in Human Cervical Intraepithelial Neoplasia. Pathol - Res Pract, 2015, 211(4):281–5., DOI 10.1016/j.prp.2015.01.002 Wu J, Liu X, and Wang Y. Predictive Value of Preoperative Serum CCL2, CCL18, and VEGF for the Patients with Gastric Cancer. BMC Clin Pathol, 2013, 13:15., DOI 10.1186/1472-6890-13-15 Chen Q, Sun W, Liao Y, Zeng H, Shan L, Yin F, et al. Monocyte Chemotactic Protein-1 Promotes the Proliferation and Invasion of Osteosarcoma Cells and Upregulates the Expression of AKT. Mol Med Rep, 2015, 12(1):219–25., DOI 10.3892/mmr.2015.3375 Singh P, Ravanan P, and Talwar P. Death Associated Protein Kinase 1, DAPK1): A Regulator of Apoptosis and Autophagy. Front Mol Neurosci, 2016, 9:46., DOI 10.3389/fnmol.2016.00046 Schneider-Stock R. Death-associated Kinase, DAPK): a Cancer "gene Chameleon". Apoptosis, 2014, 19(2):285., DOI 10.1007/s10495-013-0932-5 Kristensen LS, Asmar F, Dimopoulos K, Nygaard MK, Aslan D, Hansen JW, et al. Hypermethylation of DAPK1is an Independent Prognostic Factor Predicting Survival in Diffuse Large B-Cell Lymphoma. Oncotarget, 2014, 5(20):9798–810., DOI 10.18632/oncotarget.2394 Li L, Guo L, Wang Q, Liu X, Zeng Y, Wen Q, et al. DAPK1 as an Independent Prognostic Marker in Liver Cancer. PeerJ, 2017, 5:e3568., DOI 10.7717/peerj.3568 Lin Y, Wang X, and Jin H. EGFR-TKI Resistance in NSCLC Patients: Mechanisms and Strategies. Am J Cancer Res, 2014, 4(5):411–35., DOI 10.1016/B978-0-12-420117-0.00008-6 Rajaram P, Chandra P, Ticku S, Pallavi BK, Rudresh KB, and Mansabdar P. Epidermal Growth Factor Receptor: Role in Human Cancer. Indian J Dent Res, 2017, 28(6):687–94., DOI 10.4103/ijdr.IJDR_534_16 Nicholson S,WrightC, Richard C. Sainsbury J,Halcrow P, Kelly P,Angus B, et al. Epidermal Growth Factor Receptor, EGFr, as a Marker for Poor Prognosis in Node-Negative Breast Cancer Patients: Neu and Tamoxifen Failure. J Steroid Biochem Mol Biol, 1990, 37(6):811–4., DOI 10.1016/0960-0760(90)90424-j Wang SL, Zhong GX,Wang XW, Yu FQ,Weng DF, Wang XX, et al. Prognostic Significance of the Expression of HER Family Members in Primary Osteosarcoma. Oncol Lett, 2018, 16(2):2185–94., DOI 10.3892/ol.2018.8931 Kersting C, Gebert C, Agelopoulos K, Schmidt H, van Diest PJ, Juergens H, et al. Epidermal Growth Factor Receptor Expression in High-Grade Osteosarcomas Is Associated with a Good Clinical Outcome. Clin Cancer Res, 2007, 13(10):2998–3005., DOI 10.1158/1078-0432.ccr-06-2432 Liao C-C, Chen S-C, Huang H-P, and Wang C-J. Gallic Acid Inhibits Bladder Cancer Cell Proliferation and Migration via Regulating Fatty Acid Synthase, FAS). J Food Drug Anal, 2018, 26(2):620–7., DOI 10.1016/j.jfda.2017.06.006 Bebenek M, Du´s D, and Ko´zlak J. Fas and Fas Ligand as Prognostic Factors in Human Breast Carcinoma. Med Sci Monit, 2006, 12(11):CR457–61., DOI 10.1051/medsci/200622111003 Ali Z, Zulfiqar S, Klar J, Wikstrom J, Ullah F, Khan A, et al. Homozygous GRID2 Missense Mutation Predicts a Shift in the D-Serine Binding Domain of GluD2 in a Case with Generalized Brain Atrophy and Unusual Clinical Features. BMC Med Genet, 2017, 18(1):144., DOI 10.1186/s12881-017-0504-6 Ngollo M, Lebert A, Daures M, Judes G, Rifai K, Dubois L, et al. Global Analysis of H3K27me3 as an Epigenetic Marker in Prostate Cancer Progression. BMC Cancer, 2017, 17(1):261., DOI 10.1186/s12885-017-3256-y Qiu J, Sun M, Wang Y, and Chen B. Identification and Validation of an Individualized Autophagy-Clinical Prognostic index in Gastric Cancer Patients. Cancer Cel Int, 2020, 20:178., DOI 10.1186/s12935-020-01267-y Ura H, Denno R, Hirata K, Yamaguchi K, and Yasoshima T. Separate Functions of α2β1 and α2β1 Integrins in the Metastatic Process of Human Gastric Carcinomaintegrins in the Metastatic Process of Human Gastric Carcinoma. Surg Today, 1998, 28(10):1001–6., DOI 10.1007/bf02483952 Bauer KM, Watts TN, Buechler S, and Hummon AB. Proteomic and Functional Investigation of the colon Cancer Relapse-Associated Genes NOX4 and ITGA3. J Proteome Res, 2014, 13(11):4910–8., DOI 10.1021/ pr500557n Yang W, He M, Zhao J, and Wang Z. Association of ITGA3 Gene Polymorphisms with Susceptibility and Clinicopathological Characteristics of Osteosarcoma. Med Oncol, 2014, 31(2):826., DOI 10.1007/s12032-013- 0826-y Elliott B, Millena AC, Matyunina L, Zhang M, Zou J, Wang G, et al. Essential Role of JunD in Cell Proliferation Is Mediated via MYC Signaling in Prostate Cancer Cells. Cancer Lett, 2019, 448:155–67., DOI 10.1016/j.canlet.2019.02.005 Gabay M, Li Y, and Felsher DW. MYC Activation Is a Hallmark of Cancer Initiation and Maintenance. Cold Spring Harb Perspect Med, 2014, 4(6): a014241., DOI 10.1101/cshperspect.a014241 Wang Y, Wu M-C, Sham JST, ZhangW,WuW-Q, and Guan X-Y. Prognostic Significance Ofc-Myc andAIB1 Amplification in Hepatocellular Carcinoma. Cancer, 2002, 95(11):2346–52., DOI 10.1002/cncr.10963 Han G, Wang Y, and Bi W. C-myc Overexpression Promotes Osteosarcoma Cell Invasion via Activation of MEK-ERK Pathway. Oncol Res Featuring Preclinical Clin Cancer Ther, 2012, 20(4):149–56., DOI 10.3727/ 096504012x13522227232237 Morgan NE, Cutrona MB, and Simpson JC. Multitasking Rab Proteins in Autophagy and Membrane Trafficking: A Focus on Rab33b. Int J Mol Sci, 2019, 20(16):3916., DOI 10.3390/ijms20163916 Zhang C, LengW, Sun C, Lu T, Chen Z, Men X, et al. Urine Proteome Profiling Predicts Lung Cancer from Control Cases and Other Tumors. EBioMedicine, 2018, 30:120–8., DOI 10.1016/j.ebiom.2018.03.009 Zhang M, Hu C, Tong D, Xiang S, Williams K, Bai W, et al. Ubiquitin-specific Peptidase 10, USP10, Deubiquitinates and Stabilizes MutS Homolog 2, MSH2, to Regulate Cellular Sensitivity to DNA Damage. J Biol Chem, 2016, 291(20):10783–91., DOI 10.1074/jbc.m115.700047 Liu J, Xia H, Kim M, Xu L, Li Y, Zhang L, et al. Beclin1 Controls the Levels of P53 by Regulating the Deubiquitination Activity of USP10 and USP13. Cell, 2011, 147(1):223–34., DOI 10.1016/j.cell.2011.08.037 Sun J, Li T, Zhao Y, Huang L, Sun H, Wu H, et al. USP10 Inhibits Lung Cancer Cell Growth and Invasion by Upregulating PTEN. Mol Cel Biochem, 2018, 441(1-2):1–7., DOI 10.1007/s11010-017-3170-2 Zeng Z, Wu H-X, Zhan N, Huang Y-B, Wang Z-S, Yang G-F, et al. Prognostic Significance of USP10 as a Tumor-Associated Marker in Gastric Carcinoma. Tumor Biol, 2014, 35(4):3845–53., DOI 10.1007/s13277-013-1509-1 Tsuyuki S, Takabayashi M, Kawazu M, Kudo K, Watanabe A, Nagata Y, et al. Detection ofWIPI1mRNA as an Indicator of Autophagosome Formation. Autophagy, 2014, 10(3):497–513., DOI 10.4161/auto.27419 D’Arcangelo D, Giampietri C, Muscio M, Scatozza F, Facchiano F, and Facchiano A. WIPI1, BAG1, and PEX3 Autophagy-Related Genes Are Relevant Melanoma Markers. Oxid Med Cel Longev, 2018, 2018:1471682., DOI 10.1155/2018/1471682 Sun Y, Wang X, Chen B, Xiong L, and Liu Y. Prognostic Value of Autophagy- Related Genes Correlated with Metastasis in Uveal Melanoma Patients, 2021). Ran T, Ke S, Song X, Ma T, Xu Y, and Wang M. WIPI1 Promotes Osteosarcoma Cell Proliferation by Inhibiting CDKN1A. Gene, 2021, 782: 145537., DOI 10.1016/j.gene.2021.145537 Ouyang Z, Li G, Zhu H, Wang J, Qi T, Qu Q, et al. Construction of a Five- Super-Enhancer-Associated-Genes Prognostic Model for Osteosarcoma Patients. Front Cel Dev Biol, 2020, 8:598660., DOI 10.3389/fcell.2020.598660 Tian L, Chen S, Li C, Zhong D, and Xu C. A Novel Prognostic Model to Predict Prognosis of Patients with Osteosarcoma Based on Clinical Characteristics and Blood Biomarkers, 2020). Yang B, Su Z, Chen G, Zeng Z, Tan J,Wu G, et al. Identification of Prognostic Biomarkers Associated with Metastasis and Immune Infiltration in Osteosarcoma. Oncol Lett, 2021, 21(3):180., DOI 10.3892/ol.2021.12441 Zhang C, Zheng J-H, Lin Z-H, Lv H-Y, Ye Z-M, Chen Y-P, et al. Profiles of Immune Cell Infiltration and Immune-Related Genes in the Tumor Microenvironment of Osteosarcoma. Aging, 2020, 12(4):3486–501., DOI 10.18632/aging.102824 Gomez-Brouchet A, Illac C, Gilhodes J, Bouvier C, Aubert S, Guinebretiere JM, et al. CD163-positive Tumor-Associated Macrophages and CD8-Positive Cytotoxic Lymphocytes Are Powerful Diagnostic Markers for the Therapeutic Stratification of Osteosarcoma Patients: An Immunohistochemical Analysis of the Biopsies Fromthe French OS2006 Phase 3 Trial. Oncoimmunology, 2017, 6(9):e1331193., DOI 10.1080/2162402x.2017.1331193 Fritzsching B, Fellenberg J, Moskovszky L, Sapi Z, Krenacs T, Machado I, et al. CD8(+)/FOXP3(+)-ratio in Osteosarcoma Microenvironment Separates Survivors from Non-survivors: a Multicenter Validated Retrospective Study. Oncoimmunology, 2015, 4(3):e990800., DOI 10.4161/2162402x.2014.990800 Chen Y, Zhao B, and Wang X. Tumor Infiltrating Immune Cells, TIICs, as a Biomarker for Prognosis Benefits in Patients with Osteosarcoma. BMC Cancer, 2020, 20(1):1022., DOI 10.1186/s12885-020-07536-3 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609782 EP 1609795 DI 10.3389/pore.2021.1609782 PG 14 ER PT J AU Xu, M Yuan, L Wang, Y Chen, Sh Zhang, L Zhang, X AF Xu, Mingyue Yuan, Lijun Wang, Yan Chen, Shuo Zhang, Lin Zhang, Xipeng TI Integrative Analysis of DNA Methylation and Gene Expression Profiles Identifies Colorectal Cancer-Related Diagnostic Biomarkers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Colorectal cancer; DNA methylation; logistic regression model; CpG island methylator phenotype; The Cancer Genome Atlas; Gene Expression Omnibus; diagnosis ID Colorectal cancer; DNA methylation; logistic regression model; CpG island methylator phenotype; The Cancer Genome Atlas; Gene Expression Omnibus; diagnosis AB Background: Colorectal cancer (CRC) is a common human malignancy worldwide. The prognosis of patients is largely frustrated by delayed diagnosis or misdiagnosis. DNA methylation alterations have been previously proved to be involved in CRC carcinogenesis. Methods: In this study, we proposed to identify CRC-related diagnostic biomarkers by analyzing DNA methylation and gene expression profiles. TCGA-COAD datasets downloaded from the Cancer Genome Atlas (TCGA) were used as the training set to screen differential expression genes (DEGs) and methylation CpG sites (dmCpGs) in CRC samples. A logistic regression model was constructed based on hyper-methylated CpG sites which were located in downregulated genes for CRC diagnosis. Another two independent datasets from the Gene Expression Omnibus (GEO) were used as a testing set to evaluate the performance of the model in CRC diagnosis. Results: We found that CpG island methylator phenotype (CIMP) was a potential signature of poor prognosis by dividing CRC samples into CIMP and noCIMP groups based on a set of CpG sites with methylation standard deviation (sd) > 0.2 among CRC samples and low methylation levels (mean β < 0.05) in adjacent samples. Hypermethylated CpGs tended to be more closed to CpG island (CGI) and transcription start site (TSS) relative to hypo-methylated CpGs (p-value < 0.05, Fisher exact test). A logistic regression model was finally constructed based on two hyper-methylated CpGs, which had an area under receiver operating characteristic curve of 0.98 in the training set, and 0.85 and 0.95 in the two independent testing sets. Conclusions: In conclusion, our study identified promising DNA methylation biomarkers for CRC diagnosis. C1 [Xu, Mingyue] Tianjin Union Medical Center, Department of Colorectal SurgeryTianjin, China. [Yuan, Lijun] Tianjin Union Medical Center, Department of Colorectal SurgeryTianjin, China. [Wang, Yan] Shanghai Pudong New Area People’s Hospital, Department of Traditional Chinese MedicineShanghai, China. [Chen, Shuo] Tianjin Union Medical Center, Department of Colorectal SurgeryTianjin, China. [Zhang, Lin] Tianjin Union Medical Center, Department of Colorectal SurgeryTianjin, China. [Zhang, Xipeng] Tianjin Union Medical Center, Department of Colorectal SurgeryTianjin, China. RP Zhang, X (reprint author), Tianjin Union Medical Center, Department of Colorectal Surgery, Tianjin, China. EM zhangxipeng18212@outlook.com CR Bray C, Bell LN, Liang H, Collins D, and Yale SH. Colorectal Cancer Screening. WMJ, 2017, 116:27–33. Issa IA, and Noureddine M. Colorectal Cancer Screening: An Updated Review of the Available Options. Wjg, 2017, 23:5086–96., DOI 10.3748/ wjg.v23.i28.5086 Levin B, Lieberman DA, Mcfarland B, Smith RA, Brooks D, Andrews KS, et al. Screening and Surveillance for the Early Detection of Colorectal Cancer and Adenomatous Polyps, 2008: a Joint Guideline from the American Cancer Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of Radiology. CA: A Cancer J Clinicians, 2008, 58: 130–60., DOI 10.3322/ca.2007.0018 Vatandoost N, Ghanbari J, Mojaver M, Avan A., Ghayour-Mobarhan M., Nedaeinia R., et al. Early Detection of Colorectal Cancer: from Conventional Methods to Novel Biomarkers. J Cancer Res Clin Oncol, 2016, 142:3413–51., DOI 10.1007/s00432-015-1928-z Lao VV, and Grady WM. Epigenetics and Colorectal Cancer. Nat Rev Gastroenterol Hepatol, 2011, 8:686–700., DOI 10.1038/nrgastro.2011.173 Van Lanschot MCJ, Bosch LJW, DeWit M, Carvalho B, and Meijer GA. Early Detection: the Impact of Genomics. Virchows Arch, 2017, 471:165–73., DOI 10.1007/s00428-017-2159-2 Marmol I, Sanchez-de-diego C, Pradilla Dieste A, Dieste A. P., Cerrada E., Yoldi M. J. R., et al. Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer. Int J Mol Sci, 2017, 18., DOI 10.3390/ijms18010197 Weisenberger DJ, Liang G, and Lenz H-J. DNA Methylation Aberrancies Delineate Clinically Distinct Subsets of Colorectal Cancer and Provide Novel Targets for Epigenetic Therapies. Oncogene, 2018, 37:566–77., DOI 10.1038/ onc.2017.374 Wynter CVA, Walsh MD, and Higuchi T. Methylation Patterns Define Two Types of Hyperplastic Polyp Associated with Colorectal Cancer. Gut, 2004, 53: 573–80., DOI 10.1136/gut.2003.030841 Naumov VA, Generozov EV, Zaharjevskaya NB, Matushkina DS, Larin AK, Chernyshov SV, et al. Genome-scale Analysis of DNA Methylation in Colorectal Cancer Using Infinium HumanMethylation450 BeadChips. Epigenetics, 2013, 8:921–34., DOI 10.4161/epi.25577 Gundert M, Edelmann D, Benner A, Jansen L, Jia M, Walter V, et al. Genomewide DNA Methylation Analysis Reveals a Prognostic Classifier for Nonmetastatic Colorectal Cancer, ProMCol Classifier). Gut, 2019, 68:101–10., DOI 10.1136/gutjnl-2017-314711 Zhang F, Wang L, and Li Y. Optimizing Mesoderm Progenitor Selection and Three-Dimensional Microniche Culture Allows Highly Efficient Endothelial Differentiation and Ischemic Tissue Repair from Human Pluripotent Stem Cells. Stem Cel Res Ther, 2017, 8:6., DOI 10.1186/s13287-016-0455-4 Nazemalhosseini Mojarad E, Kuppen PJ, Aghdaei HA, and Zali MR. The CpG Island Methylator Phenotype, CIMP, in Colorectal Cancer. Gastroenterol Hepatol Bed Bench, 2013, 6:120–8. Robinson MD, Mccarthy DJ, and Smyth GK. edgeR: a Bioconductor Package for Differential Expression Analysis of Digital Gene Expression Data. Bioinformatics, 2010, 26:139–40., DOI 10.1093/bioinformatics/btp616 Robin X, Turck N, and Hainard A. pROC: an Open-Source Package for R and S+ to Analyze and Compare ROC Curves. BMC Bioinformatics, 2011, 12:77., DOI 10.1186/1471-2105-12-77 Sing T, Sander O, Beerenwinkel N, and Lengauer T. ROCR: Visualizing Classifier Performance in R. Bioinformatics, 2005, 21:3940–1., DOI 10.1093/ bioinformatics/bti623 You JS, and Jones PA. Cancer Genetics and Epigenetics: Two Sides of the Same coin? Cancer Cell, 2012, 22:9–20., DOI 10.1016/j.ccr.2012.06.008 Jones PA, Issa J-PJ, and Baylin S. Targeting the Cancer Epigenome for Therapy. Nat Rev Genet, 2016, 17:630–41., DOI 10.1038/nrg.2016.93 Okugawa Y, Grady WM, and Goel A. Epigenetic Alterations in Colorectal Cancer: Emerging Biomarkers. Gastroenterology, 2015, 149:1204–25., DOI 10.1053/j.gastro.2015.07.011 Kong X, Chen J, Xie W, Brown SM, Cai Y, Wu K, et al. Defining UHRF1 Domains that Support Maintenance of Human Colon Cancer DNA Methylation and Oncogenic Properties. Cancer Cell, 2019, 35:633–48., DOI 10.1016/j.ccell.2019.03.003 Sugimachi K, Matsumura T, Shimamura T, Hirata H, Uchi R, Ueda M, et al. Aberrant Methylation of FOXE1 Contributes to a Poor Prognosis for Patients with Colorectal Cancer. Ann Surg Oncol, 2016, 23:3948–55., DOI 10.1245/ s10434-016-5289-x Kandimalla R, Linnekamp JF, Van Hooff S, Castells A, Llor X, Andreu M, et al. Methylation of WNT Target Genes AXIN2 and DKK1 as Robust Biomarkers for Recurrence Prediction in Stage II colon Cancer. Oncogenesis, 2017, 6:e308., DOI 10.1038/oncsis.2017.9 Tse JWT, Jenkins LJ, Chionh F, and Mariadason JM. Aberrant DNA Methylation in Colorectal Cancer: What Should We Target? Trends Cancer, 2017, 3:698–712., DOI 10.1016/j.trecan.2017.08.003 Takatsu H, Hase K, Ohmae M, Ohshima S, Hashimoto K, Taniura N, et al. CD300 Antigen like Family Member G: A Novel Ig Receptor like Protein Exclusively Expressed on Capillary Endothelium. Biochem Biophysical Res Commun, 2006, 348:183–91., DOI 10.1016/j.bbrc.2006.07.047 Umemoto E, Takeda A, and Jin S. Dynamic Changes in Endothelial Cell Adhesion Molecule nepmucin/CD300LG Expression under Physiological and Pathological Conditions. PLoS One, 2013, 8:e83681., DOI 10.1371/ journal.pone.0083681 Miles FL, Pruitt FL, Van Golen KL, and Cooper CR. Stepping Out of the Flow: Capillary Extravasation in Cancer Metastasis. Clin Exp Metastasis, 2008, 25: 305–24., DOI 10.1007/s10585-007-9098-2 Taguchi T. Mannosyl, Alpha-1,3[6?]-)-Glycoprotein Beta-1,4-NAcetylglucosaminyltransferase, Isozyme C, Putative,, MGAT4C). In: Handbook of Glycosyltransferases and Related Genes, 2014, p. 257–63., DOI 10.1007/978-4-431-54240-7_134 Demichelis F, Setlur SR, Banerjee S, Chakravarty D, Chen JYH, Chen CX, et al. Identification of Functionally Active, Low Frequency Copy Number Variants at 15q21.3 and 12q21.31 Associated with Prostate Cancer Risk. Proc Natl Acad Sci, 2012, 109:6686–91., DOI 10.1073/pnas.1117405109 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609784 EP 1609790 DI 10.3389/pore.2021.1609784 PG 7 ER PT J AU Hua, Y Gao, L Li, X AF Hua, Yu Gao, Lihong Li, Xiaobo TI Comprehensive Analysis of Metabolic Genes in Breast Cancer Based on Multi-Omics Data SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; prognosis; mutation; metabolism; WGCNA ID breast cancer; prognosis; mutation; metabolism; WGCNA AB Background: Reprogramming of cell metabolism is one of the most important hallmarks of breast cancer. This study aimed to comprehensively analyze metabolic genes in the initiation, progression, and prognosis of breast cancer. Materials and Methods: Data from The Cancer Genome Atlas (TCGA) in breast cancer were downloaded including RNA-seq, copy number variation, mutation, and DNA methylation. A gene co-expression network was constructed by the weighted correlation network analysis (WGCNA) package in R. Association of metabolic genes with tumor-related immune cells and clinical parameters were also investigated. Results: We summarized 3,620 metabolic genes and observed mutations in 2,964 genes, of which the most frequently mutated were PIK3CA (51%), TNN (26%), and KMT2C (15%). Four genes (AKT1, ERBB2, KMT2C, and USP34) were associated with survival of breast cancer. Significant association was detected in the tumor mutation burden (TMB) of metabolic genes with T stage (p = 0.045) and N stage (p = 0.004). Copy number variations were significantly associated with recurrence and prognosis of breast cancer. The coexpression network for differentially expressed metabolic genes by WGCNA suggested that the modules were associated with glycerophospholipid, arachidonic acid, carbon, glycolysis/gluconeogenesis, and pyrimidine/purine metabolism. Glycerophospholipid metabolism correlated with most of the immune cells, while arachidonic acid metabolism demonstrated a significant correlation with endothelial cells. Methylation and miRNA jointly regulated 14 metabolic genes while mutation and methylation jointly regulated PIK3R1. Conclusion: Based on multi-omics data of somatic mutation, copy number variation, mRNA expression, miRNA expression, and DNA methylation, we identified a series of differentially expressed metabolic genes. Metabolic genes are associated with tumorrelated immune cells and clinical parameters, which might be therapy targets in future clinical application. C1 [Hua, Yu] The First Affiliated Hospital of China Medical University, Department of NursingShenyang, China. [Gao, Lihong] The First Affiliated Hospital of China Medical University, Department of NursingShenyang, China. [Li, Xiaobo] The First Affiliated Hospital of China Medical University, Department of NursingShenyang, China. RP Li, X (reprint author), The First Affiliated Hospital of China Medical University, Department of Nursing, Shenyang, China. EM lnshy110001@163.com CR Harbeck N, and Gnant M. Breast Cancer. The Lancet, 2017, 389(10074): 1134–50., DOI 10.1016/s0140-6736(16)31891-8 Fahad Ullah M. Breast Cancer: Current Perspectives on the Disease Status. Adv Exp Med Biol, 2019, 1152:51–64., DOI 10.1007/978-3-030-20301-6_4 Matsen CB, and Neumayer LA. Breast Cancer. JAMA Surg, 2013, 148(10): 971–9., DOI 10.1001/jamasurg.2013.3393 Yeo SK, and Guan J-L. Breast Cancer: Multiple Subtypes within a Tumor? Trends Cancer, 2017, 3(11):753–60., DOI 10.1016/j.trecan.2017.09.001 Colditz GA, and Bohlke K. Priorities for the Primary Prevention of Breast Cancer. CA A Cancer J Clinicians, 2014, 64(3):186–94., DOI 10.3322/caac.21225 Tray N, Taff J, and Adams S. Therapeutic Landscape of Metaplastic Breast Cancer. Cancer Treat Rev, 2019, 79:101888., DOI 10.1016/j.ctrv.2019.08.004 Tryfonidis K, Senkus E, Cardoso MJ, and Cardoso F. Management of Locally Advanced Breast Cancer-Perspectives and Future Directions. Nat Rev Clin Oncol, 2015, 12(3):147–62., DOI 10.1038/nrclinonc.2015.13 Sledge GW Jr. Curing Metastatic Breast Cancer. Jop, 2016, 12(1):6–10., DOI 10.1200/jop.2015.008953 Oh DY, and Bang YJ. HER2-targeted Therapies - a Role beyond Breast Cancer. Nat Rev Clin Oncol, 2020, 17(1):33–48., DOI 10.1038/s41571-019-0268-3 Kimbung S, Loman N, and Hedenfalk I. Clinical and Molecular Complexity of Breast Cancer Metastases. Semin Cancer Biol, 2015, 35:85–95., DOI 10.1016/ j.semcancer.2015.08.009 Raman D, Foo CHJ, Clement MV, and Pervaiz S. Breast Cancer: A Molecular and Redox Snapshot. Antioxid Redox Signaling, 2016, 25(6):337–70., DOI 10.1089/ars.2015.6546 Polyak K. Breast Cancer: Origins and Evolution. J Clin Invest, 2007, 117(11): 3155–63., DOI 10.1172/jci33295 Kulkoyluoglu-Cotul E, Arca A, and Madak-Erdogan Z. Crosstalk between Estrogen Signaling and Breast Cancer Metabolism. Trends Endocrinol Metab, 2019, 30(1):25–38., DOI 10.1016/j.tem.2018.10.006 Weigelt B, Geyer FC, and Reis-Filho JS. Histological Types of Breast Cancer: How Special Are They? Mol Oncol, 2010, 4(3):192–208., DOI 10.1016/ j.molonc.2010.04.004 Bianchini G, Balko JM, Mayer IA, Sanders ME, and Gianni L. Triple-negative Breast Cancer: Challenges and Opportunities of a Heterogeneous Disease. Nat Rev Clin Oncol, 2016, 13(11):674–90., DOI 10.1038/nrclinonc.2016.66 Pasculli B, Barbano R, and Parrella P. Epigenetics of Breast Cancer: Biology and Clinical Implication in the Era of Precision Medicine. Semin Cancer Biol, 2018, 51:22–35., DOI 10.1016/j.semcancer.2018.01.007 Dias AS, Almeida CR, Helguero LA, and Duarte IF. Metabolic Crosstalk in the Breast Cancer Microenvironment. Eur J Cancer, 2019, 121:154–71., DOI 10.1016/j.ejca.2019.09.002 Gandhi N, and Das GM. Metabolic Reprogramming in Breast Cancer and its Therapeutic Implications. Cells, 2019, 8(2): 89., DOI 10.3390/cells8020089 Wang T, Fahrmann JF, Lee H, Li Y-J, Tripathi SC, Yue C, et al. JAK/STAT3- Regulated Fatty Acid β-Oxidation Is Critical for Breast Cancer Stem Cell Self- Renewal and Chemoresistance. Cel Metab, 2018, 27(1):136–50., DOI 10.1016/ j.cmet.2017.11.001 Gao R, Li D, Xun J, Zhou W, Li J, Wang J, et al. CD44ICD Promotes Breast Cancer Stemness via PFKFB4-Mediated Glucose Metabolism. Theranostics, 2018, 8(22):6248–62., DOI 10.7150/thno.28721 Dasgupta S, Rajapakshe K, Zhu B, Nikolai BC, Yi P, Putluri N, et al. Metabolic Enzyme PFKFB4 Activates Transcriptional Coactivator SRC-3 to Drive Breast Cancer. Nature, 2018, 556(7700):249–54., DOI 10.1038/ s41586-018-0018-1 Chin K, DeVries S, Fridlyand J, Spellman PT, Roydasgupta R, Kuo WL, et al. Genomic and Transcriptional Aberrations Linked to Breast Cancer Pathophysiologies. Cancer Cell, 2006, 10(6):529–41., DOI 10.1016/ j.ccr.2006.10.009 Havas KM, Milchevskaya V, Radic K, Alladin A, Kafkia E, Garcia M, et al. Metabolic Shifts in Residual Breast Cancer Drive Tumor Recurrence. J Clin Invest, 2017, 127(6):2091–105., DOI 10.1172/jci89914 Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, Frederick AM, et al. Sequence Analysis of Mutations and Translocations across Breast Cancer Subtypes. Nature, 2012, 486(7403):405–9., DOI 10.1038/ nature11154 Wu W, Chen Y, Huang L, Li W, Tao C, and Shen H. Effects of AKT1 E17K Mutation Hotspots on the Biological Behavior of Breast Cancer Cells. Int J Clin Exp Pathol, 2020, 13(3):332–46. Ellis H, and Ma CX. PI3K Inhibitors in Breast Cancer Therapy. Curr Oncol Rep, 2019, 21(12):110., DOI 10.1007/s11912-019-0846-7 Gala K, Li Q, Sinha A, Razavi P, Dorso M, Sanchez-Vega F, et al. KMT2C Mediates the Estrogen Dependence of Breast Cancer through Regulation of ERα Enhancer Function. Oncogene, 2018, 37(34):4692–710., DOI 10.1038/ s41388-018-0273-5 Thomas A, Routh ED, Pullikuth A, Jin G, Su J, Chou JW, et al. TumorMutational burden Is a Determinant of Immune-Mediated Survival in Breast Cancer. Oncoimmunology, 2018, 7(10):e1490854., DOI 10.1080/2162402x.2018.1490854 Pereira B, Chin SF, Rueda OM, Vollan HK, Provenzano E, Bardwell HA, et al. The Somatic Mutation Profiles of 2,433 Breast Cancers Refines Their Genomic and Transcriptomic Landscapes. Nat Commun, 2016, 7:11479., DOI 10.1038/ncomms11479 Huang C, and Freter C. Lipid Metabolism, Apoptosis and Cancer Therapy. Ijms, 2015, 16(1):924–49., DOI 10.3390/ijms16010924 Sun Q, Zhao H, Liu Z, Wang F, He Q, Xiu C, et al. Identifying Potential Metabolic Tissue Biomarkers for Papillary Thyroid Cancer in Different Iodine Nutrient Regions. Endocrine, 2021)., DOI 10.1007/s12020-021-02773-3 Shi Y, Xu Y, Yao J, Yan C, Su H, Zhang X, et al. MTHFD2 Promotes Tumorigenesis and Metastasis in Lung Adenocarcinoma by Regulating AKT/GSK-3β/β-catenin Signalling. J Cel Mol Med, 2021, 25(14): 7013–7027. Li H, Zhang C, Zhang H, and Li H. Xanthine Oxidoreductase Promotes the Progression of Colitis-Associated Colorectal Cancer by Causing DNA Damage and Mediating Macrophage M1 Polarization. Eur J Pharmacol, 2021, 906: 174270., DOI 10.1016/j.ejphar.2021.174270 Peng F, Wang JH, Fan WJ, Meng YT, Li MM, Li TT, et al. Glycolysis Gatekeeper PDK1 Reprograms Breast Cancer Stem Cells under Hypoxia. Oncogene, 2018, 37(8):1062–74., DOI 10.1038/onc.2017.368 Ermert L, Dierkes C, and Ermert M. Immunohistochemical Expression of Cyclooxygenase Isoenzymes and Downstream Enzymes in Human Lung Tumors. Clin Cancer Res, 2003, 9(5):1604–10. Sonkar K, Ayyappan V, Tressler CM, Adelaja O, Cai R, Cheng M, et al. Focus on the Glycerophosphocholine Pathway in Choline Phospholipid Metabolism of Cancer. NMR Biomed, 2019, 32(10):e4112., DOI 10.1002/nbm.4112 Gao X, Lee K, Reid MA, Sanderson SM, Qiu C, Li S, et al. Serine Availability Influences Mitochondrial Dynamics and Function through Lipid Metabolism. Cel Rep, 2018, 22(13):3507–20., DOI 10.1016/j.celrep.2018.03.017 Ren S, Liu J, Feng Y, Li Z, He L, Li L, et al. Knockdown of circDENND4C Inhibits Glycolysis, Migration and Invasion by Up-Regulating miR-200b/c in Breast Cancer under Hypoxia. J Exp Clin Cancer Res, 2019, 38(1):388., DOI 10.1186/s13046-019-1398-2 Chen L, Yang L, Yao L, Kuang XY, Zuo WJ, Li S, et al. Characterization of PIK3CA and PIK3R1 Somatic Mutations in Chinese Breast Cancer Patients. Nat Commun, 2018, 9(1):1357., DOI 10.1038/s41467-018-03867-9 Kim S, Lee E, Jung J, Lee JW, Kim HJ, Kim J, et al. microRNA-155 Positively Regulates Glucose Metabolism via PIK3R1-FOXO3a-cMYC axis in Breast Cancer. Oncogene, 2018, 37(22):2982–91., DOI 10.1038/s41388-018-0124-4 Rahman MM, Brane AC, and Tollefsbol TO. MicroRNAs and Epigenetics Strategies to Reverse Breast Cancer. Cells, 2019, 8(10): 1214., DOI 10.3390/ cells8101214 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609789 EP 1609798 DI 10.3389/pore.2021.1609789 PG 10 ER PT J AU Zhu, L Ling, Ch Xu, T Zhang, J Zhang, Y Liu, Y Fang, Ch Yang, L Zhuang, W Wang, R Ping, J Wang, M AF Zhu, Lili Ling, Chunrun Xu, Tao Zhang, Jinglin Zhang, Yujie Liu, Yingjie Fang, Chao Yang, Lie Zhuang, Wen Wang, Rui Ping, Jie Wang, Mojin TI Clinicopathological Features and Survival of Signet-Ring Cell Carcinoma and Mucinous Adenocarcinoma of Right Colon, Left Colon, and Rectum SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE survival; colorectal carcinoma; signet-ring cell carcinoma; histological subtype; SEER ID survival; colorectal carcinoma; signet-ring cell carcinoma; histological subtype; SEER AB Histological subtype plays an important role in the different clinical characteristics and survival outcomes of patients with colorectal carcinoma (CRC). However, in previous studies, the influences of tumor locations and tumor stages have not been strictly controlled. This study focused on the assessment of the prognostic value of each histological subtype in different tumor locations and tumor stages of CRC. We used the Surveillance, Epidemiology, and End Results (SEER) database (1973–2011) to analyze 818,229 CRC patients with different clinical and pathological features, and analyzed the prognostic value of each histological subtype. Under the condition of stratification by tumor stage, signet-ring cell carcinoma (SRCC) presented the worst survival in each stage of right colon cancer (stage I, log-rank, p = 0.002, stages II, III, and IV, log-rank, p < 0.001), rectal cancer (RC) (log-rank, p < 0.001), and in stages II, III, and IV of left colon cancer (log-rank, p < 0.001). Multivariate survival analysis suggested SRCC subtype, male gender, age ≥ 70 years, tumor size ≥ 5 cm, stage progression, and poor differentiation were all significant factors worsening survival in CRC (p < 0.001, respectively). Mucinous adenocarcinoma (MC) histological subtype proved to be an independent protective factor for the prognosis of right colon cancer (p = 0.003). Overall, in our study, the results suggested SRCC had the worst survival among the three histological subtypes of CRC. MC was associated with favorable prognosis in right colon cancer but not with other tumor locations. C1 [Zhu, Lili] Sichuan University, West China Hospital, Department of Gastrointestinal SurgeryChengdu, China. [Ling, Chunrun] The People’s Hospital of Guangxi Zhuang Autonomous Region, Department of General and Pediatric SurgeryNanning, China. [Xu, Tao] Sichuan University, West China Hospital, Department of Gastrointestinal SurgeryChengdu, China. [Zhang, Jinglin] Sichuan University, West China Hospital, Department of Gastrointestinal SurgeryChengdu, China. [Zhang, Yujie] Sichuan University, West China Hospital, Department of Gastrointestinal SurgeryChengdu, China. [Liu, Yingjie] Sichuan University, West China Hospital, Department of Gastrointestinal SurgeryChengdu, China. [Fang, Chao] Sichuan University, West China Hospital, Department of Gastrointestinal SurgeryChengdu, China. [Yang, Lie] Sichuan University, West China Hospital, Department of Gastrointestinal SurgeryChengdu, China. [Zhuang, Wen] Sichuan University, West China Hospital, Department of Gastrointestinal SurgeryChengdu, China. [Wang, Rui] Sichuan University, West China Hospital, Department of GastroenterologyChengdu, China. [Ping, Jie] Vanderbilt University Medical Center, Division of EpidemiologyNashville, TN, USA. [Wang, Mojin] Sichuan University, West China Hospital, Department of Gastrointestinal SurgeryChengdu, China. RP Wang, M (reprint author), Sichuan University, West China Hospital, Department of Gastrointestinal Surgery, Chengdu, China. EM wangmojin2001@163.com CR Song IH, Hong S-M, Yu E, Yoon YS, Park IJ, Lim S-B, et al. Signet Ring Cell Component Predicts Aggressive Behaviour in Colorectal Mucinous Adenocarcinoma. Pathology, 2019, 51(4):384–91., DOI 10.1016/ j.pathol.2019.03.001 Nitsche U, Friess H, Agha A, Angele M, Eckel R, HeitlandW, et al. Prognosis of Mucinous and Signet-Ring Cell Colorectal Cancer in a Population-Based Cohort. J Cancer Res Clin Oncol, 2016, 142(11):2357–66., DOI 10.1007/ s00432-016-2224-2 Du W, Mah JTL, Lee J, Sankila R, Sankaranarayanan R, and Chia K-S. Incidence and Survival of Mucinous Adenocarcinoma of the Colorectum: a Population-Based Study from an Asian Country. Dis colon rectum, 2004, 47(1):78–85., DOI 10.1007/s10350-003-0014-9 Gao P, Song Y-x., Xu Y-y., Sun Z, Sun J-x., Xu H-m., et al. Does the Prognosis of Colorectal Mucinous Carcinoma Depend upon the Primary Tumour Site? Results from Two Independent Databases. Histopathology, 2013, 63(5): 603–15., DOI 10.1111/his.12190 Green JB, Timmcke AE, Mitchell WT, Hicks TC, Gathright BJ, Jr, and Ray JE. Mucinous Carcinoma-Just Another colon Cancer? Dis colon rectum, 1993, 36(1):49–54., DOI 10.1007/bf02050301 Hugen N, Verhoeven RHA, Radema SA, de Hingh IHJT, Pruijt JFM, Nagtegaal ID, et al. Prognosis and Value of Adjuvant Chemotherapy in Stage III Mucinous Colorectal Carcinoma. Ann Oncol, 2013, 24(11):2819–24., DOI 10.1093/annonc/mdt378 Wu C-S, Tung S-Y, Chen P-C, and Kuo Y-C. Clinicopathological Study of Colorectal Mucinous Carcinoma in Taiwan: A Multivariate Analysis. J Gastroenterol Hepatol, 1996, 11(1):77–81., DOI 10.1111/j.1440- 1746.1996.tb00014.x Kang H, O’Connell JB, Maggard MA, Sack J, and Ko CY. A 10-year Outcomes Evaluation of Mucinous and Signet-Ring Cell Carcinoma of the colon and Rectum. Dis colon rectum, 2005, 48(6):1161–8., DOI 10.1007/s10350-004- 32-1 9. Hosseini S, Bananzadeh AM, Salek R, Zare-Bandamiri M, Kermani AT, and Mohammadianpanah M. Prognostic Significance of Mucinous Histologic Subtype on Oncologic Outcomes in Patients with Colorectal Cancer. Ann Coloproctol, 2017, 33(2):57–63., DOI 10.3393/ac.2017.33.2.57 Bittorf B, Merkel S, Matzel K, Wein A, Dimmler A, and Hohenberger W. Primary Signet-Ring Cell Carcinoma of the Colorectum. Langenbecks Arch Surg, 2004, 389(3):178–83., DOI 10.1007/s00423-004-0474-y Borger M, Gosens M, Jeuken J, van Kempen L, van de Velde C, van Krieken J, et al. Signet Ring Cell Differentiation in Mucinous Colorectal Carcinoma. J Pathol, 2007, 212(3):278–86., DOI 10.1002/path.2181 Hugen N, Verhoeven RH, Lemmens VE, van Aart CJ, Elferink MA, Radema SA, et al. Colorectal Signet-Ring Cell Carcinoma: Benefit from Adjuvant Chemotherapy but a Poor Prognostic Factor. Int J Cancer, 2015, 136(2): 333–9., DOI 10.1002/ijc.28981 Lee GH, Malietzis G, Askari A, Bernardo D, Al-Hassi HO, and Clark SK. Is Right-Sided colon Cancer Different to Left-Sided Colorectal Cancer? - A Systematic Review. Eur J Surg Oncol, Ejso,, 2015, 41(3):300–8., DOI 10.1016/ j.ejso.2014.11.001 Razenberg LGEM, van Gestel YRBM, Lemmens VEPP, de Wilt JHW, Creemers G-J, and de Hingh IHJT. The Prognostic Relevance of Histological Subtype in Patients with Peritoneal Metastases from Colorectal Cancer: A Nationwide Population-Based Study. Clin Colorectal Cancer, 2015, 14(4):e13–e19., DOI 10.1016/j.clcc.2015.05.011 Shen H, Yang J, Huang Q, Jiang MJ, Tan YN, Fu JF, et al. Different Treatment Strategies and Molecular Features between Right-Sided and Left-Sided colon Cancers. Wjg, 2015, 21(21):6470–8., DOI 10.3748/ wjg.v21.i21.6470 Gao XH, Yu GY, Gong HF, Liu LJ, Xu Y, Hao LQ, et al. Differences of Protein Expression Profiles, KRAS and BRAF Mutation, and Prognosis in Right-Sided colon, Left-Sided colon and Rectal Cancer. Sci Rep, 2017, 7(1):7882., DOI 10.1038/s41598-017-08413-z He W-Z, Liao F-X, Jiang C, Kong P-F, Yin C-X, Yang Q, et al. Primary Tumor Location as a Predictive Factor for First-Line Bevacizumab Effectiveness in Metastatic Colorectal Cancer Patients. J Cancer, 2017, 8(3):388–94., DOI 10.7150/jca.16804 Bademci R, Bollo J, Martinez MC, Hernadez MP, and Targarona EM. Colorectal Cancer Prognosis: The Impact of Signet Ring Cell. Gastrointest Tumors, 2019, 6(3-4):57–63., DOI 10.1159/000501454 Hyngstrom JR, Hu C-Y, Xing Y, You YN, Feig BW, Skibber JM, et al. Clinicopathology and Outcomes for Mucinous and Signet Ring Colorectal Adenocarcinoma: Analysis from the National Cancer Data Base. Ann Surg Oncol, 2012, 19(9):2814–21., DOI 10.1245/s10434-012-2321-7 Dajani YF, Zayid I, Malatjalian DA, and Kamal MF. Colorectal Cancer in Jordan and Nova Scotia: a Comparative Epidemiologic and Histopathologic Study. Cancer, 1980, 46(2):420–8., DOI 10.1002/1097-0142(19800715)46:2 Vallam KC, Desouza A, Bal M, Patil P, Engineer R, and Saklani A. Adenocarcinoma of the Rectum-A Composite of Three Different Subtypes with Varying Outcomes? Clin Colorectal Cancer, 2016, 15(2):e47–e52., DOI 10.1016/j.clcc.2015.12.004 Ling C-R, Wang R, Wang M-J, Ping J, and ZhuangW. Prognosis and Value of Preoperative Radiotherapy in Locally Advanced Rectal Signet-Ring Cell Carcinoma. Sci Rep, 2017, 7:45334., DOI 10.1038/srep45334 Chew M-H, Yeo S-AE, Ng Z-P, Lim K-H, Koh P-K, Ng K-H, et al. Critical Analysis of Mucin and Signet Ring Cell as Prognostic Factors in an Asian Population of 2,764 Sporadic Colorectal Cancers. Int J Colorectal Dis, 2010, 25(10):1221–9., DOI 10.1007/s00384-010-1033-3 Thota R, Fang X, and Subbiah S. Clinicopathological Features and Survival Outcomes of Primary Signet Ring Cell and Mucinous Adenocarcinoma of colon: Retrospective Analysis of VACCR Database. J Gastrointest Oncol, 2014, 5(1):18–24., DOI 10.3978/j.issn.2078-6891.2013.051 Lee H-S, Soh JS, Lee S, Bae JH, Kim K-J, Ye BD, et al. Clinical Features and Prognosis of Resectable Primary Colorectal Signet-Ring Cell Carcinoma. Intest Res, 2015, 13(4):332–8., DOI 10.5217/ir.2015.13.4.332 Bermejo JL, Eng C, and Hemminki K. Cancer Characteristics in Swedish Families Fulfilling Criteria for Hereditary Nonpolyposis Colorectal Cancer. Gastroenterology, 2005, 129(6):1889–99., DOI 10.1053/j.gastro.2005.09.012 Bertagnolli MM, Niedzwiecki D, Compton CC, Hahn HP, Hall M, Damas B, et al. Microsatellite Instability Predicts Improved Response to Adjuvant Therapy with Irinotecan, Fluorouracil, and Leucovorin in Stage III colon Cancer: Cancer and Leukemia Group B Protocol 89803. Jco, 2009, 27(11): 1814–21., DOI 10.1200/jco.2008.18.2071 Des Guetz G, Lecaille C, Mariani P, Bennamoun M, Uzzan B, Nicolas P, et al. Prognostic Impact of Microsatellite Instability in Colorectal Cancer Patients Treated with Adjuvant FOLFOX. Anticancer Res, 2010, 30(10):4297–301. Kloor M, Staffa L, Ahadova A, and von Knebel Doeberitz M. Clinical Significance of Microsatellite Instability in Colorectal Cancer. Langenbecks Arch Surg, 2014, 399(1):23–31., DOI 10.1007/s00423-013- 1112-3 Lee C-T, Huang Y-C, Hung L-Y, Chow N-H, Su P-F, Ho C-L, et al. Serrated Adenocarcinoma Morphology in Colorectal Mucinous Adenocarcinoma is Associated with Improved Patient Survival. Oncotarget, 2017, 8(21):35165–75., DOI 10.18632/oncotarget.16815 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609800 EP 1609808 DI 10.3389/pore.2021.1609800 PG 9 ER PT J AU Kelany, M Barth, Th Salem, D Shakweer, M AF Kelany, Mohamed Barth, FE. Thomas Salem, Dina Shakweer, M. Marwa TI Prevalence and Prognostic Implications of PD-L1 Expression in Soft Tissue Sarcomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; prognosis; PD-L1 expression; soft tissue sarcomas; prevalence ID biomarker; prognosis; PD-L1 expression; soft tissue sarcomas; prevalence AB Background: PD-L1 expression differs from 19 to 92% in various cancer subtypes. Its expression carries a worse prognostic value in various malignancies and could also be used as a predictive marker for immune checkpoint inhibitor response. This study aimed to explore the prevalence of PD-L1 expression in soft tissue sarcomas and the correlation of PD-L1 expression with clinicopathological features. Patients and Methods: The tissue samples of 50 patients with STS were tested for PDL1 expression using immunohistochemistry (IHC). We followed a 6-step proportional scoring system. The patients were treated at Ain Shams University Hospital from 2011 to 2017. We also explored the correlation of PD-L1 expression with different clinical features of the patients. The chi-square test was used to calculate the differences among variables. Results: Twelve cases (24%) showed positive PD-L1 expression with the highest prevalence in rhabdomyosarcoma and desmoid tumors (2/2 and 2/3 cases, respectively), followed by GIST in 2/4 cases and liposarcoma in 3/11 cases. Patients with positive PD-L1 expression showed a trend for worse survival, with a median overall survival of 11 months vs. 19 months for patients with negative PD-L1 expression (p-value = 0.1) and a mean PFS of 6 months vs. 11 months for patients with negative PD-L1 expression (p-value = 0.1). However, these findings did not reach statistical significance. Conclusion: Although the results did not reach statistical significance due to the small number of cases, PD-L1 expression could represent a prognostic factor for poor outcome. Larger clinical trials are recommended for the validation of PD-L1 as a poor prognostic biomarker. C1 [Kelany, Mohamed] Ain Shams University, Department of Clinical OncologyCairo, Egypt. [Barth, FE. Thomas] Ulm University, Department of PathologyUlm, Germany. [Salem, Dina] Ain Shams University, Department of Clinical OncologyCairo, Egypt. [Shakweer, M. Marwa] Ain Shams Faculty of Medicine, Department of PathologyCairo, Egypt. RP Kelany, M (reprint author), Ain Shams University, Department of Clinical Oncology, Cairo, Egypt. CR Burningham Z, Hashibe M, Spector L, and Schiffman JD. The Epidemiology of Sarcoma. Clin Sarcoma Res, 2012, 2(1):14., DOI 10.1186/2045-3329-2-14 SEER*Stat Databases: November 2017 Submission [Internet]. [cited 2021 Feb 15]. Available from: https://seer.cancer.gov/data-software/documentation/ seerstat/nov2017/. Chen DS, and Mellman I. Oncology Meets Immunology: The Cancer- Immunity Cycle. Immunity, 2013, 39(1):1–10., DOI 10.1016/ j.immuni.2013.07.012 Martin V, Raica M, and Cimpean AM. First-Line Immunophenotyping in the Pathologic Diagnosis of Soft Tissue Tumors. 2004;54(2):122–7. Kim C, Kim EK, Jung H, Chon HJ, Han JW, Shin K-H, et al. Prognostic Implications of PD-L1 Expression in Patients with Soft Tissue Sarcoma. BMC Cancer, 2016, 16(1):434., DOI 10.1186/s12885-016-2451-6 Kim C, Kim EK, Han JW, Chon HJ, Heo SJ, Lee YH, et al. Clinical Pattern and Implication of PD-L1 Expression in Soft-Tissue Sarcoma. Jco, 2015, 33:10565., DOI 10.1200/jco.2015.33.15_suppl.10565 D’Angelo SP, Shoushtari AN, Agaram NP, Kuk LX, Qin L-X, Dickson MA, et al. Prevalence of Tumor-Infiltrating Lymphocytes and PD-L1 Expression in the Soft Tissue SarcomaMicroenvironment. Hum Pathol, 2015, 46(3):357–65., DOI 10.1016/j.humpath.2014.11.001 Jo VY, and Fletcher CDM. WHO Classification of Soft Tissue Tumours: an Update Based on the 2013, 4th, Edition. Pathology, 2014, 46(2):95–104., DOI 10.1097/PAT.0000000000000050 Herbst RS, Baas P, Kim D-W, Felip E, Perez-Gracia JL, Han J-Y, et al. Pembrolizumab versus Docetaxel for Previously Treated, PD-L1-Positive, Advanced Non-small-cell Lung Cancer, KEYNOTE-010): a Randomised Controlled Trial. Lancet, 2016, 387:1540–50., DOI 10.1016/S0140-6736(15, 01281-7 Scheel AH, Dietel M, Heukamp LC, Johrens K, Kirchner T, Reu S, et al. Harmonized PD-L1 Immunohistochemistry for Pulmonary Squamous-Cell and Adenocarcinomas. Mod Pathol, 2016, 29(10):1165–72., DOI 10.1038/ modpathol.2016.117 Boxberg M, Steiger K, Lenze U, Rechl H, von Eisenhart-Rothe R, Wortler K, et al. PD-L1 and PD-1 and Characterization of Tumor-Infiltrating Lymphocytes in High Grade Sarcomas of Soft Tissue – Prognostic Implications and Rationale for Immunotherapy. OncoImmunology, 2018, 7(3)., DOI 10.1080/2162402x.2017.1389366 Kim J, Moon YJ, Kwon KS, Bae JS, Wagle S, Kim KM, et al. Tumor Infiltrating PD1-Positive Lymphocytes and the Expression of PD-L1 Predict Poor Prognosis of Soft Tissue Sarcomas. PLoS ONE, 2013, 8(12):1–9., DOI 10.1371/journal.pone.0082870 Chowdhury F, Dunn S, Mitchell S, Mellows T, Ashton-Key M, and Gray JC. PD-L1 and CD8 + PD1 + Lymphocytes Exist as Targets in the Pediatric Tumor Microenvironment for Immunomodulatory Therapy. OncoImmunology, 2015, 4(10)., DOI 10.1080/2162402x.2015.1029701 Que Y, Xiao W, Guan Y, Liang Y, Yan S, Chen H, et al. PD-L1 Expression Is Associated with FOXP3 + Regulatory T-Cell Infiltration of Soft Tissue Sarcoma and Poor Patient Prognosis. J Cancer 2018;8:2018–25., DOI 10.7150/jca.18683 Patel KR, Martinez A, Stahl JM, Logan SJ, Perricone AJ, Ferris MJ, et al. Increase in PD-L1 Expression after Pre-operative Radiotherapy for Soft Tissue Sarcoma. OncoImmunology, 2018, 7(7)., DOI 10.1080/ 2162402x.2018.1442168 Pollack SM, He Q, Yearley JH, Emerson R, Vignali M, Zhang Y, et al. T-cell Infiltration and Clonality Correlate with Programmed Cell Death Protein 1 and Programmed Death-Ligand 1 Expression in Patients with Soft Tissue Sarcomas. Cancer, 2017, 123(17)., DOI 10.1002/cncr.30726 van Erp AEM, Versleijen-Jonkers YMH, Hillebrandt-Roeffen MHS, van Houdt L, Gorris MAJ, van Dam LS, et al. Expression and Clinical Association of Programmed Cell Death-1, Programmed Death-Ligand-1 and CD8+ Lymphocytes in Primary Sarcomas Is Subtype Dependent. Oncotarget, 2017, 8(41)., DOI 10.18632/oncotarget.19071 Paydas S, Bagir EK, Deveci MA, and Gonlusen G. Clinical and Prognostic Significance of PD-1 and PD-L1 Expression in Sarcomas. Med Oncol, 2016, 33(8)., DOI 10.1007/s12032-016-0807-z Bertucci F, Finetti P, Perrot D, Leroux A, Collin F, le Cesne A, et al. PDL1 Expression Is a Poor-Prognosis Factor in Soft-Tissue Sarcomas. OncoImmunology, 2017, 6(3)., DOI 10.1080/2162402x.2016.1278100 Budczies J, Mechtersheimer G, Denkert C, Klauschen F, Mughal SS, Chudasama P, et al. PD-L1, CD274, Copy Number Gain, Expression, and Immune Cell Infiltration as Candidate Predictors for Response to Immune Checkpoint Inhibitors in Soft-Tissue Sarcoma. OncoImmunology, 2017, 6(3)., DOI 10.1080/2162402x.2017.1279777 Thompson RH, Dong H, Lohse CM, Leibovich BC, Blute ML, Cheville JC, et al. PD-1 Is Expressed by Tumor-Infiltrating Immune Cells and Is Associated with Poor Outcome for Patients with Renal Cell Carcinoma. Clin Cancer Res, 2007, 13:1757–61., DOI 10.1158/1078-0432.CCR-06-2599 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609804 EP 1609809 DI 10.3389/pore.2021.1609804 PG 6 ER PT J AU Tian, X Xu, WH Wu, JL Gan, HL Wang, HK Gu, WJ Qu, YY Zhang, HL Ye, DW AF Tian, Xi Xu, Wen-Hao Wu, Jun-Long Gan, Hua-Lei Wang, Hong-Kai Gu, Wei-Jie Qu, Yuan-Yuan Zhang, Hai-Liang Ye, Ding-Wei TI Clear Cell Papillary Renal Cell Carcinoma Shares Distinct Molecular Characteristics and may be Significantly Associated With Higher Risk of Developing Second Primary Malignancy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE somatic mutation; clear cell papillary renal cell carcinoma; germline mutation; fanconi anemia pathway; second primary malignancy ID somatic mutation; clear cell papillary renal cell carcinoma; germline mutation; fanconi anemia pathway; second primary malignancy AB Traditionally, clear cell papillary renal cell carcinoma (ccpRCC) was considered to share similar molecular and histological characteristics with clear cell renal cell carcinoma (ccRCC) and papillary renal cell carcinoma (pRCC). Here we aimed to identify somatic and germline variants of ccpRCC. For this purpose, we conducted whole-exome sequencing to detect somatic variants in the tissues of 18 patients with pathologically confirmed ccpRCC, who underwent surgical treatment at Fudan University Shanghai Cancer Center. Targeted sequencing was conducted to detect germline variants in paired tumor or normal tissues or blood. Somatic and germline variants of ccRCC and Renal cell carcinoma included in The Cancer Genome Atlas data and other published data were analyzed as well. The molecular profiles of ccpRCC, ccRCC and pRCC were compared. Among the 387 somatic variants identified, TCEB1 (3/18) and VHL (3/18) variants occurred at the highest frequencies. Germline mutation detection showed that nine variants associated with Fanconi anemia (VAFAs) pathway (FANCA, 6/18; FANCI, 3/18) were identified in 18 ccpRCC patients. Among ccpRCC patients with VAFAs, five out of eight patients had second primary malignancy or family history of cancer. Somatic variants characteristics may distinguish ccpRCC from ccRCC or pRCC and germline VAFAs may be a molecular characterization of ccpRCC. Compared with ccRCC or pRCC, ccpRCC patients may be significantly correlated with higher risk of developing second primary malignancy. C1 [Tian, Xi] Fudan University, Shanghai Cancer Center, Department of UrologyShanghai, China. [Xu, Wen-Hao] Fudan University, Shanghai Cancer Center, Department of UrologyShanghai, China. [Wu, Jun-Long] Fudan University, Shanghai Cancer Center, Department of UrologyShanghai, China. [Gan, Hua-Lei] Fudan University, Shanghai Medical College, Department of OncologyShanghai, China. [Wang, Hong-Kai] Fudan University, Shanghai Cancer Center, Department of UrologyShanghai, China. [Gu, Wei-Jie] Fudan University, Shanghai Cancer Center, Department of UrologyShanghai, China. [Qu, Yuan-Yuan] Fudan University, Shanghai Cancer Center, Department of UrologyShanghai, China. [Zhang, Hai-Liang] Fudan University, Shanghai Cancer Center, Department of UrologyShanghai, China. [Ye, Ding-Wei] Fudan University, Shanghai Cancer Center, Department of UrologyShanghai, China. RP Ye, DW (reprint author), Fudan University, Shanghai Cancer Center, Department of Urology, Shanghai, China. EM dwyelie@163.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Truong LD, and Shen SS. Immunohistochemical Diagnosis of Renal Neoplasms. Arch Pathol Lab Med, 2011, 135(1):92–109., DOI 10.1043/2010- 0478-RAR.1 Morlote DM, Harada S, Batista D, Gordetsky J, and Rais-Bahrami S. Clear Cell Papillary Renal Cell Carcinoma: Molecular Profile and Virtual Karyotype. Hum Pathol, 2019, 91:52–60., DOI 10.1016/ j.humpath.2019.05.011 Gobbo S, Eble JN, Grignon DJ, Martignoni G, MacLennan GT, Shah RB, et al. Clear Cell Papillary Renal Cell Carcinoma. Am J Surg Pathol, 2008, 32(8): 1239–45., DOI 10.1097/pas.0b013e318164bcbb Aydin H, Chen L, Cheng L, Vaziri S, He H, Ganapathi R, et al. Clear Cell Tubulopapillary Renal Cell Carcinoma: a Study of 36 Distinctive Low-Grade Epithelial Tumors of the Kidney. Am J Surg Pathol, 2010, 34(11):1608–21., DOI 10.1097/pas.0b013e3181f2ee0b Alshenawy HA. Immunohistochemical Panel for Differentiating Renal Cell Carcinoma with Clear and Papillary Features. Pathol Oncol Res, 2015, 21(4): 893–9., DOI 10.1007/s12253-015-9898-7 Rohan SM, Xiao Y, Liang Y, Dudas ME, Al-Ahmadie HA, Fine SW, et al. Clear-Cell Papillary Renal Cell Carcinoma: Molecular and Immunohistochemical Analysis with Emphasis on the Von Hippel-Lindau Gene and Hypoxia-Inducible Factor Pathway-Related Proteins. Mod Pathol, 2011, 24(9):1207–20., DOI 10.1038/modpathol.2011.80 Williamson SR. What Is the Malignant Potential of clear Cell Papillary Renal Cell Carcinoma?. Urol Oncol Semin Original Invest, 2016, 34(9):420–1., DOI 10.1016/j.urolonc.2016.05.035 Diolombi ML, Cheng L, Argani P, and Epstein JI. Do Clear Cell Papillary Renal Cell Carcinomas Have Malignant Potential? Am J Surg Pathol, 2015, 39(12): 1621–34., DOI 10.1097/pas.0000000000000513 Gupta S, Inwards CY, Van Dyke DL, Jimenez RE, and Cheville JC. Defining Clear Cell Papillary Renal Cell Carcinoma in Routine Clinical Practice. Histopathology, 2020, 76(7):1093–5., DOI 10.1111/his.14071 Stratton MR, Campbell PJ, and Futreal PA. The Cancer Genome. Nature, 2009, 458(7239):719–24., DOI 10.1038/nature07943 Lawrie CH, Larrea E, Larrinaga G, Goicoechea I, Arestin M, Fernandez- Mercado M, et al. Targeted Next-Generation Sequencing and Non-Coding RNA Expression Analysis of clear Cell Papillary Renal Cell Carcinoma Suggests Distinct Pathological Mechanisms from Other Renal Tumour Subtypes. J Pathol, 2014, 232(1):32–42., DOI 10.1002/path.4296 Mose LE, Wilkerson MD, Hayes DN, Perou CM, and Parker JS. ABRA: Improved Coding Indel Detection via Assembly-Based Realignment. Bioinformatics, Oxford, England,, 2014, 30(19):2813–5., DOI 10.1093/ bioinformatics/btu376 Huang KL, Mashl RJ, Wu Y, Ritter DI, Wang J, Oh C, et al. Pathogenic Germline Variants in 10,389 Adult Cancers. Cell, 2018, 173(2):355–e14., DOI 10.1016/j.cell.2018.03.039 Gu Z, Eils R, and Schlesner M. Complex Heatmaps Reveal Patterns and Correlations in Multidimensional Genomic Data. Bioinformatics, 2016, 32(18):2847–9., DOI 10.1093/bioinformatics/btw313 The Cancer Genome Atlas Research Network. Comprehensive Molecular Characterization of Clear Cell Renal Cell Carcinoma. Nature, 2013, 499(7456):43–9., DOI 10.1038/nature12222 Linehan WM, Spellman PT, Ricketts CJ, Creighton CJ, Fei SS, Davis C, et al. Comprehensive Molecular Characterization of Papillary Renal-Cell Carcinoma. N Engl J Med, 2016, 374(2):135–45., DOI 10.1056/nejmoa1505917 Clague J, Lin J, Cassidy A, Matin S, Tannir NM, Tamboli P, et al. Family History and Risk of Renal Cell Carcinoma: Results from a Case-Control Study and Systematic Meta-Analysis. Cancer Epidemiol Biomarkers Prev, 2009, 18(3):801–7., DOI 10.1158/1055-9965.epi-08-0601 Chakraborty S, Tarantolo SR, Batra SK, and Hauke RJ. Incidence and Prognostic Significance of Second Primary Cancers in Renal Cell Carcinoma. Am J Clin Oncol, 2013, 36(2):132–42., DOI 10.1097/ coc.0b013e3182438ddf Joung JY, Kwon W-A, Lim J, Oh C-M, Jung K-W, Kim SH, et al. Second Primary Cancer Risk Among Kidney Cancer Patients in Korea: A Population- Based Cohort Study. Cancer Res Treat, 2018, 50(1):293–301., DOI 10.4143/ crt.2016.543 Rabbani F, Reuter VE, Katz J, and Russo P. Second Primary Malignancies Associated with Renal Cell Carcinoma: Influence of Histologic Type. Urology, 2000, 56(3):399–403., DOI 10.1016/s0090-4295(00)00682-8 Wolfe A, Dobin SM, Grossmann P, Michal M, and Donner LR. Clonal Trisomies 7,10 and 12, normal 3p and Absence of VHL Gene Mutation in a Clear Cell Tubulopapillary Carcinoma of the Kidney. Virchows Arch, 2011, 459(4):457–63., DOI 10.1007/s00428-011-1137-3 Favazza L, Chitale DA, Barod R, Rogers CG, Kalyana-Sundaram S, Palanisamy N, et al. Renal Cell Tumors with Clear Cell Histology and Intact VHL and Chromosome 3p: A Histological Review of Tumors from the Cancer Genome Atlas Database. Mod Pathol, 2017, 30(11):1603–12., DOI 10.1038/ modpathol.2017.72 Hes O, Comperat EM, and Rioux-Leclercq N. Clear Cell Papillary Renal Cell Carcinoma, Renal Angiomyoadenomatous Tumor, and Renal Cell Carcinoma with Leiomyomatous Stroma Relationship of 3 Types of Renal Tumors: A Review. Ann Diagn Pathol, 2016, 21:59–64., DOI 10.1016/ j.anndiagpath.2015.11.003 Inoue T, Matsuura K, Yoshimoto T, Nguyen LT, Tsukamoto Y, Nakada C, et al. Genomic Profiling of Renal Cell Carcinoma in Patients with End-Stage Renal Disease. Cancer Sci, 2012, 103(3):569–76., DOI 10.1111/j.1349- 7006.2011.02176.x Gonzalez ML, Alaghehbandan R, Pivovarcikova K, Michalova K, Rogala J, Martinek P, et al. Reactivity of CK7 Across the Spectrum of Renal Cell Carcinomas with Clear Cells. Histopathology, 2019, 74(4):608–17., DOI 10.1111/his.13791 Hakimi AA, Tickoo SK, Jacobsen A, Sarungbam J, Sfakianos JP, Sato Y, et al. TCEB1-Mutated Renal Cell Carcinoma: A Distinct Genomic and Morphological Subtype. Mod Pathol, 2015, 28(6):845–53., DOI 10.1038/ modpathol.2015.6 Fanconi G. Familiare Infantile Perniziosaartige Anamie, pernizioses Blutbild und Konstitution). Jahrbuch Kinderheilk, 1927, 117:257–80. Osorio A, Bogliolo M, Fernandez V, Barroso A, de la Hoya M, Caldes T, et al. Evaluation of Rare Variants in the New Fanconi Anemia GeneERCC4(FANCQ, as Familial Breast/Ovarian Cancer Susceptibility Alleles. Hum Mutat, 2013, 34(12):1615–8., DOI 10.1002/humu.22438 Kim H, and D’Andrea AD. Regulation of DNA Cross-Link Repair by the Fanconi Anemia/BRCA Pathway. Genes Develop, 2012, 26(13):1393–408., DOI 10.1101/gad.195248.112 Stecklein SR, and Jensen RA. Identifying and Exploiting Defects in the Fanconi Anemia/BRCA Pathway in Oncology. Translational Res, 2012, 160(3):178–97., DOI 10.1016/j.trsl.2012.01.022 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609809 EP 1609816 DI 10.3389/pore.2021.1609809 PG 8 ER PT J AU Dong, Y Tian, J Yan, B Lv, K Li, J Fu, D AF Dong, Yinlei Tian, Junjie Yan, Bingqian Lv, Kun Li, Ji Fu, Deliang TI Liver-Metastasis-Related Genes are Potential Biomarkers for Predicting the Clinical Outcomes of Patients with Pancreatic Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE overall survival; pancreatic cancer; nomogram; liver metastasis; prognostic signature; R0 resection ID overall survival; pancreatic cancer; nomogram; liver metastasis; prognostic signature; R0 resection AB It is widely acknowledged that metastasis determines the prognosis of pancreatic adenocarcinoma (PAAD), and the liver is the most primary distant metastatic location of PAAD. It is worth exploring the value of liver-metastasis-related genetic prognostic signature (LM-PS) in predicting the clinical outcomes of PAAD patients post R0 resection. We collected 65 tumors and 165 normal pancreatic data from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression project (GTEx), respectively. Differentially expressed genes (DEGs) between primary tumor and normal pancreatic samples were intersected with DEGs between primary tumor samples with liver metastasis and those without new tumor events. The intersected 45 genes were input into univariate Cox regression analysis to identify the prognostic genes. Thirty-three prognostic livermetastasis- related genes were identified and included in least absolute shrinkage and selection operator (LASSO) analysis to develop a seven-gene LM-PS, which included six risk genes (ANO1, FAM83A, GPR87, ITGB6, KLK10, and SERPINE1) and one protective gene (SMIM32). The PAAD patients were grouped into low- and high-risk groups based on the median value of risk scores. The LM-PS harbored an independent predictive ability to distinguish patients with a high-risk of death and liver metastasis after R0 resection. Moreover, a robust prognostic nomogram based on LM-PS, the number of positive lymph nodes, and histologic grade were established to predict the overall survival of PAAD patients. Besides, a transcription factor-microRNA coregulatory network was constructed for the seven LM-PS genes, and the immune infiltration and genomic alterations were systematically explored in the TGCA-PAAD cohort. C1 [Dong, Yinlei] Fudan University, Huashan Hospital, Department of Pancreatic Surgery, Pancreatic Disease InstituteShanghai, China. [Tian, Junjie] Zhejiang University, School of Medicine, The First Affiliated Hospital, Department of UrologyHangzhou, China. [Yan, Bingqian] National Children’s Medical Center, Children’s Hospital of Fudan UniversityShanghai, China. [Lv, Kun] Fudan University, Huashan Hospital, Department of RadiologyShanghai, China. [Li, Ji] Fudan University, Huashan Hospital, Department of Pancreatic Surgery, Pancreatic Disease InstituteShanghai, China. [Fu, Deliang] Fudan University, Huashan Hospital, Department of Pancreatic Surgery, Pancreatic Disease InstituteShanghai, China. RP Fu, D (reprint author), Fudan University, Huashan Hospital, Department of Pancreatic Surgery, Pancreatic Disease Institute, Shanghai, China. EM surgeonfu@163.com CR Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, and Matrisian LM. Projecting Cancer Incidence and Deaths to 2030: the Unexpected burden of Thyroid, Liver, and Pancreas Cancers in the United States. Cancer Res, 2014, 74(11):2913–21., DOI 10.1158/0008- 5472.CAN-14-0155 Are C, Chowdhury S, Ahmad H, Ravipati A, Song T, and Shrikandhe S. Predictive Global Trends in the Incidence and Mortality of Pancreatic Cancer Based on Geographic Location, Socio-Economic Status, and Demographic Shift. J Surg Oncol, 2016, 114(6):736–42., DOI 10.1002/jso.24410 Mizrahi JD, Surana R, Valle JW, and Shroff RT. Pancreatic Cancer. The Lancet, 2020, 395(10242):2008–20., DOI 10.1016/S0140-6736(20)30974-0 Siegel RL, Miller KD, Fuchs HE, and Jemal A. Cancer Statistics, 2021. CA A Cancer J Clin, 2021, 71(1):7–33., DOI 10.3322/caac.21654 Ryan DP, Hong TS, and Bardeesy N. Pancreatic Adenocarcinoma. N Engl J Med, 2014, 371(11):1039–49., DOI 10.1056/NEJMra1404198 Heinemann V, Haas M, and Boeck S. Systemic Treatment of Advanced Pancreatic Cancer. Cancer Treat Rev, 2012, 38(7):843–53., DOI 10.1016/ j.ctrv.2011.12.004 Shi H, Li J, and Fu D. Process of Hepatic Metastasis from Pancreatic Cancer: Biology with Clinical Significance. J Cancer Res Clin Oncol, 2016, 142(6): 1137–61., DOI 10.1007/s00432-015-2024-0 Bailey P, Chang DK, Nones K, Johns AL, Patch AM, and Gingras MC. Genomic Analyses Identify Molecular Subtypes of Pancreatic Cancer. Nature, 2016, 531(7592):47–52., DOI 10.1038/nature16965 Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin W-C, and Mansour J. Whole-exome Sequencing of Pancreatic Cancer Defines Genetic Diversity and Therapeutic Targets. Nat Commun, 2015, 6:6744., DOI 10.1038/ncomms7744 Buscail L, Bournet B, and Cordelier P. Role of Oncogenic KRAS in the Diagnosis, Prognosis and Treatment of Pancreatic Cancer. Nat Rev Gastroenterol Hepatol, 2020, 17(3):153–68., DOI 10.1038/s41575-019-0245-4 Jonckheere N, Auwercx J, Hadj Bachir E, Coppin L, Boukrout N, and Vincent A. Unsupervised Hierarchical Clustering of Pancreatic Adenocarcinoma Dataset from TCGA Defines a Mucin Expression Profile that Impacts Overall Survival. Cancers, 2020, 12(11):3309., DOI 10.3390/cancers12113309 Karasinska JM, Topham JT, Kalloger SE, Jang GH, Denroche RE, and Culibrk L. Altered Gene Expression along the Glycolysis-Cholesterol Synthesis Axis Is Associated with Outcome in Pancreatic Cancer. Clin Cancer Res, 2020, 26(1): 135–46., DOI 10.1158/1078-0432.CCR-19-1543 Zhou C, Zhao Y, Yin Y, Hu Z, Atyah M, and Chen W. A Robust 6-mRNA Signature for Prognosis Prediction of Pancreatic Ductal Adenocarcinoma. Int J Biol Sci, 2019, 15(11):2282–95., DOI 10.7150/ijbs.32899 Venkat S, Tisdale AA, Schwarz JR, Alahmari AA, Maurer HC, and Olive KP. Alternative Polyadenylation Drives Oncogenic Gene Expression in Pancreatic Ductal Adenocarcinoma. Genome Res, 2020, 30(3):347–60., DOI 10.1101/ gr.257550.119 Eisenberg E, and Levanon EY. Human Housekeeping Genes, Revisited. Trends Genet, 2013, 29(10):569–74., DOI 10.1016/j.tig.2013.05.010 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, and Aksoy BA. The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data: Figure 1. Cancer Discov, 2012, 2(5):401–4., DOI 10.1158/2159-8290.CD-12-0095 Kosti I, Jain N, Aran D, Butte AJ, and Sirota M. Cross-tissue Analysis of Gene and Protein Expression in Normal and Cancer Tissues. Sci Rep, 2016, 6:24799., DOI 10.1038/srep24799 Ogura T, Yamao K, Hara K, Mizuno N, Hijioka S, and Imaoka H. Prognostic Value of K-Ras Mutation Status and Subtypes in Endoscopic Ultrasound- Guided fine-needle Aspiration Specimens from Patients with Unresectable Pancreatic Cancer. J Gastroenterol, 2013, 48(5):640–6., DOI 10.1007/s00535- 012-0664-2 Kamisawa T, Wood LD, Itoi T, and Takaori K. Pancreatic Cancer. The Lancet, 2016, 388(10039):73–85., DOI 10.1016/S0140-6736(16)00141-0 Sauter DRP, Novak I, Pedersen SF, Larsen EH, and Hoffmann EK. ANO1, TMEM16A, in Pancreatic Ductal Adenocarcinoma, PDAC). Pflugers Arch - Eur J Physiol, 2015, 467(7):1495–508., DOI 10.1007/s00424-014-1598-8 Crottes D, Lin Y-HT, Peters CJ, Gilchrist JM, Wiita AP, and Jan YN. TMEM16A Controls EGF-Induced Calcium Signaling Implicated in Pancreatic Cancer Prognosis. Proc Natl Acad Sci USA, 2019, 116(26): 13026–35., DOI 10.1073/pnas.1900703116 Chen S, Huang J, Liu Z, Liang Q, Zhang N, and Jin Y. FAM83A Is Amplified and Promotes Cancer Stem Cell-like Traits and Chemoresistance in Pancreatic Cancer. Oncogenesis, 2017, 6(3):e300., DOI 10.1038/oncsis.2017.3 Parameswaran N, Bartel CA, Hernandez-Sanchez W, Miskimen KL, Smigiel JM, and Khalil AM. A FAM83A Positive Feed-Back Loop Drives Survival and Tumorigenicity of Pancreatic Ductal Adenocarcinomas. Sci Rep, 2019, 9(1): 13396., DOI 10.1038/s41598-019-49475-5 Jiang J, Yu C, Guo X, Zhang H, Tian S, and Cai K. G Protein-Coupled Receptor GPR87 Promotes the Expansion of PDA Stem Cells through Activating JAK2/ STAT3. Mol Ther - Oncolytics, 2020, 17:384–93., DOI 10.1016/ j.omto.2020.01.006 Wang L, Zhou W, Zhong Y, Huo Y, Fan P, and Zhan S. Overexpression of G Protein-Coupled Receptor GPR87 Promotes Pancreatic Cancer Aggressiveness and Activates NF-Κb Signaling Pathway. Mol Cancer, 2017, 16(1):61., DOI 10.1186/s12943-017-0627-6 Kryza T, Achard C, Parent C, Marchand-Adam S, Guillon-Munos A, and Iochmann S. Angiogenesis Stimulated by Human Kallikrein-related Peptidase 12 Acting via a Platelet-derived Growth Factor B-dependent Paracrine Pathway. FASEB j., 2014, 28(2):740–51., DOI 10.1096/fj.13-237503 Dong Y, Loessner D, Irving-Rodgers H, Obermair A, Nicklin JL, and Clements JA.Metastasis of Ovarian Cancer Is Mediated by Kallikrein Related Peptidases. Clin Exp Metastasis, 2014, 31(1):135–47., DOI 10.1007/s10585-013-9615-4 Cao X-Y, Zhang X-X, Yang M-W, Hu L-P, Jiang S-H, and Tian G-A. Aberrant Upregulation of KLK10 Promotes Metastasis via Enhancement of EMT and FAK/SRC/ERK axis in PDAC. Biochem Biophysical Res Commun, 2018, 499(3):584–93., DOI 10.1016/j.bbrc.2018.03.194 Botla SK, Savant S, Jandaghi P, Bauer AS, Mucke O, and Moskalev EA. Early Epigenetic Downregulation of microRNA-192 Expression Promotes Pancreatic Cancer Progression. Cancer Res, 2016, 76(14):4149–59., DOI 10.1158/0008-5472.CAN-15-0390 Xiao Y. Construction of a circRNA-miRNA-mRNA Network to Explore the Pathogenesis and Treatment of Pancreatic Ductal Adenocarcinoma. J Cel Biochem, 2020, 121(1):394–406., DOI 10.1002/jcb.29194 Akula SM, Ruvolo PP, and McCubrey JA. TP53/miR-34a-associated Signaling Targets SERPINE1 Expression in Human Pancreatic Cancer. Aging, 2020, 12(3):2777–97., DOI 10.18632/aging.102776 Zhuang H, Zhou Z, Ma Z, Li Z, Liu C, and Huang S. Characterization of the Prognostic and Oncologic Values of ITGB Superfamily Members in Pancreatic Cancer. J Cel Mol. Med., 2020, 24(22):13481–93., DOI 10.1111/jcmm.15990 Wolf K, and Friedl P. Extracellular Matrix Determinants of Proteolytic and Non-proteolytic Cell Migration. Trends Cel Biol, 2011, 21(12):736–44., DOI 10.1016/j.tcb.2011.09.006 Watt FM, and Huck WTS. Role of the Extracellular Matrix in Regulating Stem Cell Fate. Nat Rev Mol Cel Biol, 2013, 14(8):467–73., DOI 10.1038/nrm3620 Puente XS, Sanchez LM, Overall CM, and Lopez-Otin C. Human and Mouse Proteases: a Comparative Genomic Approach. Nat Rev Genet, 2003, 4(7): 544–58., DOI 10.1038/nrg1111 Liu R, Liao Y-Z, Zhang W, and Zhou H-H. Relevance of Immune Infiltration and Clinical Outcomes in Pancreatic Ductal Adenocarcinoma Subtypes. Front Oncol, 2020, 10:575264., DOI 10.3389/fonc.2020.575264 Xu C, Sui S, Shang Y, Yu Z, Han J, and Zhang G. The Landscape of Immune Cell Infiltration and its Clinical Implications of Pancreatic Ductal Adenocarcinoma. J Adv Res, 2020, 24:139–48., DOI 10.1016/j.jare.2020.03.009 DeNardo DG, and Ruffell B. Macrophages as Regulators of Tumour Immunity and Immunotherapy. Nat Rev Immunol, 2019, 19(6):369–82., DOI 10.1038/ s41577-019-0127-6 Mantovani A, Sozzani S, Locati M, Allavena P, and Sica A. Macrophage Polarization: Tumor-Associated Macrophages as a Paradigm for Polarized M2 Mononuclear Phagocytes. Trends Immunol, 2002, 23(11):549–55., DOI 10.1016/s1471-4906(02)02302-5 Ye H, Zhou Q, Zheng S, Li G, Lin Q, and Wei L. Tumor-associated Macrophages Promote Progression and the Warburg Effect via CCL18/NFkB/ VCAM-1 Pathway in Pancreatic Ductal Adenocarcinoma. Cell Death Dis, 2018, 9(5):453., DOI 10.1038/s41419-018-0486-0 Wasylishen AR, Sun C, Chau GP, Qi Y, Su X, and Kim MP. Men1 Maintains Exocrine Pancreas Homeostasis in Response to Inflammation and Oncogenic Stress. Proc Natl Acad Sci USA, 2020, 117(12):6622–9., DOI 10.1073/ pnas.1920017117 Wang Y, Liu K, Ma Q, Tan Y, Du W, and Lv Y. Pancreatic Cancer Biomarker Detection by Two Support Vector Strategies for Recursive Feature Elimination. Biomarkers Med, 2019, 13(2):105–21., DOI 10.2217/bmm-2018-0273 Richter AM, Walesch SK,Wurl P, Taubert H, and Dammann RH. The Tumor Suppressor RASSF10 Is Upregulated upon Contact Inhibition and Frequently Epigenetically Silenced in Cancer. Oncogenesis, 2012, 1:e18., DOI 10.1038/ oncsis.2012.18 Chen B, Chen XP, Wu MS, Cui W, and Zhong M. Expressions of Heparanase and Upstream Stimulatory Factor in Hepatocellular Carcinoma. Eur J Med Res, 2014, 19:45., DOI 10.1186/s40001-014-0045-9 Costa L, Corre S, Michel V, Le Luel K, Fernandes J, and Ziveri J. USF1 Defect Drives P53 Degradation during Helicobacter pylori Infection and Accelerates Gastric Carcinogenesis. Gut, 2020, 69(9):1582–91., DOI 10.1136/gutjnl-2019-318640 Wang J, Gu J, You A, Li J, Zhang Y, and Rao G. The Transcription Factor USF1 Promotes Glioma Cell Invasion and Migration by Activating lncRNA HAS2- AS1. Biosci Rep, 2020, 40(8):BSR20200487., DOI 10.1042/BSR20200487 Hessmann E, Schneider G, Ellenrieder V, and Siveke JT. MYC in Pancreatic Cancer: Novel Mechanistic Insights and Their Translation into Therapeutic Strategies. Oncogene, 2016, 35(13):1609–18., DOI 10.1038/onc.2015.216 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609822 EP 1609838 DI 10.3389/pore.2021.1609822 PG 17 ER PT J AU Wang, X Yang, Q Liu, N Bian, Q Gao, M Hou, X AF Wang, Xinzhuang Yang, Quan Liu, Nan Bian, Qilong Gao, Ming Hou, Xu TI Clinical Value of TXNDC12 Combined With IDH and 1p19q as Biomarkers for Prognosis of Glioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE glioma; prognosis; TXNDC12; IDH; 1p19q ID glioma; prognosis; TXNDC12; IDH; 1p19q AB Background: Glioma is the primary malignant tumor of the central nervous system and presents high mortality and disability rates under existing treatment measures. Thioredoxin domain-containing 12 (TXNDC12) has been shown to play an important role in various malignant tumors. Therefore, we explored the clinicopathological characteristics of TXNDC12 in glioma to bring to light new ideas in its treatment. Methods: We obtained data packages related to TXNDC12 expression status in gliomas from public databases. We analyzed glioma TXNDC12 expression and patient survival status and validated the above results using glioma specimens from our institution. Next, we analyzed the value of TXNDC12 in combination with 1p19q and isocitrate dehydrogenase (IDH) on the prognosis of glioma by regression model and receiver operating characteristic curve (ROC). Finally, we explored the function of related genes by GO analysis and KEGG analysis. Results: Compared with normal brain tissue, the expression of TXNDC12 in glioma cells, regarding bothmRNA and protein levels, was significantly upregulated. The survival time of patients with high-expression of TXNDC12 in glioma cells was shortened. In the World Health Organization pathological classification, IDH status, 1p19q status, and IDH combined with 1p19q subgroups, the expression of TXNDC12 increased with the deterioration of the above indicators. Tumor local immune analysis showed that the immune cell infiltration in TXNDC12 high-expressing glioma tissue increased, the tumor purity was reduced. GO and KEGG analyses indicated that TXNDC12 may be involved in the malignant prognosis of glioma through glycosylation and antigen processing and presentation. Conclusion: We showed that TXNDC12 is significantly highly expressed in gliomas. This high expression predicts the poor prognosis of glioma patients and is related to the gliomas’ local immune microenvironment. As a tumor-related gene, TXNDC12 may be used as a new prognostic judgment molecule. C1 [Wang, Xinzhuang] Zhengzhou University, First Affiliated Hospital, Department of NeurosurgeryZhengzhou, China. [Yang, Quan] The First Affiliated Hospital of Harbin Medical University, Department of NeurosurgeryHarbin, China. [Liu, Nan] The First Affiliated Hospital of Harbin Medical University, Department of NeurosurgeryHarbin, China. [Bian, Qilong] Heze Municipal Hospital, Department of NeurosurgeryHeze, China. [Gao, Ming] The First Affiliated Hospital of Harbin Medical University, Department of NeurosurgeryHarbin, China. [Hou, Xu] The First Affiliated Hospital of Harbin Medical University, Department of NeurosurgeryHarbin, China. RP Hou, X (reprint author), The First Affiliated Hospital of Harbin Medical University, Department of Neurosurgery, Harbin, China. EM houxu1983@hotmail.com CR Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, and Barnholtz-Sloan JS. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013-2017. Neuro Oncol, 2020, 22(12 Suppl. 2):iv1–iv96., DOI 10.1093/neuonc/noaa200 Wen PY, and Kesari S. Malignant Gliomas in Adults. N Engl J Med, 2008, 359(5):492–507., DOI 10.1056/NEJMra0708126 Brat DJ, Verhaak RG, Aldape KD, Yung WKA, Salama SR, Cooper LAD, et al. Comprehensive, Integrative Genomic Analysis of Diffuse Lower-Grade Gliomas. N Engl J Med, 2015, 372(26):2481–98., DOI 10.1056/NEJMoa1402121 Youssef G, and Miller JJ. Lower Grade Gliomas. Curr Neurol Neurosci Rep, 2020, 20(7):21., DOI 10.1007/s11910-020-01040-8 Delgado-Martin B, and Medina MA. Advances in the Knowledge of the Molecular Biology of Glioblastoma and its Impact in Patient Diagnosis, Stratification, and Treatment. Adv Sci, 2020, 7(9):1902971., DOI 10.1002/ advs.201902971 Mair MJ, Geurts M, van den Bent MJ, and Berghoff AS. A Basic Review on Systemic Treatment Options in WHO Grade II-III Gliomas. Cancer Treat Rev, 2020, 92:102124., DOI 10.1016/j.ctrv.2020.102124 Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A Summary. Acta Neuropathol, 2016, 131(6):803–20., DOI 10.1007/s00401-016-1545-1 Molinaro AM, Taylor JW, Wiencke JK, and Wrensch MR. Genetic and Molecular Epidemiology of Adult Diffuse Glioma. Nat Rev Neurol, 2019, 15(7):405–17., DOI 10.1038/s41582-019-0220-2 Lapointe S, Perry A, and Butowski NA. Primary Brain Tumours in Adults. The Lancet, 2018, 392(10145):432–46., DOI 10.1016/S0140-6736(18)30990-5 Eckel-Passow JE, Lachance DH, Molinaro AM, Walsh KM, Decker PA, Sicotte H, et al. Glioma Groups Based on 1p/19q,IDH, andTERTPromoter Mutations in Tumors. N Engl J Med, 2015, 372(26):2499–508., DOI 10.1056/NEJMoa1407279 Chen R, Smith-Cohn M, Cohen AL, and Colman H. Glioma Subclassifications and Their Clinical Significance. Neurotherapeutics, 2017, 14(2):284–97., DOI 10.1007/s13311-017-0519-x Jeong W, Lee D-Y, Park S, and Rhee SG. ERp16, an Endoplasmic Reticulum- Resident Thiol-Disulfide Oxidoreductase. J Biol Chem, 2008, 283(37): 25557–66., DOI 10.1074/jbc.M803804200 Liu F, Rong Y-P, Zeng L-C, Zhang X, and Han Z-G. Isolation and Characterization of a Novel Human Thioredoxin-Like Gene hTLP19 Encoding a Secretory Protein. Gene, 2003, 315:71–8., DOI 10.1016/s0378- 1119(03)00732-7 Galligan JJ, and Petersen DR. The Human Protein Disulfide Isomerase Gene Family. Hum Genomics, 2012, 6(1):6., DOI 10.1186/1479-7364-6-6 Zhang J, Li X, Han X, Liu R, and Fang J. Targeting the Thioredoxin System for Cancer Therapy. Trends Pharmacol Sci, 2017, 38(9):794–808., DOI 10.1016/ j.tips.2017.06.001 ClarkeHJ, Chambers JE,Liniker E, andMarciniak SJ. Endoplasmic Reticulum Stress in Malignancy. Cancer Cell, 2014, 25(5):563–73., DOI 10.1016/j.ccr.2014.03.015 Wang M, and Kaufman RJ. The Impact of the Endoplasmic Reticulum Protein-Folding Environment on Cancer Development. Nat Rev Cancer, 2014, 14(9):581–97., DOI 10.1038/nrc3800 Wu J, Chen X-H,Wang X-Q, Yu Y, Ren J-M, Xiao Y, et al. ERp19 Contributes to Tumorigenicity in Human Gastric Cancer by Promoting Cell Growth, Migration and Invasion. Oncotarget, 2015, 6(14):11794–805., DOI 10.18632/ oncotarget.3649 Lee E-K, Cho H, and Kim C-W. Proteomic Analysis of Cancer Stem Cells in Human Prostate Cancer Cells. Biochem Biophysical Res Commun, 2011, 412(2):279–85., DOI 10.1016/j.bbrc.2011.07.083 Yuan K, Xie K, Lan T, Xu L, Chen X, Li X, et al. TXNDC12 Promotes EMT and Metastasis of Hepatocellular Carcinoma Cells via Activation of β-catenin. Cell Death Differ, 2020, 27(4):1355–68., DOI 10.1038/s41418- 019-0421-7 Zheng T, Chen K, Zhang X, Feng H, Shi Y, Liu L, et al. Knockdown of TXNDC9 Induces Apoptosis and Autophagy in Glioma and Mediates Cell Differentiation by P53 Activation. Aging, 2020, 12(18):18649–59., DOI 10.18632/aging.103915 Cutts J, Brookhouser N, and Brafman DA. Generation of Regionally Specific Neural Progenitor Cells, NPCs, and Neurons from Human Pluripotent Stem Cells, hPSCs). Methods Mol Biol, 2016, 1516:121–44., DOI 10.1007/ 7651_2016_357 Omuro A, and DeAngelis LM. Glioblastoma and Other Malignant Gliomas. Jama, 2013, 310(17):1842–50., DOI 10.1001/jama.2013.280319 Lim M, Xia Y, Bettegowda C, and Weller M. Current State of Immunotherapy for Glioblastoma. Nat Rev Clin Oncol, 2018, 15(7):422–42., DOI 10.1038/ s41571-018-0003-5 Rahman M, Sawyer WG, Lindhorst S, Deleyrolle LP, Harrison JK, Karachi A, et al. Adult Immuno-Oncology: Using Past Failures to Inform the Future. Neuro Oncol, 2020, 22(9):1249–61., DOI 10.1093/neuonc/noaa116 Tan AC, Ashley DM, Lopez GY, Malinzak M, Friedman HS, and Khasraw M. Management of Glioblastoma: State of the Art and Future Directions. CA A Cancer J Clin, 2020, 70(4):299–312., DOI 10.3322/caac.21613 Gusyatiner O, and Hegi ME. Glioma Epigenetics: From Subclassification to Novel Treatment Options. Semin Cancer Biol, 2018, 51:50–8., DOI 10.1016/ j.semcancer.2017.11.010 Bi J, Chowdhry S,Wu S, Zhang W, Masui K, and Mischel PS. Altered Cellular Metabolism in Gliomas - an Emerging Landscape of Actionable Codependency Targets. Nat Rev Cancer, 2020, 20(1):57–70., DOI 10.1038/ s41568-019-0226-5 Jonsson P, Lin AL, Young RJ, DiStefano NM, Hyman DM, Li BT, et al. Genomic Correlates of Disease Progression and Treatment Response in Prospectively Characterized Gliomas. Clin Cancer Res, 2019, 25(18): 5537–47., DOI 10.1158/1078-0432.Ccr-19-0032 Reuss DE, Mamatjan Y, Schrimpf D, Capper D, Hovestadt V, Kratz A, et al. IDH Mutant Diffuse and Anaplastic Astrocytomas Have Similar Age at Presentation and Little Difference in Survival: a Grading ProblemforWHO. Acta Neuropathol, 2015, 129(6):867–73., DOI 10.1007/s00401-015-1438-8 Foote MB, Papadopoulos N, and Diaz LA, Jr. Genetic Classification of Gliomas: Refining Histopathology. Cancer Cell, 2015, 28(1):9–11., DOI 10.1016/j.ccell.2015.06.014 van den Bent MJ, Weller M, Wen PY, Kros JM, Aldape K, and Chang S. A Clinical Perspective on the 2016 WHO Brain Tumor Classification and Routine Molecular Diagnostics. Neuro Oncol, 2017, 19(5):614–24., DOI 10.1093/neuonc/now277 Reifenberger G, Wirsching H-G, Knobbe-Thomsen CB, and Weller M. Advances in the Molecular Genetics of Gliomas - Implications for Classification and Therapy. Nat Rev Clin Oncol, 2017, 14(7):434–52., DOI 10.1038/nrclinonc.2016.204 Quail DF, and Joyce JA. The Microenvironmental Landscape of Brain Tumors. Cancer Cell, 2017, 31(3):326–41., DOI 10.1016/j.ccell.2017.02.009 Klemm F, Maas RR, Bowman RL, Kornete M, Soukup K, Nassiri S, et al. Interrogation of the Microenvironmental Landscape in Brain Tumors Reveals Disease-Specific Alterations of Immune Cells. Cell, 2020, 181(7):1643–60., DOI 10.1016/j.cell.2020.05.007 Pinton L, Masetto E, Vettore M, Solito S,Magri S,D’Andolfi M, et al. The Immune Suppressive Microenvironment of Human Gliomas Depends on the Accumulation of Bone Marrow-Derived Macrophages in the center of the Lesion. J Immunotherapy Cancer, 2019, 7(1):58., DOI 10.1186/s40425-019-0536-x Saha D, Martuza RL, and Rabkin SD. Macrophage Polarization Contributes to Glioblastoma Eradication by Combination Immunovirotherapy and Immune Checkpoint Blockade. Cancer Cell, 2017, 32(2):253–67., DOI 10.1016/ j.ccell.2017.07.006 Zhang P,Miska J, Lee-Chang C, Rashidi A, Panek WK, An S, et al. Therapeutic Targeting of Tumor-Associated Myeloid Cells Synergizes with Radiation Therapy for Glioblastoma. Proc Natl Acad Sci USA, 2019, 116(47): 23714–23., DOI 10.1073/pnas.1906346116 Sampson JH, Gunn MD, Fecci PE, and Ashley DM. Brain Immunology and Immunotherapy in Brain Tumours. Nat Rev Cancer, 2020, 20(1):12–25., DOI 10.1038/s41568-019-0224-7 Roy A, Coum A, Marinescu VD, Polajeva J, Smits A, Nelander S, et al. Glioma- Derived Plasminogen Activator Inhibitor-1, PAI-1, Regulates the Recruitment of LRP1 Positive Mast Cells. Oncotarget, 2015, 6(27):23647–61., DOI 10.18632/ oncotarget.4640 Attarha S, Roy A, Westermark B, and Tchougounova E. Mast Cells Modulate Proliferation, Migration and Stemness of Glioma Cells through Downregulation of GSK3β Expression and Inhibition of STAT3 Activation. Cell Signal, 2017, 37:81–92., DOI 10.1016/j.cellsig.2017.06.004 Veillon L, Fakih C, Abou-El-Hassan H, Kobeissy F, and Mechref Y. Glycosylation Changes in Brain Cancer. ACS Chem Neurosci, 2018, 9(1): 51–72., DOI 10.1021/acschemneuro.7b00271 Lan J, Guo P, Lin Y, Mao Q, Guo L, Ge J, et al. Role of Glycosyltransferase PomGnT1 in Glioblastoma Progression. Neuro-Oncology, 2015, 17(2):211–22., DOI 10.1093/neuonc/nou151 Dusoswa SA, Verhoeff J, Abels E, Mendez-Huergo SP, Croci DO, Kuijper LH, et al. Glioblastomas Exploit Truncated O-Linked Glycans for Local and Distant Immune Modulation via the Macrophage Galactose-type Lectin. Proc Natl Acad Sci USA, 2020, 117(7):3693–703., DOI 10.1073/pnas.1907921117 Hassani Z, Saleh A, Turpault S, Khiati S, Morelle W, Vignon J, et al. Phostine PST3.1a Targets MGAT5 and Inhibits Glioblastoma-Initiating Cell Invasiveness and Proliferation. Mol Cancer Res, 2017, 15(10):1376–87., DOI 10.1158/1541-7786.Mcr-17-0120 Li Y, Liu Y, Zhu H, Chen X, Tian M, Wei Y, et al. Nacetylglucosaminyltransferase I Promotes Glioma Cell Proliferation and Migration through Increasing the Stability of the Glucose Transporter GLUT1. FEBS Lett, 2020, 594(2):358–66., DOI 10.1002/1873-3468.13596 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609825 EP 1609838 DI 10.3389/pore.2021.1609825 PG 14 ER PT J AU Umar, IM Hassan, W Murtaza, G Buabeid, M Arafa, E Irfan, MH Asmawi, ZM Huang, X AF Umar, Ihtisham Muhammad Hassan, Waseem Murtaza, Ghulam Buabeid, Manal Arafa, Elshaimaa Irfan, Muhammad Hafiz Asmawi, Zaini Mohd Huang, Xianju TI The Adipokine Component in the Molecular Regulation of Cancer Cell Survival, Proliferation and Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE cell cycle; adipokines; mesenchymal transition; leptin signaling; obesity derived cancers ID cell cycle; adipokines; mesenchymal transition; leptin signaling; obesity derived cancers AB A hormonal imbalance may disrupt the rigorously monitored cellular microenvironment by hampering the natural homeostatic mechanisms. The most common example of such hormonal glitch could be seen in obesity where the uprise in adipokine levels is in virtue of the expanding bulk of adipose tissue. Such aberrant endocrine signaling disrupts the regulation of cellular fate, rendering the cells to live in a tumor supportive microenvironment. Previously, it was believed that the adipokines support cancer proliferation and metastasis with no direct involvement in neoplastic transformations and tumorigenesis. However, the recent studies have reported discrete mechanisms that establish the direct involvement of adipokine signaling in tumorigenesis. Moreover, the individual adipokine profile of the patients has never been considered in the prognosis and staging of the disease. Hence, the present manuscript has focused on the reported extensive mechanisms that culminate the basis of poor prognosis and diminished survival rate in obese cancer patients. C1 [Umar, Ihtisham Muhammad] COMSATS University Islamabad, Lahore Campus, Department of PharmacyLahore, Pakistan. [Hassan, Waseem] COMSATS University Islamabad, Lahore Campus, Department of PharmacyLahore, Pakistan. [Murtaza, Ghulam] COMSATS University Islamabad, Lahore Campus, Department of PharmacyLahore, Pakistan. [Buabeid, Manal] Ajman University, Department of Clinical SciencesAjman, United Arab Emirates. [Arafa, Elshaimaa] Ajman University, Department of Clinical SciencesAjman, United Arab Emirates. [Irfan, Muhammad Hafiz] University of Sargodha, Department of PharmacySargodha, Pakistan. [Asmawi, Zaini Mohd] University of Science Malaysia, School of Pharmaceutical SciencesPulau Pinang, Malaysia. [Huang, Xianju] South-Central University for Nationalities, College of PharmacyWuhan, China. RP Murtaza, G (reprint author), COMSATS University Islamabad, Lahore Campus, Department of Pharmacy, Lahore, Pakistan. EM gmdogar356@gmail.com CR Madsen MS, Siersbaek R, Boergesen M, Nielsen R, and Mandrup S. Peroxisome Proliferator-Activated Receptor γ and C/EBPα Synergistically Activate Key Metabolic Adipocyte Genes by Assisted Loading. Mol Cel Biol, 2014, 34(6):939–54., DOI 10.1128/mcb.01344-13 Shamsi F, Xue R, Huang TL, Lundh M, Liu Y, Leiria L, et al. FGF6 and FGF9 Regulate UCP1 Expression Independent of Brown Adipogenesis. Nat Commun, 2020, 11(1):1421., DOI 10.1038/s41467-020-15055-9 Leitner BP, Huang S, Brychta RJ, Duckworth CJ, Baskin AS, McGehee S, et al. Mapping of Human Brown Adipose Tissue in Lean and Obese Young Men. Proc Natl Acad Sci USA, 2017, 114(32):8649–54., DOI 10.1073/pnas.1705287114 Wu J, Bostrom P, Sparks LM, Ye L, Choi JH, Giang A-H, et al. Beige Adipocytes Are a Distinct Type of Thermogenic Fat Cell in Mouse and Human. Cell, 2012, 150(2):366–76., DOI 10.1016/j.cell.2012.05.016 Garcia RA, Roemmich JN, and Claycombe KJ. Evaluation of Markers of Beige Adipocytes in white Adipose Tissue of the Mouse. Nutr Metab, 2016, 13:24., DOI 10.1186/s12986-016-0081-2 Okla M, Ha J-H, Temel RE, and Chung S. BMP7 Drives Human Adipogenic Stem Cells into Metabolically Active Beige Adipocytes. Lipids, 2015, 50(2): 111–20., DOI 10.1007/s11745-014-3981-9 Liu P, Huang S, Ling S, Xu S, Wang F, ZhangW, et al. Foxp1 Controls Brown/ beige Adipocyte Differentiation and Thermogenesis through Regulating β3- AR Desensitization. Nat Commun, 2019, 10(1):5070., DOI 10.1038/s41467- 019-12988-8 Organization WH. Obesity And Overweight. News Room: Fact Sheets 2021. Available from: https://www.who.int/news-room/fact-sheets/detail/obesityand- overweight. Reeves GK, Pirie K, Beral V, Green J, Spencer E, and Bull D. Cancer Incidence and Mortality in Relation to Body Mass index in the Million Women Study: Cohort Study. Bmj, 2007, 335(7630):1134., DOI 10.1136/bmj.39367.495995.ae Arnold M, Pandeya N, Byrnes G, Renehan AG, Stevens GA, Ezzati M, et al. Global burden of Cancer Attributable to High Body-Mass index in 2012: a Population-Based Study. Lancet Oncol, 2015, 16(1):36–46., DOI 10.1016/ s1470-2045(14)71123-4 Odutola MK, Olukomogbon T, Igbinoba F, Otu TI, Ezeome E, Hassan R, et al. Cancers Attributable to Overweight and Obesity from 2012 to 2014 in Nigeria: A Population-Based Cancer Registry Study. Front Oncol, 2019, 9: 460., DOI 10.3389/fonc.2019.00460 de Gonzalez AB, Sweetland S, and Spencer E. AMeta-Analysis of Obesity and the Risk of Pancreatic Cancer. Br J Cancer, 2003, 89(3):519–23., DOI 10.1038/ sj.bjc.6601140 Holt DJ, and Grainger DW. Senescence and Quiescence Induced Compromised Function in Cultured Macrophages. Biomaterials, 2012, 33(30):7497–507., DOI 10.1016/j.biomaterials.2012.06.099 Liu H, Adler AS, Segal E, and Chang HY. A Transcriptional Program Mediating Entry into Cellular Quiescence. Plos Genet, 2007, 3(6):e91., DOI 10.1371/journal.pgen.0030091 Seyfried TN, and Huysentruyt LC. On the Origin of Cancer Metastasis. Crit Rev oncogenesis, 2013, 18(1-2):43–73., DOI 10.1615/critrevoncog.v18.i1-2.40 Piva de Freitas P, Senna CG, Tabai M, Chone CT, and Altemani A. Metastatic Basal Cell Carcinoma: A Rare Manifestation of a Common Disease. Case Rep Med, 2017, 2017:8929745., DOI 10.1155/2017/8929745 Garcia Franco CE, Torre W, Tamura A, Guillen-Grima F, San-Julian M, Martin-Algarra S, et al. Long-term Results after Resection for Bone Sarcoma Pulmonary Metastases. Eur J Cardiothorac Surg, 2010, 37(5):1205–8., DOI 10.1016/j.ejcts.2009.11.026 Cho YK, Lee J, Kim HS, Park JY, Lee WJ, Kim YJ, et al. Metabolic Health Is a Determining Factor for Incident Colorectal Cancer in the Obese Population: A Nationwide Population-based Cohort Study. Cancer Med, 2021, 10(1): 220–9., DOI 10.1002/cam4.3607 James DB, Mary KG, and Christian W. TNM Classification of Malignant Tumours. Wiley-Blackwell, 2016). McAlpine J, Leon-Castillo A, and Bosse T. The Rise of a Novel Classification System for Endometrial Carcinoma; Integration of Molecular Subclasses. J Pathol, 2018, 244(5):538–49., DOI 10.1002/path.5034 Yanovich G, Agmon H, HarelM, Sonnenblick A, Peretz T, and Geiger T. Clinical Proteomics of Breast Cancer Reveals a Novel Layer of Breast Cancer Classification. Cancer Res, 2018, 78(20):6001–10., DOI 10.1158/0008-5472.CAN-18-1079 Ilie M, Hofman V, Dietel M, Soria J-C, and Hofman P. Assessment of the PDL1 Status by Immunohistochemistry: Challenges and Perspectives for Therapeutic Strategies in Lung Cancer Patients. Virchows Arch, 2016, 468(5):511–25., DOI 10.1007/s00428-016-1910-4 Mamatjan Y, Agnihotri S, Goldenberg A, Tonge P, Mansouri S, Zadeh G, et al. Molecular Signatures for Tumor Classification. J Mol Diagn, 2017, 19(6):881–91., DOI 10.1016/j.jmoldx.2017.07.008 Caldefie-Chezet F, Damez M, de Latour M, Konska G, Mishellani F, Fusillier C, et al. Leptin: A Proliferative Factor for Breast Cancer? Biochem Biophysical Res Commun, 2005, 334(3):737–41., DOI 10.1016/j.bbrc.2005.06.077 Yang G, Fan W, Luo B, Xu Z, Wang P, Tang S, et al. Circulating Resistin Levels and Risk of Colorectal Cancer: AMeta-Analysis. Biomed Res Int, 2016, 2016:7367485., DOI 10.1155/2016/7367485 Hu W-W, Tang C-H, Sun Y, Lu T-T, Jiang P, Wu Y-M, et al. Correlation between Resistin Gene Polymorphism and Clinical Aspects of Lung Cancer. Medicine, 2017, 96(52):e9485., DOI 10.1097/md.0000000000009485 Diakowska D, Markocka-Ma˛czka K, Szelachowski P, and Grabowski K. Serum Levels of Resistin, Adiponectin, and Apelin in Gastroesophageal Cancer Patients. Dis Markers, 2014, 2014:619649., DOI 10.1155/2014/619649 Cymbaluk-Ploska A, Chudecka-Glaz A, Pius-Sadowska E, Sompolska- Rzechula A, Machali´nski B, and Menkiszak J. Circulating Serum Level of Visfatin in Patients with Endometrial Cancer. Biomed Res Int, 2018, 2018: 8576179., DOI 10.1155/2018/8576179 Vallega KA, Liu N, Myers JS, Yu K, and Sang Q-XA. Elevated Resistin Gene Expression in African American Estrogen and Progesterone Receptor Negative Breast Cancer. PloS one, 2016, 11(6):e0157741., DOI 10.1371/ journal.pone.0157741 Funcke J-B, and Scherer PE. Beyond Adiponectin and Leptin: Adipose Tissue-Derived Mediators of Inter-organ Communication. J Lipid Res, 2019, 60–16481684., DOI 10.1194/jlr.R094060 Wang H, Liang X, Li M, Tao X, Tai S, Fan Z, et al. Chemokine, CCmotif, Ligand 18 Upregulates Slug Expression to Promote Stem-cell like Features by Activating the Mammalian Target of Rapamycin Pathway in Oral Squamous Cell Carcinoma. Cancer Sci, 2017, 108(8):1584–93., DOI 10.1111/cas.13289 Park J, and Scherer PE. Leptin and Cancer: from Cancer Stem Cells toMetastasis. Endocrine-related cancer, 2011, 18(4):C25–C29., DOI 10.1530/erc-11-0163 Hardwick JCH, Van Den Brink GR, Offerhaus GJ, Van Deventer SJH, and Peppelenbosch MP. Leptin Is a Growth Factor for Colonic Epithelial Cells. Gastroenterology, 2001, 121(1):79–90., DOI 10.1053/gast.2001.25490 Attoub S, Noe V, Pirola L, Bruyneel E, Chastre E, Mareel M, et al. Leptin Promotes Invasiveness of Kidney and Colonic Epithelial Cells via Phosphoinositide 3-kinase-, Rho-, and Rac-dependent Signaling Pathways. FASEB j, 2000, 14(14):2329–38., DOI 10.1096/fj.00-0162 Buyse M, Berlioz F, Guilmeau S, Tsocas A, Voisin T, Peranzi G, et al. PepT1- mediated Epithelial Transport of Dipeptides and Cephalexin Is Enhanced by Luminal Leptin in the Small Intestine. J Clin Invest, 2001, 108(10):1483–94., DOI 10.1172/jci13219 Koda M, Sulkowska M, Kanczuga-Koda L, Surmacz E, and Sulkowski S. Overexpression of the Obesity Hormone Leptin in Human Colorectal Cancer. J Clin Pathol, 2007, 60(8):902–6., DOI 10.1136/jcp.2006.041004 Gu L, Wang C-D, Cao C, Cai L-R, Li D-H, and Zheng Y-Z. Association of Serum Leptin with Breast Cancer: A Meta-Analysis. Medicine, 2019, 98(5): e14094., DOI 10.1097/md.0000000000014094 Jeong Y-J, Bong J-G, Park S-H, Choi J-H, and Oh H-K. Expression of Leptin, Leptin Receptor, Adiponectin, and Adiponectin Receptor in Ductal CarcinomaIn Situand Invasive Breast Cancer. J Breast Cancer, 2011, 14(2):96–103., DOI 10.4048/jbc.2011.14.2.96 Harris HR, Tworoger SS, Hankinson SE, Rosner BA, and Michels KB. Plasma Leptin Levels and Risk of Breast Cancer in Premenopausal Women. Cancer Prev Res, 2011, 4(9):1449–56., DOI 10.1158/1940-6207.capr-11-0125 Gu F, Kraft P, Rice M, and Michels KB. Leptin and Leptin Receptor Genes in Relation to Premenopausal Breast Cancer Incidence and Grade in Caucasian Women. Breast Cancer Res Treat, 2012, 131(1):17–25., DOI 10.1007/s10549- 011-1778-6 Ferla R, BonomiM, Otvos L, Jr, and Surmacz E. Glioblastoma-derived Leptin Induces Tube Formation and Growth of Endothelial Cells: Comparison with VEGF Effects. BMC cancer, 2011, 11:303., DOI 10.1186/1471-2407-11-303 Wang X-J, Yuan S-L, Lu Q, Lu Y-R, Zhang J, Liu Y, et al. Potential Involvement of Leptin in Carcinogenesis of Hepatocellular Carcinoma. Wjg, 2004, 10(17):2478–81., DOI 10.3748/wjg.v10.i17.2478 Zhou J, Lei W, Shen L, Luo H-S, and Shen Z-X. Primary Study of Leptin and Human Hepatocellular Carcinoma In Vitro. Wjg, 2008, 14(18):2900–4., DOI 10.3748/wjg.14.2900 Saxena NK, Sharma D, Ding X, Lin S, Marra F, Merlin D, et al. Concomitant Activation of the JAK/STAT, PI3K/AKT, and ERK Signaling Is Involved in Leptin-Mediated Promotion of Invasion and Migration of Hepatocellular Carcinoma Cells. Cancer Res, 2007, 67(6):2497–507., DOI 10.1158/0008- 5472.can-06-3075 Ptak A, Kolaczkowska E, and Gregoraszczuk EL. Leptin Stimulation of Cell Cycle and Inhibition of Apoptosis Gene and Protein Expression in OVCAR-3 Ovarian Cancer Cells. Endocrine, 2013, 43(2):394–403., DOI 10.1007/s12020- 012-9788-7 Pan W, Allison MB, Sabatini P, Rupp A, Adams J, Patterson C, et al. Transcriptional and Physiological Roles for STAT Proteins in Leptin Action. Mol Metab, 2019, 22:121–31., DOI 10.1016/j.molmet.2019.01.007 Saxena NK, Vertino PM, Anania FA, and Sharma D. Leptin-induced Growth Stimulation of Breast Cancer Cells Involves Recruitment of Histone Acetyltransferases and Mediator Complex to CYCLIN D1 Promoter via Activation of Stat3. J Biol Chem, 2007, 282(18):13316–25., DOI 10.1074/ jbc.m609798200 Saxena NK, Titus MA, Ding X, Floyd J, Srinivasan S, Sitaraman SV, et al. Leptin as a Novel Profibrogenic Cytokine in Hepatic Stellate Cells: Mitogenesis and Inhibition of Apoptosis Mediated by Extracellular Regulated Kinase, Erk, and Akt Phosphorylation. FASEB j, 2004, 18(13): 1612–4., DOI 10.1096/fj.04-1847fje Higurashi T, Endo H, Uchiyama T, Uchiyama S, Yamada E, Ohkubo H, et al. Conditional Knockout of the Leptin Receptor in the Colonic Epithelium Revealed the Local Effects of Leptin Receptor Signaling in the Progression of Colonic Tumors in Mice. Carcin, 2014, 35(9):2134–41., DOI 10.1093/carcin/ bgu135 Xiong H, Zhang Z-G, Tian X-Q, Sun D-F, Liang Q-C, Zhang Y-J, et al. Inhibition of JAK1, 2/STAT3 Signaling Induces Apoptosis, Cell Cycle Arrest, and Reduces Tumor Cell Invasion in Colorectal Cancer Cells. Neoplasia, 2008, 10(3):287–97., DOI 10.1593/neo.07971 Lin Q, Lai R, Chirieac LR, Li C, Thomazy VA, Grammatikakis I, et al. Constitutive Activation of JAK3/STAT3 in Colon Carcinoma Tumors and Cell Lines. Am J Pathol, 2005, 167(4):969–80., DOI 10.1016/s0002-9440(10, 61187-x Nowakowska-Zajdel E, Mazurek U, Stachowicz M, Niedworok E, Fatyga E, and Muc-Wierzgo´n M. Cellular Signal Transduction Pathways by Leptin in Colorectal Cancer Tissue: Preliminary Results. ISRN Endocrinol, 2011, 2011: 575397., DOI 10.5402/2011/575397 Mittenbuhler MJ, Sprenger H-G, Gruber S, Wunderlich CM, Kern L, Bruning JC, et al. Hepatic Leptin Receptor Expression Can Partially Compensate for IL-6Rα Deficiency in DEN-Induced Hepatocellular Carcinoma. Mol Metab, 2018, 17:122–33., DOI 10.1016/j.molmet.2018.08.010 Zheng Q, Banaszak L, Fracci S, Basali D, Dunlap SM, Hursting SD, et al. Leptin Receptor Maintains Cancer Stem-like Properties in Triple Negative Breast Cancer Cells. Endocrine-related cancer, 2013, 20(6):797–808., DOI 10.1530/erc-13-0329 Thiagarajan PS, Zheng Q, Bhagrath M, Mulkearns-Hubert EE, Myers MG, Lathia JD, et al. STAT3 Activation by Leptin Receptor Is Essential for TNBC Stem Cell Maintenance. Endocr Relat Cancer, 2017, 24(8):415–26., DOI 10.1530/erc-16-0349 Aparicio T, Kotelevets L, Tsocas A, Laigneau JP, Sobhani I, Chastre E, et al. Leptin Stimulates the Proliferation of Human colon Cancer Cells In Vitro but Does Not Promote the Growth of colon Cancer Xenografts in Nude Mice or Intestinal Tumorigenesis in ApcMin/+ Mice. Gut, 2005, 54(8):1136–45., DOI 10.1136/gut.2004.060533 Deo DD, Rao AP, Bose SS, Ouhtit A, Baliga SB, Rao SA, et al. Differential Effects of Leptin on the Invasive Potential of Androgen-dependent and -independent Prostate Carcinoma Cells. J Biomed Biotechnol, 2008, 2008: 163902., DOI 10.1155/2008/163902 Naviglio S, Di Gesto D, Sorrentino A, Illiano F, Sorvillo L, Abbruzzese A, et al. Leptin Enhances Growth Inhibition by cAMP Elevating Agents through Apoptosis of MDA-MB-231 Breast Cancer Cells. Cancer Biol Ther, 2009, 8(12):1183–90., DOI 10.4161/cbt.8.12.8562 Spina A, Di Maiolo F, Esposito A, Sapio L, Chiosi E, Sorvillo L, et al. cAMP Elevation Down-Regulates β3 Integrin and Focal Adhesion Kinase and Inhibits Leptin-Induced Migration of MDA-MB-231 Breast Cancer Cells. BioResearch open access, 2012, 1(6):324–32., DOI 10.1089/biores.2012.0270 Catalano S, Mauro L, Bonofiglio D, Pellegrino M, Qi H, Rizza P, et al. In Vivo and In Vitro Evidence that PPARγ Ligands Are Antagonists of Leptin Signaling in Breast Cancer. Am J Pathol, 2011, 179(2):1030–40., DOI 10.1016/j.ajpath.2011.04.026 Alshaker H, Krell J, Frampton AE, Waxman J, Blyuss O, Zaikin A, et al. Leptin Induces Upregulation of Sphingosine Kinase 1 in Oestrogen Receptor- Negative Breast Cancer via Src Family Kinase-Mediated, Janus Kinase 2- independent Pathway. Breast Cancer Res : BCR, 2014, 16(5):426., DOI 10.1186/ s13058-014-0426-6 Bjorbaek C, El-Haschimi K, Frantz JD, and Flier JS. The Role of SOCS-3 in Leptin Signaling and Leptin Resistance. J Biol Chem, 1999, 274(42): 30059–65., DOI 10.1074/jbc.274.42.30059 Wan J, Che Y, Kang N, and Wu W. SOCS3 Blocks HIF-1α Expression to Inhibit Proliferation and Angiogenesis of Human Small Cell Lung Cancer by Downregulating Activation of Akt, but Not STAT3. Mol Med Rep, 2015, 12(1):83–92., DOI 10.3892/mmr.2015.3368 Knobelspies H, Zeidler J, Hekerman P, Bamberg-Lemper S, and Becker W. Mechanism of Attenuation of Leptin Signaling under Chronic Ligand Stimulation. BMC Biochem, 2010, 11:2., DOI 10.1186/1471-2091-11-2 Bode J, Ludwig S, Freitas CA, Schaper F, Wammers M, Melmed S, et al. The MKK6/p38 Mitogen-Activated Protein Kinase Pathway Is Capable of Inducing SOCS3 Gene Expression and Inhibits IL-6-Induced Transcription. Biol Chem, 2001, 382:1447–53., DOI 10.1515/bc.2001.178 Thompson KJ, Lau KN, Johnson S, Martinie JB, Iannitti DA, McKillop IH, et al. Leptin Inhibits Hepatocellular Carcinoma Proliferation via P38-MAPKdependent Signalling. Hpb, 2011, 13(4):225–33., DOI 10.1111/j.1477- 2574.2010.00259.x Inagaki-Ohara K, Mayuzumi H, Kato S, Minokoshi Y, Otsubo T, Kawamura YI, et al. Enhancement of Leptin Receptor Signaling by SOCS3 Deficiency Induces Development of Gastric Tumors in Mice. Oncogene, 2014, 33(1): 74–84., DOI 10.1038/onc.2012.540 Ramani K, Yang H, Xia M, Ara AI, Mato JM, and Lu SC. Leptin’s Mitogenic Effect in Human Liver Cancer Cells Requires Induction of Both Methionine Adenosyltransferase 2A and 2beta. Hepatology, 2008, 47(2):521–31., DOI 10.1002/hep.22064 Stefanou N, Papanikolaou V, Furukawa Y, Nakamura Y, and Tsezou A. Leptin as a Critical Regulator of Hepatocellular Carcinoma Development through Modulation of Human Telomerase Reverse Transcriptase. BMC cancer, 2010, 10:442., DOI 10.1186/1471-2407-10-442 Kasiappan R, Sun Y, Lungchukiet P, Quarni W, Zhang X, and BaiW. Vitamin D Suppresses Leptin Stimulation of Cancer Growth through microRNA. Cancer Res, 2014, 74(21):6194–204., DOI 10.1158/0008-5472.can-14-1702 Fukada T, Ohtani T, Yoshida Y, Shirogane T, Nishida K, Nakajima K, et al. STAT3 Orchestrates Contradictory Signals in Cytokine-Induced G1 to S Cell- Cycle Transition.EMBO J, 1998, 17(22):6670–7., DOI 10.1093/emboj/17.22.6670 Harbuzariu A, Rampoldi A, Daley-Brown DS, Candelaria P, Harmon TL, Lipsey CC, et al. Leptin-Notch Signaling axis Is Involved in Pancreatic Cancer Progression. Oncotarget, 2017, 8(5):7740–52., DOI 10.18632/oncotarget.13946 Feldheim J, Kessler AF, Schmitt D, Salvador E, Monoranu CM, Feldheim JJ, et al. Ribosomal Protein S27/Metallopanstimulin-1, RPS27, in Glioma-A New Disease Biomarker? Cancers, 2020, 12(5):1085., DOI 10.3390/ cancers12051085 Cao D, Luo Y, Qin S, YuM,Mu Y, Ye G, et al. Metallopanstimulin-1, MPS-1, Mediates the Promotion Effect of Leptin on Colorectal Cancer through Activation of JNK/c-Jun Signaling Pathway. Cel Death Dis, 2019, 10(9): 655., DOI 10.1038/s41419-019-1911-8 Yang Z-Y, Jiang H, Qu Y, WeiM, Yan M, Zhu Z-G, et al. Metallopanstimulin- 1 Regulates Invasion and Migration of Gastric Cancer Cells Partially through Integrin β4. Carcinogenesis, 2013, 34(12):2851–60., DOI 10.1093/carcin/ bgt226 Christopoulos PF, Msaouel P, and Koutsilieris M. The Role of the Insulin-like Growth Factor-1 System in Breast Cancer. Mol Cancer, 2015, 14:43., DOI 10.1186/s12943-015-0291-7 Min DY, Jung E, Kim J, Lee YH, and Shin SY. Leptin Stimulates IGF-1 Transcription by Activating AP-1 in Human Breast Cancer Cells. BMB Rep, 2019, 52(6):385–90., DOI 10.5483/bmbrep.2019.52.6.189 Yang W-H, Chang A-C, Wang S-W,Wang S-J, Chang Y-S, Chang T-M, et al. Leptin Promotes VEGF-C Production and Induces Lymphangiogenesis by Suppressing miR-27b in Human Chondrosarcoma Cells. Scientific Rep, 2016, 6(1):28647., DOI 10.1038/srep28647 Carino C, Olawaiye AB, Cherfils S, Serikawa T, Lynch MP, Rueda BR, et al. Leptin Regulation of Proangiogenic Molecules in Benign and Cancerous Endometrial Cells. Int J Cancer, 2008, 123(12):2782–90., DOI 10.1002/ ijc.23887 Zhou W, Guo S, and Gonzalez-Perez RR. Leptin Pro-angiogenic Signature in Breast Cancer Is Linked to IL-1 Signalling. Br J Cancer, 2011, 104(1):128–37., DOI 10.1038/sj.bjc.6606013 Gonzalez-Perez RR, Xu Y, Guo S, Watters A, Zhou W, and Leibovich SJ. Leptin Upregulates VEGF in Breast Cancer via Canonic and Non-canonical Signalling Pathways and NFκB/HIF-1α Activation. Cell Signal, 2010, 22(9): 1350–62., DOI 10.1016/j.cellsig.2010.05.003 Guo S, and Gonzalez-Perez RR. Notch, IL-1 and Leptin Crosstalk Outcome, NILCO, Is Critical for Leptin-Induced Proliferation, Migration and VEGF/ VEGFR-2 Expression in Breast Cancer. PloS one, 2011, 6(6):e21467., DOI 10.1371/journal.pone.0021467 Zhao X, Dong Y, Zhang J, Li D, Hu G, Yao J, et al. Leptin Changes Differentiation Fate and Induces Senescence in Chondrogenic Progenitor Cells. Cell Death Dis, 2016, 7(4):e2188., DOI 10.1038/cddis.2016.68 He J-Y,Wei X-H, Li S-J, Liu Y, Hu H-L, Li Z-Z, et al. Adipocyte-derived IL-6 and Leptin Promote Breast Cancer Metastasis via Upregulation of Lysyl Hydroxylase-2 Expression. Cell Commun signaling : CCS, 2018, 16(1):100., DOI 10.1186/s12964-018-0309-z Jaffe T, and Schwartz B. Leptin Promotes Motility and Invasiveness in Human colon Cancer Cells by Activating Multiple Signal-Transduction Pathways. Int J Cancer, 2008, 123(11):2543–56., DOI 10.1002/ijc.23821 Strong AL, Strong TA, Rhodes LV, Semon JA, Zhang X, Shi Z, et al. Obesity Associated Alterations in the Biology of Adipose Stem Cells Mediate Enhanced Tumorigenesis by Estrogen Dependent Pathways. Breast Cancer Res, 2013, 15(5):R102., DOI 10.1186/bcr3569 Yan D, Avtanski D, Saxena NK, and Sharma D. Leptin-induced Epithelial- Mesenchymal Transition in Breast Cancer Cells Requires β-Catenin Activation via Akt/GSK3- and MTA1/Wnt1 Protein-dependent Pathways. J Biol Chem, 2012, 287(11):8598–612., DOI 10.1074/jbc.m111.322800 Liang X, Wang S, Wang X, Zhang L, Zhao H, and Zhang L. Leptin Promotes the Growth of Breast Cancer by Upregulating the Wnt/β-Catenin Pathway. Exp Ther Med, 2018, 16(2):767–71., DOI 10.3892/etm.2018.6212 Haque I, Ghosh A, Acup S, Banerjee S, Dhar K, Ray A, et al. Leptin-induced ER-α-Positive Breast Cancer Cell Viability and Migration Is Mediated by Suppressing CCN5-Signaling via Activating JAK/AKT/STAT-pathway. BMC cancer, 2018, 18(1):99., DOI 10.1186/s12885-018-3993-6 Knight BB, Oprea-Ilies GM, Nagalingam A, Yang L, Cohen C, Saxena NK, et al. Survivin Upregulation, Dependent on Leptin-EGFR-Notch1 axis, Is Essential for Leptin-Induced Migration of Breast Carcinoma Cells. Endocrine-related cancer, 2011, 18(4):413–28., DOI 10.1530/erc-11-0075 Battle M, Gillespie C, Quarshie A, Lanier V, Harmon T, Wilson K, et al. Obesity Induced a Leptin-Notch Signaling axis in Breast Cancer. Int J Cancer, 2014, 134(7):1605–16., DOI 10.1002/ijc.28496 Mocino-Rodriguez MD, Santillan-Benitez JG, Dozal-Dominguez DS, Hernandez-Navarro MD, Flores-Merino MV, Sandoval-Cabrera A, et al. Expression of AdipoR1 and AdipoR2 Receptors as Leptin-Breast Cancer Regulation Mechanisms. Dis Markers, 2017, 2017:4862016., DOI 10.1155/ 2017/4862016 Wu D-C, Zhang M-F, Su S-G, Fang H-Y, Wang X-H, He D, et al. HEY2, a Target of miR-137, Indicates Poor Outcomes and Promotes Cell Proliferation and Migration in Hepatocellular Carcinoma. Oncotarget, 2016, 7(25): 38052–63., DOI 10.18632/oncotarget.9343 Tanaka T, Terai Y, Kogata Y, Ashihara K, Maeda K, Fujiwara S, et al. CD24 Expression as aMarker for Predicting Clinical Outcome and Invasive Activity in Uterine Cervical Cancer. Oncol Rep, 2015, 34(5):2282–8., DOI 10.3892/ or.2015.4257 Xiaoping L, Xiaowei Z, Leizhen Z, and Weijian G. Expression and Significance of CD44 and P-AKT in Pancreatic Head Cancer. World J Surg Oncol, 2015, 13:334., DOI 10.1186/s12957-015-0746-8 Tsai CF, Chen JH, Wu CT, Chang PC, Wang SL, and Yeh WL. Induction of Osteoclast-like Cell Formation by Leptin-Induced Soluble Intercellular Adhesion Molecule Secreted from Cancer Cells. Ther Adv Med Oncol, 2019, 11:1758835919846806., DOI 10.1177/1758835919846806 Dong Z, Fu S, Xu X, Yang Y, Du L, Li W, et al. Leptin-mediated Regulation of ICAM-1 Is Rho/ROCK Dependent and Enhances Gastric Cancer Cell Migration. Br J Cancer, 2014, 110(7):1801–10., DOI 10.1038/bjc.2014.70 Strong AL, Ohlstein JF, Biagas BA, Rhodes LV, Pei DT, Tucker HA, et al. Leptin Produced by Obese Adipose Stromal/stem Cells Enhances Proliferation and Metastasis of Estrogen Receptor Positive Breast Cancers. Breast Cancer Res, 2015, 17(1):112., DOI 10.1186/s13058-015-0622-z Sabol RA, Bowles AC, Cˆote A, Wise R, O’Donnell B, Matossian MD, et al. Leptin Produced by Obesity-Altered Adipose Stem Cells Promotes Metastasis but Not Tumorigenesis of Triple-Negative Breast Cancer in Orthotopic Xenograft and Patient-Derived Xenograft Models. Breast Cancer Res : BCR, 2019, 21(1):67., DOI 10.1186/s13058-019-1153-9 Wei L, Li K, Pang X, Guo B, SuM, Huang Y, et al. Leptin Promotes Epithelial- Mesenchymal Transition of Breast Cancer via the Upregulation of Pyruvate Kinase M2. J Exp Clin Cancer Res : CR, 2016, 35(1):166., DOI 10.1186/s13046- 016-0446-4 Wang L, Tang C, Cao H, Li K, Pang X, Zhong L, et al. Activation of IL-8 via PI3K/Akt-dependent Pathway Is Involved in Leptin-Mediated Epithelial- Mesenchymal Transition in Human Breast Cancer Cells. Cancer Biol Ther, 2015, 16(8):1220–30., DOI 10.1080/15384047.2015.1056409 Xu M, Cao FL, Li N, Gao X, Su X, and Jiang X. Leptin Induces Epithelial-To- Mesenchymal Transition via Activation of the ERK Signaling Pathway in Lung Cancer Cells. Oncol Lett, 2018, 16(4):4782–8., DOI 10.3892/ol.2018.9230 Fazolini NP, Cruz AL, Werneck MB, Viola JP, Maya-Monteiro CM, and Bozza PT. Leptin Activation of mTOR Pathway in Intestinal Epithelial Cell Triggers Lipid Droplet Formation, Cytokine Production and Increased Cell Proliferation. Cell Cycle, 2015, 14(16):2667–76., DOI 10.1080/15384101.2015.1041684 Lee S-M, Choi H-J, Oh C-H, Oh J-W, and Han J-S. Leptin Increases TNF-α Expression and Production through Phospholipase D1 in Raw 264.7 Cells. PloS one, 2014, 9(7):e102373., DOI 10.1371/journal.pone.0102373 Wu D, Cheng J, Sun G, Wu S, Li M, Gao Z, et al. p70S6K Promotes IL-6- induced Epithelial-Mesenchymal Transition and Metastasis of Head and Neck Squamous Cell Carcinoma. Oncotarget, 2016, 7(24):36539–50., DOI 10.18632/oncotarget.9282 Hao Y, Baker D, and Ten Dijke P. TGF-β-Mediated Epithelial-Mesenchymal Transition and Cancer Metastasis. Ijms, 2019, 20(11):2767., DOI 10.3390/ ijms20112767 Xing S, Yu W, Zhang X, Luo Y, Lei Z, Huang D, et al. Isoviolanthin Extracted from Dendrobium Officinale Reverses TGF-B1-Mediated Epithelial- Mesenchymal Transition in Hepatocellular Carcinoma Cells via Deactivating the TGF-β/Smad and PI3K/Akt/mTOR Signaling Pathways. Ijms, 2018, 19(6):1556., DOI 10.3390/ijms19061556 Ogunwobi OO, Wang T, Zhang L, and Liu C. Cyclooxygenase-2 and Akt Mediate Multiple Growth-Factor-Induced Epithelial-Mesenchymal Transition in Human Hepatocellular Carcinoma. J Gastroenterol Hepatol, 2012, 27(3):566–78., DOI 10.1111/j.1440-1746.2011.06980.x Bhat AA, Ahmad R, Uppada SB, Singh AB, and Dhawan P. Claudin-1 Promotes TNF-α-Induced Epithelial-Mesenchymal Transition and Migration in Colorectal Adenocarcinoma Cells. Exp Cel Res, 2016, 349(1): 119–27., DOI 10.1016/j.yexcr.2016.10.005 Wang H, Wang H-S, Zhou B-H, Li C-L, Zhang F,Wang X-F, et al. Epithelialmesenchymal Transition, EMT, Induced by TNF-α Requires AKT/GSK-3β- mediated Stabilization of Snail in Colorectal Cancer. PloS one, 2013, 8(2): e56664., DOI 10.1371/journal.pone.0056664 Valle A, Sastre-Serra J, Pol C, Miro AM, Oliver J, and Roca P. Proteomic Analysis of MCF-7 Breast Cancer Cell Line Exposed to Leptin. Anal Cell Pathol, 2011, 34(3):147–57., DOI 10.1155/2011/405253 Katsogiannou M, Andrieu C, and Rocchi P. Heat Shock Protein 27 Phosphorylation State Is Associated with Cancer Progression. Front Genet, 2014, 5:346., DOI 10.3389/fgene.2014.00346 Mendonsa AM, ChalfantMC, Gorden LD, and VanSaunMN.Modulation of the Leptin Receptor Mediates Tumor Growth and Migration of Pancreatic Cancer Cells. PloS one, 2015, 10(4):e0126686., DOI 10.1371/journal.pone.0126686 Giordano C, Vizza D, Panza S, Barone I, Bonofiglio D, Lanzino M, et al. Leptin Increases HER2 Protein Levels through a STAT3-Mediated Up- Regulation of Hsp90 in Breast Cancer Cells. Mol Oncol, 2013, 7(3): 379–91., DOI 10.1016/j.molonc.2012.11.002 Lipsey CC, Harbuzariu A, Robey RW, Huff LM, Gottesman MM, and Gonzalez-Perez RR. Leptin Signaling Affects Survival and Chemoresistance of Estrogen Receptor Negative Breast Cancer. Ijms, 2020, 21(11):3794., DOI 10.3390/ijms21113794 Fiedor E, Zajda K, and Gregoraszczuk EL. Leptin Receptor Antagonists’ Action on HDAC Expression Eliminating the Negative Effects of Leptin in Ovarian Cancer. Cancer Genomics Proteomics, 2018, 15(4):329–36., DOI 10.21873/cgp.20091 Pachynski RK, Wang P, Salazar N, Zheng Y, Nease L, Rosalez J, et al. Chemerin Suppresses Breast Cancer Growth by Recruiting Immune Effector Cells into the Tumor Microenvironment. Front Immunol, 2019, 10:983., DOI 10.3389/fimmu.2019.00983 Harbuzariu A, and Gonzalez-Perez RR. Leptin-Notch axis Impairs 5- fluorouracil Effects on Pancreatic Cancer. Oncotarget, 2018, 9(26): 18239–53., DOI 10.18632/oncotarget.24435 Osaki M, and Okada F. Exosomes and Their Role in Cancer Progression. Yonago Acta Med, 2019, 62(2):182–90., DOI 10.33160/yam.2019.06.002 Giordano C, Gelsomino L, Barone I, Panza S, Augimeri G, Bonofiglio D, et al. Leptin Modulates Exosome Biogenesis in Breast Cancer Cells: An Additional Mechanism in Cell-To-Cell Communication. Jcm, 2019, 8(7):1027., DOI 10.3390/jcm8071027 Raut PK, Choi DY, Kim SH, Hong JT, Kwon TK, Jeong JH, et al. Estrogen Receptor Signaling Mediates Leptin-Induced Growth of Breast Cancer Cells via Autophagy Induction. Oncotarget, 2017, 8(65):109417–35., DOI 10.18632/ oncotarget.22684 Nepal S, Kim MJ, Hong JT, Kim SH, Sohn D-H, Lee SH, et al. Autophagy Induction by Leptin Contributes to Suppression of Apoptosis in Cancer Cells and Xenograft Model: Involvement of p53/FoxO3A axis. Oncotarget, 2015, 6(9):7166–81., DOI 10.18632/oncotarget.3347 Shrestha M, and Park P-H. p53 Signaling Is Involved in Leptin-Induced Growth of Hepatic and Breast Cancer Cells. Korean J Physiol Pharmacol, 2016, 20(5):487–98., DOI 10.4196/kjpp.2016.20.5.487 Yehuda-Shnaidman E, Nimri L, Tarnovscki T, Kirshtein B, Rudich A, and Schwartz B. Secreted Human Adipose Leptin Decreases Mitochondrial Respiration in HCT116 colon Cancer Cells. PloS one, 2013, 8(9):e74843., DOI 10.1371/journal.pone.0074843 Park J, Kusminski CM, Chua SC, and Scherer PE. Leptin Receptor Signaling Supports Cancer Cell Metabolism through Suppression of Mitochondrial Respiration In Vivo. Am J Pathol, 2010, 177(6):3133–44., DOI 10.2353/ ajpath.2010.100595 Wang C-Q, Wang Y, Huang B-F, Tang C-H, Du Z, Zeng Y, et al. High Expression of Both Resistin and Fascin-1 Predicts a Poor Prognosis in Patients with Colorectal Cancer. Biomed Research International, 2020, 2020:8753175., DOI 10.1155/2020/8753175 Mihajlovic M, Ninic A, Sopic M, Miljkovic M, Stefanovic A, Vekic J, et al. Association Among Resistin, Adenylate Cyclase-Associated Protein 1 and High-Density Lipoprotein Cholesterol in Patients with Colorectal Cancer: a Multi-Marker Approach, as a Hallmark of Innovative Predictive, Preventive, and Personalized Medicine. EPMA J, 2019, 10(3):307–16., DOI 10.1007/ s13167-019-00178-x Avtanski D, Garcia A, Caraballo B, Thangeswaran P, Marin S, Bianco J, et al. Resistin Induces Breast Cancer Cells Epithelial to Mesenchymal Transition, EMT, and Stemness through Both Adenylyl Cyclase-Associated Protein 1, CAP1)-dependent and CAP1-independent Mechanisms. Cytokine, 2019, 120:155–64., DOI 10.1016/j.cyto.2019.04.016 Weber D, Satlof L, Lavi A, Bahl K, Kaiser M, Chen K, et al. SAT-335 Resistin Induces Epithelial to Mesenchymal Transition, EMT, in Breast Cancer Cells through Activation of AXL Tyrosine Kinase Receptor. J Endocr Soc, 2019, 3(Suppl. 1):SAT-335., DOI 10.1210/js.2019-sat-335 Qiu L, Zhang G-F, Yu L, Wang H-Y, Jia X-J, and Wang T-J. Novel Oncogenic and Chemoresistance-Inducing Functions of Resistin in Ovarian Cancer Cells Require miRNAs-Mediated Induction of Epithelial-To-Mesenchymal Transition. Scientific Rep, 2018, 8(1):12522., DOI 10.1038/s41598-018- 30978-6 Yang C-C, Chang S-F, Chao J-K, Lai Y-L, Chang W-E, Hsu W-H, et al. Activation of AMP-Activated Protein Kinase Attenuates Hepatocellular Carcinoma Cell Adhesion Stimulated by Adipokine Resistin. BMC cancer, 2014, 14:112., DOI 10.1186/1471-2407-14-112 Huang W-S, Yang J-T, Lu C-C, Chang S-F, Chen C-N, Su Y-P, et al. Fulvic Acid Attenuates Resistin-Induced Adhesion of HCT-116 Colorectal Cancer Cells to Endothelial Cells. Ijms, 2015, 16(12):29370–82., DOI 10.3390/ ijms161226174 Hsieh Y-Y, Shen C-H, Huang W-S, Chin C-C, Kuo Y-H, Hsieh M, et al. Resistin-induced Stromal Cell-Derived Factor-1 Expression through Toll-like Receptor 4 and Activation of P38 MAPK/NFκB Signaling Pathway in Gastric Cancer Cells. J Biomed Sci, 2014, 21(1):59., DOI 10.1186/1423-0127-21-59 Deshmukh SK, Srivastava SK, Zubair H, Bhardwaj A, Tyagi N, Al-Ghadhban A, et al. Resistin Potentiates Chemoresistance and Stemness of Breast Cancer Cells: Implications for Racially Disparate Therapeutic Outcomes. Cancer Lett, 2017, 396:21–9., DOI 10.1016/j.canlet.2017.03.010 Mohammadi M, Hedayati M, Zarghami N, and Ghaemmaghami S. RESISTIN EFFECT ON TELOMERASE GENE EXPRESSION IN GASTRIC CANCER CELL LINE AGS. Acta Endo, Buc,, 2016, 12(2): 145–9., DOI 10.4183/aeb.2016.145 Malvi P, Chaube B, Singh SV,Mohammad N, Vijayakumar MV, Singh S, et al. Elevated Circulatory Levels of Leptin and Resistin Impair Therapeutic Efficacy of Dacarbazine in Melanoma under Obese State. Cancer Metab, 2018, 6(1):2., DOI 10.1186/s40170-018-0176-5 Liu Z, Shi A, Song D, Han B, Zhang Z,Ma L, et al. Resistin Confers Resistance to Doxorubicin-Induced Apoptosis in Human Breast Cancer Cells through Autophagy Induction. Am J Cancer Res, 2017, 7(3):574–83. Kim JG, Kim EO, Jeong BR, Min YJ, Park JW, Kim ES, et al. Visfatin Stimulates Proliferation of MCF-7 Human Breast Cancer Cells. Mol Cell, 2010, 30(4):341–5., DOI 10.1007/s10059-010-0124-x Buldak RJ, Buldak L, Polaniak R, Kukla M, Birkner E, Kubina R, et al. Visfatin Affects Redox Adaptative Responses and Proliferation in Me45 Human Malignant Melanoma Cells: an In Vitro Study. Oncol Rep, 2013, 29(2):771–8. Zhao H, TangW, Chen X, Wang S, Wang X, Xu H, et al. The NAMPT/E2F2/ SIRT1 axis Promotes Proliferation and Inhibits P53-dependent Apoptosis in Human Melanoma Cells. Biochem Biophysical Res Commun, 2017, 493(1): 77–84., DOI 10.1016/j.bbrc.2017.09.071 Adya R, Tan BK, Punn A, Chen J, and Randeva HS. Visfatin Induces Human Endothelial VEGF and MMP-2/9 Production via MAPK and PI3K/Akt Signalling Pathways: Novel Insights into Visfatin-Induced Angiogenesis. Cardiovasc Res, 2008, 78(2):356–65., DOI 10.1093/cvr/cvm111 Wang P, Xu T-Y, Guan Y-F, Su D-F, Fan G-R, and Miao C-Y. Perivascular Adipose Tissue-Derived Visfatin Is a Vascular Smooth Muscle Cell Growth Factor: Role of Nicotinamide Mononucleotide. Cardiovasc Res, 2009, 81(2): 370–80., DOI 10.1093/cvr/cvn288 Kim S-R, Bae S-K, Choi K-S, Park S-Y, Jun HO, Lee J-Y, et al. Visfatin Promotes Angiogenesis by Activation of Extracellular Signal-Regulated Kinase 1/2. Biochem Biophysical Res Commun, 2007, 357(1):150–6., DOI 10.1016/j.bbrc.2007.03.105 Liu P, Li H, Cepeda J, Xia Y, Kempf JA, Ye H, et al. Regulation of Inflammatory Cytokine Expression in Pulmonary Epithelial Cells by Pre- B-cell colony-enhancing Factor via a Nonenzymatic and AP-1-dependent Mechanism. J Biol Chem, 2009, 284(40):27344–51., DOI 10.1074/ jbc.m109.002519 Nowell MA, Richards PJ, Fielding CA, Ognjanovic S, Topley N, Williams AS, et al. Regulation of Pre-B Cell colony-enhancing Factor by STAT-3- dependent Interleukin-6trans-Signaling: Implications in the Pathogenesis of Rheumatoid Arthritis. Arthritis Rheum, 2006, 54(7):2084–95., DOI 10.1002/art.21942 Park H-J, Kim S-R, Kim SS, Wee H-J, Bae M-K, Ryu MH, et al. Visfatin Promotes Cell and Tumor Growth by Upregulating Notch1 in Breast Cancer. Oncotarget, 2014, 5(13):5087–99., DOI 10.18632/oncotarget.2086 Lu G-W, Wang Q-J, Xia M-M, and Qian J. Elevated Plasma Visfatin Levels Correlate with Poor Prognosis of Gastric Cancer Patients. Peptides, 2014, 58: 60–4., DOI 10.1016/j.peptides.2014.05.016 Yang J, Zhang K, Song H, Wu M, Li J, Yong Z, et al. Visfatin Is Involved in Promotion of Colorectal Carcinoma Malignancy through an Inducing EMT Mechanism. Oncotarget, 2016, 7(22):32306–17., DOI 10.18632/ oncotarget.8615 Liu T, Miao Z, Jiang J, Yuan S, Fang W, Li B, et al. Visfatin Mediates SCLC Cells Migration across Brain Endothelial Cells through Upregulation of CCL2. Int J Mol Sci, 2015, 16(5):11439–51., DOI 10.3390/ijms160511439 Lim SY, Yuzhalin AE, Gordon-Weeks AN, and Muschel RJ. Targeting the CCL2-CCR2 Signaling axis in Cancer Metastasis. Oncotarget, 2016, 7(19): 28697–710., DOI 10.18632/oncotarget.7376 Wang G-J, Shen N-J, Cheng L, Yehan Fang F, Huang H, and Li K-H. Visfatin Triggers the In Vitro Migration of Osteosarcoma Cells via Activation of NF- κB/IL-6 Signals. Eur J Pharmacol, 2016, 791:322–30., DOI 10.1016/ j.ejphar.2016.08.029 Cao Z, Liang N, Yang H, and Li S. Visfatin Mediates Doxorubicin Resistance in Human Non-small-cell Lung Cancer via Akt-Mediated Up-Regulation of ABCC1. Cel Prolif, 2017, 50(5):e12366., DOI 10.1111/cpr.12366 Kay-Pong Y, Chi LA-Y, and Samuel CM. Interactions of Omentin and Lactotransferrin in the Progression of Metastatic Ovarian Cancer. FASEB J, 2019, 33(1_Suppl. ment):704.5. Li D, Zhao X, Xiao Y, Mei H, Pu J, Xiang X, et al. Intelectin 1 Suppresses Tumor Progression and Is Associated with Improved Survival in Gastric Cancer. Oncotarget, 2015, 6(18):16168–82., DOI 10.18632/oncotarget.3753 Yeung CLA, Co NN, Onstad M, Yeung T-L, Leung CS, Schmandt R, et al. Abstract 4887: Omentin: A Novel Adipokine in the Omental Microenvironment Associated with Ovarian Cancer Progression. Cancer Res, 2014, 74(19 Suppl. ment):4887. Kenny HA, Chiang C-Y, White EA, Schryver EM, Habis M, Romero IL, et al. Mesothelial Cells Promote Early Ovarian Cancer Metastasis through Fibronectin Secretion. J Clin Invest, 2014, 124(10):4614–28., DOI 10.1172/ jci74778 Hiyoshi M, Tsuno NH, Otani K, Kawai K, Nishikawa T, Shuno Y, et al. Adiponectin Receptor 2 Is Negatively Associated with Lymph Node Metastasis of Colorectal Cancer. Oncol Lett, 2012, 3(4):756–60., DOI 10.3892/ol.2012.583 Taliaferro-Smith L, Nagalingam A, Zhong D, Zhou W, Saxena NK, and Sharma D. LKB1 Is Required for Adiponectin-Mediated Modulation of AMPK-S6k axis and Inhibition of Migration and Invasion of Breast Cancer Cells. Oncogene, 2009, 28(29):2621–33., DOI 10.1038/onc.2009.129 Corradetti MN, Inoki K, Bardeesy N, DePinho RA, and Guan K-L. Regulation of the TSC Pathway by LKB1: Evidence of a Molecular Link between Tuberous Sclerosis Complex and Peutz-Jeghers Syndrome. Genes Develop, 2004, 18(13):1533–8., DOI 10.1101/gad.1199104 Gwinn DM, Shackelford DB, Egan DF,Mihaylova MM, Mery A, Vasquez DS, et al. AMPK Phosphorylation of Raptor Mediates a Metabolic Checkpoint. Mol Cel, 2008, 30(2):214–26., DOI 10.1016/j.molcel.2008.03.003 Amaral CL, Freitas LB, Tamura RE, Tavares MR, Pavan ICB, Bajgelman MC, et al. S6Ks Isoforms Contribute to Viability, Migration, Docetaxel Resistance and Tumor Formation of Prostate Cancer Cells. BMC cancer, 2016, 16:602., DOI 10.1186/s12885-016-2629-y Zhang S, Hu B, Lv X, Chen S, Liu W, and Shao Z. The Prognostic Role of Ribosomal Protein S6 Kinase 1 Pathway in Patients with Solid Tumors: A Meta-Analysis. Front Oncol, 2019, 9(390)., DOI 10.3389/fonc.2019.00390 Mauro L, Pellegrino M, Giordano F, Ricchio E, Rizza P, De Amicis F, et al. Estrogen Receptor-α Drives Adiponectin Effects on Cyclin D1 Expression in Breast Cancer Cells. FASEB j., 2015, 29(5):2150–60., DOI 10.1096/fj.14-262808 Wang Y, Lam JB, Lam KSL, Liu J, Lam MC, Hoo RLC, et al. Adiponectin Modulates the Glycogen Synthase Kinase-3β/β-Catenin Signaling Pathway and Attenuates Mammary Tumorigenesis of MDA-MB-231 Cells in Nude Mice. Cancer Res, 2006, 66(23):11462–70., DOI 10.1158/0008-5472.can-06-1969 Miyazaki T, Bub JD, Uzuki M, and Iwamoto Y. Adiponectin Activates C-Jun NH2-terminal Kinase and Inhibits Signal Transducer and Activator of Transcription 3. Biochem Biophysical Res Commun, 2005, 333(1):79–87., DOI 10.1016/j.bbrc.2005.05.076 Partida-Perez M, de la Luz Ayala-Madrigal M, Peregrina-Sandoval J, Macias- Gomez N, Moreno-Ortiz J, Leal-Ugarte E, et al. Association of LEP and ADIPOQ Common Variants with Colorectal Cancer in Mexican Patients. Cancer Biomark, 2010, 7(3):117–21., DOI 10.3233/CBM-2010-0154 Li G, Cong L, Gasser J, Zhao J, Chen K, and Li F.Mechanisms Underlying the Anti-proliferative Actions of Adiponectin in Human Breast Cancer Cells, MCF7-Dependency on the cAMP/protein Kinase-A Pathway. Nutr Cancer, 2011, 63(1):80–8., DOI 10.1080/01635581.2010.516472 Korner A, Pazaitou-Panayiotou K, Kelesidis T, Kelesidis I, Williams CJ, Kaprara A, et al. Total and High-Molecular-Weight Adiponectin in Breast Cancer: In Vitro and In Vivo Studies. J Clin Endocrinol Metab, 2007, 92(3): 1041–8., DOI 10.1210/jc.2006-1858 Cong L, Gasser J, Zhao J, Yang B, Li F, and Zhao AZ. Human Adiponectin Inhibits Cell Growth and Induces Apoptosis in Human Endometrial Carcinoma Cells, HEC-1-A and RL95-2. Endocr Relat Cancer, 2007, 14(3):713–20., DOI 10.1677/erc-07-0065 Dieudonne M-N, Bussiere M, Dos Santos E, Leneveu M-C, Giudicelli Y, and Pecquery R. Adiponectin Mediates Antiproliferative and Apoptotic Responses in Human MCF7 Breast Cancer Cells. Biochem Biophysical Res Commun, 2006, 345(1):271–9., DOI 10.1016/j.bbrc.2006.04.076 Zakikhani M, Dowling RJO, Sonenberg N, and Pollak MN. The Effects of Adiponectin and Metformin on Prostate and colon Neoplasia Involve Activation of AMP-Activated Protein Kinase. Cancer Prev Res, 2008, 1(5): 369–75., DOI 10.1158/1940-6207.capr-08-0081 Fenton JI, Birmingham JM, Hursting SD, and Hord NG. Adiponectin Blocks Multiple Signaling Cascades Associated with Leptin-Induced Cell Proliferation inApcMin/+ colon Epithelial Cells. Int J Cancer, 2008, 122(11):2437–45., DOI 10.1002/ijc.23436 Jarde T, Perrier S, Vasson M-P, and Caldefie-Chezet F. Molecular Mechanisms of Leptin and Adiponectin in Breast Cancer. Eur J Cancer, 2011, 47(1):33–43., DOI 10.1016/j.ejca.2010.09.005 Luo Z, Saha AK, Xiang X, and Ruderman NB. AMPK, the Metabolic Syndrome and Cancer. Trends Pharmacol Sci, 2005, 26(2):69–76., DOI 10.1016/j.tips.2004.12.011 Kim AY, Lee YS, Kim KH, Lee JH, Lee HK, Jang S-H, et al. Adiponectin Represses Colon Cancer Cell Proliferation via AdipoR1- and -R2-Mediated AMPK Activation. Mol Endocrinol, 2010, 24(7):1441–52., DOI 10.1210/ me.2009-0498 Kitajima D, Kasamatsu A, Nakashima D, Miyamoto I, Kimura Y, Saito T, et al. Tie2 Regulates Tumor Metastasis of Oral Squamous Cell Carcinomas. J Cancer, 2016, 7(5):600–7., DOI 10.7150/jca.13820 KitajimaD, KasamatsuA,NakashimaD,Miyamoto I, Kimura Y, Endo-Sakamoto Y, et al. Evidence for Critical Role of Tie2/Ang1 Interaction in Metastatic Oral Cancer. Oncol Lett, 2018, 15(5):7237–42., DOI 10.3892/ol.2018.8212 Michael IP, Orebrand M, Lima M, Pereira B, Volpert O, Quaggin SE, et al. Angiopoietin-1 Deficiency Increases Tumor Metastasis in Mice. BMC cancer, 2017, 17(1):539., DOI 10.1186/s12885-017-3531-y Wu FT, Lee CR, Bogdanovic E, Prodeus A, Gariepy J, and Kerbel RS. Vasculotide Reduces Endothelial Permeability and Tumor Cell Extravasation in the Absence of Binding to or Agonistic Activation of Tie2. EMBO Mol Med, 2015, 7(6):770–87., DOI 10.15252/emmm.201404193 Ou X-L, Chen H-J, Sun W-H, Hang C, Yang L, Guan Y-Y, et al. Effects of Angiopoietin-1 on Attachment and Metastasis Ofhuman Gastric Cancer Cell Line BGC-823. Wjg, 2009, 15(43):5432–41., DOI 10.3748/wjg.15.5432 Holopainen T, Huang H, Chen C, Kim KE, Zhang L, Zhou F, et al. Angiopoietin-1 Overexpression Modulates Vascular Endothelium to Facilitate Tumor Cell Dissemination and Metastasis Establishment. Cancer Res, 2009, 69(11):4656–64., DOI 10.1158/0008-5472.can-08-4654 Xu Y, Liu Y-j., and Yu Q. Angiopoietin-3 Inhibits Pulmonary Metastasis by Inhibiting Tumor Angiogenesis. Cancer Res, 2004, 64(17):6119–26., DOI 10.1158/0008-5472.can-04-1054 Imanishi Y, Hu B, Jarzynka MJ, Guo P, Elishaev E, Bar-Joseph I, et al. Angiopoietin-2 Stimulates Breast Cancer Metastasis through the α5β1 Integrin-Mediated Pathway. Cancer Res, 2007, 67(9):4254–63., DOI 10.1158/0008-5472.can-06-4100 Li C, Li Q, Cai Y,He Y, Lan X, Wang W, et al. Overexpression of Angiopoietin 2 Promotes the Formation of Oral Squamous Cell Carcinoma by Increasing Epithelial-Mesenchymal Transition-Induced Angiogenesis. Cancer Gene Ther, 2016, 23(9):295–302., DOI 10.1038/cgt.2016.30 Han HH, Kim BG, Lee JH, Kang S, Kim JE, and Cho NH. Angiopoietin-2 Promotes ER+ Breast Cancer Cell Survival in Bone Marrow Niche. Endocr Relat Cancer, 2016, 23(8):609–23., DOI 10.1530/erc-16-0086 Sasaki H, Suzuki A, Shitara M, Hikosaka Y, Okuda K, Moriyama S, et al. Angiopoietin-like Protein ANGPTL2 Gene Expression Is Correlated with Lymph Node Metastasis in Lung Cancer. Oncol Lett, 2012, 4(6):1325–8., DOI 10.3892/ol.2012.924 Aoi J, Endo M, Kadomatsu T, Miyata K, Nakano M, Horiguchi H, et al. Angiopoietin-like Protein 2 Is an Important Facilitator of Inflammatory Carcinogenesis and Metastasis. Cancer Res, 2011, 71(24):7502–12., DOI 10.1158/0008-5472.can-11-1758 Endo M, Nakano M, Kadomatsu T, Fukuhara S, Kuroda H, Mikami S, et al. Tumor Cell-Derived Angiopoietin-like Protein ANGPTL2 Is a Critical Driver of Metastasis. Cancer Res, 2012, 72(7):1784–94., DOI 10.1158/0008-5472.can- 11-3878 Wang X, Hu Z, Wang Z, Cui Y, and Cui X. Angiopoietin-like Protein 2 Is an Important Facilitator of Tumor Proliferation, Metastasis, Angiogenesis and Glycolysis in Osteosarcoma. Am J Transl Res, 2019, 11(10):6341–55. Odagiri H, Kadomatsu T, Endo M, Masuda T, Morioka MS, Fukuhara S, et al. The Secreted Protein ANGPTL2 Promotes Metastasis of Osteosarcoma Cells through Integrin 5 1, P38 MAPK, and Matrix Metalloproteinases. Sci Signaling, 2014, 7(309):ra7., DOI 10.1126/scisignal.2004612 Galaup A, Cazes A, Le Jan S, Philippe J, Connault E, Le Coz E, et al. Angiopoietin-like 4 Prevents Metastasis through Inhibition of Vascular Permeability and Tumor Cell Motility and Invasiveness. Proc Natl Acad Sci, 2006, 103(49):18721–6., DOI 10.1073/pnas.0609025103 Zhang H, Wong CCL, Wei H, Gilkes DM, Korangath P, Chaturvedi P, et al. HIF-1-dependent Expression of Angiopoietin-like 4 and L1CAM Mediates Vascular Metastasis of Hypoxic Breast Cancer Cells to the Lungs. Oncogene, 2012, 31(14):1757–70., DOI 10.1038/onc.2011.365 Padua D, Zhang XH-F, Wang Q, Nadal C, Gerald WL, Gomis RR, et al. TGFβ Primes Breast Tumors for Lung Metastasis Seeding through Angiopoietinlike 4. Cell, 2008, 133(1):66–77., DOI 10.1016/j.cell.2008.01.046 Marchio S, Soster M, Cardaci S, Muratore A, Bartolini A, Barone V, et al. A Complex of α 6 Integrin and E-cadherin Drives Liver Metastasis of Colorectal Cancer Cells through Hepatic Angiopoietin-like 6. EMBO Mol Med, 2012, 4(11):1156–75., DOI 10.1002/emmm.201101164 Chu SH, Lee MK, Ahn KY, Im JA, Park MS, Lee DC, et al. Chemerin and Adiponectin Contribute Reciprocally to Metabolic Syndrome. PLoS One, 2012, 7(4):e34710., DOI 10.1371/journal.pone.0034710 Li J-J, Yin H-K, Guan D-X, Zhao J-S, Feng Y-X, Deng Y-Z, et al. Chemerin Suppresses Hepatocellular Carcinoma Metastasis through CMKLR1-PTENAkt axis. Br J Cancer, 2018, 118(10):1337–48., DOI 10.1038/s41416-018- 0077-y Liu-Chittenden Y, Jain M, Gaskins K, Wang S, Merino MJ, Kotian S, et al. RARRES2 Functions as a Tumor Suppressor by Promoting β-catenin Phosphorylation/degradation and Inhibiting P38 Phosphorylation in Adrenocortical Carcinoma. Oncogene, 2017, 36(25):3541–52., DOI 10.1038/ onc.2016.497 Zhang J, Zhou J, Tang X, Zhou L-Y, Zhai L-L, Vanessa ME-D, et al. Reduced Expression of Chemerin Is Associated with Poor Clinical Outcome in Acute Myeloid Leukemia. Oncotarget, 2017, 8(54):92536–44., DOI 10.18632/ oncotarget.21440 Xu C-H, Yang Y, Wang Y-C, Yan J, and Qian L-H. Prognostic Significance of Serum Chemerin Levels in Patients with Non-small Cell Lung Cancer. Oncotarget, 2017, 8(14):22483–9., DOI 10.18632/oncotarget.14956 Zhang J, Jin H-C, Zhu A-K, Ying R-C, Wei W, and Zhang F-J. Prognostic Significance of Plasma Chemerin Levels in Patients with Gastric Cancer. Peptides, 2014, 61:7–11., DOI 10.1016/j.peptides.2014.08.007 El-Sagheer G, Gayyed M, Ahmad A, Abd El-Fattah A, and Mohamed M. Expression of Chemerin Correlates with a Poor Prognosis in Female Breast Cancer Patients. Breast Cancer, Dove Med Press,, 2018, 10:169–76., DOI 10.2147/bctt.s178181 De Henau O, Degroot G-N, Imbault V, Robert V, De Poorter C, McHeik S, et al. Signaling Properties of Chemerin Receptors CMKLR1, GPR1 and CCRL2. PloS one, 2016, 11(10):e0164179., DOI 10.1371/journal.pone.0164179 Duan X, Kong Z, Liu Y, Zeng Z, Li S, Wu W, et al. β-Arrestin2 Contributes to Cell Viability and Proliferation via the Down-Regulation of FOXO1 in Castration-Resistant Prostate Cancer. J Cel Physiol., 2015, 230(10): 2371–81., DOI 10.1002/jcp.24963 Ge L, Shenoy SK, Lefkowitz RJ, and DeFea K. Constitutive Protease-Activated Receptor-2-Mediated Migration of MDA MB-231 Breast Cancer Cells Requires Both β-Arrestin-1 and -2. J Biol Chem, 2004, 279(53):55419–24., DOI 10.1074/jbc.m410312200 Lakshmikanthan V, Zou L, Kim JI, Michal A, Nie Z, Messias NC, et al. Identification of Arrestin2 as a Corepressor of Androgen Receptor Signaling in Prostate Cancer. Proc Natl Acad Sci, 2009, 106(23):9379–84., DOI 10.1073/ pnas.0900258106 Cong L, Qiu Z-y., Zhao Y, Wang W-b., Wang C-x., Shen H-c., et al. Loss of β-arrestin-2 and Activation of CXCR2 Correlate with Lymph Node Metastasis in Non-small Cell Lung Cancer. J Cancer, 2017, 8(14): 2785–92., DOI 10.7150/jca.19631 Sun W-Y, Hu S-S, Wu J-J, Huang Q, Ma Y, Wang Q-T, et al. Down-regulation of β-arrestin2 Promotes Tumour Invasion and Indicates Poor Prognosis of Hepatocellular Carcinoma. Scientific Rep, 2016, 6(1):35609., DOI 10.1038/ srep35609 DeFea KA, Zalevsky J, Thoma MS, Dery O, Mullins RD, and Bunnett NW. β-Arrestin-Dependent Endocytosis of Proteinase-Activated Receptor 2 Is Required for Intracellular Targeting of Activated Erk1/2. J Cel Biol, 2000, 148(6):1267–82., DOI 10.1083/jcb.148.6.1267 Rourke JL, Dranse HJ, and Sinal CJ. CMKLR1 and GPR1 Mediate Chemerin Signaling through the RhoA/ROCK Pathway. Mol Cell Endocrinol, 2015, 417: 36–51., DOI 10.1016/j.mce.2015.09.002 Yin J, Lv X, Hu S, Zhao X, Liu Q, and Xie H. Overexpression of Serum Response Factor Is Correlated with Poor Prognosis in Patients with Gastric Cancer. Hum Pathol, 2019, 85:10–7., DOI 10.1016/j.humpath.2018.10.018 O’Hurley G, Prencipe M, Lundon D, O’Neill A, Boyce S, O’Grady A, et al. The Analysis of Serum Response Factor Expression in Bone and Soft Tissue Prostate Cancer Metastases. Prostate, 2014, 74(3):306–13., DOI 10.1002/ pros.22752 Cen B, Selvaraj A, Burgess RC, Hitzler JK, Ma Z, Morris SW, et al. Megakaryoblastic Leukemia 1, a Potent Transcriptional Coactivator for Serum Response Factor, SRF), Is Required for Serum Induction of SRF Target Genes. Mol Cel Biol, 2003, 23(18):6597–608., DOI 10.1128/ mcb.23.18.6597-6608.2003 Selvaraj A, and Prywes R. Expression Profiling of Serum Inducible Genes Identifies a Subset of SRF Target Genes that Are MKL Dependent. BMC Mol Biol, 2004, 5(1):13., DOI 10.1186/1471-2199-5-13 Nair P, Muthukkumar S, Sells SF, Han S-S, Sukhatme VP, and Rangnekar VM. Early Growth Response-1-dependent Apoptosis Is Mediated by P53. J Biol Chem, 1997, 272(32):20131–8., DOI 10.1074/jbc.272.32.20131 Krones-Herzig A, Adamson E, and Mercola D. Early Growth Response 1 Protein, an Upstream Gatekeeper of the P53 Tumor Suppressor, Controls Replicative Senescence. Proc Natl Acad Sci, 2003, 100(6):3233–8., DOI 10.1073/pnas.2628034100 Li L, Huang C, Zhang X, Wang J, Ma P, Liu Y, et al. Chemerin-derived Peptide C-20 Suppressed Gonadal Steroidogenesis. Am J Reprod Immunol, 2014, 71(3):265–77., DOI 10.1111/aji.12164 Muruganandan S, Dranse HJ, Rourke JL, McMullen NM, and Sinal CJ. Chemerin Neutralization Blocks Hematopoietic Stem Cell Osteoclastogenesis. Stem Cells, 2013, 31(10):2172–82., DOI 10.1002/stem.1450 Guo JC, Li J, Zhao YP, Zhou L, Cui QC, Zhou WX, et al. Expression of C-Fos Was Associated with Clinicopathologic Characteristics and Prognosis in Pancreatic Cancer. PLoS One, 2015, 10(3):e0120332., DOI 10.1371/ journal.pone.0120332 Lu C, Shen Q, DuPre E, Kim H, Hilsenbeck S, and Brown PH. cFos Is Critical for MCF-7 Breast Cancer Cell Growth. Oncogene, 2005, 24(43):6516–24., DOI 10.1038/sj.onc.1208905 Mikula M, Gotzmann J, Fischer ANM, Wolschek MF, Thallinger C, Schulte- Hermann R, et al. The Proto-Oncoprotein C-Fos Negatively Regulates Hepatocellular Tumorigenesis. Oncogene, 2003, 22(43):6725–38., DOI 10.1038/sj.onc.1206781 Mahner S, Baasch C, Schwarz J, Hein S, Wolber L, Janicke F, et al. C-fos Expression Is a Molecular Predictor of Progression and Survival in Epithelial Ovarian Carcinoma. Br J Cancer, 2008, 99(8):1269–75., DOI 10.1038/ sj.bjc.6604650 Oliveira-Ferrer L, Roßler K, Haustein V, Schroder C, Wicklein D, Maltseva D, et al. c-FOS Suppresses Ovarian Cancer Progression by Changing Adhesion. Br J Cancer, 2014, 110(3):753–63., DOI 10.1038/bjc.2013.774 Zhang X, Zhang L, Yang H, Huang X, Otu H, Libermann TA, et al. c-Fos as a Proapoptotic Agent in TRAIL-Induced Apoptosis in Prostate Cancer Cells. Cancer Res, 2007, 67(19):9425–34., DOI 10.1158/0008-5472.can-07-1310 Li T, Guo H, Song Y, Zhao X, Shi Y, Lu Y, et al. Loss of Vinculin and Membrane-Bound β-catenin Promotes Metastasis and Predicts Poor Prognosis in Colorectal Cancer. Mol Cancer, 2014, 13(1):263., DOI 10.1186/ 1476-4598-13-263 Zhang M, Liu P, Xu F, He Y, Xie X, and Jiang X. Retracted : Vinculin Promotes Gastric Cancer Proliferation and Migration and Predicts Poor Prognosis in Patients with Gastric Cancer. J Cel Biochem, 2019, 120(8): 14107–15., DOI 10.1002/jcb.28686 Pan Y, Zhou F, He C, Hui L, Huang T, and Wei Y. Leptin-LepRb Expressed in Gastric Cancer Patients and Related to Cancer-Related Depression. Biomed Res Int, 2017, 2017:6482842., DOI 10.1155/2017/6482842 Cheng H, Wang Z, Fu L, and Xu T. Macrophage Polarization in the Development and Progression of Ovarian Cancers: An Overview. Front Oncol, 2019, 9:421., DOI 10.3389/fonc.2019.00421 Yafei Z, Jun G, and Guolan G. Correlation between Macrophage Infiltration and Prognosis of Ovarian Cancer-A Preliminary Study. Biomed Res, 2016, 27:305–12. Tian Z, Hou X, Liu W, Han Z, and Wei L. Macrophages and Hepatocellular Carcinoma. Cel Biosci, 2019, 9(1):79., DOI 10.1186/s13578-019-0342-7 Jeong H, Hwang I, Kang SH, Shin HC, and Kwon SY. Tumor-Associated Macrophages as Potential Prognostic Biomarkers of Invasive Breast Cancer. J Breast Cancer, 2019, 22(1):38–51., DOI 10.4048/jbc.2019.22.e5 Lee YS, Song SJ, Hong HK, Oh BY, Lee WY, and Cho YB. The FBW7-MCL-1 axis Is Key in M1 and M2 Macrophage-Related colon Cancer Cell Progression: Validating the Immunotherapeutic Value of Targeting PI3Kγ. Exp Mol Med, 2020, 52(5):815–31., DOI 10.1038/s12276-020-0436-7 Herova M, Schmid M, Gemperle C, and Hersberger M. ChemR23, the Receptor for Chemerin and Resolvin E1, Is Expressed and Functional on M1 but Not on M2 Macrophages. J.I., 2015, 194(5):2330–7., DOI 10.4049/ jimmunol.1402166 Pachynski RK, Zabel BA, Kohrt HE, Tejeda NM, Monnier J, Swanson CD, et al. The Chemoattractant Chemerin Suppresses Melanoma by Recruiting Natural Killer Cell Antitumor Defenses. J Exp Med, 2012, 209(8):1427–35., DOI 10.1084/jem.20112124 Dimitriadis GK, Kaur J, Adya R, Miras AD, Mattu HS, Hattersley JG, et al. Chemerin Induces Endothelial Cell Inflammation: Activation of Nuclear Factor-Kappa Beta and Monocyte-Endothelial Adhesion. Oncotarget, 2018, 9(24):16678–90., DOI 10.18632/oncotarget.24659 Zylla S, Pietzner M, Kuhn J-P, Volzke H, Dorr M, Nauck M, et al. Serum Chemerin Is Associated with Inflammatory and Metabolic Parameters- Results of a Population-Based Study. Obesity, 2017, 25(2):468–75., DOI 10.1002/oby.21735 Liu S, Hempe JM, McCarter RJ, Li S, and Fonseca VA. Association between Inflammation and Biological Variation in Hemoglobin A1c in U.S. Nondiabetic Adults. J Clin Endocrinol Metab, 2015, 100(6):2364–71., DOI 10.1210/jc.2014-4454 Kaur J, Adya R, Tan BK, Chen J, and Randeva HS. Identification of Chemerin Receptor, ChemR23, in Human Endothelial Cells: Chemerin-Induced Endothelial Angiogenesis. Biochem Biophysical Res Commun, 2010, 391(4):1762–8., DOI 10.1016/j.bbrc.2009.12.150 Bozaoglu K, Curran JE, Stocker CJ, Zaibi MS, Segal D, Konstantopoulos N, et al. Chemerin, a Novel Adipokine in the Regulation of Angiogenesis. J Clin Endocrinol Metab, 2010, 95(5):2476–85., DOI 10.1210/jc.2010-0042 Wang C, Wu WKK, Liu X, To K-F, Chen GG, Yu J, et al. Increased Serum Chemerin Level Promotes Cellular Invasiveness in Gastric Cancer: a Clinical and Experimental Study. Peptides, 2014, 51:131–8., DOI 10.1016/ j.peptides.2013.10.009 Nakamura N, Naruse K, Kobayashi Y, Miyabe M, Saiki T, Enomoto A, et al. Chemerin Promotes Angiogenesis In Vivo. Physiol Rep, 2018, 6(24):e13962., DOI 10.14814/phy2.13962 Skrzeczynska-Moncznik J, Wawro K, Stefanska A, Oleszycka E, Kulig P, Zabel BA, et al. Potential Role of Chemerin in Recruitment of Plasmacytoid Dendritic Cells to Diseased Skin. Biochem biophysical Res Commun, 2009, 380:323–7. Bidwell BN, Slaney CY, Withana NP, Forster S, Cao Y, Loi S, et al. Silencing of Irf7 Pathways in Breast Cancer Cells Promotes Bone Metastasis through Immune Escape. Nat Med, 2012, 18(8):1224–31., DOI 10.1038/nm.2830 Bekeredjian-Ding I, Schafer M, Hartmann E, Pries R, Parcina M, Schneider P, et al. Tumour-derived Prostaglandin E2and Transforming Growth Factor-β Synergize to Inhibit Plasmacytoid Dendritic Cell-Derived Interferon-α. Immunology, 2009, 128(3):439–50., DOI 10.1111/j.1365-2567.2009.03134.x Hartmann E,Wollenberg B, Rothenfusser S, Wagner M, Wellisch D, Mack B, et al. Identification and Functional Analysis of Tumor-Infiltrating Plasmacytoid Dendritic Cells in Head and Neck Cancer. Cancer Res, 2003, 63(19):6478–87. Auguet T, Quintero Y, Terra X, Martinez S, Lucas A, Pellitero S, et al. Upregulation of Lipocalin 2 in Adipose Tissues of Severely Obese Women: Positive Relationship with Proinflammatory Cytokines. Obesity, Silver Spring,, 2011, 19(12):2295–300., DOI 10.1038/oby.2011.61 Lee H-J, Lee E-K, Lee K-J, Hong S-W, Yoon Y, and Kim J-S. Ectopic Expression of Neutrophil Gelatinase-Associated Lipocalin Suppresses the Invasion and Liver Metastasis of colon Cancer Cells. Int J Cancer, 2006, 118(10):2490–7., DOI 10.1002/ijc.21657 Feng M, Feng J, Chen W, Wang W, Wu X, Zhang J, et al. Lipocalin2 Suppresses Metastasis of Colorectal Cancer by Attenuating NF-κb-dependent Activation of Snail and Epithelial Mesenchymal Transition. Mol Cancer, 2016, 15(1):77., DOI 10.1186/s12943-016-0564-9 Moniaux N, Chakraborty S, Yalniz M, Gonzalez J, Shostrom VK, Standop J, et al. Early Diagnosis of Pancreatic Cancer: Neutrophil Gelatinase-Associated Lipocalin as a Marker of Pancreatic Intraepithelial Neoplasia. Br J Cancer, 2008, 98(9):1540–7., DOI 10.1038/sj.bjc.6604329 Yang J, Bielenberg DR, Rodig SJ, Doiron R, Clifton MC, Kung AL, et al. Lipocalin 2 Promotes Breast Cancer Progression. Proc Natl Acad Sci, 2009, 106(10):3913–8., DOI 10.1073/pnas.0810617106 Zhang Y, Lazarus J, Steele NG, Yan W, Lee H-J, Nwosu ZC, et al. Regulatory T-Cell Depletion Alters the Tumor Microenvironment and Accelerates Pancreatic Carcinogenesis. Cancer Discov, 2020, 10(3):422–39., DOI 10.1158/2159-8290.cd-19-0958 Karlsen JR, Borregaard N, and Cowland JB. Induction of Neutrophil Gelatinase-Associated Lipocalin Expression by Co-stimulation with Interleukin-17 and Tumor Necrosis Factor-α Is Controlled by IκB-ζ but Neither by C/EBP-β Nor C/EBP-δ. J Biol Chem, 2010, 285(19):14088–100., DOI 10.1074/jbc.m109.017129 Wani NA, Nasser MW, Ahirwar DK, Zhao H, Miao Z, Shilo K, et al. C-X-C Motif Chemokine 12/C-X-C Chemokine Receptor Type 7 Signaling Regulates Breast Cancer Growth and Metastasis by Modulating the Tumor Microenvironment. Breast Cancer Res, 2014, 16(3):R54., DOI 10.1186/bcr3665 Gomez-Chou SB, Swidnicka-Siergiejko AK, Badi N, Chavez-Tomar M, Lesinski GB, Bekaii-Saab T, et al. Lipocalin-2 Promotes Pancreatic Ductal Adenocarcinoma by Regulating Inflammation in the Tumor Microenvironment. Cancer Res, 2017, 77(10):2647–60., DOI 10.1158/0008- 5472.can-16-1986 Tschesche H, Zolzer V, Triebel S, and Bartsch S. The Human Neutrophil Lipocalin Supports the Allosteric Activation of Matrix Metalloproteinases. Eur J Biochem, 2001, 268(7):1918–28., DOI 10.1046/j.1432-1327.2001.02066.x Kubben FJGM, Sier CFM, Hawinkels LJAC, Tschesche H, van Duijn W, Zuidwijk K, et al. Clinical Evidence for a Protective Role of Lipocalin-2 against MMP-9 Autodegradation and the Impact for Gastric Cancer. Eur J Cancer, 2007, 43(12):1869–76., DOI 10.1016/j.ejca.2007.05.013 Nuntagowat C, Leelawat K, and Tohtong R. NGAL Knockdown by siRNA in Human Cholangiocarcinoma Cells Suppressed Invasion by Reducing NGAL/ MMP-9 Complex Formation. Clin Exp Metastasis, 2010, 27(5):295–305., DOI 10.1007/s10585-010-9327-y Fernandez CA, Yan L, Louis G, Yang J, Kutok JL, and Moses MA. The Matrix Metalloproteinase-9/neutrophil Gelatinase-Associated Lipocalin Complex Plays a Role in Breast Tumor Growth and Is Present in the Urine of Breast Cancer Patients. Clin Cancer Res, 2005, 11(15):5390–5., DOI 10.1158/1078-0432.ccr-04-2391 Volpe V, Raia Z, Sanguigno L, Somma D, Mastrovito P, Moscato F, et al. NGAL Controls the Metastatic Potential of Anaplastic Thyroid Carcinoma Cells. J Clin Endocrinol Metab, 2013, 98(1):228–35., DOI 10.1210/jc.2012-2528 Roy R, Louis G, Loughlin KR, Wiederschain D, Kilroy SM, Lamb CC, et al. Tumor-specific Urinary Matrix Metalloproteinase Fingerprinting: Identification of High Molecular Weight Urinary Matrix Metalloproteinase Species. Clin Cancer Res, 2008, 14(20):6610–7., DOI 10.1158/1078-0432.ccr-08-1136 Leng X, Ding T, Lin H, Wang Y, Hu L, Hu J, et al. Inhibition of Lipocalin 2 Impairs Breast Tumorigenesis and Metastasis. Cancer Res, 2009, 69(22): 8579–84., DOI 10.1158/0008-5472.can-09-1934 Iannetti A, Pacifico F, Acquaviva R, Lavorgna A, Crescenzi E, Vascotto C, et al. The Neutrophil Gelatinase-Associated Lipocalin, NGAL), a NFB- Regulated Gene, Is a Survival Factor for Thyroid Neoplastic Cells. Proc Natl Acad Sci, 2008, 105(37):14058–63., DOI 10.1073/pnas.0710846105 Leung L, Radulovich N, Zhu CQ, Organ S, Bandarchi B, Pintilie M, et al. Lipocalin2 Promotes Invasion, Tumorigenicity and Gemcitabine Resistance in Pancreatic Ductal Adenocarcinoma. PLoS One, 2012, 7(10):e46677., DOI 10.1371/journal.pone.0046677 Hanai J-i., Mammoto T, Seth P, Mori K, Karumanchi SA, Barasch J, et al. Lipocalin 2 Diminishes Invasiveness and Metastasis of Ras-Transformed Cells. J Biol Chem, 2005, 280(14):13641–7., DOI 10.1074/jbc.m413047200 Shi H, Gu Y, Yang J, Xu L, Mi W, and Yu W. Lipocalin 2 Promotes Lung Metastasis of Murine Breast Cancer Cells. J Exp Clin Cancer Res, 2008, 27(1): 83., DOI 10.1186/1756-9966-27-83 Gu Y, Zhang J, Mi W, Yang J, Han F, Lu X, et al. Silencing of GM3 Synthase Suppresses Lung Metastasis of Murine Breast Cancer Cells. Breast Cancer Res, 2008, 10(1):R1., DOI 10.1186/bcr1841 Li B, Xu WW, Lam AKY, Wang Y, Hu H-F, Guan XY, et al. Significance of PI3K/AKT Signaling Pathway in Metastasis of Esophageal Squamous Cell Carcinoma and its Potential as a Target for Anti-metastasis Therapy. Oncotarget, 2017, 8(24):38755–66., DOI 10.18632/oncotarget.16333 Yue Y, Hui K, Wu S, Zhang M, Que T, Gu Y, et al. MUC15 Inhibits Cancer Metastasis via PI3K/AKT Signaling in Renal Cell Carcinoma. Cel Death Dis, 2020, 11(5):336., DOI 10.1038/s41419-020-2518-9 Liu H, Radisky DC, Nelson CM, Zhang H, Fata JE, Roth RA, et al. Mechanism of Akt1 Inhibition of Breast Cancer Cell Invasion Reveals a Protumorigenic Role for TSC2. Proc Natl Acad Sci, 2006, 103(11):4134–9., DOI 10.1073/ pnas.0511342103 Qin J-J, Li X, Wang W, Zi X, and Zhang R. Targeting the NFAT1-MDM2- MDMX Network Inhibits the Proliferation and Invasion of Prostate Cancer Cells, Independent of P53 and Androgen. Front Pharmacol, 2017, 8:917., DOI 10.3389/fphar.2017.00917 Bao G, Clifton M, Hoette TM, Mori K, Deng S-X, Qiu A, et al. Iron Traffics in Circulation Bound to a Siderocalin, Ngal)-Catechol Complex. Nat Chem Biol, 2010, 6(8):602–9., DOI 10.1038/nchembio.402 Schafer ZT, Grassian AR, Song L, Jiang Z, Gerhart-Hines Z, Irie HY, et al. Antioxidant and Oncogene rescue of Metabolic Defects Caused by Loss of Matrix Attachment. Nature, 2009, 461(7260):109–13., DOI 10.1038/ nature08268 Devireddy LR, Gazin C, Zhu X, and Green MR. A Cell-Surface Receptor for Lipocalin 24p3 Selectively Mediates Apoptosis and Iron Uptake. Cell, 2005, 123(7):1293–305., DOI 10.1016/j.cell.2005.10.027 Zhang S, Chen Y, GuoW, Yuan L, Zhang D, Xu Y, et al. Disordered Hepcidin- Ferroportin Signaling Promotes Breast Cancer Growth. Cell Signal, 2014, 26(11):2539–50., DOI 10.1016/j.cellsig.2014.07.029 Deng Y, Wang ZV, Gordillo R, An Y, Zhang C, Liang Q, et al. An Adipo- Biliary-Uridine axis that Regulates Energy Homeostasis. Science, 2017, 355(6330)., DOI 10.1126/science.aaf5375 Kohli R, Bhattacharjee J, and Inge TH. Postprandial Uridine Physiology Is Altered by Obesity. Gastroenterology, 2018, 155(5):1645–6., DOI 10.1053/ j.gastro.2018.07.043 Placet M, Arguin G, Molle CM, Babeu J-P, Jones C, Carrier JC, et al. The G Protein-Coupled P2Y6 Receptor Promotes Colorectal Cancer Tumorigenesis by Inhibiting Apoptosis. Biochim Biophys Acta, Bba, - Mol Basis Dis, 2018, 1864(5):1539–51., DOI 10.1016/j.bbadis.2018.02.008 Qiu Y, Liu Y, Li WH, Zhang HQ, Tian XX, and Fang WG. P2Y2 Receptor Promotes the Migration and Invasion of Breast Cancer Cells via EMT-Related Genes Snail and E-Cadherin. Oncol Rep, 2018, 39(1):138–50., DOI 10.3892/ or.2017.6081 Ma X, Pan X, Wei Y, Tan B, Yang L, Ren H, et al. Chemotherapy-induced Uridine Diphosphate Release Promotes Breast Cancer Metastasis through P2Y6 Activation. Oncotarget, 2016, 7(20):29036–50., DOI 10.18632/ oncotarget.8664 Miyashita H, Takebayashi Y, Eliason JF, Fujimori F, Nitta Y, Sato A, et al. Uridine Phosphorylase Is a Potential Prognostic Factor in Patients with Oral Squamous Cell Carcinoma. Cancer, 2002, 94(11):2959–66., DOI 10.1002/ cncr.10568 Shen G, He P, Mao Y, Li P, Luh F, Ding G, et al. Overexpression of Uridine- Cytidine Kinase 2 Correlates with Breast Cancer Progression and Poor Prognosis. J Breast Cancer, 2017, 20(2):132–41., DOI 10.4048/ jbc.2017.20.2.132 Guan Y, Bhandari A, Zhang X, and Wang O. Uridine Phosphorylase 1 Associates to Biological and Clinical Significance in Thyroid Carcinoma Cell Lines. J Cel Mol Med, 2019, 23(11):7438–48., DOI 10.1111/jcmm.14612 Wu Y, Jamal M, Xie T, Sun J, Song T, Yin Q, et al. Uridine-cytidine Kinase 2, UCK2): A Potential Diagnostic and Prognostic Biomarker for Lung Cancer. Cancer Sci, 2019, 110(9):2734–47., DOI 10.1111/cas.14125 Mazidi M, Katsiki N, Mikhailidis DP, and Banach M. The Link between Insulin Resistance Parameters and Serum Uric Acid Is Mediated by Adiposity. Atherosclerosis, 2018, 270:180–6., DOI 10.1016/ j.atherosclerosis.2017.12.033 Mele C, Tagliaferri MA, Saraceno G, Mai S, Vietti R, Zavattaro M, et al. Serum Uric Acid Potentially Links Metabolic Health to Measures of Fuel Use in Lean and Obese Individuals. Nutr Metab Cardiovasc Dis, 2018, 28(10):1029–35., DOI 10.1016/j.numecd.2018.06.010 Ali N, Perveen R, Rahman S, Mahmood S, Rahman S, Islam S, et al. Prevalence of Hyperuricemia and the Relationship between Serum Uric Acid and Obesity: A Study on Bangladeshi Adults. PloS one, 2018, 13(11):e0206850., DOI 10.1371/journal.pone.0206850 Levine W, Dyer AR, Shekelle RB, Schoenberger JA, and Stamler J. Serum Uric Acid and 11.5-year Mortality of Middle-Aged Women: Findings of the Chicago Heart Association Detection Project in Industry. J Clin Epidemiol, 1989, 42(3):257–67., DOI 10.1016/0895-4356(89)90061-9 Kolonel LN, Yoshizawa C, Nomura AM, and Stemmermann GN. Relationship of Serum Uric Acid to Cancer Occurrence in a Prospective Male Cohort. Cancer Epidemiol Biomarkers Prev, 1994, 3:225–8. Yan S, Zhang P, Xu W, Liu Y, Wang B, Jiang T, et al. Serum Uric Acid Increases Risk of Cancer Incidence and Mortality: A Systematic Review and Meta-Analysis. Mediators Inflamm, 2015, 2015:764250., DOI 10.1155/2015/ 764250 Hammarsten J, Damber J-E, Peeker R, Mellstrom D, and Hogstedt B. A Higher Prediagnostic Insulin Level Is a Prospective Risk Factor for Incident Prostate Cancer. Cancer Epidemiol, 2010, 34(5):574–9., DOI 10.1016/ j.canep.2010.06.014 Yim K, Bindayi A, McKay R, Mehrazin R, Raheem OA, Field C, et al. Rising Serum Uric Acid Level Is Negatively Associated with Survival in Renal Cell Carcinoma. Cancers, 2019, 11(4):536., DOI 10.3390/cancers11040536 Cetin AO, Omar M, Calp S, Tunca H, Yimaz N, Ozseker B, et al. Hyperuricemia at the Time of Diagnosis Is a Factor for Poor Prognosis in Patients with Stage II and III Colorectal Cancer, Uric Acid and Colorectal Cancer). Asian Pac J Cancer Prev, 2017, 18(2):485–90., DOI 10.22034/ APJCP.2017.18.2.485 Subandrate S, Lea K, and Safyudin. The Correlation between Uric Acid and Stages of Malignancy Among Gastric Cancer Patient in Palembang, Indonesia. J Phys Conf Ser, 2019, 1246:012062., DOI 10.1088/1742-6596/ 1246/1/012062 Wang S, Liu X, He Z, Chen X, and Li W. Hyperuricemia Has an Adverse Impact on the Prognosis of Patients with Osteosarcoma. Tumor Biol, 2016, 37(1):1205–10., DOI 10.1007/s13277-015-3830-3 Fabbrini E, SerafiniM, Colic Baric I, Hazen SL, and Klein S. Effect of Plasma Uric Acid on Antioxidant Capacity, Oxidative Stress, and Insulin Sensitivity in Obese Subjects. Diabetes, 2014, 63(3):976–81., DOI 10.2337/ db13-1396 Gersch C, Palii SP, Kim KM, Angerhofer A, Johnson RJ, and Henderson GN. Inactivation of Nitric Oxide by Uric Acid. Nucleosides, Nucleotides and Nucleic Acids, 2008, 27(8):967–78., DOI 10.1080/15257770802257952 Wan X, Xu C, Lin Y, Lu C, Li D, Sang J, et al. Uric Acid Regulates Hepatic Steatosis and Insulin Resistance through the NLRP3 Inflammasomedependent Mechanism. J Hepatol, 2016, 64(4):925–32., DOI 10.1016/ j.jhep.2015.11.022 Sautin YY, Nakagawa T, Zharikov S, and Johnson RJ. Adverse Effects of the Classic Antioxidant Uric Acid in Adipocytes: NADPH Oxidase-Mediated Oxidative/nitrosative Stress. Am J Physiology-Cell Physiol, 2007, 293(2): C584–C596., DOI 10.1152/ajpcell.00600.2006 Yu M-A, Sanchez-Lozada LG, Johnson RJ, and Kang D-H. Oxidative Stress with an Activation of the Renin-Angiotensin System in Human Vascular Endothelial Cells as a Novel Mechanism of Uric Acid-Induced Endothelial Dysfunction. J Hypertens, 2010, 28(6):1234–42., DOI 10.1097/ hjh.0b013e328337da1d Corry DB, Eslami P, Yamamoto K, Nyby MD, Makino H, and Tuck ML. Uric Acid Stimulates Vascular Smooth Muscle Cell Proliferation and Oxidative Stress via the Vascular Renin-Angiotensin System. J Hypertens, 2008, 26(2): 269–75., DOI 10.1097/hjh.0b013e3282f240bf Cirillo P, Gersch MS, Mu W, Scherer PM, Kim KM, Gesualdo L, et al. Ketohexokinase-dependent Metabolism of Fructose Induces Proinflammatory Mediators in Proximal Tubular Cells. Jasn, 2009, 20(3): 545–53., DOI 10.1681/asn.2008060576 Lanaspa MA, Sanchez-Lozada LG, Choi Y-J, Cicerchi C, Kanbay M, Roncal- Jimenez CA, et al. Uric Acid Induces Hepatic Steatosis by Generation of Mitochondrial Oxidative Stress. J Biol Chem, 2012, 287(48):40732–44., DOI 10.1074/jbc.m112.399899 DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A, Wei C, Frese K, et al. Oncogene-induced Nrf2 Transcription Promotes ROS Detoxification and Tumorigenesis. Nature, 2011, 475(7354):106–9., DOI 10.1038/nature10189 Ames BN, Cathcart R, Schwiers E, and Hochstein P. Uric Acid Provides an Antioxidant Defense in Humans against Oxidant- and Radical-Caused Aging and Cancer: a Hypothesis. Proc Natl Acad Sci, 1981, 78(11):6858–62., DOI 10.1073/pnas.78.11.6858 Linder N, Haglund C, Lundin M, Nordling S, Ristimaki A, Kokkola A, et al. Decreased Xanthine Oxidoreductase Is a Predictor of Poor Prognosis in Early-Stage Gastric Cancer. J Clin Pathol, 2006, 59(9):965–71., DOI 10.1136/ jcp.2005.032524 Linder N, Martelin E, Lundin M, Louhimo J, Nordling S, Haglund C, et al. Xanthine Oxidoreductase - Clinical Significance in Colorectal Cancer and In Vitro Expression of the Protein in Human colon Cancer Cells. Eur J Cancer, 2009, 45(4):648–55., DOI 10.1016/j.ejca.2008.10.036 Linder N, Lundin J, Isola J, Lundin M, Raivio KO, and Joensuu H. Down- Regulated Xanthine Oxidoreductase Is a Feature of Aggressive Breast Cancer. Clin Cancer Res, 2005, 11:4372–81., DOI 10.1158/1078-0432.ccr-04-2280 Cheung KJ, Tzameli I, Pissios P, Rovira I, Gavrilova O, Ohtsubo T, et al. Xanthine Oxidoreductase Is a Regulator of Adipogenesis and PPARγ Activity. Cel Metab, 2007, 5(2):115–28., DOI 10.1016/j.cmet.2007.01.005 Fini MA, Monks J, Farabaugh SM, and Wright RM. Contribution of Xanthine Oxidoreductase to Mammary Epithelial and Breast Cancer Cell Differentiation in Part Modulates Inhibitor of Differentiation-1. Mol Cancer Res, 2011, 9(9):1242–54., DOI 10.1158/1541-7786.mcr-11-0176 Fini MA, Orchard-Webb D, Kosmider B, Amon JD, Kelland R, Shibao G, et al. Migratory Activity of Human Breast Cancer Cells Is Modulated by Differential Expression of Xanthine Oxidoreductase. J Cel Biochem, 2008, 105(4):1008–26., DOI 10.1002/jcb.21901 Chen D, Forootan SS, Gosney JR, Forootan FS, and Ke Y. Increased Expression of Id1 and Id3 Promotes Tumorigenicity by Enhancing Angiogenesis and Suppressing Apoptosis in Small Cell Lung Cancer. Genes Cancer, 2014, 5(5-6):212–25., DOI 10.18632/genesandcancer.20 Sharma BK, Kolhe R, Black SM, Keller JR,Mivechi NF, and Satyanarayana A. Inhibitor of Differentiation 1 Transcription Factor Promotes Metabolic Reprogramming in Hepatocellular Carcinoma Cells. FASEB j, 2016, 30(1): 262–75., DOI 10.1096/fj.15-277749 Singh B, Berry JA, Shoher A, Ayers GD, Wei C, and Lucci A. COX-2 Involvement in Breast Cancer Metastasis to Bone. Oncogene, 2007, 26(26):3789–96., DOI 10.1038/sj.onc.1210154 Baldwin W, McRae S, Marek G, Wymer D, Pannu V, Baylis C, et al. Hyperuricemia as a Mediator of the Proinflammatory Endocrine Imbalance in the Adipose Tissue in a Murine Model of the Metabolic Syndrome. Diabetes, 2011, 60(4):1258–69., DOI 10.2337/db10-0916 Gandhi J, Khera L, Gaur N, Paul C, and Kaul R. Role of Modulator of Inflammation Cyclooxygenase-2 in Gammaherpesvirus Mediated Tumorigenesis. Front Microbiol, 2017, 8:538., DOI 10.3389/fmicb.2017.00538 Hou Z., Falcone D. J., Subbaramaiah K., and Dannenberg A. J.. Macrophages induce COX-2 expression in breast cancer cells: role of IL-1β autoamplification. Carcin, 2011, 32(5):695–702., DOI 10.1093/carcin/bgr027 Martinez-Reyes C. P., Manjarrez-Reyna A. N., Mendez-Garcia L. A., Aguayo- Guerrero J. A., Aguirre-Sierra B., Villalobos-Molina R, et al. Uric Acid Has Direct Proinflammatory Effects on Human Macrophages by Increasing Proinflammatory Mediators and Bacterial Phagocytosis Probably via URAT1. Biomolecules, 2020, 10:576., DOI 10.3390/biom10040576 Lee HE, Lee JY, Yang G, Kang HC, Cho Y-Y, Lee HS, et al. Inhibition of NLRP3 Inflammasome in TumorMicroenvironment Leads to Suppression of Metastatic Potential of Cancer Cells. Scientific Rep, 2019, 9(1):12277., DOI 10.1038/s41598-019-48794-x Bent R, Moll L, Grabbe S, and Bros M. Interleukin-1 Beta-A Friend or Foe in Malignancies? Ijms, 2018, 19(8):2155., DOI 10.3390/ijms19082155 El-Deeb MMK, El-Sheredy HG, and Mohammed AF. The Possible Role of Interleukin, IL)-18 and Nitrous Oxide and Their Relation to Oxidative Stress in the Development and Progression of Breast Cancer. Asian Pac J Cancer Prev, 2019, 20(9):2659–65., DOI 10.31557/apjcp.2019.20.9.2659 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609828 EP 1609851 DI 10.3389/pore.2021.1609828 PG 24 ER PT J AU Xie, J Kong, X Wang, W Li, Y Lin, M Li, H Chen, J Zhou, W He, J Wu, H AF Xie, Jing Kong, Xue Wang, Wei Li, Yuan Lin, Mengyu Li, Heng Chen, Jingjing Zhou, Wenchao He, Jie Wu, Haibo TI Vasculogenic Mimicry Formation Predicts Tumor Progression in Oligodendroglioma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PFS; oligodendroglioma; vasculogenic mimicry; ATM; ATR ID PFS; oligodendroglioma; vasculogenic mimicry; ATM; ATR AB Vasculogenic mimicry (VM) has been identified as an important vasculogenic mechanism in malignant tumors, but little is known about its clinical meanings and mechanisms in oligodendroglioma. In this study, VM-positive cases were detected in 28 (20.6%) out of 136 oligodendroglioma samples, significantly associated with higher WHO grade, lower Karnofsky performance status (KPS) scores, and recurrent tumor (p < 0.001, p = 0.040, and p = 0.020 respectively). Patients with VM-positive oligodendroglioma had a shorter progress-free survival (PFS) compared with those with VM-negative tumor (p < 0.001), whereas no significant difference was detected in overall survival (OS) between these patients. High levels of phosphorylate serine/threonine kinases Ataxia-telangiectasia mutated (pATM) and phosphorylate Ataxia-telangiectasia and Rad3-Related (pATR) were detected in 31 (22.8%) and 34 (25.0%), respectively out of 136 oligodendroglioma samples. Higher expressions of pATM and pATR were both associated with a shorter PFS (p < 0.001 and p < 0.001). VM-positive oligodendroglioma specimens tended to exhibit higher pATM and pATR staining than VM-negative specimens (rs = 0.435, p < 0.001 and rs = 0.317, p < 0.001). Besides, Hypoxia-inducible factor-1α (HIF1α) expression was detected in 14(10.3%) samples, correlated with higher WHO grade and non-frontal lobe (p = 0.010 and p = 0.029). However, no obvious connection was detected between HIF1α expression and VM formation (p = 0.537). Finally, either univariate or multivariate analysis suggested that VM was an independent unfavorable predictor for oligodendroglioma patients (p < 0.001, HR = 7.928, 95%CI: 3.382–18.584, and p = 0.007, HR = 4.534, 95%CI: 1.504–13.675, respectively). VM is a potential prognosticator for tumor progression in oligodendroglioma patients. Phosphorylation of ATM and ATR linked to treatment-resistance may be associated with VM formation. The role of VM in tumor progression and the implication of pATM/pATR in VM formation may provide potential therapeutic targets for oligodendroglioma treatment. C1 [Xie, Jing] Shandong University, School of MedicineJinan, China. [Kong, Xue] University of Science and Technology of China, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of PathologyHefei, China. [Wang, Wei] University of Science and Technology of China, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of PathologyHefei, China. [Li, Yuan] University of Science and Technology of China, Division of Life Sciences and Medicine, Intelligent Pathology InstituteHefei, China. [Lin, Mengyu] University of Science and Technology of China, Division of Life Sciences and Medicine, Intelligent Pathology InstituteHefei, China. [Li, Heng] University of Science and Technology of China, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of PathologyHefei, China. [Chen, Jingjing] University of Science and Technology of China, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of PathologyHefei, China. [Zhou, Wenchao] University of Science and Technology of China, Division of Life Sciences and Medicine, Intelligent Pathology InstituteHefei, China. [He, Jie] Shandong University, School of MedicineJinan, China. [Wu, Haibo] University of Science and Technology of China, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of PathologyHefei, China. RP Wu, H (reprint author), University of Science and Technology of China, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Pathology, Hefei, China. EM wuhaibo@ustc.edu.cn CR Wesseling P, and Capper D. WHO 2016 Classification of Gliomas. Neuropathol Appl Neurobiol, 2018, 44(2):139–50., DOI 10.1111/nan.12432 Wesseling P, van den Bent M, and Perry A. Oligodendroglioma: Pathology, Molecular Mechanisms and Markers. Acta Neuropathol, 2015, 129(6):809–27., DOI 10.1007/s00401-015-1424-1 Ney DE, and Lassman AB. Molecular Profiling of Oligodendrogliomas: Impact on Prognosis, Treatment, and Future Directions. Curr Oncol Rep, 2009, 11(1): 62–7., DOI 10.1007/s11912-009-0010-x Chamberlain MC, and Born D. Prognostic Significance of Relative 1p/19q Codeletion in Oligodendroglial Tumors. J Neurooncol, 2015, 125(2):249–51., DOI 10.1007/s11060-015-1906-y Van Den Bent MJ, Bromberg JEC, and Buckner J. Low-grade and Anaplastic Oligodendroglioma. Handb Clin Neurol, 2016, 134:361–80., DOI 10.1016/B978- 0-12-802997-8.00022-0 Strickland-Marmol LB, BremAM Rojiani SMV, Rojiani AM, and Rojiani MV. Vessel Morphometric Parameters-Correlation with Histologic Grade and VEGF Expression in Oligodendroglioma. Am J Cancer Res, 2017, 7(4): 973–81. Quon H, HasbiniA, Cougnard J,Djafari L, LacroixC, and Abdulkarim B. Assessment of Tumor Angiogenesis as a Prognostic Factor of Survival in Patients with Oligodendroglioma. J Neurooncol, 2010, 96(2):277–85., DOI 10.1007/s11060-009- 9961-x de Lemos ML, Markarian A, Chan E, Schaff K, and Walisser S. Clinical Effectiveness of Bevacizumab in Patients with Recurrent Brain Tumours: A Population-Based Evaluation. J Oncol Pharm Pract, 2018, 24(1):33–6., DOI 10.1177/1078155216681191 Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LMG, Pe’er J, et al. Vascular Channel Formation by Human Melanoma Cells In Vivo and In Vitro: Vasculogenic Mimicry. Am J Pathol, 1999, 155(3):739–52., DOI 10.1016/S0002-9440(10)65173-5 Cavaco A, Rezaei M, Niland S, and Eble JA. Collateral Damage Intended-Cancer- Associated Fibroblasts and Vasculature Are Potential Targets in Cancer Therapy. Int. J. Mol. Sci., 2017, 18(11):2355., DOI 10.3390/ijms18112355 Yang JP, Liao YD,Mai DM, Xie P, Qiang YY, Zheng LS, et al. TumorVasculogenic Mimicry Predicts Poor Prognosis in Cancer Patients: a Meta-Analysis. Angiogenesis, 2016, 19(2):191–200., DOI 10.1007/s10456-016-9500-2 Guo Q, Yuan Y, Jin Z, Xu T, Gao Y,Wei H, et al. Association between Tumor Vasculogenic Mimicry and the Poor Prognosis of Gastric Cancer in China: An Updated Systematic Review and Meta-Analysis. Biomed Res Int, 2016, 2016: 1–8., DOI 10.1155/2016/2408645 Vasudev NS, and Reynolds AR. Anti-angiogenic Therapy for Cancer: Current Progress, Unresolved Questions and Future Directions. Angiogenesis, 2014, 17(3):471–94., DOI 10.1007/s10456-014-9420-y Ellis LM, and Hicklin DJ. VEGF-targeted Therapy: Mechanisms of Antitumour Activity. Nat Rev Cancer, 2008, 8(8):579–91., DOI 10.1038/nrc2403 Dunleavey JM, and Dudley AC. Vascular Mimicry: Concepts and Implications for Anti-angiogenic Therapy. Curr Angiogenes, 2012, 1(2):133–8., DOI 10.2174/ 2211552811201020133 Yamamoto J, Shimajiri S, Miyaoka R, and Nishizawa S. Pitfalls of Conservative Treatments of Multiple Probable Cerebral Cavernous Malformations, CCMs): Clinicopathological Features of CCMs Coexisting with Vasculogenic Mimicry in an Anaplastic Oligodendroglioma. Brain Tumor Pathol, 2014, 31(3): 215–21., DOI 10.1007/s10014-013-0171-z Blackford AN, and Jackson SP. ATM, ATR, and DNA-PK: The Trinity at the Heart of the DNA Damage Response. Mol Cel, 2017, 66(6):801–17., DOI 10.1016/j.molcel.2017.05.015 Choy KR, and Watters DJ. Neurodegeneration in Ataxia-Telangiectasia: Multiple Roles of ATM Kinase in Cellular Homeostasis. Dev Dyn, 2018, 247(1):33–46., DOI 10.1002/dvdy.24522 Zaki-Dizaji M, Akrami SM, Azizi G, Abolhassani H, and Aghamohammadi A. Inflammation, a Significant Player of Ataxia-Telangiectasia Pathogenesis? Inflamm Res, 2018, 67(7):559–70., DOI 10.1007/s00011-018-1142-y Stagni V, Ferri A, Cirotti C, and Barila D. ATM Kinase-dependent Regulation of Autophagy: A Key Player in Senescence? Front Cel Dev. Biol., 2021, 8: 599048., DOI 10.3389/fcell.2020.599048 Bubien V, Bonnet F,Dupiot-Chiron J, Barouk-Simonet E, JonesN, de Reynies A, et al. Combined Tumor Genomic Profiling and Exome Sequencing in a Breast Cancer Family Implicates ATM in Tumorigenesis: A Proof of Principle Study. Genes Chromosomes Cancer, 2017, 56(11):788–99., DOI 10.1002/gcc.22482 Mandriota SJ, Valentijn LJ, Lesne L, Betts DR, Marino D, Boudal-Khoshbeen M, et al. Ataxia-telangiectasia Mutated, ATM, Silencing Promotes Neuroblastoma Progression through a MYCN Independent Mechanism. Oncotarget, 2015, 6(21):18558–76., DOI 10.18632/oncotarget.4061 Eich M, Roos WP, Nikolova T, and Kaina B. Contribution of ATM and ATR to the Resistance of Glioblastoma and Malignant Melanoma Cells to the Methylating Anticancer Drug Temozolomide. Mol Cancer Ther, 2013, 12(11):2529–40., DOI 10.1158/1535-716310.1158/1535-7163.mct-13-0136 Squatrito M, Brennan CW, Helmy K, Huse JT, Petrini JH, and Holland EC. Loss of ATM/Chk2/p53 Pathway Components Accelerates Tumor Development and Contributes to Radiation Resistance in Gliomas. Cancer Cell, 2010, 18(6):619–29., DOI 10.1016/j.ccr.2010.10.034 Ye Q, Chen H, Wen Z, Guo W, Huang Y, and Mo X. Abnormal Expression of P-Atm/chk2 in Nasal Extranodal NK/T Cell Lymphoma, Nasal Type, Is Correlated with Poor Prognosis. J Clin Pathol, 2020, 74:223–7., DOI 10.1136/jclinpath-2020-206476 Di Benedetto A, Ercolani C, Mottolese M, Sperati F, Pizzuti L, Vici P, et al. Analysis of the ATR-Chk1 and ATM-Chk2 Pathways in Male Breast Cancer Revealed the Prognostic Significance of ATR Expression. Sci Rep, 2017, 7(1): 8078., DOI 10.1038/s41598-017-07366-7 Okuno Y, Nakamura-Ishizu A, Otsu K, Suda T, and Kubota Y. Pathological Neoangiogenesis Depends on Oxidative Stress Regulation by ATM. Nat Med, 2012, 18(8):1208–16., DOI 10.1038/nm.2846 Shi Y, Liu Z, Zhang Q, Vallee I, Mo Z, Kishi S, et al. Phosphorylation of Seryl-tRNA Synthetase by ATM/ATR Is Essential for Hypoxia-Induced Angiogenesis. Plos Biol, 2020, 18(12):e3000991., DOI 10.1371/journal.pbio.3000991 Scheie D, Meling TR, Cvancarova M, Skullerud K, Mork S, Lote K, et al. Prognostic Variables in Oligodendroglial Tumors: a Single-Institution Study of 95 Cases. Neuro Oncol, 2011, 13(11):1225–33., DOI 10.1093/neuonc/nor114 Hendrix MJC, Seftor EA, Seftor REB, Chao D-S, and Chu Y-W. Tumor Cell Vascular Mimicry: Novel Targeting Opportunity in Melanoma. Pharmacol Ther, 2016, 159:83–92., DOI 10.1016/j.pharmthera.2016.01.006 Shen Y, Quan J, Wang M, Li S, Yang J, Lv M, et al. Tumor Vasculogenic Mimicry Formation as an Unfavorable Prognostic Indicator in Patients with Breast Cancer. Oncotarget, 2017, 8(34):56408–16., DOI 10.18632/oncotarget.16919 Zhou L, Chang Y, Xu L, Hoang STN, Liu Z, Fu Q, et al. Prognostic Value of Vascular Mimicry in Patients with Urothelial Carcinoma of the Bladder after Radical Cystectomy. Oncotarget, 2016, 7(46):76214–23., DOI 10.18632/ oncotarget.12775 Sun J, SunB, SunR, ZhuD,ZhaoX,ZhangY, et al.HMGA2 Promotes Vasculogenic Mimicry and Tumor Aggressiveness by Upregulating Twist1 in Gastric Carcinoma. Sci Rep, 2017, 7(1):2229., DOI 10.1038/s41598-017-02494-6 Wu H-B, Yang S, Weng H-Y, Chen Q, Zhao X-L, Fu W-J, et al. Autophagyinduced KDR/VEGFR-2 Activation Promotes the Formation of Vasculogenic Mimicry by Glioma Stem Cells. Autophagy, 2017, 13(9):1528–42., DOI 10.1080/ 15548627.2017.1336277 Baisiwala S, Auffinger B, Caragher SP, Shireman JM, Ahsan R, Lee G, et al. Chemotherapeutic Stress Induces Transdifferentiation of Glioblastoma Cells to Endothelial Cells and Promotes Vascular Mimicry. Stem Cell Int, 2019, 2019:6107456., DOI 10.1155/2019/6107456 Pezzella F, and Ribatti D. Vascular Co-option and Vasculogenic Mimicry Mediate Resistance to Antiangiogenic Strategies. Cancer Rep, 2020):e1318., DOI 10.1002/cnr2.1318 Sun H, Zhang D, Yao Z, Lin X, Liu J, Gu Q, et al. Anti-angiogenic Treatment Promotes Triple-Negative Breast Cancer Invasion via Vasculogenic Mimicry. Cancer Biol Ther, 2017, 18(4):205–13., DOI 10.1080/15384047.2017.1294288 Francis JH, Kim J, Lin A, Folberg R, Iyer S, and Abramson DH. Growth of Uveal Melanoma Following Intravitreal Bevacizumab. Ocul Oncol Pathol, 2017, 3(2):117–21., DOI 10.1159/000450859 Xu Y, Li Q, Li X-Y, Yang Q-Y, Xu W-W, and Liu G-L. Short-term Antivascular Endothelial Growth Factor Treatment Elicits Vasculogenic Mimicry Formation of Tumors to Accelerate Metastasis. J Exp Clin Cancer Res, 2012, 31(1):16., DOI 10.1186/1756-9966-31-16 Cremona CA, and Behrens A. ATM Signalling and Cancer. Oncogene, 2014, 33(26):3351–60., DOI 10.1038/onc.2013.275 Lecona E, and Fernandez-Capetillo O. Targeting ATR in Cancer. Nat Rev Cancer, 2018, 18(9):586–95., DOI 10.1038/s41568-018-0034-3 Nadkarni A, Shrivastav M, Mladek AC, Schwingler PM, Grogan PT, Chen J, et al. ATM Inhibitor KU-55933 Increases the TMZ Responsiveness of Only Inherently TMZ Sensitive GBM Cells. J Neurooncol, 2012, 110(3):349–57., DOI 10.1007/s11060-012-0979-0 Agnihotri S, Burrell K, Buczkowicz P, Remke M, Golbourn B, Chornenkyy Y, et al. ATM Regulates 3-Methylpurine-DNA Glycosylase and Promotes Therapeutic Resistance to Alkylating Agents. Cancer Discov, 2014, 4(10): 1198–213., DOI 10.1158/2159-8290.CD-14-0157 BianL,MengY,ZhangM, GuoZ,LiuF, ZhangW, et al. ATMExpressionIsElevated in Established Radiation-Resistant Breast Cancer Cells and Improves DNA Repair Efficiency. Int J Biol Sci, 2020, 16(7):1096–106., DOI 10.7150/ijbs.41246 Wechman SL, Emdad L, Sarkar D, Das SK, and Fisher PB. Vascular Mimicry: Triggers, Molecular Interactions and In Vivo Models. Adv Cancer Res, 2020, 148:27–67., DOI 10.1016/bs.acr.2020.06.001 Zhang J, Qiao L, Liang N, Xie J, Luo H, Deng G, et al. Vasculogenic Mimicry and Tumor Metastasis. J BUON, 2016, 21(3):533–41. Yang D, Peng M, Hou Y, Qin Y, Wan X, Zhu P, et al. Oxidized ATM Promotes Breast Cancer Stem Cell Enrichment through Energy Metabolism Reprogram- Mediated Acetyl-CoA Accumulation. Cell Death Dis, 2020, 11(7):508., DOI 10.1038/s41419-020-2714-7 Liu M, Zeng T, Zhang X, Liu C, Wu Z, Yao L, et al. ATR/Chk1 Signaling Induces Autophagy through Sumoylated RhoB-Mediated Lysosomal Translocation of TSC2 after DNA Damage. Nat Commun, 2018, 9(1):4139., DOI 10.1038/s41467-018-06556-9 Antonelli M, Strappazzon F, Arisi I, Brandi R, D’Onofrio M, Sambucci M, et al. ATM Kinase Sustains Breast Cancer Stem-like Cells by Promoting ATG4C Expression and Autophagy. Oncotarget, 2017, 8(13):21692–709., DOI 10.18632/ oncotarget.15537 Yao X-h., Ping Y-f., and Bian X-w.. Contribution of Cancer Stem Cells to Tumor Vasculogenic Mimicry. Protein Cell, 2011, 2(4):266–72., DOI 10.1007/ s13238-011-1041-2 Fallone F, Britton S, Nieto L, Salles B, and Muller C. ATR Controls Cellular Adaptation to Hypoxia through Positive Regulation of Hypoxia-Inducible Factor 1, HIF-1, Expression. Oncogene, 2013, 32(37):4387–96., DOI 10.1038/ onc.2012.462 Cam H, Easton JB, High A, and Houghton PJ. mTORC1 Signaling under Hypoxic Conditions Is Controlled by ATM-dependent Phosphorylation of HIF-1α. Mol Cel, 2010, 40(4):509–20., DOI 10.1016/j.molcel.2010.10.030 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609844 EP 1609855 DI 10.3389/pore.2021.1609844 PG 12 ER PT J AU Zhang, N Liu, Y Yang, H Liang, M Wang, X Wang, M Kong, J Yuan, X Zhou, F AF Zhang, Ning Liu, Yiwen Yang, Hong Liang, Mengxia Wang, Xiaopeng Wang, Min Kong, Jinyu Yuan, Xiang Zhou, Fuyou TI Clinical Significance of Fusobacterium nucleatum Infection and Regulatory T Cell Enrichment in Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Esophageal squamous cell carcinoma; Fusobacterium nucleatum; Prognostic value; regulatory T cells; clinical significance ID Esophageal squamous cell carcinoma; Fusobacterium nucleatum; Prognostic value; regulatory T cells; clinical significance AB A variety of pathogenic microorganisms promote tumor occurrence and development through long-term colonization in the body. Fusobacterium nucleatum (F. nucleatum) is abundant in precancerous esophageal lesions and is closely related to the malignant progression of esophageal squamous cell carcinoma (ESCC). The invasion of exogenous microorganisms can reshape the immune microenvironment, make the immune system incapacitated, and assist tumor cells in immune escape. A variety of pathogenic microorganisms induce the recruitment of regulatory T cell (Tregs) to allow tumor cells to escape immune surveillance and provide favorable conditions for their own long-term colonization. Tregs are one of the major obstacles to tumor immunotherapy and have a significant positive correlation with the occurrence and development of many kinds of tumors. Because F. nucleatum can instantly enter cells and colonize for a long time, we speculated that F. nucleatum infection could facilitate the immune escape of tumor cells through enrichment of Tregs and promote the malignant progression of ESCC. In this study, we found a significant concordance between F. nucleatum infection and Tregs infiltration. Therefore, we propose the view that chronic infection of F. nucleatum may provide favorable conditions for long-term colonization of itself by recruiting Tregs and suppressing the immune response. At the same time, the massive enrichment of Treg may also weaken the immune response and assist in the long-term colonization of F. nucleatum. We analyzed the correlation between F. nucleatum infection with the clinicopathological characteristics and survival prognosis of the patients. F. nucleatum infection was found to be closely related to sex, smoking, drinking, degree of differentiation, depth of invasion, lymph node metastasis, and clinical stage. The degree of differentiation, depth of infiltration, lymph node metastasis, clinical stage, and F. nucleatum infection are independent risk factors affecting ESCC prognosis. Additionally, the survival rate and median survival time were significantly shortened in the F. nucleatum infection positive group. Therefore, we propose that long-term smoking and alcohol consumption cause poor oral and esophageal environments, thereby significantly increasing the risk of F. nucleatum infection. In turn, F. nucleatum infection and colonization may weaken the antitumor immune response through Treg enrichment and further assist in self-colonization, promoting the malignant progression of ESCC. C1 [Zhang, Ning] The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang Tumor HospitalAnyang, China. [Liu, Yiwen] Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer InstituteLuoyang, China. [Yang, Hong] Henan University of Science and Technology, School of PELuoyang, China. [Liang, Mengxia] Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer InstituteLuoyang, China. [Wang, Xiaopeng] The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang Tumor HospitalAnyang, China. [Wang, Min] The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang Tumor HospitalAnyang, China. [Kong, Jinyu] Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer InstituteLuoyang, China. [Yuan, Xiang] Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer InstituteLuoyang, China. [Zhou, Fuyou] The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang Tumor HospitalAnyang, China. RP Zhou, F (reprint author), The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang Tumor Hospital, Anyang, China. EM ayzhoufuyou@163.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Li S, Chen H, Man J, Zhang T, Yin X, He Q, et al. Changing Trends in the Disease burden of Esophageal Cancer in China from 1990 to 2017 and its Predicted Level in 25 Years. Cancer Med, 2021, 10(5):1889–99., DOI 10.1002/ cam4.3775 Lagergren J, Smyth E, Cunningham D, and Lagergren P. Oesophageal Cancer. Lancet, 2017, 390(10110):2383–96., DOI 10.1016/S0140-6736(17)31462-9 Carmen Lia. M, Massilva. R, Christian. S, Monique. B, Claire. B, Marine. B, et al. Natural History, Dynamics, and Ecology of Human Papillomaviruses in Genital Infections of Young Women: Protocol of the PAPCLEAR Cohort Study. BMJ Open, 2019, 9(6):e025129., DOI 10.1136/bmjopen-2018-025129 Liu Y, Yuan X, Chen K, Zhou F, Yang H, Yang H, et al. Clinical Significance and Prognostic Value of Porphyromonas Gingivalis Infection in Lung Cancer. Translational Oncol, 2021, 14(1):100972., DOI 10.1016/j.tranon.2020.100972 Li N, Fan X, Wang X,Deng H, Zhang K, Zhang X, et al. PRDM1 Levels Are Associated with Clinical Diseases in Chronic HBV Infection and Survival of Patients with HBV-Related Hepatocellular Carcinoma. Int Immunopharmacology, 2019, 73(undefined):156–62., DOI 10.1016/ j.intimp.2019.05.012 Slade DJ. New Roles for Fusobacterium Nucleatum in Cancer: Target the Bacteria, Host, or Both? Trends Cancer, 2021, 7(3):185–7., DOI 10.1016/ j.trecan.2020.11.006 Han YW. Fusobacterium Nucleatum: a Commensal-Turned Pathogen. Curr Opin Microbiol, 2015, 23(undefined):141–7., DOI 10.1016/j.mib.2014.11.013 Gholizadeh P, Eslami H, and Kafil HS. Carcinogenesis Mechanisms of Fusobacterium Nucleatum. Biomed Pharmacother, 2017, 89(undefined): 918–25., DOI 10.1016/j.biopha.2017.02.102 Baba Y, Iwatsuki M, Yoshida N, Watanabe M, and Baba H. Review of the Gut Microbiome and Esophageal Cancer: Pathogenesis and Potential Clinical Implications. Ann Gastroenterol Surg, 2017, 1(2):99–104., DOI 10.1002/ ags3.12014 Yamamura K, Baba Y, Nakagawa S, Mima K, Miyake K, Nakamura K, et al. Human Microbiome Fusobacterium Nucleatum in Esophageal Cancer Tissue Is Associated with Prognosis. Clin Cancer Res, 2016, 22(22):5574–81., DOI 10.1158/1078-0432.CCR-16-1786 Altorki NK, Markowitz GJ, Gao D, Port JL, Saxena A, Stiles B, et al. McGraw Timothy., Mittal Vivek.the Lung Microenvironment: an Important Regulator of Tumour Growth and Metastasis. Nat Rev Cancer, 2019, 19(1):9–31., DOI 10.1038/s41568-018-0081-9 Roumenina LT, Daugan MV, Petitprez F, Sautes-Fridman C, and Fridman WH. Daugan Marie V., Petitprez Florent., Sautes-Fridman Catherine., Fridman Wolf Herman.Context-dependent Roles of Complement in Cancer. Nat Rev Cancer, 2019, 19(12):698–715., DOI 10.1038/s41568-019- 0210-0 Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, and Flavell RA. Inflammationinduced Cancer: Crosstalk between Tumours, Immune Cells and Microorganisms. Nat Rev Cancer, 2013, 13(11):759–71., DOI 10.1038/nrc3611 Zhuo C, Xu Y, Ying M, Li Q, Huang L, Li D, et al. FOXP3+ Tregs: Heterogeneous Phenotypes and Conflicting Impacts on Survival Outcomes in Patients with Colorectal Cancer. Immunol Res, 2015, 61(3):338–47., DOI 10.1007/s12026-014-8616-y Kumar P, Saini S, and Prabhakar BS. Cancer Immunotherapy with Check point Inhibitor Can Cause Autoimmune Adverse Events Due to Loss of Treg Homeostasis. Semin Cancer Biol, 2020, 64(undefined):29–35., DOI 10.1016/ j.semcancer.2019.01.006 Yang L, Wang G, and Xia H. Molecular Mechanism for Impaired Suppressive Function of Tregs in Autoimmune Diseases: A Summary of Cell-intrinsic and Cell-extrinsic Factors. J Cel Mol Med, 2020, 24(19):11056–63., DOI 10.1111/ jcmm.15743 LinW, Niu Z, Zhang H, Kong Y, Wang Z, Yang X, et al. Imbalance of Th1/Th2 and Th17/Treg during the Development of Uterine Cervical Cancer. Int J Clin Exp Pathol, 2019, 12(9):3604–12. Tang R, Lei Z, Wang X, Qi Q, He J, Liu D, et al. Hepatitis B Envelope Antigen Increases Tregs by Converting CD4+CD25- T Cells into CD4+CD25+Foxp3+ Tregs. Exp Ther Med, 2020, 20(4):3679–86., DOI 10.3892/etm.2020.9107 Ling Z, Shao L, Liu X, Cheng Y, Yan C, Mei Y, et al. Regulatory T Cells and Plasmacytoid Dendritic Cells within the Tumor Microenvironment in Gastric Cancer Are Correlated with Gastric Microbiota Dysbiosis: A Preliminary Study. Front Immunol, 2019, 10:533., DOI 10.3389/fimmu.2019.00533 Urakawa S, Yamasaki M, Makino T, Kurokawa Y, Yamamoto K, Goto K, et al. The Impact of ICOS+ Regulatory T Cells and Helicobacter pylori Infection on the Prognosis of Patients with Gastric and Colorectal Cancer: Potential Prognostic Benefit of Pre-operative Eradication Therapy. Cancer Immunol Immunother, 2021, 70(2):443–52., DOI 10.1007/s00262-020-02696-4 Chinese Preventive Medicine Association. Zhonghua zhong liu Za Zhi [Chinese J oncology], 2021, 43(1):1–7., DOI 10.3760/cma.j.cn112152-20210106-00019 Wu K, Zhao X, Qi L, Zhou T, Zhang W, Fan Y, et al. A Preliminary Study on the Relationship between Alcohol Consumption and Oral flora in a Natural Population of Middle-Aged and Elderly Men in the Core Region of Southwest China[J]. West China Med, 2021, 02:230–7. Wang F, Flanagan J, Su N, Wang L-C, Bui S, Bui A, et al. RNAscope. J Mol Diagn, 2012, 14:22–9., DOI 10.1016/j.jmoldx.2011.08.002 Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, et al. Analysis of Fusobacteriumpersistence and Antibiotic Response in Colorectal Cancer. Science, 2017, 358(6369):1443–8., DOI 10.1126/science.aal5240 Cioca A, Cimpean AM, Ceausu RA, Tarlui V, Toma A, Marin I, et al. Evaluation of Podoplanin Expression in Hepatocellular Carcinoma Using RNAscope and Immunohistochemistry - A Preliminary Report. Cancer genomics & proteomics, 2017, 14(5):383–7., DOI 10.21873/cgp.20048 Sparger EE, Murphy BG, Kamal FM, Arzi B, Naydan D, Skouritakis CT, et al. Investigation of Immune Cell Markers in Feline Oral Squamous Cell Carcinoma. Vet Immunol immunopathology, 2018, 202:52–62., DOI 10.1016/ j.vetimm.2018.06.011 Schwabe RF, and Jobin C. TheMicrobiome and Cancer. Nat Rev Cancer, 2013, 13(11):800–12., DOI 10.1038/nrc3610 Han YW. Fusobacterium Nucleatum: a Commensal-Turned Pathogen. Curr Opin Microbiol, 2015, 23:141–7., DOI 10.1016/j.mib.2014.11.013 Fujiwara N, Kitamura N, Yoshida K, Yamamoto T, Ozaki K, and Kudo Y. Involvement of Fusobacterium Species in Oral Cancer Progression: A Literature Review Including Other Types of Cancer. Ijms, 2020, 21(17): 6207., DOI 10.3390/ijms21176207 Li W-M, and Liu H-R. CCL20-CCR6 CytokineNetwork Facilitate TregActivity in Advanced Grades and Metastatic Variants of Hepatocellular Carcinoma. Scand J Immunol, 2016, 83(1):33–7., DOI 10.1111/sji.12367 Gallimore A, Quezada SA, and Roychoudhuri R. Regulatory T Cells in Cancer: where Are We Now? Immunology, 2019, 157(3):187–9., DOI 10.1111/ imm.13088 Raghavan S, and Quiding-Jarbrink M. Immune Modulation by Regulatory T Cells in Helicobacter Pylori-Associated Diseases. Emiddt, 2012, 12(1): 71–85., DOI 10.2174/187153012799278974 Yang P, Li Q-J, Feng Y, Zhang Y, Markowitz GJ, Ning S, et al. TGF-β-miR-34a- CCL22 Signaling-Induced Treg Cell Recruitment Promotes Venous Metastases of HBV-Positive Hepatocellular Carcinoma. Cancer Cell, 2012, 22(3):291–303., DOI 10.1016/j.ccr.2012.07.023 Baumforth KRN, Birgersdotter A, Reynolds GM, WeiW, Kapatai G, Flavell JR, et al. Expression of the Epstein-Barr Virus-Encoded Epstein-Barr Virus Nuclear Antigen 1 in Hodgkin’s Lymphoma Cells Mediates Up-Regulation of CCL20 and the Migration of Regulatory T Cells. Am J Pathol, 2008, 173(1): 195–204., DOI 10.2353/ajpath.2008.070845 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609846 EP 1609854 DI 10.3389/pore.2021.1609846 PG 9 ER PT J AU Ma, W Li, W Xu, L Liu, L Xia, Y Yang, L Da, M AF Ma, Weijun Li, Weidong Xu, Lei Liu, Lu Xia, Yu Yang, Liping Da, Mingxu TI Identification of a Gene Prognostic Model of Gastric Cancer Based on Analysis of Tumor Mutation Burden SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; gastric cancer; TCGA; immune infiltration; tumor mutation burden ID prognosis; gastric cancer; TCGA; immune infiltration; tumor mutation burden AB Introduction: Gastric cancer is one of the most common cancers. Although some progress has been made in the treatment of gastric cancer with the improvement of surgical methods and the application of immunotherapy, the prognosis of gastric cancer patients is still unsatisfactory. In recent years, there has been increasing evidence that tumor mutational load (TMB) is strongly associated with survival outcomes and response to immunotherapy. Given the variable response of patients to immunotherapy, it is important to investigate clinical significance of TMB and explore appropriate biomarkers of prognosis in patients with gastric cancer (GC). Material and Methods: All data of patients with gastric cancer were obtained from the database of The Cancer Genome Atlas (TCGA). Samples were divided into two groups based on median TMB. Differently expressed genes (DEGs) between the high- and low- TMB groups were identified and further analyzed. We identified TMB-related genes using Lasso, univariate and multivariate Cox regression analysis and validated the survival result of 11 hub genes using Kaplan-Meier Plotter. In addition, “CIBERSORT” package was utilized to estimate the immune infiltration. Results: Single nucleotide polymorphism (SNP), C > T transition were the most common variant type and single nucleotide variant (SNV), respectively. Patients in the high-TMB group had better survival outcomes than those in the low-TMB group. Besides, eleven TMB-related DEGs were utilized to construct a prognostic model that could be an independent risk factor to predict the prognosis of patients with GC. What’s more, the infiltration levels of CD4+ memory-activated T cells, M0 and M1 macrophages were significantly increased in the high-TMB group compared with the low-TMB group. Conclusions: Herein, we found that patients with high TMB had better survival outcomes in GC. In addition, higher TMB might promote immune infiltration, which could provide new ideas for immunotherapy. C1 [Ma, Weijun] Ningxia Medical University, College of Clinical MedicineYinchuan, China. [Li, Weidong] Ningxia Medical University, College of Clinical MedicineYinchuan, China. [Xu, Lei] Gansu Provincial Hospital, Department of Surgical OncologyLanzhou, China. [Liu, Lu] Gansu Provincial Hospital, Department of Surgical OncologyLanzhou, China. [Xia, Yu] Gansu Provincial Hospital, Department of Surgical OncologyLanzhou, China. [Yang, Liping] Gansu Provincial Hospital, Department of Surgical OncologyLanzhou, China. [Da, Mingxu] Ningxia Medical University, College of Clinical MedicineYinchuan, China. RP Da, M (reprint author), Ningxia Medical University, College of Clinical Medicine, Yinchuan, China. EM hxdamingxu@hotmail.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Mph RLS, Mph KDM, and Dvm AJ. Cancer Statistics. CA Cancer J Clin, 2020, 2020(1):70, 2020 . PhD., DOI 10.3322/caac.21590 Chen XS, Moon JJ, and Cheon J. New Opportunities in Cancer Immunotherapy and Theranostics. Acc Chem Res, 2020, 53(12):2763–4., DOI 10.1021/acs.accounts.0c00724 Zhao Z, Zheng L, Chen W, Weng W, Song J, and Ji J. Delivery Strategies of Cancer Immunotherapy: Recent Advances and Future Perspectives. J Hematol Oncol, 2019, 12(1):126., DOI 10.1186/s13045-019-0817-3 Davies D, and Maher J. Crosstown Traffic: Lymphodepleting Chemotherapy Drives CAR T Cells. Cancer Cell, 2020, 39(2):138–40., DOI 10.1016/ j.ccell.2020.12.019 Gong N, Sheppard NC, Billingsley MM, June CH, and Mitchell MJ. Nanomaterials for T-Cell Cancer Immunotherapy. Nat Nanotechnol, 2021, 16(1):25–36., DOI 10.1038/s41565-020-00822-y Braun D, Bakouny Z, Hirsch L, Flippot R, Van Allen E, Wu C, et al. Beyond Conventional Immune-Checkpoint Inhibition - Novel Immunotherapies for Renal Cell Carcinoma. Nat Rev Clin Oncol, 2021). 18(4):199–214., DOI 10.1038/ s41571-020-00455-z Robert C, Ribas A, Schachter J, Arance A, Grob J-J, Mortier L, et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma, KEYNOTE- 006): post-hoc 5-year Results from an Open-Label, Multicentre, Randomised, Controlled, Phase 3 Study. Lancet Oncol, 2019, 20(9): 1239–51., DOI 10.1016/s1470-2045(19)30388-2 Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, et al. Nivolumab in Previously Untreated Melanoma withoutBRAFMutation. N Engl J Med, 2015, 372(4):320–30., DOI 10.1056/nejmoa1412082 Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N Engl J Med, 2010, 363(8):711–23., DOI 10.1056/nejmoa1003466 Galuppini F, Dal Pozzo CA, Deckert J, Loupakis F, Fassan M, and Baffa R. Tumor Mutation burden: from Comprehensive Mutational Screening to the Clinic. Cancer Cel Int, 2019, 19:209., DOI 10.1186/s12935-019-0929-4 Valero C, Lee M, Hoen D, Wang J, Nadeem Z, Patel N, et al. The Association between TumorMutational burden and Prognosis Is Dependent on Treatment Context. Nat Genet, 2021, 53(1):11–5., DOI 10.1038/s41588-020-00752-4 Yarchoan M, Hopkins A, and Jaffee EM. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N Engl J Med, 2017, 377(25):2500–1., DOI 10.1056/nejmc1713444 Robbins PF, Lu Y-C, El-Gamil M, Li YF, Gross C, Gartner J, et al. Mining Exomic Sequencing Data to Identify Mutated Antigens Recognized by Adoptively Transferred Tumor-Reactive T Cells. Nat Med, 2013, 19(6): 747–52., DOI 10.1038/nm.3161 Martinez-Jimenez F, Muinos F, Sentis I, Deu-Pons J, Reyes-Salazar I, Arnedo- Pac C, et al. A Compendium of Mutational Cancer Driver Genes. Nat Rev Cancer, 2020, 20(10):555–72., DOI 10.1038/s41568-020-0290-x Bailey MH, Tokheim C, Porta-Pardo E, Sengupta S, Bertrand D, Weerasinghe A, et al. Comprehensive Characterization of Cancer Driver Genes and Mutations. Cell, 2018, 174(4):1034–5., DOI 10.1016/j.cell.2018.07.034 Alborelli I, Leonards K, Rothschild SI, Leuenberger LP, Savic Prince S, Mertz KD, et al. Tumor Mutational burden Assessed by Targeted NGS Predicts Clinical Benefit from Immune Checkpoint Inhibitors in Nonsmall Cell Lung Cancer. J Pathol, 2020, 250(1):19–29., DOI 10.1002/ path.5344 Chen Y, Liu Q, Chen Z, Wang Y, Yang W, Hu Y, et al. PD-L1 Expression and TumorMutational burden Status for Prediction of Response to Chemotherapy and Targeted Therapy in Non-small Cell Lung Cancer. J Exp Clin Cancer Res, 2019, 38(1):193., DOI 10.1186/s13046-019-1192-1 Zhang Y, Chang L, Yang Y, Fang W, Guan Y, Wu A, et al. The Correlations of Tumor Mutational burden Among Single-Region Tissue, Multi-Region Tissues and Blood in Non-small Cell Lung Cancer. J Immunotherapy Cancer, 2019, 7(1):98., DOI 10.1186/s40425-019-0581-5 Karn T, Denkert C, Weber KE, Holtrich U, Hanusch C, Sinn BV, et al. Tumor Mutational burden and Immune Infiltration as Independent Predictors of Response to Neoadjuvant Immune Checkpoint Inhibition in Early TNBC in GeparNuevo. Ann Oncol, 2020, 31(9):1216–22., DOI 10.1016/ j.annonc.2020.05.015 Barroso-Sousa R, Keenan TE, Pernas S, Exman P, Jain E, Garrido-Castro AC, et al. Tumor Mutational Burden and PTEN Alterations as Molecular Correlates of Response to PD-1/L1 Blockade in Metastatic Triple-Negative Breast Cancer. Clin Cancer Res, 2020, 26(11):2565–72., DOI 10.1158/1078- 0432.ccr-19-3507 Marsavela G, Johansson PA, Pereira MR, McEvoy AC, Reid AL, Robinson C, et al. The Prognostic Impact of Circulating Tumour DNA in Melanoma Patients Treated with Systemic Therapies-Beyond BRAF Mutant Detection. Cancers, Basel,, 2020, 12(12):3793., DOI 10.3390/cancers12123793 Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 Human Cancer Genomes Reveals the Landscape of Tumor Mutational burden. Genome Med, 2017, 9(1):34., DOI 10.1186/s13073-017- 0424-2 Chen B, Khodadoust MS, Liu CL, Newman AM, and Alizadeh AA. Profiling Tumor Infiltrating Immune Cells with CIBERSORT. Methods Mol Biol, Clifton, NJ,, 2018, 1711:243–59., DOI 10.1007/978-1-4939-7493-1_12 Kang Y-K, Boku N, Satoh T, Ryu M-H, Chao Y, Kato K, et al. Nivolumab in Patients with Advanced Gastric or Gastro-Oesophageal junction Cancer Refractory to, or Intolerant of, at Least Two Previous Chemotherapy Regimens, ONO-4538-12, ATTRACTION-2): a Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. The Lancet, 2017, 390(10111):2461–71., DOI 10.1016/s0140-6736(17)31827-5 Wang R, Yang Y, Ye WW, Xiang J, Chen S, Zou WB, et al. Case Report: Significant Response to Immune Checkpoint Inhibitor Camrelizumab in a Heavily Pretreated Advanced ER+/HER2- Breast Cancer Patient with High Tumor Mutational Burden. Front Oncol, 2020, 10:588080., DOI 10.3389/ fonc.2020.588080 Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, et al. Tumor Mutational burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol Cancer Ther, 2017, 16(11): 2598–608., DOI 10.1158/1535-7163.mct-17-0386 Ciferri A, and Crumbliss AL. The Assembling and Contraction Mechanisms of Striated Muscles. Front Chem, 2018, 6:570., DOI 10.3389/fchem.2018.00570 Jia Q, Wang J, He N, He J, and Zhu B. Titin Mutation Associated with Responsiveness to Checkpoint Blockades in Solid Tumors. JCI insight, 2019, 4(10):e127901., DOI 10.1172/jci.insight.127901 Li X, Pasche B, Zhang W, and Chen K. Association of MUC16 Mutation with Tumor Mutation Load and Outcomes in Patients with Gastric Cancer. JAMA Oncol, 2018, 4(12):1691–8., DOI 10.1001/jamaoncol.2018.2805 Zhao D-Y, Sun X-Z, and Yao S-K. Mining the Cancer Genome Atlas Database for Tumor Mutation burden and its Clinical Implications in Gastric Cancer. Wjgo, 2021, 13(1):37–57., DOI 10.4251/wjgo.v13.i1.37 Yu J, Zhang Q, Wang M, Liang S, Huang H, Xie L, et al. Comprehensive Analysis of Tumor Mutation burden and Immune Microenvironment in Gastric Cancer. Biosci Rep, 2021, 41(2)., DOI 10.1042/bsr20203336 Wang D, Wang N, Li X, Chen X, Shen B, Zhu D, et al. TumorMutation burden as a Biomarker in Resected Gastric Cancer via its Association with Immune Infiltration and Hypoxia. Gastric Cancer, 2021, 24(4):823–34., DOI 10.1007/ s10120-021-01175-8 Herbst RS, Giaccone G, de Marinis F, Reinmuth N, Vergnenegre A, Barrios CH, et al. Atezolizumab for First-Line Treatment of PD-L1-Selected Patients with NSCLC. N Engl J Med, 2020, 383(14):1328–39., DOI 10.1056/ nejmoa1917346 Gandara DR, Paul SM, Kowanetz M, Schleifman E, Zou W, Li Y, et al. Bloodbased Tumor Mutational burden as a Predictor of Clinical Benefit in Nonsmall- cell Lung Cancer Patients Treated with Atezolizumab. Nat Med, 2018, 24(9):1441–8., DOI 10.1038/s41591-018-0134-3 Reynolds SD, Reynolds PR, Pryhuber GS, Finder JD, and Stripp BR. Secretoglobins SCGB3A1 and SCGB3A2 Define Secretory Cell Subsets in Mouse and Human Airways. Am J Respir Crit Care Med, 2002, 166(11): 1498–509., DOI 10.1164/rccm.200204-285oc Tian R, Hu J, Ma X, Liang L, and Guo S. Immune-related Gene Signature Predicts Overall Survival of Gastric Cancer Patients with Varying Microsatellite Instability Status. Aging, 2020, 13(2):2418–35., DOI 10.18632/ aging.202271 Ho C-M, Huang C-J, Huang C-Y, Wu Y-Y, Chang S-F, and Cheng W-F. Promoter Methylation Status of HIN-1 Associated with Outcomes of Ovarian clear Cell Adenocarcinoma. Mol Cancer, 2012, 11:53., DOI 10.1186/1476-4598- 11-53 Zhang ZY, Zhang JL, Zhao LX, Yang Y, Guo R, Zhou N, et al. NAA10 Promotes Proliferation of Renal Cell Carcinoma by Upregulating UPK1B. Eur Rev Med Pharmacol Sci, 2020, 24(22):11553–60., DOI 10.26355/ eurrev_202011_23796 Meynet O, Scotlandi K, Pradelli E, Manara MC, Colombo MP, Schmid- Antomarchi H, et al. Xg Expression in Ewing’s Sarcoma Is of Prognostic Value and Contributes to Tumor Invasiveness. Cancer Res, 2010, 70(9): 3730–8., DOI 10.1158/0008-5472.can-09-2837 Jiao X, Shu G, Liu H, Zhang Q, Ma Z, Ren C, et al. The Diagnostic Value of Chemokine/Chemokine Receptor Pairs in Hepatocellular Carcinoma and Colorectal Liver Metastasis. J Histochem Cytochem, 2019, 67(5):299–308., DOI 10.1369/0022155418824274 Zhang L, Huo Q, Ge C, Zhao F, Zhou Q, Chen X, et al. ZNF143-Mediated H3K9 Trimethylation Upregulates CDC6 by Activating MDIG in Hepatocellular Carcinoma. Cancer Res, 2020, 80(12):2599–611., DOI 10.1158/0008-5472.can-19-3226 Wu C, Su J, Wang X, Wang J, Xiao K, Li Y, et al. Overexpression of the Phospholipase A2 Group V Gene in Glioma Tumors Is Associated with Poor Patient Prognosis. Cmar, 2019, Vol. 11:3139–52., DOI 10.2147/ cmar.s199207 Gracia-Maldonado G, Clark J, Burwinkel M, Greenslade B, Wunderlich M, Salomonis N, et al. LAMP-5 Is an Essential Inflammatory-Signaling Regulator and Novel Immunotherapy Target for Mixed Lineage Leukemia-Rearranged Acute Leukemia. Haematologica, 2021)., DOI 10.3324/haematol.2020.257451 Zhao M, Li M, Chen Z, Bian Y, Zheng Y, Hu Z, et al. Identification of Immune- Related Gene Signature Predicting Survival in the Tumor Microenvironment of Lung Adenocarcinoma. Immunogenetics, 2020, 72:455–65., DOI 10.1007/ s00251-020-01189-z Li S, Guo R, Peng Z, Quan B, Hu Y, Wang Y, et al. NPR3, Transcriptionally Regulated by POU2F1, Inhibits Osteosarcoma Cell Growth through Blocking the PI3K/AKT Pathway. Cell Signal, 2021, 86:110074., DOI 10.1016/ j.cellsig.2021.110074 Huang H, Reed CP, Zhang JS, Shridhar V, Wang L, and Smith DI. Carboxypeptidase A3, CPA3): a Novel Gene Highly Induced by Histone Deacetylase Inhibitors during Differentiation of Prostate Epithelial Cancer Cells. Cancer Res, 1999, 59(12):2981–8. Tiwari A, Tashiro K, Dixit A, Soni A, Vogel K, Hall B, et al. Loss of HIF1A from Pancreatic Cancer Cells Increases Expression of PPP1R1B and Degradation of P53 to Promote Invasion and Metastasis. Gastroenterology, 2020, 159(5): 1882–97.e5., DOI 10.1053/j.gastro.2020.07.046 Stahl D, Gentles AJ, Thiele R, and Gutgemann I. Prognostic Profiling of the Immune Cell Microenvironment in Ewing´s Sarcoma Family of Tumors. Oncoimmunology, 2019, 8(12):e1674113., DOI 10.1080/2162402x.2019.1674113 Xu F, Zhang H, Chen J, Lin L, and Chen Y. Immune Signature of T Follicular Helper Cells Predicts Clinical Prognostic and Therapeutic Impact in Lung Squamous Cell Carcinoma. Int Immunopharmacology, 2020, 81:105932., DOI 10.1016/j.intimp.2019.105932 Chaumette B, Kebir O, Dion PA, Rouleau GA, and Krebs M-O. Reliability and Correlation of Mixture Cell Correction in Methylomic and Transcriptomic Blood Data. BMC Res Notes, 2020, 13(1):74., DOI 10.1186/s13104-020-4936-2 Tang C, Ma J, Liu X, and Liu Z. Development and Validation of a Novel Stem Cell Subtype for Bladder Cancer Based on Stem Genomic Profiling. Stem Cel Res Ther, 2020, 11(1):457., DOI 10.1186/s13287-020-01973-4 Li F, Sun Y, Huang J, Xu W, Liu J, and Yuan Z. CD4/CD8 + T Cells, DC Subsets, Foxp3, and Ido Expression Are Predictive Indictors of Gastric Cancer Prognosis. Cancer Med, 2019, 8(17):7330–44., DOI 10.1002/cam4.2596 Xia Y, Rao L, Yao H,Wang Z, Ning P, and Chen X. Engineering Macrophages for Cancer Immunotherapy and Drug Delivery. Adv Mater, 2020, 32(40): e2002054., DOI 10.1002/adma.202002054 Qiu Q, Li C, Song Y, Shi T, Luo X, Zhang H, et al. Targeted Delivery of Ibrutinib to Tumor-Associated Macrophages by Sialic Acid-Stearic Acid Conjugate Modified Nanocomplexes for Cancer Immunotherapy. Acta Biomater, 2019, 92:184–95., DOI 10.1016/j.actbio.2019.05.030 Song Y, Tang C, and Yin C. Combination Antitumor Immunotherapy with VEGF and PIGF siRNA via Systemic Delivery of Multi-Functionalized Nanoparticles to Tumor-Associated Macrophages and Breast Cancer Cells. Biomaterials, 2018, 185:117–32., DOI 10.1016/j.biomaterials.2018.09.017 Gu Y, Liu Y, Fu L, Zhai L, Zhu J, Han Y, et al. Tumor-educated B Cells Selectively Promote Breast Cancer Lymph Node Metastasis by HSPA4- Targeting IgG. Nat Med, 2019, 25(2):312–22., DOI 10.1038/s41591-018- 0309-y Olkhanud PB, Damdinsuren B, Bodogai M, Gress RE, Sen R, Wejksza K, et al. Tumor-Evoked Regulatory B Cells Promote Breast Cancer Metastasis by Converting Resting CD4+ T Cells to T-Regulatory Cells. Cancer Res, 2011, 71(10):3505–15., DOI 10.1158/0008-5472.can-10-4316 Marone G, and Granata F. Angiogenesis, Lymphangiogenesis and Clinical Implications. Preface. Chem Immunol Allergy, 2014, 99:XI–XII., DOI 10.1159/ 000352074 Li L, Ng DSW, Mah W-C, Almeida FF, Rahmat SA, Rao VK, et al. A Unique Role for P53 in the Regulation of M2 Macrophage Polarization. Cell Death Differ, 2015, 22(7):1081–93., DOI 10.1038/cdd.2014.212 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609852 EP 1609866 DI 10.3389/pore.2021.1609852 PG 15 ER PT J AU Diao, B Yang, P AF Diao, Bowen Yang, Ping TI Comprehensive Analysis of the Expression and Prognosis for Laminin Genes in Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ovarian cancer; metastasis; laminins; cancer prognosis; bioinformatical analysis ID ovarian cancer; metastasis; laminins; cancer prognosis; bioinformatical analysis AB Survival is low in ovarian cancer (OC). Most OC patients demonstrate advanced metastases, and recurrence is common. Dysregulation of laminin interactions is associated with cancer development. However, it is unknown whether laminin subunits can be considered as biomarkers for OC diagnosis, prognosis, and treatment. We used cBioPortal, GEO, ONCOMINE, GEPIA, Human Protein Atlas, Kaplan-Meier Plotter, TIMER, and Metascape to determine the associations among laminin expression, prognosis, and immune cell infiltration in OC. LAMA5, LAMB3, and LAMC2 mRNAs and LAMA3, LAMB1/ B2/B3, and LAMC1/C2 proteins were overexpressed in OC tissues compared with normal ovaries. LAMA4, LAMB1, and LAMC1 mRNA upregulation was positively correlated with worse overall survival (OS) and progression-free survival (PFS) in OC. Elevated LAMA2 and LAMC2 mRNA expression levels were related to better PFS or OS, respectively. The results speculated that LAMA5 could potentially be a good prognostic factor in OC. Its expression proves valuable for predicting OS in patients diagnosed with stage Ⅳ and grade 3 OC and PFS in patients diagnosed with all OC stages or grades. LAMB3 and LAMC2 expression was correlated with platinum resistance development. ROC analysis of laminins in OC sets revealed that LAMA2/A4/A5, LAMB1/B2/B3, and LAMC2 could be used to differentiate between malignant tumors and non-neoplastic tissues. LAMA1/A5 and LAMC1 were significantly and negatively correlated with various tumor immune infiltrates (TILs), especially with dendritic cells, CD8+ T cells or neutrophil. LAMA4 and LAMB1 might be associated with tumor purity in OC. Overall, LAMA5 and LAMC1 could help predict OC survival and diagnosis and might be deemed important OC oncogenes. C1 [Diao, Bowen] Shihezi University, School of Medicine, First Affiliated Hospital, Department of GynecologyShihezi, China. [Yang, Ping] Shihezi University, School of Medicine, First Affiliated Hospital, Department of GynecologyShihezi, China. RP Yang, P (reprint author), Shihezi University, School of Medicine, First Affiliated Hospital, Department of Gynecology, Shihezi, China. EM pingy2018@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Wu A, Zhang S, Liu J, Huang Y, Deng W, Shu G, et al. Integrated Analysis of Prognostic and Immune Associated Integrin Family in Ovarian Cancer. Front Genet, 2020, 11:705., DOI 10.3389/fgene.2020.00705 Torre LA, Trabert B, DeSantis CE, Miller KD, Samimi G, Runowicz CD, et al. Ovarian Cancer Statistics. CA: A Cancer J Clinicians, 2018, 68(4):284–96., DOI 10.3322/caac.21456 Krzystyniak J, Ceppi L, Dizon DS, and Birrer MJ. Epithelial Ovarian Cancer: the Molecular Genetics of Epithelial Ovarian Cancer. Ann Oncol, 2016, 27: i4–i10., DOI 10.1093/annonc/mdw083 Bizama C, Benavente F, Salvatierra E, Gutierrez-Moraga A, Espinoza JA, Fernandez EA, et al. The Low-Abundance Transcriptome Reveals Novel Biomarkers, Specific Intracellular Pathways and Targetable Genes Associated With Advanced Gastric Cancer. Int J Cancer, 2014, 134(4): 755–64., DOI 10.1002/ijc.28405 Ye G, Qin Y,Wang S, Pan D, Xu S,Wu C, et al. Lamc1 Promotes theWarburg Effect in Hepatocellular Carcinoma Cells by Regulating PKM2 Expression through AKT Pathway. Cancer Biol Ther, 2019, 20(5):711–9., DOI 10.1080/ 15384047.2018.1564558 Wragg JW, Finnity JP, Anderson JA, Ferguson HJM, Porfiri E, Bhatt RI, et al. MCAM and LAMA4 Are Highly Enriched in Tumor Blood Vessels of Renal Cell Carcinoma and Predict Patient Outcome. Cancer Res, 2016, 76(8): 2314–26., DOI 10.1158/0008-5472.can-15-1364 Shkurnikov M, Nikulin S, Nersisyan S, Poloznikov A, Zaidi S, Baranova A, et al. LAMA4-Regulating miR-4274 and its Host Gene SORCS2 Play a Role in IGFBP6-dependent Effects on Phenotype of Basal-Like Breast Cancer. Front Mol Biosci, 2019, 6:122., DOI 10.3389/fmolb.2019.00122 Bartolini A, Cardaci S, Lamba S, Oddo D, Marchio C, Cassoni P, et al. BCAM and LAMA5 Mediate the Recognition Between Tumor Cells and the Endothelium in the Metastatic Spreading of KRAS-Mutant Colorectal Cancer. Clin Cancer Res, 2016, 22(19):4923–33., DOI 10.1158/1078-0432.ccr- 15-2664 Sathyanarayana UG, Toyooka S, Padar A, Takahashi T, Brambilla E, Minna JD, et al. Epigenetic Inactivation of Laminin-5-Encoding Genes in Lung Cancers. Clin Cancer Res, 2003, 9(7):2665–72., DOI 10.1158/0008-5472.can-03-0701 Fujita M, Khazenzon NM, Bose S, Sekiguchi K, Sasaki T, Carter WG, et al. Overexpression of β1-Chain-Containing Laminins in Capillary Basement Membranes of Human Breast Cancer and its Metastases. Breast Cancer Res, 2005, 7(4):R411–21., DOI 10.1186/bcr1011 Gordon-Weeks A, Lim SY, Yuzhalin A, Lucotti S, Vermeer JAF, Jones K, et al. Tumour-Derived Laminin α5, LAMA5, Promotes Colorectal Liver Metastasis Growth, Branching Angiogenesis and Notch Pathway Inhibition. Cancers, Basel,, 2019, 11(5):630., DOI 10.3390/cancers11050630 Tzu J, Marinkovich MP, and biology c. Bridging Structure With Function: Structural, Regulatory, and Developmental Role of Laminins. Int J Biochem Cel Biol, 2008, 40(2):199–214., DOI 10.1016/j.biocel.2007.07.015 Hao J, Tuck AR, Prakash CR, Damdimopoulos A, Sjodin MOD, Lindberg J, et al. Culture of Human Ovarian Tissue in Xeno-Free Conditions Using Laminin Components of the Human Ovarian Extracellular Matrix. J Assist Reprod Genet, 2020, 37(9):2137–50., DOI 10.1007/s10815-020-01886-4 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Sci Signaling, 2013, 6(269):pl1., DOI 10.1126/ scisignal.2004088 Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, et al. ONCOMINE: a Cancer Microarray Database and Integrated Data-Mining Platform. Neoplasia, 2004, 6(1):1–6., DOI 10.1016/s1476-5586(04)80047-2 Tang Z, Kang B, Li C, Chen T, and Zhang Z. GEPIA2: an Enhanced Web Server for Large-Scale Expression Profiling and Interactive Analysis. Nucleic Acids Res, 2019, 47(W1):W556–W560., DOI 10.1093/nar/gkz430 Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Proteomics. Tissue-Based Map of the Human Proteome. Science, 2015, 347(6220):1260419., DOI 10.1126/science.1260419 Gyorffy B, Lanczky A, and Szallasi Z. Implementing an Online Tool for Genome-Wide Validation of Survival-Associated Biomarkers in Ovarian- Cancer Using Microarray Data from 1287 Patients. Endocr Relat Cancer, 2012, 19(2):197–208., DOI 10.1530/ERC-11-0329 Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. Cancer Res, 2017, 77(21):e108–e110., DOI 10.1158/0008-5472.can-17-0307 Barrett T, Troup DB, Wilhite SE, Ledoux P, Rudnev D, Evangelista C, et al. NCBI GEO: Archive for High-Throughput Functional Genomic Data. Nucleic Acids Res, 2009, 37:D885–D890., DOI 10.1093/nar/gkn764 Tassi RA, Gambino A, Ardighieri L, Bignotti E, Todeschini P, Romani C, et al. FXYD5, Dysadherin, Upregulation Predicts Shorter Survival and Reveals Platinum Resistance in High-Grade Serous Ovarian Cancer Patients. Br J Cancer, 2019, 121(7):584–92., DOI 10.1038/s41416-019-0553-z Meyniel J-P, Cottu PH, Decraene C, Stern M-H, Couturier J, Lebigot I, et al. A Genomic and Transcriptomic Approach for a Differential Diagnosis Between Primary and Secondary Ovarian Carcinomas in Patients With a Previous History of Breast Cancer. BMC Cancer, 2010, 10:222., DOI 10.1186/1471-2407- 10-222 Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape Provides a Biologist-Oriented Resource for the Analysis of Systems- Level Datasets. Nat Commun, 2019, 10(1):1523., DOI 10.1038/s41467-019- 09234-6 Szklarczyk D, Morris JH, Cook H, Kuhn M,Wyder S, Simonovic M, et al. The STRING Database in 2017: Quality-Controlled Protein-Protein Association Networks, Made Broadly Accessible. Nucleic Acids Res, 2017, 45:D362–D368., DOI 10.1093/nar/gkw937 Bobisse S, Genolet R, Roberti A, Tanyi JL, Racle J, Stevenson BJ, et al. Sensitive and Frequent Identification of High Avidity Neo-Epitope Specific CD8 + T Cells in Immunotherapy-Naive Ovarian Cancer. Nat Commun, 2018, 9(1): 1092., DOI 10.1038/s41467-018-03301-0 Santoiemma PP, Reyes C, Wang L-P, McLane MW, Feldman MD, Tanyi JL, et al. Systematic Evaluation of Multiple Immune Markers Reveals Prognostic Factors in Ovarian Cancer. Gynecol Oncol, 2016, 143(1):120–7., DOI 10.1016/ j.ygyno.2016.07.105 Arrighetti N, Cossa G, De Cecco L, Stucchi S, Carenini N, Corna E, et al. PKCAlpha Modulation by miR-483-3p in Platinum-Resistant Ovarian Carcinoma Cells. Toxicol Appl Pharmacol, 2016, 310:9–19., DOI 10.1016/ j.taap.2016.08.005 Warde-Farley D, Donaldson SL, Comes O, Zuberi K, Badrawi R, Chao P, et al. The GeneMANIA Prediction Server: Biological Network Integration for Gene Prioritization and Predicting Gene Function. Nucleic Acids Res, 2010, 38: W214–W220., DOI 10.1093/nar/gkq537 Maltseva D, Raygorodskaya M, Knyazev E, Zgoda V, Tikhonova O, Zaidi S, et al. Knockdown of the α5 Laminin Chain Affects Differentiation of Colorectal Cancer Cells and Their Sensitivity to Chemotherapy. Biochimie, 2020, 174:107–16., DOI 10.1016/j.biochi.2020.04.016 Kuhn E, Kurman RJ, Soslow RA, Han G, Sehdev AS, Morin PJ, et al. The Diagnostic and Biological Implications of Laminin Expression in Serous Tubal Intraepithelial Carcinoma. American Journal of Surgical Pathology, 2012, 36(12):1826–34., DOI 10.1097/pas.0b013e31825ec07a Kunitomi H, Kobayashi Y, Wu RC, Takeda T, Tominaga E, Banno K, et al. LAMC1 Is a Prognostic Factor and a Potential Therapeutic Target in Endometrial Cancer. J Gynecol Oncol, 2020, 31(2):e11., DOI 10.3802/ jgo.2020.31.e11 Zhang Y, Xi S, Chen J, Zhou D, Gao H, Zhou Z, et al. Overexpression of LAMC1 Predicts Poor Prognosis and Enhances Tumor Cell Invasion and Migration in Hepatocellular Carcinoma. J Cancer, 2017, 8(15):2992–3000., DOI 10.7150/jca.21038 Zhou J, Zhang Y, Han Z, Dong Z, Cao T,Wei A, et al. miR-506 Contributes to Malignancy of Cutaneous Squamous Cell Carcinoma via Targeting of P65 and LAMC1. Cell Cycle, 2019, 18(3):333–45., DOI 10.1080/15384101.2019.1568747 Nishikawa R, Goto Y, Kojima S, Enokida H, Chiyomaru T, Kinoshita T, et al. Tumor-Suppressive microRNA-29s Inhibit Cancer Cell Migration and Invasion via Targeting LAMC1 in Prostate Cancer. Int J Oncol, 2014, 45(1):401–10., DOI 10.3892/ijo.2014.2437 Peng X, Yu M, and Chen J. Transcriptome Sequencing Identifies Genes Associated with Invasion of Ovarian Cancer. J Int Med Res, 2020, 48(9): 300060520950912., DOI 10.1177/0300060520950912 Zhang D, Guo H, FengW, and Qiu H. LAMC2 Regulated by microRNA-125a- 5p Accelerates the Progression of Ovarian Cancer via Activating P38 MAPK Signalling. Life Sci, 2019, 232:116648., DOI 10.1016/j.lfs.2019.116648 Qiu L, Wang J, Chen M, Chen F, and Tu W. Exosomal MicroRNA-146a Derived from Mesenchymal Stem Cells Increases the Sensitivity of Ovarian Cancer Cells to Docetaxel and Taxane via a LAMC2-Mediated PI3K/Akt axis. Int J Mol Med, 2020, 46(2):609–20., DOI 10.3892/ijmm.2020.4634 Jiang P, He S, Li Y, and Xu Z. Identification of Therapeutic and Prognostic Biomarkers of Lamin C, LAMC, Family Members in Head and Neck Squamous Cell Carcinoma. Med Sci Monit, 2020, 26:e925735., DOI 10.12659/MSM.925735 Huang D, Du C, Ji D, Xi J, and Gu J. Overexpression of LAMC2 Predicts Poor Prognosis in Colorectal Cancer Patients and Promotes Cancer Cell Proliferation, Migration, and Invasion. Tumour Biol, 2017, 39(6): 1010428317705849., DOI 10.1177/1010428317705849 Wang Y, Shi M, Yang N, Zhou X, and Xu L. GPR115 Contributes to Lung Adenocarcinoma Metastasis Associated With LAMC2 and Predicts a Poor Prognosis. Front Oncol, 2020, 10:577530., DOI 10.3389/fonc.2020.577530 Yang JL, Wang CCN, Cai JH, Chou CY, Lin YC, and Hung CC. Identification of GSN and LAMC2 as Key Prognostic Genes of Bladder Cancer by Integrated Bioinformatics Analysis. Cancers, Basel,, 2020, 12(7):1809., DOI 10.3390/ cancers12071809 Li L, Shirkey MW, Zhang T, Xiong Y, Piao W, Saxena V, et al. The Lymph Node Stromal Laminin α5 Shapes Alloimmunity. J Clin Invest, 2020, 130(5): 2602–19., DOI 10.1172/jci135099 Simon T, Li L, Wagner C, Zhang T, Saxena V, Brinkman CC, et al. Differential Regulation of T-Cell Immunity and Tolerance by Stromal Laminin Expressed in the Lymph Node. Node, 2019, 103(10):2075–89., DOI 10.1097/ tp.0000000000002774 Kenne E, Soehnlein O, Genove G, Rotzius P, Eriksson EE, and Lindbom L. Immune Cell Recruitment to Inflammatory Loci Is Impaired in Mice Deficient in Basement Membrane Protein Laminin α4. J Leukoc Biol, 2010, 88(3):523–8., DOI 10.1189/jlb.0110043 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609855 EP 1609867 DI 10.3389/pore.2021.1609855 PG 13 ER PT J AU Jain, D Guleria, P Singh, V Parshad, R Kumar, S Gaiser, T Kurz, K Ott, G Porubsky, S Preissler, G Sauer, Ch Scholch, S Strobel, P Hielscher, Th Marx, A Popovic, Z AF Jain, Deepali Guleria, Prerna Singh, Varsha Parshad, Rajinder Kumar, Sunil Gaiser, Timo Kurz, S. Katrin Ott, German Porubsky, Stefan Preissler, Gerhard Sauer, G. Christian Scholch, Sebastian Strobel, Philipp Hielscher, Thomas Marx, Alexander Popovic, V. Zoran TI GTF2I Mutation in Thymomas: Independence From Racial-Ethnic Backgrounds. An Indian/German Comparative Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE epidemiology; thymoma; GTF2I mutation; myasthenia gravis; racial-ethnic factors ID epidemiology; thymoma; GTF2I mutation; myasthenia gravis; racial-ethnic factors AB Thymomas are the most frequent adult mediastinal cancers. Their etiology is unknown and their pathogenesis poorly understood. Racial, ethnic and environmental factors influence tumorigenesis in many cancers, but their role in thymomas remains unclear to date. In this study that included pretreatment thymoma cases from India and Germany (n = 37 and n = 77, respectively) we compared i) the prevalence of the thymoma-specific chromosome 7 c.74146970T > A mutation of the GTF2I gene in type A and AB thymomas; ii) epidemiological features; and iii) the frequency of myasthenia gravis (MG). Due to a known predominance of GTF2I mutation in A and AB histotypes, we included only a marginal number of type B thymomas as a control group in both cohorts. While the distribution of histological types between the cohorts was similar (p = 0.1622), Indian patients were strikingly younger (p < 0.0001; median age 50 vs. 65 years) and showed significantly lower tumour stage (Masaoka-Koga stage I) at primary diagnosis (p = 0.0005) than the German patients. In patients with known MG status (n = 17 in Indian and n = 25 in German cohort), a clear trend towards more frequent MG was observed in the Indian group (p = 0.0504; 48 vs. 82%). The prevalence of the GTF2I mutation (analysed in n = 34 Indian and n = 77 German patients) was identical in the two cohorts. We conclude that racial-ethnic and environmental factors do not significantly influence the most common molecular feature of thymomas but may have an impact on the timing of clinical presentation. C1 [Jain, Deepali] All India Institute of Medical Sciences, Department of PathologyNew Delhi, India. [Guleria, Prerna] All India Institute of Medical Sciences, Department of PathologyNew Delhi, India. [Singh, Varsha] All India Institute of Medical Sciences, Department of PathologyNew Delhi, India. [Parshad, Rajinder] All India Institute of Medical Sciences, Department of SurgeryNew Delhi, India. [Kumar, Sunil] All India Institute of Medical Sciences, Dr BRA Institute-Rotary Cancer Hospital, Department of Surgical OncologyNew Delhi, India. [Gaiser, Timo] University of Heidelberg, University Medical Centre Mannheim, Institute of PathologyMannheim, Germany. [Kurz, S. Katrin] Dr. Margarete Fischer-Bosch Institute for Clinical PharmacologyStuttgart, Germany. [Ott, German] Dr. Margarete Fischer-Bosch Institute for Clinical PharmacologyStuttgart, Germany. [Porubsky, Stefan] University of Heidelberg, University Medical Centre Mannheim, Institute of PathologyMannheim, Germany. [Preissler, Gerhard] Robert-Bosch-Krankenhaus, Schillerhohe Clinics, Department of Thoracic SurgeryGerlingen, Germany. [Sauer, G. Christian] University of Heidelberg, University Medical Centre Mannheim, Institute of PathologyMannheim, Germany. [Scholch, Sebastian] University of Heidelberg, University Medical Centre Mannheim, Department of SurgeryMannheim, Germany. [Strobel, Philipp] University of Gottingen, Institute of PathologyGottingen, Germany. [Hielscher, Thomas] German Cancer Research Center (DKFZ), Department of BiostatisticsHeidelberg, Germany. [Marx, Alexander] University of Heidelberg, University Medical Centre Mannheim, Institute of PathologyMannheim, Germany. [Popovic, V. Zoran] University of Heidelberg, University Medical Centre Mannheim, Institute of PathologyMannheim, Germany. RP Jain, D (reprint author), All India Institute of Medical Sciences, Department of Pathology, New Delhi, India. EM deepalijain76@gmail.com CR Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, et al. The 2015 World Health Organization Classification of Lung Tumors. J Thorac Oncol, 2015, 10(9):1243–60., DOI 10.1097/ jto.0000000000000630 Badve S, Goswami C, Gokmen–Polar Y, Nelson RP, Henley J, Miller N, et al. Molecular Analysis of Thymoma. PLoS One, 2012, 7(8):e42669., DOI 10.1371/ journal.pone.0042669 Enkner F, Pichlhofer B, Zaharie AT, Krunic M, Holper TM, Janik S, et al. Molecular Profiling of Thymoma and Thymic Carcinoma: Genetic Differences and Potential Novel Therapeutic Targets. Pathol Oncol Res, 2017, 23(3): 551–64., DOI 10.1007/s12253-016-0144-8 Girard N, Shen R, Guo T, Zakowski MF, Heguy A, Riely GJ, et al. Comprehensive Genomic Analysis Reveals Clinically Relevant Molecular Distinctions between Thymic Carcinomas and Thymomas. Clin Cancer Res, 2009, 15(22):6790–9., DOI 10.1158/1078-0432.ccr-09-0644 Lee H-S, Jang H-J, Shah R, Yoon D, Hamaji M, Wald O, et al. Genomic Analysis of Thymic Epithelial Tumors Identifies Novel Subtypes Associated with Distinct Clinical Features. Clin Cancer Res, 2017, 23(16):4855–64., DOI 10.1158/1078-0432.ccr-17-0066 Radovich M, Pickering CR, Felau I, Ha G, Zhang H, Jo H, et al. The Integrated Genomic Landscape of Thymic Epithelial Tumors. Cancer Cell, 2018, 33(2): 244–e10., DOI 10.1016/j.ccell.2018.01.003 Feng Y, Lei Y, Wu X, Huang Y, Rao H, Zhang Y, et al. GTF2I Mutation Frequently Occurs in More Indolent Thymic Epithelial Tumors and Predicts Better Prognosis. Lung Cancer, 2017, 110:48–52., DOI 10.1016/ j.lungcan.2017.05.020 Petrini I, Meltzer PS, Kim I-K, Lucchi M, Park K-S, Fontanini G, et al. A Specific Missense Mutation in GTF2I Occurs at High Frequency in Thymic Epithelial Tumors. Nat Genet, 2014, 46(8):844–9., DOI 10.1038/ng.3016 Detterbeck FC, Nicholson AG, Kondo K, Van Schil P, and Moran C. The Masaoka-Koga Stage Classification for Thymic Malignancies: Clarification and Definition of Terms. J Thorac Oncol, 2011, 6(7 Suppl. 3):S1710–6., DOI 10.1097/JTO.0b013e31821e8cff Porubsky S, Popovic ZV, Badve S, Banz Y, Berezowska S, Borchert D, et al. Thymic Hyperplasia with Lymphoepithelial Sialadenitis, LESA)- Like Features: Strong Association with Lymphomas and Nonmyasthenic Autoimmune Diseases. Cancers, Basel,, 2021, 13(2)., DOI 10.3390/cancers13020315 Weis C-A, Yao X, Deng Y, Detterbeck FC,Marino M, Nicholson AG, et al. The Impact of Thymoma Histotype on Prognosis in a Worldwide Database. J Thorac Oncol, 2015, 10(2):367–72., DOI 10.1097/jto.0000000000000393 Reich D, Thangaraj K, Patterson N, Price AL, and Singh L. Reconstructing Indian Population History. Nature, 2009, 461(7263):489–94., DOI 10.1038/ nature08365 Bhupathiraju SN, Guasch-Ferre M, Gadgil MD, Newgard CB, Bain JR, Muehlbauer MJ, et al. Dietary Patterns Among Asian Indians Living in the United States Have Distinct Metabolomic Profiles that Are Associated with Cardiometabolic Risk. J Nutr, 2018, 148(7):1150–9., DOI 10.1093/jn/nxy074 Detterbeck FC. Clinical Value of the WHO Classification System of Thymoma. Ann Thorac Surg, 2006, 81(6):2328–34., DOI 10.1016/ j.athoracsur.2005.11.067 Ruffini E, Guerrera F, Brunelli A, Passani S, Pellicano D, Thomas P, et al. Report from the European Society of Thoracic Surgeons Prospective Thymic Database 2017: a Powerful Resource for a Collaborative Global Effort to Manage Thymic Tumours. Eur J Cardiothorac Surg, 2019, 55(4):601–9., DOI 10.1093/ejcts/ezy448 Strobel P, Bauer A, Puppe B, Kraushaar T, Krein A, Toyka K, et al. Tumor Recurrence and Survival in Patients Treated for Thymomas and Thymic Squamous Cell Carcinomas: a Retrospective Analysis. Jco, 2004, 22(8): 1501–9., DOI 10.1200/jco.2004.10.113 Kim DJ, Yang WI, Choi SS, Kim KD, and Chung KY. Prognostic and Clinical Relevance of the World Health Organization Schema for the Classification of Thymic Epithelial Tumors. Chest, 2005, 127(3):755–61., DOI 10.1378/ chest.127.3.755 Boldingh MI, Maniaol A, Brunborg C, Dekker L, Lipka A, Niks EH, et al. Prevalence and Clinical Aspects of Immigrants with Myasthenia Gravis in Northern Europe. Muscle Nerve, 2017, 55(6):819–27., DOI 10.1002/mus.25408 Huang X, Liu WB, Men LN, Feng HY, Li Y, Luo CM, et al. Clinical Features of Myasthenia Gravis in Southern China: a Retrospective Review of 2,154 Cases over 22 Years. Neurol Sci, 2013, 34(6):911–7., DOI 10.1007/s10072-012-1157-z Murai H, Yamashita N,Watanabe M, Nomura Y, Motomura M, Yoshikawa H, et al. Characteristics of Myasthenia Gravis According to Onset-Age: Japanese Nationwide Survey. J Neurol Sci, 2011, 305(1-2):97–102., DOI 10.1016/ j.jns.2011.03.004 Yamada Y, Simon-Keller K, Belharazem-Vitacolonnna D, Bohnenberger H, Kriegsmann M, Kriegsmann K, et al. A Tuft Cell-like Signature Is Highly Prevalent in Thymic Squamous Cell Carcinoma and Delineates New Molecular Subsets Among the Major Lung Cancer Histotypes. J Thorac Oncol, 2021)., DOI 10.1016/j.jtho.2021.02.008 Guleria P, Parshad R, Malik PS, Ray R, Pandey RM, and Jain D. Histotyping of Indian Thymomas: A Clinicopathologic Study from north India. Indian J Med Res, 2019, 150(2):153–60., DOI 10.4103/ ijmr.IJMR_530_18 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609858 EP 1609867 DI 10.3389/pore.2021.1609858 PG 10 ER PT J AU An, Y Zhou, J Lin, G Wu, H Cong, L Li, Y Qiu, X Shi, W AF An, Yang Zhou, Jiaolin Lin, Guole Wu, Huanwen Cong, Lin Li, Yunhao Qiu, Xiaoyuan Shi, Weikun TI Clinicopathological and Molecular Characteristics of Colorectal Signet Ring Cell Carcinoma: A Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE colorectal cancer; clinicopathology; signet ring cell carcinoma; molecular features; review ID colorectal cancer; clinicopathology; signet ring cell carcinoma; molecular features; review AB Colorectal signet ring cell carcinoma (SRCC) is a rare subtype of colorectal cancer (CRC) with unique characteristics. Due to the limited researches on it, a comprehensive and indepth understanding of this subtype is still lacking. In this article, we summarize the clinicopathological features and molecular characteristics of colorectal SRCC based on a literature review. Clinically, SRCC has been associated with young age, proximal site preference, advanced tumor stage, high histological grade, high rate of lymph node involvement, frequent peritoneal metastasis, and a significantly poor prognosis. Regarding molecular characteristics, in SRCC, the mutation burden of the classic signaling pathways that include WNT/β-catenin, RAS/RAF/MAPK, and PI3K/AKT/mTOR signaling pathways are generally reduced. In contrast, some genes related to the “epithelial-mesenchymal transition (EMT) process” and the “stem cell properties”, including RNF43, CDH1, and SMAD4, as well as the related TGF-β signaling pathway have been observed more frequently altered in SRCC than in conventional adenocarcinoma (AC). In many studies but not in others, SRCC showed a higher frequency of BRAF mutation, microsatellite instability-high (MSI-H) and CpG island methylator phenotype (CIMP) positive status compared to AC. It has been proposed that colorectal SRCC consists of two subtypes, in which the MSI+/CIMP+/BRAF+/CD3+/PD-L1+ hypermethylated genotype is more common in the proximal colon, and may represent the potential candidate for immunotherapy. Understanding the special molecular mechanisms related to the aggressive biology of SRCC is of great importance, which may provide a theoretical basis for the development of more targeted and effective treatments for this refractory disease. C1 [An, Yang] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General SurgeryBeijing, China. [Zhou, Jiaolin] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General SurgeryBeijing, China. [Lin, Guole] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General SurgeryBeijing, China. [Wu, Huanwen] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of PathologyBeijing, China. [Cong, Lin] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General SurgeryBeijing, China. [Li, Yunhao] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General SurgeryBeijing, China. [Qiu, Xiaoyuan] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General SurgeryBeijing, China. [Shi, Weikun] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General SurgeryBeijing, China. RP Zhou, J (reprint author), Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of General Surgery, Beijing, China. EM conniezhjl@163.com CR Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA, et al. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology, 2020, 159(1):335–49., DOI 10.1053/j.gastro.2020.02.068 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Laufman H, and Saphir O. Primary Linitis Plastica Type of Carcinoma of the colon. Arch Surg, 1951, 62(1):79–91., DOI 10.1001/ archsurg.1951.01250030082009 Shi T, Huang M, Han D, Tang X, Chen Y, Li Z, et al. Chemotherapy Is Associated with Increased Survival from Colorectal Signet Ring Cell Carcinoma with Distant Metastasis: A Surveillance, Epidemiology, and End Results Database Analysis. Cancer Med, 2019, 8(4):1930–40., DOI 10.1002/ cam4.2054 Hugen N, Verhoeven RH, Lemmens VE, van Aart CJ, Elferink MA, Radema SA, et al. Colorectal Signet-Ring Cell Carcinoma: Benefit from Adjuvant Chemotherapy but a Poor Prognostic Factor. Int J Cancer, 2015, 136(2): 333–9., DOI 10.1002/ijc.28981 Hyngstrom JR, Hu C-Y, Xing Y, You YN, Feig BW, Skibber JM, et al. Clinicopathology and Outcomes for Mucinous and Signet Ring Colorectal Adenocarcinoma: Analysis from the National Cancer Data Base. Ann Surg Oncol, 2012, 19(9):2814–21., DOI 10.1245/s10434-012-2321-7 Benesch MGK, and Mathieson A. Epidemiology of Signet Ring Cell Adenocarcinomas. Cancers, 2020, 12(6):1544., DOI 10.3390/cancers12061544 Nitsche U, Zimmermann A, Spath C, Muller T, Maak M, Schuster T, et al. Mucinous and Signet-Ring Cell Colorectal Cancers Differ from Classical Adenocarcinomas in Tumor Biology and Prognosis. Ann Surg, 2013, 258(5):775–83., DOI 10.1097/SLA.0b013e3182a69f7e Tawadros PS, Paquette IM, Hanly AM, Mellgren AF, Rothenberger DA, and Madoff RD. Adenocarcinoma of the Rectum in Patients under Age 40 Is Increasing. Dis Colon Rectum, 2015, 58(5):474–8., DOI 10.1097/ DCR.0000000000000318 Siegel RL, Jemal A, and Ward EM. Increase in Incidence of Colorectal Cancer Among Young Men and Women in the United States. Cancer Epidemiol Biomarkers Prev, 2009, 18(6):1695–8., DOI 10.1158/1055-9965.EPI-09-0186 Bailey CE, Hu C-Y, You YN, Bednarski BK, Rodriguez-Bigas MA, Skibber JM, et al. Increasing Disparities in the Age-Related Incidences of colon and Rectal Cancers in the United States, 1975-2010. JAMA Surg, 2015, 150(1):17–22., DOI 10.1001/jamasurg.2014.1756 Meyer JE, Narang T, Schnoll-Sussman FH, Pochapin MB, Christos PJ, and Sherr DL. Increasing Incidence of Rectal Cancer in Patients Aged Younger Than 40 Years. Cancer, 2010, 116(18):4354–9., DOI 10.1002/cncr.25432 Arai T. Where Does Signet-Ring Cell Carcinoma Come from and where Does it Go? Gastric Cancer, 2019, 22(4):651–2., DOI 10.1007/s10120-019-00960-w Nitsche U, FriessH, Agha A, Angele M, Eckel R, HeitlandW, et al. Prognosis of Mucinous and Signet-Ring Cell Colorectal Cancer in a Population-Based Cohort. J Cancer Res Clin Oncol, 2016, 142(11):2357–66., DOI 10.1007/ s00432-016-2224-2 Pande R, Sunga A, Levea C, Wilding GE, Bshara W, Reid M, et al. Significance of Signet-Ring Cells in Patients with Colorectal Cancer. Dis Colon Rectum, 2008, 51(1):50–5., DOI 10.1007/s10350-007-9073-7 Kang H, O’Connell JB, Maggard MA, Sack J, and Ko CY. A 10-year Outcomes Evaluation of Mucinous and Signet-Ring Cell Carcinoma of the colon and Rectum. Dis Colon Rectum, 2005, 48(6):1161–8., DOI 10.1007/s10350-004- 0932-1 Zhou JL, Zhao XY, Lin GL, Qiu HZ, Xiao Y, Wu B, et al. [Clinicopathological Characteristics, Diagnosis, and Treatment of 29 Cases of Signet Ring Cell Carcinoma of the Rectum and Sigmoid colon]. Zhonghua Zhong Liu Za Zhi, 2020, 42(10):897–902., DOI 10.3760/cma.j.cn112152-20200228-00142 Chang DT, Pai RK, Rybicki LA, Dimaio MA, Limaye M, Jayachandran P, et al. Clinicopathologic and Molecular Features of Sporadic Early-Onset Colorectal Adenocarcinoma: an Adenocarcinoma with Frequent Signet Ring Cell Differentiation, Rectal and Sigmoid Involvement, and Adverse Morphologic Features. Mod Pathol, 2012, 25(8):1128–39., DOI 10.1038/modpathol.2012.61 Chou C-L, Tseng C-J, and Shiue Y-L. The Impact of Young Age on the Prognosis for Colorectal Cancer: a Population-Based Study in Taiwan. Jpn J Clin Oncol, 2017, 47(11):1010–8., DOI 10.1093/jjco/hyx110 Korphaisarn K, Morris V, Davis JS, Overman MJ, Fogelman DR, Kee BK, et al. Signet Ring Cell Colorectal Cancer: Genomic Insights into a Rare Subpopulation of Colorectal Adenocarcinoma. Br J Cancer, 2019, 121(6): 505–10., DOI 10.1038/s41416-019-0548-9 Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, et al. Colorectal Cancer Statistics, 2020. CA A Cancer J Clin, 2020, 70(3): 145–64., DOI 10.3322/caac.21601 Chen JS, Hsieh PS, Hung SY, Tang R, Tsai WS, Changchien CR, et al. Clinical Significance of Signet Ring Cell Rectal Carcinoma. Int J Colorectal Dis, 2004, 19(2):102–7., DOI 10.1007/s00384-003-0515-y Shirouzu K, Isomoto H, Morodomi T, Ogata Y, Akagi Y, and Kakegawa T. Primary Linitis Plastica Carcinoma of the colon and Rectum. Cancer, 1994, 74(7):1863–8., DOI 10.1002/1097-0142(19941001)74:7<1863::aidcncr2820740706> 3.0.co;2-3 Psathakis D, Schiedeck THK, Krug F, Oevermann E, Kujath P, and Bruch H-P. Ordinary Colorectal Adenocarcinoma vs. Primary Colorectal Signet-Ring Cell Carcinoma. Dis Colon Rectum, 1999, 42(12):1618–25., DOI 10.1007/ BF02236218 Imperial R, Ahmed Z, Toor OM, Erdogan C, Khaliq A, Case P, et al. Comparative Proteogenomic Analysis of Right-Sided colon Cancer, Left- Sided colon Cancer and Rectal Cancer Reveals Distinct Mutational Profiles. Mol Cancer, 2018, 17(1):177., DOI 10.1186/s12943-018-0923-9 Distler P, and Holt PR. Are Right- and Left-Sided colon Neoplasms Distinct Tumors? Dig Dis, 1997, 15(4-5):302–11., DOI 10.1159/000171605 Sung CO, Seo JW, Kim K-M, Do I-G, Kim SW, and Park C-K. Clinical Significance of Signet-Ring Cells in Colorectal Mucinous Adenocarcinoma. Mod Pathol, 2008, 21(12):1533–41., DOI 10.1038/modpathol.2008.170 Kakar S, Deng G, Smyrk TC, Cun L, Sahai V, and Kim YS. Loss of Heterozygosity, Aberrant Methylation, BRAF Mutation and KRAS Mutation in Colorectal Signet Ring Cell Carcinoma. Mod Pathol, 2012, 25(7):1040–7., DOI 10.1038/modpathol.2012.44 Thota R, Fang X, and Subbiah S. Clinicopathological Features and Survival Outcomes of Primary Signet Ring Cell and Mucinous Adenocarcinoma of colon: Retrospective Analysis of VACCR Database. J Gastrointest Oncol, 2014, 5(1):18–24., DOI 10.3978/j.issn.2078-6891.2013.051 Yang L-l., Wang M, and He P. Clinicopathological Characteristics and Survival in Colorectal Signet Ring Cell Carcinoma: a Population-Based Study. Sci Rep, 2020, 10(1):10460., DOI 10.1038/s41598-020-67388-6 Siegel RL, Miller KD, Fuchs HE, and Jemal A. Cancer Statistics, 2021. CA A Cancer J Clin, 2021, 71(1):7–33., DOI 10.3322/caac.21654 Hugen N, van de Velde CJH, de Wilt JHW, and Nagtegaal ID. Metastatic Pattern in Colorectal Cancer Is Strongly Influenced by Histological Subtype. Ann Oncol, 2014, 25(3):651–7., DOI 10.1093/annonc/mdt591 Wu Y, Zhou J, Liu T, Xu L, and Xiao Y. Multiple Polypoid Colonic Metastases from Rectal Adenocarcinoma with Signet Ring Cells Features: a Case Report. BMC Gastroenterol, 2020, 20(1):337., DOI 10.1186/s12876-020-01493-8 Nagahisa Y, Kai C, Hattori K, Sakurai R, Matsuba Y, Hashida K, et al. [A Case of Signet-Ring Cell Carcinoma of the Sigmoid Colon with Disseminated Carcinomatosis Successfully Treated with CPT-11/Panitumumab]. Gan To Kagaku Ryoho, 2015, 42(13):2477–9. Borger M, Gosens M, Jeuken J, van Kempen L, van de Velde C, van Krieken J, et al. Signet Ring Cell Differentiation in Mucinous Colorectal Carcinoma. J Pathol, 2007, 212(3):278–86., DOI 10.1002/path.2181 Zhu L, Ling C, Xu T, Zhang J, Zhang Y, Liu Y, et al. Clinicopathological Features and Survival of Signet-Ring Cell Carcinoma and Mucinous Adenocarcinoma of Right Colon, Left Colon, and Rectum. Pathol Oncol Res, 2021, 27., DOI 10.3389/pore.2021.1609800 Benedix F, Kuester D, Meyer F, and Lippert H. Einfluss des muzinosen und siegelringzelligen Subtyps auf epidemiologische, histologische und molekularbiologische Eigenschaften sowie auf die Prognose des kolorektalen Karzinoms. Zentralbl Chir, 2013, 138(4):427–33., DOI 10.1055/ s-0031-1283870 Mizushima T,NomuraM, Fujii M, Akamatsu H, MizunoH, Tominaga H, et al. Primary Colorectal Signet-Ring Cell Carcinoma: Clinicopathological Features and Postoperative Survival. Surg Today, 2010, 40(3):234–8., DOI 10.1007/ s00595-009-4057-y Gopalan V, Smith RA, Ho Y-H, and Lam AK-Y. Signet-ring Cell Carcinoma of Colorectum-Current Perspectives and Molecular Biology. Int J Colorectal Dis, 2011, 26(2):127–33., DOI 10.1007/s00384-010-1037-z Verhulst J, Ferdinande L, Demetter P, and CeelenW.Mucinous Subtype as Prognostic Factor in Colorectal Cancer: a Systematic Review and Meta- Analysis. J Clin Pathol, 2012, 65(5):381–8., DOI 10.1136/jclinpath-2011- 200340 Klaver YLB, Lemmens VEPP, Creemers GJ, Rutten HJT, Nienhuijs SW, and de Hingh IHJT. Population-based Survival of Patients with Peritoneal Carcinomatosis from Colorectal Origin in the Era of Increasing Use of Palliative Chemotherapy. Ann Oncol, 2011, 22(10):2250–6., DOI 10.1093/ annonc/mdq762 Fodde R, Smits R, and Clevers H. APC, Signal Transduction and Genetic Instability in Colorectal Cancer. Nat Rev Cancer, 2001, 1(1):55–67., DOI 10.1038/35094067 Comprehensive Molecular Characterization of Human colon and Rectal Cancer, 2012). Nature 487, 7407):330–7., DOI 10.1038/nature11252 Li Y, Li J, Wang R, Zhang L, Fu G, Wang X, et al. Frequent RNF43 Mutation Contributes to Moderate Activation of Wnt Signaling in Colorectal Signet- Ring Cell Carcinoma. Protein Cell, 2020, 11(4):292–8., DOI 10.1007/s13238- 020-00691-0 Nam J-Y, Oh BY, Hong HK, Bae JS, Kim TW, Ha SY, et al. Molecular Characterization of Colorectal Signet-Ring Cell Carcinoma Using Whole- Exome and RNA Sequencing. Translational Oncol, 2018, 11(4):836–44., DOI 10.1016/j.tranon.2018.04.007 Leggett B, and Whitehall V. Role of the Serrated Pathway in Colorectal Cancer Pathogenesis. Gastroenterology, 2010, 138(6):2088–100., DOI 10.1053/ j.gastro.2009.12.066 Wang R, Ma X, Li Y, He Y, Huang D, Cai S, et al. The Characteristics and Prognostic Effect of E-Cadherin Expression in Colorectal Signet Ring Cell Carcinoma. PLoS One, 2016, 11(8):e0160527., DOI 10.1371/ journal.pone.0160527 Nusse R, and Clevers H. Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell, 2017, 169(6):985–99., DOI 10.1016/ j.cell.2017.05.016 Nanki K,ToshimitsuK, TakanoA, FujiiM, ShimokawaM, Ohta Y, et al.Divergent Routes toward Wnt and R-Spondin Niche Independency during Human Gastric Carcinogenesis. Cell, 2018, 174(4):856–69., DOI 10.1016/j.cell.2018.07.027 Orsulic S, Huber O, Aberle H, Arnold S, and Kemler R. E-cadherin Binding Prevents Beta-Catenin Nuclear Localization and Beta-catenin/LEF-1- Mediated Transactivation. J Cel Sci, 1999, 112(Pt 8):1237–45., DOI 10.1242/ jcs.112.8.1237 Gottardi CJ, Wong E, and Gumbiner BM. E-cadherin Suppresses Cellular Transformation by Inhibiting β-Catenin Signaling in an Adhesionindependent Manner. J Cel Biol, 2001, 153(5):1049–60., DOI 10.1083/ jcb.153.5.1049 Aitchison A, Hakkaart C, Whitehead M, Khan S, Siddique S, Ahmed R, et al. CDH1 Gene Mutation in Early-Onset, Colorectal Signet-Ring Cell Carcinoma. Pathol - Res Pract, 2020, 216(5):152912., DOI 10.1016/j.prp.2020.152912 Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ, Tu D, Tebbutt NC, et al. K-rasMutations and Benefit from Cetuximab in Advanced Colorectal Cancer. N Engl J Med, 2008, 359(17):1757–65., DOI 10.1056/NEJMoa0804385 Pai RK, Jayachandran P, Koong AC, Chang DT, Kwok S, Ma L, et al. BRAFmutated, Microsatellite-Stable Adenocarcinoma of the Proximal Colon. Am J Surg Pathol, 2012, 36(5):744–52., DOI 10.1097/PAS.0b013e31824430d7 Li WQ, Kawakami K, Ruszkiewicz A, Bennett G, Moore J, and Iacopetta B. BRAF Mutations Are Associated with Distinctive Clinical, Pathological and Molecular Features of Colorectal Cancer Independently of Microsatellite Instability Status. Mol Cancer, 2006, 5:2., DOI 10.1186/1476-4598-5-2 Nagasaka T, Sasamoto H, Notohara K, Cullings HM, Takeda M, Kimura K, et al. Colorectal Cancer with Mutation in BRAF, KRAS, and Wild-type with Respect to Both Oncogenes Showing Different Patterns of DNA Methylation. Jco, 2004, 22(22):4584–94., DOI 10.1200/jco.2004.02.154 Fang M, Ou J, Hutchinson L, and Green MR. The BRAF Oncoprotein Functions through the Transcriptional Repressor MAFG to Mediate the CpG Island Methylator Phenotype. Mol Cell, 2014, 55(6):904–15., DOI 10.1016/j.molcel.2014.08.010 Wei Q, Wang X, Gao J, Li J, Li J, Qi C, et al. Clinicopathologic and Molecular Features of Colorectal Adenocarcinoma with Signet-Ring Cell Component. PLoS One, 2016, 11(6):e0156659., DOI 10.1371/journal.pone.0156659 Kim H, Kim B-H, Lee D, and Shin E. Genomic Alterations in Signet Ring and Mucinous Patterned Colorectal Carcinoma. Pathol - Res Pract, 2019, 215(10): 152566., DOI 10.1016/j.prp.2019.152566 Ogino S, Brahmandam M, Cantor M, Namgyal C, Kawasaki T, Kirkner G, et al. Distinct Molecular Features of Colorectal Carcinoma with Signet Ring Cell Component and Colorectal Carcinoma with Mucinous Component. Mod Pathol, 2006, 19(1):59–68., DOI 10.1038/modpathol.3800482 Inamura K, Yamauchi M, Nishihara R, Kim SA, Mima K, Sukawa Y, et al. Prognostic Significance and Molecular Features of Signet-Ring Cell and Mucinous Components in Colorectal Carcinoma. Ann Surg Oncol, 2015, 22(4):1226–35., DOI 10.1245/s10434-014-4159-7 Alvi MA, Loughrey MB, Dunne P,McQuaid S, Turkington R, FuchsM-A, et al. Molecular Profiling of Signet Ring Cell Colorectal Cancer Provides a strong Rationale for Genomic Targeted and Immune Checkpoint Inhibitor Therapies. Br J Cancer, 2017, 117(2):203–9., DOI 10.1038/bjc.2017.168 Mei ZB, Duan CY, Li CB, Cui L, and Ogino S. Prognostic Role of Tumor PIK3CA Mutation in Colorectal Cancer: a Systematic Review and Meta- Analysis. Ann Oncol, 2016, 27(10):1836–48., DOI 10.1093/annonc/mdw264 Liao X, Lochhead P, Nishihara R, Morikawa T, Kuchiba A, Yamauchi M, et al. Aspirin Use, TumorPIK3CAMutation, and Colorectal-Cancer Survival.NEngl J Med, 2012, 367(17):1596–606., DOI 10.1056/NEJMoa1207756 Kim A, Lee J-E, Lee S-S, Kim C, Lee S-J, Jang W-S, et al. Coexistent Mutations ofKRASandPIK3CAaffect the Efficacy of NVP-Bez235, a Dual PI3K/MTOR Inhibitor, in Regulating the PI3K/MTOR Pathway in Colorectal Cancer. Int J Cancer, 2013, 133(4):984–96., DOI 10.1002/ijc.28073 Wasserman I, Lee LH, Ogino S, Marco MR, Wu C, Chen X, et al. SMAD4 Loss in Colorectal Cancer Patients Correlates with Recurrence, Loss of Immune Infiltrate, and Chemoresistance. Clin Cancer Res, 2019, 25(6):1948–56., DOI 10.1158/1078-0432.CCR-18-1726 Fleming NI, Jorissen RN, Mouradov D, Christie M, Sakthianandeswaren A, Palmieri M, et al. SMAD2, SMAD3 and SMAD4 Mutations in Colorectal Cancer. Cancer Res, 2013, 73(2):725–35., DOI 10.1158/0008-5472.Can-12- 2706 Lamouille S, Xu J, and Derynck R. Molecular Mechanisms of Epithelial- Mesenchymal Transition. Nat Rev Mol Cell Biol, 2014, 15(3):178–96., DOI 10.1038/nrm3758 Ziemke M, Patil T, Nolan K, Tippimanchai D, and Malkoski SP. Reduced Smad4 Expression and DNA Topoisomerase Inhibitor Chemosensitivity in Non-small Cell Lung Cancer. Lung Cancer, 2017, 109:28–35., DOI 10.1016/ j.lungcan.2017.04.017 Siraj AK, Pratheeshkumar P, Divya SP, Parvathareddy SK, Bu R, Masoodi T, et al. Tgfβ-Induced SMAD4-dependent Apoptosis Proceeded by EMT in CRC. Mol Cancer Ther, 2019, 18(7):1312–22., DOI 10.1158/1535-7163.mct-18-1378 Marzouk O, and Schofield J. Review of Histopathological and Molecular Prognostic Features in Colorectal Cancer. Cancers, 2011, 3(2):2767–810., DOI 10.3390/cancers3022767 Boland CR, and Goel A. Microsatellite Instability in Colorectal Cancer. Gastroenterology, 2010, 138(6):2073–87., DOI 10.1053/ j.gastro.2009.12.064e2073 Sinicrope FA. Lynch Syndrome-Associated Colorectal Cancer. N Engl J Med, 2018, 379(8):764–73., DOI 10.1056/NEJMcp1714533 Ogino S, Nosho K, Kirkner GJ, Kawasaki T,Meyerhardt JA, LodaM, et al. CpG Island Methylator Phenotype, Microsatellite Instability, BRAF Mutation and Clinical Outcome in colon Cancer. Gut, 2009, 58(1):90–6., DOI 10.1136/ gut.2008.155473 Kakar S, and Smyrk TC. Signet Ring Cell Carcinoma of the Colorectum: Correlations between Microsatellite Instability, Clinicopathologic Features and Survival. Mod Pathol, 2005, 18(2):244–9., DOI 10.1038/modpathol.3800298 Hartman DJ, Nikiforova MN, Chang DT, Chu E, Bahary N, Brand RE, et al. Signet Ring Cell Colorectal Carcinoma. Am J Surg Pathol, 2013, 37(7):969–77., DOI 10.1097/PAS.0b013e3182851e2b Yalcin S, and Onguru O. BRAF Mutation in Colorectal Carcinomas with Signet Ring Cell Component. Cancer Biol Med, 2017, 14(3):287–92., DOI 10.20892/ j.issn.2095-3941.2017.0053 Tajiri K, Sudou T, Fujita F, Hisaka T, Kinugasa T, and Akagi Y. Clinicopathological and Corresponding Genetic Features of Colorectal Signet Ring Cell Carcinoma. Anticancer Res, 2017, 37(7):3817–23., DOI 10.21873/anticanres.11760 Bruni D, Angell HK, and Galon J. The Immune Contexture and Immunoscore in Cancer Prognosis and Therapeutic Efficacy. Nat Rev Cancer, 2020, 20(11): 662–80., DOI 10.1038/s41568-020-0285-7 Galon J, and Bruni D. Tumor Immunology and Tumor Evolution: Intertwined Histories. Immunity, 2020, 52(1):55–81., DOI 10.1016/j.immuni.2019.12.018 Kishore C, and Bhadra P. Current Advancements and Future Perspectives of Immunotherapy in Colorectal Cancer Research. Eur J Pharmacol, 2021, 893: 173819., DOI 10.1016/j.ejphar.2020.173819 Ishihara S,Watanabe T, Akahane T, Shimada R, Horiuchi A, Shibuya H, et al. Tumor Location Is a Prognostic Factor in Poorly Differentiated Adenocarcinoma, Mucinous Adenocarcinoma, and Signet-Ring Cell Carcinoma of the colon. Int J Colorectal Dis, 2012, 27(3):371–9., DOI 10.1007/s00384-011-1343-0 Lee W-S, Chun H-K, Lee WY, Yun SH, Cho YB, Yun H-R, et al. Treatment Outcomes in Patients with Signet Ring Cell Carcinoma of the Colorectum. Am J Surg, 2007, 194(3):294–8., DOI 10.1016/j.amjsurg.2006.12.041 Zhao Z, Yan N, Pan S, Wang D-w., and Li Z-w. The Value of Adjuvant Chemotherapy in Stage II/III Colorectal Signet Ring Cell Carcinoma. Sci Rep, 2020, 10(1):14126., DOI 10.1038/s41598-020-70985-0 Prabhu A, Brandl A, Wakama S, Sako S, Ishibashi H, Mizumoto A, et al. Retrospective Analysis of Patients with Signet Ring Subtype of Colorectal Cancer with Peritoneal Metastasis Treated with CRS & HIPEC. Cancers, 2020, 12(9):2536., DOI 10.3390/cancers12092536 Winer J, Zenati M, Ramalingam L, Jones H, Zureikat A, Holtzman M, et al. Impact of Aggressive Histology and Location of Primary Tumor on the Efficacy of Surgical Therapy for Peritoneal Carcinomatosis of Colorectal Origin. Ann Surg Oncol, 2013, 21(5):1456–62., DOI 10.1245/s10434-013- 3328-4 Narasimhan V, Tan S, Kong J, Pham T, Michael M, Ramsay R, et al. Prognostic Factors Influencing Survival in Patients Undergoing Cytoreductive Surgery with Hyperthermic Intraperitoneal Chemotherapy for Isolated Colorectal Peritoneal Metastases: a Systematic Review and Meta-analysis. Colorectal Dis, 2020, 22(11):1482–95., DOI 10.1111/codi.15003 Zhou J, Wang C, Lin G, Xiao Y, Jia W, Xiao G, et al. Serial Circulating Tumor DNA in Predicting and Monitoring the Effect of Neoadjuvant Chemoradiotherapy in Patients with Rectal Cancer: A Prospective Multicenter Study. Clin Cancer Res, 2021, 27(1):301–10., DOI 10.1158/1078- 0432.CCR-20-2299 Zhang J, Zheng J, Yang Y, Lu J, Gao J, Lu T, et al. Molecular Spectrum of KRAS, NRAS, BRAF and PIK3CA Mutations in Chinese Colorectal Cancer Patients: Analysis of 1,110 Cases. Sci Rep, 2015, 5:18678., DOI 10.1038/ srep18678 Chew M-H, Yeo S-AE, Ng Z-P, Lim K-H, Koh P-K, Ng K-H, et al. Critical Analysis of Mucin and Signet Ring Cell as Prognostic Factors in an Asian Population of 2,764 Sporadic Colorectal Cancers. Int J Colorectal Dis, 2010, 25(10):1221–9., DOI 10.1007/s00384-010-1033-3 Liang Z, Yan D, Li G, and Cheng H. Clinical Analysis of Primary Colorectal Signet-Ring Cell Carcinoma. Clin Colorectal Cancer, 2018, 17(1):e39–e44., DOI 10.1016/j.clcc.2017.06.010 Kawabata Y, Tomita N, Monden T, Ohue M, Ohnishi T, Sasaki M, et al. Molecular Characteristics of Poorly Differentiated Adenocarcinoma and Signet-Ring-Cell Carcinoma of Colorectum. Int J Cancer, 1999, 84(1):33–8., DOI 10.1002/(sici)1097-0215(19990219)84:1<33::aid-ijc7>3.0.co;2-z Chen J, Zhou L, Gao J, Lu T, Wang J, Wu H, et al. Clinicopathological Characteristics and Mutation Spectrum of Colorectal Adenocarcinoma with Mucinous Component in a Chinese Cohort: Comparison with Classical Adenocarcinoma. Front Oncol, 2020, 10:917., DOI 10.3389/fonc.2020.00917 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609859 EP 1609871 DI 10.3389/pore.2021.1609859 PG 13 ER PT J AU Oh, ES Oh, YM An, YJ Lee, HJ Sohn, ST Bae, MJ Choi, MG Kim, KM AF Oh, Eun Sung Oh, Yun Mi An, Yeong Ji Lee, Ho Jun Sohn, Sung Tae Bae, Moon Jae Choi, Min-Gew Kim, Kyoung-Mee TI Prognostic Value of Highly Expressed Type VII Collagen (COL7A1) in Patients With Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; prognosis; gastric cancer; tumor microenvironment; type VII collagen (COL7A1); distant metastasis ID biomarker; prognosis; gastric cancer; tumor microenvironment; type VII collagen (COL7A1); distant metastasis AB Collagen is a major component in the tumor microenvironment. This study reveals a novel biomarker candidate, type VII collagen (COL7A1), in patients with gastric cancer. To identify genes differentially expressed in gastric cancer tissue, we analyzed cancerous (n = 20) and noncancerous tissues (n = 13) using a DNA microarray. To perform immunohistochemistry and validate the upregulation of COL7A1 expression, we collected 200 more gastric cancer tissues and 100 normal gastric tissues from 200 randomly selected patients who underwent gastrectomy for gastric cancer between January 2010 and December 2013. The correlations between COL7A1 expression and clinicopathological parameters and patients’ overall survival (OS) were analyzed. In the microarray, COL7A1 was upregulated in gastric cancer tissue compared with normal tissue. In the immunohistochemistry study, COL7A1 was more highly expressed in cancer tissue than in normal tissue (p = 0.001). Patients with intracellular COL7A1 expression had significantly poorer five-year OS than those with only extracellular expression (41.5 versus 69.7%, p = 0.001), and the site of expression was an independent prognostic factor of OS (hazard ratio 2.00, 95% CI 1.26–3.16, p = 0.003). Also, we found a significant association between the COL7A1 immunohistochemistry score and distant metastasis (high versus low, odds ratio 4.45, 95% CI 1.40–14.16, p = 0.011). The site and total immunohistochemistry score of COL7A1 expression in gastric cancer showed prognostic significance for OS and distant metastasis, respectively. COL7A1 could be a novel biomarker with diagnostic and therapeutic value. C1 [Oh, Eun Sung] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of SurgerySeoul, South Korea. [Oh, Yun Mi] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of SurgerySeoul, South Korea. [An, Yeong Ji] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of SurgerySeoul, South Korea. [Lee, Ho Jun] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of SurgerySeoul, South Korea. [Sohn, Sung Tae] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of SurgerySeoul, South Korea. [Bae, Moon Jae] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of SurgerySeoul, South Korea. [Choi, Min-Gew] Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of SurgerySeoul, South Korea. [Kim, Kyoung-Mee] Sungkyunkwan University School of Medicine, Samsung Medical Center, Department of PathologySeoul, South Korea. RP Choi, MG (reprint author), Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Department of Surgery, Seoul, South Korea. EM mingew.choi@samsung.com CR Jun JK, Choi KS, Lee H-Y, Suh M, Park B, Song SH, et al. Effectiveness of the Korean National Cancer Screening Programin Reducing Gastric Cancer Mortality. Gastroenterology, 2017, 152:1319–28., DOI 10.1053/j.gastro.2017.01.029 Noh SH, Park SR, Yang H-K, Chung HC, Chung I-J, Kim S-W, et al. Adjuvant Capecitabine Plus Oxaliplatin for Gastric Cancer after D2 Gastrectomy, CLASSIC): 5-year Follow-Up of an Open-Label, Randomised Phase 3 Trial. Lancet Oncol, 2014, 15:1389–96., DOI 10.1016/s1470-2045(14)70473-5 Sakuramoto S, Sasako M, Yamaguchi T, Kinoshita T, Fujii M, Nashimoto A, et al. Adjuvant Chemotherapy for Gastric Cancer with S-1, an Oral Fluoropyrimidine. N Engl J Med, 2007, 357:1810–20., DOI 10.1056/nejmoa072252 Jung KW, Won YJ, Kong HJ, and Lee ES, Community of Population-Based Regional Cancer R. Cancer Statistics in Korea: Incidence, Mortality, Survival, and Prevalence in 2015. Cancer Res Treat, 2018, 50:303–16., DOI 10.4143/crt.2018.143 Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer Incidence and Mortality Worldwide: Sources, Methods and Major Patterns in GLOBOCAN 2012. Int J Cancer, 2015, 136:E359–E386., DOI 10.1002/ijc.29210 Sawayama H, Ishimoto T, and Baba H. Microenvironment in the Pathogenesis of Gastric Cancer Metastasis. Jcmt, 2018, 4:10., DOI 10.20517/2394-4722.2017.79 Gelse K, Poschl E, and Aigner T. Collagens-structure, Function, and Biosynthesis. Adv Drug Deliv Rev, 2003, 55:1531–46., DOI 10.1016/j.addr.2003.08.002 Li J, Ding Y, and Li A. Identification of COL1A1 and COL1A2 as Candidate Prognostic Factors in Gastric Cancer. World J Surg Oncol, 2016, 14:297., DOI 10.1186/s12957-016-1056-5 Zhou Z-H, Ji C-D, Xiao H-L, Zhao H-B, Cui Y-H, and Bian X-W. Reorganized Collagen in the Tumor Microenvironment of Gastric Cancer and its Association with Prognosis. J Cancer, 2017, 8:1466–76., DOI 10.7150/jca.18466 Christiano AM, Anhalt G, Gibbons S, Bauer EA, and Uitto J. Premature Termination Codons in the Type VII Collagen Gene, COL7A1, Underlie Severe, Mutilating Recessive Dystrophic Epidermolysis Bullosa. Genomics, 1994, 21:160–8., DOI 10.1006/geno.1994.1238 MartinsVL,Vyas JJ, ChenM,PurdieK,MeinCA, SouthAP, et al. IncreasedInvasive Behaviour in Cutaneous Squamous Cell Carcinoma with Loss of Basement- Membrane Type VII Collagen. J Cel Sci, 2009, 122:1788–99., DOI 10.1242/jcs.042895 Kita Y, Mimori K, Tanaka F, Matsumoto T,Haraguchi N, Ishikawa K, et al. Clinical Significance of LAMB3 and COL7A1 mRNA in Esophageal Squamous Cell Carcinoma. Eur J Surg Oncol, Ejso,, 2009, 35:52–8., DOI 10.1016/j.ejso.2008.01.025 Huang DW, Sherman BT, and Lempicki RA. Systematic and Integrative Analysis of Large Gene Lists Using DAVID Bioinformatics Resources. Nat Protoc, 2009, 4:44–57., DOI 10.1038/nprot.2008.211 Rong L, Huang W, Tian S, Chi X, Zhao P, and Liu F. COL1A2 Is a Novel Biomarker to Improve Clinical Prediction in Human Gastric Cancer: Integrating Bioinformatics and Meta-Analysis. Pathol Oncol Res, 2018, 24: 129–34., DOI 10.1007/s12253-017-0223-5 Contal C, and O’Quigley J. An Application of Changepoint Methods in Studying the Effect of Age on Survival in Breast Cancer. Comput Stat Data Anal, 1999, 30:253–70., DOI 10.1016/s0167-9473(98)00096-6 Hippo Y, Taniguchi H, Tsutsumi S,Machida N, Chong JM, FukayamaM, et al. Global Gene Expression Analysis of Gastric Cancer by Oligonucleotide Microarrays. Cancer Res, 2002, 62:233–40. Lastraioli E, Romoli MR, and Arcangeli A. Immunohistochemical Biomarkers in Gastric Cancer Research and Management. Int J Surg Oncol, 2012, 2012: 868645., DOI 10.1155/2012/868645 Fang M, Yuan J, Peng C, and Li Y. Collagen as a Double-Edged Sword in Tumor Progression. Tumor Biol, 2014, 35:2871–82., DOI 10.1007/s13277-013- 1511-7 Matsui H, Kubochi K, Okazaki I, Yoshino K, Ishibiki K, and Kitajima M. Collagen Biosynthesis in Gastric Cancer: Immunohistochemical Analysis of Prolyl 4-hydroxylase. J Surg Oncol, 1999, 70:239–46., DOI 10.1002/(sici)1096- 9098(199904)70:4<239::aid-jso8>3.0.co;2-m Naito Y, Kino I, Horiuchi K, and Fujimoto D. Promotion of Collagen Production by Human Fibroblasts with Gastric Cancer Cellsin Vitro. Virchows Archiv B Cel Pathol, 1984, 46:145–54., DOI 10.1007/bf02890304 Ohno S, Tachibana M, Fujii T, Ueda S, Kubota H, and Nagasue N. Role of Stromal Collagen in Immunomodulation and Prognosis of Advanced Gastric Carcinoma. Int J Cancer, 2002, 97:770–4., DOI 10.1002/ijc.10144 NaitoY, SakamotoN, OueN,YashiroM,SentaniK,YanagiharaK, et al.MicroRNA -143 Regulates Collagen Type III Expression in Stromal Fibroblasts of Scirrhous Type Gastric Cancer. Cancer Sci, 2014, 105:228–35., DOI 10.1111/cas.12329 Xie X, Liu X, Zhang Q, and Yu J. Overexpression of Collagen VI α3 in Gastric Cancer. Oncol Lett, 2014, 7:1537–43., DOI 10.3892/ol.2014.1910 Ma Y, Zhu J, Chen S, Li T, Ma J, Guo S, et al. Activated Gastric Cancer- Associated Fibroblasts Contribute to the Malignant Phenotype and 5-FU Resistance via Paracrine Action in Gastric Cancer. Cancer Cel Int, 2018, 18:104., DOI 10.1186/s12935-018-0599-7 Ham IH, Lee D, and Hur H. Role of Cancer-Associated Fibroblast in Gastric Cancer Progression and Resistance to Treatments. J Oncol, 2019, 2019: 6270784., DOI 10.1155/2019/6270784 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609860 EP 1609870 DI 10.3389/pore.2021.1609860 PG 11 ER PT J AU Kopinska, JA Koclega, A Wozniczka, K Helbig, G AF Kopinska, J Anna Koclega, Anna Wozniczka, Krzysztof Helbig, Grzegorz TI Allogeneic Stem Cell Transplantation for Relapsed and Refractory Hodgkin Lymphoma: Real World Experience of a Single Center SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Hodgkin lymphoma; allogeneic stem cell transplantation; autologous stem cell transplantation; chemosensitivity; outcome ID Hodgkin lymphoma; allogeneic stem cell transplantation; autologous stem cell transplantation; chemosensitivity; outcome AB Introduction: Refractory and relapsed Hodgkin lymphoma (R/R HL) is associated with poor prognosis, and allogeneic stem cell transplantation (allo-SCT) remains the only potentially curative approach. Aim: The aim of the study was to evaluate the feasibility of allotransplantation in R/R HL setting. Material: Overall, 24 patients (17 men and 7 women) at a median age of 27 years (range 18–44) underwent allo-SCT between 2002 and 2020. Results: Nineteen patients received prior autologous stem cell transplantation (ASCT1) whereas eight patients received second ASCT (ASCT2) after failure of ASCT1. Six patients received only brentuximab vedotin (BV; n = 4) or BV followed by checkpoint inhibitors (CPI; n = 2) before entering allo-SCT. Median time from ASCT1 to allo-SCT was 17.1 months. Fifteen patients received grafts from unrelated donors. Peripheral blood was a source of stem cells for 16 patients. Reduced-intensity conditioning was used for all patients. Disease status at transplant entry was as follows: complete remission (CR; n = 4), partial response (PR; n = 10), and stable disease (SD; n = 10). Acute and chronic graft-versus-host disease (GVHD) developed in 13 (54%) and 4 (16%) patients, respectively. Median follow-up for the entire cohort was 13.3 months. At the last follow-up, 17 (71%) patients died. The main causes of death were disease progression (n = 10), infectious complications (n = 6), and steroid-resistant GVHD (n = 1). Non-relapse mortality at 12 months was 25%. At the last follow-up, seven patients were alive; six patients were in CR, and one had PR. The 2-year overall survival (OS) was 40%. Conclusion: Chemosensitive disease at transplant was associated with better outcome. Allo-SCT allows for long-term survival in refractory and relapsed HL. C1 [Kopinska, J Anna] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow TransplantationKatowice, Poland. [Koclega, Anna] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow TransplantationKatowice, Poland. [Wozniczka, Krzysztof] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow TransplantationKatowice, Poland. [Helbig, Grzegorz] Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow TransplantationKatowice, Poland. RP Helbig, G (reprint author), Medical University of Silesia, School of Medicine in Katowice, Department of Hematology and Bone Marrow Transplantation, Katowice, Poland. EM ghelbig@o2.pl CR Duarte RF, Labopin M, Labopin M, Bader P, Basak GW, Bonini C, et al. Indications for Haematopoietic Stem Cell Transplantation for Haematological Diseases, Solid Tumours and Immune Disorders: Current Practice in Europe, 2019. Bone Marrow Transpl, 2019, 54:1525–52., DOI 10.1038/s41409-019- 0516-2 Shah GL, and Moskowitz CH. Transplant Strategies in Relapsed/refractory Hodgkin Lymphoma. Blood, 2018, 131:1689–97., DOI 10.1182/blood-2017-09- 772673 Castagna L, Carlo-Stella C, Mazza R, and Santoro A. Current Role of Autologous and Allogeneic Stem Cell Transplantation for Relapsed and Refractory Hodgkin Lymphoma. Mediterr J Hematol Infect Dis, 2015, 7: e2015015., DOI 10.4084/MJHID.2015.015 Moskowitz CH. Should All Patients with HL Who Relapse after ASCT Be Considered for Allogeneic SCT? A Consult, Yes; a Transplant, Not Necessarily. Blood Adv, 2018, 2:821–4., DOI 10.1182/bloodadvances.2017011130 Anderlini P, Saliba R, Acholonu S, Giralt SA, Andersson B, Ueno NT, et al. Fludarabine-melphalan as a Preparative Regimen for Reduced-Intensity Conditioning Allogeneic Stem Cell Transplantation in Relapsed and Refractory Hodgkin’s Lymphoma: the Updated M.D. Anderson Cancer Center Experience. Haematologica, 2008, 93:257–64., DOI 10.3324/ haematol.11828 Sureda A, Robinson S, Canals C, Carella AM, Boogaerts MA, Caballero D, et al. Reduced-Intensity Conditioning Compared with Conventional Allogeneic Stem-Cell Transplantation in Relapsed or Refractory Hodgkin’s Lymphoma: An Analysis from the Lymphoma Working Party of the European Group for Blood and Marrow Transplantation. Jco, 2008, 26: 455–62., DOI 10.1200/JCO.2007.13.2415 Lister TA, Crowther D, Sutcliffe SB, Glatstein E, Canellos GP, Young RC, et al. Report of a Committee Convened to Discuss the Evaluation and Staging of Patients with Hodgkin’s Disease: Cotswolds Meeting. Jco, 1989, 7:1630–6., DOI 10.1200/JCO.1989.7.11.1630 Cheson BD, Pfistner B, Juweid ME, Gascoyne RD, Specht L, Horning SJ, et al. Revised Response Criteria for Malignant Lymphoma. Jco, 2007, 25:579–86., DOI 10.1200/JCO.2006.09.2403 Chen R, Palmer JM, Tsai N-C, Thomas SH, Siddiqi T, Popplewell L, et al. Brentuximab Vedotin Is Associated with Improved Progression-free Survival after Allogeneic Transplantation for Hodgkin Lymphoma. Biol Blood Marrow Transplant, 2014, 20:1864–8., DOI 10.1016/j.bbmt.2014.06.037 Martinez C, Carpio C, Heras I, Rios-Herranz E, Buch J, Gutierrez A, et al. Potential Survival Benefit for Patients Receiving Allogeneic Hematopoietic Stem Cell Transplantation after Nivolumab Therapy for Relapse/Refractory Hodgkin Lymphoma: Real-Life Experience in Spain. Biol Blood Marrow Transplant, 2020, 26:1534–42., DOI 10.1016/j.bbmt.2020.02.003 Chen R, Gopal AK, Smith SE, Ansell SM, Rosenblatt JD, Savage KJ, et al. Five-year Survival and Durability Results of Brentuximab Vedotin in Patients with Relapsed or Refractory Hodgkin Lymphoma. Blood, 2016, 128:1562–6., DOI 10.1016/j.bbmt.2020.02.00310.1182/blood-2016-02- 699850 Merryman RW, Kim HT, Zinzani PL, Carlo-Stella C, Ansell SM, Perales M-A, et al. Safety and Efficacy of Allogeneic Hematopoietic Stem Cell Transplant after PD-1 Blockade in Relapsed/refractory Lymphoma. Blood, 2017, 129: 1380–8., DOI 10.1182/blood-2016-09-738385 Schoch LK, Borrello I, Fuchs EJ, Bolanos-Meade J, Huo JS, Gojo I, et al. Checkpoint Inhibitor Therapy and Graft versus Host Disease in Allogeneic Bone Marrow Transplant Recipients of Haploidentical and Matched Products with post-transplant Cyclophosphamide. Blood, 2016, 128:4571., DOI 10.1182/ blood.v128.22.4571.4571 Merryman RW, Castagna L, Giordano L, Ho VT, Corradini P, Guidetti A, et al. Allogeneic Transplantation after PD-1 Blocade for Classic Hodgkin Lymphoma. Leukemia, 2021, 5:1648–1659., DOI 10.1038/s41375-021-01193-6 Rivas MM, Berro M, Berro M, Prates MV, Yantorno S, Fiad L, et al. Allogeneic Stem Cell Transplantation Improves Survival in Relapsed Hodgkin Lymphoma Patients Achieving Complete Remission after Salvage Treatment. Bone Marrow Transpl, 2020, 55:117–25., DOI 10.1038/s41409-019-0640-z Gajewski JL, Phillips GL, Sobocinski KA, Armitage JO, Gale RP, Champlin RE, et al. Bone Marrow Transplants from HLA-Identical Siblings in Advanced Hodgkin’s Disease. Jco, 1996, 14:572–8., DOI 10.1200/JCO.1996.14.2.572 Genadieva-Stavrik S, Boumendil A, Dreger P, Peggs K, Briones J, Corradini P, et al. Myeloablative versus Reduced Intensity Allogeneic Stem Cell Transplantation for Relapsed/refractory Hodgkin’s Lymphoma in Recent Years: a Retrospective Analysis of the Lymphoma Working Party of the European Group for Blood and Marrow Transplantation. Ann Oncol, 2016, 27:2251–7., DOI 10.1093/ANNONC/MDW421 Peggs KS, Sureda A, Qian W, Caballero D, Hunter A, Urbano-Ispizua A, et al. Reduced-intensity Conditioning for Allogeneic Haematopoietic Stem Cell Transplantation in Relapsed and Refractory Hodgkin Lymphoma: Impact of Alemtuzumab and Donor Lymphocyte Infusions on Long-Term Outcomes. Br J Haematol, 2007, 139:70–80., DOI 10.1111/j.1365-2141.2007.06759.x Burroughs LM,O’Donnell PV, Sandmaier BM, Storer BE, Luznik L, SymonsHJ, et al. Comparison of Outcomes of HLA-Matched Related, Unrelated, or HLAHaploidentical Related Hematopoietic Cell Transplantation Following Nonmyeloablative Conditioning for Relapsed or Refractory Hodgkin Lymphoma. Biol Blood Marrow Transplant, 2008, 14:1279–87., DOI 10.1016/j.bbmt.2008.08.014 Martinez C, Gayoso J, Canals C, Finel H, Peggs K, Dominietto A, et al. Posttransplantation Cyclophosphamide-Based Haploidentical Transplantation as Alternative to Matched Sibling or Unrelated Donor Transplantation for Hodgkin Lymphoma: a Registry Study of the Lymphoma Working Party of the European Society for Blood and Marrow Transplantation. Jco, 2017, 35: 3425–32., DOI 10.1200/JCO.2017.72.6869 Ahmed S, Kanakry JA, Ahn KW, Litovich C, Abdel-Azim H, Aljurf M, et al. Lower Graft-Versus-Host Disease and Relapse Risk in Post-Transplant Cyclophosphamide-Based Haploidentical versus Matched Sibling Donor Reduced-Intensity Conditioning Transplant for Hodgkin Lymphoma. Biol Blood Marrow Transplant, 2019, 25:1859–68., DOI 10.1016/j.bbmt.2019.05.025 Castagna L, Busca A, Bramanti S, Raiola Anna M, Malagola M, Ciceri F, et al. Haploidentical Related Donor Compared to HLA-Identical Donor Transplantation for Chemosensitive Hodgkin Lymphoma Patients. BMC Cancer, 2020, 20:1140., DOI 10.1186/s12885-020-07602-w Rashidi A, Ebadi M, and Cashen AF. Allogeneic Hematopoietic Stem Cell Transplantation in Hodgkin Lymphoma: a Systematic Review and Meta- Analysis. Bone Marrow Transpl, 2016, 51:521–8., DOI 10.1038/ bmt.2015.332 Sureda A, Genadieva Stavrik S, Boumendil A, Finel H, Khvedelidze I, Dietricht S, et al. Changes in Patients Population and Characteristics of Hematopoietic Stem Cell Transplantation for Relapsed/refractory Hodgkin Lymphoma: an Analysis of the LymphomaWorking Party of the EBMT. Bone Marrow Transpl, 2020, 55:2170–9., DOI 10.1038/s41409-020-0929-y NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609867 EP 1609873 DI 10.3389/pore.2021.1609867 PG 7 ER PT J AU Liu, Sh Cai, Y Sheng, J Zhang, X AF Liu, Shui Cai, Yan Sheng, Jiyao Zhang, Xuewen TI Screening and Validation ofIndependent Predictors of PoorSurvival in Pancreatic Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; pancreatic cancer; the cancer genome atlas; tetraspanin-1; erb-b2 receptor tyrosine kinase 3; gene expression omnibus ID prognosis; pancreatic cancer; the cancer genome atlas; tetraspanin-1; erb-b2 receptor tyrosine kinase 3; gene expression omnibus AB Pancreatic cancer is a digestive system malignant tumor with high mortality and poor prognosis, but the mechanisms of progression remain unclear in pancreatic cancer. It’s necessary to identify the hub genes in pancreatic cancer and explore the novel potential predictors in the prognosis of pancreatic cancer. We downloaded two mRNA expression profiles from Gene Expression Omnibus and The Cancer Genome Atlas Pancreatic Cancer (TCGA-PAAD) datasets to screen the commonly differentially expressed genes in pancreatic cancer by limma package in R. Subsequently, measurement of the functional similarity among the 38 DEGs in common was performed to identify the hub genes using GOSemSim package. Then, survival analysis and Cox regression were applied to explore prognosis-related hub genes using the survival package. Statistics analysis by two-tailed Student’s t-test or one-way based on TCGA-PAAD datasets and qPCR detection in clinical samples were performed to explore the correlations between expression of hub genes in pancreatic cancer tissues and clinical parameters. Based on integrated analysis of TCGA and GEO datasets, we screened 38 DEGs in common, which were all up-regulated. The functional similarity results showed that 10 DEGs including TSPAN1, MSLN, C1orf116, PKP3, CEACAM6, BAIAP2L1, PPL, RAB25, ERBB3, and AP1M2 in the DEGs in common, which had the higher average functional similarity, were considered as the hub genes. Survival analysis results and Cox regression analysis showed that TSPAN1, CEACAM6, as well as ERBB3 were all associated with poor overall survival of PC. qPCR results showed that the expression levels of TSPAN1 and ERBB3 were significantly upregulated in the PC tissues. The statistical analysis results revealed that TSPAN1 expression correlated significantly with histologic grade, T stage, clinical stage, and vital status by two-tailed Student’s t-test or one-way ANOVA; ERBB3 expression correlated significantly with T stage, clinical stage, and vital status by two-tailed Student’s t-test or one-way ANOVA. We found that TSPAN1 and ERBB3 could be independent predictors of poor survival in pancreatic cancer. C1 [Liu, Shui] Jilin University, The Second Hospital of Jilin University, Department of Hepatobiliary and Pancreatic SurgeryChangchun, China. [Cai, Yan] Jilin University, Hospital of StomatologyChangchun, China. [Sheng, Jiyao] Jilin University, The Second Hospital of Jilin University, Department of Hepatobiliary and Pancreatic SurgeryChangchun, China. [Zhang, Xuewen] Jilin University, The Second Hospital of Jilin University, Department of Hepatobiliary and Pancreatic SurgeryChangchun, China. RP Sheng, J (reprint author), Jilin University, The Second Hospital of Jilin University, Department of Hepatobiliary and Pancreatic Surgery, Changchun, China. EM shengjiyao@jlu.edu.cn CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: a Cancer J clinicians, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Institute NC. Surveillance, Epidemiology, and End Results, SEER, 18 Registries, 2020). Available at: https://cancerstatisticscenter.cancer.org/#!/, Accessed June 30, 2021). Cui M, Yao X, Lin Y, Zhang D, Cui R, and Zhang X. Interactive Functions of microRNAs in the miR-23a-27a-24-2 Cluster and the Potential for Targeted Therapy in Cancer. J Cel Physiol, 2020, 235(1):6–16., DOI 10.1002/jcp.28958 Hou J, Li X, and Xie KP. Coupled Liquid Biopsy and Bioinformatics for Pancreatic Cancer Early Detection and Precision Prognostication. Mol Cancer, 2021, 20(1):34., DOI 10.1186/s12943-021-01309-7 Pei H, Li L, Fridley BL, Jenkins GD, Kalari KR, Lingle W, et al. FKBP51 Affects Cancer Cell Response to Chemotherapy by Negatively Regulating Akt. Cancer cell, 2009, 16(3):259–66., DOI 10.1016/j.ccr.2009.07.016 Donahue TR, Tran LM, Hill R, Li Y, Kovochich A, Calvopina JH, et al. Integrative Survival-Based Molecular Profiling of Human Pancreatic Cancer. Clin Cancer Res, 2012, 18(5):1352–63., DOI 10.1158/1078-0432.ccr-11-1539 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res, 2015, 43(7):e47., DOI 10.1093/nar/gkv007 Kolde R, and Kolde MR. Package ‘pheatmap’. R Package, 2015, 1(7):790. Yu G, Li F, Qin Y, Bo X, Wu Y, and Wang S. GOSemSim: an R Package for Measuring Semantic Similarity Among GO Terms and Gene Products. Bioinformatics, 2010, 26(7):976–8., DOI 10.1093/bioinformatics/ btq064 Therneau TM, and Grambsch PM. Modeling Survival Data: Extending the Cox Model. Springer Science & Business Media, 2013). Wickham H. ggplot2: Elegant Graphics for Data Analysis. Springer, 2016). Detchokul S, Williams ED, Parker MW, and Frauman AG. Tetraspanins as Regulators of the Tumour Microenvironment: Implications for Metastasis and Therapeutic Strategies. Br J Pharmacol, 2014, 171(24):5462–90., DOI 10.1111/ bph.12260 Wright MD, Moseley GW, and van Spriel AB. Tetraspanin Microdomains in Immune Cell Signalling and Malignant Disease. Tissue antigens, 2004, 64(5): 533–42., DOI 10.1111/j.1399-0039.2004.00321.x Wang Y, Liang Y, Yang G, Lan Y, Han J, Wang Y, et al. Tetraspanin 1 Promotes Epithelial-To-Mesenchymal Transition and Metastasis of Cholangiocarcinoma Via PI3K/AKT Signaling. J Exp Clin Cancer Res, 2018, 37:300., DOI 10.1186/s13046-018-0969-y Zhou C, Liang Y, Zhou L, Yan Y, Liu N, Zhang R, et al. TSPAN1 Promotes Autophagy Flux and Mediates Cooperation between WNT-CTNNB1 Signaling and Autophagy via the MIR454-Fam83a-TSPAN1 axis in Pancreatic Cancer. Autophagy, 2020, 28:1–21., DOI 10.1080/ 15548627.2020.1826689 Hou F-Q, Lei X-F, Yao J-L, Wang Y-J, and Zhang W. Tetraspanin 1 Is Involved in Survival, Proliferation and Carcinogenesis of Pancreatic Cancer. Oncol Rep, 2015, 34(6):3068–76., DOI 10.3892/or.2015.4272 Tian J, Zhang R, Piao H, Li X, Sheng W, Zhou J, et al. Silencing Tspan1 Inhibits Migration and Invasion, and Induces the Apoptosis of Human Pancreatic Cancer Cells. Mol Med Rep, 2018, 18(3):3280–8., DOI 10.3892/mmr.2018.9331 Zhang X, Shi G, Gao F, Liu P, Wang H, and Tan X. TSPAN1 Upregulates MMP2 to Promote Pancreatic Cancer Cell Migration and Invasion via PLCγ. Oncol Rep, 2019, 41(4):2117–25., DOI 10.3892/or.2019.6989 Ye H, Li T, Wang H, Wu J, Yi C, Shi J, et al. TSPAN1, TMPRSS4, SDR16C5, and CTSE as Novel Panel for Pancreatic Cancer: A Bioinformatics Analysis and Experiments Validation. Front Immunol, 2021, 12:649551., DOI 10.3389/ fimmu.2021.649551 Ma C, Cui Z,Wang Y, Zhang L, Wen J, Guo H, et al. Bioinformatics Analysis Reveals TSPAN1 as a Candidate Biomarker of Progression and Prognosis in Pancreatic Cancer. Bosn J Basic Med Sci, 2021, 21(1):47–60., DOI 10.17305/bjbms.2020.5096 Roskoski R, Jr. The ErbB/HER Family of Protein-Tyrosine Kinases and Cancer. Pharmacol Res, 2014, 79:34–74., DOI 10.1016/j.phrs.2013.11.002 Ghasemi R, Rapposelli IG, Capone E, Rossi C, Lattanzio R, Piantelli M, et al. Dual Targeting of ErbB-2/ErbB-3 Results in Enhanced Antitumor Activity in Preclinical Models of Pancreatic Cancer. Oncogenesis, 2014, 3(8):e117., DOI 10.1038/oncsis.2014.31 Feng H, Wang Y, Su J, Liang H, Zhang C-Y, Chen X, et al. MicroRNA-148a Suppresses the Proliferation and Migration of Pancreatic Cancer Cells by Down-Regulating ErbB3. Pancreas, 2016, 45(9):1263–71., DOI 10.1097/ mpa.0000000000000677 Liles JS, Arnoletti JP, Tzeng C-WD, Howard JH, Kossenkov AV, Kulesza P, et al. ErbB3 Expression Promotes Tumorigenesis in Pancreatic Adenocarcinoma. Cancer Biol Ther, 2010, 10(6):555–63., DOI 10.4161/cbt.10.6.12532 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609868 EP 1609876 DI 10.3389/pore.2021.1609868 PG 9 ER PT J AU Yang, H Wang, K Li, Sh Li, Y Yuan, L AF Yang, Hui Wang, Kunlun Li, Shenglei Li, Yan Yuan, Ling TI The Prognostic Role of PORT and EGFR Mutation Status in Completely Resected Stage IIIA/N2 Non-Small Cell Lung Cancer Patients with Postoperative Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR; NSCLC; stage IIIA/N2; postoperative radiotherapy; prognostic factors ID EGFR; NSCLC; stage IIIA/N2; postoperative radiotherapy; prognostic factors AB Background: The treatment choice for completely resected stage IIIA/N2 non-small cell lung cancer (NSCLC) patients is still controversial now. Our study aims to identify potential prognostic factors in stage IIIA/N2 NSCLC patients with complete surgical resection and postoperative chemotherapy. Methods: In this study, we screened the stage IIIA/N2 NSCLC patients diagnosed in the Affiliated Cancer Hospital of Zhengzhou University from 2015 to 2019. Completely resected patients with postoperative chemotherapy (PCT) were enrolled. The univariate and multivariate COX proportional hazards regression analyses were used to identify the prognostic factors. The Kaplan-Meier survival curve was used to compare the disease-free survival (DFS) and overall survival (OS) in the subgroup analyses. Results: 180 patients were collected, including 142 patients with PCT treatment alone and 38 patients with postoperative radiotherapy (PORT) treatment. The median DFS was 17.8months (95% CI: 16.5–19.1months) and the median OS was 50.6months (47.4–53.9months) in all the patients. The median DFS of the PORT group was significantly longer than the PCT group (38.7 vs 16.7months, p < 0.001). Epidermal growth factor receptor (EGFR) mutation-positive patients had a worse DFS compared with EGFR mutation-negative patients (16.8 vs 18.0months, p = 0.032). Possible prognostic factors were evaluated through univariateCOX regression analysis. The furthermultivariate COX regression analysis showed that patients with PORT(HR: 0.318,95%CI: 0.185–0.547, p < 0.001), EGFRmutation-negative (HR: 0.678, 95% CI: 0.492–0.990, p = 0.044), T1 (HR: 0.661, 95% CI: 0.472–0.925, p = 0.016), and lobectomy (HR: 0.423, 95% CI: 0.191–0.935, p = 0.034), had better DFS. The only independent prognostic factor of OS was the type of surgery (p = 0.013). Conclusion: PORT might improve the DFS of stage IIIA/N2 NSCLC patients with complete surgical resection and PCT, but it cannot increase OS. Besides, EGFR mutation status, T stage, and type of surgery are possible independent prognostic factors for DFS, and type of surgery is associated with OS. These factors remain to be clarified in further studies. C1 [Yang, Hui] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. [Wang, Kunlun] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. [Li, Shenglei] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. [Li, Yan] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. [Yuan, Ling] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. RP Yuan, L (reprint author), The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation Oncology, Zhengzhou, China. EM HNHNYL@126.com CR Siegel RL, Miller KD, and Jemal A. Cancer Statistics, 2020. CA A Cancer J Clin, 2020, 70(1):7–30., DOI 10.3322/caac.21590 Rusch VW, Chansky K, Kindler HL, Nowak AK, Pass HI, Rice DC, et al. The IASLC Mesothelioma Staging Project: Proposals for the MDescriptors and for Revision of the TNM Stage Groupings in the Forthcoming, Eighth, Edition of the TNM Classification for Mesothelioma. J Thorac Oncol, 2016, 11(12): 2112–9., DOI 10.1016/j.jtho.2016.09.124 Veeramachaneni NK, Feins RH, Stephenson BJK, Edwards LJ, and Fernandez FG. Management of Stage IIIA Non-small Cell Lung Cancer by Thoracic Surgeons in North America. Ann Thorac Surg, 2012, 94(3):922–8., DOI 10.1016/j.athoracsur.2012.04.087 Arriagada R, Bergman B, Dunant A, Le Chevalier T, Pignon JP, and Vansteenkiste J. Cisplatin-based Adjuvant Chemotherapy in Patients with Completely Resected Non-small-cell Lung Cancer. N Engl JMed, 2004, 350(4): 351–60., DOI 10.1056/NEJMoa031644 Lally BE, Zelterman D, Colasanto JM, Haffty BG, Detterbeck FC, and Wilson LD. Postoperative Radiotherapy for Stage II or III Non-small-cell Lung Cancer Using the Surveillance, Epidemiology, and End Results Database. Jco, 2006, 24(19):2998–3006., DOI 10.1200/JCO.2005.04.6110 Douillard J-Y, Rosell R, De LenaM, RiggiM, Hurteloup P, andMahe M-A. Impact of Postoperative Radiation Therapy on Survival in Patients with Complete Resection and Stage I, II, or IIIA Non-small-cell Lung Cancer Treated with Adjuvant Chemotherapy: The Adjuvant Navelbine International Trialist Association, ANITA, Randomized Trial. Int J Radiat Oncology*Biology*Physics, 2008, 72(3):695–701., DOI 10.1016/j.ijrobp.2008.01.044 Kim BH, Kim HJ, Wu H-G, Kang CH, Kim YT, Lee S-H, et al. Role of Postoperative Radiotherapy after Curative Resection and Adjuvant Chemotherapy for Patients with Pathological Stage N2 Non-small-cell Lung Cancer: a Propensity Score Matching Analysis. Clin Lung Cancer, 2014, 15(5):356–64., DOI 10.1016/j.cllc.2014.05.005 Drake JA, Portnoy DC, Tauer K, and Weksler B. Adding Radiotherapy to Adjuvant Chemotherapy Does Not Improve Survival of Patients with N2 Lung Cancer. Ann Thorac Surg, 2018, 106(4):959–65., DOI 10.1016/ j.athoracsur.2018.04.074 Hui Z, Men Y, Hu C, Zhou Z, Liang J, Feng Q, et al. OA12.06 A Prospective Randomized Phase Ⅲ Study of Precise PORT for Patients with pⅢA-N2 NSCLC after Complete Resection and Adjuvant Chemotherapy. J Thorac Oncol, 2019, 14(10):S238–S239., DOI 10.1016/j.jtho.2019.08.475 Le Pechoux C, Pourel N, Barlesi F, Faivre-Finn C, Lerouge D, Zalcman G, et al. LBA3_PR an International Randomized Trial, Comparing post-operative Conformal Radiotherapy, PORT, to No PORT, in Patients with Completely Resected Non-small Cell Lung Cancer, NSCLC, and Mediastinal N2 Involvement: Primary End-point Analysis of LungART, IFCT-0503, UK NCRI, SAKK, NCT00410683. Ann Oncol, 2020, 31(4): S1178., DOI 10.1016/j.annonc.2020.08.2280 Zhong W-Z, Wang Q, MaoW-M, Xu S-T,Wu L, Shen Y, et al. Gefitinib versus Vinorelbine Plus Cisplatin as Adjuvant Treatment for Stage II-IIIA, N1-N2, EGFR-Mutant NSCLC, ADJUVANT/CTONG1104): a Randomised, Open- Label, Phase 3 Study. Lancet Oncol, 2018, 19(1):139–48., DOI 10.1016/S1470- 2045(17)30729-5 Wu Y-L, Tsuboi M, He J, John T, Grohe C, Majem M, et al. Osimertinib in Resected EGFR-Mutated Non-small-cell Lung Cancer. N Engl J Med, 2020, 383(18):1711–23., DOI 10.1056/NEJMoa2027071 Zou B, Xu Y, Li T, Li W, Tang B, Zhou L, et al. A Multicenter Retrospective Analysis of Survival Outcome Following Postoperative Chemoradiotherapy in Non-small-cell Lung Cancer Patients with N2 Nodal Disease. Int J Radiat Oncology*Biology*Physics, 2010, 77(2):321–8., DOI 10.1016/ j.ijrobp.2009.05.044 Shen W-y., Ji J, Zuo Y-s., Pu J, Xu Y-m., Zong C-d., et al. Comparison of Efficacy for Postoperative Chemotherapy and Concurrent Radiochemotherapy in Patients with IIIA-pN2 Non-small Cell Lung Cancer: an Early Closed Randomized Controlled Trial. Radiother Oncol, 2014, 110(1):120–5., DOI 10.1016/j.radonc.2013.10.008 Mikell JL, Gillespie TW, Hall WA, Nickleach DC, Liu Y, Lipscomb J, et al. Postoperative Radiotherapy Is Associated with Better Survival in Non-small Cell Lung Cancer with Involved N2 Lymph Nodes: Results of an Analysis of the National Cancer Data Base. J Thorac Oncol, 2015, 10(3):462–71., DOI 10.1097/JTO.0000000000000411 Corso CD, Rutter CE, Wilson LD, Kim AW, Decker RH, and Husain ZA. Reevaluation of the Role of Postoperative Radiotherapy and the Impact of Radiation Dose for Non-small-cell Lung Cancer Using the National Cancer Database. J Thorac Oncol, 2015, 10(1):148–55., DOI 10.1097/ JTO.0000000000000406 Herskovic A, Mauer E, Christos P, and Nagar H. Role of Postoperative Radiotherapy in Pathologic Stage IIIA, N2, Non-small Cell Lung Cancer in a Prospective Nationwide Oncology Outcomes Database. J Thorac Oncol, 2017, 12(2):302–13., DOI 10.1016/j.jtho.2016.09.135 Wang W, Men Y, Wang J, Zhou Z, Chen D, Xiao Z, et al. Postoperative Radiotherapy Is Effective in Improving Survival of Patients with Stage pIII-N2 Non-small-cell Lung Cancer after Pneumonectomy. BMC Cancer, 2019, 19(1):478., DOI 10.1186/s12885-019-5692-3 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609898 EP 1609902 DI 10.3389/pore.2021.1609898 PG 5 ER PT J TI Pathological Features in 100 Deceased Patients With COVID-19 in Correlation With Clinical and Laboratory Data SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE histological subtype; COVID-19; pathological features; large cohort; morphology ID histological subtype; COVID-19; pathological features; large cohort; morphology AB Background: Autopsies on COVID-19 deceased patients have many limitations due to necessary epidemiologic and preventative measures. The ongoing pandemic has caused a significant strain on healthcare systems and is being extensively studied around the world. Clinical data does not always corelate with post-mortem findings. The goal of our study was to find pathognomonic factors associated with COVID-19 mortality in 100 postmortem full body autopsies. Materials and Methods: Following necessary safety protocol, we performed 100 autopsies on patients who were diagnosed with COVID-19 related death. The macroscopic and microscopic pathologies were evaluated along with clinical and laboratory findings. Results: Extensive coagulopathic changes are seen throughout the bodies of diseased patients. Diffuse alveolar damage is pathognomonic of COVID-19 viral pneumonia, and is the leading cause of lethal outcome in younger patients. Extrapulmonary pathology is predominantly seen in the liver and spleen. Intravascular thrombosis is often widespread and signs of septic shock are often present. Conclusion: The described pathological manifestations of COVID-19 in deceased patients are an insight into the main mechanisms of SARS-CoV-2 associated lethal outcome. The disease bears no obvious bias in severity, but seems to be more severe in some patients, hinting at genetic or epigenetic factors at play. CR Reicher S, and Stott C. On order and disorder during the COVID-19 pandemic. Br J Soc Psychol, 2020, 59(3):694–702., DOI 10.1111/bjso.12398 Dong E, Du H, andGardner L. An interactive web-based dashboard to track COVID- 19 in real time. Lancet Inf Dis 20(5):533–4., DOI 10.1016/S1473-3099(20)30120-1 Kobak D. Excess mortality reveals Covid’s true toll in Russia. Significance, 2021, 18(1):16–9., DOI 10.1111/1740-9713.01486 Pomeranz JL, and Schwid AR. Governmental actions to address COVID-19 misinformation. J Public Health Pol, 2021, 42:201–10., DOI 10.1057/s41271- 020-00270-x Partinen M, Bjorvatn B, Holzinger B, Chung F, Penzel T, and Espie C. A ICOSS; collaboration group. Sleep and circadian problems during the coronavirus disease 2019, COVID-19, pandemic: the International COVID-19 Sleep Study, ICOSS). J Sleep Res, 2021, 30(1):e13206., DOI 10.1111/jsr.13206 Riiser K, Helseth S, Haraldstad K, Torbjornsen A, and Richardsen K. R. Adolescents’ health literacy, health protective measures, and health-related quality of life during the Covid-19 pandemic. PloS one, 2020, 15(8):e0238161., DOI 10.1371/journal.pone.0238161 Liu C. H, Stevens C, Conrad R. C, and Hahm H. C. Evidence for elevated psychiatric distress, poor sleep, and quality of life concerns during the COVID-19 pandemic among U.S. young adults with suspected and reported psychiatric diagnoses. Psychiatry Res, 2020, 292:113345., DOI 10.1016/j.psychres.2020.113345 Calabrese F, Fortarezza F, Fortarezza F, Hofman P, Kern I, Panizo A, and Lunardi F. Diffuse Idiopathic Pulmonary Neuroendocrine Cell Hyperplasia, DIPNECH). Virchows archiv, 2020, 1–7., DOI 10.1007/978-3-319-28845- 1_5073-1 Batah S. S, and Fabro A. T. Pulmonary pathology of ARDS in COVID-19: A pathological review for clinicians. Respir Med, 2020, 106239. Barton L. M, Duval E. J, Stroberg E, Ghosh S, and Mukhopadhyay S. COVID- 19 Autopsies, Oklahoma, USA. Am J Clin Pathol, 2020, 153(6):725–33., DOI 10.1093/ajcp/aqaa062 Hanley B, Lucas S. B, Youd E, Swift B, and Osborn M. Autopsy in suspected COVID-19 cases. J Clin Pathol, 2020, 73:239–42., DOI 10.1136/jclinpath-2020- 206522 Mauad T, Hajjar L. A, Callegari G. D, da Silva L. F. F, Schout D, Galas F. R. B. G, et al. Lung pathology in fatal novel human influenza A, H1N1, infection. Am J Respir Crit Care Med, 2010, 181(1):72–9., DOI 10.1164/rccm.200909-1420oc Centers for Disease Control and Prevention. Collection and Submission of Postmortem Specimens from Deceased Persons with Known or Suspected COVID-19, March 2020, Interim Guidance). Available from: www.cdc.gov/coronavirus/2019-ncov/ hcp/guidance-post mortem-specimens.html, Accessed May, 2020). College of American Pathologists, CAP). Amended COVID-19 autopsy guideline statement from the CAP autopsy committee, 2020). Available from: https://covid19.who.int/, Accessed May, 2020). Carsana L, Sonzogni A, Nasr A, Rossi RS, Pellegrinelli A, Zerbi P, et al. Pulmonary post-mortem findings in a series of COVID-19 cases from northern Italy: a two-centre descriptive study. Lancet Infect Dis, 2020, 20(20):1135–40., DOI 10.1016/S1473-3099(20)30434-5 Wichmann D, Sperhake JP, Lutgehetmann M, Steurer S, Edler C, Heinemann A, et al. Autopsy Findings and Venous Thromboembolism in Patients With COVID-19 [published online ahead of print, 2020 May 6]. Ann Intern Med, 2020, M20–2003., DOI 10.7326/M20-2003 Centers for Disease Control and Prevention, CDC). Interim laboratory biosafety guidelines for handling and processing specimens associated with coronavirus disease 2019, COVID-19,, 2020). https://www.cdc.gov/coronavirus/ 2019-nCoV/lab/lab-biosafetyguidelines.html, Accessed May, 2020). World Health Organization. Laboratory biosafety guidance related to the novel coronavirus, 2019-nCoV): interim guidance, 2020). https://www.who.int/ publications-detail/laboratory-biosafetyguidance-related-to-coronavirusdisease- 2019-(covid-19, Accessed May, 2020). Iwen P. C, Stiles K. L, and Pentella M. A. Safety Considerations in the Laboratory Testing of Specimens Suspected or Known to Contain the Severe Acute Respiratory Syndrome Coronavirus 2, SARS-CoV-2). Lab Med, 2020, 51:239–42., DOI 10.1093/labmed/lmaa018 Barbareschi M, Ascoli V, Bonoldi E, Cavazza A, Colombari R, Cozzi I, et al. Biosafety in surgical pathology in the era of SARS-Cov2 pandemia. A statement of the Italian Society of Surgical Pathology and Cytology. Pathologica, 2020, 12(2):59–63., DOI 10.32074/1591-951X-14-20 Henwood A. F. Coronavirus disinfection in histopathology. J Histotechnology, 2020, 43:102–4., DOI 10.1080/01478885.2020 Duarte-Neto AN, Aparecida de Almeida Monteiro R, Ferraz da Silva LF, Costa Malheiros DM. A, de Oliveira E. P, Filho JT, et al. Pulmonary and systemic involvement of COVID-19 assessed by ultrasound-guided minimally invasive autopsy. Histopathology 77(2):186–97., DOI 10.1111/his.14160 Zhang H, Zhou P, Wei Y, Yue H, Hu M, Zhang S, et al. Histopathologic Changes and SARS-CoV-2 Immunostaining in the Lung of a Patient With COVID-19. Ann Intern Med, 2020, 172(9):629–32. Tang N, Li D, Wang X, and Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost, 2020, 18(4):844–7., DOI 10.1111/jth.14768 Tang N, Bai H, Chen X, Gong J, Li D, and Sun Z. Anticoagulant treatment is associated with decreasedmortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost, 2020, 18(5):1094–9., DOI 10.1111/jth.14817 Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical course and risk factors for mortality of adult inpatients t with COVID-19 in Wuhan, China: a retrospective cohort study [published correction appears in Lancet. 2020; 395(10229):1038., DOI 10.1016/s0140-6736(20)30566-3 Wang Y, Lu X, Li Y, Chen H, Chen T, Su N, et al. Clinical Course and Outcomes of 344 Intensive Care Patients with COVID-19. Am J Respir Crit Care Med, 2020, 201(11):1430–4., DOI 10.1164/rccm.202003-0736le Du Y, Tu L, Zhu P, Mu M,Wang R, Yang P, et al. Clinical Features of 85 Fatal Cases of COVID-19 from Wuhan. A Retrospective Observational Study. Am J Respir Crit Care Med, 2020, 201(11):1372–9., DOI 10.1164/rccm.202003- 0543oc Guan W-j., Ni Z-y., Hu Y, Liang W-h., Ou C-q., He J-x., et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med, 2020, 382:1708–20., DOI 10.1056/NEJMoa2002032 Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia inWuhan, China. JAMAInternMed, 2020, 180(7): 934–43., DOI 10.1001/jamainternmed.2020.0994 Zayratyants OV, Samsonova MV, Mikhaleva LM, Chernyaev AL, Mishnev OD, Krupnov NM, et al. Moscow, GBU «NIIOOZM DZM»,.–, 2020). p. 142. Xu Z, Shi L,Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med, 2020, 8:420–2., DOI 10.1016/S2213-2600(20)30076-X Fox S. E, Akmatbekov A, Harbert J. L, Li G, Brown J. Q, and Heide R. S. V. Pulmonary and Cardiac Pathology in Covid-19: The First Autopsy Series from New Orleans. Lancet Respir Med. 8(7):681–6., DOI 10.1101/2020.04.06.20050575 Dolhnikoff M, Duarte-Neto AN, de Almeida Monteiro RA, da Silva LFF, de Oliveira EP, Nascimento Saldiva PH, et al. Pathological evidence of pulmonary thrombotic phenomena in severe COVID-19. J Thromb Haemost, 2020, 18(6): 1517–9. Lillicrap D. Disseminated intravascular coagulation in patients with 2019- nCoV pneumonia. J Thromb Haemost, 2020, 18(4):786–7., DOI 10.1111/ jth.14781 Yao XH, Li TY, He ZC, Ping YF, Liu HW, Yu SC, et al. [A pathological report of three COVID-19 cases by minimal invasive autopsies]. Zhonghua Bing Li Xue Za Zhi, 2020, 49(0):411–7., DOI 10.3760/cma.j.cn112151-20200312-00193 Tian S, Xiong Y, Liu H, and Niu L. Pathological study of the 2019 novel coronavirus disease, COVID-19, through post-mortem core biopsies. Mod Pathol, 2020, 33:1007–1014. Mason R. J. Pathogenesis of COVID-19 from a cell biology perspective. Eur Respir J, 2020, 55(4):2000607, 2020 . Published 2020 Apr 16., DOI 10.1183/ 13993003.00607-2020 Lukassen S, Chua R. L, Trefzer T, Kahn N. C, Schneider M. A, Muley T, et al. SARS -CoV-2 receptor ACE 2 and TMPRSS 2 are primarily expressed in bronchial transient secretory cells. EMBO J, 2020, 39:e105114., DOI 10.15252/ embj.20105114 Ackermann M, Verleden S. E, Kuehnel M, Haverich A, Welte T, Laenger F, et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N Engl J Med, 2020, 383(2):120–8., DOI 10.1056/nejmoa2015432 Kollias A, Kyriakoulis K. G, Dimakakos E, Poulakou G, Stergiou G. S, and Syrigos K. Thromboembolic risk and anticoagulant therapy in COVID-19 patients: emerging evidence and call for action. Br J Haematol, 2020, 189(5): 846–7., DOI 10.1111/bjh.16727 Grasselli G, Zangrillo A, Zanella A, Antonelli M, Cabrini L, Castelli A, et al. Baseline Characteristics and Outcomes of 1591 Patients Infected With SARSCoV- 2 Admitted to ICUs of the Lombardy Region, Italy [published online ahead of print, 2020 Apr 6]. JAMA, 2020, 323(16):1574–81., DOI 10.1001/ jama.2020.4031 Klok F. A, Kruip M. J. H. A, van der Meer N. J. M, Arbous M. S, Gommers D. A. M. P. J, Kant K. M, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res, 2020, 191:145–7., DOI 10.1016/ j.thromres.2020.04.013 Oudkerk M, Buller HR, Kuijpers D, Van Es N, Oudkerk S. F, McLoud T, et al. Diagnosis, Prevention, and Treatment of Thromboembolic Complications in COVID-19. Radiology, 2020, 201629, 2020 . Report of the National Institute for Public Health of the Netherlands [published online ahead of print, 2020 Apr 23]., DOI 10.1148/radiol.2020201629 Kovrigina A. M, Shalamova EA, Shalamova E. A, Berezovskiy Y. S, Kalinin D. V, Gretsov E. M, et al. Pathomorphological and immunohistochemical features of lymph nodes in COVID-19 patients, autopsy study). Clin Exp Morphol, 2020, 9(Dec. 2020):12–23., DOI 10.31088/CEM2020.9.4.12-23 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609900 EP 1609911 DI 10.3389/pore.2021.1609900 PG 12 ER PT J AU Xu, F Tian, D Shi, X Sun, K Chen, Y AF Xu, Fangfang Tian, Dandan Shi, Xiaoyang Sun, Kai Chen, Yuqing TI Analysis of the Expression and Prognostic Potential of a Novel Metabolic Regulator ANGPTL8/ Betatrophin in Human Cancers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE inflammation; prognostic biomarker; ANGPTL8/betatrophin; differential expression analysis; lipid metabolism; glucose homeostasis; Akt signaling ID inflammation; prognostic biomarker; ANGPTL8/betatrophin; differential expression analysis; lipid metabolism; glucose homeostasis; Akt signaling AB The angiopoietin-like protein (ANGPTL) family members, except for the novel atypical member ANGPTL8/betatrophin, have been reported to participate in angiogenesis, inflammation and cancer. ANGPTL8/betatrophin is a metabolic regulator that is involved in lipid metabolism and glucose homeostasis. However, little is known about the expression and prognostic value of ANGPTL8/betatrophin in human cancers. In this study, we first conducted detailed analyses of ANGPTL8/betatrophin expression in cancer/normal samples via the Human Protein Atlas (HPA), Gene Expression Profiling Interactive Analysis (GEPIA), DriverDBv3, ENCORI and UALCAN databases. ANGPTL8/ betatrophin showed high tissue specificity (enriched in the liver) and cell-type specificity (enriched in HepG2 and MCF7 cell lines). More than one databases demonstrated that the gene expression of ANGPTL8/betatrophin was significantly lower in cholangiocarcinoma (CHOL), breast invasive carcinoma (BRCA), lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), uterine corpus endometrial carcinoma (UCEC), and significantly higher in kidney renal clear cell carcinoma (KIRC) compared with that in normal samples. However, the protein expression of ANGPTL8/betatrophin displayed opposite results in clear cell renal cell carcinoma (ccRCC)/KIRC. Based on the expression profiles, the prognostic value was evaluated with the GEPIA, DriverDBv3, Kaplan Meier plotter and ENCORI databases. Two or more databases demonstrated that ANGPTL8/betatrophin significantly affected the survival of KIRC, uterine corpus endometrial carcinoma (UCEC), pheochromocytoma and paraganglioma (PCPG) and sarcoma (SARC); patients with PCPG and SARC may benifit from high ANGPTL8/betatrophin expression while high ANGPTL8/betatrophin expression was associated with poor prognosis in KIRC and UCEC. Functional analyses with the GeneMANIA, Metascape and STRING databases suggested that ANGPTL8/betatrophin was mainly involved in lipid homeostasis, especially triglyceride and cholesterol metabolism; glucose homeostasis, especially insulin resistance; AMPK signaling pathway; PI3K/Akt signaling pathway; PPAR signaling pathway; mTOR signaling pathway; HIF-1 signaling pathway; autophagy; regulation of inflammatory response. ANGPTL8/betatrophin may be a promising prognostic biomarker and therapeutic target, thus providing evidence to support further exploration of its role in defined human cancers. C1 [Xu, Fangfang] Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital, Clinical Medical Research CenterZheng Zhou, China. [Tian, Dandan] Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital, Department of HypertensionZheng Zhou, China. [Shi, Xiaoyang] Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital, Department of EndocrinologyZheng Zhou, China. [Sun, Kai] Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital, Department of HematologyZheng Zhou, China. [Chen, Yuqing] Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital, Department of HematologyZheng Zhou, China. RP Sun, K (reprint author), Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital, Department of Hematology, Zheng Zhou, China. EM sunkai@cellscience.org CR Tseng YH, Yeh YH, Chen WJ, and Lin KH. Emerging Regulation and Function of Betatrophin. Int J Mol Sci, 2014, 15(15):23640–57., DOI 10.3390/ ijms151223640 Zhang R, and Abou-Samra AB. A Dual Role of Lipasin, Betatrophin, in Lipid Metabolism and Glucose Homeostasis: Consensus and Controversy. Cardiovasc Diabetol, 2014, 13:133., DOI 10.1186/s12933-014-0133-8 Wang Y, Quagliarini F, Gusarova V, Gromada J, Valenzuela DM, Cohen JC, et al. Mice Lacking ANGPTL8, Betatrophin, Manifest Disrupted Triglyceride Metabolism without Impaired Glucose Homeostasis. Proc Natl Acad Sci U S A, 2013, 110(110):16109–14., DOI 10.1073/pnas.1315292110 Izumi R, Kusakabe T, Noguchi M, Iwakura H, Tanaka T, Miyazawa T, et al. CRISPR/Cas9-mediated Angptl8 Knockout Suppresses Plasma Triglyceride Concentrations and Adiposity in Rats. J Lipid Res, 2018, 59(59):1575–85., DOI 10.1194/jlr.M082099 Siddiqa A, Cirillo E, Tareen SHK, Ali A, Kutmon M, Eijssen LMT, et al. Visualizing the Regulatory Role of Angiopoietin-like Protein 8, ANGPTL8, in Glucose and Lipid Metabolic Pathways. Genomics, 2017, 109(109):408–18., DOI 10.1016/j.ygeno.2017.06.006 Zhang D, Yu YJ, Xu FS, Yuan JH, Wang R, Zhang CS, et al. Recombinant Betatrophin, Angptl-8/lipasin, Ameliorates Streptozotocin-Induced Hyperglycemia and β-cell Destruction in Neonatal Rats. Mol Med Rep, 2019, 20(20):4523–32., DOI 10.3892/mmr.2019.10719 Cheng C, Geng F, Cheng X, and Guo D. Lipid Metabolism Reprogramming and its Potential Targets in Cancer. Cancer Commun, Lond,, 2018, 38(38):27., DOI 10.1186/s40880-018-0301-4 Huang C, and Freter C. Lipid Metabolism, Apoptosis and Cancer Therapy. Int J Mol Sci, 2015, 16(16):924–49., DOI 10.3390/ijms16010924 Santos CR, and Schulze A. Lipid Metabolism in Cancer. Febs J, 2012, 279(279): 2610–23., DOI 10.1111/j.1742-4658.2012.08644.x Annibaldi A, and Widmann C. Glucose Metabolism in Cancer Cells. Curr Opin Clin Nutr Metab Care, 2010, 13(13):466–70., DOI 10.1097/ MCO.0b013e32833a5577 Ghanbari Movahed Z, Rastegari-Pouyani M, Mohammadi Mh., and Mansouri K. Cancer Cells Change Their Glucose Metabolism to Overcome Increased ROS: One Step from Cancer Cell to Cancer Stem Cell? Biomed Pharmacother, 2019, 112:108690., DOI 10.1016/j.biopha.2019.108690 Endo M. The Roles of ANGPTL Families in Cancer Progression. J UOEH, 2019, 41(41):317–25., DOI 10.7888/juoeh.41.317 Rong Guo X, Li Wang X, Chen Y, Hong Yuan Y, Mei Chen Y, Ding Y, et al. ANGPTL8/betatrophin Alleviates Insulin Resistance via the Akt-Gsk3β or Akt-FoxO1 Pathway in HepG2 Cells. Exp Cel Res, 2016, 345(345):158–67., DOI 10.1016/j.yexcr.2015.09.012 Zhang Z, Wu H, Dai L, Yuan Y, Zhu Y, Ma Z, et al. ANGPTL8 Enhances Insulin Sensitivity by Directly Activating Insulin-Mediated AKT Phosphorylation. Gene, 2020, 749:144707., DOI 10.1016/j.gene.2020.144707 Shariati M, and Meric-Bernstam F. Targeting AKT for Cancer Therapy. Expert Opin Investig Drugs, 2019, 28(28):977–88., DOI 10.1080/13543784. 2019.1676726 Song M, Bode AM, Dong Z, and Lee MH. AKT as a Therapeutic Target for Cancer. Cancer Res, 2019, 79(79):1019–31., DOI 10.1158/0008-5472.CAN-18- 2738 Revathidevi S, and Munirajan AK. Akt in Cancer: Mediator and More. Semin Cancer Biol, 2019, 59:80–91., DOI 10.1016/j.semcancer.2019.06.002 Pazgan-Simon M, Zuwala-Jagiello J, Menzyk T, Bator M, Derra A, Lekstan A, et al. Serum Betatrophin and Irisin Levels in Hepatocellular Carcinoma. J Physiol Pharmacol, 2020, 71(71):1., DOI 10.26402/jpp.2020.1.11 Monzavi N, Zargar SJ, Gheibi N, Azad M, and Rahmani B. Angiopoietin-like Protein 8, Betatrophin, May Inhibit Hepatocellular Carcinoma through Suppressing of the Wnt Signaling Pathway. Iran J Basic Med Sci, 2019, 22(22):1166–71., DOI 10.22038/ijbms.2019.36612.8764 Maurer L, Schwarz F, Fischer-Rosinsky A, Schlueter N, Brachs S, Mohlig M, et al. Renal Function Is Independently Associated with Circulating Betatrophin. Plos One, 2017, 12(12):e0173197., DOI 10.1371/ journal.pone.0173197 Tang Z, Kang B, Li C, Chen T, and Zhang Z. GEPIA2: an Enhanced Web Server for Large-Scale Expression Profiling and Interactive Analysis. Nucleic Acids Res, 2019, 47(47):W556–60., DOI 10.1093/nar/gkz430 Liu SH, Shen PC, Chen CY, Hsu AN, Cho YC, Lai YL, et al. DriverDBv3: a Multi-Omics Database for Cancer Driver Gene Research. Nucleic Acids Res, 2020, 48(48):D863–70., DOI 10.1093/nar/gkz964 Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce- Rodriguez I, Chakravarthi BVSK, et al. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses. Neoplasia, 2017, 19(19):649–58., DOI 10.1016/j.neo.2017.05.002 Chen F, Chandrashekar DS, Varambally S, and Creighton CJ. Pan-cancer Molecular Subtypes Revealed by Mass-Spectrometry-Based Proteomic Characterization of More Than 500 Human Cancers. Nat Commun, 2019, 10(10):5679., DOI 10.1038/s41467-019-13528-0 Nagy A, Munkacsy G, and Gyorffy B. Pancancer Survival Analysis of Cancer Hallmark Genes. Sci Rep, 2021, 11(11):6047., DOI 10.1038/s41598-021-84787-5 Warde-Farley D, Donaldson SL, Comes O, Zuberi K, Badrawi R, Chao P, et al. The GeneMANIA Prediction Server: Biological Network Integration for Gene Prioritization and Predicting Gene Function. Nucleic Acids ResWeb Server issue, 2010, 38(38):W214–20., DOI 10.1093/nar/gkq537 Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. Metascape Provides a Biologist-Oriented Resource for the Analysis of Systems- Level Datasets. Nat Commun, 2019, 10(10):1523., DOI 10.1038/s41467-019- 09234-6 Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, et al. The STRING Database in 2021: Customizable Protein-Protein Networks, and Functional Characterization of User-Uploaded Gene/measurement Sets. Nucleic Acids Res, 2021, 49(49):D605–12., DOI 10.1093/nar/gkaa1074 Ehrlich KC, Lacey M, and Ehrlich M. Tissue-specific Epigenetics of Atherosclerosis-Related ANGPT and ANGPTL Genes. Epigenomics, 2019, 11(11):169–86., DOI 10.2217/epi-2018-0150 Wang Q, Zhang H, Chen Q, Wan Z, Gao X, and Qian W. Identification of METTL14 in Kidney Renal Clear Cell Carcinoma Using Bioinformatics Analysis. Dis Markers, 2019, 2019:5648783., DOI 10.1155/2019/5648783 Chen C-C, Susanto H, Chuang W-H, Liu T-Y, and Wang C-H. Higher Serum Betatrophin Level in Type 2 Diabetes Subjects Is Associated with Urinary Albumin Excretion and Renal Function. Cardiovasc Diabetol, 2016, 15:3., DOI 10.1186/s12933-015-0326-9 Fang C, Huang Y, Guo H, Gao Y, Ji X, and Hu J. Lipasin, a Biomarker of Diabetic Retinopathy. Diabetes Res Clin Pract, 2016, 115:96–8., DOI 10.1016/ j.diabres.2016.01.029 Zheng T, Ge B, Liu H, Chen B, Qin L, Xiao L, et al. Triglyceride-mediated Influence of Serum Angiopoietin-like Protein 8 on Subclinical Atherosclerosis in Type 2 Diabetic Patients: Results from the GDMD Study in China. Cardiovasc Diabetol, 2018, 17(17):84., DOI 10.1186/s12933-018-0687-y Ejarque M, Borlaug M, Vilarrasa N, Martinez-Perez B, Llaurado G, Megia A, et al. Angiopoietin-like Protein 8/betatrophin as a New Determinant of Type 2 Diabetes Remission after Bariatric Surgery. Translational Res, 2017, 184: 35–44., DOI 10.1016/j.trsl.2017.03.001 Wang R, Yuan J, Zhang C, Wang L, Liu Y, Song L, et al. Neuropeptide Y-Positive Neurons in the Dorsomedial Hypothalamus Are Involved in the Anorexic Effect of Angptl8. Front Mol Neurosci, 2018, 11:451., DOI 10.3389/ fnmol.2018.00451 Chung HS, Lee MJ, Hwang SY, Lee HJ, Yoo HJ, Seo J-A, et al. Circulating Angiopoietin-like Protein 8, ANGPTL8, and ANGPTL3 Concentrations in Relation to Anthropometric and Metabolic Profiles in Korean Children: a Prospective Cohort Study. Cardiovasc Diabetol, 2016, 15:1., DOI 10.1186/ s12933-015-0324-y Li Z, and Zhang H. Reprogramming of Glucose, Fatty Acid and Amino Acid Metabolism for Cancer Progression. Cell Mol Life Sci, 2016, 73(73):377–92., DOI 10.1007/s00018-015-2070-4 Abdel-Wahab AF, Mahmoud W, and Al-Harizy RM. Targeting Glucose Metabolism to Suppress Cancer Progression: Prospective of Anti-glycolytic Cancer Therapy. Pharmacol Res, 2019, 150:104511., DOI 10.1016/j.phrs.2019.104511 Lu P, Chen X, Zhang Z, Zhang J, Yang Y, Liu Z, et al. Insulin Upregulates Betatrophin Expression via PI3K/Akt Pathway. Sci Rep, 2017, 7(7):5594., DOI 10.1038/s41598-017-06052-y Zhang L, Shannon CE, Bakewell TM, Abdul-Ghani MA, Fourcaudot M, and Norton L. Regulation of ANGPTL8 in Liver and Adipose Tissue by Nutritional and Hormonal Signals and its Effect on Glucose Homeostasis in Mice. Am J Physiol Endocrinol Metab, 2020, 318(318):E613–24., DOI 10.1152/ ajpendo.00339.2019 Hahnefeld L, Gruber L, Schomel N, Fischer C, Mattjus P, Gurke R, et al. Ether Lipid and Sphingolipid Expression Patterns Are Estrogen Receptor- Dependently Altered in Breast Cancer Cells. Int J Biochem Cel Biol, 2020, 127:105834., DOI 10.1016/j.biocel.2020.105834 Hoerner CR, Miao SY, Hsieh JJ, and Fan AC. Targeting Metabolic Pathways in Kidney Cancer: Rationale and Therapeutic Opportunities. Cancer J, 2020, 26(26):407–18., DOI 10.1097/PPO.0000000000000472 Dijk W, and Kersten S. Regulation of LipidMetabolism by Angiopoietin-like Proteins. Curr Opin Lipidol, 2016, 27(27):249–56., DOI 10.1097/MOL.0000000000000290 Kersten S. Angiopoietin-like 3 in Lipoprotein Metabolism. Nat Rev Endocrinol, 2017, 13(13):731–9., DOI 10.1038/nrendo.2017.119 Abu-Farha M, Ghosh A, Al-Khairi I, Madiraju SRM, Abubaker J, and Prentki M. The Multi-Faces of Angptl8 in Health and Disease: Novel Functions beyond Lipoprotein Lipase Modulation. Prog Lipid Res, 2020, 80:101067., DOI 10.1016/j.plipres.2020.101067 Chang WH, and Lai AG. The Pan-Cancer Mutational Landscape of the PPAR Pathway Reveals Universal Patterns of Dysregulated Metabolism and Interactions with Tumor Immunity and Hypoxia. Ann N Y Acad Sci, 2019, 1448(1448):65–82., DOI 10.1111/nyas.14170 Stalin A, Lin D, Josephine Princy J, Feng Y, Xiang H, Ignacimuthu S, et al. Computational Analysis of Single Nucleotide Polymorphisms, SNPs, in PPAR Gamma Associated with Obesity, Diabetes and Cancer. J Biomol Struct Dyn, 2020, 1–15., DOI 10.1080/07391102.2020.1835724 Lee J, Hong S-W, Park SE, Rhee E-J, Park C-Y, Oh K-W, et al. AMP-activated Protein Kinase Suppresses the Expression of LXR/SREBP-1 Signaling-Induced ANGPTL8 in HepG2 Cells. Mol Cell Endocrinol, 2015, 414:148–55., DOI 10.1016/j.mce.2015.07.031 Carbone C, Piro G, Merz V, Simionato F, Santoro R, Zecchetto C, et al. Angiopoietin-Like Proteins in Angiogenesis, Inflammation and Cancer. Int J Mol Sci, 2018, 19(19)., DOI 10.3390/ijms19020431 Zheng J, Umikawa M, Cui C, Li J, Chen X, Zhang C, et al. Inhibitory Receptors Bind ANGPTLs and Support Blood Stem Cells and Leukaemia Development. Nature, 2012, 485(485):656–60., DOI 10.1038/nature11095 Kadomatsu T, and Oike Y. Roles of Angiopoietin-like Proteins in Regulation of Stem Cell Activity. J Biochem, 2019, 165(165):309–15., DOI 10.1093/jb/mvz005 Santulli G. Angiopoietin-like Proteins: a Comprehensive Look. Front Endocrinol, 2014, 5:4., DOI 10.3389/fendo.2014.00004 Yang Y, Jiao X, Li L, Hu C, Zhang X, Pan L, et al. Increased Circulating Angiopoietin-like Protein 8 Levels Are Associated with Thoracic Aortic Dissection and Higher Inflammatory Conditions. Cardiovasc Drugs Ther, 2020, 34(34):65–77., DOI 10.1007/s10557-019-06924-7 Zhang Y, Guo X, Yan W, Chen Y, Ke M, Cheng C, et al. ANGPTL8 Negatively Regulates NF-Κb Activation by Facilitating Selective Autophagic Degradation of IKKγ. Nat Commun, 2017, 8(8):2164., DOI 10.1038/s41467-017-02355-w DiDonato JA, Mercurio F, and Karin M. NF-κB and the Link between Inflammation and Cancer. Immunol Rev, 2012, 246(246):379–400., DOI 10.1111/j.1600-065X.2012.01099.x Perkins ND. The Diverse and Complex Roles of NF-Κb Subunits in Cancer. Nat Rev Cancer, 2012, 12(12):121–32., DOI 10.1038/nrc3204 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609914 EP 1609928 DI 10.3389/pore.2021.1609914 PG 15 ER PT J TI Late Radiation–Related Toxicities in Patients Treated for Early-Stage Cervical Carcinoma by Surgery and Adjuvant Radiotherapy: A Retrospective Imaging Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer; toxicity; radiation-induced tumor; complication; bone; comorbidity ID cancer; toxicity; radiation-induced tumor; complication; bone; comorbidity AB Surgical treatment is preferred therapy of early-stage cervical carcinoma. In the risk of cancer recurrence surgery is often followed by adjuvant radiotherapy. In our retrospective study we aimed at identifying late (≥6 months) and very late (≥5 years) radiation adverse effects on imaging scans as CT, PET/CT and MRI in patients who underwent successful treatment for cervical carcinoma by radical surgery combined with radiotherapy ± chemotherapy. We correlated imaging results with clinical manifestations. We selected young and middle-aged patients with long life expectancy, as late radiation-related toxicities may significantly affect their quality of life. Patients were selected from those who were primary diagnosed and treated between the years 1987–2011 and regularly visited our Oncology department in years 2011–2012. Following inclusion criteria were applied: age ≤55 years at diagnosis, clinical follow-up ≥5 years and at least one tomography scan ≥3 years after finished treatment. One hundred and three subjects were reviewed: 73 patients met all inclusion criteria, while 30 patients fulfilled the inclusion criteria except for available tomography scan ≥3 years after therapy. The mean imaging follow-up was 11.2 ± 7.6 years and the mean clinical follow-up was 15.0 ± 6.9 years. In 20 (27%) subjects 27 cases grade I radiation-related toxicities were found; 9 (33%) of those 27 cases were clinically silent. In 14 (19%) females only grade I toxicities were observed. Grade III-IV toxicities were found in 5 (6.8%) subjects. No grade V toxicities were observed. We concluded that severe late side effects caused by radiotherapy were exceedingly rare in females successfully treated for early-stage cervical carcinoma, only 1 bilateral osteonecrosis, 2 cases of ileus, and 2 potentially radiation-induced tumors were found. The majority of radiation-related comorbidities found on imaging scans were clinically silent. CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Bhatla N, Aoki D, Sharma DN, and Sankaranarayanan R. Cancer of the cervix uteri. Int J Gynecol Obstet, 2018, 143(Suppl. 2):22–36., DOI 10.1002/ijgo.12611 Gaffney DK. Optimal therapy for IB2 and IIA2 cervical cancer: surgery or chemoradiotherapy? J Gynecol Oncol, 2012, 23(4):207–20., DOI 10.3802/ jgo.2012.23.4.207 Hricak H, Gatsonis C, Chi DS, Amendola MA, Brandt K, Schwartz LH, et al. Role of Imaging in Pretreatment Evaluation of Early Invasive Cervical Cancer: Results of the Intergroup Study American College of Radiology Imaging Network 6651-Gynecologic Oncology Group 183. Jco, 2005, 23(36): 9329–37., DOI 10.1200/jco.2005.02.0354 Kraljevic Z, Viskovic K, Ledinsky M, Zadravec D, Grbavac I, Bilandzija M, et al. Primary uterine cervical cancer: correlation of preoperative magnetic resonance imaging and clinical staging, FIGO, with histopathology findings. Coll Antropol, 2013, 37(2):561–8. Viswanathan AN, Lee LJ, Eswara JR, Horowitz NS, Konstantinopoulos PA, Mirabeau-Beale KL, et al. Complications of pelvic radiation in patients treated for gynecologic malignancies. Cancer, 2014, 120(24):3870–83., DOI 10.1002/ cncr.28849 Singh GK, Yadav V, Singh P, and Bhowmik KT. Radiation-Induced Malignancies Making Radiotherapy a "Two-Edged Sword": A Review of Literature. World J Oncol, 2017, 8(1):1–6., DOI 10.14740/wjon996w Vale C, Tierney JF, Stewart LA, Brady M, Dinshaw K, Jakobsen A, et al. Reducing uncertainties about the effects of chemoradiotherapy for cervical cancer: a systematic review and meta-analysis of individual patient data from 18 randomized trials. Jco, 2008, 26(35):5802–12., DOI 10.1200/jco.2008.16.4368 Malikova H, Burghardtova M, Fejfarova K, Nadova K, and Weichet J. Advanced cervical cancer in young women: imaging study of late and very late radiation-related side effects after successful treatment by combined radiotherapy. Quant Imaging Med Surg, 2021, 11(1):21–31., DOI 10.21037/ qims-20-553 Yuce Sari S, Guler OC, Gultekin M, Onal HC, and Yildiz F. Adjuvant Small Pelvic Radiotherapy in Patients with Cervical Cancer Having Intermediate Risk Factors Only - Is it Sufficient? Oncol Res Treat, 2017, 40(9):523–7., DOI 10.1159/000476037 Common terminology criteria for adverse events v5.0 Available at: www. meddra.org, Accessed Jan 6, 2021). Yu H, Zhang L, Du X, and Sheng X. Postoperative adjuvant chemotherapy combined with intracavitary brachytherapy in early-stage cervical cancer patients with intermediate risk factors. Ott, 2016, Vol. 9:7331–5., DOI 10.2147/ott.s107146 Davila Fajardo R, van Os R, Buist MR, Uitterhoeve L, Westermann AM, and Kenter GG. Postoperative radiotherapy in patients with early stage cervical cancer. Gynecol Oncol, 2014, 134:52–9., DOI 10.1016/ j.ygyno.2014.04.045 Braunstein S, and Nakamura JL. Radiotherapy-induced malignancies: review of clinical features, pathobiology, and evolving approaches for mitigating risk. Front Oncol, 2013, 3:73., DOI 10.3389/fonc.2013.00073 Cahan WG, Woodard HQ, Higinbotham NL, Stewart FW, and Coley BL. Sarcoma arising in irradiated bone: report of eleven cases. Cancer, 1998, 82(1):8–34., DOI 10.1002/(sici)1097-0142(19980101)82:1<8:aidcncr3> 3.0.co;2-w Hall EJ, and Wuu CS. Radiation -induced second cancers: the impact of 3DCRT and IMRT. Int J Radiat Oncol Biol Phys, 2003, 56(1):83–8., DOI 10.1016/ s0360-3016(03)00073-7 Kwon JW, Huh SJ, Yoon YC, Choi SH, Jung JY, Oh D, et al. Pelvic bone complications after radiation therapy of uterine cervical cancer: evaluation with MRI. Am J Roentgenol, 2008, 191(4):987–94., DOI 10.2214/ajr.07.3634 Papadopoulou I, Stewart V, Barwick TD, Park WH, Soneji N, Rockall AG, et al. Post-Radiation Therapy Imaging Appearances in Cervical Carcinoma. Radiographics, 2016, 36(2):538–53., DOI 10.1148/rg.2016150117 Lundin B, Bjorkholm E, Lundell M, and Jacobsson H. Insufficiency fractures of the sacrum after radiotherapy for gynecological malignancy. Acta Oncol, 1990, 29:211–5., DOI 10.3109/02841869009126547 Schmeler KM, Jhingran A, Iyer RB, Sun CC, Eifel PJ, Soliman PT, et al. Pelvic fractures after radiotherapy for cervical cancer: implications for survivors. Cancer, 2010, 116(3):625., DOI 10.1002/cncr.24811 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609915 EP 1609923 DI 10.3389/pore.2021.1609915 PG 9 ER PT J AU Galffy, G Vastag, A Bogos, K Kiss, Z Ostoros, Gy Muller, V Urban, L Bittner, N Sarosi, V Polanyi, Z Nagy-Erdei, Zs Daniel, A Knollmajer, K Varnai, M Szegner, P Voko, Z Nagy, B Horvath, K Rokszin, Gy Abonyi-Toth, Zs Pozsgai, E Barcza, Zs Moldvay, J Tamasi, L AF Galffy, Gabriella Vastag, Aladar Bogos, Krisztina Kiss, Zoltan Ostoros, Gyula Muller, Veronika Urban, Laszlo Bittner, Nora Sarosi, Veronika Polanyi, Zoltan Nagy-Erdei, Zsofia Daniel, Andrea Knollmajer, Kata Varnai, Mate Szegner, Peter Voko, Zoltan Nagy, Balazs Horvath, Krisztian Rokszin, Gyorgy Abonyi-Toth, Zsolt Pozsgai, Eva Barcza, Zsofia Moldvay, Judit Tamasi, Lilla TI Significant Regional Differences in Lung Cancer Incidence in Hungary: Epidemiological Study Between 2011 and 2016 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE epidemiology; Hungary; mortality; lung cancer; incidence; Hungarian regions ID epidemiology; Hungary; mortality; lung cancer; incidence; Hungarian regions AB Objective: Hungary has one of the highest incidences and mortality rates of lung cancer (LC), therefore the objective of this study was to analyse and compare LC incidence and mortality rates between the main Hungarian regions. Methods: This nationwide, retrospective study used data from the National Health Insurance Fund and included patients aged ≥20 years who were diagnosed with lung cancer (ICD-10 C34) between Jan 1, 2011 and Dec 31, 2016. Age-standardized incidence and mortality rates were calculated and compared for the main regions. Results: The highest incidence rate in males was recorded in Northern Hungary (146.8/ 100,000 person-years [PY]), while the lowest rate was found in Western Transdanubia (94.7/100,000 PY in 2011). All rates showed a declining trend between 2011 and 2016, with the largest decrease in the Northern Great Plain (−20.0%; p = 0.008). LC incidence and mortality rates in women both showed a rising tendency in all regions of Hungary, reaching the highest in Central Hungary (59.86/100,000 PY in 2016). Lung cancer incidence and mortality rates in males correlated with the level of education and smoking prevalence (p = 0.006 and p = 0.01, respectively) in the regions. A correlation with GDP per capita and Health Development Index (HDI) index could also be observed in the Hungarian regions, although these associations were not statistically significant. No correlations could be detected between these parameters among females. Conclusion: This analysis revealed considerable differences in the epidemiology of LC between the 7 main Hungarian regions. LC incidence and mortality rates significantly correlated with smoking and certain socioeconomic factors in men, but not in women. Further research is needed to explain the regional differences. C1 [Galffy, Gabriella] County Hospital of PulmonologyTorokbalint, Hungary. [Vastag, Aladar] MSD Pharma Hungary Kft.Budapest, Hungary. [Bogos, Krisztina] National Koranyi Institute for TB and Pulmonology, Department of PulmonologyBudapest, Hungary. [Kiss, Zoltan] MSD Pharma Hungary Kft.Budapest, Hungary. [Ostoros, Gyula] County Hospital of PulmonologyTorokbalint, Hungary. [Muller, Veronika] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Urban, Laszlo] Matrahaza Healthcare Center and University Teaching Hospital, Department of PulmonologyMatrahaza, Hungary. [Bittner, Nora] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Sarosi, Veronika] University of Pecs, Faculty of Medicine, Department of PulmonologyPecs, Hungary. [Polanyi, Zoltan] MSD Pharma Hungary Kft.Budapest, Hungary. [Nagy-Erdei, Zsofia] MSD Pharma Hungary Kft.Budapest, Hungary. [Daniel, Andrea] MSD Pharma Hungary Kft.Budapest, Hungary. [Knollmajer, Kata] MSD Pharma Hungary Kft.Budapest, Hungary. [Varnai, Mate] MSD Pharma Hungary Kft.Budapest, Hungary. [Szegner, Peter] MSD Pharma Hungary Kft.Budapest, Hungary. [Voko, Zoltan] Eotvos Lorand University, Department of Health Policy and Health EconomicsBudapest, Hungary. [Nagy, Balazs] Eotvos Lorand University, Department of Health Policy and Health EconomicsBudapest, Hungary. [Horvath, Krisztian] Eotvos Lorand University, Department of Health Policy and Health EconomicsBudapest, Hungary. [Rokszin, Gyorgy] RxTarget LtdSzolnok, Hungary. [Abonyi-Toth, Zsolt] RxTarget LtdSzolnok, Hungary. [Pozsgai, Eva] University of Pecs, Medical School, Department of Primary Health CarePecs, Hungary. [Barcza, Zsofia] Syntesia LtdBudapest, Hungary. [Moldvay, Judit] National Koranyi Institute of Pulmonology, Department of Tumor BiologyBudapest, Hungary. [Tamasi, Lilla] Semmelweis University, Department of PulmonologyBudapest, Hungary. RP Kiss, Z (reprint author), MSD Pharma Hungary Kft., Budapest, Hungary. CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2018, 68(6): 394–424., DOI 10.3322/caac.21492 Ferlay J, Colombet M, Soerjomataram I, Dyba T, Randi G, Bettio M, et al. Cancer Incidence and Mortality Patterns in Europe: Estimates for 40 Countries and 25 Major Cancers in 2018. Eur J Cancer, 2018, 103:356–87., DOI 10.1016/ j.ejca.2018.07.005 Bogos K, Kiss Z, Galffy G, Tamasi L, Ostoros G, Muller V, et al. Revising Incidence and Mortality of Lung Cancer in Central Europe: An Epidemiology Review from Hungary. Front Oncol, 2019, 9:1051., DOI 10.3389/fonc.2019.01051 Loomis D, Grosse Y, Lauby-Secretan B, El Ghissassi F, Bouvard V, Benbrahim- Tallaa L, et al. The Carcinogenicity of Outdoor Air Pollution. Lancet Oncol, 2013, 14(13):1262–3., DOI 10.1016/s1470-2045(13)70487-x Jemal A, Center MM, DeSantis C, and Ward EM. Global Patterns of Cancer Incidence and Mortality Rates and Trends. Cancer Epidemiol Biomarkers Prev, 2010, 19(8):1893–907., DOI 10.1158/1055-9965.epi-10-0437 Hecht SS. Tobacco Smoke Carcinogens and Lung Cancer. J Natl Cancer Inst, 1999, 91(14):1194–210., DOI 10.1093/jnci/91.14.1194 Jung KJ, Jeon C, and Jee SH. The Effect of Smoking on Lung Cancer: Ethnic Differences and the Smoking Paradox. Epidemiol Health, 2016, 38:e2016060., DOI 10.4178/epih.e2016060 Wong MCS, Lao XQ, Ho K-F, Goggins WB, and Tse SLA. Incidence and Mortality of Lung Cancer: Global Trends and Association with Socioeconomic Status. Sci Rep, 2017, 7(1):14300., DOI 10.1038/s41598-017-14513-7 United Nations Development Programme, UNDP). Human Development Report 2013. The Rise of the South: Human Progress in a Diverse World. New York. Available at: http://hdr.undp.org/en/content/human-developmentreport- 2013, Accessed April 10, 2021). Fogleman AJ,Mueller GS, and JenkinsWD. Does where You Live Play an Important Role in Cancer Incidence in the U.S.? Am J Cancer Res, 2015, 5(7):2314–9. Ou JY, Fowler B, Ding Q, Kirchhoff AC, Pappas L, Boucher K, et al. A Statewide Investigation of Geographic Lung Cancer Incidence Patterns and Radon Exposure in a Low-Smoking Population. BMC Cancer, 2018, 18(1):115., DOI 10.1186/s12885-018-4002-9 Cheng TY, Cramb SM, Baade PD, Youlden DR, Nwogu C, and Reid ME. The International Epidemiology of Lung Cancer: Latest Trends, Disparities, and Tumor Characteristics. J Thorac Oncol, 2016, 11(10):1653–71., DOI 10.1016/ j.jtho.2016.05.021 Jemal A, Thun MJ, Ries LAG, Howe HL, Weir HK, Center MM, et al. Annual Report to the Nation on the Status of Cancer, 1975-2005, Featuring Trends in Lung Cancer, Tobacco Use, and Tobacco Control. J Natl Cancer Inst, 2008, 100(23):1672–94., DOI 10.1093/jnci/djn389 Youlden DR, Cramb SM, and Baade PD. The International Epidemiology of Lung Cancer: Geographical Distribution and Secular Trends. J Thorac Oncol, 2008, 3(8):819–31., DOI 10.1097/JTO.0b013e31818020eb Lortet-Tieulent J, Renteria E, Sharp L, Weiderpass E, Comber H, Baas P, et al. Convergence of Decreasing Male and Increasing Female Incidence Rates in Major Tobacco-Related Cancers in Europe in 1988-2010. Eur J Cancer, 2015, 51(9):1144–63., DOI 10.1016/j.ejca.2013.10.014 Parkin DM, Bray FI, and Devesa SS. Cancer burden in the Year 2000. The Global Picture. Eur J Cancer, 2001, 37(Suppl. 8):S4–66., DOI 10.1016/s0959- 8049(01)00267-2 Mitra D, ShawA, TjepkemaM, and Peters P. SocialDeterminants of LungCancer Incidence in Canada: A 13-year Prospective Study. Health Rep, 2015, 26(6):12–20. PolakM, Genowska A, Szafraniec K, Fryc J, Jamiolkowski J, and Pajak A.Area-Based Socio-Economic Inequalities in Mortality from Lung Cancer and Respiratory Diseases. Int J Environ Res Public Health, 2019, 16(10):1791., DOI 10.3390/ijerph16101791 Nolasco A, Moncho J, Quesada JA, Melchor I, Pereyra-Zamora P, Tamayo- Fonseca N, et al. Trends in Socioeconomic Inequalities in Preventable Mortality in Urban Areas of 33 Spanish Cities, 1996-2007, MEDEA Project). Int J Equity Health, 2015, 14:33., DOI 10.1186/s12939-015-0164-0 Huang C-Y, Au K-K, Chen S-L, Wang S-C, Liao C-Y, Hsu H-H, et al. Unfavorable Mortality-To-Incidence Ratio of Lung Cancer Is Associated with Health Care Disparity. Int J Environ Res Public Health, 2018, 15(12): 2889., DOI 10.3390/ijerph15122889 Podralska M, Dzikiewicz-Krawczyk A, Mosor M, Zurawek M, Izykowska K, Slomski R, et al. The Most Frequent Polish ATM Mutations Are Not Susceptibility Factors for Tobacco-Related Cancers. Arch Med Sci, 2020, 17:1158., DOI 10.5114/aoms.2020.94155 Piatek L, Wilczek K, Kurzawski J, Gierlotka M, Gasior M, Polonski L, et al. Gender-related Disparities in the Treatment and Outcomes in Patients with Non-ST-segment Elevation Myocardial Infarction: Results from the Polish Registry of Acute Coronary Syndromes, PL-ACS, in the Years 2012-2014. Arch Med Sci, 2020, 16(4):781–8., DOI 10.5114/aoms.2018.76112 Koczkodaj P, Sulkowska U, Gotlib J, and Manczuk M. Breast Cancer Mortality Trends in Europe Among Women in Perimenopausal and Postmenopausal Age, 45+). Arch Med Sci, 2019, 16(1):146–56., DOI 10.5114/aoms.2019.85198 Fucito LM, Czabafy S, Hendricks PS, Kotsen C, Richardson D, and Toll BA. Pairing Smoking-Cessation Services with Lung Cancer Screening: A Clinical Guideline from the Association for the Treatment of Tobacco Use and Dependence and the Society for Research on Nicotine and Tobacco. Cancer, 2016, 122(8):1150–9., DOI 10.1002/cncr.29926 Huang C-Y, Au K-K, Chen S-L, Wang S-C, Liao C-Y, Hsu H-H, et al. Unfavorable Mortality-To-Incidence Ratio of Lung Cancer Is Associated with Health Care Disparity. Int J Environ Res Public Health, 2018, 15(12): 2889., DOI 10.3390/ijerph15122889 Raaschou-Nielsen O, Andersen ZJ, Beelen R, Samoli E, Stafoggia M,Weinmayr G, et al.Air Pollution and Lung Cancer Incidence in 17 European Cohorts: Prospective Analyses from the European Study of Cohorts for Air Pollution Effects, ESCAPE). Lancet Oncol, 2013, 14(9):813–22., DOI 10.1016/s1470-2045(13)70279-1 YueS,WangY,Wang J,andChenJ.Relationships between Lung Cancer Incidences and Air Pollutants. Technol Health Care, 2017, 25(S1):411–22., DOI 10.3233/thc-171344 Kulhanova I, Morelli X, Le Tertre A, Loomis D, Charbotel B, Medina S, et al. The Fraction of Lung Cancer Incidence Attributable to fine Particulate Air Pollution in France: Impact of Spatial Resolution of Air Pollution Models. Environ Int, 2018, 121(Pt 2):1079–86., DOI 10.1016/j.envint.2018.09.055 Zhang Y, Zhang R, and Ni H. Eriodictyol Exerts Potent Anticancer Activity against A549 Human Lung Cancer Cell Line by Inducing Mitochondrial- Mediated Apoptosis, G2/M Cell Cycle Arrest and Inhibition of M-TOR/PI3K/ Akt Signalling Pathway. Arch Med Sci, 2019, 16(2):446–52., DOI 10.5114/ aoms.2019.85152 Xu R, and Han Y. Long Non-coding RNA FOXF1 Adjacent Non-coding Developmental Regulatory RNA Inhibits Growth and Chemotherapy Resistance in Non-small Cell Lung Cancer. Arch Med Sci, 2019, 15(6): 1539–46., DOI 10.5114/aoms.2019.86707 Huang H-T, Xu Y-M, Ding S-G, Yu X-Q,Wang F,Wang H-F, et al. The Novel lncRNA PTTG3P Is Downregulated and Predicts Poor Prognosis in Non-small Cell Lung Cancer. Arch Med Sci, 2020, 16(4):931–40., DOI 10.5114/ aoms.2020.93535 Uzzoli A. Health Inequalities Regarding Territorial Differences in Hungary by Discussing Life Expectancy. Reg Stat, 2016, 6(1):139–63. Available at: https:// www.ksh.hu/docs/hun/xftp/terstat/2016/rs06108.pdf, Accessed May 15, 2021). Hungarian Central Statistical Office, CSO). [Tarsadalmi Helyzetkep 2010 – Egeszsegi Allapot, Egeszsegugy]. Available at: http://www.ksh.hu/docs/hun/ xftp/idoszaki/thk/thk10_egeszseg.pdf, Accessed May 15, 2021). Molnar T, and M Barna K. [Demografiai Jellemzok Magyarorszagon Es Az Europai Unioban, Ku€lo€no€s Tekintettel a Daganatos Megbetegedesek Okozta Halalozasra]. Hungarian Central Statistical Office, CSO). Available at: https:// www.ksh.hu/statszemle_archive/2012/2012_06/2012_06_544.pdf, Accessed May 15, 2021). Paldy A, Pinter A, Nador G, Vincze I, and Malnasi T. Regional Differences of Mortality from Malignancies in Hungary. Orv Hetil, 2003, 144(25): 1227–33. Eurostat. Health Statistics at Regional Level 2016. Available at: https://ec. europa.eu/eurostat/statistics-explained/index.php?titleHealth_statistics_at_ regional_level&oldid189405, Accessed April 18, 2021). Adany R, and Sandor J. Health Behaviour and Health Status of the Roma Population Living in Segregated Colonies in North-East Hungary. Magyar Tudomany, 2019, 180(11):1596–611., DOI 10.1556/2065.180.2019.11.2 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609916 EP 1609926 DI 10.3389/pore.2021.1609916 PG 11 ER PT J TI Retraction: Effects of microRNA-708 on Epithelial-Mesenchymal Transition, Cell Proliferation and Apoptosis in Melanoma Cells by Targeting LEF1 Through the Wnt Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Retraction AB Song, X., Wang, Q. and Huo, R. Effects of microRNA-708 on Epithelial-Mesenchymal Transition, Cell Proliferation and Apoptosis in Melanoma Cells by Targeting LEF1 Through the Wnt Signaling Pathway. Pathol. Oncol. Res. 25, 377–389 (2019). doi: https://doi.org/10.1007/s12253-017-0334-z NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609917 EP 1609917 DI 10.3389/pore.2021.1609917 PG 1 ER PT J AU Radeczky, P Moldvay, J Fillinger, J Szeitz, B Ferencz, B Boettiger, K Rezeli, M Bogos, K Renyi-Vamos, F Hoetzenecker, K Hegedus, B Megyesfalvi, Zs Dome, B AF Radeczky, Peter Moldvay, Judit Fillinger, Janos Szeitz, Beata Ferencz, Bence Boettiger, Kristiina Rezeli, Melinda Bogos, Krisztina Renyi-Vamos, Ferenc Hoetzenecker, Konrad Hegedus, Balazs Megyesfalvi, Zsolt Dome, Balazs TI Bone-Specific Metastasis Pattern of Advanced-Stage Lung Adenocarcinoma According to the Localization of the Primary Tumor SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lung cancer; adenocarcinoma; metastasis pattern; bone metastasis; tumor localization ID lung cancer; adenocarcinoma; metastasis pattern; bone metastasis; tumor localization AB Background: Patients with advanced-stage lung adenocarcinoma (LADC) often develop distant metastases in the skeletal system. Yet, the bone-specific metastasis pattern is still controversial. We, therefore, aimed to examine how the primary tumor location affects bone specificity and survival in LADC patients diagnosed with skeletal metastases. Methods: In total, 209 bone-metastatic Caucasian LADC patients from two thoracic centers were included in this study. Focusing on the specific location of primary tumors and bone metastatic sites, clinicopathological variables were included in a common database and analyzed retrospectively. Skeletal metastases were diagnosed according to the contemporary diagnostic guidelines and confirmed by bone scintigraphy. Besides region- and side-specific localization, primary tumors were also classified as central or peripheral tumors based on their bronchoscopic visibility. Results: The most common sites for metastasis were the spine (n = 103) and the ribs (n = 60), followed by the pelvis (n = 36) and the femur (n = 22). Importantly, femoral (p = 0.022) and rib (p = 0.012) metastases were more frequently associated with peripheral tumors, whereas centrally located LADCs were associated with humeral metastases (p = 0.018). Moreover, we deduced that left-sided tumors give rise to skull metastases more often than right-sided primary tumors (p = 0.018). Of note, however, the localization of the primary tumor did not significantly influence the type of affected bones. Multivariate Cox regression analysis adjusted for clinical parameters demonstrated that central localization of the primary tumor was an independent negative prognostic factor for overall survival (OS). Additionally, as expected, both chemotherapy and bisphosphonate therapy conferred a significant benefit for OS. Conclusion: The present study demonstrates unique bone-specific metastasis patterns concerning primary tumor location. Peripherally located LADCs are associated with rib and femoral metastases and improved survival outcomes. Our findings might contribute to the development of individualized follow-up strategies in bone-metastatic LADC patients and warrant further clinical investigations on a larger sample size. C1 [Radeczky, Peter] Orszagos Onkologiai Intezet, Mellkassebeszeti OsztalyBudapest, Hungary. [Moldvay, Judit] Hungarian Academy of Sciences - Semmelweis University, MTA-SE NAP, Brain Metastasis Research GroupBudapest, Hungary. [Fillinger, Janos] National Koranyi Institute of PulmonologyBudapest, Hungary. [Szeitz, Beata] Semmelweis University, 1st Department of Internal MedicineBudapest, Hungary. [Ferencz, Bence] Orszagos Onkologiai Intezet, Mellkassebeszeti OsztalyBudapest, Hungary. [Boettiger, Kristiina] Medical University of Vienna, Department of Thoracic SurgeryVienna, Austria. [Rezeli, Melinda] Lund University, Department of Biomedical EngineeringLund, Sweden. [Bogos, Krisztina] National Koranyi Institute of PulmonologyBudapest, Hungary. [Renyi-Vamos, Ferenc] Orszagos Onkologiai Intezet, Mellkassebeszeti OsztalyBudapest, Hungary. [Hoetzenecker, Konrad] Medical University of Vienna, Department of Thoracic SurgeryVienna, Austria. [Hegedus, Balazs] University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Megyesfalvi, Zsolt] Orszagos Onkologiai Intezet, Mellkassebeszeti OsztalyBudapest, Hungary. [Dome, Balazs] Orszagos Onkologiai Intezet, Mellkassebeszeti OsztalyBudapest, Hungary. RP Dome, B (reprint author), Orszagos Onkologiai Intezet, Mellkassebeszeti Osztaly, Budapest, Hungary. EM balazs.dome@meduniwien.ac.at CR Rinaldi S, and Berardi R Lung Cancer Prognosis: Can Histological Patterns and Morphological Features Have a Role in the Management of Lung Cancer Patients? Ann Transl Med, 2017, 5(17):353., DOI 10.21037/ atm.2017.05.18 Zappa C, and Mousa SA Non-small Cell Lung Cancer: Current Treatment and Future Advances. Transl Lung Cancer Res, 2016, 5(3):288–300., DOI 10.21037/ tlcr.2016.06.07 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Jones GS, and Baldwin DR Recent Advances in the Management of Lung Cancer. Clin Med, Lond,, 2018, 18(Suppl. 2):s41–s6., DOI 10.7861/ clinmedicine.18-2-s41 Zhou Y, Yu Q-F, Peng A-F, Tong W-L, Liu J-M, and Liu Z-L The Risk Factors of Bone Metastases in Patients with Lung Cancer. Sci Rep, 2017, 7(1):8970., DOI 10.1038/s41598-017-09650-y D’Antonio C, Passaro A, Gori B, Del Signore E, Migliorino MR, Ricciardi S, et al. Bone and Brain Metastasis in Lung Cancer: Recent Advances in Therapeutic Strategies. Ther Adv Med Oncol, 2014, 6(3):101–14., DOI 10.1177/1758834014521110 Klikovits T, Lohinai Z, Fabian K, Gyulai M, Szilasi M, Varga J, et al. New Insights into the Impact of Primary Lung Adenocarcinoma Location on Metastatic Sites and Sequence: A Multicenter Cohort Study. Lung Cancer, 2018, 126:139–48., DOI 10.1016/j.lungcan.2018.11.004 Niu Y-J, Wen Y-T, Shen W-W, Deng L, Liu L-L, and Zhang H-L Risk Factors for Bone Metastasis in Patients with Primary Lung Cancer: Study Protocol for a Systematic Review. BMJ Open, 2014, 4(7):e005202., DOI 10.1136/bmjopen- 2014-005202 Sugiura H, Yamada K, Sugiura T, Hida T, and Mitsudomi T Predictors of Survival in Patients with Bone Metastasis of Lung Cancer. Clin Orthop Relat Res, 2008, 466(3):729–36., DOI 10.1007/s11999-007-0051-0 Oliveira MBd. R, Mello FCd. Q, and Paschoal MEM The Relationship between Lung Cancer Histology and the Clinicopathological Characteristics of Bone Metastases. Lung Cancer, 2016, 96:19–24., DOI 10.1016/j.lungcan.2016.03.014 Gralow J, and Tripathy D Managing Metastatic Bone Pain: The Role of Bisphosphonates. J Pain Symptom Manage, 2007, 33(4):462–72., DOI 10.1016/j.jpainsymman.2007.01.001 Langer C, and Hirsh V Skeletal Morbidity in Lung Cancer Patients with Bone Metastases: Demonstrating the Need for Early Diagnosis and Treatment with Bisphosphonates. Lung Cancer, 2010, 67(1):4–11., DOI 10.1016/ j.lungcan.2009.08.020 Kuchuk M, Addison CL, Clemons M, Kuchuk I, and Wheatley-Price P Incidence and Consequences of Bone Metastases in Lung Cancer Patients. J Bone Oncol, 2013, 2(1):22–9., DOI 10.1016/j.jbo.2012.12.004 Al Husaini H, Wheatley-Price P, Clemons M, and Shepherd FA Prevention and Management of Bone Metastases in Lung Cancer: a Review. J Thorac Oncol, 2009, 4(2):251–9., DOI 10.1097/jto.0b013e31819518fc Felice FD, Piccioli A, Musio D, and Tombolini V The Role of Radiation Therapy in Bone Metastases Management. Oncotarget, 2017, 8(15):25691–9., DOI 10.18632/oncotarget.14823 Fairchild A Palliative Radiotherapy for Bone Metastases from Lung Cancer: Evidence-Based Medicine? Wjco, 2014, 5(5):845–57., DOI 10.5306/ wjco.v5.i5.845 Radeczky P, Megyesfalvi Z, Laszlo V, Fillinger J, Moldvay J, Raso E, et al. The Effects of Bisphosphonate and Radiation Therapy in Bone-Metastatic Lung Adenocarcinoma: the Impact of KRAS Mutation. Transl Lung Cancer Res, 2021, 10(2):675–84., DOI 10.21037/tlcr-20-754 Ketchedjian A, Daly BDT, Fernando HC, Florin L, Hunter CJ, Morelli DM, et al. Location as an Important Predictor of Lymph Node Involvement for Pulmonary Adenocarcinoma. J Thorac Cardiovasc Surg, 2006, 132(3):544–8., DOI 10.1016/j.jtcvs.2006.05.023 Coleman R, Hadji P, Body J-J, Santini D, Chow E, Terpos E, et al. Bone Health in Cancer: ESMO Clinical Practice Guidelines. Ann Oncol, 2020, 31(12): 1650–63., DOI 10.1016/j.annonc.2020.07.019 Institute NC Classification of Bones 2021. Available from: https://training.seer. cancer.gov/anatomy/skeletal/classification.html, Accessed May 10, 2021). Ettinger DS, Wood DE, Aggarwal C, Aisner DL, AkerleyW, Bauman JR, et al. NCCN Guidelines Insights: Non-small Cell Lung Cancer, Version 1.2020. J Natl Compr Canc Netw, 2019, 17(12):1464–72., DOI 10.6004/jnccn.2019.0059 Cho YJ, Cho YM, Kim SH, Shin K-H, Jung S-T, and Kim HS Clinical Analysis of Patients with Skeletal Metastasis of Lung Cancer. BMC Cancer, 2019, 19(1): 303., DOI 10.1186/s12885-019-5534-3 Tsuya A, Kurata T, Tamura K, and Fukuoka M Skeletal Metastases in Nonsmall Cell Lung Cancer: a Retrospective Study. Lung Cancer, 2007, 57(2): 229–32., DOI 10.1016/j.lungcan.2007.03.013 Amelot A, Terrier L-M, Cristini J, Buffenoir K, Pascal-Moussellard H, Carpentier A, et al. Spinal Metastases from Lung Cancer: Survival Depends Only on Genotype, Neurological and Personal Status, Scarcely of Surgical Resection. Surg Oncol, 2020, 34:51–6., DOI 10.1016/j.suronc.2020.03.005 Dougall B The RANK/RANKL/OPG axis in Bone Metastasis. Cancer Res, 2005, 65(9 Suppl. ment):1478. Ziu E, Viswanathan KV, and Mesfin FB Spinal Metastasis. Treasure Island, FL: StatPearls Publishing, 2020). Batson OV THE FUNCTION OF THE VERTEBRAL VEINS AND THEIR ROLE IN THE SPREAD OF METASTASES. Ann Surg, 1940, 112(1):138–49., DOI 10.1097/00000658-194007000-00016 Mujoomdar A, Austin JHM, Malhotra R, Powell CA, Pearson GDN, Shiau MC, et al. Clinical Predictors of Metastatic Disease to the Brain from Nonsmall Cell Lung Carcinoma: Primary Tumor Size, Cell Type, and Lymph Node Metastases. Radiology, 2007, 242(3):882–8., DOI 10.1148/ radiol.2423051707 Fabian K, Gyulai M, Furak J, Varallyay P, Jackel M, Bogos K, et al. Significance of Primary Tumor Location and Histology for Brain Metastasis Development and Peritumoral Brain Edema in Lung Cancer. Oncology, 2016, 91(5):237–42., DOI 10.1159/000447517 Megyesfalvi Z, Tallosy B, Pipek O, Fillinger J, Lang C, Klikovits T, et al. The Landscape of Small Cell Lung Cancer Metastases: Organ Specificity and Timing. Thorac Cancer, 2021, 12(6):914–23., DOI 10.1111/1759- 7714.13854 Hansen IO, and Jess P Possible Better Long-Term Survival in Left versus Right-Sided colon Cancer - a Systematic Review. Dan Med J, 2012, 59(6): A4444. Snaebjornsson P, Jonasson L, Jonsson T, Moller PH, Theodors A, and Jonasson JG Colon Cancer in Iceland-A Nationwide Comparative Study on Various Pathology Parameters with Respect to Right and Left Tumor Location and Patients Age. Int J Cancer, 2010, 127(11):2645–53., DOI 10.1002/ijc.25258 Suttie SA, Shaikh I,Mullen R, Amin AI, Daniel T, and Yalamarthi S Outcome of Right- and Left-Sided Colonic and Rectal Cancer Following Surgical Resection. Colorectal Dis, 2011, 13(8):884–9., DOI 10.1111/j.1463-1318.2010.02356.x Ling Q, Xu X, Ye P, Xie H, Gao F, Hu Q, et al. The Prognostic Relevance of Primary Tumor Location in Patients Undergoing Resection for Pancreatic Ductal Adenocarcinoma. Oncotarget, 2017, 8(9):15159–67., DOI 10.18632/ oncotarget.14768 Amri R, Bordeianou LG, Sylla P, and Berger DL Variations in Metastasis Site by Primary Location in Colon Cancer. J Gastrointest Surg, 2015, 19(8):1522–7., DOI 10.1007/s11605-015-2837-9 QiuM, Hu J, Yang D, Cosgrove DP, and Xu R Pattern of Distant Metastases in Colorectal Cancer: a SEER Based Study. Oncotarget, 2015, 6(36):38658–66., DOI 10.18632/oncotarget.6130 Schmid-Alliana A, Schmid-Antomarchi H, Al-Sahlanee R, Lagadec P, Scimeca J-C, and Verron E Understanding the Progression of Bone Metastases to Identify Novel Therapeutic Targets. Ijms, 2018, 19(1):148., DOI 10.3390/ ijms19010148 Langley RR, and Fidler IJ The Seed and Soil Hypothesis Revisited-The Role of Tumor-Stroma Interactions in Metastasis to Different Organs. Int J Cancer, 2011, 128(11):2527–35., DOI 10.1002/ijc.26031 Clay TD, Do H, Sundararajan V, Moore MM, Conron M, Wright GM, et al. The Clinical Relevance of Pathologic Subtypes in Metastatic Lung Adenocarcinoma. J Thorac Oncol, 2014, 9(5):654–63., DOI 10.1097/ jto.0000000000000150 Sahin S, and Karatas F The Impact of Primary Tumor Localization on Survival and Treatment Outcomes in Patients with Metastatic Colorectal Cancer-A Multicenter Study. J Buon, 2019, 24(2):479–87. Sakin A, Arici S, Secmeler S, Can O, Geredeli C, Yasar N, et al. Prognostic Significance of Primary Tumor Localization in Stage II and III colon Cancer. Wjgo, 2018, 10(11):410–20., DOI 10.4251/wjgo.v10.i11.410 Sohn VY, Arthurs ZM, Sebesta JA, and Brown TA Primary Tumor Location Impacts Breast Cancer Survival. Am J Surg, 2008, 195(5):641–4., DOI 10.1016/ j.amjsurg.2007.12.039 Sun W, Yang X, Liu Y, Yuan Y, and Lin D Primary Tumor Location Is a Useful Predictor for Lymph Node Metastasis and Prognosis in Lung Adenocarcinoma. Clin Lung Cancer, 2017, 18(1):e49–e55., DOI 10.1016/ j.cllc.2016.06.002 Moon Y, Lee KY, Sung SW, and Park JK Differing Histopathology and Prognosis in Pulmonary Adenocarcinoma at central and Peripheral Locations. J Thorac Dis, 2016, 8(1):169–77., DOI 10.3978/j.issn.2072- 1439.2016.01.15 Jia B, Zheng Q, Qi X, Zhao J, Wu M, An T, et al. Survival Comparison of Right and Left Side Non-small Cell Lung Cancer in Stage I-IIIA Patients: A Surveillance Epidemiology and End Results, SEER, Analysis. Thorac Cancer, 2019, 10(3):459–71., DOI 10.1111/1759-7714.12959 Puri V, Garg N, Engelhardt EE, Kreisel D, Crabtree TD, Meyers BF, et al. Tumor Location Is Not an Independent Prognostic Factor in Early Stage Nonsmall Cell Lung Cancer. Ann Thorac Surg, 2010, 89(4):1053–9., DOI 10.1016/ j.athoracsur.2010.01.020 Whitson BA, Groth SS, Andrade RS, Habermann EB, Maddaus MA, and D’Cunha J T1/T2 Non-small-cell Lung Cancer Treated by Lobectomy: Does Tumor Anatomic Location Matter? J Surg Res, 2012, 177(2):185–90., DOI 10.1016/j.jss.2012.05.022 Mitsuya K, Nakasu Y, Horiguchi S, Harada H, Nishimura T, Yuen S, et al. Metastatic Skull Tumors: MRI Features and a New Conventional Classification. J Neurooncol, 2011, 104(1):239–45., DOI 10.1007/s11060-010- 0465-5 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609926 EP 1609935 DI 10.3389/pore.2021.1609926 PG 10 ER PT J AU Khatun, M Urpilainen, E Ahtikoski, A Arffman, R Pasanen, A Puistola, U Tapanainen, J Andersson, L Butzow, R Loukovaara, M Piltonen, T AF Khatun, Masuma Urpilainen, Elina Ahtikoski, Anne Arffman, K. Riikka Pasanen, Annukka Puistola, Ulla Tapanainen, S. Juha Andersson, C. Leif Butzow, Ralf Loukovaara, Mikko Piltonen, T. Terhi TI Low Expression of Stanniocalcin 1 (STC-1) Protein Is Associated With Poor Clinicopathologic Features of Endometrial Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE stanniocalcin-1; uterine cancer; type 2 diabetes mellitus; disease-specific survival; endometrioid carcinoma; metformin ID stanniocalcin-1; uterine cancer; type 2 diabetes mellitus; disease-specific survival; endometrioid carcinoma; metformin AB Stanniocalcin-1 (STC-1) is a glycoprotein hormone involved in diverse biological processes, including regulation of calcium phosphate homeostasis, cell proliferation, apoptosis, inflammation, oxidative stress responses, and cancer development. The role of STC-1 in endometrial cancer (EC) is yet to be elucidated. In this study, we investigated the protein expression pattern of STC-1 in a tissue microarray (TMA) cohort of hysterectomy specimens from 832 patients with EC. We then evaluated the prognostic value of STC-1 expression regarding the clinicopathologic features and patients survival over a period of 140 months. Our results revealed that in EC tissue samples, STC-1 is mainly localized in the endometrial epithelium, although some expression was also observed in the stroma. Decreased STC-1 expression was associated with factors relating to a worse prognosis, such as grade 3 endometrioid tumors (p = 0.030), deep myometrial invasion (p = 0.003), lymphovascular space invasion (p = 0.050), and large tumor size (p = 0.001). Moreover, STC-1 expression was decreased in tumors obtained from obese women (p = 0.014) and in women with diabetes mellitus type 2 (DMT2; p = 0.001). Interestingly, the data also showed an association between DNA mismatch repair (MMR) deficiency and weak STC-1 expression, specifically in the endometrial epithelium (p = 0.048). No association was observed between STC-1 expression and disease-specific survival. As STC-1 expression was particularly low in cases with obesity and DMT2 in the TMA cohort, we also evaluated the correlation between metformin use and STC-1 expression in an additional EC cohort that only included women with DMT2 (n = 111). The analysis showed no difference in STC-1 expression in either the epithelium or the stroma in women undergoing metformin therapy compared to metformin non-users. Overall, our data may suggest a favorable role for STC- 1 in EC behavior; however, further studies are required to elucidate the detailed mechanism and possible applications to cancer treatment. C1 [Khatun, Masuma] University of Oulu, Medical Research Center, Oulu University Hospital, Department of Obstetrics and Gynaecology, PEDEGO Research UnitOulu, Finland. [Urpilainen, Elina] University of Oulu, Medical Research Center, Oulu University Hospital, Department of Obstetrics and Gynaecology, PEDEGO Research UnitOulu, Finland. [Ahtikoski, Anne] University of Oulu, Department of PathologyOulu, Finland. [Arffman, K. Riikka] University of Oulu, Medical Research Center, Oulu University Hospital, Department of Obstetrics and Gynaecology, PEDEGO Research UnitOulu, Finland. [Pasanen, Annukka] Helsinki University Central Hospital, Department of PathologyHelsinki, Finland. [Puistola, Ulla] University of Oulu, Medical Research Center, Oulu University Hospital, Department of Obstetrics and Gynaecology, PEDEGO Research UnitOulu, Finland. [Tapanainen, S. Juha] University of Oulu, Medical Research Center, Oulu University Hospital, Department of Obstetrics and Gynaecology, PEDEGO Research UnitOulu, Finland. [Andersson, C. Leif] Helsinki University Central Hospital, Department of PathologyHelsinki, Finland. [Butzow, Ralf] Helsinki University Central Hospital, Department of PathologyHelsinki, Finland. [Loukovaara, Mikko] Helsinki University Hospital and University of Helsinki, Department of Obstetrics and GynaecologyHelsinki, Finland. [Piltonen, T. Terhi] University of Oulu, Medical Research Center, Oulu University Hospital, Department of Obstetrics and Gynaecology, PEDEGO Research UnitOulu, Finland. RP Piltonen, T (reprint author), University of Oulu, Medical Research Center, Oulu University Hospital, Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Oulu, Finland. CR Brooks RA, Fleming GF, Lastra RR, Lee NK, Moroney JW, Son CH, et al. Current Recommendations and Recent Progress in Endometrial Cancer. CA Cancer J Clin, 2019, 69(4):258–79., DOI 10.3322/caac.21561 Raglan O, Kalliala I, Markozannes G, Cividini S, Gunter MJ, Nautiyal J, et al. Risk Factors for Endometrial Cancer: An Umbrella Review of the Literature. Int J Cancer, 2019, 145(7):1719–30., DOI 10.1002/ijc.31961 Sorosky JI Endometrial Cancer. Obstet Gynecol, 2012, 120(2 Pt 1):383–97., DOI 10.1097/AOG.0b013e3182605bf1 Meyer LA, Broaddus RR, and Lu KH Endometrial Cancer and Lynch Syndrome: Clinical and Pathologic Considerations. Cancer Control, 2009, 16(1):14–22., DOI 10.1177/107327480901600103 Ryan NAJ, Glaire MA, Blake D, Cabrera-Dandy M, Evans DG, and Crosbie EJ The Proportion of Endometrial Cancers Associated with Lynch Syndrome: a Systematic Review of the Literature and Meta-Analysis. Genet Med, 2019, 21(10):2167–80., DOI 10.1038/s41436-019-0536-8 Deol HK, Varghese R, Wagner GF, and Dimattia GE Dynamic Regulation of Mouse Ovarian Stanniocalcin Expression during Gestation and Lactation*. Endocrinology, 2000, 141(9):3412–21., DOI 10.1210/endo.141.9.7658 Yeung BHY, Law AYS, andWong CKC Evolution and Roles of Stanniocalcin. Mol Cell Endocrinol, 2012, 349(2):272–80., DOI 10.1016/j.mce.2011.11.007 Juhanson P, Rull K, Kikas T, Laivuori H, Vaas P, Kajantie E, et al. Stanniocalcin-1 Hormone in Nonpreeclamptic and Preeclamptic Pregnancy: Clinical, Life-Style, and Genetic Modulators. J Clin Endocrinol Metab, 2016, 101(12):4799–807., DOI 10.1210/jc.2016-1873 Zhang K-z., Lindsberg PJ, Tatlisumak T, Kaste M, Olsen HS, and Andersson LC Stanniocalcin: A Molecular Guard of Neurons during Cerebral Ischemia. Proc Natl Acad Sci, 2000, 97(7):3637–42., DOI 10.1073/pnas.97.7.3637 Westberg JA, Serlachius M, Lankila P, and Andersson LC Hypoxic Preconditioning Induces Elevated Expression of Stanniocalcin-1 in the Heart. Am J Physiology-Heart Circulatory Physiol, 2007, 293(3): H1766–H1771., DOI 10.1152/ajpheart.00017.2007 Huang L, Belousova T, Chen M, DiMattia G, Liu D, and Sheikh-Hamad D Overexpression of Stanniocalcin-1 Inhibits Reactive Oxygen Species and Renal Ischemia/reperfusion Injury in Mice. Kidney Int, 2012, 82(8):867–77., DOI 10.1038/ki.2012.223 Pan JS-C, Huang L, Belousova T, Lu L, Yang Y, Reddel R, et al. Stanniocalcin-1 Inhibits Renal Ischemia/reperfusion Injury via an AMP-Activated Protein Kinase-dependent Pathway. Jasn, 2015, 26(2):364–78., DOI 10.1681/ asn.2013070703 Mohammadipoor A, Lee RH, Prockop DJ, and Bartosh TJ Stanniocalcin-1 Attenuates Ischemic Cardiac Injury and Response of Differentiating Monocytes/macrophages to Inflammatory Stimuli. Translational Res, 2016, 177:127–42., DOI 10.1016/j.trsl.2016.06.011 Sheikh-Hamad D Mammalian Stanniocalcin-1 Activates Mitochondrial Antioxidant Pathways: New Paradigms for Regulation of Macrophages and Endothelium. Am J Physiology-Renal Physiol, 2010, 298(2):F248–F254., DOI 10.1152/ajprenal.00260.2009 Chang AC-M, Jellinek DA, and Reddel RR Mammalian Stanniocalcins and Cancer. Endocr Relat Cancer, 2003, 10(3):359–73., DOI 10.1677/erc.0.0100359 Guo F, Li Y, Wang J, Li Y, Li Y, and Li G Stanniocalcin1, STC1, Inhibits Cell Proliferation and Invasion of Cervical Cancer Cells. PLoS One, 2013, 8(1): e53989., DOI 10.1371/journal.pone.0053989 Chen F, Zhang Z, and Pu F. Role of Stanniocalcin-1 in Breast Cancer Oncol Lett, 2019, 18(4):3946–53., DOI 10.3892/ol.2019.10777 Yang Y, Yin S, Li S, Chen Y, and Yang L Stanniocalcin 1 in Tumor Microenvironment Promotes Metastasis of Ovarian Cancer. Ott, 2019, 12: 2789–98., DOI 10.2147/ott.s196150 Zhao F, Yang G, Feng M, Cao Z, Liu Y, Qiu J, et al. Expression, Function and Clinical Application of Stanniocalcin-1 in Cancer. J Cell Mol Med, 2020, 24(14):7686–7696., DOI 10.1111/jcmm.15348 Giovannucci E, Harlan DM, Archer MC, Bergenstal RM, Gapstur SM, Habel LA, et al. Diabetes and Cancer: a Consensus Report. Diabetes Care, 2010, 33(7):1674–85., DOI 10.2337/dc10-0666 Menke A, Rust KF, Fradkin J, Cheng YJ, and Cowie CC Associations between Trends in Race/Ethnicity, Aging, and Body Mass Index with Diabetes Prevalence in the United States. Ann Intern Med, 2014, 161(5):328–35., DOI 10.7326/m14-0286 Liao C, Zhang D,Mungo C, Andrew Tompkins D, and Zeidan AM Is Diabetes Mellitus Associated with Increased Incidence and Disease-specificMortality in Endometrial Cancer? A Systematic Review and Meta-Analysis of Cohort Studies. Gynecol Oncol, 2014, 135(1):163–71., DOI 10.1016/j.ygyno.2014.07.095 Saed L, Varse F, Baradaran HR, Moradi Y, Khateri S, Friberg E, et al. The Effect of Diabetes on the Risk of Endometrial Cancer: an Updated a Systematic Review and Meta-Analysis. BMC Cancer, 2019, 19(1):527., DOI 10.1186/ s12885-019-5748-4 Chatterjee S, Khunti K, and Davies MJ Type 2 Diabetes. The Lancet, 2017, 389(10085):2239–51., DOI 10.1016/s0140-6736(17)30058-2 Flory J, and Lipska K Metformin in 2019. JAMA, 2019, 321(19):1926–7., DOI 10.1001/jama.2019.3805 Erdemoglu E, Guney M, Giray SG, Take G, and Mungan T Effects of Metformin on Mammalian Target of Rapamycin in a Mouse Model of Endometrial Hyperplasia. Eur J Obstet Gynecol Reprod Biol, 2009, 145(2): 195–9., DOI 10.1016/j.ejogrb.2009.04.034 Zhang Q, Celestino J, Schmandt R, McCampbell AS, Urbauer DL, Meyer LA, et al. Chemopreventive Effects of Metformin on Obesity-Associated Endometrial Proliferation. Am J Obstet Gynecol, 2013, 209(1):24., DOI 10.1016/j.ajog.2013.03.008 Cantrell LA, Zhou C, Mendivil A, Malloy KM, Gehrig PA, and Bae-Jump VL Metformin Is a Potent Inhibitor of Endometrial Cancer Cell Proliferation- Implications for a Novel Treatment Strategy. Gynecol Oncol, 2010, 116(1): 92–8., DOI 10.1016/j.ygyno.2009.09.024 Ezewuiro O, Grushko TA, Kocherginsky M, Habis M, Hurteau JA, Mills KA, et al. Association of Metformin Use with Outcomes in Advanced Endometrial Cancer Treated with Chemotherapy. PLoS One, 2016, 11(1):e0147145., DOI 10.1371/journal.pone.0147145 Tan B-X, Yao W-X, Ge J, Peng X-C, Du X-B, Zhang R, et al. Prognostic Influence of Metformin as First-Line Chemotherapy for Advanced Nonsmall Cell Lung Cancer in Patients with Type 2 Diabetes. Cancer, 2011, 117(22): 5103–11., DOI 10.1002/cncr.26151 Sivalingam VN, Kitson S, McVey R, Roberts C, Pemberton P, Gilmour K, et al. Measuring the Biological Effect of Presurgical Metformin Treatment in Endometrial Cancer. Br J Cancer, 2016, 114(3):281–9., DOI 10.1038/ bjc.2015.453 Kitson SJ, Maskell Z, Sivalingam VN, Allen JL, Ali S, Burns S, et al. PREsurgical Metformin in Uterine Malignancy, PREMIUM): a Multi-Center, Randomized Double-Blind, Placebo-Controlled Phase III Trial. Clin Cancer Res, 2019, 25(8):2424–32., DOI 10.1158/1078-0432.ccr-18-3339 Urpilainen E, Arima R, Karihtala P, Puistola U, and Ahtikoski A Metformin Associates with Aggressive Features of Endometrial Cancer inWomen with Type 2 Diabetes. Anticancer Res, 2021, 41(2):821–8., DOI 10.21873/anticanres.14834 Mokhtar NM, Ramzi NH, Yin-Ling W, Rose IM, Hatta Mohd Dali AZ, and Jamal R Laser Capture Microdissection with Genome-wide Expression Profiling Displayed Gene Expression Signatures in Endometrioid Endometrial Cancer. Cancer Invest, 2012, 30(2):156–64., DOI 10.3109/ 07357907.2011.633290 Lopez JJ, Jardin I, Cantonero Chamorro C, Duran ML, Tarancon Rubio MJ, Reyes Panadero M, et al. Involvement of Stanniocalcins in the Deregulation of Glycaemia in Obese Mice and Type 2 Diabetic Patients. J Cel Mol Med, 2018, 22(1):684–94., DOI 10.1111/jcmm.13355 Schein V, Kucharski LC, Guerreiro PMG, Martins TL, Morgado I, Power DM, et al. Stanniocalcin 1 Effects on the Renal Gluconeogenesis Pathway in Rat and Fish. Mol Cell Endocrinol, 2015, 414:1–8., DOI 10.1016/ j.mce.2015.07.010 Pasanen A, Tuomi T, Isola J, Staff S, Butzow R, and Loukovaara M L1 Cell Adhesion Molecule as a Predictor of Disease-specific Survival and Patterns of Relapse in Endometrial Cancer. Int J Gynecol Cancer, 2016, 26(8):1465–71., DOI 10.1097/igc.0000000000000801 Loukovaara M, Pasanen A, and Butzow RMismatch Repair Protein and MLH1 Methylation Status as Predictors of Response to Adjuvant Therapy in Endometrial Cancer. Cancer Med, 2021, 10(3):1034–42., DOI 10.1002/ cam4.3691 Morice P, Leary A, Creutzberg C, Abu-Rustum N, and Darai E Endometrial Cancer. The Lancet, 2016, 387(10023):1094–108., DOI 10.1016/s0140-6736(15, 00130-0 AlHilli MM, Podratz KC, Dowdy SC, Bakkum-Gamez JN,Weaver AL, McGree ME, et al. Preoperative Biopsy and Intraoperative Tumor Diameter Predict Lymph Node Dissemination in Endometrial Cancer. Gynecol Oncol, 2013, 128(2):294–9., DOI 10.1016/j.ygyno.2012.10.009 Pecorelli S Revised FIGO Staging for Carcinoma of the Vulva, Cervix, and Endometrium. Int J Gynecol Obstet, 2009, 105(2):103–4., DOI 10.1016/ j.ijgo.2009.02.012 Kansikas M, Kasela M, Kantelinen J, and Nystrom M Assessing How Reduced Expression Levels of the Mismatch Repair GenesMLH1,MSH2, andMSH6Affect Repair Efficiency. Hum Mutat, 2014, 35(9):1123–7., DOI 10.1002/humu.22605 Aghajanova L, Altmae S, Kasvandik S, Salumets A, Stavreus-Evers A, and Giudice LC Stanniocalcin-1 Expression in normal Human Endometrium and Dysregulation in Endometriosis. Fertil Sterility, 2016, 106(3):681–91., DOI 10.1016/j.fertnstert.2016.05.023 Khatun M, Arffman RK, Lavogina D, Kangasniemi M, Laru J, Ahtikoski A, et al. Women with Polycystic Ovary Syndrome Present with Altered Endometrial Expression of Stanniocalcin-1†. Biol Reprod, 2020, 102(2): 306–15., DOI 10.1093/biolre/ioz180 Rajaram M, Li J, Egeblad M, and Powers RS System-wide Analysis Reveals a Complex Network of Tumor-Fibroblast Interactions Involved in Tumorigenicity. Plos Genet, 2013, 9(9):e1003789., DOI 10.1371/ journal.pgen.1003789 Pastushenko I, and Blanpain C EMT Transition States during Tumor Progression and Metastasis. Trends Cell Biol, 2019, 29(3):212–26., DOI 10.1016/j.tcb.2018.12.001 Su J, Guo B, Zhang T, Wang K, Li X, and Liang G Stanniocalcin-1, a New Biomarker of Glioma Progression, Is Associated with Prognosis of Patients. Tumor Biol, 2015, 36(8):6333–9., DOI 10.1007/s13277-015-3319-0 Liu G, Yang G, Chang B, Mercado-Uribe I, Huang M, Zheng J, et al. Stanniocalcin 1 and Ovarian Tumorigenesis. J Natl Cancer Inst, 2010, 102(11):812–27., DOI 10.1093/jnci/djq127 Arigami T, Uenosono Y, Ishigami S, Hagihara T, Haraguchi N, Matsushita D, et al. Expression of Stanniocalcin 1 as a Potential Biomarker of Gastric Cancer. Oncology, 2012, 83(3):158–64., DOI 10.1159/000341390 Ding H, Wei M, Bao Y, Xiong X, and Yi W Prognostic Value of STC 1 Expression in Ovarian Cancer. Int J Clin Exp Med, 2019, 12(5):5433–5439. LuoW, Chen D, Wang H, and Hu J Stanniocalcin 1 Is a Prognostic Biomarker in Glioma. Oncol Lett, 2020, 20(3):2248–56., DOI 10.3892/ol.2020.11792 Tamura S, Oshima T, Yoshihara K, Kanazawa A, Yamada T, Inagaki D, et al. Clinical Significance of STC1 Gene Expression in Patients with Colorectal Cancer. Anticancer Res, 2011, 31(1):325–9. Zhou X, Hua L, Zhang W, Zhu M, Shi Q, Li F, et al. FRK Controls Migration and Invasion of Human Glioma Cells by Regulating JNK/c-Jun Signaling. J Neurooncol, 2012, 110(1):9–19., DOI 10.1007/s11060-012-0933-1 Han J, Jeon M, Shin I, and Kim S Elevated STC-1 Augments the Invasiveness of Triple-Negative Breast Cancer Cells through Activation of the JNK/c-Jun Signaling Pathway. Oncol Rep, 2016, 36(3):1764–71., DOI 10.3892/or.2016.4977 Chang M-C, Chen C-A, Chen P-J, Chiang Y-C, Chen Y-L, Mao T-L, et al. Mesothelin Enhances Invasion of Ovarian Cancer by Inducing MMP-7 through MAPK/ERK and JNK Pathways. Biochem J, 2012, 442(2):293–302., DOI 10.1042/bj20110282 Sarapio E, De Souza SK, Model JFA, Trapp M, and Da Silva RSM Stanniocalcin-1 and -2 Effects on Glucose and Lipid Metabolism in white Adipose Tissue from Fed and Fasted Rats. Can J Physiol Pharmacol, 2019, 97(10):916–23., DOI 10.1139/cjpp-2019-0023 Zhao J, Jiao Y, Song Y, Liu J, Li X, Zhang H, et al. Stanniocalcin 2 Ameliorates Hepatosteatosis through Activation of STAT3 Signaling. Front Physiol, 2018, 9:873., DOI 10.3389/fphys.2018.00873 Shirakawa M, Fujiwara Y, Sugita Y, Moon J-H, Takiguchi S, Nakajima K, et al. Assessment of Stanniocalcin-1 as a Prognostic Marker in Human Esophageal Squamous Cell Carcinoma. Oncol Rep, 2012, 27(4):940–6., DOI 10.3892/ or.2011.1607 Silverberg SG Problems in the Differential Diagnosis of Endometrial Hyperplasia and Carcinoma. Mod Pathol, 2000, 13(3):309–27., DOI 10.1038/ modpathol.3880053 Hussein YR, Broaddus R,Weigelt B, Levine DA, and Soslow RA The Genomic Heterogeneity of FIGO Grade 3 Endometrioid Carcinoma Impacts Diagnostic Accuracy and Reproducibility. Int J Gynecol Pathol, 2016, 35(1):16–24., DOI 10.1097/pgp.0000000000000212 Zhang K-z., Westberg JA, Paetau A, von Boguslawsky K, Lindsberg P, Erlander M, et al. High Expression of Stanniocalcin in Differentiated Brain Neurons. Am J Pathol, 1998, 153(2):439–45., DOI 10.1016/s0002-9440(10)65587-3 Serlachius M, and Andersson LC Upregulated Expression of Stanniocalcin-1 during Adipogenesis. Exp Cell Res, 2004, 296(2):256–64., DOI 10.1016/j.yexcr.2004.02.016 Serlachius M, Zhang K-z., and Andersson LC Stanniocalcin in Terminally Differentiated Mammalian Cells. Peptides, 2004, 25(10):1657–62., DOI 10.1016/ j.peptides.2004.03.031 Pan X, Jiang B, Liu J, Ding J, Li Y, Sun R, et al. STC1 Promotes Cell Apoptosis via NF-Κb Phospho-P65 Ser536 in Cervical Cancer Cells. Oncotarget, 2017, 8(28):46249–61., DOI 10.18632/oncotarget.17641 Yeung BHY, Shek FH, Lee NP, and Wong CKC Stanniocalcin-1 Reduces Tumor Size in Human Hepatocellular Carcinoma. PLoS One, 2015, 10(10): e0139977., DOI 10.1371/journal.pone.0139977 Leung CCT, and Wong CKC Characterization of Stanniocalcin-1 Expression in Macrophage Differentiation. Translational Oncol, 2021, 14(1):100881., DOI 10.1016/j.tranon.2020.100881 Joensuu K, Heikkila P, and Andersson LC Tumor Dormancy: Elevated Expression of Stanniocalcins in Late Relapsing Breast Cancer. Cancer Lett, 2008, 265(1):76–83., DOI 10.1016/j.canlet.2008.02.022 Onitilo AA, Engel JM, Glurich I, Stankowski RV, Williams GM, and Doi SA Diabetes and Cancer I: Risk, Survival, and Implications for Screening. Cancer Causes Control, 2012, 23(6):967–81., DOI 10.1007/s10552-012-9972-3 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609936 EP 1609944 DI 10.3389/pore.2021.1609936 PG 9 ER PT J AU Cerina, D Matkovic, V Katic, K Lovasic, BI Separovic, R Canjko, I Jaksic, B Frobe, A Plestina, S Bajic, Vrdoljak, E AF Cerina, Dora Matkovic, Visnja Katic, Kristina Lovasic, Belac Ingrid Separovic, Robert Canjko, Ivana Jaksic, Blanka Frobe, Ana Plestina, Stjepko Bajic, Zarko Vrdoljak, Eduard TI Precision Oncology in Metastatic Uterine Cancer; Croatian First-Year Experience of the Comprehensive Genomic Profiling in Everyday Clinical Practice SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE targeted therapy; genomic profiling; uterine cancer; precision oncology; mutation ID targeted therapy; genomic profiling; uterine cancer; precision oncology; mutation AB Comprehensive genomic profiling (CGP) is gradually becoming an inevitable part of the everyday oncology clinical practice. The interpretation and optimal implementation of the results is one of the hot topics of modern-day oncology. According to the recent findings, uterine cancer harbors a high level of gene alterations but is still insufficiently explored. The primary goal of this project was to assess the proportion of patients with targetable mutations. Also, the aim was to define and emphasize potential opportunities as well as the problems we have faced in the first year of testing on the national level. We performed a multicentric, retrospective, nested cross-sectional analysis on the total population of Croatian patients with advanced/metastatic uterine cancer where the tumor CGP was performed during 2020. CGP of the tumor tissue of 32 patients revealed clinically relevant genomic alterations (CRGA) in 27 patients (84%) with a median of 3 (IQR 1-4) CRGA per patient. The most common CRGAs were those of phosphatide-inositol-3 kinases (PIK3) in 22 patients (69%), with 13/22 (59%) of those patients harboring PIK3CA mutation. The next most common CGRAs were ARID1A and PTEN mutations in 13 (41%) and 11 (34%) patients, respectively. Microsatellite status was determined as stable in 21 patients (66%) and highly unstable in 10 patients (31%). A high tumor mutational burden (≥10Muts/Mb) was reported in 12 patients (38%). CGP analysis reported some kind of targeted therapy for 28 patients (88%). CGP determined clinically relevant genomic alterations in the significant majority of patients with metastatic uterine cancer, defining it as a rich ground for further positioning and development of precision oncology. C1 [Cerina, Dora] University of Split, School of Medicine, Clinical Hospital Center Split, Department of OncologySplit, Croatia. [Matkovic, Visnja] University Hospital Center Zagreb, Department of Gynecologic OncologyZagreb, Croatia. [Katic, Kristina] University Hospital Center Zagreb, Department of Gynecologic OncologyZagreb, Croatia. [Lovasic, Belac Ingrid] University Hospital Center Rijeka, Department of Radiotherapy and OncologyRijeka, Croatia. [Separovic, Robert] Sestre Milosrdnice University Hospital Center, University Hospital for Tumors, Department of Medical Oncology, Division of Medical OncologyZagreb, Croatia. [Canjko, Ivana] University Hospital Center Osijek, Department of Radiotherapy OncologyOsijek, Croatia. [Jaksic, Blanka] Sestre Milosrdnice University Hospital Center, Department of Oncology and Nuclear MedicineZagreb, Croatia. [Frobe, Ana] Sestre Milosrdnice University Hospital Center, Department of Oncology and Nuclear MedicineZagreb, Croatia. [Plestina, Stjepko] University Hospital Center Zagreb and Zagreb Medical School, Department of Medical OncologyZagreb, Croatia. [Bajic, Zarko] University Psychiatric Hospital “Sveti Ivan”, Research Unit “Dr. Mirko Grmek”Zagreb, Croatia. [Vrdoljak, Eduard] University of Split, School of Medicine, Clinical Hospital Center Split, Department of OncologySplit, Croatia. RP Vrdoljak, E (reprint author), University of Split, School of Medicine, Clinical Hospital Center Split, Department of Oncology, Split, Croatia. EM edo.vrdoljak@gmail.com CR Janne PA, Yang JC-H, Kim D-W, Planchard D, Ohe Y, Ramalingam SS, et al. AZD9291 in EGFR Inhibitor-Resistant Non-small-cell Lung Cancer.N Engl J Med, 2015, 372:1689–99. Accessed: August 8, 2021., DOI 10.1056/NEJMoa1411817 Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, et al. space missing throghout the references pls fix Combined BRAF and MEK Inhibition in Melanoma with BRAF V600 Mutations. N Engl J Med, 2012, 367:1694–703., DOI 10.1056/nejmoa1210093 Dagher R, Cohen M, Williams G, Rothmann M, Gobburu J, Robbie G, et al. Approval Summary: Imatinib Mesylate in the Treatment of Metastatic And/or Unresectable Malignant Gastrointestinal Stromal Tumors. Clin Cancer Res, 2002, 8:3034–8. Poveda A, Floquet A, Ledermann JA, Asher R, Penson RT, Oza AM, et al. Final Overall Survival, OS, Results from SOLO2/ENGOT-Ov21: A Phase III Trial Assessing Maintenance Olaparib in Patients, Pts, with Platinum-Sensitive, Relapsed Ovarian Cancer and a BRCA Mutation. Jco, 2020, 38:6002., DOI 10.1200/JCO.2020.38.15_suppl.6002 Ascierto PA, and Schadendorf D. Immunotherapy in Non-melanoma Skin Cancer: Updates and New Perspectives. Dic, 2019, 8:1–6., DOI 10.7573/ dic.212583 Muller M, Schouten RD, De Gooijer CJ, and Baas P. Pembrolizumab for the Treatment of Non-small Cell Lung Cancer. Expert Rev Anticancer Ther, 2017, 17:399–409., DOI 10.1080/14737140.2017.1311791 Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al. Pembrolizumab Plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med, 2019, 380:1116–27., DOI 10.1056/NEJMoa1816714 Balar AV, Castellano D, O’Donnell PH, Grivas P, Vuky J, Powles T, et al. Firstline Pembrolizumab in Cisplatin-Ineligible Patients with Locally Advanced and Unresectable or Metastatic Urothelial Cancer, KEYNOTE-052): a Multicentre, Single-Arm, Phase 2 Study. Lancet Oncol, 2017, 18:1483–92., DOI 10.1016/S1470-2045(17)30616-2 von Minckwitz G, Procter M, de Azambuja E, Zardavas D, Benyunes M, Viale G, et al. Adjuvant Pertuzumab and Trastuzumab in Early HER2-Positive Breast Cancer. N Engl J Med, 2017, 377:122–31., DOI 10.1056/NEJMoa1703643 Swain SM, Kim S-B, Cortes J, Ro J, Semiglazov V, Campone M, et al. Pertuzumab, Trastuzumab, and Docetaxel for HER2-Positive Metastatic Breast Cancer, CLEOPATRA Study): Overall Survival Results from a Randomised, Double-Blind, Placebo-Controlled, Phase 3 Study. Lancet Oncol, 2013, 14:461–71., DOI 10.1016/S1470-2045(13)70130-X Cremolini C, Loupakis F, Antoniotti C, LupiC, Sensi E, Lonardi S, et al. FOLFOXIRI Plus Bevacizumab versus FOLFIRI Plus Bevacizumab as First-Line Treatment of Patients with Metastatic Colorectal Cancer: Updated Overall Survival andMolecular Subgroup Analyses of the Open-Label, Phase 3 TRIBE Study. Lancet Oncol, 2015, 16:1306–15., DOI 10.1016/S1470-2045(15)00122-9 Lyons TG, and Ku GY. Systemic Therapy for Esophagogastric Cancer: Immune Checkpoint Inhibition. Chin Clin Oncol, 2017, 6:53., DOI 10.21037/cco.2017.09.03 Coleman RL, Brady MF, Herzog TJ, Sabbatini P, Armstrong DK, Walker JL, et al. Bevacizumab and Paclitaxel-Carboplatin Chemotherapy and Secondary Cytoreduction in Recurrent, Platinum-Sensitive Ovarian Cancer, NRG Oncology/Gynecologic Oncology Group Study GOG-0213): a Multicentre, Open-Label, Randomised, Phase 3 Trial. Lancet Oncol, 2017, 18:779–91., DOI 10.1016/S1470-2045(17)30279-6 Tewari KS, Sill MW, Long HJ, Penson RT, Huang H, Ramondetta LM, et al. Improved Survival with Bevacizumab in Advanced Cervical Cancer. N Engl J Med, 2014, 370:734–43., DOI 10.1056/NEJMoa1309748 Henley SJ, Miller JW, Dowling NF, Benard VB, and Richardson LC. Uterine Cancer Incidence and Mortality - United States, 1999-2016. MMWR Morb Mortal Wkly Rep, 2018, 67:1333–8., DOI 10.15585/mmwr.mm6748a1 Shaw E, Farris M, McNeil J, and Friedenreich C. Obesity and Endometrial Cancer. Recent Results Cancer Res, 2016, 208:107–36., DOI 10.1007/978-3-319- 42542-9_7 National Cancer Institute. Cancer Stat Facts: Uterine Cancer Surveillance, Epidemiol. End Results Progr, 2021). Available at: https://seer.cancer.gov/ statfacts/html/corp.html. Accessed: June 22, 2021. McMeekin DS, Filiaci VL, Thigpen JT, Gallion HH, Fleming GF, and Rodgers WH. The Relationship between Histology and Outcome in Advanced and Recurrent Endometrial Cancer Patients Participating in First-Line Chemotherapy Trials: A Gynecologic Oncology Group Study. Gynecol Oncol, 2007, 106:16–22., DOI 10.1016/j.ygyno.2007.04.032 Levine DA. Integrated Genomic Characterization of Endometrial Carcinoma. Nature, 2013, 497:67–73., DOI 10.1038/nature12113 Babic D, Plestina S, and Samarzija M. Preporuke Za Odabir Bolesnika/tumora Za SGP, 2021). Availableat: http://www.hrvatsko-onkolosko-drustvo.com/wpcontent/ uploads/2021/02/Preporuke-za-SGP_Izdanje-23.2.2021.pdf. Accessed: June 22, 2021. Croatian Institue of Public Health. Cancer Incidence in Croatia Croat. Natl. Cancer Regist. Bull, 2018). Availableat: https://www.hzjz.hr/wp-content/ uploads/2020/12/Bilten_2018_final.pdf Accessed: July 15, 2021. Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, et al. Development and Validation of a Clinical Cancer Genomic Profiling Test Based on Massively Parallel DNA Sequencing. Nat Biotechnol, 2013, 31(11): 1023–31., DOI 10.1038/nbt.2696 Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 Human Cancer Genomes Reveals the Landscape of Tumor Mutational burden. Genome Med, 2017, 9(1):34., DOI 10.1186/s13073-017- 0424-2 Foundationmedicine. foundationmedicine. Available at: https://www. foundationmedicine.com/genomic-testing/foundation-one-cdx Accessed: August 14, 2021. World Medical Association.World Medical Association Declaration of Helsinki. JAMA, 2013, 310:2191–4., DOI 10.1001/jama.2013.281053 Park JY, Kricka LJ, and Fortina P. Next-generation Sequencing in the Clinic. Nat Biotechnol, 2013, 31:990–2., DOI 10.1038/nbt.2743 Schwaederle M, Daniels GA, Piccioni DE, Fanta PT, Schwab RB, Shimabukuro KA, et al. On the Road to Precision Cancer Medicine: Analysis of Genomic Biomarker Actionability in 439 Patients. Mol Cancer Ther, 2015, 14:1488–94., DOI 10.1158/1535-7163.MCT-14-1061 Chae YK, Pan AP, Davis AA, Patel SP, Carneiro BA, Kurzrock R, et al. Path toward Precision Oncology: Review of Targeted Therapy Studies and Tools to Aid in Defining “Actionability” of a Molecular Lesion and Patient Management Support. Mol Cancer Ther, 2017, 16:2645–55., DOI 10.1158/ 1535-7163.MCT-17-0597 Roberts N, James S, Delaney M, and Fitzmaurice C. The Global Need and Availability of Blood Products: a Modelling Study. Lancet Haematol, 2019, 6: e606–e615., DOI 10.1016/S2352-3026(19)30200-5 Massard C, Michiels S, Ferte C, Le Deley M-C, Lacroix L, Hollebecque A, et al. High-Throughput Genomics and Clinical Outcome in Hard-To-Treat Advanced Cancers: Results of the MOSCATO 01 Trial. Cancer Discov, 2017, 7:586–95., DOI 10.1158/2159-8290.CD-16-1396 Le Tourneau C, Delord J-P, Goncalves A, Gavoille C, Dubot C, Isambert N, et al. Molecularly Targeted Therapy Based on Tumour Molecular Profiling versus Conventional Therapy for Advanced Cancer, SHIVA): a Multicentre, Open-Label, Proof-Of-Concept, Randomised, Controlled Phase 2 Trial. Lancet Oncol, 2015, 16:1324–34., DOI 10.1016/S1470-2045(15)00188-6 Tsimberidou AM, and Kurzrock R. Precision Medicine: Lessons Learned from the SHIVA Trial. Lancet Oncol, 2015, 16(16):e579–e580., DOI 10.1016/S1470-2045(15, 00397-6 van der Velden DL, Hoes LR, van der Wijngaart H, van Berge Henegouwen JM, van Werkhoven E, Roepman P, et al. The Drug Rediscovery Protocol Facilitates the Expanded Use of Existing Anticancer Drugs. Nature, 2019, 574(7776):127–31., DOI 10.1038/s41586-019-1600-x Dalton WB, Forde PM, Kang H, Connolly RM, Stearns V, Gocke CD, et al. Personalized Medicine in the Oncology Clinic: Implementation and Outcomes of the Johns HopkinsMolecular Tumor Board. JCO Precision Oncol, 2017, 16: 1–19., DOI 10.1200/PO.16.00046 Schwaederle M, Zhao M, Lee JJ, Eggermont AM, Schilsky RL, Mendelsohn J, et al. Impact of Precision Medicine in Diverse Cancers: A Meta-Analysis of Phase II Clinical Trials. Jco, 2015, 33:3817–25., DOI 10.1200/JCO.2015.61.5997 Fontes Jardim DL, Schwaederle M, Wei C, Lee JJ, Hong DS, Eggermont AM, et al. Impact of a Biomarker-Based Strategy on Oncology Drug Development: A Meta-Analysis of Clinical Trials Leading to FDA Approval. JNCI.J, 2015, 107:djv253., DOI 10.1093/jnci/djv253 Barlesi F, Mazieres J, Merlio J-P, Debieuvre D, Mosser J, Lena H, et al. Routine Molecular Profiling of Patients with Advanced Non-small-cell Lung Cancer: Results of a 1-year Nationwide Programme of the French Cooperative Thoracic Intergroup, IFCT). The Lancet, 2016, 387:1415–26., DOI 10.1016/S0140-6736(16)00004-0 Juric D, Rodon J, Tabernero J, Janku F, Burris HA, Schellens JHM, et al. Phosphatidylinositol 3-Kinase α-Selective Inhibition with Alpelisib, BYL719, in PIK3CA-Altered Solid Tumors: Results from the First-In-Human Study. Jco, 2018, 36(13):1291–9., DOI 10.1200/JCO.2017.72.7107 Chakravarty D, and Solit DB. Clinical Cancer Genomic Profiling. Nat Rev Genet, 2021, 22(8):483–501., DOI 10.1038/s41576-021-00338-8 Rodriguez-Rodriguez L, Hirshfield KM, Rojas V, DiPaola RS, Gibbon D, Hellmann M, et al. Use of Comprehensive Genomic Profiling to Direct point-of-care Management of Patients with Gynecologic Cancers. Gynecol Oncol, 2016, 141:2–9., DOI 10.1016/j.ygyno.2016.02.021 Prendergast EN, Holman LL, Liu AY, Lai TS, Campos MP, Fahey JN, et al. Comprehensive Genomic Profiling of Recurrent Endometrial Cancer: Implications for Selection of Systemic Therapy. Gynecol Oncol, 2019, 154: 461–6., DOI 10.1016/j.ygyno.2019.06.016 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609963 EP 1609970 DI 10.3389/pore.2021.1609963 PG 8 ER PT J AU Zhang, C Qian, J Wu, Y Zhu, Z Yu, W Gong, Y Li, X He, Z Zhou, L AF Zhang, Cuijian Qian, Jinqin Wu, Yucai Zhu, Zhenpeng Yu, Wei Gong, Yanqing Li, Xuesong He, Zhisong Zhou, Liqun TI Identification of Novel Diagnosis Biomarkers for Therapy-Related Neuroendocrine Prostate Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; WGCNA; signature; neuroendocrine prostate cancer (NEPC); LASSO; mCRPC ID biomarker; WGCNA; signature; neuroendocrine prostate cancer (NEPC); LASSO; mCRPC AB Background: Therapy-related neuroendocrine prostate cancer (NEPC) is a lethal castration-resistant prostate cancer (CRPC) subtype that, at present, lacks wellcharacterized molecular biomarkers. The clinical diagnosis of this disease is dependent on biopsy and histological assessment: methods that are experience-based and easily misdiagnosed due to tumor heterogeneity. The development of robust diagnostic tools for NEPC may assist clinicians in making medical decisions on the choice of continuing antiandrogen receptor therapy or switching to platinum-based chemotherapy. Methods: Gene expression profiles and clinical characteristics data of 208 samples of metastatic CRPC, including castration-resistant prostate adenocarcinoma (CRPC-adeno) and castration-resistant neuroendocrine prostate adenocarcinoma (CRPC-NE), were obtained from the prad_su2c_2019 dataset. Weighted Gene Co-expression Network Analysis (WGCNA) was subsequently used to construct a free-scale gene co-expression network to study the interrelationship between the potential modules and clinical features of metastatic prostate adenocarcinoma and to identify hub genes in the modules. Furthermore, the least absolute shrinkage and selection operator (LASSO) regression analysis was used to build a model to predict the clinical characteristics of CRPC-NE. The findings were then verified in the nepc_wcm_2016 dataset. Results: A total of 51 co-expression modules were successfully constructed using WGCNA, of which three co-expression modules were found to be significantly associated with the neuroendocrine features and the NEPC score. In total, four novel genes, including NPTX1, PCSK1, ASXL3, and TRIM9, were all significantly upregulated in NEPC compared with the adenocarcinoma samples, and these genes were all associated with the neuroactive ligand receptor interaction pathway. Next, the expression levels of these four genes were used to construct an NEPC diagnosis model, which was successfully able to distinguish CRPC-NE from CRPC-adeno samples in both the training and the validation cohorts. Moreover, the values of the area under the receiver operating characteristic (AUC) were 0.995 and 0.833 for the training and validation cohorts, respectively. Conclusion: The present study identified four specific novel biomarkers for therapyrelated NEPC, and these biomarkers may serve as an effective tool for the diagnosis of NEPC, thereby meriting further study. C1 [Zhang, Cuijian] Peking University, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Department of UrologyBeijing, China. [Qian, Jinqin] Peking University, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Department of UrologyBeijing, China. [Wu, Yucai] Peking University, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Department of UrologyBeijing, China. [Zhu, Zhenpeng] Peking University, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Department of UrologyBeijing, China. [Yu, Wei] Peking University, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Department of UrologyBeijing, China. [Gong, Yanqing] Peking University, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Department of UrologyBeijing, China. [Li, Xuesong] Peking University, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Department of UrologyBeijing, China. [He, Zhisong] Peking University, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Department of UrologyBeijing, China. [Zhou, Liqun] Peking University, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Department of UrologyBeijing, China. RP Zhang, C (reprint author), Peking University, Peking University First Hospital Institute of Urology, National Urological Cancer Center, Department of Urology, Beijing, China. EM surgeon_zhang@126.com CR MaryBeth BC, Isabelle S, Jason AE, Freddie B, and Ahmedin J. Recent Global Patterns in Prostate Cancer Incidence and Mortality Rates. Amsterdam, Netharlands: European Urology, 2019). Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Rebello RJ, Oing C, Knudsen KE, Loeb S, Johnson DC, Reiter RE, et al. Prostate Cancer. Nat Rev Dis Primers, 2021, 7(1):9., DOI 10.1038/s41572-020-00243-0 Yamada Y, and Beltran H. Clinical and Biological Features of Neuroendocrine Prostate Cancer. Curr Oncol Rep, 2021, 23(2):15., DOI 10.1007/s11912-020- 01003-9 Vincenza C, Clara O, Kenneth WE, Rohan B, Michael S, Ana M, et al. Clinical Features of Neuroendocrine Prostate Cancer. Eur J Cancer, 2019). Aparicio AM, Harzstark AL, Corn PG, Wen S, Araujo JC, Tu S-M, et al.Platinum- Based Chemotherapy for Variant Castrate-Resistant Prostate Cancer. Clin Cancer Res, 2013, 19(13):3621–30., DOI 10.1158/1078-0432.ccr-12-3791 Bhagirath D, Liston M, Akoto T, Lui B, Bensing BA, Sharma A, et al. Novel, Non-invasive Markers for Detecting Therapy Induced Neuroendocrine Differentiation in Castration-Resistant Prostate Cancer Patients. Sci Rep, 2021, 11(1):8279., DOI 10.1038/s41598-021-87441-2 Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, et al. Divergent Clonal Evolution of Castration-Resistant Neuroendocrine Prostate Cancer. Nat Med, 2016, 22(3):298–305., DOI 10.1038/nm.4045 Abida W, Cyrta J, Heller G, Prandi D, Armenia J, Coleman I, et al. Genomic Correlates of Clinical Outcome in Advanced Prostate Cancer. Proc Natl Acad Sci USA, 2019, 116(23):11428–36., DOI 10.1073/pnas.1902651116 Bakht MK, Derecichei I, Li Y, Ferraiuolo RM, Dunning M, Oh SW, et al. Neuroendocrine Differentiation of Prostate Cancer Leads to PSMA Suppression. Endocr Relat Cancer, 2018, 26(2):131–46., DOI 10.1530/ERC- 18-0226 Epstein JI, Amin MB, Beltran H, Lotan TL, Mosquera J-M, Reuter VE, et al. Proposed Morphologic Classification of Prostate Cancer with Neuroendocrine Differentiation. Am J Surg Pathol, 2014, 38(6):756–67., DOI 10.1097/ pas.0000000000000208 Lovnicki J, Gan Y, Feng T, Li Y, Xie N, Ho C-H, et al. LIN28B Promotes the Development of Neuroendocrine Prostate Cancer. J Clin Invest, 2020, 130(10): 5338–48., DOI 10.1172/jci135373 Tiwari R, Manzar N, Bhatia V, Yadav A, Nengroo MA, Datta D, et al. Androgen Deprivation Upregulates SPINK1 Expression and Potentiates Cellular Plasticity in Prostate Cancer. Nat Commun, 2020, 11(1):384., DOI 10.1038/s41467-019-14184-0 Yamada Y, and Beltran H. Clinical and Biological Features of Neuroendocrine Prostate Cancer. Curr Oncol Rep, 2021, 23(2):15., DOI 10.1007/s11912-020- 01003-9 Aggarwal RR, and Small EJ. Small-cell/neuroendocrine Prostate Cancer: a Growing Threat? Oncology, Williston Park,, 2014, 28(10):838–40. Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, et al. Divergent Clonal Evolution of Castration-Resistant Neuroendocrine Prostate Cancer. Nat Med, 2016, 22(3):298–305., DOI 10.1038/nm.4045 Qiuli L, Jian P, Lin-ang W, Zhuowei H, Jing X, Xingxia Y, et al. Histone Demethylase PHF8 Drives Neuroendocrine Prostate Cancer Progression by Epigenetically Upregulating FOXA2. J Pathol, 2020)(1, 253106–118., DOI 10.1002/path.5557 Sauerbrei W, Royston P, and Binder H. Selection of Important Variables and Determination of Functional Form for Continuous Predictors in Multivariable Model Building. Statist Med, 2007, 26(30):5512–28., DOI 10.1002/sim.3148 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: a Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci, 2005, 102(43):15545–50., DOI 10.1073/pnas.0506580102 Dong B, Miao J, Wang Y, Luo W, Ji Z, Lai H, et al. Single-cell Analysis Supports a Luminal-Neuroendocrine Transdifferentiation in Human Prostate Cancer. Commun Biol, 2020, 3(1):778., DOI 10.1038/s42003- 020-01476-1 Mariot P, Vanoverberghe K, Lalevee N, Rossier MF, and Prevarskaya N. Overexpression of an α1H, Cav3.2, T-type Calcium Channel during Neuroendocrine Differentiation of Human Prostate Cancer Cells. J Biol Chem, 2002, 277(13):10824–33., DOI 10.1074/jbc.m108754200 FukamiK, Sekiguchi F, YasukawaM,Asano E, Kasamatsu R, UedaM, et al. Functional Upregulation of the H2S/Cav3.2 Channel Pathway Accelerates Secretory Function in Neuroendocrine-Differentiated Human Prostate Cancer Cells. Biochem Pharmacol, 2015, 97(3):300–9., DOI 10.1016/ j.bcp.2015.08.005 Lemonnier L, Lazarenko R, Shuba Y, Thebault S, Roudbaraki M, Lepage G, et al. Alterations in the Regulatory Volume Decrease, RVD, and Swelling- Activated Cl-Current Associated with Neuroendocrine Differentiation of Prostate Cancer Epithelial Cells. Endocr Relat Cancer, 2005, 12(2):335–49., DOI 10.1677/erc.1.00898 Fletcher PA, Sherman A, and Stojilkovic SS. Common and Diverse Elements of Ion Channels and Receptors Underlying Electrical Activity in Endocrine Pituitary Cells. Mol Cell Endocrinol, 2018, 463:23–36., DOI 10.1016/ j.mce.2017.06.022 Tsai HK, Lehrer J, Alshalalfa M, Erho N, Davicioni E, and Lotan TL. Gene Expression Signatures of Neuroendocrine Prostate Cancer and Primary Small Cell Prostatic Carcinoma. BMC cancer, 2017, 17(1):759., DOI 10.1186/s12885- 017-3729-z Boles NC, Hirsch SE, Le S, Corneo B, Najm F, Minotti AP, et al. NPTX1 Regulates Neural Lineage Specification from Human Pluripotent Stem Cells. Cel Rep, 2014, 6(4):724–36., DOI 10.1016/j.celrep.2014.01.026 Amilcar F-M, Tobias BB, Charlotte L, Tanveer SB, Dong L, Mads L, et al. Proteogenomic Characterization of Patient-Derived Xenografts Highlights the Role of REST in Neuroendocrine Differentiation of Castration-Resistant Prostate Cancer. Clin Cancer Res, 2018, 25(2):595–608., DOI 10.1158/1078- 0432.CCR-18-0729 Zhou C,Qin Y, Xie Z, Zhang J, Yang M, Li S, et al. NPTX1 Is a Novel Epigenetic Regulation Gene and Associated with Prognosis in Lung Cancer. Biochem Biophysical Res Commun, 2015, 458(2):381–6., DOI 10.1016/j.bbrc.2015.01.124 Peng X, Pan K, Zhao W, Zhang J, Yuan S, Wen X, et al.NPTX1 Inhibits colon Cancer Cell Proliferation through Down-Regulating Cyclin A2 and CDK2 Expression. Cell Biol Int, 2018, 42(5):589–97., DOI 10.1002/cbin.10935 Yan H, Zheng C, Li Z, Bao B, Yang B, Hou K, et al.NPTX1 Promotes Metastasis via Integrin/FAK Signaling in Gastric Cancer. Cmar, 2019, Vol. 11:3237–51., DOI 10.2147/cmar.s196509 Yue W, Wang T, Zachariah E, Lin Y, Yang CS, Xu Q, et al. Transcriptomic Analysis of Pancreatic Cancer Cells in Response to Metformin and Aspirin: an Implication of Synergy. Sci Rep, 2015, 5(1):13390., DOI 10.1038/srep13390 Kidd M, Modlin IM, and Drozdov I. Gene Network-Based Analysis Identifies Two Potential Subtypes of Small Intestinal Neuroendocrine Tumors. Bmc Genomics, 2014, 15:595., DOI 10.1186/1471-2164-15-595 Jansen E, Ayoubi TAY, Meulemans SMP, and Van de Ven WJM. Neuroendocrine-specific Expression of the Human Prohormone Convertase 1 Gene. J Biol Chem, 1995, 270(25):15391–7., DOI 10.1074/jbc.270.25.15391 Ramalingam S, Eisenberg A, Foo WC, Freedman J, Armstrong AJ, Moss LG, et al.Treatment-related Neuroendocrine Prostate Cancer Resulting in Cushing’s Syndrome. Int J Urol, 2016, 23(12):1038–41., DOI 10.1111/iju.13225 Brennen WN, Yezi Z, Ilsa MC, Susan LD, Lizamma A, Radhika AP, et al.Resistance to Androgen Receptor Signaling Inhibition Does Not Necessitate Development of Neuroendocrine Prostate Cancer. JCI Insight 6(8):e146827., DOI 10.1172/jci.insight.146827 Katoh M. Functional and Cancer Genomics of ASXL Family Members. Br J Cancer, 2013, 109(2):299–306., DOI 10.1038/bjc.2013.281 Shukla V, Rao M, Zhang H, Beers J, Wangsa D, Wangsa D, et al. ASXL3 Is a Novel Pluripotency Factor in Human Respiratory Epithelial Cells and a Potential Therapeutic Target in Small Cell Lung Cancer. Cancer Res, 2017, 77(22):6267–81., DOI 10.1158/0008-5472.can-17-0570 Rossi G, Bertero L, Marchio C, and Papotti M. Molecular Alterations of Neuroendocrine Tumours of the Lung. Histopathology, 2018, 72(1):142–52., DOI 10.1111/his.13394 Szczepanski AP, Zhao Z, Sosnowski T, Goo YA, Bartom ET, and Wang L. ASXL3 Bridges BRD4 to BAP1 Complex and Governs Enhancer Activity in Small Cell Lung Cancer. Genome Med, 2020, 12(1):63., DOI 10.1186/s13073- 020-00760-3 Zeng J, Wang Y, Luo Z, Chang L-C, Yoo JS, Yan H, et al. TRIM9-Mediated Resolution of Neuroinflammation Confers Neuroprotection upon Ischemic Stroke in Mice. Cel Rep, 2019, 27(2):549–60.546., DOI 10.1016/ j.celrep.2018.12.055 Yang F, Liu H, Yu Y, and Xu L. TRIM9 Overexpression Promotes Uterine Leiomyoma Cell Proliferation and Inhibits Cell Apoptosis via NF-Κb Signaling Pathway. Life Sci, 2020, 257:118101., DOI 10.1016/j.lfs.2020.118101 Amorino GP, and Parsons SJ. Neuroendocrine Cells in Prostate Cancer. Crit Rev Eukaryot Gene Expr, 2004, 14(4):287–300., DOI 10.1615/ critreveukaryotgeneexpr.v14.i4.40 McNally CJ, Ruddock MW, Moore T, and McKenna DJ. Biomarkers that Differentiate Benign Prostatic Hyperplasia from Prostate Cancer: A Literature Review. Cmar, 2020, Vol. 12:5225–41., DOI 10.2147/cmar.s250829 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609968 EP 1609980 DI 10.3389/pore.2021.1609968 PG 13 ER PT J TI Retraction: MiR-424-5p Inhibits Proliferation, Invasion and Promotes Apoptosis and Predicts Good Prognosis in Glioma by Directly Targeting BFAR SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Retraction AB MiR-424-5p Inhibits Proliferation, Invasion and Promotes Apoptosis and Predicts Good Prognosis in Glioma by Directly Targeting BFAR by Cheng, Z., Shu, H., Cui, Y., Zhang, Q., Zhao, B., Pan, D., Chao, Q., and Wang, D. (2020). MiR-424- 5p Inhibits Proliferation, Invasion and Promotes Apoptosis and Predicts Good Prognosis in Glioma by Directly Targeting BFAR. Pathol. Oncol. Res. 26, 2327–2335. doi: 10.1007/s12253-020-00831-1 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2021 VL 27 IS 3 BP 1609970 EP 1609970 DI 10.3389/pore.2021.1609970 PG 1 ER PT J AU Chen, L Chen, Y Zhang, L Xue, Y Zhang, Sh Li, X Song, H AF Chen, Li Chen, Yong Zhang, Lele Xue, Yingwei Zhang, Shiwei Li, Xingrui Song, Hongjiang TI In Gastric Cancer Patients Receiving Neoadjuvant Chemotherapy Systemic Inflammation Response Index is a Useful Prognostic Indicator SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; neoadjuvant chemotherapy; advanced gastric cancer; systemic inflammation response index (SIRI); tumor indicator ID prognosis; neoadjuvant chemotherapy; advanced gastric cancer; systemic inflammation response index (SIRI); tumor indicator AB Background: The preoperative systemic inflammation response index (SIRI), based on peripheral neutrophil (N), monocyte (M), and lymphocyte (L) counts, has shown mounting evidence as an effective prognostic indicator in some malignant tumors. The aim of the present study was to evaluate the prognostic significance of pretreatment SIRI in gastric cancer patients who received neoadjuvant chemotherapy (NACT). Methods: This retrospective study comprised 107 patients with advanced gastric cancer treated with NACT between July 2007 and September 2015 in our hospital. SIRI was calculated from peripheral venous blood samples obtained prior to treatment. The best cutoff value for SIRI by receiver operating characteristic (ROC) curve was 1.2 (low SIRI <1.21, high SIRI ≥1.21). The clinical outcomes of disease-free survival (DFS) and overall survival (OS) were analyzed by Kaplan-Meier survival analysis and compared using the logrank test. Univariate and multivariate analyses were performed by the Cox proportional hazards regression model. Results: The results demonstrated that the low SIRI group was statistically associated with gender, primary tumor site, white blood cell, neutrophil, and monocyte counts, NLR (neutrophil to lymphocyte ratio), MLR (monocyte to lymphocyte ratio), and PLR (platelet to lymphocyte ratio). The SIRI was predictive for DFS and OS by univariate and multivariate analysis; the low SIRI group had better median DFS and OS than the high SIRI group (median DFS 27.03 vs. 22.33 months, median OS 29.73 vs. 24.43 months). The DFS and OS in the low SIRI group were longer than the high SIRI group. Conclusions: SIRI may qualify as a useful, reliable, and convenient prognostic indicator in patients with advanced gastric cancer to help physicians to provide personalized prognostication for gastric cancer patients treated with NACT. C1 [Chen, Li] Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Department of Thyroid and Breast SurgeryWuhan, China. [Chen, Yong] Huai’an Second People’s Hospital and the Affiliated Huai’an Hospital of Xuzhou Medical University, Department of General SurgeryHuai’an, China. [Zhang, Lele] Harbin Medical University, Harbin Medical University Cancer Hospital, Department of Gastrointestinal SurgeryHarbin, China. [Xue, Yingwei] Harbin Medical University, Harbin Medical University Cancer Hospital, Department of Gastrointestinal SurgeryHarbin, China. [Zhang, Shiwei] The First People’s Hospital of Fuyang Hangzhou, Department of Oncology SurgeryHangzhou, China. [Li, Xingrui] Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital, Department of Thyroid and Breast SurgeryWuhan, China. [Song, Hongjiang] Harbin Medical University, Harbin Medical University Cancer Hospital, Department of Gastrointestinal SurgeryHarbin, China. RP Song, H (reprint author), Harbin Medical University, Harbin Medical University Cancer Hospital, Department of Gastrointestinal Surgery, Harbin, China. EM hongjiangsong2016@163.com CR Siegel RL, Miller KD, and Jemal A. Cancer Statistics, 2020. CA A Cancer J Clin, 2020, 70(1):7–30., DOI 10.3322/caac.21590 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, and Lordick F. Gastric Cancer. The Lancet, 2020, 396(10251):635–48., DOI 10.1016/S0140-6736(20, 31288-5 Lordick F, Shitara K, and Janjigian YY. New Agents on the Horizon in Gastric Cancer. Ann Oncol, 2017, 28(8):1767–75., DOI 10.1093/annonc/mdx051 Neves Filho EHC, de Sant’Ana RO, Nunes LVSC, Pires APB, and da Cunha Md. PSS. Histopathological Regression of Gastric Adenocarcinoma after Neoadjuvant Therapy: a Critical Review. APMIS, 2017, 125(2):79–84., DOI 10.1111/apm.12642 Rausei S, Bali CD, and Lianos GD. Neoadjuvant Chemotherapy for Gastric Cancer. Has the Time to Decelerate the Enthusiasm Passed Us by. Semin Oncol, 2020, 47(6):355–60., DOI 10.1053/j.seminoncol.2020.07.003 Lin JX, Yoon C, Desiderio J, Yi BC, Li P, Zheng CH, et al. Development and Validation of a Staging System for Gastric Adenocarcinoma after Neoadjuvant Chemotherapy and Gastrectomy with D2 Lymphadenectomy. Br J Surg, 2019, 106(9):1187–96., DOI 10.1002/bjs.11181 Diakos CI, Charles KA, McMillan DC, and Clarke SJ. Cancer-related Inflammation and Treatment Effectiveness. Lancet Oncol, 2014, 15(11): e493–e503., DOI 10.1016/S1470-2045(14)70263-3 Miyamoto R, Inagawa S, Sano N, Tadano S, Adachi S, and Yamamoto M. The Neutrophil-To-Lymphocyte Ratio, NLR, Predicts Short-Term and Long- Term Outcomes in Gastric Cancer Patients. Eur J Surg Oncol, 2018, 44(5): 607–12., DOI 10.1016/j.ejso.2018.02.003 Shoji F, Kozuma Y, Toyokawa G, Yamazaki K, and Takeo S. Complete Blood Cell Count-Derived Inflammatory Biomarkers in Early-Stage Nonsmall- cell Lung Cancer. Atcs, 2020, 26(5):248–55., DOI 10.5761/ atcs.oa.19-00315 Li B, Zhou P, Liu Y, Wei H, Yang X, Chen T, et al. Platelet-to-lymphocyte Ratio in Advanced Cancer: Review and Meta-Analysis. Clinica Chim Acta, 2018, 483:48–56., DOI 10.1016/j.cca.2018.04.023 Chen Y, Jin M, Shao Y, and Xu G., 2019). Prognostic Value of the Systemic Inflammation Response Index in Patients with Adenocarcinoma of the Oesophagogastric Junction: A Propensity Score-Matched Analysis. Dis Markers, 2019 1, 9., DOI 10.1155/2019/4659048 Chao B, Ju X, Zhang L, Xu X, and Zhao Y. A Novel PrognosticMarker Systemic Inflammation Response Index, SIRI, for Operable Cervical Cancer Patients. Front Oncol, 2020, 10:766., DOI 10.3389/fonc.2020.00766 Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, et al. The Eighth Edition AJCC Cancer Staging Manual: Continuing to Build a Bridge from a Population-Based to a More "personalized" Approach to Cancer Staging. CA: A Cancer J Clinicians, 2017, 67(2): 93–9., DOI 10.3322/caac.21388 Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New Response Evaluation Criteria in Solid Tumours: Revised RECIST Guideline, Version 1.1). Eur J Cancer, 2009, 45(2):228–47., DOI 10.1016/ j.ejca.2008.10.026 Caussanel J-P, Levi F, Brienza S, Misset J-L, Itzhaki M, Adam R, et al. Phase I Trial of 5-Day Continuous Venous Infusion of Oxaliplatin at Circadian Rhythm-Modulated Rate Compared with Constant Rate. J Natl Cancer Inst, 1990, 82(12):1046–50., DOI 10.1093/jnci/82.12.1046 Li S, Lan X, Gao H, Li Z, Chen L, Wang W, et al. Systemic Inflammation Response Index, SIRI), Cancer Stem Cells and Survival of Localised Gastric Adenocarcinoma after Curative Resection. J Cancer Res Clin Oncol, 2017, 143(12):2455–68., DOI 10.1007/s00432-017-2506-3 Reddavid R, Sofia S, Chiaro P, Colli F, Trapani R, Esposito L, et al. Neoadjuvant Chemotherapy for Gastric Cancer. Is it a Must or a Fake. World. J. Gastroentrol., 2018, 24(2):274–89., DOI 10.3748/wjg.v24.i2.274 Grivennikov SI, Greten FR, and Karin M. Immunity, Inflammation, and Cancer. Cell, 2010, 140:883–99., DOI 10.1016/j.cell.2010.01.025 Mantovani A, Allavena P, Sica A, and Balkwill F. Cancer-related Inflammation. Nature, 2008, 454(7203):436–44., DOI 10.1038/nature07205 Kim EY, Lee JW, Yoo HM, Park CH, and Song KY. The Platelet-To- Lymphocyte Ratio versus Neutrophil-To-Lymphocyte Ratio: Which Is Better as a Prognostic Factor in Gastric Cancer. Ann Surg Oncol, 2015, 22(13):4363–70., DOI 10.1245/s10434-015-4518-z Li T-J, Jiang Y-M, Hu Y-F, Huang L, Yu J, Zhao L-Y, et al. Interleukin-17- Producing Neutrophils Link Inflammatory Stimuli to Disease Progression by Promoting Angiogenesis in Gastric Cancer. Clin Cancer Res, 2017, 23(6): 1575–85., DOI 10.1158/1078-0432.CCR-16-0617 Kilincalp S, Ekiz F, Basar O, Ayte MR, Coban S, Yilmaz B, et al. Mean Platelet Volume Could Be Possible Biomarker in Early Diagnosis andMonitoring of Gastric Cancer. Platelets, 2014, 25(8):592–4., DOI 10.3109/09537104.2013.783689 Eo WK, Jeong DW, Chang HJ, Won KY, Choi SI, Kim SH, et al. Absolute Monocyte and Lymphocyte Count Prognostic Score for Patients with Gastric Cancer. Wjg, 2015, 21(9):2668–76., DOI 10.3748/wjg.v21.i9.2668 Ock C-Y, Nam A-R, Lee J, Bang J-H, Lee K-H, Han S-W, et al. Prognostic Implication of Antitumor Immunity Measured by the Neutrophil-Lymphocyte Ratio and Serum Cytokines and Angiogenic Factors in Gastric Cancer. Gastric Cancer, 2017, 20(2):254–62., DOI 10.1007/s10120-016-0613-5 Chen L, Hao Y, Zhu L, Li S, Zuo Y, Zhang Y, et al. Monocyte to Lymphocyte Ratio Predicts Survival in Patients with Advanced Gastric Cancer Undergoing Neoadjuvant Chemotherapy. Ott, 2017, Vol. 10:4007–16., DOI 10.2147/OTT.S140118 Koh C-H, Bhoo-Pathy N, Ng K-L, Jabir RS, Tan G-H, See M-H, et al. Utility of Pre-treatment Neutrophil-Lymphocyte Ratio and Platelet-Lymphocyte Ratio as Prognostic Factors in Breast Cancer. Br J Cancer, 2015, 113(1):150–8., DOI 10.1038/bjc.2015.183 Kolaczkowska E, and Kubes P. Neutrophil Recruitment and Function in Health and Inflammation. Nat Rev Immunol, 2013, 13(3):159–75., DOI 10.1038/nri3399 Tan KW, Chong SZ, Wong FHS, Evrard M, Tan SM-L, Keeble J, et al. Neutrophils Contribute to Inflammatory Lymphangiogenesis by Increasing VEGF-A Bioavailability and Secreting VEGF-D. Blood, 2013, 122(22): 3666–77., DOI 10.1182/blood-2012-11-466532 Shi C, and Pamer EG. Monocyte Recruitment during Infection and Inflammation. Nat Rev Immunol, 2011, 11:762–74., DOI 10.1038/nri3070 Gabrilovich DI, Ostrand-Rosenberg S, and Bronte V. Coordinated Regulation of Myeloid Cells by Tumours. Nat Rev Immunol, 2012, 12:253–68., DOI 10.1038/nri3175 Ogiya R, Niikura N, Kumaki N, Bianchini G, Kitano S, Iwamoto T, et al. Comparison of Tumor-Infiltrating Lymphocytes between Primary and Metastatic Tumors in Breast Cancer Patients. Cancer Sci, 2016, 107(12): 1730–5., DOI 10.1111/cas.13101 Ferrone C, and Dranoff G. Dual Roles for Immunity in Gastrointestinal Cancers. J. Clin. Oncol., 2010, 28(26):4045–51., DOI 10.1200/JCO.2010.27.9992 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609811 EP 1609820 DI 10.3389/pore.2021.1609811 PG 10 ER PT J AU Qadir, J Majid, S Khan, SM Rashid, F Wani, DM Bhat, ASh AF Qadir, Jasiya Majid, Sabhiya Khan, Saleem Mosin Rashid, Fouzia Wani, Din Mumtaz Bhat, Ahmad Showkat TI Implication of ARID1A Undercurrents and PDL1, TP53 Overexpression in Advanced Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gastric cancer; ARID1A; mutation; mRNA expression 3; mRNA expression; qRT-PCR; TP53; PDL1 ID gastric cancer; ARID1A; mutation; mRNA expression 3; mRNA expression; qRT-PCR; TP53; PDL1 AB AT-rich interactive domain-containing protein 1A (ARID1A), TP53 and programmed cell deathligand 1 (PDL1) are involved in several protein interactions that regulate the expression of various cancer-related genes involved in the progression of the cell cycle, cell proliferation, DNA repair, and apoptosis. In addition, gene expression analysis identified some common downstream targets of ARID1A and TP53. It has been established that tumors formed by ARID1A-deficient cancer cells exhibited elevated PDL1 expression. However, the aberrations in these molecules have not been studied in this population especially in Gastric Cancer (GC). In this backdrop we aimed to investigate the role of the ARID1A mutation and expression of ARID1A, TP53 and PDL1 genes in the etiopathogenesis of Gastric Cancer (GC) in the ethnic Kashmiri population (North India). The study included 103 histologically confirmed GC cases. The mutations, if any, in exon-9 of ARID1A gene was analysed by Polymerase Chain Reaction (PCR) followed by Sanger sequencing. ThemRNA expression of the ARID1A, TP53 and PDL1 genes was analysed by Quantitative real time-PCR (qRT-PCR). We identified a nonsense mutation (c.3219; C > T) in exon-9 among two GC patients (∼2.0%), which introduces a premature stop codon at protein position 1073. The mRNA expression of the ARID1A, TP53 and PDL1 gene was significantly reduced in 25.3% and elevated in 47.6 and 39.8% of GC cases respectively with a mean fold change of 0.63, 2.93 and 2.43. The data revealed that reducedmRNA expression of ARID1A and elevatedmRNA expression of TP53 and PDL1 was significantly associated with the high-grade and advanced stage of cancer. Our study proposes that ARAD1A under-expression and overexpression of TP53 and PDL1 might be crucial for tumor progression with TP53 and PDL1 acting synergistically. C1 [Qadir, Jasiya] Government Medical College Srinagar and Associated Hospitals, Department of BiochemistrySrinagar, India. [Majid, Sabhiya] Government Medical College Srinagar and Associated Hospitals, Department of BiochemistrySrinagar, India. [Khan, Saleem Mosin] Government Medical College Srinagar and Associated Hospitals, Department of BiochemistrySrinagar, India. [Rashid, Fouzia] University of Kashmir, Department of Clinical BiochemistrySrinagar, India. [Wani, Din Mumtaz] Government Medical College Srinagar and Associated Hospitals, Department of SurgerySrinagar, India. [Bhat, Ahmad Showkat] Government Medical College, Department of BiochemistryDoda, India. RP Majid, S (reprint author), Government Medical College Srinagar and Associated Hospitals, Department of Biochemistry, Srinagar, India. EM sabuumajid@gmail.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Qurieshi MA, Khan SMS, Masoodi MA, Qurieshi U, Ain Q, Jan Y, et al. Epidemiology of Cancers in Kashmir, India: an Analysis of Hospital Data. Adv Prev Med, 2016, 2016:1–6., DOI 10.1155/2016/ 1896761 Oki E, Kakeji Y, Zhao Y, Yoshida R, Ando K, Masuda T, et al. Chemosensitivity and Survival in Gastric Cancer Patients with Microsatellite Instability. Ann Surg Oncol, 2009, 16:2510–5., DOI 10.1245/s10434-009-0580-8 Wilson BG, Roberts CWM. SWI/SNF Nucleosome Remodellers and Cancer. Nat Rev Cancer, 2011, 11:481–92., DOI 10.1038/nrc3068 Samartzis E, Noske A, Dedes K, Fink D, Imesch P. ARID1A Mutations and PI3K/AKT Pathway Alterations in Endometriosis and Endometriosis- Associated Ovarian Carcinomas. Int J Mol Sci, 2013, 14(9):18824–49., DOI 10.3390/ijms140918824 Qadir J, Majid S, Khan MS, Rashid F, Wani MD, Din I, et al. AT-rich Interaction Domain 1A Gene Variations: Genetic Associations and Susceptibility to Gastric Cancer Risk. Pathol Oncol Res, 2020, 26(4): 2237–46., DOI 10.1007/s12253-020-00815-1 Mathur R, Roberts CWM. SWI/SNF, BAF, Complexes: Guardians of the Epigenome. Annu Rev Cancer Biol, 2018, 2:413–27., DOI 10.1146/annurevcancerbio- 030617-050151 Jones S, Li M, Parsons DW, Zhang X, Wesseling J, Kristel P, et al. Somatic Mutations in the Chromatin Remodeling Gene ARID1A Occur in Several Tumor Types. Hum Mutat, 2012, 33(1):100–3., DOI 10.1002/ humu.21633 Abe H, Maeda D, Hino R, Otake Y, Isogai M, Ushiku AS, et al. ARID1A Expression Loss in Gastric Cancer: Pathway-dependent Roles with and without Epstein-Barr Virus Infection and Microsatellite Instability. Virchows Arch, 2012, 461:367–77., DOI 10.1007/s00428-012-1303-2 Wang D-d., Chen Y-b., Pan K, Wang W, Chen S-p., Chen J-g., et al. Decreased Expression of the ARID1A Gene Is Associated with Poor Prognosis in Primary Gastric Cancer. PLoS One, 2012, 7:e40364., DOI 10.1371/journal.pone.0040364 Levine AJ. p53, the Cellular Gatekeeper for Growth and Division. Cell, 1997, 88:323–31., DOI 10.1016/s0092-8674(00)81871-1 Ho L, Crabtree GR. Chromatin Remodelling during Development. Nature, 2010, 463:474–84., DOI 10.1038/nature08911 Bosse T, ter Haar NT, Seeber LM, Diest PJv., Hes FJ, Vasen HF, et al. Loss of ARID1A Expression and its Relationship with PI3K-Akt Pathway Alterations, TP53 and Microsatellite Instability in Endometrial Cancer. Mod Pathol, 2013, 26:1525–35., DOI 10.1038/ modpathol.2013.96 Hanahan D, Weinberg RA. Hallmarks of Cancer: the Next Generation. Cell, 2011, 144:646–74., DOI 10.1016/j.cell.2011.02.013 Hou J, Yu Z, Xiang R, Li C, Wang L, Chen S, et al. Correlation between Infiltration of FOXP3+ Regulatory T Cells and Expression of B7-H1 in the Tumor Tissues of Gastric Cancer. Exp Mol Pathol, 2014, 96:284–91., DOI 10.1016/j.yexmp.2014.03.005 Naito T, Udagawa H, Umemura S, Sakai T, Zenke Y, Kirita K, et al. Nonsmall Cell Lung Cancer with Loss of Expression of the SWI/SNF Complex Is Associated with Aggressive Clinicopathological Features, PD-L1-Positive Status, and High Tumor Mutation burden. Lung Cancer, 2019, 138:35–42., DOI 10.1016/ j.lungcan.2019.10.009 Kim YB, Ahn JM, Bae WJ, Sung CO, Lee D. Functional Loss of ARID1A Is Tightly Associated with High PD-L1 Expression in Gastric Cancer. Int J Cancer, 2019, 145:916–26., DOI 10.1002/ijc.32140 Silva R, Gullo I, Carneiro F. The PD-1:PD-L1 Immune Inhibitory Checkpoint in Helicobacter pylori Infection and Gastric Cancer: a Comprehensive Review and Future Perspectives. Porto Biomed J, 2016, 1(1):4–11., DOI 10.1016/ j.pbj.2016.03.004 Gu L, Chen M, Guo D, Zhu H, Zhang W, Pan J, et al. PD-L1 and Gastric Cancer Prognosis: A Systematic Review and Meta-Analysis. PLoS One, 2017, 12(8):e0182692., DOI 10.1371/journal.pone.0182692 Waddell T, Chau I, Cunningham D, Gonzalez D, Okines AFC, Wotherspoon A, et al. Epirubicin, Oxaliplatin, and Capecitabine with or without Panitumumab for Patients with Previously Untreated Advanced Oesophagogastric Cancer, REAL3): a Randomised, Open- Label Phase 3 Trial. Lancet Oncol, 2013, 14:481–9., DOI 10.1016/s1470- 2045(13)70096-2 Liu H, Zhou M. Evaluation of P53 Gene Expression and Prognosis Characteristics in Uveal Melanoma Cases. Onco Targets Ther, 2017, 10: 3429–34., DOI 10.2147/ott.s136785 Ito S, Masuda T, Noda M, Hu Q, Shimizu D, Kuroda Y, et al. Prognostic Significance of PD-1, PD-L1 and CD8 Gene Expression Levels in Gastric Cancer. Oncology, 2020, 98(7):501–11., DOI 10.1159/000506075 Livak KJ, Schmittgen TD. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2−ΔΔCT Method. Methods, 2001, 25(4): 402–8., DOI 10.1006/meth.2001.1262 Hu N, Qian L, Hu Y, Shou J-Z, Wang C, Giffen C, et al. Quantitative Real- Time RT-PCR Validation of Differential mRNA Expression of SPARC, FADD, Fascin, COL7A1, CK4, TGM3, ECM1, PPL and EVPLin Esophageal Squamous Cell Carcinoma. BMC Cancer, 2006, 6:33., DOI 10.1186/1471-2407-6-33 Nobili S, Bruno L, Landini I, Napoli C, Bechi P, Tonelli F, et al. Genomic and Genetic Alterations Influence the Progression of Gastric Cancer. World J Gastroenterol, 2011, 17(3):290–9., DOI 10.3748/wjg.v17.i3.290 Pavlidou EN, Balis V. Diagnostic Significance and Prognostic Role of the ARID1A Gene in Cancer Outcomes, Review). World Acad Sci J, 2020, 2: 49–64., DOI 10.3892/wasj.2020.37 Mamo A, Cavallone L, Tuzmen S, Chabot C, Ferrario C, Hassan S, et al. An Integrated Genomic Approach Identifies ARID1A as a Candidate Tumor- Suppressor Gene in Breast Cancer. Oncogene, 2012, 31(16):2090–100., DOI 10.1038/onc.2011.386 SjoblomT, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, et al. The Consensus Coding Sequences of Human Breast and Colorectal Cancers. Science, 2006, 314(5797):268–74., DOI 10.1126/ science.1133427 Erfani M, Hosseini SV, Mokhtari M, Zamani M, Tahmasebi K, Alizadeh Naini M, et al. Altered ARID1A Expression in Colorectal Cancer. BMC Cancer, 2020, 20(1):350., DOI 10.1186/s12885-020-6706-x Yang Y, Wang X, Yang J, Duan J, Wu Z, Yang F, et al. Loss of ARID1A Promotes Proliferation, Migration and Invasion via the Akt Signaling Pathway in NPC. Cancer Manag Res, 2019, 11:4931–46., DOI 10.2147/cmar.s207329 Lai T, Vierkoetter K, Ayabe A, Jun Ahn H, Shimizu D, Keith YT. Ethnic Variations in ARID1a Expression in clear Cell and Endometrioid Ovarian Carcinoma. J Clin Oncol, 2016, 34(15Suppl. l):e17073., DOI 10.1200/ jco.2016.34.15_suppl.e17073 Chan-On W, Nairismagi M-L, Ong CK, Lim WK, Dima S, Pairojkul C, et al. Exome Sequencing Identifies Distinct Mutational Patterns in Liver Fluke- Related and Non-infection-related Bile Duct Cancers. Nat Genet, 2013, 45(12):1474–8., DOI 10.1038/ng.2806 Lee SY, Kim D-W, Lee HS, Ihn MH, Oh H-K, Park DJ, et al. Loss of AT-Rich Interactive Domain 1A Expression in Gastrointestinal Malignancies. Oncology, 2015, 88(4):234–40., DOI 10.1159/000369140 Chou A, Toon CW, Clarkson A, Sioson L, Houang M, Watson N, et al. Loss of ARID1A Expression in Colorectal Carcinoma Is Strongly Associated with Mismatch Repair Deficiency. Hum Pathol, 2014, 45(8):1697–703., DOI 10.1016/j.humpath.2014.04.009 Jopling CL. Stop that Nonsense!!. eLife, 2014, 3:e04300., DOI 10.7554/ eLife.04300 Olivier M, Hollstein M, Hainaut P. TP53 Mutations in Human Cancers: Origins, Consequences, and Clinical Use. Cold Spring Harbor Perspect Biol, 2010, 2:a001008., DOI 10.1101/cshperspect.a001008 Ogawa M, Onoda N, Maeda K, Kato Y, Nakata B, Kang S-M, et al. A Combination Analysis of P53 and P21 in Gastric Carcinoma as a strong Indicator for Prognosis. Int J Mol Med, 2001, 7:479–83., DOI 10.3892/ ijmm.7.5.479 Avery-Kiejda KA, Morten B, Wong-Brown MW, Mathe A, Scott RJ. The Relative mRNA Expression of P53 Isoforms in Breast Cancer Is Associated with Clinical Features and Outcome. Carcinogenesis, 2014, 35(3):586–96., DOI 10.1093/carcin/bgt411 Gope ML, Chun M, Gope R. Comparative Study of the Expression of Rb and P53 Genes in Human Colorectal Cancers, colon Carcinoma Cell Lines and Synchronized Human Fibroblasts. Mol Cel Biochem, 1991, 107:55–63., DOI 10.1007/bf02424576 Lothe RA, Fossli T, Danielsen HE, Stenwig AE, Nesland JM, Gallie B, et al. Molecular Genetic Studies of Tumor Suppressor Gene Regions on Chromosomes 13 and 17 in Colorectal Tumors. JNCI J Natl Cancer Inst, 1992, 84:1100–8., DOI 10.1093/jnci/84.14.1100 Takahashi R, Markovic SN, Scrable HJ. Dominant Effects of Δ40p53 on P53 Function and Melanoma Cell Fate. J Invest Dermatol, 2014, 134(3):791–800., DOI 10.1038/jid.2013.391 Bourdon J-C, Fernandes K, Murray-Zmijewski F, Liu G, Diot A, Xirodimas DP, et al. p53 Isoforms Can Regulate P53 Transcriptional Activity. Genes Dev, 2005, 19:2122–37., DOI 10.1101/gad.1339905 Fenoglio-Preiser CM, Wang J, Stemmermann GN, Noffsinger A. TP53 and Gastric Carcinoma: a Review. Hum Mutat, 2003, 21(3):258–70., DOI 10.1002/ humu.10180 Hofstetter G, Berger A, Fiegl H, Slade N, Zoric A, Holzer B, et al. Alternative Splicing of P53 and P73: the Novel P53 Splice Variant P53δ Is an Independent Prognostic Marker in Ovarian Cancer. Oncogene, 2010, 29:1997–2004., DOI 10.1038/onc.2009.482 Ioachim E, Goussia A, Stefanou D, Agnantis NJ. Expression of P53 Protein in Gastric Cancer: an Immunohistochemical Study with Correlation to Proliferative Activity. Anticancer Res, 1997, 17:513–7. Sasaki I, Yao T, Nawata H, Tsuneyoshi M. Minute Gastric Carcinoma of Differentiated Type with Special Reference to the Significance of Intestinal Metaplasia, Proliferative Zone, and P53 Protein during Tumor Development. Cancer, 1999, 85:1719–29., DOI 10.1002/(sici)1097-0142(19990415)85:8<1719: aid-cncr11>3.0.co;2-v Kakeji Y, Korenaga D, Tsujitani S, Baba H, Anai H, Maehara Y, et al. Gastric Cancer with P53 Overexpression Has High Potential for Metastasising to Lymph Nodes. Br J Cancer, 1993, 67:589–93., DOI 10.1038/bjc.1993.108 Monig SP, Eidt S, Zirbes TK, Stippel D, Baldus SE, Pichlmaier H. p53 Expression in Gastric Cancer: Clinicopathological Correlation and Prognostic Significance. Dig Dis Sci, 1997, 42:2463–7., DOI 10.1023/a: 1018844008068 El-Mahdani N, Vaillant J-C, GuiguetM, Prevot S, Bertrand V, Bernard C, et al. Overexpression of P53 mRNA in Colorectal Cancer and its Relationship to P53 Gene Mutation. Br J Cancer, 1997, 75(4):528–36., DOI 10.1038/bjc.1997.92 Muhlmann G, Ofner D, Zitt M, Muller HM, Maier H, Moser P, et al. 14-3-3 Sigma and P53 Expression in Gastric Cancer and its Clinical Applications. Dis Markers, 2010, 29(1):21–9., DOI 10.1155/2010/470314 Carneiro F, David L, Sobrinho-Simoes M, Seruca R, Nesland JM. Oncogene S and Onco-Suppressor Genes in Gastric Cancer Carcinoma. Surg Pathol, 1994, 5(3):225–38. Haronikova L, Olivares-Illana V, Wang L, Karakostis K, Chen S, Fahraeus R. The P53 mRNA: an Integral Part of the Cellular Stress Response. Nucleic Acids Res, 2019, 47(7):3257–71., DOI 10.1093/nar/gkz124 Shiao YH, Rugge M, Correa P, Lehmann HP, Scheer WD. p53 Alteration in Gastric Precancerous Lesions. Am J Pathol, 1994, 144:511–7. Misiewicz JJ. The Sydney System: A New Classification of Gastritis. Introduction. J Gastroenterol Hepatol, 1991, 6(3):207–8., DOI 10.1111/j.1440- 1746.1991.tb01467.x Morales-Fuentes GA, Zarate-Osorno A, Quinonez-Urrego EE, Antonio- Manrique M, Martinez-Garcia CL, Figueroa-Barojas P, et al. p53 expresado en la mucosa gastrica de pacientes infectados por Helicobacter pylori. Rev Gastroenterol Mex, 2013, 78(1):12–20., DOI 10.1016/j.rgmx.2012.11.001 Wei J, Nagy TA, Vilgelm A, Zaika E, Ogden SR, Romero–Gallo J, et al. Regulation of P53 Tumor Suppressor by Helicobacter pylori in Gastric Epithelial Cells. Gastroenterology, 2010, 139:1333–43., DOI 10.1053/ j.gastro.2010.06.018 Ahmed A, Smoot D, Littleton G, Tackey R, Walters CS, Kashanchi F, et al. Helicobacter pylori Inhibits Gastric Cell Cycle Progression. Microbes Infect, 2000, 2:1159–69., DOI 10.1016/s1286-4579(00)01270-3 Wu C, Zhu Y, Jiang J, Zhao J, Zhang X-G, Xu N. Immunohistochemical Localization of Programmed Death-1 Ligand-1, PD-L1, in Gastric Carcinoma and its Clinical Significance. Acta Histochem, 2006, 108:19–24., DOI 10.1016/ j.acthis.2006.01.003 Sun J, Xu K, Wu C, Wang Y, Hu Y, Zhu Y, et al. PD-L1 Expression Analysis in Gastric Carcinoma Tissue and Blocking of Tumor-Associated PD-L1 Signaling by Two Functional Monoclonal Antibodies. Tissue Antigens, 2007, 69:19–27., DOI 10.1111/j.1399-0039.2006.00701.x Chen XL, Cao XD, Kang AJ, Wang KM, Su BS, Wang YL. In Situ expression and Significance of B7 Costimulatory Molecules within Tissues of Human Gastric Carcinoma. World J Gastroenterol, 2003, 9:1370–3., DOI 10.3748/wjg.v9.i6.1370 Afreen S, Dermime S. The Immunoinhibitory B7-H1 Molecule as a Potential Target in Cancer: Killing many Birds with One Stone. Hematol Oncol Stem Cel Ther, 2014, 7:1–17., DOI 10.1016/j.hemonc.2013.09.005 CortezMA, Ivan C, Valdecanas D, Wang X, PeltierHJ, Ye Y, et al. PDL1 Regulation by P53 via miR-34. J Natl Cancer Inst, 2016, 108(1):djv303., DOI 10.1093/jnci/djv303 Cha YJ, Kim HR, Lee CY, Cho BC, Shim HS. Clinicopathological and Prognostic Significance of Programmed Cell Death Ligand-1 Expression in Lung Adenocarcinoma and its Relationship with P53 Status. Lung Cancer, 2016, 97:73–80., DOI 10.1016/j.lungcan.2016.05.001 Tojyo I, Shintani Y, Nakanishi T, Okamoto K, Hiraishi Y, Fujita S, et al. PD-L1 Expression Correlated with P53 Expression in Oral Squamous Cell Carcinoma. Maxillofac Plast Reconstr Surg, 2019, 41(1):56., DOI 10.1186/s40902-019- 0239-8 Chang T-C, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, Lee KH, et al. Transactivation of miR-34a by P53 Broadly Influences Gene Expression and Promotes Apoptosis. Mol Cel, 2007, 26:745–52., DOI 10.1016/ j.molcel.2007.05.010 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609826 EP 1609838 DI 10.3389/pore.2021.1609826 PG 13 ER PT J AU Chen, Ch Wang, T Yang, M Song, J Huang, M Bai, Y Su, H AF Chen, Chen Wang, Tao Yang, Mengmei Song, Jia Huang, Mengli Bai, Yuezong Su, Hao TI Genomic Profiling of Blood-Derived Circulating Tumor DNA from Patients with Advanced Biliary Tract Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE next-generation sequencing; signaling pathway; circulating tumor DNA; biliary tract cancer; genomic feature ID next-generation sequencing; signaling pathway; circulating tumor DNA; biliary tract cancer; genomic feature AB Background: Biliary tract cancer is a highly lethal malignancy with poor clinical outcome. Accumulating evidence indicates targeted therapeutics may provide new hope for improving treatment response in BTC, hence better understanding the genomic profile is particularly important. Since tumor tissue may not be available for some patients, a complementary method is urgently needed. Circulating tumor DNA (ctDNA) provides a noninvasive means for detecting genomic alterations, and has been regarded as a promising tool to guide clinical therapies. Methods: Next-generation sequencing of 150 cancer-related genes was used to detect gene alterations in blood-derived ctDNA from 154 Chinese patients with BTC. Genomic alterations were analyzed and compared with an internal tissue genomic database and TCGA database. Results: 94.8% patients had at least one change detected in their ctDNA. The median maximumsomatic allele frequency was 6.47% (ranging 0.1–34.8%). TP53 and KRAS were the most often mutated genes. The frequencies of single nucleotide variation in commonly mutated genes in ctDNA were similar to those detected in tissue samples, TP53 (35.1 vs. 40.4%) and KRAS (20.1 vs. 22.6%). Pathway analysis revealed that mutated genes were mapped to several key pathways including PI3K-Akt, p53, ErbB and Ras signaling pathway. In addition, patients harboring LRP1B, TP53, and ErbB family mutations presented significantly higher tumor mutation burden. Conclusions: These findings demonstrated that ctDNA testing by NGS was feasible in revealing genomic changes and could be a viable alternative to tissue biopsy in patients with metastatic BTC. C1 [Chen, Chen] Hunan Provincial People’s Hospital, Department of Hepatobiliary and Pancreatic SurgeryChangsha, China. [Wang, Tao] Hunan Provincial People’s Hospital, Department of Hepatobiliary and Pancreatic SurgeryChangsha, China. [Yang, Mengmei] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Song, Jia] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Huang, Mengli] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Bai, Yuezong] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Su, Hao] First Affiliated Hospital of Guangxi Medical University, Department of Hepatobiliary SurgeryNanning, China. RP Su, H (reprint author), First Affiliated Hospital of Guangxi Medical University, Department of Hepatobiliary Surgery, Nanning, China. EM tntboy1982@163.com CR Goodman MT, and Yamamoto J. Descriptive Study of Gallbladder, Extrahepatic Bile Duct, and Ampullary Cancers in the United States, 1997- 2002. Cancer Causes Control, 2007, 18(4):415–22., DOI 10.1007/s10552-006- 0109-4 Fornaro L, Cereda S, Aprile G, Di Girolamo S, Santini D, Silvestris N, et al. Multivariate Prognostic Factors Analysis for Second-Line Chemotherapy in Advanced Biliary Tract Cancer. Br J Cancer, 2014, 110(9):2165–9., DOI 10.1038/bjc.2014.190 Jepsen P, Vilstrup H, Tarone RE, Friis S, and Sorensen HT. Incidence Rates of Intra- and Extrahepatic Cholangiocarcinomas in Denmark from 1978 through 2002. JNCI J Natl Cancer Inst, 2007, 99(11):895–7., DOI 10.1093/ jnci/djk201 Shaib YH, Davila JA, McGlynn K, and El-Serag HB. Rising Incidence of Intrahepatic Cholangiocarcinoma in the United States: a True Increase. J Hepatol, 2004, 40(3):472–7., DOI 10.1016/j.jhep.2003.11.030 Athauda A, Fong C, Lau DK, Javle M, Abou-Alfa GK, Morizane C, et al. Broadening the Therapeutic Horizon of Advanced Biliary Tract Cancer through Molecular Characterisation. Cancer Treat Rev, 2020, 86:101998., DOI 10.1016/j.ctrv.2020.101998 Hezel AF, and Zhu AX. Systemic Therapy for Biliary Tract Cancers. Oncol, 2008, 13(4):415–23., DOI 10.1634/theoncologist.2007-0252 Wang SJ, Lemieux A, Kalpathy-Cramer J, Ord CB, Walker GV, Fuller CD, et al. Nomogram for Predicting the Benefit of Adjuvant Chemoradiotherapy for Resected Gallbladder Cancer. Jco, 2011, 29(35):4627–32., DOI 10.1200/ jco.2010.33.8020 Wang Y, Li J, Xia Y, Gong R,Wang K, Yan Z, et al. Prognostic Nomogram for Intrahepatic Cholangiocarcinoma after Partial Hepatectomy. Jco, 2013, 31(9): 1188–95., DOI 10.1200/jco.2012.41.5984 Malka D, Cervera P, Foulon S, Trarbach T, de la Fouchardiere C, Boucher E, et al. Gemcitabine and Oxaliplatin with or without Cetuximab in Advanced Biliary-Tract Cancer, BINGO): a Randomised, Open-Label, Non-comparative Phase 2 Trial. Lancet Oncol, 2014, 15(8):819–28., DOI 10.1016/s1470-2045(14, 70212-8 Nakamura H, Arai Y, Totoki Y, Shirota T, Elzawahry A, Kato M, et al. Genomic Spectra of Biliary Tract Cancer. Nat Genet, 2015, 47(9):1003–10., DOI 10.1038/ng.3375 Jusakul A, Cutcutache I, Yong CH, Lim JQ, Huang MN, Padmanabhan N, et al. Whole-Genome and Epigenomic Landscapes of Etiologically Distinct Subtypes of Cholangiocarcinoma. Cancer Discov, 2017, 7(10):1116–35., DOI 10.1158/2159-8290.cd-17-0368 Abou-Alfa GK, Macarulla T, Javle MM, Kelley RK, Lubner SJ, Adeva J, et al. Ivosidenib in IDH1-Mutant, Chemotherapy-Refractory Cholangiocarcinoma, ClarIDHy): a Multicentre, Randomised, Double-Blind, Placebo-Controlled, Phase 3 Study. Lancet Oncol, 2020, 21(6):796–807., DOI 10.1016/s1470- 2045(20)30157-1 Abou-Alfa GK, Sahai V, Hollebecque A, Vaccaro G, Melisi D, Al-Rajabi R, et al. Pemigatinib for Previously Treated, Locally Advanced or Metastatic Cholangiocarcinoma: a Multicentre, Open-Label, Phase 2 Study. Lancet Oncol, 2020, 21(5):671–84., DOI 10.1016/s1470-2045(20, 30109-1 Hiley C, de Bruin EC, McGranahan N, and Swanton C. Deciphering Intratumor Heterogeneity and Temporal Acquisition of Driver Events to Refine Precision Medicine. Genome Biol, 2014, 15(8):453., DOI 10.1186/ s13059-014-0453-8 Gerlinger M, Rowan AJ, Horswell S, Math M, Larkin J, Endesfelder D, et al. Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing. N Engl J Med, 2012, 366(10):883–92., DOI 10.1056/ NEJMoa1113205 Chae YK, Davis AA, Jain S, Santa-Maria C, Flaum L, Beaubier N, et al. Concordance of Genomic Alterations by Next-Generation Sequencing in Tumor Tissue versus Circulating Tumor DNA in Breast Cancer. Mol Cancer Ther, 2017, 16(7):1412–20., DOI 10.1158/1535-7163.mct-17- 0061 Kinugasa H, Nouso K, Ako S, Dohi C, Matsushita H, Matsumoto K, et al. Liquid Biopsy of Bile for the Molecular Diagnosis of Gallbladder Cancer. Cancer Biol Ther, 2018, 19(10):934–8., DOI 10.1080/ 15384047.2018.1456604 Wang Z, Duan J, Cai S, Han M, Dong H, Zhao J, et al..Assessment of Blood Tumor Mutational Burden as a Potential Biomarker for Immunotherapy in Patients with Non-small Cell Lung Cancer with Use of a Next-Generation Sequencing Cancer Gene Panel. JAMA Oncol, 2019, 5(5):696–702., DOI 10.1001/jamaoncol.2018.7098 Lowery MA, Ptashkin R, Jordan E, Berger MF, Zehir A, Capanu M, et al. Comprehensive Molecular Profiling of Intrahepatic and Extrahepatic Cholangiocarcinomas: Potential Targets for Intervention. Clin Cancer Res, 2018, 24(17):4154–61., DOI 10.1158/1078-0432.ccr-18-0078 Li M, Zhang Z, Li X, Ye J,Wu X, Tan Z, et al.Whole-exome and Targeted Gene Sequencing of Gallbladder Carcinoma Identifies Recurrent Mutations in the ErbB Pathway. Nat Genet, 2014, 46(8):872–6., DOI 10.1038/ng.3030 Zill OA, Greene C, Sebisanovic D, Siew LM, Leng J, VuM, et al. Cell-Free DNA Next-Generation Sequencing in Pancreatobiliary Carcinomas. Cancer Discov, 2015, 5(10):1040–8., DOI 10.1158/2159-8290.cd-15-0274 Zou S, Li J, Zhou H, Frech C, Jiang X, Chu JSC, et al. Mutational Landscape of Intrahepatic Cholangiocarcinoma. Nat Commun, 2014, 5:5696., DOI 10.1038/ ncomms6696 Goeppert B, Frauenschuh L, Renner M, Roessler S, Stenzinger A, Klauschen F, et al. BRAF V600E-specific Immunohistochemistry Reveals Low Mutation Rates in Biliary Tract Cancer and Restriction to Intrahepatic Cholangiocarcinoma. Mod Pathol, 2014, 27(7):1028–34., DOI 10.1038/ modpathol.2013.206 Abdel-Wahab R, and Mahamed-Ali S. Variations in DNA Repair Genomic Alterations and Tumor Mutation burden in Biliary Tract Cancer, BTC, Subtypes. ASCO, 2018, 36:263., DOI 10.1200/ jco.2018.36.4_suppl.263 Cotterchio M, Lowcock E, Bider-Canfield Z, Lemire M, Greenwood C, Gallinger S, et al. Association between Variants in Atopy-Related Immunologic Candidate Genes and Pancreatic Cancer Risk. PLoS One, 2015, 10(5):e0125273., DOI 10.1371/journal.pone.0125273 Ding D, Lou X, Hua D, YuW, Li L,Wang J, et al. Recurrent Targeted Genes of Hepatitis B Virus in the Liver Cancer Genomes Identified by a Next- Generation Sequencing-Based Approach. Plos Genet, 2012, 8(12):e1003065., DOI 10.1371/journal.pgen.1003065 Liu C-X, Li Y, Obermoeller-McCormick LM, Schwartz AL, and Bu G. The Putative Tumor Suppressor LRP1B, a Novel Member of the Low Density Lipoprotein, LDL, Receptor Family, Exhibits Both Overlapping and Distinct Properties with the LDL Receptor-Related Protein. J Biol Chem, 2001, 276(31): 28889–96., DOI 10.1074/jbc.m102727200 Chen H, Chong W, Wu Q, Yao Y, Mao M, and Wang X. Association of LRP1B Mutation with Tumor Mutation Burden and Outcomes in Melanoma and Non-small Cell Lung Cancer Patients Treated with Immune Check-Point Blockades. Front Immunol, 2019, 10:1113., DOI 10.3389/fimmu.2019.01113 Kruiswijk F, Labuschagne CF, and Vousden KH. p53 in Survival, Death and Metabolic Health: a Lifeguard with a Licence to Kill. Nat Rev Mol Cel Biol, 2015, 16(7):393–405., DOI 10.1038/nrm4007 Dong Z-Y, Zhong W-Z, Zhang X-C, Su J, Xie Z, Liu S-Y, et al. Potential Predictive Value of TP53 and KRAS Mutation Status for Response to PD-1 Blockade Immunotherapy in Lung Adenocarcinoma. Clin Cancer Res, 2017, 23(12):3012–24., DOI 10.1158/1078-0432.ccr-16-2554 Zhang Y, Shen N, Yu W, and Fu X. Tumor Mutation burden in Biliary Tract Cancers with ERBB Family or DNA Damage Repair Gene Mutations. ASCO, 2019, 37(15):e15602., DOI 10.1200/ JCO.2019.37.15_suppl.e15602 Valle JW, Lamarca A, Goyal L, Barriuso J, and Zhu AX. New Horizons for Precision Medicine in Biliary Tract Cancers. Cancer Discov, 2017, 7(9): 943–62., DOI 10.1158/2159-8290.cd-17-0245 Mody K, Kasi PM, Yang J, Surapaneni PK, Bekaii-Saab T, Ahn DH, et al. Circulating Tumor DNA Profiling of Advanced Biliary Tract Cancers. JCO Precision Oncol, 2019, 3:1–9., DOI 10.1200/po.18.00324 Goyal L, Kelley RK, Kiedrowski L, Catenacci D, Mody K, Shroff R, and Parikh A. Blood-based Genomic Profiling of Cell-free Tumor DNA, ctDNA, in Patients with Biliary Tract Cancer. In: Proceedings of the AACR-NCI-EORTC International Conference; October 26-30, 2017; Philadelphia, PA. Philadelphia, PA: Molecular Targets and Cancer Therapeutics, 2017)., DOI 10.1158/1535-7163 Ettrich TJ, Schwerdel D, Dolnik A, Beuter F, Blatte TJ, Schmidt SA, et al. Genotyping of Circulating Tumor DNA in Cholangiocarcinoma Reveals Diagnostic and Prognostic Information. Sci Rep, 2019, 9(1):13261., DOI 10.1038/s41598-019-49860-0 Yang N, Li Y, Liu Z, Qin H, Du D, Cao X, et al. The Characteristics of ctDNA Reveal the High Complexity in Matching the Corresponding Tumor Tissues. BMC Cancer, 2018, 18(1):319., DOI 10.1186/s12885-018- 4199-7 Chen K-Z, Lou F, Yang F, Zhang J-B, Ye H, ChenW, et al. Circulating Tumor DNA Detection in Early-Stage Non-small Cell Lung Cancer Patients by Targeted Sequencing. Sci Rep, 2016, 6:31985., DOI 10.1038/srep31985 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609879 EP 1609887 DI 10.3389/pore.2021.1609879 PG 9 ER PT J TI The Advance and Correlation of KRAS Mutation With the Fertility-Preservation Treatment of Endometrial Cancer in the Background of Molecular Classification Application SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE KRAS mutation; endometrial cancer; fertility-preservation; fertility-spared; conservative treatment; molecular classification ID KRAS mutation; endometrial cancer; fertility-preservation; fertility-spared; conservative treatment; molecular classification AB The annually increasing incidence of endometrial cancer in younger women has created a growing demand for fertility preservation. However, the diverse therapeutic efficacy among patients under the same histological subtype and the same tumor grade suggests the potential interference of the innate molecular characteristics. The molecular classification has now been applied in clinical practice and might help to stratify the endometrial cancer patients and individualize the therapy, but the candidates for the fertility-spared treatment are most likely to be subdivided in the subgroup lacking the specific signature. KRAS mutation has been linked to the malignant transition of the endometrium, while its role in molecular classification and fertility preservation is vague. Here, we mainly review the advance of molecular classification and the role of KRAS in endometrial cancer, as well as their correlation with fertility-preservation treatment. CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Wang XQ, Sun ZJ, Lang JH. Screening for Endometrial Cancer. Chin J Pract Gynecol Obstet, 2019, 35:1273–7., DOI 10.19538/j.fk2019110122 Sideris M, Emin EI, Abdullah Z, Hanrahan J, Stefatou KM, Sevas V, et al. The Role of KRAS in Endometrial Cancer: A Mini-Review. Anticancer Res, 2019, 39(2):533–9., DOI 10.21873/anticanres.13145 Bokhman JV. Two Pathogenetic Types of Endometrial Carcinoma. Gynecol Oncol, 1983, 15(1):10–7., DOI 10.1016/0090-8258(83)90111-7 Kim YB, Holschneider CH, Ghosh K, Nieberg RK, Montz FJ. Progestin Alone as Primary Treatment of Endometrial Carcinoma in Premenopausal Women. Cancer, 1997, 79(2):320–7., DOI 10.1002/(sici)1097-0142(19970115)79:2<320: aid-cncr15>3.0.co;2-2 Abu-RustumNR, Yashar CM, Bradley K, Campos SM, Chino J, Chon HS, et al. NCCN Guidelines Insights: Uterine Neoplasms, Version 3.2021. J Natl Compr Cancer Netw, 2021, 19(8):888–95., DOI 10.6004/jnccn.2021.0038 Zazhi ZA. Guidelines for the Diagnosis and Treatment of Endometrial Cancer, 2021 Edition). China Oncol, 2021, 31(06):501–12., DOI 10.19401/j.cnki.1007- 3639.2021.06.08 Concin N, Creutzberg CL, Vergote I, Cibula D, Mirza MR, Marnitz S, et al. ESGO/ESTRO/ESP Guidelines for the Management of Patients with Endometrial Carcinoma. Virchows Arch, 2021, 478(2):153–90., DOI 10.1007/ s00428-020-03007-z Yamagami W, Mikami M, Nagase S, Tabata T, Kobayashi Y, Kaneuchi M, et al. Japan Society of Gynecologic Oncology 2018 Guidelines for Treatment of Uterine Body Neoplasms. J Gynecol Oncol, 2020, 31(1)., DOI 10.3802/jgo.2020.31.e18 Zhou R, Lu Q, Liu GL, Wang YQ, Wang JL. Expert Consensus on Fertility- Sparing Treatment of Early Endometrial Cancer. Gynecol Obstet Clin Med, 2019, 20(04):369–73., DOI 10.1016/j.gocm.2020.10.002 Lee S-W, Lee TS, Hong DG, No JH, Park DC, Bae JM, et al. Practice Guidelines for Management of Uterine Corpus Cancer in Korea: a Korean Society of Gynecologic Oncology Consensus Statement. J Gynecol Oncol, 2017, 28(1): e12., DOI 10.3802/jgo.2017.28.e12 Shen K, Lang JH, Yang JX, Cao DQ, Wei LH, Kong BH, et al. Guidelines for Clinical Diagnosis and Treatment of Gynecological Malignant Tumors with Preserved Fertility. Chin J Obstet Gynecol, 2014, 49(04):243–8. Andress J, Pasternak J, Walter C, Kommoss S, Kramer B, Hartkopf A, et al. Fertility Preserving Management of Early Endometrial Cancer in a Patient Cohort at the Department of Women’s Health at the university of Tuebingen. Arch Gynecol Obstet, 2021, 304(1):215–21., DOI 10.1007/s00404-020-05905-8 Park J-Y, Kim D-Y, Kim J-H, Kim Y-M, Kim K-R, Kim Y-T, et al. Long-term Oncologic Outcomes after Fertility-Sparing Management Using Oral Progestin for Young Women with Endometrial Cancer, KGOG 2002). Eur J Cancer, 2013, 49(4):868–74., DOI 10.1016/j.ejca.2012.09.017 Yang B, Xu Y, Zhu Q, Xie L, Shan W, Ning C, et al. Treatment Efficiency of Comprehensive Hysteroscopic Evaluation and Lesion Resection Combined with Progestin Therapy in Young Women with Endometrial Atypical Hyperplasia and Endometrial Cancer. Gynecol Oncol, 2019, 153(1):55–62., DOI 10.1016/j.ygyno.2019.01.014 Casadio P, La Rosa M, Alletto A, Magnarelli G, Arena A, Fontana E, et al. Fertility Sparing Treatment of Endometrial Cancer with and without Initial Infiltration of Myometrium: A Single Center Experience. Cancers, 2020, 12(12):3571., DOI 10.3390/cancers12123571 Levine DA, Schultz N, Cherniack AD, Akbani R, Liu Y, Shen H, et al. Integrated Genomic Characterization of Endometrial Carcinoma. Nature, 2013, 497(7447):67–73., DOI 10.1038/nature12113 Talhouk A, McConechyMK, Leung S, Li-ChangHH,Kwon JS, Melnyk N, et al.A Clinically Applicable Molecular-Based Classification for Endometrial Cancers. Br J Cancer, 2015, 113(2):299–310., DOI 10.1038/bjc.2015.190 Talhouk A, McConechy MK, Leung S, Yang W, Lum A, Senz J, et al. Confirmation of ProMisE: A Simple, Genomics-Based Clinical Classifier for Endometrial Cancer. Cancer, 2017, 123(5):802–13., DOI 10.1002/ cncr.30496 Kommoss S, McConechy MK, Kommoss F, Leung S, Bunz A, Magrill J, et al. Final Validation of the ProMisE Molecular Classifier for Endometrial Carcinoma in a Large Population-Based Case Series. Ann Oncol, 2018, 29(5):1180–8., DOI 10.1093/annonc/mdy058 Travaglino A, Raffone A, Saccone G, Insabato L, Mollo A, De Placido G, et al. Immunohistochemical Predictive Markers of Response to Conservative Treatment of Endometrial Hyperplasia and Early Endometrial Cancer: A Systematic Review. Acta Obstet Gynecol Scand, 2019, 98(9):1086–99., DOI 10.1111/aogs.13587 Falcone F, Normanno N, Losito NS, Scognamiglio G, Esposito Abate R, Chicchinelli N, et al. Application of the Proactive Molecular Risk Classifier for Endometrial Cancer, ProMisE, to Patients Conservatively Treated: Outcomes from an Institutional Series. Eur J Obstet Gynecol Reprod Biol, 2019, 240:220–5., DOI 10.1016/j.ejogrb.2019.07.013 Chung YS, Woo HY, Lee J-Y, Park E, Nam EJ, Kim S, et al. Mismatch Repair Status Influences Response to Fertility-Sparing Treatment of Endometrial Cancer. Am J Obstet Gynecol, 2021, 224(4):e1–370., DOI 10.1016/ j.ajog.2020.10.003 Puechl AM, Spinosa D, Berchuck A, Secord AA, Drury KE, Broadwater G, et al. Molecular Classification to Prognosticate Response in Medically Managed Endometrial Cancers and Endometrial Intraepithelial Neoplasia. Cancers, 2021, 13(11):2847., DOI 10.3390/cancers13112847 Dong P, Xiong Y, Yue J, Hanley S. J. B., Koboyashi N, Todo Y, et al. Exploring lncRNA-Mediated Regulatory Networks in Endometrial Cancer Cells and the Tumor Microenvironment: Advances and Challenges. Cancers, 2019, 11(2): 234., DOI 10.3390/cancers11020234 Chi S, Liu Y, Zhou X, Feng D, Xiao X, LiW, et al. Knockdown of Long Noncoding HOTAIR Enhances the Sensitivity to Progesterone in Endometrial Cancer by Epigenetic Regulation of Progesterone Receptor Isoform B. Cancer Chemother Pharmacol, 2019, 83(2):277–87., DOI 10.1007/s00280- 018-3727-0 Wang W, Ge L, Xu X-J, Yang T, Yuan Y, Ma X-L, et al. LncRNA NEAT1 Promotes Endometrial Cancer Cell Proliferation, Migration and Invasion by Regulating the miR-144-3p/EZH2 axis. Radiol Oncol, 2019, 53(4):434–42., DOI 10.2478/raon-2019-0051 Malumbres M, Barbacid M. RAS Oncogenes: the First 30 Years. Nat Rev Cancer, 2003, 3(6):459–65., DOI 10.1038/nrc1097 Liu P,Wang Y, Li X. Targeting the Untargetable KRAS in Cancer Therapy. Acta Pharmaceutica Sinica B, 2019, 9(5):871–9., DOI 10.1016/j.apsb.2019.03.002 Boguski MS, McCormick F. Proteins Regulating Ras and its Relatives. Nature, 1993, 366(6456):643–54., DOI 10.1038/366643a0 Donovan S, Shannon KM, Bollag G. GTPase Activating Proteins: Critical Regulators of Intracellular Signaling. Biochim Biophys Acta, Bba, - Rev Cancer, 2002, 1602(1):23–45., DOI 10.1016/s0304-419x(01)00041-5 Schubbert S, Shannon K, Bollag G. Hyperactive Ras in Developmental Disorders and Cancer. Nat Rev Cancer, 2007, 7(4):295–308., DOI 10.1038/ nrc2109 Indini A, Rijavec E, Ghidini M, Cortellini A, Grossi F. Targeting KRAS in Solid Tumors: Current Challenges and Future Opportunities of Novel KRAS Inhibitors. Pharmaceutics, 2021, 13(5):653., DOI 10.3390/pharmaceutics13050653 Vetter IR, Wittinghofer A. The Guanine Nucleotide-Binding Switch in Three Dimensions. Science, 2001, 294(5545):1299–304., DOI 10.1126/science.1062023 Jancik S, Drabek J, Radzioch D, HajduchM. Clinical Relevance of KRAS in Human Cancers. J Biomed Biotechnol, 2010, 2010:1–13., DOI 10.1155/2010/150960 Wittinghofer A, Vetter IR. Structure-function Relationships of the G Domain, a Canonical Switch Motif. Annu Rev Biochem, 2011, 80:943–71., DOI 10.1146/ annurev-biochem-062708-134043 Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D. RAS Oncogenes: Weaving a Tumorigenic Web. Nat Rev Cancer, 2011, 11(11):761–74., DOI 10.1038/ nrc3106 Gideon P, John J, Frech M, Lautwein A, Clark R, Scheffler JE, et al. Mutational and Kinetic Analyses of the GTPase-Activating Protein, GAP)-p21 Interaction: the C-Terminal Domain of GAP Is Not Sufficient for Full Activity. Mol Cel Biol, 1992, 12(5):2050–6., DOI 10.1128/mcb.12.5.2050-2056.1992 Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. Biochemical and Structural Analysis of Common Cancer-Associated KRAS Mutations. Mol Cancer Res, 2015, 13(9):1325–35., DOI 10.1158/1541- 7786.Mcr-15-0203 Bos JL, Rehmann H,Wittinghofer A. GEFs and GAPs: Critical Elements in the Control of Small G Proteins. Cell, 2007, 129(5):865–77., DOI 10.1016/ j.cell.2007.05.018 SmithMJ, Neel BG, Ikura M. NMR-based Functional Profiling of RASopathies and Oncogenic RAS Mutations. Proc Natl Acad Sci, 2013, 110(12):4574–9., DOI 10.1073/pnas.1218173110 Cook JH, Melloni GEM, Gulhan DC, Park PJ, Haigis KM. The Origins and Genetic Interactions of KRAS Mutations Are Allele- and Tissue-specific. Nat Commun, 2021, 12(1):1808., DOI 10.1038/s41467-021-22125-z Veluswamy R, Mack PC, Houldsworth J, Elkhouly E, Hirsch FR. KRAS G12CMutant Non-small Cell Lung Cancer. J Mol Diagn, 2021, 23(5):507–20., DOI 10.1016/j.jmoldx.2021.02.002 Ihle NT, Byers LA, Kim ES, Saintigny P, Lee JJ, Blumenschein GR, et al. Effect of KRAS Oncogene Substitutions on Protein Behavior: Implications for Signaling and Clinical Outcome. J Natl Cancer Inst, 2012, 104(3):228–39., DOI 10.1093/jnci/djr523 Yang H, Liang S-Q, Schmid RA, Peng R-W. New Horizons in KRAS-Mutant Lung Cancer: Dawn after Darkness. Front Oncol, 2019, 9:953., DOI 10.3389/ fonc.2019.00953 Banno K, Yanokura M, Iida M, Masuda K, Aoki D. Carcinogenic Mechanisms of Endometrial Cancer: Involvement of Genetics and Epigenetics. J Obstet Gynaecol Res, 2014, 40(8):1957–67., DOI 10.1111/jog.12442 Hung P-S, Huang M-H, Kuo Y-Y, Yang JC-H. The Inhibition of Wnt Restrain KRASG12V-Driven Metastasis in Non-small-cell Lung Cancer. Cancers, 2020, 12(4):837., DOI 10.3390/cancers12040837 Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, et al. The Clinical KRAS(G12C, Inhibitor AMG 510 Drives Anti-tumour Immunity. Nature, 2019, 575(7781):217–23., DOI 10.1038/s41586-019-1694-1 Hobbs GA, Baker NM, Miermont AM, Thurman RD, Pierobon M, Tran TH, et al. Atypical KRASG12R Mutant Is Impaired in PI3K Signaling and Macropinocytosis in Pancreatic Cancer. Cancer Discov, 2020, 10(1):104–23., DOI 10.1158/2159-8290.CD-19-1006 Reck M, Carbone DP, Garassino M, Barlesi F. Targeting KRAS in Non-smallcell Lung Cancer: Recent Progress and New Approaches. Ann Oncol, 2021, 32: 1101–10., DOI 10.1016/j.annonc.2021.06.001 Meng M, Zhong K, Jiang T, Liu Z, Kwan HY, Su T. The Current Understanding on the Impact of KRAS on Colorectal Cancer. Biomed Pharmacother, 2021, 140:111717., DOI 10.1016/j.biopha.2021.111717 Yarla NS, Madka V, Pathuri G, Rao CV. Molecular Targets in Precision Chemoprevention of Colorectal Cancer: An Update from Pre-clinical to Clinical Trials. Ijms, 2020, 21(24):9609., DOI 10.3390/ijms21249609 Jones NL, Xiu J, Chatterjee-Paer S, Buckley de Meritens A, Burke WM, Tergas AI, et al. Distinct Molecular Landscapes between Endometrioid and Nonendometrioid Uterine Carcinomas. Int J Cancer, 2017, 140(6): 1396–404., DOI 10.1002/ijc.30537 Byron SA, Gartside M, Powell MA, Wellens CL, Gao F, Mutch DG, et al. FGFR2 point Mutations in 466 Endometrioid Endometrial Tumors: Relationship with MSI, KRAS, PIK3CA, CTNNB1 Mutations and Clinicopathological Features. PLoS One, 2012, 7(2):e30801., DOI 10.1371/ journal.pone.0030801 McConechy MK, Ding J, Cheang MC, Wiegand KC, Senz J, Tone AA, et al. Use of Mutation Profiles to Refine the Classification of Endometrial Carcinomas. J Pathol, 2012, 228(1):20–30., DOI 10.1002/path.4056 Levine DA, Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated Genomic Characterization of Endometrial Carcinoma. Nature, 2013, 497(7447):67–73., DOI 10.1038/nature12113 Shintani D, Hihara T, Ogasawara A, Sato S, Yabuno A, Tai K, et al. Tumorrelated Mutations in Cell-free DNA in Pre-operative Plasma as a Prognostic Indicator of Recurrence in Endometrial Cancer. Int J Gynecol Cancer, 2020, 30(9):1340–6., DOI 10.1136/ijgc-2019-001053 Zauber P, Denehy TR, Taylor RR, Ongcapin EH, Marotta S, Sabbath-Solitare M. Strong Correlation between Molecular Changes in Endometrial Carcinomas and Concomitant Hyperplasia. Int J Gynecol Cancer, 2015, 25(5):863–8., DOI 10.1097/igc.0000000000000421 Dobrzycka B, Terlikowski SJ, Mazurek A, Kowalczuk O, Niklinska W, Chyczewski L, et al. Circulating Free DNA, P53 Antibody and Mutations of KRAS Gene in Endometrial Cancer. Int J Cancer, 2010, 127(3):612–21., DOI 10.1002/ijc.25077 Watanabe T, Nanamiya H, Kojima M,Nomura S, Furukawa S, Soeda S, et al.Clinical Relevance of Oncogenic Driver Mutations Identified in Endometrial Carcinoma. Translational Oncol, 2021, 14(3):101010., DOI 10.1016/j.tranon.2021.101010 Singh H, Longo DL, Chabner BA. Improving Prospects for Targeting RAS. Jco, 2015, 33(31):3650–9., DOI 10.1200/jco.2015.62.1052 Duggan BD, Felix JC, Muderspach LI, Tsao JL, Shibata DK. Early Mutational Activation of the C-Ki-Ras Oncogene in Endometrial Carcinoma. Cancer Res, 1994, 54(6):1604–7. Wang Y, Yu M, Yang J-X, Cao D-Y, Zhang Y, Zhou H-M, et al. Genomic Comparison of Endometrioid Endometrial Carcinoma and its Precancerous Lesions in Chinese Patients by High-Depth Next Generation Sequencing. Front Oncol, 2019, 9:123., DOI 10.3389/fonc.2019.00123 Alomari A, Abi-Raad R, Buza N, Hui P. Frequent KRAS Mutation in Complex Mucinous Epithelial Lesions of the Endometrium. Mod Pathol, 2014, 27(5): 675–80., DOI 10.1038/modpathol.2013.186 Xiong J, He M, Hansen K, Jackson CL, Breese V, Quddus MR, et al. The Clinical Significance of K-Ras Mutation in Endometrial "surface Epithelial Changes" and Their Associated Endometrial Adenocarcinoma. Gynecol Oncol, 2016, 142(1):163–8., DOI 10.1016/j.ygyno.2016.05.001 van der Putten LJM, van Hoof R, Tops BBJ, Snijders MPLM, van den Berg-van Erp SH, van der Wurff AAM, et al. Molecular Profiles of Benign and, Pre, malignant Endometrial Lesions. Carcinogenesis, 2017, 38(3):329–35., DOI 10.1093/carcin/bgx008 Sideris M, Moorhead J, Diaz-Cano S, Haji A, Papagrigoriadis S. KRAS Mutant Status May Be Associated with Distant Recurrence in Early-Stage Rectal Cancer. Ar, 2017, 37(3):1349–58., DOI 10.21873/anticanres.11454 Ryan BM, Robles AI, Harris CC. KRAS-LCS6 Genotype as a Prognostic Marker in Early-Stage CRC-Letter. Clin Cancer Res, 2012, 18(12):3487–8. author reply 9., DOI 10.1158/1078-0432.Ccr-12-0250 Zheng XZ, Zhan Y, Ma JH, Xu HW, ZhiWX, Chen TB, et al. KRAS Mutations Analysis in Mucinous Epithelial Lesions of the Endometrium. Zhonghua Bing Li Xue Za Zhi, 2018, 47(9):687–90., DOI 10.3760/cma.j.issn.0529-5807.2018.09.007 Feng Y-Z, Shiozawa T, Miyamoto T, Kashima H, Kurai M, Suzuki A, et al. BRAF Mutation in Endometrial Carcinoma and Hyperplasia: Correlation with KRAS and P53 Mutations and Mismatch Repair Protein Expression. Clin Cancer Res, 2005, 11(17):6133–8., DOI 10.1158/1078-0432.ccr-04-2670 D’Angelo E, Espinosa I, Cipriani V, Szafranska J, Barbareschi M, Prat J. Atypical Endometrial Hyperplasia, Low-Grade. Am J Surg Pathol, 2021, 45(7): 988–96., DOI 10.1097/pas.0000000000001705 Tsuda H, Jiko K, Yajima M, Yamada T, Tanemura K, Tsunematsu R, et al. Frequent Occurrence of C-Ki-Ras Gene Mutations in Well Differentiated Endometrial Adenocarcinoma Showing Infiltrative Local Growth with Fibrosing Stromal Response. Int J Gynecol Pathol, 1995, 14(3):255–9., DOI 10.1097/00004347-199507000-00010 Birkeland E, Wik E, Mjos S, Hoivik EA, Trovik J, Werner HMJ, et al. KRAS Gene Amplification and Overexpression but Not Mutation Associates with Aggressive and Metastatic Endometrial Cancer. Br J Cancer, 2012, 107(12): 1997–2004., DOI 10.1038/bjc.2012.477 Berg A, Hoivik EA, Mjos S, Holst F, Werner HMJ, Tangen IL, et al. Molecular Profiling of Endometrial Carcinoma Precursor, Primary and Metastatic Lesions Suggests Different Targets for Treatment in Obese Compared to Non-obese Patients. Oncotarget, 2015, 6(2):1327–39., DOI 10.18632/oncotarget.2675 Lu KH, Broaddus RR. Endometrial Cancer. N Engl J Med, 2020, 383(21): 2053–64., DOI 10.1056/NEJMra1514010 Ring KL, Yates MS, Schmandt R, Onstad M, Zhang Q, Celestino J, et al. Endometrial Cancers with Activating KRas Mutations Have Activated Estrogen Signaling and Paradoxical Response to MEK Inhibition. Int J Gynecol Cancer, 2017, 27(5):854–62., DOI 10.1097/IGC.0000000000000960 Knez J, Al Mahdawi L, Takac I, Sobocan M. The Perspectives of Fertility Preservation in Women with Endometrial Cancer. Cancers, 2021, 13(4):602., DOI 10.3390/cancers13040602 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609906 EP 1609911 DI 10.3389/pore.2021.1609906 PG 6 ER PT J AU Reedy, M Jonnalagadda, Sh Palle, K AF Reedy, Mark Jonnalagadda, Shirisha Palle, Komaraiah TI Case Report: Intra-Tumoral Vaccinations of Quadrivalent HPV-L1 Peptide Vaccine With Topical TLR-7 Agonist Following Recurrence: Complete Resolution of HPV-HR-Associated Gynecologic Squamous Cell Carcinomas in Two Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE human papilloma virus high-risk variant; radiation refractory squamous cell carcinoma; quadrivalent HPV-L1 vaccine; intratumoral injection; toll-like receptor 7 agonist; immunotherapy ID human papilloma virus high-risk variant; radiation refractory squamous cell carcinoma; quadrivalent HPV-L1 vaccine; intratumoral injection; toll-like receptor 7 agonist; immunotherapy AB The human papilloma virus (HPV) high-risk variants (HPV-HR) such as HPV16 and HPV18 are responsible for most HPV related cancers, including anogenital and head and neck cancers. Here, we present two patients with HPV-HR-associated gynecological malignancies who, after failing radiation therapy, were treated with experimental salvage immunotherapy regimen resulting in complete, durable responses in both patients. Each patient was diagnosed with recurrent, radiation-refractory, HPV-HR positive, squamous cell carcinoma of the lower genital tract. Patient A was a 90-yearold, African American, with metastatic vulvar cancer to the right inguinal-femoral triangle and pulmonary metastases. Patient B was a 41-year-old, Caucasian, with a centralrecurrence of cervix cancer. Each patient received at least two intratumoral quadrivalent HPV-L1 vaccine (Gardasil™) injections and daily topical TLR-7 agonist (imiquimod) to the tumor surface 2 weeks apart. This combination of intratumoral vaccinations and topical TLR-7 agonist produced unexpected complete resolution of disease in both patients. The importance of radiation therapy, despite being considered a treatment failure by current definitions, cannot be understated. Radiation therapy appears to have offered a therapeutic immune advantage by modifying the tumor microenvironment. This immune protocol has potential to help patients with advanced HPV-HR-related malignancies previously considered incurable. C1 [Reedy, Mark] Texas Tech University Health Sciences Center, School of Medicine, Department of Obstetrics and GynecologyLubbock, TX, USA. [Jonnalagadda, Shirisha] Texas Tech University Health Sciences Center, Department of Cell Biology and BiochemistryLubbock, TX, USA. [Palle, Komaraiah] Texas Tech University Health Sciences Center, Department of Cell Biology and BiochemistryLubbock, TX, USA. RP Reedy, M (reprint author), Texas Tech University Health Sciences Center, School of Medicine, Department of Obstetrics and Gynecology, Lubbock, USA. EM Mark.Reedy@ttuhsc.edu CR Bellone S, El-Sahwi K, Cocco E, Casagrande F, Cargnelutti M, Palmieri M, et al. Human Papillomavirus Type 16, HPV-16, Virus-like Particle L1-specific CD8 + Cytotoxic T Lymphocytes, CTLs, Are Equally Effective as E7-specific CD8 + CTLs in Killing Autologous HPV-16-Positive Tumor Cells in Cervical Cancer Patients: Implications for L1 Dendritic Cell-Based Therapeutic Vaccines. J Virol, 2009, 83:6779–89., DOI 10.1128/JVI.02443-08 Chuang C-M, Monie A, Hung C-F, Wu T-C. Treatment with Imiquimod Enhances Antitumor Immunity Induced by Therapeutic HPV DNA Vaccination. J Biomed Sci, 2010, 17:32., DOI 10.1186/1423-0127-17-32 Cadena A, Cushman T, Anderson C, Barsoumian H, Welsh J, Cortez M. Radiation and Anti-cancer Vaccines: A Winning Combination. Vaccines, 2018, 6:9., DOI 10.3390/vaccines6010009 Cho JH, Lee H-J, Ko H-J, Yoon B-I, Choe J, Kim K-C, et al. The TLR7 Agonist Imiquimod Induces Anti-cancer Effects via Autophagic Cell Death and Enhances Anti-tumoral and Systemic Immunity during Radiotherapy for Melanoma. Oncotarget, 2017, 8:24932–48., DOI 10.18632/oncotarget.15326 Papadavid E, Stratigos AJ, Falagas ME. Imiquimod: an Immune Response Modifier in the Treatment of Precancerous Skin Lesions and Skin Cancer. Expert Opin Pharmacother, 2007, 8:1743–55., DOI 10.1517/14656566.8.11.1743 Green DS, Dalgleish AG, Belonwu N, Fischer MD, Bodman-SmithMD. Topical Imiquimod and Intralesional Interleukin-2 Increase Activated Lymphocytes and Restore the Th1/Th2 Balance in Patients with Metastatic Melanoma. Br J Dermatol, 2008, 159:606–14., DOI 10.1111/j.1365-2133.2008.08709.x McKee SJ, Bergot A-S, Leggatt GR. Recent Progress in Vaccination against Human Papillomavirus-Mediated Cervical Cancer. Rev Med Virol, 2015, 25: 54–71., DOI 10.1002/rmv.1824 Daayana S, Elkord E, Winters U, Pawlita M, Roden R, Stern PL, et al. Phase II Trial of Imiquimod and HPV Therapeutic Vaccination in Patients with Vulval Intraepithelial Neoplasia. Br J Cancer, 2010, 102:1129–36., DOI 10.1038/ sj.bjc.6605611 Murray ML,Meadows J, Dore CJ, Copas AJ, Haddow LJ, Lacey C, et al. Human Papillomavirus Infection: Protocol for a Randomised Controlled Trial of Imiquimod Cream, 5%, versus Podophyllotoxin Cream, 0.15%), in Combination with Quadrivalent Human Papillomavirus or Control Vaccination in the Treatment and Prevention of Recurrence of Anogenital Warts, HIPvac Trial). BMC Med Res Methodol, 2018, 18:125., DOI 10.1186/ s12874-018-0581-z Nichols AJ, Gonzalez A, Clark ES, Khan WN, Rosen AC, Guzman W, et al. Combined Systemic and Intratumoral Administration of Human Papillomavirus Vaccine to Treat Multiple Cutaneous Basaloid Squamous Cell Carcinomas. JAMA Dermatol, 2018, 154:927–30., DOI 10.1001/ jamadermatol.2018.1748 Kreiter S, Vormehr M, van de Roemer N, Diken M, Lower M, Diekmann J, et al. Mutant MHC Class II Epitopes Drive Therapeutic Immune Responses to Cancer. Nature, 2015, 520:692–6., DOI 10.1038/nature14426 Garnett-Benson C, Hodge JW, Gameiro SR. Combination Regimens of Radiation Therapy and Therapeutic Cancer Vaccines: Mechanisms and Opportunities. Semin Radiat Oncol, 2015, 25:46–53., DOI 10.1016/ j.semradonc.2014.07.002 Kaur P, Asea A. Radiation-induced Effects and the Immune System in Cancer. Front Oncol, 2012, 2:191., DOI 10.3389/fonc.2012.00191 Huang W, Chen J-J, Xing R, Zeng Y-C. Combination Therapy: Future Directions of Immunotherapy in Small Cell Lung Cancer. Translational Oncol, 2021, 14:100889., DOI 10.1016/j.tranon.2020.100889 De Mattos-Arruda L, VazquezM, Finotello F, Lepore R, Porta E, Hundal J, et al. Neoantigen Prediction and Computational Perspectives towards Clinical Benefit: Recommendations from the ESMO Precision Medicine Working Group. Ann Oncol, 2020, 31:978–90., DOI 10.1016/ j.annonc.2020.05.008 Ng C-P, Bonavida B. A New challenge for Successful Immunotherapy by Tumors that Are Resistant to Apoptosis: Two Complementary Signals to Overcome Cross-Resistance. Adv Cancer Res, 2002, 85:145–74., DOI 10.1016/ s0065-230x(02)85005-9 Vasan N, Baselga J, Hyman DM. A View on Drug Resistance in Cancer. Nature, 2019, 575:299–309., DOI 10.1038/s41586-019-1730-1 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609922 EP 1609927 DI 10.3389/pore.2021.1609922 PG 6 ER PT J AU Yao, ZG Wei, ZG Cheng, XK Huang, GH Zong, YY Meng, M Li, JM Han, XY Xu, JW Wang, J Jing, HY Li, WH Cao, ZX Ni, Y Sun, XCh Yang, X Ye, X AF Yao, Zhi-Gang Wei, Zhi-Gang Cheng, Xian-Kui Huang, Guang-Hui Zong, Yuan-Yuan Meng, Min Li, Jia-Mei Han, Xiao-Ying Xu, Jia-Wen Wang, Jiao Jing, Hai-Yan Li, Wen-Hong Cao, Zhi-Xin Ni, Yang Sun, Xi-Chao Yang, Xia Ye, Xin TI Comparison of Multi-Gene Testing Data Between Fresh and Formalin-Fixed Specimens From Core Needle Biopsy in Patients With NSCLC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE pathology; EGFR; lung cancers; frozen section examination; fresh frozen tissue ID pathology; EGFR; lung cancers; frozen section examination; fresh frozen tissue AB Purpose: Currently, formalin-fixed paraffin-embedded (FFPE) tissue specimens are the conventional material for gene testing for non-small cell lung cancer (NSCLC) patients. In our study, we aimed to develop a quick gene testing procedure using fresh core needle biopsy samples from NSCLC patients. Methods: In total, 77 fresh NSCLC samples obtained from core needle biopsy were evaluated by frozen section examination. If the NSCLC diagnosis and adequate tumor cell counts were confirmed by histopathology, the fresh tissues were used to extract DNA and subsequent gene testing by ARMS-PCR. Meanwhile, the paired FFPE core needle biopsy samples from 30 NSCLC patients also underwent gene testing. Results: In total, 77 fresh samples showed an EGFR mutation rate of 61.0%, higher than the levels in the Asian. Following a comparison of gene testing results with fresh tissues and paired FFPE tissues from the 30 patients, no significant difference in the DNA concentration extracted from fresh tissues and FFPE tissues was found. However, DNA purity was significantly higher in fresh tissues than that in FFPE tissues. Gene testing detected the same gene mutations in 93.3% of cases in fresh tissues and paired FFPE tissues. The gene testing procedure using fresh biopsy samples greatly shortens the waiting time of patients. Conclusion: The multi-gene mutation testing using fresh core needle biopsy samples from NSCLC patients is a reasonable, achievable, and quick approach. Fresh tissues may serve as a potential alternative to FFPE tissues for gene testing in NSCLC patients. C1 [Yao, Zhi-Gang] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Wei, Zhi-Gang] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. [Cheng, Xian-Kui] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Huang, Guang-Hui] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. [Zong, Yuan-Yuan] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Meng, Min] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. [Li, Jia-Mei] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Han, Xiao-Ying] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. [Xu, Jia-Wen] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Wang, Jiao] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. [Jing, Hai-Yan] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Li, Wen-Hong] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. [Cao, Zhi-Xin] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Ni, Yang] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. [Sun, Xi-Chao] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Yang, Xia] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. [Ye, Xin] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. RP Ye, X (reprint author), Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Jinan, China. EM yexintaian2020@163.com CR Valentino F, Borra G, Allione P, Rossi L. Emerging Targets in AdvancedNon-Small- Cell Lung Cancer. Future Oncol, 2018, 14(13s):61–72., DOI 10.2217/fon-2018-0099 Chen C-M, Chang JW-C, Cheung Y-C, Lin G, Hsieh J-J, Hsu T, et al. Computed Tomography-Guided Core-Needle Biopsy Specimens Demonstrate Epidermal Growth Factor Receptor Mutations in Patients with Non-Small-Cell Lung Cancer. Acta Radiol, 2008, 49(9):991–4., DOI 10.1080/02841850802294410 Lai Y, Zhang Z, Li J, Sun D, Zhou YA, Jiang T, et al. EGFR Mutations in Surgically Resected Fresh Specimens from 697 Consecutive Chinese Patients with Non-Small Cell Lung Cancer and Their Relationships with Clinical Features. Int J Mol Sci, 2013, 14(12):24549–59., DOI 10.3390/ijms141224549 Ye X, Wang J, Ni Y, Yang X, Huang G, Wei Z, et al. Diagnostic Ability of Percutaneous Core Biopsy Immediately after Microwave Ablation for Lung Ground-Glass Opacity. J Can Res Ther, 2019, 15(4):755–9., DOI 10.4103/ jcrt.JCRT_399_19 Wu S, Shi X, Si X, Liu Y, Lu T, Zhang L, et al. EGFR T790M Detection in Formalin-Fixed Paraffin-Embedded Tissues of Patients with Lung Cancer Using RNA-Based In Situ Hybridization: A Preliminary Feasibility Study. Thorac Cancer, 2019, 10(10):1936–44., DOI 10.1111/1759-7714.13169 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 ChenW, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer Statistics in China, 2015. CA: A Cancer J Clinicians, 2016, 66(2):115–32., DOI 10.3322/caac.21338 Zhou C. Lung Cancer Molecular Epidemiology in China: Recent Trends. Transl Lung Cancer Res, 2014, 3(5):270–9., DOI 10.3978/j.issn.2218-6751.2014.09.01 Wu F, Wang L, Zhou C. Lung Cancer in China: Current and Prospect. Curr Opin Oncol, 2021, 33(1):40–6., DOI 10.1097/CCO.0000000000000703 Chinese Association of Oncologists. Chinese Society for Clinical Cancer Chemotherapy, Chinese Association of Oncologists Chinese Society for Clinical Cancer Chemotherapy. The Guideline for Diagnosis and Treatment of Chinese Patients with Sensitizing EGFR Mutation or ALK Fusion Gene-Positive Non-small Cell Lung Cancer, 2015 Version). Zhonghua Zhong Liu Za Zhi, 2015, 37(10):796–9., DOI 10.3760/ cma.j.issn.0253-3766.2015.10.018 Campos PF, Gilbert TMP. DNA Extraction from Formalin-Fixed Material. Methods Mol Biol, 2012, 840:81–5., DOI 10.1007/978-1-61779-516-9_11 Ulivi P, Romagnoli M, Chiadini E, Casoni GL, Capelli L, Gurioli C. Assessment of EGFR and K-Ras Mutations in Fixed and Fresh Specimens from Transesophageal Ultrasound-Guided Fine Needle Aspiration in Non-Small Cell Lung Cancer Patients. Int J Oncol, 2012, 41(1):147–52., DOI 10.3892/ ijo.2012.1432 Suciu BA, Pap Z, Denes L, Brinzaniuc K, Copotoiu C, Pavai Z. Allele-Specific PCR Method for Identification of EGFR Mutations in Non-Small Cell Lung Cancer: Formalin-Fixed Paraffin-Embedded Tissue Versus Fresh Tissue. Rom J Morphol Embryol, 2016, 57(2):495–500. Spencer DH, Sehn JK, Abel HJ, Watson MA, Pfeifer JD, Duncavage EJ. Comparison of Clinical Targeted Next-Generation Sequence Data from Formalin-Fixed and Fresh-Frozen Tissue Specimens. J Mol Diagn, 2013, 15(5):623–33., DOI 10.1016/j.jmoldx.2013.05.004 Cheung Y-C, Chang JW-C, Hsieh J-J, Lin G, Tsai Y-H. Adequacy and Complications of Computed Tomography-Guided Core Needle Biopsy on Non-Small Cell Lung Cancers for Epidermal Growth Factor Receptor Mutations Demonstration: 18-Gauge or 20-Gauge Biopsy Needle. Lung Cancer, 2010, 67(2):166–9., DOI 10.1016/j.lungcan.2009.04.007 Shi Y, Au JS-K, Thongprasert S, Srinivasan S, Tsai C-M, Khoa MT, et al. A Prospective, Molecular Epidemiology Study of EGFR Mutations in Asian Patients with Advanced Non-Small-Cell Lung Cancer of Adenocarcinoma Histology, PIONEER). J Thorac Oncol, 2014, 9(2):154–62., DOI 10.1097/ JTO.0000000000000033 Shi Y, Li J, Zhang S, Wang M, Yang S, Li N.,et al. Molecular Epidemiology of EGFR Mutations in Asian Patients with Advanced Non-Small-Cell Lung Cancer of Adenocarcinoma Histology - Mainland China Subset Analysis of the PIONEER Study. PLoS One, 2015, 10(11):e0143515., DOI 10.1371/ journal.pone.0143515 Verhoest G, Patard J-J, Fergelot P, Jouan F, Zerrouki S, Dreano S, et al. Paraffin-Embedded Tissue Is Less Accurate Than Frozen Section Analysis for Determining VHL Mutational Status in Sporadic Renal Cell Carcinoma. Urol Oncol Semin Original Invest, 2012, 30(4):469–75., DOI 10.1016/ j.urolonc.2010.07.005 Marino FZ, Bianco R, Accardo M, Ronchi A, Cozzolino I, Morgillo F, et al. Molecular Heterogeneity in Lung Cancer: From Mechanisms of Origin to Clinical Implications. Int J Med Sci, 2019, 16(7):981–9., DOI 10.7150/ ijms.34739 MatsumuraM, Okudela K, Kojima Y,Umeda S, Tateishi Y, SekineA, et al. A Histopathological Feature of EGFR-Mutated Lung Adenocarcinomas with Highly Malignant Potential - An Implication of Micropapillary Element. Plos One, 2016, 11(11):e0166795., DOI 10.1371/ journal.pone.0166795 Zhang C, Schmidt LA, Hatanaka K, Thomas D, Lagstein A, Myers JL. Evaluation of Napsin A, TTF-1, P63, P40, and CK5/6 Immunohistochemical Stains in Pulmonary Neuroendocrine Tumors. Am J Clin Pathol, 2014, 142:320–4., DOI 10.1309/AJCPGA0IUA8BHQEZ Sheinson D, Wong WB, Wu N, Mansfield AS. Impact of Delaying Initiation of Anaplastic Lymphoma Kinase Inhibitor Treatment on Survival in Patients with Advanced Non-Small-Cell Lung Cancer. Lung Cancer, 2020, 143:86–92., DOI 10.1016/j.lungcan.2020.03.005 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609931 EP 1609939 DI 10.3389/pore.2021.1609931 PG 9 ER PT J AU Cai, H Chen, H Huang, Q Zhu, JM Ke, ZB Lin, YZ Zheng, QSh Wei, Y Xu, N Xue, XY AF Cai, Hai Chen, Hang Huang, Qi Zhu, Jun-Ming Ke, Zhi-Bin Lin, Yun-Zhi Zheng, Qing-Shui Wei, Yong Xu, Ning Xue, Xue-Yi TI Ubiquitination-Related Molecular Subtypes and a Novel Prognostic Index for Bladder Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; bladder cancer; ubiquitination; molecular subtypes; prognostic index ID prognosis; bladder cancer; ubiquitination; molecular subtypes; prognostic index AB Objective: To develop and validate ubiquitination-related molecular subtypes and a novel prognostic index using ubiquitination-related genes (URGs) for patients with bladder cancer (BCa). Materials and Methods: We downloaded the clinical data and transcriptome data of BCa from TCGA and GEO database. Consensus clustering analysis was conducted to identify ubiquitination-related molecular subtypes for BCa. Besides, we performed univariate and multivariate Cox regression analysis to develop a novel prognostic URGs-related index for BCa. We conducted internal and external verification in TCGA cohort and GEO cohort, respectively. Furthermore, the associations of ubiquitinationrelated molecular subtypes and prognostic index with tumor immune environment were also investigated. Results: A total of four ubiquitination-related molecular subtypes of BCa were finally identified. These four molecular subtypes had significantly different clinical characteristics, prognosis, PD-L1 expression level and tumor microenvironment. Besides, we developed a novel prognostic index using six URGs (including HLA-A, TMEM129, UBE2D1, UBE2N, UBE2T and USP5). The difference in OS between high and low-risk group was statistically significant in training cohort, testing cohort, and validating cohort. The area under ROC curve (AUC) for OS prediction was 0.736, 0.723, and 0.683 in training cohort, testing cohort, and validating cohort, respectively. Multivariate survival analysis showed that this index was an independent predictor for OS. This prognostic index was especially suitable for subtype 1 and 3, older, male, high grade, AJCC stage III-IV, stage N0, stage T3-4 BCa patients. Conclusions: This study identified a total of four ubiquitination-related molecular subtypes with significantly different tumor microenvironment, prognosis, clinical characteristics and PD-L1 expression level. Besides, a novel ubiquitination-related prognostic index for BCa patients was developed and successfully verified, which performed well in predicting prognosis of BCa. C1 [Cai, Hai] The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of UrologyFuzhou, China. [Chen, Hang] The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of UrologyFuzhou, China. [Huang, Qi] The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of UrologyFuzhou, China. [Zhu, Jun-Ming] The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of UrologyFuzhou, China. [Ke, Zhi-Bin] The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of UrologyFuzhou, China. [Lin, Yun-Zhi] The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of UrologyFuzhou, China. [Zheng, Qing-Shui] The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of UrologyFuzhou, China. [Wei, Yong] The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of UrologyFuzhou, China. [Xu, Ning] The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of UrologyFuzhou, China. [Xue, Xue-Yi] The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of UrologyFuzhou, China. RP Xue, XY (reprint author), The First Affiliated Hospital, Fujian Medical University, Urology Research Institute, Department of Urology, Fuzhou, China. EM xuexueyi@fjmu.edu.cn CR Lenis A. T., Lec P. M., Chamie K., and Mshs M., Bladder Cancer: A Review. Jama, 2020, 324(19):1980–91., DOI 10.1001/jama.2020.17598 Richters A., Aben K. K. H., and Kiemeney L. A. L. M., The Global burden of Urinary Bladder Cancer: an Update. World J Urol, 2020, 38(8):1895–904., DOI 10.1007/s00345-019-02984-4 Ru Y., Dancik G. M., and Theodorescu D., Biomarkers for Prognosis and Treatment Selection in Advanced Bladder Cancer Patients. Curr Opin Urol, 2011, 21(5):420–7., DOI 10.1097/MOU.0b013e32834956d6 Sjodahl G., Lauss M., Lovgren K., Chebil G., Gudjonsson S., Veerla S., et al.,A Molecular Taxonomy for Urothelial Carcinoma. Clin Cancer Res, 2012, 18(12):3377–86., DOI 10.1158/1078-0432.Ccr-12-0077-t Choi W., Porten S., Kim S., Willis D., Plimack E. R., Hoffman-Censits J., et al.,Identification of Distinct Basal and Luminal Subtypes of Muscle-Invasive Bladder Cancer with Different Sensitivities to Frontline Chemotherapy. Cancer Cell, 2014, 25(2):152–65., DOI 10.1016/j.ccr.2014.01.009 Damrauer J. S., Hoadley K. A., Chism D. D., Fan C., Tiganelli C. J., Wobker S. E., et al.,Intrinsic Subtypes of High-Grade Bladder Cancer Reflect the Hallmarks of Breast Cancer Biology. Proc Natl Acad Sci, 2014, 111(8): 3110–5., DOI 10.1073/pnas.1318376111 Hedegaard J., Lamy P., Nordentoft I., Algaba F., Hoyer S., Ulhoi B. P., et al.,Comprehensive Transcriptional Analysis of Early-Stage Urothelial Carcinoma. Cancer Cell, 2016, 30(1):27–42., DOI 10.1016/j.ccell.2016.05.004 Robertson A. G, Kim J., Al-Ahmadie H., Bellmunt J., Guo G., Cherniack A. D., et al.,Comprehensive Molecular Characterization of Muscle- Invasive Bladder Cancer. Cell, 2017, 171(3):540–556. e525., DOI 10.1016/j.cell.2017.09.007 Kamoun A., de Reynies A., Allory Y., Sjodahl G., Robertson A. G., Seiler R., et al.,A Consensus Molecular Classification of Muscle-Invasive Bladder Cancer. Eur Urol, 2020, 77(4):420–33., DOI 10.1016/j.eururo.2019.09.006 Mansour M. A., Ubiquitination: Friend and Foe in Cancer. Int J Biochem Cel Biol, 2018, 101:80–93., DOI 10.1016/j.biocel.2018.06.001 Setiawan B., Rosalina R., Pangarsa E. A., Santosa D., and Suharti C., Clinical Evaluation for the Role of High-Sensitivity C-Reactive Protein in Combination with D-Dimer and Wells Score Probability Test to Predict the Incidence of Deep Vein Thrombosis Among Cancer Patients. Ijgm, 2020, 13:587–94., DOI 10.2147/ijgm.S261718 Sato A., Asano T., Okubo K., Isono M., and Asano T., Ritonavir and Ixazomib Kill Bladder Cancer Cells by Causing Ubiquitinated Protein Accumulation. Cancer Sci, 2017, 108(6):1194–202., DOI 10.1111/cas.13242 Wang X., Liu Z., Zhang L., Yang Z., Chen X., Luo J., et al.,Targeting Deubiquitinase USP28 for Cancer Therapy. Cell Death Dis, 2018, 9(2):186., DOI 10.1038/s41419-017-0208-z Xu N., Ke Z-B., Lin X-D., Chen Y-H., Wu Y-P., Chen Y., et al.,Development and Validation of a Molecular Prognostic index of Bladder Cancer Based on Immunogenomic Landscape Analysis. Cancer Cel Int, 2020, 20:302., DOI 10.1186/s12935-020-01343-3 Ke Z. B., Wu Y. P., Huang P., Hou J., Chen Y. H., Dong R. N., et al.,Identification of Novel Genes in Testicular Cancer Microenvironment Based on ESTIMATE Algorithm-derived Immune Scores. J Cel Physiol, 2020, 236:706–13., DOI 10.1002/jcp.29898 Mou Z., Yang C., Zhang Z., Wu S., Xu C., Cheng Z., et al.,Transcriptomic Analysis of Glycolysis-Related Genes Reveals an Independent Signature of Bladder Carcinoma. Front Genet, 2020, 11:566918., DOI 10.3389/ fgene.2020.566918 El Sabbagh R., Azar N. S., Eid A. A., and Azar S. T., Thyroid Dysfunctions Due to Immune Checkpoint Inhibitors: A Review. Ijgm, 2020, 13:1003–9., DOI 10.2147/ijgm.S261433 Siegel R., Ma J., Zou Z., and Jemal A., Cancer Statistics, 2014. CA A Cancer J Clinicians, 2014, 64(1):9–29., DOI 10.3322/caac.21208 Grayson M., Bladder Cancer. Nature, 2017, 551(7679):S33., DOI 10.1038/551S33a Cao J., Li J., Yang X., Li P., Yao Z., Han D., et al.,Integrative Analysis of Immune Molecular Subtypes and Microenvironment Characteristics of Bladder Cancer. Cancer Med, 2021, 10(15):5375–91., DOI 10.1002/cam4.4071 Kim H. S., and Seo H. K., Immune Checkpoint Inhibitors for Urothelial Carcinoma. Investig Clin Urol, 2018, 59(5):285–96., DOI 10.4111/icu.2018.59.5.285 Kirkali Z., Chan T., Manoharan M., Algaba F., Busch C., Cheng L., et al.,Bladder Cancer: Epidemiology, Staging and Grading, and Diagnosis. Urology, 2005, 66(6 Suppl. 1):4–34., DOI 10.1016/j.urology.2005.07.062 Bellmunt J., Bladder Cancer. Hematology/Oncology Clin North America, 2015, 29(2):xiii–xiv., DOI 10.1016/j.hoc.2014.12.001 Zhang F., Wang X., Bai Y., Hu H., Yang Y., Wang J., et al.,Development and Validation of a Hypoxia-Related Signature for Predicting Survival Outcomes in Patients with Bladder Cancer. Front Genet, 2021, 12:670384., DOI 10.3389/ fgene.2021.670384 Liu J., Ma H.,Meng L., Liu X., Lv Z., Zhang Y., et al.,Construction and External Validation of a Ferroptosis-Related Gene Signature of Predictive Value for the Overall Survival in Bladder Cancer. Front Mol Biosci, 2021, 8:675651., DOI 10.3389/fmolb.2021.675651 Qu G., Liu Z., Yang G., Xu Y., Xiang M., and Tang C., Development of a Prognostic index and Screening of Prognosis Related Genes Based on an Immunogenomic Landscape Analysis of Bladder Cancer. Aging, 2021, 13(8): 12099–112., DOI 10.18632/aging.202917 Luo C., Ye W., Hu J., Othmane B., Li H., Chen J., et al.,A Poliovirus Receptor, CD155)-Related Risk Signature Predicts the Prognosis of Bladder Cancer. Front Oncol, 2021, 11:660273., DOI 10.3389/fonc.2021.660273 Du Y., Wang B., Jiang X., Cao J., Yu J., Wang Y., et al.,Identification and Validation of a Stromal EMT Related LncRNA Signature as a Potential Marker to Predict Bladder Cancer Outcome. Front Oncol, 2021, 11:620674., DOI 10.3389/fonc.2021.620674 Dard C., Senoussi O., Buhler S., Bardy B., and Masson D., Identification of Four Novel HLA-A Alleles. Hla, 2020, 96(2):202–3., DOI 10.1111/tan.13899 Golnik R., Lehmann A., Kloetzel P-M., and Ebstein F., Major Histocompatibility Complex, MHC, Class I Processing of the NY-ESO-1 Antigen Is Regulated by Rpn10 and Rpn13 Proteins and Immunoproteasomes Following Non-lysine Ubiquitination. J Biol Chem, 2016, 291(16):8805–15., DOI 10.1074/jbc.M115.705178 Zhou C., Bi F., Yuan J., Yang F., and Sun S., Gain of UBE2D1 Facilitates Hepatocellular Carcinoma Progression and Is Associated with DNA Damage Caused by Continuous IL-6. J Exp Clin Cancer Res, 2018, 37(1):290., DOI 10.1186/s13046-018-0951-8 Figueroa J. D., Koutros S., Colt J. S., Kogevinas M., Garcia-Closas M., Real F. X., et al.,Modification of Occupational Exposures on Bladder Cancer Risk by Common Genetic Polymorphisms. J Natl Cancer Inst, 2015, 107(11):djv223., DOI 10.1093/jnci/djv223 YanH., Ren S., LinQ., Yu Y.,Chen C., Hua X., et al.,Inhibition of UBE2N-dependent CDK6 Protein Degradation by miR-934 Promotes Human Bladder Cancer Cell Growth. FASEB j., 2019, 33(11):12112–23., DOI 10.1096/fj.201900499RR Gong Y. Q., Peng D., Ning X. H., Yang X. Y., Li X. S., Zhou L. Q., et al.,UBE2T Silencing Suppresses Proliferation and Induces Cell Cycle Arrest and Apoptosis in Bladder Cancer Cells. Oncol Lett, 2016, 12(6):4485–92., DOI 10.3892/ol.2016.5237 Xiao M., Liu J., Xiang L., Zhao K., He D., Zeng Q., et al.,MAFG-AS1 Promotes Tumor Progression via Regulation of the HuR/PTBP1 axis in Bladder Urothelial Carcinoma. Clin Translational Med, 2020, 10(8):e241., DOI 10.1002/ctm2.241 HahnM.A.,DicksonK-A., Jackson S., ClarksonA.,GillA. J., andMarshD. J.,The Tumor Suppressor CDC73 Interacts with the Ring finger Proteins RNF20 and RNF40 and Is Required for the Maintenance of Histone 2B Monoubiquitination. Hum Mol Genet, 2012, 21(3):559–68., DOI 10.1093/hmg/ddr490 Shang Z-F., Tan W., Liu X-D., Yu L., Li B., Li M., et al.,DNA-PKcs Negatively Regulates Cyclin B1 Protein Stability through Facilitating its Ubiquitination Mediated by Cdh1-APC/C Pathway. Int J Biol Sci, 2015, 11(9):1026–35., DOI 10.7150/ijbs.12443 Fu J., Liao L., Balaji K. S., Wei C., Kim J., and Peng J., Epigenetic Modification and a Role for the E3 Ligase RNF40 in Cancer Development and Metastasis. Oncogene, 2021, 40(3):465–74., DOI 10.1038/s41388-020-01556-w So E. Y., and Ouchi T., The Application of Toll like Receptors for Cancer Therapy. Int J Biol Sci, 2010, 6(7):675–81., DOI 10.7150/ijbs.6.675 Cheng S., Liu J., Zhang Y., Lin Y., Liu Q., Li H., et al.,Association Detection between Genetic Variants in the microRNA Binding Sites of Toll-like Receptors Signaling Pathway Genes and Bladder Cancer Susceptibility. Int J Clin Exp Pathol, 2014, 7(11):8118–26. Ohadian Moghadam S., and Nowroozi M. R., Toll-like Receptors: The Role in Bladder Cancer Development, Progression and Immunotherapy. Scand J Immunol, 2019, 90(6):e12818., DOI 10.1111/sji.12818 Tschernig T., Connexins and Gap Junctions in Cancer of the Urinary Tract. Cancers, 2019, 11(5):704., DOI 10.3390/cancers11050704 Ai X-l., Chi Q., Qiu Y., Li H-y., Li D-j., Wang J-x., et al.,Gap junction Protein Connexin43 Deregulation Contributes to Bladder Carcinogenesis via Targeting MAPK Pathway. Mol Cel Biochem, 2017, 428(1-2):109–18., DOI 10.1007/s11010-016-2921-9 Grossman H. B, Liebert M., Lee I. W., and Lee S. W., Decreased Connexin Expression and Intercellular Communication in Human Bladder Cancer Cells. Cancer Res, 1994, 54(11):3062–5. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609941 EP 1609957 DI 10.3389/pore.2021.1609941 PG 17 ER PT J AU Jiang, Y Gu, H Zheng, X Pan, B Liu, P Zheng, M AF Jiang, Yinan Gu, Haifeng Zheng, Xiaojing Pan, Baoyue Liu, Pingping Zheng, Min TI Pretreatment C-Reactive Protein/ Albumin Ratio is Associated With Poor Survival in Patients With 2018 FIGO Stage IB-IIA HPV-Positive Cervical Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE recurrence; survival; CAR; prognostic markers; UCC; HPV ID recurrence; survival; CAR; prognostic markers; UCC; HPV AB Objectives: The present study aimed to identify the predictive value of inflammatory indexes stratified according to human papillomavirus (HPV) infection status in women with FIGO 2018 stage IB∼IIA cervical cancer. We also explored the influences of HPV infection status on the survival of cervical cancer patients. Methods: We collected data for 583 women with stage IB∼IIA cervical cancer in Sun Yatsen University Cancer Center between 2009 and 2017. The t-test, chi-squared (χ2) test and Fisher’s exact test were applied to compare the differences of inflammatory indexes and clinicopathological features between HPV-positive and HPV-negative groups. Univariate and multivariate analyses were used to identify clinicopathological factors that were associated with the prognosis of cervical cancer patients. Results: There were no differences in overall survival (OS) and progression-free survival (PFS) between HPV-positive and HPV-negative groups. In HPV-positive group, the maximum tumor size, neoadjuvant chemotherapy and the body mass index (BMI) correlated significantly with C-reactive protein/albumin ratio (CAR). The maximum tumor size and the prognostic nutritional index (PNI) correlated significantly with the platelet-lymphocyte ratio (PLR). The maximum tumor size, neoadjuvant chemotherapy and PLR correlated significantly with PNI. Univariate and multivariate analyses showed that the depth of tumor invasion (HR: 3.651, 95% CI: 1.464–9.103, p = 0.005; HR: 2.478, 95% CI: 1.218–5.043, p = 0.012) and CAR (HR: 5.201, 95% CI: 2.080–13.004, p < 0.0001; HR: 2.769, 95% CI: 1.406–5.455, p = 0.003) were independent predictors of poor OS and PFS. PNI was an independent protective factor of OS (HR: 0.341, 95% CI: 0.156–0.745, p = 0.007). PLR was an independent factor of PFS (HR: 1.991, 95% CI: 1.018–3.894, p = 0.044). In HPV-negative group, BMI correlated significantly with CAR. Only depth of invasion (HR: 9.192, 95% CI: 1.016–83.173, p = 0.048) was the independent predictor of poor OS, and no inflammation indexes were independent predictors of prognosis. Conclusion: In patients with HPV-positive cervical cancer, depth of invasion, PNI and CAR are independent factors of OS, and depth of invasion, PLR and CAR are independent factors for PFS. For patients with HPV-negative disease, no inflammation indexes had predictive value for prognosis. The predictive value of inflammation indexes on prognosis is more significant in patients with HPV-positive cervical cancer. Stratification of HPV infection status promotes a more precise clinical application of inflammation indexes, thus improving their accuracy and feasibility. C1 [Jiang, Yinan] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. [Gu, Haifeng] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. [Zheng, Xiaojing] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. [Pan, Baoyue] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. [Liu, Pingping] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. [Zheng, Min] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. RP Zheng, M (reprint author), Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecology, Guangzhou, China. EM zhengmin@sysucc.org.cn CR 2. Sadri Nahand J, Moghoofei M, Salmaninejad A, Bahmanpour Z, Karimzadeh M, Nasiri M, et al. Pathogenic Role of Exosomes and microRNAs in HPVmediated Inflammation and Cervical Cancer: A Review. Int J Cancer, 2020, 146(2):305–20., DOI 10.1002/ijc.32688 3. Roden RBS, Stern PL. Opportunities and Challenges for Human Papillomavirus Vaccination in Cancer. Nat Rev Cancer, 2018, 18(4): 240–54., DOI 10.1038/nrc.2018.13 4. Zhao F, Qiao Y. Cervical Cancer Prevention in China: a Key to Cancer Control. The Lancet, 2019, 393(10175):969–70., DOI 10.1016/s0140- 6736(18)32849-6 5. Cohen PA, Jhingran A, Oaknin A, Denny L. Cervical Cancer. The Lancet, 2019, 393(10167):169–82., DOI 10.1016/s0140-6736(18)32470-x 6. Bhatla N, Aoki D, Sharma DN, Sankaranarayanan R. Cancer of the Cervix Uteri. Int J Gynecol Obstet, 2018, 143(Suppl. 2):22–36., DOI 10.1002/ijgo.12611 7. Namikawa T, Shimizu S, Yokota K, Tanioka N, Munekage M, Uemura S, et al. Neutrophil-to-lymphocyte Ratio and C-Reactive Protein-To-Albumin Ratio as Prognostic Factors for Unresectable Advanced or Recurrent Gastric Cancer. Langenbecks Arch Surg, 2021)., DOI 10.1007/s00423-021-02356-w 8. Yang Y, Gao P, Song Y, Sun J, Chen X, Zhao J, et al. The Prognostic Nutritional index Is a Predictive Indicator of Prognosis and Postoperative Complications in Gastric Cancer: A Meta-Analysis. Eur J Surg Oncol, Ejso,, 2016, 42(8): 1176–82., DOI 10.1016/j.ejso.2016.05.029 9. McMillan DC, Canna K, McArdle CS. Systemic Inflammatory Response Predicts Survival Following Curative Resection of Colorectal Cancer. Br J Surg, 2003, 90(2):215–9., DOI 10.1002/bjs.4038 10. Ayhan A, Gunakan E, Alyazici I, Haberal N, Altundag O, Dursun P. The Preoperative Albumin Level Is an Independent Prognostic Factor for Optimally Debulked Epithelial Ovarian Cancer. Arch Gynecol Obstet, 2017, 296(5):989–95., DOI 10.1007/s00404-017-4511-9 11. Zhang F, Liu Z, Liang J, Liu S, Wu K, Zhang F, et al. Association between Preoperative Serum Albumin and Prognosis in Patients with Adrenocortical Carcinoma after Primary Resection: a Retrospective Study. BMC Cancer, 2021, 21(1):961., DOI 10.1186/s12885-021-08689-5 12. Zhou T, Zhan J, Hong S, Hu Z, Fang W, Qin T, et al. Ratio of C-Reactive Protein/Albumin Is an Inflammatory Prognostic Score for Predicting Overall Survival of Patients with Small-Cell Lung Cancer. Sci Rep, 2015, 5:10481., DOI 10.1038/srep10481 13. Deng Y, Zhao Y, Qin J, Huang X, Wu R, Zhou C, et al. Prognostic Value of the C-Reactive Protein/Albumin Ratio and Systemic Immune-Inflammation Index for Patients with Colorectal Liver Metastasis Undergoing Curative Resection. Pathol Oncol Res, 2021, 27:633480., DOI 10.3389/pore.2021.633480 14. Stevens L, Pathak S, Nunes QM, Pandanaboyana S, Macutkiewicz C, Smart N, et al. Prognostic Significance of Pre-operative C-Reactive Protein and the Neutrophil-Lymphocyte Ratio in Resectable Pancreatic Cancer: a Systematic Review. HPB, 2015, 17(4):285–91., DOI 10.1111/hpb.12355 15. Kinoshita A, Onoda H, Imai N, Iwaku A, Oishi M, Tanaka K, et al. The C-Reactive Protein/albumin Ratio, a Novel Inflammation-Based Prognostic Score, Predicts Outcomes in Patients with Hepatocellular Carcinoma. Ann Surg Oncol, 2015, 22(3):803–10., DOI 10.1245/s10434-014-4048-0 16. Smith RA, Bosonnet L, Raraty M, Sutton R, Neoptolemos JP, Campbell F, et al. Preoperative Platelet-Lymphocyte Ratio Is an Independent Significant Prognostic Marker in Resected Pancreatic Ductal Adenocarcinoma. Am J Surg, 2009, 197(4):466–72., DOI 10.1016/j.amjsurg.2007.12.057 17. Davis AN, Afshar-Kharghan V, Sood AK. Platelet Effects on Ovarian Cancer. Semin Oncol, 2014, 41(3):378–84., DOI 10.1053/j.seminoncol.2014.04.004 18. Zhang Y, Wang L, Liu Y, Wang S, Shang P, Gao Y, et al. Preoperative Neutrophil-Lymphocyte Ratio before Platelet-Lymphocyte Ratio Predicts Clinical Outcome in Patients with Cervical Cancer Treated with Initial Radical Surgery. Int J Gynecol Cancer, 2014, 24(7):1319–25., DOI 10.1097/ IGC.0000000000000219 19. Asher V, Lee J, Innamaa A, Bali A. Preoperative Platelet Lymphocyte Ratio as an Independent Prognostic Marker in Ovarian Cancer. Clin Transl Oncol, 2011, 13(7):499–503., DOI 10.1007/s12094-011-0687-9 20. Chan AWH, Chan SL, Wong GLH, Wong VWS, Chong CCN, Lai PBS, et al. Prognostic Nutritional Index, PNI, Predicts Tumor Recurrence of Very Early/ Early Stage Hepatocellular Carcinoma after Surgical Resection. Ann Surg Oncol, 2015, 22(13):4138–48., DOI 10.1245/s10434-015-4516-1 21. Migita K, Takayama T, Saeki K, Matsumoto S, Wakatsuki K, Enomoto K, et al. The Prognostic Nutritional index Predicts Long-Term Outcomes of Gastric Cancer Patients Independent of Tumor Stage. Ann Surg Oncol, 2013, 20(8): 2647–54., DOI 10.1245/s10434-013-2926-5 22. Kanda M, Fujii T, Kodera Y, Nagai S, Takeda S, Nakao A. Nutritional Predictors of Postoperative Outcome in Pancreatic Cancer. Br J Surg, 2010, 98(2):268–74., DOI 10.1002/bjs.7305 23. Wang W-J, Li Y, Zhu J, Gao M-j., Shi J-p., Huang Y-q. Prognostic Values of Systemic Inflammation Response, SIR, Parameters in Resectable Cervical Cancer. Dose-Response, 2019, 17(1):155932581982954., DOI 10.1177/ 1559325819829543 24. Han X, Liu S, Yang G, Hosseinifard H, Imani S, Yang L, et al. Prognostic Value of Systemic Hemato-Immunological Indices in Uterine Cervical Cancer: A Systemic Review, Meta-Analysis, and Meta-Regression of Observational Studies. Gynecol Oncol, 2021, 160(1):351–60., DOI 10.1016/j.ygyno.2020.10.011 25. Nishijima TF, Muss HB, Shachar SS, Tamura K, Takamatsu Y. Prognostic Value of Lymphocyte-To-Monocyte Ratio in Patients with Solid Tumors: A Systematic Review and Meta-Analysis. Cancer Treat Rev, 2015, 41(10):971–8., DOI 10.1016/j.ctrv.2015.10.003 26. National Comprehensive Cancer Network. Clinical Practice Guidelines in Oncology. Cervical Cancer. Version 1. 2022 Available At: https://www.nccn. org/professionals/physician_gls/pdf/cervical.pdf, Accessed October 26, 2021). 27. Yurkovetskiy LA, Pickard JM, Chervonsky AV. Microbiota and Autoimmunity: Exploring New Avenues. Cell Host & Microbe, 2015, 17(5): 548–52., DOI 10.1016/j.chom.2015.04.010 28. Chen Y-P, Chan ATC, Le Q-T, Blanchard P, Sun Y, Ma J. Nasopharyngeal Carcinoma. The Lancet, 2019, 394(10192):64–80., DOI 10.1016/s0140-6736(19, 30956-0 29. Amieva M, Peek RM, Jr. Pathobiology of helicobacter Pylori-Induced Gastric Cancer. Gastroenterology, 2016, 150(1):64–78., DOI 10.1053/ j.gastro.2015.09.004 30. de Visser KE, Eichten A, Coussens LM. Paradoxical Roles of the Immune System during Cancer Development. Nat Rev Cancer, 2006, 6(1):24–37., DOI 10.1038/nrc1782 31. Janssen LME, Ramsay EE, Logsdon CD, Overwijk WW. The Immune System in Cancer Metastasis: Friend or Foe? J Immunotherapy Cancer, 2017, 5(1):79., DOI 10.1186/s40425-017-0283-9 32. Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related Inflammation and Treatment Effectiveness. Lancet Oncol, 2014, 15(11):e493–e503., DOI 10.1016/s1470-2045(14)70263-3 33. Crusz SM, Balkwill FR. Inflammation and Cancer: Advances and New Agents. Nat Rev Clin Oncol, 2015, 12(10):584–96., DOI 10.1038/nrclinonc.2015.105 34. Lin Y, Xu J, Lan H. Tumor-associated Macrophages in Tumor Metastasis: Biological Roles and Clinical Therapeutic Applications. J Hematol Oncol, 2019, 12(1):76., DOI 10.1186/s13045-019-0760-3 35. Hoppe-Seyler K, Bossler F, Braun JA, Herrmann AL, Hoppe-Seyler F. The HPV E6/E7 Oncogenes: Key Factors for Viral Carcinogenesis and Therapeutic Targets. Trends Microbiol, 2018, 26(2):158–68., DOI 10.1016/j.tim.2017.07.007 36. Zhou J, Peng C, Li B, Wang F, Zhou C, Hong D, et al. Transcriptional Gene Silencing of HPV16 E6/E7 Induces Growth Inhibition via Apoptosis In Vitro and In Vivo. Gynecol Oncol, 2012, 124(2):296–302., DOI 10.1016/ j.ygyno.2011.10.028 37. Nie D, Gong H, Mao X, Li Z. Systemic Immune-Inflammation index Predicts Prognosis in Patients with Epithelial Ovarian Cancer: A Retrospective Study. Gynecol Oncol, 2019, 152(2):259–64., DOI 10.1016/j.ygyno.2018.11.034 38. Schlesinger M. Role of Platelets and Platelet Receptors in Cancer Metastasis. J Hematol Oncol, 2018, 11(1):125., DOI 10.1186/s13045-018-0669-2 39. Maraz A, Furak J, Varga Z, Kahan Z, Tiszlavicz L, Hideghety K. Thrombocytosis Has a Negative Prognostic Value in Lung Cancer. Anticancer Res, 2013, 33(4):1725–30. 40. Siemes C, Visser LE, Coebergh J-WW, Splinter TAW, Witteman JCM, Uitterlinden AG, et al. C-reactive Protein Levels, Variation in the C-Reactive Protein Gene, and Cancer Risk: the Rotterdam Study. J Clin Oncol, 2006, 24(33):5216–22., DOI 10.1200/JCO.2006.07.1381 41. Obermair A, Simunovic M, Isenring L, Janda M. Nutrition Interventions in Patients with Gynecological Cancers Requiring Surgery. Gynecol Oncol, 2017, 145(1):192–9., DOI 10.1016/j.ygyno.2017.01.028 42. Gupta D, Lis CG. Pretreatment Serum Albumin as a Predictor of Cancer Survival: A Systematic Review of the Epidemiological Literature. Nutr J, 2010, 9:69., DOI 10.1186/1475-2891-9-69 43. Ferrone C, Dranoff G. Dual Roles for Immunity in Gastrointestinal Cancers. J Clin Oncol, 2010, 28(26):4045–51., DOI 10.1200/JCO.2010.27.9992 44. Duchemann B, Remon J, Naigeon M, Mezquita L, Ferrara R, Cassard L, et al. Integrating Circulating Biomarkers in the Immune Checkpoint Inhibitor Treatment in Lung Cancer. Cancers, 2020, 12(12):3625., DOI 10.3390/ cancers12123625 45. Shoji F, Takeoka H, Kozuma Y, Toyokawa G, Yamazaki K, Ichiki M, et al. Pretreatment Prognostic Nutritional index as a Novel Biomarker in Non-small Cell Lung Cancer Patients Treated with Immune Checkpoint Inhibitors. Lung Cancer, 2019, 136:45–51., DOI 10.1016/j.lungcan.2019.08.006 46. Oya Y, Yoshida T, Kuroda H, Mikubo M, Kondo C, Shimizu J, et al. Predictive Clinical Parameters for the Response of Nivolumab in Pretreated Advanced Non-small-cell Lung Cancer. Oncotarget, 2017, 8(61):103117–28., DOI 10.18632/oncotarget.21602 47. Huang H, Liu Q, Zhu L, Zhang Y, Lu X,Wu Y, et al. Prognostic Value of Preoperative Systemic Immune-Inflammation index in Patients with Cervical Cancer. Sci Rep, 2019, 9(1):3284., DOI 10.1038/s41598-019- 39150-0 48. Zhang W, Liu K, Ye B, Liang W, Ren Y. Pretreatment C-Reactive Protein/ albumin Ratio Is Associated with Poor Survival in Patients with Stage IBIIA Cervical Cancer. Cancer Med, 2018, 7(1):105–13., DOI 10.1002/ cam4.1270 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609946 EP 1609959 DI 10.3389/pore.2021.1609946 PG 14 ER PT J AU Govoni, M Rossi, V Stefano, DG Manerba, M AF Govoni, Marzia Rossi, Valentina Stefano, Di Giuseppina Manerba, Marcella TI Lactate Upregulates the Expression of DNA Repair Genes, Causing Intrinsic Resistance of Cancer Cells to Cisplatin SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE DNA repair; cisplatin; glycolysis; lactate; DNA damage ID DNA repair; cisplatin; glycolysis; lactate; DNA damage AB Intrinsic or acquired drug resistance is one of the major problems compromising the success of antineoplastic treatments. Several evidences correlated some therapeutic failures with changes in cell metabolic asset and in line with these findings, hindering the glycolytic metabolism of cancer cells via lactate dehydrogenase (LDH) inhibition was found to overcome the resistance to chemotherapeutic agents. Lactate, the product of LDH reaction, was shown to be involved in epigenetic regulation of gene expression. The experiments described in this paper were aimed at highlighting a possible direct effect of lactate in modifying the response of cancer cells to a chemotherapeutic treatment. To discriminate between the effects potentially caused by glycolytic metabolism from those directly referable to lactate, we selected cancer cell lines able to grow in glucose deprived conditions and evaluated the impact of lactate on the cellular response to cisplatin-induced DNA damage. In lactate-exposed cells we observed a reduced efficacy of cisplatin, which was associated with reduced signatures of DNA damage, enhanced DNA recombination competence and increased expression of a panel of genes involved in DNA repair. The identified genes take part in mismatch and nucleotide excision repair pathways, which were found to contribute in restoring the cisplatin-induced DNA damage. The obtained results suggest that this metabolite could play a role in reducing the efficacy of antineoplastic treatments. C1 [Govoni, Marzia] University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine (DIMES)Bologna, Italy. [Rossi, Valentina] University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine (DIMES)Bologna, Italy. [Stefano, Di Giuseppina] University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine (DIMES)Bologna, Italy. [Manerba, Marcella] University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine (DIMES)Bologna, Italy. RP Stefano, DG (reprint author), University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Bologna, Italy. EM giuseppina.distefano@unibo.it CR DeBerardinis RJ, Chandel NS. Fundamentals of Cancer Metabolism. Sci Adv, 2016, 2:e1600200., DOI 10.1126/sciadv.1600200 Bui T, Thompson CB. Cancer’s Sweet Tooth. Cancer Cell, 2006, 9:419–20., DOI 10.1016/j.ccr.2006.05.012 Warburg O. Uber den Stoffwechsel der Tumoren. Berlin: Verlag Springer, 1926). Translated into English by Dickens F. under the title: The metabolism of tumours. Constable, 1930), London. Liu X-S, Little JB, Yuan Z-M. Glycolytic Metabolism Influences Global Chromatin Structure. Oncotarget, 2015, 6:4214–25., DOI 10.18632/ oncotarget.2929 van der Knaap JA, Verrijzer CP. Undercover: Gene Control by Metabolites and Metabolic Enzymes. Genes Dev, 2016, 30:2345–69., DOI 10.1101/gad.289140.116 Bhatt AN, Chauhan A, Khanna S, Rai Y, Singh S, Soni R. Transient Elevation of Glycolysis Confers Radio-Resistance by Facilitating DNA Repair in Cells. BMC Cancer, 2015, 15:335., DOI 10.1186/s12885-015-1368-9 van Vugt MATM. Shutting Down the Power Supply for DNA Repair in Cancer Cells. J Cel Biol, 2017, 216:295–7., DOI 10.1083/jcb.201701026 Bhattacharya B, Mohd Omar MF, Soong R. The Warburg Effect and Drug Resistance. Br J Pharmacol, 2016, 173:970–9., DOI 10.1111/bph.13422 Fiume L, Manerba M, Vettraino M, Di Stefano G. Inhibition of Lactate Dehydrogenase Activity as an Approach to Cancer Therapy. Future Med Chem, 2014, 6:429–45., DOI 10.4155/fmc.13.206 LeA, Cooper CR,GouwAM, Dinavahi R, Maitra A,Deck LM. Inhibition of Lactate Dehydrogenase A Induces Oxidative Stress and Inhibits Tumor Progression. Proc Natl Acad Sci USA, 2010, 107:2037–42., DOI 10.1073/pnas.0914433107 Fiume L,Vettraino M, ManerbaM,Di Stefano G. Inhibition of Lactic Dehydrogenase as aWay to Increase theAnti-proliferative Effect ofMulti-Targeted Kinase Inhibitors. Pharmacol Res, 2011, 63:328–34., DOI 10.1016/j.phrs.2010.12.005 ManerbaM,Di Ianni L, Fiume L, Roberti M, Recanatini M,Di Stefano G. Lactate Dehydrogenase Inhibitors Sensitize Lymphoma Cells to Cisplatin without Enhancing the Drug Effects on Immortalized normal Lymphocytes. Eur J Pharm Sci, 2015, 74:95–102., DOI 10.1016/j.ejps.2015.04.022 Zhao Y, Butler EB, Tan M. Targeting Cellular Metabolism to Improve Cancer Therapeutics. Cell Death Dis, 2013, 4:e532., DOI 10.1038/cddis.2013.60 Boukouris AE, Zervopoulos SD, Michelakis ED. Metabolic Enzymes Moonlighting in the Nucleus: Metabolic Regulation of Gene Transcription. Trends Biochem Sci, 2016, 41:712–30., DOI 10.1016/j.tibs.2016.05.013 Latham T, Mackay L, Sproul D, Karim M, Culley J, Harrison DJ. Lactate, a Product of Glycolytic Metabolism, Inhibits Histone Deacetylase Activity and Promotes Changes in Gene Expression. Nucleic Acids Res, 2012, 40:4794–803., DOI 10.1093/nar/gks066 Wagner W, Ciszewski WM, Kania KD. L- and D-Lactate Enhance DNA Repair and Modulate the Resistance of Cervical Carcinoma Cells to Anticancer Drugs via Histone Deacetylase Inhibition and Hydroxycarboxylic Acid Receptor 1 Activation. Cell Commun Signal, 2015, 13:36., DOI 10.1186/s12964-015-0114-x San-Millan I, Julian CG, Matarazzo C, Martinez J, Brooks GA. Is Lactate an Oncometabolite? Evidence Supporting a Role for Lactate in the Regulation of Transcriptional Activity of Cancer-Related Genes in MCF7 Breast Cancer Cells. Front Oncol, 2020, 9:1536., DOI 10.3389/fonc.2019.01536 Baltazar F, Afonso J, Costa M, Granja S. Lactate beyond a Waste Metabolite: Metabolic Affairs and Signaling in Malignancy. Front Oncol, 2020, 10:231., DOI 10.3389/fonc.2020.00231 Pereira-Nunes A, Afonso J, Granja S, Baltazar F. Lactate and Lactate Transporters as Key Players in the Maintenance of the Warburg Effect. Adv Exp Med Biol, 2020, 1219:51–74., DOI 10.1007/978-3-030-34025-4_3 Zhang D, Tang Z, Huang H, Zhou G, Cui C,Weng Y. Metabolic Regulation of Gene Expression by Histone Lactylation. Nature, 2019, 574:575–80., DOI 10.1038/s41586-019-1678-1 Wagner W, Kania KD, Blauz A, Ciszewski WM. The Lactate Receptor, HCAR1/GPR81, Contributes to Doxorubicin Chemoresistance via ABCB1 Transporter Up-Regulation in Human Cervical Cancer HeLa Cells. J Physiol Pharmacol, 2017, 68:555–64. Wagner W, Kania KD, Ciszewski WM. Stimulation of Lactate Receptor, HCAR1, Affects Cellular DNA Repair Capacity. DNA Repair, 2017, 52: 49–58., DOI 10.1016/j.dnarep.2017.02.007 Sprowl-Tanio S, Habowski AN, Pate KT, McQuade MM, Wang K, Edwards RA. Lactate/pyruvate Transporter MCT-1 Is a Direct Wnt Target that Confers Sensitivity to 3-bromopyruvate in colon Cancer. Cancer Metab, 2016, 4:20., DOI 10.1186/s40170-016-0159-3c Fan Q, Yang L, Zhang X, Ma Y, Li Y, Dong L. Autophagy Promotes Metastasis and Glycolysis by Upregulating MCT1 Expression and Wnt/β-Catenin Signaling Pathway Activation in Hepatocellular Carcinoma Cells. J Exp Clin Cancer Res, 2018, 37:9., DOI 10.1186/s13046-018-0673-y Repetto G, del Peso A, Zurita JL. Neutral Red Uptake Assay for the Estimation of Cell Viability/cytotoxicity. Nat Protoc, 2008, 3:1125–31., DOI 10.1038/nprot.2008.75 Asaeda A, Ide H, Terato H, Takamori Y, Kubo K. Highly Sensitive Assay of DNA Abasic Sites in Mammalian Cells-Optimization of the Aldehyde Reactive Probe Method. Analytica Chim Acta, 1998, 365:35–41., DOI 10.1016/s0003- 2670(97)00648-x Green MR, Sambrook J. Precipitation of DNA with Ethanol. Cold Spring Harb Protoc, 20162016, 2016:pdb.prot093377., DOI 10.1101/pdb.prot093377 Sharma A, Singh K, Almasan A. Histone H2AX Phosphorylation: aMarker for DNA Damage. Methods Mol Biol, 2012, 920:613–26., DOI 10.1007/978-1- 61779-998-3_40 Papacostantinou J, Colowick SP. The Role of Glycolysis in the Growth of Tumor Cells. I. Effects of Oxamic Acid on the Metabolism of Ehrlich Tumor Cells In Vitro. J Biol Chem, 1961, 236:278–84. Chomczynski P, Sacchi N. Single-step Method of RNA Isolation by Acid Guanidinium Thiocyanate-Phenol-Chloroform Extraction. Anal Biochem, 1987, 162:156–9., DOI 10.1006/abio.1987.999910.1016/0003-2697(87)90021-2 Basu A, Krishnamurthy S. Cellular Responses to Cisplatin-Induced DNA Damage. J Nucleic Acids, 2010, 2010:1–16. pii 201367., DOI 10.4061/2010/201367 Liska DJ. The Detoxification Enzyme Systems. Altern Med Rev, 1998, 3:187–98. Stoimenov I, Helleday T. PCNA on the Crossroad of Cancer. Biochem Soc Trans, 2009, 37:605–13., DOI 10.1042/BST0370605 Wang S-C. PCNA: a Silent Housekeeper or a Potential Therapeutic Target? Trends Pharmacol Sci, 2014, 35:178–86., DOI 10.1016/j.tips.2014.02.004 Rodriguez N, Pelaez A, Barderas R, Dominguez G. Clinical Implications of the Deregulated TP73 Isoforms Expression in Cancer. Clin Transl Oncol, 2018, 20: 827–36., DOI 10.1007/s12094-017-1802-3 Loh C-Y, Chai J, Tang T, WongW, Sethi G, ShanmugamM. The E-Cadherin and N-Cadherin Switch in Epithelial-To-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells, 2019, 8:1118., DOI 10.3390/ cells8101118 Sun X, Kaufman PD. Ki-67: More Than a Proliferation Marker. Chromosoma, 2018, 127:175–86., DOI 10.1007/s00412-018-0659-8 Katoh M. Network of WNT and Other Regulatory Signaling Cascades in Pluripotent Stem Cells and Cancer Stem Cells. Curr Pharm Biotechnol, 2011, 12:160–70., DOI 10.2174/138920111794295710 Liu X, Tan N, Liao H, Pan G, Xu Q, Zhu R. High GSTP1 Inhibits Cell Proliferation by Reducing Akt Phosphorylation and Is Associated with a Better Prognosis in Hepatocellular Carcinoma. Oncotarget, 2018, 9:8957–71., DOI 10.18632/oncotarget.23420 Vassalli G. AldehydeDehydrogenases:Not Just Markers, but Functional Regulators of Stem Cells. Stem Cell Int, 2019, 2019:1–15., DOI 10.1155/2019/3904645 Formby B, Stern R. Lactate-sensitive Response Elements in Genes Involved in Hyaluronan Catabolism. Biochem Biophysical Res Commun, 2003, 305:203–8., DOI 10.1016/s0006-291x(03)00723-x Walenta S,Mueller-Klieser WF. Lactate:Mirror andMotor of Tumor Malignancy. Semin Radiat Oncol, 2004, 14:267–74., DOI 10.1016/j.semradonc.2004.04.004 Ippolito L, Morandi A, Giannoni E, Chiarugi P. Lactate: a Metabolic Driver in the Tumour Landscape. Trends Biochem Sci, 2019, 44:153–66., DOI 10.1016/ j.tibs.2018.10.011 de la Cruz-Lopez KG, Castro-Munoz LJ, Reyes-Hernandez DO, Garcia-Carranca A, Manzo-Merino J. Lactate in the Regulation of Tumor Microenvironment and Therapeutic Approaches. Front Oncol, 2019, 9:1143., DOI 10.3389/fonc.2019.01143 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609951 EP 1609959 DI 10.3389/pore.2021.1609951 PG 9 ER PT J AU Zhao, X Wu, Sh Jing, J AF Zhao, Xu Wu, Shuang Jing, Jingjing TI Identifying Diagnostic and Prognostic Biomarkers and Candidate Therapeutic Drugs of Gastric Cancer Based on Transcriptomics and Single-Cell Sequencing SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bioinformatics; biomarkers; prognosis; gastric cancer; hub genes; molecular drugs ID bioinformatics; biomarkers; prognosis; gastric cancer; hub genes; molecular drugs AB Background and Objective: Gastric cancer (GC) is an important health burden and the prognosis of GC is poor. We aimed to explore new diagnostic and prognostic indicators as well as potential therapeutic targets for GC in the current study. Methods: We screened the overlapped differentially expressed genes (DEGs) from GSE54129 and TCGA STAD datasets. Protein-protein interaction network analysis recognized the hub genes among the DEGs. The roles of these genes in diagnosis, prognosis, and their relationship with immune infiltrates and drug sensitivity of GC were analyzed using R studio. Finally, the clinically significant hub genes were verified using single-cell RNA sequencing (scRNA-seq) data. Results: A total of 222 overlapping genes were screened, which were enriched in extracellular matrix-related pathways. Further, 17 hub genes were identified, and our findings demonstrated that BGN, COMP, COL5A2, and SPARC might be important diagnostic and prognostic indicators of GC, which were also correlated with immune cell infiltration, tumor mutation burden (TMB), microsatellite instability (MSI), and sensitivity of therapeutic drugs. The scRNAseq results further confirmed that all four hub genes were highly expressed in GC. Conclusion: Based on transcriptomics and single-cell sequencing, we identified four diagnostic and prognostic biomarkers of GC, including BGN, COMP, COL5A2, and SPARC, which can help predict drug sensitivity for GC as well. C1 [Zhao, Xu] Liaoning Vocational College of Medicine, Mathematical Computer Teaching and Research OfficeShenyang, China. [Wu, Shuang] Changchun Normal University, College of Computer Science and TechnologyChangchun, China. [Jing, Jingjing] China Medical University, The First Affiliated Hospital, Department of General SurgeryShenyang, China. RP Jing, J (reprint author), China Medical University, The First Affiliated Hospital, Department of General Surgery, Shenyang, China. EM hellojjjing@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinic, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Van Cutsem E, Sagaert X, Topal B, Haustermans K, Prenen H. Gastric Cancer. Lancet, 2016, 388(10060):2654–64., DOI 10.1016/s0140-6736(16)30354-3 Song Z,Wu Y, Yang J, Yang D, Fang X. Progress in the Treatment of Advanced Gastric Cancer. Tumour Biol, 2017, 39(7):101042831771462., DOI 10.1177/ 1010428317714626 Chau I. Checkpoint Inhibition: an ATTRACTION in Advanced Gastric Cancer? Lancet, 2017, 390(10111):2418–9., DOI 10.1016/s0140-6736(17)32131-1 Rubinstein JC, Nicolson NG, Ahuja N. Next-generation Sequencing in the Management of Gastric and Esophageal Cancers. Surg Clin North Am, 2019, 99(3):511–27., DOI 10.1016/j.suc.2019.02.005 Liu X, Wu J, Zhang D, Bing Z, Tian J, Ni M, et al. Identification of Potential Key Genes Associated with the Pathogenesis and Prognosis of Gastric Cancer Based on Integrated Bioinformatics Analysis. Front Genet, 2018, 9:265., DOI 10.3389/fgene.2018.00265 Nie K, Shi L, Wen Y, Pan J, Li P, Zheng Z, et al. Identification of Hub Genes Correlated with the Pathogenesis and Prognosis of Gastric Cancer via Bioinformatics Methods. Minerva Med, 2020, 111:213., DOI 10.23736/s0026- 4806.19.06166-4 Goldman M, Craft B, Swatloski T, Cline M, Morozova O, Diekhans M, et al. The UCSC Cancer Genomics Browser: Update 2015. Nucleic Acids Res, 2015, 43(Database issue):D812–D817., DOI 10.1093/nar/gku1073 Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a Web Server for Cancer and normal Gene Expression Profiling and Interactive Analyses. Nucleic Acids Res, 2017, 45(W1):W98–W102., DOI 10.1093/nar/gkx247 Bardou P, Mariette J, Escudie F, Djemiel C, Klopp C. Jvenn: an Interactive Venn Diagram Viewer. BMC Bioinformatics, 2014, 15(1):293., DOI 10.1186/ 1471-2105-15-293 Gene Ontology Consortium. The Gene Ontology, GO, Project in 2006. Nucleic Acids Res, 2006, 34(Database issue):D322–6., DOI 10.1093/nar/gkj021 Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: New Perspectives on Genomes, Pathways, Diseases and Drugs. Nucleic Acids Res, 2017, 45(D1):D353–D361., DOI 10.1093/nar/gkw1092 Wang J, Vasaikar S, Shi Z, Greer M, Zhang B. WebGestalt 2017: a More Comprehensive, Powerful, Flexible and Interactive Gene Set Enrichment Analysis Toolkit. Nucleic Acids Res, 2017, 45(W1):W130–W137., DOI 10.1093/nar/gkx356 Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING V11: Protein-Protein Association Networks with Increased Coverage, Supporting Functional Discovery in Genome-wide Experimental Datasets. Nucleic Acids Res, 2019, 47(D1):D607–D613., DOI 10.1093/nar/gky1131 Su G, Morris JH, Demchak B, Bader GD. Biological Network Exploration with Cytoscape 3. Curr Protoc Bioinformatics, 2014, 47, 8.13.1–24., DOI 10.1002/ 0471250953.bi0813s47 Goldman MJ, Craft B, Hastie M, Repecka K, McDade F, Kamath A, et al. Visualizing and Interpreting Cancer Genomics Data via the Xena Platform. Nat Biotechnol, 2020, 38(6):675–8., DOI 10.1038/s41587-020-0546-8 Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. Cancer Res, 2017, 77(21):e108–e110., DOI 10.1158/0008-5472.can-17-0307 Tian C, Clauser KR, Ohlund D, Rickelt S, Huang Y, Gupta M, et al. Proteomic Analyses of ECM during Pancreatic Ductal Adenocarcinoma Progression Reveal Different Contributions by Tumor and Stromal Cells. Proc Natl Acad Sci USA, 2019, 116(39):19609–18., DOI 10.1073/pnas.1908626116 Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, et al. Matrix Crosslinking Forces Tumor Progression by Enhancing Integrin Signaling. Cell, 2009, 139(5):891–906., DOI 10.1016/j.cell.2009.10.027 McBride OW, Fisher LW, Young MF. Localization of PGI, Biglycan, BGN, and PGII, Decorin, DCN, PG-40, Genes onHuman Chromosomes Xq13-Qter and 12q, Respectively. Genomics, 1990, 6(2):219–25., DOI 10.1016/0888-7543(90)90560-h Jacobsen F, Kraft J, Schroeder C, Hube-MaggC,Kluth M, Lang DS, et al. Up-regulation of Biglycan Is Associated with Poor Prognosis and PTEN Deletion in Patients with Prostate Cancer. Neoplasia, 2017, 19(9):707–15., DOI 10.1016/j.neo.2017.06.003 Xing X, Gu X, Ma T, Ye H. Biglycan Up-Regulated Vascular Endothelial Growth Factor, VEGF, Expression and Promoted Angiogenesis in colon Cancer. Tumor Biol, 2015, 36(3):1773–80., DOI 10.1007/s13277-014-2779-y Hu L, Zang M-d., Wang H-x., Li J-f., Su L-p., Yan M, et al. Biglycan Stimulates VEGF Expression in Endothelial Cells by Activating the TLR Signaling Pathway. Mol Oncol, 2016, 10(9):1473–84., DOI 10.1016/j.molonc.2016.08.002 Wang B, Li G-X, Zhang S-G, Wang Q, Wen Y-G, Tang H-M, et al. Biglycan Expression Correlates with Aggressiveness and Poor Prognosis of Gastric Cancer. Exp Biol Med, Maywood,, 2011, 236(11):1247–53., DOI 10.1258/ ebm.2011.011124 Gundewar C, Sasor A, Hilmersson KS, Andersson R, Ansari D. The Role of SPARC Expression in Pancreatic Cancer Progression and Patient Survival. Scand J Gastroenterol, 2015, 50(9):1170–4., DOI 10.3109/00365521.2015.1024281 Che Y, Luo A, Wang H, Qi J, Guo J, Liu Z. The Differential Expression of SPARC in Esophageal Squamous Cell Carcinoma. Int J Mol Med, 2006, 17(6): 1027–33., DOI 10.3892/ijmm.17.6.1027 Wang J, Gao P, Song Y, Sun J, Chen X, Yu H, et al. Prognostic Value of Gastric Cancer-Associated Gene Signatures: Evidence Based on aMeta-Analysis Using Integrated Bioinformatics Methods. J Cel Mol Med, 2018, 22(11):5743–7., DOI 10.1111/jcmm.13823 Said N, Frierson HF, Sanchez-Carbayo M, Brekken RA, Theodorescu D. Loss of SPARC in Bladder Cancer Enhances Carcinogenesis and Progression. J Clin Invest, 2013, 123(2):751–66., DOI 10.1172/jci64782 Di Martino JF, Lacayo NJ, Varadi M, Li L, Saraiya C, Ravindranath Y, et al. Low or Absent SPARC Expression in Acute Myeloid Leukemia with MLL Rearrangements Is Associated with Sensitivity to Growth Inhibition by Exogenous SPARC Protein. Leukemia, 2006, 20(3):426–32., DOI 10.1038/ sj.leu.2404102 Tai IT, Tang MJ. SPARC in Cancer Biology: its Role in Cancer Progression and Potential for Therapy. Drug Resist Updates, 2008, 11(6):231–46., DOI 10.1016/ j.drup.2008.08.005 Li Z, Li A-D, Xu L, Bai D-W, Hou K-Z, Zheng H-C, et al. SPARC Expression in Gastric Cancer Predicts Poor Prognosis: Results from a Clinical Cohort, Pooled Analysis and GSEA Assay. Oncotarget, 2016, 7(43):70211–22., DOI 10.18632/oncotarget.12191 Wu D, Chen K, Bai Y, Zhu X, Chen Z, Wang C, et al. Screening of Diagnostic Markers for Osteosarcoma. Mol Med Rep, 2014, 10(5):2415–20., DOI 10.3892/ mmr.2014.2546 Zeng X-T, Liu X-P, Liu T-Z,Wang X-H. The Clinical Significance of COL5A2 in Patients with Bladder Cancer. Medicine, 2018, 97(10):e0091., DOI 10.1097/ md.0000000000010091 Wang Y. Transcriptional Regulatory Network Analysis for Gastric Cancer Based on mRNA Microarray. Pathol Oncol Res, 2017, 23(4):785–91., DOI 10.1007/s12253-016-0159-1 Liang Y, Zhang C, Dai D-Q. Identification of DNA Methylation- Regulated Differentially-Expressed Genes and Related Pathways Using Illumina 450K BeadChip and Bioinformatic Analysis in Gastric Cancer. Pathol Res Pract, 2019, 215(10):152570., DOI 10.1016/ j.prp.2019.152570 Zhou YY, Kang YT, Chen C, Xu FF, Wang HN, Jin R. Combination of TNM Staging and Pathway Based Risk Score Models in Patients with Gastric Cancer. J Cel Biochem., 2018, 119(4):3608–17., DOI 10.1002/jcb.26563 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609955 EP 1609968 DI 10.3389/pore.2021.1609955 PG 14 ER PT J AU Yang, ML Zhang, JH Li, Sh Zhu, R Wang, L AF Yang, Meng-Ling Zhang, Jia-Hua Li, Sheng Zhu, Rui Wang, Li TI SLC13A4 Might Serve as a Prognostic Biomarker and be Correlated with Immune Infiltration into Head and Neck Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; tumor-infiltrating immune cells; SLC13A4; head and neck squamous cell carcinoma; database analysis ID biomarker; tumor-infiltrating immune cells; SLC13A4; head and neck squamous cell carcinoma; database analysis AB SLC13A4 is a sodium sulfate co-transporter, which is expressed in brains, placentas, thymes and other tissues, plays an essential role in maintaining the metabolic balance of sulfate in vivo. The TCGA database shows that it is differentially expressed in a variety of tumors, but its prognostic value in tumors has not been clarified. TCGA, Oncomine and Timer databases were used to analyze SLC13A4 mRNA expression in cancer tissues and normal tissues, and its correlation with clinical prognosis in head and neck tumor. The CIBERSORT database was used to analyze the correlation between SLC13A4 expression and the infiltration of immune cells. SLC13A4 enrichment analysis was carried out by GSEA. SLC13A4 mRNA levels were significantly lower in head and neck tumors than in paracancer tissues. SLC13A4 expression in Head and neck squamous cell carcinoma (HNSCC) was closely related to tumor pathological grade and clinical stage. Decreased SLC13A4 expression was associated with poor overall survival (OS), progression free survival (PFS), disease specific survival (DSS) and recurrence free survival (RFS) in HNSCC patients. The expression of SLC13A4 was negatively correlated with Monocytes, M1 macrophages, M2 macrophages, resting CD4+ memory T cells, resting NK cells and activated NK cells, but positively correlated with neutrophils, plasma cells, T follicular helper cells, gamma delta T cells, regulatory T cells and naive B cells. In addition, the genes in SLC13A4 low-expression group were mainly concentrated in immunity-related activities, viral diseases, typical tumor pathways and metabolism. The SLC13A4 high expression group was mainly enriched in metabolic pathways. These suggest that SLC13A4 may be a potential prognostic biomarker in HNSC and correlated with immune infiltrates. C1 [Yang, Meng-Ling] Huazhong University of Science and Technology, Tongji Medical College, Union Hospital, Department of Integrated Traditional Chinese and Western MedicineWuhan, China. [Zhang, Jia-Hua] Huazhong University of Science and Technology, Tongji Medical College, Center of Stem cellWuhan, China. [Li, Sheng] Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Department of SurgeryWuhan, China. [Zhu, Rui] Huazhong University of Science and Technology, Tongji Medical College, Union Hospital, Department of Integrated Traditional Chinese and Western MedicineWuhan, China. [Wang, Li] Huazhong University of Science and Technology, Tongji Medical College, Union Hospital, Department of Emergency SurgeryWuhan, China. RP Zhu, R (reprint author), Huazhong University of Science and Technology, Tongji Medical College, Union Hospital, Department of Integrated Traditional Chinese and Western Medicine, Wuhan, China. EM zhur@hust.edu.cn CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer Incidence and Mortality Worldwide: Sources, Methods and Major Patterns in GLOBOCAN 2012. Int J Cancer, 2015, 136(5):E359–E386., DOI 10.1002/ ijc.29210 Miller KD, Fidler-Benaoudia M, Keegan TH, Hipp HS, Jemal A, and Siegel RL. Cancer Statistics for Adolescents and Young Adults, 2020. CA Cancer J Clin, 2020, 70(1):443–59., DOI 10.3322/caac.21637 Solomon B, Young RJ, and Rischin D. Head and Neck Squamous Cell Carcinoma: Genomics and Emerging Biomarkers for Immunomodulatory Cancer Treatments. Semin Cancer Biol, 2018, 52(Pt 2):228–40., DOI 10.1016/j.semcancer.2018.01.008 Peltanova B, Raudenska M, and Masarik M. Effect of Tumor Microenvironment on Pathogenesis of the Head and Neck Squamous Cell Carcinoma: a Systematic Review. Mol Cancer, 2019, 18(1):63., DOI 10.1186/ s12943-019-0983-5 Cohen EEW, Soulieres D, Le Tourneau C, Dinis J, Licitra L, Ahn MJ, et al. KEYNOTE-040 Investigators. Pembrolizumab versus Methotrexate, Docetaxel, or Cetuximab for Recurrent or Metastatic Head-And-Neck Squamous Cell Carcinoma, KEYNOTE-040): a Randomised, Open-Label, Phase 3 Study. Lancet, 2019, 393(10167):156–67., DOI 10.1016/S0140-6736(18)31999-8 Ferris RL, Blumenschein G, Jr, Fayette J, Guigay J, Colevas AD, Licitra L, et al. Nivolumab vs Investigator’s Choice in Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck: 2-year Long-Term Survival Update of CheckMate 141 with Analyses by Tumor PD-L1 Expression. Oral Oncol, 2018, 81:45–51., DOI 10.1016/j.oraloncology.2018.04.008 Bergeron MJ, Clemencon B, Hediger MA, and Markovich D. SLC13 Family of Na⁺-Coupled Di- and Tri-carboxylate/sulfate Transporters. Mol Aspects Med, 2013, 34(2-3):299–312., DOI 10.1016/j.mam.2012.12.001 Marutani E, Yamada M, Ida T, Tokuda K, Ikeda K, Kai S, et al. Thiosulfate Mediates Cytoprotective Effects of Hydrogen Sulfide against Neuronal Ischemia. J Am Heart Assoc, 2015, 4(11):e002125., DOI 10.1161/ JAHA.115.002125 Dawson PA, Pirlo KJ, Steane SE, Kunzelmann K, Chien YJ, and Markovich D. Molecular Cloning and Characterization of the Mouse Na+ Sulfate Cotransporter Gene, Slc13a4): Structure and Expression. Genes Genet Syst, 2006, 81(4):265–72., DOI 10.1266/ggs.81.265 Markovich D, Regeer RR, Kunzelmann K, and Dawson PA. Functional Characterization and Genomic Organization of the Human Na+-Sulfate Cotransporter hNaS2 Gene, SLC13A4). Biochem Biophysical Res Commun, 2005, 326(4):729–34., DOI 10.1016/j.bbrc.2004.11.102 Rakoczy J, Zhang Z, Bowling FG, Dawson PA, and Simmons DG. Loss of the Sulfate Transporter Slc13a4 in Placenta Causes Severe Fetal Abnormalities and Death in Mice. Cell Res, 2015, 25(11):1273–6., DOI 10.1038/cr.2015.100 Zhang Z, Aung ZT, Simmons DG, and Dawson PA.Molecular Analysis of Sequence and Splice Variants of the Human SLC13A4 Sulfate Transporter. Mol Genet Metab, 2017, 121(1):35–42., DOI 10.1016/ j.ymgme.2017.03.010 Hastbacka J, de la Chapelle A, Mahtani MM, Clines G, Reeve-Daly MP, Daly M, et al. The Diastrophic Dysplasia Gene Encodes a Novel Sulfate Transporter: Positional Cloning by fine-structure Linkage Disequilibrium Mapping. Cell, 1994, 78(6):1073–87., DOI 10.1016/0092-8674(94)90281-x Demydchuk M, Hill CH, Zhou A, Bunkoczi G, Stein PE, Marchesan D, et al. Insights into Hunter Syndrome from the Structure of Iduronate-2-Sulfatase. Nat Commun, 2017, 8:15786., DOI 10.1038/ncomms15786 Zhang Z, Dawson PA, Piper M, and Simmons DG. Postnatal N-Acetylcysteine Administration Rescues Impaired Social Behaviors and Neurogenesis in Slc13a4 Haploinsufficient Mice. EBioMedicine, 2019, 43:435–46., DOI 10.1016/j.ebiom.2019.03.081 Markovich D. Physiological Roles and Regulation of Mammalian Sulfate Transporters. Physiol Rev, 2001, 81(4):1499–533., DOI 10.1152/ physrev.2001.81.4.1499 Dawson PA. Role of Sulphate in Development. Reproduction, 2013, 146(3): R81–R89., DOI 10.1530/rep-13-0056 Chwatko G, Forma E, Wilkosz J, Glowacki R, Jozwiak P, Rozanski W, et al. Thiosulfate in Urine as a Facilitator in the Diagnosis of Prostate Cancer for Patients with Prostate-specific Antigen Less or Equal 10 ng/mL. Clin Chem Lab Med, 2013, 51(9):1825–31., DOI 10.1515/cclm-2013-0069 Baskar R, and Bian J. Hydrogen Sulfide Gas Has Cell Growth Regulatory Role. Eur J Pharmacol, 2011, 656(1-3):5–9., DOI 10.1016/j.ejphar.2011.01.052 Cai WJ, Wang MJ, Ju LH, Wang C, and Zhu YC. Hydrogen Sulfide Induces Human colon Cancer Cell Proliferation: Role of Akt, ERK and P21. Cell. Biol. Int., 2010, 34(6):565–72., DOI 10.1042/cbi20090368 Cao Q, Zhang L, Yang G, Xu C, and Wang R. Butyrate-stimulated H2S Production in colon Cancer Cells. Antioxid Redox Signaling, 2010, 12(9): 1101–9., DOI 10.1089/ars.2009.2915 Wang Z, Jensen MA, and Zenklusen JC. A Practical Guide to the Cancer Genome Atlas, TCGA). Methods Mol Biol, 2016, 1418:111–41., DOI 10.1007/ 978-1-4939-3578-9_6 Jiao Y, Fu Z, Li Y, Meng L, and Liu Y. High EIF2B5 mRNA Expression and its Prognostic Significance in Liver Cancer: A Study Based on the TCGA and GEO Database. Cmar, 2018, Vol. 10:6003–14., DOI 10.2147/cmar.s185459 Rhodes DR, Kalyana-Sundaram S, Mahavisno V, Varambally R, Yu J, Briggs BB, et al. Oncomine 3.0: Genes, Pathways, and Networks in a Collection of 18,000 Cancer Gene Expression Profiles. Neoplasia, 2007, 9(2):166–80., DOI 10.1593/neo.07112 Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. Cancer Res, 2017, 77(21):e108–e110., DOI 10.1158/0008-5472.can-17-0307 Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Tissue-based Map of the Human Proteome. Science, 2015, 347(6220): 1260419., DOI 10.1126/science.1260419 Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust Enumeration of Cell Subsets from Tissue Expression Profiles. Nat Methods, 2015, 12(5):453–7., DOI 10.1038/nmeth.3337 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: a Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci, 2005, 102(43):15545–50., DOI 10.1073/pnas.0506580102 Miyauchi S, Srinivas SR, Fei YJ, Gopal E, Umapathy NS, Wang H, et al. Functional Characteristics of NaS2, a Placenta-specific Na+-Coupled Transporter for Sulfate and Oxyanions of the Micronutrients Selenium and Chromium. Placenta, 2006, 27(6-7):550–9., DOI 10.1016/ j.placenta.2005.05.004 Liang B, Tao Y, and Wang T. Profiles of Immune Cell Infiltration in Head and Neck Squamous Carcinoma. Biosci Rep, 2020, 40(2):BSR20192724., DOI 10.1042/BSR20192724 Hanna GJ, Liu H, Jones RE, Bacay AF, Lizotte PH, Ivanova EV, et al. Defining an Inflamed Tumor Immunophenotype in Recurrent, Metastatic Squamous Cell Carcinoma of the Head and Neck. Oral Oncol, 2017, 67:61–9., DOI 10.1016/j.oraloncology.2017.02.005 Troiano G, Rubini C, Togni L, Caponio VCA, Zhurakivska K, Santarelli A, et al. The Immune Phenotype of Tongue Squamous Cell Carcinoma Predicts Early Relapse and Poor Prognosis. Cancer Med, 2020, 9(22):8333–44., DOI 10.1002/cam4.3440 Schoenfeld JD, Hanna GJ, Jo VY, Rawal B, Chen Y-H, Catalano PS, et al. Neoadjuvant Nivolumab or Nivolumab Plus Ipilimumab in Untreated Oral Cavity Squamous Cell Carcinoma. JAMA Oncol, 2020, 6(10):1563–70., DOI 10.1001/jamaoncol.2020.2955 Jiang H, Ni H, Zhang P, Guo X,Wu M, Shen H, et al. PD-L1/LAG-3 Bispecific Antibody Enhances Tumor-specific Immunity. Oncoimmunology, 2021, 10(1):1943180., DOI 10.1080/2162402x.2021.1943180 Liu J-F, Wu L, Yang L-L, Deng W-W, Mao L, Wu H, et al. Blockade of TIM3 Relieves Immunosuppression through Reducing Regulatory T Cells in Head and Neck Cancer. J Exp Clin Cancer Res, 2018, 37(1):44., DOI 10.1186/s13046- 018-0713-7 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609967 EP 1609981 DI 10.3389/pore.2021.1609967 PG 15 ER PT J AU Brunner, Sz Varga, D Bozo, R Polanek, R Tokes, T Szabo, RE Molnar, R Gemes, N Szebeni, G Puskas, L Erdelyi, M Hideghety, K AF Brunner, Szilvia Varga, Daniel Bozo, Renata Polanek, Robert Tokes, Tunde Szabo, Rita Emilia Molnar, Reka Gemes, Nikolett Szebeni, J. Gabor Puskas, G. Laszlo Erdelyi, Miklos Hideghety, Katalin TI Analysis of Ionizing Radiation Induced DNA Damage by Superresolution dSTORM Microscopy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ionizing radiation; γ-H2AX; DNA-DSB; confocal microscopy; dSTORM; superresolution ID ionizing radiation; γ-H2AX; DNA-DSB; confocal microscopy; dSTORM; superresolution AB The quantitative detection of radiation caused DNA double-strand breaks (DSB) by immunostained γ-H2AX foci using direct stochastic optical reconstruction microscopy (dSTORM) provides a deeper insight into the DNA repair process at nanoscale in a timedependent manner. Glioblastoma (U251) cells were irradiated with 250 keV X-ray at 0, 2, 5, 8 Gy dose levels. Cell cycle phase distribution and apoptosis of U251 cells upon irradiation was assayed by flow cytometry. We studied the density, topology and volume of the γ-H2AX foci with 3D confocal microscopy and the dSTORM superresolution method. A pronounced increase in γ-H2AX foci and cluster density was detected by 3D confocal microscopy after 2 Gy, at 30 min postirradiation, but both returned to the control level at 24 h. Meanwhile, at 24 h a considerable amount of residual foci could be measured from 5 Gy, which returned to the normal level 48 h later. The dSTORM based γ-H2AX analysis revealed that the micron-sized γ-H2AX foci are composed of distinct smaller units with a few tens of nanometers. The density of these clusters, the epitope number and the dynamics of γ-H2AX foci loss could be analyzed. Our findings suggest a discrete level of repair enzyme capacity and the restart of the repair process for the residual DSBs, even beyond 24 h. The dSTORM superresolution technique provides a higher precision over 3D confocal microscopy to study radiation induced γ-H2AX foci and molecular rearrangements during the repair process, opening a novel perspective for radiation research. C1 [Brunner, Szilvia] ELI-HU Non-Profit Ltd, ELI-ALPS Research Institute, Biomedical Applications GroupSzeged, Hungary. [Varga, Daniel] University of Szeged, Department of Optics and Quantum ElectronicsSzeged, Hungary. [Bozo, Renata] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Polanek, Robert] ELI-HU Non-Profit Ltd, ELI-ALPS Research Institute, Biomedical Applications GroupSzeged, Hungary. [Tokes, Tunde] ELI-HU Non-Profit Ltd, ELI-ALPS Research Institute, Biomedical Applications GroupSzeged, Hungary. [Szabo, Rita Emilia] ELI-HU Non-Profit Ltd, ELI-ALPS Research Institute, Biomedical Applications GroupSzeged, Hungary. [Molnar, Reka] ELI-HU Non-Profit Ltd, ELI-ALPS Research Institute, Biomedical Applications GroupSzeged, Hungary. [Gemes, Nikolett] Szegedi Biologiai Kutatokozpont, Funkcionalis Genomika LaboratoriumSzeged, Hungary. [Szebeni, J. Gabor] Szegedi Biologiai Kutatokozpont, Funkcionalis Genomika LaboratoriumSzeged, Hungary. [Puskas, G. Laszlo] Szegedi Biologiai Kutatokozpont, Funkcionalis Genomika LaboratoriumSzeged, Hungary. [Erdelyi, Miklos] University of Szeged, Department of Optics and Quantum ElectronicsSzeged, Hungary. [Hideghety, Katalin] ELI-HU Non-Profit Ltd, ELI-ALPS Research Institute, Biomedical Applications GroupSzeged, Hungary. RP Hideghety, K (reprint author), ELI-HU Non-Profit Ltd, ELI-ALPS Research Institute, Biomedical Applications Group, Szeged, Hungary. EM katalin.hideghety@eli-alps.hu CR Murad H, Alghamian Y, Aljapawe A, and Madania A. Effects of Ionizing Radiation on the Viability and Proliferative Behavior of the Human Glioblastoma T98G Cell Line. BMC Res Notes, 2018, 11:330., DOI 10.1186/ s13104-018-3438-y Man I, Szebeni GJ, Plangar I, Szabo ER, Tokes T, Szabo Z, et al..Novel Real- Time Cell Analysis Platform for the Dynamic Monitoring of Ionizing Radiation Effects on Human Tumor Cell Lines and Primary Fibroblasts. Mol Med Rep, 2015, 12:4610–9., DOI 10.3892/mmr.2015.4004 Sun D, Mu Y, and Piao H. MicroRNA-153-3p Enhances Cell Radiosensitivity by Targeting BCL2 in Human Glioma. Biol Res, 2018, 51:56., DOI 10.1186/ s40659-018-0203-6 Eccles LJ, O’Neill P, and Lomax ME. Delayed Repair of Radiation Induced Clustered DNA Damage: Friend or Foe? Mutat Res/Fundam Mol Mech Mutagen, 2011, 711:134–41., DOI 10.1016/j.mrfmmm.2010.11.003 Nickoloff JA, Sharma N, and Taylor L. Clustered DNA Double-Strand Breaks: Biological Effects and Relevance to Cancer Radiotherapy. Genes, 2020, 11:99., DOI 10.3390/genes11010099 Sage E, and Shikazono N. Radiation-induced Clustered DNA Lesions: Repair and Mutagenesis. Free Radic Biol Med, 2017, 107:125–35., DOI 10.1016/ j.freeradbiomed.2016.12.008 Rogakou EP, Boon C, Redon C, and Bonner WM. Megabase Chromatin Domains Involved in DNA Double-Strand Breaks In Vivo. J Cel Biol, 1999, 146:905–16., DOI 10.1083/jcb.146.5.905 Paull TT. Mechanisms of ATM Activation. Annu Rev Biochem, 2015, 84: 711–38., DOI 10.1146/annurev-biochem-060614-034335 Bekker-Jensen S, and Mailand N. Assembly and Function of DNA Double- Strand Break Repair Foci in Mammalian Cells. DNA Repair, 2010, 9:1219–28., DOI 10.1016/j.dnarep.2010.09.010 Podhorecka M, Skladanowski A, and Bozko P. H2AX Phosphorylation: Its Role in DNA Damage Response and Cancer Therapy. J Nucleic Acids, 2010, 2010:920161., DOI 10.4061/2010/920161 Vitor AC, Huertas P, Legube G, and de Almeida SF. Studying DNA Double- Strand Break Repair: An Ever-Growing Toolbox. Front Mol Biosci, 2020, 7:24., DOI 10.3389/fmolb.2020.00024 Lorat Y, Schanz S, Schuler N, Wennemuth G, Rube C, and Rube CE. Beyond Repair Foci: DNA Double-Strand Break Repair in Euchromatic and Heterochromatic Compartments Analyzed by Transmission Electron Microscopy. PLoS One, 2012, 7:e38165., DOI 10.1371/ journal.pone.0038165 Falk M, and Hausmann M. A Paradigm Revolution or Just Better Resolution—Will Newly Emerging Superresolution Techniques Identify Chromatin Architecture as a Key Factor in Radiation-Induced Dna Damage and Repair Regulation? Cancers, Basel,, 2021, 13:1–30., DOI 10.3390/cancers13010018 Jakob B, Dubiak-Szepietowska M, Janiel E, Schmidt A, Durante M, and Taucher-Scholz G. Differential Repair Protein Recruitment at Sites of Clustered and Isolated DNA Double-Strand Breaks Produced by High- Energy Heavy Ions. Sci Rep, 2020, 10:1443., DOI 10.1038/s41598-020- 58084-6 Rogakou EP, Pilch DR, Orr AH, Ivanova VS, and Bonner WM. DNA Double- Stranded Breaks Induce Histone H2AX Phosphorylation on Serine 139. J Biol Chem, 1998, 273:5858–68., DOI 10.1074/jbc.273.10.5858 Rothkamm K, and Horn S. Gamma-H2AX as Protein Biomarker for Radiation Exposure. Ann Ist Super Sanita, 2009, 45:265–71. Kruhlak MJ, Celeste A, and Nussenzweig A. Spatio-Temporal Dynamics of Chromatin Containing DNA Breaks. Cell Cycle, 2006, 5:1910–2., DOI 10.4161/ cc.5.17.3169 Pilch DR, Sedelnikova OA, Redon C, Celeste A, Nussenzweig A, and Bonner WM. Characteristics of γ-H2AX Foci at DNA Double-Strand Breaks Sites. Biochem Cel Biol., 2003, 81:123–9., DOI 10.1139/o03-042 Macphail SH, BanAth JP, Yu TY, Chu EHM, Lambur H, and Olive PL. Expression of Phosphorylated Histone H2AX in Cultured Cell Lines Following Exposure to X-rays. Int J Radiat Biol, 2003, 79:351–9., DOI 10.1080/ 0955300032000093128 Taneja N, Davis M, Choy JS, Beckett MA, Singh R, Kron SJ, et al..Histone H2AX Phosphorylation as a Predictor of Radiosensitivity and Target for Radiotherapy. J Biol Chem, 2004, 279:2273–80., DOI 10.1074/jbc.M310030200 Banath JP, MacPhail SH, and Olive PL. Radiation Sensitivity, H2AX Phosphorylation, and Kinetics of Repair of DNA Strand Breaks in Irradiated Cervical Cancer Cell Lines. Cancer Res, 2004, 64:7144–9., DOI 10.1158/0008-5472.CAN-04-1433 Sanders JT, Freeman TF, Xu Y, Golloshi R, Stallard MA, Hill AM, et al..Radiation-induced DNA Damage and Repair Effects on 3D Genome Organization. Nat Commun, 2020, 11:6178., DOI 10.1038/ s41467-020-20047-w Firsanov D, Vasilishina A, Kropotov A, and Mikhailov V. Dynamics of γH2AX Formation and Elimination in Mammalian Cells after X-Irradiation. Biochimie, 2012, 94:2416–22., DOI 10.1016/j.biochi.2012.06.019 HeilemannM, van de Linde SS, Schuttpelz M, Kasper R, Seefeldt B, Mukherjee A, et al..Subdiffraction-Resolution Fluorescence Imaging with Conventional Fluorescent Probes. Angew Chem Int Ed, 2008, 47:6172–6., DOI 10.1002/ anie.200802376 Varga D, Majoros H, Ujfaludi Z, Erdelyi M, and Pankotai T. Quantification of DNA Damage Induced Repair Focus Formation via Super-Resolution dSTORM Localization Microscopy. Nanoscale, 2019, 11:14226–36., DOI 10.1039/c9nr03696b Liddle P, Jara-Wilde J, Lafon-Hughes L, Castro I, Hartel S, and Folle G. dSTORM Microscopy Evidences in HeLa Cells Clustered and Scattered γH2AX Nanofoci Sensitive to ATM, DNA-PK, and ATR Kinase Inhibitors. Mol Cel Biochem, 2020, 473:77–91., DOI 10.1007/s11010-020- 03809-4 Endesfelder U, and Heilemann M. Direct Stochastic Optical Reconstruction Microscopy, Dstorm). Methods Mol Biol, 2014, 1251:263–76., DOI 10.1007/ 978-1-4939-2080-8_14 Polanek R, Varga Z, Fodor E, Brunner S, Szabo ER, Tokes T, et al..Improved FBX Chemical Dosimeter System with Enhanced Radiochemical Yield for Reference Dosimetry in Radiobiology and Radiotherapy Research. Radiat Phys Chem, 2020, 174:108899., DOI 10.1016/j.radphyschem.2020.108899 Kotogany E, Balog JA, Nagy LI, Alfoldi R, Bertagnolo V, Brugnoli F, et al. Imidazo[1,2-b]pyrazole-7-Carboxamide Derivative Induces Differentiation-Coupled Apoptosis of Immature Myeloid Cells Such as Acute Myeloid Leukemia and Myeloid-Derived Suppressor Cells. Ijms, 2020, 21:5135., DOI 10.3390/ijms21145135 File Exchange. Fatma Gargouri Thresholding the Maximum Entropy - File Exchange - MATLAB Central., 2012). Available at: https://www.mathworks. com/matlabcentral/fileexchange/35158-thresholding-the-maximum-entropy, Accessed Feb 22, 2021). Meyer F. Topographic Distance and Watershed Lines. Signal Process., 1994, 38:113–25., DOI 10.1016/0165-1684(94)90060-4 Bradley D, and Roth G. Adaptive Thresholding Using the Integral Image. J Graphics Tools, 2007, 12:13–21., DOI 10.1080/2151237x.2007.10129236 Soille P, and Soille P. Geodesic Transformations. In: Morphological Image Analysis. Springer Berlin Heidelberg, 1999). p. 155–83., DOI 10.1007/978-3- 662-03939-7_6 Maurer CR, Rensheng Qi R, and Raghavan V. A Linear Time Algorithm for Computing Exact Euclidean Distance Transforms of Binary Images in Arbitrary Dimensions. IEEE Trans Pattern Anal Machine Intell, 2003, 25: 265–70., DOI 10.1109/TPAMI.2003.1177156 rainSTORM. Advanced Optical Imaging Group., 2013).Available at: http://titan.physx.u-szeged.hu/∼adoptim/?page_id582, Accessed Feb 22, 2021). Erdelyi M, Rees E, Metcalf D, Schierle GSK, Dudas L, Sinko J, et al..Correcting Chromatic Offset in Multicolor Super-resolution Localization Microscopy. Opt Express, 2013, 21:10978., DOI 10.1364/oe.21.010978 Miller H, Zhou Z, Wollman AJM, and Leake MC. Superresolution Imaging of Single DNA Molecules Using Stochastic Photoblinking of Minor Groove and Intercalating Dyes. Methods, 2015, 88:81–8., DOI 10.1016/j.ymeth.2015.01.010 Rees EJ, Erdelyi M, Schierle GSK, Knight A, and Kaminski CF. Elements of Image Processing in Localization Microscopy. J Opt, 2013, 15:094012., DOI 10.1088/2040-8978/15/9/094012 Van De Linde S, Loschberger A, Klein T, Heidbreder M, Wolter S, Heilemann M, et al..Direct Stochastic Optical Reconstruction Microscopy with Standard Fluorescent Probes. Nat Protoc, 2011, 6:991–1009., DOI 10.1038/ nprot.2011.336 Ester M, Kriegel H-P, Sander J, and Xu X. A Density-Based Algorithm for Discovering Clusters in Large Spatial Databases with Noise. In: KDD’99: Procedings of the Second International Conference on Knowledge Discovery and Data Mining. Portland Oregon: AAAI Press, 1996). p. 226–231. Nieuwenhuizen RPJ, Bates M, Szymborska A, Lidke KA, Rieger B, and Stallinga S. Quantitative Localization Microscopy: Effects of Photophysics and Labeling Stoichiometry. PLoS One, 2015, 10:e0127989., DOI 10.1371/ journal.pone.0127989 Perez RL, Best G, Nicolay NH, Greubel C, Rossberger S, Reindl J, et al..Superresolution Light Microscopy Shows Nanostructure of Carbon Ion Radiation-induced DNA Double-strand Break Repair Foci. FASEB j., 2016, 30:2767–76., DOI 10.1096/FJ.201500106R Natale F, Rapp A, Yu W, Maiser A, Harz H, Scholl A, et al..Identification of the Elementary Structural Units of the DNA Damage Response. Nat Commun, 2017, 8:15760., DOI 10.1038/ncomms15760 Kuo LJ, and Yang LX. Gamma-H2AX - a Novel Biomarker for DNA Double- Strand Breaks. In Vivo, 2008, 22(3):305–9. Wu RS, Panusz HT, Hatch CL, and Bonner WM. Histones and Their Modification. Crit Rev Biochem, 1986, 20:201–63., DOI 10.3109/ 10409238609083735 Sisario D, Memmel S, Doose S, Neubauer J, Zimmermann H, Flentje M, et al..Nanostructure of DNA Repair Foci Revealed by Superresolution Microscopy. FASEB j., 2018, 32:6469–77., DOI 10.1096/fj.201701435 Martin OA, and Bonner WM. γ-H2AX in Cancer Cells: A Potential Biomarker for Cancer Diagnostics, Prediction and Recurrence. Cell Cycle, 2006, 5: 2909–13., DOI 10.4161/cc.5.24.3569 Wu R, Liu W, Sun Y, Shen C, Guo J, Zhao J, et al..Nanoscale Insight into Chromatin Remodeling and DNA Repair Complex in HeLa Cells after Ionizing Radiation. DNA Repair, 2020, 96:102974., DOI 10.1016/ j.dnarep.2020.102974 Staaf E, Brehwens K, Haghdoost S, Czub J, and Wojcik A. Gamma-H2AX Foci in Cells Exposed to a Mixed Beam of X-Rays and Alpha Particles. Genome Integr, 2012, 3:8., DOI 10.1186/2041-9414-3-8 Irianto J, Xia Y, Pfeifer CR, Athirasala A, Ji J, Alvey C, et al..DNA Damage Follows Repair Factor Depletion and Portends Genome Variation in Cancer Cells after Pore Migration. Curr Biol, 2017, 27:210–23., DOI 10.1016/ j.cub.2016.11.049 Pfeifer CR, Vashisth M, Xia Y, and Discher DE. Nuclear Failure, DNA Damage, and Cell Cycle Disruption after Migration through Small Pores: A Brief Review. Essays Biochem, 2019, 63:569–77., DOI 10.1042/ ebc20190007 Ruprecht N, Hungerbuhler MN, Bohm IB, and Heverhagen JT. Improved Identification of DNA Double Strand Breaks: γ-H2AX-epitope Visualization by Confocal Microscopy and 3D Reconstructed Images. Radiat Environ Biophys, 2019, 58:295–302., DOI 10.1007/s00411-019-00778-1 Anderson DL, Mirzayans R, Andrais B, Siegbahn EA, Fallone BG, and Warkentin B. Spatial and Temporal Distribution of γH2AX Fluorescence in Human Cell Cultures Following Synchrotron-Generated X-ray Microbeams: Lack of Correlation between Persistent γH2AX Foci and Apoptosis. J Synchrotron Radiat, 2014, 21:801–10., DOI 10.1107/ S1600577514011424 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609971 EP 1609983 DI 10.3389/pore.2021.1609971 PG 13 ER PT J AU Tan, Sh Fu, X Xu, Sh Qiu, P Lv, Z Xu, Y Zhang, Q AF Tan, Shirong Fu, Xin Xu, Shouping Qiu, Pengfei Lv, Zhidong Xu, Yingying Zhang, Qiang TI Quantification of Ki67 Change as a Valid Prognostic Indicator of Luminal B Type Breast Cancer After Neoadjuvant Therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; prognosis; neoadjuvant chemotherapy; Ki67; tumor response ID breast cancer; prognosis; neoadjuvant chemotherapy; Ki67; tumor response AB Introduction: Ki67 value and its variation before and after neoadjuvant chemotherapy are commonly tested in relation to breast cancer patient prognosis. This study aims to quantify the extent of changes in Ki67 proliferation pre- and post-neoadjuvant chemotherapy, confirm an optimal cut-off point, and evaluate its potential value for predicting survival outcomes in patients with different molecular subtypes of breast cancer. Methods: This retrospective real-world study recruited 828 patients at the Department of Breast Surgery of the First Affiliated Hospital of China Medical University and the Cancer Hospital of China Medical University from Jan 2014 to Nov 2020. Patient demographic features and disease pathology characteristics were recorded, and biomarkers were verified through immunohistochemistry. Various statistical methods were used to validate the relationships between different characteristics and survival outcomes irrespective of disease-free and overall survival. Results: Among 828 patients, statistically significant effects between pathological complete response and survival outcome were found in both HER2-enriched and triple-negative breast cancer (p < 0.05) but not in Luminal breast cancer (p > 0.05). Evident decrease of Ki67 was confirmed after neoadjuvant chemotherapy. To quantify the extent of Ki67 changes between pre- and post-NAC timepoints, we adopted a computational equation termed ΔKi67% for research. We found the optimal cut-off value to be “ΔKi67% = −63%” via the operating characteristic curve, defining ΔKi67% ≤ −63% as positive status and ΔKi67% > −63% as negative status. Patients with positive ΔKi67% status were 37.1% of the entire cohort. Additionally, 4.7, 39.9, 34.5 and 39.6% of patients with Luminal A, Luminal B, HER2-enriched and triple negative breast cancer were also validated with positive ΔKi67% status. The statistically significant differences between ΔKi67% status and prognostic outcomes were confirmed by univariate and multivariate analysis in Luminal B (univariate and multivariate analysis: p < 0.05) and triple negative breast cancer (univariate and multivariate analysis: p < 0.05). We proved ΔKi67% as a statistically significant independent prognostic factor irrespective of disease-free or overall survival among patients with Luminal B and triple-negative breast cancer. Conclusions: ΔKi67% can aid in predicting patient prognostic outcome, provide a measurement of NAC efficacy, and assist in further clinical decisions, especially for patients with Luminal B breast cancer. C1 [Tan, Shirong] The First Affiliated Hospital of China Medical University, Department of Breast SurgeryShenyang, China. [Fu, Xin] Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Department of Breast SurgeryShenyang, China. [Xu, Shouping] Harbin Medical University Cancer Hospital, Department of Breast SurgeryHarbin, China. [Qiu, Pengfei] Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Breast Cancer CenterJinan, China. [Lv, Zhidong] The Affiliated Hospital of Qingdao University, Breast CenterQingdao, China. [Xu, Yingying] The First Affiliated Hospital of China Medical University, Department of Breast SurgeryShenyang, China. [Zhang, Qiang] Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Department of Breast SurgeryShenyang, China. RP Zhang, Q (reprint author), Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Department of Breast Surgery, Shenyang, China. EM zhangqiang8220@163.com CR Siegel RL,Miller KD, Jemal A, et al. Cancer Statistics, 2020. CA A Cancer J Clin, 2020, 70(1):7–30., DOI 10.3322/caac.21590 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, et al. Personalizing the Treatment of Women With Early Breast Cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol, 2013, 24(9): 2206–23., DOI 10.1093/annonc/mdt303 Cancer Genome Atlas N. Comprehensive Molecular Portraits of Human Breast Tumours. Nature, 2012, 490(7418):61–70., DOI 10.1038/nature11412 Van de Wiel M, Dockx Y, Van den Wyngaert T, Stroobants S, Tjalma WAA, and Huizing MT. Neoadjuvant Systemic Therapy in Breast Cancer: Challenges and Uncertainties. Eur J Obstet Gynecol Reprod Biol, 2017, 210:144–56., DOI 10.1016/j.ejogrb.2016.12.014 von Minckwitz G, Blohmer JU, Costa SD, Denkert C, Eidtmann H, Eiermann W, et al. Response-Guided Neoadjuvant Chemotherapy for Breast Cancer. Jco, 2013, 31(29):3623–30., DOI 10.1200/JCO.2012.45.0940 Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological Complete Response and Long-Term Clinical Benefit in Breast Cancer: the CTNeoBC Pooled Analysis. The Lancet, 2014, 384(9938):164–72., DOI 10.1016/s0140-6736(13)62422-8 Wang-Lopez Q, Chalabi N, Abrial C, Radosevic-Robin N, Durando X, Mouret- Reynier M-A, et al. Can Pathologic Complete Response, pCR, Be Used as a Surrogate Marker of Survival After Neoadjuvant Therapy for Breast Cancer? Crit Rev Oncology/Hematology, 2015, 95(1):88–104., DOI 10.1016/ j.critrevonc.2015.02.011 von Minckwitz G, Fontanella C, et al. Comprehensive Review on the Surrogate Endpoints of Efficacy Proposed or Hypothesized in the Scientific Community Today. JNCI Monogr, 2015, 2015(51):29–31., DOI 10.1093/jncimonographs/ lgv007 Rodrigues-Ferreira S, Nehlig A, Moindjie H, Monchecourt C, Seiler C, Marangoni E, et al. Improving Breast Cancer Sensitivity to Paclitaxel by Increasing Aneuploidy. Proc Natl Acad Sci USA, 2019, 116(47):23691–7., DOI 10.1073/pnas.1910824116 Li L, Han D,Wang X, Wang Q, Tian J, Yao J, et al. Prognostic Values of Ki-67 in Neoadjuvant Setting for Breast Cancer: a Systematic Review and Meta- Analysis. Future Oncol, 2017, 13(11):1021–34., DOI 10.2217/fon-2016-0428 von Minckwitz G, Untch M, Blohmer J-U, Costa SD, Eidtmann H, Fasching PA, et al. Definition and Impact of Pathologic Complete Response on Prognosis After Neoadjuvant Chemotherapy in Various Intrinsic Breast Cancer Subtypes. Jco, 2012, 30(15):1796–804., DOI 10.1200/JCO.2011.38.8595 Fasching PA, Heusinger K, Haeberle L, Niklos M, Hein A, Bayer CM, et al. Ki67, Chemotherapy Response, and Prognosis in Breast Cancer Patients Receiving Neoadjuvant Treatment. BMC Cancer, 2011, 11:486., DOI 10.1186/1471-2407-11-486 Leung SCY, Nielsen TO, Nielsen TO, Zabaglo L, Arun I, Badve SS, et al. Analytical Validation of a Standardized Scoring Protocol for Ki67: Phase 3 of an International Multicenter Collaboration. NPJ Breast Cancer, 2016, 2:16014., DOI 10.1038/npjbcancer.2016.14 Andre F, Arnedos M, Goubar A, Ghouadni A, and Delaloge S. Ki67-no Evidence for its Use in Node-Positive Breast Cancer. Nat Rev Clin Oncol, 2015, 12(5):296–301., DOI 10.1038/nrclinonc.2015.46 Yoshioka T, Hosoda M, Yamamoto M, Taguchi K, Hatanaka KC, Takakuwa E, et al. Prognostic Significance of Pathologic Complete Response and Ki67 Expression after Neoadjuvant Chemotherapy in Breast Cancer. Breast Cancer, 2015, 22(2):185–91., DOI 10.1007/s12282-013-0474-2 Nishimura R, Osako T, Nishiyama Y, Tashima R, Nakano M, Fujisue M, et al. Prognostic Significance of Ki-67 Index Value at the Primary Breast Tumor in Recurrent Breast Cancer. Mol Clin Oncol, 2014, 2(6):1062–8., DOI 10.3892/ mco.2014.400 Curigliano G, Burstein HJ, Winer EP, Gnant M, Dubsky P, Loibl S, et al. De- Escalating and Escalating Treatments for Early-Stage Breast Cancer: the St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol, 2017, 28(8):1700–12., DOI 10.1093/ annonc/mdx308 Criscitiello C, Disalvatore D, De Laurentiis M, Gelao L, Fumagalli L, Locatelli M, et al. High Ki-67 Score Is Indicative of a Greater Benefit From Adjuvant Chemotherapy When Added to Endocrine Therapy in Luminal B HER2 Negative and Node-Positive Breast Cancer. The Breast, 2014, 23(1):69–75., DOI 10.1016/j.breast.2013.11.007 Smith I, Robertson J, Kilburn L,WilcoxM, Evans A,Holcombe C, et al. Long-Term Outcome and Prognostic Value of Ki67 After Perioperative Endocrine Therapy in PostmenopausalWomenWithHormone-Sensitive Early Breast Cancer, POETIC): an Open-Label, Multicentre, Parallel-Group, Randomised, Phase 3 Trial. Lancet Oncol, 2020, 21(11):1443–54., DOI 10.1016/s1470-2045(20)30458-7 Ianza A, Giudici F, Pinello C, Corona S, Strina C, Bernocchi O, et al. ΔKi67 Proliferation Index as Independent Predictive and Prognostic Factor of Outcome in Luminal Breast Cancer: Data from Neoadjuvant Letrozole-Based Treatment. Tumour Biol, 2020, 42(6):101042832092530., DOI 10.1177/1010428320925301 Bazzola L, Foroni C, Andreis D, Zanoni V, Milani M, Cappelletti G, et al. Combination of Letrozole, Metronomic Cyclophosphamide and Sorafenib Is Well-Tolerated and Shows Activity in Patients With Primary Breast Cancer. Br J Cancer, 2015, 112(1):52–60., DOI 10.1038/bjc.2014.563 Tanei T, Shimomura A, Shimazu K, Nakayama T, Kim SJ, Iwamoto T, et al. Prognostic Significance of Ki67 Index after Neoadjuvant Chemotherapy in Breast Cancer. Eur J Surg Oncol, Ejso,, 2011, 37(2):155–61., DOI 10.1016/ j.ejso.2010.10.009 Avci N, Deligonul A, Tolunay S, Cubukcu E, Fatih Olmez O, Ulas A, et al. Neoadjuvant Chemotherapy-Induced Changes in Immunohistochemical Expression of Estrogen Receptor, Progesterone Receptor, HER2, and Ki-67 in Patients with Breast Cancer. J BUON, 2015, 20(1):45–9. https://pubmed. ncbi.nlm.nih.gov/25778295/#affiliation-1. Kim KI, Lee KH, Kim TR, Chun YS, Lee TH, and Park HK. Ki-67 as a Predictor of Response to Neoadjuvant Chemotherapy in Breast Cancer Patients. J Breast Cancer, 2014, 17(1):40–6., DOI 10.4048/jbc.2014.17.1.40 Chen R, Ye Y, Yang C, Peng Y, Zong B, Qu F, et al. Assessment of the Predictive Role of Pretreatment Ki-67 and Ki-67 Changes in Breast Cancer Patients Receiving Neoadjuvant Chemotherapy According to the Molecular Classification: a Retrospective Study of 1010 Patients. Breast Cancer Res Treat, 2018, 170(1):35–43., DOI 10.1007/s10549-018-4730-1 Guarneri V, Piacentini F, Ficarra G, Frassoldati A, D’Amico R, Giovannelli S, et al. A Prognostic Model Based on Nodal Status and Ki-67 Predicts the Risk of Recurrence and Death in Breast Cancer Patients With Residual Disease After Preoperative Chemotherapy. Ann Oncol, 2009, 20(7):1193–8., DOI 10.1093/ annonc/mdn761 Wolff AC, Hammond MEH, Hicks DG, Dowsett M, McShane LM, Allison KH, et al. Recommendations for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Update. Arch Pathol Lab Med, 2014, 138(2):241–56., DOI 10.5858/arpa.2013-0953-SA Hammond MEH, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for Immunohistochemical Testing of Estrogen and Progesterone Receptors in Breast Cancer. Jco, 2010, 28(16):2784–95., DOI 10.1200/JCO.2009.25.6529 Kobayashi T, Iwaya K, Moriya T, Yamasaki T, Tsuda H, Yamamoto J, et al. A Simple Immunohistochemical Panel Comprising 2 Conventional Markers, Ki67 and P53, Is a Powerful Tool for Predicting Patient Outcome in Luminal-Type Breast Cancer. BMC Clin Pathol, 2013, 13:5., DOI 10.1186/1472-6890-13-5 Nielsen TO, Leung SCY, Rimm DL, Dodson A, Acs B, Badve S, et al. Assessment of Ki67 in Breast Cancer: Updated Recommendations From the International Ki67 in Breast Cancer Working Group. J Natl Cancer Inst, 2021, 113(7):808–19., DOI 10.1093/jnci/djaa201 Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn H-J, et al. Strategies for Subtypes-Dealing With the Diversity of Breast Cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol, 2011, 22(8):1736–47., DOI 10.1093/annonc/mdr304 Schemper M, and Smith TL. A Note on Quantifying Follow-Up in Studies of Failure Time. Controlled Clin Trials, 1996, 17(4):343–6., DOI 10.1016/0197- 2456(96)00075-x Schott AF, and Hayes DF. Defining the Benefits of Neoadjuvant Chemotherapy for Breast Cancer. Jco, 2012, 30(15):1747–9., DOI 10.1200/ jco.2011.41.3161 Ellis MJ, Suman VJ, Hoog J, Goncalves R, Sanati S, Creighton CJ, et al. Ki67 Proliferation Index as a Tool for Chemotherapy Decisions During and After Neoadjuvant Aromatase Inhibitor Treatment of Breast Cancer: Results from the American College of Surgeons Oncology Group Z1031 Trial, Alliance). Jco, 2017, 35(10):1061–9., DOI 10.1200/JCO.2016.69.4406 Tao M, Chen S, Zhang X, and Zhou Q. Ki-67 Labeling index Is a Predictive Marker for a Pathological Complete Response to Neoadjuvant Chemotherapy in Breast Cancer. Medicine, Baltimore,, 2017, 96(51):e9384., DOI 10.1097/ MD.0000000000009384 Cabrera-Galeana P, Munoz-Montano W, Lara-Medina F, Alvarado-Miranda A, Perez-Sanchez V, Villarreal-Garza C, et al. Ki67 Changes Identify Worse Outcomes in Residual Breast Cancer Tumors after Neoadjuvant Chemotherapy. Oncol, 2018, 23(6):670–8., DOI 10.1634/theoncologist.2017- 0396 Acs B, Zambo V, Vizkeleti L, Szasz AM, Madaras L, Szentmartoni G, et al. Ki- 67 as a Controversial Predictive and Prognostic Marker in Breast Cancer Patients Treated With Neoadjuvant Chemotherapy. Diagn Pathol, 2017, 12(1):20., DOI 10.1186/s13000-017-0608-5 Kim HS, Park S, Koo JS, Kim S, Kim JY, Nam S, et al. Risk Factors Associated With Discordant Ki-67 Levels Between Preoperative Biopsy and Postoperative Surgical Specimens in Breast Cancers. PLoS One, 2016, 11(3):e0151054., DOI 10.1371/journal.pone.0151054 Keam B, Im S-A, Lee K-H, Han S-W, Oh D-Y, Kim JH, et al. Ki-67 Can Be Used for Further Classification of Triple Negative Breast Cancer Into Two Subtypes With Different Response and Prognosis. Breast Cancer Res, 2011, 13(2):R22., DOI 10.1186/bcr2834 Munzone E, Botteri E, Sciandivasci A, Curigliano G, Nole F, Mastropasqua M, et al. Prognostic Value of Ki-67 Labeling Index in Patients With Node- Negative, Triple-Negative Breast Cancer. Breast Cancer Res Treat, 2012, 134(1):277–82., DOI 10.1007/s10549-012-2040-6 Battisti NML, True V, Chaabouni N, Chopra N, Lee K, Shepherd S, et al. Pathological Complete Response to Neoadjuvant Systemic Therapy in 789 Early and Locally Advanced Breast Cancer Patients: The Royal Marsden Experience. Breast Cancer Res Treat, 2020, 179(1):101–11., DOI 10.1007/ s10549-019-05444-0 Tyanova S, Albrechtsen R, Kronqvist P, Cox J, Mann M, Geiger T, et al. Proteomic Maps of Breast Cancer Subtypes. Nat Commun, 2016, 7:10259., DOI 10.1038/ncomms10259 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609972 EP 1609987 DI 10.3389/pore.2021.1609972 PG 16 ER PT J AU Guo, Y Liu, Y Yang, H Dai, N Zhou, F Yang, H Sun, W Kong, J Yuan, X Gao, Sh AF Guo, Yibo Liu, Yiwen Yang, Haijun Dai, Ningtao Zhou, Fuyou Yang, Hong Sun, Wei Kong, Jinyu Yuan, Xiang Gao, Shegan TI Associations of Porphyromonas gingivalis Infection and Low Beclin1 Expression With Clinicopathological Parameters and Survival of Esophageal Squamous Cell Carcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE P.gingivalis; ESCC; Autophagy; Beclin1; Prognosis ID P.gingivalis; ESCC; Autophagy; Beclin1; Prognosis AB Purpose: The present study focused on exploring the associations of Porphyromonas gingivalis (P. gingivalis) infection and low Beclin1 expression with clinicopathological parameters and survival of esophageal squamous cell carcinoma (ESCC) patients, so as to illustrate its clinical significance and prognostic value. Methods: Immunohistochemistry (IHC) was used to detect P. gingivalis infection status and Beclin1 expression in 370 ESCC patients. The chi-square test was adopted to illustrate the relationship between categorical variables, and Cohen’s kappa coefficient was used for correlation analysis. Kaplan-Meier survival curves with the log-rank test were used to analyse the correlation of P. gingivalis infection and low Beclin1 expression with survival time. The effects of P. gingivalis infection and Beclin1 downregulation on the proliferation, migration and antiapoptotic abilities of ESCC cells in vitro were detected by Cell Counting Kit-8, wound healing and flow cytometry assays. For P. gingivalis infection of ESCC cells, cell culture medium was replaced with antibiotic-free medium when the density of ESCC cells was 70–80%, cells were inoculated with P. gingivalis at a multiplicity of infection (MOI) of 10. Result: P. gingivalis infection was negatively correlated with Beclin1 expression in ESCC tissues, and P. gingivalis infection and low Beclin1 expression were associated with differentiation status, tumor invasion depth, lymph node metastasis, clinical stage and prognosis in ESCC patients. In vitro experiments confirmed that P. gingivalis infection and Beclin1 downregulation potentiate the proliferation, migration and antiapoptotic abilities of ESCC cells (KYSE150 and KYSE30). Our results provide evidence that P. gingivalis infection and low Beclin1 expression were associated with the development and progression of ESCC. Conclusion: Long-term smoking and alcohol consumption causes poor oral and esophageal microenvironments and ESCC patients with these features were more susceptible to P. gingivalis infection and persistent colonization, and exhibited lower Beclin1 expression, worse prognosis and more advanced clinicopathological features. Our findings indicate that effectively eliminating P. gingivalis colonization and restoring Beclin1 expression in ESCC patients may contribute to preventation and targeted treatment, and yield new insights into the aetiological research on ESCC. C1 [Guo, Yibo] Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer InstituteLuoyang, China. [Liu, Yiwen] Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer InstituteLuoyang, China. [Yang, Haijun] Anyang Tumor Hospital, Department of Thoracic SurgeryAnyang, China. [Dai, Ningtao] Anyang Tumor Hospital, Department of Thoracic SurgeryAnyang, China. [Zhou, Fuyou] Anyang Tumor Hospital, Department of Thoracic SurgeryAnyang, China. [Yang, Hong] Henan University of Science and Technology, School of PELuoyang, China. [Sun, Wei] Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer InstituteLuoyang, China. [Kong, Jinyu] Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer InstituteLuoyang, China. [Yuan, Xiang] Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer InstituteLuoyang, China. [Gao, Shegan] Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer InstituteLuoyang, China. RP Gao, Sh (reprint author), Henan University of Science and Technology, College of Clinical Medicine, The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, Luoyang, China. EM gsg112258@163.com CR Abnet CC, Arnold M, Wei W-Q. Epidemiology of Esophageal Squamous Cell Carcinoma. Gastroenterology, 2018, 154:360–73., DOI 10.1053/j.gastro.2017.08.023 Lin Y, Totsuka Y, He Y, Kikuchi S, Qiao Y, Ueda J. Epidemiology of Esophageal Cancer in Japan and China. J Epidemiol, 2013, 23:233–42., DOI 10.2188/ jea.je20120162 Bird-Lieberman EL, Fitzgerald RC. Early Diagnosis of Oesophageal Cancer. Br J Cancer, 2009, 101:1–6., DOI 10.1038/sj.bjc.6605126 Zhang SW, Zheng RS, Zuo TT, Zeng HM, Chen WQ, He J. Mortality and Survival Analysis of Esophageal Cancer in China. Zhonghua Zhong Liu Za Zhi, 2016, 38:709–15., DOI 10.3760/cma.j.issn.0253-3766.2016.09.014 Freedman ND, Abnet CC, Leitzmann MF,Mouw T, Subar AF, Hollenbeck AR. A Prospective Study of Tobacco, Alcohol, and the Risk of Esophageal and Gastric Cancer Subtypes. Am J Epidemiol, 2007, 165:1424–33., DOI 10.1093/aje/ kwm051 Wang Y-P, Han X-Y, Su W, Wang Y-L, Zhu Y-W, Sasaba T. Esophageal Cancer in Shanxi Province, People’s Republic of China: a Case-Control Study in High and Moderate Risk Areas. Cancer Causes Control, 1992, 3:107–13., DOI 10.1007/bf00051650 Chen X, Winckler B, Lu M, Cheng H, Yuan Z, Yang Y. Oral Microbiota and Risk for Esophageal Squamous Cell Carcinoma in a High-Risk Area of China. PLoS One, 2015, 10:e0143603., DOI 10.1371/journal.pone.0143603 Gao S, Li S, Ma Z, Liang S, Shan T, Zhang M. Presence of Porphyromonas Gingivalis in Esophagus and its Association with the Clinicopathological Characteristics and Survival in Patients with Esophageal Cancer. Infect Agents Cancer, 2016, 11:3., DOI 10.1186/s13027-016-0049-x Yuan X, Liu Y, Kong J, Gu B, Qi Y, Wang X. Different Frequencies of Porphyromonas Gingivalis Infection in Cancers of the Upper Digestive Tract. Cancer Lett, 2017, 404:1–7., DOI 10.1016/j.canlet.2017.07.003 Gao S, Liu Y, Duan X, Liu K, MohammedM, Gu Z. Porphyromonas Gingivalis Infection Exacerbates Oesophageal Cancer and Promotes Resistance to Neoadjuvant Chemotherapy. Br J Cancer, 2021, 125:433–44., DOI 10.1038/ s41416-021-01419-5 Dorn BR, Dunn WA, Jr, Progulske-Fox A. Bacterial Interactions with the Autophagic Pathway. Cell Microbiol, 2002, 4:1–10., DOI 10.1046/j.1462- 5822.2002.00164.x Mukhopadhyay S, Panda PK, Sinha N, Das DN, Bhutia SK. Autophagy and Apoptosis: where Do They Meet? Apoptosis, 2014, 19:555–66., DOI 10.1007/ s10495-014-0967-2 Mizushima N. Autophagy: Process and Function. Genes Dev, 2007, 21: 2861–73., DOI 10.1101/gad.1599207 Galluzzi L, Pietrocola F, Bravo-San Pedro JM, Amaravadi RK, Baehrecke EH, Cecconi F. Autophagy in Malignant Transformation and Cancer Progression. Embo j, 2015, 34:856–80., DOI 10.15252/embj.201490784 Xu H-D, Qin Z-H. Beclin 1, Bcl-2 and Autophagy. Adv Exp Med Biol, 2019, 1206:109–26., DOI 10.1007/978-981-15-0602-4_5 Liang C, Feng P, Ku B, Dotan I, Canaani D, Oh B-H. Autophagic and Tumour Suppressor Activity of a Novel Beclin1-Binding Protein UVRAG. Nat Cel Biol, 2006, 8:688–98., DOI 10.1038/ncb1426 Heras-Sandoval D, Perez-Rojas JM, Pedraza-Chaverri J. Novel Compounds for the Modulation of mTOR and Autophagy to Treat Neurodegenerative Diseases. Cell Signal, 2020, 65:109442., DOI 10.1016/j.cellsig.2019.109442 Du H, Luo F, Shi M, Che J, Zhu L, Li H. Beclin-1 Is a Promising Prognostic Biomarker in a Specific Esophageal Squamous Cell Carcinoma Population. Pathol Oncol Res, 2021, 27:594724., DOI 10.3389/pore.2021.594724 Li X, Gong Z, Zhang L, Zhao C, Zhao X, Gu X. Autophagy Knocked Down by High-Risk HPV Infection and Uterine Cervical Carcinogenesis. Int J Clin Exp Med, 2015, 8:10304–14. Zhang F, Chen C, Hu J, Su R, Zhang J, Han Z. Molecular Mechanism of Helicobacter Pylori-Induced Autophagy in Gastric Cancer. Oncol Lett, 2019, 18:6221–7., DOI 10.3892/ol.2019.10976 Hu Z, Zhong Z,Huang S, WenH, Chen X, Chu H. Decreased Expression of Beclin- 1 Is Significantly Associated with a Poor Prognosis in Oral Tongue Squamous Cell Carcinoma. Mol Med Rep, 2016, 14:1567–73., DOI 10.3892/mmr.2016.5437 Weh KM, Howell AB, Kresty LA. Expression, Modulation, and Clinical Correlates of the Autophagy Protein Beclin-1 in Esophageal Adenocarcinoma. Mol Carcinog, 2016, 55:1876–85., DOI 10.1002/mc.22432 Chen Y-P, Zhao B-C, Chen C, Lei X-X, Shen L-J, Chen G. Alcohol Drinking as an Unfavorable Prognostic Factor for Male Patients with Nasopharyngeal Carcinoma. Sci Rep, 2016, 6:19290., DOI 10.1038/srep19290 Shi J, Yao D, Liu W, Wang N, Lv H, Zhang G. Highly Frequent PIK3CA Amplification Is Associated with Poor Prognosis in Gastric Cancer. BMC Cancer, 2012, 12:50., DOI 10.1186/1471-2407-12-50 Yilmaz O, Young PA, Lamont RJ, Kenny GE. Gingival Epithelial Cell Signalling and Cytoskeletal Responses to PorphyromonasGingivalis Invasion.Microbiology, Reading,, 2003, 149:2417–26., DOI 10.1099/mic.0.26483-0 Chen H-I, Tsai H-P, Chen Y-T, Tsao S-C, Chai C-Y. Autophagy and Apoptosis Play Opposing Roles in Overall Survival of Esophageal Squamous Cell Carcinoma. Pathol Oncol Res, 2016, 22:699–705., DOI 10.1007/s12253-016-0051-z Qi Y-J, Jiao Y-L, Chen P, Kong J-Y, Gu B-L, Liu K. Porphyromonas Gingivalis Promotes Progression of Esophageal Squamous Cell Cancer via TGFβ- dependent Smad/YAP/TAZ Signaling. Plos Biol, 2020, 18:e3000825., DOI 10.1371/journal.pbio.3000825 McHugh ML. The Chi-Square Test of independence. Biochem Med, 2013, 23: 143–9., DOI 10.11613/bm.2013.018 Hongo M, Nagasaki Y, Shoji T. Epidemiology of Esophageal Cancer: Orient to Occident. Effects of Chronology, Geography and Ethnicity. J Gastroenterol Hepatol, 2009, 24:729–35., DOI 10.1111/j.1440-1746.2009.05824.x Lin Y, Totsuka Y, Shan B, Wang C, Wei W, Qiao Y. Esophageal Cancer in High-Risk Areas of China: Research Progress and Challenges. Ann Epidemiol, 2017, 27:215–21., DOI 10.1016/j.annepidem.2016.11.004 Ahn J, Segers S, Hayes RB. Periodontal Disease, Porphyromonas Gingivalis Serum Antibody Levels and Orodigestive Cancer Mortality. Carcinogenesis, 2012, 33:1055–8., DOI 10.1093/carcin/bgs112 Hajishengallis G, Lamont RJ. Breaking Bad: Manipulation of the Host Response byPorphyromonas Gingivalis. Eur J Immunol, 2014, 44:328–38., DOI 10.1002/eji.201344202 Lafuente Ibanez de Mendoza I, Maritxalar Mendia X, Garcia de la Fuente A. M., Quindos Andres G., Aguirre Urizar J. M.. Role of Porphyromonas Gingivalis in Oral Squamous Cell Carcinoma Development: A Systematic Review. J Periodontal Res, 2020, 55:13–22. Liu Y, Yuan X, Chen K, Zhou F, Yang H, Yang H. Clinical Significance and Prognostic Value of Porphyromonas Gingivalis Infection in Lung Cancer. Translational Oncol, 2021, 14:100972., DOI 10.1016/j.tranon.2020.100972 Kong J, Yuan X, Wang J, Liu Y, Sun W, Gu B, et al. Frequencies of Porphyromonas Gingivalis Detection in Oral-Digestive Tract Tumors. Pathol Oncol Res, 2021, 27., DOI 10.3389/pore.2021.628942 Wu Y-W, Li F. Bacterial Interaction with Host Autophagy. Virulence, 2019, 10:352–62., DOI 10.1080/21505594.2019.1602020 Miao G, Zhao H, Li Y, Ji M, Chen Y, Shi Y. ORF3a of the COVID-19 Virus SARS-CoV-2 Blocks HOPS Complex-Mediated Assembly of the SNARE Complex Required for Autolysosome Formation. Develop Cel, 2021, 56: 427–42. e425., DOI 10.1016/j.devcel.2020.12.010 Ding B, Zhang G, Yang X, Zhang S, Chen L, Yan Q. Phosphoprotein of Human Parainfluenza Virus Type 3 Blocks Autophagosome-Lysosome Fusion to Increase Virus Production. Cell Host & Microbe, 2014, 15:564–77., DOI 10.1016/j.chom.2014.04.004 Yamatake K, Maeda M, Kadowaki T, Takii R, Tsukuba T, Ueno T. Role for Gingipains in Porphyromonas Gingivalis Traffic to Phagolysosomes and Survival in Human Aortic Endothelial Cells. Infect Immun, 2007, 75: 2090–100., DOI 10.1128/iai.01013-06 Moreno SM, Contreras A. Functional Differences of Porphyromonas Gingivalis Fimbriae’s Determining Periodontal Disease Pathogenesis: A Literature Review. Colomb Med, Cali,, 2013, 44:48–56., DOI 10.25100/ cm.v44i1.800 El-Awady AR, Miles B, Scisci E, Kurago ZB, Palani CD, Arce RM. Porphyromonas Gingivalis Evasion of Autophagy and Intracellular Killing by Human Myeloid Dendritic Cells Involves DC-SIGN-TLR2 Crosstalk. Plos Pathog, 2015, 11:e1004647., DOI 10.1371/journal.ppat.1004647 Matsunaga K, Saitoh T, Tabata K, Omori H, Satoh T, Kurotori N. Two Beclin 1-binding Proteins, Atg14L and Rubicon, Reciprocally Regulate Autophagy at Different Stages. Nat Cel Biol, 2009, 11:385–96., DOI 10.1038/ncb1846 Liang C, Lee J-s., Inn K-S, Gack MU, Li Q, Roberts EA. Beclin1-binding UVRAG Targets the Class C Vps Complex to Coordinate Autophagosome Maturation and Endocytic Trafficking. Nat Cel Biol, 2008, 10:776–87., DOI 10.1038/ncb1740 Hill SM,Wrobel L, Rubinsztein DC. Post-translational Modifications of Beclin 1 Provide Multiple Strategies for Autophagy Regulation. Cell Death Differ, 2019, 26:617–29., DOI 10.1038/s41418-018-0254-9 Niu H, Xiong Q, Yamamoto A, Hayashi-Nishino M, Rikihisa Y. Autophagosomes Induced by a Bacterial Beclin 1 Binding Protein Facilitate Obligatory Intracellular Infection. Proc Natl Acad Sci, 2012, 109:20800–7., DOI 10.1073/pnas.1218674109 Valente G, Morani F, Nicotra G, Fusco N, Peracchio C, Titone R. Expression and Clinical Significance of the Autophagy Proteins BECLIN 1 and LC3 in Ovarian Cancer. Biomed Res Int, 2014, 2014:462658., DOI 10.1155/2014/ 462658 Zhang Y, Lin S, Zhang Y, Chang S. Effect of Beclin 1 Expression on the Biological Behavior and Chemotherapy Sensitivity of Cervical Cancer Cells. Oncol Lett, 2016, 11:4089–94., DOI 10.3892/ol.2016.4542 Weng J, Wang C, Wang Y, Tang H, Liang J, Liu X. Beclin1 Inhibits Proliferation, Migration and Invasion in Tongue Squamous Cell Carcinoma Cell Lines. Oral Oncol, 2014, 50:983–90., DOI 10.1016/j.oraloncology.2014.06.020 Du H, Che J, Shi M, Zhu L, Hang JB, Chen Z. Beclin 1 Expression Is Associated with the Occurrence and Development of Esophageal Squamous Cell Carcinoma. Oncol Lett, 2017, 14:6823–8., DOI 10.3892/ol.2017.7015 Yang M, Zhao H, Guo L, Zhang Q, Zhao L, Bai S. Autophagy-based Survival Prognosis in Human Colorectal Carcinoma. Oncotarget, 2015, 6:7084–103., DOI 10.18632/oncotarget.3054 ZhouW, Yue C, Deng J, Hu R, Xu J, Feng L. Autophagic Protein Beclin 1 Serves as an Independent Positive Prognostic Biomarker for Non-small Cell Lung Cancer. PLoS One, 2013, 8:e80338., DOI 10.1371/ journal.pone.0080338 Dikic I, Elazar Z. Mechanism and Medical Implications of Mammalian Autophagy. Nat Rev Mol Cel Biol, 2018, 19:349–64., DOI 10.1038/s41580- 018-0003-4 Miller K, McGrath ME, Hu Z, Ariannejad S, Weston S, Frieman M. Coronavirus Interactions with the Cellular Autophagy Machinery. Autophagy, 2020, 16:2131–9., DOI 10.1080/15548627.2020.1817280 Mimouna S, Bazin M, Mograbi B, Darfeuille-Michaud A, Brest P, Hofman P. HIF1A Regulates Xenophagic Degradation of Adherent and invasiveEscherichia Coli(AIEC). Autophagy, 2014, 10:2333–45., DOI 10.4161/ 15548627.2014.984275 Koepke L, Hirschenberger M, Hayn M, Kirchhoff F, Sparrer KM. Manipulation of Autophagy by SARS-CoV-2 Proteins. Autophagy, 2021, 17:2659–61., DOI 10.1080/15548627.2021.1953847 Gholizadeh P, Eslami H, Kafil HS. Carcinogenesis Mechanisms of Fusobacterium Nucleatum. Biomed Pharmacother, 2017, 89:918–25., DOI 10.1016/j.biopha.2017.02.102 Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D. Analysis of Fusobacterium Persistence and Antibiotic Response in Colorectal Cancer. Science, 2017, 358:1443–8., DOI 10.1126/science.aal5240 Haruki K, Kosumi K, Hamada T, Twombly TS, Vayrynen JP, Kim SA. Association of Autophagy Status with Amount of Fusobacterium Nucleatum in Colorectal Cancer. J Pathol, 2020, 250:397–408., DOI 10.1002/ path.5381 Chen Y, Chen Y, Zhang J, Cao P, Su W, Deng Y. Fusobacterium Nucleatum Promotes Metastasis in Colorectal Cancer by Activating Autophagy Signaling via the Upregulation of CARD3 Expression. Theranostics, 2020, 10:323–39., DOI 10.7150/thno.38870 Ma L, Liu H, Wang X, Jiang C, Yao S, Guo Y. CXXC5 Orchestrates Stat3/Erk/ Akt Signaling Networks to Modulate P. Gingivalis-Elicited Autophagy in Cementoblasts. Biochim Biophys Acta, Bba, - Mol Cel Res, 2021, 1868: 118923., DOI 10.1016/j.bbamcr.2020.118923 Hirasawa M, Kurita-Ochiai T. Porphyromonas Gingivalis Induces Apoptosis and Autophagy via ER Stress in Human Umbilical Vein Endothelial Cells. Mediators Inflamm, 2018, 2018:1967506., DOI 10.1155/2018/1967506 Oka S, Li X, Zhang F, Tewari N, Kim I-S, Chen C. Loss of Dec1 Prevents Autophagy in Inflamed Periodontal Ligament Fibroblast. Mol Biol Rep, 2021, 48:1423–31., DOI 10.1007/s11033-021-06162-x He S, Zhou Q, Luo B, Chen B, Li L, Yan F. Chloroquine and 3-Methyladenine Attenuates Periodontal Inflammation and Bone Loss in Experimental Periodontitis. Inflammation, 2020, 43:220–30., DOI 10.1007/s10753-019- 01111-0 Belanger M, Rodrigues PH, Dunn WA, Progulske-Fox A. Autophagy: a Highway for Porphyromonas Gingivalis in Endothelial Cells. Autophagy, 2006, 2:165–70., DOI 10.4161/auto.2828 Carvalho-Filho PC, Gomes-Filho IS, Meyer R, Olczak T, Xavier MT, Trindade SC. Role of Porphyromonas Gingivalis HmuY in Immunopathogenesis of Chronic Periodontitis. Mediators Inflamm, 2016, 2016:7465852., DOI 10.1155/ 2016/7465852 Lee K, Roberts JS, Choi CH, Atanasova KR, Yilmaz O. Porphyromonas Gingivalis Traffics into Endoplasmic Reticulum-Rich-Autophagosomes for Successful Survival in Human Gingival Epithelial Cells. Virulence, 2018, 9: 845–59., DOI 10.1080/21505594.2018.1454171 Zheng Y, Chen Z, Han Y, Han L, Zou X, Zhou B. Immune Suppressive Landscape in the Human Esophageal Squamous Cell Carcinoma Microenvironment. Nat Commun, 2020, 11:6268., DOI 10.1038/s41467-020- 20019-0 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609976 EP 1609987 DI 10.3389/pore.2021.1609976 PG 12 ER PT J AU Zheng, H Ning, Y Yang, Y Zhan, Y Wang, H Wen, Q Peng, J Fan, S AF Zheng, Hongmei Ning, Yue Yang, Yang Zhan, Yuting Wang, Haihua Wen, Qiuyuan Peng, Jinwu Fan, Songqing TI Aberrant Expression of β-Catenin Correlates with Infiltrating Immune Cells and Prognosis in NSCLC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; NSCLC; β-catenin; tumor microenvironment; tumor immunity ID biomarker; NSCLC; β-catenin; tumor microenvironment; tumor immunity AB Aims: β-catenin is a critical regulating factor of the Wnt pathway, which is closely linked to tumorigenesis, tumor growth, metastasis, and tumor immunity. Our study focused on exploring the relationship between β-catenin and clinicopathological features, prognosis, as well as infiltrating immune cells and immune scores, so as to illustrate its clinical significance in NSCLC. Materials and Methods: The β-catenin mRNA (CTNNB1) and protein expression data were downloaded from the UALCAN and the UCSC Xena website, respectively. All tumorimmune infiltrating cells’ data were downloaded from the TIMER platform and immune scores were downloaded from ESTIMATE website. The expression of β-catenin protein in our cohort was measured by immunohistochemistry. Results: β-catenin mRNA level was higher in lung adenocarcinoma (LUAD) compared to normal tissues (p < 0.001) and was related to overall survival (OS) (p < 0.001) and postprogression survival (PPS) (both p = 0.049) in LUAD. Aberrant β-catenin protein expression was higher in male and lung squamous cell carcinoma (LUSC) patients (both p = 0.001). Also, it was considered to be a prognosis factor independently (p = 0.034). In addition, β-catenin protein was negatively correlated with CD8+T cells (r = −0.128, p = 0.008), neutrophils (r = −0.198, p < 0.001), immune score (r = −0.109, p = 0.024), stromal score (r = −0.097, p = 0.045), and ESTIMATE score (r = −0.113, p = 0.020). Conclusions: Aberrant β-catenin protein expression was evidently higher in NSCLC and might serve as a biomarker for poor prognosis. Most importantly, β-catenin protein might play an important part in tumor immunity and the tumor microenvironment by inhibiting the infiltration of CD8+ T cells and neutrophils. C1 [Zheng, Hongmei] Central South University, Second Xiangya Hospital, Department of PathologyChangsha, China. [Ning, Yue] Central South University, Second Xiangya Hospital, Department of PathologyChangsha, China. [Yang, Yang] Central South University, Second Xiangya Hospital, Department of PathologyChangsha, China. [Zhan, Yuting] Central South University, Second Xiangya Hospital, Department of PathologyChangsha, China. [Wang, Haihua] Central South University, Second Xiangya Hospital, Department of PathologyChangsha, China. [Wen, Qiuyuan] Central South University, Second Xiangya Hospital, Department of PathologyChangsha, China. [Peng, Jinwu] Central South University, Xiangya Basic Medical College, Department of PathologyChangsha, China. [Fan, Songqing] Central South University, Second Xiangya Hospital, Department of PathologyChangsha, China. RP Fan, S (reprint author), Central South University, Second Xiangya Hospital, Department of Pathology, Changsha, China. EM songqingfan@csu.edu.cn CR Siegel RL, Miller KD, and Jemal A. Cancer Statistics, 2019. CA A Cancer J Clin, 2019, 69(1):7–34., DOI 10.3322/caac.21551 Zheng H, Zhan Y, Liu S, Lu J, Luo J, and Feng J. The Roles of Tumor- Derived Exosomes in Non-small Cell Lung Cancer and Their Clinical Implications. J Exp Clin Cancer Res, 2018, 37(1):226., DOI 10.1186/s13046- 018-0901-5 Reck M, Heigener DF, Mok T, Soria J-C, and Rabe KF. Management of Nonsmall- cell Lung Cancer: Recent Developments. Lancet, 2013, 382(9893): 709–19., DOI 10.1016/s0140-6736(13)61502-0 Miller KD, Nogueira L, Mariotto AB, Rowland JH, Yabroff KR, and Alfano CM. Cancer Treatment and Survivorship Statistics, 2019. CA A Cancer J Clin, 2019, 69(5):363–85., DOI 10.3322/caac.21565 Zheng H, Zhang Y, Zhan Y, Liu S, Lu J, and Feng J. Prognostic Analysis of Patients with Mutant and Wild-type EGFR Gene Lung Adenocarcinoma. Cancer Manag Res, 2019, 11:6139–50., DOI 10.2147/cmar.s200126 Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ, and Wu Y-L. Lung Cancer: Current Therapies and New Targeted Treatments. Lancet, 2017, 389(10066):299–311., DOI 10.1016/s0140-6736(16)30958-8 Nishijima TF, Shachar SS, Muss HB, and Tamura K. Patient-Reported Outcomes with PD-1/PD-L1 Inhibitors for Advanced Cancer: A Meta- Analysis. Oncologist, 2019, 24(7):e565–e573., DOI 10.1634/ theoncologist.2018-0449 Garon EB, Hellmann MD, Rizvi NA, Carcereny E, Leighl NB, and Ahn M-J. Five-Year Overall Survival for Patients With Advanced Non‒Small-Cell Lung Cancer Treated With Pembrolizumab: Results from the Phase I KEYNOTE- 001 Study. J Clin Oncol, 2019, 37(28):2518–27., DOI 10.1200/jco.19.00934 McCrea P, Turck C, and Gumbiner B. A Homolog of the Armadillo Protein in Drosophila, Plakoglobin, Associated with E-Cadherin. Science, 1991, 254(5036):1359–61., DOI 10.1126/science.1962194 Fagotto F. Looking beyond the Wnt Pathway for the Deep Nature of β-catenin. EMBO Rep, 2013, 14(5):422–33., DOI 10.1038/embor.2013.45 Valenta T, Hausmann G, and Basler K. The many Faces and Functions of β-catenin. EMBO J, 2012, 31(12):2714–36., DOI 10.1038/emboj.2012.150 Yamaguchi K, Nagatoishi S, Tsumoto K, and Furukawa Y. Discovery of Chemical Probes that Suppress Wnt/β-catenin Signaling through Highthroughput Screening. Cancer Sci, 2020, 111(3):783–94., DOI 10.1111/ cas.14297 Cui Y, Wu X, Lin C, Zhang X, Ye L, and Ren L. AKIP1 Promotes Early Recurrence of Hepatocellular Carcinoma through Activating the Wnt/ β-Catenin/CBP Signaling Pathway. Oncogene, 2019, 38(27):5516–29., DOI 10.1038/s41388-019-0807-5 Fodde R, and Brabletz T. Wnt/β-catenin Signaling in Cancer Stemness and Malignant Behavior. Curr Opin Cel Biol, 2007, 19(2):150–8., DOI 10.1016/ j.ceb.2007.02.007 Monga SP. β-Catenin Signaling and Roles in Liver Homeostasis, Injury, and Tumorigenesis. Gastroenterology, 2015, 148(7):1294–310., DOI 10.1053/ j.gastro.2015.02.056 Shang S, Hua F, and Hu Z-W. The Regulation of β-catenin Activity and Function in Cancer: Therapeutic Opportunities. Oncotarget, 2017, 8(20): 33972–89., DOI 10.18632/oncotarget.15687 Albini A, and Sporn MB. The Tumour Microenvironment as a Target for Chemoprevention. Nat Rev Cancer, 2007, 7(2):139–47., DOI 10.1038/nrc2067 Quail DF, and Joyce JA. Microenvironmental Regulation of Tumor Progression and Metastasis. Nat Med, 2013, 19(11):1423–37., DOI 10.1038/nm.3394 Patel S, Alam A, Pant R, and Chattopadhyay S. Wnt Signaling and its Significance Within the Tumor Microenvironment: Novel Therapeutic Insights. Front Immunol, 2019, 10:2872., DOI 10.3389/fimmu.2019.02872 Gyorffy B, Surowiak P, Budczies J, and Lanczky A. Online Survival Analysis Software to Assess the Prognostic Value of Biomarkers Using Transcriptomic Data in Non-small-cell Lung Cancer[J]. PLoS One, 2013, 8(12):e82241., DOI 10.1371/journal.pone.0082241 Detterbeck FC, Boffa DJ, Kim AW, and Tanoue LT. The Eighth Edition Lung Cancer Stage Classification. Chest, 2017, 151(1):193–203., DOI 10.1016/ j.chest.2016.10.010 Chen L, Xie G, Feng J, Wen Q, Zang H, and Lu J. Overexpression of FADD and Bcl-XS Proteins as Novel Prognostic Biomarkers for Surgically Resected Nonsmall Cell Lung Cancer. Cancer Biomark, 2021, 30(2):145–54., DOI 10.3233/ cbm-190018 Song Y, Yang QX, Zhang F, Meng F, Li H, and Dong Y. Suppression of Nasopharyngeal Carcinoma Cell by Targeting β-catenin Signaling Pathway. Cancer Epidemiol, 2012, 36(2):e116–e121., DOI 10.1016/ j.canep.2011.11.002 Wang H, Wu X, and Chen Y. Stromal-Immune Score-Based Gene Signature: A Prognosis Stratification Tool in Gastric Cancer. Front Oncol, 2019, 9:1212., DOI 10.3389/fonc.2019.01212 Kim H, Yoo SB, Sun P, Jin Y, Jheon S, and Lee CT. Alteration of the E-Cadherin/β-Catenin Complex Is an Independent Poor Prognostic Factor in Lung Adenocarcinoma. Korean J Pathol, 2013, 47(1):44–51., DOI 10.4132/ koreanjpathol.2013.47.1.44 Muto S, Ozaki Y, Yamaguchi H, Mine H, Takagi H, and Watanabe M. Tumor β-catenin Expression Is Associated with Immune Evasion in Non-small Cell Lung Cancer with High Tumor Mutation burden. Oncol Lett, 2021, 21(3):203., DOI 10.3892/ol.2021.12464 Xu X, Sun P-L, Li J-Z, Jheon S, Lee C-T, and Chung J-H. Aberrant Wnt1/ β-Catenin Expression Is an Independent Poor Prognostic Marker of Non- Small Cell Lung Cancer After Surgery. J Thorac Oncol, 2011, 6(4):716–24., DOI 10.1097/jto.0b013e31820c5189 Sun H, Liu M, Wu X, Yang C, Zhang Y, and Xu Z. Overexpression of N-Cadherin and β-catenin Correlates with Poor Prognosis in Patients with Nasopharyngeal Carcinoma. Oncol Lett, 2017, 13(3):1725–30., DOI 10.3892/ ol.2017.5645 Fang X, Gu P, Zhou C, Liang A, Ren S, and Liu F. β-Catenin Overexpression Is Associated with Gefitinib Resistance in Non-small Cell Lung Cancer Cells. Pulm Pharmacol Ther, 2014, 28(1):41–8., DOI 10.1016/j.pupt.2013.05.005 To SKY, Mak ASC, Eva Fung YM, Che C-M, Li S-S, and Deng W. β-Catenin Downregulates Dicer to Promote Ovarian Cancer Metastasis. Oncogene, 2017, 36(43):5927–38., DOI 10.1038/onc.2017.185 Hsu P-K, Li AF-Y, Wang Y-C, Hsieh C-C,Huang M-H, and Hsu W-H. Reduced Membranous β-catenin Protein Expression Is Associated with Metastasis and Poor Prognosis in Squamous Cell Carcinoma of the Esophagus. J Thorac Cardiovasc Surg, 2008, 135(5):1029–35., DOI 10.1016/j.jtcvs.2007.11.007 Wang W, Wen Q, Luo J, Chu S, Chen L, and Xu L. Suppression of β-catenin Nuclear Translocation by CGP57380 Decelerates Poor Progression And Potentiates Radiation-Induced Apoptosis in Nasopharyngeal Carcinoma. Theranostics, 2017, 7(7):2134–49., DOI 10.7150/thno.17665 Tsai MC, Huang CC, Wei YC, Liu TT, Lin MT, and Yi LN. Combined Chibby and β-Catenin Predicts Clinical Outcomes in Patients with Hepatocellular Carcinoma. Int J Mol Sci, 2020, 21(6), 2060., DOI 10.3390/ijms21062060 Yoshihara K, ShahmoradgoliM,Martinez E, Vegesna R, Kim H, and Torres- Garcia W. Inferring Tumour Purity and Stromal and Immune Cell Admixture from Expression Data. Nat Commun, 2013, 4:2612., DOI 10.1038/ncomms3612 Pai SG, Carneiro BA, Mota JM, Costa R, Leite CA, and Barroso-Sousa R. Wnt/ beta-catenin Pathway: Modulating Anticancer Immune Response. J Hematol Oncol, 2017, 10(1):101., DOI 10.1186/s13045-017-0471-6 Li X, Xiang Y, Li F, Yin C, Li B, and Ke X. WNT/β-Catenin Signaling Pathway Regulating T Cell-Inflammation in the Tumor Microenvironment. Front Immunol, 2019, 10:2293., DOI 10.3389/fimmu.2019.02293 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609981 EP 1609989 DI 10.3389/pore.2021.1609981 PG 9 ER PT J AU Jiang, X Zhang, W Li, L Xie, Sh AF Jiang, Xili Zhang, Wei Li, Lifeng Xie, Shucai TI Integrated Transcriptomic Analysis Revealed Hub Genes and Pathways Involved in Sorafenib Resistance in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE hepatocellular carcinoma; hub gene; sorafenib resistance; integrated bioinformatics analysis; transcriptomic analysis ID hepatocellular carcinoma; hub gene; sorafenib resistance; integrated bioinformatics analysis; transcriptomic analysis AB Hepatocellular carcinoma (HCC), a high mortality malignancy, has become a worldwide public health concern. Acquired resistance to the multikinase inhibitor sorafenib challenges its clinical efficacy and the survival benefits it provides to patients with advanced HCC. This study aimed to identify critical genes and pathways associated with sorafenib resistance in HCC using integrated bioinformatics analysis. Differentially expressed genes (DEGs) were identified using four HCC gene expression profiles (including 34 sorafenib-resistant and 29 sorafenib-sensitive samples) based on the robust rank aggregation method and R software. Gene ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool. A protein–protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes (STRING), and small molecules reversing sorafenib resistance were searched for using the connectivity map (CMAP) database. Pearson correlation and survival analyses of hub genes were performed using cBioPortal and Gene Expression Profiling and Interactive Analysis (GEPIA). Finally, the expression levels of hub genes in sorafenib-resistant HCC cells were verified using quantitative polymerase chain reaction (q-PCR). A total of 165 integrated DEGs (66 upregulated and 99 downregulated in sorafenib resistant samples compared sorafenib sensitive ones) primarily enriched in negative regulation of endopeptidase activity, extracellular exosome, and protease binding were identified. Some pathways were commonly shared between the integrated DEGs. Seven promising therapeutic agents and 13 hub genes were identified. These findings provide a strategy and theoretical basis for overcoming sorafenib resistance in HCC patients. C1 [Jiang, Xili] The Second People’s Hospital of Hunan Province/Brain Hospital of Hunan Province, Department of RadiologyChangsha, China. [Zhang, Wei] The Second People’s Hospital of Hunan Province/Brain Hospital of Hunan Province, Department of RadiologyChangsha, China. [Li, Lifeng] Changsha Central Hospital, Department of RadiologyChangsha, China. [Xie, Shucai] Central South University, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Department of Critical Care MedicineChangsha, China. RP Xie, Sh (reprint author), Central South University, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Department of Critical Care Medicine, Changsha, China. EM 282791444@qq.com CR Grandhi MS, Kim AK, Ronnekleiv-Kelly SM, Kamel IR, Ghasebeh MA, and Pawlik TM. Hepatocellular Carcinoma: From Diagnosis to Treatment. Surg Oncol, 2016, 25:74–85., DOI 10.1016/j.suronc.2016.03.002 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. Global Cancer Statistics 2018: Globocan Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 68:394–424., DOI 10.3322/caac.21492 El-Serag HB. Epidemiology of Viral Hepatitis and Hepatocellular Carcinoma. Gastroenterology, 2012, 142:1264–73., DOI 10.1053/j.gastro.2011.12.061 Chen Z, XieH,HuM,Huang T, Hu Y, Sang N, et al.Recent Progress in Treatment of Hepatocellular Carcinoma. Am J Cancer Res, 2020, 10:2993–3036. Couri T, and Pillai A. Goals and Targets for Personalized Therapy for HCC. Hepatol Int, 2019, 13:125–37., DOI 10.1007/s12072-018-9919-1 Zhu Y-J., Zheng B, Wang H-Y., and Chen L. New Knowledge of the Mechanisms of Sorafenib Resistance in Liver Cancer. Acta Pharmacol Sin, 2017, 38:614–22., DOI 10.1038/aps.2017.5 Schlachterman A, Craft Jr. WW, Hilgenfeldt E, Mitra A, and Cabrera R. Current and Future Treatments for Hepatocellular Carcinoma. World J Gastroenterol, 2015, 21:8478–91., DOI 10.3748/wjg.v21.i28.8478 Dufour JF, Bargellini I, De Maria N, De Simone P, Goulis I, and Marinho RT. Intermediate Hepatocellular Carcinoma: Current Treatments and Future Perspectives. Ann Oncol, 2013, 24(Suppl. 2):ii24–9., DOI 10.1093/annonc/mdt054 Cheng A-L, Kang Y-K, Lin D-Y, Park J-W, Kudo M, Qin S, et al.Sunitinib Versus Sorafenib in Advanced Hepatocellular Cancer: Results of a Randomized Phase III Trial. J. Clin. Oncol., 2013, 31:4067–75., DOI 10.1200/jco.2012.45.8372 Cervello M, Bachvarov D, Lampiasi N, Cusimano A, Azzolina A, McCubrey JA, et al.Molecular Mechanisms of Sorafenib Action in Liver Cancer Cells. Cell cycle, 2012, 11:2843–55., DOI 10.4161/cc.21193 Sun W, He B, Yang B, HuW, Cheng S, Xiao H, et al.Genome-Wide CRISPR Screen Reveals SGOL1 as a Druggable Target of Sorafenib-Treated Hepatocellular Carcinoma. Lab Invest, 2018, 98:734–44., DOI 10.1038/s41374-018-0027-6 Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc J-F, et al.Sorafenib in Advanced Hepatocellular Carcinoma. N Engl J Med, 2008, 359:378–90., DOI 10.1056/nejmoa0708857 Cheng A-L, Kang Y-K, Chen Z, Tsao C-J, Qin S, Kim JS, et al.Efficacy and Safety of Sorafenib in Patients in the Asia-Pacific Region with Advanced Hepatocellular Carcinoma: A Phase III Randomised, Double-Blind, Placebo- Controlled Trial. Lancet Oncol, 2009, 10:25–34., DOI 10.1016/s1470-2045(08, 70285-7 Tang W, Chen Z, ZhangW, Cheng Y, Zhang B,Wu F, et al.The Mechanisms of Sorafenib Resistance in Hepatocellular Carcinoma: Theoretical Basis and Therapeutic Aspects. Sig Transduct Target Ther, 2020, 5:87., DOI 10.1038/ s41392-020-0187-x Xie S, Jiang X, Zhang J, Xie S, Hua Y,Wang R, et al.Identification of Significant Gene and Pathways Involved in HBV-Related Hepatocellular Carcinoma by Bioinformatics Analysis. PeerJ, 2019, 7:e7408., DOI 10.7717/peerj.7408 Kolde R, Laur S, Adler P, and Vilo J. Robust Rank Aggregation for Gene List Integration and Meta-Analysis. Bioinformatics, 2012, 28:573–80., DOI 10.1093/bioinformatics/btr709 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al.Limma powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res, 2015, 43:e47., DOI 10.1093/nar/gkv007 Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al.STRING V10: Protein-Protein Interaction Networks, Integrated over the Tree of Life. Nucleic Acids Res, 2015, 43:D447–D452., DOI 10.1093/nar/gku1003 Bader GD, and Hogue CW. An Automated Method for Finding Molecular Complexes in Large Protein Interaction Networks[J]. BMC bioinformatics, 2003, 4:2., DOI 10.1186/1471-2105-4-2 Lamb J. The Connectivity Map: a New Tool for Biomedical Research. Nat Rev Cancer, 2007, 7:54–60., DOI 10.1038/nrc2044 Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, et al.The Connectivity Map: Using Gene-Expression Signatures to Connect Small Molecules, Genes, and Disease. Science, 2006, 313:1929–35., DOI 10.1126/ science.1132939 Tang Z, Li C, Kang B, Gao G, Li C, and Zhang Z. GEPIA: A Web Server for Cancer and normal Gene Expression Profiling and Interactive Analyses. Nucleic Acids Res, 2017, 45:W98–W102., DOI 10.1093/nar/gkx247 Xie S, Jiang X, Wang R, Xie S, Hua Y, Zhou S, et al.Low-intensity Pulsed Ultrasound Promotes the Proliferation of Human Bone Mesenchymal Stem Cells by Activating PI3K/AKt Signaling Pathways. J Cel Biochem, 2019, 120: 15823–33., DOI 10.1002/jcb.28853 Xie S-C, Zhang J-Q, Jiang X-L, Hua Y-Y, Xie S-W, Qin Y-A, et al.LncRNA CRNDE Facilitates Epigenetic Suppression of CELF2 and LATS2 to Promote Proliferation, Migration and Chemoresistance in Hepatocellular Carcinoma. Cell Death Dis, 2020, 11:676., DOI 10.1038/s41419-020-02853-8 Zhao Y, Zheng X, Zheng Y, Chen Y, Fei W, Wang F, et al.Extracellular Matrix: Emerging Roles and Potential Therapeutic Targets for Breast Cancer. Front Oncol, 2021, 11:650453., DOI 10.3389/fonc.2021.650453 Mashouri L, Yousefi H, Aref AR, Ahadi AM., Molaei F, and Alahari SK. Exosomes: Composition, Biogenesis, and Mechanisms in Cancer Metastasis and Drug Resistance. Mol Cancer, 2019, 18:75., DOI 10.1186/s12943-019- 0991-5 Kaps L, and Schuppan D. Targeting Cancer Associated Fibroblasts in Liver Fibrosis and Liver Cancer Using Nanocarriers. Cells, 2020, 9:2027., DOI 10.3390/cells9092027 Chen R, Xu X, Tao Y, Qian Z, and Yu Y. Exosomes in Hepatocellular Carcinoma: A New Horizon. Cell Commun Signal, 2019, 17:1., DOI 10.1186/ s12964-018-0315-1 Chen K-F, Chen H-L, Tai W-T, Feng W-C, Hsu C-H, Chen P-J, et al.Activation of Phosphatidylinositol 3-Kinase/Akt Signaling Pathway Mediates Acquired Resistance to Sorafenib in Hepatocellular Carcinoma Cells. J Pharmacol Exp Ther, 2011, 337:155–61., DOI 10.1124/jpet.110.175786 Tan W, Luo X, Li W, Zhong J, Cao J, Zhu S, et al.TNF-α Is a Potential Therapeutic Target to Overcome Sorafenib Resistance in Hepatocellular Carcinoma. EBioMedicine, 2019, 40:446–56., DOI 10.1016/ j.ebiom.2018.12.047 Feng J, Dai W, Mao Y, Wu L, Li J, Chen K, et al.Simvastatin Re-Sensitizes Hepatocellular Carcinoma Cells to Sorafenib by Inhibiting HIF-1α/ppar- Γ/pkm2-Mediated Glycolysis. J Exp Clin Cancer Res, 2020, 39:24., DOI 10.1186/s13046-020-1528-x Kim M-J, Choi Y-K, Park SY, Jang SY, Lee JY, Ham HJ, et al.PPARδ Reprograms Glutamine Metabolism in Sorafenib-Resistant HCC. Mol Cancer Res, 2017, 15:1230–42., DOI 10.1158/1541-7786.mcr-17-0061 Li W, Dong X, He C, Tan G, Li Z, Zhai B, et al.LncRNA SNHG1 Contributes to Sorafenib Resistance by Activating the Akt Pathway and Is Positively Regulated by miR-21 in Hepatocellular Carcinoma Cells. J Exp Clin Cancer Res, 2019, 38:183., DOI 10.1186/s13046-019-1177-0 Gao Y, Fan X, Li N, Du C, Yang B, QinW, et al.CCL22 Signaling Contributes to Sorafenib Resistance in Hepatitis B Virus-Associated Hepatocellular Carcinoma. Pharmacol Res, 2020, 157:104800., DOI 10.1016/ j.phrs.2020.104800 Zhang P-F, Li K-S, Shen Y-H., Gao P-T, Dong Z-R, Cai J-B, et al.Galectin-1 Induces Hepatocellular Carcinoma EMT and Sorafenib Resistance by Activating FAK/PI3K/AKT Signaling. Cel Death Dis, 2016, 7, e2201., DOI 10.1038/cddis.2015.324 Li S, Dai W, Mo W, Li J, Feng J, Wu L, et al.By Inhibiting PFKFB3, Aspirin Overcomes Sorafenib Resistance in Hepatocellular Carcinoma. Int J Cancer, 2017, 141:2571–84., DOI 10.1002/ijc.31022 Feng J, Li J,Wu L, Yu Q, Ji J,Wu J, et al.Emerging Roles and the Regulation of Aerobic Glycolysis in Hepatocellular Carcinoma. J Exp Clin Cancer Res, 2020, 39:126., DOI 10.1186/s13046-020-01629-4 Datta A, Kim H, McGee L, Johnson AE, Talwar S, Marugan J, et al.High- Throughput Screening Identified Selective Inhibitors of Exosome Biogenesis and Secretion: A Drug Repurposing Strategy for Advanced Cancer. Sci Rep, 2018, 8:8161., DOI 10.1038/s41598-018-26411-7 Nazari M, Ghorbani A, Hekmat-Doost A, Jeddi-Tehrani M, and Zand H. Inactivation of Nuclear Factor-Κb by Citrus Flavanone Hesperidin Contributes to Apoptosis and Chemo-Sensitizing Effect in Ramos Cells. Eur J Pharmacol, 2011, 650:526–33., DOI 10.1016/j.ejphar.2010.10.053 Febriansah R, Dyaningtyas DPP, SarmokoNurulita NA, Meiyanto E, and Nugroho AE. Hesperidin as a Preventive Resistance Agent in MCF-7 Breast Cancer Cells Line Resistance to Doxorubicin. Asian Pac J Trop Biomed, 2014, 4:228–33., DOI 10.1016/s2221-1691(14)60236-7 Samanta D, Gilkes DM, Chaturvedi P, Xiang L, and Semenza GL. Hypoxia- Inducible Factors Are Required for Chemotherapy Resistance of Breast Cancer Stem Cells. Proc Natl Acad Sci USA, 2014, 111:E5429–E5438., DOI 10.1073/pnas.1421438111 Liu G, Fan X, Tang M, Chen R, Wang H, Jia R, et al.Osteopontin Induces Autophagy to Promote Chemo-Resistance in Human Hepatocellular Carcinoma Cells. Cancer Lett, 2016, 383:171–82., DOI 10.1016/ j.canlet.2016.09.033 Cheng H, Wu Z, Wu C, Wang X, Liow SS, Li Z, et al.Overcoming STC2 Mediated Drug Resistance through Drug and Gene Co -delivery by PHBPDMAEMA Cationic Polyester in Liver Cancer Cells. Mater Sci Eng C, 2018, 83:210–7., DOI 10.1016/j.msec.2017.08.075 Liu J, Qiu WC, QiuW, Shen X, and Sun G. Bioinformatics Analysis Revealed Hub Genes and Pathways Involved in Sorafenib Resistance in Hepatocellular Carcinoma. Math Biosci Eng, 2019, 16:6319–34., DOI 10.3934/mbe.2019315 Seyhan AA, Varadarajan U, Choe S, Liu W, and Ryan TE. A Genome-wide RNAi Screen Identifies Novel Targets of Neratinib Resistance Leading to Identification of Potential Drug Resistant Genetic Markers. Mol Biosyst, 2012, 8:1553–70., DOI 10.1039/c2mb05512k Vaziri-Gohar A, Zheng Y, and Houston KD. IGF-1 Receptor Modulates FoxO1-Mediated Tamoxifen Response in Breast Cancer Cells. Mol Cancer Res, 2017, 15:489–97., DOI 10.1158/1541-7786.mcr-16-0176 Zheng Y, Sowers JY, and Houston KD. IGFBP-1 Expression Promotes Tamoxifen Resistance in Breast Cancer Cells via Erk Pathway Activation. Front Endocrinol, 2020, 11:233., DOI 10.3389/fendo.2020.00233 Berberich A, Kessler T, Thome CM, Pusch S, Hielscher T, Sahm F, et al.Targeting Resistance against the MDM2 Inhibitor RG7388 in Glioblastoma Cells by the MEK Inhibitor Trametinib. Clin Cancer Res, 2019, 25:253–65., DOI 10.1158/1078-0432.ccr-18-1580 Lee C, Safdie FM, Raffaghello L, Wei M, Madia F, Parrella E, et al.Reduced Levels of IGF-I Mediate Differential Protection of Normal and Cancer Cells in Response to Fasting and Improve Chemotherapeutic Index. Cancer Res, 2010, 70:1564–72., DOI 10.1158/0008-5472.can-09-3228 Zhu D, Zha X, Hu M, Tao A, Zhou H, Zhou X, et al.High Expression of TIMP-1 in Human Breast Cancer Tissues Is a Predictive of Resistance to Paclitaxel-Based Chemotherapy. Med Oncol, 2012, 29:3207–15., DOI 10.1007/ s12032-012-0239-3 Schrohl A-S, Meijer-van Gelder ME, Holten-Andersen MN, Christensen IJ, Look MP, Mouridsen HT, et al.Primary Tumor Levels of Tissue Inhibitor of Metalloproteinases-1 Are Predictive of Resistance to Chemotherapy in Patients with Metastatic Breast Cancer. Clin Cancer Res, 2006, 12:7054–8., DOI 10.1158/1078-0432.ccr-06-0950 Sorensen NM, Bystrom P, Christensen IJ, Berglund A, Jorgen Nielsen H, Brunner N, et al.Timp-1 Is Significantly Associated with Objective Response and Survival in Metastatic Colorectal Cancer Patients Receiving Combination of Irinotecan, 5-fluorouracil, and Folinic Acid. Clin Cancer Res, 2007, 13: 4117–22., DOI 10.1158/1078-0432.CCR-07-0186 Davidsen ML,Wurtz S, Romer MU, Sorensen NM, Johansen SK, Christensen IJ, et al.TIMP-1 Gene Deficiency Increases Tumour Cell Sensitivity to Chemotherapy-Induced Apoptosis. Br J Cancer, 2006, 95:1114–20., DOI 10.1038/sj.bjc.6603378 Wang T, Lv J-H, Zhang X-F, Li C-J, Han X, and Sun Y-J. Tissue Inhibitor of Metalloproteinase-1 Protects MCF-7 Breast Cancer Cells from Paclitaxel- Induced Apoptosis by Decreasing the Stability of Cyclin B1. Int J Cancer, 2010, 126:362–70., DOI 10.1002/ijc.24753 Tsagaraki I, Tsilibary EC, and Tzinia AK. TIMP-1 Interaction with αvβ3 Integrin Confers Resistance to Human Osteosarcoma Cell Line MG-63 against TNF-α-Induced Apoptosis. Cell Tissue Res, 2010, 342:87–96., DOI 10.1007/s00441-010-1025-1 Tan Y, Li X, Tian Z, Chen S, Zou J, Lian G, et al.TIMP1 Down-Regulation Enhances Gemcitabine Sensitivity and Reverses Chemoresistance in Pancreatic Cancer. Biochem Pharmacol, 2020, 189:114085., DOI 10.1016/ j.bcp.2020.114085 D’Costa Z, Jones K, Azad A, van Stiphout R, Lim SY, Gomes AL, et al.Gemcitabine-Induced TIMP1 Attenuates Therapy Response and Promotes Tumor Growth and Liver Metastasis in Pancreatic Cancer. Cancer Res, 2017, 77:5952–62., DOI 10.1158/0008-5472.can-16-2833 Bjerre C, Vinther L, Belling KC, Wurtz SO, Yadav R, Lademann U, et al.TIMP1 Overexpression Mediates Resistance of MCF-7 Human Breast Cancer Cells to Fulvestrant and Down-Regulates Progesterone Receptor Expression. Tumor Biol, 2013, 34:3839–51., DOI 10.1007/s13277-013-0969-7 Sonego M, Poletto E, Pivetta E, Nicoloso MS, Pellicani R, Vinciguerra GLR, et al.TIMP-1 Is Overexpressed and Secreted by Platinum Resistant Epithelial Ovarian Cancer Cells. Cells, 2019, 9:6., DOI 10.3390/cells9010006 Chen J, Lan T, Hou J, Zhang J, An Y, Tie L, et al.Atorvastatin Sensitizes Human Non-small Cell Lung Carcinomas to Carboplatin via Suppression of AKT Activation and Upregulation of TIMP-1. Int J Biochem Cel Biol, 2012, 44:759–69., DOI 10.1016/j.biocel.2012.01.015 Pennanen PT, Sarvilinna NS, and Ylikomi TJ. Gene Expression Changes during the Development of Estrogen-Independent and Antiestrogen- Resistant Growth in Breast Cancer Cell Culture Models. Anticancer Drugs, 2009, 20:51–8., DOI 10.1097/cad.0b013e32831845e1 Bao Q, Niess H, Djafarzadeh R, Zhao Y, Schwarz B, Angele MK, et al.Recombinant TIMP-1-GPI Inhibits Growth of Fibrosarcoma and Enhances Tumor Sensitivity to Doxorubicin. Targ Oncol, 2014, 9:251–61., DOI 10.1007/s11523-013-0294-5 Zhang L, Huang Y, Zhuo W, Zhu Y, Zhu B, and Chen Z. Identification and Characterization of Biomarkers and Their Functions for Lapatinib-Resistant Breast Cancer. Med Oncol, 2017, 34:89., DOI 10.1007/s12032-017-0953-y Januchowski R, Sterzynska K, Zawierucha P, Rucinski M, Swierczewska M, Partyka M, et al.Microarray-based Detection and Expression Analysis of New Genes Associated with Drug Resistance in Ovarian Cancer Cell Lines. Oncotarget, 2017, 8:49944–58., DOI 10.18632/oncotarget.18278 Zhou M, Ye Z, Gu Y, Tian B, Wu B, and Li J. Genomic Analysis of Drug Resistant Pancreatic Cancer Cell Line by Combining Long Non-Coding RNA and mRNA Expression Profling. Int J Clin Exp Pathol, 2015, 8: 38–52. Pang X, Xie R, Zhang Z, Liu Q, Wu S, and Cui Y. Identification of SPP1 as an Extracellular Matrix Signature for Metastatic Castration-Resistant Prostate Cancer. Front Oncol, 2019, 9:924., DOI 10.3389/fonc.2019.00924 Xu X, Jiang X, Chen L, Zhao Y, Huang Z, Zhou H, et al.Retracted: MiR-181a Promotes Apoptosis and Reduces Cisplatin Resistance by Inhibiting Osteopontin in Cervical Cancer Cells. Cancer Biother Radiopharm, 2019, 34:559–65., DOI 10.1089/cbr.2019.2858 Chen X, Xiong D, Ye L, Yang H, Mei S,Wu J, et al.SPP1 Inhibition Improves the Cisplatin Chemo-Sensitivity of Cervical Cancer Cell Lines. Cancer Chemother Pharmacol, 2019, 83:603–13., DOI 10.1007/s00280-018-3759-5 Liu K, Hu H, Jiang H, Liu C, Zhang H, Gong S, et al.Upregulation of Secreted Phosphoprotein 1 Affects Malignant Progression, Prognosis, and Resistance to Cetuximab via the KRAS/MEK Pathway in Head and Neck Cancer. Mol carcinogenesis, 2020, 59:1147–58., DOI 10.1002/mc.23245 Wang X, Zhang F, Yang X, XueM, Li X, Gao Y, et al.Secreted Phosphoprotein 1, SPP1, Contributes to Second-Generation EGFR Tyrosine Kinase Inhibitor Resistance in Non-Small Cell Lung Cancer. Oncol Res, 2019, 27:871–7., DOI 10.3727/096504018x15426271404407 Mirzaei A, Mohammadi S, Ghaffari SH, Nikbakht M, Bashash D, Alimoghaddam K, et al.Osteopontin B and C Isoforms: Molecular Candidates Associated with Leukemic Stem Cell Chemoresistance in Acute Myeloid Leukemia. Asian Pac J Cancer Prev, 2017, 18:1707–15., DOI 10.22034/APJCP.2017.18.6.1707 GraessmannM, Berg B, Fuchs B, Klein A, and Graessmann A. Chemotherapy Resistance of Mouse WAP-SVT/t Breast Cancer Cells Is Mediated by Osteopontin, Inhibiting Apoptosis Downstream of Caspase-3. Oncogene, 2007, 26:2840–50., DOI 10.1038/sj.onc.1210096 Insua-Rodriguez J, Pein M, Hongu T, Meier J, Descot A, Lowy CM, et al.Stress Signaling in Breast Cancer Cells Induces Matrix Components that Promote Chemoresistant Metastasis. EMBO Mol Med, 2018, 10., DOI 10.15252/ emmm.201809003 Hsieh I-S, Huang W-H, Liou H-C, Chuang W-J, Yang R-S, and Fu W-M. Upregulation of Drug Transporter Expression by Osteopontin in Prostate Cancer Cells. Mol Pharmacol, 2013, 83:968–77., DOI 10.1124/ mol.112.082339 Gu M, and Zheng X. Osteopontin and Vasculogenic Mimicry Formation Are Associated with Response to Neoadjuvant Chemotherapy in Advanced Breast Cancer. OncoTargets Ther, 2017, 10:4121–7., DOI 10.2147/ott.s129414 Zheng X, Naiditch J, Czurylo M, Jie C, Lautz T, Clark S, et al.Differential Effect of Long-Term Drug Selection with Doxorubicin and Vorinostat on Neuroblastoma Cells with Cancer Stem Cell Characteristics. Cel Death Dis, 2013, 4–e740., DOI 10.1038/cddis.2013.264 Xie X-Q, Zhao Q-H, Wang H, and Gu K-S. Dysregulation of mRNA Profile in Cisplatin-Resistant Gastric Cancer Cell Line SGC7901. WJG, 2017, 23: 1189–202., DOI 10.3748/wjg.v23.i7.1189 von der Heyde S, Wagner S, Czerny A, Nietert M, Ludewig F, Salinas-Riester G, et al.mRNA Profiling Reveals Determinants of Trastuzumab Efficiency in HER2-Positive Breast Cancer. PloS one, 2015, 10:e0117818., DOI 10.1371/ journal.pone.0117818 Yamaoka T, Ohmori T, Ohba M, Arata S, Murata Y, Kusumoto S, et al.Distinct Afatinib Resistance Mechanisms Identified in Lung Adenocarcinoma Harboring an EGFR Mutation. Mol Cancer Res, 2017, 15:915–28., DOI 10.1158/1541-7786.mcr-16-0482 Macleod K, Mullen P, Sewell J, Rabiasz G, Lawrie S, Miller E, et al.Altered ErbB Receptor Signaling and Gene Expression in Cisplatin-Resistant Ovarian Cancer. Cancer Res, 2005, 65:6789–800., DOI 10.1158/0008-5472.can-04-2684 Pasini A, Paganelli G, Tesei A, Zoli W, Giordano E, and Calistri D. Specific Biomarkers Are Associated with Docetaxeland Gemcitabine-Resistant NSCLC Cell Lines. Translational Oncol, 2012, 5:461–8., DOI 10.1593/ tlo.12256 Rodvold JJ, Xian S, Nussbacher J, Tsui B, Cameron Waller T, Searles SC, et al.IRE1α and IGF Signaling Predict Resistance to an Endoplasmic Reticulum Stress-Inducing Drug in Glioblastoma Cells. Sci Rep, 2020, 10: 8348., DOI 10.1038/s41598-020-65320-6 Hensley PJ, Cao Z, Pu H, Dicken H, He D, Zhou Z, et al.Predictive and Targeting Value of IGFBP-3 in Therapeutically Resistant Prostate Cancer. Am J Clin Exp Urol, 2019, 7:188–202. Guix M, Faber AC, Wang SE, Olivares MG, Song Y, Qu S, et al.Acquired Resistance to EGFR Tyrosine Kinase Inhibitors in Cancer Cells Is Mediated by Loss of IGF-Binding Proteins. J Clin Invest, 2008, 118:2609–19., DOI 10.1172/JCI34588 Li C, Harada A, and Oh Y. IGFBP-3 Sensitizes Antiestrogen-Resistant Breast Cancer Cells through Interaction with GRP78. Cancer Lett, 2012, 325:200–6., DOI 10.1016/j.canlet.2012.07.004 Qiu N, He Y-F., Zhang S-M., Zhan Y-T., Han G-D., Jiang M, et al.Cullin7 Enhances Resistance to Trastuzumab Therapy in Her2 Positive Breast Cancer via Degrading IRS-1 and Downregulating IGFBP-3 to Activate the PI3K/AKT Pathway. Cancer Lett, 2019, 464:25–36., DOI 10.1016/j.canlet.2019.08.008 Day TF, Kallakury BVS, Ross JS, Voronel O, Vaidya S, Sheehan CE, et al.Dual Targeting of EGFR and IGF1R in the TNFAIP8 Knockdown Non-Small Cell Lung Cancer Cells. Mol Cancer Res, 2019, 17:1207–19., DOI 10.1158/1541- 7786.mcr-18-0731 Wang EA, ChenW-Y, and Wong C-H. Multiple Growth Factor Targeting by Engineered Insulin-like Growth Factor Binding Protein-3 Augments EGF Receptor Tyrosine Kinase Inhibitor Efficacy. Sci Rep, 2020, 10:2735., DOI 10.1038/s41598-020-59466-6 Liu YN, Tsai MF, Wu SG, Chang TH, Tsai TH, Gow CH, et al.Acquired Resistance to EGFR Tyrosine Kinase Inhibitors Is Mediated by the Reactivation of STC2/JUN/AXL Signaling in Lung Cancer. Int J Cancer, 2019, 145:1609–24., DOI 10.1002/ijc.32487 Wang Y, Gao Y, ChengH, Yang G, and TanW. Stanniocalcin 2 Promotes Cell Proliferation and Cisplatin Resistance in Cervical Cancer. Biochem biophysical Res Commun, 2015, 466:362–8., DOI 10.1016/j.bbrc.2015.09.029 Yuan Q, Zhan L, Zhang L-L, Wang Q, Liu J, Jiang Z-Y, et al.Stanniocalcin 2 Induces Oxaliplatin Resistance in Colorectal Cancer Cells by Upregulating P-Glycoprotein. Can J Physiol Pharmacol, 2016, 94:929–35., DOI 10.1139/ cjpp-2015-0530 Miyazaki S, Kikuchi H, Iino I, Uehara T, Setoguchi T, Fujita T, et al.Anti- VEGF Antibody Therapy Induces Tumor Hypoxia and Stanniocalcin 2 Expression and Potentiates Growth of Human colon Cancer Xenografts. Int J Cancer, 2014, 135:295–307., DOI 10.1002/ijc.28686 Jansen MPHM, Sas L, Sieuwerts AM, Van Cauwenberghe C, Ramirez-ArdilaD, Look M, et al.Decreased Expression of ABAT and STC2Hallmarks ER-Positive Inflammatory Breast Cancer and Endocrine Therapy Resistance in Advanced Disease. Mol Oncol, 2015, 9:1218–33., DOI 10.1016/j.molonc.2015.02.006 Wu W, Wang Q, Yin F, Yang Z, Zhang W, Gabra H, et al.Identification of Proteomic and Metabolic Signatures Associated with Chemoresistance of Human Epithelial Ovarian Cancer. Int J Oncol, 2016, 49:1651–65., DOI 10.3892/ijo.2016.3652 Mora-Lagos B, Cartas-Espinel I, Riquelme I, Parker AC, Piccolo SR, Viscarra T, et al.Functional and Transcriptomic Characterization of Cisplatin- Resistant AGS and MKN-28 Gastric Cancer Cell Lines. PloS one, 2020, 15:e0228331., DOI 10.1371/journal.pone.0228331 Hultsch S, Kankainen M, Paavolainen L, Kovanen R-M, Ikonen E, Kangaspeska S, et al.Association ofTamoxifen Resistance and Lipid Reprogramming in Breast Cancer. BMC cancer, 2018, 18:850., DOI 10.1186/s12885-018-4757-z Cruz IN, Coley HM, Kramer HB, Madhuri TK, Safuwan NAM, Angelino AR, et al.Proteomics Analysis of Ovarian Cancer Cell Lines and Tissues Reveals Drug Resistance-Associated Proteins. Cancer genomics & proteomics, 2017, 14:35–52., DOI 10.21873/cgp.20017 Barik SK,MohantyKK,MohantyAK, Rawat P,Gopal G, BishtD, et al.Identification andDifferential Expression of Serotransferrin and Apolipoprotein A-I in the Plasma of HIV-1 Patients Treated with First-Line Antiretroviral Therapy. BMC Infect Dis, 2020, 20:898., DOI 10.1186/s12879-020-05610-6 Li C, Singh B, Graves-Deal R, Ma H, Starchenko A, Fry WH, et al.Three- Dimensional Culture System Identifies a New Mode of Cetuximab Resistance and Disease-Relevant Genes in Colorectal Cancer. Proc Natl Acad Sci USA, 2017, 114:E2852–E2861., DOI 10.1073/pnas.1618297114 Fujii K, KarpovaMB, Asagoe K, Georgiev O, Dummer R, and Urosevic-Maiwald M. Versican Upregulation in Sezary Cells Alters Growth, Motility and Resistance to Chemotherapy. Leukemia, 2015, 29:2024–32., DOI 10.1038/leu.2015.103 Hyung SW, Lee MY, Yu JH, Shin B, Jung HJ, and Park JMA Serum Protein Profile Predictive of the Resistance to Neoadjuvant Chemotherapy in Advanced Breast Cancers. Mol Cel Proteomics, 2011, 10:M111–011023., DOI 10.1074/mcp.M111.011023 Lin H, Zhang R, Wu W, and Lei L. Comprehensive Network Analysis of the Molecular Mechanisms Associated with Sorafenib Resistance in Hepatocellular Carcinoma. Cancer Genet, 2020, 245:27–34., DOI 10.1016/ j.cancergen.2020.04.076 Huang D, Yuan W, Li H, Li S, Chen Z, and Yang H. Identification of Key Pathways and Biomarkers in Sorafenib-Resistant Hepatocellular Carcinoma Using Bioinformatics Analysis. Exp Ther Med, 2018, 16:1850–8., DOI 10.3892/ etm.2018.6427 van Malenstein H, Dekervel J, Verslype C, Van Cutsem E, Windmolders P, Nevens F, et al. Long-Term Exposure to Sorafenib of Liver Cancer Cells Induces Resistance with Epithelial-to-Mesenchymal Transition, Increased Invasion and Risk of Rebound Growth[J]. Cancer Lett., 2013, 329:74–83. Dekervel J, Popovic D, van Malenstein H, Windmolders P, Heylen L, Libbrecht L, et al. A Global Risk Score, GRS, to Simultaneously Predict Early and Late Tumor Recurrence Risk after Resection of Hepatocellular Carcinoma[j]. Transl Oncol, 2016, 9:139–146. Tovar V, Cornella H, Moeini A, Vidal S, Hoshida Y, Sia D, et al. Tumour Initiating Cells and IGF/FGF Signalling Contribute to Sorafenib Resistance in Hepatocellular Carcinoma[j]. Gut, 2017, 66:530–540. Wu M, Shen X, Tang Y, Zhou C, Li H, and Luo X. Identification and Validation of Potential Key Long Noncoding RNAs in Sorafenib-Resistant Hepatocellular Carcinoma Cells[j]. PeerJ, 2020, 8:e8624. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609985 EP 1609999 DI 10.3389/pore.2021.1609985 PG 15 ER PT J AU Remenyi, Gy Bereczky, Zs Gindele, R Ujfalusi, A Illes, Udvardy, M AF Remenyi, Gyula Bereczky, Zsuzsanna Gindele, Reka Ujfalusi, Aniko Illes, Arpad Udvardy, Miklos TI rs779805 Von Hippel-Lindau Gene Polymorphism Induced/Related Polycythemia Entity, Clinical Features, Cancer Association, and Familiar Characteristics SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE single nucleotide polymorphism; secondary erythrocytosis; von Hippel-Lindau gene; rs779805 polymorphism; phlebotomy ID single nucleotide polymorphism; secondary erythrocytosis; von Hippel-Lindau gene; rs779805 polymorphism; phlebotomy AB Increased red blood cell count may result from primary erythrocytosis (polycythemia vera), but it is often due to secondary causes with increased erythropoietin levels. Secondary erythrocytosis may also be congenital due to different gene mutations of hemoglobin, hemoglobin stabilization proteins, EPO receptors, or oxygen sensing pathways. Von Hippel- Lindau gene mutation causes altered tissue oxygen sensation in VHL disease, usually with normal hemoglobin. Germline VHL mutations associate with classical VHL disease and represent genetic susceptibility for pheochromocytoma. VHL polymorphisms are mostly considered an innocent phenomenon. Still, some data indicate that these polymorphisms are not always harmless and can occur with prostate, renal, and colon cancer or even with isolated erythrocytosis. Seventy-eight patients referred to our department with elevated hemoglobin were screened for VHL mutations. There were no classical somatic VHL mutations. However, we found heterozygous (GA) or homozygous (AA) rs779805 VHL c.-195G>A polymorphism accompanied by erythrocytosis. These patients are Jak-2 negative, with normal or elevated EPO levels, sometimes with family accumulations and often phlebotomy needs, and in some cases with malignancies in the family. No other cause of erythrocytosis was found. We use phlebotomy regularly, and for those with cardiovascular risk factors, we recommend aspirin. C1 [Remenyi, Gyula] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Bereczky, Zsuzsanna] University of Debrecen, Faculty of Medicine, Department of Laboratory MedicineDebrecen, Hungary. [Gindele, Reka] University of Debrecen, Faculty of Medicine, Department of Laboratory MedicineDebrecen, Hungary. [Ujfalusi, Aniko] University of Debrecen, Faculty of Medicine, Department of Laboratory MedicineDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Udvardy, Miklos] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. RP Remenyi, Gy (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM gremenyi@med.unideb.hu CR McMullin MF. The Classification and Diagnosis of Erythrocytosis. Int J Lab Hematol, 2008, 30:447., DOI 10.1111/j.1751-553x.2008.01102.x Patnaik MM, Tefferi A. The Complete Evaluation of Erythrocytosis: Congenital and Acquired. Leukemia, 2009, 834–44., DOI 10.1038/leu.2009.54 Sokol L, Prchal JF, Prchal J. Primary Familial and Congenital Polycythaemia. The Lancet, 1993, 342:115–6., DOI 10.1016/0140-6736(93)91317-f Lonser RR, Glenn GM, Walther M, Chew EY, Libutti SK, Linehan WM, et al. von Hippel-Lindau disease. Lancet 361, 2003). p. 2059., DOI 10.1016/S0140- 6736(03)13643-4 Neumann HPH, Wiestler OD. Clustering of features of von Hippel-Lindau syndrome: evidence for a complex genetic locus. Lancet, 1991). 337:1052., DOI 10.1016/0140-6736(91)91705-y Lorenzo FR, Yang C, Lanikova L, Butros L, Zhuang Z, Prchal JT. Novel Compound VHL Heterozygosity, VHLT124A/L188V, Associated with Congenital Polycythaemia. Br J Haematol, 2013, 162(6):851–3., DOI 10.1111/bjh.12431 Sidhu A, Bhambhani K, Callaghan MU. Novel mutations in the von Hippel- Lindau gene associated with congenital polycythemia. Pediatr Blood Cancer, 2015, 62:1113–4., DOI 10.1002/pbc.25407 Sarangi S, Lanikova L, Kapralova K, Acharya S, Swierczek S, Lipton JM, et al. The Homozygous VHL D126N Missense Mutation Is Associated with Dramatically Elevated Erythropoietin Levels, Consequent Polycythemia, and Early Onset Severe Pulmonary Hypertension. Pediatr Blood Cancer, 2014, 61(11):2104–6., DOI 10.1002/pbc.25056 Lanikova L, Lorenzo F, Yang C, Vankayalapati H, Drachtman R, Divoky V, et al. Novel Homozygous VHL Mutation in Exon 2 Is Associated with Congenital Polycythemia but Not with Cancer. Blood, 2013). 121:3918., DOI 10.1182/blood-2012-11-469296 Tefferi A, Vardiman JW. Classification and Diagnosis of Myeloproliferative Neoplasms: the 2008 World Health Organization Criteria and point-of-care Diagnostic Algorithms. Leukemia, 2008). 22:14., DOI 10.1038/sj.leu.2404955 Wang W-C, Tsou M-H, Chen H-J, Hsu W-F, Lai Y-C. Two single nucleotide polymorphisms in the von Hippel-Lindau tumor suppressor gene in Taiwanese with renal cell carcinoma. BMC Res Notes, 2014, 7:638., DOI 10.1186/1756-0500-7-638 Demirel HS, Tasdemir P, Cetinkaya S, Cinar I, Kucukkartallar T, Dursun G. Colorectal Cancer Risk in Relation to Hypoxia Inducible Factor-1α, Hif-1 α, and Von Hippel-Lindau, Vhl, Gene Polymorphisms. Int J Hematol Oncol 29(3):13–20., DOI 10.4999/uhod.171674 Chen J, Wu Y, Shao P, Cao Q, Qin C, Li P, et al. Association between VHL Single Nucleotide Polymorphism, Rs779805, and the Susceptibility to Prostate Cancer in Chinese. DNA Cel Biol, 2012, 31:790., DOI 10.1089/dna.2011.1408 Mf M. Diagnostic Workflow for Hereditary Erythrocytosis and Thrombocytosis. Hematol Am Soc Hematol Educ Progr, 2019, 2019:391., DOI 10.1182/HEMATOLOGY.2019000047 Ang SO, Chen H, Gordeuk VR, Sergueeva AI, Polyakova LA, Miasnikova GY, et al. Endemic Polycythemia in Russia: Mutation in the VHL Gene. Blood Cell Mol Dis, 2002, 28(1):57–62., DOI 10.1006/bcmd.2002.0488 Tomasic NL, Piterkova L, Huff C, Bilic E, Yoon D, Miasnikova GY, et al. The Phenotype of Polycythemia Due to Croatian Homozygous VHL, 571C>G: H191D, Mutation Is Different from that of Chuvash Polycythemia. Haematologica, 2013, 98:560., DOI 10.3324/haematol.2012.070508 Liu E, Percy MJ, Amos CI, Guan Y, Shete S, Stockton DW, et al. The Worldwide Distribution of the VHL 598C>T Mutation Indicates a Single Founding Event. Blood 103, 2004). 1937., DOI 10.1182/blood-2003-07-2550 Couve S, Ladroue C, Laine E, Mahtouk K, Guegan J, Gad S, et al. Genetic evidence of a precisely tuned dysregulation in the hypoxia signaling pathway during oncogenesis. Cancer Res, 74, 2014). 6554., DOI 10.1158/0008-5472.CAN- 14-1161 van de Pol JAA, van den Brandt PA, van Engeland M, Godschalk RWL, van Schooten FJ, Hogervorst JGF, et al. Germline Polymorphisms in the Von Hippel-Lindau and Hypoxia-Inducible Factor 1-alpha Genes, Gene- Environment and Gene-Gene Interactions and Renal Cell Cancer. Sci Rep, 2020, 10., DOI 10.1038/s41598-019-56980-0 Lv C, Bai Z, Liu Z, Luo P, Zhang J. Renal Cell Carcinoma Risk Is Associated with the Interactions of APOE, VHL and MTHFR Gene Polymorphisms. Int J Clin Exp Pathol, 2015, 8:5781. Qin C, Cao Q, Li P, Ju X, Wang M, Chen J, et al. Functional Promoter -31G>C Variant in Survivin Gene Is Associated with Risk and Progression of Renal Cell Cancer in a Chinese Population. PLoS One, 2012, 7., DOI 10.1371/journal.pone.0028829 Moore LE, Nickerson ML, Brennan P, Toro JR, Jaeger E, Rinsky J, et al. Von Hippel-Lindau, VHL, Inactivation in Sporadic clear Cell Renal Cancer: Associations with Germline VHL Polymorphisms and Etiologic Risk Factors. PLoS Gene, 2011, 7:1002312., DOI 10.1371/ journal.pgen.1002312 McMullin MFF, Mead AJ, Ali S, Cargo C, Chen F, Ewing J, et al. A Guideline for the Management of Specific Situations in Polycythaemia Vera and Secondary Erythrocytosis. Br J Haematol, 2019, 184(2):161–75., DOI 10.1111/bjh.15647 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609987 EP 1609992 DI 10.3389/pore.2021.1609987 PG 6 ER PT J AU Youssef, H Radi, D Abd El-Azeem, M AF Youssef, M. K. Heba Radi, A. Dina Abd El-Azeem, A. Marwa TI Expression of TSP50, SERCA2 and IL-8 in Colorectal Adenoma and Carcinoma: Correlation to Clinicopathological Factors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE clinicopathological factors; colorectal adenoma; colorectal carcinoma; immunohistochemistry; IL-8; SERCA2 TSP50 ID clinicopathological factors; colorectal adenoma; colorectal carcinoma; immunohistochemistry; IL-8; SERCA2 TSP50 AB Background: Colorectal cancer (CRC) is the third most common type of cancer, it is considered a genetically heterogeneous disease with different molecular pathways being involved in its initiation and progression. Testes-specific protease 50 (TSP50) gene is a member of cancer/testis antigens that encodes for threonine protease enzyme. Overexpression of TSP50 was found to enhance the progression and invasion of breast cancer and other malignant tumors. SERCA2 is widely expressed in several body tissues; its aberrant expression has been involved in many cancers. IL-8 is an inflammatory cytokine. Alongside its role in inflammation, its expression was reported to induce the migration of tumor cells. Aim: Study the expression of TSP50, SERCA2 and IL-8 in colorectal adenoma (CRA), CRC and normal colonic tissues to compare the expression of these biomarkers in relation to clinicopathological parameters and prognostic factors. Results: TSP50, SERCA2 and IL-8 expression varied between normal colonic tissues, CRA and CRC. Significant statistical association was detected between the three biomarkers’ overexpression and degree of dysplasia in CRA. Also, significant statistical relation was found between the three biomarkers’ overexpression and presence of lympho-vascular invasion, advanced TNM staging and high intra-tumoral inflammatory infiltrate. Multivariable analysis showed that the overexpression of the three biomarkers is significantly associated with worse prognosis. Conclusion: The expression of TSP50, SERCA2 and IL-8 was different between the normal tissue and neoplastic colorectal tissue on one hand and between CRA and CRC on the other. Increased expression of these biomarkers in neoplastic epithelial cells of colorectal carcinoma is associated with adverse prognostic factors and could be considered as independent prognostic factors. C1 [Youssef, M. K. Heba] Tanta University, Faculty of Medicine, Department of PathologyTanta, Egypt. [Radi, A. Dina] Tanta University, Faculty of Medicine, Department of PathologyTanta, Egypt. [Abd El-Azeem, A. Marwa] Tanta University, Faculty of Medicine, Department of PathologyTanta, Egypt. RP Abd El-Azeem, M (reprint author), Tanta University, Faculty of Medicine, Department of Pathology, Tanta, Egypt. CR Ferlay J, Ervik M, Lam F, Colombet M, Mery L, Pineros M, et al. Global Cancer Observatory: Cancer Today. Lyon, France: International Agency for Research on Cancer, 2020). Available from https://gco.iarc.fr/today/home, Accessed December 2020). Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjmataram I, Jemal A, et al. Erratum: Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 70:313–41., DOI 10.3322/caac.21609 Jones RP, Jackson R, Dunne DFJ, Malik HZ, Fenwick SW, Poston GJ, et al. Systematic Review and Meta-Analysis of Follow-Up after Hepatectomy for Colorectal Liver Metastases2. Br J Surg, 2012, 99:477–86., DOI 10.1002/bjs.8667 Guinney J, Dienstmann R, Wang X, de Reynies A, Schlicker A, Soneson C, et al. The Consensus Molecular Subtypes of Colorectal Cancer. Nat Med, 2015, 21: 1350–6., DOI 10.1038/nm.3967 Shan J, Yuan L, Xiao Q, Chiorazzi N, Budman D, Teichberg S, et al. TSP50, a Possible Protease in Human Testes, Is Activated in Breast Cancer Epithelial Cells. Cancer Res, 2002, 62:290–4., DOI 10.1371/journal.pone.0035030 Cao Q-H, Liu F, Li C-Z, Liu N, Shu M, Lin Y, et al. Testes-specific Protease 50, TSP50, Promotes Invasion and Metastasis by Inducing EMT in Gastric Cancer. BMC Cancer, 2018, 18:94–104., DOI 10.1186/ s12885-018-4000-y Song Z-B, Bao Y-L, Zhang Y, Mi X-G, Wu P, Wu Y, et al. Testes-specific Protease 50, TSP50, Promotes Cell Proliferation through the Activation of the Nuclear Factor κB, NF-Κb, Signalling Pathway. Biochem J, 2011, 436:457–67., DOI 10.1042/bj20101780 Qiao W, Shi B, Han Y, Tang HM, Lin J, Hu HY, et al. Testes-Specific Protease 50 and Non-Small Cell Lung Cancer Prognosis. Oncol Lett, 2018, 15: 8796–804., DOI 10.3892/ol.2018.8387 Ke J, Lou J, Zhong R, Chen X, Li J, Liu C, et al. Identification of a Potential Regulatory Variant for Colorectal Cancer Risk Mapping to 3p21.31 in Chinese Population. Sci Rep, 2016, 6:25194., DOI 10.1038/srep25194 Monteith GR, McAndrew D, Faddy HM, and Roberts-Thomson SJ. Calcium and Cancer: Targeting Ca2+ Transport. Nat Rev Cancer, 2007, 7:519–30., DOI 10.1038/nrc2171 Roberts-Thomson SJ, Chalmers SB, and Monteith GR. The Calcium-Signaling Toolkit in Cancer: Remodeling and Targeting. Cold Spring Harb Perspect Biol, 2019, 11:a035204., DOI 10.1101/cshperspect.a035204 Arai M, Otsu K, MacLennan DH, and Periasamy M. Regulation of Sarcoplasmic Reticulum Gene Expression during Cardiac and Skeletal Muscle Development. Am J Physiology-Cell Physiol, 1992, 262:C614–C620., DOI 10.1152/ajpcell.1992.262.3.c614 Papp S, Dziak E, Michalak M, and Opas M. Is All of the Endoplasmic Reticulum Created Equal? The Effects of the Heterogeneous Distribution of Endoplasmic Reticulum Ca2+-Handling Proteins. J Cel Biol, 2003, 160:475–9., DOI 10.1083/jcb.200207136 Pierro C, Sneyers F, Bultynck G, and Roderick HL. ER Ca2+ Release and Store- Operated Ca2+ Entry - Partners in Crime or Independent Actors in Oncogenic Transformation? Cell Calcium, 2019, 82:102061., DOI 10.1016/ j.ceca.2019.102061 Wilson AJ, Byron K, and Gibson PR. Interleukin-8 Stimulates the Migration of Human Colonic Epithelial Cells In Vitro. Clin Sci, 1999, 97:385–90., DOI 10.1042/cs0970385 Sebok K, Woodside D, al-Aoukaty A, Ho AD, Gluck S, and Maghazachi AA. IL-8 Induces the Locomotion of Human IL-2-activated Natural Killer Cells. Involvement of a Guanine Nucleotide Binding, Go, Protein. J Immunol, 1993, 150:1524–34. Masood R, Cai J, Tulpule A, Zheng T, Hamilton A, Sharma S, et al. Interleukin 8 Is an Autocrine Growth Factor and a Surrogate Marker for Kaposi’s Sarcoma. Clin Cancer Res, 2001, 7:2693–702. Zhu YM, Webster SJ, Flower D, and Woll PJ. Interleukin-8/CXCL8 Is a Growth Factor for Human Lung Cancer Cells. Br J Cancer, 2004, 91:1970–6., DOI 10.1038/sj.bjc.6602227 Rubie C, Frick VO, Pfeil S, Wagner M, Kollmar O, Kopp B, et al. Correlation of IL-8 with Induction, Progression and Metastatic Potential of Colorectal Cancer. World J Gastroenterol, 2007, 13(37):4996–5002., DOI 10.3748/wjg.v13.i37.4996 Vieth M, Quirke P, Lambert R, Karsa LV, and Risio M. European Guidelines for Quality Assurance in Colorectal Cancer Screening and Diagnosis. First Edition–Annotations of Colorectal Lesions. Endoscopy, 2012, 44(3):131–9., DOI 10.1055/s-0032-1309798 Hamilton SR, Bosman FT, Boffetta P, Ilyas M, Morreau H, Nakamura SI, et al. Carcinoma of the colon and Rectum In: Bosman FT, Carneiro F, Hruban RH, et al. editors. WHO Classification of Tumors of the Digestive System. Lyon: IARC Press, 2010). p. 134–46. American Joint Committee on Cancer. Colon and Rectum. In: Amin MB, Edge S, Greene F, et al. editors. AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017). Wang F, He W, Jiang C, Guo G, Ke B, Dai Q, et al. Prognostic Value of Inflammation-Based Scores in Patients Receiving Radical Resection for Colorectal Cancer. BMC Cancer, 2018, 18:1102–11., DOI 10.1186/s12885- 018-4842-3 Zheng L, Xie G, Duan G, Yan X, and Li Q. High Expression of Testes-Specific Protease 50 Is Associated with Poor Prognosis in Colorectal Carcinoma. PLoS ONE, 2011, 6(7):e22203., DOI 10.1371/journal.pone.0022203 Chung F-Y, Lin S-R, Lu C-Y, Yeh C-S, Chen F-M, Hsieh J-S, et al. Sarco/ Endoplasmic Reticulum Calcium-ATPase 2 Expression as a Tumor Marker in Colorectal Cancer. Am J Surg Pathol, 2006, 30(8):969–74., DOI 10.1097/ 00000478-200608000-00006 Lee YS, Choi I, Ning Y, Kim NY, Khatchadourian V, Yang D, et al. Interleukin- 8 and its Receptor CXCR2 in the Tumour Microenvironment Promote colon Cancer Growth, Progression and Metastasis. Br J Cancer, 2012, 106:1833–41., DOI 10.1038/bjc.2012.177 Bhalla A, Zulfiqar M, and Bluth MH. Molecular Diagnostics in Colorectal Carcinoma. Clin Lab Med, 2018, 38:311–42., DOI 10.1016/ j.cll.2018.02.008 Wang Y, Yang M, Bao Y, and Li Y. The TSP50 Gene Is Re-expressed in Different Types of Human Cancers. In: Proceedings 2011 International Conference on Human Health and Biomedical Engineering; August 2011; Jilin, China, 2011). p. 1300–2. Yuan J, Wu C, Huang M, Zhou J, Ben W, and Zhang G. TSP50 Depends on its Threonine Protease Activity and its Interactions with TNF-α-Induced NF-Κb for its Role in Human Cervical Tumorigenesis. Cell Biochem Biophys, 2015, 71(2):891–6., DOI 10.1007/s12013-014-0279-8 Liu F, Cao Q, Liu N, Li C, You C, Liu C, et al. Overexpression of Testes-specific Protease 50, TSP50, Predicts Poor Prognosis in Patients with Gastric Cancer. Gastroenterol Res Pract, 2014, 2014:1–7., DOI 10.1155/2014/498246 Xu H-P, Yuan L, Shan J, and Feng H. Localization and Expression of TSP50 Protein in Human and Rodent Testes. Urology, 2004, 64:826–32., DOI 10.1016/ j.urology.2004.05.012 Lee C-J, Ahn H, Jeong D, Pak M, Moon JH, and Kim S. Impact of Mutations in DNA Methylation Modification Genes on Genome-wide Methylation Landscapes and Downstream Gene Activations in Pan-Cancer. BMC Med Genomics, 2020, 13:27–40., DOI 10.1186/s12920-020-0659-4 Evangelista AM, Thompson MD, Weisbrod RM, Pimental DR, Tong X, Bolotina VM, et al. Redox Regulation of SERCA2 Is Required for Vascular Endothelial Growth Factor-Induced Signaling and Endothelial Cell Migration. Antioxid Redox Signaling, 2012, 17:1099–108., DOI 10.1089/ ars.2011.4022 Fan L, Li A, Li W, Cai P, Yang B, Zhang M, et al. Novel Role of Sarco/ endoplasmic Reticulum Calcium ATPase 2 in Development of Colorectal Cancer and its Regulation by F36, a Curcumin Analog. Biomed Pharmacother, 2014, 68(8):1141–8., DOI 10.1016/j.biopha.2014.10.014 Dang D, and Rao R. Calcium-ATPases: Gene Disorders and Dysregulation in Cancer. Biochim Biophys Acta, 2016, 1863(6 Pt B):1344–50., DOI 10.1016/ j.bbamcr.2015.11.016 Izquierdo-Torres E, Hernandez-Oliveras A, Fuentes-Garcia G, and Zarain- Herzberg A. Calcium Signaling and Epigenetics: A Key point to Understand Carcinogenesis. Cell Calcium, 2020, 91:102285., DOI 10.1016/ j.ceca.2020.102285 Zhong T, Pan X, Wang J, Yang B, and Ding L. The Regulatory Roles of Calcium Channels in Tumors. Biochem Pharmacol, 2019, 169:113603., DOI 10.1016/ j.bcp.2019.08.005 Lu M, Sun L, Zhou J, Zhao Y, and Deng X. Dihydroartemisinin-induced Apoptosis Is Associated with Inhibition of Sarco/endoplasmic Reticulum Calcium ATPase Activity in Colorectal Cancer. Cel Biochem Biophys, 2015, 73:137–45., DOI 10.1007/s12013-015-0643-3 Krebs J, Agellon LB, andMichalakM.Ca2+Homeostasis and Endoplasmic Reticulum, ER, Stress: An Integrated View of Calcium Signaling. Biochem Biophysical Res Commun, 2015, 460:114–21., DOI 10.1016/ j.bbrc.2015.02.004 Yang B, Zhang M, Gao J, Li J, Fan L, Xiang G, et al. Small Molecule RL71 Targets SERCA2 at a Novel Site in the Treatment of Human Colorectal Cancer. Oncotarget, 2015, 6:37613–25., DOI 10.18632/oncotarget.6068 Asfaha S, Dubeykovskiy AN, Tomita H, Yang X, Stokes S, Shibata W, et al. Mice that Express Human Interleukin-8 Have Increased Mobilization of Immature Myeloid Cells, Which Exacerbates Inflammation and Accelerates colon Carcinogenesis. Gastroenterology, 2013, 144:155–66., DOI 10.1053/ j.gastro.2012.09.057 Cui G, Yuan A, Goll R, Vonen B, and Florholmen J. Dynamic Changes of Interleukin-8 Network along the Colorectal Adenoma-Carcinoma Sequence. Cancer Immunol Immunother, 2009, 58:1897–905., DOI 10.1007/s00262-009- 0702-y Xia W, Chen W, Zhang Z, Wu D, Wu P, Chen Z, et al. Prognostic Value, Clinicopathologic Features and Diagnostic Accuracy of Interleukin-8 in Colorectal Cancer: a Meta-Analysis. PloS one, 2015, 10(4):e0123484., DOI 10.1371/journal.pone.0123484 Wei H, Li J, Xie M, Lei R, and Hu B. Comprehensive Analysis of Metastasis- Related Genes Reveals a Gene Signature Predicting the Survival of colon Cancer Patients. Peer J, 2018, 6:e5433., DOI 10.7717/peerj.5433 Fisher RC, Bellamkonda K, Alex Molina L, Xiang S, Liska D, Sarvestani SK, et al. Disrupting Inflammation-Associated CXCL8-CXCR1 Signaling Inhibits Tumorigenicity Initiated by Sporadic- and Colitis-Colon Cancer Stem Cells. Neoplasia, 2019, 21:269–81., DOI 10.1016/j.neo.2018.12.007 ShimizuM, and Tanaka N. IL-8-induced O-GlcNAc Modification via GLUT3 and GFAT Regulates Cancer Stem Cell-like Properties in colon and Lung Cancer Cells. Oncogene, 2019, 38:1520–33., DOI 10.1038/s41388-018-0533-4 Bie Y, GeW, Yang Z, Cheng X, Zhao Z, Li S, et al. The Crucial Role of CXCL8 and its Receptors in Colorectal Liver Metastasis. Dis Markers, 2019, 2019: 1–12., DOI 10.1155/2019/8023460 Chan LP, Liu C, Chiang FY, Wang LF, Lee KW, Chen WT, et al. IL-8 Promotes Inflammatory Mediators and Stimulates Activation of P38 MAPK/ERKNF- κB Pathway and Reduction of JNK in HNSCC. Oncotarget, 2017, 8(34):56375–88., DOI 10.18632/oncotarget.16914 Hussain SP, Hofseth LJ, and Harris CC. Radical Causes of Cancer. Nat Rev Cancer, 2003, 3:276–85., DOI 10.1038/nrc1046 Luo B, and Lee AS. The Critical Roles of Endoplasmic Reticulum Chaperones and Unfolded Protein Response in Tumorigenesis and Anticancer Therapies. Oncogene, 2013, 32:805–18., DOI 10.1038/ onc.2012.130 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609990 EP 1610001 DI 10.3389/pore.2021.1609990 PG 12 ER PT J AU Csurgay, K Zalatnai, A Benczik, M Csomo, KB Horvath, F Lorincz, Komlos, Gy Nemeth, Zs AF Csurgay, Katalin Zalatnai, Attila Benczik, Marta Csomo, Krisztian Benedek Horvath, Ferenc Lorincz, Adam Komlos, Gyorgy Nemeth, Zsolt TI A Study of Prognostic Factors in Young Patients With Non-HPV Oral Cancer in Central Europe SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; young adult; risk factors; oral cancer; p16 ID immunohistochemistry; young adult; risk factors; oral cancer; p16 AB The etiological factors of squamous cell carcinomas of the head and neck have been well known for a long time. It is also well known that the incidence of oral cancer diagnosed in younger patients is on the rise. Due to the young age of these patients, the increase in the number of these cases and the fact that many of them neither smoke nor drink alcohol it has been suggested that other factors might be at play in the carcinogenesis of oral cancer. Thus, along the classic etiological factors of smoking and alcohol abuse certain molecular marker anomalies and the human papilloma virus (HPV) have emerged as potential factors. The aim of the present study is to verify the potential prognostic factors and to map the differences in biomarker expression between the young and the old patient groups. In the present study the immunohistochemical profile of samples obtained from oral squamous cell carcinomas was studied and compared with various clinico-pathological parameters. In 88 samples the expressions of p16, p53, Ki67, EGFR were studied with a tissue microarray technique under standard reaction conditions as well as the detection and typing of HPV infection with the Full Spectrum HPV DNA method. The biomarker expression profile of young patients with oral squamous cell carcinoma was compared to that of older patients (above 50). A significant difference was found between the immunohistochemical profile of the young and old patient groups in p16, Ki67 expression. The overall survival and progression free survival were influenced by p16 expression in young age. C1 [Csurgay, Katalin] Semmelweis University, Department of Oral and Maxillofacial SurgeryBudapest, Hungary. [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Benczik, Marta] GenoID Molecular Diagnostic Laboratory, SYNLAB Hungary Ltd.Budapest, Hungary. [Csomo, Krisztian Benedek] Semmelweis University, Department of Oral and Maxillofacial SurgeryBudapest, Hungary. [Horvath, Ferenc] Semmelweis University, Department of Public HealthBudapest, Hungary. [Lorincz, Adam] King’s College, Department of Oral SurgeryLondon, UK. [Komlos, Gyorgy] Semmelweis University, Department of Oral and Maxillofacial SurgeryBudapest, Hungary. [Nemeth, Zsolt] Semmelweis University, Department of Oral and Maxillofacial SurgeryBudapest, Hungary. RP Nemeth, Zs (reprint author), Semmelweis University, Department of Oral and Maxillofacial Surgery, Budapest, Hungary. EM nemeth.zsolt@dent.semmelweisuniv.hu CR World Health Organization. Global Cancer Observatory. Cancer Today, 2018). Available at: https://gco.iarc.fr/today/online-analysis-multi-bars, Accessed November 21, 2021). Turi K, Barabas P, Csurgay K, Lehner G, Lorincz A, Nemeth Z. An Analysis of the Epidemiological and Etiological Factors of Oral Tumors of Young Adults in a Central-Eastern European Population. Pathol Oncol Res, 2013, 19:353–63., DOI 10.1007/s12253-013-9628-y Castellsague X, Alemany L, Quer M, Halec G, Quiros B, Tous S, et al. ICO International HPV in Head and Neck Cancer Study GroupHPV Involvement in Head and Neck Cancers: Comprehensive Assessment of Biomarkers in 3680 Patients. JNCI.J, 2016, 108:djv403., DOI 10.1093/jnci/djv403 Candotto V, Lauritano D, Nardone M, Baggi L, Arcuri C, Gatto R, et al. HPV Infection in the Oral Cavity: Epidemiology, Clinical Manifestations and Relationship with Oral Cancer. Orl, 2017, 10:209–20., DOI 10.11138/orl/ 2017.10.3.209 Belobrov S, Cornall AM, Young RJ, Koo K, Angel C, Wiesenfeld D, et al. The Role of Human Papillomavirus in P16-Positive Oral Cancers. J Oral Pathol Med, 2018, 47:18–24., DOI 10.1111/jop.12649 Lewis A, Kang R, Levine A, Maghami E. The New Face of Head and Neck Cancer: The HPV Epidemic. Oncology, Williston Park,, 2015, 29:616–26. Kreimer AR, Bhatia RK, Messeguer AL, Gonzalez P, Herrero R, Giuliano AR. Oral Human Papillomavirus in Healthy Individuals: a Systematic Review of the Literature. Sex Transm Dis, 2010, 37:386–91., DOI 10.1097/ OLQ.0b013e3181c94a3b Harris SL, Thorne LB, Seaman WT, Neil Hayes D, Couch ME, Kimple RJ. Association of p16INK4a Overexpression with Improved Outcomes in Young Patients with Squamous Cell Cancers of the Oral Tongue. Head Neck, 2011, 33:1622–7., DOI 10.1002/hed.21650 Wang Z, Sturgis EM, Zhang Y, Huang Z, Zhou Q, Wei Q, et al. Combined P53- Related Genetic Variants Together with HPV Infection Increase Oral Cancer Risk. Int J Cancer, 2011, 131:E251–E258., DOI 10.1002/ijc.27335 Sarkaria JN, Harari PM. Oral Tongue Cancer in Young Adults Less Than 40 Years of Age: Rationale for Aggressive Therapy. Head Neck, 1994, 16: 107–11., DOI 10.1002/hed.2880160202 Annertz K, Anderson H, Biorklund A, Moller T, Kantola S, Mork J, et al. Incidence and Survival of Squamous Cell Carcinoma of the Tongue in Scandinavia, with Special Reference to Young Adults. Int J Cancer, 2002, 101:95–9., DOI 10.1002/ijc.10577 Nemeth Z, Turi K, Lehner G, Veres DS, Csurgay K. The Prognostic Role of Age in Oral Cancer. A Clinical Study. Magy Onkol, 2013, 57:166–72. Dobrpossy L. Epidemiology of Head and Neck Cancer: Magnitude of the Problem. Cancer Metastasis Rev, 2005, 24:9–17., DOI 10.1007/s10555-005- 5044-4 Kato K, Kawashiri S, Tanaka A, Noguchi N, Nakaya H, Hase T, et al. Predictive Value of Measuring P53 Labeling Index at the Invasive Front of Oral Squamous Cell Carcinomas. Pathol Oncol Res, 2008, 14:57–61., DOI 10.1007/s12253-008-9022-3 Rodrigo JP, Hermsen MA, Fresno MF, Brakenhoff RH, Garcia-Velasco F, Snijders PJF, et al. Prevalence of Human Papillomavirus in Laryngeal and Hypopharyngeal Squamous Cell Carcinomas in Northern Spain. Cancer Epidemiol, 2015, 39:37–41., DOI 10.1016/j.canep.2014.11.003 Jeney C, Takacs T, Sebe A, Schaff Z. Detection and Typing of 46 Genital Human Papillomaviruses by the L1F/L1R Primer System Based Multiplex PCR and Hybridization. J Virol Methods, 2007, 140:32–42., DOI 10.1016/ j.jviromet.2006.10.013 McCarty KS, Jr, Szabo E, Flowers JL, Cox EB, Leight GS,Miller L, et al. Use of a Monoclonal Anti-estrogen Receptor Antibody in the Immunohistochemical Evaluation of Human Tumors. Cancer Res, 1986, 46:4244s–4248s. Available at: https://cancerres.aacrjournals.org/content/46/8_Supplement/4244s.long, Accessed November 21, 2021). Llewellyn CD, Johnson NW, Warnakulasuriya KAAS. Risk Factors for Squamous Cell Carcinoma of the Oral Cavity in Young People - a Comprehensive Literature Review. Oral Oncol, 2001, 37:401–18., DOI 10.1016/S1368-8375(00)00135-4 Garavello W, Spreafico R, Gaini RM. Oral Tongue Cancer in Young Patients: A Matched Analysis. Oral Oncol, 2007, 43:894–7., DOI 10.1016/ j.oraloncology.2006.10.013 Shiboski CH, Schmidt BL, Jordan RCK. Tongue and Tonsil Carcinoma. Cancer, 2005, 103:1843–9., DOI 10.1002/cncr.20998 Szentkuti G, Danos K, Brauswetter D, Kiszner G, Krenacs T, Csako L, et al. Correlations between Prognosis and Regional Biomarker Profiles in Head and Neck Squamous Cell Carcinomas. Pathol Oncol Res, 2015, 21:643–50., DOI 10.1007/s12253-014-9869-4 Tamas L, Szentkuti G, Eros M, Danos K, Brauswetter D, Szende B, et al. Differential Biomarker Expression in Head and Neck Cancer Correlates with Anatomical Localization. Pathol Oncol Res, 2011, 17:721–7., DOI 10.1007/ s12253-011-9376-9 Adduri RS, Kotapalli V, Gupta NA, Gowrishankar S, Srinivasulu M, Ali MM, et al. P53 Nuclear Stabilization Is Associated with FHIT Loss and Younger Age of Onset in Squamous Cell Carcinoma of Oral Tongue. BMC Clin Pathol, 2014, 14:37., DOI 10.1186/1472-6890-14-37 Brauswetter D, Birtalan E, Danos K, Kocsis A, Krenacs T, Timar J, et al. p16INK4 Expression Is of Prognostic and Predictive Value in Oropharyngeal Cancers Independent of Human Papillomavirus Status: a Hungarian Study. Eur Arch Otorhinolaryngol, 2017, 274:1959–65., DOI 10.1007/s00405-016- 4412-8 Vankos JB, Piurko V, Suba Z, Nemeth Z, Timar J, Kenessey I. The Prognostic Role of Expression of P16 Tumor Suppressor Gene in Hungarian Patients with Oral Squamous Cell Carcinoma. Magy Onkol, 2015, 59:352–9. Adelstein DJ, Ridge JA, Gillison ML, Chaturvedi AK, D’Souza G, Gravitt PE, et al. Head and Neck Squamous Cell Cancer and the Human Papillomavirus: Summary of a National Cancer Institute State of the Science Meeting, November 9-10, 2008, Washington, D.C. Head Neck, 2009, 31:1393–422. Washington, D.C. Head Neck., DOI 10.1002/hed.21269 Gillison ML, Chaturvedi AK, Anderson WF, Fakhry C. Epidemiology of Human Papillomavirus-Positive Head and Neck Squamous Cell Carcinoma. Jco, 2015, 33:3235–42., DOI 10.1200/JCO.2015.61.6995 Mensch K, Szarka K, Mensch H, Dobai A, Magyar Z, Pacurar M, et al. PCR Technique Assisting the Early Diagnosis of Human Papillomavirus a Retrospective Clinical Study. Rev Chim, 2018, 69:2781–7., DOI 10.37358/ rc.18.10.6624 Pytynia KB, Dahlstrom KR, Sturgis EM. Epidemiology of HPV-Associated Oropharyngeal Cancer. Oral Oncol, 2014, 50:380–6., DOI 10.1016/ j.oraloncology.2013.12.019 Kabeya M, Furuta R, Kawabata K, Takahashi S, Ishikawa Y. Prevalence of Human Papillomavirus in mobile Tongue Cancer with Particular Reference to Young Patients. Cancer Sci, 2012, 103:161–8., DOI 10.1111/j.1349- 7006.2011.02149.x Liang X-H, Lewis J, Foote R, Smith D, Kademani D. Prevalence and Significance of Human Papillomavirus in Oral Tongue Cancer: the Mayo Clinic Experience. J Oral Maxill Surg, 2008, 66:1875–80., DOI 10.1016/ j.joms.2008.04.009 Ragin CCR, Taioli E. Survival of Squamous Cell Carcinoma of the Head and Neck in Relation to Human Papillomavirus Infection: Review and Meta- Analysis. Int J Cancer, 2007, 121:1813–20., DOI 10.1002/ijc.22851 Moolmuang B, Tainsky MA. CREG1 Enhances p16INK4a-Induced Cellular Senescence. Cell Cycle, 2011, 10:518–30., DOI 10.4161/cc.10.3.14756 Bu J, Li H, Liu LH, Ouyang YR, Guo HB, Li XY, et al. P16INK4a Overexpression and Survival in Osteosarcoma Patients: a Meta Analysis. Int J Clin Exp Pathol, 2014, 7:6091–6. Wang L, Li J, Yu X, Zhang Z, Pang L, Li S, et al. Prognostic Significance of Overexpressed p16INK4A in Esophageal Squamous Cell Carcinoma: a Meta-Analysis. Biomarkers Med, 2016, 10:537–46., DOI 10.2217/bmm- 2015-0057 Weinberger PM, Yu Z, Haffty BG, Kowalski D, Harigopal M, Sasaki C, et al. Prognostic Significance of P16 Protein Levels in Oropharyngeal Squamous Cell Cancer. Clin Cancer Res, 2004, 10:5684–91., DOI 10.1158/1078-0432.CCR-04-0448 Xu L, Cai J, Yang Q, Ding H, Wu L, Li T, et al. Prognostic Significance of Several Biomarkers in Epithelial Ovarian Cancer: aMeta-Analysis of Published Studies. J Cancer Res Clin Oncol, 2013, 139:1257–77., DOI 10.1007/s00432-013- 1435-z Mihic-Probst D,Mnich CD, Oberholzer PA, Seifert B, Sasse B, Moch H, et al. p16 Expression in Primary Malignant Melanoma Is Associated with Prognosis and Lymph Node Status. Int J Cancer, 2006, 118:2262–8., DOI 10.1002/ijc.21608 Ko E, Kim Y, Kim S-J, Joh J-W, Song S, Park C-K, et al. Promoter Hypermethylation of the P16 Gene Is Associated with Poor Prognosis in Recurrent Early-Stage Hepatocellular Carcinoma. Cancer Epidemiol Biomarkers Prev, 2008, 17:2260–7., DOI 10.1158/1055-9965.EPI-08-0236 Takita J, Hayashi Y, Kohno T, Yamaguchi N, Hanada R, Yamamoto K, et al. Deletion Map of Chromosome 9 and P16, CDKN2A, Gene Alterations in Neuroblastoma. Cancer Res, 1997, 57:907–12. Available at: https://cancerres. aacrjournals.org/content/57/5/907, Accessed November 21, 2021). Halvorsen OJ, Hostmark J, Haukaas S, Hoisater P, Akslen LA. Prognostic Significance of P16 and CDK4 Proteins in Localized Prostate Carcinoma. Cancer, 2000, 88:416–24., DOI 10.1002/(sici)1097-0142(20000115)88:2<416: aid-cncr24>3.0.co;2-# Miller C, Shay A, Tajudeen B, Sen N, Fidler M, Stenson K, et al. Clinical Features and Outcomes in Young Adults with Oral Tongue Cancer. Am J Otolaryngol, 2019, 40:93–6., DOI 10.1016/j.amjoto.2018.09.022 Costa V, Kowalski LP, Coutinho-Camillo CM, Begnami MD, Calsavara VF, Neves JI, et al. EGFR Amplification and Expression in Oral Squamous Cell Carcinoma in Young Adults. Int J Oral Maxill Surg, 2018, 47:817–23., DOI 10.1016/j.ijom.2018.01.002 Kumar B, Cordell KG, Lee JS, Worden FP, Prince ME, Tran HH, et al. EGFR, P16, HPV Titer, Bcl-xL and P53, Sex, and Smoking as Indicators of Response to Therapy and Survival in Oropharyngeal Cancer. Jco, 2008, 26:3128–37., DOI 10.1200/JCO.2007.12.7662 Szabo B, Nelhubel GA, Karpati A, Kenessey I, Jori B, Szekely C, et al. Clinical Significance of Genetic Alterations and Expression of Epidermal Growth Factor Receptor, EGFR, in Head and Neck Squamous Cell Carcinomas. Oral Oncol, 2011, 47:487–96., DOI 10.1016/j.oraloncology.2011.03.020 Hiraishi Y, Wada T, Nakatani K, Negoro K, Fujita S. Immunohistochemical Expression of EGFR and P-EGFR in Oral Squamous Cell Carcinomas. Pathol Oncol Res, 2006, 12:87–91., DOI 10.1007/BF02893450 Sittel C, Ruiz S, Volling P, Kvasnicka HM, Jungehulsing M, Eckel HE. Prognostic Significance of Ki-67, MIB1), PCNA and P53 in Cancer of the Oropharynx and Oral Cavity. Oral Oncol, 1999, 35:583–9., DOI 10.1016/S1368- 8375(99)00041-X Xie S, Liu Y, Qiao X, Hua R-X,Wang K, Shan X-F, et al. What Is the Prognostic Significance of Ki-67 Positivity in Oral Squamous Cell Carcinoma?. J Cancer, 2016, 7:758–67. Available at:., DOI 10.7150/jca.14214http://www.jcancer.org/ v07p0758.htm, Accessed November 21, 2021). NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609991 EP 1610001 DI 10.3389/pore.2021.1609991 PG 11 ER PT J AU Tang, J Li, Y Liu, B Liang, W Hu, S Shi, M Zeng, J Li, M Huang, M AF Tang, Jiefu Li, Yaling Liu, Boxuan Liang, Wei Hu, Sanbao Shi, Meilian Zeng, Jie Li, Mingzhen Huang, Minjiang TI Uncovering a Key Role of ETS1 on Vascular Abnormality in Glioblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE glioblastoma; ETS1; endothelial; tumor vessel; vascular abnormality ID glioblastoma; ETS1; endothelial; tumor vessel; vascular abnormality AB Glioblastoma (GBM) is the most aggressive type of brain tumor. Microvascular proliferation and abnormal vasculature are the hallmarks of the GBM, aggravating disease progression and increasing patient morbidity. Here, we uncovered a key role of ETS1 on vascular abnormality in glioblastoma. ETS1 was upregulated in endothelial cells from human tumors compared to endothelial cells from paired control brain tissue. Knockdown of Ets1 in mouse brain endothelial cells inhibited cell migration and proliferation, and suppressed expression of genes associated with vascular abnormality in GBM. ETS1 upregulation in tumor ECs was dependent on TGFβ signaling, and targeting TGFβ signaling by inhibitor decreased tumor angiogenesis and vascular abnormality in CT-2A glioma model. Our results identified ETS1 as a key factor regulating tumor angiogenesis, and suggested that TGFβ inhibition may suppress the vascular abnormality driven by ETS1. C1 [Tang, Jiefu] The First Affiliated Hospital of Hunan University of Medicine, Trauma CenterHuaihua, China. [Li, Yaling] Xi’an People’s Hospital (Xi’an Fourth Hospital), Department of Obstetrics and GynaecologyXi’an, China. [Liu, Boxuan] The Second People’s Hospital of Huaihua, Precision Medicine CenterHuaihua, China. [Liang, Wei] The Second People’s Hospital of Huaihua, Department of OrthopaedicsHuaihua, China. [Hu, Sanbao] Capital Medical University, Beijing Anzhen Hospital, Department of OrthopaedicsBeijing, China. [Shi, Meilian] The Second People’s Hospital of Huaihua, Department of Infectious DiseasesHuaihua, China. [Zeng, Jie] The Second People’s Hospital of Huaihua, Department of OrthopaedicsHuaihua, China. [Li, Mingzhen] The Second People’s Hospital of Huaihua, Precision Medicine CenterHuaihua, China. [Huang, Minjiang] Hunan University of MedicineHuaihua, China. RP Huang, M (reprint author), Hunan University of Medicine, Huaihua, China. EM zhenduan2010@126.com CR Ostrom QT, Gittleman H, Liao P, Vecchione-Koval T, Wolinsky Y, Kruchko C, et al. CBTRUS Statistical Report: Primary Brain and Other central Nervous System Tumors Diagnosed in the United States in 2010-2014. Neuro Oncol, 2017, 19:v1–v88., DOI 10.1093/neuonc/nox158 Seano G, Jain RK. Vessel Co-option in Glioblastoma: Emerging Insights and Opportunities. Angiogenesis, 2020, 23(1):9–16., DOI 10.1007/s10456-019- 09691-z Yang F, Xie Y, Tang J, Liu B, Luo Y, He Q, et al. Uncovering a Distinct Gene Signature in Endothelial Cells Associated with Contrast Enhancement in Glioblastoma. Front Oncol, 2021, 11:683367., DOI 10.3389/fonc.2021.683367 Xie Y, He L, Lugano R, Zhang Y, Cao H, He Q, et al. KeyMolecular Alterations in Endothelial Cells in Human Glioblastoma Uncovered through Single-Cell RNA Sequencing. JCI Insight, 2021, 6(15):e1508., DOI 10.1172/ jci.insight.150861 de Groot JF, Fuller G, Kumar AJ, Piao Y, Eterovic K, Ji Y, et al. Tumor Invasion after Treatment of Glioblastoma with Bevacizumab: Radiographic and Pathologic Correlation in Humans and Mice. Neuro-Oncology, 2010, 12(3): 233–42., DOI 10.1093/neuonc/nop027 Du R, Lu KV, Petritsch C, Liu P, Ganss R, Passegue E, et al. HIF1α Induces the Recruitment of Bone Marrow-Derived Vascular Modulatory Cells to Regulate Tumor Angiogenesis and Invasion. Cancer Cell, 2008, 13(3):206–20., DOI 10.1016/j.ccr.2008.01.034 Dimberg A. The Glioblastoma Vasculature as a Target for Cancer Therapy. Biochem Soc Trans, 2014, 42(6):1647–52., DOI 10.1042/BST20140278 Langenkamp E, Zhang L, Lugano R, Huang H, Elhassan TEA, Georganaki M, et al. Elevated Expression of the C-type Lectin CD93 in the Glioblastoma Vasculature Regulates Cytoskeletal Rearrangements that Enhance Vessel Function and Reduce Host Survival. Cancer Res, 2015, 75(21):4504–16., DOI 10.1158/0008-5472.CAN-14-3636 Zhang L, Dimberg A. Pleiotrophin Is a Driver of Vascular Abnormalization in Glioblastoma. Mol Cell Oncol, 2016, 3(6):e1141087., DOI 10.1080/ 23723556.2016.1141087 Zhang L, He L, Lugano R, Roodakker K, Bergqvist M, Smits A, et al. IDH Mutation Status Is Associated with Distinct Vascular Gene Expression Signatures in Lower-Grade Gliomas. Neuro Oncol, 2018, 20(11):1505–16., DOI 10.1093/neuonc/noy088 Zhang L, Kundu S, Feenstra T, Li X, Jin C, Laaniste L, et al. Pleiotrophin Promotes Vascular Abnormalization in Gliomas and Correlates with Poor Survival in Patients with Astrocytomas. Sci Signal, 2015, 8(406):ra125., DOI 10.1126/scisignal.aaa1690 Zhang L, Laaniste L, Jiang Y, Alafuzoff I, Uhrbom L, Dimberg A. Pleiotrophin Enhances PDGFB-Induced Gliomagenesis through Increased Proliferation of Neural Progenitor Cells. Oncotarget, 2016, 7(49):80382–90., DOI 10.18632/ oncotarget.12983 Cohen MH, Shen YL, Keegan P, Pazdur R. FDA Drug Approval Summary: Bevacizumab, Avastin, as Treatment of Recurrent Glioblastoma Multiforme. The Oncologist, 2009, 14(11):1131–8., DOI 10.1634/theoncologist.2009-0121 Arvanitis CD, Ferraro GB, Jain RK. The Blood-Brain Barrier and Blood- Tumour Barrier in Brain Tumours and Metastases. Nat Rev Cancer, 2020, 20(1):26–41., DOI 10.1038/s41568-019-0205-x Craig MP, Sumanas S. ETS Transcription Factors in Embryonic Vascular Development. Angiogenesis, 2016, 19(3):275–85., DOI 10.1007/s10456-016- 9511-z Bories J-C, Willerford DM, Grevin D, Davidson L, Camus A, Martin P, et al. Increased T-Cell Apoptosis and Terminal B-Cell Differentiation Induced by Inactivation of the Ets-1 Proto-Oncogene. Nature, 1995, 377(6550):635–8., DOI 10.1038/377635a0 Muthusamy N, Barton K, Leiden JM. Defective Activation and Survival of T Cells Lacking the Ets-1 Transcription Factor. Nature, 1995, 377(6550): 639–42., DOI 10.1038/377639a0 Garrett-Sinha LA. Review of Ets1 Structure, Function, and Roles in Immunity. Cell. Mol. Life Sci., 2013, 70(18):3375–90., DOI 10.1007/s00018-012-1243-7 Iwasaka C, Tanaka K, Abe M, Sato Y. Ets-1 Regulates Angiogenesis by Inducing the Expression of Urokinase-type Plasminogen Activator and Matrix Metalloproteinase-1 and the Migration of Vascular Endothelial Cells. J Cel Physiol., 1996, 169(3):522–31., DOI 10.1002/(SICI)1097-4652 Tanaka K, Oda N, Iwasaka C, AbeM, Sato Y. Induction of Ets-1 in Endothelial Cells during Reendothelialization after Denuding Injury. J Cel Physiol., 1998, 176(2):235–44., DOI 10.1002/(SICI)1097-4652 Chen Z, Fisher RJ, Riggs CW, Rhim JS, Lautenberger JA. Inhibition of Vascular Endothelial Growth Factor-Induced Endothelial Cell Migration by ETS1 Antisense Oligonucleotides. Cancer Res, 1997, 57(10):2013–9. Sato Y, Teruyama K, Nakano T, Oda N, Abe M, Tanaka K, et al. Role of Transcription Factors in Angiogenesis: Ets-1 Promotes Angiogenesis as Well as Endothelial Apoptosis. Ann N Y Acad Sci, 2001, 947:117–23., DOI 10.1111/ j.1749-6632.2001.tb03934.x Teruyama K, Abe M, Nakano T, Iwasaka-Yagi C, Takahashi S, Yamada S, et al. Role of Transcription Factor Ets-1 in the Apoptosis of Human Vascular Endothelial Cells. J Cel Physiol., 2001, 188(2):243–52., DOI 10.1002/jcp.1112 Wernert N, Stanjek A, Kiriakidis S, Hugel A, Jha HC, Mazitschek R, et al. Inhibition of Angiogenesis In Vivo Byets-1 Antisense Oligonucleotides- Inhibition of Ets-1 Transcription Factor Expression by the Antibiotic Fumagillin. Angew Chem Int Ed, 1999, 38(21):3228–31., DOI 10.1002/(sici, 1521-3773(19991102) Casie Chetty S, Sumanas S. Ets1 Functions Partially Redundantly with Etv2 to Promote Embryonic Vasculogenesis and Angiogenesis in Zebrafish. Developmental Biol, 2020, 465(1):11–22., DOI 10.1016/j.ydbio.2020.06.007 Wei G, Srinivasan R, Cantemir-Stone CZ, Sharma SM, Santhanam R, Weinstein M, et al. Ets1 and Ets2 Are Required for Endothelial Cell Survival during Embryonic Angiogenesis. Blood, 2009, 114(5):1123–30., DOI 10.1182/blood-2009-03-211391 Shaik S, Maegawa S, Haltom AR, Wang F, Xiao X, Dobson T, et al. REST Promotes ETS1-dependent Vascular Growth in Medulloblastoma. Mol Oncol, 2021, 15(5):1486–506., DOI 10.1002/1878-0261.12903 Dieterich LC, Mellberg S, Langenkamp E, Zhang L, Zieba A, Salomaki H, et al. Transcriptional Profiling of Human Glioblastoma Vessels Indicates a Key Role of VEGF-A and TGFβ2 in Vascular Abnormalization. J Pathol, 2012, 228(3): 378–90., DOI 10.1002/path.4072 Martin Sobral L, Sechler M, Parrish JK, McCann TS, Jones KL, Black JC, et al. KDM3A/Ets1/MCAM axis Promotes Growth and Metastatic Properties in Rhabdomyosarcoma. Genes Cancer, 2020, 11(1-2):53–65., DOI 10.18632/ genesandcancer.200 Brandt MM, van Dijk CGM, Chrifi I, Kool HM, Burgisser PE, Louzao- Martinez L, et al. Endothelial Loss of Fzd5 Stimulates PKC/Ets1-mediated Transcription of Angpt2 and Flt1. Angiogenesis, 2018, 21(4):805–21., DOI 10.1007/s10456-018-9625-6 Yang L-X, Guo H-B, Liu S-Y, Feng H-P, Shi J. ETS1 Promoted Cell Growth, Metastasis and Epithelial-Mesenchymal Transition Process in Melanoma by Regulating miR-16-Mediated SOX4 Expression. Melanoma Res, 2021, 31(4): 298–308., DOI 10.1097/CMR.0000000000000743 Bhattacharya R, Ray Chaudhuri S, Roy SS. FGF9-induced Ovarian Cancer Cell Invasion Involves VEGF-A/VEGFR2 Augmentation by Virtue of ETS1 Upregulation and Metabolic Reprogramming. J Cel Biochem., 2018, 119(10):8174–89., DOI 10.1002/jcb.26820 Srivastava N, Bishnoi A, Mehta S, Rani S, Kumar R, Bhardwaj S, et al. Aberrant ETS-1 Signalling Impedes the Expression of Cell Adhesion Molecules and Matrix Metalloproteinases in Non-segmental Vitiligo. Exp Dermatol, 2020, 29(6):539–47., DOI 10.1111/exd.14107 Sindi HA,RussomannoG, Satta S, Abdul-SalamVB, JoKB,Qazi-Chaudhry B, et al. Therapeutic Potential of KLF2-Induced Exosomal microRNAs in Pulmonary Hypertension. Nat Commun, 2020, 11(1):1185., DOI 10.1038/s41467-020-14966-x Zhu Y, Ye M, Xu H, Gu R, Ma X, Chen M, et al. Methylation Status of CpG Sites in the NOTCH4 Promoter Region Regulates NOTCH4 Expression in Patients with Tetralogy of Fallot. Mol Med Rep, 2020, 22(5):4412–22., DOI 10.3892/mmr.2020.11535 Gundersen HJG, Bendtsen TF, Korbo L, Marcussen N, Moller A, Nielsen K, et al. Some New, Simple and Efficient Stereological Methods and Their Use in Pathological Research and Diagnosis. APMIS, 1988, 96(5):379–94., DOI 10.1111/j.1699-0463.1988.tb05320.x Wassberg E, Hedborg F, Skoldenberg E, Stridsberg M, Christofferson R. Inhibition of Angiogenesis Induces Chromaffin Differentiation and Apoptosis in Neuroblastoma. Am J Pathol, 1999, 154(2):395–403., DOI 10.1016/ S0002-9440(10)65286-8 Wang L, Babikir H, Muller S, Yagnik G, Shamardani K, Catalan F, et al. The Phenotypes of Proliferating Glioblastoma Cells Reside on a Single Axis of Variation. Cancer Discov, 2019, 9(12):1708–19., DOI 10.1158/2159-8290.CD- 19-0329 Colas-Algora N, Garcia-Weber D, Cacho-Navas C, Barroso S, Caballero A, Ribas C, et al. Compensatory Increase of VE-Cadherin Expression through ETS1 Regulates Endothelial Barrier Function in Response to TNFα. Cel Mol. Life Sci., 2020, 77(11):2125–40., DOI 10.1007/s00018-019-03260-9 Sato Y, Abe M, Tanaka K, Iwasaka C, Oda N, Kanno S, et al. Signal Transduction and Transcriptional Regulation of Angiogenesis. Adv Exp Med Biol, 2000, 476:109–15., DOI 10.1007/978-1-4615-4221-6_9 Okano K, Hibi A, Miyaoka T, Inoue T, Sugimoto H, Tsuchiya K, et al. Inhibitory Effects of the Transcription Factor Ets-1 on the Expression of Type I Collagen in TGF-B1-Stimulated Renal Epithelial Cells. Mol Cel Biochem, 2012, 369(1-2):247–54., DOI 10.1007/s11010-012-1388-6 Rodon J, Carducci MA, Sepulveda-Sanchez JM, Azaro A, Calvo E, Seoane J, et al. First-in-Human Dose Study of the Novel Transforming Growth Factor-β Receptor I Kinase Inhibitor LY2157299 Monohydrate in Patients with Advanced Cancer and Glioma. Clin Cancer Res, 2015, 21(3):553–60., DOI 10.1158/1078-0432.CCR-14-1380 Zhang Y, Xie Y, He L, Tang J, He Q, Cao Q, et al. 1p/19q Co-deletion Status Is Associated with Distinct Tumor-Associated Macrophage Infiltration in IDH Mutated Lower-Grade Gliomas. Cell Oncol., 2021, 44(1):193–204., DOI 10.1007/s13402-020-00561-1 Dejana E, Taddei A, Randi A. Foxs and Ets in the Transcriptional Regulation of Endothelial Cell Differentiation and Angiogenesis. Biochim Biophys Acta, Bba, - Rev Cancer, 2007, 1775(2):298–312., DOI 10.1016/j.bbcan.2007.05.003 Hashiya N, Jo N, Aoki M, Matsumoto K, Nakamura T, Sato Y, et al. In Vivo Evidence of Angiogenesis Induced by Transcription Factor Ets-1. Circulation, 2004, 109(24):3035–41., DOI 10.1161/01.CIR.0000130643.41587 Nakano T, Abe M, Tanaka K, Shineha R, Satomi S, Sato Y. Angiogenesis Inhibition by Transdominant Mutant Ets-1. J Cel Physiol., 2000, 184(2): 255–62., DOI 10.1002/1097-4652 Watanabe D, Takagi H, Suzuma K, Suzuma I, Oh H, Ohashi H, et al. Transcription Factor Ets-1 Mediates Ischemia- and Vascular Endothelial Growth Factor-dependent Retinal Neovascularization. Am J Pathol, 2004, 164(5):1827–35., DOI 10.1016/S0002-9440(10)63741-8 Chen J, Fu Y, Day DS, Sun Y, Wang S, Liang X, et al. VEGF Amplifies Transcription through ETS1 Acetylation to Enable Angiogenesis. Nat Commun, 2017, 8(1):383., DOI 10.1038/s41467-017-00405-x Lobov IB, Brooks PC, Lang RA. Angiopoietin-2 Displays VEGF-dependent Modulation of Capillary Structure and Endothelial Cell Survival In Vivo. Proc Natl Acad Sci, 2002, 99(17):11205–10., DOI 10.1073/pnas.172161899 Scholz A, Harter PN, Cremer S, Yalcin BH, Gurnik S, Yamaji M, et al. Endothelial Cell-derived Angiopoietin-2 Is a Therapeutic Target in Treatment-naive and Bevacizumab-resistant Glioblastoma. EMBO Mol Med, 2016, 8(1):39–57., DOI 10.15252/emmm.201505505 Tsai C-N, Yu S-C, Lee C-W, Pang J-HS, Wu C-H, Lin S-E, et al. SOX4 Activates CXCL12 in Hepatocellular Carcinoma Cells to Modulate Endothelial Cell Migration and Angiogenesis In Vivo. Oncogene, 2020, 39(24):4695–710., DOI 10.1038/s41388-020-1319-z Vervoort SJ, de Jong OG, Roukens MG, Frederiks CL, Vermeulen JF, Lourenco AR, et al. Global Transcriptional Analysis Identifies a Novel Role for SOX4 in Tumor-Induced Angiogenesis. Elife, 2018, e27706:7., DOI 10.7554/eLife.27706 Wragg JW, Finnity JP, Anderson JA, Ferguson HJM, Porfiri E, Bhatt RI, et al. MCAM and LAMA4 Are Highly Enriched in Tumor Blood Vessels of Renal Cell Carcinoma and Predict Patient Outcome. Cancer Res, 2016, 76(8): 2314–26., DOI 10.1158/0008-5472.CAN-15-1364 Han J, Alvarez-Breckenridge CA, Wang QE, Yu J. TGF-β Signaling and its Targeting for Glioma Treatment. Am J Cancer Res, 2015, 5(3):945–55. Mangani D, Weller M, Seyed Sadr E, Willscher E, Seystahl K, Reifenberger G, et al. Limited Role for Transforming Growth Factor-β Pathway Activation- Mediated Escape from VEGF Inhibition in Murine Glioma Models. Neuro Oncol, 2016, 18(12):1610–21., DOI 10.1093/neuonc/now112 Schafer CM, Gurley JM, Kurylowicz K, Lin PK, Chen W, Elliott MH, et al. An Inhibitor of Endothelial ETS Transcription Factors Promotes Physiologic and Therapeutic Vessel Regression. Proc Natl Acad Sci USA, 2020, 117(42): 26494–502., DOI 10.1073/pnas.2015980117 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1609997 EP 1610006 DI 10.3389/pore.2021.1609997 PG 10 ER PT J AU Mell, L Herrera, F AF Mell, K. Loren Herrera, G. Fernanda TI SMARCA4-Deficient Carcinoma of Uterine Cervix Resembling SCCOHT—Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE cervical cancer; diagnostic biomarker; predictive marker; gynecological cancer; high-risk; personalized treatment; case report ID cervical cancer; diagnostic biomarker; predictive marker; gynecological cancer; high-risk; personalized treatment; case report AB Small cell carcinoma of hypercalcemic type (SCCOHT) is a rare gynaecological neoplasm, originating mostly in the ovaries. Cervical origin of this very aggressive malignancy with unknown histogenesis is an extremely rare condition, without published management recommendations. Alterations in SMARCA4 gene are supposed to play the major role in SCCOHT oncogenesis and their identification is crucial for the diagnosis. Adequate genetic counselling of the patients and their families seems to be of great importance. Optimal management and treatment approaches are not known yet but may extremely influence the prognosis of young female patients that suffer from this very resistant disease. Nowadays, a translational research seems to be the key for the further diagnostic and treatment strategies of SCCOHT. The purpose of the case report is to provide practical information and useful recommendations on the diagnosis, management, and treatment of SMARCA4-deficient carcinoma of the uterine cervix resembling SCCOHT. C1 [Mell, K. Loren] University of California, Department of Radiation Medicine and Applied Sciences, La JollaSan Diego, CA, USA. [Herrera, G. Fernanda] University of Lausanne, Lausanne University Hospital, Ludwig Institute for Cancer ResearchLausanne, Switzerland. CR Patibandla JR, Fehniger JE, Levine DA, Jelinic P. Small Cell Cancers of the Female Genital Tract: Molecular and Clinical Aspects. Gynecol Oncol, 2018, 149(2):420–7., DOI 10.1016/j.ygyno.2018.02.004 Richard Dickersin G, Kline IW, Scully RE. Small Cell Carcinoma of the Ovary with Hypercalcemia: A Report of Eleven Cases. Cancer, 1982, 49(1):188–97., DOI 10.1002/1097-0142(19820101)49:1<188:aid-cncr2820490137>3.0.co;2-d Young RH, Oliva E, Scully RE. Small Cell Carcinoma of the Ovary, Hypercalcemic Type. Am J Surg Pathol, 1994, 18(11):1102–16., DOI 10.1097/ 00000478-199411000-00004 Lavrut P-M, Le Loarer F, Normand C, Grosos C, Dubois R, Buenerd A, et al. Small Cell Carcinoma of the Ovary, Hypercalcemic Type: Report of a Bilateral Case in a Teenager Associated with SMARCA4 Germline Mutation. Pediatr Dev Pathol, 2016, 19(1):56–60., DOI 10.2350/15-04-1624-CR.1 Witkowski L, Donini N, Byler-Dann R, Knost JA, Albrecht S, Berchuck A, et al. The Hereditary Nature of Small Cell Carcinoma of the Ovary, Hypercalcemic Type: Two New Familial Cases. Fam Cancer, 2017, 16(3):395–9., DOI 10.1007/ s10689-016-9957-6 Chen L, Dinh TA, Haque A. Small Cell Carcinoma of the Ovary with Hypercalcemia and Ectopic Parathyroid Hormone Production. Arch Pathol Labor Med, 2005, 129(4):531–3., DOI 10.5858/2005-129-531-sccoto Matias-Guiu X, Prat J, Young RH, Capen CC, Rosol TJ, Delellis RA, et al. Human Parathyroid Hormone-related Protein in Ovarian Small Cell Carcinoma. An Immunohistochemical Study. Cancer, 1994, 73(7):1878–81., DOI 10.1002/1097- 0142(19940401)73:7<1878:AID-CNCR2820730717>3.0.CO;2-D Wynn D, Everett GD, Boothby RA. Small Cell Carcinoma of the Ovary with Hypercalcemia Causes Severe Pancreatitis and Altered Mental Status. Gynecol Oncol, 2004, 95(3):716–8., DOI 10.1016/j.ygyno.2004.08.038 Bourgain A, Acker O, Lambaudie E, Boukerrou M, Chevalier-Place A, Meignie P, et al. Small Cell Carcinoma of the Ovary of the Hypercalcemic Type Revealed by a Severe Acute Pancreatitis: about One Case. Gynecol Obstet Fertil, 2005, 33(1–2):35–8., DOI 10.1016/j.gyobfe.2004.11.004 Nasioudis D, Chapman-Davis E, Frey MK, Caputo TA, Witkin SS, Holcomb K. Small Cell Carcinoma of the Ovary: A Rare Tumor with a Poor Prognosis. Int J Gynecol Cancer, 2018, 28(5):932–8., DOI 10.1097/IGC.0000000000001243 Lu B, Shi H. An In-Depth Look at Small Cell Carcinoma of the Ovary, Hypercalcemic Type, SCCOHT): Clinical Implications from Recent Molecular Findings. J Cancer, 2019, 10(1):223–37., DOI 10.7150/jca.26978 Tischkowitz M, Huang S, Banerjee S, Hague J, Hendricks WPD, Huntsman DG, et al. Small-Cell Carcinoma of the Ovary, Hypercalcemic Type-Genetics, New Treatment Targets, and Current Management Guidelines. Clin Cancer Res, 2020, 26(15):3908–17., DOI 10.1158/1078-0432.CCR-19-3797 Foulkes WD, Clarke BA, Hasselblatt M, Majewski J, Albrecht S, McCluggage WG. No Small surprise - Small Cell Carcinoma of the Ovary, Hypercalcaemic Type, Is a Malignant Rhabdoid Tumour. J Pathol, 2014, 233(3):209–14., DOI 10.1002/path.4362 Fahiminiya S, Witkowski L, Nadaf J, Carrot-Zhang J, Goudie C, Hasselblatt M, et al. Molecular Analyses Reveal Close Similarities between Small Cell Carcinoma of the Ovary, Hypercalcemic Type and Atypical Teratoid/ rhabdoid Tumor. Oncotarget, 2016, 7(2):1732–40., DOI 10.18632/ oncotarget.6459 Schrader KA, Hurlburt J, Kalloger SE, Hansford S, Young S, Huntsman DG, et al. Germline BRCA1 and BRCA2 Mutations in Ovarian Cancer. Obstet Gynecol, 2012, 120(2):235–40., DOI 10.1097/AOG.0b013e31825f3576 Ramos P, Karnezis AN, Craig DW, Sekulic A, Russell ML, Hendricks WPD, et al. Small Cell Carcinoma of the Ovary, Hypercalcemic Type, Displays Frequent Inactivating Germline and Somatic Mutations in SMARCA4. Nat Genet, 2014, 46(5):427–9., DOI 10.1038/ng.2928 Witkowski L, Carrot-Zhang J, Albrecht S, Fahiminiya S, Hamel N, Tomiak E, et al. Germline and Somatic SMARCA4 Mutations Characterize Small Cell Carcinoma of the Ovary, Hypercalcemic Type. Nat Genet, 2014, 46(5):438–43., DOI 10.1038/ng.2931 Jelinic P, Mueller JJ, Olvera N, Dao F, Scott SN, Shah R, et al. Recurrent SMARCA4 Mutations in Small Cell Carcinoma of the Ovary. Nat Genet, 2014, 46(5):424–6., DOI 10.1038/ng.2922 Jelinic P, Ricca J, Van Oudenhove E, Olvera N, Merghoub T, Levine DA, et al. Immune-active Microenvironment in Small Cell Carcinoma of the Ovary, Hypercalcemic Type: Rationale for Immune Checkpoint Blockade. J Natl Cancer Instit, 2018, 110(7):787–90., DOI 10.1093/jnci/djx277 Dong A, Lu Y, Lu B. Genomic/epigenomic Alterations in Ovarian Carcinoma: Translational Insight into Clinical Practice. J Cancer, 2016, 7(11):1441–51., DOI 10.7150/jca.15556 Lin DI, Chudnovsky Y, Duggan B, Zajchowski D, Greenbowe J, Ross JS, et al. Comprehensive Genomic Profiling Reveals Inactivating SMARCA4 Mutations and Low Tumor Mutational burden in Small Cell Carcinoma of the Ovary, Hypercalcemic-type. Gynecol Oncol, 2017, 147(3):626–33., DOI 10.1016/ j.ygyno.2017.09.031 Neigeborn L, Carlson M. Genes Affecting the Regulation of SUC2 Gene Expression by Glucose Repression in Saccharomyces cerevisiae. Genetics, 1984, 108(4):845–58., DOI 10.1093/genetics/108.4.845 Pulice JL, Kadoch C. Composition and Function of Mammalian SWI/SNF Chromatin Remodeling Complexes in Human Disease. Cold Spring Harb Symp Quant Biol, 2016, 81(1):53–60., DOI 10.1101/sqb.2016.81.031021 Stern M, Jensen R, Herskowitz I. Five SWI Genes Are Required for Expression of the HO Gene in Yeast. J Mol Biol, 1984, 178(4):853–68., DOI 10.1016/0022- 2836(84)90315-2 Clapier CR, Iwasa J, Cairns BR, Peterson CL. Mechanisms of Action and Regulation of ATP-dependent Chromatin-Remodelling Complexes. Nat Rev Mol Cel Biol, 2017, 18(7):407–22., DOI 10.1038/nrm.2017.26 Shain AH, Pollack JR. The Spectrum of SWI/SNF Mutations, Ubiquitous in Human Cancers. PLoS ONE, 2013, 8(1):e55119., DOI 10.1371/journal.pone.0055119 Callegaro-Filho D, Gershenson DM, Nick AM, Munsell MF, Ramirez PT, Eifel PJ, et al. Small Cell Carcinoma of the Ovary-Hypercalcemic Type, SCCOHT): A Review of 47 Cases. Gynecol Oncol, 2016, 140(1):53–7., DOI 10.1016/ j.ygyno.2015.11.004 Witkowski L, Goudie C, Ramos P, Boshari T, Brunet J-S, Karnezis AN, et al. The Influence of Clinical and Genetic Factors on Patient Outcome in Small Cell Carcinoma of the Ovary, Hypercalcemic Type. Gynecol Oncol, 2016, 141(3):454–60., DOI 10.1016/j.ygyno.2016.03.013 Pautier P, Ribrag V, Duvillard P, Rey A, Elghissassi I, Sillet-Bach I, et al. Results of a Prospective Dose-Intensive Regimen in 27 Patients with Small Cell Carcinoma of the Ovary of the Hypercalcemic Type. Ann Oncol, 2007, 18(12):1985–9., DOI 10.1093/annonc/mdm376 Distelmaier F, Calaminus G, Harms D, Strater R, Kordes U, Fleischhack G, et al. Ovarian Small Cell Carcinoma of the Hypercalcemic Type in Children and Adolescents. Cancer, 2006, 107(9):2298–306., DOI 10.1002/cncr.22213 Bakhru A, Liu JR, Lagstein A. A Case of Small Cell Carcinoma of the Ovary Hypercalcemic Variant in a Teenager. Gynecol Oncol Case Rep, 2012, 2(4): 139–42., DOI 10.1016/j.gynor.2012.09.001 Isonishi S, Nishii H, Saitou M, Yasuda M, Kiyokawa T, Fukunaga M, et al. Small Cell Carcinoma of the Ovary: Clinical and Biological Study. Int J Clin Oncol, 2008, 13(2):161–5., DOI 10.1007/s10147-007-0698-2 Harrison ML, Hoskins P, du Bois A, Quinn M, Rustin GJS, Ledermann JA, et al. Small Cell of the Ovary, Hypercalcemic Type-Analysis of Combined Experience and Recommendation for Management. A GCIG Study. Gynecol Oncol, 2006, 100(2):233–8., DOI 10.1016/j.ygyno.2005.10.024 Gamwell LF, Gambaro K, Merziotis M, Crane C, Arcand SL, Bourada V, et al. Small Cell Ovarian Carcinoma: Genomic Stability and Responsiveness to Therapeutics. Orphanet J Rare Dis, 2013, 8(1):33., DOI 10.1186/1750-1172-8-33 Otte A, Rauprich F, Hillemanns P, Park-Simon T-W, von der Ohe J, Hass R. In Vitro and in Vivotherapeutic Approach for a Small Cell Carcinoma of the Ovary Hypercalcaemic Type Using a SCCOHT-1 Cellular Model. Orphanet J Rare Dis, 2014, 9(1):126., DOI 10.1186/s13023-014-0126-4 Pressey JG, Kelly DR, Hawthorne HT. Successful Treatment of Preadolescents with Small Cell Carcinoma of the Ovary Hypercalcemic Type. J Ped Hematol Oncol, 2013, 35(7):566–9., DOI 10.1097/MPH.0b013e318282cca8 Qin Q, Ajewole VB, Sheu TG, Donohue R, Singh M. Successful Treatment of a Stage IIIC Small-Cell Carcinoma of the Ovary Hypercalcemic Subtype Using Multi-Modality Therapeutic Approach. ecancer, 2018, 12., DOI 10.3332/ ecancer.2018.832 Stephens B, Anthony SP, Han H, Kiefer J, Hostetter G, Barrett M, et al. Molecular Characterization of a Patient’s Small Cell Carcinoma of the Ovary of the Hypercalcemic Type. J Cancer, 2012, 3(1):58–66., DOI 10.7150/jca.3872 Henon C, Blay J-Y, Massard C, Mir O, Bahleda R, Dumont S, et al. Long Lasting Major Response to Pembrolizumab in a Thoracic Malignant Rhabdoid-like SMARCA4-Deficient Tumor. Ann Oncol, 2019, 30(8): 1401–3., DOI 10.1093/annonc/mdz160 Shen J, Ju Z, Zhao W, Wang L, Peng Y, Ge Z, et al. ARID1A Deficiency Promotes Mutability and Potentiates Therapeutic Antitumor Immunity Unleashed by Immune Checkpoint Blockade. Nat Med, 2018, 24(5): 556–62., DOI 10.1038/s41591-018-0012-z Okamura R, Kato S, Lee S, Jimenez RE, Sicklick JK, Kurzrock R. ARID1Aalterations Function as a Biomarker for Longer Progression-free Survival after Anti-PD-1/pd-L1 Immunotherapy. J Immunother Cancer, 2020, 8(1):e000438., DOI 10.1136/jitc-2019-000438 Auguste A, Blanc-Durand F, Deloger M, Le Formal A, Bareja R, Wilkes DC, et al. Small Cell Carcinoma of the Ovary, Hypercalcemic Type, SCCOHT, beyond SMARCA4 Mutations: A Comprehensive Genomic Analysis. Cells, 2020, 9(6):1496., DOI 10.3390/cells9061496 Kim KH, Kim W, Howard TP, Vazquez F, Tsherniak A, Wu JN, et al. SWI/ SNF-mutant Cancers Depend on Catalytic and Non-catalytic Activity of EZH2. Nat Med, 2015, 21(12):1491–6., DOI 10.1038/nm.3968 Kurmasheva RT, Sammons M, Favours E, Wu J, Kurmashev D, Cosmopoulos K, et al. Initial Testing, Stage 1, of Tazemetostat, EPZ-6438), a Novel EZH2 Inhibitor, by the Pediatric Preclinical Testing Program. Pediatr Blood Cancer, 2017, 64(3):e26218., DOI 10.1002/pbc.26218 Chan-Penebre E, Armstrong K, Drew A, Grassian AR, Feldman I, Knutson SK, et al. Selective Killing of SMARCA2- and SMARCA4-Deficient Small Cell Carcinoma of the Ovary, Hypercalcemic Type Cells by Inhibition of EZH2: In Vitro and In Vivo Preclinical Models. Mol Cancer Ther, 2017, 16(5):850–60., DOI 10.1158/1535-7163.MCT-16-0678 Wong JP, Todd JR, Finetti MA, McCarthy F, Broncel M, Vyse S, et al. Dual Targeting of PDGFRα and FGFR1 Displays Synergistic Efficacy in Malignant Rhabdoid Tumors. Cel Rep, 2016, 17(5):1265–75., DOI 10.1016/ j.celrep.2016.10.005 Lang JD, Hendricks WPD, Orlando KA, Yin H, Kiefer J, Ramos P, et al. Ponatinib Shows Potent Antitumor Activity in Small Cell Carcinoma of the Ovary Hypercalcemic Type, SCCOHT, through Multikinase Inhibition. Clin Cancer Res, 2018, 24(8):1932–43., DOI 10.1158/1078-0432.CCR-17-1928 Xue Y, Meehan B, Macdonald E, Venneti S, Wang XQD, Witkowski L, et al. CDK4/6 Inhibitors Target SMARCA4-Determined Cyclin D1 Deficiency in Hypercalcemic Small Cell Carcinoma of the Ovary. Nat Commun, 2019, 10(1): 558., DOI 10.1038/s41467-018-06958-9 Callegaro-Filho D, Burke TW, Eifel PJ, Ramirez PT, Euscher EE, Schmeler KM. Radiotherapy for Recurrent Small Cell Carcinoma of the Ovary: A Case Report and Review of the Literature. Gynecol Oncol Rep, 2015, 11:23–5., DOI 10.1016/ j.gore.2014.12.003 Kolin DL, Dong F, Baltay M, Lindeman N, MacConaill L, Nucci MR, et al. SMARCA4-deficient Undifferentiated Uterine Sarcoma, Malignant Rhabdoid Tumor of the Uterus): a Clinicopathologic Entity Distinct from Undifferentiated Carcinoma. Mod Pathol, 2018, 31:1442–56., DOI 10.1038/ s41379-018-0049-z Lin DI, Allen JM, Hecht JL, Killian JK, Ngo NT, Edgerly C, et al. SMARCA4 Inactivation Defines a Subset of Undifferentiated Uterine Sarcomas with Rhabdoid and Small Cell Features and Germline Mutation Association. Mod Pathol, 2019, 32:1675–87., DOI 10.1038/s41379-019-0303-z Connor YD, Miao D, Lin DI, Hayne C, Howitt BE, Dalrymple JL, et al. Germline Mutations of SMARCA4 in Small Cell Carcinoma of the Ovary, Hypercalcemic Type and in SMARCA4-Deficient Undifferentiated Uterine Sarcoma: Clinical Features of a Single Family and Comparison of Large Cohorts. Gynecol Oncol, 2020, 157:106–14., DOI 10.1016/ j.ygyno.2019.10.031 Kolin DL, Quick CM, Dong F, Fletcher CDM, Stewart CJR, Soma A, et al. SMARCA4-deficient Uterine Sarcoma and Undifferentiated Endometrial Carcinoma Are Distinct Clinicopathologic Entities. Am J Surg Pathol, 2020, 44:263–70., DOI 10.1097/PAS.0000000000001375 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1610003 EP 1610011 DI 10.3389/pore.2021.1610003 PG 9 ER PT J AU Bai, R Diao, B Li, K Xu, X Yang, P AF Bai, Rui Diao, Bowen Li, Kaili Xu, Xiaohan Yang, Ping TI Serum Tie-1 is a Valuable Marker for Predicting the Progression and Prognosis of Cervical Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE survival; cervical cancer; diagnostic biomarker; Tie-1; angiopoietin ID survival; cervical cancer; diagnostic biomarker; Tie-1; angiopoietin AB Objective: To investigate whether serum Tie-1 (sTie-1) is a valuable marker for predicting progression and prognosis of cervical cancer. Methods: Enzyme-linked immunosorbent assay (ELISA) was used to detect serum sTie-1 concentrations in 75 cervical cancer patients, 40 cervical intraepithelial neoplasia (CIN) patients, and 55 healthy controls without cervical lesions, and sTie-1 levels were compared between the groups. Receiver operating characteristic curves was used to evaluate the diagnostic value of sTie-1. The relationship between sTie-1 concentrations in patients with cervical cancer and clinicopathological features and prognosis were analyzed, and the risk factors for postoperative recurrence were determined using univariate and multivariable Cox proportional hazards regression. Results: We found that sTie-1 concentrations gradually increased according to lesion severity (i.e., cancer vs. CIN; p < 0.05) and were significantly elevated in adenocarcinoma compared with healthy controls. sTie-1 levels strongly distinguished between cervical cancer patients and the healthy controls (area under the curve = 0.846; cut-off value = 1,882.64 pg/ml; sensitivity = 74.6%; specificity = 96.4%). Moreover, sTie-1 levels in cervical cancer patients were significantly associated with tumor size, advanced tumor stage, lymph node metastasis, and reduced 4-years progression-free survival. Cervical cancer patients with high sTie-1 concentrations had a 3.123-fold [95% confidence interval (CI): 1.087–8.971, p = 0.034] higher risk for tumor recurrence. Conclusions: Elevated sTie-1 levels in patients with cervical carcinoma were associated with tumor progression and poor prognosis, indicating that sTie-1 may be a valuable marker for predicting progression and prognosis of cervical cancer. C1 [Bai, Rui] Shihezi University, School of Medicine, First Affiliated Hospital, Department of GynecologyShihezi, China. [Diao, Bowen] Shihezi University, School of Medicine, First Affiliated Hospital, Department of GynecologyShihezi, China. [Li, Kaili] Shihezi University, School of Medicine, First Affiliated Hospital, Department of GynecologyShihezi, China. [Xu, Xiaohan] Shihezi University, School of Medicine, First Affiliated Hospital, Department of GynecologyShihezi, China. [Yang, Ping] Shihezi University, School of Medicine, First Affiliated Hospital, Department of GynecologyShihezi, China. RP Yang, P (reprint author), Shihezi University, School of Medicine, First Affiliated Hospital, Department of Gynecology, Shihezi, China. EM pingy2018@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 68(6): 394–424., DOI 10.3322/caac.21492 Bao HL, Liu YN, Wang LJ, Fang LW, Cong S, Zhou MG, et al. Analysis on Mortality of Cervical Cancer and its Temporal Trend in Women in China, 2006-2012. Zhonghua Liu Xing Bing Xue Za Zhi, 2017, 38(1):58–64., DOI 10.3760/cma.j.issn.0254-6450.2017.01.011 Eklund L, Kangas J, Saharinen P. Angiopoietin-Tie Signalling in the Cardiovascular and Lymphatic Systems. Clin Sci, Lond,, 2017, 131(1): 87–103., DOI 10.1042/cs20160129 Saharinen P, Eklund L, Alitalo K. Therapeutic Targeting of the Angiopoietin- TIE Pathway. Nat Rev Drug Discov, 2017, 16(9):635–61., DOI 10.1038/ nrd.2016.278 Yang P, Chen N, Yang D, Crane J, Huang B, Dong R, et al. Cervical Cancer Cell-Derived Angiopoietins Promote Tumor Progression. Tumour Biol, 2017, 39(7):1010428317711658., DOI 10.1177/1010428317711658 Yang P, Chen N, Jia J-h., Gao X-j., Li S-h., Cai J, et al. Tie-1: A Potential Target for Anti-angiogenesis Therapy. J Huazhong Univ Sci Technol [Med Sci, 2015, 35(5):615–22., DOI 10.1007/s11596-015-1479-1 Korhonen EA, Lampinen A, Giri H, Anisimov A, Kim M, Allen B, et al. Tie1 Controls Angiopoietin Function in Vascular Remodeling and Inflammation. J Clin Invest, 2016, 126(9):3495–510., DOI 10.1172/jci84923 D’Amico G, Korhonen EA, Anisimov A, Zarkada G, Holopainen T, Hagerling R, et al. Tie1 Deletion Inhibits Tumor Growth and Improves Angiopoietin Antagonist Therapy. J Clin Invest, 2014, 124(2):824–34., DOI 10.1172/JCI68897 La Porta S, Roth L, Singhal M, Mogler C, Spegg C, Schieb B, et al. Endothelial Tie1-Mediated Angiogenesis and Vascular Abnormalization Promote Tumor Progression and Metastasis. J Clin Invest, 2018, 128(2):834–45., DOI 10.1172/ jci94674 Tiainen L, Korhonen EA, Leppanen V-M, LuukkaalaT, HamalainenM, TannerM, et al. High Baseline Tie1 Level Predicts Poor Survival in Metastatic Breast Cancer. BMC Cancer, 2019, 19(1):732., DOI 10.1186/s12885-019-5959-8 Torigata M, Yamakawa D, Takakura N. Elevated Expression of Tie1 Is Accompanied by Acquisition of Cancer Stemness Properties in Colorectal Cancer. Cancer Med, 2017, 6(6):1378–88., DOI 10.1002/ cam4.1072 Liu H, Ye X, Li D, Yao Q, Li Y. Incidence, Clinical Risk and Prognostic Factors for Liver Metastasis in Patients with Cervical Cancer: a Population-Based Retrospective Study. BMC Cancer, 2021, 21(1):421., DOI 10.1186/s12885-021-08127-6 Arbyn M, Weiderpass E, Bruni L, de Sanjose S, Saraiya M, Ferlay J, et al. Estimates of Incidence and Mortality of Cervical Cancer in 2018: a Worldwide Analysis. Lancet Glob Health, 20182020, 8(2):e191–e203., DOI 10.1016/S2214-109X(19)30482-6 Savant S, La Porta S, Budnik A, Busch K, Hu J, Tisch N, et al. The Orphan Receptor Tie1 Controls Angiogenesis and Vascular Remodeling by Differentially Regulating Tie2 in Tip and Stalk Cells. Cel Rep, 2015, 12(11):1761–73., DOI 10.1016/j.celrep.2015.08.024 Wu B, Cui J, Yang X-M, Liu Z-Y, Song F, Li L, et al. Cytoplasmic Fragment of CD147 Generated by Regulated Intramembrane Proteolysis Contributes to HCC by Promoting Autophagy. Cell Death Dis, 2017, 8(7):e2925., DOI 10.1038/cddis.2017.251 Yang P, Chen N, Yang D, Crane J, Yang S, Wang H, et al. The Ratio of Serum Angiopoietin-1 to Angiopoietin-2 in Patients with Cervical Cancer Is a Valuable Diagnostic and Prognostic Biomarker. PeerJ, 2017, 5:e3387., DOI 10.7717/peerj.3387 Ishibashi M, Toyoshima M, Zhang X, Hasegawa-Minato J, Shigeta S, Usui T, et al. Tyrosine Kinase Receptor TIE-1 Mediates Platinum Resistance by Promoting Nucleotide Excision Repair in Ovarian Cancer. Sci Rep, 2018, 8(1):13207., DOI 10.1038/s41598-018-31069-2 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1610006 EP 1610012 DI 10.3389/pore.2021.1610006 PG 7 ER PT J AU Hu, F Guo, L Yu, J Dai, D Xiong, Y He, Y Zhou, W AF Hu, Fangling Guo, Liang Yu, Jieqing Dai, Daofeng Xiong, Yuanping He, Yuanqiao Zhou, Wensheng TI Using Patient-Derived Xenografts to Explore the Efficacy of Treating Head-and-Neck Squamous Cell Carcinoma With Anlotinib SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE head-and-neck squamous cell carcinoma; drug sensitivity; patient-derived xenograft; anlotinib; tumor inhibition ID head-and-neck squamous cell carcinoma; drug sensitivity; patient-derived xenograft; anlotinib; tumor inhibition AB Objective: The efficacy of anlotinib as a treatment for head-and-neck squamous cell carcinoma (HNSCC) has been little explored. Here, we used patient-derived xenografts (PDXs) to this end. Methods: Fresh tumor tissues of HNSCC patients were screened in terms of in vitro drug sensitivity using the MTT assay. Patient PDXs were used to confirm the anti-tumor effects of anlotinib in vivo. After the medication regimen was complete, the tumor volume changes in mice were calculated. Apoptosis was measured using the TUNEL assay. The cell proliferation and apoptosis levels of PDXs yielded data on the utility of anlotinib treatment in vivo. Results: Anlotinib suppressed the in vitro proliferation of nine tumor tissues by an average of 51.05 ± 13.74%. Anlotinib also significantly inhibited the growth of three PDXs in mice (tumor growth inhibition 79.02%). The expression levels of Ki-67 and proliferating cell nuclear antigen after anlotinib treatment were significantly lower than those in the controls. The negative and positive controls exhibited no and some apoptosis, respectively, whereas the anlotinib group evidenced extensive apoptosis. Conclusion: Anlotinib suppressed HNSCC growth in vitro and in vivo (by inhibiting cell proliferation and promoting apoptosis), suggesting that anlotinib can potentially treat HNSCC. C1 [Hu, Fangling] Nanchang University, First Affiliated Hospital, Department of Otolaryngology-Head and Neck SurgeryNanchang, China. [Guo, Liang] Nanchang University, First Affiliated Hospital, Department of Otolaryngology-Head and Neck SurgeryNanchang, China. [Yu, Jieqing] Jiangxi Institute of Otorhinolaryngology-Head and Neck SurgeryNanchang, China. [Dai, Daofeng] Nanchang University, First Affiliated Hospital, Department of Otolaryngology-Head and Neck SurgeryNanchang, China. [Xiong, Yuanping] Nanchang University, First Affiliated Hospital, Department of Otolaryngology-Head and Neck SurgeryNanchang, China. [He, Yuanqiao] Nanchang University, Laboratory Animal Science CenterNanchang, China. [Zhou, Wensheng] Nanchang University, First Affiliated Hospital, Department of Otolaryngology-Head and Neck SurgeryNanchang, China. RP Zhou, W (reprint author), Nanchang University, First Affiliated Hospital, Department of Otolaryngology-Head and Neck Surgery, Nanchang, China. EM zhouwesh@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: a Cancer J clinicians, 2018, 68(6): 394–424., DOI 10.3322/caac.21492 Vigneswaran N, Williams MD. Epidemiologic Trends in Head and Neck Cancer and Aids in Diagnosis. Oral Maxill Surg Clin North America, 2014, 26(2):123–41., DOI 10.1016/j.coms.2014.01.001 Bray I, Brennan P, Boffetta P. Projections of Alcohol- and Tobacco- Related Cancer Mortality in Central Europe. Int J Cancer, 2000, 87(1): 122–8., DOI 10.1002/1097-0215(20000701)87:1<122:aid-ijc18>3.0.co; 2-w Ghantous Y, Schussel JL, Brait M. Tobacco and Alcohol-Induced Epigenetic Changes in Oral Carcinoma. Curr Opin Oncol, 2018, 30(3):152–8., DOI 10.1097/CCO.0000000000000444 Tribius S, Wurdemann N, Laban S, Hoffmann TK, Sharma SJ, Klussmann JP. Update zu HPV-assoziierten Kopf-Hals-Karzinomen - Highlights der ASCOJahrestagung 2019. Hno, 2019, 67(12):912–7., DOI 10.1007/s00106-019- 00766-3 Allen CT, Law JH, Dunn GP, Uppaluri R. Emerging Insights into Head and Neck Cancer Metastasis. Head Neck, 2013, 35(11):1669–78., DOI 10.1002/ hed.23202 Pignon J-P, Maitre Al., Maillard E, Bourhis J, Group M-NC. Meta-analysis of Chemotherapy in Head and Neck Cancer, MACH-NC): an Update on 93 Randomised Trials and 17,346 Patients. Radiother Oncol, 2009, 92(1):4–14., DOI 10.1016/j.radonc.2009.04.014 Taurin S, Yang C-H, Reyes M, Cho S, Coombs DM, Jarboe EA, et al. Endometrial Cancers Harboring Mutated Fibroblast Growth Factor Receptor 2 Protein Are Successfully Treated with a New Small Tyrosine Kinase Inhibitor in an Orthotopic Mouse Model. Int J Gynecol Cancer, 2018, 28(1):152–60., DOI 10.1097/ igc.0000000000001129 Jayson GC, Kerbel R, Ellis LM, Harris AL. Antiangiogenic Therapy in Oncology: Current Status and Future Directions. The Lancet, 2016, 388(10043):518–29., DOI 10.1016/s0140-6736(15)01088-0 El-Kenawi AE, El-Remessy AB. Angiogenesis Inhibitors in Cancer Therapy: Mechanistic Perspective on Classification and Treatment Rationales. Br J Pharmacol, 2013, 170(4):712–29., DOI 10.1111/ bph.12344 Shweiki D, Itin A, Soffer D, Keshet E. Vascular Endothelial Growth Factor Induced by Hypoxia May Mediate Hypoxia-Initiated Angiogenesis. Nature, 1992, 359(6398):843–5., DOI 10.1038/359843a0 Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips HS, et al. Inhibition of Vascular Endothelial Growth Factor-Induced Angiogenesis Suppresses Tumour Growth In Vivo. Nature, 1993, 362(6423):841–4., DOI 10.1038/ 362841a0 Ferrara N, Gerber H-P, LeCouter J. The Biology of VEGF and its Receptors. Nat Med, 2003, 9(6):669–76., DOI 10.1038/nm0603-669 Wang C, Chen J, Cao W, Sun L, Sun H, Liu Y. Aurora-B and HDAC Synergistically Regulate Survival and Proliferation of Lymphoma Cell via AKT, mTOR and Notch Pathways. Eur J Pharmacol, 2016, 779:1–7., DOI 10.1016/j.ejphar.2015.11.049 Sun Y, Niu W, Du F, Du C, Li S, Wang J, et al. Safety, Pharmacokinetics, and Antitumor Properties of Anlotinib, an Oral Multi-Target Tyrosine Kinase Inhibitor, in Patients with Advanced Refractory Solid Tumors. J Hematol Oncol, 2016, 9(1):105., DOI 10.1186/s13045-016-0332-8 Xie C,Wan X, Quan H, Zheng M, Fu L, Li Y, et al. Preclinical Characterization of Anlotinib, a Highly Potent and Selective Vascular Endothelial Growth Factor Receptor-2 Inhibitor. Cancer Sci, 2018, 109(4):1207–19., DOI 10.1111/ cas.13536 Lin B, Song X, Yang D, Bai D, Yao Y, Lu N. Anlotinib Inhibits Angiogenesis via Suppressing the Activation of VEGFR2, PDGFRβ and FGFR1. Gene, 2018, 654:77–86., DOI 10.1016/j.gene.2018.02.026 Syed YY. Anlotinib: First Global Approval. Drugs, 2018, 78(10):1057–62., DOI 10.1007/s40265-018-0939-x Karamboulas C, Meens J, Ailles L. Establishment and Use of Patient- Derived Xenograft Models for Drug Testing in Head and Neck Squamous Cell Carcinoma. STAR Protoc, 2020, 1(1):100024., DOI 10.1016/ j.xpro.2020.100024 Loo DT. TUNEL Assay: An Overview of Techniques. Methods Mol Biol, 2002, 203:21–30., DOI 10.1385/1-59259-179-5:21 Lang L, Xiong Y, Prieto-Dominguez N, Loveless R, Jensen C, Shay C, et al. FGF19/FGFR4 Signaling axis Confines and Switches the Role of Melatonin in Head and Neck Cancer Metastasis. J Exp Clin Cancer Res, 2021, 40(1):93., DOI 10.1186/s13046-021-01888-9 Huang L, David O, Cabay RJ, Valyi-Nagy K, Macias V, Zhong R, et al. Molecular Classification of Lymph Node Metastases Subtypes Predict for Survival in Head and Neck Cancer. Clin Cancer Res, 2019, 25(6):1795–808., DOI 10.1158/1078-0432.CCR-18-1884 Ramjiawan RR, Griffioen AW, Duda DG. Anti-angiogenesis for Cancer Revisited: Is There a Role for Combinations with Immunotherapy?. Angiogenesis, 2017, 20(2):185–204., DOI 10.1007/s10456-017-9552-y Tian S, Quan H, Xie C, Guo H, Lu F, Xu Y, et al. YN968D1 Is a Novel and Selective Inhibitor of Vascular Endothelial Growth Factor Receptor-2 Tyrosine Kinase with Potent Activity In Vitro and In Vivo. Cancer Sci, 2011, 102(7):1374–80., DOI 10.1111/j.1349-7006.2011.01939.x Shen G, Zheng F, Ren D, Du F, Dong Q, Wang Z, et al. Anlotinib: a Novel Multi-Targeting Tyrosine Kinase Inhibitor in Clinical Development. J Hematol Oncol, 2018, 11(1):120., DOI 10.1186/s13045-018-0664-7 Lin B, Song X, Yang D, Bai D, Yao Y, Lu N. Anlotinib Inhibits Angiogenesis via Suppressing the Activation of VEGFR2, PDGFRβ and FGFR1. Gene, 2018, 654:77–86., DOI 10.1016/j.gene.2018.02.026 Gao L, Lang L, Zhao X, Shay C, Shull AY, Teng Y. FGF19 Amplification Reveals an Oncogenic Dependency upon Autocrine FGF19/FGFR4 Signaling in Head and Neck Squamous Cell Carcinoma. Oncogene, 2019, 38(13): 2394–404., DOI 10.1038/s41388-018-0591-7 Cohen EE, Davis DW, Karrison TG, Seiwert TY, Wong SJ, Nattam S, et al. Erlotinib and Bevacizumab in Patients with Recurrent or Metastatic Squamous-Cell Carcinoma of the Head and Neck: a Phase I/II Study. Lancet Oncol, 2009, 10(3):247–57., DOI 10.1016/s1470- 2045(09)70002-6 Folkman J. Antiangiogenesis in Cancer Therapy-Endostatin and its Mechanisms of Action. Exp Cel Res, 2006, 312(5):594–607., DOI 10.1016/ j.yexcr.2005.11.015 Li J, Qin S, Xu J, Xiong J, Wu C, Bai Y, et al. Randomized, Double-Blind, Placebo-Controlled Phase III Trial of Apatinib in Patients with Chemotherapy-Refractory Advanced or Metastatic Adenocarcinoma of the Stomach or Gastroesophageal Junction. Jco, 2016, 34(13):1448–54., DOI 10.1200/jco.2015.63.5995 Olsson A-K, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF Receptor Signalling ? in Control of Vascular Function. Nat Rev Mol Cel Biol, 2006, 7(5):359–71., DOI 10.1038/nrm1911 Siveen KS, Prabhu K, Krishnankutty R, Kuttikrishnan S, Tsakou M, Alali FQ, et al. Vascular Endothelial Growth Factor, VEGF, Signaling in Tumour Vascularization: Potential and Challenges. Curr Vasc Pharmacol, 2017, 15(4): 339–51., DOI 10.2174/1570161115666170105124038 Liang L, Hui K, Hu C,Wen Y, Yang S, Zhu P, et al. Autophagy Inhibition Potentiates the Anti-angiogenic Property of Multikinase Inhibitor Anlotinib through JAK2/STAT3/VEGFA Signaling in Non-small Cell Lung Cancer Cells. J Exp Clin Cancer Res, 2019, 38(1):71., DOI 10.1186/ s13046-019-1093-3 Wang G, Sun M, Jiang Y, Zhang T, Sun W, Wang H, et al. Anlotinib, a Novel Small Molecular Tyrosine Kinase Inhibitor, Suppresses Growth and Metastasis via Dual Blockade of VEGFR2 and MET in Osteosarcoma. Int J Cancer, 2019, 145(4):979–93., DOI 10.1002/ijc.32180 Ruan X, Shi X, Dong Q, Yu Y, Hou X, Song X, et al. Antitumor Effects of Anlotinib in Thyroid Cancer. Endocrine-related cancer, 2019, 26(1):153–64., DOI 10.1530/erc-17-0558 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1610008 EP 1610016 DI 10.3389/pore.2021.1610008 PG 9 ER PT J AU Kobayashi, Y Kitazono, I Akahane, T Yanazume, Sh Kamio, M Togami, Sh Nohara, S Sakamoto, I Yokoyama, S Tabata, K Kobayashi, H Tanimoto, A AF Kobayashi, Yusuke Kitazono, Ikumi Akahane, Toshiaki Yanazume, Shintaro Kamio, Masaki Togami, Shinichi Nohara, Sachio Sakamoto, Ippei Yokoyama, Seiya Tabata, Kazuhiro Kobayashi, Hiroaki Tanimoto, Akihide TI Molecular Evaluation of Endometrial Dedifferentiated Carcinoma, Endometrioid Carcinoma, Carcinosarcoma, and Serous Carcinoma Using a Custom-Made Small Cancer Panel SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE microsatellite instability; tumor mutation burden; integrated molecular diagnosis; endometrial cancers; solid proliferation ID microsatellite instability; tumor mutation burden; integrated molecular diagnosis; endometrial cancers; solid proliferation AB It is often difficult to histologically differentiate among endometrial dedifferentiated carcinoma (DC), endometrioid carcinoma (EC), serous carcinoma (SC), and carcinosarcoma (CS) due to the presence of solid components. In this study, we aimed to categorize these carcinomas according to The Cancer Genome Atlas (TCGA) classification using a small custom-made cancer genome panel (56 genes and 17 microsatellite regions) for integrated molecular diagnosis. A total of 36 endometrial cancer cases with solid components were assessed using IHC, next-generation sequencing (NGS), and the custom-made panel. Among 19 EC cases, six were categorized as MMR-deficient (MMR-d) and eight were classified as having a nonspecific molecular profile. Three EC cases were classified as POLE mutation (POLEmut)-type, which had a very high tumor mutation burden (TMB) and low microsatellite instability (MSI). Increased TMB and MSI were observed in all three DC cases, classified as MMR-d with mutations in MLH1 and POLD1. Except for one case classified as MMR-d, all SC cases exhibited TP53 mutations and were classified as p53 mutation-type. SC cases also exhibited amplification of CCND1, CCNE1, and MYC. CS cases were classified as three TCGA types other than the POLEmut-type. The IHC results for p53 and ARID1A were almost consistent with their mutation status. NGS analysis using a small panel enables categorization of endometrial cancers with solid proliferation according to TCGA classification. As TCGA molecular classification does not consider histological findings, an integrated analytical procedure including IHC and NGS may be a practical diagnostic tool for endometrial cancers. C1 [Kobayashi, Yusuke] Course of Advanced Cancer Medicine for Gynecologic CancerKagoshima, Japan. [Kitazono, Ikumi] Kagoshima University Graduate School of Medical and Dental Sciences, Department of PathologyKagoshima, Japan. [Akahane, Toshiaki] Kagoshima University Graduate School of Medical and Dental Sciences, Department of PathologyKagoshima, Japan. [Yanazume, Shintaro] Kagoshima University, Field of Oncology, Faculty of Medicine, Department of Obstetrics and Gynecology Department of Molecular and Cellular PathologyKagoshima, Japan. [Kamio, Masaki] Kagoshima University, Field of Oncology, Faculty of Medicine, Department of Obstetrics and Gynecology Department of Molecular and Cellular PathologyKagoshima, Japan. [Togami, Shinichi] Kagoshima University, Field of Oncology, Faculty of Medicine, Department of Obstetrics and Gynecology Department of Molecular and Cellular PathologyKagoshima, Japan. [Nohara, Sachio] Mitsubishi Space Software, Department of Biomedical InformaticsAmagasaki, Japan. [Sakamoto, Ippei] Mitsubishi Space Software, Department of Biomedical InformaticsAmagasaki, Japan. [Yokoyama, Seiya] Kagoshima University Graduate School of Medical and Dental Sciences, Department of PathologyKagoshima, Japan. [Tabata, Kazuhiro] Kagoshima University Graduate School of Medical and Dental Sciences, Department of PathologyKagoshima, Japan. [Kobayashi, Hiroaki] Course of Advanced Cancer Medicine for Gynecologic CancerKagoshima, Japan. [Tanimoto, Akihide] Kagoshima University Graduate School of Medical and Dental Sciences, Department of PathologyKagoshima, Japan. RP Tanimoto, A (reprint author), Kagoshima University Graduate School of Medical and Dental Sciences, Department of Pathology, Kagoshima, Japan. EM akit09@m3.kufm.kagoshima-u.ac.jp CR Kim K-R, Lax SF, Lazar AJ, Longacre TA, Malpica A, Matias-Guiu X, et al. Tumours of Uterine Corpus. In: The WHO Classification of Tumours Editorial boardFemale Genital Tumours. 5th ed. Lyon: IARC, 2020). p. 245–308. Azumi N, Battifora H. The Distribution of Vimentin and Keratin in Epithelial and Nonepithelial Neoplasms: A Comprehensive Immunohistochemical Formalin- and Alcohol-Fixed Tumors. Am J Clin Pathol, 1987, 88:286–96., DOI 10.1093/ajcp/88.3.286 Murali R, Davidson B, Fadare O, Carlson JA, Crum CP, Gilks CB, et al. Highgrade Endometrial Carcinomas. Int J Gynecol Pathol, 2019, 38(Suppl. 1): S40–S63., DOI 10.1097/pgp.0000000000000491 Piulats JM, Guerra E, Gil-Martin M, Roman-Canal B, Gatius S, Sanz- Pamplona R, et al. Molecular Approaches for Classifying Endometrial Carcinoma. Gynecol Oncol, 2017, 145:200–7., DOI 10.1016/ j.ygyno.2016.12.015 Bell DW, Ellenson LH. Molecular Genetics of Endometrial Carcinoma. Annu Rev Pathol Mech Dis, 2019, 14:339–67., DOI 10.1146/annurev-pathol-020117- 043609 Carlson J, McCluggage WG. Reclassifying Endometrial Carcinomas with a Combined Morphological and Molecular Approach. Curr Opin Oncol, 2019, 31:411–9., DOI 10.1097/cco.0000000000000560 Horak P, Frohling S, Glimm H. Integrating Next-Generation Sequencing into Clinical Oncology: Strategies, Promises and Pitfalls. ESMO Open, 2016, 1: e000094., DOI 10.1136/esmoopen-2016-000094 Chang Y-S, Huang H-D, Yeh K-T, Chang J-G. Identification of Novel Mutations in Endometrial Cancer Patients by Whole-Exome Sequencing. Int J Mol Med, 2017, 50:1778–84., DOI 10.3892/ijo.2017.3919 Akahane T, Yamaguchi T, Kato Y, Yokoyama S, Hamada T, Nishida Y, et al. Comprehensive Validation of Liquid-Based Cytology Specimens for Next- Generation Sequencing in Cancer Genome Analysis. PLoS One, 2019, 14: e0217724., DOI 10.1371/journal.pone.0217724 Yamaguchi T, Akahane T, Harada O, Kato Y, Aimono E, Takei H, et al. Nextgeneration Sequencing in Residual Liquid-based Cytology Specimens for Cancer Genome Analysis. Diagn Cytopathology, 2020, 48:965–71., DOI 10.1002/dc.24511 Akahane T, Kitazono I, Yanazume S, Kamio M, Togami S, Sakamoto I, et al. Next-generation Sequencing Analysis of Endometrial Screening Liquid-Based Cytology Specimens: a Comparative Study to Tissue Specimens. BMC Med Genomics, 2020, 13:101., DOI 10.1186/s12920-020-00753-6 Tafe LJ, Garg K, Chew I, Tornos C, Soslow RA. Endometrial and Ovarian Carcinomas with Undifferentiated Components: Clinically Aggressive and Frequently Underrecognized Neoplasms. Mod Pathol, 2010, 23:781–9., DOI 10.1038/modpathol.2010.41 Vita G, Borgia L, Di Giovannantonio L, Bisceglia M. Dedifferentiated Endometrioid Adenocarcinoma of the Uterus. Int J Surg Pathol, 2011, 19: 649–52., DOI 10.1177/1066896911405987 Raffone A, Travaglino A, Cerbone M, Gencarelli A, mollo A, Insabato L, et al. Diagnostic Accuracy of Immunohistochemistry for Mismatch Repair Proteins as Surrogate of Microsatellite Instability Molecular Testing in Endometrial Cancer. Pathol Oncol Res, 2020, 26:1417–27., DOI 10.1007/ s12253-020-00811-5 Xu C, Gu X, Padmanabhan R,Wu Z, Peng Q, DiCarlo J, et al. smCounter2: an Accurate Low-Frequency Variant Caller for Targeted Sequencing Data with Unique Molecular Identifiers. Bioinformatics, 2019, 35:1299–309., DOI 10.1093/bioinformatics/bty790 Saotome K, Chiyoda T, Aimono E, Nakamura K, Tanishima S, Nohara S, et al. Clinical Implications of Next-generation Sequencing-based Panel Tests for Malignant Ovarian Tumors. Cancer Med, 2020, 9:7407–17., DOI 10.1002/ cam4.3383 Galuppini F, Dal Pozzo CA, Deckert J, Loupakis F, Fassan M, Baffa R. Tumor Mutation burden: from Comprehensive Mutational Screening to the Clinic. Cancer Cel Int, 2019, 19:209., DOI 10.1186/s12935-019-0929-4 Yarchoan M, Hopkins A, Jaffee EM. Tumor Mutational burden and Response Rate to PD-1 Inhibition. N Engl J Med, 2017, 377:2500–1., DOI 10.1056/ nejmc1713444 Niu B, Ye K, Zhang Q, Lu C, Xie M, McLellan MD, et al. MSIsensor: Microsatellite Instability Detection Using Paired Tumor-normal Sequence Data. Bioinformatics, 2014, 30:1015–6., DOI 10.1093/bioinformatics/btt755 Johansen AFB, Kassentoft CG, Knudsen M, Laursen MB, Madsen AH, Iversen LH, et al. Validation of Computational Determination of Microsatellite Status Using Whole Exome Sequencing Data from Colorectal Cancer Patients. BMC Cancer, 2019, 19:971., DOI 10.1186/s12885-019-6227-7 Djordjevic B, Hennessy BT, Li J, Barkoh BA, Luthra R, Mills GB, et al. Clinical Assessment of PTEN Loss in Endometrial Carcinoma: Immunohistochemistry Outperforms Gene Sequencing. Mod Pathol, 2012, 25:699–708., DOI 10.1038/ modpathol.2011.208 Khalique S, Naidoo K, Attygalle AD, Kriplani D, Daley F, Lowe A, et al. Optimised ARID1A Immunohistochemistry Is an Accurate Predictor ofARID1Amutational Status in Gynaecological Cancers. J Path: Clin Res, 2018, 4:154–66., DOI 10.1002/cjp2.103 Cancer Genome Atlas Research NetworkKandoth C, Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated Genomic Characterization of Endometrial Carcinoma. Nature, 2013, 497:67–73., DOI 10.1038/ nature12113 Jenkins TM, Hanley KZ, Schwartz LE, Cantrell LA, Stoler MH, Mills AM. Mismatch Repair Deficiency in Uterine Carcinosarcoma. Am J Surg Pathol, 2020, 44:782–92., DOI 10.1097/pas.0000000000001434 Garg K, Shih K, Barakat R, Zhou Q, Iasonos A, Soslow RA. Endometrial Carcinomas in Women Aged 40 Years and Younger: Tumors Associated with Loss of DNA Mismatch Repair Proteins Comprise a Distinct Clinicopathologic Subset. Am J Surg Pathol, 2009, 33:1869–77., DOI 10.1097/ pas.0b013e3181bc9866 Han G, Sidhu D, Duggan MA, Arseneau J, Cesari M, Clement PB, et al. Reproducibility of Histological Cell Type in High-Grade Endometrial Carcinoma. Mod Pathol, 2013, 26:1594–604., DOI 10.1038/ modpathol.2013.102 McConechy MK, Ding J, Cheang MC, Wiegand KC, Senz J, Tone AA, et al. Use of Mutation Profiles to Refine the Classification of Endometrial Carcinomas. J Pathol, 2012, 228:20–30., DOI 10.1002/path.4056 Cuevas D, Valls J, Gatius S, Roman-Canal B, Estaran E, Dorca E, et al. Targeted Sequencing with a Customized Panel to Assess Histological Typing in Endometrial Carcinoma. Virchows Arch, 2019, 474:585–98., DOI 10.1007/ s00428-018-02516-2 Jones NL, Xiu J, Chatterjee-Paer S, Buckley de Meritens A, Burke WM, Tergas AI, et al. Distinct Molecular Landscapes between Endometrioid and Nonendometrioid Uterine Carcinomas. Int J Cancer, 2017, 140:1396–404., DOI 10.1002/ijc.30537 Cherniack AD, Shen H, Walter V, Stewart C, Murray BA, Bowlby R, et al. Integrated Molecular Characterization of Uterine Carcinosarcoma. Cancer Cell, 2017, 31:411–23., DOI 10.1016/j.ccell.2017.02.010 Chen X, Arend R, Hamele-Bena D, Tergas AI, Hawver M, Tong G-X, et al. Uterine Carcinosarcomas. Int J Gynecol Pathol, 2017, 36:412–9., DOI 10.1097/ pgp.0000000000000346 Talhouk A, McConechy MK, Leung S, Li-Chang HH, Kwon JS, Melnyk N, et al. A Clinically Applicable Molecular-Based Classification for Endometrial Cancers. Br J Cancer, 2015, 113:299–310., DOI 10.1038/bjc.2015.190 Talhouk A, McConechy MK, Leung S, Yang W, Lum A, Senz J, et al. Confirmation of ProMisE: a Simple, Genomics-Based Clinical Classifier for Endometrial Cancer. Cancer, 2017, 123:802–13., DOI 10.1002/cncr.30496 Meyer LA, Broaddus RR, Lu KH. Endometrial Cancer and Lynch Syndrome: Clinical and Pathologic Considerations. Cancer Control, 2009, 16:14–22., DOI 10.1177/107327480901600103 Mills AM, Liou S, Ford JM, Berek JS, Pai RK, Longacre TA. Lynch Syndrome Screening Should Be Considered for All Patients with Newly Diagnosed Endometrial Cancer. Am J Surg Pathol, 2014, 38:1501–9., DOI 10.1097/ pas.0000000000000321 Hoang LN, Lee Y-S, Karnezis AN, Tessier-Cloutier B, Almandani N, Coatham M, et al. Immunophenotypic Features of Dedifferentiated Endometrial Carcinoma - Insights from BRG1/INI1-Deficient Tumours. Histopathology, 2016, 69:560–9., DOI 10.1111/his.12989 Kuhn E, Wu R-C, Guan B, Wu G, Zhang J, Wang Y, et al. Identification of Molecular Pathway Aberrations in Uterine Serous Carcinoma by Genomewide Analyses. J Natl Cancer Inst, 2012, 104:1503–13., DOI 10.1093/jnci/djs345 Leskela S, Perez-Mies B, Rosa-Rosa JM, Cristobal E, Biscuola M, Palacios- Berraquero ML, et al. Molecular Basis of Tumor Heterogeneity in Endometrial Carcinosarcoma. Cancers, 2019, 11:964., DOI 10.3390/cancers11070964 Cuevas D, Velasco A, Vaquero M, Santacana M, Gatius S, Eritja N, et al. Intratumour Heterogeneity in Endometrial Serous Carcinoma Assessed by Targeted Sequencing and Multiplex Ligation-dependent Probe Amplification: a Descriptive Study. Histopathology, 2020, 76:447–60., DOI 10.1111/his.14001 Saijo M, Nakamura K, Ida N, Nasu A, Yoshino T, Masuyama H, et al. Histologic Appearance and Immunohistochemistry of DNA Mismatch Repair Protein and P53 in Endometrial Carcinosarcoma. Am J Surg Pathol, 2019, 43:1493–500., DOI 10.1097/pas.0000000000001353 De Jong RA, Nijman HW, Wijbrandi TF, Reyners AK, Boezen HM, Hollema H. Molecular Markers and Clinical Behavior of Uterine Carcinosarcomas: Focus on the Epithelial Tumor Component. Mod Pathol, 2011, 24:1368–79., DOI 10.1038/modpathol.2011.88 Rosa-Rosa JM, Leskela S, Cristobal-Lana E, Santon A, Lopez-Garcia MA, Munoz G, et al. Molecular Genetic Heterogeneity in Undifferentiated Endometrial Carcinomas. Mod Pathol, 2016, 29:1390–8., DOI 10.1038/ modpathol.2016.132 Coatham M, Li X, Karnezis AN, Hoang LN, Tessier-Cloutier B, Meng B, et al. Concurrent ARID1A and ARID1B Inactivation in Endometrial and Ovarian Dedifferentiated Carcinomas. Mod Pathol, 2016, 29:1586–93., DOI 10.1038/ modpathol.2016.156 Zaino RJ, Kurman RJ, Diana KL, PaulMorrow C. The Utility of the Revised International Federation of Gynecology and Obstetrics Histologic Grading of Endometrial Adenocarcinoma Using a Defined Nuclear Grading System. A Gynecologic Oncology Group Study. Cancer, 1995, 75: 812–86., DOI 10.1002/1097-0142(19950101)75:1<81:aid-cncr2820750114> 3.0.co;2-f Leon-Castillo A, Gilvazquez E, Nout R, Smit VT, McAlpine JN, McConechy M, et al. Clinicopathological and Molecular Characterisation of ‘multiple-Classifier’ Endometrial Carcinomas. J Pathol, 2020, 250:312–22., DOI 10.1002/path.5373 Wang L, Piskorz A, Bosse T, Jimenez-Linan M, Rous B, Gilks CB, et al. Immunohistochemistry and Next-Generation Sequencing Are Complementary Tests in Identifying PTEN Abnormality in Endometrial Carcinoma Biopsies. Int J Gynecol Pathol, 2021)., DOI 10.1097/ pgp.0000000000000763 Tashiro H, Blazes MS, Wu R, Cho KR, Bose S, Wang SI, et al. Mutations in PTEN Are Frequent in Endometrial Carcinoma but Rare in Other Common Gynecological Malignancies. Cancer Res, 1997, 57:3935–40. Risinger JI, Hayes K, Maxwell GL, Carney ME, Dodge RK, Barrett JC, et al. PTEN Mutation in Endometrial Cancers Is Associated with Favorable Clinical and Pathologic Characteristics. Clin Cancer Res, 1998, 4:3005–10. Sun H, Enomoto T, Fujita M, Wada H, Yoshino K, Ozaki K, et al. Mutational Analysis of thePTENGene in Endometrial Carcinoma and Hyperplasia. Am J Clin Pathol, 2001, 115:32–8., DOI 10.1309/7jx6-b9u9-3p0r-eqny Rudd ML, Price JC, Fogoros S, Godwin AK, Sgroi DC, Merino MJ, et al. A Unique Spectrum of Somatic PIK3CA, P110α, Mutations within Primary Endometrial Carcinomas. Clin Cancer Res, 2011, 17:1331–40., DOI 10.1158/ 1078-0432.ccr-10-0540 Goebel EA, Vidal A, Matias-Guiu X, Blake Gilks C. The Evolution of Endometrial Carcinoma Classification through Application of Immunohistochemistry and Molecular Diagnostics: Past, Present and Future. Virchows Arch, 2018, 472:885–96., DOI 10.1007/s00428-017-2279-8 Maiques O, Santacana M, Valls J, Pallares J, Mirantes C, Gatius S, et al. Optimal Protocol for PTEN Immunostaining; Role of Analytical and Preanalytical Variables in PTEN Staining in normal and Neoplastic Endometrial, Breast, and Prostatic Tissues. Hum Pathol, 2014, 45:522–32., DOI 10.1016/ j.humpath.2013.10.018 Church JM. Polymerase Proofreading-Associated Polyposis. Dis Colon Rectum, 2014, 57:396–7., DOI 10.1097/dcr.0000000000000084 Wong A, Kuick CH, Wong WL, Tham JM, Mansor S, Loh E, et al. Mutation Spectrum of POLE and POLD1 Mutations in South East Asian Women Presenting with Grade 3 Endometrioid Endometrial Carcinomas. Gynecol Oncol, 2016, 141:113–20., DOI 10.1016/j.ygyno.2015.12.031 Cree IA, Malpica A, McCluggage WG. Neuroendocrine Neoplasia. In: The WHO Classification of Tumours Editorial boardFemale Genital Tumours. 5th ed. Lyon: IARC, 2020). p. 451–9. Espinosa I, De Leo A, D’Angelo E, Rosa-Rosa JM, CorominasM, Gonzalez A, et al. Dedifferentiated Endometrial Carcinomas with Neuroendocrine Features: a Clinicopathologic, Immunohistochemical, and Molecular Genetic Study. Hum Pathol, 2018, 72:100–6., DOI 10.1016/j.humpath.2017.11.006 Howitt BE, Dong F, Vivero M, Shah V, Lindeman N, Schoolmeester JK, et al. Molecular Characterization of Neuroendocrine Carcinomas of the Endometrium. Am J Surg Pathol, 2020, 44:1541–8., DOI 10.1097/ pas.0000000000001560 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1610013 EP 1610022 DI 10.3389/pore.2021.1610013 PG 10 ER PT J AU Refaat, B Zekri, J Aslam, A Ahmad, J Baghdadi, M Meliti, A Idris, Sh Sultan, S Alardati, H Saimeh, AH Alsaegh, A Alhadrami, M Hamid, T Naeem, M Elsamany, ASh AF Refaat, Bassem Zekri, Jamal Aslam, Akhmed Ahmad, Jawwad Baghdadi, A. Mohammed Meliti, Abdelrazak Idris, Shakir Sultan, Sufian Alardati, Hosam Saimeh, Akram Haitham Alsaegh, Aiman Alhadrami, Mai Hamid, Tahira Naeem, E. Mohammed Elsamany, Ahmed Shereef TI Profiling Activins and Follistatin in Colorectal Cancer According to Clinical Stage, Tumour Sidedness and Smad4 Status SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apoptosis; cell cycle; activin-A; activin-AB; activin-B; follistatin ID apoptosis; cell cycle; activin-A; activin-AB; activin-B; follistatin AB This study explored the roles of activins and follistatin in colorectal cancers. Paired malignant and normal colonic tissues were collected from archived paraffin-embedded (n = 90 patients) alongside fresh (n = 40 patients) specimen cohorts. Activin β-subunits, follistatin and Smad4 mRNAs and proteins were measured by real-time PCR and immunohistochemistry (IHC). Mature activin-A, -B, -AB and follistatin proteins were measured by ELISA. Cancer tissues having ≤ the 20th percentile of the Smad4 IHC score were considered as low (L-S4) group. The Smad4-intact SW480 and Smad4-null HT29 colon cancer cell lines were treated with activins and follistatin, and cell cycle was analysed by flow cytometry. The cell cycle inducing (CCND1/CCND3) and inhibitory (p21/ p27) proteins alongside the survival (survivin/BCL2) and pro-apoptosis (Casp-8/Casp-3) markers were measured by immunofluorescence. Thirty-nine patients had right-sided cancers (30%) and showed higher rates of L-S4 tumours (n = 17; 13.1%) alongside worse clinicopathological characteristics relative to left-sided cancers. The βA-subunit and activin-A increased, whilst βB-subunit and activin-AB decreased, in malignant sites and the late-stage cancers revealed the greatest abnormalities. Interestingly, follistatin declined markedly in early-stage malignant tissues, whilst increased significantly in the advanced stages. All activin molecules were comparable between the early stage right- and left-sided tumours, whereas the late-stage right-sided cancers and L-S4 tumours showed more profound deregulations. In vitro, activin-A increased the numbers of the SW480 cells in sub-G1 and G0/G1-phases, whereas reduced the HT29 cell numbers in the sub-G1 phase with simultaneous increases in the G0/G1 and S phases. The p21/p27/Casp-8/ Casp-3 proteins escalated, whilst CCND1/CCND3/BCL2/survivin declined in the SW480 cells following activin-A, whereas activin-A only promoted p21 and p27 alongside reduced CCND3 in the HT29 cells. By contrast, activin-AB increased the numbers of SW480 and HT29 cells in Sub-G1 and G0/G1-phases and promoted the anti-cancer and reduced the oncogenic proteins in both cell lines. In conclusion, activins and follistatin displayed stagedependent dysregulations and were markedly altered during the advanced stages of right-sided and L-S4 cancers. Moreover, the activin-A actions in CRC could be Smad4- dependent, whereas activin-AB may act as a Smad4-independent tumour suppressor protein. C1 [Refaat, Bassem] Umm Al-Qura University, Faculty of Applied Medical Sciences, Laboratory Medicine DepartmentMakkah, Saudi Arabia. [Zekri, Jamal] King Faisal Specialist Hospital and Research Centre, Oncology DepartmentJeddah, Saudi Arabia. [Aslam, Akhmed] Umm Al-Qura University, Faculty of Applied Medical Sciences, Laboratory Medicine DepartmentMakkah, Saudi Arabia. [Ahmad, Jawwad] Umm Al-Qura University, Faculty of Applied Medical Sciences, Laboratory Medicine DepartmentMakkah, Saudi Arabia. [Baghdadi, A. Mohammed] King Faisal Specialist Hospital and Research Centre, Research CentreJeddah, Saudi Arabia. [Meliti, Abdelrazak] King Faisal Specialist Hospital and Research Centre, Pathology DepartmentJeddah, Saudi Arabia. [Idris, Shakir] Umm Al-Qura University, Faculty of Applied Medical Sciences, Laboratory Medicine DepartmentMakkah, Saudi Arabia. [Sultan, Sufian] King Faisal Specialist Hospital and Research Centre, Department of SurgeryJeddah, Saudi Arabia. [Alardati, Hosam] King Faisal Specialist Hospital and Research Centre, Pathology DepartmentJeddah, Saudi Arabia. [Saimeh, Akram Haitham] King Faisal Specialist Hospital and Research Centre, Department of SurgeryJeddah, Saudi Arabia. [Alsaegh, Aiman] Umm Al-Qura University, Faculty of Applied Medical Sciences, Laboratory Medicine DepartmentMakkah, Saudi Arabia. [Alhadrami, Mai] Umm Al-Qura University, Faculty of Medicine, Pathology DepartmentMakkah, Saudi Arabia. [Hamid, Tahira] King Abdullah Medical City, Histopathology DepartmentMakkah, Saudi Arabia. [Naeem, E. Mohammed] King Abdullah Medical City, Histopathology DepartmentMakkah, Saudi Arabia. [Elsamany, Ahmed Shereef] King Abdullah Medical City, Oncology Centre, Medical Oncology DepartmentMakkah, Saudi Arabia. RP Refaat, B (reprint author), Umm Al-Qura University, Faculty of Applied Medical Sciences, Laboratory Medicine Department, Makkah, Saudi Arabia. EM bassem.refaat@yahoo.co.uk CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 68:394–424., DOI 10.3322/caac.21492 Testa U, Pelosi E, and Castelli G. Colorectal Cancer: Genetic Abnormalities, Tumor Progression, Tumor Heterogeneity, Clonal Evolution and Tumor- Initiating Cells. Med Sci, 2018, 6:31., DOI 10.3390/medsci6020031 Petrelli F, Tomasello G, Borgonovo K, Ghidini M, Turati L, Dallera P, et al. Prognostic Survival Associated with Left-Sided vs Right-Sided Colon Cancer. JAMA Oncol, 2017, 3:211–9., DOI 10.1001/jamaoncol.2016.4227 Narayanan S, Gabriel E, Attwood K, Boland P, and Nurkin S. Association of Clinicopathologic and Molecular Markers on Stage-specific Survival of Right versus Left Colon Cancer. Clin Colorectal Cancer, 2018, 17:e671–e678., DOI 10.1016/j.clcc.2018.07.001 Oyanagi H, Shimada Y, Nagahashi M, Ichikawa H, Tajima Y, Abe K, et al. SMAD4alteration Associates with Invasive-front Pathological Markers and Poor Prognosis in Colorectal Cancer. Histopathology, 2019, 74:873–82., DOI 10.1111/his.13805 Hsu Y-L, Lin C-C, Jiang J-K, Lin H-H, Lan Y-T, Wang H-S, et al. Clinicopathological and Molecular Differences in Colorectal Cancer According to Location. Int J Biol Markers, 2019, 34:47–53., DOI 10.1177/ 1724600818807164 Refaat B, El-Shemi AG, Mohamed AM, Kensara OA, Ahmad J, and Idris S. Activins and Their Related Proteins in colon Carcinogenesis: Insights from Early and Advanced Azoxymethane RatModels of colon Cancer. BMC Cancer, 2016, 16:879., DOI 10.1186/s12885-016-2914-9 Refaat B, and Bahathiq AO. The Performances of Serum Activins and Follistatin in the Diagnosis of Ectopic Pregnancy: A Prospective Case- Control Study. Clinica Chim Acta, 2020, 500:69–74., DOI 10.1016/ j.cca.2019.09.019 Refaat B, and Ledger W. The Expression of Activins, Their Type II Receptors and Follistatin in Human Fallopian Tube during the Menstrual Cycle and in Pseudo-pregnancy. Hum Reprod, 2011, 26:3346–54., DOI 10.1093/humrep/ der331 Bauer J, Ozden O, Akagi N, Carroll T, Principe DR, Staudacher JJ, et al. Activin and TGFβ Use Diverging Mitogenic Signaling in Advanced colon Cancer. Mol Cancer, 2015, 14:182., DOI 10.1186/s12943-015-0456-4 Tsuchida K, Nakatani M, Yamakawa N, Hashimoto O, Hasegawa Y, and Sugino H. Activin Isoforms Signal through Type I Receptor Serine/threonine Kinase ALK7. Mol Cell Endocrinol, 2004, 220:59–65., DOI 10.1016/ j.mce.2004.03.009 Bernard DJ, Lee KB, and Santos MM. Activin B Can Signal through Both ALK4 and ALK7 in Gonadotrope Cells. Reprod Biol Endocrinol, 2006, 4:52., DOI 10.1186/1477-7827-4-52 Loomans H, and Andl C. Intertwining of Activin A and TGFβ Signaling: Dual Roles in Cancer Progression and Cancer Cell Invasion. Cancers, 2014, 7: 70–91., DOI 10.3390/cancers7010070 Jung BH, Beck SE, Cabral J, Chau E, Cabrera BL, Fiorino A, et al. Activin Type 2 Receptor Restoration in MSI-H colon Cancer Suppresses Growth and Enhances Migration with Activin. Gastroenterology, 2007, 132:633–44., DOI 10.1053/j.gastro.2006.11.018 Deacu E, Mori Y, Sato F, Yin J, Olaru A, Sterian A, et al. Activin Type II Receptor Restoration in ACVR2-Deficient Colon Cancer Cells Induces Transforming Growth Factor-β Response Pathway Genes. Cancer Res, 2004, 64:7690–6., DOI 10.1158/0008-5472.CAN-04-2082 Zhang Y, Bao Y-L, Yang M-T, Wu Y, Yu C-L, Huang Y-X, et al. Activin A Induces SLC5A8 Expression through the Smad3 Signaling Pathway in Human colon Cancer RKO Cells. Int J Biochem Cel Biol, 2010, 42:1964–72., DOI 10.1016/j.biocel.2010.08.007 Wu S, Qi Y, Niu LM, Xie DX, Cui XL, and Liu ZH. Activin A as a Novel Biomarker for Colorectal Adenocarcinoma in Humans. Eur Rev Med Pharmacol Sci, 2015, 19:4371–8. Staudacher JJ, Bauer J, Jana A, Tian J, Carroll T, Mancinelli G, et al. Activin Signaling Is an Essential Component of the TGF-β Induced Pro-metastatic Phenotype in Colorectal Cancer. Sci Rep, 2017, 7:5569., DOI 10.1038/s41598- 017-05907-8 Loumaye A, de Barsy M, Nachit M, Lause P, van Maanen A, Trefois P, et al. Circulating Activin A Predicts Survival in Cancer Patients. J Cachexia, Sarcopenia Muscle, 2017, 8:768–77., DOI 10.1002/jcsm.12209 Li X, Yu W, Liang C, Xu Y, Zhang M, Ding X, et al. INHBA Is a Prognostic Predictor for Patients with colon Adenocarcinoma. BMC Cancer, 2020, 20: 305., DOI 10.1186/s12885-020-06743-2 Karagiannis GS, Treacy A, Messenger D, Grin A, Kirsch R, Riddell RH, et al. Expression Patterns of BoneMorphogenetic Protein Antagonists in Colorectal Cancer Desmoplastic Invasion Fronts. Mol Oncol, 2014, 8:1240–52., DOI 10.1016/j.molonc.2014.04.004 Refaat B, Abdelghany AH, BaSalamah MA, El-Boshy M, Ahmad J, and Idris S. Acute and Chronic Iron Overloading Differentially Modulates the Expression of Cellular Iron-Homeostatic Molecules in Normal Rat Kidney. J Histochem Cytochem, 2018, 66:825–39., DOI 10.1369/0022155418782696 Nguyen D, Zhou T, Shu J, and Mao J. Quantifying Chromogen Intensity in Immunohistochemistry via Reciprocal Intensity. Cancer InCytes, 2013, 2:e., DOI 10.1038/protex.2013.097 Lin Z, Zhang L, Zhou J, and Zheng J. Silencing Smad4 Attenuates Sensitivity of Colorectal Cancer Cells to Cetuximab by Promoting Epithelial-Mesenchymal Transition. Mol Med Rep, 2019, 20:3735–45., DOI 10.3892/mmr.2019.10597 Pholnukulkit P, Sonoyama K, and Kawabata J. Activin A Induces Phosphorylation of Smad2 but Not Complex Formation of Smad2 with Smad4 in Human colon Cancer Cell Line HT-29. Biosci Biotechnol Biochem, 2003, 67:2042–4., DOI 10.1271/bbb.67.2042 Aslam A, Ahmad J, Baghdadi MA, Idris S, Almaimani R, Alsaegh A, et al. Chemopreventive Effects of Vitamin D3 and its Analogue, Paricalcitol, in Combination with 5-fluorouracil against Colorectal Cancer: The Role of Calcium Signalling Molecules. Biochim Biophys Acta, Bba, - Mol Basis Dis, 2021, 1867:166040., DOI 10.1016/j.bbadis.2020.166040 Moser B, Hochreiter B, Herbst R, and Schmid JA. Fluorescence Colocalization Microscopy Analysis Can Be Improved by Combining Object-Recognition with Pixel-Intensity-Correlation. Biotechnol J, 2017, 12:1600332., DOI 10.1002/ biot.201600332 Mambelli-Lisboa N, Sciani JM, Brandao Prieto da Silva AR, and Kerkis I. Co- Localization of Crotamine with Internal Membranes and Accentuated Accumulation in Tumor Cells. Molecules, 2018, 23:968., DOI 10.3390/ molecules23040968 Aldiab A, Al Khayal KA, Al Obaid OA, Alsheikh A, Alsaleh K, Shahid M, et al. Clinicopathological Features and Predictive Factors for Colorectal Cancer Outcome in the Kingdom of Saudi Arabia. Oncology, 2017, 92:75–86., DOI 10.1159/000450857 Omer AAA. The Clinical and Pathological Features of Colorectal Cancer in Tabuk Region, Saudi Arabia: Trends in the Young and Elderly Patients. Int J Surg Surg Procedures, 2017, 2:122., DOI 10.15344/2456-4443/ 2017/122 Zhou S, Buckhaults P, Zawel L, Bunz F, Riggins G, Le Dai J, et al. Targeted Deletion of Smad4 Shows it Is Required for Transforming Growth Factor and Activin Signaling in Colorectal Cancer Cells. Proc Natl Acad Sci, 1998, 95: 2412–6., DOI 10.1073/pnas.95.5.2412 Bauer J, Sporn JC, Cabral J, Gomez J, and Jung B. Effects of Activin and TGFβ on P21 in Colon Cancer. PLoS One, 2012, 7:e39381., DOI 10.1371/ journal.pone.0039381 El-Deek SEM, Abd-Elghaffar SKH, Hna RS, Mohamed HG, and El-Deek HEM. Effect of Hesperidin against Induced Colon Cancer in Rats: Impact of Smad4 and Activin A Signaling Pathway. Nutr Cancer, 2021, 1–18., DOI 10.1080/01635581.2021.1907424 Zhang B, Zhang B, Chen X, Bae S, Singh K, Washington MK, et al. Loss of Smad4 in Colorectal Cancer Induces Resistance to 5-fluorouracil through Activating Akt Pathway. Br J Cancer, 2014, 110:946–57., DOI 10.1038/ bjc.2013.789 Zhao M, Mishra L, and Deng C-X. The Role of TGF-B/smad4 Signaling in Cancer. Int J Biol Sci, 2018, 14:111–23., DOI 10.7150/ijbs.23230 Zessner-Spitzenberg J, Thomas AL, Krett NL, and Jung B. TGFβ and Activin A in the Tumor Microenvironment in Colorectal Cancer. Gene Rep, 2019, 17: 100501., DOI 10.1016/j.genrep.2019.100501 Jueckstock J, Burkhardt N, Kuhn C, Blankenstein T, Mahner S, Schindlbeck C, et al. Expression of Activin during and after Chemotherapy in Peripheral Blood of Patients with Primary Breast Cancer. Anticancer Res, 2016, 36: 2153–9. Olsen OE, Hella H, Elsaadi S, Jacobi C, Martinez-Hackert E, and Holien T. Activins as Dual Specificity TGF-β Family Molecules: SMAD-Activation via Activin- and BMP-type 1 Receptors. Biomolecules, 2020, 10:519., DOI 10.3390/ biom10040519 Niimi S, HorikawaM, Seki T, Ariga T, Kobayashi T, and Hayakawa T. Effect of Activins AB and B on DNA Synthesis Stimulated by Epidermal Growth Factor in Primary Cultured Rat Hepatocytes. Biol Pharm Bull, 2002, 25:437–40., DOI 10.1248/bpb.25.437 Liu C, Yang Z, Li D, Liu Z, Miao X, Yang L, et al. Overexpression of B2M and Loss of ALK7 Expression Are Associated with Invasion, Metastasis, and Poor- Prognosis of the Pancreatic Ductal Adenocarcinoma. Cbm, 2015, 15:735–43., DOI 10.3233/cbm-150515 Hu T, Su F, Jiang W, and Dart DA. Overexpression of Activin Receptor-like Kinase 7 in Breast Cancer Cells Is Associated with Decreased Cell Growth and Adhesion. Ar, 2017, 37:3441–51., DOI 10.21873/anticanres.11712 Michael IP, Saghafinia S, Tichet M, Zangger N, Marinoni I, Perren A, et al. ALK7 Signaling Manifests a Homeostatic Tissue Barrier that Is Abrogated during Tumorigenesis and Metastasis. Dev Cel, 2019, 49:409–24., DOI 10.1016/ j.devcel.2019.04.015 Besson-Fournier C, Latour C, Kautz L, Bertrand J, Ganz T, Roth M-P, et al. Induction of Activin B by Inflammatory Stimuli Up-Regulates Expression of the Iron-Regulatory Peptide Hepcidin through Smad1/5/8 Signaling. Blood, 2012, 120:431–9., DOI 10.1182/blood-2012-02-411470 Canali S, Core AB, Zumbrennen-Bullough KB, Merkulova M, Wang C-Y, Schneyer AL, et al. Activin B Induces Noncanonical SMAD1/5/8 Signaling via BMP Type I Receptors in Hepatocytes: Evidence for a Role in Hepcidin Induction by Inflammation in Male Mice. Endocrinology, 2016, 157:1146–62., DOI 10.1210/en.2015-1747 Norgaard K, Muller C, Christensen N, Chiloeches ML,Madsen CL, Nielsen SS, et al. Loss of Mismatch Repair Signaling Impairs the WNT-Bone Morphogenetic Protein Crosstalk and the Colonic Homeostasis. J Mol Cel Biol, 2020, 12:410–23., DOI 10.1093/jmcb/mjz031 Karagiannis GS, Musrap N, Saraon P, Treacy A, Schaeffer DF, Kirsch R, et al. Bone Morphogenetic Protein Antagonist Gremlin-1 Regulates colon Cancer Progression. Biol Chem, 2015, 396:163–83., DOI 10.1515/hsz-2014- 0221 Shi L, Resaul J, Owen S, Ye L, and Jiang WG. Clinical and Therapeutic Implications of Follistatin in Solid Tumours. Cgp, 2016, 13:425–36., DOI 10.21873/cgp.20005 Karagiannis GS, Petraki C, Prassas I, Saraon P, Musrap N, Dimitromanolakis A, et al. Proteomic Signatures of the Desmoplastic Invasion Front Reveal Collagen Type XII as a Marker of Myofibroblastic Differentiation during Colorectal Cancer Metastasis. Oncotarget, 2012, 3:267–85., DOI 10.18632/ oncotarget.451 Dreikhausen L, Blank S, Sisic L, Heger U, Weichert W, Jager D, et al. Association of Angiogenic Factors with Prognosis in Esophageal Cancer. BMC Cancer, 2015, 15:121., DOI 10.1186/s12885-015-1120-5 Zhang P, Ruan Y, Xiao J, Chen F, and Zhang X. Association of Serum Follistatin Levels with Histological Types and Progression of Tumor in Human Lung Cancer. Cancer Cel Int, 2018, 18:162., DOI 10.1186/s12935- 018-0664-2 Yu M, Xiao L, Chen Y, Wang H, Gao Y, andWang A. Identification of a Potential Target for Treatment of Squamous Cell Carcinoma of the Tongue: Follistatin. Br J Oral Maxill Surg, 2020, 58:437–42., DOI 10.1016/j.bjoms.2020.01.028 Gold E, and Risbridger G. Activins and Activin Antagonists in the Prostate and Prostate Cancer. Mol Cell Endocrinol, 2012, 359:107–12., DOI 10.1016/ j.mce.2011.07.005 Sepporta MV, Tumminello FM, Flandina C, Crescimanno M, Giammanco M, La Guardia M, et al. Follistatin as Potential Therapeutic Target in Prostate Cancer. Targ Oncol, 2013, 8:215–23., DOI 10.1007/s11523-013-0268-7 Ottley E, and Gold E. Insensitivity to the Growth Inhibitory Effects of Activin A: An Acquired Capability in Prostate Cancer Progression. Cytokine Growth Factor Rev, 2012, 23:119–25., DOI 10.1016/j.cytogfr.2012.04.004 Antsiferova M, and Werner S. The Bright and the Dark Sides of Activin in Wound Healing and Cancer. J Cel Sci, 2012, 125:3929–37., DOI 10.1242/ jcs.094789 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1610032 EP 1610051 DI 10.3389/pore.2021.1610032 PG 20 ER PT J AU Turkevi-Nagy, S Bathori, Bocz, J Krenacs, L Cserni, G Kovari, B AF Turkevi-Nagy, Sandor Bathori, Agnes Bocz, Janos Krenacs, Laszlo Cserni, Gabor Kovari, Bence TI Syntaxin-1 and Insulinoma-Associated Protein 1 Expression in Breast Neoplasms with Neuroendocrine Features SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; syntaxin-1; insulinoma-associated protein 1; breast neuroendocrine neoplasms; solid papillary carcinoma; mucinous carcinoma; carcinoma no special type ID immunohistochemistry; syntaxin-1; insulinoma-associated protein 1; breast neuroendocrine neoplasms; solid papillary carcinoma; mucinous carcinoma; carcinoma no special type AB Introduction: A subset of breast neoplasia is characterized by features of neuroendocrine differentiation. Positivity for Neuroendocrine markers by immunohistochemistry is required for the diagnosis. Sensitivity and specificity of currently used markers are limited; based on the definitions of WHO Classification of Tumours, 5th edition, about 50% of breast tumors with features of neuroendocrine differentiation express chromogranin-A and 16% express synaptophysin. We assessed the applicability of two novel markers, syntaxin-1 and insulinoma-associated protein 1 (INSM1) in breast carcinomas. Methods: Hypercellular (Type B) mucinous carcinomas, solid papillary carcinomas, invasive carcinomas of no special type with neuroendocrine features and ductal carcinomas in situ of neuroendocrine subtype were included in our study. The immunohistochemical panel included chromogranin A, synaptophysin, CD56, syntaxin- 1 and INSM1. The specificity of syntaxin-1 and INSM1 was determined using samples negative for chromogranin A, synaptophysin and CD56. Results: The sensitivity of syntaxin-1 was 84.7% (50/59), with diffuse positivity in more than 60% of the cases. Syntaxin-1 also had an excellent specificity (98.1%). Depending on the definition for positivity, the sensitivity of INSM1 was 89.8% (53/59) or 86.4% (51/59), its specificity being 57.4% or 88.9%. The sensitivities of chromogranin A, synaptophysin and CD56 were 98.3, 74.6 and 22.4%, respectively. Discussion: Syntaxin-1 and INSM1 are sensitive and specific markers of breast tumors with neuroendocrine features, outperforming chromogranin A and CD56. We recommend syntaxin-1 and INSM1 to be included in the routine neuroendocrine immunohistochemical panel. C1 [Turkevi-Nagy, Sandor] University of Szeged, Department of PathologySzeged, Hungary. [Bathori, Agnes] University of Szeged, Department of PathologySzeged, Hungary. [Bocz, Janos] University of Szeged, Department of PathologySzeged, Hungary. [Krenacs, Laszlo] Bay Zoltan Foundation for Applied Research, Institute for Biotechnology, Laboratory of Tumor Pathology and Molecular DiagnosticsSzeged, Hungary. [Cserni, Gabor] University of Szeged, Department of PathologySzeged, Hungary. [Kovari, Bence] University of Szeged, Department of PathologySzeged, Hungary. RP Turkevi-Nagy, S (reprint author), University of Szeged, Department of Pathology, Szeged, Hungary. CR Feyrter F, and Hartmann G. On the Carcinoid Growth Form of the Carcinoma Mammae, Especially the Carcinoma Solidum, Gelatinosum)Mammae. Frankf Z Pathol, 1963, 73:24–39. Visscher DW, and Yasir S. Neuroendocrine Tumors of the Breast. Endocr Pathol, 2017, 28(2):121–7., DOI 10.1007/s12022-017-9477-4 Osamura RY, Matsui N, Okubo M, Chen L, and Field AS. Histopathology and Cytopathology of Neuroendocrine Tumors and Carcinomas of the Breast: A Review. Acta Cytol, 2019, 63(4):340–6., DOI 10.1159/000500705 Hejjane L, Oualla K, Bouchbika Z, Bourhafour M, Lhlou Mimi A, Boubacar E, et al. Primary Neuroendocrine Tumors of the Breast: Two Case Reports and Review of the Literature. J Med Case Rep, 2020, 14(1):41., DOI 10.1186/s13256- 020-02361-5 Ozdirik B, Kayser A, Ullrich A, Savic LJ, Reiss M, Tacke F, et al. Primary Neuroendocrine Neoplasms of the Breast: Case Series and Literature Review. Cancers, 2020, 12(3):733., DOI 10.3390/ cancers12030733 FA Tavassoli and P Devilee, editors. World Health Organization Classification of Tumours. Pathology and Genetics of Tumours of the Breast and Female Genital Organs. 3rd ed. Lyon: IARC Press, 2003). SR Lakhani, IO Ellis, SJ Schnitt, PH Tan, and MJ van de Vijver, editors. WHO Classification of Tumours of the Breast. 4th ed. Lyon: IARC Press, 2012). WHO Classification of Tumours Editorial Board. WHO Classification of Tumours. In: Breast Tumours. 5th ed. Lyon: IARC Press, 2019). Rindi G, Klimstra DS, Abedi-Ardekani B, Asa SL, Bosman FT, Brambilla E, et al. A Common Classification Framework for Neuroendocrine Neoplasms: an International Agency for Research on Cancer, IARC, and World Health Organization, WHO, Expert Consensus Proposal. Mod Pathol, 2018, 31(12): 1770–86., DOI 10.1038/s41379-018-0110-y Kovari B, Turkevi-Nagy S, Bathori A, Fekete Z, and Krenacs L. Syntaxin 1: A Novel Robust Immunophenotypic Marker of Neuroendocrine Tumors. Int J Mol Sci, 2020, 21(4):1213., DOI 10.3390/ijms21041213 Rosenbaum JN, Guo Z, Baus RM, Werner H, Rehrauer WM, and Lloyd RV. INSM1: A Novel Immunohistochemical and Molecular Marker for Neuroendocrine and Neuroepithelial Neoplasms. Am J Clin Pathol, 2015, 144(4):579–91., DOI 10.1309/AJCPGZWXXBSNL4VD Kovari B, Szasz AM, Kulka J, Marusic Z, Sarcevic B, Tiszlavicz Ls., et al. Evaluation of P40 as a Myoepithelial Marker in Different Breast Lesions. Pathobiology, 2015, 82(3-4):166–71., DOI 10.1159/ 000375127 Zombori T, and Cserni G. Immunohistochemical Analysis of the Expression of Breast Markers in Basal-like Breast Carcinomas Defined as Triple Negative Cancers Expressing Keratin 5. Pathol Oncol Res, 2018, 24(2):259–67., DOI 10.1007/s12253-017-0246-y Kovari B, Vranic S, Marchio C, Sapino A, and Cserni G. The Expression of GHRH and its Receptors in Breast Carcinomas with Apocrine Differentiation-Further Evidence of the Presence of a GHRH Pathway in These Tumors. Hum Pathol, 2017, 64:164–70., DOI 10.1016/ j.humpath.2017.03.026 Cserni G. Histological Type and Typing of Breast Carcinomas and the WHO Classification Changes over Time. Pathologica, 2020, 112(1):25–41., DOI 10.32074/1591-951X-1-20 Kawasaki T, and Kaira K. Insulinoma-associated Protein 1, INSM1, Expression in Breast Carcinomas with Neuroendocrine Morphologies: Application and Future Prospective. Virchows Arch, 2020, 479:191–4., DOI 10.1007/s00428-020-02935-0 Razvi H, Tsang JY, Poon IK, Chan S-K, Cheung S-Y, Shea K-H, et al. INSM1 Is a Novel Prognostic Neuroendocrine Marker for Luminal B Breast Cancer. Pathology, 2021, 53(2):170–8., DOI 10.1016/j.pathol.2020.07.004 Seijnhaeve E, Galant C, and Van Bockstal MR. Nuclear Insulinoma-Associated Protein 1 Expression as a Marker of Neuroendocrine Differentiation in Neoplasms of the Breast. Int J Surg Pathol, 2021, 29:496., DOI 10.1177/ 1066896920985938 Zombori T, Turkevi-Nagy S, Sejben A, Juhasz-Nagy G, Cserni G, Furak J, et al. The Panel of Syntaxin 1 and Insulinoma-associated Protein 1 Outperforms Classic Neuroendocrine Markers in Pulmonary Neuroendocrine Neoplasms. Apmis, 2021, 129:186–94., DOI 10.1111/apm.13113 Kudo N, Takano J, Kudoh S, Arima N, and Ito T. INSM1 Immunostaining in Solid Papillary Carcinoma of the Breast. Pathol Int, 2021, 71(1):51–9., DOI 10.1111/pin.13043 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1610039 EP 1610045 DI 10.3389/pore.2021.1610039 PG 7 ER PT J AU Kiss, Z Bogos, K Tamasi, L Ostoros, Gy Muller, V Urban, L Bittner, N Sarosi, V Vastag, A Polanyi, Z Nagy-Erdei, Zs Knollmajer, K Varnai, M Nagy, B Horvath, K Rokszin, Gy Abonyi-Toth, Zs Barcza, Zs Moldvay, J Galffy, G Voko, Z AF Kiss, Zoltan Bogos, Krisztina Tamasi, Lilla Ostoros, Gyula Muller, Veronika Urban, Laszlo Bittner, Nora Sarosi, Veronika Vastag, Aladar Polanyi, Zoltan Nagy-Erdei, Zsofia Knollmajer, Kata Varnai, Mate Nagy, Balazs Horvath, Krisztian Rokszin, Gyorgy Abonyi-Toth, Zsolt Barcza, Zsofia Moldvay, Judit Galffy, Gabriella Voko, Zoltan TI Increase in the Length of Lung Cancer Patient Pathway Before First-Line Therapy: A 6-Year Nationwide Analysis From Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lung cancer; patient pathway; time to treatment; episode of care; system interval ID lung cancer; patient pathway; time to treatment; episode of care; system interval AB Objective: This study aimed to examine the characteristics of the lung cancer (LC) patient pathway in Hungary during a 6-years period. Methods: This nationwide, retrospective study included patients newly diagnosed with LC (ICD-10 C34) between January 1, 2011, and December 31, 2016, using data from the National Health Insurance Fund (NHIF) of Hungary. The following patient pathway intervals were examined: system, diagnostic and treatment interval by age, gender, tumor type, study year and first-line LC therapy. Results: During the 6-years study period, 17,386 patients had at least one type of imaging (X-ray or CT/MRI) prior to diagnosis, and 12,063 had records of both X-ray and CT/MRI. The median system interval was 64.5 days, and it was 5 days longer among women, than in men (68.0 vs. 63.0 days). The median system interval was significantly longer in patients with adenocarcinoma compared to those with squamous cell carcinoma or small cell lung cancer (70.4 vs. 64.0 vs. 48.0 days, respectively). Patients who received surgery as firstline treatment had significantly longer median system intervals compared to those receiving chemotherapy (81.4 vs. 62.0 days). The median system interval significantly increased from 62.0 to 66.0 days during the 6-years study period. Conclusion: The LC patient pathway significantly increased in Hungary over the 6-years study period. There were no significant differences in the length of the whole LC patient pathway according to age, however, female sex, surgery as first-line treatment, and adenocarcinoma were associated with longer system intervals. C1 [Kiss, Zoltan] MSD Pharma Hungary Kft.Budapest, Hungary. [Bogos, Krisztina] National Koranyi Institute of PulmonologyBudapest, Hungary. [Tamasi, Lilla] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Ostoros, Gyula] National Koranyi Institute of PulmonologyBudapest, Hungary. [Muller, Veronika] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Urban, Laszlo] Matrahaza Healthcare Center and University Teaching HospitalMatrahaza, Hungary. [Bittner, Nora] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Sarosi, Veronika] University of Pecs, Faculty of MedicinePecs, Hungary. [Vastag, Aladar] MSD Pharma Hungary Kft.Budapest, Hungary. [Polanyi, Zoltan] MSD Pharma Hungary Kft.Budapest, Hungary. [Nagy-Erdei, Zsofia] MSD Pharma Hungary Kft.Budapest, Hungary. [Knollmajer, Kata] MSD Pharma Hungary Kft.Budapest, Hungary. [Varnai, Mate] MSD Pharma Hungary Kft.Budapest, Hungary. [Nagy, Balazs] Semmelweis University, Center for Health Technology AssessmentBudapest, Hungary. [Horvath, Krisztian] Semmelweis University, Center for Health Technology AssessmentBudapest, Hungary. [Rokszin, Gyorgy] RxTarget LtdSzolnok, Hungary. [Abonyi-Toth, Zsolt] RxTarget LtdSzolnok, Hungary. [Barcza, Zsofia] Syntesia LtdBudapest, Hungary. [Moldvay, Judit] National Koranyi Institute of Pulmonology, Semmelweis University, Ist Department of PulmonologyBudapest, Hungary. [Galffy, Gabriella] County Hospital of PulmonologyTorokbalint, Hungary. [Voko, Zoltan] Semmelweis University, Center for Health Technology AssessmentBudapest, Hungary. RP Kiss, Z (reprint author), MSD Pharma Hungary Kft., Budapest, Hungary. EM zoltan_kiss2@merck.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, et al. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 7068(46):313394–424. 2018 Jul., DOI 10.3322/caac.21492 Carter-Harris L, Hermann CP, Draucker CB. Pathways to a Lung Cancer Diagnosis. J Am Assoc Nurse Pract, 2015, 27(10):576–83., DOI 10.1002/2327- 6924.12242 International Agency for Research on Cancer,, 2019, Globocan: Lung Cancer Fact Sheet 2018. Available at: http://gco.iarc.fr/today/data/factsheets/cancers/ 15-Lung-fact-sheet.pdf Last downloaded at 2019.04.16. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JWW, Comber H, et al. Cancer Incidence and Mortality Patterns in Europe: Estimates for 40 Countries in 2012. Eur J Cancer, 2013, 49:1374–403., DOI 10.1016/ j.ejca.2012.12.027 Bogos K, Kiss Z, Galffy G, Tamasi L, Ostoros G, Muller V, et al. Revising Incidence and Mortality of Lung Cancer in Central Europe: An Epidemiology Review from Hungary. Front Oncol, 2019, 9:1051, 2019 . Published 2019 Oct 23., DOI 10.3389/fonc.2019.01051 Bogos K, Kiss Z, Galffy G, Tamasi L, Ostoros G, Muller V, et al. Lung Cancer in Hungary. J Thorac Oncol, 2020, 15(5):692–9. May., DOI 10.1016/ j.jtho.2019.11.001 Vinod SK, Chandra A, Berthelsen A, Descallar J. Does Timeliness of Care in Non-small Cell Lung Cancer Impact on Survival. Lung Cancer, 2017, 112: 16–24., DOI 10.1016/j.lungcan.2017.07.032 Coleman M, Forman D, Bryant H, Butler J, Rachet B, Maringe C, et al. Cancer Survival in Australia, Canada, Denmark, Norway, Sweden, and the UK, 1995- 2007, The International Cancer Benchmarking Partnership): an Analysis of Population-Based Cancer Registry Data. The Lancet, 2011, 377(9760):127–38., DOI 10.1016/S0140-6736(10)62231-3 Heuvers ME, Hegmans JP, Stricker BH, Aerts JG. Improving Lung Cancer Survival; Time to Move on. BMC Pulm Med, 2012, 12:77, 2012 . Published 2012 Dec 13., DOI 10.1186/1471-2466-12-77 Purdie S, Creighton N, White KM, Baker D, Ewald D, Lee CK, et al. Pathways to Diagnosis of Non-small Cell Lung Cancer: a Descriptive Cohort Study. Npj Prim Care RespMed, 2019, 29(12). Published 2019 Feb 8., DOI 10.1038/s41533-018-0113-7 Olsson JK, Schultz EM, Gould MK. Timeliness of Care in Patients with Lung Cancer: a Systematic Review. Thorax, 2009, 64(9):749–56., DOI 10.1136/ thx.2008.109330 Mohammed N, Kestin LL,Grills IS, BattuM, Fitch DL,Wong C-y.O, et al. Rapid Disease Progression with Delay in Treatment of Non-small-cell Lung Cancer. Int J Radiat Oncology*Biology*Physics, 2011, 79(2):466–72., DOI 10.1016/ j.ijrobp.2009.11.029 BTS Recommendations to Respiratory Physicians for Organising the Care of Patients with Lung Cancer. The Lung Cancer Working Party of the British Thoracic Society Standards of Care Committee. Thorax. 1998;53 Suppl 1 Suppl. 1(Suppl. 1):S1–8., DOI 10.1136/thx.53.suppl_1.s1 Reifel JL. Lung Cancer. In: SM Asch, EA Kerr, EG Hamilton, JL Reifel, EA McGlynn, editors. Quality of Care for Oncologic Conditions and HIV: A Review of the Literature and Quality Indicators. Santa Monica, CA: RAND Corporation, 2000). Available at: www.rand.org/content/dam/rand/pubs/ monograph_reports/MR1281/mr1281.ch7.pdf, accessed September 4, 2020). Myrdal G, Lambe M, Hillerdal G, Lamberg K, Agustsson T, Stahle E. Effect of Delays on Prognosis in Patients with Non-small Cell Lung Cancer. Thorax, 2004, 59(1):45–9. National Collaborating Centre for Cancer, UK). The Diagnosis and Treatment of Lung Cancer, Update). Cardiff, UK)UK): National Collaborating Centre for Cancer, 2011). Kim JOA, Davis F, Butts C, Winget M. Waiting Time Intervals for Non-small Cell Lung Cancer Diagnosis and Treatment in Alberta: Quantification of Intervals and Identification of Risk Factors Associated with Delays. Clin Oncol, 2016, 28(12):750–9., DOI 10.1016/j.clon.2016.06.010 Salomaa E-R, Sallinen S, Hiekkanen H, Liippo K. Delays in the Diagnosis and Treatment of Lung Cancer. Chest, 2005, 128(4):2282–8., DOI 10.1378/ chest.128.4.2282 Quarterman RL, McMillan A, Ratcliffe MB, Block MI. Effect of Preoperative Delay on Prognosis for Patients with Early Stage Non-small Cell Lung Cancer. J Thorac Cardiovasc Surg, 2003, 125(1):108–14., DOI 10.1067/mtc.2003.93 Yorio JT, Xie Y, Yan J, Gerber DE. Lung Cancer Diagnostic and Treatment Intervals in the United States: a Health Care Disparity. J Thorac Oncol, 2009, 4(11):1322–30., DOI 10.1097/JTO.0b013e3181bbb130 Jacobsen MM, Silverstein SC, QuinnM,Waterston LB, Thomas CA, Benneyan JC, et al. Timeliness of Access to Lung Cancer Diagnosis and Treatment: A Scoping Literature Review. Lung Cancer, 2017, 112:156–64., DOI 10.1016/ j.lungcan.2017.08.011 Dalton SO, Frederiksen BL, Jacobsen E, Steding-Jessen M, Osterlind K, Schuz J, et al. Socioeconomic Position, Stage of Lung Cancer and Time between Referral and Diagnosis in Denmark, 2001-2008. Br J Cancer, 2011, 105(7): 1042–8., DOI 10.1038/bjc.2011.342 Berglund A, Holmberg L, Tishelman C, Wagenius G, Eaker S, Lambe M. Social Inequalities in Non-small Cell Lung Cancer Management and Survival: a Population-Based Study in central Sweden. Thorax, 2010, 65(4):327–33., DOI 10.1136/thx.2009.125914 Gomez DR, Liao K-P, Swisher SG, Blumenschein GR, Erasmus JJ, Buchholz TA, et al. Time to Treatment as a Quality Metric in Lung Cancer: Staging Studies, Time to Treatment, and Patient Survival. Radiother Oncol, 2015, 115(2):257–63., DOI 10.1016/j.radonc.2015.04.010 Hall SE, Holman CDAJ, Threlfall T, Sheiner H, Phillips M, Katriss P, et al. Lung Cancer: an Exploration of Patient and General Practitioner Perspectives on the Realities of Care in Rural Western Australia. Aust J Rural Health, 2008, 16(6):355–62. Dec., DOI 10.1111/j.1440-1584.2008.01016.x Verma R, Pathmanathan S, Otty ZA, Binder J, Vangaveti VN, Buttner P, et al. Delays in Lung Cancer Management Pathways between Rural and Urban Patients in North Queensland: a Mixed Methods Study. Intern Med J, 2018, 48(10):1228–33., DOI 10.1111/imj.13934 Malalasekera A, Nahm S, Blinman PL, Kao SC, Dhillon HM, Vardy JL. How Long Is Too Long? A Scoping Review of Health System Delays in Lung Cancer. Eur Respir Rev, 2018, 27(149):180045, 2018 . Published 2018 Aug 29., DOI 10.1183/16000617.0045-2018 Bogos K, Kiss Z, Tamasi L, Ostoros G, Muller V, Urban L, et al. Improvement in Lung Cancer Survival: 6-Year Trends of Overall Survival at Hungarian Patients Diagnosed in 2011-2016. Pathol Oncol Res, 2021, 27:603937., DOI 10.3389/pore.2021.603937 Hunnibell LS, Rose MG, Connery DM, Grens CE, Hampel JM, Rosa M, et al. Using Nurse Navigation to Improve Timeliness of Lung Cancer Care at a Veterans Hospital. Clin J Oncol Nurs, 2012, 16(1):29–36., DOI 10.1188/ 12.CJON.29-36 Neal RD, Din NU, Hamilton W, Ukoumunne OC, Carter B, Stapley S, et al. Comparison of Cancer Diagnostic Intervals before and after Implementation of NICE Guidelines: Analysis of Data from the UK General Practice Research Database. Br J Cancer, 2014, 110(3):584–92., DOI 10.1038/bjc.2013.791 Schultz EM, Powell AA, McMillan A, Olsson JK, Enderle MA, Graham BA, et al. Hospital Characteristics Associated with Timeliness of Care in Veterans with Lung Cancer. Am J Respir Crit Care Med, 2009, 179(7):595–600., DOI 10.1164/rccm.200806-890oc Faris N, Yu X, Sareen S, Signore RS, McHugh LM, Roark K, et al. Preoperative Evaluation of Lung Cancer in a Community Health Care Setting. Ann Thorac Surg, 2015, 100(2):394–400. Aug., DOI 10.1016/ j.athoracsur.2015.03.008 Verma A, Lim AYH, Tai DYH, Goh SK, Kor AC, A. DBA, et al. Timeliness of Diagnosing Lung Cancer. Medicine, 2015, 94(29):e1216., DOI 10.1097/ md.0000000000001216 Wang B-Y, Huang J-Y, Chen H-C, Lin C-H, Lin S-H, Hung W-H, et al. The Comparison between Adenocarcinoma and Squamous Cell Carcinoma in Lung Cancer Patients. J Cancer Res Clin Oncol, 2020, 146(1):43–52., DOI 10.1007/s00432-019-03079-8 Degree of Hungarian Ministry of Human Resources, 2015, Degree of Hungarian Ministry of Human Resources Available at: http://www. kozlonyok.hu/nkonline/mkpdf/hiteles/mk15210.pdf(Accessed 1 Jun 2015) Hanna TP, King WD, Thibodeau S, Jalink M, Paulin GA, Harvey-Jones E, et al. Mortality Due to Cancer Treatment Delay: Systematic Review and Meta- Analysis. Bmj, 2020, 371:m4087., DOI 10.1136/bmj.m4087 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1610041 EP 1610051 DI 10.3389/pore.2021.1610041 PG 11 ER PT J AU Forika, G Kiss, Petovari, G Danko, T Gellert, BA Krenacs, T AF Forika, Gertrud Kiss, Eva Petovari, Gabor Danko, Titanilla Gellert, Bertram Aron Krenacs, Tibor TI Modulated Electro-Hyperthermia Supports the Effect of Gemcitabine Both in Sensitive and Resistant Pancreas Adenocarcinoma Cell Lines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apoptosis; cell cycle arrest; pancreas adenocarcinoma; modulated electro-hyperthermia; gemcitabine resistance; combined treatment ID apoptosis; cell cycle arrest; pancreas adenocarcinoma; modulated electro-hyperthermia; gemcitabine resistance; combined treatment AB The poor prognosis of pancreatic ductal adenocarcinoma (PDAC) is frequently associated to high treatment resistance. Gemcitabine (GEM) alone or in combination is the most used chemotherapy for unresecable PDACs. Here we studied whether modulated electrohyperthermia (mEHT), a non-invasive complementary treatment, can support the effect of GEM on PDAC cells in vitro. The LD20 for the GEM-resistant Panc1 cells proved to be 200× higher than for the drug-sensitive Capan1. The mEHT alone caused significant apoptosis in Capan1 cultures as confirmed by the elevated SubG1 phase cell fraction and increased number of cleaved Caspase-3 positive cells 48 h after treatment, with an additive effect when GEM was used after hyperthermia. These were accompanied by reduced number of G1, S, and G2/M phase cells and elevated expression of the cyclindependent kinase inhibitor p21waf1 protein. In GEM-resistant Panc1 cells, an initial apoptosis was detected by flow cytometry 24 h after mEHT ± GEM treatment, which however diminished by 48 h at persistent number of cleaved Caspase-3 positive tumor cells. Though GEM monotherapy reduced the number of tumor progenitor colonies in Capan1 cell line, an additive colony inhibitory effect of mEHT was observed after mEHT + GEM treatment. The heat shock induced Hsp27 and Hsp70 proteins, which are known to sensitize PDAC cells to GEM were upregulated in both Capan1 and Panc1 cells 24 h after mEHT treatment. The level of E-Cadherin, a cell adhesion molecule, increased in Capan1 cells after mEHT + GEM treatment. In conclusion, in GEM-sensitive PDAC cells mEHT treatment alone induced cell death and cell cycle inhibition and improved GEM efficiency in combination, which effects were milder and short-term up to 24 h in the GEM-resistant Panc1 cells. Our data further support the inclusion of hyperthermia, in particular of mEHT, into the traditional oncotherapy regimens of PDAC. C1 [Forika, Gertrud] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kiss, Eva] Semmelweis University, 1st Department of Internal Medicine and Oncology, Oncology ProfileBudapest, Hungary. [Petovari, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Danko, Titanilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Gellert, Bertram Aron] Orszagos Onkologiai Intezet, Mellkassebeszeti OsztalyBudapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Krenacs, T (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM krenacs.tibor@med.semmelweis-univ.hu CR Sarantis P, Koustas E, Papadimitropoulou A, Papavassiliou AG, Karamouzis MV. Pancreatic Ductal Adenocarcinoma: Treatment Hurdles, Tumor Microenvironment and Immunotherapy. World J Gastrointest Oncol, 2020, 12(2):173–81., DOI 10.4251/wjgo.v12.i2.173 Oberstein PE, Olive KP. Pancreatic Cancer: Why Is it So Hard to Treat? Therap Adv Gastroenterol, 2013, 6(4):321–37., DOI 10.1177/1756283x13478680 National Cancer Institute. Pancreatic Cancer, 2009-2017). Available from: https:// seer.cancer.gov/statfacts/html/pancreas.html. Accessed: August 25, 2021. Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus Gemcitabine for Metastatic Pancreatic Cancer. N Engl J Med, 2011, 364(19):1817–25., DOI 10.1056/nejmoa1011923 Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased Survival in Pancreatic Cancer with Nab-Paclitaxel Plus Gemcitabine. N Engl J Med, 2013, 369(18):1691–703., DOI 10.1056/nejmoa1304369 Amrutkar M, Vethe NT, Verbeke CS, Aasrum M, Finstadsveen AV, Santha P, et al. Differential Gemcitabine Sensitivity in Primary Human Pancreatic Cancer Cells and Paired Stellate Cells Is Driven by Heterogenous Drug Uptake and Processing. Cancers, 2020, 12(12):3628., DOI 10.3390/ cancers12123628 Cheng Y, Weng S, Yu L, Zhu N, Yang M, Yuan Y. The Role of Hyperthermia in the Multidisciplinary Treatment of Malignant Tumors. Integr Cancer Ther, 2019, 18:1534735419876345., DOI 10.1177/1534735419876345 Hildebrandt B, Wust P, Ahlers O, Dieing A, Sreenivasa G, Kerner T, et al. The Cellular and Molecular Basis of Hyperthermia. Crit Rev Oncol Hematol, 2002, 43(1):33–56., DOI 10.1016/s1040-8428(01)00179-2 Pascale RM, Calvisi DF, Simile MM, Feo CF, Feo F. The Warburg Effect 97 Years after its Discovery. Cancers, 2020, 12(10):2819., DOI 10.3390/ cancers12102819 Andocs G, Meggyeshazi N, Balogh L, Spisak S, Maros ME, Balla P, et al. Upregulation of Heat Shock Proteins and the Promotion of Damage- Associated Molecular Pattern Signals in a Colorectal Cancer Model by Modulated Electrohyperthermia. Cell Stress and Chaperones, 2015, 20(1): 37–46., DOI 10.1007/s12192-014-0523-6 Hegyi G, Szigeti GP, Szasz A. Hyperthermia versus Oncothermia: Cellular Effects in Complementary Cancer Therapy. Evid Based Complement Alternat Med, 2013, 2013:672873., DOI 10.1155/2013/672873 Lee SY, LeeNR,ChoD-H, KimJS. TreatmentOutcomeAnalysis of Chemotherapy Combined with Modulated Electro-Hyperthermia Compared with Chemotherapy Alone for Recurrent Cervical Cancer, Following Irradiation. Oncol Lett, 2017, 14(1):73–8., DOI 10.3892/ol.2017.6117 Fiorentini G, Sarti D, Casadei V, Milandri C, Dentico P, Mambrini A, et al. Modulated Electro-Hyperthermia as Palliative Treatment for PancreaticCancer: A Retrospective Observational Study on 106 Patients. Integr Cancer Ther, 2019, 18: 1534735419878505., DOI 10.1177/1534735419878505 Minnaar CA, Kotzen JA, Ayeni OA, Naidoo T, Tunmer M, Sharma V, et al. The Effect of Modulated Electro-Hyperthermia on Local Disease Control in HIV-Positive and -negative Cervical Cancer Women in South Africa: Early Results from a Phase III Randomised Controlled Trial. PloS one, 2019, 14(6): e0217894., DOI 10.1371/journal.pone.0217894 Fiorentini G, Sarti D, Milandri C, Dentico P, Mambrini A, Fiorentini C, et al. Modulated Electrohyperthermia in Integrative Cancer Treatment for Relapsed Malignant Glioblastoma and Astrocytoma: Retrospective Multicenter Controlled Study. Integr Cancer Ther, 2019, 18:1534735418812691., DOI 10.1177/1534735418812691 Szasz AM, Minnaar CA, Szentmartoni G, Szigeti GP, Dank M. Review of the Clinical Evidences of Modulated Electro-Hyperthermia, mEHT, Method: An Update for the Practicing Oncologist. Front Oncol, 2019, 9:1012., DOI 10.3389/ fonc.2019.01012 Krenacs T, Meggyeshazi N, Forika G, Kiss E, Hamar P, Szekely T, et al. Modulated Electro-Hyperthermia-Induced Tumor Damage Mechanisms Revealed in Cancer Models. Int J Mol Sci, 2020, 21(17):6270., DOI 10.3390/ ijms21176270 Vancsik T, Kovago C,Kiss E, Papp E, Forika G, Benyo Z, et al. Modulated Electro- Hyperthermia Induced Loco-Regional and Systemic Tumor Destruction in Colorectal Cancer Allografts. J Cancer, 2018, 9(1):41–53., DOI 10.7150/jca.21520 Vancsik T, Forika G, Balogh A, Kiss E, Krenacs T. Modulated Electrohyperthermia Induced P53 Driven Apoptosis and Cell Cycle Arrest Additively Support Doxorubicin Chemotherapy of Colorectal Cancer In Vitro. Cancer Med, 2019, 8(9):4292–303., DOI 10.1002/cam4.2330 Meggyeshazi N, Andocs G, Balogh L, Balla P, Kiszner G, Teleki I, et al. DNA Fragmentation and Caspase-independent Programmed Cell Death by Modulated Electrohyperthermia. Strahlenther Onkol, 2014, 190(9):815–22., DOI 10.1007/s00066-014-0617-1 Amaral MVS, De Sousa Portilho AJ, Da Silva EL, De Oliveira Sales L, Da Silva Maues JH, De Moraes MEA, et al. Establishment of Drug-Resistant Cell Lines as aModel in Experimental Oncology: A Review. Anticancer Res, 2019, 39(12): 6443–55., DOI 10.21873/anticanres.13858 Forika G, Balogh A, Vancsik T, Zalatnai A, Petovari G, Benyo Z, et al. Modulated Electro-Hyperthermia Resolves Radioresistance of Panc1 Pancreas Adenocarcinoma and Promotes DNA Damage and Apoptosis In Vitro. Int J Mol Sci, 2020, 21(14):5100., DOI 10.3390/ijms21145100 Vitellius C, Fizanne L, Menager-Tabourel E, Nader J, Baize N, Laly M, et al. The Combination of Everolimus and Zoledronic Acid Increase the Efficacy of Gemcitabine in a Mouse Model of Pancreatic Adenocarcinoma. Oncotarget, 2018, 9(46):28069–82., DOI 10.18632/oncotarget.25560 Du J, Gu J, Li J. Mechanisms of Drug Resistance of Pancreatic Ductal Adenocarcinoma at Different Levels. Biosci Rep, 2020, 40(7):BSR20200401., DOI 10.1042/BSR20200401 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence andMortality Worldwide for 36 Cancers in 185 Countries. CA: a Cancer J clinicians, 2018, 68(6): 394–424., DOI 10.3322/caac.21492 Amrutkar M, Gladhaug IP. Pancreatic Cancer Chemoresistance to Gemcitabine. Cancers, Basel,, 2017, 9(11):157., DOI 10.3390/cancers9110157 Riggs MJ, Pandalai PK, Kim J, Dietrich CS. Hyperthermic Intraperitoneal Chemotherapy in Ovarian Cancer. Diagnostics, Basel,, 2020, 10(1):10043., DOI 10.3390/diagnostics10010043 You SH, Kim S. Feasibility of Modulated Electro-Hyperthermia in Preoperative Treatment for Locally Advanced Rectal Cancer: Early Phase 2 Clinical Results. Neoplasma, 2020, 67(3):677–83., DOI 10.4149/ neo_2020_190623N538 Funamizu N, Lacy CR, Kamada M, Yanaga K, Manome Y. MicroRNA-200b and -301 Are Associated with Gemcitabine Response as Biomarkers in Pancreatic Carcinoma Cells. Int J Oncol, 2019, 54(3):991–1000., DOI 10.3892/ijo.2019.4676 Jin H, Zhao Y, Yang J, Zhang X, Ma S. Hyperthermia Enhances the Sensitivity of Pancreatic Cancer SW1990 Cells to Gemcitabine through ROS/JNK Signaling. Oncol Lett, 2018, 16(5):6742–8., DOI 10.3892/ol.2018.9455 Sanhaji M, Goring J, Couleaud P, Aires A, Cortajarena AL, Courty J, et al. The Phenotype of Target Pancreatic Cancer Cells Influences Cell Death by Magnetic Hyperthermia with Nanoparticles Carrying Gemicitabine and the Pseudo-peptide NucAnt. Nanomedicine: Nanotechnology, Biol Med, 2019, 20: 101983., DOI 10.1016/j.nano.2018.12.019 Morales-Cruz M, Figueroa CM, Gonzalez-Robles T, Delgado Y, Molina A, Mendez J, et al. Activation of Caspase-dependent Apoptosis by Intracellular Delivery of Cytochrome C-Based Nanoparticles. J Nanobiotechnol, 2014, 12: doi:10.1186/s12951-014-0033-9 33. Thomas MJ, Major E, Benedek A, Horvath I, Mathe D, Bergmann R, et al. Suppression of Metastatic Melanoma Growth in Lung by Modulated Electro- HyperthermiaMonitored by a Minimally InvasiveHeat Stress Testing Approach in Mice. Cancers, Basel,, 2020, 12(12):3872., DOI 10.3390/cancers12123872 Yang Y, Yang CL, Zhao ZJ, Zuo XX, Liang TS, Yang Y, et al. Microwave Hyperthermia Enhances the Sensitivity of Lung Cancer Cells to Gemcitabine through Reactive Oxygen Species-Induced Autophagic Death. Oncol Rep, 2019, 41(5):3100–10., DOI 10.3892/or.2019.7085 Vertrees RA, Das GC, Popov VL, Coscio AM, Goodwin TJ, Logrono R, et al. Synergistic Interaction of Hyperthermia and Gemcitabine in Lung Cancer. Cancer Biol Ther, 2005, 4(10):1144–53., DOI 10.4161/cbt.4.10.2074 Guo Y, Ziesch A, Hocke S, Kampmann E, Ochs S, De Toni EN, et al. Overexpression of Heat Shock Protein 27, HSP 27, Increases Gemcitabine Sensitivity in Pancreatic Cancer Cells through S-phase Arrest and Apoptosis. J Cel Mol. Med., 2015, 19(2):340–50., DOI 10.1111/jcmm.12444 Zhao YY, Wu Q, Wu ZB, Zhang JJ, Zhu LC, Yang Y, et al. Microwave Hyperthermia Promotes Caspase-3-dependent Apoptosis and Induces G2/M Checkpoint Arrest via the ATM Pathway in Non-small Cell Lung Cancer Cells. Int J Oncol, 2018, 53(2):539–50., DOI 10.3892/ijo.2018.4439 Schafer C, Seeliger H, Bader DC, Assmann G, Buchner D, Guo Y, et al. Heat Shock Protein 27 as a Prognostic and Predictive Biomarker in Pancreatic Ductal Adenocarcinoma. J Cel Mol Med, 2012, 16(8):1776–91., DOI 10.1111/ j.1582-4934.2011.01473.x Danics L, Schvarcz CA, Viana P, Vancsik T, Krenacs T, Benyo Z, et al. Exhaustion of Protective Heat Shock Response Induces Significant Tumor Damage by Apoptosis after Modulated Electro-Hyperthermia Treatment of Triple Negative Breast Cancer Isografts in Mice. Cancers, Basel,, 2020, 12(9): 2581., DOI 10.3390/cancers12092581 Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, et al. Epithelial-tomesenchymal Transition Is Dispensable for Metastasis but Induces Chemoresistance in Pancreatic Cancer. Nature, 2015, 527(7579):525–30., DOI 10.1038/nature16064 Park SJ, Kim S-M, Moon J-H, Kim JH, Shin J-S, Hong S-W, et al. SAHA, an HDAC Inhibitor, Overcomes Erlotinib Resistance in Human Pancreatic Cancer Cells by Modulating E-Cadherin. Tumor Biol, 2016, 37(4):4323–30., DOI 10.1007/s13277-015-4216-2 Jin H, Zhao Y, Zhang S, Yang J, Zhang X, Ma S. Hyperthermia Inhibits the Motility of Gemcitabine-Resistant Pancreatic Cancer PANC-1 Cells through the Inhibition of Epithelial-Mesenchymal Transition. Mol Med Rep, 2018, 17(5):7274–80., DOI 10.3892/mmr.2018.8763 Ercan G, Karlitepe A, Ozpolat B. Pancreatic Cancer Stem Cells and Therapeutic Approaches. Anticancer Res, 2017, 37(6):2761–75., DOI 10.21873/anticanres.11628 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1610048 EP 1610062 DI 10.3389/pore.2021.1610048 PG 15 ER PT J AU Deng, Y Yu, L Zhao, Y Peng, J Xu, Y Qin, J Xiao, B Liu, S Li, M Fang, Y Pan, Z AF Deng, Yuxiang Yu, Long Zhao, Yujie Peng, Jianhong Xu, Yanbo Qin, Jiayi Xiao, Binyi Liu, Songran Li, Mei Fang, Yujing Pan, Zhizhong TI RCC1 Expression as a Prognostic Marker in Colorectal Liver Oligometastases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; expression; RCC1; colorectal liver oligometastases; pathway ID prognosis; expression; RCC1; colorectal liver oligometastases; pathway AB Introduction: Regulator of chromatin condensation 1 (RCC1) is a major guaninenucleotide exchange factor for Ran GTPase, and it plays key roles in various biological processes. Previous studies have found that RCC1 may play a role in the development of tumors, but little is known about the relationship between RCC1 and colorectal liver oligometastases (CLOs). Methods: One hundred and twenty-nine pairs of matched human CLO samples, including both primary tumor and its liver metastasis specimens, were subjected to immunohistochemistry to determine the location and expression levels of RCC1. Associations between RCC1 and survival as well as gene expression profiling were explored. Results: In this study, we first observed that RCC1 was mildly increased in CLO tumor tissues compared with normal tissues, and the localization was primarily nuclear. In addition, our study found that high RCC1 expression in liver oligometastases was an independent prognostic marker for unfavorable recurrence-free survival and overall survival (p = 0.036 and p = 0.016). Gene expression profiles generated from microarray analysis showed that RCC1 was involved in pathways including “Myc targets,” “E2F targets” and “DNA repair” pathways. Conclusion: Our data indicated that RCC1 was expressed mainly in the nucleus, and strong and significant associations were found between RCC1 expression levels and the survival of CLO patients. These findings indicated that RCC1 may play a role in CLO development. C1 [Deng, Yuxiang] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Yu, Long] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Zhao, Yujie] Peking University Shenzhen Hospital, Department of Radiation OncologyShenzhen, China. [Peng, Jianhong] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Xu, Yanbo] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Qin, Jiayi] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Xiao, Binyi] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Liu, Songran] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of PathologyGuangzhou, China. [Li, Mei] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of PathologyGuangzhou, China. [Fang, Yujing] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. [Pan, Zhizhong] Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal SurgeryGuangzhou, China. RP Pan, Z (reprint author), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Colorectal Surgery, Guangzhou, China. EM panzhzh@sysucc.org.cn CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clinjournal Article, 2021, 71:209–49., DOI 10.3322/caac.21660 Feng R-M, Zong Y-N, Cao S-M, Xu R-H. Current Cancer Situation in China: Good or Bad News from the 2018 Global Cancer Statistics?. Cancer Commun, 2019, 39:22, 2019 . Journal Article., DOI 10.1186/s40880-019- 68-6 3. Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, et al. Colorectal Cancer Statistics, 2020. CA A Cancer J Clin, 2020, 70: 70145–164., DOI 10.3322/caac.21601 Miller KD, Nogueira L, Mariotto AB, Rowland JH, Yabroff KR, Alfano CM, et al. Cancer Treatment and Survivorship Statistics, 2019. CA A Cancer J Clin, 2019, 69:363–85., DOI 10.3322/caac.21565 Engstrand J, Nilsson H, Stromberg C, Jonas E, Freedman J. Colorectal Cancer Liver Metastases - a Population-Based Study on Incidence, Management and Survival. Bmc Cancer, 2018, 18:78., DOI 10.1186/ s12885-017-3925-x Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO Consensus Guidelines for the Management of Patients with Metastatic Colorectal Cancer. Ann Oncol, 2016, 27:1386–422., DOI 10.1093/ annonc/mdw235 Lin J, Peng J, Zhao Y, Luo B, Zhao Y, Deng Y, et al. Early Recurrence in Patients Undergoing Curative Resection of Colorectal Liver Oligometastases: Identification of its Clinical Characteristics, Risk Factors, and Prognosis. J Cancer Res Clin Oncol, 2018, 144:359–69., DOI 10.1007/s00432-017-2538-8 Nishimoto T, Eilen E, Basilico C. Premature Chromosome Condensation in a Ts DNA-Mutant of BHK Cells. Cell, 1978, 15:475–83., DOI 10.1016/0092- 8674(78)90017-x Yang J, Guo Y, Lu C, Zhang R, Wang Y, Luo L, et al. Inhibition of Karyopherin Beta 1 Suppresses Prostate Cancer Growth. Oncogene, 2019, 38:4700–14., DOI 10.1038/s41388-019-0745-2 Ghobrial IM, McCormick DJ, Kaufmann SH, Leontovich AA, Loegering DA, Dai NT. Proteomic Analysis of Mantle-Cell Lymphoma by Protein Microarray. Blood, 2005, 105:3722–30., DOI 10.1182/blood-2004-10- 3999 Peters D, Freund J, Ochs RL. Genome-wide Transcriptional Analysis of Carboplatin Response in Chemosensitive and Chemoresistant Ovarian Cancer Cells. Mol Cancer Ther, 2005, 4:1605–16., DOI 10.1158/1535- 7163.mct-04-0311 Alhopuro P, Sammalkorpi H, Niittymaki I, Bistrom M, Raitila A, Saharinen J, et al. Candidate Driver Genes in Microsatellite-Unstable Colorectal Cancer. Int J Cancer, 2012, 130:1558–66., DOI 10.1002/ijc.26167 Scotto L, Narayan G, Nandula SV, Arias-Pulido H, Subramaniyam S, Schneider A, et al. Identification of Copy Number Gain and Overexpressed Genes on Chromosome Arm 20q by an Integrative Genomic Approach in Cervical Cancer: Potential Role in Progression. Genes Chromosom Cancer, 2008, 47:755–65., DOI 10.1002/gcc.20577 Hsu C-H, Hsu C-W, Hsueh C, Wang C-L, Wu Y-C, Wu C-C, et al. Identification and Characterization of Potential Biomarkers by Quantitative Tissue Proteomics of Primary Lung Adenocarcinoma. Mol Cell Proteomics, 2016, 15:2396–410., DOI 10.1074/mcp.M115.057026 Zhang B, Huang B, Guan H, Zhang S-M, Xu Q-Z, He X-P, et al. Proteomic Profiling Revealed the Functional Networks Associated with Mitotic Catastrophe of HepG2 Hepatoma Cells Induced by 6-Bromine-5-Hydroxy- 4-Methoxybenzaldehyde. Toxicol Appl Pharmacol, 2011, 252:307–17., DOI 10.1016/j.taap.2011.03.003 Lin YL, Chen HL, Cheng SB, Yeh DC, Huang CC, P’Eng FK, et al. Methylation-silencing RCC1 Expression Is Associated with Tumorigenesis and Depth of Invasion in Gastric Cancer. Int J Clin Exp Pathol, 8, 2015). p. 14257–69. Tracey L, Aggarwal M, Garcia-Cosio M, Villuendas R, Algara P, Sanchez-Beato M, et al. Somatic Hypermutation Signature in B-Cell Low-Grade Lymphomas. Haematologica, 2008, 93:1186–94., DOI 10.3324/haematol.12999 Zeng X, Zhong M, Yang Y, Wang Z, Zhu Y. Down-regulation of RCC1 Sensitizes Immunotherapy by Up-regulating PD-L1 via P27 Kip1/CDK4 axis in Non-small Cell Lung Cancer. J Cel Mol. Med.Journal Article, 2021, 25: 4136–47., DOI 10.1111/jcmm.16383 Bischoff FR, Ponstingl H. Catalysis of Guanine Nucleotide Exchange on Ran by the Mitotic Regulator RCC1. Nature, 1991, 354:80–2., DOI 10.1038/ 354080a0 Cekan P, Hasegawa K, Pan Y, Tubman E, Odde D, Chen J-Q, et al. RCC1- dependent Activation of Ran Accelerates Cell Cycle and DNA Repair, Inhibiting DNA Damage-Induced Cell Senescence. MBoC, 2016, 27: 1346–57., DOI 10.1091/mbc.E16-01-0025 Dasso M. RCC1 in the Cell Cycle: the Regulator of Chromosome Condensation Takes on New Roles. Trends Biochem Sci, 1993, 18:96–101., DOI 10.1016/0968- 0004(93)90161-f Ohtsubo M, Okazaki H, Nishimoto T. The RCC1 Protein, a Regulator for the Onset of Chromosome Condensation Locates in the Nucleus and Binds to DNA. J Cel Biol., 1989, 109:1389–97., DOI 10.1083/ jcb.109.4.1389 Riahi A, Radmanesh H, Schurmann P, Bogdanova N, Geffers R, Meddeb R, et al. Exome Sequencing and Case-Control Analyses identifyRCC1as a Candidate Breast Cancer Susceptibility Gene. Int J Cancer, 2018, 142: 2512–7., DOI 10.1002/ijc.31273 Haggag Y, Abu Ras B, El-Tanani Y, Tambuwala MM, McCarron P, Isreb M, et al. Co-delivery of a RanGTP Inhibitory Peptide and Doxorubicin Using Dual-Loaded Liposomal Carriers to Combat Chemotherapeutic Resistance in Breast Cancer Cells. Expert Opin Drug Deliv, 2020, 17:1655–69., DOI 10.1080/ 17425247.2020.1813714 Qiao L, Zheng J, Tian Y, Zhang Q,Wang X, Chen JJ, et al. Regulator of Chromatin Condensation 1 Abrogates the G1 Cell Cycle Checkpoint via Cdk1 in Human Papillomavirus E7-Expressing Epithelium and Cervical Cancer Cells. Cell Death Dis, 2018, 9:583., DOI 10.1038/s41419-018- 0584-z Deschamps T, Bazot Q, Leske DM, MacLeod R, Mompelat D, Tafforeau L, et al. Epstein-Barr Virus Nuclear Antigen 1 Interacts with Regulator of Chromosome Condensation 1 Dynamically throughout the Cell Cycle. J Gen Virol, 2017, 98:251–65. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD DEC PY 2021 VL 27 IS 4 BP 1610077 EP 1610086 DI 10.3389/pore.2021.1610077 PG 10 ER PT J AU Bienkiewicz, J Smolarz, B Wilczynski, M Stepowicz, A Jablonski, G Oblekowska, A Malinowski, A Romanowicz, H AF Bienkiewicz, Jan Smolarz, Beata Wilczynski, Milosz Stepowicz, Anna Jablonski, Grzegorz Oblekowska, Anna Malinowski, Andrzej Romanowicz, Hanna TI Is Single Nucleotide Polymorphism ADIPOQ (NM_004797.4):c.214+62G>T (rs1501299) Associated With Uterine Leiomyomas? A Pilot Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE single nucleotide polymorphism; obesity; uterine leiomyomas; uterine fibroids; c.276 G>T; adiponectin; ADIPOQ; (NM_004797.4):c.214+62G>T ID single nucleotide polymorphism; obesity; uterine leiomyomas; uterine fibroids; c.276 G>T; adiponectin; ADIPOQ; (NM_004797.4):c.214+62G>T AB Objective: Although polymorphisms of adiponectin gene (ADIPOQ) in obesity-related conditions have been the target of research efforts, little is known about this genetic marker in uterine leiomyomas. The aim of this pilot study was to analyze the frequencies of alleles and genotypes of Single Nucleotide Polymorphism ADIPOQ (NM_004797.4):c.214+62G>T (rs1501299) and to correlate it with the risk of uterine fibroids. Study Design: The Test Group comprised 90 women treated surgically for uterine leiomyomas in the Department of Operative Gynecology, Endoscopy and Gynecologic Oncology, PolishMother’sMemorial Hospital-Research Institute. 90 disease-free individuals were used as Controls. Patients within both groups were additionally stratified into lean, overweight and obese, according to Body Mass Index. Statistical analysis was performed between the two major groups and, furthermore, within the abovementioned subgroups. Results: The study revealed no statistically significant differences in the distribution of alleles and genotypes of SNP ADIPOQ (NM_004797.4):c.214+62G>T (rs1501299) between the two main groups. A weak correlation within distributions of alleles was observed between obese Test Patients and lean Controls. Conclusion: This pilot study has revealed no association between SNP ADIPOQ (NM_004797.4):c.214+62G>T (rs1501299) and uterine fibroids. Further studies on larger groups are warranted to elucidate whether this SNP may be correlated with uterine leiomyomas. C1 [Bienkiewicz, Jan] Polish Mothers’ Memorial Hospital-Research Institute, Department of Surgical & Endoscopic Gynecology and Gynecologic OncologyLodz, Poland. [Smolarz, Beata] Polish Mothers’ Memorial Hospital-Research Institute, Department of Pathology, Laboratory of Molecular GeneticsLodz, Poland. [Wilczynski, Milosz] Polish Mothers’ Memorial Hospital-Research Institute, Department of Surgical & Endoscopic Gynecology and Gynecologic OncologyLodz, Poland. [Stepowicz, Anna] Polish Mother’s Memorial Hospital- Research Institute, Department of Obstetrics, Perinatology and GynecologyLodz, Poland. [Jablonski, Grzegorz] Polish Mothers’ Memorial Hospital-Research Institute, Department of Surgical & Endoscopic Gynecology and Gynecologic OncologyLodz, Poland. [Oblekowska, Anna] Polish Mothers’ Memorial Hospital-Research Institute, Department of Surgical & Endoscopic Gynecology and Gynecologic OncologyLodz, Poland. [Malinowski, Andrzej] Medical University of Lodz, Department of Operative and Endoscopic GynecologyLodz, Poland. [Romanowicz, Hanna] Polish Mothers’ Memorial Hospital-Research Institute, Department of PathologyLodz, Poland. RP Bienkiewicz, J (reprint author), Polish Mothers’ Memorial Hospital-Research Institute, Department of Surgical & Endoscopic Gynecology and Gynecologic Oncology, Lodz, Poland. EM jan.bienkiewicz@iczmp.edu.pl CR Commandeur A. E., Styer A. K., Teixeira J. M. Epidemiological and Genetic Clues for Molecular Mechanisms Involved in Uterine Leiomyoma Development and Growth. Hum Reprod Update, 2015, 21(5):593–615., DOI 10.1093/humupd/dmv030 Giuliani E., As Sanie S., Marsh E. E. Epidemiology and Management of Uterine Fibroids. Int J Gynecol Obstet, 2020, 149(1):3–9. Epub 2020 Feb 17., DOI 10.1002/ijgo.13102 Gurusamy K. S., Vaughan J., Fraser I. S., Best L. M. J., Richards T. Medical Therapies for Uterine Fibroids - A Systematic Review and Network Meta- Analysis of Randomised Controlled Trials. PLoS One, 2016, 11(2):e0149631., DOI 10.1371/journal.pone.0149631 Ayyad I. Relationship between Fibroid Uterus and Obesity. MEJFM, 2011, 9(7):p7–9. 3p., DOI 10.5742/mejfm.2011.97071 Shikora S. A., Niloff J. M., Bistrian B. R., Forse R. A., Blackburn G. L. Relationship between Obesity and Uterine Leiomyomata. Nutrition, 1991, 7(4):251–5. Valimaki N., Kuisma H., Pasanen A., Heikinheimo O., Sjoberg J., Butzow R., et al. Genetic Predisposition to Uterine Leiomyoma Is Determined by Loci for Genitourinary Development and Genome Stability. Elife, 2018, 7:e37110. Published 2018 Sep 18., DOI 10.7554/eLife.37110 Moravek M. B., Bulun S. E. Endocrinology of Uterine Fibroids. Curr Opin Obstet Gynecol, 2015, 27(4):276–83., DOI 10.1097/gco.0000000000000185 Simon C. Introduction: Are We Advancing in Our Scientific Understanding and Therapeutic Improvement of Uterine Fibroids or Not. Fertil Steril, 2014, 102(3):611–2., DOI 10.1016/j.fertnstert.2014.07.1210 Mehine M.,Makinen N.,HeinonenH.-R.,Aaltonen L. A., Vahteristo P. Genomics of Uterine Leiomyomas: Insights from High-Throughput Sequencing. Fertil Sterility, 2014, 102(3):621–9., DOI 10.1016/j.fertnstert.2014.06.050 Giralt M., Cereijo R., Villarroya F. Adipokines and the Endocrine Role of Adipose Tissues. Handb Exp Pharmacol, 2016, 233:265–82., DOI 10.1007/164_2015_6 Tilg H, Moschen A. R. Adipocytokines: Mediators Linking Adipose Tissue, Inflammation and Immunity. Nat Rev Immunol, 2006, 6(10):772–83., DOI 10.1038/nri1937 Arita Y., Kihara S., Ouchi N., Takahashi M., Maeda K., Miyagawa J.-I., et al. Paradoxical Decrease of an Adipose-specific Protein, Adiponectin, in Obesity. Biochem Biophysical Res Commun, 1999, 257(1):79–83., DOI 10.1006/ bbrc.1999.0255 Hotta K., Funahashi T., Bodkin N. L., Ortmeyer H. K., Arita Y., Hansen B. C, et al. Circulating Concentrations of the Adipocyte Protein Adiponectin Are Decreased in Parallel with Reduced Insulin Sensitivity during the Progression to Type 2 Diabetes in Rhesus Monkeys. Diabetes, 2001, 50(5):1126–33., DOI 10.2337/diabetes.50.5.1126 Yamauchi T., Kamon J.,Waki H., Terauchi Y., Kubota N., Hara K., et al. The Fat- Derived Hormone Adiponectin Reverses Insulin Resistance Associated with Both Lipoatrophy and Obesity. Nat Med, 2001, 7(8):941–6., DOI 10.1038/90984 Soliman P. T., Wu D., Tortolero-Luna G., Schmeler K. M., Slomovitz B. M., Bray M. S., et al. Association between Adiponectin, Insulin Resistance, and Endometrial Cancer. Cancer, 2006, 106(11):2376–81., DOI 10.1002/cncr.21866 Petridou E., Mantzoros C., Dessypris N., Koukoulomatis P., Addy C., Voulgaris Z, et al. Plasma Adiponectin Concentrations in Relation to Endometrial Cancer: a Case-Control Study in Greece. J Clin Endocrinol Metab, 2003, 88(3):993–7., DOI 10.1210/jc.2002-021209 Rzepka-Gorska I., Bedner R., Cymbaluk-Ploska A., Chudecka-Glaz A. Serum Adiponectin in Relation to Endometrial Cancer and Endometrial Hyperplasia with Atypia in Obese Women. Eur J Gynaecol Oncol, 2008, 29(6):594–7. Cust A. E., Kaaks R., Friedenreich C., Bonnet F., Laville M., Lukanova A., et al. Plasma Adiponectin Levels and Endometrial Cancer Risk in Pre- and Postmenopausal Women. J Clin Endocrinol Metab, 2007, 92(1):255–63. Epub 2006 Oct 24., DOI 10.1210/jc.2006-1371 Ma Y., Liu Z., Zhang Y., Lu B. Serum Leptin, Adiponectin and Endometrial Cancer Risk in Chinese Women. J Gynecol Oncol, 2013, 24(4):336–41., DOI 10.3802/jgo.2013.24.4.336 Melistas L.,Mantzoros C. S., Kontogianni M., Antonopoulou S., Ordovas J.M., Yiannakouris N. Association of the +45T>G and +276G>T Polymorphisms in the Adiponectin Gene with Insulin Resistance in Nondiabetic Greek Women. Eur J Endocrinol, 2009, 161(6):845–52. Epub 2009 Sep 15. PMID: 19755407; PMCID: PMC2896503., DOI 10.1530/EJE-09-0492 Ukkola O., Ravussin E., Jacobson P., Sjostrom L., Bouchard C. Mutations in the Adiponectin Gene in Lean and Obese Subjects from the Swedish Obese Subjects Cohort. Metabolism, 2003, 52(7):881–4. PMID: 12870165., DOI 10.1016/s0026-0495(03)00074-x Salmenniemi U., Zacharova J., Ruotsalainen E., Vauhkonen I., Pihlajamaki J., Kainulainen S., et al. Association of Adiponectin Level and Variants in the Adiponectin Gene with Glucose Metabolism, Energy Expenditure, and Cytokines in Offspring of Type 2 Diabetic Patients. J Clin Endocrinol Metab, 2005, 90(7):4216–23. Epub 2005 Apr 26. PMID: 15855264., DOI 10.1210/jc.2004-2289 Zayani N., Omezzine A., Boumaiza I., Achour O., Rebhi L., Rejeb J., et al. Association of ADIPOQ, Leptin, LEPR, and Resistin Polymorphisms with Obesity Parameters in Hammam Sousse Sahloul Heart Study. J Clin Lab Anal, 2017, 31(6):e22148. Epub 2017 Feb 13., DOI 10.1002/jcla.22148 Christodoulou A., Ierodiakonou D., Awofala A. A., Petrou M., Kales S. N., Christiani D. C., et al. Variants in ADIPOQ Gene Are Linked to Adiponectin Levels and Lung Function in Young Males Independent of Obesity. PLoS One, 2020, 15(1):e0225662., DOI 10.1371/journal.pone.0225662 Kaur H., Badaruddoza B., Bains V., Kaur A. Genetic Association of ADIPOQ Gene Variants, -3971A>G and +276G>T, with Obesity and Metabolic Syndrome in North Indian Punjabi Population. PLoS One, 2018, 13(9): e0204502., DOI 10.1371/journal.pone.0204502 Filippi E., Sentinelli F., Trischitta V., Romeo S., Arca M., Leonetti F., et al. Association of the Human Adiponectin Gene and Insulin Resistance. Eur J Hum Genet, 2004, 12(3):199–205., DOI 10.1038/sj.ejhg.5201120 Xita N., Georgiou I., Chatzikyriakidou A., Vounatsou M., Papassotiriou G.-P., Papassotiriou I., et al. Effect of Adiponectin Gene Polymorphisms on Circulating Adiponectin and Insulin Resistance Indexes in Women with Polycystic Ovary Syndrome. Clin Chem, 2005, 51(2):416–23. Epub 2004 Dec 8. PMID: 15590747., DOI 10.1373/clinchem.2004.043109 Warodomwichit D., Shen J., Arnett D. K., Tsai M. Y., Kabagambe E. K., Peacock J. M., et al. ADIPOQ Polymorphisms, Monounsaturated Fatty Acids, and Obesity Risk: the GOLDN Study. Obesity, Silver Spring,, 2009, 17(3): 510–7., DOI 10.1038/oby.2008.583 National Center For Biotechnology Information, 2021). Short Genetic Variations. Available from: https://www.ncbi.nlm.nih.gov/snp/rs1501299. Cheung C. Y. Y., Hui E. Y. L., Cheung B. M. Y., Woo Y. C., Xu A., Fong C. H. Y., et al. Adiponectin Gene Variants and the Risk of Coronary Heart Disease: a 16-year Longitudinal Study. Eur J Endocrinol, 2014, 171(1):107–15. Epub 2014 Apr 23. PMID: 24760538., DOI 10.1530/EJE-14-0079 Bienkiewicz J., Smolarz B., Malinowski A. Association Between Single Nucleotide Polymorphism +276G > T, Rs1501299, in ADIPOQ and Endometrial Cancer. Pathol Oncol Res, 2016, 22(1):135–8. Epub 2015 Sep 19., DOI 10.1007/s12253-015-9985-9 Hong E. P., Park J.W. Sample Size and Statistical Power Calculation in Genetic Association Studies. Genomics Inform, 2012, 10(2):117–22. Epub 2012 Jun 30., DOI 10.5808/GI.2012.10.2.117 B-Rao C. Sample Size Considerations in Genetic Polymorphism Studies. Hum Hered, 2001, 52(4):191–200., DOI 10.1159/000053376 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1609966 EP 1609972 DI 10.3389/pore.2021.1609966 PG 7 ER PT J AU Li, Z Zhao, Z Wang, Ch Wang, D Mao, H Liu, F Yang, Y Tao, F Lu, Z AF Li, Zhiheng Zhao, Zhenhua Wang, Chuchu Wang, Dandan Mao, Haijia Liu, Fang Yang, Ye Tao, Feng Lu, Zengxin TI Association Between DCE-MRI Perfusion Histogram Parameters and EGFR and VEGF Expressions in Different Lauren Classifications of Advanced Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EGFR; VEGF; advanced gastric cancer; DCE-MRI; histogram parameters ID EGFR; VEGF; advanced gastric cancer; DCE-MRI; histogram parameters AB Objective: To investigate the correlations between dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) perfusion histogram parameters and vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) expressions in advanced gastric cancer (AGC). Methods: This retrospective study included 80 pathologically confirmed patients with AGC who underwent DCE-MRI before surgery from February 2017 to May 2021. The DCE-MRI perfusion histogram parameters were calculated by Omni Kinetics software in four quantitative parameter maps. Immunohistochemical methods were used to detect VEGF and EGFR expressions and calculate the immunohistochemical score. Results: VEGF expression was relatively lower in patients with intestinal-type AGC than those with diffuse-type AGC (p < 0.05). For VEGF, Receiver operating characteristics (ROC) curve analysis revealed that Quantile 90 of Ktrans, Meanvalue of Kep and Quantile 50 of Ve provided the perfect combination of sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV) for distinguishing high and low VEGF expression, For EGFR, Skewness of Ktrans, Energy of Kep and Entropy of Vp provided the perfect combination of sensitivity, specificity, PPV and NPV for distinguishing high and low EGFR expression. Ktrans (Quantile 90, Entropy) showed the strongest correlation with VEGF and EGFR in patients with intestinal-type AGC (r = 0.854 and r = 0.627, respectively); Ktrans (Mean value, Entropy) had the strongest correlation with VEGF and EGFR in patients with diffuse-type AGC (r = 0.635 and 0.656, respectively). Conclusion: DCE-MRI perfusion histogram parameters can serve as imaging biomarkers to reflect VEGF and EGFR expressions and estimate their difference in different Lauren classifications of AGC. C1 [Li, Zhiheng] Shaoxing University School of MedicineShaoxing, China. [Zhao, Zhenhua] Zhejiang University School of Medicine, Shaoxing People’s Hospital, Shaoxing Hospital, Department of RadiologyShaoxing, China. [Wang, Chuchu] Shaoxing University School of MedicineShaoxing, China. [Wang, Dandan] Zhejiang University School of Medicine, Shaoxing People’s Hospital, Shaoxing Hospital, Department of RadiologyShaoxing, China. [Mao, Haijia] Zhejiang University School of Medicine, Shaoxing People’s Hospital, Shaoxing Hospital, Department of RadiologyShaoxing, China. [Liu, Fang] Zhejiang University School of Medicine, Shaoxing People’s Hospital, Shaoxing Hospital, Department of PathologyShaoxing, China. [Yang, Ye] Zhejiang University School of Medicine, Shaoxing People’s Hospital, Shaoxing Hospital, Department of PathologyShaoxing, China. [Tao, Feng] Zhejiang University School of Medicine, Shaoxing People’s Hospital, Shaoxing Hospital, Department of Gastrointestinal SurgeryShaoxing, China. [Lu, Zengxin] Zhejiang University School of Medicine, Shaoxing People’s Hospital, Shaoxing Hospital, Department of RadiologyShaoxing, China. RP Lu, Z (reprint author), Zhejiang University School of Medicine, Shaoxing People’s Hospital, Shaoxing Hospital, Department of Radiology, Shaoxing, China. EM luzx777@163.com CR Smyth EC,NilssonM, GrabschHI, vanGriekenNC, Lordick F. GastricCancer. The Lancet, 2020, 396(10251):635–48., DOI 10.1016/S0140-6736(20)31288-5 Song H, Zhu J, Lu D. Molecular-Targeted First-Line Therapy for Advanced Gastric Cancer. Cochrane Database Syst Rev, 2016, 2016:CD011461., DOI 10.1002/14651858.CD011461.pub2 Chen Z, Li Y, Tan B, Zhao Q, Fan L, Li F, et al. Progress and Current Status of Molecule-Targeted Therapy and Drug Resistance in Gastric Cancer. Drugs Today, 2020, 56(7):469–82., DOI 10.1358/dot.2020.56.7.3112071 Watari J, Chen N, Amenta PS, Fukui H, Oshima T, Tomita T, et al. Helicobacter Pylori Associated Chronic Gastritis, Clinical Syndromes, Precancerous Lesions, and Pathogenesis of Gastric Cancer Development. Wjg, 2014, 20(18):5461–73., DOI 10.3748/wjg.v20.i18.5461 Seshacharyulu P, Ponnusamy MP, Haridas D, Jain M, Ganti AK, Batra SK Targeting the EGFR Signaling Pathway in Cancer Therapy. Expert Opin Ther Targets, 2012, 16(1):15–31., DOI 10.1517/14728222.2011.648617 Li X, Zhu X, Wang Y, Wang R, Wang L, Zhu ML, et al. Prognostic Value and Association of Lauren Classification with VEGF and VEGFR-2 Expression in Gastric Cancer. Oncol Lett, 2019, 18(5):4891–9., DOI 10.3892/ol.2019.10820 Ma L, Xu X, Zhang M, Zheng S, Zhang B, Zhang W, et al. Dynamic Contrast- Enhanced MRI of Gastric Cancer: Correlations of the Pharmacokinetic Parameters with Histological Type, Lauren Classification, and Angiogenesis. Magn Reson Imaging, 2017, 37:27–32., DOI 10.1016/ j.mri.2016.11.004 Meyer HJ, Leifels L, Hamerla G, Hohn AK, Surov A Associations between Histogram Analysis Parameters Derived from DCE-MRI and Histopathological Features Including Expression of EGFR, P16, VEGF, Hif1-Alpha, and P53 in HNSCC. Contrast Media Mol Imaging, 2019, 2019:1–10., DOI 10.1155/2019/5081909 Liang J, Cheng Q, Huang J, Ma M, Zhang D, Lei X, et al. Monitoring Tumour Microenvironment Changes during Anti-angiogenesis Therapy Using Functional MRI. Angiogenesis, 2019, 22(3):457–70., DOI 10.1007/s10456- 019-09670-4 Zhu Y, Zhou Y, Zhang W, Xue L, Li Y, Jiang J, et al. Value of Quantitative Dynamic Contrast-Enhanced and Diffusion-Weighted Magnetic Resonance Imaging in Predicting Extramural Venous Invasion in Locally Advanced Gastric Cancer and Prognostic Significance. Quant Imaging Med Surg, 2021, 11(1):328–40., DOI 10.21037/qims-20-246 Just N Improving Tumour Heterogeneity MRI Assessment with Histograms. Br J Cancer, 2014, 111(12):2205–13., DOI 10.1038/bjc.2014.512 Liu W, Wei C, Bai J, Gao X, Zhou L Histogram Analysis of Diffusion Kurtosis Imaging in the Differentiation of Malignant from Benign Breast Lesions. Eur J Radiol, 2019, 117:156–63., DOI 10.1016/ j.ejrad.2019.06.008 Tofts PS, Brix G, Buckley DL, Evelhoch JL, Henderson E, Knopp MV, et al. Estimating Kinetic Parameters from Dynamic Contrast-Enhanced T(1)- weighted MRI of a Diffusable Tracer: Standardized Quantities and Symbols. J Magn Reson Imaging, 1999, 10(3):223–32., DOI 10.1002/(sici, 1522-2586(199909)10:3<223:aid-jmri2>3.0.co;2-s Ilie MD, Vasiljevic A, Louvet C, Jouanneau E, Raverot G Gonadotroph Tumors Show Subtype Differences that Might Have Implications for Therapy. Cancers, 2020, 12(4):1012., DOI 10.3390/cancers12041012 Lauren P The Two Histological Main Types of Gastric Carcinoma: Diffuse and So-Called Intestinal-type Carcinoma. An Attempt at a Histo-Clinical Classification. Acta Pathol Microbiol Scand, 1965, 64:31–49., DOI 10.1111/ apm.1965.64.1.31 Pyo JH, Lee H, Min B-H, Lee JH, Choi MG, Lee JH, et al. Early Gastric Cancer with a Mixed-type Lauren Classification Is More Aggressive and Exhibits Greater Lymph Node Metastasis. J Gastroenterol, 2017, 52(5):594–601., DOI 10.1007/s00535-016-1254-5 Ma J, Shen H, Kapesa L, Zeng S Lauren Classification and Individualized Chemotherapy in Gastric Cancer. Oncol Lett, 2016, 11(5):2959–64., DOI 10.3892/ol.2016.4337 Bergamino M, Bonzano L, Levrero F, Mancardi GL, Roccatagliata L A Review of Technical Aspects of T1-Weighted Dynamic Contrast-Enhanced Magnetic Resonance Imaging, DCE-MRI, in Human Brain Tumors. Physica Med, 2014, 30(6):635–43., DOI 10.1016/j.ejmp.2014.04.005 Sun N-N, Ge X-L, Liu X-S, Xu L-L Histogram Analysis of DCE-MRI for Chemoradiotherapy Response Evaluation in Locally Advanced Esophageal Squamous Cell Carcinoma. Radiol Med, 2020, 125(2):165–76., DOI 10.1007/ s11547-019-01081-1 Gillies RJ, Kinahan PE, Hricak H Radiomics: Images Are More Than Pictures, They Are Data. Radiology, 2016, 278(2):563–77., DOI 10.1148/ radiol.2015151169 Murayama K, Nishiyama Y, Hirose Y, Abe M, Ohyu S, Ninomiya A, et al. Differentiating between Central Nervous System Lymphoma and High-Grade Glioma Using Dynamic Susceptibility Contrast and Dynamic Contrast- Enhanced MR Imaging with Histogram Analysis. Magn Reson Med Sci, 2018, 17(1):42–9., DOI 10.2463/mrms.mp.2016-0113 Turkbey B, Thomasson D, Pang Y, Bernardo M, Choyke PL The Role of Dynamic Contrast-Enhanced MRI in Cancer Diagnosis and Treatment. Diagn Interv Radiol, 2009, 16(3):186–92., DOI 10.4261/1305- 3825.DIR.2537-08.1 Siveen KS, Prabhu K, Krishnankutty R, Kuttikrishnan S, Tsakou M, Alali FQ, et al. Vascular Endothelial Growth Factor, VEGF, Signaling in Tumour Vascularization: Potential and Challenges. Cvp, 2017, 15(4):339–51., DOI 10.2174/1570161115666170105124038 Hu Y, Chen Y, Wang J, Kang JJ, Shen DD, Jia ZZ Non-Invasive Estimation of Glioma IDH1 Mutation and VEGF Expression by Histogram Analysis of Dynamic Contrast-Enhanced MRI. Front Oncol, 2020, 10:593102., DOI 10.3389/fonc.2020.593102 Di N, Yao C, Cheng W, Ren Y, Qu J, Wang B, et al. Correlation of Dynamic Contrast-Enhanced MRI Derived Volume Transfer Constant with Histological Angiogenic Markers in High-Grade Gliomas. J Med Imaging Radiat Oncol, 2018, 62:464–70., DOI 10.1111/1754-9485.12701 Chen Y-C, Fang W-L, Wang R-F, Liu C-A, Yang M-H, Lo S-S, et al. Clinicopathological Variation of Lauren Classification in Gastric Cancer. Pathol Oncol Res, 2016, 22(1):197–202., DOI 10.1007/s12253-015-9996-6 Wang X, Cao W, Mo M, Wang W, Wu H, Wang J J. VEGF and Cortactin Expression Are Independent Predictors of Tumor Recurrence Following Curative Resection of Gastric Cancer. J Surg Oncol, 2010, 102(4):325–30., DOI 10.1002/jso.21644 Chen C-N, Hsieh F-J, Cheng Y-M, Cheng W-F, Su Y-N, Chang K-J, et al. The Significance of Placenta Growth Factor in Angiogenesis and Clinical Outcome of Human Gastric Cancer. Cancer Lett, 2004, 213(1):73–82., DOI 10.1016/ j.canlet.2004.05.020 Galizia G, Lieto E, Orditura M, Castellano P, Mura AL, Imperatore V, et al. Epidermal Growth Factor Receptor, EGFR, Expression Is Associated with a Worse Prognosis in Gastric Cancer Patients Undergoing Curative Surgery. World J Surg, 2007, 31(7):1458–68., DOI 10.1007/s00268-007-9016-4 Wang X, Fu R, Hu Y, Du H, Li S, Li Z, et al. EGFR Gene Status Predicts Response and Survival Benefit in a Preclinical Gastric Cancer Trial Treating Patientderived Xenografts with Cetuximab. Oncol Rep, 2017, 38(4):2387–93., DOI 10.3892/or.2017.5907 Wang KL, Wu T-T, Choi IS, Wang H, Resetkova E, Correa AM, et al. Expression of Epidermal Growth Factor Receptor in Esophageal and Esophagogastric Junction Adenocarcinomas: Association with Poor Outcome. Cancer, 2007, 109(4):658–67., DOI 10.1002/cncr.22445 Surov A, Meyer HJ, Gawlitza M, Hohn A-K, Boehm A, Kahn T, et al. Correlations between DCE MRI and Histopathological Parameters in Head and Neck Squamous Cell Carcinoma. Translational Oncol, 2017, 10(1):17–21., DOI 10.1016/j.tranon.2016.10.001 Zhang Y, Li Z, Gao C, Shen J, Chen M, Liu Y, et al. Preoperative Histogram Parameters of Dynamic Contrast-Enhanced MRI as a Potential Imaging Biomarker for Assessing the Expression of Ki-67 in Prostate Cancer. Cancer Med, 2021, 10:4240–9., DOI 10.1002/cam4.3912 Ye W, Li Y, Fan L, Zhao Q, Yuan H, Tan B, et al. Annexin A7 Expression Is Downregulated in Late-Stage Gastric Cancer and Is Negatively Correlated with the Differentiation Grade and Apoptosis Rate. Oncol Lett, 2018, 15(6): 9836–44., DOI 10.3892/ol.2018.8576 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610001 EP 1610012 DI 10.3389/pore.2021.1610001 PG 12 ER PT J AU Deme, D Kovacs, S Telekes, A AF Deme, Daniel Kovacs, Sandor Telekes, Andras TI Overall Survival Prediction of Advanced Cancer Patients by Selection of the Most Significant Baseline Serum Biomarker Combination SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE overall survival; advanced cancer; serum biomarkers; prognostic importance; CRP; albumin; PLR ID overall survival; advanced cancer; serum biomarkers; prognostic importance; CRP; albumin; PLR AB Introduction: Consistent association between elevated baseline serumvalues and C-reactive protein (CRP), cross-linked fibrin degradation products (D-dimer), lactate dehydrogenase (LDH), decreased baseline serum albumin, absolute lymphocyte count to absolute monocyte count ratio (LMR), elevated absolute neutrophil count to absolute lymphocyte count ratio (NLR), elevated platelet count to absolute lymphocyte count ratio (PLR), and between some combinations of these biomarkers and the short overall survival of patients with malignant diseases has already been reported. These biomarkers are independent prognostic factors for cancer. Here, the most significant biomarker combination of these values was searched and studied in real-life advanced cancer patients of a single center. Methods: The authors retrospectively analyzed the association of the aforementioned biomarkers and their combination and OS of 75 consecutive cancer patients with locally advanced, recurrent, or metastatic diseases. Validated cut-off determination was used. Results: CRP, albumin, and PLR showed marked association with OS. Cut-off values for significant shorter OS were 30.65 mg/L (p < 0.001), 44.35 g/L (p < 0.001), and 168.20 (p < 0.001), respectively. Based on assessed biomarker cut-offs, four patient groups were created to determine whether biomarker values were out of range (ORV) compared to cutoff: 1) No ORV biomarkers (n = 24; OS = 26.07 months); 2) one ORV biomarker (n = 21; OS = 13.50 months); 3) two ORV biomarkers (n = 20; OS = 7.97 months), and 4) three ORV biomarkers (n = 10; OS = 3.91 months). Significant differences in OS were detected between the groups: For 1. vs. 2. hazard ratio (HR) = 3.0 (95% CI: 1.5–6.2), p = 0.003; for 1. vs. 3. HR = 4.1 (95% CI: 2.0–8.3), p < 0.001; and for 1. vs. 4. HR = 10.2 (95% CI: 4.2–24.6), p < 0.001. Conclusion: Based on our analysis, we can confirm that the complex monitoring of CRP, albumin, and PLR would provide a good estimation of OS. Large scale prospective studies are warranted to explore this and other useful combinations of prognostic biomarkers and their relationship to the well-established prognostic systems in real-life. C1 [Deme, Daniel] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. [Kovacs, Sandor] University of Debrecen, Department of Economical and Financial MathematicsDebrecen, Hungary. [Telekes, Andras] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. RP Deme, D (reprint author), Szent Lazar Megyei Korhaz, Onkologiai Osztaly, Salgotarjan, Hungary. EM danieldeme_md@ymail.com CR Nixon AB, Pang H, Starr MD, Friedman PN, Bertagnolli MM, Kindler HL, et al. Prognostic and Predictive Blood-Based Biomarkers in Patients with Advanced Pancreatic Cancer: Results from CALGB80303, Alliance). Clin Cancer Res, 2013, 19:6957–66., DOI 10.1158/1078-0432.CCR-13-0926 Nakagawa K, Tanaka K, Nojiri K, Kumamoto T, Takeda K, Ueda M, et al. The Modified Glasgow Prognostic Score as a Predictor of Survival after Hepatectomy for Colorectal Liver Metastases. Ann Surg Oncol, 2014, 21: 1711–8., DOI 10.1245/s10434-013-3342-6 Liao DW, Hu X, Wang Y, Yang ZQ, Li X. C-reactive Protein Is a Predictor of Prognosis of Prostate Cancer: A Systematic Review and Meta-Analysis. Ann Clin Lab Sci, 2020, 50:161–71. Chen Y, Cong R, Ji C, Ruan W. The Prognostic Role of C-reactive Protein in Patients with Head and Neck Squamous Cell Carcinoma: A Meta-analysis. Cancer Med, 2020, 9:9541–53., DOI 10.1002/cam4.3520 Wang Y, Wang Z. Predictive Value of Plasma D-Dimer Levels in Patients with Advanced Non-small-cell Lung Cancer. Ott, 2015, 8:805–8., DOI 10.2147/OTT. S78154 Liu P, Zhu y., Liu l. Elevated Pretreatment Plasma D-Dimer Levels And platelet Counts Predict Poor Prognosis In pancreatic Adenocarcinoma. Ott, 2015, 8:1335–40., DOI 10.2147/OTT.S82329 Wu J, Fu Z, Liu G, Xu P, Xu J, Jia X. Clinical Significance of Plasma D-Dimer in Ovarian Cancer. Medicine, Baltimore,, 2017, 96:e7062., DOI 10.1097/MD. 0000000000007062 Ma M, Cao R,Wang W, Wang B, Yang Y, Huang Y, et al. The D-Dimer Level Predicts the Prognosis in Patients with Lung Cancer: a Systematic Review and Meta-Analysis. J Cardiothorac Surg, 2021, 16:243., DOI 10.1186/s13019-021- 01618-4 Ji F, Fu S-J, Guo Z-Y, Pang H, Ju W-Q, Wang D-P, et al. Prognostic Value of Combined Preoperative Lactate Dehydrogenase and Alkaline Phosphatase Levels in Patients with Resectable Pancreatic Ductal Adenocarcinoma. Medicine, Baltimore,, 2016, 95:e4065., DOI 10.1097/MD.0000000000004065 Liu R, Cao J, Gao X, Zhang J,Wang L, Wang B, et al. Overall Survival of Cancer Patients with Serum Lactate Dehydrogenase Greater Than 1000 IU/L. Tumor Biol, 2016, 37:14083–8., DOI 10.1007/s13277-016-5228-2 Li F, Xiang H, Pang Z, Chen Z, Dai J, Chen S, et al. Association between Lactate Dehydrogenase Levels and Oncologic Outcomes in Metastatic Prostate Cancer: A Meta-analysis. Cancer Med, 2020, 9:7341–51., DOI 10.1002/cam4.3108 Deng T, Zhang J, Meng Y, Zhou Y, Li W. Higher Pretreatment Lactate Dehydrogenase Concentration Predicts Worse Overall Survival in Patients with Lung Cancer. Medicine, Baltimore,, 2018, 97:e12524., DOI 10.1097/MD. 0000000000012524 Win T, Sharples L, Groves AM, Ritchie AJ,Wells FC, Laroche CM. Predicting Survival in Potentially Curable Lung Cancer Patients. Lung, 2008, 186:97–102., DOI 10.1007/s00408-007-9067-1 Shibutani M, Maeda K, Nagahara H, Iseki Y, Ikeya T, Hirakawa K. Prognostic Significance of the Preoperative Ratio of C-Reactive Protein to Albumin in Patients with Colorectal Cancer. Anticancer Res, 2016, 36:995–1001. Liu J, Wang F, Li S, Huang W, Jia Y, Wei C. The Prognostic Significance of Preoperative Serum Albumin in Urothelial Carcinoma: a Systematic Review and Meta-Analysis. Biosci Rep, 2018, 38:BSR20180214., DOI 10.1042/ BSR20180214 Deme D, Telekes A. A C-Reaktiv Protein, CRP, Plazmaszintjenek Prognosztikai Jelentosege Az Onkologiaban. Orvosi Hetilap, 2017, 158: 243–56. Hungarian., DOI 10.1556/650.2017.30646 Deme D, Telekes A. Prognostic Importance of Cross-Linked Fibrin Degradation Products, D-Dimer, in Oncology. Magy Onkol, 2017, 61: 319–26. Hungarian. Deme D, Telekes A. A Laktatdehidrogenaz, LDH, Prognosztikai Jelentosege Az Onkologiaban. Orvosi Hetilap, 2017, 158:1977–88. Hungarian., DOI 10. 1556/650.2017.30890 Deme D, Telekes A. Az Albumin Prognosztikai Jelentosege Az Onkologiaban. Orvosi Hetilap, 2018, 159:96–106. Hungarian., DOI 10.1556/650.2018.30885 Song L, Zhu J, Li Z, Wei T, Gong R, Lei J. The Prognostic Value of the Lymphocyte-To-Monocyte Ratio for High-Risk Papillary Thyroid Carcinoma. Cmar, 2019, Vol. 11:8451–62., DOI 10.2147/CMAR.S219163 Chen X-Q, Xue C-R, Hou P, Lin B-Q, Zhang J-R. Lymphocyte-tomonocyte Ratio Effectively Predicts Survival Outcome of Patients with Obstructive Colorectal Cancer. Wjg, 2019, 25:4970–84., DOI 10.3748/wjg. v25.i33.4970 Song Q, Wu J-z., Wang S. Low Preoperative Lymphocyte to Monocyte Ratio Serves as a Worse Prognostic Marker in Patients with Esophageal Squamous Cell Carcinoma Undergoing Curative Tumor Resection. J Cancer, 2019, 10: 2057–62., DOI 10.7150/jca.29383 Lu C, Zhou L, Ouyang J, Yang H. Prognostic Value of Lymphocyte-To- Monocyte Ratio in Ovarian Cancer. Medicine, Baltimore,, 2019, 98:e15876., DOI 10.1097/MD.0000000000015876 Zhou W, Kuang T, Han X, Chen W, Xu X, LouW, et al. Prognostic Role of Lymphocyte-To-Monocyte Ratio in Pancreatic Neuroendocrine Neoplasms. Endocr Connect, 2020, 9:289–98. pii: EC-19-0541.R1., DOI 10.1530/EC-19-0541 Templeton AJ, McNamara MG, Seruga B, Vera-Badillo FE, Aneja P, Ocana A, et al. Prognostic Role of Neutrophil-To-Lymphocyte Ratio in Solid Tumors: a Systematic Review and Meta-Analysis. J Natl Cancer Inst, 2014, 106(6):dju124., DOI 10.1093/jnci/dju124 Yang Y, Liu R, Ren F, Guo R, Zhang P. Prognostic and Clinicopathological Significance of Neutrophil-To-Lymphocyte Ratio in Patients with Oral Cancer. Biosci Rep, 2018, 38:BSR20181550. pii., DOI 10.1042/BSR20181550 Chen G, Zhu L, Yang Y, Long Y, Li X, Wang Y. Prognostic Role of Neutrophil to Lymphocyte Ratio in Ovarian Cancer: AMeta-Analysis. Technol Cancer Res Treat, 2018, 17:153303381879150., DOI 10.1177/1533033818791500 Liu G, Ke L-c., Sun S-R. Prognostic Value of Pretreatment Neutrophil-To- Lymphocyte Ratio in Patients with Soft Tissue Sarcoma. Medicine, Baltimore,, 2018, 97:e12176., DOI 10.1097/MD.0000000000012176 Pirozzolo G, Gisbertz SS, Castoro C, van Berge Henegouwen MI, Scarpa M. Neutrophil-to-lymphocyte Ratio as Prognostic Marker in Esophageal Cancer: a Systematic Review and Meta-Analysis. J Thorac Dis, 2019, 11:3136–45., DOI 10.21037/jtd.2019.07.30 Wang Z, Zhan P, Lv Y, Shen K, Wei Y, Liu H, et al. Prognostic Role of Pretreatment Neutrophil-To-Lymphocyte Ratio in Non-small Cell Lung Cancer Patients Treated with Systemic Therapy: a Meta-Analysis. Transl Lung Cancer Res, 2019, 8:214–26., DOI 10.21037/tlcr.2019.06.10 Ding Y, Zhang S, Qiao J. Prognostic Value of Neutrophil-To-Lymphocyte Ratio in Melanoma. Medicine, Baltimore,, 2018, 97:e11446., DOI 10.1097/MD. 0000000000011446 Maretty-Nielsen K. Prognostic Factors in Soft Tissue Sarcoma. Dan Med J, 2014, 61:B4957. Cho J-K, Kim MW, Choi IS, Moon UY, KimM-J, Sohn I, et al. Optimal Cutoff of Pretreatment Neutrophil-To-Lymphocyte Ratio in Head and Neck Cancer Patients: a Meta-Analysis and Validation Study. BMC Cancer, 2018, 18:969., DOI 10.1186/s12885-018-4876-6 Tian C, Song W, Tian X, Sun Y. Prognostic Significance of Platelet-To- Lymphocyte Ratio in Patients with Ovarian Cancer: A Meta-Analysis. Eur J Clin Invest, 2018, 48(5):e12917., DOI 10.1111/eci.12917 Wang J, Zhou X, He Y, Chen X, Liu N, Ding Z, et al. Prognostic Role of Platelet to Lymphocyte Ratio in Prostate Cancer. Medicine, Baltimore,, 2018, 97(40): e12504., DOI 10.1097/MD.0000000000012504 CaoW, Yao X, Cen D, Zhi Y, Zhu N, Xu L. The Prognostic Role of Platelet-To- Lymphocyte Ratio on Overall Survival in Gastric Cancer: a Systematic Review and Meta-Analysis. BMC Gastroenterol, 2020, 20:16., DOI 10.1186/s12876-020- 1167-x Bardash Y, Olson C, Herman W, Khaymovich J, Costantino P, Tham T. Platelet-Lymphocyte Ratio as a Predictor of Prognosis in Head and Neck Cancer: A Systematic Review and Meta-Analysis. Oncol Res Treat, 2019, 42: 665–77., DOI 10.1159/000502750 Wang X, Ni X, Tang G. Prognostic Role of Platelet-To-Lymphocyte Ratio in Patients with Bladder Cancer: A Meta-Analysis. Front Oncol, 2019, 9:757., DOI 10.3389/fonc.2019.00757 Zhang M, Huang X-z., Song Y-x., Gao P, Sun J-x., Wang Z-N. High Platelet- To-Lymphocyte Ratio Predicts Poor Prognosis and Clinicopathological Characteristics in Patients with Breast Cancer: A Meta-Analysis. Biomed Res Int, 2017, 2017:1–11. Article ID 9503025., DOI 10.1155/2017/9503025 Xu W, Wang W, Yang M, Song L, Xiong J, Lin J, et al. Prognostic Significance of the Platelet-To-Lymphocyte Ratio in Ovarian Cancer: a Meta-Analysis. Translational Cancer Res, 2018, 7:552–60., DOI 10.21037/21627 Song W, Tian C, Wang K, Zhang R-J, Zou S-B. Preoperative Platelet Lymphocyte Ratio as Independent Predictors of Prognosis in Pancreatic Cancer: A Systematic Review and Meta-Analysis. PLOS ONE, 2017, 12: e0178762., DOI 10.1371/journal.pone.0178762 You J, Zhu G-Q, Xie L, Liu W-Y, Shi L,Wang O-C, et al. Preoperative Platelet to Lymphocyte Ratio Is a Valuable Prognostic Biomarker in Patients with Colorectal Cancer. Oncotarget, 2016, 7:25516–27., DOI 10.18632/oncotarget. 8334 Zhang X, Wang Y, Zhao L, Sang S, Zhang L. Prognostic Value of Platelet-To- Lymphocyte Ratio in Oncologic Outcomes of Esophageal Cancer: A Systematic Review and Meta-Analysis. Int J Biol Markers, 2018, 33:335–44., DOI 10.1177/1724600818766889 Lim JU, Yeo CD, Kang HS, Park CK, Kim JS, Kim JW, et al. Elevated Pretreatment Platelet-To-Lymphocyte Ratio Is Associated with Poor Survival in Stage IV Non-small Cell Lung Cancer with Malignant Pleural Effusion. Sci Rep, 2019, 9:4721., DOI 10.1038/s41598-019-41289-9 Lee Y, Kim YW, Park DK, Hwang IC. Inverse Association between Platelet- Lymphocyte Ratio and Prognosis in Terminally Ill Cancer Patients: A Preliminary Study. J Palliat Med, 2017, 20:533–7., DOI 10.1089/jpm.2016.0338 Budczies J, Klauschen F, Sinn BV, Gyorffy B, Schmitt WD, Darb-Esfahani S, et al. Cutoff Finder: a Comprehensive and Straightforward Web Application Enabling Rapid Biomarker Cutoff Optimization. PLoS One, 2012, 7:e51862. Online Cutoff Finder tool is available at https://molpathoheidelberg.shinyapps. io/CutoffFinder_v1/, Accessed September 18, 2021)., DOI 10.1371/journal. pone.0051862 R Core Team. R: A Language and Environment for Statistical Computing. Vienna: R Foundation for Statistical Computing, 2019). Available at: https:// www.R-project.org., Accessed July 11, 2021). Shuster JJ. Median Follow-Up in Clinical Trials. Jco, 1991, 9:191–2., DOI 10. 1200/JCO.1991.9.1.191 Greten FR, Grivennikov SI. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity, 2019, 51:27–41., DOI 10.1016/j.immuni.2019. 06.025 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610004 EP 1610012 DI 10.3389/pore.2022.1610004 PG 9 ER PT J AU Yao, JM Zhao, JY Lv, FF Yang, XB Wang, HJ AF Yao, Jin-Ming Zhao, Jun-Yu Lv, Fang-Fang Yang, Xue-Bo Wang, Huan-Jun TI A Potential Nine-lncRNAs Signature Identification and Nomogram Diagnostic Model Establishment for Papillary Thyroid Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE risk score; WGCNA; papillary thyroid cancer; lncRNAs; LASSO ID risk score; WGCNA; papillary thyroid cancer; lncRNAs; LASSO AB The purpose of our current study was to establish a long non-coding RNA(lncRNA) signature and assess its prognostic and diagnostic power in papillary thyroid cancer (PTC). LncRNA expression profiles were obtained from the Cancer Genome Atlas (TCGA). The key module and hub lncRNAs related to PTC were determined by weighted gene coexpression network analysis (WGCNA) and LASSO Cox regression analyses, respectively. Functional enrichment analyses, including Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene set enrichment analysis were implemented to analyze the possible biological processes and signaling pathways of hub lncRNAs. Associations between key lncRNA expressions and tumor-infiltrating immune cells were identified using the TIMER website, and proportions of immune cells in high/low risk score groups were compared. Kaplan-Meier Plotter was used to evaluate the prognostic significance of hub genes in PTC. A diagnostic model was conducted with logistic regression analysis, and its diagnostic performance was assessed by calibration/ receiver operating characteristic curves and principal component analysis. A nine-lncRNAs signature (SLC12A5-AS1, LINC02028, KIZ-AS1, LINC02019, LINC01877, LINC01444, LINC01176, LINC01290, and LINC00581) was established in PTC, which has significant diagnostic and prognostic power. Functional enrichment analyses elucidated the regulatory mechanism of the nine-lncRNAs signature in the development of PTC. This signature and expressions of nine hub lncRNAs were correlated with the distributions of tumor infiltrating immune cells. A diagnostic nomogram was also established for PTC. By comparing with the published models with less than or equal to nine lncRNAs, our signature showed a preferable performace for prognosis prediction. In conclusion, our present research established an innovative nine-lncRNAs signature and a six-lncRNAs nomogram that might act as a potential indicator for PTC prognosis and diagnosis, which could be conducive to the PTC treatment. C1 [Yao, Jin-Ming] The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Department of Endocrinology and MetabologyJinan, China. [Zhao, Jun-Yu] The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Department of Endocrinology and MetabologyJinan, China. [Lv, Fang-Fang] The 960th hospital of the PLA Joint Logistics Support Force, Department of Endocrinology and MetabologyJinan, China. [Yang, Xue-Bo] Beijing Splinger Institute of MedicineJinan, China. [Wang, Huan-Jun] The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Department of Endocrinology and MetabologyJinan, China. RP Wang, HJ (reprint author), The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Department of Endocrinology and Metabology, Jinan, China. EM huanjun-w@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: a Cancer J clinicians, 2018, 68:394–424., DOI 10.3322/caac.21492 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Fagin JA, Wells SA. Biologic and Clinical Perspectives on Thyroid Cancer. N Engl J Med, 2016, 375:1054–67., DOI 10.1056/nejmra1501993 Singer PA, Cooper DS, Daniels GH, Ladenson PW, Greenspan FS, Levy EG, et al. Treatment Guidelines for Patients with Thyroid Nodules and Well-Differentiated Thyroid Cancer. American Thyroid Association. Am Thyroid Assoc Arch Intern Med, 1996, 156:2165–72., DOI 10.1001/ archinte.156.19.2165 Mirian C, Gronhoj C, Jensen DH, Jakobsen KK, Karnov K, Jensen JS, et al. Trends in Thyroid Cancer: Retrospective Analysis of Incidence and Survival in Denmark 1980-2014. Cancer Epidemiol, 2018, 55:81–7., DOI 10.1016/j.canep. 2018.05.009 Clarke CA, Reynolds P, Oakley-Girvan I, Lee E, Lu Y, Yang J, et al. Indicators of Microbial-Rich Environments and the Development of Papillary Thyroid Cancer in the California Teachers Study. Cancer Epidemiol, 2015, 39:548–53., DOI 10.1016/j.canep.2015.04.014 Toniato A, Boschin I, Casara D, Mazzarotto R, Rubello D, Pelizzo M. Papillary Thyroid Carcinoma: Factors Influencing Recurrence and Survival. Ann Surg Oncol, 2008, 15:1518–22., DOI 10.1245/s10434-008-9859-4 Garzon R, Calin GA, Croce CM. MicroRNAs in Cancer. Annu Rev Med, 2009, 60:167–79., DOI 10.1146/annurev.med.59.053006.104707 Jae N, Heumuller AW, Fouani Y, Dimmeler S. Long Non-coding RNAs in Vascular Biology and Disease. Vasc Pharmacol, 2019, 114:13–22., DOI 10.1016/ j.vph.2018.03.003 Rinn JL, Chang HY. Genome Regulation by Long Noncoding RNAs. Annu Rev Biochem, 2012, 81:145–66., DOI 10.1146/annurev-biochem-051410-092902 Mercer TR, Dinger ME, Mattick JS. Long Non-coding RNAs: Insights into Functions. Nat Rev Genet, 2009, 10:155–9., DOI 10.1038/nrg2521 Wilusz JE, Sunwoo H, Spector DL. Long Noncoding RNAs: Functional Surprises from the RNA World. Genes Dev, 2009, 23:1494–504., DOI 10. 1101/gad.1800909 Spizzo R, Almeida MI, Colombatti A, Calin GA. Long Non-coding RNAs and Cancer: a New Frontier of Translational Research. Oncogene, 2012, 31: 4577–87., DOI 10.1038/onc.2011.621 Fatica A, Bozzoni I. Long Non-coding RNAs: New Players in Cell Differentiation and Development. Nat Rev Genet, 2014, 15:7–21., DOI 10. 1038/nrg3606 Kornienko AE, Guenzl PM, Barlow DP, Pauler FM. Gene Regulation by the Act of Long Non-coding RNA Transcription. BMC Biol, 2013, 11:59., DOI 10. 1186/1741-7007-11-59 Lei H, Gao Y, Xu X. LncRNA TUG1 Influences Papillary Thyroid Cancer Cell Proliferation, Migration and EMT Formation through Targeting miR-145. Acta Biochim Biophys Sinica, 2017, 49:588–97., DOI 10.1093/abbs/gmx047 Gu Y, Feng C, Liu T, Zhang B, Yang L The Downregulation of lncRNA EMX2OS Might Independently Predict Shorter Recurrence-free Survival of Classical Papillary Thyroid Cancer. PLoS ONE, 2018). 13:e0209338., DOI 10. 1371/journal.pone.0209338 Wang Z, Cheng Y, Zhao D, Pliss A, Liu J, Luan P. Synergic Treatment of Alzheimer’s Disease with Brain Targeted Nanoparticles Incorporating NgRsiRNA and Brain Derived Neurotrophic Factor. Smart Mater Med, 2020, 1: 125–30., DOI 10.1016/j.smaim.2020.08.001 Chen F, Yin S, Zhu J, Liu P, Yang C, Feng Z, et al. lncRNA DGCR5 Acts as a Tumor Suppressor in Papillary Thyroid Carcinoma via Sequestering miR- 2861. Exp Ther Med, 2019, 17:895–900., DOI 10.3892/etm.2018.7012 Liu K, Kang M, Liao X, Wang R. Genome-wide Investigation of the Clinical Significance and Prospective Molecular Mechanism of Minichromosome Maintenance Protein Family Genes in Patients with Lung Adenocarcinoma. PLoS ONE, 2019, 14:e0219467., DOI 10.1371/journal.pone.0219467 Subbannayya Y, Di Fiore R. The Role of Omics Approaches to Characterize Molecular Mechanisms of Rare Ovarian Cancers. Recent Adv Future Perspect, 2021, 9., DOI 10.3390/biomedicines9101481 Tang J, Cui Q, Zhang D, Kong D, Liao X, Ren J, et al. A Prognostic eightlncRNA Expression Signature in Predicting Recurrence of ER-positive Breast Cancer Receiving Endocrine Therapy. J Cel Physiol, 2020, 235:4746–55., DOI 10.1002/jcp.29352 Tang J, Ren J, Cui Q, Zhang D, Kong D, Liao X, et al. A Prognostic 10-lncRNA Expression Signature for Predicting the Risk of Tumour Recurrence in Breast Cancer Patient. J Cel Mol Med, 2019, 23:6775–84., DOI 10.1111/jcmm.14556 Wang S, Chai K, Chen J. A Novel Prognostic Nomogram Based on 5 Long Non-coding RNAs in clear Cell Renal Cell Carcinoma. Oncol Lett, 2019, 18: 6605–13., DOI 10.3892/ol.2019.11009 Wu M, Yuan H, Li X, Liao Q, Liu Z. Identification of a Five-Gene Signature and Establishment of a Prognostic Nomogram to Predict Progression-free Interval of Papillary Thyroid Carcinoma. Front Endocrinol, 2019, 10:790., DOI 10.3389/ fendo.2019.00790 Yang G, Zhang Y, Yang J. A Five-microRNA Signature as Prognostic Biomarker in Colorectal Cancer by Bioinformatics Analysis. Front Oncol, 2019, 9:1207., DOI 10.3389/fonc.2019.01207 Niemira M, Collin F, Szalkowska A, Bielska A, Chwialkowska K, Reszec J, et al. Molecular Signature of Subtypes of Non-small-cell Lung Cancer by Large- Scale Transcriptional Profiling: Identification of Key Modules and Genes by Weighted Gene Co-expression Network Analysis, WGCNA). Cancers, Basel,, 2019, 12:12., DOI 10.3390/cancers12010037 Wu S, Zheng J, Li Y, Wu Z, Shi S, Huang M, et al. Development and Validation of an MRI-Based Radiomics Signature for the Preoperative Prediction of Lymph Node Metastasis in Bladder Cancer. EBioMedicine, 2018, 34:76–84., DOI 10.1016/j.ebiom.2018.07.029 Weinstein JN, Collisson EA, Collisson EA, Mills GB, Shaw KRM, Ozenberger BA, et al. The Cancer Genome Atlas Pan-Cancer Analysis Project. Nat Genet, 2013, 45:1113–20., DOI 10.1038/ng.2764 Li Z, LinW, Zheng J, Hong W, Zou J, Zhang T, et al. Identification of Immune- Related lncRNAs to Improve the Prognosis Prediction for Patients with Papillary Thyroid Cancer. Biosci Rep, 2021, 41., DOI 10.1042/bsr20204086 Huang Z, Xiao C, Zhang F, Zhou Z, Yu L, Ye C, et al. A Novel Framework to Predict Breast Cancer Prognosis Using Immune-Associated LncRNAs. Front Genet, 2020, 11:634195., DOI 10.3389/fgene.2020.634195 Shan Y, He R, Yang X, Zang S, Yao S, Gao M, et al. An Autophagy-Related lncRNA Prognostic Risk Model for Thyroid Cancer. Eur Arch Otorhinolaryngol, 2021):1–14., DOI 10.1007/s00405-021-07134-4 Schlumberger M, Leboulleux S. Current Practice in Patients with Differentiated Thyroid Cancer. Nat Rev Endocrinol, 2021, 17:176–88., DOI 10.1038/s41574-020-00448-z Luo Y, Shen D, Chen L, Wang G, Liu X, Qian K, et al. Identification of 9 Key Genes and Small Molecule Drugs in clear Cell Renal Cell Carcinoma. Aging, 2019, 11:6029–52., DOI 10.18632/aging.102161 Sun J, Zhang Z, Bao S, Yan C, Hou P, Wu N, et al. Identification of Tumor Immune Infiltration-Associated lncRNAs for Improving Prognosis and Immunotherapy Response of Patients with Non-small Cell Lung Cancer. J Immunother Cancer, 2020, 8:e000110., DOI 10.1136/jitc-2019-000110 Zhai X, Xue Q, Liu Q, Guo Y, Chen Z. Colon Cancer Recurrence-Associated Genes Revealed by WGCNA Co-expression Network Analysis. Mol Med Rep, 2017, 16:6499–505., DOI 10.3892/mmr.2017.7412 Wu Y, Liang S, Zhu H, Zhu Y. Analysis of Immune-Related Key Genes in Alzheimer’s Disease. Bioengineered, 2021)., DOI 10.1080/21655979.2021. 1999553 Lin R, Fogarty CE, Ma B, Li H, Ni G, Liu X, et al. Identification of Ferroptosis Genes in Immune Infiltration and Prognosis in Thyroid Papillary Carcinoma Using Network Analysis. BMC genomics, 2021, 22:576., DOI 10.1186/s12864- 021-07895-6 Zhai T, Muhanhali D, Jia X, Wu Z, Cai Z, Ling Y. Identification of Gene Coexpression Modules and Hub Genes Associated with Lymph Node Metastasis of Papillary Thyroid Cancer. Endocrine, 2019, 66:573–84., DOI 10.1007/s12020- 019-02021-9 Shuwen H, Xi Y, Miao D, Jiamin X, Jing Z, Weili G. Nine Genes Mediate the Therapeutic Effects of Iodine-131 Radiotherapy in Thyroid Carcinoma Patients. Dis Markers, 2020, 2020:9369341., DOI 10.1155/2020/9369341 Zhang T, Yan C, Ye Z, Yin X, Jiang TA. The Identification of Three Key Genes Related to Stemness in Thyroid Carcinoma through Comprehensive Analysis. Comb Chem High Throughput Screen, 2021, 24:423–32., DOI 10.2174/ 1386207323666200806164003 Li J, Han L, Roebuck P, Diao L, Liu L, Yuan Y, et al. TANRIC: An Interactive Open Platform to Explore the Function of lncRNAs in Cancer. Cancer Res, 2015, 75:3728–37., DOI 10.1158/0008-5472.can-15-0273 Song P, Jiang B, Liu Z, Ding J, Liu S, Guan W. A Three-lncRNA Expression Signature Associated with the Prognosis of Gastric Cancer Patients. Cancer Med, 2017, 6:1154–64., DOI 10.1002/cam4.1047 Bi JW, Zou YL, Qian JT, Chen WB. MiR-599 Serves a Suppressive Role in Anaplastic Thyroid Cancer by Activating the T-Cell Intracellular Antigen. Exp Ther Med, 2019, 18:2413–20., DOI 10.3892/etm.2019.7864 Song J,Wu S, Xia X,Wang Y, Fan Y, Yang Z. Cell Adhesion-Related Gene Somatic Mutations Are Enriched in Aggressive Papillary Thyroid Microcarcinomas. J Transl Med, 2018, 16:269., DOI 10.1186/s12967-018-1642-0 Pozdeyev N, Berlinberg A, Zhou Q, Wuensch K, Shibata H, Wood WM, et al. Targeting the NF-Κb Pathway as a Combination Therapy for Advanced Thyroid Cancer. PloS one, 2015, 10:e0134901., DOI 10.1371/journal.pone.0134901 Tzanavari T, Tasoulas J, Vakaki C, Mihailidou C, Tsourouflis G, Theocharis S. The Role of Adipokines in the Establishment and Progression of Head and Neck Neoplasms. Cmc, 2019, 26:4726–48., DOI 10.2174/ 0929867325666180713154505 Halestrap AP, Wilson MC. The Monocarboxylate Transporter Family-Role and Regulation. IUBMB life, 2012, 64:109–19., DOI 10.1002/iub.572 Seo YG, Choi HC. The Association between Type 2 Diabetes Mellitus and Thyroid Cancer, 2017, 2017:5850879., DOI 10.1155/2017/5850879 Park J-I, Strock CJ, Ball DW, Nelkin BD. Interleukin-1β Can Mediate Growth Arrest and Differentiation via the Leukemia Inhibitory factor/JAK/STAT Pathway in Medullary Thyroid Carcinoma Cells. Cytokine, 2005, 29: 125–34., DOI 10.1016/j.cyto.2004.10.005 Melillo RM, Guarino V, Avilla E, Galdiero MR, Liotti F, Prevete N, et al. Mast Cells Have a Protumorigenic Role in Human Thyroid Cancer. Oncogene, 2010, 29:6203–15., DOI 10.1038/onc.2010.348 Matsubara T, Kanto T, Kuroda S, Yoshio S, Higashitani K, Kakita N, et al. TIE2-expressing Monocytes as a Diagnostic Marker for Hepatocellular Carcinoma Correlates with Angiogenesis. Hepatology, 2013, 57:1416–25., DOI 10.1002/hep.25965 Brambilla E, Le Teuff G, Marguet S, Lantuejoul S, Dunant A, Graziano S, et al. Prognostic Effect of Tumor Lymphocytic Infiltration in Resectable Non-smallcell Lung Cancer. Jco, 2016, 34:1223–30., DOI 10.1200/jco.2015.63.0970 Sulaieva O, Chernenko O, Selesnov O, Nechay O, Maievskyi O, Falalyeyeva T, et al. Mechanisms of the Impact of Hashimoto Thyroiditis on Papillary Thyroid Carcinoma Progression: Relationship with the Tumor Immune Microenvironment. Endocrinol Metab, 2020, 35:443–55., DOI 10.3803/enm. 2020.35.2.443 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610012 EP 1610026 DI 10.3389/pore.2022.1610012 PG 15 ER PT J AU Zhang, X Ma, H Zhang, L Li, F AF Zhang, Xuefeng Ma, Hongfu Zhang, Liang Li, Fenghuan TI Predictive Role of Tumor-Stroma Ratio for Survival of Patients With Non-Small Cell Lung Cancer: A Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE recurrence; survival; meta-analysis; non-small cell lung cancer; tumor-stroma ratio ID recurrence; survival; meta-analysis; non-small cell lung cancer; tumor-stroma ratio AB Background: Role of tumor-stroma ratio (TSR) as a predictor of survival in patients with non-small cell lung cancer (NSCLC) remains not clear. A systematic review and metaanalysis was conducted to summarize current evidence for the role of TSR in NSCLC. Methods: Relevant cohort studies were retrieved via search of Medline, Embase, and Web of Science databases. The data was combined with a random-effect model by incorporating the between-study heterogeneity. Specifically, subgroup and metaregression analyses were performed to explore the association between TSR and survival in patients with squamous cell carcinoma (SCC) or adenocarcinoma (AC). Results: Nine cohort studies with 2031 patients with NSCLC were eligible for the metaanalysis. Pooled results showed that compared to those stroma-poor tumor, patients with stroma rich NSCLC were associated with worse recurrence-free survival (RFS, hazard ratio [HR] = 1.52, 95% confidence interval [CI]: 1.07 to 2.16, p = 0.02) and overall survival (OS, HR = 1.48, 95% CI: 1.20 to 1.82, p < 0.001). Subgroup analyses showed that stroma-rich tumor may be associated with a worse survival of SCC (HR = 1.89 and 1.47 for PFS and OS), but a possibly favorable survival of AC (HR = 0.28 and 0.69 for PFS and OS). Results of meta-regression analysis also showed that higher proportion of patients with SCC was correlated with higher HRs for RFS (Coefficient = 0.012, p = 0.03) and OS (Coefficient = 0.014, p = 0.02) in the included patients, while higher proportion of patients with AC was correlated with lower HRs for RFS (Coefficient = −0.012, p = 0.03) and OS (Coefficient = −0.013, p = 0.04), respectively. Conclusion: Tumor TSR could be used as a predictor of survival in patients with NSCLC. The relative proportion of patients with SCC/AC in the included NSCLC patients may be an important determinant for the association between TSR and survival in NSCLC. Stroma richness may be a predictor of poor survival in patients with lung SCC, but a predictor of better survival in patients with lung AC. C1 [Zhang, Xuefeng] Yantai Mountain Hospital, Department of Respiratory and Critical Care MedicineYantai, China. [Ma, Hongfu] Yantai Mountain Hospital, Department of Respiratory and Critical Care MedicineYantai, China. [Zhang, Liang] Yantai Mountain Hospital, Department of Respiratory and Critical Care MedicineYantai, China. [Li, Fenghuan] Yantai Mountain Hospital, Department of Respiratory and Critical Care MedicineYantai, China. RP Li, F (reprint author), Yantai Mountain Hospital, Department of Respiratory and Critical Care Medicine, Yantai, China. EM lifenghuan_255@21cn.com CR Bade BC, Dela Cruz CS. Lung Cancer 2020. Clin Chest Med, 2020, 41, 1):1–24., DOI 10.1016/j.ccm.2019.10.001 Schabath MB, Cote ML. Cancer Progress and Priorities: Lung Cancer. Cancer Epidemiol Biomarkers Prev, 2019, 28, 10):1563–79., DOI 10.1158/1055- 9965.epi-19-0221 Duma N, Santana-Davila R, Molina JR. Non-Small Cell Lung Cancer: Epidemiology, Screening, Diagnosis, and Treatment. Mayo Clinic Proc, 2019, 94, 8):1623–40., DOI 10.1016/j.mayocp.2019.01.013 Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ, Jr, Wu Y-L, et al. Lung Cancer: Current Therapies and New Targeted Treatments. The Lancet, 2017, 389, 10066):299–311., DOI 10.1016/s0140-6736(16)30958-8 Herbst RS,Morgensztern D, Boshoff C. The Biology andManagement of Nonsmall Cell Lung Cancer. Nature, 2018, 553, 7689):446–54., DOI 10.1038/ nature25183 Augustus E, Zwaenepoel K, Siozopoulou V, Raskin J, Jordaens S, Baggerman G, et al. Prognostic and Predictive Biomarkers in Non-small Cell Lung Cancer Patients on Immunotherapy-The Role of Liquid Biopsy in Unraveling the Puzzle. Cancers, Basel,, 2021, 13, 7)., DOI 10.3390/cancers13071675 Thakur MK, Gadgeel SM. Predictive and Prognostic Biomarkers in Non-small Cell Lung Cancer. Semin Respir Crit Care Med, 2016, 37, 5):760–70., DOI 10.1055/s-0036-1592337 Lee SS, Cheah YK. The Interplay between MicroRNAs and Cellular Components of Tumour Microenvironment, TME, on Non-small-cell Lung Cancer, NSCLC, Progression. J Immunol Res, 2019, 2019:3046379., DOI 10.1155/2019/3046379 Lin A, Wei T, Meng H, Luo P, Zhang J. Role of the Dynamic Tumor Microenvironment in Controversies Regarding Immune Checkpoint Inhibitors for the Treatment of Non-small Cell Lung Cancer, NSCLC, with EGFR Mutations. Mol Cancer, 2019, 18, 1):139., DOI 10.1186/s12943- 019-1062-7 Bremnes RM, Donnem T, Al-Saad S, Al-Shibli K, Andersen S, Sirera R, et al. The Role of Tumor Stroma in Cancer Progression and Prognosis: Emphasis on Carcinoma-Associated Fibroblasts and Non-small Cell Lung Cancer. J Thorac Oncol, 2011, 6, 1):209–17., DOI 10.1097/ jto.0b013e3181f8a1bd van Pelt GW, Sandberg TP, Morreau H, Gelderblom H, van Krieken JHJM, Tollenaar RAEM, et al. The Tumour-Stroma Ratio in colon Cancer: the Biological Role and its Prognostic Impact. Histopathology, 2018, 73, 2): 197–206., DOI 10.1111/his.13489 Kramer CJH, Vangangelt KMH, van Pelt GW, Dekker TJA, Tollenaar RAEM, Mesker WE. The Prognostic Value of Tumour-Stroma Ratio in Primary Breast Cancer with Special Attention to Triple-Negative Tumours: a Review. Breast Cancer Res Treat, 2019, 173, 1):55–64., DOI 10.1007/s10549-018-4987-4 Wu J, Liang C, Chen M, Su W. Association between Tumor-Stroma Ratio and Prognosis in Solid Tumor Patients: a Systematic Review and Meta-Analysis. Oncotarget, 2016, 7, 42):68954–65., DOI 10.18632/oncotarget.12135 Wang Z, Liu H, Zhao R, Zhang H, Liu C, Song Y. Tumor-stroma Ratio Is an Independent Prognostic Factor of Non-small Cell Lung Cancer. Zhongguo Fei Ai Za Zhi, 2013, 16, 4):191–6., DOI 10.3779/j.issn.1009-3419.2013.04.04 Zhang T, Xu J, Shen H, Dong W, Ni Y, Du J. Tumor-stroma Ratio Is an Independent Predictor for Survival in NSCLC. Int J Clin Exp Pathol, 2015, 8, 9):11348–55. Gurel D, Ulukus C, Karacam V, Ellidokuz H, Umay C, Oztop I, et al. The Prognostic Value of Morphologic Findings for Lung Squamous Cell Carcinoma Patients. Pathol - Res Pract, 2016, 212, 1):1–9., DOI 10.1016/ j.prp.2015.10.006 Xi K-X, Wen Y-S, Zhu C-M, Yu X-Y, Qin R-Q, Zhang X-W, et al. Tumorstroma Ratio, TSR, in Non-small Cell Lung Cancer, NSCLC, Patients after Lung Resection Is a Prognostic Factor for Survival. J Thorac Dis, 2017, 9, 10): 4017–26., DOI 10.21037/jtd.2017.09.29 Ichikawa T, Aokage K, Sugano M, Miyoshi T, Kojima M, Fujii S, et al. The Ratio of Cancer Cells to Stroma within the Invasive Area Is a Histologic Prognostic Parameter of Lung Adenocarcinoma. Lung Cancer, 2018, 118: 30–5., DOI 10.1016/j.lungcan.2018.01.023 Zhang X, Zhang Y, Sun Z, Ma H. Research on Correlation between Tumor- Stroma Ratio and Prognosis in Non-small Cell Lung Cancer. Minerva Med, 2019, 110, 6):590–2., DOI 10.23736/S0026-4806.19.06001-4 Koike Y, Aokage K, Ikeda K, Nakai T, Tane K, Miyoshi T, et al. Machine Learning-Based Histological Classification that Predicts Recurrence of Peripheral Lung Squamous Cell Carcinoma. Lung Cancer, 2020, 147:252–8., DOI 10.1016/j.lungcan.2020.07.011 Smit MA, Philipsen MW, Postmus PE, Putter H, Tollenaar RA, Cohen D, et al. The Prognostic Value of the Tumor-Stroma Ratio in Squamous Cell Lung Cancer, a Cohort Study. Cancer Treat Res Commun, 2020, 25:100247., DOI 10.1016/j.ctarc.2020.100247 Micke P, Strell C, Mattsson J, Martin-Bernabe A, Brunnstrom H, Huvila J, et al. The Prognostic Impact of the Tumour Stroma Fraction: A Machine Learning- Based Analysis in 16 Human Solid Tumour Types. EBioMedicine, 2021, 65: 103269., DOI 10.1016/j.ebiom.2021.103269 Faruki H, MayhewGM, Serody JS, Hayes DN, Perou CM, Lai-GoldmanM. Lung Adenocarcinoma and Squamous Cell Carcinoma Gene Expression Subtypes Demonstrate Significant Differences in Tumor Immune Landscape. J Thorac Oncol, 2017, 12, 6):943–53., DOI 10.1016/j.jtho.2017.03.010 Ojlert AK, Halvorsen AR, Nebdal D, Lund-Iversen M, Solberg S, Brustugun OT, et al. The Immune Microenvironment in Non-small Cell Lung Cancer Is Predictive of Prognosis after Surgery. Mol Oncol, 2019, 13, 5):1166–79., DOI 10.1002/1878-0261.12475 Chen L, Cao MF, Zhang X, Dang WQ, Xiao JF, Liu Q, et al. The Landscape of Immune Microenvironment in Lung Adenocarcinoma and Squamous Cell Carcinoma Based on PD-L1 Expression and Tumor-infiltrating Lymphocytes. Cancer Med, 2019, 8, 17):7207–18., DOI 10.1002/cam4.2580 Stroup DF, Berlin JA, Morton SC, Olkin I, Williamson GD, Rennie D, et al. Meta-analysis of Observational Studies in EpidemiologyA Proposal for Reporting. JAMA, 2000, 283, 15):2008–12., DOI 10.1001/ jama.283.15.2008 Higgins J, Green S. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0. China. The Cochrane Collaboration, 2011). AvaliableAt: www. cochranehandbook.org. Wells GA, Shea B, O’Connell D, Peterson J, Welch V, Losos M, et al. The Newcastle-Ottawa Scale, NOS, for Assessing the Quality of Nonrandomised Studies in Meta-Analyses. China., 2010). AvaliableAt: http://www.ohri.ca/ programs/clinical_epidemiology/oxford.asp. Higgins JPT, Thompson SG. Quantifying Heterogeneity in a Meta-Analysis. Statist Med, 2002, 21, 11):1539–58., DOI 10.1002/sim.1186 Patsopoulos NA, Evangelou E, Ioannidis JP. Sensitivity of Between-Study Heterogeneity in Meta-Analysis: Proposed Metrics and Empirical Evaluation. Int J Epidemiol, 2008, 37, 5):1148–57., DOI 10.1093/ije/dyn065 Egger M, Smith GD, Schneider M, Minder C. Bias in Meta-Analysis Detected by a Simple, Graphical Test. BMJ, 1997, 315, 7109):629–34., DOI 10.1136/ bmj.315.7109.629 van Pelt GW, Kjaer-Frifeldt S, van Krieken JHJM, Al Dieri R, Morreau H, Tollenaar RAEM, et al. Scoring the Tumor-Stroma Ratio in colon Cancer: Procedure and Recommendations. Virchows Arch, 2018, 473, 4):405–12., DOI 10.1007/s00428-018-2408-z Sung P-J, Rama N, Imbach J, Fiore S, Ducarouge B, Neves D, et al. Cancer- Associated Fibroblasts Produce Netrin-1 to Control Cancer Cell Plasticity. Cancer Res, 2019, 79, 14):3651–61., DOI 10.1158/0008-5472.can-18-2952 Wood SL, Pernemalm M, Crosbie PA, Whetton AD. The Role of the Tumor- Microenvironment in Lung Cancer-Metastasis and its Relationship to Potential Therapeutic Targets. Cancer Treat Rev, 2014, 40, 4):558–66., DOI 10.1016/ j.ctrv.2013.10.001 Souza da Silva RM, Queiroga EM, Paz AR, Neves FFP, Cunha KS, Dias EP. Standardized Assessment of the Tumor-Stroma Ratio in Colorectal Cancer: Interobserver Validation and Reproducibility of a Potential Prognostic Factor. Clin Pathol, 2021, 14:2632010X21989686., DOI 10.1177/2632010X21989686 Ludwig N, Rubenich DS, Zareba L, Siewiera J, Pieper J, Braganhol E, et al. Potential Roles of Tumor Cell- and Stroma Cell-Derived Small Extracellular Vesicles in Promoting a Pro-angiogenic Tumor Microenvironment. Cancers, Basel,, 2020, 12, 12). 3599., DOI 10.3390/cancers12123599 Fujiwara A, Funaki S, Fukui E, Kimura K, Kanou T, Ose N, et al. Effects of Pirfenidone Targeting the Tumor Microenvironment and Tumor-Stroma Interaction as a Novel Treatment for Non-small Cell Lung Cancer. Sci Rep, 2020, 10, 1):10900., DOI 10.1038/s41598-020-67904-8 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610021 EP 1610032 DI 10.3389/pore.2021.1610021 PG 12 ER PT J AU Xie, Y Fu, R Xiao, Z Li, G AF Xie, Yan Fu, Ruimin Xiao, Zheng Li, Gang TI A Risk Model Based on Immune-Related Genes Predicts Prognosis and Characterizes the Immune Landscape in Esophageal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunotherapy; chemotherapy; prognosis; esophageal cancer; immune-related genes; risk model ID immunotherapy; chemotherapy; prognosis; esophageal cancer; immune-related genes; risk model AB Aberrant immune gene expression has been shown to have close correlations with the occurrence and progression of esophageal cancer (EC). We aimed to generate a prognostic signature based on immune-related genes (IRGs) capable of predicting prognosis, immune checkpoint gene (ICG) expressions, and half-inhibitory concentration (IC50) for chemotherapy agents for EC patients. Transcriptome, clinical, and mutation data on tumorous and paratumorous tissues from EC patients were collected from The Cancer Genome Atlas (TCGA) database. Then, we performed differential analysis to identify IRGs differentially expressed in EC. Their biofunctions and related pathways were explored using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. These gene expression profiling data were merged with survival information and subjected to univariate Cox regression to select prognostic genes, which were then included in a Lasso-Cox model for signature generation (risk score calculation). Patients were divided into the high- and low-risk groups using the median risk score as a cutoff. The accuracy of the signature in overall survival prediction was assessed, so were its performances in predicting ICG expressions and IC50 for chemotherapy and targeted therapy agents and immune cell landscape characterization. Fifteen prognostic IRGs were identified, seven of which were optimal for risk score calculation. As expected, high-risk patients had worse overall survival than low-risk individuals. Significant differences were found in tumor staging, immune cell infiltration degree, frequency of tumor mutations, tumor mutation burden (TMB), and immune checkpoint gene expressions between high- vs. low-risk patients. Further, high-risk patients exhibited high predicted IC50 for paclitaxel, cisplatin, doxorubicin, and erlotinib compared to low-risk patients. The seven-IRG-based signature can independently and accurately predict overall survival and tumor progression, characterize the tumor immune microenvironment (TIME) and estimate ICG expressions and IC50 for antitumor therapies. It shows the potential of guiding personalized treatment for EC patients. C1 [Xie, Yan] Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Department of Clinical LaboratoryZhengzhou, China. [Fu, Ruimin] Henan Finance University, College of Health ManagementZhengzhou, China. [Xiao, Zheng] Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Department of Clinical LaboratoryZhengzhou, China. [Li, Gang] Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Department of Clinical LaboratoryZhengzhou, China. RP Li, G (reprint author), Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Department of Clinical Laboratory, Zhengzhou, China. EM gangli086@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Smyth EC, Lagergren J, Fitzgerald RC, Lordick F, Shah MA, Lagergren P, et al. Oesophageal Cancer. Nat Rev Dis Primers, 2017, 3:17048., DOI 10.1038/nrdp. 2017.48 Siegel R, Ma J, Zou Z, Jemal A. Cancer Statistics, 2014. CA A Cancer J Clin, 2014, 64(1):9–29., DOI 10.3322/caac.21208 Ammannagari N, Atasoy A. Current Status of Immunotherapy and Immune Biomarkers in Gastro-Esophageal Cancers. J Gastrointest Oncol, 2018, 9(1): 196–207., DOI 10.21037/jgo.2017.06.12 Doi T, Piha-Paul SA, Jalal SI, Saraf S, Lunceford J, Koshiji M, et al. Safety and Antitumor Activity of the Anti-programmed Death-1 Antibody Pembrolizumab in Patients with Advanced Esophageal Carcinoma. Jco, 2018, 36(1):61–7., DOI 10.1200/jco.2017.74.9846 Kato K, Shah MA, Enzinger P, Bennouna J, Shen L, Adenis A, et al. KEYNOTE-590: Phase III Study of First-Line Chemotherapy with or without Pembrolizumab for Advanced Esophageal Cancer. Future Oncol, 2019, 15(10):1057–66., DOI 10.2217/fon-2018-0609 Zhao Q, Yu J,Meng X. A Good Start of Immunotherapy in Esophageal Cancer. Cancer Med, 2019, 8(10):4519–26., DOI 10.1002/cam4.2336 Michaud D, Steward CR, Mirlekar B, Pylayeva-Gupta Y. Regulatory B Cells in Cancer. Immunol Rev, 2021, 299(1):74–92., DOI 10.1111/imr.12939 Sun C, Mezzadra R, Schumacher TN. Regulation and Function of the PD-L1 Checkpoint. Immunity, 2018, 48(3):434–52., DOI 10.1016/j.immuni.2018. 03.014 Dhupar R, Van Der Kraak L, Pennathur A, Schuchert MJ, Nason KS, Luketich JD, et al. Targeting Immune Checkpoints in Esophageal Cancer: A High Mutational Load Tumor. Ann Thorac Surg, 2017, 103(4):1340–9., DOI 10.1016/ j.athoracsur.2016.12.011 Kollmann D, Ignatova D, Jedamzik J, Chang Y-T, Jomrich G, Paireder M, et al. Expression of Programmed Cell Death Protein 1 by Tumor-Infiltrating Lymphocytes and Tumor Cells Is Associated with Advanced Tumor Stage in Patients with Esophageal Adenocarcinoma. Ann Surg Oncol, 2017, 24(9): 2698–706., DOI 10.1245/s10434-017-5858-7 Huang J, Xu B, Mo H, Zhang W, Chen X, Wu D, et al. Safety, Activity, and Biomarkers of SHR-1210, an Anti-PD-1 Antibody, for Patients with Advanced Esophageal Carcinoma. Clin Cancer Res, 2018, 24(6):1296–304., DOI 10.1158/ 1078-0432.ccr-17-2439 Zhou S, Zhao L, Liang Z, Liu S, Li Y, Liu S, et al. Indoleamine 2,3-dioxygenase 1 and Programmed Cell Death-Ligand 1 Co-expression Predicts Poor Pathologic Response and Recurrence in Esophageal Squamous Cell Carcinoma after Neoadjuvant Chemoradiotherapy. Cancers, 2019, 11(2):169., DOI 10.3390/ cancers11020169 Yang WF, Wang SZ. Expression of CD1a, CD80, CD86 in Dendritic Cell of Tumor Tissue and Regional Lymph Node in Esophageal Carcinoma. Ai Zheng, 2004, 23(2):189–92. Kelly RJ. The Emerging Role of Immunotherapy for Esophageal Cancer. Curr Opin Gastroenterol, 2019, 35(4):337–43., DOI 10.1097/mog. 0000000000000542 Yagi T, Baba Y, Okadome K, Kiyozumi Y, Hiyoshi Y, Ishimoto T, et al. Tumour-associated Macrophages Are Associated with Poor Prognosis and Programmed Death Ligand 1 Expression in Oesophageal Cancer. Eur J Cancer, 2019, 111:38–49., DOI 10.1016/j.ejca.2019.01.018 Valero C, Lee M, Hoen D, Wang J, Nadeem Z, Patel N, et al. The Association between TumorMutational Burden and Prognosis Is Dependent on Treatment Context. Nat Genet, 2021, 53(1):11–5., DOI 10.1038/s41588-020-00752-4 Ritterhouse LL. Tumor Mutational burden. Cancer Cytopathology, 2019, 127(12):735–6., DOI 10.1002/cncy.22174 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res, 2015, 43(7):e47., DOI 10.1093/nar/gkv007 Ito K, Murphy D. Application of Ggplot2 to Pharmacometric Graphics. CPT Pharmacometrics Syst Pharmacol, 2013, 2(10):e79., DOI 10.1038/psp.2013.56 Jia A, Xu L, Wang Y. Venn Diagrams in Bioinformatics. Brief Bioinform, 2021, 22(5):22., DOI 10.1093/bib/bbab108 Yu G, Wang L-G, Han Y, He Q-Y. clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters. OMICS: A J Integr Biol, 2012, 16(5): 284–7., DOI 10.1089/omi.2011.0118 Friedman J, Hastie T, Tibshirani R. Regularization Paths for Generalized Linear Models via Coordinate Descent. J Stat Softw, 2010, 33(1):1–22., DOI 10. 18637/jss.v033.i01 Blanche P, Dartigues J-F, Jacqmin-Gadda H. Estimating and Comparing Time-dependent Areas under Receiver Operating Characteristic Curves for Censored Event Times with Competing Risks. Statist Med, 2013, 32(30): 5381–97., DOI 10.1002/sim.5958 Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust Enumeration of Cell Subsets from Tissue Expression Profiles. Nat Methods, 2015, 12(5):453–7., DOI 10.1038/nmeth.3337 Mayakonda A, Lin D-C, Assenov Y, Plass C, Koeffler HP. Maftools: Efficient and Comprehensive Analysis of Somatic Variants in Cancer. Genome Res, 2018, 28(11):1747–56., DOI 10.1101/gr.239244.118 Pennathur A, Gibson MK, Jobe BA, Luketich JD. Oesophageal Carcinoma. The Lancet, 2013, 381(9864):400–12., DOI 10.1016/s0140-6736(12)60643-6 Watanabe M, Otake R, Kozuki R, Toihata T, Takahashi K, Okamura A, et al. Recent Progress in Multidisciplinary Treatment for Patients with Esophageal Cancer. Surg Today, 2020, 50(1):12–20., DOI 10.1007/s00595- 019-01878-7 Abdo J, Agrawal DK, Mittal SK. Basis for Molecular Diagnostics and Immunotherapy for Esophageal Cancer. Expert Rev Anticancer Ther, 2017, 17(1):33–45., DOI 10.1080/14737140.2017.1260449 Cowie A, Noble F, Underwood T. Strategies to Improve Outcomes in Esophageal Adenocarcinoma. Expert Rev Anticancer Ther, 2014, 14(6): 677–87., DOI 10.1586/14737140.2014.895668 Vrana D, Matzenauer M, Neoral C, Aujesky R, Vrba R, Melichar B, et al. From Tumor Immunology to Immunotherapy in Gastric and Esophageal Cancer. Ijms, 2018, 20(1):13., DOI 10.3390/ijms20010013 Wang L, Wei Q, Zhang M, Chen L, Li Z, Zhou C, et al. Identification of the Prognostic Value of Immune Gene Signature and Infiltrating Immune Cells for Esophageal Cancer Patients. Int Immunopharmacol, 2020, 87:106795., DOI 10. 1016/j.intimp.2020.106795 Zhang Z, Chen C, Fang Y, Li S, Wang X, Sun L, et al. Development of a Prognostic Signature for Esophageal Cancer Based on Nine Immune Related Genes. BMC Cancer, 2021, 21(1):113., DOI 10.1186/s12885-021-07813-9 Dorner BG, Dorner MB, Zhou X, Opitz C, Mora A, Guttler S, et al. Selective Expression of the Chemokine Receptor XCR1 on Cross-Presenting Dendritic Cells Determines Cooperation with CD8+ T Cells. Immunity, 2009, 31(5): 823–33., DOI 10.1016/j.immuni.2009.08.027 Abdullah Zubir AZ, Whawell SA, Wong TS, Khurram SA. The Chemokine Lymphotactin and its Recombinant Variants in Oral Cancer Cell Regulation. Oral Dis, 2020, 26(8):1668–76., DOI 10.1111/odi.13500 Audsley KM, McDonnell AM, Waithman J. Cross-Presenting XCR1+ Dendritic Cells as Targets for Cancer Immunotherapy. Cells, 2020, 9(3): 565., DOI 10.3390/cells9030565 Schneider G, Nieznanski K, Jozwiak J, Slomnicki LP, Redowicz MJ, Filipek A. Tubulin Binding Protein, CacyBP/SIP, Induces Actin Polymerization and May Link Actin and Tubulin Cytoskeletons. Biochim Biophys Acta, Bba, - Mol Cel Res, 2010, 1803(11):1308–17., DOI 10.1016/j.bbamcr.2010.07.003 Kilanczyk E, Filipek S, Filipek A. ERK1/2 Is Dephosphorylated by a Novel Phosphatase - CacyBP/SIP. Biochem Biophys Res Commun, 2011, 404(1): 179–83., DOI 10.1016/j.bbrc.2010.11.088 Zhao M, Zhang RZ, Qi DW, Chen HY, Zhang GC. CacyBP/SIP Promotes Tumor Progression by Regulating Apoptosis and Arresting the Cell Cycle in Osteosarcoma. Exp Ther Med, 2020, 20(2):1397–404., DOI 10.3892/etm.2020. 8843 Shi H, Gao Y, Tang Y, Wu Y, Gong H, Du J, et al. CacyBP/SIP Protein Is Important for the Proliferation of Human Glioma Cells. IUBMB Life, 2014, 66(4):286–91., DOI 10.1002/iub.1263 Zhai H, Shi Y, Chen X, Wang J, Lu Y, Zhang F, et al. CacyBP/SIP Promotes the Proliferation of colon Cancer Cells. PLoS One, 2017, 12(2):e0169959., DOI 10. 1371/journal.pone.0169959 Chen X, Hu S-L, Feng Y, Li P, Mao Q-S, Xue W-J. Expression of Fatty Acid- Binding Protein-3 in Gastrointestinal Stromal Tumors and its Significance for Prognosis. J Surg Res, 2021, 260:462–6., DOI 10.1016/j.jss.2020.11.003 Tang Z, Shen Q, Xie H, Zhou X, Li J, Feng J, et al. Elevated Expression of FABP3 and FABP4 Cooperatively Correlates with Poor Prognosis in Nonsmall Cell Lung Cancer, NSCLC). Oncotarget, 2016, 7(29):46253–62., DOI 10. 18632/oncotarget.10086 Jacenik D, Beswick EJ, Krajewska WM, Prossnitz ER. G Protein-Coupled Estrogen Receptor in colon Function, Immune Regulation and Carcinogenesis. Wjg, 2019, 25(30):4092–104., DOI 10.3748/wjg.v25.i30.4092 Xu E, Xia X, Jiang C, Li Z, Yang Z, Zheng C, et al. GPER1 Silencing Suppresses the Proliferation,Migration, and Invasion of Gastric Cancer Cells by Inhibiting PI3K/AKT-Mediated EMT. Front Cel Dev Biol, 2020, 8:591239., DOI 10.3389/ fcell.2020.591239 Zhang C, Luo Y, Zhang Z, Zhang Z, Zhang G,Wang F, et al. Identification of a Prognostic Immune Signature for Esophageal Squamous Cell Carcinoma to Predict Survival and Inflammatory Landscapes. Front Cel Dev Biol, 2020, 8: 580005., DOI 10.3389/fcell.2020.580005 Olivier M, Langerod A, Carrieri P, Bergh J, Klaar S, Eyfjord J, et al. The Clinical Value of Somatic TP53 Gene Mutations in 1,794 Patients with Breast Cancer. Clin Cancer Res, 2006, 12(4):1157–67., DOI 10.1158/1078- 0432.CCR-05-1029 La Fleur L, Falk-Sorqvist E, Smeds P, Berglund A, Sundstrom M, Mattsson JS, et al. Mutation Patterns in a Population-Based Non-small Cell Lung Cancer Cohort and Prognostic Impact of Concomitant Mutations in KRAS and TP53 or STK11. Lung cancer, 2019, 130:50–8., DOI 10.1016/j.lungcan. 2019.01.003 Zhang X-F, Pan K,Weng D-S, Chen C-L, Wang Q-J, Zhao J-J, et al. Cytotoxic T Lymphocyte Antigen-4 Expression in Esophageal Carcinoma: Implications for Prognosis. Oncotarget, 2016, 7(18):26670–9., DOI 10.18632/oncotarget.8476 Kiyozumi Y, Baba Y, Okadome K, Yagi T, Ishimoto T, Iwatsuki M, et al. IDO1 Expression Is Associated with Immune Tolerance and Poor Prognosis in Patients with Surgically Resected Esophageal Cancer. Ann Surg, 2019, 269(6): 1101–8., DOI 10.1097/sla.0000000000002754 Loeser H, Kraemer M, Gebauer F, Bruns C, Schroder W, Zander T, et al. The Expression of the Immune Checkpoint Regulator VISTA Correlates with Improved Overall Survival in pT1/2 Tumor Stages in Esophageal Adenocarcinoma. Oncoimmunology, 2019, 8(5):e1581546., DOI 10.1080/ 2162402x.2019.1581546 Rosenberg AJ,Wainwright DA, Rademaker A, Galvez C, GenetM, Zhai L, et al. Indoleamine 2,3-dioxygenase 1 and Overall Survival of Patients Diagnosed with Esophageal Cancer. Oncotarget, 2018, 9(34):23482–93., DOI 10.18632/ oncotarget.25235 Gibney GT, Weiner LM, Atkins MB. Predictive Biomarkers for Checkpoint Inhibitor-Based Immunotherapy. Lancet Oncol, 2016, 17(12):e542–e551., DOI 10.1016/s1470-2045(16)30406-5 Wu L, Saxena S, Awaji M, Singh RK. Tumor-Associated Neutrophils in Cancer: Going Pro. Cancers, 2019, 11(4):564., DOI 10.3390/cancers11040564) Dyduch G, Kaczmarczyk K, Okon K. Mast Cells and Cancer: Enemies or Allies? Pol J Pathol, 2012, 63(1):1–7. Li Y, Liang L, DaiW, Cai G, Xu Y, Li X, et al. Prognostic Impact of Programed Cell Death-1, PD-1, and PD-Ligand 1, PD-L1, Expression in Cancer Cells and Tumor Infiltrating Lymphocytes in Colorectal Cancer. Mol Cancer, 2016, 15(1):55., DOI 10.1186/s12943-016-0539-x Webb JR, Milne K, Kroeger DR, Nelson BH. PD-L1 Expression Is Associated with Tumor-Infiltrating T Cells and Favorable Prognosis in High-Grade Serous Ovarian Cancer. Gynecol Oncol, 2016, 141(2):293–302., DOI 10.1016/ j.ygyno.2016.03.008 Lin A, Wei T, Meng H, Luo P, Zhang J. Role of the Dynamic Tumor Microenvironment in Controversies Regarding Immune Checkpoint Inhibitors for the Treatment of Non-small Cell Lung Cancer, NSCLC, with EGFR Mutations. Mol Cancer, 2019, 18(1):139., DOI 10.1186/s12943-019- 1062-7 Kandioler D, Schoppmann SF, Zwrtek R, Kappel S, Wolf B, Mittlbock M, et al. The Biomarker TP53 Divides Patients with Neoadjuvantly Treated Esophageal Cancer into 2 Subgroups with Markedly Different Outcomes. A P53 Research Group Study. J Thorac Cardiovasc Surg, 2014, 148(5):2280–6., DOI 10.1016/j. jtcvs.2014.06.079 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610030 EP 1610042 DI 10.3389/pore.2022.1610030 PG 13 ER PT J AU Long, C Xu, Qb Ding, L Huang, Lj Ji, Y AF Long, Cong Xu, Qiu-bo Ding, Li Huang, Li-juan Ji, Yong TI Circular RNAs as Diagnostic and Prognostic Indicators of Colorectal Cancer: A Pooled Analysis of Individual Studies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal cancer; circular RNA; diagnosis; prognosis; pooled analysis ID colorectal cancer; circular RNA; diagnosis; prognosis; pooled analysis AB Background: Circular RNAs (circRNAs) have proven as a special subset of endogenous RNAs that are implicated in the tumorigenesis of various cancers. This study sought to evaluate the role of circRNAs in the diagnosis and prognosis of colorectal cancer (CRC). Methods: The online databases were searched for collecting relevant studies on circRNAs as diagnostic and prognostic biomarkers of CRC. Two researchers independently screened literature, extracted data, and evaluated the bias and risks of included studies. The diagnostic and prognostic indicatorsweremerged and analyzed using STATA 12.0 software, and sources of heterogeneity were traced by the sensitivity analysis and the meta-regression test. Results: A total of 29 articles representing 2639 CRC patients were included. The pooled sensitivity, specificity, and area under the curve (AUC) of circRNAs in differentiating CRC from non-tumor control were 0.75 (95% CI: 0.69–0.80) and 0.74 (95% CI: 0.69–0.78) and 0.81, respectively. The survival analysis showed that up-regulations of up-regulated circRNAs were significantly related to dismal survival inCRCpatients (HR = 2.38, p < 0.001). A stratified analysis showed that the comprehensive diagnostic value of up-regualted circRNAs in CRC was higher than that of down-regualted circRNAs (AUC: 0.83 vs. 0.77; Z test, p < 0.05). The efficacy of tissue-derived circRNAs in the diagnosis of CRC was equal to that of plasma/serum-derived ones (AUC: 0.81 vs. 0.82; Z test, p > 0.05). Conclusion: Abnormally expressed circRNAs as auxiliary biomarkers present underlying value in the diagnosis and prognosis prediction of CRC. C1 [Long, Cong] Jingjiang People’s Hospital, Department of Clinical LaboratoryTaizhou, China. [Xu, Qiu-bo] Jingjiang People’s Hospital, Department of Clinical LaboratoryTaizhou, China. [Ding, Li] Jingjiang People’s Hospital, Department of Clinical LaboratoryTaizhou, China. [Huang, Li-juan] Jingjiang People’s Hospital, Department of Clinical LaboratoryTaizhou, China. [Ji, Yong] Jingjiang People’s Hospital, Department of General SurgeryTaizhou, China. RP Ji, Y (reprint author), Jingjiang People’s Hospital, Department of General Surgery, Taizhou, China. EM jjrmyongji@163.com CR Siegel RL,Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA A Cancer J Clin, 2021, 71(1):7–33. PMID: 33433946., DOI 10.3322/caac.21654 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer Statistics in China, 2015. CA: a Cancer J clinicians, 2016, 66(2):115–32. PMID: 26808342., DOI 10.3322/caac.21338 Simon K. Colorectal Cancer Development and Advances in Screening. Clin Interv Aging, 2016, 11:967–76. PMID: 27486317., DOI 10.2147/CIA.S109285 Diaz-Tasende J. Colorectal Cancer Screening and Survival. Rev Esp Enferm Dig, 2018, 110(11):681–3. PMID: 30284905., DOI 10.17235/reed.2018.5870/2018 Maida M, Macaluso FS, Ianiro G, Mangiola F, Sinagra E, Hold G, et al. Screening of Colorectal Cancer: Present and Future. Expert Rev Anticancer Ther, 2017, 17(12):1131–46. PMID: 29022408., DOI 10.1080/14737140.2017. 1392243 Dumoulin FL, Hildenbrand R. Endoscopic Resection Techniques for Colorectal Neoplasia: Current Developments. Wjg, 2019, 25(3):300–7. PMID: 30686899., DOI 10.3748/wjg.v25.i3.300 Nicholson BD, Shinkins B, Pathiraja I, Roberts NW, James TJ, Mallett S, et al. Blood CEA Levels for Detecting Recurrent Colorectal Cancer. Cochrane Database Syst Rev, 2015, 2015(12):CD011134. PMID: 26661580., DOI 10. 1002/14651858.CD011134.pub2 Fischer JW, Leung AKL. CircRNAs: a Regulator of Cellular Stress. Crit Rev Biochem Mol Biol, 2017, 52(2):220–33. PMID: 28095716., DOI 10.1080/ 10409238.2016.1276882 Patop IL, Wust S, Kadener S. Past, Present, and Future of Circ RNA S. EMBO J, 2019, 38(16):e100836. PMID: 31343080., DOI 10.15252/embj.2018100836 Wilusz JE. A 360° View of Circular RNAs: From Biogenesis to Functions. Wiley Interdiscip Rev RNA, 2018, 9(4):e1478. PMID: 29655315., DOI 10.1002/wrna. 1478 Salzman J. Circular RNA Expression: Its Potential Regulation and Function. Trends Genet, 2016, 32(5):309–16. [PMID: 27050930]., DOI 10.1016/j.tig.2016. 03.002 Kristensen LS, Hansen TB, Veno MT, Kjems J. Circular RNAs in Cancer: Opportunities and Challenges in the Field. Oncogene, 2018, 37(5):555–65. PMID: 28991235., DOI 10.1038/onc.2017.361 Meng S, Zhou H, Feng Z, Xu Z, Tang Y, Li P, et al. CircRNA: Functions and Properties of a Novel Potential Biomarker for Cancer. Mol Cancer, 2017, 16(1):94. PMID: 28535767., DOI 10.1186/s12943-017-0663-2 Fang G, Ye B-L, Hu B-R, Ruan X-J, Shi Y-X. CircRNA_100290 Promotes Colorectal Cancer Progression through miR-516b-Induced Downregulation of FZD4 Expression and Wnt/β-Catenin Signaling. Biochem Biophysical Res Commun, 2018, 504(1):184–9. PMID: 30173892., DOI 10.1016/j.bbrc.2018. 08.152 Ge Z, Li LF, Wang CY,Wang Y, Ma WL. CircMTO1 Inhibits Cell Proliferation and Invasion by Regulating Wnt/β-Catenin Signaling Pathway in Colorectal Cancer. Eur Rev Med Pharmacol Sci, 2018, 22(23):8203–9. PMID: 30556859., DOI 10.26355/eurrev_201812_16513 Hsiao K-Y, Lin Y-C, Gupta SK, Chang N, Yen L, Sun HS, et al. Noncoding Effects of Circular RNA CCDC66 Promote Colon Cancer Growth and Metastasis. Cancer Res, 2017, 77(9):2339–50. PMID: 28249903., DOI 10. 1158/0008-5472.CAN-16-1883 Ji W, Qiu C, Wang M, Mao N, Wu S, Dai Y. Hsa_circ_0001649: A Circular RNA and Potential Novel Biomarker for Colorectal Cancer. Biochem Biophysical Res Commun, 2018, 497(1):122–6. PMID: 29421663., DOI 10.1016/j.bbrc.2018.02.036 Li J, Ni S, Zhou C, Ye M. The Expression Profile and Clinical Application Potential of Hsa_circ_0000711 in Colorectal Cancer. Cmar, 2018, Vol. 10: 2777–84. PMID: 30147374., DOI 10.2147/cmar.s172388 Li R, Wu B, Xia J, Ye L, Yang X. Circular RNA hsa_circRNA_102958 Promotes Tumorigenesis of Colorectal Cancer via miR-585/CDC25B axis. Cmar, 2019, Vol. 11:6887–93. PMID: 31413634., DOI 10.2147/cmar.s212180 Li X-N, Wang Z-J, Ye C-X, Zhao B-C, Huang X-X, Yang L. Circular RNA circVAPA Is Up-Regulated and Exerts Oncogenic Properties by Sponging miR-101 in Colorectal Cancer. Biomed Pharmacother, 2019, 112:108611. PMID: 30797148., DOI 10.1016/j.biopha.2019.108611 Ruan H, Deng X, Dong L, Yang D, Xu Y, Peng H, et al. Circular RNA Circ_0002138 Is Down-Regulated and Suppresses Cell Proliferation in Colorectal Cancer. Biomed Pharmacother, 2019, 111:1022–8. PMID: 30841415., DOI 10.1016/j.biopha.2018.12.150 Wang F, Wang J, Cao X, Xu L, Chen L. Hsa_circ_0014717 Is Downregulated in Colorectal Cancer and Inhibits Tumor Growth by Promoting P16 Expression. Biomed Pharmacother, 2018, 98:775–82. PMID: 29571246., DOI 10.1016/j. biopha.2018.01.015 Wang J, Li X, Lu L, He L, Hu H, Xu Z. Circular RNA Hsa_circ_0000567 Can Be Used as a Promising Diagnostic Biomarker for Human Colorectal Cancer. J Clin Lab Anal, 2018, 32(5):e22379. PMID: 29333615., DOI 10.1002/jcla.22379 Wang X, Zhang Y, Huang L, Zhang J, Pan F, Li B, et al. Decreased Expression of Hsa_circ_001988 in Colorectal Cancer and its Clinical Significances. Int J Clin Exp Pathol, 2015, 8(12):16020–5. PMID: 26884878. Weng W, Wei Q, Toden S, Yoshida K, Nagasaka T, Fujiwara T, et al. Circular RNA ciRS-7-A Promising Prognostic Biomarker and a Potential Therapeutic Target in Colorectal Cancer. Clin Cancer Res, 2017, 23(14):3918–28. PMID: 28174233., DOI 10.1158/1078-0432.CCR-16-2541 Yuan Y, Liu W, Zhang Y, Zhang Y, Sun S. CircRNA Circ_0026344 as a Prognostic Biomarker Suppresses Colorectal Cancer Progression via microRNA-21 and microRNA-31. Biochem Biophysical Res Commun, 2018, 503(2):870–5. PMID: 29928882., DOI 10.1016/j.bbrc.2018.06.089 Zeng K, Chen X, Xu M, Liu X, Hu X, Xu T, et al. CircHIPK3 Promotes Colorectal Cancer Growth and Metastasis by Sponging miR-7. Cell Death Dis, 2018, 9(4):417. PMID: 29549306., DOI 10.1038/s41419-018-0454-8 Zhang P, Zuo Z, Shang W, Wu A, Bi R, Wu J, et al. Identification of Differentially Expressed Circular RNAs in Human Colorectal Cancer. Tumour Biol, 2017, 39(3):101042831769454–1010428317694546. PMID: 28349836., DOI 10.1177/1010428317694546 Zhang W, Yang S, Liu Y, Wang Y, Lin T, Li Y, et al. Hsa_circ_0007534 as a Blood-Based Marker for the Diagnosis of Colorectal Cancer and its Prognostic Value. Int J Clin Exp Pathol, 2018, 11(3):1399–406. PMID: 31938236. Zheng X, Chen L, Zhou Y, Wang Q, Zheng Z, Xu B, et al. A Novel Protein Encoded by a Circular RNA circPPP1R12A Promotes Tumor Pathogenesis and Metastasis of colon Cancer via Hippo-YAP Signaling. Mol Cancer, 2019, 18(1):47. PMID: 30925892., DOI 10.1186/s12943-019-1010-6 Zhuo F, Lin H, Chen Z, Huang Z, Hu J. The Expression Profile and Clinical Significance of circRNA0003906 in Colorectal Cancer. Ott, 2017, Vol. 10: 5187–93. PMID: 29123417., DOI 10.2147/ott.s147378 Liu F, Xiao X-L, Liu Y-J, Xu R-H, Zhou W-J, Xu H-C, et al. CircRNA_0084927 Promotes Colorectal Cancer Progression by Regulating miRNA-20b-3p/ glutathione S-Transferase Mu 5 axis. Wjg, 2021, 27(36):6064–78. PMID: 34629820 PMCID: PMC8476332., DOI 10.3748/wjg.v27.i36.6064 Wang J, Wang T, Hu S, Li J, Ni C, Ye M. Hsa_circ_0043278 Inhibits Tumorigenesis and Is Downregulated in Colorectal Cancer. Cmar, 2021, Vol. 13:965–75. PMID: 33568942 PMCID: PMC7868253., DOI 10.2147/cmar. s289775 Li J, Song Y, Wang J, Huang J. Plasma Circular RNA Panel Acts as a Novel Diagnostic Biomarker for Colorectal Cancer Detection. Am J Transl Res, 2020, 12(11):7395–403. PMID: 33312376 PMCID: PMC7724351. Alkhizzi B, KhanM, Al-Ghafari A, Choudhry H. Upregulation of Circular and Linear METTL3 and USP3 in Colorectal Cancer. Oncol Lett, 2021, 22(3):675. PMID: 34345300 PMCID: PMC8323003., DOI 10.3892/ol.2021.12936 Mai S, Zhang Z, Mi W. Upregulation of circ_PVT1 and Circ_001569 Indicate Unfavorable Prognosis in Colorectal Cancer. Ann Clin Lab Sci, 2021, 51(1): 55–60. PMID: 33653781. Xu Y, Wang C, Lu X, Qi R, Wang X, Zhao J. CircRNA_002178 as a ceRNA Promotes the Development of Colorectal Cancer by Regulating miR-542-3p/ CREB1. Am J Transl Res, 2021, 13(9):10038–55. PMID: 34650680 PMCID: PMC8507087. Wang J, Zhang Y, Song H, Yin H, Jiang T, Xu Y, et al. The Circular RNA circSPARC Enhances the Migration and Proliferation of Colorectal Cancer by Regulating the JAK/STAT Pathway. Mol Cancer, 2021, 20(1):81. PMID: 34074294 PMCID: PMC8167978., DOI 10.1186/s12943-021-01375-x Song Y, Cao P, Li J. Plasma Circular RNA Hsa_circ_0001821 Acts as a Novel Diagnostic Biomarker for Malignant Tumors. J Clin Lab Anal, 2021, 35(11): e24009. PMID: 34523755., DOI 10.1002/jcla.24009 Karousi P, Artemaki PI, Sotiropoulou CD, Christodoulou S, Scorilas A, Kontos CK. Identification of Two Novel Circular RNAs Deriving from BCL2L12 and Investigation of Their Potential Value as a Molecular Signature in Colorectal Cancer. Ijms, 2020, 21(22):8867. PMID: 33238574 PMCID: PMC7709015., DOI 10.3390/ijms21228867 Wang G, Yang H. CircRNA DUSP16 Knockdown Suppresses Colorectal Cancer Progression by Regulating the miR-432-5p/E2F6 Axis. Cmar, 2021, Vol. 13: 6599–609. PMID: 34456589 PMCID: PMC8387644., DOI 10.2147/cmar.s323437 Gao L, Tang X, He Q, Sun G, Wang C, Qu H. Exosome-transmitted circCOG2 Promotes Colorectal Cancer Progression via miR-1305/tgf-B2/smad3 Pathway. Cell Death Discov., 2021, 7(1):281. PMID: 34635639 PMCID: PMC8505430., DOI 10.1038/s41420-021-00680-0 Whiting PF, Rutjes AWS, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, et al.QUADAS-2 Group. QUADAS-2: a Revised Tool for the Quality Assessment of Diagnostic Accuracy Studies. Ann Intern Med, 2011, 155(8): 529–36. PMID: 22007046., DOI 10.7326/0003-4819-155-8-201110180-00009 Stang A. Critical Evaluation of the Newcastle-Ottawa Scale for the Assessment of the Quality of Nonrandomized Studies in Meta-Analyses. Eur J Epidemiol, 2010, 25(9):603–5. PMID: 20652370., DOI 10.1007/s10654-010-9491-z Lin N, Lin Y, Fu X, Wu C, Xu J, Cui Z, et al. MicroRNAs as a Novel Class of Diagnostic Biomarkers in Detection of Oral Carcinoma: a Meta-Analysis Study. Clin Lab, 2016, 62(3):451–61. PMID: 27156336., DOI 10.7754/clin.lab. 2015.150802 Glas AS, Lijmer JG, Prins MH, Bonsel GJ, Bossuyt PMM. The Diagnostic Odds Ratio: a Single Indicator of Test Performance. J Clin Epidemiol, 2003, 56(11): 1129–35. PMID: 14615004., DOI 10.1016/s0895-4356(03)00177-x Wang M, Yang Y, Xu J, Bai W, Ren X, Wu H. CircRNAs as Biomarkers of Cancer: a Meta-Analysis. BMC Cancer, 2018, 18(1):303. PMID: 29554887., DOI 10.1186/s12885-018-4213-0 Lin Y, Luo X, Cui Z, Chen Y. Diagnostic Potential for Circular RNAs in Gastric Carcinoma: a Meta-Analysis. Clin Lab, 2019, 65(3). PMID: 30868864., DOI 10. 7754/Clin.Lab.2018.180810 Hao Q, Han Y, XiaW,Wang Q, Qian H. Systematic Review and Meta-Analysis of the Utility of Circular RNAs as Biomarkers of Hepatocellular Carcinoma. Can J Gastroenterol Hepatol, 2019, 2019:1–13. PMID: 31187026., DOI 10.1155/ 2019/1684039 Huang X, Zhang W, Shao Z. Prognostic and Diagnostic Significance of circRNAs Expression in Lung Cancer. J Cel Physiol, 2019, 234(10): 18459–65. PMID: 30895620., DOI 10.1002/jcp.28481 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610037 EP 1610048 DI 10.3389/pore.2022.1610037 PG 12 ER PT J AU Wang, B Sun, Y AF Wang, Bingqi Sun, Yufu TI SELPLG Expression Was Potentially Correlated With Metastasis and Prognosis of Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; prognosis; metastasis; osteosarcoma; SELPLG ID biomarker; prognosis; metastasis; osteosarcoma; SELPLG AB Background: Osteosarcoma (OS) is the most prevalent malignant primary bone tumor in children. Selectin P ligand gene (SELPLG) has been studied in several cancers. Our research aimed to explore the role of SELPLG in OS. Methods: All OS patient data was obtained from TARGET and GEO databases. Differential expression analyses were conducted in limma package of R. Functional analyses included GO and KEGG enrichment analyses. Immune cell infiltration analysis was done in CIBERSORT software. The overall survival was calculated using survival and survminer package of R. Results: Significantly lower SELPLG expression was observed in metastatic OS samples compared with non-metastatic OS samples, both in TARGET and in GSE21257. Low SELPLG expression was an independent undesirable prognostic factor for OS patients, in both TARGET and GEO datasets. Totally 62 differentially expressed gene (DEG) overlaps were found between high SELPLG vs. low SELPLG and non-metastatic vs. metastatic OS samples, affecting metastases and thereby influencing the prognosis, which were significantly enriched in 40 GO and six KEGG terms. Five types of immune cells were significantly differentially infiltrated between high and low SELPLG expression OS patients. Conclusion: SELPLG is closely correlated with metastases and prognosis of OS patients. The OS patients with low SELPLG expression have relatively poorer prognosis and SELPLG is a potential prognostic biomarker for OS. C1 [Wang, Bingqi] Tianjin First Central Hospital, Department of Orthopedic SurgeryTianjin, China. [Sun, Yufu] Tianjin First Central Hospital, Department of Orthopedic SurgeryTianjin, China. RP Sun, Y (reprint author), Tianjin First Central Hospital, Department of Orthopedic Surgery, Tianjin, China. EM sunyftj@outlook.com CR Luetke A, Meyers PA, Lewis I, Juergens H. Osteosarcoma Treatment - where Do We Stand? A State of the Art Review. Cancer Treat Rev, 2014, 40(4): 523–32., DOI 10.1016/j.ctrv.2013.11.006 Cortini M, Avnet S, Baldini N. Mesenchymal Stroma: Role in Osteosarcoma Progression. Cancer Lett, 2017, 405:90–9., DOI 10.1016/ j.canlet.2017.07.024 Allison DC, Carney SC, Ahlmann ER, Hendifar A, Chawla S, Fedenko A, et al. A Meta-Analysis of Osteosarcoma Outcomes in the Modern Medical Era. Sarcoma, 2012, 2012:1–10., DOI 10.1155/2012/704872 Anninga JK, Gelderblom H, Fiocco M, Kroep JR, Taminiau AHM, Hogendoorn PCW, et al. Chemotherapeutic Adjuvant Treatment for Osteosarcoma: where Do We Stand? Eur J Cancer, 2011, 47(16):2431–45., DOI 10.1016/j.ejca.2011.05.030 Wang X, Qin G, Liang X, Wang W, Wang Z, Liao D, et al. Targeting the CK1α/ CBX4 axis for Metastasis in Osteosarcoma. Nat Commun, 2020, 11(1):1141., DOI 10.1038/s41467-020-14870-4 Isakoff MS, Bielack SS, Meltzer P, Gorlick R. Osteosarcoma: Current Treatment and a Collaborative Pathway to Success. Jco, 2015, 33(27): 3029–35., DOI 10.1200/JCO.2014.59.4895 Yang C, Tian Y, Zhao F, Chen Z, Su P, Li Y, et al. Bone Microenvironment and Osteosarcoma Metastasis. Ijms, 2020, 21(19):6985., DOI 10.3390/ijms21196985 Botter SM, Neri D, Fuchs B. Recent Advances in Osteosarcoma. Curr Opin Pharmacol, 2014, 16:15–23., DOI 10.1016/j.coph.2014.02.002 Czarnecka AM, Synoradzki K, Firlej W, Bartnik E, Sobczuk P, Fiedorowicz M, et al. Molecular Biology of Osteosarcoma. Cancers, 2020, 12(8):2130., DOI 10.3390/cancers12082130 Saraf AJ, Fenger JM, Roberts RD. Osteosarcoma: Accelerating Progress Makes for a Hopeful Future. Front Oncol, 2018, 8:4., DOI 10.3389/fonc.2018.00004 Shen S, Yao T, Xu Y, Zhang D, Fan S, Ma J. CircECE1 Activates Energy Metabolism in Osteosarcoma by Stabilizing C-Myc. Mol Cancer, 2020, 19(1): 151., DOI 10.1186/s12943-020-01269-4 Bime C, Pouladi N, Sammani S, Batai K, Casanova N, Zhou T, et al. Genome-Wide Association Study in African Americans with Acute Respiratory Distress Syndrome Identifies the Selectin P Ligand Gene as a Risk Factor. Am J Respir Crit Care Med, 2018, 197(11):1421–32., DOI 10.1164/rccm.201705-0961OC Tinoco R, Otero DC, Takahashi AA, Bradley LM. PSGL-1: A New Player in the Immune Checkpoint Landscape. Trends Immunol, 2017, 38(5):323–35., DOI 10.1016/j.it.2017.02.002 Li J, Zhou Z, Zhang X, Zheng L, He D, Ye Y, et al. Inflammatory Molecule, PSGL-1, Deficiency Activates Macrophages to Promote Colorectal Cancer Growth through NFκB Signaling. Mol Cancer Res, 2017, 15(4):467–77., DOI 10.1158/1541-7786.MCR-16-0309 Sultana DA, Zhang SL, Todd SP, Bhandoola A. Expression of Functional P-Selectin Glycoprotein Ligand 1 on Hematopoietic Progenitors Is Developmentally Regulated. J.I., 2012, 188(9):4385–93., DOI 10.4049/ jimmunol.1101116 Erbani J, Tay J, Barbier V, Levesque J-P,Winkler IG. Acute Myeloid Leukemia Chemo-Resistance Is Mediated by E-Selectin Receptor CD162 in Bone Marrow Niches. Front Cel Dev. Biol., 2020, 8:668., DOI 10.3389/fcell.2020.00668 Molnar Z, Banlaki Z, Somogyi A, Herold Z, Herold M, Guttman A, et al. Diabetes-specific Modulation of Peripheral Blood Gene Expression Signatures in Colorectal Cancer. Cmm, 2021, 20(10):773–80., DOI 10.2174/ 1566524020666200504084626 Sanati N, Iancu OD, Wu G, Jacobs JE, McWeeney SK. Network-Based Predictors of Progression in Head and Neck Squamous Cell Carcinoma. Front Genet, 2018, 9:183., DOI 10.3389/fgene.2018.00183 Belmonte B, Cancila V, Gulino A, Navari M, Arancio W, Macor P, et al. Constitutive PSGL-1 Correlates with CD30 and TCR Pathways and Represents a Potential Target for Immunotherapy in Anaplastic Large T-Cell Lymphoma. Cancers, 2021, 13(12):2958., DOI 10.3390/cancers13122958 Sun S, Shi R, Xu L, Sun F. Identification of Heterogeneity and Prognostic Key Genes Associated with Uveal Melanoma Using Single-Cell RNA-Sequencing Technology. Melanoma Res, 2022, 32:18–26., DOI 10.1097/ cmr.0000000000000783 Buddingh EP, Kuijjer ML, Duim RAJ, Burger H, Agelopoulos K, Myklebost O, et al. Tumor-infiltrating Macrophages Are Associated with Metastasis Suppression in High-Grade Osteosarcoma: a Rationale for Treatment with Macrophage Activating Agents. Clin Cancer Res, 2011, 17(8):2110–9., DOI 10.1158/1078-0432.CCR-10-2047 Paoloni M, Davis S, Lana S, Withrow S, Sangiorgi L, Picci P, et al. Canine Tumor Cross-Species Genomics Uncovers Targets Linked to Osteosarcoma Progression. BMC Genomics, 2009, 10:625., DOI 10.1186/1471-2164-10-625 Yu G, Wang L-G, Han Y, He Q-Y. clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters. OMICS: A J Integr Biol, 2012, 16(5): 284–7., DOI 10.1089/omi.2011.0118 Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust Enumeration of Cell Subsets from Tissue Expression Profiles. Nat Methods, 2015, 12(5):453–7., DOI 10.1038/nmeth.3337 Hoos A, Protsyuk D, Borsig L. Metastatic Growth Progression Caused by PSGL-1-Mediated Recruitment of Monocytes to Metastatic Sites. Cancer Res, 2014, 74(3):695–704., DOI 10.1158/0008-5472.CAN-13-0946 Ponnusamy K, Kohrs N, Ptasinska A, Assi SA, Herold T, Hiddemann W, et al. RUNX1/ETO Blocks Selectin-Mediated Adhesion via Epigenetic Silencing of PSGL-1. Oncogenesis, 2015, 4:e146., DOI 10.1038/oncsis.2015.6 Gruszka A, Valli D, Restelli C, Alcalay M. Adhesion Deregulation in Acute Myeloid Leukaemia. Cells, 2019, 8(1):66., DOI 10.3390/cells8010066 Luan S-L, Boulanger E, Ye H, Chanudet E, Johnson N, Hamoudi RA, et al. Primary Effusion Lymphoma: Genomic Profiling Revealed Amplification of SELPLG and CORO1C Encoding for Proteins Important for Cell Migration. J Pathol, 2010, 222(2):166–79., DOI 10.1002/path.2752 Kappelmayer J, Kiss A, Karaszi E, Veszpremi A, Jako J, Kiss C. Identification of P-Selectin Glycoprotein Ligand-1 as a Useful Marker in Acute Myeloid Leukaemias. Br J Haematol, 2001, 115(4):903–9., DOI 10.1046/j.1365- 2141.2001.03179.x Xu J, Li D, Cai Z, Zhang Y, Huang Y, Su B, et al. An Integrative Analysis of DNA Methylation in Osteosarcoma. J Bone Oncol, 2017, 9:34–40., DOI 10.1016/ j.jbo.2017.05.001 Chen XG, Ma L, Xu JX. Abnormal DNA Methylation May Contribute to the Progression of Osteosarcoma. Mol Med Rep, 2018, 17(1):193–9., DOI 10.3892/ mmr.2017.7869 Honoki K, Honoki K, Fujii H, Tohma Y, Kido A, Mori T, et al. Mesenchymal Stem Cells Promote Tumor Engraftment and Metastatic Colonization in Rat Osteosarcoma Model. Int J Oncol, 2012, 40(1):163–9., DOI 10.3892/ ijo.2011.1220 Laubli H, Borsig L. Selectins Promote Tumor Metastasis. Semin Cancer Biol, 2010, 20(3):169–77., DOI 10.1016/j.semcancer.2010.04.005 Witz IP. The Selectin-Selectin Ligand axis in Tumor Progression. Cancer Metastasis Rev, 2008, 27(1):19–30., DOI 10.1007/s10555-007-9101-z Tinoco R, Carrette F, Barraza ML, Otero DC, Magana J, Bosenberg MW, et al. PSGL-1 Is an Immune Checkpoint Regulator that Promotes T Cell Exhaustion. Immunity, 2016, 44(5):1190–203., DOI 10.1016/j.immuni.2016.04.015 Kelleher FC, O’Sullivan H. Monocytes, Macrophages, and Osteoclasts in Osteosarcoma. J Adolesc Young Adult Oncol, 2017, 6(3):396–405., DOI 10.1089/jayao.2016.0078 Cersosimo F, Lonardi S, Bernardini G, Telfer B, Mandelli GE, Santucci A, et al. Tumor-Associated Macrophages in Osteosarcoma: From Mechanisms to Therapy. Ijms, 2020, 21(15):5207., DOI 10.3390/ijms21155207 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610047 EP 1610055 DI 10.3389/pore.2022.1610047 PG 9 ER PT J AU Tuncer, BS Celik, B Odemis, AD Erciyas, KS Erdogan, SO Avsar, M Turkcan, KG Yazici, H AF Tuncer, Bugra Seref Celik, Betul Odemis, Akdeniz Demet Erciyas, Kilic Seda Erdogan, Sukruoglu Ozge Avsar, Mukaddes Turkcan, Kuru Gozde Yazici, Hulya TI miRNA Sequence Analysis in Patients With Kaposi’s Sarcoma-Associated Herpesvirus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; diagnosis; Kaposi’s sarcoma; Virus; KSHV; miRNA-seq ID prognosis; diagnosis; Kaposi’s sarcoma; Virus; KSHV; miRNA-seq AB MicroRNAs (miRNAs) are the non-coding RNAs that can both attach to the untranslated and coding sections of target mRNAs, triggering destruction or post-transcriptional alteration. miRNAs regulate various cellular processes such as immune function, apoptosis, and tumorigenesis. About 35,000 miRNAs have been discovered in the human genome. The increasing evidence suggests that the dysregulation of human miRNAs may have a role in the etiology of some disorders including cancer. Only a small sub-set of human miRNAs has functionally been validated in the pathogenesis of oncogenic viruses such as Kaposi’s sarcoma-associated herpesvirus (KSHV). KSHV is the cause of various human malignancies including primary effusion lymphoma (PEL) and Kaposi’s sarcoma (KS), which are mainly seen in AIDS patients or other immunocompromised people. We aimed to identify the miRNAs in Kaposi’s sarcoma cases, with the comparison of KSHV seropositive and seronegative tumors with the controls and in each other in Turkish Kaposi’s sarcoma patients. We performed the miRNA-sequencing at genome level in the peripheral blood mononuclear cells of 16 Kaposi’s sarcoma patients, and in 8 healthy controls matched for age, gender, and ethnicity. A total of 642 miRNA molecules with different expression profiles were identified between the patients and the healthy controls. Currently, out of 642 miRNAs, 7 miRNAs (miR-92b-3p, miR-490-3p, miR-615-3p, miR-629-5p, miR-1908, miR-3180, miR-4433b- 3p) which have not been described in the literature in the context of Kaposi’s sarcoma were addressed in the study for the first time and 9 novel miRNAs, not found previously in the database, have been detected in Kaposi’s sarcoma using the miRNA-sequencing technique. This study demonstrates the identification of differently expressed miRNAs which might be the new therapeutic targets for Kaposi’s sarcoma, that has limited treatment options and can be used in the etiology, diagnosis, and prognosis of this cancer. C1 [Tuncer, Bugra Seref] Istanbul University, Istanbul Faculty of Medicine, Oncology Institute, Department of Cancer GeneticsIstanbul, Turkey. [Celik, Betul] Istanbul University, Istanbul Faculty of Medicine, Oncology Institute, Department of Cancer GeneticsIstanbul, Turkey. [Odemis, Akdeniz Demet] Istanbul University, Istanbul Faculty of Medicine, Oncology Institute, Department of Cancer GeneticsIstanbul, Turkey. [Erciyas, Kilic Seda] Istanbul University, Istanbul Faculty of Medicine, Oncology Institute, Department of Cancer GeneticsIstanbul, Turkey. [Erdogan, Sukruoglu Ozge] Istanbul University, Istanbul Faculty of Medicine, Oncology Institute, Department of Cancer GeneticsIstanbul, Turkey. [Avsar, Mukaddes] T.C. Istanbul Aydin University, Health Services Vocational School of Higher EducationIstanbul, Turkey. [Turkcan, Kuru Gozde] Halic University, Faculty of Arts and Sciences, Department of Molecular Biology and GeneticsIstanbul, Turkey. [Yazici, Hulya] Istanbul Arel University, Arel Medical Faculty, Department of Medical Biology and GeneticsIstanbul, Turkey. RP Yazici, H (reprint author), Istanbul Arel University, Arel Medical Faculty, Department of Medical Biology and Genetics, Istanbul, Turkey. EM hy2188@istanbul.edu.tr@email.com CR Zeng Y. Principles of Micro-RNA Production and Maturation. Oncogene, 2006, 25(46):6156–62., DOI 10.1038/sj.onc.1209908 Tuncer SB, Akdeniz D, Celik B, Kilic S, Sukruoglu O, Avsar M, et al. The Expression Levels of miRNA-15a and miRNA-16-1 in Circulating Tumor Cells of Patients with Diffuse Large B-Cell Lymphoma. Mol Biol Rep, 2019, 46(1): 975–80., DOI 10.1007/s11033-018-4554-4 Engels BM, Hutvagner G. Principles and Effects of microRNA-Mediated posttranscriptional Gene Regulation. Oncogene, 2006, 25(46):6163–9., DOI 10.1038/ sj.onc.1209909 Tuncer SB, Erdogan OS, Erciyas SK, Saral MA, Celik B, Odemis DA, et al. miRNA Expression Profile Changes in the Peripheral Blood of Monozygotic Discordant Twins for Epithelial Ovarian Carcinoma: Potential New Biomarkers for Early Diagnosis and Prognosis of Ovarian Carcinoma. J Ovarian Res, 2020, 13(1):99., DOI 10.1186/s13048-020-00706-8 Wu X-J, Pu X-M, Zhao Z-F, Zhao Y-N, Kang X-J, Wu W-D, et al. The Expression Profiles of microRNAs in Kaposi’s Sarcoma. Tumor Biol, 2015, 36(1):437–46., DOI 10.1007/s13277-014-2626-1 Stefani G, Slack FJ. Small Non-coding RNAs in Animal Development. Nat Rev Mol Cell Biol, 2008, 9(3):219–30., DOI 10.1038/nrm2347 Antman K, Chang Y. Kaposi’s Sarcoma. N Engl JMed, 2000, 342(14):1027–38., DOI 10.1056/nejm200004063421407 Sternbach G, Varon J. Moritz Kaposi: Idiopathic Pigmented Sarcoma of the Skin. J Emerg Med, 1995, 13(5):671–4., DOI 10.1016/0736-4679(95, 00077-n Kaloterakis A, Papasteriades C, Filiotou A, Economidou J, Hadjiyannis S, Stratigos J. HLA in Familial and Nonfamilial Mediterranean Kaposi’s Sarcoma in Greece. Tissue Antigens, 1995, 45(2):117–9., DOI 10.1111/j.1399-0039.1995. tb02427.x Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71, 3), 209–249., DOI 10.3322/caac.21660 Morrison K,Manzano M, Chung K, SchipmaMJ, Bartom ET, Gottwein E. The Oncogenic Kaposi’s Sarcoma-Associated Herpesvirus Encodes a Mimic of the Tumor-Suppressive miR-15/16 miRNA Family. Cell Rep, 2019, 29(10): 2961–9., DOI 10.1016/j.celrep.2019.11.005 Chang Y, Cesarman E, Pessin MS, Lee F, Culpepper J, Knowles DM, et al. Identification of Herpesvirus-like DNA Sequences in AIDS-Sssociated Kaposi’s Sarcoma. Science, 1994, 266(5192):1865–9., DOI 10.1126/science. 7997879 Sen F, Tambas M, Ciftci R, Toz B, Kilic L, Bozbey HU, et al. Factors Affecting Progression-free Survival in Non-HIV-related Kaposi Sarcoma. J Dermatol Treat, 2016, 27(3):275–7., DOI 10.3109/09546634.2015.1094177 The Helsinki Declaration of the World Medical Association, Wma). Ethical Principles of Medical Research Involving Human Subjects. Pol Merkur Lekarski, 2014, 36(215):298–301. Gourzones C, Jimenez A-S, Busson P. Profiling of Epstein-Barr Virus-Encoded microRNAs in Nasopharyngeal Carcinoma Reveals Potential Biomarkers and Oncomirs. Cancer, 2012, 118(18):4634., DOI 10.1002/cncr.26514 Cai X, Lu S, Zhang Z, Gonzalez CM, Damania B, Cullen BR. Kaposi’s Sarcoma- Associated Herpesvirus Expresses an Array of Viral microRNAs in Latently Infected Cells. Proc Natl Acad Sci, 2005, 102(15):5570–5., DOI 10.1073/pnas. 0408192102 Pfeffer S, Sewer A, Lagos-QuintanaM, Sheridan R, Sander C, Grasser FA, et al. Identification of microRNAs of the Herpesvirus Family. Nat Methods, 2005, 2(4):269–76., DOI 10.1038/nmeth746 Samols MA, Skalsky RL, Maldonado AM, Riva A, Lopez MC, Baker HV, et al. Identification of Cellular Genes Targeted by KSHV-Encoded microRNAs. Plos Pathog, 2007, 3(5):e65., DOI 10.1371/journal.ppat.0030065 Gottwein E, Cai X, Cullen BR. Expression and Function of microRNAs Encoded by Kaposi’s Sarcoma-Associated Herpesvirus. Cold Spring Harbor Symposia Quantitative Biol, 2006, 71:357–64., DOI 10.1101/sqb.2006.71.004 Guo Y, Li W, Qin J, Lu C, Fan W. Kaposi’s Sarcoma-Associated Herpesvirus, KSHV)-encoded microRNAs Promote Matrix Metalloproteinases, MMPs, Expression and Pro-angiogenic Cytokine Secretion in Endothelial Cells. J Med Virol, 2017, 89(7):1274–80., DOI 10.1002/jmv.24773 Ueda K. KSHV Genome Replication and Maintenance in Latency. Adv Exp Med Biol, 2018, 1045:299–320., DOI 10.1007/978-981-10-7230-7_14 Choi HS, Jain V, Krueger B, Marshall V, Kim CH, Shisler JL, et al. Kaposi’s Sarcoma-Associated Herpesvirus, KSHV, Induces the Oncogenic miR-17-92 Cluster and Down-Regulates TGF-β Signaling. Plos Pathog, 2015, 11(11): e1005255., DOI 10.1371/journal.ppat.1005255 Qin Z, Peruzzi F, Reiss K, Dai L. Role of Host microRNAs in Kaposi’s Sarcoma- Associated Herpesvirus Pathogenesis. Viruses, 2014, 6(11):4571–80., DOI 10. 3390/v6114571 Wang W, Wang C, Jiang Y, Wu B. Effect of miR-92b on Migration, Adhesion and Invasion of Human Gastric Cancer Cell Line SGC7901. Nan Fang Yi Ke Da Xue Xue Bao, 2014, 34(12):1748–52., DOI 10.3892/ijo.2016.3480 Miyamoto Y, Mauer AS, Kumar S, Mott JL, Malhi H. Mmu-miR-615-3p Regulates Lipoapoptosis by Inhibiting C/EBP Homologous Protein. PLoS One, 2014, 9(10):e109637., DOI 10.1371/journal.pone.0109637 Zhang L-y., Liu M, Li X, Tang H. miR-490-3p Modulates Cell Growth and Epithelial to Mesenchymal Transition of Hepatocellular Carcinoma Cells by Targeting Endoplasmic Reticulum-Golgi Intermediate Compartment Protein 3, ERGIC3). J Biol Chem, 2013, 288(6):4035–47., DOI 10.1074/jbc.m112.410506 Yuan H, Gao Y.MicroRNA-1908 Is Upregulated in Human Osteosarcoma and Regulates Cell Proliferation and Migration by Repressing PTEN Expression. Oncol Rep, 2015, 34(5):2706–14., DOI 10.3892/or.2015.4242 Xia X, Li Y,Wang W, Tang F, Tan J, Sun L, et al. MicroRNA-1908 Functions as a Glioblastoma Oncogene by Suppressing PTEN Tumor Suppressor Pathway. Mol Cancer, 2015, 14:154., DOI 10.1186/s12943-015-0423-0 Guan P, Yin Z, Li X, Wu W, Zhou B. Meta-analysis of Human Lung Cancer microRNA Expression Profiling Studies Comparing Cancer Tissues with normal Tissues. J Exp Clin Cancer Res, 2012, 31:54., DOI 10.1186/1756- 9966-31-54 Xie X, Wu W, Liang L, Han S, Chen T, Pan S, et al. Prognostic Role of microRNA-210 in Various Carcinomas: a Meta-Analysis. Int J Clin Exp Med, 2015, 8(9):15283–9. Tang X, Chen L, Yan X, Li Y, Xiong Y, Zhou X. Overexpression of miR-210 Is Associated with Poor Prognosis of Acute Myeloid Leukemia. Med Sci Monit, 2015, 21:3427–33., DOI 10.12659/msm.894812 Wang T, Ji F, Dai Z, Xie Y, Yuan D. Increased Expression of microRNA-191 as a Potential Serum Biomarker for Diagnosis and Prognosis in Human Osteosarcoma. Cbm, 2015, 15(5):543–50., DOI 10.3233/cbm-150493 Zhang X-F, Li K-k., Gao L, Li S-Z, Chen K, Zhang J-B, et al. miR-191 Promotes Tumorigenesis of Human Colorectal Cancer through Targeting C/EBPβ. Oncotarget, 2015, 6(6):4144–58., DOI 10.18632/oncotarget.2864 Liu H, Xu X-F, Zhao Y, Tang M-C, Zhou Y-Q, Lu J, et al. MicroRNA-191 Promotes Pancreatic Cancer Progression by Targeting USP10. Tumor Biol, 2014, 35(12):12157–63., DOI 10.1007/s13277-014-2521-9 Miyata R, Kakuki T, Nomura K, Ohkuni T, Ogasawara N, Takano K-i., et al. Poly(I:C, Induced microRNA-146a Regulates Epithelial Barrier and Secretion of Proinflammatory Cytokines in Human Nasal Epithelial Cells. Eur J Pharmacol, 2015, 761:375–82., DOI 10.1016/j.ejphar.2015.04.031 Nie J, Liu L, Zheng W, Chen L, Wu X, Xu Y, et al. microRNA-365, Down- Regulated in colon Cancer, Inhibits Cell Cycle Progression and Promotes Apoptosis of colon Cancer Cells by Probably Targeting Cyclin D1 and Bcl-2. Carcinogenesis, 2012, 33(1):220–5., DOI 10.1093/carcin/bgr245 Wang J, Wang X, Wu G, Hou D, Hu Q. MiR-365b-3p, Down-regulated in Retinoblastoma, Regulates Cell Cycle Progression and Apoptosis of Human Retinoblastoma Cells by Targeting PAX6. FEBS Lett, 2013, 587(12):1779–86., DOI 10.1016/j.febslet.2013.04.029 Chickooree D, Zhu K, Ram V, Wu HJ, He ZJ, Zhang S. A Preliminary Microarray Assay of the miRNA Expression Signatures in Buccal Mucosa of Oral Submucous Fibrosis Patients. J Oral Pathol Med, 2016, 45, 9), 691–697., DOI 10.1111/jop.12431 de Jong YP, Jacobson IM. Antisense Therapy for Hepatitis C Virus Infection. J Hepatol, 2014, 60(1):227–8., DOI 10.1016/j.jhep.2013.08.028 Drury RE, O’Connor D, Pollard AJ. The Clinical Application of MicroRNAs in Infectious Disease. Front Immunol, 2017, 8:1182., DOI 10.3389/fimmu.2017. 01182 Barnes D, Kunitomi M, Vignuzzi M, Saksela K, Andino R. Harnessing Endogenous miRNAs to Control Virus Tissue Tropism as a Strategy for Developing Attenuated Virus Vaccines. Cell Host & Microbe, 2008, 4(3): 239–48., DOI 10.1016/j.chom.2008.08.003 Heiss BL,Maximova OA, Pletnev AG. Insertion of microRNA Targets into the Flavivirus Genome Alters its Highly Neurovirulent Phenotype. J Virol, 2011, 85(4):1464–72., DOI 10.1128/jvi.02091-10 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610055 EP 1610064 DI 10.3389/pore.2022.1610055 PG 10 ER PT J AU Zhang, ChL Fan, K Gao, MQ Pang, B AF Zhang, Chuan-Long Fan, Kui Gao, Meng-Qi Pang, Bo TI Prognostic Value of Glasgow Prognostic Score in Non-small Cell Lung Cancer: A Systematic Review and Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE meta-analysis; non-small cell lung cancer; systematic review; prognostic; GPS ID meta-analysis; non-small cell lung cancer; systematic review; prognostic; GPS AB Background: Systemic inflammation is a key factor in tumor growth. The Glasgow Prognostic Score (GPS) has a certain value in predicting the prognosis of lung cancer. However, these results still do not have a unified direction. Methods: A systematic review and meta-analysis were performed to investigate the relationship between GPS and the prognosis of patients with non-small cell lung cancer (NSCLC). We set patients as follows: GPS = 0 vs. GPS = 1 or 2, GPS = 0 vs. GPS = 1, GPS = 0 vs. GPS = 2. We collected the hazard ratio (HR) and the 95% confidence interval (CI). Results: A total of 21 studies were included, involving 7333 patients. We observed a significant correlation with GPS and poor OS in NSCLC patients (HRGPS=0 vs. GPS=1 or 2 = 1.62, 95% CI: 1.27–2.07, p ≤ .001; HRGPS=0 vs GPS=1 = 2.14, 95% CI:1.31–3.49, p ≤ .001; HRGPS=0 vs. GPS=2 = 2.64, 95% CI: 1.45–4.82, p ≤ .001). Moreover, we made a subgroup analysis of surgery and stage. The results showed that when divided into GPS = 0 group and GPS = 1 or 2 group, the effect of high GPS on OS was more obvious in surgery (HR = 1.79, 95% CI: 1.08–2.97, p = .024). When GPS was divided into two groups (GPS = 0 and GPS = 1 or 2), the III-IV stage, higher GPS is associated with poor OS (HR = 1.73, 95% CI: 1.43–2.09, p ≤ .001). In the comparison of GPS = 0 and GPS = 1 group (HR = 1.56, 95% CI: 1.05–2.31, p = .026) and the grouping of GPS = 0 and GPS = 2(HR = 2.23, 95% CI: 1.17–4.26, p = .015), we came to the same conclusion. Conclusion: For patients with NSCLC, higher GPS is associated with poor prognosis, and GPS may be a reliable prognostic indicator. The decrease of GPS after pretreatment may be an effective way to improve the prognosis of NSCLC. C1 [Zhang, Chuan-Long] China Academy of Chinese Medical Sciences, Guang’anmen Hospital, International Medical DepartmentBeijing, China. [Fan, Kui] Integrated TCM-WM Hebei, Cangzhou Hospital, Department of Radiation OncologyCangzhou, China. [Gao, Meng-Qi] Shandong University of Traditional Chinese Medicine, College of Traditional Chinese MedicineJinan, China. [Pang, Bo] China Academy of Chinese Medical Sciences, Guang’anmen Hospital, International Medical DepartmentBeijing, China. RP Pang, B (reprint author), China Academy of Chinese Medical Sciences, Guang’anmen Hospital, International Medical Department, Beijing, China. EM pangbotommy@gmail.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Barta JA, Powell CA, Wisnivesky JP. Global Epidemiology of Lung Cancer. Ann Glob Health, 2019, 85., DOI 10.5334/aogh.2419 Siegel RL,Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA A Cancer J Clin, 2021, 71:7–33., DOI 10.3322/caac.21654 Chan JE, Mann AK, Kapp DS, Rehkopf DH. Income, Inflammation and Cancer Mortality: a Study of U.S. National Health and Nutrition Examination Survey Mortality Follow-Up Cohorts. BMC Public Health, 2020, 20:1805., DOI 10.1186/s12889-020-09923-8 Zitvogel L, Pietrocola F, Kroemer G. Nutrition, Inflammation and Cancer. Nat Immunol, 2017, 18:843–50., DOI 10.1038/ni.3754 McMillan DC. Systemic Inflammation, Nutritional Status and Survival in Patients with Cancer. Curr Opin Clin Nutr Metab Care, 2009, 12:223–6., DOI 10.1097/MCO.0b013e32832a7902 Forrest LM,McMillan DC, McArdle CS, Angerson WJ, Dunlop DJ. Evaluation of Cumulative Prognostic Scores Based on the Systemic Inflammatory Response in Patients with Inoperable Non-small-cell Lung Cancer. Br J Cancer, 2003, 89:1028–30., DOI 10.1038/sj.bjc.6601242 Kishi T, Matsuo Y, Ueki N, Iizuka Y, Nakamura A, Sakanaka K, et al. Pretreatment Modified Glasgow Prognostic Score Predicts Clinical Outcomes after Stereotactic Body Radiation Therapy for Early-Stage Nonsmall Cell Lung Cancer. Int J Radiat Oncology*Biology*Physics, 2015, 92: 619–26., DOI 10.1016/j.ijrobp.2015.02.018 Yang J-R, Xu J-Y, Chen G-C, Yu N, Yang J, Zeng D-X, et al. Post-diagnostic C-Reactive Protein and Albumin Predict Survival in Chinese Patients with Non-small Cell Lung Cancer: a Prospective Cohort Study. Sci Rep, 2019, 9: 8143., DOI 10.1038/s41598-019-44653-x Shoji F, Matsubara T, Kozuma Y, Haratake N, Akamine T, Takamori S, et al. Preoperative Geriatric Nutritional Risk Index: A Predictive and Prognostic Factor in Patients with Pathological Stage I Non-small Cell Lung Cancer. Surg Oncol, 2017, 26:483–8., DOI 10.1016/j.suronc.2017.09.006 Watanabe I, Kanauchi N, Watanabe H. Preoperative Prognostic Nutritional index as a Predictor of Outcomes in Elderly Patients after Surgery for Lung Cancer. Jpn J Clin Oncol, 2018, 48:382–7., DOI 10.1093/jjco/hyy014 Mandaliya H, Jones M, Oldmeadow C, Nordman IIC. Prognostic Biomarkers in Stage IV Non-small Cell Lung Cancer, NSCLC): Neutrophil to Lymphocyte Ratio, NLR), Lymphocyte to Monocyte Ratio, LMR), Platelet to Lymphocyte Ratio, PLR, and Advanced Lung Cancer Inflammation index, ALI). Transl Lung Cancer Res, 2019, 8:886–94., DOI 10.21037/tlcr.2019.11.16 Jafri SH, Shi R, Mills G. Advance Lung Cancer Inflammation index, ALI, at Diagnosis Is a Prognostic Marker in Patients with Metastatic Non-small Cell Lung Cancer, NSCLC): a Retrospective Review. BMC Cancer, 2013, 13:158., DOI 10.1186/1471-2407-13-158 Forrest LM, McMillan DC, McArdle CS, Angerson WJ, Dunlop DJ. Comparison of an Inflammation-Based Prognostic Score, GPS, with Performance Status, ECOG, in Patients Receiving Platinum-Based Chemotherapy for Inoperable Non-small-cell Lung Cancer. Br J Cancer, 2004, 90:1704–6., DOI 10.1038/sj.bjc.6601789 Takamori S, Takada K, Shimokawa M, Matsubara T, Fujishita T, Ito K, et al. Clinical Utility of Pretreatment Glasgow Prognostic Score in Non-small-cell Lung Cancer Patients Treated with Immune Checkpoint Inhibitors. Lung Cancer, 2021, 152:27–33., DOI 10.1016/j.lungcan.2020.11.026 Kikuchi R, Takoi H, Tsuji T, Nagatomo Y, Tanaka A, Kinoshita H, et al. Glasgow Prognostic Score Predicts Chemotherapy-triggered Acute Exacerbation-interstitial Lung Disease in Patients with Non-small Cell Lung Cancer. Thorac Cancer, 2021, 12:667–75., DOI 10.1111/1759-7714.13792 Lindenmann J, Fink-Neuboeck N, Taucher V, Pichler M, Posch F, Brcic L, et al. Prediction of Postoperative Clinical Outcomes in Resected Stage I Non-small Cell Lung Cancer Focusing on the Preoperative Glasgow Prognostic Score. Cancers, 2020, 12:152., DOI 10.3390/cancers12010152 Kasahara N, Imai H, Naruse I, Tsukagoshi Y, Kotake M, Sunaga N, et al. Glasgow Prognostic Score Predicts Efficacy and Prognosis in Patients with Advanced Non-small Cell Lung Cancer Receiving EGFR-TKI Treatment. Thorac Cancer, 2020, 11:2188–95., DOI 10.1111/1759-7714.13526 Topkan E, Bolukbasi Y, Ozdemir Y, Besen AA, Mertsoylu H, Selek U. Prognostic Value of Pretreatment Glasgow Prognostic Score in Stage IIIB Geriatric Non-small Cell Lung Cancer Patients Undergoing Radical Chemoradiotherapy. J GeriatrOncol, 2019, 10:567–72., DOI 10.1016/j.jgo.2018.10.014 Kasahara N, Sunaga N, Tsukagoshi Y, Miura Y, Sakurai R, Kitahara S, et al. Post-treatment Glasgow Prognostic Score Predicts Efficacy in Advanced Nonsmall- cell Lung Cancer Treated with Anti-PD1. Anticancer Res, 2019, 39: 1455–61., DOI 10.21873/anticanres.13262 Tomita M, Ayabe T, Maeda R, Nakamura K. Comparison of Inflammation- Based Prognostic Scores in Patients Undergoing Curative Resection for Nonsmall Cell Lung Cancer. World J Oncol, 2018, 9:85–90., DOI 10.14740/ wjon1097w Ni X-F, Wu J, Ji M, Shao Y-J, Xu B, Jiang J-T, et al. Effect of C-Reactive Protein/ albumin Ratio on Prognosis in Advanced Non-small-cell Lung Cancer. Asia-pac J Clin Oncol, 2018, 14:402–9., DOI 10.1111/ajco.13055 Miyazaki T, Yamasaki N, Tsuchiya T, Matsumoto K, Kunizaki M, Kamohara R, et al. Ratio of C-Reactive Protein to Albumin Is a Prognostic Factor for Operable Non-small-cell Lung Cancer in Elderly Patients. Surg Today, 2017, 47:836–43., DOI 10.1007/s00595-016-1448-8 Yotsukura M, Ohtsuka T, Kaseda K, Kamiyama I, Hayashi Y, Asamura H. Value of the Glasgow Prognostic Score as a Prognostic Factor in Resectable Non-small Cell Lung Cancer. J Thorac Oncol, 2016, 11:1311–8., DOI 10.1016/j. jtho.2016.04.029 Su K, Wang X, Chi L, Liu Y, Jin L, Li W High glasgow Prognostic Score Associates with a Poor Survival in Chinese Advanced Non-small Cell Lung Cancer Patients Treated with Platinum-Based First-Line Chemotherapy. Int J Clin Exp Med, 2016, 9:16353–9. Suzuki H, Osugi J, Muto S, Matsumura Y, Higuchi M, Gotoh M. Prognostic Impact of the High-Sensitivity Modified Glasgow Prognostic Score in Patients with Resectable Non-small Cell Lung Cancer. J Can Res Ther, 2016, 12:945–51., DOI 10.4103/0973-1482.176168 Machida Y, Sagawa M, Tanaka M, Motono N, Matsui T, Usuda K, et al. Postoperative Survival According to the Glasgow Prognostic Score in Patients with Resected Lung Adenocarcinoma. Asian Pac J Cancer Prev, 2016, 17: 4677–80., DOI 10.22034/apjcp.2016.17.10.4677 Fan H, Shao Z-Y, Xiao Y-Y, Xie Z-H, Chen W, Xie H, et al. Comparison of the Glasgow Prognostic Score, GPS, and the Modified Glasgow Prognostic Score, mGPS, in Evaluating the Prognosis of Patients with Operable and Inoperable Non-small Cell Lung Cancer. J Cancer Res Clin Oncol, 2016, 142:1285–97., DOI 10.1007/s00432-015-2113-0 Miyazaki T, Yamasaki N, Tsuchiya T, Matsumoto K, KunizakiM, Taniguchi D, et al. Inflammation-based Scoring Is a Useful Prognostic Predictor of Pulmonary Resection for Elderly Patients with Clinical Stage I Non-small-cell Lung Cancer. Eur J Cardiothorac Surg, 2015, 47:e140–e145., DOI 10.1093/ejcts/ezu514 Kawashima M, Murakawa T, Shinozaki T, Ichinose J, Hino H, Konoeda C, et al. Significance of the Glasgow Prognostic Score as a Prognostic Indicator for Lung Cancer Surgery. Interact Cardiovasc Thorac Surg, 2015, 21:637–43., DOI 10.1093/icvts/ivv223 Jiang A-G, Lu H-Y. The Glasgow Prognostic Score as a Prognostic Factor in Patients with Advanced Non-small Cell Lung Cancer Treated with Cisplatin- Based First-Line Chemotherapy. J Chemother, 2015, 27:35–9., DOI 10.1179/ 1973947814Y.0000000188 Tomita M, Ayabe T, Chosa E, Nakamura K. Prognostic Significance of Preand Postoperative glasgow Prognostic Score for Patients with Non-small Cell Lung Cancer. Anticancer Res, 2014, 34:3137–40. Forrest LM, McMillan DC, McArdle CS, Angerson WJ, Dagg K, Scott HR. A Prospective Longitudinal Study of Performance Status, an Inflammation- Based Score, GPS, and Survival in Patients with Inoperable Non-small-cell Lung Cancer. Br J Cancer, 2005, 92:1834–6., DOI 10.1038/sj.bjc.6602591 Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: the PRISMA Statement. BMJ, 2009, 339:b2535., DOI 10.1136/bmj.b2535 Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR. Practical Methods for Incorporating Summary Time-To-Event Data into Meta-Analysis. Trials, 2007, 8:16., DOI 10.1186/1745-6215-8-16 Stang A. Critical Evaluation of the Newcastle-Ottawa Scale for the Assessment of the Quality of Nonrandomized Studies in Meta-Analyses. Eur J Epidemiol, 2010, 25:603–5., DOI 10.1007/s10654-010-9491-z Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring Inconsistency in Meta-Analyses. BMJ, 2003, 327:557–60., DOI 10.1136/bmj.327.7414.557 Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related Inflammation and Treatment Effectiveness. Lancet Oncol, 2014, 15:e493–e503., DOI 10.1016/ S1470-2045(14)70263-3 Sin DD, Man SFP,McWilliams A, Lam S. Progression of Airway Dysplasia and C-Reactive Protein in Smokers at High Risk of Lung Cancer. Am J Respir Crit Care Med, 2006, 173:535–9., DOI 10.1164/rccm.200508-1305OC Zhou B, Liu J, Wang Z-M, Xi T. C-reactive Protein, Interleukin 6 and Lung Cancer Risk: aMeta-Analysis. PLoS One, 2012, 7:e43075., DOI 10.1371/journal. pone.0043075 Xu M, Zhu M, Du Y, Yan B,Wang Q,Wang C, et al. Serum C-Reactive Protein and Risk of Lung Cancer: a Case-Control Study. Med Oncol, 2013, 30:319., DOI 10.1007/s12032-012-0319-4 Pletnikoff PP, Laukkanen JA, Tuomainen T-P, Kauhanen J, Rauramaa R, Ronkainen K, et al. Cardiorespiratory Fitness, C-Reactive Protein and Lung Cancer Risk: A Prospective Population-Based Cohort Study. Eur J Cancer, 2015, 51:1365–70., DOI 10.1016/j.ejca.2015.04.020 Tolia M, Tsoukalas N, Kyrgias G, Mosa E, Maras A, Kokakis I, et al. Prognostic Significance of Serum Inflammatory Response Markers in Newly Diagnosed Non-small Cell Lung Cancer before Chemoirradiation. Biomed Res Int, 20152015, 2015:1–5., DOI 10.1155/ 2015/485732 Li S-Q, Jiang Y-H, Lin J, Zhang J, Sun F, Gao Q-F, et al. Albumin-tofibrinogen Ratio as a Promising Biomarker to Predict Clinical Outcome of Non-small Cell Lung Cancer Individuals. Cancer Med, 2018, 7:1221–31., DOI 10.1002/cam4.1428 Li X, Qin S, Sun X, Liu D, Zhang B, Xiao G, et al. Prognostic Significance of Albumin-Globulin Score in Patients with Operable Non-small-cell Lung Cancer. Ann Surg Oncol, 2018, 25:3647–59., DOI 10.1245/s10434-018- 6715-z McMillan DC, Watson WS, O’Gorman P, Preston T, Scott HR, McArdle CS. Albumin Concentrations Are Primarily Determined by the Body Cell Mass and the Systemic Inflammatory Response in Cancer Patients with Weight Loss. Nutr Cancer, 2001, 39:210–3., DOI 10.1207/ S15327914nc392_8 McMillan DC. The Systemic Inflammation-Based Glasgow Prognostic Score: a Decade of Experience in Patients with Cancer. Cancer Treat Rev, 2013, 39:534–40., DOI 10.1016/j.ctrv.2012.08.003 Jin J, Hu K, Zhou Y, Li W. Clinical Utility of the Modified Glasgow Prognostic Score in Lung Cancer: A Meta-Analysis. PLoS One, 2017, 12: e0184412., DOI 10.1371/journal.pone.0184412 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610109 EP 1610118 DI 10.3389/pore.2022.1610109 PG 10 ER PT J AU Liang, Y Wang, T Gao, R Jia, X Ji, T Shi, P Xue, J Yang, A Chen, M Han, P AF Liang, Yiqian Wang, Ting Gao, Rui Jia, Xi Ji, Ting Shi, Puyu Xue, Jianjun Yang, Aimin Chen, Mingwei Han, Peng TI Fucosyltransferase 8 is Overexpressed and Influences Clinical Outcomes in Lung Adenocarcinoma Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE FUT8; lung adenocarcinoma; bioinformatics analysis; prognosis; proliferation ID FUT8; lung adenocarcinoma; bioinformatics analysis; prognosis; proliferation AB Background: Lung adenocarcinoma (LUAD), the most prevalent type of lung cancer, is often metastatic and has a poor prognosis. Recent studies have demonstrated an important role for fucosyltransferase 8 (FUT8) in carcinogenesis and cancer progression. Methods: A meta-analysis with 15 eligible datasets from Gene Expression Omnibus (GEO) was performed to explore the expression of FUT8 in LUAD. The results were further verified in The Cancer Genome Atlas (TCGA) database, followed by survival analysis using Kaplan-Meier plotter. We also validated the protein expression of FUT8 by immunohistochemistry (IHC). In vitro experiments were conducted to determine the biological effects of FUT8 in LUAD cells. Results: The meta-analysis showed the FUT8 expression in LUAD tissues was significantly higher than those in normal lung tissues [standard mean difference (SMD): 1.40; 95% confidence interval (CI): .95–1.85]. The results of TCGA database verified the expression of FUT8 increased in LUAD tissues versus normal tissues. IHC analyses indicated that the protein levels of FUT8 were up-regulated in LUAD, and elevated FUT8 expression was significantly correlated with poor prognosis in LUAD patients. Multivariable Cox regression analysis revealed that FUT8 expression was an independent prognostic factor. Besides, in vitro experiments showed that knockdown of FUT8 in LUAD cells markedly restrained cell proliferation, and stimulated cell apoptosis. Conclusion: This study indicates that increased FUT8 expression is correlated with shortened survival of LUAD patients and might favor the progression of the disease. C1 [Liang, Yiqian] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Nuclear MedicineXi’an, China. [Wang, Ting] Xi’an No. 4 Hospital, Department of Respiratory MedicineXi’an, China. [Gao, Rui] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Nuclear MedicineXi’an, China. [Jia, Xi] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Nuclear MedicineXi’an, China. [Ji, Ting] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Nuclear MedicineXi’an, China. [Shi, Puyu] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Respiratory and Critical Care MedicineXi’an, China. [Xue, Jianjun] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Nuclear MedicineXi’an, China. [Yang, Aimin] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Nuclear MedicineXi’an, China. [Chen, Mingwei] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Respiratory and Critical Care MedicineXi’an, China. [Han, Peng] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Otolaryngology-Head and Neck SurgeryXi’an, China. RP Han, P (reprint author), The First Affiliated Hospital of Xi’an Jiaotong University, Department of Otolaryngology-Head and Neck Surgery, Xi’an, China. EM penghanent@hotmail.com CR Feng R-M, Zong Y-N, Cao S-M, Xu R-H Current Cancer Situation in China: Good or Bad News from the 2018 Global Cancer Statistics? Cancer Commun, 2019, 39(1):22., DOI 10.1186/s40880-019-0368-6 Zhou M, Wang H, Zeng X, Yin P, Zhu J, Chen W, et al. Mortality, Morbidity, and Risk Factors in China and its Provinces, 1990-2017: a Systematic Analysis for the Global Burden of Disease Study 2017. The Lancet, 2019, 394(10204): 1145–58., DOI 10.1016/s0140-6736(19)30427-1 Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, et al. editors. based on November 2019 SEER data submission, posted to the SEER web site. SEER Cancer Statistics Review, 1975-2017. Bethesda, MD: National Cancer Institute, 2020). Available at: https://seer.cancer.gov/csr/ 1975_2017/, Accessed March 1, 2021). Ruiz-Ceja KA, Chirino YI Current FDA-Approved Treatments for Non-small Cell Lung Cancer and Potential Biomarkers for its Detection. Biomed Pharmacother, 2017, 90:24–37., DOI 10.1016/j.biopha.2017.03.018 Camidge DR, Pao W, Sequist LV Acquired Resistance to TKIs in Solid Tumours: Learning from Lung Cancer. Nat Rev Clin Oncol, 2014, 11(8): 473–81., DOI 10.1038/nrclinonc.2014.104 Mollicone R, Moore SEH, Bovin N, Garcia-Rosasco M, Candelier J-J, Martinez-Duncker I, et al. Activity, Splice Variants, Conserved Peptide Motifs, and Phylogeny of Two New α1,3-Fucosyltransferase Families, FUT10 and FUT11). J Biol Chem, 2009, 284(7):4723–38., DOI 10.1074/jbc. M809312200 Ma B, Simala-Grant JL, Taylor DE Fucosylation in Prokaryotes and Eukaryotes. Glycobiology, 2006, 16(12):158R–184R., DOI 10.1093/glycob/ cwl040 Becker DJ, Lowe JB Fucose: Biosynthesis and Biological Function in Mammals. Glycobiology, 2003, 13(7):41R–53R., DOI 10.1093/glycob/ cwg054 Pinho SS, Reis CA Glycosylation in Cancer: Mechanisms and Clinical Implications. Nat Rev Cancer, 2015, 15(9):540–55., DOI 10.1038/nrc3982 Abbott KL, Nairn AV, Hall EM, Horton MB, McDonald JF, Moremen KW, et al. Focused Glycomic Analysis of the N-Linked Glycan Biosynthetic Pathway in Ovarian Cancer. Proteomics, 2008, 8(16):3210–20., DOI 10.1002/ pmic.200800157 Osumi D, Takahashi M, Miyoshi E, Yokoe S, Lee SH, Noda K, et al. Core Fucosylation of E-Cadherin Enhances Cell-Cell Adhesion in Human colon Carcinoma WiDr Cells. Cancer Sci, 2009, 100(5):888–95., DOI 10.1111/j.1349- 7006.2009.01125.x Comunale MA, Lowman M, Long RE, Krakover J, Philip R, Seeholzer S, et al. Proteomic Analysis of Serum Associated Fucosylated Glycoproteins in the Development of Primary Hepatocellular Carcinoma. J Proteome Res, 2006, 5(2):308–15., DOI 10.1021/pr050328x Potapenko IO, Haakensen VD, Luders T, Helland A, Bukholm I, Sorlie T, et al. Glycan Gene Expression Signatures in normal and Malignant Breast Tissue; Possible Role in Diagnosis and Progression. Mol Oncol, 2010, 4(2):98–118., DOI 10.1016/j.molonc.2009.12.001 Okazaki A, Shoji-Hosaka E, Nakamura K, Wakitani M, Uchida K, Kakita S, et al. Fucose Depletion from Human IgG1 Oligosaccharide Enhances Binding Enthalpy and Association Rate between IgG1 and FcγRIIIa. J Mol Biol, 2004, 336(5):1239–49., DOI 10.1016/j.jmb.2004.01.007 Shinkawa T, Nakamura K, Yamane N, Shoji-Hosaka E, Kanda Y, Sakurada M, et al. The Absence of Fucose but Not the Presence of Galactose or Bisecting N-Acetylglucosamine of Human IgG1 Complex-type Oligosaccharides Shows the Critical Role of Enhancing Antibodydependent Cellular Cytotoxicity. J Biol Chem, 2003, 278(5):3466–73., DOI 10.1074/jbc.M210665200 Wang X, Gu J, Ihara H, Miyoshi E, Honke K, Taniguchi N Core Fucosylation Regulates Epidermal Growth Factor Receptor-Mediated Intracellular Signaling. J Biol Chem, 2006, 281(5):2572–7., DOI 10.1074/ jbc.M510893200 Wang X, Inoue S, Gu J, Miyoshi E, Noda K, Li W, et al. From the Cover: Dysregulation of TGF- 1 Receptor Activation Leads to Abnormal Lung Development and Emphysema-like Phenotype in Core Fucose-Deficient Mice. Proc Natl Acad Sci, 2005, 102(44):15791–6., DOI 10.1073/pnas. 0507375102 Liang Y, Han P, Wang T, Ren H, Gao L, Shi P, et al. Stage-associated Differences in the Serum N- and O-Glycan Profiles of Patients with Nonsmall Cell Lung Cancer. Clin Proteom, 2019, 16:20., DOI 10.1186/s12014-019- 9240-6 Xiong D-d., Li Z-y., Liang L, He R-q., Ma F-c., Luo D-z., et al. The LncRNA NEAT1 Accelerates Lung Adenocarcinoma Deterioration and Binds to Mir- 193a-3p as a Competitive Endogenous RNA. Cell Physiol Biochem, 2018, 48(3):905–18., DOI 10.1159/000491958 Gyorffy B, Surowiak P, Budczies J, Lanczky A Online Survival Analysis Software to Assess the Prognostic Value of Biomarkers Using Transcriptomic Data in Non-small-cell Lung Cancer. PloS one, 2013, 8(12):e82241., DOI 10.1371/journal.pone.0082241 Honma R, Kinoshita I, Miyoshi E, Tomaru U, Matsuno Y, Shimizu Y, et al. Expression of Fucosyltransferase 8 Is Associated with an Unfavorable Clinical Outcome in Non-small Cell Lung Cancers. Oncology, 2015, 88(5):298–308., DOI 10.1159/000369495 Tada K, Ohta M, Hidano S, Watanabe K, Hirashita T, Oshima Y, et al. Fucosyltransferase 8 Plays a Crucial Role in the Invasion and Metastasis of Pancreatic Ductal Adenocarcinoma. Surg Today, 2020, 50(7):767–77., DOI 10. 1007/s00595-019-01953-z Ito Y, Miyauchi A, Yoshida H, Uruno T, Nakano K, Takamura Y, et al. Expression of α1,6-fucosyltransferase, FUT8, in Papillary Carcinoma of the Thyroid: its Linkage to Biological Aggressiveness and Anaplastic Transformation. Cancer Lett, 2003, 200(2):167–72., DOI 10.1016/s0304- 3835(03)00383-5 Muinelo-Romay L, Vazquez-Martin C, Villar-Portela S, Cuevas E, Gil-Martin E, Fernandez-Briera A Expression and Enzyme Activity of α(1,6, fucosyltransferase in Human Colorectal Cancer. Int J Cancer, 2008, 123(3): 641–6., DOI 10.1002/ijc.23521 Takahashi T, Ikeda Y, Miyoshi E, Yaginuma Y, Ishikawa M, Taniguchi N ? 1,6fucosyltransferase Is Highly and Specifically Expressed in Human Ovarian Serous Adenocarcinomas. Int J Cancer, 2000, 88(6):914–9., DOI 10.1002/1097- 0215(20001215)88:6<914::aid-ijc12>3.0.co;2-1 Chen CY, Jan YH, Juan YH, Yang CJ, Huang MS, Yu CJ, et al. Fucosyltransferase 8 as a Functional Regulator of Nonsmall Cell Lung Cancer. Proc Natl Acad Sci United States America, 2013, 110(2):630–5., DOI 10. 1073/pnas.1220425110 Zhou W, Ma H, Deng G, Tang L, Lu J, Chen X Clinical Significance and Biological Function of Fucosyltransferase 2 in Lung Adenocarcinoma. Oncotarget, 2017, 8(57):97246–59., DOI 10.18632/oncotarget.21896 Zhao Y, Itoh S, Wang X, Isaji T, Miyoshi E, Kariya Y, et al. Deletion of Core Fucosylation on α3β1 Integrin Down-Regulates its Functions. J Biol Chem, 2006, 281(50):38343–50., DOI 10.1074/jbc.M608764200 Wang Y, Fukuda T, Isaji T, Lu J, Im S, Hang Q, et al. Loss of αl,6- fucosyltransferase Inhibits Chemical-induced Hepatocellular Carcinoma and Tumorigenesis by Down-regulating Several Cell Signaling Pathways. FASEB j., 2015, 29(8):3217–27., DOI 10.1096/fj.15-270710 Tu C-F, Wu M-Y, Lin Y-C, Kannagi R, Yang R-B FUT8 Promotes Breast Cancer Cell Invasiveness by Remodeling TGF-β Receptor Core Fucosylation. Breast Cancer Res, 2017, 19(1):19., DOI 10.1186/s13058- 017-0904-8 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610116 EP 1610127 DI 10.3389/pore.2022.1610116 PG 12 ER PT J AU Micsik, T Jakab, A Lehoczki, Cs Patai, AF Micsik, Tamas Jakab, Anna Lehoczki, Csaba Patai, V. Arpad TI Traditional Serrated Adenoma of the Gallbladder, a Case Report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE traditional serrated adenoma; gallbladder; dysplasia; serrated-pathway; TSA ID traditional serrated adenoma; gallbladder; dysplasia; serrated-pathway; TSA AB While overwhelming majority of laparoscopic cholecystectomy specimens performed for gallstones or cholecystitis show rather typical findings, sometimes polypoid structures are also removed. These can be related to cholesterolosis or conventional adenomas, but occasionally extraordinary findings do emerge. In our case, a 67-year old lady with typical complaints of cholecystitis underwent routine laparoscopic cholecystectomy. Preoperative ultrasound revealed a polypoid mass with inflammation and without suspicion for malignancy. Microscopic examination showed partly conventional, lowgrade dysplastic crypts forming a villous and rather complex structure. Ectopic crypt foci, slit-like serration pattern and serrated dysplasia with eosinophylic cytoplasm and centrally located nuclei were seen throughout the lesion, thus a traditional serrated adenoma (TSA) of the gallbladder was diagnosed. TSA represents the rarest subtype of serrated lesions in the colon and extracolonic manifestations are sporadically reported. Until now only a single case of a serrated adenoma was reported from the gallbladder. Here we describe the detailed clinical, pathological and molecular findings of our case and discuss these in the light of current literature data regarding this field. C1 [Micsik, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Jakab, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Lehoczki, Csaba] Bajcsy Hospital, Department of SurgeryBudapest, Hungary. [Patai, V. Arpad] Semmelweis University, Interdisciplinary Gastroenterology Working GroupBudapest, Hungary. RP Micsik, T (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM micsikt@gmail.com CR Goldin RD, Roa JC. Gallbladder Cancer: a Morphological and Molecular Update. Histopathology, 2009, 55(2):218–29., DOI 10.1111/j.1365-2559.2008. 03192.x Matlok M, Migaczewski M, Major P, Pedziwiatr M, Budzynski P, WiniarskiM, et al. Laparoscopic Cholecystectomy in the Treatment of Gallbladder Polypoid Lesions - 15 Years of Experience. Polski przeglad chirurgiczny, 2013, 85(11): 625–9., DOI 10.2478/pjs-2013-0094 Terada T. Histopathologic Features and Frequency of Gall Bladder Lesions in Consecutive 540 Cholecystectomies. Int J Clin Exp Pathol, 2013, 6(1):91–6. Albores-Saavedra J, Chable-Montero F, Gonzalez-Romo MA, Ramirez Jaramillo M, Henson DE. Adenomas of the Gallbladder. Morphologic Features, Expression of Gastric and Intestinal Mucins, and Incidence of High-Grade Dysplasia/carcinoma In Situ and Invasive Carcinoma. Hum Pathol, 2012, 43(9):1506–13., DOI 10.1016/j.humpath.2011.11.011 Cheng Y, Jia Q, Xiong X, Ye H, Cheng N. Gallbladder Tubulovillous Adenoma in a Patient with Liver Fluke Infection. J Gastrointestin Liver Dis, 2013, 22(4):374. Xu Y, Yuan J, Chong V, Ding Z. Villous Adenoma of Gallbladder in a Patient with Systemic Lupus Erythematosus. J Res Med Sci, 2012, 17(8):811–3. Jin W, Zhang C, Liu J, Zhao Z. Large Villous Adenoma of Gallbladder: A Case Report. Int J Surg case Rep, 2013, 4(2):175–7., DOI 10.1016/j.ijscr.2012.11.018 Bettington ML, Walker NI, Rosty C, Brown IS, Clouston AD, McKeone DM, et al. A Clinicopathological and Molecular Analysis of 200 Traditional Serrated Adenomas. Mod Pathol, 2015, 28(3):414–27., DOI 10.1038/modpathol.2014.122 RKMM Pai C Rosty, editors. Colorectal Serrated Lesions and Polyps, Vol. Digestive System Tumours: WHOClassification of Tumours. 5th ed. edn, Vol. 1. Lyon, France: World Health Organization, 2019). Bettington ML, Chetty R. Traditional Serrated Adenoma: an Update. Hum Pathol, 2015, 46(7):933–8., DOI 10.1016/j.humpath.2015.04.002 Chetty R, Hafezi-Bakhtiari S, Serra S, Colling R, Wang LM. Traditional Serrated Adenomas, TSAs, Admixed with Other Serrated, So-called Precursor, Polyps and Conventional Adenomas: a Frequent Occurrence. J Clin Pathol, 2015, 68(4):270–3., DOI 10.1136/jclinpath-2014-202827 Yoon JY, Kim HT, Hong SP, Kim HG, Kim J-O, Yang D-H, et al. High-risk Metachronous Polyps AreMore Frequent in Patients with Traditional Serrated Adenomas Than in Patients with Conventional Adenomas: a Multicenter Prospective Study. Gastrointest Endosc, 2015, 82(6):1087–93. e1083., DOI 10. 1016/j.gie.2015.05.016 Wiland HO, Shadrach B, Allende D, Carver P, Goldblum JR, Liu X, et al. Morphologic and Molecular Characterization of Traditional Serrated Adenomas of the Distal colon and Rectum. Am J Surg Pathol, 2014, 38(9): 1290–7., DOI 10.1097/pas.0000000000000253 Rubio C. Traditional Serrated Adenomas of the Upper Digestive Tract. J Clin Pathol, 2016, 69(1):1–5., DOI 10.1136/jclinpath-2015-203258 Kiremitci S, Ersoz C, Ersoz C, Savas B, Ensari A. Gastric and Small Intestinal Traditional Serrated Adenomas: a Detailed Morphologic and Immunohistochemical Analysis. Turk J Gastroenterol, 2020, 31(6):441–50., DOI 10.5152/tjg.2020.19931 Rubio CA, Grimelius L, Von Sivers K, Hoog A. Intraductal Serrated Adenoma of the Pancreas. A Case Report. Anticancer Res, 2005, 25(4):3099–102. Rubio CA, Bjork J. Serrated Adenoma of the Stomach: Case Report and Literature Review. Wjge, 2013, 5(5):261–4., DOI 10.4253/wjge.v5.i5.261 Rubio CA, Schmidt PT. An Additional Case of Gastric Serrated Adenoma. Anticancer Res, 2014, 34(6):3007–10. Rubio CA. Serrated Adenoma of the Gallbladder: a Case Report. Anticancer Res, 2015, 35(6):3485–7. Pai RK, Bettington M, Srivastava A, Rosty C. An Update on the Morphology and Molecular Pathology of Serrated Colorectal Polyps and Associated Carcinomas. Mod Pathol, 2019, 32(10):1390–415., DOI 10.1038/s41379-019- 0280-2 Klimstra DSLA, Paraids V, Schirmacher P. WHO Digestive System Tumours. In: B WCoTE, editor. WHO Classification of Tumours. 5th ed., Vol. 1. Lyon, France: WHO, 2019). p. 265–92. Cansiz Ersoz C, Kiremitci S, Savas B, Ensari A. Differential Diagnosis of Traditional Serrated Adenomas and Tubulovillous Adenomas: a Compartmental Morphologic and Immunohistochemical Analysis. Acta Gastroenterol Belg, 2020, 83(4):549–56. O’Brien MJ, Zhao Q, Yang S. Colorectal Serrated Pathway Cancers and Precursors. Histopathology, 2015, 66(1):49–65., DOI 10.1111/his.12564 Patai AV, Molnar B, Tulassay Z, Sipos F. Serrated Pathway: Alternative Route to Colorectal Cancer. Wjg, 2013, 19(5):607–15., DOI 10.3748/wjg.v19.i5.607 Rubio CA, Tanaka K, Befrits R. Traditional Serrated Adenoma in a Patient with Barrett’s Esophagus. Anticancer Res, 2013, 33(4):1743–5. Rubio CA. Serrated Adenoma of the Duodenum. J Clin Pathol, 2004, 57(11): 1219–21., DOI 10.1136/jcp.2004.016360 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610133 EP 1610137 DI 10.3389/pore.2022.1610133 PG 5 ER PT J AU Niwa, A Tomita, H Watanabe, N Kiriyama, Sh Hara, A Tanaka, T AF Niwa, Ayumi Tomita, Hiroyuki Watanabe, Naoki Kiriyama, Shunya Hara, Akira Tanaka, Takuji TI Case Report: A Case of Gallbladder Carcinosarcoma With Osteoclast-like Multinucleated Giant Cells that Was Associated With RANK-RANKL Signaling SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE case report; gallbladder; carcinosarcoma; osteoclast-like multinucleated giant cell; RANK-RANKL signaling ID case report; gallbladder; carcinosarcoma; osteoclast-like multinucleated giant cell; RANK-RANKL signaling AB Introduction: Gallbladder carcinosarcoma with osteoclast-like multinucleated giant cells is known to be most uncommon form of gallbladder cancer. Owing to its rarity, the pathogenesis of gallbladder carcinosarcoma with osteoclast-like multinucleated giant cells is largely unknown. Case Presentation: We present a case of carcinosarcoma with osteoclast-like multinucleated giant cells in the gallbladder. A 57-year-old woman visited our hospital due to jaundice. An examination revealed calculous cholecystitis and gallbladder carcinoma. After cholecystectomy, macroscopic examination disclosed one whitish mass and another distinct brown and pendulous mass in the body of the gallbladder. A pathological examination revealed that each mass had a different histological type: adenosquamous carcinoma and carcinosarcoma with osteoclast-like multinucleated giant cells. Immunohistochemistry revealed that these osteoclast-like multinucleated giant cells are CD68(+), CD163(−), and MIB-1(−). In addition, the osteoclast-like multinucleated giant cells showed the strong expression of RANK and sarcoma cells around the osteoclast-like multinucleated giant cells, were positive for RANKL. Furthermore, RUNX2 was positive for some sarcoma cells. The result indicated that osteoclastic and osteoblast-like differentiation occurred in our case. Conclusion: To our knowledge, this is the first case to show the interaction of RANKRANKL signaling in gallbladder carcinosarcoma with osteoclast-like multinucleated giant cells. C1 [Niwa, Ayumi] Gifu University, Graduate School of Medicine, Department of Tumor PathologyGifu, Japan. [Tomita, Hiroyuki] Gifu University, Graduate School of Medicine, Department of Tumor PathologyGifu, Japan. [Watanabe, Naoki] Gifu Municipal Hospital, Research Center of Diagnostic Pathology (RC-DiP), Department of Diagnostic Pathology (DDP)Gifu, Japan. [Kiriyama, Shunya] Gifu Municipal Hospital, Department of SurgeryGifu, Japan. [Hara, Akira] Gifu University, Graduate School of Medicine, Department of Tumor PathologyGifu, Japan. [Tanaka, Takuji] Gifu Municipal Hospital, Research Center of Diagnostic Pathology (RC-DiP), Department of Diagnostic Pathology (DDP)Gifu, Japan. RP Tomita, H (reprint author), Gifu University, Graduate School of Medicine, Department of Tumor Pathology, Gifu, Japan. EM h_tomita@gifu-u.ac.jp CR Reid KM, Ramos-De la Medina A, Donohue JH. Diagnosis and Surgical Management of Gallbladder Cancer: A Review. J Gastrointest Surg, 2007, 11(5):671–81., DOI 10.1007/s11605-006-0075-x Rahman R, Simoes EJ, Schmaltz C, Jackson CS, Ibdah JA. Trend Analysis and Survival of PrimaryGallbladderCancer in theUnited States: a 1973-2009 Population- Based Study. Cancer Med, 2017, 6(4):874–80., DOI 10.1002/cam4.1044 Stinton LM, Shaffer EA. Epidemiology of Gallbladder Disease: Cholelithiasis and Cancer. Gut Liver, 2012, 6(2):172–87., DOI 10.5009/gnl.2012.6.2.172 Taskin OC, Akkas G, Memis B, Seven IE, Basturk O, Jang K-T, et al. Sarcomatoid Carcinomas of the Gallbladder: Clinicopathologic Characteristics. Virchows Arch, 2019, 475(1):59–66., DOI 10.1007/s00428-019-02583-z Zhang L, Chen Z, Fukuma M, Lee LY, Wu M. Prognostic Significance of Race and Tumor Size in Carcinosarcoma of Gallbladder: a Meta-Analysis of 68 Cases. Int J Clin Exp Pathol, 2008, 1(1):75 Terasaki M, Terasaki Y,Wakamatsu K, Kuwahara N, Yoneyama K, Kawase R, et al. Uterine Leiomyosarcomas with Osteoclast-like Giant Cells Associated with High Expression of RUNX2 and RANKL. Virchows Arch, 2021, 478(5): 893–904., DOI 10.1007/s00428-020-02996-1 Renema N, Navet B, Heymann MF, Lezot F, Heymann D. RANK-RANKL Signalling in Cancer. Biosci Rep, 2016, 36:36., DOI 10.1042/BSR20160150 Gibbons CLMH, Sun SG, Vlychou M, Kliskey K, Lau YS, Sabokbar A, et al. Osteoclast-like Cells in Soft Tissue Leiomyosarcomas. Virchows Arch, 2010, 456(3):317–23., DOI 10.1007/s00428-010-0882-z Terasaki M, Terasaki Y, Yoneyama K, Kuwahara N, Wakamatsu K, Nagahama K, et al. Uterine Leiomyosarcoma with Osteoclast-like Giant Cells Associated with High Expression of Receptor Activator of Nuclear Factor κB Ligand. Hum Pathol, 2015, 46(11):1679–84., DOI 10.1016/j.humpath.2015.04.018 Ikeda T, Seki S, Maki M, Noguchi N, Kawamura T, Arii S, et al. Hepatocellular Carcinoma with Osteoclast-like Giant Cells: Possibility of Osteoclastogenesis by Hepatocyte-Derived Cells. Pathol Int, 2003, 53(7):450–6., DOI 10.1046/j. 1440-1827.2003.01503.x Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi K, et al. Targeted Disruption of Cbfa1 Results in a Complete Lack of Bone Formation Owing to Maturational Arrest of Osteoblasts. Cell., 1997, 89(5):755–64., DOI 10. 1016/s0092-8674(00)80258-5 Ikebuchi Y, Aoki S, Honma M, Hayashi M, Sugamori Y, Khan M, et al. Coupling of Bone Resorption and Formation by RANKL Reverse Signalling. Nature, 2018, 561(7722):195–200., DOI 10.1038/s41586-018- 0482-7 Furuya Y, Hiroshima K, Wakahara T, Akimoto H, Yanagie H, Harigaya K, et al. Undifferentiated Carcinoma of the Gallbladder with Endothelial Differentiation: A Case Report and Literature Review. Mol Clin Oncol, 2016, 5(6):773–6., DOI 10.3892/mco.2016.1046 Mochizuki K, Hata H, Naitou K, Motosugi U, Kondo T. Carcinosarcoma, Adenocarcinoma, Neuroendocrine Carcinoma, Undifferentiated Carcinoma and Chondrosarcoma, of the Gallbladder. Clin J Gastroenterol, 2020, 13(1): 110–5., DOI 10.1007/s12328-019-01012-7 Kubota K, Kakuta Y, Kawamura S, Abe Y, Inamori M, Kawamura H, et al. Undifferentiated Spindle-Cell Carcinoma of the Gallbladder: an Immunohistochemical Study. J Hepatobiliary Pancreat Surg, 2006, 13(5): 468–71., DOI 10.1007/s00534-006-1100-x Wada Y, Takami Y, Tateishi M, Ryu T, Mikagi K, Momosaki S, et al. Carcinosarcoma of the Gallbladder: Report of a Case. Clin J Gastroenterol, 2014, 7(5):455–9., DOI 10.1007/s12328-014-0522-2 Roppongi T, Takeyoshi I, Ohwada S, Sato Y, Fujii T, Honma M, et al. Minute Squamous Cell Carcinoma of the Gallbladder: a Case Report. Jpn J Clin Oncol, 2000, 30(1):43–5., DOI 10.1093/jjco/hyd010 Nishihara K, Nagai E, Izumi Y, Yamaguchi K, TsuneyoshiM. Adenosquamous Carcinoma of the Gallbladder: A Clinicopathological, Immunohistochemical and Flow-Cytometric Study of Twenty Cases. Gann, 1994, 85(4):389–99., DOI 10.1111/j.1349-7006.1994.tb02372.x Grosso LF, Gonzalez JG. Stromal Osteoclast-like Giant Cells in an Adenosquamous Carcinoma of the Gallbladder. Hum Pathol, 1992, 23(6): 703–6., DOI 10.1016/0046-8177(92)90328-z Akatsu T, Kameyama K, Kawachi S, Tanabe M, Aiura K, Wakabayashi G, et al. Gallbladder Carcinoma with Osteoclast-like Giant Cells. J Gastroenterol, 2006, 41(1):83–7., DOI 10.1007/s00535-005-1726-5 Albores-Saavedra J, Grider DJ, Wu J, Henson DE, Goodman ZD. Giant Cell Tumor of the Extrahepatic Biliary Tree. Am J Surg Pathol, 2006, 30(4): 495–500., DOI 10.1097/00000478-200604000-00010 Manouras A, Genetzakis M, Lagoudianakis EE, Markogiannakis H, Papadima A, Agrogiannis G, et al. Undifferentiated Giant Cell Type Carcinoma of the Gallbladder with Sarcomatoid Dedifferentiation: a Case Report and Review of the Literature. J Med Case Rep, 2009, 3: 6496., DOI 10.1186/1752-1947-3-6496 Xiao Y, Xiang C, Yang D, Zhao B, Li Y, Yin H. Targeted-gene Sequencing of an Undifferentiated Gallbladder Carcinoma: a Case Report. Diagn Pathol, 2020, 15(1):66., DOI 10.1186/s13000-020-00981-5 Shishido-Hara Y, Kurata A, Fujiwara M, Itoh H, Imoto S, Kamma H. Two Cases of Breast Carcinoma with Osteoclastic Giant Cells: Are the Osteoclastic Giant Cells Pro-tumoural Differentiation of Macrophages? Diagn Pathol, 2010, 5:55., DOI 10.1186/1746-1596-5-55 Tajima S, Koda K, Fukayama M. Primary Leiomyosarcoma of the Breast with Prominent Osteoclastic Giant Cells: A Case Expressing Receptor Activator of NF-kappaB Ligand, RANKL). Pathol Int, 2015)., DOI 10.1111/ pin.12328 Dioscoridi L, Bisogni D, Freschi G. Hepatocellular Carcinoma with Osteoclastlike Giant Cells: Report of the Seventh Case in the Literature. Case Rep Surg, 2015, 2015:836105., DOI 10.1155/2015/836105 Cai G, Simsir A, Cangiarella J. InvasiveMammary Carcinoma with Osteoclast-like Giant Cells Diagnosed by fine-needle Aspiration Biopsy: Review of the Cytologic Literature and Distinction from Other Mammary Lesions Containing Giant Cells. Diagn Cytopathol, 2004, 30(6):396–400., DOI 10.1002/dc.20069 Hatano Y, Nakahama K-i., Isobe M, Morita I. Tumor Associated Osteoclastlike Giant Cells Promote Tumor Growth and Lymphangiogenesis by Secreting Vascular Endothelial Growth Factor-C. Biochem Biophysical Res Commun, 2014, 446(1):149–54., DOI 10.1016/j.bbrc.2014.02.113 Lau SK, Chu PG, Weiss LM. Cd163. Am J Clin Pathol, 2004, 122(5):794–801., DOI 10.1309/qhd6yfn81kqxuuh6 Branstetter DG, Nelson SD,Manivel JC, Blay J-Y, Chawla S, Thomas DM, et al. Denosumab Induces Tumor Reduction and Bone Formation in Patients with Giant-Cell Tumor of Bone. Clin Cancer Res, 2012, 18(16):4415–24., DOI 10. 1158/1078-0432.ccr-12-0578 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610134 EP 1610140 DI 10.3389/pore.2022.1610134 PG 7 ER PT J AU Kiss, A AF Kiss, L. Anna TI Inflammation in Focus: The Beginning and the End SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE EMT; inflammatory cytokines; reactive oxygen species; receptor internalization; chronic inflammation ID EMT; inflammatory cytokines; reactive oxygen species; receptor internalization; chronic inflammation AB The inflammation is an important biological response induced by various harmful stimuli, like viruses, bacterial infections, toxins, toxic compounds, tissue injury. During inflammation inflammatory cytokines and reactive oxygen species are produced. Inflammatory cytokines act on various receptors present on the plasma membrane of target cells. To initiate signaling cascade, and activate transcription factors, receptors should be internalized and enter the early endosomes, where the members of the signaling cascade can meet. The further cytoplasmic fate of the receptor plays crucial role in the progression and the course of inflammation. Usually acute inflammation removes injurious stimuli and helps to regain the normal healthy status of the organism. In contrast to this the uncontrolled chronic inflammation—stimulating other than immune cells, inducing transdifferentiation—can provide base of various serious diseases. This paper draws the attention of the long-lasting consequence of chronic inflammation, pointing out that one of the most important step in medication is to identify in time the factors initiating and maintaining inflammation. C1 [Kiss, L. Anna] Semmelweis University, 1st Department of AnatomyBudapest, Hungary. RP Kiss, A (reprint author), Semmelweis University, 1st Department of Anatomy, Budapest, Hungary. EM kiss.anna@med.semmelweis-univ.hu CR Medzhitov R. Inflammation 2010: New Adventures of an Old Flame. Cell, 2010, 140:771–6., DOI 10.1016/j.cell.2010.03.006 Jabbour HN, Sales KJ, Catalano RD, Norman JE. Inflammatory Pathways in Female Reproductive Health and Disease. Reprod, 2009, 138:903–19., DOI 10.1530/rep-09-0247 Kaminska B. MAPK Signalling Pathways as Molecular Targets for Antiinflammatory Therapy-From Molecular Mechanisms to Therapeutic Benefits. Biochim Biophys Acta, Bba, - Proteins Proteomics, 2005, 1754: 253–62., DOI 10.1016/j.bbapap.2005.08.017 Lawrence T. The Nuclear Factor NF-kB Pathway in Inflammation. CSH Perspect Biol, 2009, 1:a001651., DOI 10.1101/cshperspect.a001651 Libby P. Inflammatory Mechanisms: the Molecular Basis of Inflammation and Disease. Nutr Rev, 2007, 65:S140–6., DOI 10.1111/j.1753-4887.2007.tb00352.x Ferrero-Miliani L, Nielsen OH, Andersen PS, Girardin SE. Chronic Inflammation: Importance of NOD2 and NALP3 in Interleukin-1beta Generation. Clin Exp Immunol, 2007, 147:227–35., DOI 10.1111/j.1365- 2249.2006.03261.x Nathan C, Ding A. Nonresolving Inflammation. Cell, 2010, 140:871–82., DOI 10.1016/j.cell.2010.02.029 Zhou Y, Hong Y, Huang H. Triptolide Attenuates Inflammatory Response in Membranous Glomerulo-Nephritis Rat via Downregulation of NF-Κb Signaling Pathway. Kidney Blood Press Res, 2016, 41:901–10., DOI 10.1159/ 000452591 Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, et al. Inflammatory Responses and Inflammation-Associated Diseases in Organs. Oncotarget, 2018, 9: 7204–18., DOI 10.18632/oncotarget.23208 Palfy M, Remenyi A, Korcsmaros T. Endosomal Crosstalk: Meeting Points for Signaling Pathways. Trends Cel Biol, 2012, 22:447–56., DOI 10.1016/ j.tcb.2012.06.004 Ticha T, Lochman J, Cincalova L, Luhova L, Petrivalsky M. Redox Regulation of Plant S-Nitrosoglutathione Reductase Activity through post-translational Modifications of Cysteine Residues. Biochem Biophysical Res Commun, 2017, 494:27–33., DOI 10.1016/j.bbrc.2017.10.090 Thannickai VJ, Fanburg BL. Reactive Oxygen Spesies in Cell Signaling. Am J Physiol Lung Cel Mol Physiol, 2000, 279:1005–28. Chelomstitko MA. Role of Reactive Oxygen Species in Inflammation: a Minireview. Mosc Univ Biol Sci Bull, 2018, 73:199–202., DOI 10.3103/ S009639251804003X Flohe L, Brigelius-Flohe R, Saliou C, Traber MG, Packer L. Redox Regulation of NF-Kappa B Activation. Free Radic Biol Med, 1997, 22:1115–26., DOI 10.1016/s0891-5849(96)00501-1 Ranneh Y, Ali F, Akim AM, Hamid HA, Khazaai H, Fadel A. Crosstalk between Reactive Oxygen Species and Pro-inflammatory Markers in Developing Various Chronic Diseases: a Review. Appl Biol Chem, 2017, 60(3):327–38., DOI 10.1007/s13765-017-0285-9 Henriquez-Olguin C, Altamirano F, Valladares D, Lopez JR, Allen PD, Jaimovich E. Altered ROS Production, NF-Κb Activation and Interleukin-6 Gene Expression Induced by Electrical Stimulation in Dystrophic Mdx Skeletal Muscle Cells. Biochim Biophys Acta, Bba, - Mol Basis Dis, 2015, 1852:1410–9., DOI 10.1016/j.bbadis.2015.03.012 Leto TL, Geiszt M. Role of NOX Family NADPH Oxidases in Host Defense. Antioxid Redox Signal, 2006, 8(9-10):1549–61., DOI 10.1089/ars.2006.8.1549 Walker JG, Smith MD. The Jak-STAT Pathway in Rheumatoid Arthritis. J Rheumatol, 2005, 32:1650–3. Balic JJ, Albargy H, Luu K, Kirby FJ, Jayasekara WSN, Mansell F, et al. STAT3 Serine Phosphorylation Is Required for TLR4 Metabolic Reprogramming and IL-1β Expression. Nat Commun, 2020, 11:3816., DOI 10.1038/s41467-020- 17669-5 Wegrzyn J, Potla R, Chwae Y-J, Sepuri NBV, Zhang Q, Koeck T, et al. Function of Mitochondrial Stat3 in Cellular Respiration. Science, 2009, 323:793–7., DOI 10.1126/science.1164551 Gough DJ, Corlett A, Schlessinger K, Wegrzyn J, Larner AC, Levy DE. Mitochondrial STAT3 Supports Ras-dependent Oncogenic Transformation. Science, 2009, 324:1713–6., DOI 10.1126/science.1171721 Gough DJ, Marie IJ, Lobry C, Aifantis I, Levy DE. STAT3 Supports Experimental K-RasG12D-Induced Murine Myeloproliferative Neoplasms Dependent on Serine Phosphorylation. Blood, 2014, 124:2252–61., DOI 10.1182/blood-2013-02-484196 Boyer B, Valles AM, Edme N. Induction and Regulation of Epithelial- Mesenchymal Transitions. Biochem Pharmacol, 2000, 60:1091–9., DOI 10.1016/s0006-2952(00)00427-5 Lee JM, Dedhar S, Kalluri R, Thompson EW. The Epithelial-Mesenchymal Transition: New Insights in Signaling, Development, and Disease. J Cel Biol, 2006, 172:973–81., DOI 10.1083/jcb.200601018 Strutz F, Zeisberg M, Ziyadeh FN, Yang C-Q, Kalluri R, Muller GA, et al. Role of Basic Fibroblast Growth Factor-2 in Epithelial-Mesenchymal Transformation. Kidney Int, 2002, 61:1714–28., DOI 10.1046/j.1523- 1755.2002.00333.x Kalluri R, Weinberg RA. The Basics of Epithelial-Mesenchymal Transition. J Clin Invest, 2009, 119:1420–8., DOI 10.1172/jci39104 Savagner P. Leaving the Neighborhood: Molecular Mechanisms Involved during Epithelial-Mesenchymal Transition. BioEssays, 2001, 23:912–23., DOI 10.1002/bies.1132 Zsiros V, Kiss AL. Cellular and Molecular Events of Inflammation-Induced Transdifferentiation, EMT, and Regeneration, MET, in Mesenteric Mesothelial Cells. Inflamm Res, 2020, 69(12):1173–9., DOI 10.1007/s00011- 020-01400-7 Zsiros V, Katz S, Doczi N, Kiss AL. Autophagy Is the Key Process in the Reestablishment of the Epitheloid Phenotype during Mesenchymal-Epithelial Transition, MET). Exp Cel Res, 2017, 352:382–92., DOI 10.1016/ j.yexcr.2017.02.031 Katz S, Balogh P, Kiss AL. Mesothelial Cells Can Detach from the Mesentery and Differentiate into Macrophage-like Cells. APMIS, 2011, 119:782–93., DOI 10.1111/j.1600-0463.2011.02803.x Katz S, Balogh P, Nagy N, Kiss AL. Epithelial-To-Mesenchymal Transition Induced by Freund’s Adjuvant Treatment in Rat Mesothelial Cells: A Morphological and Immunocytochemical Study. Pathol Oncol Res, 2012, 18:641–9., DOI 10.1007/s12253-011-9489-1 Katz S, Zsiros V, Doczi N, Kiss AL. Inflammation-Induced Epithelial-To- Mesenchymal Transition and GM-CSF Treatment Stimulate Mesenteric Mesothelial Cells to Transdifferentiate into Macrophages. Inflammation, 2018, 41:1825–34., DOI 10.1007/s10753-018-0825-4 Katz S, Zsiros V, Kiss AL. Under Inflammatory Stimuli Mesenteric Mesothelial Cells Transdifferentiate into Macrophages and Produce Pro-inflammatory Cytokine IL-6. Inflamm Res, 2019, 68:525–8., DOI 10.1007/s00011-019-01247-7 Laslo P, Spooner CJ, Warmflash A, Lancki DW, Lee H-J, Sciammas R, et al. Multilineage Transcriptional Priming and Determination of Alternate Hematopoietic Cell Fates. Cell, 2006, 126:755–66., DOI 10.1016/ j.cell.2006.06.052 Krishnaraju K, Nguyen HQ, Liebermann DA, Hoffman B. The Zinc finger Transcription Factor Egr-1 Potentiates Macrophage Differentiation of Hematopoietic Cells. Mol Cel Biol, 1995, 15:5499–507., DOI 10.1128/ mcb.15.10.5499 Krishnaraju K, Hoffman B, Liebermann DA. The Zinc finger Transcription Factor Egr-1 Activates Macrophage Differentiation in M1 Myeloblastic Leukemia Cells. Blood, 1998, 92:1957–66., DOI 10.1182/blood.v92.6.1957 Krishnaraju K, Hoffman B, Liebermann DA. Early Growth Response Gene 1 Stimulates Development of Hematopoietic Progenitor Cells along the Macrophage Lineage at the Expense of the Granulocyte and Erythroid Lineages. Blood, 2001, 97:1298–305., DOI 10.1182/blood.v97.5.1298 Kay AB, Ying S, Varney V, Gaga M, Durham SR, Moqbel R, et al. Messenger RNA Expression of the Cytokine Gene Cluster, Interleukin 3, IL-3), IL-4, IL-5, and Granulocyte/macrophage colony-stimulating Factor, in Allergen-Induced Late-phase Cutaneous Reactions in Atopic Subjects. J Exp Med, 1991, 173: 775–8., DOI 10.1084/jem.173.3.775 Al-Saffar N, Khwaja HA, Kadoya Y, Revell PA. Assessment of the Role of GMCSF in the Cellular Transformation and the Development of Erosive Lesions Around Orthopaedic Implants. Am J Clin Pathol, 1996, 105:628–39., DOI 10.1093/ajcp/105.5.628 Carrieri PB, Provitera V, Rosa TD, Tartaglia G, Gorga F, Perrella O. Profile of Cerebrospinal Fluid and Serum Cytokines in Patients with Relapsing- Remitting Multiple Sclerosis. A Correlation with Clinical Activity. Immunopharmacology and Immunotoxicology, 1998, 20:373–82., DOI 10.3109/08923979809034820 Johnson JA, Bacon CM, Riedy MC, O1Shea JJ. Signaling by IL-2 and Related Cytokines: JAKs, STATs and Relationship to Immunodeficiency. J Leukocyt Biol, 1996, 60:441–52. Campbell PM, Halloran PF. T Cell Activation. In: NL Tilney, TB Strom, LC Paul, editors. Transplantation Biology: Cellular and Molecular Aspects. Philadelphia: Lippicott-Raven, 1996). p. 411–34. Hendrayani S-F, Al-Harbi B, Al-Ansari MM, Silva G, Aboussekhra A. The Inflammatory/cancer-Related IL-6/STAT3/NF-κB Positive Feedback Loop Includes AUF1 and Maintains the Active State of Breast Myofibroblasts. Oncotarget, 2016, 7:41974–85., DOI 10.18632/oncotarget.9633 Kyriakis JM, Avruch J. Mammalian Mitogen-Activated Protein Kinase Signal Transduction Pathways Activated by Stress and Inflammation. Physiol Rev, 2001, 81:807–69., DOI 10.1152/physrev.2001.81.2.807 Le Roy C, Wrana JL. Clathrin- and Non-clathrin-mediated Endocytic Regulation of Cell Signalling. Nat Rev Mol Cel Biol, 2005, 6:112–26., DOI 10.1038/nrm1571 Zsiros V, Katz S, Doczi N, Kiss AL. Endocytosis of GM-CSF Receptor β Is Essential for Signal Transduction Regulating Mesothelial-Macrophage Transition. Biochim Biophys Acta, Bba, - Mol Cel Res, 2019, 1866: 1450–62., DOI 10.1016/j.bbamcr.2019.06.005 Miaczynska M, Pelkmans L, Zerial M. Not just a Sink: Endosomes in Control of Signal Transduction. Curr Opin Cel Biol, 2004, 16:400–6., DOI 10.1016/ j.ceb.2004.06.005 Howe CL, Mobley WC. Signaling Endosome Hypothesis: A Cellular Mechanism for Long Distance Communication. J Neurobiol, 2004, 58: 207–16., DOI 10.1002/neu.10323 Polo S, Di Fiore PP. Endocytosis Conducts the Cell Signaling Orchestra. Cell, 2006, 124:897–900., DOI 10.1016/j.cell.2006.02.025 Macia E, Ehrlich M, Massol R, Boucrot E, Brunner C, Kirchhausen T. Dynasore, a Cell-Permeable Inhibitor of Dynamin. Develop Cel, 2006, 10: 839–50., DOI 10.1016/j.devcel.2006.04.002 Carrosco L, Pisa D, Alonso R. Polymicrobial Infections and Neurodegeneration. Curr Clin Micro Rpt, 2020, 7:20–30., DOI 10.1007/ s40588-020-00139-3 Miklossy J. Chronic Inflammation and Amyloidogenesis in Alzheimer’s Disease - Role of Spirochetes1. Jad, 2008, 13:381–91., DOI 10.3233/jad-2008- 13404 Vigasova D, Nemergut M, Liskova B, Damborsky J. Multi-pathogen Infections and Alzheimer’s Disease. Microb Cel Fact, 2021, 20:25., DOI 10.1186/s12934- 021-01520-7 Allan SM, Rothwell NJ. Inflammation in central Nervous System Injury. Phil Trans R Soc Lond B, 2003, 358:1669–77., DOI 10.1098/rstb.2003.1358 Pollmacher T, Haack M, Schuld A, Reichenberg A, Yirmiya R. Low Levels of Circulating Inflammatory Cytokines-Do They Affect Human Brain Functions? Brain Behav Immun, 2002, 16:525–32., DOI 10.1016/s0889- 1591(02)00004-1 Lucas S-M, Rothwell NJ, Gibson RM. The Role of Inflammation in CNS Injury and Disease. Br J Pharmacol, 2006, 147:S232–S240., DOI 10.1038/sj.bjp.0706400 Ueha S, Shand FHW, Matsushima K. Cellular and Molecular Mechanisms of Chronic Inflammation-Associated Organ Fibrosis. Front Immun, 2012, 3:71., DOI 10.3389/fimmu.2012.00071 Deng T, Lyon CJ, Bergin S, Caligiuri MA, Hsueh WA. Obesity, Inflammation, and Cancer. Annu Rev Pathol Mech Dis, 2016, 11:421–49., DOI 10.1146/ annurev-pathol-012615-044359 Picon-Ruiz M, Pan C, Drews-Elger K, Jang K, Besser AH, Zhao D, et al. Interactions between Adipocytes and Breast Cancer Cells Stimulate Cytokine Production and Drive Src/Sox2/miR-302b-Mediated Malignant Progression. Cancer Res, 2016, 76:491–504., DOI 10.1158/0008-5472.can-15-0927 Diaz-Ruano A, Martinez-Alarcon N, Ferrandiz-Cabrera I, Marchal JA, Picon- Ruiz M. Estron But Not Estradiol, Potentiates NF-kB-Driven Inflammation, EMT and Stemness in ER+epithelial Cells. Int J Mol Sci, 2019, 20:528–47. Howe LR, Subbaramaiah K, Hudis CA, Dannenberg AJ. Molecular Pathways: Adipose Inflammation as a Mediator of Obesity-Associated Cancer. Clin Cancer Res, 2013, 19:6074–83., DOI 10.1158/1078-0432.ccr-12- 2603 Picon-RuizM,Morata-Tarifa C, Valle-Goffin JJ, Friedman ER, Slingerland JM. Obesity and Adverse Breast Cancer Risk and Outcome: Mechanistic Insights and Strategies for Intervention. CA: A Cancer J Clinicians, 2017, 67:378–97., DOI 10.3322/caac.21405 Tornatore L, Thotakura AK, Bennett J, Moretti M, Franzoso G. The Nuclear Factor Kappa B Signaling Pathway: Integrating Metabolism with Inflammation. Trends Cel Biol, 2012, 22:557–66., DOI 10.1016/ j.tcb.2012.08.001 Bazdyrev E, Rusina P, Panova M, Novikov F, Grishagin I, Nebolsin V. Lung Fibrosis after COVID-19: Treatment Prospects. Pharmaceuticals, 2021, 14: 807., DOI 10.3390/ph14080807 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610136 EP 1610142 DI 10.3389/pore.2021.1610136 PG 7 ER PT J AU Yang, Ch Chen, K AF Yang, Chenbo Chen, Kuisheng TI Long Non-Coding RNA in Esophageal Cancer: A Review of Research Progress SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE biomarkers; long non-coding RNA; molecular mechanism; esophageal cancer (EC); genomics ID biomarkers; long non-coding RNA; molecular mechanism; esophageal cancer (EC); genomics AB In recent years, there has been significant progress in the diagnosis and treatment of esophageal cancer. However, owing to the lack of early diagnosis strategies and treatment targets, the prognosis of patients with esophageal cancer remains unsatisfactory. There is an urgent need to identify novel biomarkers and treatment targets for esophageal cancer. With the development of genomics, long-chain non-coding RNAs (LncRNAs), which were once considered transcriptional “noise,” are being identified and characterized rapidly in large numbers. Recent research shows that LncRNAs are closely related to a series of steps in tumor development and play an important regulatory role in DNA replication, transcription, and post-transcriptional regulation. The abnormal expression of LncRNAs leads to tumor cell proliferation, migration, invasion, and treatment resistance. This review focuses on the latest progress in research on the abnormal expression and functional mechanisms of LncRNAs in esophageal cancer. Further, it discusses the potential applications of these findings towards achieving an early diagnosis, improving treatment efficacy, and evaluating the prognosis of esophageal cancer. C1 [Yang, Chenbo] The First Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. [Chen, Kuisheng] The First Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. RP Chen, K (reprint author), The First Affiliated Hospital of Zhengzhou University, Department of Pathology, Zhengzhou, China. EM chenksh2002@163.com CR Fan J, Liu Z, Mao X, Tong X, Zhang T, Suo C, et al. Global Trends in the Incidence and Mortality of Esophageal Cancer from 1990 to 2017. Cancer Med, 2020, 9:e03338., DOI 10.1002/cam4.3338 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 68(6): 394–424., DOI 10.3322/caac.21492 Wong MCS, Hamilton W, Whiteman DC, Jiang JY, Qiao Y, Fung FDH, et al. Global Incidence and Mortality of Oesophageal Cancer and Their Correlation with Socioeconomic Indicators Temporal Patterns and Trends in 41 Countries. Sci Rep, 2018, 8(1):4522., DOI 10.1038/s41598- 018-19819-8 Ohashi S, Miyamoto Si., Kikuchi O, Goto T, Amanuma Y, Muto M. Recent Advances from Basic and Clinical Studies of Esophageal Squamous Cell Carcinoma. Gastroenterology, 2015, 149(7):1700–15., DOI 10.1053/j.gastro. 2015.08.054 Maher B. ENCODE: The Human Encyclopaedia. Nature, 2012, 489(7414): 46–8., DOI 10.1038/489046a Lee JT. Epigenetic Regulation by Long Noncoding RNAs. Science, 2012, 338(6113):1435–9., DOI 10.1126/science.1231776 Yoon J-H, Kim J, Gorospe M. Long Noncoding RNA Turnover. Biochimie, 2015, 117:15–21., DOI 10.1016/j.biochi.2015.03.001 Wang X, Sun W, Shen W, Xia M, Chen C, Xiang D, et al. Long Non-coding RNA DILC Regulates Liver Cancer Stem Cells via IL-6/STAT3 axis. J Hepatol, 2016, 64(6):1283–94., DOI 10.1016/j.jhep.2016.01.019 Zhou J, Yang L, Zhong T, Mueller M, Men Y, Zhang N, et al. H19 lncRNA Alters DNA Methylation Genome Wide by Regulating S-Adenosylhomocysteine Hydrolase. Nat Commun, 2015, 6:10221., DOI 10.1038/ncomms10221 Wu Y, Zhang L, Wang Y, Li H, Ren X, Wei F, et al. Long Noncoding RNA HOTAIR Involvement in Cancer. Tumor Biol, 2014, 35(10):9531–8., DOI 10. 1007/s13277-014-2523-7 Yang F, Zhang L, Huo X-s., Yuan J-h., Xu D, Yuan S-x., et al. Long Noncoding RNA High Expression in Hepatocellular Carcinoma Facilitates Tumor Growth through Enhancer of Zeste Homolog 2 in Humans. Hepatology, 2011, 54(5): 1679–89., DOI 10.1002/hep.24563 Thin KZ, Liu X, Feng X, Raveendran S, Tu JC, LncRNA-Dancr. LncRNADANCR: A Valuable Cancer Related Long Non-coding RNA for Human Cancers. Pathol - Res Pract, 2018, 214(6):801–5., DOI 10.1016/j.prp.2018.04.003 Qi Y, Song C, Zhang J, Guo C, Yuan C. Oncogenic LncRNA CASC9 in Cancer Progression. Curr Pharm Des, 2021, 27(4):575–82., DOI 10.2174/ 1381612826666200917150130 Ji P, Diederichs S, Wang W, Boing S, Metzger R, Schneider PM, et al. MALAT- 1, a Novel Noncoding RNA, and Thymosin β4 Predict Metastasis and Survival in Early-Stage Non-small Cell Lung Cancer. Oncogene, 2003, 22(39):8031–41., DOI 10.1038/sj.onc.1206928 Hu L, Wu Y, Tan D, Meng H, Wang K, Bai Y, et al. Up-regulation of Long Noncoding RNA MALAT1 Contributes to Proliferation and Metastasis in Esophageal Squamous Cell Carcinoma. J Exp Clin Cancer Res, 2015, 34:7., DOI 10.1186/s13046-015-0123-z Chen M, Xia Z, Chen C, Hu W, Yuan Y. LncRNA MALAT1 Promotes Epithelial-To-Mesenchymal Transition of Esophageal Cancer through Ezh2-Notch1 Signaling Pathway. Anti-Cancer Drugs, 2018, 29(8):767–73., DOI 10.1097/cad.0000000000000645 Wang X, Li M, Wang Z, Han S, Tang X, Ge Y, et al. Silencing of Long Noncoding RNA MALAT1 by miR-101 and miR-217 Inhibits Proliferation, Migration, and Invasion of Esophageal Squamous Cell Carcinoma Cells. J Biol Chem, 2015, 290(7):3925–35., DOI 10.1074/jbc.m114.596866 Tan D,Wu Y, Hu L, He P, Xiong G, Bai Y, et al. Long Noncoding RNA H19 Is Up-Regulated in Esophageal Squamous Cell Carcinoma and Promotes Cell Proliferation and Metastasis. Dis Esophagus, 2017, 30(1):1–9., DOI 10.1111/ dote.12481 Chen M-J, Deng J, Chen C, Hu W, Yuan Y-C, Xia Z-K. LncRNA H19 Promotes Epithelial Mesenchymal Transition and Metastasis of Esophageal Cancer via STAT3/EZH2 axis. Int J Biochem Cel Biol, 2019, 113:27–36., DOI 10.1016/j. biocel.2019.05.011 Yang X, Song JH, Cheng Y, Wu W, Bhagat T, Yu Y, et al. Long Non-coding RNAHNF1A-AS1regulates Proliferation and Migration in Oesophageal Adenocarcinoma Cells. Gut, 2014, 63(6):881–90., DOI 10.1136/gutjnl-2013- 305266 Wang P, Liu X, Han G, Dai S, Ni Q, Xiao S, et al. Downregulated lncRNA UCA1 Acts as ceRNA to Adsorb microRNA-498 to Repress Proliferation, Invasion and Epithelial Mesenchymal Transition of Esophageal Cancer Cells by Decreasing ZEB2 Expression. Cell Cycle, 2019, 18(18):2359–76., DOI 10. 1080/15384101.2019.1648959 Jiao C, Song Z, Chen J, Zhong J, CaiW, Tian S, et al. lncRNA-UCA1 Enhances Cell Proliferation through Functioning as a ceRNA of Sox4 in Esophageal Cancer. Oncol Rep, 2016, 36(5):2960–6., DOI 10.3892/or.2016.5121 Liu HE, Shi HH, Luo XJ. Upregulated Long Noncoding RNA UCA1 Enhances Warburg Effect via miR-203/HK2 Axis in Esophagal Cancer. J Oncol, 2020, 2020:8847687., DOI 10.1155/2020/8847687 Zhang W, Chen Q, Lei C. lncRNA MIAT Promotes Cell Invasion and Migration in Esophageal Cancer. Exp Ther Med, 2020, 19(5):3267–74., DOI 10.3892/etm.2020.8588 Zhang C, Xie L, Fu Y, Yang J, Cui Y. lncRNA MIAT Promotes Esophageal Squamous Cell Carcinoma Progression by Regulating miR-1301-3p/INCENP axis and Interacting with SOX2. J Cel Physiol, 2020, 235(11):7933–44., DOI 10. 1002/jcp.29448 Xu Y, Li Y, Jin J, Han G, Sun C, Pizzi MP, et al. LncRNA PVT1 Up-Regulation Is a Poor Prognosticator and Serves as a Therapeutic Target in Esophageal Adenocarcinoma. Mol Cancer, 2019, 18(1):141., DOI 10.1186/s12943-019- 1064-5 Hu J, GaoW. Long Noncoding RNA PVT1 Promotes Tumour Progression via the miR-128/ZEB1 axis and Predicts Poor Prognosis in Esophageal Cancer. Clin Res Hepatol Gastroenterol, 2021, 45(4):101701., DOI 10.1016/j.clinre.2021. 101701 Kasagi Y, Oki E, Ando K, Ito S, Iguchi T, Sugiyama M, et al. The Expression of CCAT2, a Novel Long Noncoding RNA Transcript, and Rs6983267 Single- Nucleotide Polymorphism Genotypes in Colorectal Cancers. Oncology, 2017, 92(1):48–54., DOI 10.1159/000452143 Yang C, Li F, Zhou W, Huang J. Knockdown of Long Non-coding RNA CCAT2 Suppresses Growth and Metastasis of Esophageal Squamous Cell Carcinoma by Inhibiting the β-catenin/WISP1 Signaling Pathway. J Int Med Res, 2021, 49(5):3000605211019938., DOI 10.1177/03000605211019938 Wang X, Wang X. Long Non-coding RNA colon Cancer-Associated Transcript 2 May Promote Esophageal Cancer Growth and Metastasis by Regulating the Wnt Signaling Pathway. Oncol Lett, 2019, 18(2):1745–54., DOI 10.3892/ol.2019.10488 Liang Y, Chen X, Wu Y, Li J, Zhang S, Wang K, et al. LncRNA CASC9 Promotes Esophageal Squamous Cell Carcinoma Metastasis through Upregulating LAMC2 Expression by Interacting with the CREB-Binding Protein. Cell Death Differ, 2018, 25(11):1980–95., DOI 10.1038/s41418-018- 0084-9 Wu Y, Hu L, Liang Y, Li J, Wang K, Chen X, et al. Up-regulation of lncRNA CASC9 Promotes Esophageal Squamous Cell Carcinoma Growth by Negatively Regulating PDCD4 Expression through EZH2. Mol Cancer, 2017, 16(1):150., DOI 10.1186/s12943-017-0715-7 Zhong X, Hu X, Zhang L. Oncogenic Long Noncoding RNA FAL1 in Human Cancer. Mol Cell Oncol, 2015, 2(2):e977154., DOI 10.4161/23723556.2014. 977154 Liu T,Wang Z, Zhou R, Liang W. Focally Amplified lncRNA on Chromosome 1 Regulates Apoptosis of Esophageal Cancer Cells via DRP1 and Mitochondrial Dynamics. IUBMB Life, 2019, 71(2):254–60., DOI 10.1002/ iub.1971 Liang XS, Sun Y, Liu T. Long Non-coding RNA FAL1 Regulated Cell Proliferation through Akt Pathway via Targeting PDK1 in Esophageal Cancer Cells. Eur Rev Med Pharmacol Sci, 2018, 22(16):5214–22., DOI 10. 26355/eurrev_201808_15719 Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, et al. Functional Demarcation of Active and Silent Chromatin Domains in Human HOX Loci by Noncoding RNAs. Cell, 2007, 129(7):1311–23., DOI 10.1016/j.cell.2007.05.022 Ma J, Fan Y, Feng T, Chen F, Xu Z, Li S, et al. HOTAIR Regulates HK2 Expression by Binding Endogenous miR-125 and miR-143 in Oesophageal Squamous Cell Carcinoma Progression. Oncotarget, 2017, 8(49):86410–22., DOI 10.18632/oncotarget.21195 Wang W, Wu D, He X, Hu X, Hu C, Shen Z, et al. CCL18-induced HOTAIR Upregulation Promotes Malignant Progression in Esophageal Squamous Cell Carcinoma through the miR-130a-5p-ZEB1 axis. Cancer Lett, 2019, 460: 18–28., DOI 10.1016/j.canlet.2019.06.009 Su W, Guo C, Wang L, Wang Z, Yang X, Niu F, et al. LncRNA MIR22HG Abrogation Inhibits Proliferation and Induces Apoptosis in Esophageal Adenocarcinoma Cells via Activation of the STAT3/c-Myc/FAK Signaling. Aging, 2019, 11(13):4587–96., DOI 10.18632/aging.102071 Li F-Z, Zang W-Q. Knockdown of lncRNAXLOC_001659 Inhibits Proliferation and Invasion of Esophageal Squamous Cell Carcinoma Cells. World J Gastroenterol, 2019, 25(42):6299–310., DOI 10.3748/wjg. v25.i42.6299 Xu LJ, Yu XJ, Wei B, Hui HX, Sun Y, Dai J, et al. Long Non-coding RNA DUXAP8 Regulates Proliferation and Invasion of Esophageal Squamous Cell Cancer. Eur Rev Med Pharmacol Sci, 2018, 22(9):2646–52., DOI 10.26355/ eurrev_201805_14959 Wang Y, Zhang W, Liu W, Huang L, Wang Y, Li D, et al. Long Noncoding RNA VESTAR Regulates Lymphangiogenesis and Lymph Node Metastasis of Esophageal Squamous Cell Carcinoma by Enhancing VEGFC mRNA Stability. Cancer Res, 2021, 81(12):3187–99., DOI 10.1158/0008-5472.can-20-1713 Ma C, Shi X, Zhu Q, Li Q, Liu Y, Yao Y, et al. The Growth Arrest-specific Transcript 5, GAS5): a Pivotal Tumor Suppressor Long Noncoding RNA in Human Cancers. Tumor Biol, 2016, 37(2):1437–44., DOI 10.1007/s13277-015- 4521-9 Wang G, Sun J, Zhao H, Li H. Long Non-coding RNA, lncRNA, Growth Arrest Specific 5, GAS5, Suppresses Esophageal Squamous Cell Carcinoma Cell Proliferation and Migration by Inactivating Phosphatidylinositol 3-kinase, PI3K)/AKT/Mammalian Target of Rapamycin, mTOR, Signaling Pathway. Med Sci Monit, 2018, 24:7689–96., DOI 10.12659/msm.910867 Huang J, Li Y, Lu Z, Che Y, Sun S, Mao S, et al. Long Non-coding RNA GAS5 Is Induced by Interferons and Plays an Antitumor Role in Esophageal Squamous Cell Carcinoma. Cancer Med, 2018, 7:3157–67., DOI 10.1002/cam4.1524 Wang K, Li J, Xiong G, He G, Guan X, Yang K, et al. Negative Regulation of lncRNA GAS5 by miR-196a Inhibits Esophageal Squamous Cell Carcinoma Growth. Biochem Biophysical Res Commun, 2018, 495(1):1151–7., DOI 10. 1016/j.bbrc.2017.11.119 Liang W-C, Ren J-L, Wong C-W, Chan S-O, Waye MM-Y, Fu W-M, et al. LncRNA-NEF Antagonized Epithelial to Mesenchymal Transition and Cancer Metastasis via Cis-Regulating FOXA2 and Inactivating Wnt/β-Catenin Signaling. Oncogene, 2018, 37(11):1445–56., DOI 10.1038/s41388-017-0041-y Zhang J, Hu SL, Qiao CH, Ye JF, Li M, Ma HM, et al. LncRNA-NEF Inhibits Proliferation, Migration and Invasion of Esophageal Squamous-Cell Carcinoma Cells by Inactivating Wnt/β-Catenin Pathway. Eur Rev Med Pharmacol Sci, 2018, 22(20):6824–31., DOI 10.26355/eurrev_201810_16150 Modarresi F, Faghihi MA, Lopez-Toledano MA, Fatemi RP, Magistri M, Brothers SP, et al. Inhibition of Natural Antisense Transcripts In Vivo Results in Gene-specific Transcriptional Upregulation. Nat Biotechnol, 2012, 30(5):453–9., DOI 10.1038/nbt.2158 Zhao H, Diao C, Wang X, Xie Y, Liu Y, Gao X, et al. LncRNA BDNF-AS Inhibits Proliferation, Migration, Invasion and EMT in Oesophageal Cancer Cells by Targeting miR-214. J Cel Mol Med, 2018, 22:3729–39., DOI 10.1111/ jcmm.13558 Dong Z, Zhang A, Liu S, Lu F, Guo Y, Zhang G, et al. Aberrant Methylation- Mediated Silencing of lncRNA MEG3 Functions as a ceRNA in Esophageal Cancer. Mol Cancer Res, 2017, 15(7):800–10., DOI 10.1158/1541-7786.mcr-16- 0385 Huang Z-L, Chen R-P, Zhou X-T, Zhan H-L, Hu M-M, Liu B, et al. Long Noncoding RNA MEG3 Induces Cell Apoptosis in Esophageal Cancer through Endoplasmic Reticulum Stress. Oncol Rep, 2017, 37(5):3093–9., DOI 10.3892/ or.2017.5568 Yao J, Zhou B, Zhang J, Geng P, Liu K, Zhu Y, et al. A New Tumor Suppressor LncRNA ADAMTS9-AS2 Is Regulated by DNMT1 and Inhibits Migration of Glioma Cells. Tumor Biol, 2014, 35(8):7935–44., DOI 10.1007/s13277-014- 1949-2 Liu D, Wu K, Yang Y, Zhu D, Zhang C, Zhao S. Long Noncoding RNA ADAMTS9-AS2 Suppresses the Progression of Esophageal Cancer by Mediating CDH3 Promoter Methylation. Mol Carcinog, 2020, 59(1):32–44., DOI 10.1002/mc.23126 Dong Z, Li S, Wu X, Niu Y, Liang X, Yang L, et al. Aberrant Hypermethylation- Mediated Downregulation of Antisense lncRNA ZNF667-AS1 and its Sense Gene ZNF667 Correlate with Progression and Prognosis of Esophageal Squamous Cell Carcinoma. Cell Death Dis, 2019, 10(12):930., DOI 10.1038/ s41419-019-2171-3 Guo W, Liu S, Dong Z, Guo Y, Ding C, Shen S, et al. Aberrant Methylation- Mediated Silencing of lncRNA CTC-276P9.1 Is Associated with Malignant Progression of Esophageal Squamous Cell Carcinoma. Clin Exp Metastasis, 2018, 35(1-2):53–68., DOI 10.1007/s10585-018-9881-2 Zhao B, Cao P, Hu S, Li F, Kong K, Zu Y. LncRNA-NBAT-1 Modulates Esophageal Cancer Proliferation via PKM2. Am J Transl Res, 2019, 11(9): 5978–87. Dong Z, Liang X, Wu X, Kang X, Guo Y, Shen S, et al. Promoter Hypermethylation-Mediated Downregulation of Tumor Suppressor Gene SEMA3B and lncRNA SEMA3B-AS1 Correlates with Progression and Prognosis of Esophageal Squamous Cell Carcinoma. Clin Exp Metastasis, 2019, 36(3):225–41., DOI 10.1007/s10585-019-09964-3 Bieging KT, Mello SS, Attardi LD. Unravelling Mechanisms of P53-Mediated Tumour Suppression. Nat Rev Cancer, 2014, 14(5):359–70., DOI 10.1038/ nrc3711 Zhang A, Xu M, Mo Y-Y. Role of the lncRNA-P53 Regulatory Network in Cancer. J Mol Cel Biol, 2014, 6(3):181–91., DOI 10.1093/jmcb/mju013 Yao J, Zhang H, Li H, Qian R, Liu P, Huang J. P53-regulated lncRNA uc061hsf.1 Inhibits Cell Proliferation and Metastasis in Human Esophageal Squamous Cell Cancer. IUBMB Life, 2020, 72(3):401–12., DOI 10.1002/iub. 2196 Wang C, Zhang Q, Hu Y, Zhu J, Yang J. Emerging Role of Long Non-coding RNA MALAT1 in Predicting Clinical Outcomes of Patients with Digestive System Malignancies: A Meta-Analysis. Oncol Lett, 2019, 17(2):2159–70., DOI 10.3892/ol.2018.9875 Gao GD, Liu XY, Lin Y, Liu HF, Zhang GJ. LncRNA CASC9 Promotes Tumorigenesis by Affecting EMT and Predicts Poor Prognosis in Esophageal Squamous Cell Cancer. Eur Rev Med Pharmacol Sci, 2018, 22(2):422–9., DOI 10. 26355/eurrev_201801_14191 Zhao Y, Wang N, Zhang X, Liu H, Yang S. LncRNA ZEB1-AS1 Down- Regulation Suppresses the Proliferation and Invasion by Inhibiting ZEB1 Expression in Oesophageal Squamous Cell Carcinoma. J Cel Mol Med, 2019, 23(12):8206–18., DOI 10.1111/jcmm.14692 Li X, Yang H, Wang J, Li X, Fan Z, Zhao J, et al. High Level of lncRNA H19 Expression Is Associated with Shorter Survival in Esophageal Squamous Cell Cancer Patients. Pathol - Res Pract, 2019, 215(11):152638., DOI 10.1016/j.prp. 2019.152638 Liu Z, Yang T, Xu Z, Cao X. Upregulation of the Long Non-coding RNA BANCR Correlates with Tumor Progression and Poor Prognosis in Esophageal Squamous Cell Carcinoma. Biomed Pharmacother, 2016, 82:406–12., DOI 10. 1016/j.biopha.2016.05.014 Kang K, Huang YH, Li HP, Guo SM. Expression of UCA1 and MALAT1 Long- Chain Non-coding RNAs in Esophageal Squamous Cell Carcinoma Tissues Is Predictive of Patient Prognosis. Arch Med Sci, 2018, 14(4):752–9., DOI 10.5114/ aoms.2018.73713 Li X, Wu Z, Mei Q, Li X, Guo M, Fu X, et al. Long Non-coding RNA HOTAIR, a Driver of Malignancy, Predicts Negative Prognosis and Exhibits Oncogenic Activity in Oesophageal Squamous Cell Carcinoma. Br J Cancer, 2013, 109(8): 2266–78., DOI 10.1038/bjc.2013.548 Wu H, Zheng J, Deng J, Zhang L, Li N, Li W, et al. LincRNA-uc002yug.2 Involves in Alternative Splicing of RUNX1 and Serves as a Predictor for Esophageal Cancer and Prognosis. Oncogene, 2015, 34(36):4723–34., DOI 10. 1038/onc.2014.400 Yan S, Du L, Jiang X, DuanW, Li J, Xie Y, et al. Evaluation of Serum Exosomal lncRNAs as Diagnostic and Prognostic Biomarkers for Esophageal Squamous Cell Carcinoma. Cancer Manag Res, 2020, 12:9753–63., DOI 10.2147/cmar. s250971 Li M, Liu L, Zhang W, Zhan H, Chen R, Feng J, et al. Long Non-coding RNA MEG3 Suppresses Epithelial-To-Mesenchymal Transition by Inhibiting the PSAT1-dependent GSK-3β/Snail Signaling Pathway in Esophageal Squamous Cell Carcinoma. Oncol Rep, 2020, 44(5):2130–42., DOI 10.3892/or.2020.7754 Zhang J, Zhao H, Gao Y, Zhang W. Secretory miRNAs as Novel Cancer Biomarkers. Biochim Biophys Acta, Bba, - Rev Cancer, 2012, 1826(1):32–43., DOI 10.1016/j.bbcan.2012.03.001 Tong Y-S, Wang X-W, Zhou X-L, Liu Z-H, Yang T-X, Shi W-H, et al. Identification of the Long Non-coding RNA POU3F3 in Plasma as a Novel Biomarker for Diagnosis of Esophageal Squamous Cell Carcinoma. Mol Cancer, 2015, 14:3., DOI 10.1186/1476-4598-14-3 Shao H, Im H, Castro CM, Breakefield X, Weissleder R, Lee H. New Technologies for Analysis of Extracellular Vesicles. Chem Rev, 2018, 118(4):1917–50., DOI 10.1021/acs.chemrev.7b00534 Prensner JR, Iyer MK, Balbin OA, Dhanasekaran SM, Cao Q, Brenner JC, et al. Transcriptome Sequencing across a Prostate Cancer Cohort Identifies PCAT- 1, an Unannotated lincRNA Implicated in Disease Progression. Nat Biotechnol, 2011, 29(8):742–9., DOI 10.1038/nbt.1914 Shi W-h., Wu Q-q., Li S-q., Yang T-x., Liu Z-h., Tong Y-s., et al. Upregulation of the Long Noncoding RNA PCAT-1 Correlates with Advanced Clinical Stage and Poor Prognosis in Esophageal Squamous Carcinoma. Tumor Biol, 2015, 36(4):2501–7., DOI 10.1007/s13277-014-2863-3 Huang L, Wang Y, Chen J, Wang Y, Zhao Y, Wang Y, et al. Long Noncoding RNA PCAT1, a Novel Serum-Based Biomarker, Enhances Cell Growth by Sponging miR-326 in Oesophageal Squamous Cell Carcinoma. Cel Death Dis, 2019, 10(7):513., DOI 10.1038/s41419-019-1745-4 Zhang YG, Zhou MW, Bai L, Han RY, Lv K, Wang Z. Extracellular Vesicles Promote Esophageal Cancer Progression by Delivering lncZEB1-AS1 between Cells. Eur Rev Med Pharmacol Sci, 2018, 22(9):2662–70., DOI 10.26355/ eurrev_201805_14962 Li Z, Zhou Y, Tu B, Bu Y, Liu A, Kong J. Long Noncoding RNA MALAT1 Affects the Efficacy of Radiotherapy for Esophageal Squamous Cell Carcinoma by Regulating Cks1 Expression. J Oral Pathol Med, 2017, 46(8):583–90., DOI 10. 1111/jop.12538 Farooqi A, Legaki E, Gazouli M, Rinaldi S, Berardi R. MALAT1 as a Versatile Regulator of Cancer: Overview of the Updates from Predatory Role as Competitive Endogenous RNA to Mechanistic Insights. Curr Cancer Drug Targets, 2020, 21:192–202., DOI 10.2174/1568009620999200730183110 Luo W, Liu W, Yao J, Zhu W, Zhang H, Sheng Q, et al. Downregulation of H19 Decreases the Radioresistance in Esophageal Squamous Cell Carcinoma Cells. OncoTargets Ther, 2019, 12:4779–88., DOI 10.2147/ott.s203235 Lin J, Liu Z, Liao S, Li E, Wu X, Zeng W. Elevation of Long Non-coding RNA GAS5 and Knockdown of microRNA-21 Up-Regulate RECK Expression to Enhance Esophageal Squamous Cell Carcinoma Cell Radio-Sensitivity after Radiotherapy. Genomics, 2020, 112(3):2173–85., DOI 10.1016/j.ygeno.2019. 12.013 Nissan A, Stojadinovic A, Mitrani-Rosenbaum S, Halle D, Grinbaum R, Roistacher M, et al. Colon Cancer Associated Transcript-1: a Novel RNA Expressed in Malignant and Pre-malignant Human Tissues. Int J Cancer, 2012, 130(7):1598–606., DOI 10.1002/ijc.26170 Hu M, Zhang Q, Tian XH, Wang JL, Niu YX, Li G. lncRNA CCAT1 Is a Biomarker for the Proliferation and Drug Resistance of Esophageal Cancer via the miR-143/PLK1/BUBR1 axis. Mol Carcinog, 2019, 58(12):2207–17., DOI 10. 1002/mc.23109 Kang M, Ren M, Li Y, Fu Y, Deng M, Li C. Exosome-mediated Transfer of lncRNA PART1 Induces Gefitinib Resistance in Esophageal Squamous Cell Carcinoma via Functioning as a Competing Endogenous RNA. J Exp Clin Cancer Res, 2018, 37(1):171., DOI 10.1186/s13046-018-0845-9 Tong Y, Yang L, Yu C, Zhu W, Zhou X, Xiong Y, et al. Tumor-Secreted Exosomal lncRNA POU3F3 Promotes Cisplatin Resistance in ESCC by Inducing Fibroblast Differentiation into CAFs. Mol Ther - Oncolytics, 2020, 18:1–13., DOI 10.1016/j.omto.2020.05.014 Moran VA, Perera RJ, Khalil AM. Emerging Functional and Mechanistic Paradigms of Mammalian Long Non-coding RNAs. Nucleic Acids Res, 2012, 40(14):6391–400., DOI 10.1093/nar/gks296 Chen G, Wang Z, Wang D, Qiu C, Liu M, Chen X, et al. LncRNADisease: a Database for Long-Non-Coding RNA-Associated Diseases. Nucleic Acids Res, 2013, 41(Database issue):D983–6., DOI 10.1093/nar/gks1099 Ning S, Zhang J, Wang P, Zhi H, Wang J, Liu Y, et al. Lnc2Cancer: a Manually Curated Database of Experimentally Supported lncRNAs Associated with Various Human Cancers. Nucleic Acids Res, 2016, 44(D1):D980–D985., DOI 10.1093/nar/gkv1094 Bao Z, Yang Z, Huang Z, Zhou Y, Cui Q, Dong D. LncRNADisease 2.0: an Updated Database of Long Non-coding RNA-Associated Diseases. Nucleic Acids Res, 2019, 47(D1):D1034–d1037., DOI 10.1093/nar/gky905 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610140 EP 1610152 DI 10.3389/pore.2022.1610140 PG 13 ER PT J AU Song, J Yin, H Zhu, Y Fei, Sh AF Song, Jiawei Yin, Huanhuan Zhu, Yong Fei, Shengqi TI Identification of Predictive Factors for Lymph Node Metastasis in pT1 Stage Colorectal Cancer Patients: A Retrospective Analysis Based on the Population Database SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal cancer; lymph node metastasis; SEER; predictive factors; pT1 stage ID colorectal cancer; lymph node metastasis; SEER; predictive factors; pT1 stage AB Objective: The purpose of this study was to identify predictive factors for lymph node metastasis (LNM) in pT1 stage colorectal cancer (CRC) patients. Methods: From the Surveillance, Epidemiology, and End Results (SEER) database, 2,697 consecutive pT1 stage patients who underwent surgical resection were retrospectively reviewed. Predictive factors for LNM were identified by the univariate and multivariate logistic regression analysis. The Kaplan-Meier curves and multivariate Cox regression analysis were used to evaluate the relationships between LNM and overall survival (OS) as well as cancer specific survival (CSS) of pT1 stage CRC patients. Results: The prevalence of LNM in pT1 stage CRC patients was 15.2% (410/2,697). Patient age <60 years (OR:1.869, 95% CI: 1.505–2.321, p < 0.001), poorly differentiated or mucinous or signet ring cell adenocarcinoma (OR:2.075, 95% CI: 1.584–2.717, p < 0.001), elevated carcinoembryonic antigen (CEA) level (OR:1.343, 95% CI: 1.022–1.763, p = 0.033) and perineural invasion (PNI) (OR:6.212, 95% CI: 3.502–11.017, p < 0.001) were significantly associated with LNM in pT1 stage patients. The survival analysis demonstrated that pT1 stage patients with LNM had a worse OS (5-year OS: 82.2% vs 88.7%, p = 0.020) and CSS (5-year CSS: 74.9% vs 81.5%, p = 0.041) than those without lymph node metastasis. Lymph node metastasis was an independent predictor of poor OS (HR: 1.543, 95% CI: 1.156–2.060, p = 0.003) and CSS (HR: 1.614, 95% CI: 1.121–2.324, p = 0.010) for pT1 stage colorectal cancer patients. Conclusion: Age, differentiation type, CEA level and perineural invasion were independent predictive factors for LNM in pT1 stage CRC patients. These findings might provide further risk stratification for pT1 stage patients and help clinicians identify high-risk individuals. C1 [Song, Jiawei] Changxing People’s Hospital, Department of Gastrointestinal SurgeryChangxing, China. [Yin, Huanhuan] Changxing People’s Hospital, Department of GastroenterologyChangxing, China. [Zhu, Yong] Changxing People’s Hospital, Department of Gastrointestinal SurgeryChangxing, China. [Fei, Shengqi] Changxing People’s Hospital, Department of Gastrointestinal SurgeryChangxing, China. RP Zhu, Y (reprint author), Changxing People’s Hospital, Department of Gastrointestinal Surgery, Changxing, China. EM zy785184535@126.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer J Clinicians, 2018, 68:394–424., DOI 10.3322/caac.21492 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer Statistics in China, 2015. CA: A Cancer J Clinicians, 2016, 66:115–32., DOI 10.3322/caac. 21338 De Ceglie A, Hassan C, Mangiavillano B, Matsuda T, Saito Y, Ridola L, et al. Endoscopic Mucosal Resection and Endoscopic Submucosal Dissection for Colorectal Lesions: A Systematic Review. Crit Rev Oncology/Hematology, 2016, 104:138–55., DOI 10.1016/j.critrevonc.2016.06.008 Saitoh Y, Inaba Y, Sasaki T, Sugiyama R, Sukegawa R, Fujiya M Management of Colorectal T1 Carcinoma Treated by Endoscopic Resection. Dig Endosc, 2016, 28:324–9., DOI 10.1111/den.12503 Mou S, Soetikno R, Shimoda T, Rouse R, Kaltenbach T Pathologic Predictive Factors for Lymph Node Metastasis in Submucosal Invasive, T1, Colorectal Cancer: a Systematic Review and Meta-Analysis. Surg Endosc, 2013, 27: 2692–703., DOI 10.1007/s00464-013-2835-5 Yasue C, Chino A, Takamatsu M, Namikawa K, Ide D, Saito S, et al. Pathological Risk Factors and Predictive Endoscopic Factors for Lymph Node Metastasis of T1 Colorectal Cancer: a Single-center Study of 846 Lesions. J Gastroenterol, 2019, 54:708–17., DOI 10.1007/s00535-019- 01564-y Miyachi H, Kudo Se., Ichimasa K, Hisayuki T, Oikawa H, Matsudaira S, et al. Management of T1 Colorectal Cancers after Endoscopic Treatment Based on the Risk Stratification of Lymph Node Metastasis. J Gastroenterol Hepatol, 2016, 31:1126–32., DOI 10.1111/jgh.13257 Oh JR, Park B, Lee S, Han KS, Youk E-G, Lee D-H, et al. Nomogram Development and External Validation for Predicting the Risk of Lymph Node Metastasis in T1 Colorectal Cancer. Cancer Res Treat, 2019, 51: 1275–84., DOI 10.4143/crt.2018.569 Hashiguchi Y, Muro K, Muro K, Saito Y, Ito Y, Ajioka Y, et al. Japanese Society for Cancer of the Colon and Rectum, JSCCR, Guidelines 2019 for the Treatment of Colorectal Cancer. Int J Clin Oncol, 2020, 25:1–42., DOI 10. 1007/s10147-019-01485-z Pimentel-Nunes P, Dinis-Ribeiro M, Ponchon T, Repici A, Vieth M, De Ceglie A, et al. Endoscopic Submucosal Dissection: European Society of Gastrointestinal Endoscopy, ESGE, Guideline. Endoscopy, 2015, 47:829–54., DOI 10.1055/s-0034-1392882 Oka S, Tanaka S, Nakadoi K, Kanao H, Chayama K Risk Analysis of Submucosal Invasive Rectal Carcinomas for Lymph Node Metastasis to Expand Indication Criteria for Endoscopic Resection. Dig Endosc, 2013, 25(Suppl. 2):21–5., DOI 10.1111/den.12089 Ha RK, Han KS, Sohn DK, Kim BC, Hong CW, Chang HJ, et al. Histopathologic Risk Factors for Lymph Node Metastasis in Patients with T1 Colorectal Cancer. Ann Surg Treat Res, 2017, 93:266–71., DOI 10.4174/astr. 2017.93.5.266 Kim JH, Cheon JH, Kim TI, Baik SH, Kim NK, Kim H, et al. Effectiveness of Radical Surgery after Incomplete Endoscopic Mucosal Resection for Early Colorectal Cancers: a Clinical Study Investigating Risk Factors of Residual Cancer. Dig Dis Sci, 2008, 53:2941–6., DOI 10.1007/s10620-008- 0248-4 Sun Z-Q, Ma S, Zhou Q-B, Yang S-X, Chang Y, Zeng X-Y, et al. Prognostic Value of Lymph Node Metastasis in Patients with T1-Stage Colorectal Cancer from Multiple Centers in China. Wjg, 2017, 23:8582–90., DOI 10.3748/wjg.v23. i48.8582 Tateishi Y, Nakanishi Y, Taniguchi H, Shimoda T, Umemura S Pathological Prognostic Factors Predicting Lymph Node Metastasis in Submucosal Invasive, T1, Colorectal Carcinoma. Mod Pathol, 2010, 23:1068–72., DOI 10.1038/ modpathol.2010.88 Suh J, Han K, Kim B, Hong C, Sohn D, Chang H, et al. Predictors for Lymph Node Metastasis in T1 Colorectal Cancer. Endoscopy, 2012, 44:590–5., DOI 10. 1055/s-0031-1291665 Wang H-S, Liang W-Y, Lin T-C, Chen W-S, Jiang J-K, Yang S-H, et al. Curative Resection of T1 Colorectal Carcinoma: Risk of Lymph Node Metastasis and Long-Term Prognosis. Dis Colon Rectum, 2005, 48: 1182–92., DOI 10.1007/s10350-004-0935-y Suzuki T, Sadahiro S, Mukoyama S, Ishikawa K, Yasuda S, Tajima T, et al. Risk of Lymph Node and Distant Metastases in Patients with Early Invasive Colorectal Cancer Classified as Haggitt’s Level 4 Invasion. Dis Colon Rectum, 2003, 46:203–8., DOI 10.1007/s10350-004-6525-1 Wada H, Shiozawa M, Katayama K, Okamoto N, Miyagi Y, Rino Y, et al. Systematic Review and Meta-Analysis of Histopathological Predictive Factors for Lymph Node Metastasis in T1 Colorectal Cancer. J Gastroenterol, 2015, 50: 727–34., DOI 10.1007/s00535-015-1057-0 Knijn N, Mogk SC, Teerenstra S, Simmer F, Nagtegaal ID Perineural Invasion Is a Strong Prognostic Factor in Colorectal Cancer. Am J Surg Pathol, 2016, 40: 103–12., DOI 10.1097/PAS.0000000000000518 Alotaibi AM, Lee JL, Kim J, Lim S-B, Yu CS, Kim TW, et al. Prognostic and Oncologic Significance of Perineural Invasion in Sporadic Colorectal Cancer. Ann Surg Oncol, 2017, 24:1626–34., DOI 10.1245/s10434-016-5748-4 Huh JW, Kim HR, Kim YJ Lymphovascular or Perineural Invasion May Predict Lymph Node Metastasis in Patients with T1 and T2 Colorectal Cancer. J Gastrointest Surg, 2010, 14:1074–80., DOI 10.1007/s11605-010-1206-y Liebig C, Ayala G, Wilks JA, Berger DH, Albo D Perineural Invasion in Cancer. Cancer, 2009, 115:3379–91., DOI 10.1002/cncr.24396 Kim CW, Yoon YS, Park IJ, Lim S-B, Yu CS, Kim JC Elevation of Preoperative S-CEA Concentration in Stage IIA Colorectal Cancer Can Also Be a High Risk Factor for Stage II Patients. Ann Surg Oncol, 2013, 20:2914–20., DOI 10.1245/ s10434-013-2919-4 Baqar AR, Wilkins S, Staples M, Angus Lee CH, Oliva K, McMurrick P The Role of Preoperative CEA in the Management of Colorectal Cancer: A Cohort Study from Two Cancer Centres. Int J Surg, 2019, 64:10–5., DOI 10.1016/j.ijsu. 2019.02.014 Wang H, Wei XZ, Fu CG, Zhao RH, Cao FA Patterns of Lymph Node Metastasis Are Different in colon and Rectal Carcinomas. Wjg, 2010, 16: 5375–9., DOI 10.3748/wjg.v16.i42.5375 Aytac E, Gorgun E, Costedio MM, Stocchi L, Remzi FH, Kessler H Impact of Tumor Location on Lymph Node Metastasis in T1 Colorectal Cancer. Langenbecks Arch Surg, 2016, 401:627–32., DOI 10.1007/s00423-016-1452-x Ichimasa K, Kudo S-E,Miyachi H, Kouyama Y, Hayashi T,Wakamura K, et al. Comparative Clinicopathological Characteristics of colon and Rectal T1 Carcinoma. Oncol Lett, 2017, 13:805–10., DOI 10.3892/ol.2016.5464 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610191 EP 1610198 DI 10.3389/pore.2022.1610191 PG 8 ER PT J AU Wang, A Yuan, Y Chu, H Gao, Y Jin, Z Jia, Q Zhu, B AF Wang, Aoyun Yuan, Yixiao Chu, Han Gao, Yixing Jin, Zheng Jia, Qingzhu Zhu, Bo TI Somatostatin Receptor 2: A Potential Predictive Biomarker for Immune Checkpoint Inhibitor Treatment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bioinformatics; tumor microenvironment; immune checkpoint inhibitors; somatostatin receptor 2; predictive biomarker ID bioinformatics; tumor microenvironment; immune checkpoint inhibitors; somatostatin receptor 2; predictive biomarker AB Somatostatin receptor 2 (SSTR2), the most abundant receptor of somatostatin (SST), possesses immunoreactivity and is altered in many cancers. However, the association between SSTR2 and efficacy of immune checkpoint inhibitors (ICIs) has not yet been reported. Immunohistochemistry (IHC) information across 20 cancers was collected from the Human Protein Atlas (HPA) and used to analyze the expression of SSTR2. Immune signatures collected from public databases, such as BioCarta or Reactome, were used to investigate the association between SSTR2 and the tumor microenviroment in the Cancer Genome Atlas (TCGA). Data from cohorts treated with ICIs were collected to assess whether SSTR2 is associated with benefits from ICIs treatment. In the HPA, we found the SSTR2 IHC-positive rate of 13 cancers to be above 50%. Five types of cancer express SSTR2 mildly (positive rate: 25%–50%), while the remaining two types of cancer barely stained SSTR2-positive (positive rate: 0%–24%). In TCGA analysis, immune cell signatures and immune function pathways were enriched in high SSTR2 expression groups in most cancers. In each ICIs treated cohort, patients with high SSTR2 expression experienced numerically superior objective response rate (Braun: 14.8% vs 13.4%, p = 0.85; Gide: 69.4% vs 40.5%, p = 0.025; Mariathasan: 22.4% vs 16.7%, p = 0.233; Miao: 37.5% vs 11.8%; Riaz: 32.0% vs 7.7%, p = 0.067) and overall survival (Braun: HR (95%CI): 0.80 [0.62–1.04], p = 0.80; Gide: HR (95%CI): 0.61 [0.29–1.30], p = 0.20; Mariathasan: HR (95%CI): 0.83 [0.64–1.08], p = 0.16; Miao: HR (95%CI): 0.24 [0.086–0.65], p = 0.0028; Nathanson cohort: HR (95%CI): 0 [0-inf], p = 0.18; Riaz: HR (95%CI): 0.24 [0.086–0.65], p = 0.028) than patients with low SSTR2 expression. In pooled cohort, we found these differences were significant (Pool: 24.6% vs 16.7%, p = 0.0077; HR (95% CI): 0.77 [0.65–0.91], p = 0.0018). Our results suggest that SSTR2 is a potential predictive biomarker for response to ICIs. C1 [Wang, Aoyun] Third Military Medical University, Xinqiao Hospital, Institute of CancerChongqing, China. [Yuan, Yixiao] The Third Affiliated Hospital of Kunming Medical University, Department of Thoracic SurgeryKunming, China. [Chu, Han] Sichuan University, College of Life Sciences, Key Laboratory of Bio-Resources and Eco-Environment, Center of Growth, Metabolism and AgingChengdu, China. [Gao, Yixing] Third Military Medical University, Xinqiao Hospital, Institute of CancerChongqing, China. [Jin, Zheng] GloriousMed Clinical Laboratory (Shanghai) Co., Ltd., Research InstituteShanghai, China. [Jia, Qingzhu] Third Military Medical University, Xinqiao Hospital, Institute of CancerChongqing, China. [Zhu, Bo] Third Military Medical University, Xinqiao Hospital, Institute of CancerChongqing, China. RP Jia, Q (reprint author), Third Military Medical University, Xinqiao Hospital, Institute of Cancer, Chongqing, China. EM jiaqingzhu0801@outlook.com CR Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab Plus Chemotherapy in Metastatic Non-small-cell Lung Cancer. N Engl J Med, 2018, 378:2078–92., DOI 10.1056/ NEJMoa1801005 Garon EB, Hellmann MD, Rizvi NA, Carcereny E, Leighl NB, Ahn M-J, et al. Five-Year Overall Survival for Patients with Advanced Non‒Small-Cell Lung Cancer Treated with Pembrolizumab: Results from the Phase I KEYNOTE- 001 Study. Jco, 2019, 37:2518–27., DOI 10.1200/JCO.19.00934 Larkin J, Chiarion-Sileni V, Gonzalez R, Grob J-J, Rutkowski P, Lao CD, et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med, 2019, 381:1535–46., DOI 10.1056/NEJMoa1910836 Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, et al. Atezolizumab in Patients with Locally Advanced and Metastatic Urothelial Carcinoma Who Have Progressed Following Treatment with Platinum-Based Chemotherapy: a Single-Arm, Multicentre, Phase 2 Trial. The Lancet, 2016, 387:1909–20., DOI 10.1016/S0140-6736(16, 00561-4 Mok TSK, Wu YL, Kudaba I, Kowalski DM, Cho BC, Turna HZ, et al. Pembrolizumab versus Chemotherapy for Previously Untreated, PD-L1- Expressing, Locally Advanced or Metastatic Non-small-cell Lung Cancer, KEYNOTE-042): a Randomised, Open-Label, Controlled, Phase 3 Trial. Lancet, 2019, 393:1819–30., DOI 10.1016/S0140-6736(18)32409-7 Ouwerkerk W, van den Berg M, van der Niet S, Limpens J, Luiten RM. Biomarkers, Measured during Therapy, for Response of Melanoma Patients to Immune Checkpoint Inhibitors: a Systematic Review. Melanoma Res, 2019, 29:453–64., DOI 10.1097/CMR.0000000000000589 Havel JJ, Chowell D, Chan TA. The Evolving Landscape of Biomarkers for Checkpoint Inhibitor Immunotherapy. Nat Rev Cancer, 2019, 19:133–50., DOI 10.1038/s41568-019-0116-x ZouW,Wolchok JD, Chen L. PD-L1, B7-H1, and PD-1 Pathway Blockade for Cancer Therapy: Mechanisms, Response Biomarkers, and Combinations. Sci Transl Med, 2016, 8:328rv324., DOI 10.1126/scitranslmed.aad7118 Marcus L, Lemery SJ, Keegan P, Pazdur R. FDA Approval Summary: Pembrolizumab for the Treatment of Microsatellite Instability-High Solid Tumors. Clin Cancer Res, 2019, 25:3753–8., DOI 10.1158/1078-0432.CCR-18- 4070 Prat A, Navarro A, Pare L, Reguart N, Galvan P, Pascual T, et al. Immune- Related Gene Expression Profiling after PD-1 Blockade in Non-small Cell Lung Carcinoma, Head and Neck Squamous Cell Carcinoma, and Melanoma. Cancer Res, 2017, 77:3540–50., DOI 10.1158/0008-5472.CAN-16-3556 Riaz N, Havel JJ, Kendall SM, Makarov V, Walsh LA, Desrichard A, et al. Recurrent SERPINB3 and SERPINB4 Mutations in Patients Who Respond to Anti-CTLA4 Immunotherapy. Nat Genet, 2016, 48:1327–9., DOI 10.1038/ng. 3677 Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Mutational Landscape Determines Sensitivity to PD-1 Blockade in Non-small Cell Lung Cancer. Science, 2015, 348:124–8., DOI 10.1126/science.aaa1348 Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, et al. Association of PD- 1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti-PD-1 Therapy. Clin Cancer Res, 2014, 20:5064–74., DOI 10.1158/1078-0432.CCR-13-3271 Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJM, Robert L, et al. PD-1 Blockade Induces Responses by Inhibiting Adaptive Immune Resistance. Nature, 2014, 515:568–71., DOI 10.1038/nature13954 Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee J-L, Fong L, et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med, 2017, 376:1015–26., DOI 10.1056/ NEJMoa1613683 Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WEE, Poddubskaya E, et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-small-cell Lung Cancer. N Engl J Med, 2015, 373:123–35., DOI 10.1056/NEJMoa1504627 Mehnert JM, Monjazeb AM, Beerthuijzen JMT, Collyar D, Rubinstein L, Harris LN. The Challenge for Development of Valuable Immuno-Oncology Biomarkers. Clin Cancer Res, 2017, 23:4970–9., DOI 10.1158/1078-0432.CCR- 16-3063 Cristescu R, Mogg R, Ayers M, Albright A, Murphy E, Yearley J, et al. Pantumor Genomic Biomarkers for PD-1 Checkpoint Blockade-Based Immunotherapy. Science, 2018, 362:eaar3593., DOI 10.1126/science.aar3593 Wu W, Zhou Y, Wang Y, Liu L, Lou J, Deng Y, et al. Clinical Significance of Somatostatin Receptor, SSTR, 2 in Meningioma. Front Oncol, 2020, 10:1633., DOI 10.3389/fonc.2020.01633 Dutour A, Kumar U, Panetta R, Ouafik LH, Fina F, Sasi R, et al. Expression of Somatostatin Receptor Subtypes in Human Brain Tumors. Int J Cancer, 1998, 76:620–7., DOI 10.1002/(sici)1097-0215(19980529)76:5<620:aid-ijc2>3.0.co;2-s Schulz S, Pauli SU, Schulz S, Handel M, Dietzmann K, Firsching R, et al. Immunohistochemical Determination of Five Somatostatin Receptors in Meningioma Reveals Frequent Overexpression of Somatostatin Receptor Subtype sst2A. Clin Cancer Res, 2000, 6:1865–74. Li J, Chen C, Bi X, Zhou C, Huang T, Ni C, et al. DNA Methylation of CMTM3 , SSTR2 , and MDFI Genes in Colorectal Cancer. Gene, 2017, 630:1–7., DOI 10. 1016/j.gene.2017.07.082 Tang LH, Modlin IM. Somatostatin Receptor Regulation of Gastric Carcinoid Tumours. Digestion, 1996, 57(Suppl. 1):11–4., DOI 10.1159/000201385 Yin X, Wang P, Yang T, Li G, Teng X, Huang W, et al. Identification of Key Modules and Genes Associated with Breast Cancer Prognosis Using WGCNA and ceRNA Network Analysis. Aging, 2020, 13:2519–38., DOI 10.18632/aging. 202285 Patel YC. Somatostatin and its Receptor Family. Front Neuroendocrinology, 1999, 20:157–98., DOI 10.1006/frne.1999.0183 Hagen PMV, Krenning EP, Kwekkeboom DJ, Reubi JC, Anker-Lugtenburg v. PJ, Lowenberg B, et al. Somatostatin and the Immune and Haematopoetic System; a Review. Eur J Clin Invest, 1994, 24:91–9., DOI 10.1111/j.1365-2362. 1994.tb00972.x Braun DA, Hou Y, Bakouny Z, Ficial M, Sant’ Angelo M, Forman J, et al. Interplay of Somatic Alterations and Immune Infiltration Modulates Response to PD-1 Blockade in Advanced clear Cell Renal Cell Carcinoma. Nat Med, 2020, 26:909–18., DOI 10.1038/s41591-020-0839-y Gide TN, Quek C, Menzies AM, Tasker AT, Shang P, Holst J, et al. Distinct Immune Cell Populations Define Response to Anti-PD-1 Monotherapy and Anti-PD-1/Anti-CTLA-4 Combined Therapy. Cancer Cell, 2019, 35:238–55., DOI 10.1016/j.ccell.2019.01.003 Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFβ Attenuates Tumour Response to PD-L1 Blockade by Contributing to Exclusion of T Cells. Nature, 2018, 554:544–8., DOI 10.1038/ nature25501 Miao D, Margolis CA, Gao W, Voss MH, Li W, Martini DJ, et al. Genomic Correlates of Response to Immune Checkpoint Therapies in clear Cell Renal Cell Carcinoma. Science, 2018, 359:801–6., DOI 10.1126/science.aan5951 Nathanson T, Ahuja A, Rubinsteyn A, Aksoy BA, Hellmann MD, Miao D, et al. Somatic Mutations and Neoepitope Homology in Melanomas Treated with CTLA-4 Blockade. Cancer Immunol Res, 2017, 5:84–91., DOI 10.1158/2326- 6066.CIR-16-0019 Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, et al. Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab. Cell, 2017, 171:934–49., DOI 10.1016/j.cell.2017.09.028 Angelova M, Charoentong P, Hackl H, Fischer ML, Snajder R, Krogsdam AM, et al. Characterization of the Immunophenotypes and Antigenomes of Colorectal Cancers Reveals Distinct Tumor Escape Mechanisms and Novel Targets for Immunotherapy. Genome Biol, 2015, 16:64., DOI 10.1186/s13059- 015-0620-6 Charoentong P, Finotello F, Angelova M, Mayer C, Efremova M, Rieder D, et al. Pan-cancer Immunogenomic Analyses Reveal Genotype- Immunophenotype Relationships and Predictors of Response to Checkpoint Blockade. Cell Rep, 2017, 18:248–62., DOI 10.1016/j.celrep.2016. 12.019 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: a Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci, 2005, 102:15545–50., DOI 10.1073/pnas.0506580102 Theodoropoulou M, Stalla GK. Somatostatin Receptors: from Signaling to Clinical Practice. Front Neuroendocrinology, 2013, 34:228–52., DOI 10.1016/j. yfrne.2013.07.005 Benali N, Cordelier P, Calise D, Pages P, Rochaix P, Nagy A, et al. Inhibition of Growth and Metastatic Progression of Pancreatic Carcinoma in Hamster after Somatostatin Receptor Subtype 2, Sst2, Gene Expression and Administration of Cytotoxic Somatostatin Analog AN-238. Proc Natl Acad Sci, 2000, 97: 9180–5., DOI 10.1073/pnas.130196697 Li M, Fisher WE, Kim HJ,Wang X, Brunicardi CF, Chen C, et al. Somatostatin, Somatostatin Receptors, and Pancreatic Cancer. World J Surg, 2005, 29:293–6., DOI 10.1007/s00268-004-7814-5 Buscail L, Saint-Laurent N, Chastre E, Vaillant JC, Gespach C, Capella G, et al. Loss of Sst2 Somatostatin Receptor Gene Expression in Human Pancreatic and Colorectal Cancer. Cancer Res, 1996, 56:1823–7. Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, et al. First-Line Nivolumab in Stage IV or Recurrent Non-small-cell Lung Cancer. N Engl J Med, 2017, 376:2415–26., DOI 10.1056/NEJMoa1613493 Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, Activity, and Immune Correlates of Anti-PD-1 Antibody in Cancer. N Engl J Med, 2012, 366:2443–54., DOI 10.1056/NEJMoa1200690 McGrail DJ, Pilie PG, Rashid NU, Voorwerk L, Slagter M, Kok M, et al. High Tumor Mutation burden Fails to Predict Immune Checkpoint Blockade Response across All Cancer Types. Ann Oncol, 2021, 32:661–72., DOI 10. 1016/j.annonc.2021.02.006 Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, et al. IFN-γ-related mRNA Profile Predicts Clinical Response to PD-1 Blockade. J Clin Invest, 2017, 127:2930–40., DOI 10.1172/JCI91190 Ott PA, Bang Y-J, Piha-Paul SA, Razak ARA, Bennouna J, Soria J-C, et al. T-Cell-Inflamed Gene-Expression Profile, Programmed Death Ligand 1 Expression, and Tumor Mutational Burden Predict Efficacy in Patients Treated with Pembrolizumab across 20 Cancers: KEYNOTE-028. Jco, 2019, 37:318–27., DOI 10.1200/JCO.2018.78.2276 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610196 EP 1610205 DI 10.3389/pore.2022.1610196 PG 10 ER PT J AU Uchida, Sh Kojima, T Sugino, T AF Uchida, Shiro Kojima, Takaaki Sugino, Takashi TI Frequency and Clinicopathological Characteristics of Patients With KRAS/BRAF Double-Mutant Colorectal Cancer: An In Silico Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer; KRAS; BRAF; bioinformatic analysis; colon; double mutation ID cancer; KRAS; BRAF; bioinformatic analysis; colon; double mutation AB KRAS and BRAF mutations are currently thought to be mutually exclusive as their cooccurrence is extremely rare. Therefore, clinicopathological and molecular characteristics of colorectal carcinoma with KRAS/BRAF double mutations are unclear. We aimed to investigate the frequency and clinicopathological characteristics of double-mutant colorectal carcinoma and its differences from KRAS/BRAF single-mutant colorectal carcinoma using bioinformatics tools. We estimated the KRAS/BRAF double mutation frequency in the whole exon and coding sequences via bioinformatic analyses of three datasets from cBioPortal. We compared the clinicopathological characteristics, microsatellite instability status, BRAF classification, and tumor mutation burden of patients harboring the double mutants with those of patients harboring KRAS or BRAF single mutations. We integrated three large datasets and found that the frequency of the KRAS/BRAF double mutation in the dataset was 1.2% (29/2347). The double mutation occurred more frequently in males, with a slightly higher occurrence in the right side of the colon. Sex, histological type, histological grade, microsatellite instability, and tumor mutation burden of the patients harboring KRAS-mutant, BRAF-mutant, and doublemutant colorectal carcinoma varied significantly. The frequency of double-mutant colorectal carcinoma was 60 times higher than that previously reported. Significantly fewer double-mutant colorectal carcinoma cases were classified as BRAF class 1 and more were classified as unknown. Our findings indicate that the biological characteristics of double-mutant tumors are different from those of single-mutant tumors. C1 [Uchida, Shiro] Kikuna Memorial Hospital, Division of Diagnostic PathologyYokohama, Japan. [Kojima, Takaaki] Nagoya University, Graduate School of Bioagricultural SciencesNagoya, Japan. [Sugino, Takashi] Shizuoka Cancer Center, Division of PathologyShizuoka, Japan. RP Uchida, Sh (reprint author), Kikuna Memorial Hospital, Division of Diagnostic Pathology, Yokohama, Japan. EM Dr.Uchida@gmail.com CR WHO Classification of Tumours Editorial Board. WHO Classification of Tumours In: Digestive System Tumours. 5th ed. Lyon: IARC, 2019). Cancer Genome Atlas Network. Comprehensive Molecular Characterization of Human colon and Rectal Cancer. Nature, 2012, 487:330–7., DOI 10.1038/ nature11252 Morkel M, Riemer P, Blaker H, Sers C. Similar but Different: Distinct Roles for KRAS and BRAF Oncogenes in Colorectal Cancer Development and Therapy Resistance. Oncotarget, 2015, 6:20785–800., DOI 10.18632/oncotarget.4750 Lin JS, Webber EM, Senger CA, Holmes RS, Whitlock EP. Systematic Review of Pharmacogenetic Testing for Predicting Clinical Benefit to Anti-EGFR Therapy in Metastatic Colorectal Cancer. Am J Cancer Res, 2011, 1:650–62. Castellano E, Downward J. RAS Interaction with PI3K: More Than Just Another Effector Pathway. Genes & Cancer, 2011, 2:261–74., DOI 10.1177/ 1947601911408079 Eklof V, Wikberg M.L, Edin S, Dahlin A.M, Jonsson B-A, Oberg A, et al. The Prognostic Role of KRAS, BRAF, PIK3CA and PTEN in Colorectal Cancer. Br J Cancer, 2013, 108:2153–63., DOI 10.1038/bjc.2013.212 Afrasanie V.-A, Marinca MV, Alexa-Stratulat T, Gafton B, Paduraru M, Adavidoaiei AM, et al. KRAS, NRAS, BRAF, HER2 and Microsatellite Instability in Metastatic Colorectal Cancer - Practical Implications for the Clinician. Radiol Oncol, 2019, 53:265–74., DOI 10.2478/raon-2019-0033 Dankner M, Rose AAN, Rajkumar S, Siegel PM, Watson IR Classifying BRAF Alterations in Cancer: New Rational Therapeutic Strategies for Actionable Mutations. Oncogene, 2018, 37:3183–99., DOI 10.1038/s41388-018-0171-x Yao Z, Yaeger R, Rodrik-Outmezguine VS, Tao A, Torres NM, Chang MT, et al. Tumours with Class 3 BRAF Mutants Are Sensitive to the Inhibition of Activated RAS. Nature, 2017, 548:234–8., DOI 10.1038/nature23291 Yoshino T, Arnold D, Taniguchi H, Pentheroudakis G, Yamazaki K, Xu R-H, et al. Pan-Asian Adapted ESMO Consensus Guidelines for the Management of Patients with Metastatic Colorectal Cancer: A JSMO-ESMO Initiative Endorsed by CSCO, KACO, MOS, SSO and TOS. Ann Oncol, 2018, 29: 44–70., DOI 10.1093/annonc/mdx738 Rajagopalan H, Bardelli A, Lengauer C, Kinzler KW, Vogelstein B, Velculescu VE RAF/RAS Oncogenes and Mismatch-Repair Status. Nature, 2002, 418:934., DOI 10.1038/418934a Nakayama I, Hirota T, Shinozaki E BRAF Mutation in Colorectal Cancers: From Prognostic Marker to Targetable Mutation. Cancers, 2020, 12:3236., DOI 10.3390/cancers12113236 Van Cutsem E, Kohne C-H, Lang I, Folprecht G, Nowacki MP, Cascinu S, et al. Cetuximab Plus Irinotecan, Fluorouracil, and Leucovorin as First-Line Treatment for Metastatic Colorectal Cancer: Updated Analysis of Overall Survival According to Tumor KRAS and BRAF Mutation Status. Jco, 2011, 29: 2011–9., DOI 10.1200/JCO.2010.33.5091 Lamy A, Blanchard F, Le Pessot F, Sesboue R, Di Fiore F, Bossut J, et al. Metastatic Colorectal Cancer KRAS Genotyping in Routine Practice: Results and Pitfalls. Mod Pathol, 2011, 24:1090–100., DOI 10.1038/modpathol.2011.60 Price TJ, Hardingham JE, Lee CK, Weickhardt A, Townsend AR, Wrin JW, et al. Impact of KRAS and BRAF Gene Mutation Status on Outcomes from the Phase III AGITG MAX Trial of Capecitabine Alone or in Combination with Bevacizumab and Mitomycin in Advanced Colorectal Cancer. Jco, 2011, 29: 2675–82., DOI 10.1200/JCO.2010.34.5520 Maughan TS, Adams RA, Smith CG, Meade AM, Seymour MT, Wilson RH, et al. Addition of Cetuximab to Oxaliplatin-Based First-Line Combination Chemotherapy for Treatment of Advanced Colorectal Cancer: Results of the Randomised Phase 3 MRC COIN Trial. The Lancet, 2011, 377:2103–14., DOI 10.1016/S0140-6736(11)60613-2 Wang X, Lu YY, An YX,Wang X, Zhao QC KRAS, BRAF and PIK3CAMutations in Human Colorectal Cancer: Relationship with Metastatic Colorectal Cancer. Oncol Rep, 2011, 25:1691–7., DOI 10.3892/or.2011.1217 Bokemeyer C, Bondarenko I, Hartmann JT, de Braud F, Schuch G, Zubel A, et al. Efficacy According to Biomarker Status of Cetuximab Plus FOLFOX-4 as First-Line Treatment for Metastatic Colorectal Cancer: The OPUS Study. Ann Oncol, 2011, 22:1535–46., DOI 10.1093/annonc/mdq632 De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, et al. Effects of KRAS, BRAF, NRAS, and PIK3CA Mutations on the Efficacy of Cetuximab Plus Chemotherapy in Chemotherapy-Refractory Metastatic Colorectal Cancer: a Retrospective Consortium Analysis. Lancet Oncol, 2010, 11:753–62., DOI 10.1016/S1470-2045(10)70130-3 Sahin IH, Kazmi SMA, Yorio JT, Bhadkamkar NA, KeeGarrett BKCR, Garrett CR Rare Though Not Mutually Exclusive: A Report of Three Cases of Concomitant KRAS and BRAF Mutation and a Review of the Literature. J Cancer, 2013, 4:320–2., DOI 10.7150/jca.3619 Larki P, Gharib E, Yaghoob Taleghani M, Khorshidi F, Nazemalhosseini- Mojarad E, Asadzadeh Aghdaei H Coexistence of KRAS and BRAF Mutations in Colorectal Cancer: A Case Report Supporting the Concept of Tumoral Heterogeneity. Cell J, 2017, 19:113–7., DOI 10.22074/cellj.2017.5123 Vittal A, Middinti A, Kasi Loknath Kumar A Are All Mutations the Same? A Rare Case Report of Coexisting Mutually Exclusive KRAS and BRAF Mutations in a Patient with Metastatic colon Adenocarcinoma. Case Rep Oncological Med, 2017, 2017:1–3., DOI 10.1155/2017/2321052 Deshwar A, Margonis GA, Andreatos N, Barbon C, Wang J, Buettner S, et al. Double KRAS and BRAF Mutations in Surgically Treated Colorectal Cancer Liver Metastases: An International, Multi-Institutional Case Series. Ar, 2018, 38:2891–5., DOI 10.21873/anticanres.12535 Midthun L, Shaheen S, Deisch J, Senthil M, Tsai J, Hsueh C-T Concomitant KRAS and BRAF Mutations in Colorectal Cancer. J Gastrointest Oncol, 2019, 10:577–81., DOI 10.21037/jgo.2019.01.10 Cafiero C, Re A, D’Amato G, Surico PL, Surico G, Pirrelli M, et al. KRAS and BRAF Concomitant Mutations in a Patient with Metastatic colon Adenocarcinoma: An Interesting Case Report. Case Rep Oncol, 2020, 13: 595–600., DOI 10.1159/000507882 Yaeger R, Chatila WK, Lipsyc MD, Hechtman JF, Cercek A, Sanchez-Vega F, et al. Clinical Sequencing Defines the Genomic Landscape of Metastatic Colorectal Cancer. Cancer Cell, 2018, 33:125–36., DOI 10.1016/j.ccell.2017. 12.004 Giannakis M, Mu XJ, Shukla SA, Qian ZR, Cohen O, Nishihara R, et al. Genomic Correlates of Immune-Cell Infiltrates in Colorectal Carcinoma. Cel Rep, 2016, 15:857–65., DOI 10.1016/j.celrep.2016.03.075 Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, et al. COSMIC: The Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res, 2019, 47: D941–D947., DOI 10.1093/nar/gky1015 Cancer Genome Atlas Research Network. Integrated Genomic Analyses of Ovarian Carcinoma. Nature, 2011, 474:609–15., DOI 10.1038/nature10166 Liu Y, Sethi NS, Hinoue T, Schneider BG, Cherniack AD, Sanchez-Vega F, et al. Comparative Molecular Analysis of Gastrointestinal Adenocarcinomas. Cancer Cell, 2018, 33:735–21., DOI 10.1016/j.ccell.2018.03.010 Cheng DT, Mitchell TN, Zehir A, Shah RH, Benayed R, Syed A, et al. Memorial Sloan Kettering-IntegratedMutation Profiling of Actionable Cancer Targets, MSKIMPACT). J Mol Diagn, 2015, 17:251–64., DOI 10.1016/j.jmoldx.2014.12.006 Niu B, Ye K, Zhang Q, Lu C, Xie M, McLellan MD, et al. MSIsensor: Microsatellite Instability Detection Using Paired Tumor-normal Sequence Data. Bioinformatics, 2014, 30:1015–6., DOI 10.1093/bioinformatics/btt755 Latham A, Srinivasan P, Kemel Y, Shia J, Bandlamudi C, Mandelker D, et al. Microsatellite Instability Is Associated with the Presence of Lynch Syndrome Pan-Cancer. Jco, 2019, 37:286–95., DOI 10.1200/JCO.18.00283 Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 Human Cancer Genomes Reveals the Landscape of Tumor Mutational burden. Genome Med, 2017, 9:34., DOI 10.1186/s13073-017- 0424-2 Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of Tumor Mutation burden as an Immunotherapy Biomarker: Utility for the Oncology Clinic. Ann Oncol, 2019, 30:44–56., DOI 10.1093/ annonc/mdy495 Fabrizio DA, George Jr TJ, Jr, Dunne RF, Frampton G, Sun J, Gowen K, et al. Beyond Microsatellite Testing: Assessment of Tumor Mutational burden Identifies Subsets of Colorectal Cancer Who May Respond to Immune Checkpoint Inhibition. J Gastrointest Oncol, 2018, 9:610–7., DOI 10.21037/ jgo.2018.05.06 Gilson P, Franczak C, Dubouis L, Husson M, Rouyer M, Demange J, et al. Evaluation of KRAS, NRAS and BRAF Hotspot Mutations Detection for Patients with Metastatic Colorectal Cancer Using Direct DNA Pipetting in a Fully-Automated Platform and Next-Generation Sequencing for Laboratory Workflow Optimisation. PLOS ONE, 2019, 14:e0219204., DOI 10.1371/journal. pone.0219204 Wu H-X, Wang Z-X, Zhao Q, Chen D-L, He M-M, Yang L-P, et al. Tumor Mutational and Indel burden: A Systematic Pan-Cancer Evaluation as Prognostic Biomarkers. Ann Transl Med, 2019, 7:640., DOI 10.21037/atm. 2019.10.116 Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ, Tu D, Tebbutt NC, et al. K-rasMutations and Benefit from Cetuximab in Advanced Colorectal Cancer. N Engl J Med, 2008, 359:1757–65., DOI 10.1056/NEJMoa0804385 Van Cutsem E, Kohne C-H, Hitre E, Zaluski J, Chang Chien C-R, Makhson A, et al. Cetuximab and Chemotherapy as Initial Treatment for Metastatic Colorectal Cancer. N Engl J Med, 2009, 360:1408–17., DOI 10.1056/NEJMoa0805019 De Roock W, Jonker DJ, Di Nicolantonio F, Sartore-Bianchi A, Tu D, Siena S, et al. Association of KRAS p.G13D Mutation with Outcome in Patients with Chemotherapy-Refractory Metastatic Colorectal Cancer Treated with Cetuximab. JAMA, 2010, 304:1812–20., DOI 10.1001/jama.2010.1535 Shinozaki E, Yoshino T, Yamazaki K, Muro K, Yamaguchi K, Nishina T, et al. Clinical Significance of BRAF Non-v600e Mutations on the Therapeutic Effects of Anti-EGFR Monoclonal Antibody Treatment in Patients with Pretreated Metastatic Colorectal Cancer: The Biomarker Research for Anti- EGFRMonoclonal Antibodies by Comprehensive Cancer Genomics, BREAC, Study. Br J Cancer, 2017, 117:1450–8., DOI 10.1038/bjc.2017.308 Hsu H-C, Thiam TK, Lu Y-J, Yeh CY, Tsai W-S, You JF, et al. Mutations of KRAS/NRAS/BRAF Predict Cetuximab Resistance in Metastatic Colorectal Cancer Patients. Oncotarget, 2016, 7:22257–70., DOI 10.18632/oncotarget.8076 Irahara N, Baba Y, Nosho K, Shima K, Yan L, Dias-Santagata D, et al. NRAS Mutations Are Rare in Colorectal Cancer. Diagn Mol Pathol, 2010, 19:157–63., DOI 10.1097/PDM.0b013e3181c93fd1 Sanchez-Ibarra HE, Jiang X, Gallegos-Gonzalez EY, Cavazos-Gonzalez AC, Chen Y, Morcos F, et al. KRAS, NRAS, and BRAF Mutation Prevalence, Clinicopathological Association, and Their Application in a Predictive Model in Mexican Patients with Metastatic Colorectal Cancer: A Retrospective Cohort Study. PLOS ONE, 2020, 15:e0235490., DOI 10.1371/journal.pone. 0235490 Vittal A, Sharma D, Samanta I, Kasi A Rare Case of Triple Mutant, KRAS + NRAS + BRAF, Metastatic colon Adenocarcinoma. BMJ Case Rep, 2019, 12: e221816., DOI 10.1136/bcr-2017-221816 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610206 EP 1610214 DI 10.3389/pore.2022.1610206 PG 9 ER PT J AU Szekely, T Krenacs, T Maros, EM Bodor, Cs Daubner, V Csizmadia, A Vrabely, B Timar, B AF Szekely, Tamas Krenacs, Tibor Maros, Elod Mate Bodor, Csaba Daubner, Viktoria Csizmadia, Annamaria Vrabely, Brigitta Timar, Botond TI Correlations Between the Expression of Stromal Cell Activation Related Biomarkers, L-NGFR, Phospho-ERK1-2 and CXCL12, and Primary Myelofibrosis Progression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE primarymyelofibrosis progression; -stromal cell activation; -L-NGFR/CD271; -CXCL12; -phospho-ERK1- 2; -connexin 43 channels ID primarymyelofibrosis progression; -stromal cell activation; -L-NGFR/CD271; -CXCL12; -phospho-ERK1- 2; -connexin 43 channels AB In myelofibrosis, pathologically enhanced extracellular matrix production due to aberrant cytokine signalling and clonal megakaryocyte functions result(s) in impaired hemopoiesis. Disease progression is still determined by detecting reticulin and collagen fibrosis with Gomori’s silver impregnation. Here, we tested whether the expression growth related biomarkers L-NGFR/CD271, phospho-ERK1-2 and CXCL12 can be linked to the functional activation of bone marrow stromal cells during primary myelofibrosis progression. Immunoscores for all tested biomarkers showed varying strength of positive statistical correlation with the silver impregnation based myelofibrosis grades. The intimate relationship between spindle shaped stromal cells positive for all three markers and aberrant megakaryocytes was likely to reflect their functional cooperation. L-NGFR reaction was restricted to bone marrow stromal cells and revealed the whole length of their processes. Also, L-NGFR positive cells showed themost intersections, the best statistical correlations with myelofibrosis grades and the strongest interrater agreements. CXCL12 reaction highlighted stromal cell bodies and a weak extracellular staining in line with its constitutive release. Phospho-ERK1-2 reaction showed a similar pattern to CXCL12 in stromal cells with an additional nuclear staining in agreement with its role as a transcription factor. Both p-ERK1-2 and CXCL12 were also expressed at a moderate level in sinus endothelial cells. Connexin 43 gap junction communication channels, known to be required for CXCL12 release to maintain stem cell niche, were also expressed progressively in the myelofibrotic stromal network as a support of compartmental functions. Our results suggest that, diverse growth related pathways are activated in the functionally coupled bone marrow stromal cells during myelofibrosis progression. L-NGFR expression can be a useful biological marker of stromal cell activation which deserves diagnostic consideration for complementing Gomori’s silver impregnation. C1 [Szekely, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Maros, Elod Mate] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Bodor, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Daubner, Viktoria] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Csizmadia, Annamaria] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Vrabely, Brigitta] Peterfy Hospital - National Institute of Traumatology, Department of PathologyBudapest, Hungary. [Timar, Botond] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Krenacs, T (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM krenacs.tibor@med.semmelweis-univ.hu CR Barbui T, Thiele J, Gisslinger H, Kvasnicka HM, Vannucchi AM, Guglielmelli P, et al. The 2016 WHO Classification and Diagnostic Criteria for Myeloproliferative Neoplasms: Document Summary and In-Depth Discussion. Blood Cancer J, 2018, 8:15., DOI 10.1038/s41408-018-0054-y Tefferi A. Primary Myelofibrosis: 2019 Update on Diagnosis, Risk- Stratification and Management. Am J Hematol, 2018, 93:1551–60., DOI 10. 1002/ajh.25230 Schieber M, Crispino JD, Stein B. Myelofibrosis in 2019: Moving beyond JAK2 Inhibition. Blood Cancer J, 2019, 9:74., DOI 10.1038/s41408-019-0236-2 Gomori G. Silver Impregnation of Reticulum in Paraffin Sections. Am J Pathol, 1937, 13:993–5. Kvasnicka HM, Beham-Schmid C, Bob R, Dirnhofer S, Hussein K, Kreipe H, et al. Problems and Pitfalls in Grading of Bone Marrow Fibrosis, Collagen Deposition and Osteosclerosis - a Consensus-based Study. Histopathology, 2016, 68:905–15., DOI 10.1111/his.12871 Cattoretti G, Schiro R, Orazi A, Soligo D, Colombo M. Bone Marrow Stroma in Humans: Anti-nerve Growth Factor Receptor Antibodies Selectively Stain Reticular Cells In Vivo and In Vitro. Blood, 1993, 81:1726–38., DOI 10.1182/ blood.v81.7.1726.bloodjournal8171726 Covaceuszach S, Konarev PV, Cassetta A, Paoletti F, Svergun DI, Lamba D, et al. The Conundrum of the High-Affinity NGF Binding Site Formation Unveiled? Biophysical J, 2015, 108:687–97., DOI 10.1016/j.bpj.2014.11.3485 Micera A, Lambiase A, Stampachiacchiere B, Bonini S, Bonini S, Levischaffer F. Nerve Growth Factor and Tissue Repair Remodeling: trkANGFR and p75NTR, Two Receptors One Fate. Cytokine Growth Factor Rev, 2007, 18: 245–56., DOI 10.1016/j.cytogfr.2007.04.004 Barilani M, Banfi F, Sironi S, Ragni E, Guillaumin S, Polveraccio F, et al. Lowaffinity Nerve Growth Factor Receptor, CD271, Heterogeneous Expression in Adult and Fetal Mesenchymal Stromal Cells. Sci Rep, 2018, 8:9321., DOI 10. 1038/s41598-018-27587-8 Tomlinson RE, Li Z, Zhang Q, Goh BC, Li Z, Thorek DLJ, et al. NGF-TrkA Signaling by Sensory Nerves Coordinates the Vascularization and Ossification of Developing Endochondral Bone. Cel Rep, 2016, 16:2723–35., DOI 10.1016/j. celrep.2016.08.002 Zheng MG, Sui WY, He ZD, Liu Y, Huang YL, Mu SH, et al. TrkA Regulates the Regenerative Capacity of Bone Marrow Stromal Stem Cells in Nerve Grafts. Neural Regen Res, 2019, 14:1765–71., DOI 10.4103/1673-5374.257540 Bock O, Loch G, Busche G, von Wasielewski R, Schlue J, Kreipe H. Aberrant Expression of Platelet-Derived Growth Factor, PDGF, and PDGF Receptor- Alpha Is Associated with Advanced Bone Marrow Fibrosis in Idiopathic Myelofibrosis. Haematologica, 2005, 90:133–4. Bedekovics J, Kiss A, Beke L, Karolyi K, Mehes G. Platelet Derived Growth Factor Receptor-Beta, PDGFRβ, Expression Is Limited to Activated Stromal Cells in the Bone Marrow and Shows a strong Correlation with the Grade of Myelofibrosis. Virchows Arch, 2013, 463:57–65., DOI 10.1007/s00428-013- 1434-0 Bedekovics J, Szeghalmy S, Beke L, Fazekas A, Mehes G. Image Analysis of Platelet Derived Growth Factor Receptor-Beta, PDGFRβ, Expression to Determine the Grade and Dynamics of Myelofibrosis in Bone Marrow Biopsy Samples. Cytometry, 2014, 86:319–28., DOI 10.1002/cyto.b.21167 Mehes G, Tzankov A, Hebeda K, Anagnostopoulos I, Krenacs L, Bedekovics J. Platelet-derived Growth Factor Receptor β, PDGFRβ, Immunohistochemistry Highlights Activated Bone Marrow Stroma and Is Potentially Predictive for Fibrosis Progression in Prefibrotic Myeloproliferative Neoplasia. Histopathology, 2015, 67:617–24., DOI 10.1111/his.12704 Kitagawa M, Kurata M, Onishi I, Yamamoto K. Bone Marrow Niches in Myeloid Neoplasms. Pathol Int, 2020, 70:63–71., DOI 10.1111/pin.12870 Singh P, Mohammad KS, Pelus LM. CXCR4 Expression in the Bone Marrow Microenvironment Is Required for Hematopoietic Stem and Progenitor Cell Maintenance and Early Hematopoietic Regeneration after Myeloablation. Stem cells, Dayton, Ohio,, 2020, 38:849–59., DOI 10.1002/stem.3174 Schajnovitz A, Itkin T, D’Uva G, Kalinkovich A, Golan K, Ludin A, et al. CXCL12 Secretion by Bone Marrow Stromal Cells Is Dependent on Cell Contact and Mediated by Connexin-43 and Connexin-45 gap Junctions. Nat Immunol, 2011, 12:391–8., DOI 10.1038/ni.2017 Krenacs T, Rosendaal M. Connexin43 gap Junctions in normal, Regenerating, and Cultured Mouse Bone Marrow and in Human Leukemias: Their Possible Involvement in Blood Formation. Am J Pathol, 1998, 152:993–1004. Abdelouahab H, Zhang Y, Wittner M, Oishi S, Fujii N, Besancenot R, et al. CXCL12/CXCR4 Pathway Is Activated by Oncogenic JAK2 in a PI3Kdependent Manner. Oncotarget, 2017, 8:54082–95., DOI 10.18632/oncotarget. 10789 Lopez-Gil JC, Martin-Hijano L, Hermann PC, Sainz B, Jr. The CXCL12 Crossroads in Cancer Stem Cells and Their Niche. Cancers, Basel,, 2021, 13., DOI 10.3390/cancers13030469 Schepers K, Pietras EM, Reynaud D, Flach J, Binnewies M, Garg T, et al. Myeloproliferative Neoplasia Remodels the Endosteal Bone Marrow Niche into a Self-Reinforcing Leukemic Niche. Cell stem cell, 2013, 13:285–99., DOI 10. 1016/j.stem.2013.06.009 Thiele J, Kvasnicka HM, Facchetti F, Franco V, van der Walt J, Orazi A. European Consensus on Grading Bone Marrow Fibrosis and Assessment of Cellularity. Haematologica, 2005, 90:1128–32. Balla P, Maros ME, Barna G, Antal I, Papp G, Sapi Z, et al. Prognostic Impact of Reduced Connexin43 Expression and gap junction Coupling of Neoplastic Stromal Cells in Giant Cell Tumor of Bone. PloS one, 2015, 10:e0125316., DOI 10.1371/journal.pone.0125316 Maros ME, Wenz R, Forster A, Froelich MF, Groden C, Sommer WH, et al. Objective Comparison Using Guideline-Based Query of Conventional Radiological Reports and Structured Reports. In Vivo, 2018, 32:843–9., DOI 10.21873/invivo.11318 Canossa M, Twiss JL, Verity AN, Shooter EM. p75(NGFR, and TrkA Receptors Collaborate to Rapidly Activate a p75(NGFR)-associated Protein Kinase. EMBO J, 1996, 15:3369–76., DOI 10.1002/j.1460-2075.1996.tb00702.x Barker PA. p75NTR: A Study in Contrasts. Cell Death Differ, 1998, 5:346–56., DOI 10.1038/sj.cdd.4400375 Casaccia-Bonnefil P, Kong H, Chao MV. Neurotrophins: the Biological Paradox of Survival Factors Eliciting Apoptosis. Cel Death Differ, 1998, 5: 357–64., DOI 10.1038/sj.cdd.4400377 Zha K, Yang Y, Tian G, Sun Z, Yang Z, Li X, et al. Nerve Growth Factor, NGF, and NGF Receptors in Mesenchymal Stem/stromal Cells: Impact on Potential Therapies. Stem Cell translational Med, 2021, 10:1008–20., DOI 10.1002/sctm. 20-0290 Labouyrie E, Dubus P, Groppi A, Mahon FX, Ferrer J, Parrens M, et al. Expression of Neurotrophins and Their Receptors in Human Bone Marrow. Am J Pathol, 1999, 154:405–15., DOI 10.1016/s0002-9440(10)65287-x Garcia R, Aguiar J, Alberti E, de la Cuetara K, Pavon N. Bone Marrow Stromal Cells Produce Nerve Growth Factor and Glial Cell Line-Derived Neurotrophic Factors. Biochem Biophys Res Commun, 2004, 316:753–4., DOI 10.1016/j.bbrc. 2004.02.111 Caneva L, Soligo D, Cattoretti G, De Harven E, Lambertenghi Deliliers G. Immuno-electron Microscopy Characterization of Human Bone Marrow Stromal Cells with Anti-NGFR Antibodies. Blood Cell Mol Dis, 1995, 21: 73–85., DOI 10.1006/bcmd.1995.0011 Calabrese G, Giuffrida R, Lo Furno D, Parrinello N, Forte S, Gulino R, et al. Potential Effect of CD271 on Human Mesenchymal Stromal Cell Proliferation and Differentiation. Ijms, 2015, 16:15609–24., DOI 10.3390/ijms160715609 Quirici N, Soligo D, Bossolasco P, Servida F, Lumini C, Deliliers GL. Isolation of Bone Marrow Mesenchymal Stem Cells by Anti-nerve Growth Factor Receptor Antibodies. Exp Hematol, 2002, 30:783–91., DOI 10.1016/s0301- 472x(02)00812-3 Yigit N, Covey S, Barouk-Fox S, Turker T, Geyer JT, Orazi A. Nuclear Factor- Erythroid 2, Nerve Growth Factor Receptor, and CD34-Microvessel Density Are Differentially Expressed in Primary Myelofibrosis, Polycythemia Vera, and Essential Thrombocythemia. Hum Pathol, 2015, 46:1217–25., DOI 10.1016/ j.humpath.2015.05.004 Beyer C, Distler JHW. Tyrosine Kinase Signaling in Fibrotic Disorders. Biochim Biophys Acta, Bba, - Mol Basis Dis, 2013, 1832:897–904., DOI 10. 1016/j.bbadis.2012.06.008 Satomura K, Derubeis AR, Fedarko NS, Ibaraki-O’Connor K, Kuznetsov SA, Rowe DW, et al. Receptor Tyrosine Kinase Expression in Human Bone Marrow Stromal Cells. J Cel Physiol., 1998, 177:426–38., DOI 10.1002/(sici, 1097-4652(199812)177:3<426::aid-jcp6>3.0.co;2-f Dupree MA, Pollack SR, Levine EM, Laurencin CT. Fibroblast Growth Factor 2 Induced Proliferation in Osteoblasts and Bone Marrow Stromal Cells: a Whole Cell Model. Biophysical J, 2006, 91:3097–112., DOI 10.1529/biophysj.106.087098 Pages G, Lenormand P, L’Allemain G, Chambard JC, Meloche S, Pouyssegur J. Mitogen-activated Protein Kinases P42mapk and P44mapk Are Required for Fibroblast Proliferation. Proc Natl Acad Sci, 1993, 90:8319–23., DOI 10.1073/ pnas.90.18.8319 Plotnikov A, Zehorai E, Procaccia S, Seger R. The MAPK Cascades: Signaling Components, Nuclear Roles and Mechanisms of Nuclear Translocation. Biochim Biophys Acta, Bba, - Mol Cel Res, 2011, 1813:1619–33., DOI 10. 1016/j.bbamcr.2010.12.012 Song M-K, Park B-B, Uhm J-E. Understanding Splenomegaly in Myelofibrosis: Association with Molecular Pathogenesis. Ijms, 2018, 19:898., DOI 10.3390/ ijms19030898 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610217 EP 1610228 DI 10.3389/pore.2022.1610217 PG 12 ER PT J AU Mohas, A Krencz, I Varadi, Zs Arato, G Felkai, L Kiss, JD Moldvai, D Sebestyen, A Csoka, M AF Mohas, Anna Krencz, Ildiko Varadi, Zsofia Arato, Gabriella Felkai, Luca Kiss, Judit Dorottya Moldvai, Dorottya Sebestyen, Anna Csoka, Monika TI In Situ Analysis of mTORC1/C2 and Metabolism-Related Proteins in Pediatric Osteosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE osteosarcoma; mTOR; pathways; metabolic; metabolic adaptation; pediatric ID osteosarcoma; mTOR; pathways; metabolic; metabolic adaptation; pediatric AB Activation of the mTOR pathway has been observed in osteosarcoma, however the inhibition of mammalian target of rapamycin (mTOR) complex 1 has had limited results in osteosarcoma treatment. Certain metabolic pathways can be altered by mTOR activation, which can affect survival. Our aim was to characterize the mTOR profile and certain metabolic alterations in pediatric osteosarcoma to determine the interactions between the mTOR pathway and metabolic pathways. We performed immunohistochemistry on 28 samples to analyze the expression of mTOR complexes such as phospho-mTOR (pmTOR), phosphorylated ribosomal S6 (pS6), and rapamycin-insensitive companion of mTOR (rictor). To characterize metabolic pathway markers, we investigated the expression of phosphofructokinase (PFK), lactate dehydrogenase-A (LDHA), β-F1- ATPase (ATPB), glucose-6-phosphate dehydrogenase (G6PDH), glutaminase (GLS), fatty acid synthetase (FASN), and carnitin-O-palmitoyltransferase-1 (CPT1A). In total, 61% of the cases showed low mTOR activity, but higher pmTOR expression was associated with poor histological response to chemotherapy and osteoblastic subtype. Rictor expression was higher in metastatic disease and older age at the time of diagnosis. Our findings suggest the importance of the Warburg-effect, pentose-phosphate pathway, glutamine demand, and fatty-acid beta oxidation in osteosarcoma cells. mTOR activation is linked to several metabolic pathways. We suggest performing a detailed investigation of the mTOR profile before considering mTORC1 inhibitor therapy. Our findings highlight that targeting certain metabolic pathways could be an alternative therapeutic approach. C1 [Mohas, Anna] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Krencz, Ildiko] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Varadi, Zsofia] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Arato, Gabriella] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Felkai, Luca] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kiss, Judit Dorottya] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Moldvai, Dorottya] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Csoka, Monika] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Mohas, A (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM mohas.anna@med.semmelweis-univ.hu CR Pizzo PA, Poplack DG. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia PA ,USA: Lippincott Williams and Wilkins, 2015). Smeland S, Bielack SS, Whelan J, Bernstein M, Hogendoorn P, Krailo MD, et al. Survival and Prognosis with Osteosarcoma: Outcomes in More Than 2000 Patients in the EURAMOS-1, European and American Osteosarcoma Study, Cohort. Eur J Cancer, 2019, 109:36–50., DOI 10.1016/j.ejca.2018.11.027 Coller HA. Is Cancer a Metabolic Disease? Am J Pathol, 2014, 184(1):4–17., DOI 10.1016/j.ajpath.2013.07.035 Mossmann D, Park S, Hall MN. mTOR Signalling and Cellular Metabolism Are Mutual Determinants in Cancer. Nat Rev Cancer, 2018, 18(12):744–57., DOI 10.1038/s41568-018-0074-8 Wan X, Helman LJ. The Biology behind mTOR Inhibition in Sarcoma. The Oncologist, 2007, 12:1007–18., DOI 10.1634/theoncologist.12-8-1007 Wander SA, Hennessy BT, Slingerland JM. Next-generation mTOR Inhibitors in Clinical Oncology: How Pathway Complexity Informs Therapeutic Strategy. J Clin Invest, 2011, 121(4):1231–41., DOI 10.1172/jci44145 Zhou Q, Deng Z, Zhu Y, Long H, Zhang S, Zhao J. mTOR/p70S6K Signal Transduction Pathway Contributes to Osteosarcoma Progression and Patients’ Prognosis. Med Oncol, 2010, 27:1239–45., DOI 10.1007/s12032-009-9365-y Perry JA, Kiezun A, Tonzi P, Van Allen EM, Carter SL, Baca SC, et al. Complementary Genomic Approaches Highlight the PI3K/mTOR Pathway as a Common Vulnerability in Osteosarcoma. Proc Natl Acad Sci U S A, 2014, 111:E5564–73., DOI 10.1073/pnas.1419260111 Wang X, Lai P, Zhang Z, Huang M, Wang L, Yin M, et al. Targeted Inhibition of mTORC2 Prevents Osteosarcoma Cell Migration and Promotes Apoptosis. Oncol Rep, 2014, 32:382–8., DOI 10.3892/or.2014.3182 Fleuren EDG, Versleijen-Jonkers YMH, Roeffen MHS, Franssen GM, Flucke UE, Houghton PJ, et al. Temsirolimus Combined with Cisplatin or Bevacizumab Is Active in Osteosarcoma Models. Int J Cancer, 2014, 135: 2770–82., DOI 10.1002/ijc.28933 Horie R, Nakamura O, Yamagami Y, Mori M, Nishimura H, Fukuoka N, et al. Apoptosis and Antitumor Effects Induced by the Combination of an mTOR Inhibitor and an Autophagy Inhibitor in Human Osteosarcoma MG63 Cells. Int J Oncol, 2015, 48:37–44., DOI 10.3892/ijo.2015.3227 Hu K, Dai H-B, Qiu Z-L. mTOR Signaling in Osteosarcoma: Oncogenesis and Therapeutic Aspects, Review). Oncol Rep, 2016, 36:1219–25., DOI 10.3892/or. 2016.4922 Wagner LM, Fouladi M, Ahmed A, Krailo MD, Weigel B, DuBois SG, et al. Phase II Study of Cixutumumab in Combination with Temsirolimus in Pediatric Patients and Young Adults with Recurrent or Refractory Sarcoma: A Report from the Children’s Oncology Group. Pediatr Blood Cancer, 2015, 62:440–4., DOI 10.1002/pbc.25334 Whelan JS, Bielack SS, Marina N, Smeland S, Jovic G, Hook JM, et al. EURAMOS-1, an International Randomised Study for Osteosarcoma: Results from Pre-randomisation Treatment. Ann Oncol, 2015, 26:407–14., DOI 10.1093/annonc/mdu526 Adamo B, Deal AM, Burrows E, Geradts J, Hamilton E, Blackwell KL. Phosphatidylinositol 3-kinase Pathway Activation in Breast Cancer Brain Metastases. Breast Cancer Res, 2011, 13:125., DOI 10.1186/bcr3071 Krencz I, Sebestyen A, Fabian K, Mark A, Moldvay J, Khoor A. Expression of mTORC1/2-Related Proteins in Primaryand Brain Metastatic Lung Adenocarcinoma. Hum Pathol, 2017, 62:66–73., DOI 10.1016/j.humpath. 2016.12.012 Ottaviani G. The Epidemiology of Osteosarcoma. Cancer Trear Res, 2009, 152: 3–13., DOI 10.1007/978-1-4419-0284-9_1 Zhu YR, Min H. Activity of the Novel Dual Phosphatidylinositol 3-kinase/ mammalian Target of Rapamycin Inhibitor NVP-Bez235 against Osteosarcoma. Canc Biol Ther, 2015, 602–9., DOI 10.1080/15384047.2015. 1017155 Hu X. The Anti-osteosarcoma Cell Activity by a mTORC1/2 Dual inhibitorRES-529. Biochem Biophysical Res Commun, 2018, 497:499–505., DOI 10.1016/j.bbrc.2018.02.050 Gazouli I, Kyriazoglou A, Kotsantis I, Anastasiou M, Pantazopoulos A, Prevezanou M, et al. Systematic Review of Recurrent Osteosarcoma Systemic Therapy. Cancers, 2021, 13:1757., DOI 10.3390/cancers13081757 Grignani G. Sorafenib and Everolimus for Patients with Unresectable High- Grade Osteosarcoma Progressing after Standard Treatment: a Nonrandomised Phase 2 Clinical Trial. Lancet Oncol, 2015, 16:98–107., DOI 10. 1016/s1470-2045(14)71136-2 Martin-Broto J, Redondo A, Valverde C, Vaz MA, Mora J, Garcia del Muro X, et al. Gemcitabine Plus Sirolimus for Relapsed and Progressing Osteosarcoma Patients after Standard Chemotherapy: a Multicenter, Single-Arm Phase II Trial of Spanish Group for Research on Sarcoma, GEIS). Ann Oncol, 2017, 28: 2994–9., DOI 10.1093/annonc/mdx536 Wan X, Mendoza A, Khanna C, Helman LJ. Rapamycin Inhibits Ezrin- Mediated Metastatic Behavior in a Murine Model of Osteosarcoma. Cancer Res, 2005, 65(6):2406–11., DOI 10.1158/0008-5472.can-04-3135 Krencz I, Anna S, Fabian K, Mark A, Moldvay J, Khoor A, et al. Expression of mTORC1/2-Related Proteins in Primary and Brain Metastatic Lung Adenocarcinoma. Hum Pathol(2017, 62:66–73., DOI 10.1016/j.humpath.2016.12.012 Hegyi M, Semsei AF, Jakab Z, Antal I, Kiss J, Szendroi M, et al. Good Prognosis of Localized Osteosarcoma in Young Patients Treated with Limb-Salvage Surgery and Chemotherapy. Pediatr Blood Cancer, 2011, 57(3):415–22., DOI 10.1002/pbc.23172 Spraker-Perlman HL, Barkauskas DA, Krailo MD, Meyers PA, Schwartz CL, Doski J, et al. Factors Influencing Survival after Recurrence in Osteosarcoma: A Report from the Children’s Oncology Group. Pediatr Blood Cancer, 2019, 66(1):e27444., DOI 10.1002/pbc.27444 Kennedy BK, Lamming DW. The Mechanistic Target of Rapamycin: The Grand ConducTOR of Metabolism and Aging. Cell Metab. ., 2016, 23: 990–1003., DOI 10.1016/j.cmet.2016.05.009 Saxton RA, Sabatini DM. mTOR Signaling in Growth, Metabolism, and Disease. Cell(2017, 169:361–71., DOI 10.1016/j.cell.2017.03.035 Pavlova NN, Thompson CB. The Emerging Hallmarks of Cancer Metabolism. Cell Metab, 2016, 23:27–47., DOI 10.1016/j.cmet.2015.12.006 Mizushima E, Torigoe T. Osteosarcoma-initiating Cells Show High Aerobic Glycolysis and Attenuation of Oxidative Phosphorylation Mediated by LIN28B. Cancer Sci, 2020, 111:36–46., DOI 10.1111/cas.14229 Giang A-H, Raymond T, Paul B, de Mesy Bentley K, Schwarz E, O’Keefe R, et al. Mitochondrial Dysfunction and Permeability Transition inOsteosarcoma Cells Showing the Warburg Effect. J Biol Chem, 2013, 288:33303–11., DOI 10. 1074/jbc.m113.507129 Bonuccelli G, Avnet S, Grisedni G, Salerno M, Granchi D, Dominici M, et al. Role of Mesenchymal Stem Cells in Osteosarcoma and Metabolic Reprogramming of Tumor Cells. Oncotarget, 2017, 5:7575–88., DOI 10. 18632/oncotarget.2243 Wang J, Yuan W, Chen Z, Wu S, Chen J, Ge J, et al. Overexpression of G6PD Is Associated with Poor Clinical Outcome in Gastric Cancer. Tumour Biol, 2012, 33(1):95–101., DOI 10.1007/s13277-011-0251-9 Fan TW, Kucia M, Jankowski K, Higashi RM, Ratajczak J, Ratajczak MZ, et al. Rhabdomyosarcoma Cells Show an Energy Producing Anabolic Metabolic Phenotype Compared with Primary Myocytes. Mol Cancer, 2008, 7:79., DOI 10. 1186/1476-4598-7-79 Wang X, Chen K, Zhao Z. LncRNA OR3A4 Regulated the Growth of Osteosarcoma Cells by Modulating the miR-1207- 5p/G6PD Signaling. OncoTargets Ther, 2020, 13:3117–28., DOI 10.2147/ott.s234514 Altman BJ, Stine ZE, Dang CV. From Krebs to Clinic: Glutamine Metabolism to Cancer Therapy. Nat Rev Cancer, 2016, 16(10):619–34., DOI 10.1038/nrc. 2016.71 Ren L, Ruiz-Rodado V, Dowdy T, Huang S, Issaq SH, Beck J, et al. Glutaminase-1, GLS1, Inhibition Limits Metastatic Progression in Osteosarcoma. Cancer Metab, 2020, 8:4., DOI 10.1186/s40170-020-0209-8 Beloribi-Djefaflia S, Vasseur S, Guillaumond F. Lipid Metabolic Reprogramming in Cancer Cells. Oncogenesis, 2016, 5:e189., DOI 10.1038/ oncsis.2015.49 Tan Z, Xiao L, Tang M, Bai F, Li J, Li L. Targeting CPT1A-Mediated Fatty Acid Oxidation Sensitizes Nasopharyngeal Carcinoma to Radiation Therapy. Theranostics, 2018, 8:2329–47., DOI 10.7150/thno.21451 Alo PL, Amini M, Piro F. Immunohistochemical Expression and Prognostic Significance of Fatty Acid Synthase in Pancreatic Carcinoma. Anticancer Res, 2007, 27:2523–7. Orita H, Coulter J, Tully E. Inhibiting Fatty Acid Synthase for Chemoprevention of Chemically Induced Lung Tumors. Clin Cancer Res, 2008, 14:2458–64., DOI 10.1158/1078-0432.ccr-07-4177 Chen XY, Ruan HB, Long XH, Peng AF, Zhou LD, Liu M, et al. Blocking Fatty Acid Synthase Inhibits Tumor Progression of Human Osteosarcoma by Regulating the Human Epidermal Growth Factor Receptor 2/ phosphoinositide 3-kinase/protein Kinase B Signaling Pathway in Xenograft Models. Exp Ther Med, 2017, 13(5):2411–6., DOI 10.3892/etm. 2017.4284 Liu ZL, Wang G, Peng AF, Luo QF, Zhou Y, Huang SH. Fatty Acid Synthase Expression in Osteosarcoma and its Correlation with Pulmonary Metastasis. Oncol Lett, 2012, 4(5):878–82., DOI 10.3892/ol.2012.862 Felkai L, Krencz I, Nagy N, Petovari G, Danko T, Micsik T, et al. Characterization of mTOR Activity and Metabolic Profile in Pediatric Rhabdomyosarcoma. Cancers(2020, 12:1947., DOI 10.3390/cancers12071947 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610231 EP 1610241 DI 10.3389/pore.2022.1610231 PG 11 ER PT J AU Mohamed, F Kurdi, M Baeesa, S Sabbagh, JA Hakamy, S Maghrabi, Y Alshedokhi, M Dallol, A Halawa, T Najjar, A Fdl-Elmula, I AF Mohamed, Fawaz Kurdi, Maher Baeesa, Saleh Sabbagh, Jafar Abdulrahman Hakamy, Sahar Maghrabi, Yazid Alshedokhi, Mohammed Dallol, Ashraf Halawa, F. Taher Najjar, A. Ahmed Fdl-Elmula, Imad TI The Diagnostic Value of Pan-Trk Expression to Detect Neurotrophic Tyrosine Receptor Kinase (NTRK) Gene Fusion in CNS Tumours: A Study Using Next-Generation Sequencing Platform SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; next-generation sequencing; CNS tumours; NTRK-fusions; Pan-Trk; TruSight Oncology500 ID immunohistochemistry; next-generation sequencing; CNS tumours; NTRK-fusions; Pan-Trk; TruSight Oncology500 AB Background: Neurotrophic tyrosine receptor kinase (NTRK) fusion has been detected in rare types of CNS tumours, which can promote tumorigenesis. The efficacy of Trk inhibitor became a significant therapeutic interest. Our aim was to investigate whether Pan-Trk immunohistochemistry (IHC) is a reliable and efficient marker for detecting NTRK-fusion in different brain tumours. Methods: This study included 23 patients diagnosed with different types of CNS tumours. Testing for Pan-Trk IHC with monoclonal Ab (EPR17341) has been performed on all FFPE tissues. Parallelly, NTRK-rearrangements were tested using both DNA and RNA-based next-generation sequencing (NGS) assay using TruSight Onco500 platform. Results: The cohort included eight pilocytic astrocytomas, one oligodendroglioma, six IDHwildtype glioblastomas, four IDHmutant grade four astrocytomas, and one sample of each (astroblastoma, central neurocytoma, medulloblastoma, and liponeurocytoma). The mean age was 35 years; seven cases were in the paediatric age group, and 16 were adult. Pan- Trk expression was detected in 11 (47.8%) tumours, and 12 (52.1%) tumours showed no Pan-Trk expression. Nine Cases (82%) with different Pan-Trk expressions did not reveal NTRK-rearrangement. The other two positively expressed cases (liponeurocytoma and glioblastoma) were found to have NTRK2-fusions (SLC O 5A1-NTRK2, AGBL4-NTRK2, BEND5-NTRK2). All the 12 cases (100%) with no Pan-Trk expression have shown no NTRK-fusions. There was no statistically significant association between Pan-Trk expression and NTRK-fusion (p = 0.217). The detection of NTRK- fusions using NGS had high specificity over NTRK-fusion detection by using Pan-Trk IHC. Conclusion: Pan-Trk IHC is not a suitable tissue-efficient biomarker to screen for NTRKfusions in CNS tumours, however RNA-based NGS sequencing should be used as an alternative method. C1 [Mohamed, Fawaz] King Abdulaziz University, Faculty of Medicine, Department of PathologyRabigh, Saudi Arabia. [Kurdi, Maher] King Abdulaziz University, Faculty of Medicine, Department of PathologyRabigh, Saudi Arabia. [Baeesa, Saleh] King Abdulaziz University, Faculty of Medicine in Rabigh, Division of NeurosurgeryJeddah, Saudi Arabia. [Sabbagh, Jafar Abdulrahman] King Abdulaziz University, Faculty of Medicine in Rabigh, Division of NeurosurgeryJeddah, Saudi Arabia. [Hakamy, Sahar] King Abdulaziz University, Neuromuscular and Brain Tumour UnitJeddah, Saudi Arabia. [Maghrabi, Yazid] King Faisal Specialist Hospital, Department of NeuroscienceJeddah, Saudi Arabia. [Alshedokhi, Mohammed] King Abdulaziz University, Center of Excellence in Genomic Medicine ResearchJeddah, Saudi Arabia. [Dallol, Ashraf] King Abdulaziz University, Center of Excellence in Genomic Medicine ResearchJeddah, Saudi Arabia. [Halawa, F. Taher] King Abdulaziz University, Faculty of Medicine, Department of PediatricsRabigh, Saudi Arabia. [Najjar, A. Ahmed] Taibah University, College of MedicineAlmadinah Almunawwarah, Saudi Arabia. [Fdl-Elmula, Imad] Al-Neelain University, Faculty of Medicine, Department of Clinical GeneticsKhartoum, Sudan. RP Kurdi, M (reprint author), King Abdulaziz University, Faculty of Medicine, Department of Pathology, Rabigh, Saudi Arabia. EM Ahkurdi@kau.edu.sa CR Solomon JP, Benayed R, Hechtman JF, Ladanyi M. Identifying Patients with NTRK Fusion Cancer. Ann Oncol, 2019, 30:viii16–viii22. Epub 2019 Dec 24., DOI 10.1093/annonc/mdz384 Torre M, Vasudevaraja V, Serrano J, DeLorenzo M, Malinowski S, Blandin A-F, et al. Molecular and Clinicopathologic Features of Gliomas Harboring NTRK Fusions. Acta Neuropathol Commun, 2020, 8:107., DOI 10.1186/s40478- 020-00980-z Amatu A, Sartore-Bianchi A, Siena S. NTRK Gene Fusions as Novel Targets of Cancer Therapy across Multiple Tumour Types. ESMO Open, 2016, 1: e000023., DOI 10.1136/esmoopen-2015-000023 Lange A, Lo H-W. Inhibiting TRK Proteins in Clinical Cancer Therapy. Cancers, 2018, 10:105., DOI 10.3390/cancers10040105 Vaishnavi A, Le AT, Doebele RC. TRKing Down an OldOncogene in a new era of Targeted Therapy. Cancer Discov, 2015, 5:25–34., DOI 10.1158/2159-8290. cd-14-0765 Hechtman JF, Benayed R, Hyman DM, Drilon A, Zehir A, Frosina D, et al. Pan-Trk Immunohistochemistry Is an Efficient and Reliable Screen for the Detection of NTRK Fusions. Am J Surg Pathol, 2017, 41:1547–51., DOI 10.1097/ pas.0000000000000911 Solomon JP, Linkov I, Rosado A, Mullaney K, Rosen EY, Frosina D, et al. NTRK Fusion Detection across Multiple Assays and 33,997 Cases: Diagnostic Implications and Pitfalls. Mod Pathol, 2020, 33:38–46., DOI 10.1038/s41379- 019-0324-7 Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: a Summary. Neuro Oncol, 2021, 23:1231–51., DOI 10.1093/neuonc/noab106 Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, et al. EANO Guidelines on the Diagnosis and Treatment of Diffuse Gliomas of Adulthood. Nat Rev Clin Oncol, 2021, 18:170–86., DOI 10.1038/s41571-020- 00447-z Kurdi M, Alghamdi B, Butt NS, Baeesa S. The Relationship between CD204 M2-Polarized Tumour-Associated Macrophages, TAMs), Tumour- Infiltrating Lymphocytes, TILs), and Microglial Activation in Glioblastoma Microenvironment: a Novel Immune Checkpoint Receptor Target. Discov Onc, 2021, 12:28., DOI 10.1007/s12672-021-00423-8 Drilon A, Laetsch TW, Kummar S, DuBois SG, Lassen UN, Demetri GD, et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Childrenficacy of Larotrectinib in TRK Fusionepositive Cancers in Adults and Children. N Engl J Med, 2018, 378:731–9., DOI 10.1056/NEJMoa1714448 Bourhis A, Caumont C, Quintin-Roue I, Magro E, Dissaux G, Detection of NTRK Fusions in Glioblastoma: Fluorescent In Situ Hybridisation Is More Useful Than Pan-TRK Immunohistochemistry as a Screening Tool Prior to RNA Sequencing, A Remoue, et al. Detection of NTRK Fusions in Glioblastoma: Fluorescent In Situ Hybridisation Is More Useful Than Pan- TRK Immunohistochemistry as a Screening Tool Prior to RNA Sequencing. Pathology, 2021, S0031-3025(21):00429–3., DOI 10.1016/j.pathol.2021.05.100 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610233 EP 1610242 DI 10.3389/pore.2022.1610233 PG 10 ER PT J AU Nozzoli, F Lazar, A Castiglione, F Campanacci, AD Beltrami, G De Logu, F Caporalini, Ch Massi, D Roviello, G AF Nozzoli, Filippo Lazar, J. Alexander Castiglione, Francesca Campanacci, Andrea Domenico Beltrami, Giovanni De Logu, Francesco Caporalini, Chiara Massi, Daniela Roviello, Giandomenico TI NTRK Fusions Detection in Paediatric Sarcomas to Expand the Morphological Spectrum and Clinical Relevance of Selected Entities SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Pan-Trk; NTRK; ETV6-NTRK3; NTRK-rearranged tumors; paediatric sarcomas ID Pan-Trk; NTRK; ETV6-NTRK3; NTRK-rearranged tumors; paediatric sarcomas AB Undifferentiated round cell sarcomas (URCS) of soft tissue and bone and tumours of uncertain differentiation (TUD) are commonly ascribed to a subset of neoplasms with low frequency of NTRK gene fusions. However, more recently NTRK-rearranged round and spindle cell tumours have been noted in case reports and in limited or heterogeneous cohorts. The aim of our study was to investigate the presence of NTRK gene fusions in a large retrospective cohort of paediatric URCS and TUD after a systematic review of the diagnosis, according to the recently updated WHO classification scheme. One-hundred and five patients with diagnosis of URCS or TUD, involving the bone or soft tissue, were retrospectively evaluated. After the case selection and the histopathological review of the case cohort, pan-Trk immunohistochemistry (IHC) testing was performed on formalin-fixed paraffinembedded (FFPE) tissues. Tumour RNA was extracted from FFPE tissue and subjected to next-generation sequencing (NGS) library preparation, using a 10-gene NGS fusion panel, sequenced on an Illumina MiSeq. The NGS-positive cases were further confirmed by real-time PCR. On immunohistochemical screening, 12/105 (11.4%) caseswere positive using the pan- Trk antibody, showing three different staining patterns with the cytoplasmic distribution being most common. Molecular analysis using NGS and confirmed by the real-rime PCR detected two positive cases for the ETV6-NTRK3 fusion. The histological pattern of the two positive cases, together with the demonstration of the NTRK rearrangement, leaded to re-classify these previously not otherwise specified sarcomas with uncertain differentiation into the emerging category of NTRK-rearranged neoplasms. In addition, we found the two NTRK fused neoplasms showing a clinical indolent course, in contrast with literature. C1 [Nozzoli, Filippo] University of Florence, Department of Health Sciences, Section of Anatomic PathologyFlorence, Italy. [Lazar, J. Alexander] The University of Texas, M.D. Anderson Cancer Center, Departments of Pathology and Genomic MedicineHouston, TX, USA. [Castiglione, Francesca] University of Florence, Department of Health Sciences, Section of Anatomic PathologyFlorence, Italy. [Campanacci, Andrea Domenico] Careggi University Hospital, Department of Orthopaedic Oncology and Reconstructive SurgeryFlorence, Italy. [Beltrami, Giovanni] Meyer Children’s Hospital, Department of Paediatric Orthopaedic OncologyFlorence, Italy. [De Logu, Francesco] University of Florence, Department of Health Sciences, Clinical Pharmacology and Oncology UnitFlorence, Italy. [Caporalini, Chiara] University of Florence, Meyer Children’s Hospital, Pathology UnitFlorence, Italy. [Massi, Daniela] University of Florence, Department of Health Sciences, Section of Anatomic PathologyFlorence, Italy. [Roviello, Giandomenico] University of Florence, Department of Health Sciences, Medical OncologyFlorence, Italy. RP Nozzoli, F (reprint author), University of Florence, Department of Health Sciences, Section of Anatomic Pathology, Florence, Italy. EM filippo.nozzoli@unifi.it CR Sbaraglia M, Bellan E, Dei Tos AP. The 2020WHOClassification of Soft Tissue Tumours: News and Perspectives. Pathologica, 2020, 113:70–84., DOI 10.32074/ 1591-951X-213 Albert CM, Davis JL, Federman N, Casanova M, Laetsch TW. TRK Fusion Cancers in Children: A Clinical Review and Recommendations for Screening. Jco, 2019, 37(6):513–24., DOI 10.1200/JCO.18.00573 Wong DD, Vargas AC, Bonar F, Maclean F, Kattampallil J, Stewart C, et al. NTRK-rearranged Mesenchymal Tumours: Diagnostic Challenges, Morphological Patterns and Proposed Testing Algorithm. Pathology, 2020, 52(4):401–9., DOI 10.1016/j.pathol.2020.02.004 Vargas AC, Ardakani NM, Wong DD, Maclean FM, Kattampallil J, Boyle R, et al. Chromosomal Imbalances Detected in NTRK -rearranged Sarcomas by the Use of Comparative Genomic Hybridisation. Histopathology, 2020, 78(7): 932–42., DOI 10.1111/his.14295 Kao Y-C, Sung Y-S, Argani P, Swanson D, Alaggio R, Tap W, et al. NTRK3 Overexpression in Undifferentiated Sarcomas with YWHAE and BCOR Genetic Alterations. Mod Pathol, 2020, 33(7):1341–9., DOI 10.1038/s41379- 020-0495-2 Solomon JP, Linkov I, Rosado A,Mullaney K, Rosen EY, Frosina D, et al. NTRK Fusion Detection across Multiple Assays and 33,997 Cases: Diagnostic Implications and Pitfalls. Mod Pathol, 2020, 33(1):38–46., DOI 10.1038/s41379-019-0324-7 Davis JL, Lockwood CM, Stohr B, Boecking C, Al-Ibraheemi A, DuBois SG, et al. Expanding the Spectrum of Pediatric NTRK-Rearranged Mesenchymal Tumors. Am J Surg Pathol, 2019, 43(4):435–45., DOI 10.1097/PAS. 0000000000001203 Marchio C, Scaltriti M, Ladanyi M, Iafrate AJ, Bibeau F, Dietel M, et al. ESMO Recommendations on the Standard Methods to Detect NTRK Fusions in Daily Practice and Clinical Research. Ann Oncol, 2019, 30(9):1417–27., DOI 10.1093/ annonc/mdz204 Enneking WF, Spanier SS, Goodman MA. A System for the Surgical Staging of Musculoskeletal Sarcoma. Clin Orthop Relat Res, 1980, 153(153):106–120. PMID: 7449206., DOI 10.1097/00003086-198011000-00013 Tanaka K, Ozaki T. New TNM Classification, AJCC Eighth Edition, of Bone and Soft Tissue Sarcomas: JCOG Bone and Soft Tissue Tumor Study Group. Jpn J Clin Oncol, 2019, 49(2):103–7. PMID: 30423153., DOI 10.1093/jjco/hyy157 Picci P, Rougraff BT, Bacci G, Neff JR, Sangiorgi L, Cazzola A, et al. Prognostic Significance of Histopathologic Response to Chemotherapy in Nonmetastatic Ewing’s Sarcoma of the Extremities. Jco, 1993, 11(9):1763–9. PMID: 8355043., DOI 10.1200/JCO.1993.11.9.1763 Rudzinski ER, Lockwood CM, Stohr BA, Vargas SO, Sheridan R, Black JO, et al. Pan-Trk Immunohistochemistry Identifies NTRK Rearrangements in Pediatric Mesenchymal Tumors. Am J Surg Pathol, 2018, 42(7):927–35., DOI 10.1097/PAS.0000000000001062 Drilon A, Laetsch TW, Kummar S, DuBois SG, Lassen UN, Demetri GD, et al. Efficacy of Larotrectinib inTRKFusion-Positive Cancers in Adults and Children. N Engl J Med, 2018, 378(8):731–9., DOI 10.1056/NEJMoa1714448 Kheder ES, Hong DS. Emerging Targeted Therapy for Tumors with NTRK Fusion Proteins. Clin Cancer Res, 2018, 24(23):5807–14., DOI 10.1158/1078- 0432.CCR-18-1156 Tognon C, Knezevich SR, Huntsman D, Roskelley CD, Melnyk N, Mathers JA, et al. Expression of the ETV6-NTRK3 Gene Fusion as a Primary Event in Human Secretory Breast Carcinoma. Cancer Cell, 2002, 2:367–76., DOI 10. 1016/s1535-6108(02)00180-0 Rubin BP, Chen C-J, Morgan TW, Xiao S, Grier HE, Kozakewich HP, et al. Congenital Mesoblastic Nephroma T(12;15, Is Associated withETV6-NTRK3 Gene Fusion. Am J Pathol, 1998, 153:1451–8., DOI 10.1016/S0002-9440(10)65732-X Knezevich SR, McFadden DE, TaoW, Lim JF, Sorensen PHB. A Novel ETV6- NTRK3 Gene Fusion in Congenital Fibrosarcoma. Nat Genet, 1998, 18:184–7., DOI 10.1038/ng0298-184 Suurmeijer AJ, Dickson BC, Swanson D, Zhang L, Sung YS, Huang HY, et al. The Histologic Spectrum of Soft Tissue Spindle Cell Tumors with NTRK3 Gene Rearrangements. Genes Chromosomes Cancer, 2019, 58(11):739–46., DOI 10.1002/gcc.22767 Solomon JP, Hechtman JF. Detection of NTRK Fusions: Merits and Limitations of Current Diagnostic Platforms. Cancer Res, 2019, 79(13): 3163–8., DOI 10.1158/0008-5472.CAN-19-0372 Hechtman JF, Benayed R, Hyman DM, Drilon A, Zehir A, Frosina D, et al. Pan-Trk Immunohistochemistry Is an Efficient and Reliable Screen for the Detection of NTRK Fusions. Am J Surg Pathol, 2017, 41(11):1547–51., DOI 10. 1097/PAS.0000000000000911 Murphy DA, Ely HA, Shoemaker R, Boomer A, Culver BP, Hoskins I, et al. Detecting Gene Rearrangements in Patient Populations through a 2-Step Diagnostic Test Comprised of Rapid IHC Enrichment Followed by Sensitive Next-Generation Sequencing. Appl Immunohistochem Mol Morphol, 2017, 25(7):513–23., DOI 10.1097/PAI.0000000000000360 Chiang S, Cotzia P, Hyman DM, Drilon A, Tap WD, Zhang L, et al. NTRK Fusions Define a Novel Uterine Sarcoma Subtype with Features of Fibrosarcoma. Am J Surg Pathol, 2018, 42(6):791–8., DOI 10.1097/PAS. 0000000000001055 Hung YP, Fletcher CDM, Hornick JL. Evaluation of Pan-TRK Immunohistochemistry in Infantile Fibrosarcoma, Lipofibromatosis-like Neural Tumour and Histological Mimics. Histopathology, 2018, 73(4): 634–44., DOI 10.1111/his.13666 Penault-Llorca F, Rudzinski ER, Sepulveda AR. Testing Algorithm for Identification of Patients with TRK Fusion Cancer. J Clin Pathol, 2019, 72(7):460–7., DOI 10.1136/jclinpath-2018-205679 Siozopoulou V, Smits E, De Winne K, Marcq E, Pauwels P. NTRK Fusions in Sarcomas: Diagnostic Challenges and Clinical Aspects. Diagnostics, 2021, 11(3):478., DOI 10.3390/diagnostics11030478 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610237 EP 1610245 DI 10.3389/pore.2022.1610237 PG 9 ER PT J AU Yang, A Li, M Fang, M AF Yang, Ai Li, Min Fang, Mingzhi TI Corrigendum: The Research Progress of Direct KRAS G12C Mutation Inhibitors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE KRAS mutation; targeted drugs; oncogene; inhibitor; oncology ID KRAS mutation; targeted drugs; oncogene; inhibitor; oncology AB A Corrigendum on The Research Progress of Direct KRAS G12C Mutation Inhibitors by Yang, A., Li, M., and Fang, M. (2021). Pathol. Oncol. Res. 27:631095. doi: 10.3389/pore.2021. 631095 In the original article, there were mistakes in the legend for all of the figures as published. The source of the pictures cited in the article were not included. The correct legends appear below. The sources for the figures have been added to the reference list, and the reference numbering has been updated to accommodate this. The new references are appear below. The authors apologize for this error and state that authorization has been obtained from the author of each picture, and this does not change the scientific conclusions of the article in any way. The original article has been updated. C1 [Yang, Ai] Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Department of OncologyNanjing, China. [Li, Min] Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Department of OncologyNanjing, China. [Fang, Mingzhi] Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Department of OncologyNanjing, China. RP Li, M (reprint author), Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Department of Oncology, Nanjing, China. EM doctorlimin@163.com CR 3. Prior IA, Lewis PD, and Mattos C. A Comprehensive Survey of RasMutations in Cancer. Cancer Res, 2012, 72(10):2457–67., DOI 10.1158/0008-5472.can-11-2612 4. Simanshu DK, Nissley DV, and Mccormick F. RAS Proteins and Their Regulators in Human Disease. Cell, 2017, 170(1):17. 14. Huili L, Fei J, Jiwei R, Tao L, Yadong C. Research Advances on KRAS and Its Inhibitors. Acta Pharmaceutica Sinica, 2020, 44(1):43–55. 20. Xin L, Yijun W, and Pingyu L. Recent Advancement In Targeting The KRASG12C Mutant For Cancer Therapy. Acta Pharmaceutica Sinica, 2021, 56(2):374–82. 30. Fell JB, Fischer JP, Baer BR, Blake JF, Bouhana K, Briere DM, et al. Identification of the Clinical Development Candidate MRTX849, a Covalent KRASG12C Inhibitor for the Treatment of Cancer. JMed Chem, 2020, 63(13):6679–93., DOI 10.1021/ acs.jmedchem.9b02052 33. Tran TH, Alexander P, Dharmaiah S, Agamasu C, and Balius TE. The Small Molecule BI-2852 Induces A Nonfunctional Dimer Of KRAS Proc National Academy Sci., 2020, 117(7). NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610250 EP 1610254 DI 10.3389/pore.2021.1610250 PG 5 ER PT J AU Diao, B Yang, P AF Diao, Bowen Yang, Ping TI Corrigendum: Comprehensive Analysis of the Expression and Prognosis for Laminin Genes in Ovarian Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE ovarian cancer; metastasis; laminins; cancer prognosis; bioinformatical analysis ID ovarian cancer; metastasis; laminins; cancer prognosis; bioinformatical analysis AB A Corrigendum on Comprehensive Analysis of the Expression and Prognosis for Laminin Genes in Ovarian Cancer by Diao, B., Yang, P. (2021). Pathol. Oncol. Res. 27:1609855. doi: 10.3389/pore.2021.1609855 There is an error in the Funding statement. The correct number for National Natural Science Foundation of China is “82072893”. In the original article, there was an error. In the results section, the database name in the first paragraph was incorrect. A correction has been made to Results, Genetic Mutations in Laminins and Their Associations With Overall Survival and Disease-free Survival in Patients With Ovarian Cancer, paragraph one: “We evaluated genetic alterations in laminins and their relationships with the OS and DFS of OC patients registered in the cBioPortal database”. The authors apologize for this error and state that this does not change the scientific conclusions of the article in any way. The original article has been updated. C1 [Diao, Bowen] Shihezi University, School of Medicine, First Affiliated Hospital, Department of GynecologyShihezi, China. [Yang, Ping] Shihezi University, School of Medicine, First Affiliated Hospital, Department of GynecologyShihezi, China. RP Yang, P (reprint author), Shihezi University, School of Medicine, First Affiliated Hospital, Department of Gynecology, Shihezi, China. EM pingy2018@163.com NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610258 EP 1610258 DI 10.3389/pore.2022.1610258 PG 1 ER PT J AU Muller, J Szucs-Farkas, D Szegedi, I Csoka, M Garami, M Tiszlavicz, GyL Hauser, P Krivan, G Csanadi, K Ottoffy, G Nagy, B Kiss, Cs Kovacs, G AF Muller, Judit Szucs-Farkas, Dora Szegedi, Istvan Csoka, Monika Garami, Miklos Tiszlavicz, Gyorgyi Lilla Hauser, Peter Krivan, Gergely Csanadi, Krisztina Ottoffy, Gabor Nagy, Bela Kiss, Csongor Kovacs, Gabor TI Clinical Course of COVID-19 Disease in Children Treated With Neoplastic Diseases in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SARS-CoV-2; COVID-19; pediatric malignancy; chemotherapy delay; SARS-CoV-2S antibodies ID SARS-CoV-2; COVID-19; pediatric malignancy; chemotherapy delay; SARS-CoV-2S antibodies AB We report on children with cancer in Hungary suffering from COVID-19, surveying a 13- months-long period of time. We performed a retrospective clinical trial studying the medical documentation of children treated in seven centers of the Hungarian Pediatric Oncology- Hematology Group. About 10% of children admitted to tertiary hemato-oncological centers for anti-neoplastic treatment or diagnosis for de novo malignancies were positive for SARS-CoV-2 infection. Nearly two-thirds of the infected patients were asymptomatic or had only mild symptoms but showed seropositivity by 1–4.5 months after positive PCR. One third of the SARS-CoV-2-positive children were hospitalized due to symptomatic COVID-19. Five children required antiviral treatment with remdesivir. One child was referred to the intensive care unit, requiring intubation and mechanical ventilation. Delay in the scheduled anti-cancer treatment did not exceed 2 weeks in the majority (89%) of cases. There was only one patient requiring treatment deferral longer than a month. There was no COVID-19-related death in patients under 18 years of age, and nor was multisystem inflammatory syndrome diagnosed. In conclusion, SARS-CoV-2 infection did not represent an untoward risk factor among children with cancer in Hungary. C1 [Muller, Judit] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Szucs-Farkas, Dora] Medical and Health Science Center, University of Debrecen, Department of PediatricsDebrecen, Hungary. [Szegedi, Istvan] Medical and Health Science Center, University of Debrecen, Department of PediatricsDebrecen, Hungary. [Csoka, Monika] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Garami, Miklos] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Tiszlavicz, Gyorgyi Lilla] University of Szeged, Department of PediatricsSzeged, Hungary. [Hauser, Peter] Borsod-Abauj-Zemplen County Hospital, Child Health Centre, Haematology/ Oncology and Pediatric Bone Marrow Transplantation UnitMiskolc, Hungary. [Krivan, Gergely] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Csanadi, Krisztina] Heim Pal Children’s Hospital, Hemato-Oncology UnitBudapest, Hungary. [Ottoffy, Gabor] University of Pecs, Department of PediatricsPecs, Hungary. [Nagy, Bela] University of Debrecen, Faculty of Medicine, Department of Laboratory MedicineDebrecen, Hungary. [Kiss, Csongor] Medical and Health Science Center, University of Debrecen, Department of PediatricsDebrecen, Hungary. [Kovacs, Gabor] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Kiss, Cs (reprint author), Medical and Health Science Center, University of Debrecen, Department of Pediatrics, Debrecen, Hungary. EM kisscs@med.unideb.hu CR Pollard CA, Morran MP, Nestor-Kalinoski AL. The COVID-19 Pandemic: a Global Health Crisis. Physiol Genomics, 2020, 52(11):549–57., DOI 10.1152/ physiolgenomics.00089.2020 Dong Y, Mo X, Hu Y, Qi X, Jiang F, Jiang Z, et al. Epidemiology of COVID-19 Among Children in China. Pediatrics, 2020, 145:e20200702., DOI 10.1542/peds. 2020-0702 Tezer H, Bedi_r Demi_rdag T. Novel Coronavirus Disease, COVID-19, in Children. Turk J Med Sci, 2020, 50(SI-1):592–603., DOI 10.3906/sag- 2004-174 Guo C-X, He L, Yin J-Y, Meng X-G, Tan W, Yang G-P, et al. Epidemiological and Clinical Features of Pediatric COVID-19. BMC Med, 2020, 18(1):250., DOI 10.1186/s12916-020-01719-2 Loeffen EAH, Knops RRG, Boerhof J, Feijen EAM, Merks JHM, Reedijk AMJ, et al. Treatment-related Mortality in Children with Cancer: Prevalence and Risk Factors. Eur J Cancer, 2019, 121:113–22., DOI 10. 1016/j.ejca.2019.08.008 Inaba H, Pei D, Wolf J, Howard SC, Hayden RT, Go M, et al. Infectionrelated Complications during Treatment for Childhood Acute Lymphoblastic Leukemia. Ann Oncol, 2017, 28(2):386–92., DOI 10. 1093/annonc/mdw557 Liang W, Guan W, Chen R, Wang W, Li J, Xu K, et al. Cancer Patients in SARS-CoV-2 Infection: a Nationwide Analysis in China. Lancet Oncol, 2020, 21:335–7., DOI 10.1016/S1470-2045(20)30096-6 National Public Health Center. National Public Health Center, 2020). Available from: https://www.nnk.gov.hu/attachments/article/567/ Aktualiz%C3%A1lt%20elj%C3%A1r%C3%A1srend%2011.07.pdf. Hungarian Central Statistical Office. Population of Hungary by Sex and Age, 1 January. Hungarian Central Statistical Office, 2021). Available from: https://www.ksh.hu/stadat_files/nep/hu/nep0003.html, Accessed 06 21, 2021). Schuler D. Systemizing Childhood Cancer Care in Hungary: Twenty-Five Years of Progress. Med Pediatr Oncol, 1999, 32:68–70., DOI 10.1002/(sici)1096- 911x(199901)32:1<68::aid-mpo16>3.0.co;2-3 Andre N, Rouger-Gaudichon J, Brethon B, Phulpin A, Thebault E, Pertuisel S, et al. COVID-19 in Pediatric Oncology from French Pediatric Oncology and Hematology Centers: High Risk of Severe Forms? Pediatr Blood Cancer, 2020, 67:e28392., DOI 10.1002/pbc.28392 Balduzzi A, Brivio E, Rovelli A, Rizzari C, Gasperini S, Melzi ML, et al. Lessons after the Early Management of the COVID-19 Outbreak in a Pediatric Transplant and Hemato-Oncology center Embedded within a COVID-19 Dedicated Hospital in Lombardia, Italy. Estote Parati. Bone Marrow Transpl, 2020, 55(10):1900–5., DOI 10.1038/ s41409-020-0895-4 Bouffet E, Challinor J, Sullivan M, Biondi A, Rodriguez-Galindo C, Pritchard-Jones K. Early Advice on Managing Children with Cancer during the COVID-19 Pandemic and a Call for Sharing Experiences. Pediatr Blood Cancer, 2020, 67:e28327., DOI 10.1002/pbc.28327 Mayanskiy N, Luchkina P, Fedorova N, Lebedin Y, Ponomareva N. Seroconversion and Dynamics of the Anti-SARS-CoV-2 Antibody Response Related to a Hospital COVID-19 Outbreak Among Pediatric Oncology Patients. Leukemia, 2021, 35(6):1820–2., DOI 10.1038/s41375-021- 01288-0 Boulad F, Kamboj M, Bouvier N, Mauguen A, Kung AL. COVID-19 in Children with Cancer in New York City. JAMA Oncol, 2020, 6(9):1459–60., DOI 10.1001/jamaoncol.2020.2028 Madhusoodhan PP, Pierro J, Musante J, Kothari P, Gampel B, Appel B, et al. Characterization of COVID-19 Disease in Pediatric Oncology Patients: The New York-New Jersey Regional Experience. Pediatr Blood Cancer, 2021, 68(3):e28843., DOI 10.1002/pbc.28843 Millen GC, Arnold R, Cazier J-B, Curley H, Feltbower RG, Gamble A, et al. Severity of COVID-19 in Children with Cancer: Report from the United Kingdom Paediatric Coronavirus Cancer Monitoring Projec. Br J Cancer, 2021, 124:754–9., DOI 10.1038/s41416-020-01181-0 Kutluk MT, Ahmed F, Kirazli M, Bajin IY, Mungen E, Ekinci S, et al. The Effect of the COVID-19 Pandemic on Paediatric Cancer Care: Lessons Learnt from a Major Paediatric Oncology Department in Turkey. Ecancermedicalscience, 2021, 15:1172., DOI 10.3332/ecancer.2021.1172 Graetz D, Agulnik A, Ranadive R, Vedaraju Y, Chen Y, Chantada G, et al. Global Effect of the COVID-19 Pandemic on Paediatric Cancer Care: a Cross- Sectional Study. Lancet Child Adolesc Health, 2021, 5:332–40., DOI 10.1016/ S2352-4642(21)00031-6 Mukkada S, Bhakta N, Chantada GL, Chen Y, Vedarajus Y, Faughnan L, et al. Global Characteristics and Outcomes of SARS-CoV-2 Infection in Children and Adolescentswithcancer, GRCCC): a Cohortstudy. Lancet Oncol, 2021, 22(10):1416–26., DOI 10.1016/S1470-2045(21)00454-X Parambil BC, Moulik NR, Dhamne C, Dhariwal N, Narula G, Vora T, et al. COVID-19 in Children with Cancer and Continuation of Cancer-Directed Therapy during the Infection. Indian J Pediatr, 2021, 2021:1., DOI 10.1007/ s12098-021-03894-3 Liu C, Zhao Y, Okwan-Duodu D, Basho R, Cui X. COVID-19 in Cancer Patients: Risk, Clinicalf Eatures, and Management. Cancer Biol Med, 2020, 17(3):519–27., DOI 10.20892/j.issn.2095-3941.2020.0289 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610261 EP 1610266 DI 10.3389/pore.2022.1610261 PG 6 ER PT J AU Chai, Y Jiao, Sh Peng, X Gan, Q Chen, L Hu, X Hao, L Zhang, Sh Tao, Q AF Chai, Yong Jiao, Shoufeng Peng, Xin Gan, Qiang Chen, Leifeng Hu, Xiaolu Hao, Liang Zhang, Shouhua Tao, Qiang TI RING-Finger Protein 6 promotes Drug Resistance in Retinoblastoma via JAK2/STAT3 Signaling Pathway SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; signaling pathway; drug resistance; retinoblastoma; RING-finger protein 6 ID biomarker; signaling pathway; drug resistance; retinoblastoma; RING-finger protein 6 AB Chemotherapy is the first-line treatment for human retinoblastoma (RB), but the occurrence of drug resistance greatly limited its efficacy in practice. RING-finger protein 6 (RNF6) is an E3 ubiquitin ligase that is aberrantly upregulated in a range of cancers and plays important roles in cancer progression. However, the role of RNF6 in RB is largely unknown. In this study, we investigated the role of RNF6 in RB drug resistance. Two carboplatin-resistant RB cells, Y-79/CR and SO-Rb50/CR, were generated based on Y-79 and SO-Rb50 cells. RT-PCR and western blot analyses showed that RNF6 expression on both mRNA and protein levels was significantly increased in Y-79/CR and SO-Rb50/CR cells comparing to their parental cells. Knockdown of RNF6 using siRNA in Y-79/CR and SO-Rb50/CR cells resulted in cells sensitive to carboplatin on a RNF6 siRNA dose dependent manner. Similarly, RNF6 overexpression in parental Y-79 and SORb50 cells could help cells gain resistance to carboplatin on a RNF6 expression dependent manner. Signaling pathway analyses revealed that JAK2/STAT3 pathway was involved in the RNF6-induced carboplatin resistance in RB cells. We further revealed that RNF6 expression in both Y-79 and SO-Rb50 cells could render cells resistant to multiple anticancer drugs including carboplatin, vincristine and etoposide, an implication of RNF6 as a biomarker for RB drug resistance. Taken together, our study has revealed that RNF6 is upregulated in drug-resistant RB cells and RNF6 promotes drug resistance through JAK2/ STAT3 signaling pathway. The importance of RNF6 in RB cells drug resistance may represent this protein as a potential biomarker and treatment target for drug resistance in RB. C1 [Chai, Yong] The Affiliated Children’s Hospital of Nanchang University, Department of OphthalmologyNanchang, China. [Jiao, Shoufeng] The First Affiliated Hospital of Nanchang University, Department of PharmacyNanchang, China. [Peng, Xin] The Affiliated Children’s Hospital of Nanchang University, Department of General SurgeryNanchang, China. [Gan, Qiang] The Affiliated Children’s Hospital of Nanchang University, Department of OphthalmologyNanchang, China. [Chen, Leifeng] Second Affiliated Hospital of Nanchang University, Department of General SurgeryNanchang, China. [Hu, Xiaolu] The Affiliated Children’s Hospital of Nanchang University, Department of General SurgeryNanchang, China. [Hao, Liang] Second Affiliated Hospital of Nanchang University, Department of General SurgeryNanchang, China. [Zhang, Shouhua] The Affiliated Children’s Hospital of Nanchang University, Department of General SurgeryNanchang, China. [Tao, Qiang] The Affiliated Children’s Hospital of Nanchang University, Department of General SurgeryNanchang, China. RP Zhang, Sh (reprint author), The Affiliated Children’s Hospital of Nanchang University, Department of General Surgery, Nanchang, China. EM zshouhua416@163.com CR Fabian ID, Onadim Z, Karaa E, Duncan C, Chowdhury T, Scheimberg I, et al. TheManagement of Retinoblastoma. Oncogene, 2018, 37(12):1551–60., DOI 10. 1038/s41388-017-0050-x Yin X, Liao Y, Xiong W, Zhang Y, Zhou Y, Yang Y. Hypoxia-induced lncRNA ANRIL Promotes Cisplatin Resistance in Retinoblastoma Cells through Regulating ABCG2 Expression. Clin Exp Pharmacol Physiol, 2020, 47(6): 1049–57., DOI 10.1111/1440-1681.13279 Zhu X, Xue L, Yao Y, Wang K, Tan C, Zhuang M, et al. The FoxM1-ABCC4 axis Mediates Carboplatin Resistance in Human Retinoblastoma Y-79 Cells. Acta Biochim Biophys Sin, Shanghai,, 2018, 50(9):914–20., DOI 10.1093/abbs/ gmy080 Chai Y, Xiao J, Du Y, Luo Z, Lei J, Zhang S, et al. A Novel Treatment Approach for Retinoblastoma by Targeting Epithelial Growth Factor Receptor Expression with a shRNA Lentiviral System. Iran J Basic Med Sci, 2017, 20(7):739–44., DOI 10.22038/IJBMS.2017.9003 Chai Y, Xiao J, Zhang S, Du Y, Luo Z, Zhou X, et al. High-mobility Group protein B1 Silencing Promotes Susceptibility of Retinoblastoma Cells to Chemotherapeutic Drugs through Downregulating Nuclear Factor-Κb. Int J Mol Med, 2018, 41(3):1651–8., DOI 10.3892/ijmm.2018.3379 Tursun B, Schluter A, Peters MA, Viehweger B, Ostendorff HP, Soosairajah J, et al. The Ubiquitin Ligase Rnf6 Regulates Local LIM Kinase 1 Levels in Axonal Growth Cones. Genes Dev, 2005, 19(19):2307–19., DOI 10.1101/gad.1340605 Huang Z, Cai Y, Yang C, Chen Z, Sun H, Xu Y, et al. Knockdown of RNF6 Inhibits Gastric Cancer Cell Growth by Suppressing STAT3 Signaling. Ott, 2018, 11:6579–87., DOI 10.2147/ott.s174846 Liu L, Zhang Y, Wong CC, Zhang J, Dong Y, Li X, et al. RNF6 Promotes Colorectal Cancer by Activating the Wnt/β-Catenin Pathway via Ubiquitination of TLE3. Cancer Res, 2018, 78(8):1958–71., DOI 10.1158/ 0008-5472.can-17-2683 Liang Q, Ma D, Zhu X,Wang Z, Sun T-T, Shen C, et al. RING-finger Protein 6 Amplification Activates JAK/STAT3 Pathway by Modifying SHP-1 Ubiquitylation and Associates with Poor Outcome in Colorectal Cancer. Clin Cancer Res, 2018, 24(6):1473–85., DOI 10.1158/1078-0432.ccr-17-2133 Xu X, Han K, Tang X, Zeng Y, Lin X, Zhao Y, et al. The Ring finger Protein RNF6 Induces Leukemia Cell Proliferation as a Direct Target of Pre-B-cell Leukemia Homeobox 1. J Biol Chem, 2016, 291(18):9617–28., DOI 10.1074/jbc. m115.701979 Zeng Y, Xu X, Wang S, Zhang Z, Liu Y, Han K, et al. Ring finger Protein 6 Promotes Breast Cancer Cell Proliferation by Stabilizing Estrogen Receptor Alpha. Oncotarget, 2017, 8(12):20103–12., DOI 10.18632/oncotarget.15384 Xu K, Shimelis H, Linn DE, Jiang R, Yang X, Sun F, et al. Regulation of Androgen Receptor Transcriptional Activity and Specificity by RNF6-Induced Ubiquitination. Cancer cell, 2009, 15(4):270–82., DOI 10.1016/j.ccr.2009.02.021 Shukla S, Srivastava A, Kumar S, Singh U, Goswami S, Chawla B, et al. Expression of Multidrug Resistance Proteins in Retinoblastoma. Int J Ophthalmol, 2017, 10(11):1655–61., DOI 10.18240/ijo.2017.11.04 Hu K, Fu M, Wang J, Luo S, Barreto M, Singh R, et al. HSV-2 Infection of Human Genital Epithelial Cells Upregulates TLR9 Expression through the SP1/JNK Signaling Pathway. Front Immunol, 2020, 11(356):356., DOI 10.3389/ fimmu.2020.00356 Xu X, Han K, Tang X, Zeng Y, Lin X, Zhao Y, et al. The Ring Finger Protein RNF6 Induces Leukemia Cell Proliferation as a Direct Target of Pre-B-cell Leukemia Homeobox 1. J Biol Chem, 2016, 291(18):9617–28., DOI 10.1074/jbc. m115.701979 Qi L, Wang Y, Wang H, Deng J. Adenovirus 7 Induces Interlukin-6 Expression in Human Airway Epithelial Cells via p38/NF-Κb Signaling Pathway. Front Immunol, 2020, 11:551413., DOI 10.3389/fimmu.2020.551413 Zhang S, Chen K, Li W, Chai Y, Zhu J, Chu B, et al. Varied Thyroid Disrupting Effects of Perfluorooctanoic Acid, PFOA, and its Novel Alternatives Hexafluoropropylene-Oxide-Dimer-Acid, GenX, and Ammonium 4,8- Dioxa-3h-Perfluorononanoate, ADONA, In Vitro. Environ Int, 2021, 156: 106745., DOI 10.1016/j.envint.2021.106745 Zhang S, Li N, Sheng Y, Chen W, Ma Q, Yu X, et al. Hepatitis B Virus Induces Sorafenib Resistance in Liver Cancer via Upregulation of cIAP2 Expression. Infect Agents Cancer, 2021, 16(1):20., DOI 10.1186/s13027-021-00359-2 Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 Years of Image Analysis. Nat Methods, 2012, 9(7):671–5., DOI 10.1038/nmeth.2089 Sun J, Xi H-Y, Shao Q, Liu Q-H. Biomarkers in Retinoblastoma. Int J Ophthalmol, 2020, 13(2):325–41., DOI 10.18240/ijo.2020.02.18 McGranahan N, Swanton C. Biological and Therapeutic Impact of Intratumor Heterogeneity in Cancer Evolution. Cancer cell, 2015, 27(1):15–26., DOI 10. 1016/j.ccell.2014.12.001 Song W-p., Zheng S, Yao H-j., Zhou X-f., Li R, Zhang C-y., et al. Different Transcriptome Profiles between Human Retinoblastoma Y79 Cells and an Etoposide-Resistant Subline Reveal a Chemoresistance Mechanism. BMC Ophthalmol, 2020, 20(1):92., DOI 10.1186/s12886-020-01348-6 Zapolnik P, Pyrkosz A. RNF6 as an Oncogene and Potential Therapeutic Target-A Review. BioTech, 2020, 9(4):22., DOI 10.3390/biotech9040022 Lu Q, He Y, Wang Y, Gao L, Zheng Y, Zhang Z, et al. Saponins from Paris Forrestii, Takht., H. Li Display Potent Activity against Acute Myeloid Leukemia by Suppressing the RNF6/AKT/mTOR Signaling Pathway. Front Pharmacol, 2018, 9(673):673., DOI 10.3389/fphar.2018.00673 Deshaies RJ, Joazeiro CA. RING Domain E3 Ubiquitin Ligases. Annu Rev Biochem, 2009, 78:399–434., DOI 10.1146/annurev.biochem.78.101807.093809 Dimaras H, Corson TW, Cobrinik D, White A, Zhao J, Munier FL, et al. Retinoblastoma. Nat Rev Dis Primers, 2015, 1:15021., DOI 10.1038/nrdp.2015.21 Ashrafizadeh M, Zarrabi A, Orouei S, Zarrin V, Rahmani Moghadam E, Zabolian A, et al. STAT3 Pathway in Gastric Cancer: Signaling, Therapeutic Targeting and Future Prospects. Biology, 2020, 9(6):126., DOI 10.3390/ biology9060126 DeVaux RS, Ropri AS, GrimmSL, Hall PA, Herrera EO, Chittur SV, et al. Long Noncoding RNA BHLHE40-AS1 Promotes Early Breast Cancer Progression through Modulating IL-6/STAT3 Signaling. J Cel Biochem, 2020, 121(7): 3465–78., DOI 10.1002/jcb.29621 Zheng T, Ma G, Tang M, Li Z, Xu R. IL-8 Secreted from M2 Macrophages Promoted Prostate Tumorigenesis via STAT3/MALAT1 Pathway. Int J Mol Sci, 2019, 20(1):98., DOI 10.3390/ijms20010098 Yuan L, Ye J, Fan D. The B7-H4 Gene Induces Immune Escape Partly via Upregulating the PD-1/Stat3 Pathway in Non-small Cell Lung Cancer. Hum Immunol, 2020, 81(5):254–61., DOI 10.1016/j.humimm.2020.02.004 Yun HH, Kim S, Kuh H-J, Lee J-H. Downregulation of BIS Sensitizes A549 Cells for Digoxin-Mediated Inhibition of Invasion and Migration by the STAT3-dependent Pathway. Biochem Biophysical Res Commun, 2020, 524(3):643–8., DOI 10.1016/j.bbrc.2020.01.154 Matsumoto Y, Ichikawa T, Kurozumi K, Otani Y, Fujimura A, Fujii K, et al. Annexin A2-STAT3-Oncostatin M Receptor axis Drives Phenotypic and Mesenchymal Changes in Glioblastoma. Acta Neuropathol Commun, 2020, 8(1):42., DOI 10.1186/s40478-020-00916-7 Johnson DE, O’Keefe RA, Grandis JR. Targeting the IL-6/JAK/STAT3 Signalling axis in Cancer. Nat Rev Clin Oncol, 2018, 15(4):234–48., DOI 10. 1038/nrclinonc.2018.8 Kitamura H, Ohno Y, Toyoshima Y, Ohtake J, Homma S, Kawamura H, et al. Interleukin-6/STAT3 Signaling as a Promising Target to Improve the Efficacy of Cancer Immunotherapy. Cancer Sci, 2017, 108(10):1947–52., DOI 10.1111/ cas.13332 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610273 EP 1610281 DI 10.3389/pore.2022.1610273 PG 9 ER PT J AU Szita, RV Mikala, G Kozma, A Fabian, J Hardi, A Alizadeh, H Rajnics, P Rejto, L Szendrei, T Varoczy, L Nagy, Zs Illes, Valyi-Nagy, I Masszi, T Varga, G AF Szita, Reka Virag Mikala, Gabor Kozma, Andras Fabian, Janos Hardi, Apor Alizadeh, Hussain Rajnics, Peter Rejto, Laszlo Szendrei, Tamas Varoczy, Laszlo Nagy, Zsolt Illes, Arpad Valyi-Nagy, Istvan Masszi, Tamas Varga, Gergely TI Targeted Venetoclax Therapy in t(11; 14) Multiple Myeloma: Real World Data From Seven Hungarian Centers SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE multiple myeloma; venetoclax; t(11; 14); relapsed/refractory; plasma cell leukemia; amyloidosis ID multiple myeloma; venetoclax; t(11; 14); relapsed/refractory; plasma cell leukemia; amyloidosis AB Despite the introduction of novel agents, multiple myeloma remains incurable for most patients, necessitating further therapeutic options. Venetoclax, a selective BCL-2 inhibitor, had shown promising results in patients with translocation t(11;14), but questions remain open about its optimal use. We have contacted all Hungarian haematology centers for their experience treating t(11;14) myeloma patients with venetoclax. 58 patients were reported. 37 received venetoclax in the relapsed/ refractory setting with few or no other therapeutic options available. 21 patients started venetoclax as salvage after failing to achieve satisfactory response to first line therapy. In the relapsed/refractory setting objective response rate (ORR) was 94%, median progression-free survival (PFS) 10.0 months and median overall survival (OS) 14.6 months. In reinduction patients, ORR was 100%, median PFS and OS were not reached. Importantly, we found no adverse effect of high risk features such as deletion 17p or renal failure, in fact renal failure ameliorated in 42% of the cases, including three patients who became dialysis independent. Our study also reports the highest number of plasma cell leukemia cases successfully treated with venetoclax published in literature, with refractory plasma cell leukemia patients achieving a median PFS of 10.0 and a median OS of 12.2 months. C1 [Szita, Reka Virag] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Mikala, Gabor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Kozma, Andras] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Molekularis Genetikai LaboratoriumBudapest, Hungary. [Fabian, Janos] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Hardi, Apor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Alizadeh, Hussain] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Rajnics, Peter] Kaposi Mor Teaching Hospital, Department of HematologyKaposvar, Hungary. [Rejto, Laszlo] Josa Andras County HospitalNyiregyhaza, Hungary. [Szendrei, Tamas] Vas County Markusovszky HospitalSzombathely, Hungary. [Varoczy, Laszlo] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Nagy, Zsolt] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Valyi-Nagy, Istvan] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Masszi, Tamas] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Varga, Gergely] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. RP Varga, G (reprint author), Semmelweis University, Department of Internal Medicine and Haematology, Budapest, Hungary. EM vargager@gmail.com CR Gandhi UH, Cornell RF, Lakshman A, Gahvari ZJ, McGehee E, Jagosky MH, et al. Outcomes of Patients with Multiple Myeloma Refractory to CD38- Targeted Monoclonal Antibody Therapy. Leukemia, 2019, 33:2266–75., DOI 10.1038/s41375-019-0435-7 Veltri LW, Milton DR, Delgado R, Shah N, Patel K, Nieto Y, et al. Outcome of Autologous Hematopoietic Stem Cell Transplantation in Refractory Multiple Myeloma. Cancer, 2017, 123:3568–75., DOI 10.1002/cncr.30770 Jurczyszyn A, Waszczuk-Gajda A, Castillo JJ, Krawczyk K, Stork M, Pour L, et al. Primary Refractory Multiple Myeloma: a Real-World Experience with 85 Cases. Leuk Lymphoma, 2020, 61:2868–75., DOI 10.1080/10428194.2020. 1788014 Jackson GH, Davies FE, Pawlyn C, Cairns DA, Striha A, Collett C, et al. Response-adapted Intensification with Cyclophosphamide, Bortezomib, and Dexamethasone versus No Intensification in Patients with Newly Diagnosed Multiple Myeloma, Myeloma XI): a Multicentre, Open-Label, Randomised, Phase 3 Trial. Lancet Haematol, 2019, 6:e616–e629., DOI 10.1016/s2352- 3026(19)30167-x Touzeau C, Maciag P, Amiot M, Moreau P. Targeting Bcl-2 for the Treatment of Multiple Myeloma. Leukemia, 2018, 32:1899–907., DOI 10.1038/s41375-018- 0223-9 Kale J, Osterlund EJ, Andrews DW. BCL-2 Family Proteins: Changing Partners in the Dance towards Death. Cell Death Differ, 2018, 25:65–80., DOI 10.1038/ cdd.2017.186 Kapoor I, Bodo J, Hill BT, Hsi ED, Almasan A. Targeting BCL-2 in B-Cell Malignancies and Overcoming Therapeutic Resistance. Cell Death Dis, 2020, 11:941–11., DOI 10.1038/s41419-020-03144-y Kumar S, Kaufman JL, Gasparetto C, Mikhael J, Vij R, Pegourie B, et al. Efficacy of Venetoclax as Targeted Therapy for Relapsed/refractory T(11;14, Multiple Myeloma. Blood, 2017, 130:2401–9., DOI 10.1182/blood-2017-06- 788786 Kumar S, Rajkumar SV. Surrogate Endpoints in Randomised Controlled Trials: a Reality Check. The Lancet, 2019, 394:281–3., DOI 10.1016/s0140- 6736(19)31711-8 Kumar S, Harrison SJ, Cavo M, Rubia JDL, Popat R, Gasparetto CJ, et al. A Phase 3 Study of Venetoclax or Placebo in Combination with Bortezomib and Dexamethasone in Patients with Relapsed/Refractory Multiple Myeloma. Clin Lymphoma Myeloma Leuk, 2019, 19:e31., DOI 10.1016/j.clml.2019.09.046 Kumar SK, Harrison SJ, Cavo M, de la Rubia J, Popat R, Gasparetto C, et al. Venetoclax or Placebo in Combination with Bortezomib and Dexamethasone in Patients with Relapsed or Refractory Multiple Myeloma, BELLINI): a Randomised, Double-Blind, Multicentre, Phase 3 Trial. Lancet Oncol, 2020, 21:1630–42., DOI 10.1016/s1470-2045(20)30525-8 Kaufman JL, Gasparetto C, Schjesvold FH, Moreau P, Touzeau C, Facon T, et al. Targeting BCL -2 with Venetoclax and Dexamethasone in Patients with Relapsed/refractory T(11;14, Multiple Myeloma. Am J Hematol, 2021, 96: 418–27., DOI 10.1002/ajh.26083 Basali D, Chakraborty R, Rybicki L, Rosko N, Reed J, Karam M, et al. Realworld Data on Safety and Efficacy of Venetoclax-based Regimens in Relapsed/ refractory T(11;14, Multiple Myeloma. Br J Haematol, 2020, 189:1136–40., DOI 10.1111/bjh.16454 BahlisNJ, Baz R,Harrison SJ,Quach H, Ho S-J, Vangsted AJ, et al. Phase I Study of Venetoclax Plus Daratumumab and Dexamethasone, with or without Bortezomib, in Patients with Relapsed or Refractory Multiple Myeloma with and without T(11;14). Jco, 2021, 39:3602–12., DOI 10.1200/ jco.21.00443 Gasparetto C, Bowles KM, Abdallah A-O, Morris L, Mander G, Coppola S, et al. A Phase II Study of Venetoclax in Combination with Pomalidomide and Dexamethasone in Relapsed/Refractory Multiple Myeloma. Clin Lymphoma Myeloma Leuk, 2021, 21:775–84., DOI 10.1016/j.clml.2021.07.029 Costa LJ, Davies FE, Monohan GP, Kovacsovics T, Burwick N, Jakubowiak A, et al. Phase 2 Study of Venetoclax Plus Carfilzomib and Dexamethasone in Patients with Relapsed/refractory Multiple Myeloma. Blood Adv, 2021, 5(19): 3748–59., DOI 10.1182/bloodadvances.2020004146 Turesson I, Bjorkholm M, Blimark CH, Kristinsson S, Velez R, Landgren O. Rapidly Changing Myeloma Epidemiology in the General Population: Increased Incidence, Older Patients, and Longer Survival. Eur J Haematol, 2018, 101:237–44., DOI 10.1111/ejh.13083 Gran C, Uttervall K, Borg Bruchfeld J, Wallblom A, Alici E, Gahrton G, et al. Translocation, 11;14, in Newly Diagnosed Multiple Myeloma, Time to Reclassify This Standard Risk Chromosomal Aberration? Eur J Haematol, 2019, 103:588–96., DOI 10.1111/ejh.13325 Lakshman A, Alhaj Moustafa M, Rajkumar SV, Dispenzieri A, Gertz MA, Buadi FK, et al. Natural History of T(11;14, Multiple Myeloma. Leukemia, 2018, 32:131–8., DOI 10.1038/leu.2017.204 Muchtar E, Dispenzieri A, Kumar SK, Ketterling RP, Dingli D, Lacy MQ, et al. Interphase Fluorescence In Situ Hybridization in Untreated AL Amyloidosis Has an Independent Prognostic Impact by Abnormality Type and Treatment Category. Leukemia, 2017, 31:1562–9., DOI 10.1038/leu.2016.369 Tiedemann RE, Gonzalez-Paz N, Kyle RA, Santana-Davila R, Price-Troska T, VanWier SA, et al. Genetic Aberrations and Survival in Plasma Cell Leukemia. Leukemia, 2008, 22:1044–52., DOI 10.1038/leu.2008.4 Sidiqi MH, Al Saleh AS, Leung N, Jevremovic D, Aljama MA, Gonsalves WI, et al. Venetoclax for the Treatment of Translocation, 11;14, AL Amyloidosis. Blood Cancer J, 2020, 10:55., DOI 10.1038/s41408-020-0321-6 Le Bras F, Dupuis J, Lemonnier F, Oghina S, Bodez D, Ladaique A, et al. Venetoclax Induces Sustained Complete Responses in Refractory/relapsed Patients with Cardiac AL Amyloidosis. Jco, 2019, 37:e19538., DOI 10.1200/ jco.2019.37.15_suppl.e19538 Premkumar V, Comenzo RL, Lentzsch S. Preliminary Evidence of Efficacy of Venetoclax in Relapsed and Refractory AL Amyloidosis. Clin Lymphoma Myeloma Leuk, 2019, 19:e327–e328., DOI 10.1016/j.clml.2019.09.538 Leung N, Thome SD, Dispenzieri A. Venetoclax Induced a Complete Response in a Patient with Immunoglobulin Light Chain Amyloidosis Plateaued on Cyclophosphamide, Bortezomib and Dexamethasone. Haematologica, 2018, 103:e135–e137., DOI 10.3324/haematol.2017.183749 Gonsalves WI, Buadi FK, Kumar SK. Combination Therapy Incorporating Bcl- 2 Inhibition with Venetoclax for the Treatment of Refractory Primary Plasma Cell Leukemia with T, 11;14). Eur J Haematol, 2018, 100:215–7., DOI 10.1111/ ejh.12986 Jelinek T, Mihalyova J, Kascak M, Duras J, Popkova T, Benkova K, et al. Singleagent Venetoclax Induces MRD-Negative Response in Relapsed Primary Plasma Cell Leukemia with T(11;14). Am J Hematol, 2019, 94:E35–E37., DOI 10.1002/ajh.25331 Nalghranyan S, Singh AP, Schinke C. The Combination of Venetoclax, Daratumumab and Dexamethasone for the Treatment of Refractory Primary Plasma Cell Leukemia. Am J Hematol, 2020, 95:E34–5., DOI 10. 1002/ajh.25676 Glavey SV, Flanagan L, Bleach R, Kelly C, Quinn J, Ni Chonghaile T, et al. Secondary Plasma Cell Leukaemia Treated with Single Agent Venetoclax. Br J Haematol, 2020, 190:e242–5., DOI 10.1111/bjh.16858 Kupsh A, Arnall J, Voorhees P. A Successful Case of Venetoclax-Based Therapy in Relapsed/refractory Secondary Plasma Cell Leukemia. J Oncol Pharm Pract, 2020, 26:1274–8., DOI 10.1177/1078155219895072 Wang H, Zhou H, Zhang Z, Geng C, Chen W. Bortezomib-based Regimens Improve the Outcome of Patients with Primary or Secondary Plasma Cell Leukemia: A Retrospective Cohort Study. Turk J Haematol, 2020, 37:91–7., DOI 10.4274/tjh.galenos.2019.2019.0254 Durie BGM, Harousseau J-L, Harousseau J-L, Miguel JS, Blade J, Barlogie B, et al. International Uniform Response Criteria for Multiple Myeloma. Leukemia, 2006, 20:1467–73., DOI 10.1038/sj.leu.2404284 Hardi A, Bors A, Andrikovics H, Mikala G. Unexpected Equal Long-Term Overall Survival of Multiple Myeloma Harboring T(11;14), or T(4;14): A Retrospective Single Center Study. Clin Lymphoma Myeloma Leuk, 2019, 19: S328–S329., DOI 10.1016/j.clml.2019.07.316 Kumar SK, Dimopoulos MA, Kastritis E, Terpos E, Nahi H, Goldschmidt H, et al. Natural History of Relapsed Myeloma, Refractory to Immunomodulatory Drugs and Proteasome Inhibitors: a Multicenter IMWG Study. Leukemia, 2017, 31:2443–8., DOI 10.1038/leu.2017.138 Cardona-Benavides IJ, de Ramon C, Gutierrez NC. Genetic Abnormalities in Multiple Myeloma: Prognostic and Therapeutic Implications. Cells, 2021, 10: 336., DOI 10.3390/cells10020336 Hanamura I. Gain/Amplification of Chromosome Arm 1q21 in Multiple Myeloma. Cancers, 2021, 13:256., DOI 10.3390/cancers13020256 Schmidt TM, Barwick BG, Joseph N, Heffner LT, Hofmeister CC, Bernal L, et al. Gain of Chromosome 1q Is Associated with Early Progression in Multiple Myeloma Patients Treated with Lenalidomide, Bortezomib, and Dexamethasone. Blood Cancer J, 2019, 9:94., DOI 10.1038/s41408-019-0254-0 An G, Xu Y, Shi L, Shizhen Z, Deng S, Xie Z, et al. Chromosome 1q21 Gains Confer Inferior Outcomes in Multiple Myeloma Treated with Bortezomib but Copy Number Variation and Percentage of Plasma Cells Involved Have No Additional Prognostic Value. Haematologica, 2014, 99:353–9., DOI 10.3324/ haematol.2013.088211 Shah GL, Landau H, Londono D, Devlin SM, Kosuri S, Lesokhin AM, et al. Gain of Chromosome 1q Portends Worse Prognosis in Multiple Myeloma Despite Novel Agent-Based Induction Regimens and Autologous Transplantation. Leuk Lymphoma, 2017, 58:1823–31., DOI 10.1080/ 10428194.2016.1260126 Slomp A, Moesbergen LM, Gong J-n., Cuenca M, von dem Borne PA, Sonneveld P, et al. Multiple Myeloma with 1q21 Amplification Is Highly Sensitive to MCL-1 Targeting. Blood Adv, 2019, 3:4202–14., DOI 10.1182/ bloodadvances.2019000702 Wei AH, Roberts AW, Spencer A, Rosenberg AS, Siegel D, Walter RB, et al. Targeting MCL-1 in Hematologic Malignancies: Rationale and Progress. Blood Rev, 2020, 44:100672., DOI 10.1016/j.blre.2020.100672 VENCLEXTA, venetoclax). Potential Interactions for CLL/SLL [Internet], 2021). Available from: https://www.venclextahcp.com/cll/dosing/druginteractions. html., Accessed May 13, 2021). Wei AH, Montesinos P, Ivanov V, DiNardo CD, Novak J, Laribi K, et al. Venetoclax Plus LDAC for Newly Diagnosed AML Ineligible for Intensive Chemotherapy: a Phase 3 Randomized Placebo-Controlled Trial. Blood, 2020, 135:2137–45., DOI 10.1182/blood.2020004856 Jonas BA, Dinardo CD, Fracchiolla N, Pristupa A, Ishizawa K, Jin J, et al. CYP3A Inhibitors and Impact of These Agents on Outcomes in Patients with Acute Myeloid Leukemia Treated with Venetoclax Plus Azacitidine on the VIALE-A Study. Blood, 2020, 136:50–2., DOI 10.1182/blood-2020-134850 Salem AH, Agarwal SK, Dunbar M, Enschede SLH, Humerickhouse RA, Wong SL. Pharmacokinetics of Venetoclax, a Novel BCL-2 Inhibitor, in Patients with Relapsed or Refractory Chronic Lymphocytic Leukemia or Non-hodgkin Lymphoma. J Clin Pharmacol, 2017, 57:484–92., DOI 10. 1002/jcph.821 Hetavi M, Bharatsinh G, Nene U, Obiora M, Arif A, Dawn C. Abstract 11835: Evaluation of Cardiovascular Adverse Events Associated with Ibrutinib, Venetoclax and Idelalisib Used in Treatment of Chronic Lymphocytic Leukemia. Circulation, 2018, 138(Suppl. l_1):A11835. Belicza E, Janosi A. Study of Incidence and Treatment of Acute Myocardial Infarction by Evaluating the Financing Database: 2004-2009. Orvosi Hetilap, 2012, 153(3):102–12., DOI 10.1556/oh.2012.29270 Libourel EJ, Sonneveld P, van der Holt B, de Maat MPM, Leebeek FWG. High Incidence of Arterial Thrombosis in Young Patients Treated for Multiple Myeloma: Results of a Prospective Cohort Study. Blood, 2010, 116(1):22–6., DOI 10.1182/blood-2009-12-257519 Plummer C, Driessen C, Szabo Z, Mateos MV. Management of Cardiovascular Risk in Patients with Multiple Myeloma. Blood Cancer J, 2019, 9(3):26–12., DOI 10.1038/s41408-019-0183-y Stewart AK, Rajkumar SV, Dimopoulos MA, Masszi T, Spicka I, Oriol A, et al. Carfilzomib, Lenalidomide, and Dexamethasone for Relapsed Multiple Myeloma. N Engl J Med, 2015, 372(2):142–52., DOI 10.1056/ nejmoa1411321 Swan D, Delaney C, Natoni A, O’Dwyer M, Krawczyk J. Successful Venetoclax Salvage in the Setting of Refractory, Dialysis-dependent Multiple Myeloma with T(11;14). Haematologica, 2020, 105:e141–e143., DOI 10.3324/haematol. 2019.228338 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610276 EP 1610284 DI 10.3389/pore.2022.1610276 PG 9 ER PT J AU Jiang, Y Zhang, Z Wang, X Feng, Z Hong, B Yu, D Wang, Y AF Jiang, Yu Zhang, Zhiqiang Wang, Xian Feng, Zhenzhong Hong, Bo Yu, Dexin Wang, Yi TI A Novel Prognostic Factor TIPE2 in Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE epithelial-mesenchymal transition; bladder cancer; TIPE2; cystectomy; urothelial carcinoma ID epithelial-mesenchymal transition; bladder cancer; TIPE2; cystectomy; urothelial carcinoma AB Objective: We sought to identify tumor necrosis factor (TNF)-alpha-induced protein 8-like 2 (TIPE2/TNFAIP8L2) expression in bladder cancer and its relationship to clinicopathological findings and prognosis. Methods: Immunohistochemical (IHC) staining for TIPE2 was performed on 110 archived radical cystectomy specimens. Ten high-power fields were randomly selected from each specimen to observe and record the percentage of immunoreactive cells of TIPE2 in tumor cells (grade 0–4) and the corresponding immunostaining intensity (grade 0–3). The expression score of TIPE2 was obtained by multiplying the results of the above two scores, which ranged from 0 to 12 points. The cut-off point of the sum of the scores were defined as follows: 0–3 scores were defined as negative expression (-); >3 scores were classified as positive expression, < 7, low expression, ≥7, high expression. Results: In 110 cases, TIPE2 was stained in various degrees in bladder cancer tissues, and expressed in both nucleus and cytoplasm. 4.5% (5/110) showed negative expression, 40.9% (45/110) showed low expression, and 54.5% (60/110) showed high expression. TIPE2 expression was negatively correlated with lymph node metastasis (p = 0.004) and disease progression (p = 0.021). Survival curves were plotted to show that patients with high TIPE2 expression had a progression-free survival curve above those with negative/ low TIPE2 expression (p = 0.027). In multivariate Cox proportional hazard regression analysis, TIPE2 was a protective factor for progression-free survival in bladder urothelial carcinoma (p = 0.031), pT stage (p = 0.016) was a risk factor for progression-free survival, and age was a risk factor for overall survival (p = 0.020). Conclusion: TIPE2 may be a new biomarker to predict the disease progression and prognosis of patients with urothelial carcinoma of the bladder. C1 [Jiang, Yu] Second Affiliated Hospital of Anhui Medical University, Department of UrologyHefei, China. [Zhang, Zhiqiang] Second Affiliated Hospital of Anhui Medical University, Department of UrologyHefei, China. [Wang, Xian] Second Affiliated Hospital of Anhui Medical University, Department of PathologyHefei, China. [Feng, Zhenzhong] Second Affiliated Hospital of Anhui Medical University, Department of PathologyHefei, China. [Hong, Bo] Chinese Academy of Sciences, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Anhui Province Key Laboratory of Medical Physics and TechnologyHefei, China. [Yu, Dexin] Second Affiliated Hospital of Anhui Medical University, Department of UrologyHefei, China. [Wang, Yi] Second Affiliated Hospital of Anhui Medical University, Department of UrologyHefei, China. RP Wang, Y (reprint author), Second Affiliated Hospital of Anhui Medical University, Department of Urology, Hefei, China. EM wangyi2035@ahmu.edu.cn CR Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2020. CA A Cancer J Clin, 20202020, 70(1):7–30., DOI 10.3322/caac.21590 Global Burden of Disease Cancer CollaborationAbate D, Fitzmaurice C, Abate D, Abbasi N, Abbastabar H, Abd-Allah F, et al. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived with Disability, and Disability-Adjusted Life-Years for 29 Cancer Groups, 1990 to 2017: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol, 2019, 5(12):1749–68., DOI 10.1001/jamaoncol.2019.2996 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer Statistics in China, 2015. CA: A Cancer J Clinicians, 2016, 66(2):115–32., DOI 10.3322/ caac.21338 Gakis G, Stenzl A. CanWe Rely on Lymphovascular Invasion for Determining the Need for Adjuvant Chemotherapy in Organ-Confined Bladder Cancer? BJU Int, 2015, 116(1):3–4., DOI 10.1111/bju.13047 Bordoloi D, Banik K, Shabnam B, Padmavathi G, Monisha J, Arfuso F, et al. TIPE Family of Proteins and its Implications in Different Chronic Diseases. Ijms, 2018, 19(10):2974., DOI 10.3390/ijms19102974 Padmavathi G, Banik K, Monisha J, Bordoloi D, Shabnam B, Arfuso F, et al. Novel Tumor Necrosis Factor-α Induced Protein Eight, TNFAIP8/TIPE, Family: Functions and Downstream Targets Involved in Cancer Progression. Cancer Lett, 2018, 432:260–71., DOI 10.1016/j.canlet.2018.06.017 Niture S, Dong X, Arthur E, Chimeh U, Niture S, Zheng W, et al. Oncogenic Role of Tumor Necrosis Factor α-Induced Protein 8, TNFAIP8). Cells, 2018, 8(1):9., DOI 10.3390/cells8010009 Gus-Brautbar Y, Johnson D, Zhang L, Sun H, Wang P, Zhang S, et al. The Antiinflammatory TIPE2 Is an Inhibitor of the Oncogenic Ras. Mol Cel, 2012, 45(5):610–8., DOI 10.1016/j.molcel.2012.01.006 Liu Q-Q, Zhang F-F, Wang F, Qiu J-H, Luo C-H, Zhu G-Y, et al. TIPE2 Inhibits Lung Cancer Growth Attributing to Promotion of Apoptosis by Regulating Some Apoptotic Molecules Expression. PLoS One, 2015, 10(5): e0126176., DOI 10.1371/journal.pone.0126176 Zhao Q, Zhao M, Dong T, Zhou C, Peng Y, Zhou X, et al. Tumor Necrosis Factor-α-Induced Protein-8 Like-2, TIPE2, Upregulates P27 to Decrease Gastic Cancer Cell Proliferation. J Cel Biochem., 2015, 116(6):1121–9., DOI 10.1002/jcb.25068 Zhu L, Zhang X, Fu X, Li Z, Sun Z, Wu J, et al. TIPE2 Suppresses Progression and Tumorigenesis of Esophageal Carcinoma via Inhibition of the Wnt/β- Catenin Pathway. J Transl Med, 2018, 16(1):7., DOI 10.1186/s12967-018- 1383-0 Lu Q, Liu Z, Li Z, Chen J, Liao Z, Wu W-r., et al. TIPE2 Overexpression Suppresses the Proliferation, Migration, and Invasion in Prostate Cancer Cells by Inhibiting PI3K/Akt Signaling Pathway. Oncol Res, 2016, 24(5):305–13., DOI 10.3727/096504016X14666990347437 Zhang Z, Qi H, Hou S, Jin X. TIPE2 mRNA Overexpression Correlates with TNM Staging in Renal Cell Carcinoma Tissues. Oncol Lett, 2013, 6(2):571–5., DOI 10.3892/ol.2013.1388 Li X-M, Su J-R, Yan S-P, Cheng Z-L, Yang T-T, Zhu Q. A Novel Inflammatory Regulator TIPE2 Inhibits TLR4-Mediated Development of colon Cancer via Caspase-8. Cbm, 2014, 14(4):233–40., DOI 10.3233/ CBM-140402 Jia W, Li Z, Chen J, Sun L, Liu C, Wang S, et al. TIPE2 Acts as a Biomarker for Tumor Aggressiveness and Suppresses Cell Invasiveness in Papillary Thyroid Cancer, PTC). Cell Biosci, 2018, 8:49., DOI 10.1186/s13578-018- 0247-x Mostofi FK, Sobin LH, Tosoni I. Histological Typing of Urinary Bladder Tumours, International Classification of Tumours, No 19. Geneva: World Health Organisation, 1973). Brierley JD, Gospodarowicz MK, Wittekind C. TNM Classification of Malignant Tumours, 8th Edition. J Clin Pathol, 1998, 51(1):84., DOI 10. 1136/jcp.43.4.350-e Yoo SH, KimH, Kwak C, Kim HH, Jung JH, Ku JH. Late Recurrence of Bladder Cancer Following Radical Cystectomy: Characteristics and Outcomes. Urol Int, 2019, 103:291–6., DOI 10.1159/000502656 Babjuk M, Bohle A, Burger M, Capoun O, Cohen D, Comperat EM, et al. EAU Guidelines on Non-muscle-invasive Urothelial Carcinoma of the Bladder: Update 2016. Eur Urol, 2017, 71:447–61., DOI 10.1016/j.eururo. 2016.05.041 Pastushenko I, Blanpain C. EMT Transition States during Tumor Progression and Metastasis. Trends Cel Biol, 2019, 29(3):212–26., DOI 10.1016/j.tcb.2018. 12.001 Cai Z, Li H. Circular RNAs and Bladder Cancer. Ott, 2020, Vol. 13:9573–86., DOI 10.2147/OTT.S268859 Garg M, Maurya N. WNT/β-catenin Signaling in Urothelial Carcinoma of Bladder. Wjn, 2019, 8(5):83–94., DOI 10.5527/wjn.v8.i5.83 Zhang L, Shi Y, Wang Y, Zhu F, Wang Q, Ma C, et al. The Unique Expression Profile of Human TIPE2 Suggests New Functions beyond its Role in Immune Regulation. Mol Immunol, 2011, 48(9-10):1209–15., DOI 10.1016/j.molimm. 2011.03.001 Liu Z-j., Liu H-l., Zhou H-c., Wang G-c. TIPE2 Inhibits Hypoxia-Induced Wnt/β-Catenin Pathway Activation and EMT in Glioma Cells. Oncol Res, 2016, 24(4):255–61., DOI 10.3727/096504016X14666990347356 Wu J, Zhang H, Xu C, Xu H, Zhou X, Xie Y, et al. TIPE2 Functions as a Metastasis Suppressor via Negatively Regulating β-catenin through Activating GSK3β in Gastric Cancer. Int J Oncol, 2016, 48(1):199–206., DOI 10.3892/ijo. 2015.3224 Liu Y, Wang X, Wan L, Liu X, Yu H, Zhang D, et al. TIPE2 Inhibits the Migration and Invasion of Endometrial Cells by Targeting β-catenin to Reverse Epithelial-Mesenchymal Transition. Hum Reprod, 2020, 35(6):1377–90., DOI 10.1093/humrep/deaa062 Najafi M, Goradel NH, Farhood B, Salehi E, Solhjoo S, Toolee H, et al. Tumor Microenvironment: Interactions and Therapy. J Cel Physiol, 2019, 234(5): 5700–21., DOI 10.1002/jcp.27425 Terry S, Savagner P, Ortiz-Cuaran S, Mahjoubi L, Saintigny P, Thiery J-P, et al. New Insights into the Role of EMT in Tumor Immune Escape. Mol Oncol, 2017, 11(7):824–46., DOI 10.1002/1878-0261.12093 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610282 EP 1610289 DI 10.3389/pore.2022.1610282 PG 8 ER PT J AU Luo, W Nagaria, T Sun, H Ma, J Lombardo, J Bassett, R Cao, A Tan, D AF Luo, Wenyi Nagaria, S. Teddy Sun, Hongxia Ma, Junsheng Lombardo, L. Jamie Bassett, Roland Cao, C. Austin Tan, Dongfeng TI Expression and Potential Prognostic Value of SOX9, MCL-1 and SPOCK1 in Gastric Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; gastric cancer; SPOCK1; SOX9; MCL-1 ID prognosis; gastric cancer; SPOCK1; SOX9; MCL-1 AB Gastric cancer is a common malignancy and remains one of the leading causes of cancerrelated deaths, though its incidence is in decline in most developed countries. One of the major challenges of treating gastric cancer is tumor heterogeneity, which portends a high degree of prognostic variance and the necessity for different treatment modalities. Tumor heterogeneity is at least in part due to divergent differentiation of tumor cells to clones harboring different molecular alterations. Here we studied the expression of emerging prognostic markers SOX9, MCL-1, and SPOCK1 (Testican-1) in a cohort of gastric cancer by immunohistochemistry and investigated how individual biomarkers and their combinations predict disease prognosis. We found frequent expression of SPOCK1 (in both nuclei and cytoplasm), MCL-1 and SOX9 in gastric cancer. In univariate analysis, nuclear SPOCK1 expression and pathologic TNM stage were negative prognostic markers in this cohort. In multivariate analysis, SOX9 expression stood out as a predictor of poor prognosis. Further subgroup analysis suggested prognostic value of SOX9 expression in poorly differentiated gastric adenocarcinoma. MCL-1 showed no prognostic role in this cohort. C1 [Luo, Wenyi] University of Oklahoma, Health Sciences Center, Department of PathologyOklahoma City, OK, USA. [Nagaria, S. Teddy] The University of Texas M.D. Anderson Cancer Center, Department of PathologyHouston, TX, USA. [Sun, Hongxia] The University of Texas M.D. Anderson Cancer Center, Department of PathologyHouston, TX, USA. [Ma, Junsheng] The University of Texas M.D. Anderson Cancer Center, Department of BiostatisticsHouston, TX, USA. [Lombardo, L. Jamie] The University of Texas M.D. Anderson Cancer Center, Department of PathologyHouston, TX, USA. [Bassett, Roland] The University of Texas M.D. Anderson Cancer Center, Department of BiostatisticsHouston, TX, USA. [Cao, C. Austin] University of Pennsylvania, School of MedicinePhiladelphia, PA, USA. [Tan, Dongfeng] The University of Texas M.D. Anderson Cancer Center, Department of PathologyHouston, TX, USA. RP Luo, W (reprint author), University of Oklahoma, Health Sciences Center, Department of Pathology, Oklahoma City, USA. EM wenyi-luo@ouhsc.edu CR Rawla P, Barsouk A Epidemiology of Gastric Cancer: Global Trends, Risk Factors and Prevention. pg, 2019, 14:26–38., DOI 10.5114/pg.2018.80001 Bergquist JR, Leiting JL, Habermann EB, Cleary SP, Kendrick ML, Smoot RL, et al. Early-onset Gastric Cancer Is a Distinct Disease with Worrisome Trends and Oncogenic Features. Surgery, 2019, 166:547–55., DOI 10.1016/j.surg.2019. 04.036 D’Angelica M, Gonen M, Brennan MF, Turnbull AD, Bains M, Karpeh MS Patterns of Initial Recurrence in Completely Resected Gastric Adenocarcinoma. Ann Surg, 2004, 240:808–16., DOI 10.1097/01.sla. 0000143245.28656.15 Ajani JA, Lee J, Sano T, Janjigian YY, Fan D, Song S Gastric Adenocarcinoma. Nat Rev Dis Primers, 2017, 3:17036., DOI 10.1038/nrdp.2017.36 Bang Y-J, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, et al. Trastuzumab in Combination with Chemotherapy versus Chemotherapy Alone for Treatment of HER2-Positive Advanced Gastric or Gastro- Oesophageal junction Cancer, ToGA): a Phase 3, Open-Label, Randomised Controlled Trial. The Lancet, 2010, 376:687–97., DOI 10.1016/s0140-6736(10, 61121-x Hudler P Challenges of Deciphering Gastric Cancer Heterogeneity. Wjg, 2015, 21:10510–27., DOI 10.3748/wjg.v21.i37.10510 Cancer Genome Atlas Research N. Comprehensive Molecular Characterization of Gastric Adenocarcinoma. Nature, 2014, 513:202–9., DOI 10.1038/nature13480 Lim B, Kim JH, Kim M, Kin SY. Genomic and Epigenomic Heterogeneity in Molecular Subtypes of Gastric Cancer. Wjg, 2016, 22:1190–201., DOI 10.3748/ wjg.v22.i3.1190 Truong CD, Feng W, Li W, Khoury T, Li Q, Alrawi S, et al. Characteristics of Epstein-Barr Virus-Associated Gastric Cancer: a Study of 235 Cases at a Comprehensive Cancer center in U.S.A. J Exp Clin Cancer Res, 2009, 28:14., DOI 10.1186/1756-9966-28-14 Campbell KJ, Dhayade S, Ferrari N, Sims AH, Johnson E, Mason SM, et al. MCL-1 Is a Prognostic Indicator and Drug Target in Breast Cancer. Cell Death Dis, 2018, 9:19., DOI 10.1038/s41419-017-0035-2 Bartley AN, Washington MK, Ventura CB, Ismaila N, Colasacco C, Benson AB, et al. HER2Testing and Clinical Decision Making in Gastroesophageal Adenocarcinoma. Am J Clin Pathol, 2016, 146:647–69., DOI 10.1093/ajcp/aqw206 Kulangara K, Zhang N, Corigliano E, Guerrero L, Waldroup S, Jaiswal D, et al. Clinical Utility of the Combined Positive Score for Programmed Death Ligand-1 Expression and the Approval of Pembrolizumab for Treatment of Gastric Cancer. Arch Pathol Lab Med, 2019, 143:330–7., DOI 10.5858/arpa.2018-0043-oa Mesquita P, Freire AF, Lopes N, Gomes R, Azevedo D, Barros R, et al. Expression and Clinical Relevance of SOX9 in Gastric Cancer. Dis Markers, 2019, 2019:8267021., DOI 10.1155/2019/8267021 Lee W-S, Park Y-L, Kim N, Oh H-H, Son D-J, Kim M-Y, et al. Myeloid Cell Leukemia-1 Regulates the Cell Growth and Predicts Prognosis in Gastric Cancer. Int J Oncol, 2015, 46:2154–1262., DOI 10.3892/ijo.2015.2890 Kawaguchi Y Sox9 and Programming of Liver and Pancreatic Progenitors. J Clin Invest, 2013, 123:1881–6., DOI 10.1172/jci66022 Matheu A, Collado M, Wise C, Manterola L, Cekaite L, Tye AJ, et al. Oncogenicity of the Developmental Transcription Factor Sox9. Cancer Res, 2012, 72:1301–15., DOI 10.1158/0008-5472.can-11-3660 Larsimont J-C, Youssef KK, Sanchez-Danes A, Sukumaran V, Defrance M, Delatte B, et al. Sox9 Controls Self-Renewal of Oncogene Targeted Cells and Links Tumor Initiation and Invasion. Cell Stem Cell, 2015, 17:60–73., DOI 10. 1016/j.stem.2015.05.008 Kawai T, Yasuchika K, Ishii T, Miyauchi Y, Kojima H, Yamaoka R, et al. SOX9 Is a Novel Cancer Stem Cell Marker Surrogated by Osteopontin in Human Hepatocellular Carcinoma. Sci Rep, 2016, 6:30489., DOI 10.1038/srep30489 Santos JC, Carrasco-Garcia E, Garcia-PugaM, Aldaz P, Montes M, Fernandez- Reyes M, et al. SOX9 Elevation Acts with Canonical WNT Signaling to Drive Gastric Cancer Progression. Cancer Res, 2016, 76:6735–46., DOI 10.1158/0008- 5472.can-16-1120 Song S, Maru DM, Ajani JA, Chan C-H, Honjo S, Lin H-K, et al. Loss of TGF-β Adaptor β2SP Activates Notch Signaling and SOX9 Expression in Esophageal Adenocarcinoma. Cancer Res, 2013, 73:2159–69., DOI 10.1158/0008-5472.can- 12-1962 Sashikawa Kimura M, Mutoh H, Sugano K SOX9 Is Expressed in normal Stomach, Intestinal Metaplasia, and Gastric Carcinoma in Humans. J Gastroenterol, 2011, 46:1292–9., DOI 10.1007/s00535-011-0443-5 Sun M, Uozaki H, Hino R, Kunita A, Shinozaki A, Ushiku T, et al. SOX9 Expression and its Methylation Status in Gastric Cancer. Virchows Arch, 2012, 460:271–9., DOI 10.1007/s00428-012-1201-7 Zhou C-J, Guo J-Q, Zhu K-X, Zhang Q-H, Pan C-R, Xu W-H, et al. Elevated Expression of SOX9 Is Related with the Progression of Gastric Carcinoma. Diagn Cytopathol, 2011, 39:105–9., DOI 10.1002/dc.21348 Choi YJ, Song JH, Yoon JH, Choi WS, Nam SW, Lee JY, et al. Aberrant Expression of SOX9 Is Associated with Gastrokine 1 Inactivation in Gastric Cancers. Gastric Cancer, 2014, 17:247–54., DOI 10.1007/s10120- 013-0277-3 Zhang N, Chai D, Du H, Li K, Xie W, Li X, et al. Expression of Reg IV and SOX9 and Their Correlation in Human Gastric Cancer. BMC Cancer, 2018, 18:344., DOI 10.1186/s12885-018-4285-x Croft B, Ohnesorg T, Hewitt J, Bowles J, Quinn A, Tan J, et al. Human Sex Reversal Is Caused by Duplication or Deletion of Core Enhancers Upstream of SOX9. Nat Commun, 2018, 9:5319., DOI 10.1038/s41467-018-07784-9 Diaz P, Valenzuela ValderramaM, Bravo J, Quest AFG Helicobacter pylori and Gastric Cancer: Adaptive Cellular Mechanisms Involved in Disease Progression. Front Microbiol, 2018, 9:5., DOI 10.3389/fmicb.2018.00005 Bradshaw AD, Sage EH SPARC, a Matricellular Protein that Functions in Cellular Differentiation and Tissue Response to Injury. J Clin Invest, 2001, 107:1049–54., DOI 10.1172/jci12939 Wang T, Liu X, Tian Q, Liang T, Chang P. Reduced SPOCK1 Expression Inhibits Non-small Cell Lung Cancer Cell Proliferation and Migration through Wnt/β-Catenin Signaling. Eur Rev Med Pharmacol Sci, 2018, 22:637–44., DOI 10.26355/eurrev_201802_14288 Zhang LQ, Wang Y, Zhang L. Effects of shRNA-Mediated Knockdown of SPOCK1 on Ovarian Cancer Growth and Metastasis. Cel Mol Biol, Noisy-legrand,, 2015, 61:102–9., DOI 10.14715/cmb/2015.61.7.16 Yang J, Yang Q, Yu J, Li X, Yu S, Zhang X SPOCK1 Promotes the Proliferation, Migration and Invasion of Glioma Cells through PI3K/AKT and Wnt/β-Catenin Signaling Pathways. Oncol Rep, 2016, 35:3566–76., DOI 10.3892/or.2016.4757 Veenstra VL, Damhofer H, Waasdorp C, Steins A, Kocher HM, Medema JP, et al. Stromal SPOCK1 Supports Invasive Pancreatic Cancer Growth. Mol Oncol, 2017, 11:1050–64., DOI 10.1002/1878-0261.12073 Marimuthu A, Jacob HK, Jakharia A, Subbannayya Y, Keerthikumar S, Kashyap MK, et al. Gene Expression Profiling of Gastric Cancer. J Proteomics Bioinform, 2011, 4:74–82., DOI 10.4172/jpb.1000170 Chen D, Zhou H, Liu G, Zhao Y, Cao G, Liu Q. SPOCK1 Promotes the Invasion and Metastasis of Gastric Cancer through Slug-Induced Epithelial- Mesenchymal Transition. J Cel Mol Med, 2018, 22:797–807., DOI 10.1111/ jcmm.13357 Mondal T, Juvvuna PK, Kirkeby A, Mitra S, Kosalai ST, Traxler L, et al. Sense- Antisense lncRNA Pair Encoded by Locus 6p22.3 Determines Neuroblastoma Susceptibility via the USP36-CHD7-SOX9 Regulatory Axis. Cancer Cell, 2018, 33:417–34., DOI 10.1016/j.ccell.2018.01.020 Kaufmann SH, Karp JE, Svingen PA, Krajewski S, BurkePJ, Gore SD, et al.Elevated Expression of the Apoptotic Regulator Mcl-1 at the Time of Leukemic Relapse. Blood, 1998, 91:991–1000., DOI 10.1182/blood.v91.3.991.991_991_1000 Willis SN, Chen L, Dewson G, Wei A, Naik E, Fletcher JI, et al. Proapoptotic Bak Is Sequestered by Mcl-1 and Bcl-xL, but Not Bcl-2, until Displaced by BH3-Only Proteins. Genes Dev, 2005, 19:1294–305., DOI 10.1101/gad.1304105 Lee W-S, Kim N, Park Y-R, Oh H-H, Myung E, Kim S-H, et al. Myeloid Cell Leukemia-1 Promotes Epithelial-Mesenchymal Transition of Human Gastric Cancer Cells. Oncol Rep, 2015, 34:1011–6., DOI 10.3892/or.2015.4040 Maeta Y, Tsujitani S, Matsumoto S, Yamaguchi K, Tatebe S, Kondo A, et al. Expression of Mcl-1 and P53 Proteins Predicts the Survival of Patients with T3 Gastric Carcinoma. Gastric Cancer, 2004, 7:78–84., DOI 10.1007/s10120-004- 0272-9 Hu C-J, Wang B, Tang B, Chen B-j., Xiao Y-F, Qin Y, et al. The FOXM1- Induced Resistance to Oxaliplatin Is Partially Mediated by its Novel Target Gene Mcl-1 in Gastric Cancer Cells. Biochim Biophys Acta, Bba, - Gene Regul Mech, 2015, 1849:290–9., DOI 10.1016/j.bbagrm.2014.11.008 WacheckV, CejkaD, Sieghart W, LosertD, Strommer S, Crevenna R, et al.Mcl-1 Is a Relevant Molecular Target for Antisense Oligonucleotide Strategies in Gastric Cancer Cells. Cancer Biol Ther, 2006, 5:1348–54., DOI 10.4161/cbt.5.10.3224 Bhattacharyya A, Chattopadhyay R, Hall EH, Mebrahtu ST, Ernst PB, Crowe SE Mechanism of Hypoxia-Inducible Factor 1α-Mediated Mcl1 Regulation in Helicobacter Pylori-Infected Human Gastric Epithelium. Am J Physiology- Gastrointestinal Liver Physiol, 2010, 299:G1177–G1186., DOI 10.1152/ajpgi. 00372.2010 Zhan Z, Li Q, Wu P, Ye Y, Tseng H-Y, Zhang L, et al. Autophagy-mediated HMGB1 Release Antagonizes Apoptosis of Gastric Cancer Cells Induced by Vincristine via Transcriptional Regulation of Mcl-1. Autophagy, 2012, 8: 109–21., DOI 10.4161/auto.8.1.18319 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD MAR PY 2022 VL 28 IS 1 BP 1610293 EP 1610299 DI 10.3389/pore.2022.1610293 PG 7 ER PT J AU Chen, B Li, R Zhang, J Xu, L Jiang, F AF Chen, Bing Li, Rutao Zhang, Junling Xu, Lin Jiang, Feng TI Genomic Landscape of Metastatic Lymph Nodes and Primary Tumors in Non-Small-Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gene mutation; ARID1A; non-small-cell lung cancer; metastatic; CTNNB1 ID gene mutation; ARID1A; non-small-cell lung cancer; metastatic; CTNNB1 AB Objective: To investigate the genetic mutation characteristics of non-small cell lung cancers (NSCLC) with and without lymph node metastasis. Methods: Primary lesions and metastatic lymph node lesions of 36 Chinese NSCLC patients were tested for somatic mutations, tumor mutation burden, phylogenetic and clonal evolutional analysis using a 1021-gene panel by next-generation sequencing (NGS) with an average sequencing depth of 671X. Results: In this study, eighteen patients with lung adenocarcinoma (LUAD) and 18 with lung squamous cell carcinoma (LUSC) were included. Different groups had distinct characteristics of gene mutations. CTNNB1 gene mutations were only present in Nome_LC LUAD patients (p < 0.05). ARID1A mutation was however the only gene with significant alterations (p < 0.05) in Nome_LC in LUSC. Phylogenetic trees of mutated genes were also constructed. Linear and parallel evolutions of metastatic lymph nodes were observed both in LUAD and LUSC. Conclusion: LUSC exhibited more genetic mutations than LUAD. Intriguingly, there was significant difference in gene mutations between Meta_LC and Nome_LC. CTNNB1 gene alteration was the key mutation in LUAD that seems to promote proliferation of the tumor and then determine T stage. On the other hand, proliferation of the tumor was characterized by ARID1A missense mutation in LUSC, thus influencing the T stage as well. Lymph node metastasis could display both linear and parallel evolutionary characteristics in NSCLC. Different metastatic lymph nodes might have exactly the same or different mutated genes, underlining the heterogeneous genomic characteristics of these cancer types. C1 [Chen, Bing] Jiangsu Cancer Hospital, the Affiliated Hospital of Nanjing Medical UniversityNanjing, China. [Li, Rutao] Jiangsu Cancer Hospital, the Affiliated Hospital of Nanjing Medical UniversityNanjing, China. [Zhang, Junling] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Xu, Lin] Jiangsu Cancer Hospital, the Affiliated Hospital of Nanjing Medical UniversityNanjing, China. [Jiang, Feng] Jiangsu Cancer Hospital, the Affiliated Hospital of Nanjing Medical UniversityNanjing, China. RP Xu, L (reprint author), Jiangsu Cancer Hospital, the Affiliated Hospital of Nanjing Medical University, Nanjing, China. EM szl_xl@126.com CR Siegel RL,Miller KD, Jemal A. Cancer Statistics, 2020. CA Cancer J Clin, 2020, 70:7–30., DOI 10.3322/caac.21590 Howlader N, Noone AM, Krapcho M, Miller D, Bishop K, Kosary CL, et al. SEER Cancer Statistics Review, 1975-2014. Bethesda, MD: National Cancer Institute, 2017). Based on November 2016 SEER Data Submission, posted to the SEER web site. Xu S, Yang J, Xu S, Zhu Y, Zhang C, Liu L, et al. Lymphatic Vessel Density as a Prognostic Indicator in Asian NSCLC Patients: A Meta-Analysis. BMC Pulm Med, 2018, 18:128., DOI 10.1186/s12890-018-0702-9 Adachi Y, Nakamura H, Kitamura Y, Taniguchi Y, Araki K, Shomori K, et al. Lymphatic Vessel Density in Pulmonary Adenocarcinoma Immunohistochemically Evaluated with Anti-podoplanin or Anti-D2- 40 Antibody is Correlated with Lymphatic Invasion or Lymph Node Metastases. Pathol Int, 2007, 57:171–7., DOI 10.1111/j.1440-1827.2007. 02077.x Sobin LH, Compton CC TNM Seventh Edition: What’s New, What’s Changed. Cancer, 2010, 116:5336–9., DOI 10.1002/cncr.25537 Chen B, Wang X, Yu X, XiaWj., Zhao H, Li Xf., et al. Lymph Node Metastasis in Chinese Patients with Clinical T1 Non-small Cell Lung Cancer: A Multicenter Real-world Observational Study. Thorac Cancer, 2019, 10: 533–42., DOI 10.1111/1759-7714.12970 Cho S, Song IH, Yang HC, Kim K, Jheon S. Predictive Factors for Node Metastasis in Patients with Clinical Stage I Non-small Cell Lung Cancer. Ann Thorac Surg, 2013, 96:239–45., DOI 10.1016/j.athoracsur.2013.03.050 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data. Cancer Discov, 2012, 2:401–4., DOI 10.1158/2159- 8290.cd-12-0095 Zhang Y, Chang L, Yang Y, Fang W, Guan Y, Wu A, et al. Intratumor Heterogeneity Comparison Among Different Subtypes of Non-small-cell Lung Cancer through Multi-Region Tissue and Matched ctDNA Sequencing. Mol Cancer, 2019, 18:7., DOI 10.1186/s12943-019-0939-9 Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Mutational Landscape Determines Sensitivity to PD-1 Blockade in Non-small Cell Lung Cancer. Science, 2015, 348:124–8., DOI 10.1126/science.aaa1348 Hoshide R, Jandial R. The Role of the Neural Niche in Brain Metastasis. Clin Exp Metastasis, 2017, 34:369–76., DOI 10.1007/s10585-017-9857-7 Comen E, Norton L, Massague J. Clinical Implications of Cancer Self-Seeding. Nat Rev Clin Oncol, 2011, 8:369–77., DOI 10.1038/nrclinonc.2011.64 Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, et al. Mutational Landscape of Metastatic Cancer Revealed from Prospective Clinical Sequencing of 10,000 Patients. Nat Med, 2017, 23:703–13., DOI 10.1038/nm. 4333 Jamal-Hanjani M, Wilson GA, McGranahan N, Birkbak NJ, Watkins TBK, Veeriah S, et al. Tracking the Evolution of Non-small-cell Lung Cancer. N Engl J Med, 2017, 376:2109–21., DOI 10.1056/nejmoa1616288 Willert K, Jones KA. Wnt Signaling: Is the Party in the Nucleus? Genes Dev, 2006, 20:1394–404., DOI 10.1101/gad.1424006 Tenbaum SP, Ordonez-Moran P, Puig I, Chicote I, Arques O, Landolfi S, et al. β-Catenin Confers Resistance to PI3K and AKT Inhibitors and Subverts FOXO3a to Promote Metastasis in colon Cancer. Nat Med, 2012, 18: 892–901., DOI 10.1038/nm.2772 Wen J, Min X, Shen M, Hua Q, Han Y, Zhao L, et al. ACLY Facilitates colon Cancer Cell Metastasis by CTNNB1. J Exp Clin Cancer Res, 2019, 38:401., DOI 10.1186/s13046-019-1391-9 Tissier F, Cavard C, Groussin L, Perlemoine K, Fumey G, Hagnere A-M, et al. Mutations of β-Catenin in Adrenocortical Tumors: Activation of the Wnt Signaling Pathway is a Frequent Event in Both Benign and Malignant Adrenocortical Tumors. Cancer Res, 2005, 65:7622–7., DOI 10.1158/0008- 5472.can-05-0593 Durand J, Lampron A, Mazzuco TL, Chapman A, Bourdeau I. Characterization of Differential Gene Expression in Adrenocortical Tumors Harboring β-Catenin, CTNNB1, Mutations. J Clin Endocrinol Metab, 2011, 96:E1206–E1211., DOI 10.1210/jc.2010-2143 Umphlett M, Shea S, Tome-Garcia J, Zhang Y, Hormigo A, Fowkes M, et al. Widely Metastatic Glioblastoma with BRCA1 and ARID1A Mutations: A Case Report. BMC cancer, 2020, 20:47., DOI 10.1186/s12885-020-6540-1 Gibson WJ, Hoivik EA, Halle MK, Taylor-Weiner A, Cherniack AD, Berg A, et al. The Genomic Landscape and Evolution of Endometrial Carcinoma Progression and Abdominopelvic Metastasis. Nat Genet, 2016, 48:848–55., DOI 10.1038/ng.3602 Zhu YP, Sheng LL, Wu J, Yang M, Cheng XF, Wu NN, et al. Loss of ARID1A Expression is Associated with Poor Prognosis in Patients with Gastric Cancer. Hum Pathol, 2018, 78:28–35., DOI 10.1016/j.humpath.2018.04.003 Jiang T, Fang Z, Tang S, Cheng R, Li Y, Ren S, et al. Mutational Landscape and Evolutionary Pattern of Liver and Brain Metastasis in Lung Adenocarcinoma. J Thorac Oncol, 2021, 16:237–49., DOI 10.1016/j.jtho.2020.10.128 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610020 EP 1610029 DI 10.3389/pore.2022.1610020 PG 10 ER PT J TI Methodological Challenges of Digital PCR Detection of the Histone H3 K27M Somatic Variant in Cerebrospinal Fluid SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cerebrospinal fluid; liquid biopsy; dPCR; cell-free DNA; CNS tumor; diffuse midline glioma; H3 K27M ID cerebrospinal fluid; liquid biopsy; dPCR; cell-free DNA; CNS tumor; diffuse midline glioma; H3 K27M AB Cell-free DNA (cfDNA) in body fluids is invaluable for cancer diagnostics. Despite the impressive potential of liquid biopsies for the diagnostics of central nervous system (CNS) tumors, a number of challenges prevent introducing this approach into routine laboratory practice. In this study, we adopt a protocol for sensitive detection of the H3 K27M somatic variant in cerebrospinal fluid (CSF) by using digital polymerase chain reaction (dPCR). Optimization of the protocol was carried out stepwise, including preamplification of the H3 target region and adjustment of dPCR conditions. The optimized protocol allowed detection of the mutant allele starting from DNA quantities as low as 9 picograms. Analytical specificity was tested using a representative group of tumor tissue samples with known H3 K27M status, and no false-positive cases were detected. The protocol was applied to a series of CSF samples collected from patients with CNS tumors (n = 18) using two alternative dPCR platforms, QX200 Droplet Digital PCR system (Bio-Rad) and QIAcuity Digital PCR System (Qiagen). In three out of four CSF specimens collected from patients with H3 K27M-positive diffuse midline glioma, both platforms allowed detection of the mutant allele. The use of ventricular access for CSF collection appears preferential, as lumbar CSF samples may produce ambiguous results. All CSF samples collected from patients with H3 wild-type tumors were qualified as H3 K27M-negative. High agreement of the quantitative data obtained with the two platforms demonstrates universality of the approach. CR Ostrom QT, Gittleman H, Truitt G, Boscia A, Kruchko C, Barnholtz-Sloan JS CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2011-2015. Neuro Oncol, 2018, 20: iv1–iv86., DOI 10.1093/neuonc/noy131 Walker DA, Liu J, Kieran M, Jabado N, Picton S, Packer R, et al. A Multi- Disciplinary Consensus Statement Concerning Surgical Approaches to Low- Grade, High-Grade Astrocytomas and Diffuse Intrinsic Pontine Gliomas in Childhood, CPN Paris 2011, Using the Delphi Method. Neuro-Oncology, 2013, 15(4):462–8., DOI 10.1093/neuonc/nos330 Wu G, Broniscer A, McEachron TA, Lu C, Paugh BS, Becksfort J, et al. Somatic Histone H3 Alterations in Pediatric Diffuse Intrinsic Pontine Gliomas and Non-brainstem Glioblastomas. Nat Genet, 2012, 44(3):251–3., DOI 10.1038/ng. 1102 Zaytseva MA, Yasko LA, Papusha LI, Druy AE. Molecular Genetic Features of Pediatric Gliomas. Pediatr Hematology/Oncology Immunopathology, 2019, 18(4):109–17., In Russ.)., DOI 10.24287/1726-1708-2019-18-4-109-117 Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A Summary. Acta Neuropathol, 2016, 131(6):803–20., DOI 10.1007/s00401-016-1545-1 Nazarian J, Druy AE, Yasko LA, Papusha LI, Novichkova GA. Liquid Biopsy in Pediatric Brain Cancers: A Theragnostic Opportunity. Pediatr Hematology/ Oncology Immunopathology, 2018, 17(1):133–5., In Russ.)., DOI 10.24287/ 1726-1708-2018-17-1-133-135 Scarlotta M, Simsek C, Kim AK. Liquid Biopsy in Solid Malignancy. Genet Test Mol Biomarkers, 2019, 23(4):284–96., DOI 10.1089/gtmb.2018.0237 Kwapisz D. The First Liquid Biopsy Test Approved. Is it a New Era ofMutation Testing for Non-small Cell Lung Cancer? Ann Transl Med, 2017, 5(3):46., DOI 10.21037/atm.2017.01.32 Oxnard GR, Thress KS, Alden RS, Lawrance R, Paweletz CP, Cantarini M, et al. Association between Plasma Genotyping and Outcomes of Treatment with Osimertinib, AZD9291, in Advanced Non–small-cell Lung Cancer. J Clin Oncol, 2016, 34(28):3375–82., DOI 10.1200/jco.2016.66.7162 Tzanikou E, Markou A, Politaki E, Koutsopoulos A, Psyrri A, Mavroudis D, et al. PIK3CA Hotspot Mutations in Circulating Tumor Cells and Paired Circulating Tumor DNA in Breast Cancer: A Direct Comparison Study. Mol Oncol, 2019, 13(12):2515–30., DOI 10.1002/1878-0261.12540 De Mattos-Arruda L, Mayor R, Ng CKY, Weigelt B, Martinez-Ricarte F, Torrejon D, et al. Cerebrospinal Fluid-Derived Circulating Tumour DNA Better Represents the Genomic Alterations of Brain Tumours Than Plasma. Nat Commun, 2015, 6(1):8839., DOI 10.1038/ncomms9839 Miller AM, Shah RH, Pentsova EI, Pourmaleki M, Briggs S, Distefano N, et al. Tracking Tumour Evolution in Glioma through Liquid Biopsies of Cerebrospinal Fluid. Nature, 2019, 565(7741):654–8., DOI 10.1038/s41586- 019-0882-3 Pan C, Diplas BH, Chen X, Wu Y, Xiao X, Jiang L, et al. Molecular Profiling of Tumors of the Brainstem by Sequencing of CSF-Derived Circulating Tumor DNA. Acta Neuropathol, 2019, 137(2):297–306., DOI 10.1007/s00401-018- 1936-6 Martinez-Ricarte F, Mayor R, Martinez-Saez E, Rubio-Perez C, Pineda E, Cordero E, et al. Molecular Diagnosis of Diffuse Gliomas through Sequencing of Cell-free Circulating Tumor DNA from Cerebrospinal Fluid. Clin Cancer Res, 2018, 24(12):2812–9., DOI 10.1158/1078-0432.CCR-17-3800 Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: a Summary. Neuro Oncol, 2021, 23(8):1231–51., DOI 10.1093/neuonc/noab106 Stallard S, Savelieff MG, Wierzbicki K, Mullan B, Miklja Z, Bruzek A, et al. CSF H3F3A K27M Circulating Tumor DNA Copy Number Quantifies Tumor Growth and In Vitro Treatment Response. Acta Neuropathol Commun, 2018, 6(1):80., DOI 10.1186/s40478-018-0580-7 Watanabe K, Fukuhara T, Tsukita Y, Morita M, Suzuki A, Tanaka N, et al. EGFR Mutation Analysis of Circulating Tumor DNA Using an Improved PNA-LNA PCR Clamp Method. Can Respir J, 2016, 2016:1–7., DOI 10.1155/ 2016/5297329 Milbury CA, Zhong Q, Lin J, WilliamsM, Olson J, Link DR, et al. Determining Lower Limits of Detection of Digital PCR Assays for Cancer-Related Gene Mutations. Biomol Detect Quantif, 2014, 1(1):8–22., DOI 10.1016/j.bdq.2014. 08.001 Wang Y, Springer S, Zhang M, McMahon KW, Kinde I, Dobbyn L, et al. Detection of Tumor-Derived DNA in Cerebrospinal Fluid of Patients with Primary Tumors of the Brain and Spinal Cord. Proc Natl Acad Sci USA, 2015, 112(31):9704–9., DOI 10.1073/pnas.1511694112 Zhao Z, Zhang C, Li M, Shen Y, Feng S, Liu J, et al. Applications of Cerebrospinal Fluid Circulating Tumor DNA in the Diagnosis of Gliomas. Jpn J Clin Oncol, 2020, 50(3):325–32., DOI 10.1093/jjco/hyz156 Connolly ID, Li Y, Pan W, Johnson E, You L, Vogel H, et al. A Pilot Study on the Use of Cerebrospinal Fluid Cell-free DNA in Intramedullary Spinal Ependymoma. J Neurooncol, 2017, 135(1):29–36., DOI 10.1007/s11060-017- 2557-y Fujioka Y, Hata N, Akagi Y, Kuga D, Hatae R, Sangatsuda Y, et al. Molecular Diagnosis of Diffuse Glioma Using a Chip-Based Digital PCR System to Analyze IDH, TERT, and H3 Mutations in the Cerebrospinal Fluid. J Neurooncol, 2021, 152(1):47–54., DOI 10.1007/ s11060-020-03682-7 On J, Natsumeda M, Watanabe J, Saito S, Kanemaru Y, Abe H, et al. Low Detection Rate of H3K27M Mutations in Cerebrospinal Fluid Obtained from Lumbar Puncture in Newly Diagnosed Diffuse Midline Gliomas. Diagnostics, 2021, 11(4):681., DOI 10.3390/diagnostics11040681 Dong L, Meng Y, Sui Z, Wang J, Wu L, Fu B. Comparison of Four Digital PCR Platforms for Accurate Quantification of DNA Copy Number of a Certified Plasmid DNA ReferenceMaterial. Sci Rep, 2015, 5:13174., DOI 10. 1038/srep13174 Li D, Bonner ER, Wierzbicki K, Panditharatna E, Huang T, Lulla R, et al. Standardization of the Liquid Biopsy for Pediatric Diffuse Midline Glioma Using DdPCR. Sci Rep, 2021, 11(1):5098., DOI 10.1038/s41598-021- 84513-1 Jackson JB, Choi DS, Luketich JD, Pennathur A, Stahlberg A, Godfrey TE. Multiplex Preamplification of Serum DNA to Facilitate Reliable Detection of Extremely Rare Cancer Mutations in Circulating DNA by Digital PCR. J Mol Diagn, 2016, 18(2):235–43., DOI 10.1016/j.jmoldx.2015.10.004 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610024 EP 1610036 DI 10.3389/pore.2022.1610024 PG 13 ER PT J AU Zhang, P Chen, L Wu, Sh Ye, B Chen, Ch Shi, L AF Zhang, Peichen Chen, Liping Wu, Shengjie Ye, Bailiang Chen, Chao Shi, Lingyan TI Construction of a Metabolism-Related Long Non-Coding RNAs-Based Risk Score Model of Hepatocellular Carcinoma for Prognosis and Personalized Treatment Prediction SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; lncRNA; hepatocellular carcinoma; metabolism; therapeutic response ID prognosis; lncRNA; hepatocellular carcinoma; metabolism; therapeutic response AB Background: Long non-coding RNAs (lncRNAs) play a key regulatory role in tumor metabolism. Although hepatocellular carcinoma (HCC) is a metabolic disease, there have been few systematic reports on the association between lncRNA expression and metabolism in HCC. Results: In this study, we screened 557 metabolism-related lncRNAs in HCC. A risk score model based on 13 metabolism-related lncRNA pairs was constructed to predict the outcome and drug response in HCC. The risk score model presented a better prediction of the outcomes than that with common clinicopathological characteristics, such as tumor stage, grade, and status and aneuploidy score in both training and testing cohorts. In addition, patients in the high-risk group exhibited higher responses to gemcitabine and epothilone, whereas those in the low-risk group were more sensitive to metformin and nilotinib. Conclusion: The metabolism-related lncRNAs-based risk score model and the other findings of this study may be helpful for HCC prognosis and personalized treatment prediction. C1 [Zhang, Peichen] The First Affiliated Hospital of Wenzhou Medical University, Department of Gastrointestinal SurgeryWenzhou, China. [Chen, Liping] Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Department of PharmacyHangzhou, China. [Wu, Shengjie] Zhejiang University, School of Medicine, Sir Run Run Shaw Hospital, Department of PharmacyHangzhou, China. [Ye, Bailiang] The First Affiliated Hospital of Wenzhou Medical University, Department of GastroenterologyWenzhou, China. [Chen, Chao] The First Affiliated Hospital of Wenzhou Medical University, Department of GastroenterologyWenzhou, China. [Shi, Lingyan] The First Affiliated Hospital of Wenzhou Medical University, Department of GastroenterologyWenzhou, China. RP Shi, L (reprint author), The First Affiliated Hospital of Wenzhou Medical University, Department of Gastroenterology, Wenzhou, China. EM shilingyan@wmu.edu.cn CR Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, et al. Hepatocellular Carcinoma. Nat Rev Dis Primers, 2016, 2:16018., DOI 10. 1038/nrdp.2016.18 Villanueva A. Hepatocellular Carcinoma. N Engl J Med, 2019, 380:1450–62., DOI 10.1056/nejmra1713263 European Association For The Study Of The Liver. EASL-EORTC Clinical Practice Guidelines: Management of Hepatocellular Carcinoma. Eur J Cancer, 2012, 48:599–641., DOI 10.1016/j.ejca.2011.12.021 Ward PS, Thompson CB. Metabolic Reprogramming: a Cancer Hallmark Even Warburg Did Not Anticipate. Cancer Cell, 2012, 21:297–308., DOI 10.1016/j. ccr.2012.02.014 Pavlova NN, Thompson CB. The Emerging Hallmarks of Cancer Metabolism. Cel Metab, 2016, 23:27–47., DOI 10.1016/j.cmet.2015. 12.006 Vander Heiden MG, Cantley LC, Thompson CB. Understanding theWarburg Effect: the Metabolic Requirements of Cell Proliferation. Science, 2009, 324: 1029–33., DOI 10.1126/science.1160809 Brosnan CA, Voinnet O. The Long and the Short of Noncoding RNAs. Curr Opin Cel Biol, 2009, 21:416–25., DOI 10.1016/j.ceb.2009.04.001 Marques AC, Ponting CP. Catalogues of Mammalian Long Noncoding RNAs: Modest Conservation and Incompleteness. Genome Biol, 2009, 10:R124., DOI 10.1186/gb-2009-10-11-r124 Lin R,Maeda S, Liu C, Karin M, Edgington TS. A Large Noncoding RNA Is a Marker for Murine Hepatocellular Carcinomas and a Spectrum of Human Carcinomas. Oncogene, 2007, 26:851–8., DOI 10.1038/sj.onc. 1209846 Matouk IJ, Abbasi I, Hochberg A, Galun E, Dweik H, Akkawi M. Highly Upregulated in Liver Cancer Noncoding RNA Is Overexpressed in Hepatic Colorectal Metastasis. Eur J Gastroenterol Hepatol, 2009, 21:688–92., DOI 10. 1097/meg.0b013e328306a3a2 Gabory A, Ripoche M-A, Yoshimizu T, Dandolo L. The H19 Gene: Regulation and Function of a Non-coding RNA. Cytogenet Genome Res, 2006, 113: 188–93., DOI 10.1159/000090831 Tsang WP, Wong TWL, Cheung AHH, Co CNN, Kwok TT. Induction of Drug Resistance and Transformation in Human Cancer Cells by the Noncoding RNA CUDR. RNA, 2007, 13:890–8., DOI 10.1261/rna.359007 Prasanth KV, Prasanth SG, Xuan Z, Hearn S, Freier SM, Bennett CF, et al. Regulating Gene Expression through RNA Nuclear Retention. Cell, 2005, 123: 249–63., DOI 10.1016/j.cell.2005.08.033 Zhao X-Y, Lin JD. Long Noncoding RNAs: a New Regulatory Code in Metabolic Control. Trends Biochem Sci, 2015, 40:586–96., DOI 10.1016/j.tibs. 2015.08.002 Possemato R, Marks KM, Shaul YD, Pacold ME, Kim D, Birsoy K, et al. Functional Genomics Reveal that the Serine Synthesis Pathway Is Essential in Breast Cancer. Nature, 2011, 476:346–50., DOI 10.1038/ nature10350 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res, 2015, 43:e47., DOI 10.1093/nar/ gkv007 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: a Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci U.S.A, 2005, 102:15545–50., DOI 10.1073/pnas.0506580102 Mootha VK, Lindgren CM, Eriksson K-F, Subramanian A, Sihag S, Lehar J, et al. PGC-1α-responsive Genes Involved in Oxidative Phosphorylation Are Coordinately Downregulated in Human Diabetes. Nat Genet, 2003, 34: 267–73., DOI 10.1038/ng1180 Geeleher P, Cox N, Huang RS. pRRophetic: an R Package for Prediction of Clinical Chemotherapeutic Response from Tumor Gene Expression Levels. PLoS One, 2014, 9:e107468., DOI 10.1371/ journal.pone.0107468 Panzitt K, Tschernatsch MMO, Guelly C, Moustafa T, Stradner M, Strohmaier HM, et al. Characterization of HULC, a Novel Gene with Striking Up- Regulation in Hepatocellular Carcinoma, as Noncoding RNA. Gastroenterology, 2007, 132:330–42., DOI 10.1053/j.gastro.2006.08.026 Cui M, Xiao Z, Wang Y, Zheng M, Song T, Cai X, et al. Long Noncoding RNA HULC Modulates Abnormal Lipid Metabolism in Hepatoma Cells through an miR-9-Mediated RXRA Signaling Pathway. Cancer Res, 2015, 75:846–57., DOI 10.1158/0008-5472.can-14-1192 Liang Y, Zhang D, Zheng T, Yang G, Wang J, Meng F, et al. LncRNA-SOX2OT Promotes Hepatocellular Carcinoma Invasion and Metastasis through miR- 122-5p-Mediated Activation of PKM2. Oncogenesis, 2020, 9:54., DOI 10.1038/ s41389-020-0242-z Li X, Zhao Q, Qi J, Wang W, Zhang D, Li Z, et al. lncRNA Ftx Promotes Aerobic Glycolysis and Tumor Progression through the PPARγ Pathway in Hepatocellular Carcinoma. Int J Oncol, 2018, 53:551–66., DOI 10.3892/ijo.2018. 4418 Malakar P, Stein I, Saragovi A, Winkler R, Stern-Ginossar N, Berger M, et al. Long Noncoding RNA MALAT1 Regulates Cancer Glucose Metabolism by Enhancing mTOR-Mediated Translation of TCF7L2. Cancer Res, 2019, 79: 2480–93., DOI 10.1158/0008-5472.can-18-1432 Hu M, Fu Q, Jing C, Zhang X, Qin T, Pan Y. LncRNA HOTAIR Knockdown Inhibits Glycolysis by Regulating miR-130a-3p/HIF1A in Hepatocellular Carcinoma under Hypoxia. Biomed Pharmacother, 2020, 125:109703., DOI 10.1016/j.biopha.2019.109703 Zhao J, Song X, Xu T, Yang Q, Liu J, Jiang B, et al. Identification of Potential Prognostic Competing Triplets in High-Grade Serous Ovarian Cancer. Front Genet, 2021, 11:607722., DOI 10.3389/fgene.2020.607722 Ye J, Li H, Wei J, Luo Y, Liu H, Zhang J, et al. Risk Scoring System Based on lncRNA Expression for Predicting Survival in Hepatocellular Carcinoma with Cirrhosis. Asian Pac J Cancer Prev, 2020, 21:1787–95., DOI 10.31557/apjcp. 2020.21.6.1787 Jiang Y, Gou X, Wei Z, Tan J, Yu H, Zhou X, et al. Bioinformatics Profiling Integrating a Three Immune-Related Long Non-coding RNA Signature as a Prognostic Model for clear Cell Renal Cell Carcinoma. Cancer Cel Int, 2020, 20:166., DOI 10.1186/s12935-020-01242-7 Wu L, Wen Z, Song Y, Wang L. A Novel Autophagy-Related lncRNA Survival Model for Lung Adenocarcinoma. J Cel Mol Med, 2021, 25:5681–90., DOI 10. 1111/jcmm.16582 Luo H, Yang L, Liu C, Wang X, Dong Q, Liu L, et al. TMPO-AS1/miR-98- 5p/EBF1 Feedback Loop Contributes to the Progression of Bladder Cancer. Int J Biochem Cel Biol, 2020, 122:105702., DOI 10.1016/j.biocel. 2020.105702 Nicolaou KC, Ritzen A, Namoto K. Recent Developments in the Chemistry, Biology and Medicine of the Epothilones. Chem Commun, 2001, 2001: 1523–35., DOI 10.1039/b104949f Bollag DM, McQueney PA, Zhu J, Hensens O, Koupal L, Liesch J, et al. Epothilones, a New Class of Microtubule-Stabilizing Agents with a Taxol-like Mechanism of Action. Cancer Res, 1995, 55:2325–33. Mok TSK, Choi E, Yau D, Johri A, Yeo W, Chan ATC, et al. Effects of Patupilone, Epothilone B; EPO906), a Novel Chemotherapeutic Agent, in Hepatocellular Carcinoma: an In Vitro Study. Oncology, 2006, 71:292–6., DOI 10.1159/000106450 Yu H-C, Lin C-S, Tai W-T, Liu C-Y, Shiau C-W, Chen K-F. Nilotinib Induces Autophagy in Hepatocellular Carcinoma through AMPK Activation. J Biol Chem, 2013, 288:18249–59., DOI 10.1074/jbc.m112. 446385 Nayak A, Kapur A, Barroilhet L, Patankar M. Oxidative Phosphorylation: A Target for Novel Therapeutic Strategies against Ovarian Cancer. Cancers, 2018, 10:337., DOI 10.3390/cancers10090337 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610066 EP 1610075 DI 10.3389/pore.2022.1610066 PG 10 ER PT J AU Xia, H Deng, L Meng, Sh Liu, X Zheng, Ch AF Xia, Hailong Deng, Lei Meng, Shu Liu, Xipeng Zheng, Chao TI Single-Cell Transcriptome Profiling Signatures and Alterations of Microglia Associated With Glioblastoma Associate Microglia Contribution to Tumor Formation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE GBM; single-sell RNA-seq; microglia; tumor immune microenvironment; aged microglia ID GBM; single-sell RNA-seq; microglia; tumor immune microenvironment; aged microglia AB Glioblastoma (GBM), which occasionally occurs in pediatric patients, is the most common tumor of the central nervous system in adults. Clinically, GBM is classified as low-grade to high-grade (from 1 to 4) and is characterized by late discovery, limited effective treatment methods, and poor efficacy. With the development of immunotherapy technology, effective GBM treatment strategies are of great significance. The main immune cells found in the GBM tumor microenvironment are macrophages and microglia (MG). Both these monocytes play important roles in the occurrence and development of GBM. Macrophages are recruited during tumorigenesis, whereas MG is present in the brain during embryonic development. Interestingly, the accumulation of these monocytes is inversely proportional to the survival of adult GBM patients but not the pediatric GBM patients. This study used single-cell RNA-seq data to reveal the heterogeneity of MG in tumor lesions and to explore the role of different MG subtypes in the occurrence and development of GBM. The results may help find new targets for immunotherapy of GBM. C1 [Xia, Hailong] Chongqing Red Cross Hospital (Jiangbei District People’s Hospital), Department of NeurosurgeryChongqing, China. [Deng, Lei] Bishan District People’s Hospital, Department of NeurosurgeryChongqing, China. [Meng, Shu] Chongqing Red Cross Hospital (Jiangbei District People’s Hospital), Internal MedicineChongqing, China. [Liu, Xipeng] Chongqing Red Cross Hospital (Jiangbei District People’s Hospital), Department of NeurosurgeryChongqing, China. [Zheng, Chao] Chongqing Red Cross Hospital (Jiangbei District People’s Hospital), Department of NeurosurgeryChongqing, China. RP Zheng, Ch (reprint author), Chongqing Red Cross Hospital (Jiangbei District People’s Hospital), Department of Neurosurgery, Chongqing, China. EM lemanarc@dreamway-china.com CR Villa C, Miquel C, Mosses D, Bernier M, Di Stefano AL.The 2016 World Health Organization Classification of Tumours of the central Nervous System. Presse Med, 2018, 47(11-12):E187–E200., DOI 10.1016/j.lpm.2018.04.015 Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy Plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N Engl J Med, 2005, 352(10):987–96., DOI 10.1056/ nejmoa043330 Wen PY, Kesari S Malignant Gliomas in Adults. N Engl J Med, 2008, 359(17): 492–507., DOI 10.1056/NEJMra0708126 Mukherjee S, Baidoo J, Fried A, Atwi D, Dolai S, Boockvar J, et al. Curcumin Changes the Polarity of Tumor-Associated Microglia and Eliminates Glioblastoma. Int J Cancer, 2016, 139(12):2838–49., DOI 10.1002/ijc.30398 Simmons GW, Pong WW, Emnett RJ, White CR, Gianino SM, Rodriguez FJ, et al. Neurofibromatosis-1 Heterozygosity Increases Microglia in a Spatially and Temporally Restricted Pattern Relevant to Mouse Optic Glioma Formation and Growth. J Neuropathol Exp Neurol, 2011, 70(1):51–62., DOI 10.1097/nen.0b013e3182032d37 Gutmann DH, Kettenmann H Microglia/Brain Macrophages as Central Drivers of Brain Tumor Pathobiology. Neuron, 2019, 104(3):442–9., DOI 10. 1016/j.neuron.2019.08.028 Roesch S, Rapp C, Dettling S, Herold-Mende C When Immune Cells Turn Bad-Tumor-Associated Microglia/Macrophages in Glioma. Int J Mol Sci, 2018, 19(2), 436., DOI 10.3390/ijms19020436 Solga AC, Pong WW, Kim K-Y, Cimino PJ, Toonen JA, Walker J, et al. RNA Sequencing of Tumor-Associated Microglia Reveals Ccl5 as a Stromal Chemokine Critical for Neurofibromatosis-1 Glioma Growth. Neoplasia, 2015, 17(10):776–88., DOI 10.1016/j.neo.2015.10.002 Chen Z, Feng X, Herting CJ, Garcia VA, Nie K, Pong WW, et al. Cellular and Molecular Identity of Tumor-Associated Macrophages in Glioblastoma. Cancer Res, 2017, 77(9):2266–78., DOI 10.1158/0008-5472.can-16-2310 Guo X, Pan Y, Gutmann DH Genetic and Genomic Alterations Differentially Dictate Low-Grade Glioma Growth through Cancer Stem Cell-specific Chemokine Recruitment of T Cells and Microglia. Neuro Oncol, 2019, 21(10):1250–62., DOI 10.1093/neuonc/noz080 Hambardzumyan D, Gutmann DH, Kettenmann H The Role of Microglia and Macrophages in Glioma Maintenance and Progression. Nat Neurosci, 2016, 19(1):20–7., DOI 10.1038/nn.4185 Sorensen MD, Dahlrot RH, Boldt HB, Hansen S, Kristensen BW Tumourassociated Microglia/macrophages Predict Poor Prognosis in High-Grade Gliomas and Correlate with an Aggressive Tumour Subtype. Neuropathol Appl Neurobiol, 2018, 44(2):185–206., DOI 10.1111/nan.12428 Pyonteck SM, Akkari L, Schuhmacher AJ, Bowman RL, Sevenich L, Quail DF, et al. CSF-1R Inhibition Alters Macrophage Polarization and Blocks Glioma Progression. Nat Med, 2013, 19(10):1264–72., DOI 10.1038/nm.3337 Szulzewsky F, Pelz A, Feng X, Synowitz M, Markovic D, Langmann T, et al. Glioma-Associated Microglia/Macrophages Display an Expression Profile Different from M1 and M2 Polarization and Highly Express Gpnmb and Spp1. Plos One, 2015, 10(2):e0116644., DOI 10.1371/journal.pone.0116644 Daginakatte GC, Gutmann DH Neurofibromatosis-1, Nf1, Heterozygous Brain Microglia Elaborate Paracrine Factors that Promote Nf1-Deficient Astrocyte and Glioma Growth. Hum Mol Genet, 2007, 16(9):1098–112., DOI 10.1093/hmg/ddm059 Markovic DS, Vinnakota K, van Rooijen N, Kiwit J, Synowitz M, Glass R, et al. Minocycline Reduces Glioma Expansion and Invasion by Attenuating Microglial MT1-MMP Expression. Brain Behav Immun, 2011, 25(4):624–8., DOI 10.1016/j.bbi.2011.01.015 Hu F, Dzaye OD, Hahn A, Yu Y, Scavetta RJ, Dittmar G, et al. Glioma-derived Versican Promotes Tumor Expansion via Glioma-Associated Microglial/ macrophages Toll-like Receptor 2 Signaling. Neuro Oncol, 2015, 17(2): 200–10., DOI 10.1093/neuonc/nou324 Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM, et al. Comprehensive Integration of Single-Cell Data. Cell, 2019, 177(7):1888–902., DOI 10.1016/j.cell.2019.05.031 Aran D, Looney AP, Liu L, Wu E, Fong V, Hsu A, et al. Reference-based Analysis of Lung Single-Cell Sequencing Reveals a Transitional Profibrotic Macrophage. Nat Immunol, 2019, 20(2):163–72., DOI 10.1038/s41590-018- 0276-y Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, et al. The Dynamics and Regulators of Cell Fate Decisions are Revealed by Pseudotemporal Ordering of Single Cells. Nat Biotechnol, 2014, 32(4): 381–6., DOI 10.1038/nbt.2859 Love MI, Huber W, Anders S Moderated Estimation of Fold Change and Dispersion for RNA-Seq Data with DESeq2. Genome Biol, 2014, 15(12):550., DOI 10.1186/s13059-014-0550-8 Yu G, Wang L-G, Han Y, He Q-Y clusterProfiler: An R Package for Comparing Biological Themes Among Gene Clusters. OMICS: A J Integr Biol, 2012, 16(5): 284–7., DOI 10.1089/omi.2011.0118 Wainwright DA, Balyasnikova IV, Chang AL, Ahmed AU, Moon K-S, Auffinger B, et al. IDO Expression in Brain Tumors Increases the Recruitment of Regulatory T Cells and Negatively Impacts Survival. Clin Cancer Res, 2012, 18(22):6110–21., DOI 10.1158/1078-0432.ccr-12-2130 Bloch O, Crane CA, Kaur R, Safaee M, Rutkowski MJ, Parsa AT Gliomas Promote Immunosuppression through Induction of B7-H1 Expression in Tumor-Associated Macrophages. Clin Cancer Res, 2013, 19(12):3165–75., DOI 10.1158/1078-0432.ccr-12-3314 Yu JS, Wheeler CJ, Zeltzer PM, Ying H, Finger DN, Lee PK, et al. Vaccination of Malignant Glioma Patients with Peptide-Pulsed Dendritic Cells Elicits Systemic Cytotoxicity and Intracranial T-Cell Infiltration. Cancer Res, 2001, 61(3):842–7. Mukherjee S, Fried A, Hussaini R, White R, Baidoo J, Yalamanchi S, et al. Phytosomal Curcumin Causes Natural Killer Cell-dependent Repolarization of Glioblastoma, GBM, Tumor-Associated Microglia/macrophages and Elimination of GBM and GBM Stem Cells. J Exp Clin Cancer Res, 2018, 37:168., DOI 10.1186/s13046-018-0792-5 Shah S, Mostafa MM, McWhae A, Traves SL, Newton R Negative Feed- Forward Control of Tumor Necrosis Factor, TNF, by Tristetraprolin, ZFP36, is Limited by the Mitogen-Activated Protein Kinase Phosphatase, Dual- Specificity Phosphatase 1, DUSP1). J Biol Chem, 2016, 291(1):110–25., DOI 10.1074/jbc.m115.697599 Dranoff G Cytokines in Cancer Pathogenesis and Cancer Therapy. Nat Rev Cancer, 2004, 4(1):11–22., DOI 10.1038/nrc1252 Okada H, Lieberman FS, Walter KA, Lunsford LD, Kondziolka DS, Bejjani GK, et al. Autologous Glioma Cell Vaccine Admixed with Interleukin-4 Gene Transfected Fibroblasts in the Treatment of Patients with Malignant Gliomas. J Transl Med, 2007, 5:67., DOI 10.1186/1479-5876-5-67 Natsume A, Wakabayashi T, Ishii D, Maruta H, Fujii M, Shimato S, et al. A Combination of IFN-β and Temozolomide in Human Glioma Xenograft Models: Implication of P53-Mediated MGMT Downregulation. Cancer Chemother Pharmacol, 2008, 61(4):653–9., DOI 10.1007/s00280-007-0520-x Qian J, Luo F, Yang J, Liu J, Liu R, Wang L, et al. TLR2 Promotes Glioma Immune Evasion by Downregulating MHC Class II Molecules in Microglia. Cancer Immunol Res, 2018, 6(10):1220–33., DOI 10.1158/2326-6066.CIR-18- 0020 Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-T Immunotherapy for Castration-Resistant Prostate Cancer.NEngl J Med, 2010, 363(5):411–22., DOI 10.1056/nejmoa1001294 Pong WW, Higer SB, Gianino SM, Emnett RJ, Gutmann DH Reduced Microglial CX3CR1 Expression Delays Neurofibromatosis-1 Glioma Formation. Ann Neurol, 2013, 73(2):303–8., DOI 10.1002/ana.23813 Ntanasis-Stathopoulos I, Fotiou D, Terpos E CCL3 Signaling in the Tumor Microenvironment. Tumor Microenviron Role of Chemokines Pt A, 2020, 1231:13–21., DOI 10.1007/978-3-030-36667-4_2 Shaik S, Martin EC, Hayes DJ, Gimble JM, Devireddy RV Transcriptomic Profiling of Adipose Derived Stem Cells Undergoing Osteogenesis by RNA-Seq. Sci Rep, 2019, 9:11800., DOI 10.1038/s41598- 019-48089-1 Caracciolo V, Young J, Gonzales D, Ni Y, Flowers SJ, Summer R, et al. Myeloid-specific Deletion of Zfp36 Protects against Insulin Resistance and Fatty Liver in Diet-Induced Obese Mice. Am J Physiol Endocrinol Metab, 2018, 315(4):E676–E693., DOI 10.1152/ajpendo.00224.2017 Calcinotto A, Kohli J, Zagato E, Pellegrini L, Demaria M, Alimonti A Cellular Senescence: Aging, Cancer, and Injury. Physiol Rev, 2019, 99(2):1047–78., DOI 10.1152/physrev.00020.2018 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610067 EP 1610076 DI 10.3389/pore.2022.1610067 PG 10 ER PT J AU Egyed, B Horvath, A Semsei, Szalai, Cs Muller, J Erdelyi, D Kovacs, G AF Egyed, Balint Horvath, Anna Semsei, F. Agnes Szalai, F. Csaba Muller, Judit Erdelyi, J. Daniel Kovacs, T. Gabor TI Co-Detection of VEGF-A and Its Regulator, microRNA-181a, May Indicate Central Nervous System Involvement in Pediatric Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cerebrospinal fluid; biomarkers; liquid biopsy; central nervous system involvement; pediatric leukemia; enzyme-linked immunosorbent assay ID cerebrospinal fluid; biomarkers; liquid biopsy; central nervous system involvement; pediatric leukemia; enzyme-linked immunosorbent assay AB Central nervous system (CNS) involvement is a leading cause of therapy-refractory pediatric acute lymphoblastic leukemia (pALL), which is aggravated by underdiagnosing CNS disease with the currently used cell-based approach of cerebrospinal fluid (CSF) diagnostics. Our study focused on developing novel subcellular CNS leukemia indicators in the CSF and the bone marrow (BM) of patients with pALL. Serial liquid biopsy samples (n = 65) were analyzed by Elisas to measure the level of essential proteins associated with blast cell CNS trafficking, vascular endothelial growth factor A (VEGF-A) and integrin alpha 6 (ITGA6). In CSF samples from early induction chemotherapy, VEGF-A concentration were uniformly elevated in the CNS-positive group compared to those patients without unambiguous meningeal infiltration (9 vs Nine patients, Δc = 17.2 pg/ml, p = 0.016). Expression of miR-181a, a VEGFA-regulating microRNA which showed increased level in CNS leukemia in our previous experiments, was then paralleled with VEGF-A concentration. A slight correlation between the levels of miR-181a and VEGF-A indicators in CSF and BM samples was revealed (n = 46, Pearson’s r = 0.36, p = 0.015). After validating in international cohorts, the joint quantification of miR-181a and VEGF-A might provide a novel tool to precisely diagnose CNS involvement and adjust CNS-directed therapy in pALL. C1 [Egyed, Balint] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Horvath, Anna] Semmelweis University, Department of Genetics, Cell- and Immunobiology, Clinical Genomics Research GroupBudapest, Hungary. [Semsei, F. Agnes] Semmelweis University, Department of Genetics, Cell- and Immunobiology, Clinical Genomics Research GroupBudapest, Hungary. [Szalai, F. Csaba] Semmelweis University, Department of Genetics, Cell- and Immunobiology, Clinical Genomics Research GroupBudapest, Hungary. [Muller, Judit] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Erdelyi, J. Daniel] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. [Kovacs, T. Gabor] Semmelweis University, 2nd Department of PediatricsBudapest, Hungary. RP Kovacs, G (reprint author), Semmelweis University, 2nd Department of Pediatrics, Budapest, Hungary. EM kovacs.gabor1@med.semmelweis-univ.hu CR Force LM, Abdollahpour I, Advani SM, Agius D, Ahmadian E, Alahdab F, et al. The Global burden of Childhood and Adolescent Cancer in 2017: an Analysis of the Global Burden of Disease Study 2017. Lancet Oncol, 2019, 20(9): 1211–25., DOI 10.1016/S1470-2045(19)30339-0 Nguyen K, Devidas M, Devidas M, Cheng S-C, La M, Raetz EA, et al. Factors Influencing Survival after Relapse from Acute Lymphoblastic Leukemia: a Children’s Oncology Group Study. Leukemia, 2008, 22(12):2142–50., DOI 10. 1038/leu.2008.251 Martinez-Laperche C, Gomez-Garcia AM, Lassaletta A, Moscardo C, Vivanco JL,Molina J, et al. Detection of Occult Cerebrospinal Fluid Involvement during Maintenance Therapy Identifies a Group of Children with Acute Lymphoblastic Leukemia at High Risk for Relapse. Am J Hematol, 2013, 88(5):359–64., DOI 10.1002/ajh.23407 Crespo-Solis E, Lopez-Karpovitch X, Higuera J, Vega-Ramos B. Diagnosis of Acute Leukemia in Cerebrospinal Fluid, CSF-Acute Leukemia). Curr Oncol Rep, 2012, 14(5):369–78., DOI 10.1007/s11912-012-0248-6 Munch V, Trentin L, Herzig J, Demir S, Seyfried F, Kraus JM, et al. Central Nervous System Involvement in Acute Lymphoblastic Leukemia Is Mediated by Vascular Endothelial Growth Factor. Blood, 2017, 130(5):643–54., DOI 10. 1182/blood-2017-03-769315 Yao H, Price TT, Cantelli G, Ngo B, Warner MJ, Olivere L, et al. Leukaemia Hijacks a Neural Mechanism to Invade the central Nervous System. Nature, 2018, 560(7716):55–60., DOI 10.1038/s41586-018-0342-5 Egyed B, Kutszegi N, Sagi JC, Gezsi A, Rzepiel A, Visnovitz T, et al. MicroRNA-181a as Novel Liquid Biopsy Marker of central Nervous System Involvement in Pediatric Acute Lymphoblastic Leukemia. J Transl Med, 2020, 18(1):250., DOI 10.1186/s12967-020-02415-8 Sun W, Wang X, Li J, You C, Lu P, Feng H, et al. MicroRNA-181a Promotes Angiogenesis in Colorectal Cancer by Targeting SRCIN1 to Promote the SRC/ VEGF Signaling Pathway. Cell Death Dis, 2018, 9(4):438., DOI 10.1038/s41419- 018-0490-4 Kato I, Nishinaka Y, Nakamura M, Akarca AU, Niwa A, Ozawa H, et al. Hypoxic Adaptation of Leukemic Cells Infiltrating the CNS Affords a Therapeutic Strategy Targeting VEGFA. Blood, 2017, 129(23):3126–9., DOI 10.1182/BLOOD-2016-06-721712 Tang Y-T, Jiang F, Guo L, Si M-Y, Jiao X-Y. The Soluble VEGF Receptor 1 and 2 Expression in Cerebral Spinal Fluid as an Indicator for Leukemia central Nervous System Metastasis. J Neurooncol, 2013, 112(3):329–38., DOI 10.1007/ S11060-013-1066-X Bartram J, Goulden N, Wright G, Adams S, Brooks T, Edwards D, et al. High Throughput Sequencing in Acute Lymphoblastic Leukemia Reveals Clonal Architecture of central Nervous System and Bone Marrow Compartments. Haematologica, 2018, 103(3):e110–e114., DOI 10.3324/ haematol.2017.174987 Sun X, Wei L, Chen Q, Terek RM. MicroRNA Regulates Vascular Endothelial Growth Factor Expression in Chondrosarcoma Cells. Clin Orthop Relat Res, 2015, 473(3):907–13., DOI 10.1007/S11999-014-3842-0 Shah Scharff BFS, Modvig S, Thastrup M, Levinsen M, Degn M, Ryder LP, et al. A Comprehensive Clinical Study of Integrins in Acute Lymphoblastic Leukemia Indicates a Role of α6/CD49f in Persistent Minimal Residual Disease and α5 in the Colonization of Cerebrospinal Fluid. Leuk Lymphoma, 2020, 61(7):1714–8., DOI 10.1080/10428194.2020. 1731500 Yu X, Zhang H, Yuan M, Zhang P,Wang Y, Zheng M, et al. Identification and Characterization of a Murine Model of BCR-Abl1+ Acute B-Lymphoblastic Leukemia with central Nervous System Metastasis. Oncol Rep, 2019, 42(2): 532., DOI 10.3892/OR.2019.7184 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610096 EP 1610101 DI 10.3389/pore.2022.1610096 PG 6 ER PT J AU Boniface, Ch Spellman, P AF Boniface, T. Christopher Spellman, T. Paul TI Blood, Toil, and Taxoteres: Biological Determinates of Treatment-Induce ctDNA Dynamics for Interpreting Tumor Response SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarkers; liquid biopsy; circulating tumor DNA; ctDNA; tumor growth; treatment response ID biomarkers; liquid biopsy; circulating tumor DNA; ctDNA; tumor growth; treatment response AB Collection and analysis of circulating tumor DNA (ctDNA) is one of the few methods of liquid biopsy that measures generalizable and tumor specific molecules, and is one of the most promising approaches in assessing the effectiveness of cancer care. Clinical assays that utilize ctDNA are commercially available for the identification of actionable mutations prior to treatment and to assess minimal residual disease after treatment. There is currently no clinical ctDNA assay specifically intended to monitor disease response during treatment, partially due to the complex challenge of understanding the biological sources of ctDNA and the underlying principles that govern its release. Although studies have shown preand post-treatment ctDNA levels can be prognostic, there is evidence that early, ontreatment changes in ctDNA levels are more accurate in predicting response. Yet, these results also vary widely among cohorts, cancer type, and treatment, likely due to the driving biology of tumor cell proliferation, cell death, and ctDNA clearance kinetics. To realize the full potential of ctDNA monitoring in cancer care, we may need to reorient our thinking toward the fundamental biological underpinnings of ctDNA release and dissemination from merely seeking convenient clinical correlates. C1 [Boniface, T. Christopher] Oregon Health & Science University, Knight Cancer InstitutePortland, OR, USA. [Spellman, T. Paul] Oregon Health & Science University, Knight Cancer InstitutePortland, OR, USA. RP Boniface, Ch (reprint author), Oregon Health & Science University, Knight Cancer Institute, Portland, USA. EM boniface@ohsu.edu CR Wan JCM, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, et al. Liquid Biopsies Come of Age: towards Implementation of Circulating Tumour DNA. Nat Rev Cancer, 2017, 17(4):223–38., DOI 10.1038/nrc.2017.7 Heitzer E, Auinger L, Speicher MR. Cell-Free DNA and Apoptosis: How Dead Cells Inform about the Living. Trends Mol Med, 2020, 26(5):519–28., DOI 10.1016/j.molmed.2020.01.012 Merker JD, Oxnard GR, Compton C, Diehn M, Hurley P, Lazar AJ, et al. Circulating Tumor DNA Analysis in Patients with Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J Clin Oncol, 2018, 36(16):1631–41., DOI 10.1200/jco.2017.76.8671 Abbosh C, Birkbak NJ,Wilson GA, Jamal-HanjaniM, Constantin T, Salari R, et al. Phylogenetic ctDNA Analysis Depicts Early-Stage Lung Cancer Evolution. Nature, 2017, 545(7655):446–51., DOI 10.1038/nature22364 Keller L, Belloum Y, Wikman H, Pantel K. Clinical Relevance of Blood-Based ctDNA Analysis: Mutation Detection and beyond. Br J Cancer, 2021, 124(2): 345–58., DOI 10.1038/s41416-020-01047-5 Adashek JJ, Janku F, Kurzrock R. Signed in Blood: Circulating Tumor DNA in Cancer Diagnosis, Treatment and Screening. Cancers, Basel,, 2021, 13(14), 3600., DOI 10.3390/cancers13143600 De Michino S, Aparnathi M, Rostami A, Lok BH, Bratman SV. The Utility of Liquid Biopsies in Radiation Oncology. Int J Radiat Oncol Biol Phys, 2020, 107(5):873–86., DOI 10.1016/j.ijrobp.2020.05.008 Khier S, Lohan L. Kinetics of Circulating Cell–free DNA for Biomedical Applications: Critical Appraisal of the Literature. Future Sci OA, 2018, 4(4): FSO295., DOI 10.4155/fsoa-2017-0140 Butler TM, Boniface CT, Johnson-Camacho K, Tabatabaei S, Melendez D, Kelley T, et al. Circulating Tumor DNA Dynamics Using Patient-Customized Assays are Associated with Outcome in Neoadjuvantly Treated Breast Cancer. Cold Spring Harb Mol Case Stud, 2019, 5(2):a003772., DOI 10. 1101/mcs.a003772 Boniface C, Deig C, Halsey C, Kelley T, Heskett MB, Thomas CR Jr, et al. The Feasibility of Patient-specific Circulating Tumor DNA Monitoring throughout Multi-Modality Therapy for Locally Advanced Esophageal and Rectal Cancer: A Potential Biomarker for Early Detection of Subclinical Disease. Diagnostics, Basel,, 2021, 11(1):73., DOI 10.3390/ diagnostics11010073 Magbanua MJM, Swigart LB, Wu H-T, Hirst GL, Yau C, Wolf DM, et al. Circulating Tumor DNA in Neoadjuvant-Treated Breast Cancer Reflects Response and Survival. Ann Oncol, 2021, 32(2):229–39., DOI 10.1016/j. annonc.2020.11.007 Morbelli S, Alama A, Ferrarazzo G, Coco S, Genova C, Rijavec E, et al. Circulating Tumor DNA Reflects Tumor Metabolism rather Than Tumor Burden in Chemotherapy-Naive Patients with Advanced Non-small Cell Lung Cancer: 18F-FDG PET/CT Study. J Nucl Med, 2017, 58(11):1764–9., DOI 10.2967/jnumed.117.193201 Garcia-Olmo DC, Samos J, Picazo MG, Asensio AI, Toboso I, Garcia-Olmo D. Release of Cell-free DNA into the Bloodstream Leads to High Levels of Non-tumor Plasma DNA during Tumor Progression in Rats. Cancer Lett, 2008, 272(1):133–40., DOI 10.1016/j.canlet.2008.07.003 Xi L, Pham TH-T, Payabyab EC, Sherry RM, Rosenberg SA, Raffeld M. Circulating Tumor DNA as an Early Indicator of Response to T-Cell Transfer Immunotherapy in Metastatic Melanoma. Clin Cancer Res, 2016, 22(22): 5480–6., DOI 10.1158/1078-0432.ccr-16-0613 Lo YM, Leung SF, Chan LY, Chan AT, Lo KW, Johnson PJ, et al. Kinetics of Plasma Epstein-Barr Virus DNA during Radiation Therapy for Nasopharyngeal Carcinoma. Cancer Res, 2000, 60(9):2351–5. Hilke FJ, Muyas F, Admard J, Kootz B, Nann D, Welz S, et al. Dynamics of Cell-free Tumour DNA Correlate with Treatment Response of Head and Neck Cancer Patients Receiving Radiochemotherapy. Radiother Oncol, 2020, 151:182–9., DOI 10.1016/j.radonc.2020.07.027 Zhang Q, Luo J, Wu S, Si H, Gao C, Xu W, et al. Prognostic and Predictive Impact of Circulating Tumor DNA in Patients with Advanced Cancers Treated with Immune Checkpoint Blockade. Cancer Discov, 2020, 10(12): 1842–53., DOI 10.1158/2159-8290.cd-20-0047 Song Y, Hu C, Hu C, Xie Z, Wu L, Zhu Z, et al. Circulating Tumor DNA Clearance Predicts Prognosis across Treatment Regimen in a Large Real- World Longitudinally Monitored Advanced Non-small Cell Lung Cancer Cohort. Transl Lung Cancer Res, 2020, 9(2):269–79., DOI 10.21037/tlcr.2020. 03.17 Wang Y, Yang L, Bao H, Fan X, Xia F, Wan J, et al. Utility of ctDNA in Predicting Response to Neoadjuvant Chemoradiotherapy and Prognosis Assessment in Locally Advanced Rectal Cancer: A Prospective Cohort Study. Plos Med, 2021, 18(8):e1003741., DOI 10.1371/journal.pmed. 1003741 Anagnostou V, Forde PM, White JR, Niknafs N, Hruban C, Naidoo J, et al. Dynamics of Tumor and Immune Responses during Immune Checkpoint Blockade in Non-small Cell Lung Cancer. Cancer Res, 2019, 79(6):1214–25., DOI 10.1158/0008-5472.can-18-1127 Adalsteinsson VA, Ha G, Freeman SS, Choudhury AD, Stover DG, Parsons HA, et al. Scalable Whole-Exome Sequencing of Cell-free DNA Reveals High Concordance with Metastatic Tumors. Nat Commun, 2017, 8(1):1324., DOI 10.1038/s41467-017-00965-y Xia S, Ye J, Chen Y, Lizaso A, Huang L, Shi L, et al. Parallel Serial Assessment of Somatic Mutation and Methylation Profile from Circulating Tumor DNA Predicts Treatment Response and Impending Disease Progression in Osimertinib-Treated Lung Adenocarcinoma Patients. Transl Lung Cancer Res, 2019, 8(6):1016–28., DOI 10.21037/tlcr.2019.12.09 Silva R, Moran B, Baird A-M, O’Rourke CJ, Finn SP, McDermott R, et al. Longitudinal Analysis of Individual cfDNA Methylome Patterns in Metastatic Prostate Cancer. Clin Epigenet, 2021, 13(1):168., DOI 10.1186/ s13148-021-01155-w Aucamp J, Bronkhorst AJ, Badenhorst CPS, Pretorius PJ. The Diverse Origins of Circulating Cell-free DNA in the Human Body: A Critical Reevaluation of the Literature. Biol Rev, 2018, 93(3):1649–83., DOI 10.1111/ brv.12413 Gold B, Cankovic M, Furtado LV, Meier F, Gocke CD. Do circulating Tumor Cells, Exosomes, and Circulating Tumor Nucleic Acids Have Clinical Utility? A Report of the Association for Molecular Pathology. J Mol Diagn, 2015, 17(3):209–24., DOI 10.1016/j.jmoldx.2015.02.001 Grabuschnig S, Bronkhorst AJ, Holdenrieder S, Rosales Rodriguez I, Schliep KP, Schwendenwein D, et al. Putative Origins of Cell-free DNA in Humans: A Review of Active and Passive Nucleic Acid Release Mechanisms. Int J Mol Sci, 2020, 21(21):8062., DOI 10.3390/ijms21218062 Kustanovich A, Schwartz R, Peretz T, Grinshpun A. Life and Death of Circulating Cell-free DNA. Cancer Biol Ther, 2019, 20(8):1057–67., DOI 10. 1080/15384047.2019.1598759 de Miranda FS, Barauna VG, Dos Santos L, Costa G, Vassallo PF, Campos LCG. Properties and Application of Cell-free DNA as a Clinical Biomarker. Int J Mol Sci, 2021, 22(17):9110., DOI 10.3390/ijms22179110 Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of Circulating Tumor DNA in Early- and Late-Stage Human Malignancies. Sci Transl Med, 2014, 6(224):224ra24., DOI 10.1126/ scitranslmed.3007094 Phallen J, Sausen M, Adleff V, Leal A, Hruban C, White J, et al. Direct Detection of Early-Stage Cancers Using Circulating Tumor DNA. Sci Transl Med, 2017, 9(403):eaan2415., DOI 10.1126/scitranslmed.aan2415 Hojbjerg JA, Madsen AT, Schmidt HH, Sorensen SF, Stougaard M, Meldgaard P, et al. Intra-individual Variation of Circulating Tumour DNA in Lung Cancer Patients. Mol Oncol, 2019, 13(10):2098–106., DOI 10. 1002/1878-0261.12546 Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, et al. Circulating Mutant DNA to Assess Tumor Dynamics. Nat Med, 2008, 14(9):985–90., DOI 10.1038/nm.1789 Cheng ML, Pectasides E, Hanna GJ, Parsons HA, Choudhury AD, Oxnard GR. Circulating Tumor DNA in Advanced Solid Tumors: Clinical Relevance and Future Directions. CA Cancer J Clin, 2021, 71(2):176–90., DOI 10.3322/ caac.21650 Aucamp J, Bronkhorst AJ, Peters DL, Van Dyk HC, Van der Westhuizen FH, Pretorius PJ. Kinetic Analysis, Size Profiling, and Bioenergetic Association of DNA Released by Selected Cell Lines In Vitro. Cell. Mol. Life Sci., 2017, 74(14):2689–707., DOI 10.1007/s00018-017-2495-z Wang W, Kong P, Ma G, Li L, Zhu J, Xia T, et al. Characterization of the Release and Biological Significance of Cell-free DNA from Breast Cancer Cell Lines. Oncotarget, 2017, 8(26):43180–91., DOI 10.18632/oncotarget.17858 Rostami A, Lambie M, Yu CW, Stambolic V, Waldron JN, Bratman SV. Senescence, Necrosis, and Apoptosis Govern Circulating Cell-free DNA Release Kinetics. Cel Rep, 2020, 31(13):107830., DOI 10.1016/j.celrep.2020. 107830 Otandault A, Abraham J-D, Al Amir Dache Z, Khalyfa A, Jariel-Encontre I, Forne T, et al. Hypoxia Differently Modulates the Release of Mitochondrial and Nuclear DNA. Br J Cancer, 2020, 122(5):715–25., DOI 10.1038/s41416- 019-0716-y Bronkhorst AJ, Wentzel JF, Aucamp J, van Dyk E, du Plessis L, Pretorius PJ. Characterization of the Cell-free DNA Released by Cultured Cancer Cells. Biochim Biophys Acta Mol Cel Res, 2016, 1863(1):157–65., DOI 10.1016/j. bbamcr.2015.10.022 Bronkhorst AJ, Wentzel JF, Ungerer V, Peters DL, Aucamp J, de Villiers EP, et al. Sequence Analysis of Cell-free DNA Derived from Cultured Human Bone Osteosarcoma, 143B, Cells. Tumour Biol, 2018, 40(9): 1010428318801190., DOI 10.1177/1010428318801190 Hu Z-Y, Xie N, Tian C, Yang X, Liu L, Li J, et al. Identifying Circulating Tumor DNA Mutation Profiles in Metastatic Breast Cancer Patients with Multiline Resistance. EBioMedicine, 2018, 32:111–8., DOI 10.1016/j.ebiom. 2018.05.015 Zhou Y, Xu Y, Gong Y, Zhang Y, Lu Y, Wang C, et al. Clinical Factors Associated with Circulating Tumor DNA, Ct DNA , in Primary Breast Cancer. Mol Oncol, 2019, 13(5):1033–46., DOI 10.1002/1878-0261.12456 Ortolan E, Appierto V, Silvestri M, Miceli R, Veneroni S, Folli S, et al. Bloodbased Genomics of Triple-Negative Breast Cancer Progression in Patients Treated with Neoadjuvant Chemotherapy. ESMO Open, 2021, 6(2):100086., DOI 10.1016/j.esmoop.2021.100086 Abbosh C, Birkbak NJ, Wilson GA, Jamal-Hanjani M, Constantin T, Salari R, et al. Corrigendum: Phylogenetic ctDNA Analysis Depicts Early- Stage Lung Cancer Evolution. Nature, 2018, 554(7691):264., DOI 10.1038/ nature25161 Fornvik D, Aaltonen KE, Chen Y, George AM, Brueffer C, Rigo R, et al. Detection of Circulating Tumor Cells and Circulating Tumor DNA before and after Mammographic Breast Compression in a Cohort of Breast Cancer Patients Scheduled for Neoadjuvant Treatment. Breast Cancer Res Treat, 2019, 177(2):447–55., DOI 10.1007/s10549-019- 05326-5 Chabon JJ, Hamilton EG, Kurtz DM, Esfahani MS, Moding EJ, Stehr H, et al. Integrating Genomic Features for Non-invasive Early Lung Cancer Detection. Nature, 2020, 580(7802):245–51., DOI 10.1038/s41586-020-2140-0 Chen K-Z, Lou F, Yang F, Zhang J-B, Ye H, Chen W, et al. Circulating Tumor DNA Detection in Early-Stage Non-small Cell Lung Cancer Patients by Targeted Sequencing. Sci Rep, 2016, 6:31985., DOI 10.1038/srep31985 Cho M-S, Park CH, Lee S, Park HS. Clinicopathological Parameters for Circulating Tumor DNA Shedding in Surgically Resected Non-small Cell Lung Cancer with EGFR or KRAS Mutation. PLoS One, 2020, 15(3): e0230622., DOI 10.1371/journal.pone.0230622 Avanzini S, Kurtz DM, Chabon JJ, Moding EJ, Hori SS, Gambhir SS, et al. A Mathematical Model of ctDNA Shedding Predicts Tumor Detection Size. Sci Adv, 2020, 6(50):eabc4308., DOI 10.1126/sciadv.abc4308 McEvoy AC, Warburton L, Al-Ogaili Z, Celliers L, Calapre L, Pereira MR, et al. Correlation between Circulating Tumour DNA and Metabolic Tumour burden in Metastatic Melanoma Patients. BMC Cancer, 2018, 18(1):726., DOI 10.1186/s12885-018-4637-6 Chen Z, Fadiel A, Naftolin F, Eichenbaum KD, Xia Y. Circulation DNA: Biological Implications for Cancer Metastasis and Immunology. Med Hypotheses, 2005, 65(5):956–61., DOI 10.1016/j.mehy.2005.04.042 Ungerer V, Bronkhorst AJ, Van den Ackerveken P, Herzog M, Holdenrieder S. Serial Profiling of Cell-free DNA and Nucleosome Histone Modifications in Cell Cultures. Sci Rep, 2021, 11(1):9460., DOI 10.1038/s41598-021-88866-5 Fleischhacker M, Schmidt B. Circulating Nucleic Acids, CNAs, and Cancer- A Survey. Biochim Biophys Acta Rev Cancer, 2007, 1775(1):181–232., DOI 10. 1016/j.bbcan.2006.10.001 Choi JJ, Reich CF, 3rd, Pisetsky DS. Release of DNA from Dead and Dying Lymphocyte andMonocyte Cell Lines In Vitro. Scand J Immunol, 2004, 60(1- 2):159–66., DOI 10.1111/j.0300-9475.2004.01470.x Utani K-i., Okamoto A, Shimizu N. Generation of Micronuclei during Interphase by Coupling between Cytoplasmic Membrane Blebbing and Nuclear Budding. PLoS One, 2011, 6(11):e27233., DOI 10.1371/journal. pone.0027233 Kisurina-Evgenieva OP, Sutiagina OI, Onishchenko GE. Biogenesis of Micronuclei. Biochem Mosc, 2016, 81(5):453–64., DOI 10.1134/ s0006297916050035 Ryoo HD, Bergmann A. The Role of Apoptosis-Induced Proliferation for Regeneration and Cancer. Cold Spring Harbor Perspect Biol, 2012, 4(8): a008797., DOI 10.1101/cshperspect.a008797 Lugano R, Ramachandran M, Dimberg A. Tumor Angiogenesis: Causes, Consequences, Challenges and Opportunities. Cel Mol. Life Sci., 2020, 77(9): 1745–70., DOI 10.1007/s00018-019-03351-7 Wang J, Huang A, Wang Y-P, Yin Y, Fu P-Y, Zhang X, et al. Circulating Tumor DNA Correlates with Microvascular Invasion and Predicts Tumor Recurrence of Hepatocellular Carcinoma. Ann Transl Med, 2020, 8(5):237., DOI 10.21037/atm.2019.12.154 Shirasu H, Taniguchi H, Watanabe J, Kotaka M, Yamazaki K, Hirata K, et al. O-11 Monitoring Molecular Residual Disease by Circulating Tumor DNA in Resectable Colorectal Cancer: Molecular Subgroup Analyses of a Prospective Observational Study GALAXY in CIRCULATE-Japan. Ann Oncol, 2021, 32:S222–S223., DOI 10.1016/j. annonc.2021.05.015 Nabavizadeh SA, Ware JB, Guiry S, Nasrallah MP, Mays JJ, Till JE, et al. Imaging and Histopathologic Correlates of Plasma Cell-free DNA Concentration and Circulating Tumor DNA in Adult Patients with Newly Diagnosed Glioblastoma. Neurooncol Adv, 2020, 2(1):vdaa016., DOI 10.1093/ noajnl/vdaa016 Bagley SJ, Nabavizadeh SA, Mays JJ, Till JE, Ware JB, Levy S, et al. Clinical Utility of Plasma Cell-free DNA in Adult Patients with Newly Diagnosed Glioblastoma: A Pilot Prospective Study. Clin Cancer Res, 2020, 26(2): 397–407., DOI 10.1158/1078-0432.ccr-19-2533 Mair R, Mouliere F, Smith CG, Chandrananda D, Gale D, Marass F, et al. Measurement of Plasma Cell-free Mitochondrial Tumor DNA Improves Detection of Glioblastoma in Patient-Derived Orthotopic Xenograft Models. Cancer Res, 2019, 79(1):220–30., DOI 10.1158/0008-5472.can-18-0074 McKeown SR. Defining Normoxia, Physoxia and Hypoxia in Tumours- Implications for Treatment Response. Br J Radiol, 2014, 87(1035): 20130676., DOI 10.1259/bjr.20130676 Harris AL. Hypoxia - A Key Regulatory Factor in Tumour Growth. Nat Rev Cancer, 2002, 2(1):38–47., DOI 10.1038/nrc704 Giacona MB, Ruben GC, Iczkowski KA, Roos TB, Porter DM, Sorenson GD. Cell-free DNA in Human Blood Plasma: Length Measurements in Patients with Pancreatic Cancer and Healthy Controls. Pancreas, 1998, 17(1):89–97., DOI 10.1097/00006676-199807000-00012 Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al. DNA Fragments in the Blood Plasma of Cancer Patients: Quantitations and Evidence for Their Origin from Apoptotic and Necrotic Cells. Cancer Res, 2001, 61(4):1659–65. Cortese R, Almendros I, Wang Y, Gozal D. Tumor Circulating DNA Profiling in Xenografted Mice Exposed to Intermittent Hypoxia. Oncotarget, 2015, 6(1):556–69., DOI 10.18632/oncotarget.2785 Lindberg HK, Wang X, Jarventaus H, Falck GC, Norppa H, Fenech M. Origin of Nuclear Buds and Micronuclei in normal and Folate-Deprived Human Lymphocytes. Mutat Res, 2007, 617(1-2):33–45., DOI 10.1016/j.mrfmmm. 2006.12.002 Al-Asmakh M, Hedin L. Microbiota and the Control of Blood-Tissue Barriers. Tissue Barriers, 2015, 3(3):e1039691., DOI 10.1080/21688370.2015. 1039691 Lo YMD, Tein MSC, Lau TK, Haines CJ, Leung TN, Poon PMK, et al. Quantitative Analysis of Fetal DNA in Maternal Plasma and Serum: Implications for Noninvasive Prenatal Diagnosis. Am J Hum Genet, 1998, 62(4):768–75., DOI 10.1086/301800 Zhu L, Nazeri A, Pacia CP, Yue Y, Chen H. Focused Ultrasound for Safe and Effective Release of Brain Tumor Biomarkers into the Peripheral Circulation. PLoS One, 2020, 15(6):e0234182., DOI 10.1371/journal.pone.0234182 Pacia CP, Zhu L, Yang Y, Yue Y, Nazeri A, Michael Gach H, et al. Feasibility and Safety of Focused Ultrasound-Enabled Liquid Biopsy in the Brain of a Porcine Model. Sci Rep, 2020, 10(1):7449., DOI 10.1038/s41598-020-64440-3 Choi J-J, Reich CF, 3rd, Pisetsky DS. The Role of Macrophages in the In Vitro Generation of Extracellular DNA from Apoptotic and Necrotic Cells. Immunology, 2005, 115(1):55–62., DOI 10.1111/j.1365-2567.2005.02130.x Jiang N, Reich CF, 3rd, Pisetsky DS. Role of Macrophages in the Generation of Circulating Blood Nucleosomes from Dead and Dying Cells. Blood, 2003, 102(6):2243–50., DOI 10.1182/blood-2002-10-3312 Herbreteau G, Vallee A, Knol AC, Theoleyre S, Quereux G, Varey E, et al. Circulating Tumor DNA Early Kinetics Predict Response of Metastatic Melanoma to Anti-PD1 Immunotherapy: Validation Study. Cancers, Basel,, 2021, 13(8):1826., DOI 10.3390/cancers13081826 Westman J, Grinstein S, Marques PE. Phagocytosis of Necrotic Debris at Sites of Injury and Inflammation. Front Immunol, 2019, 10:3030., DOI 10.3389/ fimmu.2019.03030 Lo YMD, Zhang J, Leung TN, Lau TK, Chang AMZ, Hjelm NM. Rapid Clearance of Fetal DNA from Maternal Plasma. Am J Hum Genet, 1999, 64(1):218–24., DOI 10.1086/302205 Patel PS, Patel BP, Rawal RM, Raval GN, Patel MM, Patel JB, et al. Evaluation of Serum Alkaline DNase Activity in Treatment Monitoring of Head and Neck Cancer Patients. Tumor Biol, 2000, 21(2):82–9., DOI 10. 1159/000030113 Martin M, Leffler J, Smolag KI, Mytych J, Bjork A, Chaves LD, et al. Factor H Uptake Regulates Intracellular C3 Activation during Apoptosis and Decreases the Inflammatory Potential of Nucleosomes. Cell Death Differ, 2016, 23(5):903–11., DOI 10.1038/cdd.2015.164 Kirsch C, Weickmann S, Schmidt B, Fleischhacker M. An Improved Method for the Isolation of Free-Circulating Plasma DNA and Cell-free DNA from Other Body Fluids. Ann N Y Acad Sci, 2008, 1137:135–9., DOI 10.1196/annals. 1448.035 Gauthier VJ, Tyler LN, Mannik M. Blood Clearance Kinetics and Liver Uptake of Mononucleosomes in Mice. J Immunol, 1996, 156(3):1151–6. Korabecna M, Opatrna S,Wirth J, Rulcova K, Eiselt J, Sefrna F, et al. Cell-free Plasma DNA during Peritoneal Dialysis and Hemodialysis and in Patients with Chronic Kidney Disease. Ann N Y Acad Sci, 2008, 1137:296–301., DOI 10. 1196/annals.1448.014 Cheng THT, Jiang P, Tam JCW, Sun X, Lee W-S, Yu SCY, et al. Genomewide Bisulfite Sequencing Reveals the Origin and Time-dependent Fragmentation of Urinary cfDNA. Clin Biochem, 2017, 50(9):496–501., DOI 10.1016/j. clinbiochem.2017.02.017 Hung ECW, Shing TKF, Chim SSC, Yeung PC, Chan RWY, Chik KW, et al. Presence of Donor-Derived DNA and Cells in the Urine of Sex-Mismatched Hematopoietic Stem Cell Transplant Recipients: Implication for the Transrenal Hypothesis. Clin Chem, 2009, 55(4):715–22., DOI 10.1373/ clinchem.2008.113530 Costa DB, Halmos B, Kumar A, Schumer ST,Huberman MS, Boggon TJ, et al. BIM Mediates EGFR Tyrosine Kinase Inhibitor-Induced Apoptosis in Lung Cancers with Oncogenic EGFR Mutations. Plos Med, 2007, 4(10):1669–80., DOI 10.1371/journal.pmed.0040315 Phallen J, Leal A, Woodward BD, Forde PM, Naidoo J, Marrone KA, et al. Early Noninvasive Detection of Response to Targeted Therapy in Non-small Cell Lung Cancer. Cancer Res, 2019, 79(6):1204–13., DOI 10.1158/0008-5472. can-18-1082 Blankenberg FG. In Vivo imaging of Apoptosis. Cancer Biol Ther, 2008, 7(10):1525–32., DOI 10.4161/cbt.7.10.6934 Maier P, Hartmann L,Wenz F, Herskind C. Cellular Pathways in Response to Ionizing Radiation and Their Targetability for Tumor Radiosensitization. Int J Mol Sci, 2016, 17(1):102., DOI 10.3390/ijms17010102 Patsch K, Matasci N, Soundararajan A, Diaz P, Agus DB, Ruderman D, et al. Monitoring Dynamic Cytotoxic Chemotherapy Response in Castration- Resistant Prostate Cancer Using Plasma Cell-free DNA, cfDNA). BMC Res Notes, 2019, 12(1):275., DOI 10.1186/s13104-019-4312-2 Moser T, Waldispuehl-Geigl J, Belic J, Weber S, Zhou Q, Hasenleithner SO, et al. On-treatment Measurements of Circulating Tumor DNA during FOLFOX Therapy in Patients with Colorectal Cancer. NPJ Precis Oncol, 2020, 4(1):30., DOI 10.1038/s41698-020-00134-3 Lyskjaer I, Kronborg CS, Rasmussen MH, Sorensen BS, Demuth C, Rosenkilde M, et al. Correlation between Early Dynamics in Circulating Tumour DNA and Outcome from FOLFIRI Treatment in Metastatic Colorectal Cancer. Sci Rep, 2019, 9(1):11542., DOI 10.1038/s41598-019- 47708-1 Wang H, Zhang B, Chen D, Xia W, Zhang J, Wang F, et al. Real-time Monitoring Efficiency and Toxicity of Chemotherapy in Patients with Advanced Lung Cancer. Clin Epigenet, 2015, 7:119., DOI 10.1186/s13148- 015-0150-9 Kruger S, Heinemann V, Ross C, Diehl F, Nagel D, Ormanns S, et al. Repeated mutKRAS ctDNA Measurements Represent a Novel and Promising Tool for Early Response Prediction and Therapy Monitoring in Advanced Pancreatic Cancer. Ann Oncol, 2018, 29(12):2348–55., DOI 10.1093/annonc/mdy417 Chera BS, Kumar S, Shen C, Amdur R, Dagan R, Green R, et al. Plasma Circulating Tumor HPV DNA for the Surveillance of Cancer Recurrence in HPV-Associated Oropharyngeal Cancer. J Clin Oncol, 2020, 38(10):1050–8., DOI 10.1200/jco.19.02444 Moding EJ, Liu Y, Hui AB, He J, Qiao Y, Xu T, et al. Abstract PO-069: Circulating Tumor DNA Kinetics to Identify Genomic Predictors of Rapid Response to Chemoradiation in Non-small Cell Lung Cancer. Clin Cancer Res, 2021, 27(8 Suppl.):PO-069., DOI 10.1158/1557-3265.radsci21-po-069 Breadner DA, Vincent MD, Correa R, Black M, Warner A, Sanatani M, et al. Exploitation of Treatment Induced Tumor Lysis to Enhance the Sensitivity of ctDNA Analysis: A First-In-Human Pilot Study. Lung Cancer, 2022, 165: 145–51., DOI 10.1016/j.lungcan.2022.01.013 Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the Serum of Cancer Patients and the Effect of Therapy. Cancer Res, 1977, 37(3):646–50. Medvedeva NG, Panyutin IV, Panyutin IG, Neumann RD. Phosphorylation of Histone H2AX in Radiation-Induced Micronuclei. Radiat Res, 2007, 168(4):493–8., DOI 10.1667/rr0788.1 Durante M, Formenti SC. Radiation-Induced Chromosomal Aberrations and Immunotherapy: Micronuclei, Cytosolic DNA, and Interferon-Production Pathway. Front Oncol, 2018, 8:192., DOI 10.3389/fonc.2018.00192 Walls GM, McConnell L, McAleese J, Murray P, Lynch TB, Savage K, et al. Early Circulating Tumour DNA Kinetics Measured by Ultra-deep Next- Generation Sequencing during Radical Radiotherapy for Non-small Cell Lung Cancer: A Feasibility Study. Radiat Oncol, 2020, 15(1):132., DOI 10. 1186/s13014-020-01583-7 Boniface C, Baker K, Dieg C, Halsey C, Rahmani R, Deffenbach M, et al. Abstract PR05: Radiation-Assisted Amplification Sequencing, RAMP-Seq): Evaluating the Use of Stereotactic Body Radiation Therapy, SBRT, for Enriching Circulating Tumor DNA in Liquid Biopsies. Clin Cancer Res, 2020, 26(11 Suppl.):PR05., DOI 10.1158/1557-3265.liqbiop20-pr05 Chen EL, Chaudhuri AA, Nabet BY, Chabon JJ, Merriott DJ, Loo BW, et al. Analysis of Circulating Tumor DNA Kinetics during Stereotactic Ablative Radiation Therapy for Non-small Cell Lung Cancer. Int J Radiat Oncol Biol Phys, 2018, 102(3 Suppl.):e676., DOI 10.1016/j.ijrobp.2018.07.1826 Chaudhuri AA, Lovejoy AF, Chabon JJ, Newman A, Stehr H, Merriott DJ, et al. Circulating Tumor DNA Analysis during Radiation Therapy for Localized Lung Cancer Predicts Treatment Outcome. Int J Radiat Oncol Biol Phys, 2017, 99(2 Suppl.):S1–S2., DOI 10.1016/j.ijrobp.2017.06.021 Johnson BE, Creason AL, Stommel JM, Keck JM, Parmar S, Betts CB, et al. An Omic and Multidimensional Spatial Atlas from Serial Biopsies of an Evolving Metastatic Breast Cancer. Cel Rep Med, 2022, 3(2):100525., DOI 10.1016/j. xcrm.2022.100525 Mole RH. Whole Body Irradiation-Radiobiology or Medicine? Br J Radiol, 1953, 26(305):234–41., DOI 10.1259/0007-1285-26-305-234 Neumann MHD, Bender S, Krahn T, Schlange T. ctDNA and CTCs in Liquid Biopsy - Current Status and where We Need to Progress. Comput Struct Biotechnol J, 2018, 16:190–5., DOI 10.1016/j.csbj.2018.05.002 Ng SP, Meas S, Bahig H, Skinner HD, Garden AS, Phan J, et al. Detection of Circulating Tumor Cells during Radiation Therapy in Patients with Head and Neck Cancer. Int J Radiat Oncol Biol Phys, 2018, 102(3 Suppl.):e177., DOI 10. 1016/j.ijrobp.2018.07.657 Lee JH, Long GV, Menzies AM, Lo S, Guminski A, Whitbourne K, et al. Association between Circulating Tumor DNA and Pseudoprogression in Patients with Metastatic Melanoma Treated with Anti-programmed Cell Death 1 Antibodies. JAMA Oncol, 2018, 4(5):717–21., DOI 10.1001/ jamaoncol.2017.5332 Ma Y, Wang Q, Dong Q, Zhan L, Zhang J. How to Differentiate Pseudoprogression from True Progression in Cancer Patients Treated with Immunotherapy. Am J Cancer Res, 2019, 9(8):1546–53. Varaljai R, Elouali S, Lueong SS, Wistuba-Hamprecht K, Seremet T, Siveke JT, et al. The Predictive and Prognostic Significance of Cell-free DNA Concentration in Melanoma. J Eur Acad Dermatol Venereol, 2021, 35(2): 387–95., DOI 10.1111/jdv.16766 Goldberg SB, Narayan A, Kole AJ, Decker RH, Teysir J, Carriero NJ, et al. Early Assessment of Lung Cancer Immunotherapy Response via Circulating Tumor DNA. Clin Cancer Res, 2018, 24(8):1872–80., DOI 10.1158/1078-0432. ccr-17-1341 Kato K, Uchida J, Kukita Y, Kumagai T, Nishino K, Inoue T, et al. Transient Appearance of Circulating Tumor DNA Associated with De Novo Treatment. Sci Rep, 2016, 6:38639., DOI 10. 1038/srep38639 Riediger AL, Dietz S, Schirmer U, Meister M, Heinzmann-Groth I, Schneider M, et al. Mutation Analysis of Circulating Plasma DNA to Determine Response to EGFR Tyrosine Kinase Inhibitor Therapy of Lung Adenocarcinoma Patients. Sci Rep, 2016, 6:33505., DOI 10.1038/ srep33505 Horn L, Whisenant JG, Wakelee H, Reckamp KL, Qiao H, Leal TA, et al. Monitoring Therapeutic Response and Resistance: Analysis of Circulating Tumor DNA in Patients with ALK+ Lung Cancer. J Thorac Oncol, 2019, 14(11):1901–11., DOI 10.1016/j.jtho.2019.08.003 Wyatt AW, Azad AA, Volik SV, Annala M, Beja K, McConeghy B, et al. Genomic Alterations in Cell-free DNA and Enzalutamide Resistance in Castration-Resistant Prostate Cancer. JAMA Oncol, 2016, 2(12):1598–606., DOI 10.1001/jamaoncol.2016.0494 Romanel A, Gasi Tandefelt D, Conteduca V, Jayaram A, Casiraghi N, Wetterskog D, et al. Plasma AR and Abiraterone-Resistant Prostate Cancer. Sci Transl Med, 2015, 7(312):312re10., DOI 10.1126/scitranslmed. aac9511 Breadner DA, Vincent MD, Correa RJM, Black M, Warner A, Qu M, et al. Exploitation of Treatment Induced Tumor Lysis to Enhance Sensitivity of ctDNA Analysis: A First-In-Human Pilot Study. J Clin Oncol, 2020, 38(15_Suppl. l):3530., DOI 10.1200/jco.2020.38.15_suppl.3530 van de Donk PP, Kist de Ruijter L, Lub-de Hooge MN, Brouwers AH, van der Wekken AJ, Oosting SF, et al. Molecular Imaging Biomarkers for Immune Checkpoint Inhibitor Therapy. Theranostics, 2020, 10(4):1708–18., DOI 10. 7150/thno.38339 Wagner JT, Kim HJ, Johnson-Camacho KC, Kelley T, Newell LF, Spellman PT, et al. Diurnal Stability of Cell-free DNA and Cell-free RNA in Human Plasma Samples. Sci Rep, 2020, 10(1):16456., DOI 10.1038/s41598-020- 73350-3 O’Leary B, Hrebien S, Morden JP, Beaney M, Fribbens C, Huang X, et al. Early Circulating Tumor DNA Dynamics and Clonal Selection with Palbociclib and Fulvestrant for Breast Cancer. Nat Commun, 2018, 9(1):896., DOI 10. 1038/s41467-018-03215-x NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610103 EP 1610114 DI 10.3389/pore.2022.1610103 PG 12 ER PT J TI Senescence-Associated miRNAs and Their Role in Pancreatic Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE oncogene; pancreatic ductal adenocarcinoma; cellular senescence; senescence-associated miRNA; senescence bypass; tumor suppressor ID oncogene; pancreatic ductal adenocarcinoma; cellular senescence; senescence-associated miRNA; senescence bypass; tumor suppressor AB Replicative senescence is irreversible cell proliferation arrest for somatic cells which can be circumvented in cancers. Cellular senescence is a process, which may play two opposite roles. On the one hand, this is a natural protection of somatic cells against unlimited proliferation and malignant transformation. On the other hand, cellular secretion caused by senescence can stimulate inflammation and proliferation of adjacent cells that may promote malignancy. The main genes controlling the senescence pathways are also well known as tumor suppressors. Almost 140 genes regulate both cellular senescence and cancer pathways. About two thirds of these genes (64%) are regulated by microRNAs. Senescence-associated miRNAs can stimulate cancer progression or act as tumor suppressors. Here we review the role playing by senescence-associated miRNAs in development, diagnostics and treatment of pancreatic cancer. CR Hayflick L. The Limited In Vitro Lifetime of Human Diploid Cell Strains. Exp Cel Res, 1965, 37:614–36., DOI 10.1016/0014-4827(65, 90211-9 Miura T, Mattson MP, RaoMS. Cellular Lifespan and Senescence Signaling in Embryonic Stem Cells. Aging Cell, 2004, 3:333–43., DOI 10.1111/j.1474-9728. 2004.00134.x Ag Moir J, AWhite S, Mann J. Arrested Development and the Great Escape - the Role of Cellular Senescence in Pancreatic Cancer. Int J Biochem Cel Biol, 2014, 57:142–8., DOI 10.1016/j.biocel.2014.10.018 Porciuncula A, Hajdu C, David G. The Dual Role of Senescence in Pancreatic Ductal Adenocarcinoma. Adv Cancer Res, 2016, 131:1–20., DOI 10.1016/bs. acr.2016.05.006 Rodier F, Campisi J. Four Faces of Cellular Senescence. J Cel Biol, 2011, 192: 547–56., DOI 10.1083/jcb.201009094 Ou HL, Hoffmann R, Gonzalez-Lopez C, Doherty GJ, Korkola JE, Munoz- Espin D. Cellular Senescence in Cancer: from Mechanisms to Detection. Mol Oncol, 2021, 15:2634–71., DOI 10.1002/1878-0261.12807 Nakamura AJ, Chiang YJ, Hathcock KS, Horikawa I, Sedelnikova OA, Hodes RJ, et al. Both telomeric and Non-telomeric DNA Damage Are Determinants of Mammalian Cellular Senescence. Epigenetics Chromatin, 2008, 1:6., DOI 10. 1186/1756-8935-1-6 Campisi J. Cellular Senescence: Putting the Paradoxes in Perspective. Curr Opin Genet Develop, 2011, 21:107–12., DOI 10.1016/j.gde.2010.10.005 Collado M, Blasco MA, Serrano M. Cellular Senescence in Cancer and Aging. Cell, 2007, 130:223–33., DOI 10.1016/j.cell.2007.07.003 Gire V, Dulic V. Senescence from G2 Arrest, Revisited. Cell Cycle, 2015, 14: 297–304., DOI 10.1080/15384101.2014.1000134 Kumari R, Jat P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front Cel Dev Biol, 2021, 9: 645593., DOI 10.3389/fcell.2021.645593 Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Oncogenic Ras Provokes Premature Cell Senescence Associated with Accumulation of P53 and p16INK4a. Cell, 1997, 88:593–602., DOI 10.1016/s0092-8674(00)81902-9 Ko A, Han SY, Song J. Dynamics of ARF Regulation that Control Senescence and Cancer. BMB Rep, 2016, 49:598–606., DOI 10.5483/BMBRep.2016.49. 11.120 Dimri GP. What Has Senescence Got to Do with Cancer? Cancer Cell, 2005, 7:505–12., DOI 10.1016/j.ccr.2005.05.025 Silva J, Silva JM, Dominguez G, Garcia JM, Cantos B, Rodriguez R, et al. Concomitant Expression ofp16INK4aandp14ARFin Primary Breast Cancer and Analysis of Inactivation Mechanisms. J Pathol, 2003, 199:289–97., DOI 10. 1002/path.1297 Dominguez G, Silva J, Garcia JM, Silva JM, Rodriguez R, Munoz C, et al. Prevalence of Aberrant Methylation of p14ARF over p16INK4a in Some Human Primary Tumors. Mutat Res Fundam Mol Mech Mutagenesis, 2003, 530:9–17., DOI 10.1016/s0027-5107(03)00133-7 Tannapfel A, Busse C, Geissler F, Witzigmann H, Hauss J, Wittekind C. INK4a-ARF Alterations in Liver Cell Adenoma. Gut, 2002, 51:253–8., DOI 10. 1136/gut.51.2.253 Hansel DE, Kern SE, Hruban RH. Molecular Pathogenesis of Pancreatic Cancer. Annu Rev Genom Hum Genet, 2003, 4:237–56., DOI 10.1146/annurev. genom.4.070802.110341 Astle MV, Hannan KM, Ng PY, Lee RS, George AJ, Hsu AK, et al. AKT Induces Senescence in Human Cells via mTORC1 and P53 in the Absence of DNA Damage: Implications for Targeting mTOR during Malignancy. Oncogene, 2012, 31:1949–62., DOI 10.1038/onc.2011.394 Murthy D, Attri KS, Singh PK. Phosphoinositide 3-Kinase Signaling Pathway in Pancreatic Ductal Adenocarcinoma Progression, Pathogenesis, and Therapeutics. Front Physiol, 2018, 9:335., DOI 10.3389/fphys.2018.00335 Lin S, Yang J, Elkahloun AG, Bandyopadhyay A, Wang L, Cornell JE, et al. Attenuation of TGF-β Signaling Suppresses Premature Senescence in a P21- dependent Manner and Promotes Oncogenic Ras-Mediated Metastatic Transformation in Human Mammary Epithelial Cells. MBoC, 2012, 23: 1569–81., DOI 10.1091/mbc.E11-10-0849 Roupakia E, Markopoulos GS, Kolettas E. Genes and Pathways Involved in Senescence Bypass Identified by Functional Genetic Screens. Mech Ageing Develop, 2021, 194:111432., DOI 10.1016/j.mad.2021.111432 Jing H, Lee S. NF-κB in Cellular Senescence and Cancer Treatment. Mol Cell, 2014, 37:189–95., DOI 10.14348/molcells.2014.2353 Hoare M, Ito Y, Kang T-W, Weekes MP, Matheson NJ, Patten DA, et al. NOTCH1 Mediates a Switch between Two Distinct Secretomes during Senescence. Nat Cel Biol, 2016, 18:979–92., DOI 10.1038/ncb3397 Parry AJ, Hoare M, Bihary D, Hansel-Hertsch R, Smith S, Tomimatsu K, et al. NOTCH-mediated Non-cell Autonomous Regulation of Chromatin Structure during Senescence. Nat Commun, 2018, 9:1840., DOI 10.1038/ s41467-018-04283-9 Aster JC, Pear WS, Blacklow SC. TheVaried Roles ofNotch inCancer. Annu Rev Pathol Mech Dis, 2017, 12:245–75., DOI 10.1146/annurev-pathol-052016-100127 Chien Y, Scuoppo C, Wang X, Fang X, Balgley B, Bolden JE, et al. Control of the Senescence-Associated Secretory Phenotype by NF-κB Promotes Senescence and Enhances Chemosensitivity. Genes Dev, 2011, 25: 2125–36., DOI 10.1101/gad.17276711 Laberge R-M, Sun Y, Orjalo AV, Patil CK, Freund A, Zhou L, et al. MTOR Regulates the Pro-tumorigenic Senescence-Associated Secretory Phenotype by Promoting IL1A Translation. Nat Cel Biol, 2015, 17:1049–61., DOI 10.1038/ ncb3195 Teo YV, Rattanavirotkul N, Olova N, Salzano A, Quintanilla A, Tarrats N, et al. Notch Signaling Mediates Secondary Senescence. Cel Rep, 2019, 27: 997–1007., DOI 10.1016/j.celrep.2019.03.104 Coppe J-P, Desprez P-Y, Krtolica A, Campisi J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu Rev Pathol Mech Dis, 2010, 5:99–118., DOI 10.1146/annurev-pathol-121808-102144 Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, et al. Cellular Senescence: Defining a Path Forward. Cell, 2019, 179:813–27., DOI 10. 1016/j.cell.2019.10.005 Kuilman T, Michaloglou C, Vredeveld LCW, Douma S, van Doorn R, Desmet CJ, et al. Oncogene-Induced Senescence Relayed by an Interleukin-dependent Inflammatory Network. Cell, 2008, 133:1019–31., DOI 10.1016/j.cell.2008. 03.039 Acosta JC, O’Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, et al. Chemokine Signaling via the CXCR2 Receptor Reinforces Senescence. Cell, 2008, 133:1006–18., DOI 10.1016/j.cell.2008.03.038 Kojima H, Inoue T, Kunimoto H, Nakajima K. IL-6-STAT3 Signaling and Premature Senescence. JAK-STAT, 2013, 2:e25763., DOI 10.4161/jkst.25763 Laberge R-M, Awad P, Campisi J, Desprez P-Y. Epithelial-mesenchymal Transition Induced by Senescent Fibroblasts. Cancer Microenviron, 2012, 5: 39–44., DOI 10.1007/s12307-011-0069-4 Kapoor P, Deshmukh R. VEGF: A Critical Driver for Angiogenesis and Subsequent Tumor Growth: An IHC Study. J Oral Maxillofac Pathol, 2012, 16:330–7., DOI 10.4103/0973-029X.102478 Olivieri F, Rippo MR, Monsurro V, Salvioli S, Capri M, Procopio AD, et al. MicroRNAs Linking Inflamm-Aging, Cellular Senescence and Cancer. Ageing Res Rev, 2013, 12:1056–68., DOI 10.1016/j.arr.2013.05.001 Collado M, Gil J, Efeyan A, Guerra C, Schuhmacher AJ, Barradas M, et al. Senescence in Premalignant Tumours. Nature, 2005, 436:642., DOI 10.1038/ 436642a Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, et al. Pancreatic Cancer Genomes Reveal Aberrations in Axon Guidance Pathway Genes. Nature, 2012, 491:399–405., DOI 10.1038/nature11547 Waters AM, Der CJ. KRAS: The Critical Driver and Therapeutic Target for Pancreatic Cancer. Cold Spring Harb Perspect Med, 2018, 8:a031435., DOI 10. 1101/cshperspect.a031435 Caldwell ME, DeNicola GM, Martins CP, Jacobetz MA, Maitra A, Hruban RH, et al. Cellular Features of Senescence during the Evolution of Human and Murine Ductal Pancreatic Cancer. Oncogene, 2012, 31:1599–608., DOI 10. 1038/onc.2011.350 Deschenes-Simard X, Parisotto M, Rowell M-C, Le Calve B, Igelmann S, Moineau-Vallee K, et al. Circumventing Senescence Is Associated with Stem Cell Properties and Metformin Sensitivity. Aging Cell, 2019, 18:e12889., DOI 10.1111/acel.12889 Guerra C, Collado M, Navas C, Schuhmacher AJ, Hernandez-Porras I, Canamero M, et al. Pancreatitis-induced Inflammation Contributes to Pancreatic Cancer by Inhibiting Oncogene-Induced Senescence. Cancer Cell, 2011, 19:728–39., DOI 10.1016/j.ccr.2011.05.011 Carriere C, Gore AJ, Norris AM, Gunn JR, Young AL, Longnecker DS, et al. Deletion of Rb Accelerates Pancreatic Carcinogenesis by Oncogenic Kras and Impairs Senescence in Premalignant Lesions. Gastroenterology, 2011, 141: 1091–101., DOI 10.1053/j.gastro.2011.05.041 Schild C, Wirth M, Reichert M, Schmid RM, Saur D, Schneider G. PI3K Signaling Maintains C-Myc Expression to Regulate Transcription of E2F1 in Pancreatic Cancer Cells. Mol Carcinog, 2009, 48:1149–58., DOI 10.1002/mc. 20569 Ciernikova S, Earl J, Garcia Bermejo ML, Stevurkova V, Carrato A, Smolkova B. Epigenetic Landscape in Pancreatic Ductal Adenocarcinoma: On the Way to Overcoming Drug Resistance? Int J Mol Sci, 2020, 21:4091., DOI 10.3390/ ijms21114091 Gao J, Wang L, Xu J, Zheng J, Man X, Wu H, et al. Aberrant DNA Methyltransferase Expression in Pancreatic Ductal Adenocarcinoma Development and Progression. J Exp Clin Cancer Res, 2013, 32:86., DOI 10. 1186/1756-9966-32-86 Ueki T, Toyota M, Sohn T, Yeo CJ, Issa JP, Hruban RH, et al. Hypermethylation of Multiple Genes in Pancreatic Adenocarcinoma. Cancer Res, 2000, 60:1835–9. Hong L, Sun G, Peng L, Tu Y, Wan Z, Xiong H, et al. The Interaction between miR-148a and DNMT1 Suppresses Cell Migration and Invasion by Reactivating Tumor Suppressor Genes in Pancreatic Cancer. Oncol Rep, 2018, 40:2916–25., DOI 10.3892/or.2018.6700 Lowery MA, Jordan EJ, Basturk O, Ptashkin RN, Zehir A, Berger MF, et al. Real-Time Genomic Profiling of Pancreatic Ductal Adenocarcinoma: Potential Actionability and Correlation with Clinical Phenotype. Clin Cancer Res, 2017, 23:6094–100., DOI 10.1158/1078-0432.CCR-17-0899 Maniati E, Bossard M, Cook N, Candido JB, Emami-Shahri N, Nedospasov SA, et al. Crosstalk between the Canonical NF-κB and Notch Signaling Pathways Inhibits Pparγ Expression and Promotes Pancreatic Cancer Progression in Mice. J Clin Invest, 2011, 121:4685–99., DOI 10.1172/JCI45797 Prabhu L, Mundade R, Korc M, Loehrer PJ, Lu T. Critical Role of NF-κB in Pancreatic Cancer. Oncotarget, 2014, 5:10969–75., DOI 10.18632/oncotarget. 2624 Penfield JD, Anderson M, Lutzke L, Wang KK. The Role of Cellular Senescence in the Gastrointestinal Mucosa. Gut Liver, 2013, 7:270–7., DOI 10.5009/gnl.2013.7.3.270 Fane M, Weeraratna AT. How the Ageing Microenvironment Influences Tumour Progression. Nat Rev Cancer, 2020, 20:89–106., DOI 10.1038/s41568- 019-0222-9 Xue R, Jia K, Wang J, Yang L, Wang Y, Gao L, et al. A Rising Star in Pancreatic Diseases: Pancreatic Stellate Cells. Front Physiol, 2018, 9:754., DOI 10.3389/ fphys.2018.00754 Petroni G, Galluzzi L. Senescence Inflames the Pancreatic Tumor Microenvironment. Cel Rep Med, 2020, 1:100020., DOI 10.1016/j.xcrm. 2020.100020 Shao C, Tu C, Cheng X, Xu Z, Wang X, Shen J, et al. Inflammatory and Senescent Phenotype of Pancreatic Stellate Cells Induced by Sqstm1 Downregulation Facilitates Pancreatic Cancer Progression. Int J Biol Sci, 2019, 15:1020–9., DOI 10.7150/ijbs.27825 Wang T, Notta F, Navab R, Joseph J, Ibrahimov E, Xu J, et al. Senescent Carcinoma-Associated Fibroblasts Upregulate IL8 to Enhance Prometastatic Phenotypes. Mol Cancer Res, 2017, 15:3–14., DOI 10.1158/1541-7786.MCR- 16-0192 Tacutu R, Budovsky A, Yanai H, Fraifeld VE. Molecular Links between Cellular Senescence, Longevity and Age-Related Diseases - a Systems Biology Perspective. Aging, 2011, 3:1178–91., DOI 10.18632/aging.100413 Bayraktar R, Van Roosbroeck K, Calin GA. Cell-to-cell Communication: microRNAs as Hormones. Mol Oncol, 2017, 11:1673–86., DOI 10.1002/1878- 0261.12144 Bueno MJ, Malumbres M. MicroRNAs and the Cell Cycle. Biochim Biophys Acta Mol Basis Dis, 2011, 1812:592–601., DOI 10.1016/j.bbadis.2011.02.002 Romano R, Picca A, Eusebi LHU, Marzetti E, Calvani R, Moro L, et al. Extracellular Vesicles and Pancreatic Cancer: Insights on the Roles of miRNA, lncRNA, and Protein Cargos in Cancer Progression. Cells, 2021, 10:1361., DOI 10.3390/cells10061361 Conti I, Varano G, Simioni C, Laface I, Milani D, Rimondi E, et al. miRNAs as Influencers of Cell-Cell Communication in Tumor Microenvironment. Cells, 2020, 9:220., DOI 10.3390/cells9010220 Uddin MH, Al-Hallak MN, Philip PA, Mohammad RM, Viola N, Wagner K-U, et al. Exosomal microRNA in Pancreatic Cancer Diagnosis, Prognosis, and Treatment: From Bench to Bedside. Cancers, 2021, 13:2777., DOI 10.3390/ cancers13112777 Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, et al. Pancreatic Cancer Exosomes Initiate Pre-metastatic Niche Formation in the Liver. Nat Cel Biol, 2015, 17:816–26., DOI 10.1038/ncb3169 Sun W, Ren Y, Lu Z, Zhao X. The Potential Roles of Exosomes in Pancreatic Cancer Initiation and Metastasis. Mol Cancer, 2020, 19:135., DOI 10.1186/ s12943-020-01255-w Vicentini C, Calore F, Nigita G, Fadda P, Simbolo M, Sperandio N, et al. Exosomal miRNA Signatures of Pancreatic Lesions. BMC Gastroenterol, 2020, 20:137., DOI 10.1186/s12876-020-01287-y Ling H, Fabbri M, Calin GA. MicroRNAs and Other Non-coding RNAs as Targets for Anticancer Drug Development. Nat Rev Drug Discov, 2013, 12: 847–65., DOI 10.1038/nrd4140 Chang X, Yu C, Li J, Yu S, Chen J. hsa-miR-96 and Hsa-miR-217 Expression Down-Regulates with Increasing Dysplasia in Pancreatic Intraepithelial Neoplasias and Intraductal Papillary Mucinous Neoplasms. Int J Med Sci, 2017, 14:412–8., DOI 10.7150/ijms.18641 Terlecki-Zaniewicz L, Lammermann I, Latreille J, Bobbili MR, Pils V, Schosserer M, et al. Small Extracellular Vesicles and their miRNA Cargo are Anti-apoptotic Members of the Senescence-Associated Secretory Phenotype. Aging, 2018, 10:1103–32., DOI 10.18632/aging.101452 Wallis R, Mizen H, Bishop CL. The Bright and Dark Side of Extracellular Vesicles in the Senescence-Associated Secretory Phenotype. Mech Ageing Develop, 2020, 189:111263., DOI 10.1016/j.mad.2020.111263 Wang Z, Tan Y, Yu W, Zheng S, Zhang S, Sun L, et al. Small Role with Big Impact: miRNAs as Communicators in the Cross-Talk between Cancer- Associated Fibroblasts and Cancer Cells. Int J Biol Sci, 2017, 13:339–48., DOI 10.7150/ijbs.17680 Zhang Y, Yang P, Wang X-F. Microenvironmental Regulation of Cancer Metastasis by miRNAs. Trends Cel Biol, 2014, 24:153–60., DOI 10.1016/j.tcb. 2013.09.007 Gascard P, Tlsty TD. Carcinoma-associated Fibroblasts: Orchestrating the Composition of Malignancy. Genes Dev, 2016, 30:1002–19., DOI 10.1101/gad. 279737.116 Pang W, Su J, Wang Y, Feng H, Dai X, Yuan Y, et al. Pancreatic Cancersecreted miR-155 Implicates in the Conversion from normal Fibroblasts to Cancer-associated Fibroblasts. Cancer Sci, 2015, 106:1362–9., DOI 10.1111/ cas.12747 Sun Q, Zhang B, Hu Q, Qin Y, Xu W, Liu W, et al. The Impact of Cancer- Associated Fibroblasts on Major Hallmarks of Pancreatic Cancer. Theranostics, 2018, 8:5072–87., DOI 10.7150/thno.26546 Olivieri F, Albertini MC, Orciani M, Ceka A, Cricca M, Procopio AD, et al. DNA Damage Response, DDR, and Senescence: Shuttled InflammamiRNAs on the Stage of Inflamm-Aging. Oncotarget, 2015, 6:35509–21., DOI 10.18632/oncotarget.5899 Cortesi M, Zanoni M, Pirini F, Tumedei MM, Ravaioli S, Rapposelli IG, et al. Pancreatic Cancer and Cellular Senescence: Tumor Microenvironment under the Spotlight. Int J Mol Sci, 2021, 23:254., DOI 10.3390/ijms23010254 Feliciano A, Sanchez-Sendra B, Kondoh H, Lleonart ME. MicroRNAs Regulate Key Effector Pathways of Senescence. J Aging Res, 2011, 2011: 1–11., DOI 10.4061/2011/205378 Nakata K, Ohuchida K, Mizumoto K, Kayashima T, Ikenaga N, Sakai H, et al. MicroRNA-10b Is Overexpressed in Pancreatic Cancer, Promotes its Invasiveness, and Correlates with a Poor Prognosis. Surgery, 2011, 150: 916–22., DOI 10.1016/j.surg.2011.06.017 Lee EJ, Gusev Y, Jiang J, Nuovo GJ, Lerner MR, Frankel WL, et al. Expression Profiling Identifies microRNA Signature in Pancreatic Cancer. Int J Cancer, 2007, 120:1046–54., DOI 10.1002/ijc.22394 Buscaglia LEB, Li Y. Apoptosis and the Target Genes of microRNA-21. Chin J Cancer, 2011, 30:371–80., DOI 10.5732/cjc.011.10132 Park J-K, Lee EJ, Esau C, Schmittgen TD. Antisense Inhibition of microRNA- 21 or -221 Arrests Cell Cycle, Induces Apoptosis, and Sensitizes the Effects of Gemcitabine in Pancreatic Adenocarcinoma. Pancreas, 2009, 38:e190–e199., DOI 10.1097/MPA.0b013e3181ba82e1 Liu J, Xu D, Wang Q, Zheng D, Jiang X, Xu L. LPS Induced miR-181a Promotes Pancreatic Cancer Cell Migration via Targeting PTEN and MAP2K4. Dig Dis Sci, 2014, 59:1452–60., DOI 10.1007/s10620-014- 3049-y Du Rieu MC, Torrisani J, Selves J, Al Saati T, Souque A, Dufresne M, et al. MicroRNA-21 Is Induced Early in Pancreatic Ductal Adenocarcinoma Precursor Lesions. Clin Chem, 2010, 56:603–12., DOI 10.1373/clinchem. 2009.137364 Giovannetti E, Funel N, Peters GJ, Del Chiaro M, Erozenci LA, Vasile E, et al. MicroRNA-21 in Pancreatic Cancer: Correlation with Clinical Outcome and Pharmacologic Aspects Underlying its Role in the Modulation of Gemcitabine Activity. Cancer Res, 2010, 70:4528–38., DOI 10.1158/0008- 5472.CAN-09-4467 Moriyama T, Ohuchida K, Mizumoto K, Yu J, Sato N, Nabae T, et al. MicroRNA-21 Modulates Biological Functions of Pancreatic Cancer Cells Including Their Proliferation, Invasion, and Chemoresistance. Mol Cancer Ther, 2009, 8:1067–74., DOI 10.1158/1535-7163.MCT-08-0592 Hu G-y., Tao F, Wang W, Ji K-w. Prognostic Value of microRNA-21 in Pancreatic Ductal Adenocarcinoma: a Meta-Analysis. World J Surg Onc, 2016, 14:82., DOI 10.1186/s12957-016-0842-4 Ouyang H, Gore J, Deitz S, Korc M. microRNA-10b Enhances Pancreatic Cancer Cell Invasion by Suppressing TIP30 Expression and Promoting EGF and TGF-β Actions. Oncogene, 2014, 33:4664–74., DOI 10.1038/onc.2013.405 Zhang W-L, Zhang J-H, Wu X-Z, Yan T, Lv W. miR-15b Promotes Epithelial-Mesenchymal Transition by Inhibiting SMURF2 in Pancreatic Cancer. Int J Oncol, 2015, 47:1043–53., DOI 10.3892/ijo.2015.3076 Cloonan N, Brown MK, Steptoe AL, Wani S, Chan W, Forrest AR, et al. The miR-17-5p microRNA Is a Key Regulator of the G1/S Phase Cell Cycle Transition. Genome Biol, 2008, 9:R127., DOI 10.1186/gb-2008-9-8-r127 Dellago H, Bobbili MR, Grillari J. MicroRNA-17-5p: At the Crossroads of Cancer and Aging - A Mini-Review. Gerontology, 2017, 63:20–8., DOI 10. 1159/000447773 Yu J, Ohuchida K, Mizumoto K, Fujita H, Nakata K, Tanaka M. MicroRNAmiR-17-5pis Overexpressed in Pancreatic Cancer, Associated with a Poor Prognosis, and Involved in Cancer Cell Proliferation and Invasion. Cancer Biol Ther, 2010, 10:748–57., DOI 10. 4161/cbt.10.8.13083 Zhu Y, Gu J, Li Y, Peng C, Shi M, Wang X, et al. MiR-17-5p Enhances Pancreatic Cancer Proliferation by Altering Cell Cycle Profiles via Disruption of RBL2/E2F4-Repressing Complexes. Cancer Lett, 2018, 412:59–68., DOI 10. 1016/j.canlet.2017.09.044 Gironella M, Seux M, Xie M-J, Cano C, Tomasini R, Gommeaux J, et al. Tumor Protein 53-induced Nuclear Protein 1 Expression is Repressed by miR-155, and its Restoration Inhibits Pancreatic Tumor Development. Proc Natl Acad Sci U.S.A, 2007, 104:16170–5., DOI 10.1073/pnas. 0703942104 Wang P, Zhu C-f., Ma M-z., Chen G, Song M, Zeng Z-l., et al. Micro-RNA- 155 is Induced by K-Ras Oncogenic Signal and Promotes ROS Stress in Pancreatic Cancer. Oncotarget, 2015, 6:21148–58., DOI 10.18632/oncotarget. 4125 Huang C, Li H, Wu W, Jiang T, Qiu Z. Regulation of miR-155 Affects Pancreatic Cancer Cell Invasiveness and Migration by Modulating the STAT3 Signaling Pathway through SOCS1. Oncol Rep, 2013, 30:1223–30., DOI 10.3892/or.2013.2576 Fasanaro P, D’Alessandra Y, Di Stefano V, Melchionna R, Romani S, Pompilio G, et al. MicroRNA-210 Modulates Endothelial Cell Response to Hypoxia and Inhibits the Receptor Tyrosine Kinase Ligand Ephrin-A3. J Biol Chem, 2008, 283:15878–83., DOI 10.1074/jbc.M800731200 Zhang Z, Sun H, Dai H, Walsh R, Imakura M, Schelter J, et al. MicroRNA miR-210 Modulates Cellular Response to Hypoxia through the MYC Antagonist MNT. Cell Cycle, 2009, 8:2756–68., DOI 10.4161/cc.8.17.9387 Huang X, Le Q-T, Giaccia AJ. MiR-210 - Micromanager of the Hypoxia Pathway. Trends Mol Med, 2010, 16:230–7., DOI 10.1016/j.molmed.2010. 03.004 Huang X, Zuo J. Emerging Roles of miR-210 and Other Non-coding RNAs in the Hypoxic Response. Acta Biochim Biophys Sinica, 2014, 46:220–32., DOI 10. 1093/abbs/gmt141 Xu Q, Li P, Chen X, Zong L, Jiang Z, Nan L, et al. miR-221/222 Induces Pancreatic Cancer Progression through the Regulation of Matrix Metalloproteinases. Oncotarget, 2015, 6:14153–64., DOI 10.18632/ oncotarget.3686 Sarkar S, Dubaybo H, Ali S, Goncalves P, Kollepara SL, Sethi S, et al. Downregulation of miR-221 Inhibits Proliferation of Pancreatic Cancer Cells through Up-Regulation of PTEN, P27(kip1), P57(kip2), and PUMA. Am J Cancer Res, 2013, 3:465–77. Balzeau J, Menezes MR, Cao S, Hagan JP. The LIN28/let-7 Pathway in Cancer. Front Genet, 2017, 8:31., DOI 10.3389/fgene.2017.00031 Wang H, Chirshev E, Hojo N, Suzuki T, Bertucci A, Pierce M, et al. The Epithelial-Mesenchymal Transcription Factor SNAI1 Represses Transcription of the Tumor Suppressor miRNA Let-7 in Cancer. Cancers, 2021, 13:1469., DOI 10.3390/cancers13061469 Bhutia YD, Hung SW, Krentz M, Patel D, Lovin D, Manoharan R, et al. Differential Processing of Let-7a Precursors Influences RRM2 Expression and Chemosensitivity in Pancreatic Cancer: Role of LIN-28 and SET Oncoprotein. PLoS One, 2013, 8:e53436., DOI 10.1371/journal.pone. 0053436 Li XJ, Ren ZJ, Tang JH. MicroRNA-34a: a Potential Therapeutic Target in Human Cancer. Cell Death Dis, 2014, 5:e1327., DOI 10.1038/cddis. 2014.270 Iliopoulos D, Drakaki A. MicroRNA-gene Signaling Pathways in Pancreatic Cancer. Biomed J, 2013, 36:200–8., DOI 10.4103/2319-4170.119690 Kong YW, Ferland-McCollough D, Jackson TJ, Bushell M. microRNAs in Cancer Management. Lancet Oncol, 2012, 13:e249–e258., DOI 10.1016/S1470- 2045(12)70073-6 Guo S, Fesler A, Wang H, Ju J. microRNA Based Prognostic Biomarkers in Pancreatic Cancer. Biomark Res, 2018, 6:18., DOI 10.1186/s40364-018- 0131-1 Tang Y, Tang Y, Cheng Y-s. miR-34a Inhibits Pancreatic Cancer Progression through Snail1-Mediated Epithelial-Mesenchymal Transition and the Notch Signaling Pathway. Sci Rep, 2017, 7:38232., DOI 10.1038/srep38232 Hidalgo-Sastre A, Lubeseder-Martellato C, Engleitner T, Steiger K, Zhong S, Desztics J, et al. Mir34a Constrains Pancreatic Carcinogenesis. Sci Rep, 2020, 10:9654., DOI 10.1038/s41598-020-66561-1 Lee K-H, Lotterman C, Karikari C, Omura N, Feldmann G, Habbe N, et al. Epigenetic Silencing of MicroRNA miR-107 Regulates Cyclin-dependent Kinase 6 Expression in Pancreatic Cancer. Pancreatology, 2009, 9: 293–301., DOI 10.1159/000186051 Xiong J, Wang D, Wei A, Lu H, Tan C, Li A, et al. Deregulated Expression of miR-107 Inhibits Metastasis of PDAC through Inhibition PI3K/Akt Signaling via Caveolin-1 and PTEN. Exp Cel Res, 2017, 361:316–23., DOI 10.1016/j. yexcr.2017.10.033 Huang W, Gu J, Tao T, Zhang J, Wang H, Fan Y. MiR-24-3p Inhibits the Progression of Pancreatic Ductal Adenocarcinoma through LAMB3 Downregulation. Front Oncol, 2020, 9:1499., DOI 10.3389/fonc.2019. 01499 Sun Y, Wang P, Zhang Q, Wu H. CDK14/β-catenin/TCF4/miR-26b Positive Feedback Regulation Modulating Pancreatic Cancer Cell Phenotypes In Vitro and Tumor Growth in Mice Model In Vivo. J Gene Med, 2021, 24:e3343., DOI 10.1002/jgm.3343 Trehoux S, Lahdaoui F, Delpu Y, Renaud F, Leteurtre E, Torrisani J, et al. Micro-RNAs miR-29a and miR-330-5p Function as Tumor Suppressors by Targeting the MUC1 Mucin in Pancreatic Cancer Cells. Biochim Biophys Acta Mol Cel Res, 2015, 1853:2392–403., DOI 10.1016/j.bbamcr. 2015.05.033 Dey S, Kwon JJ, Liu S, Hodge GA, Taleb S, Zimmers TA, et al. miR-29a is Repressed by MYC in Pancreatic Cancer and its Restoration Drives Tumor- Suppressive Effects via Downregulation of LOXL2. Mol Cancer Res, 2020, 18: 311–23., DOI 10.1158/1541-7786.MCR-19-0594 Sun X-J, Liu B-Y, Yan S, Jiang T-H, Cheng H-Q, Jiang H-S, et al. MicroRNA- 29a Promotes Pancreatic Cancer Growth by Inhibiting Tristetraprolin. Cell Physiol Biochem, 2015, 37:707–18., DOI 10.1159/000430389 Wang T, Chen G, Ma X, Yang Y, Chen Y, Peng Y, et al. MiR-30a Regulates Cancer Cell Response to Chemotherapy through SNAI1/IRS1/AKT Pathway. Cel Death Dis, 2019, 10:1–15., DOI 10.1038/s41419-019-1326-6 Xu Y-F, Hannafon BN, Ding W-Q. microRNA Regulation of Human Pancreatic Cancer Stem Cells. Stem Cel Investig., 2017, 4:5., DOI 10.21037/ sci.2017.01.01 Lu Y, Lu J, Li X, Zhu H, Fan X, Zhu S, et al. MiR-200a Inhibits Epithelial- Mesenchymal Transition of Pancreatic Cancer Stem Cell. BMC Cancer, 2014, 14:85., DOI 10.1186/1471-2407-14-85 Zhao G, Wang B, Liu Y, Zhang J-g., Deng S-c., Qin Q, et al. miRNA-141, Downregulated in Pancreatic Cancer, Inhibits Cell Proliferation and Invasion by Directly Targeting MAP4K4. Mol Cancer Ther, 2013, 12:2569–80., DOI 10. 1158/1535-7163.MCT-13-0296 Hamada S, Satoh K, Fujibuchi W, Hirota M, Kanno A, Unno J, et al. MiR-126 Acts as a Tumor Suppressor in Pancreatic Cancer Cells via the Regulation of ADAM9. Mol Cancer Res, 2012, 10:3–10., DOI 10.1158/1541-7786.MCR-11- 0272 Xu WX, Liu Z, Deng F, Wang DD, Li XW, Tian T, et al. MiR-145: a Potential Biomarker of Cancer Migration and Invasion. Am J Transl Res, 2019, 11: 6739–53. Khan S, Ebeling MC, ZamanMS, Sikander M, Yallapu MM, Chauhan N, et al. MicroRNA-145 Targets MUC13 and Suppresses Growth and Invasion of Pancreatic Cancer. Oncotarget, 2014, 5:7599–609., DOI 10.18632/oncotarget. 2281 Han T, Yi X-P, Liu B, Ke M-J, Li Y-X. MicroRNA-145 Suppresses Cell Proliferation, Invasion and Migration in Pancreatic Cancer Cells by Targeting NEDD9. Mol Med Rep, 2015, 11:4115–20., DOI 10.3892/ mmr.2015.3294 Deng S, Zhu S, Wang B, Li X, Liu Y, Qin Q, et al. Chronic Pancreatitis and Pancreatic Cancer Demonstrate Active Epithelial-Mesenchymal Transition Profile, Regulated by miR-217-SIRT1 Pathway. Cancer Lett, 2014, 355: 184–91., DOI 10.1016/j.canlet.2014.08.007 Yang Y, Tao X, Li C-B, Wang C-M. MicroRNA-494 Acts as a Tumor Suppressor in Pancreatic Cancer, Inhibiting Epithelial-Mesenchymal Transition, Migration and Invasion by Binding to SDC1. Int J Oncol, 2018, 53:1204–14., DOI 10.3892/ijo.2018.4445 Chen G, Shi Y, Zhang Y, Sun J. CircRNA_100782 Regulates Pancreatic Carcinoma Proliferation through the IL6-STAT3 Pathway. OncoTargets Ther, 2017, 10:5783–94., DOI 10.2147/OTT.S150678 Wu D-H, Liang H, Lu S-N, Wang H, Su Z-L, Zhang L, et al. miR-124 Suppresses Pancreatic Ductal Adenocarcinoma Growth by Regulating Monocarboxylate Transporter 1-Mediated Cancer Lactate Metabolism. Cel Physiol Biochem, 2018, 50:924–35., DOI 10.1159/000494477 Gao L, Yang Y, Xu H, Liu R, Li D, Hong H, et al. MiR-335 Functions as a Tumor Suppressor in Pancreatic Cancer by Targeting OCT4. Tumor Biol, 2014, 35:8309–18., DOI 10.1007/s13277-014-2092-9 Neault M, Mallette FA, Richard S. miR-137 Modulates a Tumor Suppressor Network-Inducing Senescence in Pancreatic Cancer Cells. Cel Rep, 2016, 14: 1966–78., DOI 10.1016/j.celrep.2016.01.068 Li Y, VandenBoom TG, Wang Z, Kong D, Ali S, Philip PA, et al. miR-146a Suppresses Invasion of Pancreatic Cancer Cells. Cancer Res, 2010, 70: 1486–95., DOI 10.1158/0008-5472.CAN-09-2792 Delpu Y, Lulka H, Sicard F, Saint-Laurent N, Lopez F, Hanoun N, et al. The rescue of miR-148a Expression in Pancreatic Cancer: an Inappropriate Therapeutic Tool. PLoS One, 2013, 8:e55513., DOI 10.1371/journal.pone. 0055513 Zhao W-G, Yu S-N, Lu Z-H, Ma Y-H, Gu Y-M, Chen J. The miR-217 microRNA Functions as a Potential Tumor Suppressor in Pancreatic Ductal Adenocarcinoma by Targeting KRAS. Carcinogenesis, 2010, 31:1726–33., DOI 10.1093/carcin/bgq160 Wang B, Du R, Xiao X, Deng Z-L, Jian D, Xie H-F, et al. Microrna-217 Modulates Human Skin Fibroblast Senescence by Directly Targeting DNA Methyltransferase 1. Oncotarget, 2017, 8:33475–86., DOI 10.18632/oncotarget. 16509 Tesfaye AA, Azmi AS, Philip PA. miRNA and Gene Expression in Pancreatic Ductal Adenocarcinoma. Am J Pathol, 2019, 189:58–70., DOI 10.1016/j.ajpath. 2018.10.005 Friedman RC, Farh KK-H, Burge CB, Bartel DP. Most Mammalian mRNAs Are Conserved Targets of microRNAs. Genome Res, 2009, 19:92–105., DOI 10. 1101/gr.082701.108 Zhang Y, Li M, Wang H, Fisher WE, Lin PH, Yao Q, et al. Profiling of 95 MicroRNAs in Pancreatic Cancer Cell Lines and Surgical Specimens by Real- Time PCR Analysis. World J Surg, 2009, 33:698–709., DOI 10.1007/s00268- 008-9833-0 Hong TH, Park IY. MicroRNA Expression Profiling of Diagnostic Needle Aspirates from Surgical Pancreatic Cancer Specimens. Ann Surg Treat Res, 2014, 87:290., DOI 10.4174/astr.2014.87.6.290 Lee LS, Szafranska-Schwarzbach AE, Wylie D, Doyle LA, Bellizzi AM, Kadiyala V, et al. Investigating MicroRNA Expression Profiles in Pancreatic Cystic Neoplasms. Clin Transl Gastroenterol, 2014, 5:e47., DOI 10.1038/ctg.2013.18 Szafranska-Schwarzbach AE, Adai AT, Lee LS, Conwell DL, Andruss BF. Development of a miRNA-Based Diagnostic Assay for Pancreatic Ductal Adenocarcinoma. Expert Rev Mol Diagn, 2011, 11:249–57., DOI 10.1586/erm.11.10 Vila-Navarro E, Duran-Sanchon S, Vila-Casadesus M, Moreira L, Gines A, Cuatrecasas M, et al. Novel Circulating miRNA Signatures for Early Detection of Pancreatic Neoplasia. Clin Transl Gastroenterol, 2019, 10: e00029., DOI 10.14309/ctg.0000000000000029 Wei L, Yao K, Gan S, Suo Z. Clinical Utilization of Serum- or Plasma- Based miRNAs as Early Detection Biomarkers for Pancreatic Cancer. Medicine, Baltimore,, 2018, 97:e12132., DOI 10.1097/MD. 0000000000012132 Peng C, Wang J, Gao W, Huang L, Liu Y, Li X, et al. Meta-analysis of the Diagnostic Performance of Circulating MicroRNAs for Pancreatic Cancer. Int J Med Sci, 2021, 18:660–71., DOI 10.7150/ijms.52706 Alemar B, Izetti P, Gregorio C, Macedo GS, Castro MAA, Osvaldt AB, et al. miRNA-21 and miRNA-34a Are Potential Minimally Invasive Biomarkers for the Diagnosis of Pancreatic Ductal Adenocarcinoma. Pancreas, 2016, 45: 84–92., DOI 10.1097/MPA.0000000000000383 Sun B, Liu X, Gao Y, Li L, Dong Z. Downregulation of miR-124 Predicts Poor Prognosis in Pancreatic Ductal Adenocarcinoma Patients. Br J Biomed Sci, 2016, 73:152–7., DOI 10.1080/09674845.2016.1220706 Wald P, Liu XS, Pettit C, Dillhoff M, Manilchuk A, Schmidt C, et al. Prognostic Value of microRNA Expression Levels in Pancreatic Adenocarcinoma: a Review of the Literature. Oncotarget, 2017, 8: 73345–61., DOI 10.18632/oncotarget.20277 Frampton AE, Krell J, Jamieson NB, Gall TMH, Giovannetti E, Funel N, et al. microRNAs with Prognostic Significance in Pancreatic Ductal Adenocarcinoma: A Meta-Analysis. Eur J Cancer, 2015, 51:1389–404., DOI 10.1016/j.ejca.2015.04.006 Papaconstantinou IG, Manta A, Gazouli M, Lyberopoulou A, Lykoudis PM, Polymeneas G, et al. Expression of microRNAs in Patients with Pancreatic Cancer and its Prognostic Significance. Pancreas, 2013, 42:67–71., DOI 10. 1097/MPA.0b013e3182592ba7 Mikamori M, Yamada D, Eguchi H, Hasegawa S, Kishimoto T, Tomimaru Y, et al. MicroRNA-155 Controls Exosome Synthesis and Promotes Gemcitabine Resistance in Pancreatic Ductal Adenocarcinoma. Sci Rep, 2017, 7:42339., DOI 10.1038/srep42339 Yu Q, Xu C, Yuan W, Wang C, Zhao P, Chen L, et al. Evaluation of Plasma MicroRNAs as Diagnostic and Prognostic Biomarkers in Pancreatic Adenocarcinoma: miR-196a and miR-210 Could Be Negative and Positive Prognostic Markers, Respectively. Biomed Res Int, 2017, 2017:1–10., DOI 10. 1155/2017/6495867 Greither T, Grochola LF, Udelnow A, Lautenschlager C, Wurl P, Taubert H. Elevated Expression of microRNAs 155, 203, 210 and 222 in Pancreatic Tumors is Associated with Poorer Survival. Int J Cancer, 2010, 126:73–80., DOI 10.1002/ijc.24687 Bader AG, Brown D, Stoudemire J, Lammers P. Developing Therapeutic microRNAs for Cancer. Gene Ther, 2011, 18:1121–6., DOI 10.1038/gt.2011.79 Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M, et al. Silencing of microRNAs In Vivo with ’antagomirs’. Nature, 2005, 438:685–9., DOI 10.1038/nature04303 Cheng CJ, Bahal R, Babar IA, Pincus Z, Barrera F, Liu C, et al. MicroRNA Silencing for Cancer Therapy Targeted to the Tumour Microenvironment. Nature, 2015, 518:107–10., DOI 10.1038/nature13905 Ebert MS, Sharp PA. MicroRNA Sponges: Progress and Possibilities. RNA, 2010, 16:2043–50., DOI 10.1261/rna.2414110 Tay FC, Lim JK, Zhu H, Hin LC, Wang S. Using Artificial microRNA Sponges to Achieve microRNA Loss-Of-Function in Cancer Cells. Adv Drug Deliv Rev, 2015, 81:117–27., DOI 10.1016/j.addr.2014.05.010 Ben-Shushan D, Markovsky E, Gibori H, Tiram G, Scomparin A, Satchi- Fainaro R. Overcoming Obstacles in microRNA Delivery towards Improved Cancer Therapy. Drug Deliv Transl Res, 2014, 4:38–49., DOI 10.1007/s13346- 013-0160-0 Pramanik D, Campbell NR, Karikari C, Chivukula R, Kent OA, Mendell JT, et al. Restitution of Tumor Suppressor microRNAs Using a Systemic Nanovector Inhibits Pancreatic Cancer Growth in Mice. Mol Cancer Ther, 2011, 10:1470–80., DOI 10.1158/1535-7163.MCT-11-0152 Trang P, Wiggins JF, Daige CL, Cho C, Omotola M, Brown D, et al. Systemic Delivery of Tumor Suppressor microRNA Mimics Using a Neutral Lipid Emulsion Inhibits Lung Tumors in Mice. Mol Ther, 2011, 19:1116–22., DOI 10.1038/mt.2011.48 Kota J, Chivukula RR, O’Donnell KA, Wentzel EA, Montgomery CL, Hwang H-W, et al. Therapeutic microRNA Delivery Suppresses Tumorigenesis in a Murine Liver Cancer Model. Cell, 2009, 137:1005–17., DOI 10.1016/j.cell.2009. 04.021 Li L, Xie X, Luo J, Liu M, Xi S, Guo J, et al. Targeted Expression of miR-34a Using the T-VISA System Suppresses Breast Cancer Cell Growth and Invasion. Mol Ther, 2012, 20:2326–34., DOI 10.1038/mt.2012.201 Pai P, Rachagani S, Are C, Batra S. Prospects of miRNA-Based Therapy for Pancreatic Cancer. Cdt, 2013, 14:1101–9., DOI 10.2174/ 13894501113149990181 Lindow M, Kauppinen S. Discovering the First microRNA-Targeted Drug. J Cel Biol, 2012, 199:407–12., DOI 10.1083/jcb.201208082 Gurbuz N, Ozpolat B. MicroRNA-based Targeted Therapeutics in Pancreatic Cancer. Anticancer Res, 2019, 39:529–32., DOI 10.21873/anticanres.13144 Passadouro M, Faneca H. Managing Pancreatic Adenocarcinoma: A Special Focus in MicroRNA Gene Therapy. Int J Mol Sci, 2016, 17:718., DOI 10.3390/ ijms17050718 Hu QL, Jiang QY, Jin X, Shen J, Wang K, Li YB, et al. Cationic microRNADelivering Nanovectors with Bifunctional Peptides for Efficient Treatment of PANC-1 Xenograft Model. Biomaterials, 2013, 34:2265–76., DOI 10.1016/j. biomaterials.2012.12.016 Idichi T, Seki N, Kurahara H, Fukuhisa H, Toda H, Shimonosono M, et al. Molecular Pathogenesis of Pancreatic Ductal Adenocarcinoma: Impact of Passenger Strand of Pre-miR-148aon Gene Regulation. Cancer Sci, 2018, 109: 2013–26., DOI 10.1111/cas.13610 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610156 EP 1610167 DI 10.3389/pore.2022.1610156 PG 12 ER PT J AU Kertesz, G Kallay, K Kassa, Cs Zombori, M Bodo, I Kiss, Cs Szegedi, I Krivan, G AF Kertesz, Gabriella Kallay, Krisztian Kassa, Csaba Zombori, Marianna Bodo, Imre Kiss, Csongor Szegedi, Istvan Krivan, Gergely TI Case Report: A Child With Hemophilia A Serves as Donor for Hematopoietic Stem Cell Transplantation to Cure His Brother’s Severe Aplastic Anemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE hemophilia a; severe aplastic anemia (SAA); hematopoietic stem cell transplantation (HSCT); hepatitis associated bone marrow failure (HABMF); children ID hemophilia a; severe aplastic anemia (SAA); hematopoietic stem cell transplantation (HSCT); hepatitis associated bone marrow failure (HABMF); children AB The first-line treatment of severe aplastic anemia is allogeneic hematopoietic stem cell transplantation with a matched sibling donor. However, co-morbidities of the identical donor can make donation difficult. We present a transplantation where in parallel with the patient’s conditioning treatment, the preparation of the donor with severe hemophilia A required a special management with perioperative factor VIII substitution. Donation was successful without complications, and 18 months after transplantation, the patient and his donor are well without any long-term sequelae. To our knowledge, this is the first reported succesfull transplantation with hemophilic child serving as a bone marrow donor. The procedure did not mean a significant risk to donor health, so donors with hemophilia should not be excluded from donation. C1 [Kertesz, Gabriella] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Kallay, Krisztian] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Kassa, Csaba] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Zombori, Marianna] Heim Pal Children’s Hospital, Hemato-Oncology UnitBudapest, Hungary. [Bodo, Imre] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Kiss, Csongor] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-OncologyDebrecen, Hungary. [Szegedi, Istvan] University of Debrecen, Medical and Health Science Center, Department of Pediatric Hematology-OncologyDebrecen, Hungary. [Krivan, Gergely] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. RP Bodo, I (reprint author), Semmelweis University, Department of Internal Medicine and Haematology, Budapest, Hungary. EM bodoimre.md@gmail.com CR Alonso Madrigal C, Dobon Rebollo M, Laredo de la Torre V, Palomera Bernal L, Garcia Gil FA. Liver Transplantation in Hemophilia A and VonWillebrand Disease Type 3: Perioperative Management and Post-Transplant Outcome. Rev Esp Enferm Dig, 2018, 110(8):522–6., DOI 10.17235/reed.2018.5204/2017 Hermans C, Altisent C, Batorova A, Chambost H, De Moerloose P, Karafoulidou A, et al. Replacement Therapy for Invasive Procedures in Patients with Haemophilia: Literature Review, European Survey and Recommendations. Haemophilia, 2009, 15(3):639–58., DOI 10.1111/j.1365- 2516.2008.01950.x Lin PS, Yao YT. Perioperative Management of Hemophilia A Patients Undergoing Cardiac Surgery: A Literature Review of Published Cases. J Cardiothorac Vasc Anesth, 2020, 35:1341., DOI 10.1053/j.jvca.2020.06.074 Ljung RCR, Knobe K. How to Manage Invasive Procedures in Children with Haemophilia. Br J Haematol, 2012, 157(5):519–28., DOI 10.1111/j.1365-2141. 2012.09089.x Garagiola I, Palla R, Peyvandi F. Risk Factors for Inhibitor Development in Severe Hemophilia a. Thromb Res, 2018, 168:20–7., DOI 10.1016/j.thromres. 2018.05.027 Peyvandi F, Cannavo A, Garagiola I, Palla R, Mannucci PM, Rosendaal FR, et al. Timing and Severity of Inhibitor Development in Recombinant versus Plasma-Derived Factor VIII Concentrates: A SIPPET Analysis. J Thromb Haemost, 2018, 16(1):39–43., DOI 10.1111/jth.13888 Gouw SC, van der Bom JG, Ljung R, Escuriola C, Cid AR, Claeyssens-Donadel S, et al. Factor VIII Products and Inhibitor Development in Severe Hemophilia A. N Engl J Med, 2013, 368(3):231–9., DOI 10.1056/nejmoa1208024 Federici AB, Mannucci PM, Stabile F, Rossi G, Piseddu G. Orthotopic Liver Transplantation in a Patient with Severe Hemophilia A: A Life-Saving Treatment for the First Italian Case. Int J Clin Lab Res, 1995, 25(1):44–6., DOI 10.1007/bf02592576 Fahs SA, Hille MT, Shi Q, Weiler H, Montgomery RR. A Conditional Knockout Mouse Model Reveals Endothelial Cells as the Principal and Possibly Exclusive Source of Plasma Factor VIII. Blood, 2014, 123(24): 3706–13., DOI 10.1182/blood-2014-02-555151 Fomin ME, Zhou Y, Beyer AI, Publicover J, Baron JL,Muench MO. Production of Factor VIII by Human Liver Sinusoidal Endothelial Cells Transplanted in Immunodeficient uPA Mice. PLoS One, 2013, 8(10):e77255., DOI 10.1371/ journal.pone.0077255 Tuddenham E. In Search of the Source of Factor VIII. Blood, 2014, 123(24): 3691., DOI 10.1182/blood-2014-05-568857 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610171 EP 1610175 DI 10.3389/pore.2022.1610171 PG 5 ER PT J AU Chen, X Li, M Tang, Y Liang, Q Hua, Ch He, H Song, Y Cheng, H AF Chen, Xianzhen Li, Ma Tang, Yi Liang, Qichang Hua, Chunting He, Huiqin Song, Yinjing Cheng, Hao TI Gene Expression Profile Analysis of Human Epidermal Keratinocytes Expressing Human Papillomavirus Type 8 E7 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE RNA-sequencing; HPV 8E7; immune regulations; metabolic reprogramming; epigenetic modifications ID RNA-sequencing; HPV 8E7; immune regulations; metabolic reprogramming; epigenetic modifications AB Background: Human papillomavirus type 8 (HPV8) has been implicated in the progress of non-melanoma skin cancers and their precursor lesions. The HPV8 E7 oncoprotein plays a key role in the tumorigenesis of HPV-associated cutaneous tumors. However, the exact role of HPV8 E7 in human epidermal carcinogenesis has not been fully elucidated. Methods: To investigate the potential carcinogenic effects of HPV8 E7 on epithelial cells, we used RNA-sequencing technology to analyze the gene expression profile of HPV8 E7- overexpressed normal human epidermal keratinocytes (NHEKs). Results: RNA-sequencing revealed 831 differentially expressed genes (DEGs) between HPV8 E7-expressing NHEKs and control cells, among which, 631 genes were significantly upregulated, and 200 were downregulated. Gene ontology annotation enrichment analysis showed that HPV8 E7 mainly affected the expression of genes associated with protein heterodimerization activity, DNA binding, nucleosomes, and nucleosome assembly. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis revealed that overexpression of HPV8 E7 affected the expression of gene clusters associated with viral carcinogenesis and transcriptional misregulation in cancer and necroptosis signaling pathways that reportedly play crucial roles in HPV infection promotion and cancer progression. We also found the DEGs, such as HKDC1 and TNFAIP3, were associated with epigenetic modifications, immune regulation, and metabolic pathways. Conclusion: Our results demonstrate that the pro-carcinogenic effect of HPV8 expression in epithelial cells may be attributed to the regulatory effect of oncogene E7 on gene expression associated with epigenetic modifications and immune and metabolic status-associated gene expression. Although our data are based on an in vitro experiment, it provides the theoretical evidence that the development of squamous cell carcinoma can be caused by HPV. C1 [Chen, Xianzhen] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Dermatology and VenereologyHangzhou, China. [Li, Ma] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Dermatology and VenereologyHangzhou, China. [Tang, Yi] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Dermatology and VenereologyHangzhou, China. [Liang, Qichang] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Dermatology and VenereologyHangzhou, China. [Hua, Chunting] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Dermatology and VenereologyHangzhou, China. [He, Huiqin] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of GastroenterologyHangzhou, China. [Song, Yinjing] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Dermatology and VenereologyHangzhou, China. [Cheng, Hao] Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Dermatology and VenereologyHangzhou, China. RP Cheng, H (reprint author), Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Department of Dermatology and Venereology, Hangzhou, China. EM chenghao1@zju.edu.cn CR Mitsiogianni M, Amery T, Franco R, Zoumpourlis V, Pappa A, Panayiotidis MI From Chemo-Prevention to Epigenetic Regulation: The Role of Isothiocyanates in Skin Cancer Prevention. Pharmacol Ther, 2018, 190: 187–201., DOI 10.1016/j.pharmthera.2018.06.001 Schiffman M, Doorbar J, Wentzensen N, de Sanjose S, Fakhry C, Monk BJ, et al. Carcinogenic Human Papillomavirus Infection. Nat Rev Dis Primers, 2016, 2:16086., DOI 10.1038/nrdp.2016.86 Smola S. Human Papillomaviruses and Skin Cancer. Adv Exp Med Biol, 2020, 1268:195–209., DOI 10.1007/978-3-030-46227-7_10 Ding X, Lucas T, Marcuzzi GP, Pfister H, Eming SA. Distinct Functions of Epidermal and Myeloid-Derived VEGF-A in Skin Tumorigenesis Mediated by HPV8. Cancer Res, 2015, 75:330–43., DOI 10.1158/0008-5472.CAN-13-3007 Akgul B, Kirschberg M, Storey A, Hufbauer M. Human Papillomavirus Type 8 Oncoproteins E6 and E7 Cooperate in Downregulation of the Cellular Checkpoint Kinase-1. Int J Cancer, 2019, 145:797–806., DOI 10.1002/ijc.32223 Heuser S, Hufbauer M, Steiger J, Marshall J, Sterner-Kock A, Mauch C, et al. The Fibronectin/α3β1 Integrin axis Serves as Molecular Basis for Keratinocyte Invasion Induced by βHPV. Oncogene, 2016, 35:4529–39., DOI 10.1038/onc. 2015.512 Tommasino M. The Biology of Beta Human Papillomaviruses. Virus Res, 2017, 231:128–38., DOI 10.1016/j.virusres.2016.11.013 Pfister H. HPV und Neoplasien der Haut. Hautarzt, 2008, 59:26–30., DOI 10. 1007/s00105-007-1442-6 Cruz-Gregorio A, Aranda-Rivera AK, Ortega-Lozano AJ, Pedraza-Chaverri J, Mendoza-Hoffmann F. Lipid Metabolism and Oxidative Stress in HPVRelated Cancers. Free Radic Biol Med, 2021, 172:226–36., DOI 10.1016/j. freeradbiomed.2021.06.009 Al-Eitan LN, Alghamdi MA, Tarkhan AH, Al-Qarqaz FA. Genome-Wide CpG Island Methylation Profiles of Cutaneous Skin with and without HPV Infection. Int J Mol Sci, 2019, 20:4822., DOI 10.3390/ijms20194822 Georgescu SR, Mitran CI, MitranMI, Caruntu C, Sarbu MI, Matei C, et al. New Insights in the Pathogenesis of HPV Infection and the Associated Carcinogenic Processes: The Role of Chronic Inflammation and Oxidative Stress. J Immunol Res, 2018, 2018:1–10., DOI 10.1155/2018/ 5315816 Liu X, Ma X, Lei Z, Feng H, Wang S, Cen X, et al. Chronic Inflammation- Related HPV: A Driving Force Speeds Oropharyngeal Carcinogenesis. PLoS One, 2015, 10:e0133681., DOI 10.1371/journal.pone.0133681 Arizmendi-Izazaga A, Navarro-Tito N, Jimenez-Wences H, Mendoza-Catalan MA, Martinez-Carrillo DN, Zacapala-Gomez AE, et al. Metabolic Reprogramming in Cancer: Role of HPV 16 Variants. Pathogens, 2021, 10: 347., DOI 10.3390/pathogens10030347 Wang Y, Mashock M, Tong Z, Mu X, Chen H, Zhou X, et al. Changing Technologies of RNA Sequencing and Their Applications in Clinical Oncology. Front Oncol, 2020, 10:447., DOI 10.3389/fonc.2020.00447 Zheng H, Zou Z, Wu X, Xu Y, Zhu J, Zhou Q, et al. HPV11E7 Inhibits IMQInduced Chemokine and colony-stimulating Factor Production in Keratinocytes. Gene, 2020, 760:145003., DOI 10.1016/j.gene.2020.145003 Zhang J, Zhang C, Sun P, Huang M, Fan M, Liu M. RNA-sequencing and Pathway Analysis Reveal Alteration of Hepatic Steroid Biosynthesis and Retinol Metabolism by Tributyltin Exposure in Male Rare Minnow, Gobiocypris Rarus). Aquat Toxicol, 2017, 188:109–18., DOI 10.1016/j. aquatox.2017.03.015 Wang T, Zhang Y, Guo Y, Zhang X, Yang H, Tian X, et al. RNA-sequence Reveals Differentially Expressed Genes Affecting the Crested Trait of Wumeng Crested Chicken. Poult Sci, 2021, 100:101357., DOI 10.1016/j.psj.2021.101357 Shamseddine AA, Burman B, Lee NY, Zamarin D, Riaz N. Tumor Immunity and Immunotherapy for HPV-Related Cancers. Cancer Discov, 2021, 11: 1896–912., DOI 10.1158/2159-8290.CD-20-1760 Saraiya M, Unger ER, Thompson TD, Lynch CF, Hernandez BY, Lyu CW, et al. US Assessment of HPV Types in Cancers: Implications for Current and 9- valent HPV Vaccines. J Natl Cancer Inst, 2015, 107:djv086., DOI 10.1093/jnci/ djv086 Roden RBS, Stern PL. Opportunities and Challenges for Human Papillomavirus Vaccination in Cancer. Nat Rev Cancer, 2018, 18:240–54., DOI 10.1038/nrc.2018.13 Pal A, Kundu R. Human Papillomavirus E6 and E7: The Cervical Cancer Hallmarks and Targets for Therapy. Front Microbiol, 2019, 10:3116., DOI 10. 3389/fmicb.2019.03116 McLaughlin-Drubin ME, Munger K. The Human Papillomavirus E7 Oncoprotein. Virology, 2009, 384:335–44., DOI 10.1016/j.virol.2008.10.006 Kyrgiou M, Moscicki A-B. Vaginal Microbiome and Cervical Cancer. Semin Cancer Biol, 2022)., DOI 10.1016/j.semcancer.2022.03.005 Hufbauer M, Cooke J, van der Horst GTJ, Pfister H, Storey A, Akgul B. Human Papillomavirus Mediated Inhibition of DNA Damage Sensing and Repair Drives Skin Carcinogenesis. Mol Cancer, 2015, 14:183., DOI 10.1186/s12943-015-0453-7 Wendel SO, Wallace NA. Loss of Genome Fidelity: Beta HPVs and the DNA Damage Response. Front Microbiol, 2017, 8:2250., DOI 10.3389/fmicb.2017. 02250 Kirschberg M, Syed AS, Donmez HG, Heuser S, Wilbrand-Hennes A, Alonso A, et al. Novel Insights into Cellular Changes in HPV8-E7 Positive Keratinocytes: A Transcriptomic and Proteomic Analysis. Front Microbiol, 2021, 12:672201., DOI 10.3389/fmicb.2021.672201 Haedicke J, Iftner T. Human Papillomaviruses and Cancer. Radiother Oncol, 2013, 108:397–402., DOI 10.1016/j.radonc.2013.06.004 Oswald E, Reinz E, Voit R, Aubin F, Alonso A, Auvinen E. Human Papillomavirus Type 8 E7 Protein Binds Nuclear Myosin 1c and Downregulates the Expression of Pre-rRNA. Virus Genes, 2017, 53:807–13., DOI 10.1007/s11262-017-1491-6 The Gene Ontology C. The Gene Ontology Resource: 20 Years and Still Going Strong. Nucleic Acids Res, 2019, 47:D330–D338., DOI 10.1093/nar/gky1055 Sultana S, Zarreen F, Chakraborty S. Insights into the Roles of Histone Chaperones in Nucleosome Assembly and Disassembly in Virus Infection. Virus Res, 2021, 297:198395., DOI 10.1016/j.virusres.2021.198395 Pismataro MC, Felicetti T, Bertagnin C, Nizi MG, Bonomini A, Barreca ML, et al. 1,2,4-Triazolo[1,5-a]pyrimidines: Efficient One-step Synthesis and Functionalization as Influenza Polymerase PA-PB1 Interaction Disruptors. Eur J Med Chem, 2021, 221:113494., DOI 10.1016/j.ejmech. 2021.113494 Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. KEGG: Integrating Viruses and Cellular Organisms. Nucleic Acids Res, 2021, 49: D545–D551., DOI 10.1093/nar/gkaa970 Akgul B, Ghali L, Davies D, Pfister H, Leigh IM, Storey A. HPV8 Early Genes Modulate Differentiation and Cell Cycle of Primary Human Adult Keratinocytes. Exp Dermatol, 2007, 16:590–9., DOI 10.1111/j.1600-0625. 2007.00569.x Li L, Yu S, Zang C. Low Necroptosis Process Predicts Poor Treatment Outcome of Human Papillomavirus Positive Cervical Cancers by Decreasing Tumor-Associated Macrophages M1 Polarization. Gynecol Obstet Invest, 2018, 83:259–67., DOI 10.1159/000487434 Kirschberg M, Heuser S, Marcuzzi GP, Hufbauer M, Seeger JM, Dukic A, et al. ATP Synthase Modulation Leads to an Increase of Spare Respiratory Capacity in HPV Associated Cancers. Sci Rep, 2020, 10:17339., DOI 10.1038/s41598-020- 74311-6 Arii J, Fukui A, Shimanaka Y, Kono N, Arai H, Maruzuru Y, et al. Role of Phosphatidylethanolamine Biosynthesis in Herpes Simplex Virus 1- Infected Cells in Progeny Virus Morphogenesis in the Cytoplasm and in Viral Pathogenicity In Vivo. J Virol, 2020, 94:e01572., DOI 10.1128/JVI. 01572-20 Guardado-Calvo P, Atkovska K, Jeffers SA, Grau N, Backovic M, Perez-Vargas J, et al. A Glycerophospholipid-specific Pocket in the RVFV Class II Fusion Protein Drives Target Membrane Insertion. Science, 2017, 358:663–7., DOI 10. 1126/science.aal2712 Yang Y-C, Chang T-Y, Chen T-C, Chang S-C, Chen W-F, Chan H-W, et al. Genetic Polymorphisms in the ITPKC Gene and Cervical Squamous Cell Carcinoma Risk. Cancer Immunol Immunother, 2012, 61:2153–9., DOI 10. 1007/s00262-012-1280-y Hsu C-H, Peng K-L, Jhang H-C, Lin C-H, Wu S-Y, Chiang C-M, et al. The HPV E6 Oncoprotein Targets Histone Methyltransferases for Modulating Specific Gene Transcription. Oncogene, 2012, 31:2335–49., DOI 10.1038/onc. 2011.415 Di Domenico M, Giovane G, Kouidhi S, Iorio R, Romano M, De Francesco F, et al. HPV Epigenetic Mechanisms Related to Oropharyngeal and Cervix Cancers. Cancer Biol Ther, 2018, 19:850–7., DOI 10.1080/15384047.2017. 1310349 Lo Cigno I, Calati F, Borgogna C, Zevini A, Albertini S, Martuscelli L, et al. Human Papillomavirus E7 Oncoprotein Subverts Host Innate Immunity via SUV39H1-Mediated Epigenetic Silencing of Immune Sensor Genes. J Virol, 2020, 94:e01812–e01819., DOI 10.1128/JVI.01812-19 Degli Esposti D, Sklias A, Lima SC, Beghelli-de la Forest Divonne S, Cahais V, Fernandez-Jimenez N, et al. Unique DNA Methylation Signature in HPVPositiveHead and Neck Squamous Cell Carcinomas. GenomeMed, 2017, 9:33., DOI 10.1186/s13073-017-0419-z Roy M,Mukherjee S. Reversal of Resistance towards Cisplatin by Curcumin in Cervical Cancer Cells. Asian Pac J Cancer Prev, 2014, 15:1403–10., DOI 10. 7314/apjcp.2014.15.3.1403 Lourenco de Freitas N, Deberaldini MG, Gomes D, Pavan AR, Sousa A, Dos Santos JL, et al. Histone Deacetylase Inhibitors as Therapeutic Interventions on Cervical Cancer Induced by Human Papillomavirus. Front Cel Dev. Biol., 2020, 8:592868., DOI 10.3389/fcell.2020.592868 Al-Eitan LN, Alghamdi MA, Tarkhan AH, Al-Qarqaz FA. Epigenome-wide Analysis of Common Warts Reveals Aberrant Promoter Methylation. Int J Med Sci, 2020, 17:191–206., DOI 10.7150/ijms.39261 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610176 EP 1610186 DI 10.3389/pore.2022.1610176 PG 11 ER PT J TI Chronometric Administration of Cyclophosphamide and a Double-Stranded DNA-Mix at Interstrand Crosslinks Repair Timing, Called “Karanahan” Therapy, Is Highly Efficient in a Weakly Immunogenic Lewis Carcinoma Model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer stem cells; Karanahan approach; Lewis lung carcinoma; committed tumor cells; tumor growth site; cytostatic agent; dsDNA; interstrand crosslink repair ID cancer stem cells; Karanahan approach; Lewis lung carcinoma; committed tumor cells; tumor growth site; cytostatic agent; dsDNA; interstrand crosslink repair AB Background and Aims: A new technology based on the chronometric administration of cyclophosphamide and complex composite double-stranded DNA-based compound, which is scheduled in strict dependence on interstrand crosslinks repair timing, and named “Karanahan”, has been developed. Being applied, this technology results in the eradication of tumor-initiating stem cells and full-scale apoptosis of committed tumor cells. In the present study, the efficacy of this novel approach has been estimated in the model of Lewis carcinoma. Methods: To determine the basic indicative parameters for the approach, the duration of DNA repair in tumor cells, as well as their distribution along the cell cycle, have been assessed. Injections were done into one or both tumors in femoral region of the engrafted mice in accordance with the developed regimen. Four series of experiments were carried out at different periods of time. The content of poorly differentiated CD34+/TAMRA+ cells in the bone marrow and peripheral blood has been determined. Immunostaining followed by the flow cytometry was used to analyze the subpopulations of immune cells. Results: The high antitumor efficacy of the new technology against the developed experimental Lewis carcinoma was shown. It was found that the therapy efficacy depended on the number of tumor growth sites, seasonal and annual peculiarities. In some experiments, a long-term remission has been reached in 70% of animals with a single tumor and in 60% with two tumors. In mice with two developed grafts, mobilization capabilities of both poorly differentiated hematopoietic cells of the host and tumor stemlike cells decrease significantly. Being applied, this new technology was shown to activate a specific immune response. There is an increase in the number of NK cell populations in the blood, tumor, and spleen, killer T cells and T helper cells in the tumor and spleen, CD11b+Ly-6C+ and CD11b+Ly-6G+ cells in the tumor. A population of mature dendritic cells is found in the tumor. Conclusion: The performed experiments indicate the efficacy of the Karanahan approach against incurable Lewis carcinoma. Thus, the discussed therapy is a new approach for treating experimental neoplasms, which has a potential as a personalized anti-tumor therapeutic approach in humans. CR Shafi AA, Knudsen KE. Cancer and the Circadian Clock. Cancer Res, 2019, 79(15):3806–14., DOI 10.1158/0008-5472.CAN-19-0566 Adam D. Emerging Science of Chronotherapy Offers Big Opportunities to Optimize Drug Delivery. Proc Natl Acad Sci U.S.A, 2019, 116(44):21957–9., DOI 10.1073/pnas.1916118116 Chen DS, Mellman I. Elements of Cancer Immunity and the Cancer-Immune Set point. Nature, 2017, 541(7637):321–30., DOI 10.1038/nature21349 Sagiv-Barfi I, Czerwinski DK, Levy S, Alam IS, Mayer AT, Gambhir SS, et al. Eradication of Spontaneous Malignancy by Local Immunotherapy. Sci Transl Med, 2018, 10(426):eaan4488., DOI 10.1126/scitranslmed.aan4488 Simsek C, Esin E, Yalcin S. Metronomic Chemotherapy: A Systematic Review of the Literature and Clinical Experience. J Oncol, 2019, 2019:1–31., DOI 10. 1155/2019/5483791 Cazzaniga ME, Cordani N, Capici S, Cogliati V, Riva F, Cerrito MG. Metronomic Chemotherapy. Cancers, 2021, 1313(9):22362236., DOI 10.3390/ CANCERS13092236 Munoz R, Girotti A, Hileeto D, Arias FJ. Metronomic Anti-cancer Therapy: A Multimodal Therapy Governed by the Tumor Microenvironment. Cancers, 2021, 1313(21):54145414., DOI 10.3390/ CANCERS13215414 Leong WI, Ames RY, Haverkamp JM, Torres L, Kline J, Bans A, et al. Low-dose Metronomic Cyclophosphamide Complements the Actions of an Intratumoral C-Class CpG TLR9 Agonist to Potentiate Innate Immunity and Drive Potent T Cell-Mediated Anti-tumor Responses. Oncotarget, 2019, 10(68):7220–37., DOI 10.18632/ONCOTARGET. 27322 Dolgova EV, Proskurina AS, Nikolin VP, Popova NA, Alyamkina EA, Orishchenko KE, et al. "Delayed Death" Phenomenon: A Synergistic Action of Cyclophosphamide and Exogenous DNA. Gene, 2012, 495(2):134–45., DOI 10.1016/j.gene.2011.12.032 Dolgova EV, Efremov YR, Orishchenko KE, Andrushkevich OM, Alyamkina EA, Proskurina AS, et al. Delivery and Processing of Exogenous Double- Stranded DNA in Mouse CD34+ Hematopoietic Progenitor Cells and Their Cell Cycle Changes upon Combined Treatment with Cyclophosphamide and Double-Stranded DNA. Gene, 2013, 528(2):74–83., DOI 10.1016/j.gene.2013. 06.058 Likhacheva AS, Nikolin VP, Popova NA, Dubatolova TD, Strunkin DN, Rogachev VA, et al. Integration of Human DNA Fragments into the Cell Genomes of Certain Tissues from Adult Mice Treated with Cytostatic Cyclophosphamide in Combination with Human DNA. Gene Ther Mol Biol, 2007, 11(2):185–202. Likhacheva AS, Nikolin VP, Popova NA, Rogachev VA, ProkhorovichMA, Sebeleva TE, et al. Exogenous DNA Can Be Captured by Stem Cells and Be Involved in Their rescue from Death after Lethal-Dose γ-radiation. Gene Ther Mol Biol, 2007, 11(2):305–14. Dolgova EV, Nikolin VP, Popova NA, Proskurina AS, Orishenko KE, Alyamkina EA, et al. Internalization of Exogenous DNA into Internal Compartments of Murine Bone Marrow Cells. Russ J Genet Appl Res, 2012, 16(2):397–414., DOI 10.1134/s2079059712060056 Dolgova EV, Nikolin VP, Popova NA, Proskurina AS, Orishchenko KE, Alyamkina EA, et al. Pathological Changes in Mice Treated with Cyclophosphamide and Exogenous DNA. Russ J Genet Appl Res, 2013, 17(1):129–46., DOI 10.1134/s2079059713040035 Dolgova EV, Alyamkina EA, Efremov YR, Nikolin VP, Popova NA, Tyrinova TV, et al. Identification of Cancer Stem Cells and a Strategy for Their Elimination. Cancer Biol Ther, 2014, 15(10):1378–94., DOI 10.4161/cbt.29854 Dolgova EV, Mishinov SV, Proskurina AS, Potter EA, Efremov YR, Bayborodin SI, et al. Novel Cancer Stem Marker and its Applicability for Grading Primary Human Gliomas. Technol Cancer Res Treat, 2018, 17: 153303461775381., DOI 10.1177/1533034617753812 Potter EA, Dolgova EV, Proskurina AS, Efremov YR, Minkevich AM, Rozanov AS, et al. Gene Expression Profiling of Tumor-Initiating Stem Cells from Mouse Krebs-2 Carcinoma Using a Novel Marker of Poorly Differentiated Cells. Oncotarget, 2017, 8(6):9425–41., DOI 10.18632/ONCOTARGET.14116 Dolgova EV, Evdokimov AN, Proskurina AS, Efremov YR, Bayborodin SI, Potter EA, et al. Double-Stranded DNA Fragments Bearing Unrepairable Lesions and Their Internalization into Mouse Krebs-2 Carcinoma Cells. Nucleic Acid Ther, 2019, 29(5):278–90., DOI 10.1089/NAT.2019.0786 Ahrens TD, Bang-Christensen SR, Jorgensen AM, Loppke C, Spliid CB, Sand NT, et al. The Role of Proteoglycans in Cancer Metastasis and Circulating Tumor Cell Analysis. Front Cel Dev. Biol., 2020, 8:749., DOI 10.3389/FCELL. 2020.00749/BIBTEX Barnes JM, Kaushik S, Bainer RO, Sa JK, Woods EC, Kai F, et al. A Tension- Mediated Glycocalyx-Integrin Feedback Loop Promotes Mesenchymal-like Glioblastoma. Nat Cel Biol, 2018, 20(10):1203–14., DOI 10.1038/S41556-018- 0183-3 Knelson EH, Nee JC, Blobe GC. Heparan Sulfate Signaling in Cancer. Trends Biochem Sci, 2014, 39(6):277–88., DOI 10.1016/J.TIBS.2014.03.001 Kuo JC-H, Gandhi JG, Zia RN, Paszek MJ. Physical Biology of the Cancer Cell Glycocalyx. Nat Phys, 2018, 14(7):658–69., DOI 10.1038/S41567-018-0186-9 Mallard BW, Tiralongo J. Cancer Stem Cell Marker Glycosylation: Nature, Function and Significance. Glycoconj J, 2017, 34(4):441–52., DOI 10.1007/ s10719-017-9780-9 Mitchell MJ, King MR. Physical Biology in Cancer. 3. The Role of Cell Glycocalyx in Vascular Transport of Circulating Tumor Cells. Am J Physiology-Cell Physiol, 2014, 306(2):C89–C97., DOI 10.1152/AJPCELL. 00285.2013 Paszek MJ, Dufort CC, Rossier O, Bainer R, Mouw JK, Godula K, et al. The Cancer Glycocalyx Mechanically Primes Integrin-Mediated Growth and Survival. Nature, 2014, 511(7509):319–25., DOI 10.1038/NATURE13535 Vitale D, Kumar Katakam S, Greve B, Jang B, Oh ES, Alaniz L, et al. Proteoglycans and Glycosaminoglycans as Regulators of Cancer Stem Cell Function and Therapeutic Resistance. FEBS J, 2019, 286(15):2870–82., DOI 10. 1111/FEBS.14967 Alyamkina EA, Nikolin VP, Popova NA, Minkevich AM, Kozel AV, Dolgova EV, et al. Combination of Cyclophosphamide and Double-Stranded DNA Demonstrates Synergistic Toxicity against Established Xenografts. Cancer Cel Int, 2015, 15(1):32., DOI 10.1186/s12935-015-0180-6 Dolgova EV, Efremov YR, Taranov OS, Potter EA, Nikolin VP, Popova NA, et al. Comparative Analysis of Pathologic Processes Developing in Mice Housed in SPF vs Non-SPF Conditions and Treated with Cyclophosphamide and dsDNA Preparation. Pathol - Res Pract, 2015, 211(10):754–8., DOI 10.1016/J.PRP.2015.07.002 Potter EA, Dolgova EV, Minkevich AM, Efremov YR, Taranov OS, Omigov VV, et al. Therapeutic Effects of Cyclophosphamide, dsDNA Preparations and Combinations Thereof against Krebs-2 Ascites Cancer Cells and Various Cancer Transplants. Vestn Vogis, 2016, 20(1):96–107., DOI 10.18699/vj15.116 Potter EA, Dolgova EV, Minkevich AM, Nikolin VP, Popova NA, Efremov YR, et al. Analysis of Different Therapeutic Schemes Combining Cyclophosphamide and Doublestranded DNA Preparation for Eradication of Krebs-2 Primary Ascites in Mice. Vestn Vogis, 2016, 20(1):108–24., DOI 10. 18699/VJ15.117 Potter EA, Dolgova EV, Minkevich AM, Nikolin VP, Popova NA, Efremov YR, et al. Eradication of Krebs-2 Primary Ascites via a Single-Injection Regimen of Cyclophosphamide and Double-Stranded DNA. Vestn Vogis, 2016, 20(5): 716–22., DOI 10.18699/vj16.161 Potter EA, Dolgova EV, Proskurina AS, Efremov YR, Taranov OS, Nikolin VP, et al. Development of the Therapeutic Regimen Based on the Synergistic Activity of Cyclophosphamide and Double-Stranded DNA Preparation Which Results in Complete Cure of Mice Engrafted with Krebs-2 Ascites. Vestn Vogis, 2016, 20(5):723–35., DOI 10.18699/vj16.162 Potter EA, Dolgova EV, Proskurina AS,Minkevich AM, Efremov YR, Taranov OS, et al. A Strategy to Eradicate Well-Developed Krebs-2 Ascites in Mice. Oncotarget, 2016, 7(10):11580–94., DOI 10.18632/oncotarget.7311 Dolgova EV, Likhacheva AS, Orishchenko KE, Alyamkina EA, Bogachev SS, Shurdov MA. Repair of Interstrand Crosslinks in a DNA Molecule. Vavilov J Genet Breed, 2010, 14(2):332–56. Dolgova EV, Potter EA, Proskurina AS, Minkevich AM, Chernych ER,Ostanin AA, et al. Properties of Internalization Factors Contributing to the Uptake of Extracellular DNA into Tumor-Initiating Stem Cells of Mouse Krebs-2 Cell Line. Stem Cel Res Ther, 2016, 7(1):76., DOI 10.1186/s13287-016-0338-8 Potter EA, Proskurina AS, Ritter GS, Dolgova EV, Nikolin VP, Popova NA, et al. Efficacy of a New Cancer Treatment Strategy Based on Eradication of Tumor-Initiating Stem Cells in a Mouse Model of Krebs-2 Solid Adenocarcinoma. Oncotarget, 2018, 9(47):28486–99., DOI 10.18632/ oncotarget.25503 Kisaretova PE, Kirikovich SS, Ritter GS, Efremov YR, Taranov OS, Dubatolova TD, et al. Approbation of the Cancer Treatment Approach Based on the Eradication of TAMRA+ Cancer Stem Cells in a Model of Murine Cyclophosphamide Resistant Lymphosarcoma. Anticancer Res, 2020, 40(2): 795–805., DOI 10.21873/anticanres.14011 Ruzanova VS, Proskurina AS, Ritter GS, Efremov YR, Nikolin VP, Popova NA, et al. Experimental Comparison of the In Vivo Efficacy of Two Novel Anticancer Therapies. Anticancer Res, 2021, 41(7):3371–87., DOI 10.21873/ anticanres.15125 Dolgova EV, Andrushkevich OM, Kisaretova PE, Proskurina AS, Ritter GS, Dubatolova TD, et al. Efficacy of the New Therapeutic Approach in Curing Malignant Neoplasms on the Model of Human Glioblastoma. Cancer Biol Med, 2021, 18(3):910–30., DOI 10.20892/j.issn.2095-3941.2020.0511 Proskurina AS, Ruzanova VS, Ostanin AA, Chernykh ER, Bogachev SS. Theoretical Premises of a “Three in One” Therapeutic Approach to Treat Immunogenic and Nonimmunogenic Cancers: a Narrative Review. Transl Cancer Res TCR, 2021, 10(11):4958–72., DOI 10.21037/TCR-21-919 Mayo JG. Biologic Characterization of the Subcutaneously Implanted Lewis Lung Tumor. Cancer Chemother Rep 2, 1972, 3(1):325–30. Moller P. The Comet Assay: Ready for 30 More Years. Mutagenesis, 2018, 33(1):1–7., DOI 10.1093/mutage/gex046 Olive PL, Banath JP, Durand RE, Banath JP. Heterogeneity in Radiation- Induced DNA Damage and Repair in Tumor and Normal Cells Measured Using the "Comet" Assay. Radiat Res, 1990, 122(1):86., DOI 10.2307/3577587 Bogachev S, Dolgova E, Potter E, Proskurina A, Ritter G, Nikolin V, et al. A Method for the Treatment of Cancer. Russia; RU 2662354 C1, 2018). Ishikawa M, Inoue T, Inui T, Kuchiike D, Kubo K, Uto Y, et al. A Novel Assay System for Macrophage-Activating Factor Activity Using a Human U937 Cell Line. Anticancer Res, 2014, 34(8):4577–81. Stockert JC, Blazquez-Castro A, Canete M, Horobin RW, Villanueva A. MTT Assay for Cell Viability: Intracellular Localization of the Formazan Product Is in Lipid Droplets. Acta Histochem, 2012, 114(8):785–96., DOI 10.1016/j.acthis. 2012.01.006 Dolgova Y, Potter Y, Proskurina A, Nikolin V, Popova N, Romanenko M, et al. Evaluating the Effectiveness of the Tumor-Initiating Stem Cells Eradication Strategy on the Example of Human Glioblastoma Cell Line U87. Probl Oncol, 2019, 65(6):904–19., DOI 10.37469/0507-3758-2019-65-6-904-919 Dolgova EV, Proskurina AS, Potter EA, Tyrinova TV, Taranov OS, Efremov YR, et al. Evaluation of a Strategy for Tumor-Initiating Stem Cell Eradication in Primary Human Glioblastoma Cultures as a Model. Vestn Vogis, 2018, 22(7):825–36., DOI 10.18699/VJ18.31-o Potter Y, Ritter G, Bogachev S, Kolchanov N, Sidorov S, Chernykh Y, et al. Evaluating the Efficiency of the Tumor-Initiating Stem Cells Eradication Strategy on the Example of Ascite Form of Mouse Gepatocarcinoma G-29. Probl Oncol, 2018, 64(6):818–29., DOI 10.37469/0507-3758-2018-64-6-818-829 Alyamkina EA, Dolgova EV, Likhacheva AS, Rogachev VA, Sebeleva TE, Nikolin VP, et al. Exogenous Allogenic Fragmented Double-Stranded DNA Is Internalized into Human Dendritic Cells and Enhances Their Allostimulatory Activity. Cell Immunol, 2010, 262(2):120–6., DOI 10.1016/j.cellimm.2010. 01.005 Alyamkina EA, Leplina OY, Ostanin AA, Chernykh ER, Nikolin VP, Popova NA, et al. Effects of Human Exogenous DNA on Production of Perforin- Containing CD8+ Cytotoxic Lymphocytes in Laboratory Setting and Clinical Practice. Cell Immunol, 2012, 276(1–2):59–66., DOI 10.1016/j.cellimm.2012. 04.004 Alyamkina EA, Leplina OY, Sakhno LV, Chernykh ER, Ostanin AA, Efremov YR, et al. Effect of Double-Stranded DNA on Maturation of Dendritic Cells In Vitro. Cell Immunol, 2010, 266(1):46–51., DOI 10.1016/j. cellimm.2010.08.011 Orishchenko KE, Ryzhikova SL, Druzhinina YG, Ryabicheva TG, Varaksin NA, Alyamkina EA, et al. Effect of Human Double-Stranded DNA Preparation on the Production of Cytokines by Dendritic Cells and Peripheral Blood Cells from Relatively Healthy Donors. Cancer Ther, 2013, 8:191–205. Law AMK, Valdes-Mora F, Gallego-Ortega D. Myeloid-derived Suppressor Cells as a Therapeutic Target for Cancer. Cells, 2020, 9(3):561., DOI 10.3390/ cells9030561 Ostrand-Rosenberg S, Fenselau C. Myeloid-Derived Suppressor Cells: Immune-Suppressive Cells that Impair Antitumor Immunity and Are Sculpted by Their Environment. J.I., 2018, 200(2):422–31., DOI 10.4049/ jimmunol.1701019 Wang H, Shi G, LiM, Fan H, Ma H, Sheng L. Correlation of IL-1 and HB-EGF with Endometrial Receptivity. Exp Ther Med, 2018, 16(6):5130–6., DOI 10. 3892/etm.2018.6840 Zheng X, Turkowski K, Mora J, Brune B, Seeger W, Weigert A, et al. Redirecting Tumor-Associated Macrophages to Become Tumoricidal Effectors as a Novel Strategy for Cancer Therapy. Oncotarget, 2017, 8(29): 48436–52., DOI 10.18632/oncotarget.17061 Jain A, Pasare C. Innate Control of Adaptive Immunity: beyond the Three- Signal Paradigm. J.I., 2017, 198(10):3791–800., DOI 10.4049/jimmunol. 1602000 Sonnenberg GF, Hepworth MR. Functional Interactions between Innate Lymphoid Cells and Adaptive Immunity. Nat Rev Immunol, 2019, 19(10): 599–613., DOI 10.1038/s41577-019-0194-8 Proskurina AS, Ruzanova VS, Tyrinova TV, Strunkin DN, Kirikovich SS, Ritter GS, et al. The Basis of Anticancer Immunity Mechanism Induced by In Situ Vaccination. J Sib Fed Univ Biol, 2020, 2020(3):235–69., DOI 10.17516/1997- 1389-0326 Abel AM, Yang C, Thakar MS, Malarkannan S. Natural Killer Cells: Development, Maturation, and Clinical Utilization. Front Immunol, 2018, 9(AUG):1–23., DOI 10.3389/fimmu.2018.01869 Sharma A, Rudra D. Emerging Functions of Regulatory T Cells in Tissue Homeostasis. Front Immunol, 2018, 9(APR):883., DOI 10.3389/fimmu.2018. 00883 Fuhr L, Abreu M, Pett P, Relogio A. Circadian Systems Biology: When Time Matters. Comput Struct Biotechnol J, 2015, 13:417–26., DOI 10.1016/j.csbj.2015. 07.001 Magnusson A, Boivin D. Seasonal Affective Disorder: An Overview. Chronobiology Int, 2003, 20(2):189–207., DOI 10.1081/CBI-120019310 Ortiz-Tudela E, Innominato PF, Rol MA, Levi F, Madrid JA. Relevance of Internal Time and Circadian Robustness for Cancer Patients. BMC Cancer, 2016, 16(1)., DOI 10.1186/s12885-016-2319-9 Smolensky MH, Hermida RC, Reinberg A, Sackett-Lundeen L, Portaluppi F. Circadian Disruption: New Clinical Perspective of Disease Pathology and Basis for Chronotherapeutic Intervention. Chronobiology Int, 2016, 33(8):1101–19., DOI 10.1080/07420528.2016.1184678 Larson TA, Lent KL, Bammler TK, MacDonald JW, Wood WE, Caras ML, et al. Network Analysis of microRNA and mRNA Seasonal Dynamics in a Highly Plastic Sensorimotor Neural Circuit. BMC Genomics, 2015, 16(1)., DOI 10.1186/s12864-015-2175-z Longpre KM, Kinstlinger NS, Mead EA, Wang Y, Thekkumthala AP, Carreno KA, et al. Seasonal Variation of Urinary microRNA Expression in Male Goats, Capra hircus, as Assessed by Next Generation Sequencing. Gen Comp Endocrinol, 2014, 199:1–15., DOI 10.1016/j.ygcen.2014.01.002 Novoselova EG, Kolaeva SG, Makar VR, Agaphonova TA. Production of Tumor Necrosis Factor in Cells of Hibernating Ground Squirrels Citellus Undulatus during Annual Cycle. Life Sci, 2000, 67(9):1073–80., DOI 10.1016/ S0024-3205(00)00698-6 Moan JE, Lagunova Z, Bruland O, Juzeniene A. Seasonal Variations of Cancer Incidence and Prognosis. Dermato-Endocrinology, 2010, 2(2):55–7., DOI 10. 4161/derm.2.2.12664 Oh E-Y, Wood PA, Du-Quiton J, Hrushesky WJM. Seasonal Modulation of post-resection Breast Cancer Metastasis. Breast Cancer Res Treat, 2008, 111(2):219–28., DOI 10.1007/s10549-007-9780-8 Holdaway I, Mason B, Gibbs E, Rajasoorya C, Hopkins K. Seasonal Changes in Serum Melatonin in Women with Previous Breast Cancer. Br J Cancer, 1991, 64(1):149–53., DOI 10.1038/bjc.1991.259 Paradiso A, Serio G, Fanelli M, Mangia A, Cellamare G, Schittulli F. Predictability of Monthly and Yearly Rhythms of Breast Cancer Features. Breast Cancer Res Treat, 2001, 67(1):41–9., DOI 10.1023/A:1010658804640 Blottner S, Hingst O, Meyer HHD. Inverse Relationship between Testicular Proliferation and Apoptosis in Mammalian Seasonal Breeders. Theriogenology, 1995, 44(3):321–8., DOI 10.1016/0093-691x(95)00187-d Hazlerigg DG, Lincoln GA. Hypothesis. J Biol Rhythms, 2011, 26(6):471–85., DOI 10.1177/0748730411420812 Hrushesky W, Rich IN. Measuring Stem Cell Circadian Rhythm. Methods Mol Biol, 2014, 1235:81–95., DOI 10.1007/978-1-4939-1785-3_8 Margotta V. Interaction between Autumnal Temperature-Photoperiod and Experimentally Induced Transient Cold Shock Influences Proliferative Activity in the Brain of an Adult Terrestrial Heterothermic Vertebrate, Rana Bergeri, Gunther, 1986). Ital J Anat Embryol, 2015, 120(3):192–200., DOI 10.13128/IJAE-17816 Margotta V. Relationships between Seasonal thermal Variations and Cell Proliferation in Heterothermic Vertebrates, as Revealed by PCNA Expression in the Brain of Adult Podarcis sicula. Ital J Anat Embryol, 2014, 119(1):29–37., DOI 10.13128/IJAE-14637 Migaud M, Batailler M, Pillon D, Franceschini I, Malpaux B. Seasonal Changes in Cell Proliferation in the Adult Sheep Brain and Pars Tuberalis. J Biol Rhythms, 2011, 26(6):486–96., DOI 10.1177/0748730411420062 Zaldibar B, Cancio I, Marigomez I. Epithelial Cell Renewal in the Digestive Gland and Stomach of Mussels: Season, Age and Tidal Regime Related Variations. Histol Histopathol, 2008, 23(3):281–90., DOI 10.14670/HH-23.281 Ballesta A, Innominato PF, Dallmann R, Rand DA, Levi FA. Systems Chronotherapeutics. Pharmacol Rev, 2017, 69(2):161–99., DOI 10.1124/pr. 116.013441 Kiessling S, Beaulieu-Laroche L, Blum ID, Landgraf D, Welsh DK, Storch K-F, et al. Enhancing Circadian Clock Function in Cancer Cells Inhibits Tumor Growth. BMC Biol, 2017, 15(1):13., DOI 10.1186/s12915-017-0349-7 Pierre K, Rao RT, Hartmanshenn C, Androulakis IP.Modeling the Influence of Seasonal Differences in the HPA axis on Synchronization of the Circadian Clock and Cell Cycle. Endocrinology, 2018, 159(4):1808–26., DOI 10.1210/en. 2017-03226 Shostak A. Circadian Clock, Cell Division, and Cancer: from Molecules to Organism. Ijms, 2017, 18(4):873., DOI 10.3390/ijms18040873 Sulli G, Lam MTY, Panda S. Interplay between Circadian Clock and Cancer: New Frontiers for Cancer Treatment. Trends Cancer, 2019, 5(8):475–94., DOI 10.1016/j.trecan.2019.07.002 Fadini GP, Mehta A, Dhindsa DS, Bonora BM, Sreejit G, Nagareddy P, et al. Circulating Stem Cells and Cardiovascular Outcomes: from Basic Science to the Clinic. Eur Heart J, 2020, 41(44):4271–82., DOI 10.1093/eurheartj/ehz923 Labusca L, Herea DD, Mashayekhi K. Stem Cells as Delivery Vehicles for Regenerative Medicine-Challenges and Perspectives. Wjsc, 2018, 10(5):43–56., DOI 10.4252/wjsc.v10.i5.43 Ratajczak MZ. A Novel View of the Adult Bone Marrow Stem Cell Hierarchy and Stem Cell Trafficking. Leukemia, 2015, 29(4):776–82., DOI 10.1038/leu. 2014.346 Karpova D, Rettig MP, DiPersio JF. Mobilized Peripheral Blood: an Updated Perspective. F1000Res, 2019, 8:2125., DOI 10.12688/ f1000research.21129.1 Schreier S, TriampoW. The Blood Circulating Rare Cell Population. What Is it and what Is it Good for? Cells, 2020, 9(4):790., DOI 10.3390/ cells9040790 Ming Y, Li Y, Xing H, Luo M, Li Z, Chen J, et al. Circulating Tumor Cells: From Theory to Nanotechnology-Based Detection. Front Pharmacol, 2017, 08(FEB):35., DOI 10.3389/fphar.2017.00035 Yang MH, Imrali A, Heeschen C. Circulating Cancer Stem Cells: the Importance to Select. Chin J Cancer Res, 2015, 27(5):437–49., DOI 10.3978/j. issn.1000-9604.2015.04.08 El-Badawy A, Ghoneim MA, Gabr MM, Salah RA, Mohamed IK, Amer M, et al. Cancer Cell-Soluble Factors Reprogram Mesenchymal Stromal Cells to Slow Cycling, Chemoresistant Cells with a More Stem-like State. Stem Cel Res Ther, 2017, 8(1):254., DOI 10.1186/s13287-017-0709-9 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610180 EP 1610199 DI 10.3389/pore.2022.1610180 PG 20 ER PT J AU Ma, T Wu, FH Wu, HX Fa, Q Chen, Y AF Ma, Tao Wu, Fa-Hong Wu, Hong-Xia Fa, Qiong Chen, Yan TI Long Non-Coding RNA MCM3AP-AS1: A Crucial Role in Human Malignancies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE cancer; biomarker; lncRNA; MCM3AP-AS1; clinical features ID cancer; biomarker; lncRNA; MCM3AP-AS1; clinical features AB The incidence of cancer continues to grow and is one of the leading causes of death in the world. Long noncoding RNAs (LncRNAs) is a group of RNA transcripts greater than 200 nucleotides in length, and although it cannot encode proteins, it can regulate different biological functions by controlling gene expression, transcription factors, etc. LncRNA micro-chromosome maintenance protein 3-associated protein antisense RNA 1 (MCM3AP-AS1) is involved in RNA processing and cell cycle-related functions, and MCM3AP-AS1 is dysregulated in expression in various types of cancers. This biomarker is involved in many processes related to carcinogens, such as cell proliferation, apoptosis, cell cycle, and migration. In this review, we summarize the roles of MCM3AP-AS1 in different human cancers and its biological functions with a view to providing ideas for future research. C1 [Ma, Tao] The Affiliated Hospital of Southwest Medical University, Department of HematologyLuzhou, China. [Wu, Fa-Hong] The Second Hospital of Lanzhou University, The Department of General SurgeryLanzhou, China. [Wu, Hong-Xia] Lanzhou University Second Hospital, Department of Nuclear MedicineLanzhou, China. [Fa, Qiong] The 940th Hospital of the People’s Liberation Army Joint Service Support Force, Department of Nuclear MedicineLanzhou, China. [Chen, Yan] The Affiliated Hospital of Southwest Medical University, Department of HematologyLuzhou, China. RP Chen, Y (reprint author), The Affiliated Hospital of Southwest Medical University, Department of Hematology, Luzhou, China. EM chenyan1211@swmu.edu.cn CR Torre LA, Siegel RL, Ward EM, Jemal A. Global Cancer Incidence and Mortality Rates and Trends-An Update. Cancer Epidemiol Biomarkers Prev, 2016, 25(1):16–27., DOI 10.1158/1055-9965.epi-15-0578 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Brucher BL, Jamall IS. Epistemology of the Origin of Cancer: A New Paradigm. BMC Cancer, 2014, 14:331., DOI 10.1186/1471-2407-14-331 Chi Y, Wang D, Wang J, Yu W, Yang J. Long Non-coding RNA in the Pathogenesis of Cancers. Cells, 2019, 8(9):1015., DOI 10.3390/cells8091015 Burrell RA, McGranahan N, Bartek J, Swanton C. The Causes and Consequences of Genetic Heterogeneity in Cancer Evolution. Nature, 2013, 501(7467):338–45., DOI 10.1038/nature12625 Fang Y, Fullwood MJ. Roles, Functions, and Mechanisms of Long Non-coding RNAs in Cancer. Genomics Proteomics Bioinformatics, 2016, 14(1):42–54., DOI 10.1016/j.gpb.2015.09.006 Lekka E, Hall J. Noncoding RNAs in Disease. FEBS Lett, 2018, 592(17): 2884–900., DOI 10.1002/1873-3468.13182 Novikova IV, Hennelly SP, Sanbonmatsu KY. Tackling Structures of Long Noncoding RNAs. Int J Mol Sci, 2013, 14(12):23672–84., DOI 10.3390/ ijms141223672 Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, et al. Exosome-Transmitted lncARSR Promotes Sunitinib Resistance in Renal Cancer by Acting as a Competing Endogenous RNA. Cancer Cell, 2016, 29(5):653–68., DOI 10. 1016/j.ccell.2016.03.004 Rajagopal T, Talluri S, Akshaya RL, Dunna NR. HOTAIR LncRNA: A Novel Oncogenic Propellant in Human Cancer. Clin Chim Acta, 2020, 503:1–18., DOI 10.1016/j.cca.2019.12.028 Peng WX, Koirala P, Mo YY. LncRNA-mediated Regulation of Cell Signaling in Cancer. Oncogene, 2017, 36(41):5661–7., DOI 10.1038/onc.2017.184 Li J, Meng H, Bai Y, Wang K. Regulation of lncRNA and its Role in Cancer Metastasis. Oncol Res, 2016, 23(5):205–17., DOI 10.3727/ 096504016x14549667334007 Luo H, Xu C, Le W, Ge B, Wang T. lncRNA CASC11 Promotes Cancer Cell Proliferation in Bladder Cancer through miRNA-150. J Cell. Biochem, 2019, 120(8):13487–93., DOI 10.1002/jcb.28622 Zhao W, Geng D, Li S, Chen Z, Sun M. LncRNA HOTAIR Influences Cell Growth, Migration, Invasion, and Apoptosis via the miR-20a-5p/HMGA2 axis in Breast Cancer. Cancer Med, 2018, 7(3):842–55., DOI 10.1002/cam4.1353 Jia Y, Duan Y, Liu T, Wang X, Lv W, Wang M, et al. LncRNA TTN-AS1 Promotes Migration, Invasion, and Epithelial Mesenchymal Transition of Lung Adenocarcinoma via Sponging miR-142-5p to Regulate CDK5. Cell Death Dis, 2019, 10(8):573., DOI 10.1038/s41419-019-1811-y Lai Y, Feng B, Abudoureyimu M, Zhi Y, Zhou H, Wang T, et al. Non-coding RNAs: Emerging Regulators of Sorafenib Resistance in Hepatocellular Carcinoma. Front Oncol, 2019, 9:1156., DOI 10.3389/fonc.2019.01156 Reymond A, Camargo AA, Deutsch S, Stevenson BJ, Parmigiani RB, Ucla C, et al. Nineteen Additional Unpredicted Transcripts from Human Chromosome 21. Genomics, 2002, 79(6):824–32., DOI 10.1006/geno.2002.6781 Gao Y, Zhao H, Li Y. LncRNA MCM3AP-AS1 Regulates miR-142-3p/HMGB1 to Promote LPS-Induced Chondrocyte Apoptosis. BMC Musculoskelet Disord, 2019, 20(1):605., DOI 10.1186/s12891-019-2967-4 Mei H, Xian H, Ke J. LncRNA-MCM3AP-AS1 Promotes the Progression of Infantile Hemangiomas by Increasing miR-138-5p/HIF-1α Axis- Regulated Glycolysis. Front Mol Biosci, 2021, 8:753218., DOI 10.3389/ fmolb.2021.753218 Zhao K, Tu C, Liang K, Li Y, Yu Y. Long Noncoding RNA MCM3AP Antisense RNA 1 is Downregulated in Chronic Obstructive Pulmonary Disease and Regulates Human Bronchial Smooth Muscle Cell Proliferation. J Int Med Res, 2020, 48(9):300060520935215., DOI 10.1177/0300060520935215 Yang C, Xu X, Lin P, Luo B, Luo S, Huang H, et al. Overexpression of Long Noncoding RNA MCM3AP-AS1 Promotes Osteogenic Differentiation of Dental Pulp Stem Cells via miR-143-3p/IGFBP5 axis. Hum Cel, 2022, 35(1):150–62., DOI 10.1007/s13577-021-00648-3 Zhang L, Liu B, Han J,Wang T, Han L. Competing Endogenous RNA Network Analysis for Screening Inflammation-Related Long Non-coding RNAs for Acute Ischemic Stroke. Mol Med Rep, 2020, 22(4):3081–94., DOI 10.3892/mmr. 2020.11415 Saygili H, Bozgeyik I, Yumrutas O, Akturk E, Bagis H. Differential Expression of Long Noncoding RNAs in Patients with Coronary Artery Disease. Mol Syndromol, 2021, 12(6):372–8., DOI 10.1159/000517077 Cao Y, Wang P, Ning S, Xiao W, Xiao B, Li X. Identification of Prognostic Biomarkers in Glioblastoma Using a Long Non-coding RNA-Mediated, Competitive Endogenous RNA Network. Oncotarget, 2016, 7(27): 41737–47., DOI 10.18632/oncotarget.9569 Wu F, Zhao Z, Chai R, Liu Y, Wang K, Wang Z, et al. Expression Profile Analysis of Antisense Long Non-coding RNA Identifies WDFY3-AS2 as a Prognostic Biomarker in Diffuse Glioma. Cancer Cel Int, 2018, 18:107., DOI 10. 1186/s12935-018-0603-2 Yan Y, Yu J, Liu H, Guo S, Zhang Y, Ye Y, et al. Construction of a Long Noncoding RNA-Associated ceRNA Network Reveals Potential Prognostic lncRNA Biomarkers in Hepatocellular Carcinoma. Pathol Res Pract, 2018, 214(12):2031–8., DOI 10.1016/j.prp.2018.09.022 Wang Y, Yang L, Chen T, Liu X, Guo Y, Zhu Q, et al. A Novel lncRNA MCM3AP-AS1 Promotes the Growth of Hepatocellular Carcinoma by Targeting miR-194-5p/FOXA1 axis. Mol Cancer, 2019, 18(1):28., DOI 10. 1186/s12943-019-0957-7 Zhang H, Luo C, Zhang G. LncRNA MCM3AP-AS1 Regulates Epidermal Growth Factor Receptor and Autophagy to Promote Hepatocellular Carcinoma Metastasis by Interacting with miR-455. DNA Cel Biol, 2019, 38(8):857–64., DOI 10.1089/dna.2019.4770 Wu J, Lv Y, Li Y, Jiang Y,Wang L, Zhang X, et al.MCM3AP-AS1/miR-876-5p/ WNT5A axis Regulates the Proliferation of Prostate Cancer Cells. Cancer Cel Int, 2020, 20:307., DOI 10.1186/s12935-020-01365-x Li X, Lv J, Liu S. MCM3AP-AS1 KD Inhibits Proliferation, Invasion, and Migration of PCa Cells via DNMT1/DNMT3, A/B, Methylation-Mediated Upregulation of NPY1R. Mol Ther Nucleic Acids, 2020, 20:265–78., DOI 10. 1016/j.omtn.2020.01.016 Jia Z, Li W, Bian P, Liu H, Pan D, Dou Z. LncRNA MCM3AP-AS1 Promotes Cell Proliferation and Invasion through Regulating miR-543-3p/SLC39A10/ PTEN Axis in Prostate Cancer. Onco Targets Ther, 2020, 13:9365–76., DOI 10. 2147/ott.s245537 Lan L, Liang Z, Zhao Y, Mo Y. LncRNA MCM3AP-AS1 Inhibits Cell Proliferation in Cervical Squamous Cell Carcinoma by Down-Regulating miRNA-93. Biosci Rep, 2020, 40(2):BSR20193794., DOI 10.1042/BSR20193794 Chen Q, Xu H, Zhu J, Feng K, Hu C. LncRNA MCM3AP-AS1 Promotes Breast Cancer Progression via Modulating miR-28-5p/CENPF axis. Biomed Pharmacother, 2020, 128:110289., DOI 10.1016/j.biopha.2020.110289 Ren G, Han G, Song Z, Zang A, Liu B, Hu L, et al. LncRNA MCM3AP-AS1 Downregulates LncRNA MEG3 in Triple Negative Breast Cancer to Inhibit the Proliferation of Cancer Cells. Crit Rev Eukaryot Gene Expr, 2021, 31(4):81–7., DOI 10.1615/critreveukaryotgeneexpr.2021038296 Ma X, Luo J, Zhang Y, Sun D, Lin Y. LncRNA MCM3AP-AS1 Upregulates CDK4 by Sponging miR-545 to Suppress G1 Arrest in Colorectal Cancer. Cancer Manag Res, 2020, 12:8117–24., DOI 10.2147/cmar.s247330 Zhou M, Bian Z, Liu B, Zhang Y, Cao Y, Cui K, et al. Long Noncoding RNA MCM3AP-AS1 Enhances Cell Proliferation and Metastasis in Colorectal Cancer by Regulating miR-193a-5p/SENP1. Cancer Med, 2021, 10(7): 2470–81., DOI 10.1002/cam4.3830 Yu Y, Lai S, Peng X. Long Non-coding RNA MCM3AP-AS1 Facilitates Colorectal Cancer Progression by Regulating the microRNA-599/ARPP19 axis. Oncol Lett, 2021, 21(3):225., DOI 10.3892/ol.2021.12486 Liang M, Jia J, Chen L, Wei B, Guan Q, Ding Z, et al. LncRNA MCM3AP-AS1 Promotes Proliferation and Invasion through Regulating miR-211-5p/SPARC axis in Papillary Thyroid Cancer. Endocrine, 2019, 65(2):318–26., DOI 10.1007/ s12020-019-01939-4 Guo C, Gong M, Li Z. Knockdown of lncRNA MCM3AP-AS1 Attenuates Chemoresistance of Burkitt Lymphoma to Doxorubicin Treatment via Targeting the miR-15a/EIF4E Axis. Cancer Manag Res, 2020, 12:5845–55., DOI 10.2147/cmar.s248698 Yang M, Sun S, Guo Y, Qin J, Liu G. Long Non-coding RNA MCM3AP-AS1 Promotes Growth and Migration through Modulating FOXK1 by Sponging miR-138-5p in Pancreatic Cancer. Mol Med, 2019, 25(1):55., DOI 10.1186/ s10020-019-0121-2 Wang H, Xu T, Wu L, Xu HL, Liu RM. Molecular Mechanisms of MCM3APAS1 Targeted the Regulation of miR-708-5p on Cell Proliferation and Apoptosis in Gastric Cancer Cells. Eur Rev Med Pharmacol Sci, 2020, 24(5):2452–61., DOI 10.26355/eurrev_202003_20512 Sun H, Wu P, Zhang B, Wu X, Chen W. MCM3AP-AS1 Promotes Cisplatin Resistance in Gastric Cancer Cells via the miR-138/FOXC1 axis. Oncol Lett, 2021, 21(3):211., DOI 10.3892/ol.2021.12472 Li X, Yu M, Yang C. YY1-mediated Overexpression of Long Noncoding RNA MCM3AP-AS1 Accelerates Angiogenesis and Progression in Lung Cancer by Targeting miR-340-5p/KPNA4 axis. J Cel Biochem, 2020, 121(3):2258–67., DOI 10.1002/jcb.29448 Qiu L, Ma Y, Yang Y, Ren X, Wang D, Jia X. Pro-Angiogenic and Proinflammatory Regulation by lncRNA MCM3AP-AS1-Mediated Upregulation of DPP4 in Clear Cell Renal Cell Carcinoma. Front Oncol, 2020, 10:705., DOI 10.3389/fonc.2020.00705 Li H, Jiang J. LncRNA MCM3AP-AS1 Promotes Proliferation, Migration and Invasion of Oral Squamous Cell Carcinoma Cells via Regulating miR-204-5p/ FOXC1. J Investig Med, 2020, 68(7):1282–8., DOI 10.1136/jim-2020-001415 Hou C,Wang X, Du B. lncRNA MCM3AP-AS1 Promotes the Development of Oral Squamous Cell Carcinoma by Inhibiting miR-363-5p. Exp Ther Med, 2020, 20(2):978–84., DOI 10.3892/etm.2020.8738 Sun P, Feng Y, Guo H, Li R, Yu P, Zhou X, et al. MiR-34a Inhibits Cell Proliferation and Induces Apoptosis inHuman Nasopharyngeal Carcinoma by Targeting lncRNA MCM3AP-AS1. Cancer Manag Res, 2020, 12:4799–806., DOI 10.2147/cmar.s245520 Forner A, Reig M, Bruix J. Hepatocellular Carcinoma. Lancet, 2018, 391(10127):1301–14., DOI 10.1016/s0140-6736(18)30010-2 Wang G, Zhao D, Spring DJ, DePinho RA. Genetics and Biology of Prostate Cancer. Genes Dev, 2018, 32(17-18):1105–40., DOI 10.1101/gad. 315739.118 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global Cancer Statistics, 2012. CA Cancer J Clin, 2015, 65(2):87–108., DOI 10.3322/caac.21262 Chen Y, Chen Z, Mo J, Pang M, Chen Z, Feng F, et al. Identification of HCG18 and MCM3AP-AS1 that Associate with Bone Metastasis, Poor Prognosis and Increased Abundance of M2 Macrophage Infiltration in Prostate Cancer. Technol Cancer Res Treat, 2021, 20:1533033821990064., DOI 10.1177/ 1533033821990064 Peng L, Yuan X, Jiang B, Tang Z, Li G-C. LncRNAs: Key Players and Novel Insights into Cervical Cancer. Tumor Biol, 2016, 37(3):2779–88., DOI 10.1007/ s13277-015-4663-9 Harbeck N, Gnant M. Breast Cancer. Lancet, 2017, 389(10074):1134–50., DOI 10.1016/s0140-6736(16)31891-8 Riahi A, Hosseinpour-Feizi M, Rajabi A, Akbarzadeh M, Montazeri V, Safaralizadeh R. Overexpression of Long Non-coding RNA MCM3APAS1 in Breast Cancer Tissues Compared to Adjacent Non-tumour Tissues. Br J Biomed Sci, 2021, 78(2):53–7., DOI 10.1080/09674845. 2020.1798058 Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal Cancer. Lancet, 2019, 394(10207):1467–80., DOI 10.1016/S0140-6736(19)32319-0 Dai W, Zeng W, Lee D. lncRNA MCM3AP-AS1 Inhibits the Progression of Colorectal Cancer via the miR-19a-3p/FOXF2 axis. J Gene Med, 2021, 23(3): e3306., DOI 10.1002/jgm.3306 Liu Z, Lv T, Xie C, Di Z. BRAF V600E Gene Mutation is Associated with Bilateral Malignancy of Papillary Thyroid Cancer. Am JMed Sci, 2018, 356(2): 130–4., DOI 10.1016/j.amjms.2018.04.012 Sui F, Ji M, Hou P. Long Non-coding RNAs in Thyroid Cancer: Biological Functions and Clinical Significance. Mol Cel Endocrinol, 2018, 469:11–22., DOI 10.1016/j.mce.2017.07.020 Dunleavy K. Approach to the Diagnosis and Treatment of Adult Burkitt’s Lymphoma. J Oncol Pract, 2018, 14(11):665–71., DOI 10.1200/jop.18.00148 Zhao L, Liu Y, Zhang J, Liu Y, Qi Q. LncRNA SNHG14/miR-5590-3p/ZEB1 Positive Feedback Loop Promoted Diffuse Large B Cell Lymphoma Progression and Immune Evasion through Regulating PD-1/PD-L1 Checkpoint. Cel Death Dis, 2019, 10(10):731., DOI 10.1038/s41419-019- 1886-5 Wang QM, Lian GY, Song Y, Huang YF, Gong Y. LncRNA MALAT1 Promotes Tumorigenesis and Immune Escape of Diffuse Large B Cell Lymphoma by Sponging miR-195. Life Sci, 2019, 231:116335., DOI 10.1016/ j.lfs.2019.03.040 Siyu G, Linqing Z, Linling K, Hong L, Guoqi S, Cho WC. Long Noncoding RNA Identification in Lymphoma. Future Oncol, 2017, 13(27):2479–87., DOI 10.2217/fon-2017-0230 Conroy T, Bachet J-B, Ayav A, Huguet F, Lambert A, Caramella C, et al. Current Standards and New Innovative Approaches for Treatment of Pancreatic Cancer. Eur J Cancer, 2016, 57:10–22., DOI 10.1016/j.ejca.2015. 12.026 Previdi MC, Carotenuto P, Zito D, Pandolfo R, Braconi C. Noncoding RNAs as Novel Biomarkers in Pancreatic Cancer: What Do We Know? Future Oncol, 2017, 13(5):443–53., DOI 10.2217/fon-2016-0253 Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric Cancer. Lancet, 2020, 396(10251):635–48., DOI 10.1016/S0140-6736(20, 31288-5 Wei L, Sun J, Zhang N, Zheng Y, Wang X, Lv L, et al. Noncoding RNAs in Gastric Cancer: Implications for Drug Resistance. Mol Cancer, 2020, 19(1):62., DOI 10.1186/s12943-020-01185-7 Chen Z, Lei T, Chen X, Gu J, Huang J, Lu B, et al. Long Non-coding RNA in Lung Cancer. Clinica Chim Acta, 2020, 504:190–200., DOI 10.1016/j.cca.2019.11.031 Jonasch E, Walker CL, Rathmell WK. Clear Cell Renal Cell Carcinoma Ontogeny and Mechanisms of Lethality. Nat Rev Nephrol, 2021, 17(4): 245–61., DOI 10.1038/s41581-020-00359-2 Panarese I, Aquino G, Ronchi A, Longo F, Montella M, Cozzolino I, et al. Oral and Oropharyngeal Squamous Cell Carcinoma: Prognostic and Predictive Parameters in the Etiopathogenetic Route. Expert Rev Anticancer Ther, 2019, 19(2):105–19., DOI 10.1080/14737140.2019. 1561288 Zhang L, Meng X, Zhu XW, Yang DC, Chen R, Jiang Y, et al. Long Non-coding RNAs in Oral Squamous Cell Carcinoma: Biologic Function, Mechanisms and Clinical Implications. Mol Cancer, 2019, 18(1):102., DOI 10.1186/s12943-019- 1021-3 Roberts TC, Langer R, Wood MJA. Advances in Oligonucleotide Drug Delivery. Nat Rev Drug Discov, 2020, 19(10):673–94., DOI 10.1038/s41573- 020-0075-7 Crooke ST, Witztum JL, Bennett CF, Baker BF. RNA-targeted Therapeutics. Cell Metab, 2018, 27(4):714–39., DOI 10.1016/j.cmet.2018.03.004 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610194 EP 1610202 DI 10.3389/pore.2022.1610194 PG 9 ER PT J AU Zhang, R Tian, X Luo, Y Dong, H Tian, W Zhang, Y Li, D Sun, H Meng, Z AF Zhang, Ruyi Tian, Xin Luo, Ying Dong, Haiwei Tian, Weijun Zhang, Yujie Li, Dong Sun, Haoran Meng, Zhaowei TI Case Report: Recurrent Malignant Struma Ovarii With Hyperthyroidism and Metastases, A Rare Case Report and Review of the Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE case report; surgery; struma ovarii; hyperthyroidism; CA125; thyroglobulin ID case report; surgery; struma ovarii; hyperthyroidism; CA125; thyroglobulin AB Background: SO (Struma ovarii) is a rare form of ovarian teratoma which originates from ovarian dermoid cysts. Due to the rarity of this disease, relevant studies might not be sufficiently documented, especially cases with hyperthyroidism and multiple metastases. Case Presentation: A 40-year-old female patient was admitted to our hospital due to management of early pregnancy along with a recurrent abdominal and pelvic mass. Contrast-enhanced CT images showed an irregular mass (10.7 × 8.6 × 12.8 cm) located in the right side from the hypogastrium to the pelvic cavity and another mass (3.8 × 3.7 cm) in the liver. Laboratory examination showed that CA125 (Carbohydrate Antigen-125) was 118.10 U/mL, Tg (thyroglobulin) was >300 ng/ml, FT4 (free thyroxine) was 22.11 pmol/L, and TSH (thyroid-stimulating hormone) was <0.004 mIU/L. She subsequently underwent liver mass dissection, omentectomy, tumor dissection, peritoneal nodule resection, as well as rectal anterior wall nodule resection. The patient was diagnosed with malignant SO (papillary type) along with multiple metastases. Also, we conducted a literature review based on 290 SO cases from 257 articles. Conclusion: This study showed that malignant SO might be prone to relapse and metastasize (a metastatic rate of 52.94%) and therefore aggressive management might need to be recommended for malignant SO. Also, laparotomy might need to be recommended for large tumors that cannot be resected by laparoscopic surgery since these tumors might be prone to rupture and thus produce peritoneal implants. Furthermore, Graves’ disease might need to be considered in the differential diagnosis. C1 [Zhang, Ruyi] Tianjin Medical University General Hospital, Department of Nuclear MedicineTianjin, China. [Tian, Xin] Tianjin Medical University General Hospital, Department of Nuclear MedicineTianjin, China. [Luo, Ying] Tianjin Medical University General Hospital, Department of Obstetrics and GynecologyTianjin, China. [Dong, Haiwei] Tianjin Medical University General Hospital, Department of Obstetrics and GynecologyTianjin, China. [Tian, Weijun] Tianjin Medical University General Hospital, Department of General SurgeryTianjin, China. [Zhang, Yujie] Tianjin Medical University General Hospital, Department of PathologyTianjin, China. [Li, Dong] Tianjin Medical University General Hospital, Department of Medical ImagingTianjin, China. [Sun, Haoran] Tianjin Medical University General Hospital, Department of Medical ImagingTianjin, China. [Meng, Zhaowei] Tianjin Medical University General Hospital, Department of Nuclear MedicineTianjin, China. RP Meng, Z (reprint author), Tianjin Medical University General Hospital, Department of Nuclear Medicine, Tianjin, China. EM zmeng@tmu.edu.cn CR Gonet A, Slusarczyk R, Gasior-Perczak D, Kowalik A, Kopczynski J, Kowalska A. Papillary Thyroid Cancer in a Struma Ovarii in a 17-Year-Old Nulliparous Patient: A Case Report. Diagnostics, 2020, 10(1):45., DOI 10.3390/ diagnostics10010045 Fujiwara S, Tsuyoshi H, Nishimura T, Takahashi N, Yoshida Y. Precise Preoperative Diagnosis of Struma Ovarii with Pseudo-meigs’ Syndrome Mimicking Ovarian Cancer with the Combination of 131I Scintigraphy and 18F-FDG PET: Case Report and Review of the Literature. J Ovarian Res, 2018, 11(1):11., DOI 10.1186/s13048-018-0383-2 Jean S, Tanyi JL, Montone K, McGrath C, Lage-Alvarez MM, Chu CS. Papillary Thyroid Cancer Arising in Struma Ovarii. J Obstet Gynaecol, 2012, 32(3):222–6., DOI 10.3109/01443615.2011.645921 Goffredo P, Sawka AM, Pura J, Adam MA, Roman SA, Sosa JA. Malignant Struma Ovarii: A Population-Level Analysis of a Large Series of 68 Patients. Thyroid, 2015, 25(2):211–5., DOI 10.1089/thy.2014.0328 Dardik RB, Dardik M, Westra W, Montz FJ. Malignant Struma Ovarii: Two Case Reports and a Review of the Literature. Gynecol Oncol, 1999, 73(3): 447–51., DOI 10.1006/gyno.1999.5355 Makani S, Kim W, Gaba AR. Struma Ovarii with a Focus of Papillary Thyroid Cancer: A Case Report and Review of the Literature. Gynecol Oncol, 2004, 94(3):835–9., DOI 10.1016/j.ygyno.2004.06.003 Wang L, Chen X, Liu B. Lower Lip Teratoma with Ventral Capillary Malformation in an Infant: Case Report and Literature Review. Int J Oral Maxill Surg, 2009, 38(12):1330–3., DOI 10.1016/j.ijom.2009.06.018 Outwater EK, Siegelman ES, Hunt JL. Ovarian Teratomas: Tumor Types and Imaging Characteristics. RadioGraphics, 2001, 21(2):475–90., DOI 10.1148/ radiographics.21.2.g01mr09475 Stanojevic B, Dzodic R, Saenko V, Milovanovic Z, Krstevski V, Radlovic P, et al. Unilateral Follicular Variant of Papillary Thyroid Carcinoma with Unique KRAS Mutation in Struma Ovarii in Bilateral Ovarian Teratoma: A Rare Case Report. BMC Cancer, 2012, 12:224., DOI 10.1186/1471-2407-12-224 Coyne C, Nikiforov YE. RAS Mutation-Positive Follicular Variant of Papillary Thyroid Carcinoma Arising in a Struma Ovarii. Endocr Pathol, 2010, 21(2): 144–7., DOI 10.1007/s12022-009-9097-8 Kabukcuoglu F, Baksu A, Yilmaz B, Aktumen A, Evren I. Malignant Struma Ovarii. Pathol Oncol Res, 2002, 8(2):145–7., DOI 10.1007/bf03033726 Rosenblum NG, LiVolsi VA, Edmonds PR, Mikuta JJ. Malignant Struma Ovarii. Gynecol Oncol, 1989, 32(2):224–7., DOI 10.1016/s0090-8258(89, 80037-x Volpi E, Ferrero A, Nasi PG, Sismondi P. Malignant Struma Ovarii: a Case Report of Laparoscopic Management. Gynecol Oncol, 2003, 90(1):191–4., DOI 10.1016/s0090-8258(03)00142-2 Shrimali RK, Shaikh G, Reed NS. Malignant Struma Ovarii: The West of Scotland Experience and Review of Literature with Focus on Postoperative Management. JMed Imaging Radiat Oncol, 2012, 56(4):478–82., DOI 10.1111/j. 1754-9485.2012.02394.x Devaney K, Snyder R, Morris HJ, Tavassoli FA. Proliferative and Histologically Malignant Struma Ovarii. Int J Gynecol Pathol, 1993, 12(4):333–43., DOI 10. 1097/00004347-199310000-00008 Boutross-Tadross O, Saleh R, Asa SL. Follicular Variant Papillary Thyroid Carcinoma Arising in Struma Ovarii. Endocr Pathol, 2007, 18(3):182–6., DOI 10.1007/s12022-007-0022-8 Pardo-Mindan FJ, Vazquez JJ. Malignant Struma Ovarii: Light and Electron Microscopic Study. Cancer, 1983, 51(2):337–43., DOI 10.1002/1097- 0142(19830115)51:2<337::aid-cncr2820510229>3.0.co;2-5 Shaco-Levy R, Bean SM, Bentley RC, Robboy SJ. Natural History of Biologically Malignant Struma Ovarii: Analysis of 27 Cases with Extraovarian Spread. Int J Gynecol Pathol, 2010, 29(3):212–27., DOI 10.1097/ PGP.0b013e3181bfb133 Shaco-Levy R, Peng RY, Snyder MJ, Osmond GW, Veras E, Bean SM, et al. Malignant Struma Ovarii: A Blinded Study of 86 Cases Assessing Which Histologic Features Correlate with Aggressive Clinical Behavior. Arch Pathol Lab Med, 2012, 136(2):172–8., DOI 10.5858/arpa.2011- 0092-OA Zhang X, Axiotis C. Thyroid-type Carcinoma of Struma Ovarii. Arch Pathol Lab Med, 2010, 134(5):786–91., DOI 10.5858/134.5.786 Wei S, Baloch ZW, LiVolsi VA. Pathology of Struma Ovarii: A Report of 96 Cases. Endocr Pathol, 2015, 26(4):342–8., DOI 10.1007/s12022-015-9396-1 Yoo S-C, Chang K-H, Lyu M-O, Chang S-J, Ryu H-S, Kim H-S. Clinical Characteristics of Struma Ovarii. J Gynecol Oncol, 2008, 19(2):135–8., DOI 10. 3802/jgo.2008.19.2.135 Hatami M, Breining D, Owers RL, Del Priore G, Goldberg GL. Malignant Struma Ovarii - A Case Report and Review of the Literature. Gynecol Obstet Invest, 2008, 65(2):104–7., DOI 10.1159/000108654 Zakhem A, Aftimos G, Kreidy R, Salem P. Malignant Struma Ovarii: Report of Two Cases and Selected Review of the Literature. J Surg Oncol, 1990, 43(1): 61–5., DOI 10.1002/jso.2930430116 Robboy SJ, Scully RE. Strumal Carcinoid of the Ovary: An Analysis of 50 Cases of a Distinctive Tumor Composed of Thyroid Tissue and Carcinoid. Cancer, 1980, 46(9):2019–34., DOI 10.1002/1097-0142(19801101)46:9<2019::aidcncr2820460921> 3.0.co;2-w Lazarus JH, Richards AR, MacPherson MJ, Dinnen JS, Williams ED, Owen GM, et al. Struma Ovarii: A Case Report. Clin Endocrinol, 1987, 27(6):715–20., DOI 10.1111/j.1365-2265.1987.tb02955.x Anastasilakis AD, Ruggeri R-M, Polyzos SA, Makras P, Molyva D, Campenni A, et al. Coexistence of Graves’ Disease, Papillary Thyroid Carcinoma and Unilateral Benign Struma Ovarii: Case Report and Review of the Literature. Metabolism, 2013, 62(10):1350–6., DOI 10.1016/j.metabol.2013.05.013 Huh JJ, Montz FJ, Bristow RE. Struma Ovarii Associated with Pseudo-Meigs’ Syndrome and Elevated Serum CA 125. Gynecol Oncol, 2002, 86(2):231–4., DOI 10.1006/gyno.2002.6741 Yanaranop M, Tiyayon J, Siricharoenthai S, Nakrangsee S, Thinkhamrop B. Rajavithi-ovarian Cancer Predictive Score, R-OPS): A New Scoring System for Predicting Ovarian Malignancy in Women Presenting with a Pelvic Mass. Gynecol Oncol, 2016, 141(3):479–84., DOI 10.1016/j.ygyno.2016.03.019 Elisei R, Romei C, Castagna MG, Lisi S, Vivaldi A, Faviana P, et al. RET/PTC3 Rearrangement and Thyroid Differentiation Gene Analysis in a Struma Ovarii Fortuitously Revealed by Elevated Serum Thyroglobulin Concentration. Thyroid, 2005, 15(12):1355–61., DOI 10.1089/thy.2005.15.1355 Kraemer B, Grischke E-M, Staebler A, Hirides P, Rothmund R. Laparoscopic Excision of Malignant Struma Ovarii and 1 Year Follow-Up without Further Treatment. Fertil sterility, 2011, 95(6):2124., DOI 10.1016/j.fertnstert.2010. 12.047 Ukita M, Nakai H, Kotani Y, Tobiume T, Koike E, Tsuji I, et al. Long-term Survival in Metastatic Malignant Struma Ovarii Treated with Oral Chemotherapy: A Case Report. Oncol Lett, 2014, 8(6):2458–62., DOI 10. 3892/ol.2014.2587 Marti JL, Clark VE, Harper H, Chhieng DC, Sosa JA, Roman SA. Optimal Surgical Management of Well-Differentiated Thyroid Cancer Arising in Struma Ovarii: A Series of 4 Patients and a Review of 53 Reported Cases. Thyroid, 2012, 22(4):400–6., DOI 10.1089/thy.2011.0162 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610221 EP 1610229 DI 10.3389/pore.2022.1610221 PG 9 ER PT J TI The Diagnostic Significance of CXCL13 in M2 Tumor Immune Microenvironment of Human Astrocytoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; gliomas; M2 macrophages; astrocytoma; CXCL13 ID immunohistochemistry; gliomas; M2 macrophages; astrocytoma; CXCL13 AB Background: CXCL13 may act as a mediator of tumor-associated macrophage immunity during malignant progression. Objective: The present study clarifies the clinicopathological significances of CXCL13 and its corresponding trend with M2 macrophage in human astrocytoma. Methods: The predictive potential of CXCL13 was performed using 695 glioma samples derived from TCGA lower-grade glioma and glioblastoma (GBMLGG) dataset. CXCL13 and M2 biomarker CD163 were observed by immunohistochemistry in 112 astrocytoma tissues. Results: An in-depth analysis showed that CXCL13 expression was related to the poor prognosis of glioma patients (p = 0.0002) derive fromTCGA analysis. High level of CXCL13 was detected in 43 (38.39%) astrocytoma and CXCL13/CD163 coexpression was expressed in 33 (29.46%) cases. The immunoreactivities of CXCL13 and CXCL13/ CD163 were found in the malignant lesions, which were both significantly associated with grade, patient survival, and IDH1 mutation. Single CXCL13 and CXCL13/CD163 coexpression predicted poor overall survival in astrocytoma (p = 0.0039 and p = 0.0002, respectively). Multivariate Cox regression analyses manifested CXCL13/CD163 phenotype was a significant independent prognostic indicator of patient outcome in astrocytoma (CXCL13, p = 0.0642; CXCL13/CD163, p = 0.0368). Conclusion: CXCL13 overexpression is strongly linked to CD163+ M2 infiltration in malignant astrocytoma. CXCL13/CD163 coexpression would imply M2c-related aggressive characteristics existing in astrocytoma progression could also provide predictive trends of patient outcomes. CR Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a Summary. Acta Neuropathol, 2016, 131(6):803–20., DOI 10.1007/s00401-016-1545-1 Aum DJ, Kim DH, Beaumont TL, Leuthardt EC, Dunn GP, Kim AH. Molecular and Cellular Heterogeneity: the Hallmark of Glioblastoma. Foc, 2014, 37(6):E11., DOI 10.3171/2014.9.focus14521 Kraboth Z, Kalman B. Longitudinal Characteristics of Glioblastoma in Genome-wide Studies. Pathol Oncol Res, 2019, 26(4):2035–47., DOI 10.1007/ s12253-019-00705-1 Hambardzumyan D, Bergers G. Glioblastoma: Defining Tumor Niches. Trends Cancer, 2015, 1(4):252–65., DOI 10.1016/j.trecan.2015.10.009 Chen Z, Hambardzumyan D. Immune Microenvironment in Glioblastoma Subtypes. Front Immunol, 2018, 9:1004., DOI 10.3389/fimmu.2018.01004 Martinez-LageM, Lynch TM, Bi Y, Cocito C, WayGP, Pal S, et al. Immune Landscapes Associated with Different Glioblastoma Molecular Subtypes. Acta Neuropathol Commun, 2019, 7(1):203., DOI 10.1186/s40478-019- 0803-6 Vidyarthi A, Agnihotri T, Khan N, Singh S, Tewari MK, Radotra BD, et al. Predominance of M2 Macrophages in Gliomas Leads to the Suppression of Local and Systemic Immunity. Cancer Immunol Immunother, 2019, 68(12): 1995–2004., DOI 10.1007/s00262-019-02423-8 Morisse MC, Jouannet S, Dominguez-Villar M, Sanson M, Idbaih A. Interactions between Tumor-Associated Macrophages and Tumor Cells in Glioblastoma: Unraveling Promising Targeted Therapies. Expert Rev Neurotherapeutics, 2018, 18(9):729–37., DOI 10.1080/14737175.2018.1510321 Lin Y, Xu J, Lan H. Tumor-associated Macrophages in Tumor Metastasis: Biological Roles and Clinical Therapeutic Applications. J Hematol Oncol, 2019, 12(1):76., DOI 10.1186/s13045-019-0760-3 Jayasingam SD, Citartan M, Thang TH, Mat Zin AA, Ang KC, Ch’ng ES. Evaluating the Polarization of Tumor-Associated Macrophages into M1 and M2 Phenotypes in Human Cancer Tissue: Technicalities and Challenges in Routine Clinical Practice. Front Oncol, 2019, 9:1512., DOI 10.3389/fonc.2019. 01512 Bercovici N, Guerin MV, Trautmann A, Donnadieu E. The Remarkable Plasticity of Macrophages: A Chance to Fight Cancer. Front Immunol, 2019, 10:1563., DOI 10.3389/fimmu.2019.01563 Das A, Sinha M, Datta S, Abas M, Chaffee S, Sen CK, et al. Monocyte and Macrophage Plasticity in Tissue Repair and Regeneration. Am J Pathol, 2015, 185(10):2596–606., DOI 10.1016/j.ajpath.2015.06.001 Lisi L, Ciotti GMP, Braun D, Kalinin S, Curro D, Dello Russo C, et al. Expression of iNOS, CD163 and ARG-1 Taken as M1 and M2 Markers of Microglial Polarization in Human Glioblastoma and the Surrounding normal Parenchyma. Neurosci Lett, 2017, 645:106–12., DOI 10.1016/j.neulet.2017. 02.076 Biswas SK, Mantovani A. Macrophage Plasticity and Interaction with Lymphocyte Subsets: Cancer as a Paradigm. Nat Immunol, 2010, 11(10): 889–96., DOI 10.1038/ni.1937 Wang N, Liang H, Zen K. Molecular Mechanisms that Influence the Macrophage M1a€"M2 Polarization Balance. Front Immunol, 2014, 5:614., DOI 10.3389/fimmu.2014.00614 Brown JM, Recht L, Strober S. The Promise of Targeting Macrophages in Cancer Therapy. Clin Cancer Res, 2017, 23(13):3241–50., DOI 10.1158/1078- 0432.ccr-16-3122 Movahedi K, Laoui D, Gysemans C, Baeten M, Stange G, Van den Bossche J, et al. Different Tumor Microenvironments Contain Functionally Distinct Subsets of Macrophages Derived from Ly6C(high, Monocytes. Cancer Res, 2010, 70(14):5728–39., DOI 10.1158/0008-5472.can-09-4672 Laoui D, Van Overmeire E, Di Conza G, Aldeni C, Keirsse J, Morias Y, et al. Tumor Hypoxia Does Not Drive Differentiation of Tumor-Associated Macrophages but rather fine-tunes the M2-like Macrophage Population. Cancer Res, 2014, 74(1):24–30., DOI 10.1158/0008-5472.can-13-1196 Amici SA, Dong J, Guerau-de-Arellano M. Molecular Mechanisms Modulating the Phenotype of Macrophages and Microglia. Front Immunol, 2017, 8:1520., DOI 10.3389/fimmu.2017.01520 Lopes RL, Borges TJ, Zanin RF, Bonorino C. IL-10 Is Required for Polarization of Macrophages to M2-like Phenotype by Mycobacterial DnaK, Heat Shock Protein 70). Cytokine, 2016, 85:123–9., DOI 10.1016/j.cyto.2016.06.018 Roszer T. Understanding the Mysterious M2 Macrophage through Activation Markers and Effector Mechanisms. Mediators Inflamm, 2015, 2015:816460. Ferrante CJ, Leibovich SJ. Regulation of Macrophage Polarization and Wound Healing. Adv Wound Care, 2012, 1(1):10–6., DOI 10.1089/wound.2011.0307 Ferrante CJ, Pinhal-Enfield G, Elson G, Cronstein BN, Hasko G, Outram S, et al. The Adenosine-dependent Angiogenic Switch of Macrophages to an M2- like Phenotype Is Independent of Interleukin-4 Receptor Alpha, IL-4Rα, Signaling. Inflammation, 2013, 36(4):921–31., DOI 10.1007/s10753-013-9621-3 Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, et al. Macrophage Activation and Polarization: Nomenclature and Experimental Guidelines. Immunity, 2014, 41(1):14–20., DOI 10.1016/j.immuni.2014.06.008 Komohara Y, Ohnishi K, Kuratsu J, Takeya M. Possible Involvement of the M2 Anti-inflammatory Macrophage Phenotype in Growth of Human Gliomas. J Pathol, 2008, 216(1):15–24., DOI 10.1002/path.2370 Gjorgjevski M, Hannen R, Carl B, Li Y, Landmann E, Buchholz M, et al. Molecular Profiling of the Tumor Microenvironment in Glioblastoma Patients: Correlation of Microglia/macrophage Polarization State with Metalloprotease Expression Profiles and Survival. Biosci Rep, 2019, 39(6)., DOI 10.1042/BSR20182361 Wei J, Chen P, Gupta P, Ott M, Zamler D, Kassab C, et al. Immune Biology of Glioma-Associated Macrophages and Microglia: Functional and Therapeutic Implications. Neuro Oncol, 2020, 22(2):180–94., DOI 10.1093/neuonc/noz212 Hattermann K, Sebens S, Helm O, Schmitt AD, Mentlein R, Mehdorn HM, et al. Chemokine Expression Profile of Freshly Isolated Human Glioblastoma- Associated Macrophages/microglia. Oncol Rep, 2014, 32(1):270–6., DOI 10. 3892/or.2014.3214 Hambardzumyan D, Gutmann DH, Kettenmann H. The Role of Microglia and Macrophages in Glioma Maintenance and Progression. Nat Neurosci, 2016, 19(1):20–7., DOI 10.1038/nn.4185 Chow MT, Luster AD. Chemokines in Cancer. Cancer Immunol Res, 2014, 2(12):1125–31., DOI 10.1158/2326-6066.cir-14-0160 Nagarsheth N, Wicha MS, Zou W. Chemokines in the Cancer Microenvironment and Their Relevance in Cancer Immunotherapy. Nat Rev Immunol, 2017, 17(9):559–72., DOI 10.1038/nri.2017.49 Tan P, Shi M, Lai L, Tang Z, Xie N, Xu H, et al. Regulative Role of the CXCL13- CXCR5 axis in the Tumor Microenvironment. Precision Clin Med, 2018, 1(1): 49–56. Kazanietz MG, Durando M, Cooke M. CXCL13 and its Receptor CXCR5 in Cancer: Inflammation, Immune Response, and beyond. Front Endocrinol, 2019, 10:471., DOI 10.3389/fendo.2019.00471 Sciume G, Santoni A, Bernardini G. Chemokines and Glioma: Invasion and More. J Neuroimmunol, 2010, 224(1-2):8–12., DOI 10.1016/j.jneuroim.2010. 05.019 Ansel KM, Ngo VN, Hyman PL, Luther SA, Forster R, Sedgwick JD, et al. A Chemokine-Driven Positive Feedback Loop Organizes Lymphoid Follicles. Nature, 2000, 406(6793):309–14., DOI 10.1038/35018581 van de Pavert SA, Olivier BJ, Goverse G, Vondenhoff MF, Greuter M, Beke P, et al. Chemokine CXCL13 Is Essential for Lymph Node Initiation and Is Induced by Retinoic Acid and Neuronal Stimulation. Nat Immunol, 2009, 10(11):1193–9., DOI 10.1038/ni.1789 Fischer L, Korfel A, Pfeiffer S, Kiewe P, Volk H-D, Cakiroglu H, et al. CXCL13 and CXCL12 in central Nervous System Lymphoma Patients. Clin Cancer Res, 2009, 15(19):5968–73., DOI 10.1158/1078-0432.ccr-09-0108 Moser B. CXCR5, the Defining Marker for Follicular B Helper T, TFH, Cells. Front Immunol, 2015, 6:296., DOI 10.3389/fimmu.2015.00296 Krumbholz M, Theil D, Cepok S, Hemmer B, Kivisakk P, Ransohoff RM, et al. Chemokines in Multiple Sclerosis: CXCL12 and CXCL13 Up-Regulation Is Differentially Linked to CNS Immune Cell Recruitment. Brain, 2006, 129(Pt 1):200–11., DOI 10.1093/brain/awh680 Burkle A, Niedermeier M, Schmitt-Graff A, Wierda WG, Keating MJ, Burger JA. Overexpression of the CXCR5 Chemokine Receptor, and its Ligand, CXCL13 in B-Cell Chronic Lymphocytic Leukemia. Blood, 2007, 110(9): 3316–25., DOI 10.1182/blood-2007-05-089409 Panse J, Friedrichs K, Marx A, Hildebrandt Y, Luetkens T, Bartels K, et al. Chemokine CXCL13 Is Overexpressed in the Tumour Tissue and in the Peripheral Blood of Breast Cancer Patients. Br J Cancer, 2008, 99(6):930–8., DOI 10.1038/sj.bjc.6604621 Zheng Z, Cai Y, Chen H, Chen Z, Zhu D, Zhong Q, et al. CXCL13/CXCR5 Axis Predicts Poor Prognosis and Promotes Progression through PI3K/AKT/ mTOR Pathway in Clear Cell Renal Cell Carcinoma. Front Oncol, 2018, 8: 682., DOI 10.3389/fonc.2018.00682 Gu-Trantien C, Migliori E, Buisseret L, de Wind A, Brohee S, Garaud S, et al. CXCL13-producing TFH Cells Link Immune Suppression and Adaptive Memory in Human Breast Cancer. JCI Insight, 2017, 2(11)., DOI 10.1172/jci. insight.91487 Rubio AJ, Porter T, Zhong X. Duality of B Cell-CXCL13 Axis in Tumor Immunology. Front Immunol, 2020, 11:521110., DOI 10.3389/fimmu.2020. 521110 Chunsong H, Yuling H, Li W, Jie X, Gang Z, Qiuping Z, et al. CXC Chemokine Ligand 13 and CC Chemokine Ligand 19 Cooperatively Render Resistance to Apoptosis in B Cell Lineage Acute and Chronic Lymphocytic Leukemia CD23+CD5+B Cells. J Immunol, 2006, 177(10):6713–22., DOI 10.4049/ jimmunol.177.10.6713 Qiuping Z, Jie X, Youxin J, Qun W, Wei J, Chun L, et al. Selectively Frequent Expression of CXCR5 Enhances Resistance to Apoptosis in CD8+CD34+ T Cells from Patients with T-Cell-Lineage Acute Lymphocytic Leukemia. Oncogene, 2005, 24(4):573–84., DOI 10.1038/sj.onc.1208184 Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The Chemokine System in Diverse Forms of Macrophage Activation and Polarization. Trends Immunol, 2004, 25(12):677–86., DOI 10.1016/j.it.2004. 09.015 Rubenstein JL, Wong VS, Kadoch C, Gao H-X, Barajas R, Chen L, et al. CXCL13 Plus Interleukin 10 Is Highly Specific for the Diagnosis of CNS Lymphoma. Blood, 2013, 121(23):4740–8., DOI 10.1182/blood-2013-01-476333 Annovazzi L, Mellai M, Bovio E, Mazzetti S, Pollo B, Schiffer D. Microglia Immunophenotyping in Gliomas. Oncol Lett, 2018, 15(1):998–1006., DOI 10. 3892/ol.2017.7386 Hsieh CH, Jian CZ, Lin LI, Low GS, Ou PY, Hsu C, et al. Potential Role of CXCL13/CXCR5 Signaling in Immune Checkpoint Inhibitor Treatment in Cancer. Cancers, Basel,, 2022, 14(2)., DOI 10.3390/cancers14020294 Quail DF, Joyce JA. The Microenvironmental Landscape of Brain Tumors. Cancer Cell, 2017, 31(3):326–41., DOI 10.1016/j.ccell.2017.02.009 Tomaszewski W, Sanchez-Perez L, Gajewski TF, Sampson JH. Brain Tumor Microenvironment and Host State: Implications for Immunotherapy. Clin Cancer Res, 2019, 25(14):4202–10., DOI 10.1158/1078-0432.ccr-18-1627 Akins EA, Aghi MK, Kumar S. Incorporating Tumor-Associated Macrophages into Engineered Models of Glioma. iScience, 2020, 23(12):101770., DOI 10. 1016/j.isci.2020.101770 Zhou Y, Wang Y, Chen H, Xu Y, Luo Y, Deng Y, et al. Immuno-oncology: Are TAM Receptors in Glioblastoma Friends or Foes? Cell Commun Signal, 2021, 19(1):11., DOI 10.1186/s12964-020-00694-8 Cheng Z, Zhang D, Gong B, Wang P, Liu F. CD163 as a Novel Target Gene of STAT3 Is a Potential Therapeutic Target for Gastric Cancer. Oncotarget, 2017, 8(50):87244–62., DOI 10.18632/oncotarget.20244 Zhao J, Yuan W, Tao C, Sun P, Yang Z, Xu W. M2 Polarization of Monocytes in Ankylosing Spondylitis and Relationship with Inflammation and Structural Damage. APMIS, 2017, 125(12):1070–5., DOI 10.1111/apm. 12757 Chang SJ, Tu HP, Lai YC, Luo CW, Nejo T, Tanaka S, et al. Increased Vascular Adhesion Protein 1, VAP-1, Levels Are Associated with Alternative M2 Macrophage Activation and Poor Prognosis for Human Gliomas. Diagnostics, Basel,, 2020, 10(5)., DOI 10.3390/diagnostics10050256 Poon CC, Gordon PMK, Liu K, Yang R, Sarkar S, Mirzaei R, et al. Differential Microglia and Macrophage Profiles in Human IDH-Mutant and -wild Type Glioblastoma. Oncotarget, 2019, 10(33):3129–43., DOI 10.18632/oncotarget. 26863 Mehani B, Asanigari S, Chung H-J, Dazelle K, Singh A, Hannenhalli S, et al. Immune Cell Gene Expression Signatures in Diffuse Glioma Are Associated with IDH Mutation Status, Patient Outcome and Malignant Cell State, and Highlight the Importance of Specific Cell Subsets in Glioma Biology. Acta Neuropathol Commun, 2022, 10(1):19., DOI 10.1186/s40478- 022-01323-w Gao SH, Liu SZ, Wang GZ, Zhou GB. CXCL13 in Cancer and Other Diseases: Biological Functions, Clinical Significance, and Therapeutic Opportunities. Life, Basel,, 2021, 11(12)., DOI 10.3390/life11121282 Xie Y, Chen Z, Zhong Q, Zheng Z, Chen Y, Shangguan W, et al. M2 Macrophages Secrete CXCL13 to Promote Renal Cell Carcinoma Migration, Invasion, and EMT. Cancer Cell Int, 2021, 21(1):677., DOI 10. 1186/s12935-021-02381-1 Wang Lx., Zhang Sx., Wu Hj., Rong Xl., Guo J. M2b Macrophage Polarization and its Roles in Diseases. J Leukoc Biol, 2019, 106(2):345–58., DOI 10.1002/jlb. 3ru1018-378rr Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, et al. Macrophage Plasticity, Polarization, and Function in Health and Disease. J Cell Physiol, 2018, 233(9):6425–40., DOI 10.1002/jcp. 26429 Guo Y, Hong W, Zhang P, Han D, Fang Y, Tu J, et al. Abnormal Polarization of Macrophage-like Cells in the Peripheral Blood of Patients with Glioma. Oncol Lett, 2020, 20(1):947–54., DOI 10.3892/ol.2020.11602 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610230 EP 1610239 DI 10.3389/pore.2022.1610230 PG 10 ER PT J AU Li, Y Wang, K Zhao, E Li, B Li, Sh Dong, X Yuan, L Yang, H AF Li, Yan Wang, Kunlun Zhao, Erjiang Li, Bingxu Li, Shenglei Dong, Xiaotao Yuan, Ling Yang, Hui TI Prognostic Value of Lactate Dehydrogenase in Second-Line Immunotherapy for Advanced Esophageal Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunotherapy; prognosis; esophageal squamous cell carcinoma; lactate dehydrogenase; programmed death -1 ID immunotherapy; prognosis; esophageal squamous cell carcinoma; lactate dehydrogenase; programmed death -1 AB Background: Immunotherapy is recommended by the NCCN (National Comprehensive Cancer Network) guidelines as the standard second-line treatment for advanced esophageal squamous cell carcinoma (ESCC). Patients with advanced ESCC can benefit from immunotherapy, but the overall survival time (OS) is still not satisfactory. Therefore, it is of great importance to select effective prognostic indicators. Methods: A retrospective follow-up study was conducted from January 2018 to January 2020 among 44 patients with advanced ESCC treated with second-line immune checkpoint inhibitors (programmed death -1 blocking agents) in our hospital. The cutoff values of baseline lactate dehydrogenase (LDH), LDH level at week 8, serum albumin, hemoglobin, neutrophils, monocytes, and platelets were obtained by receiver operating characteristic (ROC) curves. The Kaplan-Meier method was used to analyze the relationship between LDH at baseline, LDH level at week 8, and LDH changes during treatment with progression-free survival (PFS) and OS time. The Cox proportional hazards model was used for univariate and multivariate analyses to determine the predictors of OS. Results: In univariate analysis, we found patients with lower baseline LDH levels (cutoff value: 200 U/L) had a better median PFS (8 months vs. 3 months; HR = 2.420, 95% CI: 1.178–4.971, p = 0.016) and OS (14 months vs. 6 months; HR = 3.637, 95% CI: 1.638–8.074, p = 0.004). The level of LDH at week 8 and the changes in LDH during treatment were not significantly associated with PFS or OS. The multivariate analyses showed that baseline LDH was an independent predictor of PFS (HR = 2.712, 95% CI: 1.147–6.409, p = 0.023) and OS (HR = 6.260, 95%CI: 2.320–16.888, p < 0.001), and the monocyte count (HR = 0.389, 95% CI: 0.162–0.934, p = 0.035) was significantly associated with OS. Conclusion: Serum LDH is a powerful independent factor for PFS and OS in advanced ESCC patients treated with anti-PD-1 therapy. C1 [Li, Yan] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. [Wang, Kunlun] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. [Zhao, Erjiang] The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Department of BiostatisticsZhengzhou, China. [Li, Bingxu] Anyang Tumour Hospital, Department of Radiation OncologyAnyang, China. [Li, Shenglei] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. [Dong, Xiaotao] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. [Yuan, Ling] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. [Yang, Hui] The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation OncologyZhengzhou, China. RP Yang, H (reprint author), The Affiliated Cancer Hospital of Zhengzhou University, Department of Radiation Oncology, Zhengzhou, China. EM dr.huiyang@gmail.com CR Xiao Y, Chen W, Xie Z, Shao Z, Xie H, Qin G, et al.Prognostic Relevance of Lactate Dehydrogenase in Advanced Pancreatic Ductal Adenocarcinoma Patients. Bmc Cancer, 2017, 17(1):25., DOI 10.1186/s12885-016-3012-8 Zhang Z, Li Y, Yan X, Song Q, Wang G, Hu Y, et al.Pretreatment Lactate Dehydrogenase May Predict Outcome of Advanced Non Small-cell Lung Cancer Patients Treated with Immune Checkpoint Inhibitors: A Metaanalysis. Cancer Med, 2019, 8(4):1467–73., DOI 10.1002/cam4.2024 Deme D, Telekes AA Laktatdehidrogenaz, LDH, Prognosztikai Jelentosege Az Onkologiaban. Orvosi Hetilap, 2017, 158(50):1977–88., DOI 10.1556/650.2017. 30890 Zhang X, Guo M, Fan J, Lv Z, Huang Q, Han J, et al.Prognostic Significance of Serum LDH in Small Cell Lung Cancer: A Systematic Review with Meta- Analysis. Cbm, 2016, 16(3):415–23., DOI 10.3233/CBM-160580 Deng T, Zhang J, Meng Y, Zhou Y, Li WHigher Pretreatment Lactate Dehydrogenase Concentration Predicts Worse Overall Survival in Patients with Lung Cancer. Medicine, Baltimore,, 2018, 97(38):e12524., DOI 10.1097/ MD.0000000000012524 Lee DS, Park KR, Kim SJ, Chung MJ, Lee YH, Chang JH, et al.Serum Lactate Dehydrogenase Levels at Presentation in Stage IV Non-small Cell Lung Cancer: Predictive Value of Metastases and Relation to Survival Outcomes. Tumor Biol, 2016, 37(1):619–25., DOI 10.1007/s13277-015-3776-5 Koukourakis MI, Giatromanolaki A, Sivridis E, Gatter KC, Harris ALLactate Dehydrogenase 5 Expression in Operable Colorectal Cancer: Strong Association with Survival and Activated Vascular Endothelial Growth Factor Pathway-A Report of the Tumour Angiogenesis Research Group. Jco, 2006, 24(26):4301–8., DOI 10.1200/JCO.2006.05.9501 Wang X, Zhang B, Chen X, Mo H, Wu D, Lan B, et al.Lactate Dehydrogenase and Baseline Markers Associated with Clinical Outcomes of Advanced Esophageal Squamous Cell Carcinoma Patients Treated with Camrelizumab, SHR-1210), a Novel anti-PD-1 Antibody. Thorac Cancer, 2019, 10(6): 1395–401., DOI 10.1111/1759-7714.13083 Gatenby RA, Gillies RJWhy Do Cancers Have High Aerobic Glycolysis? Nat Rev Cancer, 2004, 4(11):891–9., DOI 10.1038/nrc1478 Yamada Y, Nakamura K, Aoki S, TobiumeM, Zennami K, Kato Y, et al.Lactate Dehydrogenase, Gleason Score and HER-2 Overexpression Are Significant Prognostic Factors for M1b Prostate Cancer. Oncol Rep, 2011, 25(4):937–44., DOI 10.3892/or.2011.1154 Koukourakis MI, Giatromanolaki A, Giatromanolaki A, Sivridis E, Bougioukas G, Didilis V, et al.Lactate Dehydrogenase-5, LDH-5, Overexpression in Nonsmall- cell Lung Cancer Tissues Is Linked to Tumour Hypoxia, Angiogenic Factor Production and Poor Prognosis. Br J Cancer, 2003, 89(5):877–85., DOI 10.1038/sj.bjc.6601205 Jong C, Deneer VHM, Kelder JC, Ruven H, Egberts TCG, Herder GJMAssociation between Serum Biomarkers CEA and LDH and Response in Advanced Non-small Cell Lung Cancer Patients Treated with Platinumbased Chemotherapy. Thorac Cancer, 2020, 11(7):1790–800., DOI 10.1111/ 1759-7714.13449 Wei X-l., Zhang D-s., He M-m., Jin Y, Wang D-s., Zhou Y-x., et al.The Predictive Value of Alkaline Phosphatase and Lactate Dehydrogenase for Overall Survival in Patients with Esophageal Squamous Cell Carcinoma. Tumor Biol, 2016, 37(2):1879–87., DOI 10.1007/s13277- 015-3851-y Rawat K, Syeda S, Shrivastava AHyperactive Neutrophils Infiltrate Vital Organs of Tumor Bearing Host and Contribute to Gradual Systemic Deterioration via Upregulated NE, MPO and MMP-9 Activity. Immunol Lett, 2022, 241:35–48., DOI 10.1016/j.imlet.2021.12.001 Liu LT, Liang YJ, Guo SS, Xie Y, Jia GD, Wen DX, et al.Identifying Distinct Risks of Treatment Failure in Nasopharyngeal Carcinoma: Study Based on the Dynamic Changes in Peripheral Blood Lymphocytes, Monocytes, N Classification, and Plasma Epstein-Barr Virus DNA. Head & Neck, 2022, 44(1):34–45., DOI 10.1002/hed.26897 Tu J, Wen L, Huo Z, Wang B, Wang Y, Liao H, et al.Predictive Value of Dynamic Change of Haemoglobin Levels during Therapy on Treatment Outcomes in Patients with Enneking Stage IIB Extremity Osteosarcoma. Bmc Cancer, 2018, 18(1):428., DOI 10.1186/s12885-018-4279-8 Jung K, Heishi T, Khan OF, Kowalski PS, Incio J, Rahbari NN, et al.Ly6Clo Monocytes Drive Immunosuppression and Confer Resistance to Anti- VEGFR2 Cancer Therapy. J Clin Invest, 2017, 127(8):3039–51., DOI 10.1172/ JCI93182 Sidibe A, Ropraz P, Jemelin S, Emre Y, Poittevin M, Pocard M, et al.Angiogenic Factor-Driven Inflammation Promotes Extravasation of Human Proangiogenic Monocytes to Tumours. Nat Commun, 2018, 9(1):355., DOI 10.1038/s41467-017-02610-0 Jeong J, Kim DK, Park J-H, Park DJ, Lee H-J, Yang H-K, et al.Tumor- Infiltrating Neutrophils and Non-classical Monocytes May Be Potential Therapeutic Targets for HER2negative Gastric Cancer. Immune Netw, 2021, 21(4):e31., DOI 10.4110/in.2021.21.e31 Sheng SL, Liu JJ, Dai YH, Sun XG, Xiong XP, Huang GKnockdown of Lactate Dehydrogenase A Suppresses Tumor Growth and Metastasis of Human Hepatocellular Carcinoma. Febs J, 2012, 279(20):3898–910., DOI 10.1111/j. 1742-4658.2012.08748.x Xie H, Hanai J-i., Ren J-G, Kats L, Burgess K, Bhargava P, et al.Targeting Lactate Dehydrogenase-A Inhibits Tumorigenesis and Tumor Progression in Mouse Models of Lung Cancer and Impacts Tumor-Initiating Cells. Cell Metab, 2014, 19(5):795–809., DOI 10.1016/j.cmet.2014.03.003 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610245 EP 1610253 DI 10.3389/pore.2022.1610245 PG 9 ER PT J AU Evans, H O’Sullivan, B Hughes, F Charles, K Robertson, L Taniere, P Diaz-Cano, S AF Evans, Harriet O’Sullivan, Brendan Hughes, Frances Charles, Kathryn Robertson, Lee Taniere, Philippe Diaz-Cano, Salvador TI PD-L1 Testing in Urothelial Carcinoma: Analysis of a Series of 1401 Cases Using Both the 22C3 and SP142 Assays SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PD-L1; urothelial bladder carcinoma; predictive marker; Pembrolizumab; Atezolizumab ID PD-L1; urothelial bladder carcinoma; predictive marker; Pembrolizumab; Atezolizumab AB Immune checkpoint blockade (ICB) drugs are a novel, effective treatment for advanced urothelial carcinoma. Worldwide, several different ICB drugs are approved, each developed and clinically validated with a specific PD-L1 compound diagnostic assay. As a result, PD-L1 testing workflows in routine practice are complex: requiring multiple assays across two platforms, with each assay having a different method of interpretation. Our service tested 1,401 urothelial carcinoma cases for PD-L1 expression, using both the 22C3 PharmDx assay (required prior to Pembrolizumab therapy) and SP142 assay (required prior to Atezolizumab therapy). Of the 1,401 cases tested, 621 cases (44%) were tested with both the 22C3 PharmDx and SP142 assays, 492 cases (35%) with 22C3 PharmDx only, and 288 cases (21%) with SP142 only. Each assay was used and interpreted according to the manufacturer’s guidelines. The rate of positivity we observed was 26% with the 22C3 assay and 31% with the SP142 assay, similar to the pre-licensing studies for both drugs. The discrepancy observed between the assays was 11%, which reinforces the requirement for utilisation of the correct assay for each agent, and limits potential cross-utility of assays. This aspect must be considered when setting up a PD-L1 testing strategy in laboratories where both Pembrolizumab and Atezolizumab are available for the treatment of urothelial carcinoma but also has broader implications for testing of other cancers where multiple ICB drugs and their respective assays are approved. C1 [Evans, Harriet] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic ServiceBirmingham, UK. [O’Sullivan, Brendan] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic ServiceBirmingham, UK. [Hughes, Frances] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic ServiceBirmingham, UK. [Charles, Kathryn] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic ServiceBirmingham, UK. [Robertson, Lee] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic ServiceBirmingham, UK. [Taniere, Philippe] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic ServiceBirmingham, UK. [Diaz-Cano, Salvador] Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic ServiceBirmingham, UK. RP Evans, H (reprint author), Queen Elizabeth Hospital Birmingham, Molecular Pathology Diagnostic Service, Birmingham, UK. EM harriet.evans4@nhs.net CR Cheung CC, Barnes P, Bigras G, Boerner S, Butany J, Calabrese F, et al. Fit-forpurpose PD-L1 Biomarker Testing for Patient Selection in Immuno-Oncology: Guidelines for Clinical Laboratories from the Canadian Association of Pathologists- Association Canadienne Des Pathologistes, CAP-ACP). Appl Immunohistochem Mol Morphol, 2019, 27(10):699–714., DOI 10.1097/pai.0000000000000800 Eckstein M, Cimadamore A, Hartmann A, Lopez-Beltran A, Cheng L, Scarpelli M, et al. PD-L1 Assessment in Urothelial Carcinoma: a Practical Approach. Ann Transl Med, 2019, 7:690–22., DOI 10.21037/atm.2019.10.24 Powles T, Walker J, Andrew Williams J, Bellmunt J. The Evolving Role of PDL1 Testing in Patients with Metastatic Urothelial Carcinoma. Cancer Treat Rev, 2020, 82:101925., DOI 10.1016/j.ctrv.2019.101925 Zajac M, Scott M, Ratcliffe M, Scorer P, Barker C, Al-Masri H, et al. Concordance Among Four Commercially Available, Validated Programmed Cell Death Ligand-1 Assays in Urothelial Carcinoma. Diagn Pathol, 2019, 14(1):99–10., DOI 10.1186/s13000-019-0873-6 National Institute for Health and Care Excellence. Atezolizumab for Untreated PD-L1-Positive Advanced Urothelial Cancer when Cisplatin Is Unsuitable. last update, 2021). Available at https://www.nice.org.uk/guidance/ta739/chapter/ 1-Recommendations Oct 27, Accessed February 28, 2022) National Institute for Health and Care Excellence.Atezolizumab for Treating Locally Advanced or Metastatic Urothelial Carcinoma after Platinum-Containing Chemotherapy. last update, 2018). Available at https://www.nice.org.uk/guidance/ ta525/chapter/1-Recommendations June 13, Accessed February 28, 2022) European Medicines Agency, -last Update, Tecentriq/Atezolizumab. Summary of Product Characteristics. last update, 2021). Available at: https://www.ema.europa. eu/en/documents/product-information/tecentriq-epar-product-information_en.pdf Nov 10, Accessed February 28, 2022) European Medicines Agency. Keytruda/Pembrolizumab. Summary of Product Characteristics. last update, 2022). Available at https://www.ema.europa.eu/en/ documents/product-information/keytruda-epar-product-information_en.pdf Jan 11, Accessed February 28, 2022) Balar AV, Castellano D, O’Donnell PH, Grivas P, Vuky J, Powles T, et al. Firstline Pembrolizumab in Cisplatin-Ineligible Patients with Locally Advanced and Unresectable or Metastatic Urothelial Cancer, KEYNOTE-052): a Multicentre, Single-Arm, Phase 2 Study. Lancet Oncol, 2017, 18(11): 1483–92., DOI 10.1016/s1470-2045(17)30616-2 Fradet Y, Bellmunt J, Vaughn DJ, Lee JL, Fong L, Vogelzang NJ, et al. Randomized Phase III KEYNOTE-045 Trial of Pembrolizumab versus Paclitaxel, Docetaxel, or Vinflunine in Recurrent Advanced Urothelial Cancer: Results of >2 Years of Follow-Up. Ann Oncol, 2019, 30(6):970–6., DOI 10.1093/annonc/mdz127 Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, et al. Atezolizumab as First-Line Treatment in Cisplatin-Ineligible Patients with Locally Advanced and Metastatic Urothelial Carcinoma: a Single-Arm, Multicentre, Phase 2 Trial. The Lancet, 2017, 389(10064):67–76., DOI 10. 1016/s0140-6736(16)32455-2 Rosenberg JE, Hoffman-Censits J, Powles T, Van Der Heijden MS, Balar AV, Necchi A, et al. Atezolizumab in Patients with Locally Advanced and Metastatic Urothelial Carcinoma Who Have Progressed Following Treatment with Platinum-Based Chemotherapy: a Single-Arm, Multicentre, Phase 2 Trial. The Lancet, 2016, 387(10031):1909–20., DOI 10.1016/s0140-6736(16)00561-4 Lee KS, Choe G. Programmed Cell Death-Ligand 1 Assessment in Urothelial Carcinoma: Prospect and Limitation. J Pathol Transl Med, 2021, 55(3): 163–70., DOI 10.4132/jptm.2021.02.22 Eckstein M, Erben P, Kriegmair MC, Worst TS, Weiss C-A, Wirtz RM, et al. Performance of the Food and Drug Administration/EMA-Approved Programmed Cell Death Ligand-1 Assays in Urothelial Carcinoma with Emphasis on Therapy Stratification for First-Line Use of Atezolizumab and Pembrolizumab. Eur J Cancer, 2019, 106:234–43., DOI 10.1016/j.ejca.2018. 11.007 Hodgson A, Slodkowska E, Jungbluth A, Liu SK, Vesprini D, Enepekides D, et al. PD-L1 Immunohistochemistry Assay Concordance in Urothelial Carcinoma of the Bladder and Hypopharyngeal Squamous Cell Carcinoma. Am J Surg Pathol, 2018, 42(8):1059–66., DOI 10.1097/pas. 0000000000001084 Schwamborn K,Ammann JU, Knuchel R, Hartmann A, Baretton G, Lasitschka F, et al. Multicentric Analytical Comparability Study of Programmed Death- Ligand 1 Expression on Tumor-Infiltrating Immune Cells and Tumor Cells in Urothelial Bladder Cancer Using Four Clinically Developed Immunohistochemistry Assays. Virchows Arch, 2019, 475(5):599–608., DOI 10.1007/s00428-019-02610-z Tretiakova M, Fulton R, Kocherginsky M, Long T, Ussakli C, Antic T, et al. Concordance Study of PD-L1 Expression in Primary and Metastatic Bladder Carcinomas: Comparison of Four Commonly Used Antibodies and RNA Expression. Mod Pathol, 2018, 31(4):623–32., DOI 10.1038/modpathol. 2017.188 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610260 EP 1610264 DI 10.3389/pore.2022.1610260 PG 5 ER PT J AU Benke, M Farkas, N Hegyi, P Tinusz, B Sarlos, P Eross, B Szemes, K Vorhendi, N Szakacs, Zs Szucs, AF Benke, Marton Farkas, Nelli Hegyi, Peter Tinusz, Benedek Sarlos, Patricia Eross, Balint Szemes, Kata Vorhendi, Nora Szakacs, Zsolt Szucs, Akos TI Preoperative Serum Carbohydrate Antigen 19-9 Levels Cannot Predict the Surgical Resectability of Pancreatic Cancer: A Meta-Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE biomarker; prognosis; pancreas adenocarcinoma; carbohydrate antigen 19-9; pancreatic surgery ID biomarker; prognosis; pancreas adenocarcinoma; carbohydrate antigen 19-9; pancreatic surgery AB Background and Aims: Pancreatic ductal adenocarcinoma has one of theworst prognosis of all malignancies. This investigated the relationship between the preoperative serum carbohydrate antigen 19-9 and surgical resectability. Methods: A systematic search was performed in three databases (MEDLINE, EMBASE, and Web of Science) to compare the surgical resectability of pancreatic ductal adenocarcinoma in patients with high and low preoperative serum carbohydrate antigen 19-9 values. The receiving operating characteristic curves were constructed and the weighted mean differences for preoperative serumcarbohydrate antigen 19-9 levels of resectable and unresectable groups of patients were calculated. The PROSPERO registration number is CRD42019132522. Results: Results showed that there was a significant difference in resectability between the low and high carbohydrate antigen 19-9 groups. Six out of the eight studies utilised receiver operating characteristic curves in order to find the cut-off preoperative carbohydrate antigen 19-9 levels marking unresectability. The overall result from the pooled area under curve values from the receiver operating characteristic curves was 0.794 (CI: 0.694–0.893), showing that the preoperative carbohydrate antigen 19-9 level is a “fair” marker of resectability. The result of the pooled weighted mean differences was 964 U/ml (p < 0.001) showing that there is a significant carbohydrate antigen 19-9 difference between the resectable and unresectable groups. Based on the results of the I-squared test, the result was 87.4%, accounting for “considerable” heterogeneity within the population. Conclusion: Carbohydrate antigen 19-9 is not a reliable marker of unresectability, it should not be used on its own in surgical decision-making. C1 [Benke, Marton] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. [Farkas, Nelli] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Hegyi, Peter] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Tinusz, Benedek] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Sarlos, Patricia] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Eross, Balint] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Szemes, Kata] University of Pecs, Medical School, First Department of Medicine, Division of GastroenterologyPecs, Hungary. [Vorhendi, Nora] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Szakacs, Zsolt] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Szucs, Akos] Semmelweis University, 1st Department of SurgeryBudapest, Hungary. RP Szucs, (reprint author), Semmelweis University, 1st Department of Surgery, Budapest, Hungary. EM szucs.akos@gmail.com CR Adamska A, Domenichini A, Falasca M. Pancreatic Ductal Adenocarcinoma: Current and Evolving Therapies. Int J Mol Sci, 2017, 18(7)., DOI 10.3390/ ijms18071338 Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, et al. Pancreatic Cancer. Nat Rev Dis primers, 2016, 2:16022., DOI 10.1038/nrdp.2016.22 KimKS,Kwon J, KimK,Chie EK. Impact of ResectionMarginDistance on Survival of Pancreatic Cancer: A Systematic Review and Meta-Analysis. Cancer Res Treat, 2017, 49(3):824–33., DOI 10.4143/crt.2016.336 Tempero MA,MalafaMP, Al-HawaryM, Asbun H, Bain A, Behrman SW, et al. PancreaticAdenocarcinoma,Version 2.2017,NCCNClinical Practice Guidelines in Oncology. J NatlComprCancNetw(2017, 15(8):1028–61., DOI 10.6004/jnccn.2017. 0131 Buanes TA. Role of Surgery in Pancreatic Cancer. World J Gastroenterol, 2017, 23(21):3765–70., DOI 10.3748/wjg.v23.i21.3765 Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: the PRISMA Statement. Int J Surg, 2010, 8(5):336–41., DOI 10.1016/j.ijsu.2010.02.007 Kamarudin AN, Cox T, Kolamunnage-Dona R. Time-dependent ROC Curve Analysis in Medical Research: Current Methods and Applications. BMC Med Res Methodol, 2017, 17(1):53., DOI 10.1186/s12874-017-0332-6 Jones CM, Athanasiou T. Summary Receiver Operating Characteristic Curve Analysis Techniques in the Evaluation of Diagnostic Tests. Ann Thorac Surg, 2005, 79(1):16–20., DOI 10.1016/j.athoracsur.2004.09.040 Albatanony A, Alseesi A. The Value of Serum Carbohydrate Antigen 19-9 as a Predictor of Resectability in Pancreatic Adenocarcinoma. Egypt J Surg, 2015, 34:226., DOI 10.4103/1110-1121.167382 Almadi M, Alharbi O, Azzam N, Altayeb M, Javed M, Alsaif F, et al. Clinical Predictors of Resectability of Pancreatic Adenocarcinoma. Saudi J Gastroenterol, 2013, 19(6):278–85., DOI 10.4103/1319-3767.121036 Kilic M, Gocmen E, Tez M, Ertan T, Keskek M, Koc M. Value of Preoperative Serum CA 19-9 Levels in Predicting Resectability for Pancreatic Cancer. Can J Surg, 2006, 49(4):241–4. Retriewed from: https://www.ncbi.nlm.nih.gov/pmc/issues/202076/ Kim YC, Kim HJ, Park JH, Park DI, Cho YK, Sohn CI, et al. Can Preoperative CA19-9 and CEA Levels Predict the Resectability of Patients with Pancreatic Adenocarcinoma? J Gastroenterol Hepatol, 2009, 24(12):1869–75., DOI 10.1111/j.1440-1746.2009.05935.x Luo G, Xiao Z, Long J, Liu Z, Liu L, Liu C, et al. CA125 Is Superior to CA19-9 in Predicting the Resectability of Pancreatic Cancer. J Gastrointest Surg, 2013, 17(12):2092–8., DOI 10.1007/s11605-013- 2389-9 Ong SL, Garcea G, Thomasset SC, Mann CD, Neal CP, Abu AmaraM, et al. Surrogate Markers of Resectability in Patients Undergoing Exploration of Potentially Resectable Pancreatic Adenocarcinoma. J Gastrointest Surg, 2008, 12(6):1068–73., DOI 10.1007/s11605-007-0422-6 Santucci N, Facy O, Ortega-Deballon P, Lequeu J-B, Rat P, Rat P. CA 19-9 Predicts Resectability of Pancreatic Cancer Even in Jaundiced Patients. Pancreatology, 2018, 18(6):666–70., DOI 10.1016/j.pan.2018.07.001 Zhang S, Wang Y-M, Sun C-D, Lu Y, Wu L-Q. Clinical Value of Serum CA19-9 Levels in Evaluating Resectability of Pancreatic Carcinoma. World J Gastroenterol, 2008, 14(23):3750–3., DOI 10.3748/wjg.14.3750 Poruk KE, Firpo MA, Adler DG, Mulvihill SJ. Screening for Pancreatic Cancer: why, how, and who? Ann Surg, 2013, 257(1):17–26., DOI 10.1097/ sla.0b013e31825ffbfb Isaji S, Mizuno S, Windsor JA, Bassi C, Fernandez-del Castillo C, Hackert T, et al. International Consensus on Definition and Criteria of Borderline Resectable Pancreatic Ductal Adenocarcinoma 2017. Pancreatology, 2018, 18(1):2–11., DOI 10.1016/j.pan.2017.11.011 Daamen LA, Groot VP, Goense L, Wessels FJ, Borel Rinkes IH, Intven MPW, et al. The Diagnostic Performance of CT versus FDG PET-CT for the Detection of Recurrent Pancreatic Cancer: a Systematic Review and Meta-Analysis. Eur J Radiol, 2018, 106:128–36., DOI 10.1016/j.ejrad.2018. 07.010 Tempero M. Active Systemic Treatment of Pancreatic Cancer. J Natl Compr Canc Netw, 2017, 15(5s):723–5., DOI 10.6004/jnccn.2017.0084 Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic Cancer. Lancet, 2016, 388(10039):73–85., DOI 10.1200/JCO.2006.08.2644 Ryan DP, Hong TS, Bardeesy N. Pancreatic Adenocarcinoma. N Engl J Med, 2014, 371(11):1039–49., DOI 10.1056/nejmra1404198 Takacs T, Czako L, Dubravcsik Z, Farkas G, Hegyi P, Hritz I, et al. Chronic Pancreatitis Evidence Based Management Guidelines of the Hungarian Pancreatic Study Group. Orvosi hetilap, 2015, 156(7):262–88., DOI 10.4103/1110-1121.167382 Rawla P, Sunkara T, Gaduputi V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J Oncol, 2019, 10(1): 10–27., DOI 10.14740/wjon1166 Osteikoetxea X, Benke M, Rodriguez M, Paloczi K, Sodar BW, Szvicsek Z, et al. Detection and Proteomic Characterization of Extracellular Vesicles in Human Pancreatic Juice. Biochem biophysical Res Commun, 2018, 499(1):37–43., DOI 10.1016/j.bbrc.2018.03.107 Chang JC, KundrandaM. Novel Diagnostic and Predictive Biomarkers in Pancreatic Adenocarcinoma. Int J Mol Sci, 2017, 18(3):667., DOI 10.3390/ijms18030667 Herreros-Villanueva M, Gironella M, Castells A, Bujanda L. Molecular Markers in Pancreatic Cancer Diagnosis. Clinica Chim Acta, 2013, 418: 22–9., DOI 10.1016/j.cca.2012.12.025 Ballehaninna UK, Chamberlain RS. The Clinical Utility of Serum CA 19-9 in the Diagnosis, Prognosis and Management of Pancreatic Adenocarcinoma: An Evidence Based Appraisal. J Gastrointest Oncol, 2012, 3(2):105–19., DOI 10. 3978/j.issn.2078-6891.2011.021 Okano K, Suzuki Y. Strategies for Early Detection of Resectable Pancreatic Cancer. World J Gastroenterol, 2014, 20(32):11230–40., DOI 10.3748/wjg.v20.i32.11230 LockerGY, Hamilton S, Harris J, Jessup JM,KemenyN,Macdonald JS, et al. ASCO 2006 Update of Recommendations for the Use of Tumor Markers inGastrointestinal Cancer. J Clin Oncol, 2006, 24(33):5313–27., DOI 10.1200/jco.2006.08.2644 Scara S, Bottoni P, Scatena R. CA 19-9: Biochemical and Clinical Aspects. Adv Exp Med Biol, 2015, 867:247–60., DOI 10.1007/978-94-017-7215-0_15 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610266 EP 1610274 DI 10.3389/pore.2022.1610266 PG 9 ER PT J AU Yang, Y Yu, J Xiong, Y Xiao, J Dai, D Zhang, F AF Yang, Yong Yu, Jieqing Xiong, Yuanping Xiao, Jiansheng Dai, Daofeng Zhang, Feng TI Prognostic Analysis of Differentially Expressed DNA Damage Repair Genes in Bladder Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarkers; prognosis; bladder cancer; differentially expressed genes; DNA damage repair genes ID biomarkers; prognosis; bladder cancer; differentially expressed genes; DNA damage repair genes AB Bladder cancer (BCa) is the tenth most common tumor in humans. DNA damage repair genes (DDRGs) play important roles in many malignant tumors; thus, their functions in BCa should also be explored. We performed a comprehensive analysis of the expression profiles of DDRGs in 410 BCa tumors and 19 normal tissues from The Cancer Genome Atlas database. We identified 123 DDRGs differentially expressed between BCa tumors and normal tissues, including 95 upregulated and 28 downregulated genes. We detected 22 DDRGs associated with overall survival (OS) of patients with BCa by performing univariate Cox regression analysis. To explore the interactions between OS-associated DDRGs, we constructed a PPI network, which showed that the top six DDRGs (CDCA2, FOXM1, PBK, RRM2, ORC1, and HDAC4) with the highest scores in the PPI network might play significant roles in OS of BCa. Moreover, to investigate the latent regulatory mechanism of these OS-associated DDRGs, we analyzed the transcription factors (TFs)- DDRGs regulatory network. The core seven TFs (NCAPG, DNMT1, LMNB1, BRCA1, E2H2, CENPA, and E2F7) were shown to be critical regulators of the OS-related DDRGs. The 22 DDRGs were incorporated into a stepwise multivariable Cox analysis. Then, we built the index of risk score based on the expression of 8 DDRGs (CAD, HDAC10, JDP2, LDLR, PDGFRA, POLA2, SREBF1, and STAT1). The p-value < 0.0001 in the Kaplan–Meier survival plot and an area under the ROC curve (AUC) of 0.771 in TCGA-BLCA training dataset suggested the high specificity and sensitivity of the prognostic index. Furthermore, we validated the risk score in the internal TCGA-BLCA and an independent GSE32894 dataset, with AUC of 0.743 and 0.827, respectively. More importantly, the multivariate Cox regression and stratification analysis demonstrated that the predictor was independent of various clinical parameters, including age, tumor stage, grade, and number of positive tumor lymph nodes. In summary, a panel of 8 DNA damage repair genes associated with overall survival in bladder cancer may be a useful prognostic tool. C1 [Yang, Yong] Jiangxi Provincial People’s Hospital Affiliated to Nanchang Medical College, Department of Otolaryngology Head and Neck SurgeryNanchang, China. [Yu, Jieqing] Nanchang University, First Affiliated Hospital, Department of Otolaryngology-Head and Neck SurgeryNanchang, China. [Xiong, Yuanping] Nanchang University, First Affiliated Hospital, Department of Otolaryngology-Head and Neck SurgeryNanchang, China. [Xiao, Jiansheng] The First Affiliated Hospital of Nanchang University, Department of General SurgeryNanchang, China. [Dai, Daofeng] Nanchang University, First Affiliated Hospital, Department of Otolaryngology-Head and Neck SurgeryNanchang, China. [Zhang, Feng] Nanchang University, The Institute of Translational Medicine, The National Engineering Research Center for Bioengineering Drugs and the TechnologiesNanchang, China. RP Zhang, F (reprint author), Nanchang University, The Institute of Translational Medicine, The National Engineering Research Center for Bioengineering Drugs and the Technologies, Nanchang, China. EM fengzhang0709@hotmail.com CR Siegel RL, Miller KD, Jemal A Cancer Statistics, 2019. CA Cancer J Clin, 2019, 69(1):7–34., DOI 10.3322/caac.21551 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Kamat AM, Hahn NM, Efstathiou JA, Lerner SP, Malmstrom P-U, Choi W, et al. Bladder Cancer. Lancet, 2016, 388(10061):2796–810., DOI 10.1016/s0140- 6736(16)30512-8 Antoni S, Ferlay J, Soerjomataram I, Znaor A, Jemal A, Bray F Bladder Cancer Incidence and Mortality: A Global Overview and Recent Trends. Eur Urol, 2017, 71(1):96–108., DOI 10.1016/j.eururo.2016.06.010 Baumann BC, Guzzo TJ, He J, Keefe SM, Tucker K, Bekelman JE, et al. A Novel Risk Stratification to Predict Local-Regional Failures in Urothelial Carcinoma of the Bladder after Radical Cystectomy. Int J Radiat Oncol Biol Phys, 2012, 85(1):81–8., DOI 10.1016/j.ijrobp.2012.03.007 Paner GP, Stadler WM, Hansel DE, Montironi R, Lin DW, Amin MB Updates in the Eighth Edition of the Tumor-Node-Metastasis Staging Classification for Urologic Cancers. Eur Urol, 2018, 73(4):560–9., DOI 10.1016/j.eururo.2017. 12.018 Gallan AJ, Choy B, Paner GP Contemporary Grading and Staging of Urothelial Neoplasms of the Urinary Bladder. Surg Pathol Clin, 2018, 11(4):775–95., DOI 10.1016/j.path.2018.07.006 Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G The Hallmarks of Aging. Cell, 2013, 153(6):1194–217., DOI 10.1016/j.cell.2013.05.039 Lieber MR The Mechanism of Double-Strand DNA Break Repair by the Nonhomologous DNA End-Joining Pathway. Annu Rev Biochem, 2010, 79: 181–211., DOI 10.1146/annurev.biochem.052308.093131 Shimizu I, Yoshida Y, Suda M, Minamino T DNA Damage Response and Metabolic Disease. Cel Metab, 2014, 20(6):967–77., DOI 10.1016/j.cmet.2014. 10.008 Wang G, Guo S, Zhang W, Li Z, Xu J, Li D, et al. A Comprehensive Analysis of Alterations in DNA Damage Repair Pathways Reveals a Potential Way to Enhance the Radio-Sensitivity of Esophageal Squamous Cell Cancer. Front Oncol, 2020, 10:575711., DOI 10.3389/fonc.2020.575711 Xu N, Ke Z-B, Lin X-D, Chen Y-H, Wu Y-P, Chen Y, et al. Development and Validation of a Molecular Prognostic index of Bladder Cancer Based on Immunogenomic Landscape Analysis. Cancer Cel Int, 2020, 20:302., DOI 10. 1186/s12935-020-01343-3 Xiao H, Wang B, Xiong HX, Guan JF, Wang J, Tan T, et al. A Novel Prognostic index of Hepatocellular Carcinoma Based on Immunogenomic Landscape Analysis. J Cel Physiol, 2020, 236:2572–91., DOI 10.1002/jcp.30015 Wang L, Shi J, Huang Y, Liu S, Zhang J, Ding H, et al. A Six-Gene Prognostic Model Predicts Overall Survival in Bladder Cancer Patients. Cancer Cel Int, 2019, 19:229., DOI 10.1186/s12935-019-0950-7 Heagerty PJ, Lumley T, Pepe MS Time-dependent ROC Curves for Censored Survival Data and a Diagnostic Marker. Biometrics, 2000, 56(2):337–44., DOI 10.1111/j.0006-341x.2000.00337.x Mei S, Meyer CA, Zheng R, Qin Q, Wu Q, Jiang P, et al. Cistrome Cancer: A Web Resource for Integrative Gene Regulation Modeling in Cancer. Cancer Res, 2017, 77(21):e19–e22., DOI 10.1158/0008-5472.CAN-17-0327 Bader GD, Hogue CW An Automated Method for Finding Molecular Complexes in Large Protein Interaction Networks. BMC Bioinform, 2003, 4:2., DOI 10.1186/1471-2105-4-2 Yin M, Grivas P, Wang Q-E, Mortazavi A, Emamekhoo H, Holder SL, et al. Prognostic Value of DNA Damage Response Genomic Alterations in Relapsed/Advanced Urothelial Cancer. Oncologist, 2020, 25(8):680–8., DOI 10.1634/theoncologist.2019-0851 Curia MC, Catalano T, Aceto GM MUTYH: Not Just Polyposis. World J Clin Oncol, 2020, 11(7):428–49., DOI 10.5306/wjco.v11.i7.428 Lappalainen T, Scott AJ, Brandt M, Hall IM Genomic Analysis in the Age of Human Genome Sequencing. Cell, 2019, 177(1):70–84., DOI 10.1016/j.cell. 2019.02.032 Zhao L, Lee VHF, Ng MK, Yan H, BijlsmaMFMolecular Subtyping of Cancer: Current Status and Moving toward Clinical Applications. Brief Bioinform, 2019, 20(2):572–84., DOI 10.1093/bib/bby026 Lerner SP, McConkey DJ, Hoadley KA, Chan KS, Kim WY, Radvanyi F, et al. Bladder Cancer Molecular Taxonomy: Summary from a Consensus Meeting. Bladder Cancer, 2016, 2(1):37–47., DOI 10.3233/BLC-150037 Sanli O, Dobruch J, Knowles MA, Burger M, Alemozaffar M, Nielsen ME, et al. Bladder Cancer. Nat Rev Dis Primers, 2017, 3:17022., DOI 10.1038/nrdp. 2017.22 Aine M, Eriksson P, Liedberg F, Hoglund M, Sjodahl G On Molecular Classification of Bladder Cancer: Out of One, Many. Eur Urol, 2015, 68(6): 921–3., DOI 10.1016/j.eururo.2015.07.021 Allione A, Pardini B, Viberti C, Oderda M, Allasia M, Gontero P, et al. The Prognostic Value of Basal DNA Damage Level in Peripheral Blood Lymphocytes of Patients Affected by Bladder Cancer. Urol Oncol Semin Original Invest, 2018, 36(5):241.e15–241.e23., DOI 10.1016/j.urolonc.2018. 01.006 Duan W, Du L, Jiang X, Wang R, Yan S, Xie Y, et al. Identification of a Serum Circulating lncRNA Panel for the Diagnosis and Recurrence Prediction of Bladder Cancer. Oncotarget, 2016, 7(748):78850–8., DOI 10.18632/oncotarget. 12880 Fang Z, Dai W, Wang X, Chen W, Shen C, Ye G, et al. Circulating miR-205: A Promising Biomarker for the Detection and Prognosis Evaluation of Bladder Cancer. Tumor Biol, 2016, 37(6):8075–82., DOI 10.1007/s13277- 015-4698-y Abudurexiti M, Xie H, Jia Z, Zhu Y, Zhu Y, Shi G, et al. Development and External Validation of a Novel 12-Gene Signature for Prediction of Overall Survival in Muscle-Invasive Bladder Cancer. Front Oncol, 2019, 9(9):856., DOI 10.3389/fonc.2019.00856 Ingelmo-Torres M, Lozano JJ, Izquierdo L, Carrion A, Costa M, Gomez L, et al. Urinary Cell microRNA-Based Prognostic Classifier for Non-muscle Invasive Bladder Cancer. Oncotarget, 2017, 8(11):18238–47., DOI 10.18632/oncotarget. 15315 Chen S, Zhang N, Shao J, Wang T, Wang X A Novel Gene Signature Combination Improves the Prediction of Overall Survival in Urinary Bladder Cancer. J Cancer, 2019, 10(23):5744–53., DOI 10.7150/jca.30307 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610267 EP 1610278 DI 10.3389/pore.2022.1610267 PG 12 ER PT J AU Brenner, S Hartzendorf, S Vogt, P Maier, E Etminan, N Jung, E Wick, W Sahm, F Winkler, F Ratliff, M AF Brenner, Steffen Hartzendorf, Sebastian Vogt, Philip Maier, Elena Etminan, Nima Jung, Erik Wick, Wolfgang Sahm, Felix Winkler, Frank Ratliff, Miriam TI Progression Patterns in Non-Contrast-Enhancing Gliomas Support Brain Tumor Responsiveness to Surgical Lesions SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tumor progression; astrocytoma; surgical lesioning; tumor cell network; tumor microtubes ID tumor progression; astrocytoma; surgical lesioning; tumor cell network; tumor microtubes AB Purpose: The overall benefit of surgical treatments for patients with glioma is undisputed. We have shown preclinically that brain tumor cells form a network that is capable of detecting damage to the tumor, and repair itself. The aim of this study was to determine whether a similar mechanism might contribute to local recurrence in the clinical setting. Methods: We evaluated tumor progression patterns of 24 initially non-contrast-enhancing gliomas that were partially resected or biopsied. We measured the distance between the new contrast enhancement developing over time, and prior surgical lesioning, and evaluated tumor network changes in response to sequential resections by quantifying tumor cells and tumor networks with specific stainings against IDH1-R132H. Results: We found that new contrast enhancement appeared within the residual, nonenhancing tumor mass in 21/24 patients (87.5%). The location of new contrast enhancement within the residual tumor region was non-random; it occurred adjacent to the wall of the resection cavity in 12/21 patients (57.1%). Interestingly, the density of the glioma cell network increased in all patient tumors between initial resection or biopsy and recurrence. In line with the histological and radiological malignization, Ki67 expression increased from initial to final resections in 14/17 cases. Conclusion: The non-random distribution of glioma malignization in patients and unidirectional increase of anatomical tumor networks after surgical procedures provides evidence that surgical lesions, in the presence of residual tumor cells, can stimulate local tumor progression and tumor cell network formation. This argues for the development of intraoperative treatments increasing the benefits from surgical resection by specifically disrupting the mechanisms of local recurrence, particularly tumor cell network functionality. C1 [Brenner, Steffen] University of Heidelberg, University Hospital Mannheim, Department of NeurosurgeryMannheim, Germany. [Hartzendorf, Sebastian] University of Heidelberg, University Hospital Mannheim, Department of NeurosurgeryMannheim, Germany. [Vogt, Philip] University of Heidelberg, University Hospital Mannheim, Department of NeurosurgeryMannheim, Germany. [Maier, Elena] University of Heidelberg, University Hospital Mannheim, Department of NeurosurgeryMannheim, Germany. [Etminan, Nima] University of Heidelberg, University Hospital Mannheim, Department of NeurosurgeryMannheim, Germany. [Jung, Erik] University Hospital Heidelberg, Neurology Clinic and National Center for Tumor DiseasesHeidelberg, Germany. [Wick, Wolfgang] University Hospital Heidelberg, Neurology Clinic and National Center for Tumor DiseasesHeidelberg, Germany. [Sahm, Felix] University of Heidelberg, University Hospital Heidelberg, Department of NeuropathologyHeidelberg, Germany. [Winkler, Frank] University Hospital Heidelberg, Neurology Clinic and National Center for Tumor DiseasesHeidelberg, Germany. [Ratliff, Miriam] University of Heidelberg, University Hospital Mannheim, Department of NeurosurgeryMannheim, Germany. RP Ratliff, M (reprint author), University of Heidelberg, University Hospital Mannheim, Department of Neurosurgery, Mannheim, Germany. EM miriam.ratliff@umm.de CR Brown TJ, Brennan MC, Li M, Church EW, Brandmeir NJ, Rakszawski KL, et al. Association of the Extent of Resection with Survival in Glioblastoma: A Systematic Review and Meta-Analysis. JAMA Oncol, 2016, 2:1460–9., DOI 10. 1001/jamaoncol.2016.1373 Aghi MK, Nahed BV, Sloan AE, Ryken TC, Kalkanis SN, Olson JJ. The Role of Surgery in the Management of Patients with Diffuse Low Grade Glioma. J Neurooncol, 2015, 125:503–30., DOI 10.1007/s11060-015- 1867-1 Lacroix M, Abi-Said D, Fourney DR, Gokaslan ZL, Shi W, DeMonte F, et al. A Multivariate Analysis of 416 Patients with Glioblastoma Multiforme: Prognosis, Extent of Resection, and Survival. J Neurosurg, 2001, 95:190–8., DOI 10.3171/jns.2001.95.2.0190 Marko NF, Weil RJ, Schroeder JL, Lang FF, Suki D, Sawaya RE. Extent of Resection of Glioblastoma Revisited: Personalized Survival Modeling Facilitates More Accurate Survival Prediction and Supports a Maximum- Safe-Resection Approach to Surgery. J Clin Oncol, 2014, 32:774–82., DOI 10. 1200/jco.2013.51.8886 Molinaro AM, Hervey-Jumper S, Morshed RA, Young J, Han SJ, Chunduru P, et al. Association of Maximal Extent of Resection of Contrast-Enhanced and Non-contrast-enhanced Tumor with Survival within Molecular Subgroups of Patients with Newly Diagnosed Glioblastoma. JAMA Oncol, 2020, 6:495., DOI 10.1001/jamaoncol.2019.6143 Jakola AS, Myrmel KS, Kloster R, Torp SH, Lindal S, Unsgard G, et al. Comparison of a Strategy Favoring Early Surgical Resection vs a Strategy Favoring Watchful Waiting in Low-Grade Gliomas. JAMA, 2012, 308:1881–8., DOI 10.1001/jama.2012.12807 Xu DS, Awad A-W, Mehalechko C, Wilson JR, Ashby LS, Coons SW, et al. An Extent of Resection Threshold for Seizure freedom in Patients with Low-Grade Gliomas. J Neurosurg, 2018, 128:1084–90., DOI 10.3171/2016.12.jns161682 Kilic T, Ozduman K, Elmaci I, Sav A, Necmettin PamirM. Effect of Surgery on Tumor Progression and Malignant Degeneration in Hemispheric Diffuse Low- Grade Astrocytomas. J Clin Neurosci, 2002, 9:549–52., DOI 10.1054/jocn.2002. 1136 Rossi M, Gay L, Ambrogi F, Nibali MC, Sciortino T, Puglisi G, et al. Association of Supratotal Resection with Progression-free Survival, Malignant Transformation, and Overall Survival in Lower-Grade Gliomas. Neuro Oncol, 2020, 23:812., DOI 10.1093/neuonc/noaa225 Smith JS, Chang EF, Lamborn KR, Chang SM, Prados MD, Cha S, et al. Role of Extent of Resection in the Long-Term Outcome of Low-Grade Hemispheric Gliomas. J Clin Oncol, 2008, 26:1338–45., DOI 10.1200/jco.2007.13.9337 Petrecca K, Guiot M-C, Panet-Raymond V, Souhami L. Failure Pattern Following Complete Resection Plus Radiotherapy and Temozolomide Is at the Resection Margin in Patients with Glioblastoma. J Neurooncol, 2013, 111: 19–23., DOI 10.1007/s11060-012-0983-4 Okolie O, Bago JR, Schmid RS, Irvin DM, Bash RE, Miller CR, et al. Reactive Astrocytes Potentiate Tumor Aggressiveness in a Murine Glioma Resection and Recurrence Model. Neuonc, 2016, 18:1622–33., DOI 10.1093/neuonc/ now117 Weil S, Osswald M, Solecki G, Grosch J, Jung E, Lemke D, et al. Tumor Microtubes Convey Resistance to Surgical Lesions and Chemotherapy in Gliomas. Neuro Oncol, 2017, 19:1316–26., DOI 10.1093/neuonc/nox070 Osswald M, Jung E, Sahm F, Solecki G, Venkataramani V, Blaes J, et al. Brain Tumour Cells Interconnect to a Functional and Resistant Network. Nature, 2015, 528:93–8., DOI 10.1038/nature16071 Jung E, Osswald M, Blaes J, Wiestler B, Sahm F, Schmenger T, et al. Tweety- Homolog 1 Drives Brain Colonization of Gliomas. J Neurosci, 2017, 37: 6837–50., DOI 10.1523/jneurosci.3532-16.2017 Venkataramani V, Tanev DI, Strahle C, Studier-Fischer A, Fankhauser L, Kessler T, et al. Glutamatergic Synaptic Input to Glioma Cells Drives Brain Tumour Progression. Nature, 2019, 573:532–8., DOI 10.1038/s41586-019- 1564-x Weller M, van den Bent M, Tonn JC, Stupp R, Preusser M, Cohen-Jonathan- Moyal E, et al. European Association for Neuro-Oncology, EANO, Guideline on the Diagnosis and Treatment of Adult Astrocytic and Oligodendroglial Gliomas. Lancet Oncol, 2017, 18:e315–e329., DOI 10.1016/s1470-2045(17, 30194-8 Wen PY, Weller M, Lee EQ, Alexander BM, Barnholtz-Sloan JS, Barthel FP, et al. Glioblastoma in Adults: A Society for Neuro-Oncology, SNO, and European Society of Neuro-Oncology, EANO, Consensus Review on Current Management and Future Directions. Neuro Oncol, 2020, 22:1073–113., DOI 10. 1093/neuonc/noaa106 Ratel D, van der Sanden B, Wion D. Glioma Resection and Tumor Recurrence: Back to Semmelweis. Neuonc, 2016, 18:1688–9., DOI 10.1093/neuonc/now201 Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A Summary. Acta Neuropathol, 2016, 131:803–20., DOI 10.1007/s00401-016-1545-1 Wiestler B, Capper D, Holland-Letz T, Korshunov A, von Deimling A, Pfister SM, et al. ATRX Loss Refines the Classification of Anaplastic Gliomas and Identifies a Subgroup of IDH Mutant Astrocytic Tumors with Better Prognosis. Acta Neuropathol, 2013, 126:443–51., DOI 10.1007/s00401-013- 1156-z Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, et al. Fiji: An Open-Source Platform for Biological-Image Analysis. Nat Methods, 2012, 9:676–82., DOI 10.1038/nmeth.2019 Gritsenko PG, Atlasy N, Dieteren CEJ, Navis AC, Venhuizen J-H, Veelken C, et al. p120-catenin-dependent Collective Brain Infiltration by Glioma Cell Networks. Nat Cel Biol, 2020, 22:97–107., DOI 10.1038/s41556-019- 0443-x Sahm F, Capper D, Jeibmann A, Habel A, Paulus W, Troost D, et al. Addressing Diffuse Glioma as a Systemic Brain Disease with Single-Cell Analysis. Arch Neurol, 2012, 69:523–6., DOI 10.1001/archneurol.2011. 2910 Konishi Y, Muragaki Y, Iseki H, Mitsuhashi N, Okada Y. Patterns of Intracranial Glioblastoma Recurrence after Aggressive Surgical Resection and Adjuvant Management: Retrospective Analysis of 43 Cases. Neurol Med Chir, Tokyo,, 2012, 52:577–86., DOI 10.2176/nmc.52.577 Lemee J-M, Clavreul A,Menei P. Intratumoral Heterogeneity in Glioblastoma: Don’t Forget the Peritumoral Brain Zone. Neuro Oncol, 2015, 17:1322–32., DOI 10.1093/neuonc/nov119 StummerW, Pichlmeier U, Meinel T, Wiestler OD, Zanella F, Reulen H-J, et al. Fluorescence-guided Surgery with 5-aminolevulinic Acid for Resection of Malignant Glioma: A Randomised Controlled Multicentre Phase III Trial. Lancet Oncol, 2006, 7:392–401., DOI 10.1016/s1470-2045(06)70665-9 Campos B, Olsen LR, Urup T, Poulsen HS. A Comprehensive Profile of Recurrent Glioblastoma. Oncogene, 2016, 35:5819–25., DOI 10.1038/onc. 2016.85 Combs SE, Schmid TE, Vaupel P, Multhoff G. Stress Response Leading to Resistance in Glioblastoma-The Need for Innovative Radiotherapy, iRT, Concepts. Cancers, Basel,, 2016, 8:15., DOI 10.3390/cancers8010015 Cordner R, Black KL, Wheeler CJ. Exploitation of Adaptive Evolution in Glioma Treatment. CNS Oncol, 2013, 2:171–9., DOI 10.2217/cns.12.46 Venkatesh HS, Morishita W, Geraghty AC, Silverbush D, Gillespie SM, Arzt M, et al. Electrical and Synaptic Integration of Glioma into Neural Circuits. Nature, 2019, 573:539–45., DOI 10.1038/s41586-019-1563-y NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610268 EP 1610276 DI 10.3389/pore.2022.1610268 PG 9 ER PT J AU Fan, X Liu, Y Liang, Z Wang, Sh Yang, J Wu, A AF Fan, Xin Liu, Yanqing Liang, Zhigang Wang, Shanshan Yang, Jing Wu, Aihua TI Diagnostic Value of Six Tumor Markers for Malignant Pleural Effusion in 1,230 Patients: A Single-Center Retrospective Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE diagnostic performance; malignant pleural effusion; tumor markers; carcinoembryonic antigen; cytokeratin 19 fragment; area under the curve ID diagnostic performance; malignant pleural effusion; tumor markers; carcinoembryonic antigen; cytokeratin 19 fragment; area under the curve AB Background: The diagnostic value of tumor markers in pleural effusion (PE) and serum for malignant pleural effusion (MPE) is still in debate. This study aimed to evaluate the diagnostic value of six tumor markers in PE, serum, and the corresponding PE/serum (PE/S) ratio in distinguishing MPE from benign pleural effusion (BPE). Methods: A total of 1,230 patients with PE (452 MPEs and 778 BPEs) were retrospectively included in the study. PE and serum levels of carcinoembryonic antigen (CEA), carbohydrate antigen 15-3 (CA15-3), carbohydrate antigen 125 (CA125), carbohydrate antigen 19-9 (CA19-9), cytokeratin 19 fragment (CYFRA 21-1), and neuron-specific enolase (NSE) were measured. The area under the curve (AUC) was used to assess the single and combined diagnostic values of the six tumor markers for MPE. Results: The levels of the six tumor markers in PE, serum, and PE/S were significantly higher in MPE than that in BPE, except for serum CA125. PE CEA showed the highest AUC [0.890 (0.871–0.907)] at a cut-off value of 3.7 ng/ml compared to any single tumor marker using receiver operating characteristic (ROC) analysis. The specificity, sensitivity, positive predictive value (PPV), negative predictive value (NPV), positive likelihood ratio (PLR), and negative likelihood ratio (NLR) of PE CEA were 74.1%, 95.5%, 90.5%, 86.4%, 16.47, and 0.27, respectively. The combination of PE CEA and serum CYFRA21-1 showed the best diagnostic performance with an AUC of 0.934 (sensitivity, 79.9%; specificity, 95.7%, PPV, 90.5; PLR, 17.35) among all two or three combinations. Besides, serum CYFRA21-1 was the best diagnostic tumormarker in distinguishing cytology-negativeMPE from BPE at a cut-off value of 3.0 ng/ml. Conclusion: PE CEA was the best diagnostic tumor marker in distinguishing MPE from BPE. Serum CYFRA21-1 was the best diagnostic tumor marker in distinguishing cytology-negative MPE from BPE. The combination of PE CEA and serum CYFRA21-1 could increase the diagnostic performance in distinguishingMPE from BPE and cytology-negativeMPE fromBPE. C1 [Fan, Xin] Ningbo First Hospital, Department of DermatologyNingbo, China. [Liu, Yanqing] Ningbo First Hospital, Department of Laboratory MedicineNingbo, China. [Liang, Zhigang] Ningbo First Hospital, Department of Thoracic SurgeryNingbo, China. [Wang, Shanshan] Ningbo First Hospital, Department of Laboratory MedicineNingbo, China. [Yang, Jing] Ningbo First Hospital, Department of Respiratory and Critical CareNingbo, China. [Wu, Aihua] Ningbo First Hospital, Department of Laboratory MedicineNingbo, China. RP Wu, A (reprint author), Ningbo First Hospital, Department of Laboratory Medicine, Ningbo, China. EM wuah910602@126.com CR Feller-Kopman DJ, Reddy CB, DeCamp MM, Diekemper RL, Gould MK, Henry T, et al. Management of Malignant Pleural Effusions. An Official ATS/ STS/STR Clinical Practice Guideline. Am J Respir Crit Care Med, 2018, 198(7): 839–49., DOI 10.1164/rccm.201807-1415ST Wang S, Tian S, Li Y, Zhan N, Guo Y, Liu Y, et al. Development and Validation of a Novel Scoring System Developed from a Nomogram to Identify Malignant Pleural Effusion. EBioMedicine, 2020, 58:102924., DOI 10.1016/j.ebiom.2020. 102924 ShafiqM, Feller-Kopman D. Management of Malignant Pleural Effusions. Clin Chest Med, 2020, 41(2):259–67., DOI 10.1016/j.ccm.2020.02.009 Porcel JM, Esquerda A, Vives M, Bielsa S. Etiology of Pleural Effusions: Analysis of More Than 3,000 Consecutive Thoracenteses. Archivos de Bronconeumologia, English Edition,, 2014, 50(5):161–5., DOI 10.1016/j. arbres.2013.11.007 Arnold DT, De Fonseka D, Perry S, Morley A, Harvey JE, Medford A, et al. Investigating Unilateral Pleural Effusions: the Role of Cytology. Eur Respir J, 2018, 52(5):1801254., DOI 10.1183/13993003.01254-2018 Maturu VN, Dhooria S, Bal A, Singh N, Aggarwal AN, Gupta D, et al. Role of Medical Thoracoscopy and Closed-Blind Pleural Biopsy in Undiagnosed Exudative Pleural Effusions. J Bronchology Interv Pulmonol, 2015, 22(2): 121–9., DOI 10.1097/lbr.0000000000000145 Antonangelo L, Sales RK, Cora AP, Acencio MMP, Teixeira LR, Vargas FS. Pleural Fluid Tumour Markers in Malignant Pleural Effusion with Inconclusive Cytologic Results. Curr Oncol, 2015, 22(5):336–41., DOI 10. 3747/co.22.2563 Nguyen AH, Miller EJ,Wichman CS, Berim IG, Agrawal DK. Diagnostic Value of Tumor Antigens in Malignant Pleural Effusion: a Meta-Analysis. Translational Res, 2015, 166(5):432–9., DOI 10.1016/j.trsl.2015.04.006 Yang Y, Liu Y-L, Shi H-Z. Diagnostic Accuracy of Combinations of Tumor Markers for Malignant Pleural Effusion: An Updated Meta-Analysis. Respiration, 2017, 94(1):62–9., DOI 10.1159/000468545 Huang W-W, Tsao S-M, Lai C-L, Su C-C, Tseng C-E. Diagnostic Value of Her- 2/neu, Cyfra 21-1, and Carcinoembryonic Antigen Levels in Malignant Pleural Effusions of Lung Adenocarcinoma. Pathology, 2010, 42(3):224–8., DOI 10. 3109/00313021003631320 Son S-M, Han H-S, An JY, Choe KH, Lee KM, Lee KH, et al. Diagnostic Performance of CD66c in Lung Adenocarcinoma-Associated Malignant Pleural Effusion: Comparison with CEA, CA 19-9, and CYFRA 21-1. Pathology, 2015, 47(2):123–9., DOI 10.1097/pat.0000000000000215 Feng M, Zhu J, Liang L, Zeng N, Wu Y, Wan C, et al. Diagnostic Value of Tumor Markers for Lung Adenocarcinoma-Associated Malignant Pleural Effusion: a Validation Study and Meta-Analysis. Int J Clin Oncol, 2017, 22(2):283–90., DOI 10.1007/s10147-016-1073-y Zhang H, Li C, Hu F, Zhang X, Shen Y, Chen Y, et al. Auxiliary Diagnostic Value of Tumor Biomarkers in Pleural Fluid for Lung Cancer-Associated Malignant Pleural Effusion. Respir Res, 2020, 21(1):284., DOI 10.1186/s12931- 020-01557-z PaganuzziM, OnettoM,Marroni P, Filiberti R, Tassara E, Parodi S, et al. Diagnostic Value of CYFRA 21-1 Tumor Marker and CEA in Pleural Effusion Due to Mesothelioma. Chest, 2001, 119(4):1138–42., DOI 10.1378/chest.119.4.1138 Villena V, Lopez-Encuentra A, Echave-Sustaeta J, Marti´n-Escribano P, Ortuno-de-Solo B, Estenoz-Alfaro J. Diagnostic Value of CA 549 in Pleural Fluid. Comparison with CEA, CA 15.3 and CA 72.4. Lung Cancer, 2003, 40(3): 289–94., DOI 10.1016/S0169-5002(03)00120-X Hackbarth JS, Murata K, Reilly WM, Algeciras-Schimnich A. Performance of CEA and CA19-9 in Identifying Pleural Effusions Caused by Specific Malignancies. Clin Biochem, 2010, 43(13-14):1051–5., DOI 10.1016/j. clinbiochem.2010.05.016 Filiberti R, Parodi S, Libener R, Ivaldi GP, Canessa PA, Ugolini D, et al. Diagnostic Value ofMesothelin in Pleural Fluids: Comparison with CYFRA 21-1 and CEA. Med Oncol, 2013, 30(2):543., DOI 10.1007/s12032-013- 0543-6 Porcel JM, Vives M, Esquerda A, Salud A, Perez B, Rodriguez-Panadero F. Use of a Panel of Tumor Markers, Carcinoembryonic Antigen, Cancer Antigen 125, Carbohydrate Antigen 15-3, and Cytokeratin 19 Fragments, in Pleural Fluid for the Differential Diagnosis of Benign and Malignant Effusions. Chest, 2004, 126(6):1757–63., DOI 10.1378/chest.126.6.1757 Shitrit D, Zingerman B, Shitrit AB-G, Shlomi D, Kramer MR. Diagnostic Value of CYFRA 21-1, CEA, CA 19-9, CA 15-3, and CA 125 Assays in Pleural Effusions: Analysis of 116 Cases and Review of the Literature. Oncologist, 2005, 10(7):501–7., DOI 10.1634/theoncologist.10-7-501 Gaspar MJ, De Miguel J, Garcia Diaz JD, Diez M. Clinical Utility of a Combination of Tumour Markers in the Diagnosis of Malignant Pleural Effusions. Anticancer Res, 2008, 28(5b):2947–52. Zhai K, Wang W, Wang Y, Liu J-Y, Zhou Q, Shi H-Z. Diagnostic Accuracy of Tumor Markers for Malignant Pleural Effusion: a Derivation and Validation Study. J Thorac Dis, 2017, 9(12):5220–9., DOI 10.21037/jtd.2017.11.62 Volaric D, Flego V, Zauhar G, Bulat-Kardum L. Diagnostic Value of Tumour Markers in Pleural Effusions. Biochem Med, Zagreb,, 2018, 28(1):010706., DOI 10.11613/bm.2018.010706 Grunnet M, Sorensen JB. Carcinoembryonic Antigen, CEA, as TumorMarker in Lung Cancer. Lung Cancer, 2012, 76(2):138–43., DOI 10.1016/j.lungcan. 2011.11.012 Tozzoli R, Basso SMM, D’Aurizio F, Metus P, Lumachi F. Evaluation of Predictive Value of Pleural CEA in Patients with Pleural Effusions and Histological Findings: A Prospective Study and Literature Review. Clin Biochem, 2016, 49(16-17):1227–31., DOI 10.1016/j.clinbiochem.2016.08.006 Rodriguez-Panadero F, Borderas Naranjo F, Lopez Mejias J. Pleural Metastatic Tumours and Effusions. Frequency and Pathogenic Mechanisms in a postmortem Series. Eur Respir J, 1989, 2(4):366–9. Chen Z,Wang Y, Fang M. Analysis of Tumor Markers in Pleural Effusion and Serum to Verify the Correlations between Serum Tumor Markers and Tumor Size, TNM Stage of Lung Adenocarcinoma. Cancer Med, 2020, 9(4):1392–9., DOI 10.1002/cam4.2809 Psallidas I, Kalomenidis I, Porcel JM, Robinson BW, Stathopoulos GT. Malignant Pleural Effusion: from Bench to Bedside. Eur Respir Rev, 2016, 25(140):189–98., DOI 10.1183/16000617.0019-2016 Hsieh T-C, Huang W-W, Lai C-L, Tsao S-M, Su C-C. Diagnostic Value of Tumor Markers in Lung Adenocarcinoma-Associated Cytologically Negative Pleural Effusions. Cancer Cytopathology, 2013, 121(9):483–8., DOI 10.1002/ cncy.21283 Wang YX, Hu D, Yan X. Diagnostic Accuracy of Cyfra 21-1 for Head and Neck Squamous Cell Carcinoma: a Meta-Analysis. Eur Rev Med Pharmacol Sci, 2013, 17(17):2383–9. Hooper C, Lee YCG, Maskell N. Investigation of a Unilateral Pleural Effusion in Adults: British Thoracic Society Pleural Disease Guideline 2010. Thorax, 2010, 65(Suppl. 2):ii4–ii17., DOI 10.1136/thx.2010.136978 Wu A, Liang Z, Yuan S, Wang S, Peng W, Mo Y, et al. Development and Validation of a Scoring System for Early Diagnosis of Malignant Pleural Effusion Based on a Nomogram. Front Oncol, 2021, 11:775079., DOI 10.3389/ fonc.2021.775079 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610280 EP 1610287 DI 10.3389/pore.2022.1610280 PG 8 ER PT J AU Yang, Sh Xie, Sh Shi, X Su, D He, B Xu, Y Liu, Z AF Yang, Shuming Xie, Shengzhi Shi, Xinying Su, Dan He, Bo Xu, Yang Liu, Zhefeng TI Characterizing HDAC Pathway Copy Number Variation in Pan-Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; copy number variation; immune; histone deacetylase; tumor characteristics ID prognosis; copy number variation; immune; histone deacetylase; tumor characteristics AB Background: Histone deacetylase (HDAC) plays a crucial role in regulating the expression and activity of a variety of genes associated with tumor progression and immunotherapeutic processes. The aim of this study was to characterize HDAC pathway copy number variation (CNV) in pan-cancer. Methods: A total of 10,678 tumor samples involving 33 types of tumors from The Cancer Genome Atlas (TCGA) were included in the study. Results: HDAC pathway CNV and CNV gain were identified as prognostic risk factors for pan-cancer species. The differences of tumor characteristics including tumor mutational burden, tumor neoantigen burden, high-microsatellite instability, and microsatellite stable between HDAC pathway CNV altered-type group and wild-type group varied among the various cancer species. In some cancer types, HDAC pathway CNV alteration was positively correlated with loss of heterozygosity, CNV burden, ploidy, and homologous recombination defect score markers, while it was significantly negatively correlated with immune score and stroma score. There were significant differences in immune characteristics such as major histocompatibility complex class I (MHC-I), MHC-II, chemokines, cytolytic-activity, and IFN-γ between the two groups. Immune cycle characteristics varied from one cancer type to another. Conclusion: This study reveals a tumor and immune profile of HDAC pathway CNV as well as its unlimited potential in immune prognosis. C1 [Yang, Shuming] The Fifth Medical Center of PLA General Hospital, Senior Department of Oncology, Department of OncologyBeijing, China. [Xie, Shengzhi] The Fifth Medical Center of PLA General Hospital, Senior Department of Oncology, Department of OncologyBeijing, China. [Shi, Xinying] Genecast Biotechnology Co., Ltd.Wuxi, China. [Su, Dan] The Fifth Medical Center of PLA General Hospital, Senior Department of Oncology, Department of OncologyBeijing, China. [He, Bo] The Fifth Medical Center of PLA General Hospital, Senior Department of Oncology, Department of OncologyBeijing, China. [Xu, Yang] The Fifth Medical Center of PLA General Hospital, Senior Department of Oncology, Department of OncologyBeijing, China. [Liu, Zhefeng] The Fifth Medical Center of PLA General Hospital, Senior Department of Oncology, Department of OncologyBeijing, China. RP Liu, Z (reprint author), The Fifth Medical Center of PLA General Hospital, Senior Department of Oncology, Department of Oncology, Beijing, China. EM lzf1220@sina.com CR Sun Y, Sun Y, Yue S, Wang Y, Lu F. Histone Deacetylase Inhibitors in Cancer Therapy. Curr Top Med Chem, 2018, 18(28):2420–8., DOI 10.2174/ 1568026619666181210152115 Seto E, Yoshida M. Erasers of Histone Acetylation: The Histone Deacetylase Enzymes. Cold Spring Harbor Perspect Biol, 2014, 6(4):a018713., DOI 10.1101/ cshperspect.a018713 Teodori L, Sestili P, Madiai V, Coppari S, Fraternale D, Rocchi MBL, et al. MicroRNAs Bioinformatics Analyses Identifying HDAC Pathway as a Putative Target for Existing Anti-COVID-19 Therapeutics. Front Pharmacol, 2020, 11: 582003., DOI 10.3389/fphar.2020.582003 Zhao L-M, Zhang J-H. Histone Deacetylase Inhibitors in Tumor Immunotherapy. Curr Med Chem, 2019, 26(17):2990–3008., DOI 10.2174/ 0929867324666170801102124 De Simone A, Milelli A. Histone Deacetylase Inhibitors as Multitarget Ligands: New Players in Alzheimer’s Disease Drug Discovery? ChemMedChem, 2019, 14(11):1067–73., DOI 10.1002/cmdc.201900174 Shah RR. Safety and Tolerability of Histone Deacetylase, HDAC, Inhibitors in Oncology. Drug Saf, 2019, 42(2):235–45., DOI 10.1007/s40264-018-0773-9 Knowles DA, Bouchard G, Plevritis S. Sparse Discriminative Latent Characteristics for Predicting Cancer Drug Sensitivity from Genomic Features. Plos Comput Biol, 2019, 15(5):e1006743., DOI 10.1371/journal.pcbi.1006743 Butler RM, McKenzie RC, Jones CL, Flanagan CE, Woollard WJ, Demontis M, et al. Contribution of STAT3 and RAD23B in Primary Sezary Cells to Histone Deacetylase Inhibitor FK228 Resistance. J Invest Dermatol, 2019, 139(9): 1975–84., DOI 10.1016/j.jid.2019.03.1130 Cohen AL, Piccolo SR, Cheng L, Soldi R, Han B, Johnson WE, et al. Genomic Pathway Analysis Reveals that EZH2 and HDAC4 Represent Mutually Exclusive Epigenetic Pathways across Human Cancers. BMC Med Genomics, 2013, 6:35., DOI 10.1186/1755-8794-6-35 Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The Immune Landscape of Cancer. Immunity, 2018, 48(4):812., DOI 10.1016/j. immuni.2018.03.023 Angelova M, Charoentong P, Hackl H, Fischer ML, Snajder R, Krogsdam AM, et al. Characterization of the Immunophenotypes and Antigenomes of Colorectal Cancers Reveals Distinct Tumor Escape Mechanisms and Novel Targets for Immunotherapy. Genome Biol, 2015, 16:64., DOI 10.1186/s13059- 015-0620-6 Charoentong P, Finotello F, Angelova M, Mayer C, Efremova M, Rieder D, et al. Pan-cancer Immunogenomic Analyses Reveal Genotype- Immunophenotype Relationships and Predictors of Response to Checkpoint Blockade. Cel Rep, 2017, 18(1):248–62., DOI 10.1016/j.celrep. 2016.12.019 Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and Genetic Properties of Tumors Associated with Local Immune Cytolytic Activity. Cell, 2015, 160(1-2):48–61., DOI 10.1016/j.cell.2014.12.033 Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, et al. IFN-γ-related mRNA Profile Predicts Clinical Response to PD-1 Blockade. J Clin Invest, 2017, 127(8):2930–40., DOI 10.1172/JCI91190 Karasaki T, Nagayama K, Kuwano H, Nitadori J-i., Sato M, Anraku M, et al. An Immunogram for the Cancer-Immunity Cycle: Towards Personalized Immunotherapy of Lung Cancer. J Thorac Oncol, 2017, 12(5):791–803., DOI 10.1016/j.jtho.2017.01.005 Montgomery MR, Hull EE. Alterations in the Glycome after HDAC Inhibition Impact Oncogenic Potential in Epigenetically Plastic SW13 Cells. BMC Cancer, 2019, 19(1):79., DOI 10.1186/s12885-018-5129-4 Delaney JR, Stupack DG. Whole Genome Pathway Analysis Identifies an Association of Cadmium Response Gene Loss with Copy Number Variation in Mutant P53 Bearing Uterine Endometrial Carcinomas. PLoS One, 2016, 11(7): e0159114., DOI 10.1371/journal.pone.0159114 Garmpis N, Damaskos C, Garmpi A, Dimitroulis D, Spartalis E, Margonis GA, et al. Targeting Histone Deacetylases in Malignant Melanoma: A Future Therapeutic Agent or Just Great Expectations? Anticancer Res, 2017, 37(10):5355–62., DOI 10.21873/anticanres.11961 Xiao J, Li W, Huang Y, Huang M, Li S, Zhai X, et al. A Next-Generation Sequencing-Based Strategy Combining Microsatellite Instability and Tumor Mutation burden for Comprehensive Molecular Diagnosis of Advanced Colorectal Cancer. BMC Cancer, 2021, 21(1):282., DOI 10.1186/s12885-021- 07942-1 Liu L, Bai X,Wang J, Tang X-R, Wu D-H, Du S-S, et al. Combination of TMB and CNA Stratifies Prognostic and Predictive Responses to Immunotherapy across Metastatic Cancer. Clin Cancer Res, 2019, 25(24):7413–23., DOI 10.1158/ 1078-0432.CCR-19-0558 Lu M, Zhang P, Zhang Y, Li Z, Gong J-f., Li J, et al. Efficacy, Safety and Biomarkers of Toripalimab in Patients with Recurrent or Metastatic Neuroendocrine Neoplasms: A Multiple-center Phase Ib Trial. Clin Cancer Res, 2020, 26(10):2337–45., DOI 10.1158/1078-0432.CCR-19-4000 Hanigan CL, Van Engeland M, De Bruine AP, Wouters KA, Weijenberg MP, Eshleman JR, et al. An Inactivating Mutation in HDAC2 Leads to Dysregulation of Apoptosis Mediated by APAF1. Gastroenterology, 2008, 135(5):1654–64., DOI 10.1053/j.gastro.2008.07.078 Sefrioui D, Vermeulin T, Blanchard F, Chapusot C, Beaussire L, Armengol- Debeir L, et al. Copy Number Variations in DCC/18q and ERBB2/17q are Associated with Disease-free Survival in Microsatellite Stable colon Cancer. Int J Cancer, 2017, 140(7):1653–61., DOI 10.1002/ijc.30584 Chen Y, Chen C. DNA Copy Number Variation and Loss of Heterozygosity in Relation to Recurrence of and Survival from Head and Neck Squamous Cell Carcinoma: A Review. Head Neck, 2008, 30(10):1361–83., DOI 10.1002/hed. 20861 Kong L, Liu P, Zheng M, Xue B, Liang K, Tan X. Multi-omics Analysis Based on Integrated Genomics, Epigenomics and Transcriptomics in Pancreatic Cancer. Epigenomics, 2020, 12(6):507–24., DOI 10.2217/epi-2019-0374 Shen S, Li H, Liu J, Sun L, Yuan Y. The panoramic Picture of Pepsinogen Gene Family with pan-cancer. Cancer Med, 2020, 9(23):9064–80., DOI 10.1002/cam4. 3489 Hsu C-C, Chang W-C, Hsu T-I, Liu J-J, Yeh S-H, Wang J-Y, et al. Suberoylanilide Hydroxamic Acid Represses Glioma Stem-like Cells. J Biomed Sci, 2016, 23(1):81., DOI 10.1186/s12929-016-0296-6 Nakajima NI, Niimi A, Isono M, Oike T, Sato H, Nakano T, et al. Inhibition of the HDAC/Suv39/G9a Pathway Restores the Expression of DNA Damagedependent Major Histocompatibility Complex Class I-Related Chain A and B in Cancer Cells. Oncol Rep, 2017, 38(2):693–702., DOI 10.3892/or.2017.5773 Neuwelt AJ, Kimball AK, Johnson AM, Arnold BW, Bullock BL, Kaspar RE, et al. Cancer Cell-Intrinsic Expression of MHC II in Lung Cancer Cell Lines is Actively Restricted by MEK/ERK Signaling and Epigenetic Mechanisms. J Immunother Cancer, 2020, 8(1):e000441., DOI 10.1136/jitc-2019-000441 Palomino DCT, Marti LC. Chemokines and Immunity. Einstein, Sao Paulo,, 2015, 13(3):469–73., DOI 10.1590/S1679-45082015RB3438 Zheng H, Zhao W, Yan C, Watson CC, Massengill M, Xie M, et al. HDAC Inhibitors Enhance T-Cell Chemokine Expression and Augment Response to PD-1 Immunotherapy in Lung Adenocarcinoma. Clin Cancer Res, 2016, 22(16):4119–32., DOI 10.1158/1078-0432.CCR-15-2584 Bretz AC, Parnitzke U, Kronthaler K, Dreker T, Bartz R, Hermann F, et al. Domatinostat Favors the Immunotherapy Response by Modulating the Tumor Immune Microenvironment, TIME). J Immunotherapy Cancer, 2019, 7(1): 294., DOI 10.1186/s40425-019-0745-3 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610288 EP 1610297 DI 10.3389/pore.2022.1610288 PG 10 ER PT J AU Liu, P Jiang, Y Zheng, X Pan, B Xiang, H Zheng, M AF Liu, Pingping Jiang, Yinan Zheng, Xiaojing Pan, Baoyue Xiang, Huiling Zheng, Min TI Pretreatment Systemic Immune-Inflammation Index Can Predict Response to Neoadjuvant Chemotherapy in Cervical Cancer at Stages IB2-IIB SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cervical cancer; prognosis; neoadjuvant chemotherapy; pathological complete response; systemic immune-inflammation index ID cervical cancer; prognosis; neoadjuvant chemotherapy; pathological complete response; systemic immune-inflammation index AB Background: The systemic immune-inflammation index (SII) has been identified as a predictor of chemotherapy efficacy for a variety of cancers, and we aimed to determine its ability to predict the response to chemotherapy and its long-term prognosis for patients with cervical squamous cell carcinoma (CSCC) who have underwent platinum-based neoadjuvant chemotherapy (NACT). Methods: The date from 210 patients (133 in the training cohort and 77 in the validation cohort) with CSCC who received NACT were analyzed retrospectively. The association between SII and the pathological complete response (pCR) was determined using Pearson’s chi-square test, receiver operating characteristic (ROC) curve, and Logistic regression analysis. The Kaplan-Meier method and Cox proportional regression model were used to assess the relationship between SII and progression-free survival (PFS) or overall survival (OS). Results: The calculated optimal SII cutoff values for pCR and survival were 568.7051 and 600.5683, respectively, and patients were divided into two groups: a low SII group (≤568.7051 or ≤600.5683) and a high SII group (>568.7051 or >600.5683). A high SII was associated significantly with a lower pCR. Further analysis determined that SII was a more efficient predictor of pCR than the prognostic nutritional index, platelet-to-lymphocyte ratio, and lymphocyte-to-monocyte ratio. Upon multivariate logistic analysis, SII proved to be an independent risk factor to predict the pCR of patients with CSCC. Kaplan-Meier analysis demonstrated that PFS and OS rates were significantly higher in the low-SII group compared with those in the high-SII group. Additional multivariate analysis indicated that the SII is an independent prognostic factor for patients with CSCC treated with NACT. Conclusion: The results confirmed that the pre-treatment SII is not only an independent predictor of pCR but also an independent prognostic factor of CSCC patients treated with platinum based NACT. C1 [Liu, Pingping] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. [Jiang, Yinan] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. [Zheng, Xiaojing] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. [Pan, Baoyue] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. [Xiang, Huiling] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. [Zheng, Min] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of GynecologyGuangzhou, China. RP Zheng, M (reprint author), Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecology, Guangzhou, China. EM zhengmin@sysucc.org.cn CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer Statistics in China, 2015. CA Cancer J Clin, 2016, 66(2):115–32., DOI 10.3322/caac.21338 Licitra L, Vermorken JB Is There Still a Role for Neoadjuvant Chemotherapy in Head and Neck Cancer? Ann Oncol, 2004, 15(1):7–11., DOI 10.1093/annonc/ mdh001 Shi L, Qin X, Wang H, Xia Y, Li Y, Chen X, et al. Elevated Neutrophil-To- Lymphocyte Ratio and Monocyte-To-Lymphocyte Ratio and Decreased Platelet-To-Lymphocyte Ratio Are Associated with Poor Prognosis in Multiple Myeloma. Oncotarget, 2017, 8:18792–801., DOI 10.18632/ oncotarget.13320 Templeton AJ, Ace O, McNamara MG, Al-Mubarak M, Vera-Badillo FE, Hermanns T, et al. Prognostic Role of Platelet to Lymphocyte Ratio in Solid Tumors: a Systematic Review and Meta-Analysis. Cancer Epidemiol Biomarkers Prev, 2014, 23(7):1204–12., DOI 10.1158/1055-9965.EPI-14-0146 Mellman I, Coukos G, Dranoff G Cancer Immunotherapy Comes of Age. Nature, 2011, 480(7378):480–9., DOI 10.1038/nature10673 Hu B, Yang X-R, Xu Y, Sun Y-F, Sun C, Guo W, et al. Systemic Immune- Inflammation index Predicts Prognosis of Patients after Curative Resection for Hepatocellular Carcinoma. Clin Cancer Res, 2014, 20(23):6212–22., DOI 10. 1158/1078-0432.CCR-14-0442 Jiang C, Huang Y Predictive Effect of Systemic Immune-Inflammation Index on Pathological Complete Response of Breast Cancer to Neoadjuvant Chemotherapy and its Relation with P53. Cancer Prev Treat Res, 2020, 47(10):756–60., DOI 10.3971/j.issn.1000-8578.2020.20.0273 Eraslan E, Adas YG, Yildiz F, Gulesen AI, Karacin C, Arslan UY Systemic Immune-Inflammation Index, SII, Predicts Pathological Complete Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer. J Coll Phys Surg Pak, 2021, 31(4):399–404., DOI 10.29271/jcpsp.2021.04.399 Chen L, Yan Y, Zhu L, Cong X, Li S, Song S, et al. Systemic Immune- Inflammation index as a Useful Prognostic Indicator Predicts Survival in Patients with Advanced Gastric Cancer Treated with Neoadjuvant Chemotherapy. Cancer Manag Res, 2017, 9:849–67., DOI 10.2147/CMAR. S151026 Murthy P, Boone BA ASO Author Reflections: Systemic Immune- Inflammation Index, SII, as a Biomarker of Response to Neoadjuvant Therapy in Patients with Pancreatic Adenocarcinoma. Ann Surg Oncol, 2020, 27(3):907–8., DOI 10.1245/s10434-019-08136-7 Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, et al. Tumour-infiltrating Lymphocytes and Prognosis in Different Subtypes of Breast Cancer: a Pooled Analysis of 3771 Patients Treated with Neoadjuvant Therapy. Lancet Oncol, 2018, 19(1):40–50., DOI 10.1016/S1470- 2045(17)30904-X D’AlessandrisN, Palaia I, Pernazza A, Tomao F, Di Pinto A, Musacchio L, et al. PD-L1 Expression Is Associated with Tumor Infiltrating Lymphocytes that Predict Response to NACT in Squamous Cell Cervical Cancer. Virchows Arch, 2021, 478(3):517–25., DOI 10.1007/s00428-020-02922-5 Sheu B-C, Hsu S-M, Ho H-N, Lin R-H, Torng P-L, Huang S-C Reversed CD4/ CD8 Ratios of Tumor-Infiltrating Lymphocytes are Correlated with the Progression of Human Cervical Carcinoma. Cancer, 1999, 86(8):1537–43., DOI 10.1002/(sici)1097-0142(19991015)86:8<1537:aid-cncr21>3.0.co;2-d Ray-Coquard I, Cropet C, Van Glabbeke M, Sebban C, Le Cesne A, Judson I, et al. Lymphopenia as a Prognostic Factor for Overall Survival in Advanced Carcinomas, Sarcomas, and Lymphomas. Cancer Res, 2009, 69(13):5383–91., DOI 10.1158/0008-5472.CAN-08-3845 Cools-Lartigue J, Spicer J, McDonald B, Gowing S, Chow S, Giannias B, et al. Neutrophil Extracellular Traps Sequester Circulating Tumor Cells and Promote Metastasis. J Clin Invest, 2013, 123:3446–58., DOI 10.1172/JCI67484 Martinez PJ, Farhan A, Mustafa M, Javaid N, Darkoh C, Garrido-Sanabria E, et al. PspA Facilitates Evasion of Pneumococci from Bactericidal Activity of Neutrophil Extracellular Traps, NETs). Microb Pathogenesis, 2019, 136: 103653., DOI 10.1016/j.micpath.2019.103653 Groth C, Hu X, Weber R, Fleming V, Altevogt P, Utikal J, et al. Immunosuppression Mediated by Myeloid-Derived Suppressor Cells, MDSCs, during Tumour Progression. Br J Cancer, 2019, 120(1):16–25., DOI 10.1038/s41416-018-0333-1 Jaillon S, Galdiero MR, Del Prete D, Cassatella MA, Garlanda C, Mantovani A Neutrophils in Innate and Adaptive Immunity. Semin Immunopathol, 2013, 35(4):377–94., DOI 10.1007/s00281-013-0374-8 Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, et al. The Prognostic Landscape of Genes and Infiltrating Immune Cells across Human Cancers. Nat Med, 2015, 21(8):938–45., DOI 10.1038/nm.3909 Murakami Y, Saito H, Shimizu S, Kono Y, Shishido Y, Miyatani K, et al. Neutrophil-to-Lymphocyte Ratio as a Prognostic Indicator in Patients with Unresectable Gastric Cancer. Anticancer Res, 2019, 39(5):2583–9., DOI 10. 21873/anticanres.13381 Wisdom AJ, Hong CS, Lin AJ, Xiang Y, Cooper DE, Zhang J, et al. Neutrophils Promote Tumor Resistance to Radiation Therapy. Proc Natl Acad Sci U.S.A, 2019, 116(37):18584–9., DOI 10.1073/pnas.1901562116 Matsumoto Y, Mabuchi S, Kozasa K, Kuroda H, Sasano T, Yokoi E, et al. The Significance of Tumor-Associated Neutrophil Density in Uterine Cervical Cancer Treated with Definitive Radiotherapy. Gynecol Oncol, 2017, 145(3): 469–75., DOI 10.1016/j.ygyno.2017.02.009 Franco AT, Corken A, Ware J Platelets at the Interface of Thrombosis, Inflammation, and Cancer. Blood, 2015, 126(5):582–8., DOI 10.1182/blood- 2014-08-531582 Lou XL, Sun J, Gong SQ, Yu XF, Gong R, Deng H Interaction between Circulating Cancer Cells and Platelets: Clinical Implication. Chin J Cancer Res, 2015, 27(5):450–60., DOI 10.3978/j.issn.1000-9604. 2015.04.10 Dong M, Shi Y, Yang J, Zhou Q, Lian Y, Wang D, et al. Prognostic and Clinicopathological Significance of Systemic Immune-Inflammation index in Colorectal Cancer: a Meta-Analysis. Ther Adv Med Oncol, 2020, 12: 175883592093742., DOI 10.1177/1758835920937425 Jomrich G, Paireder M, Kristo I, Baierl A, Ilhan-Mutlu A, Preusser M, et al. High Systemic Immune-Inflammation Index is an Adverse Prognostic Factor for Patients with Gastroesophageal Adenocarcinoma. Ann Surg, 2021, 273(3): 532–41., DOI 10.1097/SLA.0000000000003370 Zeng Q, Li J, Sun N, Xue Q, Gao Y, Zhao J, et al. Preoperative Systemic Immune-Inflammation index Predicts Survival and Recurrence in Patients with Resected Primary Pulmonary Sarcomatoid Carcinoma. Transl Lung Cancer Res, 2021, 10(1):18–31., DOI 10.21037/tlcr-20-960 Aziz MH, Sideras K, Aziz NA, Mauff K, Haen R, Roos D, et al. The Systemic- Immune-Inflammation Index Independently Predicts Survival and Recurrence in Resectable Pancreatic Cancer and its Prognostic Value Depends on Bilirubin Levels. Ann Surg, 2019, 270(1):139–46., DOI 10.1097/SLA.0000000000002660 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610294 EP 1610305 DI 10.3389/pore.2022.1610294 PG 12 ER PT J AU Ladanyi, A Hegyi, B Balatoni, T Liszkay, G Rohregger, R Waldnig, Ch Dudas, J Ferrone, S AF Ladanyi, Andrea Hegyi, Barbara Balatoni, Timea Liszkay, Gabriella Rohregger, Raphael Waldnig, Christoph Dudas, Jozsef Ferrone, Soldano TI HLA Class I Downregulation in Progressing Metastases of Melanoma Patients Treated With Ipilimumab SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunotherapy; melanoma; ipilimumab; HLA class I expression; longitudinal study ID immunotherapy; melanoma; ipilimumab; HLA class I expression; longitudinal study AB Characterization of the molecular mechanisms underlying antitumor immune responses and immune escape mechanisms has resulted in the development of more effective immunotherapeutic strategies, including immune checkpoint inhibitor (ICI) therapy. ICIs can induce durable responses in patients with advanced cancer in a wide range of cancer types, however, the majority of the patients fail to respond to this therapy or develop resistance in the course of the treatment. Information about the molecular mechanisms underlying primary and acquired resistance is limited. Although HLA class I molecules are crucial in the recognition of tumor antigens by cytotoxic T lymphocytes, only a few studies have investigated the role of their expression level on malignant cells in ICI resistance. To address this topic, utilizing immunohistochemical staining with monoclonal antibodies (mAbs) we analyzed HLA class I expression level in pre-treatment and post-treatment tumor samples from melanoma patients treated with ipilimumab. Twenty-nine metastases removed from six patients were available for the study, including 18 pre-treatment and 11 post-treatment lesions. Compared to metastases excised before ipilimumab therapy, post-treatment lesions displayed a significantly lower HLA class I expression level on melanoma cells; HLA class I downregulation was most marked in progressing metastases from nonresponding patients. We also evaluated the level of infiltration by CD8+ T cells and NK cells but did not find consistent changes between pre- and post-treatment samples. Our results indicate the potential role of HLA class I downregulation as a mechanism of ICI resistance. C1 [Ladanyi, Andrea] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Hegyi, Barbara] Orszagos Onkologiai Intezet, Mellkasi es Hasuregi Daganatok es Klinikai Farmakologiai OsztalyBudapest, Hungary. [Balatoni, Timea] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Liszkay, Gabriella] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Rohregger, Raphael] Medical University Innsbruck, Department of OtorhinolaryngologyInnsbruck, Austria. [Waldnig, Christoph] Medical University Innsbruck, Department of OtorhinolaryngologyInnsbruck, Austria. [Dudas, Jozsef] Medical University Innsbruck, Department of OtorhinolaryngologyInnsbruck, Austria. [Ferrone, Soldano] Massachusetts General Hospital, Harvard Medical School, Department of SurgeryBoston, MA, USA. RP Ladanyi, A (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Budapest, Hungary. EM ladanyi.andrea@oncol.hu CR Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N Engl J Med, 2010, 363:711–23., DOI 10.1056/ NEJMoa1003466 Robert C, Thomas L, Bondarenko I, O’Day S, Weber J, Garbe C, et al. Ipilimumab Plus Dacarbazine for Previously Untreated Metastatic Melanoma. N Engl J Med, 2011, 364:2517–26., DOI 10.1056/ NEJMoa11046210.1056/nejmoa1104621 Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell, 2017, 168:707–23., DOI 10.1016/j.cell.2017.01.017 Gide TN, Wilmott JS, Scolyer RA, Long GV. Primary and Acquired Resistance to Immune Checkpoint Inhibitors in Metastatic Melanoma. Clin Cancer Res, 2018, 24:1260–70., DOI 10.1158/1078-0432.CCR-17-2267 Zhou B, Gao Y, Zhang P, Chu Q. Acquired Resistance to Immune Checkpoint Blockades: the Underlying Mechanisms and Potential Strategies. Front Immunol, 2021, 12:693609., DOI 10.3389/fimmu.2021. 693609 Anagnostou V, Smith KN, Forde PM, Niknafs N, Bhattacharya R,White J, et al. Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non-small Cell Lung Cancer. Cancer Discov, 2017, 7:264–76., DOI 10.1158/2159-8290.CD-16-0828 Kakavand H, Jackett LA, Menzies AM, Gide TN, Carlino MS, Saw RPM, et al. Negative Immune Checkpoint Regulation by VISTA: aMechanism of Acquired Resistance to Anti-PD-1 Therapy in Metastatic Melanoma Patients. Mod Pathol, 2017, 30:1666–76., DOI 10.1038/modpathol.2017.8 Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu- Lieskovan S, et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med, 2016, 375:819–29., DOI 10.1056/ NEJMoa1604958 Gettinger S, Choi J, Hastings K, Truini A, Datar I, Sowell R, et al. Impaired HLA Class I Antigen Processing and Presentation as a Mechanism of Acquired Resistance to Immune Checkpoint Inhibitors in Lung Cancer. Cancer Discov, 2017, 7:1420–35., DOI 10.1158/2159-8290.CD-17-0593 Sade-Feldman M, Jiao YJ, Chen JH, Rooney MS, Barzily-Rokni M, Eliane J-P, et al. Resistance to Checkpoint Blockade Therapy through Inactivation of Antigen Presentation. Nat Commun, 2017, 8:1136., DOI 10.1038/s41467-017-01062-w Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, et al. Primary Resistance to PD-1 BlockadeMediated by JAK1/2 Mutations. Cancer Discov, 2017, 7:188–201., DOI 10.1158/2159-8290.CD- 16-1223 Chowell D, Morris LGT, Grigg CM, Weber JK, Samstein RM, Makarov V, et al. Patient HLA Class I Genotype Influences Cancer Response to Checkpoint Blockade Immunotherapy. Science, 2018, 359:582–7., DOI 10.1126/science.aao4572 Chowell D, Krishna C, Pierini F, Makarov V, Rizvi NA, Kuo F, et al. Evolutionary Divergence of HLA Class I Genotype Impacts Efficacy of Cancer Immunotherapy. Nat Med, 2019, 25:1715–20., DOI 10.1038/ s41591-019-0639-4 Manczinger M, Koncz B, Balogh GM, Papp BT, Asztalos L, Kemeny L, et al. Negative Trade-Off between Neoantigen Repertoire Breadth and the Specificity of HLA-I Molecules Shapes Antitumor Immunity. Nat Cancer, 2021, 2:950–61., DOI 10.1038/s43018-021-00226-4 Cai L,Michelakos T, Yamada T, Fan S,Wang X, Schwab JH, et al. Defective HLA Class I Antigen Processing Machinery in Cancer. Cancer Immunol Immunother, 2018, 67:999–1009., DOI 10.1007/s00262-018-2131-2 Maggs L, Sadagopan A, Moghaddam AS, Ferrone S. HLA Class I Antigen Processing Machinery Defects in Antitumor Immunity and Immunotherapy. Trends Cancer, 2021, 7:1089–101., DOI 10.1016/j.trecan.2021.07.006 Campoli M, Ferrone S. HLA Antigen Changes in Malignant Cells: Epigenetic Mechanisms and Biologic Significance. Oncogene, 2008, 27:5869–85., DOI 10. 1038/onc.2008.273 Rodig SJ, Gusenleitner D, Jackson DG, Gjini E, Giobbie-Hurder A, Jin C, et al. MHC Proteins Confer Differential Sensitivity to CTLA-4 and PD-1 Blockade in Untreated Metastatic Melanoma. Sci Transl Med, 2018, 10:eaar3342., DOI 10. 1126/scitranslmed.aar3342 Ladanyi A, Papp E, Mohos A, Balatoni T, Liszkay G, Olah J, et al. Role of the Anatomic Site in the Association of HLA Class I Antigen Expression Level in Metastases with Clinical Response to Ipilimumab Therapy in Patients with Melanoma. J Immunother Cancer, 2020, 8:e000209., DOI 10.1136/jitc-2019- 000209 Ugurel S, Spassova I, Wohlfarth J, Drusio C, Cherouny A, Melior A, et al.MHC Class-I Downregulation in PD-1/PD-L1 Inhibitor Refractory Merkel Cell Carcinoma and its Potential Reversal by Histone Deacetylase Inhibition: a Case Series. Cancer Immunol Immunother, 2019, 68:983–90., DOI 10.1007/ s00262-019-02341-9 Wolchok JD, Hoos A, O’Day S,Weber JS, Hamid O, Lebbe C, et al. Guidelines for the Evaluation of Immune Therapy Activity in Solid Tumors: Immune- Related Response Criteria. Clin Cancer Res, 2009, 15:7412–20., DOI 10.1158/ 1078-0432.CCR-09-1624 Pellegrino MA, Ng A-K, Russo C, Ferrone S. Heterogeneous Distribution of the Determinants Defined by Monoclonal Antibodies on HLA-A and B Antigens Bearing Molecules. Transplantation, 1982, 34:18–23., DOI 10.1097/ 00007890-198207000-00004 Stam NJ, Vroom TM, Peters PJ, Pastoors EB, Ploegh HL. HLA-A- and HLA-Bspecific Monoclonal Antibodies Reactive with Free Heavy Chains in Western Blots, in Formalin-Fixed, Paraffin-Embedded Tissue Sections and in Cryo- Immuno-Electron Microscopy. Int Immunol, 1990, 2:113–25., DOI 10.1093/ intimm/2.2.113 Balatoni T, Mohos A, Papp E, Sebestyen T, Liszkay G, Olah J, et al. Tumorinfiltrating Immune Cells as Potential Biomarkers Predicting Response to Treatment and Survival in Patients with Metastatic Melanoma Receiving Ipilimumab Therapy. Cancer Immunol Immunother, 2018, 67:141–51., DOI 10.1007/s00262-017-2072-1 Ingruber J, Savic D, Steinbichler TB, Sprung S, Fleischer F, Glueckert R, et al. KLF4, Slug and EMT in Head and Neck Squamous Cell Carcinoma. Cells, 2021, 10:539., DOI 10.3390/cells10030539 Steinbichler TB, Dudas J, Ingruber J, Glueckert R, Sprung S, Fleischer F, et al. Slug Is a Surrogate Marker of Epithelial to Mesenchymal Transition, EMT, in Head and Neck Cancer. J Clin Med, 2020, 9:2061., DOI 10.3390/jcm9072061 Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S. Escape of Human Solid Tumors from T-Cell Recognition: Molecular Mechanisms and Functional Significance. Adv Immunol, 2000, 74:181–273., DOI 10.1016/s0065-2776(08, 60911-6 Restifo NP, Marincola FM, Kawakami Y, Taubenberger J, Yannelli JR, Rosenberg SA. Loss of Functional Beta2-Microglobulin in Metastatic Melanomas from Five Patients Receiving Immunotherapy. JNCI J Natl Cancer Inst, 1996, 88:100–8., DOI 10.1093/jnci/88.2.100 Benitez R, Godelaine D, Lopez-Nevot MA, Brasseur F, Jimenez P, Marchand M, et al. Mutations of the β2-Microglobulin Gene Result in a Lack of HLA Class IMolecules on Melanoma Cells of Two Patients Immunized with MAGE Peptides. Tissue Antigens, 1998, 52:520–9., DOI 10.1111/j.1399-0039.1998. tb03082.x Tran E, Robbins PF, Lu Y-C, Prickett TD, Gartner JJ, Jia L, et al. T-cell Transfer Therapy Targeting Mutant KRAS in Cancer. N Engl JMed, 2016, 375:2255–62., DOI 10.1056/NEJMoa1609279 Liu D, Schilling B, Liu D, Sucker A, Livingstone E, Jerby-Arnon L, et al. Integrative Molecular and Clinical Modeling of Clinical Outcomes to PD1 Blockade in Patients with Metastatic Melanoma. Nat Med, 2019, 25:1916–27., DOI 10.1038/s41591-019-0654-5 Shim JH, Kim HS, Cha H, Kim S, Kim TM, Anagnostou V, et al. HLAcorrected Tumor Mutation burden and Homologous Recombination Deficiency for the Prediction of Response to PD-(L)1 Blockade in Advanced Non-small-cell Lung Cancer Patients. Ann Oncol, 2020, 31: 902–11., DOI 10.1016/j.annonc.2020.04.004 Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, et al. Genomic Correlates of Response to CTLA-4 Blockade in Metastatic Melanoma. Science, 2015, 350:207–11., DOI 10.1016/j.annonc.2020. 04.004 Thompson JC, Davis C, Deshpande C, Hwang WT, Jeffries S, Huang A, et al. Gene Signature of Antigen Processing and Presentation Machinery Predicts Response to Checkpoint Blockade in Non-small Cell Lung Cancer, NSCLC, and Melanoma. J Immunother Cancer, 2020, 8:e000974., DOI 10.1136/jitc- 2020-000974 Chen PL, Roh W, Reuben A, Cooper ZA, Spencer CN, Prietov PA, et al. Analysis of Immune Signatures in Longitudinal Tumor Samples Yields Insight into Biomarkers of Response and Mechanisms of Resistance to Immune Checkpoint Blockade. Cancer Discov, 2016, 6:827–37., DOI 10.1158/2159- 8290.CD-15-154 Yoo SH, Yun J, Keam B, Hong S-P, Ock C-Y, Koh J, et al. Discovery of Acquired Molecular Signature on Immune Checkpoint Inhibitors in Paired Tumor Tissues. Cancer Immunol Immunother, 2021, 70:1755–69., DOI 10. 1007/s00262-020-02799-y Lee JH, Shklovskaya E, Lim SY, Carlino MS, Menzies AM, Stewart A, et al. Transcriptional Downregulation of MHC Class I and Melanoma De- Differentiation in Resistance to PD-1 Inhibition. Nat Commun, 2020, 11: 1897., DOI 10.1038/s41467-020-15726-7 HamidO, Schmidt H, Nissan A, Ridolfi L, Aamdal S, Hansson J, et al.AProspective Phase II Trial Exploring the Association between Tumor Microenvironment Biomarkers and Clinical Activity of Ipilimumab in Advanced Melanoma. J Transl Med, 2011, 9:204., DOI 10.1186/1479-5876-9-204 Ji R-R, Chasalow SD, Wang L, HamidO, Schmidt H, Cogswell J, et al. An Immune- Active TumorMicroenvironment Favors Clinical Response to Ipilimumab. Cancer Immunol Immunother, 2012, 61:1019–31., DOI 10.1007/s00262-011-1172-6 TumehPC, HarviewCL, Yearley JH, Shintaku IP,TaylorEJM,Robert L, et al. PD-1 Blockade Induces Responses by Inhibiting Adaptive Immune Resistance. Nature, 2014, 515:568–71., DOI 10.1038/nature13954 Vilain RE, Menzies AM, Wilmott JS, Kakavand H, Madore J, Guminski A, et al. Dynamic Changes in PD-L1 Expression and Immune Infiltrates Early during Treatment Predict Response to PD-1 Blockade in Melanoma. Clin Cancer Res, 2017, 23:5024–33., DOI 10.1158/1078-0432.CCR-16-0698 Sharma A, Subudhi SK, Blando J, Scutti J, Vence L, Wargo J, et al. Anti- CTLA-4 Immunotherapy Does Not Deplete FOXP3+ Regulatory T Cells, Tregs, in Human Cancers. Clin Cancer Res, 2019, 25:1233–8., DOI 10. 1158/1078-0432.CCR-18-0762 Huang RR, Jalil J, Economou JS, Chmielowski B, Koya RC, Mok S, et al. CTLA4 Blockade Induces Frequent Tumor Infiltration by Activated Lymphocytes Regardless of Clinical Responses in Humans. Clin Cancer Res, 2011, 17:4101–9., DOI 10.1158/1078-0432.CCR-11-040 Erdag G, Schaefer JT, SchaeferME, Deacon DH, Shea SM, Dengel LT, et al. Immunotype and Immunohistologic Characteristics of Tumor-Infiltrating Immune Cells Are Associated with Clinical Outcome in Metastatic Melanoma. Cancer Res, 2012, 72:1070–80., DOI 10.1158/0008-5472. CAN-11-3218 Lee H, Quek C, Silva I, Tasker A, Batten M, Rizos H, et al. Integrated Molecular and Immunophenotypic Analysis of NK Cells in Anti-PD-1 Treated Metastatic Melanoma Patients. Oncoimmunology, 2018, 8: e1537581., DOI 10.1080/2162402X.2018.1537581 Edwards J, Wilmott JS, Madore J, Gide TN, Quek C, Tasker A, et al. CD103+ Tumor-Resident CD8+ T Cells Are Associated with Improved Survival in Immunotherapy-Naive Melanoma Patients and Expand Significantly during Anti-PD-1 Treatment. Clin Cancer Res, 2018, 24: 3036–45., DOI 10.1158/1078-0432.CCR-17-2257 Ladanyi A, Timar J. Immunologic and Immunogenomic Aspects of Tumor Progression. Semin Cancer Biol, 2020, 60:249–61., DOI 10.1016/j. semcancer.2019.08.011 Wu Y, Biswas D, Swanton C. Impact of Cancer Evolution on Immune Surveillance and Checkpoint Inhibitor Response. Semin Cancer Biol, 2021)., DOI 10.1016/j.semcancer.2021.02.013 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610297 EP 1610304 DI 10.3389/pore.2022.1610297 PG 8 ER PT J AU Gao, L Chen, G Liang, ZQ Li, JD Li, DM Tang, YL Tang, D Huang, ZG Chen, JH Luo, JY Zeng, JH Dang, YW Feng, ZB AF Gao, Li Chen, Gang Liang, Zi-Qian Li, Jian-Di Li, Dong-Ming Tang, Yu-Lu Tang, Deng Huang, Zhi-Guang Chen, Jun-Hong Luo, Jia-Yuan Zeng, Jiang-Hui Dang, Yi-Wu Feng, Zhen-Bo TI Expression Profile and Molecular Basis of Cyclin-Dependent Kinases Regulatory Subunit 2 in Endometrial Carcinoma Detected by Diversified Methods SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE molecular mechanism; endometrial carcinoma; RNA-seq; CKS2; in-house tissue microarray ID molecular mechanism; endometrial carcinoma; RNA-seq; CKS2; in-house tissue microarray AB Purpose: Our purpose was to systematically appraise the clinicopathological significance and explore the molecular bases of CKS2 in endometrial carcinoma. Patients and Methods: We measured the clinicopathological significance of CKS2 using diverse methods of public RNA-seq, microarrays, and in-house tissue microarrays to investigate the molecular basis of CKS2 in endometrial carcinoma through upstream transcriptional analysis, immune infiltration correlation analysis, and co-expression analysis. Results: Both the analysis for public RNA-seq plus the microarray data and in-house tissue microarray confirmed the significant overexpression of CKS2 in a total of 1,021 endometrial carcinoma samples compared with 279 non-cancer endometrium samples (SMD = 2.10, 95% CI = 0.72–3.48). The upregulated CKS2 was significantly related to the lymph node metastasis and advanced clinical grade of endometrial carcinoma patients (p < 0.001). Mutation types such as amplification and mRNA occurred with high frequency in the CKS2 gene in endometrial carcinoma patients. A series of miRNAs and transcription factors, such as hsa-miR-26a, hsa-miR-130a, hsa-miR-30, E2F4, MAX, and GABPA, were predicted to regulate the transcription and expression of CKS2. Significant links were found between CKS2 expression and the infiltration level of B cells, CD4+ T cells, and neutrophils in endometrial carcinoma. CKS2-coexpressed genes were actively involved in pathways such as the mitotic cell cycle process, PID aurora B pathway, and prolactin signaling pathway. Conclusion: The overexpressed CKS2 showed positive correlations with the clinical progression of endometrial carcinoma and was associated with various cancer-related biological processes and pathways, showing potential as a promising clinical biomarker for endometrial carcinoma. C1 [Gao, Li] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Chen, Gang] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Liang, Zi-Qian] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Li, Jian-Di] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Li, Dong-Ming] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Tang, Yu-Lu] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Tang, Deng] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Huang, Zhi-Guang] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Chen, Jun-Hong] Guangxi Maternal and Child Health Hospital, Department of PathologyNanning, China. [Luo, Jia-Yuan] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Zeng, Jiang-Hui] The Third Affiliated Hospital of Guangxi Medical University/Nanning Second People’s Hospital, Department of Clinical LaboratoryNanning, China. [Dang, Yi-Wu] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Feng, Zhen-Bo] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. RP Feng, ZB (reprint author), The First Affiliated Hospital of Guangxi Medical University, Department of Pathology, Nanning, China. EM fengzhenbo_gxmu@163.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Fader AN, Arriba LN, Frasure HE, von Gruenigen VE Endometrial Cancer and Obesity: Epidemiology, Biomarkers, Prevention and Survivorship. Gynecol Oncol, 2009, 114:121–7., DOI 10.1016/j.ygyno.2009.03.039 Crosbie EJ, Kitson SJ, McAlpine JN, Mukhopadhyay A, Powell ME, Singh N Endometrial Cancer. Lancet, 2022, 399:1412–28., DOI 10.1016/S0140-6736(22, 00323-3 ZhangK, Li H, YanY,ZangY, LiK,WangY, et al. Identification of Key Genes and Pathways between Type I and Type II Endometrial Cancer Using Bioinformatics Analysis. Oncol Lett, 2019, 18:2464–76., DOI 10.3892/ol.2019.10550 Setiawan VW, Yang HP, Pike MC, McCann SE, Yu H, Xiang Y-B, et al. Type I and II Endometrial Cancers: Have They Different Risk Factors? J Clin Oncol, 2013, 31:2607–18., DOI 10.1200/JCO.2012.48.2596 Eriksson AGZ, Davidson B, Bjerre Trent P, Eyjolfsdottir B, Dahl GF, Wang Y, et al. Update on Sentinel Lymph Node Biopsy in Surgical Staging of Endometrial Carcinoma. J Clin Med, 2021, 10:3094., DOI 10.3390/jcm10143094 Straubhar AM, Parsons MW, Francis S, Gaffney D, Maurer KA Refusal of Surgery and Survival Outcomes in Endometrial Cancer. Int J Gynecol Cancer, 2021, 31:1236–41., DOI 10.1136/ijgc-2021-002692 Wang H, Li W, Li N Efficacy of Laparoscopic, Pelvic and Para-Aortic Lymphadenectomy in the Treatment of Endometrial Carcinoma. J BUON, 2021, 26:1796–801. Jiang P, Yuan R Analysis of Factors Related to Lymph Node Metastasis in Early-Stage Type 1 Endometrial Cancer: Verifying the Clinical Value of Positive Threshold of the Immunohistochemical Parameter Ki67. Cancer Manag Res, 2021, 13:6319–28., DOI 10.2147/CMAR.S316211 Sasano H,Watanabe K, Ito K, Sato S, Yajima A New Concepts in the Diagnosis and Prognosis of Endometrial Carcinoma. Pathol Annu, 1994, 29(Pt 2):31–49. Wang J-J, Fang Z-X, Ye H-M, You P, Cai M-J, Duan H-B, et al. Clinical Significance of Overexpressed Cyclin-dependent Kinase Subunits 1 and 2 in Esophageal Carcinoma. Dis Esophagus, 2013, 26:729., DOI 10.1111/dote.12013 Wu Z, Lu B, Li X, Miao W, Li J, Shi Y, et al. MicroRNA-26a Inhibits Proliferation and Tumorigenesis via Targeting CKS2 in Laryngeal Squamous Cell Carcinoma. Clin Exp Pharmacol Physiol, 2018, 45:444–51., DOI 10.1111/1440-1681.12890 Zhang X-M, Wang J, Liu Z-L, Liu H, Cheng Y-F, Wang T LINC00657/miR- 26a-5p/CKS2 ceRNA Network Promotes the Growth of Esophageal Cancer Cells via the MDM2/p53/Bcl2/Bax Pathway. Biosci Rep, 2020, 40: BSR20200525., DOI 10.1042/BSR20200525 Xu JH,Wang Y, Xu D CKS2 Promotes Tumor Progression andMetastasis and Is an Independent Predictor of Poor Prognosis in Epithelial Ovarian Cancer. Eur Rev Med Pharmacol Sci, 2019, 23:3225–34., DOI 10.26355/eurrev_201904_17681 Shen D-Y, Zhan Y-H, Wang Q-M, Rui G, Zhang Z-M Oncogenic Potential of Cyclin Kinase Subunit-2 in Cholangiocarcinoma. Liver Int, 2013, 33:137–48., DOI 10.1111/liv.12014 Kita Y, Nishizono Y, Okumura H, Uchikado Y, Sasaki K, Matsumoto M, et al. Clinical and Biological Impact of Cyclin-dependent Kinase Subunit 2 in Esophageal Squamous Cell Carcinoma. Oncol Rep, 2014, 31:1986–92., DOI 10.3892/or.2014.3062 Gao L, He R-q., Wu H-y., Zhang T-t., Liang H-w., Ye Z-h., et al. Expression Signature and Role of miR-30d-5p in Non-small Cell Lung Cancer: a Comprehensive Study Based on In Silico Analysis of Public Databases and In Vitro Experiments. Cell Physiol Biochem, 2018, 50:1964–87., DOI 10.1159/ 000494875 Gao L, Pang Y-Y, Guo X-Y, Zeng J-J, Tang Z-Q, Xiong D-D, et al. Polo like Kinase 1 Expression in Cervical Cancer Tissues Generated from Multiple Detection Methods. PeerJ, 2020, 8:e10458., DOI 10.7717/peerj.10458 Huang W-J, He W-Y, Li J-D, He R-Q, Huang Z-G, Zhou X-G, et al. Clinical Significance and Molecular Mechanism of Angiotensin-Converting Enzyme 2 in Hepatocellular Carcinoma Tissues. Bioengineered, 2021, 12:4054–69., DOI 10.1080/21655979.2021.1952791 Li G-S, Hou W, Chen G, Yao Y-X, Chen X-Y, Zhang X-G, et al. Clinical Significance of Integrin Subunit Beta 4 in Head and Neck Squamous Cell Carcinoma. Cancer Biother Radiopharm, 2022, 37:256–75., DOI 10.1089/cbr. 2020.3943 Yang Z, Cheng H, Zhang Y, Zhou Y Identification of NDRG Family Member 4, NDRG4, and CDC28 Protein Kinase Regulatory Subunit 2, CKS2, as Key Prognostic Genes in Adrenocortical Carcinoma by Transcriptomic Analysis. Med Sci Monit, 2021, 27:e928523., DOI 10.12659/MSM.928523 Xiao D, Dong S, Yang S, Liu Z CKS2 and RMI2 Are Two Prognostic Biomarkers of Lung Adenocarcinoma. PeerJ, 2020, 8:e10126., DOI 10.7717/ peerj.10126 Yu MH, Luo Y, Qin SL, Wang ZS, Mu YF, Zhong M Up-regulated CKS2 Promotes Tumor Progression and Predicts a Poor Prognosis in Human Colorectal Cancer. Am J Cancer Res, 2015, 5:2708–18. Gao F, Li C, Zhao X, Xie J, Fang G, Li Y CKS2 Modulates Cell-cycle Progression of Tongue Squamous Cell Carcinoma Cells Partly via Modulating the Cellular Distribution of DUTPase. J Oral Pathol Med, 2021, 50:175–82., DOI 10.1111/jop.13116 Zhang J, Song Q, Liu J, Lu L, Xu Y, Zheng W Cyclin-Dependent Kinase Regulatory Subunit 2 Indicated Poor Prognosis and Facilitated Aggressive Phenotype of Hepatocellular Carcinoma. Dis Markers, 2019, 2019:1–13., DOI 10.1155/2019/8964015 del Rincon SV, Widschwendter M, Sun D, Ekholm-Reed S, Tat J, Teixeira LK, et al. Cks Overexpression Enhances Chemotherapeutic Efficacy by Overriding DNA Damage Checkpoints. Oncogene, 2015, 34:1961–7., DOI 10.1038/onc. 2014.137 Liberal V, Martinsson-Ahlzen H-S, Liberal J, Spruck CH, Widschwendter M, McGowan CH, et al. Cyclin-dependent Kinase Subunit, Cks, 1 or Cks2 Overexpression Overrides the DNA Damage Response Barrier Triggered by Activated Oncoproteins. Proc Natl Acad Sci U.S.A, 2012, 109:2754–9., DOI 10. 1073/pnas.1102434108 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610307 EP 1610319 DI 10.3389/pore.2022.1610307 PG 13 ER PT J TI Detection and Quantification of ctDNA for Longitudinal Monitoring of Treatment in Non-Small Cell Lung Cancer Patients Using a Universal Mutant Detection Assay by Denaturing Capillary Electrophoresis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE liquid biopsy; NSCLC; ctDNA; KRAS mutations; minimal residual disease; capillary electrophoresis; TP53 mutations ID liquid biopsy; NSCLC; ctDNA; KRAS mutations; minimal residual disease; capillary electrophoresis; TP53 mutations AB Background: Observation of anticancer therapy effect by monitoring of minimal residual disease (MRD) is becoming an important tool in management of non-small cell lung cancer (NSCLC). The approach is based on periodic detection and quantification of tumorspecific somatic DNA mutation in circulating tumor DNA (ctDNA) extracted from patient plasma. For such repetitive testing, complex liquid-biopsy techniques relying on ultra-deep NGS sequencing are impractical. There are other, cost-effective, methods for ctDNA analysis, typically based on quantitative PCR or digital PCR, which are applicable for detecting specific individual mutations in hotspots. While such methods are routinely used in NSCLC therapy prediction, however, extension to cover broader spectrum of mutations (e.g., in tumor suppressor genes) is required for universal longitudinal MRD monitoring. Methods: For a set of tissue samples from 81 NSCLC patients we have applied a denaturing capillary electrophoresis (DCE) for initial detection of somatic mutations within 8 predesigned PCR amplicons covering oncogenes and tumor suppressor genes. Mutationnegative samples were then subjected to a large panel NGS sequencing. For each patient mutation found in tissue was then traced over time in ctDNA by DCE. Results: In total we have detected a somatic mutation in tissue of 63 patients. For those we have then prospectively analyzed ctDNA from collected plasma samples over a period of up to 2 years. The dynamics of ctDNA during the initial chemotherapy therapy cycles as well as in the long-term follow-up matched the clinically observed response. Conclusion: Detection and quantification of tumor-specific mutations in ctDNA represents a viable complement to MRD monitoring during therapy of NSCLC patients. The presented approach relying on initial tissue mutation detection by DCE combined with NGS and a subsequent ctDNA mutation testing by DCE only represents a cost-effective approach for its routine implementation. CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Ettinger DS,Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, et al. NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 2.2021. J Natl Compr Canc Netw, 2021, 19(3):254–66., DOI 10.6004/jnccn.2021. 0013 Gubens MA, Davies M. NCCN Guidelines Updates: New Immunotherapy Strategies for Improving Outcomes in Non-Small Cell Lung Cancer. J Natl Compr Canc Netw, 2019, 17:574–8., DOI 10.6004/jnccn.2019.5005 Malone ER, Oliva M, Sabatini PJB, Stockley TL, Siu LL. Molecular Profiling for Precision Cancer Therapies. Genome Med, 2020, 12(1):8., DOI 10.1186/s13073- 019-0703-1 Theodoropoulos AS, Gkiozos I, Kontopyrgias G, Charpidou A, Kotteas E, Kyrgias G, et al. Modern Radiopharmaceuticals for Lung Cancer Imaging with Positron Emission Tomography/Computed Tomography Scan: A Systematic Review. SAGE Open Med, 2020, 8:205031212096159., DOI 10.1177/ 2050312120961594 Boonstra PA, Wind TT, van Kruchten M, Schuuring E, Hospers GAP, van der Wekken AJ, et al. Clinical Utility of Circulating Tumor DNA as a Response and Follow-Up Marker in Cancer Therapy. Cancer Metastasis Rev, 2020, 39(3):999–1013., DOI 10.1007/s10555-020-09876-9 Schwarzenbach H, Hoon DSB, Pantel K. Cell-Free Nucleic Acids as Biomarkers in Cancer Patients. Nat Rev Cancer, 2011, 11(6):426–37., DOI 10.1038/nrc3066 Diaz LA, Bardelli A. Liquid Biopsies: Genotyping Circulating Tumor DNA. J Clin Oncol, 2014, 32(6):579–86., DOI 10.1200/JCO.2012.45.2011 Peng Y,Mei W, Ma K, Zeng C. Circulating Tumor DNA andMinimal Residual Disease, MRD, in Solid Tumors: Current Horizons and Future Perspectives. Front Oncol, 2021, 11:763790., DOI 10.3389/fonc.2021.763790 Zheng D, Ye X, Zhang MZ, Sun Y, Wang JY, Ni J, et al. Plasma EGFR T790M ctDNA Status is Associated with Clinical Outcome in Advanced NSCLC Patients with Acquired EGFR-TKI Resistance. Sci Rep, 2016, 6:20913., DOI 10. 1038/srep20913 Karlovich C, Goldman JW, Sun J-M, Mann E, Sequist LV, Konopa K, et al. Assessment of EGFR Mutation Status in Matched Plasma and Tumor Tissue of NSCLC Patients from a Phase I Study of Rociletinib, CO-1686). Clin Cancer Res, 2016, 22(10):2386–95., DOI 10.1158/1078-0432.CCR-15-1260 Garrido P, Paz-Ares L, Majem M, Moran T, Trigo JM, Bosch-Barrera J, et al. LungBEAM: A Prospective Multicenter Study to Monitor Stage IV NSCLC Patients with EGFR Mutations Using BEAMing Technology. Cancer Med, 2021, 10(17):5878–88., DOI 10.1002/cam4.4135 Fernandes MGO, Sousa C, Pereira Reis J, Cruz-Martins N, Souto Moura C, Guimaraes S, et al. Liquid Biopsy for Disease Monitoring in Non-Small Cell Lung Cancer: The Link Between Biology and the Clinic. Cells, 2021, 10(8): 1912., DOI 10.3390/cells10081912 Metzenmacher M, Hegedus B, Forster J, Schramm A, Horn PA, Klein CA, et al. Combined Multimodal ctDNA Analysis and Radiological Imaging for Tumor Surveillance in Non-Small Cell Lung Cancer. Transl Oncol, 2022, 15(1): 101279., DOI 10.1016/j.tranon.2021.101279 de Kock R, Borne Bv. d., Soud MYE, Belderbos H, Stege G, de Saegher M, et al. Circulating Biomarkers for Monitoring Therapy Response and Detection of Disease Progression in Lung Cancer Patients. Cancer Treat Res Commun, 2021, 28:100410., DOI 10.1016/j.ctarc.2021.100410 Di Capua D, Bracken-Clarke D, Ronan K, Baird A-M, Finn S. The Liquid Biopsy for Lung Cancer: State of the Art, Limitations and Future Developments. Cancers, 2021, 13(16):3923., DOI 10.3390/cancers13163923 Thierry AR, El Messaoudi S, Gahan PB, Anker P, Stroun M. Origins, Structures, and Functions of Circulating DNA in Oncology. Cancer Metastasis Rev, 2016, 35(3):347–76., DOI 10.1007/s10555-016-9629-x Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, et al. Detection and Quantification of Mutations in the Plasma of Patients with Colorectal Tumors. Proc Natl Acad Sci U.S.A, 2005, 102(45):16368–73., DOI 10.1073/pnas.0507904102 Elazezy M, Joosse SA. Techniques of Using Circulating Tumor DNA as a Liquid Biopsy Component in Cancer Management. Comput Struct Biotechnol J, 2018, 16:370–8., DOI 10.1016/j.csbj.2018.10.002 Sefrioui D, Mauger F, Leclere L, Beaussire L, Di Fiore F, Deleuze J-F, et al. Comparison of the Quantification of KRAS Mutations by Digital PCR and E-Ice-COLD-PCR in Circulating-Cell-Free DNA from Metastatic Colorectal Cancer Patients. Clinica Chim Acta, 2017, 465:1–4., DOI 10.1016/j.cca.2016. 12.004 Chen H, Zhang J, Chen H-Y, Su B, Lu D. Establishment of Multiplex Allele- Specific Blocker PCR for Enrichment and Detection of 4 Common EGFR Mutations in Non-Small Cell Lung Cancer. Ann Transl Med, 2020, 8(22):1509., DOI 10.21037/atm-20-6754 Soussi T. The P53 Tumor Suppressor Gene: From Molecular Biology to Clinical Investigation. Ann NY Acad Sci, 2000, 910:121–39., DOI 10.1111/j. 1749-6632.2000.tb06705.x van Es JH, Giles RH, Clevers HC. The Many Faces of the Tumor Suppressor Gene APC. Exp Cel Res, 2001, 264(1):126–34., DOI 10.1006/excr.2000.5142 Minarikova P, Benesova L, Halkova T, Belsanova B, Suchanek S, Cyrany J, et al. Longitudinal Molecular Characterization of Endoscopic Specimens from Colorectal Lesions. World J Gastroenterol, 2016, 22(20):4936–45., DOI 10. 3748/wjg.v22.i20.4936 Benesova L, Halkova T, Ptackova R, Semyakina A, Menclova K, Pudil J, et al. Significance of Postoperative Follow-Up of Patients with Metastatic Colorectal Cancer Using Circulating Tumor DNA. World J Gastroenterol, 2019, 25(48): 6939–48., DOI 10.3748/wjg.v25.i48.6939 Pazdirek F, Minarik M, Benesova L, Halkova T, Belsanova B, Macek M, et al. Monitoring of Early Changes of Circulating Tumor DNA in the Plasma of Rectal Cancer Patients Receiving Neoadjuvant Concomitant Chemoradiotherapy: Evaluation for Prognosis and Prediction of Therapeutic Response. Front Oncol, 2020, 10:1028., DOI 10.3389/fonc.2020.01028 Lian D-S, Zhao S-J. Capillary Electrophoresis Based on the Nucleic Acid Detection in the Application of Cancer Diagnosis and Therapy. Analyst, 2014, 139(14):3492–506., DOI 10.1039/c4an00400k Fiala O, Pesek M, Finek J, Svaton M, Minarik M, Benesova L, et al. Pemetrexed versus Erlotinib in the Second-Line Treatment of Patients with Advanced- Stage Non-Squamous NSCLC Harboring Wild-Type EGFR Gene. Anticancer Res, 2016, 36(1):447–53. Bunganic B, Halkova T, Benesova L, Belsanova B, Laclav M, Hruzova M, et al. KRAS Mutation Assay on EUS-FNA Specimens from Pacients with Pancreatic Mass. Cas Lek Cesk, 2016, 155(1):48–51. Benesova L, Halkova T, Bunganic B, Belsanova B, Zavoral M, Traboulsi E, et al. Comparison of Native Aspirates and Cytological Smears Obtained by EUSGuided Biopsies for Effective DNA/RNA Marker Testing in Pancreatic Cancer. Pathol Oncol Res, 2020, 26(1):379–85., DOI 10.1007/s12253-018-0490-9 Benesova L, Belsanova B, Kramar F, Halkova T, Benes V, Minarik M. Application of Denaturing Capillary Electrophoresis for the Detection of Prognostic Mutations in Isocitrate Dehydrogenase 1 and Isocitrate Dehydrogenase 2 Genes in Brain Tumors. J Sep Sci, 2018, 41(13):2819–27., DOI 10.1002/jssc.201701473 Benesova L, Belsanova B, Suchanek S, Kopeckova M, Minarikova P, Lipska L, et al. Mutation-Based Detection and Monitoring of Cell-Free Tumor DNA in Peripheral Blood of Cancer Patients. Anal Biochem, 2013, 433(2):227–34., DOI 10.1016/j.ab.2012.06.018 Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New Response Evaluation Criteria in Solid Tumours: Revised RECIST Guideline, Version 1.1). Eur J Cancer, 2009, 45(2):228–47., DOI 10.1016/j. ejca.2008.10.026 Salek C, Minarikova P, Benesova L, Nosek V, Strnad R, Zavoral M, et al. Mutation Status of K-Ras, P53 and Allelic Losses at 9p and 18q are Not Prognostic Markers in Patients with Pancreatic Cancer. Anticancer Res, 2009, 29:1803–10. Bjorheim J, Minarik M, Gaudernack G, Ekstrom PO. Mutation Detection in KRAS Exon 1 by Constant Denaturant Capillary Electrophoresis in 96 Parallel Capillaries. Anal Biochem, 2002, 304(2):200–5., DOI 10.1006/abio.2002.5629 Newman AM, Bratman SV, To J,Wynne JF, Eclov NCW, Modlin LA, et al. An Ultrasensitive Method for Quantitating Circulating Tumor DNA with Broad Patient Coverage. Nat Med, 2014, 20(5):548–54., DOI 10.1038/nm.3519 Mogi A, Kuwano H. TP53Mutations in Nonsmall Cell Lung Cancer. J Biomed Biotechnol, 2011, 2011:1–9., DOI 10.1155/2011/583929 Baeissa H, Benstead-Hume G, Richardson CJ, Pearl FMG. Identification and Analysis of Mutational Hotspots in Oncogenes and Tumour Suppressors. Oncotarget, 2017, 8(13):21290–304., DOI 10.18632/oncotarget.15514 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610308 EP 1610317 DI 10.3389/pore.2022.1610308 PG 10 ER PT J AU Polk, N Budai, B Hitre, E Patocs, A Mersich, T AF Polk, Nandor Budai, Barna Hitre, Erika Patocs, Attila Mersich, Tamas TI High Neutrophil-To-Lymphocyte Ratio (NLR) and Systemic Immune-Inflammation Index (SII) Are Markers of Longer Survival After Metastasectomy of Patients With Liver-Only Metastasis of Rectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE neutrophil-to-lymphocyte ratio; liver-only metastases of rectal cancer; metastasectomy; relapse-free survival; systemic immune-inflammation index; liver-only metastases of colon cancer; preoperative treatment ID neutrophil-to-lymphocyte ratio; liver-only metastases of rectal cancer; metastasectomy; relapse-free survival; systemic immune-inflammation index; liver-only metastases of colon cancer; preoperative treatment AB Background: The literature data regarding colon cancer patients with liver-only metastases (CLM) show that NLR determined before metastasectomy is a prognostic marker of shorter relapse-free survival (RFS), but no results has been reported to date for rectal cancer patients with liver-only metastases (RLM). This study aimed to investigate the NLR and SII in CLM and RLM. Methods: Relapse-free (RFS) and overall survival (OS) were evaluated in 67 CLM and 103 RLM patients with a median follow-up of 46.5 and 59.8 months, respectively. Pre- and/or postoperative chemotherapy ± targeted treatment was applied in 96% and 87% of CLM and RLM patients, respectively. The cut-off level for hematologic parameters were determined by receiver operating characteristic (ROC) analysis. Univariate analysis was performed by Kaplan-Meier method and log rank test. For multivariate analysis Cox regression was applied. Results: In univariate analysis low NLR (cut-off 2) and SII (535) were predictors of longer RFS in case of CLM (p < 0.01). In contrast, for RLMhigh NLR (2.42) and SII (792) were predictors of longer RFS (p < 0.001). For RLM both NLR and SII proved to be independent markers of RFS (HR 0.66 (95% CI 0.52–0.84) and 0.73 (0.57–0.91), respectively) and OS (0.76 (0.58–0.99) and 0.66 (0.5–0.87), respectively). Only NLR (1.44 (1.04–1.99)) was independent marker of RFS for CLM. The preoperative treatment has not influenced the role of NLR or SII. Conclusion: In contrast to CLM, in RLM the high NLR or SII determined before metastasectomy proved to be independent prognostic factors of longer RFS and OS. C1 [Polk, Nandor] Orszagos Onkologiai Intezet, Emlo- es Lagyreszsebeszeti OsztalyBudapest, Hungary. [Budai, Barna] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Hitre, Erika] Orszagos Onkologiai Intezet, „B” Belgyogyaszati-Onkologiai es Klinikai Farmakologiai OsztalyBudapest, Hungary. [Patocs, Attila] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Mersich, Tamas] Orszagos Onkologiai Intezet, Emlo- es Lagyreszsebeszeti OsztalyBudapest, Hungary. RP Budai, B (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Budapest, Hungary. EM budai.barna@oncol.hu CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Liu W, Zhang W, Xu Y, Li Y-H, Xing B-C. A Prognostic Scoring System to Predict Survival Outcome of Resectable Colorectal Liver Metastases in This Modern Era. Ann Surg Oncol, 2021, 28:7709–18., DOI 10.1245/s10434-021- 10143-6 Alabraba E, Ibrahim H, Olaru A, Cameron I, Gomez D, Group NHS. Retrospective Cohort Study of Statin Therapy Effect on Resected Colorectal Liver Metastases. Wjgs, 2020, 12:34–44., DOI 10.4240/wjgs.v12.i2.34 Dupre A, Jones RP, Diaz-Nieto R, Fenwick SW, Poston GJ, Malik HZ. Preoperative Leucocyte-Based Inflammatory Scores in Patients with Colorectal Liver Metastases: Can We Count on Them? World J Surg, 2019, 43:1351–9., DOI 10.1007/s00268-019-04914-2 KimW-J, Lim T-W, Kang S-H, Park P-J, Choi S-B, Lee S-i., et al. Development and Validation of Novel Scoring System for the Prediction of Disease Recurrence Following Resection of Colorectal Liver Metastasis. Asian J Surg, 2020, 43(2):438–46., DOI 10.1016/j.asjsur.2019.06.001 Halazun KJ, Aldoori A, Malik HZ, Al-Mukhtar A, Prasad KR, Toogood GJ, et al. Elevated Preoperative Neutrophil to Lymphocyte Ratio Predicts Survival Following Hepatic Resection for Colorectal Liver Metastases. Eur J Surg Oncol, Ejso,, 2008, 34:55–60., DOI 10.1016/j.ejso.2007.02.014 Neal CP, Mann CD, Sutton CD, Garcea G, Ong SL, Steward WP, et al. Evaluation of the Prognostic Value of Systemic Inflammation and Socioeconomic Deprivation in Patients with Resectable Colorectal Liver Metastases. Eur J Cancer, 2009, 45:56–64., DOI 10.1016/j.ejca.2008.08.019 Western CE, Wiggans M, Aroori S, Bowles M, Stell D. PMO-089 Preoperative Neutrophil: Lymphocyte Ratio Is Not a Predictor of Outcome Following Hepatic Resection for Colorectal Metastases. Gut, 2012, 61:2–A109., DOI 10. 1136/gutjnl-2012-302514b.89 Yamashita S, Sheth RA, Niekamp AS, Aloia TA, Chun YS, Lee JE, et al. Comprehensive Complication index Predicts Cancer-specific Survival after Resection of Colorectal Metastases Independent of RAS Mutational Status. Ann Surg, 2017, 266:1045–54., DOI 10.1097/SLA.0000000000002018 Lu Y, Xin D, Wang F. Predictive Significance of Preoperative Systemic Immune-Inflammation index Determination in Postoperative Liver Metastasis of Colorectal Cancer. Ott, 2019, Vol. 12:7791–9., DOI 10.2147/ OTT.S223419 Wang Y-Y, Liu Z-Z, Xu D, Liu M, Wang K, Xing B-C. Fibrinogen-albumin Ratio index, FARI): a More Promising Inflammation-Based Prognostic Marker for Patients Undergoing Hepatectomy for Colorectal Liver Metastases. Ann Surg Oncol, 2019, 26:3682–92., DOI 10.1245/s10434-019- 07586-3 Deng Y, Zhao Y, Qin J, Huang X, Wu R, Zhou C, et al. Prognostic Value of the C-Reactive Protein/Albumin Ratio and Systemic Immune-Inflammation Index for Patients with Colorectal Liver Metastasis Undergoing Curative Resection. Pathol Oncol Res, 2021, 27:633480., DOI 10.3389/pore.2021.633480 Giakoustidis A, Neofytou K, Khan AZ, Mudan S. Neutrophil to Lymphocyte Ratio Predicts Pattern of Recurrence in Patients Undergoing Liver Resection for Colorectal Liver Metastasis and Thus the Overall Survival. J Surg Oncol, 2015, 111:445–50., DOI 10.1002/jso.23845 Neofytou K, Smyth EC, Giakoustidis A, Khan AZ, Cunningham D, Mudan S. Elevated Platelet to Lymphocyte Ratio Predicts Poor Prognosis after Hepatectomy for Liver-Only Colorectal Metastases, and it Is superior to Neutrophil to Lymphocyte Ratio as an Adverse Prognostic Factor. Med Oncol, 2014, 31:239., DOI 10.1007/s12032-014-0239-6 Mao R, Zhao J-J, Bi X-Y, Zhang Y-F, Li Z-Y, Huang Z, et al. A Low Neutrophil to Lymphocyte Ratio before Preoperative Chemotherapy Predicts Good Outcomes after the Resection of Colorectal Liver Metastases. J Gastrointest Surg, 2019, 23:563–70., DOI 10.1007/s11605-018-3796-8 Zhang Y, Peng Z, Chen M, Liu F, Huang J, Xu L, et al. Elevated Neutrophil to Lymphocyte Ratio Might Predict Poor Prognosis for Colorectal Liver Metastasis after Percutaneous Radiofrequency Ablation. Int J Hyperthermia, 2012, 28:132–40., DOI 10.3109/02656736.2011.654374 Kim H, Jung HI, Kwon SH, Bae SH, Kim HC, Baek M-J, et al. Preoperative Neutrophil-Lymphocyte Ratio and CEA Is Associated with Poor Prognosis in Patients with Synchronous Colorectal Cancer Liver Metastasis. Ann Surg Treat Res, 2019, 96:191–200., DOI 10.4174/astr.2019.96.4.191 Hand F, Ryan EJ, Harrington C, Durand M, Maguire D, O’Farrelly C, et al. Chemotherapy and Repeat Resection Abrogate the Prognostic Value of Neutrophil Lymphocyte Ratio in Colorectal Liver Metastases. Hpb, 2020, 22:670–6., DOI 10.1016/j.hpb.2019.09.003 Hamada T, Ishizaki H, Haruyama Y, Hamada R, Yano K, Kondo K, et al. Neutrophil-to-lymphocyte Ratio and Intratumoral CD45RO-Positive T Cells as Predictive Factors for Longer Survival of Patients with Colorectal Liver Metastasis after Hepatectomy. Tohoku J Exp Med, 2020, 251:303–11., DOI 10. 1620/tjem.251.303 Kishi Y, Kopetz S, Chun YS, Palavecino M, Abdalla EK, Vauthey J-N. Blood Neutrophil-To-Lymphocyte Ratio Predicts Survival in Patients with Colorectal Liver Metastases Treated with Systemic Chemotherapy. Ann Surg Oncol, 2009, 16:614–22., DOI 10.1245/s10434-008-0267-6 Komlosi V, Hitre E, Pap E, Adleff V, Reti A, Szekely E, et al. SHMT1 1420 and MTHFR 677 Variants Are Associated with Rectal but Not colon Cancer. BMC Cancer, 2010, 10:525., DOI 10.1186/1471-2407-10-525 Paschke S, Jafarov S, Staib L, Kreuser E-D, Maulbecker-Armstrong C, Roitman M, et al. Are colon and Rectal Cancer Two Different Tumor Entities? A Proposal to Abandon the Term Colorectal Cancer. Ijms, 2018, 19:2577., DOI 10. 3390/ijms19092577 Slattery ML, Wolff RK, Herrick JS, Caan BJ, Potter JD. IL6 Genotypes and colon and Rectal Cancer. Cancer Causes Control, 2007, 18:1095–105., DOI 10. 1007/s10552-007-9049-x Slattery ML, Curtin K, Poole EM, Duggan DJ, Samowitz WS, Peters U, et al. Genetic Variation in C-Reactive Protein in Relation to colon and Rectal Cancer Risk and Survival. Int J Cancer, 2011, 128:2726–34., DOI 10.1002/ijc.25721 Chang Z, Zheng J, Ma Y, Zhao J, Wang C, Liu Z. The Neutrophil-To- Lymphocyte Ratio as a Predictor for Recurrence of Colorectal Liver Metastases Following Radiofrequency Ablation. Med Oncol, 2014, 31:855., DOI 10.1007/s12032-014-0855-1 Sanyal S, Alabraba E, Ibrahim H, Olaru A, Cameron I, Gomez D. ACE Inhibitor Therapy Does Not Influence the Survival Outcomes of Patients with Colorectal Liver Metastases Following Liver Resection. J Gastrointest Canc, 2021, 52:106–12., DOI 10.1007/s12029-019-00350-6 Cimino MM, Donadon M, Giudici S, Sacerdote C, Di Tommaso L, Roncalli M, et al. Peri-tumoural CD3+ Inflammation and Neutrophil-To-Lymphocyte Ratio Predict Overall Survival in Patients Affected by Colorectal Liver Metastases Treated with Surgery. J Gastrointest Surg, 2020, 24:1061–70., DOI 10.1007/s11605-019-04458-9 Liu Y-W, Lu C-C, Chang C-D, Lee K-C, Chen HH, Yeh WS, et al. Prognostic Value of Sarcopenia in Patients with Colorectal Liver Metastases Undergoing Hepatic Resection. Sci Rep, 2020, 10:6459., DOI 10.1038/s41598-020-63644-x Peng J, Li H, Ou Q, Lin J, Wu X, Lu Z, et al. Preoperative Lymphocyte-To- Monocyte Ratio Represents a superior Predictor Compared with Neutrophil- To-Lymphocyte and Platelet-To-Lymphocyte Ratios for Colorectal Liver-Only Metastases Survival. Ott, 2017, Vol. 10:3789–99., DOI 10.2147/OTT.S140872 Taniai T, Haruki K, Hamura R, Fujiwara Y, Furukawa K, Gocho T, et al. The Prognostic Significance of C-Reactive Protein-To-Lymphocyte Ratio in Colorectal Liver Metastases. J Surg Res, 2021, 258:414–21., DOI 10.1016/j.jss.2020.08.059 Verter E, Berger Y, Perl G, Peretz I, Tovar A, Morgenstern S, et al. Neutrophilto- lymphocyte Ratio Predicts Recurrence Pattern in Patients with Resectable Colorectal Liver Metastases. Ann Surg Oncol, 2021, 28:4320–9., DOI 10.1245/ s10434-021-10000-6 Zeman M, Maciejewski A, Poltorak S, Kryj M. Evaluation of Outcomes and Treatment Safety of Patients with Metastatic Colorectal Cancer to the Liver with Estimation of Prognostic Factors. Pol Przegl Chir, 2013, 85:333–9., DOI 10. 2478/pjs-2013-0050 Neal CP, Mann CD, Garcea G, Briggs CD, Dennison AR, Berry DP. Preoperative Systemic Inflammation and Infectious Complications after Resection of Colorectal Liver Metastases. Arch Surg, 2011, 146:471–8., DOI 10.1001/archsurg.2011.50 Nemoto T, Endo S, Isohata N, Takayanagi D, Nemoto D, Aizawa M, et al. Change in the Neutrophil-To-Lymphocyte Ratio during Chemotherapy May Predict Prognosis in Patients with Advanced or Metastatic Colorectal Cancer. Mol Clin Oncol, 2021, 14:107., DOI 10.3892/mco.2021.2269 Eefsen RL, Engelholm L, Alpizar-Alpizar W, Van den Eynden GGE, Vermeulen PB, Christensen IJ, et al. Inflammation and uPAR-Expression in Colorectal Liver Metastases in Relation to Growth Pattern and Neo- Adjuvant Therapy. Cancer Microenvironment, 2015, 8:93–100., DOI 10.1007/ s12307-015-0172-z McCoyMJ, Hemmings C,Miller TJ, Austin SJ, Bulsara MK, Zeps N, et al. Low Stromal Foxp3+ Regulatory T-Cell Density Is Associated with Complete Response to Neoadjuvant Chemoradiotherapy in Rectal Cancer. Br J Cancer, 2015, 113:1677–86., DOI 10.1038/bjc.2015.427 Gunnarsson U, Strigard K, Edin S, Gkekas I, Mustonen H, Kaprio T, et al. Association between Local Immune Cell Infiltration, Mismatch Repair Status and Systemic Inflammatory Response in Colorectal Cancer. J Transl Med, 2020, 18:178., DOI 10.1186/s12967-020-02336-6 Kubo H, Murayama Y, Arita T, Kuriu Y, Nakanishi M, Otsuji E. The Prognostic Value of Preoperative Neutrophil-To-Lymphocyte Ratio in Colorectal Cancer. World J Surg, 2016, 40:2796–802., DOI 10.1007/s00268- 016-3595-x Pine JK,Morris E, Hutchins GG, West NP, Jayne DG, Quirke P, et al. Systemic Neutrophil-To-Lymphocyte Ratio in Colorectal Cancer: the Relationship to Patient Survival, Tumour Biology and Local Lymphocytic Response to Tumour. Br J Cancer, 2015, 113:204–11., DOI 10.1038/bjc.2015.87 Xiao B, Peng J, Wang Y, Deng Y, Ou Q, WuX, et al. Prognostic Value of Tumor Infiltrating Lymphocytes Combined with PD-L1 Expression for Patients with Solitary Colorectal Cancer Liver Metastasis. Ann Transl Med, 2020, 8:1221., DOI 10.21037/atm-20-2762a Peng J, Wang Y, Zhang R, Deng Y, Xiao B, Ou Q, et al. Immune Cell Infiltration in the Microenvironment of Liver Oligometastasis from Colorectal Cancer: Intratumoural CD8/CD3 Ratio Is a Valuable Prognostic index for Patients Undergoing Liver Metastasectomy. Cancers, 2019, 11:1922., DOI 10.3390/cancers11121922 Halama N, Spille A, Lerchl T, Brand K, Herpel E, Welte S, et al. Hepatic Metastases of Colorectal Cancer Are rather Homogeneous but Differ from Primary Lesions in Terms of Immune Cell Infiltration. Oncoimmunology, 2013, 2:e24116., DOI 10.4161/onci.24116 Lin H-C, Shao Q, Liang J-Y, Wang Y, Zhang H-Z, Yuan Y-F, et al. Primary Tumor Immune Score Fails to Predict the Prognosis of Colorectal Cancer Liver Metastases after Hepatectomy in Chinese Populations. Ann Transl Med, 2021, 9:310., DOI 10.21037/atm-20-4932 Ahtiainen M, Elomaa H, Vayrynen JP, Wirta E-V, Kuopio T, Helminen O, et al. Immune Contexture of MMR-Proficient Primary Colorectal Cancer and Matched Liver and Lung Metastases. Cancers, 2021, 13:1530., DOI 10.3390/ cancers13071530 Wang K, Xu D, Yan X-L, Poston G, Xing B-C. The Impact of Primary Tumour Location in Patients Undergoing Hepatic Resection for Colorectal Liver Metastasis. Eur J Surg Oncol, 2018, 44:771–7., DOI 10.1016/j.ejso.2018.02.210 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610315 EP 1610328 DI 10.3389/pore.2022.1610315 PG 14 ER PT J AU Sipos, F Bohusne Barta, B Simon, Nagy, L Danko, T Raffay, ER Petovari, G Zsiros, V Wichmann, B Sebestyen, A Muzes, Gy AF Sipos, Ferenc Bohusne Barta, Bettina Simon, Agnes Nagy, Lorinc Danko, Titanilla Raffay, Eszter Regina Petovari, Gabor Zsiros, Viktoria Wichmann, Barnabas Sebestyen, Anna Muzes, Gyorgyi TI Survival of HT29 Cancer Cells Is Affected by IGF1R Inhibition via Modulation of Self-DNA-Triggered TLR9 Signaling and the Autophagy Response SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE autophagy; CD133; IGF1R; TLR9; HT29 cancer cell; self-DNA ID autophagy; CD133; IGF1R; TLR9; HT29 cancer cell; self-DNA AB Purpose: In HT29 colon cancer cells, a close interplay between self-DNA-induced TLR9 signaling and autophagy response was found, with remarkable effects on cell survival and differentiation. IGF1R activation drives the development and malignant progression of colorectal cancer. IGF1R inhibition displays a controversial effect on autophagy. The interrelated roles of IGF1R inhibition and TLR9/autophagy signaling in HT29 cancer cells have not yet been clarified. In our study, we aimed to investigate the complex interplay of IGF1R inhibition and TLR9/autophagy signaling in HT29 cells. Methods: HT29 cells were incubated with tumor-originated self-DNA with or without inhibitors of IGF1R (picropodophyllin), autophagy (chloroquine), and TLR9 (ODN2088), respectively. Cell proliferation and metabolic activity measurements, direct cell counting, NanoString and Taqman gene expression analyses, immunocytochemistry, WES Simple Western blot, and transmission electron microscopy investigations were performed. Results: The concomitant use of tumor-derived self-DNA and IGF1R inhibitors displays anti-proliferative potential, which can be reversed by parallel TLR9 signaling inhibition. The distinct effects of picropodophyllin, ODN2088, and chloroquine per se or in combination on HT29 cell proliferation and autophagy suggest that either the IGF1R-associated or nonassociated autophagy machinery is “Janus-faced” regarding its actions on cell proliferation. Autophagy, induced by different combinations of self-DNA and inhibitors is not sufficient to rescue HT29 cells from death but results in the survival of some CD133- positive stem-like HT29 cells. Conclusion: The creation of new types of combined IGF1R, autophagy, and/or TLR9 signaling inhibitors would play a significant role in the development of more personalized anti-tumor therapies for colorectal cancer. C1 [Sipos, Ferenc] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Bohusne Barta, Bettina] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Simon, Agnes] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Nagy, Lorinc] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Danko, Titanilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Raffay, Eszter Regina] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Petovari, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Zsiros, Viktoria] Semmelweis University, 1st Department of AnatomyBudapest, Hungary. [Wichmann, Barnabas] Lufthansa Systems HungariaBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Muzes, Gyorgyi] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. RP Sipos, F (reprint author), Semmelweis University, Department of Internal Medicine and Haematology, Budapest, Hungary. EM sipos.ferenc@med.semmelweis-univ.hu CR Girnita L, Worrall C, Takahashi S-I, Seregard S, Girnita A. Something Old, Something New and Something Borrowed: Emerging Paradigm of Insulin-like Growth Factor Type 1 Receptor, IGF-1R, Signaling Regulation. Cell. Mol. Life Sci., 2014, 71(13):2403–27., DOI 10.1007/s00018-013-1514-y Chen P-C, Kuo Y-C, Chuong C-M, Huang Y-H. Niche Modulation of IGF-1R Signaling: Its Role in Stem Cell Pluripotency, Cancer Reprogramming, and Therapeutic Applications. Front Cel Dev. Biol., 2021, 8:625943., DOI 10.3389/ fcell.2020.625943 Wit JM, Walenkamp MJ. Role of Insulin-like Growth Factors in Growth, Development and Feeding. World Rev Nutr Diet, 2013, 106:60–5., DOI 10.1159/ 000342546 Sell C, Rubini M, Rubin R, Liu JP, Efstratiadis A, Baserga R. Simian Virus 40 Large Tumor Antigen Is Unable to Transform Mouse Embryonic Fibroblasts Lacking Type 1 Insulin-like Growth Factor Receptor. Proc Natl Acad Sci U.S.A, 1993, 90(23):11217–21., DOI 10.1073/pnas.90.23.11217 Sell C, Dumenil G, Deveaud C, Miura M, Coppola D, DeAngelis T, et al. Effect of a Null Mutation of the Insulin-like Growth Factor I Receptor Gene on Growth and Transformation of Mouse Embryo Fibroblasts. Mol Cel Biol, 1994, 14(6):3604–12., DOI 10.1128/mcb.14.6.3604-3612.1994 Weber MM, Fottner C, Liu SB, Jung MC, Engelhardt D, Baretton GB. Overexpression of the Insulin-like Growth Factor I Receptor in Human colon Carcinomas. Cancer, 2002, 95(10):2086–95., DOI 10.1002/cncr.10945 Hakam A, Yeatman TJ, Lu L, Mora L, Marcet G, Nicosia SV, et al. Expression of Insulin-like Growth Factor-1 Receptor in Human Colorectal Cancer. Hum Pathol, 1999, 30(10):1128–33., DOI 10.1016/s0046-8177(99)90027-8 Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Shringarpure R, Akiyama M, et al. Inhibition of the Insulin-like Growth Factor Receptor-1 Tyrosine Kinase Activity as a Therapeutic Strategy for Multiple Myeloma, Other Hematologic Malignancies, and Solid Tumors. Cancer Cell, 2004, 5(3): 221–30., DOI 10.1016/s1535-6108(04)00050-9 White E. The Role for Autophagy in Cancer. J Clin Invest, 2015, 125:42–6., DOI 10.1172/JCI73941 Lin X, Peng Z, Wang X, Zou J, Chen D, Chen Z, et al. Targeting Autophagy Potentiates Antitumor Activity of Met-TKIs against Met-Amplified Gastric Cancer. Cell Death Dis, 2019, 10:139., DOI 10.1038/s41419-019-1314-x Sipos F, Szekely H, Kis ID, Tulassay Z, Muzes G. Relation of the IGF/IGF1R System to Autophagy in Colitis and Colorectal Cancer. World J Gastroenterol, 2017, 23(46):8109–19., DOI 10.3748/wjg.v23.i46.8109 Wu Q, Tian A-L, Kroemer G, Kepp O. Autophagy Induction by IGF1R Inhibition with Picropodophyllin and Linsitinib. Autophagy, 2021, 17:2046–7., DOI 10.1080/15548627.2021.1936934 Wu Q, Tian A-L, Li B, Leduc M, Forveille S, Hamley P, et al. IGF1 Receptor Inhibition Amplifies the Effects of Cancer Drugs by Autophagy and Immunedependent Mechanisms. J Immunother Cancer, 2021, 9(6):e002722., DOI 10. 1136/jitc-2021-002722 Renna M, Bento CF, Fleming A, Menzies FM, Siddiqi FH, Ravikumar B, et al. IGF-1 Receptor Antagonism Inhibits Autophagy. Hum Mol Genet, 2013, 22(22):4528–44., DOI 10.1093/hmg/ddt300 Wu W, Ma J, Shao N, Shi Y, Liu R, Li W, et al. Co-Targeting IGF-1R and Autophagy Enhances the Effects of Cell Growth Suppression and Apoptosis Induced by the IGF-1R Inhibitor NVP-AEW541 in Triple-Negative Breast Cancer Cells. PLoS One, 2017, 12(1):e0169229., DOI 10.1371/journal.pone. 9229 16. Mandel P, Metais P. Nuclear Acids in Human Blood Plasma. C R Seances Soc Biol Fil, 1948, 142:241–3. Mansour H. Cell-free Nucleic Acids as Noninvasive Biomarkers for Colorectal Cancer Detection. Front Genet, 2014, 5:182., DOI 10.3389/fgene.2014.00182 Butt AN, Swaminathan R. Overview of Circulating Nucleic Acids in Plasma/ serum. Ann N Y Acad Sci, 2008, 1137:236–42., DOI 10.1196/annals.1448.002 Gormally E, Caboux E, Vineis P, Hainaut P. Circulating Free DNA in Plasma or Serum as Biomarker of Carcinogenesis: Practical Aspects and Biological Significance. Mutat Res Reviews Mutat Res, 2007, 635:105–17., DOI 10.1016/j. mrrev.2006.11.002 Rauh P, Rickes S, Fleischhacker M. Microsatellite Alterations in Free- Circulating Serum DNA in Patients with Ulcerative Colitis. Dig Dis, 2003, 21:363–6., DOI 10.1159/000075361 Gosu V, Basith S, Kwon O-P, Choi S. Therapeutic Applications of Nucleic Acids and Their Analogues in Toll-like Receptor Signaling. Molecules, 2012, 17:13503–29., DOI 10.3390/molecules171113503 Furi I, Sipos F, Spisak S, Kiszner G, Wichmann B, Scholler A, et al. Association of Self-DNA Mediated TLR9-Related Gene, DNA Methyltransferase, and Cytokeratin Protein Expression Alterations in HT29-Cells to DNA Fragment Length and Methylation Status. Scientific World J, 2013, 2013: 1–8., DOI 10.1155/2013/293296 Ge R-T, Mo L-H, Wu R, Liu J-Q, Zhang H-P, Liu Z, et al. Insulin-like Growth Factor-1 Endues Monocytes with Immune Suppressive Ability to Inhibit Inflammation in the Intestine. Sci Rep, 2015, 5:7735., DOI 10.1038/srep07735 Hofmann C, Dunger N, Doser K, Lippert E, Siller S, Edinger M, et al. Physiologic TLR9-CpG-DNA Interaction Is Essential for the Homeostasis of the Intestinal Immune System. Inflamm Bowel Dis, 2014, 20(1):136–43., DOI 10.1097/01.MIB.0000436276.19755.c1 Sipos F, Kiss AL, Constantinovits M, Tulassay Z,Muzes G. Modified Genomic Self-DNA Influences In Vitro Survival of HT29 Tumor Cells via TLR9- and Autophagy Signaling. Pathol Oncol Res, 2019, 25:1505–17., DOI 10.1007/ s12253-018-0544-z Wang C, Ji L, Yuan X, Jin Y, Cardona CJ, Xing Z. Differential Regulation of TLR Signaling on the Induction of Antiviral Interferons in Human Intestinal Epithelial Cells Infected with Enterovirus 71. PLoS One, 2016, 11(3):e0152177., DOI 10.1371/journal.pone.0152177 Li Y, Lu K, Zhao B, Zeng X, Xu S, Ma X, et al. Depletion of Insulin-like Growth Factor 1 Receptor Increases Radiosensitivity in Colorectal Cancer. J Gastrointest Oncol, 2020, 11(6):1135–45., DOI 10.21037/jgo-20-210 Buck E, Gokhale PC, Koujak S, Brown E, Eyzaguirre A, Tao N, et al. Compensatory Insulin Receptor, IR, Activation on Inhibition of Insulinlike Growth Factor-1 Receptor, IGF-1R): Rationale for Cotargeting IGF-1R and IR in Cancer. Mol Cancer Ther, 2010, 9(10):2652–64., DOI 10.1158/1535- 7163.MCT-10-0318 Nowakowska M, Pospiech K, Lewandowska U, Piastowska-Ciesielska AW, Bednarek AK. Diverse Effect of WWOX Overexpression in HT29 and SW480 colon Cancer Cell Lines. Tumor Biol, 2014, 35(9):9291–301., DOI 10.1007/ s13277-014-2196-2 Ogoshi K, Hashimoto S-i., Nakatani Y, Qu W, Oshima K, Tokunaga K, et al. Genome-wide Profiling of DNA Methylation in Human Cancer Cells. Genomics, 2011, 98(4):280–7., DOI 10.1016/j.ygeno.2011.07.003 Moulis M, Vindis C. Methods for Measuring Autophagy in Mice. Cells, 2017, 6(2):14., DOI 10.3390/cells6020014 Rampersad SN. Multiple Applications of Alamar Blue as an Indicator of Metabolic Function and Cellular Health in Cell Viability Bioassays. Sensors, 2012, 12(9):12347–60., DOI 10.3390/s120912347 R Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing, 2019). Available at: https://www.R-project.org/. Bronkhorst AJ, Wentzel JF, Aucamp J, van Dyk E, du Plessis L, Pretorius PJ. Characterization of the Cell-free DNA Released by Cultured Cancer Cells. Biochim Biophys Acta Mol Cel Res, 2016, 1863(1):157–65., DOI 10.1016/j. bbamcr.2015.10.022 Ungerer V, Bronkhorst AJ, Van den Ackerveken P, Herzog M, Holdenrieder S. Serial Profiling of Cell-free DNA and Nucleosome Histone Modifications in Cell Cultures. Sci Rep, 2021, 11(1):9460., DOI 10.1038/s41598-021-88866-5 Souza AGD, Bastos VAF, Fujimura PT, Ferreira ICC, Leal LF, da Silva LS, et al. Cell-free DNA Promotes Malignant Transformation in Non-tumor Cells. Sci Rep, 2020, 10(1):21674., DOI 10.1038/s41598-020-78766-5 Zhou J, Rossi J. Aptamers as Targeted Therapeutics: Current Potential and Challenges. Nat Rev Drug Discov, 2017, 16(3):181–202., DOI 10.1038/nrd. 2016.199 Akhtar M, Watson JL, Nazli A, McKay DM. Bacterial DNA Evokes Epithelial IL-8 Production by a MAPK-dependent, NFκB-independent Pathway. FASEB J., 2003, 17:1319–21., DOI 10.1096/fj.03-0950fje10.1096/fj.02-0950fje Shahriari S, Rezaeifard S, Moghimi HR, Khorramizadeh MR, Faghih Z. Cell Membrane and Intracellular Expression of Toll-like Receptor 9, TLR9, in Colorectal Cancer and Breast Cancer Cell-Lines. Cancer Biomark, 2017, 18: 375–80., DOI 10.3233/cbm-160260 Sun H, Li X, Fan L, Wu G, Li M, Fang J. TRAF6 is Upregulated in colon Cancer and Promotes Proliferation of colon Cancer Cells. Int J Biochem Cel Biol, 2014, 53:195–201., DOI 10.1016/j.biocel.2014.04.010 Marcuello M, Mayol X, Felipe-Fumero E, Costa J, Lopez-Hierro L, Salvans S, et al. Modulation of the colon Cancer Cell Phenotype by Pro-inflammatory Macrophages: A Preclinical Model of Surgery-Associated Inflammation and Tumor Recurrence. PLoS One, 2018, 13:e0192958., DOI 10.1371/journal.pone. 0192958 Ning Y, Manegold PC, Hong YK, Zhang W, Pohl A, Lurje G, et al. Interleukin- 8 is Associated with Proliferation, Migration, Angiogenesis and Chemosensitivity In Vitro and In Vivo in colon Cancer Cell Line Models. Int J Cancer, 2011, 128:2038–49., DOI 10.1002/ijc.25562 Liu H, Li G, Zhang B, Sun D, Wu J, Chen F, et al. Suppression of the NF-κB Signaling Pathway in colon Cancer Cells by the Natural Compound Riccardin D from Dumortierahirsute. Mol Med Rep, 2018, 17:5837–43., DOI 10.3892/ mmr.2018.8617 Li X, Jiang S, Tapping RI. Toll-like Receptor Signaling in Cell Proliferation and Survival. Cytokine, 2010, 49(1):1–9., DOI 10.1016/j. cyto.2009.08.010 Chand HS, Harris JF, Mebratu Y, Chen Y, Wright PS, Randell SH, et al. Intracellular Insulin-like Growth Factor-1 Induces Bcl-2 Expression in Airway Epithelial Cells. J Immunol, 2012, 188(9):4581–9., DOI 10.4049/jimmunol. 1102673 Veleeparambil M, Poddar D, Abdulkhalek S, Kessler PM, Yamashita M, Chattopadhyay S, et al. Constitutively Bound EGFR-Mediated Tyrosine Phosphorylation of TLR9 is Required for its Ability to Signal. J Immunol, 2018, 200(8):2809–18., DOI 10.4049/jimmunol.1700691 Bertin S, Pierrefite-Carle V. Autophagy and Toll-like Receptors: A New Link in Cancer Cells. Autophagy, 2008, 4:1086–9., DOI 10.4161/auto.7138 Colell A, Ricci J-E, Tait S, Milasta S, Maurer U, Bouchier-Hayes L, et al. GAPDH and Autophagy Preserve Survival after Apoptotic Cytochrome C Release in the Absence of Caspase Activation. Cell, 2007, 129:983–97., DOI 10. 1016/j.cell.2007.03.045 Lemasters JJ. Selective Mitochondrial Autophagy, or Mitophagy, as a Targeted Defense against Oxidative Stress, Mitochondrial Dysfunction, and Aging. Rejuvenation Res, 2005, 8:3–5., DOI 10.1089/rej.2005.8.3 Zhao Z-Q, Yu Z-Y, Li J, Ouyang X-N. Gefitinib Induces Lung Cancer Cell Autophagy and Apoptosis via Blockade of the PI3K/AKT/mTOR Pathway. Oncol Lett, 2016, 12(1):63–8., DOI 10.3892/ol.2016.4606 Wang H, Liu Y, Wang D, Xu Y, Dong R, Yang Y, et al. The Upstream Pathway of mTOR-Mediated Autophagy in Liver Diseases. Cells, 2019, 8(12):1597., DOI 10.3390/cells8121597 Le Roith D, Zick Y. Recent Advances in Our Understanding of Insulin Action and Insulin Resistance. Diabetes Care, 2001, 24(3):588–97., DOI 10.2337/ diacare.24.3.588 Cheng Z, Tseng Y, White MF. Insulin Signaling Meets Mitochondria in Metabolism. Trends Endocrinol Metab, 2010, 21(10):589–98., DOI 10.1016/j. tem.2010.06.005 Ning J, Xi G, Clemmons DR. Suppression of AMPK Activation via S485 Phosphorylation by IGF-I during Hyperglycemia is Mediated by AKT Activation in Vascular Smooth Muscle Cells. Endocrinology, 2011, 152(8): 3143–54., DOI 10.1210/en.2011-0155 De Leo MG, Staiano L, Vicinanza M, Luciani A, Carissimo A, Mutarelli M, et al. Autophagosome-lysosome Fusion Triggers a Lysosomal Response Mediated by TLR9 and Controlled by OCRL. Nat Cel Biol, 2016, 18(8): 839–50., DOI 10.1038/ncb3386 Liu L, Han C, Yu H, Zhu W, Cui H, Zheng L, et al. Chloroquine Inhibits Cell Growth in Human A549 Lung Cancer Cells by Blocking Autophagy and Inducing Mitochondrial-Mediated Apoptosis. Oncol Rep, 2018, 39(6): 2807–16., DOI 10.3892/or.2018.6363 NgoM-HT, Jeng H-Y, Kuo Y-C, Diony Nanda J, Brahmadhi A, Ling T-Y, et al. The Role of IGF/IGF-1R Signaling in Hepatocellular Carcinomas: Stemness- Related Properties and Drug Resistance. Int J Mol Sci, 2021, 22(4):1931., DOI 10. 3390/ijms22041931 Wang X-H, Wu H-Y, Gao J, Wang X-H, Gao T-H, Zhang S-F. IGF1R Facilitates Epithelial-Mesenchymal Transition and Cancer Stem Cell Properties in Neuroblastoma via the STAT3/AKT axis. Cancer Manag Res, 2019, 11:5459–72., DOI 10.2147/CMAR.S196862 Singh RK, Dhadve A, Sakpal A, De A, Ray P. An Active IGF-1r-AKT Signaling Imparts Functional Heterogeneity in Ovarian CSC Population. Sci Rep, 2016, 6:36612., DOI 10.1038/srep36612 Xue F, Hu L, Ge R, Yang L, Liu K, Li Y, et al. Autophagy-deficiency in Hepatic Progenitor Cells Leads to the Defects of Stemness and Enhances Susceptibility to Neoplastic Transformation. Cancer Lett, 2016, 371(1):38–47., DOI 10.1016/j. canlet.2015.11.022 Kantara C, O’Connell M, Sarkar S, Moya S, Ullrich R, Singh P. Curcumin Promotes Autophagic Survival of a Subset of colon Cancer Stem Cells, Which are Ablated by DCLK1-siRNA. Cancer Res, 2014, 74(9):2487–98., DOI 10.1158/ 0008-5472.CAN-13-3536 Nazio F, Bordi M, Cianfanelli V, Locatelli F, Cecconi F. Autophagy and Cancer Stem Cells: Molecular Mechanisms and Therapeutic Applications. Cell Death Differ, 2019, 26(4):690–702., DOI 10.1038/s41418-019-0292-y Wollert T, Hurley JH. Molecular Mechanism of Multivesicular Body Biogenesis by ESCRT Complexes. Nature, 2010, 464(7290):864–9., DOI 10. 1038/nature08849 DeRita RM, Zerlanko B, Singh A, Lu H, Iozzo RV, Benovic JL, et al. c-Src, Insulin-like Growth Factor I Receptor, G-Protein-Coupled Receptor Kinases and Focal Adhesion Kinase Are Enriched into Prostate Cancer Cell Exosomes. J Cel Biochem., 2017, 118(1):66–73., DOI 10.1002/jcb.25611 Crudden C, Song D, Cismas S, Trocme E, Pasca S, Calin GA, et al. Below the Surface: IGF-1R Therapeutic Targeting and its Endocytic Journey. Cells, 2019, 8(10):1223., DOI 10.3390/cells8101223 Valcz G, Buzas EI, Kittel A, Krenacs T, Visnovitz T, Spisak S, et al. En Bloc Release of MVB-like Small Extracellular Vesicle Clusters by Colorectal Carcinoma Cells. J Extracellular Vesicles, 2019, 8(1):1596668., DOI 10.1080/ 20013078.2019.1596668 Fader CM, Colombo MI. Autophagy and Multivesicular Bodies: Two Closely Related Partners. Cel Death Differ, 2009, 16:70–8., DOI 10.1038/cdd.2008.168 Berg TO, Fengsrud M, Stromhaug PE, Berg T, Seglen PO. Isolation and Characterization of Rat Liver Amphisomes. Evidence for Fusion of Autophagosomes with Both Early and Late Endosomes. J Biol Chem, 1998, 273:21883–92., DOI 10.1074/jbc.273.34.21883 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610322 EP 1610336 DI 10.3389/pore.2022.1610322 PG 15 ER PT J AU Li, L Song, Q Cao, D Jiao, Y Yuan, G Song, Y AF Li, Lihong Song, Qianqian Cao, Dandan Jiao, Yuchen Yuan, Guangwen Song, Yan TI Whole-Exome Sequencing Could Distinguish Primary Pulmonary Squamous Cell Carcinoma From Lung Metastases in Individuals With Cervical Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE metastasis; cervix; squamous cell carcinoma; sequence; lung ID metastasis; cervix; squamous cell carcinoma; sequence; lung AB Aims: Metastatic cervical carcinoma is hard to cure using traditional treatment and new therapeutic approaches are needed. However, the process of clonal evolution and the molecular alterations that contribute to tumor progression from primary to metastatic carcinoma remain unclear. It is currently difficult to distinguish between the primary pulmonary squamous cell carcinoma (PPSCC) and metastatic cervical squamous cell carcinoma (CSCC). Methods: Paired primary CSCC and lung/lymph nodes metastatic lesions from eight patients were analyzed by whole-exome sequencing (WES). WES data of matched specimens and normal samples were aligned to the human reference genome and analyzed to identify somatic mutations in primary and metastatic lesions. Results: A total of 1,254 somatic variants were identified. All the primary lesions and metastatic lesions shared mutations, the percentage of shared mutations between primary lesions and corresponding metastatic lesions varied significantly, ranging from6%to 70%. In other words, all the metastatic lesions are clonally related to primary lesions, confirming WES could prove they are metastatic from the cervix but not PPSCC. We tried to apply a gene panel to help distinguish PPSCC and metastatic CSCC but failed because the mutations were widely distributed in CSCC. Interestingly, lymph nodes metastasis (LNM) harbored fewer cancer driver mutations than primary CSCC specimens with a significant difference. Besides this, there was no significant difference in somatic mutations and copy number variation (CNV) between primary and metastatic CSCC. Conclusion: Our data demonstrate that WES is an additional helpful tool in distinguishing PPSCC and metastatic CSCC, especially for certain difficult cases. C1 [Li, Lihong] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Song, Qianqian] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, State Key Lab of Molecular Oncology, Laboratory of Cell and Molecular BiologyBeijing, China. [Cao, Dandan] Genetron Health (Beijing) Co. Ltd.Beijing, China. [Jiao, Yuchen] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, State Key Lab of Molecular Oncology, Laboratory of Cell and Molecular BiologyBeijing, China. [Yuan, Guangwen] Chinese Academy of Medvdical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Department of Gynecology OncologyBeijing, China. [Song, Yan] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. RP Song, Y (reprint author), Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of Pathology, Beijing, China. EM songyan@cicams.ac.cn CR Siegel RL,Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA A Cancer J Clin, 2021, 71(1):7–33., DOI 10.3322/caac.21654 Walboomers JMM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, et al. Human Papillomavirus Is a Necessary Cause of Invasive Cervical Cancer Worldwide. J Pathol, 1999, 189(1):12–9., DOI 10.1002/(sici)1096-9896(199909, 189:1<12::aid-path431>3.0.co;2-f Fulcher AS, O’Sullivan SG, Segreti EM, Kavanagh BD. Recurrent Cervical Carcinoma: Typical and Atypical Manifestations. Radiographics, 1999, 19: S103–S116., DOI 10.1148/radiographics.19.suppl_1.g99oc19s103 Ki EY, Lee KH, Park JS, Hur SY A Clinicopathological Review of Pulmonary Metastasis from Uterine Cervical Cancer. Cancer Res Treat, 2016, 48(1):266-72 Chang SY, Keeney M, Law M, Donovan J, Aubry M-C, Garcia J. Detection of Human Papillomavirus in Non-Small Cell Carcinoma of the Lung. Hum Pathol, 2015, 46(11):1592–7., DOI 10.1016/j.humpath. 2015.07.012 Lin S, Zhang X, Li X, Qin C, Zhang L, Lu J, et al. Detection of Human Papillomavirus Distinguishes Second Primary Tumors from Lung Metastases in Patients with Squamous Cell Carcinoma of the Cervix. Thorac Cancer, 2020, 11(8):2297–305., DOI 10.1111/1759-7714.13544 Fan X, Yu K, Wu J, Shao J, Zhu L, Zhang J. Correlation Between Squamous Cell Carcinoma of the Lung and Human Papillomavirus Infection and the Relationship to Expression of P53 and P16. Tumor Biol, 2015, 36(4): 3043–9., DOI 10.1007/s13277-014-2940-7 Weichert W, Schewe C, Denkert C, Morawietz L, Dietel M, Petersen I. Molecular HPV Typing as a Diagnostic Tool to Discriminate Primary from Metastatic Squamous Cell Carcinoma of the Lung. Am J Surg Pathol, 2009, 33(4):513–20., DOI 10.1097/PAS.0b013e3181938319 Crook T, Wrede D, Tidy JA, Vousden KH, Wrede D, Tidy J, et al. Clonal P53 Mutation in Primary Cervical Cancer: Association with Human- Papillomavirus-Negative Tumours. The Lancet, 1992, 339(8801):1070–3., DOI 10.1016/0140-6736(92)90662-m McIntyre JB, Wu JS, Craighead PS, Phan T, Kobel M, Lees-Miller SP, et al. PIK3CA Mutational Status and Overall Survival in Patients with Cervical Cancer Treated with Radical Chemoradiotherapy. Gynecol Oncol, 2013, 128(3):409–14., DOI 10.1016/j.ygyno.2012.12.019 Kang S, Kim H-S, Seo SS, Park S-Y, Sidransky D, Dong SM. Inverse Correlation Between RASSF1A Hypermethylation, KRAS and BRAF Mutations in Cervical Adenocarcinoma. Gynecol Oncol, 2007, 105(3): 662–6., DOI 10.1016/j.ygyno.2007.01.045 Wingo SN, Gallardo TD, Akbay EA, Liang M-C, Contreras CM, Boren T, et al. Somatic LKB1 Mutations Promote Cervical Cancer Progression. PLoS One, 2009, 4(4);e5137., DOI 10.1371/journal.pone.0005137 Cancer Genome Atlas Research N, Albert Einstein College of M. Integrated Genomic and Molecular Characterization of Cervical Cancer. Nature, 2017, 543(7645):378–84., DOI 10.1038/nature21386 Dietlein F, Weghorn D, Taylor-Weiner A, Richters A, Reardon B, Liu D, et al. Identification of Cancer Driver Genes Based on Nucleotide Context. Nat Genet, 2020, 52(2):208–18., DOI 10.1038/s41588-019-0572-y Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative Genomics Viewer. Nat Biotechnol, 2011, 29(1):24–6., DOI 10. 1038/nbt.1754 Talevich E, Shain AH, Botton T, Bastian BC. CNVkit: Genome-Wide Copy Number Detection and Visualization from Targeted DNA Sequencing. Plos Comput Biol, 2016, 12(4);e1004873., DOI 10.1371/journal.pcbi. 1004873 Phylip. Phylogeny Inference Package, Version 3.6. WA: Department of Genome Sciences, University of Washington, Seattle., 2015). Bodelon C, Vinokurova S, Sampson JN, den Boon JA,Walker JL,Horswill MA, et al. Chromosomal Copy Number Alterations and HPV Integration in Cervical Precancer and Invasive Cancer. Carcin, 2016, 37(2):188–96., DOI 10.1093/carcin/bgv171 Yan D, Yi S, Chiu WC, Qin LG, Kin WH, Kwok Hung CT, et al. Integrated Analysis of Chromosome Copy Number Variation and Gene Expression in Cervical Carcinoma. Oncotarget, 2017, 8(65):108912–22., DOI 10.18632/ oncotarget.22403 WangM, Fan W, Ye M, Tian C, Zhao L, Wang J, et al.Molecular Profiles and Tumor Mutational Burden Analysis in Chinese Patients with Gynecologic Cancers. Sci Rep, 2018, 8(1):8990., DOI 10.1038/s41598- 018-25583-6 Tomasetti CC, Marchionni MA, Nowak B, Parmigiani G, Vogelstein B. Only Three Driver Gene Mutations Are Required for the Development of Lung and Colorectal Cancers. Proc Natl Acad Sci U.S.A, 2015, 112(1):118–23., DOI 10. 1073/pnas.1421839112 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610325 EP 1610332 DI 10.3389/pore.2022.1610325 PG 8 ER PT J AU Chen, Z Zhou, H Hu, H Chen, L AF Chen, Zhe Zhou, Hong Hu, Haoliang Chen, Linxi TI Blocking the Metabolic Switch Toward Cytosolic 1C Flux: A Novel Therapeutic Approach for Tumors With Low SLC19A1 Expression SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE One-carbon units; SHMT1; SHMT2; RPMI; TSH; MTHFD1 ID One-carbon units; SHMT1; SHMT2; RPMI; TSH; MTHFD1 C1 [Chen, Zhe] University of South China, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, College of Basic Medical Science, Hengyang Medical School, Institute of Pharmacy and PharmacologyHengyang, China. [Zhou, Hong] The First Affiliated Hospital of University of South China, Radiology DepartmentHengyang, China. [Hu, Haoliang] Hunan University of Arts and Science, College of Life and Environmental Sciences, Changde Research Centre for Artificial Intelligence and BiomedicineChangde, China. [Chen, Linxi] University of South China, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, College of Basic Medical Science, Hengyang Medical School, Institute of Pharmacy and PharmacologyHengyang, China. RP Hu, H (reprint author), Hunan University of Arts and Science, College of Life and Environmental Sciences, Changde Research Centre for Artificial Intelligence and Biomedicine, Changde, China. EM haolianghu6@126.com CR Ducker GS, Chen L, Morscher RJ, Ghergurovich JM, EspositoM, Teng X, et al. Reversal of Cytosolic One-Carbon Flux Compensates for Loss of the Mitochondrial Folate Pathway. Cell Metab, 2016, 24(4):640–1., DOI 10.1016/ j.cmet.2016.09.011 Lee WD, Pirona AC, Sarvin B, Stern A, Nevo-Dinur K, Besser E, et al. Tumor Reliance on Cytosolic versus Mitochondrial One-Carbon Flux Depends on Folate Availability. Cell Metab, 2020, 33:190–8., DOI 10.1016/j.cmet.2020. 12.002 Paone A, Marani M, Fiascarelli A, Rinaldo S, Giardina G, Contestabile R, et al. SHMT1 Knockdown Induces Apoptosis in Lung Cancer Cells by Causing Uracil Misincorporation. Cell Death Dis, 2014, 5:e1525., DOI 10.1038/cddis. 2014.482 Vlashi E, Lagadec C, Vergnes L, Matsutani T, Masui K, Poulou M, et al. Metabolic State of Glioma Stem Cells and Nontumorigenic Cells. Proc Natl Acad Sci U.S.A, 2011, 108(38):16062–7., DOI 10.1073/pnas.1106704108 Marani M, Paone A, Fiascarelli A, Macone A, Gargano M, Rinaldo S, et al. A Pyrazolopyran Derivative Preferentially Inhibits the Activity of Human Cytosolic Serine Hydroxymethyltransferase and Induces Cell Death in Lung Cancer Cells. Oncotarget, 2016, 7(4):4570–83., DOI 10.18632/oncotarget.6726 Perry C, Sastry R, Nasrallah IM, Stover PJ. Mimosine Attenuates Serine Hydroxymethyltransferase Transcription by Chelating Zinc. J Biol Chem, 2005, 280(1):396–400., DOI 10.1074/jbc.m410467200 Kamynina E, Lachenauer ER, DiRisio AC, Liebenthal RP, Field MS, Stover PJ. Arsenic Trioxide Targets MTHFD1 and SUMO-dependent Nuclear De Novo Thymidylate Biosynthesis. Proc Natl Acad Sci U S A, 2017, 114(12): E2319–E2326., DOI 10.1073/pnas.1619745114 Schwertz G, Witschel MC, Rottmann M, Leartsakulpanich U, Chitnumsub P, Jaruwat A, et al. Potent Inhibitors ofPlasmodialSerine Hydroxymethyltransferase, SHMT, Featuring a Spirocyclic Scaffold. ChemMedChem, 2018, 13(9):931–43., DOI 10.1002/cmdc.201800053 Tramonti A, Nardella C, di Salvo ML, Barile A, Cutruzzola F, Contestabile R. Human Cytosolic and Mitochondrial Serine Hydroxymethyltransferase Isoforms in Comparison: Full Kinetic Characterization and Substrate Inhibition Properties. Biochemistry, 2018, 57(51):6984–96., DOI 10.1021/acs. biochem.8b01074 Paiardini A, TramontiSchirch A, Schirch D, Guiducci G, di Salvo ML, Fiascarelli A, et al. Differential 3-bromopyruvate Inhibition of Cytosolic and Mitochondrial Human Serine Hydroxymethyltransferase Isoforms, Key Enzymes in Cancer Metabolic Reprogramming. Biochim Biophys Acta, Bba, - Proteins Proteomics, 2016, 1864(11):1506–17., DOI 10.1016/j.bbapap.2016. 08.010 Dekhne AS, Shah K, Ducker GS, Katinas JM, Wong-Roushar J, Nayeen MJ, et al. Novel Pyrrolo[3,2-D]pyrimidine Compounds Target Mitochondrial and Cytosolic One-Carbon Metabolism with Broad-Spectrum Antitumor Efficacy. Mol Cancer Ther, 2019, 18(10):1787–99., DOI 10.1158/1535-7163.mct-19-0037 Scaletti E, Jemth AS, Helleday T, Stenmark P. Structural Basis of Inhibition of the Human Serine Hydroxymethyltransferase SHMT 2 by Antifolate Drugs. FEBS Lett, 2019, 593(14):1863–73., DOI 10.1002/1873-3468.13455 Wu S, Zhang G, Li P, Chen S, Zhang F, Li J, et al. miR-198 Targets SHMT1 to Inhibit Cell Proliferation and Enhance Cell Apoptosis in Lung Adenocarcinoma. Tumor Biol, 2016, 37(4):5193–202., DOI 10.1007/s13277- 015-4369-z Yang Q, Li J, Hu Y, Tang X, Yu L, Dong L, et al. MiR-218-5p Suppresses the Killing Effect of Natural Killer Cell to Lung Adenocarcinoma by Targeting SHMT1. Yonsei Med J, 2019, 60(6):500–8., DOI 10.3349/ymj.2019.60.6.500 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610337 EP 1610341 DI 10.3389/pore.2022.1610337 PG 5 ER PT J AU Nagy, Zs Ferenczi, K Istenes, I Eid, H Bodor, Cs Timar, B Demeter, J AF Nagy, F. Zsofia Ferenczi, Kata Istenes, Ildiko Eid, Hanna Bodor, Csaba Timar, Botond Demeter, Judit TI Case Report: Development of Diffuse Large B Cell Lymphoma a Long Time After Hairy Cell Leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE hairy cell leukaemia; non-Hodgkin lymphoma; diffuse large B cell lymphoma; transformation; case report ID hairy cell leukaemia; non-Hodgkin lymphoma; diffuse large B cell lymphoma; transformation; case report AB Hairy cell leukaemia (HCL) is a rare B cell malignancy with an indolent course leading to pancytopaenia due to bone marrow infiltration. It has been proposed that HCL patients are at risk of developing a secondary malignancy, with a marked likelihood of the development of other hematologic malignancies including Hodgkin lymphoma and high-grade non- Hodgkin lymphomas. Here, we present the case of two patients who developed diffuse large B cell lymphoma after a long course of hairy cell leukaemia. In the case of the female patient, we report on the occurrence of a third malignant disease, which is very uncommon. With our case descriptions we contribute to the very small number of similar cases reported. C1 [Nagy, F. Zsofia] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Ferenczi, Kata] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Istenes, Ildiko] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Eid, Hanna] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Bodor, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, HCEMM-SE Molecular Oncohematology Research GroupBudapest, Hungary. [Timar, Botond] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, HCEMM-SE Molecular Oncohematology Research GroupBudapest, Hungary. [Demeter, Judit] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. RP Timar, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, HCEMM-SE Molecular Oncohematology Research Group, Budapest, Hungary. EM timar.botond@med.semmelweis-univ.hu CR Cross M, Dearden C. Hairy Cell Leukaemia. Curr Oncol Rep, 2020, 22:42., DOI 10.1007/s11912-020-00911-0 Grever MR, Abdel-Wahab O, Andritsos LA, Banerji V, Barrientos J, Blachly JS, et al. Consensus Guidelines for the Diagnosis and Management of Patients with Classic Hairy Cell Leukemia. Blood, 2017, 129:553–60., DOI 10.1182/ blood-2016-01-689422 Else M, Ruchlemer R, Osuji N, Del Giudice I, Matutes E, Woodman A, et al. Long Remissions in Hairy Cell Leukemia with Purine Analogs. Cancer, 2005, 104:2442–8., DOI 10.1002/cncr.21447 Tiacci E, Trifonov V, Schiavoni G, Holmes A, Kern W, Martelli MP, et al. BRAF Mutations in Hairy-Cell Leukemia. N Engl J Med, 2011, 364:2305–15., DOI 10.1056/NEJMoa1014209.BRAF Chihara D, Kreitman RJ. Treatment of Hairy Cell Leukemia. Expert Rev Hematol, 2020, 13:1107–17., DOI 10.1080/17474086.2020.1819231 Tiacci E, De Carolis L, Simonetti E, Capponi M, Ambrosetti A, Lucia E, et al. Vemurafenib Plus Rituximab in Refractory or Relapsed Hairy-Cell Leukemia. N Engl J Med, 2021, 384:1810–23., DOI 10.1056/nejmoa2031298 Paillassa J, Cornet E,Noel S, TomowiakC, Lepretre S, Vaudaux S, et al.Analysis of a Cohort of 279 Patients with Hairy-Cell Leukemia, HCL): 10 Years of Follow-Up. Blood Cancer J, 2020, 10:62., DOI 10.1038/s41408-020-0328-z ZhengG, Chattopadhyay S, SudA, SundquistK, Sundquist J, Forsti A, et al.Types of Second Primary Cancers Influence Survival in Chronic Lymphocytic and Hairy Cell Leukemia Patients. Blood Cancer J, 2019, 9:0–3., DOI 10.1038/s41408-019-0201-0 Sun T, Grupka N, Klein C. Transformation of Hairy Cell Leukemia to High- Grade Lymphoma: A Case Report and Review of the Literature. Hum Pathol, 2004, 35:1423–6., DOI 10.1016/j.humpath.2004.08.016 Zinzani PL, Pellegrini C, Gandolfi L, Stefoni V, Quirini F, Derenzini E, et al. Combination of Lenalidomide and Rituximab in Elderly Patients with Relapsed or Refractory Diffuse Large B-Cell Lymphoma: A Phase 2 Trial. Clin Lymphoma Myeloma Leuk, 2011, 11:462–6., DOI 10.1016/j.clml.2011.02.001 Hisada M, Chen BE, Jaffe ES, Travis LB. Second Cancer Incidence and CausespecificMortalityAmong 3104 PatientswithHairy Cell Leukemia:APopulation- Based Study. J Natl Cancer Inst, 2007, 99:215–22., DOI 10.1093/jnci/djk030 Rosenberg JD, Burian C, Waalen J, Saven A. Clinical Characteristics and Long- Term Outcome of Young Hairy Cell Leukemia Patients Treated with Cladribine: A Single-Institution Series. Blood, 2014, 123:177–83., DOI 10. 1182/blood-2013-06-508754 Balasubramanian R, Rolph R, Morgan C, Hamed H. Genetics of Breast Cancer: Management Strategies and Risk-Reducing Surgery. Br J Hosp Med, 2019, 80: 720–5., DOI 10.12968/hmed.2019.80.12.720 Cheson BD, Vena DA, Barrett J, Freidlin B. Second Malignancies as a Consequence of Nucleoside Analog Therapy for Chronic Lymphoid Leukemias. Jco, 1999, 17:2454., DOI 10.1200/jco.1999.17.8.2454 Pawson R, A’hern R, Catovsky D. SecondMalignancy in Hairy Cell Leukaemia: No Evidence of Increased Incidence after Treatment with Interferon Alpha. Leuk Lymphoma, 1996, 22:103–6., DOI 10.3109/10428199609051735 Troussard X,Henry-AmarM, Flandrin G. Second CancerRisk after InterferonTherapy? [letter; Comment]. Blood, 1994, 84:3242–4., DOI 10.1182/blood.v84.9.3242.3242 Cornet E, Tomowiak C, Tanguy-Schmidt A, Lepretre S, Dupuis J, Feugier P, et al. Long-term Follow-Up and Second Malignancies in 487 Patients with Hairy Cell Leukaemia. Br J Haematol, 2014, 166:390–400., DOI 10.1111/bjh.12908 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610338 EP 1610342 DI 10.3389/pore.2022.1610338 PG 5 ER PT J AU Ujfaludi, Zs Kuthi, L Pankotai-Bodo, G Banko, S Sukosd, F Pankotai, T AF Ujfaludi, Zsuzsanna Kuthi, Levente Pankotai-Bodo, Gabriella Banko, Sarolta Sukosd, Farkas Pankotai, Tibor TI Novel Diagnostic Value of Driver Gene Transcription Signatures to Characterise Clear Cell Renal Cell Carcinoma, ccRCC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE ccRCC; gene expression signature; tumour profiling; driver gene; statistical analyses ID ccRCC; gene expression signature; tumour profiling; driver gene; statistical analyses AB Routine molecular tumour diagnostics are augmented by DNA-based qualitative and quantitative molecular techniques detecting mutations of DNA. However, in the past decade, it has been unravelled that the phenotype of cancer, as it’s an extremely complex disease, cannot be fully described and explained by single or multiple genetic variants affecting only the coding regions of the genes. Moreover, studying the manifestation of these somatic mutations and the altered transcription programming—driven by genomic rearrangements, dysregulation of DNA methylation and epigenetic landscape—standing behind the tumorigenesis and detecting these changes could provide a more detailed characterisation of the tumour phenotype. Consequently, novel comparative cancer diagnostic pipelines, including DNA- and RNA-based approaches, are needed for a global assessment of cancer patients. Here we report, that by monitoring the expression patterns of key tumour driver genes by qPCR, the normal and the tumorous samples can be separated into distinct categories. Furthermore, we also prove that by examining the transcription signatures of frequently affected genes at 3p25, 3p21 and 9p21.3 genomic regions, the ccRCC (clear cell renal cell carcinoma) and non-tumorous kidney tissues can be distinguished based on the mRNA level of the selected genes. Our results open new diagnostics possibilities where the mRNA signatures of tumour drivers can supplement the DNA-based approaches providing a more precise diagnostics opportunity leading to determine more precise therapeutic protocols. C1 [Ujfaludi, Zsuzsanna] University of Szeged, Department of PathologySzeged, Hungary. [Kuthi, Levente] University of Szeged, Department of PathologySzeged, Hungary. [Pankotai-Bodo, Gabriella] University of Szeged, Department of PathologySzeged, Hungary. [Banko, Sarolta] University of Szeged, Department of PathologySzeged, Hungary. [Sukosd, Farkas] University of Szeged, Department of PathologySzeged, Hungary. [Pankotai, Tibor] University of Szeged, Department of PathologySzeged, Hungary. RP Pankotai, T (reprint author), University of Szeged, Department of Pathology, Szeged, Hungary. EM pankotai.tibor@szte.hu CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM The 2016 WHO Classification of Tumours of the Urinary System and Male Genital Organs-Part A: Renal, Penile, and Testicular Tumours. Eur Urol, 2016, 70(1):93–105., DOI 10.1016/j.eururo.2016. 02.029 Deng F-M, Melamed J Histologic Variants of Renal Cell Carcinoma: Does Tumor Type Influence Outcome? Urol Clin North America, 2012, 39(2): 119–32., DOI 10.1016/j.ucl.2012.02.001 Gnarra JR, Tory K, Weng Y, Schmidt L, Wei MH, Li H, et al. Mutations of the VHL Tumour Suppressor Gene in Renal Carcinoma. Nat Genet, 1994, 7(1): 85–90., DOI 10.1038/ng0594-85 Brugarolas J Molecular Genetics of clear-cell Renal Cell Carcinoma. J Clin Oncol, 2014, 32(18):1968–76., DOI 10.1200/jco.2012.45.2003 Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, et al. Genomic Architecture and Evolution of clear Cell Renal Cell Carcinomas Defined by Multiregion Sequencing. Nat Genet, 2014, 46(3):225–33., DOI 10. 1038/ng.2891 Linehan WM, Ricketts CJ The Cancer Genome Atlas of Renal Cell Carcinoma: Findings and Clinical Implications. Nat Rev Urol, 2019, 16(9):539–52., DOI 10. 1038/s41585-019-0211-5 Turajlic S, Xu H, Litchfield K, Rowan A, Horswell S, Chambers T, et al. Deterministic Evolutionary Trajectories Influence Primary Tumor Growth: TRACERx Renal. Cell, 2018, 173(3):595–610., DOI 10.1016/j. cell.2018.03.043 Turajlic S, Xu H, Litchfield K, Rowan A, Chambers T, Lopez JI, et al. Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal. Cell, 2018, 173(3):581–94., DOI 10.1016/j.cell.2018. 03.057 Ren Y, Xiao L, Weng G, Shi B Clinical Significance of p16INK4A and p14ARF Promoter Methylation in Renal Cell Carcinoma: a Meta-Analysis. Oncotarget, 2017, 8(38):64385–94., DOI 10.18632/oncotarget.18826 Easton DF, Pharoah PDP, Antoniou AC, Tischkowitz M, Tavtigian SV, Nathanson KL, et al. Gene-panel Sequencing and the Prediction of Breast- Cancer Risk. N Engl J Med, 2015, 372(23):2243–57., DOI 10.1056/ nejmsr1501341 Rainville IR, Rana HQ Next-generation Sequencing for Inherited Breast Cancer Risk: Counseling through the Complexity. Curr Oncol Rep, 2014, 16(3):371., DOI 10.1007/s11912-013-0371-z LaDuca H, Stuenkel AJ, Dolinsky JS, Keiles S, Tandy S, Pesaran T, et al. Utilization of Multigene Panels in Hereditary Cancer Predisposition Testing: Analysis of More Than 2,000 Patients. Genet Med, 2014, 16(11):830–7., DOI 10. 1038/gim.2014.40 Susswein LR, Marshall ML, Nusbaum R, Vogel Postula KJ,Weissman SM, Yackowski L, et al. Pathogenic and Likely Pathogenic Variant Prevalence Among the First 10,000 Patients Referred for Next-Generation Cancer Panel Testing. Genet Med, 2016, 18(8):823–32., DOI 10.1038/ gim.2015.166 Kamps R, Brandao RD, Bosch BJ, Paulussen AD, Xanthoulea S, Blok MJ, et al. Next-Generation Sequencing in Oncology: Genetic Diagnosis, Risk Prediction and Cancer Classification. Int J Mol Sci, 2017, 18(2):308., DOI 10.3390/ ijms18020308 Johnson MR, Wang K, Smith JB, Heslin MJ, Diasio RB Quantitation of Dihydropyrimidine Dehydrogenase Expression by Real-Time Reverse Transcription Polymerase Chain Reaction. Anal Biochem, 2000, 278(2): 175–84., DOI 10.1006/abio.1999.4461 Ricketts CJ, De Cubas AA, Fan H, Smith CC, Lang M, Reznik E, et al. The Cancer Genome Atlas Comprehensive Molecular Characterization of Renal Cell Carcinoma. Cell Rep, 2018, 23(1):3698–326 e315., DOI 10.1016/j.celrep. 2018.06.032 Herman JG, Latif F, Weng Y, Lerman MI, Zbar B, Liu S, et al. Silencing of the VHL Tumor-Suppressor Gene by DNA Methylation in Renal Carcinoma. Proc Natl Acad Sci U.S.A, 1994, 91(21):9700–4., DOI 10. 1073/pnas.91.21.9700 Sun X-J, Wei J, Wu X-Y, Hu M, Wang L, Wang H-H, et al. Identification and Characterization of a Novel Human Histone H3 Lysine 36-specific Methyltransferase. J Biol Chem, 2005, 280(42):35261–71., DOI 10.1074/jbc. m504012200 Bihr S, Ohashi R, Moore AL, Ruschoff JH, Beisel C, Hermanns T, et al. Expression and Mutation Patterns of PBRM1, BAP1 and SETD2 Mirror Specific Evolutionary Subtypes in Clear Cell Renal Cell Carcinoma. Neoplasia, 2019, 21(2):247–56., DOI 10.1016/j.neo.2018.12.006 Xi X, Li T, Huang Y, Sun J, Zhu Y, Yang Y, et al. RNA Biomarkers: Frontier of Precision Medicine for Cancer. Noncoding RNA, 2017, 3(1):9., DOI 10.3390/ ncrna3010009 Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. MicroRNA Expression Profiles Classify Human Cancers. Nature, 2005, 435(7043):834–8., DOI 10.1038/nature03702 Cuzick J, Swanson GP, Fisher G, Brothman AR, Berney DM, Reid JE, et al. Prognostic Value of an RNA Expression Signature Derived from Cell Cycle Proliferation Genes in Patients with Prostate Cancer: a Retrospective Study. Lancet Oncol, 2011, 12(3):245–55., DOI 10.1016/s1470-2045(10, 70295-3 Parker JS, Mullins M, Cheang MCU, Leung S, Voduc D, Vickery T, et al. Supervised Risk Predictor of Breast Cancer Based on Intrinsic Subtypes. J Clin Oncol, 2009, 27(8):1160–7., DOI 10.1200/jco.2008.18.1370 Chen Y-T, Tu JJ, Kao J, Zhou XK, Mazumdar M Messenger RNA Expression Ratios Among Four Genes Predict Subtypes of Renal Cell Carcinoma and Distinguish Oncocytoma from Carcinoma. Clin Cancer Res, 2005, 11(18): 6558–66., DOI 10.1158/1078-0432.ccr-05-0647 Youssef YM, White NMA, Grigull J, Krizova A, Samy C, Mejia-Guerrero S, et al. Accurate Molecular Classification of Kidney Cancer Subtypes Using microRNA Signature. Eur Urol, 2011, 59(5):721–30., DOI 10.1016/j.eururo. 2011.01.004 Wang Q, Gan H, Chen C, Sun Y, Chen J, Xu M, et al. Identification and Validation of a 44-gene Expression Signature for the Classification of Renal Cell Carcinomas. J Exp Clin Cancer Res, 2017, 36(1):176., DOI 10.1186/s13046- 017-0651-9 Borys AM, Seweryn M, Golabek T, Belch L, Klimkowska A, Toton-Zuranska J, et al. Patterns of Gene Expression Characterize T1 and T3 clear Cell Renal Cell Carcinoma Subtypes. PLoS One, 2019, 14(5):e0216793., DOI 10.1371/journal. pone.0216793 Chen L, Yuan L, Qian K, Qian G, Zhu Y, Wu C-L, et al. Identification of Biomarkers Associated with Pathological Stage and Prognosis of Clear Cell Renal Cell Carcinoma by Co-expression Network Analysis. Front Physiol, 2018, 9:399., DOI 10.3389/fphys.2018.00399 Wang Y, Chen L, Wang G, Cheng S, Qian K, Liu X, et al. Fifteen Hub Genes Associated with Progression and Prognosis of clear Cell Renal Cell Carcinoma Identified by Coexpression Analysis. J Cell Physiol, 2019, 234(7):10225–37., DOI 10.1002/jcp.27692 Chen L, Luo Y, Wang G, Qian K, Qian G, Wu CL, et al. Prognostic Value of a Gene Signature in clear Cell Renal Cell Carcinoma. J Cell Physiol, 2019, 234(7): 10324–35., DOI 10.1002/jcp.27700 Aran D, Camarda R, Odegaard J, Paik H, Oskotsky B, Krings G, et al. Comprehensive Analysis of normal Adjacent to Tumor Transcriptomes. Nat Commun, 2017, 8(1):1077., DOI 10.1038/s41467-017-01027-z NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610345 EP 1610357 DI 10.3389/pore.2022.1610345 PG 13 ER PT J AU Matrai, Z Kelemen, P Kosa, Cs Maraz, R Paszt, A Pavlovics, G Savolt, Simonka, Zs Toth, D Kasler, M Vicko, F Pluta, P Kolacinska-Wow, A Murawa, D Jankau, J Ciesla, S Dyttert, D Sabol, M Zhygulin, A Avetisyan, A Lazar, Gy AF Matrai, Zoltan Kelemen, Peter Kosa, Csaba Maraz, Robert Paszt, Attila Pavlovics, Gabor Savolt, Akos Simonka, Zsolt Toth, Dezso Kasler, Miklos Vicko, Ferenc Pluta, Piotr Kolacinska-Wow, Agnieszka Murawa, Dawid Jankau, Jerzy Ciesla, Slawomir Dyttert, Daniel Sabol, Martin Zhygulin, Andrii Avetisyan, Artur Lazar, Gyorgy TI Modern Breast Cancer Surgery 1st Central-Eastern European Professional Consensus Statement on Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Guideline DE breast cancer; surgery; consensus statement; oncoplastic surgery; oncology ID breast cancer; surgery; consensus statement; oncoplastic surgery; oncology AB This text is based on the recommendations accepted by the 4th Hungarian Consensus Conference on Breast Cancer, modified on the basis of the international consultation and conference within the frames of the Central-Eastern European Academy of Oncology. The recommendations cover non-operative, intraoperative and postoperative diagnostics, determination of prognostic and predictive markers and the content of cytology and histology reports. Furthermore, they address some specific issues such as the current status of multigene molecular markers, the role of pathologists in clinical trials and prerequisites for their involvement, and some remarks about the future. C1 [Matrai, Zoltan] National Institute of Oncology, Department of SurgeryBudapest, Hungary. [Kelemen, Peter] National Institute of Oncology, Department of SurgeryBudapest, Hungary. [Kosa, Csaba] University of Debrecen, Department of Surgery and Operative TechniquesDebrecen, Hungary. [Maraz, Robert] Bacs-Kiskun County HospitalKecskemet, Hungary. [Paszt, Attila] University of Szeged, Department of SurgerySzeged, Hungary. [Pavlovics, Gabor] University of Pecs, Department of SurgeryPecs, Hungary. [Savolt, Akos] National Institute of Oncology, Department of SurgeryBudapest, Hungary. [Simonka, Zsolt] University of Szeged, Department of SurgerySzeged, Hungary. [Toth, Dezso] University of Debrecen, Department of Surgery and Operative TechniquesDebrecen, Hungary. [Kasler, Miklos] Government of Hungary, Minister of Human CapacitiesBudapest, Hungary. [Vicko, Ferenc] University of Novi Sad, Medical Faculty Novi Sad, Oncology Institute of Vojvodina Sremska KamenicaNovi Sad, Serbia. [Pluta, Piotr] Polish Mother’s Memorial Hospital - Research Institute, Department of Oncological Surgery and Breast DiseasesLodz, Poland. [Kolacinska-Wow, Agnieszka] Medical University of Lodz, department of Head and Neck Cancer SurgeryLodz, Poland. [Murawa, Dawid] Poznan University of Medical Sciences, Department of Oncology, Department of Oncological SurgeryPoznan, Poland. [Jankau, Jerzy] Medical University of Gdansk, University Hospitals, Plastic Surgery DepartmentGdansk, Poland. [Ciesla, Slawomir] Karol Marcinkowski University Hospital, General and Oncological Surgery ClinicZielona Gora, Poland. [Dyttert, Daniel] Comenius University, Medical Faculty, St. Elisabeth Cancer Institute, Department of Surgical OncologyBratislava, Slovakia. [Sabol, Martin] Comenius University, Medical Faculty, St. Elisabeth Cancer Institute, Department of Surgical OncologyBratislava, Slovakia. [Zhygulin, Andrii] LISOD Hospital of Israeli OncologyKyiv, Ukraine. [Avetisyan, Artur] National Center of OncologyYerevan, Armenia. [Lazar, Gyorgy] University of Szeged, Department of SurgerySzeged, Hungary. RP Matrai, Z (reprint author), National Institute of Oncology, Department of Surgery, Budapest, Hungary. EM matraidoc@gmail.com CR Lazar G, Kelemen P, Kosa C, Maraz R, Paszt A, Pavlovics G, et al. Modern Surgical Treatment of Breast Cancer. 4th Breast Cancer Consensus Conference. Magy Onkol, 2020, 64(4):329–46. Coates AS,Winer EP, Goldhirsch A, Gelber RD, Gnant M, Piccart-Gebhart M, et al. Tailoring Therapies-Iimproving the Management of Early Breast Cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015. Ann Oncol, 2015, 26:1533–46., DOI 10.1093/annonc/ mdv221 Senkus E, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rutgers E, et al. Primary Breast Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol, 2015, 26(Suppl. 5):v8–v30., DOI 10. 1093/annonc/mdv298 National Comprehensive Cancer Network, NCCN). Guidelines. Breast Cancer. Version 2, 2022). Available at: www.nccn.org, Accessed January 12, 2022). Burstein HJ, Curigliano G, Thurlimann B, Weber WP, Poortmans P, Regan MM, et al. Customizing Local and Systemic Therapies for Women with Early Breast Cancer: The St. Gallen International Consensus Guidelines for Treatment of Early Breast Cancer 2021. Ann Oncol, 2021, 32(10):1216–35., DOI 10.1016/j.annonc.2021.06.023 CardosoF, Kyriakides S,OhnoS, Penault-LlorcaF, PoortmansP, RubioIT, et al.Early Breast Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol, 2019, 30:1674., DOI 10.1093/annonc/mdz189 Burstein HJ, Curigliano G, Loibl S, Dubsky P, Gnant M, Poortmans P, et al. Estimating the Benefits of Therapy for Early-Stage Breast Cancer: The St. Gallen International Consensus Guidelines for the Primary Therapy of Early Breast Cancer 2019. Ann Oncol, 2019, 30:1541–57., DOI 10.1093/annonc/ mdz235 Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, et al. Early Breast Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol, 2019, 30:1194–220., DOI 10.1093/ annonc/mdz173 Balic M, Thomssen C, Wurstlein R, Gnant M, Harbeck N. St. Gallen/Vienna 2019: A Brief Summary of the Consensus Discussion on the Optimal Primary Breast Cancer Treatment. Breast Care, 2019, 14:103–10., DOI 10.1159/ 000499931 Pukancsik D, Kelemen P, Ujhelyi M, Kovacs E, Udvarhelyi N, Meszaros N, et al. Objective Decision Making between Conventional and Oncoplastic Breast-Conserving Surgery or Mastectomy: An Aesthetic and Functional Prospective Cohort Study. Eur J Surg Oncol, EJSO,, 2017, 43:303–10., DOI 10.1016/j.ejso.2016.11.010 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, et al. Personalizing the Treatment of Women with Early Breast Cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol, 2013, 24: 2206–23., DOI 10.1093/annonc/mdt303 Narod SA. The Impact of Contralateral Mastectomy on Mortality in BRCA1 and BRCA2 Mutation Carriers with Breast Cancer. Breast Cancer Res Treat, 2011, 128:581–3., DOI 10.1007/s10549-011-1479-1 Nijenhuis MV, Rutgers EJ. Conservative Surgery for Multifocal/multicentric Breast Cancer. Breast, 2015, 24(Suppl. 2):S96–9., DOI 10.1016/j.breast.2015. 07.023 Gentilini O, Botteri E, Rotmensz N, Da Lima L, Caliskan M, Garcia-Etienne CA, et al. Conservative Surgery in Patients with Multifocal/multicentric Breast Cancer. Breast Cancer Res Treat, 2009, 113:577–83., DOI 10.1007/s10549-008- 9959-7 Goldhirsch A, Ingle JN, Gelber RD, Coates AS, Thurlimann B, Senn H-J. Thresholds for Therapies: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2009. Ann Oncol, 2009, 20:1319–29., DOI 10.1093/annonc/mdp322 Rubio IT, Wyld L, Esgueva A, Kovacs T, Cardoso MJ, Leidenius M, et al. Variability in Breast Cancer Surgery Training across Europe: An ESSOEUSOMA International Survey. Eur J Surg Oncol, 2019, 45:567–72., DOI 10. 1016/j.ejso.2019.01.003 Hennigs A, Hartmann B, Rauch G, Golatta M, Tabatabai P, Domschke C, et al. Long-term Objective Esthetic Outcome after Breast-Conserving Therapy. Breast Cancer Res Treat, 2015, 153:345–51., DOI 10.1007/s10549-015-3540-y Losken A, Dugal CS, Styblo TM, Carlson GW. A Meta-Analysis Comparing Breast Conservation Therapy Alone to the Oncoplastic Technique. Ann Plast Surg, 2014, 72:145–9., DOI 10.1097/sap.0b013e3182605598 Kelemen P, Pukancsik D, Ujhelyi M, Savolt A, Kovacs E, Ivady G, et al. Comparison of Clinicopathologic, Cosmetic and Quality of Life Outcomes in 700 Oncoplastic and Conventional Breast-Conserving Surgery Cases: A Singlecentre Retrospective Study. Eur J Surg Oncol, 2019, 45:118–24., DOI 10.1016/j. ejso.2018.09.006 Association of Breast Surgery at BASO; Association of Breast Surgery at BAPRAS; Training Interface Group in Breast SurgeryBaildam A, Bishop H, Boland G, et al. Oncoplastic Breast Surgery-Aa Guide to Good Practice. Eur J Surg Oncol, 2007, 33(Suppl. 1):S1–23., DOI 10.1016/j.ejso.2007.04.014 Matrai Z, Gulyas G, Kovacs T, Kasler M. Principles and Practice of Oncoplastic Breast Surgery. Budapest, Hungary: Medicina, 2019). Rezai M, Knispel S, Kellersmann S, Lax H, Kimmig R, Kern P. Systematization of Oncoplastic Surgery: Selection of Surgical Techniques and Patient-Reported Outcome in a Cohort of 1,035 Patients. Ann Surg Oncol, 2015, 22:3730–7., DOI 10.1245/s10434-015-4396-4 Weber WP, Haug M, Kurzeder C, Bjelic-Radisic V, Koller R, Reitsamer R, et al. Oncoplastic Breast Consortium Consensus Conference on Nipple-Sparing Mastectomy. Breast Cancer Res Treat, 2018, 172(3):523–37., DOI 10.1007/ s10549-018-4937-1 Dorogi B, Ujhelyi M, Kenessey I, Ivady G, Matrai Z. Clinicopathological Correlations of Areola-sparing Mastectomies versus Nipple-sparing Mastectomies: Analysis of the Oncological and Cosmetic Importance of the Components of the Nipple-areola Complex. Breast J, 2020, 26(10):2081–6., DOI 10.1111/tbj.13957 Ujhelyi M, Savolt A, Fulop R, Meszaros N, Matrai Z. Evaluation of theMedially Pedicled Skin-Reducing Nipple-Sparing Mastectomy as a Standard Mastectomy Technique for Large and Ptotic Breasts. Breast J, 2020, 26(11): 2276–9., DOI 10.1111/tbj.13985 Moran MS, Schnitt SJ, Giuliano AE, Harris JR, Khan SA,Horton J, et al. Society of Surgical Oncology-American Society for Radiation Oncology Consensus Guideline on Margins for Breast-Conserving Surgery with Whole-Breast Irradiation in Stages I and II Invasive Breast Cancer. J Clin Oncol, 2014, 32:1507–15., DOI 10.1200/jco.2013.53.3935 Masannat YA, Bains SK, Pinder SE, Purushotham AD. Challenges in the Management of Pleomorphic Lobular Carcinoma In Situ of the Breast. Breast, 2013, 22:194–6., DOI 10.1016/j.breast.2013.01.003 Flanagan MR, Rendi MH, Calhoun KE, Anderson BO, Javid SH. Pleomorphic Lobular Carcinoma In Situ: Radiologic-Pathologic Features and Clinical Management. Ann Surg Oncol, 2015, 22:4263–9., DOI 10.1245/s10434-015- 4552-x Takacs T, Paszt A, Simonka Z, Abraham S, Borda B, Ottlakan A, et al. Radioguided Occult Lesion Localisation versus Wire-Guided Lumpectomy in the Treatment of Non-palpable Breast Lesions. Pathol Oncol Res, 2013, 19: 267–73., DOI 10.1007/s12253-012-9578-9 Chan BK, Wiseberg-Firtell JA, Jois RH, Jensen K, Audisio RA. Localization Techniques for Guided Surgical Excision of Non-palpable Breast Lesions. Cochrane Database Syst Rev, 2015, 12:CD009206., DOI 10.1002/14651858. CD009206.pub2 Hoffmann J, Marx M, Hengstmann A, Seeger H, Oberlechner E, Helms G, et al. Ultrasound-Assisted Tumor Surgery in Breast Cancer - A Prospective, Randomized, Single-Center Study, MAC 001). Ultraschall Med, 2019, 40(3):326–32., DOI 10.1055/a-0637-1725 Maraz R, Boross G, Pap-Szekeres J, Rajtar M, Ambrozay E, Cserni G. Internal Mammary sentinel Node Biopsy in Breast Cancer. Is it Indicated? Pathol Oncol Res, 2014, 20:169–77., DOI 10.1007/s12253-013-9680-7 Banys-Paluchowski M, Gasparri M, de Boniface J, Gentilini O, Stickeler E, Hartmann S, et al. Surgical Management of the Axilla in Clinically Node- Positive Breast Cancer Patients Converting to Clinical Node Negativity through Neoadjuvant Chemotherapy: Current Status, Knowledge Gaps, and Rationale for the EUBREAST-03 AXSANA Study. Cancers, 2021, 13(7):1565., DOI 10.3390/cancers13071565 Kuemmel S, Heil J, Rueland A, Seiberling C, Harrach H, Schindowski D, et al. A Prospective, Multicenter Registry Study to Evaluate the Clinical Feasibility of Targeted Axillary Dissection, TAD, in Node-Positive Breast Cancer Patients. Surg, 2020). [Epub ahead of print]., DOI 10.1097/SLA.0000000000004572 Cserni G. Orszemnyirokcsomo-biopszia Es Axillaris Blokkdissectio Korai Emlorakban – Algoritmus Magyarazatokkal, Nyitott Kerdesekkel, Sentinel Lymph Node Biopsy and Axillary Block Dissection in Early Breast Cancer – Algorithm with Explanations, Open Questions). Magyar Sebeszet, Hungarian Surgery, J, 2016, 69:3–12., DOI 10.1556/1046.69.2016.1.2 Savolt A, Peley G, Polgar C, Udvarhelyi N, Rubovszky G, Kovacs E, et al. Eightyear Follow up Result of the OTOASOR Trial: The Optimal Treatment of the Axilla – Surgery or Radiotherapy after Positive sentinel Lymph Node Biopsy in Early-Stage Breast Cancer: A Randomized, Single centre, Phase III, Noninferiority Trial. Eur J Surg Oncol, 2017, 43:672–9., DOI 10.1016/j.ejso.2016. 12.011 Donker M, van Tienhoven G, Straver ME, Meijnen P, van de Velde CJH, Mansel RE, et al. Radiotherapy or Surgery of the Axilla after a Positive sentinel Node in Breast Cancer, EORTC 10981-22023 AMAROS): A Randomised, Multicentre, Open-Label, Phase 3 Non-inferiority Trial. Lancet Oncol, 2014, 15:1303–10., DOI 10.1016/s1470-2045(14)70460-7 Giuliano AE, Ballman KV, McCall L, Beitsch PD, BrennanMB, Kelemen PR, et al. Effect of Axillary Dissection vs No Axillary Dissection on 10- Year Overall Survival Among Women with Invasive Breast Cancer and Sentinel Node Metastasis. JAMA, 2017, 318(10):918–26., DOI 10.1001/ jama.2017.11470 Horvath Z, Paszt A, Simonka Z, Latos M, Olah V, Nagyszegi D, et al. Is Intraoperative Touch Imprint Cytology Indicated in the Surgical Treatment of Early Breast Cancers? Eur J Surg Oncol, 2017, 43:1252–7., DOI 10.1016/j.ejso. 2017.01.003 Sabel MS. The Need for Axillary Lymph Node Dissection in T1/T2 Breast Cancer Surgery-Counterpoint. Cancer Res, 2013, 73:7156–60., DOI 10.1158/ 0008-5472.can-13-2094 Kuba MG, Murray MP, Coffey K, Calle C, Morrow M, Brogi E. Morphologic Subtypes of Lobular Carcinoma In Situ Diagnosed on Core Needle Biopsy: Clinicopathologic Features and Findings at Follow-Up Excision. Mod Pathol, 2021, 34:1495–506., DOI 10.1038/ s41379-021-00796-9 Sopik V, Sun P, Narod SA. Impact of Microinvasion on Breast Cancer Mortality in Women with Ductal Carcinoma In Situ. Breast Cancer Res Treat, 2018, 167:787–95., DOI 10.1007/s10549-017-4572-2 Kuerer HM, Smith BD, Chavez-MacGregor M, Albarracin C, Barcenas CH, Santiago L, et al. DCIS Margins and Breast Conservation: MD Anderson Cancer Center Multidisciplinary Practice Guidelines and Outcomes. J Cancer, 2017, 8:2653–62., DOI 10.7150/jca.20871 Kuhl CK, Schrading S, Bieling HB, Wardelmann E, Leutner CC, Koenig R, et al. MRI for Diagnosis of Pure Ductal Carcinoma In Situ: a Prospective Observational Study. The Lancet, 2007, 370:485–92., DOI 10.1016/s0140- 6736(07)61232-x Rageth CJ, O’Flynn EAM, Pinker K, Kubik-Huch RA, Mundinger A, Decker T, et al. Second International Consensus Conference on Lesions of Uncertain Malignant Potential in the Breast, B3 Lesions). Breast Cancer Res Treat, 2019, 174:279–96., DOI 10.1007/s10549-018-05071-1 Lee JS, Yoon K, Onyshchenko M. Sarcoma of the Breast: Clinical Characteristics and Outcomes of 991 Patients from the National Cancer Database. Sarcoma, 2021, 2021:8828158., DOI 10.1155/2021/8828158 van Uden DJP, van Laarhoven HWM, Westenberg AH, de Wilt JHW, Blanken-Peeters CFJM. Inflammatory Breast Cancer: an Overview. Crit Rev Oncol Hematol, 2015, 93:116–26., DOI 10.1016/j.critrevonc.2014.09.003 Guidroz JA, Scott-Conner CEH, Weigel RJ.Management of Pregnant Women with Breast Cancer. J Surg Oncol, 2011, 103:337–40., DOI 10.1002/jso.21673 Matrai Z, Banhidy F, Teglas M, Kovacs E, Savolt A, Udvarhelyi N, et al. Sentinel Lymph Node Biopsy in Gestational Breast Cancer. Orv Hetil, 2013, 154:1991–7., DOI 10.1556/OH.2013.29771 Gropper AB, Calvillo KZ, Dominici L, Troyan S, Rhei E, Economy KE, et al. Sentinel Lymph Node Biopsy in Pregnant Women with Breast Cancer. Ann Surg Oncol, 2014, 21:2506–11., DOI 10.1245/s10434-014-3718-2 Lee H-B, Han W. Unique Features of Young Age Breast Cancer and its Management. J Breast Cancer, 2014, 17:301–7., DOI 10.4048/jbc.2014.17.4.301 Kroman N, Holtveg H, Wohlfahrt J, Jensen M-B, Mouridsen HT, Blichert-Toft M, et al. Effect of Breast-Conserving Therapy versus Radical Mastectomy on Prognosis for Young Women with Breast Carcinoma. Cancer, 2004, 100: 688–93., DOI 10.1002/cncr.20022 Szollar A, Ujhelyi M, Polgar C, Olah E, Pukancsik D, Rubovszky G, et al. A Long-Term Retrospective Comparative Study of the Oncological Outcomes of 598 Very Young, ≤35 Years, and Young, 36-45 Years, Breast Cancer Patients. Eur J Surg Oncol, 2019, 45:2009–15., DOI 10.1016/j.ejso.2019.06.007 Fancellu A, Sanna V. High Mastectomy Rates in Young and Very Young Patients with Breast Cancer. Are They Justified? Eur J Surg Oncol, 2020, 46: 1391–2., DOI 10.1016/j.ejso.2019.12.024 Patten DK, Sharifi LK, Fazel M. New Approaches in the Management of Male Breast Cancer. Clin Breast Cancer, 2013, 13:309–14., DOI 10.1016/j.clbc.2013. 04.003 Antoniou A, Pharoah PDP, Narod S, Risch HA, Eyfjord JE, Hopper JL, et al. Average Risks of Breast and Ovarian Cancer Associated with BRCA1 or BRCA2 Mutations Detected in Case Series Unselected for Family History: A Combined Analysis of 22 Studies. Am J Hum Genet, 2003, 72:1117–30., DOI 10. 1086/375033 Paluch-Shimon S, Cardoso F, Sessa C, Balmana J, Cardoso MJ, Gilbert F, et al. Prevention and Screening in BRCA Mutation Carriers and Other Breast/ ovarian Hereditary Cancer Syndromes: ESMO Clinical Practice Guidelines for Cancer Prevention and Screening. Ann Oncol, 2016, 27(Suppl. 5):v103–v110., DOI 10.1093/annonc/mdw327 Kurian AW, Lichtensztajn DY, Keegan THM, Nelson DO, Clarke CA, Gomez SL. Use of and Mortality after Bilateral Mastectomy Compared with Other Surgical Treatments for Breast Cancer in California, 1998-2011. JAMA, 2014, 312:902–14., DOI 10.1001/jama.2014.10707 Guth U, Myrick ME, Viehl CT, Weber WP, Lardi AM, Schmid SM. Increasing Rates of Contralateral Prophylactic Mastectomy - A Trend Made in USA? Eur J Surg Oncol, 2012, 38:296–301., DOI 10.1016/j.ejso. 2011.12.014 Yao K, Winchester DJ, Czechura T, Huo D. Contralateral Prophylactic Mastectomy and Survival: Report from the National Cancer Data Base, 1998-2002. Breast Cancer Res Treat, 2013, 142:465–76., DOI 10.1007/s10549- 013-2745-1 Hwang ES, Lichtensztajn DY, Gomez SL, Fowble B, Clarke CA. Survival after Lumpectomy and Mastectomy for Early Stage Invasive Breast Cancer. Cancer, 2013, 119:1402–11., DOI 10.1002/cncr.27795 Kronowitz SJ. State of the Art and Science in Postmastectomy Breast Reconstruction. Plast Reconstr Surg, 2015, 135:755e–71e., DOI 10.1097/PRS. 0000000000001118 Potter S, Conroy EJ, Cutress RI, Williamson PR, Whisker L, Thrush S, et al. Short-term Safety Outcomes of Mastectomy and Immediate Implant-Based Breast Reconstruction with and without Mesh, iBRA): A Multicentre, Prospective Cohort Study. Lancet Oncol, 2019, 20:254–66., DOI 10.1016/ S1470-2045(18)30781-2 Fisher B, Brown A, Mamounas E, Wieand S, Robidoux A, Margolese RG, et al. Effect of Preoperative Chemotherapy on Local-Regional Disease in Women with Operable Breast Cancer: Findings from National Surgical Adjuvant Breast and Bowel Project B-18. J Clin Oncol, 1997, 15:2483–93., DOI 10.1200/jco.1997. 15.7.2483 van Nes JGH, Putter H, Putter H, Julien J-P, Tubiana-Hulin M, van de Vijver M, et al. Preoperative Chemotherapy Is Safe in Early Breast Cancer, Even after 10 Years of Follow-Up; Clinical and Translational Results from the EORTC Trial 10902. Breast Cancer Res Treat, 2009, 115:101–13., DOI 10.1007/s10549- 008-0050-1 Kaufmann M, von Minckwitz G, Mamounas EP, Cameron D, Carey LA, Cristofanilli M, et al. Recommendations from an International Consensus Conference on the Current Status and Future of Neoadjuvant Systemic Therapy in Primary Breast Cancer. Ann Surg Oncol, 2012, 19:1508–16., DOI 10.1245/s10434-011-2108-2 Early Breast Cancer Trialists’ Collaborative Group, EBCTCG). Long-term Outcomes for Neoadjuvant versus Adjuvant Chemotherapy in Early Breast Cancer: Meta-Analysis of Individual Patient Data from Ten Randomised Trials. Lancet Oncol, 2018, 19(27–39):27–39., DOI 10.1016/S1470-2045(17, 30777-5 Ataseven B, Lederer B, Blohmer JU, Denkert C, Gerber B, Heil J, et al. Impact of Multifocal or Multicentric Disease on Surgery and Locoregional, Distant and Overall Survival of 6,134 Breast Cancer Patients Treated with Neoadjuvant Chemotherapy. Ann Surg Oncol, 2015, 22:1118–27., DOI 10.1245/s10434-014- 4122-7 Kuehn T, Bauerfeind I, Fehm T, Fleige B, Hausschild M, Helms G, et al. Sentinel-lymph-node Biopsy in Patients with Breast Cancer before and after Neoadjuvant Chemotherapy, SENTINA): a Prospective, Multicentre Cohort Study. Lancet Oncol, 2013, 14:609–18., DOI 10.1016/s1470-2045(13)70166-9 Boileau J-F, Poirier B, Basik M, Holloway CMB, Gaboury L, Sideris L, et al. Sentinel Node Biopsy after Neoadjuvant Chemotherapy in Biopsy-Proven Node-Positive Breast Cancer: the SN FNAC Study. J Clin Oncol, 2015, 33: 258–64., DOI 10.1200/jco.2014.55.7827 Boughey JC, Suman VJ, Mittendorf EA, Ahrendt GM, Wilke LG, Taback B, et al. Factors Affecting sentinel Lymph Node Identification Rate after Neoadjuvant Chemotherapy for Breast Cancer Patients Enrolled in ACOSOG Z1071, Alliance). Ann Surg, 2015, 261:547–52., DOI 10.1097/sla. 0000000000000551 Galimberti V. Feasibility of sentinel Node Biopsy in Breast Cancer after Neoadjuvant Treatment. Breast, 2015, 24(Suppl. 1):PG 9.02., DOI 10.1016/ s0960-9776(15)70035-4 Simons JM, van Nijnatten TJA, van der Pol CC, Luiten EJT, Koppert LB, Smidt ML. Diagnostic Accuracy of Different Surgical Procedures for Axillary Staging after Neoadjuvant Systemic Therapy in Node-Positive Breast Cancer. Ann Surg, 2019, 269:432–42., DOI 10.1097/sla.0000000000003075 Cardoso F, Senkus E, Costa A, Papadopoulos E, Aapro M, Andre F, et al. 4th ESO-ESMO International Consensus Guidelines for Advanced Breast Cancer, ABC 4). Ann Oncol, 2018, 29:1634–57., DOI 10.1093/annonc/mdy192 Walstra CJEF, Schipper R-J, Poodt IGM, van Riet YE, Voogd AC, van der Sangen MJC, et al. Repeat Breast-Conserving Therapy for Ipsilateral Breast Cancer Recurrence: A Systematic Review. Eur J Surg Oncol, 2019, 45:1317–27., DOI 10.1016/j.ejso.2019.02.008 Ishitobi M, Fukui R, Hashimoto Y, Kittaka N, Nakayama T, Tamaki Y. pTis and pT1a Ipsilateral Breast Tumor Recurrence Is Associated with Good Prognosis after Salvage Surgery. Oncology, 2018, 94:12–8., DOI 10.1159/ 000480516 Wapnir IL, Price KN, Anderson SJ, Robidoux A, Martin M, Nortier JWR, et al. Efficacy of Chemotherapy for ER-Negative and ER-Positive Isolated Locoregional Recurrence of Breast Cancer: Final Analysis of the CALOR Trial. J Clin Oncol, 2018, 36:1073–9., DOI 10.1200/jco.2017.76.5719 Cardoso F, Spence D, Mertz S, Corneliussen-James D, Sabelko K, Gralow J, et al. Global Analysis of Advanced/metastatic Breast Cancer: Decade Report, 2005-2015). Breast, 2018, 39:131–8., DOI 10.1016/j.breast.2018.03.002 Gennari A, Andre F, Barrios CH, Cortes J, de Azambuja E, DeMichele A, et al. ESMO Clinical Practice Guideline for the Diagnosis, Staging and Treatment of Patients with Metastatic Breast Cancer. Ann Oncol, 2021, 32:1475–95., DOI 10. 1016/j.annonc.2021.09.019 Kwapisz D. Oligometastatic Breast Cancer. Breast Cancer, 2019, 26:138–46., DOI 10.1007/s12282-018-0921-1 Khan SA, Zhao F, Solin LJ, Goldstein LJ, Cella D, BasikM, et al. A Randomized Phase III Trial of Systemic Therapy Plus Early Local Therapy versus Systemic Therapy Alone in Women with De Novo Stage IV Breast Cancer: A Trial of the ECOG-ACRIN Research Group, E2108). J Clin Oncol, 2020, 38:LBA2., DOI 10. 1200/jco.2020.38.18_suppl.lba2 Soran A, Dogan L, Isik A, Ozbas S, Trabulus DC, Demirci U, et al. The Effect of Primary Surgery in Patients with De Novo Stage IV Breast Cancer with Bone Metastasis Only, Protocol BOMET MF 14-01): a multi-center, Prospective Registry Study. Ann Surg Oncol, 2020). Available at: http://link.springer.com/10.1245/s10434- 021-09621-8, In press, 38., DOI 10.1200/JCO.2020.38.18_suppl.LBA2 Bjelic-Radisic V, Fitzal F, Knauer FM, Steger G, Egle D, Greil R, et al. Primary Surgery versus No Surgery in Synchronous Metastatic Breast Cancer: Patient- Reported Quality-Of-Life Outcomes of the Prospective Randomized Multicenter ABCSG-28 Posytive Trial. BMC Cancer, 2020, 20:392., DOI 10. 1186/s12885-020-06894-2 Khan SA, Zhao F, Solin LJ, Goldstein LJ, Cella D, BasikM, et al. A Randomized Phase III Trial of Systemic Therapy Plus Early Local Therapy versus Systemic Therapy Alone in Women with De Novo Stage IV Breast Cancer: A Trial of the ECOG-ACRIN Research Group, E2108). J Clin Oncol, 2020, 38(Suppl. l): LBA2., DOI 10.1200/jco.2020.38.18_suppl.lba2 Ruiz A, Wicherts DA, Sebagh M, Giacchetti S, Castro-Benitez C, van Hillegersberg R, et al. Predictive Profile-Nomogram for Liver Resection for Breast Cancer Metastases: An Aggressive Approach with Promising Results. Ann Surg Oncol, 2017, 24:535–45., DOI 10.1245/s10434-016-5522-7 Motomura K, Inaji H, Komoike Y, Kasugai T, Noguchi S, Koyama H. Sentinel Node Biopsy Guided by Indocyanin Green Dye in Breast Cancer Patients. Jpn J Clin Oncol, 1999, 29:604–7., DOI 10.1093/jjco/29.12.604 Lin J, Lin L-S, Chen D-R, Lin K-J, Wang Y-F, Chang Y-J. Indocyanine green FluorescenceMethod for sentinel Lymph Node Biopsy in Breast Cancer. Asian J Surg, 2020, 43:1149–53., DOI 10.1016/j.asjsur.2020.02.003 Thill M, Kurylcio A, Blechmann R, van Haasteren V, Grosse B, Berclaz G, et al. The SentiMag Study: sentinel Node Biopsy with Superparamagnetic Iron Oxide, SPIO, vs. Radioisotope. Eur J Cancer, 2013, 49:S260–S261., DOI 10. 1016/j.breast.2014.01.004 Mok CW, Tan SM, Zheng Q, Shi L. Network Meta-analysis of Novel and Conventional sentinel Lymph Node Biopsy Techniques in Breast Cancer. BJS Open, 2019, 3:445–52., DOI 10.1002/bjs5.50157 Goonawardena J, Yong C, Law M. Use of Indocyanine green Fluorescence Compared to Radioisotope for sentinel Lymph Node Biopsy in Early-Stage Breast Cancer: Systematic Review and Meta- Analysis. Am J Surg, 2020, 220:665–76., DOI 10.1016/j.amjsurg.2020. 02.001 Forrai G, Kovacs E, Ambrozay E, BartaM, Bobely K, Lengyel Z, et al. Use of Diagnostic Imaging Modalities in Modern Screening, Diagnostics and Management of Breast Tumours. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022, 220, 3). in press., DOI 10.1016/j.amjsurg.2020.02.001 Cserni G, Francz M, Jaray B, Kalman E, Kovacs I, Krenacs T, et al. Pathological Diagnosis, Work-Up and Reporting of Breast Cancer 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, Forthcoming 2022). Rubovszky G, Kocsis J, Boer K, Chilingirova N, Dank M, Kahan Z, et al. Systemic Treatment of Breast Cancer. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, Forthcoming 2022). Polgar C, Kahan Z, Ivanov O, Chorvath M, Ligacova A, Csejtei A, et al. Radiotherapy of Breast Cancer – Professional Guideline. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, Forthcoming 2022). Kahan Z, Szanto I, Dudas R, Kapitany Z, Molnar M, Koncz Z, et al. Breast Cancer: Follow-Up, Rehabilitation, Psycho-Oncology. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, Forthcoming 2022). NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610377 EP 1610395 DI 10.3389/pore.2022.1610377 PG 19 ER PT J AU Polgar, Cs Kahan, Zs Ivanov, O Chorvath, M Ligacova, A Csejtei, A Gabor, G Landherr, L Mangel, L Mayer, Fodor, J AF Polgar, Csaba Kahan, Zsuzsanna Ivanov, Olivera Chorvath, Martin Ligacova, Andrea Csejtei, Andras Gabor, Gabriella Landherr, Laszlo Mangel, Laszlo Mayer, Arpad Fodor, Janos TI Radiotherapy of Breast Cancer— Professional Guideline 1st Central-Eastern European Professional Consensus Statement on Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Guideline DE breast cancer; radiotherapy; guidelines; radiation oncology; consensus ID breast cancer; radiotherapy; guidelines; radiation oncology; consensus AB The international radiotherapy (RT) expert panel has revised and updated the RT guidelines that were accepted in 2020 at the 4th Hungarian Breast Cancer Consensus Conference, based on new scientific evidence. Radiotherapy after breast-conserving surgery (BCS) is indicated in ductal carcinoma in situ (stage 0), as RT decreases the risk of local recurrence (LR) by 50–60%. In early stage (stage I-II) invasive breast cancer RT remains a standard treatment following BCS. However, in elderly (≥70 years) patients with stage I, hormone receptor-positive tumour, hormonal therapy without RT can be considered. Hypofractionated whole breast irradiation (WBI) and for selected cases accelerated partial breast irradiation are validated treatment alternatives to conventional WBI administered for 5 weeks. Following mastectomy, RT significantly decreases the risk of LR and improves overall survival of patients who have 1 to 3 or ≥4 positive axillary lymph nodes. In selected cases of patients with 1 to 2 positive sentinel lymph nodes axillary dissection can be substituted with axillary RT. After neoadjuvant systemic treatment (NST) followed by BCS, WBI is mandatory, while after NST followed by mastectomy, locoregional RT should be given in cases of initial stage III–IV and ypN1 axillary status. C1 [Polgar, Csaba] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of OncotherapySzeged, Hungary. [Ivanov, Olivera] University of Novi Sad, Medical Faculty Novi SadNovi Sad, Serbia. [Chorvath, Martin] Slovak Medical University, St. Elisabeth Cancer Institute, Department of Radiation OncologyBratislava, Slovakia. [Ligacova, Andrea] Slovak Medical University, St. Elisabeth Cancer Institute, Department of Radiation OncologyBratislava, Slovakia. [Csejtei, Andras] Vas Megyei Markusovszky Korhaz, OnkoradiologiaSzombathely, Hungary. [Gabor, Gabriella] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. [Landherr, Laszlo] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai KozpontBudapest, Hungary. [Mangel, Laszlo] University of Pecs, Oncotherapy InstitutePecs, Hungary. [Mayer, Arpad] Fovarosi Onkormanyzat Uzsoki utcai Korhaza, Onkoradiologiai KozpontBudapest, Hungary. [Fodor, Janos] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. RP Polgar, Cs (reprint author), National Institute of Oncology, Center of Radiotherapy, Budapest, Hungary. EM polgar.csaba@oncol.hu CR Bijker N, Meijnen P, Peterse JL, Bogaerts J, Van Hoorebeeck I, Julien JP, et al. Breast-conserving Treatment with or without Radiotherapy in Ductal Carcinoma In Situ: Ten-Year Results of European Organisation for Research and Treatment of Cancer Randomized Phase III Trial 10853 – a Study by the EORTC Breast Cancer Cooperative Group and EORTC Radiotherapy Group. J Clin Oncol, 2006, 24(1–8):3381–7., DOI 10.1200/ JCO.2006.06.1366 Cutulli B, Bernier J, Poortmans P. Radiotherapy in DCIS, an Underestimated Benefit? Radiother Oncol, 2014, 112:1–8., DOI 10.1016/j. radonc.2014.06.011 CuzickSestak JI, Pinder SE, Ellis IO, Forsyth S, Bundred NJ, et al. Effect of Tamoxifen and Radiotherapy in Women with Locally Excised Ductal Carcinoma In Situ: Long-Term Results from the UK/ANZ DCIS Trial. Lancet Oncol, 2011, 12:21–9., DOI 10.1016/S1470-2045(10)70266-7 Early Breast Cancer Trialists’ Collaborative Group. Overview of Randomised Trials of Radiotherapy in Ductal Carcinoma In Situ of the Breast. J Natl Cancer Inst Monogr, 2010, 41:162–77., DOI 10.1093/jncimonographs/lgq039 Emdin SO, Granstrand B, Ringberg A, Sandelin K, Arnesson LG, Nordgren H, et al. SweDCIS: Radiotherapy after Sector Resection for Ductal Carcinoma In Situ of the Breast. Results of a Randomised Trial in a Population Offered Mammography Screening. Acta Oncol, 2006, 45:536–43., DOI 10.1080/ 02841860600681569 Fisher ER, Dignam J, Tan-Chiu E, Costantino J, Fisher B, Paik S, et al. Pathologic Findings from the National Surgical Adjuvant Breast Project, NSABP, Eight-Year Update of Protocol B-17: Intraductal Carcinoma. Cancer, 1999, 86:429–38., DOI 10.1002/(sici)1097-0142(19990801)86: 3<429::aid-cncr11>3.0.co;2-y Meattini I, Livi L, Franceschini D, Saieva C, Meacci F, Marrazzo L, et al. Role of Radiotherapy Boost in Women with Ductal Carcinoma In Situ: A Singlecenter Experience in a Series of 389 Patients. Eur J Surg Oncol, 2013, 39: 613–8., DOI 10.1016/j.ejso.2013.03.002 Polgar C, Kahan Z, Orosz Z, Gabor G, Hadijev J, Cserni G, et al. The Role of Radiotherapy in the Conservative Treatment of Ductal Carcinoma In Situ of the Breast. Pathol Oncol Res, 2008, 14:179–92., DOI 10.1007/s12253-008- 9044-x Stuart KE, Houssami N, Taylor R, Hayen A, Boyages J. Long-term Outcomes of Ductal Carcinoma In Situ of the Breast: a Systematic Review, Meta- Analysis and Meta-Regression Analysis. BMC Cancer, 2015, 15:890., DOI 10. 1186/s12885-015-1904-7 Early Breast Cancer Trialists’ Collaborative Group, Darby S, McGale P, Correa C, Taylor C, ARRiagada R, et al. Effect of Radiotherapy after Breast-Conserving Surgery on 10-year Recurrence and 15-year Breast Cancer Death: Meta-Analysis of Individual Patient Data for 10801 Women in 17 Randomised Trials. Lancet, 2011, 378:1707–16., DOI 10. 1016/S0140-6736(11)61629-2 Fisher B, Anderson S, Bryant J, Margolese RG, Deutsch M, Fisher ER, et al. Twenty-year Follow-Up of a Randomized Trial Comparing Total Mastectomy, Lumpectomy, and Lumpectomy Plus Irradiation for the Treatment of Invasive Breast Cancer. N Engl J Med, 2002, 347:1233–41., DOI 10.1056/nejmoa022152 Hughes KS, Schnaper LA, Bellon JR, Cirrincione CT, Berry DA, McCormick B, et al. Lumpectomy Plus Tamoxifen with or without Irradiation in Women Age 70 Years or Older with Early Breast Cancer: Long-Term Follow-Up of CALGB 9343. J Clin Oncol, 2013, 31:2382–7., DOI 10.1200/jco.2012.45.2615 Kunkler IH, Williams LJ, Jack WJL, Cameron DA, Dixon JM. Breastconserving Surgery with or without Irradiation in Women Aged 65 Years or Older with Early Breast Cancer, PRIME II): a Randomised Controlled Trial. Lancet Oncol, 2015, 16:266–73., DOI 10.1016/s1470-2045(14)71221-5 Litiere S, Werutsky G, Fentiman IS, Rutgers E, Christiaens MR, Van Limbergen E, et al. Breast Conserving Therapy versus Mastectomy for Stage I-II Breast Cancer: 20 Year Follow-Up of the EORTC 10801 Phase 3 Randomised Trial. Lancet Oncol, 2012, 13:412–9., DOI 10.1016/S1470- 2045(12)70042-6 NCCN. NCCN Practice Guidelines in Oncology. Plymouth Meeting, PA: Breast Cancer, 2019). Polgar C, Major T, Fodor J. [Modern Radiotherapy after Breast-Conserving Surgery].. Orv Hetil, 2012, 153:45–55., DOI 10.1556/OH.2012.29248 Veronesi U, Cascinelli N, Mariani L, Greco M, Saccozzi R, Luini A, et al. Twenty-year Follow-Up of a Randomized Study Comparing Breast- Conserving Surgery with Radical Mastectomy for Early Breast Cancer. N Engl J Med, 2002, 347:1227–32., DOI 10.1056/nejmoa020989 Guenzi M, Blandino G, Vidili MG, Aloi D, Configliacco E, Verzanini E, et al. Hypofractionated Irradiation of Infra-supraclavicular Lymph Nodes after Axillary Dissection in Patients with Breast Cancer post-conservative Surgery: Impact on Late Toxicity. Radiat Oncol, 2015, 10:177., DOI 10.1186/s13014- 015-0480-y Haviland JS, Owen JR, Dewar JA, Agrawal RK, Barrett J, Barrett-Lee PJ, et al. The UK Standardisation of Breast Radiotherapy, START, Trials of Radiotherapy Hypofractionation for Treatment of Early Breast Cancer: 10-year Follow-Up Results of Two Randomised Controlled Trials. Lancet Oncol, 2013, 14:1086–94., DOI 10.1016/s1470-2045(13)70386-3 Valle LF, Agarwal S, Bickel KE, Herchek HA, Nalepinski DC, Kapadia NS. Hypofractionated Whole Breast Radiotherapy in Breast Conservation for Early-Stage Breast Cancer: a Systematic Review and Meta-Analysis of Randomized Trials. Breast Cancer Res Treat, 2017, 162:409–17., DOI 10. 1007/s10549-017-4118-7 Wang SL, Fang H, Song YW, Wang WH, Hu C, Liu YP, et al. Hypofractionated versus Conventional Fractionated Postmastectomy Radiotherapy for Patients with High-Risk Breast Cancer: a Randomised, Non-inferiority, Open-Label, Phase 3 Trial. Lancet Oncol, 2019, 20:352–60., DOI 10.1016/s1470-2045(18)30813-1 Whelan TJ, Pignol JP, Levine MN, Julian JA, MacKenzie R, Parpia S, et al. Long-term Results of Hypofractionated Radiation Therapy for Breast Cancer. New Engl J Med, 2010, 362:513–20., DOI 10.1056/NEJMoa0906260 Yarnold J, Somaiah N, Bliss JM. Hypofractionated Radiotherapy in Early Breast Cancer: Clinical, Dosimetric and Radiogenomic Issues. The Breast, 2015, 24:S108–13., DOI 10.1016/j.breast.2015.07.025 Coles CE, Griffin CI, Kirby AM, Titley J, Agrawal RK, Alhasso A, et al. Partial Breast Radiotherapy after Breast Conservation Surgery for Patients with Early Breast Cancer, UK IMPORT LOW Trial): 5-year Results from a Multicentre, Randomised, Controlled, Phase 3, Non-inferiority Trial. The Lancet, 2017, 390:1048–60., DOI 10.1016/s0140-6736(17)31145-5 Hannoun-Levi JM, Lam Cham Kee D, Gal J, Schiappa R, Hannoun A, Fouche Y, et al. Accelerated Partial Breast Irradiation in the Elderly: 5- year Results of the Single Fraction Elderly Breast Irradiation, SiFEBI, Phase I/II Trial. Brachytherapy, 2020, 19:90–6., DOI 10.1016/j.brachy. 2019.10.007 Khan AJ, Chen PY, Yashar C, Poppe MM, Li L, Abou Yehia Z, et al. Threefraction Accelerated Partial Breast Irradiation, APBI, Delivered with Brachytherapy Applicators Is Feasible and Safe: First Results from the TRIUMPH-T Trial. Int J Radiat Oncol Biol Phys, 2019, 104:67–74., DOI 10. 1016/j.ijrobp.2018.12.050 Livi L, Meattini I, Marrazzo L, Simontacchi G, Pallotta S, Saieva C, et al. Accelerated Partial Breast Irradiation Using Intensity-Modulated Radiotherapy versus Whole Breast Irradiation: 5-year Survival Analysis of a Phase 3 Randomised Controlled Trial. Eur J Cancer, 2015, 51:451–63., DOI 10.1016/j.ejca.2014.12.013 Meszaros N, Major T, Stelczer G, Zaka Z, Mozsa E, Pukancsik D, et al. Implementation of Image-Guided Intensity-Modulated Accelerated Partial Breast Irradiation: Three-Year Results of a Phase II Clinical Study. Strahlenther Onkol, 2017, 193:70–9., DOI 10.1007/s00066-016-1074-9 Polgar C, Fodor J, Major T, Sulyok Z, Kasler M. Breast-conserving Therapy with Partial or Whole Breast Irradiation: Ten-Year Results of the Budapest Randomized Trial. Radiother Oncol, 2013, 108:197–202., DOI 10.1016/j. radonc.2013.05.008 Polgar C, Ott OJ, Hildebrandt G, Kauer-Dorner D, Knauerhase H, Major T, et al. Late Side-Effects and Cosmetic Results of Accelerated Partial Breast Irradiation with Interstitial Brachytherapy versus Whole-Breast Irradiation after Breast-Conserving Surgery for Low-Risk Invasive and In-Situ Carcinoma of the Female Breast: 5-year Results of a Randomised, Controlled, Phase 3 Trial. Lancet Oncol, 2017, 18:259–68., DOI 10.1016/ S1470-2045(17)30011-6 Polgar C, van Limbergen E, Potter R, Kovacs G, Polo A, Lyczek J, et al. Patient selection for accelerated partial-breast irradiation, APBI, after breastconserving surgery: recommendations of the Groupe Europeen de Curietherapie-European Society for Therapeutic Radiology and Oncology, GEC-ESTRO, breast cancer working group based on clinical evidence, 2009). Radiother Oncol, 2009, 94:264–73., DOI 10.1016/j.radonc.2010.01.014 Schafer R, Strnad V, Polgar C, Uter W, Hildebrandt G, Ott OJ, et al. Qualityof- life Results for Accelerated Partial Breast Irradiation with Interstitial Brachytherapy versus Whole-Breast Irradiation in Early Breast Cancer after Breast-Conserving Surgery, GEC-ESTRO): 5-year Results of a Randomised, Phase 3 Trial. Lancet Oncol, 2018, 19:834–44., DOI 10.1016/ S1470-2045(18)30195-5 Strnad V, Ott OJ, Hildebrandt G, Kauer-Dorner D, Knauerhase H, Major T, et al. 5-year Results of Accelerated Partial Breast Irradiation Using Sole Interstitial Multicatheter Brachytherapy versus Whole-Breast Irradiation with Boost after Breast-Conserving Surgery for Low-Risk Invasive and In- Situ Carcinoma of the Female Breast: a Randomised, Phase 3, Non-inferiority Trial.. The Lancet, 2016, 387:229–38., DOI 10.1016/s0140-6736(15)00471-7 Vicini FA, Cecchini RS, White JR, Arthur DW, Julian TB, Rabinovitch RA, et al. Long-term Primary Results of Accelerated Partial Breast Irradiation after Breast-Conserving Surgery for Early-Stage Breast Cancer: a Randomised, Phase 3, Equivalence Trial. The Lancet, 2019, 394:2155–64., DOI 10.1016/s0140-6736(19)32514-0 Wilkinson JB, Chen PY, Wallace MF, Shah CS, Benitez PR, Martinez AA, et al. Six-year Results from a Phase I/II Trial for Hypofractionated Accelerated Partial Breast Irradiation Using a 2-day Dose Schedule. Am J Clin Oncol, 2018, 41:986–91., DOI 10.1097/coc.0000000000000402 Whelan TJ, Julian JA, Berrang TS, Kim DH, Germain I, Nichol AM, et al. External Beam Accelerated Partial Breast Irradiation versus Whole Breast Irradiation after Breast Conserving Surgery in Women with Ductal Carcinoma In Situ and Node-Negative Breast Cancer, RAPID): a Randomised Controlled Trial. The Lancet, 2019, 394:2165–72., DOI 10. 1016/S0140-6736(19)32515-2 Early Breast Cancer Trialists’ Collaborative Group, McGale P, Taylor C, Correa C, Cutter D, Duane F, et al. Effect of Radiotherapy after Mastectomy and Axillary Surgery on 10-year Recurrence and 20-year Breast Cancer Mortality: Meta-Analysis of Individual Patient Data for 8135 Women in 22 Randomised Trials. The Lancet, 2014, 383:2127–35., DOI 10.1016/S0140- 6736(14)60488-8 Fodor J, Major T, Polgar C. Sugarkezeles Mastectomia Utan: Evidenciak Es Nyitott Kerdesek., Radiotherapy after Mastectomy: Evidence and Open Questions). Onkologia, 2014, 2:35–9. Fodor J, Polgar C, Major T, Nemeth G. Locoregional Failure 15 Years after Mastectomy in Women with One to Three Positive Axillary Nodes with or without Irradiation: The Significance of Tumour Size. Strahlenther Onkol, 2003, 179:197–202., DOI 10.1007/s00066-003-1010-7 Krug D, Baumann R, Budach W, Dunst J, Feyer P, Fietkau R, et al. Current Controversies in Radiotherapy for Breast Cancer. Radiat Oncol, 2017, 12:25., DOI 10.1186/s13014-017-0766-3 Nielsen HM, Overgaard M, Grau C, Jensen AR, Overgaard J. Study of Failure Pattern Among High-Risk Breast Cancer Patients with or without Postmastectomy Radiotherapy in Addition to Adjuvant Systemic Therapy: Long-Term Results from the Danish Breast Cancer Cooperative Group DBCG 82 B and C Randomized Studies. J Clin Oncol, 2006, 24:2268–75., DOI 10.1200/jco.2005.02.8738 Overgaard M, Hansen PS, Overgaard J, Rose C, Andersson M, Bach F, et al. Postoperative Radiotherapy in High-Risk Premenopausal Women with Breast Cancer Who Receive Adjuvant Chemotherapy. N Engl J Med, 1997, 337:949–55., DOI 10.1056/nejm199710023371401 Overgaard M, Jensen MB, Overgaard J, Hansen PS, Rose C, Andersson M, et al. Postoperative Radiotherapy in High-Risk Postmenopausal Breast- Cancer Patients Given Adjuvant Tamoxifen: Danish Breast Cancer Cooperative Group DBCG 82c Randomised Trial. The Lancet, 1999, 353: 1641–8., DOI 10.1016/s0140-6736(98)09201-0 Overgaard M, Nielsen HM, Overgaard J. Is the Benefit of Postmastectomy Irradiation Limited to Patients with Four or More Positive Nodes, as Recommended in International Consensus Reports? A Subgroup Analysis of the DBCG 82 B&c Trials. Radiother Oncol, 2007, 82:247–53., DOI 10.1016/j. radonc.2007.02.001 Poortmans P. Postmastectomy Radiation in Breast Cancer with One to Three Involved Lymph Nodes: Ending the Debate.. The Lancet, 2014, 383:2104–6., DOI 10.1016/s0140-6736(14)60192-6 Poortmans P, Collette S, Kirkove C, Van Limbergen E, Budach V, Struikmans H, et al. Internal Mammary and Medial Supraclavicular Irradiation in Breast Cancer. N Engl J Med, 2015, 373:317–27., DOI 10.1056/nejmoa1415369 Whelan TJ, Olivotto IA, Parulekar WR, Ackerman I, Chua BH, Nabid A, et al. Regional Nodal Irradiation in Early-Stage Breast Cancer. N Engl JMed, 2015, 373:307–16., DOI 10.1056/nejmoa1415340 Donker M, van Tienhoven G, Straver ME, Meijnen P, van de Velde CJH, Mansel RE, et al. Radiotherapy or Surgery of the Axilla after a Positive sentinel Node in Breast Cancer, EORTC 10981-22023 AMAROS): a Randomised, Multicentre, Open-Label, Phase 3 Non-inferiority Trial. Lancet Oncol, 2014, 15:1303–10., DOI 10.1016/s1470-2045(14)70460-7 Mansel RE, Fallowfield L, Kissin M, Goyal A, Newcombe RG, Dixon JM, et al. Randomized Multicenter Trial of sentinel Node Biopsy versus Standard Axillary Treatment in Operable Breast Cancer: the ALMANAC Trial. J Natl Cancer Inst, 2006, 98:599–609., DOI 10.1093/jnci/djj158 Savolt A, Peley G, Polgar C, Udvarhelyi N, Rubovszky G, Kovacs E, et al. Eight-year Follow up Result of the OTOASOR Trial: The Optimal Treatment of the Axilla - Surgery or Radiotherapy after Positive sentinel Lymph Node Biopsy in Early-Stage Breast Cancer: A Randomized, Single centre, Phase III, Non-inferiority Trial. Eur J Surg Oncol, 2017, 43:672–9., DOI 10.1016/j.ejso. 2016.12.011 Veronesi U, Orecchia R, Zurrida S, Zurrida S, Luini A, Veronesi P, et al. Avoiding Axillary Dissection in Breast Cancer Surgery: a Randomized Trial to Assess the Role of Axillary Radiotherapy. Ann Oncol, 2005, 16:383–8., DOI 10.1093/annonc/mdi089 Louis-Sylvestre C, Clough K, Asselain B, Vilcoq JR, Salmon RJ, Campana F, et al. Axillary Treatment in Conservative Management of Operable Breast Cancer: Dissection or Radiotherapy? Results of a Randomized Study with 15 Years of Follow-Up. J Clin Oncol, 2004, 22:97–101., DOI 10.1200/jco.2004. 12.108 Speers C, Pierce LJ. Molecular Signatures of Radiation Response in Breast Cancer: towards Personalized Decision-Making in Radiation Treatment. Int J Breast Cancer, 2017, 2017:1–7., DOI 10.1155/2017/4279724 Akay CL, Meric-Bernstam F, Hunt KK, Grubbs EG, Bedrosian I, Tucker SL, et al. Evaluation of the MD Anderson Prognostic Index for Local-Regional Recurrence after Breast Conserving Therapy in Patients Receiving Neoadjuvant Chemotherapy. Ann Surg Oncol, 2012, 19:901–7., DOI 10. 1245/s10434-011-2006-7 Chapman CH, Jagsi R. Postmastectomy Radiotherapy after Neoadjuvant Chemotherapy: a Review of the Evidence. Oncology, Williston Park,, 2015, 29:657–66. Chen AM, Meric-Bernstam F, Hunt KK, Thames HD, Oswald MJ, Outlaw ED, et al. Breast Conservation after Neoadjuvant Chemotherapy: the MD Anderson Cancer Center Experience. J Clin Oncol, 2004, 22:2303–12., DOI 10. 1200/jco.2004.09.062 Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological Complete Response and Long-Term Clinical Benefit in Breast Cancer: the CTNeoBC Pooled Analysis. The Lancet, 2014, 384:164–72., DOI 10.1016/s0140-6736(13)62422-8 Fowble BL, Einck JP, Kim DN, McCloskey S, Mayadev J, Yashar C, et al. Role of Postmastectomy Radiation after Neoadjuvant Chemotherapy in Stage IIIII Breast Cancer. Int J Radiat Oncol Biol Phys, 2012, 83:494–503., DOI 10. 1016/j.ijrobp.2012.01.068 Garg AK, Buchholz TA. Influence of Neoadjuvant Chemotherapy on Radiotherapy for Breast Cancer. Ann Surg Oncol, 2015, 22:1434–40., DOI 10.1245/s10434-015-4402-x Garg AK, Strom EA, McNeese MD, Buzdar AU, Hortobagyi GN, Kuerer HM, et al. T3 Disease at Presentation or Pathologic Involvement of Four or More Lymph Nodes Predict for Locoregional Recurrence in Stage II Breast Cancer Treated with Neoadjuvant Chemotherapy and Mastectomy without Radiotherapy. Int J Radiat Oncology*Biology*Physics, 2004, 59:138–45., DOI 10.1016/j.ijrobp.2003.10.037 Gnant M, Thomssen C, Harbeck N. St. Gallen/Vienna 2015: A Brief Summary of the Consensus Discussion. Breast Care, Basel,, 2015, 10: 124–30., DOI 10.1159/000430488 Huang EH, Tucker SL, Strom EA, McNeese MD, Kuerer HM, Buzdar AU, et al. Postmastectomy Radiation Improves Local-Regional Control and Survival for Selected Patients with Locally Advanced Breast Cancer Treated with Neoadjuvant Chemotherapy and Mastectomy. J Clin Oncol, 2004, 22:4691–9., DOI 10.1200/jco.2004.11.129 Huang EH, Tucker SL, Strom EA, McNeese MD, Kuerer HM, Hortobagyi GN, et al. Predictors of Locoregional Recurrence in Patients with Locally Advanced Breast Cancer Treated with Neoadjuvant Chemotherapy, Mastectomy, and Radiotherapy. Int J Radiat Oncology*Biology*Physics, 2005, 62:351–7., DOI 10.1016/j.ijrobp.2004.09.056 Mamounas EP, Anderson SJ, Dignam JJ, Bear HD, Julian TB, Geyer CE, et al. Predictors of Locoregional Recurrence after Neoadjuvant Chemotherapy: Results from Combined Analysis of National Surgical Adjuvant Breast and Bowel Project B-18 and B-27. J Clin Oncol, 2012, 30:3960–6., DOI 10.1200/jco. 2011.40.8369 McGuire SE, Gonzalez-Angulo AM, Huang EH, Tucker SL, Kau SWC, Yu TK, et al. Postmastectomy Radiation Improves the Outcome of Patients with Locally Advanced Breast Cancer Who Achieve a Pathologic Complete Response to Neoadjuvant Chemotherapy. Int J Radiat Oncology*Biology*Physics, 2007, 68:1004–9., DOI 10.1016/j.ijrobp.2007.01.023 Bukovszky B, Fodor J, Zongor Z, Mihaly D, Matrai Z, Polgar C, et al. [Dose Coverage of Axillary Target Volumes Using Different Field Arrangements Following Breast Conserving Surgery for Invasive Breast Cancer]. Magy Onkol, 2019, 63:102–9. Major T, Gutierrez C, Guix B, van Limbergen E, Strnad V, Polgar C, et al. Recommendations from GEC ESTRO Breast Cancer Working Group, II): Target Definition and Target Delineation for Accelerated or Boost Partial Breast Irradiation Using Multicatheter Interstitial Brachytherapy after Breast Conserving Open Cavity Surgery. Radiother Oncol, 2016, 118:199–204., DOI 10.1016/j.radonc.2015.12.006 Darby SC, Ewertz M, McGale P, Bennet AM, Blom-Goldman U, Bronnum D, et al. Risk of Ischemic Heart Disease in Women after Radiotherapy for Breast Cancer. N Engl J Med, 2013, 368:987–98., DOI 10.1056/nejmoa1209825 Kahan Z, Csenki M, Varga Z, Szil E, Cserhati A, Balogh A, et al. The Risk of Early and Late Lung Sequelae after Conformal Radiotherapy in Breast Cancer Patients. Int J Radiat Oncol Biol Phys, 2007, 68:673–81., DOI 10.1016/j.ijrobp. 2006.12.016 Zs K, Varga Z, Csenki M, Szabo J, Szil E, Fekete G, et al. Torekves a Sugarterapia Individualizalasara Emlorakban: Egyeni Rizikobecsles Es Egyenileg Alkalmazott Technikak., Approaches to Individual Radiotherapy in Breast Cancer: Individual Risk Estimation and Individualized Techniques). Orv Hetil, 2007, 148:833–41. Zs K, Janvary L. Radiotherapy of Breast Cancer: Risk of Damage to Normal Tissues. In: Zs K, editor. Technical Development and Clinical Practice Expert Committee of the Hungarian Society for Radiation Oncology – Damage to normal Tissues during Radiotherapy: Tolerance and Risk Reduction – Guidelines Based on Scientific Evidence, 2007). p. 16–26. Taylor CW, Wang Z, Macaulay E, Jagsi R, Duane F, Darby SC. Exposure of the Heart in Breast Cancer Radiation Therapy: A Systematic Review of Heart Doses Published during 2003 to 2013. Int J Radiat Oncol Biol Phys, 2015, 93: 845–53., DOI 10.1016/j.ijrobp.2015.07.2292 Nissen HD, Appelt AE. Improved Heart, Lung and Target Dose with Deep Inspiration Breath Hold in a Large Clinical Series of Breast Cancer Patients. Radiother Oncol, 2013, 106:28–32., DOI 10.1016/j.radonc.2012.10.016 Varga Z, Cserhati A, Rarosi F, Boda K, Gulyas G, Egyud Z, et al. Individualized Positioning for Maximum Heart protection during Breast Irradiation. Acta Oncol, 2014, 53:58–64., DOI 10.3109/0284186X.2013.781674 Varga Z, Hideghety K, Mezo T, Nikolenyi A, Thurzo L, Kahan Z. Individual Positioning: a Comparative Study of Adjuvant Breast Radiotherapy in the Prone versus Supine Position. Int J Radiat Oncol Biol Phys, 2009, 75:94–100., DOI 10.1016/j.ijrobp.2008.10.045 Piroth MD, Baumann R, Budach W, Dunst J, Feyer P, Fietkau R, et al. Heart Toxicity from Breast Cancer Radiotherapy: Current Findings, Assessment, and Prevention. Strahlenther Onkol, 2019, 195:1–12., DOI 10.1007/s00066- 018-1378-z Ranger A, Dunlop A, Dunlop A, Convery H, Maclennan MK, Twyman N, et al. A Dosimetric Comparison of Breast Radiotherapy Techniques to Treat Locoregional Lymph Nodes Including the Internal Mammary Chain. Clin Oncol, R Coll Radiol, 2018, 30:346–53., DOI 10.1016/j.clon.2018.01.017 Moiseenko V, Einck J, Murphy J, Oden J, Bjohle J, Uzan J, et al. Clinical Evaluation of QUANTEC Guidelines to Predict the Risk of Cardiac Mortality in Breast Cancer Patients. Acta Oncol, 2016, 55:1506–10., DOI 10.1080/ 0284186x.2016.1234067 Taylor CW, Kirby AM. Cardiac Side-Effects from Breast Cancer Radiotherapy. Clin Oncol, R Coll Radiol, 2015, 27:621–9., DOI 10.1016/j. clon.2015.06.007 Mukesh MB, Barnett GC, Wilkinson JS, Moody AM, Wilson C, Dorling L, et al. Randomized Controlled Trial of Intensity-Modulated Radiotherapy for Early Breast Cancer: 5-year Results Confirm superior Overall Cosmesis. J Clin Oncol, 2013, 31:4488–95., DOI 10.1200/jco.2013.49.7842 Bartelink H, Maingon P, Poortmans P, Weltens C, Fourquet A, Jager J, et al. Whole-breast Irradiation with or without a Boost for Patients Treated with Breast-Conserving Surgery for Early Breast Cancer: 20-year Follow-Up of a Randomised Phase 3 Trial. Lancet Oncol, 2015, 16:47–56., DOI 10.1016/s1470- 2045(14)71156-8 Kindts I, Laenen A, Depuydt T,Weltens C. Tumour Bed Boost Radiotherapy for Women after Breast-Conserving Surgery. Cochrane Database Syst Rev, 2017, 11:CD011987., DOI 10.1002/14651858.CD011987.pub2 Polo A, Polgar C, Hannoun-Levi JM, Guinot JL, Gutierrez C, Galalae R, et al. Risk Factors and State-Of-The-Art Indications for Boost Irradiation in Invasive Breast Carcinoma. Brachytherapy, 2017, 16:552–64., DOI 10.1016/j. brachy.2017.03.003 Major T, Gutierrez C, Guix B, Mozsa E, Hannoun-Levi JM, Lossl K, et al. Interobserver Variations of Target Volume Delineation in Multi-Catheter Partial Breast Brachytherapy after Open Cavity Surgery. Brachytherapy, 2015, 14:925–32., DOI 10.1016/j.brachy.2015.06.008 Petersen RP, Truong PT, Kader HA, Berthelet E, Lee JC, Hilts ML, et al. Target Volume Delineation for Partial Breast Radiotherapy Planning: Clinical Characteristics Associated with Low Interobserver Concordance. Int J Radiat Oncol Biol Phys, 2007, 69:41–8., DOI 10.1016/j.ijrobp.2007.01.070 Olivotto IA, Whelan TJ, Parpia S, Kim DH, Berrang T, Truong PT, et al. Interim Cosmetic and Toxicity Results from RAPID: A Randomized Trial of Accelerated Partial Breast Irradiation Using Three-Dimensional Conformal External Beam Radiation Therapy. J Clin Oncol, 2013, 31:4038–45., DOI 10. 1200/jco.2013.50.5511 Vaidya JS, Wenz F, Bulsara M, Tobias JS, Joseph DJ, Keshtgar M, et al. Riskadapted Targeted Intraoperative Radiotherapy versus Whole-Breast Radiotherapy for Breast Cancer: 5-year Results for Local Control and Overall Survival from the TARGIT-A Randomised Trial. The Lancet, 2014, 383:603–13., DOI 10.1016/s0140-6736(13)61950-9 Veronesi U, Orecchia R, Maisonneuve P, Viale G, Rotmensz N, Sangalli C, et al. Intraoperative Radiotherapy versus External Radiotherapy for Early Breast Cancer, ELIOT): a Randomised Controlled Equivalence Trial. Lancet Oncol, 2013, 14:1269–77., DOI 10.1016/s1470-2045(13)70497-2 Offersen BV, Boersma LJ, Kirkove C, Hol S, Aznar MC, Sola AB, et al. ESTRO Consensus Guideline on Target Volume Delineation for Elective Radiation Therapy of Early Stage Breast Cancer, Version 1.1. Radiother Oncol, 2016, 118:205–8., DOI 10.1016/j.radonc.2015.12.027 Csenki M, Ujhidy D, Cserhati A, Cserhati A, Kahan Z, Varga Z. Radiation Dose to the Nodal Regions during Prone versus Supine Breast Irradiation. Ther Clin Risk Manag, 2014, 10:367., DOI 10.2147/TCRM.S59483 Offersen BV, Boersma LJ, Kirkove C, Hol S, Aznar MC, Biete Sola A, et al. ESTRO Consensus Guideline on Target Volume Delineation for Elective Radiation Therapy of Early Stage Breast Cancer. Radiother Oncol, 2015, 114: 3–10., DOI 10.1016/j.radonc.2014.11.030 Nielsen MH, Berg M, Pedersen AN, Andersen K, Glavicic V, Jakobsen EH, et al. Delineation of Target Volumes and Organs at Risk in Adjuvant Radiotherapy of Early Breast Cancer: National Guidelines and Contouring Atlas by the Danish Breast Cancer Cooperative Group. Acta Oncol, 2013, 52: 703–10., DOI 10.3109/0284186x.2013.765064 Verhoeven K, Weltens C, Remouchamps V, Mahjoubi K, Veldeman L, Lengele B, et al. Vessel Based Delineation Guidelines for the Elective Lymph Node Regions in Breast Cancer Radiation Therapy – PROCAB Guidelines. Radiother Oncol, 2015, 114:11–6., DOI 10.1016/j.radonc.2014. 11.008 Li XA, Tai A, Arthur DW, Buchholz TA, Macdonald S, Marks LB, et al. Variability of Target and normal Structure Delineation for Breast Cancer Radiotherapy: an RTOG Multi-Institutional and Multiobserver Study. Int J Radiat Oncology*Biology*Physics, 2009, 73:944–51., DOI 10.1016/j.ijrobp. 2008.10.034 Omlin A, Amichetti M, Azria D, Cole BF, Fourneret P, Poortmans P, et al. Boost Radiotherapy in Young Women with Ductal Carcinoma In Situ: a Multicentre, Retrospective Study of the Rare Cancer Network. Lancet Oncol, 2006, 7:652–6., DOI 10.1016/s1470-2045(06)70765-3 Brunt AM, Haviland JS, Wheatley DA, Sydenham MA, Alhasso A, Bloomfield DJ, et al. Hypofractionated Breast Radiotherapy for 1 Week versus 3 Weeks, FAST-Forward): 5-year Efficacy and Late normal Tissue Effects Results from a Multicentre, Non-inferiority, Randomised, Phase 3 Trial. The Lancet, 2020, 395:1613–26., DOI 10.1016/s0140-6736(20, 30932-6 Polgar C, Major T. Current Status and Perspectives of Brachytherapy for Breast Cancer. Int J Clin Oncol, 2009, 14:7–24., DOI 10.1007/s10147-008- 0867-y Romestaing P, Lehingue Y, Carrie C, CoquaRd R, Montbarbon X, Ardiet JM, et al. Role of a 10-Gy Boost in the Conservative Treatment of Early Breast Cancer: Results of a Randomized Clinical Trial in Lyon, France. J Clin Oncol, 1997, 15:963–8., DOI 10.1200/jco.1997.15.3.963 Zhu L, Jin L, Li S, Chen K, Jia W, Shan Q, et al. Which Nomogram Is Best for Predicting Non-sentinel Lymph Node Metastasis in Breast Cancer Patients? A Meta-Analysis. Breast Cancer Res Treat, 2013, 137:783–95., DOI 10.1007/ s10549-012-2360-6 Budach W, Bolke E, Kammers K, Gerber PA, Nestle-Kramling C, Matuschek C. Adjuvant Radiation Therapy of Regional Lymph Nodes in Breast Cancer – a Meta-Analysis of Randomized Trials – an Update. Radiat Oncol, 2015, 10: 258., DOI 10.1186/s13014-015-0568-4 Thorsen LBJ, Offersen BV, Dano H, Berg M, Jensen I, Pedersen AN, et al. DBCG-IMN: A Population-Based Cohort Study on the Effect of Internal Mammary Node Irradiation in Early Node-Positive Breast Cancer. J Clin Oncol, 2016, 34:314–20., DOI 10.1200/jco.2015.63.6456 Kantor O, Pesce C, Singh P, Miller M, Tseng J, Wang CH, et al. Post-mastectomy Radiation Therapy and Overall Survival after Neoadjuvant Chemotherapy. J Surg Oncol, 2017, 115:668–76., DOI 10.1002/ jso.24551 Currey A, Patten CR, Bergom C, Wilson JF, Kong AL. Management of the Axilla after Neo-Adjuvant Chemotherapy for Breast Cancer: Sentinel Node Biopsy and Radiotherapy Considerations. Breast J, 2018, 24:902–10., DOI 10. 1111/tbj.13116 Bristol IJ, Woodward WA, Strom EA, Cristofanilli M, Domain D, Singletary SE, et al. Locoregional Treatment Outcomes after Multimodality Management of Inflammatory Breast Cancer. Int J Radiat Oncol Biol Phys, 2008, 72:474–84., DOI 10.1016/j.ijrobp.2008.01.039 Brito RA, Valero VV, Buzdar AU, Booser DJ, Ames F, Strom E, et al. Longterm Results of Combined-Modality Therapy for Locally Advanced Breast Cancer with Ipsilateral Supraclavicular Metastases: The University of Texas M.D. Anderson Cancer Center Experience. J Clin Oncol, 2001, 19:628–33., DOI 10.1200/jco.2001.19.3.628 Chia S, Swain SM, Byrd DR, Mankoff DA. Locally Advanced and Inflammatory Breast Cancer. J Clin Oncol, 2008, 26:786–90., DOI 10.1200/ jco.2008.15.0243 Montagna E, Bagnardi V, Rotmensz N, Rodriguez J, Veronesi P, Luini A, et al. Factors that Predict Early Treatment Failure for Patients with Locally Advanced, T4, Breast Cancer. Br J Cancer, 2008, 98:1745–52., DOI 10. 1038/sj.bjc.6604384 Woodward WA, Buchholz TA. The Role of Locoregional Therapy in Inflammatory Breast Cancer. Semin Oncol, 2008, 35:78–86., DOI 10.1053/j. seminoncol.2007.11.009 Fodor J, Gulyas G, Polgar C, Major T, Szabo E, Koves I, et al. Radiotherapy and Delayed Breast Reconstruction with Implant: Examination of Compatibility]. Magy Onkol, 2002, 46:323–6. Fodor J, Gulyas G, Polgar C, Major T, Kasler M. [Radiotherapy and Breast Reconstruction: the Issue of Compatibility]. Orv Hetil, 2003, 144:549–55. Adamovicz K, Marczewska M, Jassem J. Combining Systemic Therapies with Radiation in Breast Cancer. Cancer Treat Rev, 2009, 35:409–16., DOI 10.1016/ j.ctrv.2009.04.012 Varga Z, Cserhati A, Kelemen G, Boda K, Thurzo L, Kahan Z. Role of Systemic Therapy in the Development of Lung Sequelae after Conformal Radiotherapy in Breast Cancer Patients. Int J Radiat Oncol Biol Phys, 2011, 80:1109–16., DOI 10.1016/j.ijrobp.2010.03.044 Marinko T, Dolenc J, Biban-Jakopin C. Cardiotoxicity of Concomitant Radiotherapy and Trastuzumab for Early Breast Cancer. Radiol Oncol, 2014, 48:105–12., DOI 10.2478/raon-2013-0040 Cecchini MJ, Yu E, Potvin K, D’souza D, Lock M. Concurrent or Sequential Hormonal and Radiation Therapy in Breast Cancer: A Literature Review. Cureus, 2015, 7:e364., DOI 10.7759/cureus.364 Azria D, Gourgou S, Sozzi WJ, ZouhAir A, Mirimanoff RO, Mirimanoff RO, et al. Concomitant Use of Tamoxifen with Radiotherapy Enhances Subcutaneous Breast Fibrosis in Hypersensitive Patients. Br J Cancer, 2004, 91:1251–60., DOI 10.1038/sj.bjc.6602146 Masinghe SP, Faluyi OO, Kerr GR, Kunkler IH. Breast Radiotherapy for Occult Breast Cancer with Axillary Nodal Metastases – Does it Reduce the Local Recurrence Rate and Increase Overall Survival? Clin Oncol, R Coll Radiol, 2011, 23:95–100., DOI 10.1016/j.clon.2010.10.001 Tan BY, Acs G, Apple SK, Badve S, Bleiweiss IJ, Brogi E, et al. Phyllodes Tumours of the Breast: a Consensus Review. Histopathology, 2016, 68:5–21., DOI 10.1111/his.12876 Lahat G, Lev D, Gerstenhaber F, Madewell J, Le-Petross H, Pollock RE. Sarcomas of the Breast. Expert Rev Anticancer Ther, 2012, 12:1045–51., DOI 10.1586/era.12.82 Depla AL, Scharloo-Karels CH, de Jong MA, Oldenborg S, Kolff MW, Oei SB, et al. Treatment and Prognostic Factors of Radiation-Associated Angiosarcoma, RAAS, after Primary Breast Cancer: a Systematic Review. Eur J Cancer, 2014, 50:1779–88., DOI 10.1016/j.ejca.2014.03.002 Fodor J, Orosz Z, Szabo E, Sulyok Z, Polgar C, Zaka Z, et al. Angiosarcoma after Conservation Treatment for Breast Carcinoma: Our Experience and a Review of the Literature. J Am Acad Dermatol, 2006, 54:499–504., DOI 10. 1016/j.jaad.2005.10.017 Shickman R, Leibman AJ, Handa P, Kornmehl A, Abadi M. Mesenchymal Breast Lesions. Clin Radiol, 2015, 70:567–75., DOI 10.1016/j.crad.2014.12.015 Fodor J, Major T, Polgar C, OrosZ Z, Sulyok Z, Kasler M. Prognosis of Patients with Local Recurrence after Mastectomy or Conservative Surgery for Early-Stage Invasive Breast Cancer. The Breast, 2008, 17:302–8., DOI 10.1016/ j.breast.2007.11.004 Arthur DW, Winter KA, Kuerer HM, Haffty B, Cuttino L, Todor DA, et al. Effectiveness of Breast-Conserving Surgery and 3-dimensional Conformal Partial Breast Reirradiation for Recurrence of Breast Cancer in the Ipsilateral Breast: The NRG Oncology/RTOG 1014 Phase 2 Clinical Trial. JAMA Oncol, 2019, 6:75–82., DOI 10.1001/jamaoncol.2019.4320 Hannoun-Levi JM, Resch A, Kauer-Dorner D, Strnad V, Niehoff P, Loessl K, et al. Accelerated Partial Breast Irradiation with Interstitial Brachytherapy as Second Conservative Treatment for Ipsilateral Breast Tumour Recurrence: Multicentric Study of the GEC-ESTRO Breast Cancer Working Group. Radiother Oncol, 2013, 108:226–31., DOI 10.1016/j.radonc.2013.03.026 Kuerer HM, Arthur DW, Haffty BG. Repeat Breast-Conserving Surgery for In-Breast Local Breast Carcinoma Recurrence: the Potential Role of Partial Breast Irradiation. Cancer, 2004, 100:2269–80., DOI 10.1002/cncr.20257 Polgar C, Major T, Sulyok Z, Frohlich G, Szabo E, Savolt A, et al. Second Breast-Conserving Surgery and Reirradiation with Interstitial High-Dose- Rate Brachytherapy for the Management of Intra-breast Recurrences -- 5- year Results. Magy Onkol, 2012, 56:68–74. Mayer A, Naszaly A, Patyanik M, Zarand P, Polgar I, Klinko T. Perioperative Brachytherapy for Pretreated Chest wall Recurrence of Breast Cancer. Strahlenther Onkol, 2002, 178:633–6., DOI 10.1007/ s00066-002-0933-8 Smanyko V, Meszaros N, Ujhelyi M, Frohlich G, Stelczer G, Major T, et al. Second Breast-Conserving Surgery and Interstitial Brachytherapy vs. Salvage Mastectomy for the Treatment of Local Recurrences: 5-year Results. Brachytherapy, 2019, 18:411–9., DOI 10.1016/j.brachy.2019.02.004 Polgar C. A Palliativ Sugarkezeles Indikacioi., Indications for Palliative Radiotherapy. LAM, 2003, 13:373–8. Sahgal A, Aoyama A, Kocher M, Neupane B, Collette S, Tago M, et al. Phase 3 Trials of Stereotactic Radiosurgery with or without Whole-Brain Radiation Therapy for 1 to 4 BrainMetastases: Individual Patient DataMeta-Analysis. Int J Radiat Oncol Biol Phys, 2015, 91:710–7., DOI 10.1016/j.ijrobp.2014.10.024 Forrai G, Kovacs E, Ambrozay E, Barta M, Bobely K, Lengyel Z, et al. Use of Diagnostic Imaging Modalities in Modern Screening, Diagnostics and Management of Breast Tumours 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022, 28:1610382., DOI 10.3389/pore.2022.1610382 Cserni G, Francz M, Jaray B, Kalman E, Kovacs I, Krenacs T, et al. Pathological Diagnosis, Work-Up and Reporting of Breast Cancer 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, Forthcoming 2022)., DOI 10.3389/pore.2022.1610373 Matrai Z, Kelemen P, Kosa C, Maraz R, Paszt A, Pavlovics G, et al. Modern Breast Cancer Surgery. 1stCentral-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022, 28:1610377., DOI 10.3389/pore.2022.1610377 Rubovszky G, Kocsis J, Boer K, Chilingirova N, Dank M, Kahan Z, et al. Systemic Treatment of Breast Cancer. 1st Central-eastern Eur Prof Consensus Statement Breast Cancer Pathol Oncol Res, Forthcoming 2022)., DOI 10.3389/ pore.2022.1610383 Kahan Z, Szanto I, Dudas R, Kapitany Z, Molnar M, Koncz Z, et al. Breast Cancer: Follow-Up, Rehabilitation, Psycho-Oncology. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022, 28:1610391., DOI 10.3389/pore.2022.1610391 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610378 EP 1610392 DI 10.3389/pore.2022.1610378 PG 15 ER PT J AU Forrai, G Kovacs, E Ambrozay, Barta, M Borbely, K Lengyel, Zs Ormandi, K Pentek, Z Tunde, T Eva, S AF Forrai, Gabor Kovacs, Eszter Ambrozay, Eva Barta, Miklos Borbely, Katalin Lengyel, Zsolt Ormandi, Katalin Pentek, Zoltan Tunde, Tasnadi Eva, Sebo TI Use of Diagnostic Imaging Modalities in Modern Screening, Diagnostics and Management of Breast Tumours 1st Central-Eastern European Professional Consensus Statement on Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Guideline DE mammography; breast ultrasound; breast MRI; breast screening; conventional nuclear medicine; SPECT/CT; PET/CT; biopsy ID mammography; breast ultrasound; breast MRI; breast screening; conventional nuclear medicine; SPECT/CT; PET/CT; biopsy AB Breast radiologists and nuclear medicine specialists updated their previous recommendation/guidance at the 4th Hungarian Breast Cancer Consensus Conference in Kecskemet. A recommendation is hereby made that breast tumours should be screened, diagnosed and treated according to these guidelines. These professional guidelines include the latest technical developments and research findings, including the role of imaging methods in therapy and follow-up. It includes details on domestic development proposals and also addresses related areas (forensic medicine, media, regulations, reimbursement). The entire material has been agreed with the related medical disciplines. C1 [Forrai, Gabor] GE-RAD Kft.Budapest, Hungary. [Kovacs, Eszter] GE-RAD Kft.Budapest, Hungary. [Ambrozay, Eva] MaMMa Egeszsegugyi RtBudapest, Hungary. [Barta, Miklos] Royal Cornwall HospitalTruro, UK. [Borbely, Katalin] National Institute of OncologyBudapest, Hungary. [Lengyel, Zsolt] Hamad Medical CorporationDoha, Qatar. [Ormandi, Katalin] University of SzegedSzeged, Hungary. [Pentek, Zoltan] MaMMa Egeszsegugyi RtBudapest, Hungary. [Tunde, Tasnadi] Rethy Pal HospitalBekescsaba, Hungary. [Eva, Sebo] Kenezy Teaching HospitalDebrecen, Hungary. RP Forrai, G (reprint author), GE-RAD Kft., Budapest, Hungary. EM drforraigabor@gmail.com CR Karsa L, Holland R, Broeders M, Wolf C, Perry N, Tornberg S. European Guidelines for Quality Assurance in Breast Cancer Screening and Diagnosis. 4th ed. Publications Office Luxembourg, 2013). Tabar L, Yen M-F, Vitak B, Chen H-HT, Smith RA, Duffy SW. Mammography Service Screening and Mortality in Breast Cancer Patients: 20-year Follow-Up before and after Introduction of Screening. Lancet, 2003, 361:1405–10., DOI 10.1016/s0140-6736(03)13143-1 Yang WT, Lai C-J, Whitman GJ, Murphy WA, Dryden MJ, Kushwaha AC, et al. Comparison of Full-Field Digital Mammography and Screen-Film Mammography for Detection and Characterization of Simulated Small Masses. Am J Roentgenol, 2006, 187:W576–W581., DOI 10.2214/ajr.05.0126 Skaane P, Bandos AI, Eben EB, Jebsen IN, Krager M, Haakenaasen U, et al. Two-view Digital Breast Tomosynthesis Screening with Synthetically Reconstructed Projection Images: Comparison with Digital Breast Tomosynthesis with Full-Field Digital Mammographic Images. Radiology, 2014, 271:655–63., DOI 10.1148/radiol.13131391 McDonald ES, Oustimov A, Weinstein SP, Synnestvedt MB, Schnall M, Conant EF. Effectiveness of Digital Breast Tomosynthesis Compared with Digital Mammography: Outcomes Analysis from 3 Years of Breast Cancer Screening. JAMA Oncol, 2016, 2:737–43., DOI 10.1001/jamaoncol.2015.5536 Dromain C, Balleyguier C, Adler G, Garbay JR, Delaloge S. Contrastenhanced Digital Mammography. Eur J Radiol, 2009, 69:34–42., DOI 10. 1016/j.ejrad.2008.07.035 Kim EY, Youn I, Lee KH, Yun J-S, Park YL, Park CH, et al. Diagnostic Value of Contrast-Enhanced Digital Mammography versus Contrast-Enhanced Magnetic Resonance Imaging for the Preoperative Evaluation of Breast Cancer. J Breast Cancer, 2018, 21:453–62., DOI 10.4048/jbc.2018.21.e62 Perry H, Phillips J, Dialani V, Slanetz PJ, Fein-Zachary VJ, Karimova EJ, et al. Contrast-enhanced Mammography: A Systematic Guide to Interpretation and Reporting. Am J Roentgenol, 2019, 212:222–31., DOI 10.2214/ajr.17.19265 Fallenberg EM, Dromain C, Diekmann F, Engelken F, Krohn M, Singh JM, et al. Contrast-enhanced Spectral Mammography versus MRI: Initial Results in the Detection of Breast Cancer and Assessment of Tumour Size. Eur Radiol, 2014, 24:256–64., DOI 10.1007/s00330-013-3007-7 Chou C-P, Lewin JM, Chiang C-L, Hung B-H, Yang T-L, Huang J-S, et al. Clinical Evaluation of Contrast-Enhanced Digital Mammography and Contrast Enhanced Tomosynthesis-Comparison to Contrast-Enhanced Breast MRI. Eur J Radiol, 2015, 84:2501–8., DOI 10.1016/j.ejrad.2015.09.019 Magyar Radiologusok Tarsasaga Emlodiagnosztikai Szekcio es Ultrahang Szekcio. Kozos Allasfoglalas Az Emlo Ultrahangvizsgalatarol. Magy Radiol, 2001). [Hungarian Society of Radiologists, Breast Diagnostic Section and Ultrasound Section, Joint Resolution on Breast Ultrasound]. Berg WA, Blume JD, Cormack JB, Mendelson EB, Lehrer D, Bohm-Velez M, et al. Combined Screening with Ultrasound and Mammography vs Mammography Alone in Women at Elevated Risk of Breast Cancer. JAMA, 2008, 299:2151–63., DOI 10.1001/jama.299.18.2151 Lander MR, Tabar L. Automated 3-D Breast Ultrasound as a Promising Adjunctive Screening Tool for Examining Dense Breast Tissue. Semin Roentgenol, 2011, 46:302–8., DOI 10.1053/j.ro.2011.06.003 Brem RF, Tabar L, Duffy SW, Inciardi MF, Guingrich JA, Hashimoto BE, et al. Assessing Improvement in Detection of Breast Cancer with Three- Dimensional Automated Breast US in Women with Dense Breast Tissue: the SomoInsight Study. Radiology, 2015, 274:663–73., DOI 10.1148/radiol. 14132832 Karst I, Henley C, Gottschalk N, Floyd S, Mendelson EB. Three-dimensional Automated Breast US: Facts and Artifacts. RadioGraphics, 2019, 39:913–31., DOI 10.1148/rg.2019180104 Nazari SS, Mukherjee P. An Overview of Mammographic Density and its Association with Breast Cancer. Breast Cancer, 2018, 25:259–67., DOI 10. 1007/s12282-018-0857-5 Emons J, Wunderle M, Hartmann A, Radicke M, Rauh C, Uder M, et al. Initial Clinical Results with a Fusion Prototype for Mammography and Three-Dimensional Ultrasound with a Standard Mammography System and a Standard Ultrasound Probe. Acta Radiol, 2018, 59:1406–13., DOI 10. 1177/0284185118762249 Schaefgen B, Heil J, Barr RG, Radicke M, Harcos A, Gomez C, et al. Initial Results of the FUSION-X-US Prototype Combining 3D Automated Breast Ultrasound and Digital Breast Tomosynthesis. Eur Radiol, 2018, 28: 2499–506., DOI 10.1007/s00330-017-5235-8 Berg WA, Leung JWT. Diagnostic Imaging: Breast. 3nd ed. Altona, Manitoba, Canada: Elsevier Science Health Science, 2019). Barr RG, Nakashima K, Amy D, Cosgrove D, Farrokh A, Schafer F, et al. WFUMB Guidelines and Recommendations for Clinical Use of Ultrasound Elastography: Part 2: Breast. Ultrasound Med Biol, 2015, 41:1148–60., DOI 10. 1016/j.ultrasmedbio.2015.03.008 Borbola GY, Kardos K, Tasnadi T. Az Emlo Betegsegeinek Vizsgalata Szonoelasztografiaval. Kezdeti Tapasztalatok. Magy Radiol, 2008, 82: 27–33. [Assessment of breast diseases by sonoelastography. Initial experience]. Sidhu PS, Cantisani V, Dietrich CF, Gilja OH, Saftoiu A, Bartels E, et al. The EFSUMB Guidelines and Recommendations for the Clinical Practice of Contrast-Enhanced Ultrasound, CEUS, in Non-hepatic Applications: Update 2017, Long Version). Ultraschall Med, 2018, 39:e2–44., DOI 10. 1055/a-0586-1107 ACR. ACR BI-RADS Atlas. 5th ed. American Collage of Radiology, 2013). Sardanelli F, Boetes C, Borisch B, Decker T, Federico M, Gilbert FJ, et al. Magnetic Resonance Imaging of the Breast: Recommendations from the EUSOMA Working Group. Eur J Cancer, 2010, 46:1296–316., DOI 10.1016/j. ejca.2010.02.015 Biglia N, Bounous VE, Martincich L, Panuccio E, Liberale V, Ottino L, et al. Role of MRI, Magnetic Resonance Imaging, versus Conventional Imaging for Breast Cancer Presurgical Staging in Young Women or with Dense Breast. Eur J Surg Oncol, 2011, 37:199–204., DOI 10.1016/j.ejso. 2010.12.011 Kuhl CK. High-risk Screening: Multi-Modality Surveillance of Women at High Risk for Breast Cancer, Proven or Suspected Carriers of a Breast Cancer Susceptibility Gene). J Exp Clin Cancer Res, 2002, 21: 103–6. Mann RM, Balleyguier C, Balleyguier C, Baltzer PA, Bick U, Colin C, et al. Breast MRI: EUSOBI Recommendations for Women’s Information. Eur Radiol, 2015, 25:3669–78., DOI 10.1007/s00330-015-3807-z Berger N, Luparia A, Di Leo G, Carbonaro LA, Trimboli RM, Ambrogi F, et al. Diagnostic Performance of MRI versus Galactography in Women with Pathologic Nipple Discharge: A Systematic Review and Meta-Analysis. Am J Roentgenol, 2017, 209:465–71., DOI 10.2214/ajr.16.16682 Lee JH, Rosen EL, Mankoff DA. The Role of Radiotracer Imaging in the Diagnosis and Management of Patients with Breast Cancer: Part 1-Overview, Detection, and Staging. J Nucl Med, 2009, 50:569–81., DOI 10.2967/jnumed. 108.053512 Lee JH, Rosen EL, Mankoff DA. The Role of Radiotracer Imaging in the Diagnosis and Management of Patients with Breast Cancer: Part 2-Response to Therapy, Other Indications, and Future Directions. J Nucl Med, 2009, 50: 738–48., DOI 10.2967/jnumed.108.061416 Podoloff DA, Advani RH, Allred C, Benson AB, Brown E, Burstein HJ, et al. NCCN Task Force Report: Positron Emission Tomography, PET)/computed Tomography, CT, Scanning in Cancer. J Natl Compr Canc Netw, 2007, 5(Suppl. 1):S1–2., DOI 10.6004/jnccn.2007.2001 Lavayssiere R, Cabee AE, Filmont JE. Positron Emission Tomography, PET, and Breast Cancer in Clinical Practice. Eur J Radiol, 2009, 69(50–58):50–8., DOI 10.1016/j.ejrad.2008.07.039 Poeppel TD, Krause BJ, Heusner TA, Boy C, Bockisch A, Antoch G. PET/CT for the Staging and Follow-Up of Patients with Malignancies. Eur J Radiol, 2009, 70:382–92., DOI 10.1016/j.ejrad.2009.03.051 Borbely K, Szilagyi I, Kasler M. IV. PET/CT Multidiszciplinaris Nemzeti Konszenzus Konferencia Allasfoglalasa. [Resolution of the 4th National PET/ CT Multidisciplinary Consensus Conference]. Magy Onkol, 2011, 55:117–27. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Breast Cancer Screening and Diagnosis Guidelines, Breast Cancer Version 3, 2019). Available at: https://www2.tri-kobe.org/nccn/ guideline/archive/breast2019/english/breast_v3.pdf. Pan H, Gray R, Braybrooke J, Davies C, Taylor C, McGale P, et al. 20-year Risks of Breast-Cancer Recurrence after Stopping Endocrine Therapy at 5 Years. N Engl J Med, 2017, 377:1836–46., DOI 10.1056/nejmoa1701830 Borbely K. Ujdonsagok Es Uj Lehetosegek a Nuklearis Medicina Kepalkotasban. [Novelties and New Possibilities in Nuclear Medicine Imaging]. Magy Onkol, 2014, 58:232–8. Forrai G, Ambrozay E, Bidlek M, Miklos B, Katalin B, Zsolt L, et al. A Kepalkoto VIZSGALO MODSZERek Alkalmazasa Az Emlodaganatok Korszeru Szureseben, Diagnosztikajaban Es Ellatasaban – Szakmai Utmutato a III. Emlorak Konszenzus Konferencia Alapjan. [Use of diagnostic imaging MODALITIES in modern screening, diagnostics and management of breast tumours – professional guidelines based on the 3rd Breast Cancer Consensus Conference]. Magy Onkol, 2016, 60: 181–93. Glass SB, Shah ZA. Clinical Utility of Positron Emission Mammography. Baylor Univ Med Cent Proc, 2013, 26:314–9., DOI 10.1080/08998280.2013. 11928996 Borbely K. Ujdonsagok Es Uj Lehetosegek Az Onkologiai Betegek Terapias Vezeteseben: PET/MR Klinikai Alkalmazasok. [Novelties and new possibilities in the therapeutic management of oncology patients: clinical applications of PET/MRI]. Magy Onkol, 2015, 59:10–16. Borbely K. A PET/MR Technologia Jelene Es Jovoje. [Present and future of the PET/MRI technology]. Magy Radiol, 2018, 92:43–52. Bhole S, Neuschler E. MRI-guided Breast Interventions. Appl Radiol, 2015, 44:7–13., DOI 10.37549/ar2218 The UK MammotomeTrial GroupTeh W, Michell MJ,Wilson ARM, Britton P, Shah B. UK National Health Service Breast Screening Programme, NHSBSP, Multicentre Image Guided Biopsy Trial: an Update. Breast Cancer Res, 2002, 4:15., DOI 10.1186/bcr470 Dobrossy L. Szervezett Szures Az Onkologiaban. Budapest: Egeszsegugyi Miniszterium, 2000). [Organized Screening in Oncology, Ministry of Health]. Dobrossy L. Nepegeszsegugyi Onkologiai Szuresek. Public Health Oncology Screenings. Budapest: ANTSZ -National Public Health and Medical Officer Service, 2005). Dobrossy L. Daganatok Szűrese Minősegbiztositasi Keziko€nyv EsModszertani Utmutato. Budapest: Orszagos Tisztifőorvosi Hivatal, 2013). [Tumour Screening-Quality Assurance Manual and Methodological Guide. Budapest: Office of the Chief Medical Officer]. Forrai G, Pemtek Z, Ormandi K. A Mammografias Emloszures Es a Korai Emlorak Diagnosztikajara Es Terapiajara Vonatkozo Protokollok Gyujtemenye. 02. verzio. Budapest: OTH Mammografias Emloszuresi Albizottsag, 2004). [Collection of Protocols for Mammographic Breast Screening and for the Diagnosis and Therapy of Early Breast Cancer, OTH Breast Screening Mammography Subcommittee 2004, Version 02)]. Smith RA, Cokkinides V, Brawley OW. Cancer Screening in the United States, 2009: a Review of Current American Cancer Society Guidelines and Issues in Cancer Screening. CA Cancer J Clin, 2009, 59: 27–41., DOI 10.3322/caac.20008 Saslow D, Boetes C, Burke W, Harms S, Leach MO, Lehman CD, et al. American Cancer Society Guidelines for Breast Screening with MRI as an Adjunct to Mammography. CA Cancer J Clin, 2007, 57:75–89., DOI 10.3322/ canjclin.57.2.75 Sauven P, Bishop H, Patnick J, Walton J, Wheeler E, Lawrence G. The National Health Service Breast Screening Programme and British Association of Surgical Oncology Audit of Quality Assurance in Breast Screening 1996- 2001. Br J Surg, 2003, 90:82–7., DOI 10.1002/bjs.4013 Moss SM, Wale C, Smith R, Evans A, Cuckle H, Duffy SW. Effect of Mammographic Screening from Age 40 Years on Breast Cancer Mortality in the UK Age Trial at 17 years’ Follow-Up: a Randomised Controlled Trial. Lancet Oncol, 2015, 16:1123–32., DOI 10.1016/s1470-2045(15)00128-x Sardanelli F, Fallenberg EM, Fallenberg EM, Clauser P, Trimboli RM, Camps- Herrero J, et al. Mammography: An Update of the EUSOBI Recommendations on Information for Women. Insights Imaging, 2017, 8: 11–8., DOI 10.1007/s13244-016-0531-4 Adany R. Megelozo Orvostan Es Nepegeszsegtan. Budapest: Debreceni Egyetem, 2011). [Preventive Medicine and Public Health, University of Debrecen]. Schneerg T, Mitchell G, Taylor D, Saunders C. MRI Screening for Breast Cancer in Women at High Risk; Is the Australian Breast MRI Screening Access Program Addressing the Needs of Women at High Risk of Breast Cancer? J Med Radiat, 2015, 62:212–25., DOI 10.1002/jmrs.116 Liort G, Chirivella I, Morales R, Serrano R, Sanchez AB, Teule A, et al. SEOM Clinical Guidelines in Hereditary Breast and Ovarian Cancer. On Behalf of the Hereditary Cancer Working Group. Clin Transl Oncol, 2015, 17:956–61., DOI 10.1007/s12094-015-1435-3 Podo F, Sardanelli F, Canese R, D’Agnolo G, Natali PG, Crecco M, et al. The Italian Multi-centre Project on Evaluation of MRI and Other Imaging Modalities in Early Detection of Breast Cancer in Subjects at High Genetic Risk. J Exp Clin Cancer Res, 2002, 21:115–24. Lech MO, Eeles RA, Turnbull LW, Dixon AK, Brown J, Hoff RJ, et al. The UK National Study ofMagnetic Resonance Imaging as aMethod of Screening for Breast Cancer, MARIBS). J Exp Clin Cancer Res, 2002, 21:107–14. NICE. CG41 Familial Breast Cancer Guideline, NICE,, 2006). Available at: http://www.nice.org.uk/guidance/cg164. Sardanelli F, Podo F, D’Agnolo G, Verdecchia A, Santaquilani M, Musumeci R, et al. Multicenter Comparative Multimodality Surveillance of Women at Genetic-Familial High Risk for Breast Cancer, HIBCRIT Study): Interim Results. Radiology, 2007, 242: 698–715., DOI 10.1148/radiol.2423051965 Olah E. Herediter Emlo- Es Petefeszekrak-Szindroma, a Gyanutol a Rizikocsokkentesig. In: Z Matrai, G Gulyas, M Kasler, editors. Az Emlorak Korszeru Sebeszete. Budapest: Medicina Kiado, 2015). p. 389–409. [Olah E. Hereditary Breast and Ovarian Cancer Syndrome, from Suspicion to Risk Reduction. In: Matrai Z, Gulyas G, Kasler M, editors. Modern Surgery of Breast Cancer. Budapest: Medicina Publisher, 2015). p. 389–409]. Olah E. Molekularis Onkogenetika. In: M Kasler, editor. Az Onkologia Alapjai. Budapest: Medicina Kiado, 2011). p. 49–69. [Olah E. Molecular Oncogenetics. In: Kasler M, editor. Basic Oncology. Budapest: Medicina Publisher, 2011). p. 49–69]. Antoniou AC, Cunningham AP, Peto J, Evans DG, Lalloo F, Narod SA, et al. The BOADICEA Model of Genetic Susceptibility to Breast and Ovarian Cancers: Updates and Extensions. Br J Cancer, 2008, 98:1457–66., DOI 10. 1038/sj.bjc.6604305 Paulch-Shimon S, Cardoso F, Partidge AH, Abulkhair O, Azim HA, Bianchi- Micheli G, et al. ESO–ESMO 4th International Consensus Guidelines for Breast Cancer in Young Women, BCY4). Ann Oncol, 2020, 31(6):674–96., DOI 10.1016/j.annonc.2020.03.284 Momenimovahed Z, Taheri TA. Do the Fertility Drugs Increase the Risk of Cancer? A Review Study. Front Endocrinol, 2019, 1–13. Senkus E, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rutgers E, et al. Primary Breast Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol, 2015, 26(Suppl. 5): v8–30., DOI 10.1093/annonc/mdv298 Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, et al. Early Breast Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol, 2019, 30:1194–220., DOI 10.1093/ annonc/mdz173 Slanetz PJ, Freer PE, Birdwell RL. Breast-Density Legislation - Practical Considerations. N Engl J Med, 2015, 372:593–5., DOI 10.1056/nejmp1413728 EUR-Lex. Artificial Intelligence for Europe a Bizottsag Ko€zlemenye Az Europai Parlamentnek, a Tanacsnak, Az Europai Gazdasagi Es Szocialis Bizottsagnak Es a Regiok Bizottsaganak. [Artificial Intelligence for Europe – Communication from the Commission to the European Parliament, the Council, the European Economic and Social Committee and the Committee of the Regions], 2018). Available at: https://eur-lex.europa.eu/legal-content/ HU/TXT/PDF/?uri=CELEX:52018DC0237&from=EN. Lehman CD, Wellman RD, Buist DSM, Kerlikowske K, Tosteson ANA, Miglioretti DL. Diagnostic Accuracy of Digital Screening Mammography with and without Computer-Aided Detection. JAMA Intern Med, 2015, 175: 1828–37., DOI 10.1001/jamainternmed.2015.5231 Gilbert FJ, Astley SM, Gillan MGC, Agbaje OF, Wallis MG, James J, et al. Single reading with Computer-Aided Detection for Screening Mammography. N Engl J Med, 2008, 359:1675–84., DOI 10.1056/ nejmoa0803545 Kim E-K, Kim H-E, Han K, Kang BJ, Sohn Y-M, Woo OH, et al. Applying Data-Driven Imaging Biomarker in Mammography for Breast Cancer Screening: Preliminary Study. Sci Rep, 2018, 8:2762., DOI 10.1038/s41598- 018-21215-1 Chougrad H, Zouaki H, Alheyane O. Deep Convolutional Neural Networks for Breast Cancer Screening. Comp Methods Programs Biomed, 2018, 157: 19–30., DOI 10.1016/j.cmpb.2018.01.011 Dhungel N, Carneiro G, Bradley AP. A Deep Learning Approach for the Analysis of Masses in Mammograms with Minimal User Intervention. Med Image Anal, 2017, 37:114–28., DOI 10.1016/j.media.2017.01.009 Lee HN, Sohn Y-M, Han KH. Comparison of Mammographic Density Estimation by Volpara Software with Radiologists’ Visual Assessment: Analysis of Clinical-Radiologic Factors Affecting Discrepancy between Them. Acta Radiol, 2015, 56:1061–8., DOI 10.1177/0284185114554674 U.S. Food & Drug Administration. Available at: https://www.accessdata.fda. gov/cdrh_docs/pdf16/K163623.pdf. U.S. Food & Drug Administration. Available at: https://www.accessdata.fda. gov/cdrh_docs/pdf16/K170540.pdf. Teesside University. Artificial Intelligence to Improve Breast Cancer Screening, 2018). Available at: https://www.tees.ac.uk/sections/news/pressreleases_ story.cfm?story_id=6784. Mendelson EB. Artificial Intelligence in Breast Imaging: Potentials and Limitations. AJR Am J Roentgenol, 2018, 212:293–9., DOI 10.2214/AJR.18. 20532 Le EPV,Wang Y, Huang Y,Hickman S, Gilbert FJ. Artificial Intelligence in Breast Imaging. Clin Radiol, 2019, 74:357–66., DOI 10.1016/j.crad.2019. 02.006 Bahl M, Barzilay R, Yedidia AB, Locascio NJ, Yu L, Lehman CD. High-risk Breast Lesions: aMachine LearningModel to Predict Pathologic Upgrade and Reduce Unnecessary Surgical Excision. Radiology, 2018, 286:810–8., DOI 10. 1148/radiol.2017170549 Sanders LM, Sharma P, El Madany M, King AB, Goodman KS, Sanders AE. Clinical Breast Concerns in Low-Risk Pediatric Patients: Practice Review with Proposed Recommendations. Pediatr Radiol, 2018, 48:186–95., DOI 10.1007/ s00247-017-4007-6 National Institute for Health and Clinical Excellence. Image-guided Vacuum- Assisted Excision Biopsy of Benign Breast Lesions, 2006). Available at: http:// www.nice.org.uk/nicemedia/pdf/ip/IPG156guidance.pdf. Smith GEC, Burrows P. Ultrasound Diagnosis of Fibroadenoma - Is Biopsy Always Necessary? Clin Radiol, 2008, 63:511–5., DOI 10.1016/j.crad.2007. 10.015 Taylor K, Lowes S, Stanley E, Hamilton P, Redman A, Leaver A, et al. Evidence for Avoiding the Biopsy of Typical Fibroadenomas in Women Aged 25-29 Years. Clin Radiol, 2019, 74:676–81., DOI 10.1016/j.crad.2019. 02.019 NICE, 2017). Available at: https://www.nice.org.uk/guidance/ipg592. Zhang BN, Cao XC, Chen JY, Chen J, Fu L, Hu XC, et al. Guidelines on the Diagnosis and Treatment of Breast Cancer, 2011 Edition). Gland Surg, 2012, 1:39–61., DOI 10.3978/j.issn.2227-684X.2012.04.07 British Columbia. BCGuidelines.ca, 2013). Available at: https://www2.gov.bc. ca/assets/gov/health/practitioner-pro/bc-guidelines/brdisease.pdf. University Hospitals Birmingham NHS. Guideline for the Imaging of Patients Presenting with Breast Symptoms Incorporating the Guideline for the Use of MRI in Breast Cancer Online, 2013). Available at: https://www.uhb.nhs.uk/ Downloads/pdf/CancerPbImagingBreastCancer.pdf. Evans A, Pinder S, Wilson R. Breast Calcification-A Diagnostic Manual. London: Greenwich Medical Media, 2002). Forrai G, Toth ZsSebo E, et al. Emlodiagnosztikai Asszisztensek Elmeleti Es Gyakorlati Kezikonyve. [Theoretical and Practical Handbook on Breast Diagnostics for Medical Technicians]. Budapest: Saxum, 2019). OKI 2017. Lehman CD. Magnetic Resonance Imaging in the Evaluation of Ductal Carcinoma In Situ. JNCI Monogr, 2010, 2010:150–1., DOI 10.1093/ jncimonographs/lgq030 Lim HS, Jeong SJ, Lee JS, Park MH, Kim JW, Shin SS, et al. Paget Disease of the Breast: Mammographic, US, and MR Imaging Findings with Pathologic Correlation. RadioGraphics, 2011, 31:1973–87., DOI 10.1148/rg.317115070 Van der Ploeg IMC, Hobbelink M, van den Bosch MAAJ, Mali WPTM, Rinkes IHMB, van Hillegersberg R. ’Radioguided Occult Lesion Localisation’, ROLL, for Non-palpable Breast Lesions: A Review of the Relevant Literature. Eur J Surg Oncol, EJSO,, 2008, 34:1–5., DOI 10.1016/j.ejso.2007.03.002 Borbely K, Sinkovics I, Madaras B, et al. Az Emlorak Korszeru Kepalkoto Diagnosztikaja: Nuklearis Medicina Technikak. [Advanced Imaging Diagnostics of Breast Cancer: Nuclear Medicine Techniques]. Orv Hetil, 2012, 153:15–22. McCormick JT, Tikhomirov VB, Budway RJ, Caushaj PF. Analysis of the Use of Specimen Mammography in Breast Conservation Therapy. Am J Surg, 2004, 188:433–6., DOI 10.1016/j.amjsurg.2004.06.030 Kim T, Giuliano AE, Lyman GH. Lymphatic Mapping and sentinel Lymph Node Biopsy in Early-Stage Breast Carcinoma. Cancer, 2006, 106:4–16., DOI 10.1002/cncr.21568 Postma EL, Verkooijen HM, Verkooijen HM, van Esser S, Hobbelink MG, van der Schelling GP, et al. Efficacy of ’radioguided Occult Lesion Localisation’, ROLL, versus ’wire-Guided Localisation’, WGL, in Breast Conserving Surgery for Non-palpable Breast Cancer: A Randomised Controlled Multicentre Trial. Breast Cancer Res Treat, 2012, 136:469–78., DOI 10.1007/s10549-012-2225-z Takacs T, Paszt A, Simonka Z, Abraham S, Borda B, Ottlakan A, et al. Radioguided Occult Lesion Localisation versus Wire-Guided Lumpectomy in the Treatment of Non-palpable Breast Lesions. Pathol Oncol Res, 2013, 19: 267–73., DOI 10.1007/s12253-012-9578-9 Harvey JR, Lim Y, Murphy J, Howe M, Morris J, Goyal A, et al. Safety and Feasibility of Breast Lesion Localization Using Magnetic Seeds, Magseed): A multi-centre, Open-Label Cohort Study. Breast Cancer Res Treat, 2018, 169: 531–6., DOI 10.1007/s10549-018-4709-y Kapoor MM, Patel MM, Scoggins ME. The Wire and beyond: Recent Advances in Breast Imaging Preoperative Needle Localization. RadioGraphics, 2019, 39:1886–906., DOI 10.1148/rg.2019190041 McGuire KP, Toro-Burguete J, Dang H, Young J, Soran A, Zuley M, et al. MRI Staging after Neoadjuvant Chemotherapy for Breast Cancer: Does Tumor Biology Affect Accuracy? Ann Surg Oncol, 2011, 18:3149–54., DOI 10.1245/ s10434-011-1912-z Schmitz AC, Gianfelice D, Daniel BL, Mali WPTM, van den Bosch MAAJ. Image-guided Focused Ultrasound Ablation of Breast Cancer: Current Status, Challenges, and Future Directions. Eur Radiol, 2008, 18:1431–41., DOI 10. 1007/s00330-008-0906-0 Littrup PJ, Freeman-Gibb L, Andea A, White M, Amerikia KC, Bouwman D, et al. Cryotherapy for Breast Fibroadenomas. Radiology, 2005, 234:63–72., DOI 10.1148/radiol.2341030931 Edwards MJ, Broadwater R, Tafra L, Jarowenki D, Mabry C, Beitsch P, et al. Progressive Adoption of Cryoablative Therapy for Breast Fibroadenoma in Community Practice. Am J Surg, 2004, 188:221–4., DOI 10.1016/j.amjsurg. 2004.07.002 Kaufman CS, Littrup PJ, Freeman-Gibb LA, Smith JS, Francescatti D, Simmons R, et al. Office-based Cryoablation of Breast Fibroadenomas with Long-Term Follow-Up. Breast J, 2005, 11:344–50., DOI 10.1111/j. 1075-122x.2005.21700.x Lakoma A, Kim ES. Minimally Invasive Surgical Management of Benign Breast Lesions. Gland Surg, 2014, 3:142–8., DOI 10.3978/j.issn.2227-684X. 2014.04.01 ClinicalTrials.gov. Cryoablation Therapy in Treating Patients with Invasive Ductal Breast Cancer, 2017). Available at: https://clinicaltrials.gov/ct2/show/ NCT00723294?term=z1072&rank=1. Littrup PJ, Jallad B, Chandiwala-Mody P, D’Agostini M, Adam BA, Bouwman D. Cryotherapy for Breast Cancer: a Feasibility Study without Excision. J Vasc Interv Radiol, 2009, 20:1329–41., DOI 10.1016/j.jvir.2009. 06.029 Manenti G, Perretta T, Gaspari E, Pistolese CA, Scarano L, Cossu E, et al. Percutaneous Local Ablation of Unifocal Subclinical Breast Cancer: Clinical Experience and Preliminary Results of Cryotherapy. Eur Radiol, 2011, 21:2344–53., DOI 10.1007/s00330- 011-2179-2 Manenti G, Bolacchi F, Perretta T, Cossu E, Pistolese CA, Buonomo OC, et al. Small Breast Cancers: In Vivo Percutaneous US-Guided Radiofrequency Ablation with Dedicated Cool-Tip Radiofrequency System. Radiology, 2009, 251:339–46., DOI 10.1148/radiol.2512080905 Palussiere J, Henriques C, Mauriac L, Asad-Syed M, Valentin F, Brouste V, et al. Radiofrequency Ablation as a Substitute for Surgery in Elderly Patients with Nonresected Breast Cancer: Pilot Study with Long-Term Outcomes. Radiology, 2012, 264:597–605., DOI 10.1148/ radiol.12111303 Nguyen T, Hattery E, Khatri VP. Radiofrequency Ablation and Breast Cancer: A Review. Gland Surg, 2014, 3:128–35., DOI 10.3978/j.issn.2227-684X.2014. 03.05 Linda A, Zuiani C, Furlan A, Lorenzon M, Londero V, Girometti R, et al. Nonsurgical Management of High-Risk Lesions Diagnosed at Core Needle Biopsy: Can Malignancy Be Ruled Out Safely with Breast MRI? AJR Am J Roentgenol, 2012, 198:272–80., DOI 10.2214/ AJR.11.7040 Pediconi F, Padula S, Dominelli V, Luciani M, Telesca M, Casali V, et al. Role of Breast MR Imaging for Predicting Malignancy of Histologically Borderline Lesions Diagnosed at Core Needle Biopsy: Prospective Evaluation. Radiology, 2010, 257:653–61., DOI 10.1148/radiol.10100732 Rageth CJ, O’Flynn EA, Comstock C, Kurtz C, Kubik R, Madjar H, et al. First International Consensus Conference on Lesions of Uncertain Malignant Potential in the Breast, B3 Lesions). Breast Cancer Res Treat, 2016, 159: 203–13., DOI 10.1007/s10549-016-3935-4 Rageth CJ, O’Flynn EAM, Pinker K, Kubik-Huch RA, Mundinger A, Decker T, et al. Second International Consensus Conference on Lesions of Uncertain Malignant Potential in the Breast, B3 Lesions). Breast Cancer Res Treat, 2019, 174:279–96., DOI 10.1007/s10549-018-05071-1 Association of Breast Surgery. NHS Breast Screening Programme Clinical Guidance for Breast Cancer Screening Assessment, 2016). Available at: https:// associationofbreastsurgery.org.uk/media/1414/nhs-bsp-clinical-guidancefor- breast-cancer-screening-assessment.pdf. Clemens MW, Jacobsen ED, Horwitz SM. 2019 NCCN Consensus Guidelines on the Diagnosis and Treatment of Breast Implant-Associated Anaplastic Large Cell Lymphoma, BIA-ALCL). Aesthet Surg J, 2019, 39(S1):S3–S13., DOI 10.1093/asj/sjy331 Johansen Taber KA, Morisy LR, Osbahr AJ, Dickinson BD. Male Breast Cancer: Risk Factors, Diagnosis, and Management, Review). Oncol Rep, 2010, 24:1115–20., DOI 10.3892/or_00000962 Gao Y, Goldberg JE, Young TK, Babb JS, Moy L, Heller SL. Breast Cancer Screening in High-Risk Men: A 12-year Longitudinal Observational Study of Male Breast Imaging Utilization and Outcomes. Radiology, 2019, 293: 282–91., DOI 10.1148/radiol.2019190971 Marino MA, Gucalp A, Leithner D, Keating D, Avendano D, Bernard-Davila B, et al. Mammographic Screening in Male Patients at High Risk for Breast Cancer: Is it worth it? Breast Cancer Res Treat, 2019, 177:705–11., DOI 10. 1007/s10549-019-05338-1 Mitchell KB, Johnson HM, Eglash A, Young M, Noble L, Reece-Stremtan S, et al. ABM Clinical Protocol #30: Breast Masses, Breast Complaints, and Diagnostic Breast Imaging in the Lactating Woman. Breastfeed Med, 2019, 14:208–14., DOI 10.1089/bfm.2019.29124.kjm National Pathology Advisory Board. Az Egeszsegugyi Miniszterium Szakmai Protokollja a Mammografias Emloszuresrol Es a Korai Emlorak Diagnosztikajarol. [Professional Protocol of the Ministry of Health on Mammography Breast Screening and Early Breast Cancer Diagnostics. Hungarian Health Bulletin]. Egeszsegugyi Kozlony, 2008, 10:2990–3012. Brierley JD, Gospodarowicz MK, Wittekind C, et al. A Rosszindulatu Daganatok TNM-Klasszifikacioja Es Stadiumbesorolasa. [TNM Classification and Staging of Malignancies]. Oriold es Tarsai Kiado Kft., 2017). Kasler M. Ajanlas Az Emlorak Korszeru Diagnosztikajara, Kezelesere Es Gondozasara. Elso Magyar Nemzeti Emlorak Konszenzus Konferencia Iranyelvei. [Recommendation for State-Of-The-Art Diagnostics, Treatment and Care of Breast Cancer. Guidelines of the First Hungarian National Breast Cancer Consensus Conference]. Magy Onkol, 2000, 44:11–38. Coates AS, Winer EP, Goldhirsch A, Gelber RD, Gnant M, Piccart-Gebhart M, et al. Tailoring Therapies-Iimproving the Management of Early Breast Cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015. Ann Oncol, 2015, 26:1533–46., DOI 10.1093/ annonc/mdv221 Cardoso F, Costa A, Norton L, Senkus E, Aapro M, Andre F, et al. ESO-ESMO 2nd International Consensus Guidelines for Advanced Breast Cancer, ABC2). Ann Oncol, 2014, 25:1871–88., DOI 10.1093/annonc/mdu385 Tasnadi T, Forrai G. Emlo-MR-vizsgalatok Leletezesi Protokollja a BIRADS Lexikon Alapjan I. A Leletezes Elvei Es Alapfogalmai. Magy Radiol, 2017, 8(2):1–16. [Tasnadi T, Forrai G. Reporting protocol for breast MRI scans based on BIRADS lexicon I. Principles and basic concepts of reporting. Hungarian Radiology journal, 2017, 8(2):1–16]. Tasnadi T, Forrai G. Emlo-MR-vizsgalatok Leletezesi Protokollja a BIRADS Lexikon Alapjan II. Leletsablonok. Magy Radiol, 2017, 8(3):1–6. [Tasnadi T, Forrai G. Reporting Protocol for Breast MRI Scans based on BIRADS Lexicon II. Reporting templates. Hungarian Radiology Journal, 2017, 8(3):1–6]. EMMI. Decree 23/2012, IX. 14., EMMI of the Minister of Human Capacities. ESzCsM. Decree 60/2003, X. 20., ESzCsM of the Minister of Health, Social and Family Affairs. Bordas P, Jonsson H, Nystrom L, Lenner P. Interval Cancer Incidence and Episode Sensitivity in the Norrbotten Mammography Screening Programme, Sweden. J Med Screen, 2009, 16:39–45., DOI 10.1258/jms.2009.008098 Oeffinger KC, Fontham ETH, Etzioni R, Herzig A, Michaelson JS, Shih Y-CT, et al. Breast Cancer Screening for Women at Average Risk. JAMA, 2015, 314: 1599–614., DOI 10.1001/jama.2015.12783 Bluekens AMJ, Holland R, Karssemeijer N, Broeders MJM, den Heeten GJ. Comparison of Digital Screening Mammography and Screen-Film Mammography in the Early Detection of Clinically Relevant Cancers: a Multicenter Study. Radiology, 2012, 265:707–14., DOI 10.1148/radiol. 12111461 Seradour B, Heid P, Esteve J. Comparison of Direct Digital Mammography, Computed Radiography, and Film-Screen in the French National Breast Cancer Screening Program. Am J Roentgenol, 2014, 202:229–36., DOI 10.2214/ AJR.12.10419 Pinker K, Perry N, Vinnicombe S, Shiel S, Weber M. Conspicuity of Breast Cancer According to Histopathological Type and Breast Density when Imaged by Full-Field Digital Mammography Compared with Screen-Film Mammography. Eur Radiol, 2011, 21:18–25., DOI 10. 1007/s00330-010-1906-4 Yamada T, Ishibashi T, Sato A, Saito M, Saito H, Matsuhashi T, et al. Comparison of Screen-Film and Full-Field Digital Mammography: Image Contrast and Lesion Characterization. Radiat Med, 2003, 21:166–71. Fischer U, Baum F, Obenauer S, Luftner-Nagel S, von Heyden D, Vosshenrich R, et al. Comparative Study in Patients with Microcalcifications: Full-Field Digital Mammography vs Screen-Film Mammography. Eur Radiol, 2002, 12: 2679–83., DOI 10.1007/s00330-002-1354-x Pisano ED, Hendrick RE, Yaffe MJ, Baum JK, Acharyya S, Cormack JB, et al. Diagnostic Accuracy of Digital versus Film Mammography: Exploratory Analysis of Selected Population Subgroups in DMIST. Radiology, 2008, 246:376–83., DOI 10.1148/radiol.2461070200 Juel I-M, Skaane P, Hoff SR, Johannessen G, Hofvind S. Screen-film Mammography versus Full-Field Digital Mammography in a Population- Based Screening Program: The Sogn and Fjordane Study. Acta Radiol, 2010, 51:962–8., DOI 10.3109/02841851.2010.504969 Ranger NT, Lo JY, Samei E. A Technique Optimization Protocol and the Potential for Dose Reduction in Digital Mammography. Med Phys, 2010, 37: 962–9., DOI 10.1118/1.3276732 Schueller G, Riedl CC, Mallek R, Eibenberger K, Langenberger H, Kaindl E, et al. Image Quality, Lesion Detection, and Diagnostic Efficacy in Digital Mammography: Full-Field Digital Mammography versus Computed Radiography-Based Mammography Using Digital Storage Phosphor Plates. Eur J Radiol, 2008, 67:487–96., DOI 10.1016/j.ejrad.2007.08.016 Kasler M. A Komplex Onkodiagnosztika Es Onkoterapia Iranyelvei. Semmelweis Kiado. [Principles of Complex Oncological Diagnostics and Oncotherapy]. Budapest: Semmelweis, 2008). p. 329–68. Tabar L, Dean PB, Chen TH, Yen AM, Chen SL, Fann JC, et al. The Incidence of Fatal Breast Cancer Measures the Increased Effectiveness of Therapy in Women Participating in Mammography Screening. Cancer, 2019, 125: 515–23., DOI 10.1002/cncr.31840 ANTSZ. Lakossagi Szűrővizsgalatok, Populations Screening Investigations). Available at: https://www.antsz.hu/data/cms41690/lakossagi_ szurovizsgalatok.pdf. National Radiology Advisory Board. A Radiologiai Szakmai Kollegium Allasfoglalasa a Radiologia Digitalizalasaval Kapcsolatos Kerdesekrol, A Digitalis Radiologia, a PACS Es a Teleradiologia Fejlodesi Iranyai Szakmai Technikai Jogi Feltetelrendszere). Radiologiai Szakmai Kollegium, 2007). Available at: www.radiologia.hu. [Resolution of the National Radiology Advisory Board on Issues Related to the Digitization of Radiology, Professional Technical Legal Conditions of Development Directions for Digital Radiology, PACS and Teleradiology). National Radiology Advisory Board, 2007). Available at: www.radiologia.hu]. Gennaro G, Avramova-Cholakova S, Azzalini A, Luisa Chapel M, Chevalier M, Ciraj O, et al. Quality Controls in Digital Mammography Protocol of the EFOMP Mammo Working Group. Phys Med, 2018, 48:55–64., DOI 10.1016/j. ejmp.2018.03.016 Gresz M. Relationship between Breast Cancer and Breast Screening in Hungary Analysed on the Basis of the National Health Insurance Fund Database. Orvosi Hetilap, 2012, 153:1745–51., DOI 10.1556/oh.2012. 29427 Laubs-Secretan B, Scoccianti C, Loomis D, Benbrahim-Tallaa L, Bouvard V, Bianchini F, et al. Breast-cancer Screening-Vviewpoint of the IARC Working Group. N Engl J Med, 2015, 372:2353–8., DOI 10.1056/NEJMsr1504363 Cserni G, Francz M, Jaray B, Kalman E, Kovacs I, Krenacs T, et al. Pathological Diagnosis, Work-Up and Reporting of Breast Cancer 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022). in press. Matrai Z, Kelemen P, Kosa C, Maraz R, Paszt A, Pavlovics G, et al. Modern Breast Cancer Surgery. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022). in press. Rubovszky G, Kocsis J, Boer K, Chilingirova N, Dank M, Kahan Z, et al. Systemic Treatment of Breast Cancer. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022). in press. Polgar C, Kahan Z, Ivanov O, Chorvath M, Ligacova A, Csejtei A, et al. Radiotherapy of Breast Cancer – Professional Guideline. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022). in press. Kahan Z, Szanto I, Dudas R, Kapitany Z, Molnar M, Koncz Z, et al. Breast Cancer: Follow-Up, Rehabilitation, Psycho-Oncology. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022). in press. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610382 EP 1610405 DI 10.3389/pore.2022.1610382 PG 24 ER PT J AU Xu, R Ke, X Shang, W Liu, Sh Fu, X Wang, T Jin, Sh AF Xu, Rui Ke, Xing Shang, Wenwen Liu, Shuna Fu, Xin Wang, Ting Jin, Shuxian TI Distribution and Clinical Significance of IL-17A in Tumor-Infiltrating Lymphocytes of Non-Small Cell Lung Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NSCLC; TILs; Th17 cells; Tc17 cells; γδT cells; IL-17 ID NSCLC; TILs; Th17 cells; Tc17 cells; γδT cells; IL-17 AB Objective: To investigate the distribution of IL-17A and its clinical significance in tumor infiltrating lymphocytes (TILs) of patients with non-small cell lung cancer (NSCLC). Methods: Expression level of IL-17A in TILs of 3 paired NSCLC and paracancerous specimens was measured by qRT-PCR. The distribution of IL-17A in immune cell subsets of 15 paired NSCLC and paracancerous specimens was examined by flow cytometry. The correlation between IL-17A and clinical features of NSCLC was identified. Results: IL-17A was significantly upregulated in TILs of NSCLC specimens than those of paracancerous ones (p < 0.0001). Meanwhile, T helper 17 cells (Th17 cells, p < 0.001), IL- 17-secreting CD8+ T cells (Tc17 cells, p < 0.001) and IL-17-producing cells (γδT17 cells, p < 0.0001) were significantly abundant in TILs of NSCLC specimens than those of controls, and the higher abundance of the latter was much pronounced than that of the former two. Moreover, γδT17 cells in TILs were significantly correlated with lymphatic metastasis and CYFRA 21-1 level of NSCLC patients (p < 0.05). Conclusion: Tumor infiltrated γδT cells are the main source of IL-17 in early-stage NSCLC, and IL-17 may be a vital regulator involved in the development of NSCLC. C1 [Xu, Rui] The First Affiliated Hospital of Nanjing Medical University, Department of Laboratory MedicineNanjing, China. [Ke, Xing] Shanghai Jiao Tong University School of Medicine, Xinhua Hospital, Department of Clinical LaboratoryShanghai, China. [Shang, Wenwen] The First Affiliated Hospital of Nanjing Medical University, Department of Laboratory MedicineNanjing, China. [Liu, Shuna] The First Affiliated Hospital of Nanjing Medical University, Department of Laboratory MedicineNanjing, China. [Fu, Xin] The First Affiliated Hospital of Nanjing Medical University, Department of Laboratory MedicineNanjing, China. [Wang, Ting] The First Affiliated Hospital of Nanjing Medical University, Department of Laboratory MedicineNanjing, China. [Jin, Shuxian] The First Affiliated Hospital of Nanjing Medical University, Department of Respiratory and Critical Care MedicineNanjing, China. RP Jin, Sh (reprint author), The First Affiliated Hospital of Nanjing Medical University, Department of Respiratory and Critical Care Medicine, Nanjing, China. EM jin_sx@aliyun.com CR Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin, 2021, 71(1):7–33., DOI 10.3322/caac.21654 Bouras E, Karhunen V, Gill D, Huang J, Haycock PC, Gunter MJ, et al. Circulating Inflammatory Cytokines and Risk of Five Cancers : A Mendelian Randomization Analysis. BMC Med, 2022, 20:1–15., DOI 10.1186/s12916-021- 02193-0 Corsale AM, Di Simone M, Lo Presti E, Picone C, Dieli F, Meraviglia S. Metabolic Changes in Tumor Microenvironment: How Could They Affect γδ T Cells Functions? Cells, 2021, 10(11):2896., DOI 10.3390/ cells10112896 Wu X, Yang T, Liu X, Guo Jn., Xie T, Ding Y, et al. IL-17 Promotes Tumor Angiogenesis through Stat3 Pathway Mediated Upregulation of VEGF in Gastric Cancer. Tumor Biol, 2016, 37(4):5493–501., DOI 10.1007/s13277-015-4372-4 Hurtado CG, Wan F, Housseau F, Sears CL. Roles for Interleukin 17 and Adaptive Immunity in Pathogenesis of Colorectal Cancer. Gastroenterology, 2018, 155(6):1706–15., DOI 10.1053/j.gastro.2018.08.056 Bugaut H, Aractingi S. Major Role of the IL17/23 Axis in Psoriasis Supports the Development of New Targeted Therapies. Front Immunol, 2021, 12:621956., DOI 10.3389/fimmu.2021.621956 Cheng M, Qian L, Shen G, Bian G, Xu T, Xu W, et al. Microbiota Modulate Tumoral Immune Surveillance in Lung through a γδT17 Immune Celldependent Mechanism. Cancer Res, 2014, 74(15):4030–41., DOI 10.1158/ 0008-5472.can-13-2462 Lockhart E, Green AM, Flynn JL. IL-17 Production Is Dominated by γδ T Cells rather Than CD4 T Cells during Mycobacterium tuberculosis Infection. J Immunol, 2006, 177(7):4662–9., DOI 10.4049/jimmunol.177. 7.4662 Wilharm A, Brigas HC, Sandrock I, Ribeiro M, Amado T, Reinhardt A, et al. Microbiota-dependent Expansion of Testicular IL-17-producing Vγ6+ γδ T Cells upon Puberty Promotes Local Tissue Immune Surveillance. Mucosal Immunol, 2021, 14(1):242–52., DOI 10.1038/s41385-020-0330-6 Marie IJ, Brambilla L, Azzouz D, Chen Z, Baracho GV, Arnett A, et al. Tonic Interferon Restricts Pathogenic IL-17-driven Inflammatory Disease via Balancing the Microbiome. Elife, 2021, 10:e68371., DOI 10.7554/eLife. 68371 Patil RS, Shah SU, Shrikhande SV, Goel M, Dikshit RP, Chiplunkar SV. IL17 Producing γδT Cells Induce Angiogenesis and Are Associated with Poor Survival in Gallbladder Cancer Patients. Int J Cancer, 2016, 139(4):869–81., DOI 10.1002/ijc.30134 Song MH, Gupta A, Kim HO, Oh K. Lysophosphatidylcholine Aggravates Contact Hypersensitivity by Promoting Neutrophil Infiltration and IL17 Expression. BMB Rep, 2021, 54(4):203–8., DOI 10.5483/bmbrep.2021.54. 4.193 Zhang Y, Chandra V, Riquelme Sanchez E, Dutta P, Quesada PR, Rakoski A, et al. Interleukin-17-induced Neutrophil Extracellular Traps Mediate Resistance to Checkpoint Blockade in Pancreatic Cancer. J Exp Med, 2020, 217(12):e20190354., DOI 10.1084/jem.20190354 Zhao C, Li Y, ZhangW, Zhao D, Ma L, Ma P, et al. IL-17 Induces NSCLC A549 Cell Proliferation via the Upregulation of HMGA1, Resulting in an Increased Cyclin D1 Expression. Int J Oncol, 2018, 52(5):1579–92., DOI 10.3892/ijo.2018. 4307 Jiang Y-X, Yang S-W, Li P-A, Luo X, Li Z-Y, Hao Y-X, et al. The Promotion of the Transformation of Quiescent Gastric Cancer Stem Cells by IL-17 and the Underlying Mechanisms. Oncogene, 2017, 36(9):1256–64., DOI 10.1038/onc. 2016.291 Chen G, Zhang PG, Li JS, Duan JJ, Su W, Guo SP, et al. Th17 Cell Frequency and IL-17A Production in Peripheral Blood of Patients with Non-small-cell Lung Cancer. J Int Med Res, 2020, 48(6):300060520925948., DOI 10.1177/ 0300060520925948 Song L, Ma S, Chen L, Miao L, Tao M, Liu H. Long-term Prognostic Significance of Interleukin-17-producing T Cells in Patients with Nonsmall Cell Lung Cancer. Cancer Sci, 2019, 110(7):2100–9., DOI 10.1111/cas. 14068 Cui K, Mei X, Cheng M. Increased Interleukin-17a-Producing γδT Cells Predict Favorable Survival in Elderly Patients with LUAD and LUSC. J Cancer Res Clin Oncol, 2021, 147(11):3289–98., DOI 10.1007/s00432-021- 03742-z Zhang F, Wu X, Zhu J, Huang Y, Song X, Jiang L. 18F-FDG PET/CT and Circulating Tumor Cells in Treatment-Naive Patients with Non-small-cell Lung Cancer. Eur J Nucl Med Mol Imaging, 2021, 48(10):3250–9., DOI 10.1007/ s00259-021-05260-z NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610384 EP 1610390 DI 10.3389/pore.2022.1610384 PG 7 ER PT J AU Wang, Sh Yu, A Han, M Chen, X Li, Z Ke, M Cai, X Ai, M Xing, Y AF Wang, Shun Yu, Aihua Han, Mengyao Chen, Xiaomin Li, Zhi Ke, Min Cai, Xiaojun Ai, Ming Xing, Yiqiao TI Pathological Changes and Expression of JAK-STAT Signaling Pathway Hallmark Proteins in Rat Retinas at Different Time Points After Retinal Ischemia Reperfusion Injury SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apoptosis; pathology; retina ischemia reperfusion injury; retinal ganglion cells; Janus kinase signal transducer and activator of transcription ID apoptosis; pathology; retina ischemia reperfusion injury; retinal ganglion cells; Janus kinase signal transducer and activator of transcription AB Retinal ischemia reperfusion injury (RIRI) is a conventional pathological process in various retinal vascular diseases. Many studies select only one specific time point to apply drugs and then assess the therapeutic effect of drugs; however, the baselines are not the same at different time points, which may cause variation in the judgement. Therefore, further investigation is needed. Accordingly, this study aimed to investigate the pathological changes of retinal structure, expression of JAK-STAT signaling pathway hallmark proteins, and apoptosis at different time points after retinal ischemia reperfusion injury in rats. Sixtysix male SPF Sprague-Dawley rats were randomly divided into six groups: control group, RIRI 0, 6-, 24-, 72-, and 144-h groups. RIRI models were induced by perfusing equilibrium solution into the right eye anterior chamber to increase intraocular pressure to 110 mmHg for 60 min. Rats were sacrificed at different time points after reperfusion. Then hematoxylin-eosin staining, transmission electron microscope, immunohistochemistry, western blot, and TUNEL were used. Hematoxylin-eosin showed the pathological changes while transmission electron microscope revealed the ultra-structure changes of retina after RIRI. Immunohistochemistry showed that JAK2, STAT3, p-JAK2, p-STAT3, Bax, and Bcl-2 proteins mainly located in ganglion cell layer and inner nuclear layer, the relative expression of former five proteins had significant differences vs. control group (p < 0.05), while Bcl-2 had no significant difference. In western blot, the protein expressing of JAK2, STAT3, p-JAK2, p-STAT3, p-Akt, and Bax had significant differences vs. control group (p < 0.05), while Akt and Bcl-2 had no significant differences. TUNEL staining showed the number of apoptosis positive cells rose initially but declined later, with a peak value at RIRI 24 h group. The dynamic changes of hallmark proteins at different time points after RIRI indicate that JAK-STAT signaling pathway activates rapidly but weakens later and plays a vital role in RIRI, and apoptosis is involved in RIRI with a peak value at 24 h in the process, suggesting a potential therapeutic direction and time window for treating RIRI. C1 [Wang, Shun] Renmin Hospital of Wuhan University, Eye CenterWuhan, China. [Yu, Aihua] Renmin Hospital of Wuhan University, Eye CenterWuhan, China. [Han, Mengyao] Wuhan Aier Eye Hospital of Wuhan University, Retinal and Vitreous Diseases DepartmentWuhan, China. [Chen, Xiaomin] Zhongnan Hospital of Wuhan University, Department of OphthalmologyWuhan, China. [Li, Zhi] Zhongnan Hospital of Wuhan University, Department of OphthalmologyWuhan, China. [Ke, Min] Zhongnan Hospital of Wuhan University, Department of OphthalmologyWuhan, China. [Cai, Xiaojun] Zhongnan Hospital of Wuhan University, Department of OphthalmologyWuhan, China. [Ai, Ming] Renmin Hospital of Wuhan University, Eye CenterWuhan, China. [Xing, Yiqiao] Renmin Hospital of Wuhan University, Eye CenterWuhan, China. RP Ai, M (reprint author), Renmin Hospital of Wuhan University, Eye Center, Wuhan, China. EM wan@whu.edu.cn CR Minhas G, Sharma J, Khan N. Cellular Stress Response and Immune Signaling in Retinal Ischemia-Reperfusion Injury. Front Immunol, 2016, 7:444., DOI 10. 3389/fimmu.2016.00444 Osborne NN, Casson RJ,Wood JPM, Chidlow G, Graham M, Melena J. Retinal Ischemia: Mechanisms of Damage and Potential Therapeutic Strategies. Prog Retin Eye Res, 2004, 23(1):91–147., DOI 10.1016/j.preteyeres.2003.12.001 Villarino AV, Gadina M, O’Shea JJ, Kanno Y. SnapShot: Jak-STAT Signaling II. Cell, 2020, 181(7):1696., DOI 10.1016/j.cell.2020.04.052 Goodman MD, Koch SE, Fuller-Bicer GA, Butler KL. Regulating RISK: a Role for JAK-STAT Signaling in Postconditioning? Am J Physiology-Heart Circulatory Physiol, 2008, 295(4):H1649–H1656., DOI 10.1152/ajpheart.00692.2008 Li B, Yu J, Liu P, Zeng T, Zeng X. Astragaloside IV Protects Cardiomyocytes against Hypoxia Injury via HIF-1α and the JAK2/STAT3 Pathway. Ann Transl Med, 2021, 9(18):1435., DOI 10.21037/atm-21-4080 Zhao X, Zhao B, Zhao Y, Zhang Y, Qian M. Protective Effect of Anisodamine on Bleomycin-Induced Acute Lung Injury in Immature Rats via Modulating Oxidative Stress, Inflammation, and Cell Apoptosis by Inhibiting the JAK2/ STAT3 Pathway. Ann Transl Med, 2021, 9(10):859., DOI 10.21037/atm-21- 1750 Shao F, Pang X, Baeg GH. Targeting the JAK/STAT Signaling Pathway for Breast Cancer. Cmc, 2021, 28(25):5137–51., DOI 10.2174/ 0929867328666201207202012 Hin Tang JJJJ, Hao Thng DK, Lim JJ, Toh TB. JAK/STAT Signaling in Hepatocellular Carcinoma. Hepatic Oncol, 2020, 7(1):HEP18., DOI 10.2217/ hep-2020-0001 Owen KL, Brockwell NK, Parker BS. JAK-STAT Signaling: A Double-Edged Sword of Immune Regulation and Cancer Progression. Cancers, 2019, 11(12): 2002., DOI 10.3390/cancers11122002 Malemud CJ. The Role of the JAK/STAT Signal Pathway in Rheumatoid Arthritis. Ther Adv Musculoskelet, 2018, 10(5-6):117–27., DOI 10.1177/ 1759720X18776224 Fasouli ES, Katsantoni E. JAK-STAT in Early Hematopoiesis and Leukemia. Front Cell Dev. Biol., 2021, 9:669363., DOI 10.3389/fcell.2021.669363 Gurzov EN, Stanley WJ, Pappas EG, Thomas HE, Gough DJ. The JAK/STAT Pathway in Obesity and Diabetes. FEBS J, 2016, 283(16):3002–15., DOI 10.1111/ febs.13709 Li C-D, Zhao J-Y, Chen J-L, Lu J-H, ZhangM-B, Huang Q, et al. Mechanismof the JAK2/STAT3-CAV-1-NR2B Signaling Pathway in Painful Diabetic Neuropathy. Endocrine, 2019, 64(1):55–66., DOI 10.1007/s12020-019-01880-6 Tang Y, Tong X, Li Y, Jiang G, Yu M, Chen Y, et al. JAK2/STAT3 Pathway Is Involved in the Protective Effects of Epidermal Growth Factor Receptor Activation against Cerebral Ischemia/reperfusion Injury in Rats. Neurosci Lett, 2018, 662:219–26., DOI 10.1016/j.neulet.2017.10.037 Kim H-C, Kim E, Bae JI, Lee KH, Jeon Y-T, Hwang J-W, et al. Sevoflurane Postconditioning Reduces Apoptosis by Activating the JAK-STAT Pathway after Transient Global Cerebral Ischemia in Rats. J Neurosurg Anesthesiol, 2017, 29(1):37–45., DOI 10.1097/ANA.0000000000000331 London A, Benhar I, Schwartz M. The Retina as aWindow to the Brain-From Eye Research to CNS Disorders. Nat Rev Neurol, 2013, 9(1):44–53., DOI 10. 1038/nrneurol.2012.227 Liu W, Xia F, Ha Y, Zhu S, Li Y, Folorunso O, et al. Neuroprotective Effects of HSF1 in Retinal Ischemia-Reperfusion Injury. Invest Ophthalmol Vis Sci, 2019, 60(4):965–77., DOI 10.1167/iovs.18-26216 Hartsock MJ, Cho H, Wu L, Chen W-J, Gong J, Duh EJ. A Mouse Model of Retinal Ischemia-Reperfusion Injury through Elevation of Intraocular Pressure. JoVE, 2016, 2016(113):54065., DOI 10.3791/54065 San Cristobal Epalza J, Palomares T, Garcia-Alonso I, Herrero de la Parte B. Histological Assessment of Rat Retinas with Ischemia-Reperfusion Injury. Eur Surg Res, 2021, 62(3):1–7., DOI 10.1159/000515832 Hayden CM, Meseck EK. Transmission Electron Microscopy of the Retina: A Method for Sample Preparation and Evaluation. Toxicol Pathol, 2021, 49(3): 521–7., DOI 10.1177/0192623320954124 Tham Y-C, Li X, Wong TY, Quigley HA, Aung T, Cheng C-Y. Global Prevalence of Glaucoma and Projections of Glaucoma Burden through 2040. Ophthalmology, 2014, 121(11):2081–90., DOI 10.1016/j.ophtha.2014. 05.013 Niwa M, Aoki H, Hirata A, Tomita H, Green P, Hara A. Retinal Cell Degeneration in Animal Models. Ijms, 2016, 17(1):110., DOI 10.3390/ ijms17010110 Zhong Y, Yin B, Ye Y, Dekhel OYAT, Xiong X, Jian Z, et al. The Bidirectional Role of the JAK2/STAT3 Signaling Pathway and Related Mechanisms in Cerebral Ischemia-Reperfusion Injury. Exp Neurol, 2021, 341:113690., DOI 10.1016/j.expneurol.2021.113690 Youle RJ, Strasser A. The BCL-2 Protein Family: Opposing Activities that Mediate Cell Death. Nat Rev Mol Cell Biol, 2008, 9(1):47–59., DOI 10.1038/nrm2308 Yang J, Yang N, Luo J, Cheng G, Zhang X, He T, et al. Overexpression of S100A4 Protects Retinal Ganglion Cells against Retinal Ischemia-Reperfusion Injury in Mice. Exp Eye Res, 2020, 201:108281., DOI 10.1016/j.exer.2020.108281 OlaMS, NawazM, Ahsan H. Role of Bcl-2 Family Proteins and Caspases in the Regulation of Apoptosis. Mol Cell Biochem, 2011, 351(1-2):41–58., DOI 10. 1007/s11010-010-0709-x D’Arcy MS. Cell Death: a Review of the Major Forms of Apoptosis, Necrosis and Autophagy. Cell Biol Int, 2019, 43(6):582–92., DOI 10.1002/cbin.11137 Ju W-K, Shim MS, Kim K-Y, Bu JH, Park TL, Ahn S, et al. Ubiquinol Promotes Retinal Ganglion Cell Survival and Blocks the Apoptotic Pathway in Ischemic Retinal Degeneration. Biochem Biophysical Res Commun, 2018, 503(4): 2639–45., DOI 10.1016/j.bbrc.2018.08.016 Wu J, Wang R, Yang D, Tang W, Chen Z, Sun Q, et al. Hydrogen Postconditioning Promotes Survival of Rat Retinal Ganglion Cells against Ischemia/reperfusion Injury through the PI3K/Akt Pathway. Biochem Biophysical Res Commun, 2018, 495(4):2462–8., DOI 10.1016/j.bbrc.2017. 12.146 Xin P, Xu X, Deng C, Liu S, Wang Y, Zhou X, et al. The Role of JAK/STAT Signaling Pathway and its Inhibitors in Diseases. Int Immunopharmacology, 2020, 80:106210., DOI 10.1016/j.intimp.2020.106210 Pan S, Chen Y, Zhang X, Xie Y. The JAK2/STAT3 Pathway Is Involved in Dexmedetomidine-Induced Myocardial protection in Rats Undergoing Cardiopulmonary Bypass. Ann Transl Med, 2020, 8(7):483., DOI 10.21037/ atm.2020.03.67 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610385 EP 1610393 DI 10.3389/pore.2022.1610385 PG 9 ER PT J AU Kahan, Zs Szanto, I Dudas, R Kapitany, Zs Molnar, M Koncz, Zs Mailath, M AF Kahan, Zsuzsanna Szanto, Istvan Dudas, Rita Kapitany, Zsuzsanna Molnar, Maria Koncz, Zsuzsa Mailath, Monika TI Breast Cancer Survivorship Programme: Follow-Up, Rehabilitation, Psychosocial Oncology Care. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Guideline DE follow-up; healthy lifestyle; physical rehabilitation; psychosocial oncology care; social rehabilitation; side-effect management ID follow-up; healthy lifestyle; physical rehabilitation; psychosocial oncology care; social rehabilitation; side-effect management AB Follow-up includes ongoing contact with and health education of the patient, surveillance and control of the adverse effects of surgery, oncological therapies or radiotherapy, screening of metachronous cancers, and comprehensive (physical, psychological and social) patient rehabilitation, which may be enhanced by a healthy lifestyle. Primary attention should be paid to early detection and, when needed, curative treatment of local/regional tumour recurrences. Similarly, with the hope of curative solution, it is important to recognize the entity of a low-mass and relatively indolent recurrence or metastasis (oligometastasis); however, there is still no need to investigate distant metastases by routine diagnostic imaging or assess tumour markers. Below there is a list of possible sources of support, with respect to adjuvant hormone therapy continued during long-term care, social support resources, pivotal points and professional opportunities for physical and mental rehabilitation. Individual solutions for specific issues (breast cancer risk/genetic mutation, pregnancy) are provided by constantly widening options. Ideally, a complex breast cancer survivorship programme is practised by a specially trained expert supported by a cooperative team of oncologists, surgeons, breast radiologists, social workers, physiotherapists, psychooncologists and psychiatrists. The approach of follow-up should be comprehensive and holistic. C1 [Kahan, Zsuzsanna] University of Szeged, Department of OncotherapySzeged, Hungary. [Szanto, Istvan] Fejer County Szent Gyorgy HospitalSzekesfehervar, Hungary. [Dudas, Rita] University of Szeged, Department of OncotherapySzeged, Hungary. [Kapitany, Zsuzsanna] Semmelweis University, Department of PhysiotherapyBudapest, Hungary. [Molnar, Maria] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. [Koncz, Zsuzsa] Semmelweis University, Institute of Behavioural SciencesBudapest, Hungary. [Mailath, Monika] University of Debrecen, Department of OncologyDebrecen, Hungary. RP Kahan, Zs (reprint author), University of Szeged, Department of Oncotherapy, Szeged, Hungary. EM kahan.zsuzsanna@med.u-szeged.hu CR Runowicz CD, Leach CR, Henry NL, Henry KS, Mackey HT, Cowens- Alvarado RL, et al. American Cancer Society/American Society of Clinical Oncology Breast Cancer Survivorship Care Guideline. J Clin Oncol, 2016, 34: 611–35., DOI 10.1200/jco.2015.64.3809 Giordano SH, Elias AD, Gradishar WJ. NCCN Guidelines Updates: Breast Cancer. J Natl Compr Canc Netw, 2018, 16(5S):605–10., DOI 10.6004/jnccn. 2018.0043 Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, et al. Early Breast Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol, 2019, 30:1674., DOI 10.1093/annonc/ mdz189 Waggoner J, Carline JD, Durning SJ. Is There a Consensus on Consensus Methodology? Descriptions and Recommendations for Future Consensus Research. Acad Med, 2016, 91:663–8., DOI 10.1097/acm.0000000000001092 Gilligan T, Coyle N, Frankel RM, Berry DL, Bohlke K, Epstein RM, et al. Patient-Clinician Communication: American Society of Clinical Oncology Consensus Guideline. J Clin Oncol, 2017, 35:3618–32., DOI 10.1200/jco.2017. 75.2311 Heitz AE, Baumgartner RN, Baumgartner KB, Boone SD. Healthy Lifestyle Impact on Breast Cancer-specific and All-Cause Mortality. Breast Cancer Res Treat, 2018, 167:171–81., DOI 10.1007/s10549-017-4467-2 Zeinomar N, Knight JA, Genkinger JM, Phillips KA, Daly MB, Milne RL, et al. Alcohol Consumption, Cigarette Smoking, and Familial Breast Cancer Risk: Findings from the Prospective Family Study Cohort, ProF-SC). Breast Cancer Res, 2019, 21:128., DOI 10.1186/s13058-019-1213-1 Hattori M, Iwata H. Advances in Treatment and Care in Metastatic Breast Cancer, MBC): Are There MBC Patients Who are Curable? Chin Clin Oncol, 2018, 7(23):23., DOI 10.21037/cco.2018.05.01 deSouza NM, Tempany CM. A Risk-based Approach to Identifying Oligometastatic Disease on Imaging. Int J Cancer, 2019, 144:422–30., DOI 10.1002/ijc.31793 deSouza NM, Liu Y, Chiti A, Oprea-Lager D, Gebhart G, Van Beers BE, et al. Strategies and Technical Challenges for Imaging Oligometastatic Disease: Recommendations from the European Organisation for Research and Treatment of Cancer Imaging Group. Eur J Cancer, 2019, 91:153–63., DOI 10.1016/j.ejca.2017.12.012 Shapiro CL, Van Poznak C, Lacchetti C, Kirshner J, Eastell R, Gagel R, et al. Management of Osteoporosis in Survivors of Adult Cancers with Nonmetastatic Disease: ASCO Clinical Practice Guideline. J Clin Oncol, 2019, 37:2916–46., DOI 10.1200/jco.19.01696 Paice JA, Portenoy R, Lacchetti C, Campbell T, Cheville A, Citron M, et al. Management of Chronic Pain in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol, 2019, 34(27): 3325–45., DOI 10.1200/JCO.2016.68.5206 Andersen BL, DeRubeis RJ, Berman BS, Gruman J, Champion VL, Massie MJ, et al. Screening, Assessment, and Care of Anxiety and Depressive Symptoms in Adults with Cancer: An American Society of Clinical Oncology Guideline Adaptation. J Clin Oncol, 2014, 32:1605–19., DOI 10. 1200/jco.2013.52.4611 Jakes AD, Twelves C. Breast Cancer-Related Lymphoedema and Venepuncture: A Review and Evidence-Based Recommendations. Breast Cancer Res Treat, 2015, 154:455–61., DOI 10.1007/s10549-015-3639-1 Zamorano JL, Lancellotti P, Rodriguez Munoz D, Aboyans V, Asteggiano R, Galderisi M, et al. 2016 ESC Position Paper on Cancer Treatments and Cardiovascular Toxicity Developed under the Auspices of the ESC Committee for Practice Guidelines: The Task Force for Cancer Treatments and Cardiovascular Toxicity of the European Society of Cardiology, ESC). Eur Heart J, 2016, 37:2768–801., DOI 10.1093/eurheartj/ehw211 Armenian SH, Lacchetti C, Barac A, Carver J, Constine LS, Denduluri N, et al. Prevention and Monitoring of Cardiac Dysfunction in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol, 2017, 35:893–911., DOI 10.1200/jco.2016.70.5400 Bower JE, Bak K, Berger A, Breitbart W, Escalante CP, Ganz PA, et al. Screening, Assessment, and Management of Fatigue in Adult Survivors of Cancer: An American Society of Clinical Oncology Clinical Practice Guideline Adaptation. J Clin Oncol, 2014, 32:1840–50., DOI 10.1200/jco. 2013.53.4495 Riba MB, Donovan KA, Andersen B, Braun I, Breitbart WS, Brewer BW, et al. Distress Management, Version 3.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw, 2019, 17:1229–49., DOI 10.6004/jnccn. 2019.0048 Cascella M, Di Napoli R, Carbone D, Cuomo GF, Bimonte S, Muzio MR. Chemotherapy-related Cognitive Impairment: Mechanisms, Clinical Features and Research Perspectives. Recenti Prog Med, 2018, 109:523–30., DOI 10.1701/ 3031.30289 Szentmartoni G, Makkos Z, Dank M. Chemobrain. Neuropsychopharmacol Hung, 2018, 20:112–6. Biro E, Kahan Z, Kalman J, Rusz O, Pakaski M, Irinyi T, et al. Cognitive Functioning and Psychological Well-Being in Breast Cancer Patients on Endocrine Therapy. In Vivo, 2019, 33:1381–92., DOI 10.21873/invivo. 11615 Carter J, Lacchetti C, Andersen BL, Barton DL, Bolte S, Damast S, et al. Interventions to Address Sexual Problems in People with Cancer: American Society of Clinical Oncology Clinical Practice Guideline Adaptation of Cancer Care Ontario Guideline. J Clin Oncol, 2018, 36:492–511., DOI 10.1200/jco.2017. 75.8995 Mazzarello S, Hutton B, Ibrahim MFK, Jacobs C, Shorr R, Smith S, et al. Management of Urogenital Atrophy in Breast Cancer Patients: A Systematic Review of Available Evidence from Randomized Trials. Breast Cancer Res Treat, 2015, 152:1–8., DOI 10.1007/s10549-015-3434-z Areas F, Valadares ALR, Conde DM, Costa-Paiva L. The Effect of Vaginal Erbium Laser Treatment on Sexual Function and Vaginal Health in Women with aHistory of Breast Cancer and Symptoms of the Genitourinary Syndrome of Menopause. Menopause, 2019, 26:1052–8., DOI 10.1097/gme. 0000000000001353 Forbes C, Fayter D, de Kock S, Quek RGW. A Systematic Review of International Guidelines and Recommendations for the Genetic Screening, Diagnosis, Genetic Counseling, and Treatment of BRCAMutated Breast Cancer. Cancer Manag Res, 2019, 11:2321–37., DOI 10. 2147/cmar.s189627 Singer CF, Balmana J, Burki N, Delaloge S, Filieri ME, Gerdes A-M, et al. Genetic Counselling and Testing of Susceptibility Genes for Therapeutic Decision-Making in Breast Cancer-An European Consensus Statement and Expert Recommendations. Eur J Cancer, 2019, 106:54–60., DOI 10.1016/j.ejca. 2018.10.007 Paluch-Shimon S, Cardoso F, Sessa C, Balmana J, Cardoso MJ, Gilbert F, et al. Prevention and Screening in BRCA Mutation Carriers and Other Breast/ ovarian Hereditary Cancer Syndromes: ESMO Clinical Practice Guidelines for Cancer Prevention and Screening. Ann Oncol, 2016, 27(Suppl. 5):v103., DOI 10. 1093/annonc/mdw327 Oktay K, Harvey BE, Partridge AH, Quinn GP, Reinecke J, Taylor HS, et al. Fertility Preservation in Patients with Cancer: ASCO Clinical Practice Guideline Update. J Clin Oncol, 2018, 36:1994–2001., DOI 10.1200/jco.2018. 78.1914 Snowden A, Young J, White C, Murray E, Richard C, Lussier M-T, et al. Evaluating Holistic Needs Assessment in Outpatient Cancer Care-Aa Randomised Controlled Trial: The Study Protocol. BMJ Open, 2015, 5: e006840., DOI 10.1136/bmjopen-2014-006840 Johnston L, Young J, Campbell K. The Implementation and Impact of Holistic Needs Assessments for People Affected by Cancer: A Systematic Review and Thematic Synthesis of the Literature. Eur J Cancer Care, Engl,, 2019, 28: e13087., DOI 10.1111/ecc.13087 Wagner JA. The Ever-Changing Face of Breast Advocacy. Breast, 2010, 19: 280–3., DOI 10.1016/j.breast.2010.03.016 Kozma J, editor. Kezikonyv a Szocialis Munka Gyakorlatahoz. Budapest: Szocialis Szakmai Szovetseg, 2001). [Kozma, J, editor. Handbook for Social Work Practice. Budapest: Association of Social Professionals, 2001)]. Mewes JC, Steuten LMG, Groeneveld IF, de Boer AGEM, Frings-Dresen MHW, IJzerman MJ, et al. Return-to-work Intervention for Cancer Survivors: Budget Impact and Allocation of Costs and Returns in the Netherlands and Six Major EU-Countries. BMC Cancer, 2015, 15:899., DOI 10.1186/s12885-015-1912-7 de Boer AG, Taskila TK, Tamminga SJ, Feuerstein M, Frings-Dresen MH, Verbeek JH. Interventions to Enhance Return-To-Work for Cancer Patients. Cochrane Database Syst Rev, 2015, 9:CD007569., DOI 10.1002/14651858. CD007569.pub3 Rock CL, Doyle C, Demark-Wahnefried W, Meyerhardt J, Courneya KS, Schwartz AL, et al. Nutrition and Physical Activity Guidelines for Cancer Survivors. CA Cancer J Clin, 2012, 62:242–74., DOI 10.3322/caac.21142 Jung AY, Behrens S, Schmidt M, Thoene K, Obi N, Husing A, et al. Pre-to Postdiagnosis Leisure-Time Physical Activity and Prognosis in Postmenopausal Breast Cancer Survivors. Breast Cancer Res, 2019, 21:117., DOI 10.1186/s13058-019-1206-0 Fukushima KFP, Carmo LA, Borinelli AC, Ferreira CWS. Frequency and Associated Factors of Axillary Web Syndrome in Women Who Had Undergone Breast Cancer Surgery: A Transversal and Retrospective Study. SpringerPlus, 2015, 4:112., DOI 10.1186/s40064-015-0889-7 Neil-Sztramko SE, Kirkham AA, Hung SH, Niksirat N, Nishikawa K, Campbell KL. Aerobic Capacity and Upper Limb Strength are Reduced in Women Diagnosed with Breast Cancer: A Systematic Review. J Physiother, 2014, 60: 189–200., DOI 10.1016/j.jphys.2014.09.005 Johansen S, Fossa K, Nesvold IL, Malinen E, Fossa SD. Arm and Shoulder Morbidity Following Surgery and Radiotherapy for Breast Cancer. Acta Oncol, 2014, 53:521–9., DOI 10.3109/0284186x.2014.880512 Luz C, Deitos J, Siqueira T, Palu M, Heck A. Management of Axillary Web Syndrome after Breast Cancer: Evidence-Based Practice. Rev Bras Ginecol Obstet, 2017, 39:632–9., DOI 10.1055/s-0037-1604181 Bruce J, Williamson E, Lait C, Richmond H, Betteley L, Lall R, et al. Randomised Controlled Trial of Exercise to Prevent Shoulder Problems in Women Undergoing Breast Cancer Treatment: Study Protocol for the Prevention of Shoulder Problems Trial, UK PROSPER). BMJ Open, 2018, 8:e019078., DOI 10.1136/bmjopen-2017-019078 Harrington S, Michener LA, Kendig T, Miale S, George SZ. Patient-Reported Upper Extremity Outcome Measures Used in Breast Cancer Survivors: A Systematic Review. Arch Phys Med Rehabil, 2014, 95:153–62., DOI 10.1016/j. apmr.2013.07.022 Binkley JM, Stratford P, Kirkpatrick S, Farley CR, Okoli J, Gabram S. Estimating the Reliability and Validity of the Upper Extremity Functional index in Women after Breast Cancer Surgery. Clin Breast Cancer, 2018, 18: e1267., DOI 10.1016/j.clbc.2018.02.008 Campbell KL, Pusic AL, Zucker DS, McNeely ML, Binkley JM, Cheville AL, et al. A Prospective Model of Care for Breast Cancer Rehabilitation: Function. Cancer, 2012, 118:2300–11., DOI 10.1002/cncr.27464 Andrade Ortega JA, Millan Gomez AP, Ribeiro Gonzalez M, Martinez Piro P, Jimenez Anula J, Sanchez Andujar MB. Validation of the FACT-B+4-UL Questionnaire and Exploration of its Predictive Value in Women Submitted to Surgery for Breast Cancer. Med Clin, English Edition,, 2017, 148:555–8., DOI 10.1016/j.medcle.2017.05.015 Torres Lacomba M, Yuste Sanchez MJ, Zapico Goni A, Prieto Merino D, Mayoral del Moral O, Cerezo Tellez E, et al. Effectiveness of Early Physiotherapy to Prevent Lymphoedema after Surgery for Breast Cancer: Randomised, Single Blinded, Clinical Trial. BMJ, 2010, 340:b5396., DOI 10. 1136/bmj.b5396 Mobarakeh ZS, Mokhtari-Hesari P, Lotfi-Tokaldany M, Montazeri A, Heidari M, Zekri F. Combined Decongestive Therapy and Reduction of Pain and Heaviness in Patients with Breast Cancer-Related Lymphedema. Support Care Cancer, 2019, 27:3805–11., DOI 10.1007/ s00520-019-04681-9 Holland J, Watson M, Dunn J. The IPOS New International Standard of Quality Cancer Care: Integrating the Psychosocial Domain into Routine Care. Psycho Oncol, 2011, 20:677–80., DOI 10.1002/pon.1978 Risko A. Az Onkopszichologia Szakmai Protokollja. In: Bagdy E, Tury F, editors. A Klinikai Es Mentalhigieniai Szakpszichologia Szakmai Protokollja, Masodik, Atdolgozott Valtozat). Budapest: Animula Kiado, 2005). [Risko A. Professional Protocols for Oncopsychology. In: Bagdy E, Tury F, editors. Professional Protocols of Clinical and Mental Health Psychology [Second Revision]. Budapest: Animula Kiado, 2005)]. Professional Guidelines on Oncopsychological Care of the Ministry of Human Resources. Health Bulletin., 2021). LXXI. Available at: http:// www.kozlonyok.hu/kozlonyok/Kozlonyok/6/PDF/2021/19.pdf, Accessed October 28, 2021). Gradishar WJ, Anderson BO, Abraham J, Aft R, Agnese D, Allison KH, et al. Breast Cancer, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw, 2020, 18:452–78., DOI 10.6004/jnccn. 2020.0016 Khatcheressian JL, Hurley P, Bantug E, Esserman LJ, Grunfeld E, Halberg F, et al. Breast Cancer Follow-Up and Management after Primary Treatment: American Society of Clinical Oncology Clinical Practice Guideline Update. J Clin Oncol, 2013, 31:961–5., DOI 10.1200/jco.2012.45.9859 Hershman DL, Lacchetti C, Dworkin RH, Lavoie Smith EM, Bleeker J, Cavaletti G, et al. Prevention and Management of Chemotherapy-Induced Peripheral Neuropathy in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol, 2014, 32:1941–67., DOI 10.1200/jco.2013.54.0914 Janelsins MC, Kohli S, Mohile SG, Usuki K, Ahles TA,Morrow GR. An Update on Cancer- and Chemotherapy-Related Cognitive Dysfunction: Current Status. Semin Oncol, 2011, 38:431–8., DOI 10.1053/j.seminoncol.2011.03.014 Reid-Arndt SA, Yee A, Perry MC, Hsieh C. Cognitive and Psychological Factors Associated with Early Posttreatment Functional Outcomes in Breast Cancer Survivors. J Psychosocial Oncol, 2009, 27:415–34., DOI 10.1080/ 07347330903183117 Bultz BD, Carlson LE. Emotional Distress: the Sixth Vital Sign-Future Directions in Cancer Care. Psycho Oncol, 2006, 15:93–5., DOI 10.1002/pon.1022 Letters endorsing the IPOS Statement on Quality Cancer Care, website,, 2021). Available at: https://ipos-society.org/endorsements/organizations, Accessed October 15, 2021). Harju E, Michel G, Roser K. A Systematic Review on the Use of the Emotion Thermometer in Individuals Diagnosed with Cancer. Psycho Oncol, 2019, 28: 1803–18., DOI 10.1002/pon.5172 Hinz A, Mitchell AJ, Degi CL, Mehnert-Theuerkauf A. Normative Values for the Distress Thermometer, DT, and the Emotion Thermometers, ET), Derived from a German General Population Sample. Qual Life Res, 2019, 28:277–82., DOI 10.1007/s11136-018-2014-1 Muszbek K, Szekely A, Balogh EM, Molnar M, Rohanszky M, Ruzsa A, et al. Validation of the Hungarian Translation of Hospital Anxiety and Depression Scale. Qual Life Res, 2006, 15:761–6., DOI 10.1007/s11136-005-3627-8 Millar K, Jelicic M, Bonke B, Asbury AJ. Assessment of Preoperative Anxiety: Comparison of Measures in Patients Awaiting Surgery for Breast Cancer. Br J Anaesth, 1995, 74:180–3., DOI 10.1093/bja/74.2.180 Bjelland I, Dahl AA, Haug TT, Neckelmann D. The Validity of the Hospital Anxiety and Depression Scale. J Psychosomatic Res, 2002, 52:69–77., DOI 10. 1016/s0022-3999(01)00296-3 NCCN. NCCN Guidelines Version 1.2020 Distress Management. National Comprehensive Cancer Network, 2020). Forrai G, Kovacs E, Ambrozay E, Barta M, Bobely K, Lengyel Z, et al. Use of Diagnostic Imaging Modalities in Modern Screening, Diagnostics and Management of Breast Tumours. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022). in press. Cserni G, Francz M, Jaray B, Kalman E, Kovacs I, Krenacs T, et al. Pathological Diagnosis, Work-Up and Reporting of Breast Cancer 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022). in press. Matrai Z, Kelemen P, Kosa C, Maraz R, Paszt A, Pavlovics G, et al. Modern Breast Cancer Surgery. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022). in press. Rubovszky G, Kocsis J, Boer K, Chilingirova N, Dank M, Kahan Z, et al. Systemic Treatment of Breast Cancer. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022). in press. Polgar C, Kahan Z, Ivanov O, Chorvath M, Ligacova A, Csejtei A, et al. Radiotherapy of Breast Cancer-Professional Guideline. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022). in press. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610391 EP 1610404 DI 10.3389/pore.2022.1610391 PG 14 ER PT J AU Siozopoulou, V Marcq, E De Winne, K Norga, K Schmitz, G Duwel, V Delvenne, P Smits, E Pauwels, P AF Siozopoulou, Vasiliki Marcq, Elly De Winne, Koen Norga, Koen Schmitz, Gertjan Duwel, Valerie Delvenne, Philippe Smits, Evelien Pauwels, Patrick TI NTRK Fusions in a Sarcomas Series: Pathology, Molecular and Clinical Aspects SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE sarcoma; prognosis; NTRK fusion; TRK immunohistochemistry; NTRK fusion partner; histology ID sarcoma; prognosis; NTRK fusion; TRK immunohistochemistry; NTRK fusion partner; histology AB Targeting molecular alterations has been proven to be an inflecting point in tumor treatment. Especially in recent years, inhibitors that target the tyrosine receptor kinase show excellent response rates and durable effects in all kind of tumors that harbor fusions of one of the three neurotrophic tyrosine receptor kinase genes (NTRK1, NTRK2 and NTRK3). Today, the therapeutic options in most metastatic sarcomas are rather limited. Therefore, identifying which sarcoma types are more likely to harbor these targetable NTRK fusions is of paramount importance. At the moment, identification of these fusions is solely based on immunohistochemistry and confirmed by molecular techniques. However, a first attempt has been made to describe the histomorphology of NTRK-fusion positive sarcomas, in order to pinpoint which of these tumors are the best candidates for testing. In this study, we investigate the immunohistochemical expression of pan-TRK in 70 soft tissue and bone sarcomas. The pan-TRK positive cases were further investigated with molecular techniques for the presence of a NTRK fusion. Seven out of the 70 cases showed positivity for pan-TRK, whereas two of these seven cases presented an NTRK3 fusion. Further analysis of the fused sarcomas revealed some unique histological, molecular and clinical findings. The goal of this study is to expand the histomorphological spectrum of the NTRK-fused sarcomas, to identify their fusion partners and to correlate these parameters with the clinical outcome of the disease. In addition, we evaluated the immunohistochemical expression pattern of the pan-TRK and its correlation with the involved NTRK gene. C1 [Siozopoulou, Vasiliki] Antwerp University Hospital, Department of PathologyEdegem, Belgium. [Marcq, Elly] University of Antwerp, Integrated Personalized and Precision Oncology Network (IPPON), Center for Oncological Research (CORE)Wilrijk, Belgium. [De Winne, Koen] Antwerp University Hospital, Department of PathologyEdegem, Belgium. [Norga, Koen] Antwerp University Hospital, Department of PediatricsEdegem, Belgium. [Schmitz, Gertjan] Hospital of Klina, Department of OrthopaedicsAntwerp, Belgium. [Duwel, Valerie] Hospital of Klina, Department of PathologyAntwerp, Belgium. [Delvenne, Philippe] CHU Sart Tilman, Department of AnatomopathologyLiege, Belgium. [Smits, Evelien] University of Antwerp, Integrated Personalized and Precision Oncology Network (IPPON), Center for Oncological Research (CORE)Wilrijk, Belgium. [Pauwels, Patrick] Antwerp University Hospital, Department of PathologyEdegem, Belgium. RP Siozopoulou, V (reprint author), Antwerp University Hospital, Department of Pathology, Edegem, Belgium. EM vasiliki.siozopoulou@uantwerpen.be CR Gamboa AC, Gronchi A, Cardona K. Soft-tissue Sarcoma in Adults: An Update on the Current State of Histiotype-specific Management in an Era of Personalized Medicine. CA A Cancer J Clin, 2020, 70(3):200–29., DOI 10. 3322/caac.21605 Dangoor A, Seddon B, Gerrand C, Grimer R, Whelan J, Judson I. UK Guidelines for the Management of Soft Tissue Sarcomas. Clin Sarcoma Res, 2016, 6:20., DOI 10.1186/s13569-016-0060-4 Gerrand C, Athanasou N, Athanasou N, Brennan B, Grimer R, Judson I, et al. UK Guidelines for the Management of Bone Sarcomas. Clin Sarcoma Res, 2016, 6:7., DOI 10.1186/s13569-016-0047-1 Bessen T, Caughey GE, Shakib S, Potter JA, Reid J, Farshid G, et al. A Population-Based Study of Soft Tissue Sarcoma Incidence and Survival in Australia: An Analysis of 26,970 Cases. Cancer Epidemiol, 2019, 63:101590., DOI 10.1016/j.canep.2019.101590 Reichardt P. The Story of Imatinib in GIST - a Journey through the Development of a Targeted Therapy. Oncol Res Treat, 2018, 41(7-8):472–7., DOI 10.1159/000487511 Drilon A. TRK Inhibitors in TRK Fusion-Positive Cancers. Ann Oncol, 2019, 30(Suppl. 8):viii23–viii30., DOI 10.1093/annonc/mdz282 Drilon A, Nagasubramanian R, Blake JF, Ku N, Tuch BB, Ebata K, et al. A Next-Generation TRK Kinase Inhibitor Overcomes Acquired Resistance to Prior TRK Kinase Inhibition in Patients with TRK Fusion-Positive Solid Tumors. Cancer Discov, 2017, 7(9):963–72., DOI 10.1158/2159-8290.CD-17- 0507 Drilon A, Ou S-HI, Cho BC, Kim D-W, Lee J, Lin JJ, et al. Repotrectinib, TPX- 0005, Is a Next-Generation ROS1/TRK/ALK Inhibitor that Potently Inhibits ROS1/TRK/ALK Solvent- Front Mutations. Cancer Discov, 2018, 8(10): 1227–36., DOI 10.1158/2159-8290.CD-18-0484 Cocco E, Scaltriti M, Drilon A. NTRK Fusion-Positive Cancers and TRK Inhibitor Therapy. Nat Rev Clin Oncol, 2018, 15(12):731–47., DOI 10.1038/ s41571-018-0113-0 Siozopoulou V, Smits E, De Winne K, Marcq E, Pauwels P. NTRK Fusions in Sarcomas: Diagnostic Challenges and Clinical Aspects. Diagnostics, 2021, 11(3):478., DOI 10.3390/diagnostics11030478 Gatalica Z, Xiu J, Swensen J, Vranic S. Molecular Characterization of Cancers with NTRK Gene Fusions. Mod Pathol, 2019, 32(1):147–53., DOI 10.1038/ s41379-018-0118-3 De Winne K, Sorber L, Lambin S, Siozopoulou V, Beniuga G, Dedeurwaerdere F, et al. Immunohistochemistry as a Screening Tool for NTRK Gene Fusions: Results of a First Belgian Ring Trial. Virchows Arch, 2021, 478(2):283–91., DOI 10.1007/s00428-020-02921-6 Detre S, Saclani Jotti G, Dowsett M. A "quickscore" Method for Immunohistochemical Semiquantitation: Validation for Oestrogen Receptor in Breast Carcinomas. J Clin Pathol, 1995, 48(9):876–8., DOI 10.1136/jcp.48. 9.876 Kummar S, Lassen UN. TRK Inhibition: A New Tumor-Agnostic Treatment Strategy. Targ Oncol, 2018, 13(5):545–56., DOI 10.1007/s11523-018-0590-1 Vaishnavi A, Le AT, Doebele RC. TRKing Down an Old Oncogene in a new era of Targeted Therapy. Cancer Discov, 2015, 5(1):25–34., DOI 10.1158/2159- 8290.CD-14-0765 Hong DS, Bauer TM, Lee JJ, Dowlati A, Brose MS, Farago AF, et al. Larotrectinib in Adult Patients with Solid Tumours: a multi-centre, Open- Label, Phase I Dose-Escalation Study. Ann Oncol, 2019, 30(2):325–31., DOI 10. 1093/annonc/mdy539 Demetri GD, Antonescu CR, Bjerkehagen B, Bovee JVMG, Boye K, Chacon M, et al. Diagnosis and Management of Tropomyosin Receptor Kinase, TRK, Fusion Sarcomas: Expert Recommendations from the World Sarcoma Network. Ann Oncol, 2020, 31(11):1506–17., DOI 10.1016/j.annonc.2020.08. 2232 Kao YC, Suurmeijer AJH, Argani P, Dickson BC, Zhang L, Sung YS, et al. Soft Tissue Tumors Characterized by a Wide Spectrum of Kinase Fusions Share a Lipofibromatosis-like Neural Tumor Pattern. Genes Chromosomes Cancer, 2020, 59(10):575–83., DOI 10.1002/gcc.22877 Agaram NP, Zhang L, Sung Y-S, Chen C-L, Chung CT, Antonescu CR, et al. Recurrent NTRK1 Gene Fusions Define a Novel Subset of Locally Aggressive Lipofibromatosis-like Neural Tumors. Am J Surg Pathol, 2016, 40(10): 1407–16., DOI 10.1097/PAS.0000000000000675 Haller F, Knopf J, Ackermann A, Bieg M, Kleinheinz K, Schlesner M, et al. Paediatric and Adult Soft Tissue Sarcomas withNTRK1gene Fusions: a Subset of Spindle Cell Sarcomas Unified by a Prominent Myopericytic/ haemangiopericytic Pattern. J Pathol, 2016, 238(5):700–10., DOI 10.1002/ path.4701 Croce S, Hostein I, Longacre TA, Mills AM, Perot G, Devouassoux- Shisheboran M, et al. Uterine and Vaginal Sarcomas Resembling Fibrosarcoma: a Clinicopathological and Molecular Analysis of 13 Cases Showing Common NTRK-Rearrangements and the Description of a COL1A1-PDGFB Fusion Novel to Uterine Neoplasms. Mod Pathol, 2019, 32(7):1008–22., DOI 10.1038/s41379-018-0184-6 Chiang S, Cotzia P, Hyman DM, Drilon A, Tap WD, Zhang L, et al. NTRK Fusions Define a Novel Uterine Sarcoma Subtype with Features of Fibrosarcoma. Am J Surg Pathol, 2018, 42(6):791–8., DOI 10.1097/PAS. 0000000000001055 Rabban JT, Devine WP, Sangoi AR, Poder L, Alvarez E, Davis JL, et al. NTRK Fusion Cervical Sarcoma: a Report of Three Cases, Emphasising Morphological and Immunohistochemical Distinction from Other Uterine Sarcomas, Including Adenosarcoma. Histopathology, 2020, 77(1):100–11., DOI 10.1111/his.14069 Alassiri AH, Ali RH, Shen Y, Lum A, Strahlendorf C, Deyell R, et al. ETV6- NTRK3 Is Expressed in a Subset of ALK-Negative Inflammatory Myofibroblastic Tumors. Am J Surg Pathol, 2016, 40(8):1051–61., DOI 10. 1097/PAS.0000000000000677 Yamamoto H, Yoshida A, Taguchi K, Kohashi K, Hatanaka Y, Yamashita A, et al. ALK,ROS1andNTRK3gene Rearrangements in Inflammatory Myofibroblastic Tumours. Histopathology, 2016, 69(1):72–83., DOI 10.1111/ his.12910 Olson N, Rouhi O, Zhang L, Angeles C, Bridge J, Lopez-Terrada D, et al. A Novel Case of an Aggressive Superficial Spindle Cell Sarcoma in an Adult Resembling Fibrosarcomatous Dermatofibrosarcoma Protuberans and Harboring anEML4-NTRK3fusion. J Cutan Pathol, 2018, 45(12):933–9., DOI 10.1111/cup.13348 So YK, Chow C, To KF, Chan JKC, Cheuk W. Myxoid Spindle Cell Sarcoma with LMNA-NTRK Fusion: Expanding the Morphologic Spectrum of NTRKRearranged Tumors. Int J Surg Pathol, 2020, 28(5):574–8., DOI 10.1177/ 1066896920905888 Kao Y-C, Flucke U, Eijkelenboom A, Zhang L, Sung Y-S, Suurmeijer AJH, et al. Novel EWSR1-SMAD3 Gene Fusions in a Group of Acral Fibroblastic Spindle Cell Neoplasms. Am J Surg Pathol, 2018, 42(4):522–8., DOI 10.1097/PAS. 0000000000001002 Brcic I, Godschachner TM, Bergovec M, Igrec J, Till H, Lackner H, et al. Broadening the Spectrum of NTRK Rearranged Mesenchymal Tumors and Usefulness of Pan-TRK Immunohistochemistry for Identification of NTRK Fusions. Mod Pathol, 2021, 34(2):396–407., DOI 10.1038/s41379-020- 00657-x Davis JL, Lockwood CM, Stohr B, Boecking C, Al-Ibraheemi A, DuBois SG, et al. Expanding the Spectrum of Pediatric NTRK-Rearranged Mesenchymal Tumors. Am J Surg Pathol, 2019, 43(4):435–45., DOI 10.1097/PAS. 0000000000001203 Hechtman JF, Benayed R, Hyman DM, Drilon A, Zehir A, Frosina D, et al. Pan-Trk Immunohistochemistry Is an Efficient and Reliable Screen for the Detection of NTRK Fusions. Am J Surg Pathol, 2017, 41(11):1547–51., DOI 10. 1097/PAS.0000000000000911 Solomon JP, Benayed R, Hechtman JF, Ladanyi M. Identifying Patients with NTRK Fusion Cancer. Ann Oncol, 2019, 30(Suppl. l_8):viii16–viii22., DOI 10. 1093/annonc/mdz384 Nozzoli F, Lazar AJ, Castiglione F, Campanacci DA, Beltrami G, De Logu F, et al. NTRK Fusions Detection in Paediatric Sarcomas to Expand the Morphological Spectrum and Clinical Relevance of Selected Entities. Pathol Oncol Res. 2022;28., DOI 10.3389/pore.2022.1610237 Rudzinski ER, Lockwood CM, Stohr BA, Vargas SO, Sheridan R, Black JO, et al. Pan-Trk Immunohistochemistry Identifies NTRK Rearrangements in Pediatric Mesenchymal Tumors. Am J Surg Pathol, 2018, 42(7):927–35., DOI 10.1097/PAS.0000000000001062 Solomon JP, Linkov I, Rosado A, Mullaney K, Rosen EY, Frosina D, et al. NTRK Fusion Detection across Multiple Assays and 33,997 Cases: Diagnostic Implications and Pitfalls. Mod Pathol, 2020, 33(1):38–46., DOI 10.1038/s41379- 019-0324-7 Hung YP, Fletcher CDM, Hornick JL. Evaluation of Pan-TRK Immunohistochemistry in Infantile Fibrosarcoma, Lipofibromatosis-like Neural Tumour and Histological Mimics. Histopathology, 2018, 73(4): 634–44., DOI 10.1111/his.13666 Csanyi-Bastien M, Lanic M-D, Beaussire L, Ferric S, Francois A, Meseure D, et al. Pan-TRK Immunohistochemistry Is Highly Correlated with NTRK3 Gene Rearrangements in Salivary Gland Tumors. Am J Surg Pathol, 2021, 45: 1487–98., DOI 10.1097/PAS.0000000000001718 Suurmeijer AJH, Dickson BC, Swanson D, Zhang L, Sung Y-S, Cotzia P, et al. A Novel Group of Spindle Cell Tumors Defined by S100 and CD34 Coexpression Shows Recurrent Fusions Involving RAF1, BRAF, and NTRK1/2 Genes. Genes Chromosomes Cancer, 2018, 57(12):611–21., DOI 10.1002/gcc. 22671 Suurmeijer AJ, Dickson BC, Swanson D, Zhang L, Sung YS, Huang HY, et al. The Histologic Spectrum of Soft Tissue Spindle Cell Tumors with NTRK3 Gene Rearrangements. Genes Chromosomes Cancer, 2019, 58(11):739–46., DOI 10.1002/gcc.22767 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610423 EP 1610430 DI 10.3389/pore.2022.1610423 PG 8 ER PT J AU Fogarasi, IA Benczik, M Moravcsik-Kornyicki, Kocsis, A Gyulai, A Kosa, Zs AF Fogarasi, Istvan Andras Benczik, Marta Moravcsik-Kornyicki, Agota Kocsis, Adrienn Gyulai, Aniko Kosa, Zsigmond TI The Prevalence of High-Risk Human Papillomavirus in Hungary —A Geographically Representative, Cross-Sectional Study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cervical cancer; cancer screening; cross-sectional studies; genotype; human papillomavirus infection; human papillomavirus vaccines; prevalence studies; rural population ID cervical cancer; cancer screening; cross-sectional studies; genotype; human papillomavirus infection; human papillomavirus vaccines; prevalence studies; rural population AB Background: The estimated age-standardized incidence and mortality rates of cervical cancer in Hungary are substantially higher than the European average. In many countries, human papillomavirus (HPV) testing is the first-line method of cervical cancer screening in women >30 years. According to the European guidelines, evidence-based improvement of a national prevention strategy requires the monitoring of representative data. Methods: ThinPrep cervical samples were collected over a period of 8months at 84 sampling sites, including 4,000 eligible samples with valid laboratory results from the screening target population of females aged 25–65 years, with addresses in the representative geographic area (19 counties and four major settlement types). Genotyping of high-risk HPV (hrHPV) was performed using the Confidence HPV-X (Neumann Diagnostics) and Linear Array HPV Genotyping (Roche) tests. Demographic data were collected using a questionnaire, enabling the analysis of hrHPV genotype distribution by age, geography, education, and HPV vaccination. Results: Overall, 446 samples were hrHPV-positive, showing a prevalence of 11.15% (9.73% age-representative), similar to the world average, higher than the European average, and lower than the Eastern-European average. After age standardization, no significant geographic differences were found, except for low hrHPV prevalence in villages (p = 0.036) and in those with elementary education (p = 0.013). Following genotypes 16 and 31, in order of frequency, certain non-vaccine hrHPV genotypes (HPV51, 66, 56) showed unexpectedly higher prevalence than international data. Conclusion: Our study provides the first geographically representative genotype-specific hrHPV prevalence baseline database in Hungary to support policy-making efforts. Significant correlations with demographic data have transferable conclusions. C1 [Fogarasi, Istvan Andras] SYNLAB Hungary Ltd., SYNLAB Genoid Molecular Diagnostic LaboratoryBudapest, Hungary. [Benczik, Marta] SYNLAB Hungary Ltd., SYNLAB Genoid Molecular Diagnostic LaboratoryBudapest, Hungary. [Moravcsik-Kornyicki, Agota] University of Debrecen, Department of Health Visitors Methodology and PreventionNyiregyhaza, Hungary. [Kocsis, Adrienn] SYNLAB Hungary Ltd., SYNLAB Genoid Molecular Diagnostic LaboratoryBudapest, Hungary. [Gyulai, Aniko] University of Miskolc, Institute of Applied Health Sciences, Department of Preventive Health SciencesMiskolc, Hungary. [Kosa, Zsigmond] University of Debrecen, Department of Health Visitors Methodology and PreventionNyiregyhaza, Hungary. RP Fogarasi, IA (reprint author), SYNLAB Hungary Ltd., SYNLAB Genoid Molecular Diagnostic Laboratory, Budapest, Hungary. EM andras.fogarasi.dr@gmail.com CR Ferlay J, Ervik M, Lam F, Colombet M, Mery L, Pineros M, et al. Global Cancer Observatory: Cancer Today. Lyon, France: International Agency for Research on Cancer, 2020). Available from: https://gco.iarc.fr/today, Accessed March 2, 2022). Ronco G, Dillner J, Elfstrom KM, Tunesi S, Snijders PJF, Arbyn M, et al. Efficacy of HPV-Based Screening for Prevention of Invasive Cervical Cancer: Follow-Up of Four European Randomised Controlled Trials. Lancet, 2014, 383:524–32., DOI 10.1016/S0140-6736(13)62218-7 von Karsa L, Arbyn M, De Vuyst H, Dillner J, Dillner L, Franceschi S, et al. European Guidelines for Quality Assurance in Cervical Cancer Screening. Summary of the Supplements on HPV Screening and Vaccination. Papillomavirus Res, 2015, 1:22–31., DOI 10.1016/j.pvr.2015.06.006 Kocsis A, Takacs T, Jeney C, Schaff Z, Koiss R, Jaray B, et al. Performance of a NewHPV and Biomarker Assay in the Management of hrHPV PositiveWomen: Subanalysis of the Ongoing Multicenter TRACE Clinical Trial, N > 6,000, to Evaluate POU4F3 Methylation as a Potential Biomarker of Cervical Precancer and Cancer. Int J Cancer, 2017, 140:1119–33., DOI 10.1002/ijc.30534 Forman D, de Martel C, Lacey CJ, Soerjomataram I, Lortet-Tieulent J, Bruni L, et al. Global burden of Human Papillomavirus and Related Diseases. Vaccine, 2012, 30(Suppl. 5):F12–F23., DOI 10.1016/j.vaccine.2012.07.055 De Vuyst H, Clifford G, Li N, Franceschi S. HPV Infection in Europe. Eur J Cancer, 2009, 45:2632–9., DOI 10.1016/j.ejca.2009.07.019 Poljak M, Seme K, Maver PJ, Kocjan BJ, Cuschieri KS, Rogovskaya SI, et al. Human Papillomavirus Prevalence and Type-Distribution, Cervical Cancer Screening Practices and Current Status of Vaccination Implementation in Central and Eastern Europe. Vaccine, 2013, 31:H59–H70., DOI 10.1016/j. vaccine.2013.03.029 Tachezy R, Smahelova J, Kaspirkova J, Salakova M. Human Papillomavirus Type-specific Prevalence in the Cervical Cancer Screening Population of Czech Women. PLOS ONE, 2013, 8:e79156., DOI 10.1371/journal.pone.0079156 IlisiuMB, HashimD, Andreassen T, StoerNC, Nicula F, Weiderpass E. HPVTesting for Cervical Cancer in Romania: High-Risk HPV Prevalence Among Ethnic Subpopulations and Regions. Ann Glob Health, 2019, 85(1):89., DOI 10.5334/aogh.2502 Nya´ri T, Cseh I, Woodward M, Szo€llo€si J, Bak M, Dea´k J. Screening for Human Papillomavirus Infection in Asymptomatic Women in Hungary. Hum Reprod, 2001, 16:2235–7., DOI 10.1093/humrep/16.10.2235 Kornya L, Cseh I, Deak J, Bak M, Fulop V. The Diagnostics and Prevalence of Genital Human Papillomavirus, HPV, Infection in Hungary. Eur J Obstet Gynecol Reprod Biol, 2002, 100:231–6., DOI 10.1016/S0301-2115(01)00474-2 Benczik M. HPV Genotypes Detected by GenoID Laboratory in Hungary at 2005/2006. [article in Hu]. STD es Genitalis Infektol, 2008, 2:10–6. Galamb A, Pajor A, Langmar Z, Sobel G. Results of the First Human Papilloma Virus center in Hungary, 2007-2011). Orvosi Hetilap, 2011, 152:1804–7., DOI 10.1556/OH.2011.29233 Szentirmay Z, Veleczki Z, Kasler M. Human Papillomavirus Associated Cervix Uteri Morbidity in Hungary: Epidemiology and Correlation with the HPV Types and the Simultaneous Cytological Diagnosis. [article in Hu]. Orvosi Hetilap, 2017, 158:1213–21., DOI 10.1556/650.2017.30807 Public Health Analysis Centre Information System, NEKIR). Methodology Development of Health Service System Project No. EFOP 1.8.0-VEKOP-17-2017- 00001. National Public Health Centre, 2019). Available from: https://efop180.antsz. hu/nepegeszsegugyi-elemzesi-kozpont/75-nepegeszsegugyi-elemzesi-kozpont/168- nekir-adatvalaszto.html, Accessed Apr 28, 2021). Lin W, Chen B, Hu H, Yuan S, Wu B, Zhong C, et al. Joint Effects of HPVRelated Knowledge and Socio-Demographic Factors on HPV Testing Behaviour Among Females in Shenzhen. Eur J Public Health, 2021, 31: 582–8., DOI 10.1093/eurpub/ckab049 Wright TC, Stoler MH, Behrens CM, Apple R, Derion T, Wright TL. The ATHENA Human Papillomavirus Study:Design, Methods, and Baseline Results. Am J Obstet Gynecol, 2012, 206:e1–e46., DOI 10.1016/j.ajog.2011.07.024 Kjaer SK, Frederiksen K, Munk C, Iftner T. Long-term Absolute Risk of Cervical Intraepithelial Neoplasia Grade 3 or Worse Following Human Papillomavirus Infection: Role of Persistence. J Natl Cancer Inst, 2010, 102:1478–88., DOI 10.1093/jnci/djq356 Lutringer-Magnin D, Kalecinski J, Cropet C, Barone G, Ronin V, Regnier V, et al. Prevention of Sexually Transmitted Infections Among Girls and Young Women in Relation to Their HPV Vaccination Status. Eur J Public Health, 2013, 23(6):1046–53., DOI 10.1093/eurpub/ckt018 Guan P, Howell-Jones R, Li N, Bruni L, de Sanjose S, Franceschi S, et al. Human Papillomavirus Types in 115,789 HPV-Positive Women: a Meta- Analysis from Cervical Infection to Cancer. Int J Cancer, 2012, 131:2349–59., DOI 10.1002/ijc.27485 Bruni L, Diaz M, Castellsague X, Ferrer E, Bosch FX, de Sanjose S. Cervical Human Papillomavirus Prevalence in 5 Continents: Meta-Analysis of 1Million Women with Normal Cytological Findings. J Infect Dis, 2010, 202:1789–99., DOI 10.1086/657321 Monsonego J, Cox JT, Behrens C, Sandri M, Franco EL, Yap P-S, et al. Prevalence of High-Risk Human Papilloma Virus Genotypes and Associated Risk of Cervical Precancerous Lesions in a Large U.S. Screening Population: Data from the ATHENA Trial. Gynecol Oncol, 2015, 137:47–54., DOI 10.1016/j. ygyno.2015.01.551 Clifford G, Gallus S, Herrero R, Munoz N, Snijders P, Vaccarella S, et al. Worldwide Distribution of Human Papillomavirus Types in Cytologically normal Women in the International Agency for Research on Cancer HPV Prevalence Surveys: A Pooled Analysis. Lancet, 2005, 366:991–8., DOI 10.1016/ S0140-6736(05)67069-9 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610424 EP 1610432 DI 10.3389/pore.2022.1610424 PG 9 ER PT J AU Zheng, P Gao, H Xie, X Lu, P AF Zheng, Peiming Gao, Huijie Xie, Xuanhu Lu, Peipei TI Plasma Exosomal hsa_circ_0015286 as a Potential Diagnostic and Prognostic Biomarker for Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; gastric cancer; diagnosis; CircRNA; exosomes ID biomarker; gastric cancer; diagnosis; CircRNA; exosomes AB Circular RNA (circRNA) is stable and abundant in exosomes as a potential biomarker for the diagnosis and prognosis of tumor. In this study, cancer specific exosomal circRNAs were identified through circRNA microarray, and 58 circRNAs were significantly upregulated in cancer cells derived exosomes. Then 60 patients with newly diagnosed gastric cancer (GC), 30 chronic gastritis patients and 30 healthy subjects were enrolled for further clinical validation. We detected that hsa_circ_0015286 was remarkably highly expressed in GC tissue, plasma and cancer cells compared with normal controls. Results of ROC curve analysis showed that the area under curve (AUC) of hsa_circ_0015286, CEA and CA 19-9 was 0.778, 0.673, and 0.665, respectively. The combined detection of three indicators had the highest AUC (0.843). Exosomal hsa_circ_0015286 expression was closely associated with tumor size, TNM stage and lymph node metastasis. The expression level of exosomal hsa_circ_0015286 in GC patients decreased significantly after surgery. Overall survival of patients with low hsa_circ_0015286 expression was longer than those with high expression. Our data demonstrated that exosomal hsa_circ_0015286 might be a promising noninvasive biomarker for the diagnosis and prognosis evaluation of GC. C1 [Zheng, Peiming] Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Department of Clinical LaboratoryZhengzhou, China. [Gao, Huijie] The First Affiliated Hospital of Henan University, Department of OncologyKaifeng, China. [Xie, Xuanhu] The First Affiliated Hospital of Henan University, Department of OncologyKaifeng, China. [Lu, Peipei] The First Affiliated Hospital of Henan University, Department of OncologyKaifeng, China. RP Zheng, P (reprint author), Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Department of Clinical Laboratory, Zhengzhou, China. EM zpm8266@163.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer Statistics in China, 2015. CA: A Cancer J Clinicians, 2016, 66:115–32., DOI 10.3322/caac. 21338 Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric Cancer. The Lancet, 2020, 396:635–48., DOI 10.1016/s0140-6736(20)31288-5 Kalluri R, LeBleu VS. The Biology, Function, and Biomedical Applications of Exosomes. New York, N.Y.: Science, 2020). p. 367. Tkach M, Thery C. Communication by Extracellular Vesicles: Where We Are and where We Need to Go. Cell, 2016, 164:1226–32., DOI 10.1016/j.cell.2016. 01.043 Ruivo CF, Adem B, Silva M, Melo SA. The Biology of Cancer Exosomes: Insights and New Perspectives. Cancer Res, 2017, 77:6480–8., DOI 10.1158/ 0008-5472.can-17-0994 Mohrmann L, Huang HJ, Hong DS, Tsimberidou AM, Fu S, Piha-Paul SA, et al. Liquid Biopsies Using Plasma Exosomal Nucleic Acids and Plasma Cellfree DNA Compared with Clinical Outcomes of Patients with Advanced Cancers. Clin Cancer Res, 2018, 24:181–8., DOI 10.1158/1078-0432.ccr-17-2007 Chen B, Huang S. Circular RNA: An Emerging Non-coding RNA as a Regulator and Biomarker in Cancer. Cancer Lett, 2018, 418:41–50., DOI 10. 1016/j.canlet.2018.01.011 Li Y, Zheng Q, Bao C, Li S, Guo W, Zhao J, et al. Circular RNA Is Enriched and Stable in Exosomes: a Promising Biomarker for Cancer Diagnosis. Cell Res, 2015, 25:981–4., DOI 10.1038/cr.2015.82 Wang M, Yu F, Li P, Wang K. Emerging Function and Clinical Significance of Exosomal circRNAs in Cancer. Mol Ther - Nucleic Acids, 2020, 21:367–83., DOI 10.1016/j.omtn.2020.06.008 Wang S, Dong Y, Gong A, Kong H, Gao J, Hao X, et al. Exosomal circRNAs as Novel Cancer Biomarkers: Challenges and Opportunities. Int J Biol Sci, 2021, 17:562–73., DOI 10.7150/ijbs.48782 Zheng P, Zhang H, Gao H, Sun J, Li J, Zhang X, et al. Plasma Exosomal Long Noncoding RNA Lnc-Slc2a12-10:1 as a Novel Diagnostic Biomarker for Gastric Cancer. Ott, 2020, 13:4009–18., DOI 10.2147/ott.s253600 Shimada H, Noie T, Ohashi M, Oba K, Takahashi Y. Clinical Significance of Serum Tumor Markers for Gastric Cancer: a Systematic Review of Literature by the Task Force of the Japanese Gastric Cancer Association. Gastric Cancer, 2014, 17:26–33., DOI 10.1007/s10120-013-0259-5 Sawaki K, Kanda M, Kodera Y. Review of Recent Efforts to Discover Biomarkers for Early Detection, Monitoring, Prognosis, and Prediction of Treatment Responses of Patients with Gastric Cancer. Expert Rev Gastroenterol Hepatol, 2018, 12:657–70., DOI 10.1080/17474124.2018. 1489233 Eusebi LH, Telese A, Marasco G, Bazzoli F, Zagari RM. Gastric Cancer Prevention Strategies: A Global Perspective. J Gastroenterol Hepatol, 2020, 35:1495–502., DOI 10.1111/jgh.15037 Siravegna G, Marsoni S, Siena S, Bardelli A. Integrating Liquid Biopsies into the Management of Cancer. Nat Rev Clin Oncol, 2017, 14:531–48., DOI 10.1038/ nrclinonc.2017.14 Sorber L, Zwaenepoel K, Deschoolmeester V, Van Schil PEY, VanMeerbeeck J, Lardon F, et al. Circulating Cell-free Nucleic Acids and Platelets as a Liquid Biopsy in the Provision of Personalized Therapy for Lung Cancer Patients. Lung Cancer, 2017, 107:100–7., DOI 10.1016/j.lungcan.2016.04.026 Kalluri R. The Biology and Function of Exosomes in Cancer. J Clin Invest, 2016, 126:1208–15., DOI 10.1172/jci81135 Jiang L, Gu Y, Du Y, Liu J. Exosomes: Diagnostic Biomarkers and Therapeutic Delivery Vehicles for Cancer. Mol Pharmaceutics, 2019, 16:3333–49., DOI 10. 1021/acs.molpharmaceut.9b00409 Lasda E, Parker R. Circular RNAs Co-precipitate with Extracellular Vesicles: A Possible Mechanism for circRNA Clearance. PloS one, 2016, 11:e0148407., DOI 10.1371/journal.pone.0148407 Bai H, Lei K, Huang F, Jiang Z, Zhou X. Exo-circRNAs: a New Paradigm for Anticancer Therapy. Mol Cancer, 2019, 18:56., DOI 10.1186/s12943-019- 0986-2 Zhang H, Zhu L, Bai M, Liu Y, Zhan Y, Deng T, et al. Exosomal circRNA Derived from Gastric Tumor Promotes white Adipose browning by Targeting the miR-133/PRDM16 Pathway. Int J Cancer, 2019, 144:2501–15., DOI 10. 1002/ijc.31977 Chen W, Quan Y, Fan S, Wang H, Liang J, Huang L, et al. Exosometransmitted Circular RNA Hsa_circ_0051443 Suppresses Hepatocellular Carcinoma Progression. Cancer Lett, 2020, 475:119–28., DOI 10.1016/j. canlet.2020.01.022 Li T, Sun X, Chen L. Exosome Circ_0044516 Promotes Prostate Cancer Cell Proliferation and Metastasis as a Potential Biomarker. J Cel Biochem, 2020, 121:2118–26., DOI 10.1002/jcb.28239 Shao Y, Tao X, Lu R, Zhang H, Ge J, Xiao B, et al. Hsa_circ_0065149 Is an Indicator for Early Gastric Cancer Screening and Prognosis Prediction. Pathol Oncol Res, 2020, 26:1475–82., DOI 10.1007/s12253-019-00716-y Tao X, Shao Y, Lu R, Ye Q, Xiao B, Ye G, et al. Clinical Significance of Hsa_circ_0000419 in Gastric Cancer Screening and Prognosis Estimation. Pathol - Res Pract, 2020, 216:152763., DOI 10.1016/j.prp.2019.152763 Tang W, Fu K, Sun H, Rong D, Wang H, Cao H. CircRNA Microarray Profiling Identifies a Novel Circulating Biomarker for Detection of Gastric Cancer. Mol Cancer, 2018, 17:137., DOI 10.1186/s12943-018-0888-8 Li J, Li Z, Jiang P, Peng M, Zhang X, Chen K, et al. Circular RNA IARS, Circ- IARS, Secreted by Pancreatic Cancer Cells and Located within Exosomes Regulates Endothelial Monolayer Permeability to Promote Tumor Metastasis. J Exp Clin Cancer Res, 2018, 37:177., DOI 10.1186/s13046-018-0822-3 Lu J, Wang Y-h., Yoon C, Huang X-y., Xu Y, Xie J-w., et al. Circular RNA Circ- RanGAP1 Regulates VEGFA Expression by Targeting miR-877-3p to Facilitate Gastric Cancer Invasion and Metastasis. Cancer Lett, 2020, 471:38–48., DOI 10. 1016/j.canlet.2019.11.038 Li L, Li W, Chen N, Zhao H, Xu G, Zhao Y, et al. FLI1 Exonic Circular RNAs as a Novel Oncogenic Driver to Promote Tumor Metastasis in Small Cell Lung Cancer. Clin Cancer Res, 2019, 25:1302–17., DOI 10.1158/1078-0432.ccr-18- 1447 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610446 EP 1610453 DI 10.3389/pore.2022.1610446 PG 8 ER PT J AU Zhang, K Gao, L Wang, J Chu, X Zhang, Z Zhang, Y Fang, F Tao, Y Li, X Tian, Y Li, Z Sang, X Ma, L Lu, L Chen, Y Yu, J Zhuo, R Wu, Sh Pan, J Hu, Sh AF Zhang, Kunlong Gao, Li Wang, Jianwei Chu, Xinran Zhang, Zimu Zhang, Yongping Fang, Fang Tao, Yanfang Li, Xiaolu Tian, Yuanyuan Li, Zhiheng Sang, Xu Ma, Li Lu, Lihui Chen, Yanling Yu, Juanjuan Zhuo, Ran Wu, Shuiyan Pan, Jian Hu, Shaoyan TI A Novel BRD Family PROTAC Inhibitor dBET1 Exerts Great Anti-Cancer Effects by Targeting c-MYC in Acute Myeloid Leukemia Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE acute myeloid leukemia; C-MYC; BRD4; dBET1; PROTAC ID acute myeloid leukemia; C-MYC; BRD4; dBET1; PROTAC AB Acute myeloid leukemia (AML) represents an aggressive hematopoietic malignancy with a prognosis inferior to that of other leukemias. Recent targeted therapies offer new opportunities to achieve better treatment outcomes. However, due to the complex heterogeneity of AML, its prognosis remains dismal. In this study, we first identified the correlation between high expression of BRD4 and overall survival of patients with AML. Targeted degradation of BRD2, BRD3, and BRD4 proteins by dBET1, a proteolysistargeting chimera (PROTAC) against the bromodomain and extra-terminal domain (BET) family members, showed cytotoxic effects on Kasumi (AML1-ETO), NB4 (PML-RARa), THP-1 (MLL-AF9), and MV4-11 (MLL-AF4) AML cell lines representing different molecular subtypes of AML. Furthermore, we determined that dBET1 treatment arrested cell cycling and enhanced apoptosis and c-MYC was identified as the downstream target. Collectively, our results indicated that dBET1 had broad anti-cancer effects on AML cell lines with different molecular lesions and provided more benefits to patients with AML. C1 [Zhang, Kunlong] Children’s Hospital of Soochow UniversitySuzhou, China. [Gao, Li] Children’s Hospital of Soochow University, Department of HaematologySuzhou, China. [Wang, Jianwei] Children’s Hospital of Soochow University, Institute of Pediatric ResearchSuzhou, China. [Chu, Xinran] Children’s Hospital of Soochow University, Department of HaematologySuzhou, China. [Zhang, Zimu] Children’s Hospital of Soochow University, Institute of Pediatric ResearchSuzhou, China. [Zhang, Yongping] Children’s Hospital of Soochow UniversitySuzhou, China. [Fang, Fang] Children’s Hospital of Soochow University, Institute of Pediatric ResearchSuzhou, China. [Tao, Yanfang] Children’s Hospital of Soochow University, Institute of Pediatric ResearchSuzhou, China. [Li, Xiaolu] Children’s Hospital of Soochow University, Institute of Pediatric ResearchSuzhou, China. [Tian, Yuanyuan] Children’s Hospital of Soochow University, Department of HaematologySuzhou, China. [Li, Zhiheng] Children’s Hospital of Soochow UniversitySuzhou, China. [Sang, Xu] Children’s Hospital of Soochow UniversitySuzhou, China. [Ma, Li] Children’s Hospital of Soochow UniversitySuzhou, China. [Lu, Lihui] Children’s Hospital of Soochow UniversitySuzhou, China. [Chen, Yanling] Children’s Hospital of Soochow UniversitySuzhou, China. [Yu, Juanjuan] Children’s Hospital of Soochow UniversitySuzhou, China. [Zhuo, Ran] Children’s Hospital of Soochow UniversitySuzhou, China. [Wu, Shuiyan] Children’s Hospital of Soochow University, Intensive Care UnitSuzhou, China. [Pan, Jian] Children’s Hospital of Soochow University, Institute of Pediatric ResearchSuzhou, China. [Hu, Shaoyan] Children’s Hospital of Soochow University, Department of HaematologySuzhou, China. RP Pan, J (reprint author), Children’s Hospital of Soochow University, Institute of Pediatric Research, Suzhou, China. EM panjian2008@163.com CR Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, et al. The 2008 Revision of the World Health Organization, WHO, Classification of Myeloid Neoplasms and Acute Leukemia: Rationale and Important Changes. Blood, 2009, 114(5):937–51., DOI 10.1182/ blood-2009-03-209262 Dohner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid Leukemia. N Engl J Med, 2015, 373(12):1136–52., DOI 10.1056/nejmra1406184 Kantarjian H, Kadia T, DiNardo C, Daver N, Borthakur G, Jabbour E, et al. Acute Myeloid Leukemia: Current Progress and Future Directions. Blood Cancer J, 2021, 11(2):41., DOI 10.1038/s41408-021-00425-3 Kadia TM, Ravandi F, O’Brien S, Cortes J, Kantarjian HM. Progress in Acute Myeloid Leukemia. Clin Lymphoma Myeloma Leuk, 2015, 15(3):139–51., DOI 10.1016/j.clml.2014.08.006 Dohner H, Estey EH, Amadori S, AppelbaumFR, Buchner T, Burnett AK, et al. Diagnosis and Management of Acute Myeloid Leukemia in Adults: Recommendations from an International Expert Panel, on Behalf of the European LeukemiaNet. Blood, 2010, 115(3):453–74., DOI 10.1182/blood- 2009-07-235358 Estey EH. Acute Myeloid Leukemia: 2014 Update on Risk-Stratification and Management. Am J Hematol, 2014, 89(11):1063–81., DOI 10.1002/ajh.23834 Tamamyan G, Kadia T, Ravandi F, Borthakur G, Cortes J, Jabbour E, et al. Frontline Treatment of Acute Myeloid Leukemia in Adults. Crit Rev Oncol Hematol, 2017, 110:20–34., DOI 10.1016/j.critrevonc.2016.12.004 Dovey OM, Cooper JL, Mupo A, Grove CS, Lynn C, Conte N, et al. Molecular Synergy Underlies the Co-occurrence Patterns and Phenotype of NPM1- Mutant Acute Myeloid Leukemia. Blood, 2017, 130(17):1911–22., DOI 10. 1182/blood-2017-01-760595 Ambinder AJ, Levis M. Potential Targeting of FLT3 Acute Myeloid Leukemia. Haematologica, 2021, 106(3):671–81., DOI 10.3324/haematol.2019.240754 Ferret Y, Boissel N, Helevaut N, Madic J, Nibourel O, Marceau-Renaut A, et al. Clinical Relevance of IDH1/2 Mutant Allele burden during Follow-Up in Acute Myeloid Leukemia. A Study by the French ALFA Group. Haematologica, 2018, 103(5):822–9., DOI 10.3324/haematol.2017.183525 Klein K, Kaspers G, Harrison CJ, Beverloo HB, Reedijk A, Bongers M, et al. Clinical Impact of Additional Cytogenetic Aberrations, cKIT and RAS Mutations, and Treatment Elements in Pediatric T(8;21)-AML: Results from an International Retrospective Study by the International Berlin- Frankfurt-Munster Study Group. J Clin Oncol, 2015, 33(36):4247–58., DOI 10.1200/jco.2015.61.1947 Wei Y, Cao Y, Sun R, Cheng L, Xiong X, Jin X, et al. Targeting Bcl-2 Proteins in Acute Myeloid Leukemia. Front Oncol, 2020, 10:584974., DOI 10.3389/fonc. 2020.584974 Wong TN, Ramsingh G, YoungAL, MillerCA,ToumaW, Welch JS, et al. Role of TP53 Mutations in the Origin and Evolution of Therapy-Related Acute Myeloid Leukaemia. Nature, 2015, 518(7540):552–5., DOI 10.1038/nature13968 Larrosa-Garcia M, Baer MR. FLT3 Inhibitors in Acute Myeloid Leukemia: Current Status and Future Directions. Mol Cancer Ther, 2017, 16(6): 991–1001., DOI 10.1158/1535-7163.mct-16-0876 Lam SSY, Leung AYH. Overcoming Resistance to FLT3 Inhibitors in the Treatment of FLT3-Mutated AML. Int J Mol Sci, 2020, 21(4)., DOI 10.3390/ ijms21041537 Dawson MA, Kouzarides T, Huntly BJ. Targeting Epigenetic Readers in Cancer. N Engl J Med, 2012, 367:7. Shi J, Vakoc CR. The Mechanisms behind the Therapeutic Activity of BET Bromodomain Inhibition. Mol Cel, 2014, 54(5):728–36., DOI 10.1016/j.molcel. 2014.05.016 Roe JS, Mercan F, Rivera K, Pappin DJ, Vakoc CR. BET Bromodomain Inhibition Suppresses the Function of Hematopoietic Transcription Factors in Acute Myeloid Leukemia. Mol Cel, 2015, 58(6):1028–39., DOI 10.1016/j. molcel.2015.04.011 Stathis A, Bertoni F. BET Proteins as Targets for Anticancer Treatment. Cancer Discov, 2018, 8(1):24–36., DOI 10.1158/2159-8290.cd-17-0605 Wang N, Wu R, Tang D, Kang R. The BET Family in Immunity and Disease. Sig Transduct Target Ther, 2021, 6(1):23., DOI 10.1038/s41392-020-00384-4 Baratta MG, Schinzel AC, Zwang Y, Bandopadhayay P, Bowman-Colin C, Kutt J, et al. An In-Tumor Genetic Screen Reveals that the BET Bromodomain Protein, BRD4, Is a Potential Therapeutic Target in Ovarian Carcinoma. Proc Natl Acad Sci U.S.A, 2015, 112(1):232–7., DOI 10.1073/pnas.1422165112 Toyoshima M, Howie HL, Imakura M, Walsh RM, Annis JE, Chang AN, et al. Functional Genomics Identifies Therapeutic Targets forMYC-Driven Cancer. Proc Natl Acad Sci U.S.A, 2012, 109(24):9545–50., DOI 10.1073/pnas.1121119109 Zuber J, Shi J, Wang E, Rappaport AR, Herrmann H, Sison EA, et al. RNAi Screen Identifies Brd4 as a Therapeutic Target in Acute Myeloid Leukaemia. Nature, 2011, 478(7370):524–8., DOI 10.1038/nature10334 Marcotte R, Sayad A, Brown KR, Sanchez-Garcia F, Reimand J,Haider M, et al. Functional Genomic Landscape of Human Breast Cancer Drivers, Vulnerabilities, and Resistance. Cell, 2016, 164(1-2):293–309., DOI 10.1016/j. cell.2015.11.062 Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, et al. BET Bromodomain Inhibition as a Therapeutic Strategy to Target C-Myc. Cell, 2011, 146(6):904–17., DOI 10.1016/j.cell.2011.08.017 Sun X, Gao H, Yang Y, He M, Wu Y, Song Y, et al. PROTACs: Great Opportunities for Academia and Industry. Sig Transduct Target Ther, 2019, 4:64., DOI 10.1038/s41392-019-0101-6 Toure M, Crews CM. Small-Molecule PROTACS: New Approaches to Protein Degradation. Angew Chem Int Ed Engl, 2016, 55(6):1966–73., DOI 10.1002/anie. 201507978 Winter GE, Buckley DL, Paulk J, Roberts JM, Souza A, Dhe-Paganon S, et al. Drug Development. Phthalimide Conjugation as a Strategy for In Vivo Target Protein Degradation. Science. Drug Dev, 2015, 348(6241):1376–81., DOI 10. 1126/science.aab1433 Rose D, Haferlach T, Schnittger S, Perglerova K, Kern W, Haferlach C. Subtype-specific Patterns of Molecular Mutations in Acute Myeloid Leukemia. Leukemia, 2017, 31(1):11–7., DOI 10.1038/leu.2016.163 Lim SL, Damnernsawad A, Shyamsunder P, Chng WJ, Han BC, Xu L, et al. Proteolysis Targeting Chimeric Molecules as Therapy for Multiple Myeloma: Efficacy, Biomarker and Drug Combinations. Haematologica, 2019, 104(6): 1209–20., DOI 10.3324/haematol.2018.201483 Xu L, Chen Y, Mayakonda A, Koh L, Chong YK, Buckley DL, et al. Targetable BET Proteins- and E2F1-dependent Transcriptional Program Maintains the Malignancy of Glioblastoma. Proc Natl Acad Sci U S A, 2018, 115(22): E5086–E5095., DOI 10.1073/pnas.1712363115 Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, et al. clusterProfiler 4.0: A Universal Enrichment Tool for Interpreting Omics Data. Innovation, Camb,, 2021, 2(3): 100141., DOI 10.1016/j.xinn.2021.100141 Li Z, Yang C, Li X, Du X, Tao Y, Ren J, et al. The Dual Role of BI 2536, a Small- Molecule Inhibitor that Targets PLK1, in Induction of Apoptosis and Attenuation of Autophagy in Neuroblastoma Cells. J Cancer, 2020, 11(11): 3274–87., DOI 10.7150/jca.33110 Wu S, Jiang Y, Hong Y, Chu X, Zhang Z, Tao Y, et al. BRD4 PROTAC Degrader ARV-825 Inhibits T-Cell Acute Lymphoblastic Leukemia by Targeting ’Undruggable’ Myc-Pathway Genes. Cancer Cel Int, 2021, 21(1): 230., DOI 10.1186/s12935-021-01908-w Boyson SP, Gao C, Quinn K, Boyd J, Paculova H, Frietze S, et al. Functional Roles of Bromodomain Proteins in Cancer. Cancers, Basel,, 2021, 13(14)., DOI 10.3390/cancers13143606 Carter JL, Hege K, Yang J, KalpageHA, Su Y, EdwardsH, et al. TargetingMultiple Signaling Pathways: the New Approach to Acute Myeloid Leukemia Therapy. Signal Transduct Target Ther, 2020, 5(1):288., DOI 10.1038/s41392-020-00361-x Brondfield S, Umesh S, Corella A, Zuber J, Rappaport AR, Gaillard C, et al.Direct and Indirect Targeting of MYC to Treat Acute Myeloid Leukemia. Cancer Chemother Pharmacol, 2015, 76(1):35–46., DOI 10.1007/s00280-015-2766-z Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Buchner T, et al. Diagnosis and Management of AML in Adults: 2017 ELN Recommendations from an International Expert Panel. Blood, 2017, 129(4):424–47., DOI 10.1182/ blood-2016-08-733196 Yu J, Jiang PYZ, Sun H, Zhang X, Jiang Z, Li Y, et al. Advances in Targeted Therapy for Acute Myeloid Leukemia. Biomark Res, 2020, 8:17., DOI 10.1186/ s40364-020-00196-2 Cucchi DGJ, Polak TB, Ossenkoppele GJ, Uyl–De Groot CA, Cloos J, Zweegman S, et al. Two Decades of Targeted Therapies in Acute Myeloid Leukemia. Leukemia, 2021, 35(3):651–60., DOI 10.1038/s41375-021-01164-x Levis M, Perl AE. Gilteritinib: Potent Targeting of FLT3 Mutations in AML. Blood Adv, 2020, 4(6):1178–91., DOI 10.1182/bloodadvances.2019000174 Ahmadi SE, Rahimi S, Zarandi B, Chegeni R, Safa M. MYC: a Multipurpose Oncogene with Prognostic and Therapeutic Implications in BloodMalignancies. J Hematol Oncol, 2021, 14(1):121., DOI 10.1186/s13045-021-01111-4 Kawabata KC, Zong H, Meydan C, Wyman S, Wouters BJ, Sugita M, et al. BCL6 Maintains Survival and Self-Renewal of Primary Human Acute Myeloid Leukemia Cells. Blood, 2021, 137(6):812–25., DOI 10.1182/blood.2019001745 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610447 EP 1610457 DI 10.3389/pore.2022.1610447 PG 11 ER PT J AU Luo, D Liao, S Liu, Y Lin, Y Li, Y Liao, X AF Luo, Dongcheng Liao, Sina Liu, Yu Lin, Youzhi Li, Yongqiang Liao, XiaoLi TI Holliday Cross-Recognition Protein HJURP: Association With the Tumor Microenvironment in Hepatocellular Carcinoma and With Patient Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HJURP; hepatocellular carcinoma; tumor microenvironment; prognosis; GSEA; WGCNA; RNA-seq; single-cell RNA-seq ID HJURP; hepatocellular carcinoma; tumor microenvironment; prognosis; GSEA; WGCNA; RNA-seq; single-cell RNA-seq AB Background: Hepatocellular carcinoma is the most common type of primary liver cancer, and it is associated with poor prognosis. It often fails to respond to immunotherapy, highlighting the need to identify genes that are associated with the tumor microenvironment and may be good therapeutic targets. We and others have shown that the Holliday cross-recognition protein HJURP can promote the proliferation, migration, and invasion by hepatocellular carcinoma cells, and that HJURP overexpression is associated with poor survival. Here we explored the potential relationship between HJURP and the tumor microenvironment in hepatocellular carcinoma. Methods: We used the Immuno-Oncology-Biological-Research (IOBR) software package to analyze the potential roles of HJURP in the tumor microenvironment. Using single-cell RNA sequencing data, we identified the cell clusters expressing abundant HJURP, then linked some of these clusters to certain bioprocesses using Gene Set Enrichment Analysis (GSEA). We validated the differential expression of HJURP in tumor-infiltrating CD8+ T cells, sorted by flow cytometry into populations based on the expression level of PD-1. We used weighted gene co-expression network analysis (WGCNA) to identify immunity-related genes whose expression strongly correlated with that of HJURP. The function of these genes was validated based on enrichment in Gene Ontology (GO) terms, and they were used to establish a prognosis prediction model. Results: IOBR analysis suggested that HJURP is significantly related to the immunosuppressive tumor microenvironment and was significantly related to T cells, dendritic cells, and B cells. Based on single-cell RNA sequencing, HJURP was strongly expressed in T cells, erythrocytes, and B cells from normal liver tissues, as well as in CD8+ T cells, dendritic cells, and one cluster of hepatocytes in hepatocellular carcinoma tissues. Malignant hepatocytes strongly expressing HJURP were associated with the downregulation of immune bioprocesses. HJURP expression was significantly higher in CD8+ T cells strongly expressing PD-1 than in those expressing no or intermediate levels of PD1. WGCNA identified two module eigengenes (comprising 397 and 84 genes) related to the tumor microenvironment. We identified 24 hub genes and confirmed that they were related to immune regulation. A prognostic risk score model based on expression of HJURP, PPT1, PML, and CLEC7A showed moderate ability to predict survival. Conclusion: HJURP is associated with tumor-infiltrating immune cells, immune checkpoints, and immune suppression in hepatocellular carcinoma. HJURP-related genes involved in immune responses may be useful for predicting patient prognosis. C1 [Luo, Dongcheng] Guangxi Medical University Cancer Hospital, Department of First ChemotherapyNanning, China. [Liao, Sina] Guangxi Medical University Cancer Hospital, Department of First ChemotherapyNanning, China. [Liu, Yu] Guangxi Medical University Cancer Hospital, Department of First ChemotherapyNanning, China. [Lin, Youzhi] Guangxi Medical University Cancer Hospital, Hepatobiliary Surgery DepartmentNanning, China. [Li, Yongqiang] Guangxi Medical University Cancer Hospital, Department of First ChemotherapyNanning, China. [Liao, XiaoLi] Guangxi Medical University Cancer Hospital, Department of First ChemotherapyNanning, China. RP Liao, X (reprint author), Guangxi Medical University Cancer Hospital, Department of First Chemotherapy, Nanning, China. EM nllxl@163.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Gordan JD, Kennedy EB, Abou-Alfa GK, Beg MS, Brower ST, Gade TP, et al. Systemic Therapy for Advanced Hepatocellular Carcinoma: ASCO Guideline. J Clin Oncol, 2020, 38(36):4317–45., DOI 10.1200/jco.20.02672 Golabi P, Fazel S, Otgonsuren M, Sayiner M, Locklear CT, Younossi ZM. Mortality Assessment of Patients with Hepatocellular Carcinoma According to Underlying Disease and Treatment Modalities. Medicine, Baltimore,, 2017, 96(9):e5904., DOI 10.1097/md.0000000000005904 El-Serag HB. Hepatocellular Carcinoma. N Engl J Med, 2011, 365(12): 1118–27., DOI 10.1056/nejmra1001683 Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D, et al. Pembrolizumab in Patients with Advanced Hepatocellular Carcinoma Previously Treated with Sorafenib, KEYNOTE-224): A Non-randomised, Open-Label Phase 2 Trial. Lancet Oncol, 2018, 19(7):940–52., DOI 10.1016/ S1470-2045(18)30351-6 Giannone G, Ghisoni E, Genta S, Scotto G, Tuninetti V, Turinetto M, et al. Immuno-Metabolism and Microenvironment in Cancer: Key Players for Immunotherapy. Int J Mol Sci, 2020, 21(12):4414., DOI 10.3390/ ijms21124414 Fu Y, Liu S, Zeng S, Shen H. From Bench to Bed: The Tumor Immune Microenvironment and Current Immunotherapeutic Strategies for Hepatocellular Carcinoma. J Exp Clin Cancer Res, 2019, 38(1):396., DOI 10. 1186/s13046-019-1396-4 Dunleavy EM, Roche D, Tagami H, Lacoste N, Ray-Gallet D, Nakamura Y, et al. HJURP is a Cell-cycle-dependent Maintenance and Deposition Factor of CENP-A at Centromeres. Cell, 2009, 137(3):485–97., DOI 10.1016/j.cell.2009. 02.040 Shuaib M, Ouararhni K, Dimitrov S, Hamiche A. HJURP Binds CENP-A via a Highly Conserved N-Terminal Domain and Mediates its Deposition at Centromeres. Proc Natl Acad Sci U.S.A, 2010, 107(4):1349–54., DOI 10.1073/ pnas.0913709107 Zasadzinska E, Huang J, Bailey AO, Guo LY, Lee NS, Srivastava S, et al. Inheritance of CENP-A Nucleosomes during DNA Replication Requires HJURP. Dev Cel, 2018, 47(3):348–e7., DOI 10.1016/j.devcel.2018.09.003 Lai W, Zhu W, Xiao C, Li X, Wang Y, Han Y, et al. HJURP Promotes Proliferation in Prostate Cancer Cells through Increasing CDKN1A Degradation via the GSK3β/JNK Signaling Pathway. Cell Death Dis, 2021, 12(6):583., DOI 10.1038/s41419-021-03870-x Wei Y, Ouyang GL, Yao WX, Zhu YJ, Li X, Huang LX, et al. Knockdown of HJURP Inhibits Non-small Cell Lung Cancer Cell Proliferation, Migration, and Invasion by Repressing Wnt/β-Catenin Signaling. Eur Rev Med Pharmacol Sci, 2019, 23(9):3847–56., DOI 10.26355/eurrev_201905_17812 Chen T, Huang H, Zhou Y, Geng L, Shen T, Yin S, et al. HJURP Promotes Hepatocellular Carcinoma Proliferation by Destabilizing P21 via the MAPK/ERK1/2 and AKT/GSK3β Signaling Pathways. J Exp Clin Cancer Res, 2018, 37(1):193., DOI 10.1186/s13046-018-0866-4 Wang C-j., Li X, Shi P, Ding H-y., Liu Y-p., Li T, et al. Holliday junction Recognition Protein Promotes Pancreatic Cancer Growth and Metastasis via Modulation of the MDM2/p53 Signaling. Cel Death Dis, 2020, 11(5):386., DOI 10.1038/s41419-020-2595-9 Kang DH, Woo J, Kim H, Kim SY, Ji S, Jaygal G, et al. Prognostic Relevance of HJURP Expression in Patients with Surgically Resected Colorectal Cancer. Int J Mol Sci, 2020, 21(21):7928., DOI 10.3390/ijms21217928 Hu Z, Huang G, Sadanandam A, Gu S, Lenburg ME, Pai M, et al. The Expression Level of HJURP Has an Independent Prognostic Impact and Predicts the Sensitivity to Radiotherapy in Breast Cancer. Breast Cancer Res, 2010, 12(2):R18., DOI 10.1186/bcr2487 Li Y, Yi Q, Liao X, Han C, Zheng L, Li H, et al. Hypomethylation-driven Overexpression of HJURP Promotes Progression of Hepatocellular Carcinoma and Is Associated with Poor Prognosis. Biochem Biophys. Res Commun, 2021, 566:67–74., DOI 10.1016/j.bbrc.2021.05.102 Shixiang W, Liu XS. The UCSCXena Tools R Package: A Toolkit for Accessing Genomics Data from UCSC Xena Platform, from Cancer Multi-Omics to Single-Cell RNA-Seq. J Open Source Softw, 2019, 4(40):1627., DOI 10.21105/ joss.01627 Kim H-D, Song G-W, Park S, Jung MK, Kim MH, Kang HJ, et al. Association between Expression Level of PD1 by Tumor-Infiltrating CD8+ T Cells and Features of Hepatocellular Carcinoma. Gastroenterology, 2018, 155(6): 1936–50., DOI 10.1053/j.gastro.2018.08.030 Therneau T. A Package for Survival Analysis in R. R package version 3.2-13, 2021). Available at: https://CRAN.R-project.org/package=survival. Zeng D, Ye Z, Shen R, Yu G, Wu J, Xiong Y, et al. IOBR: Multi-Omics Immuno-Oncology Biological Research to Decode Tumor Microenvironment and Signatures. Front Immunol, 2021, 12:687975., DOI 10.3389/fimmu.2021. 687975 Sharma A, Seow JJW, Dutertre C-A, Pai R, Bleriot C, Mishra A, et al. Oncofetal Reprogramming of Endothelial Cells Drives Immunosuppressive Macrophages in Hepatocellular Carcinoma. Cell, 2020, 183(2):377–94., DOI 10.1016/j.cell.2020.08.040 Hao Y, Hao S, Andersen-Nissen E, Mauck WM, Zheng S, Butler A, et al. Integrated Analysis of Multimodal Single-Cell Data. Cell, 2021, 184:3573., DOI 10.1016/j.cell.2021.04.048 Langfelder P, Horvath S. WGCNA: an R Package for Weighted Correlation Network Analysis. BMC Bioinformatics, 2008, 9:559., DOI 10.1186/1471-2105- 9-559 Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: A Software Environment for Integrated Models of Biomolecular Interaction Networks. Genome Res, 2003, 13(11):2498–504., DOI 10.1101/gr. 1239303 Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, et al. clusterProfiler 4.0: A Universal Enrichment Tool for Interpreting Omics Data. The Innovation, 2021, 2(3): 100141., DOI 10.1016/j.xinn.2021.100141 Yu G. Enrichplot: Visualization of Functional Enrichment Result. R Package Version 1.12.3, 2021). Available at: https://yulab-smu.top/biomedicalknowledge- mining-book/. Foltz DR, Jansen LET, Bailey AO, Yates JR, Bassett EA, Wood S, et al. Centromere-specific Assembly of CENP-A Nucleosomes is Mediated by HJURP. Cell, 2009, 137(3):472–84., DOI 10.1016/j.cell.2009.02.039 Hu B, Wang Q, Wang Y, Chen J, Li P, Han M. Holliday junction-recognizing Protein Promotes Cell Proliferation and Correlates with Unfavorable Clinical Outcome of Hepatocellular Carcinoma. Onco Targets Ther, 2017, 10:2601–7., DOI 10.2147/ott.s127738 Huang W, Zhang H, Hao Y, Xu X, Zhai Y,Wang S, et al. A Non-synonymous Single Nucleotide Polymorphism in the HJURP Gene Associated with Susceptibility to Hepatocellular Carcinoma Among Chinese. PLoS One, 2016, 11(2):e0148618., DOI 10.1371/journal.pone.0148618 Luo C, Chen J, Chen L. Exploration of Gene Expression Profiles and Immune Microenvironment between High and Low Tumor Mutation burden Groups in Prostate Cancer. Int Immunopharmacol, 2020, 86:106709., DOI 10.1016/j. intimp.2020.106709 Zhang F, Yuan D, Song J, Chen W, Wang W, Zhu G, et al. HJURP is a Prognostic Biomarker for clear Cell Renal Cell Carcinoma and is Linked to Immune Infiltration. Int Immunopharmacol, 2021, 99:107899., DOI 10.1016/j. intimp.2021.107899 Peng Q, Qiu X, Zhang Z, Zhang S, Zhang Y, Liang Y, et al. PD-L1 on Dendritic Cells Attenuates T Cell Activation and Regulates Response to Immune Checkpoint Blockade. Nat Commun, 2020, 11(1):4835., DOI 10.1038/s41467- 020-18570-x Thommen DS, Schumacher TN. T Cell Dysfunction in Cancer. Cancer Cell, 2018, 33(4):547–62., DOI 10.1016/j.ccell.2018.03.012 Heo J-I, Cho JH, Kim J-R. HJURP Regulates Cellular Senescence in Human Fibroblasts and Endothelial Cells via a P53-dependent Pathway. J Gerontol Ser A Biol Sci Med Sci, 2013, 68(8):914–25., DOI 10.1093/gerona/gls257 Mao M, Jia Y, Chen Y, Yang J, Xu L, Zhang X, et al. HJURP Regulates Cell Proliferation and Chemo-Resistance via YAP1/NDRG1 Transcriptional axis in Triple-Negative Breast Cancer. Cel Death Dis, 2022, 13(4):396., DOI 10.1038/ s41419-022-04833-6 Chen T, Zhou L, Zhou Y, Zhou W, Huang H, Yin S, et al. HJURP Promotes Epithelial-To-Mesenchymal Transition via Upregulating SPHK1 in Hepatocellular Carcinoma. Int J Biol Sci, 2019, 15(6):1139–47., DOI 10.7150/ ijbs.30904 Chakraborty P, Vaena SG, Thyagarajan K, Chatterjee S, Al-Khami A, Selvam SP, et al. Pro-Survival Lipid Sphingosine-1-Phosphate Metabolically Programs T Cells to Limit Anti-tumor Activity. Cel Rep, 2019, 28(7):1879–93., DOI 10. 1016/j.celrep.2019.07.044 Gupta P, Kadamberi IP, Mittal S, Tsaih SW, George J, Kumar S, et al. Tumor Derived Extracellular Vesicles Drive T Cell Exhaustion in Tumor Microenvironment through Sphingosine Mediated Signaling and Impacting Immunotherapy Outcomes in Ovarian Cancer. Adv Sci, Weinh,, 2022, 9(14): e2104452., DOI 10.1002/advs.202104452 Takeuchi Y, Tanegashima T, Sato E, Irie T, Sai A, Itahashi K, et al. Highly Immunogenic Cancer Cells Require Activation of the WNT Pathway for Immunological Escape. Sci Immunol, 2021, 6(65):eabc6424., DOI 10.1126/ sciimmunol.abc6424 Fang DD, Tang Q, Kong Y, Wang Q, Gu J, Fang X, et al. MDM2 Inhibitor APG-115 Synergizes with PD-1 Blockade through Enhancing Antitumor Immunity in the Tumor Microenvironment. J Immunother Cancer, 2019, 7(1):327., DOI 10.1186/s40425-019-0750-6 Shibata M, Ham K, Hoque MO. A Time for YAP1: Tumorigenesis, Immunosuppression and Targeted Therapy. Int J Cancer, 2018, 143(9): 2133–44., DOI 10.1002/ijc.31561 Rebecca VW, Nicastri MC, Fennelly C, Chude CI, Barber-Rotenberg JS, Ronghe A, et al. PPT1 Promotes Tumor Growth and is the Molecular Target of Chloroquine Derivatives in Cancer. Cancer Discov, 2019, 9(2): 220–9., DOI 10.1158/2159-8290.cd-18-0706 Xu J, Su Z, Cheng X, Hu S, Wang W, Zou T, et al. High PPT1 Expression Predicts Poor Clinical Outcome and PPT1 Inhibitor DC661 Enhances Sorafenib Sensitivity in Hepatocellular Carcinoma. Cancer Cel Int, 2022, 22(1):115., DOI 10.1186/s12935-022-02508-y Sharma G, Ojha R, Noguera-Ortega E, Rebecca VW, Attanasio J, Liu S, et al. PPT1 Inhibition Enhances the Antitumor Activity of Anti-PD-1 Antibody in Melanoma. JCI Insight, 2020, 5(17):e133225., DOI 10.1172/jci.insight.133225 Maarifi G, Chelbi-Alix MK, Nisole S. PML Control of Cytokine Signaling. Cytokine Growth Factor Rev, 2014, 25(5):551–61., DOI 10.1016/j.cytogfr.2014. 04.008 Daley D, Mani VR, Mohan N, Akkad N, Ochi A, Heindel DW, et al. Dectin 1 Activation on Macrophages by Galectin 9 Promotes Pancreatic Carcinoma and Peritumoral Immune Tolerance. Nat Med, 2017, 23(5):556–67., DOI 10.1038/ nm.4314 Shiao SL, Kershaw KM, Limon JJ, You S, Yoon J, Ko EY, et al. Commensal Bacteria and Fungi Differentially Regulate Tumor Responses to Radiation Therapy. Cancer Cell, 2021, 39(9):1202–13., DOI 10.1016/j.ccell.2021.07.002 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610506 EP 1610524 DI 10.3389/pore.2022.1610506 PG 19 ER PT J AU Deme, D Kovacs, S Telekes, A AF Deme, Daniel Kovacs, Sandor Telekes, Andras TI Corrigendum: Overall Survival Prediction of Advanced Cancer Patients by Selection of the Most Significant Baseline Serum Biomarker Combination SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE overall survival; advanced cancer; serum biomarkers; prognostic importance; CRP; albumin; PLR ID overall survival; advanced cancer; serum biomarkers; prognostic importance; CRP; albumin; PLR AB A Corrigendum on Overall Survival Prediction of Advanced Cancer Patients by Selection of the Most Significant Baseline Serum Biomarker Combination by Deme D, Kovacs S and Telekes A (2022). Pathol. Oncol. Res. 28:1610004. doi: 10.3389/pore.2022.1610004 C1 [Deme, Daniel] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. [Kovacs, Sandor] University of Debrecen, Department of Economical and Financial MathematicsDebrecen, Hungary. [Telekes, Andras] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. RP Deme, D (reprint author), Szent Lazar Megyei Korhaz, Onkologiai Osztaly, Salgotarjan, Hungary. EM danieldeme_md@ymail.com NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUN PY 2022 VL 28 IS 2 BP 1610517 EP 1610518 DI 10.3389/pore.2022.1610517 PG 2 ER PT J AU Chen, W Zhang, F Xu, H Hou, X Tang, D Dai, Y AF Chen, Wenbiao Zhang, Feng Xu, Huixuan Hou, Xianliang Tang, Donge Dai, Yong TI Identification and Characterization of Genes Related to the Prognosis of Hepatocellular Carcinoma Based on Single-Cell Sequencing SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gene; hepatocellular carcinoma; prognostic model; single-cell sequencing; molecular cluster ID gene; hepatocellular carcinoma; prognostic model; single-cell sequencing; molecular cluster AB The heterogeneity of hepatocellular carcinoma (HCC) highlights the importance of precision therapy. In recent years, single-cell RNA sequencing has been used to reveal the expression of genes at the single-cell level and comprehensively study cell heterogeneity. This study combined big data analytics and single-cell data mining to study the influence of genes on HCC prognosis. The cells and genes closely related to the HCC were screened through single-cell RNA sequencing (71,915 cells, including 34,414 tumor cells) and big data analysis. Comprehensive bioinformatics analysis of the key genes of HCC was conducted formolecular classification and multi-dimensional correlation analyses, and a prognostic model for HCC was established. Finally, the correlation between the prognosticmodel and clinicopathological features was analyzed. 16,880 specific cells, screened from the single-cell expression profile matrix, were divided into 20 sub-clusters. Cell typing revealed that 97% of these cells corresponded to HCC cell lines, demonstrating the high specificity of cells derived from single-cell sequencing. 2,038 genes with high variability were obtained. The 371 HCC samples were divided into two molecular clusters. Cluster 1 (C1) was associated with tumorigenesis, high immune score, immunotherapy targets (PD-L1 and CYLA-4), high pathological stage, and poor prognosis. Cluster 2 (C2) was related to metabolic and immune function, low immune score, low pathological stage, and good prognosis. Seven differentially expressed genes (CYP3A4, NR1I2, CYP2C9, TTR, APOC3, CYP1A2, and AFP) identified between the two molecular clusters were used to construct a prognosticmodel.We further validated the correlation between the seven key genes and clinical features, and the established prognostic model could effectively predict HCC prognosis. Our study identified seven key genes related to HCC that were used to construct a prognostic model through single-cell sequencing and big data analytics. This study provides new insights for further research on clinical targets of HCC and new biomarkers for clinical application. C1 [Chen, Wenbiao] Chinese Academy of Sciences, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Research Center for Human Tissue and Organs DegenerationShenzhen, China. [Zhang, Feng] The First Affiliated Hospital of Jinan University, Intensive Care UnitGuangzhou, China. [Xu, Huixuan] Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen, China. [Hou, Xianliang] Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen, China. [Tang, Donge] Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen, China. [Dai, Yong] Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen, China. RP Chen, W (reprint author), Chinese Academy of Sciences, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Research Center for Human Tissue and Organs Degeneration, Shenzhen, China. CR Kulik L, El-Serag HB. Epidemiology and Management of Hepatocellular Carcinoma. Gastroenterology, 2019, 156(2):477–91., DOI 10.1053/j.gastro. 2018.08.065 Hartke J, Johnson M, Ghabril M. The Diagnosis and Treatment of Hepatocellular Carcinoma. Semin Diagn Pathol, 2017, 34(2):153–9., DOI 10. 1053/j.semdp.2016.12.011 Lurje I, Czigany Z, Bednarsch J, Roderburg C, Isfort P, Neumann UP, et al. Treatment Strategies for Hepatocellular Carcinoma ⁻ a Multidisciplinary Approach. Int J Mol Sci, 2019, 20(6):E1465., DOI 10.3390/ijms20061465 Li L, Wang H. Heterogeneity of Liver Cancer and Personalized Therapy. Cancer Lett, 2016, 379(2):191–7., DOI 10.1016/j.canlet.2015.07.018 Prasetyanti PR, Medema JP. Intra-Tumor Heterogeneity from a Cancer Stem Cell Perspective. Mol Cancer, 2017, 16(1):41., DOI 10.1186/s12943-017-0600-4 Zhang Q, Lou Y, Yang J, Wang J, Feng J, Zhao Y, et al. Integrated Multiomic Analysis Reveals Comprehensive Tumour Heterogeneity and Novel Immunophenotypic Classification in Hepatocellular Carcinomas. Gut, 2019, 68(11):2019–31., DOI 10.1136/gutjnl-2019-318912 Hong M, Tao S, Zhang L, Diao LT, Huang X, Huang S, et al. RNA Sequencing: New Technologies and Applications in Cancer Research. J Hematol Oncol, 2020, 13(1):166., DOI 10.1186/s13045-020-01005-x Hwang B, Lee JH, Bang D. Single-cell RNA Sequencing Technologies and Bioinformatics Pipelines. Exp Mol Med, 2018, 50(8):96., DOI 10.1038/s12276- 018-0071-8 Kumar MP, Du J, Lagoudas G, Jiao Y, Sawyer A, Drummond DC, et al. Analysis of Single-Cell RNA-Seq Identifies Cell-Cell Communication Associated with Tumor Characteristics. Cell Rep, 2018, 25(6):1458–68. e4., DOI 10.1016/j.celrep.2018.10.047 Huang X, Liu S, Wu L, Jiang M, Hou Y. High Throughput Single Cell RNA Sequencing, Bioinformatics Analysis and Applications. Adv Exp Med Biol, 2018, 1068:33–43., DOI 10.1007/978-981-13-0502-3_4 Zhou S, Yung Chan S, Cher Goh B, Chan E, Duan W, Huang M, et al. Mechanism-based Inhibition of Cytochrome P450 3A4 by Therapeutic Drugs. Clin Pharmacokinet, 2005, 44(3):279–304., DOI 10.2165/00003088-200544030- 00005 Ma X, Chen J, Tian Y. Pregnane X Receptor as the "Sensor and Effector" in Regulating Epigenome. J Cell Physiol, 2015, 230(4):752–7., DOI 10.1002/jcp. 24838 Nalivaeva NN, Belyaev ND, Kerridge C, Turner AJ. Amyloid-clearing Proteins and Their Epigenetic Regulation as a Therapeutic Target in Alzheimer’s Disease. Front Aging Neurosci, 2014, 6:235., DOI 10.3389/ fnagi.2014.00235 Kozlitina J, Boerwinkle E, Cohen JC, Hobbs HH. Dissociation between APOC3 Variants, Hepatic Triglyceride Content and Insulin Resistance. Hepatology, Baltimore, Md,, 2011, 53(2):467–74., DOI 10.1002/hep.24072 Chen Z, Zou Y, Zhang Y, Chen Z, Wu F, Jin H, et al. A Pyroptosis-Based Prognostic Model for Immune Microenvironment Estimation of Hepatocellular Carcinoma. Dis Markers, 2022, 2022:8109771., DOI 10.1155/ 2022/8109771 Yu H, Bai X, Zheng W. Identification of the Pyroptosis-Related Prognosis Gene Signature and Immune Infiltration in Hepatocellular Carcinoma. Dis Markers, 2022, 2022:9124216., DOI 10.1155/2022/9124216 Chen R, Zhao M, An Y, Liu D, Tang Q, Teng G, et al. A Prognostic Gene Signature for Hepatocellular Carcinoma. Front Oncol, 2022, 12:841530., DOI 10.3389/fonc.2022.841530 Wang G, Ding B, Sun L, Guo J, Wang S, Li W, et al. Construction and Validation of a Necroptosis-Related Signature Associated with the Immune Microenvironment in Liver Hepatocellular Carcinoma. Front Genet, 2022, 13: 859544., DOI 10.3389/fgene.2022.859544 Whitfield ML, George LK, Grant GD, Perou CM. Common Markers of Proliferation. Nat Rev Cancer, 2006, 6(2):99–106., DOI 10.1038/nrc1802 Lockwood DS, Yeadon TM, Clouston AD, Crawford DG, Fawcett J, Callaghan SA, et al. Tumor Progression in Hepatocellular Carcinoma: Relationship with Tumor Stroma and Parenchymal Disease. J Gastroenterol Hepatol, 2003, 18(6):666–72., DOI 10.1046/j.1440-1746.2003.03018.x Chen W, Tang D, Ou M, Dai Y. Mining Prognostic Biomarkers of Hepatocellular Carcinoma Based on Immune-Associated Genes. DNA Cell Biol, 2020, 39(4):499–512., DOI 10.1089/dna.2019.5099 Chen W, Zhang X, Bi K, Zhou H, Xu J, Dai Y, et al. Comprehensive Study of Tumor Immune Microenvironment and Relevant Genes in Hepatocellular Carcinoma Identifies Potential Prognostic Significance. Front Oncol, 2020, 10: 554165., DOI 10.3389/fonc.2020.554165 Zhang Z, Wang Z, Huang Y. Comprehensive Analyses of the Infiltrating Immune Cell Landscape and its Clinical Significance in Hepatocellular Carcinoma. Int J Gen Med, 2021, 14:4695–704., DOI 10.2147/IJGM.S326535 Gajewski TF, Schreiber H, Fu YX. Innate andAdaptive Immune Cells in the Tumor Microenvironment. Nat Immunol, 2013, 14(10):1014–22., DOI 10.1038/ni.2703 Borst J, Ahrends T, Babala N, Melief CJM, Kastenmuller W. CD4(+, T Cell Help in Cancer Immunology and Immunotherapy. Nat Rev Immunol, 2018, 18(10):635–47., DOI 10.1038/s41577-018-0044-0 Xie G, Dong H, Liang Y, Ham JD, Rizwan R, Chen J, et al. CAR-NK Cells: A Promising Cellular Immunotherapy for Cancer. EBioMedicine, 2020, 59: 102975., DOI 10.1016/j.ebiom.2020.102975 Rotte A. Combination of CTLA-4 and PD-1 Blockers for Treatment of Cancer. J Exp Clin Cancer Res, 2019, 38(1):255., DOI 10.1186/s13046-019- 1259-z Luo P, Wu S, Yu Y, Ming X, Li S, Zuo X, et al. Current Status and Perspective Biomarkers in AFP Negative HCC: Towards Screening for and Diagnosing Hepatocellular Carcinoma at an Earlier Stage. Pathol Oncol Res, 2020, 26(2): 599–603., DOI 10.1007/s12253-019-00585-5 Wang W, Wei C. Advances in the Early Diagnosis of Hepatocellular Carcinoma. Genes Dis, 2020, 7(3):308–19., DOI 10.1016/j.gendis.2020.01.014 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610199 EP 1610212 DI 10.3389/pore.2022.1610199 PG 14 ER PT J AU Amiri, M Komi, EAD Vaisi-Raygani, A Kiani, A Moradi, M Aliyari, M Rahimi, Z Mohammadi-Noori, E Bashiri, H AF Amiri, Mohammad Komi, Elieh Ali Daniel Vaisi-Raygani, Asad Kiani, Amir Moradi, Mahmoudreza Aliyari, Mahdieh Rahimi, Zohreh Mohammadi-Noori, Ehsan Bashiri, Homayoon TI Association Between Vitamin D Binding Protein Gene Polymorphism (rs7041), Vitamin D Receptor, and 25-Hydroxyvitamin D Serum Levels With Prostate Cancer in Kurdish Population in West of Iran SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prostate cancer; polymorphism; vitamin D receptor; vitamin D binding protein; 25(OH)-vitamin D ID prostate cancer; polymorphism; vitamin D receptor; vitamin D binding protein; 25(OH)-vitamin D AB Prostate cancer (PCa) pathology has been linked to vitamin D, vitamin D receptors (VDRs), and vitamin D binding proteins (VDBPs). We sought to investigate the association between VDR rs2228570 and rs1544410 as well as VDBP rs7041 polymorphisms and serum 25- hydroxyvitamin D (25(OH)-vitamin D) levels in PCa patients. Blood samples were collected from 111 PCa patients and 150 age-matched healthy volunteers. The VDR rs2228570 T/ C, rs1544410 G/A, and VDBP rs7041 T/G genotypes were determined using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). 25(OH)-vitamin D and PSA (Total and Free) serum levels were measured. The frequencies of VDBP genotypes T/G vs. T/T (56.5% vs. 44.5%, p = 0.01) according to the dominant model T/G + G/G vs. T/T (84.3% vs. 71.5%, p = 0.01) were significantly higher in PCa patients when compared to control group and considerably increased the risk of disease by 2.29, 1.44, and 2.13 folds respectively. Interestingly, the results demonstrated that PCa patients with the dominant model (T/G + G/G vs. T/T) of VDBP had significantly lower serum levels of vitamin D and higher serum levels of total and free PSA in comparison to the controls. Furthermore, when compared to controls, PCa patients with the dominant model T allele (T/G + G/G vs. TT) of VDBP had significantly higher vitamin D, total PSA, and free PSA concentrations. Serum levels of 25(OH)-vitamin D and rs7041 T/G polymorphism of the VDBP gene could be potential risk factors for PCa. C1 [Amiri, Mohammad] Kermanshah University of Medical Sciences, Student Research CommitteeKermanshah, Iran. [Komi, Elieh Ali Daniel] Kermanshah University of Medical Sciences, Regenerative Medicine Research Center (RMRC)Kermanshah, Iran. [Vaisi-Raygani, Asad] Kermanshah University of Medical Sciences, Fertility and Infertility Research CenterKermanshah, Iran. [Kiani, Amir] Kermanshah University of Medical Sciences, Regenerative Medicine Research Center (RMRC)Kermanshah, Iran. [Moradi, Mahmoudreza] Kermanshah University of Medical Sciences, Regenerative Medicine Research Center (RMRC)Kermanshah, Iran. [Aliyari, Mahdieh] Kermanshah University of Medical Sciences, Student Research CommitteeKermanshah, Iran. [Rahimi, Zohreh] Kermanshah University of Medical Sciences, Medical Biology Research CenterKermanshah, Iran. [Mohammadi-Noori, Ehsan] Kermanshah University of Medical Sciences, Health Institute, Pharmaceutical Sciences Research CenterKermanshah, Iran. [Bashiri, Homayoon] Kermanshah University of Medical Sciences, Department of Internal MedicineKermanshah, Iran. RP Bashiri, H (reprint author), Kermanshah University of Medical Sciences, Department of Internal Medicine, Kermanshah, Iran. EM h.bashiri@kums.ac.ir CR Aliyari M, Elieh Ali Komi D, Kiani A, Moradi M, Tanhapour M, Rahimi Z, et al. The Role of Caveolin-1 and Endothelial Nitric Oxide Synthase Polymorphisms in Susceptibility to Prostate Cancer. Int J Exp Path, 2021, 102:260–7., DOI 10.1111/iep.12394 Pernar CH, Ebot EM, Wilson KM, Mucci LA. The Epidemiology of Prostate Cancer. Cold Spring Harb Perspect Med, 2018, 8(12):a030361., DOI 10.1101/ cshperspect.a030361 Kimura T, Egawa S. Epidemiology of Prostate Cancer in Asian Countries. Int J Urol, 2018, 25(6):524–31., DOI 10.1111/iju.13593 Bouillon R, Schuit F, Antonio L, Rastinejad F. Vitamin D Binding Protein: A Historic Overview. Front Endocrinol, 2019, 10:910., DOI 10.3389/fendo.2019. 00910 Zacharioudaki M, Messaritakis I, Galanakis E. Vitamin D Receptor, Vitamin D Binding Protein and CYP27B1 Single Nucleotide Polymorphisms and Susceptibility to Viral Infections in Infants. Sci Rep, 2021, 11(1):13835., DOI 10.1038/s41598-021-93243-3 Speeckaert MM, Speeckaert R, van Geel N, Delanghe JR. Vitamin D Binding Protein. Adv Clin Chem, 2014, 63:1–57., DOI 10.1016/b978-0-12-800094-6. 00001-7 Yang L, Chen H, Zhao M, Peng P. Prognostic Value of Circulating Vitamin D Binding Protein, Total, Free and Bioavailable 25-hydroxy Vitamin D in Patients with Colorectal Cancer. Oncotarget, 2017, 8(25):40214–21., DOI 10. 18632/oncotarget.16597 Rozmus D, Ciesielska A, Plominski J, Grzybowski R, Fiedorowicz E, Kordulewska N, et al. Vitamin D Binding Protein, VDBP, and its Gene Polymorphisms-The Risk of Malignant Tumors and Other Diseases. Int J Mol Sci, 2020, 21(21):7822., DOI 10.3390/ijms21217822 Yuan C, Song M, Zhang Y, Wolpin BM, Meyerhardt JA, Ogino S, et al. Prediagnostic Circulating Concentrations of Vitamin D Binding Protein and Survival Among Patients with Colorectal Cancer. Cancer Epidemiol Biomarkers Prev, 2020, 29(11):2323–31., DOI 10.1158/1055-9965.Epi-20-0291 Samuel S, Sitrin MD. Vitamin D’s Role in Cell Proliferation and Differentiation. Nutr Rev, 2008, 66(10 Suppl. 2):S116–S124., DOI 10.1111/j. 1753-4887.2008.00094.x Haussler MR, Whitfield GK, Haussler CA, Hsieh J-C, Thompson PD, Selznick SH, et al. The Nuclear Vitamin D Receptor: Biological and Molecular Regulatory Properties Revealed. J Bone Miner Res, 1998, 13(3):325–49., DOI 10.1359/jbmr.1998.13.3.325 Krill D, DeFlavia P, Dhir R, Luo J, Becich MJ, Lehman E, et al. Expression Patterns of Vitamin D Receptor in Human Prostate. J Cel Biochem., 2001, 82(4):566–72., DOI 10.1002/jcb.1185 Miyamoto KI, Kesterson RA, Yamamoto H, Taketani Y, Nishiwaki E, Tatsumi S, et al. Structural Organization of the Human Vitamin D Receptor Chromosomal Gene and its Promoter. Mol Endocrinol, 1997, 11(8): 1165–79., DOI 10.1210/mend.11.8.9951 Li L, Shang F, ZhangW, Zhang C, Li J, Wang C, et al. Role of Vitamin D Receptor Gene Polymorphisms in Pancreatic Cancer: A Case-Control Study in China. Tumor Biol, 2015, 36(6):4707–14., DOI 10.1007/s13277- -3119-6 15. Bai Y, Yu Y, Yu B, Ge J, Ji J, Lu H, et al. Association of Vitamin D Receptor Polymorphisms with the Risk of Prostate Cancer in the Han Population of Southern China. BMC Med Genet, 2009, 10:125., DOI 10.1186/1471-2350- 10-125 Maciejewski A, Kowalczyk MJ, Herman W, Czyzyk A, Kowalska M, Zaba R, et al. Vitamin D Receptor Gene Polymorphisms and Autoimmune Thyroiditis: Are They Associated with Disease Occurrence and its Features? Biomed Res Int, 2019, 2019:1–9., DOI 10.1155/2019/8197580 Hashemi SM, Arbabi N, Hashemi M, Mashhadi MA, Allahyari A, Sadeghi M. Association between VDR Gene Polymorphisms, Rs 1544410, Rs 7975232, Rs 2228570, Rs 731236 and Rs 11568820, and Susceptibility to Breast Cancer in a Sample of Southeastern Iranian Population. Int J Cancer Manag, 2017, 10(3): e8807., DOI 10.5812/ijcm.8807 Lurie G, Wilkens LR, Thompson PJ, Carney ME, Palmieri RT, Pharoah PDP, et al. Vitamin D Receptor Rs2228570 Polymorphism and Invasive Ovarian Carcinoma Risk: Pooled Analysis in Five Studies within the Ovarian Cancer Association Consortium. Int J Cancer, 2011, 128(4):936–43., DOI 10.1002/ijc. 25403 Verstuyf A, Carmeliet G, Bouillon R, Mathieu C. Vitamin D: A Pleiotropic Hormone. Kidney Int, 2010, 78(2):140–5., DOI 10.1038/ki.2010.17 Deeb KK, Trump DL, Johnson CS. Vitamin D Signalling Pathways in Cancer: Potential for Anticancer Therapeutics. Nat Rev Cancer, 2007, 7(9):684–700., DOI 10.1038/nrc2196 Welsh J. Targets of Vitamin D Receptor Signaling in the Mammary Gland. J Bone Miner Res, 2007, 22(Suppl. 2):V86–V90., DOI 10.1359/jbmr.07s204 Stewart LV, Weigel NL. Vitamin D and Prostate Cancer. Exp Biol Med, Maywood,, 2004, 229(4):277–84., DOI 10.1177/153537020422900401 Hendrickson WK, Flavin R, Kasperzyk JL, Fiorentino M, Fang F, Lis R, et al. Vitamin D Receptor Protein Expression in Tumor Tissue and Prostate Cancer Progression. J Clin Oncol, 2011, 29(17):2378–85., DOI 10.1200/jco.2010.30.9880 Kim H, Giovannucci E. Vitamin D Status and Cancer Incidence, Survival, and Mortality. Adv Exp Med Biol, 2020, 1268:39–52., DOI 10.1007/978-3-030- 46227-7_3 Beer TM, Myrthue A. Calcitriol in the Treatment of Prostate Cancer. Anticancer Res, 2006, 26(4a):2647–51. Peehl DM, Skowronski RJ, Leung GK, Wong ST, Stamey TA, Feldman D. Antiproliferative Effects of 1,25-dihydroxyvitamin D3 on Primary Cultures of Human Prostatic Cells. Cancer Res, 1994, 54(3):805–10. Flanagan JN, Young MV, Persons KS, Wang L, Mathieu JS, Whitlatch LW, et al. Vitamin D Metabolism in Human Prostate Cells: Implications for Prostate Cancer Chemoprevention by Vitamin D. Anticancer Res, 2006, 26(4a):2567–72. Kittaka A, Yoshida A, Chiang K-C, Takano M, Sawada D, Sakaki T, et al. Potent 19-norvitamin D Analogs for Prostate and Liver Cancer Therapy. Future Med Chem, 2012, 4(16):2049–65., DOI 10.4155/fmc.12.130 Elieh Ali Komi D, Sadeghi-Shabestari M, Shanebandi D, Babaloo Z, Razavi A, Sadigh-Eteghad S, et al. Investigation of Chitinase3like-1, Chiti3L1, Gene Polymorphism, Rs4950928, with Susceptibility to Allergic Asthma in Iranian Northwestern Azeri Population. J Res Mol Med, 2019, 7(4):17–24., DOI 10. 32598/rmm.7.4.17 Zeinaly I, Vossoughi N, Elieh Ali Komi D, Kazemi T, Babaloo Z, Razavi A, et al. Investigating the Association of Orosomucoid 1-like 3, ORMDL3, Gene Polymorphism, Rs12603332, with Susceptibility to Allergic Asthma in Iranian Northwestern Azeri Population. Iran J Allergy Asthma Immunol, 2018, 17(6):526–32., DOI 10.18502/ijaai.v17i6.615 Quint JK, Donaldson GC, Wassef N, Hurst JR, Thomas M, Wedzicha JA. 25- hydroxyvitamin D Deficiency, Exacerbation Frequency and Human Rhinovirus Exacerbations in Chronic Obstructive Pulmonary Disease. BMC Pulm Med, 2012, 12:28., DOI 10.1186/1471-2466-12-28 Cakir OO, Yilmaz A, Demir E, Incekara K, Kose MO, Ersoy N. Association of the BsmI, ApaI, TaqI, Tru9I and FokI Polymorphisms of the Vitamin D Receptor Gene with Nephrolithiasis in the Turkish Population. Urol J, 2016, 13(1):2509–18., DOI 10.22037/uj.v13i1.3383 Li F, Jiang L, Willis-Owen SA, Zhang Y, Gao J. Vitamin D Binding Protein Variants Associate with Asthma Susceptibility in the Chinese Han Population. BMC Med Genet, 2011, 12:103., DOI 10.1186/1471-2350-12-103 Ahonen MH, Tenkanen L, Teppo L, HakamaM, Tuohimaa P. Prostate Cancer Risk and Prediagnostic Serum 25-hydroxyvitamin D Levels, Finland). Cancer Causes Control, 2000, 11(9):847–52., DOI 10.1023/a:1008923802001 Atoum MF, AlKateeb D, Mahmoud SAA. The Fok1 Vitamin D Receptor Gene Polymorphism and 25(OH, D Serum Levels and Prostate Cancer Among Jordanian Men. Asian Pac J Cancer Prev, 2015, 16(6):2227–30., DOI 10.7314/ apjcp.2015.16.6.2227 Deschasaux M, Souberbielle J-C, Latino-Martel P, Sutton A, Charnaux N, Druesne-Pecollo N, et al. A Prospective Study of Plasma 25-hydroxyvitamin D Concentration and Prostate Cancer Risk. Br J Nutr, 2016, 115(2):305–14., DOI 10.1017/s0007114515004353 Majewski S, Skopinska M, Marczak M, Szmurlo A, Bollag W, Jablonska S. Vitamin D3 is a Potent Inhibitor of Tumor Cell-Induced Angiogenesis. J Investig Dermatol Symp Proc, 1996, 1(1):97–101. Nomura AMY, Stemmermann GN, Lee J, Kolonel LN, Chen TC, Turner A, et al. Serum Vitamin D Metabolite Levels and the Subsequent Development of Prostate Cancer, Hawaii, United States). Cancer Causes Control, 1998, 9(4): 425–32., DOI 10.1023/a:1008875819232 Ahn J, Peters U, Albanes D, Purdue MP, Abnet CC, Chatterjee N, et al. Serum Vitamin D Concentration and Prostate Cancer Risk: a Nested Case-Control Study. JNCI J Natl Cancer Inst, 2008, 100(11):796–804., DOI 10.1093/jnci/ djn152 Travis RC, Crowe FL, Allen NE, Appleby PN, Roddam AW, Tjonneland A, et al. Serum Vitamin D and Risk of Prostate Cancer in a Case-Control Analysis Nested within the European Prospective Investigation into Cancer and Nutrition, EPIC). Am J Epidemiol, 2009, 169(10):1223–32., DOI 10.1093/aje/ kwp022 Gilbert R, Bonilla C, Metcalfe C, Lewis S, Evans DM, Fraser WD, et al. Associations of Vitamin D Pathway Genes with Circulating 25- Hydroxyvitamin-D, 1,25-Dihydroxyvitamin-D, and Prostate Cancer: A Nested Case-Control Study. Cancer Causes Control, 2015, 26(2):205–18., DOI 10.1007/s10552-014-0500-5 Maneechay W, Boonpipattanapong T, Kanngurn S, Puttawibul P, Geater SL, Sangkhathat S. Single Nucleotide Polymorphisms in the Gc Gene for Vitamin D Binding Protein in Common Cancers in Thailand. Asian Pac J Cancer Prev, 2015, 16(8):3339–44., DOI 10.7314/apjcp.2015.16.8.3339 Kirac D, Dincer Yazan C, Gezmis H, Yaman A, Haklar G, Sirikci O, et al. VDBP, VDR Mutations and Other Factors Related with Vitamin D Metabolism May Be Associated with Type 1 Diabetes Mellitus. Cel Mol Biol, Noisy-le-grand,, 2018, 64(3):11–6. Newton DA, Baatz JE, Kindy MS, Gattoni-Celli S, Shary JR, Hollis BW, et al. Vitamin D Binding Protein Polymorphisms Significantly Impact Vitamin D Status in Children. Pediatr Res, 2019, 86(5):662–9., DOI 10.1038/s41390-019- 0322-y Chokkalingam AP, McGlynn KA, Gao YT, Pollak M, Deng J, Sesterhenn IA, et al. Vitamin D Receptor Gene Polymorphisms, Insulin-like Growth Factors, and Prostate Cancer Risk: a Population-Based Case-Control Study in China. Cancer Res, 2001, 61(11):4333–6. Ingles SA, Coetzee GA, Ross RK, Henderson BE, Kolonel LN, Crocitto L, et al. Association of Prostate Cancer with Vitamin D Receptor Haplotypes in African-Americans. Cancer Res, 1998, 58(8):1620–3. Huang S-P, Huang C-Y,Wu W-J, Pu Y-S, Chen J, Chen Y-Y, et al. Association of Vitamin D receptorFokI Polymorphism with Prostate Cancer Risk, Clinicopathological Features and Recurrence of Prostate Specific Antigen after Radical Prostatectomy. Int J Cancer, 2006, 119(8):1902–7., DOI 10.1002/ijc.22053 Ahn J, Albanes D, Berndt SI, Peters U, Chatterjee N, Freedman ND, et al. Vitamin D-Related Genes, Serum Vitamin D Concentrations and Prostate Cancer Risk. Carcinogenesis, 2009, 30(5):769–76., DOI 10.1093/carcin/bgp055 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610246 EP 1610253 DI 10.3389/pore.2022.1610246 PG 8 ER PT J AU Kothalawala, W Bartak, B Nagy, Zs Zsigrai, S Szigeti, K Valcz, G Takacs, I Kalmar, A Molnar, B AF Kothalawala, J. William Bartak, K. Barbara Nagy, B. Zsofia Zsigrai, Sara Szigeti, A. Krisztina Valcz, Gabor Takacs, Istvan Kalmar, Alexandra Molnar, Bela TI A Detailed Overview About the Single-Cell Analyses of Solid Tumors Focusing on Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE bioinformatics; colorectal cancer; multi-omics; heterogeneity; single-cell sequencing ID bioinformatics; colorectal cancer; multi-omics; heterogeneity; single-cell sequencing AB In recent years, the evolution of the molecular biological technical background led to the widespread application of single-cell sequencing, a versatile tool particularly useful in the investigation of tumor heterogeneity. Even 10 years ago the comprehensive characterization of colorectal cancers by The Cancer Genome Atlas was based on measurements of bulk samples. Nowadays, with single-cell approaches, tumor heterogeneity, the tumor microenvironment, and the interplay between tumor cells and their surroundings can be described in unprecedented detail. In this review article we aimed to emphasize the importance of single-cell analyses by presenting tumor heterogeneity and the limitations of conventional investigational approaches, followed by an overview of the whole single-cell analytic workflow from sample isolation to amplification, sequencing and bioinformatic analysis and a review of recent literature regarding the single-cell analysis of colorectal cancers. C1 [Kothalawala, J. William] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Bartak, K. Barbara] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Nagy, B. Zsofia] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Zsigrai, Sara] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Szigeti, A. Krisztina] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Valcz, Gabor] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Takacs, Istvan] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Kalmar, Alexandra] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Molnar, Bela] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. RP Kothalawala, W (reprint author), Semmelweis University, Department of Internal Medicine and Oncology, Budapest, Hungary. CR The Cancer Genome Atlas Network. Comprehensive Molecular Characterization of Human colon and Rectal Cancer. Nature, 2012, 487: 330–7., DOI 10.1038/nature11252 Bonnet D, Dick JE. Human Acute Myeloid Leukemia is Organized as a Hierarchy that Originates from a Primitive Hematopoietic Cell. Nat Med, 1997, 3(7):730–7., DOI 10.1038/nm0797-730 Shackleton M, Quintana E, Fearon ER, Morrison SJ. Heterogeneity in Cancer: Cancer Stem Cells versus Clonal Evolution. Cell, 2009, 138(5):822–9., DOI 10. 1016/j.cell.2009.08.017 Nowell PC. The Clonal Evolution of Tumor Cell Populations. Science, 1976, 194(4260):23–8., DOI 10.1126/science.959840 Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective Identification of Tumorigenic Breast Cancer Cells. Proc Natl Acad Sci U.S.A, 2003, 100(7):3983–8., DOI 10.1073/pnas.0530291100 Dirks PB. Brain Tumour Stem Cells: the Undercurrents of Human Brain Cancer and Their Relationship to Neural Stem Cells. Phil Trans R Soc B, 2008, 363:139–52., DOI 10.1098/rstb.2006.2017 Munro MJ, Wickremesekera SK, Peng L, Tan ST, Itinteang T. Cancer Stem Cells in Colorectal Cancer: A Review. J Clin Pathol, 2018, 71(2):110–6., DOI 10. 1136/jclinpath-2017-204739 Navin N, Krasnitz A, Rodgers L, Cook K, Meth J, Kendall J, et al. Inferring Tumor Progression from Genomic Heterogeneity. Genome Res, 2010, 20(1): 68–80., DOI 10.1101/gr.099622.109 Snuderl M, Fazlollahi L, Le LP, Nitta M, Zhelyazkova BH, Davidson CJ, et al. Mosaic Amplification of Multiple Receptor Tyrosine Kinase Genes in Glioblastoma. Cancer Cell, 2011, 20(6):810–7., DOI 10.1016/j.ccr.2011.11.005 Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing. N Engl J Med, 2012, 366(10):883–92., DOI 10.1056/ NEJMoa1113205 Li H, Fan X, Houghton J. Tumor Microenvironment: The Role of the Tumor Stroma in Cancer. J Cel Biochem., 2007, 101(4):805–15., DOI 10.1002/jcb.21159 Valcz G, Sipos F, Tulassay Z, Molnar B, Yagi Y. Importance of Carcinoma- Associated Fibroblast-Derived Proteins in Clinical Oncology. J Clin Pathol, 2014, 67(12):1026–31., DOI 10.1136/jclinpath-2014-202561 Wei R, Liu S, Zhang S,Min L, Zhu S. Cellular and Extracellular Components in Tumor Microenvironment and Their Application in Early Diagnosis of Cancers. Anal Cell Pathol, 2020, 2020:1–13., DOI 10.1155/2020/6283796 Idos GE, Kwok J, Bonthala N, Kysh L, Gruber SB, Qu C. The Prognostic Implications of Tumor Infiltrating Lymphocytes in Colorectal Cancer: A Systematic Review and Meta-Analysis. Sci Rep, 2020, 10(1):1–14., DOI 10. 1038/s41598-020-60255-4 Grigore A, Jolly M, Jia D, Farach-Carson M, Levine H. Tumor Budding: The Name Is EMT. Partial EMT. J Clin Med, 2016, 5(5):51., DOI 10.3390/ jcm5050051 Guo S, Ye Y, Liu X, Gong Y, Xu M, Song L, et al. Intra-Tumor Heterogeneity of Colorectal Cancer Necessitates the Multi-Regional Sequencing for Comprehensive Mutational Profiling. Cancer Manag Res, 2021, 13: 9209–23., DOI 10.2147/CMAR.S327596 Alizadeh AA, Aranda V, Bardelli A, Blanpain C, Bock C, Borowski C, et al. Toward Understanding and Exploiting Tumor Heterogeneity. Nat Med, 2015, 21(8):846–53., DOI 10.1038/nm.3915 Losi L, Baisse B, Bouzourene H, Benhattar J. Evolution of Intratumoral Genetic Heterogeneity during Colorectal Cancer Progression. Carcinogenesis, 2005, 26(5):916–22., DOI 10.1093/carcin/bgi044 Yohe S, Thyagarajan B. Review of Clinical Next-Generation Sequencing. Arch Pathol Lab Med, 2017, 141(11):1544–57., DOI 10.5858/arpa.2016- 0501-RA Gross A, Schoendube J, Zimmermann S, Steeb M, Zengerle R, Koltay P. Technologies for Single-Cell Isolation. Int J Mol Sci, 2015, 16(8):16897–919., DOI 10.3390/ijms160816897 Hu P, Zhang W, Xin H, Deng G. Single Cell Isolation and Analysis. Front Cel Dev. Biol., 2016, 4(Oct)., DOI 10.3389/fcell.2016.00116 Lu Z, Moraes C, Zhao Y, You L, Simmons CA, Sun Y. A Micromanipulation System for Single Cell Deposition. In: 2010 IEEE International Conference on Robotics and Automation, Anchorage, AK, May 03–07, 2010. Anchorage, AK, 2010). p. 494–9., DOI 10.1109/ROBOT.2010.5509784 Liao X, Makris M, Luo XM. Fluorescence-activated Cell Sorting for Purification of Plasmacytoid Dendritic Cells from the Mouse Bone Marrow. J Vis Exp, 2016, 117:54641., DOI 10.3791/54641 Penter L, Dietze K, Bullinger L, Westermann J, Rahn HP, Hansmann L. FACS Single Cell index Sorting is Highly Reliable and Determines Immune Phenotypes of Clonally Expanded T Cells. Eur J Immunol, 2018, 48(7): 1248–50., DOI 10.1002/eji.201847507 Zhang H, Liu K-K. Optical Tweezers for Single Cells. J R Soc Interf, 2008, 5(24):671–90., DOI 10.1098/rsif.2008.0052 Ladanyi A, Sipos F, Szoke D, Galamb O, Molnar B, Tulassay Z. Laser Microdissection in Translational and Clinical Research. Cytometry, 2006, 69A(9):947–60., DOI 10.1002/cyto.a.20322 Huang L, Ma F, Chapman A, Lu S, Xie XS. Single-Cell Whole-Genome Amplification and Sequencing: Methodology and Applications. Annu Rev Genom Hum Genet, 2015, 16(1):79–102., DOI 10.1146/annurev-genom- 090413-025352 Zong C. Multiple Annealing and Looping-Based Amplification Cycles, MALBAC, for the Analysis of DNA Copy Number Variation. In: JM Frade FH Gage, editors. Genomic Mosaicism in Neurons and Other Cell Types, 131. New York, NY: Springer New York, 2017). p. 133–42., DOI 10.1007/ 978-1-4939-7280-7_7 Saliba A-E, Westermann AJ, Gorski SA, Vogel J. Single-cell RNA-Seq: Advances and Future Challenges. Nucleic Acids Res, 2014, 42(14):8845–60., DOI 10.1093/nar/gku555 Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, et al. mRNA-Seq Whole-Transcriptome Analysis of a Single Cell. Nat Methods, 2009, 6(5): 377–82., DOI 10.1038/nmeth.1315 Eberwine J, Yeh H, Miyashiro K, Cao Y, Nair S, Finnell R, et al. Analysis of Gene Expression in Single Live Neurons. Proc Natl Acad Sci U.S.A, 1992, 89(7):3010–4., DOI 10.1073/pnas.89.7.3010 Picelli S, Bjorklund AK, Faridani OR, Sagasser S, Winberg G, Sandberg R. Smart-seq2 for Sensitive Full-Length Transcriptome Profiling in Single Cells. Nat Methods, 2013, 10(11):1096–8., DOI 10.1038/nmeth.2639 Soumillon M, Cacchiarelli D, Semrau S, van Oudenaarden A, Mikkelsen TS. Characterization of Directed Differentiation by High-Throughput Single-Cell RNA-Seq. bioRxiv, 2014). Preprint., DOI 10.1101/003236 Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, et al. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell, 2015, 161(5):1202–14., DOI 10.1016/j.cell.2015.05.002 Jaitin DA, Kenigsberg E, Keren-Shaul H, Elefant N, Paul F, Zaretsky I, et al. Massively Parallel Single-Cell RNA-Seq for Marker-free Decomposition of Tissues into Cell Types. Science, 2014, 343(6172):776–9., DOI 10.1126/science. 1247651 Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, et al. Droplet Barcoding for Single-Cell Transcriptomics Applied to Embryonic Stem Cells. Cell, 2015, 161(5):1187–201., DOI 10.1016/j.cell.2015.04.044 Hashimshony T,Wagner F, Sher N, Yanai I. CEL-seq: Single-Cell RNA-Seq by Multiplexed Linear Amplification. Cel Rep, 2012, 2(3):666–73., DOI 10.1016/j. celrep.2012.08.003 Clark SJ, Smallwood SA, Lee HJ, Krueger F, Reik W, Kelsey G. Genome-wide Base-Resolution Mapping of DNA Methylation in Single Cells Using Single- Cell Bisulfite Sequencing, scBS-Seq). Nat Protoc, 2017, 12(3):534–47., DOI 10. 1038/nprot.2016.187 Rotem A, Ram O, Shoresh N, Sperling RA, Goren A, Weitz DA, et al. Singlecell ChIP-Seq Reveals Cell Subpopulations Defined by Chromatin State. Nat Biotechnol, 2015, 33(11):1165–72., DOI 10.1038/nbt.3383 Buenrostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales ML, Snyder MP, et al. Single-cell Chromatin Accessibility Reveals Principles of Regulatory Variation. Nature, 2015, 523(7561):486–90., DOI 10.1038/nature14590 Schwartzman O, Tanay A. Single-cell Epigenomics: Techniques and Emerging Applications. Nat Rev Genet, 2015, 16(12):716–26., DOI 10. 1038/nrg3980 Li X, Wang C-Y. From Bulk, Single-Cell to Spatial RNA Sequencing. Int J Oral Sci, 2021, 13(1):36., DOI 10.1038/s41368-021-00146-0 Vannay A, Fekete A, Adori C, Toth T, Losonczy G, Laszlo L, et al. Divergence of Renal Vascular Endothelial Growth Factor mRNA Expression and Protein Level in post-ischaemic Rat Kidneys: Post-transcriptional Regulation of Renal VEGF Synthesis. Exp Physiol, 2004, 89(4):435–44., DOI 10.1113/expphysiol. 2004.027516 Luquette LJ, Bohrson CL, Sherman MA, Park PJ. Identification of Somatic Mutations in Single Cell DNA-Seq Using a Spatial Model of Allelic Imbalance. Nat Commun, 2019, 10(1):3908., DOI 10.1038/s41467-019-11857-8 Mallory XF, Edrisi M, Navin N, Nakhleh L. Methods for Copy Number Aberration Detection from Single-Cell DNA-Sequencing Data. Genome Biol, 2020, 21(1)., DOI 10.1186/s13059-020-02119-8 Daniel Lai GH. HMMcopy: Copy Number Prediction with Correction for GC and Mappability Bias for HTS Data. Bioconductor, 2021)., DOI 10.18129/B9. BIOC.HMMCOPY Bakker B, Taudt A, Belderbos ME, Porubsky D, Spierings DCJ, de Jong TV, et al. Single-cell Sequencing Reveals Karyotype Heterogeneity in Murine and HumanMalignancies. Genome Biol, 2016, 17(1):115., DOI 10.1186/s13059-016- 0971-7 Garvin T, Aboukhalil R, Kendall J, Baslan T, Atwal GS, Hicks J, et al. Interactive Analysis and Assessment of Single-Cell Copy-Number Variations. Nat Methods, 2015, 12(11):1058–60., DOI 10.1038/nmeth.3578 Wang X, Chen H, Zhang NR. DNA Copy Number Profiling Using Single-Cell Sequencing. Brief Bioinform, 2018, 19(5):731–6., DOI 10.1093/bib/bbx004 Yu X, Abbas-Aghababazadeh F, Chen YA, Fridley BL. Statistical and Bioinformatics Analysis of Data from Bulk and Single-Cell RNA Sequencing Experiments. Methods Mol Biol, 2021, 2194:143–75., DOI 10. 1007/978-1-0716-0849-4_9 Trapnell C, Pachter L, Salzberg SL. TopHat: Discovering Splice Junctions with RNA-Seq. Bioinformatics, 2009, 25(9):1105–11., DOI 10.1093/bioinformatics/ btp120 Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: Ultrafast Universal RNA-Seq Aligner. Bioinformatics, 2013, 29(1):15–21., DOI 10.1093/bioinformatics/bts635 Li H, Durbin R. Fast and Accurate Short Read Alignment with Burrows- Wheeler Transform. Bioinformatics, 2009, 25(14):1754–60., DOI 10.1093/ bioinformatics/btp324 Langmead B, Salzberg SL. Fast Gapped-Read Alignment with Bowtie 2. Nat Methods, 2012, 9(4):357–9., DOI 10.1038/nmeth.1923 Lu B, Zeng Z, Shi T. Comparative Study of De Novo Assembly and Genome- Guided Assembly Strategies for Transcriptome Reconstruction Based on RNA-Seq. Sci China Life Sci, 2013, 56(2):143–55., DOI 10.1007/s11427-013- 4442-z Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, et al. Transcript Assembly and Quantification by RNA-Seq Reveals Unannotated Transcripts and Isoform Switching during Cell Differentiation. Nat Biotechnol, 2010, 28(5):511–5., DOI 10.1038/nbt.1621 Bankevich A, Nurk S, Antipov D, Gurevich AA, Dvorkin M, Kulikov AS, et al. SPAdes: A New Genome Assembly Algorithm and its Applications to Single- Cell Sequencing. J Comput Biol, 2012, 19(5):455–77., DOI 10.1089/cmb.2012. 0021 Lun ATL, McCarthy DJ, Marioni JC. A Step-by-step Workflow for Low-Level Analysis of Single-Cell RNA-Seq Data with Bioconductor. F1000Res, 2016, 5: 2122., DOI 10.12688/f1000research.9501.2 Bacher R, Chu L-F, Leng N, Gasch AP, Thomson JA, Stewart RM, et al. SCnorm: Robust Normalization of Single-Cell RNA-Seq Data. Nat Methods, 2017, 14(6):584–6., DOI 10.1038/nmeth.4263 Jia C, Hu Y, Kelly D, Kim J, Li M, Zhang NR. Accounting for Technical Noise in Differential Expression Analysis of Single-Cell RNA Sequencing Data. Nucleic Acids Res, 2017, 45(19):10978–88., DOI 10.1093/nar/gkx754 Kharchenko PV, Silberstein L, Scadden DT. Bayesian Approach to Single-Cell Differential Expression Analysis. Nat Methods, 2014, 11(7):740–2., DOI 10. 1038/nmeth.2967 Cole MB, Risso D, Wagner A, DeTomaso D, Ngai J, Purdom E, et al. Performance Assessment and Selection of Normalization Procedures for Single-Cell RNA-Seq. Cel Syst, 2019, 8(4):315–28., DOI 10.1016/j.cels.2019. 03.010 Wang T, Li B, Nelson CE, Nabavi S. Comparative Analysis of Differential Gene Expression Analysis Tools for Single-Cell RNA Sequencing Data. BMC Bioinform, 2019, 20(1):40., DOI 10.1186/s12859-019-2599-6 Yu C, Yu J, Yao X, Wu WK, Lu Y, Tang S, et al. Discovery of Biclonal Origin and a Novel Oncogene SLC12A5 in colon Cancer by Single-Cell Sequencing. Cell Res, 2014, 24(6):701–12., DOI 10.1038/cr.2014.43 Wu H, Zhang X-Y, Hu Z, Hou Q, Zhang H, Li Y, et al. Evolution and Heterogeneity of Non-hereditary Colorectal Cancer Revealed by Single-Cell Exome Sequencing. Oncogene, 2017, 36(20):2857–67., DOI 10.1038/onc. 2016.438 Roerink SF, Sasaki N, Lee-Six H, Young MD, Alexandrov LB, Behjati S, et al. Intra-tumour Diversification in Colorectal Cancer at the Single-Cell Level. Nature, 2018, 556(7702):457–62., DOI 10.1038/s41586-018-0024-3 Bian S, Hou Y, Zhou X, Li X, Yong J, Wang Y, et al. Single-cell Multiomics Sequencing and Analyses of Human Colorectal Cancer. Science, 2018, 362(6418):1060–3., DOI 10.1126/science.aao3791 Zhou Y, Bian S, Zhou X, Cui Y, Wang W, Wen L, et al. Single-Cell Multiomics Sequencing Reveals Prevalent Genomic Alterations in Tumor Stromal Cells of Human Colorectal Cancer. Cancer Cell, 2020, 38(6):818–82., DOI 10.1016/j. ccell.2020.09.015 Wang Q, Wang Z, Zhang Z, Zhang W, Zhang M, Shen Z, et al. Landscape of Cell Heterogeneity and Evolutionary Trajectory in Ulcerative Colitis- Associated Colon Cancer Revealed by Single-Cell RNA Sequencing. Chin J Cancer Res, 2021, 33(2):271–88., DOI 10.21147/j.issn.1000-9604.2021.02.13 Liu Y, Liu X, Xu Q, Gao X, Linghu E. A Prognostic Model of Colon Cancer Based on the Microenvironment Component Score via Single Cell Sequencing. In Vivo, 2022, 36(2):753–63., DOI 10.21873/invivo.12762 Arneson N, Hughes S, Houlston R, Done S. Whole-Genome Amplification by Degenerate Oligonucleotide Primed PCR, DOP-PCR). Cold Spring Harb Protoc, 2008, 2008:pdb.prot4919., DOI 10.1101/pdb.prot4919 Silander K, Saarela J. Whole Genome Amplification with Phi29 DNA Polymerase to Enable Genetic or Genomic Analysis of Samples of Low DNA Yield. In: M Starkey R Elaswarapu, editors. Genomics Protocols, 439. Totowa, NJ: Humana Press, 2008). p. 1–18., DOI 10.1007/978-1-59745-188-8_1 Sasagawa Y, Nikaido I, Hayashi T, Danno H, Uno KD, Imai T, et al. Quartz- Seq: A Highly Reproducible and Sensitive Single-Cell RNA Sequencing Method, Reveals Non-genetic Gene-Expression Heterogeneity. Genome Biol, 2013, 14(4):R31., DOI 10.1186/gb-2013-14-4-r31 Ramskold D, Luo S, Wang Y-C, Li R, Deng Q, Faridani OR, et al. Fulllength mRNA-Seq from Single-Cell Levels of RNA and Individual Circulating Tumor Cells. Nat Biotechnol, 2012, 30(8):777–82., DOI 10. 1038/nbt.2282 Nygaard V. Options Available for Profiling Small Samples: A Review of Sample Amplification Technology when Combined with Microarray Profiling. Nucleic Acids Res, 2006, 34(3):996–1014., DOI 10.1093/nar/ gkj499 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610342 EP 1610351 DI 10.3389/pore.2022.1610342 PG 10 ER PT J AU Li, G Liu, D Flandrin, P Zhang, Y Lambert, C Mallouk, N Cottier, M AF Li, Guorong Liu, Dongdong Flandrin, Pascale Zhang, Yang Lambert, Claude Mallouk, Nora Cottier, Michele TI Tumor-Derived Exosomal RNA From Fine-Needle Aspiration Supernatant as a Novel Liquid Biopsy for Molecular Diagnosis of Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE exosomes; pancreatic adenocarcinoma; liquid biopsy; FNA supernatant; molecular diagnosis; glypican 1 ID exosomes; pancreatic adenocarcinoma; liquid biopsy; FNA supernatant; molecular diagnosis; glypican 1 AB Background: We hypothesized that the fine needle aspiration (FNA) supernatant from tumor might contain tumor-derived exosomes. The objective of this pilot study was to test if tumor-derived exosomal RNA could be found in FNA supernatants for molecular diagnosis of cancer. Methods: 10 FNA samples from pancreatic tumor were included. After the routine recuperation of cellular material by centrifugation, the cell-free Cytolyt liquid was collected instead of being discarded. 10 ml Cytolyt was used to isolate the exosomes. Transmission electronic microscopy (TEM) was used to examine the presence of exosomes. The exosomal marker CD63 was analyzed by flow cytometry. The exosomal RNA was extracted. RT-qPCR was performed to detect the GAPDH and the tumor marker of glypican 1 gene expression. Results: TEM confirmed the presence of exosomes from FNA supernatants. Flow cytometry showed a strong positive expression of exosome marker CD63. The concentration of exosomal RNA ranged from 18.81 to 354.75 ng/μl with an average of 81.76 ng/μl. The average exosomal RNA quantity was 1390.01 ng (range from 319.77 to 6030.75 ng) with an average 260/280 ratio of 2.12. GAPDH was detectable in all samples. Exosomal glypican 1 was detected in all samples of pancreatic ductal adenorcarcinomas (3/3) and absent from benign cystic samples (3/3). Furthermore, exosomal glypican 1 was positive in one sample with a non-contributive cytology and in one sample in which no malignant cell was found. Conclusion: This is the first report that the supernatants from FNA biopsy may contain tumor-derived exosomal RNA. These tumor-derived exosomes from FNA may provide a new liquid biopsy for the molecular diagnosis of cancer. C1 [Li, Guorong] North Hospital, CHU Saint-Etienne, Department of Digestive Surgery and UrologySaint-Etienne, France. [Liu, Dongdong] The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Department of Laboratory ScienceGuangzhou, China. [Flandrin, Pascale] North Hospital, CHU Saint-Etienne, Laboratory of Molecular BiologySaint-Etienne, France. [Zhang, Yang] Guangzhou HopeTech Biological Technology Co., Ltd.Guangzhou, China. [Lambert, Claude] North Hospital, CHU Saint- Etienne, Immunology Laboratory, Section of Flow CytometrySaint-Etienne, France. [Mallouk, Nora] University Jean Monnet, Faculty of Medicine, Center of Electronic Microscopy, CMESSaint-Etienne, France. [Cottier, Michele] North Hospital, CHU Saint-Etienne, Laboratory of CytopathologySaint-Etienne, France. RP Li, G (reprint author), North Hospital, CHU Saint-Etienne, Department of Digestive Surgery and Urology, Saint-Etienne, France. EM grli2001@yahoo.fr CR McGuigan A, Kelly P, Turkington RC, Jones C, Coleman HG, McCain RS, et al. Pancreatic Cancer: A Review of Clinical Diagnosis, Epidemiology, Treatment and Outcomes. World J Gastroenterol, 2018, 24(43):4846–61., DOI 10.3748/wjg. v24.i43.4846 Siegel RL,Miller KD, Jemal A. Cancer Statistics, 2020. CA Cancer J Clin, 2020, 70(1):7–30., DOI 10.3322/caac.21590 Groot VP, Gemenetzis G, Blair AB, Rivero-Soto RJ, Yu J, Javed AA, et al. Defining and Predicting Early Recurrence in 957 Patients with Resected Pancreatic Ductal Adenocarcinoma. Ann Surg, 2019, 269(6):1154–62., DOI 10.1097/SLA.0000000000002734 Smith EH, Bartrum RJ, Chang YC, D’Orsi CJ, Lokich J, Abbruzzese A, et al. Percutaneous Aspiration Biopsy of the Pancreas under Ultrasonic Guidance. N Engl J Med, 1975, 292(16):825–8., DOI 10.1056/nejm197504172921603 Zamboni GA, D’Onofrio M, Idili A, Malago R, Iozzia R, Manfrin E, et al. Ultrasound-guided Percutaneous fine-needle Aspiration of 545 Focal Pancreatic Lesions. Am J Roentgenol, 2009, 193(6):1691–5., DOI 10.2214/ AJR.09.2958 Jhala N, Jhala D, Vickers SM, Eltoum I, Batra SK, Manne U, et al. Biomarkers in Diagnosis of Pancreatic Carcinoma in Fine-Needle Aspirates. Am J Clin Pathol, 2006, 126(4):572–9., DOI 10.1309/cev30be088cbdqd9 Palmirotta R, Lovero D, Cafforio P, Felici C, Mannavola F, Pelle E, et al. Liquid Biopsy of Cancer: A Multimodal Diagnostic Tool in Clinical Oncology. Ther AdvMed Oncol, 2018, 10:1758835918794630., DOI 10.1177/1758835918794630 Keller S, Ridinger J, Rupp AK, Janssen JW, Altevogt P. Body Fluid Derived Exosomes as a Novel Template for Clinical Diagnostics. J Transl Med, 2011, 9: 86., DOI 10.1186/1479-5876-9-86 Finkelstein SD, Bibbo M, Kowalski TE, Loren DE, Siddiqui AA, Solomides C, et al. Mutational Analysis of Cytocentrifugation Supernatant Fluid from Pancreatic Solid Mass Lesions. Diagn Cytopathol, 2014, 42(8):719–25., DOI 10.1002/dc.23048 Deftereos G, Finkelstein SD, Jackson SA, Ellsworth EM, Krishnamurti U, Liu Y, et al. The Value of Mutational Profiling of the Cytocentrifugation Supernatant Fluid from fine-needle Aspiration of Pancreatic Solid Mass Lesions. Mod Pathol, 2014, 27(4):594–601., DOI 10.1038/modpathol. 2013.147 Roy-Chowdhuri S, Mehrotra M, Bolivar AM, Kanagal-Shamanna R, Barkoh BA, Hannigan B, et al. Salvaging the Supernatant: Next Generation Cytopathology for Solid Tumor Mutation Profiling. Mod Pathol, 2018, 31(7):1036–45., DOI 10.1038/s41379-018-0006-x Wen J, Li G, Berremila SA, Klein JP, Peoc’h M, Cottier M, et al. Assessment of Cellular Adequacy of fine Needle Aspiration Biopsy for Small Solid Renal Tumors. Cytopathology, 2018, 29(5):444–8., DOI 10.1111/cyt.12579 VanderLaan PA.MolecularMarkers: Implications for Cytopathology and Specimen Collection. Cancer Cytopathol, 2015, 123(8):454–60., DOI 10.1002/cncy.21560 Coley SM, Crapanzano JP, Saqi A. FNA, Core Biopsy, or Both for the Diagnosis of Lung Carcinoma: Obtaining Sufficient Tissue for a Specific Diagnosis andMolecular Testing. Cancer Cytopathol, 2015, 123(5):318–26., DOI 10.1002/cncy.21527 Schneider F, Smith MA, Lane MC, Pantanowitz L, Dacic S, Ohori NP, et al. Adequacy of Core Needle Biopsy Specimens and fine-needle Aspirates for Molecular Testing of Lung Adenocarcinomas. Am J Clin Pathol, 2015, 143(2): 193–200., DOI 10.1309/AJCPMY8UI7WSFSYY Fetzer R, Duey M, Pena V, Wanzer D, Kirkpatrick J, Chau D, et al. Role of Cytotechnologists in Rapid Onsite Adequacy Assessment of Cytology Materials for Diagnostic Workup and Specimen Allocation for Ancillary Testing Using a Standardized Protocol. J Am Soc Cytopathol, 2020, 9(2): 67–75., DOI 10.1016/j.jasc.2019.08.005 Hannigan B, YeW, MehrotraM, Lam V, Bolivar A, Zalles S, et al. Liquid Biopsy Assay for Lung Carcinoma Using Centrifuged Supernatants from fine-needle Aspiration Specimens. Ann Oncol, 2019, 30(6):963–9., DOI 10.1093/annonc/mdz102 Ye W, Hannigan B, Zalles S, Mehrotra M, Barkoh BA, Williams MD, et al. Centrifuged Supernatants from FNA Provide a Liquid Biopsy Option for Clinical Next-Generation Sequencing of Thyroid Nodules. Cancer Cytopathol, 2019, 127(3):146–60., DOI 10.1002/cncy.22098 Lund ME, Campbell DH, Walsh BJ. The Role of Glypican-1 in the Tumour Microenvironment. Adv Exp Med Biol, 2020, 1245:163–76., DOI 10.1007/978-3- 030-40146-7_8 Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, et al. Glypican-1 Identifies Cancer Exosomes and Detects Early Pancreatic Cancer. Nature, 2015, 523(7559):177–82., DOI 10.1038/nature14581 Frampton AE, Prado MM, Lopez-Jimenez E, Fajardo-Puerta AB, Jawad ZAR, Lawton P, et al. Glypican-1 Is Enriched in Circulating-Exosomes in Pancreatic Cancer and Correlates with Tumor burden. Oncotarget, 2018, 9(27):19006–13., DOI 10.18632/oncotarget.24873 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610344 EP 1610349 DI 10.3389/pore.2022.1610344 PG 6 ER PT J AU Wang, J Akter, R AF Wang, Jie Akter, Rehana TI Cancer-Associated Stromal Fibroblast-Derived Transcriptomes Predict Poor Clinical Outcomes and Immunosuppression in Colon Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colon cancer; survival time; immunosuppressive roles; metastatic scores; cancer-associated fibroblasts ID colon cancer; survival time; immunosuppressive roles; metastatic scores; cancer-associated fibroblasts AB Background: Previous studies revealed that colonic cancer-associated fibroblasts (CAFs) are associated with the modulation of the colon tumor microenvironment (TME). However, identification of key transcriptomes and their correlations with the survival prognosis, immunosuppression, tumor progression, and metastasis in colon cancer remains lacking. Methods: We used the GSE46824, GSE70468, GSE17536, GSE35602, and the cancer genome atlas (TCGA) colon adenocarcinoma (COAD) datasets for this study.We identified the differentially expressed genes (DEGs), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, hub genes, and survival-associated genes in colon cancer. Finally, we investigated the correlation of key genes with the survival prognosis, immunosuppression, and metastasis. Results: We identified 246 common DEGs between the GSE46824 and GSE70468 datasets of colonic CAFs, which included 72 upregulated and 174 downregulated genes. The upregulated pathways are mainly involved with cancers and cellular signaling, and downregulated pathways are involvedwith immune regulation and cellular metabolism. The search tool for the retrieval of interacting genes (STRING)-based analysis identified 15 hub genes and 9 significant clusters in colonic CAFs. The upregulation of CTHRC1, PDGFC, PDLIM3, NTM, and SLC16A3 and downregulation of FBN2 are correlated with a shorter survival time in colon cancer. The CTHRC1, PDGFC, PDLIM3, and NTM genes are positively correlated with the infiltration of tumor-associated macrophages (TAM), macrophages, M2 macrophages, the regulatory T cells (Tregs), T cell exhaustion, and myeloid-derived suppressor cells (MDSCs), indicating the immunosuppressive roles of these transcriptomes in colon cancer. Moreover, the CTHRC1, PDGFC, PDLIM3, NTM, and SLC16A3 genes are gradually increased from normal tissue to the tumor and tumor to the metastatic tumor, and FBN2 showed the reverse pattern. Furthermore, the CTHRC1, FBN2, PDGFC, PDLIM3, and NTM genes are positively correlated with the metastatic scores in colon cancer. Then, we revealed that the expression value of CTHRC1, FBN2, PDGFC, PDLIM3, NTM, and SLC16A3 showed the diagnostic efficacy in colonic CAFs. Finally, the expression level of CTHRC1, PDGFC, and NTM genes are consistently altered in colon tumor stroma as well as in the higher CAFs-group of TCGA COAD patients. Conclusion: The identified colonic CAFs-derived key genes are positively correlated with survival prognosis, immunosuppression, tumor progression, and metastasis. C1 [Wang, Jie] First Affiliated Hospital of Xinjiang Medical University, Department of PharmacyUrumqi, China. [Akter, Rehana] Center for Research Innovation and Development (CRID), Bioinformatics Research Lab.Dhaka, Bangladesh. CR Valkenburg KC, de Groot AE, Pienta KC. Targeting the Tumour Stroma to Improve Cancer Therapy. Nat Rev Clin Oncol, 2018, 15:366–81., DOI 10.1038/ s41571-018-0007-1 Zhang R, Qi F, Shao S, Li G, Feng Y. Human Colorectal Cancer-Derived Carcinoma Associated Fibroblasts Promote CD44-Mediated Adhesion of Colorectal Cancer Cells to Endothelial Cells by Secretion of HGF. Cancer Cel Int, 2019, 19:192., DOI 10.1186/s12935-019-0914-y Musa M, Ali A. Cancer-associated Fibroblasts of Colorectal Cancer and Their Markers: Updates, Challenges and Translational Outlook. Future Oncol, 2020, 16:2329–44., DOI 10.2217/fon-2020-0384 Unterleuthner D, Neuhold P, Schwarz K, Janker L, Neuditschko B, Nivarthi H, et al. Cancer-associated Fibroblast-Derived WNT2 Increases Tumor Angiogenesis in colon Cancer. Angiogenesis, 2020, 23:159–77., DOI 10.1007/ s10456-019-09688-8 Wu J, Liang C, Chen M, Su W. Association between Tumor-Stroma Ratio and Prognosis in Solid Tumor Patients: A Systematic Review and Meta-Analysis. Oncotarget, 2016, 7:68954–65., DOI 10.18632/oncotarget.12135 Berdiel-Acer M, Sanz-Pamplona R, Calon A, Cuadras D, Berenguer A, Sanjuan X, et al. Differences between CAFs and Their Paired NCF from Adjacent Colonic Mucosa Reveal Functional Heterogeneity of CAFs, Providing Prognostic Information. Mol Oncol, 2014, 8:1290–305., DOI 10.1016/j. molonc.2014.04.006 Sandberg TP, Stuart MPME, Oosting J, Tollenaar RAEM, Sier CFM, Mesker WE. Increased Expression of Cancer-Associated Fibroblast Markers at the Invasive Front and its Association with Tumor-Stroma Ratio in Colorectal Cancer. BMC Cancer, 2019, 19:284., DOI 10.1186/s12885-019-5462-2 Xing F, Saidou J, Watabe K. Cancer Associated Fibroblasts, CAFs, in Tumor Microenvironment. Front Biosci, 2010, 15:166–79., DOI 10.2741/3613 Gascard P, Tlsty TD. Carcinoma-associated Fibroblasts: Orchestrating the Composition of Malignancy. Genes Dev, 2016, 30:1002–19., DOI 10.1101/gad. 279737.116 Cirri P, Chiarugi P. Cancer-associated-fibroblasts and Tumour Cells: A Diabolic Liaison Driving Cancer Progression. Cancer Metastasis Rev, 2012, 31:195–208., DOI 10.1007/s10555-011-9340-x Liu T, Han C,Wang S, Fang P, Ma Z, Xu L, et al. Cancer-associated Fibroblasts: an Emerging Target of Anti-cancer Immunotherapy. J Hematol Oncol, 2019, 12:86., DOI 10.1186/s13045-019-0770-1 Isella C, Terrasi A, Bellomo SE, Petti C, Galatola G, Muratore A, et al. Stromal Contribution to the Colorectal Cancer Transcriptome. Nat Genet, 2015, 47: 312–9., DOI 10.1038/ng.3224 Becht E, de Reynies A, Giraldo NA, Pilati C, Buttard B, Lacroix L, et al. Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy. Clin Cancer Res, 2016, 22:4057–66., DOI 10.1158/1078-0432.CCR-15-2879 Zheng H, Liu H, Ge Y, Wang X. Integrated Single-Cell and Bulk RNA Sequencing Analysis Identifies a Cancer Associated Fibroblast-Related Signature for Predicting Prognosis and Therapeutic Responses in Colorectal Cancer. Cancer Cel Int, 2021, 21:552., DOI 10.1186/s12935-021-02252-9 Dienstmann R, Villacampa G, Sveen A, Mason MJ, NieDzwiecki D, NesbAkken A, et al. Relative Contribution of Clinicopathological Variables, Genomic Markers, Transcriptomic Subtyping and Microenvironment Features for Outcome Prediction in Stage II/III Colorectal Cancer. Ann Oncol, 2019, 30:1622–9., DOI 10.1093/annonc/mdz287 Baulida J. Epithelial-to-mesenchymal Transition Transcription Factors in Cancer-Associated Fibroblasts. Mol Oncol, 2017, 11:847–59., DOI 10.1002/ 1878-0261.12080 Liu B, Pan S, Liu J, Kong C. Cancer-associated Fibroblasts and the Related Runt-Related Transcription Factor 2, RUNX2, Promote Bladder Cancer Progression. Gene, 2021, 775:145451., DOI 10.1016/j.gene.2021.145451 NCBI. GEO Accession Viewer, 2019). Available at: https://www.ncbi.nlm.nih. gov/geo/query/acc.cgi?acc=GSE46824, Accessed Feb 13, 2019). Ferrer-Mayorga G, Gomez-Lopez G, Barbachano A, Fernandez-Barral A, Pena C, Pisano DG, et al. Vitamin D Receptor Expression and Associated Gene Signature in Tumour Stromal Fibroblasts Predict Clinical Outcome in Colorectal Cancer. Gut, 2017, 66:1449–62., DOI 10.1136/gutjnl-2015-310977 Cancer Genome Atlas Network. Comprehensive Molecular Characterization of Human colon and Rectal Cancer. Nature, 2012, 487:330–7., DOI 10.1038/ nature11252 Tang Z, Li C, Kang B, Gao G, Zhang Z. GEPIA: A Web Server for Cancer and normal Gene Expression Profiling and Interactive Analyses. Nucleic Acids Res, 2017, 45:W98–W102., DOI 10.1093/nar/gkx247 Mizuno H, Kitada K, Nakai K, Sarai A. PrognoScan: A New Database for Meta- Analysis of the Prognostic Value of Genes. BMC Med Genomics, 2009, 2:18., DOI 10.1186/1755-8794-2-18 Smith JJ, Deane NG, Wu F, Merchant NB, Zhang B, Jiang A, et al. Experimentally Derived Metastasis Gene Expression Profile Predicts Recurrence and Death in Patients with colon Cancer. Gastroenterology, 2010, 138:958–68., DOI 10.1053/j.gastro.2009.11.005 Nishida N, Nagahara M, Sato T, Mimori K, Sudo T, Tanaka F, et al. Microarray Analysis of Colorectal Cancer Stromal Tissue Reveals Upregulation of Two Oncogenic miRNA Clusters. Clin Cancer Res, 2012, 18:3054–70., DOI 10.1158/ 1078-0432.CCR-11-1078 Davis S, Meltzer PS. GEOquery: a Bridge between the Gene Expression Omnibus, GEO, and BioConductor. Bioinformatics, 2007, 23:1846–7., DOI 10.1093/bioinformatics/btm254 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res, 2015, 43:e47., DOI 10.1093/nar/gkv007 Wang J, Uddin MN, Akter R, Wu Y. Contribution of Endothelial Cell-Derived Transcriptomes to the colon Cancer Based on Bioinformatics Analysis. Math Biosci Eng, 2021, 18:7280–300., DOI 10.3934/mbe.2021360 Kim N, Kim HK, Lee K, Hong Y, Cho JH, Choi JW, et al. Single-cell RNA Sequencing Demonstrates the Molecular and Cellular Reprogramming of Metastatic Lung Adenocarcinoma. Nat Commun, 2020, 11:2285., DOI 10. 1038/s41467-020-16164-1 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: A Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci U S A, 2005, 102:15545–50., DOI 10.1073/pnas.0506580102 Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: New Perspectives on Genomes, Pathways, Diseases and Drugs. Nucleic Acids Res, 2017, 45:D353–D361., DOI 10.1093/nar/gkw1092 Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING V11: Protein–Protein Association Networks with Increased Coverage, Supporting Functional Discovery in Genome-wide Experimental Datasets. Nucleic Acids Res, 2019, 47:D607–D613., DOI 10.1093/nar/gky1131 Chin C-H, Chen S-H, Wu H-H, Ho CW, Ko MT, Lin CY. cytoHubba: Identifying Hub Objects and Sub-networks from Complex Interactome. BMC Syst Biol, 2014, 8:S11., DOI 10.1186/1752-0509-8-S4-S11 Shannon P,Markiel A, OzierO, BaligaNS, Wang JT, Ramage D, et al. Cytoscape: A Software Environment for Integrated Models of Biomolecular Interaction Networks. Genome Res, 2003, 13:2498–504., DOI 10.1101/gr.1239303 Bader GD, Hogue CW. An Automated Method for Finding Molecular Complexes in Large Protein Interaction Networks. BMC Bioinformatics, 2003, 4:2., DOI 10.1186/1471-2105-4-2 Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, KimH, Torres-Garcia W, et al. Inferring Tumour Purity and Stromal and Immune Cell Admixture from Expression Data. Nat Commun, 2013, 4:2612., DOI 10.1038/ncomms3612 Hanzelmann S, Castelo R, Guinney J. GSVA: Gene Set Variation Analysis for Microarray and RNA-Seq Data. BMC Bioinformatics, 2013, 14:7., DOI 10.1186/ 1471-2105-14-7 Tirosh I, Izar B, Prakadan SM, Wadsworth MH, Treacy D, Trombetta JJ, et al. Dissecting the Multicellular Ecosystem of Metastatic Melanoma by Single-Cell RNA-Seq. Science, 2016, 352:189–96., DOI 10.1126/science.aad0501 Aran D, Hu Z, Butte AJ. xCell: Digitally Portraying the Tissue Cellular Heterogeneity Landscape. Genome Biol, 2017, 18:220., DOI 10.1186/s13059- 017-1349-1 Davoli T, Uno H, Wooten EC, Elledge SJ. Tumor Aneuploidy Correlates with Markers of Immune Evasion and with Reduced Response to Immunotherapy. Science, 2017, 355:eaaf8399., DOI 10.1126/science.aaf8399 Liu Z, Li M, Jiang Z, Wang X. A Comprehensive Immunologic Portrait of Triple-Negative Breast Cancer. Transl Oncol, 2018, 11:311–29., DOI 10.1016/j. tranon.2018.01.011 van der Weyden L, Arends MJ, Campbell AD, Bald T, Wardle-Jones H, Griggs N, et al. Genome-wide In Vivo Screen Identifies Novel Host Regulators of Metastatic Colonization. Nature, 2017, 541:233–6., DOI 10.1038/nature20792 Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez JC, et al. pROC: An Open-Source Package for R and S+ to Analyze and Compare ROC Curves. BMC Bioinformatics, 2011, 12:77., DOI 10.1186/1471-2105-12-77 Yang Q, Wang R, Wei B, Peng C,Wang L, Hu G, et al. Candidate Biomarkers and Molecular Mechanism Investigation for Glioblastoma Multiforme Utilizing WGCNA. Biomed Res Int, 2018, 2018:e4246703., DOI 10.1155/ 2018/4246703 Liu Y. Survival Correlation of Immune Response in Human Cancers. Oncotarget, 2019, 10:6885–97., DOI 10.18632/oncotarget.27360 Uddin MN, Wang X. The Landscape of Long Non-coding RNAs in Tumor Stroma. Life Sci, 2021, 264:118725., DOI 10.1016/j.lfs.2020.118725 Mousavi Ardehaie R, Hashemzadeh S, Behrouz Sharif S, Ghojazadeh M, Teimoori-Toolabi L, Sakhinia E. Aberrant Methylated EDNRB Can Act as a Potential Diagnostic Biomarker in Sporadic Colorectal Cancer while KISS1 Is Controversial. Bioengineered, 2017, 8:555–64., DOI 10.1080/21655979.2017. 1283458 Gao D, Han Y, Yang Y, Herman JG, Linghu E, Zhan Q, et al. Methylation of TMEM176A Is an Independent Prognostic Marker and Is Involved in Human Colorectal Cancer Development. Epigenetics, 2017, 12:575–83., DOI 10.1080/ 15592294.2017.1341027 Xu Y, Xu J, Yang Y, Zhu L, Li X, Zhao W. SRGN Promotes Colorectal Cancer Metastasis as a Critical Downstream Target of HIF-1α. Cell Physiol Biochem, 2018, 48:2429–40., DOI 10.1159/000492657 Tang J, Jiang W, Liu D, Luo J, Wu X, Pan Z, et al. The Comprehensive Molecular Landscape of the Immunologic Co-stimulator B7 and TNFR Ligand Receptor Families in Colorectal Cancer: Immunotherapeutic Implications with Microsatellite Instability. OncoImmunology, 2018, 7:e1488566., DOI 10.1080/ 2162402X.2018.1488566 Jung B, Staudacher JJ, Beauchamp D. Transforming Growth Factor β Superfamily Signaling in Development of Colorectal Cancer. Gastroenterology, 2017, 152:36–52., DOI 10.1053/j.gastro.2016.10.015 Barrett CW, Reddy VK, Short SP, Motley AK, Lintel MK, Bradley AM, et al. Selenoprotein P Influences Colitis-Induced Tumorigenesis by Mediating Stemness and Oxidative Damage. J Clin Invest, 2015, 125:2646–60., DOI 10. 1172/JCI76099 Li D, Guo J,Wang S, Zhu L, Shen Z. Identification of Novel Methylated Targets in Colorectal Cancer by Microarray Analysis and Construction of Coexpression Network. Oncol Lett, 2017, 14:2643–8., DOI 10.3892/ol.2017.6506 Uddin MN, Li M, Wang X. Identification of Transcriptional Signatures of Colon Tumor Stroma by a Meta-Analysis. J Oncol, 2019, 2019:8752862., DOI 10.1155/2019/8752862 Uddin MN, Li M, Wang X. Identification of Transcriptional Markers and microRNA–mRNA Regulatory Networks in Colon Cancer by Integrative Analysis of mRNA and microRNA Expression Profiles in Colon Tumor Stroma. Cells, 2019, 8:E1054., DOI 10.3390/cells8091054 Bhagwat AS, Vakoc CR. Targeting Transcription Factors in Cancer. Trends Cancer, 2015, 1:53–65., DOI 10.1016/j.trecan.2015.07.001 Vishnoi K, Viswakarma N, Rana A, Rana B. Transcription Factors in Cancer Development and Therapy. Cancers, 2020, 12:E2296., DOI 10.3390/ cancers12082296 Li D, Wei P, Peng Z, Huang C, Tang H, Jia Z, et al. The Critical Role of Dysregulated FOXM1-PLAUR Signaling in Human colon Cancer Progression and Metastasis. Clin Cancer Res, 2013, 19:62–72., DOI 10.1158/1078-0432. CCR-12-1588 Joshi S, Singh AR, Zulcic M, Bao L, Messer K, Ideker T, et al. Rac2 Controls Tumor Growth, Metastasis and M1-M2 Macrophage Differentiation In Vivo. PLoS ONE, 2014, 9:e95893., DOI 10.1371/journal.pone.0095893 Xu Y, Pasche B. TGF-β Signaling Alterations and Susceptibility to Colorectal Cancer. Hum Mol Genet, 2007, 16:R14–20., DOI 10.1093/hmg/ddl486 Mazzarelli P, Pucci S, Spagnoli LG. CLU and colon Cancer. The Dual Face of CLU: from normal to Malignant Phenotype. Adv Cancer Res, 2009, 105:45–61., DOI 10.1016/S0065-230X(09)05003-9 O’Brien RM, Cannon A, Reynolds JV, Lysaght J, Lynam-Lennon N. Complement in Tumourigenesis and the Response to Cancer Therapy. Cancers, 2021, 13:1209., DOI 10.3390/cancers13061209 Itatani Y, Kawada K, Inamoto S, Yamamoto T, Ogawa R, Taketo MM, et al. The Role of Chemokines in Promoting Colorectal Cancer Invasion/Metastasis. Int J Mol Sci, 2016, 17:643., DOI 10.3390/ijms17050643 Ni S, Ren F, Xu M, Tan C, Weng W, Huang Z, et al. CTHRC1 Overexpression Predicts Poor Survival and Enhances Epithelial mesenchymal Transition in Colorectal Cancer. Cancer Med, 2018, 7:5643–54., DOI 10.1002/cam4.1807 Yu S, Wu Y, Li C, Qu Z, Lou G, Guo X, et al. Comprehensive Analysis of the SLC16A Gene Family in Pancreatic Cancer via Integrated Bioinformatics. Sci Rep, 2020, 10:7315., DOI 10.1038/s41598-020-64356-y Lu C, Rong D, Zhang B, Zheng W, Wang X, Chen Z, et al. Current Perspectives on the Immunosuppressive Tumor Microenvironment in Hepatocellular Carcinoma: Challenges and Opportunities. Mol Cancer, 2019, 18:130., DOI 10.1186/s12943-019-1047-6 Lorenzo-Sanz L, Munoz P. Tumor-Infiltrating Immunosuppressive Cells in Cancer-Cell Plasticity, Tumor Progression and Therapy Response. Cancer Microenviron, 2019, 12:119–32., DOI 10.1007/s12307-019-00232-2 Manzat Saplacan RM, Balacescu L, Gherman C, Chira RI, Craiu A, Mircea PA, et al. The Role of PDGFs and PDGFRs in Colorectal Cancer. Mediators Inflamm, 2017, 2017:e4708076., DOI 10.1155/2017/4708076 Hibi K, Mizukami H, Saito M, Kigawa G, Nemoto H, Sanada Y. FBN2 Methylation Is Detected in the Serum of Colorectal Cancer Patients with Hepatic Metastasis. Anticancer Res, 2012, 32:4371–4. Zhang H, Apfelroth SD, Hu W, Davis EC, Sanguineti C, Bonadio J, et al. Structure and Expression of Fibrillin-2, a Novel Microfibrillar Component Preferentially Located in Elastic Matrices. J Cel Biol, 1994, 124:855–63., DOI 10. 1083/jcb.124.5.855 Lian X, Bond JS, Bharathy N, Boudko SP, Pokidysheva E, Shern JF, et al. Defining the Extracellular Matrix of Rhabdomyosarcoma. Front Oncol, 2021, 11:601957., DOI 10.3389/fonc.2021.601957 Gu H-Y, Zhang C, Guo J, Yang M, Zhong HC, Jin W, et al. Risk Score Based on Expression of Five Novel Genes Predicts Survival in Soft Tissue Sarcoma. Aging, 2020, 12:3807–27., DOI 10.18632/aging.102847 Chu L, Qu Y, An Y, Hou L, Li J, LiW, et al. Induction of Senescence-Associated Secretory Phenotype Underlies the Therapeutic Efficacy of PRC2 Inhibition in Cancer. Cell Death Dis, 2022, 13:155., DOI 10.1038/s41419-022-04601-6 Yi JM, Dhir M, Guzzetta AA, Iacobuzio-Donahue CA, Heo K, Yang KM, et al. DNA Methylation Biomarker Candidates for Early Detection of colon Cancer. Tumour Biol, 2012, 33:363–72., DOI 10.1007/s13277-011-0302-2 Yi JM. DNA Methylation Change Profiling of Colorectal Disease: Screening towards Clinical Use. Life, 2021, 11:412., DOI 10.3390/life11050412 Yan M, Sun H, Li G, Chen Q, Xie Q, Zhang W, et al. miR-139-5p Was Identified as Biomarker of Different Molecular Subtypes of Breast Carcinoma. Front Oncol, 2022, 12:857714., DOI 10.3389/fonc.2022.857714 Song W, Bai Y, Zhu J, Zeng F, Yang C, Hu B, et al. A Novel Prognostic Model Based on Epithelial-Mesenchymal Transition-Related Genes Predicts Patient Survival in Gastric Cancer. World J Surg Oncol, 2021, 19:216., DOI 10.1186/ s12957-021-02329-9 Zhang S, Zeng X, Lin S, Liang M, Huang H. Identification of Seven-Gene Marker to Predict the Survival of Patients with Lung Adenocarcinoma Using Integrated Multi-Omics Data Analysis. J Clin Lab Anal, 2022, 36:e24190., DOI 10.1002/jcla.24190 Meng J, Zhang J, Xiu Y, Jin Y, Xiang J, Nie Y, et al. Prognostic Value of an Immunohistochemical Signature in Patients with Esophageal Squamous Cell Carcinoma Undergoing Radical Esophagectomy. Mol Oncol, 2018, 12: 196–207., DOI 10.1002/1878-0261.12158 Poschel A, Beebe E, Kunz L, Amini P, Guscetti F, Malbon A, et al. Identification of Disease-Promoting Stromal Components by Comparative Proteomic and Transcriptomic Profiling of Canine Mammary Tumors Using Laser-Capture Microdissected FFPE Tissue. Neoplasia, 2021, 23:400–12., DOI 10.1016/j.neo. 2021.03.001 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610350 EP 1610365 DI 10.3389/pore.2022.1610350 PG 16 ER PT J AU Han, D Cho, SE Park, J Kim, D AF Han, Dawool Cho, Sandra Eunae Park, Jiho Kim, Dongwook TI Case Report: Papillary Lesions at the Mouth Floor May Mimic Sialadenoma Papilliferum SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE salivary gland; tumor; sialadenoma papilliferum; oral pathology; papillary lesion; oral cavity; oromaxillofacial ID salivary gland; tumor; sialadenoma papilliferum; oral pathology; papillary lesion; oral cavity; oromaxillofacial AB Salivary gland tumor Sialadenoma papilliferum (SialP) clinically resembles papillary epithelial lesions, such as squamous papilloma (SqP) or verrucous leukoplakia. Pathological sampling including an adequate depth of both the mucosa and submucosa layer is required for discrimination between the diseases. Though ductal proliferation in the submucosa is characteristic in SialP, papillary lesions arising at the mouth floor, specifically near the ductal orifice, are more problematic. Salivary gland ductal ectasia, along with the overlying papillary hyperplasia, may mimic the biphasic tumorous growth pattern of SialP, making discrimination extremely difficult. Further cellular dysplasia in the papillary mucosal lesion raises the possibility of malignant transformation in a known benign lesion, SialP. Herein, we present a case of SqP at the mouth floor which mimicked both clinical and pathological features of SialP and compared it with a definite case of SialP. Moreover, we discuss major differential points that clinicians and pathologists should consider during diagnosis of oral papillary lesions arising near the salivary glands. C1 [Han, Dawool] Yonsei University College of Dentistry, Oral Cancer Research Institute, Department of Oral PathologySeoul, South Korea. [Cho, Sandra Eunae] Yonsei University College of Dentistry, Oral Cancer Research Institute, Department of Oral PathologySeoul, South Korea. [Park, Jiho] Yonsei University College of Dentistry, Department of Oral and Maxillofacial SurgerySeoul, South Korea. [Kim, Dongwook] Yonsei University College of Dentistry, Department of Oral and Maxillofacial SurgerySeoul, South Korea. RP Kim, D (reprint author), Yonsei University College of Dentistry, Department of Oral and Maxillofacial Surgery, Seoul, South Korea. EM dwkimomfs@yuhs.ac CR Abrams AM, Franck FM. Sialadenoma papilliferum.A Previously Unreported Salivary Gland Tumor. Cancer, 1969, 24:1057–63., DOI 10. 1002/1097-0142(196911)24:5<1057::aid-cncr2820240529>3.0.co;2-l Kronenberg J, Horowitz A, Leventon G. Sialadenoma Papilliferum of the Parotid Gland. J Laryngol Otol, 1989, 103:1089–90., DOI 10.1017/ s0022215100111089 Nakaguro M, Mino-Kenudson M, Urano M, Ogawa I, Honda Y, Hirai H, et al. Sialadenoma Papilliferum of the Bronchus: An Unrecognized Bronchial Counterpart of the Salivary Gland Tumor with Frequent: BRAF: V600E Mutations. Am J Surg Pathol, 2021, 45:662–71., DOI 10. 1097/pas.0000000000001657 Bobos M, Hytiroglou P, Karkavelas G, Papakonstantinou C, Papadimitriou CS. Sialadenoma Papilliferum of Bronchus. Virchows Archiv, 2003, 443:695–9., DOI 10.1007/s00428-003-0879-y Hamilton J, Osborne RF, Smith LM. Sialadenoma Papilliferum Involving the Nasopharynx. Ear Nose Throat J, 2005, 84:474–5., DOI 10.1177/ 014556130508400807 Miyamoto S, Ogawa T, Chikazu D. Sialadenoma Papilliferum in the BuccalMucosa Detected on 18 F-Fluorodeoxyglucose-Positron Emission Tomography. Br J Oral Maxillofacial Surg, 2017, 55:727–9., DOI 10.1016/ j.bjoms.2017.05.004 Chen S, Peng J, Yuan C, Sun L, Zhang R, Sun Y. Sialadenoma Papilliferum: Clinicopathologic, Immunohistochemical, Molecular Analyses of New Five Cases and Review of the Literature. Diagn Pathol, 2021, 16:22., DOI 10.1186/s13000-021-01084-5 Shirasuna K, Watatani K, Miyazaki T. Ultrastructure of a Sialadenoma Papilliferum. Cancer, 1984, 53:468–74., DOI 10.1002/1097- 0142(19840201)53:3<468::aid-cncr2820530317>3.0.co;2-r Fowler CB, Damm DD. Sialadenoma Papilliferum: Analysis of Seven New Cases and Review of the Literature. Head Neck Pathol, 2018, 12: 193–201., DOI 10.1007/s12105-017-0852-8 de Vasconcelos Ii AJ, Gerber LR, Saraiva SH, de Vasconcelos PO, Cabral LN, Pinheiro TN. Sialadenoma Papilliferum: Bibliometric Analysis. J Clin Exp Dent, 2019, 11:e839–e44., DOI 10.4317/jced.56055 MahajanD, Khurana N, SetiaN. Sialadenoma Papilliferum in a Young Patient: a Case Report and Review of the Literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endodontology., 2007, 103:e51–e54., DOI 10.1016/j.tripleo.2006.01.012 Ide F, Kikuchi K, Kusama K, Kanazawa H. Sialadenoma Papilliferum with Potentially Malignant Features. J Clin Pathol, 2010, 63:362–4., DOI 10.1136/jcp. 2009.072660 Solomon MP, Rosen Y, Alfonso A. Intraoral Papillary Squamous Cell Tumor of the Soft Palate with Features of Sialadenoma Papilliferum-? Malignant Sialadenoma Papilliferum. Cancer, 1978, 42:1859–69., DOI 10.1002/1097-0142(197810)42: 4<1859::aid-cncr2820420425>3.0.co;2-c Shimoda M, Kameyama K, Morinaga S, Tanaka Y, Hashiguchi K, ShimadaM, et al. Malignant Transformation of Sialadenoma Papilliferum of the Palate: a Case Report. Virchows Arch, 2004, 445:641–6., DOI 10.1007/s00428-004-1091-4 Ponniah I. A Rare Case of Sialadenoma Papilliferum with Epithelial Dysplasia and Carcinoma In Situ. Oral Surg Oral Med Oral Pathol Oral Radiol Endodontology, 2007, 104:e27–e29., DOI 10.1016/j.tripleo.2006.06.053 Liu W, GneppDR, de Vries E, BibawyH, Solomon M, Gloster ES.Mucoepidermoid Carcinoma Arising in a Background of Sialadenoma Papilliferum: A Case Report. Head Neck Pathol, 2009, 3:59–62., DOI 10.1007/s12105-009-0106-5 Carneiro TE, Marinho SA, Verli FD, Mesquita ATM, Lima NL, Miranda JL. Oral Squamous Papilloma: Clinical, Histologic and Immunohistochemical Analyses. J Oral Sci, 2009, 51:367–72., DOI 10.2334/josnusd.51.367 Pimentel MTY, Amado ML, Sarandeses AG. Recurrent Sialadenoma Papilliferum of the Buccal Mucosa. J Laryngol Otol, 2007, 109:787–90., DOI 10.1017/ s0022215100131330 Maiorano E, Favia G, Ricco R. Sialadenoma Papilliferum: an Immunohistochemical Study of Five Cases. J Oral Pathol Med, 1996, 25:336–42., DOI 10.1111/j.1600-0714. 1996.tb00273.x Gomes APN, Sobral APV, Loducca SVL, de Araujo VC. Sialadenoma Papilliferum: Immunohistochemical Study. Int J Oral Maxillofacial Surg, 2004, 33:621–4., DOI 10. 1016/j.ijom.2003.10.019 Ohtomo R, Mori T, Shibata S, Tsuta K, Maeshima AM, Akazawa C, et al. SOX10 Is a Novel Marker of Acinus and Intercalated Duct Differentiation in Salivary Gland Tumors: a Clue to the Histogenesis for Tumor Diagnosis. Mod Pathol, 2013, 26:1041–50., DOI 10.1038/modpathol. 2013.54 Hashmi AA, Hussain ZF, Hashmi SK, Irfan M, Khan EY, Faridi N, et al. Immunohistochemical over Expression of P53 in Head and Neck Squamous Cell Carcinoma: Clinical and Prognostic Significance. BMC Res Notes, 2018, 11:433., DOI 10.1186/s13104-018-3547-7 Hsieh L-L, Wang P-F, Chen I-H, Liao C-T, Wang H-M, Chen M-C, et al. Characteristics of Mutations in the P53 Gene in Oral Squamous Cell Carcinoma Associated with Betel Quid Chewing and Cigarette Smoking in Taiwanese. Carcinogenesis, 2001, 22:1497–503., DOI 10.1093/carcin/22.9. 1497 Sawada K, Momose S, Kawano R, Kohda M, Irie T, Mishima K, et al. Immunohistochemical Staining Patterns of P53 Predict the Mutational Status of TP53 in Oral Epithelial Dysplasia. Mod Pathol, 2022, 35:177–85., DOI 10.1038/s41379-021-00893-9 Gomes CC, Diniz MG, Orsine LA, Duarte AP, Fonseca-Silva T, Conn BI, et al. Assessment of TP53 Mutations in Benign and Malignant Salivary Gland Neoplasms. PLoS One, 2012, 7:e41261., DOI 10.1371/journal.pone.0041261 Lazzaro B, Cleveland D. P53 and Ki-67 Antigen Expression in Small Oral Biopsy Specimens of Salivary Gland Tumors. Oral Surg Oral Med Oral Pathol Oral Radiol Endodontology, 2000, 89:613–7., DOI 10.1067/moe.2000.105765 Luukkaa H, Klemi P, Leivo I, Vahlberg T, Grenman R. Prognostic Significance of Ki-67 and P53 as Tumor Markers in Salivary Gland Malignancies in Finland: An Evaluation of 212 Cases. Acta Oncologica, 2006, 45:669–75., DOI 10.1080/02841860500543208 Hsieh M-S, Bishop JA,Wang Y-P, Poh CF, Cheng Y-SL, Lee Y-H, et al. Salivary Sialadenoma Papilliferum Consists of Two Morphologically, Immunophenotypically, and Genetically Distinct Subtypes. Head Neck Pathol, 2020, 14:489–96., DOI 10.1007/s12105-019-01068-4 Nakaguro M, Urano M, Ogawa I, Hirai H, Yamamoto Y, Yamaguchi H, et al. Histopathological Evaluation of Minor Salivary Gland Papillary-Cystic Tumours: Focus onGeneticAlterations inSialadenoma Papilliferum and Intraductal Papillary Mucinous Neoplasm. Histopathology, 2020, 76:411–22., DOI 10.1111/his.13990 Levinsohn JL, Sugarman JL, Bilguvar K, McNiff JM, Choate KAThe Yale Center for Mendelian Genomics. Somatic V600E BRAF Mutation in Linear and Sporadic Syringocystadenoma Papilliferum. J Invest Dermatol, 2015, 135:2536–8., DOI 10. 1038/jid.2015.180 Konstantinova AM, Kyrpychova L, Nemcova J, Sedivcova M, Bisceglia M, Kutzner H, et al. Syringocystadenoma Papilliferum of the Anogenital Area and Buttocks: A Report of 16 Cases, Including Human Papillomavirus Analysis and HRAS and BRAF V600 Mutation Studies. The Am J Dermatopathology, 2019, 41:281–5., DOI 10.1097/dad.0000000000001285 Pimentel MTY, Amado ML, Sarandeses AG. Recurrent Sialadenoma Papilliferum of the Buccal Mucosa. J Laryngol Otol, 1995, 109:787–90., DOI 10.1017/s0022215100131330 Rennie JS, MacDonald DG, Critchlow HA. Sialadenoma Papilliferum. Int J Oral Surg, 1984, 13:452–4., DOI 10.1016/s0300-9785(84)80074-5 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610352 EP 1610359 DI 10.3389/pore.2022.1610352 PG 8 ER PT J AU Wang, H Miao, J Wen, Y Xia, X Chen, Y Huang, M Chen, Sh Zhao, Z Zhang, Y Chen, Ch Zhu, X AF Wang, Hao Miao, Ji Wen, Yazhou Xia, Xihua Chen, Yanan Huang, Mengli Chen, Shiqing Zhao, Zhengyi Zhang, Yuzi Chen, Chunzhu Zhu, Xinhua TI Molecular Landscape of ERBB2 Alterations in 14,956 Solid Tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE next-generation sequencing; TMB; ERBB2; solid tumors; anti-HER2 agents ID next-generation sequencing; TMB; ERBB2; solid tumors; anti-HER2 agents AB ERBB2 abnormalities frequently occur and serve as rationale therapeutic targets in cancer. In this study, clinical and next-generation sequencing data from 14,956 patients across more than 20 tumor types were collected. A total of 406 (2.7%) patients were identified with ERBB2 amplifications, and 303 (2.0%) patients with pathogenic somatic ERBB2 mutations. ERBB2 amplifications fell most frequently in breast (15.9%) and stomach (8.3%) cancers. Somatic ERBB2 SNVs/indels occurred most common in bladder/urinary tract (7.3%) and intestine (6.1%) cancers. The top mutated ERBB2 SNVs/indels were p.Y772_A775dup (25.5%) and p.S310F/Y (19.9%). Significantly higher rates of ERBB2 SNV/indels were found in women compared to men (2.8% vs. 1.5%, p < 0.0001). CDK12 was the most common co-amplification gene with ERBB2 in cancers with a high frequency of ERBB2 amplifications. Patients with ERBB2 amplifications or mutations had higher TMB compared with patients with non-ERBB2 alterations. The study provided the landscape of ERBB2 alterations across a variety of solid tumors that may benefit fromanti-HER2 agents. C1 [Wang, Hao] The Affiliated Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Department of General SurgeryNanjing, China. [Miao, Ji] The Affiliated Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Department of General SurgeryNanjing, China. [Wen, Yazhou] Nanjing Maternity and Child Health Care Hospital, Women’s Hospital of Nanjing Medical University, Department of AnesthesiologyNanjing, China. [Xia, Xihua] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Chen, Yanan] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Huang, Mengli] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Chen, Shiqing] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Zhao, Zhengyi] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Zhang, Yuzi] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Chen, Chunzhu] 3D Medicines Inc., The Medical DepartmentShanghai, China. [Zhu, Xinhua] The Affiliated Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Department of General SurgeryNanjing, China. RP Zhu, X (reprint author), The Affiliated Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Department of General Surgery, Nanjing, China. EM drzhuxh@163.com CR Yarden Y, Sliwkowski MX. Untangling the ErbB Signalling Network. Nat Rev Mol Cel Biol, 2001, 2(2):127–37., DOI 10.1038/35052073 Prenzel N, Fischer OM, Streit S, Hart S, Ullrich A. The Epidermal Growth Factor Receptor Family as a central Element for Cellular Signal Transduction and Diversification. Endocr Relat Cancer, 2001, 8(1):11–31., DOI 10.1677/erc.0. 0080011 Roskoski R. The ErbB/HER Family of Protein-Tyrosine Kinases and Cancer. Pharmacol Res, 2014, 79:34–74., DOI 10.1016/j.phrs.2013.11.002 Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human Breast Cancer: Correlation of Relapse and Survival with Amplification of the HER-2/Neu Oncogene. Science, 1987, 235(4785):177–82., DOI 10.1126/science. 3798106 Wang YK, Gao CF, Yun T, Chen Z, Zhang XW, Lv XX, et al. Assessment of ERBB2 and EGFR Gene Amplification and Protein Expression in Gastric Carcinoma by Immunohistochemistry and Fluorescence In Situ Hybridization. Mol Cytogenet, 2011, 4(1):14., DOI 10.1186/1755-8166-4-14 Wei XW, Gao X, Zhang XC, Yang JJ, Chen ZH, Wu YL, et al.Mutational Landscape and Characteristics of ERBB2 in Non-small Cell Lung Cancer. Thorac Cancer, 2020, 11(6):1512–21., DOI 10.1111/1759-7714.13419 Emde A, Kostler WJ, Yarden Y. Association of Radiotherapy and Oncology of the Mediterranean arEa. Therapeutic Strategies and Mechanisms of Tumorigenesis of HER2-Overexpressing Breast Cancer. Crit Rev Oncol Hematol, 2012, 84(Suppl. 1):e49–e57., DOI 10.1016/j.critrevonc.2010.09.002 Bang Y-J, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, et al. Trastuzumab in Combination with Chemotherapy versus Chemotherapy Alone for Treatment of HER2-Positive Advanced Gastric or Gastro- Oesophageal junction Cancer, ToGA): A Phase 3, Open-Label, Randomised Controlled Trial. Lancet, 2010, 376(9742):687–97., DOI 10. 1016/S0140-6736(10)61121-X Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang G, Rahmeh M, et al. Activity of the Dual Kinase Inhibitor Lapatinib, GW572016, against HER-2- Overexpressing and Trastuzumab-Treated Breast Cancer Cells. Cancer Res, 2006, 66(3):1630–9., DOI 10.1158/0008-5472.CAN-05-1182 O’Brien NA, Browne BC, Chow L, Wang Y, Ginther C, Arboleda J, et al. Activated Phosphoinositide 3-kinase/AKT Signaling Confers Resistance to Trastuzumab but Not Lapatinib. Mol Cancer Ther, 2010, 9(6):1489–502., DOI 10.1158/1535-7163.MCT-09-1171 Verma S,Miles D, Gianni L, Krop IE,Welslau M, Baselga J, et al. Trastuzumab Emtansine for HER2-Positive Advanced Breast Cancer. N Engl J Med, 2012, 367(19):1783–91., DOI 10.1056/NEJMoa1209124 Krop IE, Kim S-B, Gonzalez-Martin A, LoRusso PM, Ferrero J-M, Smitt M, et al. Trastuzumab Emtansine versus Treatment of Physician’s Choice for Pretreated HER2-Positive Advanced Breast Cancer, TH3RESA): A Randomised, Open-Label, Phase 3 Trial. Lancet Oncol, 2014, 15(7):689–99., DOI 10.1016/S1470-2045(14)70178-0 Perez EA, Barrios C, Eiermann W, Toi M, Im Y-H, Conte P, et al. Trastuzumab Emtansine with or without Pertuzumab versus Trastuzumab Plus Taxane for Human Epidermal Growth Factor Receptor 2-Positive, Advanced Breast Cancer: Primary Results from the Phase III MARIANNE Study. J Clin Oncol, 2017, 35(2):141–8., DOI 10. 1200/JCO.2016.67.4887 Modi S. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Metastatic Breast Cancer: Plain Language Summary of the DESTINYBreast01 Study. Future Oncol, 2021, 17(26):3415–23., DOI 10.2217/fon- 2021-0427 Shitara K, Bang Y-J, Iwasa S, Sugimoto N, Ryu M-H, Sakai D, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N Engl J Med, 2020, 382(25):2419–30., DOI 10.1056/ NEJMoa2004413 Elamin YY, Robichaux JP, Carter BW, Altan M, Gibbons DL, Fossella FV, et al. Poziotinib for Patients with HER2 Exon 20 Mutant Non-small-cell Lung Cancer: Results from a Phase II Trial. J Clin Oncol, 2022, 40:702–9., DOI 10.1200/JCO.21.01113 Li BT, Shen R, Buonocore D, Olah ZT, Ni A, Ginsberg MS, et al. Ado- Trastuzumab Emtansine for Patients with HER2-Mutant Lung Cancers: Results from a Phase II Basket Trial. J Clin Oncol, 2018, 36(24):2532–7., DOI 10.1200/JCO.2018.77.9777 Li BT, Smit EF, Goto Y, Nakagawa K, Udagawa H, Mazieres J, et al. Trastuzumab Deruxtecan in HER2-Mutant Non-small-cell Lung Cancer. N Engl J Med, 2022, 386:241–51., DOI 10.1056/NEJMoa2112431 Su D, Zhang D, Chen K, Lu J, Wu J, Cao X, et al. High Performance of Targeted Next Generation Sequencing on Variance Detection in Clinical Tumor Specimens in Comparison with Current Conventional Methods. J Exp Clin Cancer Res, 2017, 36(1):121., DOI 10.1186/s13046-017-0591-4 Xi R, Lee S, Xia Y, Kim T-M, Park PJ. Copy Number Analysis of Whole- Genome Data Using BIC-Seq2 and its Application to Detection of Cancer Susceptibility Variants. Nucleic Acids Res, 2016, 44(13):6274–86., DOI 10. 1093/nar/gkw491 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Sci Signal, 2013, 6(269):pl1., DOI 10.1126/scisignal. 2004088 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data. Cancer Discov, 2012, 2(5): 401–4., DOI 10.1158/2159-8290.CD-12-0095 Robertson AG, Kim J, Al-Ahmadie H, Bellmunt J, Guo G, Cherniack AD, et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell, 2018, 174(4):1033., DOI 10.1016/j.cell.2018.07.036 Robichaux JP, Elamin YY, Vijayan RSK, Nilsson MB, Hu L, He J, et al. Pan- Cancer Landscape and Analysis of ERBB2 Mutations Identifies Poziotinib as a Clinically Active Inhibitor and Enhancer of T-DM1 Activity. Cancer Cell, 2019, 36(4):444–57., DOI 10.1016/j.ccell.2019.09.001 Triulzi T, Forte L, Regondi V, Di Modica M, Ghirelli C, Carcangiu ML, et al. HER2 Signaling Regulates the Tumor Immune Microenvironment and Trastuzumab Efficacy. Oncoimmunology, 2019, 8(1):e1512942., DOI 10.1080/ 2162402X.2018.1512942 Janjigian YY, Maron SB, ChatilaWK, Millang B, Chavan SS, Alterman C, et al. Firstline Pembrolizumab and Trastuzumab in HER2-Positive Oesophageal, Gastric, or Gastro-Oesophageal junction Cancer: An Open-Label, Single-Arm, Phase 2 Trial. Lancet Oncol, 2020, 21(6):821–31., DOI 10.1016/S1470-2045(20)30169-8 Robichaux JP, Elamin YY, Tan Z, Carter BW, Zhang S, Liu S, et al.Mechanisms and Clinical Activity of an EGFR and HER2 Exon 20-selective Kinase Inhibitor in Nonsmall Cell Lung Cancer. Nat Med, 2018, 24(5):638–46., DOI 10.1038/s41591-018- 0007-9 Kirchner M, Kluck K, Brandt R, Volckmar A-L, Penzel R, Kazdal D, et al. The Immune Microenvironment in EGFR- and ERBB2-Mutated Lung Adenocarcinoma. ESMO Open, 2021, 6(5):100253., DOI 10.1016/j.esmoop.2021. 100253 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610360 EP 1610367 DI 10.3389/pore.2022.1610360 PG 8 ER PT J AU Rubovszky, G Kocsis, J Boer, K Chilingirova, N Dank, M Kahan, Zs Kaidarova, D Kover, E Vertakova Krakovska, B Mahr, K Mrinakova, B Piko, B Bozovic-Spasojevic, I Horvath, Zs AF Rubovszky, Gabor Kocsis, Judit Boer, Katalin Chilingirova, Nataliya Dank, Magdolna Kahan, Zsuzsanna Kaidarova, Dilyara Kover, Erika Vertakova Krakovska, Bibiana Mahr, Karoly Mrinakova, Bela Piko, Bela Bozovic-Spasojevic, Ivana Horvath, Zsolt TI Systemic Treatment of Breast Cancer. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Guideline DE early breast cancer; locally advanced breast cancer; adjuvant treatment; neoadjuvant treatment; metastatic breast cancer; inflammatory breast cancer; guideline ID early breast cancer; locally advanced breast cancer; adjuvant treatment; neoadjuvant treatment; metastatic breast cancer; inflammatory breast cancer; guideline AB This text is based on the recommendations accepted by the 4th Hungarian Consensus Conference on Breast Cancer, modified based on the international consultation and conference within the frames of the Central-Eastern European Academy of Oncology. The professional guideline primarily reflects the resolutions and recommendations of the current ESMO, NCCN and ABC5, as well as that of the St. Gallen Consensus Conference statements. The recommendations cover classical prognostic factors and certain multigene tests, which play an important role in therapeutic decision-making. From a didactic point of view, the text first addresses early and then locally advanced breast cancer, followed by locoregionally recurrent and metastatic breast cancer. Within these, we discuss each group according to the available therapeutic options. At the end of the recommendations, we summarize the criteria for treatment in certain rare clinical situations. C1 [Rubovszky, Gabor] Orszagos Onkologiai Intezet, „B” Belgyogyaszati-Onkologiai es Klinikai Farmakologiai OsztalyBudapest, Hungary. [Kocsis, Judit] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. [Boer, Katalin] St. Margit Hospital, Clinical Oncology DepartmentBudapest, Hungary. [Chilingirova, Nataliya] Medical University-Pleven, Heart and Brain Hospital, Science and Research Institute, Clinic Center of ExcellencePleven, Bulgaria. [Dank, Magdolna] Semmelweis University, Department of OncologyBudapest, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of OncotherapySzeged, Hungary. [Kaidarova, Dilyara] Kazakh Institute of Oncology and RadiologyAlmaty, Kazakhstan. [Kover, Erika] University of Pecs, Oncotherapy InstitutePecs, Hungary. [Vertakova Krakovska, Bibiana] Comenius University, Faculty of Medicine, 1st Department of OncologyBratislava, Slovakia. [Mahr, Karoly] Zala Megyei Szent Rafael Korhaz, OnkologiaZalaegerszeg, Hungary. [Mrinakova, Bela] Comenius University, Faculty of Medicine, 1st Department of OncologyBratislava, Slovakia. [Piko, Bela] Bekes Megyei Kozponti Korhaz, Pandy Kalman Tagkorhaz, Megyei Onkologiai CentrumGyula, Hungary. [Bozovic-Spasojevic, Ivana] Daily Chemotherapy Hospital, Institute for Oncology and Radiology of SerbiaBelgrade, Serbia. [Horvath, Zsolt] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. RP Rubovszky, G (reprint author), Orszagos Onkologiai Intezet, „B” Belgyogyaszati-Onkologiai es Klinikai Farmakologiai Osztaly, Budapest, Hungary. EM rubovszkyg@gmail.com CR Horvath Z, Boer K, Dank M, Kahan Z, Kocsis J, Kover E, et al. Systemic Therapy of Breast Cancer: Practice Guideline. Magy Onkol, 2016, 60(3): 241–57. Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, et al. Early Breast Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol, 2019, 30(8):1194–220., DOI 10.1093/ annonc/mdz173 NCCN. NCCN Clinical Practice Guidelines in Oncology, NCCN Guidelines(R)): Breast Cancer: NCCN.Org; 2020. [1/2020:. Available from: http://www.nccn.org/professionals/physician_gls/pdf/breast.pdf, Accessed April 2022). Cardoso F, Paluch-Shimon S, Senkus E, Curigliano G, Aapro MS, Andre F, et al. 5th ESO-ESMO International Consensus Guidelines for Advanced Breast Cancer, ABC 5). Ann Oncol, 2020, 31(12):1623–49., DOI 10.1016/j. annonc.2020.09.010 Burstein HJ, Curigliano G, Loibl S, Dubsky P, Gnant M, Poortmans P, et al. Estimating the Benefits of Therapy for Early-Stage Breast Cancer: the St. Gallen International Consensus Guidelines for the Primary Therapy of Early Breast Cancer 2019. Ann Oncol, 2019, 30(10):1541–57., DOI 10.1093/annonc/ mdz235 Wilson ARM, Marotti L, Bianchi S, Biganzoli L, Claassen S, Decker T, et al. The Requirements of a Specialist Breast Centre. Eur J Cancer, 2013, 49(17): 3579–87., DOI 10.1016/j.ejca.2013.07.017 Pons-Tostivint E, Daubisse-Marliac L, Grosclaude P, Oum Sack E, Goddard J, Morel C, et al. Multidisciplinary Team Meeting and EUSOMA Quality Indicators in Breast Cancer Care: A French Regional Multicenter Study. The Breast, 2019, 46:170–7., DOI 10.1016/j.breast.2019.06.001 Hammond MEH, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for Immunohistochemical Testing of Estrogen and Progesterone Receptors in Breast Cancer. Jco, 2010, 28(16):2784–95., DOI 10.1200/jco.2009.25.6529 Yi M, Huo L, Koenig KB, Mittendorf EA,Meric-Bernstam F, Kuerer HM, et al. Which Threshold for ER Positivity? a Retrospective Study Based on 9639 Patients. Ann Oncol, 2014, 25(5):1004–11., DOI 10.1093/annonc/mdu053 Giannakeas V. Single Hormone Receptor-Positive Breast Cancer-Signal or Noise? JAMA Netw Open, 2020, 3(1):e1918176., DOI 10.1001/ jamanetworkopen.2019.18176 Giuliano AE, Edge SB, Hortobagyi GN. Eighth Edition of the AJCC Cancer Staging Manual: Breast Cancer. Ann Surg Oncol, 2018, 25(7):1783–5., DOI 10. 1245/s10434-018-6486-6 Margolese RG, Cecchini RS, Julian TB, Ganz PA, Costantino JP, Vallow LA, et al. Anastrozole versus Tamoxifen in Postmenopausal Women with Ductal Carcinoma In Situ Undergoing Lumpectomy Plus Radiotherapy, NSABP B- 35): a Randomised, Double-Blind, Phase 3 Clinical Trial. The Lancet, 2016, 387(10021):849–56., DOI 10.1016/s0140-6736(15)01168-x Narod SA, Iqbal J, Giannakeas V, Sopik V, Sun P. Breast Cancer Mortality after a Diagnosis of Ductal Carcinoma In Situ. JAMA Oncol, 2015, 1(7): 888–96., DOI 10.1001/jamaoncol.2015.2510 Phung MT, Tin Tin S, Elwood JM. Prognostic Models for Breast Cancer: a Systematic Review. BMC cancer, 2019, 19(1):230., DOI 10.1186/s12885-019- 5442-6 Gray E, Donten A, Payne K, Hall PS. Survival Estimates Stratified by the Nottingham Prognostic Index for Early Breast Cancer: a Systematic Review and Meta-Analysis of Observational Studies. Syst Rev, 2018, 7(1):142., DOI 10. 1186/s13643-018-0803-9 Hearne BJ, Teare MD, Butt M, Donaldson L. Comparison of Nottingham Prognostic Index and Adjuvant Online Prognostic Tools in Young Women with Breast Cancer: Review of a Single-Institution Experience. BMJ open, 2015, 5(1):e005576., DOI 10.1136/bmjopen-2014-005576 Lee AHS, Ellis IO. The Nottingham Prognostic index for Invasive Carcinoma of the Breast. Pathol Oncol Res, 2008, 14(2):113–5., DOI 10.1007/s12253-008- 9067-3 Harnan S, Tappenden P, Cooper K, Stevens J, Bessey A, Rafia R, et al. Tumour Profiling Tests to Guide Adjuvant Chemotherapy Decisions in Early Breast Cancer: a Systematic Review and Economic Analysis. Health Technol Assess, 2019, 23(30):1–328., DOI 10.3310/hta23300 Candido Dos Reis FJ, Wishart GC, Dicks EM, Greenberg D, Rashbass J, Schmidt MK, et al. An Updated PREDICT Breast Cancer Prognostication and Treatment Benefit Prediction Model with Independent Validation. Breast Cancer Res, 2017, 19(1):58., DOI 10.1186/s13058-017-0852-3 Wishart GC, Bajdik CD, Dicks E, Provenzano E, Schmidt MK, Sherman M, et al. PREDICT Plus: Development and Validation of a Prognostic Model for Early Breast Cancer that Includes HER2. Br J Cancer, 2012, 107(5):800–7., DOI 10.1038/bjc.2012.338 Wishart GC, Azzato EM, Greenberg DC, Rashbass J, Kearins O, Lawrence G, et al. PREDICT: a New UK Prognostic Model that Predicts Survival Following Surgery for Invasive Breast Cancer. Breast Cancer Res, 2010, 12(1):R1., DOI 10. 1186/bcr2464 Sparano JA, Gray RJ, Makower DF, Pritchard KI, Albain KS, Hayes DF, et al. Adjuvant Chemotherapy Guided by a 21-Gene Expression Assay in Breast Cancer. N Engl J Med, 2018, 379(2):111–21., DOI 10.1056/nejmoa1804710 Paik S, Tang G, Shak S, Kim C, Baker J, Kim W, et al. Gene Expression and Benefit of Chemotherapy in Women with Node-Negative, Estrogen Receptor-Positive Breast Cancer. Jco, 2006, 24(23):3726–34., DOI 10.1200/ jco.2005.04.7985 Massarweh SA, Sledge GW, Miller DP, McCullough D, Petkov VI, Shak S. Molecular Characterization and Mortality from Breast Cancer in Men. Jco, 2018, 36(14):1396–404., DOI 10.1200/jco.2017.76.8861 Sparano J, O’Neill A, Alpaugh K, Wolff AC, Northfelt DW, Dang CT, et al. Association of Circulating Tumor Cells with Late Recurrence of Estrogen Receptor-Positive Breast Cancer. JAMA Oncol, 2018, 4(12):1700–6., DOI 10. 1001/jamaoncol.2018.2574 Hibler EA, Kauderer J, Greene MH, Rodriguez GC, Alberts DS. Bone Loss after Oophorectomy Among High-Risk Women: an NRG Oncology/ gynecologic Oncology Group Study. Menopause, 2016, 23(11):1228–32., DOI 10.1097/gme.0000000000000692 Francis PA, Pagani O, Fleming GF, Walley BA, Colleoni M, Lang I, et al. Tailoring Adjuvant Endocrine Therapy for Premenopausal Breast Cancer. N Engl J Med, 2018, 379(2):122–37., DOI 10.1056/nejmoa1803164 Gibson L, Lawrence D, Dawson C, Bliss J. Aromatase Inhibitors for Treatment of Advanced Breast Cancer in Postmenopausal Women. Cochrane Database Syst Rev, 2009, 2009(4):CD003370., DOI 10.1002/14651858.CD003370.pub3 Regan MM, Francis PA, Pagani O, Fleming GF, Walley BA, Viale G, et al. Absolute Benefit of Adjuvant Endocrine Therapies for Premenopausal Women with Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Early Breast Cancer: TEXT and SOFT Trials. Jco, 2016, 34(19):2221–31., DOI 10.1200/jco.2015.64.3171 Pagani O, Francis PA, Fleming GF, Walley BA, Viale G, Colleoni M, et al. Absolute Improvements in Freedom from Distant Recurrence to Tailor Adjuvant Endocrine Therapies for Premenopausal Women: Results from TEXT and SOFT. Jco, 2020, 38(12):1293–303., DOI 10.1200/jco.18.01967 Regan MM, Walley BA, Francis PA, Fleming GF, Lang I, Gomez HL, et al. Concurrent and Sequential Initiation of Ovarian Function Suppression with Chemotherapy in Premenopausal Women with Endocrine-Responsive Early Breast Cancer: an Exploratory Analysis of TEXT and SOFT. Ann Oncol, 2017, 28(9):2225–32., DOI 10.1093/annonc/mdx285 van Hellemond IEG, Geurts SME, Tjan-Heijnen VCG. Current Status of Extended Adjuvant Endocrine Therapy in Early Stage Breast Cancer. Curr Treat Options Oncol, 2018, 19(5):26., DOI 10.1007/s11864-018-0541-1 Ribnikar D, Sousa B, Cufer T, Cardoso F. Extended Adjuvant Endocrine Therapy - A Standard to All or Some? The Breast, 2017, 32:112–8., DOI 10. 1016/j.breast.2017.01.004 Clement Z, Kollias J, Bingham J, Whitfield R, Bochner M. Extended Duration of Adjuvant Aromatase Inhibitor in Breast Cancer: a Meta-Analysis of Randomized Controlled Trials. Gland Surg, 2018, 7(5):449–57., DOI 10. 21037/gs.2018.08.03 Burstein HJ, Lacchetti C, Anderson H, Buchholz TA, Davidson NE, Gelmon KA, et al. Adjuvant Endocrine Therapy for Women with Hormone Receptor- Positive Breast Cancer: ASCO Clinical Practice Guideline Focused Update. Jco, 2019, 37(5):423–38., DOI 10.1200/jco.18.01160 Gray RTE. Effects of Prolonging Adjuvant Aromatase Inhibitor Therapy beyond Five Years on Recurrence and Cause-specific Mortality: an EBCTCG Meta-Analysis of Individual Patient Data from 12 Randomised Trials Including 24,912 Women. Cancer Res, 2019, 79., DOI 10.1158/1538-7445. SABCS18-GS3-03 Kalinsky K, Barlow WE, Gralow h, Meric-Bernstam F, Albain KS, Hayes DF, et al. 21-Gene Assay to Inform Chemotherapy Benefit in Node-Positive Breast Cancer. N Engl J Med, 2021, 385(25):2336–47., DOI 10.1056/ nejmoa2108873 LeVasseur N, Sun J, Fenton D, Baxter S, Chan A, Roberts S, et al. Impact of the 21-Gene Recurrence Score Assay on the Treatment of Estrogen Receptor- Positive, HER2-Negative, Breast Cancer Patients with 1-3 Positive Nodes: A Prospective Clinical Utility Study. Clin Breast Cancer, 2021, 22:e74–e79., DOI 10.1016/j.clbc.2021.09.004 Jones S, Holmes FA, O’Shaughnessy J, Blum JL, Vukelja SJ, McIntyre KJ, et al. Docetaxel with Cyclophosphamide Is Associated with an Overall Survival Benefit Compared with Doxorubicin and Cyclophosphamide: 7-Year Follow- Up of US Oncology Research Trial 9735. Jco, 2009, 27(8):1177–83., DOI 10. 1200/jco.2008.18.4028 Fisher B, Jeong J-H, Dignam J, Anderson S, Mamounas E, Wickerham DL, et al. Findings from Recent National Surgical Adjuvant Breast and Bowel Project Adjuvant Studies in Stage I Breast Cancer. JNCI Monogr, 2001, 2001(30):62–6., DOI 10.1093/oxfordjournals.jncimonographs.a003463 Early Breast Cancer Trialists’ Collaborative G. Effects of Chemotherapy and Hormonal Therapy for Early Breast Cancer on Recurrence and 15-year Survival: an Overview of the Randomised Trials. The Lancet, 2005, 365(9472):1687–717., DOI 10.1016/s0140-6736(05)66544-0 Bines J, Earl H, Buzaid AC, Saad ED. Anthracyclines and Taxanes in the Neo/ adjuvant Treatment of Breast Cancer: Does the Sequence Matter? Ann Oncol, 2014, 25(6):1079–85., DOI 10.1093/annonc/mdu007 Citron ML, Berry DA, Cirrincione C, Hudis C, Winer EP, GradisharWJ, et al. Randomized Trial of Dose-Dense versus Conventionally Scheduled and Sequential versus Concurrent Combination Chemotherapy as Postoperative Adjuvant Treatment of Node-Positive Primary Breast Cancer: First Report of Intergroup Trial C9741/Cancer and Leukemia Group B Trial 9741. Jco, 2003, 21(8):1431–9., DOI 10.1200/jco.2003.09.081 Sparano JA, Wang M, Martino S, Jones V, Perez EA, Saphner T, et al. Weekly Paclitaxel in the Adjuvant Treatment of Breast Cancer. N Engl J Med, 2008, 358(16):1663–71., DOI 10.1056/nejmoa0707056 Mackey JR, Martin M, Pienkowski T, Rolski J, Guastalla J-P, Sami A, et al. Adjuvant Docetaxel, Doxorubicin, and Cyclophosphamide in Node-Positive Breast Cancer: 10-year Follow-Up of the Phase 3 Randomised BCIRG 001 Trial. Lancet Oncol, 2013, 14(1):72–80., DOI 10.1016/s1470-2045(12)70525-9 Swain SM, Tang G, Geyer CE, Rastogi P, Atkins JN, Donnellan PP, et al. Definitive Results of a Phase III Adjuvant Trial Comparing Three Chemotherapy Regimens in Women with Operable, Node-Positive Breast Cancer: the NSABP B-38 Trial. Jco, 2013, 31(26):3197–204., DOI 10.1200/jco. 2012.48.1275 Chen H, Xiao L, Li J, Cui L, Huang W. Adjuvant Gonadotropin-Releasing Hormone Analogues for the Prevention of Chemotherapy-Induced Premature Ovarian Failure in Premenopausal Women. Cochrane Database Syst Rev, 2019, 3:CD008018., DOI 10.1002/14651858.CD008018.pub3 Recht A, Come SE, Henderson IC, Gelman RS, Silver B, Hayes DF, et al. The Sequencing of Chemotherapy and Radiation Therapy after Conservative Surgery for Early-Stage Breast Cancer. N Engl J Med, 1996, 334(21): 1356–61., DOI 10.1056/nejm199605233342102 Hickey BE, Francis DP, Lehman M. Sequencing of Chemotherapy and Radiotherapy for Early Breast Cancer. Cochrane Database Syst Rev, 2013, 2013(4):CD005212., DOI 10.1002/14651858.CD005212.pub3 Masuda N, Lee S-J, Ohtani S, Im Y-H, Lee E-S, Yokota I, et al. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med, 2017, 376(22):2147–59., DOI 10.1056/nejmoa1612645 Schneeweiss A, Chia S, Hickish T, Harvey V, Eniu A, Hegg R, et al. Pertuzumab Plus Trastuzumab in Combination with Standard Neoadjuvant Anthracycline-Containing and Anthracycline-free Chemotherapy Regimens in Patients with HER2-Positive Early Breast Cancer: a Randomized Phase II Cardiac Safety Study, TRYPHAENA). Ann Oncol, 2013, 24(9):2278–84., DOI 10.1093/annonc/mdt182 Schneeweiss A, Chia S, Hickish T, Harvey V, Eniu A, Waldron-Lynch M, et al. Long-term Efficacy Analysis of the Randomised, Phase II TRYPHAENA Cardiac Safety Study: Evaluating Pertuzumab and Trastuzumab Plus Standard Neoadjuvant Anthracycline-Containing and Anthracycline-free Chemotherapy Regimens in Patients with HER2-Positive Early Breast Cancer. Eur J Cancer, 2018, 89:27–35., DOI 10.1016/j.ejca.2017.10.021 Gianni L, Pienkowski T, Im Y-H, Roman L, Tseng L-M, Liu M-C, et al. Efficacy and Safety of Neoadjuvant Pertuzumab and Trastuzumab in Women with Locally Advanced, Inflammatory, or Early HER2-Positive Breast Cancer, NeoSphere): a Randomised Multicentre, Open-Label, Phase 2 Trial. Lancet Oncol, 2012, 13(1):25–32., DOI 10.1016/s1470- 2045(11)70336-9 Gianni L, Pienkowski T, Im Y-H, Tseng L-M, Liu M-C, Lluch A, et al. 5-year Analysis of Neoadjuvant Pertuzumab and Trastuzumab in Patients with Locally Advanced, Inflammatory, or Early-Stage HER2-Positive Breast Cancer, NeoSphere): a Multicentre, Open-Label, Phase 2 Randomised Trial. Lancet Oncol, 2016, 17(6):791–800., DOI 10.1016/s1470-2045(16, 00163-7 Harbeck N, Gluz O, Christgen M, Kuemmel S, Grischke E-M, Braun M, et al. De-escalated Neoadjuvant Pertuzumab+trastuzumab with or without Paclitaxel Weekly in HR-/HER2+ Early Breast Cancer: ADAPT-HR-/ HER2+ Biomarker and Survival Results. Jco, 2021, 39(15_Suppl. l):503., DOI 10.1200/jco.2021.39.15_suppl.503 O’Sullivan CC, Ballman KV, McCall L, Kommalapati A, Zemla T, Weiss A, et al. Alliance A011801, compassHER2 RD): Postneoadjuvant T-DM1 + Tucatinib/placebo in Patients with Residual HER2-Positive Invasive Breast Cancer. Future Oncol, 2021, 17(34):4665–76., DOI 10.2217/fon-2021-0753 von Minckwitz G, Procter M, de Azambuja E, Zardavas D, Benyunes M, Viale G, et al. Adjuvant Pertuzumab and Trastuzumab in Early HER2-Positive Breast Cancer. N Engl J Med, 2017, 377(2):122–31., DOI 10.1056/ nejmoa1703643 von Minckwitz G, Huang C-S, Mano MS, Loibl S, Mamounas EP, Untch M, et al. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer. N Engl J Med, 2019, 380(7):617–28., DOI 10.1056/nejmoa1814017 Baselga J, Bradbury I, Eidtmann H, Di Cosimo S, de Azambuja E, Aura C, et al. Lapatinib with Trastuzumab for HER2-Positive Early Breast Cancer, NeoALTTO): a Randomised, Open-Label, Multicentre, Phase 3 Trial. The Lancet, 2012, 379(9816):633–40., DOI 10.1016/s0140-6736(11)61847-3 Tolaney SM, Guo H, Pernas S, Barry WT, Dillon DA, Ritterhouse L, et al. Seven-Year Follow-Up Analysis of Adjuvant Paclitaxel and Trastuzumab Trial for Node-Negative, Human Epidermal Growth Factor Receptor 2- Positive Breast Cancer. Jco, 2019, 37(22):1868–75., DOI 10.1200/jco.19.00066 De Cock E, Pivot X, Hauser N, Verma S, Kritikou P, Millar D, et al. A Time and Motion Study of Subcutaneous versus Intravenous Trastuzumab in Patients with HER2-Positive Early Breast Cancer. Cancer Med, 2016, 5(3):389–97., DOI 10.1002/cam4.573 Tjalma WAA, Van den Mooter T, Mertens T, Bastiaens V, Huizing MT, Papadimitriou K. Subcutaneous Trastuzumab, Herceptin, versus Intravenous Trastuzumab for the Treatment of Patients with HER2- Positive Breast Cancer: A Time, Motion and Cost Assessment Study in a Lean Operating Day Care Oncology Unit. Eur J Obstet Gynecol Reprod Biol, 2018, 221:46–51., DOI 10.1016/j.ejogrb.2017.12.006 Ismael G, Hegg R, Muehlbauer S, Heinzmann D, Lum B, Kim S-B, et al. Subcutaneous versus Intravenous Administration of, Neo)adjuvant Trastuzumab in Patients with HER2-Positive, Clinical Stage I-III Breast Cancer, HannaH Study): a Phase 3, Open-Label, Multicentre, Randomised Trial. Lancet Oncol, 2012, 13(9):869–78., DOI 10.1016/s1470-2045(12)70329-7 Tadros AB, Wen HY, Morrow M. Breast Cancers of Special Histologic Subtypes Are Biologically Diverse. Ann Surg Oncol, 2018, 25(11):3158–64., DOI 10.1245/s10434-018-6687-z Martin M, Holmes FA, Ejlertsen B, Delaloge S, Moy B, Iwata H, et al. Neratinib after Trastuzumab-Based Adjuvant Therapy in HER2-Positive Breast Cancer, ExteNET): 5-year Analysis of a Randomised, Double- Blind, Placebo-Controlled, Phase 3 Trial. Lancet Oncol, 2017, 18(12): 1688–700., DOI 10.1016/S1470-2045(17)30717-9 Robson M, Im S-A, Senkus E, Xu B, Domchek SM, Masuda N, et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N Engl J Med, 2017, 377(6):523–33., DOI 10.1056/nejmoa1706450 Ellis MJ, Tao Y, Luo J, A’Hern R, Evans DB, Bhatnagar AS, et al. Outcome Prediction for Estrogen Receptor-Positive Breast Cancer Based on Postneoadjuvant Endocrine Therapy Tumor Characteristics. JNCI J Natl Cancer Inst, 2008, 100(19):1380–8., DOI 10.1093/jnci/djn309 Dawood S, Merajver SD, Viens P, Vermeulen PB, Swain SM, Buchholz TA, et al. International Expert Panel on Inflammatory Breast Cancer: Consensus Statement for Standardized Diagnosis and Treatment. Ann Oncol, 2011, 22(3):515–23., DOI 10.1093/annonc/mdq345 Ueno NT, Espinosa Fernandez JR, Cristofanilli M, Overmoyer B, Rea D, Berdichevski F, et al. International Consensus on the Clinical Management of Inflammatory Breast Cancer from the Morgan Welch Inflammatory Breast Cancer Research Program 10th Anniversary Conference. J Cancer, 2018, 9(8):1437–47., DOI 10.7150/jca.23969 Aebi S, Gelber S, Anderson SJ, Lang I, Robidoux A, Martin M, et al. Chemotherapy for Isolated Locoregional Recurrence of Breast Cancer, CALOR): a Randomised Trial. Lancet Oncol, 2014, 15(2):156–63., DOI 10. 1016/s1470-2045(13)70589-8 Johnston SRD, Harbeck N, Hegg R, Toi M, Martin M, Shao ZM, et al. Abemaciclib Combined with Endocrine Therapy for the Adjuvant Treatment of HR+, HER2−, Node-Positive, High-Risk, Early Breast Cancer, monarchE). Jco, 2020, 38(34):3987–98., DOI 10.1200/jco.20.02514 Harbeck N, Rastogi P, Martin M, Tolaney SM, Shao ZM, Fasching PA, et al. Adjuvant Abemaciclib Combined with Endocrine Therapy for High-Risk Early Breast Cancer: Updated Efficacy and Ki-67 Analysis from the monarchE Study. Ann Oncol : official J Eur Soc Med Oncol, 2021, 32(12): 1571–81. Loibl S, Schneeweiss A, Huober JB, Braun M, Rey J, Blohmer JU, et al. Durvalumab Improves Long-Term Outcome in TNBC: Results from the Phase II Randomized GeparNUEVO Study Investigating Neodjuvant Durvalumab in Addition to an Anthracycline/taxane Based Neoadjuvant Chemotherapy in Early Triple-Negative Breast Cancer, TNBC). Jco, 2021, 39(15_Suppl. l):506., DOI 10.1200/jco.2021.39.15_suppl.506 Schmid Jc P, Dent R, Pusztai L, McArthur HL, Kuemmel S, Bergh J, et al. KEYNOTE-522: Phase 3 Study of Pembrolizumab, Pembro, + Chemotherapy, Chemo, vs Placebo, Pbo, + Chemo as Neoadjuvant Treatment, Followed by Pembro vs Pbo as Adjuvant Treatment for Early Triple-Negative Breast Cancer, TNBC). Ann Oncol, 2019, 30:v851–v934., DOI 10.1093/annonc/mdz394.003 Tripathy D, Im S-A, Colleoni M, Franke F, Bardia A, Harbeck N, et al. Ribociclib Plus Endocrine Therapy for Premenopausal Women with Hormone-Receptor-Positive, Advanced Breast Cancer, MONALEESA-7): a Randomised Phase 3 Trial. Lancet Oncol, 2018, 19(7):904–15., DOI 10. 1016/s1470-2045(18)30292-4 Hortobagyi Sms GN, Burris HA, III, Yap YS, Sonke GS, Hart L, Campone M, et al. Overall Survival, OS, Results from the Phase III MONALEESA-2, ML- 2, Trial of Postmenopausal Patients, Pts, with Hormone Receptor Positive/ human Epidermal Growth Factor Receptor 2 Negative, HR+/HER2−, Advanced Breast Cancer, ABC, Treated with Endocrine Therapy, ET, ± Ribociclib, RIB). Ann Oncol, 2021, 32:1283–S346., DOI 10.1016/j.annonc. 2021.08.2090 Finn RS, Martin M, Rugo HS, Jones S, Im S-A, Gelmon K, et al. Palbociclib and Letrozole in Advanced Breast Cancer. N Engl J Med, 2016, 375(20): 1925–36., DOI 10.1056/nejmoa1607303 Hortobagyi GN, Stemmer SM, Burris HA, Yap Y-S, Sonke GS, Paluch- Shimon S, et al. Ribociclib as First-Line Therapy for HR-Positive, Advanced Breast Cancer. N Engl J Med, 2016, 375(18):1738–48., DOI 10.1056/ nejmoa1609709 Hortobagyi GN, Stemmer SM, Burris HA, Yap YS, Sonke GS, Paluch-Shimon S, et al. Updated Results from MONALEESA-2, a Phase III Trial of First-Line Ribociclib Plus Letrozole versus Placebo Plus Letrozole in Hormone Receptor-Positive, HER2-Negative Advanced Breast Cancer. Ann Oncol, 2019, 30(11):1842., DOI 10.1093/annonc/mdz215 Goetz MP, Toi M, Campone M, Sohn J, Paluch-Shimon S, Huober J, et al. MONARCH 3: Abemaciclib as Initial Therapy for Advanced Breast Cancer. Jco, 2017, 35(32):3638–46., DOI 10.1200/jco.2017.75.6155 Turner NC, Slamon DJ, Ro J, Bondarenko I, Im S-A, Masuda N, et al. Overall Survival with Palbociclib and Fulvestrant in Advanced Breast Cancer. N Engl J Med, 2018, 379(20):1926–36., DOI 10.1056/nejmoa1810527 Cristofanilli M, Turner NC, Bondarenko I, Ro J, Im S-A, Masuda N, et al. Fulvestrant Plus Palbociclib versus Fulvestrant Plus Placebo for Treatment of Hormone-Receptor-Positive, HER2-Negative Metastatic Breast Cancer that Progressed on Previous Endocrine Therapy, PALOMA-3): Final Analysis of the Multicentre, Double-Blind, Phase 3 Randomised Controlled Trial. Lancet Oncol, 2016, 17(4):425–39., DOI 10.1016/s1470- 2045(15)00613-0 Slamon DJ, Neven P, Chia S, Jerusalem G, De Laurentiis M, Im S, et al. Ribociclib Plus Fulvestrant for Postmenopausal Women with Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-negative Advanced Breast Cancer in the Phase III Randomized MONALEESA-3 Trial: Updated Overall Survival. Ann Oncol, 2021, 32(8):1015–24., DOI 10.1016/j. annonc.2021.05.353 Slamon DJ, Neven P, Chia S, Fasching PA, De Laurentiis M, Im S-A, et al. Overall Survival with Ribociclib Plus Fulvestrant in Advanced Breast Cancer. N Engl J Med, 2020, 382(6):514–24., DOI 10.1056/nejmoa1911149 Slamon DJ, Neven P, Chia S, Fasching PA, De Laurentiis M, Im S-A, et al. Phase III Randomized Study of Ribociclib and Fulvestrant in Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Advanced Breast Cancer: MONALEESA-3. Jco, 2018, 36(24):2465–72., DOI 10.1200/jco.2018.78.9909 Sledge GW, ToiM, Neven P, Sohn J, Inoue K, Pivot X, et al.MONARCH 2: Abemaciclib in Combination with Fulvestrant in Women with HR+/ HER2− Advanced Breast Cancer Who Had Progressed while Receiving Endocrine Therapy. Jco, 2017, 35(25):2875–84., DOI 10.1200/jco.2017.73. 7585 Sledge GW, Toi M, Neven P, Sohn J, Inoue K, Pivot X, et al. The Effect of Abemaciclib Plus Fulvestrant on Overall Survival in Hormone Receptor- Positive, ERBB2-Negative Breast Cancer that Progressed on Endocrine Therapy-MONARCH 2. JAMA Oncol, 2020, 6(1):116–24., DOI 10.1001/ jamaoncol.2019.4782 O’Shaughnessy J, Petrakova K, Sonke GS, Conte P, Arteaga CL, Cameron DA, et al. Ribociclib Plus Letrozole versus Letrozole Alone in Patients with De Novo HR+, HER2- Advanced Breast Cancer in the Randomized MONALEESA-2 Trial. Breast Cancer Res Treat, 2018, 168(1):127–34., DOI 10.1007/s10549-017-4518-8 Kaufman PA, Toi M, Neven P, Sohn J, Grischke E-M, Andre V, et al. Health- Related Quality of Life in MONARCH 2: Abemaciclib Plus Fulvestrant in Hormone Receptor-Positive, HER2-Negative Advanced Breast Cancer after Endocrine Therapy. The oncologist, 2020, 25(2):e243–e251., DOI 10.1634/ theoncologist.2019-0551 Turner NC, Ro J, Andre F, Loi S, Verma S, Iwata H, et al. Palbociclib in Hormone-Receptor-Positive Advanced Breast Cancer. N Engl J Med, 2015, 373(3):209–19., DOI 10.1056/nejmoa1505270 Baselga J, Campone M, Piccart M, Burris HA, 3rd, Rugo HS, Sahmoud T, et al. Everolimus in Postmenopausal Hormone-Receptor-Positive Advanced Breast Cancer. N Engl J Med, 2012, 366(6):520–9., DOI 10.1056/ nejmoa1109653 Im S-A, Lu Y-S, Bardia A, Harbeck N, Colleoni M, Franke F, et al. Overall Survival with Ribociclib Plus Endocrine Therapy in Breast Cancer. N Engl J Med, 2019, 381(4):307–16., DOI 10.1056/nejmoa1903765 Sledge GW, Toi M, Neven P, Sohn J, Inoue K, Pivot X, et al. The Effect of Abemaciclib Plus Fulvestrant on Overall Survival in Hormone Receptor- Positive, ERBB2-Negative Breast Cancer that Progressed on Endocrine Therapy-MONARCH 2: A Randomized Clinical Trial. JAMA Oncol, 2020, 6:116–24., DOI 10.1001/jamaoncol.2019.4782 Spring LM, Wander SA, Zangardi M, Bardia A. CDK 4/6 Inhibitors in Breast Cancer: Current Controversies and Future Directions. Curr Oncol Rep, 2019, 21(3):25., DOI 10.1007/s11912-019-0769-3 Piccart M, Hortobagyi GN, Campone M, Pritchard KI, Lebrun F, Ito Y, et al. Everolimus Plus Exemestane for Hormone-Receptor-Positive, Human Epidermal Growth Factor Receptor-2-Negative Advanced Breast Cancer: Overall Survival Results from BOLERO-2. Ann Oncol, 2014, 25(12):2357–62., DOI 10.1093/annonc/mdu456 Bachelot T, Bourgier C, Cropet C, Ray-Coquard I, Ferrero J-M, Freyer G, et al. Randomized Phase II Trial of Everolimus in Combination with Tamoxifen in Patients with Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-negative Metastatic Breast Cancer with Prior Exposure to Aromatase Inhibitors: a GINECO Study. Jco, 2012, 30(22):2718–24., DOI 10.1200/jco.2011.39.0708 Massarweh S, Romond E, Black EP, Van Meter E, Shelton B, Kadamyan- Melkumian V, et al. A Phase II Study of Combined Fulvestrant and Everolimus in Patients with Metastatic Estrogen Receptor, ER)-positive Breast Cancer after Aromatase Inhibitor, AI, Failure. Breast Cancer Res Treat, 2014, 143(2):325–32., DOI 10.1007/s10549-013-2810-9 Mehta RS, Barlow WE, Albain KS, Vandenberg TA, Dakhil SR, Tirumali NR, et al. Combination Anastrozole and Fulvestrant in Metastatic Breast Cancer. N Engl J Med, 2012, 367(5):435–44., DOI 10.1056/nejmoa1201622 Dickler MN, Tolaney SM, Rugo HS, Cortes J, Dieras V, Patt D, et al. MONARCH 1, A Phase II Study of Abemaciclib, a CDK4 and CDK6 Inhibitor, as a Single Agent, in Patients with Refractory HR+/HER2− Metastatic Breast Cancer. Clin Cancer Res, 2017, 23(17):5218–24., DOI 10. 1158/1078-0432.ccr-17-0754 Andre F, Ciruelos E, Rubovszky G, Campone M, Loibl S, Rugo HS, et al. Alpelisib for PIK3CA-Mutated, Hormone Receptor-Positive Advanced Breast Cancer. N Engl J Med, 2019, 380(20):1929–40., DOI 10.1056/ nejmoa1813904 Kaufman B, Mackey JR, Clemens MR, Bapsy PP, Vaid A, Wardley A, et al. Trastuzumab Plus Anastrozole versus Anastrozole Alone for the Treatment of Postmenopausal Women with Human Epidermal Growth Factor Receptor 2-positive, Hormone Receptor-Positive Metastatic Breast Cancer: Results from the Randomized Phase III TAnDEM Study. Jco, 2009, 27(33):5529–37., DOI 10.1200/jco.2008.20.6847 Johnston S, Pippen J, Pivot X, Lichinitser M, Sadeghi S, Dieras V, et al. Lapatinib Combined with Letrozole versus Letrozole and Placebo as First-Line Therapy for Postmenopausal Hormone Receptor-Positive Metastatic Breast Cancer. Jco, 2009, 27(33):5538–46., DOI 10.1200/jco. 2009.23.3734 Schwartzberg LS, Franco SX, Florance A, O’Rourke L, Maltzman J, Johnston S. Lapatinib Plus Letrozole as First-Line Therapy for HER-2+ Hormone Receptor-Positive Metastatic Breast Cancer. The oncologist, 2010, 15(2): 122–9., DOI 10.1634/theoncologist.2009-0240 Liu Y, Gu F, Liang J, Dai X, Wan C, Hong X, et al. The Efficacy and Toxicity Profile of Metronomic Chemotherapy for Metastatic Breast Cancer: A Meta-Analysis. PloS one, 2017, 12(3):e0173693., DOI 10. 1371/journal.pone.0173693 Cazzaniga ME, Cortesi L, Cortesi L, Ferzi A, Scaltriti L, Cicchiello F, et al. Metronomic Chemotherapy with Oral Vinorelbine, mVNR, and Capecitabine, mCAPE, in Advanced HER2-Negative Breast Cancer Patients: Is it a Way to Optimize Disease Control? Final Results of the VICTOR-2 Study. Breast Cancer Res Treat, 2016, 160(3):501–9., DOI 10.1007/ s10549-016-4009-3 Fedele P, Marino A, Orlando L, Schiavone P, Nacci A, Sponziello F, et al. Efficacy and Safety of Low-Dose Metronomic Chemotherapy with Capecitabine in Heavily Pretreated Patients with Metastatic Breast Cancer. Eur J Cancer, 2012, 48(1):24–9., DOI 10.1016/j.ejca.2011.06.040 Li Q, Yan H, Zhao P, Yang Y, Cao B. Efficacy and Safety of Bevacizumab Combined with Chemotherapy for Managing Metastatic Breast Cancer: A Meta-Analysis of Randomized Controlled Trials. Sci Rep, 2015, 5:15746., DOI 10.1038/srep15746 Tutt A, Tovey H, Cheang MCU, Kernaghan S, Kilburn L, Gazinska P, et al. Carboplatin in BRCA1/2-Mutated and Triple-Negative Breast Cancer BRCAness Subgroups: the TNT Trial. Nat Med, 2018, 24(5):628–37., DOI 10.1038/s41591-018-0009-7 Smyth LM, Iyengar NM, Chen MF, Popper SM, Patil S, Wasserheit-Lieblich C, et al. Weekly Paclitaxel with Trastuzumab and Pertuzumab in Patients with HER2-Overexpressing Metastatic Breast Cancer: Overall Survival and Updated Progression-free Survival Results from a Phase II Study. Breast Cancer Res Treat, 2016, 158(1):91–7., DOI 10.1007/s10549-016-3851-7 Swain SM, Baselga J, Kim S-B, Ro J, Semiglazov V, Campone M, et al. Pertuzumab, Trastuzumab, and Docetaxel in HER2-Positive Metastatic Breast Cancer. N Engl J Med, 2015, 372(8):724–34., DOI 10.1056/ nejmoa1413513 Burstein HJ, Keshaviah A, Baron AD, Hart RD, Lambert-Falls R, Marcom PK, et al. Trastuzumab Plus Vinorelbine or Taxane Chemotherapy for HER2-Overexpressing Metastatic Breast Cancer: the Trastuzumab and Vinorelbine or Taxane Study. Cancer, 2007, 110(5):965–72., DOI 10.1002/ cncr.22885 Robert N, Leyland-Jones B, Asmar L, Belt R, Ilegbodu D, Loesch D, et al. Randomized Phase III Study of Trastuzumab, Paclitaxel, and Carboplatin Compared with Trastuzumab and Paclitaxel in Women with HER-2- Overexpressing Metastatic Breast Cancer. Jco, 2006, 24(18):2786–92., DOI 10.1200/jco.2005.04.1764 Bartsch R, Wenzel C, Altorjai G, Pluschnig U, Rudas M, Mader RM, et al. Capecitabine and Trastuzumab in Heavily Pretreated Metastatic Breast Cancer. Jco, 2007, 25(25):3853–8., DOI 10.1200/jco.2007.11.9776 Schaller G, Fuchs I, Gonsch T, Weber J, Kleine-Tebbe A, Klare P, et al. Phase II Study of Capecitabine Plus Trastuzumab in Human Epidermal Growth Factor Receptor 2-Overexpressing Metastatic Breast Cancer Pretreated with Anthracyclines or Taxanes. Jco, 2007, 25(22):3246–50., DOI 10.1200/jco.2006. 09.6826 Huober J, Fasching PA, Barsoum M, Petruzelka L, Wallwiener D, Thomssen C, et al. Higher Efficacy of Letrozole in Combination with Trastuzumab Compared to Letrozole Monotherapy as First-Line Treatment in Patients with HER2-Positive, Hormone-Receptor-Positive Metastatic Breast Cancer - Results of the eLEcTRA Trial. The Breast, 2012, 21(1):27–33., DOI 10.1016/j. breast.2011.07.006 Rimawi M, Ferrero J-M, de la Haba-Rodriguez J, Poole C, De Placido S, Osborne CK, et al. First-Line Trastuzumab Plus an Aromatase Inhibitor, with or without Pertuzumab, in Human Epidermal Growth Factor Receptor 2- Positive and Hormone Receptor-Positive Metastatic or Locally Advanced Breast Cancer, PERTAIN): A Randomized, Open-Label Phase II Trial. Jco, 2018, 36(28):2826–35., DOI 10.1200/jco.2017.76.7863 Dieras V, Miles D, Verma S, Pegram M, Welslau M, Baselga J, et al. Trastuzumab Emtansine versus Capecitabine Plus Lapatinib in Patients with Previously Treated HER2-Positive Advanced Breast Cancer, EMILIA): a Descriptive Analysis of Final Overall Survival Results from a Randomised, Open-Label, Phase 3 Trial. Lancet Oncol, 2017, 18(6):732–42., DOI 10.1016/s1470-2045(17)30312-1 Johnston SRD, Hegg R, Im S-A, Park IH, Burdaeva O, Kurteva G, et al. Phase III, Randomized Study of Dual Human Epidermal Growth Factor Receptor 2, HER2, Blockade with Lapatinib Plus Trastuzumab in Combination with an Aromatase Inhibitor in Postmenopausal Women with HER2-Positive, Hormone Receptor-Positive Metastatic Breast Cancer: ALTERNATIVE. Jco, 2018, 36(8):741–8., DOI 10.1200/jco.2017.74.7824 Cameron D, Casey M, Oliva C, Newstat B, Imwalle B, Geyer CE. Lapatinib Plus Capecitabine in Women with HER-2-Positive Advanced Breast Cancer: Final Survival Analysis of a Phase III Randomized Trial. The oncologist, 2010, 15(9):924–34., DOI 10.1634/theoncologist.2009-0181 Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, et al. Lapatinib Plus Capecitabine for HER2-Positive Advanced Breast Cancer. N Engl J Med, 2006, 355(26):2733–43., DOI 10.1056/nejmoa064320 Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, et al. Trastuzumab Emtansine for HER2-Positive Advanced Breast Cancer. N Engl J Med, 2012, 367(19):1783–91., DOI 10.1056/nejmoa1209124 Cortes JSK, Chung W, Im S, Park YH, Hegg R, Kim MH, et al. Trastuzumab Deruxtecan, T-DXd, vs Trastuzumab Emtansine, T-DM1, in Patients, Pts, with HER2+ Metastatic Breast Cancer, mBC): Results of the Randomized Phase III DESTINY-Breast03 Study. Ann Oncol, 2021, 32:S1283–S346., DOI 10.1016/j.annonc.2021.08.2087 Baez-Vallecillo L, Raghavendra AS, Hess KR, Barcenas CH, Moulder SL, Tripathy D, et al. Lapatinib Activity in Metastatic Human Epidermal Growth Factor Receptor 2-positive Breast Cancers that Received Prior Therapy with Trastuzumab, Pertuzumab, And/or Ado-Trastuzumab Emtansine, T-DM1). Breast Cancer Res Treat, 2019, 176(1):227–34., DOI 10.1007/s10549-018- 05081-z Blackwell KL, Burstein HJ, Storniolo AM, Rugo HS, Sledge G, Aktan G, et al. Overall Survival Benefit with Lapatinib in Combination with Trastuzumab for Patients with Human Epidermal Growth Factor Receptor 2-positive Metastatic Breast Cancer: Final Results from the EGF104900 Study. Jco, 2012, 30(21):2585–92., DOI 10.1200/jco.2011.35.6725 Krop IE, Kim S-B, Gonzalez-Martin A, LoRusso PM, Ferrero J-M, Smitt M, et al. Trastuzumab Emtansine versus Treatment of Physician’s Choice for Pretreated HER2-Positive Advanced Breast Cancer, TH3RESA): a Randomised, Open-Label, Phase 3 Trial. Lancet Oncol, 2014, 15(7): 689–99., DOI 10.1016/s1470-2045(14)70178-0 Krop IE, Kim S-B,Martin AG, LoRusso PM, Ferrero J-M, Badovinac-Crnjevic T, et al. Trastuzumab Emtansine versus Treatment of Physician’s Choice in Patients with Previously Treated HER2-Positive Metastatic Breast Cancer, TH3RESA): Final Overall Survival Results from a Randomised Open-Label Phase 3 Trial. Lancet Oncol, 2017, 18(6):743–54., DOI 10.1016/s1470-2045(17, 30313-3 Blackwell KL, Burstein HJ, Storniolo AM, Rugo H, Sledge G, Koehler M, et al. Randomized Study of Lapatinib Alone or in Combination with Trastuzumab in Women with ErbB2-Positive, Trastuzumab-Refractory Metastatic Breast Cancer. Jco, 2010, 28(7):1124–30., DOI 10.1200/jco. 2008.21.4437 Curigliano G, Mueller V, Borges V, Hamilton E, Hurvitz S, Loi S, et al. Tucatinib vs Placebo Added to Trastuzumab and Capecitabine for Patients with Pretreated HER2+ Metastatic Breast Cancer with and without Brain Metastases, HER2CLIMB): Final Overall Survival Analysis. Ann Oncol : official J Eur Soc Med Oncol, 2022, 33:321–9., DOI 10.1016/j.annonc.2021. 12.005 Murthy RK, Loi S, Okines A, Paplomata E, Hamilton E, Hurvitz SA, et al. Tucatinib, Trastuzumab, and Capecitabine for HER2-Positive Metastatic Breast Cancer. N Engl J Med, 2020, 382(7):597–609., DOI 10.1056/ nejmoa1914609 Modi S, Saura C, Yamashita T, Park YH, Kim S-B, Tamura K, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Breast Cancer. N Engl J Med, 2020, 382(7):610–21., DOI 10.1056/ nejmoa1914510 Saura C, Oliveira M, Feng Y-H, Dai M-S, Chen S-W, Hurvitz SA, et al. Neratinib Plus Capecitabine versus Lapatinib Plus Capecitabine in HER2- Positive Metastatic Breast Cancer Previously Treated with ≥ 2 HER2- Directed Regimens: Phase III NALA Trial. Jco, 2020, 38(27):3138–49., DOI 10.1200/jco.20.00147 Tarantino P, Morganti S, Uliano J, Giugliano F, Crimini E, Curigliano G. Margetuximab for the Treatment of HER2-Positive Metastatic Breast Cancer. Expert Opin Biol Ther, 2021, 21(2):127–33., DOI 10.1080/14712598.2021. 1856812 Winer EP, Lipatov O, Im S-A, Goncalves A, Munoz-Couselo E, Lee KS, et al. Pembrolizumab versus Investigator-Choice Chemotherapy for Metastatic Triple-Negative Breast Cancer, KEYNOTE-119): a Randomised, Open- Label, Phase 3 Trial. Lancet Oncol, 2021, 22(4):499–511., DOI 10.1016/ s1470-2045(20)30754-3 Schmid P, Rugo HS, Adams S, Schneeweiss A, Barrios CH, Iwata H, et al. Atezolizumab Plus Nab-Paclitaxel as First-Line Treatment for Unresectable, Locally Advanced or Metastatic Triple-Negative Breast Cancer, IMpassion130): Updated Efficacy Results from a Randomised, Double- Blind, Placebo-Controlled, Phase 3 Trial. Lancet Oncol, 2020, 21(1): 44–59., DOI 10.1016/s1470-2045(19)30689-8 Emens LA, Adams S, Barrios CH, Dieras V, Iwata H, Loi S, et al. First-line Atezolizumab Plus Nab-Paclitaxel for Unresectable, Locally Advanced, or Metastatic Triple-Negative Breast Cancer: IMpassion130 Final Overall Survival Analysis. Ann Oncol, 2021, 32(8):983–93., DOI 10.1016/j.annonc. 2021.05.355 Miles D, Gligorov J, Andre F, Cameron D, Schneeweiss A, Barrios C, et al. Primary Results from IMpassion131, a Double-Blind, Placebo-Controlled, Randomised Phase III Trial of First-Line Paclitaxel with or without Atezolizumab for Unresectable Locally Advanced/metastatic Triple- Negative Breast Cancer. Ann Oncol, 2021, 32(8):994–1004., DOI 10.1016/j. annonc.2021.05.801 Rugo Jc HS, Cescon DW, Im S, Md Yusof M, Gallardo C, Lipatov O, et al. KEYNOTE-355: Final Results from a Randomized, Double-Blind Phase III Study of First-Line Pembrolizumab + Chemotherapy vs Placebo + Chemotherapy for Metastatic TNBC. Ann Oncol, 2021, 32:S1283–S346. Coleman R, Hadji P, Body J-J, Santini D, Chow E, Terpos E, et al. Bone Health in Cancer: ESMO Clinical Practice Guidelines. Ann Oncol, 2020, 31(12): 1650–63., DOI 10.1016/j.annonc.2020.07.019 Early Breast Cancer Trialists’ Collaborative G. Adjuvant Bisphosphonate Treatment in Early Breast Cancer: Meta-Analyses of Individual Patient Data from Randomised Trials. The Lancet, 2015, 386(10001):1353–61., DOI 10. 1016/s0140-6736(15)60908-4 Hadji P, Aapro MS, Body J-J, Gnant M, Brandi ML, Reginster JY, et al. Management of Aromatase Inhibitor-Associated Bone Loss, AIBL, in Postmenopausal Women with Hormone Sensitive Breast Cancer: Joint Position Statement of the IOF, CABS, ECTS, IEG, ESCEO, IMS, and SIOG. J bone Oncol, 2017, 7:1–12., DOI 10.1016/j.jbo.2017.03.001 Coleman R, Finkelstein DM, Barrios C, Martin M, Iwata H, Hegg R, et al. Adjuvant Denosumab in Early Breast Cancer, D-CARE): an International, Multicentre, Randomised, Controlled, Phase 3 Trial. Lancet Oncol, 2020, 21(1):60–72., DOI 10.1016/s1470-2045(19)30687-4 Brufsky A, Mathew A. Bisphosphonate Choice as Adjuvant Therapy for Breast Cancer: Does it Matter? J Natl Cancer Inst, 2020, 112(7):659–60., DOI 10.1093/jnci/djz216 Byrski T, Huzarski T, Dent R, Marczyk E, Jasiowka M, Gronwald J, et al. Pathologic Complete Response to Neoadjuvant Cisplatin in BRCA1-Positive Breast Cancer Patients. Breast Cancer Res Treat, 2014, 147(2):401–5., DOI 10. 1007/s10549-014-3100-x Zimmer AS, Gillard M, Lipkowitz S, Lee J-M. Update on PARP Inhibitors in Breast Cancer. Curr Treat Options Oncol, 2018, 19(5):21., DOI 10.1007/ s11864-018-0540-2 Tutt ANJ, Garber JE, Kaufman B, Viale G, Fumagalli D, Rastogi P, et al. Adjuvant Olaparib for Patients with BRCA1- or BRCA2-Mutated Breast Cancer. N Engl J Med, 2021, 384(25):2394–405., DOI 10.1056/ nejmoa2105215 Sonnenblick A, de Azambuja E, Azim HA, Piccart M. An Update on PARP Inhibitors-Moving to the Adjuvant Setting. Nat Rev Clin Oncol, 2015, 12(1): 27–41., DOI 10.1038/nrclinonc.2014.163 Matulonis UA, Penson RT, Domchek SM, Kaufman B, Shapira-Frommer R, Audeh MW, et al. Olaparib Monotherapy in Patients with Advanced Relapsed Ovarian Cancer and a Germline BRCA1/2 Mutation: a Multistudy Analysis of Response Rates and Safety. Ann Oncol, 2016, 27(6):1013–9., DOI 10.1093/annonc/mdw133 Gelmon KA, Tischkowitz M, Mackay H, Swenerton K, Robidoux A, Tonkin K, et al. Olaparib in Patients with Recurrent High-Grade Serous or Poorly Differentiated Ovarian Carcinoma or Triple-Negative Breast Cancer: a Phase 2, Multicentre, Open-Label, Non-randomised Study. Lancet Oncol, 2011, 12(9):852–61., DOI 10.1016/s1470-2045(11)70214-5 Deb S, Lakhani SR, Ottini L, Fox SB. The Cancer Genetics and Pathology of Male Breast Cancer. Histopathology, 2016, 68(1):110–8., DOI 10.1111/his. 12862 Johansson I, Ringner M, Hedenfalk I. The Landscape of Candidate Driver Genes Differs between Male and Female Breast Cancer. PloS one, 2013, 8(10): e78299., DOI 10.1371/journal.pone.0078299 Ottini L, Palli D, Rizzo S, Federico M, Bazan V, Russo A. Male Breast Cancer. Crit Rev oncology/hematology, 2010, 73(2):141–55., DOI 10.1016/j.critrevonc. 2009.04.003 Eggemann H, Ignatov A, Smith BJ, Altmann U, von Minckwitz G, Rohl FW, et al. Adjuvant Therapy with Tamoxifen Compared to Aromatase Inhibitors for 257 Male Breast Cancer Patients. Breast Cancer Res Treat, 2013, 137(2): 465–70., DOI 10.1007/s10549-012-2355-3 Korde LA, Zujewski JA, Kamin L, Giordano S, Domchek S, Anderson WF, et al. Multidisciplinary Meeting on Male Breast Cancer: Summary and Research Recommendations. Jco, 2010, 28(12):2114–22., DOI 10.1200/jco. 2009.25.5729 Sousa B, Moser E, Cardoso F. An Update on Male Breast Cancer and Future Directions for Research and Treatment. Eur J Pharmacol, 2013, 717(1-3): 71–83., DOI 10.1016/j.ejphar.2013.03.037 Cardoso F, Bartlett JMS, Slaets L, van Deurzen CHM, van Leeuwen-Stok E, Porter P, et al. Characterization of Male Breast Cancer: Results of the EORTC 10085/TBCRC/BIG/NABCG International Male Breast Cancer Program. Ann Oncol, 2018, 29(2):405–17., DOI 10.1093/annonc/mdx651 Gucalp A, Traina TA, Eisner JR, Parker JS, Selitsky SR, Park BH, et al. Male Breast Cancer: a Disease Distinct from Female Breast Cancer. Breast Cancer Res Treat, 2019, 173(1):37–48., DOI 10.1007/s10549-018- 4921-9 Davies C, Pan H, Godwin J, Gray R, Arriagada R, Raina V, et al. Long-term Effects of Continuing Adjuvant Tamoxifen to 10 Years versus Stopping at 5 Years after Diagnosis of Oestrogen Receptor-Positive Breast Cancer: ATLAS, a Randomised Trial. The Lancet, 2013, 381(9869):805–16., DOI 10. 1016/s0140-6736(12)61963-1 Rodriguez AO, Chew H, Cress R, Xing G, McElvy S, Danielsen B, et al. Evidence of Poorer Survival in Pregnancy-Associated Breast Cancer. Obstet Gynecol, 2008, 112(1):71–8., DOI 10.1097/aog.0b013e31817c4ebc Zubor P, Kubatka P, Kapustova I, Miloseva L, Dankova Z, Gondova A, et al. Current Approaches in the Clinical Management of Pregnancy-Associated Breast Cancer-Pros and Cons. EPMA J, 2018, 9(3):257–70., DOI 10.1007/s13167-018- 0139-5 Bandoli G, Palmsten K, Forbess Smith CJ, Chambers CD. A Review of Systemic Corticosteroid Use in Pregnancy and the Risk of Select Pregnancy and Birth Outcomes. Rheum Dis Clin North America, 2017, 43(3):489–502., DOI 10.1016/j.rdc.2017.04.013 Yu HH, Cheung PS, Leung RC, Leung TN, Kwan WH. Current Management of Pregnancy-Associated Breast Cancer. Hong Kong Med J, 2017, 23(4): 387–94., DOI 10.12809/hkmj166049 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, et al. Personalizing the Treatment of Women with Early Breast Cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol, 2013, 24(9): 2206–23., DOI 10.1093/annonc/mdt303 Goldhirsch A, Wood WC, Gelber RD, Coates AS, Thurlimann B, Senn H-J, et al. Progress and Promise: Highlights of the International Expert Consensus on the Primary Therapy of Early Breast Cancer 2007. Ann Oncol, 2007, 18(7): 1133–44., DOI 10.1093/annonc/mdm271 Harlow SD, Gass M, Hall JE, Lobo R, Maki P, Rebar RW, et al. Executive Summary of the Stages of Reproductive Aging Workshop + 10: Addressing the Unfinished Agenda of Staging Reproductive Aging. J Clin Endocrinol Metab, 2012, 97(4):1159–68., DOI 10.1210/jc.2011-3362 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610383 EP 1610408 DI 10.3389/pore.2022.1610383 PG 26 ER PT J AU Liang, L Chai, Y Chai, F Liu, H Ma, N Zhang, H Zhang, Sh Nong, L Li, T Zhang, B AF Liang, Li Chai, Yijie Chai, Fei Liu, Haijing Ma, Ningning Zhang, Hong Zhang, Shuang Nong, Lin Li, Ting Zhang, Bo TI Expression of SASP, DNA Damage Response, and Cell Proliferation Factors in Early Gastric Neoplastic Lesions: Correlations and Clinical Significance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; gastric cancer; p53; DNA damage response; endoscopic submucosal dissection; senescence-associated secretory phenotype ID immunohistochemistry; gastric cancer; p53; DNA damage response; endoscopic submucosal dissection; senescence-associated secretory phenotype AB The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-mediated senescence-associated secretory phenotype (SASP) pathway has recently been identified in the suppression and promotion of cancers. However, its practical role in carcinogenesis remains to be comprehensively elucidated. Here, we describe an investigation analysing SASP activity and its correlations with DNA damage response (DDR), genomic mutations, and cell proliferation in gastric carcinogenesis among 30 cases with available endoscopic submucosal dissection (ESD) specimens of early neoplastic lesions (including low-grade dysplasia [LGD], high-grade dysplasia [HGD], and intramucosal carcinoma). The positive cells of senescence-associated β- galactosidase staining and cGAS, STING, interferon-regulatory factor 3 (IRF3), and signal transducer and activator of transcription 6 (STAT6) expression levels using immunostaining were elevated in HGD and in cancers. Similarly, increased expression of the Fanconi anemia group D2 (FANCD2) protein, tumour suppressor p53 binding protein 1 (TP53BP1), and replication protein A (RPA2) (i.e., primary DDR factors) was detected in HGD and in cancers; these increased expression levels were closely correlated with high expression of Ki67 and minichromosome maintenance complex component 7 (MCM7) proteins. Moreover, genomic mutations in TP53 gene were detected in 56.67% of the evaluated cases (17/30) using next-generation sequencing, and positive staining was verified in HGD and in cancers. Statistical analysis revealed that cell proliferation closely correlated with the expression of DDR factors, of which TP53BP1 was positively associated with SASP factors and IRF3 was positively correlated with cell proliferation. In addition, an analysis evaluating clinical features demonstrated that STAT6-positive cases showed a longer progression-free survival time than STAT6-negative cases. Our evaluation, conducted using a limited number of specimens, suggests SASP may be prevalent in early gastric neoplastic lesions and could be activated by accelerated cell proliferation-induced DDR. The clinical significance of SASP still needs to be determined. C1 [Liang, Li] Peking University First Hospital, Department of PathologyBeijing, China. [Chai, Yijie] Health Science Center, Peking University, Department of PathologyBeijing, China. [Chai, Fei] Shanxi Medical University, Fenyang College, Department of PathologyFenyang, China. [Liu, Haijing] Health Science Center, Peking University, Department of PathologyBeijing, China. [Ma, Ningning] Health Science Center, Peking University, Department of PathologyBeijing, China. [Zhang, Hong] Peking University First Hospital, Department of PathologyBeijing, China. [Zhang, Shuang] Peking University First Hospital, Department of PathologyBeijing, China. [Nong, Lin] Peking University First Hospital, Department of PathologyBeijing, China. [Li, Ting] Peking University First Hospital, Department of PathologyBeijing, China. [Zhang, Bo] Health Science Center, Peking University, Department of PathologyBeijing, China. RP Zhang, B (reprint author), Health Science Center, Peking University, Department of Pathology, Beijing, China. EM 13601150230@163.com CR Acosta JC, O’Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, et al. Chemokine Signaling via the CXCR2 Receptor Reinforces Senescence. Cell, 2018, 133:1006–18., DOI 10.1016/j.cell.2008.03.038 Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, et al. Oncogene-induced Senescence Relayed by an Interleukin-dependent Inflammatory Network. Cell, 2008, 133(6):1019–31., DOI 10.1016/j.cell.2008. .039 3. Gluck S, Guey B, Gulen MF, Wolter K, Kang TW, Schmacke NA, et al. Innate Immune Sensing of Cytosolic Chromatin Fragments through cGAS Promotes Senescence. Nat Cel Biol, 2017, 19(9):1061–70., DOI 10.1038/ncb3586 Li T, Chen ZJ. The cGAS-cGAMP-STING Pathway Connects DNA Damage to Inflammation, Senescence, and Cancer. J Exp Med, 2018, 215(5):1287–99., DOI 10.1084/jem.20180139 Pepin G, Gantier MP. cGAS-STING Activation in the Tumor Microenvironment and its Role in Cancer Immunity. Adv Exp Med Biol, 2017, 1024:175–94., DOI 10.1007/978-981-10-5987-2_8 Chen Q, Sun L, Chen ZJ. Regulation and Function of the cGAS-STING Pathway of Cytosolic DNA Sensing. Nat Immunol, 2016, 17(10):1142–9., DOI 10.1038/ni.3558 Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP Synthase Is a Cytosolic DNA Sensor that Activates the Type I Interferon Pathway. Science, 2013, 339(6121):786–91., DOI 10.1126/science.1232458 Loaiza N, Demaria M. Cellular Senescence and Tumor Promotion: Is Aging the Key? Biochim Biophys Acta, 2016, 1865(2):155–67., DOI 10.1016/j.bbcan. 2016.01.007 Loo TM, Miyata K, Tanaka Y, Takahashi A. Cellular Senescence and Senescence-Associated Secretory Phenotype via the cGAS-STING Signaling Pathway in Cancer. Cancer Sci, 2020, 111(2):304–11., DOI 10. 1111/cas.14266 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence andMortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Tan P, Yeoh KG. Genetics and Molecular Pathogenesis of Gastric Adenocarcinoma. Gastroenterology, 2015, 149(5):1153–62., DOI 10.1053/j. gastro.2015.05.059 Joshi SS, Badgwell BD. Current Treatment and Recent Progress in Gastric Cancer. CA Cancer J Clin, 2021, 71(3):264–79., DOI 10.3322/caac.21657 Hanazono K, Natsugoe S, Stein HJ, Aikou T, Hoefler H, Siewert JR. Distribution of P53 Mutations in Esophageal and Gastric Carcinomas and the Relationship with P53 Expression. Oncol Rep, 2006, 15(4):821–4., DOI 10. 3892/or.15.4.821 Flejou JF, Gratio V, Muzeau F, Hamelin R. p53 Abnormalities in Adenocarcinoma of the Gastric Cardia and Antrum. Mol Pathol, 1999, 52(5):263–8., DOI 10.1136/mp.52.5.263 Cancer Genome Atlas Research N. Comprehensive Molecular Characterization of Gastric Adenocarcinoma. Nature, 2014, 513(7517): 202–9., DOI 10.1038/nature13480 Baek DH, Kim GH, Park DY, Lee BE, Jeon HK, Lim W, et al. Gastric Epithelial Dysplasia: Characteristics and Long-Term Follow-Up Results after Endoscopic Resection According to Morphological Categorization. BMC Gastroenterol, 2015, 15:17., DOI 10.1186/s12876-015-0249-7 Schlemper RJ, Kato Y, Stolte M. Review of Histological Classifications of Gastrointestinal Epithelial Neoplasia: Differences in Diagnosis of Early Carcinomas between Japanese and Western Pathologists. J Gastroenterol, 2001, 36(7):445–56., DOI 10.1007/s005350170067 Lokuhetty D, White VA, Watanabe R. WHO Classification of Digestive System Tumours. 5th ed. Lyon, France: IARC Press, 2018). p. 64–109. Hwang HJ, Nam SK, Park H, Park Y, Koh J, Na HY, et al. Prediction of TP53 Mutations by P53 Immunohistochemistry and Their Prognostic Significance in Gastric Cancer. J Pathol Transl Med, 2020, 54(5):378–86., DOI 10.4132/jptm.2020.06.01 Sunter JP. Cell Proliferation in Gastrointestinal Carcinogenesis. Scand J Gastroenterol Suppl, 1984)(104, 45–55. Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, et al. Molecular Analysis of Gastric Cancer Identifies Subtypes Associated with Distinct Clinical Outcomes. Nat Med, 2015, 21(5):449–56., DOI 10.1038/nm.3850 Serra O, Galan M, Ginesta MM, Calvo M, Sala N, Salazar R. Comparison and Applicability of Molecular Classifications for Gastric Cancer. Cancer Treat Rev, 2019, 77:29–34., DOI 10.1016/j.ctrv Sethi NS, Kikuchi O, Duronio GN, Stachler MD, McFarland JM, Ferrer-Luna R, et al. Early TP53 Alterations Engage Environmental Exposures to Promote Gastric Premalignancy in an Integrative Mouse Model. Nat Genet, 2020, 52(2):219–30., DOI 10.1038/s41588-019-0574-9 Sun Y, Coppe JP, Lam EW. Cellular Senescence: The Sought or the Unwanted? Trends Mol Med, 2018, 24(10):871–85., DOI 10.1016/j.molmed. 2018.08.002 Wang Y, Luo J, Alu A, Han X, Wei Y, Wei X. cGAS-STING Pathway in Cancer Biotherapy. Mol Cancer, 2020, 19(1):136., DOI 10.1186/s12943-020- 01247-w Matsumoto Y, Marusawa H, Kinoshita K, Endo Y, Kou T, Morisawa T, et al. Helicobacter pylori Infection Triggers Aberrant Expression of Activation- Induced Cytidine Deaminase in Gastric Epithelium. Nat Med, 2007, 13(4): 470–6., DOI 10.1038/nm1566 Nishizawa T, Suzuki H. Gastric Carcinogenesis and Underlying Molecular Mechanisms: Helicobacter pylori and Novel Targeted Therapy. Biomed Res Int, 2015, 2015:794378., DOI 10.1155/2015/794378 Macheret M, Halazonetis TD. DNA Replication Stress as a Hallmark of Cancer. Annu Rev Pathol, 2015, 10:425–48., DOI 10.1146/annurev-pathol- 012414-040424 Primo LMF, Teixeira LK. DNA Replication Stress: Oncogenes in the Spotlight. Genet Mol Biol, 2019, 43(1):e20190138., DOI 10.1590/1678-4685GMB-2019- 0138 Gaillard H, Garcia-Muse T, Aguilera A. Replication Stress and Cancer. Nat Rev Cancer, 2015, 15(5):276–89., DOI 10.1038/nrc3916 Smith J, Tho LM, Xu N, Gillespie DA. The ATM-Chk2 and ATR-Chk1 Pathways in DNA Damage Signaling and Cancer. Adv Cancer Res, 2010, 108:73–112., DOI 10.1016/B978-0-12-380888-2.00003-0 Murina O, von Aesch C, Karakus U, Ferretti LP, Bolck HA, Hanggi K, et al. FANCD2 and CtIP Cooperate to Repair DNA Interstrand Crosslinks. Cell Rep, 2014, 7(4):1030–8., DOI 10.1016/j.celrep.2014.03.069 Cuella-Martin R, Hayward SB, Fan X, Chen X, Huang JW, Taglialatela A, et al. Functional Interrogation of DNA Damage Response Variants with Base Editing Screens. Cell, 2021, 184(4):1081–97., DOI 10.1016/j.cell.2021. 01.041 Kais Z, Rondinelli B, Holmes A, O’Leary C, Kozono D, D’Andrea AD, et al. FANCD2 Maintains Fork Stability in BRCA1/2-Deficient Tumors and Promotes Alternative End-Joining DNA Repair. Cel Rep, 2016, 15(11): 2488–99., DOI 10.1016/j.celrep.2016.05.031 Schlemper RJ, Kato Y, Stolte M. Review of Histological Classifications of Gastrointestinal Epithelial Neoplasia: Differences in Diagnosis of Early Carcinomas between Japanese and Western Pathologists. J Gastroenterol, 2001, 36(7):445–56., DOI 10.1007/s005350170067 Sung JK. Diagnosis and Management of Gastric Dysplasia. Korean J Intern Med, 2016, 31(2):201–9., DOI 10.3904/kjim.2016.021 Yao M, Wu Y, Cao Y, Liu H, Ma N, Chai Y, et al. Autophagy-Mediated Clearance of Free Genomic DNA in the Cytoplasm Protects the Growth and Survival of Cancer Cells. Front Oncol, 2021, 11:667920., DOI 10.3389/fonc.2021. 667920 Verhoeven Y, Tilborghs S, Jacobs J, De Waele J, Quatannens D, Deben C, et al. The Potential and Controversy of Targeting STAT Family Members in Cancer. Semin Cancer Biol, 2020, 60:41–56., DOI 10.1016/j.semcancer.2019. 10.002 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610401 EP 1610413 DI 10.3389/pore.2022.1610401 PG 13 ER PT J AU Hu, J Yang, J Chen, L Meng, X Zhang, X Li, W Li, Z Huang, G AF Hu, Jinxian Yang, Jihu Chen, Lei Meng, Xiangbao Zhang, Xiejun Li, Weiping Li, Zongyang Huang, Guodong TI Alterations of the Gut Microbiome in Patients With Pituitary Adenoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE pituitary adenoma; invasive pituitary adenoma; gut microbiome; metagenome sequencing; noninvasive pituitary adenoma ID pituitary adenoma; invasive pituitary adenoma; gut microbiome; metagenome sequencing; noninvasive pituitary adenoma AB Pituitary adenoma (PA) includes invasive pituitary adenoma (IPA) and noninvasive pituitary adenoma (NIPA), which are associated with the endocrine system. The gut microbiome plays an important role in human metabolism, but the association between the gut microbiome and pituitary adenoma remains unclear. A total of 44 subjects were enrolled in this study. Of these, 29 PA patients were further divided into IPA patients (n = 13) and NIPA patients (n = 16), while 15 healthy age-matched subjects were defined as control subjects. We collected faecal samples and characterized the gut microbial profiles by metagenomic sequencing using the Illumina X-ten platform. PLS-DA showed different microbial clusters among the three groups, and slightly different microbial ecological networks were observed. LEfSe analysis revealed significant alterations in the microbial community among PA patients. In particular, the enrichment of Clostridium innocuum, along with the reduced abundance of Oscillibacter sp. 57_20 and Fusobacterium mortiferum, were observed both in the IPA and NIPA groups compared to the control group. Moreover, PA patients could be effectively classified based on these bacteria using a support vector machine algorithm. In summary, this study demonstrated significant differences in the gut microbiome between PA patients and healthy controls. Future mechanistic experiments are needed to determine whether such alterations are a cause or consequence of pituitary adenoma. C1 [Hu, Jinxian] The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, Department of NeurosurgeryShenzhen, China. [Yang, Jihu] The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, Department of NeurosurgeryShenzhen, China. [Chen, Lei] The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, Department of NeurosurgeryShenzhen, China. [Meng, Xiangbao] The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, Department of NeurosurgeryShenzhen, China. [Zhang, Xiejun] The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, Department of NeurosurgeryShenzhen, China. [Li, Weiping] The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, Department of NeurosurgeryShenzhen, China. [Li, Zongyang] The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, Department of NeurosurgeryShenzhen, China. [Huang, Guodong] The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, Department of NeurosurgeryShenzhen, China. RP Huang, G (reprint author), The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, Department of Neurosurgery, Shenzhen, China. EM huangguodong@email.szu.edu.cn CR Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, Barnholtz-Sloan JS, et al. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013-2017. Neuro Oncol, 2020, 22: iv1–iv96., DOI 10.1093/neuonc/noaa200 Li J, Qian Y, Zhang C,Wang W, Qiao Y, Song H, et al. LncRNA LINC00473 Is Involved in the Progression of Invasive Pituitary Adenoma by Upregulating KMT5A via ceRNA-Mediated miR-502-3p Evasion. Cell Death Dis, 2021, 12: 580., DOI 10.1038/s41419-021-03861-y Yang Q, Li X. Molecular Network Basis of Invasive Pituitary Adenoma: A Review. Front Endocrinol, Lausanne,, 2019, 10:7., DOI 10.3389/fendo.2019.00007 Chiloiro S, Doglietto F, Trapasso B, Iacovazzo D, Giampietro A, Di Nardo F, et al. Typical and Atypical Pituitary Adenomas: A Single-center Analysis of Outcome and Prognosis. Neuroendocrinology, 2015, 101:143–50., DOI 10.1159/000375448 Banskota S, Adamson DC. Pituitary Adenomas: From Diagnosis to Therapeutics. Biomedicines, 2021, 9:494., DOI 10.3390/biomedicines9050494 Zheng P, Zeng B, Liu M, Chen J, Pan J, Han Y, et al. The GutMicrobiome from Patients with Schizophrenia Modulates the Glutamate-Glutamine-GABA Cycle and Schizophrenia-Relevant Behaviors in Mice. Sci Adv, 2015, 5: eaau8317., DOI 10.1126/sciadv.aau8317 Martin CR, Osadchiy V, Kalani A, Mayer EA. The Brain-Gut-Microbiome Axis. Cell Mol Gastroenterol Hepatol, 2018, 6:133–48., DOI 10.1016/j.jcmgh.2018.04.003 Mohajeri MH, La Fata G, Steinert RE, Weber P. Relationship between the Gut Microbiome and Brain Function. Nutr Rev, 2018, 76:481–96., DOI 10.1093/ nutrit/nuy009 Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, et al. A Core Gut Microbiome in Obese and Lean Twins. Nature, 2009, 457: 480–4., DOI 10.1038/nature07540 Gurung M, Li Z, You H, Rodrigues R, Jump DB, Morgun A, et al. Role of Gut Microbiota in Type 2 Diabetes Pathophysiology. EBioMedicine, 2020, 51: 102590., DOI 10.1016/j.ebiom.2019.11.051 Valdes AM, Walter J, Segal E, Spector TD. Role of the Gut Microbiota in Nutrition and Health. BMJ, 2018, 361:k2179., DOI 10.1136/bmj.k2179 Farzi A, Frohlich EE, Holzer P. Gut Microbiota and the Neuroendocrine System. Neurotherapeutics, 2018, 15:5–22., DOI 10.1007/s13311-017-0600-5 Hacioglu A, Gundogdu A, Nalbantoglu U, Karaca Z, Urhan ME, Sahin S, et al. Gut Microbiota in Patients with Newly Diagnosed Acromegaly: A Pilot Cross- Sectional Study. Pituitary, 2021, 24:600–10., DOI 10.1007/s11102-021-01137-4 Lan X, Gao H, Wang F, Feng J, Bai J, Zhao P, et al. Whole-exome Sequencing Identifies Variants in Invasive Pituitary Adenomas. Oncol Lett, 2016, 12: 2319–28., DOI 10.3892/ol.2016.5029 Zhou S, Xu R, He F, Zhou J, Wang Y, Zhou J, et al. Diversity of GutMicrobiota Metabolic Pathways in 10 Pairs of Chinese Infant Twins. PLoS One, 2016, 11: e0161627., DOI 10.1371/journal.pone.0161627 Chen S, Zhou Y, Chen Y, Gu J. Fastp: An Ultra-fast All-In-One FASTQ Preprocessor. Bioinformatics, 2018, 34:i884–90., DOI 10.1093/bioinformatics/bty560 Li H, Durbin R. Fast and Accurate Short Read Alignment with Burrows- Wheeler Transform. Bioinformatics, 2009, 25:1754–60., DOI 10.1093/ bioinformatics/btp324 Beghini F, McIver LJ, Blanco-Miguez A, Dubois L, Asnicar F, Maharjan S, et al. Integrating Taxonomic, Functional, and Strain-Level Profiling of DiverseMicrobial Communities with bioBakery 3. Elife, 2021, 10:e65088., DOI 10.7554/eLife.65088 Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. Metagenomic Biomarker Discovery and Explanation. Genome Biol, 2011, 12: R60., DOI 10.1186/gb-2011-12-6-r60 Love MI, Huber W, Anders S. Moderated Estimation of Fold Change and Dispersion for RNA-Seq Data with DESeq2. Genome Biol, 2014, 15:550., DOI 10.1186/s13059-014-0550-8 Kurtz ZD, Muller CL, Miraldi ER, Littman DR, Blaser MJ, Bonneau RA, et al. Sparse and Compositionally Robust Inference of Microbial Ecological Networks. Plos Comput Biol, 2015, 11:e1004226., DOI 10.1371/journal.pcbi.1004226 Magne F, Gotteland M, Gauthier L, Zazueta A, Pesoa S, Navarrete P, et al. The Firmicutes/Bacteroidetes Ratio: A Relevant Marker of Gut Dysbiosis in Obese Patients? Nutrients, 2020, 12:1474., DOI 10.3390/nu12051474 Cherny KE, Muscat EB, Reyna ME, Kociolek LK. Clostridium Innocuum: Microbiological and Clinical Characteristics of a Potential Emerging Pathogen. Anaerobe, 2021, 71:102418., DOI 10.1016/j.anaerobe.2021.102418 Ha CWY, Martin A, Sepich-Poore GD, Shi B, Wang Y, Gouin K, et al. Translocation of Viable Gut Microbiota to Mesenteric Adipose Drives Formation of Creeping Fat in Humans. Cell, 2020, 183:666–83., DOI 10. 1016/j.cell.2020.09.009 Thomas AM, Asnicar F, Kroemer G, Segata N. Genes EncodingMicrobial Acyl Coenzyme A Binding Protein/Diazepam-Binding Inhibitor Orthologs Are Rare in the Human Gut Microbiome and Show No Links to Obesity. Appl Environ Microbiol, 2021, 87:e0047121., DOI 10.1128/AEM.00471-21 Zeybel M, Altay O, Arif M, Li X, Yang H, Fredolini C, et al. Combined Metabolic Activators Therapy Ameliorates Liver Fat in Nonalcoholic Fatty Liver Disease Patients. Mol Syst Biol, 2021, 17:e10459., DOI 10.15252/msb. 202110459 Tarallo S, Ferrero G, De Filippis F, Francavilla A, Pasolli E, Panero V, et al. Stool microRNA Profiles Reflect Different Dietary and Gut Microbiome Patterns in Healthy Individuals. Gut, 2021, 71:1302–14., DOI 10.1136/gutjnl- 2021-325168 Liang S, Mao Y, Liao M, Xu Y, Chen Y, Huang X, et al. Gut Microbiome Associated with APC Gene Mutation in Patients with Intestinal Adenomatous Polyps. Int J Biol Sci, 2020, 16:135–46., DOI 10.7150/ijbs.37399 Portrait V, Cottenceau G, Pons AM. A Fusobacterium Mortiferum Strain Produces a Bacteriocin-like Substance(s, Inhibiting Salmonella Enteritidis. Lett Appl Microbiol, 2000, 31:115–7., DOI 10.1046/j.1365-2672.2000.00774.x Mocking RJ, Verburg HF,Westerink AM, Assies J, Vaz FM, Koeter MW, et al. Fatty Acid Metabolism and its Longitudinal Relationship with the Hypothalamic-Pituitary-Adrenal axis in Major Depression: Associations with Prospective Antidepressant Response. Psychoneuroendocrinology, 2015, 59:1–13., DOI 10.1016/j.psyneuen.2015.04.027 Casanueva FF, Villanueva L, Dieguez C, Diaz Y, Cabranes JA, Szoke B, et al. Free Fatty Acids Block Growth Hormone, GH, Releasing Hormone- Stimulated GH Secretion in Man Directly at the Pituitary. J Clin Endocrinol Metab, 1987, 65:634–42., DOI 10.1210/jcem-65-4-634 Sari DK, Kuwahara S, Tsukamoto Y, Hori H, Kunugita N, Arashidani K, et al. Effect of Prolonged Exposure to Low Concentrations of Formaldehyde on the Corticotropin Releasing Hormone Neurons in the Hypothalamus and Adrenocorticotropic Hormone Cells in the Pituitary Gland in Female Mice. Brain Res, 2004, 1013:107–16., DOI 10.1016/j.brainres.2004.03.070 Schwarzer M, Makki K, Storelli G, Machuca-Gayet I, Srutkova D, Hermanova P, et al. Lactobacillus Plantarum Strain Maintains Growth of Infant Mice during Chronic Undernutrition. Science, 2016, 351:854–7., DOI 10.1126/ science.aad8588 Fu X, Liu Z, Zhu C, Mou H, Kong Q. Nondigestible Carbohydrates, Butyrate, and Butyrate-Producing Bacteria. Crit Rev Food Sci Nutr, 2019, 59:S130–S152., DOI 10.1080/10408398.2018.1542587 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610402 EP 1610409 DI 10.3389/pore.2022.1610402 PG 8 ER PT J AU Liang, ZQ He, RQ Luo, JY Huang, ZG Li, J Zhong, LY Chen, JH Huang, SN Shi, L Wei, KL Zeng, JH Zeng, JJ Chen, G AF Liang, Zi-Qian He, Rong-Quan Luo, Jia-Yuan Huang, Zhi-Guang Li, Jie Zhong, Lu-Yang Chen, Jun-Hong Huang, Su-Ning Shi, Lin Wei, Kang-Lai Zeng, Jiang-Hui Zeng, Jing-Jing Chen, Gang TI Downregulated Dual-Specificity Protein Phosphatase 1 in Ovarian Carcinoma: A Comprehensive Study With Multiple Methods SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE molecular docking; dual-specificity protein phosphatase 1; immune landscape; cancer associated fibroblasts; ovarian carcinoma ID molecular docking; dual-specificity protein phosphatase 1; immune landscape; cancer associated fibroblasts; ovarian carcinoma AB Introduction: We aimed to explore the abnormal expression of dual-specificity protein phosphatase 1 (DUSP1) and its latent molecular mechanisms in ovarian carcinoma (OVCA). Materials and Methods: Two clinical cohorts collected from two different hospitals were used to evaluate the expression of DUSP1 protein in OVCA tissues. RNA-sequencing and microarray datasets were utilised to verify DUSP1 expression at mRNA levels in both OVCA tissues and in the peripheral blood of OVCA patients. Furthermore, an integrated calculation was performed to pool the standard mean difference (SMD) fromeach cohort in order to comprehensively assess the expression of DUSP1 in OVCA. Furthermore, we examined the relationship among DUSP1, tumour microenvironment (TME), and chemotherapy resistance in OVCA. Moreover, we used pathway enrichment analysis to explore the underlying mechanisms of DUSP1 in OVCA. Results: A pooled SMD of −1.19 (95% CI [−2.00, −0.38], p = 0.004) with 1,240 samples revealed that DUSP1 was downregulated in OVCA at both mRNA and protein levels. The area under the receiver operating characteristic curve of 0.9235 indicated the downregulated DUSP1 in peripheral blood may have a non-invasive diagnostic value in OVCA. Through six algorithms, we identified that DUSP1 may related to tumour-infiltrating T cells and cancer associated fibroblasts (CAFs) in OVCA. Pathway enrichment demonstrated that DUSP1 might participate in the mitogen-activated protein kinase (MAPK) signalling pathway. Furthermore, DUSP1 may have relations with chemotherapy resistance, and a favourable combining affinity was observed in the paclitaxel-DUSP1 docking model. Conclusion: DUSP1 was downregulated in OVCA, and this decreasing trend may affect the infiltration of CAFs. Finally, DUSP1 may have a targeting relation with paclitaxel and participate in MAPK signaling pathways. C1 [Liang, Zi-Qian] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [He, Rong-Quan] The First Affiliated Hospital of Guangxi Medical University, Department of Medical OncologyNanning, China. [Luo, Jia-Yuan] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Huang, Zhi-Guang] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Li, Jie] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Zhong, Lu-Yang] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Chen, Jun-Hong] Guangxi Maternal and Child Health Hospital, Department of PathologyNanning, China. [Huang, Su-Ning] Guangxi Medical University Cancer Hospital, Department of RadiotherapyNanning, China. [Shi, Lin] The Second Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Wei, Kang-Lai] The Second Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Zeng, Jiang-Hui] The Third Affiliated Hospital of Guangxi Medical University/Nanning Second People’s Hospital, Department of Clinical LaboratoryNanning, China. [Zeng, Jing-Jing] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. [Chen, Gang] The First Affiliated Hospital of Guangxi Medical University, Department of PathologyNanning, China. RP Chen, G (reprint author), The First Affiliated Hospital of Guangxi Medical University, Department of Pathology, Nanning, China. EM chengang@gxmu.edu.cn CR Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin, 2021, 71(1):7–33., DOI 10.3322/caac.21654 Shin W, Park S-Y, Kang S, Lim MC, Seo S-S. How to Manage Synchronous Endometrial and Ovarian Cancer Patients? BMC Cancer, 2021, 21(1):489., DOI 10.1186/s12885-021-08220-w Adambekov S, Lopa S, Edwards RP, Lemon L, Wang S, Taylor SE, et al. Survival and Recurrence after Intraperitoneal Chemotherapy Use: Retrospective Review of Ovarian Cancer Hospital Registry Data. Cancer Med, 2020, 9(20):7388–97., DOI 10.1002/cam4.3340 Pan X, Bi F. A Potential Immune-Related Long Non-coding RNA Prognostic Signature for Ovarian Cancer. Front Genet, 2021, 12:694009., DOI 10.3389/ fgene.2021.694009 Sun X, Liu Q, Huang J, Diao G, Liang Z. Transcriptome-based Stemness Indices Analysis Reveals Platinum-Based Chemo-Theraputic Response Indicators in Advanced-Stage Serous Ovarian Cancer. Bioengineered, 2021, 12(1):3753–71., DOI 10.1080/21655979.2021.1939514 Ma Q, Song J, Wang S, He N. MUC1 Regulates AKT Signaling Pathway by Upregulating EGFR Expression in Ovarian Cancer Cells. Pathol Res Pract, 2021, 224:153509., DOI 10.1016/j.prp.2021.153509 Xue B, Li S, Jin X, Liu L. Bioinformatics Analysis of mRNA and miRNA Microarray to Identify the Key miRNA-mRNA Pairs in Cisplatin-Resistant Ovarian Cancer. BMC Cancer, 2021, 21(1):452., DOI 10.1186/s12885-021- 166-z 8. Yin L, Wang Y. Long Non-coding RNA NEAT1 Facilitates the Growth, Migration, and Invasion of Ovarian Cancer Cells via the Let-7 g/MEST/ ATGL axis. Cancer Cell Int, 2021, 21(1):437., DOI 10.1186/s12935-021-02018-3 Guo J, Zhu Y, Yu L, Li Y, Guo J, Cai J, et al. Aspirin Inhibits Tumor Progression and Enhances Cisplatin Sensitivity in Epithelial Ovarian Cancer. PeerJ, 2021, 9:e11591., DOI 10.7717/peerj.11591 Zhu D, Huang J, Liu N, Li W, Yan L. PSMC2/CCND1 axis Promotes Development of Ovarian Cancer through Regulating Cell Growth, Apoptosis and Migration. Cell Death Dis, 2021, 12(8):730., DOI 10.1038/ s41419-021-03981-5 Falzone L, Scandurra G, Lombardo V, Gattuso G, Lavoro A, Distefano AB, et al. A Multidisciplinary Approach Remains the Best Strategy to Improve and Strengthen the Management of Ovarian Cancer, Review). Int J Oncol, 2021, 59(1):53., DOI 10.3892/ijo.2021.5233 Segura-Sampedro JJ, Morales-Soriano R, Arjona-Sanchez A, Cascales-Campos P. Secondary Surgical Cytoreduction Needs to Be Assessed Taking into Account Surgical Technique, Completeness of Cytoreduction, and Extent of Disease. World J Surg Onc, 2020, 18(1):92., DOI 10.1186/s12957-020-01853-4 Huang X, Xie C, Tang J, HeW, Yang F, Tian W, et al. Adipose Tissue Area as a Predictor for the Efficacy of Apatinib in Platinum-Resistant Ovarian Cancer: An Exploratory Imaging Biomarker Analysis of the AEROC Trial. BMC Med, 2020, 18(1):267., DOI 10.1186/s12916-020-01733-4 Gao L, Pang Y-Y, Guo X-Y, Zeng J-J, Tang Z-Q, Xiong D-D, et al. Polo like Kinase 1 Expression in Cervical Cancer Tissues Generated from Multiple Detection Methods. PeerJ, 2020, 8:e10458., DOI 10.7717/peerj.10458 Kato MK, Yunokawa M, Bun S, Shimoi T, Yonemori K, Miyasaka N, et al. Treatment Strategies for Recurrent Ovarian Cancer in Older Adult Patients in Japan: A Study Based on Real-World Data. J Cancer Res Clin Oncol, 2020, 146(5):1335–41., DOI 10.1007/s00432-020-03168-z He L, Bulanova D, Oikkonen J, Hakkinen A, Zhang K, Zheng S, et al. Networkguided Identification of Cancer-Selective Combinatorial Therapies in Ovarian Cancer. Brief Bioinformatics, 2021, 22:bbab272., DOI 10.1093/bib/bbab272 Majidi A, Na R, Jordan SJ, De Fazio A, Webb PM. Statin Use and Survival Following a Diagnosis of Ovarian Cancer: A Prospective Observational Study. Int J Cancer, 2021, 148(7):1608–15., DOI 10.1002/ijc.33333 Li S, Yang T, Xiang Y, Li X, Zhang L, Deng S. Clinical Characteristics and Survival Outcomes of Malignant Struma Ovarii Confined to the Ovary. BMC Cancer, 2021, 21(1):383., DOI 10.1186/s12885-021-08118-7 Schuler-Toprak S, Weber F, Skrzypczak M, Ortmann O, Treeck O. Expression of Estrogen-Related Receptors in Ovarian Cancer and Impact on Survival. J Cancer Res Clin Oncol, 2021, 147(9):2555–67., DOI 10.1007/s00432-021- 03673-9 Leandersson P, Hogberg T, Dickman PW, Malander S, Borgfeldt C. Incidence and Survival of Epithelial Ovarian, Fallopian Tube, Peritoneal, and Undesignated Abdominal/pelvic Cancers in Sweden 1960-2014: A Population-Based Cohort Study. BMC Cancer, 2021, 21(1):465., DOI 10. 1186/s12885-021-08169-w Teixeira LA, Candido Dos Reis FJ. BRCA1 Expression by Immunohistochemistry and Prognosis in Ovarian Cancer: A Systematic Review and Meta-Analysis. Targ Oncol, 2020, 15(1):37–46., DOI 10.1007/ s11523-020-00697-y Jin Q, Li R, Hu N, Xin T, Zhu P, Hu S, et al. DUSP1 Alleviates Cardiac Ischemia/reperfusion Injury by Suppressing the Mff-Required Mitochondrial Fission and Bnip3-Related Mitophagy via the JNK Pathways. Redox Biol, 2018, 14:576–87., DOI 10.1016/j.redox.2017.11.004 Shen J, Zhang Y, Yu H, Shen B, Liang Y, Jin R, et al. Role of DUSP1/MKP1 in Tumorigenesis, Tumor Progression and Therapy. Cancer Med, 2016, 5(8): 2061–8., DOI 10.1002/cam4.772 Chen X, Yu C, Kang R, Kroemer G, Tang D. Cellular Degradation Systems in Ferroptosis. Cell Death Differ, 2021, 28(4):1135–48., DOI 10.1038/s41418-020-00728-1 Prager BC, Vasudevan HN, Dixit D, Bernatchez JA, Wu Q, Wallace LC, et al. The Meningioma Enhancer Landscape Delineates Novel Subgroups and Drives Druggable Dependencies. Cancer Discov, 2020, 10(11):1722–41., DOI 10.1158/2159-8290.cd-20-0160 Martinez-Martinez D, Toledo Lobo MV, Baquero P, Ropero S, Angulo JC, Chiloeches A, et al. Downregulation of Snail by DUSP1 Impairs Cell Migration and Invasion through the Inactivation of JNK and ERK and Is Useful as a Predictive Factor in the Prognosis of Prostate Cancer. Cancers, 2021, 13(5): 1158., DOI 10.3390/cancers13051158 Du Y, Lu S, Ge J, Long D, Wen C, Tan S, et al. ROCK2 Disturbs MKP1 Expression to Promote Invasion and Metastasis in Hepatocellular Carcinoma. Am J Cancer Res, 2020, 10(3):884–96. PLOS ONE Editors. Expression of Concern: DUSP1 Is a Novel Target for Enhancing Pancreatic Cancer Cell Sensitivity to Gemcitabine. PloS one, 2020, 15(5):e0233098., DOI 10.1371/journal.pone.0233098 Kang Y-S, Seok H-J, Jeong E-J, Kim Y, Yun S-J, Min J-K, et al. DUSP1 Induces Paclitaxel Resistance through the Regulation of P-Glycoprotein Expression in Human Ovarian Cancer Cells. Biochem Biophys Res Commun, 2016, 478(1): 403–9., DOI 10.1016/j.bbrc.2016.07.035 Zhang X, Su Y, Wu X, Xiao R, Wu Y, Yang B, et al. Integrative Analysis of the Common Genetic Characteristics in Ovarian Cancer Stem Cells Sorted byMultiple Approaches. J Ovarian Res, 2020, 13(1):116., DOI 10.1186/s13048-020-00715-7 Li L, Cai S, Liu S, Feng H, Zhang J. Bioinformatics Analysis to Screen the Key Prognostic Genes in Ovarian Cancer. J Ovarian Res, 2017, 10(1):27., DOI 10. 1186/s13048-017-0323-6 Yang Y, Zhou J-Y, Zhao L-J, Gao B-R, Wan X-P, Wang J-L. Dual-specificity Phosphatase 1 Deficiency Induces Endometrioid Adenocarcinoma Progression via Activation of Mitogen-Activated Protein Kinase/ Extracellular Signal-Regulated Kinase Pathway. Chin Med J, 2016, 129(10): 1154–60., DOI 10.4103/0366-6999.181954 Liang Z-Q, Zhong LY, Li J, Shen JH, Tu XY, Zhong ZH, et al. Clinicopathological Significance and Underlying Molecular Mechanism of Downregulation of Basonuclin 1 Expression in Ovarian Carcinoma. Exp Biol Med, 2021, 247:15353702211052036., DOI 10.1177/ 15353702211052036 Liang ZQ, Gao L, Chen JH, Dai WB, Su YS, Chen G. Downregulation of the Coiled-Coil Domain Containing 80 and its Perspective Mechanisms in Ovarian Carcinoma: A Comprehensive Study. Int J Genomics, 2021, 2021: 3752871., DOI 10.1155/2021/3752871 Gautier L, Cope L, Bolstad BM, Irizarry RA. Affy-Analysis of Affymetrix GeneChip Data at the Probe Level. Bioinformatics, 2004, 20(3):307–15., DOI 10. 1093/bioinformatics/btg405 Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The Sva Package for Removing Batch Effects and Other Unwanted Variation in High-Throughput Experiments. Bioinformatics, Oxford, England,, 2012, 28(6):882–3., DOI 10. 1093/bioinformatics/bts034 Gao L, Yan S-b., Yang J, Kong J-l., Shi K, Ma F-c., et al. MiR-182-5p and its Target HOXA9 in Non-small Cell Lung Cancer: A Clinical and In-Silico Exploration with the Combination of RT-qPCR, miRNA-Seq and miRNAChip. BMC Med Genomics, 2020, 13(1):3., DOI 10.1186/s12920-019-0648-7 Pang YY, Li JD, Gao L, Yang X, Dang YW, Lai ZF, et al. The Clinical Value and PotentialMolecularMechanism of the Downregulation of MAOA in Hepatocellular Carcinoma Tissues. Cancer Med, 2020, 9(21):8004–19., DOI 10.1002/cam4.3434 Sturm G, Finotello F, Petitprez F, Zhang JD, Baumbach J, Fridman WH, et al. Comprehensive Evaluation of Transcriptome-Based Cell-type Quantification Methods for Immuno-Oncology. Bioinformatics, Oxford, England,, 2019, 35(14):i436–i445., DOI 10.1093/bioinformatics/btz363 Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, et al. clusterProfiler 4.0: A Universal Enrichment Tool for Interpreting Omics Data. Innovation, 2021, 2:100141., DOI 10.1016/j.xinn.2021.100141 Bu D, Luo H, Huo P, Wang Z, Zhang S, He Z, et al. KOBAS-I: Intelligent Prioritization and Exploratory Visualization of Biological Functions for Gene Enrichment Analysis. Nucleic Acids Res, 2021, 49(W1):W317–W325., DOI 10. 1093/nar/gkab447 Geeleher P, Cox N, Huang RS. pRRophetic: An R Package for Prediction of Clinical Chemotherapeutic Response from Tumor Gene Expression Levels. PloS one, 2014, 9(9):e107468., DOI 10.1371/journal.pone.0107468 Trott O, Olson AJ. AutoDock Vina: Improving the Speed and Accuracy of Docking with a New Scoring Function, Efficient Optimization, and Multithreading. J Comput Chem, 2010, 31(2):455–61., DOI 10.1002/jcc.21334 Zhang Y, Zhang Y, Chen M, Liu C, Xiang C. DUSP1 Is Involved in the Progression of Small Cell Carcinoma of the Prostate. Saudi J Biol Sci, 2018, 25(5):858–62., DOI 10.1016/j.sjbs.2017.09.015 Chiofalo B, Bruni S, Certelli C, Sperduti I, Baiocco E, Vizza E. Primary Debulking Surgery vs. Interval Debulking Surgery for Advanced Ovarian Cancer: Review of the Literature and Meta-Analysis. Minerva Med, 2019, 110(4):330–40., DOI 10.23736/S0026-4806.19.06078-6 Chen Z, Chen Q, Cheng Z, Gu J, Feng W, Lei T, et al. Long Non-coding RNA CASC9 Promotes Gefitinib Resistance in NSCLC by Epigenetic Repression of DUSP1. Cell Death Dis, 2020, 11(10):858., DOI 10.1038/s41419-020-03047-y Du Y, Wei N, Ma R, Jiang S-H, Song D. Long Noncoding RNA MIR210HG Promotes the Warburg Effect and Tumor Growth by Enhancing HIF-1α Translation in Triple-Negative Breast Cancer. Front Oncol, 2020, 10:580176., DOI 10.3389/fonc.2020.580176 Baci D, Bosi A, Gallazzi M, Rizzi M, Noonan DM, Poggi A, et al. The Ovarian Cancer Tumor Immune Microenvironment, TIME, as Target for Therapy: A Focus on Innate Immunity Cells as Therapeutic Effectors. Int J Mol Sci, 2020, 21(9):3125., DOI 10.3390/ijms21093125 Odunsi K. Immunotherapy in Ovarian Cancer. Ann Oncol, 2017, 28(Suppl. l_8):viii1–viii7., DOI 10.1093/annonc/mdx444 Ghisoni E, Imbimbo M, Zimmermann S, Valabrega G. Ovarian Cancer Immunotherapy: Turning up the Heat. Int J Mol Sci, 2019, 20(12):2927., DOI 10.3390/ijms20122927 Micek HM, Visetsouk MR, Fleszar AJ, Kreeger PK. The Many Microenvironments of Ovarian Cancer. Adv Exp Med Biol, 2020, 1296: 199–213., DOI 10.1007/978-3-030-59038-3_12 Kalluri R. The Biology and Function of Fibroblasts in Cancer. Nat Rev Cancer, 2016, 16(9):582–98., DOI 10.1038/nrc.2016.73 Li M, Wang J, Wang C, Xia L, Xu J, Xie X, et al. Microenvironment Remodeled by Tumor and Stromal Cells Elevates Fibroblast-Derived COL1A1 and Facilitates Ovarian Cancer Metastasis. Exp Cel Res, 2020, 394(1):112153., DOI 10.1016/j.yexcr.2020.112153 Yamauchi M, Gibbons DL, Zong C, Fradette JJ, Bota-Rabassedas N, Kurie JM. Fibroblast Heterogeneity and its Impact on Extracellular Matrix and Immune Landscape Remodeling in Cancer. Matrix Biol, 2020, 91-92:8–18., DOI 10.1016/ j.matbio.2020.05.001 Givel A-M, Kieffer Y, Scholer-Dahirel A, Sirven P, Cardon M, Pelon F, et al. miR200-regulated CXCL12β Promotes Fibroblast Heterogeneity and Immunosuppression in Ovarian Cancers. Nat Commun, 2018, 9(1):1056., DOI 10.1038/s41467-018-03348-z Hussain A, Voisin V, Poon S, Karamboulas C, Bui NHB, Meens J, et al. Distinct Fibroblast Functional States Drive Clinical Outcomes in Ovarian Cancer and are Regulated by TCF21. J ExpMed, 2020, 217(8):e20191094., DOI 10.1084/jem.20191094 Wang W, Zou W, Liu JR. Tumor-infiltrating T Cells in Epithelial Ovarian Cancer: Predictors of Prognosis and Biological Basis of Immunotherapy. Gynecol Oncol, 2018, 151(1):1–3., DOI 10.1016/j.ygyno.2018.09.005 Edwards SC, Sutton CE, Ladell K, Grant EJ, McLaren JE, Roche F, et al. A Population of Proinflammatory T Cells Coexpresses αβ and γδ T Cell Receptors in Mice and Humans. J Exp Med, 2020, 217(5):e20190834., DOI 10.1084/jem.20190834 Pauza CD, Liou M-L, Lahusen T, Xiao L, Lapidus RG, Cairo C, et al. Gamma Delta T Cell Therapy for Cancer: It Is Good to Be Local. Front Immunol, 2018, 9:1305., DOI 10.3389/fimmu.2018.01305 Morandi F, Yazdanifar M, Cocco C, Bertaina A, Airoldi I. Engineering the Bridge between Innate and Adaptive Immunity for Cancer Immunotherapy: Focus on γδ T and NK Cells. Cells, 2020, 9(8):1757., DOI 10.3390/cells9081757 Wang J, Zhou J-Y, Kho D, Reiners JJ, Wu GS. Role for DUSP1, Dual-specificity Protein Phosphatase 1, in the Regulation of Autophagy. Autophagy, 2016, 12(10):1791–803., DOI 10.1080/15548627.2016.1203483 Zhu J, Zheng Y, Zhang H, Liu Y, Sun H, Zhang P. Galectin-1 Induces Metastasis and Epithelial-Mesenchymal Transition, EMT, in Human Ovarian Cancer Cells via Activation of the MAPK JNK/p38 Signalling Pathway. Am J Transl Res, 2019, 11(6):3862–78. Liu Y, Ding Y, Nie Y, Yang M. EMP1 Promotes the Proliferation and Invasion of Ovarian Cancer Cells through Activating the MAPK Pathway. Onco Targets Ther, 2020, 13:2047–55., DOI 10.2147/ott.s240028 Lv Y, Wang X, Li X, Xu G, Bai Y, Wu J, et al. Nucleotide De Novo Synthesis Increases Breast Cancer Stemness andMetastasis via cGMP-PKG-MAPK Signaling Pathway. Plos Biol, 2020, 18(11):e3000872., DOI 10.1371/journal.pbio.3000872 Wang Z, Zhang C, Chang J, Tian X, Zhu C, XuW. LncRNA EMX2OS, Regulated by TCF12, Interacts with FUS to Regulate the Proliferation,Migration and Invasion of Prostate Cancer Cells through the cGMP-PKG Signaling Pathway. Onco Targets Ther, 2020, 13:7045–56., DOI 10.2147/ott.s243552 Lin W, Ye H. Anticancer Activity of Ursolic Acid on Human Ovarian Cancer Cells via ROS and MMP Mediated Apoptosis, Cell Cycle Arrest and Downregulation of PI3K/AKT Pathway. J BUON, 2020, 25(2):750–6. Wang P, Zhang J, Xiong X, Yuan W, Qin S, Cao W, et al. Icariin Suppresses Cell Cycle Transition and Cell Migration in Ovarian Cancer Cells. Oncol Rep, 2019, 41(4):2321–8., DOI 10.3892/or.2019.6986 Bakhtiari H, Gheysarzadeh A, Ghanadian M, Aghaei M. 15-Hydroxy- 8(17),13(E)-labdadiene-19-carboxylic Acid, HLCA, Inhibits Proliferation and Induces Cell Cycle Arrest and Apoptosis in Ovarian Cancer Cells. Life Sci, 2021, 267:118981., DOI 10.1016/j.lfs.2020.118981 Orr B, Edwards RP. Diagnosis and Treatment of Ovarian Cancer. Hematol Oncol Clin North Am, 2018, 32(6):943–64., DOI 10.1016/j.hoc.2018.07.010 Kang Y, Nagaraja AS, Armaiz-Pena GN, Dorniak PL, Hu W, Rupaimoole R, et al. Adrenergic Stimulation of DUSP1 Impairs Chemotherapy Response in Ovarian Cancer. Clin Cancer Res, 2016, 22(7):1713–24., DOI 10.1158/1078- 0432.ccr-15-1275 Zhou H-H, Chen X, Cai L-Y, Nan X-W, Chen J-H, Chen X-X, et al. Erastin Reverses ABCB1-Mediated Docetaxel Resistance in Ovarian Cancer. Front Oncol, 2019, 9:1398., DOI 10.3389/fonc.2019.01398 Hu Z, Cai M, Zhang Y, Tao L, Guo R. miR-29c-3p Inhibits Autophagy and Cisplatin Resistance in Ovarian Cancer by Regulating FOXP1/ ATG14 Pathway. Cell Cycle, 2020, 19(2):193–206., DOI 10.1080/15384101. 2019.1704537 Hu Y, RanM,Wang B, Lin Y, Cheng Y, Zheng S. Co-Delivery of Docetaxel and Curcumin via Nanomicelles for Enhancing Anti-ovarian Cancer Treatment. Int J Nanomedicine, 2020, 15:9703–15., DOI 10.2147/ijn.s274083 Hosomi Y, Morita S, Sugawara S, Kato T, Fukuhara T, Gemma A, et al. Gefitinib Alone versus Gefitinib Plus Chemotherapy for Non-small-cell Lung Cancer with Mutated Epidermal Growth Factor Receptor: NEJ009 Study. J Clin Oncol, 2020, 38(2):115–23., DOI 10.1200/jco.19.01488 Noronha V, Patil VM, Joshi A, Menon N, Chougule A, Mahajan A, et al. Gefitinib versus Gefitinib Plus Pemetrexed and Carboplatin Chemotherapy in EGFR-Mutated Lung Cancer. J Clin Oncol, 2020, 38(2):124–36., DOI 10.1200/ jco.19.01154 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610404 EP 1610418 DI 10.3389/pore.2022.1610404 PG 15 ER PT J AU Zhou, J Lai, M Ni, Y Li, Sh Zhen, J Du, F Zhang, X Song, Ch Cai, L AF Zhou, Jiangfen Lai, Mingyao Ni, Yang Li, Shaoqun Zhen, Junjie Du, Furong Zhang, Xing Song, Chao Cai, Linbo TI Case Report: Clinicopathological and Genetic Features of IDH-Mutant Brainstem Glioma in Adults: Report of Five Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE prognosis; IDH1 mutation; molecular features; temozolomide; brainstem glioma ID prognosis; IDH1 mutation; molecular features; temozolomide; brainstem glioma AB Adult brainstem gliomas are rare central nervous system tumors that represent a heterogeneous group of tumors. Somatic IDH mutations are uncommon in adult brainstem gliomas and there are few relevant clinical studies. Here, we reported five patients with IDH1 mutations associated with brainstem gliomas, including four cases of IDH1 R132H mutations and one case of R132G mutation. All patients were treated with focal intensity-modulated radiation therapy (IMRT) with concurrent temozolomide (TMZ). One patient died, one relapsed, and three survived to date. All these cases carried a pathogenic variant of TP53, among whom 1 harbored ATRX mutation and 1 had H3K27M mutation. Moreover, we also found some genes related to a worse prognosis, such as CDK4/6 amplification. These findings demonstrate that the specific characteristics of IDHmutant brainstem gliomas should be considered in diagnostic workflows to make therapeutic regimens and improve the prognosis. C1 [Zhou, Jiangfen] Guangdong Sanjiu Brain Hospital, Department of Neuro-OncologyGuangzhou, China. [Lai, Mingyao] Guangdong Sanjiu Brain Hospital, Department of Neuro-OncologyGuangzhou, China. [Ni, Yang] Jiangsu Simcere Diagnostics Co., Ltd., State Key Laboratory of Translational Medicine and Innovative Drug DevelopmentNanjing, China. [Li, Shaoqun] Guangdong Sanjiu Brain Hospital, Department of Neuro-OncologyGuangzhou, China. [Zhen, Junjie] Guangdong Sanjiu Brain Hospital, Department of Neuro-OncologyGuangzhou, China. [Du, Furong] Jiangsu Simcere Diagnostics Co., Ltd., State Key Laboratory of Translational Medicine and Innovative Drug DevelopmentNanjing, China. [Zhang, Xing] Jiangsu Simcere Diagnostics Co., Ltd., State Key Laboratory of Translational Medicine and Innovative Drug DevelopmentNanjing, China. [Song, Chao] Jiangsu Simcere Diagnostics Co., Ltd., State Key Laboratory of Translational Medicine and Innovative Drug DevelopmentNanjing, China. [Cai, Linbo] Guangdong Sanjiu Brain Hospital, Department of Neuro-OncologyGuangzhou, China. RP Cai, L (reprint author), Guangdong Sanjiu Brain Hospital, Department of Neuro-Oncology, Guangzhou, China. EM cailinbogz@126.com CR Ostrom QT, Gittleman H, Fulop J, Liu M, Blanda R, Kromer C, et al. CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2008-2012. Neuro Oncol, 2015, 17(Suppl. 4):iv1–iv62., DOI 10.1093/neuonc/nov189 Eisele SC, Reardon DA. Adult Brainstem Gliomas. Cancer, 2016, 122: 2799–809., DOI 10.1002/cncr.29920 Salmaggi A, Fariselli L, Milanesi I, Lamperti E, SilvAni A, Bizzi A, et al. Natural History and Management of Brainstem Gliomas in Adults. A Retrospective Italian Study. J Neurol, 2008, 255:171–7., DOI 10.1007/s00415-008-0589-0 Johnson DR, Brown PD, Galanis E, Hammack JE. Pilocytic Astrocytoma Survival in Adults: Analysis of the Surveillance, Epidemiology, and End Results Program of the National Cancer Institute. J Neurooncol, 2012, 108:187–93., DOI 10.1007/s11060-012-0829-0 Wang Y, Wang L, Blumcke I, Zhang W, Fu Y, Shan Y, et al. Integrated Genotype-Phenotype Analysis of Long-Term Epilepsy-Associated Ganglioglioma. Brain Pathol, 2022, 32:e13011., DOI 10.1111/bpa.13011 Hartmann C, Meyer J, Balss J, Capper D, Mueller W, Christians A, et al. Type and Frequency of IDH1 and IDH2 Mutations Are Related to Astrocytic and Oligodendroglial Differentiation and Age: a Study of 1, 010 Diffuse Gliomas. Acta Neuropathol, 2009, 118:469–74., DOI 10.1007/s00401-009-0561-9 Carlos-Escalante JA, Gomez-Flores-Ramos L, Bian X, Perdomo-Pantoja A, de Andrade KC, Mejia-Perez SI, et al. Landscape of Germline Genetic Variants in AGT, MGMT, and TP53 in Mexican Adult Patients with Astrocytoma. Cell Mol Neurobiol, 2021, 41:1285–97., DOI 10.1007/s10571-020-00901-7 Wang Z, Rajaraman P, Melin BS, Chung CC, Zhang W, McKean-Cowdin R, et al. Further Confirmation of Germline Glioma Risk Variant Rs78378222 in TP53 and its Implication in Tumor Tissues via Integrative Analysis of TCGA Data. Hum Mutat, 2015, 36:684–8., DOI 10.1002/humu.22799 Aoki K, Nakamura H, Suzuki H, Matsuo K, Kataoka K, Shimamura T, et al. Prognostic Relevance of Genetic Alterations in Diffuse Lower-Grade Gliomas. Neuro Oncol, 2018, 20:66–77., DOI 10.1093/neuonc/nox132 Cao Y, Li X, Kong S, Shang S, Qi Y. CDK4/6 Inhibition Suppresses Tumour Growth and Enhances the Effect of Temozolomide in Glioma Cells. J Cel Mol Med, 2020, 24:5135–45., DOI 10.1111/jcmm.15156 University of Texas Southwestern Medical Center. Pilot Study of Abemaciclib with Bevacizumab in Recurrent Glioblastoma Patients with Loss of CDKN2A/B or Gain or Amplification of CDK4/6. Available online: https://ClinicalTrials. gov/show/NCT04074785, Accessed May 16, 2020). Barrow Neurological Institute. A Phase 0/II Study of Ribociclib, LEE011, in Combination with Everolimus in Preoperative Recurrent High-Grade Glioma Patients Scheduled for Resection. Available online: https://ClinicalTrials.gov/ show/NCT03834740, Accessed May 16, 2020). Taylor JW, Parikh M, Phillips JJ, James CD, Molinaro AM, Butowski NA, et al. Phase-2 Trial of Palbociclib in Adult Patients with Recurrent RB1-Positive Glioblastoma. J Neurooncol, 2018, 140:477–83., DOI 10.1007/s11060-018-2977-3 Wick W, Weller M, van den Bent M, Sanson M, Weiler M, von Deimling A, et al. MGMT Testing-Tthe Challenges for Biomarker-Based Glioma Treatment. Nat Rev Neurol, 2014, 10:372–85., DOI 10.1038/nrneurol.2014.100 Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT Gene Silencing and Benefit from Temozolomide in Glioblastoma. N Engl J Med, 2005, 352:997–1003., DOI 10.1056/NEJMoa043331 Banan R, Stichel D, Bleck A, Hong B, Lehmann U, Suwala A, et al. Infratentorial IDH-Mutant Astrocytoma Is a Distinct Subtype. Acta Neuropathol, 2020, 140:569–81., DOI 10.1007/s00401-020-02194-y Suzuki H, Aoki K, Chiba K, Sato Y, Shiozawa Y, Shiraishi Y, et al. Mutational Landscape and Clonal Architecture in Grade II and III Gliomas. Nat Genet, 2015, 47:458–68., DOI 10.1038/ng.3273 Leeper HE, Caron AA, Decker PA, Jenkins RB, Lachance DH, Giannini C, et al. IDH Mutation, 1p19q Codeletion and ATRX Loss in WHO Grade II Gliomas. Oncotarget, 2015, 6:30295–305., DOI 10.18632/oncotarget.4497 Wiestler B, Capper D, Holland-Letz T, Korshunov A, von Deimling A, Pfister SM, et al. ATRX Loss Refines the Classification of Anaplastic Gliomas and Identifies a Subgroup of IDH Mutant Astrocytic Tumors with Better Prognosis. Acta Neuropathol, 2013, 126:443–51., DOI 10.1007/s00401-013-1156-z Richardson TE, Sathe AA, Kanchwala M, Jia G, Habib AA, Xiao G, et al. Genetic and Epigenetic Features of Rapidly Progressing IDH-Mutant Astrocytomas. J Neuropathol Exp Neurol, 2018, 77:542–8., DOI 10.1093/jnen/nly026 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610408 EP 1610412 DI 10.3389/pore.2022.1610408 PG 5 ER PT J AU Kontsek, E Pesti, A Gordon, P Tornoczki, T Smuk, G Gergely, Sz Kiss, A AF Kontsek, Endre Pesti, Adrian Gordon, Peter Tornoczki, Tamas Smuk, Gabor Gergely, Szilveszter Kiss, Andras TI Mid-Infrared Imaging Characterization to Differentiate Lung Cancer Subtypes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lung cancer; fingerprint region; transflectance; SVM; LDA; FTIR; infrared ID lung cancer; fingerprint region; transflectance; SVM; LDA; FTIR; infrared AB Introduction: Lung cancer is the most common malignancy worldwide. Squamous cell carcinoma (SQ) and adenocarcinoma (LUAD) are the two most frequent histological subtypes. Small cell carcinoma (SCLC) subtype has the worst prognosis. Differential diagnosis is essential for proper oncological treatment. Life science associated mid- and near-infrared based microscopic techniques have been developed exponentially, especially in the past decade. Vibrational spectroscopy is a potential non-destructive approach to investigate malignancies. Aims: Our goal was to differentiate lung cancer subtypes by their label-free mid-infrared spectra using supervised multivariate analyses. Material and Methods: Formalin-fixed paraffin-embedded (FFPE) samples were selected from the archives. Three subtypes were selected for each group: 10- 10 cases SQ, LUAD and SCLC. 2 μm thick sections were cut and laid on aluminium coated glass slides. Transflection optical setup was applied on Perkin-Elmer infrared microscope. 250 × 600 μm areas were imaged and the so-called mid-infrared fingerprint region (1800-648cm−1) was further analysed with linear discriminant analysis (LDA) and support vector machine (SVM) methods. Results: Both “patient-based” and “pixel-based” approaches were examined. Patientbased analysis by using 3 LDA models and 2 SVM models resulted in different separations. The higher the cut-off value the lower is the accuracy. The linear C-support vector classification (C-SVC) SVM resulted in the best (100%) accuracy for the three subtypes using a 50% cut-off value. The pixel-based analysis gave, similarly, the linear C-SVC SVM model to be the most efficient in the statistical indicators (SQ sensitivity 81.65%, LUAD sensitivity 82.89% and SCLC sensitivity 88.89%). The spectra cut-off, the kernel function and the algorithm function influence the accuracy. Conclusion: Mid-Infrared imaging could be used to differentiate FFPE lung cancer subtypes. Supervised multivariate tools are promising to accurately separate lung tumor subtypes. The long-term perspective is to develop a spectroscopy-based diagnostic tool, revolutionizing medical differential diagnostics, especially cancer identification. C1 [Kontsek, Endre] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Pesti, Adrian] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Gordon, Peter] Budapest University of Technology and Economics, Department of Electronics TechnologyBudapest, Hungary. [Tornoczki, Tamas] University of Pecs, Department of PathologyPecs, Hungary. [Smuk, Gabor] University of Pecs, Department of PathologyPecs, Hungary. [Gergely, Szilveszter] Budapest University of Technology and Economics, Department of Applied Biotechnology and Food ScienceBudapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Kontsek, E (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM kontsek.endre@med.semmelweis-univ.hu CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Finlayson D, Rinaldi C, Baker MJ. Is Infrared Spectroscopy Ready for the Clinic? Anal Chem, 2019, 91(19):12117–28., DOI 10.1021/acs.analchem. 9b02280 Fernandes S, Williams G, Williams E, Ehrlich K, Stone J, Finlayson N, et al. Solitary Pulmonary Nodule Imaging Approaches and the Role of Optical Fibre-Based Technologies. Eur Respir J, 2021, 57(3):2002537., DOI 10.1183/ 13993003.02537-2020 Loo PS, Thomas SC, Nicolson MC, Fyfe MN, Kerr KM. Subtyping of Undifferentiated Non-small Cell Carcinomas in Bronchial Biopsy Specimens. J Thorac Oncol, 2010, 5(4):442–7., DOI 10.1097/JTO. 0b013e3181d40fac Rossi G, Pelosi G, Graziano P, BarbareschiM, Papotti M. A Reevaluation of the Clinical Significance of Histological Subtyping of Non-small-cell Lung Carcinoma: Diagnostic Algorithms in the Era of Personalized Treatments. Int J Surg Pathol, 2009, 17(3):206–18., DOI 10.1177/1066896909336178 Su KY, Lee WL. Fourier Transform Infrared Spectroscopy as a Cancer Screening and Diagnostic Tool: A Review and Prospects. Cancers, 2020, 12(1):E115., DOI 10.3390/cancers12010115 Yatabe Y, Dacic S, Borczuk AC, Warth A, Russell PA, Lantuejoul S, et al. Best Practices Recommendations for Diagnostic Immunohistochemistry in Lung Cancer. J Thorac Oncol, 2019, 14(3):377–407., DOI 10.1016/j.jtho.2018.12.005 Balan V, Mihai CT, Cojocaru FD, Uritu CM, Dodi G, Botezat D, et al. Vibrational Spectroscopy Fingerprinting in Medicine: From Molecular to Clinical Practice. Materials, 2019, 12(18):E2884., DOI 10.3390/ma12182884 Baumann WJ, Madson TH, Weseman BJ. “Plasmalogen-type” Cyclic Acetals: Formation and Conformation of the 1, 3-dioxanes and 1, 3-dioxolanes from 1- O-Cis-Alk-1′-Enyl-Sn-Glycerols. J Lipid Res, 1972, 13(5):640–50., DOI 10.1016/ s0022-2275(20)39369-x O’Sullivan A, O’Connor B, Kelly A, McGrath MJ. The Use of Chemical and Infrared Methods for Analysis of Milk and Dairy Products. Int J Dairy Technol, 1999, 52(4):139–48., DOI 10.1111/j.1471-0307.1999.tb02856.x Goulden JDS. Analysis of Milk by Infra-red Absorption. J Dairy Res, 1964, 31(03):273–84., DOI 10.1017/s0022029900018203 GarciaJares CM, Medina B. Application of Multivariate Calibration to the Simultaneous Routine Determination of Ethanol, Glycerol, Fructose, Glucose and Total Residual Sugars in Botrytized-Grape Sweet Wines by Means of Near- Infrared Reflectance Spectroscopy. Fresenius’ J Anal Chem, 1997, 357(1): 86–91., DOI 10.1007/s002160050117 Nieuwoudt HH, Prior BA, Pretorius IS, Manley M, Bauer FF. Principal Component Analysis Applied to Fourier Transform Infrared Spectroscopy for the Design of Calibration Sets for Glycerol Prediction Models in Wine and for the Detection and Classification of Outlier Samples. J Agric Food Chem, 2004, 52(12):3726–35., DOI 10.1021/jf035431q Kays SE, Barton FE, Windham WR. Predicting Protein Content by Near Infrared Reflectance Spectroscopy in Diverse Cereal Food Products. J Near Infrared Spectrosc, 2000, 8(1):35–43., DOI 10.1255/jnirs.262 Gergely S, Salgo A. Changes in Carbohydrate Content during Wheat Maturation - what Is Measured by Near Infrared Spectroscopy? J Near Infrared Spectrosc, 2005, 13(1):9–17., DOI 10.1255/jnirs.452 Huang H, Yu H, Xu H, Ying Y. Near Infrared Spectroscopy for On/in-Line Monitoring of Quality in Foods and Beverages: A Review. J Food Eng, 2008, 87(3):303–13., DOI 10.1016/j.jfoodeng.2007.12.022 Juriga D, Laszlo I, Ludanyi K, Klebovich I, Chae CH, Zrinyi M. Kinetics of Dopamine Release from Poly(aspartamide)-Based Prodrugs. Acta Biomater, 2018, 76:225–38., DOI 10.1016/j.actbio.2018.06.030 Juriga D, Kalman EE, Toth K, Barczikai D, Szollosi D, Foldes A, et al. Analysis of Three-Dimensional Cell Migration in Dopamine-Modified Poly(aspartic Acid)-Based Hydrogels. Gels, 2022, 8(2):65., DOI 10.3390/gels8020065 Bogomolov A, Belikova V, Zabarylo UJ, Bibikova O, Usenov I, Sakharova T, et al. Synergy Effect of Combining Fluorescence and Mid Infrared Fiber Spectroscopy for Kidney Tumor Diagnostics. Sensors, Basel,, 2017, 17(11): E2548., DOI 10.3390/s17112548 Sojka L, Tang Z, Furniss D, Sakr H, Fang Y, Beres-Pawlik E, et al. Mid-infrared Emission in Tb3+-Doped Selenide Glass Fiber. J Opt Soc Am B, 2017, 34(3): A70–A79., DOI 10.1364/josab.34.000a70 Nallala J, Lloyd GR, Hermes M, Shepherd N, Stone N. Enhanced Spectral Histology in the colon Using High-Magnification Benchtop FTIR Imaging. Vibrational Spectrosc, 2017, 91:83–91., DOI 10.1016/j.vibspec.2016.08.013 Seddon AB, Benson TM, Sujecki S, Abdel-Moneim N, Tang ZQ, Furniss D, et al. Towards the Mid-infrared Optical Biopsy. In: Optical Biopsy Xiv: Toward Real-Time Spectroscopic Imaging and Diagnosis; 2016 7 Mar; San Francisco, CA, 2016). p. 9703. Joro R, Laaperi AL, Dastidar P, Soimakallio S, Kuukasjarvi T, Toivonen T, et al. Imaging of Breast Cancer with Mid- and Long-Wave Infrared Camera. J Med Eng Technol, 2008, 32(3):189–97., DOI 10.1080/03091900701234358 Paraskevaidi M, Morais CLM, Lima KMG, Ashton KM, Stringfellow HF, Martin-Hirsch PL, et al. Potential of Mid-infrared Spectroscopy as a Noninvasive Diagnostic Test in Urine for Endometrial or Ovarian Cancer. Analyst, 2018, 143(13):3156–63., DOI 10.1039/c8an00027a Sreedhar H, Varma VK, Nguyen PL, Davidson B, Akkina S, Guzman G, et al. High-definition Fourier Transform Infrared, FT-IR, Spectroscopic Imaging of Human Tissue Sections towards Improving Pathology. J Vis Exp, 2015)(95, 52332., DOI 10.3791/52332 Zohdi V, Whelan DR, Wood BR, Pearson JT, Bambery KR, Black MJ. Importance of Tissue Preparation Methods in FTIR Micro-spectroscopical Analysis of Biological Tissues: ’Traps for New Users. PLoS One, 2015, 10(2): e0116491., DOI 10.1371/journal.pone.0116491 Grosserueschkamp F, Kallenbach-Thieltges A, Behrens T, Bruning T, Altmayer M, Stamatis G, et al. Marker-free Automated Histopathological Annotation of Lung Tumour Subtypes by FTIR Imaging. Analyst, 2015, 140(7):2114–20., DOI 10.1039/c4an01978d Gaydou V, Polette M, Gobinet C, Kileztky C, Angiboust JF, Birembaut P, et al. New Insights into Spectral Histopathology: Infrared-Based Scoring of Tumour Aggressiveness of Squamous Cell Lung Carcinomas. Chem Sci, 2019, 10(15): 4246–58., DOI 10.1039/c8sc04320e Akalin A, Mu X, Kon MA, Ergin A, Remiszewski SH, Thompson CM, et al. Classification of Malignant and Benign Tumors of the Lung by Infrared Spectral Histopathology, SHP). Lab Invest, 2015, 95(4):697–21., DOI 10. 1038/labinvest.2015.55 Le Naour F, Bralet MP, Debois D, Sandt C, Guettier C, Dumas P, et al. Chemical Imaging on Liver Steatosis Using Synchrotron Infrared and ToFSIMS Microspectroscopies. Plos One, 2009, 4(10):e7408., DOI 10.1371/journal. pone.0007408 Kontsek E, Pesti A, Bjornstedt M, Uveges T, Szabo E, Garay T, et al. Mid- Infrared Imaging is Able to Characterize and Separate Cancer Cell Lines. Pathol Oncol Res, 2020, 26(4):2401–7., DOI 10.1007/s12253-020-00825-z James G, Witten D, Hastie T, Tibshirani R. Classification. In: An Introduction to Statistical Learning: With Applications in R. New York, NY: Springer US, 2021). p. 129–95. Paech DC, Weston AR, Pavlakis N, Gill A, Rajan N, Barraclough H, et al. A Systematic Review of the Interobserver Variability for Histology in the Differentiation between Squamous and Nonsquamous Non-small Cell Lung Cancer. J Thorac Oncol, 2011, 6(1):55–63., DOI 10.1097/JTO. 0b013e3181fc0878 Irwig L, Macaskill P, Farnsworth A, Wright RG, McCool J, Barratt A, et al. A Randomized Crossover Trial of PAPNET for Primary Cervical Screening. J Clin Epidemiol, 2004, 57(1):75–81., DOI 10.1016/S0895-4356(03)00259-2 Phelps DL, Balog J, Gildea LF, Bodai Z, Savage A, El-Bahrawy MA, et al. The Surgical Intelligent Knife Distinguishes normal, Borderline and Malignant Gynaecological Tissues Using Rapid Evaporative Ionisation Mass Spectrometry, REIMS). Br J Cancer, 2018, 118(10):1349–58., DOI 10.1038/ s41416-018-0048-3 Petersen D, Mavarani L, Niedieker D, Freier E, Tannapfel A, Kotting C, et al. Virtual Staining of colon Cancer Tissue by Label-free Raman Microspectroscopy. Analyst, 2017, 142(8):1207–15., DOI 10.1039/c6an02072k Tian P, Wang Y, Li L, Zhou Y, Luo W, Li W. CT-Guided Transthoracic Core Needle Biopsy for Small Pulmonary Lesions: Diagnostic Performance and Adequacy for Molecular Testing. J Thorac Dis, 2017, 9(2):333–43., DOI 10. 21037/jtd.2017.02.16 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610439 EP 1610446 DI 10.3389/pore.2022.1610439 PG 8 ER PT J AU Truszkowska, E Andrzejewska, M Szymanska, C Wziatek, A Derwich, K AF Truszkowska, Ewelina Andrzejewska, Marta Szymanska, Cyntia Wziatek, Agnieszka Derwich, Katarzyna TI Case Report: Brentuximab Vedotin Associated Acute Pancreatitis in a Pediatric Hodgkin Lymphoma Patient: Case Report and Literature Review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Hodgkin lymphoma; pediatric oncology; brentuximab vedotin; acute pancreatitis; side effects ID Hodgkin lymphoma; pediatric oncology; brentuximab vedotin; acute pancreatitis; side effects AB Brentuximab vedotin is a conjugate drug used mainly in Hodgkin lymphoma, systemic and primary cutaneous anaplastic large cell lymphomas, and CD30-expressing peripheral T-cell lymphoma. We report a unique case of acute pancreatitis associated with brentuximab vedotin in a 17-year-old male patient suffering from classical Hodgkin lymphoma. Diagnosed in 2020, the patient was classified to an intermediate therapeutic group and disease’s grade was IIIAE. The patient was treated with brentuximab vedotin and bendamustine in the third line. Two weeks after the drug administration, the patient developed acute epigastric pain. Laboratory and radiological findings confirmed the clinical suspicion of acute pancreatitis that was managed with opioid pain medications, meropenem, parenteral nutrition, ondansetron and omeprazole. This is the first case report of brentuximab vedotin-associated acute pancreatitis in the pediatric patient reported in the literature to the best of our knowledge. C1 [Truszkowska, Ewelina] Poznan University of Medical Sciences, Faculty of MedicinePoznan, Poland. [Andrzejewska, Marta] Poznan University of Medical Sciences, Faculty of MedicinePoznan, Poland. [Szymanska, Cyntia] Poznan University of Medical Sciences, Faculty of MedicinePoznan, Poland. [Wziatek, Agnieszka] Poznan University of Medical Sciences, Institute of Pediatrics, Department of Pediatric Oncology, Hematology and TransplantologyPoznan, Poland. [Derwich, Katarzyna] Poznan University of Medical Sciences, Institute of Pediatrics, Department of Pediatric Oncology, Hematology and TransplantologyPoznan, Poland. RP Derwich, K (reprint author), Poznan University of Medical Sciences, Institute of Pediatrics, Department of Pediatric Oncology, Hematology and Transplantology, Poznan, Poland. EM kderwich@ump.edu.pl CR Miller KD, Fidler-Benaoudia M, Keegan TH, Hipp HS, Jemal A, Siegel RL, et al. Cancer Statistics for Adolescents and Young Adults, 2020. CA Cancer J Clin, 2020, 443:6–59., DOI 10.3322/caac.21637 Daw S, Hasenclever D, Mascarin M, Fernandez-Teijeiro A, Balwierz W, Beishuizen A, et al. Risk and Response Adapted Treatment Guidelines for Managing First Relapsed and Refractory Classical Hodgkin Lymphoma in Children and Young People. Recommendations from the EuroNet Pediatric Hodgkin Lymphoma Group. HemaSphere, 2020, 4:e329., DOI 10.1097/HS9. 0000000000000329 Smith RS,ChenQ,HudsonMM, LinkMP,KunM, WeinsteinH, et al. Prognostic Factors for Children with Hodgkin’s Disease Treated with Combined-Modality Therapy. J Clin Oncol, 2003, 21:2026–33., DOI 10.1200/JCO.2003.07.124 Schwartz CL, Chen L, McCarten K, Wolden S, Constine LS, Hutchison RE, et al. Childhood Hodgkin International Prognostic Score, CHIPS, Predicts Event-free Survival in Hodgkin Lymphoma: A Report from the Children’s Oncology Group. Pediatr Blood Cancer, 2017, 64(4):e26278., DOI 10.1002/pbc. 2627810.1002/pbc.26278 McCarten KM, Metzger ML, Drachtman RA, Pei Q, Friedman DL, Schwartz CL, et al. Significance of Pleural Effusion at Diagnosis in Pediatric Hodgkin Lymphoma: A Report from Children’s Oncology Group Protocol AHOD0031. Pediatr Radiol, 2018, 48(12):1736–44., DOI 10.1007/s00247-018-4197-6 Marks LJ, McCarten KM, Pei Q, Friedman DL, Schwartz CL, Kelly KM, et al. Pericardial Effusion in Hodgkin Lymphoma: A Report from the Children’s Oncology Group AHOD0031 Protocol. Blood, 2018, 132(11):1208–11., DOI 10. 1182/blood-2018-02-834465 Hutchings M, Loft A, Hansen M, Pedersen LM, Buhl T, Jurlander J, et al. FDGPET after Two Cycles of Chemotherapy Predicts Treatment Failure and Progression-free Survival in Hodgkin Lymphoma. Blood, 2006, 107(1): 52–9., DOI 10.1182/blood-2005-06-2252 Joseph PD, Craig JC, Caldwell PH. Clinical Trials in Children. Br J Clin Pharmacol, 2015, 79:357–69., DOI 10.1111/bcp.12305 Bazarbachi A, Boumendil A, Finel H, Mohty M, Castagna L, Blaise D, et al. Brentuximab Vedotin for Recurrent Hodgkin Lymphoma after Allogeneic Hematopoietic Stem Cell Transplantation: A Report from the EBMT Lymphoma Working Party. Cancer, 2019, 125:90–8., DOI 10.1002/cncr.31755 O’Connor OA, Lue JK, Sawas A, Amengual JE, Deng C, Kalac M, et al. Brentuximab Vedotin Plus Bendamustine in Relapsed or Refractory Hodgkin’s Lymphoma: an International, Multicentre, Single-Arm, Phase 1-2 trialErratum in. Lancet Oncol, 2018, 1919(23):257e137–66., DOI 10.1016/S1470-2045(17, 30912-9 Cole PD, McCarten KM, Pei Q, Spira M, Metzger ML, Drachtman RA, et al. Brentuximab Vedotin with Gemcitabine for Paediatric and Young Adult Patients with Relapsed or Refractory Hodgkin’s Lymphoma, AHOD1221): A Children’s Oncology Group, Multicentre Single-Arm, Phase 1–2 Trial. Lancet Oncol, 2018, 19(9):1229–38., DOI 10.1016/S1470-2045(18)30426-1 Locatelli F, Mauz-Koerholz C, Neville K, Llort A, Beishuizen A, Daw S, et al. Brentuximab Vedotin for Paediatric Relapsed or Refractory Hodgkin’s Lymphoma and Anaplastic Large-Cell Lymphoma: a Multicentre, Open- Label, Phase 1/2 Study. Lancet Haematol, 2018, 5:e450–61., DOI 10.1016/ S2352-3026(18)30153-4 Maradana S, Akella P, Nalluru SS, Jindal V, Siddiqui AD. Hypertriglyceridemia Induced Pancreatitis Due to Brentuximab Therapy: First Case Report. Cureus, 2019, 11(7):e5138., DOI 10.7759/cureus.5138 Urru SAM, Mariotti E, Carta P, Massidda S, Marcias M, Murru R, et al. Acute Pancreatitis Following Brentuximab Vedotin Therapy for Refractory Hodgkin Lymphoma: A Case Report. Drugs R D, 2014, 14:9–11., DOI 10.1007/s40268- 014-0036-x Gandhi MD, Evens AM, Fenske TS, Hamlin P, Coiffier B, Engert A, et al. Pancreatitis in Patients Treated with Brentuximab Vedotin: A Previously Unrecognized Serious Adverse Event. Blood, 2014, 123(18):2895–7., DOI 10. 1182/blood-2014-03-561878 Vinti L, Locatelli F, Merli P, Parasole R, Buffardi S, Pillon M, et al. Brentuximab Vedotin in Combination with Bendamustine in Relapsed or Refractory Hodgkin Lymphoma: A Retrospective Analysis on 23 Paediatric Patients or Young Adults. Blood, 2017, 130:1., DOI 10.1182/blood.V130.Suppl_1.4090.4090 McMillan A, O’Neil AT, Townsend W, Lambert J, Virchi A, Shah R, et al. The Addition of Bendamustine to Brentuximab Vedotin Leads to Improved Rates of Complete Metabolic Remission in Children, Adolescents and Young Adults with Relapsed and Refractory Classical Hodgkin Lymphoma: A Retrospective Single-centre Series. Br J Haematol, 2021, 192:e84–e87., DOI 10.1111/bjh.17274 Korholz D, Wallace WH, Landman-Parker J. Euro-net-Paediatric Hodgkin’s Lymphoma Group, Euro-Net-PHL-C2): Second International Inter-group Study for Classical Hodgkin Lymphoma in Children and Adolescents, 2015). Available at: https://clinicaltrials.gov/ct2/show/NCT02684708, Accessed February 20, 2022). Suri A, Mould DR, Song G, Kinley J, Venkatakrishnan K. Population Pharmacokinetics of Brentuximab Vedotin in Adult and Pediatric Patients with Relapsed/Refractory Hematologic Malignancies: Model-Informed Hypothesis Generation for Pediatric Dosing Regimens. J Clin Pharmacol, 2020, 60(12):1585–97., DOI 10.1002/jcph.1682 Pang Y, Kartsonaki C, Turnbull I, Guo Y, Yang L, Bian Z, et al. Metabolic and Lifestyle Risk Factors for Acute Pancreatitis in Chinese Adults: A Prospective Cohort Study of 0.5 Million People. Plos Med, 2018, 15(8):e1002618., DOI 10. 1371/journal.pmed.1002618 Aune D,Mahamat-Saleh Y, Norat T, Riboli E. High BodyMass Index and Central Adiposity Is Associated with Increased Risk of Acute Pancreatitis: AMeta-Analysis. Dig Dis Sci, 2021, 66(4):1249–67., DOI 10.1007/s10620-020-06275-6 Li X, Guo X, Ji H, Niu J, Gao P. Relationships between Metabolic Comorbidities and Occurrence, Severity, and Outcomes in Patients with Acute Pancreatitis: A Narrative Review. Biomed Res Int, 2019, 2019: 2645926., DOI 10.1155/2019/2645926 Wang SQ, Shu-jun L, Quan-xin F, Xiang-ying F, Xu L, Zhao QC, et al. Overweight Is an Additional Prognostic Factor in Acute Pancreatitis: A Meta- Analysis. Pancreatology, 2011, 11(2):92–8., DOI 10.1159/000327688 Evens AM, Advani RH, Helenowski IB, Fanale M, Smith SM, Jovanovic BD, et al. Multicenter Phase II Study of Sequential Brentuximab Vedotin and Doxorubicin, Vinblastine, and Dacarbazine Chemotherapy for Older Patients with Untreated Classical Hodgkin Lymphoma. J Clin Oncol, 2018, 36(30): 3015–22., DOI 10.1200/JCO.2018.79.0139 Ching Chen C, Su-Peng Y. Fatal Pancreatitis Occurred in a Patient with Refractory CD30+ Anaplastic Large Cell Lymphoma after Brentuximab Vedotin Treatment. J Cancer Res Pract, 2017, 4(1):35–7., DOI 10.1016/j. jcrpr.2016.09.002 Anderson K, Shehata M, Singh D, Al-Ourani M. Acute Pancreatitis Induced by Polatuzumab-Vedotin-Piiq in Combination with Bendamustine and Rituximab for Diffuse Large B-Cell Lymphoma. Cureus, 2020, 12(9): e10299., DOI 10.7759/cureus.10299 Whitcomb DC. Genetic Risk Factors for Pancreatic Disorders. Gastroenterology, 2013, 144(6):1292–302., DOI 10.1053/j.gastro.2013.01.069 Tenner S. Drug-Induced Acute Pancreatitis: Underdiagnosis and Overdiagnosis. Dig Dis Sci, 2010, 55:2706–8., DOI 10.1007/s10620-010-1367-2 Stefanovic M, Jazbec J, Lindgren F, Bulajic M, Lohr M. Acute Pancreatitis as a Complication of Childhood Cancer Treatment.. Cancer Med, 2016, 5(5): 827–36., DOI 10.1002/cam4.649 Raja RA, Schmiegelow K, Frandsen TL. Asparaginase-associated Pancreatitis in Children. Br J Haematol, 2012, 159:18–27., DOI 10.1111/bjh.12016 Hijiya N, van der Sluis IM. Asparaginase-associated Toxicity in Children with Acute Lymphoblastic Leukemia. Leuk Lymphoma, 2016, 57(4):748–57., DOI 10. 3109/10428194.2015.1101098 Teuffel O, Kuster SP, Hunger SP, Conter V, Hitzler J, Ethier MC, et al. Dexamethasone versus Prednisone for Induction Therapy in Childhood Acute Lymphoblastic Leukemia: A Systematic Review and Meta-Analysis. Leukemia, 2011, 25(8):1232–8., DOI 10.1038/leu.2011.84 Kearney SL, DahlbergLevy SEDE, Voss SD, Sallan SE, Silverman LB. Clinical Course and Outcome in Children with Acute Lymphoblastic Leukemia and Asparaginase-Associated Pancreatitis. Pediatr Blood Cancer, 2009, 53:162–7., DOI 10.1002/pbc.22076 Junquera Alonso E, Seoane Blanco L, Cano Calderero FX. Bortezomib-induced Acute Pancreatitis, an Uncommon Adverse Event. Rev Esp Enferm Dig, 2021, 113(1):77., DOI 10.17235/reed.2020.7120/2020 Brulc E, Seehaus C, Schutz N, Fantl D. Management of Pancreatitis Related to Bortezomib Treatment: Report of Two Cases. Hematol Transfus Cel Ther, 2018, 40(4):382–4., DOI 10.1016/j.htct.2018.03.003 Halalsheh H, Bazzeh F, Alkayed K, Salami K, Madanat F. 6-Mercaptopurineinduced Recurrent Acute Pancreatitis in Children with Acute Lymphoblastic Leukemia/Lymphoma. J Pediatr Hematol Oncol, 2013, 35(6):470–2., DOI 10. 1097/mph.0b013e318271c92f Zerra P, Bergsagel J, Keller FG, Glen L, Pauly M. Maintenance Treatment with Low-Dose Mercaptopurine in Combination with Allopurinol in Children with Acute Lymphoblastic Leukemia and Mercaptopurine- Induced Pancreatitis. Pediatr Blood Cancer, 2016, 63(4):712–5., DOI 10. 1002/pbc.25841 McGrail LH, Sehn LH, Weiss RB, Robson MR, Antin JH, Byrd JC, et al. Pancreatitis during Therapy of Acute Myeloid Leukemia: Cytarabine Related? Ann Oncol, 1999, 10(11):1373–6., DOI 10.1023/a:1008342320532 Sastry J, Young S, Shaw P. Acute Pancreatitis Due to Tacrolimus in a Case of Allogeneic Bone Marrow Transplantation. Bone Marrow Transpl, 2004, 33: 867–8., DOI 10.1038/sj.bmt.1704429 Xu J, Xu L, Wei X, Li X, Cai M. A Case Report: Acute Pancreatitis Associated with Tacrolimus in Kidney Transplantation. BMC Nephrol, 2019, 20:209., DOI 10.1186/s12882-019-1395-x Stock W, Douer D, DeAngelo DJ, Arellano M, Advani A, Damon L, et al. Prevention and Management of Asparaginase/pegasparaginase-Associated Toxicities in Adults and Older Adolescents: Recommendations of an Expert Panel. Leuk Lymphoma, 2011, 52(12):2237–53., DOI 10.3109/ 10428194.2011.596963 Abu-El-Haija M, Kumar S, Quiros JA, Balakrishnan K, Barth B, Bitton S, et al. Management of Acute Pancreatitis in the Pediatric Population: A Clinical Report From the North American Society for Pediatric Gastroenterology, Hepatology and Nutrition Pancreas Committee. J Pediatr Gastroenterol Nutr, 2018, 66(1):159–76., DOI 10.1097/MPG.0000000000001715 Isenmann R, Runzi M, Kron M, Kahl S, Kraus D, Jung N, et al. Prophylactic Antibiotic Treatment in Patients with Predicted Severe Acute Pancreatitis: A Placebo-Controlled, Double-Blind Trial. Gastroenterology, 2004, 126(4): 997–1004., DOI 10.1053/j.gastro.2003.12.050 Garcia-Barrasa A, Borobia FG, Pallares R, Jorba R, Poves I, Busquets J, et al. A Double-Blind, Placebo-Controlled Trial of Ciprofloxacin Prophylaxis in Patients with Acute Necrotizing Pancreatitis. J Gastrointest Surg, 2009, 13(4): 768–74., DOI 10.1007/s11605-008-0773-7 Dambrauskas Z, Gulbinas A, Pundzius J, Barauskas G. Meta-analysis of Prophylactic Parenteral Antibiotic Use in Acute Necrotizing Pancreatitis. Medicina, Kaunas,, 2007, 43(4):291., DOI 10.3390/medicina43040036 Thu Huynh V, Bergeron S. Asparaginase Toxicities: Identification and Management in Patients with Acute Lymphoblastic Leukemia. Clin J Oncol Nurs, 2017, 21(5):E248–E259., DOI 10.1188/17.CJON.E248-E259 Burke PW, Hoelzer D, Park JH, Schmiegelow K, Douer D. Managing Toxicities with Asparaginase Based Therapies in Adult ALL: Summary of an ESMO Open– Cancer Horizons Roundtable Discussion. ESMO Open, 2020, 5: e000858., DOI 10.1136/esmoopen-2020-000858 Oak E, Bartlett NL. A Safety Evaluation of Brentuximab Vedotin for the Treatment of Hodgkin Lymphoma. Expert Opin Drug Saf, 2016, 15:875–82., DOI 10.1080/14740338.2016.1179277 Carson KR, Newsome SD, Kim EJ, Wagner-Johnston ND, von Geldern G, Moskowitz CH, et al. Progressive Multifocal Leukoencephalopathy Associated with Brentuximab Vedotin Therapy: A Report of 5 Cases from the Southern Network on Adverse Reactions, SONAR, Project. Cancer, 2014, 120:2464–71., DOI 10.1002/cncr.28712 Berger JR, Aksamit AJ, Clifford DB, Davis L, Koralnik IJ, Sejvar JJ, et al. PML Diagnostic Criteria: Consensus Statement from the AAN Neuroinfectious Disease Section. Neurology, 2013, 80(15):1430–8., DOI 10.1212/WNL.0b013e31828c2fa1 EMA Europa. Adcetris. [Internet], 2022). Available from: https://www.ema.europa. eu/en/documents/product-information/adcetris-epar-product-information_en.pdf, Accessed February 23, 2022). NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610445 EP 1610451 DI 10.3389/pore.2022.1610445 PG 7 ER PT J AU Kawasaki, Y Suzuki, H Suzuki, Sh Yamada, T Suzuki, M Ito, A Hatakeyama, H Miura, M Omori, Y AF Kawasaki, Yohei Suzuki, Hitomi Suzuki, Shinsuke Yamada, Takechiyo Suzuki, Maya Ito, Ayumi Hatakeyama, Haruka Miura, Masahito Omori, Yasufumi TI GPNMB-Positive Cells in Head and Neck Squamous Cell Carcinoma —Their Roles in Cancer Stemness, Therapy Resistance, and Metastasis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE epithelial-mesenchymal transition; cancer stem cells; radiotherapy; head and neck squamous cell carcinoma; glycoprotein nonmetastatic melanoma protein B; prognostic factor ID epithelial-mesenchymal transition; cancer stem cells; radiotherapy; head and neck squamous cell carcinoma; glycoprotein nonmetastatic melanoma protein B; prognostic factor AB Objective: Despite the use of surgical and chemoradiation therapies, head and neck squamous cell carcinoma (HNSCC) still has a poor prognosis. Immune checkpoint inhibitors have been shown to prolong life expectancy but have limited efficacy. Glycoprotein nonmetastatic melanoma protein B (GPNMB) has received significant attention in breast cancer treatment, in which it has been associated with cancer stem cells (CSCs) and epithelial-mesenchymal transition (EMT); however, the function of GPNMB in HNSCC is completely unknown. This study aimed to clarify the characteristics of GPNMB-positive cells in vitro and their association with the prognosis by immunostaining clinical specimens. Methods: We examined the sphere formation, invasion, and migration ability of GPNMBpositive cells in four HNSCC cell lines in vitro. We also immunostained biopsy specimens with GPNMB from 174 patients with HNSCC diagnosed, treated, and followed-up in our institution to evaluate overall survival and progression-free survival. Results: GPNMB-positive cells showed enhanced sphere formation, invasion, and migration, suggesting that they could have CSC characteristics and the ability to induce EMT, as reported for breast cancer. Clinical specimens showed that overall survival was 39.4% and 57.8% (p = 0.045) and that progression-free survival was 27.6% and 51.6% (p = 0.013) for the high-expression and the low-expression groups, respectively, indicating poor prognosis for the high GPNMB group. The high GPNMB group was also more resistant to chemoradiation and bioradiotherapy. GPNMB was more highly expressed in metastatic lymph nodes than in the primary tumor. Conclusion: GPNMB-positive cells might have CSC characteristics and induce EMT. Detailed functional analyses of GPNMB in HNSCC and the establishment of therapies targeting GPNMB will lead to improved prognoses. C1 [Kawasaki, Yohei] Akita University Graduate School of Medicine, Department of Otorhinolaryngology and Head-and-Neck SurgeryAkita, Japan. [Suzuki, Hitomi] Akita University Graduate School of Medicine, Department of Otorhinolaryngology and Head-and-Neck SurgeryAkita, Japan. [Suzuki, Shinsuke] Akita University Graduate School of Medicine, Department of Otorhinolaryngology and Head-and-Neck SurgeryAkita, Japan. [Yamada, Takechiyo] Akita University Graduate School of Medicine, Department of Otorhinolaryngology and Head-and-Neck SurgeryAkita, Japan. [Suzuki, Maya] Akita University Graduate School of Medicine, Department of Molecular and Tumour PathologyAkita, Japan. [Ito, Ayumi] Akita University Graduate School of Medicine, Department of Molecular and Tumour PathologyAkita, Japan. [Hatakeyama, Haruka] Akita University Graduate School of Medicine, Department of Molecular and Tumour PathologyAkita, Japan. [Miura, Masahito] Akita University Graduate School of Medicine, Department of Molecular and Tumour PathologyAkita, Japan. [Omori, Yasufumi] Akita University Graduate School of Medicine, Department of Molecular and Tumour PathologyAkita, Japan. RP Omori, Y (reprint author), Akita University Graduate School of Medicine, Department of Molecular and Tumour Pathology, Akita, Japan. EM yasu@med.akita-u.ac.jp CR Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer Incidence and Mortality Worldwide: Sources, Methods and Major Patterns in GlOBOCAN 2012. Int J Cancer, 2015, 136:E359–86., DOI 10.1002/ijc.29210 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A, et al. Global Cancer Statistics, 2012. CA Cancer J Clin, 2015, 65:87–108., DOI 10.3322/caac. 21262 Pignon JP, le Maitre A, Maillard E, Bourhis J, MACH-NC Collaborative Group. Meta-analysis of Chemotherapy in Head and Neck Cancer, MACHNC): An Update on 93 Randomised Trials and 17, 346 Patients. Radiother Oncol, 2009, 92:4–14., DOI 10.1016/j.radonc.2009.04.014 Ripoll VM, Meadows NA, Raggatt LJ, Chang MK, Pettit AR, Cassady AI, et al. Microphthalmia Transcription Factor Regulates the Expression of the Novel Osteoclast Factor GPNMB. Gene, 2008, 413:32–41., DOI 10.1016/j.gene.2008. 01.014 Sheng MH, Wegedal JE, Mohan S, Amoui M, Baylink DJ, Lau KH. Targeted Overexpression of Osteoactivin in Cells of Osteoclastic Lineage Promotes Osteoclastic Resorption and Bone Loss in Mice. PLoS One, 2012, 7:e35280., DOI 10.1371/journal.pone.0035280 Rich JN, Shi Q, Hjelmeland M, Cummings TJ, Kuan CT, Bigner DD, et al. Bone-related Genes Expressed in Advanced Malignancies Induce Invasion and Metastasis in a Genetically Defined Human Cancer Model. J Biol Chem, 2003, 278:15951–7., DOI 10.1074/jbc.M211498200 Tian F, Liu C, Wu Q, Qu K, Wang R, Wei J, et al. Upregulation of Glycoprotein Nonmetastatic B by colony-stimulating Factor-1 and Epithelial Cell Adhesion Molecule in Hepatocellular Carcinoma Cells. Oncol Res, 2013, 20:341–50., DOI 10.3727/096504013X13657689382851 Eldai H, Periyasamy S, Al Qarni S, Al Rodayyan M, Muhammed Mustafa S, Deeb A, et al. Novel Genes Associated with Colorectal Cancer are Revealed by High Resolution Cytogenetic Analysis in a Patient Specific Manner. PLoS One, 2013, 8:e76251., DOI 10.1371/journal.pone.0076251 Williams MD, Esmaeli B, Soheili A, Simantov R, Gombos DS, Bedikian AY, et al. GPNMB Expression in Uveal Melanoma: A Potential for Targeted Therapy. Melanoma Res, 2010, 20:184–90., DOI 10.1097/CMR. 0b013e3283364a08 Zhao Y, Qiao ZG, Shan SJ, Sun QM, Zhang JZ. Expression of Glycoprotein Non-metastatic Melanoma Protein B in Cutaneous Malignant and Benign Lesions: a Tissue Microarray Study. Chin Med J, Engl,, 2012, 125:3279–82., DOI 10.3760/cma.j.issn.0366-6999.2012.18.015 Okita Y, Chen C, Kato M. Cell-surface GPNMB and Induction of Stemness. Oncotarget, 2018, 9:37289–90., DOI 10.18632/oncotarget.26472 Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The Epithelial-Mesenchymal Transition Generates Cells with Properties of Stem Cells. Cell, 2008, 133:704–15., DOI 10.1016/j.cell.2008.03.027 Chen C, Okita Y, Watanabe Y, Abe F, Fikry MA, Ichikawa Y, et al. Glycoprotein Nmb Is Exposed on the Surface of Dormant Breast Cancer Cells and Induces Stem Cell-like Properties. Cancer Res, 2018, 78:6424–35., DOI 10.1158/0008-5472.CAN-18-0599 Li H, Xiao Y, Wu CC, Yang LL, Cao LY, Chen DR, et al. High Expression of GPNMB Predicts Poor Prognosis in Head and Neck Squamous Cell Carcinoma. Histol Histopathol, 2019, 34:803–10., DOI 10.14670/HH-18-084 Arosarena OA, Dela Cadena RA, Denny MF, Bryant E, Barr EW, Thorpe R, et al. Osteoactivin Promotes Migration of Oral Squamous Cell Carcinomas. J Cel Physiol, 2016, 231:1761–70., DOI 10.1002/jcp.25279 Li Q, Omori Y, Nishikawa Y, Yoshioka T, Yamamoto Y, Enomoto K. Cytoplasmic Accumulation of Connexin32 Protein Enhances Motility and Metastatic Ability ofHuman Hepatoma Cells In Vitro and In Vivo. Int J Cancer, 2007, 121:536–46., DOI 10.1002/ijc.22696 Rietbergen MM, Martens-de Kemp SR, Bloemena E, Witte BI, Brink A, Baatenburg de Jong RJ, et al. Cancer Stem Cell Enrichment Marker CD98: A Prognostic Factor for Survival in Patients with Human Papillomavirus- Positive Oropharyngeal Cancer. Eur J Cancer, 2014, 50:765–73., DOI 10.1016/j. ejca.2013.11.010 Kawasaki Y, Omori Y, Li Q, Nishikawa Y, Yoshioka T, Yoshida M, et al. Cytoplasmic Accumulation of Connexin32 Expands Cancer Stem Cell Population in Human HuH7 Hepatoma Cells by Enhancing its Self- Renewal. Int J Cancer, 2011, 128:51–62., DOI 10.1002/ijc.25308 Kawasaki Y, Omori Y, Suzuki S, Yamada T. CD98hc as a Marker of Radiotherapy-Resistant Cancer Stem Cells in Head and Neck Squamous Cell Carcinoma. Arch Med Sci, 2020)., DOI 10.5114/aoms.2020.92872 Okita Y, Kimura M, Xie R, Chen C, Shen LT, Kojima Y, et al. The Transcription Factor MAFK Induces EMT and Malignant Progression of Triple-Negative Breast Cancer Cells through its Target GPNMB. Sci Signal, 2017, 10:eaak9397., DOI 10.1126/scisignal.aak9397 Huang R, Rofstad EK. Cancer Stem Cells, CSCs), Cervical CSCs and Targeted Therapies. Oncotarget, 2017, 8:35351–67., DOI 10.18632/oncotarget.10169 Fiorentini C, Bodei S, Bedussi F, Fragni M, Bonini SA, Simeone C, et al. GPNMB/OA Protein Increases the Invasiveness of HumanMetastatic Prostate Cancer Cell Lines DU145 and PC3 through MMP-2 and MMP-9 Activity. Exp Cel Res, 2014, 323:100–11., DOI 10.1016/j.yexcr.2014.02.025 Ramani V, Teshima T, Tamura K, Chung JS, Kobayashi M, Cruz PD, et al. Melanoma-derived Soluble DC-HIL/GPNMB Promotes Metastasis by Excluding T-Lymphocytes from the Pre-metastatic Niches. J Invest Dermatol, 2018, 138:2443–51., DOI 10.1016/j.jid.2018.05.012 Bhattacharyya S, Feferman L, Sharma G, Tobacman JK. Increased GPNMB, Phospho-ERK1/2, and MMP-9 in Cystic Fibrosis in Association with Reduced Arylsulfatase B. Mol Genet Metab, 2018, 124:168–75., DOI 10.1016/j.ymgme. 2018.02.012 Arosarena OA, Barr EW, Thorpe R, Yankey H, Tarr JT, Safadi FF. Osteoactivin Regulates Head and Neck Squamous Cell Carcinoma Invasion by Modulating Matrix Metalloproteases. J Cel Physiol, 2018, 233:409–21., DOI 10.1002/jcp. 25900 Zhang Q, He Y, Luo N, Patel SJ, Han Y, Gao R, et al. Landscape and Dynamics of Single Immune Cells in Hepatocellular Carcinoma. Cell, 2019, 179:829–45., DOI 10.1016/j.cell.2019.10.003 Ma RQ, Tang ZJ, Ye X, Cheng HY, Sun KK, Chang XH, et al. Overexpression of GPNMB Predicts an Unfavorable Outcome of Epithelial Ovarian Cancer. Arch Gynecol Obstet, 2018, 297:1235–44., DOI 10.1007/s00404-018-4699-3 Feng X, Zhang L, Ke S, Liu T, Hao L, Zhao P, et al. High Expression of GPNMB Indicates an Unfavorable Prognosis in Glioma: Combination of Data from the GEO and CGGA Databases and Validation in Tissue Microarray. Oncol Lett, 2020, 20:2356–68., DOI 10.3892/ol.2020.11787 Huang YH, Chu PY, Chen JL, Huang CT, Huang CC, Tsai YF, et al. Expression Pattern and Prognostic Impact of Glycoprotein Non-metastatic B, GPNMB, in Triple-Negative Breast Cancer. Sci Rep, 2021, 11:12171., DOI 10.1038/ s41598-021-91588-3 Zhai JP, Liu ZH, Wang HD, Huang GL, Man LB. GPNMB Overexpression is Associated with Extensive Bone Metastasis and Poor Prognosis in Renal Cell Carcinoma. Oncol Lett, 2022, 23:36., DOI 10.3892/ol.2021.13154 Marie-Egyptienne DT, Lohse I, Hill RP. Cancer Stem Cells, the Epithelial to Mesenchymal Transition, EMT, and Radioresistance: Potential Role of Hypoxia. Cancer Lett, 2013, 341:63–72., DOI 10.1016/j.canlet.2012.11.019 Kopp LM, Malempati S, Krailo M, Gao Y, Buxton A, Weigel BJ, et al. Phase II Trial of the Glycoprotein Non-metastatic B-Targeted Antibody-Drug Conjugate, Glembatumumab Vedotin, CDX-011), in Recurrent Osteosarcoma AOST1521: A Report from the Children’s Oncology Group. Eur J Cancer, 2019, 121:177–83., DOI 10.1016/j.ejca.2019.08.015 Hasanov M, Rioth MJ, Kendra K, Hernandez-Aya L, Joseph RW, Williamson S, et al. A Phase II Study of Glembatumumab Vedotin for Metastatic Uveal Melanoma. Cancers, Basel,, 2020, 12:2270., DOI 10.3390/cancers12082270 Vahdat LT, Schmid P, Forero-Torres A, Blackwell K, Telli ML, MeliskoM, et al. Glembatumumab Vedotin for Patients withMetastatic, gpNMB Overexpressing, Triple-Negative Breast Cancer, “METRIC”): a Randomized Multicenter Study. NPJ Breast Cancer, 2021, 7:57., DOI 10.1038/s41523-021-00244-6 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610450 EP 1610462 DI 10.3389/pore.2022.1610450 PG 13 ER PT J AU Xie, Y Wu, H Hu, W Zhang, H Li, A Zhang, Z Ren, Sh Zhang, X AF Xie, Yuning Wu, Hongjiao Hu, Wenqian Zhang, Hongmei Li, Ang Zhang, Zhi Ren, Shuhua Zhang, Xuemei TI Identification of Hub Genes of Lung Adenocarcinoma Based on Weighted Gene Co-Expression Network in Chinese Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE next-generation sequencing; survival analysis; lung adenocarcinoma; WGCNA; PPI network ID next-generation sequencing; survival analysis; lung adenocarcinoma; WGCNA; PPI network AB Purpose: Lung adenocarcinoma is one of the most common malignancies. Though some historic breakthroughs have been made in lung adenocarcinoma, its molecular mechanisms of development remain elusive. The aim of this study was to identify the potential genes associated with the lung adenocarcinoma progression and to provide new ideas for the prognosis evaluation of lung adenocarcinoma. Methods: The transcriptional profiles of ten pairs of snap-frozen tumor and adjacent normal lung tissues were obtained by performing RNA-seq. Weighted gene co-expression network analysis (WGCNA) was used to construct free-scale gene co-expression networks in order to explore the associations of gene sets with the clinical features and to investigate the functional enrichment analysis of co-expression genes. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, and Gene Set Enrichment Analysis (GSEA) analyses were performed using clusterProfiler. The protein-protein network (PPI) was established using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and hub genes were identified using Cytohubba in Cytoscape. Transcription factor enrichment analysis was performed by the RcisTarget program in R language. Results: Based on RNA-seq data, 1,545 differentially expressed genes (DEGs) were found. Eight co-expression modules were identified among these DEGs. The blue module exhibited a strong correlation with LUAD, in which ADCY4, RXFP1, AVPR2, CALCRL, ADRB1, RAMP3, RAMP2 and VIPR1 were hub genes. A low expression level of RXFP1, AVPR2, ADRB1 and VIPR1 was detrimental to the survival of LUAD patients. Genes in the blue module enriched in 86 Gene Ontology terms and five KEGG pathways. We also found that transcription factors EGR3 and EXOSC3 were related to the biological function of the blue module. Overall, this study brings a new perspective to the understanding of LUAD and provides possible molecular biomarkers for prognosis evaluation of LUAD. C1 [Xie, Yuning] North China University of Science and Technology, School of Public HealthTangshan, China. [Wu, Hongjiao] North China University of Science and Technology, School of Public HealthTangshan, China. [Hu, Wenqian] North China University of Science and Technology, School of Public HealthTangshan, China. [Zhang, Hongmei] North China University of Science and Technology, School of Public HealthTangshan, China. [Li, Ang] North China University of Science and Technology, School of Public HealthTangshan, China. [Zhang, Zhi] North China University of Science and Technology, Affiliated Tangshan Gongren HospitalTangshan, China. [Ren, Shuhua] North China University of Science and Technology, Affiliated Tangshan Gongren HospitalTangshan, China. [Zhang, Xuemei] North China University of Science and Technology, School of Public HealthTangshan, China. RP Zhang, X (reprint author), North China University of Science and Technology, School of Public Health, Tangshan, China. EM jyxuemei@gmail.com CR Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin, 2021, 71(1):7–33., DOI 10.3322/caac.21654 Malhotra J, Malvezzi M, Negri E, La Vecchia C, Boffetta P. Risk Factors for Lung Cancer Worldwide. Eur Respir J, 2016, 48(3):889–902., DOI 10.1183/ 13993003.00359-2016 Herbst RS, Morgensztern D, Boshoff C. The Biology and Management of Nonsmall Cell LungCancer. Nature, 2018, 553(7689):446–54., DOI 10.1038/nature25183 Thomas A, Liu SV, Subramaniam DS, Giaccone G. Refining the Treatment of NSCLC According to Histological and Molecular Subtypes. Nat Rev Clin Oncol, 2015, 12(9):511–26., DOI 10.1038/nrclinonc.2015.90 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: a Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci U S A, 2005, 102(43):15545–50., DOI 10.1073/pnas.0506580102 Langfelder P, Horvath S. WGCNA: an R Package for Weighted Correlation Network Analysis. BMC Bioinformatics, 2008, 9:559., DOI 10.1186/1471-2105- 9-559 Yin L, Cai Z, Zhu B, Xu C. Identification of Key Pathways and Genes in the Dynamic Progression of HCC Based on WGCNA. Genes, Basel,, 2018, 9(2): E92., DOI 10.3390/genes9020092 Liu Z, Li M, Hua Q, Li Y, Wang G. Identification of an Eight-lncRNA Prognostic Model for Breast Cancer Using WGCNA Network Analysis and a Cox-Proportional Hazards Model Based on L1-Penalized Estimation. Int J Mol Med, 2019, 44(4):1333–43., DOI 10.3892/ijmm.2019.4303 Tian Z, He W, Tang J, Liao X, Yang Q, Wu Y, et al. Identification of Important Modules and Biomarkers in Breast Cancer Based on WGCNA. Onco Targets Ther, 2020, 13:6805–17., DOI 10.2147/ott.S258439 Yin X, Wang P, Yang T, Li G, Teng X, Huang W, et al. Identification of Key Modules and Genes Associated with Breast Cancer Prognosis Using WGCNA and ceRNA Network Analysis. Aging, Albany NY,, 2020, 13(2):2519–38., DOI 10.18632/aging.202285 Tang J, Kong D, Cui Q,Wang K, Zhang D, Gong Y, et al. Prognostic Genes of Breast Cancer Identified by Gene Co-expression Network Analysis. Front Oncol, 2018, 8:374., DOI 10.3389/fonc.2018.00374 Yan G, An Y, Xu B, Wang N, Sun X, Sun M, et al. Potential Impact of ALKBH5 and YTHDF1 on Tumor Immunity in Colon Adenocarcinoma. Front Oncol, 2021, 11:670490., DOI 10.3389/fonc.2021.670490 Jiang C, Liu Y, Wen S, Xu C, Gu L. In Silico development and Clinical Validation of Novel 8 Gene Signature Based on Lipid Metabolism Related Genes in colon Adenocarcinoma. Pharmacol Res, 2021, 169:105644., DOI 10. 1016/j.phrs.2021.105644 Li W, Liu J, Zhao H. Identification of a Nomogram Based on Long Non-coding RNA to Improve Prognosis Prediction of Esophageal Squamous Cell Carcinoma. Aging, Albany NY,, 2020, 12(2):1512–26., DOI 10.18632/aging. 102697 Zhang H, Luo YB, Wu W, Zhang L, Wang Z, Dai Z, et al. The Molecular Feature of Macrophages in Tumor Immune Microenvironment of Glioma Patients. Comput Struct Biotechnol J, 2021, 19:4603–18., DOI 10.1016/j.csbj. 2021.08.019 Zhang J, Wu Y, Jin HY, Guo S, Dong Z, Zheng ZC, et al. Prognostic Value of Sorting Nexin 10 Weak Expression in Stomach Adenocarcinoma Revealed by Weighted Gene Co-expression Network Analysis. World J Gastroenterol, 2018, 24(43):4906–19., DOI 10.3748/wjg.v24.i43.4906 Zhu Y, Zhao Y, Cao Z, Chen Z, Pan W. Identification of Three Immune Subtypes Characterized by Distinct Tumor Immune Microenvironment and Therapeutic Response in Stomach Adenocarcinoma. Gene, 2022, 818:146177., DOI 10.1016/j.gene.2021.146177 Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based Genome Alignment and Genotyping with HISAT2 and HISAT-Genotype. Nat Biotechnol, 2019, 37(8):907–15., DOI 10.1038/s41587-019-0201-4 Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL, et al. StringTie Enables Improved Reconstruction of a Transcriptome from RNA-Seq Reads. Nat Biotechnol, 2015, 33(3):290–5., DOI 10.1038/nbt.3122 Love MI, Huber W, Anders S. Moderated Estimation of Fold Change and Dispersion for RNA-Seq Data with DESeq2. Genome Biol, 2014, 15(12):550., DOI 10.1186/s13059-014-0550-8 Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters. Omics, 2012, 16(5):284–7., DOI 10. 1089/omi.2011.0118 Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, et al. ClueGO: a Cytoscape Plug-In to Decipher Functionally Grouped Gene Ontology and Pathway Annotation Networks. Bioinformatics, 2009, 25(8): 1091–3., DOI 10.1093/bioinformatics/btp101 Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING V11: Protein-Protein Association Networks with Increased Coverage, Supporting Functional Discovery in Genome-wide Experimental Datasets. Nucleic Acids Res, 2019, 47(D1):D607-D613., DOI 10.1093/nar/gky1131 Chin CH, Chen SH, Wu HH, Ho CW, Ko MT, Lin CY, et al. CytoHubba: Identifying Hub Objects and Sub-Networks from Complex Interactome. BMC Syst Biol, 2014, 8(4):S11., DOI 10.1186/1752-0509-8-s4-s11 Aibar S, Gonzalez-Blas CB, Moerman T, Huynh-Thu VA, Imrichova H, Hulselmans G, et al. SCENIC: Single-Cell Regulatory Network Inference and Clustering. Nat Methods, 2017, 14(11):1083–6., DOI 10.1038/nmeth. 4463 Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, et al. TCGAbiolinks: an R/Bioconductor Package for Integrative Analysis of TCGA Data. Nucleic Acids Res, 2016, 44(8):e71., DOI 10.1093/nar/gkv1507 Berridge MJ, Lipp P, Bootman MD. The Versatility and Universality of Calcium Signalling. Nat Rev Mol Cel Biol, 2000, 1(1):11–21., DOI 10.1038/ 35036035 Monteith GR, Prevarskaya N, Roberts-Thomson SJ. The Calcium-Cancer Signalling Nexus. Nat Rev Cancer, 2017, 17(6):367–80., DOI 10.1038/nrc. 2017.18 Piazza GA, Ward A, Chen X, Maxuitenko Y, Coley A, Aboelella NS, et al. PDE5 and PDE10 Inhibition Activates cGMP/PKG Signaling to Block Wnt/β- Catenin Transcription, Cancer Cell Growth, and Tumor Immunity. Drug Discov Today, 2020, 25(8):1521–7., DOI 10.1016/j.drudis.2020.06.008 Kong X, Wang JS, Yang H. Upregulation of lncRNA DARS-AS1 Accelerates Tumor Malignancy in Cervical Cancer by Activating cGMP-PKG Pathway. J Biochem Mol Toxicol, 2021, 35(6):1–11., DOI 10.1002/jbt.22749 Wurth R, Bajetto A, Harrison JK, Barbieri F, Florio T. CXCL12 Modulation of CXCR4 and CXCR7 Activity in Human Glioblastoma Stem-like Cells and Regulation of the Tumor Microenvironment. Front Cel Neurosci, 2014, 8:144., DOI 10.3389/fncel.2014.00144 Nazari A, Khorramdelazad H, Hassanshahi G. Biological/pathological Functions of the CXCL12/CXCR4/CXCR7 Axes in the Pathogenesis of Bladder Cancer. Int J Clin Oncol, 2017, 22(6):991–1000., DOI 10.1007/ s10147-017-1187-x Khare T, Bissonnette M, Khare S. CXCL12-CXCR4/CXCR7 Axis in Colorectal Cancer: Therapeutic Target in Preclinical and Clinical Studies. Int J Mol Sci, 2021, 22(14):7371., DOI 10.3390/ijms22147371 Daniel SK, Seo YD, Pillarisetty VG. The CXCL12-Cxcr4/cxcr7 axis as a Mechanism of Immune Resistance in Gastrointestinal Malignancies. Semin Cancer Biol, 2020, 65:176–88., DOI 10.1016/j.semcancer.2019.12.007 Hussain M, Adah D, Tariq M, Lu Y, Zhang J, Liu J, et al. CXCL13/ CXCR5 Signaling axis in Cancer. Life Sci, 2019, 227:175–86., DOI 10.1016/j. lfs.2019.04.053 He H, Wu J, Zang M, Wang W, Chang X, Chen X, et al. CCR6(+, B Lymphocytes Responding to Tumor Cell-Derived CCL20 Support Hepatocellular Carcinoma Progression via Enhancing Angiogenesis. Am J Cancer Res, 2017, 7(5):1151–63. Tan H, Wang S, Zhao L. A Tumour-Promoting Role of Th9 Cells in Hepatocellular Carcinoma through CCL20 and STAT3 Pathways. Clin Exp Pharmacol Physiol, 2017, 44(2):213–21., DOI 10.1111/1440-1681.12689 Wang D, Yuan W, Wang Y, Wu Q, Yang L, Li F, et al. Serum CCL20 Combined with IL-17A as Early Diagnostic and Prognostic Biomarkers for Human Colorectal Cancer. J Transl Med, 2019, 17(1):253., DOI 10.1186/s12967-019- 2008-y Wang D, Yang L, Yu W, Wu Q, Lian J, Li F, et al. Colorectal Cancer Cell- Derived CCL20 Recruits Regulatory T Cells to Promote Chemoresistance via FOXO1/CEBPB/NF-κB Signaling. J Immunother Cancer, 2019, 7(1):215., DOI 10.1186/s40425-019-0701-2 Lee SK, Park KK, Kim HJ, Park J, Son SH, Kim KR, et al. Human Antigen R-Regulated CCL20 Contributes to Osteolytic Breast Cancer Bone Metastasis. Sci Rep, 2017, 7(1):9610., DOI 10.1038/s41598-017-09040-4 Chen W, Qin Y, Wang D, Zhou L, Liu Y, Chen S, et al. CCL20 Triggered by Chemotherapy Hinders the Therapeutic Efficacy of Breast Cancer. PLoS Biol, 2018, 16(7):e2005869., DOI 10.1371/journal.pbio.2005869 D’Amico L, Belisario D, Migliardi G, Grange C, Bussolati B, D’Amelio P, et al. C-Met Inhibition Blocks Bone Metastasis Development Induced by Renal Cancer Stem Cells. Oncotarget, 2016, 7(29):45525–37., DOI 10.18632/ oncotarget.9997 Denton CP, Wells AU, Coghlan JG. Major Lung Complications of Systemic Sclerosis. Nat Rev Rheumatol, 2018, 14(9):511–27., DOI 10.1038/s41584-018- 0062-0 Samuel CS, Zhao C, Bond CP, Hewitson TD, Amento EP, Summers RJ, et al. Relaxin-1-deficient Mice Develop an Age-Related Progression of Renal Fibrosis. Kidney Int, 2004, 65(6):2054–64., DOI 10.1111/j.1523-1755.2004. 00628.x Devarakonda T, Salloum FN. Heart Disease and Relaxin: New Actions for an Old Hormone. Trends Endocrinol Metab, 2018, 29(5):338–48., DOI 10.1016/j. tem.2018.02.008 Bigazzi M, Brandi ML, Bani G, Sacchi TB. Relaxin Influences the Growth of MCF-7 Breast Cancer Cells. Mitogenic and Antimitogenic Action Depends on Peptide Concentration. Cancer, 1992, 70(3):639–43., DOI 10.1002/1097- 0142(19920801)70:3<639::aid-cncr2820700316>3.0.co;2-v Binder C, Hagemann T, Husen B, Schulz M, Einspanier A. Relaxin Enhances In-Vitro Invasiveness of Breast Cancer Cell Lines by Up-Regulation of Matrix Metalloproteases. Mol Hum Reprod, 2002, 8(9):789–96., DOI 10.1093/molehr/8. 9.789 Feng S, Agoulnik IU, Bogatcheva NV, Kamat AA, Kwabi-Addo B, Li R, et al. Relaxin Promotes Prostate Cancer Progression. Clin Cancer Res, 2007, 13(6): 1695–702., DOI 10.1158/1078-0432.Ccr-06-2492 Hombach-Klonisch S, Bialek J, Trojanowicz B, Weber E, Holzhausen HJ, Silvertown JD, et al. Relaxin Enhances the Oncogenic Potential of Human Thyroid Carcinoma Cells. Am J Pathol, 2006, 169(2):617–32., DOI 10.2353/ ajpath.2006.050876 Vinall RL, Mahaffey CM, Davis RR, Luo Z, Gandour-Edwards R, Ghosh PM, et al. Dual Blockade of PKA and NF-Κb Inhibits H2 Relaxin-Mediated Castrate-Resistant Growth of Prostate Cancer Sublines and Induces Apoptosis. Horm Cancer, 2011, 2(4):224–38., DOI 10.1007/s12672-011-0076-4 Feng S, Agoulnik IU, Truong A, Li Z, Creighton CJ, Kaftanovskaya EM, et al. Suppression of Relaxin Receptor RXFP1 Decreases Prostate Cancer Growth and Metastasis. Endocr Relat Cancer, 2010, 17(4):1021–33., DOI 10.1677/erc- 10-0073 Radestock Y, Willing C, Kehlen A, Hoang-Vu C, Hombach-Klonisch S. Relaxin Enhances S100A4 and Promotes Growth of Human Thyroid Carcinoma Cell Xenografts. Mol Cancer Res, 2010, 8(4):494–506., DOI 10. 1158/1541-7786.Mcr-09-0307 Thanasupawat T, Glogowska A, Burg M, Krcek J, Beiko J, Pitz M, et al. C1q/ TNF-related Peptide 8, CTRP8, Promotes Temozolomide Resistance in Human Glioblastoma. Mol Oncol, 2018, 12(9):1464–79., DOI 10.1002/1878- 0261.12349 Lodhi RS, Nakabayashi K, Suzuki K, Yamada AY, Hazama R, Ebina Y, et al. Relaxin Has Anti-apoptotic Effects on Human Trophoblast-Derived HTR-8/ SV Neo Cells. Gynecol Endocrinol, 2013, 29(12):1051–4., DOI 10.3109/ 09513590.2013.829444 Silvertown JD, Summerlee AJ, Klonisch T. Relaxin-like Peptides in Cancer. Int J Cancer, 2003, 107(4):513–9., DOI 10.1002/ijc.11424 Jung KH, Choi IK, Lee HS, Yan HH, Son MK, Ahn HM, et al. Oncolytic Adenovirus Expressing Relaxin, YDC002, Enhances Therapeutic Efficacy of Gemcitabine against Pancreatic Cancer. Cancer Lett, 2017, 396:155–66., DOI 10.1016/j.canlet.2017.03.009 Garona J, Sobol NT, Pifano M, Segatori VI, Gomez DE, Ripoll GV, et al. Preclinical Efficacy of [V4 Q5 ]dDAVP, a Second Generation Vasopressin Analog, on Metastatic Spread and Tumor-Associated Angiogenesis in Colorectal Cancer. Cancer Res Treat, 2019, 51(2):438–50., DOI 10.4143/crt. 2018.040 Wang J, Zhang X, Li J, Ma X, Feng F, Liu L, et al. ADRB1 Was Identified as a Potential Biomarker for Breast Cancer by the Co-analysis of Tumor Mutational burden and Immune Infiltration. Aging, Albany NY,, 2020, 13(1):351–63., DOI 10.18632/aging.104204 Zhao L, Yu Z, Zhao B. Mechanism of VIPR1 Gene Regulating Human Lung Adenocarcinoma H1299 Cells. Med Oncol, 2019, 36(11):91., DOI 10.1007/ s12032-019-1312-y Shin SH, Kim I, Lee JE, Lee M, Park JW. Loss of EGR3 Is an Independent Risk Factor for Metastatic Progression in Prostate Cancer. Oncogene, 2020, 39(36): 5839–54., DOI 10.1038/s41388-020-01418-5 Li X, Yuan M, Song L, Wang Y. Silencing of microRNA-210 Inhibits the Progression of Liver Cancer and Hepatitis B Virus-Associated Liver Cancer via Targeting EGR3. BMC Med Genet, 2020, 21(1):48., DOI 10.1186/s12881-020- 0974-9 Chien MH, Lee WJ, Yang YC, Li YL, Chen BR, Cheng TY, et al. KSRP Suppresses Cell Invasion and Metastasis through miR-23a-Mediated EGR3 mRNA Degradation in Non-small Cell Lung Cancer. Biochim Biophys Acta Gene Regul Mech, 2017, 1860(10):1013–24., DOI 10.1016/j. bbagrm.2017.08.005 Ansari D, Andersson R, Bauden MP, Andersson B, Connolly JB, Welinder C, et al. Protein Deep Sequencing Applied to Biobank Samples from Patients with Pancreatic Cancer. J Cancer Res Clin Oncol, 2015, 141(2):369-80., DOI 10.1007/ s00432-014-1817-x NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610455 EP 1610465 DI 10.3389/pore.2022.1610455 PG 11 ER PT J AU Liu, J Liu, L Su, Y Wang, Y Zhu, Y Sun, X Guo, Y Shan, J AF Liu, Jia Liu, Lei Su, Yang Wang, Yi Zhu, Yuchun Sun, Xiaobin Guo, Yuanbiao Shan, Jing TI IL-33 Participates in the Development of Esophageal Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE EMT; esophageal adenocarcinoma; interleukin-33; ST2; EAC rat model ID EMT; esophageal adenocarcinoma; interleukin-33; ST2; EAC rat model AB Background: The progression from chronic gastroesophageal reflux disease (GERD) to Barrett esophagus (BE) and esophageal adenocarcinoma (EAC) is an inflammatory-driven neoplastic change. Interleukin-33 (IL-33) has identified as a crucial factor in several inflammatory disorders and malignancies. Methods: The high-density tissue microarray of the human EAC was analyzed with IL-33 immunohistochemistry staining (IHC). By anastomosing the jejunum with the esophagus, the rat model of EAC with mixed gastroduodenal reflux was established. The expression of IL-33 was determined using quantitative real-time polymerase chain reaction (RT-qPCR), western blot (WB), IHC and enzyme-linked immunosorbent assay (ELISA). Esophageal adenocarcinoma cells (OE19 and OE33) and human esophageal epithelial cells (HEECs) were used. Results: In the cytoplasm of human EAC tissue, IL-33 expression was substantially greater than in adjacent normal tissue. In rat model, the expression of IL-33 in the EAC group was considerably greater than in the control group, and this expression increased with the upgrade of pathological stage. In in vitro experiment, the mRNA and protein levels of IL-33 were considerably greater in OE19 and OE33 than in HEECs. The stimulation of IL- 33 enhanced the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of OE19 and OE33, but soluble ST2 (sST2) inhibited these effects. IL-33 stimulated the release of IL-6 by OE19 and OE33 cells. Conclusion: This study demonstrated the overexpression of IL-33 in the transition from GERD to EAC and that IL-33 promoted carcinogenesis in EAC cells through ST2. IL-33 might be a possible preventive target for EAC. C1 [Liu, Jia] Southwest Jiaotong University, School of MedicineChengdu, China. [Liu, Lei] The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Medical Research CenterChengdu, China. [Su, Yang] Southwest Jiaotong University, School of MedicineChengdu, China. [Wang, Yi] North Sichuan Medical CollegeNanchong, China. [Zhu, Yuchun] North Sichuan Medical CollegeNanchong, China. [Sun, Xiaobin] The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Department of GastroenterologyChengdu, China. [Guo, Yuanbiao] The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Medical Research CenterChengdu, China. [Shan, Jing] The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Department of GastroenterologyChengdu, China. RP Shan, J (reprint author), The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Department of Gastroenterology, Chengdu, China. EM tulip-sj@163.com CR Hur CMM, Kong CY, Dowling EC, Nattinger KJ, Dunn M, Feuer EJ, et al. Trends in Esophageal Adenocarcinoma Incidence and Mortality. cancer, 2013, 119(6):1149–58., DOI 10.1002/cncr.27834 Corley DA, Mehtani K, Quesenberry C, Zhao W, de Boer J, Weiss NS. Impact of Endoscopic Surveillance on Mortality from Barrett’s Esophagus-Associated Esophageal Adenocarcinomas. Gastroenterology, 2013, 145(2):312–9., DOI 10. 1053/j.gastro.2013.05.004 Kamboj AK, Katzka DA, Iyer PG. Endoscopic Screening for Barrett’s Esophagus and Esophageal Adenocarcinoma: Rationale, Candidates, and Challenges. Gastrointest Endosc Clin N Am, 2021, 31(1):27–41., DOI 10.1016/j. giec.2020.08.002 Schlottmann F, Molena D, Patti MG. Gastroesophageal Reflux and Barrett’s Esophagus: a Pathway to Esophageal Adenocarcinoma. Updates Surg, 2018, 70(3):339–42., DOI 10.1007/s13304-018-0564-y Quante M, Graham TA, Jansen M. Insights into the Pathophysiology of Esophageal Adenocarcinoma. Gastroenterology, 2018, 154(2):406–20., DOI 10.1053/j.gastro.2017.09.046 Schmitz J, Owyang A, Oldham E, Song Y, Murphy E, McClanahan TK, et al. IL-33, an Interleukin-1-like Cytokine that Signals via the IL-1 Receptor-Related Protein ST2 and Induces T Helper Type 2-associated Cytokines. Immunity, 2005, 23(5):479–90., DOI 10.1016/j.immuni.2005. 09.015 Martin NT, Martin MU. Interleukin 33 Is a Guardian of Barriers and a Local Alarmin. Nat Immunol, 2016, 17(2):122–31., DOI 10.1038/ni.3370 Shan J, Oshima T, Muto T, Yasuda K, Fukui H, Watari J, et al. Epithelialderived Nuclear IL-33 Aggravates Inflammation in the Pathogenesis of Reflux Esophagitis. J Gastroenterol, 2015, 50(4):414–23., DOI 10.1007/s00535-014- 88-1 9. Pastorelli L, Garg RR, Hoang SB, Spina L, Mattioli B, Scarpa M, et al. Epithelial-derived IL-33 and its Receptor ST2 Are Dysregulated in Ulcerative Colitis and in Experimental Th1/Th2 Driven Enteritis. Proc Natl Acad Sci U S A, 2010, 107(17):8017–22., DOI 10.1073/pnas. 0912678107 Buzzelli JN, Chalinor HV, Pavlic DI, Sutton P, Menheniott TR, Giraud AS, et al. IL33 Is a Stomach Alarmin that Initiates a Skewed Th2 Response to Injury and Infection. Cell Mol Gastroenterol Hepatol, 2015, 1(2):203–21., DOI 10.1016/ j.jcmgh.2014.12.003 Yu XX, Hu Z, Shen X, Dong LY, Zhou WZ, Hu WH. IL-33 Promotes Gastric Cancer Cell Invasion and Migration via ST2-Erk1/2 Pathway. Dig Dis Sci, 2015, 60(5):1265–72., DOI 10.1007/s10620-014-3463-1 Mertz KD, Mager LF, Wasmer MH, Thiesler T, Koelzer VH, Ruzzante G, et al. The IL-33/ST2 Pathway Contributes to Intestinal Tumorigenesis in Humans and Mice. Oncoimmunology, 2016, 5(1):e1062966., DOI 10.1080/2162402X. 2015.1062966 Chen SF, Nieh S, Jao SW, Wu MZ, Liu CL, Chang YC, et al. The Paracrine Effect of Cancer-Associated Fibroblast-Induced Interleukin-33 Regulates the Invasiveness of Head and Neck Squamous Cell Carcinoma. J Pathol, 2013, 231(2):180–9., DOI 10.1002/path.4226 Zhang Y, Davis C, Shah S, Hughes D, Ryan JC, Altomare D, et al. IL-33 Promotes Growth and Liver Metastasis of Colorectal Cancer in Mice by Remodeling the Tumor Microenvironment and Inducing Angiogenesis. Mol Carcinog, 2017, 56(1):272–87., DOI 10.1002/mc.22491 Akimoto M, Hayashi JI, Nakae S, Saito H, Takenaga K. Interleukin-33 Enhances Programmed Oncosis of ST2L-Positive Low-Metastatic Cells in the Tumour Microenvironment of Lung Cancer. Cell Death Dis, 2016, 7: e2057., DOI 10.1038/cddis.2015.418 Fang D, Kuzel TM, Zhang B, Wang C, Chen Z, Bu X, et al. IL-33 Signaling Fuels Outgrowth and Metastasis of Human Lung Cancer. Biochem Biophys Res Commun, 2016, 479(3):461–8., DOI 10.1016/j.bbrc.2016.09.081 Shan J, Oshima T, Wu L, Fukui H, Watari J, Miwa H. Interferon Gamma- Induced Nuclear Interleukin-33 Potentiates the Release of Esophageal Epithelial Derived Cytokines. PLoS One, 2016, 11(3):e0151701., DOI 10. 1371/journal.pone.0151701 Mukaisho KI, Kanai S, Kushima R, Nakayama T, Hattori T, Sugihara H. Barretts’s Carcinogenesis. Pathol Int, 2019, 69(6):319–30., DOI 10.1111/pin. 12804 Greten FR, Grivennikov SI. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity, 2019, 51(1):27–41., DOI 10.1016/j.immuni.2019. 06.025 Wang F, Meng W, Wang B, Qiao L. Helicobacter Pylori-Induced Gastric Inflammation and Gastric Cancer. Cancer Lett, 2014, 345(2):196–202., DOI 10. 1016/j.canlet.2013.08.016 Terzic J, Grivennikov S, Karin E, Karin M. Inflammation and colon Cancer. Gastroenterology, 2010, 138(6):2101–14., DOI 10.1053/j.gastro. 2010.01.058 Kunze B, Wein F, Fang HY, Anand A, Baumeister T, Strangmann J, et al. Notch SignalingMediates Differentiation in Barrett’s Esophagus and Promotes Progression to Adenocarcinoma. Gastroenterology, 2020, 159(2):575–90., DOI 10.1053/j.gastro.2020.04.033 Liew FY, Girard J-P, Turnquist HR. Interleukin-33 in Health and Disease. Nat Rev Immunol, 2016, 16(11):676–89., DOI 10.1038/nri.2016.95 Cayrol C, Girard JP. Interleukin-33, IL-33): A Nuclear Cytokine from the IL-1 Family. Immunol Rev, 2018, 281(1):154–68., DOI 10.1111/imr.12619 Cayrol C, Girard JP. IL-33: an Alarmin Cytokine with Crucial Roles in Innate Immunity, Inflammation and Allergy. Curr Opin Immunol, 2014, 31:31–7., DOI 10.1016/j.coi.2014.09.004 Tu L, Yang L. IL-33 at the Crossroads of Metabolic Disorders and Immunity. Front Endocrinol, Lausanne,, 2019, 10:26., DOI 10.3389/fendo.2019.00026 Afferni C, Buccione C, Andreone S, GaldieroMR, Varricchi G, Marone G, et al. The Pleiotropic Immunomodulatory Functions of IL-33 and its Implications in Tumor Immunity. Front Immunol, 2018, 9:2601., DOI 10.3389/fimmu.2018.02601 Yang ZP, Ling DY, Xie YH, Wu WX, Li JR, Jiang J, et al. The Association of Serum IL-33 and sST2 with Breast Cancer. Dis Markers, 2015, 2015:516895., DOI 10.1155/2015/516895 Huang N, Cui X, LiW, Zhang C, Liu L, Li J. IL33/ST2 Promotes theMalignant Progression of Gastric Cancer via the MAPK Pathway. Mol Med Rep, 2021, 23(5):361., DOI 10.3892/mmr.2021.12000 Shani O, Vorobyov T, Monteran L, Lavie D, Cohen N, Raz Y, et al. Fibroblast- Derived IL33 Facilitates Breast Cancer Metastasis by Modifying the Immune Microenvironment and Driving Type 2 Immunity. Cancer Res, 2020, 80(23): 5317–29., DOI 10.1158/0008-5472.CAN-20-2116 Jovanovic IP, Pejnovic NN, Radosavljevic GD, Pantic JM, Milovanovic MZ, Arsenijevic NN, et al. Interleukin-33/ST2 axis Promotes Breast Cancer Growth and Metastases by Facilitating Intratumoral Accumulation of Immunosuppressive and Innate Lymphoid Cells. Int J Cancer, 2014, 134(7):1669–82., DOI 10.1002/ijc.28481 Zhou Q, Wu X, Wang X, Yu Z, Pan T, Li Z, et al. The Reciprocal Interaction between Tumor Cells and Activated Fibroblasts Mediated by TNF-α/IL-33/ ST2L Signaling Promotes Gastric Cancer Metastasis. Oncogene, 2020, 39(7): 1414–28., DOI 10.1038/s41388-019-1078-x Pastille E, Wasmer MH, Adamczyk A, Vu VP, Mager LF, Phuong NNT, et al. The IL-33/ST2 Pathway Shapes the Regulatory T Cell Phenotype to Promote Intestinal Cancer. Mucosal Immunol, 2019, 12(4):990–1003., DOI 10.1038/ s41385-019-0176-y Yang Z, Gao X, Wang J, Xu L, Zheng Y, Xu Y. Interleukin-33 Enhanced the Migration and Invasiveness of Human Lung Cancer Cells. Onco Targets Ther, 2018, 11:843–9., DOI 10.2147/OTT.S155905 Zhang X, Hu F, Li G, Li G, Yang X, Liu L, et al. Human Colorectal Cancer- Derived Mesenchymal Stem Cells Promote Colorectal Cancer Progression through IL-6/JAK2/STAT3 Signaling. Cell Death Dis, 2018, 9(2):25., DOI 10. 1038/s41419-017-0176-3 Weng YS, Tseng HY, Chen YA, Shen PC, Al Haq AT, Chen LM, et al. MCT-1/ miR-34a/IL-6/IL-6R Signaling axis Promotes EMT Progression, Cancer Stemness and M2 Macrophage Polarization in Triple-Negative Breast Cancer. Mol Cancer, 2019, 18(1):42., DOI 10.1186/s12943-019-0988-0 Razidlo GL, Burton KM, McNiven MA. Interleukin-6 Promotes Pancreatic Cancer Cell Migration by Rapidly Activating the Small GTPase CDC42. J Biol Chem, 2018, 293(28):11143–53., DOI 10.1074/jbc.RA118.003276 Ebbing EA, van der Zalm AP, Steins A, Creemers A, Hermsen S, Rentenaar R, et al. Stromal-derived Interleukin 6 Drives Epithelial-To-Mesenchymal Transition and Therapy Resistance in Esophageal Adenocarcinoma. Proc Natl Acad Sci U S A, 2019, 116(6):2237–42., DOI 10.1073/pnas.1820459116 Karakasheva TA, Lin EW, Tang Q, Qiao E, Waldron TJ, Soni M, et al. IL-6 Mediates Cross-Talk between Tumor Cells and Activated Fibroblasts in the Tumor Microenvironment. Cancer Res, 2018, 78(17):4957–70., DOI 10.1158/ 0008-5472.CAN-17-2268 Braun H, Afonina IS, Mueller C, Beyaert R. Dichotomous Function of IL-33 in Health and Disease: From Biology to Clinical Implications. Biochem Pharmacol, 2018, 148:238–52., DOI 10.1016/j.bcp.2018.01.010 Yang Y, Andersson P, Hosaka K, Zhang Y, Cao R, Iwamoto H, et al. The PDGF-BB-SOX7 axis-modulated IL-33 in Pericytes and Stromal Cells Promotes Metastasis through Tumour-Associated Macrophages. Nat Commun, 2016, 7(1):11385., DOI 10.1038/ncomms11385 Kurowska-Stolarska M, Stolarski B, Kewin P, Murphy G, Corrigan CJ, Ying S, et al. IL-33 Amplifies the Polarization of Alternatively Activated Macrophages that Contribute to Airway Inflammation. J Immunol, 2009, 183(10):6469–77., DOI 10.4049/jimmunol.0901575 Wang C, Dong C, Xiong S. IL-33 Enhances Macrophage M2 Polarization and Protects Mice from CVB3-Induced Viral Myocarditis. J Mol Cell Cardiol, 2017, 103:22–30., DOI 10.1016/j.yjmcc.2016.12.010 EissmannMF, Dijkstra C, Jarnicki A, Phesse T, Brunnberg J, Poh AR, et al. IL-33- mediated Mast Cell Activation Promotes Gastric Cancer through Macrophage Mobilization. Nat Commun, 2019, 10(1):2735., DOI 10.1038/s41467-019-10676-1 Wei C, Yang C, Wang S, Shi D, Zhang C, Lin X, et al. Crosstalk between Cancer Cells and Tumor Associated Macrophages Is Required for Mesenchymal Circulating Tumor Cell-Mediated Colorectal Cancer Metastasis. Mol Cancer, 2019, 18(1):64., DOI 10.1186/s12943-019-0976-4 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610474 EP 1610485 DI 10.3389/pore.2022.1610474 PG 12 ER PT J AU Zhou, Z Huang, D Yang, Sh Liang, J Wang, X Rao, Q AF Zhou, Zhiyi Huang, Dandan Yang, Shudong Liang, Jiabei Wang, Xuan Rao, Qiu TI Clinicopathological Significance, Related Molecular Changes and Tumor Immune Response Analysis of the Abnormal SWI/SNF Complex Subunit PBRM1 in Gastric Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; PD-L1; gastric adenocarcinoma (GAC); PBRM1; microsatellite stability; tumor-infiltrating lymphocytes (TIL) ID prognosis; PD-L1; gastric adenocarcinoma (GAC); PBRM1; microsatellite stability; tumor-infiltrating lymphocytes (TIL) AB Background: PBRM1 gene abnormalities were recently found to play a role in tumor development and tumor immune activity. This article will explore the clinicopathological and molecular changes and tumor immune activity of the abnormal SWI/SNF complex subunit PBRM1 in gastric adenocarcinoma (GAC) and its significance. Methods: The cBioPortal, LinkedOmics and TISIDB datasets were used to analyze the abnormality of the PBRM1 gene in GAC and its relationship with prognosis, related molecular changes and tumor-infiltrating lymphocytes (TILs). In addition, 198 GAC cases were collected to further study the relationship between the loss/attenuation of PBRM1 expression and clinicopathology, prognosis, microsatellite stability, PD-L1 expression and TIL in GAC. DNA whole-exome sequencing was performed on 7 cases of gastric cancer with loss of PBRM1 expression. Results: The cBioPortal data showed that PBRM1 deletion/mutation accounted for 7.32% of GAC and was significantly associated with several molecular changes, such as molecular subtypes of GAC. The LinkedOmics dataset showed that PBRM1 mutation and its promoter DNA methylation showed lower PBRM1 mRNA expression, and PBRM1 mutation cases showed significantly higher mRNA expression of PD-L1 (CD274). TISIDB data showed that PBRM1 abnormalities were significantly positively associated with multiple TILs. In our group of 198 cases, the loss/attenuation of PBRM1 expression was significantly positively correlated with intra-tumoral tumor infiltrating lymphocytes (iTILs) and deficient MMR and PD-L1 expression. Kaplan–Meier survival analysis showed that the overall survival of GAC patients with loss/attenuation of PBRM1 expression was significantly better (p = 0.023). iTIL was an independent prognostic factor of GAC. Loss of PBRM1 expression often co-occurs with mutations in other SWI/SNF complex subunit genes, and there are some repetitive KEGG signaling changes. Conclusion: Abnormality of the PBRM1 gene may be related to the occurrence of some GACs and can affect tumor immune activity, thereby affecting clinicopathology and prognosis. It may be a potentially effective predictive marker for immunotherapy and a novel therapeutic approach associated with synthetic lethality. C1 [Zhou, Zhiyi] The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Department of PathologyWuxi, China. [Huang, Dandan] The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Digestive Endoscopic CenterWuxi, China. [Yang, Shudong] The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Department of PathologyWuxi, China. [Liang, Jiabei] The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Department of PathologyWuxi, China. [Wang, Xuan] Nanjing University School of Medicine, Jinling Hospital, Department of PathologyNanjing, China. [Rao, Qiu] Nanjing University School of Medicine, Jinling Hospital, Department of PathologyNanjing, China. RP Rao, Q (reprint author), Nanjing University School of Medicine, Jinling Hospital, Department of Pathology, Nanjing, China. EM nanjing_raoq@163.com CR Joshi SS, Badgwell BD. Current Treatment and Recent Progress in Gastric Cancer. CA Cancer J Clin, 2021, 71(3):264–79., DOI 10.3322/caac.21657 Sasaki M, Ogiwara H. Synthetic Lethal Therapy Based on Targeting the Vulnerability of SWI/SNF Chromatin Remodeling Complex-deficient Cancers. Cancer Sci, 2020, 111(3):774–82., DOI 10.1111/cas.14311 Liu X-D, Kong W, Peterson CB, McGrail DJ, Hoang A, Zhang X, et al. PBRM1 Loss Defines a Nonimmunogenic Tumor Phenotype Associated with Checkpoint Inhibitor Resistance in Renal Carcinoma. Nat Commun, 2020, 11(1):2135., DOI 10.1038/s41467-020-15959-6 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Sci Signal, 2013, 6(269):pl1., DOI 10.1126/scisignal. 2004088 Vasaikar SV, Straub P, Wang J, Zhang B. LinkedOmics: Analyzing Multi- Omics Data within and across 32 Cancer Types. Nucleic Acids Res, 2018, 46(D1):D956–D963., DOI 10.1093/nar/gkx1090 Ru B, Wong CN, Tong Y, Zhong JY, Zhong SSW, Wu WC, et al. TISIDB: An Integrated Repository portal for Tumor-Immune System Interactions. Bioinformatics, 2019, 35(20):4200–2., DOI 10.1093/bioinformatics/btz210 Huang S-C, Ng K-F, Chang IY-F, Chang C-J, Chao Y-C, Chang S-C, et al. The Clinicopathological Significance of SWI/SNF Alterations in Gastric Cancer Is Associated with the Molecular Subtypes. PLoS One, 2021, 16(1):e0245356., DOI 10.1371/journal.pone.0245356 Hendry S, Salgado R, Gevaert T, Russell PA, John T, Thapa B, et al. Assessing Tumor-Infiltrating Lymphocytes in Solid Tumors: A Practical Review for Pathologists and Proposal for a Standardized Method from the International Immuno-Oncology Biomarkers Working Group: Part 2: TILs in Melanoma, Gastrointestinal Tract Carcinomas, Non-small Cell Lung Carcinoma and Mesothelioma, Endometrial and Ovarian Carcinomas, Squamous Cell Carcinoma of the Head and Neck, Genitourinary Carcinomas, and Primary Brain Tumors. Adv Anat Pathol, 2017, 24(6):311–35., DOI 10.1097/PAP. 0000000000000161 Gonzalez IA, Bauer PS, Liu J, Chatterjee D. Intraepithelial Tumour Infiltrating Lymphocytes Are Associated with Absence of Tumour Budding and Immature/myxoid Desmoplastic Reaction, and with Better Recurrence-free Survival in Stages I-III Colorectal Cancer. Histopathology, 2021, 78(2):252–64., DOI 10.1111/his.14211 Jun S-Y, Park ES, Lee JJ, Chang H-K, Jung ES, Oh Y-H, et al. Prognostic Significance of Stromal and Intraepithelial Tumor-Infiltrating Lymphocytes in Small Intestinal Adenocarcinoma. Am J Clin Pathol, 2020, 153(1):105–18., DOI 10.1093/ajcp/aqz136 Xu H, Zhu X, Bao H,Wh Shek T, Huang Z, Wang Y, et al. Genetic and Clonal Dissection of Osteosarcoma Progression and Lung Metastasis. Int J Cancer, 2018, 143(5):1134–42., DOI 10.1002/ijc.31389 Savas S, Skardasi G. The SWI/SNF Complex Subunit Genes: Their Functions, Variations, and Links to Risk and Survival Outcomes in Human Cancers. Crit Rev Oncol Hematol, 2018, 123:114–31., DOI 10. 1016/j.critrevonc.2018.01.009 Pierre RS, Kadoch C. Mammalian SWI/SNF Complexes in Cancer: Emerging Therapeutic Opportunities. Curr Opin Genet Dev, 2017, 42:56–67., DOI 10. 1016/j.gde.2017.02.004 Hu B, Lin JZ, Yang XB, Sang XT. The Roles of Mutated SWI/SNF Complexes in the Initiation and Development of Hepatocellular Carcinoma and its Regulatory Effect on the Immune System: A Review. Cell Prolif, 2020, 53(4):e12791., DOI 10.1111/cpr.12791 Pan D, Kobayashi A, Jiang P, Ferrari de Andrade L, Tay RE, Luoma AM, et al. A Major Chromatin Regulator Determines Resistance of Tumor Cells to T Cell-Mediated Killing. Science, 2018, 359(6377):770–5., DOI 10.1126/science. aao1710 Panwalkar P, Pratt D, Chung C, Dang D, Le P, Martinez D, et al. SWI/SNF Complex Heterogeneity Is Related to Polyphenotypic Differentiation, Prognosis, and Immune Response in Rhabdoid Tumors. Neuro Oncol, 2020, 22(6):785–96., DOI 10.1093/neuonc/noaa004 Xing X, Guo J, Ding G, Li B, Dong B, Feng Q, et al. Analysis of PD1, PDL1, PDL2 Expression and T Cells Infiltration in 1014 Gastric Cancer Patients. Oncoimmunology, 2017, 7(3):e1356144., DOI 10.1080/2162402X.2017.1356144 Kim H, Heo YJ, Cho YA, Kang SY, Ahn S, Kim K-M. Tumor Immune Microenvironment Is Influenced by Frameshift Mutations and Tumor Mutational burden in Gastric Cancer. Clin Transl Oncol, 2022, 24(3): 556–67., DOI 10.1007/s12094-021-02714-6 Morikawa R, Nemoto Y, Yonemoto Y, Tanaka S, Takei Y, Oshima S, et al. Intraepithelial Lymphocytes Suppress Intestinal Tumor Growth by Cell-To- Cell Contact via CD103/E-Cadherin Signal. Cell Mol Gastroenterol Hepatol, 2021, 11(5):1483–503., DOI 10.1016/j.jcmgh.2021.01.014 Schallenberg S, Bork J, Essakly A, Alakus H, Buettner R, Hillmer AM, et al. Loss of the SWI/SNF-ATPase Subunit Members SMARCF1, ARID1A), SMARCA2, BRM), SMARCA4, BRG1, and SMARCB1, INI1, in Oesophageal Adenocarcinoma. BMC Cancer, 2020, 20(1):12., DOI 10.1186/ s12885-019-6425-3 Peng L, Li J, Wu J, Xu B, Wang Z, Giamas G, et al. A Pan-Cancer Analysis of SMARCA4 Alterations in Human Cancers. Front Immunol, 2021, 12:762598., DOI 10.3389/fimmu.2021.762598 Wang L, Yang L, Wang C, Zhao W, Ju Z, Zhang W, et al. Inhibition of the ATM/Chk2 axis Promotes cGAS/STING Signaling in ARID1A-Deficient Tumors. J Clin Invest, 2020, 130(11):5951–66., DOI 10.1172/JCI130445 Shen J, Ju Z, Zhao W, Wang L, Peng Y, Ge Z, et al. ARID1A Deficiency Promotes Mutability and Potentiates Therapeutic Antitumor Immunity Unleashed by Immune Checkpoint Blockade. Nat Med, 2018, 24(5): 556–62., DOI 10.1038/s41591-018-0012-z Tokunaga R, Xiu J, Goldberg RM, Philip PA, Seeber A, Battaglin F, et al. The Impact of ARID1A Mutation on Molecular Characteristics in Colorectal Cancer. Eur J Cancer, 2020, 140:119–29., DOI 10.1016/j.ejca. 2020.09.006 Li L, Li M, Jiang Z, Wang X. ARID1A Mutations are Associated with Increased Immune Activity in Gastrointestinal Cancer. Cells, 2019, 8(7):678., DOI 10. 3390/cells8070678 Botta GP, Kato S, Patel H, Fanta P, Lee S, Okamura R, et al. SWI/SNF Complex Alterations as a Biomarker of Immunotherapy Efficacy in Pancreatic Cancer. JCI Insight, 2021, 6(18):e150453., DOI 10.1172/jci.insight.150453 Xu S, Tang C. The Role of ARID1A in Tumors: Tumor Initiation or Tumor Suppression? Front Oncol, 2021, 11:745187., DOI 10.3389/fonc.2021.745187 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610479 EP 1610490 DI 10.3389/pore.2022.1610479 PG 12 ER PT J AU Fang, Z Wang, F Zhang, M Huang, H Lin, Z AF Fang, Zhijie Wang, Feifei Zhang, Mengya Huang, Hua Lin, Zhiqiang TI Identification of Co-Expression Modules and Genes Associated With Tumor Progression in Oral Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tumor progression; WGCNA; prognostic markers; OSCC; hub-gene ID tumor progression; WGCNA; prognostic markers; OSCC; hub-gene AB Oral squamous cell carcinoma (OSCC) is a common head-and-neck cancer with a deficiency of early diagnosis and poor prognosis. To identify potential diagnostic and prognostic markers of OSCC, we firstly used weighted gene co-expression network analysis (WGCNA) to build a co-expression module from GSE42743. Next, we performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on specified units from selected modules utilizing Database for Annotation, Visualization, and Integrated Discovery (DAVID). Additionally, we identified and validate hub genes of these specified modules from multiple datasets like GEPIA and TCGA. In total 16 co-expression modules were built by 17,238 genes of 74 tumor samples utilizing WGCNA. Through pathway and functional enrichment analysis, the turquoise module was most firmly relevant to the cell cycle, oocyte meiosis, and p53 signaling pathway. Hub genes VRK1, NUP37, HMMR, SPC25, and RUVBL1 were identified to be related to oral cancer at both molecular level and clinical levels. The expressions of these genes differed in tumor tissues and normal tissues. Meanwhile, patients with high hub gene expression had a poor prognosis clinically. To conclude, five hub genes were identified to be relevant to oral cancer from the molecular level and the clinical level. Therefore, the detection of these genes was of great significance. They can be regarded as diagnostic and prognostic biomarkers for oral cancer. Also, they could shed light on the improvement of patients’ overall survival and prognosis, which needs further analysis in the future. C1 [Fang, Zhijie] Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Department of OtolaryngologySuzhou, China. [Wang, Feifei] The Affiliated BenQ Hospital of Nanjing Medical University, Suzhou BenQ Medical Center, Department of NursingSuzhou, China. [Zhang, Mengya] Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Department of OtolaryngologySuzhou, China. [Huang, Hua] Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Department of OtolaryngologySuzhou, China. [Lin, Zhiqiang] Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Department of OtolaryngologySuzhou, China. RP Lin, Z (reprint author), Gusu School, Nanjing Medical University, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Department of Otolaryngology, Suzhou, China. EM 865205697@qq.com CR Thomson PJ. Perspectives on Oral Squamous Cell Carcinoma Prevention- Proliferation, Position, Progression and Prediction. J Oral Pathol Med, 2018, 47(9):803–7., DOI 10.1111/jop.12733 Gandini S, Botteri E, Iodice S, Boniol M, Lowenfels AB, Maisonneuve P, et al. Tobacco Smoking and Cancer: a Meta-Analysis. Int J Cancer, 2008, 122(1): 155–64., DOI 10.1002/ijc.23033 Turati F, Garavello W, Tramacere I, PeluCChi C, Galeone C, Bagnardi V, et al. A Meta-Analysis of Alcohol Drinking and Oral and Pharyngeal Cancers: Results from Subgroup Analyses. Alcohol Alcohol, 2013, 48(1):107–18., DOI 10. 1093/alcalc/ags100 Guha N, Warnakulasuriya S, Vlaanderen J, Straif K. Betel Quid Chewing and the Risk of Oral and Oropharyngeal Cancers: a Meta-Analysis with Implications for Cancer Control. Int J Cancer, 2014, 135(6):1433–43., DOI 10.1002/ijc.28643 Mirghani H, Amen F, Moreau F, Lacau St Guily J. Do high-risk Human Papillomaviruses Cause Oral Cavity Squamous Cell Carcinoma? Oral Oncol, 2015, 51(3):229–36., DOI 10.1016/j.oraloncology.2014.11.011 Chi AC, Day TA, Neville BW. Oral Cavity and Oropharyngeal Squamous Cell Carcinoma-Aan Update. CA Cancer J Clin, 2015, 65(5):401–21., DOI 10.3322/ caac.21293 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Siegel RL,Miller KD, Jemal A. Cancer Statistics, 2019. CA Cancer J Clin, 2019, 69(1):7–34., DOI 10.3322/caac.21551 Kowalski LP, Carvalho AL. Natural History of Untreated Head and Neck Cancer. Eur J Cancer, 2000, 36(8):1032–7., DOI 10.1016/s0959-8049(00, 00054-x Hashim D, GEndEn E, Posner M, Hashibe M, Boffetta P. Head and Neck Cancer Prevention: from Primary Prevention to Impact of Clinicians on Reducing burden. Ann Oncol, 2019, 30(5):744–56., DOI 10.1093/annonc/ mdz084 Langfelder P, Horvath S. WGCNA: an R Package for Weighted Correlation Network Analysis. BMC Bioinformatics, 2008, 9:559., DOI 10.1186/1471-2105-9-559 Lohavanichbutr P, Mendez E, Holsinger FC, Rue TC, Zhang Y, Houck J, et al. A 13-gene Signature Prognostic of HPV-Negative OSCC: Discovery and External Validation. Clin Cancer Res, 2013, 19(5):1197–203., DOI 10.1158/ 1078-0432.CCR-12-2647 Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a Web Server for Cancer and normal Gene Expression Profiling and Interactive Analyses. Nucleic Acids Res, 2017, 45(W1):W98–W102., DOI 10.1093/nar/gkx247 Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al. PGC-Lalpha-Responsive Genes Involved in Oxidative Phosphorylation Are Coordinately Downregulated in Human Diabetes. Nat Genet, 2003, 34(3): 267–73., DOI 10.1038/ng1180 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: a Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci U S A, 2005, 102(43):15545–50., DOI 10.1073/pnas.0506580102 Hanzelmann S, Castelo R, Guinney J. GSVA: Gene Set Variation Analysis for Microarray and RNA-Seq Data. BMC Bioinformatics, 2013, 14:7., DOI 10.1186/ 1471-2105-14-7 Klutstein M, Nejman D, Greenfield R, Cedar H. DNA Methylation in Cancer and Aging. Cancer Res, 2016, 76(12):3446–50., DOI 10.1158/0008-5472.CAN- 15-3278 Fu C, Jiang A. Dendritic Cells and CD8 T Cell Immunity in Tumor Microenvironment. Front Immunol, 2018, 9:3059., DOI 10.3389/fimmu.2018. 03059 Valbuena A, Sanz-Garcia M, Lopez-Sanchez I, Vega FM, Lazo PA. Roles of VRK1 as a New Player in the Control of Biological Processes Required for Cell Division. Cell Signal, 2011, 23(8):1267–72., DOI 10.1016/j.cellsig.2011. 04.002 Valbuena A, Lopez-Sanchez I, Lazo PA. Human VRK1 Is an Early Response Gene and its Loss Causes a Block in Cell Cycle Progression. Plos One, 2008, 3(2):e1642., DOI 10.1371/journal.pone.0001642 Santos CR, Rodriguez-Pinilla M, Vega FM, Rodriguez-Peralto JL, Blanco S, Sevilla A, et al. VRK1 Signaling Pathway in the Context of the Proliferation Phenotype in Head and Neck Squamous Cell Carcinoma. Mol Cancer Res, 2006, 4(3):177–85., DOI 10.1158/1541-7786.MCR-05- 0212 Valbuena A, Suarez-Gauthier A, Lopez-Rios F, Lopez-Encuentra A, Blanco S, Fernandez PL, et al. Alteration of the VRK1-P53 Autoregulatory Loop in Human Lung Carcinomas. Lung Cancer, 2007, 58(3):303–9., DOI 10.1016/j. lungcan.2007.06.023 Lee N, Kwon JH, Kim YB, Kim SH, Park SJ, Xu W, et al. Vaccinia-related Kinase 1 Promotes Hepatocellular Carcinoma by Controlling the Levels of Cell Cycle Regulators Associated with G1/S Transition. Oncotarget, 2015, 6(30): 30130–48., DOI 10.18632/oncotarget.4967 Finetti P, Cervera N, Charafe-Jauffret E, Chabannon C, Charpin C, Chaffanet M, et al. Sixteen-kinase Gene Expression Identifies Luminal Breast Cancers with Poor Prognosis. Cancer Res, 2008, 68(3):767–76., DOI 10.1158/0008-5472. CAN-07-5516 Zhang Q, Zhang J, Jin H, Sheng S.Whole Transcriptome Sequencing Identifies Tumor-specific Mutations in Human Oral Squamous Cell Carcinoma. BMC Med Genomics, 2013, 6:28., DOI 10.1186/1755-8794-6-28 Luo X, Liu Y, Feng W, Lei L, Du Y,Wu J, et al. NUP37, a Positive Regulator of YAP/TEAD Signaling, Promotes the Progression of Hepatocellular Carcinoma. Oncotarget, 2017, 8(58):98004–13., DOI 10.18632/oncotarget.20336 Huang L, Wang T, Wang F, Hu X, Zhan G, Jin X, et al. NUP37 Silencing Induces Inhibition of Cell Proliferation, G1 Phase Cell Cycle Arrest and Apoptosis in Non-small Cell Lung Cancer Cells. Pathol Res Pract, 2020, 216(3):152836., DOI 10.1016/j.prp.2020.152836 He Z, Mei L, Connell M, Maxwell CA. Hyaluronan MediatedMotility Receptor, HMMR, Encodes an Evolutionarily Conserved Homeostasis, Mitosis, and Meiosis Regulator rather Than a Hyaluronan Receptor. Cells, 2020, 9(4):E819., DOI 10.3390/cells9040819 Mantripragada KK, Spurlock G, Kluwe L, Chuzhanova N, Ferner RE, Frayling IM, et al. High-resolution DNA Copy Number Profiling of Malignant Peripheral Nerve Sheath Tumors Using Targeted Microarray-Based Comparative Genomic Hybridization. Clin Cancer Res, 2008, 14(4): 1015–24., DOI 10.1158/1078-0432.CCR-07-1305 Li H, Guo L, Li JW, Liu N, Qi R, Liu J. Expression of Hyaluronan Receptors CD44 and RHAMM in Stomach Cancers: Relevance with Tumor Progression. Int J Oncol, 2000, 17(5):927–32., DOI 10.3892/ijo.17.5.927 Assmann V, Gillett CE, Poulsom R, Ryder K, Hart IR, Hanby AM. The Pattern of Expression of the Microtubule-Binding Protein RHAMM/IHABP in Mammary Carcinoma Suggests a Role in the Invasive Behaviour of Tumour Cells. J Pathol, 2001, 195(2):191–6., DOI 10.1002/path.941 Chen J, Chen H, Yang H, Dai H. SPC25 Upregulation Increases Cancer Stem Cell Properties in Non-small Cell Lung Adenocarcinoma Cells and Independently Predicts Poor Survival. Biomed Pharmacother, 2018, 100: 233–9., DOI 10.1016/j.biopha.2018.02.015 Cui F, Tang H, Tan J, Hu J. Spindle Pole Body Component 25 Regulates Stemness of Prostate Cancer Cells. Aging, Albany NY,, 2018, 10(11):3273–82., DOI 10.18632/aging.101631 Wang Q, Zhu Y, Li Z, Bu Q, Sun T, Wang H, et al. Up-regulation of SPC25 Promotes Breast Cancer. Aging, Albany NY,, 2019, 11(15): 5689–704., DOI 10.18632/aging.102153 Guo H, Zhang XY, Peng J, Huang Y, Yang Y, Liu Y, et al. RUVBL1, a Novel C-RAF-Binding Protein, Activates the RAF/MEK/ERK Pathway to Promote Lung Cancer Tumorigenesis. Biochem Biophys Res Commun, 2018, 498(4): 932–9., DOI 10.1016/j.bbrc.2018.03.084 Yuan XS,Wang ZT, Hu YJ, Bao FC, Yuan P, Zhang C, et al. Downregulation of RUVBL1 Inhibits Proliferation of Lung Adenocarcinoma Cells by G1/S Phase Cell Cycle Arrest via Multiple Mechanisms. Tumor Biol, 2016, 37:16015–27., DOI 10.1007/s13277-016-5452-9 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610481 EP 1610492 DI 10.3389/pore.2022.1610481 PG 12 ER PT J TI The Prognostic Value of Eight Immunohistochemical Markers Expressed in the Tumor Microenvironment and on Hodgkin Reed-Sternberg Cells in Pediatric Patients With Classical Hodgkin Lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; prognostic value; children; pediatric; classical Hodgkin lymphoma; immunohistochemical markers; prognostic markers ID prognosis; prognostic value; children; pediatric; classical Hodgkin lymphoma; immunohistochemical markers; prognostic markers AB Immunohistochemical markers are associated with treatment outcome in adults with classical Hodgkin Lymphoma (cHL). Studies in children are scarce and inconsistent. We investigated in 67 children with cHL, whether the expression of CD15, CD30, PAX5, PD-1, PD-L1, CD68, CD163 and TARC at diagnosis is associated with disease free survival (DFS) and with interim remission status. Low CD15 and low TARC expression were associated with relapsed disease. Low expression of PD-L1 was associated with complete remission at interim PET-scan. Our data suggest a difference between pediatric and adult cHL. This underlines the importance of future research into specific prognostic factors in pediatric cHL, indispensable for improvement of treatment in this population. CR AldinucciD, Gloghini A, PintoA,DeFilippi R, CarboneA. The ClassicalHodgkin’s Lymphoma Microenvironment and its Role in Promoting Tumour Growth and Immune Escape. J Pathol, 2010, 221(3):248–63., DOI 10.1002/path.2711 Jacobson CA, Longo DL. Hodgkin’s Lymphoma. In: JL Jameson, AS Fauci, DL Kasper, SL Hauser, DL Longo, J Loscalzo, editors. Harrison’s Principles of Internal Medicine. New York, NY: McGraw-Hill Education, 2018). Cuccaro A, Bartolomei F, Cupelli E, Hohaus S, Giachelia M, Hohaus S. Prognostic Factors in Hodgkin Lymphoma. Mediterr J Hematol Infect Dis, 2014, 6(1):e2014053., DOI 10.4084/mjhid.2014.053 Guo B, Cen H, Tan X, Ke Q. Meta-Analysis of the Prognostic and Clinical Value of Tumor-Associated Macrophages in Adult Classical Hodgkin Lymphoma. BMC Med, 2016, 14(1):159., DOI 10.1186/s12916-016-0711-6 Hollander P, Kamper P, Smedby KE, Enblad G, Ludvigsen M, Mortensen J, et al. High Proportions of PD-1+ and PD-L1+ Leukocytes in Classical Hodgkin Lymphoma Microenvironment are Associated with Inferior Outcome. Blood Adv, 2017, 1(18):1427–39., DOI 10.1182/bloodadvances.2017006346 Nguyen TT, Frater JL, Klein J, Chen L, Bartlett NL, Foyil KV, et al. Expression of TIA1 and PAX5 in Classical Hodgkin Lymphoma at Initial Diagnosis May Predict Clinical Outcome. Appl Immunohistochem Mol Morphol, 2016, 24(6): 383–91., DOI 10.1097/pai.0000000000000200 Steidl C, Lee T, Shah SP, Farinha P, Han G, Nayar T, et al. Tumor-Associated Macrophages and Survival in Classic Hodgkin’s Lymphoma. N Engl J Med, 2010, 362(10):875–85., DOI 10.1056/nejmoa0905680 Barros MHM, Hassan R, Niedobitek G. Tumor-Associated Macrophages in Pediatric Classical Hodgkin Lymphoma: Association with Epstein-Barr Virus, Lymphocyte Subsets, and Prognostic Impact. Clin Cancer Res, 2012, 18(14): 3762–71., DOI 10.1158/1078-0432.ccr-12-0129 Dinand V, Malik A, Unni R, Arya LS, Pandey RM, Dawar R. Proliferative Index and CD15 Expression in Pediatric Classical Hodgkin Lymphoma. Pediatr Blood Cancer, 2008, 50(2):280–3., DOI 10.1002/pbc.21204 Gupta S, Yeh S, Chami R, Punnett A, Chung C. The Prognostic Impact of Tumour-Associated Macrophages and Reed-Sternberg Cells in Paediatric Hodgkin Lymphoma. Eur J Cancer, 2013, 49(15):3255–61., DOI 10.1016/j. ejca.2013.05.024 Hsi ED, Li H, Nixon AB, Schoder H, Bartlett NL, LeBlanc M, et al. Serum Levels of TARC, MDC, IL-10, and Soluble CD163 in Hodgkin Lymphoma: A SWOG S0816 Correlative Study. Blood, 2019, 133(16):1762–5., DOI 10.1182/ blood-2018-08-870915 Jimenez O, Colli S, Garcia Lombardi M, Preciado MV, De Matteo E, Chabay P. Epstein-Barr Virus Recruits PDL1-Positive Cells at the Microenvironment in Pediatric Hodgkin Lymphoma. Cancer Immunol Immunother, 2021, 70(6): 1519–26., DOI 10.1007/s00262-020-02787-2 Mavili HS, Isisag A, Tan A, Miskioglu M, Baraz LS, Nese N. Relationship of Tumor-Associated Macrophage Population Detected by CD68 PG-M1, CD68 KP1, and CD163 with Latent EBV Infection and Prognosis in Classical Hodgkin Lymphoma. Turk Patoloji Derg, 2021, 37(2):130–8., DOI 10.5146/ tjpath.2020.01514 Barros MHM, Vera-Lozada G, Soares FA, Niedobitek G, Hassan R. Tumor Microenvironment Composition in Pediatric Classical Hodgkin Lymphoma Is Modulated by Age and Epstein-Barr Virus Infection. Int J Cancer, 2012, 131(5):1142–52., DOI 10.1002/ijc.27314 Keegan THM, Glaser SL, Clarke CA, Gulley ML, Craig FE, Digiuseppe JA, et al. Epstein-Barr Virus as a Marker of Survival after Hodgkin’s Lymphoma: A Population-Based Study. J Clin Oncol, 2005, 23(30):7604–13., DOI 10.1200/jco. 2005.02.6310 Nagpal P, Descalzi-Montoya DB, Lodhi N. The Circuitry of the Tumor Microenvironment in Adult and Pediatric Hodgkin Lymphoma: Cellular Composition, Cytokine Profile, EBV, and Exosomes. Cancer Rep, Hoboken,, 2021, 4(2):e1311., DOI 10.1002/cnr2.1311 RoemerMGM, Advani RH, Ligon AH, Natkunam Y, Redd RA, Homer H, et al. PD-L1 and PD-L2 Genetic Alterations Define Classical Hodgkin Lymphoma and Predict Outcome. J Clin Oncol, 2016, 34(23):2690–7., DOI 10.1200/jco. 2016.66.4482 Peh SC, Kim LH, Poppema S. TARC, a CC Chemokine, Is Frequently Expressed in Classic Hodgkin’s Lymphoma but Not in NLP Hodgkin’s Lymphoma, T-Cell-Rich B-Cell Lymphoma, and Most Cases of Anaplastic Large Cell Lymphoma. Am J Surg Pathol, 2001, 25(7):925–9., DOI 10.1097/ 00000478-200107000-00011 Zijtregtop EAM, van der Strate I, Beishuizen A, Zwaan CM, Scheijde-Vermeulen MA, Brandsma AM, et al. Biology and Clinical Applicability of Plasma Thymus and Activation-Regulated Chemokine, TARC, in ClassicalHodgkin Lymphoma. Cancers, Basel,, 2021, 13(4):884., DOI 10.3390/cancers13040884 Friedman DL, Chen L, Wolden S, Buxton A, McCarten K, FitzGerald TJ, et al. Dose-Intensive Response-Based Chemotherapy and Radiation Therapy for Children and Adolescents with Newly Diagnosed Intermediate-Risk Hodgkin Lymphoma: A Report from the Children’s Oncology Group Study AHOD0031. J Clin Oncol, 2014, 32(32):3651–8., DOI 10.1200/jco.2013.52.5410 Hakvoort-Cammel FGAJ, Buitendijk S, van den Heuvel-Eibrink M, Hahlen K. Treatment of Pediatric Hodgkin Disease Avoiding Radiotherapy: Excellent Outcome with the Rotterdam-HD-84-Protocol. Pediatr Blood Cancer, 2004, 43(1):8–16., DOI 10.1002/pbc.20031 Mauz-Korholz C, Landman-Parker J, Balwierz W, Ammann RA, Anderson RA, Attarbaschi A, et al. Response-Adapted Omission of Radiotherapy and Comparison of Consolidation Chemotherapy in Children and Adolescents With Intermediate-Stage and Advanced-Stage Classical Hodgkin Lymphoma, EuroNet-PHL-C1): A Titration Study With an Open-Label, Embedded, Multinational, Non-Inferiority, Randomised Controlled Trial. Lancet Oncol, 2022, 23(1):125–137., DOI 10.1016/S1470-2045(21)00470-8 SKION. Second International Inter-group Study for Classical Hodgkin’s Lymphoma in Children and Adolescents. E-PHsL, 2014). Available from: https://www.skion.nl/workspace/uploads/EuroNet-PHL-C2_trial_protocol_ final2-0_2015-07-27.pdf, Accessed February 20, 2022). Vassallo J, Traina F, de Souza CA, Lorand-Metze I. Expression of Epstein-Barr Virus in Classical Hodgkin’s Lymphomas in Brazilian Adult Patients. Haematologica, 2001, 86(11):1227–8. von Wasielewski R, Mengel M, Fischer R, Hansmann ML, Hubner K, Franklin J, et al. Classical Hodgkin’s Disease. Clinical Impact of the Immunophenotype. Am J Pathol, 1997, 151(4):1123–30. Wirnsberger G, Hebenstreit D, Posselt G, Horejs-Hoeck J, Duschl A. IL-4 Induces Expression of TARC/CCL17 via Two STAT6 Binding Sites. Eur J Immunol, 2006, 36(7):1882–91., DOI 10.1002/eji.200635972 Buglio D, Georgakis GV, Hanabuchi S, Arima K, Khaskhely NM, Liu Y-J, et al. Vorinostat Inhibits STAT6-Mediated TH2 Cytokine and TARC Production and Induces Cell Death in Hodgkin Lymphoma Cell Lines. Blood, 2008, 112(4):1424–33., DOI 10.1182/blood-2008-01-133769 Plattel WJ, Visser L, Diepstra A, Glaudemans AWJM, Nijland M, Meerten T, et al. Interim Thymus and Activation Regulated Chemokine versus Interim 18 F-Fluorodeoxyglucose Positron-Emission Tomography in Classical Hodgkin Lymphoma Response Evaluation. Br J Haematol, 2020, 190(1):40–4., DOI 10. 1111/bjh.16514 Green MR, Monti S, Rodig SJ, Juszczynski P, Currie T, O’Donnell E, et al. Integrative Analysis Reveals Selective 9p24.1 Amplification, Increased PD-1 Ligand Expression, and Further Induction via JAK2 in Nodular Sclerosing Hodgkin Lymphoma and Primary Mediastinal Large B-Cell Lymphoma. Blood, 2010, 116(17):3268–77., DOI 10.1182/blood-2010-05- 282780 XieW, Medeiros LJ, Li S, Yin CC, Khoury JD, Xu J. PD-1/PD-L1 Pathway and its Blockade in Patients with Classic Hodgkin Lymphoma and Non-Hodgkin Large-Cell Lymphomas. Curr Hematol Malig Rep, 2020, 15(4):372–81., DOI 10. 1007/s11899-020-00589-y Carey CD, Gusenleitner D, Lipschitz M, Roemer MGM, Stack EC, Gjini E, et al. Topological Analysis Reveals a PD-L1-Associated Microenvironmental Niche for Reed-Sternberg Cells in Hodgkin Lymphoma. Blood, 2017, 130(22): 2420–30., DOI 10.1182/blood-2017-03-770719 Shanbhag S, Ambinder RF. Hodgkin Lymphoma: A Review and Update on Recent Progress. CA Cancer J Clin, 2018, 68(2):116–32., DOI 10.3322/caac. 21438 Ramchandren R, Domingo-Domenech E, Rueda A, Trneny M, Feldman TA, Lee HJ, et al. Nivolumab for Newly Diagnosed Advanced-Stage Classic Hodgkin Lymphoma: Safety and Efficacy in the Phase II CheckMate 205 Study. J Clin Oncol, 2019, 37(23):1997–2007., DOI 10. 1200/jco.19.00315 Tan KL, Scott DW, Hong F, Kahl BS, Fisher RI, Bartlett NL, et al. Tumor- Associated Macrophages Predict Inferior Outcomes in Classic Hodgkin Lymphoma: A Correlative Study from the E2496 Intergroup Trial. Blood, 2012, 120(16):3280–7., DOI 10.1182/blood-2012-04-421057 Yoon DH, Koh YW, Kang HJ, Kim S, Park C-S, Lee S-w., et al. CD68 and CD163 as Prognostic Factors for Korean Patients with Hodgkin Lymphoma. Eur J Haematol, 2012, 88(4):292–305., DOI 10.1111/j.1600-0609.2011.01731.x Locatelli F, Mauz-Koerholz C, Neville K, Llort A, Beishuizen A, Daw S, et al. Brentuximab Vedotin for Paediatric Relapsed or Refractory Hodgkin’s Lymphoma and Anaplastic Large-Cell Lymphoma: A Multicentre, Open- Label, Phase 1/2 Study. Lancet Haematol, 2018, 5(10):e450–e461., DOI 10. 1016/s2352-3026(18)30153-4 MetzgerML, LinkMP, Billett AL, Flerlage J, Lucas JT, Mandrell BN, et al. Excellent Outcome for Pediatric Patients with High-Risk Hodgkin Lymphoma Treated with Brentuximab Vedotin and Risk-Adapted Residual Node Radiation. J Clin Oncol, 2021, 39(20):2276–83., DOI 10. 1200/jco.20.03286 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610482 EP 1610489 DI 10.3389/pore.2022.1610482 PG 8 ER PT J AU Zhao, Y Song, X Song, X Xie, L AF Zhao, Yajing Song, Xingguo Song, Xianrang Xie, Li TI Identification of Diagnostic Exosomal LncRNA-miRNA-mRNA Biomarkers in Colorectal Cancer Based on the ceRNA Network SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal cancer; lncRNA; miRNA; exosomes; diagnostic biomarker; mRNA ID colorectal cancer; lncRNA; miRNA; exosomes; diagnostic biomarker; mRNA AB Background: Colorectal cancer (CRC) is currently the fourth most common cancer worldwide. The roles of exosomal competing endogenous RNAs (ceRNAs) in CRC remain unclear. In this study, we constructed an exosomal ceRNA network to identify the core ceRNAs and investigate the diagnostic biomarkers in CRC. Methods and Patients: Serum exosomes were isolated from four CRC patients and two healthy donors by ultracentrifugation, and then subjected to RNA isolation, sequencing and microarray. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) analyses were performed to identify functional enrichment implications of differentially expressed exosomal mRNAs. TargetScan and miRanda were used for identifying the miRNA-mRNA and miRNA-LncRNA interactions. The predicted lncRNAs and mRNAs were intersected with the differentially expressed genes, for which the screening criterion was fold change >1.5 in the microarray. Differentially expressed exosomal miRNAs were identified in the GSE71008 dataset, and differentially expressed mRNAs (DEmRNAs) were further summarized from The Cancer Genome Atlas (TCGA) database. Results: A total of 1186 exosomal DEmRNAs, 2088 exosomal DElncRNAs and 29 exosomal miRNAs were detected in CRC patients compared to the healthy donors. Functional enrichment analysis suggested that exosomal DEmRNAs might participate in pathways related to carcinogenesis and development of cancer. An exosomal ceRNA regulatory network of CRC was constructed based on 40 lncRNAs, two miRNAs, and five mRNAs. Exosomal miR-150-5p and miR-10b-5p expression levels were increased in healthy donors compared with CRC patients in the GSE71008 dataset, and five DEmRNAs (TOMM70A, RBM48, BEND3, RHOBTB1, and ADAMTS2) were significantly upregulated in TCGA database. Two potential exosomal regulatory axes of lncRNA G016261-miR-150-5p-RBM48 and lncRNA XLOC_011677-miR-10b-5p-BEND3 were identified from the network. Conclusion: The current study revealed potential molecular biological regulation pathways and diagnostic biomarkers through the exosomal ceRNA regulatory network. C1 [Zhao, Yajing] Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Department of Clinical LaboratoryJinan, China. [Song, Xingguo] Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Department of Clinical LaboratoryJinan, China. [Song, Xianrang] Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Department of Clinical LaboratoryJinan, China. [Xie, Li] Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Department of Clinical LaboratoryJinan, China. RP Xie, L (reprint author), Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Department of Clinical Laboratory, Jinan, China. EM l_xie2001@126.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 De Rubis G, Rajeev Krishnan S, Bebawy M. Liquid Biopsies in Cancer Diagnosis, Monitoring, and Prognosis. Trends Pharmacol Sci, 2019, 40(3): 172–86., DOI 10.1016/j.tips.2019.01.006 Pegtel DM, Gould SJ. Exosomes. Annu Rev Biochem, 2019, 88:487–514., DOI 10.1146/annurev-biochem-013118-111902 Kulkarni B, Kirave P, Gondaliya P, Jash K, Jain A, Tekade RK, et al. Exosomal miRNA in Chemoresistance, Immune Evasion, Metastasis and Progression of Cancer. Drug Discov Today, 2019, 24:2058–67., DOI 10.1016/j.drudis.2019.06.010 Malla B, Zaugg K, Vassella E, Aebersold DM, Dal Pra A. Exosomes and Exosomal MicroRNAs in Prostate Cancer Radiation Therapy. Int J Radiat Oncol Biol Phys, 2017, 98(5):982–95., DOI 10.1016/j.ijrobp.2017.03.031 Zhang X, Yuan X, Shi H, Wu L, Qian H, Xu W. Exosomes in Cancer: Small Particle, Big Player. J Hematol Oncol, 2015, 8:83., DOI 10.1186/s13045-015- 0181-x Milman N, Ginini L, Gil Z. Exosomes and Their Role in Tumorigenesis and Anticancer Drug Resistance. Drug Resist Updat, 2019, 45:1–12., DOI 10.1016/j. drup.2019.07.003 Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, et al. Suppression of Exosomal PD-L1 Induces Systemic Anti-tumor Immunity and Memory. Cell, 2019, 177(2):414–27., DOI 10.1016/j.cell.2019.02.016 Kim J, Kim TY, Lee MS, Mun JY, Ihm C, Kim SA. Exosome Cargo Reflects TGF-β1-Mediated Epithelial-To-Mesenchymal Transition, EMT, Status in A549 Human Lung Adenocarcinoma Cells. Biochem Biophys Res Commun, 2016, 478(2):643–8., DOI 10.1016/j.bbrc.2016.07.124 Zhang W, Cai X, Yu J, Lu X, Qian Q, Qian W. Exosome-mediated Transfer of lncRNA RP11838N2.4 Promotes Erlotinib Resistance in Non-small Cell Lung Cancer. Int J Oncol, 2018, 53(2):527–38., DOI 10.3892/ijo.2018.4412 Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA Hypothesis: the Rosetta Stone of a Hidden RNA Language? Cell, 2011, 146(3):353–8., DOI 10. 1016/j.cell.2011.07.014 Thomson DW, Dinger ME. Endogenous microRNA Sponges: Evidence and Controversy. Nat Rev Genet, 2016, 17(5):272–83., DOI 10.1038/nrg. 2016.20 Xie C, Zhang LZ, Chen ZL, Zhong WJ, Fang JH, Zhu Y, et al. A Novel hMTR4- PDIA3P1-miR-125/124-TRAF6 Regulatory Axis and its Function in NF-K B Signaling and Chemoresistance. Hepatology, 2020, 71(5):1660–77., DOI 10. 1002/hep.30931 Cheng Z, Yu C, Cui S, Wang H, Jin H, Wang C, et al. circTP63 Functions as a ceRNA to Promote Lung Squamous Cell Carcinoma Progression by Upregulating FOXM1. Nat Commun, 2019, 10(1):3200., DOI 10.1038/ s41467-019-11162-4 Liu Y, XueM, Du S, FengW, Zhang K, Zhang L, et al. Competitive Endogenous RNA Is an Intrinsic Component of EMT Regulatory Circuits and Modulates EMT. Nat Commun, 2019, 10(1):1637., DOI 10.1038/s41467-019-09649-1 Zheng ZQ, Li ZX, Zhou GQ, Lin L, Zhang LL, Lv JW, et al. Long Noncoding RNA FAM225A Promotes Nasopharyngeal Carcinoma Tumorigenesis and Metastasis by Acting as ceRNA to Sponge miR-590-3p/miR-1275 and Upregulate ITGB3. Cancer Res, 2019, 79(18):4612–26., DOI 10.1158/0008- 5472.CAN-19-0799 Song YX, Sun JX, Zhao JH, Yang YC, Shi JX, Wu ZH, et al. Non-coding RNAs Participate in the Regulatory Network of CLDN4 via ceRNA Mediated miRNA Evasion. Nat Commun, 2017, 8(1):289., DOI 10.1038/ s41467-017-00304-1 Li Z, Jiang P, Li J, Peng M, Zhao X, Zhang X, et al. Tumor-derived Exosomal Lnc-Sox2ot Promotes EMT and Stemness by Acting as a ceRNA in Pancreatic Ductal Adenocarcinoma. Oncogene, 2018, 37(28):3822–38., DOI 10.1038/ s41388-018-0237-9 Barbagallo C, Brex D, Caponnetto A, Cirnigliaro M, Scalia M, Magnano A, et al. LncRNA UCA1, Upregulated in CRC Biopsies and Downregulated in Serum Exosomes, Controls mRNA Expression by RNA-RNA Interactions. Mol Ther Nucleic Acids, 2018, 12:229–41., DOI 10.1016/j.omtn.2018.05.009 Wang N, Song X, Liu L, Niu L, Wang X, Song X, et al. Circulating Exosomes Contain Protein Biomarkers of Metastatic Non-small-cell Lung Cancer. Cancer Sci, 2018, 109(5):1701–9., DOI 10.1111/cas.13581 Zhao YJ, Song X,Niu L, Tang Y, Song X, Xie L. Circulating Exosomal miR-150- 5p and miR-99b-5p as Diagnostic Biomarkers for Colorectal Cancer. Front Oncol, 2019, 9:1129., DOI 10.3389/fonc.2019.01129 Keum N, Giovannucci E. Global burden of Colorectal Cancer: Emerging Trends, Risk Factors and Prevention Strategies. Nat Rev Gastroenterol Hepatol, 2019, 16:713–32., DOI 10.1038/s41575-019-0189-8 Li X, Luo Y, Liu L, Cui S, Chen W, Zeng A, et al. The Long Noncoding RNA ZFAS1 Promotes the Progression of Glioma by Regulating the miR- 150-5p/PLP2 axis. J Cel Physiol, 2019, 235:2937–46., DOI 10.1002/jcp. 29199 Xiao G, Wang P, Zheng X, Liu D, Sun X. FAM83A-AS1 Promotes Lung Adenocarcinoma Cell Migration and Invasion by Targeting miR-150-5p and Modifying MMP14. Cell Cycle, 2019, 18:2972–85., DOI 10.1080/15384101.2019. 1664225 Li Y, Chen D, Li Y, Jin L, Liu J, Su Z, et al. Oncogenic cAMP Responsive Element Binding Protein 1 Is Overexpressed upon Loss of Tumor Suppressive miR-10b-5p and miR-363-3p in Renal Cancer. Oncol Rep, 2016, 35(4): 1967–78., DOI 10.3892/or.2016.4579 Paunescu IA, Bardan R, Marcu A, Nitusca D, Dema A, Negru S, et al. Biomarker Potential of Plasma MicroRNA-150-5p in Prostate Cancer. Medicina, Kaunas,, 2019, 55(9):E564., DOI 10.3390/medicina55090564 Zou SL, Chen YL, Ge ZZ, Qu YY, Cao Y, Kang ZX. Downregulation of Serum Exosomal miR-150-5p Is Associated with Poor Prognosis in Patients with Colorectal Cancer. Cancer Biomark, 2019, 26(1):69–77., DOI 10.3233/CBM- 190156 Chen X, Xu X, Pan B, Zeng K, Xu M, Liu X, et al. miR-150-5p Suppresses Tumor Progression by Targeting VEGFA in Colorectal Cancer. Aging, Albany NY,, 2018, 10(11):3421–37., DOI 10.18632/aging.101656 Ouyang M, Li Y, Ye S, Ma J, Lu L, Lv W, et al. MicroRNA Profiling Implies New Markers of Chemoresistance of Triple-Negative Breast Cancer. PLoS One, 2014, 9(5):e96228., DOI 10.1371/journal.pone.0096228 Jin X, Chen Y, Chen H, Fei S, Chen D, Cai X, et al. Evaluation of Tumor- Derived Exosomal miRNA as Potential Diagnostic Biomarkers for Early-Stage Non-Small Cell Lung Cancer Using Next-Generation Sequencing. Clin Cancer Res, 2017, 23(17):5311–9., DOI 10.1158/1078-0432.CCR-17-0577 Lu C, Jiang W, Hui B, Rong D, Fu K, Dong C, et al. The circ_0021977/miR- 10b-5p/P21 and P53 Regulatory axis Suppresses Proliferation, Migration, and Invasion in Colorectal Cancer. J Cel Physiol, 2020, 235(3):2273–85., DOI 10. 1002/jcp.29135 Yuan L, Zhou M, Huang D, Wasan HS, Zhang K, Sun L, et al. Resveratrol Inhibits the Invasion and Metastasis of colon Cancer through Reversal of Epithelial‑ Mesenchymal Transition via the AKT/GSK‑3β/Snail Signaling Pathway. Mol Med Rep, 2019, 20(3):2783–95., DOI 10.3892/mmr.2019.10528 Khan A, Prasanth SG. BEND3 Mediates Transcriptional Repression and Heterochromatin Organization. Transcription, 2015, 6(5):102–5., DOI 10. 1080/21541264.2015.1100228 Savci-Heijink CD, Halfwerk H, Koster J, Horlings HM, van de Vijver MJ. A Specific Gene Expression Signature for Visceral Organ Metastasis in Breast Cancer. BMC Cancer, 2019, 19(1):333., DOI 10.1186/s12885-019-5554-z Edmonson AM, Mayfield DK, Vervoort V, DuPont BR, Argyropoulos G. Characterization of a Human Import Component of the Mitochondrial Outer Membrane, TOMM70A. Cell Commun Adhes, 2002, 9(1):15–27., DOI 10.1080/ 15419060212186 Rangel R, Guzman-Rojas L, Kodama T, Kodama M, Newberg JY, Copeland NG, et al. Identification of New Tumor Suppressor Genes in Triple-Negative Breast Cancer. Cancer Res, 2017, 77:4089–101., DOI 10.1158/0008-5472.CAN-17-0785 Sotgia F, Fiorillo M, Lisanti MP. Mitochondrial Markers Predict Recurrence, Metastasis and Tamoxifen-Resistance in Breast Cancer Patients: Early Detection of Treatment Failure with Companion Diagnostics. Oncotarget, 2017, 8(40):68730–45., DOI 10.18632/oncotarget.19612 de Alcantara BBR, Cruz FM, Fonseca FLA, da Costa Aguiar Alves B, Perez MM, Varela P, et al. Chemotherapy-induced Fatigue Is Associated with Changes in Gene Expression in the Peripheral Blood Mononuclear Cell Fraction of Patients with Locoregional Breast Cancer. Support Care Cancer, 2019, 27(7):2479–86., DOI 10.1007/s00520-018-4519-0 Lin N, Zhou Y, Lian X, Tu Y. MicroRNA-31 Functions as an Oncogenic microRNA in Cutaneous Squamous Cell Carcinoma Cells by Targeting RhoTBT1. Oncol Lett, 2017, 13(3):1078–82., DOI 10.3892/ol.2017.5554 Xu RS,Wu XD, Zhang SQ, Li CF, Yang L, Li DD, et al. The Tumor Suppressor Gene RhoBTB1 Is a Novel Target of miR-31 in Human colon Cancer. Int J Oncol, 2013, 42(2):676–82., DOI 10.3892/ijo.2012.1746 Haga RB, Garg R, Collu F, Borda D’Agua B, Menendez ST, Colomba A, et al. RhoBTB1 Interacts with ROCKs and Inhibits Invasion. Biochem J, 2019, 476(17):2499–514., DOI 10.1042/BCJ20190203 Kirana C, Peng L, Miller R, Keating JP, Glenn C, Shi H, et al. Combination of Laser Microdissection, 2D-DIGE and MALDI-TOF MS to Identify Protein Biomarkers to Predict Colorectal Cancer Spread. Clin Proteomics, 2019, 16:3., DOI 10.1186/s12014-019-9223-7 Jiang C, Zhou Y, Huang Y, Wang Y, Wang W, Kuai X. Overexpression of ADAMTS-2 in Tumor Cells and Stroma Is Predictive of Poor Clinical Prognosis in Gastric Cancer. Hum Pathol, 2019, 84:44–51., DOI 10.1016/j. humpath.2018.08.030 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610493 EP 1610503 DI 10.3389/pore.2022.1610493 PG 11 ER PT J AU Wang, L Xiao, Sh Zheng, Y Gao, Z AF Wang, Longyue Xiao, Shuaishuai Zheng, Yiming Gao, Zefeng TI Interaction Between Vascular Endothelial Growth Factor Gene Polymorphism and Smoking on Gastric Cancer Risk in Chinese Han Population SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gastric cancer; smoking; interaction; single nucleotide polymorphisms; vascular endothelial growth factor ID gastric cancer; smoking; interaction; single nucleotide polymorphisms; vascular endothelial growth factor AB Aim: In this study, we aimed to evaluate the associations of vascular endothelial growth factor (VEGF) gene single nucleotide polymorphisms (SNPs) and its interaction with current smoking with gastric cancer (GC) risk in the Chinese Han population. Methods: We used logistic regression model to test the association between VEGF gene polymorphism and the risk of GC. The association strength was evaluated by odds ratio (OR) and 95% confidence interval (CI) calculated using logistic regression. Generalized multifactor dimensionality reduction (GMDR) was used to analyze the effect of the interaction between VEGF gene and current smoking on GC risk. Results: Logistic regression analysis showed that the risk of GC was significantly higher in rs10434 -G allele carriers than that in AA genotype carriers (AG + GG and AA), and the adjusted OR (95% CI) = 1.64 (1.24–2.08). In addition, we found a significantly higher GC risk in subjects with rs833061-T allele than those with CC allele (CT + TT and CC), adjusted or (95% CI) = 1.43 (1.10–1.87). We also found a statistically significant two- locus model (p = 0.018), including rs10434 and current smoking, indicating a significant interaction between rs10434 and current smoking on the risk of GC. Hierarchical analysis found that current smokers with AG or GG genotype have the highest GC risk, compared to neversmokers with AA genotype, OR (95% CI) = 2.43 (1.64–3.28). Conclusion: We found that rs10434 -G and rs833061-T alleles, gene- environment interaction between rs10434, and current smoking were all related to increased GC risk. C1 [Wang, Longyue] Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Department of General Surgery TwoTaiyuan, China. [Xiao, Shuaishuai] Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Department of General Surgery TwoTaiyuan, China. [Zheng, Yiming] Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Department of General Surgery TwoTaiyuan, China. [Gao, Zefeng] Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Department of General Surgery TwoTaiyuan, China. RP Wang, L (reprint author), Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Department of General Surgery Two, Taiyuan, China. EM wanglongyue661@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Rawla P, Barsouk A. Epidemiology of Gastric Cancer: Global Trends, Risk Factors and Prevention. Prz Gastroenterol, 2019, 14(1):26–38., DOI 10.5114/pg. 2018.80001 Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric Cancer: Descriptive Epidemiology, Risk Factors, Screening, and Prevention. Cancer Epidemiol Biomarkers Prev, 2014, 23:700–13., DOI 10.1158/1055-9965. EPI-13-1057 Alipour M. Molecular Mechanism of Helicobacter Pylori-Induced Gastric Cancer. J Gastrointest Cancer, 2021, 52(1):23–30., DOI 10.1007/s12029-020- 00518-5 Alagappan VK, Willems-Widyastuti A, Seynhaeve AL, Garrelds IM, ten Hagen TL, Saxena PR, et al. Vasoactive Peptides Upregulate mRNA Expression and Secretion of Vascular Endothelial Growth Factor in Human Airway Smooth Muscle Cells. Cell Biochem Biophys, 2007, 47(1):109–18., DOI 10.1385/cbb:47:1:109 Eroglu A, Ersoz C, Karasoy D, Sak S. Vascular Endothelial Growth Factor, VEGF)-C, VEGF-D, VEGFR-3 and D2-40 Expressions in Primary Breast Cancer: Association with Lymph Node Metastasis. Adv Clin Exp Med, 2017, 26(2):245–9., DOI 10.17219/acem/58784 Vincenti V, Cassano C, Rocchi M, Persico G. Assignment of the Vascular Endothelial Growth Factor Gene to Human Chromosome 6p21.3. Circulation, 1996, 93:1493–5., DOI 10.1161/01.cir.93.8.1493 Fu D, Li P, Cheng W, Tian F, Xu X, Yi X, et al. Impact of Vascular Endothelial Growth Factor Gene-Gene and Gene-Smoking Interaction and Haplotype Combination on Bladder Cancer Risk in Chinese Population. Oncotarget, 2017, 8(14):22927–35., DOI 10.18632/oncotarget.15287 Naykoo NA, Dil-Afroze, Rasool R, Shah S, Ahangar AG, Bhat IA, et al. Single Nucleotide Polymorphisms, Haplotype Association and Tumour Expression of the Vascular Endothelial Growth Factor, VEGF, Gene with Lung Carcinoma. Gene, 2017, 608:95–102., DOI 10.1016/j.gene.2017.01.007 Ratnasari N, Nurdjanah S, Sadewa AH, Hakimi M. The Role of Vascular Endothelial Growth Factor -634 G/C and its Soluble Receptor on Chronic Liver Disease and Hepatocellular Carcinoma. Arab J Gastroenterol, 2016, 17(2):61–6., DOI 10.1016/j.ajg.2016.06.005 Rezaei M, Hashemi M, Sanaei S, Mashhadi MA, Taheri M. Association between Vascular Endothelial Growth Factor Gene Polymorphisms with Breast Cancer Risk in an Iranian Population. Breast Cancer, Auckl,, 2016, 10: 85–91., DOI 10.4137/BCBCR.S39649 Tie Z, Bai R, Zhai Z, Zhang G, Zhang H, Zhao Z, et al. Single Nucleotide Polymorphisms in VEGF Gene are Associated with an Increased Risk of Osteosarcoma. Int J Clin Exp Pathol, 2014, 7(11):8143–9. Xu HM, Xu LF, Hou TT, Luo LF, Chen GB, Sun XW, et al. GMDR: Versatile Software for Detecting Gene-Gene and Gene-Environment Interactions Underlying Complex Traits. Curr Genomics, 2016, 17(5):396–402., DOI 10. 2174/1389202917666160513102612 Zhou Y, Li N, ZhuangW,Wu X. Vascular Endothelial Growth Factor, VEGF, Gene Polymorphisms and Gastric Cancer Risk in a Chinese Han Population. Mol Carcinog, 2011, 50(3):184–8., DOI 10.1002/mc.20703 Tahara T, Shibata T, Nakamura M, Yamashita H, Yoshioka D, Hirata I, et al. Effect of Polymorphisms in the 3’ Untranslated Region, 3’-UTR, of Vascular Endothelial Growth Factor Gene on Gastric Cancer and Peptic Ulcer Diseases in Japan. Mol Carcinog, 2009, 48(11):1030–7., DOI 10.1002/mc.20554 Hong TT, Cai DY, Wu XH, Hua D. Three Vascular Endothelial Growth Factor Polymorphisms, −460C>T, −2578C>a, 1612G>A, with Cancer Risk: A Meta- Analysis Based on 30 Case-Control Studies. Cancer Investig, 2011, 29(7): 472–7., DOI 10.3109/07357907.2011.597811 Zhuang M, Peng Z, Wang J, Su X. Vascular Endothelial Growth Factor Gene Polymorphisms and Gastric Cancer Risk: A Meta-Analysis. J BUON, 2017, 22(3):714–24. Liu W, Dong Z, Hu R, Wang C. Association of Vascular Endothelial Growth Factor, VEGF, Gene Polymorphisms with Gastric Cancer and its Development, Prognosis, and Survival. Technol Cancer Res Treat, 2018, 17: 1533034617753810., DOI 10.1177/1533034617753810 Zhou LP, Luan H, Dong XH, Jin GJ, Man DL, Shang H. Vascular Endothelial Growth Factor +936C/T Polymorphism and Gastric Cancer Risk: A Meta-Analysis. Exp Ther Med, 2011, 2(5):931–6., DOI 10.3892/etm. 2011.278 Takahashi Y, Cleary KR, Mai M, Kitadai Y, Bucana CD, Ellis LM. Significance of Vessel Count and Vascular Endothelial Growth Factor and its Receptor, KDR, in Intestinal-type Gastric Cancer. Clin Cancer Res, 1996, 2:1679–84. Maeda K, Kang SM, Onoda N, Ogawa M, Kato Y, Sawada T, et al. Vascular Endothelial Growth Factor Expression in Preoperative Biopsy Specimens Correlates with Disease Recurrence in Patients with Early Gastric Carcinoma. Cancer, 1999, 86:566–71., DOI 10.1002/(sici)1097- 0142(19990815)86:4<566::aid-cncr4>3.0.co;2-1 Feng CW, Wang LD, Jiao LH, Liu B, Zheng S, Xie XJ. Expression of P53, Inducible Nitric Oxide Synthase and Vascular Endothelial Growth Factor in Gastric Precancerous and Cancerous Lesions: Correlation with Clinical Features. BMC Cancer, 2000, 29:8., DOI 10.1186/1471-2407-2-8 Lao X, Peng Q, Lu Y, Li S, Qin X, Chen Z, et al. Glutathione S-Transferase Gene GSTM1, Gene-Gene Interaction, and Gastric Cancer Susceptibility: Evidence from an Updated Meta-Analysis. Cancer Cel Int, 2014, 14(1):127., DOI 10.1186/ s12935-014-0127-3 Praud D, Rota M, Pelucchi C, Bertuccio P, Rosso T, Galeone C, et al. Cigarette Smoking and Gastric Cancer in the Stomach Cancer Pooling, StoP, Project. Eur J Cancer Prev, 2018, 27(2):124–33., DOI 10.1097/CEJ.0000000000000290 Kim SA, Choi BY, Song KS, Park CH, Eun CS, Han DS, et al. Prediagnostic Smoking and Alcohol Drinking and Gastric Cancer Survival: A Korean Prospective Cohort Study. Korean J Gastroenterol, 2019, 73(3):141–51., DOI 10.4166/kjg.2019.73.3.141 Ferro A, Morais S, Rota M, Pelucchi C, Bertuccio P, Bonzi R, et al. Tobacco Smoking and Gastric Cancer: Meta-Analyses of Published Data versus Pooled Analyses of Individual Participant Data, StoP Project). Eur J Cancer Prev, 2018, 27(3):197–204., DOI 10.1097/CEJ.0000000000000401 Ugur MG, Kutlu R, Kilinc I. The Effects of Smoking on Vascular Endothelial Growth Factor and Inflammation Markers: A Case-Control Study. Clin Respir J, 2018, 12(5):1912–8., DOI 10.1111/crj.12755 Rovina N, Papapetropoulos A, Kollintza A, Michailidou M, Simoes DC, Roussos C, et al. Vascular Endothelial Growth Factor: an Angiogenic Factor Reflecting Airway Inflammation in Healthy Smokers and in Patients with Bronchitis Type of Chronic Obstructive Pulmonary Disease? Respir Res, 2007, 8(1):53., DOI 10.1186/1465-9921-8-53 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610495 EP 1610500 DI 10.3389/pore.2022.1610495 PG 6 ER PT J AU Chien, ChYCh Beke, L Mehes, G Mokanszki, A AF Chien, Chang Yi-Che Beke, Livia Mehes, Gabor Mokanszki, Attila TI Anastomosing Haemangioma: Report of Three Cases With Molecular and Immunohistochemical Studies and Comparison With Well-Differentiated Angiosarcoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE angiosarcoma; anastomosing haemangioma; immunohistochemistry (IHC); fluorescence in situ hybridization (FISH); next-generation sequencing (NGS); GNA11 mutation ID angiosarcoma; anastomosing haemangioma; immunohistochemistry (IHC); fluorescence in situ hybridization (FISH); next-generation sequencing (NGS); GNA11 mutation AB Anastomosing haemangioma (AH) is a newly described distinct vascular neoplasm that histologically may confuse with well-differentiated angiosarcoma (AS) for those who are unfamiliar with this rare entity. We aimed to identify molecular genetic differences between AHs and ASs by carrying out immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), and next-generation sequencing (NGS). Immunohistochemically, all six cases showed positivity for cyclinD1 and pERK. All cases of AH showed focal weak positive reaction for p53 and MIB-1, and the IHCs for HIF-1α were all negative for all three cases. Those three cases of angiosarcoma revealed strong, diffuse positivity for p53, 50%–70% MIB-1 labelling, and multifocal, moderate to strong HIF-1α expression. To further clarify the difference in p53 expression, we carried out a FISH which revealed 17p polysomy in all three ASs whereas copy number aberration was absent in the AH group. In one AH case, the GNA11 c.627G > T nucleotide variant was detected. Due to the rarity and overlapping morphological features, AH might be difficult to separate from other vascular tumours, in particular from well-differentiated AS also featured by mild hyperchromatic, hobnail-like endothelial cells. The potential molecular differences between these two entities presented here may be used in support of the correct diagnosis. C1 [Chien, Chang Yi-Che] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Beke, Livia] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Mokanszki, Attila] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. RP Chien, ChYCh (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, Debrecen, Hungary. EM dr.changchien.yiche@med.unideb.hu CR Montgomery E, Epstein JI. Anastomosing Hemangioma of the Genitourinary Tract. Am J Surg Pathol, 2009, 33:1364–9., DOI 10.1097/PAS. 0b013e3181ad30a7 Lin J, Bigge J, Ulbright TM, Montgomery E. Anastomosing Hemangioma of the Liver and Gastrointestinal Tract. Am J Surg Pathol, 2013, 37:1761–5., DOI 10.1097/PAS.0b013e3182967e6c John I, Folpe AL. Anastomosing Hemangiomas Arising in Unusual Locations. Am J Surg Pathol, 2016, 40:1084–9., DOI 10.1097/PAS.0000000000000627 Bean GR, Joseph NM, Gill RM, Folpe AL, Horvai AE, Umetsu SE. Recurrent GNAQ Mutations in Anastomosing Hemangiomas. Mod Pathol, 2017, 30: 722–7., DOI 10.1038/modpathol.2016.234 Lim YH, Bacchiocchi A, Qiu J, Straub R, Bruckner A, Bercovitch L, et al. GNA14 Somatic Mutation Causes Congenital and Sporadic Vascular Tumors by MAPK Activation. Am J Hum Genet, 2016, 99:443–50., DOI 10.1016/j.ajhg. 2016.06.010 Liau J-Y, Lee J-C, Tsai J-H, Chen C-C, Chung Y-C, Wang Y-H. High Frequency of GNA14, GNAQ, and GNA11 Mutations in Cherry Hemangioma: a Histopathological and Molecular Study of 85 Cases Indicating GNA14 as the Most Commonly Mutated Gene in Vascular Neoplasms. Mod Pathol, 2019, 32:1657–65., DOI 10.1038/s41379-019-0284-y Cheon PM, Rebello R, Naqvi A, Popovic S, Bonert M, Kapoor A. Anastomosing Hemangioma of the Kidney: Radiologic and Pathologic Distinctions of a Kidney Cancer Mimic. Curr Oncol, 2018, 25:220–3., DOI 10.3747/co.25.3927 Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L, O’Brien JM, et al. Frequent Somatic Mutations of GNAQ in Uveal Melanoma and Blue Naevi. Nature, 2009, 457:599–602., DOI 10.1038/nature07586 Urtatiz O, Haage A, Tanentzapf G, Van Raamsdonk CD. Crosstalk with Keratinocytes Causes GNAQ Oncogene Specificity in Melanoma. Elife, 2021, 10:e71825., DOI 10.7554/eLife.71825 Ayturk UM, Couto JA, Hann S, Mulliken JB, Williams KL, Huang AY, et al. Somatic Activating Mutations in GNAQ and GNA11 Are Associated with Congenital Hemangioma. Am J Hum Genet, 2016, 98:1271., DOI 10.1016/j.ajhg. 2016.05.010 Couto JA, Ayturk UM, Konczyk DJ, Goss JA, Huang AY, Hann S, et al. A Somatic GNA11 Mutation Is Associated with Extremity Capillary Malformation and Overgrowth. Angiogenesis, 2017, 20:303–6., DOI 10.1007/ s10456-016-9538-1 Flores K, Yadav SS, Katz AA, Seger R. The Nuclear Translocation of Mitogen- Activated Protein Kinases: Molecular Mechanisms and Use as Novel Therapeutic Target. Neuroendocrinology, 2019, 108:121–31., DOI 10.1159/ 000494085 Xu F, Zhao L-J, Liao T, Li Z-C, Wang L-L, Lin P-Y, et al. Ononin Ameliorates Inflammation and Cartilage Degradation in Rat Chondrocytes with IL-1β-induced Osteoarthritis by Downregulating the MAPK and NF-κB Pathways. BMC Complement Med Ther, 2022, 22:25., DOI 10.1186/s12906-022-03504-5 Tsai H-L, Yeh Y-S, Chang Y-T, Yang I-P, Lin C-H, Kuo C-H, et al. Coexistence of Cyclin D1 and Vascular Endothelial Growth Factor Protein Expression Is a Poor Prognostic Factor for UICC Stage I-III Colorectal Cancer Patients after Curative Resection. J Surg Oncol, 2013, 107:148–54., DOI 10.1002/jso.23243 Salter DMM, GriffinM, CulleyTeo JSJR, Gomez-Cuadrado LMK, Sims AHHL, Henderson NCBVG. Development of Mouse Models of Angiosarcoma Driven by P53. Dis Model Mech, 2019, 12:dmm038612., DOI 10.1242/dmm.038612 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610498 EP 1610503 DI 10.3389/pore.2022.1610498 PG 6 ER PT J TI The Role of Tumor Microenvironment and Immune Response in Colorectal Cancer Development and Prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE colorectal cancer; tumor microenvironment; immune cells; tumorigenesis; consensus molecular subtypes; immunoscore ID colorectal cancer; tumor microenvironment; immune cells; tumorigenesis; consensus molecular subtypes; immunoscore AB Colorectal cancer (CRC) is one of the most common cancers worldwide. The patient’s prognosis largely depends on the tumor stage at diagnosis. The pathological TNM Classification of Malignant Tumors (pTNM) staging of surgically resected cancers represents the main prognostic factor and guidance for decision-making in CRC patients. However, this approach alone is insufficient as a prognostic predictor because clinical outcomes in patients at the same histological tumor stage can still differ. Recently, significant progress in the treatment of CRC has been made due to improvements in both chemotherapy and surgical management. Immunotherapybased approaches are one of the most rapidly developing areas of tumor therapy. This review summarizes the current knowledge about the tumor microenvironment (TME), immune response and its interactions with CRC development, immunotherapy and prognosis. CR Creasy JM, Sadot E, Koerkamp BG, Chou JF, Gonen M, Kemeny NE, et al. Actual 10-year Survival after Hepatic Resection of Colorectal Liver Metastases: What Factors Preclude Cure? Surgery, 2018, 163:1238–44., DOI 10.1016/j.surg.2018.01.004 Tomasek J, Kiss I. Immunotherapy of Colorectal and Anal Cancer. Klin Onkol, 2017, 30(3):3562–5., DOI 10.14735/amko20173S62 Grady WM, Pritchard CC. Molecular Alterations and Biomarkers in Colorectal Cancer. Toxicol Pathol, 2014, 42:124–39., DOI 10.1177/ 0192623313505155 McGranahan N, Furness AJS, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, et al. Clonal Neoantigens Elicit T Cell Immunoreactivity and Sensitivity to Immune Checkpoint Blockade. Science, 2016, 351(6280):1463–9., DOI 10. 1126/science.aaf1490 Tsang AHF, Cheng KH, Wong ASP, Ng SS, Ma BB, Chan CML, et al. Current and Future Molecular Diagnostics in Colorectal Cancer and Colorectal Adenoma. World J Gastroenterol, 2014, 20:3847–57., DOI 10.3748/wjg.v20. i14.3847 Damilakis E, Mavroudis D, Sfakianaki M, Souglakos J. Immunotherapy in Metastatic Colorectal Cancer: Could the Latest Developments Hold the Key to Improving Patient Survival? Cancers, 2020, 12:889., DOI 10.3390/ cancers12040889 Advani SM, Advani PS, Brown DW, DeSantis SM, Korphaisarn K, VonVille HM, et al. Global Differences in the Prevalence of the CpG Island Methylator Phenotype of Colorectal Cancer. BMC Cancer, 2019, 19:964., DOI 10.1186/ s12885-019-6144-9 Guinney J, Dienstmann R, Wang X, de Reynies A, Schlicker A, Soneson C, et al. The Consensus Molecular Subtypes of Colorectal Cancer. Nat Med, 2015, 21:1350–6., DOI 10.1038/nm.3967 Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A, Ruschoff J, et al. Revised Bethesda Guidelines for Hereditary Nonpolyposis Colorectal Cancer, Lynch Syndrome, and Microsatellite Instability. J Natl Cancer Inst, 2004, 96:261–8., DOI 10.1093/jnci/djh034 Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM, et al. The Vigorous Immune Microenvironment of Microsatellite Instable colon Cancer is Balanced by Multiple Counter-inhibitory Checkpoints. Cancer Discov, 2015, 5(1):43–51., DOI 10.1158/2159- 8290.CD-14-0863 Fridman WH, Zitvogel L, Sautes-Fridman C, Kroemer G. The Immune Contexture in Cancer Prognosis and Treatment. Nat Rev Clin Oncol, 2017, 14:717–34., DOI 10.1038/nrclinonc.2017.101 Fiegle E, Doleschel D, Koletnik S, Rix A, Weiskirchen R, Borkham- Kamphorst E, et al. Dual CTLA-4 and PD-L1 Blockade Inhibits Tumor Growth and Liver Metastasis in a Highly Aggressive Orthotopic Mouse Model of Colon Cancer. Neoplasia, 2019, 21:932–44., DOI 10.1016/j.neo.2019. 07.006 Imai K, Yamamoto H. Carcinogenesis and Microsatellite Instability: The Interrelationship between Genetics and Epigenetics. Carcinogenesis, 2008, 29:673–80., DOI 10.1093/carcin/bgm228 Lynch H, Lynch P, Lanspa S, Snyder C, Lynch J, Boland C, et al. Review of the Lynch Syndrome: History, Molecular Genetics, Screening, Differential Diagnosis, and Medicolegal Ramifications. Clin Genet, 2009, 76:1–18., DOI 10.1111/j.1399-0004.2009.01230.x Jenkins MA, Hayashi S, O´Shea AM, Burgart LJ, Smyrk TC, Shimizu D, et al. Pathology Features in Bethesda Guidelines Predict Colorectal Cancer Microsatellite Instability: A Population-Based Study. Gastroenterology, 2007, 133:48–56., DOI 10.1053/j.gastro.2007.04.044 Nosho K, Baba Y, Tanaka N, Shima K, Hayashi M, Meyerhardt JA, et al. Tumor-infiltrating T-Cell Subsets, Molecular Changes in Colorectal Cancer, and Prognosis: Cohort Study and Literature Review. J Pathol, 2010, 12: 298–306., DOI 10.1002/path.2774 Ogino S, Nosho K, Irahara N, Meyerhardt JA, Baba Y, Shima K, et al. Lymphocytic Reaction to Colorectal Cancer is Associated with Longer Survival, Independent of Lymph Node Count, Microsatellite Instability, and CpG Island Methylator Phenotype. Clin Cancer Res, 2009, 15: 6412–20., DOI 10.1158/1078-0432.CCR-09-1438 Fabrizio DA, George TJ, Dunne RF, Frampton G, Sun J, Gowen K, et al. Beyond Microsatellite Testing: Assessment of Tumor Mutational burden Identifies Subsets of Colorectal Cancer Who May Respond to Immune Checkpoint Inhibition. J Gastrointest Oncol, 2018, 9(4):610–7., DOI 10. 21037/jgo.2018.05.06 Salama P, Phillips M, Grieu F, Morris M, Zeps N, Joseph D, et al. Tumorinfiltrating FOXP3+ T Regulatory Cells Show strong Prognostic Significance in Colorectal Cancer. J Clin Oncol, 2009, 27:186–92., DOI 10.1200/JCO.2008. 18.7229 Angell HK, Bruni D, Barett JC, Herbst R, Galon J. The Immunoscore: Colon Cancer and beyond. Clin Cancer Res, 2020, 26:332–9., DOI 10.1158/1078- 0432.CCR-18-1851 Krummel MF, Allison JP. CD28 and CTLA-4 Have Opposing Effects on the Response of T Cells to Stimulation. J Exp Med, 1995, 182:459–65., DOI 10. 1084/jem.182.2.459 Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and Activity of Anti-PD-L1 Antibody in Patients with Advanced Cancer. N Engl J Med, 2012, 366(26):2455–65., DOI 10.1056/NEJMoa1200694 Pardoll DM. The Blockade of Immune Checkpoints in Cancer Immunotherapy. Nat Rev Cancer, 2012, 4:252–64., DOI 10.1038/nrc3239 Han Y, Yang Y, Chen Z, Jiang Z, Gu Y, Liu Y, et al. Human Hepatocellular Carcinoma-Infiltrating CD4+ CD69+ Foxp3- Regulatory T Cell Suppresses T Cell Response via Membrane-Bound TGF-Beta1. J Mol Med, 2014, 95(5): 539–50., DOI 10.1007/s00109-014-1143-4 Svensson H, Olofsson V, Lundin S, Yakkala C, Bjorck S, Borjesson L, et al. Accumulation of CCR4+CTLA-4hi FOXP3+CD25hi Regulatory T Cells in colon Adenocarcinomas Correlate to Reduced Activation of Conventional T Cells. PLoS One, 2012, 7:e30695., DOI 10.1371/journal.pone.0030695 Liu Y, Xia T, Jin CH, Gu D, Yu J, Shi W, et al. FOXP3 and CEACAM6 Expression and T Cell Infiltration in the Occurrence and Development of colon Cancer. Oncol Lett, 2016, 11:3693–701., DOI 10. 3892/ol.2016.4439 Deschoolmeester V, Baay M, Lardon F, Pauwels P, Peeters M. Immune Cells in Colorectal Cancer: Prognostic Relevance and Role of MSI. Cancer Microenviron, 2011, 4:377–92., DOI 10.1007/s12307-011- 0068-5 Xu W, Liu H, Song J, Fu HX, Qiu L, Zhang BF, et al. The Appearance of Tregs in Cancer Nest is a Promising Independent Risk Factor in colon Cancer. J Cancer Res Clin Oncol, 2013, 139:1845–52., DOI 10.1007/s00432-013-1500-7 Poschke I, Mougiakakos D, Kiessling R. Camouflage and Sabotage: Tumor Escape from the Immune System. Cancer Immunol Immunother, 2011, 60: 1161–71., DOI 10.1007/s00262-011-1012-8 Fife BT, Bluestone JA. Control of Peripheral T-Cell Tolerance and Autoimmunity via the CTLA-4 and PD-1 Pathways. Immunol Rev, 2008, 224:166–82., DOI 10.1111/j.1600-065X.2008.00662.x Takasu C, Nishi M, Yoshikawa K, Tokunaga T, Kashihara H, Yoshimoto T, et al. Impact of Sidedness of Colorectal Cancer on Tumor Immunity. PLoS ONE, 2020, 15(10):e0240408., DOI 10.1371/journal.pone.0240408 Fontenot JD, Rasmussen JP,Williams LM, Dooley JL, Farr AG, Rudensky AY, et al. Regulatory T Cell Lineage Specification by the Forkhead Transcription Factor Foxp3. Immunity, 2005, 22:329–41., DOI 10.1016/j.immuni.2005. 01.016 Li W, Wang L, Katoh H, Liu R, Zheng P, Liu Y, et al. Identification of a Tumor Suppressor Relay between the FOXP3 and the Hippo Pathways in Breast and Prostate Cancers. Cancer Res, 2011, 71:2162–71., DOI 10.1158/0008-5472. CAN-10-3268 Zuo T, Liu R, Zhang H, Chang X, Liu Y, Wang L, et al. FOXP3 is a Novel Transcriptional Repressor for the Breast Cancer Oncogene SKP2. J Clin Invest, 2007, 117:3765–73., DOI 10.1172/JCI32538 Kim M, Grimmig T, Grimm M, Lazariotou M, Meier E, Rosenwald A, et al. Expression of Foxp3 in Colorectal Cancer but Not in Treg Cells Correlates with Disease Progression in Patients with Colorectal Cancer. PLoS One, 2013, 8(1):e53630., DOI 10.1371/journal.pone.0053630 Yoon HH, Orrock JM, Foster NR, Sargent DJ, Smyrk TC, Sinicrope FA, et al. Prognostic Impact of FoxP3+ Regulatory T Cells in Relation to CD8+ T Lymphocyte Density in Human Colon Carcinomas. PLoS ONE, 2012, 7(8): e42274., DOI 10.1371/journal.pone.0042274 Ling A, Edin S, Wikberg ML, Oberg A, Palmqvist R. The Intratumoural Subsite and Relation of CD8(+, and FOXP3(+, T Lymphocytes in Colorectal Cancer Provide Important Prognostic Clues. Br J Cancer, 2014, 110:2551–9., DOI 10.1038/bjc.2014.161 Ladoire S, Martin F, Ghiringhelli F. Prognostic Role of FOXP3+ Regulatory T Cells Infiltrating Human Carcinomas: The Paradox of Colorectal Cancer. Cancer Immunol Immunother, 2011, 60(7):909–18., DOI 10.1007/s00262-011- 1046-y Sharpe AH, Pauken KE. The Diverse Functions of the PD1 Inhibitory Pathway. Nat Rev Immunol, 2018, 18:153–67., DOI 10.1038/nri.2017.108 Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its Ligands in Tolerance and Immunity. Annu Rev Immunol, 2008, 26:677–704., DOI 10. 1146/annurev.immunol.26.021607.090331 Okazaki T, Honjo T. The PD-1-PD-L Pathway in Immunological Tolerance. Trends Immunol, 2006, 27:195–201., DOI 10.1016/j.it.2006.02.001 Bardhan K, Anagnostou T, Boussiotis VA. The PD1:PD-L1/2 Pathway from Discovery to Clinical Implementation. Front Immunol, 2016, 7:550., DOI 10. 3389/fimmu.2016.00550 Greenfield EA, Nguyen KA, Kuchroo VK. CD28/B7 Costimulation: A Review. Crit Rev Immunol, 1998, 18(5):389–418., DOI 10.1615/ critrevimmunol.v18.i5.10 Calik I, Calik M, Turken G, Ozercan IH, Dagli AF, Artas G, et al. Intratumoral Cytotoxic T-Lymphocyte Density and PD-L1 Expression Are Prognostic Biomarkers for Patients with Colorectal Cancer. Medicina, 2019, 55:723., DOI 10.3390/medicina55110723 Berntsson J, Eberhard J, Nodin B, Leandersson K, Larsson AH, Jirstrom K, et al. Expression of Programmed Cell Death Protein 1, PD-1, and its Ligand PD-L1 in Colorectal Cancer: Relationship with Sidedness and Prognosis. Oncoloimmunology, 2018, 7:e1465165., DOI 10.1080/2162402X.2018.1465165 Yi M, Niu M, Xu L, Luo S, Wu K. Regulation of PD-L1 Expression in the Tumor Microenvironment. J Hematol Oncol, 2021, 14:10., DOI 10.1186/ s13045-020-01027-5 Akinleye A, Rasool Z. Immune Checkpoint Inhibitors of PD-L1 as Cancer Therapeutics. J Hematol Oncol, 2019, 12:92., DOI 10.1186/s13045-019-0779-5 Noh BJ, Kwak JY, Eom DW. Immune Classification for the PD-L1 Expression and Tumour-Infiltrating Lymphocytes in Colorectal Adenocarcinoma. BMC Cancer, 2020, 20:58., DOI 10.1186/s12885-020-6553-9 Panda A, Betigeri A, Subramanian K, Ross JS, Pavlick DC, Ali S, et al. Identifying a Clinically Applicable Mutational Burden Threshold as a Potential Biomarker of Response to Immune Checkpoint Therapy in Solid Tumors. JCO Precis Oncol, 2017, 2017:1–13., DOI 10.1200/PO.17.00146 Liu S, Gӧnen M, Stadler ZK, Weiser MR, Hechtman JF, Vakiani E, et al. Cellular Localization of PD-L1 Expression in Mismatch-Repair-Deficient and Proficient Colorectal Carcinomas. Mod Pathol, 2019, 32:110–21., DOI 10. 1038/s41379-018-0114-7 Li Y, Liang L, DaiW, Cai G, Xu Y, Li X, et al. Prognostic Impact of Programed Cell Death-1, PD-1, and PD-Ligand 1, PD-L1, Expression in Cancer Cells and Tumor Infiltrating Lymphocytes in Colorectal Cancer. Mol Cancer, 2016, 15:55., DOI 10.1186/s12943-016-0539-x Ahtiainen M, Wirta EV, Kuopio T, Seppala T, Rantala J, Mecklin JP, et al. Combined Prognostic Value of CD274, PD-L1)/PDCDI, PD-1, Expression and Immune Cell Infiltration in Colorectal Cancer as Per Mismatch Repair Status. Mod Pathol, 2019, 32:866–83., DOI 10.1038/ s41379-019-0219-7 Le DT, Uram JN,Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med, 2015, 372:2509–20., DOI 10.1056/NEJMoa1500596 Shen Z, Gu L, Mao D, Chen M, Jin R. Clinicopathological and Prognostic Significance of PD-L1 Expression in Colorectal Cancer: A Systematic Review and Meta-Analysis. World J Surg Oncol, 2019, 17:4., DOI 10.1186/s12957-018- 1544-x Buckowitz A, Knaebel HP, Benner A, Blaker H, Gebert J, Kienle P, et al. Microsatellite Instability in Colorectal Cancer Is Associated with Local Lymphocyte Infiltration and Low Frequency of Distant Metastases. Br J Cancer, 2005, 92:1746–53., DOI 10.1038/sj.bjc.6602534 Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, et al. Immunotherapy in Colorectal Cancer: Rationale, Challenges and Potential. Nat Rev Gastroenterol Hepatol, 2019, 16:361–75., DOI 10.1038/s41575-019- 0126-x Yu G, Wu Y, Wang W, Xu J, Lv X, Cao X, et al. Low-dose Decitabine Enhances the Effect of PD-1 Blockade in Colorectal Cancer with Microsatellite Stability by Re-modulating the Tumor Microenvironment. Cell Mol Immunol, 2019, 16(4):401–9., DOI 10.1038/s41423-018-0026-y Venderbosch S, Nagtegaal ID, Maughan TS, Smith CG, Cheadle JP, Fisher D, et al. Mismatch Repair Status and BRAF Mutation Status in Metastatic Colorectal Cancer Patients: A Pooled Analysis of the CAIRO, CAIRO2, COIN, and FOCUS Studies. Clin Cancer Res, 2014, 20:5322–30., DOI 10.1158/ 1078-0432.CCR-14-0332 Wherry EJ. T Cell Exhaustion. Nat Immunol, 2011, 12:492–9., DOI 10.1038/ni. 2035 Zarour HM. Reversing T-Cell Dysfunction and Exhaustion in Cancer. Clin Cancer Res, 2016, 22:1856–64., DOI 10.1158/1078-0432.CCR-15-1849 Yassin M, Sadowska Z, Djurhuus D, Nielsen B, Tougaard P, Olsen J, et al. Upregulation of PD-1 Follows Tumour Development in the AOM/DSS Model of Inflammation-Induced Colorectal Cancer in Mice. Immunology, 2019, 158:35–46., DOI 10.1111/imm.13093 Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, Shi J, et al. Human Gut Microbiome and Risk for Colorectal Cancer. J Natl Cancer Inst, 2013, 105: 1907–11., DOI 10.1093/jnci/djt300 Huipeng W, Lifeng G, Chuang G, Jiaying Z, Yuankun C. The Differences in Colonic Mucosal Microbiota between normal Individual and colon Cancer Patients by Polymerase Chain Reaction-Denaturing Gradient Gel Electrophoresis. J Clin Gastroenterol, 2014, 48:138–44., DOI 10.1097/MCG. 0b013e3182a26719 Kuwahara T, Hazama S, Suzuki N, Yoshida S, Tomochika S, Nakagami Y, et al. Intratumoural-infiltrating CD4 + and FOXP3 + T Cells as strong Positive Predictive Markers for the Prognosis of Resectable Colorectal Cancer. Br J Cancer, 2019, 121:659–65., DOI 10.1038/s41416-019-0559-6 Bourgeois C, Veiga-Fernandes H, Joret AM, Rocha B, Tanchot C. CD8 Lethargy in the Absence of CD4 Help. Eur J Immunol, 2002, 32: 2199–207., DOI 10.1002/1521-4141(200208)32:8<2199:AID-IMMU2199>3.0. CO;2-L Kurts C, Carbone FR, Barnden M, Blanas E, Allison J, Heath WR, et al. CD4+ T Cell Help Impairs CD8+ T Cell Deletion Induced by Cross-Presentation of Self-Antigens and Favors Autoimmunity. J Exp Med, 1997, 186:2057–62., DOI 10.1084/jem.186.12.2057 Xie Y, Akpinarli A, Maris C, Hipkiss EL, Lane M, Kwon EK, et al. Naive Tumor-specific CD4(+, T Cells Differentiated In Vivo Eradicate Established Melanoma. J Exp Med, 2010, 207:651–67., DOI 10.1084/jem.20091921 Gianchecchi E, Delfino DV, Fierabracci A. Recent Insights into the Role of the PD-1/PD-L1 Pathway in Immunological Tolerance and Autoimmunity. Autoimmun Rev, 2013, 12:1091–100., DOI 10.1016/j.autrev.2013.05.003 Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, et al. Rescue of Exhausted CD8 T Cells by PD-1-Targeted Therapies is CD28- dependent. Science, 2017, 355:1423–7., DOI 10.1126/science.aaf0683 Gajewski TF, Schreiber H, Fu YX. Innate and Adaptive Immune Cells in the Tumor Microenvironment. Nat Immunol, 2013, 14:1014–22., DOI 10.1038/ni. 2703 Palucka AK, Coussens LM. The Basis of Oncoimmunology. Cell, 2016, 164: 1233–47., DOI 10.1016/j.cell.2016.01.049 Sharma P, Allison JP. The Future of Immune Checkpoint Therapy. Science, 2015, 348:56–61., DOI 10.1126/science.aaa8172 Reissfelder C, Stamova S, Gossmann C, Braun M, Bonertz A, Walliczek U, et al. Tumor-specific Cytotoxic T Lymphocyte Activity Determines Colorectal Cancer Patient Prognosis. J Clin Invest, 2015, 125:739–51., DOI 10.1172/JCI74894 Kwak Y, Koh J, Kim DW, Kang SB, Kim WH, Lee HS, et al. Immunoscore Encompassing CD3+ and CD8+ T Cell Densities in Distant Metastasis is a Robust Prognostic Marker for Advanced Colorectal Cancer. Oncotarget, 2016, 7:81778–90., DOI 10.18632/oncotarget.13207 Miller TJ, Anyaegbu CC, Lee-Pullen TF, Spalding LJ, Platell CF, McCoy MJ, et al. PD-L1+ Dendritic Cells in the Tumor Microenvironment Correlate with Good Prognosis and CD8+ T Cell Infiltration in colon Cancer. Cancer Sci, 2021, 112:1173–83., DOI 10.1111/cas.14781 Dahlin AM, Henriksson ML, Van Guelpen B, Stenling R, Oberg A, Rutegard J, et al. Colorectal Cancer Prognosis Depends on T-Cell Infiltration and Molecular Characteristics of the Tumor. Mod Pathol, 2011, 24:671–82., DOI 10.1038/modpathol.2010.234 Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pages C, et al. Type, Density, and Location of Immune Cells within Human Colorectal Tumors Predict Clinical Outcome. Science, 2006, 313:1960–4., DOI 10.1126/science.1129139 Huang CY, Chiang SF, Ke TW, Chen TW, You YS, Chen WT, et al. Clinical Significance of Programmed Death 1 Ligand-1, CD274/PD-L1, and Intratumoral CD8+ T-Cell Infiltration in Stage II-III Colorectal Cancer. Sci Rep, 2018, 8:15658., DOI 10.1038/s41598-018-33927-5 Anfossi N, Andre P, Guia S, Falk CS, Roetynck S, Stewart CA, et al. Human NK Cell Education by Inhibitory Receptors for MHC Class I. Immunity, 2006, 25:331–42., DOI 10.1016/j.immuni.2006.06.013 Coppola A, Arriga R, Lauro D, Del PrincipeMI, Buccisano F, Maurillo L, et al. NK Cell Inflammation in the Clinical Outcome of Colorectal Carcinoma. Front Med, Lausanne,, 2015, 2:33., DOI 10.3389/fmed.2015.00033 Schwartz M, Zhang Y, Rosenblatt JD. B Cell Regulation of the Anti-tumor Response and Role in Carcinogenesis. J Immunother Cancer, 2016, 4:40., DOI 10.1186/s40425-016-0145-x Shimabukuro-Vornhagen A, Schloßer HA, Gryschok L, Malcher J, Wennhold K, Garcia-Marquez M, et al. Characterization of Tumor- Associated B-Cell Subsets in Patients with Colorectal Cancer. Oncotarget, 2014, 5:4651–64., DOI 10.18632/oncotarget.1701 Toor SM, Sasidharan Nair V, Murshed K, Abu Nada M, Elkord E. Tumor- Infiltrating Lymphoid Cells in Colorectal Cancer Patients with Varying Disease Stages and Microsatellite Instability-High/Stable Tumors. Vaccines, Basel,, 2021, 9:64., DOI 10.3390/vaccines9010064 Nirschl CJ, Drake CG. Molecular Pathways: Coexpression of Immune Checkpoint Molecules: Signaling Pathways and Implications for Cancer Immunotherapy. Clin Cancer Res, 2013, 19:4917–24., DOI 10.1158/1078- 0432.CCR-12-1972 Ren D, Hua Y, Yu B, Ye X, He Z, Li C, et al. Predictive Biomarkers and Mechanisms Underlying Resistance to PD1/PD-L1 Blockade Cancer Immunotherapy. Mol Cancer, 2020, 19:19., DOI 10.1186/s12943-020-1144-6 Turvey SE, Broide DH. Innate Immunity. J Allergy Clin Immunol, 2010, 125: S24–S32., DOI 10.1016/j.jaci.2009.07.016 Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated Macrophages as Major Players in the Tumor Microenvironment. Cancers, Basel,, 2014, 6(3): 1670–90., DOI 10.3390/cancers6031670 Sica A, Mantovani A. Macrophage Plasticity and Polarization: In Vivo Veritas. J Clin Invest, 2012, 122(3):787–95., DOI 10.1172/JCI59643 Ngambenjawong C, Gustafson HH, Pun SH. Progress in Tumor-Associated Macrophage, TAM)-targeted Therapeutics. Adv Drug Deliv Rev, 2017, 114: 206–21., DOI 10.1016/j.addr.2017.04.010 Liu Y, Cao X. The Origin and Function of Tumor-Associated Macrophages. Cel Mol Immunol, 2015, 12:1–4., DOI 10.1038/cmi.2014.83 Qian BZ, Pollard JW. Macrophage Diversity Enhances Tumor Progression and Metastasis. Cell, 2010, 141:39–51., DOI 10.1016/j.cell.2010.03.014 Lian G, Chen S, Ouyang M, Li F, Chen L, Yang J, et al. Colon Cancer Cell Secretes EGF to Promote M2 Polarization of TAM through EGFR/PI3K/ AKT/mTOR Pathway. Technol Cancer Res Treat, 2019, 18: 1533033819849068., DOI 10.1177/1533033819849068 Kobie JJ, Wu RS, Kurt RA, Lou S, Adelman MK, Whitesell LJ, et al. Transforming Growth Factor Inhibits the Antigen-Presenting Functions and Antitumor Activity of Dendritic Cell Vaccines. Cancer Res, 2003, 63(8):1860–4. Orsini G, Legitimo A, Failli A, Ferrari P, Nicolini A, Spisni R, et al. Defective Generation and Maturation of Dendritic Cells from Monocytes in Colorectal Cancer Patients during the Course of Disease. Int J Mol Sci, 2013, 14(11): 22022–41., DOI 10.3390/ijms141122022 Hsu YL, Chen YJ, Chang WA, Jian SF, Fan HL, Wang JY, et al. Interaction between Tumor-Associated Dendritic Cells and colon Cancer Cells Contributes to Tumor Progression via CXCL1. Int J Mol Sci, 2018, 19(8): 2427., DOI 10.3390/ijms19082427 Della Porta M, Danova M, Rigolin GM, Brugnatelli S, Rovati B, Tronconi C, et al. Dendritic Cells and Vascular Endothelial Growth Factor in Colorectal Cancer: Correlations with Clinicobiological Findings. Oncology, 2005, 68(2- 3):276–84., DOI 10.1159/000086784 Ammendola M, Sacco R, Sammarco G, Donato G, Montemurro S, Ruggieri E, et al. Correlation between Serum Tryptase, Mast Cells Positive to Tryptase and Microvascular Density in colo-rectal Cancer Patients: Possible Biological-Clinical Significance. PLoS One, 2014, 9(6):e99512., DOI 10.1371/ journal.pone.0099512 Suzuki S, Ichikawa Y, Nakagawa K, Kumamoto T, Mori R, Matsuyama R, et al. High Infiltration of Mast Cells Positive to Tryptase Predicts Worse Outcome Following Resection of Colorectal Liver Metastases. BMC Cancer, 2015, 15:840., DOI 10.1186/s12885-015-1863-z Zhao P, Zhou P, Tang T, Si R, Ji Y, Hu X, et al. Levels of CirculatingMast Cell Progenitors and Tumour-Infiltrating Mast Cells in Patients with Colorectal Cancer. Oncol Rep, 2022, 47:89., DOI 10.3892/or.2022.8300 Sinha P, Chornoguz O, Clements VK, Artemenko KA, Zubarev RA, Ostrand- Rosenberg S, et al. Myeloid-derived Suppressor Cells Express the Death Receptor Fas and Apoptose in Response to T Cell-Expressed FasL. Blood, 2011, 117(20):5381–90., DOI 10.1182/blood-2010-11-321752 Fleming V, Hu X, Weber R, Nagibin V, Groth C, Altevogt P, et al. Targeting Myeloid-Derived Suppressor Cells to Bypass Tumor-Induced Immunosuppression. Front Immunol, 2018, 9:398., DOI 10.3389/fimmu. 2018.00398 Dosset M, Vargas TR, Lagrange A, Boidot R, Vegran F, Roussey A, et al. PD- 1/PD-L1 Pathway: An Adaptive Immune Resistance Mechanism to Immunogenic Chemotherapy in Colorectal Cancer. Oncoimmunology, 2018, 7(6):e1433981., DOI 10.1080/2162402X.2018.1433981 McClellan JL, Davis JM, Steiner JL, Enos RT, Jung SH, Carson JA, et al. Linking Tumor-Associated Macrophages, Inflammation, and Intestinal Tumorigenesis: Role of MCP-1. Am J Physiol Gastrointest Liver Physiol, 2012, 303(10):G1087–95., DOI 10.1152/ajpgi.00252.2012 Vermeulen L, De Sousa EMelo F, van der Heijden M, Cameron K, de Jong JH, Borovski T, et al. Wnt Activity Defines colon Cancer Stem Cells and is Regulated by the Microenvironment. Nat Cel Biol, 2010, 5:468–76., DOI 10. 1038/ncb2048 Hawinkels LJ, PaauweM, Verspaget HW, Wiercinska E, van der Zon JM, van der Ploeg K, et al. Interaction with colon Cancer Cells Hyperactivates TGF-β Signaling in Cancer-Associated Fibroblasts. Oncogene, 2014, 1:97–107., DOI 10.1038/onc.2012.536 Vellinga TT, den Uil S, Rinkes IH, Marvin D, Ponsioen B, Alvarez-Varela A, et al. Collagen-rich Stroma in Aggressive colon Tumors Induces Mesenchymal Gene Expression and Tumor Cell Invasion. Oncogene, 2016, 35:5263–71., DOI 10.1038/onc.2016.60 Kalluri R. The Biology and Function of Fibroblasts in Cancer. Nat Rev Cancer, 2016, 9:582–98., DOI 10.1038/nrc.2016.73 Mele V, Muraro MG, Calabrese D, Pfaff D, Amatruda N, Amicarella F, et al. Mesenchymal Stromal Cells Induce Epithelial-To-Mesenchymal Transition in Human Colorectal Cancer Cells through the Expression of Surface-Bound TGF-β. Int J Cancer, 2014, 11:2583–94., DOI 10.1002/ijc.28598 Brabletz T, Kalluri R, Nieto MA, Weinberg RA. EMT in Cancer. Nat Rev Cancer, 2018, 18:128–34., DOI 10.1038/nrc.2017.118 Burger GA, Danen EHJ, Beltman JB. Deciphering Epithelial-Mesenchymal Transition Regulatory Networks in Cancer through Computational Approaches. Front Oncol, 2017, 7:162., DOI 10.3389/fonc.2017.00162 Jiang Y, Zhan H. Communication between EMT and PD-L1 Signaling: New Insights into Tumor Immune Evasion. Cancer Lett, 2020, 468:72–81., DOI 10. 1016/j.canlet.2019.10.013 Terry S, Savagner P, Ortiz-Cuaran S, Mahjoubi L, Saintigny P, Thiery JP, et al. New Insights into the Role of EMT in Tumor Immune Escape. Mol Oncol, 2017, 11:824–46., DOI 10.1002/1878-0261.12093 Zhan HX, Zhou B, Cheng YG, Xu JW, Wang L, Zhang GY, et al. Crosstalk between Stromal Cells and Cancer Cells in Pancreatic Cancer: New Insights into Stromal Biology. Cancer Lett, 2017, 392:83–93., DOI 10.1016/j.canlet. 2017.01.041 Ma HY, Liu XZ, Liang CM. Inflammatory Microenvironment Contributes to Epithelial-Mesenchymal Transition in Gastric Cancer. World J Gastroenterol, 2016, 22:6619–28., DOI 10.3748/wjg.v22.i29.6619 ZippiM,De TomaG,Minervini G, Cassieri C, Pica R, ColarussoD, et al. Desmoplasia Influenced Recurrence of Disease and Mortality in Stage III Colorectal Cancer within Five Years after Surgery and Adjuvant Therapy. Saudi J Gastroenterol, 2017, 23:39–44., DOI 10.4103/1319- 3767.199114 Ueno H, Shinto E, Shimazaki H, Kajiwara Y, Sueyama T, Yamamoto J, et al. Histologic Categorization of Desmoplastic Reaction: Its Relevance to the Colorectal Cancer Microenvironment and Prognosis. Ann Surg Oncol, 2015, 22:1504–12., DOI 10.1245/s10434-014-4149-9 Sueyama T, Kajiwara Y, Mochizuki S, Shimazaki H, Shinto E, Hase K, et al. Periostin as a Key Molecule Defining Desmoplastic Environment in Colorectal Cancer. Virchows Arch, 2021, 478:865–74., DOI 10.1007/s00428- 020-02965-8 Becht E, de Reynies A, Giraldo NA, Pilati C, Buttard B, Lacroix L, et al. Immune and Stromal Classification of Colorectal Cancer is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy. Clin Cancer Res, 2016, 22(16):4057–66., DOI 10.1158/1078-0432.CCR-15-2879 Tougeron D, Fauquembergue E, Rouquette A, Le Pessot F, Sesboue R, Laurent M, et al. Tumor-infiltrating Lymphocytes in Colorectal Cancers with Microsatellite Instability are Correlated with the Number and Spectrum of Frameshift Mutations. Mod Pathol, 2009, 22(9):1186–95., DOI 10.1038/ modpathol.2009.80 Hodi FS, O’Day SJ, McDermott DF,Weber RW, Sosman JA, Haanen JB, et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N Engl J Med, 2010, 363(8):711–23., DOI 10.1056/NEJMoa1003466 Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, Activity, and Immune Correlates of Anti-PD-1 Antibody in Cancer. N Engl J Med, 2012, 366(26):2443–54., DOI 10.1056/NEJMoa1200690 Fan J, Shang D, Han B, Song J, Chen H, Yang JM, et al. Adoptive Cell Transfer: Is it a Promising Immunotherapy for Colorectal Cancer? Theranostics, 2018, 8:5784–800., DOI 10.7150/thno.29035 Beak JH, Kim KJ. Expansion of Tumor-Infiltrating Lymphocytes and Their Potential for Application to Adoptive Cell Therapy in Patients with Colorectal Cancer. Eur J Surg Oncol, 2019, 45(2):e58–9., DOI 10.1016/j.ejso. 2018.10.223 Koelzer VH, Lugli A, Dawson H, Hadrich M, Berger MD, Borner M, et al. CD8/CD45RO T-Cell Infiltration in Endoscopic Biopsies of Colorectal Cancer Predicts Nodal Metastasis and Survival. J Transl Med, 2014, 12: 81., DOI 10.1186/1479-5876-12-81 Morris M, Platell C, Iacopetta B. Tumor-infiltrating Lymphocytes and Perforation in colon Cancer Predict Positive Response to 5-fluorouracil Chemotherapy. Clin Cancer Res, 2008, 14(5):1413–7., DOI 10.1158/1078- 0432.CCR-07-1994 Halama N, Michel S, Kloor M, Zoernig I, Benner A, Spille A, et al. Localization and Density of Immune Cells in the Invasive Margin of Human Colorectal Cancer Liver Metastases are Prognostic for Response to Chemotherapy. Cancer Res, 2011, 71(17):5670–7., DOI 10.1158/0008-5472. CAN-11-0268 Valenzuela G, Canepa J, Simonetti C, Solo de Zaldivar L, Marcelain K, Gonzalez-Montero J, et al. Consensus Molecular Subtypes of Colorectal Cancer in Clinical Practice: A Translational Approach. World J Clin Oncol, 2021, 12(11):1000–8., DOI 10.5306/wjco.v12.i11.1000 Eide PW, Moosavi SH, Eilertsen IA, Brunsell TH, Langerud J, Berg KCG, et al. Metastatic Heterogeneity of the Consensus Molecular Subtypes of Colorectal Cancer. NPJ Genom Med, 2021, 6:59., DOI 10.1038/s41525-021-00223-7 Fontana E, Eason K, Cervantes A, Salazar R, Sadanandam A. Context Matters-Consensus Molecular Subtypes of Colorectal Cancer as Biomarkers for Clinical Trials. Ann Oncol, 2019, 30(4):520–7., DOI 10. 1093/annonc/mdz052 Kamal Y, Schmit SL, Hoehn HJ, Amos CI, Frost HR. Transcriptomic Differences between Primary Colorectal Adenocarcinomas and Distant Metastases Reveal Metastatic Colorectal Cancer Subtypes. Cancer Res, 2019, 79:4227–41., DOI 10.1158/0008-5472.CAN-18-3945 Akishima-Fukasawa Y, Ino Y, Nakanishi Y, Miura A, Moriya Y, Kondo T, et al. Significance of PGP9.5 Expression in Cancer-Associated Fibroblasts for Prognosis of Colorectal Carcinoma. Am J Clin Pathol, 2010, 134:71–9., DOI 10. 1309/AJCPRJP39MIDSGBH Glentis A, Oertle P, Mariani P, Chikina A, El Marjou F, Attieh Y, et al. Cancer-associated Fibroblasts Induce Metalloprotease-independent Cancer Cell Invasion of the BasementMembrane. Nat Commun, 2017, 8:924., DOI 10. 1038/s41467-017-00985-8 Ren J, Ding L, Zhang D, Shi G, Xu Q, Shen S, et al. Carcinoma-associated Fibroblasts Promote the Stemness and Chemoresistance of Colorectal Cancer by Transferring Exosomal lncRNA H19. Theranostics, 2018, 8(14):3932–48., DOI 10.7150/thno.25541 Zhang R, Qi F, Shao S, Li G, Feng Y. Human Colorectal Cancer-Derived Carcinoma Associated Fibroblasts Promote CD44-Mediated Adhesion of Colorectal Cancer Cells to Endothelial Cells by Secretion of HGF. Cancer Cel Int, 2019, 19:192., DOI 10.1186/s12935-019-0914-y Miyazaki K, Togo S, Okamoto R, Idiris A, Kumagai H, Miyagi Y, et al. Collective Cancer Cell Invasion in Contact with Fibroblasts through Integrin- α5β1/fibronectin Interaction in Collagen Matrix. Cancer Sci, 2020, 111: 4381–92., DOI 10.1111/cas.14664 Unterleuthner D, Neuhold P, Schwarz K, Janker L, Neuditschko B, Nivarthi H, et al. Cancer-associated Fibroblast-Derived WNT2 Increases Tumor Angiogenesis in Colon Cancer. Angiogenesis, 2020, 23:159–77., DOI 10. 1007/s10456-019-09688-8 Bauer K, Michel S, Reuschenbach M, Nelius N, von Knebel Doeberitz M, Kloor M, et al. Dendritic Cell and Macrophage Infiltration in Microsatellite- Unstable andMicrosatellite -stable Colorectal Cancer. Fam Cancer, 2011, 10: 557–65., DOI 10.1007/s10689-011-9449-7 Gulubova MV, Ananiev JR, Vlaykova TI, Yovchev Y, Tsoneva V, Manolova IM, et al. Role of Dendritic Cells in Progression and Clinical Outcome of colon Cancer. Int J Colorectal Dis, 2012, 27:159–69., DOI 10.1007/s00384-011- 1334-1 Hu Z, Ma Y, Shang Z, Hu S, Liang K, Liang W, et al. Improving Immunotherapy for Colorectal Cancer Using Dendritic Cells Combined with Anti-programmed Death-Ligand In Vitro. Oncol Lett, 2018, 15: 5345–51., DOI 10.3892/ol.2018.7978 Berntsson J, Nodin B, Eberhard J, Micke P, Jirstrom K. Prognostic Impact of Tumour-Infiltrating B Cells and Plasma Cells in Colorectal Cancer. Int J Cancer, 2016, 139:1129–39., DOI 10.1002/ijc.30138 Edin S, Kaprio T, Hagstrom J, Larsson P, Mustonen H, Bockelman C, et al. The Prognostic Importance of CD20+ B Lymphocytes in Colorectal Cancer and the Relation to Other Immune Cell Subsets. Sci Rep, 2019, 9:19997., DOI 10.1038/s41598-019-56441-8 Mullins CS, Gock M, Krohn M, Linnebacher M. Human Colorectal Carcinoma Infiltrating B Lymphocytes are Active Secretors of the Immunoglobulin Isotypes A, G, and M. Cancers, Basel,, 2019, 11:776., DOI 10.3390/cancers11060776 Berntsson J, Svensson MC, Leandersson K, Nodin B, Micke P, Larsson AH, et al. The Clinical Impact of Tumour-Infiltrating Lymphocytes in Colorectal Cancer Differs by Anatomical Subsite: A Cohort Study. Int J Cancer, 2017, 141:1654–66., DOI 10.1002/ijc.30869 Digiacomo N, Bolzacchini E, Veronesi G, Cerutti R, Sahnane N, Pinotti G, et al. Neuroendocrine Differentiation, Microsatellite Instability, and Tumor- Infiltrating Lymphocytes in Advanced Colorectal Cancer with BRAF Mutation. Clin Colorectal Cancer, 2019, 18(2):e251–60., DOI 10.1016/j.clcc. 2018.12.003 Glaire MA, Domingo E, Sveen A, Bruun J, Nesbakken A, Nicholson G, et al. Tumour-infiltrating CD8+ Lymphocytes and Colorectal Cancer Recurrence by Tumour and Nodal Stage. Br J Cancer, 2019, 121:474–82., DOI 10.1038/ s41416-019-0540-4 Craig SG, Humphries MP, Alderdice M, Bingham V, Richman SD, Loughrey MB, et al. Immune Status Is Prognostic for Poor Survival in Colorectal Cancer Patients and is Associated with Tumour Hypoxia. Br J Cancer, 2020, 123: 1280–8., DOI 10.1038/s41416-020-0985-5 Hartman DJ, Frank M, Seigh L, Choudry H, Pingpank J, Holtzman M, et al. Automated Quantitation of CD8-Positive T Cells Predicts Prognosis in Colonic Adenocarcinoma with Mucinous, Signet Ring Cell, or Medullary Differentiation Independent of Mismatch Repair Protein Status. Am J Surg Pathol, 2020, 44:991–1001., DOI 10.1097/PAS.0000000000001468 Fuchs TL, Sioson L, Sheen A, Jafari-Nejad K, Renaud CJ, Andrici J, et al. Assessment of Tumor-Infiltrating Lymphocytes Using International TILs Working Group, ITWG, System is a Strong Predictor of Overall Survival in Colorectal Carcinoma: A Study of 1034 Patients. Am J Surg Pathol, 2020, 44: 536–44., DOI 10.1097/PAS.0000000000001409 Lalos A, Tulek A, Tosti N, Mechera R, Wilhelm A, Soysal S, et al. Prognostic Significance of CD8+ T-Cells Density in Stage III Colorectal Cancer Depends Od SDF-1 Expression. Sci Rep, 2021, 11:775., DOI 10.1038/s41598-020- 80382-2 Al-Badran SS, Grant L, Campo MV, Inthagard J, Pennel K, Quinn J, et al. Relationship between Immune Checkpoint Proteins, Tumour Microenvironment Characteristics, and Prognosis in Primary Operable Colorectal Cancer. J Pathol Clin Res, 2021, 7:121–34., DOI 10.1002/cjp2.193 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610502 EP 1610513 DI 10.3389/pore.2022.1610502 PG 12 ER PT J AU Zhou, M Chen, Y Gu, X Wang, C AF Zhou, Min Chen, Yan Gu, Xuyu Wang, Cailian TI A Comprehensive Bioinformatic Analysis for Identification of Myeloid-Associated Differentiation Marker as a Potential Negative Prognostic Biomarker in Non-Small-Cell Lung Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bioinformatics; biomarker; NSCLC; survival-related gene; MYADM ID bioinformatics; biomarker; NSCLC; survival-related gene; MYADM AB Objectives: This study aimed to identify a molecular marker associated with the prognosis of non-small-cell lung cancer (NSCLC). Materials and Methods: The RNA sequencing data and clinical information of NSCLC patients were obtained from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO). The weighted gene co-expression network analysis (WGCNA) was used to identify the co-expression gene modules and differentially expressed genes (DEGs) by comparing gene expression between NSCLC tumor tissues and normal tissues. Subsequently, the functional enrichment analysis of the DEGs was performed. Kaplan- Meier survival analysis and the GEPIA2 online tool were performed to investigate the relationship between the expression of these genes of interest and the survival of NSCLC patients, and to validate one most survival-relevent hub gene, as well as validated the hub gene using independent datasets from the GEO database. Further analysis was carried out to characterize the relationship between the hub gene and tumor immune cell infiltration, tumor mutation burden (TMB), microsatellite instability (MSI), and other known biomarkers of lung cancer. The related genes were screened by analyzing the protein-protein interaction (PPI) network and the survival model was constructed. GEPIA2 was applied in the potential analysis of pan-cancer biomarker of hub gene. Results: 57 hub genes were found to be involved in intercellular connectivity from the 779 identified differentially co-expressed genes. Myeloid-associated differentiation marker (MYADM) was strongly associated with overall survival (OS) and disease-free survival (DFS) of NSCLC patients, and high MYADM expression was associated with poor prognosis. Thus, MYADM was identified as a risk factor. Additionally, MYADM was validated as a survival risk factor in NSCLC patients in two independent datasets. Further analysis showed that MYADM was nagetively associated with TMB, and was positively correlated with macrophages, neutrophils, and dendritic cells, suggesting its role in regulating tumor immunity. The MYADM expression differed across many types of cancer and had the potential to serve as a pan-cancer marker. Conclusion: MYADM is an independent prognostic factor for NSCLC patients, which can predict the progression of cancer and play a role in the tumor immune cell infiltration in NSCLC. C1 [Zhou, Min] Southeast University, School of Medicine, Zhongda Hospital, Department of OncologyNanjing, China. [Chen, Yan] Southeast University, School of Medicine, Zhongda Hospital, Department of OncologyNanjing, China. [Gu, Xuyu] Southeast University, School of Medicine, Zhongda Hospital, Department of OncologyNanjing, China. [Wang, Cailian] Southeast University, School of Medicine, Zhongda Hospital, Department of OncologyNanjing, China. RP Wang, C (reprint author), Southeast University, School of Medicine, Zhongda Hospital, Department of Oncology, Nanjing, China. EM wangcailian65@hotmail.com CR Molina JR, Yang P, Cassivi SD, Schild SE, Adjei AA. Non-Small Cell Lung Cancer: Epidemiology, Risk Factors, Treatment, and Survivorship. Mayo Clin Proc, 2008, 83(5):584–94., DOI 10.4065/83.5.584 Garcia MA, Nelson WJ, Chavez N. Cell-Cell Junctions Organize Structural and Signaling Networks. Cold Spring Harb Perspect Biol, 2018, 10:a029181., DOI 10. 1101/cshperspect.a029181 Wu M, Tong X, Wang D, Wang L, Fan H. Soluble Intercellular Cell Adhesion Molecule-1 in Lung Cancer: AMeta-Analysis. Pathol Res Pract, 2020, 216(10): 153029., DOI 10.1016/j.prp.2020.153029 Liang J, Oyang L, Rao S, Han Y, Luo X, Yi P, et al. Rac1, a Potential Target for Tumor Therapy. Front Oncol, 2021, 11:674426., DOI 10.3389/fonc.2021. 674426 Liu Y, Xie J. Accurate and Efficient P-Value Calculation via Gaussian Approximation: A Novel Monte-Carlo Method. J Am Stat Assoc, 2019, 114:525384–392., DOI 10.1080/01621459.2017.1407776 Cui W, Yu L, He H, Chu Y, Gao J, Wan B, et al. Cloning of Human Myeloid- Associated Differentiation Marker, MYADM, Gene Whose Expression Was Up-Regulated in NB4 Cells Induced by All-Trans Retinoic Acid. Mol Biol Rep, 2001, 28(3):123–38., DOI 10.1023/a:1015288412047 Dannaeus K, Bessonova M, Jonsson J-I. Characterization of the Mouse Myeloid-Associated Differentiation Marker, Myadm, Gene: Promoter Analysis and Protein Localization. Mol Biol Rep, 2005, 32(3):149–57., DOI 10.1007/s11033-005-0753-x Aranda JF, Reglero-Real N, Kremer L, Marcos-Ramiro B, Ruiz-Saenz A, Calvo M, et al. MYADM Regulates Rac1 Targeting to Ordered Membranes Required for Cell Spreading and Migration. Mol Biol Cel, 2011, 22(8):1252–62., DOI 10. 1091/mbc.E10-11-0910 Wang Q, Li N, Wang X, Shen J, Hong X, Yu H, et al. Membrane Protein hMYADM Preferentially Expressed in Myeloid Cells Is Up-Regulated during Differentiation of Stem Cells and Myeloid Leukemia Cells. Life Sci, 2007, 80(5):420–9., DOI 10.1016/j.lfs.2006.09.043 Lorenzo PI, Brendeford EM, Gilfillan S, Gavrilov AA, Leedsak M, Razin SV, et al. Identification of c-Myb Target Genes in K562 Cells Reveals a Role for c-Myb as a Master Regulator. Genes Cancer, 2011, 28:805–17., DOI 10.1177/1947601911428224 de Wit NJ, Rijntjes J, Diepstra JH, van Kuppevelt TH, Weidle UH, Ruiter DJ, et al. Analysis of Differential Gene Expression in HumanMelanocytic Tumour Lesions by Custom Made Oligonucleotide Arrays. Br J Cancer, 2005, 92: 2249–61., DOI 10.1038/sj.bjc.6602612 Megger DA, Naboulsi W, Meyer HE, Sitek B. Proteome Analyses of Hepatocellular Carcinoma. J Clin Transl Hepatol, 2014, 2:23–30., DOI 10. 14218/JCTH.2013.00022 Papasotiriou I, Apostolou P, Ntanovasilis DA, Parsonidis P, Osmonov D, Junemann KP. Study and Detection of Potential Markers for Predicting Metastasis into Lymph Nodes in Prostate Cancer. Biomark Med, 2020, 1414:1317–27., DOI 10.2217/bmm-2020-0372 Echevarria MI, Awasthi S, Cheng C-H, AndersBerglundRounbehler ERJ, Gerke TA, Davicioni E, et al. African American Specific Gene Panel Predictive of Poor Prostate Cancer Outcome. J Urol, 2019, 2022:247–55., DOI 10.1097/JU.0000000000000193 McGrail DJ, Pilie PG, Rashid NU, Voorwerk L, Slagter M, Kok M, et al. High Tumor Mutation Burden Fails to Predict Immune Checkpoint Blockade Response across All Cancer Types. Ann Oncol, 2021, 32(5):661–72., DOI 10. 1016/j.annonc.2021.02.006 De Vita F, Infusino S, Auriemma A, Orditura M, Catalano G. Circulating Levels of Soluble Intercellular Adhesion Molecule-1 in Non-small Cell Lung Cancer Patients. Oncol Rep, 1998, 5(2):393–6., DOI 10.3892/or.5. 2.393 Akunuru S, James ZQ, Zheng Y. Non-Small Cell Lung Cancer Stem/ Progenitor Cells Are Enriched in Multiple Distinct Phenotypic Subpopulations and Exhibit Plasticity. Cell Death Dis, 2012, 3(7):e352., DOI 10.1038/cddis.2012.93 Aranda JF, Reglero-Real N, Marcos-Ramiro B, Ruiz-Saenz A, Fernandez-Martin L, Bernabe-Rubio M, et al. MYADM Controls Endothelial Barrier Function through ERM-dependent Regulation of ICAM-1 Expression. Mol Biol Cel, 2013, 24(4):483–94., DOI 10.1091/ mbc.E11-11-0914 Luo K, Li Y, Yin Y, Li L, Wu C, Chen Y, et al. USP49 Negatively Regulates Tumorigenesis and Chemoresistance through FKBP51-AKT Signaling. EMBO JOURNAL, 2017, 36(10):1434–46., DOI 10.15252/embj. 201695669 Tu R, Kang W, Yang X, Zhang Q, Xie X, Liu W, et al. USP49 Participates in the DNA Damage Response by Forming a Positive Feedback Loop with P53. Cel Death Dis, 2018, 9(5):553., DOI 10.1038/s41419-018-0475-3 Berger A, Le Fourn V, Masternak J, Regamey A, Bodenmann I, Girod PA, et al. Overexpression of Transcription Factor Foxa1 and Target Genes Remediate Therapeutic Protein Production Bottlenecks in Chinese Hamster Ovary Cells. Biotechnol Bioeng, 2020, 117(4):1101–16., DOI 10.1002/bit.27274 Pourcel L, Buron F, Arib G, Le Fourn V, Regamey A, Bodenmann I, et al. Influence of Cytoskeleton Organization on Recombinant Protein Expression by CHO Cells. Biotechnol Bioeng, 2020, 117(4):1117–26., DOI 10.1002/bit. 27277 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610504 EP 1610515 DI 10.3389/pore.2022.1610504 PG 12 ER PT J AU Jiang, L Li, D Wang, Ch Liao, J Liu, J Wei, Q Wang, Y AF Jiang, Lucen Li, Dan Wang, Chao Liao, Jia Liu, Jianghuan Wei, Qingzhu Wang, Yiyang TI Decreased Expression of Karyopherin-α 1 is Related to the Malignant Degree of Cervical Cancer and is Critical for the Proliferation of Hela Cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cervical cancer; proliferation; KPNA1; Hela cell; malignant ID cervical cancer; proliferation; KPNA1; Hela cell; malignant AB Karyopherin α (KPNA) proteins are involved in nucleocytoplasmic trafficking and are critical for protein subcellular localization. Recent studies have suggested that KPNA proteins are abnormally expressed in various solid tumors. The objective of this study was to investigate the expression of KPNA1 and KPNA2 in cervical cancer tissue with different histologic grades and cell lines, as well as the effects of the KPNA1 expression level on Hela cell proliferation. We collected the medical data of 106 patients with cervical cancer and investigated the protein expression of KPNA1 and KPNA2 by immunohistochemistry and western blot. The results revealed a significantly lower expression of KPNA1 in cervical cancer compared to normal tissue. Conversely, stronger staining intensity for KPNA2 was observed in cervical tumor samples. The expression levels of KPNA1 and KPNA2 were significantly associated with the tumor histologic grade. The weakest KPNA1 expression and strongest staining for KPNA2 were observed in grade III tumor tissue. The expression levels of KPNA1 were lower in Hela and C33A cells compared with normal human cervical epithelial cells; however, the expression of KPNA2 exhibited an opposite trend. The upregulation of KPNA1 significantly suppressed the proliferation of Hela cells and relevant proteins expression, as well as promoted transportation of IRF3 into nucleus. Our results suggest the downregulation of KPNA1 expression is related to the malignant degree of cervical cancer and is closely associated with the proliferation of cervical cancer cells. C1 [Jiang, Lucen] Southern Medical University, College of Basic Medicine, Department of PathologyGuangzhou, China. [Li, Dan] Southern Medical University, College of Basic Medicine, Department of PathologyGuangzhou, China. [Wang, Chao] The Sixth Affiliated Hospital of Sun Yat-Sen University, Department of PathologyGuangzhou, China. [Liao, Jia] Jinan University, School of Medicine, Department of PathophysiologyGuangzhou, China. [Liu, Jianghuan] Southern Medical University, College of Basic Medicine, Department of PathologyGuangzhou, China. [Wei, Qingzhu] Southern Medical University, College of Basic Medicine, Department of PathologyGuangzhou, China. [Wang, Yiyang] Jinan University, School of Medicine, Department of PathophysiologyGuangzhou, China. RP Wang, Y (reprint author), Jinan University, School of Medicine, Department of Pathophysiology, Guangzhou, China. EM wangyiyang@jnu.edu.cn CR Brisson M, Drolet M. Global Elimination of Cervical Cancer as a Public Health Problem. Lancet Oncol, 2019, 20(3):319–21., DOI 10.1016/S1470-2045(19, 30072-5 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Si MY, Su XY, Jiang Y, Qiao YL, Liu YL. Interventions to Improve Human Papillomavirus Vaccination Among Chinese Female College Students: Study Protocol for a Randomized Controlled Trial. BMC Public Health, 2019, 19(1): 1546., DOI 10.1186/s12889-019-7903-x Ramirez PT, Pareja R, Rendon G, Millan C, Frumovitz M, Schmeler KM, et al. Management of Low-Risk Early-Stage Cervical Cancer: Should Conization, Simple Trachelectomy, or Simple Hysterectomy Replace Radical Surgery as the New Standard of Care? Gynecol Oncol, 2014, 132(1):254–9., DOI 10.1016/j. ygyno.2013.09.004 Rose GP. Concurrent Chemoradiation for Locally Advanced Carcinoma of the Cervix: Where are we in 2006? Ann Oncol, 2006, 17(10):x224–9., DOI 10.1093/ annonc/mdl264 Bosh FX, Lorincz A, Munoz N, Meijer C, Shah KV. The Causal Relation between Human Papillomavirus and Cervical Cancer. J Clin Pathol, 2002, 55(4):244–65., DOI 10.1136/jcp.55.4.244 Schiffman M, Wentzensen N, Wacholder S, Kinney W, Gage JC, Castle PE, et al. Human Papillomavirus Testing in the Prevention of Cervical Cancer. J Natl Cancer Inst, 2011, 103(5):368–83., DOI 10.1093/jnci/ djq562 Pimple SA, Mishra GA. Global Strategies for Cervical Cancer Prevention and Screening. Minerva Ginecol, 2019, 71(4):313–20., DOI 10.23736/S0026-4784.19. 04397-1 Lange A, Mills RE, Lange CJ, Stewart M, Devine SE, Corbett AH, et al. Classical Nuclear Localization Signals: Definition, Function, and Interaction with Importin α. J Biol Chem, 2011, 282(8):5101–5., DOI 10.1074/jbc.R600026200 Moroianu J, Hijikata M, Blobel G, Radu A. Mammalian Karyopherin Alpha 1 Beta and Alpha 2 Beta Heterodimers: Alpha 1 or Alpha 2 Subunit Binds Nuclear Localization Signal and Beta Subunit Interacts with Peptide Repeat- Containing Nucleoporins. Proc Natl Acad Sci U S A, 1995, 92(14):6532–6., DOI 10.1073/pnas.92.14.6532 Mosammaparast N, Pemberton LF. Karyopherins: From Nuclear-Transport Mediators to Nuclear-Function Regulators. Trends Cel Biol, 2004, 14(10): 547–56., DOI 10.1016/j.tcb.2004.09.004 Alshareeda AT, Negm OH, Green AR, Nolan CC, Tighe P, Albarakati N, et al. KPNA2 is a Nuclear export Protein that Contributes to Aberrant Localisation of Key Proteins and Poor Prognosis of Breast Cancer. Br J Cancer, 2015, 112(12):1929–37., DOI 10.1038/bjc.2015.165 Christiansen A, Dyrskjot L. The Functional Role of the Novel Biomarker Karyopherin α 2, KPNA2, in Cancer. Cancer Lett, 2013, 331(1):18–23., DOI 10. 1016/j.canlet.2012.12.013 Sakai M, Sohda M, Miyazaki T, Suzuki S, Sano A, Tanaka N, et al. Significance of Karyopherin-{alpha} 2, KPNA2, Expression in Esophageal Squamous Cell Carcinoma. Anticancer Res, 2010, 30(3):851–6. Zhang Y, Zhang M, Yu F, Lu S, Sun H, Tang H, et al. Karyopherin Alpha 2 is a Novel Prognostic Marker and a Potential Therapeutic Target for colon Cancer. J Exp Clin Cancer Res, 2015, 34:145., DOI 10.1186/s13046-015-0261-3 PaulineVan Der Watt J, Christopher PM, Denver TH, Iqbal PM, Lynette D, Dhirendra G, et al. The Karyopherin Proteins, Crm1 and Karyopherin Beta1, are Overexpressed in Cervical Cancer and are Critical for Cancer Cell Survival and Proliferation. Int J Cancer, 2009, 124(8):1829–40., DOI 10.1002/ijc.24146 Jiang L, Liu J, Wei Q, Wang Y. KPNA2 Expression is a Potential Marker for Differential Diagnosis between Osteosarcomas and Other Malignant Bone Tumor Mimics. Diagn Pathol, 2020, 15(1):135., DOI 10.1186/s13000-020- 01051-6 Bolag A, Takehiko Y, Erito M, Tetsuro O, Kyoichi O, Atsushi O, et al. Nuclear Karyopherin-α2 Expression in Primary Lesions and Metastatic Lymph Nodes Was Associated with Poor Prognosis and Progression in Gastric Cancer. Carcinogenesis, 2013, 34(10):2314–21., DOI 10.1093/carcin/bgt214 Huang L, Zhou Y, Cao XP, Lin JX, Zhang L, Huang ST, et al. KPNA2 is a Potential Diagnostic Serum Biomarker for Epithelial Ovarian Cancer and Correlates with Poor Prognosis. Tumour Biol, 2017, 39(6):1010428317706289., DOI 10.1177/10104283177062-89 Jiang P, Tang Y, He L, Tang H, Liang M, Mai C, et al. Aberrant Expression of Nuclear KPNA2 is Correlated with Early Recurrence and Poor Prognosis in Patients with Small Hepatocellular Carcinoma after Hepatectomy. Med Oncol, 2014, 31(8):131., DOI 10.1007/s12032-014-0131-4 Bermudez A, Bhatla N, Leung E. Cancer of the Cervix Uteri. Int J Gynaecol Obstet, 2015, 131(2):S88–95., DOI 10.1016/j.ijgo.2015.06.004 Wang YY, Wang Y, Yang DM, Yu XH, Li H, Lv XX, et al. β-Adrenoceptor Stimulation Promotes LPS-Induced Cardiomyocyte Apoptosis through Activating PKA and Enhancing CaMKII and IκBα Phosphorylation. Crit Care, 2015, 19(1):76., DOI 10.1186/s13054-015-0820-1 Liu S, Cai X, Wu J, Cong Q, Chen X, Li T, et al. Phosphorylation of Innate Immune Adaptor Proteins MAVS, STING, and TRIF Induces IRF3 Activation. Science, 2015, 347(6227):aaa2630., DOI 10.1126/science.aaa2630 Yeon SI, Youn JH, Lim MH, Lee HJ, Kim YM, Choi JE, et al. Development of Monoclonal Antibodies against Human IRF-5 and Their Use in Identifying the Binding of IRF-5 to Nuclear Import Proteins Karyopherin-Alpha1 and -beta1. Yonsei Med J, 2008, 49(6):1023–31., DOI 10.3349/ymj.2008.49.6.1023 Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Pineros M, et al. Estimating the Global Cancer Incidence and Mortality in 2018: GLOBOCAN Sources and Methods. Int J Cancer, 2019, 144(8):1941–53., DOI 10.1002/ijc.31937 Kohler M, Speck C, Christiansen M, Bischoff FR, Prehn S, Haller H, et al. Evidence for Distinct Substrate Specificities of Importin α Family Members in Nuclear Protein Import. Mol Cel Biol, 1999, 19(11):7782–91., DOI 10.1128/ MCB.19.11.7782 Cui X, Wang H, Wu X, Huo K, Jing X. Increased Expression of KPNA2 Predicts Unfavorable Prognosis in Ovarian Cancer Patients, Possibly by Targeting KIF4A Signaling. J Ovarian Res, 2021, 14(1):71., DOI 10.1186/s13048-021-00818-9 Zeng F, Luo L, Li D, Guo J, Guo M. KPNA2 Interaction with CBX8 Contributes to the Development and Progression of Bladder Cancer by Mediating the PRDM1/c-FOS Pathway. J Transl Med, 2021, 19(1):112., DOI 10.1186/s12967- 021-02709-5 Kubo N, Araki K, Altan B, Hoshino K, Ishii N, Tsukagoshi M, et al. Enhanced Karyopherin -α2 Expression is Associated with Carcinogenesis in Patients with Intraductal Papillary Mucinous Neoplasms. Pancreatology, 2017, 17:611–6., DOI 10.1016/j.pan.2017.04.011 Poon I, Jans DA. Regulation of Nuclear Transport: Central Role in Development and Transformation? Traffic, 2005, 6(3):173–86., DOI 10.1111/ j.1600-0854.2005.00268.x Kau TR, Way JC, Silver PA. Nuclear Transport and Cancer: From Mechanism to Intervention. Nat Rev Cancer, 2004, 4(2):106–17., DOI 10.1038/nrc1274 Quensel C, Friedrich B, Sommer T, Hartmann E, Kohler M. In Vivo analysis of Importin Alpha Proteins Reveals Cellular Proliferation Inhibition and Substrate Specificity. Mol Cel Biol, 2005, 24(23):10246–55., DOI 10.1128/ MCB.24.23.10246-10255.2004 Noetzel E, Rose M, Bornemann J, Gajewski M, Knuchel R, Dahl E, et al. Nuclear Transport Receptor Karyopherin-α2 Promotes Malignant Breast Cancer Phenotypes In Vitro. Oncogene, 2012, 31(16):2101–14., DOI 10.1038/ onc.2011.403 Wang YY, Liu RC, Liao J, Jiang LC, Jeong GH, Zhou L, et al. Orthogonal Ubiquitin Transfer Reveals Human Papillomavirus E6 Downregulates Nuclear Transport to Disarm Interferon-γ Dependent Apoptosis of Cervical Cancer Cells. FASEB J, 2021, 35(11):e21986., DOI 10.1096/fj.202101232RR Kosyna FK, Depping R. Controlling the Gatekeeper: Therapeutic Targeting of Nuclear Transport. Cells, 2018, 7(11):221., DOI 10.3390/cells7110221 Angus L, van derWatt PJ, Leaner VD. Inhibition of the Nuclear Transporter, Kpnβ1, Results in Prolonged Mitotic Arrest and Activation of the Intrinsic Apoptotic Pathway in Cervical Cancer Cells. Carcinogenesis, 2014, 35(5): 1121–31., DOI 10.1093/carcin/bgt491 Kosyna FK, Nagel M, Kluxen L, Kraushaar K, Depping R. The Importin α/ β-specific Inhibitor Ivermectin Affects HIF-dependent Hypoxia Response Pathways. Biol Chem, 2015, 396(12):1357–67., DOI 10.1515/ hsz-2015-0171 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610518 EP 1610527 DI 10.3389/pore.2022.1610518 PG 10 ER PT J AU Kiraly, Zs Szepesi, Sebestyen, A Kuroli, E Rencz, F Toth, B Bokor, L Szakonyi, J Medvecz, M Hidvegi, B AF Kiraly, Zsofia Szepesi, Agota Sebestyen, Anna Kuroli, Eniko Rencz, Fanni Toth, Bela Bokor, Laura Szakonyi, Jozsef Medvecz, Marta Hidvegi, Bernadett TI Immunohistochemical Study of the PD-1/PD-L1 Pathway in Cutaneous Lupus Erythematosus SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PD-L1; PD-1; subacute cutaneous lupus erythematosus; discoid lupus erythematosus; PD-1 inhibitorinduced SCLE; TEN-like lupus ID PD-L1; PD-1; subacute cutaneous lupus erythematosus; discoid lupus erythematosus; PD-1 inhibitorinduced SCLE; TEN-like lupus AB The pathomechanism of various autoimmune diseases is known to be associated with the altered function of programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) axis. We aimed to investigate the role of this pathway and inflammatory cell markers in subtypes of cutaneous lupus erythematosus (CLE): discoid lupus erythematosus (DLE), subacute CLE (SCLE) and toxic epidermal necrolysis (TEN)-like lupus, a hyperacute form of acute CLE (ACLE). Ten skin biopsy samples from 9 patients were analyzed with immunohistochemistry regarding the following markers: CD3, CD4, CD8, Granzyme B, CD123, CD163, PD-1, PD-L1. Our group consisted of 4 SCLE (2 idiopathic (I-SCLE) and 2 PD-1 inhibitor-induced (DI-SCLE)), 4 DLE and 1 TEN-like lupus cases. From the latter patient two consecutive biopsies were obtained 1 week apart. Marker expression patterns were compared through descriptive analysis. Higher median keratinocyte (KC) PD-L1 expression was observed in the SCLE group compared to the DLE group (65% and 5%, respectively). Medians of dermal CD4, Granzyme B (GB), PD-1 positive cell numbers and GB+/CD8+ ratio were higher in the DLE group than in the SCLE group. The I-SCLE and DISCLE cases showed many similarities, however KC PD-L1 expression and dermal GB positive cell number was higher in the former. The consecutive samples of the TEN-like lupus patient showed an increase by time within the number of infiltrating GB+ cytotoxic T-cells and KC PD-L1 expression (from 22 to 43 and 30%–70%, respectively). Alterations of the PD-1/PD-L1 axis seems to play a role in the pathogenesis of CLE. C1 [Kiraly, Zsofia] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Szepesi, Agota] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kuroli, Eniko] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Rencz, Fanni] Corvinus University of Budapest, Department of Health EconomicsBudapest, Hungary. [Toth, Bela] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Bokor, Laura] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Szakonyi, Jozsef] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Medvecz, Marta] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Hidvegi, Bernadett] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. RP Kiraly, Zs (reprint author), Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Budapest, Hungary. EM kiraly.zsofia@phd.semmelweis.hu CR Curran CS, Gupta S, Sanz I, Sharon E. PD-1 Immunobiology in Systemic Lupus Erythematosus. J Autoimmun, 2019, 97:1–9., DOI 10.1016/j.jaut.2018. 10.025 Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the Pd-1 Immunoinhibitory Receptor by a Novel B7 Family Member Leads to Negative Regulation of Lymphocyte Activation. J Exp Med, 2000, 192(7):1027–34., DOI 10.1084/jem.192.7.1027 Ramos-Casals M, Brahmer JR, Callahan MK, Flores-Chavez A, Keegan N, Khamashta MA, et al. Immune-related Adverse Events of Checkpoint Inhibitors. Nat Rev Dis Primers, 2020, 6(1):38., DOI 10.1038/s41572-020- 0160-6 Sibaud V. Dermatologic Reactions to Immune Checkpoint Inhibitors: Skin Toxicities and Immunotherapy. Am J Clin Dermatol, 2018, 19(3):345–61., DOI 10.1007/s40257-017-0336-3 Bui AN, Hirner J, Singer S, Eberly-Puleo A, Larocca C, Lian CG, et al. De Novo subacute Cutaneous Lupus Erythematosus like Eruptions in the Setting of PD- 1 or PD-L1 Inhibitor Therapy: Clinical-pathological Correlation. Clin Exp Dermatol, 2020, 46:328., DOI 10.1111/ced.14449 Diago A, Hueso L, Ara-Martin M, Abadias-Granado I. Subacute Cutaneous Lupus Erythematosus Induced by PD-1 Inhibitor Therapy: Two Case Reports and Literature Review. Australas J Dermatol, 2021, 62(2):e347–9., DOI 10.1111/ ajd.13538 Blakeway EA, Elshimy N, Muinonen-Martin A, Marples M, Mathew B, Mitra A. Cutaneous Lupus Associated with Pembrolizumab Therapy for Advanced Melanoma: A Report of Three Cases. Melanoma Res, 2019, 29(3):338–41., DOI 10.1097/cmr.0000000000000587 Biazar C, Sigges J, Patsinakidis N, Ruland V, Amler S, Bonsmann G, et al. Cutaneous Lupus Erythematosus: First Multicenter Database Analysis of 1002 Patients from the European Society of Cutaneous Lupus Erythematosus, EUSCLE). Autoimmun Rev, 2013, 12(3):444–54., DOI 10.1016/j.autrev.2012. 08.019 Kuhn A, Landmann A. The Classification and Diagnosis of Cutaneous Lupus Erythematosus. J Autoimmun, 2014, 48-49:14–9., DOI 10.1016/j.jaut.2014. 01.021 Guicciardi F, Atzori L, Marzano AV, Tavecchio S, Girolomoni G, Colato C, et al. Are There Distinct Clinical and Pathological Features Distinguishing Idiopathic from Drug-Induced Subacute Cutaneous Lupus Erythematosus? A European Retrospective Multicenter Study. J Am Acad Dermatol, 2019, 81(2): 403–11., DOI 10.1016/j.jaad.2019.02.009 Borucki R, Werth VP. Cutaneous Lupus Erythematosus Induced by Drugs - Novel Insights. Expert Rev Clin Pharmacol, 2020, 13:35–42., DOI 10.1080/ 17512433.2020.1698290 Kuhn A, Lehmann P, Ruzicka T. Cutaneous Lupus Erythematosus. Berlin: Springer-Verlag, 2004). p. 53–8. Kuhn A, Wenzel J, Bijl M. Lupus Erythematosus Revisited. Semin Immunopathol, 2016, 38(1):97–112., DOI 10.1007/s00281-015-0550-0 Ceccarelli F, Mancuso S, Lucchetti R, Conti F. Systemic Lupus Erythematosus Onset in Patient Receiving Anti-PD1 Treatment with Pembrolizumab: A Case Report. Rheumatology, United Kingdom,, 2021, 60(2):E39–40., DOI 10.1093/ rheumatology/keaa389 Leonardi GC, Gainor JF, Altan M, Kravets S, Dahlberg SE, Gedmintas L, et al. Safety of Programmed Death-1 Pathway Inhibitors Among Patients with Nonsmall- cell Lung Cancer and Preexisting Autoimmune Disorders. J Clin Oncol, 2018, 36(19):1905–12., DOI 10.1200/jco.2017.77.0305 Liu RC, Sebaratnam DF, Jackett L, Kao S, Lowe PM. Subacute Cutaneous Lupus Erythematosus Induced by Nivolumab. Australas J Dermatol, 2018, 59(2):e152–e154., DOI 10.1111/ajd.12681 Bolton C, Chen Y, Hawthorne R, Schepel IRM, Harriss E, Hofmann SC, et al. Systematic Review: Monoclonal Antibody-Induced Subacute Cutaneous Lupus Erythematosus. Drugs RD, 2020, 20:1–12., DOI 10.1007/s40268-020- 00320-5 Han X, Vesely MD, Yang W, Sanmamed MF, Badri T, Alawa J, et al. PD-1H, VISTA)-mediated Suppression of Autoimmunity in Systemic and Cutaneous Lupus Erythematosus. Sci Transl Med, 2019, 11(522):1–15., DOI 10.1126/ scitranslmed.aax1159 Magro CM, Segal JP, Crowson AN, Chadwick P. The Phenotypic Profile of Dermatomyositis and Lupus Erythematosus: A Comparative Analysis. J Cutan Pathol, 2010, 37(6):659–71., DOI 10.1111/j.1600-0560.2009.01443.x Wenzel J, Zahn S, Mikus S, Wiechert A, Bieber T, Tuting T. The Expression Pattern of Interferon-Inducible Proteins Reflects the Characteristic Histological Distribution of Infiltrating Immune Cells in Different Cutaneous Lupus Erythematosus Subsets. Br J Dermatol, 2007, 157(4): 752–7., DOI 10.1111/j.1365-2133.2007.08137.x Schaberg KB, Novoa RA, Wakelee HA, Kim J, Cheung C, Srinivas S, et al. Immunohistochemical Analysis of Lichenoid Reactions in Patients Treated with Anti-PD-L1 and Anti-PD-1 Therapy. J Cutan Pathol, 2016, 43(4): 339–46., DOI 10.1111/cup.12666 Vivar KL, Deschaine M, Messina J, Divine JM, Rabionet A, Patel N, et al. Epidermal Programmed Cell Death-ligand 1 Expression in TEN Associated with Nivolumab Therapy. J Cutan Pathol, 2017, 44(4):381–4., DOI 10.1111/cup. 12876 Okiyama N, Katz SI. Programmed Cell Death 1, PD-1, Regulates the Effector Function of CD8 T Cells via PD-L1 Expressed on Target Keratinocytes. J Autoimmun, 2014, 53(C):1–9., DOI 10.1016/j.jaut.2014.06.005 Youngnak-Piboonratanakit P, Tsushima F, Otsuki N, Igarashi H, Machida U, Iwai H, et al. The Expression of B7-H1 on Keratinocytes in Chronic Inflammatory Mucocutaneous Disease and its Regulatory Role. Immunol Lett, 2004, 94(3):215–22., DOI 10.1016/j.imlet.2004.05.007 Ritprajak P, Hashiguchi M, Tsushima F, Chalermsarp N, Azuma M. Keratinocyte-Associated B7-H1 Directly Regulates Cutaneous Effector CD8+ T Cell Responses. J Immunol, 2010, 184(9):4918–25., DOI 10.4049/ jimmunol.0902478 Costa NL, Goncalves JAM, Lima SLG, Arruda JAA, Miranda ACC, Mesquita RA, et al. Evaluation of PD-L1, PD-L2, PD-1 and Cytotoxic Immune Response in Oral Lichen Planus. Oral Dis, 2020, 26(6):1246–54., DOI 10.1111/odi.13344 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610521 EP 1610527 DI 10.3389/pore.2022.1610521 PG 7 ER PT J AU Zhang, Y Li, L Chu, F Xiao, X Zhang, L Li, K Wu, H AF Zhang, Yong Li, Lu Chu, Feifei Xiao, Xingguo Zhang, Li Li, Kunkun Wu, Huili TI Identification and Validation of an m6A-Related LncRNA Signature to Predict Progression-Free Survival in Colorectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal cancer; lncRNA; signature; m6A; progression free survival ID colorectal cancer; lncRNA; signature; m6A; progression free survival AB The RNA methylation of N6 adenosine (m6A) plays a crucial role in various biological processes. Strong evidence reveals that the dysregulation of long non-coding RNAs (lncRNA) brings about the abnormality of downstream signaling in multiple ways, thus influencing tumor initiation and progression. Currently, it is essential to discover effective and succinct molecular biomarkers for predicting colorectal cancer (CRC) prognosis. However, the prognostic value of m6A-related lncRNAs for CRC remains unclear, especially for progression-free survival (PFS). Here, we screened 24 m6A-related lncRNAs in 622 CRC patients and identified five lncRNAs (SLCO4A1-AS1, MELTFAS1, SH3PXD2A-AS1, H19 and PCAT6) associated with patient PFS. Compared to normal samples, their expression was up-regulated in CRC tumors from TCGA dataset, which was validated in 55 CRC patients fromour in-house cohort. We established an m6ALnc signature for predicting patient PFS, which was an independent prognostic factor by classification analysis of clinicopathologic features. Moreover, the signature was validated in 1,077 patients from six independent datasets (GSE17538, GSE39582, GSE33113, GSE31595, GSE29621, and GSE17536), and it showed better performance than three known lncRNA signatures for predicting PFS. In summary, our study demonstrates that the m6A-Lnc signature is a promising biomarker for forecasting patient PFS in CRC. C1 [Zhang, Yong] Zhengzhou Central Hospital Affiliated to Zhengzhou University, Department of GastroenterologyZhengzhou, China. [Li, Lu] Zhengzhou Central Hospital Affiliated to Zhengzhou University, Department of GastroenterologyZhengzhou, China. [Chu, Feifei] Zhengzhou Central Hospital Affiliated to Zhengzhou University, Department of GastroenterologyZhengzhou, China. [Xiao, Xingguo] Zhengzhou Central Hospital Affiliated to Zhengzhou University, Department of GastroenterologyZhengzhou, China. [Zhang, Li] Zhengzhou Central Hospital Affiliated to Zhengzhou University, Department of GastroenterologyZhengzhou, China. [Li, Kunkun] Zhengzhou Central Hospital Affiliated to Zhengzhou University, Department of GastroenterologyZhengzhou, China. [Wu, Huili] Zhengzhou Central Hospital Affiliated to Zhengzhou University, Department of GastroenterologyZhengzhou, China. RP Wu, H (reprint author), Zhengzhou Central Hospital Affiliated to Zhengzhou University, Department of Gastroenterology, Zhengzhou, China. EM wuhuili660912@zzu.edu.cn CR Fitzmaurice C, Allen C, Barber RM, Barregard L, Bhutta ZA, Brenner H, et al. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived with Disability, and Disability-Adjusted Life-Years for 32 Cancer Groups, 1990 to 2015: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol, 2017, 3(4):524–48., DOI 10.1001/jamaoncol.2016.5688 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Marisa L, de Reynies A, Duval A, Selves J, Gaub MP, Vescovo L, et al. Gene Expression Classification of colon Cancer into Molecular Subtypes: Characterization, Validation, and Prognostic Value. Plos Med, 2013, 10(5): e1001453., DOI 10.1371/journal.pmed.1001453 Kandimalla R, Ozawa T, Gao F, Wang X, Goel A, Nozawa H, et al. Gene Expression Signature in Surgical Tissues and Endoscopic Biopsies Identifies High-Risk T1 Colorectal Cancers. Gastroenterology, 2019, 156(8):2338–41., DOI 10.1053/j.gastro.2019.02.027 Quinn JJ, Chang HY. Unique Features of Long Non-coding RNA Biogenesis and Function. Nat Rev Genet, 2016, 17(1):47–62., DOI 10.1038/nrg.2015.10 Han D,Wang M, Ma N, Xu Y, Jiang Y, Gao X. Long Noncoding RNAs: Novel Players in Colorectal Cancer. Cancer Lett, 2015, 361(1):13–21., DOI 10.1016/j. canlet.2015.03.002 Huarte M. The Emerging Role of lncRNAs in Cancer. Nat Med, 2015, 21(11): 1253–61., DOI 10.1038/nm.3981 Yang W, Ning N, Jin X. The lncRNA H19 Promotes Cell Proliferation by Competitively Binding to miR-200a and Derepressing β-Catenin Expression in Colorectal Cancer. Biomed Res Int, 2017, 2017:2767484., DOI 10.1155/2017/2767484 Bian Z, Zhang J, Li M, Feng Y, Wang X, Zhang J, et al. LncRNA-FEZF1-AS1 Promotes Tumor Proliferation and Metastasis in Colorectal Cancer by Regulating PKM2 Signaling. Clin Cancer Res, 2018, 24(19):4808–19., DOI 10. 1158/1078-0432.ccr-17-2967 Zhao J, Xu J, Shang A-q., Zhang R. A Six-LncRNA Expression Signature Associated with Prognosis of Colorectal Cancer Patients. Cell Physiol Biochem, 2018, 50(5):1882–90., DOI 10.1159/000494868 Huang R, Zhou L, Chi Y, Wu H, Shi L. LncRNA Profile Study Reveals a SevenlncRNA Signature Predicts the Prognosis of Patients with Colorectal Cancer. Biomark Res, 2020, 8:8., DOI 10.1186/s40364-020-00187-3 Gu L, Yu J, Wang Q, Xu B, Ji L, Yu L, et al. Identification of a 5-lncRNA Signature-Based Risk Scoring System for Survival Prediction in Colorectal Cancer. Mol Med Rep, 2018, 18(1):279–91., DOI 10.3892/mmr.2018.8963 Wang S, Chai P, Jia R, Jia R. Novel Insights on m6A RNA Methylation in Tumorigenesis: A Double-Edged Sword. Mol Cancer, 2018, 17(1):101., DOI 10. 1186/s12943-018-0847-4 Yang Y, Hsu PJ, Chen Y-S, Yang Y-G. Dynamic Transcriptomic m6A Decoration: Writers, Erasers, Readers and Functions in RNA Metabolism. Cell Res, 2018, 28(6):616–24., DOI 10.1038/s41422-018-0040-8 Roundtree IA, Evans ME, Pan T, He C. Dynamic RNA Modifications in Gene Expression Regulation. Cell, 2017, 169(7):1187–200., DOI 10.1016/j.cell.2017. 05.045 Fu Y, Dominissini D, Rechavi G, He C. Gene Expression Regulation Mediated through Reversible m6A RNA Methylation. Nat Rev Genet, 2014, 15(5): 293–306., DOI 10.1038/nrg3724 Wang S, Zou X, Chen Y, Cho WC, Zhou X. Effect of N6-Methyladenosine Regulators on Progression and Prognosis of Triple-Negative Breast Cancer. Front Genet, 2020, 11:580036., DOI 10.3389/fgene.2020.580036 Ji L, Chen S, Gu L, Zhang X. Exploration of Potential Roles of m6A Regulators in Colorectal Cancer Prognosis. FrontOncol, 2020, 10:768., DOI 10.3389/fonc.2020.00768 Zhao X, Cui L. Development and Validation of a m6A RNA Methylation Regulators-Based Signature for Predicting the Prognosis of Head and Neck Squamous Cell Carcinoma. Am J Cancer Res, 2019, 9(10):2156–69. Dai F, Wu Y, Lu Y, An C, Zheng X, Dai L, et al. Crosstalk between RNA m6 A ModificationandNon-codingRNAContributes toCancerGrowth andProgression. Mol Ther Nucleic Acids, 2020, 22:62–71., DOI 10.1016/j.omtn.2020.08.004 Chang Y-Z, Chai R-C, Pang B, Chang X, An SY, Zhang K-N, et al. METTL3 Enhances the Stability ofMALAT1 with theAssistance of HuR viam6AModification and Activates NF-κB to Promote the Malignant Progression of IDH-Wildtype Glioma. Cancer Lett, 2021, 511:36–46., DOI 10.1016/j.canlet.2021.04.020 Yu J, Mao W, Sun S, Hu Q, Wang C, Xu Z, et al. Identification of an m6ARelated lncRNA Signature for Predicting the Prognosis in Patients with Kidney Renal Clear Cell Carcinoma. Front Oncol, 2021, 11:663263., DOI 10.3389/fonc. 2021.663263 Tu Z, Wu L, Wang P, Hu Q, Tao C, Li K, et al. N6-Methylandenosine-Related lncRNAs Are Potential Biomarkers for Predicting the Overall Survival of Lower-Grade Glioma Patients. Front Cel Dev. Biol., 2020, 8:642., DOI 10.3389/ fcell.2020.00642 Wang H, Meng Q, Ma B. Characterization of the Prognostic m6A-Related lncRNA Signature in Gastric Cancer. Front Oncol, 2021, 11:630260., DOI 10. 3389/fonc.2021.630260 Wang K, Zhao B, Liang Y, Ma B. Identification and Validation of a Novel 2- LncRNAs Signature Associated with m6A Regulation in Colorectal Cancer. J Cancer, 2022, 13(1):21–33., DOI 10.7150/jca.64817 Yu ZL, Zhu ZM. Construction of an N6-Methyladenosine lncRNA- and Immune Cell Infiltration-Related Prognostic Model in Colorectal Cancer. Protoplasma, 2021, 259:1029., DOI 10.1007/s00709-021-01718-x Song W, Ren J, Yuan W, Xiang R, Ge Y, Fu T. N6-Methyladenosine-Related lncRNA Signature Predicts the Overall Survival of Colorectal Cancer Patients. Genes, Basel,, 2021, 12(9):1375., DOI 10.3390/genes12091375 Li Y, Xiao J, Bai J, Tian Y, Qu Y, Chen X, et al. Molecular Characterization and Clinical Relevance of m6A Regulators across 33 Cancer Types. Mol Cancer, 2019, 18(1):137., DOI 10.1186/s12943-019-1066-3 Love MI, Huber W, Anders S. Moderated Estimation of Fold Change and Dispersion for RNA-Seq Data with DESeq2. Genome Biol, 2014, 15(12):550., DOI 10.1186/s13059-014-0550-8 Deng S, Zhang H, Zhu K, Li X, Ye Y, Li R, et al. M6A2Target: A Comprehensive Database for Targets of m6A Writers, Erasers and Readers. Brief Bioinform, 2021, 22(3):bbaa055., DOI 10.1093/bib/bbaa055 Teng X, Chen X, Xue H, Tang Y, Zhang P, Kang Q, et al. NPInter v4.0: An Integrated Database of ncRNA Interactions. Nucleic Acids Res, 2020, 48(D1): D160–D165., DOI 10.1093/nar/gkz969 Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: Decoding miRNAceRNA, miRNA-ncRNA and Protein-RNA Interaction Networks from Large- Scale CLIP-Seq Data. Nucleic Acids Res, 2014, 42:D92–7., DOI 10.1093/nar/ gkt1248 Karagkouni D, Paraskevopoulou MD, Tastsoglou S, Skoufos G, Karavangeli A, Pierros V, et al. DIANA-LncBase V3: Indexing Experimentally Supported miRNA Targets on Non-coding Transcripts. Nucleic Acids Res, 2020, 48(D1): D101–D110., DOI 10.1093/nar/gkz1036 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: A Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci U.S.A, 2005, 102(43):15545–50., DOI 10.1073/pnas.0506580102 Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular Signatures Database, MSigDB, 3.0. Bioinformatics, 2011, 27(12):1739–40., DOI 10.1093/bioinformatics/btr260 Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, et al. clusterProfiler 4.0: A Universal Enrichment Tool for Interpreting Omics Data. The Innovation, 2021, 2(3): 100141., DOI 10.1016/j.xinn.2021.100141 Wang T, Kong S, TaoM, Ju S. The Potential Role of RNA N6-Methyladenosine in Cancer Progression. Mol Cancer, 2020, 19(1):88., DOI 10.1186/s12943-020- 01204-7 Jiang X, Liu B, Nie Z, Duan L, Xiong Q, Jin Z, et al. The Role of m6A Modification in the Biological Functions and Diseases. Sig Transduct Target Ther, 2021, 6(1):74., DOI 10.1038/s41392-020-00450-x Ma S, Chen C, Ji X, Liu J, Zhou Q, Wang G, et al. The Interplay between m6A RNA Methylation and Noncoding RNA in Cancer. J Hematol Oncol, 2019, 12(1):121., DOI 10.1186/s13045-019-0805-7 He L, Li H, Wu A, Peng Y, Shu G, Yin G. Functions of N6-Methyladenosine and its Role in Cancer. Mol Cancer, 2019, 18(1):176., DOI 10.1186/s12943-019- 1109-9 Alarcon CR, Lee H, Goodarzi H, Halberg N, Tavazoie SF. N6-methyladenosine marks Primary microRNAs for Processing. Nature, 2015, 519(7544):482–5., DOI 10.1038/nature14281 Chen Y, Lin Y, Shu Y, He J, Gao W. Interaction between N6-Methyladenosine, m6A, Modification and Noncoding RNAs in Cancer. Mol Cancer, 2020, 19(1):94., DOI 10.1186/s12943-020-01207-4 Monfort A, Di Minin G, Postlmayr A, Freimann R, Arieti F, Thore S, et al. Identification of Spen as a Crucial Factor for Xist Function through Forward Genetic Screening in Haploid Embryonic Stem Cells. Cel Rep, 2015, 12(4): 554–61., DOI 10.1016/j.celrep.2015.06.067 Patil DP, Chen C-K, Pickering BF, Chow A, Jackson C, Guttman M, et al. m6A RNA Methylation Promotes XIST-Mediated Transcriptional Repression. Nature, 2016, 537(7620):369–73., DOI 10.1038/nature19342 Wu Y, Yang X, Chen Z, Tian L, Jiang G, Chen F, et al. m6A-induced lncRNA RP11 Triggers the Dissemination of Colorectal Cancer Cells via Upregulation of Zeb1. Mol Cancer, 2019, 18(1):87., DOI 10.1186/s12943-019-1014-2 Guo T, Liu DF, Peng SH, Xu AM. ALKBH5 Promotes colon Cancer Progression by Decreasing Methylation of the lncRNA NEAT1. Am J Transl Res, 2020, 12(8):4542–9. Yan J, Huang X, Zhang X, Chen Z, Ye C, Xiang W, et al. LncRNA LINC00470 Promotes the Degradation of PTEN mRNA to Facilitate Malignant Behavior in Gastric Cancer Cells. Biochem Biophys Res Commun, 2020, 521(4):887–93., DOI 10.1016/j.bbrc.2019.11.016 Wang Y, Lu J-H,Wu Q-N, Jin Y, Wang D-S, Chen Y-X, et al. LncRNA LINRIS Stabilizes IGF2BP2 and Promotes the Aerobic Glycolysis in Colorectal Cancer. Mol Cancer, 2019, 18(1):174., DOI 10.1186/s12943-019-1105-0 Lu S, Yu Z, Xiao Z, Zhang Y. Gene Signatures and Prognostic Values of m6A Genes in Nasopharyngeal Carcinoma. Front Oncol, 2020, 10:875., DOI 10.3389/ fonc.2020.00875 Zhao Y, Tao Z, Chen X. Identification of a Three-m6A Related Gene Risk Score Model as a Potential Prognostic Biomarker in clear Cell Renal Cell Carcinoma. PeerJ, 2020, 8:e8827., DOI 10.7717/peerj.8827 Chu F, Cui Y, Li K, Xiao X, Zhang L, Zhang L, et al. Long Noncoding RNA THOR is Highly Expressed in Colorectal Cancer and Predicts a Poor Prognosis. Future Oncol, 2020, 16(25):1911–20., DOI 10.2217/fon-2020-0393 Weng C, Wang L, Liu G, Guan M, Lu L. Identification of a N6- Methyladenosine, m6A)-Related lncRNA Signature for Predicting the Prognosis and Immune Landscape of Lung Squamous Cell Carcinoma. Front Oncol, 2021, 11:763027., DOI 10.3389/fonc.2021.763027 Li L, Xie R, Lu G. Identification of m6A Methyltransferase-Related lncRNA Signature for Predicting Immunotherapy and Prognosis in Patients with Hepatocellular Carcinoma. Biosci Rep, 2021, 41(6):BSR20210760., DOI 10. 1042/BSR20210760 Zhou C,Wang S, Shen Z, Shen Y, Li Q, Shen Y, et al. Construction of an m6ARelated lncRNA Pair Prognostic Signature and Prediction of the Immune Landscape in Head and Neck Squamous Cell Carcinoma. J Clin Lab Anal, 2022, 36(1):e24113., DOI 10.1002/jcla.24113 Chen S, Li X, Guo L, Zhang J, Li L, Wang X, et al. Characterization of the m6A-Related lncRNA Signature in Predicting Prognosis and Immune Response in Patients with colon Cancer. J BUON, 2021, 26(5):1931–41. Liu C, Yang Z, Li R, Wu Y, Chi M, Gao S, et al. Potential Roles of N6- Methyladenosine, m6A, in Immune Cells. J Transl Med, 2021, 19(1):251., DOI 10.1186/s12967-021-02918-y Shulman Z, Stern-Ginossar N. The RNA Modification N6-Methyladenosine as a Novel Regulator of the Immune System. Nat Immunol, 2020, 21(5):501–12., DOI 10.1038/s41590-020-0650-4 Han D, Liu J, Chen C, Dong L, Liu Y, Chang R, et al. Anti-tumour Immunity Controlled through mRNA m6AMethylation and YTHDF1 in Dendritic Cells. Nature, 2019, 566(7743):270–4., DOI 10.1038/s41586-019-0916-x Luo J, Xu T, Sun K. N6-Methyladenosine RNA Modification in Inflammation: Roles, Mechanisms, and Applications. Front Cel Dev. Biol., 2021, 9:670711., DOI 10.3389/fcell.2021.670711 Yu R, Li Q, Feng Z, Cai L, Xu Q. m6A Reader YTHDF2 Regulates LPS-Induced Inflammatory Response. Int J Mol Sci, 2019, 20(6):1323., DOI 10.3390/ijms20061323 Zhang M, Song J, Yuan W, Zhang W, Sun Z. Roles of RNA Methylation on Tumor Immunity and Clinical Implications. Front Immunol, 2021, 12:641507., DOI 10.3389/fimmu.2021.641507 Li H-B, Tong J, Zhu S, Batista PJ, Duffy EE, Zhao J, et al. m6A mRNA Methylation Controls T Cell Homeostasis by Targeting the IL-7/STAT5/SOCS Pathways. Nature, 2017, 548(7667):338–42., DOI 10.1038/nature23450 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610536 EP 1610548 DI 10.3389/pore.2022.1610536 PG 13 ER PT J AU Kispal, M Janvary, ZsL Balatoni, T Gabor, S Fedorcsak, I Katalin, B Kenessey, I Liszkay, G AF Kispal, Mihaly Janvary, Zsolt Levente Balatoni, Timea Gabor, Stelczer Fedorcsak, Imre Katalin, Bocs Kenessey, Istvan Liszkay, Gabriella TI The Role of Stereotactic Radiotherapy in the Management of Melanoma, A Retrospective Single Institute Preliminary Study of 30 Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE skin cancer; stereotactic radiation therapy; targeted therapy; immunotherapy; melanoma ID skin cancer; stereotactic radiation therapy; targeted therapy; immunotherapy; melanoma AB Cutaneous melanoma is the third most common type of skin cancer in the world. The incidence of melanoma is increasing in most countries, however, mortality seems to be slowly decreasing. The treatment of advanced cutaneous melanoma changed radically since 2011. The new therapeutic modalities, such as immuno- and targeted therapies give a chance to successfully reach more prolonged progression-free survival (PFS) and overall survival (OS) in patients with metastatic melanoma. Despite the great therapeutic benefit, most patients eventually develop resistance to these therapies, and the disease will progress. In some cases oligoprogression develops. In those cases local therapy, such as stereotactic radiotherapy can make it possible to continue the previously applied effective medical treatment for the benefit of patients. In our study of a total of 30 patients—20 of them received pre-treatment with systemic medical therapy—received stereotactic radiotherapy using various systems, in the National Institute of Oncology, Hungary, Budapest. We managed to prolong the systemic therapy for 12.5 months median period with the assistance of CyberKnife technique. Therapy related adverse events were mostly tolerable with only 3% of Grade 3 toxicity. We concluded that stereotactic radiotherapy and stereotactic radiosurgery, are safe, and effective therapeutic modalities for regional tumor control in cases of oligoprogression C1 [Kispal, Mihaly] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Janvary, Zsolt Levente] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Balatoni, Timea] National Institute of Oncology, Department of DermatologyBudapest, Hungary. [Gabor, Stelczer] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Fedorcsak, Imre] Orszagos Pszichiatriai es Neurologiai IntezetBudapest, Hungary. [Katalin, Bocs] National Institute of Oncology, Center of RadiologyBudapest, Hungary. [Kenessey, Istvan] National Institute of Oncology, National Cancer RegistryBudapest, Hungary. [Liszkay, Gabriella] National Institute of Oncology, Department of DermatologyBudapest, Hungary. RP Kispal, M (reprint author), National Institute of Oncology, Department of Dermatology, Budapest, Hungary. EM kispal.mihaly90@gmail.com CR International Agency for Reserch on Cancer. Melanoma of Skin ASR. Glob Cancer Obs, 2020). Available from: http://globocan.iarc.fr/old/bar_sex_site.asp?selection= 16120&title=Melanoma+of+skin&statistic=2&populations=6&window=1&grid= 1&color1=5&color1e=&color2=4&color2e=&submit= Execute. American Cancer Society. American Cancer Society. Cancer Facts & Figures 2021. Atlanta: American Cancer Society, 2021). p. 1–72. Macdonald EJ. Epidemiology of Melanoma. Prog Clin Cancer, 1975, 6:139–49. Goulart CR, Mattei TA, Ramina R. Cerebral Melanoma Metastases: A Critical Review on Diagnostic Methods and Therapeutic Options. ISRN Surg, 2011, 2011:276908., DOI 10.5402/2011/276908 Robert C, Grob JJ, Stroyakovskiy D, Karaszewska B, Hauschild A, Levchenko E, et al. Five-Year Outcomes with Dabrafenib Plus Trametinib in Metastatic Melanoma. N Engl J Med, 2019, 381:626–36., DOI 10.1056/NEJMoa1904059 Comito F, Leslie I, Boos L, Furness A, Pickering L, Turajlic S, et al. Oligoprogression after Checkpoint Inhibition in Metastatic Melanoma Treated with Locoregional Therapy: A Single-center Retrospective Analysis. J Immunother, 2020, 43:250–5., DOI 10.1097/CJI.0000000000000333 Koenig JL, Shi S, Sborov K, Gensheimer MF, Li G, Nagpal S, et al. Adverse Radiation Effect and Disease Control in Patients Undergoing Stereotactic Radiosurgery and Immune Checkpoint Inhibitor Therapy for Brain Metastases. World Neurosurg, 2019, 126:e1399–e1411., DOI 10.1016/j.wneu. 2019.03.110 Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New Response Evaluation Criteria in Solid Tumours: Revised RECIST Guideline, Version 1.1). Eur J Cancer, 2009, 45(2):228–47., DOI 10.1016/j. ejca.2008.10.026 Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S, et al. iRECIST: Guidelines for Response Criteria for Use in Trials Testing Immunotherapeutics. Lancet Oncol, 2017, 18:e143–e152., DOI 10.1016/ S1470-2045(17)30074-8 Cancer Therapy Evaluation Program, CTEP). Common Terminology Criteria for Adverse Events, CTCAE).v.5.0 [5x7], 2017). Available from: https://ctep.cancer.gov/protocolDevelopment/electronic_ applications/ctc.htm#ctc_50. Domingues B, Lopes J, Soares P, Populo H. Melanoma Treatment in Review. Immunotargets Ther, 2018, 7:35–49., DOI 10.2147/ITT.S134842 Sindhu KK, Leiter A, Moshier E, Lin JY, Carroll E, Brooks D, et al. Durable Disease Control with Local Treatment for Oligoprogression of Metastatic Solid Tumors Treated with Immune Checkpoint Blockade. Cancer Treat Res Commun, 2020, 25:100216., DOI 10.1016/j.ctarc.2020.100216 Sobrevilla-Moreno N, Rivera-Sanchez D, Garcia-Ortega DY, Lopez-Macias D, Garcilazo-Reyes A, Alvarez-Avitia MA. Oligoprogression after Immunotherapy and Targeted Therapy in Metastatic Melanoma. Oncol, United States,, 2021, 35:562–6., DOI 10.46883/ONC.2021.3509.0562 Arozarena I, Wellbrock C. Phenotype Plasticity as Enabler of Melanoma Progression and Therapy Resistance. Nat Rev Cancer, 2019, 19:377–91., DOI 10.1038/s41568-019-0154-4 Guida M, Bartolomeo N, De Risi I, Fucci L, Armenio A, Filannino R, et al. The Management of Oligoprogression in the Landscape of New Therapies for Metastatic Melanoma. Cancers, Basel,, 2019, 11:1559., DOI 10.3390/ cancers11101559 Versluis JM, Hendriks AM, Weppler AM, Brown LJ, de Joode K, Suijkerbuijk KPM, et al. The Role of Local Therapy in the Treatment of Solitary Melanoma Progression on Immune Checkpoint Inhibition: A Multicentre Retrospective Analysis. Eur J Cancer, 2021, 151:72–83., DOI 10.1016/j.ejca.2021.04.003 Tsao MN, Rades D, Wirth A, Lo SS, Danielson BL, Gaspar LE, et al. Radiotherapeutic and Surgical Management for Newly Diagnosed Brain Metastasis(es): An American Society for Radiation Oncology Evidence- Based Guideline. Pract Radiat Oncol, 2012, 2:210–25., DOI 10.1016/j.prro. 2011.12.004 Goyal S, Silk AW, Tian S, Mehnert J, Danish S, Ranjan S, et al. Clinical Management of Multiple Melanoma Brain Metastases a Systematic Review. JAMA Oncol, 2015, 1:668–76., DOI 10.1001/jamaoncol.2015.1206 Liu Y, Dong Y, Kong L, Shi F, Zhu H, Yu J. Abscopal Effect of Radiotherapy Combined with Immune Checkpoint Inhibitors. J Hematol Oncol, 2018, 11: 104., DOI 10.1186/s13045-018-0647-8 Xing D, Siva S, Hanna GG. The Abscopal Effect of Stereotactic Radiotherapy and Immunotherapy: Fool’s Gold or El Dorado? Clin Oncol, 2019, 31:432–43., DOI 10.1016/j.clon.2019.04.006 Watanabe T, Firat E, Scholber J, Gaedicke S, Heinrich C, Luo R, et al. Deep Abscopal Response to Radiotherapy and Anti-PD-1 in an Oligometastatic Melanoma Patient with Unfavorable Pretreatment Immune Signature. Cancer Immunol Immunother, 2020, 69:1823–32., DOI 10.1007/s00262-020-02587-8 Guimond DE, Tsai DCJ, Hosni DA, O’Kane DG, Yang DJ, Barry DA. Safety and Tolerability of Metastasis Directed Radiotherapy in the Era of Evolving Systemic, Immune and Targeted Therapies. Adv Radiat Oncol, 2022, 2022: 101022., DOI 10.1016/j.adro.2022.101022 Robert C, Ribas A, Hamid O, Daud A, Wolchok JD, Joshua AM, et al. Threeyear Overall Survival for Patients with Advanced Melanoma Treated with Pembrolizumab in KEYNOTE-001. J Clin Oncol, 2016, 34:9503., DOI 10.1200/ JCO.2016.34.15_suppl.9503 Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Five-year Survival Outcomes for Patients with Advanced Melanoma Treated with Pembrolizumab in KEYNOTE-001. Ann Oncol, 2019, 30:582–8., DOI 10. 1093/annonc/mdz011 Long GV, Flaherty KT, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, et al. Dabrafenib Plus Trametinib versus Dabrafenib Monotherapy in Patients with Metastatic BRAF V600E/ K-Mutant Melanoma: Long-Term Survival and Safety Analysis of a Phase 3 Study. Ann Oncol, 2017, 28:1631–9., DOI 10.1093/ annonc/mdx176 Robert C, Ribas A, Schachter J, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma, KEYNOTE- 006): post-hoc 5-year Results from an Open-Label, Multicentre, Randomised, Controlled, Phase 3 Study. Lancet Oncol, 2019, 20:1239–51., DOI 10.1016/S1470-2045(19)30388-2 Ratnayake G, Reinwald S, Shackleton M, Moore M, Voskoboynik M, Ruben J, et al. Stereotactic Radiation Therapy Combined with Immunotherapy against Metastatic Melanoma: Long-Term Results of a Phase 1 Clinical Trial. Int J Radiat Oncol Biol Phys, 2020, 108:150–6., DOI 10.1016/j.ijrobp.2020.05.022 Feng R, Oermann EK, Shrivastava R, Gold A, Collins BT, Kondziolka D, et al. Stereotactic Radiosurgery for Melanoma Brain Metastases: A Comprehensive Clinical Case Series. World Neurosurg, 2017, 100:297–304., DOI 10.1016/j. wneu.2017.01.014 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610550 EP 1610559 DI 10.3389/pore.2022.1610550 PG 10 ER PT J AU Zhou, L Wu, H Bai, X Min, Sh Zhang, J Li, C AF Zhou, Lixia Wu, Huiqin Bai, Xingli Min, Shuyun Zhang, Jiawen Li, Cunli TI O-Glycosylating Enzyme GALNT2 Predicts Worse Prognosis in Cervical Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE survival; cervical cancer; GALNT2; anti-tumor immune response; immune-related biomarker ID survival; cervical cancer; GALNT2; anti-tumor immune response; immune-related biomarker AB Identification of novel biomarkers is helpful for the diagnosis and treatment of cervical cancer. Mucin glycosylating enzyme GALNT2 modulates mucin O-glycosylation, and has been revealed as a regulator of tumorigenesis in various cancers. However, the expression pattern of GALNT2 in cervical cancer is still unclear. In this study, we demonstrated that the mRNA expression and protein level of GALNT2 were increased in cervical high-grade intraepithelial neoplasia and tumor tissues compared with normal cervix tissues. Kaplan-Meier plotter showed that overexpression of GALNT2 was associated with worse overall survival in TCGA cohort (p < 0.001, HR = 2.65, 95% CI = 1.62–4.34) and poor disease free survival in GSE44001 cohort (p = 0.0218, HR = 2.15, 95% CI = 1.14–4.06). In addition, GSEA analysis showed that various immune-related pathways were closely related to the expression of GALNT2 in cervical cancer. Moreover, coexpression of GALNT2 and IL1A, IL1B, IL11, CXCL1, CXCL2, CXCL5, CXCL6, CXCR1, or CCR3 predicted poor overall survival, and the expression of GALNT2 also affected the prognostic value of CD47, CD274, CD276, CSF1R, TNFSF9, and TNFSF11 in cervical cancer patients. These findings suggest that GALNT2 might be used as a prognostic biomarker in cervical cancer. C1 [Zhou, Lixia] Shanghai Jiao Tong University School of Medicine, Jiading Branch of Shanghai General Hospital, Department of Obstetrics and GynecologyShanghai, China. [Wu, Huiqin] Shanghai Songjiang DistrictMaternal and Child Health Hospital, Department of Obstetrics and GynecologyShanghai, China. [Bai, Xingli] Shanghai Jiao Tong University School of Medicine, Jiading Branch of Shanghai General Hospital, Department of Obstetrics and GynecologyShanghai, China. [Min, Shuyun] Shanghai Jiao Tong University School of Medicine, Jiading Branch of Shanghai General Hospital, Department of Obstetrics and GynecologyShanghai, China. [Zhang, Jiawen] Shanghai Jiao Tong University School of Medicine, Jiading Branch of Shanghai General Hospital, Department of Obstetrics and GynecologyShanghai, China. [Li, Cunli] Shanghai Jiao Tong University School of Medicine, Jiading Branch of Shanghai General Hospital, Department of Obstetrics and GynecologyShanghai, China. RP Li, C (reprint author), Shanghai Jiao Tong University School of Medicine, Jiading Branch of Shanghai General Hospital, Department of Obstetrics and Gynecology, Shanghai, China. EM lcllzr2019@sina.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Guo L, Hua K. Cervical Cancer: Emerging Immune Landscape and Treatment. Onco Targets Ther, 2020, 13:8037–47., DOI 10.2147/OTT. S264312 Sherer MV, Kotha NV, Williamson C, Mayadev J. Advances in Immunotherapy for Cervical Cancer: Recent Developments and Future Directions. Int J Gynecol Cancer, 2022, 32(3):281–7., DOI 10.1136/ijgc-2021- 002492 Thomas D, Rathinavel AK, Radhakrishnan P. Altered Glycosylation in Cancer: A Promising Target for Biomarkers and Therapeutics. Biochim Biophys Acta Rev Cancer, 2021, 1875(1):188464., DOI 10.1016/j.bbcan.2020.188464 Reily C, Stewart TJ, Renfrow MB, Novak J. Glycosylation in Health and Disease. Nat Rev Nephrol, 2019, 15(6):346–66., DOI 10.1038/s41581-019- 0129-4 Santaella-Verdejo A, Gallegos B, Perez-Campos E, Hernandez P, Zenteno E. Use of amaranthus Leucocarpus Lectin to Differentiate Cervical Dysplasia, CIN). Prep Biochem Biotechnol, 2007, 37(3):219–28., DOI 10.1080/ 10826060701386703 Sahasrabudhe NM, van der Horst JC, Spaans V, Kenter G, de Kroon C, Bosse T, et al. MGL Ligand Expression is Correlated to Lower Survival and Distant Metastasis in Cervical Squamous Cell and Adenosquamous Carcinoma. Front Oncol, 2019, 9:29., DOI 10.3389/fonc.2019.00029 Solorzano C, Angel Mayoral M, de los Angeles Carlos M, Berumen J, Guevara J, Raul Chavez F, et al. Overexpression of Glycosylated Proteins in Cervical Cancer Recognized by the Machaerocereus Eruca Agglutinin. Folia Histochem Cytobiol, 2012, 50(3):398–406., DOI 10.5603/19748 Alexander KL, Serrano CA, Chakraborty A, NearingM, Council LN, Riquelme A, et al. Modulation of Glycosyltransferase ST6Gal-I in Gastric Cancer-Derived Organoids Disrupts Homeostatic Epithelial Cell Turnover. J Biol Chem, 2020, 295(41):14153–63., DOI 10.1074/jbc.RA120.014887 Britain CM, Bhalerao N, Silva AD, Chakraborty A, Buchsbaum DJ, Crowley MR, et al. Glycosyltransferase ST6Gal-I Promotes the Epithelial to Mesenchymal Transition in Pancreatic Cancer Cells. J Biol Chem, 2020, 296:100034., DOI 10.1074/jbc.RA120.014126 Kwon OS, Lee H, Kong HJ, Kwon EJ, Park JE, Lee W, et al. Connectivity Map- Based Drug Repositioning of Bortezomib to Reverse the Metastatic Effect of GALNT14 in Lung Cancer. Oncogene, 2020, 39(23):4567–80., DOI 10.1038/ s41388-020-1316-2 Lin WR, Yeh CT. GALNT14: An Emerging Marker Capable of Predicting Therapeutic Outcomes in Multiple Cancers. Int J Mol Sci, 2020, 21(4):1491., DOI 10.3390/ijms21041491 Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce- Rodriguez I, Chakravarthi BVSK, et al. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses. Neoplasia, 2017, 19(8):649–58., DOI 10.1016/j.neo.2017.05.002 Nagy A, Munkacsy G, Gyorffy B. Pancancer Survival Analysis of Cancer Hallmark Genes. Sci Rep, 2021, 11(1):6047., DOI 10.1038/s41598-021-84787-5 Vasaikar SV, Straub P, Wang J, Zhang B. LinkedOmics: Analyzing Multi- Omics Data within and across 32 Cancer Types. Nucleic Acids Res, 2018, 46(D1):D956–D963., DOI 10.1093/nar/gkx1090 Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al. PGC-1alpha-responsive Genes Involved in Oxidative Phosphorylation are Coordinately Downregulated in Human Diabetes. Nat Genet, 2003, 34(3): 267–73., DOI 10.1038/ng1180 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: A Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci U S A, 2005, 102(43):15545–50., DOI 10.1073/pnas.0506580102 Lairson LL, Henrissat B, Davies GJ, Withers SG. Glycosyltransferases: Structures, Functions, and Mechanisms. Annu Rev Biochem, 2008, 77: 521–55., DOI 10.1146/annurev.biochem.76.061005.092322 Schjoldager KT, Clausen H. Site-specific Protein O-Glycosylation Modulates Proprotein Processing - Deciphering Specific Functions of the Large Polypeptide GalNAc-Transferase Gene Family. Biochim Biophys Acta, 2012, 1820(12):2079–94., DOI 10.1016/j.bbagen.2012.09.014 Holleboom AG, Karlsson H, Lin RS, Beres TM, Sierts JA, Herman DS, et al. Heterozygosity for a Loss-Of-Function Mutation in GALNT2 Improves Plasma Triglyceride Clearance in Man. Cell Metab, 2011, 14(6):811–8., DOI 10.1016/j.cmet.2011.11.005 Marucci A, di Mauro L, Menzaghi C, Prudente S, Mangiacotti D, Fini G, et al. GALNT2 Expression Is Reduced in Patients withType 2Diabetes: Possible Role of Hyperglycemia. PLoS One, 2013, 8(7):e70159., DOI 10.1371/journal.pone.0070159 Zhang X, Zhao H, Zhang J, Han D, Zheng Y, Guo X, et al. Gene Environment Interaction of GALNT2 and APOE Gene with Hypertension in the Chinese Han Population. Biomed Mater Eng, 2015, 26(1):S1977–83., DOI 10.3233/BME-151501 Wu YM, Liu CH, Hu RH, Huang MJ, Lee JJ, Chen CH, et al. Mucin Glycosylating Enzyme GALNT2 Regulates the Malignant Character of Hepatocellular Carcinoma by Modifying the EGF Receptor. Cancer Res, 2011, 71(23):7270–9., DOI 10.1158/0008-5472.CAN-11-1161 Lin MC, Huang MJ, Liu CH, Yang TL, Huang MC. GALNT2 Enhances Migration and Invasion of Oral Squamous Cell Carcinoma by Regulating EGFR Glycosylation and Activity. Oral Oncol, 2014, 50(5):478–84., DOI 10. 1016/j.oraloncology.2014.02.003 Ho WL, Chou CH, Jeng YM, Lu MY, Yang YL, Jou ST, et al. GALNT2 Suppresses Malignant Phenotypes through IGF-1 Receptor and Predicts Favorable Prognosis in Neuroblastoma. Oncotarget, 2014, 5(23): 12247–59., DOI 10.18632/oncotarget.2627 Liu SY, Shun CT, Hung KY, Juan HF, Hsu CL, Huang MC, et al. Mucin Glycosylating Enzyme GALNT2 Suppresses Malignancy in Gastric Adenocarcinoma by Reducing MET Phosphorylation. Oncotarget, 2016, 7(10):11251–62., DOI 10.18632/oncotarget.7081 Wahby S, Jarczyk J, Fierek A, Heinkele J, Weis CA, Eckstein M, et al. POFUT1 mRNA Expression as an Independent Prognostic Parameter in Muscle-Invasive Bladder Cancer. Transl Oncol, 2021, 14(1):100900., DOI 10. 1016/j.tranon.2020.100900 Jeong HY, Park SY, Kim HJ, Moon S, Lee S, Lee SH, et al. B3GNT5 is a Novel Marker Correlated with Stem-like Phenotype and Poor Clinical Outcome in Human Gliomas. CNS Neurosci Ther, 2020, 6(11):1147–54., DOI 10.1111/cns. 13439 Lee PC, Chen ST, Kuo TC, Lin TC, Lin MC, Huang J, et al. C1GALT1 is Associated with Poor Survival and Promotes Soluble Ephrin A1-Mediated Cell Migration through Activation of EPHA2 in Gastric Cancer. Oncogene, 2020, 39(13):2724–40., DOI 10.1038/s41388-020-1178-7 Khetarpal SA, Schjoldager KT, Christoffersen C, Raghavan A, Edmondson AC, Reutter HM, et al. Loss of Function of GALNT2 Lowers High-Density Lipoproteins in Humans, Nonhuman Primates, and Rodents. Cel Metab, 2016, 24(2):234–45., DOI 10.1016/j.cmet.2016.07.012 Sun Z, Xue H, Wei Y, Wang C, Yu R, Wang C, et al. Mucin O-Glycosylating Enzyme GALNT2 Facilitates theMalignant Character of Glioma by Activating the EGFR/PI3K/Akt/mTOR axis. Clin Sci, Lond,, 2019, 133(10):1167–84., DOI 10.1042/CS20190145 Marucci A, Cozzolino F, Dimatteo C, Monti M, Pucci P, Trischitta V, et al. Role of GALNT2 in the Modulation of ENPP1 Expression, and Insulin Signaling and Action: GALNT2: a Novel Modulator of Insulin Signaling. Biochim Biophys Acta, 2013, 1833(6):1388–95., DOI 10.1016/j.bbamcr.2013. 02.032 Wang W, Sun R, Zeng L, Chen Y, Zhang N, Cao S, et al. GALNT2 Promotes Cell Proliferation, Migration, and Invasion by Activating the Notch/Hes1- PTEN-PI3K/Akt Signaling Pathway in Lung Adenocarcinoma. Life Sci, 2021, 276:119439., DOI 10.1016/j.lfs.2021.119439 Kong W, Zhao G, Chen H, Wang W, Shang X, Sun Q, et al. Analysis of Therapeutic Targets and Prognostic Biomarkers of CXC Chemokines in Cervical Cancer Microenvironment. Cancer Cel Int, 2021, 21(1):399., DOI 10.1186/s12935-021-02101-9 Chen R, Yang W, Li Y, Cheng X, Nie Y, Liu D, et al. Effect of Immunotherapy on the Immune Microenvironment in Advanced Recurrent Cervical Cancer. Int Immunopharmacol, 2022, 106:108630., DOI 10.1016/j.intimp.2022.108630 Domenici L, Tonacci A, Aretini P, Garibaldi S, Perutelli A, Bottone P, et al. Inflammatory Biomarkers as Promising Predictors of Prognosis in Cervical Cancer Patients. Oncology, 2021, 99(9):571–9., DOI 10.1159/000517320 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610554 EP 1610564 DI 10.3389/pore.2022.1610554 PG 11 ER PT J AU Wu, J He, B Miao, M Han, X Dai, H Dou, H Li, Y Zhang, X Wang, G AF Wu, Jie He, Baojun Miao, Miao Han, Xibin Dai, Hongyan Dou, Heng Li, Yanqiu Zhang, Xiaoqing Wang, Guangchuan TI Enhancing Natural Killer Cell-Mediated Cancer Immunotherapy by the Biological Macromolecule Nocardia rubra Cell-Wall Skeleton SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer; metastasis; natural killer cell; Nocardia rubra cell-wall skeleton; cytotoxicity ID cancer; metastasis; natural killer cell; Nocardia rubra cell-wall skeleton; cytotoxicity AB The biological macromolecule Nocardia rubra cell-wall skeleton (Nr-CWS) has wellestablished immune-stimulating and anti-tumor activities. However, the role of Nr-CWS on natural killer (NK) cells remains unclear. Here, we explore the function and related mechanisms of Nr-CWS on NK cells. Using a tumor-bearing model, we show that Nr-CWS has slightly effect on solid tumor. In addition, using a tumor metastasis model, we show that Nr-CWS suppresses the lung metastasis induced by B16F10 melanoma cells in mice, which indicates that Nr-CWS may up-regulate the function of NK cells. Further investigation demonstrated that Nr-CWS can increase the expression of TRAIL and FasL on spleen NK cells from Nr-CWS treated B16F10 tumor metastasis mice. The spleen index and serum levels of TNF-α, IFN-γ, and IL-2 in B16F10 tumor metastasis mice treated with Nr-CWS were significantly increased. In vitro, the studies using purified or sorted NK cells revealed that Nr-CWS increases the expression of CD69, TRAIL, and FasL, decreases the expression of CD27, and enhances NK cell cytotoxicity. The intracellular expression of IFN-γ, TNF-α, perforin (prf), granzyme-B (GrzB), and secreted TNF-α, IFN-γ, IL-6 of the cultured NK cells were significantly increased after treatment with Nr-CWS. Overall, the findings indicate that Nr-CWS could suppress the lung metastasis induced by B16F10 melanoma cells, which may be exerted through its effect on NK cells by promoting NK cell terminal differentiation (CD27lowCD11bhigh), and up-regulating the production of cytokines and cytotoxic molecules. C1 [Wu, Jie] The First Affiliated Hospital of Jinzhou Medical University, Department of OncologyJinzhou, China. [He, Baojun] The First Affiliated Hospital of Jinzhou Medical University, Department of Clinical Laboratory MedicineJinzhou, China. [Miao, Miao] Jinzhou Medical University, School of Basic Medical Science, Department of ImmunologyJinzhou, China. [Han, Xibin] Jinzhou Medical University, Laboratory Animal CenterJinzhou, China. [Dai, Hongyan] The First Affiliated Hospital of Jinzhou Medical University, Department of Outpatient PICCJinzhou, China. [Dou, Heng] Greatest Biopharma Limited CompanyBenxi, China. [Li, Yanqiu] Greatest Biopharma Limited CompanyBenxi, China. [Zhang, Xiaoqing] China Medical University, Teaching Center for Basic Medical ExperimentShenyang, China. [Wang, Guangchuan] Jinzhou Medical University, School of Basic Medical Science, Department of ImmunologyJinzhou, China. RP Wang, G (reprint author), Jinzhou Medical University, School of Basic Medical Science, Department of Immunology, Jinzhou, China. EM chuan560@163.com CR Kuyukina MS, Ivshina IB, Baeva TA, Kochina OA, Gein SV, Chereshnev VA. Trehalolipid Biosurfactants from Nonpathogenic Rhodococcus Actinobacteria with Diverse Immunomodulatory Activities. N Biotechnol, 2015, 32:559–68., DOI 10.1016/j.nbt.2015.03.006 Azuma I, Taniyama T, Yamawaki M, Sugimura K, Yamamura Y. Adjuvant and Antitumor Activities of Nocardia Cell-wall Skeletons. Gan, 1976, 67: 733–6. Meng Y, Sun J, Wang X, Ma Y, Kong C, Zhang G, et al. The Biological Macromolecule Nocardia Rubra Cell-wall Skeleton as an Avenue for Cell-Based Immunotherapy. J Cel Physiol, 2019, 2019., DOI 10.1002/jcp. 28182 Wang G, Wu J, Miao M, Dou H, Nan N, Shi M, et al. Nocardia Rubra Cell-wall Skeleton Promotes CD4(+, T Cell Activation and Drives Th1 Immune Response. Int J Biol Macromol, 2017, 101:398–407., DOI 10.1016/j.ijbiomac. 2017.03.060 Tao Y, Wang G, Zhai J, Zhang N. Functional Modulation of CD8+ T Cell by Approved Novel Immune Enhancer: Nocardia Rubra Cell-Wall Skeletons, Nr- CWS). Int Immunopharmacol, 2020, 78:106023., DOI 10.1016/j.intimp.2019. 106023 Chen W, Zhang Y, Zhao C, Shao S, Zhang Y, Li X, et al. Nocardia Rubra Cell Wall Skeleton Up-Regulates T Cell Subsets and Inhibits PD-1/pd-L1 Pathway to Promote Local Immune Status of Patients with High-Risk Human Papillomavirus Infection and Cervical Intraepithelial Neoplasia. Front Immunol, 2020, 11:612547., DOI 10.3389/fimmu.2020.612547 Wang Y, Hu Y, Ma B, Wang F, Liu S, Xu J, et al. Nocardia Rubra Cell wall Skeleton Accelerates Cutaneous Wound Healing by Enhancing Macrophage Activation and Angiogenesis. J Int Med Res, 2018, 46:2398–409., DOI 10.1177/ 0300060518764210 Rohde D, Gastl G, Biesterfeld S, Plante M, Jakse G. Local Expression of Cytokines in Rat Bladder Carcinoma Tissue after Intravesical Treatment with Nocardia Rubra Cell wall Skeleton and Bacille-Calmette-Guerin. Urol Res, 1997, 25:19–24., DOI 10.1007/BF00941901 Huang C, Tang X, Li S, Wang Q, Xie B, Xu J, et al. Immunopotentiator Aikejia Improves the Therapeutic Efficacy of PD-1/pd-L1 Immunosuppressive Pathway in CT26.WT Cancer Cell. J Cancer, 2019, 10:3472–80., DOI 10. 7150/jca.29672 Dell’Oste V, Biolatti M, Galitska G, Griffante G, Gugliesi F, Pasquero S, et al. Tuning the Orchestra: HCMV vs. Innate Immunity. Front Microbiol, 2020, 11: 661., DOI 10.3389/fmicb.2020.00661 Liu S, Galat V, Galat Y, Lee YKA, Wainwright D, Wu J. NK Cell-Based Cancer Immunotherapy: from Basic Biology to Clinical Development. J Hematol Oncol, 2021, 14:7., DOI 10.1186/s13045-020-01014-w Zhang X, Wei H. Role of Decidual Natural Killer Cells in Human Pregnancy and Related Pregnancy Complications. Front Immunol, 2021, 12:728291., DOI 10.3389/fimmu.2021.728291 Yu G, Xu X, Vu MD, Kilpatrick ED, Li XC. NK Cells Promote Transplant Tolerance by Killing Donor Antigen-Presenting Cells. J Exp Med, 2006, 203: 1851–8., DOI 10.1084/jem.20060603 Morvan MG, Lanier LL. NK Cells and Cancer: You Can Teach Innate Cells New Tricks. Nat Rev Cancer, 2016, 16:7–19., DOI 10.1038/nrc.2015.5 Grudzien M, Rapak A. Effect of Natural Compounds on NK Cell Activation. J Immunol Res, 2018, 2018:4868417., DOI 10.1155/2018/4868417 Bi J, Wang X. Molecular Regulation of NK Cell Maturation. Front Immunol, 2020, 11:1945., DOI 10.3389/fimmu.2020.01945 Debska-Zielkowska J, Moszkowska G, Zielinski M, Zielinska H, Dukat- Mazurek A, Trzonkowski P, et al. KIR Receptors as Key Regulators of NK Cells Activity in Health and Disease. Cells, 2021, 10:1777., DOI 10.3390/ cells10071777 Wu F, Ding XY, Li XH, Gong MJ, An JQ, Huang SL. Correlation between Elevated Inflammatory Cytokines of Spleen and Spleen index in Acute Spinal Cord Injury. J Neuroimmunol, 2020, 344:577264., DOI 10.1016/j.jneuroim. 2020.577264 Chen W, Liu J, Meng J, Lu C, Li X, Wang E, et al. Macrophage Polarization Induced by Neuropeptide Methionine Enkephalin, MENK, Promotes Tumoricidal Responses. Cancer Immunol Immunother, 2012, 61:1755–68., DOI 10.1007/s00262-012-1240-6 Wu X, Cheng B, Cai ZD, Lou LM. Determination of the Apoptotic index in Osteosarcoma Tissue and its Relationship with Patients Prognosis. Cancer Cel Int, 2013, 13:56., DOI 10.1186/1475-2867-13-56 Sivori S, Carlomagno S, Pesce S, Moretta A, Vitale M, Marcenaro E. TLR/NCR/ KIR: Which One to Use and when? Front Immunol, 2014, 5:105., DOI 10.3389/ fimmu.2014.00105 Kim S, Iizuka K, Aguila HL, Weissman IL, Yokoyama WM. In Vivo natural Killer Cell Activities Revealed by Natural Killer Cell-Deficient Mice. Proc Natl Acad Sci U S A, 2000, 97:2731–6., DOI 10.1073/pnas.050588297 Street SE, Zerafa N, Iezzi M, Westwood JA, Stagg J, Musiani P, et al. Host Perforin Reduces Tumor Number but Does Not Increase Survival in Oncogene-Driven Mammary Adenocarcinoma. Cancer Res, 2007, 67: 5454–60., DOI 10.1158/0008-5472.CAN-06-4084 Krneta T, Gillgrass A, Chew M, Ashkar AA. The Breast Tumor Microenvironment Alters the Phenotype and Function of Natural Killer Cells. Cell Mol Immunol, 2016, 13:628–39., DOI 10.1038/cmi. 2015.42 Eckelhart E, Warsch W, Zebedin E, Simma O, Stoiber D, Kolbe T, et al. A Novel Ncr1-Cre Mouse Reveals the Essential Role of STAT5 for NK-Cell Survival and Development. Blood, 2011, 117:1565–73., DOI 10.1182/blood- 2010-06-291633 Bjorkstrom NK, Strunz B, Ljunggren HG. Natural Killer Cells in Antiviral Immunity. Nat Rev Immunol, 2021, 22:112–23., DOI 10.1038/s41577-021- 00558-3 Sag D, Ayyildiz ZO, Gunalp S, Wingender G. The Role of TRAIL/DRs in the Modulation of Immune Cells and Responses. Cancers, Basel,, 2019, 11:1469., DOI 10.3390/cancers11101469 Rossin A, Miloro G, Hueber AO. TRAIL and FasL Functions in Cancer and Autoimmune Diseases: Towards an Increasing Complexity. Cancers, Basel,, 2019, 11:639., DOI 10.3390/cancers11050639 Fehniger TA, Cai SF, Cao X, Bredemeyer AJ, Presti RM, French AR, et al. Acquisition of Murine NK Cell Cytotoxicity Requires the Translation of a Preexisting Pool of Granzyme B and Perforin mRNAs. Immunity, 2007, 26: 798–811., DOI 10.1016/j.immuni.2007.04.010 Abel AM, Yang C, Thakar MS, Malarkannan S. Natural Killer Cells: Development, Maturation, and Clinical Utilization. Front Immunol, 2018, 9:1869., DOI 10.3389/fimmu.2018.01869 Fu B, Wang F, Sun R, Ling B, Tian Z, Wei H. CD11b and CD27 Reflect Distinct Population and Functional Specialization in Human Natural Killer Cells. Immunology, 2011, 133:350–9., DOI 10.1111/j.1365-2567.2011.03446.x Chiossone L, Chaix J, Fuseri N, Roth C, Vivier E, Walzer T. Maturation of Mouse NK Cells Is a 4-stage Developmental Program. Blood, 2009, 113: 5488–96., DOI 10.1182/blood-2008-10-187179 Bald T, Krummel MF, Smyth MJ, Barry KC. The NK Cell-Cancer Cycle: Advances and New Challenges in NK Cell-Based Immunotherapies. Nat Immunol, 2020, 21:835–47., DOI 10.1038/s41590-020-0728-z Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, et al. Innate or Adaptive Immunity? the Example of Natural Killer Cells. Science, 2011, 331:44–9., DOI 10.1126/science.1198687 Wu Y, Tian Z, Wei H. Developmental and Functional Control of Natural Killer Cells by Cytokines. Front Immunol, 2017, 8:930., DOI 10.3389/fimmu.2017.00930 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610555 EP 1610563 DI 10.3389/pore.2022.1610555 PG 9 ER PT J AU Chen, X Hu, G Xiong, L Xu, Q AF Chen, Xi Hu, Gang Xiong, Li Xu, Qingqing TI Relationships of Cuproptosis-Related Genes With Clinical Outcomes and the Tumour Immune Microenvironment in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunotherapy; hepatocellular carcinoma; immune infiltration; prognostic model; cuproptosis ID immunotherapy; hepatocellular carcinoma; immune infiltration; prognostic model; cuproptosis AB Background: Cuproptosis is a recently identified form of regulated cell death that plays a critical role in the onset and progression of various cancers. However, the effects of cuproptosis-related genes (CRGs) on hepatocellular carcinoma (HCC) are poorly understood. This study aimed to identify the cuproptosis subtypes and established a novel prognostic signature of HCC. Methods: We collected gene expression data and clinical outcomes from the TCGA, ICGC, and GEO datasets, analysed and identified 16 CRGs and the different subtypes of cuproptosis related to overall survival (OS), and further examined the differences in prognosis and immune infiltration among the subtypes. Subtypes-related differentially expressed genes (DEGs) were employed to build a prognostic signature. The relationship of the signature with the immune landscape as well as the sensitivity to different therapies was explored. Moreover, a nomogram was constructed to predict the outcome based on different clinicopathological characteristics. Results: Three cuproptosis subtypes were identified on the basis of 16 CRGs, and subtype B had an advanced clinical stage and worse OS. The immune response and function in subtype B were significantly suppressed, which may be an important reason for its poor prognosis. Based on the DEGs among the three subtypes, a prognostic model of five CRGs was constructed in the training set, and its predictive ability was validated in two external validation sets. HCC patients were classified into high and low-risk subgroups according to the risk score, and found that patients in the low-risk group showed significantly higher survival possibilities than those in the high-risk group (p < 0.001). The independent predictive performance of the risk score was assessed and verified by multivariate Cox regression analysis (p < 0.001). We further created an accurate nomogram to improve the clinical applicability of the risk score, showing good predictive ability and calibration. Low- and high-risk patients exhibit distinct immune cell infiltration and immune checkpoint changes. By further analyzing the risk score, patients in the highrisk group were found to be resistant to immunotherapy and a variety of chemotherapy drugs. Conclusion: Our study identified three cuproptosis subtypes and established a novel prognostic model that provides new insights into HCC subtype prognostic assessment and guides more effective treatment regimens. C1 [Chen, Xi] Edong Healthcare Group, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic UniversityHuangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Department of Thoracic OncologyHuangshi, China. [Hu, Gang] Edong Healthcare Group, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Department of Breast Surgery, Thyroid SurgeryHuangshi, China. [Xiong, Li] Edong Healthcare Group, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Department of RadiologyHuangshi, China. [Xu, Qingqing] Edong Healthcare Group, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Department of PathologyHuangshi, China. RP Xu, Q (reprint author), Edong Healthcare Group, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Department of Pathology, Huangshi, China. CR Kanwal F, Singal AG. Surveillance for Hepatocellular Carcinoma: Current Best Practice and Future Direction. Gastroenterology, 2019, 157:54–64., DOI 10. 1053/j.gastro.2019.02.049 Cadoux M, Caruso S, Pham S, Gougelet A, Pophillat C, Riou R, et al. Expression of NKG2D Ligands Is Downregulated by β-catenin Signalling and Associates with HCC Aggressiveness. J Hepatol, 2021, 74:1386–97., DOI 10. 1016/j.jhep.2021.01.017 Heller M, Parikh ND, Fidelman N, Owen D. Frontiers of Therapy for Hepatocellular Carcinoma. Abdom Radiol, Ny,, 2021, 46:3648–59., DOI 10. 1007/s00261-021-03065-0 Jiri T, Igor K, Mba. Hepatocellular Carcinoma Future Treatment Options. Klin Onkol, 2020, 33:26–9., DOI 10.14735/amko20203S26 Sangro B, Sarobe P, Hervas-Stubbs S, Melero I. Advances in Immunotherapy for Hepatocellular Carcinoma. Nat Rev Gastroenterol Hepatol, 2021, 18: 525–43., DOI 10.1038/s41575-021-00438-0 Zongyi Y, Xiaowu L. Immunotherapy for Hepatocellular Carcinoma. Cancer Lett, 2020, 470:8–17., DOI 10.1016/j.canlet.2019.12.002 Huang A, Yang XR, Chung WY, Dennison AR, Zhou J. Targeted Therapy for Hepatocellular Carcinoma. Signal Transduct Target Ther, 2020, 5:146., DOI 10. 1038/s41392-020-00264-x Gordan JD, Kennedy EB, Abou-Alfa GK, Beg MS, Brower ST, Gade TP, et al. Systemic Therapy for Advanced Hepatocellular Carcinoma: ASCO Guideline. J Clin Oncol, 2020, 38:4317–45., DOI 10.1200/jco.20.02672 Sauler M, Bazan IS, Lee PJ. Cell Death in the Lung: The Apoptosis-Necroptosis Axis. Annu Rev Physiol, 2019, 81:375–402., DOI 10.1146/annurev-physiol- 020518-114320 Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The Molecular Machinery of Regulated Cell Death. Cell Res, 2019, 29:347–64., DOI 10.1038/ s41422-019-0164-5 Koren E, Fuchs Y. Modes of Regulated Cell Death in Cancer. Cancer Discov, 2021, 11:245–65., DOI 10.1158/2159-8290.Cd-20-0789 Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an Iron-dependent Form of Nonapoptotic Cell Death. Cell, 2012, 149:1060–72., DOI 10.1016/j.cell.2012.03.042 Mou Y,Wang J, Wu J, He D, Zhang C, Duan C, et al. Ferroptosis, a New Form of Cell Death: Opportunities and Challenges in Cancer. J Hematol Oncol, 2019, 12:34., DOI 10.1186/s13045-019-0720-y Liang C, Zhang X, Yang M, Dong X. Recent Progress in Ferroptosis Inducers for Cancer Therapy. Adv Mater, 2019, 31:e1904197., DOI 10.1002/adma. 201904197 Tan Y, Chen Q, Li X, Zeng Z, Xiong W, Li G, et al. Pyroptosis: a New Paradigm of Cell Death for Fighting against Cancer. J Exp Clin Cancer Res, 2021, 40:153., DOI 10.1186/s13046-021-01959-x Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, et al. Copper Induces Cell Death by Targeting Lipoylated TCA Cycle Proteins. Science, 2022, 375:1254–61., DOI 10.1126/science.abf0529 Jiang Y, Huo Z, Qi X, Zuo T, Wu Z. Copper-induced Tumor Cell Death Mechanisms and Antitumor Theragnostic Applications of Copper Complexes. Nanomedicine, London, England,, 2022, 17:303–24., DOI 10.2217/nnm-2021- 0374 Yu Z, Zhou R, Zhao Y, Pan Y, Liang H, Zhang JS, et al. Blockage of SLC31A1-dependent Copper Absorption Increases Pancreatic Cancer Cell Autophagy to Resist Cell Death. Cell Prolif, 2019, 52:e12568., DOI 10.1111/ cpr.12568 Gao W, Huang Z, Duan J, Nice EC, Lin J, Huang C. Elesclomol Induces Copper-dependent Ferroptosis in Colorectal Cancer Cells via Degradation of ATP7A. Mol Oncol, 2021, 15:3527–44., DOI 10.1002/1878-0261.13079 Polishchuk EV, Merolla A, Lichtmannegger J, Romano A, Indrieri A, Ilyechova EY, et al. Activation of Autophagy, Observed in Liver Tissues from Patients with Wilson Disease and from ATP7B-Deficient Animals, Protects Hepatocytes from Copper-Induced Apoptosis. Gastroenterology, 2019, 156: 1173–89. e5., DOI 10.1053/j.gastro.2018.11.032 Saito A, Hayashi T, Okuno S, Ferrand-Drake M, Chan PH. Overexpression of Copper/zinc Superoxide Dismutase in Transgenic Mice Protects against Neuronal Cell Death after Transient Focal Ischemia by Blocking Activation of the Bad Cell Death Signaling Pathway. J Neurosci, 2003, 23:1710–8., DOI 10. 1523/jneurosci.23-05-01710.2003 Grasso M, Bond GJ, Kim YJ, Boyd S, Matson Dzebo M, Valenzuela S, et al. The Copper Chaperone CCS Facilitates Copper Binding to MEK1/2 to Promote Kinase Activation. J Biol Chem, 2021, 297:101314., DOI 10.1016/j.jbc.2021. 101314 Luo Q, Song Y, Kang J, Wu Y, Wu F, Li Y, et al. mtROS-mediated Akt/AMPK/ mTOR Pathway Was Involved in Copper-Induced Autophagy and it Attenuates Copper-Induced Apoptosis in RAW264.7 Mouse Monocytes. Redox Biol, 2021, 41:101912., DOI 10.1016/j.redox.2021.101912 Ren X, Li Y, Zhou Y, Hu W, Yang C, Jing Q, et al. Overcoming the Compensatory Elevation of NRF2 Renders Hepatocellular Carcinoma Cells More Vulnerable to Disulfiram/copper-Induced Ferroptosis. Redox Biol, 2021, 46:102122., DOI 10.1016/j.redox.2021.102122 Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, et al. Signatures of T Cell Dysfunction and Exclusion Predict Cancer Immunotherapy Response. Nat Med, 2018, 24: 1550–8., DOI 10.1038/s41591-018-0136-1 Kulik L, El-Serag HB. Epidemiology and Management of Hepatocellular Carcinoma. Gastroenterology, 2019, 156:477–91., DOI 10.1053/j.gastro.2018. 08.065 Ioannou GN. HCC Surveillance after SVR in Patients with F3/F4 Fibrosis. J Hepatol, 2021, 74:458–65., DOI 10.1016/j.jhep.2020.10.016 Carneiro BA, El-Deiry WS. Targeting Apoptosis in Cancer Therapy. Nat Rev Clin Oncol, 2020, 17:395–417., DOI 10.1038/s41571-020-0341-y Kaur P, Johnson A, Northcote-Smith J, Lu C, Suntharalingam K. Immunogenic Cell Death of Breast Cancer Stem Cells Induced by an Endoplasmic Reticulum-Targeting Copper(II, Complex. Chembiochem, 2020, 21:3618–24., DOI 10.1002/cbic.202000553 Li Y, Liang R, Zhang X, Wang J, Shan C, Liu S, et al. Copper Chaperone for Superoxide Dismutase Promotes Breast Cancer Cell Proliferation and Migration via ROS-Mediated MAPK/ERK Signaling. Front Pharmacol, 2019, 10:356., DOI 10.3389/fphar.2019.00356 Vyas A, Duvvuri U, Kiselyov K. Copper-dependent ATP7B Up-Regulation Drives the Resistance of TMEM16A-Overexpressing Head-And-Neck Cancer Models to Platinum Toxicity. Biochem J, 2019, 476:3705–19., DOI 10.1042/ bcj20190591 Gupta S. Cell Therapy to Remove Excess Copper in Wilson’s Disease. Ann N Y Acad Sci, 2014, 1315:70–80., DOI 10.1111/nyas.12450 Lorincz MT. Wilson Disease and Related Copper Disorders. Handb Clin Neurol, 2018, 147:279–92., DOI 10.1016/b978-0-444-63233-3.00018-x Chen J, Jiang Y, Shi H, Peng Y, Fan X, Li C. The Molecular Mechanisms of Copper Metabolism and its Roles in Human Diseases. Pflugers Arch, 2020, 472:1415–29., DOI 10.1007/s00424-020-02412-2 Li Y. Copper Homeostasis: Emerging Target for Cancer Treatment. IUBMB Life, 2020, 72:1900–8., DOI 10.1002/iub.2341 Singal AG, Lampertico P, Nahon P. Epidemiology and Surveillance for Hepatocellular Carcinoma: New Trends. J Hepatol, 2020, 72:250–61., DOI 10.1016/j.jhep.2019.08.025 Huang DQ, El-Serag HB, Loomba R. Global Epidemiology of NAFLD-Related HCC: Trends, Predictions, Risk Factors and Prevention. Nat Rev Gastroenterol Hepatol, 2021, 18:223–38., DOI 10.1038/s41575-020-00381-6 Antonucci L, Porcu C, Iannucci G, Balsano C, Barbaro B. Non-Alcoholic Fatty Liver Disease and Nutritional Implications: Special Focus on Copper. Nutrients, 2017, 9:E1137., DOI 10.3390/nu9101137 Porcu C, Antonucci L, Barbaro B, Illi B, Nasi S, MartiniM, et al. Copper/MYC/ CTR1 Interplay: a Dangerous Relationship in Hepatocellular Carcinoma. Oncotarget, 2018, 9:9325–43., DOI 10.18632/oncotarget.24282 Gunjan D, Shalimar, Nadda N, Kedia S, Nayak B, Paul SB, et al. Hepatocellular Carcinoma: An Unusual Complication of Longstanding Wilson Disease. J Clin Exp Hepatol, 2017, 7:152–4., DOI 10.1016/j.jceh.2016.09.012 Huo J, Cai J, Guan G, Liu H, Wu L. A Ferroptosis and Pyroptosis Molecular Subtype-Related Signature Applicable for Prognosis and Immune Microenvironment Estimation in Hepatocellular Carcinoma. Front Cel Dev Biol, 2021, 9:761839., DOI 10.3389/fcell.2021.761839 Liu Z, Wang L, Liu L, Lu T, Jiao D, Sun Y, et al. The Identification and Validation of Two Heterogenous Subtypes and a Risk Signature Based on Ferroptosis in Hepatocellular Carcinoma. Front Oncol, 2021, 11:619242., DOI 10.3389/fonc.2021.619242 Yang H, Jiang Q. A Multi-Omics-Based Investigation of the Immunological and Prognostic Impact of Necroptosis-Related Genes in Patients with Hepatocellular Carcinoma. J Clin Lab Anal, 2022, 36:e24346., DOI 10.1002/jcla.24346 Shi WK, Zhu XD, Wang CH, Zhang YY, Cai H, Li XL, et al. PFKFB3 Blockade Inhibits Hepatocellular Carcinoma Growth by Impairing DNA Repair through AKT. Cell Death Dis, 2018, 9:428., DOI 10.1038/s41419-018-0435-y Chen DP, Ning WR, Jiang ZZ, Peng ZP, Zhu LY, Zhuang SM, et al. Glycolytic Activation of Peritumoral Monocytes Fosters Immune Privilege via the PFKFB3-PD-L1 axis in Human Hepatocellular Carcinoma. J Hepatol, 2019, 71:333–43., DOI 10.1016/j.jhep.2019.04.007 Matsumoto K, Noda T, Kobayashi S, Sakano Y, Yokota Y, Iwagami Y, et al. Inhibition of Glycolytic Activator PFKFB3 Suppresses Tumor Growth and Induces Tumor Vessel Normalization in Hepatocellular Carcinoma. Cancer Lett, 2021, 500:29–40., DOI 10.1016/j.canlet.2020.12.011 Long Q, Zou X, Song Y, Duan Z, Liu L. PFKFB3/HIF-1α Feedback Loop Modulates Sorafenib Resistance in Hepatocellular Carcinoma Cells. Biochem Biophys Res Commun, 2019, 513:642–50., DOI 10.1016/j.bbrc.2019.03.109 Dai W, Wang Y, Yang T, Wang J, Wu W, Gu J. Downregulation of Exosomal CLEC3B in Hepatocellular Carcinoma Promotes Metastasis and Angiogenesis via AMPK and VEGF Signals. Cell Commun Signal, 2019, 17:113., DOI 10.1186/ s12964-019-0423-6 Xie XW, Jiang SS, Li X. CLEC3B as a Potential Prognostic Biomarker in Hepatocellular Carcinoma. Front Mol Biosci, 2020, 7:614034., DOI 10.3389/ fmolb.2020.614034 Mao X, Zhou L, Tey SK, Ma APY, Yeung CLS, Ng TH, et al. Tumour Extracellular Vesicle-Derived Complement Factor H Promotes Tumorigenesis and Metastasis by Inhibiting Complement-dependent Cytotoxicity of Tumour Cells. J Extracell Vesicles, 2020, 10:e12031., DOI 10. 1002/jev2.12031 Fu Y, Liu S, Zeng S, Shen H. From Bench to Bed: the Tumor Immune Microenvironment and Current Immunotherapeutic Strategies for Hepatocellular Carcinoma. J Exp Clin Cancer Res, 2019, 38:396., DOI 10. 1186/s13046-019-1396-4 Cheng AL, Hsu C, Chan SL, Choo SP, Kudo M. Challenges of Combination Therapy with Immune Checkpoint Inhibitors for Hepatocellular Carcinoma. J Hepatol, 2020, 72:307–19., DOI 10.1016/j. jhep.2019.09.025 Zhou G, Sprengers D, Boor PPC, Doukas M, Schutz H, Mancham S, et al. Antibodies against Immune Checkpoint Molecules Restore Functions of Tumor-Infiltrating T Cells in Hepatocellular Carcinomas. Gastroenterology, 2017, 153:1107–19. e10., DOI 10.1053/j.gastro.2017.06.017 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610558 EP 1610569 DI 10.3389/pore.2022.1610558 PG 12 ER PT J AU Lin, Y Li, Y Chen, X Chen, M Huang, J Guo, W Chen, L Wu, L Zhang, X Zhang, W Jin, X Zhang, J Fu, F Wang, Ch AF Lin, Yuxiang Li, Yan Chen, Xiaobin Chen, Minyan Huang, Jun Guo, Wenhui Chen, Lili Wu, Long Zhang, Xiang Zhang, Wenzhe Jin, Xuan Zhang, Jie Fu, Fangmeng Wang, Chuan TI Thrombospondin 2 is a Functional Predictive and Prognostic Biomarker for Triple-Negative Breast Cancer Patients With Neoadjuvant Chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; pathological response; neoadjuvant chemotherapy; triple-negative breast cancer; THBS2 ID biomarker; pathological response; neoadjuvant chemotherapy; triple-negative breast cancer; THBS2 AB Background: Triple-negative breast cancer (TNBC) is characterized by a more aggressive biological behavior and unfavorable outcome. Circulating and histological expression of THBS2 has been demonstrated to be a novel diagnostic and prognostic biomarker in patients with various types of tumors. However, few studies have evaluated the predictive and prognostic value of THBS2 in TNBC specifically. Methods: In total, 185 triple-negative breast cancer patients (TNBC) with preoperative neoadjuvant chemotherapy were enrolled in this study. Serum THBS2 (sTHBS2) level was measured both prior to the start of NAC and at surgery by enzyme-linked immunosorbent assay (ELISA). Histological THBS2 (hTHBS2) expression in patients with residual tumors was evaluated by immunohistochemistry (IHC) staining method. Correlations between variables and treatment response were studied. Kaplan-Meier plots and Cox proportional hazard regression model were applied for survival analysis. Functional activities of THBS2 in TNBC cells were determined by CCK-8 assay, colony formation, wound healing, and transwell assay. Results: Of the 185 patients, 48 (25.9%) achieved pathological complete response (pCR) after completion of NAC. Elevated pCR rates were observed in patients with a lower level of sTHBS2 at surgery and higher level of sTHBS2 change (OR = 0.88, 95%CI: 0.79–0.98, p = 0.020 and OR = 1.12, 95%CI: 1.02–1.23, p = 0.015, respectively). In survival analysis, hTHBS2 expression in residual tumor was of independent prognostic value for both disease-free survival (HR = 2.21, 95%CI = 1.24–3.94, p = 0.007) and overall survival (HR = 2.07, 95%CI = 1.09–3.92, p = 0.026). For functional studies, THBS2 was indicated to inhibit proliferation, migration, and invasion abilities of TNBC cells in vitro. Conclusion: Our findings confirmed the value of serum THBS2 level to predict pCR for TNBC patients and the prognostic performance of histological THBS2 expression in nonpCR responders after NAC. THBS2 might serve as a promising functional biomarker for patients with triple-negative breast cancer. C1 [Lin, Yuxiang] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. [Li, Yan] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. [Chen, Xiaobin] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. [Chen, Minyan] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. [Huang, Jun] Fujian Medical University Union Hospital, Department of Laboratory MedicineFuzhou, China. [Guo, Wenhui] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. [Chen, Lili] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. [Wu, Long] Fujian Medical University Union Hospital, Department of PathologyFuzhou, China. [Zhang, Xiang] Fujian Medical University Union Hospital Pingtan Branch, Department of PathologyFuzhou, China. [Zhang, Wenzhe] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. [Jin, Xuan] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. [Zhang, Jie] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. [Fu, Fangmeng] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. [Wang, Chuan] Fujian Medical University Union Hospital, Department of Breast SurgeryFuzhou, China. RP Wang, Ch (reprint author), Fujian Medical University Union Hospital, Department of Breast Surgery, Fuzhou, China. EM chuanwang1968@outlook.com CR Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, et al. Triple-negative Breast Cancer: Clinical Features and Patterns of Recurrence. Clin Cancer Res, 2007, 13(15):4429–34., DOI 10.1158/1078-0432.CCR-06- 3045 Gluz O, Liedtke C, Gottschalk N, Pusztai L, Nitz U, Harbeck N. Triple-negative Breast Cancer–Current Status and Future Directions. Ann Oncol, 2009, 20(12):1913–27., DOI 10.1093/annonc/mdp492 Huang M, O’Shaughnessy J, Zhao J, Haiderali A, Cortes J, Ramsey SD, et al. Association of Pathologic Complete Response with Long-Term Survival Outcomes in Triple-Negative Breast Cancer: A Meta-Analysis. Cancer Res, 2020, 80:5427–34., DOI 10.1158/0008-5472.CAN-20-1792 Harbeck N, Gnant M. Breast Cancer. Lancet, 2017, 389(10074):1134–50., DOI 10.1016/S0140-6736(16)31891-8 Symmans WF, Wei C, Gould R, Yu X, Zhang Y, Liu M, et al. Long-Term Prognostic Risk after Neoadjuvant Chemotherapy Associated with Residual Cancer Burden and Breast Cancer Subtype. J Clin Oncol, 2017, 35:1049–60., DOI 10.1200/JCO.2015.63.1010 Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological Complete Response and Long-Term Clinical Benefit in Breast Cancer: the CTNeoBC Pooled Analysis. Lancet, 2014, 384(9938):164–72., DOI 10.1016/S0140-6736(13)62422-8 Fornier M, Fumoleau P. The Paradox of Triple Negative Breast Cancer: Novel Approaches to Treatment. Breast J, 2012, 18(1):41–51., DOI 10.1111/j.1524- 4741.2011.01175.x Bornstein P, Armstrong LC, Hankenson KD, Kyriakides TR, Yang Z. Thrombospondin 2, a Matricellular Protein with Diverse Functions. Matrix Biol, 2012, 19(7):557–68., DOI 10.1016/s0945-053x(00)00104-9 Krady MM, Zeng J, Yu J, MacLauchlan S, Skokos EA, Tian W, et al. Thrombospondin-2 Modulates Extracellular Matrix Remodeling during Physiological Angiogenesis. Am J Pathol, 2008, 173(3):879–91., DOI 10.2353/ ajpath.2008.080128 Calabro NE, Kristofik NJ, Kyriakides TR. Thrombospondin-2 and Extracellular Matrix Assembly. Biochim Biophys Acta, 2014, 1840(8): 2396–402., DOI 10.1016/j.bbagen.2014.01.013 Jeong SY, Kim DH, Ha J, Jin HJ, Kwon SJ, Chang JW, et al. Thrombospondin-2 Secreted by Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells Promotes Chondrogenic Differentiation. Stem Cells, 2013, 31(10):2136–48., DOI 10.1002/stem.1471 Liu JF, Lee CW, Tsai MH, Tang CH, Chen PC, Lin LW, et al. Thrombospondin 2 Promotes Tumor Metastasis by Inducing Matrix Metalloproteinase-13 Production in Lung Cancer Cells. Biochem Pharmacol, 2018, 155:537–46., DOI 10.1016/j.bcp.2018.07.024 Chen PC, Tang CH, Lin LW, Tsai CH, Chu CY, Lin TH, et al. Thrombospondin-2 Promotes Prostate Cancer Bone Metastasis by the Up- Regulation of Matrix Metalloproteinase-2 through Down-Regulating miR- 376c Expression. J Hematol Oncol, 2017, 10(1)(33)., DOI 10.1186/s13045-017- 0390-6 PanW, Song XY, Hu QB, Zhang M, Xu XH. TSP2 Acts as a Suppresser of Cell Invasion, Migration and Angiogenesis in Medulloblastoma by Inhibiting the Notch Signaling Pathway. Brain Res, 2019, 1718:223–30., DOI 10.1016/j. brainres.2019.05.004 Le Large TYS, Meijer LL, Paleckyte R, Boyd LNC, Kok B, Wurdinger T, et al. Combined Expression of Plasma Thrombospondin-2 and CA19-9 for Diagnosis of Pancreatic Cancer and Distal Cholangiocarcinoma: A Proteome Approach. Oncologist, 2020, 25(4):e634–e643., DOI 10.1634/ theoncologist.2019-0680 Peng HY, Chang MC, Hu CM, Yang HI, Lee WH, Chang YT. Thrombospondin-2 Is a Highly Specific Diagnostic Marker and Is Associated with Prognosis in Pancreatic Cancer. Ann Surg Oncol, 2019, 26:807–14., DOI 10.1245/s10434-018-07109-6 Kim J, Bamlet WR, Oberg AL, Chaffee KG, Donahue G, Cao XJ, et al. Detection of Early Pancreatic Ductal Adenocarcinoma with Thrombospondin-2 and CA19-9 Blood Markers. Sci Transl Med, 2017, 9:eaah5583., DOI 10.1126/ scitranslmed.aah5583 Byrling J, KristlT,HuD, Pla I, SanchezA, SasorA, et al.Mass Spectrometry-Based Analysis of Formalin-Fixed, Paraffin-Embedded Distal Cholangiocarcinoma Identifies Stromal Thrombospondin-2 as a Potential Prognostic Marker. J Transl Med, 2020, 18(1)(343)., DOI 10.1186/s12967-020-02498-3 Jiang YM, Yu DL, Hou GX, Jiang JL, Zhou Q, Xu XF. SerumThrombospondin-2 Is a CandidateDiagnosis Biomarker for Early Non-small-cell Lung Cancer. Biosci Rep, 2019, 39:BSR20190476., DOI 10.1042/BSR2019047639(7):BSR20190476 Naumnik W, Ossolinska M, Plonska I, Chyczewska E, Niklinski J. Circulating Thrombospondin-2 and FGF-2 in Patients with Advanced Non-small Cell Lung Cancer: Correlation with Survival. Adv Exp Med Biol, 2015, 833:9–14., DOI 10.1007/5584_2014_78 Tian Q, Liu Y, Zhang Y, Song Z, Yang J, Zhang J, et al. THBS2 Is a Biomarker for AJCC Stages and a Strong Prognostic Indicator in Colorectal Cancer. J BUON, 2018, 23:1331–6. Wang X, Zhang L, Li H, Sun W, Zhang H, Lai M. THBS2 Is a Potential Prognostic Biomarker in Colorectal Cancer. Sci Rep, 2016, 6:33366., DOI 10. 1038/srep33366 Lin Y, Lin L, Fu F,Wang C, Hu A, Xie J, et al. Quantitative Proteomics Reveals Stage-specific Protein Regulation of Triple Negative Breast Cancer. Breast Cancer Res Treat, 2021, 185(1):39–52., DOI 10.1007/s10549-020-05916-8 Symmans WF, Peintinger F, Hatzis C, Rajan R, Kuerer H, Valero V, et al. Measurement of Residual Breast Cancer Burden to Predict Survival after Neoadjuvant Chemotherapy. J Clin Oncol, 2007, 25(28):4414–22., DOI 10.1200/ JCO.2007.10.6823 Zhao S, Ma D, Xiao Y, Li XM,Ma JL, Zhang H, et al. Molecular Subtyping of Triple-Negative Breast Cancers by Immunohistochemistry: Molecular Basis and Clinical Relevance. Oncologist, 2020, 25(10):e1481–e1491., DOI 10.1634/theoncologist.2019-0982 Hugo C, Daniel C. Thrombospondin in Renal Disease. Nephron Exp Nephrol, 2009, 111(3):e61–e66., DOI 10.1159/000198235 Iruela-Arispe ML, Luque A, Lee N. Thrombospondin Modules and Angiogenesis. Int J Biochem Cel Biol, 2004, 36(6):1070–8., DOI 10.1016/ j.biocel.2004.01.025 Mirochnik Y, Kwiatek A, Volpert OV. Thrombospondin and Apoptosis: Molecular Mechanisms and Use for Design of Complementation Treatments. Curr Drug Targets, 2008, 9(10):851–62., DOI 10.2174/ 138945008785909347 Rahman MT,Muppala S, Wu J, Krukovets I, Solovjev D, Verbovetskiy D, et al. Effects of Thrombospondin-4 on Pro-inflammatory Phenotype Differentiation and Apoptosis in Macrophages. Cell Death Dis, 2020, 53., DOI 10.1038/s41419-020-2237-2 Chang IW, Li CF, Lin VC, He HL, Liang PI, Wu WJ, et al. Prognostic Impact of Thrombospodin-2, THBS2, Overexpression on Patients with Urothelial Carcinomas of Upper Urinary Tracts and Bladders. J Cancer, 2016, 7:1541–9., DOI 10.7150/jca.15696 Hsu CW, Yu JS, Peng PH, Liu SC, Chang YS, Chang KP, et al. Secretome Profiling of Primary Cells Reveals that THBS2 Is a Salivary Biomarker of Oral Cavity Squamous Cell Carcinoma. J Proteome Res, 2014, 13(11): 4796–807., DOI 10.1021/pr500038k Dave RV, Millican-Slater R, Dodwell D, Horgan K, Sharma N. Neoadjuvant Chemotherapy with MRI Monitoring for Breast Cancer. Br J Surg, 2017, 104(9):1177–87., DOI 10.1002/bjs.10544 Bae MS, Shin SU, Ryu HS, Han W, Im SA, Park IA, et al. Pretreatment MR Imaging Features of Triple-Negative Breast Cancer: Association with Response to Neoadjuvant Chemotherapy and Recurrence-free Survival. Radiology, 2016, 281(2):392–400., DOI 10.1148/radiol.2016152331 Golshan M, Wong SM, Loibl S, Huober JB, O’Shaughnessy J, Rugo HS, et al. Early Assessment with Magnetic Resonance Imaging for Prediction of Pathologic Response to Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer: Results from the Phase III BrighTNess Trial. Eur J Surg Oncol, 2020, 46(2):223–8., DOI 10.1016/j.ejso.2019.10.002 Marinovich ML, Houssami N, Macaskill P, Sardanelli F, Irwig L, Mamounas EP, et al. Meta-analysis of Magnetic Resonance Imaging in Detecting Residual Breast Cancer after Neoadjuvant Therapy. J Natl Cancer Inst, 2013, 105(5):321–33., DOI 10.1093/jnci/djs528 Wang RX, Chen S, Huang L, Zhou Y, Shao ZM. Monitoring Serum VEGF in Neoadjuvant Chemotherapy for Patients with Triple-Negative Breast Cancer: A New Strategy for Early Prediction of Treatment Response and Patient Survival. Oncologist, 2019, 24(6):753–61., DOI 10.1634/theoncologist.2017- 0602 Li Y, Cui X, Yang YJ, Chen QQ, Zhong L, Zhang T, et al. SerumsPD-1 and sPD-L1 as Biomarkers for Evaluating the Efficacy of Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer Patients. Clin Breast Cancer, 2019, 19(5):326–32., DOI 10.1016/j.clbc.2019.03.008 Balko JM, Giltnane JM, Wang K, Schwarz LJ, Young CD, Cook RS, et al. Molecular Profiling of the Residual Disease of Triple-Negative Breast Cancers after Neoadjuvant Chemotherapy Identifies Actionable Therapeutic Targets. Cancer Discov, 2014, 4(2):232–45., DOI 10.1158/ 2159-8290.CD-13-0286 Romero A, Garcia-Saenz JA, Fuentes-Ferrer M, Lopez Garcia-Asenjo JA, Furio V, Roman JM, et al. Correlation between Response to Neoadjuvant Chemotherapy and Survival in Locally Advanced Breast Cancer Patients. Ann Oncol, 2013, 24:655–61., DOI 10.1093/annonc/mds493 Sejben A, Koszo R, Kahan Z, Cserni G, Zombori T. Examination of Tumor Regression Grading Systems in Breast Cancer Patients Who Received Neoadjuvant Therapy. Pathol Oncol Res, 2020, 26:2747–54., DOI 10.1007/ s12253-020-00867-3 Laas E, Labrosse J, Hamy AS, Benchimol G, de Croze D, Feron JG, et al. Determination of Breast Cancer Prognosis after Neoadjuvant Chemotherapy: Comparison of Residual Cancer Burden, RCB, and Neo-Bioscore. Br J Cancer, 2021, 124(8):1421–7., DOI 10.1038/s41416-020-01251-3 Ao R, Guan L, Wang Y, Wang JN. Silencing of COL1A2, COL6A3, and THBS2 Inhibits Gastric Cancer Cell Proliferation, Migration, and Invasion While Promoting Apoptosis through the PI3k-Akt Signaling Pathway. J Cel Biochem, 2018, 119(6):4420–34., DOI 10. 1002/jcb.26524 Liu JF, Chen PC, Chang TM, Hou CH. Thrombospondin-2 Stimulates MMP-9 Production and Promotes Osteosarcoma Metastasis via the PLC, PKC, C-Src and NF-Κb Activation. J Cel Mol Med, 2020, 24:12826–39., DOI 10.1111/jcmm. 15874 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610559 EP 1610570 DI 10.3389/pore.2022.1610559 PG 12 ER PT J AU Stukaite-Ruibiene, E Norvilas, R Dirse, V Stankeviciene, S Vaitkeviciene, EG AF Stukaite-Ruibiene, Egle Norvilas, Rimvydas Dirse, Vaidas Stankeviciene, Sigita Vaitkeviciene, Elizabeta Goda TI Case Report: Specific ABL-Inhibitor Imatinib Is an Effective Targeted Agent as the First Line Therapy to Treat B-Cell Acute Lymphoblastic Leukemia With a Cryptic NUP214::ABL1 Gene Fusion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE case report; targeted therapy; tyrosine kinase inhibitors; acute lymphoblastic leukemia; imatinib; BCRABL1- like ID case report; targeted therapy; tyrosine kinase inhibitors; acute lymphoblastic leukemia; imatinib; BCRABL1- like AB Acute lymphoblastic leukemia (ALL) with recurrent genetic lesions, affecting a series of kinase genes, is associated with unfavorable prognosis, however, it could benefit from treatment with tyrosine kinase inhibitors (TKI). NUP214::ABL1 fusion is detected in 6% of T-cell acute lymphoblastic leukemia (T-ALL), and is very rare in B-ALL. We present a case of adolescent with B-ALL and a cryptic NUP214::ABL1 fusion which was initially missed during diagnostic screening and was detected by additional RNA sequencing. Treatment with specific ABL-inhibitor Imatinib was added later in therapy with a good effect. Initial treatment according to conventional chemotherapy was complicated by severe side effects. At the end of Consolidation, the patient was stratified to a high risk group with allogeneic hematopoietic stem cell transplantation because of insufficient response to therapy. At that time, targeted RNA sequencing detected NUP214::ABL1 gene fusion which was previously missed due to a small microduplication in the 9q34 chromosome region. Gene variant analysis revealed no TKI-resistant ABL1 mutations; therefore, treatment with Imatinib was added to target the NUP214::ABL1 fusion protein. A negative minimal residual disease was achieved, and treatment was downgraded to intermediate risk protocol. Combining routine genetic assays with next-generation sequencing methods could prevent from missing atypical gene alterations. Identification of rare targetable genetic subtypes is of importance in order to introduce targeted therapy as early as possible that may improve survival and reduce toxicity. Treatment with ABL1 inhibitor imatinib mesylate revealed as a highly effective targeted therapy against the leukemia driving protein kinase. C1 [Stukaite-Ruibiene, Egle] Faculty of Medicine of Vilnius UniversityVilnius, Lithuania. [Norvilas, Rimvydas] Vilnius University Hospital Santaros Klinikos, Hematology, Oncology and Transfusion Medicine CenterVilnius, Lithuania. [Dirse, Vaidas] Vilnius University Hospital Santaros Klinikos, Hematology, Oncology and Transfusion Medicine CenterVilnius, Lithuania. [Stankeviciene, Sigita] Vilnius University Hospital Santaros Klinikos, Center for Pediatric Oncology and HematologyVilnius, Lithuania. [Vaitkeviciene, Elizabeta Goda] Faculty of Medicine of Vilnius UniversityVilnius, Lithuania. RP Stukaite-Ruibiene, E (reprint author), Faculty of Medicine of Vilnius University, Vilnius, Lithuania. EM egle.eglaite@gmail.com CR Tasian SK, Loh ML, Hunger SP. Philadelphia Chromosome-like Acute Lymphoblastic Leukemia. Blood, 2017, 130(19):2064–72., DOI 10.1182/ blood-2017-06-743252 Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, et al. The 2016 Revision to the World Health Organization Classification of Myeloid Neoplasms and Acute Leukemia. Blood, 2016, 127(20):2391–405., DOI 10.1182/ blood-2016-03-643544 Den Boer ML, van Slegtenhorst M, De Menezes RX, Cheok MH, Buijs- Gladdines JGCAM, Peters STCJM, et al. A Subtype of Childhood Acute Lymphoblastic Leukaemia with Poor Treatment Outcome: a Genome-wide Classification Study. Lancet Oncol, 2009, 10(2):125–34., DOI 10.1016/S1470- 2045(08)70339-5 De Braekeleer E, Douet-Guilbert N, Rowe D, Bown N, Morel F, Berthou C, et al. ABL1 Fusion Genes in Hematological Malignancies: a Review. Eur J Haematol, 2011, 86(5):361–71., DOI 10.1111/j.1600-0609.2011.01586.x Roberts KG, Li Y, Payne-Turner D, Harvey RC, Yang YL, Pei D, et al. Targetable Kinase-Activating Lesions in Ph-like Acute Lymphoblastic Leukemia. N Engl J Med, 2014, 371:1005–15., DOI 10.1056/ NEJMoa1403088 Deenik W, Beverloo HB, van der Poel-van de Luytgaarde Scp a. M, Wattel MM, van Esser Jwj, Valk PJM, et al. Rapid Complete Cytogenetic Remission after Upfront Dasatinib Monotherapy in a Patient with a NUP214-ABL1- Positive T-Cell Acute Lymphoblastic Leukemia. Leukemia, 2009, 23(3):627–9., DOI 10.1038/leu.2008.318 Stergianou K, Fox C, Russell NH. Fusion of NUP214 to ABL1 on Amplified Episomes in T-ALL-Iimplications for Treatment. Leukemia, 2005, 19(9): 1680–1., DOI 10.1038/sj.leu.2403877 Koschmieder S, Burmeister T, Bruggemann M, Berkemeier A, Volpert S, Wieacker P, et al. Molecular Monitoring in NUP214-ABL-Positive T-Acute Lymphoblastic Leukemia Reveals Clonal Diversity and Helps to Guide Targeted Therapy. Leukemia, 2014, 28(2):419–22., DOI 10.1038/leu.2013.272 Tsurusaki Y, Nagai J, Fujita S, Sugiyama M, Nakamura W, Hayashi A, et al. Whole-exome Sequencing Reveals the Subclonal Expression of NUP214-ABL1 Fusion Gene in T-Cell Acute Lymphoblastic Leukemia. Pediatr Blood Cancer, 2020, 67(1):e28019., DOI 10.1002/pbc.28019 Aldoss I, Pullarkat V. Response to Single Agent Dasatinib post Allogeneic Transplant in B-Cell Acute Lymphoblastic Leukemia with NUP214-ABL1. Leuk Lymphoma, 2019, 60(11):2832–4., DOI 10.1080/10428194.2019. 1605510 Chen Y, Zhang L, Huang J, Hong X, Zhao J, Wang Z, et al. Dasatinib and Chemotherapy in a Patient with Early T-Cell Precursor Acute Lymphoblastic Leukemia and NUP214-ABL1 Fusion: A Case Report. Exp Ther Med, 2017, 14(5):3979–84., DOI 10.3892/etm.2017.5046 Coccaro N, Anelli L, Zagaria A, Specchia G, Albano F. Next-Generation Sequencing in Acute Lymphoblastic Leukemia. Int J Mol Sci, 2019, 20(12): 2929., DOI 10.3390/ijms20122929 Sherali N, Hamadneh T, Aftab S, Alfonso M, Tsouklidis N. Integration of Next-Generation Sequencing in Diagnosing and Minimal Residual Disease Detection in Patients with Philadelphia Chromosome-like Acute Lymphoblastic Leukemia. Cureus, 2020, 12(9):e10696., DOI 10.7759/cureus. 10696 Norvilas R, Dirse V, Semaskeviciene R, Mickeviciute O, Gineikiene E, Stoskus M, et al. Low Incidence of ABL-Class and JAK-STAT Signaling Pathway Alterations in Uniformly Treated Pediatric and Adult B-Cell Acute Lymphoblastic Leukemia Patients Using MRD Risk-Directed Approach - a Population-Based Study. BMC Cancer, 2021, 21(1):326., DOI 10.1186/s12885- 020-07781-6 Burmeister T, Gokbuget N, Reinhardt R, Rieder H, Hoelzer D, Schwartz S. NUP214-ABL1 in Adult T-ALL: the GMALL Study Group Experience. Blood, 2006, 108(10):3556–9., DOI 10.1182/blood-2006-04-014514 Rossari F, Minutolo F, Orciuolo E. Past, Present, and Future of Bcr-Abl Inhibitors: from Chemical Development to Clinical Efficacy. J Hematol Oncol, 2018, 11:84., DOI 10.1186/s13045-018-0624-2 Hunger SP. Tyrosine Kinase Inhibitor Use in Pediatric Philadelphia Chromosome–Positive Acute Lymphoblastic Anemia. Hematol Am Soc Hematol Educ Program, 2011, 2011(1):361–5., DOI 10.1182/asheducation- 2011.1.361 Roberts KG, Morin RD, Zhang J, Hirst M, Zhao Y, Su X, et al. Genetic Alterations Activating Kinase and Cytokine Receptor Signaling in High-Risk Acute Lymphoblastic Leukemia. Cancer Cell, 2012, 22(2):153–66., DOI 10.1016/ j.ccr.2012.06.005 Duployez N, Grzych G, Ducourneau B, Fuentes MA, Grardel N, Boyer T, et al. NUP214-ABL1 Fusion Defines a Rare Subtype of B-Cell Precursor Acute Lymphoblastic Leukemia that Could Benefit from Tyrosine Kinase Inhibitors. Haematologica, 2016, 101(4):e133–4., DOI 10.3324/haematol.2015.136499 Tsujimoto SI, Nakano Y, Osumi T, Okada K, Ouchi-Uchiyama M, Kataoka K, et al. A Cryptic NUP214-ABL1 Fusion in B-Cell Precursor Acute Lymphoblastic Leukemia. J Pediatr Hematol Oncol, 2018, 40(6):e397–9., DOI 10.1097/MPH.0000000000001007 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610570 EP 1610575 DI 10.3389/pore.2022.1610570 PG 6 ER PT J AU Hu, B Hao, Sh Miao, Y Deng, Y Wang, J Wan, H Zhang, Sh Ji, N Feng, J AF Hu, Boyi Hao, Shuyu Miao, Yazhou Deng, Yuxuan Wang, Jing Wan, Hong Zhang, Shaodong Ji, Nan Feng, Jie TI Inhibiting PP2A Upregulates B7-H3 Expression and Potentially Increases the Sensitivity of Malignant Meningiomas to Immunotherapy by Proteomics SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immune checkpoint; malignant meningiomas; PP2A inhibitor; B7-H3; tumor proliferation ID immune checkpoint; malignant meningiomas; PP2A inhibitor; B7-H3; tumor proliferation AB Malignantmeningiomas have a highmortality rate and short survival time and currently have no effective treatment. In our study, proteomics analysis was performed to identify highly expressed proteins as therapeutic targets in malignant meningiomas. Cell Counting Kit-8 (CCK-8) assays were performed to verify the effect of LB-100 on the growth of malignant meningiomas. In addition, immunoblotting was used to verify the expression of B7-H3 and phosphorylation of STAT1 (Tyr701) in tissues and cells. Our results show that STAT1 and CD276 (B7-H3) regulated by PP2A were enriched in GO_IMMUNE_EFFECTOR_PROCESS and GO_REGULATION_OF_IMMUNE_SYSTEM_PROCESS. The immunotherapy target protein B7-H3 was confirmed to be upregulated in malignant meningiomas compared with meningothelial (p = 0.0001) and fibroblastic (p = 0.0046) meningiomas. In vitro, the PP2A inhibitor LB-100 suppressed the growth and invasion of malignant meningioma cells. Notably, the PP2A inhibitor LB-100 increased the phosphorylation of STAT1, thereby increasing the expression of the immune checkpoint protein B7-H3 in malignant meningioma cells in vitro. In conclusion, B7-H3 was found to be upregulated in malignant meningiomas. The PP2A inhibitor LB-100 increased the phosphorylation of STAT1 and B7-H3 expression, which could increase the sensitivity of malignant meningiomas to B7-H3 targeted immunotherapy. C1 [Hu, Boyi] Beijing Neurosurgical InstituteBeijing, China. [Hao, Shuyu] Capital Medical University, Beijing Tiantan Hospital, Department of Neurosurgery / China National Clinical Research Center for Neurological DiseasesBeijing, China. [Miao, Yazhou] Capital Medical University, Beijing Tiantan Hospital, Department of Neurosurgery / China National Clinical Research Center for Neurological DiseasesBeijing, China. [Deng, Yuxuan] Capital Medical University, Beijing Tiantan Hospital, Department of Neurosurgery / China National Clinical Research Center for Neurological DiseasesBeijing, China. [Wang, Jing] Beijing Neurosurgical InstituteBeijing, China. [Wan, Hong] Beijing Neurosurgical InstituteBeijing, China. [Zhang, Shaodong] Beijing Neurosurgical InstituteBeijing, China. [Ji, Nan] Capital Medical University, Beijing Tiantan Hospital, Department of Neurosurgery / China National Clinical Research Center for Neurological DiseasesBeijing, China. [Feng, Jie] Beijing Neurosurgical InstituteBeijing, China. RP Feng, J (reprint author), Beijing Neurosurgical Institute, Beijing, China. EM fengjie111@ccmu.edu.cn CR Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, Barnholtz-Sloan JS. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013-2017. Neuro Oncol, 2020, 22(12):iv1–iv96., DOI 10.1093/neuonc/noaa200 Buerki RA, Horbinski CM, Kruser T, Horowitz PM, James CD, Lukas RV. An Overview of Meningiomas. Future Oncol, 2018, 14(21):2161–77., DOI 10.2217/ fon-2018-0006 Kaur A, Westermarck J. Regulation of Protein Phosphatase 2A, PP2A, Tumor Suppressor Function by PME-1. Biochem Soc Trans, 2016, 44(6):1683–93., DOI 10.1042/BST20160161 Wlodarchak N, Xing Y. PP2A as a Master Regulator of the Cell Cycle. Crit Rev Biochem Mol Biol, 2016, 51(3):162–84., DOI 10.3109/10409238.2016. 1143913 Qian H, Wang J, Shang Q, Xiao J, Chen G, Gao B, et al. The Effect of Protein Phosphatase 2A Inhibitor LB100 on Regulating Methamphetamine Induced Conditioned Place Preference in Mice. Neurosci Lett, 2020, 721:134817., DOI 10.1016/j.neulet.2020.134817 Owen KL, Brockwell NK, Parker BS. JAK-STAT Signaling: A Double-Edged Sword of Immune Regulation and Cancer Progression. Cancers, Basel,, 2019, 11(12):E2002., DOI 10.3390/cancers11122002 Gao Y, Yang J, Cai Y, Fu S, Zhang N, Fu X, et al. IFN-gamma-mediated Inhibition of Lung Cancer Correlates with PD-L1 Expression and Is Regulated by PI3K-AKT Signaling. Int J Cancer, 2018, 143(4):931–43., DOI 10.1002/ijc. 31357 Lv H, Lv G, Chen C, Zong Q, Jiang G, Ye D, et al. NAD(+, Metabolism Maintains Inducible PD-L1 Expression to Drive Tumor Immune Evasion. Cell Metab, 2021, 33(1):110–27.e5. e5., DOI 10.1016/j.cmet.2020.10.021 Chen S, Crabill GA, Pritchard TS, McMiller TL, Wei P, Pardoll DM, et al. Mechanisms Regulating PD-L1 Expression on Tumor and Immune Cells. J Immunother Cancer, 2019, 7(1):305., DOI 10.1186/s40425-019-0770-2 Flem-Karlsen K, Fodstad O, Nunes-Xavier CE. B7-H3 Immune Checkpoint Protein in Human Cancer. Curr Med Chem, 2020, 27(24):4062–86., DOI 10. 2174/0929867326666190517115515 Lee YH, Martin-Orozco N, Zheng P, Li J, Zhang P, Tan H, et al. Inhibition of the B7-H3 Immune Checkpoint Limits Tumor Growth by Enhancing Cytotoxic Lymphocyte Function. Cell Res, 2017, 27(8):1034–45., DOI 10. 1038/cr.2017.90 Chen L, Chen J, Xu B, Wang Q, Zhou W, Zhang G, et al. B7-H3 Expression Associates with Tumor Invasion and Patient’s Poor Survival in Human Esophageal Cancer. Am J Transl Res, 2015, 7(12):2646–60. Xie J, Sun M, Zhang D, Chen C, Lin S, Zhang G. Fibronectin Enhances Tumor Metastasis through B7-H3 in clear Cell Renal Cell Carcinoma. FEBS Open Bio, 2021, 11(11):2977–87., DOI 10.1002/2211-5463.13280 Yu TT, Zhang T, Lu X, Wang RZ. B7-H3 Promotes Metastasis, Proliferation, and Epithelial-Mesenchymal Transition in Lung Adenocarcinoma. Onco Targets Ther, 2018, 11:4693–700., DOI 10.2147/OTT.S169811 Purvis IJ, Avilala J, Guda MR, Venkataraman S, Vibhakar R, Tsung AJ, et al. Role of MYC-miR-29-B7-H3 in Medulloblastoma Growth and Angiogenesis. J Clin Med, 2019, 8(8):E1158., DOI 10.3390/jcm8081158 Loo D, Alderson RF, Chen FZ, Huang L, Zhang W, Gorlatov S, et al. Development of an Fc-Enhanced Anti-B7-H3 Monoclonal Antibody with Potent Antitumor Activity. Clin Cancer Res, 2012, 18(14):3834–45., DOI 10. 1158/1078-0432.CCR-12-0715 Tang X, Liu F, Liu Z, Cao Y, Zhang Z, Wang Y, et al. Bioactivity and Safety of B7-H3-Targeted Chimeric Antigen Receptor T Cells against Anaplastic Meningioma. Clin Transl Immunol, 2020, 9(6):e1137., DOI 10.1002/cti2.1137 Scribner JA, Brown JG, Son T, Chiechi M, Li P, Sharma S, et al. Preclinical Development of MGC018, a Duocarmycin-Based Antibody-Drug Conjugate Targeting B7-H3 for Solid Cancer. Mol Cancer Ther, 2020, 19(11):2235–44., DOI 10.1158/1535-7163.MCT-20-0116 Feng J, Yu SY, Li CZ, Li ZY, Zhang YZ. Integrative Proteomics and Transcriptomics Revealed that Activation of the IL-6R/JAK2/STAT3/ MMP9 Signaling Pathway Is Correlated with Invasion of Pituitary Null Cell Adenomas. Mol Cell Endocrinol, 2016, 436:195–203., DOI 10.1016/j.mce. 2016.07.025 Shanker V, Trincucci G, Heim HM, Duong HT. Protein Phosphatase 2A Impairs IFNα-Induced Antiviral Activity against the Hepatitis C Virus through the Inhibition of STAT1 Tyrosine Phosphorylation. J Viral Hepat, 2013, 20(9):612–21., DOI 10.1111/jvh.12083 Garzon-Muvdi T, Bailey DD, Pernik MN, Pan E. Basis for Immunotherapy for Treatment of Meningiomas. Front Neurol, 2020, 11:945., DOI 10.3389/fneur. 2020.00945 Proctor DT, Patel Z, Lama S, Resch L, van Marle G, Sutherland GR. Identification of PD-L2, B7-H3 and CTLA-4 Immune Checkpoint Proteins in Genetic Subtypes of Meningioma. Oncoimmunology, 2019, 8(1):e1512943., DOI 10.1080/2162402X.2018.1512943 Doi T, Ishikawa T, Okayama T, Oka K, Mizushima K, Yasuda T, et al. The JAK/STAT Pathway Is Involved in the Upregulation of PD-L1 Expression in Pancreatic Cancer Cell Lines. Oncol Rep, 2017, 37(3):1545–54., DOI 10.3892/or. 2017.5399 Kang FB, Wang L, Jia HC, Li D, Li HJ, Zhang YG, et al. B7-H3 Promotes Aggression and Invasion of Hepatocellular Carcinoma by Targeting Epithelial- To-Mesenchymal Transition via JAK2/STAT3/Slug Signaling Pathway. Cancer Cell Int, 2015, 15:45., DOI 10.1186/s12935-015-0195-z Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PDL2 Expression. Cell Rep, 2017, 19(6):1189–201., DOI 10.1016/j.celrep.2017. 04.031 Liu L, Wang H, Cui J, Zhang Q, Zhang W, Xu W, et al. Inhibition of Protein Phosphatase 2A Sensitizes Mucoepidermoid Carcinoma to Chemotherapy via the PI3K-AKT Pathway in Response to Insulin Stimulus. Cell Physiol Biochem, 2018, 50(1):317–31., DOI 10.1159/000494008 Ho WS, Feldman MJ, Maric D, Amable L, Hall MD, Feldman GM, et al. PP2A Inhibition with LB100 Enhances Cisplatin Cytotoxicity and Overcomes Cisplatin Resistance in Medulloblastoma Cells. Oncotarget, 2016, 7(11): 12447–63., DOI 10.18632/oncotarget.6970 Hao S, Song H, Zhang W, Seldomridge A, Jung J, Giles AJ, et al. Protein Phosphatase 2A Inhibition Enhances Radiation Sensitivity and Reduces Tumor Growth in Chordoma. Neuro Oncol, 2018, 20(6):799–809., DOI 10. 1093/neuonc/nox241 Gordon IK, Lu J, Graves CA, Huntoon K, Frerich JM, Hanson RH, et al. Protein Phosphatase 2A Inhibition with LB100 Enhances Radiation-Induced Mitotic Catastrophe and Tumor Growth Delay in Glioblastoma. Mol Cancer Ther, 2015, 14(7):1540–7., DOI 10.1158/1535-7163.MCT-14-0614 Hu C, Yu M, Ren Y, Li K, Maggio DM, Mei C, et al. PP2A Inhibition from LB100 Therapy Enhances Daunorubicin Cytotoxicity in Secondary Acute Myeloid Leukemia via miR-181b-1 Upregulation. Sci Rep, 2017, 7(1):2894., DOI 10.1038/s41598-017-03058-4 Ho WS, Sizdahkhani S, Hao S, Song H, Seldomridge A, Tandle A, et al. LB-100, a Novel Protein Phosphatase 2A, PP2A, Inhibitor, Sensitizes Malignant Meningioma Cells to the Therapeutic Effects of Radiation. Cancer Lett, 2018, 415:217–26., DOI 10.1016/j.canlet.2017.11.035 Mirzapoiazova T, Xiao G, Mambetsariev B, Nasser MW, Miaou E, Singhal SS, et al. Protein Phosphatase 2A as a Therapeutic Target in Small Cell Lung Cancer. Mol Cancer Ther, 2021, 20(10):1820–35., DOI 10.1158/1535-7163. MCT-21-0013 Cui J, Wang H, Medina R, Zhang Q, Xu C, Indig IH, et al. Inhibition of PP2A with LB-100 Enhances Efficacy of CAR-T Cell Therapy against Glioblastoma. Cancers, Basel,, 2020, 12(1):E139., DOI 10.3390/cancers12010139 Giles AJ,Hao S, Padget M, Song H, ZhangW, Lynes J, et al. Efficient ADCC Killing of Meningioma by Avelumab and a High-Affinity Natural Killer Cell Line, haNK. JCI Insight, 2019, 4(20):130688., DOI 10.1172/jci.insight. 130688 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610572 EP 1610579 DI 10.3389/pore.2022.1610572 PG 8 ER PT J AU Kulka, J Cserni, G AF Kulka, Janina Cserni, Gabor TI Editorial: Guidelines From the Central-Eastern European Professional Consensus Statement on Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Editorial DE breast cancer; imaging; pathology; surgery; radiotherapy; systemic treatment; follow-up and rehabilitation; psycho-oncology ID breast cancer; imaging; pathology; surgery; radiotherapy; systemic treatment; follow-up and rehabilitation; psycho-oncology AB Guidelines from the Central-Eastern European Professional Consensus Statement on Breast Cancer C1 [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Cserni, Gabor] Bacs-Kiskun County Hospital, Department of PathologyKecskemet, Hungary. RP Cserni, G (reprint author), Bacs-Kiskun County Hospital, Department of Pathology, Kecskemet, Hungary. EM cserni@freemail.hu CR Kesson EM, Allardice GM, George WD, Burns HJG, Morrison DS. Effects of Multidisciplinary Team Working on Breast Cancer Survival: Retrospective, Comparative, Interventional Cohort Study of 13 722 Women. BMJ, 2012, 344: e2718., DOI 10.1136/bmj.e2718 Forrai G, Kovacs E, Ambrozay E, Barta M, Borbely K, Lengyel Z, et al. Radiology/Nuclear Medicine Group. Use of Imaging Methods in the Current Screening, Diagnostics and Treatment of Breast Cancer - Professional Guidelines. 4th Breast Cancer Consensus Conference. Magy Onkol, 2020, 64:278–99. [In Hungarian]. Cserni G, Francz M, Jaray B, Kalman E, Kovacs I, Krenacs T, et al. Pathological Diagnosis, Work-Up and Reporting of Breast Cancer. Recommendations from the 4th Breast Cancer Consensus Conference. Magy Onkol, 2020, 64: 301–28. [In Hungarian]. Lazar G, Kelemen P, Kosa C, Maraz R, Paszt A, Pavlovics G, et al. Modern Surgical Treatment of Breast Cancer. 4th Breast Cancer Consensus Conference. Magy Onkol, 2020, 64:329–46. [In Hungarian]. Polgar C, Kahan Z, Csejtei A, Gabor G, Landherr L, Mangel L, et al. 4th Hungarian Breast Cancer Consensus Conference - Radiotherapy Guidelines. Magy Onkol, 2020, 64:371–83. [In Hungarian]. Horvath Z, Boer K, Dank M, Kahan Z, Kocsis J, Kover E, et al. Systemic Treatment of Breast Cancer: Professional Guideline. Magy Onkol, 2020, 64: 348–68. [In Hungarian]. Kahan Z, Szanto I, Dudas R, Kapitany Z, Molnar M, Koncz Z, et al. Breast Cancer: Follow-Up, Rehabilitation, Psycho-Oncology. Magy Onkol, 2020, 64: 384–93. [In Hungarian]. Forrai G, Kovacs E, Ambrozay E, Barta M, Bobely K, Lengyel Z, et al. Use of Diagnostic Imaging Modalities in Modern Screening, Diagnostics and Management of Breast Tumours 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022, 28:1610382., DOI 10.3389/pore.2022.1610382 Cserni G, Francz M, Jaray B, Kalman E, Kovacs I, Krenacs T, et al. Pathological Diagnosis, Work-Up and Reporting of Breast Cancer 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022, 28:1610373., DOI 10.3389/pore.2022.1610373 Matrai Z, Kelemen P, Kosa C, Maraz R, Paszt A, Pavlovics G, et al. Modern Breast Cancer Surgery. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022, 28:1610377., DOI 10.3389/ pore.2022.1610377 Rubovszky G, Kocsis J, Boer K, Chilingirova N, Dank M, Kahan Z, et al. Systemic Treatment of Breast Cancer. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022, 28:1610383., DOI 10.3389/pore.2022.1610383 Polgar C, Kahan Z, Ivanov O, Chorvath M, Ligacova A, Csejtei A, et al. Radiotherapy of Breast Cancer – Professional Guideline. 1st Central- Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022, 28:1610378., DOI 10.3389/pore.2022. 1610378 Kahan Z, Szanto I, Dudas R, Kapitany Z, Molnar M, Koncz Z, et al. Breast Cancer: Follow-Up, Rehabilitation, Psycho-Oncology. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res, 2022, 28:1610391., DOI 10.3389/pore.2022.1610391 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610587 EP 1610589 DI 10.3389/pore.2022.1610587 PG 3 ER PT J AU Hino, Ch Pham, B Gray, A Wang, J Castillo, RD Akhtari, M Liu, Y AF Hino, Christopher Pham, Bryan Gray, L. Austin Wang, Jun Castillo, Ran Dan Akhtari, Mojtaba Liu, Yan TI Clinicopathologic Features and Treatment of CD10-Positive Mantle Cell Lymphoma: A Case Report and Review of Literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE case report; mantle cell lymphoma; CD10; CCND1; B-cell chronic lymphoproliferative disorders; immunophenotyping ID case report; mantle cell lymphoma; CD10; CCND1; B-cell chronic lymphoproliferative disorders; immunophenotyping AB Mantle cell lymphoma (MCL) is a rare and aggressive non-Hodgkin’s B cell lymphoma characterized by the translocation t(11;14) (q13;32) and overexpression of CCND1. MCL is immunophenotypically identified as CD20+, CD5+, CyclinD1+, CD43+, CD10−, BCL6−, and CD23−. It is often distinguished from B cell lymphomas of germinal center cell origin by the absence of CD10 expression. Here we report the unique clinicopathologic features of a patient with CD10+ MCL with gastrointestinal involvement and review current literature identifying this unique immunophenotype. C1 [Hino, Christopher] Loma Linda University, Medical Center, Department of Internal MedicineLoma Linda, CA, USA. [Pham, Bryan] Loma Linda University, Medical Center, Department of Internal MedicineLoma Linda, CA, USA. [Gray, L. Austin] Loma Linda University, Department of PathologyLoma Linda, CA, USA. [Wang, Jun] Loma Linda University, Department of PathologyLoma Linda, CA, USA. [Castillo, Ran Dan] Loma Linda University, Department of Oncology/HematologyLoma Linda, CA, USA. [Akhtari, Mojtaba] Loma Linda University, Department of Oncology/HematologyLoma Linda, CA, USA. [Liu, Yan] Loma Linda University, Department of PathologyLoma Linda, CA, USA. RP Liu, Y (reprint author), Loma Linda University, Department of Pathology, Loma Linda, USA. EM yanliu@llu.edu CR Jain P, Wang M. Mantle Cell Lymphoma: 2019 Update on the Diagnosis, Pathogenesis, Prognostication, and Management. Am J Hematol, 2019, 94(6): 710–25., DOI 10.1002/ajh.25487 Teras LR, DeSantis CE, Cerhan JR, Morton LM, Jemal A, Flowers CR. US Lymphoid Malignancy Statistics by World Health Organization Subtypes: 2016 US Lymphoid Malignancy Statistics by World Health Organization Subtypes. CA Cancer J Clin, 20162016, 66(6):443–59., DOI 10.3322/caac.21357 Bosch F, Lpez-Guillermo A, Campo E, Ribera JM, Conde E, et al. Mantle Cell Lymphoma: Presenting Features, Response to Therapy, and Prognostic Factors. Cancer, 1998, 82(3):567–75., DOI 10.1002/(sici)1097- 0142(19980201)82:3<567::aid-cncr20>3.0.co;2-z Campo E, Raffeld M, Jaffe ES. Mantle-cell Lymphoma. Semin Hematol, 1999, 36(2):115–27. Jares P, Colomer D, Campo E. Genetic and Molecular Pathogenesis of Mantle Cell Lymphoma: Perspectives for New Targeted Therapeutics. Nat Rev Cancer, 2007, 7(10):750–62., DOI 10.1038/nrc2230 Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, et al. The 2016 Revision of the World Health Organization Classification of Lymphoid Neoplasms. Blood, 2016, 127(20):2375–90., DOI 10.1182/blood-2016-01- 643569 Nadeu F, Martin-Garcia D, Clot G, Diaz-Navarro A, Duran-Ferrer M, Navarro A, et al. Genomic and Epigenomic Insights into the Origin, Pathogenesis, and Clinical Behavior of Mantle Cell Lymphoma Subtypes. Blood, 2020, 136(12): 1419–32., DOI 10.1182/blood.2020005289 Ghielmini M, Zucca E. How I Treat Mantle Cell Lymphoma. Blood, 2009, 114(8):1469–76., DOI 10.1182/blood-2009-02-179739 Xu J, Medeiros LJ, Saksena A, Wang M, Zhou J, Li J, et al. CD10-positive Mantle Cell Lymphoma: Clinicopathologic and Prognostic Study of 30 Cases. Oncotarget, 2018, 9(14):11441–50., DOI 10.18632/oncotarget.23571 Zanetto U, Dong H, Huang Y, Zhang K, NarbaitzM, Sapia S, et al. Mantle Cell Lymphoma with Aberrant Expression of CD10. Histopathology, 2008, 53(1): 20–9., DOI 10.1111/j.1365-2559.2008.03060.x Gao J, Peterson L, Nelson B, Goolsby C, Chen YH. Immunophenotypic Variations in Mantle Cell Lymphoma. Am J Clin Pathol, 2009, 132(5): 699–706., DOI 10.1309/AJCPV8LN5ENMZOVY Tam CS, Anderson MA, Pott C, Agarwal R, Handunnetti S, Hicks RJ, et al. Ibrutinib Plus Venetoclax for the Treatment of Mantle-Cell Lymphoma. N Engl J Med, 2018, 378(13):1211–23., DOI 10.1056/NEJMoa1715519 Herrmann A, Hoster E, Zwingers T, Brittinger G, Engelhard M, Meusers P, et al. Improvement of Overall Survival in Advanced Stage Mantle Cell Lymphoma. J Clin Oncol, 2009, 27(4):511–8., DOI 10.1200/JCO.2008.16.8435 Le Gouill S, Morschhauser F, Chiron D, Bouabdallah K, Cartron G, Casasnovas O, et al. Ibrutinib, Obinutuzumab, and Venetoclax in Relapsed and Untreated Patients with Mantle Cell Lymphoma: a Phase 1/2 Trial. Blood, 2021, 137(7): 877–87., DOI 10.1182/blood.2020008727 Hanel W, Epperla N. Emerging Therapies in Mantle Cell Lymphoma. J Hematol Oncol, 2020, 13(1):79., DOI 10.1186/s13045-020-00914-1 Maddocks K. Update on Mantle Cell Lymphoma. Blood, 2018, 132(16): 1647–56., DOI 10.1182/blood-2018-03-791392 Veloza L, Ribera-Cortada I, Campo E. Mantle Cell Lymphoma Pathology Update in the 2016 WHO Classification. Ann Lymphoma, 2019, 3:3., DOI 10. 21037/aol.2019.03.01 Aqil B, Triska G, Frater J, Hassan A, Ruzinova MB, Cashen A, et al. Immunophenotypic Variations in Mantle Cell Lymphoma and Their Impact on Clinical Behavior and Outcome. Arch Pathol Lab Med, 2018, 142(10):1268–74., DOI 10.5858/arpa.2017-0368-OA Akhter A, Mahe E, Street L, Pournazari P, Perizzolo M, Shabani-Rad MT, et al. CD10-positive Mantle Cell Lymphoma : Biologically Distinct Entity or an Aberrant Immunophenotype? Insight, through Gene Expression Profile in a Unique Case Series. J Clin Pathol, 2015, 68(10):844–8., DOI 10.1136/jclinpath- 2015-202955 Camacho FI, Garcia JF, Cigudosa JC, Mollejo M, Algara P, Ruiz-Ballesteros E, et al. Aberrant Bcl6 Protein Expression in Mantle Cell Lymphoma. Am J Surg Pathol, 2004, 28(8):1051–6., DOI 10.1097/01.pas.0000128671.92609.af Morice WG, Hodnefield JM, Kurtin PJ, Hanson CA. An Unusual Case of Leukemic Mantle Cell Lymphoma with a Blastoid Component Showing Loss of CD5 and Aberrant Expression of CD10. Am J Clin Pathol, 2004, 122(1):122–7., DOI 10.1309/UD2C-6JVP-WHXQ-Q217 Xu Y, McKenna RW, Kroft SH. Assessment of CD10 in the Diagnosis of Small B-Cell Lymphomas: A Multiparameter Flow Cytometric Study. Am J Clin Pathol, 2002, 117(2):291–300., DOI 10.1309/T88X-71U4-WC0R-2531 Dong HY, Gorczyca W, Liu Z, Tsang P, Wu CD, Cohen P, et al. B-Cell Lymphomas with Coexpression of CD5 and CD10. Am J Clin Pathol, 2003, 119(2):218–30., DOI 10.1309/u98advkuc26r2rja Bea S, Valdes-Mas R, Navarro A, Salaverria I, Martin-Garcia D, Jares P, et al. Landscape of Somatic Mutations and Clonal Evolution in Mantle Cell Lymphoma. Proc Natl Acad Sci U S A, 2013, 110(45):18250–5., DOI 10. 1073/pnas.1314608110 Argatoff LH, Connors JM, Klasa RJ, Horsman DE, Gascoyne RD. Mantle Cell Lymphoma: a Clinicopathologic Study of 80 Cases. Blood, 1997, 89(6): 2067–78., DOI 10.1182/blood.v89.6.2067 Zhang S, Jiang VC, Han G, Hao D, Lian J, Liu Y, et al. Longitudinal Single-Cell Profiling Reveals Molecular Heterogeneity and Tumor-Immune Evolution in Refractory Mantle Cell Lymphoma. Nat Commun, 2021, 12(1):2877., DOI 10. 1038/s41467-021-22872-z Kauh J, Baidas SM, Ozdemirli M, Cheson BD. Mantle Cell Lymphoma: Clinicopathologic Features and Treatments. Oncology, 2003, 17(6):879–91. Shah BD, Martin P, Sotomayor EM. Mantle Cell Lymphoma: A Clinically Heterogeneous Disease in Need of Tailored Approaches. Cancer Control, 2012, 19(3):227–35., DOI 10.1177/107327481201900307 Swerdlow SH, Williams ME. From Centrocytic to Mantle Cell Lymphoma: A Clinicopathologic and Molecular Review of 3 Decades. Hum Pathol, 2002, 33(1):7–20., DOI 10.1053/hupa.2002.30221 Thelander EF, Rosenquist R. Molecular Genetic Characterization Reveals New Subsets of Mantle Cell Lymphoma. Leuk Lymphoma, 2008, 49(6):1042–9., DOI 10.1080/10428190801947559 Basso K, Dalla-Favera R. BCL6: Master Regulator of the Germinal center Reaction and Key Oncogene in B Cell Lymphomagenesis. Adv Immunol, 2010, 105:193–210., DOI 10.1016/S0065-2776(10)05007-8 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610588 EP 1610592 DI 10.3389/pore.2022.1610588 PG 5 ER PT J TI Stat3 Tyrosine 705 and Serine 727 Phosphorylation Associate With Clinicopathological Characteristics and Distinct Tumor Cell Phenotypes in Triple-Negative Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE clinicopathological characteristics; triple-negative breast cancer; Stat3 tyrosine phosphorylation; Stat3 serine phosphorylation; tumor cell phenotypes ID clinicopathological characteristics; triple-negative breast cancer; Stat3 tyrosine phosphorylation; Stat3 serine phosphorylation; tumor cell phenotypes AB Signal transducer and activator of transcription 3 (Stat3) is responsible for many aspects of normal development and contributes to the development and progression of cancer through regulating epithelial cell identity and cancer stem cells. In breast cancer, Stat3 is associated with triple-negative breast cancers (TNBC) and its function has been related to the activation of p63, itself a marker of basal-like TNBC and a master regulator of stem cell activities. Stat3 activation is controlled by dual phosphorylation at tyrosine 705 (pTyr705) and serine 727 (pSer727), although it is unclear whether these have equivalent effects, and whether they are related or independent events. To address these issues, we investigated Stat3 phosphorylation at the two sites by immunohistochemistry in 173 patients with TNBC. Stat3 phosphorylation was assessed by automated quantitative measurements of digitized scanned images and classified into four categories based on histoscore. The results were analyzed for associations with multiple markers of tumor phenotype, proliferation, BRCA status, and clinicopathological characteristics. We show that the levels of pTyr705- and pSer727-Stat3 were independent in 34% of tumors. High pTyr705-Stat3 levels were associated with the luminal differentiation markers ERβ/AR and MUC1, whereas tumors with high levels of pSer727-Stat3 were more likely to be positive for the basal marker CK5/6, but were independent of p63 and were EGFR negative. Combined high pSer727- and low Tyr705-Stat3 phosphorylation associated with basal-like cancer. Although high Stat3 phosphorylation levels were associated with less aggressive tumor characteristics, they did not associate with improved survival, indicating that Stat3 phosphorylation is an unfavorable indicator for tumors with an otherwise good prognosis according to clinicopathological characteristics. These findings also show that pTyr705-Stat3 and pSer727-Stat3 associate with specific breast tumor phenotypes, implying that they exert distinct functional activities in breast cancer. CR GaloczovaM, Coates P, Vojtesek B. STAT3, Stem Cells, Cancer Stem Cells and P63. Cell Mol Biol Lett, 2018, 23:12., DOI 10.1186/s11658-018-0078-0 Hughes K, Watson CJ. The Multifaceted Role of STAT3 in Mammary Gland Involution and Breast Cancer. Int J Mol Sci, 2018, 19:E1695., DOI 10.3390/ ijms19061695 Resemann HK, Watson CJ, Lloyd-Lewis B. The Stat3 Paradox: a Killer and an Oncogene. Mol Cel Endocrinol, 2014, 382:603–11., DOI 10.1016/j.mce.2013. 06.029 Philp JA, Burdon TG, Watson CJ. Differential Activation of STATs 3 and 5 during Mammary Gland Development. FEBS Lett, 1996, 396:77–80., DOI 10. 1016/0014-5793(96)01069-1 Banerjee K, Resat H. Constitutive Activation of STAT3 in Breast Cancer Cells: A Review. Int J Cancer, 2016, 138:2570–8., DOI 10.1002/ijc.29923 Qin J-J, Yan L, Zhang J, Zhang W-D. STAT3 as a Potential Therapeutic Target in Triple Negative Breast Cancer: a Systematic Review. J Exp Clin Cancer Res, 2019, 38:195., DOI 10.1186/s13046-019-1206-z Pokorna Z, Vyslouzil J, Hrabal V, Vojtesek B, Coates PJ. The Foggy World(s, of P63 Isoform Regulation in normal Cells and Cancer. J Pathol, 2021, 254: 454–73., DOI 10.1002/path.5656 Fisher ML, Balinth S, Mills AA. p63-related Signaling at a Glance. J Cel Sci, 2020, 133:jcs228015., DOI 10.1242/jcs.228015 Ma J, Meng Y, Kwiatkowski DJ, Chen X, Peng H, Sun Q, et al. Mammalian Target of Rapamycin Regulates Murine and Human Cell Differentiation through STAT3/p63/Jagged/Notch cascade. J Clin Invest, 2010, 120:103–14., DOI 10.1172/JCI37964 Chu W-K, Dai P-M, Li H-L, Chen J-K. Transcriptional Activity of the DeltaNp63 Promoter Is Regulated by STAT3. J Biol Chem, 2008, 283: 7328–37., DOI 10.1074/jbc.M800183200 Aleskandarany MA, Agarwal D, Negm OH, Ball G, Elmouna A, Ashankyty I, et al. The Prognostic Significance of STAT3 in Invasive Breast Cancer: Analysis of Protein and mRNA Expressions in Large Cohorts. Breast Cancer Res Treat, 2016, 156:9–20., DOI 10.1007/s10549-016-3709-z Dolled-Filhart M, Camp RL, Kowalski DP, Smith BL, Rimm DL. Tissue Microarray Analysis of Signal Transducers and Activators of Transcription 3, Stat3, and Phospho-Stat3, Tyr705, in Node-Negative Breast Cancer Shows Nuclear Localization Is Associated with a Better Prognosis. Clin Cancer Res, 2003, 9:594–600. Li Y, Wang Y, Shi Z, Liu J, Zheng S, Yang J, et al. Clinicopathological and Prognostic Role of STAT3/p-STAT3 in Breast Cancer Patients in China: A Meta-Analysis. Sci Rep, 2019, 9:11243., DOI 10.1038/s41598-019-47556-z Liu Y, Huang J, Li W, Chen Y, Liu X, Wang J, et al. Meta-analysis of STAT3 and Phospho-STAT3 Expression and Survival of Patients with Breast Cancer. Oncotarget, 2018, 9:13060–7., DOI 10.18632/oncotarget.23962 Wu P, Wu D, Zhao L, Huang L, Shen G, Huang J, et al. Prognostic Role of STAT3 in Solid Tumors: a Systematic Review and Meta-Analysis. Oncotarget, 2016, 7:19863–83., DOI 10.18632/oncotarget.7887 Liu Z, Ge X, Gu Y, Huang Y, Liu H, Yu M, et al. Small Molecule STAT3 Inhibitor, 6Br-6a Suppresses Breast Cancer Growth In Vitro and In Vivo. Biomed Pharmacother, 2020, 121:109502., DOI 10.1016/j.biopha.2019. 109502 Levy DE, Darnell JE. Stats: Transcriptional Control and Biological Impact. Nat Rev Mol Cel Biol, 2002, 3:651–62., DOI 10.1038/nrm909 Wen Z, Zhong Z, Darnell JE. Maximal Activation of Transcription by Stat1 and Stat3 Requires Both Tyrosine and Serine Phosphorylation. Cell, 1995, 82:241–50., DOI 10.1016/0092-8674(95)90311-9 Zhang X, Blenis J, Li HC, Schindler C, Chen-Kiang S. Requirement of Serine Phosphorylation for Formation of STAT-Promoter Complexes. Science, 1995, 267:1990–4., DOI 10.1126/science.7701321 Shi X, Zhang H, Paddon H, Lee G, Cao X, Pelech S, et al. Phosphorylation of STAT3 Serine-727 by Cyclin-dependent Kinase 1 Is Critical for Nocodazole- Induced Mitotic Arrest. Biochemistry, 2006, 45:5857–67., DOI 10.1021/ bi052490j Frank DA, Mahajan S, Ritz J. B Lymphocytes from Patients with Chronic Lymphocytic Leukemia Contain Signal Transducer and Activator of Transcription, STAT, 1 and STAT3 Constitutively Phosphorylated on Serine Residues. J Clin Invest, 1997, 100:3140–8., DOI 10.1172/JCI119869 Liu H, Ma Y, Cole SM, Zander C, Chen K-H, Karras J, et al. Serine Phosphorylation of STAT3 Is Essential for Mcl-1 Expression and Macrophage Survival. Blood, 2003, 102:344–52., DOI 10.1182/blood-2002-11- 3396 Qin HR, Kim H-J, Kim J-Y, Hurt EM, Klarmann GJ, Kawasaki BT, et al. Activation of Signal Transducer and Activator of Transcription 3 through a Phosphomimetic Serine 727 Promotes Prostate Tumorigenesis Independent of Tyrosine 705 Phosphorylation. Cancer Res, 2008, 68:7736–41., DOI 10.1158/ 0008-5472.CAN-08-1125 Avalle L, Camporeale A, Morciano G, Caroccia N, Ghetti E, Orecchia V, et al. STAT3 Localizes to the ER, Acting as a Gatekeeper for ER-Mitochondrion Ca2+ Fluxes and Apoptotic Responses. Cell Death Differ, 2019, 26:932–42., DOI 10.1038/s41418-018-0171-y D’Amico S, Shi J, Martin BL, Crawford HC, Petrenko O, Reich NC, et al. STAT3 Is a Master Regulator of Epithelial Identity and KRAS-Driven Tumorigenesis. Genes Dev, 2018, 32:1175–87., DOI 10.1101/gad.311852.118 Huang G, Yan H, Ye S, Tong C, Ying Q-L. STAT3 Phosphorylation at Tyrosine 705 and Serine 727 Differentially Regulates Mouse ESC Fates. Stem Cells, 2014, 32:1149–60., DOI 10.1002/stem.1609 Coates PJ, Nenutil R, Holcakova J, Nekulova M, Podhorec J, Svoboda M, et al. p63 Isoforms in Triple-Negative Breast Cancer: ΔNp63 Associates with the Basal Phenotype whereas TAp63 Associates with Androgen Receptor, Lack of BRCA Mutation, PTEN and Improved Survival. Virchows Arch, 2018, 472: 351–9., DOI 10.1007/s00428-018-2324-2 Siersbaek R, Scabia V, Nagarajan S, Chernukhin I, Papachristou EK, Broome R, et al. IL6/STAT3 Signaling Hijacks Estrogen Receptor α Enhancers to Drive Breast Cancer Metastasis. Cancer Cell, 2020, 38:412–23., DOI 10.1016/j.ccell. 2020.06.007 Koyama Y, Sumie S, Nakano Y, Nagao T, Tokumaru S, Michinaga S, et al. Endothelin-1 Stimulates Expression of Cyclin D1 and S-phase Kinase- Associated Protein 2 by Activating the Transcription Factor STAT3 in Cultured Rat Astrocytes. J Biol Chem, 2019, 294:3920–33., DOI 10.1074/jbc. RA118.005614 Yeh Y-T, Ou-Yang F, Chen I-F, Yang S-F, Wang Y-Y, Chuang H-Y, et al. STAT3 Ser727 Phosphorylation and its Association with Negative Estrogen Receptor Status in Breast Infiltrating Ductal Carcinoma. Int J Cancer, 2006, 118:2943–7., DOI 10.1002/ijc.21771 Bankhead P, Loughrey MB, Fernandez JA, Dombrowski Y, McArt DG, Dunne PD, et al. QuPath: Open Source Software for Digital Pathology Image Analysis. Sci Rep, 2017, 7:16878., DOI 10.1038/s41598-017-17204-5 Lucijanic M. Survival Analysis in Clinical Practice: Analyze Your Own Data Using an Excel Workbook. Croat Med J, 2016, 57:77–9., DOI 10.3325/cmj.2016. 57.77 Sato T, Neilson LM, Peck AR, Liu C, Tran TH, Witkiewicz A, et al. Signal Transducer and Activator of Transcription-3 and Breast Cancer Prognosis. Am J Cancer Res, 2011, 1:347–55. Segatto I, Baldassarre G, Belletti B. STAT3 in Breast Cancer Onset and Progression: A Matter of Time and Context. Int J Mol Sci, 2018, 19:E2818., DOI 10.3390/ijms19092818 Kim S-Y, Kang JW, Song X, Kim BK, Yoo YD, Kwon YT, et al. Role of the IL-6- JAK1-STAT3-Oct-4 Pathway in the Conversion of Non-stem Cancer Cells into Cancer Stem-like Cells. Cell Signal, 2013, 25:961–9., DOI 10.1016/j.cellsig. 2013.01.007 Lehmann BD, Pietenpol JA. Identification and Use of Biomarkers in Treatment Strategies for Triple-Negative Breast Cancer Subtypes. J Pathol, 2014, 232:142–50., DOI 10.1002/path.4280 Burstein MD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SAW, et al. Comprehensive Genomic Analysis Identifies Novel Subtypes and Targets of Triple-Negative Breast Cancer. Clin Cancer Res, 2015, 21:1688–98., DOI 10.1158/1078-0432.CCR-14-0432 Ahmad R, Rajabi H, Kosugi M, Joshi MD, Alam M, Vasir B, et al. MUC1-C Oncoprotein Promotes STAT3 Activation in an Autoinductive Regulatory Loop. Sci Signal, 2011, 4:ra9., DOI 10.1126/scisignal.2001426 Larsen MJ, Kruse TA, Tan Q, Laenkholm A-V, Bak M, Lykkesfeldt AE, et al. Classifications within Molecular Subtypes Enables Identification of BRCA1/ BRCA2 Mutation Carriers by RNA Tumor Profiling. PLoS ONE, 2013, 8: e64268., DOI 10.1371/journal.pone.0064268 McIntosh MT, Koganti S, Boatwright JL, Li X, Spadaro SV, Brantly AC, et al. STAT3 Imparts BRCAness by Impairing Homologous Recombination Repair in Epstein-Barr Virus-Transformed B Lymphocytes. PLoS Pathog, 2020, 16: e1008849., DOI 10.1371/journal.ppat.1008849 Cheang MCU, Voduc D, Bajdik C, Leung S,McKinney S, Chia SK, et al. Basallike Breast Cancer Defined by Five Biomarkers Has superior Prognostic Value Than Triple-Negative Phenotype. Clin Cancer Res, 2008, 14:1368–76., DOI 10. 1158/1078-0432.CCR-07-1658 Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. Identification of Human Triple-Negative Breast Cancer Subtypes and Preclinical Models for Selection of Targeted Therapies. J Clin Invest, 2011, 121:2750–67., DOI 10.1172/JCI45014 Luond F, Tiede S, Christofori G. Breast Cancer as an Example of Tumour Heterogeneity and Tumour Cell Plasticity during Malignant Progression. Br J Cancer, 2021, 125:164–75., DOI 10.1038/s41416-021-01328-7 Pasha N, Turner NC. Understanding and Overcoming Tumor Heterogeneity in Metastatic Breast Cancer Treatment. Nat Cancer, 2021, 2:680–92., DOI 10. 1038/s43018-021-00229-1 Turner KM, Yeo SK, Holm TM, Shaughnessy E, Guan J-L. Heterogeneity within Molecular Subtypes of Breast Cancer. Am J Physiol Cel Physiol, 2021, 321:C343–C354., DOI 10.1152/ajpcell.00109.2021 Lee ATJ, Chew W, Wilding CP, Guljar N, Smith MJ, Strauss DC, et al. The Adequacy of Tissue Microarrays in the Assessment of Inter- and Intratumoural Heterogeneity of Infiltrating Lymphocyte burden in Leiomyosarcoma. Sci Rep, 2019, 9:14602., DOI 10.1038/s41598-019-50888-5 Siegmund K, Shibata D. At Least Two Well-Spaced Samples Are Needed to Genotype a Solid Tumor. BMC Cancer, 2016, 16:250., DOI 10.1186/s12885-016- 2202-8 Huynh J, Chand A, Gough D, Ernst M. Therapeutically Exploiting STAT3 Activity in Cancer - Using Tissue Repair as a Road Map. Nat Rev Cancer, 2019, 19:82–96., DOI 10.1038/s41568-018-0090-8 Liu Y, Nenutil R, Appleyard MV, Murray K, Boylan M, Thompson AM, et al. Lack of Correlation of Stem Cell Markers in Breast Cancer Stem Cells. Br J Cancer, 2014, 110:2063–71., DOI 10.1038/bjc.2014.105 Zhang R, Tu J, Liu S. Novel Molecular Regulators of Breast Cancer Stem Cell Plasticity and Heterogeneity. Semin Cancer Biol, 2022, 82:11–25., DOI 10.1016/ j.semcancer.2021.03.008 Zeng X, Liu C, Yao J, Wan H, Wan G, Li Y, et al. Breast Cancer Stem Cells, Heterogeneity, Targeting Therapies and Therapeutic Implications. Pharmacol Res, 2021, 163:105320., DOI 10.1016/j.phrs.2020.105320 Kim J, Villadsen R, Sorlie T, Fogh L, Gronlund SZ, Fridriksdottir AJ, et al. Tumor Initiating but Differentiated Luminal-like Breast Cancer Cells Are Highly Invasive in the Absence of Basal-like Activity. Proc Natl Acad Sci U S A, 2012, 109:6124–9., DOI 10.1073/pnas.1203203109 Liu Y, Nekulova M, Nenutil R, Horakova I, Appleyard MV, Murray K, et al. ΔNp63/p40 Correlates with the Location and Phenotype of Basal/ mesenchymal Cancer Stem-like Cells in Human ER+ and HER2+ Breast Cancers. J Pathol Clin Res, 2020, 6:83–93., DOI 10.1002/cjp2.149 GaloczovaM, Nenutil R, Pokorna Z, Vojtesek B, Coates PJ. TAp63 and ΔNp63, P40, in Prostate Adenocarcinomas: ΔNp63 Associates with a Basal-like Cancer Stem Cell Population but Not with Metastasis. Virchows Arch, 2021, 478:627–36., DOI 10.1007/s00428-020-02944-z Holcakova J, Nekulova M, Orzol P, Nenutil R, Podhorec J, Svoboda M, et al. ΔNp63 Activates EGFR Signaling to Induce Loss of Adhesion in Triple- Negative Basal-like Breast Cancer Cells. Breast Cancer Res Treat, 2017, 163: 475–84., DOI 10.1007/s10549-017-4216-6 Ciriello G, Gatza ML, Beck AH, Wilkerson MD, Rhie SK, Pastore A, et al. Comprehensive Molecular Portraits of Invasive Lobular Breast Cancer. Cell, 2015, 163:506–19., DOI 10.1016/j.cell.2015.09.033 Gujam FJA, McMillan DC, Edwards J. The Relationship between Total and Phosphorylated STAT1 and STAT3 Tumour Cell Expression, Components of Tumour Microenvironment and Survival in Patients with Invasive Ductal Breast Cancer. Oncotarget, 2016, 7:77607–21., DOI 10.18632/oncotarget. 12730 Nilsson L, Sanden E, Khazaei S, Tryggvadottir H, Nodin B, Jirstrom K, et al. Patient Characteristics Influence Activated Signal Transducer and Activator of Transcription 3, STAT3, Levels in Primary Breast Cancer—Impact on Prognosis. Front Oncol, 2020, 10:1278., DOI 10.3389/fonc.2020.01278 Hsieh F-C, Cheng G, Lin J. Evaluation of Potential Stat3-Regulated Genes in Human Breast Cancer. Biochem Biophys Res Commun, 2005, 335:292–9., DOI 10.1016/j.bbrc.2005.07.075 Barre B, Vigneron A, Perkins N, Roninson IB, Gamelin E, Coqueret O, et al. The STAT3 Oncogene as a Predictive Marker of Drug Resistance. Trends Mol Med, 2007, 13:4–11., DOI 10.1016/j.molmed.2006.11.001 Ma J-H, Qin L, Li X. Role of STAT3 Signaling Pathway in Breast Cancer. Cell Commun Signal, 2020, 18:33., DOI 10.1186/s12964-020-0527-z NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610592 EP 1610601 DI 10.3389/pore.2022.1610592 PG 10 ER PT J AU Jambor, K Koroknai, V Kiss, T Szasz, I Piko, P Balazs, M AF Jambor, Krisztina Koroknai, Viktoria Kiss, Timea Szasz, Istvan Piko, Peter Balazs, Margit TI Gene Expression Patterns of Osteopontin Isoforms and Integrins in Malignant Melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gene expression; melanoma progression; osteopontin; osteopontin splice variants; integrins ID gene expression; melanoma progression; osteopontin; osteopontin splice variants; integrins AB Osteopontin (OPN) is a multifunctional glycoprotein that physiologically interacts with different types of integrins. It is considered to be a possible prognostic biomarker in certain tumor types; however, various splicing isoforms exist, which have not been investigated in melanoma. We aimed to define the relative expression pattern of five OPN isoforms and clarify the prognostic significance of the splice variants in melanoma. We also aimed to investigate the expression pattern of eight integrins in the same tumors. Gene expression analyses revealed that the relative expression of OPNa, OPNb, and OPNc is significantly higher in metastatic tumors compared to primary lesions (p < 0.01), whereas the expression of OPN4 and OPN5 was low in both. The more aggressive nodular melanomas had higher expression levels compared to the superficial spreading subtype (p ≤ 0.05). The relative expression of the eight tested integrins was low, with only the expression of ITGB3 being detectable in nodular melanoma (Medianlog2 = 1.274). A positive correlation was found between Breslow thickness and the expression of OPNc variant, whereby thicker tumors (>4 mm) had significantly higher expression (p ≤ 0.05). The Breslow thickness was negatively correlated with the expression of OPN4, and similarly with ITGA2. OPNc also exhibited significant positive correlation with the presence of metastasis. Our data show that high expression of OPNa, OPNb, and especially OPNc and low expression of OPN4 and ITGA2 are associated with an advanced stage of tumor progression and poor prognosis in melanoma. C1 [Jambor, Krisztina] Debreceni Egyetem, Egeszsegtudomanyok Doktori IskolaDebrecen, Hungary. [Koroknai, Viktoria] Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai TanszekDebrecen, Hungary. [Kiss, Timea] Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai TanszekDebrecen, Hungary. [Szasz, Istvan] Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai TanszekDebrecen, Hungary. [Piko, Peter] Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai TanszekDebrecen, Hungary. [Balazs, Margit] Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai TanszekDebrecen, Hungary. RP Balazs, M (reprint author), Debreceni Egyetem, Nepegeszsegugyi Kar, Biostatisztikai es Epidemiologiai Tanszek, Debrecen, Hungary. EM balazs.margit@med.unideb.hu CR Zhao Y,Huang C. The Role of Osteopontin in the Development and Metastasis of Melanoma. Melanoma Res, 2021, 31(4):283–9., DOI 10.1097/CMR. 0000000000000753 Rangel J, Nosrati M, Torabian S, Shaikh L, Leong SP, Haqq C, et al. Osteopontin as a Molecular Prognostic Marker for Melanoma. Cancer, 2008, 112(1):144–50., DOI 10.1002/cncr.23147 Gimba ERP, Brum MCM, De Moraes GN. Full-length Osteopontin and its Splice Variants as Modulators of Chemoresistance and Radioresistance, Review). Int J Oncol, 2019, 54(2):420–30., DOI 10.3892/ijo.2018.4656 Lin J, Myers AL,Wang Z, Nancarrow DJ, Ferrer-Torres D, Handlogten A, et al. Osteopontin, OPN/SPP1, Isoforms Collectively Enhance Tumor Cell Invasion and Dissemination in Esophageal Adenocarcinoma. Oncotarget, 2015, 6(26): 22239–57., DOI 10.18632/oncotarget.4161 Yokosaki Y, Tanaka K, Higashikawa F, Yamashita K, Eboshida A. Distinct Structural Requirements for Binding of the Integrins αvβ6, αvβ3, αvβ5, α5β1 and α9β1 to Osteopontin. Matrix Biol, 2005, 24(6):418–27., DOI 10. 1016/j.matbio.2005.05.005 Inoue M, Shinohara ML. Intracellular Osteopontin, iOPN, and Immunity. Immunol Res, 2011, 49(1-3):160–72., DOI 10.1007/s12026-010-8179-5 Del Prete A, Scutera S, Sozzani S, Musso T. Role of Osteopontin in Dendritic Cell Shaping of Immune Responses. Cytokine Growth Factor Rev, 2019, 50: 19–28., DOI 10.1016/j.cytogfr.2019.05.004 Leavenworth JW, Verbinnen B, Wang Q, Shen E, Cantor H. Intracellular Osteopontin Regulates Homeostasis and Function of Natural Killer Cells. Proc Natl Acad Sci U S A, 2015, 112(2):494–9., DOI 10.1073/pnas.1423011112 Anborgh PH, Mutrie JC, Tuck AB, Chambers AF. Role of the Metastasis- Promoting Protein Osteopontin in the Tumour Microenvironment. J Cel Mol Med, 2010, 14(8):2037–44., DOI 10.1111/j.1582-4934.2010.01115.x Cho HJ, Cho HJ, Kim HS. Osteopontin: A Multifunctional Protein at the Crossroads of Inflammation, Atherosclerosis, and Vascular Calcification. Curr Atheroscler Rep, 2009, 11(3):206–13., DOI 10.1007/s11883-009-0032-8 Pang X, Gong K, Zhang X, Wu S, Cui Y, Qian BZ, et al. Osteopontin as a Multifaceted Driver of Bone Metastasis and Drug Resistance. Pharmacol Res, 2019, 144:235–44., DOI 10.1016/j.phrs.2019.04.030 Briones-Orta MA, Avendano-Vazquez SE, Aparicio-Bautista DI, Coombes JD, Weber GF, Syn WK, et al. Osteopontin Splice Variants and Polymorphisms in Cancer Progression and Prognosis. Biochim Biophys Acta Rev Cancer, 2017, 1868(1):93–108., DOI 10.1016/j.bbcan.2017.02.005 Hao C, Cui Y, Owen S, Li W, Cheng S, Jiang WG, et al. Human Osteopontin: Potential Clinical Applications in Cancer, Review). Int JMol Med, 2017, 39(6): 1327–37., DOI 10.3892/ijmm.2017.2964 Lamort AS, Giopanou I, Psallidas I, Stathopoulos GT. Osteopontin as a Link between Inflammation and Cancer: The Thorax in the Spotlight. Cells, 2019, 8(8):E815., DOI 10.3390/cells8080815 Pang H, Lu H, Song H, Meng Q, Zhao Y, Liu N, et al. Prognostic Values of Osteopontin-C, E-Cadherin and Beta-Catenin in Breast Cancer. Cancer Epidemiol, 2013, 37(6):985–92., DOI 10.1016/j.canep.2013.08.005 Sun SJ, Wu CC, Sheu GT, Chang HY, Chen MY, Lin YY, et al. Integrin β3 and CD44 Levels Determine the Effects of the OPN-A Splicing Variant on Lung Cancer Cell Growth. Oncotarget, 2016, 7(34):55572–84., DOI 10.18632/ oncotarget.10865 Han X, Wang W, He J, Jiang L, Li X. Osteopontin as a Biomarker for Osteosarcoma Therapy and Prognosis. Oncol Lett, 2019, 17(3):2592–8., DOI 10.3892/ol.2019.9905 Viana B, Gomes AVP, Gimba ERP, Ferreira LB. Osteopontin Expression in Thyroid Cancer: Deciphering EMT-Related Molecular Mechanisms. Biomedicines, 2021, 9(10):1372., DOI 10.3390/biomedicines9101372 Amilca-Seba K, Sabbah M, Larsen AK, Denis JA. Osteopontin as a Regulator of Colorectal Cancer Progression and its Clinical Applications. Cancers, Basel,, 2021, 13(15):3793., DOI 10.3390/cancers13153793 Rakosy Z, Ecsedi S, Toth R, Vizkeleti L, Hernandez-Vargas H, Lazar V, et al. Integrative Genomics Identifies Gene Signature Associated with Melanoma Ulceration. PLoS One, 2013, 8(1):e54958., DOI 10.1371/journal.pone.0054958 Kiss T, Ecsedi S, Vizkeleti L, Koroknai V, Emri G, Kovacs N, et al. The Role of Osteopontin Expression in Melanoma Progression. Tumour Biol, 2015, 36(10):7841–7., DOI 10.1007/s13277-015-3495-y Chou CF, Huang CC, Bin Dabil N, Chang PL. Assessing SPP1/Osteopontin, OPN, Splice Variants and Their Association to Nonmelanoma Skin Cancer by Absolute Quantification: Identification of OPN-5 Subvariants and Their Protein Coding Potential. Cancer Invest, 2021, 39(6-7):559–70., DOI 10.1080/ 07357907.2021.1933015 Livak KJ, Schmittgen TD. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2(-Delta Delta C(T), Method. Methods, 2001, 25(4):402–8., DOI 10.1006/meth.2001.1262 Scolyer RA, Rawson RV, Gershenwald JE, Ferguson PM, Prieto VG. Melanoma Pathology Reporting and Staging. Mod Pathol, 2020, 33(1):15–24., DOI 10. 1038/s41379-019-0402-x Patel V, Szasz I, Koroknai V, Kiss T, Balazs M. Molecular Alterations Associated with Acquired Drug Resistance during Combined Treatment with Encorafenib and Binimetinib in Melanoma Cell Lines. Cancers, 2021, 13(23):6058., DOI 10.3390/cancers13236058 Kiss T, Jambor K, Koroknai V, Szasz I, Bardos H, Mokanszki A, et al. Silencing Osteopontin Expression Inhibits Proliferation, Invasion and Induce Altered Protein Expression in Melanoma Cells. Pathol Oncol Res, 2021, 27:581395., DOI 10.3389/pore.2021.581395 Szasz I, Koroknai V, Kiss T, Vizkeleti L, Adany R, Balazs M, et al. Molecular Alterations Associated with Acquired Resistance to BRAFV600E Targeted Therapy in Melanoma Cells. Melanoma Res, 2019, 29(4):390–400., DOI 10. 1097/CMR.0000000000000588 Kijewska M, Kocyk M, Kloss M, Stepniak K, Korwek Z, Polakowska R, et al. The Embryonic Type of SPP1 Transcriptional Regulation is Re-activated in Glioblastoma. Oncotarget, 2017, 8(10):16340–55., DOI 10.18632/oncotarget. 14092 El Sharouni MA, van Diest PJ, Witkamp AJ, Sigurdsson V, van Gils CH. Subtyping Cutaneous Melanoma Matters. JNCI Cancer Spectr, 2020, 4(6): pkaa097., DOI 10.1093/jncics/pkaa097 Hao CC, Cui YX, Lane J, Jia SQ, Ji JF, Jiang WG, et al. Distinctive Prognostic Value and Cellular Functions of Osteopontin Splice Variants in Human Gastric Cancer. Cells, 2021, 10(7):1820., DOI 10.3390/cells10071820 Sivakumar S, Niranjali Devaraj S. Tertiary Structure Prediction and Identification of Druggable Pocket in the Cancer Biomarker - Osteopontin- C. J Diabetes Metab Disord, 2014, 13(1):13., DOI 10.1186/2251-6581-13-13 He B, MirzaM,Weber GF. An Osteopontin Splice Variant Induces Anchorage Independence in Human Breast Cancer Cells. Oncogene, 2006, 25(15): 2192–202., DOI 10.1038/sj.onc.1209248 Nishimichi N,Hayashita-Kinoh H, Chen C, Matsuda H, Sheppard D, Yokosaki Y, et al. Osteopontin Undergoes Polymerization In Vivo and Gains Chemotactic Activity for Neutrophils Mediated by Integrin Alpha9beta1. J Biol Chem, 2011, 286(13):11170–8., DOI 10.1074/jbc.M110.189258 Tilli TM, Franco VF, Robbs BK, Wanderley JL, da Silva FR, deMello KD, et al. Osteopontin-c Splicing Isoform Contributes to Ovarian Cancer Progression. Mol Cancer Res, 2011, 9(3):280–93., DOI 10.1158/1541-7786.MCR-10-0463 Silva GR, Mattos DS, Bastos ACF, Viana B, Brum MCM, Ferreira LB, et al. Osteopontin-4 and Osteopontin-5 Splice Variants are Expressed in Several Tumor Cell Lines. Mol Biol Rep, 2020, 47(10):8339–45., DOI 10.1007/s11033-020- 05867-9 Danen EH, Ten Berge PJ, Van Muijen GN, Van ’t Hof-Grootenboer AE, Brocker EB, Ruiter DJ, et al. Emergence of α5β1 Fibronectin- and αvβ3 Vitronectin-Receptor Expression in Melanocytic Tumour Progression. Histopathology, 1994, 24(3):249–56., DOI 10.1111/j.1365-2559.1994.tb00517.x Hieken TJ, Ronan SG, Farolan M, Shilkaitis AL, Das Gupta TK. Molecular Prognostic Markers in Intermediate-Thickness Cutaneous Malignant Melanoma. Cancer, 1999, 85(2):375–82., DOI 10.1002/(sici)1097- 0142(19990115)85:2<375:aid-cncr15>3.0.co;2-1 Arias-Mejias SM, Warda KY, Quattrocchi E, Alonso-Quinones H, Sominidi- Damodaran S, Meves A, et al. The Role of Integrins in Melanoma: A Review. Int J Dermatol, 2020, 59(5):525–34., DOI 10.1111/ijd.14850 Huang R, Rofstad EK. Integrins as Therapeutic Targets in the Organ-specific Metastasis of Human Malignant Melanoma. J Exp Clin Cancer Res, 2018, 37(1):92., DOI 10.1186/s13046-018-0763-x Vizkeleti L, Kiss T, Koroknai V, Ecsedi S, Papp O, Szasz I, et al. Altered Integrin Expression Patterns Shown by Microarray in Human Cutaneous Melanoma. Melanoma Res, 2017, 27(3):180–8., DOI 10.1097/Cmr. 0000000000000322 Martin TA, Jiang WG. Evaluation of the Expression of Stem Cell Markers in Human Breast Cancer Reveals a Correlation with Clinical Progression and Metastatic Disease in Ductal Carcinoma. Oncol Rep, 2014, 31(1):262–72., DOI 10.3892/or.2013.2813 Ramirez NE, Zhang Z, Madamanchi A, Boyd KL, O’Rear LD, Nashabi A, et al. The αβ Integrin is a Metastasis Suppressor in Mouse Models and Human Cancer. J Clin Invest, 2011, 121(1):226–37., DOI 10.1172/ JCI42328 Madamanchi A, Santoro SA, Zutter MM. α2β1 Integrin. Adv Exp Med Biol, 2014, 819:41–60., DOI 10.1007/978-94-017-9153-3_3 Bartolome RA, Torres S, Isern de Val S, Escudero-Paniagua B, Calvino E, Teixido J, et al. VE-Cadherin RGD Motifs Promote Metastasis and Constitute a Potential Therapeutic Target in Melanoma and Breast Cancers. Oncotarget, 2017, 8(1):215–27., DOI 10.18632/ oncotarget.13832 Yoshimura K, Meckel KF, Laird LS, Chia CY, Park JJ, Olino KL, et al. Integrin Alpha2 Mediates Selective Metastasis to the Liver. Cancer Res, 2009, 69(18): 7320–8., DOI 10.1158/0008-5472.CAN-09-0315 Naci D, Vuori K, Aoudjit F. Alpha2beta1 Integrin in Cancer Development and Chemoresistance. Semin Cancer Biol, 2015, 35:145–53., DOI 10.1016/j. semcancer.2015.08.004 Di Paola J, Jugessur A, Goldman T, Reiland J, Tallman D, Sayago C, et al. Platelet Glycoprotein I(b)alpha and Integrin Alpha2 Beta1 Polymorphisms: Gene Frequencies and Linkage Disequilibrium in a Population Diversity Panel. J Thromb Haemost, 2005, 3(7):1511–21., DOI 10.1111/j.1538-7836.2005. 01273.x Cheli Y, Williams SA, Ballotti R, Nugent DJ, Kunicki TJ. Enhanced Binding of poly(ADP-Ribose)polymerase-1 and Ku80/70 to the ITGA2 Promoter via an Extended Cytosine-Adenosine Repeat. PLoS One, 2010, 5(1):e8743., DOI 10. 1371/journal.pone.0008743 Adorno-Cruz V, Liu H. Regulation and Functions of Integrin α2 in Cell Adhesion and Disease. Genes Dis, 2019, 6(1):16–24., DOI 10.1016/j.gendis.2018. 12.003 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610608 EP 1610619 DI 10.3389/pore.2022.1610608 PG 12 ER PT J AU Antal, I Papai, Zs Szendroi, M Perlaky, T Dezso, K Lippai, Z Sapi, Z AF Antal, Imre Papai, Zsuzsanna Szendroi, Miklos Perlaky, Tamas Dezso, Katalin Lippai, Zoltan Sapi, Zoltan TI The Activation of PDGFRβ on Mononuclear Stromal/Tumor Cells in Giant Cell Tumor of Bone After Denosumab Treatment. An Immunohistochemical Study of Five Cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE giant cell tumor of bone; denosumab; PDGFRβ; combination therapy; sunitinib ID giant cell tumor of bone; denosumab; PDGFRβ; combination therapy; sunitinib AB Due to the relatively high recurrence rate and the destructive nature of the tumor, the treatment of giant cell tumor is still a challenge. Denosumab appeared to be a promising candidate as a therapeutic drug. However, several studies have reported that tumors can recur during/after treatment with denosumab. Based on activated receptor tyrosine kinase signaling pattern of the stromal/tumor cells, a combination treatment with denosumab and sunitinib has recently been proposed to inhibit recurrences. This prompted us to investigate the PDGFRβ expression of five denosumab treated cases using both primary and recurrent tumors during and after denosumab treatment. In addition, to recognise morphological changes, immunohistochemical analysis of H3F3A and PDGFRβ was also performed. As an effect of denosumab treatment, the permanent absence of giant cells associated with severe to mild fibrosis was the most consistent morphological change, but H3F3A positive stromal/tumor cells were observed in all cases. Furthermore, an increased immunopositivity of PDGFRβ in stromal/tumor cells was evident in all recurrent cases during denosumab treatment. Upon tumor recurrence (after the discontinuation of denosumab treatment) the intensity of PDGFRβ immunostaining in stromal/tumor cells was restored/decreased. Our results confirm (for the first time) the activation of PDGFRβ on mononuclear stromal/tumor cells at protein level as an effect of denosumab treatment, which has so far only been demonstrated by phosphoprotein array analysis (protein lysates). The decreased PDGFRβ activity after the discontinuation of denosumab treatmeant and the increased PDGFRβ activity during denosumab treatment underlines the need for denosumab and sunitinib combination therapy. C1 [Antal, Imre] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Papai, Zsuzsanna] Honved Korhaz, Onkologiai OsztalyBudapest, Hungary. [Szendroi, Miklos] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Perlaky, Tamas] Semmelweis University, Department of OrthopedicsBudapest, Hungary. [Dezso, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Lippai, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Sapi, Z (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM sapi.zoltan.dr@gmail.com CR WHO. Soft Tissue and Bone Tumours. Argonay: Naturaprint, 2020). Moskovszky L, Szuhai K, Krenacs T, Hogendoorn PC, Szendroi M, Benassi MS, et al. Genomic Instability in Giant Cell Tumor of Bone. A Study of 52 Cases Using DNA Ploidy, Relocalization FISH, and Array-CGH Analysis. Genes Chromosomes Cancer, 2009, 48(6):468–79., DOI 10.1002/gcc.20656 Amary F, Berisha F, Ye H, Gupta M, Gutteridge A, Baumhoer D, et al. H3F3A, Histone 3.3, G34W Immunohistochemistry: A Reliable Marker Defining Benign and Malignant Giant Cell Tumor of Bone. Am J Surg Pathol, 2017, 41(8):1059–68., DOI 10.1097/PAS.0000000000000859 van der Heijden L, Dijkstra PDS, Blay JY, Gelderblom H. Giant Cell Tumour of Bone in the Denosumab Era. Eur J Cancer, 2017, 77:75–83., DOI 10.1016/j.ejca. 2017.02.021 Balke M, Schremper L, Gebert C, Ahrens H, Streitbuerger A, Koehler G, et al. Giant Cell Tumor of Bone: Treatment and Outcome of 214 Cases. J Cancer Res Clin Oncol, 2008, 134(9):969–78., DOI 10.1007/s00432-008-0370-x Arbeitsgemeinschaft K, Becker WT, Dohle J, Bernd L, Braun A, Cserhati M, et al. Local Recurrence of Giant Cell Tumor of Bone after Intralesional Treatment with and without Adjuvant Therapy. J Bone Jt Surg Am, 2008, 90(5):1060–7., DOI 10.2106/JBJS.D.02771 Thomas D, Henshaw R, Skubitz K, Chawla S, Staddon A, Blay JY, et al. Denosumab in Patients with Giant-Cell Tumour of Bone: an Open-Label, Phase 2 Study. Lancet Oncol, 2010, 11(3):275–80., DOI 10.1016/S1470-2045(10, 70010-3 Jamshidi K, Gharehdaghi M, Hajialiloo SS, Mirkazemi M, Ghaffarzadehgan K, Izanloo A. Denosumab in Patients with Giant Cell Tumor and its Recurrence: A Systematic Review. Arch Bone Jt Surg, 2018, 6(4):260–8. Chen X, Li H, Zhu S, Wang Y, Qian W. Pre-operative Denosumab Is Associated with Higher Risk of Local Recurrence in Giant Cell Tumor of Bone: a Systematic Review and Meta-Analysis. BMC Musculoskelet Disord, 2020, 21(1):256., DOI 10.1186/s12891-020-03294-2 Lipplaa A, Dijkstra S, Gelderblom H. Challenges of Denosumab in Giant Cell Tumor of Bone, and Other Giant Cell-Rich Tumors of Bone. Curr Opin Oncol, 2019, 31(4):329–35., DOI 10.1097/CCO.0000000000000529 Hasenfratz M, Mellert K, Marienfeld R, von Baer A, Schultheiss M, Roitman PD, et al. Profiling of Three H3F3A-Mutated and Denosumab-Treated Giant Cell Tumors of Bone Points to Diverging Pathways during Progression and Malignant Transformation. Sci Rep, 2021, 11(1):5709., DOI 10.1038/s41598- 021-85319-x Mahdal M, Neradil J, Mudry P, Paukovcekova S, Staniczkova Zambo I, Urban J, et al. New Target for Precision Medicine Treatment of Giant-Cell Tumor of Bone: Sunitinib Is Effective in the Treatment of Neoplastic Stromal Cells with Activated PDGFRβ Signaling.. Cancers, Basel,, 2021, 13(14):3543., DOI 10. 3390/cancers13143543 Roitman PD, Jauk F, Farfalli GL, Albergo JI, Aponte-Tinao LA. Denosumabtreated Giant Cell Tumor of Bone. Its Histologic Spectrum and Potential Diagnostic Pitfalls. Hum Pathol, 2017, 63:89–97., DOI 10.1016/j.humpath.2017. 02.008 Szendroi M. Giant-cell Tumour of Bone. The J Bone Jt Surg Br volume, 2004, 86(1):5–12., DOI 10.1302/0301-620x.86b1.14053 Forsyth RG, Krenacs T, Athanasou N, Hogendoorn PCW. Cell Biology of Giant Cell Tumour of Bone: Crosstalk between M/wt Nucleosome H3.3, Telomeres and Osteoclastogenesis. Cancers, Basel,, 2021, 13(20):5119., DOI 10.3390/cancers13205119 Athanasou NABM, Forsyth R, Reid RP, Sapi Z. Giant Cell Tumor of Bone. In: Fletcher CDBJ, Hogendoorn PC, editors. WHO Classification of Tumours of Soft Tissue and Bone. 4th ed.. Lyon: IARC Press, 2013). p. 321–4. Roux S, Amazit L, Meduri G, Guiochon-Mantel A, Milgrom E, Mariette X. RANK, Receptor Activator of Nuclear Factor Kappa B, and RANK Ligand Are Expressed in Giant Cell Tumors of Bone. Am J Clin Pathol, 2002, 117(2): 210–6., DOI 10.1309/BPET-F2PE-P2BD-J3P3 Atkins GJ, Kostakis P, Vincent C, Farrugia AN, Houchins JP, Findlay DM, et al. RANK Expression as a Cell Surface Marker of Human Osteoclast Precursors in Peripheral Blood, Bone Marrow, and Giant Cell Tumors of Bone. J Bone Miner Res, 2006, 21(9):1339–49., DOI 10.1359/jbmr.060604 Behjati S, Tarpey PS, Presneau N, Scheipl S, Pillay N, Van Loo P, et al. Distinct H3F3A and H3F3B Driver Mutations Define Chondroblastoma and Giant Cell Tumor of Bone. Nat Genet, 2013, 45(12):1479–82., DOI 10.1038/ng.2814 Cottone L, Ligammari L, Knowles HJ, Lee HM, Henderson S, Bianco S, et al. Aberrant Paracrine Signalling Underlies the Mutant Histone-Driven Giant Cell Tumour of Bone. J Pathol, 2021, 255:S16. Matcuk GR, Jr, Patel DB, Schein AJ, White EA, Menendez LR. Giant Cell Tumor: Rapid Recurrence after Cessation of Long-Term Denosumab Therapy. Skeletal Radiol, 2015, 44(7):1027–31., DOI 10.1007/s00256-015-2117-5 Traub F, Singh J, Dickson BC, Leung S, Mohankumar R, Blackstein ME, et al. Efficacy of Denosumab in Joint Preservation for Patients with Giant Cell Tumour of the Bone. Eur J Cancer, 2016, 59:1–12., DOI 10.1016/j.ejca.2016. 01.006 Palmerini E, Seeger LL, Gambarotti M, Righi A, Reichardt P, Bukata S, et al. Malignancy in Giant Cell Tumor of Bone: Analysis of an Open-Label Phase 2 Study of Denosumab. BMC Cancer, 2021, 21(1):89., DOI 10.1186/s12885-020- 07739-8 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610633 EP 1610640 DI 10.3389/pore.2022.1610633 PG 8 ER PT J AU Bang, S Jee, S Son, H Cha, H Sim, J Kim, Y Park, H Myung, J Kim, H Paik, S AF Bang, Seongsik Jee, Seungyun Son, Hwangkyu Cha, Hyebin Sim, Jongmin Kim, Yeseul Park, Hosub Myung, Jaekyung Kim, Hyunsung Paik, Seungsam TI Clinicopathological Implications of ASAP1 Expression in Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; prognosis; hepatocellular carcinoma; ArfGAP with SH3 domain ankyrin repeat and PH domain 1; ASAP1 ID immunohistochemistry; prognosis; hepatocellular carcinoma; ArfGAP with SH3 domain ankyrin repeat and PH domain 1; ASAP1 AB Background: The expression of ArfGAP with SH3 domain ankyrin repeat and PH domain 1 (ASAP1) is increased in various types of cancer, showing potential as a prognostic marker. The clinicopathological implications of ASAP1 expression in patients with hepatocellular carcinoma (HCC) remain unclear. We thus investigated the clinicopathological significance and prognostic effect of ASAP1 expression in HCC patients. Materials and Methods: ASAP1 expression was assessed in 149 HCC tissue samples using immunohistochemistry (IHC). The associations between ASAP1 expression and clinicopathological characteristics were analyzed. The prognostic effect of ASAP1 expression in patients with HCC was evaluated based on survival analyses and confirmed using a web-based tool. Results: ASAP1 expression was observed in the cytoplasm of tumor cells. High ASAP1 expression was observed in 89 (59.7%) of 149 cases. High ASAP1 expression was significantly associated with male patients (p = 0.018), higher histological grade (p = 0.013), vessel invasion (p = 0.021), and higher stage (p = 0.020). High ASAP1 expression was associated with shorter overall survival (OS; p = 0.041) and recurrence-free survival (RFS; p = 0.008) based on Kaplan-Meier survival analyses. Web-based analysis using Kaplan-Meier (KM) plotter showed high mRNA ASAP1 expression to be associated with short OS (p = 0.001). Conclusion: High ASAP1 expression was associated with aggressive clinicopathological characteristics and poor clinical outcomes in patients with HCC. ASAP1 can be considered a prognostic biomarker in HCC patients. C1 [Bang, Seongsik] Seoul Hospital, Hanyang University College of Medicine, Department of PathologySeoul, South Korea. [Jee, Seungyun] Seoul Hospital, Hanyang University College of Medicine, Department of PathologySeoul, South Korea. [Son, Hwangkyu] Seoul Hospital, Hanyang University College of Medicine, Department of PathologySeoul, South Korea. [Cha, Hyebin] Seoul Hospital, Hanyang University College of Medicine, Department of PathologySeoul, South Korea. [Sim, Jongmin] Korea University College of Medicine, Anam Hospital, Department of PathologySeoul, South Korea. [Kim, Yeseul] Korea University College of Medicine, Anam Hospital, Department of PathologySeoul, South Korea. [Park, Hosub] Seoul Hospital, Hanyang University College of Medicine, Department of PathologySeoul, South Korea. [Myung, Jaekyung] Seoul Hospital, Hanyang University College of Medicine, Department of PathologySeoul, South Korea. [Kim, Hyunsung] Seoul Hospital, Hanyang University College of Medicine, Department of PathologySeoul, South Korea. [Paik, Seungsam] Seoul Hospital, Hanyang University College of Medicine, Department of PathologySeoul, South Korea. RP Paik, S (reprint author), Seoul Hospital, Hanyang University College of Medicine, Department of Pathology, Seoul, South Korea. EM sspaik@hanyang.ac.kr CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 McGlynn KA, Petrick JL, El-Serag HB. Epidemiology of Hepatocellular Carcinoma. Hepatology, 2021, 73(1):4–13., DOI 10.1002/hep.31288 Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular Carcinoma. Nat Rev Dis Primers, 2021, 7(1):6., DOI 10.1038/ s41572-020-00240-3 Koulouris A, Tsagkaris C, Spyrou V, Pappa E, Troullinou A, Nikolaou M. Hepatocellular Carcinoma: An Overview of the Changing Landscape of Treatment Options. J Hepatocell Carcinoma, 2021, 8:387–401., DOI 10.2147/ JHC.S300182 Brown MT, Andrade J, Radhakrishna H, Donaldson JG, Cooper JA, Randazzo PA. ASAP1, a Phospholipid-dependent Arf GTPase-Activating Protein that Associates with and Is Phosphorylated by Src. Mol Cel Biol, 1998, 18(12): 7038–51., DOI 10.1128/mcb.18.12.7038 Randazzo PA, Andrade J, Miura K, Brown MT, Long YQ, Stauffer S, et al. The Arf GTPase-Activating Protein ASAP1 Regulates the Actin Cytoskeleton. Proc Natl Acad Sci U S A, 2000, 97(8):4011–6., DOI 10. 1073/pnas.070552297 Donaldson JG, Jackson CL. ARF Family G Proteins and Their Regulators: Roles in Membrane Transport, Development and Disease. Nat Rev Mol Cel Biol, 2011, 12(6):362–75., DOI 10.1038/nrm3117 Onodera Y, Hashimoto S, Hashimoto A, Morishige M, Mazaki Y, Yamada A, et al. Expression of AMAP1, an ArfGAP, Provides Novel Targets to Inhibit Breast Cancer Invasive Activities. EMBO J, 2005, 24(5):963–73., DOI 10.1038/sj. emboj.7600588 Ehlers JP, Worley L, Onken MD, Harbour JW. DDEF1 Is Located in an Amplified Region of Chromosome 8q and Is Overexpressed in Uveal Melanoma. Clin Cancer Res, 2005, 11(10):3609–13., DOI 10.1158/1078- 0432.CCR-04-1941 Lin D, Watahiki A, Bayani J, Zhang F, Liu L, Ling V, et al. ASAP1, a Gene at 8q24, Is Associated with Prostate Cancer Metastasis. Cancer Res, 2008, 68(11): 4352–9., DOI 10.1158/0008-5472.CAN-07-5237 Sabe H, Hashimoto S, Morishige M, Ogawa E, Hashimoto A, Nam JM, et al. The EGFR-Gep100-Arf6-AMAP1 Signaling Pathway Specific to Breast Cancer Invasion andMetastasis. Traffic, 2009, 10(8):982–93., DOI 10.1111/j.1600-0854. 2009.00917.x Muller T, Stein U, Poletti A, Garzia L, Rothley M, Plaumann D, et al. ASAP1 Promotes Tumor Cell Motility and Invasiveness, Stimulates Metastasis Formation In Vivo, and Correlates with Poor Survival in Colorectal Cancer Patients. Oncogene, 2010, 29(16):2393–403., DOI 10.1038/ onc.2010.6 Li M, Tian L, Yao H, Lu J, Ge J, Guo Y, et al. ASAP1 Mediates the Invasive Phenotype of Human Laryngeal Squamous Cell Carcinoma to Affect Survival Prognosis. Oncol Rep, 2014, 31(6):2676–82., DOI 10.3892/or.2014.3150 Sato H, Hatanaka KC, Hatanaka Y, Hatakeyama H, Hashimoto A, Matsuno Y, et al. High Level Expression of AMAP1 Protein Correlates with Poor Prognosis and Survival after Surgery of Head and Neck Squamous Cell Carcinoma Patients. Cell Commun Signal, 2014, 12:17., DOI 10.1186/1478-811X-12-17 Hou T, Yang C, Tong C, Zhang H, Xiao J, Li J. Overexpression of ASAP1 Is Associated with Poor Prognosis in Epithelial Ovarian Cancer. Int J Clin Exp Pathol, 2014, 7(1):280–7. Hashimoto S, Mikami S, Sugino H, Yoshikawa A, Hashimoto A, Onodera Y, et al. Lysophosphatidic Acid Activates Arf6 to Promote the Mesenchymal Malignancy of Renal Cancer. Nat Commun, 2016, 7:10656., DOI 10.1038/ ncomms10656 Hashimoto S, Furukawa S, Hashimoto A, Tsutaho A, Fukao A, Sakamura Y, et al. ARF6 and AMAP1 Are Major Targets of KRAS and TP53 Mutations to Promote Invasion, PD-L1 Dynamics, and Immune Evasion of Pancreatic Cancer. Proc Natl Acad Sci U S A, 2019, 116(35):17450–9., DOI 10.1073/ pnas.1901765116 Luo Q, Zhang S, Zhang D, Yuan F, Chen X, Yang S. Expression of ASAP1 and FAK in Gastric Cancer and its Clinicopathological Significance. Oncol Lett, 2020, 20(1):974–80., DOI 10.3892/ol.2020.11612 Li J, Tian S, Guo Y, Dong W. Oncological Effects and Prognostic Value of AMAP1 in Gastric Cancer. Front Genet, 2021, 12:675100., DOI 10.3389/fgene. 2021.675100 He J, McLaughlin RP, van der Beek L, Canisius S, Wessels L, Smid M, et al. Integrative Analysis of Genomic Amplification-dependent Expression and Loss-Of-Function Screen Identifies ASAP1 as a Driver Gene in Triple-Negative Breast Cancer Progression. Oncogene, 2020, 39(20):4118–31., DOI 10.1038/s41388-020-1279-3 Wang ZG, Zheng H, Gao W, Han J, Cao JZ, Yang Y, et al. eIF5B Increases ASAP1 Expression to Promote HCC Proliferation and Invasion. Oncotarget, 2016, 7(38):62327–39., DOI 10.18632/oncotarget.11469 Cancer Genome Atlas Research Network. Electronic Address Wbe, Cancer Genome Atlas Research N. Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma. Cell, 2017, 169(7):1327–41 e23. Lanczky A, Gyorffy B. Web-Based Survival Analysis Tool Tailored for Medical Research, KMplot): Development and Implementation. J Med Internet Res, 2021, 23(7):e27633., DOI 10.2196/27633 Casalou C, Ferreira A, Barral DC. The Role of ARF Family Proteins and Their Regulators and Effectors in Cancer Progression: A Therapeutic Perspective. Front Cel Dev Biol, 2020, 8:217., DOI 10.3389/fcell.2020.00217 Kinoshita R, Nam JM, Ito YM, Hatanaka KC, Hashimoto A, Handa H, et al. Co-overexpression of GEP100 and AMAP1 Proteins Correlates with Rapid Local Recurrence after Breast Conservative Therapy. PLoS One, 2013, 8(10): e76791., DOI 10.1371/journal.pone.0076791 Tsutaho A, Hashimoto A, Hashimoto S, Hata S, Kachi S, Hirano S, et al. High Expression of AMAP1, an ARF6 Effector, Is Associated with Elevated Levels of PD-L1 and Fibrosis of Pancreatic Cancer. Cel Commun Signal, 2020, 18(1): 101., DOI 10.1186/s12964-020-00608-8 Horikawa M, Sabe H, Onodera Y. Dual Roles of AMAP1 in the Transcriptional Regulation and Intracellular Trafficking of Carbonic Anhydrase IX. Transl Oncol, 2022, 15(1):101258., DOI 10.1016/j.tranon. 2021.101258 Damgaci S, Ibrahim-Hashim A, Enriquez-Navas PM, Pilon-Thomas S, Guvenis A, Gillies RJ. Hypoxia and Acidosis: Immune Suppressors and Therapeutic Targets. Immunology, 2018, 154(3):354–62., DOI 10.1111/imm. 12917 Kuchuk O, Tuccitto A, Citterio D, Huber V, Camisaschi C, Milione M, et al. pH Regulators to Target the Tumor Immune Microenvironment in Human Hepatocellular Carcinoma. Oncoimmunology, 2018, 7(7):e1445452., DOI 10.1080/ 2162402X.2018.1445452 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610635 EP 1610642 DI 10.3389/pore.2022.1610635 PG 8 ER PT J AU Zhu, Zz Zhang, G Liu, J AF Zhu, Zhong-zhong Zhang, Guanglin Liu, Jianping TI Establishment of a Novel Prognostic Prediction Model for Gastric Cancer Based on Necroptosis-Related Genes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; prognosis; gastric cancer; treatment response; necroptosis ID biomarker; prognosis; gastric cancer; treatment response; necroptosis AB Background: Necroptosis plays a crucial role in the progression of multiple types of cancer. However, the role of necroptosis in gastric cancer (GC) remains unclear. The aim of this study is to establish a necroptosis-related prediction model, which could provide information for treatment monitoring. Methods: The TCGA-STAD cohort was employed to establish a prognostic prediction signature and the GEO dataset was employed for external validation. The correlation between the risk score and the immune landscape, tumor mutational burden (TMB), microsatellite instability (MSI), as well as therapeutic responses of different therapies were analyzed. Results: We constructed a prognostic model based on necroptosis-associated genes (NAGs), and its favorable predictive ability was confirmed in an external cohort. The risk score was confirmed as an independent determinant, and a nomogram was further established for prognosis. A high score implies higher tumor immune microenvironment (TIME) scores and more significant TIME cell infiltration. High-risk patients presented with lower TMB, and low-TMB patients had worse overall survival (OS). Meanwhile, Low-risk scores are characterized by MSI-high (MSI-H), lower Tumor Immune Dysfunction and Exclusion (TIDE) score, and higher immunogenicity in immunophenoscore (IPS) analysis. Conclusion: The developed NAG score provides a novel and effective method for predicting the outcome of GC as well as potential targets for further research. C1 [Zhu, Zhong-zhong] Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi Central Hospital, Department of Gastroenteroanrectal SurgeryHuangshi, China. [Zhang, Guanglin] Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi Central Hospital (Pu Ai Hospital), Department of Abdominal and Pelvic Medical Oncology II WardHuangshi, China. [Liu, Jianping] Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi Central Hospital (Pu Ai Hospital), Department of Abdominal and Pelvic Medical Oncology II WardHuangshi, China. RP Liu, J (reprint author), Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi Central Hospital (Pu Ai Hospital), Department of Abdominal and Pelvic Medical Oncology II Ward, Huangshi, China. EM jianpl@126.com CR Sexton RE, Al Hallak MN, Diab M, Azmi AS. Gastric Cancer: a Comprehensive Review of Current and Future Treatment Strategies. Cancer Metastasis Rev, 2020, 39:1179–203., DOI 10.1007/s10555-020-09925-3 Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric Cancer. Lancet, 2020, 396:635–48., DOI 10.1016/s0140-6736(20, 31288-5 Johnston FM, Beckman M. Updates on Management of Gastric Cancer. Curr Oncol Rep, 2019, 21:67., DOI 10.1007/s11912-019-0820-4 Rawla P, Barsouk A. Epidemiology of Gastric Cancer: Global Trends, Risk Factors and Prevention. Prz Gastroenterol, 2019, 14:26–38., DOI 10.5114/pg. 2018.80001 Frank D, Vince JE. Pyroptosis versus Necroptosis: Similarities, Differences, and Crosstalk. Cell Death Differ, 2019, 26:99–114., DOI 10.1038/s41418-018- 0212-6 Gong Y, Fan Z, Luo G, Yang C, Huang Q, Fan K, et al. The Role of Necroptosis in Cancer Biology and Therapy. Mol Cancer, 2019, 18:100., DOI 10.1186/ s12943-019-1029-8 Sprooten J, De Wijngaert P, Vanmeerbeerk I, Martin S, Vangheluwe P, Schlenner S, et al. Necroptosis in Immuno-Oncology and Cancer Immunotherapy. Cells, 2020, 9:E1823., DOI 10.3390/cells9081823 Bao JH, Li JB, Lin HS, Zhang WJ, Guo BY, Li JJ, et al. Deciphering a Novel Necroptosis-Related miRNA Signature for Predicting the Prognosis of Clear Cell Renal Carcinoma. Anal Cel Pathol, 2022, 2022:2721005., DOI 10.1155/ 2022/2721005 Huang Y, Zou Y, Xiong Q, Zhang C, Sayagues JM, Shelat VG, et al. Development of a Novel Necroptosis-Associated miRNA Risk Signature to Evaluate the Prognosis of colon Cancer Patients. Ann Transl Med, 2021, 9: 1800., DOI 10.21037/atm-21-6576 Lu Y, Luo X, Wang Q, Chen J, Zhang X, Li Y, et al. A Novel Necroptosis- Related lncRNA Signature Predicts the Prognosis of Lung Adenocarcinoma. Front Genet, 2022, 13:862741., DOI 10.3389/fgene. 2022.862741 Zheng L, Wang J, Jiang H, Dong H. A Novel Necroptosis-Related miRNA Signature for Predicting the Prognosis of Breast Cancer Metastasis. Dis Markers, 2022, 2022:3391878., DOI 10.1155/2022/3391878 Ma J, Jin Y, Gong B, Li L, Zhao Q. Pan-cancer Analysis of Necroptosis- Related Gene Signature for the Identification of Prognosis and Immune Significance. Discov Oncol, 2022, 13:17., DOI 10.1007/s12672-022- 00477-2 Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, et al. Signatures of T Cell Dysfunction and Exclusion Predict Cancer Immunotherapy Response. Nat Med, 2018, 24: 1550–8., DOI 10.1038/s41591-018-0136-1 Seeneevassen L, Bessede E, Megraud F, Lehours P, Dubus P, Varon C. Gastric Cancer: Advances in Carcinogenesis Research and New Therapeutic Strategies. Int J Mol Sci, 2021, 22:3418., DOI 10.3390/ ijms22073418 Lou S, Meng F, Yin X, Zhang Y, Han B, Xue Y. Comprehensive Characterization of RNA Processing Factors in Gastric Cancer Identifies a Prognostic Signature for Predicting Clinical Outcomes and Therapeutic Responses. Front Immunol, 2021, 12:719628., DOI 10.3389/fimmu.2021.719628 Huo J, Wu L, Zang Y. Eight-gene Prognostic Signature Associated with Hypoxia and Ferroptosis for Gastric Cancer with General Applicability. Epigenomics, 2021, 13:875–90., DOI 10.2217/epi-2020-0411 Yu S, Hu C, Cai L, Du X, Lin F, Yu Q, et al. Seven-Gene Signature Based on Glycolysis Is Closely Related to the Prognosis and Tumor Immune Infiltration of Patients With Gastric Cancer. Front Oncol, 2020, 10:1778., DOI 10.3389/fonc. 2020.01778 Moreno-Gonzalez G, Vandenabeele P, Krysko DV. Necroptosis: A Novel Cell Death Modality and its Potential Relevance for Critical Care Medicine. Am J Respir Crit Care Med, 2016, 194:415–28., DOI 10.1164/ rccm.201510-2106CI Shi H, Peng Q, Zhou X, He Y, Sun S. An Efficient Signature Based on Necroptosis-Related Genes for Prognosis of Patients With Pancreatic Cancer. Front Genet, 2022, 13:848747., DOI 10.3389/fgene.2022.848747 Chen J, Wang H, Zhou L, Liu Z, Chen H, Tan X. A Necroptosis-Related Gene Signature for Predicting Prognosis, Immune Landscape, and Drug Sensitivity in Hepatocellular Carcinoma. Cancer Med, 2022)., DOI 10. 1002/cam4.4812 Yang G, Fang J, Shi Q, Wang R, Lian M, Ma H, et al. Screening of Molecular Markers of Induced Chemotherapy in Supraglottic Laryngeal Squamouscell Carcinoma. World J Otorhinolaryngol Head Neck Surg, 2020, 6:34–40., DOI 10. 1016/j.wjorl.2019.05.001 Ewald JA, Downs TM, Cetnar JP, Ricke WA. Expression Microarray Meta-Analysis Identifies Genes Associated with Ras/MAPK and Related Pathways in Progression of Muscle-Invasive Bladder Transition Cell Carcinoma. PLoS One, 2013, 8:e55414., DOI 10.1371/journal.pone. 0055414 Zhang L, Li H, Yuan M, Li M, Zhang S. Cervical Cancer Cells-Secreted Exosomal microRNA-221-3p Promotes Invasion, Migration and Angiogenesis of Microvascular Endothelial Cells in Cervical Cancer by Down-Regulating MAPK10 Expression. Cancer Manag Res, 2019, 11:10307–19., DOI 10.2147/ cmar.S221527 Xie Y, Liu Y, Fan X, Zhang L, Li Q, Li S, et al. MicroRNA-21 Promotes Progression of Breast Cancer via Inhibition of Mitogen-Activated Protein Kinase10, MAPK10). Biosci Rep, 2019, BSR20181000., DOI 10.1042/ bsr20181000 Wu X, Li X, Xu G. Propofol Suppresses the Progression of Non-small Cell Lung Cancer via Downregulation of the miR-21-5p/MAPK10 axis. Oncol Rep, 2020, 44:487–98., DOI 10.3892/or.2020.7619 Gao Y,Wang Y,Wang X, Zhao C,Wang F, Du J, et al. miR-335-5p Suppresses Gastric Cancer Progression by Targeting MAPK10. Cancer Cel Int, 2021, 21: 71., DOI 10.1186/s12935-020-01684-z Yang KS, Xu CQ, Lv J. Identification and Validation of the Prognostic Value of Cyclic GMP-AMP Synthase-Stimulator of Interferon, cGAS-STING, Related Genes in Gastric Cancer. Bioengineered, 2021, 12:1238–50., DOI 10.1080/ 21655979.2021.1911557 van der Leun AM, Thommen DS, Schumacher TN. CD8(+, T Cell States in Human Cancer: Insights from Single-Cell Analysis. Nat Rev Cancer, 2020, 20: 218–32., DOI 10.1038/s41568-019-0235-4 Wang Y, Zhu C, Song W, Li J, Zhao G, Cao H. PD-L1 Expression and CD8(+, T Cell Infiltration Predict a Favorable Prognosis in Advanced Gastric Cancer. J Immunol Res, 2018, 2018:4180517., DOI 10.1155/2018/ 4180517 Yu PC, Long D, Liao CC, Zhang S. Association between Density of Tumor-Infiltrating Lymphocytes and Prognoses of Patients with Gastric Cancer. Medicine, Baltimore,, 2018, 97:e11387., DOI 10.1097/md. 0000000000011387 Lee JS, Won HS, Sun S, Hong JH, Ko YH. Prognostic Role of Tumor- Infiltrating Lymphocytes in Gastric Cancer: A Systematic Review and Meta-Analysis. Medicine, Baltimore,, 2018, 97:e11769., DOI 10.1097/md. 0000000000011769 Thompson ED, Zahurak M, Murphy A, Cornish T, Cuka N, Abdelfatah E, et al. Patterns of PD-L1 Expression and CD8 T Cell Infiltration in Gastric Adenocarcinomas and Associated Immune Stroma. Gut, 2017, 66:794–801., DOI 10.1136/gutjnl-2015-310839 Li K, Zhang A, Li X, Zhang H, Zhao L. Advances in Clinical Immunotherapy for Gastric Cancer. Biochim Biophys Acta Rev Cancer, 2021, 1876:188615., DOI 10.1016/j.bbcan.2021.188615 Kawazoe A, Shitara K, Boku N, Yoshikawa T, Terashima M. Current Status of Immunotherapy for Advanced Gastric Cancer. Jpn J Clin Oncol, 2021, 51: 20–7., DOI 10.1093/jjco/hyaa202 Zeng D, Wu J, Luo H, Li Y, Xiao J, Peng J, et al. Tumor Microenvironment Evaluation Promotes Precise Checkpoint Immunotherapy of Advanced Gastric Cancer. J Immunother Cancer, 2021, 9:e002467., DOI 10.1136/jitc- 2021-002467 Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor Mutational Load Predicts Survival after Immunotherapy across Multiple Cancer Types. Nat Genet, 2019, 51:202–6., DOI 10.1038/s41588-018- 0312-8 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610641 EP 1610652 DI 10.3389/pore.2022.1610641 PG 12 ER PT J AU Takacs, F Kotmayer, L Czeti, Szaloki, G Laszlo, T Mikala, G Mark, Masszi, A Farkas, P Plander, M Weisinger, J Demeter, J Fekete, S Szerafin, L Deak, MB Szaleczky, E Sulak, A Borbenyi, Z Barna, G AF Takacs, Ferenc Kotmayer, Lili Czeti, Agnes Szaloki, Gabor Laszlo, Tamas Mikala, Gabor Mark, Agnes Masszi, Andras Farkas, Peter Plander, Mark Weisinger, Julia Demeter, Judit Fekete, Sandor Szerafin, Laszlo Deak, Margit Beata Szaleczky, Erika Sulak, Adrienn Borbenyi, Zita Barna, Gabor TI Revealing a Phenotypical Appearance of Ibrutinib Resistance in Patients With Chronic Lymphocytic Leukaemia by Flow Cytometry SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE flow cytometry; targeted therapy; drug resistance; chronic lymphocytic leukaemia; ibrutinib ID flow cytometry; targeted therapy; drug resistance; chronic lymphocytic leukaemia; ibrutinib AB Background: Ibrutinib is widely known as an effective and well-tolerated therapeutical choice of the chronic lymphocytic leukaemia (CLL). However, acquired resistance may occur during the treatment, causing relapse. Early detection of ibrutinib resistance is an important issue, therefore we aimed to find phenotypic markers on CLL cells the expression of which may correlate with the appearance of ibrutinib resistance. Methods: We examined 28 patients’ peripheral blood (PB) samples (treatment naive, ibrutinib sensitive, clinically ibrutinib resistant). The surface markers’ expression (CD27, CD69, CD86, CD184, CD185) were measured by flow cytometry. Furthermore, the BTKC481S resistance mutation was assessed by digital droplet PCR. Moreover, the CLL cells’ phenotype of a patient with acquired ibrutinib resistance was observed during the ibrutinib treatment. Results: The expression of CD27 (p = 0.030) and CD86 (p = 0.031) became higher in the clinically resistant cohort than in the ibrutinib sensitive cohort. Besides, we found that high CD86 and CD27 expressions were accompanied by BTKC481S mutation. Our prospective study showed that the increase of the expression of CD27, CD69 and CD86 was noticed ahead of the clinical resistance with 3 months. Conclusion: Our study suggests that the changes of the expression of these markers could indicate ibrutinib resistance and the examination of these phenotypic changes may become a part of the patients’ follow-up in the future. C1 [Takacs, Ferenc] HCEMM-SE Molecular Oncohematology Research GroupBudapest, Hungary. [Kotmayer, Lili] HCEMM-SE Molecular Oncohematology Research GroupBudapest, Hungary. [Czeti, Agnes] HCEMM-SE Molecular Oncohematology Research GroupBudapest, Hungary. [Szaloki, Gabor] HCEMM-SE Molecular Oncohematology Research GroupBudapest, Hungary. [Laszlo, Tamas] HCEMM-SE Molecular Oncohematology Research GroupBudapest, Hungary. [Mikala, Gabor] Del-pesti Centrumkohaz, Orszagos Hematologiai es Infektologiai Intezet (DPC-OHII)Budapest, Hungary. [Mark, Agnes] HCEMM-SE Molecular Oncohematology Research GroupBudapest, Hungary. [Masszi, Andras] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Farkas, Peter] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Plander, Mark] Vas County Markusovszky Hospital, Department of HematologySzombathely, Hungary. [Weisinger, Julia] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Demeter, Judit] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Fekete, Sandor] Del-pesti Centrumkohaz, Orszagos Hematologiai es Infektologiai Intezet (DPC-OHII)Budapest, Hungary. [Szerafin, Laszlo] Josa Andras County HospitalNyiregyhaza, Hungary. [Deak, Margit Beata] National Institute of OncologyBudapest, Hungary. [Szaleczky, Erika] National Institute of OncologyBudapest, Hungary. [Sulak, Adrienn] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Borbenyi, Zita] University of Szeged, 2nd Department of Internal Medicine and Cardiology CentreSzeged, Hungary. [Barna, Gabor] HCEMM-SE Molecular Oncohematology Research GroupBudapest, Hungary. RP Barna, G (reprint author), HCEMM-SE Molecular Oncohematology Research Group, Budapest, Hungary. EM barna.gabor@med.semmelweis-univ.hu CR Zent CS, Kyasa MJ, Evans R, Schichman SA. Chronic Lymphocytic Leukemia Incidence Is Substantially Higher Than Estimated from Tumor Registry Data. Cancer, 2001, 92(5):1325–30., DOI 10.1002/1097-0142(20010901)92:5<1325:: aid-cncr1454>3.0.co;2-e Baumann T, Delgado J, Santacruz R, Martinez-Trillos A, Royo C, Navarro A, et al. Chronic Lymphocytic Leukemia in the Elderly: Clinico-Biological Features, Outcomes, and Proposal of a Prognostic Model. Haematologica, 2014, 99(10):1599–604., DOI 10.3324/haematol.2014.107326 Badar T, Burger JA, Wierda WG, O’Brien S. Ibrutinib: a Paradigm Shift in Management of CLL. Expert Rev Hematol, 2014, 7(6):705–17., DOI 10.1586/ 17474086.2014.977862 Burger JA, Tedeschi A, Barr PM, Robak T, Owen C, Ghia P, et al. Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia. N Engl J Med, 2015, 373(25):2425–37., DOI 10.1056/NEJMoa1509388 Woyach JA, Ruppert AS, Heerema NA, Zhao W, Booth AM, Ding W, et al. Ibrutinib Regimens versus Chemoimmunotherapy in Older Patients with Untreated CLL. N Engl J Med, 2018, 379(26):2517–28., DOI 10.1056/ NEJMoa1812836 Ahn IE, Underbayev C, Albitar A, Herman SE, Tian X, Maric I, et al. Clonal Evolution Leading to Ibrutinib Resistance in Chronic Lymphocytic Leukemia. Blood, 2017, 129(11):1469–79., DOI 10.1182/blood-2016-06-719294 Burger JA, Landau DA, Taylor-Weiner A, Bozic I, Zhang H, Sarosiek K, et al. Clonal Evolution in Patients with Chronic Lymphocytic Leukaemia Developing Resistance to BTK Inhibition. Nat Commun, 2016, 7:11589., DOI 10.1038/ncomms11589 Gango A, Alpar D, Galik B, Marosvari D, Kiss R, Fesus V, et al. Dissection of Subclonal Evolution by Temporal Mutation Profiling in Chronic Lymphocytic Leukemia Patients Treated with Ibrutinib. Int J Cancer, 2020, 146(1):85–93., DOI 10.1002/ijc.32502 Kiss R, Alpar D, Gango A, Nagy N, Eyupoglu E, Aczel D, et al. Spatial Clonal Evolution Leading to Ibrutinib Resistance and Disease Progression in Chronic Lymphocytic Leukemia. Haematologica, 2019, 104(1):e38–e41., DOI 10.3324/ haematol.2018.202085 Ahn IE, Farooqui MZH, Tian X, Valdez J, Sun C, Soto S, et al. Depth and Durability of Response to Ibrutinib in CLL: 5-year Follow-Up of a Phase 2 Study. Blood, 2018, 131(21):2357–66., DOI 10.1182/blood-2017-12-820910 Lew TE, Anderson MA, Lin VS, Handunnetti SM, Came NA, Blombery P, et al. Undetectable Peripheral Blood MRD Should Be the Goal of Venetoclax in CLL, but Attainment Plateaus after 24 Months. Blood Adv, 2020, 4(1):165–73., DOI 10.1182/bloodadvances.2019000864 Wang XV, Hanson CA, Tschumper RC, Lesnick CE, Braggio E, Paietta EM, et al. Measurable Residual Disease Does Not Preclude Prolonged Progressionfree Survival in CLL Treated with Ibrutinib. Blood, 2021, 138(26):2810–27., DOI 10.1182/blood.2020010146 Tissino E, BenedettiD,Herman SEM, Ten Hacken E, Ahn IE, ChaffeeKG, et al. Functional and Clinical Relevance of VLA-4, CD49d/CD29, in Ibrutinib-Treated Chronic Lymphocytic Leukemia. J Exp Med, 2018, 215(2):681–97., DOI 10.1084/jem.20171288 Damle RN, Ghiotto F, Valetto A, Albesiano E, Fais F, Yan XJ, et al. B-Cell Chronic Lymphocytic Leukemia Cells Express a Surface Membrane Phenotype of Activated, Antigen-Experienced B Lymphocytes. Blood, 2002, 99(11): 4087–93., DOI 10.1182/blood.v99.11.4087 Montraveta A, Lee-Verges E, Roldan J, Jimenez L, Cabezas S, Clot G, et al. CD69 Expression Potentially Predicts Response to Bendamustine and its Modulation by Ibrutinib or Idelalisib Enhances Cytotoxic Effect in Chronic Lymphocytic Leukemia. Oncotarget, 2016, 7(5):5507–20., DOI 10.18632/ oncotarget.6685 Chen SS, Chang BY, Chang S, Tong T, Ham S, Sherry B, et al. BTK Inhibition Results in Impaired CXCR4 Chemokine Receptor Surface Expression, Signaling and Function in Chronic Lymphocytic Leukemia. Leukemia, 2016, 30(4):833–43., DOI 10.1038/leu.2015.316 Saez de Guinoa J, Barrio L, Mellado M, Carrasco YR. CXCL13/ CXCR5 Signaling Enhances BCR-Triggered B-Cell Activation by Shaping Cell Dynamics. Blood, 2011, 118(6):1560–9., DOI 10.1182/blood-2011-01- 332106 Shen Y, Best OG, Mulligan SP, Christopherson RI. Ibrutinib and Idelalisib Block Immunophenotypic Changes Associated with the Adhesion and Activation of CLL Cells in the Tumor Microenvironment. Leuk Lymphoma, 2018, 59(8):1927–37., DOI 10.1080/10428194.2017.1403598 Takacs F, Tolnai-Kriston C, Hernadfoi M, Szabo O, Szaloki G, Szepesi A, et al. The Effect of CD86 Expression on the Proliferation and the Survival of CLL Cells. Pathol Oncol Res, 2019, 25(2):647–52., DOI 10.1007/s12253-018-0512-7 Huemer M, Rebhandl S, Zaborsky N, Gassner FJ, Hainzl S, Weiss L, et al. AID Induces Intraclonal Diversity and Genomic Damage in CD86(+, Chronic Lymphocytic Leukemia Cells. Eur J Immunol, 2014, 44(12):3747–57., DOI 10. 1002/eji.201344421 World Health Organization. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. In: FT Bosman, editors. World Health Organization Classification of Tumours. 4th ed. Lyon, France: International Agency for Research on Cancer, 2017). Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G, Dohner H, et al. iwCLL Guidelines for Diagnosis, Indications for Treatment, Response Assessment, and Supportive Management of CLL. Blood, 2018, 131(25): 2745–60., DOI 10.1182/blood-2017-09-806398 Takacs F, Mikala G, Nagy N, Reszegi A, Czeti A, Szaloki G, et al. Identification of a Novel Resistance Mechanism in Venetoclax Treatment and its Prediction in Chronic Lymphocytic Leukemia. Acta Oncol, 2021, 60:528–30., DOI 10.1080/ 0284186X.2021.1878388 Bodor C, Kotmayer L, Laszlo T, Takacs F, Barna G, Kiss R, et al. Screening and Monitoring of the BTK(C481S, Mutation in a Real-World Cohort of Patients with Relapsed/refractory Chronic Lymphocytic Leukaemia during Ibrutinib Therapy. Br J Haematol, 2021, 194(2):355–64., DOI 10. 1111/bjh.17502 Blombery P, Anderson MA, Gong JN, Thijssen R, Birkinshaw RW, Thompson ER, et al. Acquisition of the Recurrent Gly101Val Mutation in BCL2 Confers Resistance to Venetoclax in Patients with Progressive Chronic Lymphocytic Leukemia. Cancer Discov, 2019, 9(3):342–53., DOI 10.1158/2159-8290.CD-18- 1119 Sedlarikova L, Petrackova A, Papajik T, Turcsanyi P, Kriegova E. Resistance- Associated Mutations in Chronic Lymphocytic Leukemia Patients Treated with Novel Agents. Front Oncol, 2020, 10:894., DOI 10.3389/fonc.2020.00894 Thompson PA. MRD Negativity as a Surrogate for PFS in CLL? Blood, 2018, 131(9):943–4., DOI 10.1182/blood-2018-01-824177 Rendeiro AF, Krausgruber T, Fortelny N, Zhao F, Penz T, Farlik M, et al. Chromatin Mapping and Single-Cell Immune Profiling Define the Temporal Dynamics of Ibrutinib Response in CLL. Nat Commun, 2020, 11(1):577., DOI 10.1038/s41467-019-14081-6 Riether C, Schurch C, Ochsenbein AF. Modulating CD27 Signaling to Treat Cancer. Oncoimmunology, 2012, 1(9):1604–6., DOI 10.4161/onci.21425 Gobessi S, Laurenti L, Longo PG, Sica S, Leone G, Efremov DG. ZAP-70 Enhances B-Cell-Receptor Signaling Despite Absent or Inefficient Tyrosine Kinase Activation in Chronic Lymphocytic Leukemia and Lymphoma B Cells. Blood, 2007, 109(5):2032–9., DOI 10.1182/blood-2006-03-011759 Lafarge ST, Hou S, Pauls SD, Johnston JB, Gibson SB,Marshall AJ. Differential Expression and Function of CD27 in Chronic Lymphocytic Leukemia Cells Expressing ZAP-70. Leuk Res, 2015, 39(7):773–8., DOI 10.1016/j.leukres.2015. 04.016 Del Poeta G, Del Principe MI, Zucchetto A, Luciano F, Buccisano F, Rossi FM, et al. CD69 Is Independently Prognostic in Chronic Lymphocytic Leukemia: a Comprehensive Clinical and Biological Profiling Study. Haematologica, 2012, 97(2):279–87., DOI 10.3324/haematol.2011.052829 Herman SE, Mustafa RZ, Gyamfi JA, Pittaluga S, Chang S, Chang B, et al. Ibrutinib Inhibits BCR and NF-κB Signaling and Reduces Tumor Proliferation in Tissue-Resident Cells of Patients with CLL. Blood, 2014, 123(21):3286–95., DOI 10.1182/blood-2014-02-548610 Dai ZS, Chen QF, Lu HZ, Xie Y. Defective Expression and Modulation of B7-2/ CD86 on B Cells in B Cell Chronic Lymphocytic Leukemia. Int J Hematol, 2009, 89(5):656–63., DOI 10.1007/s12185-009-0320-7 Axelsson S, Magnuson A, Lange A, Alshamari A, Hornquist EH,Hultgren O. A Combination of the Activation Marker CD86 and the Immune Checkpoint Marker B and T Lymphocyte Attenuator, BTLA, Indicates a Putative Permissive Activation State of B Cell Subtypes in Healthy Blood Donors Independent of Age and Sex. BMC Immunol, 2020, 21(1):14., DOI 10.1186/ s12865-020-00343-2 Burger JA, Ghia P, Rosenwald A, Caligaris-Cappio F. The Microenvironment in Mature B-Cell Malignancies: a Target for New Treatment Strategies. Blood, 2009, 114(16):3367–75., DOI 10.1182/blood-2009-06-225326 Lim TS, Goh JK, Mortellaro A, Lim CT, Hammerling GJ, Ricciardi-Castagnoli P. CD80 and CD86 Differentially Regulate Mechanical Interactions of T-Cells with Antigen-Presenting Dendritic Cells and B-Cells. PLoS One, 2012, 7(9): e45185., DOI 10.1371/journal.pone.0045185 Long M, Beckwith K, Do P, Mundy BL, Gordon A, Lehman AM, et al. Ibrutinib Treatment Improves T Cell Number and Function in CLL Patients. J Clin Invest, 2017, 127(8):3052–64., DOI 10.1172/JCI89756 Lanham S, Hamblin T, Oscier D, Ibbotson R, Stevenson F, Packham G. Differential Signaling via Surface IgM Is Associated with VH Gene Mutational Status and CD38 Expression in Chronic Lymphocytic Leukemia. Blood, 2003, 101(3):1087–93., DOI 10.1182/blood-2002-06-1822 Duhren-von Minden M, Ubelhart R, Schneider D, Wossning T, Bach MP, Buchner M, et al. Chronic Lymphocytic Leukaemia Is Driven by Antigenindependent Cell-Autonomous Signalling. Nature, 2012, 489(7415):309–12., DOI 10.1038/nature11309 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610659 EP 1610665 DI 10.3389/pore.2022.1610659 PG 7 ER PT J AU Xu, Y He, J Li, W Zhang, W Liu, S He, J Pan, Z Lu, Z Peng, J Lin, J AF Xu, Yanbo He, Jiarui Li, Weihao Zhang, Weili Liu, Songran He, Jiahua Pan, Zhizhong Lu, Zhenhai Peng, Jianhong Lin, Junzhong TI The Pathologic Complete Response Ratio of Liver Metastases Represents a Valuable Prognostic Indicator SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE recurrence; colorectal cancer; prognosis; liver metastasis; pathologic complete response ratio ID recurrence; colorectal cancer; prognosis; liver metastasis; pathologic complete response ratio AB Background and Objectives: The aim of this study was to evaluate the role of the pathologic complete response ratio of liver metastases (PCRRLM) in predicting the prognosis and recurrence of colorectal cancer liver metastases (CRLM). Methods: A total of 305 CRLM patients who underwent preoperative chemotherapy followed by hepatectomy were included. PCRRLM was defined as the number of liver metastases exhibiting pathologic complete response (PCR) divided by the number of total resected liver metastases. The Kaplan–Meier method was used to calculate survival, and differences were examined by the log-rank test. Univariate and multivariate analyses were performed to identify the predictors of PCRRLM, recurrence-free survival (RFS) and overall survival (OS). Results: Among the 305 included patients, 44 (14.4%) achieved a PCRRLM ≥0.50 (including PCRRLM = 1), and 261 (85.6%) achieved a PCRRLM <0.50 (including PCRRLM = 0). Patients of an older age (≥55 years old) and those with higher carcinoembryonic antigen (CEA) levels (≥5 ng/ml) were less likely to achieve a PCRRLM ≥0.50. In the multivariate analysis, PCRRLM≥ 0.50 (vs. < 0.50, HR [95% CI]: 0.67 [0.46–0.99], p = 0.043) was associated with better RFS. Positive lymph node status (vs. negative, HR [95% CI]: 1.46 [1.04–2.05], p = 0.028) and TBS ≥5 (vs. < 5, HR [95% CI]: 1.44 [1.02–2.04], p = 0.038) were associated with worse RFS. Conclusion: PCRRLM was significantly associated with long-term RFS after preoperative chemotherapy and CRLM resection. Thus, it may be a valuable indicator of recurrence in CRLM patients. C1 [Xu, Yanbo] Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Colorectal SurgeryGuangzhou, China. [He, Jiarui] Sun Yat-sen University, Zhongshan School of MedicineGuangzhou, China. [Li, Weihao] Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Colorectal SurgeryGuangzhou, China. [Zhang, Weili] Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Colorectal SurgeryGuangzhou, China. [Liu, Songran] Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of PathologyGuangzhou, China. [He, Jiahua] Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of PathologyGuangzhou, China. [Pan, Zhizhong] Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Colorectal SurgeryGuangzhou, China. [Lu, Zhenhai] Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Colorectal SurgeryGuangzhou, China. [Peng, Jianhong] Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Colorectal SurgeryGuangzhou, China. [Lin, Junzhong] Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Colorectal SurgeryGuangzhou, China. RP Lin, J (reprint author), Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Colorectal Surgery, Guangzhou, China. EM linjzh@sysucc.org.cn CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Hackl C, Neumann P, Gerken M, Loss M, Klinkhammer-Schalke M, Schlitt HJ. Treatment of Colorectal Liver Metastases in Germany: A Ten-Year Population-Based Analysis of 5772 Cases of Primary Colorectal Adenocarcinoma. BMC Cancer, 2014, 14:810., DOI 10.1186/1471-2407-14-810 Engstrand J, Nilsson H, Stromberg C, Jonas E, Freedman J. Colorectal Cancer Liver Metastases - a Population-Based Study on Incidence, Management and Survival. BMC Cancer, 2018, 18(1):78., DOI 10.1186/ s12885-017-3925-x Dhir M, Sasson AR. Surgical Management of Liver Metastases from Colorectal Cancer. J Oncol Pract, 2016, 12(1):33–9., DOI 10.1200/JOP.2015.009407 Simmonds PC, Primrose JN, Colquitt JL, Garden OJ, Poston GJ, Rees M. Surgical Resection of Hepatic Metastases from Colorectal Cancer: A Systematic Review of Published Studies. Br J Cancer, 2006, 94(7):982–99., DOI 10.1038/sj.bjc.6603033 Aloia TA, Loyer J-NVEM, Ribero D, Pawlik TM, StevenWei HM, Curley SA, et al. Solitary Colorectal Liver Metastasis Resection Determines Outcome. Arch Surg, 2006, 141(5):460–6., DOI 10.1001/archsurg.141.5.460 Van Cutsem E, Nordlinger B, Adam R, Kohne CH, Pozzo C, Poston G, et al. Towards a Pan-European Consensus on the Treatment of Patients with Colorectal Liver Metastases. Eur J Cancer, 2006, 42(14):2212–21., DOI 10. 1016/j.ejca.2006.04.012 Adam R, Delvart V, Pascal G, Valeanu A, Castaing D, Azoulay D, et al. Rescue Surgery for Unresectable Colorectal Liver Metastases Downstaged by Chemotherapy: A Model to Predict Long-Term Survival. Ann Surg, 2004, 240(4):644–57., DOI 10.1097/01.sla.0000141198.92114.f6 Rossi L, Zullo A, Zoratto F, Papa A, Strudel M, Colonna M, et al. Chemotherapy and Target Therapy as Neo-Adjuvant Approach for Initially Unresectable Colorectal Liver Metastases. Oncol Rev, 2012, 6(1):e6., DOI 10. 4081/oncol.2012.e6 Nordlinger B, Sorbye H, Glimelius B, Poston GJ, Schlag PM, Rougier P, et al. Perioperative FOLFOX4 Chemotherapy and Surgery versus Surgery Alone for Resectable Liver Metastases from Colorectal Cancer, EORTC 40983): Long- Term Results of a Randomised, Controlled, Phase 3 Trial. Lancet Oncol, 2013, 14(12):1208–15., DOI 10.1016/s1470-2045(13)70447-9 de Jong MC, Pulitano C, Ribero D, Strub J, Mentha G, Schulick RD, et al. Rates and Patterns of Recurrence Following Curative Intent Surgery for Colorectal Liver Metastasis: An International Multi-Institutional Analysis of 1669 Patients. Ann Surg, 2009, 250(3):440–8., DOI 10.1097/SLA. 0b013e3181b4539b Petrelli F, Coinu A, Zaniboni A, Pietrantonio F, Barni S. Prognostic Factors after R0 Resection of Colorectal Cancer Liver Metastases: A Systematic Review and Pooled-Analysis. Rev Recent Clin Trials, 2016, 11(1):56–62., DOI 10.2174/ 1574887110666151006093403 Halazun KJ, Aldoori A, Malik HZ, Al-Mukhtar A, Prasad KR, Toogood GJ, et al. Elevated Preoperative Neutrophil to Lymphocyte Ratio Predicts Survival Following Hepatic Resection for Colorectal Liver Metastases. Eur J Surg Oncol, 2008, 34(1):55–60., DOI 10.1016/j.ejso.2007.02.014 Iwatsuki SID, Madariaga JR, Marsh JW, Dodson F, Bonham AC, Geller DA, et al. Hepatic Resection for Metastatic Colorectal Adenocarcinoma: A Proposal of a Prognostic Scoring System. J Am Coll Surg, 1999, 189(3):291–9., DOI 10. 1016/s1072-7515(99)00089-7 Umekita N, Maeshiro T, Miyamoto S, Suzuki T, Inoue S, Awane Y. Preoperative CEA Level Predicts Outcomes of Hepatic Resection for Liver Metastases from Colorectal Cancer. Gan To Kagaku Ryoho, 1998, 25(9): 1426–8. Ambiru S, Miyazaki M, Isono T, Ito H, Nakagawa K, Shimizu H, et al. Hepatic Resection for Colorectal Metastases: Analysis of Prognostic Factors. Dis Colon Rectum, 1999, 42(5):632–9., DOI 10.1007/BF02234142 Nakai T, Ishikawa H, Tokoro T, Okuno K. The Clinical Risk Score Predicts the Effectiveness of Adjuvant Chemotherapy for Colorectal Liver Metastasis. World J Surg, 2015, 39(6):1527–36., DOI 10.1007/s00268-015-2980-1 Ayez N, van der Stok EP, Grunhagen DJ, Rothbarth J, van Meerten E, Eggermont AM, et al. The Use of Neo-Adjuvant Chemotherapy in Patients with Resectable Colorectal Liver Metastases: Clinical Risk Score as Possible Discriminator. Eur J Surg Oncol, 2015, 41(7):859–67., DOI 10.1016/j.ejso.2015. 04.012 Rahbari NN, Reissfelder C, Schulze-Bergkamen H, Jager D, Buchler MW, Weitz J, et al. Adjuvant Therapy after Resection of Colorectal Liver Metastases: The Predictive Value of the MSKCC Clinical Risk Score in the Era of Modern Chemotherapy. BMC Cancer, 2014, 14:174., DOI 10.1186/1471-2407-14-174 Hayashi M, Inoue Y, Komeda K, Shimizu T, Asakuma M, Hirokawa F, et al. Clinicopathological Analysis of Recurrence Patterns and Prognostic Factors for Survival after Hepatectomy for Colorectal Liver Metastasis. BMC Surg, 2010, 10:27., DOI 10.1186/1471-2482-10-27 Blazer DG, 3rd, Kishi Y, Maru DM, Kopetz S, Chun YS, Overman MJ, et al. Pathologic Response to Preoperative Chemotherapy: A New Outcome End point after Resection of Hepatic Colorectal Metastases. J Clin Oncol, 2008, 26(33):5344–51., DOI 10.1200/JCO.2008.17.5299 Xu D, Liu XF, Yan XL, Wang K, Xing BC. Survival Prediction in Patients with Resectable Colorectal Liver Metastases: Clinical Risk Scores and Tumor Response to Chemotherapy. Oncol Lett, 2017, 14(6):8051–9., DOI 10.3892/ol. 2017.7191 Xu D, Yan XL, Liu JM, Li J, Xing BC. The Characteristics and Long-Term Survival of Patients with Colorectal Liver Metastases with Pathological Complete Response after Chemotherapy. J Cancer, 2020, 11(21):6256–63., DOI 10.7150/jca.47911 Arata R, Itamoto T, Ikeda S, Nakahara H, Oshita A, Shinozaki K, et al. Pathological Complete Response after Neoadjuvant Chemotherapy for Rectal Cancer with Synchronous Multiple Liver Metastases: A Report of an Unusual Case. Surg Case Rep, 2016, 2(1):106., DOI 10.1186/s40792-016-0231-9 Hakoda K, Yoshimitsu M, Emi M, Omori I, Kohashi T, Kaneko M, et al. Complete Pathological Response of Multiple Huge Liver Metastases of colon Cancer: a Case Report. OxfMed Case Rep, 2017, 2017(5):omx016., DOI 10.1093/ omcr/omx016 Sinukumar S, Patil P, Engineer R, Desouza A, Saklani A. Clinical Outcome of Patients with Complete Pathological Response to Neoadjuvant Chemoradiotherapy for Locally Advanced Rectal Cancers: The Indian Scenario. Gastroenterol Res Pract, 2014, 2014:867841., DOI 10.1155/2014/ 867841 Yamashita S, Chun YS, Kopetz SE, Maru D, Conrad C, Aloia TA, et al. APC and PIK3CA Mutational Cooperativity Predicts Pathologic Response and Survival in Patients Undergoing Resection for Colorectal Liver Metastases. Ann Surg, 2020, 272(6):1080–5., DOI 10.1097/SLA.0000000000002245 Adam R, Wicherts DA, de Haas RJ, Aloia T, Levi F, Paule B, et al. Complete Pathologic Response after Preoperative Chemotherapy for Colorectal Liver Metastases: Myth or Reality? J Clin Oncol, 2008, 26(10):1635–41., DOI 10.1200/ jco.2007.13.7471 Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New Response Evaluation Criteria in Solid Tumours: Revised RECIST Guideline, Version 1.1). Eur J Cancer, 2009, 45(2):228–47., DOI 10.1016/j. ejca.2008.10.026 Mekenkamp LJ, Koopman M, Teerenstra S, van Krieken JH, Mol L, Nagtegaal ID, et al. Clinicopathological Features and Outcome in Advanced Colorectal Cancer Patients with Synchronous vs Metachronous Metastases. Br J Cancer, 2010, 103(2):159–64., DOI 10.1038/sj.bjc.6605737 Sasaki K, Morioka D, Conci S, Margonis GA, Sawada Y, Ruzzenente A, et al. The Tumor Burden Score: A New “Metro-Ticket” Prognostic Tool for Colorectal Liver Metastases Based on Tumor Size and Number of Tumors. Ann Surg, 2016, 267(1):132–41., DOI 10.1097/sla.0000000000002064 Fong Y, Fortner J, Sun RL, Brennan MF, Blumgart LH. Clinical Score for Predicting Recurrence after Hepatic Resection for Metastatic Colorectal Cancer: Analysis of 1001 Consecutive Cases. Ann Surg, 1999, 230(3): 309–18., DOI 10.1097/00000658-199909000-00004 Jung SW, Kim DS, Yu YD, Han JH, Suh SO. Risk Factors for Cancer Recurrence or Death within 6 Months after Liver Resection in Patients with Colorectal Cancer Liver Metastasis. Ann Surg Treat Res, 2016, 90(5): 257–64., DOI 10.4174/astr.2016.90.5.257 Mahar AL, Compton C, Halabi S, Hess KR, Weiser MR, Groome PA. Personalizing Prognosis in Colorectal Cancer: A Systematic Review of the Quality and Nature of Clinical Prognostic Tools for Survival Outcomes. J Surg Oncol, 2017, 116(8):969–82., DOI 10.1002/jso.24774 Tanaka K, Takakura H, Takeda K, Matsuo K, Nagano Y, Endo I. Importance of Complete Pathologic Response to Prehepatectomy Chemotherapy in Treating Colorectal Cancer Metastases. Ann Surg, 2009, 250(6):935–42., DOI 10.1097/ sla.0b013e3181b0c6e4 Dworak O, Keilholz L, Hoffmann A. Pathological Features of Rectal Cancer after Preoperative Radiochemotherapy. Int J Colorectal Dis, 1997, 12(1):19–23., DOI 10.1007/s003840050072 Reddy SK, Parker RJ, Leach JW, Hill MJ, Burgart LJ. Tumor Histopathology Predicts Outcomes after Resection of Colorectal Cancer Liver Metastases Treated with and without Pre-Operative Chemotherapy. J Surg Oncol, 2016, 113(4):456–62., DOI 10.1002/jso.24144 Sebagh M, Allard MA, Cunha AS, Ruiz A, Araujo R, Lemoine A, et al. A Proposed New Method for Assessing the Pathological Response to Chemotherapy in Resected Colorectal Liver Metastases. Br J Cancer, 2014, 111(3):470–6., DOI 10.1038/bjc.2014.299 Rubbia-Brandt L, Giostra E, Brezault C, Roth AD, Andres A, Audard V, et al. Importance of Histological Tumor Response Assessment in Predicting the Outcome in Patients with Colorectal Liver Metastases Treated with Neo- Adjuvant Chemotherapy Followed by Liver Surgery. Ann Oncol, 2007, 18(2): 299–304., DOI 10.1093/annonc/mdl386 Wang Y, Yuan YF, Lin HC, Li BK, Wang FH, Wang ZQ, et al. Pathologic Response after Preoperative Therapy Predicts Prognosis of Chinese Colorectal Cancer Patients with Liver Metastases. Chin J Cancer, 2017, 36(1):78., DOI 10. 1186/s40880-017-0244-1 Auer RC, White RR, Kemeny NE, Schwartz LH, Shia J, Blumgart LH, et al. Predictors of a True Complete Response Among Disappearing Liver Metastases from Colorectal Cancer after Chemotherapy. Cancer, 2010, 116(6):1502–9., DOI 10.1002/cncr.24912 Buisman FE,Galjart B, Buettner S, GrootKoerkamp B,GrunhagenDJ,Verhoef C. Primary Tumor Location and the Prognosis of Patients after Local Treatment of Colorectal Liver Metastases: A Systematic Review and Meta-Analysis. HPB, Oxford,, 2020, 22(3):351–7., DOI 10.1016/j.hpb.2019.10.003 Imai K, Yamashita YI, Miyamoto Y, Nakao Y, Yusa T, Itoyama R, et al. Implication of Primary Tumor Location for the Indication of Preoperative Chemotherapy in Patients with Colorectal Liver Metastases. HPB, Oxford,, 2019, 21(4):405–12., DOI 10.1016/j.hpb.2018.08.012 Sunakawa Y, Ichikawa W, Tsuji A, Denda T, Segawa Y, Negoro Y, et al. Prognostic Impact of Primary Tumor Location on Clinical Outcomes of Metastatic Colorectal Cancer Treated with Cetuximab Plus Oxaliplatin- Based Chemotherapy: A Subgroup Analysis of the JACCRO CC-05/ 06 Trials. Clin Colorectal Cancer, 2017, 16(3):e171–80., DOI 10.1016/j.clcc. 2016.09.010 Wang F, Bai L, Liu TS, Yu YY, He MM, Liu KY, et al. Right-Sided Colon Cancer and Left-Sided Colorectal Cancers Respond Differently to Cetuximab. Chin J Cancer, 2015, 34(9):384–93., DOI 10.1186/s40880-015-0022-x Stintzing S, Tejpar S, Gibbs P, Thiebach L, Lenz HJ. Understanding the Role of Primary Tumour Localisation in Colorectal Cancer Treatment and Outcomes. Eur J Cancer, 2017, 84:69–80., DOI 10.1016/j.ejca.2017.07.016 Benedix F, Kube R, Meyer F, Schmidt U, Gastinger I, Lippert H, et al. Comparison of 17, 641 Patients with Right- and Left-Sided Colon Cancer: Differences in Epidemiology, Perioperative Course, Histology, and Survival. Dis Colon Rectum, 2010, 53(1):57–64., DOI 10.1007/DCR.0b013e3181c703a4 Ruzzenente A, Bagante F, Ratti F, Beal EW, Alexandrescu S, Merath K, et al. Response to Preoperative Chemotherapy: Impact of Change in Total Burden Score and Mutational Tumor Status on Prognosis of Patients Undergoing Resection for Colorectal Liver Metastases. HPB, Oxford,, 2019, 21(9):1230–9., DOI 10.1016/j.hpb.2019.01.014 Sasaki K, Margonis GA, Andreatos N, Zhang XF, Buettner S, Wang J, et al. The Prognostic Utility of the “Tumor Burden Score” Based on Preoperative Radiographic Features of Colorectal Liver Metastases. J Surg Oncol, 2017, 116(4):515–23., DOI 10.1002/jso.24678 Vigano L, Capussotti L, De Rosa G, De Saussure WO, Mentha G, Rubbia- Brandt L. Liver Resection for Colorectal Metastases after Chemotherapy: Impact of Chemotherapy-Related Liver Injuries, Pathological Tumor Response, and Micrometastases on Long-Term Survival. Ann Surg, 2013, 258(5):731–40., DOI 10.1097/SLA.0b013e3182a6183e Berg KCG, Brunsell TH, Sveen A, Alagaratnam S, Bjornslett M, Hektoen M, et al. Genomic and Prognostic Heterogeneity Among RAS/BRAF(V600E)/ TP53 Co-Mutated Resectable Colorectal Liver Metastases. Mol Oncol, 2021, 15(4):830–45., DOI 10.1002/1878-0261.12885 Brunsell TH, Sveen A, Bjornbeth BA, Rosok BI, Danielsen SA, Brudvik KW, et al. High Concordance and Negative Prognostic Impact of RAS/BRAF/ PIK3CA Mutations in Multiple Resected Colorectal Liver Metastases. Clin Colorectal Cancer, 2020, 19(1):e26–e47., DOI 10.1016/j.clcc.2019.09.003 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610663 EP 1610672 DI 10.3389/pore.2022.1610663 PG 10 ER PT J AU Kenessey, I Szoke, G Dobozi, M Szatmari, I Weber, A Fogarassy, Gy Nagy, P Kasler, M Polgar, Cs Vathy-Fogarassy, AF Kenessey, Istvan Szoke, Georgina Dobozi, Maria Szatmari, Istvan Weber, Andras Fogarassy, Gyorgy Nagy, Peter Kasler, Miklos Polgar, Csaba Vathy-Fogarassy, Agnes TI Comparison of Cancer Survival Trends in Hungary in the Periods 2001–2005 and 2011–2015 According to a Population-Based Cancer Registry SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mortality; survival; incidence; oncology; cancer registry; trend ID mortality; survival; incidence; oncology; cancer registry; trend AB Background: Assessment of population-based cancer survival may provide the most valuable feedback about the effectiveness of oncological surveillance and treatment. Aims: Based on the database of the Hungarian National Cancer Registry, standardized incidence rates of lung, breast, colorectal, prostate and cervical cancer were compared to standardized mortality data of the Hungarian Central Statistical Office in the period between 2001 and 2015. Then survival analysis was performed on cleansed database. Results: The incidence of colorectal, breast and prostate cancer increased, while standardized rates of lung and cervical cancer declined. The survival of colorectal, breast and prostate cancer showed improvement. Contrarily, lung cancer exhibited a mild decline, while that of cervical cancer did not change significantly. In earlier stages survival was improved among almost every studied tumor type, while in advanced stages improvement was not observed. Comparison of stage distribution revealed that in the 2011–2015 period colorectal, breast and prostate cancer cases were diagnosed at earlier stages, while lung and cervical cancer patients were typically discovered at more advanced stages. Discussion: The outcome of advanced cancer treatments is better in earlier stages, which highlighted the importance of screening network. However, growth of oncological treatment costs with longer patient survival imposes a constantly increasing burden on society. C1 [Kenessey, Istvan] National Institute of OncologyBudapest, Hungary. [Szoke, Georgina] Pannon Egyetem, Rendszer- es Szamitastudomanyi TanszekVeszprem, Hungary. [Dobozi, Maria] National Institute of OncologyBudapest, Hungary. [Szatmari, Istvan] National Institute of OncologyBudapest, Hungary. [Weber, Andras] National Institute of OncologyBudapest, Hungary. [Fogarassy, Gyorgy] Allami Szivkorhaz, 1. Sz. Kardiologiai OsztalyBalatonfured, Hungary. [Nagy, Peter] National Institute of OncologyBudapest, Hungary. [Kasler, Miklos] National Institute of OncologyBudapest, Hungary. [Polgar, Csaba] National Institute of OncologyBudapest, Hungary. [Vathy-Fogarassy, Agnes] Pannon Egyetem, Rendszer- es Szamitastudomanyi TanszekVeszprem, Hungary. RP Kenessey, I (reprint author), National Institute of Oncology, Budapest, Hungary. EM steveken12@yahoo.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Ferlay J, Colombet M, Soerjomataram I, Dyba T, Randi G, Bettio M, et al. Cancer Incidence and Mortality Patterns in Europe: Estimates for 40 Countries and 25 Major Cancers in 2018. Eur J Cancer, 2018, 103:356–87., DOI 10.1016/j. ejca.2018.07.005 Arnold M, Rutherford MJ, Bardot A, Ferlay J, Andersson TM, Myklebust TA, et al. Progress in Cancer Survival, Mortality, and Incidence in Seven High- Income Countries 1995-2014, ICBP SURVMARK-2): a Population-Based Study. Lancet Oncol, 2019, 20:1493–505., DOI 10.1016/S1470-2045(19, 30456-5 Tusnady G, Gaudi I, Rejto L, Kasler M, Szentirmay Z. Survival Chances of Hungarian Cancer Patients in the National Cancer Registry. Magy Onkol, 2008, 52:339–49., DOI 10.1556/MOnkol.52.2008.4.2 World Health Organization. International Statistical Classification of Diseases and Related Health Problems, Tenth Revision. 2nd ed. Geneva, Switzerland: World Health Organization, 2004). Sobin LH, Gospodarowicz MK, Wittekind C. TNM Classification of Malignant Tumours. 7th ed. Hoboken, New Jersey, US: Wiley-Blackwell, 2010). Brierley JD, Gospodarowicz MK, Wittekind C. TNM Classification of Malignant Tumours. 8th ed. Hoboken, New Jersey, US: Wiley, 2017). KSH. Hungarian Central Statistical Office, 2020). https://www.ksh.hu/shelf, Accessed January 31, 2022). Eurostat. Revision of the European Standard Population - Report of Eurostat’s Task Force - 2013 Edition, 2013). https://ec.europa.eu/eurostat/documents/3859598/ 5926869/KS-RA-13-028-EN.PDF/e713fa79-1add-44e8-b23d-5e8fa09b3f8f, Accessed November 18, 2020). Team RC. A Language and Environment for Statistical Computing, 2020). https://www.R-project.org/(Accessed November 18, 2020). Prasad V, Fojo T, Brada M. Precision Oncology: Origins, Optimism, and Potential. Lancet Oncol, 2016, 17:e81–6., DOI 10.1016/S1470-2045(15)00620-8 Michigan State University. Hungarian National Institute of Pharmacy and Nutrition, 2021). https://ogyei.gov.hu/gyogyszeradatbazis, Accessed January 31, 2022). Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, et al. Cancer Incidence and Mortality Patterns in Europe: Estimates for 40 Countries in 2012. Eur J Cancer, 2013, 49:1374–403., DOI 10.1016/j.ejca.2012.12.027 Global Burden of Disease Cancer C, Fitzmaurice C, Dicker D, Pain A, Hamavid H, Moradi-Lakeh M, et al. The Global Burden of Cancer 2013. JAMA Oncol, 2015, 1:505–27., DOI 10.1001/jamaoncol.2015.0735 Jemal A, Ward EM, Johnson CJ, Cronin KA, Ma J, Ryerson B, et al. Annual Report to the Nation on the Status of Cancer, 1975-2014, Featuring Survival. J Natl Cancer Inst, 2017, 109., DOI 10.1093/jnci/djx030 Bradley CJ, Yabroff KR, Mariotto AB, Zeruto C, Tran Q, Warren JL. Antineoplastic Treatment of Advanced-Stage Non-small-cell Lung Cancer: Treatment, Survival, and Spending, 2000 to 2011). J Clin Oncol, 2017, 35: 529–35., DOI 10.1200/JCO.2016.69.4166 Loud JT, Murphy J. Cancer Screening and Early Detection in the 21(st, Century. Semin Oncol Nurs, 2017, 33:121–8., DOI 10.1016/j.soncn.2017. 02.002 Byers T, Wender RC, Jemal A, Baskies AM, Ward EE, Brawley OW. The American Cancer Society challenge Goal to Reduce US Cancer Mortality by 50% between 1990 and 2015: Results and Reflections. CA Cancer J Clin, 2016, 66:359–69., DOI 10.3322/caac.21348 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610668 EP 1610677 DI 10.3389/pore.2022.1610668 PG 10 ER PT J TI Corrigendum: Methylation Statuses of H19DMR and KvDMR at WT2 in Wilms Tumors in Taiwan SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE methylation; multiplex ligation-dependent probe amplification; Nephroblastoma; paternal uniparental disomy; Wilms tumor ID methylation; multiplex ligation-dependent probe amplification; Nephroblastoma; paternal uniparental disomy; Wilms tumor AB In the published article, there was an error in the Funding statement. The funding statement for the grant number from Chung Shan Medical University was displayed as “CSMU-INT-108-2.” The correct statement is “This work was supported by a grant from Chung Shan Medical University (grant number CSMU-INT-108-12).” The authors apologize for this error and state that this does not change the scientific conclusions of the article in any way. NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610677 EP 1610677 DI 10.3389/pore.2022.1610677 PG 1 ER PT J AU Timar, J Uhlyarik, A AF Timar, Jozsef Uhlyarik, Andrea TI On-Target Side Effects of Targeted Therapeutics of Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE immunotherapy; target therapy; bone therapy; angiogenesis inhibitor; side effect; pathomechanism ID immunotherapy; target therapy; bone therapy; angiogenesis inhibitor; side effect; pathomechanism AB The concept of precision medicine is based on the identification of hallmarks of cancer to exploit them as drug targets. The basic idea was that in this way the therapeutic modalities will be more effective and the side effects will be less. Since the majority of these novel modalities are not specific for a cancer-related biological process or a cancer-specific (mutant) target protein, it is not a surprise that we had to learn new type of side effects, because these therapeutics also affect physiological or pathological processes. Even more, in cases of some of these novel therapies we were able to discover new molecular mechanisms of physiological and pathological processes. Identification of the on-target side effects of targeted drugs can help to prevent the development of them or better manage the patients when emerge during cancer therapy. C1 [Timar, Jozsef] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Uhlyarik, Andrea] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM jtimar@gmail.com CR Sebestyen A, Kopper L, Danko T, Timar J. Hypoxia Signaling in Cancer: from Basics to Clinical Practice. Pathol Oncol Res, 2021, 27:1609802., DOI 10.3389/ pore.2021.1609802 Cheng K, Liu C-F, Rao G-W. Anti-angiogenic Agents: A Review on Vascular Endothelial Growth Factor Receptor-2, VEGFR-2, Inhibitors. Curr Med Chem, 2021, 28:2540–64., DOI 10.2174/ 0929867327666200514082425 Salgado R, Benoy I, Bogers R, Wejtien R, Vermeulen P, Dirix L, et al. Platelets and Vascular Endothelial Growth Factor, VEGF): a Morphological and Functional Study. Angiogenesis, 2001, 4:37–43., DOI 10.1023/a:1016611230747 Touyz RM, Herrmann SMS, Herrmann J. Vascular Toxicities with VEGF Inhibitor Therapies-Focus on Hypertension and Arterial Thrombotic Events. J Am Soc Hypertens, 2018, 12:409–25., DOI 10.1016/j.jash.2018. 03.008 Timar J, Uhlyarik A. A Celzott Kezelesek Celzott Mellekhatasai. KLINIKAI ONKOLOGIA, 2022, 9:157–68., in Hungarian). Versmissen J, Mirabito Colafella KM, Koolen SLW, Danser AHJ. Vascular Cardio-Oncology: Vascular Endothelial Growth Factor Inhibitors and Hypertension. Cardiovasc Res, 2019, 113:904–14., DOI 10.1093/cvr/cvz022 Chen W, Hill H, Christie A, Kim MS, Pavia-Jimenez A, Homayoun F, et al. Targeting Renal Cell Carcinoma with a HIF-2 Antagonist. Nature, 2016, 539: 112–7., DOI 10.1038/nature19796 Food and Drug Administration. US Food and Drug Administration, 2022). Available from: www.fda.gov/resources-information-approveddrugs. Jonasch E, Donskov F, Iliopoulos O, Rathmell WK, Narayan VK, Maughan B, et al. Belzutifan for renal cell carcinoma in von Hippel-Lindau disease. N Engl J Med, 2022, 385:2036–46., DOI 10.1056/nejmoa2103425 Shapiro CL. Bone-modifying Agents, BMAs, in Breast Cancer. Clin Breast Cancer, 2021, 25:e618–e630., DOI 10.1016/j.clbc.2021.04.009 Casimiro S, Vilhais G, Gomes I, Costa L. The Roadmap of RANKL/RANK Pathway in Cancer. Cells, 1978, 10:1978., DOI 10.3390/cells10081978 Jara MA, Varghese J, Hu MI. Adverse Events Associated with Bone-Directed Therapies in Patients with Cancer. Bone, 2022, 158:115901., DOI 10.1016/j. bone.2021.115901 Jackson C, Freeman ALJ, Szlamka Z, Spiegelhalter DJ. The Adverse Effects of Bisphosphonates in Breast Cancer: A Systematic Review and Network Meta-Analysis. PLoS ONE, 2021, 16:e0246441., DOI 10.1371/journal.pone. 0246441 Lorusso L, Pieruzzi L, Gabriele M, Morinaro E. Osteonecrosis of the Jaw. J Endocrinol Invest, 2021, 44:2537–66. Zarour HM. Reversing T-Cell Dysfunction and Exhaustion in Cancer. Clin Cancer Res, 2016, 22:1856–64., DOI 10.1158/1078-0432.CCR-15-1849 Joller N, Peters A, Anderson AC, Kuchroo VK. Immune Checkpoints in central Nervous System Autoimmunity. Immunol Rev, 2012, 248:122–39., DOI 10.1111/j.1600-065X.2012.01136.x Zhai Y, moosavi R, chen M. Immune Checkpoints a Novel Class of Therapeutic Targets for Autoimmune Diseases. Front Immunol, 2021, 12: 645699., DOI 10.3389/fimmu.2021.645699 Eggermont AM, Chiarion-Sileni V, Grobb J, Dummer R, Wolchok JD, Schmidt H, et al. Prolonged Survival in stageIII Melanoma with Ipilimumab Adjuvant Therapy. N Engl J Med, 2016, 375:1845–55., DOI 10.1056/NEJMoa1611299 Robert C, Long GV, brady B, Dutriaux C,Maio M, Mortier L, et al. Nivolumab in Previously Untreated Melanoma without BRAF Mutation. N Engl J Med, 2015, 372:320–30., DOI 10.1056/NEJMoa1412082 Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, et al. Pembrolizumab versus Docetaxel for Previously Treated, PD-L1-Positive, Advanced Non-small-cell Lung Cancer, KEYNOTE-010): a Randomised Controlled Trial. Lancet, 2016, 387:1540–50., DOI 10.1016/S0140-6736(15, 01281-7 Gulley JL, Rajan A, Spigel TR, Iannotti N, Chandler J, Wong DJL, et al. Avelumab for Patients with Previously Treated Metastatic or Recurrent Nonsmall- cell Lung Cancer, JAVELIN Solid Tumor): Dose-Expansion Cohort of a Multicentre, Open-Label, Phase 1b Trial. Lancet Oncol, 2017, 18:599–610., DOI 10.1016/S1470-2045(17)30240-1 Rittmayer A, Barlesi A, Waterkamp D, Park K, Ciadiello F, Hida T, et al. Atezolizumab versus Docetaxel in Pati4nts with Previously Treated NSCLC, OAK): a Phase III Open Label Multicenter Randomized Controlled Trial. Lancet, 2017, 389:255–65. Garassino MC, Cho BC, Kim JH, Mazieres J, Vansteenkiste J, Lena H, et al. Durvalumab as Third-Line or Later Treatment for Advanced Non-smallcell Lung Cancer, ATLANTIC): an Open-Label, Single-Arm, Phase 2 Study. Lancet Oncol, 2018, 19:521–36., DOI 10.1016/S1470-2045(18, 30144-X Martins F, Sofyia L, Sykiotis GP, Lamine F,Maillard M, Fraga M, et al. Adverse Effects of Immune-Checkpoint Inhibitors: Epidemiology, Management and Surveillance. Nat Rev Clin Oncol, 2019, 16:563–80., DOI 10.1038/s41571-019- 0218-0 Ralli M, Boticelli A, Visconti IC, Angelletti D, Fiore M, Marchetti P, et al. Immunotherapy in the Treatment of Metastatic Melanoma: Current Knowledge and Future Directions. J Immunol Res, 2020, 2020:9235638., DOI 10.1155/2020/9235638 Hellmann MD, Rizvi NA, Goldman JW, Gettinger SN, Borghaei H, Brahmer JR, et al. Nivolumab Plus Ipilimumab as First-Line Treatment for Advanced Non-small-cell Lung Cancer, CheckMate 012): Results of an Open Label, Phase 1, Multicohort Study. Lancet Oncol, 2017, 18:31–41., DOI 10.1016/S1470- 2045(16)30624-6 Motzer RJ, Tannir NM, McDermott DF, Frontera OA, Melichar B, Choueiri TK, et al. Nivolumab Plus Ipilimumab versus Sunitinib in Advanced Renal- Cell Carcinoma. N Engl J Med, 2018, 378:1277–90., DOI 10.1056/ NEJMoa1712126 Hellmann MD, Ciuleanu T-E, Plizansky A, Lee JS, Otterson GA, Audigier- Valette C, et al. Nivolumab Plus Ipilimumab in Lung Cancer with a High Tumor Mutational burden. N Engl J Med, 2018, 378:2093–104., DOI 10.1056/ NEJMoa1801946 Bouwhuis MG, tenHagen TLM, Suciu S, Eggermont AMM. Autoimmunity and Treatment Outcome in Melanoma. Curr Opin Oncol, 2011, 23:170–6., DOI 10.1097/CCO.0b013e328341edff Ye W, Olsson-Brown A,Watson RA, Cheung VTF,Morgan RD, Nassiri I, et al. Checkpoint Blocker Induced Autoimmunity Is Associated with Favourable Outcome in Metastatic Melanoma and Distinct T-Cell Expression Profiles. Br J Cancer, 2021, 124:1661–9., DOI 10.1038/s41416-021-01310-3 Zhong L, Wu Q, Chen F, Liu J, Xie X. Immune-related Adverse Events: Promising Predictors for Efficacy of Immune Checkpoint Inhibitors. Cancer Immunol Immunother, 2021, 70:2559–76., DOI 10.1007/s00262-020-02803-5 Guida M, Strippoli S, Maule M, Quaglino P, Ramondetta A, Queirolo P, et al. Immune Checkpoint Inhibitor Associated Vitiligo and its Impact on Survival in Patients with Metastatic Melanoma. ESMO Open, 2021, 6:100064., DOI 10. 1016/j.esmoop.2021.100064 Wu Q, Bai B, Tian C, Li D, Yu H, Song B, et al. The Molecular Mechaisms of Cardiotoxicity Induced by HER2, VEGF, and Tyrosine Kinase Inhibitors. Cardiovasc Drugs Ther, 2021, 36:511–24., DOI 10.1007/s10557-021-07181-3 D’Uva G, Tzahor E. The Key Roles of ERBB2 in Cardiac Regeneration. Cell Cycle, 2015, 14:2383–4., DOI 10.1080/15384101.2015.1063292 Anjos M, Fontes-Oliviera M, Costa VM, Santos M, Ferreira R. An Update of the Molecular Mechanisms Undelying Doxorubicin Plus Trastuzumab Induced Cardiotoxicity. Life Sci, 2021, 280:119760., DOI 10.1016/j.lfs.2021. 119760 Hampton T. New Insight on Preventing EGFR Inhibitor-Induced Adverse Effects. JAMA, 2020, 323:814–5., DOI 10.1001/jama.2020.0812 Galimont-Collen AFS, Vos LE, Lavrijsen APM, Ouwerkerk J, Gelderblom H. Classification and Management of Skin, Hair, Nail and Mucosal Side-Effects of Epidermal Growth Factor Receptor, EGFR, Inhibitors. Eur J Cancer, 2007, 43: 845–51., DOI 10.1016/j.ejca.2006.11.016 Subbiah V, Baik C, Kirkwood JM. Clinical Development of BRAF Plus MEK Inhibitor Combinations. Trends Cancer, 2020, 6:797–810., DOI 10.1016/j. trecan.2020.05.009 Moreira A, Lebbe C, Heinzerling L. MAPK Blockade, Toxicities, Pathogenesis and Management. Curr Opin Oncol, 2021, 33:139–45., DOI 10.1097/CCO. 0000000000000710 Livingstone E, Zimmer L, Vaubel J, Schadendorf D. BRAF, MEK and KIT Inhibitors for Melanoma: Adverse Events and Their Management. Chi Clin Oncol, 2014, 29. Galanis A, Levis M. Inhibition of C-Kit by Tyrosine Kinase Inhibitors. Haematologica, 2015, 100:e77–9., DOI 10.3324/haematol.2014.117028 Fachi MM, Tonin FS, Leonart L, Rotta I, Fernandez-Llimos F, Pontarolo R. Haematological Adverse Events Associated with Tyrosine Kinase Inhibitors in Chronic Myeloid Leukaemia: A Network Meta-Analysis. Br J Clin Pharmacol, 2019, 85:2280–91., DOI 10.1111/bcp.13933 Hou H, Sun D, Jiang M, Liu D, Zhou N, Zhu J, et al. The Safety and Serious Adverse Events of Approved ALK Inhibitors in Malignancies. A Meta- Analysis. Cancer Manag Res, 2019, 11:4109–18., DOI 10.2147/CMAR.S190098 Liu D, Lin FA, Offin M, Falcon CJ, Marciano-Goroff YR, Lin A, et al. Characterization of On-Target Adverse Events Caused by TRK Inhibitor Therapy. Ann Oncol, 2020, 31:1207–15., DOI 10.1016/j.annonc.2020. 05.006 Zhang Y, Yan H, Xu Z, Yang B, Luo P, He Q.Molecular Basis for Class Side Effects Associated with PI3K/AKT/mTOR Pathway Inhibitors. Expert Opin Drug Metab Toxicol, 2019, 15:767–74., DOI 10.1080/17425255. 2019.1663169 Pallet N, Legendre C. Adverse Events Associated with mTOR Inhibitors. Expert Opin Drug Saf, 2013, 12:177–86., DOI 10.1517/14740338.2013.752814 Tankova T, Senkus E, Beloyartseva M, Bortsnar S, Catrinoiu D, Frolova MS, et al. Management Strategies for Hyprglycemia Associated with the A-Selectgive PI3K Inhibitor Alpelisib for the Treatment of Breast Cancer. Cancers, 2022, 14:1598., DOI 10.3390/cancers14071598 Caron A, Richard D, Laplante M. The Roles of mTOR Complexes in Lipid Metabolism. Annu Rev Nutr, 2015, 35:321–48., DOI 10.1146/annurev-nutr- 071714-034355 Generali D, Montemurro F, Bordonaro R, Mafodda A, Romito S, Michelotti A, et al. Everolimus Plus Exemestane in Advanced Breast Cancer: Safety Results of the BALLET Study on Patients Previously Treated without and with Chemotherapy in the Metastatic Setting. Oncologist, 2017, 22:648–54., DOI 10.1634/theoncologist.2016-0461 Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, et al. Everolimus for Advanced Pancreatic Neuroendocrine Tumors. N Engl J Med, 2011, 364:514–23., DOI 10.1056/NEJMoa1009290 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610694 EP 1610705 DI 10.3389/pore.2022.1610694 PG 12 ER PT J AU , PaORPO AF , Pathology and Oncology Research Production Office TI Erratum: The Role of Time as a Prognostic Factor in Pediatric Brain Tumors: a Multivariate Survival Analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE Symptomatic interval; Delayed diagnosis; CNS tumors; Survival outcome; pretreatment interval ID Symptomatic interval; Delayed diagnosis; CNS tumors; Survival outcome; pretreatment interval AB An Erratum on The Role of Time as a Prognostic Factor in Pediatric Brain Tumors: a Multivariate Survival Analysis by Barragan-Perez EJ, Altamirano-Vergara CE, Alvarez-Amado DE, Garcia-Beristain JC, Chico- Ponce-de-Leon F, Gonzalez-Carranza V, Juarez-Villegas L, Murata C (2020). Pathol. Oncol. Res. 26: 2693–2701. 10.1007/s12253-020-00875-3 Due to a production error at the previous publisher, the Supplementary Material in the original article incorrectly included a copy of the manuscript. The correct version of the Supplementary Material is published alongside this erratum. The original version of this article has not been updated. C1 [, Pathology and Oncology Research Production Office] Frontiers Media SALausanne, Switzerland. RP , PaORPO (reprint author), Frontiers Media SA, Lausanne, Switzerland. EM production@por-journal.com NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD SEP PY 2022 VL 28 IS 3 BP 1610756 EP 1610756 DI 10.3389/pore.2022.1610756 PG 1 ER PT J AU Huo, Q Hu, J Hou, B Zhao, M Han, X Du, Y Li, Y AF Huo, Qi Hu, Junjie Hou, Binfen Zhao, Mei Han, Xue Du, Yulin Li, Yao TI Clinicopathological Features and Prognostic Evaluation of UBR5 in Liver Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; TCGA; liver cancer; UBR5; YWHAZ ID prognosis; TCGA; liver cancer; UBR5; YWHAZ AB Background: Typically, liver cancer patients are diagnosed at an advanced stage and have a poor prognosis. N-recognin 5 (UBR5), a component of the ubiquitin protein ligase E3, is involved in the genesis and progression of several types of cancer. As of yet, it is unknown what the exact biological function of UBR5 is in liver cancer. Methods: A Kaplan-Meier survival curve (OS) was used to examine the effect of UBR5 expression on overall survival based on the TCGA database. To determine the molecular functions of UBR5 in liver cancer, we used the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. A protein-protein interaction (PPI) network was established for the screening of UBR5-related proteins in liver cancer. Western blot analysis was used to determine the expression levels of UBR5 and YWHAZ (tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta), and in order to detect cell proliferation, an MTT assay was used. Results: The expression of UBR5 in liver cancer patient samples is significantly higher than in adjacent normal tissues. A high level of UBR5 expression was associated with older patients, a higher tumor grade, lymph node metastasis, and poor survival. We discovered YWHAZ with high connectivity, and UBR5 expression correlated positively with YWHAZ expression (r = 0.83, p < 0.05). Furthermore, we found that elevated UBR5 levels directly correlated with YWHAZ overexpression, and that UBR5 promoted cell proliferation by affecting YWHAZ expression. Additionally, the TCGA databases confirmed that patients with liver cancer who expressed higher levels of YWHAZ had poorer outcomes. Conclusion: This suggests that UBR5 associated with YWHAZ may influence prognosis in patients with liver cancer, and that UBR5 may be a candidate treatment target for liver cancer. Therefore, UBR5 associated with YWHAZ may influence prognosis in patients with liver cancer, and UBR5 could serve as a potential target for liver cancer treatment. C1 [Huo, Qi] Bengbu Medical College, The Second Affiliated Hospital, Department of Medical OncologyBengbu, China. [Hu, Junjie] Bengbu Medical College, The Second Affiliated Hospital, Department of Medical OncologyBengbu, China. [Hou, Binfen] Bengbu Medical College, Department of Laboratory Medicine, Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and ImmunologyBengbu, China. [Zhao, Mei] Bengbu Medical College, The Second Affiliated Hospital, Department of Medical OncologyBengbu, China. [Han, Xue] Bengbu Medical College, Department of Laboratory Medicine, Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and ImmunologyBengbu, China. [Du, Yulin] Bengbu Medical College, Department of Laboratory Medicine, Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and ImmunologyBengbu, China. [Li, Yao] Bengbu Medical College, Department of Laboratory Medicine, Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and ImmunologyBengbu, China. RP Li, Y (reprint author), Bengbu Medical College, Department of Laboratory Medicine, Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, Bengbu, China. EM liyao@bbmc.edu.cn CR Siegel RL, Miller KD, Fuchs HE, Jemal A Cancer statistics. CA Cancer J Clin, 2022, 72(1):7–33., DOI 10.3322/caac.21708 Heimbach JK, Kulik LM, Finn RS, Sirlin CB, Abecassis MM, Roberts LR, et al. AASLD Guidelines for the Treatment of Hepatocellular Carcinoma. Hepatology, 2018, 67:358–80., DOI 10.1002/hep.29086 Pandey UB, Nie Z, Batlevi Y, McCray BA, Ritson GP, Nedelsky NB, et al. HDAC6 Rescues Neurodegeneration and Provides an Essential Link between Autophagy and the UPS. Nature, 2007, 14447:859–63., DOI 10.1038/ nature05853 Li W, Tu D, Brunger AT, Ye Y. A Ubiquitin Ligase Transfers Preformed Polyubiquitin Chains from a Conjugating Enzyme to a Substrate. Nature, 2007, 446:333–7., DOI 10.1038/nature05542 Saurabh K, Shah PP, Doll MA, Siskind LJ, Beverly LJ. UBR-Box Containing Protein, UBR5, Is Over-expressed in Human Lung Adenocarcinoma and Is a Potential Therapeutic Target. BMC Cancer, 2020, 20:824., DOI 10.1186/s12885- 020-07322-1 Hu Y, Parvin JD. Small Ubiquitin-like Modifier, SUMO, Isoforms and Conjugation-independent Function in DNA Double-Strand Break Repair Pathways. J Biol Chem, 2014, 289:21289–95., DOI 10.1074/jbc.C114.582122 Sy SM, Jiang J, Deng Y, Huen MS. The Ubiquitin Specific Protease USP34 Promotes Ubiquitin Signaling at DNA Double-Strand Breaks. Nucleic Acids Res, 2013, 41:8572–80., DOI 10.1093/nar/gkt622 Hu G, Wang X, Saunders DN, Henderson M, Russell AJ, Herring BP, et al. Modulation of Myocardin Function by the Ubiquitin E3 Ligase UBR5. J Biol Chem, 2010, 285:11800–9., DOI 10.1074/jbc.M109.079384 Song M, Yeku OO, Rafiq S, Purdon T, Dong X, Zhu L, et al. Tumor Derived UBR5 Promotes Ovarian Cancer Growth and Metastasis through Inducing Immunosuppressive Macrophages. Nat Commun, 2020, 11:6298., DOI 10.1038/ s41467-020-20140-0 Shearer RF, Iconomou M, Watts CK, Saunders DN. Functional Roles of the E3 Ubiquitin Ligase UBR5 in Cancer. Mol Cancer Res, 2015, 13:1523–32., DOI 10.1158/1541-7786.MCR-15-0383 Ding F, Zhu X, Song X, Yuan P, Ren L, Chai C, et al. UBR5 Oncogene as an Indicator of Poor Prognosis in Gastric Cancer. Exp Ther Med, 2020, 20:7., DOI 10.3892/etm.2020.9135 Qiao X, Liu Y, Prada ML, Mohan AK, Gupta A, Jaiswal A, et al. UBR5 Is Coamplified with MYC in Breast Tumors and Encodes an Ubiquitin Ligase that Limits MYC-dependent Apoptosis. Cancer Res, 2020, 80:1414–27., DOI 10. 1158/0008-5472.CAN-19-1647 Swenson SA, Gilbreath TJ, Vahle H, Hynes-Smith RW, Graham JH, Law HC, et al. UBR5 HECT Domain Mutations Identified in Mantle Cell Lymphoma Control Maturation of B Cells. Blood, 2020, 136:299–312., DOI 10.1182/blood. 2019002102 Gan Y, Ye F, He XX. The Role of YWHAZ in Cancer: AMaze of Opportunities and Challenges. J Cancer, 2020, 11:2252–64., DOI 10.7150/jca.41316 Wang W, Wei C. Advances in the Early Diagnosis of Hepatocellular Carcinoma. Genes Dis, 2020, 7:308–19., DOI 10.1016/j.gendis.2020.01.014 Akoad ME, Pomfret EA. Surgical Resection and Liver Transplantation for Hepatocellular Carcinoma. Clin Liver Dis, 2015, 19:381–99., DOI 10.1016/j.cld. 2015.01.007 Akateh C, Black SM, Conteh L, Miller ED, Noonan A, Elliott E, et al. Neoadjuvant and Adjuvant Treatment Strategies for Hepatocellular Carcinoma. World J Gastroenterol, 2019, 25:3704–21., DOI 10.3748/wjg.v25.i28.3704 Wei X, Jiang Y, Zhang X, Feng S, Zhou B, Ye X, et al. Neoadjuvant Three- Dimensional Conformal Radiotherapy for Resectable Hepatocellular Carcinoma with Portal Vein Tumor Thrombus: A Randomized, Open- Label, Multicenter Controlled Study. J Clin Oncol, 2019, 37:2141–51., DOI 10.1200/JCO.18.02184 Lee JH, Lee JH, Lim YS, Yeon JE, Song TJ, Yu SJ, et al. Adjuvant Immunotherapy with Autologous Cytokine-Induced Killer Cells for Hepatocellular Carcinoma. Gastroenterology, 2015, 148:1383–91., DOI 10. 1053/j.gastro.2015.02.055 Tang QH, Li AJ, YangGM, Lai EC, ZhouWP, Jiang ZH, et al. Surgical Resection versus Conformal Radiotherapy Combined with TACE for Resectable Hepatocellular Carcinoma with portal Vein Tumor Thrombus: a Comparative Study. World J Surg, 2013, 37:1362–70., DOI 10.1007/s00268-013-1969-x Tella SH, Kommalapati A, Mahipal A. Systemic Therapy for Advanced Hepatocellular Carcinoma: Targeted Therapies. Chin Clin Oncol, 2021, 10: 10., DOI 10.21037/cco-20-117 Choi WS, Jeong BC, Joo YJ, Lee MR, Kim J, Eck MJ, et al. Structural Basis for the Recognition of N-End Rule Substrates by the UBR Box of Ubiquitin Ligases. Nat Struct Mol Biol, 2010, 17:1175–81., DOI 10.1038/nsmb.1907 Tasaki T, Zakrzewska A, Dudgeon DD, Jiang Y, Lazo JS, Kwon YT. The Substrate Recognition Domains of the N-End Rule Pathway. J Biol Chem, 2009, 284:1884–95., DOI 10.1074/jbc.M803641200 Liu Y, Liu C, Dong W, Li W. Physiological Functions and Clinical Implications of the N-End Rule Pathway. Front Med, 2016, 10:258–70., DOI 10.1007/s11684- 016-0458-7 Li CG, Mahon C, Sweeney NM, Verschueren E, Kantamani V, Li D, et al. PPARγ Interaction with UBR5/ATMIN Promotes DNA Repair to Maintain Endothelial Homeostasis. Cell Rep, 2019, 26:1333–43., DOI 10.1016/j.celrep. 2019.01.013 Fuja TJ, Lin F, Osann KE, Bryant PJ. Somatic Mutations and Altered Expression of the Candidate Tumor Suppressors CSNK1 Epsilon, DLG1, and EDD/hHYD in Mammary Ductal Carcinoma. Cancer Res, 2004, 64: 942–51., DOI 10.1158/0008-5472.can-03-2100 Liao L, Song M, Li X, Tang L, Zhang T, Zhang L, et al. E3 Ubiquitin Ligase UBR5 Drives the Growth and Metastasis of Triple-Negative Breast Cancer. Cancer Res, 2017, 77:2090–101., DOI 10.1158/0008-5472.CAN-16-2409 Chen L, Yuan R, Wen C, Liu T, Feng Q, Deng X, et al. E3 Ubiquitin Ligase UBR5 Promotes Pancreatic Cancer Growth and Aerobic Glycolysis by Downregulating FBP1 via Destabilization of C/EBPα. Oncogene, 2021, 40: 262–76., DOI 10.1038/s41388-020-01527-1 Tang Y, Lv P, Sun Z, Han L, Luo B, Zhou W. 14-3-3ζ Up-Regulates Hypoxia- Inducible Factor-1α in Hepatocellular Carcinoma via Activation of PI3K/Akt/ NF-lB Signal Transduction Pathway. Int J Clin Exp Pathol, 2015, 8:15845–53. Du G, Yu X, Chen Y, Cai W. MiR-1-3p Suppresses Colorectal Cancer Cell Proliferation and Metastasis by Inhibiting YWHAZ-Mediated Epithelial- Mesenchymal Transition. Front Oncol, 2021, 11:634596., DOI 10.3389/fonc. 2021.634596 Deng Y, Zheng J, Ma J. The Clinical and Prognostic Significance of YWHAZ in Non-small-cell Lung Cancer Patients: Immunohistochemical Analysis. J Cel Biochem, 2019, 120:6290–8., DOI 10.1002/jcb.27915 Mei J, Liu Y, Yu X, Hao L, Ma T, Zhan Q, et al. YWHAZ Interacts with DAAM1 to Promote Cell Migration in Breast Cancer. Cell Death Discov, 2021, 7:221., DOI 10.1038/s41420-021-00609-7 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610396 EP 1610406 DI 10.3389/pore.2022.1610396 PG 11 ER PT J TI Overexpression of Dehydrogenase/ Reductase 9 Predicts Poor Response to Concurrent Chemoradiotherapy and Poor Prognosis in Rectal Cancer Patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; rectal cancer; DHRS9; epithelial cell differentiation; keratan sulfate; mucin ID biomarker; rectal cancer; DHRS9; epithelial cell differentiation; keratan sulfate; mucin AB Objective: To reduce the risk of locoregional recurrence, the addition of neoadjuvant concurrent chemoradiotherapy (CCRT) is recommended before surgical management for rectal cancer patients. However, despite identical tumor histology, individual patient response to neoadjuvant CCRT varies greatly. Accordingly, a comprehensive molecular characterization that is used to predict CCRT efficacy is instantly needed. Methods: Pearson’s chi-squared test was utilized to correlate dehydrogenase/reductase 9 (DHRS9) expression with clinicopathological features. Survival curves were created applying the Kaplan-Meier method, and the log-rank test was conducted to compare prognostic utility between high and low DHRS9 expression groups. Multivariate Cox proportional hazards regression analysis was applied to identify independent prognostic biomarkers based on variables with prognostic utility at the univariate level. Results: Utilizing a public transcriptome dataset, we identified that the DHRS9 gene is the most considerably upregulated gene related to epithelial cell differentiation (GO: 0030855) among rectal cancer patients with CCRT resistance. Employing immunohistochemical staining, we also demonstrated that high DHRS9 immunoexpression is considerably associated with an aggressive clinical course and CCRT resistance in our rectal cancer cohort. Among all variables with prognostic utility at the univariate level, only high DHRS9 immunoexpression was independently unfavorably prognostic of all three endpoints (all p ≤0.048) in the multivariate analysis. In addition, applying bioinformatic analysis, we also linked DHRS9 with unrevealed functions, such as keratan sulfate and mucin synthesis which may be implicated in CCRT resistance. Conclusion: Altogether, DHRS9 expression may serve as a helpful predictive and prognostic biomarker and assist decision-making for rectal cancer patients who underwent neoadjuvant CCRT. CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Keller DS, Berho M, Perez RO, Wexner SD, Chand M. The Multidisciplinary Management of Rectal Cancer. Nat Rev Gastroenterol Hepatol, 2020, 17(7): 414–29., DOI 10.1038/s41575-020-0275-y Fokas E, Glynne-Jones R, Appelt A, Beets-Tan R, Beets G, Haustermans K, et al. Outcome Measures in Multimodal Rectal Cancer Trials. Lancet Oncol, 2020, 21(5):e252–e64., DOI 10.1016/S1470-2045(20)30024-3 Dworak O, Keilholz L, Hoffmann A. Pathological Features of Rectal Cancer after Preoperative Radiochemotherapy. Int J Colorectal Dis, 1997, 12(1): 19–23., DOI 10.1007/s003840050072 Clevers H. The Intestinal Crypt, a Prototype Stem Cell Compartment. Cell, 2013, 154(2):274–84., DOI 10.1016/j.cell.2013.07.004 Allaire JM, Crowley SM, Law HT, Chang SY, Ko HJ, Vallance BA. The Intestinal Epithelium: Central Coordinator of Mucosal Immunity. Trends Immunol, 2018, 39(9):677–96., DOI 10.1016/j.it.2018.04.002 Van der Sluis M, De Koning BA, De Bruijn AC, Velcich A, Meijerink JP, Van Goudoever JB, et al. Muc2-deficient Mice Spontaneously Develop Colitis, Indicating that MUC2 Is Critical for Colonic protection. Gastroenterology, 2006, 131(1):117–29., DOI 10.1053/j.gastro.2006.04.020 Betge J, Schneider NI, Harbaum L, Pollheimer MJ, Lindtner RA, Kornprat P, et al. MUC1, MUC2, MUC5AC, and MUC6 in Colorectal Cancer: Expression Profiles and Clinical Significance. Virchows Arch, 2016, 469(3):255–65., DOI 10. 1007/s00428-016-1970-5 Kasprzak A, Siodla E, Andrzejewska M, Szmeja J, Seraszek-Jaros A, Cofta S, et al. Differential Expression of Mucin 1 and Mucin 2 in Colorectal Cancer. World J Gastroenterol, 2018, 24(36):4164–77., DOI 10.3748/wjg.v24.i36.4164 Chou CL, Chen TJ, Tian YF, Chan TC, Yeh CF, LiWS, et al. UpregulatedMUC2 Is an Unfavorable Prognostic Indicator for Rectal Cancer Patients Undergoing Preoperative CCRT. J Clin Med, 2021, 10(14):3030., DOI 10.3390/jcm10143030 Derwinger K, Kodeda K, Bexe-Lindskog E, Taflin H. Tumour Differentiation Grade Is Associated with TNM Staging and the Risk of Node Metastasis in Colorectal Cancer. Acta Oncol, 2010, 49(1):57–62., DOI 10.3109/ 02841860903334411 Kim SC, Shin YK, Kim YA, Jang SG, Ku JL. Identification of Genes Inducing Resistance to Ionizing Radiation in Human Rectal Cancer Cell Lines: Resensitization of Radio-Resistant Rectal Cancer Cells through Down Regulating NDRG1. BMC cancer, 2018, 18(1):594., DOI 10.1186/s12885-018-4514-3 Kropotova ES, Zinovieva OL, Zyryanova AF, Dybovaya VI, Prasolov VS, Beresten SF, et al. Altered Expression of Multiple Genes Involved in Retinoic Acid Biosynthesis in Human Colorectal Cancer. Pathol Oncol Res, 2014, 20(3): 707–17., DOI 10.1007/s12253-014-9751-4 Hu L, Chen HY, Han T, Yang GZ, Feng D, Qi CY, et al. Downregulation of DHRS9 Expression in Colorectal Cancer Tissues and its Prognostic Significance. Tumour Biol, 2016, 37(1):837–45., DOI 10.1007/s13277-015-3880-6 Chen TJ, Chou CL, Tian YF, Yeh CF, Chan TC, He HL, et al. High FRMD3 Expression Is Prognostic for Worse Survival in Rectal Cancer Patients Treated with CCRT. Int J Clin Oncol, 2021, 26(9):1689–97., DOI 10. 1007/s10147-021-01944-6 Budwit-Novotny DA, McCarty KS, Cox EB, Soper JT, Mutch DG, Creasman WT, et al. Immunohistochemical Analyses of Estrogen Receptor in Endometrial Adenocarcinoma Using a Monoclonal Antibody. Cancer Res, 1986, 46(10):5419–25. Stahl M, Tallman MS. Acute Promyelocytic Leukemia, APL): Remaining Challenges towards a Cure for All. Leuk Lymphoma, 2019, 60(13):3107–15., DOI 10.1080/10428194.2019.1613540 Siddikuzzaman GC, Berlin Grace VM, Berlin Grace VM. All Trans Retinoic Acid and Cancer. Immunopharmacol Immunotoxicol, 2011, 33(2):241–9., DOI 10.3109/08923973.2010.521507 Shimomura H, Sasahira T, Nakashima C, Shimomura-Kurihara M, Kirita T. Downregulation of DHRS9 Is Associated with Poor Prognosis in Oral Squamous Cell Carcinoma. Pathology, 2018, 50(6):642–7., DOI 10.1016/j. pathol.2018.06.002 Li HB, Zhou J, Zhao F, Yu J, Xu L. Prognostic Impact of DHRS9 Overexpression in Pancreatic Cancer. Cancer Manag Res, 2020, 12:5997–6006., DOI 10.2147/CMAR.S251897 Pomin VH, Mulloy B. Glycosaminoglycans and Proteoglycans. Pharmaceuticals, Basel, Switzerland,, 2018, 11(1):E27., DOI 10.3390/ ph11010027 Pomin VH. Keratan Sulfate: an Up-To-Date Review. Int J Biol Macromol, 2015, 72:282–9., DOI 10.1016/j.ijbiomac.2014.08.029 Henke E, Nandigama R, Ergun S. Extracellular Matrix in the Tumor Microenvironment and its Impact on Cancer Therapy. Front Mol Biosci, 2019, 6:160., DOI 10.3389/fmolb.2019.00160 Raman R, Sasisekharan V, Sasisekharan R. Structural Insights into Biological Roles of Protein-Glycosaminoglycan Interactions. Chem Biol, 2005, 12(3): 267–77., DOI 10.1016/j.chembiol.2004.11.020 Kakizaki I, Kobayashi T, Tamura S, Akagi H, Takagaki K. Effect of Glycosaminoglycan Structure on All-Trans-Retinoic Acid-Induced Neural Differentiation of P19 Embryonal Carcinoma Cells. Biochem Biophys Res Commun, 2021, 570:169–74., DOI 10.1016/j.bbrc.2021.07.012 Vitale D, Kumar Katakam S, Greve B, Jang B, Oh ES, Alaniz L, et al. Proteoglycans and Glycosaminoglycans as Regulators of Cancer Stem Cell Function and Therapeutic Resistance. FEBS J, 2019, 286(15):2870–82., DOI 10. 1111/febs.14967 Wu N, Silva LM, Liu Y, Zhang Y, Gao C, Zhang F, et al. Glycan Markers of Human Stem Cells Assigned with Beam Search Arrays. Mol Cell Proteomics, 2019, 18(10):1981–2002., DOI 10.1074/mcp.RA119.001309 Very N, Lefebvre T, El Yazidi-Belkoura I. Drug Resistance Related to Aberrant Glycosylation in Colorectal Cancer. Oncotarget, 2018, 9(1):1380–402., DOI 10. 18632/oncotarget.22377 Caterson B, Melrose J. Keratan Sulfate, a Complex Glycosaminoglycan with Unique Functional Capability. Glycobiology, 2018, 28(4):182–206., DOI 10. 1093/glycob/cwy003 Hayes AJ, Melrose J. Keratan Sulphate in the Tumour Environment. Adv Exp Med Biol, 2020, 1245:39–66., DOI 10.1007/978-3-030-40146-7_2 Iwai T, Inaba N, Naundorf A, Zhang Y, Gotoh M, Iwasaki H, et al. Molecular Cloning and Characterization of a Novel UDP-GlcNAc:GalNAc-Peptide Beta1, 3-N-Acetylglucosaminyltransferase, Beta 3Gn-T6), an Enzyme Synthesizing the Core 3 Structure of O-Glycans. J Biol Chem, 2002, 277(15):12802–9., DOI 10.1074/jbc.M112457200 Kitayama K, Hayashida Y, Nishida K, Akama TO. Enzymes Responsible for Synthesis of Corneal Keratan Sulfate Glycosaminoglycans. J Biol Chem, 2007, 282(41):30085–96., DOI 10.1074/jbc.M703695200 Lee JK, Bistrup A, van Zante A, Rosen SD. Activities and Expression Pattern of the Carbohydrate Sulfotransferase GlcNAc6ST-3, I-GlcNAc6ST): Functional Implications. Glycobiology, 2003, 13(4):245–54., DOI 10.1093/glycob/cwg018 Brockhausen I. Sulphotransferases Acting on Mucin-type Oligosaccharides. Biochem Soc Trans, 2003, 31(2):318–25., DOI 10.1042/bst0310318 Bonnans C, Chou J,Werb Z. Remodelling the ExtracellularMatrix in Development and Disease. Nat Rev Mol Cell Biol, 2014, 15(12):786–801., DOI 10.1038/nrm3904 Shan M, Gentile M, Yeiser JR, Walland AC, Bornstein VU, Chen K, et al. Mucus Enhances Gut Homeostasis and Oral Tolerance by Delivering Immunoregulatory Signals. Science, New York, NY,, 2013, 342(6157): 447–53., DOI 10.1126/science.1237910 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610537 EP 1610546 DI 10.3389/pore.2022.1610537 PG 10 ER PT J AU Li, JJ Wang, Sh Guan, ZN Zhang, JX Zhan, RX Zhu, JL AF Li, Jin-Jin Wang, Shuai Guan, Zhong-Ning Zhang, Jin-Xi Zhan, Ri-Xin Zhu, Jian-Long TI Anterior Gradient 2 is a Significant Prognostic Biomarker in Bone Metastasis of Breast Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; bone metastasis; DEGs; BRCA; AGR2 ID prognosis; bone metastasis; DEGs; BRCA; AGR2 AB Background: The study aimed to detect DEGs associated with BRCA bone metastasis, filter prognosis biomarkers, and explore possible pathways. Methods: GSE175692 dataset was used to detect DEGs between BRCA bone metastatic cases and non-bone metastatic cases, followed by the construction of a PPI network among DEGs. The main module among the PPI network was then determined and pathway analysis on genes within the module was performed. Through performing Cox regression, Kaplan-Meier, nomogram, and ROC curve analyses using GSE175692 and GSE124647 datasets at the same time, the most significant prognostic biomarker was gradually filtered. Finally, important pathways associated with prognostic biomarkers were explored by GSEA analysis. Results: The 74 DEGs were detected between bone metastasis and non-bone metastasis groups. A total of 15 nodes were included in the main module among the whole PPI network and they mainly correlated with the IL-17 signaling pathway. We then performed Cox analysis on 15 genes using two datasets and only enrolled the genes with p < 0.05 in Cox analysis into the further analyses. Kaplan-Meier analyses using two datasets showed that the common biomarker AGR2 expression was related to the survival time ofBRCAmetastatic cases. Further, the nomogram determined the greatest contribution of AGR2 on the survival probability and the ROC curve revealed its optimal prognostic performance. More importantly, high expression of AGR2 prolonged the survival time of BRCA bone metastatic patients. These results all suggested the importance of AGR2 in metastatic BRCA. Finally, we performed the GSEA analysis and found that AGR2 was negatively related to IL-17 and NF-kβ signaling pathways. Conclusion: AGR2 was finally determined as the most important prognostic biomarker in BRCA bone metastasis, and it may play a vital role in cancer progression by regulating IL- 17 and NF-kB signaling pathways. C1 [Li, Jin-Jin] Hangzhou Ninth People’s Hospital, Department of OrthopaedicsHangzhou, China. [Wang, Shuai] Hangzhou Ninth People’s Hospital, Department of PathologyHangzhou, China. [Guan, Zhong-Ning] Hangzhou Ninth People’s Hospital, Department of OrthopaedicsHangzhou, China. [Zhang, Jin-Xi] Hangzhou Ninth People’s Hospital, Department of OrthopaedicsHangzhou, China. [Zhan, Ri-Xin] Hangzhou Ninth People’s Hospital, Department of Medical Record ManagementHangzhou, China. [Zhu, Jian-Long] Hangzhou Ninth People’s Hospital, Department of OrthopaedicsHangzhou, China. RP Zhu, JL (reprint author), Hangzhou Ninth People’s Hospital, Department of Orthopaedics, Hangzhou, China. EM XSSYZJL@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/caac.21492 Weigelt B, Peterse JL, van ’t Veer LJ. Breast Cancer Metastasis: Markers and Models. Nat Rev Cancer, 2005, 5(8):591–602., DOI 10.1038/nrc1670 Brockton NT, Gill SJ, Laborge SL, Paterson AH, Cook LS, Vogel HJ, et al. The Breast Cancer to Bone, B2B, Metastases Research Program: a Multi- Disciplinary Investigation of Bone Metastases from Breast Cancer. BMC cancer, 2015, 15:512., DOI 10.1186/s12885-015-1528-y Fidler IJ. The Pathogenesis of Cancer Metastasis: the ’seed and Soil’ Hypothesis Revisited. Nat Rev Cancer, 2003, 3(6):453–8., DOI 10.1038/ nrc1098 Body JJ, Quinn G, Talbot S, Booth E, Demonty G, Taylor A, et al. Systematic Review and Meta-Analysis on the Proportion of Patients with Breast Cancer Who Develop Bone Metastases. Crit Rev Oncol Hematol, 2017, 115:67–80., DOI 10.1016/j.critrevonc.2017.04.008 Li X, Liang Y, Lian C, Peng F, Xiao Y, He Y, et al. CST6 Protein and Peptides Inhibit Breast Cancer Bone Metastasis by Suppressing CTSB Activity and Osteoclastogenesis. Theranostics, 2021, 11(20):9821–32., DOI 10.7150/thno. 62187 Coleman RE. Clinical Features of Metastatic Bone Disease and Risk of Skeletal Morbidity. Clin Cancer Res, 2006, 12(20):6243s–9s., DOI 10.1158/1078-0432. CCR-06-0931 Hankins ML, Smith CN, Hersh B, Heim T, Belayneh R, Dooley S, et al. Prognostic Factors and Survival of Patients Undergoing Surgical Intervention for Breast Cancer Bone Metastases. J Bone Oncol, 2021, 29:100363., DOI 10. 1016/j.jbo.2021.100363 Domschke C, Schuetz F. Side Effects of Bone-Targeted Therapies in Advanced Breast Cancer. Breast care, Basel, Switzerland,, 2014, 9(5):332–6., DOI 10.1159/ 000368844 Steeg PS. Targeting Metastasis. Nat Rev Cancer, 2016, 16(4):201–18., DOI 10. 1038/nrc.2016.25 Kozlowski J, Kozlowska A, Kocki J. Breast Cancer Metastasis - Insight into Selected Molecular Mechanisms of the Phenomenon. Postepy Hig Med Dosw, 2015, 69:447–51., DOI 10.5604/17322693.1148710 Wang L, Wang W, Zeng S, Zheng H, Lu Q. Construction and Validation of a 6- gene Nomogram Discriminating Lung Metastasis Risk of Breast Cancer Patients. PloS one, 2020, 15(12):e0244693., DOI 10.1371/journal.pone.0244693 Lu X, Gao C, Liu C, Zhuang J, Su P, Li H, et al. Identification of the Key Pathways and Genes Involved in HER2-Positive Breast Cancer with Brain Metastasis. Pathol Res Pract, 2019, 215(8):152475., DOI 10.1016/j.prp.2019. 152475 Tang D, Zhao X, Zhang L, Wang Z, Wang C. Identification of Hub Genes to Regulate Breast Cancer Metastasis to Brain by Bioinformatics Analyses. J Cell Biochem, 2019, 120(6):9522–31., DOI 10.1002/jcb.28228 Zhang Y, Huang X, Liu J, Chen G, Liu C, Zhang S, et al. New Insight into Long Non-coding RNAs Associated with Bone Metastasis of Breast Cancer Based on an Integrated Analysis. Cancer Cell Int, 2021, 21(1):372., DOI 10.1186/s12935- 021-02068-7 Spadazzi C, Mercatali L, Esposito M, Wei Y, Liverani C, De Vita A, et al. Trefoil Factor-1 Upregulation in Estrogen-Receptor Positive Breast Cancer Correlates with an Increased Risk of Bone Metastasis. Bone, 2021, 144:115775., DOI 10. 1016/j.bone.2020.115775 Haider M, Ridlmaier N, Smit D, Taipaleenmaki H. Interleukins as Mediators of the Tumor Cell-Bone Cell Crosstalk during the Initiation of Breast Cancer Bone Metastasis. Int J Mol Sci, 2021, 22(6):2898., DOI 10. 3390/ijms22062898 van ’t Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, et al. Gene Expression Profiling Predicts Clinical Outcome of Breast Cancer. Nature, 2002, 415(6871):530–6., DOI 10.1038/415530a Roy LD, Ghosh S, Pathangey LB, Tinder TL, Gruber HE, Mukherjee P. Collagen Induced Arthritis Increases Secondary Metastasis in MMTV-PyV MT Mouse Model of Mammary Cancer. BMC cancer, 2011, 11:365., DOI 10. 1186/1471-2407-11-365 Roy LD, Sahraei M, Schettini JL, Gruber HE, Besmer DM, Mukherjee P. Systemic Neutralization of IL-17A Significantly Reduces Breast Cancer Associated Metastasis in Arthritic Mice by Reducing CXCL12/SDF- 1 Expression in the Metastatic Niches. BMC cancer, 2014, 14:225., DOI 10. 1186/1471-2407-14-225 Fabre JAS, Giustinniani J, Garbar C, Merrouche Y, Antonicelli F, Bensussan A. The Interleukin-17 Family of Cytokines in Breast Cancer. Int J Mol Sci, 2018, 19(12):E3880., DOI 10.3390/ijms19123880 Jiang P, Gao W, Ma T, Wang R, Piao Y, Dong X, et al. CD137 Promotes Bone Metastasis of Breast Cancer by Enhancing the Migration and Osteoclast Differentiation of Monocytes/macrophages. Theranostics, 2019, 9(10): 2950–66., DOI 10.7150/thno.29617 Arciero CA, Guo Y, Jiang R, Behera M, O’Regan R, Peng L, et al. ER(+)/ HER2(+, Breast Cancer Has Different Metastatic Patterns and Better Survival Than ER(-)/HER2(+, Breast Cancer. Clin Breast Cancer, 2019, 19(4):236–45., DOI 10.1016/j.clbc.2019.02.001 Yu C-c, Wu Y-n, Hu K-m, Zhang S-z. Prominin 1 Significantly Correlated with Bone Metastasis of Breast Cancer and Influenced the Patient’s Prognosis. Biomed Res Int, 2022, 2022:4123622., DOI 10.1155/2022/4123622 Gray TA, MacLaine NJ, Michie CO, Bouchalova P, Murray E, Howie J, et al. Anterior Gradient-3: a Novel Biomarker for Ovarian Cancer that Mediates Cisplatin Resistance in Xenograft Models. J Immunol Methods, 2012, 378(1- 2):20–32., DOI 10.1016/j.jim.2012.01.013 Smid M, Wang Y, Klijn J, Sieuwerts A, Zhang Y, Atkins D, et al. Genes Associated with Breast Cancer Metastatic to Bone. J Clin Oncol, 2006, 24(15): 2261–7., DOI 10.1200/JCO.2005.03.8802 Guo J, Gong G, Zhang B. Identification and Prognostic Value of Anterior Gradient Protein 2 Expression in Breast Cancer Based on Tissue Microarray. Tumour Biol, 2017, 39(7):1010428317713392., DOI 10.1177/ 1010428317713392 Brychtova V, Vojtesek B, Hrstka R. Anterior Gradient 2: a Novel Player in Tumor Cell Biology. Cancer Lett, 2011, 304(1):1–7., DOI 10.1016/j.canlet.2010.12.023 Salmans ML, Zhao F, Andersen B. The Estrogen-Regulated Anterior Gradient 2, AGR2, Protein in Breast Cancer: a Potential Drug Target and Biomarker. Breast Cancer Res, 2013, 15(2):204., DOI 10.1186/bcr3408 Lacambra MD, Tsang JY, Ni YB, Chan SK, Tan PH, Tse GM. Anterior Gradient 2 Is a Poor Outcome Indicator in Luminal Breast Cancer. Ann Surg Oncol, 2015, 22(11):3489–96., DOI 10.1245/s10434-015-4420-8 Yenmis G, Yaprak Sarac E, Besli N, Soydas T, Tastan C, Dilek Kancagi D, et al. Anti-cancer Effect of Metformin on the Metastasis and Invasion of Primary Breast Cancer Cells throughMediating NF-kB Activity. Acta Histochem, 2021, 123(4):151709., DOI 10.1016/j.acthis.2021.151709 Zhang Z, Yao Y, Yuan Q, Lu C, Zhang X, Yuan J, et al. Gold Clusters Prevent Breast Cancer Bone Metastasis by Suppressing Tumor-Induced Osteoclastogenesis. Theranostics, 2020, 10(9):4042–55., DOI 10.7150/thno. 42218 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610538 EP 1610550 DI 10.3389/pore.2022.1610538 PG 13 ER PT J AU Bencze, E Bogos, K Kohanka, A Bathory-Fulop, L Sarosi, V Csernak, E Bittner, N Melegh, Zs Toth, E AF Bencze, Eszter Bogos, Krisztina Kohanka, Andrea Bathory-Fulop, Laszlo Sarosi, Veronika Csernak, Erzsebet Bittner, Nora Melegh, Zsombor Toth, Erika TI EGFR T790M Mutation Detection in Patients With Non-Small Cell Lung Cancer After First Line EGFR TKI Therapy: Summary of Results in a Three-Year Period and a Comparison of Commercially Available Detection Kits SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE liquid biopsy; NSCLC; ctDNA; EGFR T790M mutation; AmoyDx super-ARMS EGFR mutation detection kit; cobas EGFR test v2 ID liquid biopsy; NSCLC; ctDNA; EGFR T790M mutation; AmoyDx super-ARMS EGFR mutation detection kit; cobas EGFR test v2 AB EGFR mutation in non-small cell lung cancer (NSCLC) offers a potential therapeutic target for tyrosine kinase inhibitor (TKI) therapy. The majority of these cases, however eventually develop therapy resistance, mainly by acquiring EGFR T790M mutation. Recently, thirdgeneration TKIs have been introduced to overcome T790M mutation-related resistance. Cell free circulating tumor DNA (liquid biopsy) has emerged as a valuable alternative method for T790M mutation detection during patient follow up, when a tissue biopsy cannot be obtained for analysis. In this study, we summarized our experience with Super- ARMS EGFR Mutation Detection Kit (AmoyDx) on 401 samples of 242 NSCLC patients in a 3-year period in Hungary, comprising 364 plasma and 37 non-plasma samples. We also compared the performance of two commercially available detection kits, the cobas EGFR Mutation test v2 (Roche) and the Super-ARMS EGFR Mutation Detection Kit (AmoyDx). The same activating EGFR mutation was detected with the AmoyDx kit as in the primary tumor in 45.6% of the samples. T790M mutation was identified in 48.1% of the samples containing activating EGFR mutation. The detection rate of T790M mutation was not dependent on the DNA concentration of the plasma sample and there was no considerable improvement in mutation detection rate after a second, subsequent plasma sample. The concordance of EGFR activating mutation detection was 89% between the two methods, while this was 93% for T790M mutation detection. The AmoyDx kit, however showed an overall higher detection rate of T790M mutation compared to the cobas kit (p = 0.014). T790M mutation was detected at 29.8% of the patients if only plasma samples were available for analysis, while the detection rate was 70.2% in non-plasma samples. If the activating EGFR was detected in the plasma samples, the detection rate of T790M mutation was 42.4%. Although non-plasma samples provided a superior T790M mutation detection rate, we found that liquid biopsy can offer a valuable tool for T790M mutation detection, when a tissue biopsy is not available. Alternatively, a liquid biopsy can be used as a screening test, when re-biopsy should be considered in case of wild-type results. C1 [Bencze, Eszter] National Institute of Oncology, National Tumour Biology Laboratory, Department of Surgical and Molecular PathologyBudapest, Hungary. [Bogos, Krisztina] National Koranyi Institute of PulmonologyBudapest, Hungary. [Kohanka, Andrea] National Institute of Oncology, National Tumour Biology Laboratory, Department of Surgical and Molecular PathologyBudapest, Hungary. [Bathory-Fulop, Laszlo] National Institute of Oncology, National Tumour Biology Laboratory, Department of Surgical and Molecular PathologyBudapest, Hungary. [Sarosi, Veronika] University of Pecs, Faculty of MedicinePecs, Hungary. [Csernak, Erzsebet] National Institute of Oncology, National Tumour Biology Laboratory, Department of Surgical and Molecular PathologyBudapest, Hungary. [Bittner, Nora] Semmelweis University, Department of OncologyBudapest, Hungary. [Melegh, Zsombor] National Institute of Oncology, National Tumour Biology Laboratory, Department of Surgical and Molecular PathologyBudapest, Hungary. [Toth, Erika] National Institute of Oncology, National Tumour Biology Laboratory, Department of Surgical and Molecular PathologyBudapest, Hungary. RP Toth, E (reprint author), National Institute of Oncology, National Tumour Biology Laboratory, Department of Surgical and Molecular Pathology, Budapest, Hungary. EM dr.toth.erika@oncol.hu CR Bogos K, Kiss Z, Galffy G, Tamasi L, Ostoros G, Muller V, et al. Lung Cancer in Hungary. J Thorac Oncol, 2020, 15(5):692–9., DOI 10.1016/j.jtho.2019.11.001 Graham RP, Treece AL, Lindeman NI, Vasalos P, Shan M, Jennings LJ, et al. Worldwide Frequency of Commonly Detected EGFR Mutations. Arch Pathol Lab Med, 2018, 142(2):163–7., DOI 10.5858/arpa.2016-0579-CP Foster SA, Whalen DM, Ozen A, Wongchenko MJ, Yin J, Yen I, et al. Activation Mechanism of Oncogenic Deletion Mutations in BRAF, EGFR, and HER2. Cancer Cell, 2016, 29(4):477–93., DOI 10.1016/j.ccell.2016.02.010 Morgillo F, Della Corte CM, Fasano M, Ciardiello F. Mechanisms of Resistance to EGFR-Targeted Drugs: Lung Cancer. ESMO Open, 2016, 1(3):e000060., DOI 10.1136/esmoopen-2016-000060 Tumbrink HL, Heimsoeth A, Sos ML. The Next Tier of EGFR Resistance Mutations in Lung Cancer. Oncogene, 2021, 40(1):1–11., DOI 10.1038/s41388- 020-01510-w Yun CH, Mengwasser KE, Toms AV, Woo MS, Greulich H, Wong KK, et al. The T790M Mutation in EGFR Kinase Causes Drug Resistance by Increasing the Affinity for ATP. Proc Natl Acad Sci U S A, 2008, 105(6):2070–5., DOI 10. 1073/pnas.0709662105 Walter AO, Sjin RT, Haringsma HJ, Ohashi K, Sun J, Lee K, et al. Discovery of a Mutant-Selective Covalent Inhibitor of EGFR that Overcomes T790MMediated Resistance in NSCLC. Cancer Discov, 2013, 3(12):1404–15., DOI 10.1158/2159-8290.CD-13-0314 Diaz LA, Jr, Bardelli A. Liquid Biopsies: Genotyping Circulating Tumor DNA. J Clin Oncol, 2014, 32(6):579–86., DOI 10.1200/JCO.2012.45.2011 Fenizia F, De Luca A, Pasquale R, Sacco A, Forgione L, Lambiase M, et al. EGFRMutations in Lung Cancer: from Tissue Testing to Liquid Biopsy. Future Oncol, 2015, 11(11):1611–23., DOI 10.2217/fon.15.23 Wang M, Huang X, Li X, Guo Q, Xu W, Zhao M, et al. Performance Comparison of Commercial Kits for Isolating and Detecting Circulating Tumor DNA. Scand J Clin Lab Invest, 2021, 81(4):276–81., DOI 10.1080/ 00365513.2020.1821394 Ho HL, Huang CC, Ku WH, Ho CL, Lin CH, Yu SL, et al. Liquid Biopsy for Detection of EGFR T790M Mutation in Nonsmall Cell Lung Cancer: An Experience of Proficiency Testing in Taiwan. J Chin Med Assoc, 2019, 82(6): 473–6., DOI 10.1097/JCMA.0000000000000100 Maass KK, Schad PS, Finster AME, Puranachot P, Rosing F, Wedig T, et al. From Sampling to Sequencing: A Liquid Biopsy Pre-analytic Workflow to Maximize Multi-Layer Genomic Information from a Single Tube. Cancers, 2021, 13(12):3002., DOI 10.3390/cancers13123002 Sorber L, Zwaenepoel K, Jacobs J, De Winne K, Goethals S, Reclusa P, et al. Circulating Cell-free DNA and RNA Analysis as Liquid Biopsy: Optimal Centrifugation Protocol. Cancers, 2019, 11(4):E458., DOI 10.3390/ cancers11040458 Oxnard GR, Thress KS, Alden RS, Lawrance R, Paweletz CP, CantariniM, et al. Association between Plasma Genotyping and Outcomes of Treatment with Osimertinib, AZD9291, in Advanced Non-small-cell Lung Cancer. J Clin Oncol, 2016, 34(28):3375–82., DOI 10.1200/JCO.2016.66.7162 Jenkins S, Yang JC, Ramalingam SS, Yu K, Patel S, Weston S, et al. Plasma ctDNA Analysis for Detection of the EGFR T790M Mutation in Patients with Advanced Non-small Cell Lung Cancer. J Thorac Oncol, 2017, 12(7):1061–70., DOI 10.1016/j.jtho.2017.04.003 Li X, Zhou C. Comparison of Cross-Platform Technologies for EGFR T790M Testing in Patients with Non-small Cell Lung Cancer. Oncotarget, 2017, 8(59): 100801–18., DOI 10.18632/oncotarget.19007 Chouaid C, Dujon C, Do P, Monnet I, Madroszyk A, Le Caer H, et al. Feasibility and Clinical Impact of Re-biopsy in Advanced Non Small-Cell Lung Cancer: a Prospective Multicenter Study in a Real-World Setting, GFPC Study 12-01). Lung Cancer, 2014, 86(2):170–3., DOI 10.1016/j.lungcan.2014.08.016 Stockley T, Souza CA, Cheema PK, Melosky B, Kamel-Reid S, Tsao MS, et al. Evidence-based Best Practices for EGFR T790M Testing in Lung Cancer in Canada. Curr Oncol, 2018, 25(2):163–9., DOI 10.3747/co.25.4044 Pereira I, Gaspar C, Pina M, Azevedo I, Rodrigues A. Real-World T790M Mutation Frequency and Impact of Rebiopsy in Patients with EGFR-Mutated Advanced Non-small Cell Lung Cancer. Cureus, 2020, 12(12):e12128., DOI 10. 7759/cureus.12128 Ontario H. Cell-Free Circulating Tumour DNA Blood Testing to Detect EGFR T790M Mutation in People with Advanced Non-small Cell Lung Cancer: A Health Technology Assessment. Ont Health Technol Assess Ser, 2020, 20(5): 1–176. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610607 EP 1610612 DI 10.3389/pore.2022.1610607 PG 6 ER PT J AU Yu, T Yu, Shk Lu, Kh AF Yu, Tao Yu, Shao-kun Lu, Kai-hua TI Comprehensive Molecular Analyses of an SLC Family-Based Model in Stomach Adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunotherapy; prognosis; STAD; solute carrier; amino acid ID immunotherapy; prognosis; STAD; solute carrier; amino acid AB Background: Solute carrier (SLC) family members are crucial in transporting amino acids across membranes. Amino acids are indispensable for both cancer and immune cells. However, the clinical significance of amino acid transporting SLC members in stomach adenocarcinoma (STAD) remains unclear. This study aimed to develop an SLC familybased model to predict the prognosis and the response of STAD patients to immunotherapy. Methods: A total of 1239 tumor cases were obtained from online databases. The training set (n = 371) consisted of RNA sequencing profiles obtained from The Cancer Genome Atlas (TCGA), while those from Gene Expression Omnibus (GEO) were used as the test set. Subsequently, the clinical characteristics and immune profiles were investigated, and potential immunotherapy response prediction values of the model were assessed. Results: Based on the TCGA cohort, an SLC family-based model was developed using multivariate Cox analysis. All tumor cases were stratified into high- and low-risk groups considering the SLC model. High-risk patients had a worse overall survival (OS) than lowrisk patients, consistent with the results of GEO cohorts. Comprehensive analyses revealed that the high-risk group was correlated with aggressiveness-related pathways, whereas the low-risk group had better T helper cell infiltration and stronger immunotherapy response. Compared to the high-risk group, the low-risk group presented increased PD-L1 and tumor mutation burden. Conclusion: This SLC family-based model has the potential to predict the prognosis and immunotherapy outcomes of STAD patients. The survival of patients in the low-risk group was greatly prolonged, and the patients may benefit more from immunotherapy. C1 [Yu, Tao] Nanjing Medical University, Hangzhou First People’s Hospital, Department of OncologyNanjing, China. [Yu, Shao-kun] Nanjing Medical University, Hangzhou First People’s Hospital, Department of OncologyNanjing, China. [Lu, Kai-hua] Nanjing Medical University, Hangzhou First People’s Hospital, Department of OncologyNanjing, China. RP Lu, Kh (reprint author), Nanjing Medical University, Hangzhou First People’s Hospital, Department of Oncology, Nanjing, China. EM lukaihua@njmu.edu.cn CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, et al. Firstline Nivolumab Plus Chemotherapy versus Chemotherapy Alone for Advanced Gastric, Gastro-Oesophageal junction, and Oesophageal Adenocarcinoma, CheckMate 649): a Randomised, Open-Label, Phase 3 Trial. Lancet, 2021, 398:27–40., DOI 10.1016/S0140-6736(21)00797-2 Lin Z, Gu J, Cui X, Huang L, Li S, Feng J, et al. Deciphering Microenvironment of NSCLC Based on CD8+ TIL Density and PD-1/PD-L1 Expression. J Cancer, 2019, 10:211–22., DOI 10.7150/jca.26444 Hensley CT, Wasti AT, DeBerardinis RJ. Glutamine and Cancer: Cell Biology, Physiology, and Clinical Opportunities. J Clin Invest, 2013, 123:3678–84., DOI 10.1172/jci69600 Green CR, Wallace M, Divakaruni AS, Phillips SA, Murphy AN, Ciaraldi TP, et al. Branched-chain Amino Acid Catabolism Fuels Adipocyte Differentiation and Lipogenesis. Nat Chem Biol, 2016, 12:15–21., DOI 10.1038/nchembio.1961 Chung WJ, Lyons SA, Nelson GM, Hamza H, Gladson CL, Gillespie GY, et al. Inhibition of Cystine Uptake Disrupts the Growth of Primary Brain Tumors. J Neurosci, 2005, 25:7101–10., DOI 10.1523/jneurosci.5258-04.2005 Rodriguez PC, Zea AH, Culotta KS, Zabaleta J, Ochoa JB, Ochoa AC. Regulation of T Cell Receptor CD3zeta Chain Expression by L-Arginine. J Biol Chem, 2002, 277:21123–9., DOI 10.1074/jbc.M110675200 Geiger R, Rieckmann JC, Wolf T, Basso C, Feng Y, Fuhrer T, et al. L-arginine Modulates T Cell Metabolism and Enhances Survival and Anti-tumor Activity. Cell, 2016, 167:829–42 e13., DOI 10.1016/j.cell.2016.09.031 Carr EL, Kelman A, Wu GS, Gopaul R, Senkevitch E, Aghvanyan A, et al. Glutamine Uptake and Metabolism Are Coordinately Regulated by ERK/ MAPK during T Lymphocyte Activation. J Immunol, 2010, 185:1037–44., DOI 10.4049/jimmunol.0903586 Klysz D, Tai X, Robert PA, Craveiro M, Cretenet G, Oburoglu L, et al. Glutamine-dependent α-ketoglutarate Production Regulates the Balance between T Helper 1 Cell and Regulatory T Cell Generation. Sci Signal, 2015, 8:ra97., DOI 10.1126/scisignal.aab2610 Kedia-Mehta N, Finlay DK. Competition for Nutrients and its Role in Controlling Immune Responses. Nat Commun, 2019, 10:2123., DOI 10.1038/ s41467-019-10015-4 Sinclair LV, Howden AJ, Brenes A, Spinelli L, Hukelmann JL, Macintyre AN, et al. Antigen Receptor Control of Methionine Metabolism in T Cells. Elife, 2019, 8:e44210., DOI 10.7554/eLife.44210 Bian Y, Li W, Kremer DM, Sajjakulnukit P, Li S, Crespo J, et al. Cancer SLC43A2 Alters T Cell Methionine Metabolism and Histone Methylation. Nature, 2020, 585:277–82., DOI 10.1038/s41586-020-2682-1 Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB. Tryptophan-derived Catabolites Are Responsible for Inhibition of T and Natural Killer Cell Proliferation Induced by Indoleamine 2, 3-Dioxygenase. J Exp Med, 2002, 196:459–68., DOI 10.1084/jem.20020121 Nguyen NT, Kimura A, Nakahama T, Chinen I, Masuda K, Nohara K, et al. Aryl Hydrocarbon Receptor Negatively Regulates Dendritic Cell Immunogenicity via a Kynurenine-dependent Mechanism. Proc Natl Acad Sci U S A, 2010, 107:19961–6., DOI 10.1073/pnas.1014465107 Cesar-Razquin A, Snijder B, Frappier-Brinton T, Isserlin R, Gyimesi G, Bai X, et al. A Call for Systematic Research on Solute Carriers. Cell, 2015, 162:478–87., DOI 10.1016/j.cell.2015.07.022 Kandasamy P, Gyimesi G, Kanai Y, Hediger MA. Amino Acid Transporters Revisited: New Views in Health and Disease. Trends Biochem Sci, 2018, 43: 752–89., DOI 10.1016/j.tibs.2018.05.003 Lopes C, Pereira C, Medeiros R. ASCT2 and LAT1 Contribution to the Hallmarks of Cancer: From a Molecular Perspective to Clinical Translation. Cancers, Basel,, 2021, 13:E203., DOI 10.3390/cancers13020203 Koppula P, Zhuang L, Gan B. Cystine Transporter SLC7A11/xCT in Cancer: Ferroptosis, Nutrient Dependency, and Cancer Therapy. Protein Cell, 2021, 12:599–620., DOI 10.1007/s13238-020-00789-5 van Geldermalsen M, Wang Q, NagaRajah R, Marshall AD, Thoeng A, Gao D, et al. ASCT2/SLC1A5 Controls Glutamine Uptake and Tumour Growth in Triple-Negative Basal-like Breast Cancer. Oncogene, 2016, 35:3201–8., DOI 10. 1038/onc.2015.381 Wang W, Zou W. Amino Acids and Their Transporters in T Cell Immunity and Cancer Therapy. Mol Cel, 2020, 80:384–95., DOI 10.1016/j.molcel.2020.09.006 Li J, Xie J, Wu D, Chen L, Gong Z, Wu R, et al. A Pan-Cancer Analysis Revealed the Role of the SLC16 Family in Cancer. Channels, 2021, 15:528–40., DOI 10.1080/19336950.2021.1965422 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene Set Enrichment Analysis: a Knowledge-Based Approach for Interpreting Genome-wide Expression Profiles. Proc Natl Acad Sci U S A, 2005, 102:15545–50., DOI 10.1073/pnas.0506580102 Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, et al. Signatures of T Cell Dysfunction and Exclusion Predict Cancer Immunotherapy Response. Nat Med, 2018, 24: 1550–8., DOI 10.1038/s41591-018-0136-1 Zhao J, Guan JL. Signal Transduction by Focal Adhesion Kinase in Cancer. Cancer Metastasis Rev, 2009, 28:35–49., DOI 10.1007/s10555-008-9165-4 Kim ST, Cristescu R, Bass AJ, Kim KM, Odegaard JI, Kim K, et al. Comprehensive Molecular Characterization of Clinical Responses to PD-1 Inhibition in Metastatic Gastric Cancer. Nat Med, 2018, 24:1449–58., DOI 10. 1038/s41591-018-0101-z Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell, 2017, 168:707–23., DOI 10.1016/j.cell.2017.01.017 Kaji S, Irino T, Kusuhara M, Makuuchi R, Yamakawa Y, Tokunaga M, et al. Metabolomic Profiling of Gastric Cancer Tissues Identified Potential Biomarkers for Predicting Peritoneal Recurrence. Gastric Cancer, 2020, 23: 874–83., DOI 10.1007/s10120-020-01065-5 Lowman XH, Hanse EA, Yang Y, Ishak Gabra MB, Tran TQ, Li H, et al. p53 Promotes Cancer Cell Adaptation to Glutamine Deprivation by Upregulating Slc7a3 to Increase Arginine Uptake. Cell Rep, 2019, 26: 3051–60.e4., DOI 10.1016/j.celrep.2019.02.037 Shan YS, Hsu HP, Lai MD, Yen MC, Chen WC, Fang JH, et al. Argininosuccinate Synthetase 1 Suppression and Arginine Restriction Inhibit Cell Migration in Gastric Cancer Cell Lines. Sci Rep, 2015, 5:9783., DOI 10.1038/srep09783 Li M, Xu DM, Lin SB, Yang ZL, Xu TY, Yang JH, et al. Transcriptional Expressions of Hsa-Mir-183 Predicted Target Genes as Independent Indicators for Prognosis in Bladder Urothelial Carcinoma. Aging, Albany NY,, 2022, 14:3782–800., DOI 10.18632/aging.204040 Kong Z, Wan X, Lu Y, Zhang Y, Huang Y, Xu Y, et al. Circular RNA circFOXO3 Promotes Prostate Cancer Progression through Sponging miR- 29a-3p. J Cel Mol Med, 2020, 24:799–813., DOI 10.1111/jcmm.14791 Mitra SK, Schlaepfer DD. Integrin-regulated FAK-Src Signaling in normal and Cancer Cells. Curr Opin Cel Biol, 2006, 18:516–23., DOI 10.1016/j.ceb.2006.08.011 Zhan T, Rindtorff N, Boutros M. Wnt Signaling in Cancer. Oncogene, 2017, 36:1461–73., DOI 10.1038/onc.2016.304 Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin Signalling Prevents Anti-tumour Immunity. Nature, 2015, 523:231–5., DOI 10.1038/ nature14404 Perez-Diez A, Joncker NT, Choi K, Chan WFN, Anderson CC, Lantz O, et al. CD4 Cells Can Be More Efficient at Tumor Rejection Than CD8 Cells. Blood, 2007, 109:5346–54., DOI 10.1182/blood-2006-10-051318 Bos R, Sherman LA. CD4+ T-Cell Help in the Tumor Milieu Is Required for Recruitment and Cytolytic Function of CD8+ T Lymphocytes. Cancer Res, 2010, 70:8368–77., DOI 10.1158/0008-5472.Can-10-1322 Lorvik KB, Hammarstrom C, Fauskanger M, Haabeth OAW, Zangani M, Haraldsen G, et al. Adoptive Transfer of Tumor-Specific Th2 Cells Eradicates Tumors by Triggering an In Situ Inflammatory Immune Response. Cancer Res, 2016, 76:6864–76., DOI 10.1158/0008-5472.Can-16-1219 Aqbi HF,WallaceM, Sappal S, Payne KK,Manjili MH. IFN-Gamma Orchestrates Tumor Elimination, Tumor Dormancy, Tumor Escape, and Progression. J Leukoc Biol, 2018, 103:1219–23., DOI 10.1002/JLB.5MIR0917-351R Fuchs CS, Doi T, Jang RW, Muro K, Satoh T,Machado M, et al. Safety and Efficacy of Pembrolizumab Monotherapy in Patients with Previously Treated Advanced Gastric and Gastroesophageal Junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA Oncol, 2018, 4:e180013., DOI 10.1001/jamaoncol.2018.0013 Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of Tumor Mutation burden as an Immunotherapy Biomarker: Utility for the Oncology Clinic. Ann Oncol, 2019, 30:44–56., DOI 10.1093/ annonc/mdy495 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610610 EP 1610621 DI 10.3389/pore.2022.1610610 PG 12 ER PT J AU Ma, YT Yang, HL Yan, L Hua, F Wang, DG Xu, GY Li, Y Xue, YJ Qin, YJ Sha, D Ning, H Zhao, MQ Yao, ZG AF Ma, Yu-Ting Yang, Hong-Lan Yan, Li Hua, Fang Wang, Dong-Guan Xu, Guo-Ying Li, Yan Xue, Ying-Jie Qin, Ye-Jun Sha, Dan Ning, Hao Zhao, Miao-Qing Yao, Zhi-Gang TI Case Report: Potential Predictive Value of MMR/MSI Status and PD-1 Expression in Immunotherapy for Urothelial Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE immunotherapy; microsatellite instability; immune checkpoint inhibitors; urothelial carcinoma; lynch syndrome; PD-1/PD-L1 ID immunotherapy; microsatellite instability; immune checkpoint inhibitors; urothelial carcinoma; lynch syndrome; PD-1/PD-L1 AB Immune checkpoint inhibitors (ICIs) have shown encouraging outcomes against Lynch syndrome (LS)-associated colorectal cancer (CRC) and endometrial cancer with mismatch repair deficient/microsatellite instability–high (dMMR/MSI-H). However, there is as yet no clarity on the safety and efficacy of immunotherapy combined with chemotherapy in LSassociated urothelial carcinoma (UC). Here, we report a patient with recurrent and metastatic LS-associated UC who achieved sustained response to programmed death protein 1 (PD-1) inhibitor combined with chemotherapy over 31 months, during which the side effects of immunotherapy could be controlled and managed. Our findings indicate that the dMMR/MSI status and PD-1 expression in UC may have potential predictive value for the response to PD-1-targeted immunotherapy. Our case supports the inclusion of such combination and/or monotherapy for UC in clinical studies and using dMMR/MSI status and PD-1 expression as potential predictive biomarkers for assessment of the therapeutic response. C1 [Ma, Yu-Ting] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Yang, Hong-Lan] Dongying City People’s Hospital, Department of OncologyDongying, China. [Yan, Li] Dongying City People’s Hospital, Department of PathologyDongying, China. [Hua, Fang] Tulane University, Department of Microbiology and ImmunologyNew Orleans, LA, USA. [Wang, Dong-Guan] Dongying City People’s Hospital, Department of PathologyDongying, China. [Xu, Guo-Ying] Dongying Hospital of Traditional Chinese Medicine, Department of Urology SurgeryDongying, China. [Li, Yan] Dongying City People’s Hospital, Department of OncologyDongying, China. [Xue, Ying-Jie] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Qin, Ye-Jun] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Sha, Dan] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. [Ning, Hao] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of UrologyJinan, China. [Zhao, Miao-Qing] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Yao, Zhi-Gang] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. RP Yao, ZG (reprint author), Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of Pathology, Jinan, China. EM yzg20062009@163.com CR De Santis M, Bellmunt J, Mead G, Kerst JM, Leahy M, Maroto P, et al. Randomized Phase II/III Trial Assessing Gemcitabine/carboplatin and Methotrexate/carboplatin/vinblastine in Patients with Advanced Urothelial Cancer Who Are Unfit for Cisplatin-Based Chemotherapy: EORTC Study 30986. J Clin Oncol, 2012, 30:191–9., DOI 10.1200/JCO.2011.37.3571 Galsky MD, Pal SK, Lin SW, Ogale S, Zivkovic M, Simpson J, et al. Real-World Effectiveness of Chemotherapy in Elderly Patients with Metastatic Bladder Cancer in the United States. Bladder Cancer, 2018, 4:227–38., DOI 10.3233/ BLC-170149 Rhea LP, Mendez-Marti S, Kim D, Aragon-Ching JB. Role of Immunotherapy in Bladder Cancer. Cancer Treat Res Commun, 2021, 26:100296., DOI 10.1016/j. ctarc.2020.100296 Andre T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, et al. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. N Engl J Med, 2020, 383:2207–18., DOI 10.1056/NEJMoa2017699 O’Malley DM, Bariani GM, Cassier PA, Marabelle A, Hansen AR, De Jesus Acosta A, et al. Pembrolizumab in Patients with Microsatellite Instability-High Advanced Endometrial Cancer: Results from the KEYNOTE-158 Study. J Clin Oncol, 2022, 40:752–61., DOI 10.1200/JCO.21.01874 Skeldon SC, Semotiuk K, Aronson M, Holter S, Gallinger S, Pollett A, et al. Patients with Lynch Syndrome Mismatch Repair Gene Mutations Are at Higher Risk for Not Only Upper Tract Urothelial Cancer but Also Bladder Cancer. Eur Urol, 2013, 63:379–85., DOI 10.1016/j.eururo.2012.07.047 Huang N, Zhao C, Hu X, Zhang C, Xiong F,Huang W, et al. Safety and Efficacy of Sintilimab Combination Therapy for the Treatment of 48 Patients with Advanced Malignant Tumors. Transl Cancer Res, 2022, 11:252–61., DOI 10. 21037/tcr-22-54 Cao JZ, Wu W, Pan JF, Wang HW, Jiang JH, Ma Q. Case Report: Anlotinib Combined with Sintilimab as Third-Line Treatment in a Metastatic Urothelial Bladder Carcinoma Patient with FGFR3 Mutation. Front Oncol, 2021, 11: 643413., DOI 10.3389/fonc.2021.643413 Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A, Ruschoff J, et al. Revised Bethesda Guidelines for Hereditary Nonpolyposis Colorectal Cancer, Lynch Syndrome, and Microsatellite Instability. J Natl Cancer Inst, 2004, 96: 261–8., DOI 10.1093/jnci/djh034 Huang D, Matin SF, Lawrentschuk N, Roupret M. Systematic Review: An Update on the Spectrum of Urological Malignancies in Lynch Syndrome. Bladder Cancer, 2018, 4:261–8., DOI 10.3233/BLC-180180 van der Post RS, Kiemeney LA, Ligtenberg MJ, Witjes JA, Hulsbergen-van de Kaa CA, BoDmer D, et al. Risk of Urothelial Bladder Cancer in Lynch Syndrome Is Increased, in Particular Among MSH2 Mutation Carriers. J Med Genet, 2010, 47:464–70., DOI 10.1136/jmg.2010.076992 Castro MP, Goldstein N. Mismatch Repair Deficiency Associated with Complete Remission to Combination Programmed Cell Death Ligand Immune Therapy in a Patient with Sporadic Urothelial Carcinoma: Immunotheranostic Considerations. J Immunother Cancer, 2015, 3:58., DOI 10.1186/s40425-015-0104-y Hartmann A, Zanardo L, Bocker-Edmonston T, Blaszyk H, Dietmaier W, Stoehr R, et al. Frequent Microsatellite Instability in Sporadic Tumors of the Upper Urinary Tract. Cancer Res, 2002, 62: 6796–802. Teo MY, Seier K, Ostrovnaya I, Regazzi AM, Kania BE, Moran MM, et al. Alterations in DNA Damage Response and Repair Genes as Potential Marker of Clinical Benefit from PD-1/pd-L1 Blockade in Advanced Urothelial Cancers. J Clin Oncol, 2018, 36:1685–94., DOI 10.1200/JCO.2017.75.7740 Hodgson A, Vesprini D, Liu SK, Xu B, Downes MR. Correlation of Mismatch Repair Protein Deficiency, PD-L1 and CD8 Expression in High-Grade Urothelial Carcinoma of the Bladder. J Clin Pathol, 2020, 73:519–22., DOI 10.1136/jclinpath-2019-206256 Massard C, Gordon MS, Sharma S, Rafii S, Wainberg ZA, Luke J, et al. Safety and Efficacy of Durvalumab, MEDI4736), an Anti-programmed Cell Death Ligand-1 Immune Checkpoint Inhibitor, in Patients with Advanced Urothelial Bladder Cancer. J Clin Oncol, 2016, 34:3119–25., DOI 10.1200/JCO.2016.67. 9761 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610638 EP 1610643 DI 10.3389/pore.2022.1610638 PG 6 ER PT J AU Zeng, J Li, M Dai, K Zuo, B Guo, J Zang, L AF Zeng, Jianmin Li, Man Dai, Kefan Zuo, Bingyu Guo, Jianhui Zang, Lu TI A Novel Glycolysis-Related Long Noncoding RNA Signature for Predicting Overall Survival in Gastric Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lncRNA; gastric cancer; TCGA; immune infiltration; prognostic signature ID lncRNA; gastric cancer; TCGA; immune infiltration; prognostic signature AB Background: The aim of this study was to construct a glycolysis-related long noncoding RNA (lncRNA) signature to predict the prognosis of patients with gastric cancer (GC). Methods: Glycolysis-related genes were obtained from the Molecular Signatures Database (MSigDB), lncRNA expression profiles and clinical data of GC patients were obtained from The Cancer Genome Atlas database (TCGA). Furthermore, univariate Cox regression analysis, Least Absolute Shrinkage and Selection Operator (LASSO) and multivariate Cox regression analysis were used to construct prognostic glycolysisrelated lncRNA signature. The specificity and sensitivity of the signature was verified by receiver operating characteristic (ROC) curves. We constructed a nomogram to predict the 1-year, 3-year, and 5-year survival rates of GC patients. Besides, the relationship between immune infiltration and the risk score was analyzed in the high and low risk groups. Multi Experiment Matrix (MEM) was used to analyze glycolysis-related lncRNA target genes. R “limma” package was used to analyze the mRNA expression levels of the glycolysis-related lncRNA target genes in TCGA. Gene set enrichment analysis (GSEA) was employed to further explore the biological pathways in the high-risk group and the glycolysis-related lncRNA target gene. Results: A prognostic signature was conducted based on nine glycolysis-related lncRNAs, which are AL391152.1, AL590705.3, RHOXF1-AS1, CFAP61-AS1, LINC00412, AC005165.1, AC110995.1, AL355574.1 and SCAT1. The area under the ROC curve (AUC) values at 1-year, 3-year, and 5-year were 0.765, 0.828 and 0.707 in the training set, and 0.669, 740 and 0.807 in the testing set, respectively. In addition, the nomogram could efficaciously predict the 1-year, 3-year, and 5-year survival rates of the GC patients. Then, we discovered that GC patients with high-risk scores were more likely to respond to immunotherapy. GSEA revealed that the signature was mainly associated with the calcium signaling pathway, extracellular matrix (ECM) receptor interaction, and focal adhesion in high-risk group, also indicated that SBSPON is related to aminoacyl-tRNA biosynthesis, citrate cycle, fructose and mannose metabolism, pentose phosphate pathway and pyrimidine metabolism. Conclusion: Our study shows that the signature can predict the prognosis of GC and may provide new insights into immunotherapeutic strategies. C1 [Zeng, Jianmin] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of General SurgeryShanghai, China. [Li, Man] The First Affiliated Hospital of Bengbu Medical CollegeBengbu, China. [Dai, Kefan] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of General SurgeryShanghai, China. [Zuo, Bingyu] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of General SurgeryShanghai, China. [Guo, Jianhui] The Affiliated Hospital of Kunming University of Science and Technology, The First People’s Hospital of Yunnan Province, Second Department of General SurgeryKunming, China. [Zang, Lu] Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of General SurgeryShanghai, China. RP Zang, L (reprint author), Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Department of General Surgery, Shanghai, China. EM zanglu@yeah.net CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Matsuno K, Ishihara R, Ohmori M, Iwagami H, Shichijyo S, Maekawa A, et al. Time Trends in the Incidence of Esophageal Adenocarcinoma, Gastric Adenocarcinoma, and Superficial Esophagogastric junction Adenocarcinoma. J Gastroenterol, 2019, 54(9):784–91., DOI 10.1007/s00535- 019-01577-7 Allemani C, Weir HK, Carreira H, Harewood R, Spika D, Wang X-S, et al. Global Surveillance of Cancer Survival 1995–2009: Analysis of Individual Data for 25 676 887 Patients from 279 Population-Based Registries in 67 Countries, CONCORD-2). Lancet, 2015, 385(9972):977–1010., DOI 10.1016/s0140- 6736(14)62038-9 Hanahan D, Weinberg RA. Hallmarks of Cancer: the Next Generation. Cell, 2011, 144(5):646–74., DOI 10.1016/j.cell.2011.02.013 Faubert B, Solmonson A, DeBerardinis RJ. Metabolic Reprogramming and Cancer Progression. Science, New York, NY,, 2020, 368(6487):eaaw5473., DOI 10.1126/science.aaw5473 Yu S, Hu C, Cai L, Du X, Lin F, Yu Q, et al. Seven-Gene Signature Based on Glycolysis Is Closely Related to the Prognosis and Tumor Immune Infiltration of Patients with Gastric Cancer. Front Oncol, 2020, 10:1778., DOI 10.3389/fonc. 2020.01778 Levine AJ, Puzio-Kuter AM. The Control of the Metabolic Switch in Cancers by Oncogenes and Tumor Suppressor Genes. Science, New York, NY,, 2010, 330(6009):1340–4., DOI 10.1126/science.1193494 Warburg O. On the Origin of Cancer Cells. Science, New York, NY,, 1956, 123(3191):309–14., DOI 10.1126/science.123.3191.309 Liu Y, Zhang Z, Wang J, Chen C, Tang X, Zhu J, et al. Metabolic Reprogramming Results in Abnormal Glycolysis in Gastric Cancer: a Review. Onco Targets Ther, 2019, 12:1195–204., DOI 10.2147/OTT.S189687 Park JS, Burckhardt CJ, Lazcano R, Solis LM, Isogai T, Li L, et al. Mechanical Regulation of Glycolysis via Cytoskeleton Architecture. Nature, 2020, 578(7796):621–6., DOI 10.1038/s41586-020-1998-1 Ben-Haim S, Ell P. 18F-FDG PET and PET/CT in the Evaluation of Cancer Treatment Response. J Nucl Med, 2009, 50(1):88–99., DOI 10.2967/jnumed.108. 054205 Statello L, Guo CJ, Chen LL, Huarte M. Gene Regulation by Long Non-coding RNAs and its Biological Functions. Nat Rev Mol Cel Biol, 2021, 22(2):96–118., DOI 10.1038/s41580-020-00315-9 Li G, Kryczek I, Nam J, Li X, Li S, Li J, et al. LIMIT Is an Immunogenic lncRNA in Cancer Immunity and Immunotherapy. Nat Cel Biol, 2021, 23(5):526–37., DOI 10.1038/s41556-021-00672-3 Sun TT, He J, Liang Q, Ren LL, Yan TT, Yu TC, et al. LncRNA GClnc1 Promotes Gastric Carcinogenesis and May Act as a Modular Scaffold of WDR5 and KAT2A Complexes to Specify the Histone Modification Pattern. Cancer Discov, 2016, 6(7):784–801., DOI 10.1158/ 2159-8290.CD-15-0921 Schmitt AM, Chang HY. Long Noncoding RNAs in Cancer Pathways. Cancer Cell, 2016, 29(4):452–63., DOI 10.1016/j.ccell.2016.03.010 Napieralski R, Ott K, Kremer M, Specht K, Vogelsang H, Becker K, et al. Combined GADD45A and Thymidine Phosphorylase Expression Levels Predict Response and Survival of Neoadjuvant-Treated Gastric Cancer Patients. Clin Cancer Res, 2005, 11(8):3025–31., DOI 10.1158/1078-0432.Ccr- 04-1605 Tan IB, Ivanova T, Lim KH, Ong CW, Deng N, Lee J, et al. Intrinsic Subtypes of Gastric Cancer, Based on Gene Expression Pattern, Predict Survival and Respond Differently to Chemotherapy. Gastroenterology, 2011, 141(2): 476–85.e11., DOI 10.1053/j.gastro.2011.04.042 Jiang Y, Wang H, Wu J, Chen C, Yuan Q, Huang W, et al. Noninvasive Imaging Evaluation of Tumor Immune Microenvironment to Predict Outcomes in Gastric Cancer. Ann Oncol, 2020, 31(6):760–8., DOI 10.1016/j. annonc.2020.03.295 Jiang Y, Zhang Q, Hu Y, Li T, Yu J, Zhao L, et al. ImmunoScore Signature: A Prognostic and Predictive Tool in Gastric Cancer. Ann Surg, 2018, 267(3): 504–13., DOI 10.1097/SLA.0000000000002116 Van Cutsem E, Sagaert X, Topal B, Haustermans K, Prenen H. Gastric Cancer. Lancet, 2016, 388(10060):2654–64., DOI 10.1016/s0140-6736(16)30354-3 Huang J, Chen YX, Zhang B. IGF2-AS Affects the Prognosis and Metastasis of Gastric Adenocarcinoma via Acting as a ceRNA of miR-503 to Regulate SHOX2. Gastric Cancer, 2020, 23(1):23–38., DOI 10.1007/s10120-019-00976-2 Putzer BM, Solanki M, Herchenroder O. Advances in Cancer Stem Cell Targeting: How to Strike the Evil at its Root. Adv Drug Deliv Rev, 2017, 120:89–107., DOI 10.1016/j.addr.2017.07.013 Patel SA, Minn AJ. Combination Cancer Therapy with Immune Checkpoint Blockade: Mechanisms and Strategies. Immunity, 2018, 48(3):417–33., DOI 10. 1016/j.immuni.2018.03.007 DeVita VT, Jr, Rosenberg SA. Two Hundred Years of Cancer Research. N Engl J Med, 2012, 366(23):2207–14., DOI 10.1056/NEJMra1204479 Wayteck L, Breckpot K, Demeester J, De Smedt SC, Raemdonck K. A Personalized View on Cancer Immunotherapy. Cancer Lett, 2014, 352(1): 113–25., DOI 10.1016/j.canlet.2013.09.016 Pardoll DM. The Blockade of Immune Checkpoints in Cancer Immunotherapy. Nat Rev Cancer, 2012, 12(4):252–64., DOI 10.1038/nrc3239 Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N Engl J Med, 2010, 363(8):711–23., DOI 10.1056/NEJMoa1003466 Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the Treatment of Non-small-cell Lung Cancer. N Engl J Med, 2015, 372(21):2018–28., DOI 10.1056/NEJMoa1501824 Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med, 2017, 377(20):1919–29., DOI 10.1056/NEJMoa1709937 Leone RD, Powell JD.Metabolism of Immune Cells in Cancer. Nat Rev Cancer, 2020, 20(9):516–31., DOI 10.1038/s41568-020-0273-y Lim S, Phillips JB, Madeira da Silva L, Zhou M, Fodstad O, Owen LB, et al. Interplay between Immune Checkpoint Proteins and Cellular Metabolism. Cancer Res, 2017, 77(6):1245–9., DOI 10.1158/0008-5472.CAN-16-1647 Heward JA, Lindsay MA. Long Non-coding RNAs in the Regulation of the Immune Response. Trends Immunol, 2014, 35(9):408–19., DOI 10.1016/j.it.2014.07.005 Hogan PG, Chen L, Nardone J, Rao A. Transcriptional Regulation by Calcium, Calcineurin, and NFAT. Genes Dev, 2003, 17(18):2205–32., DOI 10.1101/gad. 1102703 Lewis RS. The Molecular Choreography of a Store-Operated CalciumChannel. Nature, 2007, 446(7133):284–7., DOI 10.1038/nature05637 Hogan PG, Rao A. Dissecting ICRAC, a Store-Operated Calcium Current. Trends Biochem Sci, 2007, 32(5):235–45., DOI 10.1016/j.tibs.2007.03.009 Xu M, Seas A, Kiyani M, Ji KSY, Bell HN. A Temporal Examination of Calcium Signaling in Cancer- from Tumorigenesis, to Immune Evasion, and Metastasis. Cell Biosci, 2018, 8:25., DOI 10.1186/s13578-018-0223-5 Theocharis AD, Manou D, Karamanos NK. The Extracellular Matrix as a Multitasking Player in Disease. FEBS J, 2019, 286(15):2830–69., DOI 10.1111/ febs.14818 Lewis JM, Truong TN, Schwartz MA. Integrins Regulate the Apoptotic Response to DNA Damage through Modulation of P53. Proc Natl Acad Sci U S A, 2002, 99(6):3627–32., DOI 10.1073/pnas.062698499 Sloan KE, Stewart JK, Treloar AF, Matthews RT, Jay DG. CD155/PVR Enhances Glioma Cell Dispersal by Regulating Adhesion Signaling and Focal Adhesion Dynamics. Cancer Res, 2005, 65(23):10930–7., DOI 10.1158/ 0008-5472.CAN-05-1890 Zhang C, Zhang Z, Zhang G, Xue L, Yang H, Luo Y, et al. A Three-lncRNA Signature of Pretreatment Biopsies Predicts Pathological Response and Outcome in Esophageal Squamous Cell Carcinoma with Neoadjuvant Chemoradiotherapy. Clin Transl Med, 2020, 10(4):e156., DOI 10.1002/ ctm2.156 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610643 EP 1610656 DI 10.3389/pore.2022.1610643 PG 14 ER PT J AU Lovas, Sz Al-Ali, ON Varga, G Szita, V Alizadeh, H Plander, M Rajnics, P Illes, Szemlaky, Zs Mikala, G Varoczy, L AF Lovas, Szilvia Al-Ali, Obajed Nora Varga, Gergely Szita, Virag Alizadeh, Hussain Plander, Mark Rajnics, Peter Illes, Arpad Szemlaky, Zsuzsa Mikala, Gabor Varoczy, Laszlo TI Pomalidomide Treatment in Relapsed/ Refractory Multiple Myeloma Patients —Real-World Data From Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE survival; toxicity; multiple myeloma; treatment response; pomalidomide ID survival; toxicity; multiple myeloma; treatment response; pomalidomide AB Pomalidomide is a third generation immunomodulatory drug in the treatment of refractory and relapsed multiple myeloma patients. Our aim was to investigate the efficacy and safety of pomalidomide therapy in a real world setting. Eighty-six Hungarian patients were included, 45 of whom received pomalidomide ± an alkylating agent, while in 38 of them pomalidomide was combined with a proteasome inhibitor. 56 patients (65%) showed any response to the treatment with 18 complete or very good partial remissions and 38 partial remissions. At a median duration of follow-up of 18.6 months, the median progression-free survival (PFS) was 9.03 months, while the median overall survival (OS) was 16.53 months in the whole cohort. Patients with early stage disease (R-ISS 1 and 2) had better survival results than those with stage 3 myeloma (p = 0.002). Neither the number of prior treatment lines, nor lenalidomide refractoriness had a significant impact on PFS. PFS was found similar between the cohort of patients with impaired renal function and the cohort without kidney involvement. During the study, eight mortal infections and two fatal bleeding complications occurred, however, mild hematologic and gastrointestinal toxicities were identified as the most frequent adverse events. The results of our investigations confirm that pomalidomide is an effective treatment option for relapsed/refractory MM, besides, the safety profile is satisfactory in subjects with both normal and impaired renal function. C1 [Lovas, Szilvia] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Al-Ali, Obajed Nora] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Varga, Gergely] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Szita, Virag] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Alizadeh, Hussain] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Plander, Mark] Vas County Markusovszky Hospital, Department of HematologySzombathely, Hungary. [Rajnics, Peter] Kaposi Mor Teaching Hospital, Department of HematologyKaposvar, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Szemlaky, Zsuzsa] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Mikala, Gabor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Varoczy, Laszlo] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. RP Varoczy, L (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM varoczy@internal.med.unideb.hu CR Cowan AJ, Green DJ, Kwok M, Lee S, Coffey DG, Holmberg LA, et al. Diagnosis and Management of Multiple Myeloma: A Review. JAMA, 2022, 327(5):464–77., DOI 10.1001/jama.2022.0003 Abe Y, Ishida T. Immunomodulatory Drugs in the Treatment of Multiple Myeloma. Jpn J Clin Oncol, 2019, 49(8):695–702., DOI 10.1093/jjco/hyz083 Charlinski G, Vesole DH, Jurczyszyn A. Rapid Progress in the Use of Immunomodulatory Drugs and Cereblon E3 Ligase Modulators in the Treatment of Multiple Myeloma. Cancers, 2021, 13(18):4666., DOI 10.3390/ cancers13184666 Hoffmann M, Kasserra C, Reyes J, Schafer P, Kosek J, Capone L, et al. Absorption, Metabolism and Excretion of [14C]pomalidomide in Humans Following Oral Administration. Cancer Chemother Pharmacol, 2013, 71(2): 489–501., DOI 10.1007/s00280-012-2040-6 Dimopoulos MA, Richardson P, Lonial S. Treatment Options for Patients with Heavily Pretreated Relapsed and Refractory Multiple Myeloma. Clin Lymphoma Myeloma Leuk, 2022, 22:460–73., DOI 10.1016/j.clml.2022.01.011 Miguel JS, Weisel K, Moreau P, Lacy M, Song K, Delforge M, et al. Pomalidomide Plus Low-Dose Dexamethasone versus High-Dose Dexamethasone Alone for Patients with Relapsed and Refractory Multiple Myeloma, MM-003): a Randomised, Open-Label, Phase 3 Trial. Lancet Oncol, 2013, 14(11):1055–66., DOI 10.1016/S1470-2045(13)70380-2 Richardson PG, Oriol A, Beksac M, Liberati AM, Galli M, Schjesvold F, et al. Pomalidomide, Bortezomib, and Dexamethasone for Patients with Relapsed or Refractory Multiple Myeloma Previously Treated with Lenalidomide, OPTIMISMM): a Randomised, Open-Label, Phase 3 Trial. Lancet Oncol, 2019, 20(6):781–94., DOI 10.1016/S1470-2045(19)30152-4 Attal M, Richardson PG, Rajkumar SV, San-Miguel J, Beksac M, Spicka I, et al. Isatuximab Plus Pomalidomide and Low-Dose Dexamethasone versus Pomalidomide and Low-Dose Dexamethasone in Patients with Relapsed and Refractory Multiple Myeloma, ICARIA-MM): a Randomised, Multicentre, Open-Label, Phase 3 Study. Lancet, 2019, 394(10214): 2096–107., DOI 10.1016/S0140-6736(19)32556-5 Dimopoulos MA, Terpos E, Boccadoro M, Delimpasi S, Beksac M, Katodritou E, et al. Daratumumab Plus Pomalidomide and Dexamethasone versus Pomalidomide and Dexamethasone Alone in Previously Treated Multiple Myeloma, APOLLO): an Open-Label, Randomised, Phase 3 Trial. Lancet Oncol, 2021, 22(6):801–12., DOI 10.1016/S1470-2045(21)00128-5 Usami E, Kimura M, Takenaka S, Iwai M, Teramachi H, Yoshimura T. Tolerability and Safety of Real-World Use of Pomalidomide in Patients with Relapsed/refractory Multiple Myeloma. Mol Clin Oncol, 2019, 10(2): 293–8., DOI 10.3892/mco.2018.1775 Del Giudice ML, Gozzetti A, Antonioli E, Orciuolo E, Ghio A, Ciofini S, et al. Real-life Experience with Pomalidomide Plus Low-Dose Dexamethasone in Patients with Relapsed and Refractory Multiple Myeloma: a Retrospective and Prospective Study. Medicina, 2021, 57(9):28900., DOI 10.3390/ medicina57090900 Maciocia N, Melville A, Cheesman S, Sharpley F, Ramasamy K, Streetly M, et al. Real-world Use of Pomalidomide and Dexamethasone in Double Refractory Multiple Myeloma Suggests Benefit in Renal Impairment and Adverse Genetics: a Multi-centre UK Experience. Br J Haematol, 2017, 176(6):908–17., DOI 10.1111/bjh.14547 Charlinski G, Grzasko N, Jurczyszyn A, Janczarski M, Szeremet A, Waszczuk- Gajda A, et al. The Efficacy and Safety of Pomalidomide in Relapsed/refractory Multiple Myeloma in a “Real-world” Study: Polish Myeloma Group Experience. Eur J Haematol, 2018, 101(3):354–61., DOI 10.1111/ejh.13106 Rodriguez-Otero P, Maialen S, Gonzalez-Rodriguez AP, Lavilla E, de Coca AG, Arguinano JM, et al. Pomalidomide, Cyclophosphamide, and Dexamethasone for the Treatment of Relapsed/refractory Multiple Myeloma: Real-World Analysis of the Pethema-GEM Experience. Clin Lymphoma Myeloma Leuk, 2021, 21(6):413–20., DOI 10.1016/j.clml.2021.02.004 Mark T, Falkenstein A, Kish J. Real-world Outcomes of Pomalidomide Therapy after Lenalidomide Induction in Relapsed/refractory Multiple Myeloma. Future Oncol, 2022, 18(5):553–64., DOI 10.2217/fon-2021-1176 Varga G, TothAD, Szita VR,Csukly Z,Hardi A, Gaal-Weisinger J, et al. Beneficial Effect of Lenalidomide-Dexamethason Treatment in Relapsed/Refractory Multiple Myeloma Patients: Results of Real-Life Data from 11 Hungarian Centers. Pathol Oncol Res, 2021, 27:613264., DOI 10.3389/pore.2021.613264 Varga G, Nagy Z, Demeter J, Kosztolanyi S, Szomor A, Alizadeh H, et al. Real World Efficacy and Safety Results of Ixazomib, Lenalidomide and Dexamethasone Combination in Relapsed/refractory Multiple Myeloma: Data Collected from the Hungarian Ixazomib Named Patient Program. Pathol Oncol Res, 2019, 25(4):1615–20., DOI 10.1007/s12253-019-00607-2 Lovas S, Varga G, Farkas P, Masszi T, Wohner N, Bereczki A, et al. Real-world Data Onthe Efficacy and Safety of Daratumumab Treatment in Hungarian Relapsed/refractory Multiple Myeloma Patients. Int J Hematol, 2019, 110(5): 559–65., DOI 10.1007/s12185-019-02715-w Szita VR, Mikala G, Kozma A, Fabian J, Hardi A, Alizadeh H, et al. Targeted Venetoclax Therapy in T(11;14, Multiple Myeloma: Real World Data from Seven Hungarian Centers. Pathol Oncol Res, 2022, 28:1610276., DOI 10.3389/ pore.2022.1610276 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610645 EP 1610651 DI 10.3389/pore.2022.1610645 PG 7 ER PT J AU Zhang, B Zhang, Y Li, Q Jiang, Q Chu, W Gong, H Li, R Ji, H AF Zhang, Bing Zhang, Yangyang Li, Quan Jiang, Qingjun Chu, Wei Gong, Haifeng Li, Ruyuan Ji, Hong TI Case report: Chronic lymphocytic leukemia/small lymphocytic lymphoma and monomorphic epitheliotropic intestinal T-cell lymphoma: A composite lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE case report; chronic lymphocytic leukemia; composite lymphoma; small lymphocytic lymphoma; monomorphic epitheliotropic intestinal T-cell lymphoma ID case report; chronic lymphocytic leukemia; composite lymphoma; small lymphocytic lymphoma; monomorphic epitheliotropic intestinal T-cell lymphoma AB Background: Composite lymphomas involving B-cell and T-cell lymphomas is very rare. Case presentation: We reported a 63-year-old gentleman with composite chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) and monomorphic epitheliotropic intestinal T-cell lymphoma (MEITL). The patient was admitted to our hospital due to abdominal pain, and was diagnosed with CLL/SLL after bone marrow (BM) biopsy, BM aspiration, and flow cytometry. Two weeks later, he was diagnosed with MEITL based on pathological analysis after intestine excision. Next gene sequencing (NGS) findings identified two hotspot mutation sites (STAT5B and DNMT3A) closely related with the pathogenesis of CLL/SLL and MEILT. Additionally, BCOR mutation was only detected in the CLL/SLL area. The likely pathogenic mutations of CLL were SETD2, NOTCH1, SF3B1, and PTPN11, while the likely pathogenic mutations related with the MEILT were TET2 and ZRSR2. Mutations of GATA3, PLCG2, and FAT1 were identified in both CLL/SLL and MEITL areas, but the clinical significance was unknown. Finally, the patient died in the 12-month follow-up after surgery. Conclusion: We report a rare case of composite CLL/SLL and MEITL that highlights the importance of careful inspection of hematologic neoplasms. We also present the results of NGS of different gene mutations in CLL and MEITL tissues. C1 [Zhang, Bing] Shandong University, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Department of UrologyQingdao, China. [Zhang, Yangyang] Binzhou Medical University Hospital, Department of PathologyBinzhou, China. [Li, Quan] Binzhou Medical University Hospital, Department of ImagingBinzhou, China. [Jiang, Qingjun] Shandong University, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Department of ImagingQingdao, China. [Chu, Wei] Binzhou Medical University Hospital, Department of PathologyBinzhou, China. [Gong, Haifeng] Binzhou Medical University Hospital, Department of PathologyBinzhou, China. [Li, Ruyuan] Shandong University, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Department of GastroenterologyQingdao, China. [Ji, Hong] Shandong University, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Department of PathologyQingdao, China. RP Ji, H (reprint author), Shandong University, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Department of Pathology, Qingdao, China. CR Campo E, Ghia P, Montserrat E, Harris N, Muller-Hermelink H, Stein H, et al. Chronic lymphocytic leukaemia/small lymphocytic lymphoma, who classification of tumours of haematopoietic and lymphoid tissues lyon. Lyon: IARC, 2017). p. 216–21. Van Der Nest BM, Leslie C, Joske D, Radeski D, White R, Cheah CY. Peripheral T-cell lymphoma arising in patients with chronic lymphocytic leukemia. Am J Clin Pathol, 2019, 152:818–27., DOI 10.1093/ajcp/aqz109 National cancer institute sponsored study of classifications of non-hodgkin’s lymphomas: summary and description of a working formulation for clinical usage. The non-hodgkin’s lymphoma pathologic classification project. Cancer, 1982, 49: 2112–35., DOI 10.1002/1097-0142(19820515)49:10<2112:aid-cncr2820491024>3.0. co;2-2 Thirumala S, Esposito M, Fuchs A. An unusual variant of composite lymphoma: A short case report and review of the literature. Arch Pathol Lab Med, 2000, 124:1376–8., DOI 10.1043/0003-9985(2000)124<1376:AUVOCL>2.0. CO;2 Strickler JG, Amsden TW, Kurtin PJ. Small B-cell lymphoid neoplasms with coexisting T-cell lymphomas. Am J Clin Pathol, 1992, 98:424–9., DOI 10.1093/ajcp/ 98.4.424 Trimech M, Letourneau A, Missiaglia E, De Prijck B, Nagy-HulligerM, Somja J, et al. Angioimmunoblastic T-Cell lymphoma and chronic lymphocytic leukemia/ small lymphocytic lymphoma: A novel form of composite lymphoma potentially mimicking richter syndrome. Am J Surg Pathol, 2021, 45:773–86., DOI 10.1097/pas. 0000000000001646 Tomita S, Kikuti YY, Carreras J, Sakai R, Takata K, Yoshino T, et al. Monomorphic epitheliotropic intestinal T-Cell lymphoma in asia frequently shows SETD2 alterations. Cancers, Basel,, 2020, 12:E3539., DOI 10.3390/ cancers12123539 Chen C, Gong Y, Yang Y, Xia Q, Rao Q, Shao Y, et al. Clinicopathological and molecular genomic features of monomorphic epitheliotropic intestinal T-Cell lymphoma in the chinese population: A Study of 20 cases. Diagn Pathol, 2021, 16:114., DOI 10.1186/s13000-021-01173-5 KimM, Lee E, Zang DY, Kim HJ, Kim HY, Han B, et al. Novel genes exhibiting DNA methylation alterations in korean patients with chronic lymphocytic leukaemia: A methyl-CpG-binding domain sequencing study. Sci Rep, 2020, 10: 1085., DOI 10.1038/s41598-020-57919-6 Diamantopoulos PT, Sofotasiou M, Georgoussi Z, Giannakopoulou N, Papadopoulou V, Galanopoulos A, et al. Prognostic significance of signal transducer and activator of transcription 5 and 5b expression in EPSTEIN-Barr virus-positive patients with chronic lymphocytic leukemia. Cancer Med, 2016, 5: 2240–8., DOI 10.1002/cam4.804 Biran A, Yin S, Kretzmer H, Ten Hacken E, Parvin S, Lucas F, et al. Activation of notch and myc Signaling via B-cell-restricted depletion of Dnmt3a generates a consistent murine model of chronic lymphocytic leukemia. Cancer Res, 2021, 81: 6117–30., DOI 10.1158/0008-5472.Can-21-1273 Mutzbauer G, Maurus K, Buszello C, Pischimarov J, Roth S, Rosenwald A, et al. SYK expression in monomorphic epitheliotropic intestinal T-Cell lymphoma. Mod Pathol, 2018, 31:505–16., DOI 10.1038/modpathol.2017.145 Quinquenel A, Fornecker LM, Letestu R, Ysebaert L, Fleury C, Lazarian G, et al. Prevalence of BTK and PLCG2 mutations in a real-life CLL cohort still on ibrutinib after 3 years: A FILO group study. Blood, 2019, 134:641–4., DOI 10.1182/ blood.2019000854 Messina M, Del Giudice I, Khiabanian H, Rossi D, Chiaretti S, Rasi S, et al. Genetic lesions associated with chronic lymphocytic leukemia chemorefractoriness. Blood, 2014, 123:2378–88., DOI 10.1182/blood-2013-10-534271 Parker H, Rose-Zerilli MJ, Larrayoz M, Clifford R, Edelmann J, Blakemore S, et al. Genomic disruption of the histone methyltransferase SETD2 in chronic lymphocytic leukaemia. Leukemia, 2016, 30:2179–86., DOI 10.1038/leu.2016.134 Roberti A, Dobay MP, Bisig B, Vallois D, Boechat C, Lanitis E, et al. Type II enteropathy-associated T-cell lymphoma features a unique genomic profile with highly recurrent SETD2 alterations. Nat Commun, 2016, 7:12602., DOI 10.1038/ ncomms12602 Moffitt AB, Ondrejka SL, McKinney M, Rempel RE, Goodlad JR, Teh CH, et al. Enteropathy-associated T cell lymphoma subtypes are characterized by loss of function of SETD2. J Exp Med, 2017, 214:1371–86., DOI 10.1084/jem.20160894 Tausch E, Schneider C, Robrecht S, Zhang C, Dolnik A, Bloehdorn J, et al. Prognostic and predictive impact of genetic markers in patients with CLL treated with obinutuzumab and venetoclax. Blood, 2020, 135:2402–12., DOI 10.1182/blood. 2019004492 Kohlhaas V, Blakemore SJ, Al-Maarri M, Nickel N, PalM, Roth A, et al. Active Akt signaling triggers CLL toward richter transformation via overactivation of Notch1. Blood, 2021, 137:646–60., DOI 10.1182/blood.2020005734 Kim JA, Hwang B, Park SN, Huh S, Im K, Choi S, et al. Genomic profile of chronic lymphocytic leukemia in Korea identified by targeted sequencing. PLoS One, 2016, 11:e0167641., DOI 10.1371/journal.pone.0167641 Riches JC, Davies JK, McClanahan F, Fatah R, Iqbal S, Agrawal S, et al. T cells from CLL patients exhibit features of T-Cell exhaustion but retain capacity for cytokine production. Blood, 2013, 121:1612–21., DOI 10.1182/blood-2012-09- 457531 Goolsby CL, Kuchnio M, Finn WG, Peterson L. Expansions of clonal and oligoclonal T cells in B-cell chronic lymphocytic leukemia are primarily restricted to the CD3(+)CD8(+, T-cell population. Cytometry, 2000, 42:188–95., DOI 10.1002/ 1097-0320(20000615)42:3<188:aid-cyto5>3.0.co;2-q Martinez A, Pittaluga S, Villamor N, Colomer D, Rozman M, Raffeld M, et al. Clonal T-cell populations and increased risk for cytotoxic T-cell lymphomas in B-CLL patients: clinicopathologic observations and molecular analysis. Am J Surg Pathol, 2004, 28:849–58., DOI 10.1097/00000478-200407000-00002 Went P, Agostinelli C, Gallamini A, Piccaluga PP, Ascani S, Sabattini E, et al. Marker expression in peripheral T-cell lymphoma: A proposed clinicalpathologic prognostic score. J Clin Oncol, 2006, 24:2472–9., DOI 10.1200/jco. 2005.03.6327 Kanavaros P, Boulland ML, Petit B, Arnulf B, Gaulard P. Expression of cytotoxic proteins in peripheral T-cell and natural killer-cell, NK, lymphomas: Association with extranodal site, NK or tgammadelta phenotype, anaplastic morphology and CD30 expression. Leuk Lymphoma, 2000, 38:317–26., DOI 10. 3109/10428190009087022 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610653 EP 1610660 DI 10.3389/pore.2022.1610653 PG 8 ER PT J AU Polk, N Budai, B Hitre, E Patocs, A Mersich, T AF Polk, Nandor Budai, Barna Hitre, Erika Patocs, Attila Mersich, Tamas TI Corrigendum: High Neutrophil-to-Lymphocyte Ratio (NLR) and Systemic Immune-Inflammation Index (SII) are Markers of Longer Survival After Metastasectomy of Patients With Liver-Only Metastasis of Rectal Cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE neutrophil-to-lymphocyte ratio; liver-only metastases of rectal cancer; metastasectomy; relapse-free survival; systemic immune-inflammation index; liver-only metastases of colon cancer; preoperative treatment ID neutrophil-to-lymphocyte ratio; liver-only metastases of rectal cancer; metastasectomy; relapse-free survival; systemic immune-inflammation index; liver-only metastases of colon cancer; preoperative treatment AB A Corrigendum on High Neutrophil-to-Lymphocyte Ratio (NLR) and Systemic Immune-Inflammation Index (SII) are Markers of Longer Survival After Metastasectomy of Patients With Liver-Only Metastasis of Rectal Cancer by Polk N, Budai B, Hitre E, Patocs A and Mersich T (2022). Pathol. Oncol. Res. 28:1610315. doi: 10. 3389/pore.2022.1610315 C1 [Polk, Nandor] Orszagos Onkologiai Intezet, Emlo- es Lagyreszsebeszeti OsztalyBudapest, Hungary. [Budai, Barna] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Hitre, Erika] Orszagos Onkologiai Intezet, „B” Belgyogyaszati-Onkologiai es Klinikai Farmakologiai OsztalyBudapest, Hungary. [Patocs, Attila] Semmelweis University, Department of Laboratory MedicineBudapest, Hungary. [Mersich, Tamas] Orszagos Onkologiai Intezet, Emlo- es Lagyreszsebeszeti OsztalyBudapest, Hungary. RP Budai, B (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Budapest, Hungary. EM budai.barna@oncol.hu NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610658 EP 1610659 DI 10.3389/pore.2022.1610658 PG 2 ER PT J AU Zhong, Z Xu, M Tan, J AF Zhong, Zixuan Xu, Minxuan Tan, Jun TI Identification of an Oxidative Stress-Related LncRNA Signature for Predicting Prognosis and Chemotherapy in Patients With Hepatocellular Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE hepatocellular carcinoma; survival analysis; immune cell infiltration; oxidative stress; drug sensitivity; lncRNA signature ID hepatocellular carcinoma; survival analysis; immune cell infiltration; oxidative stress; drug sensitivity; lncRNA signature AB Background: Oxidative stress plays a critical role in oncogenesis and tumor progression. However, the prognostic role of oxidative stress-related lncRNA in hepatocellular carcinomas (HCC) has not been fully explored. Methods: We used the gene expression data and clinical data from The Cancer Genome Atlas (TCGA) database to identify oxidative stress-related differentially expressed lncRNAs (DElncRNAs) by pearson correlation analysis. A four-oxidative stress-related DElncRNA signature was constructed by LASSO regression and Cox regression analyses. The predictive signature was further validated by Kaplan–Meier (K–M) survival analysis, receiver operating characteristic (ROC) curves, nomogram and calibration plots, and principal component analysis (PCA). Single-sample gene set enrichment analysis (ssGSEA) was used to explore the relationship between the signature and immune status. Finally, the correlation between the signature and chemotherapeutic response of HCC patients was analyzed. Results: In our study, the four-DElncRNA signature was not only proved to be a robust independent prognostic factor for overall survival (OS) prediction, but also played a crucial role in the regulation of progression and chemotherapeutic response of HCC. ssGSEA showed that the signature was correlated with the infiltration level of immune cells. HCC patients in high-risk group were more sensitive to the conventional chemotherapeutic drugs including Sorafenib, lapatinib, Nilotinib, Gefitinib, Erlotinib and Dasatinib, which pave the way for targeting DElncRNA-associated treatments for HCC patients. Conclusion: Our study has originated a prognostic signature for HCC based on oxidative stress-related DElncRNAs, deepened the understanding of the biological role of four key DElncRNAs in HCC and laid a theoretical foundation for the choice of chemotherapy. C1 [Zhong, Zixuan] Chongqing University of Education, School of Biological and Chemical Engineering, Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir RegionChongqing, China. [Xu, Minxuan] Chongqing University of Education, School of Biological and Chemical Engineering, Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir RegionChongqing, China. [Tan, Jun] Chongqing University of Education, School of Biological and Chemical Engineering, Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir RegionChongqing, China. RP Zhong, Z (reprint author), Chongqing University of Education, School of Biological and Chemical Engineering, Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, Chongqing, China. EM zhongzx@cque.edu.cn CR Balogh J, Victor D 3rd, Asham EH, Burroughs SG, Boktour M, Saharia A, et al. Hepatocellular Carcinoma: a Review. J Hepatocell Carcinoma, 2016, 3:41–53., DOI 10.2147/JHC.S61146 Uchida D, Takaki A, Oyama A, Adachi T, Wada N, Onishi H, et al. Oxidative Stress Management in Chronic Liver Diseases and Hepatocellular Carcinoma. Nutrients, 2020, 12(6):E1576., DOI 10.3390/ nu12061576 Brown ZJ, Greten TF, Heinrich B. Adjuvant Treatment of Hepatocellular Carcinoma: Prospect of Immunotherapy. Hepatology, 2019, 70(4):1437–42., DOI 10.1002/hep.30633 Heimbach JK, Kulik LM, Finn RS, Sirlin CB, Abecassis MM, Roberts LR, et al. AASLD Guidelines for the Treatment of Hepatocellular Carcinoma. Hepatology, 2018, 67(1):358–80., DOI 10.1002/hep.29086 Kirstein MM. Wirth TC: [Multimodal Treatment of Hepatocellular Carcinoma]. Internist, 2020, 61(2):164–9., DOI 10.1007/s00108-019- 00722-x Njei B, Rotman Y, Ditah I, Lim JK. Emerging Trends in Hepatocellular Carcinoma Incidence and Mortality. Hepatology, 2015, 61(1):191–9., DOI 10. 1002/hep.27388 Bartsch H, Nair J. Chronic Inflammation and Oxidative Stress in the Genesis and Perpetuation of Cancer: Role of Lipid Peroxidation, DNA Damage, and Repair. Langenbecks Arch Surg, 2006, 391(5):499–510., DOI 10.1007/s00423- 006-0073-1 Sosa V, Moline T, Somoza R, Paciucci R, Kondoh H, Lleonart ME. Oxidative Stress and Cancer: an Overview. Ageing Res Rev, 2013, 12(1):376–90., DOI 10. 1016/j.arr.2012.10.004 Huang PS,Wang CS, Yeh CT, Lin KH. Roles of Thyroid Hormone-Associated microRNAs Affecting Oxidative Stress in Human Hepatocellular Carcinoma. Int J Mol Sci, 2019, 20(20):E5220., DOI 10.3390/ijms20205220 Mossenta M, Busato D, Dal Bo M, Toffoli G. Glucose Metabolism and Oxidative Stress in Hepatocellular Carcinoma: Role and Possible Implications in Novel Therapeutic Strategies. Cancers, Basel,, 2020, 12(6): E1668., DOI 10.3390/cancers12061668 Muriel P. Role of Free Radicals in Liver Diseases. Hepatol Int, 2009, 3(4): 526–36., DOI 10.1007/s12072-009-9158-6 Kumari S, Badana AK, Malla R, Shailender G. Reactive Oxygen Species: a Key Constituent in Cancer Survival. Biomark Insights, 2018, 13:117727191875539., DOI 10.1177/1177271918755391 Wu X, Zhao J. Novel Oxidative Stress-Related Prognostic Biomarkers for Melanoma Associated with Tumor Metastasis. Medicine, Baltimore,, 2021, 100(8):e24866., DOI 10.1097/MD.0000000000024866 Ma-On C, Sanpavat A, Whongsiri P, Suwannasin S, Hirankarn N, Tangkijvanich P, et al. Oxidative Stress Indicated by Elevated Expression of Nrf2 and 8-OHdG Promotes Hepatocellular Carcinoma Progression. Med Oncol, 2017, 34(4):1–12., DOI 10.1007/s12032-017-0914-5 Yang H, Jiang Z, Wang S, Zhao Y, Song X, Xiao Y, et al. Long Non-coding Small Nucleolar RNA Host Genes in Digestive Cancers. Cancer Med, 2019, 8(18):7693–704., DOI 10.1002/cam4.2622 Li W, Chen QF, Huang T, Wu P, Shen L, Huang ZL. Identification and Validation of a Prognostic lncRNA Signature for Hepatocellular Carcinoma. Front Oncol, 2020, 10:780., DOI 10.3389/fonc.2020.00780 Schwarzmueller L, Bril O, Vermeulen L, Leveille N. Emerging Role and Therapeutic Potential of lncRNAs in Colorectal Cancer. Cancers, 2020, 12(12):3843., DOI 10.3390/cancers12123843 Zhao Y, Liu Y, Lin L, Huang Q, He W, Zhang S, et al. The lncRNA MACC1- AS1 Promotes Gastric Cancer Cell Metabolic Plasticity via AMPK/ Lin28 Mediated mRNA Stability of MACC1. Mol Cancer, 2018, 17(1): 69–16., DOI 10.1186/s12943-018-0820-2 Zhao QJ, Zhang J, Xu L, Liu FF. Identification of a Five-Long Non-coding RNA Signature to Improve the Prognosis Prediction for Patients with Hepatocellular Carcinoma. World J Gastroenterol, 2018, 24(30):3426–39., DOI 10.3748/wjg. v24.i30.3426 Yang Z, Yang Y, Zhou G, Luo Y, Yang W, Zhou Y, et al. The Prediction of Survival in Hepatocellular Carcinoma Based on A Four Long Non-coding RNAs Expression Signature. J Cancer, 2020, 11(14):4132–44., DOI 10.7150/jca. 40621 Qi W, Li Z, Xia L, Dai J, Zhang Q, Wu C, et al. LncRNA GABPB1-AS1 and GABPB1 Regulate Oxidative Stress during Erastin-Induced Ferroptosis in HepG2 Hepatocellular Carcinoma Cells. Sci Rep, 2019, 9(1):16185., DOI 10. 1038/s41598-019-52837-8 Ghafouri-Fard S, Shoorei H, Taheri M. Non-coding RNAs Are Involved in the Response to Oxidative Stress. Biomed Pharmacother, 2020, 127:110228., DOI 10.1016/j.biopha.2020.110228 Geeleher P, Cox NJ, Huang RS. Clinical Drug Response Can Be Predicted Using Baseline Gene Expression Levels and in Vitrodrug Sensitivity in Cell Lines. Genome Biol, 2014, 15(3):R47–12., DOI 10.1186/gb-2014-15-3-r47 Reinhold WC, Sunshine M, Liu H, Varma S, Kohn KW, Morris J, et al. CellMiner: a Web-Based Suite of Genomic and Pharmacologic Tools to Explore Transcript and Drug Patterns in the NCI-60 Cell Line Set. Cancer Res, 2012, 72(14):3499–511., DOI 10.1158/0008-5472.CAN-12-1370 Mayakonda A, Lin D-C, Assenov Y, Plass C, Koeffler HP. Maftools: Efficient and Comprehensive Analysis of Somatic Variants in Cancer. Genome Res, 2018, 28(11):1747–56., DOI 10.1101/gr.239244.118 Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of Tumor Mutation burden as an Immunotherapy Biomarker: Utility for the Oncology Clinic. Ann Oncol, 2019, 30(1):44–56., DOI 10.1093/ annonc/mdy495 Mossenta M, Busato D, Dal Bo M, Toffoli G. Glucose Metabolism and Oxidative Stress in Hepatocellular Carcinoma: Role and Possible Implications in Novel Therapeutic Strategies. Cancers, 2020, 12(6):1668., DOI 10.3390/cancers12061668 Nie K, Deng Z, Zheng Z, Wen Y, Pan J, Jiang X, et al. Identification of a 14- lncRNA Signature and Construction of a Prognostic Nomogram Predicting Overall Survival of Gastric Cancer. DNA Cel Biol, 2020, 39(9):1532–44., DOI 10. 1089/dna.2020.5565 Liu S, Cao Q, An G, Yan B, Lei L. Identification of the 3-lncRNA Signature as a Prognostic Biomarker for Colorectal Cancer. Int J Mol Sci, 2020, 21(24): E9359., DOI 10.3390/ijms21249359 Song JY, Li XP, Qin XJ, Zhang JD, Zhao JY,Wang R. A Fourteen-lncRNA Risk Score System for Prognostic Prediction of Patients with Non-small Cell Lung Cancer. Cancer Biomark, 2020, 29(4):493–508., DOI 10.3233/CBM-190505 Xing C, Sun SG, Yue ZQ, Bai F. Role of lncRNA LUCAT1 in Cancer. Biomed Pharmacother, 2021, 134:111158., DOI 10.1016/j.biopha.2020.111158 Wen JF, Jiang YQ, Li C, Dai XK, Wu T, Yin WZ. LncRNA-XIST Promotes the Oxidative Stress-induced Migration, Invasion, and Epithelial-to-mesenchymal Transition of Osteosarcoma Cancer Cells through miR-153-SNAI1 axis. Cell Biol Int, 2020, 44(10):1991–2001., DOI 10.1002/cbin.11405 Zhang Q, ChengM, Fan Z, Jin Q, Cao P, Zhou G. Identification of Cancer Cell Stemness-Associated Long Noncoding RNAs for Predicting Prognosis of Patients with Hepatocellular Carcinoma. DNA Cel Biol, 2021, 40(8): 1087–100., DOI 10.1089/dna.2021.0282 Deng B, Yang M, Wang M, Liu Z. Development and Validation of 9-long Noncoding RNA Signature to Predicting Survival in Hepatocellular Carcinoma. Medicine, Baltimore,, 2020, 99(21):e20422., DOI 10.1097/MD. 0000000000020422 Chen ZA, Tian H, Yao DM, Zhang Y, Feng ZJ, Yang CJ. Identification of a Ferroptosis-Related Signature Model Including mRNAs and lncRNAs for Predicting Prognosis and Immune Activity in Hepatocellular Carcinoma. Front Oncol, 2021, 11:738477., DOI 10.3389/fonc.2021.738477 Xu Q, Wang Y, Huang W. Identification of Immune-Related lncRNA Signature for Predicting Immune Checkpoint Blockade and Prognosis in Hepatocellular Carcinoma. Int Immunopharmacol, 2021, 92:107333., DOI 10. 1016/j.intimp.2020.107333 Qin G, Wang X, Ye S, Li Y, Chen M, Wang S, et al. NPM1 Upregulates the Transcription of PD-L1 and Suppresses T Cell Activity in Triple-Negative Breast Cancer. Nat Commun, 2020, 11(1):1669–16., DOI 10.1038/s41467-020- 15364-z Wong JC, Hasan MR, Rahman M, Yu AC, Chan SK, Schaeffer DF, et al. Nucleophosmin 1, Upregulated in Adenomas and Cancers of the colon, Inhibits P53-mediated Cellular Senescence. Int J Cancer, 2013, 133(7): 1567–77., DOI 10.1002/ijc.28180 Liu XS, Zhou LM, Yuan LL, Gao Y, Kui XY, Liu XY, et al. Corrigendum: NPM1 Is a Prognostic Biomarker Involved in Immune Infiltration of Lung Adenocarcinoma and Associated with m6A Modification and Glycolysis. Front Immunol, 2021, 12:751004., DOI 10.3389/fimmu.2021.751004 Orhan E, Velazquez C, Tabet I, Sardet C, Theillet C. Regulation of RAD51 at the Transcriptional and Functional Levels: What Prospects for Cancer Therapy? Cancers, 2021, 13(12):2930., DOI 10.3390/cancers13122930 Zhang S, Shao J, Su F. Prognostic Significance of STIP1 Expression in Human Cancer: A Meta-Analysis. Clin Chim Acta, 2018, 486:168–76., DOI 10.1016/j. cca.2018.07.037 Luo JP, Wang J, Huang JH. CDKN2A Is a Prognostic Biomarker and Correlated with Immune Infiltrates in Hepatocellular Carcinoma. Biosci Rep, 2021, 41(10):BSR20211103., DOI 10.1042/BSR20211103 Li L, Xie R, Lu G. Identification of m6A Methyltransferase-Related lncRNA Signature for Predicting Immunotherapy and Prognosis in Patients with Hepatocellular Carcinoma. Biosci Rep, 2021, 41(6):BSR20210760., DOI 10. 1042/BSR20210760 Guo M, Huang J, Shao X, Bai N, Li M, Niu M, et al. LINC02870 Facilitates SNAIL Translation to Promote Hepatocellular Carcinoma Progression. 2022. Chen C, Su N, Li G, Shen Y, Duan X. Long Non-coding RNA TMCC1-AS1 Predicts Poor Prognosis and Accelerates Epithelial-Mesenchymal Transition in Liver Cancer. Oncol Lett, 2021, 22(5):773., DOI 10.3892/ol.2021.13034 Guo C, Zhou S, Yi W, Yang P, Li O, Liu J, et al. Long Non-coding RNA Muskelin 1 Antisense RNA, MKLN1-AS, Is a Potential Diagnostic and Prognostic Biomarker and Therapeutic Target for Hepatocellular Carcinoma. Exp Mol Pathol, 2021, 120:104638., DOI 10.1016/j.yexmp.2021. 104638 Ouyang J, Zhu X, Chen Y, Wei H, Chen Q, Chi X, et al. NRAV, a Long Noncoding RNA, Modulates Antiviral Responses through Suppression of Interferon-Stimulated Gene Transcription. Cell Host Microbe, 2014, 16(5): 616–26., DOI 10.1016/j.chom.2014.10.001 Tang W, Chen Z, Zhang W, Cheng Y, Zhang B, Wu F, et al. The Mechanisms of Sorafenib Resistance in Hepatocellular Carcinoma: Theoretical Basis and Therapeutic Aspects. Signal Transduct Target Ther, 2020, 5(1):87., DOI 10. 1038/s41392-020-0187-x Abdelgalil AA, Alkahtani HM, Al-Jenoobi FI. Sorafenib. Profiles Drug Subst Excip Relat Methodol, 2019, 44:239–66., DOI 10.1016/bs.podrm.2018.11.003 Hong W, Liang L, Gu Y, Qi Z, Qiu H, Yang X, et al. Immune-Related lncRNA to Construct Novel Signature and Predict the Immune Landscape of Human Hepatocellular Carcinoma. Mol Ther Nucleic Acids, 2020, 22:937–47., DOI 10. 1016/j.omtn.2020.10.002 Moreira C, Kaklamani V. Lapatinib and Breast Cancer: Current Indications and Outlook for the Future. Expert Rev Anticancer Ther, 2010, 10(8):1171–82., DOI 10.1586/era.10.113 Wang S, Xie Y, Bao A, Li J, Ye T, Yang C, et al. Nilotinib, a Discoidin Domain Receptor 1, DDR1, Inhibitor, Induces Apoptosis and Inhibits Migration in Breast Cancer. Neoplasma, 2021, 68(5):975–82., DOI 10.4149/ neo_2021_201126N1282 Sacha T, Saglio G. Nilotinib in the Treatment of Chronic Myeloid Leukemia. Future Oncol, 2019, 15(9):953–65., DOI 10.2217/fon-2018-0468 Hida T, Ogawa S, Park JC, Shimizu J, Horio Y. Gefitinib for the Treatment of Non-small-cell Lung Cancer. Expert Rev Anticancer Ther, 2009, 9(1):17–35., DOI 10.1586/14737140.9.1.17 Abdelgalil AA, Al-Kahtani HM, Al-Jenoobi FI. Erlotinib. Profiles Drug Subst Excip Relat Methodol, 2020, 45:93–117., DOI 10.1016/bs.podrm.2019.10.004 Lindauer M., Hochhaus A. Dasatinib. In: Martens U, editor Small Molecules in Hematology. Cham: Springer, 2018, 212. Huang IT, Dhungel B, Shrestha R, Bridle KR, Crawford DHG, Jayachandran A, et al. Spotlight on Bortezomib: Potential in the Treatment of Hepatocellular Carcinoma. Expert Opin Investig Drugs, 2019, 28(1):7–18., DOI 10.1080/ 13543784.2019.1551359 Azmy AM, Nasr KE, Gobran NS, YassinM. Gemcitabine Plus Carboplatin in Patients with Advanced Hepatocellular Carcinoma: Results of a Phase II Study. ISRN Oncol, 2012, 2012:420931., DOI 10. 5402/2012/420931 Hsu C, Yang TS, Huo TI, Hsieh RK, Yu CW, Hwang WS, et al. Vandetanib in Patients with Inoperable Hepatocellular Carcinoma: a Phase II, Randomized, Double-Blind, Placebo-Controlled Study. J Hepatol, 2012, 56(5):1097–103., DOI 10.1016/j.jhep.2011.12.013 Zhang J, Zong Y, Xu G-Z, Xing K. Erlotinib for Advanced Hepatocellular Carcinoma. A Systematic Review of Phase II/III Clinical Trials. Saudi Med J, 2016, 37(11):1184–90., DOI 10.15537/smj.2016.11.16267 Decaens T, Barone C, Assenat E, Wermke M, Fasolo A, Merle P, et al. Phase 1b/2 Trial of Tepotinib in Sorafenib Pretreated Advanced Hepatocellular Carcinoma with MET Overexpression. Br J Cancer, 2021, 125(2):190–9., DOI 10.1038/s41416-021-01334-9 Zhu AX, Abrams TA, Miksad R, Blaszkowsky LS, Meyerhardt JA, Zheng H, et al. Phase 1/2 Study of Everolimus in Advanced Hepatocellular Carcinoma. Cancer, 2011, 117(22):5094–102., DOI 10.1002/cncr.26165 Martinez de Toda I, Ceprian N, Diaz-Del Cerro E, De la Fuente M. The Role of Immune Cells in Oxi-Inflamm-Aging. Cells, 2021, 10(11):2974., DOI 10.3390/ cells10112974 Huang Y, Wang FM, Wang T, Wang YJ, Zhu ZY, Gao YT, et al. Tumorinfiltrating FoxP3+ Tregs and CD8+ T Cells Affect the Prognosis of Hepatocellular Carcinoma Patients. Digestion, 2012, 86(4):329–37., DOI 10. 1159/000342801 Ino Y, Yamazaki-Itoh R, Shimada K, IwasakiM Kosuge T, Kanai Y, et al. Immune Cell Infiltration as an Indicator of the Immune Microenvironment of Pancreatic Cancer. Br J Cancer, 2013, 108(4):914–23., DOI 10.1038/bjc.2013.32 Liu Z, Zhou Q, Wang Z, Zhang H, Zeng H, Huang Q, et al. Intratumoral TIGIT+ CD8+ T-Cell Infiltration Determines Poor Prognosis and Immune Evasion in Patients with Muscle-Invasive Bladder Cancer. J Immunother Cancer, 2020, 8(2):e000978., DOI 10.1136/jitc-2020-000978 Ryder M, Ghossein RA, Ricarte-Filho JC, Knauf JA, Fagin JA. Increased Density of Tumor-Associated Macrophages Is Associated with Decreased Survival in Advanced Thyroid Cancer. Endocr Relat Cancer, 2008, 15(4): 1069–74., DOI 10.1677/ERC-08-0036 Tanaka A, Sakaguchi S. Regulatory T Cells in Cancer Immunotherapy. Cell Res, 2017, 27(1):109–18., DOI 10.1038/cr.2016.151 Huo J, Wu L, Zang Y. A Prognostic Model of 15 Immune-Related Gene Pairs Associated with TumorMutation Burden for Hepatocellular Carcinoma. Front Mol Biosci, 2020, 7:581354., DOI 10.3389/fmolb.2020.581354 Peng Y, Liu C, Li M, Li W, Zhang M, Jiang X, et al. Identification of a Prognostic and Therapeutic Immune Signature Associated with Hepatocellular Carcinoma. Cancer Cel Int, 2021, 21(1):98., DOI 10.1186/ s12935-021-01792-4 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610670 EP 1610684 DI 10.3389/pore.2022.1610670 PG 15 ER PT J AU Ma, W Zhang, X Ma, Ch Liu, P AF Ma, Wanshan Zhang, Xiaoning Ma, Chenchen Liu, Peng TI Highly expressed FAM189B predicts poor prognosis in hepatocellular carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; prognosis; hepatocellular carcinoma; clinical significance; FAM189B ID biomarker; prognosis; hepatocellular carcinoma; clinical significance; FAM189B AB Hepatocellular carcinoma (HCC) is one of the most malignant tumors with persistently high morbidity and mortality. However, the expression, prognostic and clinical significance of FAM189 family genes in HCC remain largely unknown. In this study, the expression levels of FAM189 family genes in HCC were analyzed through TCGA-LIHC and ICGC-LIRI-JP cohorts, and further validated in multiple independent GEO datasets. It was found that the expression of FAM189B was significantly upregulated in HCC tumor tissues, while the expression of FAM189A1 and FAM189A2 was not significantly changed between tumor and adjacent tissues. Further analysis revealed that upregulated copy number variation contributed to increased expression of FAM189B in HCC. Survival analysis showed that highly expressed FAM189B was significantly correlated with unfavorable prognosis, including overall survival, disease-specific survival, and progression-free interval. Univariate and multivariate Cox regression analysis showed that FAM189B was a potential novel prognosis factor for HCC patients. In addition, the association between FAM189B expression and clinical and molecular characteristics was analyzed. High expression of FAM189B was associated with high AFP level, high predicted risk metastasis signature, and TP53 mutation, while there was no significant association between FAM189B expression and cancer stage or tumor grade of HCC. Gene set enrichment analysis revealed that highly expressed FAM189B was closely related with signal pathways and biological processes associated with cell proliferation and cell cycle in HCC. In conclusion, this study suggested that FAM189B was highly expressed in HCC and highly expressed FAM189B may serve as an effective prognostic indicator and a potential therapeutic target for HCC patients. C1 [Ma, Wanshan] Shandong Medicine and Health Key Laboratory of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Department of Clinical Laboratory MedicineJinan, Shandong, China. [Zhang, Xiaoning] Shandong Medicine and Health Key Laboratory of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Department of Clinical Laboratory MedicineJinan, Shandong, China. [Ma, Chenchen] Affiliated Hospital of Shandong University of Chinese Traditional Medicine, Central LaboratoryJinan, Shandong, China. [Liu, Peng] Shandong Medicine and Health Key Laboratory of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Department of Clinical Laboratory MedicineJinan, Shandong, China. RP Liu, P (reprint author), Shandong Medicine and Health Key Laboratory of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Department of Clinical Laboratory Medicine, Jinan, China. EM summerivyleague@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018, 68(6):394–424. Epub 2018/09/13., DOI 10.3322/caac.21492 Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol, 2019, 16(10):589–604. Epub 2019/08/24., DOI 10.1038/ s41575-019-0186-y Kallwitz ER, Daviglus ML, Allison MA, Emory KT, Zhao L, Kuniholm MH, et al. Prevalence of suspected nonalcoholic fatty liver disease in hispanic/latino individuals differs by heritage. Clin Gastroenterol Hepatol, 2015, 13(3):569–76. Epub 2014/09/15., DOI 10.1016/j.cgh.2014.08.037 Mittal S, Sada YH, El-Serag HB, Kanwal F, Duan Z, Temple S, et al. Temporal trends of nonalcoholic fatty liver disease-related hepatocellular carcinoma in the veteran affairs population. Clin Gastroenterol Hepatol, 2015, 13(3):594–601. Epub 2014/08/26., DOI 10.1016/j.cgh.2014.08.013 Marengo A, Rosso C, Bugianesi E. Liver cancer: Connections with obesity, fatty liver, and cirrhosis. Annu Rev Med, 2016, 67:103–17., DOI 10.1146/annurev-med- 090514-013832 Kulik L, El-Serag HB. Epidemiology and management of hepatocellular carcinoma. Gastroenterology, 2019, 156(2):477–91. Epub 2018/10/28., DOI 10. 1053/j.gastro.2018.08.065 McGlynn KA, Petrick JL, El-Serag HB. Epidemiology of hepatocellular carcinoma. Hepatology, 2021, 73(Suppl. 1):4–13., DOI 10.1002/hep.31288 Gopal P, Yopp AC, Waljee AK, Chiang J, Nehra M, Kandunoori P, et al. Factors that affect accuracy of alpha-fetoprotein test in detection of hepatocellular carcinoma in patients with cirrhosis. Clin Gastroenterol Hepatol, 2014, 12(5): 870–7., DOI 10.1016/j.cgh.2013.09.053 McCallie BR, Parks JC, Patton AL, Griffin DK, Schoolcraft WB, Katz-JaffeMG. Hypomethylation and genetic instability in monosomy blastocysts may contribute to decreased implantation potential. PLoS One, 2016, 11(7):e0159507. Epub 2016/ 07/21., DOI 10.1371/journal.pone.0159507 Laqqan M, Hammadeh ME. Aberrations in sperm DNA methylation patterns of males suffering from reduced fecundity. Andrologia, 2018, 50(3):e12913. Epub 2017/10/27., DOI 10.1111/and.12913 Mazzara PG, Muggeo S, Luoni M, Massimino L, Zaghi M, Valverde PT, et al. Frataxin gene editing rescues Friedreich’s ataxia pathology in dorsal root ganglia organoid-derived sensory neurons. Nat Commun, 2020, 11(1):4178. Epub 2020/08/ 23., DOI 10.1038/s41467-020-17954-3 Tsunoda T, Riku M, Yamada N, Tsuchiya H, Tomita T, Suzuki M, et al. Entrep/Fam189a2 encodes a new itch ubiquitin ligase activator that is downregulated in breast cancer. EMBO Rep, 2022, 23(2):e51182., DOI 10.15252/ embr.202051182 Hu X, Sun G, Shi Z, Ni H, Jiang S. Identification and validation of key modules and hub genes associated with the pathological stage of oral squamous cell carcinoma by weighted gene Co-expression network analysis. PeerJ, 2020, 8: e8505. Epub 2020/03/03., DOI 10.7717/peerj.8505 Wang A, Guo H, Long Z. Integrative analysis of differently expressed genes reveals a 17-gene prognosis signature for endometrial carcinoma. Biomed Res Int, 2021, 2021:4804694., DOI 10.1155/2021/4804694 Winfield SL, Tayebi N, Martin BM, Ginns EI, Sidransky E. Identification of three additional genes contiguous to the glucocerebrosidase locus on chromosome 1q21: Implications for gaucher disease. Genome Res, 1997, 7(10):1020–6. Epub 1997/10/23., DOI 10.1101/gr.7.10.1020 Yu W, Andersson B,Worley KC, Muzny DM, Ding Y, LiuW, et al. Large-scale concatenation cdna sequencing. Genome Res, 1997, 7(4):353–8. Epub 1997/04/01., DOI 10.1101/gr.7.4.353 Wu CL, Tan QX, Liu D, Jiang JE, Lu T, Huang ZM, et al. High FAM189b expression and its prognostic value in patients with gastric cancer. Biomed Res Int, 2021, 2021:8875971., DOI 10.1155/2021/8875971 Zhang H, Tian Y, Shen J, Wang Y, Xu Y, Wang Y, et al. Upregulation of the putative oncogene COTE1 contributes to human hepatocarcinogenesis through modulation of wwox signaling. Int J Oncol, 2014, 45(2):719–31. Epub 2014/06/06., DOI 10.3892/ijo.2014.2482 Zhang H, Huang CJ, Tian Y, Wang YP, Han ZG, Li XC. Ectopic overexpression of COTE1 promotes cellular invasion of hepatocellular carcinoma. Asian Pac J Cancer Prev, 2012, 13(11):5799–804. Epub 2013/01/16., DOI 10.7314/apjcp.2012.13.11.5799 Cancer Genome Atlas Research Network. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell, 2017, 169(7): 1327–41.e23., DOI 10.1016/j.cell.2017.05.046 Roessler S, Jia HL, Budhu A, Forgues M, Ye QH, Lee JS, et al. A unique metastasis gene signature enables prediction of tumor relapse in early-stage hepatocellular carcinoma patients. Cancer Res, 2010, 70(24):10202–12. Epub 2010/12/17., DOI 10.1158/0008-5472.CAN-10-2607 Burchard J, Zhang C, Liu AM, Poon RT, Lee NP, Wong KF, et al. Microrna- 122 as a regulator of mitochondrial metabolic gene network in hepatocellular carcinoma. Mol Syst Biol, 2010, 6:402., DOI 10.1038/msb.2010.58 Tung EK, Mak CK, Fatima S, Lo RC, Zhao H, Zhang C, et al. Clinicopathological and prognostic significance of serum and tissue dickkopf-1 levels in human hepatocellular carcinoma. Liver Int, 2011, 31(10):1494–504., DOI 10. 1111/j.1478-3231.2011.02597.x Neumann O, Kesselmeier M, Geffers R, Pellegrino R, Radlwimmer B, Hoffmann K, et al. Methylome analysis and integrative profiling of human hccs identify novel protumorigenic factors. Hepatology, 2012, 56(5):1817–27., DOI 10. 1002/hep.25870 GhandiM, Huang FW, Jane-Valbuena J, Kryukov GV, Lo CC, McDonald ER, 3rd, et al. Next-generation characterization of the cancer cell line Encyclopedia. Nature, 2019, 569(7757):503–8., DOI 10.1038/s41586-019-1186-3 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A, 2005, 102(43): 15545–50., DOI 10.1073/pnas.0506580102 Wang L, Li H, Li Z, Li M, Tang Q, Wu C, et al. Smoothened loss is a characteristic of neuroendocrine prostate cancer. Prostate, 2021, 81(9):508–20., DOI 10.1002/pros.24122 Yue X, Zhang Z, Liang X, Gao L, Zhang X, Zhao D, et al. Zinc fingers and homeoboxes 2 inhibits hepatocellular carcinoma cell proliferation and represses expression of cyclins a and E. Gastroenterology, 2012, 142(7):1559–70. Epub 2012/ 03/13., DOI 10.1053/j.gastro.2012.02.049 Zucman-Rossi J, Villanueva A, Nault JC, Llovet JM. Genetic landscape and biomarkers of hepatocellular carcinoma. Gastroenterology, 2015, 149(5):1226–39. e4., DOI 10.1053/j.gastro.2015.05.061 Li Y, Roberts ND, Wala JA, Shapira O, Schumacher SE, Kumar K, et al. Patterns of somatic structural variation in human cancer genomes. Nature, 2020, 578(7793):112–21., DOI 10.1038/s41586-019-1913-9 Ye QH, Qin LX, ForguesM, He P, Kim JW, Peng AC, et al. Predicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learning. Nat Med, 2003, 9(4):416–23. Epub 2003/03/18., DOI 10.1038/nm843 Ozen C, Yildiz G, Dagcan AT, Cevik D, Ors A, Keles U, et al. Genetics and epigenetics of liver cancer. N Biotechnol, 2013, 30(4):381–4., DOI 10.1016/j.nbt.2013. 01.007 Villanueva A. Hepatocellular carcinoma. N Engl J Med, 2019, 380(15): 1450–62., DOI 10.1056/NEJMra1713263 Saran U, Humar B, Kolly P, Dufour JF. Hepatocellular carcinoma and lifestyles. J Hepatol, 2016, 64(1):203–14., DOI 10.1016/j.jhep.2015.08.028 Bressac B, Galvin KM, Liang TJ, Isselbacher KJ, Wands JR, Ozturk M. Abnormal structure and expression of P53 gene in human hepatocellular carcinoma. Proc Natl Acad Sci U S A, 1990, 87(5):1973–7., DOI 10.1073/pnas.87.5.1973 Ozturk M. Genetic aspects of hepatocellular carcinogenesis. Semin Liver Dis, 1999, 19(3):235–42., DOI 10.1055/s-2007-1007113 Ozturk M, Arslan-Ergul A, Bagislar S, Senturk S, Yuzugullu H. Senescence and immortality in hepatocellular carcinoma. Cancer Lett, 2009, 286(1):103–13., DOI 10. 1016/j.canlet.2008.10.048 Perugorria MJ, Olaizola P, Labiano I, Esparza-Baquer A, Marzioni M, Marin JJG, et al. Wnt-β-catenin signalling in liver development, Health and disease. Nat Rev Gastroenterol Hepatol, 2019, 16(2):121–36., DOI 10.1038/ s41575-018-0075-9 Sunami Y, RingelhanM, Kokai E, LuM, O’Connor T, Lorentzen A, et al. Canonical NF-κB signaling in hepatocytes acts as a tumor-suppressor in hepatitis B virus surface antigen-driven hepatocellular carcinoma by controlling the unfolded protein response. Hepatology, 2016, 63(5): 1592–607., DOI 10.1002/hep.28435 Deng Q, Wang Q, Zong WY, Zheng DL, Wen YX, Wang KS, et al. E2f8 contributes to human hepatocellular carcinoma via regulating cell proliferation. Cancer Res, 2010, 70(2):782–91., DOI 10.1158/0008-5472.CAN-09- 3082 Khemlina G, Ikeda S, Kurzrock R. The biology of hepatocellular carcinoma: Implications for genomic and immune therapies. Mol Cancer, 2017, 16(1):149., DOI 10.1186/s12943-017-0712-x NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610674 EP 1610687 DI 10.3389/pore.2022.1610674 PG 14 ER PT J AU Hiroe, T Moriya, Sh Kobayashi, S Nishijima, Y Watanabe, A Shirabe, K Ikota, H Yokoo, H Saio, M AF Hiroe, Tamaki Moriya, Shunichi Kobayashi, Sayaka Nishijima, Yoshimi Watanabe, Akira Shirabe, Ken Ikota, Hayato Yokoo, Hideaki Saio, Masanao TI Negative correlation between the nuclear size and nuclear Lamina component Lamin A in intraductal papillary mucinous neoplasms of the pancreas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Emerin; Lamin A; nuclear morphology; intraductal papillary mucinous neoplasm; computer-assisted image analysis ID Emerin; Lamin A; nuclear morphology; intraductal papillary mucinous neoplasm; computer-assisted image analysis AB Background: The nuclear laminar protein Lamin A and inner nuclear membrane protein Emerin plays important role in sustaining nuclear structure. However, They have not investigated the significance of these proteins for development of pancreatic intraductal papillary mucinous neoplasm (IPMN). Methods: We examined pancreatic IPMN specimens for nuclear morphology and nuclear protein expression pattern of Lamin A and Emerin. Forty-two IPMN specimens were included, with 30 classified as intraductal papillary mucinous adenoma (IPMA) and 12 as intraductal papillary mucinous carcinoma (IPMC). Results: Classification according to histological subtype revealed that 26 specimenswere of the gastric subtype (1 IPMC case), 8 were pancreatobiliary (6 IPMC cases), 6 were intestinal (3 IPMC cases), and 2 were oncocytic (all cases were IPMC). The frequency of IPMN subtypes in this study seemed to agree with those in previous reports. We analyzed Feulgen staining sections for nuclear morphological analysis using computer-assisted image analysis. Nuclear area and perimeter were significantly larger in IPMC than in IPMA. Finally, we examined the positive ratios of Lamin A and Emerin in immunohistochemical staining sections by image analysis. We found a negative correlation between the nuclear size and Lamin A-positive ratio, which was significantly lower in IPMC than that in IPMA. However, no significant correlation was observed between nuclear size and Emerin expression was observed, and no differences were found in the Emerin-positive ratio between IPMA and IPMC. Conclusion: Our results suggest that a decreased Lamin A positive ratio induces nuclear enlargement in adenomas, which thereby induce promotion to carcinomas. Furthermore, Lamin A expression can be a reliable biomarker for distinguishing between IPMC and IPMA. C1 [Hiroe, Tamaki] Gunma University, Graduate School of Health Sciences, Department of Laboratory SciencesMaebashi, Gunma, Japan. [Moriya, Shunichi] Gunma University, Graduate School of Health Sciences, Department of Laboratory SciencesMaebashi, Gunma, Japan. [Kobayashi, Sayaka] Gunma University, Graduate School of Health Sciences, Department of Laboratory SciencesMaebashi, Gunma, Japan. [Nishijima, Yoshimi] Gunma University, Graduate School of Health Sciences, Department of Laboratory SciencesMaebashi, Gunma, Japan. [Watanabe, Akira] Gunma University, Graduate School of Medicine, Department of Hepatobiliary and Pancreatic SurgeryMaebashi, Gunma, Japan. [Shirabe, Ken] Gunma University, Graduate School of Medicine, Department of Hepatobiliary and Pancreatic SurgeryMaebashi, Gunma, Japan. [Ikota, Hayato] Gunma University Hospital, Clinical Department of PathologyMaebashi, Gunma, Japan. [Yokoo, Hideaki] Gunma University, Graduate School of Medicine, Department of Human PathologyMaebashi, Gunma, Japan. [Saio, Masanao] Gunma University, Graduate School of Health Sciences, Department of Laboratory SciencesMaebashi, Gunma, Japan. RP Saio, M (reprint author), Gunma University, Graduate School of Health Sciences, Department of Laboratory Sciences, Maebashi, Japan. EM saio@gunma-u.ac.jp CR Andrejevic-Blant S, Kosmahl M, Sipos B, Kloppel G. Pancreatic intraductal papillary-mucinous neoplasms: A new and evolving entity. Virchows Arch, 2007, 451(5):863–9., DOI 10.1007/s00428-007-0512-6 Furukawa T, Kloppel G, Volkan Adsay N, Albores-Saavedra J, Fukushima N, Horii A, et al. Classification of types of intraductal papillary-mucinous neoplasm of the pancreas: A consensus study. Virchows Arch, 2005, 447(5):794–9., DOI 10.1007/ s00428-005-0039-7 Nakata B, Yashiro M, Nishioka N, Aya M, Yamada S, Takenaka C, et al. Very low incidence of microsatellite instability in intraductal papillary-mucinous neoplasm of the pancreas. Int J Cancer, 2002, 102(6):655–9., DOI 10.1002/ijc.10771 Kobayashi M, Fujinaga Y, Ota H. Reappraisal of the immunophenotype of pancreatic intraductal papillary mucinous neoplasms, IPMNs)-Gastric pyloric and small intestinal immunophenotype expression in gastric and intestinal type IPMNs. Acta Histochem Cytochem, 2014, 47(2):45–57., DOI 10.1267/ahc.13027 Saito M, Imada H, Suzuki T, Sata N, Yasuda Y, Maetani I, et al. Distinct patterns of peritumoral histological findings in subtypes of intraductal papillary mucinous neoplasms of the pancreas. Ann Diagn Pathol, 2015, 19(5):347–52., DOI 10.1016/j. anndiagpath.2015.07.005 Furukawa T, Hatori T, Fujita I, Yamamoto M, Kobayashi M, Ohike N, et al. Prognostic relevance of morphological types of intraductal papillary mucinous neoplasms of the pancreas. Gut, 2011, 60(4):509–16., DOI 10.1136/gut.2010.210567 Muchir A, Worman HJ. The nuclear envelope and human disease. Physiology, 2004, 19:309–14., DOI 10.1152/physiol.00022.2004 Clements L,Manilal S, Love DR, Morris GE. Direct interaction between emerin and lamin A. Biochem Biophys Res Commun, 2000, 267(3):709–14., DOI 10.1006/ bbrc.1999.2023 Sakaki M, Koike H, Takahashi N, Sasagawa N, Tomioka S, Arahata K, et al. Interaction between emerin and nuclear lamins. J Biochem, 2001, 129(2):321–7., DOI 10.1093/oxfordjournals.jbchem.a002860 Harada T, Swift J, Irianto J, Shin JW, Spinler KR, Athirasala A, et al. Nuclear lamin stiffness is a barrier to 3D migration, but softness can limit survival. J Cel Biol, 2014, 204(5):669–82., DOI 10.1083/jcb.201308029 Lammerding J, Hsiao J, Schulze PC, Kozlov S, Stewart CL, Lee RT. Abnormal nuclear shape and impaired mechanotransduction in emerin-deficient cells. J Cel Biol, 2005, 170(5):781–91., DOI 10.1083/jcb.200502148 Wang Y, Jiang J, He L, Gong G, Wu X. Effect of lamin-A expression on migration and nuclear stability of ovarian cancer cells. Gynecol Oncol, 2019, 152(1): 166–76., DOI 10.1016/j.ygyno.2018.10.030 Urciuoli E, Petrini S, D’Oria V, Leopizzi M, Rocca CD, Peruzzi B. Nuclear lamins and emerin are differentially expressed in osteosarcoma cells and scale with tumor aggressiveness. Cancers, Basel,, 2020, 12(2):443., DOI 10.3390/ cancers12020443 Alhudiri IM, Nolan CC, Ellis IO, Elzagheid A, Rakha EA, Green AR, et al. Expression of Lamin A/C in early-stage breast cancer and its prognostic value. Breast Cancer Res Treat, 2019, 174(3):661–8., DOI 10.1007/s10549- 018-05092-w Wazir U, Ahmed MH, Bridger JM, Harvey A, Jiang WG, Sharma AK, et al. The clinicopathological significance of lamin A/C, lamin B1 and lamin B receptor mRNA expression in human breast cancer. Cell Mol Biol Lett, 2013, 18(4):595–611., DOI 10.2478/s11658-013-0109-9 Gong G, Chen P, Li L, Tan H, Zhou J, Zhou Y, et al. Loss of lamin A but not lamin C expression in epithelial ovarian cancer cells is associated with metastasis and poor prognosis. Pathol Res Pract, 2015, 211(2):175–82., DOI 10.1016/j.prp.2014. 11.008 Willis ND, Cox TR, Rahman-Casans SF, Smits K, Przyborski SA, van den Brandt P, et al. Lamin A/C is a risk biomarker in colorectal cancer. Plos One, 2008, 3(8):e2988., DOI 10.1371/journal.pone.0002988 Izdebska M, Gagat M, Grzanka A. Overexpression of lamin B1 induces mitotic catastrophe in colon cancer LoVo cells and is associated with worse clinical outcomes. Int J Oncol, 2018, 52(1):89–102., DOI 10.3892/ijo.2017.4182 Guilford J. Correlation. In: Morgan C, editor. Fundamental statistics in psychology and education. New York: McGraw-Hill Book Company, 1950). p. 152–73. Adsay N, Fukushima N, Furukawa T, Hruban RH, Kd S, Kloppel G, et al. Intraductal neoplasms of the pancreas. In: Bosman FT, Carnerio F, Hruban RH, Theise ND, editors. WHO classification of tumours of the digestive system. Geneva: WHO Press, 2010). p. 304–13. Mohri D, Asaoka Y, Ijichi H, Miyabayashi K, Kudo Y, Seto M, et al. Different subtypes of intraductal papillary mucinous neoplasm in the pancreas have distinct pathways to pancreatic cancer progression. J Gastroenterol, 2012, 47(2):203–13., DOI 10.1007/s00535-011-0482-y Aiad H, Abdou A, Bashandy M, Said A, Ezz-Elarab S, Zahran A. Computerized nuclear morphometry in the diagnosis of thyroid lesions with predominant follicular pattern. Ecancermedicalscience, 2009, 3:146., DOI 10.3332/ ecancer.2009.146 Ikeguchi M, Sakatani T, Endo K, Makino M, Kaibara N. Computerized nuclear morphometry is a useful technique for evaluating the high metastatic potential of colorectal adenocarcinoma. Cancer, 1999, 86(10):1944–51., DOI 10. 1002/(sici)1097-0142(19991115)86:10<1944::aid-cncr10>3.0.co;2-2 Mulder JW, Offerhaus GJ, de Feyter EP, Floyd JJ, Kern SE, Vogelstein B, et al. The relationship of quantitative nuclear morphology to molecular genetic alterations in the adenoma-carcinoma sequence of the large bowel. Am J Pathol, 1992, 141(4):797–804. Kitamura H, Kameda Y, Nakamura N, Inayama Y, Nakatani Y, Shibagaki T, et al. Atypical adenomatous hyperplasia and bronchoalveolar lung carcinoma. Analysis by morphometry and the expressions of p53 and carcinoembryonic antigen. Am J Surg Pathol, 1996, 20(5):553–62., DOI 10.1097/00000478- 199605000-00002 Liddane AG, McNamara CA, Campbell MC, Mercier I, Holaska JM. Defects in emerin-nucleoskeleton binding disrupt nuclear structure and promote breast cancer cell motility and metastasis. Mol Cancer Res, 2021, 19:1196–207., DOI 10. 1158/1541-7786.MCR-20-0413 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610684 EP 1610696 DI 10.3389/pore.2022.1610684 PG 13 ER PT J AU He, K Li, Ch Yuan, H Jiang, K Deng, G AF He, Kangwei Li, Changjiu Yuan, Hui Jiang, Kang Deng, Gang TI Immunological role and prognostic value of SPARCL1 in pan-cancer analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; immune infiltration; pan-cancer; SPARCL1; M2 macrophage ID prognosis; immune infiltration; pan-cancer; SPARCL1; M2 macrophage AB Background: Secreted protein acidic and rich in cysteine-like 1 (SPARCL1) was a kind of extracellular matrix glycoprotein. SPARCL1 was strongly inhibited in most cancers. However, the potential functions of SPARCL1 in the pan-cancer cohort have not been widely studied. Methods: We evaluated the transcriptional level and the prognostic value of SPARCL1 in 33 types of cancer and revealed the relationship between genetic alterations of SPARCL1 and the tumor mutation burden. Meanwhile, we assessed the correlations between SPARCL1 and tumor-infiltrating lymphocytes across cancers. Results: The transcriptional level of SPARCL1 was inhibited in most cancers. Although SPARCL1 was down-regulated in most cancers, SPARCL1 might play a protective or detrimental role in different cancers. We demonstrated that mutation count was elevated in the altered SPARCL1 group in several cancers. Additionally, we found a significant positive correlation between SPARCL1 and macrophage infiltration levels in most cancers. Especially, marker sets of M2 macrophages were strongly related to SPARCL1 in cholangiocarcinoma, colon adenocarcinoma, rectum adenocarcinoma, and pancreatic adenocarcinoma. Conclusion: Our study found that SPARCL1 might work as a biomarker for prognosis and immune infiltration in pan-cancer analysis. C1 [He, Kangwei] Zhejiang Chinese Medical University, The Fourth Clinical Medical CollegeHangzhou, China. [Li, Changjiu] Zhejiang University, School of Medicine, The First Affiliated Hospital, Department of UrologyHangzhou, China. [Yuan, Hui] Zhejiang Chinese Medical University, The Fourth Clinical Medical CollegeHangzhou, China. [Jiang, Kang] Zhejiang University, School of Medicine, The First Affiliated Hospital, Department of UrologyHangzhou, China. [Deng, Gang] Zhejiang University, School of Medicine, The First Affiliated Hospital, Department of UrologyHangzhou, China. RP Deng, G (reprint author), Zhejiang University, School of Medicine, The First Affiliated Hospital, Department of Urology, Hangzhou, China. EM dfg326@aliyun.com CR Gagliardi F, Narayanan A, Mortini P. SPARCL1 a novel player in cancer biology. Crit Rev Oncol Hematol, 2017, 109:63–8., DOI 10.1016/j.critrevonc.2016. 11.013 Girard JP, Springer TA. Modulation of endothelial cell adhesion by hevin, an acidic protein associated with high endothelial venules. J Biol Chem, 1996, 271: 4511–7., DOI 10.1074/jbc.271.8.4511 Kucukdereli H, Allen NJ, Lee AT, Feng A, Ozlu MI, Conatser LM, et al. Control of excitatory CNS synaptogenesis by astrocyte-secreted proteins Hevin and SPARC. Proc Natl Acad Sci U S A, 2011, 108:E440–9., DOI 10.1073/pnas.1104977108 Jones EV, Bouvier DS. Astrocyte-secreted matricellular proteins in CNS remodelling during development and disease. Neural Plast, 2014, 2014:321209., DOI 10.1155/2014/321209 Girard JP, Springer TA. Cloning from purified high endothelial venule cells of hevin, a close relative of the antiadhesive extracellular matrix protein SPARC. Immunity, 1995, 2:113–23., DOI 10.1016/1074-7613(95)90083-7 Johnston IG, Paladino T, Gurd JW, Brown IR. Molecular cloning of SC1: a putative brain extracellular matrix glycoprotein showing partial similarity to osteonectin/BM40/SPARC. Neuron, 1990, 4:165–76., DOI 10.1016/0896-6273(90, 90452-l Singh SK, Stogsdill JA, Pulimood NS, Dingsdale H, Kim YH, Pilaz LJ, et al. Astrocytes Assemble Thalamocortical Synapses by Bridging NRX1α and NL1 via Hevin. Cell, 2016, 164:183–96., DOI 10.1016/j.cell.2015.11.034 Nelson PS, Plymate SR, Wang K, True LD, Ware JL, Gan L, et al. Hevin, an antiadhesive extracellular matrix protein, is down-regulated in metastatic prostate adenocarcinoma. Cancer Res, 1998, 58:232–6. Zhang H, Widegren E, Wang DW, Sun XF. SPARCL1: a potential molecule associated with tumor diagnosis, progression and prognosis of colorectal cancer. Tumour Biol, 2011, 32:1225–31., DOI 10.1007/s13277-011-0226-x Li P, Qian J, Yu G, Chen Y, Liu K, Li J, et al. Down-regulated SPARCL1 is associated with clinical significance in human gastric cancer. J Surg Oncol, 2012, 105:31–7., DOI 10.1002/jso.22025 Bendik I, Schraml P, Ludwig CU. Characterization of MAST9/Hevin, a SPARC-like protein, that is down-regulated in non-small cell lung cancer. Cancer Res, 1998, 58:626–9. Xiang Y, Qiu Q, Jiang M, Jin R, Lehmann BD, Strand DW, et al. SPARCL1 suppresses metastasis in prostate cancer. Mol Oncol, 2013, 7: 1019–30., DOI 10.1016/j.molonc.2013.07.008 Hu H, Zhang H, Ge W, Liu X, Loera S, Chu P, et al. Secreted protein acidic and rich in cysteines-like 1 suppresses aggressiveness and predicts better survival in colorectal cancers. Clin Cancer Res, 2012, 18:5438–48., DOI 10.1158/1078-0432. CCR-12-0124 Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, et al. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res, 2020, 48:W509-W514–w14., DOI 10.1093/nar/gkaa407 Tang Z, Kang B, Li C, Chen T, Zhang Z. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res, 2019, 47: W556-W560–w60., DOI 10.1093/nar/gkz430 Nagy A, Munkacsy G, Gyorffy B. Pancancer survival analysis of cancer hallmark genes. Sci Rep, 2021, 11:6047., DOI 10.1038/s41598-021-84787-5 Mizuno H, Kitada K, Nakai K, Sarai A. PrognoScan: a new database for metaanalysis of the prognostic value of genes. BMC Med Genomics, 2009, 2:18., DOI 10. 1186/1755-8794-2-18 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal, 2013, 6:pl1., DOI 10.1126/scisignal.2004088 Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: AWeb Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. Cancer Res, 2017, 77:e108–e10., DOI 10.1158/0008-5472.CAN-17-0307 Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science, New York, NY,, 2015, 348:124–8., DOI 10. 1126/science.aaa1348 Azimi F, Scolyer RA, Rumcheva P, Moncrieff M, Murali R, McCarthy SW, et al. Tumor-infiltrating lymphocyte grade is an independent predictor of sentinel lymph node status and survival in patients with cutaneous melanoma. J Clin Oncol, 2012, 30:2678–83., DOI 10.1200/JCO.2011.37.8539 Li T, Liu X, Yang A, Fu W, Yin F, Zeng X. Associations of tumor suppressor SPARCL1 with cancer progression and prognosis. Oncol Lett, 2017, 14:2603–10., DOI 10.3892/ol.2017.6546 Zhang HP, Wu J, Liu ZF, Gao JW, Li SY. SPARCL1 Is a Novel Prognostic Biomarker and Correlates with Tumor Microenvironment in Colorectal Cancer. Biomed Res Int, 2022, 2022:1398268., DOI 10.1155/2022/ 1398268 Ma Y, Xu Y, Li L. SPARCL1 suppresses the proliferation and migration of human ovarian cancer cells via the MEK/ERK signaling. Exp Ther Med, 2018, 16: 3195–201., DOI 10.3892/etm.2018.6575 Zhao SJ, Jiang YQ, Xu NW, Li Q, Zhang Q, Wang SY, et al. SPARCL1 suppresses osteosarcoma metastasis and recruits macrophages by activation of canonical WNT/β-catenin signaling through stabilization of the WNT-receptor complex. Oncogene, 2018, 37:1049–61., DOI 10.1038/onc. 2017.403 Lau CP, Poon RT, Cheung ST, Yu WC, Fan ST. SPARC and Hevin expression correlate with tumour angiogenesis in hepatocellular carcinoma. J Pathol, 2006, 210:459–68., DOI 10.1002/path.2068 Hambrock HO, Nitsche DP, Hansen U, Bruckner P, Paulsson M, Maurer P, et al. SC1/hevin. An extracellular calcium-modulated protein that binds collagen I. J Biol Chem, 2003, 278:11351–8., DOI 10.1074/jbc.M212291200 Tomida S, Takeuchi T, Shimada Y, Arima C, Matsuo K, Mitsudomi T, et al. Relapse-related molecular signature in lung adenocarcinomas identifies patients with dismal prognosis. J Clin Oncol, 2009, 27:2793–9., DOI 10.1200/JCO.2008.19. 7053 Okayama H, Kohno T, Ishii Y, Shimada Y, Shiraishi K, Iwakawa R, et al. Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALKnegative lung adenocarcinomas. Cancer Res, 2012, 72:100–11., DOI 10.1158/0008- 5472.CAN-11-1403 Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol, 2019, 30:44–56., DOI 10.1093/annonc/mdy495 Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell, 2006, 124:263–6., DOI 10.1016/j.cell. 2006.01.007 Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell, 2010, 141:39–51., DOI 10.1016/j.cell.2010.03.014 Mantovani A, Sica A. Macrophages, innate immunity and cancer: balance, tolerance, and diversity. Curr Opin Immunol, 2010, 22:231–7., DOI 10.1016/j.coi. 2010.01.009 Kitamura T, Qian BZ, Pollard JW. Immune cell promotion of metastasis. Nat Rev Immunol, 2015, 15:73–86., DOI 10.1038/nri3789 Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity, 2014, 41:49–61., DOI 10.1016/j.immuni.2014.06.010 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610687 EP 1610700 DI 10.3389/pore.2022.1610687 PG 14 ER PT J AU Ko, J Jung, J Kim, TS Hong, YJ Park, S Park, OJ Park, SY Lim, YH Kang, KW Lee, J AF Ko, Jihoon Jung, Jaeyun Kim, Tae Seung Hong, Yong Jung Park, Sehhoon Park, Oh Joon Park, Suk Young Lim, Yeong Ho Kang, Ki Won Lee, Jeeyun TI MET gene alterations predict poor survival following chemotherapy in patients with advanced cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE next-generation sequencing; oncogene; overall survival analysis; MET; MET alterations; chemotherapy; cancer ID next-generation sequencing; oncogene; overall survival analysis; MET; MET alterations; chemotherapy; cancer AB Background: To aid in oncology drug development, we investigated MET protooncogene receptor tyrosine kinase gene aberrations in 2,239 oncology patients who underwent next-generation sequencing (NGS) in clinical practice. Materials andmethods: FromNovember 2019 to January 2021, 2,239 patientswith advanced solid tumors who visited oncology clinics underwent NGS. The NGS panel included >500 comprehensive NGS tests using archival tissue specimens. Programmed death-ligand 1(PD-L1) 22C3 assay results and clinical records regarding initial chemotherapy were available for 1,137 (50.8%) and 1,761 (78.7%) patients, respectively for overall survival (OS) analysis. Results: The 2,239 patients represented 37 types of cancer. The NGS panel included >500 genes, microsatellite instability status, tumor mutational burden, and fusions. Themost commoncancer types were colorectal (N=702),gastric (N= 481), and sarcoma (N = 180). MET aberrations were detected in 212 patients. All MET-amplified tumors had microsatellite stable status, and 8 had a high tumor mutational burden.Of 46 patientswithMET-amplified cancers, 8 had MET-positive protein expression by immunohistochemistry (2+ and 3+). MET fusion was detected in 10 patients. Partner genes of MET fusion included ST7, TFEC, LRRD1, CFTR, CAV1, PCM1, HLA-DRB1, and CAPZA2. In survival analysis, patients with amplification of MET gene fusion had shorter OS and progressionfree survival (PFS) than thosewithout. Thus, MET aberrationwas determined to be a factor of response to chemotherapy. Conclusion: Approximately 2.1% and 0.4% of patients with advanced solid tumors demonstrated MET gene amplification and fusion, respectively, and displayed a worse response to chemotherapy and significantly shorter OS and PFS than those without MET gene amplification or fusion. C1 [Ko, Jihoon] Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-OncologySeoul, South Korea. [Jung, Jaeyun] Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-OncologySeoul, South Korea. [Kim, Tae Seung] Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-OncologySeoul, South Korea. [Hong, Yong Jung] Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-OncologySeoul, South Korea. [Park, Sehhoon] Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-OncologySeoul, South Korea. [Park, Oh Joon] Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-OncologySeoul, South Korea. [Park, Suk Young] Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-OncologySeoul, South Korea. [Lim, Yeong Ho] Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-OncologySeoul, South Korea. [Kang, Ki Won] Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-OncologySeoul, South Korea. [Lee, Jeeyun] Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-OncologySeoul, South Korea. RP Lee, J (reprint author), Sungkyunkwan University, School of Medicine, Samsung Medical Center, Department of Medicine, Division of Hematology-Oncology, Seoul, South Korea. EM jyunlee@skku.edu CR Maulik G, Shrikhande A, Kijima T, Ma-P C, Morrison P, Salgia R. Role of the hepatocyte growth factor receptor, c-Met, in oncogenesis and potential for therapeutic inhibition. Cytokine Growth Factor Rev, 2002, 13(1):41–59., DOI 10. 1016/S1359-6101(01)00029-6 Lee J, Seo JW, Jun HJ, Ki CS, Park SH, Park YS, et al. Impact of met amplification on gastric cancer: possible roles as a novel prognostic marker and a potential therapeutic target. Oncol Rep, 2011, 25(6):1517–24., DOI 10.3892/or. 2011.1219 Lee SJ, Lee J, Park SH, Park JO, Lim HY, Kang WK, et al. c-MET overexpression in colorectal cancer: a poor prognostic factor for survival. Clin Colorectal Cancer, 2018, 17(3):165–9., DOI 10.1016/j.clcc.2018.02.013 Janjigian YY, Tang LH, Coit DG, Kelsen DP, Francone TD, Weiser MR, et al. MET expression and amplification in patients with localized gastric cancer. Cancer Epidemiol Biomarkers Prev, 2011, 20(5):1021–7., DOI 10.1158/1055-9965.EPI-10- 1080 An X, Wang F, Shao Q, Wang F, Wang Z, Chen C, et al. MET amplification is not rare and predicts unfavorable clinical outcomes in patients with recurrent/ metastatic gastric cancer after chemotherapy. Cancer, 2014, 120(5):675–82., DOI 10. 1002/cncr.28454 Peruzzi B, Bottaro DP. Targeting the c-Met signaling pathway in cancer. Clin Cancer Res, 2006, 1212:3657–60., DOI 10.1158/1078-0432. CCR-06-0818 Moosavi F, Giovannetti E, Peters GJ, Firuzi O. Combination of HGF/METtargeting agents and other therapeutic strategies in cancer. Crit Rev Oncol Hematol, 2021, 160:103234., DOI 10.1016/j.critrevonc.2021.103234 Lee H, Kim M, Lee H, Jung E, Yang H, Lee B, et al. MET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer, 2012, 107(2):325–33., DOI 10.1038/bjc.2012.237 Inokuchi M, Otsuki S, Fujimori Y, Sato Y, Nakagawa M, Kojima K. Clinical significance of MET in gastric cancer. World J Gastrointest Oncol, 2015, 7(11): 317–27., DOI 10.4251/wjgo.v7.i11.317 Kawakami H, Okamoto I, Arao T, Okamoto W, Matsumoto K, Taniguchi H, et al. MET amplification as a potential therapeutic target in gastric cancer. Oncotarget, 2013, 4(1):9–17., DOI 10.18632/oncotarget.718 Chen C-T, Kim H, Liska D, Gao S, Christensen J, Weiser M. MET activation mediates resistance to lapatinib inhibition of HER2-amplified gastric cancer cells. Mol Cancer Ther, 2012, 11(3):660–9., DOI 10.1158/1535-7163.MCT-11-0754 Ha SY, Lee J, Kang SY, Do IG, Ahn S, Park JO, et al. MET overexpression assessed by new interpretation method predicts gene amplification and poor survival in advanced gastric carcinomas. Mod Pathol, 2013, 26(12):1632–41., DOI 10.1038/modpathol.2013.108 Shah MA, Cho J-Y, Tan IB, Tebbutt NC, Yen CJ, Kang A, et al. A randomized phase II study of folfox with or without the MET inhibitor onartuzumab in advanced adenocarcinoma of the stomach and gastroesophageal junction. Oncologist, 2016, 21(9):1085–90., DOI 10.1634/theoncologist.2016-0038 Lee J, Kim ST, Kim K, Lee H, Kozarewa I, Mortimer PGS, et al. Tumor genomic profiling guides patients with metastatic gastric cancer to targeted treatment: the VIKTORY umbrella trial. Cancer Discov, 2019, 9(10):1388–405., DOI 10.1158/2159-8290.CD-19-0442 Bang YJ, Su WC, Schuler M, Nam DH, Lim WT, Bauer TM, et al. Phase 1 study of capmatinib in MET-positive solid tumor patients: dose escalation and expansion of selected cohorts. Cancer Sci, 2020, 111(2):536–47., DOI 10.1111/cas. 14254 Lorenzato A, Olivero M, Patane S, Rosso E, Oliaro A, Comoglio PM, et al. Novel somatic mutations of the MET oncogene in human carcinoma metastases activating cell motility and invasion. Cancer Res, 2002, 62(23):7025–30. Raghav KPS, Gonzalez-Angulo AM, Blumenschein GR, Jr. Role of HGF/MET axis in resistance of lung cancer to contemporary management. Transl Lung Cancer Res, 2012, 1(3):179–93., DOI 10.3978/j.issn.2218-6751.2012.09.04 Mukai Y, Ueno H. Establishment and implementation of cancer genomic medicine in japan. Cancer Sci, 2021, 112(3):970–7., DOI 10.1111/ cas.14754 Fujii S, Magliocco AM, Kim J, Okamoto W, Kim JE, Sawada K, et al. International harmonization of provisional diagnostic criteria for ERBB2- amplified metastatic colorectal cancer allowing for screening by nextgeneration sequencing panel. JCO Precis Oncol, 2020, 4:6–19., DOI 10.1200/ PO.19.00154 Pestinger V, Smith M, Sillo T, Findlay JM, Laes JF, Martin G, et al. Use of an integrated pan-cancer oncology enrichment next-generation sequencing assay to measure tumour mutational burden and detect clinically actionable variants. Mol Diagn Ther, 2020, 24(3):339–49., DOI 10.1007/s40291-020-00462-x Karasaki T, Nakajima J, Kakimi K.Neoantigens and whole-exome sequencing. Gan To Kagaku Ryoho, 2016, 43(7):791–7. Comoglio PM, Giordano S, Trusolino L. Drug development of MET inhibitors: targeting oncogene addiction and expedience. Nat Rev Drug Discov, 2008, 7(6):504–16., DOI 10.1038/nrd2530 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610697 EP 1610706 DI 10.3389/pore.2022.1610697 PG 10 ER PT J AU Huang, Q Shen, YJ Hsueh, ChY Zhang, YF Yuan, XH Zhou, YJ Li, JY Lin, L Wu, ChP Hu, ChY AF Huang, Qiang Shen, Yu-Jie Hsueh, Chi-Yao Zhang, Yi-Fan Yuan, Xiao-Hui Zhou, Yu-Juan Li, Jiao-Yu Lin, Lan Wu, Chun-Ping Hu, Chun-Yan TI Plasma Extracellular Vesicles-Derived miR-99a-5p: A Potential Biomarker to Predict Early Head and Neck Squamous Cell Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; HNSCC; HPV; plasma extracellular vesicles; miR-99a-5p ID biomarker; HNSCC; HPV; plasma extracellular vesicles; miR-99a-5p AB Purpose: This study aimed to investigate the applicability of plasma extracellular vesicles (EVs) miR-99a-5p as a potential head and neck squamous cell carcinoma (HNSCC) diagnostic biomarker. Methods: The miRNA expression of HNSCC tissue and plasma EVs were profiled by small RNA sequencing. qRT-PCR was performed to detect miR-99a-5p expression in HNSCC (n = 93) and benign disease (n = 39) plasma EVs and formalin-fixed and paraffinembedded (FFPE) tissue (n = 110). We constructed receiver-operating characteristic curves to investigate the diagnostic efficiency of plasma EVs miR-99a-5p. Results: Tumor tissue exhibited lower miR-99a-5p than para-tumor tissue. Patients with high miR-99a-5p expression exhibited significantly more p16 positive status. In contrast, HNSCC plasma EVs harbored more miR-99a-5p than the benign disease group. Plasma EVs miR-99a-5p distinguished HNSCC with area under the curve (AUC) of 0.7494 (95% CI: 0.6692–0.8296; p < 0.0001), with 61.54% sensitivity and 75.27% specificity, respectively. Furthermore, plasma EVs miR-99a-5p also distinguished early HNSCC with AUC of 0.7394 (95% CI: 0.6284–0.8504; p = 0.0002), with 79.07% sensitivity and 61.54% specificity, respectively. Conclusion: Plasma EVs miR-99a-5p is a potential biomarker for predicting early HNSCC. C1 [Huang, Qiang] Fudan University, Eye & ENT Hospital, Department of OtorhinolaryngologyShanghai, China. [Shen, Yu-Jie] Fudan University, Eye & ENT Hospital, Department of OtorhinolaryngologyShanghai, China. [Hsueh, Chi-Yao] Fudan University, Eye & ENT Hospital, Department of OtorhinolaryngologyShanghai, China. [Zhang, Yi-Fan] Fudan University, Eye & ENT Hospital, Department of OtorhinolaryngologyShanghai, China. [Yuan, Xiao-Hui] Fudan University, Eye & ENT Hospital, Department of OtorhinolaryngologyShanghai, China. [Zhou, Yu-Juan] Fudan University, Eye & ENT Hospital, Department of OtorhinolaryngologyShanghai, China. [Li, Jiao-Yu] Shanghai Jiaotong University School of Medicine, Xinhua Hospital, Department of PediatricShanghai, China. [Lin, Lan] Fudan University, Eye & ENT Hospital, Department of PathologyShanghai, China. [Wu, Chun-Ping] Fudan University, Eye & ENT Hospital, Department of OtorhinolaryngologyShanghai, China. [Hu, Chun-Yan] Fudan University, Eye & ENT Hospital, Department of PathologyShanghai, China. RP Hu, ChY (reprint author), Fudan University, Eye & ENT Hospital, Department of Pathology, Shanghai, China. EM huchy2003@163.com CR Cramer JD, Burtness B, Le QT, Ferris RL. The Changing Therapeutic Landscape of Head and Neck Cancer. Nat Rev Clin Oncol, 2019, 16(11): 669–83., DOI 10.1038/s41571-019-0227-z Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE, Grandis JR. Head and Neck Squamous Cell Carcinoma. Nat Rev Dis Primers, 2020, 6(1): 92., DOI 10.1038/s41572-020-00224-3 Lin H, Wang T, Heng Y, Zhu X, Zhou L, Zhang M, et al. Risk Stratification of Postoperative Recurrence in Hypopharyngeal Squamous-Cell Carcinoma Patients with Nodal Metastasis. J Cancer Res Clin Oncol, 2021, 147(3): 803–11., DOI 10.1007/s00432-020-03337-0 Heng Y, Zhang D, Zhou L, Zhang M, Wu C, Tao L. Assessment and Treatment Strategies for Occult Contralateral Lymph Node Metastasis in Hypopharyngeal Squamous Cell Carcinoma Patients with Ipsilateral Node- Positive Necks. Oral Oncol, 2021, 114:105183., DOI 10.1016/j.oraloncology. 2021.105183 Moller A, Lobb RJ. The Evolving Translational Potential of Small Extracellular Vesicles in Cancer. Nat Rev Cancer, 2020, 20(12):697–709., DOI 10.1038/ s41568-020-00299-w LeBleu VS, Kalluri R. Exosomes as a Multicomponent Biomarker Platform in Cancer. Trends Cancer, 2020, 6(9):767–74., DOI 10.1016/j.trecan.2020.03.007 Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosomemediated Transfer of mRNAs and microRNAs Is a Novel Mechanism of Genetic Exchange between Cells. Nat Cel Biol, 2007, 9(6):654–9., DOI 10.1038/ ncb1596 Taylor DD, Gercel-Taylor C. MicroRNA Signatures of Tumor-Derived Exosomes as Diagnostic Biomarkers of Ovarian Cancer. Gynecol Oncol, 2008, 110(1):13–21., DOI 10.1016/j.ygyno.2008.04.033 Guo S, Qin H, Liu K, Wang H, Bai S, Liu S, et al. Blood Small Extracellular Vesicles Derived miRNAs to Differentiate Pancreatic Ductal Adenocarcinoma from Chronic Pancreatitis. Clin Transl Med, 2021, 11(9):e520., DOI 10.1002/ ctm2.520 Gao S, Guo W, Liu T, Liang N,Ma Q, Gao Y, et al. Plasma Extracellular Vesicle microRNA Profiling and the Identification of a Diagnostic Signature for Stage I Lung Adenocarcinoma. Cancer Sci, 2022, 113(2):648–59., DOI 10.1111/cas. 15222 Tamai M, Tatarano S, Okamura S, Fukumoto W, Kawakami I, Osako Y, et al. microRNA-99a-5p Induces Cellular Senescence in Gemcitabine-Resistant Bladder Cancer by Targeting SMARCD1. Mol Oncol, 2022, 16(6):1329–46., DOI 10.1002/1878-0261.13192 Liu Z, Wu K, Gu S, Wang W, Xie S, Lu T, et al. A Methyltransferase-like 14/ miR-99a-5p/tribble 2 Positive Feedback Circuit Promotes Cancer Stem Cell Persistence and Radioresistance via Histone Deacetylase 2-mediated Epigenetic Modulation in Esophageal Squamous Cell Carcinoma. Clin Transl Med, 2021, 11(9):e545., DOI 10.1002/ctm2.545 Sun X, Yan H. MicroRNA-99a-5p Suppresses Cell Proliferation, Migration, and Invasion by Targeting Isoprenylcysteine Carboxylmethyltransferase in Oral Squamous Cell Carcinoma. J Int Med Res, 2021, 49(5):300060520939031., DOI 10.1177/0300060520939031 Guo T, Wang Y, Jia J, Mao X, Stankiewicz E, Scandura G, et al. The Identification of Plasma Exosomal miR-423-3p as a Potential Predictive Biomarker for Prostate Cancer Castration-Resistance Development by Plasma Exosomal miRNA Sequencing. Front Cel Dev Biol, 2020, 8:602493., DOI 10.3389/fcell.2020.602493 Zheng D, Zhu Y, Zhang J, ZhangW,Wang H, Chen H, et al. Identification and Evaluation of Circulating Small Extracellular Vesicle microRNAs as Diagnostic Biomarkers for Patients with Indeterminate Pulmonary Nodules. J Nanobiotechnology, 2022, 20(1):172., DOI 10.1186/s12951-022-01366-0 Huang Q, Hsueh CY, Shen YJ, Guo Y, Huang JM, Zhang YF, et al. Small Extracellular Vesicle-Packaged TGFβ1 Promotes the Reprogramming of normal Fibroblasts into Cancer-Associated Fibroblasts by Regulating Fibronectin in Head and Neck Squamous Cell Carcinoma. Cancer Lett, 2021, 517:1–13., DOI 10.1016/j.canlet.2021.05.017 Huang Q, Hsueh C, Guo Y, Wu X, Li J, Zhou L. Lack of miR-1246 in Small Extracellular Vesicle Blunts Tumorigenesis of Laryngeal Carcinoma Cells by Regulating Cyclin G2. IUBMB Life, 2020, 72(7):1491–503., DOI 10.1002/iub. 2274 Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: Decoding miRNAceRNA, miRNA-ncRNA and Protein–RNA Interaction Networks from Large- Scale CLIP-Seq Data. Nucleic Acids Res, 2014, 42(D1):D92–7., DOI 10.1093/nar/ gkt1248 Chen Y, Yao J, Qin Y, Hu K,Wu F, Fang Y. Biological Role and Clinical Value of miR-99a-5p in Head and Neck Squamous Cell Carcinoma, HNSCC ): A Bioinformatics-based Study. FEBS Open Bio, 2018, 8(8):1280–98., DOI 10.1002/ 2211-5463.12478 Kalluri R, LeBleu VS. The Biology, Function, and Biomedical Applications of Exosomes. Science, 2020, 367(6478):eaau6977., DOI 10.1126/science.aau6977 Garrido-Cano I, Constancio V, Adam-Artigues A, Lameirinhas A, Simon S, Ortega B, et al. Circulating miR-99a-5p Expression in Plasma: A Potential Biomarker for Early Diagnosis of Breast Cancer. Int J Mol Sci, 2020, 21(19): 7427., DOI 10.3390/ijms21197427 Bigagli E, Locatello LG, Di Stadio A, Maggiore G, Valdarnini F, Bambi F, et al. Extracellular Vesicles miR-210 as a Potential Biomarker for Diagnosis and Survival Prediction of Oral Squamous Cell Carcinoma Patients. J Oral Pathol Med, 2022, 51(4):350–7., DOI 10.1111/jop.13263 Panvongsa W, Siripoon T, Worakitchanon W, Arsa L, Trachu N, Jinawath N, et al. Plasma Extracellular Vesicle microRNA-491-5p as Diagnostic and Prognostic Marker for Head and Neck Squamous Cell Carcinoma. Cancer Sci, 2021, 112(10):4257–69., DOI 10.1111/cas.15067 Zhang Q, Chen Y, Hu SQ, Pu YM, Zhang K, Wang YX. A HPV16-Related Prognostic Indicator for Head and Neck Squamous Cell Carcinoma. Ann Transl Med, 2020, 8(22):1492., DOI 10.21037/atm-20-6338 Torres A, Torres K, Pesci A, Ceccaroni M, Paszkowski T, Cassandrini P, et al. Deregulation of miR-100, miR-99a and miR-199b in Tissues and Plasma Coexists with Increased Expression of mTOR Kinase in Endometrioid Endometrial Carcinoma. BMC Cancer, 2012, 12:369., DOI 10.1186/1471- 2407-12-369 van Schooneveld E, Wouters MC, Van der Auwera I, Peeters DJ, Wildiers H, Van Dam PA, et al. Expression Profiling of Cancerous and normal Breast Tissues Identifies microRNAs that Are Differentially Expressed in Serum from Patients with, Metastatic, Breast Cancer and Healthy Volunteers. Breast Cancer Res, 2012, 14(1):R34., DOI 10.1186/bcr3127 Cuk K, Zucknick M, Heil J, Madhavan D, Schott S, Turchinovich A, et al. Circulating microRNAs in Plasma as Early Detection Markers for Breast Cancer. Int J Cancer, 2013, 132(7):1602–12., DOI 10.1002/ijc.27799 Chan M, Liaw CS, Ji SM, Tan HH, Wong CY, Thike AA, et al. Identification of Circulating microRNA Signatures for Breast Cancer Detection. Clin Cancer Res, 2013, 19(16):4477–87., DOI 10.1158/1078-0432.CCR-12-3401 Teng Y, Ren Y, Hu X, Mu J, Samykutty A, Zhuang X, et al. MVP-Mediated Exosomal Sorting of miR-193a Promotes colon Cancer Progression. Nat Commun, 2017, 8:14448., DOI 10.1038/ncomms14448 Xu X, Liu Y, Li Y, Chen H, Zhang Y, Liu J, et al. Selective Exosome Exclusion of miR-375 by Glioma Cells Promotes Glioma Progression by Activating the CTGF-EGFR Pathway. J Exp Clin Cancer Res, 2021, 40(1):16., DOI 10.1186/ s13046-020-01810-9 Qi Y, Jin C, Qiu W, Zhao R, Wang S, Li B, et al. The Dual Role of Glioma Exosomal microRNAs: Glioma Eliminates Tumor Suppressor miR-1298-5p via Exosomes to Promote Immunosuppressive Effects of MDSCs. Cell Death Dis, 2022, 13(5):426., DOI 10.1038/s41419-022-04872-z Temoche-Diaz MM, Shurtleff MJ, Nottingham RM, Yao J, Fadadu RP, Lambowitz AM, et al. Distinct Mechanisms of microRNA Sorting into Cancer Cell-Derived Extracellular Vesicle Subtypes. eLife, 2019, 8:e47544., DOI 10.7554/eLife.47544 Shurtleff MJ, Temoche-Diaz MM, Karfilis KV, Ri S, Schekman R. Y-Box Protein 1 Is Required to Sort microRNAs into Exosomes in Cells and in a Cellfree Reaction. eLife, 2019, 5:e19276., DOI 10.7554/eLife.19276 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610699 EP 1610708 DI 10.3389/pore.2022.1610699 PG 10 ER PT J AU Xiong, S Jin, L Zeng, Ch Ma, H Xie, L Liu, Sh AF Xiong, Siping Jin, Long Zeng, Chao Ma, Hongmei Xie, Linying Liu, Shuguang TI An innovative pyroptosis-related long-noncoding-RNA signature predicts the prognosis of gastric cancer via affecting immune cell infiltration landscape SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; gastric cancer; pyroptosis; overall survival; lncRNA signature ID prognosis; gastric cancer; pyroptosis; overall survival; lncRNA signature AB Background: Gastric cancer (GC) is a worldwide popular malignant tumor. However, the survival rate of advanced GC remains low. Pyroptosis and long non-coding RNAs (lncRNAs) are important in cancer progression. Thus, we aimed to find out a pyroptosis-related lncRNAs (PRLs) signature and use it to build a practical risk model with the purpose to predict the prognosis of patients with GC. Methods: Univariate Cox regression analysis was used to identify PRLs linked to GC patient’s prognosis. Subsequently, to construct a PRLs signature, the least absolute shrinkage and selection operator regression, and multivariate Cox regression analysis were used. Kaplan–Meier analysis, principal component analysis, and receiver operating characteristic curve analysis were performed to assess our novel lncRNA signature. The correlation between risk signature and clinicopathological features was also examined. Finally, the relationship of pyroptosis and immune cells were evaluated through the CIBERSORT tool and single-sample lncRNA set enrichment analysis (ssGSEA). Results: A PRLs signature comprising eight lncRNAs was discerned as a selfdetermining predictor of prognosis. GC patients were sub-divided into high-risk and low-risk groups via this risk-model. Stratified analysis of different clinical factors also displayed that the PRLs signature was a good prognosis factor. According to the risk score and clinical characteristics, a nomogram was established. Moreover, the difference between the groups is significance in immune cells and immune pathways. Conclusion: This study established an effective prognostic signature consist of eight PRLs in GC, and constructed an efficient nomogram model. Further, the PRLs correlated with immune cells and immune pathways. C1 [Xiong, Siping] Sun Yat-sen University, The Eighth Affiliated Hospital, Department of PathologyShenzhen, Guangdong, China. [Jin, Long] Fujian Medical University, Shengli Clinical Medical College, Department of PathologyFuzhou, Fujian, China. [Zeng, Chao] Sun Yat-sen University, The Eighth Affiliated Hospital, Department of PathologyShenzhen, Guangdong, China. [Ma, Hongmei] Sun Yat-sen University, The Eighth Affiliated Hospital, Department of PathologyShenzhen, Guangdong, China. [Xie, Linying] Sun Yat-sen University, The Eighth Affiliated Hospital, Department of PathologyShenzhen, Guangdong, China. [Liu, Shuguang] Sun Yat-sen University, The Eighth Affiliated Hospital, Department of PathologyShenzhen, Guangdong, China. RP Liu, Sh (reprint author), Sun Yat-sen University, The Eighth Affiliated Hospital, Department of Pathology, Shenzhen, China. EM liushg3@mail.sysu.edu.cn CR Han T, Xu D, Zhu J, Li J, Liu L, Deng Y. Identification of a robust signature for clinical outcomes and immunotherapy response in gastric cancer: Based on N6- methyladenosine related long noncoding RNAs. Cancer Cel Int, 2021, 21:432., DOI 10.1186/s12935-021-02146-w Kim Y, Choi JW, Lee JH, Kim YS. Spindle assembly checkpoint MAD2 and CDC20 overexpressions and cell-in-cell formation in gastric cancer and its precursor lesions. Hum Pathol, 2019, 85:174–83., DOI 10.1016/j.humpath.2018. 10.029 Burrus O. ["Top secret. or bum secret"] Rev Infirm, 1986, 36:46–8. Smyth EC, NilssonM, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet, London, England,, 2020, 396:635–48., DOI 10.1016/S0140-6736(20)31288-5 Gao Y, Qi W, Sun L, Lv J, Qiu W, Liu S. FOXO3 inhibits human gastric adenocarcinoma, AGS, cell growth by promoting autophagy in an acidic microenvironment. Cell Physiol Biochem, 2018, 49:335–48., DOI 10.1159/000492884 Nie Y, Wu K, Yu J, Liang Q, Cai X, Shang Y, et al. A global burden of gastric cancer: The major impact of China. Expert Rev Gastroenterol Hepatol, 2017, 11: 651–61., DOI 10.1080/17474124.2017.1312342 Diederichs S. The four dimensions of noncoding RNA conservation. Trends Genet, 2014, 30:121–3., DOI 10.1016/j.tig.2014.01.004 Ji P, Diederichs S, Wang W, Boing S, Metzger R, Schneider PM, et al. MALAT-1, a novel noncoding RNA, and thymosin beta4 predictmetastasis and survival in early-stage non-small cell lung cancer. Oncogene, 2003, 22:8031–41., DOI 10.1038/sj.onc.1206928 Yan X, Hu Z, Feng Y, Hu X, Yuan J, Zhao SD, et al. Comprehensive genomic characterization of long non-coding RNAs across human cancers. Cancer Cell, 2015, 28:529–40., DOI 10.1016/j.ccell.2015.09.006 Matkovich SJ, Edwards JR, Grossenheider TC, de Guzman Strong C, Dorn GW. Epigenetic coordination of embryonic heart transcription by dynamically regulated long noncoding RNAs. Proc Natl Acad Sci U S A, 2014, 111:12264–9., DOI 10.1073/pnas.1410622111 Ounzain S, Micheletti R, Arnan C, Plaisance I, Cecchi D, Schroen B, et al. CARMEN, a human super enhancer-associated long noncoding RNA controlling cardiac specification, differentiation and homeostasis. J Mol Cel Cardiol, 2015, 89: 98–112., DOI 10.1016/j.yjmcc.2015.09.016 Kumarswamy R, Bauters C, Volkmann I, Maury F, Fetisch J, Holzmann A, et al. Circulating long noncoding RNA, LIPCAR, predicts survival in patients with heart failure. Circ Res, 2014, 114:1569–75., DOI 10.1161/CIRCRESAHA.114.303915 Nasrollahzadeh-Khakiani M, Emadi-Baygi M, Schulz WA, Nikpour P. Long noncoding RNAs in gastric cancer carcinogenesis and metastasis. Brief Funct Genomics, 2017, 16:129–45., DOI 10.1093/bfgp/elw011 Malla RR, Padmaraju V, Marni R, Kamal MA. Natural products: Potential targets of TME related long non-coding RNAs in lung cancer. Phytomedicine, 2021, 93:153782., DOI 10.1016/j.phymed.2021.153782 Shi T, Morishita A, Kobara H, Masaki T. The role of long non-coding RNA and microRNA networks in hepatocellular carcinoma and its tumor microenvironment. Int J Mol Sci, 2021, 22:10630., DOI 10.3390/ ijms221910630 Schmitz SU, Grote P, Herrmann BG. Mechanisms of long noncoding RNA function in development and disease. Cell Mol Life Sci, 2016, 73:2491–509., DOI 10. 1007/s00018-016-2174-5 Yousefi L, Osquee HO, Ghotaslou R, Rezaee MA, Pirzadeh T, Sadeghi J, et al. Dysregulation of lncRNA in Helicobacter pylori-infected gastric cancer cells. Biomed Res Int, 2021, 2021:6911734., DOI 10.1155/2021/6911734 Chen R, Yang M, Huang W, Wang B. Cascades between miRNAs, lncRNAs and the NF-κB signaling pathway in gastric cancer, Review). Exp Ther Med, 2021, 22:769., DOI 10.3892/etm.2021.10201 Li Y, Lu L,Wu X, Li Q, Zhao Y, Du F, et al. The multifaceted role of long noncoding RNA in gastric cancer: Current status and future perspectives. Int J Biol Sci, 2021, 17:2737–55., DOI 10.7150/ijbs.61410 Burdette BE, Esparza AN, Zhu H, Wang S. Gasdermin D in pyroptosis. Acta Pharm Sin B, 2021, 11:2768–82., DOI 10.1016/j.apsb.2021.02.006 AlMamun A, Mimi AA, Aziz MA, Zaeem M, Ahmed T, Munir F, et al. Role of pyroptosis in cancer and its therapeutic regulation. Eur J Pharmacol, 2021, 910: 174444., DOI 10.1016/j.ejphar.2021.174444 Miao EA, Rajan JV, Aderem A. Caspase-1-induced pyroptotic cell death. Immunol Rev, 2011, 243:206–14., DOI 10.1111/j.1600-065X.2011.01044.x Broz P, Pelegrin P, Shao F. The gasdermins, a protein family executing cell death and inflammation. Nat Rev Immunol, 2020, 20:143–57., DOI 10.1038/s41577- 019-0228-2 Zhou CB, Fang JY. The role of pyroptosis in gastrointestinal cancer and immune responses to intestinal microbial infection. Biochim Biophys Acta Rev Cancer, 2019, 1872:1–10., DOI 10.1016/j.bbcan.2019.05.001 He T, Yuan C, Zhao C. Long intragenic non-coding RNA p53-induced transcript, LINC-PINT, as a novel prognosis indicator and therapeutic target in cancer. Biomed Pharmacother, 2021, 143:112127., DOI 10.1016/j.biopha.2021. 112127 Xue C, Cai X, Jia J. Long non-coding RNA double homeobox A pseudogene 8: A novel oncogenic propellant in human cancer. Front Cel Dev Biol, 2021, 9:709069., DOI 10.3389/fcell.2021.709069 Han X, Liu Z. Long noncoding RNA JPX promotes gastric cancer progression by regulating CXCR6 and autophagy via inhibiting miR197. Mol Med Rep, 2021, 23:60., DOI 10.3892/mmr.2020.11698 Li G, Li X, Yuan C, Zhou C, Li X, Li J, et al. Long non-coding RNA JPX contributes to tumorigenesis by regulating miR-5195-3p/VEGFA in non-small cell lung cancer. Cancer Manag Res, 2021, 13:1477–89., DOI 10.2147/CMAR.S255317 Chen X, Yang J,Wang Y. LncRNA JPX promotes cervical cancer progression by modulating miR-25-3p/SOX4 axis. Cancer Cel Int, 2020, 20:441., DOI 10.1186/ s12935-020-01486-3 Wang XQ, Xu H, Wang CH, Xie H. Long non-coding RNA GNAS-AS1 promotes cell migration and invasion via regulating Wnt/β-catenin pathway in nasopharyngeal carcinoma. Eur Rev Med Pharmacol Sci, 2020, 24:3077–84., DOI 10. 26355/eurrev_202003_20672 Li Z, Feng C, Guo J, Hu X, Xie D. GNAS-AS1/miR-4319/NECAB3 axis promotes migration and invasion of non-small cell lung cancer cells by altering macrophage polarization. Funct Integr Genomics, 2020, 20:17–28., DOI 10.1007/ s10142-019-00696-x Pang B, Wang Q, Ning S, Wu J, Zhang X, Chen Y, et al. Landscape of tumor suppressor long noncoding RNAs in breast cancer. J Exp Clin Cancer Res, 2019, 38: 79., DOI 10.1186/s13046-019-1096-0 Persson H, Kwon AT, Ramilowski JA, Silberberg G, Soderhall C, Orsmark- Pietras C, et al. Transcriptome analysis of controlled and therapy-resistant childhood asthma reveals distinct gene expression profiles. J Allergy Clin Immunol, 2015, 136:638–48., DOI 10.1016/j.jaci.2015.02.026 Huang R, Zhou L, Chi Y, Wu H, Shi L. LncRNA profile study reveals a sevenlncRNA signature predicts the prognosis of patients with colorectal cancer. Biomark Res, 2020, 8:8., DOI 10.1186/s40364-020-00187-3 Zhang H, Zhao L, Li S, Wang J, Feng C, Li T, et al. N6-Methylandenosine- Related lncRNAs in tumor microenvironment are potential prognostic biomarkers in colon cancer. Front Oncol, 2021, 11:697949., DOI 10.3389/fonc.2021.697949 Liu W, Feng Q, Liao W, Li E, Wu L. TUG1 promotes the expression of IFITM3 in hepatocellular carcinoma by competitively binding to miR-29a. J Cancer, 2021, 12:6905–20., DOI 10.7150/jca.57477 Jin Y, Cao J, Hu X, Cheng H. Long noncoding RNA TUG1 upregulates VEGFA to enhance malignant behaviors in stomach adenocarcinoma by sponging miR-29c-3p. J Clin Lab Anal, 2021, 35:e24106., DOI 10.1002/jcla.24106 Yu G, Zhou H, Yao W, Meng L, Lang B. lncRNA TUG1 promotes cisplatin resistance by regulating CCND2 via epigenetically silencing miR- 194-5p in bladder cancer. Mol Ther Nucleic Acids, 2019, 16:257–71., DOI 10. 1016/j.omtn.2019.02.017 Wang F, Li G, Ning J, Chen L, Xu H, Kong X, et al. Alcohol accumulation promotes esophagitis via pyroptosis activation. Int J Biol Sci, 2018, 14:1245–55., DOI 10.7150/ijbs.24347 Chu Q, Jiang Y, Zhang W, Xu C, Du W, Tuguzbaeva G, et al. Pyroptosis is involved in the pathogenesis of human hepatocellular carcinoma. Oncotarget, 2016, 7:84658–65., DOI 10.18632/oncotarget.12384 Wang Y, Yin B, Li D, Wang G, Han X, Sun X. GSDME mediates caspase-3- dependent pyroptosis in gastric cancer. Biochem Biophys Res Commun, 2018, 495: 1418–25., DOI 10.1016/j.bbrc.2017.11.156 Mitra SA, Mitra AP, Triche TJ. A central role for long non-coding RNA in cancer. Front Genet, 2012, 3:17., DOI 10.3389/fgene.2012.00017 Prensner JR, Chinnaiyan AM. The emergence of lncRNAs in cancer biology. Cancer Discov, 2011, 1:391–407., DOI 10.1158/2159-8290.CD-11-0209 Hung T, Chang HY. Long noncoding RNA in genome regulation: Prospects and mechanisms. RNA Biol, 2010, 7:582–5., DOI 10.4161/rna.7.5. 13216 Li C, Liang G, Yao W, Sui J, Shen X, Zhang Y, et al. Differential expression profiles of long non-coding RNAs reveal potential biomarkers for identification of human gastric cancer. Oncol Rep, 2016, 35:1529–40., DOI 10.3892/or.2015.4531 Winkler L, Dimitrova N. A mechanistic view of long noncoding RNAs in cancer. Wiley Interdiscip Rev RNA, 2021, 13:e1699., DOI 10.1002/wrna.1699 Khawar MB, Hamid SE, Jan T, Abbasi MH, Idnan M, Sheikh N. Diagnostic, prognostic and therapeutic potential of long noncoding RNAs in cancer. Mol Biol Rep, 2022, 49:2311–9., DOI 10.1007/s11033-022- 07180-z Tan Y, Sun R, Liu L, Yang D, Xiang Q, Li L, et al. Tumor suppressor DRD2 facilitates M1 macrophages and restricts NF-κB signaling to trigger pyroptosis in breast cancer. Theranostics, 2021, 11:5214–31., DOI 10.7150/thno. 58322 Chen Y, Sun Z, Chen W, Liu C, Chai R, Ding J, et al. The immune subtypes and landscape of gastric cancer and to predict based on the wholeslide images using deep learning. Front Immunol, 2021, 12:685992., DOI 10. 3389/fimmu.2021.685992 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610712 EP 1610726 DI 10.3389/pore.2022.1610712 PG 15 ER PT J AU Paszt, A Ottlakan, A Abraham, Sz Simonka, Zs Vas, M Maraz, A Szepes, Z Tiszlavicz, L Nyari, T Olah, J Lazar, Gy AF Paszt, Attila Ottlakan, Aurel Abraham, Szabolcs Simonka, Zsolt Vas, Marton Maraz, Aniko Szepes, Zoltan Tiszlavicz, Laszlo Nyari, Tibor Olah, Judit Lazar, Gyorgy TI Clinical benefits of oral capecitabine over intravenous 5-fluorouracyl regimen in case of neoadjuvant chemoradiotherapy followed by surgery for locally advanced rectal cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE surgery; chemoradiotherapy; neoadjuvant treatment; capecitabine; advanced rectal cancer ID surgery; chemoradiotherapy; neoadjuvant treatment; capecitabine; advanced rectal cancer AB Background: During the last decade, one of the most important treatment options for locally advanced, potencially resectable rectal tumours was neoadjuvant chemoradiotherapy (CRT) followed by surgery. Methods: Effects of the neoadjuvant treatment on surgical outcomes were retrospectively analysed in 185 patients with stage T2–T4 and N0–2, resectable rectal tumour among two patient groups defined by radiosensitizer agents. Group 1 (n = 94) involved radiotherapy (RT) with 50.4 Gy total dose (25 × 1.8 Gy + 3 × 1.8 Gy tumour bed boost), and intravenous 5-fluorouracil (5-FU) (350 mg/ m2) with leucovorin (20 mg/m2) on the 1–5 and 21–25 days, while Group 2 (n = 91) RT and orally administrated capecitabine (daily 2 × 825 mg/m2) on RT days. Surgery was carried out after 8–10 weeks. Side effects, perioperative complications, type of surgery, number of removed regional lymph nodes, resection margins and tumour regression grade (TRG) were analysed. Results: More favourable side effects were observed in Group 2. Despite the same rate of diarrhoea (Group 1 vs. Group 2: 54.3% vs. 56.0%), Grade 2–3 diarrhoea ratio was lower (p = 0.0352) after capecitabine (Group 2). Weight loss occurred in 17.0% and 2.2% (p = 0.00067), while nausea and vomiting was described in 38.3% and 15.4% (p = 0.00045) with 5-FU treatment and capecitabine respectively. Anaemia was observed in 33.0% and 22.0% (p = 0.0941). Complete tumour regression occurred in 25.3% after oral- and 13.8% after intravenous treatment (p = 0.049). Ratio of sphincter preservation was higher with laparoscopy than open surgery (72.3% vs. 39.7%) (p = 0.00001). Conclusion: The study confirms advantages of neoadjuvant chemoradiotherapy with oral capecitabine for rectal tumours, such as more favourable side effect profile and overall clinical outcome, with increased rate of complete tumour regression. C1 [Paszt, Attila] University of Szeged, Department of SurgerySzeged, Hungary. [Ottlakan, Aurel] University of Szeged, Department of SurgerySzeged, Hungary. [Abraham, Szabolcs] University of Szeged, Department of SurgerySzeged, Hungary. [Simonka, Zsolt] University of Szeged, Department of SurgerySzeged, Hungary. [Vas, Marton] University of Szeged, Department of SurgerySzeged, Hungary. [Maraz, Aniko] University of Szeged, Department of OncotherapySzeged, Hungary. [Szepes, Zoltan] University of Szeged, 1st Department of MedicineSzeged, Hungary. [Tiszlavicz, Laszlo] University of Szeged, Department of PathologySzeged, Hungary. [Nyari, Tibor] University of Szeged, Department of Medical InformaticsSzeged, Hungary. [Olah, Judit] University of Szeged, Department of OncotherapySzeged, Hungary. [Lazar, Gyorgy] University of Szeged, Department of SurgerySzeged, Hungary. RP Paszt, A (reprint author), University of Szeged, Department of Surgery, Szeged, Hungary. EM paszt.attila@med.u-szeged.hu CR RohMS, Colangelo LH, O’Connell MJ, Yothers G, Deutsch M, Allegra CJ, et al. Preoperative multimodality therapy improves disease-free survival in patients with carcinoma of the rectum: NSABP R-03. J Clin Oncol, 2009, 27:5124–30., DOI 10. 1200/JCO.2009.22.0467 Sauer R, Liersch T, Merkel S, Fietkau R, Hohenberger W, Hess C, et al. Preoperative versus postoperative chemoradiotherapy for locally advanced rectal cancer: Results of the German CAO/ARO/AIO-94 randomized phase III trial after a median follow-up of 11 years. J Clin Oncol, 2012, 30:1926–33., DOI 10.1200/JCO. 2011.40.1836 Azria D, Doyen J, Jarlier M, Martel-Lafay I, Hennequin C, Etienne P, et al. Late toxicities and clinical outcome at 5 years of the ACCORD 12/0405-PRODIGE 02 trial comparing two neoadjuvant chemoradiotherapy regimens for intermediaterisk rectal cancer. Ann Oncol, 2017, 28:2436–42., DOI 10.1093/annonc/mdx351 Sauer R, Becker H, Hohenberger W, Rodel C, Wittekind C, Fietkau R, et al. Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med, 2004, 351:1731–40., DOI 10.1056/NEJMoa040694 Erlandsson J, Holm T, Pettersson D, Berglund A, Cedermark B, Radu C, et al. Optimal fractionation of preoperative radiotherapy and timing to surgery for rectal cancer, Stockholm III): A multicentre, randomised, non-blinded, phase 3, noninferiority trial. Lancet Oncol, 2017, 18:336–46., DOI 10.1016/S1470-2045(17, 30086-4 Marechal R, Vos B, Polus M, Delaunoit T, Peeters M, Demetter P, et al. Short course chemotherapy followed by concomitant chemoradiotherapy and surgery in locally advanced rectal cancer: A randomized multicentric phase II study. Ann Oncol, 2012, 23:1525–30., DOI 10.1093/annonc/mdr473 Cedermark B, Dahlberg M, Glimelius B, Pahlman L, Rutqvist LE, Wilking N, et al. Improved survival with preoperative radiotherapy in resectable rectal cancer. N Engl J Med, 1997, 336:980–7., DOI 10.1056/NEJM199704033361402 Feeney G, Sehgal R, Sheehan M, Hogan A, Regan M, Joyce M, et al. Neoadjuvant radiotherapy for rectal cancer management. World J Gastroenterol, 2019, 25:4850–69., DOI 10.3748/wjg.v25.i33.4850 Van Cutsem E, Twelves C, Cassidy J, Allman D, BajEtta E, Boyer M, et al. Oral capecitabine compared with intravenous fluorouracil plus leucovorin in patients with metastatic colorectal cancer: Results of a large phase III study. J Clin Oncol, 2001, 19:4097–106., DOI 10.1200/JCO.2001.19.21.4097 Cassidy J, Twelves C, Van CutsEm E, Hoff P, BajEtta E, Boyer M, et al. Firstline oral capecitabine therapy in metastatic colorectal cancer: A favorable safety profile compared with intravenous 5-fluorouracil/leucovorin. Ann Oncol, 2002, 13: 566–75., DOI 10.1093/annonc/mdf089 Eisterer W, Piringer G, DE Vries A, Ofner D, Greil R, Tschmelitsch J, et al. Neoadjuvant chemotherapy with capecitabine, oxaliplatin and bevacizumab followed by concomitant chemoradiation and surgical resection in locally advanced rectal cancer with high risk of recurrence - a phase II study. Anticancer Res, 2017, 37(5):2683–91., DOI 10.21873/anticanres.11617 Liu GC, Yan JP, He Q, An X, Pan ZZ, Ding PR. Effect of neoadjuvant chemoradiotherapy with capecitabine versus fluorouracil for locally advanced rectal cancer: A meta-analysis. Gastroenterol Res Pract, 2016, 2016:1798285., DOI 10.1155/ 2016/1798285 Chakrabarti D, Rajan S, Akhtar N, Qayoom S, Gupta S, VermaM, et al. Shortcourse radiotherapy with consolidation chemotherapy versus conventionally fractionated long-course chemoradiotherapy for locally advanced rectal cancer: Randomized clinical trial. Br J Surg, 2021, 108:511–20., DOI 10.1093/bjs/znab020 Kav T, Bayraktar Y. How useful is rectal endosonography in the staging of rectal cancer? World J Gastroenterol, 2010, 16:691–7., DOI 10.3748/wjg.v16.i6.691 Ang CW, Dawson R, Hall C, Farmer M. The diagnostic value of digital rectal examination in primary care for palpable rectal tumour. Colorectal Dis, 2008, 10: 789–92., DOI 10.1111/j.1463-1318.2007.01381.x Rastogi R, Meena GL, Gupta Y, Sinha P, Kumar Das P, Chaudhary M, et al. CT or MRI – which is better for rectal cancer imaging? Colorec Cancer, 2016, 2:3., DOI 10.21767/2471-9943.100028 Glynne-Jones R, Wyrwicz L, Tiret E, Brown G, Rodel C, Cervantes A, et al. Rectal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol, 2018, 29:iv263., DOI 10.1093/annonc/mdy161 Gilshtein H, Ghuman A, Dawoud M, Yellinek S, Kent I, Sharp SP, et al. Total neoadjuvant treatment for rectal cancer: Preliminary experience. Am Surg, 2021, 87:708–13., DOI 10.1177/0003134820951499 Conroy T, Bosset JF, Etienne PL, Rio E, E F, Mesgouez-Nebout N, et al. Neoadjuvant chemotherapy with FOLFIRINOX and preoperative chemoradiotherapy for patients with locally advanced rectal cancer, UNICANCER-PRODIGE 23): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol, 2021, 22:702–15., DOI 10.1016/S1470-2045(21)00079-6 US Department of Health and Human Services. Common Terminology criteria for adverse events, CTCAE). Bethesda, MD: U.S.DEPARTMENT OF HEALTH AND HUMAN SERVICES National Institutes of Health, 2010). Version 4.0 Published: May 28, 2009, v4.03: June 14, 2010). Nelson H, Sargent DJ, Wieand HS, Fleshman J, Anvari M, Stryker SJ, et al. A comparison of laparoscopically assisted and open colectomy for colon cancer. N Engl J Med, 2004, 350:2050–9., DOI 10.1056/NEJMoa032651 Guillou PJ, Quirke P, Thorpe H, Walker J, Jayne DG, Smith AM, et al. Shortterm endpoints of conventional versus laparoscopic-assisted surgery in patients with colorectal cancer, MRC CLASICC trial): Multicentre, randomised controlled trial. Lancet, 2005, 365:1718–26., DOI 10.1016/S0140-6736(05)66545-2 Lacy AM, Garcia-Valdecasas JC, Delgado S, Castells A, Taura P, Pique JM, et al. Laparoscopy-assisted colectomy versus open colectomy for treatment of nonmetastatic colon cancer: A randomised trial. Lancet, 2002, 359:2224–9., DOI 10. 1016/S0140-6736(02)09290-5 Bonjer HJ, Hop WC, Nelson H, Sargent DJ, Lacy AM, Castells A, et al. Laparoscopically assisted vs open colectomy for colon cancer: Ameta-analysis. Arch Surg, 2007, 142:298–303., DOI 10.1001/archsurg.142.3.298 Jayne DG, Thorpe HC, Copeland J, Quirke P, Brown JM, Guillou PJ. Five-year follow-up of the Medical Research Council CLASICC trial of laparoscopically assisted versus open surgery for colorectal cancer. Br J Surg, 2010, 97:1638–45., DOI 10.1002/bjs.7160 Veenhof AA, Sietses C, von Blomberg BM, van Hoogstraten IM, vd Pas MH, Meijerink WJ, et al. The surgical stress response and postoperative immune function after laparoscopic or conventional total mesorectal excision in rectal cancer: A randomized trial. Int J Colorectal Dis, 2011, 26:53–9., DOI 10.1007/ s00384-010-1056-9 Mandard AM, Dalibard F, Mandard JC, Marnay J, Henry-Amar M, Petiot JF, et al. Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Cancer, 1994, 73:2680–6., DOI 10.1002/1097-0142(19940601)73:11<2680::aidcncr2820731105> 3.0.co;2-c Jarrar A, Edalatpour A, Sebikali-Potts A, Vitello D, Valente M, Liska D, et al. An up-to-date predictive model for rectal cancer survivorship reflecting tumor biology and clinical factors. Am J Surg, 2020, 219:515–20., DOI 10.1016/j.amjsurg. 2019.10.036 Zou XC, Wang QW, Zhang JM. Comparison of 5-FU-based and capecitabinebased neoadjuvant chemoradiotherapy in patients with rectal cancer: A metaanalysis. Clin Colorectal Cancer, 2017, 16:e123–39., DOI 10.1016/j.clcc.2017.01.009 Lee SW, Jeong SY, Kim K, Kim SJ. Direct comparison of F-18 FDG PET/CT and MRI to predict pathologic response to neoadjuvant treatment in locally advanced rectal cancer: A meta-analysis. Ann Nucl Med, 2021, 35:1038–47., DOI 10.1007/s12149-021-01639-y Jhaveri KS, Hosseini-Nik H. MRI of rectal cancer: An overview and update on recent advances. AJR Am J Roentgenol, 2015, 205:W42–55., DOI 10.2214/AJR.14. 14201 Krishnamurthi SS, Seo Y, Kinsella TJ. Adjuvant therapy for rectal cancer. Clin Colon Rectal Surg, 2007, 20:167–81., DOI 10.1055/s-2007-984861 Husseini ZE, Haibe Y, Bouferraa Y, Kreidieh M, Darazi MA, Mukherji D, et al. Total neoadjuvant therapy in patients with locally advanced rectal cancer: A tertiary medical center experience. Mol Clin Oncol, 2021, 15:220., DOI 10.3892/mco.2021. 2382 Cercek A, Roxburgh CSD, Strombom P, Smith JJ, Temple LKF, Nash GM, et al. Adoption of total neoadjuvant therapy for locally advanced rectal cancer. JAMA Oncol, 2018, 4(6):e180071., DOI 10.1001/jamaoncol.2018.0071 Voogt ELK, Schaap DP, van den Berg K, Nieuwenhuijzen GAP, Bloemen JG, Creemers GJ, et al. Improved response rate in patients with prognostically poor locally advanced rectal cancer after treatment with induction chemotherapy and chemoradiotherapy when compared with chemoradiotherapy alone: A matched case-control study. Eur J Surg Oncol, 2021, 47:2429–35., DOI 10.1016/j.ejso.2021. 05.017 De Caluwe L, Van Nieuwenhove Y, Ceelen WP. Preoperative chemoradiation versus radiation alone for stage II and III resectable rectal cancer. Cochrane Database Syst Rev, 2013, 2:CD006041., DOI 10.1002/14651858.CD006041.pub3 Rombouts AJM, Hugen N, Verhoeven RHA, Elferink MAG, Poortmans PMP, Nagtegaal ID, et al. Tumor response after long interval comparing 5x5Gy radiation therapy with chemoradiation therapy in rectal cancer patients. Eur J Surg Oncol, 2018, 44:1018–24., DOI 10.1016/j.ejso.2018.03.017 Musters GD, Buskens CJ, Bemelman WA, Tanis PJ. Perineal wound healing after abdominoperineal resection for rectal cancer: A systematic review and metaanalysis. Dis Colon Rectum, 2014, 57:1129–39., DOI 10.1097/DCR. 0000000000000182 Haubner F, Ohmann E, Pohl F, Strutz J, Gassner HG. Wound healing after radiation therapy: Review of the literature. Radiat Oncol, 2012, 7:162., DOI 10.1186/ 1748-717X-7-162 Pettersson D, Cedermark B, Holm T, Radu C, Pahlman L, Glimelius B, et al. Interim analysis of the Stockholm III trial of preoperative radiotherapy regimens for rectal cancer. Br J Surg, 2010, 97:580–7., DOI 10.1002/bjs.6914 Pettersson D, Glimelius B, Iversen H, Johansson H, Holm T, Martling A. Impaired postoperative leucocyte counts after preoperative radiotherapy for rectal cancer in the Stockholm III Trial. Br J Surg, 2013, 100:969–75., DOI 10.1002/bjs.9117 Ozgen Z, Ozden S, Atasoy BM, Ozyurt H, Gencosmanoglu R, Imeryuz N. Long-term effects of neoadjuvant chemoradiotherapy followed by sphincterpreserving resection on anal sphincter function in relation to quality of life among locally advanced rectal cancer patients: A cross-sectional analysis. Radiat Oncol, 2015, 10:168., DOI 10.1186/s13014-015-0479-4 Kornmann M, Staib L, Wiegel T, Kreuser ED, Kron M, Baumann W, et al. Adjuvant chemoradiotherapy of advanced resectable rectal cancer: Results of a randomised trial comparing modulation of 5-fluorouracil with folinic acid or with interferon-α. Br J Cancer, 2010, 103:1163–72., DOI 10.1038/sj.bjc.6605871 Moertel CG, Fleming TR, Macdonald JS, Haller DG, Laurie JA, Goodman PJ, et al. Levamisole and fluorouracil for adjuvant therapy of resected colon carcinoma. N Engl J Med, 1990, 322:352–8., DOI 10.1056/NEJM199002083220602 Thirion P, Michiels S, Pignon JP, Buyse M, Braud AC, Carlson RW, et al. Modulation of fluorouracil by leucovorin in patients with advanced colorectal cancer: An updated meta-analysis. J Clin Oncol, 2004, 22:3766–75., DOI 10.1200/ JCO.2004.03.104 Hofheinz RD, Wenz F, Post S, Matzdorff A, Laechelt S, Hartmann JT, et al. Chemoradiotherapy with capecitabine versus fluorouracil for locally advanced rectal cancer: A randomised, multicentre, non-inferiority, phase 3 trial. Lancet Oncol, 2012, 13:579–88., DOI 10.1016/S1470-2045(12)70116-X Han J, Noh GT, Yeo SA, Cheong C, Cho MS, Hur H, et al. The number of retrieved lymph nodes needed for accurate staging differs based on the presence of preoperative chemoradiation for rectal cancer. Medicine, Baltimore,, 2016, 95: e4891., DOI 10.1097/MD.0000000000004891 Scheidbach H, Schneider C, Konradt J, Barlehner E, Kohler L, Wittekind C, et al. Laparoscopic abdominoperineal resection and anterior resection with curative intent for carcinoma of the rectum. Surg Endosc, 2002, 16:7–13., DOI 10.1007/ s00464-001-8314-4 Van Cutsem E, Hoff PM, HarPer P, Bukowski RM, Cunningham D, Dufour P, et al. Oral capecitabine vs intravenous 5-fluorouracil and leucovorin: Integrated efficacy data and novel analyses from two large, randomised, phase III trials. Br J Cancer, 2004, 90:1190–7., DOI 10.1038/sj.bjc.6601676 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610722 EP 1610732 DI 10.3389/pore.2022.1610722 PG 11 ER PT J AU Yang, L Fan, Y Lu, H AF Yang, Lan Fan, Ying Lu, Hongyang TI Pulmonary Large Cell Neuroendocrine Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE treatment; diagnosis; LCNEC; imaging examination; pathologic features; prognosis ID treatment; diagnosis; LCNEC; imaging examination; pathologic features; prognosis AB Pulmonary large cell neuroendocrine carcinoma (LCNEC) is a rare subtype of malignant pulmonary tumor. The incidence rate of LCNEC was reported to be 0.3%–3% in lung cancers. Although LCNEC is classified as non-small cell lung cancer (NSCLC), it is more aggressive and malignant than other NSCLC, and its biological behavior is similar to that of small cell lung cancer (SCLC). Most of the LCNEC patients are elderly smoking male and the clinical manifestations are not specific. The imaging manifestations of the tumors are often located in the periphery and the upper lobes, and the enlargement of mediastinal or hilar lymph nodes is common. The diagnosis is mainly based on pathology by the histological features and immunohistochemistry (IHC). Specific neuroendocrine markers such as chromogranin A (CgA), synaptophysin (Syn) and CD56 are usually diffusely positive in LCNEC, and found that insulinoma-associated protein (INSM1) and high rate of Ki-67 are helpful for diagnosis. More differential diagnoses also increase the difficulty of correctly diagnosing LCNEC. The rise of LCNEC molecular typing in recent years may be helpful for diagnosis and subsequent treatment. This review focuses on the epidemiological features, imaging studies, pathology, diagnosis, treatment, and prognosis of LCNEC. C1 [Yang, Lan] Zhejiang Cancer Hospital, Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus)Hangzhou, China. [Fan, Ying] Zhejiang Cancer Hospital, Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus)Hangzhou, China. [Lu, Hongyang] Zhejiang Cancer Hospital, Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus)Hangzhou, China. RP Lu, H (reprint author), Zhejiang Cancer Hospital, Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Hangzhou, China. EM luhy@zjcc.org.cn CR Asamura H, Kameya T, Matsuno Y, Noguchi M, Tada H, Ishikawa Y, et al. Neuroendocrine Neoplasms of the Lung: a Prognostic Spectrum. J Clin Oncol, 2006, 24(1):70–6., DOI 10.1200/JCO.2005.04.1202 Travis WD. Advances in Neuroendocrine Lung Tumors. Ann Oncol, 2010, 21(7):vii65–vii71., DOI 10.1093/annonc/mdq380 Travis WD, Linnoila RI, Tsokos MG, Hitchcock CL, Cutler GB, Nieman L, et al. Neuroendocrine Tumors of the Lung with Proposed Criteria for Large- Cell Neuroendocrine Carcinoma. An Ultrastructural, Immunohistochemical, and Flow Cytometric Study of 35 Cases. Am J Surg Pathol, 1991, 15(6): 529–53., DOI 10.1097/00000478-199106000-00003 Arrigoni MG, Woolner LB, Bernatz PE. Atypical Carcinoid Tumors of the Lung. J Thorac Cardiovasc Surg, 1972, 64(3):413–21., DOI 10.1016/s0022- 5223(19)39836-8 Varlotto JM, Medford-Davis LN, Recht A, Flickinger JC, Schaefer E, Zander DS, et al. Hould Large Cell Neuroendocrine Lung Carcinoma Be Classified and Treated as a Small Cell Lung Cancer or with Other Large Cell Carcinomas? J Thorac Oncol, 2011, 6(6):1050–8., DOI 10.1097/JTO. 0b013e318217b6f8 Travis WD, Brambilla E, Burke AP, Marx A, Nicholson AG. Introduction to the 2015 World Health Organization Classification of Tumors of the Lung, Pleura, Thymus, and Heart. J Thorac Oncol, 2015, 10(9):1240–2., DOI 10.1097/ JTO.0000000000000663 Nicholson AG, Tsao MS, Beasley MB, Borczuk AC, Brambilla E, Cooper WA, et al. The 2021 WHO Classification of Lung Tumors: Impact of Advances since 2015. J Thorac Oncol, 2022, 17(3):362–87., DOI 10.1016/j.jtho.2021. 11.003 Kupeli M, Koseoglu RD. Large Cell Carcinoma with Adenocarcinoma in Lung. J Coll Physicians Surg Pak, 2018, 28(3):240–2., DOI 10.29271/jcpsp. 2018.03.240 Pelosi G, Fabbri A, Cossa M, Sonzogni A, Valeri B, Righi L, et al. What Clinicians Are Asking Pathologists when Dealing with Lung Neuroendocrine Neoplasms? Semin Diagn Pathol, 2015, 32(6):469–79., DOI 10.1053/j.semdp. 2015.10.009 Welter S, Aigner C, Roesel C. The Role of Surgery in High Grade Neuroendocrine Tumours of the Lung. J Thorac Dis, 2017, 9(15): S1474–S1483., DOI 10.21037/jtd.2017.01.60 Fasano M, Della Corte CM, Papaccio F, Ciardiello F, Morgillo F. Pulmonary Large-Cell Neuroendocrine Carcinoma: From Epidemiology to Therapy. J Thorac Oncol, 2015, 10(8):1133–41., DOI 10.1097/JTO.0000000000000589 Fernandez FG, Battafarano RJ. Large-cell Neuroendocrine Carcinoma of the Lung: an Aggressive Neuroendocrine Lung Cancer. Semin Thorac Cardiovasc Surg, 2006, 18(3):206–10., DOI 10.1053/j.semtcvs.2006.08.007 Veronesi G, Morandi U, Alloisio M, Terzi A, Cardillo G, Filosso P, et al. Large Cell Neuroendocrine Carcinoma of the Lung: a Retrospective Analysis of 144 Surgical Cases. Lung Cancer, 2006, 53(1):111–5., DOI 10.1016/j.lungcan. 2006.03.007 Paci M, Cavazza A, Annessi V, Putrino I, Ferrari G, De Franco S, et al. Large Cell Neuroendocrine Carcinoma of the Lung: a 10-year Clinicopathologic Retrospective Study. Ann Thorac Surg, 2004, 77(4):1163–7., DOI 10.1016/j. athoracsur.2003.09.070 Zacharias J, Nicholson AG, Ladas GP, Goldstraw P. Large Cell Neuroendocrine Carcinoma and Large Cell Carcinomas with Neuroendocrine Morphology of the Lung: Prognosis after Complete Resection and Systematic Nodal Dissection. Ann Thorac Surg, 2003, 75(2):348–52., DOI 10.1016/s0003-4975(02)04118-8 Naidoo J, Santos-Zabala ML, Iyriboz T, Woo KM, Sima CS, Fiore JJ, et al. Large Cell Neuroendocrine Carcinoma of the Lung: Clinico-Pathologic Features, Treatment, and Outcomes. Clin Lung Cancer, 2016, 17(5): e121–9., DOI 10.1016/j.cllc.2016.01.003 Isaka M, Nakagawa K, Ohde Y, Okumura T, Watanabe R, Ito I, et al. A Clinicopathological Study of Peripheral, Small-Sized High-Grade Neuroendocrine Tumours of the Lung: Differences between Small-Cell Lung Carcinoma and Large-Cell Neuroendocrine Carcinoma. Eur J Cardiothorac Surg, 2012, 41(4):841–6., DOI 10.1093/ejcts/ezr132 Iyoda A, Hiroshima K, Toyozaki T, Fujisawa T, Ohwada H. Clinical Characterization of Pulmonary Large Cell Neuroendocrine Carcinoma and Large Cell Carcinoma with Neuroendocrine Morphology. Cancer, 2001, 91(11):1992–2000., DOI 10.1002/1097-0142(20010601)91:11<1992:: aid-cncr1224>3.0.co;2-5 Sarkaria IS, Iyoda A, RohMS, Sica G, Kuk D, Sima CS, et al. Neoadjuvant and Adjuvant Chemotherapy in Resected Pulmonary Large Cell Neuroendocrine Carcinomas: a Single Institution Experience. Ann Thorac Surg, 2011, 92(4): 1180–6., DOI 10.1016/j.athoracsur.2011.05.027 Sun JM, Ahn MJ, Ahn JS, Um SW, Kim H, Kim HK, et al. Chemotherapy for Pulmonary Large Cell Neuroendocrine Carcinoma: Similar to that for Small Cell Lung Cancer or Non-small Cell Lung Cancer? Lung Cancer, 2012, 77(2): 365–70., DOI 10.1016/j.lungcan.2012.04.009 Rossi G, Cavazza A, Marchioni A, Longo L, MigaldiM, Sartori G, et al. Role of Chemotherapy and the Receptor Tyrosine Kinases KIT, PDGFRalpha, PDGFRbeta, and Met in Large-Cell Neuroendocrine Carcinoma of the Lung. J Clin Oncol, 2005, 23(34):8774–85., DOI 10.1200/JCO.2005.02.8233 Kim KW, Kim HK, Kim J, Shim YM, Ahn MJ, Choi YL. Outcomes of Curative-Intent Surgery and Adjuvant Treatment for Pulmonary Large Cell Neuroendocrine Carcinoma. World J Surg, 2017, 41(7):1820–7., DOI 10.1007/ s00268-017-3908-8 Fisseler-Eckhoff A, Demes M. Neuroendocrine Tumors of the Lung. Cancers, Basel,, 2012, 4(3):777–98., DOI 10.3390/cancers4030777 Selvaggi G, Scagliotti GV. Histologic Subtype in NSCLC: Does it Matter? Oncology, Williston Park,, 2009, 23(13):1133–40. Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, et al. The 2015 World Health Organization Classification of Lung Tumors: Impact of Genetic, Clinical and Radiologic Advances since the 2004 Classification. J Thorac Oncol, 2015, 10(9):1243–60., DOI 10.1097/ JTO.0000000000000630 Rindi G, Klersy C, Inzani F, FelleGara G, AmpoLLini L, Ardizzoni A, et al. Grading the Neuroendocrine Tumors of the Lung: an Evidence-Based Proposal. Endocr Relat Cancer, 2013, 21(1):1–16., DOI 10.1530/ERC-13-0246 Garcia-Yuste M, Matilla JM, Alvarez-Gago T, Duque JL, Heras F, Cerezal LJ, et al. Prognostic Factors in Neuroendocrine Lung Tumors: a Spanish Multicenter Study. Spanish Multicenter Study of Neuroendocrine Tumors of the Lung of the Spanish Society of Pneumonology and Thoracic Surgery, EMETNE-SEPAR). Ann Thorac Surg, 2000, 70(1):258–63., DOI 10.1016/ s0003-4975(00)01369-2 Jung KJ, Lee KS, Han J, Kwon OJ, Kim J, Shim YM, et al. Large Cell Neuroendocrine Carcinoma of the Lung: Clinical, CT, and Pathologic Findings in 11 Patients. J Thorac Imaging, 2001, 16(3):156–62., DOI 10. 1097/00005382-200107000-00003 Oshiro Y, Kusumoto M, Matsuno Y, Asamura H, Tsuchiya R, Terasaki H, et al. CT Findings of Surgically Resected Large Cell Neuroendocrine Carcinoma of the Lung in 38 Patients. AJR Am J Roentgenol, 2004, 182(1):87–91., DOI 10.2214/ajr.182.1.1820087 Lee KW, Lee Y, Oh SW, Jin KN, Goo JM. Large Cell Neuroendocrine Carcinoma of the Lung: CT and FDG PET Findings. Eur J Radiol, 2015, 84(11):2332–8., DOI 10.1016/j.ejrad.2015.07.033 Akata S, Okada S,Maeda J, Park J, YoshimuraM, Saito K, et al. Computed Tomographic Findings of Large Cell Neuroendocrine Carcinoma of the Lung. Clin Imaging, 2007, 31(6):379–84., DOI 10.1016/j.clinimag.2007. 04.027 Chong S, Lee KS, Kim BT, Choi JY, Yi CA, Chung MJ, et al. Integrated PET/ CT of Pulmonary Neuroendocrine Tumors: Diagnostic and Prognostic Implications. AJR Am J Roentgenol, 2007, 188(5):1223–31., DOI 10.2214/ AJR.06.0503 Takamochi K, Yokose T, Yoshida J, Nishimura M, Ohmatsu H, Nagai K, et al. Calcification in Large Cell Neuroendocrine Carcinoma of the Lung. Jpn J Clin Oncol, 2003, 33(1):10–3., DOI 10.1093/jjco/hyg006 Eichhorn F, Dienemann H, Muley T, Warth A, Hoffmann H. Predictors of Survival after Operation Among Patients with Large Cell Neuroendocrine Carcinoma of the Lung. Ann Thorac Surg, 2015, 99(3):983–9., DOI 10.1016/j. athoracsur.2014.10.015 Travis WD. Pathology and Diagnosis of Neuroendocrine Tumors: Lung Neuroendocrine. Thorac Surg Clin, 2014, 24(3):257–66., DOI 10.1016/j. thorsurg.2014.04.001 Rossi G, Longo L, Barbieri F, Bertolini F, Spagnolo P. Large Cell Neuroendocrine Carcinoma of the Lung: Chemotherapy Regimen Depends on How "large" Your Diagnostic Criteria Are. Eur Respir J, 2017, 50(4):1701292., DOI 10.1183/13993003.01292-2017 den Bakker MA, Willemsen S, Grunberg K, Noorduijn LA, van Oosterhout MFM, van Suylen RJ, et al. Small Cell Carcinoma of the Lung and Large Cell Neuroendocrine Carcinoma Interobserver Variability. Histopathology, 2010, 56(3):356–63., DOI 10.1111/j.1365-2559.2010.03486.x Bohnenberger H, Dinter H, Konig A, Strobel P. Neuroendocrine Tumors of the Thymus and Mediastinum. J Thorac Dis, 2017, 9(15):S1448–S1457., DOI 10.21037/jtd.2017.02.02 Peifer M, Fernandez-Cuesta L, Sos ML, George J, Seidel D, Kasper LH, et al. Integrative Genome Analyses Identify Key Somatic Driver Mutations of Small-Cell Lung Cancer. Nat Genet, 2012, 44(10):1104–10., DOI 10.1038/ng. 2396 Liang R, Chen TX, Wang ZQ, Jin KW, Zhang LY, Yan QN, et al. A Retrospective Analysis of the Clinicopathological Characteristics of Large Cell Carcinoma of the Lung. Exp Ther Med, 2015, 9(1):197–202., DOI 10.3892/ etm.2014.2075 Takei H, Asamura H, Maeshima A, Suzuki K, Kondo H, Niki T, et al. Large Cell Neuroendocrine Carcinoma of the Lung: a Clinicopathologic Study of Eighty-Seven Cases. J Thorac Cardiovasc Surg, 2002, 124(2):285–92., DOI 10. 1067/mtc.2002.122523 Meng Y, Lian YB, Xu Y, Dong JQ, Song M. Clinical and Molecular Pathological Features of Bronchopulmonary Large Cell Neuroendocrine Carcinoma. Zhonghua Yi Xue Za Zhi, 2022, 102(14):1020–7., DOI 10.3760/ cma.j.cn112137-20210814-01816 Incekara F, Aydogdu K, Sayilir E, Gulhan SSE, Demirag F, Kaya S, et al. Surgical Management of Large-Cell Neuroendocrinelung Carcinoma: an Analysis of 25 Cases. Turk J Med Sci, 2016, 46(6):1808–15., DOI 10.3906/ sag-1507-115 Baine MK, Sinard JH, Cai G, Homer RJ. A Semiquantitative Scoring System May Allow Biopsy Diagnosis of Pulmonary Large Cell Neuroendocrine Carcinoma. Am J Clin Pathol, 2020, 153(2):165–74., DOI 10.1093/ajcp/aqz149 Lyda MH, Weiss LM. Immunoreactivity for Epithelial and Neuroendocrine Antibodies Are Useful in the Differential Diagnosis of Lung Carcinomas. Hum Pathol, 2000, 31(8):980–7., DOI 10.1053/hupa.2000.9076 Sturm N, Lantuejoul S, Laverriere MH, Papotti M, Brichon PY, Brambilla C, et al. Thyroid Transcription Factor 1 and Cytokeratins 1, 5, 10, 14, 34betaE12, Expression in Basaloid and Large-Cell Neuroendocrine Carcinomas of the Lung. Hum Pathol, 2001, 32(9):918–25., DOI 10.1053/ hupa.2001.27110 Zou Q, Zhang L, Cheng Z, Guo X, Cao D. INSM1 Is Less Sensitive but More Specific Than Synaptophysin in Gynecologic High-Grade Neuroendocrine Carcinomas: An Immunohistochemical Study of 75 Cases with Specificity Test and Literature Review. Am J Surg Pathol, 2021, 45(2):147–59., DOI 10. 1097/PAS.0000000000001641 Sakakibara R, Kobayashi M, Takahashi N, Inamura K, Ninomiya H, Wakejima R, et al. Insulinoma-associated Protein 1, INSM1, Is a Better Marker for the Diagnosis and Prognosis Estimation of Small Cell Lung Carcinoma Than Neuroendocrine Phenotype Markers Such as Chromogranin A, Synaptophysin, and CD56. Am J Surg Pathol, 2020, 44(6):757–64., DOI 10.1097/PAS.0000000000001444 Rooper LM, Bishop JA, Westra WH. INSM1 Is a Sensitive and Specific Marker of Neuroendocrine Differentiation in Head and Neck Tumors. Am J Surg Pathol, 2018, 42(5):665–71., DOI 10.1097/PAS.0000000000001037 Dermawan JK, Mukhopadhyay S. Insulinoma-associated Protein 1, INSM1, Differentiates Carcinoid Tumourlets of the Lung from Pulmonary Meningothelial-like Nodules. Histopathology, 2018, 72(6):1067–9., DOI 10. 1111/his.13458 Rekhtman N, Pietanza MC, Hellmann MD, Naidoo J, Arora A, Won H, et al. Next-Generation Sequencing of Pulmonary Large Cell Neuroendocrine Carcinoma Reveals Small Cell Carcinoma-like and Non-small Cell Carcinoma-like Subsets. Clin Cancer Res, 2016, 22(14):3618–29., DOI 10. 1158/1078-0432.CCR-15-2946 Bari MF, Brown H, Nicholson AG, Kerr KM, Gosney JR, Wallace WA, et al. Bai3, CDX2 and VIL1: a Panel of Three Antibodies to Distinguish Small Cell from Large Cell Neuroendocrine Lung Carcinomas. Histopathology, 2014, 64(4):547–56., DOI 10.1111/his.12278 Saunders LR, Bankovich AJ, Anderson WC, Aujay MA, Bheddah S, Black K, et al. A DLL3-Targeted Antibody-Drug Conjugate Eradicates Highgrade Pulmonary Neuroendocrine Tumor-Initiating Cells In Vivo. Sci Transl Med, 2015, 7(302):302ra136., DOI 10.1126/scitranslmed.aac9459 Ogawa H, Sakai Y, Nishio W, Fujibayashi Y, Nishikubo M, Nishioka Y, et al. DLL3 Expression Is a Predictive Marker of Sensitivity to Adjuvant Chemotherapy for Pulmonary LCNEC. Thorac Cancer, 2020, 11(9): 2561–9., DOI 10.1111/1759-7714.13574 Ye B, Cappel J, Findeis-Hosey J, McMahon L, Yang Q, Xiao GQ, et al. hASH1 Is a Specific ImmunohistochemicalMarker for Lung Neuroendocrine Tumors. Hum Pathol, 2016, 48:142–7., DOI 10.1016/j.humpath.2015.09.019 Papaxoinis G, Bille A, McLean E, Nonaka D. Comparative Study of Rb1, Cyclin D1 and P16 Immunohistochemistry Expression to Distinguish Lung Small-Cell Carcinoma and Large-Cell Neuroendocrine Carcinoma. Histopathology, 2022, 81(2):205–14., DOI 10.1111/his.14679 Beasley MB, Lantuejoul S, Abbondanzo S, ChuWS, Hasleton PS, Travis WD, et al. The P16/cyclin D1/Rb Pathway in Neuroendocrine Tumors of the Lung. Hum Pathol, 2003, 34(2):136–42., DOI 10.1053/hupa.2003.8 Tsai H, Morais CL, Alshalalfa M, Tan HL, Haddad Z, Hicks J, et al. Cyclin D1 Loss Distinguishes Prostatic Small-Cell Carcinoma from Most Prostatic Adenocarcinomas. Clin Cancer Res, 2015, 21(24):5619–29., DOI 10.1158/ 1078-0432.CCR-15-0744 Pelosi G, Rindi G, Travis WD, Papotti M. Ki-67 Antigen in Lung Neuroendocrine Tumors: Unraveling a Role in Clinical Practice. J Thorac Oncol, 2014, 9(3):273–84., DOI 10.1097/JTO.0000000000000092 Righi L, Volante M, Rapa I, Tavaglione V, Inzani F, Pelosi G, et al. Mammalian Target of Rapamycin Signaling Activation Patterns in Neuroendocrine Tumors of the Lung. Endocr Relat Cancer, 2010, 17(4): 977–87., DOI 10.1677/ERC-10-0157 Silveira F, Basile ML, Kuga FS, Prospero JD, Paes RAP, Bernardi FDC. Neuroendocrine Tumors: An Epidemiological Study of 250 Cases at a Tertiary Hospital. Rev Assoc Med Bras, 2017, 63(10):856–61., DOI 10.1590/ 1806-9282.63.10.856 Iyoda A, Hiroshima K, Nakatani Y, Fujisawa T. Pulmonary Large Cell Neuroendocrine Carcinoma: its Place in the Spectrum of Pulmonary Carcinoma. Ann Thorac Surg, 2007, 84(2):702–7., DOI 10.1016/j.athoracsur. 2007.03.093 Iyoda A, Makino T, Koezuka S, Otsuka H, Hata Y. Treatment Options for Patients with Large Cell Neuroendocrine Carcinoma of the Lung. Gen Thorac Cardiovasc Surg, 2014, 62(6):351–6., DOI 10.1007/s11748-014- 0379-9 Derks JL, Leblay N, Thunnissen E, van Suylen RJ, den Bakker M, Groen HJM, et al. Molecular Subtypes of Pulmonary Large-Cell Neuroendocrine Carcinoma Predict Chemotherapy Treatment Outcome. Clin Cancer Res, 2018, 24(1):33–42., DOI 10.1158/1078-0432.CCR-17-1921 Zhuo M, Guan Y, Yang X, Hong L, Wang Y, Li Z, et al. The Prognostic and Therapeutic Role of Genomic Subtyping by Sequencing Tumor or Cell-free DNA in Pulmonary Large-Cell Neuroendocrine Carcinoma. Clin Cancer Res, 2020, 26(4):892–901., DOI 10.1158/1078-0432.CCR-19-0556 Le X, Desai NV, Majid A, Karp RS, Huberman MS, Rangachari D, et al. De Novo pulmonary Small Cell Carcinomas and Large Cell Neuroendocrine Carcinomas Harboring EGFR Mutations: Lack of Response to EGFR Inhibitors. Lung Cancer, 2015, 88(1):70–3., DOI 10.1016/j.lungcan.2015. 02.003 Iyoda A, Travis WD, Sarkaria IS, Jiang SX, Amano H, Sato Y, et al. Expression Profiling and Identification of Potential Molecular Targets for Therapy in Pulmonary Large-Cell Neuroendocrine Carcinoma. Exp Ther Med, 2011, 2(6):1041–5., DOI 10.3892/etm.2011.343 George J, Walter V, Peifer M, Alexandrov LB, Seidel D, Leenders F, et al. Integrative Genomic Profiling of Large-Cell Neuroendocrine Carcinomas Reveals Distinct Subtypes of High-Grade Neuroendocrine Lung Tumors. Nat Commun, 2018, 9(1):1048., DOI 10.1038/s41467-018-03099-x Hung YP. Neuroendocrine Tumors of the Lung: Updates and Diagnostic Pitfalls. Surg Pathol Clin, 2019, 12(4):1055–71., DOI 10.1016/j.path.2019. 08.012 Baine MK, Rekhtman N. Multiple Faces of Pulmonary Large Cell Neuroendocrine Carcinoma: Update with a Focus on Practical Approach to Diagnosis. Transl Lung Cancer Res, 2020, 9(3):860–78., DOI 10.21037/tlcr. 2020.02.13 Yoshimura M, Seki K, Bychkov A, Fukuoka J. Molecular Pathology of Pulmonary Large Cell Neuroendocrine Carcinoma: Novel Concepts and Treatments. Front Oncol, 2021, 11:671799., DOI 10.3389/fonc.2021.671799 Yoshida A, Kobayashi E, Kubo T, Kodaira M, Motoi T, Motoi N, et al. Clinicopathological and Molecular Characterization of SMARCA4-Deficient Thoracic Sarcomas with Comparison to Potentially Related Entities. Mod Pathol, 2017, 30:797–809., DOI 10.1038/modpathol.2017.11 Rekhtman N, Montecalvo J, Chang JC, Alex D, Ptashkin RN, Ai N, et al. SMARCA4-Deficient Thoracic Sarcomatoid Tumors Represent Primarily Smoking-Related Undifferentiated Carcinomas rather Than Primary Thoracic Sarcomas. J Thorac Oncol, 2020, 15(2):231–47., DOI 10.1016/j. jtho.2019.10.023 Yeh YC, Kao HL, Lee KL, Wu MH, Ho HL, Chou TY. Epstein-Barr Virus- Associated Pulmonary Carcinoma: Proposing an Alternative Term and Expanding the Histologic Spectrum of Lymphoepithelioma-like Carcinoma of the Lung. Am J Surg Pathol, 2019, 43(2):211–9., DOI 10. 1097/PAS.0000000000001173 Fournel L, Falcoz PE, AlifanoM, Charpentier MC, Boudaya MS,Magdeleinat P, et al. Surgical Management of Pulmonary Large Cell Neuroendocrine Carcinomas: a 10-year Experience. Eur J Cardiothorac Surg, 2013, 43(1): 111–4., DOI 10.1093/ejcts/ezs174 Iyoda A, Hiroshima K, Moriya Y, Iwadate Y, Takiguchi Y, Uno T, et al. Postoperative Recurrence and the Role of Adjuvant Chemotherapy in Patients with Pulmonary Large-Cell Neuroendocrine Carcinoma. J Thorac Cardiovasc Surg, 2009, 138(2):446–53., DOI 10.1016/j.jtcvs.2008.12.037 Kumar P, Herndon J, 2nd, Langer M, Kohman LJ, Elias AD, Kass FC, et al. Patterns of Disease Failure after Trimodality Therapy of Nonsmall Cell Lung Carcinoma Pathologic Stage IIIA, N2). Analysis of Cancer and Leukemia Group B Protocol 8935. Cancer, 1996, 77(11):2393–9., DOI 10.1002/(SICI, 1097-0142(19960601)77:11<2393::AID-CNCR31>3.0.CO;2-Q Fujiwara Y, Sekine I, Tsuta K, Ohe Y, Kunitoh H, Yamamoto N, et al. Effect of Platinum Combined with Irinotecan or Paclitaxel against Large Cell Neuroendocrine Carcinoma of the Lung. Jpn J Clin Oncol, 2007, 37(7): 482–6., DOI 10.1093/jjco/hym053 Yoshida H, Sekine I, Tsuta K, Horinouchi H, Nokihara H, Yamamoto N, et al. Amrubicin Monotherapy for Patients with Previously Treated Advanced Large-Cell Neuroendocrine Carcinoma of the Lung. Jpn J Clin Oncol, 2011, 41(7):897–901., DOI 10.1093/jjco/hyr065 Tokito T, Kenmotsu H, Watanabe R, Ito I, Shukuya T, Ono A, et al. Comparison of Chemotherapeutic Efficacy between LCNEC Diagnosed Using Large Specimens and Possible LCNEC Diagnosed Using Small Biopsy Specimens. Int J Clin Oncol, 2014, 19(1):63–7., DOI 10.1007/s10147- 012-0509-2 Niho S, Kenmotsu H, Sekine I, Ishii G, Ishikawa Y, Noguchi M, et al. Combination Chemotherapy with Irinotecan and Cisplatin for Large- Cell Neuroendocrine Carcinoma of the Lung: a Multicenter Phase II Study. J Thorac Oncol, 2013, 8(7):980–4., DOI 10.1097/JTO. 0b013e31828f6989 Kenmotsu H, Niho S, Ito T, Ishikawa Y, Noguchi M, Tada H, et al. A Pilot Study of Adjuvant Chemotherapy with Irinotecan and Cisplatin for Completely Resected High-Grade Pulmonary Neuroendocrine Carcinoma, Large Cell Neuroendocrine Carcinoma and Small Cell Lung Cancer). Lung Cancer, 2014, 84(3):254–8., DOI 10.1016/j.lungcan.2014.03.007 Shah MH, Goldner WS, Benson AB, Bergsland E, Blaszkowsky LS, Brock P, et al. Neuroendocrine and Adrenal Tumors, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw, 2021, 19(7): 839–68., DOI 10.6004/jnccn.2021.0032 Rieber J, Schmitt J, Warth A, Muley T, Kappes J, Eichhorn F, et al. Outcome and Prognostic Factors of Multimodal Therapy for Pulmonary Large-Cell Neuroendocrine Carcinomas. Eur J Med Res, 2015, 20(1):64., DOI 10.1186/ s40001-015-0158-9 Jiang Y, Lei C, Zhang X, Cui Y, Che K, Shen H. Double-edged Role of Radiotherapy in Patients with Pulmonary Large-Cell Neuroendocrine Carcinoma. J Cancer, 2019, 10(25):6422–30., DOI 10.7150/jca.32446 Prelaj A, Rebuzzi SE, Del Bene G, Giron Berrios JR, Emiliani A, De Filippis L, et al. Evaluation of the Efficacy of Cisplatin-Etoposide and the Role of Thoracic Radiotherapy and Prophylactic Cranial Irradiation in LCNEC. ERJ Open Res, 2017, 3(1):00128–2016., DOI 10.1183/23120541. 00128-2016 De Pas TM, Giovannini M, Manzotti M, Trifiro G, Toffalorio F, Catania C, et al. Large-cell Neuroendocrine Carcinoma of the Lung Harboring EGFR Mutation and Responding to Gefitinib. J Clin Oncol, 2011, 29(34):e819–22., DOI 10.1200/JCO.2011.36.2251 Hayashi N, Fujita A, Saikai T, Takabatake H, Sotoshiro M, Sekine K, et al. Large Cell Neuroendocrine Carcinoma Harboring an Anaplastic Lymphoma Kinase, ALK, Rearrangement with Response to Alectinib. Intern Med, 2018, 57(5):713–6., DOI 10.2169/internalmedicine.9368-17 Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim DW, et al. Alectinib versus Crizotinib in Untreated ALK-Positive Non-small-cell Lung Cancer. N Engl J Med, 2017, 377(9):829–38., DOI 10.1056/NEJMoa1704795 Miyoshi T, Umemura S, Matsumura Y, Mimaki S, Tada S, Makinoshima H, et al. Genomic Profiling of Large-Cell Neuroendocrine Carcinoma of the Lung. Clin Cancer Res, 2017, 23(3):757–65., DOI 10.1158/1078-0432.CCR-16- 0355 Christopoulos P, Engel-Riedel W, Grohe C, Kropf-SanChen C, von Pawel J, Gutz S, et al. Everolimus with Paclitaxel and Carboplatin as First-Line Treatment for Metastatic Large-Cell Neuroendocrine Lung Carcinoma: a Multicenter Phase II Trial. Ann Oncol, 2017, 28(8):1898–902., DOI 10.1093/ annonc/mdx268 Odate S, Nakamura K, Onishi H, Kojima M, Uchiyama A, Nakano K, et al. TrkB/BDNF Signaling Pathway Is a Potential Therapeutic Target for Pulmonary Large Cell Neuroendocrine Carcinoma. Lung Cancer, 2013, 79(3):205–14., DOI 10.1016/j.lungcan.2012.12.004 Filosso PL, Ruffini E, Oliaro A, Rena O, Casadio C, Mancuso M, et al. Largecell Neuroendocrine Carcinoma of the Lung: a Clinicopathologic Study of Eighteen Cases and the Efficacy of Adjuvant Treatment with Octreotide. J Thorac Cardiovasc Surg, 2005, 129(4):819–24., DOI 10.1016/j.jtcvs.2004. 05.023 Yokouchi H, Kitahashi M, Oshitari T, Yamamoto S. Intravitreal Bevacizumab for Iris Tumor Metastasized from Large Cell Neuroendocrine Carcinoma of Lung. Graefes Arch Clin Exp Ophthalmol, 2013, 251(9):2243–5., DOI 10.1007/ s00417-012-2218-y Weber MM, Fottner C. Immune Checkpoint Inhibitors in the Treatment of Patients with Neuroendocrine Neoplasia. Oncol Res Treat, 2018, 41(5): 306–12., DOI 10.1159/000488996 Tsuruoka K, Horinouchi H, Goto Y, Kanda S, Fujiwara Y, Nokihara H, et al. PD-L1 Expression in Neuroendocrine Tumors of the Lung. Lung Cancer, 2017, 108:115–20., DOI 10.1016/j.lungcan.2017.03.006 Wang VE, Urisman A, Albacker L, Ali S, Miller V, Aggarwal R, et al. Checkpoint Inhibitor Is Active against Large Cell Neuroendocrine Carcinoma with High Tumor Mutation burden. J Immunother Cancer, 2017, 5(1):75., DOI 10.1186/s40425-017-0281-y Sherman S, RotemO, Shochat T, ZerA,MooreA, Dudnik E. Efficacy of Immune Check-point Inhibitors, ICPi, in Large Cell Neuroendocrine Tumors of Lung, LCNEC). Lung Cancer, 2020, 143:40–6., DOI 10.1016/j.lungcan.2020.03.008 Dudnik E, Kareff S, Moskovitz M, Kim C, Liu SV, Lobachov A, et al. Realworld Survival Outcomes with Immune Checkpoint Inhibitors in Large-Cell Neuroendocrine Tumors of Lung. J Immunother Cancer, 2021, 9(2):e001999., DOI 10.1136/jitc-2020-001999 Derks JL, Hendriks LE, Buikhuisen WA, Groen HJM, Thunnissen E, van Suylen RJ, et al. Clinical Features of Large Cell Neuroendocrine Carcinoma: a Population-Based Overview. Eur Respir J, 2016, 47(2):615–24., DOI 10.1183/ 13993003.00618-2015 Morise M, Hishida T, Takahashi A, Yoshida J, Ohe Y, Nagai K, et al. Clinicopathological Significance of Cancer Stem-like Cell Markers in High-Grade Neuroendocrine Carcinoma of the Lung. J Cancer Res Clin Oncol, 2015, 141(12):2121–30., DOI 10.1007/s00432-015-1985-3 Okui M, Yamamichi T, Asakawa A, Harada M, Saito M, Horio H. Prognostic Significance of Neutrophil-Lymphocyte Ratios in Large Cell Neuroendocrine Carcinoma. Gen Thorac Cardiovasc Surg, 2017, 65(11):633–9., DOI 10.1007/ s11748-017-0804-y Christopoulos P, Schneider MA, Bozorgmehr F, Kuon J, Engel-Riedel W, Kollmeier J, et al. Large Cell Neuroendocrine Lung Carcinoma Induces Peripheral T-Cell Repertoire Alterations with Predictive and Prognostic Significance. Lung Cancer, 2018, 119:48–55., DOI 10.1016/j.lungcan.2018. 03.002 Li Y, Shi X, Mao B, Wang L, Wu L, Li J, et al. The Genomic Mutational Landscape and its Correlation with TMB, PD-L1 Expression and CD8+ T Cell Infiltration in Chinese Lung Large Cell Neuroendocrine Carcinoma. Lung Cancer, 2022, 166:161–9., DOI 10.1016/j.lungcan.2022.01.007 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610730 EP 1610740 DI 10.3389/pore.2022.1610730 PG 11 ER PT J AU Wang, LJ Liu, L Ju, W Yao, WX Yang, XH Qian, WH AF Wang, Li-Jun Liu, Liu Ju, Wei Yao, Wen-Xin Yang, Xi-Hu Qian, Wen-Hao TI 20 abnormal metabolites of Stage IV Grade C periodontitis was discovered by CPSI-MS SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE metabolomics; point-of-care test; conducting polymer spray ionization; saliva metabolomics; severe periodontitis (Stage IV and Grade C) ID metabolomics; point-of-care test; conducting polymer spray ionization; saliva metabolomics; severe periodontitis (Stage IV and Grade C) AB Saliva is a noninvasive biofluid that contains the metabolic signature of severe periodontitis (SP, Stage IV and Grade C). Conductive polymer spray ionization mass spectrometry (CPSI-MS) was used to record a wide range of metabolites within a few seconds, making this technique a promising point-of-care method for the early detection of SP (Stage IV and Grade C). Saliva samples from 31 volunteers, consisting of 16 healthy controls (HC) and 15 patients with SP (Stage IV and Grade C), were collected to identify dysregulated metabolites. Twenty metabolites were screened out, including seven amino acids. Moreover, the results showed that amino acid metabolism is closely related to the development of periodontitis. The present study further confirmed that salivary metabolites in the oral cavity were significantly altered after plaque removal. These results suggest that the combination of CPSI-MS is a feasible tool for preclinical screening of SP (Stage IV and Grade C). C1 [Wang, Li-Jun] Xuhui District Dental Center, Department of PeriodontitisShanghai, China. [Liu, Liu] Shanghai Jiao Tong University School of Medicine, Ninth People’s Hospital, Department of Oral and Maxillofacial-Head & Neck OncologyShanghai, China. [Ju, Wei] Affiliated Hospital of Jiangsu University, Department of Oral and Maxillofacial SurgeryZhenjiang, Jiangsu, China. [Yao, Wen-Xin] Xuhui District Dental Center, Department of PeriodontitisShanghai, China. [Yang, Xi-Hu] Affiliated Hospital of Jiangsu University, Department of Oral and Maxillofacial SurgeryZhenjiang, Jiangsu, China. [Qian, Wen-Hao] Xuhui District Dental Center, Department of PeriodontitisShanghai, China. RP Qian, WH (reprint author), Xuhui District Dental Center, Department of Periodontitis, Shanghai, China. EM pingyanlaoto@163.com CR Papapanou PN, Sanz M, Buduneli N, Dietrich T, Feres M, Fine DH, et al. Periodontitis: Consensus report of workgroup 2 of the 2017 world workshop on the classification of periodontal and peri-implant diseases and conditions. J Periodontol, 2018, 89:S173–S182., DOI 10.1002/JPER.17-0721 Sawle AD, Kebschull M, Demmer RT, Papapanou PN. Identification of master regulatorgenes in human periodontitis. J Dent Res, 2016, 95:1010–7., DOI 10.1177/ 0022034516653588 Trindade F, Oppenheim FG, Helmerhorst EJ, Amado F, Gomes PS, Vitorino R. Uncovering the molecular networks in periodontitis. Proteomics Clin Appl, 2014, 8: 748–61., DOI 10.1002/prca.201400028 Grover HS, Kapoor S, Saksena N. Periodontal proteomics: Wonders never cease. Int J Proteomics, 2013, 2013:850235., DOI 10.1155/2013/850235 Zhang Y, Sun J, Lin CC, Abemayor E,Wang MB,Wong DT. The emerging landscape of salivary diagnostics. Periodontol 2000, 2016, 70:38–52., DOI 10.1111/prd.12099 de Almenida Pdel V, Gregio AM, Machado MA, de Lima AA, Azevedo LR. Saliva composition and functions: A comprehensive review. J Contemp Dent Pract, 2008, 9:72–80., DOI 10.5005/jcdp-9-3-72 Yoshizawa JM, Schafer CA, Schafer JJ, Farrell JJ, Paster BJ, Wong DT. Salivary biomarkers: Toward future clinical and diagnostic utilities. Clin Microbiol Rev, 2013, 26:781–91., DOI 10.1128/CMR.00021-13 Wallner-Liebmann S, Tenori L, Mazzoleni A, Dieber-Rotheneder M, Konrad M, Hofmann P, et al. Individual human metabolic phenotype analyzed by 1H NMR of saliva samples. J Proteome Res, 2016, 15:1787–93., DOI 10.1021/acs.jproteome.5b01060 Zhang A, Sun H, Yan G, Wang P, Wang X. Metabolomics for biomarker discovery: Moving to the clinic. Biomed Res Int, 2015, 2015:354671., DOI 10.1155/2015/354671 Vignoli A, Ghini V, Meoni G, Licari C, Takis PG, Tenori L, et al. Highthroughputmetabolomics by 1D NMR. Angew Chem Int Ed Engl, 2019, 58:968–94., DOI 10.1002/anie.201804736 Takis PG, Ghini V, Tenori L, Turano P, Luchinat C. Uniqueness of the NMR approach to metabolomics. Trac Trends Anal Chem, 2019, 120:115300., DOI 10.1016/j.trac. 2018.10.036 Nannini G, Meoni G, Amedei A, Tenori L. Metabolomics profile in gastrointestinal cancers: Update and future perspectives. World J Gastroenterol, 2020, 26:2514–32., DOI 10.3748/wjg.v26.i20.2514 Kuboniwa M, Sakanaka A, Hashino E, Bamba T, Fukusaki E, Amano A, et al. Prediction of periodontal inflammation via metabolic profiling of saliva. J Dent Res, 2016, 95:1381–6., DOI 10.1177/0022034516661142 Rzeznik M, Triba MN, Levy P, Jungo S, Botosoa E, Duchemann B, et al. Identification of a discriminative metabolomic fingerprint of potential clinical relevance in saliva of patients with periodontitis using 1H nuclear magnetic resonance, NMR, spectroscopy. PLoS ONE, 2017, 12:e0182767., DOI 10.1371/ journal.pone.0182767 Pu F, Chiang S, Zhang W, Ouyang Z. Direct sampling mass spectrometry for clinical analysis. Analyst, 2019, 144:1034–51., DOI 10.1039/c8an01722k Yao YN, Di D, Yuan ZC, Wu L, Hu B. Schirmer paper noninvasive microsampling for direct mass spectrometry analysis of human tears. Anal Chem, 2020, 92:6207–12., DOI 10.1021/acs.analchem.9b05078 Mendes TPP, Pereira I, de Lima LAS, Morais CLM, Neves ACON, Martin FL, Lima KMG, et al. Paper spray ionization mass spectrometry as a potential tool for early diagnosis of cervical cancer. J Am Soc Mass Spectrom, 2020, 31:1665–72., DOI 10.1021/jasms.0c00111 Huang YC, Chung HH, Dutkiewicz EP, Chen CL, Hsieh HY, Chen BR, et al. Predicting breast cancer by paper spray ion mobility spectrometry mass spectrometry and machine learning. Anal Chem, 2020, 92:1653–7., DOI 10.1021/ acs.analchem.9b03966 Song X, Narayanan R, Shankar V, Ethiraj S, Wang X, Duan N, et al. Oral squamous cell carcinoma diagnosed from saliva metabolic profiling. Proc Natl Acad Sci U S A, 2020, 17:16167–73., DOI 10.1073/pnas.2001395117 Song XW, Chen H, Zare RN. Conductive polymer spray ionization mass spectrometry for biofluid analysis. Anal Chem, 2018, 90:12878–85., DOI 10.1021/acs. analchem.8b03460 Kuboniwa M, Sakanaka A, Hashino E, Bamba T, Fukusaki E. Prediction of periodontal inflammation via metabolic profiling of saliva. J Dent Res, 2016, 95(12): 1381–6., DOI 10.1177/0022034516661142 Barnes VM, Ciancio SG, Shibly O, Xu T, Devizio W, Trivedi HM, et al. Metabolomics reveals elevated macromolecular degradation in periodontal disease. J Dent Res, 2011, 90(11):1293–7., DOI 10.1177/0022034511416240 Barnes VM, Kennedy AD, Panagakos F, DevizioW, Trivedi HM, Jonsson T, et al. Global metabolomic analysis of human saliva and plasma from healthy and diabetic subjects, with and without periodontal disease. PLoS One, 2014, 9(8):e105181., DOI 10.1371/ journal.pone.0105181 Aimetti M, Cacciatore S, Graziano A, Tenori L. Metabonomic analysis of saliva reveals generalized chronic periodontitis signature. Metabolomics, 2012, 8(3): 465–74., DOI 10.1007/s11306-011-0331-2 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610739 EP 1610746 DI 10.3389/pore.2022.1610739 PG 8 ER PT J AU Susztak, N Besznyak, I Almasi, K Bursics, A Kelemen, D Borowski, D Banky, B AF Susztak, Nora Besznyak, Istvan Almasi, Kalman Bursics, Attila Kelemen, Dora Borowski, W. David Banky, Balazs TI Improved Accuracy of Lymph Node Staging and Long-Term Survival Benefit in Colorectal Cancer With Ex Vivo Arterial Methylene Blue Infiltration SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal cancer; overall survival; methylene blue; nodal staging accuracy; lymph node staging ID colorectal cancer; overall survival; methylene blue; nodal staging accuracy; lymph node staging AB Introduction: Ex vivo methylene blue (MB) injection into the main supplying arteries of the colorectal specimen after surgical removal is an uncomplicated technique to support lymph node harvest during pathological evaluation. The primary aim of this randomized, interventional, bicentric trialwas to evaluate the impact ofMB injection on lymph node yield, with secondary aims assessing the accuracy of lymph node staging and the effect on 5- year overall survival for patients undergoing resection of colorectal cancer. Methods: In the study period between December 2013 and August 2015, 200 colorectal resections were performed at two independent onco-surgery centers of Hungary. Following surgical resection, each specimen was randomly assigned either to the control (standard pathological work-up) or to the MB staining group before formaldehyde fixation. Patient-level surgical and clinical data were retrieved from routinely collected clinical datasets. Survival status data were obtained from the National Health Insurance Fund of Hungary. Results: A total of 162 specimens, 82 in the control and 80 in the MB groups, were included for analysis. Baseline characteristics were equally distributed among study groups, except for specimen length. Both the median of total number of lymph nodes retrieved (control 11 ± 8 [0–33] nodes vs. MB 14 ± 6 [0–42] nodes; p < 0.01), and the ratio of cases with at least 12 removed lymph nodes (36/82, 43.9% vs. 53/80, 66.3%; p < 0.01) were higher in the MB group. The rate of accurate lymph node staging was nonsignificantly improved. As for rectal cancer, nodal staging accuracy (16/31, 51.6% vs. 23/30, 76.7%; p = 0.04) and the proportion with minimum 12 lymph node retrieval (7/31, 22.6%, vs. 18/30, 60%; p < 0.01) was improved by MB injection. In Mantel–Cox regression, a statistically significant survival benefit with methylene blue injection at 5 years post-surgery was proven (51.2% vs. 68.8%; p = 0.04). Conclusion: In our experience, postoperative ex vivo arterial methylene blue injection appears to be an uncomplicated technique, improving lymph node yield and decreasing the chance of insufficient nodal staging. The technique might also associate with a 5-year overall survival benefit. C1 [Susztak, Nora] Semmelweis University, Faculty of MedicineBudapest, Hungary. [Besznyak, Istvan] Fovarosi Onkormanyzat Uzsoki utcai Oktato Korhaz, Sebeszeti-Ersebeszeti OsztalyBudapest, Hungary. [Almasi, Kalman] Petz Aladar Hospital, Department of PathologyGyor, Hungary. [Bursics, Attila] Fovarosi Onkormanyzat Uzsoki utcai Oktato Korhaz, Sebeszeti-Ersebeszeti OsztalyBudapest, Hungary. [Kelemen, Dora] Fovarosi Uzsoki utcai Korhaz, PathologiaBudapest, Hungary. [Borowski, W. David] Welwitschia Hospital, Department of SurgeryWalvis Bay, Namibia. [Banky, Balazs] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. RP Susztak, N (reprint author), Semmelweis University, Faculty of Medicine, Budapest, Hungary. EM nsusztak@gmail.com CR Observatory WGC. Estimated Number of Deaths Worldwide, Both Sexes, All Ages, Graph,, 2021). Available at: https://gco.iarc.fr/, Accessed 04 2021, 01). Wright FC, Law CH, Berry S, Smith AJ. Clinically Important Aspects of Lymph Node Assessment in colon Cancer. J Surg Oncol, 2009, 99(4):248–55., DOI 10. 1002/jso.21226 Hohenberger W, Weber K, Matzel K, Papadopoulos T, Merkel S. Standardized Surgery for Colonic Cancer: Complete Mesocolic Excision and central Ligation-Ttechnical Notes and Outcome. Colorectal Dis, 2009, 11(4): 354–64. discussion 64-5., DOI 10.1111/j.1463-1318.2008.01735.x Havenga K, Enker WE, Norstein J, Moriya Y, Heald RJ, van Houwelingen HC, et al. Improved Survival and Local Control after Total Mesorectal Excision or D3 Lymphadenectomy in the Treatment of Primary Rectal Cancer: an International Analysis of 1411 Patients. Eur J Surg Oncol, 1999, 25(4): 368–74., DOI 10.1053/ejso.1999.0659 Kulendran M, Stebbing JF, Marks CG, Rockall TA. Predictive and Prognostic Factors in Colorectal Cancer: a Personalized Approach. Cancers, Basel,, 2011, 3(2):1622–38., DOI 10.3390/cancers3021622 Le Voyer TE, Sigurdson ER, Hanlon AL, Mayer RJ,Macdonald JS, Catalano PJ, et al. Colon Cancer Survival Is Associated with Increasing Number of Lymph Nodes Analyzed: a Secondary Survey of Intergroup Trial INT-0089. J Clin Oncol, 2003, 21(15):2912–9., DOI 10.1200/JCO.2003.05.062 Sobin LH, Gospodarowicz MK, Wittekind C. 7th Ed. UICC International Union against Cancer. Wiley-Blackwell, 2009). TNM Classification of Malignant Tumours Lei P, Ruan Y, Liu J, Zhang Q, Tang X, Wu J. Prognostic Impact of the Number of Examined Lymph Nodes in Stage II Colorectal Adenocarcinoma: A Retrospective Study. Gastroenterol Res Pract, 2020, 2020:8065972., DOI 10. 1155/2020/8065972 Liu J, Huang P, Zheng Z, Chen T, Wei H. Modified Methylene Blue Injection Improves Lymph Node Harvest in Rectal Cancer. ANZ J Surg, 2017, 87(4): 247–51., DOI 10.1111/ans.12889 Hamza A, Sakhi R, Khawar S, Alrajjal A, Edens J, Khurram MS, et al. Role of "Second Look" Lymph Node Search in Harvesting Optimal Number of Lymph Nodes for Staging of Colorectal Carcinoma. Gastroenterol Res Pract, 2018, 2018:1985031., DOI 10.1155/2018/1985031 Gurawalia J, Dev K, Nayak SP, Kurpad V, Pandey A. Less Than 12 Lymph Nodes in the Surgical Specimen after Neoadjuvant Chemo-Radiotherapy: an Indicator of Tumor Regression in Locally Advanced Rectal Cancer? J Gastrointest Oncol, 2016, 7(6):946–57., DOI 10.21037/jgo.2016.09.03 Trepanier M, Erkan A, Kouyoumdjian A, Nassif G, Albert M, Monson J, et al. Examining the Relationship between Lymph Node Harvest and Survival in Patients Undergoing Colectomy for colon Adenocarcinoma. Surgery, 2019, 166(4):639–47., DOI 10.1016/j.surg.2019.03.027 Tsai HL, Lu CY, Hsieh JS, Wu DC, Jan CM, Chai CY, et al. The Prognostic Significance of Total Lymph Node Harvest in Patients with T2-4N0M0 Colorectal Cancer. J Gastrointest Surg, 2007, 11(5):660–5., DOI 10.1007/ s11605-007-0119-x Topor B, Acland R, Kolodko V, Galandiuk S. Mesorectal Lymph Nodes: Their Location and Distribution within the Mesorectum. Dis Colon Rectum, 2003, 46(6):779–85., DOI 10.1007/s10350-004-6656-4 Markl B, Rossle J, Arnholdt HM, Schaller T, Krammer I, Cacchi C, et al. The Clinical Significance of Lymph Node Size in colon Cancer. Mod Pathol, 2012, 25(10):1413–22., DOI 10.1038/modpathol.2012.92 Cawthorn SJ, Gibbs NM, Marks CG. Clearance Technique for the Detection of Lymph Nodes in Colorectal Cancer. Br J Surg, 1986, 73(1):58–60., DOI 10.1002/ bjs.1800730124 Basten O, Bandorski D, Bismarck C, Neumann K, Fisseler-Eckhoff A. Acetone Compression. A Fast, Standardized Method to Investigate Gastrointestinal Lymph Nodes. Pathologe, 2010, 31(3):218–24., DOI 10.1007/s00292-009-1256-7 Reese JA, Hall C, Bowles K, Moesinger RC. Colorectal Surgical Specimen Lymph Node Harvest: Improvement of Lymph Node Yield with a Pathology Assistant. J Gastrointest Surg, 2009, 13(8):1459–63., DOI 10.1007/s11605-009- 0820-z Markl B, Kerwel TG, Wagner T, Anthuber M, Arnholdt HM. Methylene Blue Injection into the Rectal Artery as a Simple Method to Improve Lymph Node Harvest in Rectal Cancer. Mod Pathol, 2007, 20(7):797–801., DOI 10.1038/ modpathol.3800824 Borowski DW, Banky B, Banerjee AK, Agarwal AK, Tabaqchali MA, Garg DK, et al. Intra-arterial Methylene Blue Injection into Ex Vivo Colorectal Cancer Specimens Improves Lymph Node Staging Accuracy: a Randomized Controlled Trial. Colorectal Dis, 2014, 16(9):681–9., DOI 10.1111/codi.12681 Markl B, Schaller T, Krammer I, Cacchi C, Arnholdt HM, Schenkirsch G, et al. Methylene Blue-Assisted Lymph Node Dissection Technique Is Not Associated with an Increased Detection of Lymph Node Metastases in Colorectal Cancer. Mod Pathol, 2013, 26(9):1246–54., DOI 10.1038/ modpathol.2013.61 Cai HK, He HF, Tian W, Zhou MQ, Hu Y, Deng YC. Colorectal Cancer Lymph Node Staining by Activated Carbon Nanoparticles Suspension In Vivo or Methylene Blue In Vitro. World J Gastroenterol, 2012, 18(42):6148–54., DOI 10. 3748/wjg.v18.i42.6148 Tornroos A, Shabo I, Druvefors B, Arbman G, Olsson H. Postoperative Intraarterial Methylene Blue Injection of Colorectal Cancer Specimens Increases the Number of Lymph Nodes Recovered. Histopathology, 2011, 58(3):408–13., DOI 10.1111/j.1365-2559.2011.03755.x Kerwel TG, Spatz J, Anthuber M, Wunsch K, Arnholdt H, Markl B. Injecting Methylene Blue into the Inferior Mesenteric Artery Assures an Adequate Lymph Node Harvest and Eliminates Pathologist Variability in Nodal Staging for Rectal Cancer. Dis Colon Rectum, 2009, 52(5):935–41., DOI 10.1007/DCR. 0b013e31819f28c9 Markl B, Kerwel T, Jahnig H, Anthuber M, Arnholdt H. Lymph Node Preparation in Colorectal Cancer. Ex Vivo Methylene Blue Injection as a Novel Technique to Improve Lymph Node Visualization. Pathologe, 2008, 29(4):274–9., DOI 10.1007/s00292-007-0950-6 Markl B, Olbrich G, Schenkirsch G, Kretsinger H, Kriening B, Anthuber M. Clinical Significance of International Union against Cancer pN Staging and Lymph Node Ratio in Node-Positive Colorectal Cancer after Advanced Lymph Node Dissection. Dis Colon Rectum, 2016, 59(5):386–95., DOI 10.1097/DCR. 0000000000000569 Reima H, Saar H, Innos K, Soplepmann J. Methylene Blue Intra-arterial Staining of Resected Colorectal Cancer Specimens Improves Accuracy of Nodal Staging: A Randomized Controlled Trial. Eur J Surg Oncol, 2016, 42(11):1642–6., DOI 10.1016/j.ejso.2016.06.001 Vasala A, Nair HG, Rao ST, Tagore KR, Murthy SS, Fonseca D. Impact of Methylene Blue Staining in the Retrieval of Lymph Nodes in Resected Colorectal Cancer Specimens. Indian J Pathol Microbiol, 2016, 59(4): 504–6., DOI 10.4103/0377-4929.191804 Munster M, Hanisch U, Tuffaha M, Kube R, Ptok H. Ex Vivo Intra-arterial Methylene Blue Injection in Rectal Cancer Specimens Increases the Lymph- Node Harvest, Especially after Preoperative Radiation. World J Surg, 2016, 40(2):463–70., DOI 10.1007/s00268-015-3230-2 Kir G, Alimoglu O, Sarbay BC, Bas G. Ex Vivo intra-arterial Methylene Blue Injection in the Operation Theater May Improve the Detection of Lymph Node Metastases in Colorectal Cancer. Pathol Res Pract, 2014, 210(12): 818–21., DOI 10.1016/j.prp.2014.09.003 Jepsen RK, Ingeholm P, Lund EL. Upstaging of Early Colorectal Cancers Following Improved Lymph Node Yield after Methylene Blue Injection. Histopathology, 2012, 61(5):788–94., DOI 10.1111/j.1365-2559.2012.04287.x Klepsyte E, Samalavicius NE. Injection of Methylene Blue Solution into the InferiorMesenteric Artery of Resected Rectal Specimens for Rectal Cancer as a Method for Increasing the Lymph Node Harvest. Tech Coloproctol, 2012, 16(3):207–11., DOI 10.1007/s10151-012-0816-7 Markl B, Kerwel TG, Jahnig HG, Oruzio D, Arnholdt HM, Scholer C, et al. Methylene Blue-Assisted Lymph Node Dissection in colon Specimens: a Prospective, Randomized Study. Am J Clin Pathol, 2008, 130(6):913–9., DOI 10.1309/AJCPVAPB5APABJNX Chapman B, Paquette C, Tooke C, Schwartz M, Osler T, Weaver D, et al. Impact of Schwartz Enhanced Visualization Solution on Staging Colorectal Cancer and Clinicopathological Features Associated with Lymph Node Count. Dis Colon Rectum, 2013, 56(9):1028–35., DOI 10. 1097/DCR.0b013e31829c41ba Johnson PM, Malatjalian D, Porter GA. Adequacy of Nodal Harvest in Colorectal Cancer: a Consecutive Cohort Study. J Gastrointest Surg, 2002, 6(6):883–8. discussion 9-90., DOI 10.1016/s1091-255x(02)00131-2 Marks JH, Valsdottir EB, Rather AA, Nweze IC, Newman DA, Chernick MR. Fewer Than 12 Lymph Nodes Can Be Expected in a Surgical Specimen after High-Dose Chemoradiation Therapy for Rectal Cancer. Dis Colon Rectum, 2010, 53(7):1023–9., DOI 10.1007/DCR.0b013e3181dadeb4 VatherR, Sammour T, KahokehrA,ConnollyA, HillA. Quantitative LymphNode Evaluation as an Independent Marker of Long-Term Prognosis in Stage III Rectal Cancer. ANZ J Surg, 2011, 81(12):883–8., DOI 10.1111/j.1445-2197.2010.05595.x Resch A, Langner C. Lymph Node Staging in Colorectal Cancer: Old Controversies and Recent Advances. World J Gastroenterol, 2013, 19(46): 8515–26., DOI 10.3748/wjg.v19.i46.8515 Haboubi NY, Abdalla SA, Amini S, Clark P, Dougal M, Dube A, et al. The Novel Combination of Fat Clearance and Immunohistochemistry Improves Prediction of the Outcome of Patients with Colorectal Carcinomas: a Preliminary Study. Int J Colorectal Dis, 1998, 13(2):99–102., DOI 10.1007/ s003840050143 Wijesuriya RE, Deen KI, Hewavisenthi J, Balawardana J, Perera M. Neoadjuvant Therapy for Rectal Cancer Down-Stages the Tumor but Reduces Lymph Node Harvest Significantly. Surg Today, 2005, 35(6): 442–5., DOI 10.1007/s00595-004-2956-5 Baxter NN, Morris AM, Rothenberger DA, Tepper JE. Impact of Preoperative Radiation for Rectal Cancer on Subsequent Lymph Node Evaluation: a Population-Based Analysis. Int J Radiat Oncol Biol Phys, 2005, 61(2): 426–31., DOI 10.1016/j.ijrobp.2004.06.259 Mekenkamp LJ, van Krieken JH, Marijnen CA, van de Velde CJ, Nagtegaal ID, Pathology Review C, et al. Lymph Node Retrieval in Rectal Cancer Is Dependent on many Factors-Tthe Role of the Tumor, the Patient, the Surgeon, the Radiotherapist, and the Pathologist. Am J Surg Pathol, 2009, 33(10):1547–53., DOI 10.1097/PAS.0b013e3181b2e01f Foo CC, Ku C, Wei R, Yip J, Tsang J, Chan TY, et al. How Does Lymph Node Yield Affect Survival Outcomes of Stage I and II colon Cancer? World J Surg Oncol, 2020, 18(1):22., DOI 10.1186/s12957-020-1802-6 Argiles G, Tabernero J, Labianca R, Hochhauser D, Salazar R, Iveson T, et al. Localised colon Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol, 2020, 31(10):1291–305., DOI 10.1016/j. annonc.2020.06.022 Benson AB, Venook AP, Al-Hawary MM, Arain MA, Chen YJ, Ciombor KK, et al. Colon Cancer, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw, 2021, 19(3):329–59., DOI 10.6004/jnccn. 2021.0012 Wells KO, Hawkins AT, Krishnamurthy DM, Dharmarajan S, Glasgow SC, Hunt SR, et al. Omission of Adjuvant Chemotherapy Is Associated with Increased Mortality in Patients with T3N0 Colon Cancer with Inadequate Lymph Node Harvest. Dis Colon Rectum, 2017, 60(1):15–21., DOI 10.1097/ DCR.0000000000000729 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610742 EP 1610751 DI 10.3389/pore.2022.1610742 PG 10 ER PT J AU Li, M Yang, L Lu, H AF Li, Meihui Yang, Lan Lu, Hongyang TI Pulmonary Combined Large Cell Neuroendocrine Carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE pulmonary combined large cell neuroendocrine carcinoma (CLCNEC); pathological characteristics; diagnosis; therapy; targeted therapy ID pulmonary combined large cell neuroendocrine carcinoma (CLCNEC); pathological characteristics; diagnosis; therapy; targeted therapy AB Pulmonary combined large-cell neuroendocrine carcinoma (CLCNEC) is a rare neuroendocrine tumor pertained to lung large cell neuroendocrine carcinoma (LCNEC) with aggressive behavior and poor prognosis generally. The clinical features of CLCNEC are not specific including cough, expectoration, chest distress, chest pain, etc., which are prone to have different manifestations of the mixed components. Owing to the low incidence, there are few related smallscale retrospective studies and case reports. Currently, the treatment regimen of CLCNEC mainly refers to LCNEC that complete surgical resection is preferred in the early stage and according to previous researches, platinumbased small cell lung cancer (SCLC) standard treatment regimen showed promising results in postoperative and advanced CLCNEC as compared to that of non-small cell lung cancer (NSCLC). Adenocarcinoma-CLCNEC more likely harbor driver gene mutation, and may benefit from targeted therapy. As for immunotherapy, more clinical trial data are needed to support its benefits. This article will fill the gap and will provide new insight into the clinical characteristics, pathological diagnosis and treatment endeavors of CLCNEC. C1 [Li, Meihui] Zhejiang Cancer Hospital, Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus)Hangzhou, China. [Yang, Lan] Zhejiang Cancer Hospital, Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus)Hangzhou, China. [Lu, Hongyang] Zhejiang Cancer Hospital, Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus)Hangzhou, China. RP Lu, H (reprint author), Zhejiang Cancer Hospital, Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Hangzhou, China. EM luhy@zjcc.org.cn CR Tanimoto H, Hamasaki A, Akimoto Y, Honda H, Takao Y, Okamoto K, et al. A case of large cell neuroendocrine carcinoma, LCNEC, of the uterine cervix successfully treated by postoperative CPT-11+CDDP chemotherapy after non-curative surgery. Gan To Kagaku Ryoho, 2012, 39(9):1439–41. Lee KW, Lee Y, Oh SW, Jin KN, Goo JM. Large cell neuroendocrine carcinoma of the lung: CT and FDG PET findings. Eur J Radiol, 2015, 84(11):2332–8., DOI 10. 1016/j.ejrad.2015.07.033 Hendifar AE, Marchevsky AM, Tuli R. Neuroendocrine tumors of the lung: Current challenges and advances in the diagnosis and management of welldifferentiated disease. J Thorac Oncol, 2017, 12(3):425–36., DOI 10.1016/j.jtho. 2016.11.2222 Coleman N, Wotherspoon A, Yousaf N, Popat S. Transformation to neuroendocrine carcinoma as a resistance mechanism to lorlatinib. Lung Cancer, 2019, 134(8):117–20., DOI 10.1016/j.lungcan.2019.05.025 Fasano M, Corte CM, Papaccio F, Ciardiello F,Morgillo F. Pulmonary large-cell neuroendocrine carcinoma: from epidemiology to therapy. J Thorac Oncol, 2015, 10(8):1133–41., DOI 10.1097/jto.0000000000000589 WHO Classification of Tumours Editorial Board. WHO Classification of Tumours. Thoracic Tumours [M]. 5th ed. Lyon: IARC Press, 2021). Cakir E, Demirag E, Aydin M, Unsal E. Clinicopathologic features and prognostic significance of lung tumours with mixed histologic patterns. Acta Chir Belg, 2009, 109(4):489–93., DOI 10.1080/00015458.2009.11680466 Naidoo J, Santos-ZabalaML, Iyriboz T,Woo KM, Sima CS, Fiore JJ, et al. Large cell neuroendocrine carcinoma of the lung: clinico-pathologic features, treatment, and outcomes [J]. Clin Lung Cancer, 2016, 17(5):121–9. Travis WD, Linnoila RI, Tsokos MG, Hitchcock CL, Cutler GB, Jr, Nieman L, et al. Neuroendocrine tumors of the lung with proposed criteria for large-cell neuroendocrine carcinoma. An ultrastructural, immunohistochemical, and flow cytometric study of 35 cases. Am J Surg Pathol, 1991, 15(6):529–53., DOI 10.1097/ 00000478-199106000-00003 Kujtan L, Muthukumar V, Kennedy KF, Davis JR, Masood A, Subramanian J. The role of systemic therapy in the management of stage i large cell neuroendocrine carcinoma of the lung. J Thorac Oncol, 2018, 13(5):707–14., DOI 10.1016/j.jtho.2018. 01.019 Beasley MB, Brambilla E, Travis WD. The 2004 World Health Organization classification of lung tumors. Semin Roentgenol, 2005, 40(2):90–7., DOI 10.1053/j.ro. 2005.01.001 Zhang JT, Li Y, Yan LX, Zhu ZF, Dong XR, Chu Q, et al. Disparity in clinical outcomes between pure and combined pulmonary large-cell neuroendocrine carcinoma: A multi-center retrospective study. Lung Cancer, 2020, 139:118–23., DOI 10.1016/j.lungcan.2019.11.004 Wang YN, Qian FF, Chen Y, Yang ZY, Hu MJ, Lu J, et al. Comparative study of pulmonary combined large-cell neuroendocrine carcinoma and combined smallcell carcinoma in surgically resected high-grade neuroendocrine tumors of the lung. Front Oncol, 2021, 11:714549., DOI 10.3389/fonc.2021.714549 Mangum MD, Greco FA, Hainsworth JD, Hande KR, Johnson DH. Combined small-cell and non-small-cell lung cancer. J Clin Oncol, 1989, 7(5):607–12., DOI 10. 1200/JCO.1989.7.5.607 Miyoshi T, Umemura S, Matsumura Y, Mimaki S, Tada S, Makinoshima H, et al. Genomic profiling of large-cell neuroendocrine carcinoma of the lung. Clin Cancer Res, 2017, 23(3):757–65., DOI 10.1158/1078-0432.CCR-16-0355 Shen YC, Hu F, Li CH, Xu JL, Zhong RB, Zhang XY, et al. Clinical features and outcomes analysis of surgical resected pulmonary large-cell neuroendocrine carcinoma with adjuvant chemotherapy. Front Oncol, 2020, 10:556194., DOI 10. 3389/fonc.2020.556194 Isaka M, Nakagawa K, Ohde Y, Okumura T, Watanabe R, Ito I, et al. A clinicopathological study of peripheral, small-sized high-grade neuroendocrine tumours of the lung: differences between small-cell lung carcinoma and largecell neuroendocrine carcinoma. Eur J Cardiothorac Surg, 2012, 41(4):841–6., DOI 10. 1093/ejcts/ezr132 Wang J, Ye L, Cai H, Jin ML. Comparative study of large cell neuroendocrine carcinoma and small cell lung carcinoma in high-grade neuroendocrine tumors of the lung: a large population-based study. J Cancer, 2019, 10(18):4226–36., DOI 10. 7150/jca.33367 Cirak AK, Ceylan KC, Varol Y, Karadeniz G, Aydogdu Z, Acar A, et al. Characteristics of patients with large-cell neuroendocrine carcinoma of the lung. Turk Thorac J, 2020, 21(3):150–5., DOI 10.5152/TurkThoracJ.2019.180166 Escuin SC. Diagnosis and treatment of neuroendocrine lung tumors. Arch Bronconeumol, 2014, 50(9):392–6., DOI 10.1016/j.arbres.2014.02.004 Handa Y, Tsutani Y, Ito M, Miyata Y, Mukaida H, Kaneko M, et al. Clinical behavior of combined versus pure high-grade neuroendocrine carcinoma. Clin Lung Cancer, 2022, 23(1):9–e16., DOI 10.1016/j.cllc.2021.06.010 Yang ZY, Wang YN, Chen Y, Qian FF, Zhang YW, Hu MJ, et al. Combined large cell neuroendocrine carcinoma: clinical characteristics, prognosis and postoperative management. Eur J Cardiothorac Surg, 2022, 62(2):ezac069., DOI 10.1093/ejcts/ezac069 Katsenos S, Karachaliou I, Archondakis S. Mixed squamous and large-cell carcinoma of the lung: a case study and literature review. J Cancer Res Ther, 2012, 8(3):445–7., DOI 10.4103/0973-1482.103531 Masuya D, Gotoh M, Nakashima T, Liu D, Ishikawa S, Yamamoto Y, et al. Combined large cell neuroendocrine carcinoma and squamous cell carcinoma of the lung; report of a case. Kyobu Geka, 2006, 59(6):491–5. Cai C, Su X, Lin J, Li X. Combined lung large cell neuroendocrine carcinoma with squamous cell carcinoma differentiation: a case report [J]. J China-Japan Friendship Hosp, 2018, 32(06):370–86. Goto T, Maeshima A, Kato R. Combined large cell neuroendocrine carcinoma and spindle cell carcinoma of the lung. Jpn J Clin Oncol, 2011, 41(6):797–802., DOI 10.1093/jjco/hyr034 Yang WJ, Han BH. The current status of management and research progress of large cell neuroendocrine lung carcinoma’s [J]. Oncol Prog, 2017, 15(11):1250–4. Travis WD, Brambilla E, Burke AP, Marx A, Nicholson AG. Introduction to the 2015 World Health Organization classification of tumors of the Lung, Pleura, Thymus, and Heart. J Thorac Oncol, 2015, 10(9):1240–2., DOI 10.1097/JTO. 0000000000000663 Tsutsumi R, Kataoka N, Kunimatsu Y, Sato I, Tanimura M, Nakano T, et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel for managing combined large-cell neuroendocrine carcinoma: A case report. Respirol Case Rep, 2022, 10(7):e0989., DOI 10.1002/rcr2.989 Ando T, Kage H, Shinozaki-Ushiku A, Tatsuno K, Tsutsumi S, Nagayama K, et al. Composite clonal analysis reveals transition of NSCLC subtypes through accumulation of gene mutations: A case report. JTO Clin Res Rep, 2022, 3(2): 100277., DOI 10.1016/j.jtocrr.2022.100277 Lim CA, Banyi N, Tucker T, Ionescu DN, Melosky B. A case of ALKrearranged combined lung adenocarcinoma and neuroendocrine carcinoma with diffuse bone metastasis and partial response to alectinib. Curr Oncol, 2022, 29(2): 848–52., DOI 10.3390/curroncol29020072 Sakamoto T, Arai K, Makishima K, Yamasaki A. BRAF V600E-mutated combined large cell neuroendocrine carcinoma and adenocarcinoma responding to targeted therapy. BMJ Case Rep, 2021, 14(12):e243295., DOI 10.1136/bcr-2021-243295 Rossi G, Cavazza A, Marchioni A, Longo L, MigaldiM, Sartori G, et al. Role of chemotherapy and the receptor tyrosine kinases kit, PDGFRalpha, PDGFRbeta, and Met in large-cell neuroendocrine carcinoma of the lung. J Clin Oncol, 2005, 23(34): 8774–85., DOI 10.1200/JCO.2005.02.8233 Sun JM, Ahn MJ, Ahn JS, Um SW, Kim H, Kim HK, et al. chemotherapy for pulmonary large cell neuroendocrine carcinoma: similar to that for small cell lung cancer or non-small cell lung cancer? Lung Cancer, 2012, 77(2):365–70., DOI 10. 1016/j.lungcan.2012.04.009 Niho S, Kenmotsu H, Sekine I, Ishii G, Ishikawa Y, Noguchi M, et al. Combination chemotherapy with irinotecan and cisplatin for large-cell neuroendocrine carcinoma of the lung: a multicenter phase II study. J Thorac Oncol, 2013, 8(7):980–4., DOI 10.1097/JTO.0b013e31828f6989 Treut J, Sault MC, Lena H, Souquet PJ, Vergnenegre A, Caer HL, et al. Multicentre phase II study of cisplatin-etoposide chemotherapy for advanced largecell neuroendocrine lung carcinoma: the GFPC 0302 study. Ann Oncol, 2013, 24(6): 1548–52., DOI 10.1093/annonc/mdt009 Filosso PL, Fontana EC, Ruffini E. Large-cell neuroendocrine carcinoma and combined large-cell neuroendocrine carcinoma: 2 characters in search of an author. Eur J Cardiothorac Surg, 2022, 62(2):ezac176., DOI 10.1093/ejcts/ezac176 ReckM,Rodriguez-Abreu D,RobinsonAG,Hui R,Csoszi T, FulopA, et al.Updated analysis of keynote-024: pembrolizumab versus platinum-based chemotherapy for advanced non-small-cell lung cancer with PD-L1 tumor proportion score of 50% or greater. J Clin Oncol, 2019, 37(7):537–46., DOI 10.1200/JCO.18.00149 Sabari JK, Julian RA, Ni A, Halpenny D, Hellmann MD, Drilon AE, et al. Outcomes of advanced pulmonary large cell neuroendocrine carcinoma stratified by RB1 loss, SLFN11 expression, and tumor mutational burden. JCO, 2018, 36: e20568., DOI 10.1200/jco.2018.36.15_suppl.e20568 Rekhtman N, Pietanza MC, Hellmann MD, Naidoo J, Arora A, Won H, et al. Next-generation sequencing of pulmonary large cell neuroendocrine carcinoma reveals small cell carcinoma–like and non–small cell carcinoma–like subsets. Clin Cancer Res, 2016, 22(14):3618–29., DOI 10.1158/1078-0432.ccr-15-2946 Xu J, Feng Q, Chen Y, Liu XL, Jiang O. Complete remission of combined pulmonary large cell neuroendocrine carcinoma: a case report. J Int Med Res, 2021, 49(11):3000605211055387., DOI 10.1177/03000605211055387 Lou GY, Yu XM, Song ZB. Molecular profiling and survival of completely resected primary pulmonary neuroendocrine carcinoma. Clin Lung Cancer, 2017, 18(3):197–201., DOI 10.1016/j.cllc.2016.11.014 Omachi N, Shimizu S, Kawaguchi T, Tezuka K, Kanazu M, Tamiya A, et al. A case of large-cell neuroendocrine carcinoma harboring an EML4-ALK rearrangement with resistance to the ALK inhibitor crizotinib. J Thorac Oncol, 2014, 9(6):40–2., DOI 10.1097/JTO.0000000000000103 Hayashi N, Fujita A, Saikai T, Takabatake H, Sotoshiro M, Sekine K, et al. Large cell neuroendocrine carcinoma harboring an anaplastic lymphoma kinase, ALK, rearrangement with response to alectinib. Intern Med, 2018, 57:713–6., DOI 10.2169/internalmedicine.9368-17 HotonD,HumbletY,Libbrecht L.Phenotypic variationof anALK-positive largecell neuroendocrine lung carcinomawith carcinoidmorphology during treatmentwith ALK inhibitors. Histopathology, 2018, 72(4):707–10., DOI 10.1111/his.13388 Liu DR, Li BC. Large cell neuroendocrine carcinoma of the right lung: A case report and literature review. Chin J Med Guide, 2020, 22(09):627–30. Huang WY, Xu X, Li SX. Pulmonary combined large cell neuroendocrine carcinoma: a case report and literature review [J]. J Clin Exp Pathol, 2014, 30(04): 458–60. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610747 EP 1610752 DI 10.3389/pore.2022.1610747 PG 6 ER PT J AU Wang, H Xie, H Wang, Sh Zhao, J Gao, Y Chen, J Zhao, Y Guo, G AF Wang, Hetong Xie, Haitao Wang, Shuying Zhao, Jiaying Gao, Ya Chen, Jun Zhao, Yuxia Guo, Genyan TI PARP-1 genetic polymorphism associated with radiation sensitivity of non-small cell lung cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE radiotherapy; polymorphism; non-small cell lung cancer (NSCLC); PARP-1; sensitivity ID radiotherapy; polymorphism; non-small cell lung cancer (NSCLC); PARP-1; sensitivity AB About 70% of non-small cell lung cancer (NSCLC) patients require radiotherapy. However, due to the difference in radiation sensitivity, the treatment outcome may differ for the same pathology and choice of treatment. Poly (ADP-ribose) polymerase 1 (PARP-1) is a key gene responsible for DNA repair and is involved in base excision repair as well as repair of single strand break induced by ionizing radiation and oxidative damage. In order to investigate the relationship between PARP-1 gene polymorphism and radiation sensitivity in NSCLC, we collected 141 primary NSCLC patients undergoing three-dimensional conformal radiotherapy. For each case, the gross tumor volumes (GTV) before radiation and that after 40 Gy radiation were measured to calculate the tumor regression rate. TaqMan real-time polymerase chain reaction was performed to genotype the single-nucleotide polymorphisms (SNPs). Genotype frequencies for PARP-1 genotypes were 14.2% for C/C, 44.7% for C/G and 41.1% for G/G. The average tumor regression rate after 40 Gy radiation therapy was 35.1% ± 0.192. Tumor regression rate of mid-term RT of C/C genotype was 44.6% ± 0.170, which was higher than that of genotype C/G and G/G (32.4% ± 0.196 and 34.8% ± 0.188, respectively) with statistical significance (F = 3.169 p = 0.045). The higher tumor regression rate in patients with C/C genotype suggested that G allele was a protective factor against radiation therapy. Using the median tumor regression rate of 34%, we divided the entire cohort into two groups, and found that the frequency distribution of PARP-1 gene rs3219073 had significant difference between these two groups (p < 0.05). These results showed that PARP-1 gene polymorphism may affect patient radiation sensitivity and predict the efficacy of radiotherapy. It therefore presents an opportunity for developing new therapeutic targets to improve radiotherapy outcome. C1 [Wang, Hetong] The Tenth People’s Hospital of Shenyang, Department of Radiation OncologyShenyang, China. [Xie, Haitao] Liaoning Cancer Hospital, Department of Radiation OncologyShenyang, China. [Wang, Shuying] Shandong Polytechnic CollegeJining, China. [Zhao, Jiaying] Qingdao United Family Healthcare, Department of Radiation OncologyQingdao, China. [Gao, Ya] Kailuan Hospital, Department of OncologyTangshan, Hebei, China. [Chen, Jun] The Tenth People’s Hospital of Shenyang, Department of Radiation OncologyShenyang, China. [Zhao, Yuxia] The Fourth Affiliated Hospital of China Medical University, Department of Radiation OncologyShenyang, China. [Guo, Genyan] The Fourth Affiliated Hospital of China Medical University, Department of Radiation OncologyShenyang, China. RP Guo, G (reprint author), The Fourth Affiliated Hospital of China Medical University, Department of Radiation Oncology, Shenyang, China. EM gyguo@cmu.edu.cn CR Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin, 2021, 71(1):7–33., DOI 10.3322/caac.21654 ChenW, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin, 2016, 66(2):115–32., DOI 10.3322/caac.21338 Rivera MP. Multimodality therapy in the treatment of lung cancer. Semin Respir Crit Care Med, 2004, 25:3–10., DOI 10.1055/s-2004-829639 Tyldesley S, Boyd C, Schulze K, Walker H, Mackillop WJ. Estimating the need for radiotherapy for lung cancer: An evidence-based, epidemiologic approach. Int J Radiat Oncol Biol Phys, 2001, 49(4):973–85., DOI 10.1016/s0360-3016(00)01401-2 Shimura T, Kakuda S, Ochiai Y, Nakagawa H, Kuwahara Y, Takai Y, et al. Acquired radioresistance of human tumor cells by DNA-pk/akt/gsk3beta-mediated cyclin D1 overexpression. Oncogene, 2010, 29(34):4826–37., DOI 10.1038/onc.2010.238 Peltonen JK, Vahakangas KH, Helppi HM, Bloigu R, Paakko P, Turpeenniemi- Hujanen T. Specific Tp53 mutations predict aggressive phenotype in head and neck squamous cell carcinoma: A retrospective archival study. Head Neck Oncol, 2011, 3: 20., DOI 10.1186/1758-3284-3-20 Parliament MB, Murray D. Single nucleotide polymorphisms of DNA repair genes as predictors of radioresponse. Semin Radiat Oncol, 2010, 20(4):232–40., DOI 10.1016/j.semradonc.2010.05.003 Karar J, Maity A. Modulating the tumor microenvironment to increase radiation responsiveness. Cancer Biol Ther, 2009, 8(21):1994–2001., DOI 10.4161/cbt.8.21.9988 Baumann M, Krause M, Hill R. Exploring the role of cancer stem cells in radioresistance. Nat Rev Cancer, 2008, 8(7):545–54., DOI 10.1038/nrc2419 Yanbaeva DG, Dentener MA, Creutzberg EC, Wesseling G, Wouters EFM. Systemic effects of smoking. Chest, 2007, 131(5):1557–66., DOI 10.1378/chest.06-2179 Lyakhovich VV, Vavilin VA, Zenkov NK, Menshchikova EB. Active defense under oxidative stress. The antioxidant responsive element. Biochemistry, 2006, 71(9):962–74., DOI 10.1134/S0006297906090033 Goto M, Ueda K, Hashimoto T, Fujiwara S, Matsuyama K, Kometani T, et al. A formation mechanism for 8-hydroxy-2 ’-Deoxyguanosine mediated by peroxidized 2 ’-Deoxythymidine. Free Radic Biol Med, 2008, 45(9):1318–25., DOI 10.1016/j.freeradbiomed.2008.08.003 van Helden YGJ, Keijer J, Heil SG, Pico C, Palou A, Oliver P, et al. Betacarotene affects oxidative stress-related DNA damage in lung epithelial cells and in ferret lung. Carcinogenesis, 2009, 30(12):2070–6., DOI 10.1093/carcin/bgp186 Morrison C, Smith GC, Stingl L, Jackson SP, Wagner EF, Wang ZQ. Genetic interaction between parp and DNA-pk in V(D)J recombination and tumorigenesis. Nat Genet, 1997, 17(4):479–82., DOI 10.1038/ng1297-479 Burkle A. Physiology and pathophysiology of poly(adp-Ribosyl)Ation. Bioessays, 2001, 23(9):795–806., DOI 10.1002/Bies.1115 Wang HT, Gao Y, Zhao YX, Yu H, Wang TL, Bai L, et al. Parp-1 Rs3219073 polymorphism may contribute to susceptibility to lung cancer. Genet Test Mol Biomarkers, 2014, 18(11):736–40., DOI 10.1089/gtmb.2014.0106 Fox J, Ford E, Redmond K, Zhou J,Wong J, Song DY. Quantification of tumor volume changes during radiotherapy for non-small-cell lung cancer. Int J Radiat Oncol Biol Phys, 2009, 74(2):341–8., DOI 10.1016/j.ijrobp.2008.07.063 Ramsey CR, Langen KM, Kupelian PA, Scaperoth DD, Meeks SL, Mahan SL, et al. A technique for adaptive image-guided helical tomotherapy for lung cancer. Int J Radiat Oncol Biol Phys, 2006, 64(4):1237–44., DOI 10.1016/j.ijrobp. 2005.11.012 Kupelian PA, Ramsey C, Meeks SL, Willoughby TR, Forbes A, Wagner TH, et al. Serial megavoltage ct imaging during external beam radiotherapy for nonsmall- cell lung cancer: Observations on tumor regression during treatment. Int J Radiat Oncol Biol Phys, 2005, 63(4):1024–8., DOI 10.1016/j.ijrobp.2005.04.046 Siker ML, Tome WA, Mehta MP. Tumor volume changes on serial imaging with megavoltage ct for non-small-cell lung cancer during intensity-modulated radiotherapy: How reliable, consistent, and meaningful is the effect? Int J Radiat Oncol Biol Phys, 2006, 66(1):135–41., DOI 10.1016/j.ijrobp.2006.03.064 Woodford C, Yartsev S, Dar AR, Bauman G, Van Dyk J. Adaptive radiotherapy planning on decreasing gross tumor volumes as seen on megavoltage computed tomography images. Int J Radiat Oncol Biol Phys, 2007, 69(4):1316–22., DOI 10.1016/j.ijrobp.2007.07.2369 Hecht SS. Cigarette smoking and lung cancer: Chemical mechanisms and approaches to prevention. Lancet Oncol, 2002, 3(8):461–9., DOI 10.1016/s1470- 2045(02)00815-x Bargagli E, Olivieri C, Bennett D, Prasse A, Muller-Quernheim J, Rottoli P. Oxidative stress in the pathogenesis of diffuse lung diseases: A review. Respir Med, 2009, 103(9):1245–56., DOI 10.1016/j.rmed.2009.04.014 Krishnakumar R, Kraus WL. The parp side of the nucleus: Molecular actions, physiological outcomes, and clinical targets. Mol Cel, 2010, 39(1):8–24., DOI 10. 1016/j.molcel.2010.06.017 Hegan DC, Lu Y, Stachelek GC, Crosby ME, Bindra RS, Glazer PM. Inhibition of poly(adp-ribose, polymerase down-regulates Brca1 and Rad51 in a pathway mediated by E2f4 and P130. Proc Natl Acad Sci U S A, 2010, 107(5):2201–6., DOI 10. 1073/pnas.0904783107 Masutani M, Nakagama H, Sugimura T. Poly(Adp-Ribose, and carcinogenesis. Genes Chromosomes Cancer, 2003, 38(4):339–48., DOI 10.1002/ gcc.10250 Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG. Parp inhibition: Parp1 and beyond. Nat Rev Cancer, 2010, 10(4):293–301., DOI 10.1038/nrc2812 Camero S, Ceccarelli S, De Felice F, Marampon F, Mannarino O, Camicia L, et al. Parp inhibitors affect growth, survival and radiation susceptibility of human alveolar and embryonal rhabdomyosarcoma cell lines. J Cancer Res Clin Oncol, 2019, 145(1):137–52., DOI 10.1007/s00432-018-2774-6 Chalmers AJ, Lakshman M, Chan N, Bristow RG. Poly(Adp-Ribose, polymerase inhibition as a model for synthetic lethality in developing radiation oncology targets. Semin Radiat Oncol, 2010, 20(4):274–81., DOI 10.1016/j. semradonc.2010.06.001 Wahner Hendrickson AE, Menefee ME, Hartmann LC, Long HJ, Northfelt DW, Reid JM, et al. A phase I clinical trial of the poly(adp-ribose, polymerase inhibitor veliparib and weekly topotecan in patients with solid tumors. Clin Cancer Res, 2018, 24(4):744–52., DOI 10.1158/1078-0432.CCR-17-1590 Kummar S, Kinders R, Gutierrez ME, Rubinstein L, Parchment RE, Phillips LR, et al. Phase 0 clinical trial of the poly, Adp-Ribose, polymerase inhibitor abt- 888 in patients with advanced malignancies. J Clin Oncol, 2009, 27(16):2705–11., DOI 10.1200/JCO.2008.19.7681 Gadducci A, Cosio S. Randomized clinical trials and real world prospective observational studies on bevacizumab, parp inhibitors, and immune checkpoint inhibitors in the first-line treatment of advanced ovarian carcinoma: A critical review. Anticancer Res, 2021, 41(10):4673–85., DOI 10.21873/anticanres.15281 Fong PC, Yap TA, Boss DS, Carden CP, Mergui-Roelvink M, Gourley C, et al. Poly(Adp)-Ribose polymerase inhibition: Frequent durable responses in brca carrier ovarian cancer correlating with platinum-free interval. J Clin Oncol, 2010, 28(15):2512–9., DOI 10.1200/JCO.2009.26.9589 Tutt A, Robson M, Garber JE, Domchek SM, Audeh MW, Weitzel JN, et al. Oral poly(adp-ribose, polymerase inhibitor olaparib in patients with Brca1 or Brca2 mutations and advanced breast cancer: A proof-of-concept trial. Lancet, London, England,, 2010, 376(9737):235–44., DOI 10.1016/S0140-6736(10)60892-6 Inbar D, Cohen-Armon M, Neumann D. Erythropoietin-driven signalling and cell migration mediated by polyadp-ribosylation. Br J Cancer, 2012, 107(8): 1317–26., DOI 10.1038/bjc.2012.395 Meyn RE, Munshi A, Haymach JV, Milas L, Ang KK. Receptor signaling as a regulatory mechanism of DNA repair. Radiother Oncol, 2009, 92(3):316–22., DOI 10. 1016/j.radonc.2009.06.031 Barreto-Andrade JC, Efimova EV, Mauceri HJ, Beckett MA, Sutton HG, Darga TE, et al. Response of human prostate cancer cells and tumors to combining parp inhibition with ionizing radiation. Mol Cancer Ther, 2011, 10(7):1185–93., DOI 10.1158/1535-7163.MCT-11-0061 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610751 EP 1610757 DI 10.3389/pore.2022.1610751 PG 7 ER PT J AU AlQudah, M Khalifeh, M Al-Azaizeh, R Masaadeh, A Al-Rusan, O Haddad, H AF AlQudah, Mohammad Khalifeh, Mohammad Al-Azaizeh, Rasha Masaadeh, Amr Al-Rusan, M. Omar Haddad, K. Husam TI Hyperbaric oxygen exposure alleviate metabolic side-effects of olanzapine treatment and is associated with Langerhans islet proliferation in rats SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE olanzapine; hyperbaric oxygen therapy; pancreatic Langerhans cells; metabolic disorders; insulin ID olanzapine; hyperbaric oxygen therapy; pancreatic Langerhans cells; metabolic disorders; insulin AB Introduction: Olanzapine (OLZ) is one of the second-generation antipsychotics drugs (APDs) used to treat several psychiatric illnesses. Olanzapine treatment is often associated with many metabolic side effects in a dose dependent manner such as obesity, dyslipidemia and insulin resistance, induction of type II diabetes and acute pancreatitis in some patients. Methods: Hyperbaric Oxygen therapy (HBOT) was investigated as a tool to mitigate olanzapine metabolic side effects in rats. Thirty-six female Sprague Dawley (SD) rats were divided into 4 groups; rats on olanzapine treatment either exposed to hyperbaric oxygen therapy (HBOOLZ) or left without exposure (OLZ) then non-treated rats that either exposed to hyperbaric oxygen therapy or left without exposure (control). Rats received Hyperbaric Oxygen therapy for 35 days at 2.4 atmospheres absolute (ATA) for 2.5 h daily followed by intraperitoneal injection of olanzapine at 10mg/kg or placebo. Results: Rats on either hyperbaric oxygen therapy or olanzapine had a significant loss in body weight. Olanzapine treatment showed a decrease in serum insulin level, triglyceride, highdensity lipoprotein (HDL) cholesterol, and lipase level but an increase in fasting blood sugar (FBS), insulin resistance index (HOMA-IR) and amylase, while rats’ exposure to hyperbaric oxygen therapy reversed these effects. The Pancreatic Langerhans islets were up-regulated in both hyperbaric oxygen therapy and olanzapine treatments but the combination (HBOOLZ) doubled these islets number. Discussion: This study advocated that hyperbaric oxygen therapy can be an alternative approach to control or reverse many metabolic disorders (MDs) associatedwith olanzapine treatment. In addition, it seems that hyperbaric oxygen therapy positively affect the pancreatic Langerhans cells activity and architecture. C1 [AlQudah, Mohammad] Jordan University of Science and Technology, Faculty of Medicine, Department of Pathology and MicrobiologyIrbid, Jordan. [Khalifeh, Mohammad] Jordan University of Science and Technology, Department of Veterinary Basic SciencesIrbid, Jordan. [Al-Azaizeh, Rasha] Jordan University of Science and Technology, Department of Veterinary Basic SciencesIrbid, Jordan. [Masaadeh, Amr] Jordan University of Science and Technology, Faculty of Medicine, Department of Pathology and MicrobiologyIrbid, Jordan. [Al-Rusan, M. Omar] Jordan University of Science and Technology, Faculty of Medicine, Department of Pathology and MicrobiologyIrbid, Jordan. [Haddad, K. Husam] Ministry of Health, Department of Pathology and Laboratory MedicineAmman, Jordan. RP AlQudah, M (reprint author), Jordan University of Science and Technology, Faculty of Medicine, Department of Pathology and Microbiology, Irbid, Jordan. EM m.alqudah12@just.edu.jo CR Thomas K, Saadabadi A. Olanzapine in: StatPearls, 2018). Treasure Island, FL: StatPearls Publishing. Citrome L, McEvoy JP, Todtenkopf MS, McDonnell D, Weiden PJ. A commentary on the efficacy of olanzapine for the treatment of schizophrenia: The past, present, and future. Neuropsychiatr Dis Treat, 2019, 15:2559–69., DOI 10.2147/NDT.S209284 Gautam S, Meena PS. Drug-emergent metabolic syndrome in patients with schizophrenia receiving atypical, second-generation, antipsychotics. Indian J Psychiatry, 2011, 53(2):128–33., DOI 10.4103/0019-5545.82537 Shirzadi AA, Ghaemi SN. Side effects of atypical antipsychotics: extrapyramidal symptoms and the metabolic syndrome. Harv Rev Psychiatry, 2006, 14(3):152–64., DOI 10.1080/10673220600748486 Tandon R. Safety and tolerability: how do newer generation “atypical” antipsychotics compare? Psychiatr Q, 2002, 73(4):297–311., DOI 10.1023/a: 1020464017021 Allison DB, Mentore JL, Heo M, Chandler LP, Cappelleri JC, Infante MC, et al. Antipsychotic-induced weight gain: A comprehensive research synthesis. Am J Psychiatry, 1999, 156(11):1686–96., DOI 10.1176/ajp.156.11. 1686 Ganguli R. Weight gain associated with antipsychotic drugs. J Clin Psychiatry, 1999, 60:20–4. Tadger S, Melamed Y. Weight gain due to long term antipsychotic treatment of persistent mental disorders. Psychiatr Danub, 2008, 20(1):37–41. Birkenaes AB, Birkeland KI, Engh JA, Faerden A, Jonsdottir H, Ringen PA, et al. Dyslipidemia independent of body mass in antipsychotic-treated patients under real-life conditions. J Clin Psychopharmacol, 2008, 28(2):132–7., DOI 10.1097/ JCP.0b013e318166c4f7 Cope M, Nagy TR, Fernandez JR, Geary N, Casey DE, Allison DB. Antipsychotic drug-induced weight gain: development of an animal model. Int J Obes, 2005, 29(6):607–14., DOI 10.1038/sj.ijo.0802928 Shah R, Subhan F, Ali G, Ullah I, Ullah S, Shahid M, et al. Olanzapine induced biochemical and histopathological changes after its chronic administration in rats. Saudi Pharm J, 2016, 24(6):698–704., DOI 10.1016/j.jsps.2015.06.006 Haupt DW, Newcomer JW. Hyperglycemia and antipsychotic medications. J Clin Psychiatry, 2001, 62(27):15–26; discussion 40-1. Ferno J, Ersland KM, Duus IH, Gonzalez-Garcia I, Fossan KO, Berge RK, et al. Olanzapine depot exposure in male rats: dose-dependent lipogenic effects without concomitant weight gain. Eur Neuropsychopharmacol, 2015, 25(6):923–32., DOI 10. 1016/j.euroneuro.2015.03.002 van der Zwaal EM, Luijendijk MCM, Evers SS, la Fleur SE, Adan RAH. Olanzapine affects locomotor activity and meal size in male rats. Pharmacol Biochem Behav, 2010, 97(1):130–7., DOI 10.1016/j.pbb.2010.05.009 Kinon BJ, Kaiser CJ, Ahmed S, Rotelli MD, Kollack-Walker S. Association between early and rapid weight gain and change in weight over one year of olanzapine therapy in patients with schizophrenia and related disorders. J Clin Psychopharmacol, 2005, 25(3):255–8., DOI 10.1097/01.jcp.0000161501.65890.22 Basson BR, Kinon BJ, Taylor CC, Szymanski KA, Gilmore JA, Tollefson GD. Factors influencing acute weight change in patients with schizophrenia treated with olanzapine, haloperidol, or risperidone. J Clin Psychiatry, 2001, 62(4):231–8., DOI 10.4088/jcp.v62n0404 Zhang Z-J, Yao ZJ, LiuW, Fang Q, Reynolds GP. Effects of antipsychotics on fat deposition and changes in leptin and insulin levels: magnetic resonance imaging study of previously untreated people with schizophrenia. Br J Psychiatry, 2004, 184(1):58–62., DOI 10.1192/bjp.184.1.58 Deng C. Effects of antipsychotic medications on appetite, weight, and insulin resistance. Endocrinol Metab Clin North Am, 2013, 42(3):545–63., DOI 10.1016/j.ecl. 2013.05.006 Martins PJ, Haas M, Obici S. Central nervous system delivery of the antipsychotic olanzapine induces hepatic insulin resistance. Diabetes, 2010, 59(10):2418–25., DOI 10.2337/db10-0449 Ahuja N, PalaNichamyN, Mackin P, Lloyd A. Olanzapine-induced hyperglycaemic coma and neuroleptic malignant syndrome: case report and review of literature. J Psychopharmacol, 2010, 24(1):125–30., DOI 10.1177/0269881108096901 Kohen I, Gampel M, Reddy L, Manu P. Rapidly developing hyperglycemia during treatment with olanzapine. Ann Pharmacother, 2008, 42(4):588–91., DOI 10. 1345/aph.1K607 Koller EA, Cross JT, Doraiswamy PM, Malozowski SN. Pancreatitis associated with atypical antipsychotics: from the food and drug administration’s MedWatch surveillance system and published reports. Pharmacotherapy, 2003, 23(9):1123–30., DOI 10.1592/phco.23.10.1123.32759 Waage C, Carlsson H, Nielsen EW. Olanzapine-induced pancreatitis: A case report. JOP, 2004, 5(5):388–91. Wirshing DA, Spellberg BJ, Erhart SM, Marder SR, Wirshing WC. Novel antipsychotics and new onset diabetes. Biol Psychiatry, 1998, 44(8):778–83., DOI 10. 1016/s0006-3223(98)00100-0 Kerr TA, Jonnalagadda S, Prakash C, Azar R. Pancreatitis following olanzapine therapy: A report of three cases. Case Rep Gastroenterol, 2007, 1(1): 15–20., DOI 10.1159/000104222 Ustohal L, Mayerova M, Valkova B, Sedlakova H, Kasparek T. Asymptomatic elevation of amylase and lipase after olanzapine treatment. J Clin Psychopharmacol, 2016, 36(2):181–3., DOI 10.1097/JCP.0000000000000460 Remington GJ, Teo C, Wilson V, Chintoh A, Guenette M, Ahsan Z, et al. Metformin attenuates olanzapine-induced hepatic, but not peripheral insulin resistance. J Endocrinol, 2015, 227(2):71–81., DOI 10.1530/JOE-15-0074 Venkatasubramanian G, Arasappa R, Rao NP, Behere RV, Gangadhar BN. Adjuvant metformin worsens psychosis in schizophrenia: A case report. Prim Care Companion J Clin Psychiatry, 2010, 12(2):PCC.09l00801., DOI 10.4088/PCC. 09l00801yel Boyda HN, Procyshyn RM, Tse L, Hawkes E, Jin CH, Pang CCY, et al. Differential effects of 3 classes of antidiabetic drugs on olanzapine-induced glucose dysregulation and insulin resistance in female rats. J Psychiatry Neurosci, 2012, 37(6):407–15., DOI 10.1503/jpn.110140 Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M, et al. PPARγ signaling and metabolism: The good, the bad and the future. Nat Med, 2013, 19(5): 557–66., DOI 10.1038/nm.3159 Castellani LN, Peppler WT,Miotto PM, Bush N,Wright DC. Exercise protects against olanzapine-induced hyperglycemia in male C57BL/6J mice. Sci Rep, 2018, 8(1):772–11., DOI 10.1038/s41598-018-19260-x Brown S, Birtwistle J, Roe L, Thompson C. The unhealthy lifestyle of people with schizophrenia. Psychol Med, 1999, 29(3):697–701., DOI 10.1017/s0033291798008186 Lieberman JA, Stroup TS, McEvoy JP, Swartz MS, Rosenheck RA, Perkins DO, et al. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med, 2005, 353(12):1209–23., DOI 10.1056/NEJMoa051688 Semenza GL. Oxygen-dependent regulation of mitochondrial respiration by hypoxia-inducible factor 1. Biochem J, 2007, 405(1):1–9., DOI 10.1042/BJ20070389 Wilson DF, Erecinska M, Drown C, Silver IA. The oxygen dependence of cellular energy metabolism. Arch Biochem Biophys, 1979, 195(2):485–93., DOI 10. 1016/0003-9861(79)90375-8 FaleoG, FotinoC, BoccaN,MolanoRD, Zahr-Akrawi E,Molina J, et al. Prevention of autoimmune diabetes and induction of β-cell proliferation in NOD mice by hyperbaric oxygen therapy. Diabetes, 2012, 61(7):1769–78., DOI 10.2337/db11-0516 Zakiyah A, Anggraini VS. Hyperbaric oxygen therapy to reduce blood glucose level on patients diabetes mellitus. J Nurs Care, 2019, 2(1)., DOI 10.24198/jnc.v2i1.19306 Grajales D, Vazquez P, Ruiz-RosarioM, Tuduri E,MirasierraM, Ferreira V, et al. The second-generation antipsychotic drug aripiprazole modulates the serotonergic system in pancreatic islets and induces beta cell dysfunction in female mice. Diabetologia, 2022, 65(3):490–505., DOI 10.1007/s00125-021- 05630-0 Huang J, Hei GR, Yang Y, Liu CC, Xiao JM, Long YJ, et al. Increased appetite plays a key role in olanzapine-induced weight gain in first-episode schizophrenia patients. Front Pharmacol, 2020, 11:739., DOI 10.3389/fphar. 2020.00739 National Research Council, US). Guide for the care and use of laboratory animals. Washington, DC, 1996). Mann S, Chintoh A, Giacca A, Fletcher P, Nobrega J, Hahn M, et al. Chronic olanzapine administration in rats: effect of route of administration on weight, food intake and body composition. Pharmacol Biochem Behav, 2013, 103(4):717–22., DOI 10.1016/j.pbb.2012.12.002 Davey KJ, O’Mahony SM, Schellekens H, O’Sullivan O, Bienenstock J, Cotter PD, et al. Gender-dependent consequences of chronic olanzapine in the rat: effects on body weight, inflammatory, metabolic and microbiota parameters. Psychopharmacology, 2012, 221(1):155–69., DOI 10.1007/s00213-011-2555-2 Pouzet B, Mow T, KreilgaardM,VelSchow S. Chronic treatment with antipsychotics in rats as a model for antipsychotic-induced weight gain in human. Pharmacol Biochem Behav, 2003, 75(1):133–40., DOI 10.1016/s0091-3057(03)00042-x Alazaizeh RA. Improve health parameters and reduce in body weight by oxygen therapy, exercises while breathing). In: Applied biology. Jordan: Jordan University of Science and Technology, 2020). p. 133. Nakamura M, Nagamine T. Severe hyperglycemia induced by olanzapine was improved with a recovery of insulin secretion after switching to risperidone and introducing insulin therapy. Intern Med, 2010, 49(23):2635–7., DOI 10.2169/ internalmedicine.49.4397 Nakamura M, Masaoka Y, Nagamine T. Olanzapine-induced severe hyperglycemia was completely reversed by the restoration of insulin secretion after switching to aripiprazole and initiating insulin therapy. Clin Neuropsychopharmacol Ther, 2014, 5:29–33., DOI 10.5234/cnpt.5.29 Doucette DE, Grenier JS, Robertson PS. Olanzapine-induced acute pancreatitis. Ann Pharmacother, 2000, 34(10):1128–31., DOI 10.1345/aph.19390 Hou P-H, Chang GR, Chen CP, Lin YL, Chao IS, Shen TT, et al. Long-term administration of olanzapine induces adiposity and increases hepatic fatty acid desaturation protein in female C57BL/6J mice. Iran J Basic Med Sci, 2018, 21(5): 495–501., DOI 10.22038/IJBMS.2018.22759.5780 Mir S, Taylor D. Atypical antipsychotics and hyperglycaemia. Int Clin Psychopharmacol, 2001, 16(2):63–73., DOI 10.1097/00004850-200103000-00001 Nagamine T. Hypoglycemia associated with insulin hypersecretion following the addition of olanzapine to conventional antipsychotics. Neuropsychiatr Dis Treat, 2006, 2(4):583–5., DOI 10.2147/nedt.2006.2.4.583 Wilkinson D, Chapman I, Heilbronn L. Hyperbaric oxygen therapy improves peripheral insulin sensitivity in humans. Diabet Med, 2012, 29(8):986–9., DOI 10. 1111/j.1464-5491.2012.03587.x Cruz-Villanueva SR, Ramirez-Nava JC, Moreno-Luna JA, Cardenas-Urena KG, Espin-Iturbe LT, SanchezOtero MG, et al. Effect of hyperbaric oxygen therapy, HBOT, on insulin resistance associated with abdominal obesity in wistar rats with dietary sucroseinduced metabolic syndrome. JNutr Sci Vitaminol, 2021, 67(5):292–300., DOI 10.3177/jnsv. 67.292 Finkel S. Pharmacology of antipsychotics in the elderly: A focus on atypicals. J Am Geriatr Soc, 2004, 52:S258–65., DOI 10.1111/j.1532-5415.2004.52602.x Thomas J, Linssen M, van der Vusse GJ, Hirsch B, Rosen P, Kammermeier H, et al. Acute stimulation of glucose transport by histamine in cardiac microvascular endothelial cells. Biochim Biophys Acta, 1995, 1268(1):88–96., DOI 10.1016/0167- 4889(95)00049-x Shi T, Papay RS, Perez DM. The role of α1-adrenergic receptors in regulating metabolism: increased glucose tolerance, leptin secretion and lipid oxidation. J Recept Signal Transduct Res, 2017, 37(2):124–32., DOI 10.1080/10799893.2016.1193522 Wei H, Zapata RC, Lopez-ValenciaM, Aslanoglou D, Farino ZJ, Benner V, et al. Dopamine D2 receptor signaling modulates pancreatic beta cell circadian rhythms. Psychoneuroendocrinology, 2020, 113:104551., DOI 10.1016/j.psyneuen.2019.104551 Farino ZJ, Morgenstern TJ, Maffei A, Quick M, De Solis AJ, Wiriyasermkul P, et al. New roles for dopamine D 2 and D 3 receptors in pancreatic beta cell insulin secretion. Mol Psychiatry, 2020, 25(9):2070–85., DOI 10.1038/s41380-018-0344-6 Ballon JS, Pajvani UB, Mayer LE, Freyberg Z, Freyberg R, Contreras I, et al. Pathophysiology of drug induced weight and metabolic effects: findings from an RCT in healthy volunteers treated with olanzapine, iloperidone, or placebo. J Psychopharmacol, 2018, 32(5):533–40., DOI 10.1177/0269881118754708 Weston-Green K, Huang X-F, Deng C. Second generation antipsychoticinduced type 2 diabetes: A role for the muscarinic M3 receptor. CNS drugs, 2013, 27(12):1069–80., DOI 10.1007/s40263-013-0115-5 Weston-Green K, Huang XF, Lian J, Deng C. Effects of olanzapine on muscarinic M3 receptor binding density in the brain relates to weight gain, plasma insulin and metabolic hormone levels. Eur Neuropsychopharmacol, 2012, 22(5):364–73., DOI 10.1016/j.euroneuro.2011.09.003 Qiu L, Yin G, Cheng L, Fan Y, Xiao W, Yu G, et al. Astragaloside IV ameliorates acute pancreatitis in rats by inhibiting the activation of nuclear factor- κB. Int J Mol Med, 2015, 35(3):625–36., DOI 10.3892/ijmm.2015.2070 Naxakis S, Wafer M, Collins R. Olanzapine-induced acute necrotising pancreatitis leading to recurrent multiple organ dysfunction syndrome. Gen Psychiatr, 2022, 35(1):e100687., DOI 10.1136/gpsych-2021-100687 Cuthbertson CM, Christophi C. Potential effects of hyperbaric oxygen therapy in acute pancreatitis. ANZ J Surg, 2006, 76(7):625–30., DOI 10.1111/j.1445-2197. 2006.03793.x Christophi C, Millar I, Nikfarjam M, Muralidharan V,Malcontenti-Wilson C. Hyperbaric oxygen therapy for severe acute pancreatitis. J Gastroenterol Hepatol, 2007, 22(11):2042–6., DOI 10.1111/j.1440-1746.2006.03380.x Zhao H, Ge B, Yuan Y, Wang G. Hyperbaric oxygen ameliorated acute pancreatitis in rats via the mitochondrial pathway. Dig Dis Sci, 2020, 65(12): 3558–69., DOI 10.1007/s10620-020-06070-3 Dauwan M, Begemann MJH, Heringa SM, Sommer IE. Exercise improves clinical symptoms, quality of life, global functioning, and depression in schizophrenia: A systematic review and meta-analysis. Schizophr Bull, 2016, 42(3):588–99., DOI 10.1093/schbul/sbv164 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610752 EP 1610762 DI 10.3389/pore.2022.1610752 PG 11 ER PT J AU Shen, X Mo, X Tan, W Mo, X Li, L Yu, F He, J Deng, Z Xing, Sh Chen, Z Yang, J AF Shen, Xiaoju Mo, Xiaocheng Tan, Weidan Mo, Xiaoxiang Li, Li Yu, Fei He, Jingchuan Deng, Zhihua Xing, Shangping Chen, Zhiquan Yang, Jie TI KIAA1199 Correlates With Tumor Microenvironment and Immune Infiltration in Lung Adenocarcinoma as a Potential Prognostic Biomarker SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; prognosis; lung adenocarcinoma; immune infiltration; KIAA1199 ID biomarker; prognosis; lung adenocarcinoma; immune infiltration; KIAA1199 AB Background: KIAA1199 has been considered a key regulator of carcinogenesis. However, the relationship between KIAA1199 and immune infiltrates, as well as its prognostic value in lung adenocarcinoma (LUAD) remains unclear. Methods: The expression of KIAA1199 and its influence on tumor prognosis were analyzed using a series of databases, comprising TIMER, GEPIA, UALCAN, LCE, Prognoscan and Kaplan-Meier Plotter. Further, immunohistochemistry (IHC), western blot (WB) and receiver operating characteristic (ROC) curve analyses were performed to verify our findings. The cBioPortal was used to investigate the genomic alterations of KIAA1199. Prediction of candidate microRNA (miRNAs) and transcription factor (TF) targeting KIAA1199, as well as GO and KEGG analyses, were performed based on LinkedOmics. TIMER and TISIDB databases were used to explore the relationship between KIAA1199 and tumor immune infiltration. Results: High expression of KIAA1199 was identified in LUAD and Lung squamous cell carcinoma (LUSC) patients. High expression of KIAA1199 indicated a worse prognosis in LUAD patients. The results of IHC and WB analyses showed that the expression level of KIAA1199 in tumor tissues was higher than that in adjacent tissues. GO and KEGG analyses indicated KIAA1199 was mainly involved in extracellular matrix (ECM)-receptor interaction and extracellular matrix structure constituent. KIAA1199 was positively correlated with infiltrating levels of CD4+ T cells, macrophages, neutrophil cells, dendritic cells, and showed positive relationship with immune marker subsets expression of a variety of immunosuppressive cells. Conclusion: High expression of KIAA1199 predicts a poor prognosis of LUAD patients. KIAA1199 might exert its carcinogenic role in the tumor microenvironment via participating in the extracellular matrix formation and regulating the infiltration of immune cells in LUAD. The results indicate that KIAA1199 might be a novel biomarker for evaluating prognosis and immune cell infiltration in LUAD. C1 [Shen, Xiaoju] Guangxi Medical University, School of Pharmacy, Department of PharmacologyNanning, China. [Mo, Xiaocheng] Guangxi Medical University, School of Pharmacy, Department of PharmacologyNanning, China. [Tan, Weidan] Guangxi Medical University, School of Pharmacy, Department of PharmacologyNanning, China. [Mo, Xiaoxiang] Guangxi Medical University, School of Pharmacy, Department of PharmacologyNanning, China. [Li, Li] Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Department of PharmacologyNanning, China. [Yu, Fei] Guangxi Medical University, School of Pharmacy, Department of PharmacologyNanning, China. [He, Jingchuan] Guangxi Medical University, School of Pharmacy, Department of PharmacologyNanning, China. [Deng, Zhihua] Guangxi Medical University, School of Pharmacy, Department of PharmacologyNanning, China. [Xing, Shangping] Guangxi Medical University, School of Pharmacy, Guangxi Key Laboratory of Bioactive Molecules Research and EvaluationNanning, China. [Chen, Zhiquan] Guangxi Medical University, School of Pharmacy, Department of PharmacologyNanning, China. [Yang, Jie] Guangxi Medical University, School of Pharmacy, Department of PharmacologyNanning, China. RP Yang, J (reprint author), Guangxi Medical University, School of Pharmacy, Department of Pharmacology, Nanning, China. EM jieyang2016@gxmu.edu.cn CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics. CA Cancer J Clin, 2021, 71(1):7–33., DOI 10.3322/caac.21654 Herbst RS, Morgensztern D, and Boshoff C. The Biology and Management of Non-small Cell Lung Cancer. Nature, 2018, 553(7689):446–54., DOI 10.1038/ nature25183 Peinado-Serrano J, and Carnero A. Molecular Radiobiology in Non-small Cell Lung Cancer: Prognostic and Predictive Response Factors. Cancers, Basel,, 2022, 14(9):2202., DOI 10.3390/cancers14092202 Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ, Jr, Wu YL, et al. Lung Cancer: Current Therapies and New Targeted Treatments. Lancet, 2017, 389(10066):299–311., DOI 10.1016/s0140-6736(16)30958-8 Wang M, Herbst RS, and Boshoff C. Toward Personalized Treatment Approaches for Non-small-cell Lung Cancer. Nat Med, 2021, 27(8): 1345–56., DOI 10.1038/s41591-021-01450-2 Doroshow DB, Sanmamed MF, Hastings K, Politi K, Rimm DL, Chen L, et al. Immunotherapy in Non-small Cell Lung Cancer: Facts and Hopes. Clin Cancer Res, 2019, 25(15):4592–602., DOI 10.1158/1078-0432.ccr-18-1538 Schoenfeld AJ, Antonia SJ, Awad MM, Felip E, Gainor J, Gettinger SN, et al. Clinical Definition of Acquired Resistance to Immunotherapy in Patients with Metastatic Non-small-cell Lung Cancer. Ann Oncol, 2021, 32(12):1597–607., DOI 10.1016/j.annonc.2021.08.2151 Lim Zf, and Ma PC. Emerging Insights of Tumor Heterogeneity and Drug Resistance Mechanisms in Lung Cancer Targeted Therapy. J Hematol Oncol, 2019, 12(1):134., DOI 10.1186/s13045-019-0818-2 Xu JY, Zhang C, Wang X, Zhai L, Ma Y, Mao Y, et al. Integrative Proteomic Characterization of Human Lung Adenocarcinoma. Cell, 2020, 182(1): 245–61., DOI 10.1016/j.cell.2020.05.043 Terlizzi M, Colarusso C, Pinto A, and Sorrentino R. Drug Resistance in Nonsmall Cell Lung Cancer, NSCLC): Impact of Genetic and Non-genetic Alterations on Therapeutic Regimen and Responsiveness. Pharmacol Ther, 2019, 202:140–8., DOI 10.1016/j.pharmthera.2019.06.005 Zhang W, Yin G, Zhao H, Ling H, Xie Z, Xiao C, et al. Secreted KIAA1199 Promotes the Progression of Rheumatoid Arthritis by Mediating Hyaluronic Acid Degradation in an ANXA1-dependent Manner. Cell Death Dis, 2021, 12(1):102., DOI 10.1038/s41419-021-03393-5 Abe S, Usami S, and Nakamura Y. Mutations in the Gene Encoding KIAA1199 Protein, an Inner-Ear Protein Expressed in Deiters’ Cells and the Fibrocytes, as the Cause of Nonsyndromic Hearing Loss. J Hum Genet, 2003, 48(11):564–70., DOI 10.1007/s10038-003-0079-2 Mi C, Zhang D, Li Y, Ren M, Ma W, Lu G, et al. miR-4677-3p Participates Proliferation and Metastases of Gastric Cancer Cell via CEMIP-Pi3k/AKT Signaling Pathway. Cell Cycle, 2021, 20(19):1978–87., DOI 10.1080/15384101. 2021.1971375 Xie G, Dong P, Chen H, Xu L, Liu Y, Ma Y, et al. Decreased Expression of ATF3, Orchestrated by β-catenin/TCF3, miR-17-5p and HOXA11-AS, Promoted Gastric Cancer Progression via Increased β-catenin and CEMIP. Exp Mol Med, 2021, 53(11):1706–22., DOI 10.1038/s12276-021-00694-9 Song M, Liu J, Zheng X, Zhou X, Feng Z, and Hu W. MiR-148a-3p Targets CEMIP to Suppress the Genesis of Gastric Cancer Cells. Biochem Biophys Res Commun, 2021, 575:42–9., DOI 10.1016/j.bbrc.2021.08.039 Oba T, Sato N, Adachi Y, Amaike T, Kudo Y, Koga A, et al. Hypoxia Increases KIAA1199/CEMIP Expression and Enhances Cell Migration in Pancreatic Cancer. Sci Rep, 2021, 11(1):18193., DOI 10.1038/s41598-021-97752-z Zhang D, Zhao L, Shen Q, LvQ, Jin M, Ma H, et al. Down-regulation of KIAA1199/ CEMIP by miR-216a Suppresses Tumor Invasion and Metastasis in Colorectal Cancer. Int J Cancer, 2017, 140(10):2298–309., DOI 10.1002/ijc.30656 Hua Q, Zhang B, Xu G,Wang L, Wang H, Lin Z, et al. CEMIP, a Novel Adaptor Protein of OGT, Promotes Colorectal Cancer Metastasis through Glutamine Metabolic Reprogramming via Reciprocal Regulation of β-catenin. Oncogene, 2021, 40(46):6443–55., DOI 10.1038/s41388-021-02023-w Zhang P, Song Y, Sun Y, Li X, Chen L, Yang L, et al. AMPK/GSK3β/β-catenin cascade-triggered Overexpression of CEMIP Promotes Migration and Invasion in Anoikis-Resistant Prostate Cancer Cells by Enhancing Metabolic Reprogramming. Faseb j, 2018, 32(7):3924–35., DOI 10.1096/fj. 201701078R Yu Y, Liu B, Li X, Lu D, Yang L, Chen L, et al. ATF4/CEMIP/PKCα Promotes Anoikis Resistance by Enhancing Protective Autophagy in Prostate Cancer Cells. Cel Death Dis, 2022, 13(1):46., DOI 10.1038/s41419-021-04494-x Xu Y, Xu H, Li M, Wu H, Guo Y, Chen J, et al. KIAA1199 Promotes Sorafenib Tolerance and the Metastasis of Hepatocellular Carcinoma by Activating the EGF/EGFR-dependent Epithelial-Mesenchymal Transition Program. Cancer Lett, 2019, 454:78–89., DOI 10.1016/j.canlet.2019.03.049 Liu B, Li X, Wang D, Yu Y, Lu D, Chen L, et al. CEMIP Promotes Extracellular Matrix-Detached Prostate Cancer Cell Survival by Inhibiting Ferroptosis. Cancer Sci, 2022, 113(6):2056–70., DOI 10.1111/cas.15356 Rodrigues G, Hoshino A, Kenific CM, Matei IR, Steiner L, Freitas D, et al. Tumour Exosomal CEMIP Protein Promotes Cancer Cell Colonization in Brain Metastasis. Nat Cel Biol, 2019, 21(11):1403–12., DOI 10.1038/s41556- 019-0404-4 Wang H, Zhang B, Li R, Chen J, Xu G, Zhu Y, et al. KIAA1199 Drives Immune Suppression to Promote Colorectal Cancer Liver Metastasis by Modulating Neutrophil Infiltration. Hepatology, 2022, 76:967–81., DOI 10. 1002/hep.32383 Tang Z, Ding Y, Shen Q, Zhang C, Li J, Nazar M, et al. KIAA1199 Promotes Invasion and Migration in Non-small-cell Lung Cancer, NSCLC, via PI3KAkt Mediated EMT. J Mol Med, Berl,, 2019, 97(1):127–40., DOI 10.1007/ s00109-018-1721-y Wang A, Zhu J, Li J, Du W, Zhang Y, Cai T, et al. Downregulation of KIAA1199 by miR-486-5p Suppresses Tumorigenesis in Lung Cancer. Cancer Med, 2020, 9(15):5570–86., DOI 10.1002/cam4.3210 Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. Cancer Res, 2017, 77(21):e108–10., DOI 10.1158/0008-5472.CAN-17-0307 Li C, Tang Z, Zhang W, Ye Z, and Liu F. GEPIA2021: Integrating Multiple Deconvolution-Based Analysis into GEPIA. Nucleic Acids Res, 2021, 49(W1): W242–w246., DOI 10.1093/nar/gkab418 Chand rashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce- Rodriguez I, Chakravarthi B, et al. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses. Neoplasia, 2017, 19(8): 649–58., DOI 10.1016/j.neo.2017.05.002 Lanczky A, and Gyorffy B, Gyorffy B. Web-Based Survival Analysis Tool Tailored for Medical Research, KMplot): Development and Implementation. J Med Internet Res, 2021, 23(7):e27633., DOI 10.2196/27633 Mizuno H, Kitada K, Nakai K, and Sarai A. PrognoScan: a New Database for Meta-Analysis of the Prognostic Value of Genes. BMC Med Genomics, 2009, 2: 18., DOI 10.1186/1755-8794-2-18 Cai L, Lin S, Girard L, Zhou Y, Yang L, Ci B, et al. LCE: an OpenWeb portal to Explore Gene Expression and Clinical Associations in Lung Cancer. Oncogene, 2019, 38(14):2551–64., DOI 10.1038/s41388-018-0588-2 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Sci Signal, 2013, 6(269):pl1., DOI 10.1126/scisignal.2004088 Vasaikar SV, Straub P, Wang J, and Zhang B. LinkedOmics: Analyzing Multi- Omics Data within and across 32 Cancer Types. Nucleic Acids Res, 2018, 46(D1):D956-D963–d963., DOI 10.1093/nar/gkx1090 Ru B, Wong CN, Tong Y, Zhong JY, Zhong SSW, Wu WC, et al. TISIDB: an Integrated Repository portal for Tumor-Immune System Interactions. Bioinformatics, 2019, 35(20):4200–2., DOI 10.1093/bioinformatics/btz210 Wei T, Song J, Liang K, Li L, Mo X, Huang Z, et al. Identification of a Novel Therapeutic Cand idate, NRK, in Primary Cancer-Associated Fibroblasts of Lung Adenocarcinoma Microenvironment. J Cancer Res Clin Oncol, 2021, 147(4):1049–64., DOI 10.1007/s00432-020-03489-z Li L, Pan Y,Mo X,Wei T, Song J, LuoM, et al.ANovel Metastatic Promoter CEMIP and its Downstream Molecular Targets and Signaling Pathway of CellularMigration and Invasion in SCLC Cells Based on Proteome Analysis. J Cancer Res Clin Oncol, 2020, 146(10):2519–34., DOI 10.1007/s00432-020-03308-5 Liu M, Zhao SQ, Yang L, Li X, Song X, Zheng Y, et al. A Direct Immunohistochemistry, IHC, Method Improves the Intraoperative Diagnosis of Breast Papillary Lesions Including Breast Cancer. Discov Med, 2019, 28(151):29–37. de Sousa Vml, and Carvalho L. Heterogeneity in Lung Cancer. Pathobiology, 2018, 85(1-2):96–107., DOI 10.1159/000487440 Kikuno R, Nagase T, Nakayama M, Koga H, OkazakiN,NakajimaD, et al. HUGE: a Database for Human KIAA Proteins, a 2004 Update Integrating HUGEppi and ROUGE. Nucleic Acids Res, 2004, 32, D502–4., DOI 10.1093/nar/gkh035 Liu J, Yan W, Han P, and Tian D. The Emerging Role of KIAA1199 in Cancer Development and Therapy. Biomed Pharmacother, 2021, 138:111507., DOI 10. 1016/j.biopha.2021.111507 Chen Y, Zhou H, Jiang WJ,Wang JF, Tian Y, Jiang Y, et al. The Role of CEMIP in Tumors: An Update Based on Cellular and Molecular Insights. Biomed Pharmacother, 2022, 146:112504., DOI 10.1016/j.biopha.2021.112504 Cheng J, Zhang Y, Wan R, Zhou J, Wu X, Fan Q, et al. CEMIP Promotes Osteosarcoma Progression and Metastasis Through Activating Notch Signaling Pathway. Front Oncol, 2022, 12:919108., DOI 10.3389/fonc.2022. 919108 Hsieh IY, He J, Wang L, Lin B, Liang Z, Lu B, et al. H3K27me3 Loss Plays a Vital Role in CEMIP Mediated Carcinogenesis and Progression of Breast Cancer with Poor Prognosis. Biomed Pharmacother, 2020, 123:109728., DOI 10. 1016/j.biopha.2019.109728 Przytycki Pf, and Singh M. Differential Analysis between Somatic Mutation and Germline Variation Profiles Reveals Cancer-Related Genes. Genome Med, 2017, 9(1):79., DOI 10.1186/s13073-017-0465-6 Hayes J, Peruzzi PP, and Lawler S. MicroRNAs in Cancer: Biomarkers, Functions and Therapy. Trends Mol Med, 2014, 20(8):460–9., DOI 10.1016/j. molmed.2014.06.005 Lee Y, Dutta A. MicroRNAs in Cancer. Annu Rev Pathol, 2009, 4:199–227., DOI 10.1146/annurev.pathol.4.110807.092222 O’Brien J, Hayder H, Zayed Y, and Peng C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front Endocrinol, Lausanne,, 2018, 9:402., DOI 10.3389/fendo.2018.00402 Wang L, Yu T, Li W, Li M, Zuo Q, Zou Q, et al. The miR-29c-Kiaa1199 axis Regulates Gastric Cancer Migration by Binding with WBP11 and PTP4A3. Oncogene, 2019, 38(17):3134–50., DOI 10.1038/s41388-018-0642-0 Monoe Y, Jingushi K, Kawase A, Hirono T, Hirose R, Nakatsuji Y, et al. Pharmacological Inhibition of miR-130 Family Suppresses Bladder Tumor Growth by Targeting Various Oncogenic Pathways via PTPN1. Int J Mol Sci, 2021, 22(9):4751., DOI 10.3390/ijms22094751 Li S, Jia H, Zhang Z, and Wu D. DRAIC Promotes Growth of Breast Cancer by Sponging miR-432-5p to Upregulate SLBP. Cancer Gene Ther, 2021, 29: 951–60., DOI 10.1038/s41417-021-00388-4 Liu Y, Liu R, Yang F, Cheng R, Chen X, Cui S, et al. miR-19a Promotes Colorectal Cancer Proliferation and Migration by Targeting TIA1. Mol Cancer, 2017, 16(1):53., DOI 10.1186/s12943-017-0625-8 Chen J, Zhang K, Zhi Y, Wu Y, Chen B, Bai J, et al. Tumor-derived Exosomal miR-19b-3p Facilitates M2 Macrophage Polarization and Exosomal LINC00273 Secretion to Promote Lung Adenocarcinoma Metastasis via Hippo Pathway. Clin Transl Med, 2021, 11(9):e478., DOI 10.1002/ctm2.478 Muller S, Glaß M, SinghAK, Haase J, BleyN, Fuchs T, et al. IGF2BP1 Promotes SRFdependent Transcription in Cancer in a m6A- and miRNA-dependent Manner. Nucleic Acids Res, 2019, 47(1):375–90., DOI 10.1093/nar/gky1012 Li T, Wan Y, Su Z, Li J, Han M, and Zhou C. SRF Potentiates Colon Cancer Metastasis and Progression in a microRNA-214/ptk6-dependent Manner. Cancer Manag Res, 2020, 12:6477–91., DOI 10.2147/cmar.s257422 Wang HY, Zhang B, Zhou JN, Wang DX, Xu YC, Zeng Q, et al. Arsenic Trioxide Inhibits Liver Cancer StemCells andMetastasis by Targeting SRF/MCM7 Complex. Cel Death Dis, 2019, 10(6):453., DOI 10.1038/s41419-019-1676-0 Anderson Nm, and Simon MC. The Tumor Microenvironment. Curr Biol, 2020, 30(16):R921-R925–r925., DOI 10.1016/j.cub.2020.06.081 Hinshaw Dc, and Shevde LA. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res, 2019, 79(18):4557–66., DOI 10. 1158/0008-5472.can-18-3962 Yoshida H, Nagaoka A, Kusaka-Kikushima A, Tobiishi M, Kawabata K, Sayo T, et al. KIAA1199, a Deafness Gene of Unknown Function, Is a New Hyaluronan Binding Protein Involved in Hyaluronan Depolymerization. Proc Natl Acad Sci U S A, 2013, 110(14):5612–7., DOI 10.1073/pnas.1215432110 Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S, et al. Extracellular Matrix and its Therapeutic Potential for Cancer Treatment. Signal Transduct Target Ther, 2021, 6(1):153., DOI 10.1038/s41392-021-00544-0 Kwapiszewska G, Gungl A, Wilhelm J, Marsh LM, Thekkekara Puthenparampil H, Sinn K, et al. Transcriptome Profiling Reveals the Complexity of Pirfenidone Effects in Idiopathic Pulmonary Fibrosis. Eur Respir J, 2018, 52(5):1800564., DOI 10.1183/13993003.00564-2018 Shi R, Zhang Z, Zhu A, Xiong X, Zhang J, Xu J, et al. Targeting Type I Collagen for Cancer Treatment. Int J Cancer, 2022, 151(5):665–83., DOI 10.1002/ijc. 33985 Xiang H, Ramil CP, Hai J, Zhang C, Wang H, Watkins AA, et al. Cancer- Associated Fibroblasts Promote Immunosuppression by Inducing ROSGenerating Monocytic MDSCs in Lung Squamous Cell Carcinoma. Cancer Immunol Res, 2020, 8(4):436–50., DOI 10.1158/2326-6066.cir- 19-0507 Bremnes RM, Donnem T, Al-Saad S, Al-Shibli K, Andersen S, Sirera R, et al. The Role of Tumor Stroma in Cancer Progression and Prognosis: Emphasis on Carcinoma-Associated Fibroblasts and Non-small Cell Lung Cancer. J Thorac Oncol, 2011, 6(1):209–17., DOI 10.1097/JTO.0b013e3181f8a1bd Nagarsheth N, Wicha MS, and Zou W. Chemokines in the Cancer Microenvironment and Their Relevance in Cancer Immunotherapy. Nat Rev Immunol, 2017, 17(9):559–72., DOI 10.1038/nri.2017.49 Wu J, Liu X, Wu J, Lou C, Zhang Q, Chen H, et al. CXCL12 Derived from CD248-Expressing Cancer-Associated Fibroblasts Mediates M2-Polarized Macrophages to Promote Nonsmall Cell Lung Cancer Progression. Biochim Biophys Acta Mol Basis Dis, 2022, 1868(11):166521., DOI 10.1016/j.bbadis.2022. 166521 Xue J, Song Y, Xu W, and Zhu Y. The CDK1-Related lncRNA and CXCL8 Mediated Immune Resistance in Lung Adenocarcinoma. Cells, 2022, 11(17):2688., DOI 10.3390/cells11172688 Lu Q, Yin H, Deng Y, ChenW, Diao W, Ding M, et al. circDHTKD1 Promotes Lymphatic Metastasis of Bladder Cancer by Upregulating CXCL5. Cell Death Discov, 2022, 8(1):243., DOI 10.1038/s41420-022-01037-x Santolla MF, Talia M, Cirillo F, Scordamaglia D, De Rosis S, Spinelli A, et al. The AGEs/RAGE Transduction Signaling Prompts IL-8/CXCR1/2-Mediated Interaction between Cancer-Associated Fibroblasts, CAFs, and Breast Cancer Cells. Cells, 2022, 11(15):2402., DOI 10.3390/cells11152402 Baer C, Kimura S, Rana MS, Kleist AB, Flerlage T, Feith DJ, et al. CCL22 Mutations Drive Natural Killer Cell Lymphoproliferative Disease by Deregulating Microenvironmental Crosstalk. Nat Genet, 2022, 54(5):637–48., DOI 10.1038/s41588-022-01059-2 Sarkar T, Dhar S, Chakraborty D, Pati S, Bose S, Pand a AK, et al. FOXP3/ HAT1 Axis Controls Treg Infiltration in the Tumor Microenvironment by Inducing CCR4 Expression in Breast Cancer. Front Immunol, 2022, 13: 740588., DOI 10.3389/fimmu.2022.740588 Wang T, Zhou Q, Zeng H, Zhang H, Liu Z, Shao J, et al. CCR8 Blockade Primes Anti-tumor Immunity through Intratumoral Regulatory T Cells Destabilization in Muscle-Invasive Bladder Cancer. Cancer Immunol Immunother, 2020, 69(9):1855–67., DOI 10.1007/s00262-020- 02583-y Xu J, Li JQ, Chen QL, Shestakova EA, Misyurin VA, Pokrovsky VS, et al. Advances in Research on the Effects and Mechanisms of Chemokines and Their Receptors in Cancer. Front Pharmacol, 2022, 13:920779., DOI 10.3389/ fphar.2022.920779 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2022 VL 28 IS 4 BP 1610754 EP 1610769 DI 10.3389/pore.2022.1610754 PG 16 ER PT J AU Olah, O Majlat, E Koszo, R Vereb, Z Voros, A AF Olah, Orsolya Majlat, Edit Koszo, Renata Vereb, Zoltan Voros, Andras TI Predictive role of neostromal CD10 expression in breast cancer patients treated with neoadjuvant chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE advanced breast cancer; predictive factors; CD10; neoadjuvant therapy; neostroma ID advanced breast cancer; predictive factors; CD10; neoadjuvant therapy; neostroma AB Background: The therapeutic strategy of invasive breast cancer is based on routine histopathological markers (estrogen-, progesterone receptor, HER2, Ki67) routinely evaluated in tumor cells. However, the assessment of cancer stroma could influence therapeutic strategies. Studies have shown that stromal expression of CD10, a zinc-dependent metalloproteinase, is associated with biological aggressiveness of the tumor. In the present retrospective study, we aimed to evaluate stromal CD10 expression and association between CD10 expression and response to neoadjuvant chemotherapy in invasive breast cancer. Methods: CD10 immunohistochemistry was performed on core biopsies taken before the neoadjuvant therapy. Stromal CD10 expression was determined and compared with well-known predictive and prognostic tissue markers as well as with the following groups defined according to the degree of tumor response: no regression, partial regression, and complete regression. Results: A total of 60 locally advanced invasive breast carcinomas of “no special type” were included. The proportion of CD10 positive tumors was significantly higher in the “no regression” group compared to “complete regression” group (p = 0.000). Stromal CD10 expression was found to be significantly associated with decrease in response to neoadjuvant chemotherapy. According to CD10 expression we did not find any difference in hormone receptor status, Ki67, tumor grade or neostromal area. Conclusion: Our data suggest that CD10 expression can serve as a predictive marker of the effect of neoadjuvant chemotherapy in breast cancer patients. Therefore, its inclusion into the routine assessment of biopsies to tailor tumorspecific therapeutic strategies merits consideration. C1 [Olah, Orsolya] University of Szeged, Department of PathologySzeged, Hungary. [Majlat, Edit] University of Szeged, Department of PathologySzeged, Hungary. [Koszo, Renata] University of Szeged, Department of OncotherapySzeged, Hungary. [Vereb, Zoltan] University of Szeged, Department of Dermatology and AllergologySzeged, Hungary. [Voros, Andras] University of Szeged, Department of PathologySzeged, Hungary. RP Voros, A (reprint author), University of Szeged, Department of Pathology, Szeged, Hungary. EM andrasvoros@libero.it CR De Wever O, Mareel M. Role of tissue stroma in cancer cell invasion. J Pathol, 2003, 200(4):429–47., DOI 10.1002/path.1398 Bremnes RM, Donnem T, Al-Saad S, Al-Shibli K, Andersen S, Sirera R, et al. The role of tumor stroma in cancer progression and prognosis: Emphasis on carcinoma-associated fibroblasts and non-small cell lung cancer. J Thorac Oncol, 2011, 6(1):209–17., DOI 10.1097/JTO.0b013e3181f8a1bd Orimo A, Weinberg RA. Stromal fibroblasts in cancer: A novel tumorpromoting cell type. Cell Cycle, 2006, 5(15):1597–601., DOI 10.4161/cc.5.15.3112 Paulsson J, Micke P. Prognostic relevance of cancer-associated fibroblasts in human cancer. Semin Cancer Biol, 2014, 25:61–8., DOI 10.1016/j.semcancer.2014. 02.006 Su S, Chen J, Yao H, Liu J, Yu S, Lao L, et al. CD10+GPR77+ cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell, 2018, 172(4):841–56. e16., DOI 10.1016/j.cell.2018.01.009 Song J, Aumuller G, Xiao F, Wilhelm B, Albrecht M. Cell specific expression of CD10/neutral endopeptidase 24.11 gene in human prostatic tissue and cells. Prostate, 2004, 58(4):394–405., DOI 10.1002/pros.10345 Sato Y, Itoh F, Hinoda Y, Ohe Y, Nakagawa N, Ueda R, et al. Expression of CD10/neutral endopeptidase in normal and malignant tissues of the human stomach and colon. J Gastroenterol, 1996, 31(1):12–7., DOI 10.1007/BF01211181 Loke SL, Leung CY, Chiu KY, Yau WL, Cheung KN, Ma L. Localisation of CD10 to biliary canaliculi by immunoelectron microscopical examination. J Clin Pathol, 1990, 43(8):654–6., DOI 10.1136/jcp.43.8.654 Kovari B, Bathori A, Cserni G. CD10 immunohistochemical expression in apocrine lesions of the breast. Pathobiology, 2015, 82(6):259–63., DOI 10.1159/ 000440664 Maguer-Satta V, Chapellier M, Delay E, Bachelard-Cascales E. CD10: A tool to crack the role of stem cells in breast cancer. Proc Natl Acad Sci U S A, 2011, 108(49): E1264; author reply E1265. ; author reply E1265, 2011)., DOI 10.1073/pnas. 1116567108 Shipp MA, Look AT. Hematopoietic differentiation antigens that are membrane-associated enzymes: Cutting is the key. Blood, 1993, 82(4):1052–70., DOI 10.1182/blood.v82.4.1052.1052 Makretsov NA, Hayes M, Carter BA, Dabiri S, Gilks CB, Huntsman DG. Stromal CD10 expression in invasive breast carcinoma correlates with poor prognosis, estrogen receptor negativity, and high grade. Mod Pathol, 2007, 20(1):84–9., DOI 10.1038/modpathol.3800713 Voutsadakis IA, Vlachostergios PJ, Daliani DD, Karasavvidou F, Kakkas G, Moutzouris G, et al. CD10 is inversely associated with nuclear factor-kappa B and predicts biochemical recurrence after radical prostatectomy. Urol Int, 2012, 88(2): 158–64., DOI 10.1159/000335299 Horvath Z, Boer K, Dank M, Kahan Z, Kocsis J, Kover E, et al. Systemic treatment of breast cancer: Professional guideline]. Magy Onkol, 2020, 64(4): 348–68. Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, et al. The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: Recommendations by an international TILs working group 2014. Ann Oncol, 2014, 26(2):259–71., DOI 10.1093/annonc/mdu450 Wolff AC, Hammond MEH, Allison KH, Harvey BE,Mangu PB, Bartlett JMS, et al. Human epidermal growth factor receptor 2 testing in breast cancer: American society of clinical oncology/college of American pathologists clinical practice guideline focused update. J Clin Oncol, 2018, 36(20):2105–22., DOI 10.1200/jco. 2018.77.8738 Allison KH, Hammond MEH, Dowsett M, McKernin SE, Carey LA, Fitzgibbons PL, et al. Estrogen and progesterone receptor testing in breast cancer: ASCO/CAP guideline update. J Clin Oncol, 2020, 38(12):1346–66., DOI 10.1200/jco.19.02309 Cserni G, Kulka J, Francz M, Jaray B, Kalman E, Kovacs I, et al. Pathological diagnosis, work-up and reporting of breast cancer. Recommendations of the 3rd Hungarian Consensus Conference on Breast Cancer]. Magy Onkol, 2016, 60(3):209 Amendoeira I, et al. European guidelines for quality assurance in breast cancer screening and diagnosis. In: Ed N Perry, M Broeders, C de Wolf, S Tornberg, R Holland&L. von Karsa. European Commission, Belgium, 2013) Iwaya K, Ogawa H, Izumi M, Kuroda M, Mukai K. Stromal expression of CD10 in invasive breast carcinoma: A new predictor of clinical outcome. Virchows Arch, 2002, 440(6):589–93., DOI 10.1007/s00428-002-0639-4 Mott JD, Werb Z. Regulation of matrix biology by matrix metalloproteinases. Curr Opin Cel Biol, 2004, 16(5):558–64., DOI 10.1016/j.ceb.2004.07.010 Vermeulen L, De Sousa EMelo F, van der Heijden M, Cameron K, de Jong JH, Borovski T, et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cel Biol, 2010, 12(5):468–76., DOI 10.1038/ncb2048 Lin EY, Pollard JW. Macrophages: Modulators of breast cancer progression. Novartis Found Symp, 2004, 256:158–68. ; discussion 168-72, 259-69, 2004). Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer, 2004, 4(1):71–8., DOI 10.1038/nrc1256 Jinga DC, Blidaru A, Condrea I, Ardeleanu C, Dragomir C, Szegli G, et al. MMP-9 and MMP-2 gelatinases and TIMP-1 and TIMP-2 inhibitors in breast cancer: Correlations with prognostic factors. J Cel Mol Med, 2006, 10(2):499–510., DOI 10.1111/j.1582-4934.2006.tb00415.x Curran CS, Keely PJ. Breast tumor and stromal cell responses to TGF-β and hypoxia in matrix deposition. Matrix Biol, 2013, 32(2):95–105., DOI 10.1016/j. matbio.2012.11.016 Kass L, Erler JT, Dembo M, Weaver VM. Mammary epithelial cell: Influence of extracellular matrix composition and organization during development and tumorigenesis. Int J Biochem Cel Biol, 2007, 39(11):1987–94., DOI 10.1016/j.biocel. 2007.06.025 Maguer-Satta V, Besancon R, Bachelard-Cascales E. Concise review: Neutral endopeptidase, CD10): A multifaceted environment actor in stem cells, physiological mechanisms, and cancer. Stem Cells, 2011, 29(3):389–96., DOI 10. 1002/stem.592 Smollich M, Gotte M, Yip GW, Yong ES, Kersting C, Fischgrabe J, et al. On the role of endothelin-converting enzyme-1, ECE-1, and neprilysin in human breast cancer. Breast Cancer Res Treat, 2007, 106(3):361–9., DOI 10.1007/s10549-007- 9516-9 Thomas S, Babu RJ, Agarwal K, Puri V, Jain M, Andley M, et al. Effect of neoadjuvant chemotherapy on stromal CD10 antigens in breast cancer - a preliminary study. Indian J Cancer, 2013, 50(1):46–51., DOI 10.4103/0019-509X. 112299 Jana S, Jha B, Patel C, Jana D, Agarwal A. CD10-A new prognostic stromal marker in breast carcinoma, its utility, limitations and role in breast cancer pathogenesis. Indian J Pathol Microbiol, 2014, 57(4):530–6., DOI 10.4103/0377- 4929.142639 Goodman OB, Febbraio M, Simantov R, Zheng R, Shen R, Silverstein RL, et al. Neprilysin inhibits angiogenesis via proteolysis of fibroblast growth factor-2. J Biol Chem, 2006, 281(44):33597–605., DOI 10.1074/jbc.M602490200 Puri V, Jain M, Mahajan G, Pujani M. Critical appraisal of stromal CD10 staining in fibroepithelial lesions of breast with a special emphasis on expression patterns and correlation with WHO grading. J Cancer Res Ther, 2016, 12(2):667–70., DOI 10.4103/0973-1482.177215 Dhande AN, Sinai Khandeparkar SG, Joshi AR, Kulkarni MM, Pandya N, Mohanapure N, et al. Stromal expression of CD10 in breast carcinoma and its correlation with clinicopathological parameters. South Asian J Cancer, 2019, 8(1): 18–21., DOI 10.4103/sajc.sajc_56_18 Vo TN, Mekata E, Umeda T, Abe H, Kawai Y, Mori T, et al. Prognostic impact of CD10 expression in clinical outcome of invasive breast carcinoma. Breast Cancer, 2015, 22(2):117–28., DOI 10.1007/s12282-013-0459-1 Desmedt C, Majjaj S, Kheddoumi N, Singhal SK, Haibe-Kains B, El Ouriaghli F, et al. Characterization and clinical evaluation of CD10+ stroma cells in the breast cancer microenvironment. Clin Cancer Res, 2012, 18(4):1004–14., DOI 10.1158/ 1078-0432.CCR-11-0383 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610598 EP 1610605 DI 10.3389/pore.2022.1610598 PG 8 ER PT J AU Behnke, A Cayre, A De Maglio, G Giannini, G Habran, L Tarsitano, M Chetta, M Cappellen, D Lespagnol, A Le Naoures, C Massazza, G Destro, A Bonzheim, I Rau, A Battmann, A Kah, B Watkin, E Hummel, M AF Behnke, Anke Cayre, Anne De Maglio, Giovanna Giannini, Giuseppe Habran, Lionel Tarsitano, Marina Chetta, Massimiliano Cappellen, David Lespagnol, Alexandra Le Naoures, Cecile Massazza, Gabriella Destro, Annarita Bonzheim, Irina Rau, Achim Battmann, Achim Kah, Bettina Watkin, Emmanuel Hummel, Michael TI FACILITATE: A real-world, multicenter, prospective study investigating the utility of a rapid, fully automated real-time PCR assay versus local reference methods for detecting epidermal growth factor receptor variants in NSCLC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE non-small cell lung cancer; epidermal growth factor receptor; DNA mutational analysis; clinical decision-making; turnaround time ID non-small cell lung cancer; epidermal growth factor receptor; DNA mutational analysis; clinical decision-making; turnaround time AB Accurate testing for epidermal growth factor receptor (EGFR) variants is essential for informing treatment decisions in non-small cell lung cancer (NSCLC). Automated diagnostic workflows may allow more streamlined initiation of targeted treatments, where appropriate, while comprehensive variant analysis is ongoing. FACILITATE, a real-world, prospective, multicenter, European study, evaluated performance and analytical turnaround time of the Idylla™ EGFR Mutation Test compared with local reference methods. Sixteen sites obtained formalin-fixed paraffin-embedded biopsy samples with ≥ 10% neoplastic cells from patients with NSCLC. Consecutive 5 μm sections from patient samples were tested for clinically relevant NSCLC-associated EGFR variants using the Idylla™ EGFR Mutation Test and local reference methods; performance (concordance) and analytical turnaround time were compared. Between January 2019 and November 2020, 1,474 parallel analyses were conducted. Overall percentage agreement was 97.7% [n = 1,418; 95% confidence interval (CI): 96.8–98.3], positive agreement, 87.4% (n = 182; 95% CI: 81.8–91.4) and negative agreement, 99.2% (n = 1,236; 95% CI: 98.5–99.6). There were 38 (2.6%) discordant cases. Ninety percent of results were returned with an analytical turnaround time of within 1 week using the Idylla™ EGFR Mutation Test versus ~22 days using reference methods. The Idylla™ EGFR Mutation Test performed well versus local methods and had shorter analytical turnaround time. The Idylla™ EGFR Mutation Test can thus support application of personalized medicine in NSCLC. C1 [Behnke, Anke] Charite-Universitatsmedizin Berlin, Corporate Member of Freie Universitat Berlin and Humboldt-Universitat zu Berlin, Institute of Pathology and Berlin Institute of HealthBerlin, Germany. [Cayre, Anne] Centre Jean-Perrin, Departement de PathologieClermont-Ferrand, France. [De Maglio, Giovanna] Azienda Sanitaria Universitaria Friuli Centrale, Santa Maria della Misericordia Hospital, Pathology DepartmentUdine, Italy. [Giannini, Giuseppe] Universita di Roma La Sapienza, Department Molecular MedicineRome, Italy. [Habran, Lionel] CHU Sart Tilman, Department of AnatomopathologyLiege, Belgium. [Tarsitano, Marina] U.O.C. di Genetica Medica, Medical Genetics Laboratory, and Ospedale Antonio Cardarelli, Di Laboratorio, A.O.R.N. CardarelliNaples, Italy. [Chetta, Massimiliano] U.O.C. di Genetica Medica, Medical Genetics Laboratory, and Ospedale Antonio Cardarelli, Di Laboratorio, A.O.R.N. CardarelliNaples, Italy. [Cappellen, David] Centre Hospitalier Universitaire de Bordeaux, Hopital du Haut Leveque, Service de Biologie des TumeursPessac, France. [Lespagnol, Alexandra] CHU de Rennes, Laboratoire de Genetique Somatique des CancersRennes, France. [Le Naoures, Cecile] CHU de Rennes, Service d’Anatomie et Cytologie PathologiquesRennes, France. [Massazza, Gabriella] ASST Papa Giovanni XXIII, Dipartimento Medicina di Laboratorio Anatomia PatologicaBergamo, BG, Italy. [Destro, Annarita] Humanitas Clinical and Research Center—IRCCS, Pathology DepartmentMilan, Italy. [Bonzheim, Irina] University Hospital Tubingen, Eberhard Karls University of Tubingen and Comprehensive Cancer Center, Institute of Pathology and NeuropathologyTubingen, Germany. [Rau, Achim] University Hospital Tubingen, Eberhard Karls University of Tubingen and Comprehensive Cancer Center, Institute of Pathology and NeuropathologyTubingen, Germany. [Battmann, Achim] Institut fur Pathologie und Zytodiagnostik am Krankenhaus NordwestFrankfurt, Germany. [Kah, Bettina] Institute for Hematopathology HamburgHamburg, Germany. [Watkin, Emmanuel] CYPATHVilleurbanne, France. [Hummel, Michael] Charite-Universitatsmedizin Berlin, Corporate Member of Freie Universitat Berlin and Humboldt-Universitat zu Berlin, Institute of Pathology and Berlin Institute of HealthBerlin, Germany. RP Hummel, M (reprint author), Charite-Universitatsmedizin Berlin, Corporate Member of Freie Universitat Berlin and Humboldt-Universitat zu Berlin, Institute of Pathology and Berlin Institute of Health, Berlin, Germany. EM michael.hummel@charite.de CR Brown NA, Aisner DL, Oxnard GR. Precision medicine in non–small cell lung cancer: Current standards in Pathology and biomarker interpretation. Am Soc Clin Oncol Educ Book, 2018, 38:708–15., DOI 10.1200/EDBK_209089 Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med, 2004, 350(21):2129–39., DOI 10.1056/NEJMoa040938 Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science, 2004, 304(5676):1497–500., DOI 10.1126/science.1099314 Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, et al. Metastatic non-small cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Updated version published 15 september 2020 by the ESMO guidelines committee [online]., 2020,, 2020). Available: https://www.esmo.org/guidelines/lung-and-chest-tumours/clinical-practice-livingguidelines- metastatic-non-small-cell-lung-cancer, Accessed September 25, 2020). Zhang YL, Yuan JQ, Wang KF, Fu XH, Han XR, Threapleton D, et al. The prevalence of EGFR mutation in patients with non-small cell lung cancer: A systematic review and meta-analysis. Oncotarget, 2016, 7(48):78985–93., DOI 10. 18632/oncotarget.12587 Murray S, Dahabreh IJ, Linardou H, Manoloukos M, Bafaloukos D, Kosmidis P. Somatic mutations of the tyrosine kinase domain of epidermal growth factor receptor and tyrosine kinase inhibitor response to TKIs in non-small cell lung cancer: An analytical database. J Thorac Oncol Official Publ Int Assoc Study Lung Cancer, 2008, 3(8):832–9., DOI 10.1097/JTO.0b013e31818071f3 Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer, eurtac): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol, 2012, 13(3):239–46., DOI 10.1016/S1470-2045(11)70393-X Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med, 2009, 361(10): 947–57., DOI 10.1056/NEJMoa0810699 Wu YL, Zhou C, Liam CK, Wu G, Liu X, Zhong Z, et al. First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: Analyses from the phase III, randomized, open-label, ENSURE study. Ann Oncol, 2015, 26(9):1883–9., DOI 10.1093/annonc/mdv270 Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, et al. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med, 2018, 378(2):113–25., DOI 10.1056/NEJMoa1713137 Leonetti A, Sharma S, Minari R, Perego P, Giovannetti E, Tiseo M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer, 2019, 121(9):725–37., DOI 10.1038/s41416-019-0573-8 NiederstMJ, Hu H, Mulvey HE, Lockerman EL, Garcia AR, Piotrowska Z, et al. The allelic context of the C797S mutation acquired upon treatment with third-generation EGFR inhibitors impacts sensitivity to subsequent treatment strategies. Clin Cancer Res, 2015, 21(17):3924–33., DOI 10.1158/1078-0432. Ccr-15-0560 NCCN Clinical Practice Guidelines. Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology, NCCN Guidelines®, for Non-Small Cell Lung Cancer V.3.2022. © National Comprehensive Cancer Network, Inc. 2022. All rights reserved. Accessed March 17, 2022. To view the most recent and complete version of the guideline, go online to NCCN.org. NCCN makes no warranties of any kind whatsoever regarding their content, use or application and disclaims any responsibility for their application or use in any way, 2022). Cross DA, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, et al. AZD9291, an irreversible EGFR TKI, overcomes t790m-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov, 2014, 4(9):1046–61., DOI 10.1158/ 2159-8290.CD-14-0337 Reungwetwattana T, Nakagawa K, Cho BC, Cobo M, Cho EK, Bertolini A, et al. CNS response to osimertinib versus standard epidermal growth factor receptor tyrosine kinase inhibitors in patients with untreated EGFR-mutated advanced non–small-cell lung cancer. J Clin Oncol, 2018, 36(33):3290–7., DOI 10.1200/jco. 2018.78.3118 Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, Ohe Y, et al. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med, 2020, 382(1):41–50., DOI 10.1056/NEJMoa1913662 Wu YL, Ahn MJ, Garassino MC, Han JY, Katakami N, Kim HR, et al. CNS efficacy of osimertinib in patients with t790m-positive advanced non-small-cell lung cancer: Data from a randomized phase III trial, AURA3). J Clin Oncol, 2018, 36(26):2702–9., DOI 10.1200/jco.2018.77.9363 Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, et al. Osimertinib or platinum-pemetrexed in EGFR t790m-positive lung cancer. N Engl J Med, 2017, 376(7):629–40., DOI 10.1056/NEJMoa1612674 Hanna NH, Robinson AG, Temin S, Baker S, Brahmer JR, Ellis PM, et al. Therapy for stage IV non–small-cell lung cancer with driver alterations: ASCO and OH, CCO, joint guideline update. J Clin Oncol, 2021, 20:1040–91., DOI 10.1200/ JCO.20.03570 Leighl NB, Rekhtman N, Biermann WA, Huang J, Mino-Kenudson M, Ramalingam SS, et al. Molecular testing for selection of patients with lung cancer for epidermal growth factor receptor and anaplastic lymphoma kinase tyrosine kinase inhibitors: American society of clinical Oncology endorsement of the college of American pathologists/international association for the study of lung cancer/association for molecular Pathology guideline. J Clin Oncol, 2014, 32(32):3673–9., DOI 10.1200/JCO.2014.57.3055 Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker E, et al. Updated molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors: Guideline from the college of American pathologists, the international association for the study of lung cancer, and the association for molecular Pathology. Arch Pathol Lab Med, 2018, 142(3): 321–46., DOI 10.5858/arpa.2017-0388-CP Oxnard GR, Miller VA, Robson ME, Azzoli CG, Pao W, Ladanyi M, et al. Screening for germline EGFR T790M mutations through lung cancer genotyping. J Thorac Oncol, 2012, 7(6):1049–52., DOI 10.1097/JTO.0b013e318250ed9d Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, et al. Recommendations for the use of next-generation sequencing, NGS, for patients with metastatic cancers: A report from the ESMO precision medicine working group. Ann Oncol, 2020, 31(11):1491–505., DOI 10.1016/j.annonc.2020.07.014 Mardis ER. DNA sequencing technologies: 2006–2016. Nat Protoc, 2017, 12(2):213–8., DOI 10.1038/nprot.2016.182 Grossmann V, Kohlmann A, Klein HU, Schindela S, Schnittger S, Dicker F, et al. Targeted next-generation sequencing detects point mutations, insertions, deletions and balanced chromosomal rearrangements as well as identifies novel leukemia-specific fusion genes in a single procedure. Leukemia, 2011, 25(4): 671–80., DOI 10.1038/leu.2010.309 Takeda M, Sakai K, Terashima M, Kaneda H, Hayashi H, Tanaka K, et al. Clinical application of amplicon-based next-generation sequencing to therapeutic decision making in lung cancer. Ann Oncol, 2015, 26(12):2477–82., DOI 10.1093/ annonc/mdv475 Royal College of Physicians. Spotlight report on molecular testing in advanced lung cancer, 2020). Available: https://www.hqip.org.uk/wp-content/uploads/2020/ 01/REF100_NLCA_Spotlight-Molec-Test_FINAL-TYPESET_web_20200108.pdf, Accessed July 14, 2021). Gregg JP, Li T, Yoneda KY. Molecular testing strategies in non-small cell lung cancer: Optimizing the diagnostic journey. Transl Lung Cancer Res, 2019, 8(3): 286–301., DOI 10.21037/tlcr.2019.04.14 Lassalle S, Hofman V, Heeke S, Benzaquen J, Long E, Poudenx M, et al. Targeted assessment of the EGFR status as reflex testing in treatment-naive nonsquamous cell lung carcinoma patients: A single laboratory experience, LPCE, Nice, France). Cancers, 2020, 12(4):955., DOI 10.3390/cancers12040955 Kerr KM, Bibeau F, Thunnissen E, Botling J, Ryska A, Wolf J, et al. The evolving landscape of biomarker testing for non-small cell lung cancer in europe. Lung Cancer, 2021, 154:161–75., DOI 10.1016/j.lungcan.2021.02.026 Lim C, Tsao MS, Le LW, Shepherd FA, Feld R, Burkes RL, et al. Biomarker testing and time to treatment decision in patients with advanced non-small-cell lung cancer. Ann Oncol, 2015, 26(7):1415–21., DOI 10.1093/annonc/mdv208 Rangachari D, Drake L, Huberman MS, McDonald DC, VanderLaan PA, Folch E, et al. Rapidly fatal advanced EGFR-mutated lung cancers and the need for rapid tumor genotyping in clinical practice. Cancer Treat Commun, 2016, 9:41–3., DOI 10.1016/j.ctarc.2016.07.001 Gutierrez ME, Choi K, Lanman RB, Licitra EJ, Skrzypczak SM, Pe Benito R, et al. Genomic profiling of advanced non-small cell lung cancer in community settings: Gaps and opportunities. Clin Lung Cancer, 2017, 18(6):651–9., DOI 10. 1016/j.cllc.2017.04.004 Wu Y-L, Tsuboi M, He J, John T, Grohe C, Majem M, et al. Osimertinib in resected EGFR-mutated non–small-cell lung cancer. N Engl J Med, 2020, 383(18): 1711–23., DOI 10.1056/NEJMoa2027071 Remon J, Soria JC, Peters S, clinicalguidelines@esmo.org EGCEa. Early and locally advanced non-small-cell lung cancer: An update of the ESMO clinical practice guidelines focusing on diagnosis, staging, systemic and local therapy. Ann Oncol, 2021, 32(12):1637–42., DOI 10.1016/j.annonc.2021.08.1994 Liam C-K, Mallawathantri S, Fong KM. Is tissue still the issue in detecting molecular alterations in lung cancer? Respirology, 2020, 25(9):933–43., DOI 10.1111/ resp.13823 Kris MG, Johnson BE, Berry LD, Kwiatkowski DJ, Iafrate AJ,Wistuba B, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA, 2014, 311(19):1998–2006., DOI 10.1001/jama.2014.3741 Cheema PK, Gomes M, Banerji S, Joubert P, Leighl NB, Melosky B, et al. Consensus recommendations for optimizing biomarker testing to identify and treat advanced EGFR-mutated non-small-cell lung cancer. Curr Oncol, 2020, 27(6): 321–9., DOI 10.3747/co.27.7297 Delgado-Garcia M, Weynand B, Gomez-Izquierdo L, Hernandez MJ, Blanco AM, Varela M, et al. Clinical performance evaluation of the Idylla™EGFR mutation test on formalin-fixed paraffin-embedded tissue of non-small cell lung cancer. BMC cancer, 2020, 20(1):275., DOI 10.1186/s12885-020-6697-7 Momeni-Boroujeni A, Salazar P, Zheng T, Mensah N, Rijo I, Dogan S, et al. Rapid EGFR mutation detection using the Idylla platform: Single-institution experience of 1200 cases analyzed by an in-house developed pipeline and comparison with concurrent next-generation sequencing results. J Mol Diagn, 2021, 23(3):310–22., DOI 10.1016/j.jmoldx.2020.11.009 Arcila ME, Yang S-R, Momeni A, Mata DA, Salazar P, Chan R, et al. Ultrarapid EGFR mutation screening followed by comprehensive nextgeneration sequencing: A feasible, informative approach for lung carcinoma cytology specimens with a high success rate. JTO Clin Res Rep, 2020, 1(3): 100077., DOI 10.1016/j.jtocrr.2020.100077 De Luca C, Rappa AG, Gragnano G, Malapelle U, Troncone G, Barberis M. Idylla assay and next generation sequencing: An integrated EGFR mutational testing algorithm. J Clin Pathol, 2018, 71(8):745–50., DOI 10.1136/jclinpath-2018- 205197 Biocartis. Technical sheet Idylla™ ctEGFR mutation assay, 2019). Available: https://www.biocartis.com/sites/default/files/2021-03/Idylla_ctEGFR-RUO_Tech- Sheet.pdf, Accessed May 31, 2022). Lee Y, Clark EW, Milan MSD, Champagne C, Michael KS, Awad MM, et al. Turnaround time of plasma next-generation sequencing in thoracic oncology patients: A quality improvement analysis. JCO Precis Oncol, 2020, 4:1098–108., DOI 10.1200/PO.20.00121 Evrard SM, Taranchon-Clermont E, Rouquette I, Murray S, Dintner S, Nam-Apostolopoulos YC, et al. Multicenter evaluation of the fully automated PCR-based Idylla EGFR mutation assay on formalin-fixed, paraffin-embedded tissue of human lung cancer. J Mol Diagn, 2019, 21(6):1010–24., DOI 10.1016/j. jmoldx.2019.06.010 Ilie M, Butori C, Lassalle S, Heeke S, Piton N, Sabourin J-C, et al. Optimization of EGFR mutation detection by the fully-automated qPCR-based Idylla system on tumor tissue from patients with non-small cell lung cancer. Oncotarget, 2017, 8(61):103055–62., DOI 10.18632/oncotarget.21476 Malapelle U, Pisapia P, Iaccarino A, Barberis M, Bellevicine C, Brunnstrom H, et al. Predictive molecular Pathology in the time of coronavirus disease, COVID- 19, in Europe. J Clin Pathol, 2020, 74:391–5., DOI 10.1136/jclinpath-2020-206957 Grant J, Stanley A, Balbi K, Gerrard G, Bennett P. Performance evaluation of the Biocartis Idylla EGFR mutation test using pre-extracted DNA from a cohort of highly characterised mutation positive samples. J Clin Pathol, 2021, 75:241–9., DOI 10.1136/jclinpath–2020–20733810.1136/jclinpath-2020-207338 Park S, Holmes-Tisch AJ, Cho EY, Shim YM, Kim J, Kim HS, et al. Discordance of molecular biomarkers associated with epidermal growth factor receptor pathway between primary tumors and lymph node metastasis in non-small cell lung cancer. J Thorac Oncol, 2009, 4(7):809–15., DOI 10.1097/JTO. 0b013e3181a94af4 Chang S, Shim HS, Kim TJ, Choi YL, Kim WS, Shin DH, et al. Molecular biomarker testing for non-small cell lung cancer: Consensus statement of the Korean cardiopulmonary pathology study group. J Pathol Transl Med, 2021, 55(3): 181–91., DOI 10.4132/jptm.2021.03.23 Bordi P, Del Re M, Danesi R, Tiseo M. Circulating DNA in diagnosis and monitoring EGFR gene mutations in advanced non-small cell lung cancer. Transl Lung Cancer Res, 2015, 4(5):584–97., DOI 10.3978/j.issn.2218-6751.2015.08.09 Nkosi D, Casler VL, Syposs CR, Oltvai ZN. Utility of select gene mutation detection in tumors by the Idylla rapid multiplex PCR platform in comparison to next-generation sequencing. Genes, Basel,, 2022, 13(5):799., DOI 10.3390/ genes13050799 Gilson P, Saurel C, Salleron J, Husson M, Demange J, Merlin JL, et al. Evaluation of the Idylla ctEGFR mutation assay to detect EGFR mutations in plasma from patients with non-small cell lung cancers. Sci Rep, 2021, 11(1):10470., DOI 10. 1038/s41598-021-90091-z Petiteau C, Robinet-Zimmermann G, Riot A, Dorbeau M, Richard N, Blanc- Fournier C, et al. Contribution of the IdyllaTM system to improving the therapeutic care of patients with NSCLC through early screening of EGFR mutations. Curr Oncol, 2021, 28(6):4432–45., DOI 10.3390/curroncol28060376 Harrison PT, Vyse S, Huang PH. Rare epidermal growth factor receptor, EGFR, mutations in non-small cell lung cancer. Semin Cancer Biol, 2020, 61: 167–79., DOI 10.1016/j.semcancer.2019.09.015 Friedlaender A, Subbiah V, Russo A, Banna GL, Malapelle U, Rolfo C, et al. EGFR and HER2 exon 20 insertions in solid tumours: From biology to treatment. Nat Rev Clin Oncol, 2022, 19(1):51–69., DOI 10.1038/s41571-021-00558-1 Vigliar E, Malapelle U, Bellevicine C, de Luca C, Troncone G. Outsourcing cytological samples to a referral laboratory for EGFR testing in non-small cell lung cancer: Does theory meet practice? Cytopathology, 2015, 26(5):312–7., DOI 10.1111/cyt.12221 Yang DX, Khera R, Miccio JA, Jairam V, Chang E, Yu JB, et al. Prevalence of missing data in the National Cancer Database and association with overall survival. JAMA Netw Open, 2021, 4(3):e211793., DOI 10.1001/ jamanetworkopen.2021.1793 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610707 EP 1610721 DI 10.3389/pore.2023.1610707 PG 15 ER PT J AU Wang, Ch Wang, W Xu, R Xiang, J AF Wang, Changrong Wang, Wei Xu, Rujun Xiang, Jingjing TI Case report: Large-size intramuscular nodular fasciitis, a challenging histopathologic diagnosis confirmed by molecular detection of USP6 gene rearrangement: Case report and literature review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE nodular fasciitis; gene rearrangement; large-size; intramuscular; USP6 ID nodular fasciitis; gene rearrangement; large-size; intramuscular; USP6 AB The intramuscular subtype of nodular fasciitis (NF) is rare with lesions normally not more than 2 cmin size and characterized by pseudosarcomatousmorphology.We report a case of a 27-year-old man with a large-size intramuscular NF. The patient came for treatment complaining of an increasingly enlarged mass in the left upper arm for 4months. Magnetic resonance imaging (MRI) confirmed the presence of a well-defined tumor measuring 5 cm within the outer edge of themiddle humerus. Microscopically, the neoplasm was rich in fibroblasts and myofibroblasts in an interlaced pattern with high mitotic index and evident multinuclear giant cells. Erythrocyte extravasation was easily seen in the stroma. The tumor border was infiltrative. Immunohistochemically, the tumor cells were positive for smooth muscle actin (SMA) and negative for cytokeratin, desmin, H-Caldesmon, CD34, S100, ALK, and β-catenin. Fibrosarcoma was highly suspected by histopathological and immunohistochemical examination. Molecular detection demonstrated evidence of ubiquitin-specific peptidase 6 (USP6) gene rearrangement in this tumor. Based on the findings, the tumor was diagnosed as intramuscular NF. At 56 months after the initial surgery, the patient had recovered with no evidence of recurrence or metastasis. Large-size intramuscular NF is very rare and easily overdiagnosed as malignant tumor due to its obvious pseudosarcomatoid pathological features. USP6 gene rearrangement detection can effectively avoid this major misdiagnosis. C1 [Wang, Changrong] Zhejiang University School of Medicine, Affiliated Hangzhou First People’s Hospital, Department of Surgical PathologyHangzhou, Zhejiang, China. [Wang, Wei] Zhejiang University School of Medicine, Affiliated Hangzhou First People’s Hospital, Department of Surgical PathologyHangzhou, Zhejiang, China. [Xu, Rujun] Zhejiang University School of Medicine, Affiliated Hangzhou First People’s Hospital, Department of Surgical PathologyHangzhou, Zhejiang, China. [Xiang, Jingjing] Zhejiang University School of Medicine, Affiliated Hangzhou First People’s Hospital, Department of Surgical PathologyHangzhou, Zhejiang, China. RP Xiang, J (reprint author), Zhejiang University School of Medicine, Affiliated Hangzhou First People’s Hospital, Department of Surgical Pathology, Hangzhou, China. EM xiang_cell@163.com CR Michele REJ, Margaret MC, Barbara RE, ChristopherWR, Amber RS, Jin L, et al. Nodular fasciitis: A novel model of transient neoplasia induced by MYH9- USP6 gene fusion. Lab Invest, 2011, 91:1427–33., DOI 10.1038/labinvest. 2011.118 Chen J, Ye X, Li Y,Wei C, Zheng Q, Zhong P, et al. Chromosomal translocation involving USP6 gene in nodular fasciitis. Zhonghua Bing Li Xue Za Zhi, 2014, 43: 533–6. Oliveira AM, Chou MM. USP6-induced neoplasms: The biologic spectrum of aneurysmal bone cyst and nodular fasciitis. Hum Pathol, 2014, 45:1–11., DOI 10. 1016/j.humpath.2013.03.005 Shin C, Low I, Ng D, Oei P, Miles C, Symmans P. USP6 gene rearrangement in nodular fasciitis and histological mimics. Histopathology, 2016, 69:784–91., DOI 10. 1111/his.13011 Erber R, Agaimy A. Misses and near misses in diagnosing nodular fasciitis and morphologically related reactive myofibroblastic proliferations: Experience of a referral center with emphasis on frequency of USP6 gene rearrangements. Virchows Arch, 2018, 473:351–60., DOI 10.1007/s00428-018- 2350-0 Lu LL, Lao IW, Liu XH, Yu L, Wang J. Nodular fasciitis: A retrospective study of 272 cases from China with clinicopathologic and radiologic correlation. Ann Diagn Pathol, 2015, 19:180–5., DOI 10.1016/j.anndiagpath.2015.03.013 Jan CM, Cathleen M, Nitsana S, Paula WB, Arzu K. Intramuscular nodular fasciitis of the rectus abdominis muscle in an 11-year-old girl. Skeletal Radiol, 2013, 42:147–50., DOI 10.1007/s00256-012-1498-y Guo RF, Wang XK, Margaret MC, Yan A, Doris EW, Alyaa AI, et al. PPP6R3- USP6 amplification: Novel oncogenic mechanism in malignant nodular fasciitis. Genes Chromosomes Cancer, 2016, 55:640–9., DOI 10.1002/gcc.22366 Anandhi A, Vishnu PNR, Susan R, Rajesh NG. Giant intramuscular nodular fasciitis masquerading as soft tissue sarcoma with neural involvement-A case report. J Clin Diagn Res, 2016, 10:PD13–5., DOI 10.7860/JCDR/2016/ 18399.8012 Giuseppe F, Moira R, Eugenio C, Monica F, Monica S, Veltri S, et al. Intramuscular nodular fasciitis of the pectoralis major. Breast J, 2016, 22:122–3., DOI 10.1111/tbj.12532 Cahit K, Mehmet I, Ali KS, Aylin A. Intramuscular nodular fasciitis of the erector spinae muscle mimicking soft tissue sarcoma. Spine J, 2016, 16:e491–2., DOI 10.1016/j.spinee.2016.01.177 Sankappa PS, Bharathi KV, Ramachandra VB, Basavanandaswamy CH. Intra-muscular nodular fasciitis presenting as swelling in neck: Challenging entity for diagnosis. J Clin Diagn Res, 2014, 8:155–7., DOI 10.7860/JCDR/2014/ 6424.3909 Andre MO, Antonio RPA, Carrie YI, Fabiola M, Victoria D, Bae-Li H, et al. USP6 and CDH11 oncogenes identify the neoplastic cell in primary aneurysmal bone cysts and are absent in so-called secondary aneurysmal bone cysts. Am J Pathol, 2004, 165:1773–80., DOI 10.1016/S0002-9440(10)63432-3 Oliveira AM, Hsi BL, Weremowicz S, Rosenberg AE, Dal Cin P, Joseph N, et al. USP6, Tre2, fusion oncogenes in aneurysmal bone cyst. Cancer Res, 2004, 64: 1920–3., DOI 10.1158/0008-5472.can-03-2827 Andre MO, Antonio RP, Paola DC, Mark CG, Chen CJ, James RN, et al. Aneurysmal bone cyst variant translocations upregulate USP6 transcription by promoter swapping with the ZNF9, COL1A1, TRAP150, and OMD genes. Oncogene, 2005, 24:3419–26., DOI 10.1038/sj.onc.1208506 William RS,Marcello FF, Michele EJ,Margaret MC, Unni KK, Doris EW, et al. Frequency of USP6 rearrangements in myositis ossificans, Brown tumor, and cherubism: Molecular cytogenetic evidence that a subset of “myositis ossificanslike lesions” are the early phases in the formation of soft-tissue aneurysmal bone cyst. Skeletal Radiol, 2008, 37:321–7., DOI 10.1007/s00256-007-0442-z Ioannis P, Fredrik M, Richard L, Nils M. Fusion of the COL1A1 and USP6 genes in a benign bone tumor. Cancer Genet Cytogenet, 2008, 180:70–3., DOI 10.1016/j.cancergencyto.2007.09.017 Alan WL, Lashon MP, Laura Q, Daisy NR, Ying Y, Oliveira AM, et al. TRE17/ ubiquitin-specific protease 6, USP6, oncogene translocated in aneurysmal bone cyst blocks osteoblastic maturation via an autocrine mechanism involving bone morphogenetic protein dysregulation. J Biol Chem, 2010, 285:37111–20., DOI 10. 1074/jbc.M110.175133 Nimesh RP, John SAC, Elizabeth GD, Stephen FS, Jacquelyn R, Erica K, et al. USP6 activation in nodular fasciitis by promoter-swapping gene fusions. Mod Pathol, 2017, 30:1577–88., DOI 10.1038/modpathol.2017.78 S M, Michael M, Petr M, Nikola P, Zdenek K, Peter S, et al. Fibro-osseous pseudotumor of digits and myositis ossificans show consistent col1a1-USP6 rearrangement: A clinicopathological and genetic study of 27 cases. Hum Pathol, 2015, 88:39–47., DOI 10.1016/j.humpath.2019.02.009 Carter JM, Wang X, Dong J, Westendorf J, Chou MM, Oliveira AM. USP6 genetic rearrangements in cellular fibroma of tendon sheath. Mod Pathol, 2016, 29(8):865–9., DOI 10.1038/modpathol.2016.83 Uta F, Elise MB, David C, Joost MG. COL1A1 is a fusionpartner of USP6 in myositis ossificans- FISH analysis of six cases. Ann Diagn Pathol, 2018, 36:61–2., DOI 10.1016/j.anndiagpath.2018.06.009 Uta F, Sarah JS, Elise MB, Roberto T, Paul JD, et al. Fibro-osseous pseudotumor of digits-Expanding the spectrum of clonal transient neoplasms harboring USP6 rearrangement. Ann Diagn Pathol, 2018, 35:53–5., DOI 10.1016/ j.anndiagpath.2018.05.003 Elise MB, Astrid E, Katrien G, Rona CR, Jacky WJR, Ingrid CMG, et al. Myositis ossificans- Another condition with USP6 rearrangement, providing evidence of a relationship with nodular fasciitis and aneurysmal bone cyst. Ann Diagn Pathol, 2018, 34:56–9., DOI 10.1016/j.anndiagpath.2018.01.006 Judith ASJ, Steven CS, Kyle DP, Nilesh SG, Shaheen A, Shannon C, et al. Pseudosarcomatous myofibroblastic proliferations of the genitourinary tract are genetically different from nodular fasciitis and lack USP6, ROS1 and ETV6 gene rearrangements. Histopathology, 2018, 73:321–6., DOI 10.1111/his.13526 Sapi Z, LippaiPapp ZG, Hegyi L, Sapi J, Dezso K, et al. Nodular fasciitis: A comprehensive, time-correlated investigation of 17 cases. Mod Pathol, 2021, 34(12):2192–9., DOI 10.1038/s41379-021-00883-x Jason YKC, Zhen G, Eddy WYW, Siu KN, Michael CFT, Alexander CV. Lowgrade myofibroblastic sarcoma: A population-based study. Laryngoscope, 2017, 127:116–21., DOI 10.1002/lary.26146 Bharat R, Mahesh D, Nirmala AJ. Low-grade fibromyxoid sarcoma: A clinicopathologic study of 18 cases, including histopathologic relationship with sclerosing epithelioid fibrosarcoma in a subset of cases. Ann Diagn Pathol, 2011, 15: 303–11., DOI 10.1016/j.anndiagpath.2011.02.005 Matthias HMS, Robert K, Anja G, Gunhild M, Jochen G, Stefanie W, et al. Comparable transforming capacities and differential gene expression patterns of variant FUS/CHOP fusion transcripts derived from soft tissue liposarcomas. Oncogene, 2004, 23:6798–805., DOI 10.1038/sj.onc.1207840 Carlson JW, Fletcher CD. Immunohistochemistry for beta-catenin in the differential diagnosis of spindle cell lesions: Analysis of a series and review of the literature. Histopathology, 2007, 51:509–14., DOI 10.1111/j.1365-2559.2007.02794.x Montgomery EA, Meis JM. Nodular fasciitis. Its morphologic spectrum and immunohistochemical profile. Am J Surg Pathol, 1991, 15:942–8., DOI 10.1097/ 00000478-199110000-00004 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610785 EP 1610791 DI 10.3389/pore.2023.1610785 PG 7 ER PT J AU Zhang, Sh Xu, H Li, W Ji, J Jin, Q Chen, L Gan, Q Tao, Q Chai, Y AF Zhang, Shouhua Xu, Hongyan Li, Weiming Ji, Jianfeng Jin, Qifang Chen, Leifeng Gan, Qiang Tao, Qiang Chai, Yong TI MDM2 promotes cancer cell survival through regulating the expression of HIF-1α and pVHL in retinoblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HIF-1α; retinoblastoma; MDM2; pVHL; hypoxia ID HIF-1α; retinoblastoma; MDM2; pVHL; hypoxia AB Hypoxia is an important tumor feature and hypoxia-inducible factor 1 (HIF-1) is a master regulator of cell response to hypoxia. Mouse double minute 2 homolog (MDM2) promotes cancer cell survival in retinoblastoma (RB), with the underlying mechanism remaining elusive. In this study,we investigated the role of MDM2 and its relation to HIF-1α in RB. Expression analysis on primary human RB samples showed that MDM2 expression was positively correlated with that of HIF-1α while negatively correlated with von Hippel-Lindau protein (pVHL), the regulator of HIF-1α. In agreement, RB cells with MDM2 overexpression showed increased expression of HIF-1α and decreased expression of pVHL,while cells withMDM2 siRNA knockdown or MDM2-specific inhibitor showed the opposite effect under hypoxia. Further immuno-precipitation analysis revealed that MDM2 could directly interact with pVHL and promotes its ubiquitination and degradation, which consequently led to the increase of HIF-1α. Inhibition of MDM2 and/or HIF-1α with specific inhibitors induced RB cell death and decreased the stem cell properties of primary RB cells. Taken together, our study has shown that MDM2 promotes RB survival through regulating the expression of pVHL and HIF-1α, and targeting MDM2 and/or HIF-1α represents a potential effective approach for RB treatment. C1 [Zhang, Shouhua] Jiangxi Provincial Children’s Hospital, Department of General SurgeryNanchang, Jiangxi, China. [Xu, Hongyan] Jiangxi Provincial Children’s Hospital, Department of General SurgeryNanchang, Jiangxi, China. [Li, Weiming] Jiangxi Provincial Children’s Hospital, Department of General SurgeryNanchang, Jiangxi, China. [Ji, Jianfeng] Joint Support Forces of the Chinese People’s Liberation Army 908 Hospital, Department of UltrasoundNanchang, Jiangxi, China. [Jin, Qifang] Second Affiliated Hospital of Nanchang University, Department of General SurgeryNanchang, Jiangxi, China. [Chen, Leifeng] Second Affiliated Hospital of Nanchang University, Department of General SurgeryNanchang, Jiangxi, China. [Gan, Qiang] Jiangxi Provincial Children’s Hospital, Department of OphthalmologyNanchang, Jiangxi, China. [Tao, Qiang] Jiangxi Provincial Children’s Hospital, Department of General SurgeryNanchang, Jiangxi, China. [Chai, Yong] Jiangxi Provincial Children’s Hospital, Department of OphthalmologyNanchang, Jiangxi, China. RP Chai, Y (reprint author), Jiangxi Provincial Children’s Hospital, Department of Ophthalmology, Nanchang, China. EM yc3408954524@hotmail.com CR Rojanaporn D, Boontawon T, Chareonsirisuthigul T, Thanapanpanich O, Attaseth T, Saengwimol D, et al. Spectrum of germline RB1 mutations and clinical manifestations in retinoblastoma patients from Thailand. Mol Vis, 2018, 24:778–88. Dimaras H, Corson TW, Cobrinik D, White A, Zhao J, Munier FL, et al. Retinoblastoma Nat Rev Dis primers, 2015, 1(1):1–23. Burkhart DL, Sage J. Cellular mechanisms of tumour suppression by the retinoblastoma gene. Nat Rev Cancer, 2008, 8(9):671–82., DOI 10.1038/nrc2399 Dimaras H, Corson TW. Retinoblastoma, the visible CNS tumor: A review. J Neurosci Res, 2019, 97(1):29–44., DOI 10.1002/jnr.24213 Kaewkhaw R, Rojanaporn D. Retinoblastoma: Etiology, modeling, and treatment. Cancers, 2020, 12(8):2304., DOI 10.3390/cancers12082304 Freedman D, Wu L, Levine A. Functions of the MDM2 oncoprotein. Cell Mol Life Sci CMLS, 1999, 55(1):96–107., DOI 10.1007/s000180050273 Nieminen A-L, Qanungo S, Schneider EA, Jiang B-H, Agani FH. Mdm2 and HIF-1alpha interaction in tumor cells during hypoxia. J Cell Physiol, 2005, 204(2): 364–9., DOI 10.1002/jcp.20406 Qi DL, Cobrinik D. MDM2 but not MDM4 promotes retinoblastoma cell proliferation through p53-independent regulation of MYCN translation. Oncogene, 2017, 36(13):1760–9., DOI 10.1038/onc.2016.350 Xu XL, Fang Y, Lee TC, Forrest D, Gregory-Evans C, Almeida D, et al. Retinoblastoma has properties of a cone precursor tumor and depends upon conespecific MDM2 signaling. Cell, 2009, 137(6):1018–31., DOI 10.1016/j.cell.2009.03.051 Boutrid H, Pina Y, Cebulla CM, FeuerWJ, Lampidis TJ, Jockovich M-E, et al. Increased hypoxia following vessel targeting in a murine model of retinoblastoma. Invest Ophthalmol Vis Sci, 2009, 50(12):5537–43., DOI 10.1167/iovs.09-3702 Wang GL, Semenza GL. Purification and characterization of hypoxiainducible factor 1, p). J Biol Chem, 1995, 270(3):1230–7., DOI 10.1074/jbc.270.3.1230 Safran M, Kaelin WG. HIF hydroxylation and the mammalian oxygensensing pathway. J Clin Invest, 2003, 111(6):779–83., DOI 10.1172/JCI18181 Sudhakar J, Venkatesan N, Lakshmanan S, Khetan V, Krishnakumar S, Biswas J. Hypoxic tumor microenvironment in advanced retinoblastoma. Pediatr Blood Cancer, 2013, 60(10):1598–601., DOI 10.1002/pbc.24599 Tang Z,Ma H,Mao Y, Ai S, Zhang P, Nie C, et al. Identification of stemness in primary retinoblastoma cells by analysis of stem-cell phenotypes and tumorigenicity with culture and xenograft models. Exp Cel Res, 2019, 379(1): 110–8., DOI 10.1016/j.yexcr.2019.03.034 Park KS, Kim JH, Shin HW, Chung KS, Im DS, Lim JH, et al. E2-EPF UCP regulates stability and functions of missense mutant pVHL via ubiquitin mediated proteolysis. BMC Cancer, 2015, 15:800., DOI 10.1186/s12885-015-1786-8 Hu K, Fu M, Wang J, Luo S, Barreto M, Singh R, et al. HSV-2 infection of human genital epithelial cells upregulates TLR9 expression through the SP1/JNK signaling pathway. Front Immunol, 2020, 11:356., DOI 10.3389/fimmu.2020.00356 Hu K, McKay PF, Samnuan K, Najer A, Blakney AK, Che J, et al. Presentation of antigen on extracellular vesicles using transmembrane domains from viral glycoproteins for enhanced immunogenicity. J Extracellular Vesicles, 2022, 11(3):e12199., DOI 10.1002/jev2.12199 Zhang S, Li N, Sheng Y, ChenW, Ma Q, Yu X, et al. Hepatitis B virus induces sorafenib resistance in liver cancer via upregulation of cIAP2 expression. Infect Agents Cancer, 2021, 16(1):20–11., DOI 10.1186/s13027-021-00359-2 Wolf ER, Mabry AR, Damania B, Mayo LD. Mdm2-mediated neddylation of pVHL blocks the induction of antiangiogenic factors. Oncogene, 2020, 39(29): 5228–39., DOI 10.1038/s41388-020-1359-4 Nomura K, Klejnot M, Kowalczyk D, Hock AK, Sibbet GJ, Vousden KH, et al. Structural analysis of MDM2 RING separates degradation from regulation of p53 transcription activity. Nat Struct Mol Biol, 2017, 24(7):578–87., DOI 10.1038/ nsmb.3414 Geyer RK, Yu ZK, Maki CG. The MDM2 RING-finger domain is required to promote p53 nuclear export. Nat Cel Biol, 2000, 2(9):569–73., DOI 10.1038/ 35023507 Itahana K, Mao H, Jin A, Itahana Y, Clegg HV, Lindstrom MS, et al. Targeted inactivation of Mdm2 RING finger E3 ubiquitin ligase activity in the mouse reveals mechanistic insights into p53 regulation. Cancer cell, 2007, 12(4):355–66., DOI 10.1016/j.ccr. 2007.09.007 Batlle E, Clevers H. Cancer stem cells revisited. Nat Med, 2017, 23(10): 1124–34., DOI 10.1038/nm.4409 Wienken M, Dickmanns A, Nemajerova A, Kramer D, Najafova Z, Weiss M, et al. MDM2associates with polycomb repressor complex 2 and enhances stemnesspromoting chromatin modifications independent of p53. Mol Cel, 2016, 61(1): 68–83., DOI 10.1016/j.molcel.2015.12.008 Peng G, Liu Y. Hypoxia-inducible factors in cancer stem cells and inflammation. Trends Pharmacological Sciences, 2015, 36(6):374–83., DOI 10. 1016/j.tips.2015.03.003 Najafi M, Farhood B, Mortezaee K, Kharazinejad E, Majidpoor J, Ahadi R. Hypoxia in solid tumors: A key promoter of cancer stem cell, CSC, resistance. J Cancer Res Clin Oncol, 2020, 146(1):19–31., DOI 10.1007/s00432- 019-03080-1 Kim H, Lin Q, Glazer PM, Yun Z. The hypoxic tumor microenvironment in vivo selects the cancer stem cell fate of breast cancer cells. Breast Cancer Res, 2018, 20(1):16–5., DOI 10.1186/s13058-018-0944-8 Hayashi Y, Yokota A, Harada H, Huang G. Hypoxia/pseudohypoxiamediated activation of hypoxia-inducible factor-1α in cancer. Cancer Sci, 2019, 110(5):1510–7., DOI 10.1111/cas.13990 Ryan HE, Poloni M, McNulty W, Elson D, Gassmann M, Arbeit JM, et al. Hypoxia-inducible factor-1alpha is a positive factor in solid tumor growth. Cancer Res, 2000, 60(15):4010–5. Jaakkola P, Mole DR, Tian Y-M, Wilson MI, Gielbert J, Gaskell SJ, et al. Targeting of HIF-α to the von Hippel-Lindau ubiquitylation complex by O2- regulated prolyl hydroxylation. Science, 2001, 292(5516):468–72., DOI 10.1126/ science.1059796 Corrado C, Fontana S. Hypoxia and HIF signaling: One Axis with divergent effects. Int J Mol Sci, 2020, 21(16):5611., DOI 10.3390/ijms21165611 Kietzmann T, Mennerich D, Dimova EY. Hypoxia-inducible factors, HIFs, and phosphorylation: Impact on stability, localization, and transactivity. Front Cel Dev Biol, 2016, 4:11., DOI 10.3389/fcell.2016.00011 Xiao Y, Peng H, Hong C, Chen Z, Deng X,Wang A, et al. PDGF promotes the warburg effect in pulmonary arterial smooth muscle cells via activation of the PI3K/ AKT/mTOR/HIF-1α signaling pathway. Cell Physiol Biochem, 2017, 42(4): 1603–13., DOI 10.1159/000479401 Zhang Z, Yao L, Yang J, Wang Z, Du G. PI3K/Akt and HIF‑1 signaling pathway in hypoxia‑ischemia, Review). Mol Med Rep, 2018, 18(4):3547–54., DOI 10.3892/mmr.2018. 9375 Shvetsova AN, Mennerich D, Keratar JM, Hiltunen JK, Kietzmann T. Nonelectron transfer chain mitochondrial defects differently regulate HIF-1α degradation and transcription. Redox Biol, 2017, 12:1052–61., DOI 10.1016/j. redox.2017.05.003 Semenza GL. Hypoxia-inducible factors: Coupling glucose metabolism and redox regulation with induction of the breast cancer stem cell phenotype. EMBO J, 2017, 36(3):252–9., DOI 10.15252/embj.201695204 Lacher SE, Levings DC, Freeman S, Slattery M. Identification of a functional antioxidant response element at the HIF1A locus. Redox Biol, 2018, 19:401–11., DOI 10.1016/j.redox.2018.08.014 Jeong H, Kim S, Hong B-J, Lee C-J, Kim Y-E, Bok S, et al. Tumor-associated macrophages enhance tumor hypoxia and aerobic glycolysis. Cancer Res, 2019, 79(4):795–806., DOI 10.1158/0008-5472.CAN-18-2545 Bohlman S, Manfredi JJ. p53-independent effects of Mdm2. Mutant p53 and MDM2 in Cancer. Subcell Biochem, 2014, 235–46., DOI 10.1007/978-94-017-9211- 0_13 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610801 EP 1610811 DI 10.3389/pore.2023.1610801 PG 11 ER PT J AU Liao, X Chen, J Luo, D Luo, B Huang, W Xie, W AF Liao, XiaoLi Chen, Junming Luo, Dongcheng Luo, Baohua Huang, Wenfeng Xie, Weimin TI Prognostic value of long non-coding RNA MALAT1 in hepatocellular carcinoma: A study based on multi-omics analysis and RT-PCR validation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; hepatocellular carcinoma; mutation; DNA methylation; metastasis associated lung adenocarcinoma transcript 1 (MALAT1); interacting proteins; competing endogenous RNAs (ceRNAs); multi-omics ID prognosis; hepatocellular carcinoma; mutation; DNA methylation; metastasis associated lung adenocarcinoma transcript 1 (MALAT1); interacting proteins; competing endogenous RNAs (ceRNAs); multi-omics AB Background: This study aimed to explore the relationship between MALAT1 and the prognosis of patients with hepatocellular carcinoma (HCC). Methods: We constructed a MALAT1 protein-protein interaction network using the STRING database and a network of competing endogenous RNAs (ceRNAs) using the StarBase database. Using data from the GEPIA2 database, we studied the association between genes in these networks and survival of patients with HCC. The potential mechanisms underlying the relationship between MALAT1 and HCC prognosis were studied using combined data from RNA sequencing, DNA methylation, and somatic mutation data from The Cancer Genome Atlas (TCGA) liver cancer cohort. Tumor tissues and 19 paired adjacent non-tumor tissues (PANTs) from HCC patients who underwent radical resection were analyzed for MALAT1 mRNA levels using real-time PCR, and associations of MALAT1 expression with clinicopathological features or prognosis of patients were analyzed using log-rank test and Gehan-Breslow-Wilcoxon test. Results: Five interacting proteins and five target genes of MALAT1 in the ceRNA network significantly correlated with poor survival of patients with HCC (p < 0.05). High MALAT1 expression was associated with mutations in two genes leading to poor prognosis and may upregulate some prognostic risk genes through methylation. MALAT1 was significantly co-expressed with various signatures of genes involved in HCC progression, including the cell cycle, DNA damage repair, mismatch repair, homologous recombination, molecular cancer m6A, exosome, ferroptosis, infiltration of lymphocyte (p < 0.05). The expression of MALAT1 was markedly upregulated in HCC tissues compared with PANTs. In Kaplan-Meier analysis, patients with high MALAT1 expression had significantly shorter progression-free survival (PFS) (p = 0.033) and overall survival (OS) (p = 0.023) than those with low MALAT1 expression. Median PFS was 19.2 months for patients with high MALAT1 expression and 52.8 months for patients with low expression, while the corresponding median OS was 40.5 and 78.3 months. In subgroup analysis of patients with vascular invasion, cirrhosis, and HBsAg positive or AFP positive, MALAT1 overexpression was significantly associated with shorter PFS and OS. Models for predicting PFS and OS constructed based on MALAT1 expression and clinicopathological features had moderate predictive power, with areas under the receiver operating characteristic curves of 0.661–0.731. Additionally, MALAT1 expression level was significantly associated with liver cirrhosis, vascular invasion, and tumor capsular infiltration (p < 0.05 for all). Conclusion: MALAT1 is overexpressed in HCC, and higher expression is associated with worse prognosis. MALAT1 mRNA level may serve as a prognostic marker for patients with HCC after hepatectomy. C1 [Liao, XiaoLi] Guangxi Medical University Cancer Hospital, Department of First ChemotherapyNanning, China. [Chen, Junming] Fujian Medical University Union Hospital, Department of Medical OncologyFuzhou, China. [Luo, Dongcheng] Guangxi Medical University Cancer Hospital, Department of First ChemotherapyNanning, China. [Luo, Baohua] Jiangbin Hospital, Department of GastroenterologyNanning, China. [Huang, Wenfeng] Second Affiliated Hospital of Guangxi Medical University, Department of Medical OncologyNanning, China. [Xie, Weimin] Guangxi Medical University Cancer Hospital, Department of First ChemotherapyNanning, China. RP Xie, W (reprint author), Guangxi Medical University Cancer Hospital, Department of First Chemotherapy, Nanning, China. EM xieweimin@gxmu.edu.cn CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/ caac.21492 Sun D, Cao M, Li H, He S, Chen W. Cancer burden and trends in China: A review and comparison with Japan and South Korea. Chin J Cancer Res, 2020, 32(2):129–39., DOI 10.21147/j.issn.1000-9604.2020.02.01 Chen LT, Martinelli E, Cheng AL, Pentheroudakis G, Qin S, Bhattacharyya GS, et al. Pan-asian adapted ESMO clinical practice guidelines for the management of patients with intermediate and advanced/relapsed hepatocellular carcinoma: A TOS-ESMO initiative endorsed by CSCO, ISMPO, JSMO, KSMO, MOS and SSO. Ann Oncol, 2020, 31(3):334–51., DOI 10.1016/j.annonc.2019.12.001 Marrero JA, Kulik LM, Sirlin CB, Zhu AX, Finn RS, Abecassis MM, et al. Diagnosis, staging, and management of hepatocellular carcinoma: 2018 practice guidance by the American association for the study of liver diseases. Hepatology, 2018, 68(2):723–50., DOI 10.1002/hep.29913 Hilmi M, Neuzillet C, Calderaro J, Lafdil F, Pawlotsky JM, Rousseau B. Angiogenesis and immune checkpoint inhibitors as therapies for hepatocellular carcinoma: Current knowledge and future research directions. J Immunother Cancer, 2019, 7(1):333., DOI 10.1186/s40425-019-0824-5 Pinter M, Scheiner B, Peck-Radosavljevic M. Immunotherapy for advanced hepatocellular carcinoma: A focus on special subgroups. Gut, 2021, 70(1):204–14., DOI 10.1136/gutjnl-2020-321702 Makary MS, Khandpur U, Cloyd JM, Mumtaz K, Dowell JD. Locoregional therapy approaches for hepatocellular carcinoma: Recent advances and management strategies. Cancers, Basel,, 2020, 12(7):1914., DOI 10.3390/ cancers12071914 Yarchoan M, Agarwal P, Villanueva A, Rao S, Dawson LA, Llovet JM, et al. Recent developments and therapeutic strategies against hepatocellular carcinoma. Cancer Res, 2019, 79(17):4326–30., DOI 10.1158/0008-5472.Can-19-0803 Vogel A, Cervantes A, Chau I, Meyer T, Llovet JM, Ricke J, et al. Correction to: "Hepatocellular carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up". Ann Oncol, 2019, 30(5):871–3., DOI 10.1093/annonc/ mdy510 Pinero F, Dirchwolf M, Pessoa MG. Biomarkers in hepatocellular carcinoma: Diagnosis, prognosis and treatment response assessment. Cells, 2020, 9(6):1370., DOI 10.3390/cells9061370 Huang Z, Zhou JK, Peng Y, He W, Huang C. The role of long noncoding RNAs in hepatocellular carcinoma. Mol Cancer, 2020, 19(1):77., DOI 10.1186/ s12943-020-01188-4 Forrest ME, Khalil AM. Review: Regulation of the cancer epigenome by long non-coding RNAs. Cancer Lett, 2017, 407:106–12., DOI 10.1016/j.canlet.2017.03.040 Lee JT. Epigenetic regulation by long noncoding RNAs. Science, 2012, 338(6113):1435–9., DOI 10.1126/science.1231776 Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA hypothesis: The rosetta stone of a hidden RNA language? Cell, 2011, 146(3):353–8., DOI 10.1016/j. cell.2011.07.014 Huang W, Li H, Yu Q, Xiao W, Wang DO. LncRNA-mediated DNA methylation: An emerging mechanism in cancer and beyond. J Exp Clin Cancer Res, 2022, 41(1):100., DOI 10.1186/s13046-022-02319-z He S, Chen J, Gao H, Yang G, Zhang F, Ding Y, et al. Extended transcriptome analysis reveals genome-wide lncRNA-mediated epigenetic dysregulation in colorectal cancer. Comput Struct Biotechnol J, 2020, 18:3507–17., DOI 10.1016/j. csbj.2020.11.004 Sun Y, Ma L. New insights into long non-coding RNAMALAT1 in cancer and metastasis. Cancers, Basel,, 2019, 11(2):216., DOI 10.3390/cancers11020216 Lei L, Chen J, Huang J, Lu J, Pei S, Ding S, et al. Functions and regulatory mechanisms of metastasis-associated lung adenocarcinoma transcript 1. J Cel Physiol, 2018, 234(1):134–51., DOI 10.1002/jcp.26759 Wang S, Liu X. The UCSCXenaTools R package: A toolkit for accessing genomics data from UCSC xena platform, from cancer multi-omics to single-cell RNA-seq. J Open Source Softw, 2019, 4(40):1627., DOI 10.21105/joss.01627 Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T, method. Nat Protoc, 2008, 3(6):1101–8., DOI 10.1038/nprot.2008.73 Ji P, Diederichs S,Wang W, Boing S,Metzger R, Schneider PM, et al. MALAT- 1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene, 2003, 22(39):8031–41., DOI 10. 1038/sj.onc.1206928 Cao X, Zhao R, Chen Q, Zhao Y, Zhang B, Zhang Y, et al. MALAT1 might be a predictive marker of poor prognosis in patients who underwent radical resection of middle thoracic esophageal squamous cell carcinoma. Cancer Biomark, 2015, 15(6):717–23., DOI 10.3233/cbm-150513 Liu M, Yang P, Mao G, Deng J, Peng G, Ning X, et al. Long non-coding RNA MALAT1 as a valuable biomarker for prognosis in osteosarcoma: A systematic review and meta-analysis. Int J Surg, 2019, 72:206–13., DOI 10.1016/j.ijsu.2019. 11.004 Lai MC, Yang Z, Zhou L, Zhu Q, Xie H, Zhang F, et al. Long non-coding RNA MALAT-1 overexpression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med Oncol, 2012, 29(3):1810–6., DOI 10.1007/s12032- 011-0004-z Sonohara F, Inokawa Y, Hayashi M, Yamada S, Sugimoto H, Fujii T, et al. Prognostic value of long non-coding RNA HULC and MALAT1 following the curative resection of hepatocellular carcinoma. Sci Rep, 2017, 237(1):16142., DOI 10. 1038/s41598-017-16260-1 Badalamenti G, Barraco N, Incorvaia L, Galvano A, Fanale D, Cabibi D, et al. Are long noncoding RNAs new potential biomarkers in gastrointestinal stromal tumors, GISTs)? The role of H19 and MALAT1. J Oncol, 2019, 2019:5458717., DOI 10.1155/2019/5458717 Ye ZL, Huang Y, Li LF, Zhu H, Gao H, Liu H, et al. Argonaute 2 promotes angiogenesis via the PTEN/VEGF signaling pathway in human hepatocellular carcinoma. Acta Pharmacol Sin, 2015, 36(10):1237–45., DOI 10.1038/aps.2015.18 Yang Y, Mei Q. Accumulation of AGO2 facilitates tumorigenesis of human hepatocellular carcinoma. Biomed Res Int, 2020, 2020:1631843., DOI 10.1155/2020/ 1631843 Hu J, Cai D, Zhao Z, Zhong GC, Gong J. Suppression of heterogeneous nuclear ribonucleoprotein C inhibit hepatocellular carcinoma proliferation, migration, and invasion via ras/MAPK signaling pathway. Front Oncol, 2021, 11:659676., DOI 10.3389/fonc.2021.659676 Zhou J, Che J, Xu L, Yang W, Li Y, ZhouW, et al. Enhancer of zeste homolog 2 promotes hepatocellular cancer progression and chemoresistance by enhancing protein kinase B activation through microRNA-381-mediated SET domain bifurcated 1. Bioengineered, 2022, 13(3):5737–55., DOI 10.1080/21655979.2021. 2023792 Ding H, Liu J, Wang C, Su Y. NONO promotes hepatocellular carcinoma progression by enhancing fatty acids biosynthesis through interacting with ACLY mRNA. Cancer Cel Int, 2020, 20:425., DOI 10.1186/s12935-020-01520-4 Lei S, Zhang B, Huang L, Zheng Z, Xie S, Shen L, et al. SRSF1 promotes the inclusion of exon 3 of SRA1 and the invasion of hepatocellular carcinoma cells by interacting with exon 3 of SRA1pre-mRNA. Cell Death Discov, 2021, 7(1):117., DOI 10.1038/s41420-021-00498-w Malakar P, Shilo A, Mogilevsky A, Stein I, Pikarsky E, Nevo Y, et al. Long noncoding RNA MALAT1 promotes hepatocellular carcinoma development by SRSF1 upregulation and mTOR activation. Cancer Res, 2017, 77(5):1155–67., DOI 10.1158/0008-5472.Can-16-1508 Zhang P, Ha M, Li L, Huang X, Liu C. MicroRNA-3064-5p sponged by MALAT1 suppresses angiogenesis in human hepatocellular carcinoma by targeting the FOXA1/CD24/Src pathway. Faseb j., 2020, 34(1):66–81., DOI 10.1096/fj. 201901834R Peng N, He J, Li J, Huang H, Huang W, Liao Y, et al. Long noncoding RNA MALAT1 inhibits the apoptosis and autophagy of hepatocellular carcinoma cell by targeting the microRNA-146a/PI3K/Akt/mTOR axis. Cancer Cel Int, 2020, 20:165., DOI 10.1186/s12935-020-01231-w Chen F, Zhong Z, Tan HY, Guo W, Zhang C, Cheng CS, et al. Suppression of lncRNA MALAT1 by betulinic acid inhibits hepatocellular carcinoma progression by targeting IAPs via miR-22-3p. Clin Transl Med, 2020, 10(6):e190., DOI 10.1002/ ctm2.190 Chen L, Yao H, Wang K, Liu X. Long non-coding RNA MALAT1 regulates ZEB1 expression by sponging miR-143-3p and promotes hepatocellular carcinoma progression. J Cel Biochem., 2017, 118(12):4836–43., DOI 10.1002/jcb.26158 Liu S, Qiu J, He G, Liang Y,Wang L, Liu C, et al. LncRNA MALAT1 acts as a miR-125a-3p sponge to regulate FOXM1 expression and promote hepatocellular carcinoma progression. J Cancer, 2019, 10(26):6649–59., DOI 10.7150/jca.29213 Gillespie M, Jassal B, Stephan R, Milacic M, Rothfels K, Senff-Ribeiro A, et al. The reactome pathway knowledgebase 2022. Nucleic Acids Res, 2022, 50(D1): D687–D692., DOI 10.1093/nar/gkab1028 Varallo-Rodriguez C, Freyer CW, Jr, Ontiveros EP, Griffiths EA, Wang ES, Wetzler M. Bosutinib for the treatment of philadelphia chromosome-positive leukemias. Expert Opin Orphan Drugs, 2015, 3(5):599–608., DOI 10.1517/21678707. 2015.1036027 Celia-Terrassa T, Liu DD, Choudhury A, Hang X, Wei Y, Zamalloa J, et al. Normal and cancerous mammary stem cells evade interferon-induced constraint through the miR-199a-LCOR axis. Nat Cel Biol., 2017, 19(6):711–23., DOI 10.1038/ ncb3533 Vogelsang TLR, Schmoeckel E, Kuhn C, Blankenstein T, Temelkov M, Heidegger H, et al. Regulation of LCoR and RIP140 expression in cervical intraepithelial neoplasia and correlation with CIN progression and dedifferentiation. J Cancer Res Clin Oncol, 2020, 146(7):1847–55., DOI 10.1007/ s00432-020-03178-x Yamamoto N, Nishikawa R, Chiyomaru T, Goto Y, Fukumoto I, Usui H, et al. The tumor-suppressive microRNA-1/133a cluster targets PDE7A and inhibits cancer cell migration and invasion in endometrial cancer. Int J Oncol, 2015, 47(1):325–34., DOI 10.3892/ijo.2015.2986 Lv P, Yang S, Liu W, Qin H, Tang X,Wu F, et al. Circulating plasma lncRNAs as novel markers of EGFR mutation status and monitors of epidermal growth factor receptor-tyrosine kinase inhibitor therapy. Thorac Cancer, 2020, 11(1):29–40., DOI 10.1111/1759-7714.13216 Ak Aksoy S, Mutlu M, Tunca B, Kocaeli H, Taskapilioglu MO, Bekar A, et al. Coexistence of TERT C228T mutation and MALAT1 dysregulation in primary glioblastoma: New prognostic and therapeutic targets. Neurol Res, 2021, 43(11): 916–25., DOI 10.1080/01616412.2021.1948738 Zhan P, Ji YN, Yu LK. TP53 mutation is associated with a poor outcome for patients with hepatocellular carcinoma: Evidence from a meta-analysis. Hepatobiliary Surg Nutr, 2013, 2(5):260–5., DOI 10.3978/j.issn.2304-3881.2013. 07.06 Tischoff I, Tannapfe A. DNA methylation in hepatocellular carcinoma. World J Gastroenterol, 2008, 14(11):1741–8., DOI 10.3748/wjg.14.1741 Liu A, Wu Q, Peng D, Ares I, Anadon A, Lopez-Torres B, et al. A novel strategy for the diagnosis, prognosis, treatment, and chemoresistance of hepatocellular carcinoma: DNA methylation. Med Res Rev, 2020, 40(5): 1973–2018., DOI 10.1002/med.21696 Dai X, Liu L, Liang Z, Guo K, Xu S, Wang H. Silencing of lncRNA MALAT1 inhibits cell cycle progression via androgen receptor signaling in prostate cancer cells. Pathol Res Pract, 2019, 215(4):712–21., DOI 10.1016/j.prp. 2019.01.011 Tripathi V, Shen Z, Chakraborty A, Giri S, Freier SM, Wu X, et al. Long noncoding RNA MALAT1 controls cell cycle progression by regulating the expression of oncogenic transcription factor B-MYB. Plos Genet, 2013, 9(3): e1003368., DOI 10.1371/journal.pgen.1003368 Samimi H, Haghpanah V, Irani S, Arefian E, Sohi AN, Fallah P, et al. Transcript-level regulation of MALAT1-mediated cell cycle and apoptosis genes using dual MEK/Aurora kinase inhibitor "BI-847325" on anaplastic thyroid carcinoma. Daru., 2019, 27(1):1–7., DOI 10.1007/s40199-018-0231-3 Huang J, Lin C, Dong H, Piao Z, Jin C, Han H, et al. Targeting MALAT1 induces DNA damage and sensitize non-small cell lung cancer cells to cisplatin by repressing BRCA1. Cancer Chemother Pharmacol, 2020, 86(5): 663–72., DOI 10.1007/s00280-020-04152-7 Hu Y, Lin J, Fang J, Fang H, Li C, Chen W, et al. Targeting the MALAT1/ PARP1/LIG3 complex induces DNA damage and apoptosis in multiple myeloma. Leukemia, 2018, 32(10):2250–62., DOI 10.1038/s41375-018-0104-2 Hardin H, Helein H,Meyer K, Robertson S, Zhang R, Zhong W, et al. Thyroid cancer stem-like cell exosomes: Regulation of EMT via transfer of lncRNAs. Lab Invest, 2018, 98(9):1133–42., DOI 10.1038/s41374-018-0065-0 Klinge CM. Non-coding RNAs in breast cancer: Intracellular and intercellular communication. Noncoding RNA, 2018, 4(4):40., DOI 10.3390/ ncrna4040040 Liang Z, Wu Q, Wang H, Tan J, Gou Y, Cao Y, et al. Silencing of lncRNA MALAT1 facilitates erastin-induced ferroptosis in endometriosis through miR-145- 5p/MUC1 signaling. Cel Death Discov, 2022, 8(1):190., DOI 10.1038/s41420-022- 00975-w Wang Z, Wang X, Zhang T, Su L, Liu B, Zhu Z, et al. LncRNA MALAT1 promotes gastric cancer progression via inhibiting autophagic flux and inducing fibroblast activation. Cell Death Dis, 2021, 12(4):368., DOI 10.1038/ s41419-021-03645-4 Hou ZH, Xu XW, Fu XY, Zhou LD, Liu SP, Tan DM. Long non-coding RNA MALAT1 promotes angiogenesis and immunosuppressive properties of HCC cells by sponging miR-140. Am J Physiol Cel Physiol., 2020, 318(3):C649–c663., DOI 10. 1152/ajpcell.00510.2018 Chang HL, Bamodu OA, Ong JR, Lee WH, Yeh CT, Tsai JT. Targeting the epigenetic non-coding RNA MALAT1/wnt signaling Axis as a therapeutic approach to suppress stemness and metastasis in hepatocellular carcinoma. Cells, 2020, 9(4):1020., DOI 10.3390/cells9041020 Malakar P, Stein I, Saragovi A, Winkler R, Stern-Ginossar N, Berger M, et al. Long noncoding RNA MALAT1 regulates cancer glucose metabolism by enhancing mTOR-mediated translation of TCF7L2. Cancer Res, 2019, 79(10):2480–93., DOI 10. 1158/0008-5472.Can-18-1432 He B, Peng F, LiW, Jiang Y. Interaction of lncRNA-MALAT1 and miR- 124 regulates HBx-induced cancer stem cell properties in HepG2 through PI3K/Akt signaling. J Cel Biochem., 2019, 120(3):2908–18., DOI 10.1002/jcb. 26823 Chen S, Wang G, Tao K, Cai K, Wu K, Ye L, et al. Long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 cooperates with enhancer of zeste homolog 2 to promote hepatocellular carcinoma development by modulating the microRNA-22/Snail family transcriptional repressor 1 axis. Cancer Sci, 2020, 111(5):1582–95., DOI 10.1111/cas.14372 Fattovich G, Stroffolini T, Zagni I, Donato F. Hepatocellular carcinoma in cirrhosis: Incidence and risk factors. Gastroenterology, 2004, 127(5):S35–50., DOI 10. 1053/j.gastro.2004.09.014 Yuan LT, Chang JH, Lee HL, Yang YC, Su SC, Lin CL, et al. Genetic variants of lncRNA MALAT1 exert diverse impacts on the risk and clinicopathologic characteristics of patients with hepatocellular carcinoma. J Clin Med, 2019, 8(9):1406., DOI 10.3390/jcm8091406 Toraih EA, Ellawindy A, Fala SY, Al Ageeli E, Gouda NS, Fawzy MS, et al. Oncogenic long noncoding RNA MALAT1 and HCV-related hepatocellular carcinoma. Biomed Pharmacother, 2018, 102:653–69., DOI 10.1016/j.biopha.2018. 03.105 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610808 EP 1610833 DI 10.3389/pore.2022.1610808 PG 26 ER PT J AU Fan, Z Liu, B Shang, P AF Fan, Zizheng Liu, Bing Shang, Peizhong TI Development and validation of a nomogram prediction model based on albumin-to-alkaline phosphatase ratio for predicting the prognosis of gallbladder carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; nomogram; gallbladder carcinoma; independent risk factors; AAPR ID prognosis; nomogram; gallbladder carcinoma; independent risk factors; AAPR AB Gallbladder carcinoma (GBC) is a rare biliary tract cancer with a high recurrence rate and a poor prognosis. Albumin-alkaline phosphatase ratio (AAPR) has been demonstrated to be a prognostic predictor for several cancers, but its predictive value for GBC patients remains unknown. The aim of this study was to investigate the predictive role of AAPR in GBC patients and to develop a novel nomogram prediction model for GBC patients. We retrospectively collected data from 80 patients who underwent surgery at the Hospital of 81st Group Army PLA as a training cohort. Data were collected from 70 patients with the same diagnosis who underwent surgery at the First Affiliated Hospital of Hebei North University as an external verification cohort. The optimal cut-off value of AAPR was determined using X-tile software. A nomogram for the overall survival (OS) based on multivariate Cox regression analysis was developed and validated using calibration curves, Harrell’s concordance index, the receiver operating characteristic curves, and decisive curve analyses. The optimal cut-off value of AAPR was .20. Univariate and multivariate Cox regression analyses demonstrated that BMI (p = .043), R0 resection (p = .001), TNM stage (p = .005), and AAPR (p = .017) were independent risk factors for GBC patients. In terms of consistency, discrimination, and net benefit, the nomogram incorporating these four independent risk factors performed admirably. AAPR is an independent predictor of GBC patients undergoing surgery, and a novel nomogram prediction model based on AAPR showed superior predictive ability. C1 [Fan, Zizheng] Hebei North University, Department of Graduate SchoolZhangjiakou, China. [Liu, Bing] The Hospital of 81st Group Army PLA, Department of Hepatobiliary SurgeryZhangjiakou, China. [Shang, Peizhong] The Hospital of 81st Group Army PLA, Department of Hepatobiliary SurgeryZhangjiakou, China. RP Shang, P (reprint author), The Hospital of 81st Group Army PLA, Department of Hepatobiliary Surgery, Zhangjiakou, China. EM spz251@163.com CR ChenW, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin, 2016, 66:115–32., DOI 10.3322/caac.21338 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Kakaei F, Beheshtirouy S, Nejatollahi SM, Zarrintan S, Mafi MR. Surgical treatment of gallbladder carcinoma: A critical review. Updates Surg, 2015, 67: 339–51., DOI 10.1007/s13304-015-0328-x Kanthan R, Senger JL, Ahmed S, Kanthan SC. Gallbladder cancer in the 21st century. J Oncol, 2015, 2015:967472., DOI 10.1155/2015/967472 Chan AW, Chan SL, Mo FK, Wong GL, Wong VW, Cheung YS, et al. Albumin-to-alkaline phosphatase ratio: A novel prognostic index for hepatocellular carcinoma. Dis Markers, 2015, 2015:564057., DOI 10.1155/ 2015/564057 Tan P, Xie N, Ai J, Xu H, Xu H, Liu L, et al. The prognostic significance of Albumin-to-Alkaline Phosphatase Ratio in upper tract urothelial carcinoma. Sci Rep, 2018, 8:12311., DOI 10.1038/s41598-018-29833-5 Zhang L, Zhang H, Yue D, WeiW, Chen Y, Zhao X, et al. The prognostic value of the preoperative albumin to alkaline phosphatase ratio in patients with non-small cell lung cancer after surgery. Thorac Cancer, 2019, 10:1581–9., DOI 10.1111/1759- 7714.13107 Zhang F, Lu S, Tian M, Hu K, Chen R, Zhang B, et al. Albumin-to-Alkaline phosphatase ratio is an independent prognostic indicator in combined hepatocellular and cholangiocarcinoma. J Cancer, 2020, 11:5177–86., DOI 10.7150/jca.45633 Li S, Lu S, Liu X, Chen X. Association between the pretreatment albumin-toalkaline phosphatase ratio and clinical outcomes in patients with bladder cancer treated with radical cystectomy: A retrospective cohort study. Front Oncol, 2021, 11:664392., DOI 10.3389/fonc.2021.664392 Camp RL, Dolled-Filhart M, Rimm DL. X-Tile: A new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res, 2004, 10:7252–9., DOI 10.1158/1078-0432.CCR-04-0713 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018, 68:394–424., DOI 10.3322/caac.21492 Kasumova GG, Tabatabaie O, Najarian RM, Callery MP, Ng SC, Bullock AJ, et al. Surgical management of gallbladder cancer: Simple versus extended cholecystectomy and the role of adjuvant therapy. Ann Surg, 2017, 266:625–31., DOI 10.1097/SLA.0000000000002385 Zhou T, Yu ST, Chen WZ, Xie R, Yu JC. Pretreatment albumin globulin ratio has a superior prognostic value in laryngeal squamous cell carcinoma patients: A comparison study. J Cancer, 2019, 10:594–601., DOI 10.7150/jca.28817 Nojiri S, Joh T. Albumin suppresses human hepatocellular carcinoma proliferation and the cell cycle. Int J Mol Sci, 2014, 15:5163–74., DOI 10.3390/ ijms15035163 Reebye V, Saetrom P, Mintz PJ, Huang KW, Swiderski P, Peng L, et al. Novel RNA oligonucleotide improves liver function and inhibits liver carcinogenesis in vivo. Hepatology, 2014, 59:216–27., DOI 10.1002/hep.26669 Shirai Y, Shiba H, Haruki K, Horiuchi T, Saito N, Fujiwara Y, et al. Preoperative platelet-to-albumin ratio predicts prognosis of patients with pancreatic ductal adenocarcinoma after pancreatic resection. Anticancer Res, 2017, 37:787–93., DOI 10.21873/anticanres.11378 Chen Z, Shao Y, Yao H, Zhuang Q, Wang K, Xing Z, et al. Preoperative albumin to globulin ratio predicts survival in clear cell renal cell carcinoma patients. Oncotarget, 2017, 8:48291–302., DOI 10.18632/oncotarget.15162 He X, Guo S, ChenD, Yang G, Chen X, Zhang Y, et al. Preoperative albumin to globulin ratio, AGR, as prognostic factor in renal cell carcinoma. J Cancer, 2017, 8: 258–65., DOI 10.7150/jca.16525 Bagirsakci E, Sahin E, Atabey N, Erdal E, Guerra V, Carr BI. Role of albumin in growth inhibition in hepatocellular carcinoma. Oncology, 2017, 93:136–42., DOI 10.1159/000471807 Zhang H, Ren P,MaM, Zhu X, Zhu K, Xiao W, et al. Prognostic significance of the preoperative albumin/fibrinogen ratio in patients with esophageal squamous cell carcinoma after surgical resection. J Cancer, 2021, 12:5025–34., DOI 10.7150/jca. 58022 Gupta D, Lis CG. Pretreatment serum albumin as a predictor of cancer survival: A systematic review of the epidemiological literature. Nutr J, 2010, 9:69., DOI 10.1186/1475-2891-9-69 Wheler J, Tsimberidou AM, Moulder S, Cristofanilli M, Hong D, Naing A, et al. Clinical outcomes of patients with breast cancer in a phase I clinic: The M. D. Anderson cancer center experience. Clin Breast Cancer, 2010, 10:46–51., DOI 10.3816/CBC.2010.n.006 Lei J, Wang Y, Guo X, Yan S, Ma D, Wang P, et al. Low preoperative serum ALB level is independently associated with poor overall survival in endometrial cancer patients. Future Oncol, 2020, 16:307–16., DOI 10.2217/fon-2019-0732 Li YT, Zhou XS, Han XM, Tian J, Qin Y, Zhang T, et al. Pretreatment serum albumin-to-alkaline phosphatase ratio is an independent prognosticator of survival in patients with metastatic gastric cancer. World J Gastrointest Oncol, 2022, 14: 1002–13., DOI 10.4251/wjgo.v14.i5.1002 Yu MC, Chan KM, Lee CF, Lee YS, Eldeen FZ, Chou HS, et al. Alkaline phosphatase: Does it have a role in predicting hepatocellular carcinoma recurrence. J Gastrointest Surg, 2011, 15:1440–9., DOI 10.1007/s11605-011-1537-3 Xu XS,Wan Y, Song SD, ChenW,Miao RC, Zhou YY, et al. Model based on γ- glutamyltransferase and alkaline phosphatase for hepatocellular carcinoma prognosis. World J Gastroenterol, 2014, 20:10944–52., DOI 10.3748/wjg.v20.i31.10944 Jin Y, Yuan MQ, Chen JQ, Zhang YP. Serum alkaline phosphatase predicts survival outcomes in patients with skeletal metastatic nasopharyngeal carcinoma. Clinics, Sao Paulo,, 2015, 70:264–72., DOI 10.6061/clinics/2015(04)08 Liu L, Xu Y, Jia S, Chen X, He S. Prognostic significance of serum alkaline phosphatase for all-cause mortality in patients with hypertrophic cardiomyopathy: A cohort of the hospitalized population. Ann Clin Biochem, 2022, 59:387–95., DOI 10.1177/00045632221113986 Dang C, Deng YH, Qin RY. Prognostic value of ALP and LDH in periampullary carcinoma patients undergoing surgery. Curr Med Sci, 2022, 42: 150–8., DOI 10.1007/s11596-021-2452-9 Wang Y, Xiong F, Yang J, Xia T, Jia Z, Shen J, et al. Decreased albumin-toalkaline phosphatase ratio predicted poor survival of resectable gastric cancer patients. J Gastrointest Oncol, 2021, 12:1338–50., DOI 10.21037/jgo-21-430 Liu X, Li Y, Zhao Q, Jiang H, Ni J, Cai H. Albumin-to-alkaline phosphatase ratio: A novel prognostic index for patients with driver mutation-negative advanced non-small cell lung cancer. Clin Respir J, 2021, 15:540–9., DOI 10.1111/crj.13339 Li J, Zuo M, Zhou X, Xiang Y, Zhang S, FengW, et al. Prognostic significance of preoperative albumin to alkaline phosphatase ratio in patients with glioblastoma. J Cancer, 2021, 12:5950–9., DOI 10.7150/jca.61866 Acikgoz Y, Bal O, Dogan M. Albumin-to-Alkaline phosphatase ratio: Does it predict survival in grade 1 and grade 2 neuroendocrine tumors. Pancreas, 2021, 50: 111–7., DOI 10.1097/MPA.0000000000001720 Sun L, Ke X, Wang D, Yin H, Jin B, Xu H, et al. Prognostic value of the albumin-to-γ-glutamyltransferase ratio for gallbladder cancer patients and establishing a nomogram for overall survival. J Cancer, 2021, 12:4172–82., DOI 10.7150/jca.49242 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610818 EP 1610830 DI 10.3389/pore.2022.1610818 PG 13 ER PT J AU Hou, J Guo, P Lu, Y Jin, X Liang, K Zhao, N Xue, Sh Zhou, Ch Wang, G Zhu, X Hong, H Chen, Y Lu, H Wang, W Xu, Ch Han, Y Cai, Sh Liu, Y AF Hou, Jun Guo, Peng Lu, Yujiao Jin, Xiaokang Liang, Ke Zhao, Na Xue, Shunxu Zhou, Chengmin Wang, Guoqiang Zhu, Xin Hong, Huangming Chen, Yungchang Lu, Huafei Wang, Wenxian Xu, Chunwei Han, Yusheng Cai, Shangli Liu, Yang TI A prognostic 15-gene model based on differentially expressed genes among metabolic subtypes in diffuse large B-cell lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; risk score; drug sensitivity; DEGs; diffuse large B-cell lymphoma (DLBCL); metabolic subtypes ID prognosis; risk score; drug sensitivity; DEGs; diffuse large B-cell lymphoma (DLBCL); metabolic subtypes AB The outcomes of patients with diffuse large B-cell lymphoma (DLBCL) vary widely, and about 40% of them could not be cured by the standard first-line treatment, R-CHOP, which could be due to the high heterogeneity of DLBCL. Here, we aim to construct a prognostic model based on the genetic signature of metabolic heterogeneity of DLBCL to explore therapeutic strategies for DLBCL patients. Clinical and transcriptomic data of one training and four validation cohorts of DLBCL were obtained from the GEO database. Metabolic subtypes were identified by PAM clustering of 1,916 metabolic genes in the 7 major metabolic pathways in the training cohort. DEGs among the metabolic clusters were then analyzed. In total, 108 prognosis-related DEGs were identified. Through univariable Cox and LASSO regression analyses, 15 DEGs were used to construct a risk score model. The overall survival (OS) and progression-free survival (PFS) of patients with high risk were significantly worse than those with low risk (OS: HR 2.86, 95%CI 2.04–4.01, p < 0.001; PFS: HR 2.42, 95% CI 1.77–3.31, p < 0.001). This model was also associated with OS in the four independent validation datasets (GSE10846: HR 1.65, p = 0.002; GSE53786: HR 2.05, p = 0.02; GSE87371: HR 1.85, p = 0.027; GSE23051: HR 6.16, p = 0.007) and PFS in the two validation datasets (GSE87371: HR 1.67, p = 0.033; GSE23051: HR 2.74, p = 0.049). Multivariable Cox analysis showed that in all datasets, the risk model could predict OS independent of clinical prognosis factors (p < 0.05). Compared with the high-risk group, patients in the low-risk group predictively respond to R-CHOP (p = 0.0042), PI3K inhibitor (p < 0.05), and proteasome inhibitor (p < 0.05). Therefore, in this study, we developed a signature model of 15 DEGs among 3 metabolic subtypes, which could predict survival and drug sensitivity in DLBCL patients. C1 [Hou, Jun] University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Department of PathologyChengdu, China. [Guo, Peng] University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Department of PathologyChengdu, China. [Lu, Yujiao] Burning Rock BiotechGuangzhou, China. [Jin, Xiaokang] Burning Rock BiotechGuangzhou, China. [Liang, Ke] Burning Rock BiotechGuangzhou, China. [Zhao, Na] University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Department of PathologyChengdu, China. [Xue, Shunxu] University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Department of PathologyChengdu, China. [Zhou, Chengmin] University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Department of PathologyChengdu, China. [Wang, Guoqiang] Burning Rock BiotechGuangzhou, China. [Zhu, Xin] Burning Rock BiotechGuangzhou, China. [Hong, Huangming] University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Medical OncologyChengdu, China. [Chen, Yungchang] University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Medical OncologyChengdu, China. [Lu, Huafei] Burning Rock BiotechGuangzhou, China. [Wang, Wenxian] The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Department of Clinical TrialHangzhou, China. [Xu, Chunwei] Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of SciencesHangzhou, China. [Han, Yusheng] Burning Rock BiotechGuangzhou, China. [Cai, Shangli] Burning Rock BiotechGuangzhou, China. [Liu, Yang] University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Department of PathologyChengdu, China. RP Liu, Y (reprint author), University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Department of Pathology, Chengdu, China. EM liuyang@scszlyy.org.cn CR Susanibar-Adaniya S, Barta SK. 2021 update on diffuse large B cell lymphoma: A review of current data and potential applications on risk stratification and management. Am J Hematol, 2021, 96(5):617–29., DOI 10.1002/ajh.26151 Liu Y, Barta SK. Diffuse large B-cell lymphoma: 2019 update on diagnosis, risk stratification, and treatment. Am J Hematol, 2019, 94(5):604–16., DOI 10.1002/ajh.25460 Chapuy B, Stewart C, Dunford AJ, Kim J, Kamburov A, Redd RA, et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med, 2018, 24(5):679–90., DOI 10.1038/ s41591-018-0016-8 International Non-Hodgkin’s Lymphoma Prognostic Factors Project. A predictive model for aggressive non-Hodgkin’s lymphoma. New Engl J Med, 1993, 329(14):987–94., DOI 10.1056/NEJM199309303291402 Hans CP, Weisenburger DD, Greiner TC, Gascoyne RD, Delabie J, Ott G, et al. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood, 2004, 103(1):275–82., DOI 10.1182/blood-2003-05-1545 Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, Fisher RI, et al. The use ofmolecular profiling to predict survival after chemotherapy for diffuse large-Bcell lymphoma. New Engl J Med, 2002, 346(25):1937–47., DOI 10.1056/ NEJMoa012914 Miao Y, Medeiros LJ, Xu-Monette ZY, Li J, Young KH. Dysregulation of cell survival in diffuse large B cell lymphoma: Mechanisms and therapeutic targets. Front Oncol, 2019, 9:107., DOI 10.3389/fonc.2019.00107 Ruminy P, Etancelin P, Couronne L, Parmentier F, Rainville V, Mareschal S, et al. The isotype of the BCR as a surrogate for the GCB and ABC molecular subtypes in diffuse large B-cell lymphoma. Leukemia, 2011, 25(4):681–8., DOI 10. 1038/leu.2010.302 Wilson WH, Young RM, Schmitz R, Yang Y, Pittaluga S, Wright G, et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med, 2015, 21(8):922–6., DOI 10.1038/nm.3884 Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell, 2011, 144(5):646–74., DOI 10.1016/j.cell.2011.02.013 Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science, New York, NY,, 2009, 324(5930):1029–33., DOI 10.1126/science.1160809 Martinez-Reyes I, Chandel NS. Cancer metabolism: Looking forward. Nat Rev Cancer, 2021, 21(10):669–80., DOI 10.1038/s41568-021-00378-6 Fendt SM, Frezza C, Erez A. Targeting metabolic plasticity and flexibility dynamics for cancer therapy. Cancer Discov, 2020, 10(12):1797–807., DOI 10.1158/ 2159-8290.CD-20-0844 Ricci JE, Chiche J. Metabolic reprogramming of non-hodgkin’s B-cell lymphomas and potential therapeutic strategies. Front Oncol, 2018, 8:556., DOI 10.3389/fonc.2018.00556 Gaude E, Frezza C. Tissue-specific and convergent metabolic transformation of cancer correlates with metastatic potential and patient survival. Nat Commun, 2016, 7:13041., DOI 10.1038/ncomms13041 Peng X, Chen Z, Farshidfar F, Xu X, Lorenzi PL, Wang Y, et al. Molecular characterization and clinical relevance of metabolic expression subtypes in human cancers. Cell Rep, 2018, 23(1):255–69.e4., DOI 10.1016/j.celrep.2018.03.077 Sung JY, Cheong JH. Pan-cancer analysis reveals distinct metabolic reprogramming in different epithelial-mesenchymal transition activity states. Cancers, Basel,, 2021, 13(8):1778., DOI 10.3390/cancers13081778 Cui L, Xue H, Wen Z, Lu Z, Liu Y, Zhang Y. Prognostic roles of metabolic reprogramming-associated genes in patients with hepatocellular carcinoma. Aging, Albany NY,, 2020, 12(21):22199–219., DOI 10.18632/aging.104122 Tan Z, Lei Y, Xu J, Shi S, Hua J, Zhang B, et al. The value of a metabolic reprogramming-related gene signature for pancreatic adenocarcinoma prognosis prediction. Aging, Albany NY,, 2020, 12(23):24228–41., DOI 10.18632/aging.104134 Zhang Z, Zhu H, Li Q, GaoW, Zang D, SuW, et al. Gene expression profiling of tricarboxylic acid cycle and one carbon metabolism related genes for prognostic risk signature of colon carcinoma. Front Genet, 2021, 12:647152., DOI 10.3389/fgene. 2021.647152 Caro P, Kishan AU, Norberg E, Stanley IA, Chapuy B, Ficarro SB, et al. Metabolic signatures uncover distinct targets in molecular subsets of diffuse large B cell lymphoma. Cancer cell, 2012, 22(4):547–60., DOI 10.1016/j.ccr.2012.08.014 Cokelaer T, Chen E, Iorio F, Menden MP, Lightfoot H, Saez-Rodriguez J, et al. GDSCTools for mining pharmacogenomic interactions in cancer. Bioinformatics, 2018, 34(7):1226–8., DOI 10.1093/bioinformatics/btx744 Geeleher P, Cox N, Huang RS. pRRophetic: an R package for prediction of clinical chemotherapeutic response from tumor gene expression levels. PLoS One, 2014, 9(9):e107468., DOI 10.1371/journal.pone.0107468 Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods, 2015, 12(5):453–7., DOI 10.1038/nmeth.3337 Visco C, Li Y, Xu-Monette ZY, Miranda RN, Green TM, Li Y, et al. Comprehensive gene expression profiling and immunohistochemical studies support application of immunophenotypic algorithm for molecular subtype classification in diffuse large B-cell lymphoma: A report from the international DLBCL rituximab-CHOP consortium program study. Leukemia, 2012, 26(9): 2103–13., DOI 10.1038/leu.2012.83 Ruppert AS, Dixon JG, Salles G, Wall A, Cunningham D, Poeschel V, et al. International prognostic indices in diffuse large B-cell lymphoma: A comparison of IPI, R-IPI, and NCCN-IPI. Blood, 2020, 135(23):2041–8., DOI 10.1182/blood. 2019002729 Nie Y, Liu L, Liu Q, Zhu X. Identification of ametabolic-related gene signature predicting the overall survival for patients with stomach adenocarcinoma. PeerJ, 2021, 9:e10908., DOI 10.7717/peerj.10908 Zhang ZY, Yao QZ, Liu HY, Guo QN, Qiu PJ, Chen JP, et al. Metabolic reprogramming-associated genes predict overall survival for rectal cancer. J Cel Mol Med, 2020, 24(10):5842–9., DOI 10.1111/jcmm.15254 Kollareddy M, Dimitrova E, Vallabhaneni KC, Chan A, Le T, Chauhan KM, et al. Regulation of nucleotide metabolism by mutant p53 contributes to its gain-offunction activities. Nat Commun, 2015, 6:7389., DOI 10.1038/ncomms8389 Cao Y, Chen P, Cai M, Shi Q, Xu P, Wang L, et al. Prognostic impact of B-vitamins involved in one-carbon metabolism in patients with diffuse large B-cell lymphoma. Hematol Oncol, 2020, 38(4):456–66., DOI 10.1002/hon.2752 ShenN, KormS, Karantanos T, Li D, Zhang X, Ritou E, et al. DLST-dependence dictates metabolic heterogeneity in TCA-cycle usage among triple-negative breast cancer. Commun Biol, 2021, 4(1):1289., DOI 10.1038/s42003-021-02805-8 Vacanti NM, Divakaruni AS, Green CR, Parker SJ, Henry RR, Ciaraldi TP, et al. Regulation of substrate utilization by the mitochondrial pyruvate carrier. Mol Cel, 2014, 56(3):425–35., DOI 10.1016/j.molcel.2014.09.024 Yuan X, Dong B, Xu Y, Dong L, Huang J, Zhang J, et al. TIKI2 is upregulated and plays an oncogenic role in renal cell carcinoma. Oncotarget, 2016, 7(13): 17212–9., DOI 10.18632/oncotarget.7873 Krejsgaard T, Litvinov IV, Wang Y, Xia L, Willerslev-Olsen A, Koralov SB, et al. Elucidating the role of interleukin-17F in cutaneous T-cell lymphoma. Blood, 2013, 122(6):943–50., DOI 10.1182/blood-2013-01-480889 IqbalMA, LiM, Lin J, Zhang G, ChenM,Moazzam NF, et al. Preliminary study on the sequencing of whole genomic methylation and transcriptome-related genes in thyroid carcinoma. Cancers, Basel,, 2022, 14(5):1163., DOI 10.3390/cancers14051163 Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature, 2010, 467(7319):1114–7., DOI 10.1038/nature09515 Dizeyi N, Bjartell A, Hedlund P, Tasken KA, Gadaleanu V, Abrahamsson PA. Expression of serotonin receptors 2B and 4 in human prostate cancer tissue and effects of their antagonists on prostate cancer cell lines. Eur Urol, 2005, 47(6): 895–900., DOI 10.1016/j.eururo.2005.02.006 Nakamura Y, Ise K, Yamazaki Y, Fujishima F, McNamara KM, Sasano H. Serotonin receptor 4, 5-hydroxytryptamine receptor Type 4, regulates expression of estrogen receptor beta and cell migration in hormone-naive prostate cancer. Indian J Pathol Microbiol, 2017, 60(1):33–7., DOI 10.4103/0377-4929.200022 Zapatka M, Borozan I, Brewer DS, Iskar M, Grundhoff A, Alawi M, et al. The landscape of viral associations in human cancers. Nat Genet, 2020, 52(3):320–30., DOI 10.1038/s41588-019-0558-9 Cheng AS, Li MS, Kang W, Cheng VY, Chou JL, Lau SS, et al. Helicobacter pylori causes epigenetic dysregulation of FOXD3 to promote gastric carcinogenesis. Gastroenterology, 2013, 144(1):122–33., DOI 10.1053/j.gastro.2012.10.002 Rosenbaum SR, Knecht M, Mollaee M, Zhong Z, Erkes DA, McCue PA, et al. FOXD3 regulates VISTA expression in melanoma. Cel Rep, 2020, 30(2):510–24., DOI 10.1016/j.celrep.2019.12.036 Kong L, Yang W, Chen L, Qian L. The DNA methylation-regulated MCTP1 activates the drug-resistance of esophageal cancer cells. Aging, Albany NY,, 2021, 13(3):3342–52., DOI 10.18632/aging.104173 Kovalevska L, Kashuba E. Expression pattern of MRPS18 family genes in malignantly transformed b-cells. Exp Oncol, 2020, 42(4):295–9., DOI 10.32471/exponcology. 2312-8852.vol-42-no-4.15366 Ma C, Li H, Li X, Lu S, He J. The prognostic value of faciogenital dysplasias as biomarkers in head and neck squamous cell carcinoma. Biomarkers Med, 2019, 13(16):1399–415., DOI 10.2217/bmm-2019-0273 Walker JS, Hing ZA, Sher S, Cronin J, Williams K, Harrington B, et al. Rare t(X;14)(q28;q32, translocation reveals link between MTCP1 and chronic lymphocytic leukemia. Nat Commun, 2021, 12(1):6338., DOI 10.1038/s41467-021- 26400-x Pfreundschuh M, Kuhnt E, Trumper L, Osterborg A, Trneny M, Shepherd L, et al. CHOP-Like chemotherapy with or without rituximab in young patients with good-prognosis diffuse large-B-cell lymphoma: 6-year results of an open-label randomised study of the MabThera international trial, MInT, group. Lancet Oncol, 2011, 12(11):1013–22., DOI 10.1016/S1470-2045(11)70235-2 Pfreundschuh M, Trumper L, Osterborg A, Pettengell R, Trneny M, Imrie K, et al. CHOP-Like chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with good-prognosis diffuse large-B-cell lymphoma: A randomised controlled trial by the MabThera international trial, MInT, group. Lancet Oncol, 2006, 7(5):379–91., DOI 10.1016/S1470-2045(06)70664-7 Lang R, Gill MJ. Diffuse large B-cell lymphoma. New Engl J Med, 2021, 384(23):2261–2., DOI 10.1056/NEJMc2105452 Chen W, Liang W, He Y, Liu C, Chen H, Lv P, et al. Immune microenvironment-related gene mapping predicts immunochemotherapy response and prognosis in diffuse large B-cell lymphoma. Med Oncol, 2022, 39(4):44., DOI 10.1007/s12032-021-01642-3 Keane C, Gill D, Vari F, Cross D, Griffiths L, Gandhi M. CD4(+, tumor infiltrating lymphocytes are prognostic and independent of R-IPI in patients with DLBCL receiving R-CHOP chemo-immunotherapy. Am J Hematol, 2013, 88(4): 273–6., DOI 10.1002/ajh.23398 Emens LA. Chemoimmunotherapy. Cancer J, 2010, 16(4):295–303., DOI 10. 1097/PPO.0b013e3181eb5066 Zhao Y, Niu C, Cui J. Gamma-delta, γδ, T cells: Friend or foe in cancer development? J translational Med, 2018, 16(1):3., DOI 10.1186/s12967-017- 1378-2 Tokuyama H, Hagi T, Mattarollo SR, Morley J, Wang Q, So HF, et al. V gamma 9 V delta 2 T cell cytotoxicity against tumor cells is enhanced by monoclonal antibody drugs-rituximab and trastuzumab. Int J Cancer, 2008, 122(11):2526–34., DOI 10.1002/ijc.23365 Yan M, Chang YM, Raghavan V, Dong E, Klein C, Nielsen TG, et al. Lymphoma microenvironment deconvolution links M1 macrophage infiltration to clinical outcome in diffuse large B-cell lymphoma. Blood, 2020, 136(1):29–30., DOI 10.1182/blood-2020-134867 Autio M, Leivonen SK, Bruck O, Mustjoki S, Meszaros Jorgensen J, Karjalainen-Lindsberg ML, et al. Immune cell constitution in the tumor microenvironment predicts the outcome in diffuse large B-cell lymphoma. Haematologica, 2021, 106(3):718–29., DOI 10.3324/haematol.2019.243626 Schmitz R, Wright GW, Huang DW, Johnson CA, Phelan JD,Wang JQ, et al. Genetics and pathogenesis of diffuse large B-cell lymphoma. New Engl JMed, 2018, 378(15):1396–407., DOI 10.1056/NEJMoa1801445 Wright GW, Huang DW, Phelan JD, Coulibaly ZA, Roulland S, Young RM, et al. A probabilistic classification tool for genetic subtypes of diffuse large B cell lymphoma with therapeutic implications. Cancer cell, 2020, 37(4):551–68., DOI 10. 1016/j.ccell.2020.03.015 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610819 EP 1610831 DI 10.3389/pore.2023.1610819 PG 13 ER PT J AU Deng, M Luo, R Huang, J Luo, Y Song, Q Liang, H Xu, Ch Yuan, W Hou, Y AF Deng, Minying Luo, Rongkui Huang, Jie Luo, Yuanlong Song, Qi Liang, Huaiyu Xu, Chen Yuan, Wei Hou, Yingyong TI Clinicopathologic features of gastric glomus tumor: A report of 15 cases and literature review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE glomus tumor; gastric tumor; immunohistochemistry; molecular genetics; prognosis ID glomus tumor; gastric tumor; immunohistochemistry; molecular genetics; prognosis AB Objective: Glomus tumor is a relatively uncommon soft tissue neoplasm predominantly occurring in upper extremity (fingers), less reported in stomach. This study aimed to discuss the clinicopathologic features of gastric glomus tumor (GGT) and then provide reference for clinical practice. Methods: A retrospective analysis of all cases pathologically diagnosed of GGT was performed, pathological findings were correlated with clinical information, immunohistochemical studies, next-generation sequencing, and patient follow-ups. A review of literature by searching similar cases was conducted to summarize previous knowledge of GGTs. Results: Our study identified 15GGTsincluded 5 males and 10 females, aged between 35–75 years old (median, 49 years old). The tumor was located to the gastric corpus in 6 cases (40%) and to the antrum in 9 cases (60%). The maximum tumor diameter ranged between 1–4 cm(median, 1.5 cm). There were 11 cases (73%) of solid glomus tumor, 3 cases (20%) ofmixture of solid glomus tumor and glomangioma, and 1 case (7%) of glomangiomyoma. Partial spindle cell area was observed in 3 cases (20%), moderate cellular atypia in 1 case (7%), atypicalmitosis in 1 case (7%), vascular invasion in 5 cases (33%), neural invasion in 6 cases (40%) and tumor necrosis in 1 case (7%). Tumor cells expressed Collagen type IV, α-smooth muscle actin (α-SMA), and synaptophysin in most cases. The Ki67 index varied from 1% to 30%. Nextgeneration sequencing reported EGFR, PIK3CA, KEAP1 and TP53 mutation. The outcome information was obtained in 12 (80%) cases, followed for 6–63 months, 11 patients (92%) had tumor-free survival and 1 patient (8%) developed liver metastasis 26 months after surgery. Literature review obtained 16 previously reported malignant GGT cases. In terms of the total 31 cases, univariate analysis revealed that the atypical mitosis (OS: p=0.009; DFS: p = 0.010) and severe cellular atypia (OS: p = 0.007; DFS: p = 0.004) were significantly associated with poor prognosis (patient death). Conclusion: GGT is indolent, while long-term close follow-up should be required in the presence of increasing number of risk factors. Malignant GGT is relatively uncommon and predisposes to liver metastasis, calling for accumulation of large-sample data and experience. C1 [Deng, Minying] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Luo, Rongkui] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Huang, Jie] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Luo, Yuanlong] Shanghai University of Traditional Chinese MedicineShanghai, China. [Song, Qi] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Liang, Huaiyu] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Xu, Chen] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Yuan, Wei] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Hou, Yingyong] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. RP Hou, Y (reprint author), Fudan University, Zhongshan Hospital, Department of Pathology, Shanghai, China. EM hou.yingyong@zs-hospital.sh.cn CR Smol’iannikov AA. Glomus tumors. Vopr Onkol, 1974, 20(10):104–16. Folpe AL, Fanburg-Smith JC, Miettinen M, Weiss SW. Atypical and malignant glomus tumors: Analysis of 52 cases, with a proposal for the reclassification of glomus tumors. Am J Surg Pathol, 2001, 25(1):1–12., DOI 10.1097/00000478-200101000-00001 Miettinen M, Paal E, Lasota J, Sobin LH. Gastrointestinal glomus tumors: A clinicopathologic, immunohistochemical, and molecular genetic study of 32 cases. Am J Surg Pathol, 2002, 26(3):301–11., DOI 10.1097/00000478-200203000-00003 Bray AP, Wong NA, Narayan S. Cutaneous metastasis from gastric glomus tumour. Clin Exp Dermatol, 2009, 34(8):e719–21., DOI 10.1111/j.1365-2230.2009.03445.x Lee H, Choi YS, Oh SC, Park JJ, Kim CW, Kim HC, et al. Malignant glomus tumors of the stomach - a report of 2 cases with multiple metastases. Korean J Pathol, 2009, 43(4):358–563., DOI 10.4132/KoreanJPathol.2009.43.4.358 Zhang Q, Zhang CS, Liu GQ, Yu WT, Ma Z. Malignant gastric glomus tumor and literature review. Chin J Digestion, 2009, 29(9):573–4., DOI 10.3760/cma.j.issn. 0254-1432.2009.09.019 Song SE, Lee CH, Kim KA, Lee HJ, Park CM. Malignant glomus tumor of the stomach with multiorgan metastases: Report of a case. Surg Today, 2010, 40(7): 662–7., DOI 10.1007/s00595-008-4113-z Teng TH, Huang SH, Liang CW. Malignant gastric glomus tumour mimicking GIST. Pathology, 2012, 44(3):261–3., DOI 10.1097/PAT.0b013e3283514067 Yao XF. A case of malignant gastric glomus tumor. Chin J Diagn Pathol, 2013, 20(6):380–1., DOI 10.3969/j.issn.1007-8096.2013.06.018 Tao YL, Wang ZJ, Han JG,Wei GH, Yi BQ, Wei P. A case of gastric malignant glomus tumor complicated by acute gastrointestinal bleeding. Cancer Res Clinic, 2013, 25(3):214–5., DOI 10.3760/cma.j.issn.1006-9801.2013.03.025 Zaidi S, Arafah M. Malignant gastric glomus tumor: A case report and literature review of a rare entity. Oman Med J, 2016, 31(1):60–4., DOI 10.5001/omj.2016.11 Li CX, Jiang HC, Wang QG. A case of gastric malignant glomus tumor. Chin J Med Imaging Technol, 2017, 33(3):483., DOI 10.13929/j.1003-3289.201608147 Bodolan AA, Wilcox R, Yang MX. Malignant glomus tumor of the gastric antrum with hepatic metastases: A case report and literature review. Hum Pathol Case Rep, 2018, 14:81–4., DOI 10.1016/j.ehpc.2018.09.004 Toti L, Manzia TM, Roma S, Meucci R, Blasi F, Ferlosio A, et al. Rare malignant glomus tumor of the stomach with liver metastases. Radiol Case Rep, 2019, 14(4):463–7., DOI 10.1016/j.radcr.2019.01.012 Kong FH, Chen LQ, Gao SM, Xu GD, Zhang YX. A case ofmalignant gastric glomus tumor. Chin J Diagn Pathol, 2019, 26(3):175178., DOI 10.3969/j.issn.1007-8096.2019.03.011 Alsahwan AG, Alfaraj ZM, AlSafwani J, Bunaiyan AH, AlKhalifah RH, Al- Saba’a SA, et al. Rare gastric neoplasm: Malignant glomus tumor of the stomach. A case report. Int J Surg Case Rep, 2021, 81:105802., DOI 10.1016/j.ijscr.2021.105802 Shugart RR, Soule EH, Johnson EW, Jr. Glomus tumor. Surg Gynecol Obstet, 1963, 117:334–40. Masson P. Le glomus neuromyoarterial des regions tactiles et ses tumeurs. Lyon Chir, 1924, 21:257–80. Lamba G, Rafiyath SM, Kaur H, Khan S, Singh P, Hamilton AM, et al. Malignant glomus tumor of kidney: The first reported case and review of literature. Hum Pathol, 2011, 42(8):1200–3., DOI 10.1016/j.humpath.2010.11.009 Kang G, Park HJ, Kim JY, Choi D,Min BH, Lee JH, et al. Glomus tumor of the stomach: A clinicopathologic analysis of 10 cases and review of the literature. Gut Liver, 2012, 6(1):52–7., DOI 10.5009/gnl.2012.6.1.52 Wang X, Hanif S, Wang B, Chai C. Management of gastric glomus tumor: A case report. Medicine, Baltimore,, 2019, 98(38):e16980., DOI 10.1097/MD. 0000000000016980 Kay S, Callahan WP, Jr, Murray MR, Randall HT, Stout AP. Glomus tumors of the stomach. Cancer, 1951, 4(4):726–36., DOI 10.1002/1097-0142(195107)4:4<726: aid-cncr2820040410>3.0.co;2-z Cao JX, Wang LQ, Li J. Clinicopathologic features of gastric glomus tumor. Chin J Diagn Pathol, 2021, 28(2):94–7., DOI 10.3969/j.issn.1007-8096.2021.02.004 Kirshbaum JD, Teitelman SL. Malignant tumor of the greater omentum simulating a glomangioma. Arch Path, 1939, 27:95–103. Yannopoulos K, Stout AP. Smooth muscle tumors in children. Cancer, 1962, 15: 958–71., DOI 10.1002/1097-0142(196209/10)15:5<958:aid-cncr2820150511>3.0.co;2-u Pansa A, Sama L, Ruspi L, Sicoli F, Cananzi FCM, Quagliuolo V. Glomus tumor of the stomach: A systematic review and illustrative case report. Dig Dis, 2022)., DOI 10.1159/000525513 Papke DJ, Jr, Sholl LM, Doyle LA, Fletcher C, Hornick JL. Gastroesophageal glomus tumors: Clinicopathologic and molecular genetic analysis of 26 cases with a proposal for malignancy criteria. Am J Surg Pathol, 2022, 46(10):1436–46., DOI 10. 1097/PAS.0000000000001925 Wu TT, Wu CL, Yang QX, Cai F, Zhang FR. Clinical and pathological features of gastric glomus tumor. Chin J Diagn Pathol, 2013, 20(7):410–3., DOI 10.3969/j.issn. 1007-8096.2013.07.009 Haque S, Modlin IM, West AB. Multiple glomus tumors of the stomach with intravascular spread. Am J Surg Pathol, 1992, 16(3):291–9., DOI 10.1097/00000478- 199203000-00010 Luzar B, Martin B, Fisher C, Calonje E. Cutaneous malignant glomus tumours: Applicability of currently established malignancy criteria for tumours occurring in the skin. Pathology, 2018, 50(7):711–7., DOI 10. 1016/j.pathol.2018.08.005 Zhu YZ, Li WP, Wang ZY, Yang HF, He QL, Zhu HG, et al. Glomus tumor of uncertain malignant potential arising in the bronchus. J Cardiothorac Surg, 2013, 8: 146., DOI 10.1186/1749-8090-8-146 Matevossian E, Brucher BL, Nahrig J, Feußner H, Huser N. Glomus tumor of the stomach simulating a gastrointestinal stromal tumor: A case report and review of literature. Case Rep Gastroenterol, 2008, 2(1):1–5., DOI 10.1159/000112862 Xu XD, Lu XH, Ye GX, Hu XR. Immunohistochemical analysis and biological behaviour of gastric glomus tumours: A case report and review of the literature. J Int Med Res, 2010, 38(4):1539–46., DOI 10.1177/ 147323001003800438 Wang JC, Jin XF,Weng SX, Xu C, GanMF. Gastric glomus tumors expressing synaptophysin: Clinicopathologic and immunohistochemical analyses. Zhonghua Bing Li Xue Za zhi= Chin J Pathol, 2017, 46(11):756–9., DOI 10.3760/cma.j.issn. 0529-5807.2017.11.004 Agaram NP, Zhang L, Jungbluth AA, Dickson BC, Antonescu CR. A molecular reappraisal of glomus tumors and related pericytic neoplasms with emphasis on NOTCH-gene fusions. Am J Surg Pathol, 2020, 44(11):1556–62., DOI 10.1097/PAS.0000000000001531 Karamzadeh Dashti N, Bahrami A, Lee SJ, Jenkins SM, Rodriguez FJ, Folpe AL, et al. BRAF V600E mutations occur in a subset of glomus tumors, and are associated with malignant histologic characteristics. Am J Surg Pathol, 2017, 41(11):1532–41., DOI 10.1097/PAS.0000000000000913 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610824 EP 1610838 DI 10.3389/pore.2022.1610824 PG 15 ER PT J AU Fejes, Zs Santa, F Jenei, A Kiraly, EI Varga, L Kuthi, L AF Fejes, Zsuzsanna Santa, Fanni Jenei, Alex Kiraly, Elod Istvan Varga, Linda Kuthi, Levente TI Angiomyolipoma of the kidney— Clinicopathological analysis of 52 cases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE AML; angiomyolipoma; kidney tumor; tuberous sclerosis; nephrectomy ID AML; angiomyolipoma; kidney tumor; tuberous sclerosis; nephrectomy AB The renal angiomyolipoma (AML) is a benign tumor characteristically composed of fat, smooth muscle tissue, and vessels. We collected AMLs from our nephrectomy database, reclassified them according to their histological appearance, recorded the demographic, clinical, and pathological parameters, and compared them with oncocytoma (RO) and renal cell carcinoma (RCC). Immunohistochemistry was ordered in 41 cases. In 2224 nephrectomies, we found 52 AMLs with a 53mm median size. The mean age was 52.76. Forty-eight tumors were sporadic, while four were hereditary. The revision resulted in 31 classic, 13 leiomyoma-like, five lipoma-like, two epithelioid, and one AML with epithelial cysts. SMA was diffusely positive, except for the epithelioid type,whileMelanA harbored stronger expression than HMB45. AML was more frequent in females and appeared ten and 7 years earlier than RO and RCC, respectively. The follow-up time was 7.42 years, and neither tumor-related death nor relapse occurred. AML is rare in nephrectomies and develops primarily in females in their 50swith an average sizeof 50–60mmat the surgery. The histological appearance in order of frequency is classic, leiomyoma-like, lipoma-like, epithelioid, and cystic. The MelanA, HMB45, and SMA immunohistochemistry can support the light-microscopic findings. C1 [Fejes, Zsuzsanna] University of Szeged, Department of RadiologySzeged, Hungary. [Santa, Fanni] University of Szeged, Department of PathologySzeged, Hungary. [Jenei, Alex] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kiraly, Elod Istvan] University of Szeged, Department of UrologySzeged, Hungary. [Varga, Linda] University of Szeged, Department of OncotherapySzeged, Hungary. [Kuthi, Levente] University of Szeged, Department of PathologySzeged, Hungary. RP Kuthi, L (reprint author), University of Szeged, Department of Pathology, Szeged, Hungary. EM kuthi.levente@med.u-szeged.hu CR Folpe AL, Kwiatkowski DJ. Perivascular epithelioid cell neoplasms: Pathology and pathogenesis. Hum Pathol, 2010, 41(1):1–15., DOI 10.1016/j.humpath.2009. 05.011 Martignoni G, Pea M, Reghellin D, Zamboni G, Bonetti F. PEComas: The past, the present and the future. Virchows Arch, 2008, 452(2):119–32., DOI 10.1007/ s00428-007-0509-1 Martignoni G, Pea M, Reghellin D, Zamboni G, Bonetti F. Perivascular epithelioid cell tumor, PEComa, in the genitourinary tract. Adv Anat Pathol, 2007, 14(1):36–41., DOI 10.1097/PAP.0b013e31802e0dc4 WHO Classification of Tumours Editorial Board. Urinary and male genital tumours. In: WHO classification of tumours series. 5th ed.; vol. 8. Lyon, France): International Agency for Research on Cancer, 2022). van Slegtenhorst M, de Hoogt R, Hermans C, Nellist M, Janssen B, Verhoef S, et al. Identification of the tuberous sclerosis gene TSC1 on chromosome 9q34. Science, 1997, 277(5327):805–8., DOI 10.1126/science. 277.5327.805 Eble JN. Angiomyolipoma of kidney. Semin Diagn Pathol, 1998, 15(1):21–40. Henske EP, Ao X, Short MP, Greenberg R, Neumann HP, Kwiatkowski DJ, et al. Frequent progesterone receptor immunoreactivity in tuberous sclerosisassociated renal angiomyolipomas. Mod Pathol, 1998, 11(7):665. Cook JA, Oliver K, Mueller RF, Sampson J. A cross sectional study of renal involvement in tuberous sclerosis. J Med Genet, 1996, 33(6):480–4., DOI 10.1136/ jmg.33.6.480 Smolarek TA, Wessner LL, McCormack FX, Mylet JC, Menon AG, Henske EP, et al. Evidence that lymphangiomyomatosis is caused by TSC2 mutations: Chromosome 16p13 loss of heterozygosity in angiomyolipomas and lymph nodes from women with lymphangiomyomatosis. Am J Hum Genet, 1998, 62(4):810–5., DOI 10.1086/301804 European Chromosome 16 Tuberous Sclerosis Consortium. Identification and characterization of the tuberous sclerosis gene on chromosome 16. Cell, 1993, 75(7):1305–15., DOI 10.1016/0092-8674(93)90618-z Dibble CC, Elis W, Menon S, Qin W, Klekota J, Asara JM, et al. TBC1D7 is a third subunit of the TSC1-TSC2 complex upstream of mTORC1. Mol Cel, 2012, 47(4):535–46., DOI 10.1016/j.molcel.2012.06.009 Steiner MS, Goldman SM, Fishman EK, Marshall FF. The natural history of renal angiomyolipoma. J Urol, 1993, 150(6):1782–6., DOI 10.1016/s0022-5347(17, 35895-0 Yamakado K, Tanaka N, Nakagawa T, Kobayashi S, Yanagawa M, Takeda K. Renal angiomyolipoma: Relationships between tumor size, aneurysm formation, and rupture. Radiology, 2002, 225(1):78–82., DOI 10.1148/radiol.2251011477 Fujii Y, Ajima J, Oka K, Tosaka A, Takehara Y. Benign renal tumors detected among healthy adults by abdominal ultrasonography. Eur Urol, 1995, 27:124–7., DOI 10.1159/000475142 Halpenny D, Snow A, McNeill G, TorreggianiWC. The radiological diagnosis and treatment of renal angiomyolipomacurrent status. Clin Radiol, 2010, 65(2): 99–108., DOI 10.1016/j.crad.2009.09.014 Varma S, Gupta S, Talwar J, Forte F, Dhar M. Renal epithelioid angiomyolipoma: A malignant disease. J Nephrol, 2011, 24(1):18–22., DOI 10. 5301/jn.2010.5451 Kyo CK, Won TK, Won SH, Jin SL, Hee JJ, Young DC. Trends of presentation and clinical outcome of treated renal angiomyolipoma. Yonsei Med J, 2010, 51(5): 728–34., DOI 10.3349/ymj.2010.51.5.728 Zavala-Pompa A, Folpe AL, Jimenez RE, Lim SD, Cohen C, Eble JN, et al. Immunohistochemical study of microphthalmia transcription factor and tyrosinase in angiomyolipoma of the kidney, renal cell carcinoma, and renal and retroperitoneal sarcomas: Comparative evaluation with traditional diagnostic markers. Am J Surg Pathol, 2001, 25(1):65–70., DOI 10.1097/00000478-200101000- 00007 Amin S, Kingswood JC, Bolton PF, Elmslie F, Gale DP, Harland C, et al. The UK guidelines for management and surveillance of Tuberous Sclerosis Complex. QJM, 2019, 112(3):171–82., DOI 10.1093/qjmed/hcy215 Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, et al. The 2015 world health organization classification of lung tumors: Impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol, 2015, 10(9):1243–60., DOI 10.1097/JTO.0000000000000630 Bennett JA, Braga AC, Pinto A, Van de Vijver K, Cornejo K, Pesci A, et al. Uterine PEComas: A morphologic, immunohistochemical, and molecular analysis of 32 tumors. Am J Surg Pathol, 2018, 42(10):1370–83., DOI 10.1097/PAS. 0000000000001119 Baniak N, Barletta JA, HirschMS. Key renal neoplasms with a female predominance. Adv Anat Pathol, 2021, 28(4):228–50., DOI 10.1097/PAP.0000000000000301 Jenei A, Hes O, Kuthi L. Provisional renal cell carcinoma subsets following the 2016 WHO classification. Orv Hetil, 2020, 161(3):83–94., DOI 10.1556/650.2020.31654 Yanai H, Sasagawa I, Kubota Y, Ishigooka M, Hashimoto T, Kaneko H, et al. Spontaneous hemorrhage during pregnancy secondary to renal angiomyolipoma. Urol Int, 1996, 56(3):188–91., DOI 10.1159/000282838 Oesterling JE, Fishman EK, Goldman SM, Marshall FF. The management of renal angiomyolipoma. J Urol, 1986, 135(6):1121–4., DOI 10.1016/s0022-5347(17)46013-7 Bakshi SS, Vishal K, Kalia V, Gill JS. Aggressive renal angiomyolipoma extending into the renal vein and inferior vena cava - an uncommon entity. Br J Radiol, 2011, 84(1004):e166–8., DOI 10.1259/bjr/98449202 Tong YC, Chieng PU, Tsai TC, Lin SN. Renal angiomyolipoma: Report of 24 cases. Br J Urol, 1990, 66(6):585–9., DOI 10.1111/j.1464-410x.1990.tb07187.x Song S, Park BK, Park JJ.New radiologic classification of renal angiomyolipomas. Eur J Radiol, 2016, 85(10):1835–42., DOI 10.1016/j.ejrad.2016.08.012 Park BK. Renal angiomyolipoma: Radiologic classification and imaging features according to the amount of fat. Am J Roentgenol, 2017, 209(4):826–35., DOI 10.2214/AJR.17.17973 Shetty AS, Sipe AL, Zulfiqar M, Tsai R, Raptis DA, Raptis CA, et al. In-phase and opposed-phase imaging: Applications of chemical shift and magnetic susceptibility in the chest and abdomen. Radiographics, 2019, 39(1):115–35., DOI 10.1148/rg.2019180043 Shirotake S, Yoshimura I, Kosaka T, Matsuzaki S. A case of angiomyolipoma of the renal sinus. Clin Exp Nephrol, 2011, 15(6):953–6., DOI 10.1007/s10157-011-0519-9 Lin WY, Chuang CK, Ng KF, Liao SK. Renal angiomyolipoma with lymph node involvement: A case report and literature review. Chang Gung Med J, 2003, 26(8):607. Ackerman TE, Levi CS, Lindsay DJ, Greenberg HM, Gough JC. Angiomyolipoma with lymph node involvement. Can Assoc Radiol J, 1994, 45(1):52. Stone CH, Lee MW, Amin MB, Yaziji H, Gown AM, Ro JY, et al. Renal angiomyolipoma: Further immunophenotypic characterization of an expanding morphologic spectrum. Arch Pathol Lab Med, 2001, 125(6):751–8., DOI 10.1043/ 0003-9985(2001)125<0751:RA>2.0.CO;2 Sooriakumaran P, Gibbs P, Coughlin G, Attard V, Elmslie F, Kingswood C, et al. Angiomyolipomata: Challenges, solutions, and future prospects based on over 100 cases treated. BJU Int, 2010, 105(1):101–6., DOI 10.1111/j.1464-410X.2009.08649.x Schieda N, Kielar AZ, Al Dandan O,McInnes MD, Flood TA. Ten uncommon and unusual variants of renal angiomyolipoma, AML): Radiologic-pathologic correlation. Clin Radiol, 2015, 70(2):206–20., DOI 10.1016/j.crad.2014.10.001 Aydin H, Magi-Galluzzi C, Lane BR, Sercia L, Lopez JI, Rini BI, et al. Renal angiomyolipoma: Clinicopathologic study of 194 cases with emphasis on the epithelioid histology and tuberous sclerosis association. Am J Surg Pathol, 2009, 33(2):289–97., DOI 10.1097/PAS.0b013e31817ed7a6 Katabathina VS, Vikram R, Nagar AM, Tamboli P, Menias CO, Prasad SR. Mesenchymal neoplasms of the kidney in adults: Imaging spectrum with radiologicpathologic correlation. Radiographics, 2010, 30(6):1525–40., DOI 10.1148/rg.306105517 Di BA, Ferrara G, Gogglia P. Renal capsuloma: Description of a case with predominantly muscular differentiation. Pathologica, 2003, 95(2):119. Aldughiman AW, Alzahrani A, Alzahrani T. Renal leiomyoma: Case report and literature review. J Endourol Case Rep, 2019, 5(4):181–3., DOI 10.1089/cren.2019.0049 Gobbo S, Eble JN,Huang J, Grignon DJ, WangM,Martignoni G, et al. Schwannoma of the kidney. Mod Pathol, 2008, 21(6):779–83., DOI 10.1038/modpathol.2008.52 Mehta V, Venkataraman G, Antic T, Rubinas TC, Le Poole IC, Picken MM. Renal angiomyolipoma, fat-poor variant - a clinicopathologic mimicker of malignancy. Virchows Arch, 2013, 463(1):41–6., DOI 10.1007/s00428-013-1432-2 Oh SD, Oh SJ, Suh BJ, Shin JY, Oh CK, Park JK, et al. A giant retroperitoneal liposarcoma encasing the entire left kidney and adherent to adjacent structures: A case report. Case Rep Oncol, 2016, 9(2):368–72., DOI 10.1159/000447488 Knebel C, Neumann J, Schwaiger BJ, Karampinos DC, Pfeiffer D, Specht K, et al. Differentiating atypical lipomatous tumors from lipomas with magnetic resonance imaging: A comparison with MDM2 gene amplification status. BMC Cancer, 2019, 19(1):309., DOI 10.1186/s12885-019-5524-5 Davis CJ, Barton JH, Sesterhenn IA. Cystic angiomyolipoma of the kidney: A clinicopathologic description of 11 cases. Mod Pathol, 2006, 19(5):669–74., DOI 10. 1038/modpathol.3800572 Fine SW, Reuter VE, Epstein JI, Argani P. Angiomyolipoma with epithelial cysts, AMLEC): A distinct cystic variant of angiomyolipoma. Am J Surg Pathol, 2006, 30(5):593–9., DOI 10.1097/01.pas.0000194298.19839.b4 Armah HB, Yin M, Rao UN, Parwani AV. Angiomyolipoma with epithelial cysts, AMLEC): A rare but distinct variant of angiomyolipoma. Diagn Pathol, 2007, 2:11., DOI 10.1186/1746-1596-2-11 Michal M, Hes O, Bisceglia M, Simpson RH, Spagnolo DV, Parma A, et al. Mixed epithelial and stromal tumors of the kidney. A report of 22 cases. Virchows Arch, 2004, 445(4):359–67., DOI 10.1007/s00428-004-1060-y Nese N, Martignoni G, Fletcher CD, Gupta R, Pan CC, Kim H, et al. Pure epithelioid PEComas, so-called epithelioid angiomyolipoma, of the kidney: A clinicopathologic study of 41 cases: Detailed assessment of morphology and risk stratification. Am J Surg Pathol, 2011, 35(2):161–76., DOI 10.1097/PAS. 0b013e318206f2a9 Brimo F, Robinson B, Guo C, Zhou M, LatourM, Epstein JI. Renal epithelioid angiomyolipoma with atypia: A series of 40 cases with emphasis on clinicopathologic prognostic indicators of malignancy. Am J Surg Pathol, 2010, 34(5):715–22., DOI 10.1097/PAS.0b013e3181d90370 He W, Cheville JC, Sadow PM, Gopalan A, Fine SW, Al-Ahmadie HA, et al. Epithelioid angiomyolipoma of the kidney: Pathological features and clinical outcome in a series of consecutively resected tumors. Mod Pathol, 2013, 26(10): 1355–64., DOI 10.1038/modpathol.2013.72 Mete O, van der Kwast TH. Epithelioid angiomyolipoma: A morphologically distinct variant that mimics a variety of intra-abdominal neoplasms. Arch Pathol Lab Med, 2011, 135(5):665–70., DOI 10.1043/2009-0637-RSR.1 Giannikou K, Malinowska IA, Pugh TJ, Yan R, Tseng YY, Oh C, et al. Whole exome sequencing identifies TSC1/TSC2 biallelic loss as the primary and sufficient driver event for renal angiomyolipoma development. Plos Genet, 2016, 12(8): e1006242., DOI 10.1371/journal.pgen.1006242 Li W, Guo L, Bi X, Ma J, Zheng S. Immunohistochemistry of p53 and Ki-67 and p53 mutation analysis in renal epithelioid angiomyolipoma. Int J Clin Exp Pathol, 2015, 8(8):9446. Roma AA, Magi-Galluzzi C, Zhou M. Differential expression of melanocytic markers in myoid, lipomatous, and vascular components of renal angiomyolipomas. Arch Pathol Lab Med, 2007, 131(1):122–5., DOI 10.1043/1543- 2165(2007)131[122:DEOMMI]2.0.CO;2 BonsibSM, BoilsC,GokdenN,GrignonD, GuX,Higgins JP, etal.Tuberous sclerosis complex: Hamartin and tuberin expression in renal cysts and its discordant expression in renal neoplasms. Pathol Res Pract, 2016, 212(11):972–9., DOI 10.1016/j.prp.2016.04.005 Sobiborowicz A, Czarnecka AM, Szumera-Cieckiewicz A, Rutkowski P, Switaj T. Rozpoznanie i leczenie nowotworow typu angiomyolipoma, AML). Oncol Clin Pract, 2020, 16(3):116–32., DOI 10.5603/ocp.2020.0008 Ljungberg B, Albiges L, Abu-Ghanem Y, Bensalah K, Dabestani S, Fernandez-Pello S, et al. European association of urology guidelines on renal cell carcinoma: The 2019 update. Eur Urol, 2019, 75(5):799–810., DOI 10.1016/j. eururo.2019.02.011 Fernandez-Pello S, Hora M, Kuusk T, Tahbaz R, Dabestani S, Abu-Ghanem Y, et al. Management of sporadic renal angiomyolipomas: A systematic review of available evidence to guide recommendations from the European association of urology renal cell carcinoma guidelines panel. Eur Urol Oncol, 2020, 3(1):57–72., DOI 10.1016/j.euo.2019.04.005 Vos N, Oyen R. Renal angiomyolipoma: The good, the bad, and the ugly. J Belg Soc Radiol, 2018, 102(1):41., DOI 10.5334/jbsr.1536 Kuusk T, Biancari F, Lane B, Tobert C, Campbell S, Rimon U, et al. Treatment of renal angiomyolipoma: Pooled analysis of individual patient data. BMC Urol, 2015, 15:123., DOI 10.1186/s12894-015-0118-2 Bissler JJ, Kingswood JC. Renal angiomyolipomata. Kidney Int, 2004, 66(3): 924–34., DOI 10.1111/j.1523-1755.2004.00838.x Lei JH, Liu LR, Wei Q, Song TR, Yang L, Yuan HC, et al. A four-year follow-up study of renal epithelioid angiomyolipoma: Amulti-center experience and literature review. Sci Rep, 2015, 5:10030., DOI 10.1038/srep10030 Lattanzi M, Deng FM, Chiriboga LA, Femia AN, Meehan SA, Iyer G, et al. Durable response to anti-PD-1 immunotherapy in epithelioid angiomyolipoma: A report on the successful treatment of a rare malignancy. J Immunother Cancer, 2018, 6(1):97., DOI 10.1186/s40425-018-0415-x NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610831 EP 1610841 DI 10.3389/pore.2022.1610831 PG 11 ER PT J AU Phikulsod, P Sukpanichnant, S Kunacheewa, Ch Chieochansin, Th Junking, M Yenchitsomanus, Pth AF Phikulsod, Ployploen Sukpanichnant, Sanya Kunacheewa, Chutima Chieochansin, Thaweesak Junking, Mutita Yenchitsomanus, Pa-thai TI High prevalence of Wilms tumor 1 expression in multiple myeloma and plasmacytoma: A cohort of 142 Asian patients’ samples SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE multiple myeloma; immunohistochemistry (IHC); plasmacytoma; WT1 protein; cohort study Thailand; Asian cohort; treatment outcome; clinical relevance ID multiple myeloma; immunohistochemistry (IHC); plasmacytoma; WT1 protein; cohort study Thailand; Asian cohort; treatment outcome; clinical relevance AB Wilms tumor 1 (WT1) is a promising target antigen for cancer immunotherapy. However, WT1 protein expression and its clinical correlation in multiple myeloma (MM) patients are still limited. We, therefore, investigated WT1 expression in 142 bone marrow and plasmacytoma samples of MM patients at different stages of the disease by immunohistochemistry. The correlations between WT1 expression and clinical parameters or treatment outcomes were evaluated. The overall positive rate of WT1 expression was 91.5%; this high prevalence was found in both bone marrow and plasmacytoma samples, regardless of the disease status. Cytoplasmic WT1 expression was correlated with high serum free light chain ratio at presentation. However, no significant association between WT1 expression and treatment outcome was observed. This study confirms the high prevalence of WT1 expression in an Asian cohort of MM, encouraging the development of immunotherapy targeting WT1 in MM patients, particularly in those with extramedullary plasmacytoma or relapsed disease. C1 [Phikulsod, Ployploen] Mahidol University, Faculty of Medicine Siriraj Hospital, Research Department, Division of Molecular MedicineBangkok, Thailand. [Sukpanichnant, Sanya] Mahidol University, Faculty of Medicine Siriraj Hospital, Department of PathologyBangkok, Thailand. [Kunacheewa, Chutima] Mahidol University, Faculty of Medicine Siriraj Hospital, Department of MedicineBangkok, Thailand. [Chieochansin, Thaweesak] Mahidol University, Faculty of Medicine Siriraj Hospital, Research Department, Division of Molecular MedicineBangkok, Thailand. [Junking, Mutita] Mahidol University, Faculty of Medicine Siriraj Hospital, Research Department, Division of Molecular MedicineBangkok, Thailand. [Yenchitsomanus, Pa-thai] Mahidol University, Faculty of Medicine Siriraj Hospital, Research Department, Division of Molecular MedicineBangkok, Thailand. RP Yenchitsomanus, Pth (reprint author), Mahidol University, Faculty of Medicine Siriraj Hospital, Research Department, Division of Molecular Medicine, Bangkok, Thailand. EM ptyench@gmail.com CR Call KM, Glaser T, Ito CY, Buckler AJ, Pelletier J, Haber DA, et al. Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11Wilms’ tumor locus. Cell, 1990, 60(3):509–20., DOI 10.1016/0092-8674(90)90601-a Liu HS, Zhu MS, Zhang HL, Wei SY, Wang XN, Xi XP, et al. WT1 gene expression difference in leukemia and non-leukemia and its clinical significance. Zhongguo Shi Yan Xue Ye Xue Za Zhi, 2014, 22(5):1217–21., DOI 10.7534/j.issn. 1009-2137.2014.05.005 Qiwei X, Zhang F, Wu H, Liu JL, Zongge B, Xu C, et al. Wilms’ tumor 1, WT1, expression and prognosis in solid cancer patients: a systematic review and metaanalysis. Sci Rep, 2015, 5:8924., DOI 10.1038/srep08924 Parenti R, Perris R, Vecchio GM, Salvatorelli L, Torrisi A, Gravina L, et al. Immunohistochemical expression of Wilms’ tumor protein, WT1, in developing human epithelial and mesenchymal tissues. Acta Histochem, 2013, 115(1):70–5., DOI 10.1016/j.acthis.2012.04.006 Nakatsuka S-I, Oji Y, Horiuchi T, Kanda T, Kitagawa M, Takeuchi T, et al. Immunohistochemical detection of WT1 protein in a variety of cancer cells. Mod Pathol Off J U S Can Acad Pathol Inc, 2006, 19(6):804–14., DOI 10.1038/modpathol.3800588 Hein KZ, Yao S, Fu S. Wilms’ tumor 1, WT1): The vaccine for cancer. J Immunother Precis Oncol, 2020, 3(4):165–71., DOI 10.36401/JIPO-20-12 Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res Off J Am Assoc Cancer Res, 2009, 15(17):5323–37., DOI 10.1158/1078-0432.CCR-09-0737 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Mateos MV, Martinez BP, Gonzalez-Calle V. High-risk multiplemyeloma: how to treat at diagnosis and relapse? Hematology, 2021, 2021(1):30–6., DOI 10.1182/ hematology.2021000229 Verheye E, BravoMelgar J, Deschoemaeker S, Raes G,Maes A, De Bruyne E, et al. Dendritic cell-based immunotherapy in multiple myeloma: Challenges, opportunities, and future directions. Int J Mol Sci, 2022, 23(2):904., DOI 10.3390/ijms23020904 Saatci C, Caglayan AO, Kocyigit I, Akalin H, Kaynar LG, Altuntas F, et al. Expression of WT1 gene in multiple myeloma patients at diagnosis: Is WT1 gene expression a useful marker in multiple myeloma? Hematol Amst Neth, 2010, 15(1): 39–42., DOI 10.1179/102453310X12583347009496 Azuma T, Otsuki T, Kuzushima K, Froelich CJ, Fujita S, Yasukawa M. Myeloma cells are highly sensitive to the granule exocytosis pathway mediated by WT1-specific cytotoxic T lymphocytes. Clin Cancer Res Off J Am Assoc Cancer Res, 2004, 10(21):7402–12., DOI 10.1158/1078-0432.CCR-04-0825 Tyler EM, Jungbluth AA, O’Reilly RJ, Koehne G. WT1-specific T-cell responses in high-risk multiple myeloma patients undergoing allogeneic T celldepleted hematopoietic stem cell transplantation and donor lymphocyte infusions. Blood, 2013, 121(2):308–17., DOI 10.1182/blood-2012-06-435040 Koehne G, Devlin S, Korde N, Mailankody S, Heather L, Hassoun H, et al. Galinpepimut-S, a WT1-targeting immuno-oncology treatment, induces specific, robust and durable immune responses, IRs, in patients, pts, with high-risk, HR, multiple myeloma, MM). Clin Lymphoma Myeloma Leuk, 2017, 17:S343–4., DOI 10. 1016/j.clml.2017.07.164 Tsuboi A, Oka Y, Nakajima H, Fukuda Y, Elisseeva OA, Yoshihara S, et al. Wilms tumor gene WT1 peptide-based immunotherapy induced a minimal response in a patient with advanced therapy-resistant multiple myeloma. Int J Hematol, 2007, 86(5):414–7., DOI 10.1007/BF02983998 Li GJ, Ding H, Yang L. Correlation of WT1 and VEGF expression with angiogenesis in bone marrow biopsies of multiple myeloma patients. Zhongguo Shi Yan Xue Ye Xue Za Zhi, 2015, 23(3):733–6., DOI 10.7534/j.issn.1009-2137.2015.03.026 Avivi I, Cohen YC, Suska A, Shragai T, Mikala G, Garderet L, et al. Hematogenous extramedullary relapse in multiple myeloma - a multicenter retrospective study in 127 patients. Am J Hematol, 2019, 94(10):1132–40., DOI 10.1002/ajh.25579 Tsuta K, Kato Y, Tochigi N, Hoshino T, Takeda Y, Hosako M, et al. Comparison of different clones, WT49 versus 6F-H2, of WT-1 antibodies for immunohistochemical diagnosis of malignant pleural mesothelioma. Appl Immunohistochem Mol Morphol AIMM, 2009, 17(2):126–30., DOI 10.1097/PAI. 0b013e318187dd26 Novacescu D, Cut TG, Cumpanas AA, Latcu SC, Bardan R, Ferician O, et al. Evaluating established roles, future perspectives and methodological heterogeneity for Wilms’ tumor 1, WT1, antigen detection in adult renal cell carcinoma, using a novel N-terminus targeted antibody, clone WT49). Biomedicines, 2022, 10(4):912., DOI 10.3390/biomedicines10040912 Yadav P, Cockwell P, Cook M, Pinney J, Giles H, Aung YS, et al. Serum free light chain levels and renal function at diagnosis in patients with multiple myeloma. BMC Nephrol, 2018, 19(1):178., DOI 10.1186/s12882-018-0962-x Walker BA, Mavrommatis K, Wardell CP, Ashby TC, Bauer M, Davies FE, et al. Identification of novel mutational drivers reveals oncogene dependencies in multiple myeloma. Blood, 2018, 132(6):587–97., DOI 10.1182/blood-2018-03- 840132 Bolli N, Biancon G, Moarii M, Gimondi S, Li Y, de Philippis C, et al. Analysis of the genomic landscape of multiple myeloma highlights novel prognostic markers and disease subgroups. Leukemia, 2018, 32(12):2604–16., DOI 10.1038/s41375-018- 0037-9 AACR Project GENIE Consortium. AACR project GENIE: Powering precision medicine through an international consortium. Cancer Discov, 2017, 7(8):818–31., DOI 10.1158/2159-8290.CD-17-0151 Broyl A, Hose D, Lokhorst H, de Knegt Y, Peeters J, Jauch A, et al. Gene expression profiling for molecular classification of multiple myeloma in newly diagnosed patients. Blood, 2010, 116(14):2543–53., DOI 10.1182/blood-2009-12- 261032 Niksic M, Slight J, Sanford JR, Caceres JF, Hastie ND. The Wilms’ tumour protein, WT1, shuttles between nucleus and cytoplasm and is present in functional polysomes. Hum Mol Genet, 2004, 13(4):463–71., DOI 10.1093/hmg/ddh040 Toska E, Roberts SGE. Mechanisms of transcriptional regulation by WT1, Wilms’ tumour 1). Biochem J, 2014, 461(1):15–32., DOI 10.1042/BJ20131587 DehbiM, Hiscott J, Pelletier J. Activation of the wt1 Wilms’ tumor suppressor gene by NF-kappaB. Oncogene, 1998, 16(16):2033–9., DOI 10.1038/sj.onc.1201747 Chesi M, Bergsagel PL. Molecular pathogenesis of multiple myeloma: Basic and clinical updates. Int J Hematol, 2013, 97(3):313–23., DOI 10.1007/s12185-013- 1291-2 Vicent S, Chen R, Sayles LC, Lin C, Walker RG, Gillespie AK, et al. Wilms tumor 1, WT1, regulates KRAS-driven oncogenesis and senescence in mouse and human models. J Clin Invest, 2010, 120(11):3940–52., DOI 10.1172/JCI44165 Zhou B, Jin X, Jin W, Huang X, Wu Y, Li H, et al. WT1 facilitates the selfrenewal of leukemia-initiating cells through the upregulation of BCL2L2: WT1- BCL2L2 axis as a new acute myeloid leukemia therapy target. J Transl Med, 2020, 18(1):254., DOI 10.1186/s12967-020-02384-y Han Y, San-Marina S, Liu J,Minden MD. Transcriptional activation of c-myc proto-oncogene by WT1 protein. Oncogene, 2004, 23:6933–41., DOI 10.1038/sj.onc. 1207609 Silberstein GB, Van Horn K, Strickland P, Roberts CT, Daniel CW. Altered expression of the WT1 Wilms tumor suppressor gene in human breast cancer. Proc Natl Acad Sci, 1997, 94(15):8132–7., DOI 10.1073/pnas.94.15.8132 Ohno S, Dohi S, Ohno Y, Kyo S, Sugiyama H, Suzuki N, et al. Immunohistochemical detection of WT1 protein in endometrial cancer. Anticancer Res, 2009, 29(5):1691–5. Carpentieri DF, Nichols K, Chou PM, Matthews M, Pawel B, Huff D. The expression of WT1 in the differentiation of rhabdomyosarcoma from other pediatric small round blue cell tumors. Mod Pathol Off J U S Can Acad Pathol Inc, 2002, 15(10):1080–6., DOI 10.1097/01.MP.0000028646.03760.6B Ye Y, Raychaudhuri B, Gurney A, Campbell CE, Williams BR. Regulation of WT1 by phosphorylation: Inhibition of DNA binding, alteration of transcriptional activity and cellular translocation. EMBO J, 1996, 15(20):5606–15., DOI 10.1002/j. 1460-2075.1996.tb00945.x Podar K, Raab MS, Zhang J, McMillin D, Breitkreutz I, Tai YT, et al. Targeting PKC in multiple myeloma: In vitro and in vivo effects of the novel, orally available small-molecule inhibitor enzastaurin, LY317615.HCl). Blood, 2007, 109:1669–77. Available from: https://ashpublications.org/blood/article/109/4/1669/23434/ Targeting-PKC-in-multiple-myeloma-in-vitro-and-in., DOI 10.1182/blood-2006- 08-042747 Ni H, Ergin M, Tibudan SS, Denning MF, Izban KF, Alkan S. Protein kinase C-delta is commonly expressed in multiple myeloma cells and its downregulation by rottlerin causes apoptosis. Br J Haematol, 2003, 121(6):849–56., DOI 10.1046/j. 1365-2141.2003.04368.x Kirschner KM, Wagner N, Wagner KD, Wellmann S, Scholz H. The Wilms tumor suppressor Wt1 promotes cell adhesion through transcriptional activation of the alpha4integrin gene. J Biol Chem, 2006, 281(42):31930–9., DOI 10.1074/jbc. M602668200 Velecela V, Lettice LA, Chau YY, Slight J, Berry RL, Thornburn A, et al. WT1 regulates the expression of inhibitory chemokines during heart development. Hum Mol Genet, 2013, 22(25):5083–95., DOI 10.1093/hmg/ddt358 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610844 EP 1610851 DI 10.3389/pore.2023.1610844 PG 8 ER PT J AU Chen, T Peng, Y Lei, T Wu, Ch Wang, H Shi, Y AF Chen, Tongbing Peng, Yan Lei, Ting Wu, Chao Wang, Hui Shi, Yongqiang TI Low-grade oncocytic tumour (LOT) of the kidney is characterised by GATA3 positivity, FOXI1 negativity and mTOR pathway mutations SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; clinicopathological characteristics; mTOR; differential diagnosis; low-grade oncocytic tumour (LOT) of the kidney ID immunohistochemistry; clinicopathological characteristics; mTOR; differential diagnosis; low-grade oncocytic tumour (LOT) of the kidney AB Aims: We present a 5-case series of low-grade oncocytic tumour of the kidney to further discuss their clinicopathological characteristics. Methods and results: Five patients were included in this study. There were three females and two males aged 45–66 years, with a median age of 65 years. Four tumours were located in the right kidney, and one was located in the left kidney. Most of the tumour sections were yellow-brown in colour. Tumour sizes ranged from 2.5 to 4.5 cm, with a median size of 3 cm. Microscopically, the tumours were well-circumscribed but lacked a fibrous capsule; the tumours consisted of monomorphous oncocytic cells arranged mainly in solid and nested architectural patterns. The tumour cells had uniformly round to oval nuclei and often had perinuclear halos but lacked significant irregularities. Immunohistochemically, the tumour cells showed a diffuse and strong positivity forCK7 andwere negative for CD117. The tumour cellswere also positive for GATA3, E-cadherin, Pax-8, Succinate dehydrogenase B (SDHB) and Fumarate hydratase (FH), and negative for vimentin, Carbonic anhydrase 9 (CA9), CD10, P504s, CK20, TFE3, TFEB, HMB45, ALK and Forkhead box protein I1 (FOXI1). Next-generation sequencing identified genetic variations in these tumours, including MTOR gene mutations (4/5) and PIK3CA gene mutation (1/5). All patients were alive without disease progression at a median follow-up of 32 months (range 10–57months). Conclusion: LOT is an emerging renal entity of indolent behaviour that has morphologic overlap with some renal tumours with eosinophilic cytoplasm, primarily with oncocytoma and eosinophilic variant of chromophobe renal cell carcinoma. Familiarity with the distinctive morphological features, immunophenotype and molecular genetics of LOT helps avoid misdiagnosis. C1 [Chen, Tongbing] The Third Affiliated Hospital of Soochow University, Department of PathologyChangzhou, China. [Peng, Yan] The Third Affiliated Hospital of Soochow University, Department of PathologyChangzhou, China. [Lei, Ting] The Third Affiliated Hospital of Soochow University, Department of PathologyChangzhou, China. [Wu, Chao] The Third Affiliated Hospital of Soochow University, Department of PathologyChangzhou, China. [Wang, Hui] The Third Affiliated Hospital of Soochow University, Department of PathologyChangzhou, China. [Shi, Yongqiang] The Third Affiliated Hospital of Soochow University, Department of PathologyChangzhou, China. RP Peng, Y (reprint author), The Third Affiliated Hospital of Soochow University, Department of Pathology, Changzhou, China. EM pengyan871005@163.com CR Trpkov K, Hes O. New and emerging renal entities: A perspective post-WHO 2016 classification. Histopathology, 2019, 74(1):31–59., DOI 10.1111/his.13727 Trpkov K, Williamson SR, Gao Y, Martinek P, Cheng L, Sangoi AR, et al. Lowgrade oncocytic tumour of kidney, CD117-negative, cytokeratin 7-positive): A distinct entity? Histopathology, 2019, 75(2):174–84., DOI 10.1111/his.13865 Akgul M, Al-Obaidy KI, Cheng L, Idrees MT. Low-grade oncocytic tumour expands the spectrum of renal oncocytic tumours and deserves separate classification: A review of 23 cases from a single tertiary institute. J Clin Pathol, 2021, 75:772–5., DOI 10.1136/jclinpath-2021-207478 Guo Q, Liu N, Wang F, Guo Y, Yang B, Cao Z, et al. Characterization of a distinct low-grade oncocytic renal tumor, CD117-negative and cytokeratin 7- positive, based on a tertiary oncology center experience: The new evidence from China. Virchows Arch, 2021, 478(3):449–58., DOI 10.1007/s00428-020-02927-0 Kravtsov O, Gupta S, Cheville JC, Sukov WR, Rowsey R, Herrera-Hernandez LP, et al. Low-Grade Oncocytic Tumor of Kidney, CK7-Positive, CD117-Negative): Incidence in a single institutional experience with clinicopathological andmolecular characteristics. Hum Pathol, 2021, 114:9–18., DOI 10.1016/j.humpath.2021.04.013 Lerma LA, Schade GR, Tretiakova MS. Co-existence of ESC-RCC, EVT, and LOT as synchronous and metachronous tumors in six patients with multifocal neoplasia but without clinical features of tuberous sclerosis complex. Hum Pathol, 2021, 116:1–11., DOI 10.1016/j.humpath.2021.06.002 Bai YF, Chang CD, Wang B, Zhao M, Teng XD. CK7+/CD117-low grade oncocytic tumor of the kidney: A clinicopathological analysis. Zhonghua Bing Li Xue Za Zhi, 2022, 51(10):976–80., DOI 10.3760/cma.j.cn112151-20220719-00627 Kapur P, Gao M, Zhong H, Chintalapati S, MituiM, Barnes SD, et al. Germline and sporadic mTOR pathway mutations in low-grade oncocytic tumor of the kidney. Mod Pathol, 2022, 35(3):333–43., DOI 10.1038/s41379-021-00896-6 Morini A, Drossart T, Timsit MO, Sibony M, Vasiliu V, Gimenez-Roqueplo AP, et al. Low-grade oncocytic renal tumor, LOT): Mutations in mTOR pathway genes and low expression of FOXI1. Mod Pathol, 2022, 35(3):352–60., DOI 10.1038/ s41379-021-00906-7 Xie B, Cheng LC, Yin GL, Liu BA, Hu ZL, Tong K. Clinicopathological features of low-grade oncocytic renal tumor, CD117-negative, cytokeratin 7- positive): Report of seven cases. Zhonghua Bing Li Xue Za Zhi, 2022, 51(8): 719–25., DOI 10.3760/cma.j.cn112151-20220410-00263 Zhang HZ, Xia QY, Wang SY, Shi MJ. Low-grade oncocytic tumor of kidney harboring TSC/MTOR mutation: Clinicopathologic, immunohistochemical and molecular characteristics support a distinct entity. Virchows Arch, 2022, 480(5): 999–1008., DOI 10.1007/s00428-022-03283-x Williamson SR, Hes O, Trpkov K, Aggarwal A, Satapathy A, Mishra S, et al. Low-grade oncocytic tumour of the kidney is characterised by genetic alterations of TSC1, TSC2, MTOR or PIK3CA and consistent GATA3 positivity. Histopathology, 2022, 82:296–304., DOI 10.1111/his.14816 Xia QY,WangXT, Zhao M, HeHY, Fang R, Ye SB, et al. TSC/MTOR -associated eosinophilic renal tumors exhibit a heterogeneous clinicopathologic spectrum: A targeted next-generation sequencing and gene expression profiling study. Am J Surg Pathol, 2022, 46(11):1562–76., DOI 10.1097/PAS.0000000000001955 WHO Classification of Tumours. Urinary and male genital tumors. 5th ed. Lyon, France): International Agency for Research on Cancer, 2022). Tong K, Hu Z. FOXI1 expression in chromophobe renal cell carcinoma and renal oncocytoma: A study of the cancer genome atlas transcriptome-based outlier mining and immunohistochemistry. Virchows Arch, 2021, 478(4):647–58., DOI 10. 1007/s00428-020-02900-x Miettinen M, McCue PA, Sarlomo-Rikala M, Rys J, Czapiewski P, Wazny K, et al. GATA3: A multispecific but potentially useful marker in surgical pathology: A systematic analysis of 2500 epithelial and nonepithelial tumors. Am J Surg Pathol, 2014, 38(1):13–22., DOI 10.1097/PAS.0b013e3182a0218f Mantilla JG, Antic T, Tretiakova M. GATA3 as a valuable marker to distinguish clear cell papillary renal cell carcinomas from morphologic mimics. Hum Pathol, 2017, 66:152–8., DOI 10.1016/j.humpath.2017.06.016 Al-Obaidy KI, Eble JN, Cheng L, Williamson SR, Sakr WA, Gupta N, et al. Papillary renal neoplasm with reverse polarity: A morphologic, immunohistochemical, and molecular study. Am J Surg Pathol, 2019, 43(8): 1099–111., DOI 10.1097/PAS.0000000000001288 Mansoor M, Siadat F, Trpkov K. Low-grade oncocytic tumor, LOT, - a new renal entity ready for a prime time: An updated review. Histol Histopathol, 2022, 37(5):405–13., DOI 10.14670/HH-18-435 Miricescu D, Balan DG, Tulin A, Stiru O, Vacaroiu IA,Mihai DA, et al. PI3K/ AKT/mTOR signalling pathway involvement in renal cell carcinoma pathogenesis, Review). Exp Ther Med, 2021, 21(5):540., DOI 10.3892/etm.2021.9972 Pivovarcikova K, Alaghehbandan R, Vanecek T, Ohashi R, Pitra T, Hes O. TSC/mTOR pathway mutation associated eosinophilic/oncocytic renal neoplasms: A heterogeneous group of tumors with distinct morphology, immunohistochemical profile, and similar genetic background. Biomedicines, 2022, 10(2):322., DOI 10. 3390/biomedicines10020322 Trpkov K, Abou-Ouf H, Hes O, Lopez JI, Nesi G, Comperat E, et al. Eosinophilic solid and cystic renal cell carcinoma, ESC RCC): Further morphologic and molecular characterization of ESC RCC as a distinct entity. Am J Surg Pathol, 2017, 41(10):1299–308., DOI 10.1097/PAS.0000000000000838 He H, Trpkov K, Martinek P, Isikci OT, Maggi-Galuzzi C, Alaghehbandan R, et al. High-grade oncocytic renal tumor": Morphologic, immunohistochemical, and molecular genetic study of 14 cases. Virchows Arch, 2018, 473(6):725–38., DOI 10. 1007/s00428-018-2456-4 Farcas M, Gatalica Z, Trpkov K, Swensen J, Zhou M, Alaghehbandan R, et al. Eosinophilic vacuolated tumor, EVT, of kidney demonstrates sporadic TSC/ MTOR mutations: Next-generation sequencing multi-institutional study of 19 cases. Mod Pathol, 2022, 35(3):344–51., DOI 10.1038/s41379-021-00923-6 Tsai TH, Lee WY. Succinate dehydrogenase-deficient renal cell carcinoma. Arch Pathol Lab Med, 2019, 143(5):643–7., DOI 10.5858/arpa.2018-0024-RS Xia QY, Wang XT, Fang R, Wang Z, Zhao M, Chen H, et al. Clinicopathologic and molecular analysis of the TFEB fusion variant reveals new members of TFEB translocation renal cell carcinomas, RCCs): Expanding the genomic spectrum. Am J Surg Pathol, 2020, 44(4):477–89., DOI 10.1097/PAS. 0000000000001408 Smith SC, Sirohi D, Ohe C, McHugh JB, Hornick JL, Kalariya J, et al. A distinctive, low-grade oncocytic fumarate hydratase-deficient renal cell carcinoma, morphologically reminiscent of succinate dehydrogenase-deficient renal cell carcinoma. Histopathology, 2017, 71(1):42–52., DOI 10.1111/his.13183 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610852 EP 1610858 DI 10.3389/pore.2023.1610852 PG 7 ER PT J AU Wang, Z Wang, J Zhang, W Wang, D Wang, X Liang, X AF Wang, Zhuo Wang, Jinsui Zhang, Wenwen Wang, Daoying Wang, Xiaojun Liang, Xiaoqin TI Case report: Urothelial carcinoma of the renal pelvis with trophoblastic differentiation: A rare case report and review of literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE urothelial carcinoma; renal pelvis; trophoblastic differentiation; beta-human chorionic gonadotropin; syncytiotrophoblastic cells ID urothelial carcinoma; renal pelvis; trophoblastic differentiation; beta-human chorionic gonadotropin; syncytiotrophoblastic cells AB We report a rare case of urothelial carcinoma (UC) of the renal pelvis with trophoblastic differentiation that occurred in a 55-year-old male patient. The patient presented with gross hematuria and paroxysmal lumbago pain 5 months ago. The enhanced computed tomography (CT) scan demonstrated a large space occupying lesion in the left kidney and multiple retroperitoneal lymph node enlargements. Histologically, high-grade infiltrating urothelial carcinoma (HGUC) contained giant cells which were positive for beta-human chorionic gonadotropin (β-hCG). Three weeks after resection, positron emission tomography and computed tomography (PET-CT) scan showed multiple nodules of metastasis in the left renal region, extensive systemic muscle, bone, lymph node, liver and bilateral lung metastases. The patient underwent bladder perfusion chemotherapy and gemcitabine combined with cisplatin chemotherapy regimens. This is the eighth documented case of UC of the renal pelvis with trophoblastic differentiation. Due to its rarity and extremely poor prognosis, it is important to clarify the characteristics of the disease and make an accurate and prompt diagnosis. C1 [Wang, Zhuo] Gansu Province People’s Hospital, Department of PathologyLanzhou, Gansu, China. [Wang, Jinsui] Gansu Province People’s Hospital, Department of PathologyLanzhou, Gansu, China. [Zhang, Wenwen] Gansu Province People’s Hospital, Department of RadiologyLanzhou, Gansu, China. [Wang, Daoying] Gansu Province People’s Hospital, Department of the Centre of Positron Emission Tomography-Computed TomographyLanzhou, Gansu, China. [Wang, Xiaojun] Gansu Province People’s Hospital, Department of Respiratory MedicineLanzhou, Gansu, China. [Liang, Xiaoqin] Gansu Province People’s Hospital, Department of PathologyLanzhou, Gansu, China. RP Liang, X (reprint author), Gansu Province People’s Hospital, Department of Pathology, Lanzhou, China. EM liangxq905@163.com CR Elias C, Ferran A, Antonio P. Placental proteins in high-grade urothelial neoplasms an lmmunohistochemical study of human chorionic gonadotropin, human placental lactogen, and pregnancy-specific beta-1-glycoprotein. Cancer, 1989, 63(6):2497–504. Winfried V, Urs R, Berthold W, Ihling C. Beta-human chorionic gonadotropin -positive extragonadal germcell neoplasia of the renal pelvis. Cancer, 1991, 67(6):3146–9., DOI 10.1002/1097-0142(19910615)67:12<3146:aid-cncr2820671235>3.0.co;2-g Dino G, David G, Parker E, Shepherd RR, Hearn SA, Walton JC. Transitional cell carcinoma of the renal pelvis with choriocarcinomatous differentiation. Immunohistochemical and immunoelectron microscopic assessment of human chorionic gonadotropin production by transitional cell carcinoma of the urinary bladder. Cancer, 1993, 71(8):1835–41., DOI 10.1002/1097-0142(19930301)71: 5<1835:aid-cncr2820710519>3.0.co;2-5 Zettl A, Konrad MA, Polzin S, Ehsan A, Riedmiller H, Muller-Hermelink HK, et al. Urothelial carcinoma of the renal pelvis with choriocarcinomatous features: Genetic evidence of clonal evolution. Hum Pathol, 2002, 33(12):1234–7., DOI 10. 1053/hupa.2002.129208 Preda O, Dema A, Iacob M, Goyenaga P, Dulcey I, Aneiros Fernandez J, et al. Urothelial carcinoma of the renal pelvis with simultaneous trophoblastic and malignant clear cell endodermal-type differentiation. Virchows Arch, 2012, 460(3):353–6., DOI 10.1007/s00428-012-1211-5 Msaouel P, Zhang M, Tu SM. Prolonged remission of upper urinary tract urothelial carcinoma with prominent choriocarcinomatous differentiation: A case report. Clin Genitourin Cancer, 2017, 15(1):e73–7., DOI 10.1016/j.clgc. 2016.08.025 Rolim I, Henriques V, Rolim N, Blanca A, Marques RC, Volavsek M, et al. Clinicopathologic analysis of upper urinary tract carcinoma with variant histology. Virchows Arch, 2020, 477(1):111–20., DOI 10.1007/s00428-020-02745-4 Rajabi B, Khoury J, Brewer C, Goodman OB. Urothelial bladder carcinoma with choriocarcinomatous differentiation presenting with a false-positive pregnancy test. Am J Med Sci, 2013, 346(3):256–8., DOI 10.1097/MAJ. 0b013e318270d0f9 Inoue S, Mizushima T, Fujita K, Meliti A, Ide H, Yamaguchi S, et al. GATA3 immunohistochemistry in urothelial carcinoma of the upper urinary tract as a urothelial marker and a prognosticator. Hum Pathol, 2017, 64(17): 83–90., DOI 10.1016/j.humpath.2017.04.003 Yi L, Hitoshi I, Takashi K,Miyamoto Y, Izumi K,Miyamoto H. GATA3 in the urinary bladder: Suppression of neoplastic transformation and down-regulation by androgens. Am J Cancer Res, 2014, 4(5):461–73. Miyamoto H, Izumi K, Yao JL, Li Y, Yang Q, McMahon LA, et al. GATA binding protein 3 is down-regulated in bladder cancer yet strong expression is an independent predictor of poor prognosis in invasive tumor. Hum Pathol, 2012, 43(11):2033–40., DOI 10.1016/j.humpath.2012.02.011 Cheng H-L, Chou L-P, Tsai H-W, Lee CT, Wang YW, Chung-Liang H, et al. Urothelial carcinoma with trophoblastic differentiation: Reappraisal of the clinical implication and immunohistochemically features. Urol Oncol, 2021, 39(10): 732.e17–732.732.e23., DOI 10.1016/j.urolonc.2021.03.006 John A, Norman R, Leopold G. Primary choriocarcinoma of the urinary bladder Report of a case with probable epithelial Origin. cancer, 1983, 52(6):1405–9., DOI 10.1002/ 1097-0142(19831015)52:8<1405:aid-cncr2820520812>3.0.co;2-t Amin MB. Histological variants of urothelial carcinoma: Diagnostic, therapeutic and prognostic implications. Mod Pathol, 2009, 22(2):S96–118., DOI 10.1038/modpathol.2009.26 Regalado JJ. Mixed micropapillary and trophoblastic carcinoma of bladder: Report of a first case with new immunohistochemical evidence of urothelial origin. Hum Pathol, 2004, 35(3):382–4., DOI 10.1016/j.humpath.2003.09.012 Jenkins BJ, Martin JE, Baithun SI, Zuk RJ, Oliver RTD, Blandy JP. Prediction of response to radiotherapy in invasive bladder cancer. Br Journalof Urol, 1990, 1990(65):345–8., DOI 10.1111/j.1464-410x.1990.tb14753.x Uchida M, Kawai K, Kurobe M, Ikeda A, Kandori S, Endo T, et al. Metastases of urothelial carcinoma with trophoblastic differentiation that responded to combination chemotherapy with gemcitabine and oxaliplatin: A case report. A Case Rep, 2018, 64(2):55–61., DOI 10.14989/ActaUrolJap_64_2_55 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610856 EP 1610862 DI 10.3389/pore.2023.1610856 PG 7 ER PT J AU Dapkeviciutė-Purlienė, A Augustinavicius, V Zucenka, A AF Dapkeviciutė-Purlienė, Austėja Augustinavicius, Vytautas Zucenka, Andrius TI Case report: Relapsed/refractory extranodal natural killer/T-cell lymphoma nasal type with extensive central nervous system involvement SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE case report; lymphoma; NK-cell; T-cell; central nervous system ID case report; lymphoma; NK-cell; T-cell; central nervous system AB Background: Extranodal natural killer/T-cell lymphoma (ENKL) is a rare subtype of mature T and natural killer cell lymphomas associated with Epstein-Barr virus. Case: A 20-year-old presented with severe neurological symptoms and was diagnosed with stage IV ENKL, nasal type, with CNS involvement. Overall, the patient received nine treatment lines, including chemotherapy, craniospinal irradiation, allogeneic stem cell transplant (alloSCT), donor lymphocyte infusions, and novel agents (Nivolumab, Daratumumab, Thalidomide, Lenalidomide, virus-specific T cells) combined with intrathecal chemotherapy. The treatment effect was evaluated in both blood and CSF (cerebrospinal fluid). First-line SMILE chemotherapy resulted in systemic and CNS remission. Later Cytarabine-based chemotherapy and Daratumumab combination helped to reinduce remission before alloSCT. Conclusion: We show that efficacy monitoring should include both blood and CSF analysis. High-dose Cytarabine-based chemotherapy in combination with Daratumumab and intrathecal chemotherapy may be considered as salvage CNS-directed therapies. We add to existing limited data that Daratumumab penetrates the blood-brain barrier. C1 [Dapkeviciutė-Purlienė, Austėja] Vilnius University, Faculty of Medicine, Institute of Clinical MedicineVilnius, Lithuania. [Augustinavicius, Vytautas] Vilnius University, Faculty of Medicine, Department of Radiology, Nuclear Medicine and Physics of MedicineVilnius, Lithuania. [Zucenka, Andrius] Vilnius University, Faculty of Medicine, Institute of Clinical MedicineVilnius, Lithuania. RP Dapkeviciutė-Purlienė, A (reprint author), Vilnius University, Faculty of Medicine, Institute of Clinical Medicine, Vilnius, Lithuania. EM austeja.dapkeviciute@santa.lt CR Yamaguchi M, Suzuki R, Oguchi M. Advances in the treatment of extranodal NK/T-cell lymphoma, nasal type [Internet]. Blood., 2018, 131131(23):2528–40., DOI 10.1182/blood-2017-12-791418 Yamaguchi M, Miyazaki K. Current treatment approaches for NK/T-cell lymphoma. J Clin Exp Hematop 57:98–108., DOI 10.3960/jslrt. 17018 Yang Y, Li Z, Zhiyang C, Liang H. Extranodal natural killer/T-cell lymphoma nasal type with central nervous system involvement mimicked tuberculous meningitis: A case report. Medicine, Baltimore, 98(34):e16747. Romero-Guadarrama MB, Aguilar-Martinez E. Extranodal nasal NK/T-cell lymphoma with dissemination to the central nervous system: A case report. Acta Cytol, 2010, 54(5):993–7. Tse E, Kwong YL. Diagnosis and management of extranodal NK/T cell lymphoma nasal type. Expert Rev Hematol 99(9):861–71., DOI 10.1080/17474086. 2016.1206465 Yanagisawa R, Nakazawa Y, Sakashita K, Saito S, Tanaka M, Shiohara M, et al. Intrathecal donor lymphocyte infusion for isolated leukemia relapse in the central nervous system following allogeneic stem cell transplantation: A case report and literature review. Int J Hematol 103:107–11., DOI 10.1007/s12185-015-1902-1 Zhao J, Zu Y, Han L, Zhang Y, Gui R, Yu F, et al. Treatment of Epstein–Barr virus associated central nervous system diseases after allogeneic hematopoietic stem cell transplantation with intrathecal donor lymphocyte infusion. Bone Marrow Transpl, 2019, 54(6):821–7., DOI 10.1038/s41409-018-0409-9 YamaguchiM, Oguchi M, Suzuki R. Extranodal NK/T-cell lymphoma: Updates in biology and management strategies [Internet]. Best Pract Res Clin Haematol, 2018, 3131(3):315–21., DOI 10.1016/j.beha.2018.07.002 Chan TSY, Li J, Loong F, Khong PL, Tse E, Kwong YL. PD1 blockade with lowdose nivolumab in NK/T cell lymphoma failing l-asparaginase: Efficacy and safety [internet]. Ann Hematol 9797(1):193–6., DOI 10.1007/s00277-017-3127-2 Li X, Cheng Y, ZhangM, Yan J, Li L, Fu X, et al. Activity of pembrolizumab in relapsed/refractory NK/T-cell lymphoma. J Hematol Oncol, 2018, 11(1):15., DOI 10. 1186/s13045-018-0559-7 Kwong YL, Chan TSY, Tan D, Kim SJ, Poon LM, Mow B, et al. PD1 blockade with pembrolizumab is highly effective in relapsed or refractory NK/T-cell lymphoma failing L-asparaginase. Blood., 2017, 129(17):2437–42., DOI 10.1182/ blood-2016-12-756841 AbidH,Watthanasuntorn K, Shah O, Gnanajothy R. Efficacy of pembrolizumab and nivolumab in crossing the blood brain barrier. Cureus, 2019, 11(4):e4446., DOI 10.7759/ cureus.4446 Van Bussel MTJ, Beijnen JH, Brandsma D. Intracranial antitumor responses of nivolumab and ipilimumab: A pharmacodynamic and pharmacokinetic perspective, a scoping systematic review. BMC Cancer, 2019, 19(1):519., DOI 10. 1186/s12885-019-5741-y Fan S, Ren H, Zhao L, Yin J, Feng G, Wang J, et al. Neurological immunerelated adverse events associated with immune checkpoint inhibitors: A review of the literature. Asia Pac J Clin Oncol, 2020, 16(6):291–8., DOI 10.1111/ajco.13375 Kim W-S, Eom H-S, Yeh S-P, Cho S-G, Heo DS, Kim JS, et al. Daratumumab monotherapy for patients with relapsed or refractory, R/R, natural killer/T-cell lymphoma, NKTCL), nasal type: An open-label, single-arm, multicenter phase 2 study. Blood, 2018, 132(1):1617., DOI 10.1182/blood-2018-99-111885 Varga G, Mikala G, Gopcsa L, Csukly Z, Kollai S, Balazs G, et al. Multiple myeloma of the central nervous system: 13 cases and review of the literature. J Oncol, 2018, 2018:3970169., DOI 10.1155/2018/3970169 Ferreri AJ, Reni M, Foppoli M, Martelli M, Pangalis GA, Frezzato M, et al. High-dose cytarabine plus high-dose methotrexate versus high-dose methotrexate alone in patients with primary CNS lymphoma: A randomised phase 2 trial. Lancet, 2009, 374(9700):1512–20., DOI 10.1016/S0140-6736(09)61416-1 DeMel S, Hue SSS, Jeyasekharan AD, Chng WJ, Ng SB. Molecular pathogenic pathways in extranodal NK/T cell lymphoma. J Hematol Oncol 12(1):33., DOI 10. 1186/s13045-019-0716-7 Wu H, Zhao C, Gu K, Jiao Y, Hao J, Sun G. Thalidomide plus chemotherapy exhibit enhanced efficacy in the clinical treatment of T-cell non-hodgkin’s lymphoma: A prospective study of 46 cases. Mol Clin Oncol 2: 695–700., DOI 10.3892/mco.2014.307 Wang L, Wang ZH, Chen XQ, Wang KF, Huang HQ, Xia ZJ. First-line combination of GELOX followed by radiation therapy for patients with stage IE/IIE ENKTL: An updated analysis with long-term follow-up. Oncol Lett 10(2):1036–40., DOI 10.3892/ol.2015.3327 Kumar A, Gupta P, Kaushal V. Locally advanced angiocentric nk/t cell lymphoma of nasal cavity. J Cancer Prev Curr Res, 2018, 9(6):00366., DOI 10.15406/ jcpcr.2018.09.00366 Muscal JA, Sun Y, Nuchtern JG, Dauser RC, McGuffey LH, Gibson BW, et al. Plasma and cerebrospinal fluid pharmacokinetics of thalidomide and lenalidomide in nonhuman primates. Cancer Chemother Pharmacol 69(4):943–7., DOI 10.1007/ s00280-011-1781-y Rubenstein JL, Geng H, Fraser EJ, Formaker P, Chen L, Sharma J, et al. Phase 1 investigation of lenalidomide/rituximab plus outcomes of lenalidomide maintenance in relapsed CNS lymphoma. Blood Adv 2(13):1595–607., DOI 10. 1182/bloodadvances.2017014845 McLaughlin LP, Rouce R, Gottschalk S, Torrano V, Carrum G, Wu MF, et al. EBV/LMP-specific T cells maintain remissions of T- and B-cell EBV lymphomas after allogeneic bone marrow transplantation. Blood 132(22): 2351–61., DOI 10.1182/blood-2018-07-863654 Muftuoglu M, Olson A, Marin D, Ahmed S, Mulanovich V, Tummala S, et al. Allogeneic BK virus–specific T cells for progressive multifocal leukoencephalopathy. N Engl J Med 379(15):1443–51., DOI 10.1056/ NEJMoa1801540 Seo S, Kami M, Honda H, Kashima T, Matsumura T, Moriya A, et al. Extramedullary relapse in the so-called “sanctuary” sites for chemotherapy after donor lymphocyte infusion. Bone Marrow Transplant, 2000, 25(2):226–7., DOI 10.1038/sj.bmt.1702116 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610866 EP 1610872 DI 10.3389/pore.2022.1610866 PG 7 ER PT J AU Inukai, K Kise, K Hayashi, Y Jia, W Muramatsu, F Okamoto, N Konishi, H Akuta, K Kidoya, H Takakura, N AF Inukai, Koichi Kise, Kazuyoshi Hayashi, Yumiko Jia, Weizhen Muramatsu, Fumitaka Okamoto, Naoki Konishi, Hirotaka Akuta, Keigo Kidoya, Hiroyasu Takakura, Nobuyuki TI Cancer apelin receptor suppresses vascular mimicry in malignant melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE epithelial-mesenchymal transition; melanoma; apelin; apelin receptor; neovascularization ID epithelial-mesenchymal transition; melanoma; apelin; apelin receptor; neovascularization AB Several reports indicate that apelin is often over-expressed in tumors, and therefore it has been suggested that the apelin–apelin receptor (APJ) system may induce tumor progression. In contrast, our previous research revealed high expression of the apelin–APJ system in tumor blood vessels, suggesting its involvement in the regulation of tumor vessel formation and normalization, resulting in the suppression of tumor growth by promoting the infiltration of T cells. Thus, the effect of the apelin–APJ system on tumors remains controversial. In this report, to clarify the effect of apelin in tumor cells, we analyzed the function of APJ in tumor cells using APJ knock out (KO) mice. In APJ-KO mice, Apelin overexpression in B16/BL6 (B16) melanoma cells induced greater tumor growth than controls. In an APJ-KO melanoma inoculation model, although angiogenesis is suppressed compared to wild type, no difference is evident in tumor growth. We found that APJ deficiency promoted vascular mimicry in tumors. In vitro, cultured APJ-KO B16 cells demonstrated a spindle-like shape. This phenotypic change was thought to be induced by epithelial–mesenchymal transition (EMT) based on evidence that APJ-KO B16 cells show persistently high levels of the mesenchymal maker, Zeb1; however, we found that EMT did not correlate with the transforming growth factor-β/smad signaling pathway in our model. We propose that apelin- APJ system in cancer cells induces tumor growth but negatively regulates EMT and tumor malignancy. C1 [Inukai, Koichi] Osaka University, Research Institute for Microbial Diseases, Department of Signal TransductionSuita, Japan. [Kise, Kazuyoshi] Osaka University, Research Institute for Microbial Diseases, Department of Signal TransductionSuita, Japan. [Hayashi, Yumiko] University of Fukui, Faculty of Medical Sciences, Department of Integrative Vascular BiologyFukui, Japan. [Jia, Weizhen] Osaka University, Research Institute for Microbial Diseases, Department of Signal TransductionSuita, Japan. [Muramatsu, Fumitaka] Osaka University, Research Institute for Microbial Diseases, Department of Signal TransductionSuita, Japan. [Okamoto, Naoki] Osaka University, Research Institute for Microbial Diseases, Department of Signal TransductionSuita, Japan. [Konishi, Hirotaka] Osaka University, Research Institute for Microbial Diseases, Department of Signal TransductionSuita, Japan. [Akuta, Keigo] Osaka University, Research Institute for Microbial Diseases, Department of Signal TransductionSuita, Japan. [Kidoya, Hiroyasu] University of Fukui, Faculty of Medical Sciences, Department of Integrative Vascular BiologyFukui, Japan. [Takakura, Nobuyuki] Osaka University, Research Institute for Microbial Diseases, Department of Signal TransductionSuita, Japan. RP Takakura, N (reprint author), Osaka University, Research Institute for Microbial Diseases, Department of Signal Transduction, Suita, Japan. EM ntakaku@biken.osaka-u.ac.jp CR Serene HL, Tian J, Reversade B. Elabela: A hormone essential for heart development signals via the apelin receptor. Dev Cel, 2013, 27(6):672–80., DOI 10.1016/j.devcel.2013.11.002 Perjes A, Kilpio T, Ulvila J, Magga J, Alakoski T, Szabo Z, et al. Characterization of apela, a novel endogenous ligand of apelin receptor, in the adult heart. Basic Res Cardiol, 2016, 111(1):2., DOI 10.1007/s00395-015-0521-6 Kuba K, Sato T, Imai Y, Yamaguchi T. Apelin and Elabela/Toddler; double ligands for APJ/Apelin receptor in heart development, physiology, and pathology. Peptides, 2019, 111:62–70., DOI 10.1016/j.peptides.2018.04.011 Busch R, Strohbach A, Pennewitz M, Lorenz F, Bahls M, Busch MC, et al. Regulation of the endothelial apelin/APJ system by hemodynamic fluid flow. Cell Signal, 2015, 27(7):1286–96., DOI 10.1016/j.cellsig.2015.03.011 Kidoya H, Ueno M, Yamada Y, Mochizuki N, Nakata M, Yano T, et al. Spatial and temporal role of the apelin/APJ system in the caliber size regulation of blood vessels during angiogenesis. EMBO J, 2008, 27(3):522–34., DOI 10.1038/sj.emboj. 7601982 Kidoya H, Takakura N. Maturation of blood vessels by haematopoietic stem cells and progenitor cells: Involvement of apelin/APJ and angiopoietin/ Tie2 interactions in vessel caliber size regulation. Thromb Haemost, 2009, 101(06):999–1005., DOI 10.1160/th08-06-0358 Kidoya H, Naito H, Takakura N. Apelin induces enlarged and nonleaky blood vessels for functional recovery from ischemia. Blood, 2010, 115(15):3166–74., DOI 10.1182/blood-2009-07-232306 Kidoya H, Naito H, Muramatsu F, Yamakawa D, Jia W, Ikawa M, et al. APJ regulates parallel alignment of arteries and veins in the skin. Dev Cel, 2015, 33(3): 247–59., DOI 10.1016/j.devcel.2015.02.024 Frisch A, Kalin S, Monk R, Radke J, Heppner FL, Kalin RE. Apelin controls angiogenesis-dependent glioblastoma growth. Int J Mol Sci, 2020, 21(11):4179., DOI 10.3390/ijms21114179 Masoumi J, Jafarzadeh A, Khorramdelazad H, Abbasloui M, Abdolalizadeh J, Jamali N. Role of Apelin/APJ axis in cancer development and progression. AdvMed Sci, 2020, 65(1):202–13., DOI 10.1016/j.advms.2020.02.002 Zhang L, Takara K, Yamakawa D, Kidoya H, Takakura N. Apelin as a marker for monitoring the tumor vessel normalization window during antiangiogenic therapy. Cancer Sci, 2016, 107(1):36–44., DOI 10.1111/cas.12836 Kidoya H, Kunii N, Naito H, Muramatsu F, Okamoto Y, Nakayama T, et al. The apelin/APJ system induces maturation of the tumor vasculature and improves the efficiency of immune therapy. Oncogene, 2012, 31(27):3254–64., DOI 10.1038/onc.2011.489 Hu L, Hayashi Y, Kidoya H, Takakura N. Endothelial cell-derived Apelin inhibits tumor growth by altering immune cell localization. Sci Rep, 2021, 11(1): 14047., DOI 10.1038/s41598-021-93619-5 Berta J, Torok S, Tarnoki-Zach J, Drozdovszky O, Tovari J, Paku S, et al. Apelin promotes blood and lymph vessel formation and the growth of melanoma lung metastasis. Sci Rep, 2021, 11(1):5798., DOI 10.1038/s41598-021-85162-0 Berta J, Kenessey I, Dobos J, Tovari J, Klepetko W, Jan Ankersmit H, et al. Apelin expression in human non-small cell lung cancer: Role in angiogenesis and prognosis. J Thorac Oncol, 2010, 5(8):1120–9., DOI 10.1097/jto.0b013e3181e2c1ff Ishida J, Hashimoto T, Hashimoto Y, Nishiwaki S, Iguchi T, Harada S, et al. Regulatory roles for APJ, a seven-transmembrane receptor related to angiotensintype 1 receptor in blood pressure in vivo. J Biol Chem, 2004, 279(25):26274–9., DOI 10.1074/jbc.m404149200 Carpentier G, Berndt S, Ferratge S, Rasband W, Cuendet M, Uzan G, et al. Angiogenesis analyzer for ImageJ — a comparative morphometric analysis of “endothelial tube formation assay” and “fibrin bead assay”. Sci Rep, 2020, 10(1): 11568., DOI 10.1038/s41598-020-67289-8 Zudaire E, Gambardella L, Kurcz C, Vermeren S. A computational tool for quantitative analysis of vascular networks. PLoS One, 2011, 6(11):e27385., DOI 10. 1371/journal.pone.0027385 Morales-Guadarrama G, Garcia-Becerra R, Mendez-Perez EA, Garcia-Quiroz J, Avila E, Diaz L. Vasculogenic mimicry in breast cancer: Clinical relevance and drivers. Cells, 2021, 10(7):1758., DOI 10.3390/cells10071758 Brabletz S, Schuhwerk H, Brabletz T, Stemmler MP. Dynamic EMT: A multitool for tumor progression. EMBO J, 2021, 40(18):e108647., DOI 10.15252/embj. 2021108647 Yang L, Su T, Lv D, Xie F, Liu W, Cao J, et al. ERK1/2 mediates lung adenocarcinoma cell proliferation and autophagy induced by apelin-13. Acta Biochim Biophys Sinica, 2014, 46(2):100–11., DOI 10.1093/abbs/gmt140 Chen T, Liu N, Xu GM, Liu TJ, Liu Y, Zhou Y, et al. Apelin13/APJ promotes proliferation of colon carcinoma by activating Notch3 signaling pathway. Oncotarget, 2017, 8(60):101697–706., DOI 10.18632/oncotarget. 21904 Heo K, Kim YH, Sung HJ, Li HY, Yoo CW, Kim JY, et al. Hypoxia-induced up-regulation of apelin is associated with a poor prognosis in oral squamous cell carcinoma patients. Oral Oncol, 2012, 48(6):500–6., DOI 10.1016/j.oraloncology. 2011.12.015 Wechman SL, Emdad L, Sarkar D, Das SK, Fisher PB. Vascular mimicry: Triggers, molecular interactions and in vivo models. Adv Cancer Res, 2020, 148: 27–67., DOI 10.1016/bs.acr.2020.06.001 Zhao N, Sun B-C, Sun T, Ma Y-M, Zhao X-L, Liu Z-Y, et al. Hypoxia-induced vasculogenic mimicry formation via VE-cadherin regulation by Bcl-2. Med Oncol, 2012, 29(5):3599–607., DOI 10.1007/s12032-012-0245-5 Liu Q, Qiao L, Liang N, Xie J, Zhang J, Deng G, et al. The relationship between vasculogenic mimicry and epithelial-mesenchymal transitions. J Cel Mol Med, 2016, 20(9):1761–9., DOI 10.1111/jcmm.12851 Bussolati B, Grange C, Sapino A, Camussi G. Endothelial cell differentiation of human breast tumour stem/progenitor cells. J Cel Mol Med, 2009, 13(2):309–19., DOI 10.1111/j.1582-4934.2008.00338.x Xu X, Zong Y, Gao Y, Sun X, Zhao H, Luo W, et al. VEGF induce vasculogenic mimicry of choroidal melanoma through the PI3k signal pathway. Biomed Res Int, 2019, 2019:3909102–13., DOI 10.1155/2019/3909102 Seftor RE, Seftor EA, Koshikawa N, Meltzer PS, Gardner LM, Bilban M, et al. Cooperative interactions of laminin 5 gamma2 chain, matrix metalloproteinase-2, and membrane type-1-matrix/metalloproteinase are required for mimicry of embryonic vasculogenesis by aggressive melanoma. Cancer Res, 2001, 61(17): 6322–7. Hendrix MJ, Seftor RE, Seftor EA, Gruman LM, Lee LM, Nickoloff BJ, et al. Transendothelial function of human metastatic melanoma cells: Role of the microenvironment in cell-fate determination. Cancer Res, 2002, 62(3): 665–8. Zhang S, Li M, Gu Y, Liu Z, Xu S, Cui Y, et al. Thalidomide influences growth and vasculogenic mimicry channel formation in melanoma. J Exp Clin Cancer Res, 2008, 27:60., DOI 10.1186/1756-9966-27-60 Zhang X, Luo H. Effects of thalidomide on growth and VEGF-A expression in SW480 colon cancer cells. Oncol Lett, 2018, 15(3):3313–20., DOI 10.3892/ol.2017. 7645 Wels C, Joshi S, Koefinger P, Bergler H, Schaider H. Transcriptional activation of ZEB1 by slug leads to cooperative regulation of the epithelial–mesenchymal transition-like phenotype in melanoma. J Invest Dermatol, 2011, 131(9):1877–85., DOI 10.1038/jid.2011.142 Wei X, Chen Y, Jiang X, Peng M, Liu Y, Mo Y, et al. Mechanisms of vasculogenic mimicry in hypoxic tumor microenvironments. Mol Cancer, 2021, 20(1):7., DOI 10.1186/s12943-020-01288-1 Mastrella G, Hou M, Li M, Stoecklein VM, Zdouc N, Volmar MNM, et al. Targeting APLN/APLNR improves antiangiogenic efficiency and blunts proinvasive side effects of VEGFA/VEGFR2 blockade in glioblastoma. Cancer Res, 2019, 79(9):2298–313., DOI 10.1158/0008-5472.CAN-18-0881 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610867 EP 1610877 DI 10.3389/pore.2023.1610867 PG 11 ER PT J AU Wang, Sh Zheng, Q Wang, J Chen, Sh Chen, L AF Wang, Shaoyu Zheng, Qiaomei Wang, Jinhua Chen, Shaozhan Chen, Lihong TI Long non-coding RNA MYU promotes ovarian cancer cell proliferation by sponging miR-6827-5p and upregulating HMGA1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lncRNA; ovarian cancer; HMGA1; MYU; miR-6827-5p ID lncRNA; ovarian cancer; HMGA1; MYU; miR-6827-5p AB Background: Long non-coding RNAs (lncRNAs) have been confirmed to play vital roles in tumorigenesis. LncRNA MYU has recently been reported as an oncogene in several kinds of tumors. However, MYU’s expression status and potential involvement in ovarian cancer (OC) remain unclear. In this study, we explored the underlying role of MYU in OC. Methods and results: The expression of MYU was upregulated in OC tissues, and MYU’s overexpression was significantly correlated with the FIGO stage and lymphatic metastasis. Knockdown of MYU inhibited cell proliferation in SKOV3 and A2780 cells. Mechanistically, MYU directly interacted with miR- 6827-5p in OC cells; HMGA1 is a downstream target gene of miR-6827- 5p. Furthermore, MYU knockdown increased the expression of miR-6827-5p and decreased the ex pression of HMGA1. Restoration of HMGA1 expression reversed the influence on cell proliferation caused by MYU knockdown. Conclusion: MYU functions as a ceRNA that positively regulates HMGA1 expression by sponging miR-6827-5p in OC cells, which may provide a potential target and biomarker for the diagnosis or prognosis of OC. C1 [Wang, Shaoyu] Fujian Medical University, The First Affiliated Hospital, Department of Obstetrics and GynecologyFuzhou, China. [Zheng, Qiaomei] Fujian Medical University, The First Affiliated Hospital, Department of Obstetrics and GynecologyFuzhou, China. [Wang, Jinhua] Fujian Medical University, The First Affiliated Hospital, Department of Obstetrics and GynecologyFuzhou, China. [Chen, Shaozhan] Fujian Medical University, The First Affiliated Hospital, Department of Obstetrics and GynecologyFuzhou, China. [Chen, Lihong] Fujian Medical University, The First Affiliated Hospital, Department of Obstetrics and GynecologyFuzhou, China. RP Chen, L (reprint author), Fujian Medical University, The First Affiliated Hospital, Department of Obstetrics and Gynecology, Fuzhou, China. EM wscclh@163.com CR Xia C, Dong X, Li H, Cao M, Sun D, He S, et al. Cancer statistics in China and United States, 2022: Profiles, trends, and determinants. Chin Med J, 2022, 135(5): 584–90., DOI 10.1097/cm9.0000000000002108 Moufarrij S, DandapaniM, Arthofer E, Gomez S, Srivastava A, Lopez-Acevedo M, et al. Epigenetic therapy for ovarian cancer: Promise and progress. Clin epigenetics, 2019, 11(1):7., DOI 10.1186/s13148-018-0602-0 Torre LA, Trabert B, DeSantis CE, Miller KD, Samimi G, Runowicz CD, et al. Ovarian cancer statistics. CA Cancer J Clin, 2018, 68(4):284–96., DOI 10.3322/caac. 21456 Beermann J, Piccoli MT, Viereck J, Thum T. Non-coding RNAs in development and disease: Background, mechanisms, and therapeutic approaches. Physiol Rev, 2016, 96(4):1297–325., DOI 10.1152/physrev.00041.2015 Schaukowitch K, Kim TK. Emerging epigenetic mechanisms of long noncoding RNAs. Neuroscience, 2014, 264:25–38., DOI 10.1016/j.neuroscience.2013. 12.009 Mendell J. MicroRNAs: Critical regulators of development, cellular physiology and malignancy. Cell Cycle, Georgetown, Tex,, 2005, 4(9):1179–84., DOI 10.4161/cc. 4.9.2032 Qi X, Zhang DH, Wu N, Xiao JH, Wang X, Ma W. ceRNA in cancer: possible functions and clinical implications. J Med Genet, 2015, 52(10):710–8., DOI 10.1136/ jmedgenet-2015-103334 Kawasaki Y, Komiya M, Matsumura K, Negishi L, Suda S, Okuno M, et al. MYU, a target lncRNA for wnt/c-myc signaling, mediates induction of CDK6 to promote cell cycle progression. Cel Rep, 2016, 16(10):2554–64., DOI 10.1016/j.celrep. 2016.08.015 Wang J, Yang X, Li R, Wang L, Gu Y, Zhao Y, et al. Long non-coding RNA MYU promotes prostate cancer proliferation bymediating the miR-184/c-Myc axis. Oncol Rep, 2018, 40:2814–25., DOI 10.3892/or.2018.6661 Wang X, Chen Q,Wang X, LiW, Yu G, Zhu Z, et al. ZEB1 activated-VPS9D1- AS1 promotes the tumorigenesis and progression of prostate cancer by sponging miR-4739 to upregulate MEF2D. Biomed Pharmacother, 2020, 122:109557., DOI 10. 1016/j.biopha.2019.109557 Tan J, Yang L. Long noncoding RNA VPS9D1-AS1 overexpression predicts a poor prognosis in non-small cell lung cancer. Biomed Pharmacother, 2018, 106: 1600–6., DOI 10.1016/j.biopha.2018.07.113 Han X, Huang T, Han J. Long noncoding RNA VPS9D1-AS1 augments the malignant phenotype of non-small cell lung cancer by sponging microRNA-532-3p and thereby enhancing HMGA2 expression. Aging, 2020, 12(1):370–86., DOI 10. 18632/aging.102628 Xiao S, Xu N, Ding Q, Huang S, Zha Y, Zhu H. LncRNA VPS9D1-AS1 promotes cell proliferation in acute lymphoblastic leukemia through modulating GPX1 expression by miR-491-5p and miR-214-3p evasion. Biosci Rep, 2020, 40., DOI 10.1042/bsr20193461 Fa X, Song P, Fu Y, Deng Y, Liu K. Long non-coding RNA VPS9D1-AS1 facilitates cell proliferation, migration and stemness in hepatocellular carcinoma. Cancer Cel Int, 2021, 21(1):131., DOI 10.1186/s12935-020-01741-7 Masciullo V, Baldassarre G, Pentimalli F, Berlingieri MT, Boccia A, Chiappetta G, et al. HMGA1 protein over-expression is a frequent feature of epithelial ovarian carcinomas. Carcinogenesis, 2003, 24(7):1191–8., DOI 10.1093/ carcin/bgg075 Kim DK, Seo EJ, Choi EJ, Lee SI, Kwon YW, Jang IH, et al. Crucial role of HMGA1 in the self-renewal and drug resistance of ovarian cancer stem cells. Exp Mol Med, 2016, 48:e255., DOI 10.1038/emm.2016.73 Liu Y, Wang Y, Zhang Y, Fu J, Zhang G. Knockdown of HMGA1 expression by short/small hairpin RNA inhibits growth of ovarian carcinoma cells. Biotechnol Appl Biochem, 2012, 59(1):1–5., DOI 10.1002/bab.56 Braga EA, Fridman MV, Moscovtsev AA, Filippova EA, Dmitriev AA, Kushlinskii NE. LncRNAs in ovarian cancer progression, metastasis, and main pathways: ceRNA and alternative mechanisms. Int J Mol Sci, 2020, 21(22):8855., DOI 10.3390/ijms21228855 Pa M, Naizaer G, Seyiti A, Kuerbang G. Long noncoding RNA MALAT1 functions as a sponge of MiR-200c in ovarian cancer. Oncol Res, 2017)., DOI 10.3727/096504017X15049198963076 Zhang Y, Ai H, Fan X, Chen S,Wang Y, Liu L. Knockdown of long non-coding RNA HOTAIR reverses cisplatin resistance of ovarian cancer cells through inhibiting miR-138-5p-regulated EZH2 and SIRT1. Biol Res, 2020, 53(1):18., DOI 10.1186/s40659-020-00286-3 Cao Y, Shi H, Ren F, Jia Y, Zhang R. Long non-coding RNA CCAT1 promotes metastasis and poor prognosis in epithelial ovarian cancer. Exp Cel Res, 2017, 359(1):185–94., DOI 10.1016/j.yexcr.2017.07.030 Liu Y,Wang Y, Fu X, Lu Z. Long non-coding RNA NEAT1 promoted ovarian cancer cells’ metastasis through regulation of miR-382-3p/ROCK1 axial. Cancer Sci, 2018, 109(7):2188–98., DOI 10.1111/cas.13647 Shen Y, Lv M, Fang Y, Lu J, Wu Y. LncRNA MNX1-AS1 promotes ovarian cancer process via targeting the miR-744-5p/SOX12 axis. J Ovarian Res, 2021, 14(1):161., DOI 10.1186/s13048-021-00910-0 Wu DD, Chen X, Sun KX, Wang LL, Chen S, Zhao Y. Role of the lncRNA ABHD11-AS1 in the tumorigenesis and progression of epithelial ovarian cancer through targeted regulation of RhoC. Mol Cancer, 2017, 16(1):138., DOI 10.1186/ s12943-017-0709-5 Long X, Li L, Zhou Q,Wang H, Zou D,Wang D, et al. Long non-coding RNA LSINCT5 promotes ovarian cancer cell proliferation, migration and invasion by disrupting the CXCL12/CXCR4 signalling axis. Oncol Lett, 2018, 15(5):7200–6., DOI 10.3892/ol.2018.8241 Nagpal S, Ghosn C, DiSepio D, Molina Y, Sutter M, Klein ES, et al. Retinoid-dependent recruitment of a histone H1 displacement activity by retinoic acid receptor. J Biol Chem, 1999, 274(32):22563–8., DOI 10.1074/jbc. 274.32.22563 Cleynen I, Van de Ven WJM. The HMGA proteins: A myriad of functions, Review). Int J Oncol, 2008, 32(2):289–305., DOI 10.3892/ijo.32.2.289 Benecke AG, Eilebrecht S. RNA-mediated regulation of HMGA1 function. Biomolecules, 2015, 5(2):943–57., DOI 10.3390/biom5020943 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610870 EP 1610880 DI 10.3389/pore.2023.1610870 PG 11 ER PT J AU Jia, G Lei, P Zhang, Y Zheng, Z Fang, J Yang, X Wei, H Chen, T AF Jia, Guiru Lei, Purun Zhang, Yanru Zheng, Zongheng Fang, Jiafeng Yang, Xiaofeng Wei, Hongbo Chen, Tufeng TI New staging systems for left-sided colon cancer based on the number of retrieved and metastatic lymph nodes provide a more accurate prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE left-sided colon cancer; retrieved lymph nodes; log odds of positive lymph nodes; revised nodal stage; staging system ID left-sided colon cancer; retrieved lymph nodes; log odds of positive lymph nodes; revised nodal stage; staging system AB Objectives: We aimed to explore reasonable lymph node classification strategies for left-sided colon cancer (LCC) patients. Methods: 48,425 LCC patients from 2010 to 2015 were identified in the US Surveillance, Epidemiology, and End Results database. We proposed an innovative revised nodal (rN) staging of the 8th American Joint Committee on Cancer (AJCC) Tumor/Node/Metastasis (TNM) classification based on the cut-off value of retrieved lymph nodes and survival analyses in patients with LCC. Log odds of positive lymph nodes (LODDS) stage is a numerical classification strategy obtained by a formula that incorporates the numbers of retrieved and positive lymph nodes. To develop the TrN or TLODDS classification, patients with similar survival rates were grouped by combining T and rN or LODDS stage. The TrN or TLODDS classification was further evaluated in a validation set of 12,436 LCC patients from 2016 to 2017 in the same database and a Chinese application set of 958 LCC patients. Results: Wedeveloped novel TrNand TLODDS classifications for LCC patients that incorporated 7 stageswith reference to the AJCC staging system. In comparison to the 8th AJCC TNM and TrN classifications, TLODDS classification demonstrated significantly better discrimination (area under the receiver operating characteristic curve, 0.650 vs. 0.656 vs. 0.661, p < 0.001), bettermodel-fitting (Akaike information criteria, 309,287 vs. 308,767 vs. 308,467), and superior net benefits. The predictive performance of the TrN and TLODDS classifications was further verified in the validation and application sets. Conclusion: Both the TrN and TLODDS classifications have better discriminatory ability, model-fitting, and net benefits than the existing TNM classification, and represent an alternative to the current TNM classification for LCC patients. C1 [Jia, Guiru] Sun Yat-sen University, The Third Affiliated Hospital, Department of Gastrointestinal SurgeryGuangzhou, China. [Lei, Purun] Sun Yat-sen University, The Third Affiliated Hospital, Department of Gastrointestinal SurgeryGuangzhou, China. [Zhang, Yanru] Sun Yat-sen University, The Third Affiliated Hospital, Department of Gastrointestinal SurgeryGuangzhou, China. [Zheng, Zongheng] Sun Yat-sen University, The Third Affiliated Hospital, Department of Gastrointestinal SurgeryGuangzhou, China. [Fang, Jiafeng] Sun Yat-sen University, The Third Affiliated Hospital, Department of Gastrointestinal SurgeryGuangzhou, China. [Yang, Xiaofeng] Sun Yat-sen University, The Third Affiliated Hospital, Department of Gastrointestinal SurgeryGuangzhou, China. [Wei, Hongbo] Sun Yat-sen University, The Third Affiliated Hospital, Department of Gastrointestinal SurgeryGuangzhou, China. [Chen, Tufeng] Sun Yat-sen University, The Third Affiliated Hospital, Department of Gastrointestinal SurgeryGuangzhou, China. RP Chen, T (reprint author), Sun Yat-sen University, The Third Affiliated Hospital, Department of Gastrointestinal Surgery, Guangzhou, China. EM chentuf@mail.sysu.edu.cn CR Keum N, Giovannucci E. Global burden of colorectal cancer: Emerging trends, risk factors and prevention strategies. Nat Rev Gastroenterol Hepatol, 2019, 16: 713–32., DOI 10.1038/s41575-019-0189-8 Gervaz P, Bucher P, Morel P. Two colons-two cancers: Paradigm shift and clinical implications. J Surg Oncol, 2004, 88:261–6., DOI 10.1002/jso.20156 Wu W, Li D,Ma W, Zheng S, Han D, Xu F, et al. Examining more lymph nodes may improve the prognosis of patients with right colon cancer: Determining the optimal minimum lymph node count. Cancer Control, 2021, 28: 10732748211064034., DOI 10.1177/10732748211064034 Glebov OK, Rodriguez LM, Nakahara K, Jenkins J, Cliatt J, Humbyrd CJ, et al. Distinguishing right from left colon by the pattern of gene expression. Cancer Epidemiol Biomarkers Prev, 2003, 12:755–62. Baran B, Mert ON, Yerli TN, Acar E, Bekcioglu O, Baskin Y. Difference between left-sided and right-sided colorectal cancer: A focused review of literature. Gastroenterol Res, 2018, 11:264–73., DOI 10.14740/gr1062w Marzouk O, Schofield J. Review of histopathological and molecular prognostic features in colorectal cancer. Cancers, Basel,, 2011, 3:2767–810., DOI 10.3390/ cancers3022767 Gualco G, Reissenweber N, Cliche I, Bacchi CE. Flat elevated lesions of the colon and rectum: A spectrum of neoplastic and nonneoplastic entities. Ann Diagn Pathol, 2006, 10:333–8., DOI 10.1016/j.anndiagpath.2006.03.003 Nawa T, Kato J, Kawamoto H, Okada H, Yamamoto H, Kohno H, et al. Differences between right- and left-sided colon cancer in patient characteristics, cancer morphology and histology. J Gastroenterol Hepatol, 2008, 23:418–23., DOI 10. 1111/j.1440-1746.2007.04923.x Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, et al. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more "personalized" approach to cancer staging. CA Cancer J Clin, 2017, 67: 93–9., DOI 10.3322/caac.21388 Li DG, Di Carlo I, Scilletta R, Scilletta B, Puleo S. Colorectal cancer and lymph nodes: The obsession with the number 12. World J Gastroenterol, 2014, 20:1951–60., DOI 10.3748/wjg.v20.i8.1951 Le Voyer TE, Sigurdson ER, Hanlon AL, Mayer RJ, Macdonald JS, Catalano PJ, et al. Colon cancer survival is associated with increasing number of lymph nodes analyzed: A secondary survey of intergroup trial INT-0089. J Clin Oncol, 2003, 21: 2912–9., DOI 10.1200/JCO.2003.05.062 Xie D, Song X, Tong L. Stage migration resulting from inadequate number of examined lymph nodes impacts prognosis in stage II colon cancer after radical surgery. Int J Colorectal Dis, 2021, 36:959–69., DOI 10.1007/s00384-020-03794-6 Deng W, Xu T, Wang Y, Xu Y, Yang P, Gomez D, et al. Log odds of positive lymph nodes may predict survival benefit in patients with node-positive non-small cell lung cancer. Lung Cancer, 2018, 122:60–6., DOI 10.1016/j.lungcan.2018.05.016 Xu Z, Jing J, Ma G. Development and validation of prognostic nomogram based on log odds of positive lymph nodes for patients with gastric signet ring cell carcinoma. Chin J Cancer Res, 2020, 32:778–93., DOI 10.21147/j.issn.1000-9604. 2020.06.11 Pei JP, Zhao ZM, Sun Z, Gu WJ, Zhu J, Zhu J, et al. Development and validation of a novel classification scheme for combining pathological T stage and log odds of positive lymph nodes for colon cancer. Eur J Surg Oncol, 2022, 48: 228–36., DOI 10.1016/j.ejso.2021.09.005 Wang J, Hassett JM, Dayton MT, Kulaylat MN. The prognostic superiority of log odds of positive lymph nodes in stage III colon cancer. J Gastrointest Surg, 2008, 12:1790–6., DOI 10.1007/s11605-008-0651-3 Camp RL, Dolled-Filhart M, Rimm DL. X-Tile: A new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res, 2004, 10:7252–9., DOI 10.1158/1078-0432.CCR-04-0713 Akaike H. A new look at the statistical model identification. Ieee T Automat Contr, 1974, 6:716–23., DOI 10.1109/tac.1974.1100705 Fitzgerald M, Saville BR, Lewis RJ. Decision curve analysis. JAMA, 2015, 313: 409–10., DOI 10.1001/jama.2015.37 Vickers AJ, Elkin EB. Decision curve analysis: A novel method for evaluating prediction models. Med Decis Making, 2006, 26:565–74., DOI 10.1177/ 0272989X06295361 Greenberg R, Itah R, Ghinea R, Sacham-Shmueli E, Inbar R, Avital S. Metastatic lymph node ratio, LNR, as a prognostic variable in colorectal cancer patients undergoing laparoscopic resection. Tech Coloproctol, 2011, 15:273–9., DOI 10.1007/s10151-011-0701-9 Scarinci A, Di Cesare T, Cavaniglia D, Neri T, CollettiM, Cosenza G, et al. The impact of log odds of positive lymph nodes, LODDS, in colon and rectal cancer patient stratification: A single-center analysis of 323 patients. Updates Surg, 2018, 70:23–31., DOI 10.1007/s13304-018-0519-3 Persiani R, Cananzi FC, Biondi A, Paliani G, Tufo A, Ferrara F, et al. Log odds of positive lymph nodes in colon cancer: A meaningful ratio-based lymph node classification system. World J Surg, 2012, 36:667–74., DOI 10.1007/s00268-011- 1415-x Zhang C, Mei Z, Pei J, Abe M, Zeng X, Huang Q, et al. A modified tumornode- metastasis classification for primary operable colorectal cancer. JNCI Cancer Spectr, 2021, 5:pkaa093., DOI 10.1093/jncics/pkaa093 Imamura Y, Lochhead P, Yamauchi M, Kuchiba A, Qian ZR, Liao X, et al. Analyses of clinicopathological, molecular, and prognostic associations of KRAS codon 61 and codon 146 mutations in colorectal cancer: Cohort study and literature review. Mol Cancer, 2014, 13:135., DOI 10.1186/1476-4598-13-135 Kosumi K, Hamada T, Zhang S, Liu L, da Silva A, Koh H, et al. Prognostic association of PTGS2, COX-2, over-expression according to BRAF mutation status in colorectal cancer: Results from two prospective cohorts and CALGB 89803, Alliance, trial. Eur J Cancer, 2019, 111:82–93., DOI 10.1016/j.ejca.2019.01.022 Park HS, Lloyd S, Decker RH, Wilson LD, Yu JB. Limitations and biases of the surveillance, Epidemiology, and End results database. Curr Probl Cancer, 2012, 36: 216–24., DOI 10.1016/j.currproblcancer.2012.03.011 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610874 EP 1610885 DI 10.3389/pore.2023.1610874 PG 12 ER PT J AU Zhao, Sh Zhang, D Liu, S Huang, J AF Zhao, Shimin Zhang, Dongdong Liu, Sicheng Huang, Jun TI The roles of NOP56 in cancer and SCA36 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE methylation; NOP56; malignant tumor; box C/D RNP; SCA36 ID methylation; NOP56; malignant tumor; box C/D RNP; SCA36 AB NOP56 is a highly conserved nucleolar protein. Amplification of the intron GGCCTG hexanucleotide repeat sequence of the NOP56 gene results in spinal cerebellar ataxia type 36 (SCA36). NOP56 contains an N-terminal domain, a coiled-coil domain, and a C-terminal domain. Nucleolar protein NOP56 is significantly abnormally expressed in a number of malignant tumors, and its mechanism is different in different tumors, but its regulatory mechanism in most tumors has not been fully explored. NOP56 promotes tumorigenesis in some cancers and inhibits tumorigenesis in others. In addition, NOP56 is associated with methylation in some tumors, suggesting that NOP56 has the potential to become a tumor-specific marker. This review focuses on the structure, function, related signaling pathways, and role of NOP56 in the progression of various malignancies, and discusses the progression of NOP56 in neurodegenerative and other diseases. C1 [Zhao, Shimin] Second Affiliated Hospital of Nanchang University, Department of General SurgeryNanchang, China. [Zhang, Dongdong] Second Affiliated Hospital of Nanchang University, Department of General SurgeryNanchang, China. [Liu, Sicheng] Second Affiliated Hospital of Nanchang University, Department of General SurgeryNanchang, China. [Huang, Jun] Second Affiliated Hospital of Nanchang University, Department of General SurgeryNanchang, China. RP Huang, J (reprint author), Second Affiliated Hospital of Nanchang University, Department of General Surgery, Nanchang, China. EM Huangjun0727@163.com CR Gautier T, Berges T, Tollervey D, Hurt E. Nucleolar KKE/D repeat proteins Nop56p and Nop58p interact with Nop1p and are required for ribosome biogenesis. Mol Cell Biol, 1997, 17:7088–98., DOI 10.1128/mcb.17.12.7088 GeneCards NOP56. GeneCards NOP56, 2022). Available at: https://www. genecards.org/Search/Keyword?queryString=NOP56, Accessed October 10, 2022). Omer AD, Lowe TM, Russell AG, Ebhardt H, Eddy SR, Dennis PP. Homologs of small nucleolar RNAs in Archaea. Science, 2000, 288:517–22., DOI 10.1126/ science.288.5465.517 Wajahat M, Bracken CP, Orang A. Emerging functions for snoRNAs and snoRNA-derived fragments. Int J Mol Sci, 2021, 22:10193., DOI 10.3390/ ijms221910193 Newman DR, Kuhn JF, Shanab GM, Maxwell ES. Box C/D snoRNA-associated proteins: Two pairs of evolutionarily ancient proteins and possible links to replication and transcription. RNA, 2000, 6:861–79., DOI 10.1017/ s1355838200992446 Yi Y, Li Y, Meng Q, Li Q, Li F, Lu B, et al. A PRC2-independent function for EZH2 in regulating rRNA 2′-O methylation and IRES-dependent translation. Nat Cell Biol, 2021, 23:341–54., DOI 10.1038/s41556-021-00653-6 Marcel V, Catez F, Berger CM, Perrial E, Plesa A, Thomas X, et al. Expression profiling of ribosome biogenesis factors reveals nucleolin as a novel potential marker to predict outcome in AML patients. PLoS One, 2017, 12:e0170160., DOI 10.1371/journal.pone.0170160 Hong H, Luo B, Qin Y, Li S, Peng Z. RNA-seq and integrated network analysis reveals the hub genes and key pathway of paclitaxel inhibition on Adriamycin resistant diffuse large B cell lymphoma cells. Bioengineered, 2022, 13:7607–21., DOI 10.1080/21655979.2022.2048772 Cowling VH, Turner SA, Cole MD. Burkitt’s lymphoma-associated c-Myc mutations converge on a dramatically altered target gene response and implicate Nol5a/Nop56 in oncogenesis. Oncogene, 2014, 33:3519–27., DOI 10.1038/onc. 2013.338 Yang Z, Liang S-Q, Zhao L, Yang H, Marti TM, Hegedus B, et al. Metabolic synthetic lethality by targeting NOP56 and mTOR in KRAS-mutant lung cancer. J Exp Clin Cancer Res, 2022, 41:25., DOI 10.1186/s13046-022-02240-5 Kobayashi H, Abe K, Matsuura T, Ikeda Y, Hitomi T, Akechi Y, et al. Expansion of intronic GGCCTG hexanucleotide repeat in NOP56 causes SCA36, a type of spinocerebellar ataxia accompanied by motor neuron involvement. Am J Hum Genet, 2011, 89:121–30., DOI 10.1016/j.ajhg.2011.05.015 Schols L, Bauer P, Schmidt T, Schulte T, Riess O. Autosomal dominant cerebellar ataxias: Clinical features, genetics, and pathogenesis. Lancet Neurol, 2004, 3:291–304., DOI 10.1016/S1474-4422(04)00737-9 Ikeda Y, Ohta Y, Kobayashi H, Okamoto M, Takamatsu K, Ota T, et al. Clinical features of SCA36: A novel spinocerebellar ataxia with motor neuron involvement, asidan). Neurology, 2012, 79:333–41., DOI 10.1212/WNL. 0b013e318260436f Garcia-Murias M, Quintans B, Arias M, Seixas AI, Cacheiro P, Tarrio R, et al. Costa da morte” ataxia is spinocerebellar ataxia 36: Clinical and genetic characterization. Brain, 2012, 135:1423–35., DOI 10.1093/brain/aws069 Arias M, Garcia-Murias M, Sobrido MJ. Spinocerebellar ataxia 36, SCA36): «Costa da morte ataxia. Neurologia, 2017, 32:386–93., DOI 10. 1016/j.nrl.2014.11.005 NOP56. National Center for Biotechnology information, 2022). Available at: https://www.ncbi.nlm.nih.gov/gene/10528, Accessed September 11, 2022). Lykke-Andersen S, Ardal BK, Hollensen AK, Damgaard CK, Jensen TH. Box C/D snoRNP Autoregulation by a cis-Acting snoRNA in the NOP56 Pre-mRNA. Mol Cell, 2018, 72:99–111. e5., DOI 10.1016/j.molcel.2018.08.017 Bizarro J, Charron C, Boulon S, Westman B, Pradet-Balade B, Vandermoere F, et al. Proteomic and 3D structure analyses highlight the C/D box snoRNP assembly mechanism and its control. J Cell Biol, 2014, 207:463–80., DOI 10.1083/jcb. 201404160 Westman BJ, Verheggen C, Hutten S, Lam YW, Bertrand E, Lamond AI. A proteomic screen for nucleolar SUMO targets shows SUMOylation modulates the function of Nop5/Nop58. Mol Cell, 2010, 39:618–31., DOI 10.1016/j.molcel.2010. 07.025 Ryu H, Sun X, Chen Y, Li Y, Wang X, Dai RS, et al. The deubiquitinase USP36 promotes snoRNP group SUMOylation and is essential for ribosome biogenesis. EMBO Rep, 2021, 22:e50684., DOI 10.15252/embr.202050684 Schlosser I, Holzel M, Murnseer M, Burtscher H, Weidle UH, Eick D. A role for c-Myc in the regulation of ribosomal RNA processing. Nucleic Acids Res, 2003, 31:6148–56., DOI 10.1093/nar/gkg794 Ruggero D. The role of myc-induced protein synthesis in cancer. Cancer Res, 2009, 69:8839–43., DOI 10.1158/0008-5472.CAN-09-1970 Cheng X, Roberts RJ. AdoMet-dependent methylation, DNA methyltransferases and base flipping. Nucleic Acids Res, 2001, 29:3784–95., DOI 10.1093/nar/29.18.3784 Martin JL, McMillan FM. SAM, dependent, I AM: The S-adenosylmethionine-dependent methyltransferase fold. Curr Opin Struct Biol, 2002, 12:783–93., DOI 10.1016/s0959-440x(02)00391-3 Feric M, Vaidya N, Harmon TS, Mitrea DM, Zhu L, Richardson TM, et al. Coexisting liquid phases underlie nucleolar subcompartments. Cell, 2016, 165: 1686–97., DOI 10.1016/j.cell.2016.04.047 Yu G, Zhao Y, Li H. The multistructural forms of box C/D ribonucleoprotein particles. RNA, 2018, 24:1625–33., DOI 10.1261/rna.068312.118 Yang Z, Wang J, Huang L, Lilley DMJ, Ye K. Functional organization of box C/D RNA-guided RNA methyltransferase. Nucleic Acids Res, 2020, 48:5094–105., DOI 10.1093/nar/gkaa247 Hofler S, Lukat P, Blankenfeldt W, Carlomagno T. High-resolution structure of eukaryotic Fibrillarin interacting with Nop56 amino-terminal domain. RNA, 2021, 27:496–512., DOI 10.1261/rna.077396.120 Pradet-Balade B, Girard C, Boulon S, Paul C, Azzag K, Bordonne R, et al. CRM1 controls the composition of nucleoplasmic pre-snoRNA complexes to licence them for nucleolar transport: CRM1 controls transport and assembly of snoRNPs. EMBO J, 2011, 30:2205–18., DOI 10.1038/emboj.2011.128 Shi X, Huang L, Lilley DMJ, Harbury PB, Herschlag D. The solution structural ensembles of RNA kink-turn motifs and their protein complexes. Nat Chem Biol, 2016, 12:146–52., DOI 10.1038/nchembio.1997 Omer AD, Ziesche S, Ebhardt H, Dennis PP. In vitro reconstitution and activity of a C/D box methylation guide ribonucleoprotein complex. Proc Natl Acad Sci U S A, 2002, 99:5289–94., DOI 10.1073/pnas.082101999 Tran E, Zhang X, Lackey L, Maxwell ES. Conserved spacing between the box C/D and C′/D′ RNPs of the archaeal box C/D sRNP complex is required for efficient 2′-O-methylation of target RNAs. RNA, 2005, 11:285–93., DOI 10.1261/rna.7223405 Ye K, Jia R, Lin J, JuM, Peng J, Xu A, et al. Structural organization of box C/D RNA-guided RNA methyltransferase. Proc Natl Acad Sci USA, 2009, 106:13808–13., DOI 10.1073/pnas.0905128106 Gagnon KT, Biswas S, Zhang X, Brown BA, Wollenzien P, Mattos C, et al. Structurally conserved Nop56/58 N-terminal domain facilitates archaeal box C/D ribonucleoprotein-guided methyltransferase activity. J Biol Chem, 2012, 287: 19418–28., DOI 10.1074/jbc.M111.323253 Lin J, Lai S, Jia R, Xu A, Zhang L, Lu J, et al. Structural basis for site-specific ribose methylation by box C/D RNA protein complexes. Nature, 2011, 469:559–63., DOI 10.1038/nature09688 Tran EJ, Zhang X, Maxwell ES. Efficient RNA 2’-O-methylation requires juxtaposed and symmetrically assembled archaeal box C/D and C’/D’ RNPs. EMBO J, 2003, 22:3930–40., DOI 10.1093/emboj/cdg368 Gagnon KT, Zhang X, Agris PF, Maxwell ES. Assembly of the archaeal box C/D sRNP can occur via alternative pathways and requires temperature-facilitated sRNA remodeling. J Mol Biol, 2006, 362:1025–42., DOI 10.1016/j.jmb.2006.07.091 Massenet S, Bertrand E, Verheggen C. Assembly and trafficking of box C/D and H/ACA snoRNPs. RNA Biol, 2017, 14:680–92., DOI 10.1080/15476286.2016.1243646 Jorjani H, Kehr S, Jedlinski DJ, Gumienny R, Hertel J, Stadler PF, et al. An updated human snoRNAome. Nucleic Acids Res, 2016, 44:5068–82., DOI 10.1093/ nar/gkw386 Girard C, Verheggen C, Neel H, Cammas A, Vagner S, Soret J, et al. Characterization of a short isoform of human Tgs1 hypermethylase associating with small nucleolar ribonucleoprotein core proteins and produced by limited proteolytic processing. J Biol Chem, 2008, 283:2060–9., DOI 10.1074/jbc.M704209200 Ono K, Han J. The p38 signal transduction pathway: Activation and function. Cell Signal, 2000, 12:1–13., DOI 10.1016/s0898-6568(99)00071-6 Verma IM, Stevenson JK, Schwarz EM, Van Antwerp D, Miyamoto S. Rel/NFkappa B/I kappa B family: Intimate tales of association and dissociation. Genes Dev, 1995, 9:2723–35., DOI 10.1101/gad.9.22.2723 Ren X, Zhang H, Yan X, Sun Y, Xu T. NOP56 negatively regulates MyD88- mediated NF-κB signaling in miiuy croaker, Miichthys miiuy. Fish Shellfish Immunol, 2022, 120:75–81., DOI 10.1016/j.fsi.2021.11.011 Hu X, li J, Fu M, Zhao X, Wang W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct Target Ther, 2021, 6:402., DOI 10.1038/s41392-021- 00791-1 Wang H, Chen X, He T, Zhou Y, Luo H. Evidence for tissue-specific JAK/ STAT target genes in Drosophila optic lobe development. Genetics, 2013, 195: 1291–306., DOI 10.1534/genetics.113.155945 Judd J, Abdel Karim N, Khan H, Naqash AR, Baca Y, Xiu J, et al. Characterization of KRAS mutation subtypes in non–small cell lung cancer. Mol Cancer Ther, 2021, 20:2577–84., DOI 10.1158/1535-7163.MCT-21-0201 Zhang Z, Wang Z, Jin L, Tan X, Shen L,Wei G, et al. Effect of piRNA NU13 in regulating biological behaviors of human Wilms tumor cells in vitro. Nan Fang Yi Ke Da Xue Xue Bao, 2021, 41:184–92., DOI 10.12122/j.issn.1673-4254.2021.02.04 Zhang Z, Wang Z, Tan X, Jin L, Wang Z, Shen L, et al. PiRNA MW557525 promotes the vitality and pluripotency of piwil2-iCSCs by regulating NOP56., 2021)., DOI 10.21203/rs.3.rs-439016/v1 Su H, Xu T, Ganapathy S, Shadfan M, Long M, Huang TH-M, et al. Elevated snoRNA biogenesis is essential in breast cancer. Oncogene, 2014, 33:1348–58., DOI 10.1038/onc.2013.89 Vellky JE, Ricke EA, Huang W, Ricke WA. Expression, localization, and function of the nucleolar protein BOP1 in prostate cancer progression. Am J Pathol, 2021, 191:168–79., DOI 10.1016/j.ajpath.2020.09.010 Ussowicz M, Marcel V, Nguyen Van Long F, Kazanowska B, Diaz J-J, Wolowiec D. Analysis of the rRNA methylation complex components in pediatric B-cell precursor acute lymphoblastic leukemia: A pilot study. Adv Clin Exp Med, 2020, 29:107–13., DOI 10.17219/acem/112608 Singh P, Chalertpet K, Sukbhattee J, Wongmanee N, Suwannakart P, Yanatatsaneejit P. Association between promoter methylation and gene expression of CGB3 and NOP56 in HPV-infected cervical cancer cells. Biomed Rep, 2022, 16:1., DOI 10.3892/br.2021.1484 Ng JM-K, Yu J. Promoter hypermethylation of tumour suppressor genes as potential biomarkers in colorectal cancer. Int J Mol Sci, 2015, 16:2472–96., DOI 10. 3390/ijms16022472 Jung G, Hernandez-Illan E, Moreira L, Balaguer F, Goel A. Epigenetics of colorectal cancer: Biomarker and therapeutic potential. Nat Rev Gastroenterol Hepatol, 2020, 17:111–30., DOI 10.1038/s41575-019-0230-y Soozangar N, Sadeghi MR, Jeddi F, Somi MH, Shirmohamadi M, Samadi N. Comparison of genome-wide analysis techniques to DNA methylation analysis in human cancer. J Cell Physiol, 2018, 233:3968–81., DOI 10.1002/jcp.26176 Ciribilli Y, Singh P, Inga A, Borlak J. c-Myc targeted regulators of cell metabolism in a transgenic mouse model of papillary lung adenocarcinoma. Oncotarget, 2016, 7:65514–39., DOI 10.18632/oncotarget.11804 Liu P, Dong Y, Gu J, Puthiyakunnon S, Wu Y, Chen X-G. Developmental piRNA profiles of the invasive vector mosquito Aedes albopictus. Parasit Vectors, 2016, 9:524., DOI 10.1186/s13071-016-1815-8 Liu Y, Dou M, Song X, Dong Y, Liu S, Liu H, et al. The emerging role of the piRNA/piwi complex in cancer. Mol Cancer, 2019, 18:123., DOI 10.1186/s12943- 019-1052-9 Pan Y, Liu G, Zhou F, Su B, Li Y. DNA methylation profiles in cancer diagnosis and therapeutics. Clin Exp Med, 2018, 18:1–14., DOI 10.1007/s10238-017- 0467-0 Zhu H, Zhu H, Tian M, Wang D, He J, Xu T. DNA methylation and hydroxymethylation in cervical cancer: Diagnosis, prognosis and treatment. Front Genet, 2020, 11:347., DOI 10.3389/fgene.2020.00347 Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2017. CA Cancer J Clin, 2021, 71:7–30., DOI 10.3322/caac.21387 Rosendo-Chalma P, Antonio-Vejar V, Bigoni-Ordonez GD, Patino-Morales CC, Cano-Garcia A, Garcia-Carranca A. CDH1 and SNAI1 are regulated by E7 from human papillomavirus types 16 and 18. Int J Oncol, 2020, 57:301–13., DOI 10.3892/ijo.2020.5039 Chalertpet K, Pakdeechaidan W, Patel V, Mutirangura A, Yanatatsaneejit P. Human papillomavirus type 16 E7 oncoprotein mediates CCNA1 promoter methylation. Cancer Sci, 2015, 106:1333–40., DOI 10.1111/cas.12761 Cao J, Dong J,Wang Y, Chen Y. The expressions of DNA methyltransferase 1, DNMT1, and cyclin A1, CCNA1, in cervical carcinogenesis. Int J Clin Exp Pathol, 2019, 12:40–9. Herbst J, Pantel K, Effenberger K, Wikman H. Clinical applications and utility of cell-free DNA-based liquid biopsy analyses in cervical cancer and its precursor lesions. Br J Cancer, 2022, 127:1403–10., DOI 10.1038/s41416-022-01868-6 Zhang Y, Lin Z, Lin X, Zhang X, Zhao Q, Sun Y. A gene module identification algorithm and its applications to identify gene modules and key genes of hepatocellular carcinoma. Sci Rep, 2021, 11:5517., DOI 10.1038/s41598-021- 84837-y Zhou W, Chen Y, Luo R, Li Z, Jiang G, Ou X. Identification of biomarkers related to immune cell infiltration in hepatocellular carcinoma using gene Coexpression network. Pathol Oncol Res, 2021, 27:601693., DOI 10.3389/pore.2021. 601693 Li A, Ye B, Lin F, Zhou P, Wang Y, Miao X, et al. Dissecting novel tumor antigens and immune subtypes guided mRNA vaccine development for HBV related-hepatocellular carcinoma. [preprint]. In Review, 2022)., DOI 10.21203/rs.3. rs-1962830/v1 Coccaro N, Anelli L, Zagaria A, Specchia G, Albano F. Next-generation sequencing in acute lymphoblastic leukemia. Int J Mol Sci, 2019, 20:2929., DOI 10. 3390/ijms20122929 Button L, Rogers B, Thomas E, Bradfield A, Alnafakh R, Drury J, et al. Telomere and telomerase-associated proteins in endometrial carcinogenesis and cancer-associated survival. Int J Mol Sci, 2022, 23:626., DOI 10.3390/ ijms23020626 Giridhar KV, Sosa CP, Hillman DW, Sanhueza C, Dalpiaz CL, Costello BA, et al. Whole blood mRNA expression-based prognosis of metastatic renal cell carcinoma. Int J Mol Sci, 2017, 18:2326., DOI 10.3390/ijms18112326 Bhushan A, Kumari R, Srivastava T. Scouting for common genes in the heterogenous hypoxic tumor microenvironment and their validation in glioblastoma. 3 Biotech, 2021, 11:451., DOI 10.1007/s13205-021-02987-2 Expansion of intronic. Expansion of intronic GGCCTG hexanucleotide repeat in NOP56 causes SCA36, a type of spinocerebellar ataxia accompanied by motor neuron involvement - PubMed, 2022). Available at: https://pubmed.ncbi.nlm.nih. gov/21683323/(Accessed August 18, 2022). Zeng S, Zeng J, He M, Zeng X, Zhou Y, Liu Z, et al. Genetic and clinical analysis of spinocerebellar ataxia type 36 in Mainland China. Clin Genet, 2016, 90: 141–8., DOI 10.1111/cge.12706 Kozol RA, Abrams AJ, James DM, Buglo E, Yan Q, Dallman JE. Function over form: Modeling groups of inherited neurological conditions in zebrafish. Front Mol Neurosci, 2016, 9:55., DOI 10.3389/fnmol.2016.00055 Quelle-Regaldie A, Folgueira M, Yanez J, Sobrido-Camean D, Alba-Gonzalez A, Barreiro-Iglesias A, et al. A nop56 zebrafish loss-of-function model exhibits a severe neurodegenerative phenotype. Biomedicines, 2022, 10:1814., DOI 10.3390/ biomedicines10081814 Vijayakumar S, Jones S, Jones T, Tian C, Johnson K. Spontaneous mutations of the Zpld1 gene in mice cause semicircular canal dysfunction but do not impair gravity receptor or hearing functions. Scientific Rep, 2019, 9:12430., DOI 10.1038/ s41598-019-48835-5 Miyazaki K, Yamashita T, Morimoto N, Sato K, Mimoto T, Kurata T, et al. Early and selective reduction of NOP56, Asidan, and RNA processing proteins in the motor neuron of ALS model mice. Neurol Res, 2013, 35:744–54., DOI 10.1179/ 1743132813Y.0000000196 Xu Z, Ruan Z, Huang X, Liu Q, Li Z, Zhou X, et al. Identification of aberrantly methylated differentially expressed genes in age-related macular degeneration. Medicine, 2019, 98:e15083., DOI 10.1097/MD.0000000000015083 Li W, Zhang S, Zhao Y,Wang D, Shi Q, Ding Z, et al. Revealing the key MSCs niches and pathogenic genes in influencing cep homeostasis: A conjoint analysis of single-cell and WGCNA. Front Immunol, 2022, 13:933721., DOI 10.3389/fimmu. 2022.933721 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610884 EP 1610892 DI 10.3389/pore.2023.1610884 PG 9 ER PT J AU Deng, H Li, T Wei, F Han, W Xu, X Zhang, Y AF Deng, Hongyang Li, Tengfei Wei, Fengxian Han, Wei Xu, Xiaodong Zhang, Youcheng TI High expression of TMEM200A is associated with a poor prognosis and immune infiltration in gastric cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bioinformatics; gastric cancer; prognostic; immune infiltration; TMEM200A ID bioinformatics; gastric cancer; prognostic; immune infiltration; TMEM200A AB Background: Gastric cancer (GC) is one of the global malignant tumors with high incidence and poor prognosis. Exploring new GC molecular markers is important to improve GC prognosis. Transmembrane protein 200A (TMEM200A) is a member of the family of transmembrane proteins (TMEM). This study is the first to investigate the potential function of TMEM200A and its relationship with immune infiltration in GC. Methods: The differential expression of TMEM200A was determined through the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The receiver operating characteristic (ROC) curve was drawn to assess the diagnostic value of TMEM200A for GC. The relationship between TMEM200A and the clinical characteristics of patients with GC was investigated using the Wilcoxon test and the Kruskal-Wallis test. The effect of TMEM200A on overall survival (OS) was identified using the Kaplan-Meier method, the Logrank test, the univariate/multivariate Cox regression analysis, and the nomogram prediction model. The co-expressed genes and gene set enrichment analysis (GSEA) were used to explore the potential biological functions of TMEM200A. We used the Tumor Immune Estimation Resource (TIMER) database and the ssGSEA algorithm to estimate the relationship between TMEM200A and immune cell infiltration. Furthermore, we investigated the correlation of TMEM200A with immune checkpoint/immune cell surface markers using the TCGA-STAD data set. Finally, we identified prognosis-related methylation sites in TMEM200A using MethSurv. Results: TMEM200A was highly expressed in GC tissues. TMEM200A had a good diagnostic value for GC. High expression of TMEM200A may shorten the OS of GC patients and may be an independent risk factor for OS in GC patients. TMEM200A participates in the construction of a predictive model with a good predictive effect on the survival rate of GC patients at 1, 3, and 5 years. Coexpressed genes and GSEA indicated that TMEM200A may be an adhesion molecule closely associated with tumor invasion and metastasis. In addition, TMEM200A may be significantly associated with immune cell infiltration and immune checkpoint expression. We also found that TMEM200A contains three methylation sites associated with a poor prognosis. Conclusion: Upregulated TMEM200A may be a promising prognostic marker for GC and is closely associated with the tumor microenvironment (TME). C1 [Deng, Hongyang] The Second Hospital of Lanzhou University, The Department of General SurgeryLanzhou, China. [Li, Tengfei] The Second Hospital of Lanzhou University, The Department of General SurgeryLanzhou, China. [Wei, Fengxian] The Second Hospital of Lanzhou University, The Department of General SurgeryLanzhou, China. [Han, Wei] The Second Hospital of Lanzhou University, The Department of General SurgeryLanzhou, China. [Xu, Xiaodong] The Second Hospital of Lanzhou University, The Department of General SurgeryLanzhou, China. [Zhang, Youcheng] The Second Hospital of Lanzhou University, The Department of General SurgeryLanzhou, China. RP Zhang, Y (reprint author), The Second Hospital of Lanzhou University, The Department of General Surgery, Lanzhou, China. EM zhangyouchengphd@163.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/ caac.21492 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Smyth EC, NilssonM, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet, 2020, 396(10251):635–48., DOI 10.1016/S0140-6736(20)31288-5 Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer, ToGA): A phase 3, open-label, randomised controlled trial. Lancet, 2010, 376(9742):687–97., DOI 10.1016/S0140-6736(10)61121-X Lordick F, Carneiro F, Cascinu S, Fleitas T, Haustermans K, Piessen G, et al. Gastric cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol, 2022, 33(10):1005–20., DOI 10.1016/j.annonc.2022.07.004 Zhao Y, Zhang K, PanH,Wang Y, Zhou X, Xiang Y, et al. Genetic analysis of six transmembrane protein family genes in Parkinson’s disease in a large Chinese cohort. Front Aging Neurosci, 2022, 14:889057., DOI 10.3389/fnagi.2022.889057 Marx S, Dal Maso T, Chen JW, Bury M, Wouters J, Michiels C, et al. Transmembrane, TMEM, protein family members: Poorly characterized even if essential for the metastatic process. Semin Cancer Biol, 2020, 60:96–106., DOI 10. 1016/j.semcancer.2019.08.018 Zhuang J, Huang Y, Zheng W, Yang S, Zhu G, Wang J, et al. TMEM100 expression suppresses metastasis and enhances sensitivity to chemotherapy in gastric cancer. Biol Chem, 2020, 401(2):285–96., DOI 10.1515/ hsz-2019-0161 Schmit K, Michiels C. TMEM proteins in cancer: A review. Front Pharmacol, 2018, 9:1345., DOI 10.3389/fphar.2018.01345 Zhang S, Dai H, Li W, Wang R, Wu H, Shen M, et al. TMEM116 is required for lung cancer cell motility and metastasis through PDK1 signaling pathway. Cell Death Dis, 2021, 12(12):1086., DOI 10.1038/s41419-021-04369-1 Koteluk O, Bielicka A, Lemanska Z, Jozwiak K, Klawiter W, Mackiewicz A, et al. The landscape of transmembrane protein family members in head and neck cancers: Their biological role and diagnostic utility. Cancers, Basel,, 2021, 13(19): 4737., DOI 10.3390/cancers13194737 Lundback V, Kulyte A, Arner P, Strawbridge RJ, Dahlman I. Genome-Wide association study of diabetogenic adipose morphology in the GENetics of adipocyte lipolysis, GENiAL, cohort. [J] Cell, 2020, 9(5):1085., DOI 10.3390/cells9051085 Tan M, Schaffalitzky De Muckadell OB, Joergensen MT. Gene expression network analysis of precursor lesions in familial pancreatic cancer. J Pancreat Cancer, 2020, 6(1):73–84., DOI 10.1089/pancan.2020.0007 Vieira AR, de Carvalho FM, Johnson L, DeVos L, Swailes AL,Weber ML, et al. Fine mapping of 6q23.1 identifies TULP4 as contributing to clefts. Cleft Palate Craniofac J, 2015, 52(2):128–34., DOI 10.1597/13-023 Nie L, Zhang Y, You Y, Lin C, Li Q, Deng W, et al. The signature based on seven genomic instability-related genes could predict the prognosis of acutemyeloid leukemia patients. Hematology, 2022, 27(1):840–8., DOI 10.1080/16078454.2022. 2107970 Zhang X, Zheng P, Li Z, Gao S, Liu G. The somatic mutation landscape and RNA prognostic markers in stomach adenocarcinoma. Onco Targets Ther, 2020, 13:7735–46., DOI 10.2147/OTT.S263733 Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. Timer: A web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res, 2017, 77(21):e108–10., DOI 10.1158/0008-5472.CAN-17-0307 Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. Gepia: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res, 2017, 45(W1):W98–W102., DOI 10.1093/nar/gkx247 Goldman MJ, Craft B, Hastie M, Repecka K, McDade F, Kamath A, et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol, 2020, 38(6):675–8., DOI 10.1038/s41587-020-0546-8 Oh SC, Sohn BH, Cheong JH, Kim SB, Lee JE, Park KC, et al. Clinical and genomic landscape of gastric cancer with a mesenchymal phenotype. Nat Commun, 2018, 9(1):1777., DOI 10.1038/s41467-018-04179-8 Ooi CH, Ivanova T, Wu J, Lee M, Tan IB, Tao J, et al. Oncogenic pathway combinations predict clinical prognosis in gastric cancer. Plos Genet, 2009, 5(10): e1000676., DOI 10.1371/journal.pgen.1000676 Vasaikar SV, Straub P, Wang J, Zhang B. LinkedOmics: Analyzing multiomics data within and across 32 cancer types. Nucleic Acids Res, 2018, 46(D1): D956–d963., DOI 10.1093/nar/gkx1090 Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics, 2012, 16(5):284–7., DOI 10.1089/omi. 2011.0118 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A, 2005, 102(43): 15545–50., DOI 10.1073/pnas.0506580102 Hanzelmann S, Castelo R, Guinney J. Gsva: Gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics, 2013, 14:7., DOI 10.1186/1471- 2105-14-7 Modhukur V, Iljasenko T, Metsalu T, Lokk K, Laisk-Podar T, Vilo J. MethSurv: A web tool to perform multivariable survival analysis using DNA methylation data. Epigenomics, 2018, 10(3):277–88., DOI 10.2217/epi-2017-0118 Shen K, Yu W, Yu Y, Liu X, Cui X. Knockdown of TMEM45B inhibits cell proliferation and invasion in gastric cancer. Biomed Pharmacother, 2018, 104: 576–81., DOI 10.1016/j.biopha.2018.05.016 Duan J, Qian Y, Fu X, Chen M, Liu K, Liu H, et al. TMEM106C contributes to the malignant characteristics and poor prognosis of hepatocellular carcinoma. Aging, Albany NY,, 2021, 13(4):5585–606., DOI 10.18632/aging.202487 Shiraishi T, Ikeda K, Tsukada Y, Nishizawa Y, Sasaki T, Ito M, et al. High expression of TMEM180, a novel tumour marker, is associated with poor survival in stage III colorectal cancer. BMC Cancer, 2021, 21(1):302., DOI 10.1186/s12885-021- 08046-6 Chen ZF, Wang J, Yu Y, Wei W. MicroRNA-936 promotes proliferation and invasion of gastric cancer cells by down-regulating FGF2 expression and activating P13K/Akt signaling pathway. Eur Rev Med Pharmacol Sci, 2020, 24(12):6707–15., DOI 10.26355/eurrev_202006_21658 Shah S, Brock EJ, Ji K, Mattingly RR. Ras and Rap1: A tale of two GTPases. Semin Cancer Biol, 2019, 54:29–39., DOI 10.1016/j.semcancer.2018.03.005 Feng S, Agoulnik IU, Li Z, Han HD, Lopez-Berestein G, Sood A, et al. Relaxin/ RXFP1 signaling in prostate cancer progression. Ann N Y Acad Sci, 2009, 1160: 379–80., DOI 10.1111/j.1749-6632.2008.03793.x Ng HH, Shen M, Samuel CS, Schlossmann J, Bennett RG. Relaxin and extracellular matrix remodeling: Mechanisms and signaling pathways. Mol Cel Endocrinol, 2019, 487:59–65., DOI 10.1016/j.mce.2019.01.015 Peng D, Fu M,Wang M,Wei Y, Wei X. Targeting TGF-β signal transduction for fibrosis and cancer therapy. Mol Cancer, 2022, 21(1):104., DOI 10.1186/s12943- 022-01569-x Koh V, Chakrabarti J, Torvund M, Steele N, Hawkins JA, Ito Y, et al. Hedgehog transcriptional effector GLI mediates mTOR-Induced PD-L1 expression in gastric cancer organoids. Cancer Lett, 2021, 518:59–71., DOI 10.1016/j.canlet. 2021.06.007 Mishra YG, Manavathi B. Focal adhesion dynamics in cellular function and disease. Cell Signal, 2021, 85:110046., DOI 10.1016/j.cellsig.2021.110046 Seetharaman S, Etienne-Manneville S. Cytoskeletal crosstalk in cell migration. Trends Cel Biol, 2020, 30(9):720–35., DOI 10.1016/j.tcb.2020.06.004 Gilkes DM, Semenza GL, Wirtz D. Hypoxia and the extracellular matrix: Drivers of tumour metastasis. Nat Rev Cancer, 2014, 14(6):430–9., DOI 10.1038/ nrc3726 Lv B,Wang Y,Ma D, Cheng W, Liu J, Yong T, et al. Immunotherapy: Reshape the tumor immune microenvironment. Front Immunol, 2022, 13:844142., DOI 10. 3389/fimmu.2022.844142 Fu T, Dai LJ, Wu SY, Xiao Y, Ma D, Jiang YZ, et al. Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response. J Hematol Oncol, 2021, 14(1):98., DOI 10.1186/ s13045-021-01103-4 Granito A, Muratori L, Lalanne C, Quarneti C, Ferri S, Guidi M, et al. Hepatocellular carcinoma in viral and autoimmune liver diseases: Role of CD4+ CD25+ Foxp3+ regulatory T cells in the immune microenvironment [J]. World J Gastroenterol, 2021, 27(22):2994–3009., DOI 10.3748/wjg.v27.i22.2994 Shen LS, Wang J, Shen DF, Yuan XL, Dong P, Li MX, et al. CD4(+)CD25(+, CD127(low/-, regulatory T cells express Foxp3 and suppress effector T cell proliferation and contribute to gastric cancers progression [J]. Clin Immunol, 2009, 131(1):109–18., DOI 10.1016/j.clim.2008.11.010 Gambardella V, Castillo J, Tarazona N, Gimeno-Valiente F, Martinez- Ciarpaglini C, Cabeza-Segura M, et al. The role of tumor-associated macrophages in gastric cancer development and their potential as a therapeutic target. Cancer Treat Rev, 2020, 86:102015., DOI 10.1016/j.ctrv.2020.102015 Demkow U. Neutrophil extracellular traps, NETs, in cancer invasion, evasion and metastasis. Cancers, Basel,, 2021, 13(17):4495., DOI 10.3390/cancers13174495 Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunol Rev, 2017, 276(1):97–111., DOI 10.1111/imr.12520 Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol, 2001, 2(3):261–8., DOI 10.1038/85330 Wang C, Li Y, Jia L, Kim JK, Li J, Deng P, et al. CD276 expression enables squamous cell carcinoma stem cells to evade immune surveillance. Cell Stem Cell, 2021, 28(9):1597–613.e7. e7., DOI 10.1016/j.stem.2021.04.011 Nagase H, Takeoka T, Urakawa S, Morimoto-Okazawa A, Kawashima A, Iwahori K, et al. ICOS(+, Foxp3(+, TILs in gastric cancer are prognostic markers and effector regulatory T cells associated with Helicobacter pylori [J]. Int J Cancer, 2017, 140(3):686–95., DOI 10.1002/ijc.30475 Zhang X, Xie K, Zhou H, Wu Y, Li C, Liu Y, et al. Role of non-coding RNAs and RNA modifiers in cancer therapy resistance. Mol Cancer, 2020, 19(1):47., DOI 10.1186/s12943-020-01171-z Das PM, Singal R. DNA methylation and cancer. J Clin Oncol, 2004, 22(22): 4632–42., DOI 10.1200/JCO.2004.07.151 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610893 EP 1610910 DI 10.3389/pore.2023.1610893 PG 18 ER PT J AU Lu, Ch Feng, J Yao, Z Shi, L Li, J AF Lu, Chang Feng, Jizhen Yao, Zhigang Shi, Lei Li, Jiamei TI Case report: Gastric adenosquamous carcinoma with EBV-positive component of squamous cell carcinoma mixed with gastric carcinoma with lymphoid stroma: A novel case report and literature review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE case report; gastric cancer; Epstein-Barr virus; adenosquamous carcinoma; gastric carcinoma with lymphoid stroma ID case report; gastric cancer; Epstein-Barr virus; adenosquamous carcinoma; gastric carcinoma with lymphoid stroma AB Background: Gastric adenosquamous carcinoma with EBV-positive component of squamous cell carcinoma mixed with gastric carcinoma with lymphoid stroma are extremely unusual variants of gastric carcinoma. We herein reported such a case and summarized five related cases that have been reported previously. Case presentation: A 59-year-old man was admitted to our hospital with upper abdominal discomfort and acid reflux. Gastric endoscopic examination revealed an irregular ulcer in the gastric angle. Biopsy of the lesion revealed adenocarcinoma. The patient underwent laparoscopic distal gastrectomy with lymph node dissection subsequently. Histologically, the tumor showed coexistence of GASC and GCLS. SCC and GCLS were positive for EBER in situ hybridization, while adenocarcinoma component was negative. Accordingly, the present case was diagnosed as GASC with EBV-positive component of SCC mixed with GCLS. Conclusion: GASC with EBV-positive component of SCC mixed with GCLS is extremely rare. Although the pathogenesis of GASC and the role of EBV in the development of an ASC component have not been fully elucidated, this case will help clinicians and pathologists better understand this special subtype of gastric tumor. C1 [Lu, Chang] Shandong University, Shandong Provincial Hospital, Department of PathologyJinan, Shandong, China. [Feng, Jizhen] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of RadiologyJinan, Shandong, China. [Yao, Zhigang] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, Shandong, China. [Shi, Lei] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of GastroenterologyJinan, Shandong, China. [Li, Jiamei] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, Shandong, China. RP Li, J (reprint author), Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of Pathology, Jinan, China. EM lijiamei366@126.com CR Kuroda N, Oonishi K, Inoue K, OharaM,Mizuno K, Lee GH. Lymphoepitheliomalike carcinoma of the stomach associated with adenosquamous carcinoma. Med Mol Morphol, 2010, 43(3):170–3., DOI 10.1007/s00795-009-0445-z Oya HTHKT. Adenosquamous carcinoma, with a component of carcinoma with lymphoid stroma positive for EBER-1, a case report. Stomach Intestine, 2010, 45:1987–97. Miyake H, Miyasaka C, Ishida M, Miki H, Inoue K, Tsuta K. Simultaneous gastric adenosquamous carcinoma and gastric carcinoma with lymphoid stroma: A case report. Mol Clin Oncol, 2019, 11(1):77–80., DOI 10.3892/mco.2019.1860 Cao F, Yan Y, Niu D, Huang X, Jia L, Diao X, et al. Epstein-barr virus-associated gastric adenosquamous carcinoma with concurrent gastric carcinoma with lymphoid stroma: A case report and review of the literature. Bmc Gastroenterol, 2022, 22(1):346., DOI 10.1186/s12876-022-02417-4 Ji-Hoon K, Dae-Woon E. A case of multifocal EBV associated gastric carcinoma with focal squamous differentiation. Indian J Pathol Microbiol, 2019, 62(3):505–7., DOI 10.4103/IJPM.IJPM_174_18 Tokunaga M, Land CE, Uemura Y, Tokudome T, Tanaka S, Sato E. Epstein-Barr virus in gastric carcinoma. Am J Pathol, 1993, 143(5): 1250–4. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature, 2014, 513(7517):202–9., DOI 10.1038/nature13480 Fukayama M, Abe H, Kunita A, Shinozaki-Ushiku A, Matsusaka K, Ushiku T, et al. Thirty years of Epstein-Barr virus-associated gastric carcinoma. Virchows Arch, 2020, 476(3):353–65., DOI 10.1007/s00428-019-02724-4 Feng F, Zheng G, Qi J, Xu G, Wang F, Wang Q, et al. Clinicopathological features and prognosis of gastric adenosquamous carcinoma. Sci Rep, 2017, 7(1): 4597., DOI 10.1038/s41598-017-04563-2 Bansal RK, Sharma P, Kaur R, Arora A. Primary gastric adenosquamous carcinoma in an Indian male. Indian J Pathol Microbiol, 2013, 56(4):416–8., DOI 10.4103/0377-4929.125357 Kim YS, HeoWS, Chae KH, Gang YS, Jung JH, Kim SH, et al. Clinicopathological features and differences of p53 and Ki-67 expression in adenosquamous and squamous cell carcinomas of the stomach. Korean J Gastroenterol, 2006, 47(6):425–31. Shirahige A, Suzuki H, Oda I, Sekiguchi M, Mori G, Abe S, et al. Fatal submucosal invasive gastric adenosquamous carcinoma detected at surveillance after gastric endoscopic submucosal dissection. World J Gastroenterol, 2015, 21(14):4385–90., DOI 10.3748/wjg.v21.i14.4385 Yoshida K, Manabe T, Tsunoda T, Kimoto M, Tadaoka Y, Shimizu M. Early gastric cancer of adenosquamous carcinoma type: Report of a case and review of literature. Jpn J Clin Oncol, 1996, 26(4):252–7., DOI 10.1093/oxfordjournals.jjco. a023224 Chu YX, Gong HY, Hu QY, Song QB. Adenosquamous carcinoma may have an inferior prognosis to signet ring cell carcinoma in patients with stages I and II gastric cancer. World J Gastrointest Oncol, 2020, 12(1):101–12., DOI 10.4251/wjgo. v12.i1.101 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610902 EP 1610907 DI 10.3389/pore.2023.1610902 PG 6 ER PT J AU Kong, L Yang, M Wan, Z Wang, L AF Kong, Linghong Yang, Ming Wan, Zhiyi Wang, Lining TI Cohort size required for prognostic genes analysis of stage II/III esophageal squamous cell carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE esophageal squamous cell carcinoma; prognostic genes analysis; power-law; events number; cohort size ID esophageal squamous cell carcinoma; prognostic genes analysis; power-law; events number; cohort size AB Background: Few overlaps between prognostic biomarkers are observed among different independently performed genomic studies of esophageal squamous cell carcinoma (ESCC). One of the reasons for this is the insufficient cohort size. How many cases are needed to prognostic genes analysis in ESCC? Methods: Here, based on 387 stage II/III ESCC cases analyzed by wholegenome sequencing from one single center, effects of cohort size on prognostic genes analysis were investigated. Prognostic genes analysis was performed in 100 replicates at each cohort size level using a random resampling method. Results: The number of prognostic genes followed a power-law increase with cohort size in ESCC patients with stage II and stage III, with exponents of 2.27 and 2.25, respectively. Power-law curves with increasing events number were also observed in stage II and III ESCC, respectively, and they almost overlapped. The probability of obtaining statistically significant prognostic genes shows a logistic cumulative distribution function with respect to cohort size. To achieve a 100% probability of obtaining statistically significant prognostic genes, the minimum cohort sizes required in stage II and III ESCC were approximately 95 and 60, respectively, corresponding to a number of outcome events of 33 and 36, respectively. Conclusion: In summary, the number of prognostic genes follows a power-law growth with the cohort size or events number in ESCC. The minimum events number required to achieve a 100% probability of obtaining a statistically significant prognostic gene is approximately 35. C1 [Kong, Linghong] Beijing Chuiyangliu Hospital, Department of PathologyBeijing, China. [Yang, Ming] Tsinghua University, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary CenterBeijing, China. [Wan, Zhiyi] Beijing Chuiyangliu Hospital, Department of PathologyBeijing, China. [Wang, Lining] Beijing Chuiyangliu Hospital, Department of PathologyBeijing, China. RP Wang, L (reprint author), Beijing Chuiyangliu Hospital, Department of Pathology, Beijing, China. EM wanglining1001@126.com CR Lam AK. Molecular biology of esophageal squamous cell carcinoma. Crit Rev Oncol Hematol, 2000, 33:71–90., DOI 10.1016/s1040-8428(99)00054-2 ArnalMJD, Arenas AF, Arbeloa AL. Esophageal cancer: Risk factors, screening and endoscopic treatment inWestern and Eastern countries. World J Gastroenterol, 2015, 21:7933–43., DOI 10.3748/wjg.v21.i26.7933 Lin DC, Wang MR, Koeffler HP. Genomic and epigenomic aberrations in esophageal squamous cell carcinoma and implications for patients. Gastroenterology, 2018, 154:374–89., DOI 10.1053/j.gastro.2017.06.066 Lin DC, Hao JJ, Nagata Y, Xu L, Shang L,Meng X, et al. Genomic and molecular characterization of esophageal squamous cell carcinoma. Nat Genet, 2014, 46: 467–73., DOI 10.1038/ng.2935 Gao YB, Chen ZL, Li JG, Hu XD, Shi XJ, Sun ZM, et al. Genetic landscape of esophageal squamous cell carcinoma. Nat Genet, 2014, 46:1097–102., DOI 10.1038/ ng.3076 The Cancer Genome Atlas Research Network. Integrated genomic characterization of oesophageal carcinoma. Nature, 2017, 541:169–75., DOI 10. 1038/nature20805 Moody S, Senkin S, Islam SMA, Wang J, Nasrollahzadeh D, Cortez Cardoso Penha R, et al. Mutational signatures in esophageal squamous cell carcinoma from eight countries with varying incidence. Nat Genet, 2021, 53:1553–63., DOI 10.1038/ s41588-021-00928-6 Cui Y, Chen H, Xi R, Cui H, Zhao Y, Xu E, et al. Whole-genome sequencing of 508 patients identifies key molecular features associated with poor prognosis in esophageal squamous cell carcinoma. Cell Res, 2020, 30:902–13., DOI 10.1038/ s41422-020-0333-6 Song Y, Li L, Ou Y, Gao Z, Li E, Li X, et al. Identification of genomic alterations in oesophageal squamous cell cancer. Nature, 2014, 509:91–5., DOI 10.1038/ nature13176 Sawada G, Niida A, Uchi R, Hirata H, Shimamura T, Suzuki Y, et al. Genomic landscape of esophageal squamous cell carcinoma in a Japanese population. Gastroenterology, 2016, 150:1171–82., DOI 10.1053/j.gastro.2016. 01.035 Zhang N, Shi J, Shi X, Chen W, Liu J. Mutational characterization and potential prognostic biomarkers of Chinese patients with esophageal squamous cell carcinoma. Onco Targets Ther, 2020, 13:12797–809., DOI 10.2147/OTT. S275688 Ein-Dor L, Kela I, Getz G, Givol D, Domany E. Outcome signature genes in breast cancer: Is there a unique set? Bioinformatics, 2005, 21:171–8., DOI 10.1093/ bioinformatics/bth469 Riley RD, Snell KI, Ensor J, BurkeDL,Harrell FE, Jr,Moons KG, et al. Minimum sample size for developing a multivariable prediction model: PART II - binary and time-to-event outcomes. Stat Med, 2019, 38:1276–96., DOI 10.1002/sim.7992 Schober P, Vetter TR. Survival analysis and interpretation of time-to-event data: The tortoise and the hare. Anesth Analg, 2018, 127:792–8., DOI 10.1213/ANE. 0000000000003653 In J, Lee DK. Survival analysis: Part II - applied clinical data analysis. Korean J Anesthesiol, 2019, 72:441–57., DOI 10.4097/kja.19183 Mayakonda A, Lin DC, Assenov Y, Plass C, Koeffler HP. Maftools: Efficient and comprehensive analysis of somatic variants in cancer. Genome Res, 2018, 28: 1747–56., DOI 10.1101/gr.239244.118 Moons KG, Royston P, Vergouwe Y, Grobbee DE, Altman DG. Prognosis and prognostic research: What, why, and how? Bmj, 2009, 338:b375., DOI 10.1136/bmj. b375 Jinks RC, Royston P, Parmar MK. Discrimination-based sample size calculations for multivariable prognostic models for time-to-event data. BMC Med Res Methodol, 2015, 15:82., DOI 10.1186/s12874-015-0078-y NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610909 EP 1610912 DI 10.3389/pore.2023.1610909 PG 4 ER PT J AU Cheng, H Zong, L Yu, Sh Chen, J Wan, X Xiang, Y Yang, J AF Cheng, Hongyan Zong, Liju Yu, Shuangni Chen, Jie Wan, Xirun Xiang, Yang Yang, Junjun TI Expression of the immune targets in tumor-infiltrating immunocytes of gestational trophoblastic neoplasia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gestational trophoblastic neoplasia; tumor-infiltrating immunocyte; PD-1; TIM-3; GAL-9; LAG-3 ID gestational trophoblastic neoplasia; tumor-infiltrating immunocyte; PD-1; TIM-3; GAL-9; LAG-3 AB Objectives: To evaluate the expression of emerging immune targets in the tumor-infiltrating immunocytes (TIIs) of human gestational trophoblastic neoplasia (GTN) specimens, and to analyze the correlation between the expression patterns and prognosis of GTN patients. Methods: Between January 2008 and December 2017, patients who were diagnosed histologically with GTN were included in this study. The expression densities of LAG-3, TIM-3, GAL-9, PD-1, CD68, CD8, and FOXP3 in the TIIs were assessed independently by two pathologists blinded to clinical outcomes. The expression patterns and correlation with patient outcomes were analyzed to identify prognostic factors. Results: We identified 108 patients with GTN, including 67 with choriocarcinoma, 32 with placental site trophoblastic tumor (PSTT), and 9 with epithelioid trophoblastic tumor (ETT). Almost all GTN patients showed expression of GAL-9, TIM-3, and PD-1 in TIIs (100%, 92.6%, and 90.7%, respectively); LAG-3 was expressed in 77.8% of the samples. The expression densities of CD68 and GAL-9 were significantly higher in choriocarcinoma than that in PSTT and ETT. The TIM-3 expression density in choriocarcinoma was higher than that in PSTT. In addition, the expression density of LAG-3 in the TIIs of choriocarcinoma and PSTT was higher than that in ETT. There was no statistical difference in the expression pattern of PD-1 among different pathological subtypes. The positive expression of LAG- 3 in tumor TIIs was a prognostic factor for disease recurrence, and patients with positive expression of LAG-3 in the TIIs had poorer disease-free survival (p = 0.026). Conclusion: Our study evaluated the expression of immune targets PD-1, TIM-3, LAG-3, and GAL-9 in the TIIs of GTN patients and found that they were widely expressed but not associated with patients’ prognoses, excepting the positive expression of LAG-3 was a prognostic factor for disease recurrence. C1 [Cheng, Hongyan] Peking Union Medical College Hospital, Department of Obstetrics and GynecologyBeijing, China. [Zong, Liju] Peking Union Medical College Hospital, Department of Obstetrics and GynecologyBeijing, China. [Yu, Shuangni] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of PathologyBeijing, China. [Chen, Jie] Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Department of PathologyBeijing, China. [Wan, Xirun] Peking Union Medical College Hospital, Department of Obstetrics and GynecologyBeijing, China. [Xiang, Yang] Peking Union Medical College Hospital, Department of Obstetrics and GynecologyBeijing, China. [Yang, Junjun] Peking Union Medical College Hospital, Department of Obstetrics and GynecologyBeijing, China. RP Yang, J (reprint author), Peking Union Medical College Hospital, Department of Obstetrics and Gynecology, Beijing, China. EM yangjunjun@pumch.cn CR Ngan HYS, Seckl MJ, Berkowitz RS, Xiang Y, Golfier F, Sekharan PK, et al. Update on the diagnosis and management of gestational trophoblastic disease. Int J Gynaecol Obstet, 2018, 143(2):79–85., DOI 10.1002/ijgo.12615 Zong L, Yang J, Wang X, Kong Y, Ren T, Zhao J, et al. Management and prognosis of patients with liver metastases from gestational trophoblastic neoplasia: A retrospective cohort study. Cancer Manag Res, 2018, 10:557., DOI 10.2147/CMAR. S160606 Kong Y, Yang J, Jiang F, Zhao J, Ren T, Li J, et al. Clinical characteristics and prognosis of ultra high-risk gestational trophoblastic neoplasia patients: A retrospective cohort study. Gynecol Oncol, 2017, 146:81–6., DOI 10.1016/j.ygyno. 2017.04.010 Ghorani E, Kaur B, Fisher RA, Short D, Joneborg U, Carlson JW, et al. Pembrolizumab is effective for drug-resistant gestational trophoblastic neoplasia. Lancet, 2017, 390:2343–5., DOI 10.1016/S0140-6736(17)32894-5 Nakashima A, Shima T, Aoki A, Kawaguchi M, Yasuda I, Tsuda S, et al. Molecular and immunological developments in placentas. Hum Immunol, 2021, 82:317–24., DOI 10.1016/j.humimm.2021.01.012 Veras E, Kurman RJ,Wang TL, Shih IM. PD-L1 expression in human placentas and gestational trophoblastic diseases. Int J Gynecol Pathol, 2017, 36:146–53., DOI 10.1097/pgp.0000000000000305 Bolze PA, Patrier S, Massardier J, Hajri T, Abbas F, Schott AM, et al. PD-L1 expression in premalignant and malignant trophoblasts from gestational trophoblastic diseases is ubiquitous and independent of clinical outcomes. Int J Gynrcol Cancer, 2017, 27:554–61., DOI 10.1097/IGC.0000000000000892 Zong L, Zhang M, Wang W, Wan X, Yang J, Xiang Y. PD-L1, B7-H3 and VISTA are highly expressed in gestational trophoblastic neoplasia. Histopathology, 2019, 75:421–30., DOI 10.1111/his.13882 Yang J, Zong L, Wang J, Wan X, Feng F, Xiang Y. Epithelioid trophoblastic tumors: Treatments, outcomes, and potential therapeutic targets. J Cancer, 2019, 10:11–9., DOI 10.7150/jca.28134 Kohorn EI. The new FIGO 2000 staging and risk factor scoring system for gestational trophoblastic disease: Description and critical assessment. Int J Gynrcol Cancer, 2010, 11:73–7., DOI 10.1046/j.1525-1438.2001.011001073.x Jiang F, Xiang Y, Guo LN. Laparoscopic diagnosis and treatment of an isolated epithelioid trophoblastic tumor in recto-uterine pouch. J Obstet Gynaecol Res, 2018, 44:960–5., DOI 10.1111/jog.13593 Blanc-Durand F, Genestie C, Galende EY, Gouy S, Morice P, Pautier P, et al. Distribution of novel immune-checkpoint targets in ovarian cancer tumor microenvironment: A dynamic landscape. Gynecol Oncol, 2021, 160:279–84., DOI 10.1016/j.ygyno.2020.09.045 Carvajal-Hausdorf D, Altan M, Velcheti V, Gettinger SN, Herbst RS, Rimm DL, et al. Expression and clinical significance of PD-L1, B7-H3, B7-H4 and TILs in human small cell lung Cancer, SCLC). J Immunother Cancer, 2019, 7:65., DOI 10. 1186/s40425-019-0540-1 Mattox AK, Lee J,Westra WH, Pierce RH, Ghossein R, Faquin WC, et al. PD- 1 expression in head and neck squamous cell carcinomas derives primarily from functionally anergic CD4(+, TILs in the presence of PD-L1(+, TAMs. Cancer Res, 2017, 77:6365–74., DOI 10.1158/0008-5472.can-16-3453 Gagliani N, Magnani CF, Huber S, Gianolini ME, Pala M, Licona-Limon P, et al. Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat Med, 2013, 19:739–46., DOI 10.1038/nm.3179 Grosso JF, Kelleher CC, Harris TJ, Maris CH, Hipkiss EL, Angelo DM, et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J Clin Invest, 2007, 117:3383–92., DOI 10.1172/JCI31184 Lui Y, Davis SJ. LAG-3: A very singular immune checkpoint. Nat Immunol, 2018, 19:1278–9., DOI 10.1038/s41590-018-0257-1 Sbracia M, Scarpellini F, Mastrone M, Grasso JA. HLA antigen sharing in Italian couples in which women were affected by gestational trophoblastic tumors. Am J Reprod Immunol, 1996, 35:252–5., DOI 10.1111/j.1600-0897.1996.tb00039.x Tu L, Yang H, Zhou Y, Guan R. Assessment of the expression of the immune checkpoint molecules PD-1, CTLA4, TIM-3 and LAG-3 across different cancers in relation to treatment response, tumor-infiltrating immune cells and survival. Int J Cancer, 2019, 147:423–39., DOI 10.1002/ijc.32785 Heeren AM, Rotman J, Stam AGM, Pocorni N, Gassama AA, Samuels S, et al. Efficacy of PD-1 blockade in cervical cancer is related to a CD8+FoxP3+CD25+ T-cell subset with operational effector functions despite high immune checkpoint levels. J ImmunoTherapy Cancer, 2019, 7:43., DOI 10.1186/s40425-019-0526-z Matsuzaki J, Gnjatic S, Mhawechfauceglia P, Beck A, Miller A, Tsuji T, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A, 2010, 107: 7875–80., DOI 10.1073/pnas.1003345107 Cheng H, Zong L, Kong Y, Gu Y, Yang J, Xiang Y. Emerging targets of immunotherapy in gynecologic cancer. Emerging Targets Immunother Gynecol Cancer, 2020, 13:11869–82., DOI 10.2147/ott.s282530 Kuchroo VK. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol, 2005, 6:1245–52., DOI 10.1038/ni1271 Sabatos CA, Chakravarti S, Cha E, Schubart A, Sanchez-Fueyo A, Zheng XX, et al. Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol, 2003, 4:1102–10., DOI 10.1038/ ni988 Wang K, Chen Z, Wu R, Yin J, Fan M, Xu X. Prognostic role of high gal-9 expression in solid tumours: A meta-analysis. Cell Physiol Biochem, 2018, 45: 993–1002., DOI 10.1159/000487294 Lv R, Bao Q, Li Y. Regulation of M1-type and M2-type macrophage polarization in RAW264.7 cells by Galectin-9. Mol Med Rep, 2017, 16:9111–9., DOI 10.3892/mmr.2017.7719 Dumas G, Dufresne M, Asselin E, Girouard J, Carrier C, Reyes-Moreno C. CD40 pathway activation reveals dual function for macrophages in human endometrial cancer cell survival and invasion. Cancer Immunol Immunother, 2013, 62:273–83., DOI 10.1007/s00262-012-1333-2 Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell, 2010, 141:39–51., DOI 10.1016/j.cell.2010.03.014 Bingle L, Brown NJ, Lewis CE. The role of tumour-associated macrophages in tumour progression: Implications for new anticancer therapies. J Pathol, 2002, 196: 254–65., DOI 10.1002/path.1027 Condeelis J, Pollard JW.Macrophages: Obligate partners for tumor cell migration, invasion, and metastasis. Cell, 2006, 124:263–6., DOI 10.1016/j.cell.2006.01.007 Duraiswamy J, Freeman GJ, Coukos G. Therapeutic PD-1 pathway blockade augments with other modalities of immunotherapy T-cell function to prevent immune decline in ovarian cancer. Cancer Res, 2013, 73:6900–12., DOI 10.1158/ 0008-5472.can-13-1550 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610918 EP 1610925 DI 10.3389/pore.2023.1610918 PG 8 ER PT J AU Sun, A Tian, X Chen, Y Yang, W Lin, Q AF Sun, Aiqin Tian, Xianyan Chen, Yifei Yang, Wannian Lin, Qiong TI Emerging roles of the HECT E3 ubiquitin ligases in gastric cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE gastric cancer; ubiquitination; tumor suppressor; HECT E3 ubiquitin ligases; oncoprotein ID gastric cancer; ubiquitination; tumor suppressor; HECT E3 ubiquitin ligases; oncoprotein AB Gastric cancer (GC) is one of the most pernicious gastrointestinal tumors with extraordinarily high incidence and mortality. Ubiquitination modification of cellular signaling proteins has been shown to play important roles in GC tumorigenesis, progression, and prognosis. The E3 ubiquitin ligase is the crucial enzyme in the ubiquitination reaction and determines the specificity of ubiquitination substrates, and thus, the cellular effects. The HECT E3 ligases are the second largest E3 ubiquitin ligase family characterized by containing a HECT domain that has E3 ubiquitin ligase activity. The HECT E3 ubiquitin ligases have been found to engage in GC progression. However, whether HECT E3 ligases function as tumor promoters or tumor suppressors in GC remains controversial. In this review, we will focus on recent discoveries about the role of the HECT E3 ubiquitin ligases, especially members of the NEDD4 and other HECT E3 ligase subfamilies, in GC. C1 [Sun, Aiqin] Jiangsu University, School of MedicineZhenjiang, China. [Tian, Xianyan] Jiangsu University, School of MedicineZhenjiang, China. [Chen, Yifei] Jiangsu University, School of MedicineZhenjiang, China. [Yang, Wannian] Jiangsu University, School of MedicineZhenjiang, China. [Lin, Qiong] Jiangsu University, School of MedicineZhenjiang, China. RP Lin, Q (reprint author), Jiangsu University, School of Medicine, Zhenjiang, China. EM qlin@ujs.edu.cn CR Hershko A. Ubiquitin: Roles in protein modification and breakdown. Cell, 1983, 34:11–2., DOI 10.1016/0092-8674(83)90131-9 Pickart CM. Mechanisms underlying ubiquitination. Annu Rev Biochem, 2001, 70:503–33., DOI 10.1146/annurev.biochem.70.1.503 Rape M. Ubiquitylation at the crossroads of development and disease. Nat Rev Mol Cel Biol, 2018, 19:59–70., DOI 10.1038/nrm.2017.83 Kwon YT, Ciechanover A. The ubiquitin code in the ubiquitin-proteasome system and autophagy. Trends Biochemical Sciences, 2017, 42:873–86., DOI 10.1016/ j.tibs.2017.09.002 Ciechanover A. The unravelling of the ubiquitin system. Nat Rev Mol Cel Biol, 2015, 16:322–4., DOI 10.1038/nrm3982 Schulman BA, Harper JW. Ubiquitin-like protein activation by E1 enzymes: The apex for downstream signalling pathways. Nat Rev Mol Cel Biol, 2009, 10: 319–31., DOI 10.1038/nrm2673 Ye Y, Rape M. Building ubiquitin chains: E2 enzymes at work. Nat Rev Mol Cel Biol, 2009, 10:755–64., DOI 10.1038/nrm2780 Yang Q, Zhao J, Chen D, Wang Y. E3 ubiquitin ligases: Styles, structures and functions. Mol Biomed, 2021, 2:23., DOI 10.1186/s43556-021-00043-2 Buetow L, Huang DT. Structural insights into the catalysis and regulation of E3 ubiquitin ligases. Nat Rev Mol Cel Biol, 2016, 17:626–42., DOI 10.1038/nrm. 2016.91 Deng L, Meng T, Chen L, Wei W, Wang P. The role of ubiquitination in tumorigenesis and targeted drug discovery. Signal Transduction Targeted Therapy, 2020, 5:11., DOI 10.1038/s41392-020-0107-0 Senft D, Qi J, Ronai ZA. Ubiquitin ligases in oncogenic transformation and cancer therapy. Nat Rev Cancer, 2018, 18:69–88., DOI 10.1038/nrc.2017.105 Sluimer J, Distel B. Regulating the human HECT E3 ligases. Cell Mol Life Sci : CMLS, 2018, 75:3121–41., DOI 10.1007/s00018-018-2848-2 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer J clinicians, 2021, 71: 209–49., DOI 10.3322/caac.21660 Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA, et al. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology, 2020, 159:335–49., DOI 10.1053/j.gastro.2020.02.068 Shi J, Qu YP, Hou P. Pathogenetic mechanisms in gastric cancer. World J Gastroenterol, 2014, 20:13804–19., DOI 10.3748/wjg.v20.i38.13804 Shimizu D, Kanda M, Kodera Y. Review of recent molecular landscape knowledge of gastric cancer. Histology and histopathology, 2018, 33:11–26., DOI 10. 14670/hh-11-898 Kim SS, Yoo NJ, Jeong EG, KimMS, Lee SH. Expression of NEDD4-1, a PTEN regulator, in gastric and colorectal carcinomas. APMIS : Acta pathologica, microbiologica, immunologica Scand, 2008, 116:779–84., DOI 10.1111/j.1600-0463. 2008.00999.x Wang YY, Ye ZY, Zhao ZS, Tao HQ, Li SG. Systems biology approach to identification of biomarkers for metastatic progression in gastric cancer. J Cancer Res Clin Oncol, 2010, 136:135–41., DOI 10.1007/s00432-009-0644-y Sun A, Yu G, Dou X, Yan X, Yang W, Lin Q. Nedd4-1 is an exceptional prognostic biomarker for gastric cardia adenocarcinoma and functionally associated with metastasis. Mol Cancer, 2014, 13:248., DOI 10.1186/1476-4598- 13-248 Yang Z, Yuan XG, Chen J, Lu NH. Is NEDD4-1 a negative regulator of phosphatase and tensin homolog in gastric carcinogenesis? World J Gastroenterol, 2012, 18:6345–8., DOI 10.3748/wjg.v18.i43.6345 Gao C, Pang L, Ren C, Ma T. Decreased expression of Nedd4L correlates with poor prognosis in gastric cancer patient. Med Oncol, Northwood, Lond England,, 2012, 29:1733–8., DOI 10.1007/s12032-011-0061-3 Jiang X, Zhang S, Yin Z, Sheng Y, Yan Q, Sun R, et al. The correlation between NEDD4L and HIF-1α levels as a gastric cancer prognostic marker. Int J Med Sci, 2019, 16:1517–24., DOI 10.7150/ijms.34646 Gen Y, Yasui K, Kitaichi T, Iwai N, Terasaki K, Dohi O, et al. ASPP2 suppresses invasion and TGF-β1-induced epithelial-mesenchymal transition by inhibiting Smad7 degradation mediated by E3 ubiquitin ligase ITCH in gastric cancer. Cancer Lett, 2017, 398:52–61., DOI 10.1016/j.canlet.2017. 04.002 Peng Y, Wang HH. Cir-ITCH inhibits gastric cancer migration, invasion and proliferation by regulating the Wnt/β-catenin pathway. Scientific Rep, 2020, 10: 17443., DOI 10.1038/s41598-020-74452-8 Ghasemi S, Emadi-Baygi M, Nikpour P. Down-regulation of circular RNA ITCH and circHIPK3 in gastric cancer tissues. Turkish J Med Sci, 2019, 49:687–95., DOI 10.3906/sag-1806-50 Zhang L, Wu Z, Ma Z, Liu H, Wu Y, Zhang Q. WWP1 as a potential tumor oncogene regulates PTEN-Akt signaling pathway in human gastric carcinoma. Tumour Biol : J Int Soc Oncodevelopmental Biol Med, 2015, 36:787–98., DOI 10.1007/ s13277-014-2696-0 Li Q, Li Z, Wei S, Wang W, Chen Z, Zhang L, et al. Overexpression of miR- 584-5p inhibits proliferation and induces apoptosis by targeting WW domaincontaining E3 ubiquitin protein ligase 1 in gastric cancer. J Exp Clin Cancer Res : CR, 2017, 36:59., DOI 10.1186/s13046-017-0532-2 Ma L, Chen X, Li C, Cheng R, Gao Z, Meng X, et al. miR-129-5p and -3p cotarget WWP1 to suppress gastric cancer proliferation and migration. J Cell Biochem, 2018, 120:7527–38., DOI 10.1002/jcb.28027 Wang K, Liu J, Zhao X, Li H, Luo G, Yu Y, et al. WWP2 regulates proliferation of gastric cancer cells in a PTEN-dependent manner. Biochem biophysical Res Commun, 2020, 521:652–9., DOI 10.1016/j.bbrc.2019.10.179 Tao Y, Sun C, Zhang T, Song Y. SMURF1 promotes the proliferation, migration and invasion of gastric cancer cells. Oncol Rep, 2017, 38:1806–14., DOI 10.3892/or.2017.5825 Lu L, Wu M, Lu Y, Zhao Z, Liu T, Fu W, et al. MicroRNA-424 regulates cisplatin resistance of gastric cancer by targeting SMURF1 based on GEO database and primary validation in human gastric cancer tissues. OncoTargets Ther, 2019, 12:7623–36., DOI 10.2147/ott.S208275 Jiang M, Shi L, Yang C, Ge Y, Lin L, Fan H, et al. miR-1254 inhibits cell proliferation, migration, and invasion by down-regulating Smurf1 in gastric cancer. Cel Death Dis, 2019, 10:32., DOI 10.1038/s41419-018-1262-x Ji CD, Wang YX, Xiang DF, Liu Q, Zhou ZH, Qian F, et al. Kir2.1 interaction with Stk38 promotes invasion and metastasis of human gastric cancer by enhancing MEKK2-MEK1/2-ERK1/2 signaling. Cancer Res, 2018, 78:3041–53., DOI 10.1158/ 0008-5472.Can-17-3776 He Y, Zhou J, Wan Q. The E3 ligase HUWE1 mediates TGFBR2 ubiquitination and promotes gastric cancer cell proliferation, migration, and invasion. Invest New Drugs, 2021, 39:713–23., DOI 10.1007/ s10637-020-01041-x Chen YL, Li DP, Jiang HY, Yang Y, Xu LL, Zhang SC, et al. Overexpression of HACE1 in gastric cancer inhibits tumor aggressiveness by impeding cell proliferation and migration. Cancer Med, 2018, 7:2472–84., DOI 10.1002/cam4.1496 Jiang HY, Chen YL, Xu XX, Li CY, Chen Y, Li DP, et al. Ubiquitylation of cyclin C by HACE1 regulates cisplatin-associated sensitivity in gastric cancer. Clin translational Med, 2022, 12:e770., DOI 10.1002/ctm2.770 Sakata M, Kitamura YH, Sakuraba K, Goto T, Mizukami H, Saito M, et al. Methylation of HACE1 in gastric carcinoma. Anticancer Res, 2009, 29:2231–3. Adhikary S, Marinoni F, Hock A, Hulleman E, Popov N, Beier R, et al. The ubiquitin ligase HectH9 regulates transcriptional activation by Myc and is essential for tumor cell proliferation. Cell, 2005, 123:409–21., DOI 10.1016/j.cell.2005.08.016 Kim MS, Oh JE, Eom HS, Yoo NJ, Lee SH. Mutational analysis of UBR5 gene encoding an E3 ubiquitin ligase in common human cancers. Pathology, 2010, 42: 93–4., DOI 10.3109/00313020903434322 Ding F, Zhu X, Song X, Yuan P, Ren L, Chai C, et al. UBR5 oncogene as an indicator of poor prognosis in gastric cancer. Exp Ther Med, 2020, 20:7., DOI 10. 3892/etm.2020.9135 Gu J, Mao W, Ren W, Xu F, Zhu Q, Lu C, et al. Ubiquitin-protein ligase E3C maintains non-small-cell lung cancer stemness by targeting AHNAK-p53 complex. Cancer Lett, 2019, 443:125–34., DOI 10.1016/j.canlet.2018.11.029 Zhang Y, Xu J, Fu H, Wei Z, Yang D, Yan R. UBE3C promotes proliferation and inhibits apoptosis by activating the β-catenin signaling via degradation of AXIN1 in gastric cancer. Carcinogenesis, 2021, 42:285–93., DOI 10.1093/carcin/ bgaa098 Zhao W, Jin Y, Wu P, Yang J, Chen Y, Yang Q, et al. LINC00355 induces gastric cancer proliferation and invasion through promoting ubiquitination of P53. Cel Death Discov, 2020, 6:99., DOI 10.1038/s41420-020-00332-9 Rotin D, Kumar S. Physiological functions of the HECT family of ubiquitin ligases. Nat Rev Mol Cel Biol, 2009, 10:398–409., DOI 10.1038/nrm2690 Berndsen CE,Wolberger C. New insights into ubiquitin E3 ligase mechanism. Nat Struct Mol Biol, 2014, 21:301–7., DOI 10.1038/nsmb.2780 Scheffner M, Kumar S. Mammalian HECT ubiquitin-protein ligases: Biological and pathophysiological aspects. Biochim Biophys Acta, 2014, 1843: 61–74., DOI 10.1016/j.bbamcr.2013.03.024 Deshaies RJ, Joazeiro CA. RING domain E3 ubiquitin ligases. Annu Rev Biochem, 2009, 78:399–434., DOI 10.1146/annurev.biochem.78.101807.093809 Metzger MB, Pruneda JN, Klevit RE, Weissman AM. RING-Type E3 ligases: Master manipulators of E2 ubiquitin-conjugating enzymes and ubiquitination. Biochim Biophys Acta, 2014, 1843:47–60., DOI 10.1016/j.bbamcr.2013.05.026 Spratt DE, Walden H, Shaw GS. RBR E3 ubiquitin ligases: New structures, new insights, new questions. Biochem J, 2014, 458:421–37., DOI 10.1042/bj20140006 Walden H, Rittinger K. RBR ligase-mediated ubiquitin transfer: A tale with many twists and turns. Nat Struct Mol Biol, 2018, 25:440–5., DOI 10.1038/s41594- 018-0063-3 Zheng N, Shabek N. Ubiquitin ligases: Structure, function, and regulation. Annu Rev Biochem, 2017, 86:129–57., DOI 10.1146/annurev-biochem-060815- 014922 Wang Y, Argiles-Castillo D, Kane EI, Zhou A, Spratt DE. HECT E3 ubiquitin ligases - emerging insights into their biological roles and disease relevance. J Cel Sci, 2020, 133:jcs228072., DOI 10.1242/jcs.228072 Zou X, Levy-Cohen G, BlankM. Molecular functions of NEDD4 E3 ubiquitin ligases in cancer. Biochim Biophys Acta, 2015, 1856:91–106., DOI 10.1016/j.bbcan. 2015.06.005 Plant PJ, Yeger H, Staub O, Howard P, Rotin D. The C2 domain of the ubiquitin protein ligase Nedd4 mediates Ca2+-dependent plasma membrane localization. J Biol Chem, 1997, 272:32329–36., DOI 10.1074/jbc.272.51.32329 Yamaguchi K, Ohara O, Ando A, Nagase T. Smurf1 directly targets hPEM-2, a GEF for Cdc42, via a novel combination of protein interaction modules in the ubiquitin-proteasome pathway. Biol Chem, 2008, 389:405–13., DOI 10.1515/bc. 2008.036 Tian M, Bai C, Lin Q, Lin H, Liu M, Ding F, et al. Binding of RhoA by the C2 domain of E3 ligase Smurf1 is essential for Smurf1-regulated RhoA ubiquitination and cell protrusive activity. FEBS Lett, 2011, 585:2199–204., DOI 10.1016/j.febslet.2011.06.016 Wiesner S, Ogunjimi AA, Wang HR, Rotin D, Sicheri F, Wrana JL, et al. Autoinhibition of the HECT-type ubiquitin ligase Smurf2 through its C2 domain. Cell, 2007, 130:651–62., DOI 10.1016/j.cell.2007.06.050 Dunn R, Klos DA, Adler AS, Hicke L. The C2 domain of the Rsp5 ubiquitin ligase binds membrane phosphoinositides and directs ubiquitination of endosomal cargo. J Cel Biol, 2004, 165:135–44., DOI 10.1083/jcb.200309026 Dodson EJ, Fishbain-Yoskovitz V, Rotem-Bamberger S, Schueler-Furman O. Versatile communication strategies among tandem WW domain repeats. Exp Biol Med, Maywood, N.J.,, 2015, 240:351–60., DOI 10.1177/1535370214566558 Bork P, Sudol M. The WW domain: A signalling site in dystrophin? Trends Biochemical Sciences, 1994, 19:531–3., DOI 10.1016/0968-0004(94)90053-1 Staub O, Dho S, Henry P, Correa J, Ishikawa T, McGlade J, et al.WWdomains of Nedd4 bind to the proline-rich PYmotifs in the epithelial Na+ channel deleted in Liddle’s syndrome. EMBO J, 1996, 15:2371–80., DOI 10.1002/j.1460-2075.1996.tb00593.x Nemergut ME, Mizzen CA, Stukenberg T, Allis CD, Macara IG. Chromatin docking and exchange activity enhancement of RCC1 by histones H2A and H2B. Science, New York, N.Y.,, 2001, 292:1540–3., DOI 10.1126/science.292.5521.1540 Zhang C, Clarke PR. Chromatin-independent nuclear envelope assembly induced by Ran GTPase in Xenopus egg extracts. Science, New York, N.Y.,, 2000, 288:1429–32., DOI 10.1126/science.288.5470.1429 Singh S, Ng J, Sivaraman J. Exploring the "Other" subfamily of HECT E3- ligases for therapeutic intervention. Pharmacol Ther, 2021, 224:107809., DOI 10. 1016/j.pharmthera.2021.107809 Grau-Bove X, Sebe-Pedros A, Ruiz-Trillo I. A genomic survey of HECT ubiquitin ligases in eukaryotes reveals independent expansions of the HECT system in several lineages. Genome Biol Evol, 2013, 5:833–47., DOI 10.1093/gbe/evt052 Marin I. Animal HECT ubiquitin ligases: Evolution and functional implications. BMC Evol Biol, 2010, 10:56., DOI 10.1186/1471-2148-10-56 Wang ZW, Hu X, Ye M, Lin M, Chu M, Shen X. NEDD4 E3 ligase: Functions and mechanism in human cancer. Semin Cancer Biol, 2020, 67:92–101., DOI 10. 1016/j.semcancer.2020.03.006 Yang B, Kumar S. Nedd4 and nedd4-2: Closely related ubiquitin-protein ligases with distinct physiological functions. Cel Death Differ, 2010, 17:68–77., DOI 10.1038/cdd.2009.84 Gao S, Alarcon C, Sapkota G, Rahman S, Chen PY, Goerner N, et al. Ubiquitin ligase Nedd4L targets activated Smad2/3 to limit TGF-beta signaling. Mol Cel, 2009, 36:457–68., DOI 10.1016/j.molcel.2009.09.043 Wang Z, Dang T, Liu T, Chen S, Li L, Huang S, et al. NEDD4L protein catalyzes ubiquitination of PIK3CA protein and regulates PI3K-AKT signaling. J Biol Chem, 2016, 291:17467–77., DOI 10.1074/jbc.M116.726083 Ding Y, Zhang Y, Xu C, Tao QH, Chen YG. HECT domain-containing E3 ubiquitin ligase NEDD4L negatively regulates Wnt signaling by targeting dishevelled for proteasomal degradation. J Biol Chem, 2013, 288:8289–98., DOI 10.1074/jbc.M112.433185 ChanW, Tian R, Lee YF, Sit ST, Lim L, Manser E. Down-regulation of active ACK1 is mediated by association with the E3 ubiquitin ligase Nedd4-2. J Biol Chem, 2009, 284:8185–94., DOI 10.1074/jbc.M806877200 Melino G, Gallagher E, Aqeilan RI, Knight R, Peschiaroli A, Rossi M, et al. Itch: A HECT-type E3 ligase regulating immunity, skin and cancer. Cel Death Differ, 2008, 15:1103–12., DOI 10.1038/cdd.2008.60 Lallemand F, Seo SR, Ferrand N, Pessah M, L’Hoste S, Rawadi G, et al. AIP4 restricts transforming growth factor-beta signaling through a ubiquitinationindependent mechanism. J Biol Chem, 2005, 280:27645–53., DOI 10.1074/jbc. M500188200 Park SH, Jung EH, Kim GY, Kim BC, Lim JH, Woo CH. Itch E3 ubiquitin ligase positively regulates TGF-β signaling to EMT via Smad7 ubiquitination. Mol Cell, 2015, 38:20–5., DOI 10.14348/molcells.2015.2120 Zhi X, Chen C. WWP1: A versatile ubiquitin E3 ligase in signaling and diseases. Cell Mol Life Sci : CMLS, 2012, 69:1425–34., DOI 10.1007/s00018-011- 0871-7 Chen W, Jiang X, Luo Z. WWP2: A multifunctional ubiquitin ligase gene. Pathol Oncol Res : POR, 2014, 20:799–803., DOI 10.1007/s12253-014-9838-y Cao Y, Zhang L. A Smurf1 tale: Function and regulation of an ubiquitin ligase in multiple cellular networks. Cell Mol Life Sci : CMLS, 2013, 70:2305–17., DOI 10. 1007/s00018-012-1170-7 Fu L, Cui CP, Zhang X, Zhang L. The functions and regulation of Smurfs in cancers. Semin Cancer Biol, 2020, 67:102–16., DOI 10.1016/j.semcancer.2019.12.023 Qi L, Xu X, Qi X. The giant E3 ligase HUWE1 is linked to tumorigenesis, spermatogenesis, intellectual disability, and inflammatory diseases. Front Cell Infect Microbiol, 2022, 12:905906., DOI 10.3389/fcimb.2022.905906 Zhang L, Anglesio MS, O’Sullivan M, Zhang F, Yang G, Sarao R, et al. The E3 ligase HACE1 is a critical chromosome 6q21 tumor suppressor involved in multiple cancers. Nat Med, 2007, 13:1060–9., DOI 10.1038/nm1621 Matta-Camacho E, Kozlov G, Menade M, Gehring K. Structure of the HECT C-lobe of the UBR5 E3 ubiquitin ligase. Acta Crystallographica Section F, Struct Biol crystallization Commun, 2012, 68:1158–63., DOI 10.1107/s1744309112036937 Shearer RF, Iconomou M, Watts CK, Saunders DN. Functional roles of the E3 ubiquitin ligase UBR5 in cancer. Mol Cancer Res : MCR, 2015, 13:1523–32., DOI 10.1158/1541-7786.Mcr-15-0383 Yang M, Jiang N, Cao QW, Ma MQ, Sun Q. The E3 ligase UBR5 regulates gastric cancer cell growth by destabilizing the tumor suppressor GKN1. Biochem biophysical Res Commun, 2016, 478:1624–9., DOI 10.1016/j.bbrc.2016.08.170 Michel MA, Elliott PR, Swatek KN, Simicek M, Pruneda JN,Wagstaff JL, et al. Assembly and specific recognition of k29-and k33-linked polyubiquitin. Mol Cel, 2015, 58:95–109., DOI 10.1016/j.molcel.2015.01.042 Singh S, Sivaraman J. Crystal structure of HECT domain of UBE3C E3 ligase and its ubiquitination activity. Biochem J, 2020, 477:905–23., DOI 10.1042/ bcj20200027 Pan SJ, Zhan SK, Ji WZ, Pan YX, LiuW, Li DY, et al. Ubiquitin-protein ligase E3C promotes glioma progression by mediating the ubiquitination and degrading of Annexin A7. Scientific Rep, 2015, 5:11066., DOI 10.1038/srep11066 Tang L, Yi XM, Chen J, Chen FJ, Lou W, Gao YL, et al. Ubiquitin ligase UBE3C promotes melanoma progression by increasing epithelial-mesenchymal transition in melanoma cells. Oncotarget, 2016, 7:15738–46., DOI 10.18632/ oncotarget.7393 Wen JL, Wen XF, Li RB, Jin YC,Wang XL, Zhou L, et al.UBE3C promotes growth and metastasis of renal cell carcinoma via activating Wnt/β-catenin pathway. PloS one, 2015, 10:e0115622., DOI 10.1371/journal.pone.0115622 Jiang JH, Liu YF, Ke AW, Gu FM, Yu Y, Dai Z, et al. Clinical significance of the ubiquitin ligase UBE3C in hepatocellular carcinoma revealed by exome sequencing. Hepatol, Baltimore, Md, 2014, 59:2216–27., DOI 10.1002/hep.27012 Xiong J,Wei B, Ye Q, LiuW.MiR-30a-5p/UBE3C axis regulates breast cancer cell proliferation and migration. Biochem biophysical Res Commun, 2019, 516: 1013–8., DOI 10.1016/j.bbrc.2016.03.069 Zhang G, Zhu Q, Yan X, CiM, Zhao E, Hou J, et al. HECTD3 promotes gastric cancer progression by mediating the polyubiquitination of c-MYC. Cel Death Discov, 2022, 8:185., DOI 10.1038/s41420-022-01001-9 Duan S, Pagano M. Ubiquitin ligases in cancer: Functions and clinical potentials. Cel Chem Biol, 2021, 28:918–33., DOI 10.1016/j.chembiol.2021. 04.008 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610931 EP 1610938 DI 10.3389/pore.2023.1610931 PG 8 ER PT J AU Bodis, F Orosz, G Toth, J Szabo, M Elo, GL Gal, J Elo, G AF Bodis, Fruzsina Orosz, Gabor Toth, T. Jozsef Szabo, Marcell Elo, Gergely Laszlo Gal, Janos Elo, Gabor TI Percutaneous tracheostomy: Comparison of three different methods with respect to tracheal cartilage injury in cadavers —Randomized controlled study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE percutaneous tracheostomy; tracheal cartilage injury; Tracheal stenosis; Griggs; Ciaglia Blue Rhino ID percutaneous tracheostomy; tracheal cartilage injury; Tracheal stenosis; Griggs; Ciaglia Blue Rhino AB Background: Performing tracheostomy improves patient comfort and success rate of weaning from prolonged invasive mechanical ventilation. Data suggest that patients have more benefit of percutaneous technique than the surgical procedure, however, there is no consensus on the percutaneous method of choice regarding severe complications such as late tracheal stenosis. Aim of this study was comparing incidences of cartilage injury caused by different percutaneous dilatation techniques (PDT), including Single Dilator, Griggs’ and modified (bidirectional) Griggs’ method. Materials and methods: Randomized observational study was conducted on 150 cadavers underwent post-mortem percutaneous tracheostomy. Data of cadavers including age, gender and time elapsed from death until the intervention (more or less than 72 h) were collected and recorded. Primary and secondary outcomes were: rate of cartilage injury and cannula malposition respectively. Results: Statistical analysis revealed that method of intervention was significantly associated with occurrence of cartilage injury, as comparing either standard Griggs’ with Single Dilator (p = 0.002; OR: 4.903; 95% CI: 1.834–13.105) or modified Griggs’ with Single Dilator (p < 0.001; OR: 6.559; 95% CI: 2.472–17.404), however, no statistical difference was observed between standard and modified Griggs’ techniques (p = 0.583; OR: 0.748; 95% CI: 0.347–1.610). We found no statistical difference in the occurrence of cartilage injury between the early- and late post-mortem group (p = 0.630). Neither gender (p = 0.913), nor age (p = 0.529) influenced the rate of cartilage fracture. There was no statistical difference between the applied PDT techniques regarding the cannula misplacement/malposition. Conclusion: In this cadaver study both standard and modified Griggs’ forceps dilatational methods were safer than Single dilator in respect of cartilage injury. C1 [Bodis, Fruzsina] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Orosz, Gabor] Semmelweis University, Department of Anaesthesiology and Intensive TherapyBudapest, Hungary. [Toth, T. Jozsef] Semmelweis University, Department of Anaesthesiology and Intensive TherapyBudapest, Hungary. [Szabo, Marcell] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Elo, Gergely Laszlo] Semmelweis University, Department of Anaesthesiology and Intensive TherapyBudapest, Hungary. [Gal, Janos] Semmelweis University, Department of Anaesthesiology and Intensive TherapyBudapest, Hungary. [Elo, Gabor] Semmelweis University, Department of Anaesthesiology and Intensive TherapyBudapest, Hungary. RP Bodis, F (reprint author), Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Budapest, Hungary. EM bodis.fruzsina@med.semmelweis-univ.hu CR Armstrong PA, McCarthy MC, Peoples JB. Reduced use of resources by early tracheostomy in ventilator-dependent patients with blunt trauma. Surgery, 1998, 124(4):763–6., DOI 10.1067/msy.1998.91224 Brook AD, Sherman G, Malen J, Kollef MH. Early versus late tracheostomy in patients who require prolonged mechanical ventilation. Am J Crit Care, 2000, 9(5): 352–9., DOI 10.4037/ajcc2000.9.5.352 Kluger Y, Paul DB, Lucke J, Cox P, Colella JJ, Townsend RN, et al. Early tracheostomy in trauma patients. Eur J Emerg Med, 1996, 3(2):95–101., DOI 10.1097/ 00063110-199606000-00007 Rumbak MJ, Newton M, Truncale T, Schwartz SW, Adams JW, Hazard PB. A prospective, randomized, study comparing early percutaneous dilational tracheotomy to prolonged translaryngeal intubation, delayed tracheotomy, in critically ill medical patients. Crit Care Med, 2004, 32(8):1689–94., DOI 10.1097/ 01.ccm.0000134835.05161.b6 Ciaglia P, Firsching R, Syniec C. Elective percutaneous dilatational tracheostomy. A new simple bedside procedure; preliminary report. Chest, 1985, 87(6):715–9., DOI 10.1378/chest.87.6.715 Rashid AO, Islam S. Percutaneous tracheostomy: A comprehensive review. J Thorac Dis, 2017, 9(10):S1128–S38., DOI 10.21037/jtd.2017.09.33 Zgoda MA, Berger R. Balloon-facilitated percutaneous dilational tracheostomy tube placement: Preliminary report of a novel technique. Chest, 2005, 128(5): 3688–90., DOI 10.1378/chest.128.5.3688 Frova G, Quintel M. A new simple method for percutaneous tracheostomy: Controlled rotating dilation. A preliminary report. Intensive Care Med, 2002, 28(3): 299–303., DOI 10.1007/s00134-002-1218-5 Griggs WM, Worthley LI, Gilligan JE, Thomas PD, Myburg JA. A simple percutaneous tracheostomy technique. Surg Gynecol Obstet, 1990, 170(6): 543–5. Fantoni A, Ripamonti D. A non-derivative, non-surgical tracheostomy: The translaryngeal method. Intensive Care Med, 1997, 23(4):386–92., DOI 10.1007/ s001340050345 de Kleijn BJ, Wedman J, Zijlstra JG, Dikkers FG, van der Laan B. Short- and long-term complications of surgical and percutaneous dilatation tracheotomies: A large single-centre retrospective cohort study. Eur Arch Otorhinolaryngol, 2019, 276(6):1823–8., DOI 10.1007/s00405-019-05394-9 Epstein SK. Late complications of tracheostomy. Respir Care, 2005, 50(4): 542–9. Jung Kwon O, Young Suh G, Pyo Chung M, Kim J, Han J, Kim H. Tracheal stenosis depends on the extent of cartilaginous injury in experimental caninemodel. Exp Lung Res, 2003, 29(6):329–38., DOI 10.1080/01902140303757 Pookamala S, Thakar A, Puri K, Singh P, Kumar R, Sharma S. Acquired subglottic stenosis: Aetiological profile and treatment results. The J Laryngol Otology, 2014, 128(7):641–8., DOI 10.1017/S0022215114000966 Zouk AN, Batra H. Managing complications of percutaneous tracheostomy and gastrostomy. J Thorac Dis, 2021, 13(8):5314–30., DOI 10.21037/jtd-19-3716 Law R, Carney A, Manara A. Long-term outcome after percutaneous dilational tracheostomy: Endoscopic and spirometry findings. Anaesthesia, 1997, 52(1):51–6., DOI 10.1111/j.1365-2044.1997.013-az013.x Zias N, Chroneou A, TabbaMK, Gonzalez AV, Gray AW, Lamb CR, et al. Post tracheostomy and post intubation tracheal stenosis: Report of 31 cases and review of the literature. BMC Pulm Med, 2008, 8(1):18–9., DOI 10.1186/1471-2466-8-18 Ambesh SP. Principles and practice of percutaneous tracheostomy. New Delhi, India): Jaypee Brothers Publishers, 2010). van Heurn LW, Theunissen PH, Ramsay G, Brink PR. Pathologic changes of the trachea after percutaneous dilatational tracheotomy. Chest, 1996, 109(6): 1466–9., DOI 10.1378/chest.109.6.1466 Marchioni A, Tonelli R, Andreani A, Cappiello GF, Fermi M, Trentacosti F, et al. Molecular mechanisms and physiological changes behind benign tracheal and subglottic stenosis in adults. Int J Mol Sci, 2022, 23(5):2421., DOI 10.3390/ ijms23052421 Elo G, Zubek L, Hargitai Z, Ivanyi Z, Branovics J, Gal J. Prevention of tracheal cartilage injury with modified Griggs technique during percutaneous tracheostomy - randomized controlled cadaver study. Interv Med Appl Sci, 2012, 4(4):206–9., DOI 10.1556/IMAS.4.2012.4.5 Bloss RS,Ward RE. Survival after tracheoinnominate artery fistula. Am J Surg, 1980, 139(2):251–3., DOI 10.1016/0002-9610(80)90266-4 Khanafer A, Hellstern V, Meißner H, Harmening C, Schneider K, Henkes H. Tracheoinnominate fistula: Acute bleeding and hypovolemic shock due to a trachea-innominate artery fistula after long-term tracheostomy, treated with a stent-graft. CVIR Endovasc, 2021, 4(1):30., DOI 10.1186/s42155-021-00216-8 Zouk AN, Batra H. Managing complications of percutaneous tracheostomy and gastrostomy. J Thorac Dis, 2021, 13(8):5314–30., DOI 10.21037/jtd-19-3716 Fernandez-Bussy S, Mahajan B, Folch E, Caviedes I, Guerrero J, Majid A. Tracheostomy tube placement: Early and late complications. J Bronchology Interv Pulmonol, 2015, 22(4):357–64., DOI 10.1097/LBR.0000000000000177 Cipriano A, Mao ML, Hon HH, Vazquez D, Stawicki SP, Sharpe RP, et al. An overview of complications associated with open and percutaneous tracheostomy procedures. Int J Crit Illn Inj Sci, 2015, 5(3):179–88., DOI 10.4103/2229-5151.164994 Feller-Kopman D. Acute complications of artificial airways. Clin Chest Med, 2003, 24(3):445–55., DOI 10.1016/s0272-5231(03)00047-9 Watters M, Thorne G, Cox C, Monk C. Tracheal trauma from percutaneous tracheostomy using the Griggs method. Anaesthesia, 2002, 57(3):249–52., DOI 10. 1046/j.0003-2409.2001.02452.x Paran H, Butnaru G, Hass I, Afanasyv A, GutmanM. Evaluation of a modified percutaneous tracheostomy technique without bronchoscopic guidance. Chest, 2004, 126(3):868–71., DOI 10.1378/chest.126.3.868 Ambesh SP, Pandey CK, Srivastava S, Agarwal A, Singh DK. Percutaneous tracheostomy with single dilatation technique: A prospective, randomized comparison of Ciaglia blue rhino versus Griggs’ guidewire dilating forceps. Anesth Analg, 2002, 95(6):1739–45. t., DOI 10.1097/00000539-200212000-00050 Tai HP, Lee DL, Chen CF, Huang YT. The effect of tracheostomy delay time on outcome of patients with prolonged mechanical ventilation: A STROBEcompliant retrospective cohort study. Medicine, Baltimore,, 2019, 98(35): e16939., DOI 10.1097/MD.0000000000016939 Pauliny M, Christova E, Mackova J, Liska M. Percutaneous dilation tracheostomy versus surgical tracheostomy in critically ill patients. Bratisl Lek Listy, 2012, 113(7):409–11., DOI 10.4149/bll_2012_092 Khaja M, Haider A, Alapati A, Qureshi ZA, Yapor L. Percutaneous tracheostomy: A bedside procedure. Cureus, 2022, 14(4):e24083., DOI 10.7759/ cureus.24083 Cheung NH, Napolitano LM. Tracheostomy: Epidemiology, indications, timing, technique, and outcomes. Respir Care, 2014, 59(6):895–915., DOI 10. 4187/respcare.02971 Heikkinen M, Aarnio P, Hannukainen J. Percutaneous dilational tracheostomy or conventional surgical tracheostomy? Crit Care Med, 2000, 28(5):1399–402., DOI 10.1097/00003246-200005000-00023 Kaylie DM, Andersen PE, Wax MK. An analysis of time and staff utilization for open versus percutaneous tracheostomies. Otolaryngol Head Neck Surg, 2003, 128(1):109–14., DOI 10.1067/mhn.2003.18 Melloni G,Muttini S, Gallioli G, Carretta A, Cozzi S, Gemma M, et al. Surgical tracheostomy versus percutaneous dilatational tracheostomy. A prospectiverandomized study with long-term follow-up. J Cardiovasc Surg, Torino,, 2002, 43(1):113–21. Youssef TF, Ahmed MR, Saber A. Percutaneous dilatational versus conventional surgical tracheostomy in intensive care patients. N Am J Med Sci, 2011, 3(11):508–12., DOI 10.4297/najms.2011.3508 Brass P, Hellmich M, Ladra A, Ladra J, Wrzosek A. Percutaneous techniques versus surgical techniques for tracheostomy. Cochrane Database Syst Rev, 2016, 7(7):Cd008045., DOI 10.1002/14651858.CD008045.pub2 Delaney A, Bagshaw SM, Nalos M. Percutaneous dilatational tracheostomy versus surgical tracheostomy in critically ill patients: A systematic review and metaanalysis. Crit Care, 2006, 10(2):R55., DOI 10.1186/cc4887 Dennis BM, Eckert MJ, Gunter OL, Morris JA, May AK. Safety of bedside percutaneous tracheostomy in the critically ill: Evaluation of more than 3, 000 procedures. J Am Coll Surg, 2013, 216(4):858–65., DOI 10.1016/j.jamcollsurg. 2012.12.017 Cinotti R, Voicu S, Jaber S, Chousterman B, Paugam-Burtz C,Oueslati H, et al. Tracheostomy and long-term mortality in ICU patients undergoing prolonged mechanical ventilation. PLoS One, 2019, 14(10):e0220399., DOI 10.1371/journal. pone.0220399 Johnson-Obaseki S, Veljkovic A, Javidnia H. Complication rates of open surgical versus percutaneous tracheostomy in critically ill patients. Laryngoscope, 2016, 126(11):2459–67., DOI 10.1002/lary.26019 Anon JM, Escuela MP, Gomez V, Moreno A, Lopez J, Diaz R, et al. Percutaneous tracheostomy: Ciaglia blue rhino versus Griggs’ guide wire dilating forceps. A prospective randomized trial. Acta Anaesthesiol Scand, 2004, 48(4):451–6., DOI 10.1111/j.1399-6576.2004.0313.x Fikkers BG, StaatsenM, van den Hoogen FJ, van der Hoeven JG. Early and late outcome after single step dilatational tracheostomy versus the guide wire dilating forceps technique: A prospective randomized clinical trial. Intensive Care Med, 2011, 37(7):1103–9., DOI 10.1007/s00134-011-2222-4 Cabrini L, Landoni G, Greco M, Costagliola R, Monti G, Colombo S, et al. Single dilator vs. guide wire dilating forceps tracheostomy: A meta-analysis of randomised trials. Acta Anaesthesiol Scand, 2014, 58(2):135–42., DOI 10.1111/aas. 12213 Fikkers BG, Staatsen M, Lardenoije SG, van den Hoogen FJ, van der Hoeven JG. Comparison of two percutaneous tracheostomy techniques, guide wire dilating forceps and Ciaglia blue rhino: A sequential cohort study. Crit Care, 2004, 8(5): R299–305., DOI 10.1186/cc2907 Norwood S, Vallina VL, Short K, Saigusa M, Fernandez LG, McLarty JW. Incidence of tracheal stenosis and other late complications after percutaneous tracheostomy. Ann Surg, 2000, 232(2):233–41., DOI 10.1097/00000658-200008000-00014 Steele AP, Evans HW, Afaq MA, Robson JM, Dourado J, Tayar R, et al. Long-term follow-up of Griggs percutaneous tracheostomy with spiral CT and questionnaire. Chest, 2000, 117(5):1430–3., DOI 10.1378/chest.117.5. 1430 Ronen O, Rosin I, Taitelman UZ, Altman E. Comparison of Ciaglia and Griggs percutaneous tracheostomy techniques - a biomechanical animal study. Indian J Crit Care Med, 2019, 23(6):247–50., DOI 10.5005/jp-journals-10071- 23174 Karagiannidis C, Merten ML, Heunks L, Strassmann SE, Schafer S, Magnet F, et al. Respiratory acidosis during bronchoscopy-guided percutaneous dilatational tracheostomy: Impact of ventilator settings and endotracheal tube size. BMC Anesthesiol, 2019, 19(1):147., DOI 10.1186/s12871-019-0824-5 Hotchkiss KS, McCaffrey JC. Laryngotracheal injury after percutaneous dilational tracheostomy in cadaver specimens. Laryngoscope, 2003, 113(1): 16–20., DOI 10.1097/00005537-200301000-00003 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610934 EP 1610941 DI 10.3389/pore.2023.1610934 PG 8 ER PT J AU Rubovszky, G Kocsis, J Boer, K Chilingirova, N Dank, M Kahan, Zs Kaidarova, D Kover, E Vertakova Krakovska, B Mahr, K Mrinakova, B Piko, B Bozovic-Spasojevic, I Horvath, Zs AF Rubovszky, Gabor Kocsis, Judit Boer, Katalin Chilingirova, Nataliya Dank, Magdolna Kahan, Zsuzsanna Kaidarova, Dilyara Kover, Erika Vertakova Krakovska, Bibiana Mahr, Karoly Mrinakova, Bela Piko, Bela Bozovic-Spasojevic, Ivana Horvath, Zsolt TI Corrigendum: Systemic treatment of breast cancer. 1st Central-Eastern European professional Consensus Statement on breast cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE early breast cancer; locally advanced breast cancer; adjuvant treatment; neoadjuvant treatment; metastatic breast cancer; inflammatory breast cancer; guideline ID early breast cancer; locally advanced breast cancer; adjuvant treatment; neoadjuvant treatment; metastatic breast cancer; inflammatory breast cancer; guideline AB A Corrigendum on Systemic treatment of breast cancer. 1st Central-Eastern European professional consensus statement on breast cancer by Rubovszky G, Kocsis J, Boer K, Chilingirova N, Dank M, Kahan Z, Kaidarova D, Kover E, Krakovska BV, Mahr K, Mrinakova B, Piko B, Bozovic-Spasojevic I and Horvath Z (2022). Pathol. Oncol. Res. 28:1610383. doi: 10.3389/pore.2022.1610383 C1 [Rubovszky, Gabor] Orszagos Onkologiai Intezet, „B” Belgyogyaszati-Onkologiai es Klinikai Farmakologiai OsztalyBudapest, Hungary. [Kocsis, Judit] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. [Boer, Katalin] St. Margit Hospital, Clinical Oncology DepartmentBudapest, Hungary. [Chilingirova, Nataliya] Medical University-Pleven, Heart and Brain Hospital, Science and Research Institute, Clinic Center of ExcellencePleven, Bulgaria. [Dank, Magdolna] Semmelweis University, Department of OncologyBudapest, Hungary. [Kahan, Zsuzsanna] University of Szeged, Department of OncotherapySzeged, Hungary. [Kaidarova, Dilyara] Kazakh Institute of Oncology and RadiologyAlmaty, Kazakhstan. [Kover, Erika] University of Pecs, Oncotherapy InstitutePecs, Hungary. [Vertakova Krakovska, Bibiana] Comenius University, Faculty of Medicine, 1st Department of OncologyBratislava, Slovakia. [Mahr, Karoly] Zala Megyei Szent Rafael Korhaz, OnkologiaZalaegerszeg, Hungary. [Mrinakova, Bela] Comenius University, Faculty of Medicine, 1st Department of OncologyBratislava, Slovakia. [Piko, Bela] Bekes Megyei Kozponti Korhaz, Pandy Kalman Tagkorhaz, Megyei Onkologiai CentrumGyula, Hungary. [Bozovic-Spasojevic, Ivana] Daily Chemotherapy HospitalBelgrade, Serbia. [Horvath, Zsolt] Bacs-Kiskun County Hospital, Department of OncoradiologyKecskemet, Hungary. RP Rubovszky, G (reprint author), Orszagos Onkologiai Intezet, „B” Belgyogyaszati-Onkologiai es Klinikai Farmakologiai Osztaly, Budapest, Hungary. EM rubovszkyg@gmail.com NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610954 EP 1610955 DI 10.3389/pore.2023.1610954 PG 2 ER PT J AU Yang, Y Wang, Z He, M Diao, L Yu, B Li, D AF Yang, Yuting Wang, Ze He, Mengqi Diao, Lihong Yu, Biyue Li, Dong TI NAD+ biosynthesis metabolism predicts prognosis and indicates immune microenvironment for breast cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunotherapy; breast cancer; prognosis; immune microenvironment; NAD+ biosynthesis ID immunotherapy; breast cancer; prognosis; immune microenvironment; NAD+ biosynthesis AB The growing evidence implies that tumor cells need to increase NAD+ levels by upregulating NAD+ biosynthesis to satisfy their growth demand. NAD+ biosynthesis metabolism is implicated in tumor progression. Breast cancer (BC) is the most common malignant malignancy in the world. Nevertheless, the prognostic significance of NAD+ biosynthesis and its relationship with the tumor immune microenvironment in breast cancer still need further investigation. In this study, we obtained the mRNA expression data and clinical information of BC samples from public databases and calculated the level of NAD+ biosynthesis activity by single-sample gene set enrichment analysis (ssGSEA). We then explored the relationship between the NAD+ biosynthesis score, infiltrating immune cells, prognosis significance, immunogenicity and immune checkpoint molecules. The results demonstrated that patients with high NAD+ biosynthetic score displayed poor prognosis, high immune infiltration, high immunogenicity, elevated PDL1 expression, and might more benefit from immunotherapy. Taken together, our studies not only deepened the understanding of NAD+ biosynthesis metabolism of breast cancer but also provided new insights into personalized treatment strategies and immunological therapies to improve the outcomes of breast cancer patients. C1 [Yang, Yuting] Medical College of Qingdao University, Department of ImmunologyQingdao, Shandong, China. [Wang, Ze] Beijing Institute of Lifeomics, National Center for Protein Sciences, State Key Laboratory of Proteomics, Beijing Proteome Research CenterBeijing, China. [He, Mengqi] Beijing Institute of Lifeomics, National Center for Protein Sciences, State Key Laboratory of Proteomics, Beijing Proteome Research CenterBeijing, China. [Diao, Lihong] Beijing University of Chinese Medicine, School of Traditional Chinese MedicineBeijing, China. [Yu, Biyue] Hebei University, School of Life SciencesBaoding, Hebei, China. [Li, Dong] Medical College of Qingdao University, Department of ImmunologyQingdao, Shandong, China. RP Li, D (reprint author), Medical College of Qingdao University, Department of Immunology, Qingdao, China. EM lidong.bprc@foxmail.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 McDonald ES, Clark AS, Tchou J, Zhang P, Freedman GM. Clinical diagnosis and management of breast cancer. J Nucl Med Off Publ Soc Nucl Med, 2016, 57(1): 9S–16S., DOI 10.2967/jnumed.115.157834 Januskevicienė I, Petrikaitė V. Heterogeneity of breast cancer: The importance of interaction between different tumor cell populations. Life Sci, 2019, 239:117009., DOI 10.1016/j.lfs.2019.117009 Catalano V, Turdo A, Di Franco S, Dieli F, Todaro M, Stassi G. Tumor and its microenvironment: A synergistic interplay. Semin Cancer Biol, 2013, 23:522–32., DOI 10.1016/j.semcancer.2013.08.007 Fu T, Dai L-J, Wu S-Y, Xiao Y, Ma D, Jiang YZ, et al. Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response. J Hematol Oncolj Hematol Oncol, 2021, 14:98., DOI 10.1186/s13045- 021-01103-4 Wang S, Zhang Q, Yu C, Cao Y, Zuo Y, Yang L. Immune cell infiltration-based signature for prognosis and immunogenomic analysis in breast cancer. Brief Bioinform, 2021, 22:2020–31., DOI 10.1093/bib/bbaa026 Oshi M, Asaoka M, Tokumaru Y, Yan L, Matsuyama R, Ishikawa T, et al. CD8 T cell score as a prognostic biomarker for triple negative breast cancer. Int J Mol Sci, 2020, 21:E6968., DOI 10.3390/ijms21186968 Gao W, Wen H, Liang L, Dong X, Du R, Zhou W, et al. IL20RA signaling enhances stemness and promotes the formation of an immunosuppressive microenvironment in breast cancer. Theranostics, 2021, 11:2564–80., DOI 10. 7150/thno.45280 Savas P, Salgado R, Denkert C, Sotiriou C, Darcy PK, Smyth MJ, et al. Clinical relevance of host immunity in breast cancer: From TILs to the clinic. Nat Rev Clin Oncol, 2016, 13:228–41., DOI 10.1038/nrclinonc.2015.215 Dieci MV, Miglietta F, Guarneri V. Immune infiltrates in breast cancer: Recent updates and clinical implications. Cells, 2021, 10:223., DOI 10.3390/ cells10020223 Covarrubias AJ, Perrone R, Grozio A, Verdin E. NAD+ metabolism and its roles in cellular processes during ageing. Nat Rev Mol Cel Biol, 2021, 22:119–41., DOI 10.1038/s41580-020-00313-x Navas LE, Carnero A. NAD+ metabolism, stemness, the immune response, and cancer. Signal Transduct Target Ther, 2021, 6:2., DOI 10.1038/s41392-020- 00354-w Chiarugi A, Dolle C, Felici R, Ziegler M. The NAD metabolome--a key determinant of cancer cell biology. Nat Rev Cancer, 2012, 12:741–52., DOI 10. 1038/nrc3340 Ghanem MS, Monacelli F, Nencioni A. Advances in NAD-lowering agents for cancer treatment. Nutrients, 2021, 13:1665., DOI 10.3390/ nu13051665 Zhang H, Zhang N, Liu Y, Su P, Liang Y, Li Y, et al. Epigenetic regulation of NAMPT by NAMPT-AS drives metastatic progression in triple-negative breast cancer. Cancer Res, 2019, 79:3347–59., DOI 10.1158/0008-5472.CAN-18- 3418 Gholinejad Z, Kheiripour N, Nourbakhsh M, Ilbeigi D, Behroozfar K, Hesari Z, et al. Extracellular NAMPT/Visfatin induces proliferation through ERK1/2 and AKT and inhibits apoptosis in breast cancer cells. Peptides, 2017, 92:9–15., DOI 10. 1016/j.peptides.2017.04.007 Li C, Zhu Y, Chen W, Li M, Yang M, Shen Z, et al. Circulating NAD+ metabolism-derived genes unveils prognostic and peripheral immune infiltration in amyotrophic lateral sclerosis. Front Cel Dev Biol, 2022, 10:831273., DOI 10.3389/ fcell.2022.831273 Dedeurwaerder S, Desmedt C, Calonne E, Singhal SK, Haibe-Kains B, Defrance M, et al. DNA methylation profiling reveals a predominant immune component in breast cancers. EMBO Mol Med, 2011, 3:726–41., DOI 10.1002/ emmm.201100801 Huang C-C, Tu S-H, Lien H-H, Jeng JY, Huang CS, Huang CJ, et al. Concurrent gene signatures for han Chinese breast cancers. PLoS One, 2013, 8: e76421., DOI 10.1371/journal.pone.0076421 Metzger-Filho O, Michiels S, Bertucci F, CatteAu A, Salgado R, Galant C, et al. Genomic grade adds prognostic value in invasive lobular carcinoma. Ann Oncol Off J Eur Soc Med Oncol, 2013, 24:377–84., DOI 10.1093/annonc/ mds280 Gautier L, Cope L, Bolstad BM, Irizarry RA. affy--analysis of Affymetrix GeneChip data at the probe level. Bioinforma Oxf Engl, 2004, 20:307–15., DOI 10. 1093/bioinformatics/btg405 Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostat Oxf Engl, 2007, 8: 118–27., DOI 10.1093/biostatistics/kxj037 Hanzelmann S, Castelo R, Guinney J. Gsva: Gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics, 2013, 14:7., DOI 10.1186/1471- 2105-14-7 Yu G,Wang L-G, Han Y, He Q-Y. clusterProfiler: an R Package for comparing biological themes among gene clusters. OMICS J Integr Biol, 2012, 16:284–7., DOI 10. 1089/omi.2011.0118 Charoentong P, Finotello F, Angelova M, Mayer C, Efremova M, Rieder D, et al. Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep, 2017, 18: 248–62., DOI 10.1016/j.celrep.2016.12.019 Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity, 2013, 39:782–95., DOI 10.1016/j.immuni. 2013.10.003 Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, Kim H, Torres- Garcia W, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun, 2013, 4:2612., DOI 10.1038/ ncomms3612 Mayakonda A, Lin D-C, Assenov Y, Plass C, Koeffler HP. Maftools: Efficient and comprehensive analysis of somatic variants in cancer. Genome Res, 2018, 28: 1747–56., DOI 10.1101/gr.239244.118 Shi Z, Shen J, Qiu J, Zhao Q, Hua K, Wang H. CXCL10 potentiates immune checkpoint blockade therapy in homologous recombination-deficient tumors. Theranostics, 2021, 11:7175–87., DOI 10.7150/thno.59056 Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity, 2018, 48:812–30.e14., DOI 10.1016/j. immuni.2018.03.023 Geeleher P, Cox N, Huang RS. pRRophetic: an R package for prediction of clinical chemotherapeutic response from tumor gene expression levels. PLoS One, 2014, 9:e107468., DOI 10.1371/journal.pone.0107468 Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates cancer progression. Cancer Res, 2019, 79:4557–66., DOI 10.1158/0008-5472.CAN- 18-3962 Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: A leading role for STAT3. Nat Rev Cancer, 2009, 9:798–809., DOI 10.1038/nrc2734 Marigo I, Bosio E, Solito S, Mesa C, Fernandez A, Dolcetti L, et al. Tumorinduced tolerance and immune suppression depend on the C/EBPbeta transcription factor. Immunity, 2010, 32:790–802., DOI 10.1016/j.immuni.2010.05.010 Huang Q, Liu J, Wu S, Zhang X, Xiao Z, Liu Z, et al. Spi-B promotes the recruitment of tumor-associated macrophages via enhancing CCL4 expression in lung cancer. Front Oncol, 2021, 11:659131., DOI 10. 3389/fonc.2021.659131 Zhao Z, Zheng L, ChenW,Weng W, Song J, Ji J. Delivery strategies of cancer immunotherapy: Recent advances and future perspectives. J Hematol Oncolj Hematol Oncol, 2019, 12:126., DOI 10.1186/s13045-019-0817-3 Liu C-L, Cheng S-P, Chen M-J, Lin CH, Chen SN, Kuo YH, et al. Quinolinate phosphoribosyltransferase promotes invasiveness of breast cancer through myosin light chain phosphorylation. Front Endocrinol, 2020, 11:621944., DOI 10.3389/fendo. 2020.621944 Hammerl D, Smid M, Timmermans AM, Sleijfer S, Martens JWM, Debets R. Breast cancer genomics and immuno-oncological markers to guide immune therapies. Semin Cancer Biol, 2018, 52:178–88., DOI 10.1016/j.semcancer.2017. 11.003 Gabrilovich DI.Myeloid-derived suppressor cells. Cancer Immunol Res, 2017, 5:3–8., DOI 10.1158/2326-6066.CIR-16-0297 Lin C-F, Lin C-M, Lee K-Y, Wu SY, Feng PH, Chen KY, et al. Escape from IFN-γ-dependent immunosurveillance in tumorigenesis. J Biomed Sci, 2017, 24:10., DOI 10.1186/s12929-017-0317-0 Mojic M, Takeda K, Hayakawa Y. The dark side of IFN-γ: Its role in promoting cancer immunoevasion. Int J Mol Sci, 2017, 19:89., DOI 10.3390/ ijms19010089 Lv H, Lv G, Chen C, Zong Q, Jiang G, Ye D, et al. NAD+ metabolism maintains inducible PD-L1 expression to drive tumor immune evasion. Cell Metab, 2021, 33:110–27.e5., DOI 10.1016/j.cmet.2020.10.021 Guler R, Mpotje T, Ozturk M, Nono JK, Parihar SP, Chia JE, et al. Batf2 differentially regulates tissue immunopathology in Type 1 and Type 2 diseases. Mucosal Immunol, 2019, 12:390–402., DOI 10.1038/s41385-018- 0108-2 Roy S, Guler R, Parihar SP, Schmeier S, Kaczkowski B, Nishimura H, et al. Batf2/Irf1 induces inflammatory responses in classically activated macrophages, lipopolysaccharides, and mycobacterial infection. J Immunol Baltim, 2015, 194: 6035–44., DOI 10.4049/jimmunol.1402521 Garrett-Sinha LA, Su GH, Rao S, Kabak S, Hao Z, Clark MR, et al. PU.1 and Spi-B are required for normal B cell receptor-mediated signal transduction. Immunity, 1999, 10:399–408., DOI 10.1016/s1074-7613(00)80040-0 Schotte R, Nagasawa M, Weijer K, Spits H, Blom B. The ETS transcription factor Spi-B is required for human plasmacytoid dendritic cell development. J Exp Med, 2004, 200:1503–9., DOI 10.1084/jem.20041231 Wang J, Wang X, Guo Y, Ye L, Li D, Hu A, et al. Therapeutic targeting of SPIB/SPI1-facilitated interplay of cancer cells and neutrophils inhibits aerobic glycolysis and cancer progression. Clin Transl Med, 2021, 11:e588., DOI 10.1002/ ctm2.588 Wang S, He Z, Wang X, Li H, Liu XS. Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction. eLife, 2019, 8: e49020., DOI 10.7554/eLife.49020 Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of tumor mutation burden as an immunotherapy biomarker: Utility for the oncology clinic. Ann Oncol Off J Eur Soc Med Oncol, 2019, 30:44–56., DOI 10. 1093/annonc/mdy495 Barroso-Sousa R, Jain E, Cohen O, Kim D, Buendia-Buendia J, WinEr E, et al. Prevalence and mutational determinants of high tumor mutation burden in breast cancer. Ann Oncol Off J Eur Soc Med Oncol, 2020, 31:387–94., DOI 10.1016/j.annonc. 2019.11.010 Budczies J, Kluck K, Beck S, Ourailidis I, Allgauer M, Menzel M, et al. Homologous recombination deficiency is inversely correlated with microsatellite instability and identifies immunologically cold tumors in most cancer types. J Pathol Clin Res, 2022, 8:371–82., DOI 10.1002/cjp2.271 Pellegrino B, Musolino A, Llop-Guevara A, Serra V, De Silva P, Hlavata Z, et al. Homologous recombination repair deficiency and the immune response in breast cancer: A literature review. Transl Oncol, 2020, 13:410–22., DOI 10.1016/j. tranon.2019.10.010 Solinas C, Marcoux D, Garaud S, Vitoria JR, Van den Eynden G, de Wind A, et al. BRCA gene mutations do not shape the extent and organization of tumor infiltrating lymphocytes in triple negative breast cancer. Cancer Lett, 2019, 450: 88–97., DOI 10.1016/j.canlet.2019.02.027 Madel M-B, Ibanez L, Wakkach A, de Vries TJ, Teti A, Apparailly F, et al. Immune function and diversity of osteoclasts in normal and pathological conditions. Front Immunol, 2019, 10:1408., DOI 10.3389/fimmu.2019.01408 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610956 EP 1610967 DI 10.3389/pore.2023.1610956 PG 12 ER PT J AU Chen, H Shi, X Ren, L Wan, Y Zhuo, H Zeng, L SangDan, W Wang, F AF Chen, HongMin Shi, XiaoXiao Ren, Li Wan, YuMing Zhuo, HongYu Zeng, Li SangDan, WangMu Wang, Feng TI Screening of core genes and prediction of ceRNA regulation mechanism of circRNAs in nasopharyngeal carcinoma by bioinformatics analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TCGA; nasopharyngeal carcinoma; weighted gene co-expression network analysis; GEO database; fibronectin 1 ID TCGA; nasopharyngeal carcinoma; weighted gene co-expression network analysis; GEO database; fibronectin 1 AB Background: Nasopharyngeal carcinoma (NPC) represents a highly aggressive malignant tumor. Competing endogenous RNAs (ceRNA) regulation is a common regulatory mechanism in tumors. The ceRNA network links the functions between mRNAs and ncRNAs, thus playing an important regulatory role in diseases. This study screened the potential key genes in NPC and predicted regulatory mechanisms using bioinformatics analysis. Methods: The merged microarray data of three NPC-related mRNA expression microarrays from the Gene Expression Omnibus (GEO) database and the expression data of tumor samples or normal samples from the nasopharynx and tonsil in The Cancer Genome Atlas (TCGA) database were both subjected to differential analysis and Weighted Gene Co-expression Network Analysis (WGCNA). The results from two different databases were intersected with WGCNA results to obtain potential regulatory genes in NPC, followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses. The hub-gene in candidate genes was discerned through Protein-Protein Interaction (PPI) analysis and its upstream regulatory mechanism was predicted by miRwalk and circbank databases. Results: Totally 68 upregulated genes and 96 downregulated genes in NPC were screened through GEO and TCGA. According to WGCNA, the NPCrelated modules were screened from GEO and TCGA analysis results, and the genes in the modules were obtained. After the results of differential analysis and WGCNA were intersected, 74 differentially expressed candidate genes associated with NPC were discerned. Finally, fibronectin 1 (FN1) was identified as a hub-gene in NPC. Prediction of upstream regulatory mechanisms of FN1 suggested that FN1 may be regulated by ceRNA mechanisms involving multiple circRNAs, thereby influencing NPC progression through ceRNA regulation. Conclusion: FN1 is identified as a key regulator in NPC development and is likely to be regulated by numerous circRNA-mediated ceRNA mechanisms. C1 [Chen, HongMin] Medical School, Sichuan University, West China Hospital, Cancer Center, Department of Medical OncologySichuan, China. [Shi, XiaoXiao] Sichuan University, West China Hospital, Chengdu Shangjin Nanfu Hospital, Department of Medical OncologyChengdu, China. [Ren, Li] Sichuan University, West China Hospital, Cancer Center, Department of Thoracic OncologyChengdu, China. [Wan, YuMing] Medical School, Sichuan University, West China Hospital, Cancer Center, Department of Medical OncologySichuan, China. [Zhuo, HongYu] Medical School, Sichuan University, West China Hospital, Cancer Center, Department of Medical OncologySichuan, China. [Zeng, Li] Medical School, Sichuan University, West China Hospital, Cancer Center, Department of Medical OncologySichuan, China. [SangDan, WangMu] People’s Hospital of Tibet Autonomous Region, Department of OncologyLhasa, China. [Wang, Feng] Medical School, Sichuan University, West China Hospital, Cancer Center, Department of Medical OncologySichuan, China. RP Wang, F (reprint author), Medical School, Sichuan University, West China Hospital, Cancer Center, Department of Medical Oncology, Sichuan, China. EM wangfeng5024@126.com CR Chen YP, Chan ATC, Le QT, Blanchard P, Sun Y, Ma J. Nasopharyngeal carcinoma. Lancet, 2019, 394(10192):64–80., DOI 10.1016/S0140-6736(19)30956-0 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Wu L, Li C, Pan L. Nasopharyngeal carcinoma: A review of current updates. Exp Ther Med, 2018, 15(4):3687–92., DOI 10.3892/etm.2018.5878 Engku Nur Syafirah EAR, Irekeola AA, Yean Yean C. Diagnostic and prognostic indications of nasopharyngeal carcinoma. Diagnostics, Basel,, 2020, 10(9):611., DOI 10.3390/diagnostics10090611 Bruce JP, Yip K, Bratman SV, Ito E, Liu FF. Nasopharyngeal cancer: Molecular landscape. J Clin Oncol, 2015, 33(29):3346–55., DOI 10.1200/JCO.2015.60.7846 Tang J, Aittokallio T. Network pharmacology strategies toward multi-target anticancer therapies: From computational models to experimental design principles. Curr Pharm Des, 2014, 20(1):23–36., DOI 10.2174/13816128113199990470 Lee KT, Tan JK, Lam AK, Gan SY. MicroRNAs serving as potential biomarkers and therapeutic targets in nasopharyngeal carcinoma: A critical review. Crit Rev Oncol Hematol, 2016, 103:1–9., DOI 10.1016/j.critrevonc.2016.04.006 Wang H, Wang W, Fan S. Emerging roles of lncRNA in Nasopharyngeal Carcinoma and therapeutic opportunities. Int J Biol Sci, 2022, 18(7):2714–28., DOI 10.7150/ijbs.70292 Patop IL, Kadener S. circRNAs in Cancer. Curr Opin Genet Dev, 2018, 48: 121–7., DOI 10.1016/j.gde.2017.11.007 Yang M, Huang W. Circular RNAs in nasopharyngeal carcinoma. Clin Chim Acta, 2020, 508:240–8., DOI 10.1016/j.cca.2020.05.029 Chen N, Zhao G, Yan X, Lv Z, Yin H, Zhang S, et al. A novel FLI1 exonic circular RNA promotes metastasis in breast cancer by coordinately regulating TET1 and DNMT1. Genome Biol, 2018, 19(1):218., DOI 10.1186/s13059-018-1594-y Greene JM, Larin Z, Taylor IC, Prentice H, Gwinn KA, Kingston RE. Multiple basal elements of a human hsp70 promoter function differently in human and rodent cell lines. Mol Cel Biol, 1987, 7(10):3646–55., DOI 10.1128/mcb.7.10.3646 Ye Z,Wang F, Yan F, Wang L, Li B, Liu T, et al. Bioinformatic identification of candidate biomarkers and related transcription factors in nasopharyngeal carcinoma. World J Surg Oncol, 2019, 17(1):60., DOI 10.1186/s12957-019-1605-9 Zhu D, Shao M, Yang J, Fang M, Liu S, Lou D, et al. Curcumin enhances radiosensitization of nasopharyngeal carcinoma via mediating regulation of tumor stemlike cells by a CircRNA network. J Cancer, 2020, 11(8):2360–70., DOI 10.7150/jca.39511 Zhang Z, Huang J,WangG, Jin F, Zheng J,XiaoH, et al. SerummiRNAs, a potential prognosis marker of loco-regionally advanced nasopharyngeal carcinoma patients treated with CCRT. BMC Cancer, 2020, 20(1):183., DOI 10.1186/s12885-020-6689-7 Bao YN, Cao X, Luo DH, Sun R, Peng LX, Wang L, et al. Urokinase-type plasminogen activator receptor signaling is critical in nasopharyngeal carcinoma cell growth and metastasis. Cell Cycle, 2014, 13(12):1958–69., DOI 10.4161/cc.28921 Bose S, Yap LF, Fung M, Starzcynski J, Saleh A, Morgan S, et al. The ATM tumour suppressor gene is down-regulated in EBV-associated nasopharyngeal carcinoma. J Pathol, 2009, 217(3):345–52., DOI 10.1002/path.2487 Hu C,Wei W, Chen X,Woodman CB, Yao Y, Nicholls JM, et al. A global view of the oncogenic landscape in nasopharyngeal carcinoma: An integrated analysis at the genetic and expression levels. PLoS One, 2012, 7(7):e41055., DOI 10.1371/ journal.pone.0041055 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res, 2015, 43(7):e47., DOI 10.1093/nar/gkv007 Leek JT, Storey JD. Capturing heterogeneity in gene expression studies by surrogate variable analysis. Plos Genet, 2007, 3(9):1724–35., DOI 10.1371/journal.pgen.0030161 Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics, 2012, 28(6):882–3., DOI 10.1093/bioinformatics/bts034 Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics, 2010, 26(1):139–40., DOI 10.1093/bioinformatics/btp616 McCarthy DJ, Chen Y, Smyth GK. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res, 2012, 40(10):4288–97., DOI 10.1093/nar/gks042 Chen Y, Lun AT, Smyth GK. From reads to genes to pathways: Differential expression analysis of RNA-seq experiments using rsubread and the edgeR quasilikelihood pipeline. F1000Res, 2016, 5:1438., DOI 10.12688/f1000research.8987.1 Langfelder P, Horvath S. Wgcna: an R package for weighted correlation network analysis. BMC Bioinformatics, 2008, 9:559., DOI 10.1186/1471-2105-9-559 Zhu Q, ZhangQ, GuM, Zhang K, Xia T, Zhang S, et al.MIR106A-5p upregulation suppresses autophagy and accelerates malignant phenotype in nasopharyngeal carcinoma. Autophagy, 2021, 17(7):1667–83., DOI 10.1080/15548627.2020.1781368 LeeHM,Okuda KS,Gonzalez FE, PatelV. Current perspectives on nasopharyngeal carcinoma. Adv Exp Med Biol, 2019, 1164:11–34., DOI 10.1007/978-3-030-22254-3_2 Petersson F. Nasopharyngeal carcinoma: A review. Semin Diagn Pathol, 2015, 32(1):54–73., DOI 10.1053/j.semdp.2015.02.021 Tsao SW, Tsang CM, Lo KW. Epstein-Barr virus infection and nasopharyngeal carcinoma. Philos Trans R Soc Lond B Biol Sci, 2017, 372(1732):20160270., DOI 10.1098/rstb.2016.0270 Han B, Yang X, Zhang P, Zhang Y, Tu Y, He Z, et al. DNA methylation biomarkers for nasopharyngeal carcinoma. PLoS One, 2020, 15(4):e0230524., DOI 10.1371/journal.pone.0230524 LiG, Zhang J, Liu D,Wei Q,Wang H, Lv Y, et al. Identification of hub genes and potential ceRNA networks of diabetic nephropathy by weighted gene Co-expression network analysis. Front Genet, 2021, 12:767654., DOI 10.3389/fgene.2021.767654 Tian Z, He W, Tang J, Liao X, Yang Q, Wu Y, et al. Identification of important modules and biomarkers in breast cancer based on WGCNA. Onco Targets Ther, 2020, 13:6805–17., DOI 10.2147/OTT.S258439 Wen JY, Qin LT, Chen G,Huang HQ, Shen MJ, Pang JS, et al. Downregulation of MicroRNA-1 and its potential molecular mechanism in nasopharyngeal cancer: An investigation combined with in silico and in-house immunohistochemistry validation. Dis Markers, 2022, 2022:7962220., DOI 10.1155/2022/7962220 Chu Q, Huang H, Huang T, Cao L, Peng L, Shi S, et al. Extracellular serglycin upregulates the CD44 receptor in an autocrine manner to maintain self-renewal in nasopharyngeal carcinoma cells by reciprocally activating the MAPK/β-catenin axis. Cell Death Dis, 2016, 7(11):e2456., DOI 10.1038/cddis.2016.287 Verma N, Patel S, Osborn V, McBride S, Riaz N, Lee A, et al. Prognostic significance of human papillomavirus and Epstein-Bar virus in nasopharyngeal carcinoma. Head Neck, 2020, 42(9):2364–74., DOI 10.1002/hed.26245 Huang SH, Jacinto JCK, O’Sullivan B, Su J, Kim J, Ringash J, et al. Clinical presentation and outcome of human papillomavirus-positive nasopharyngeal carcinoma in a North American cohort. Cancer, 2022, 128(15):2908–21., DOI 10.1002/cncr.34266 Liu Y, Wang C, Shan X, Wu J, Liu H, Liu H, et al. S100P is associated with proliferation and migration in nasopharyngeal carcinoma. Oncol Lett, 2017, 14(1): 525–32., DOI 10.3892/ol.2017.6198 Wang C, Wang X, Han A, Wang Y, Jiang H. Proof-of-concept study investigating the role of S100P-RAGE in nasopharyngeal carcinoma. Exp Ther Med, 2021, 21(5):470., DOI 10.3892/etm.2021.9901 Yu Y, Lin ZX, Li HW, Luo HQ, Yang DH, Zhou HC, et al. Circulating tumor cells and fibronectin 1 in the prognosis of nasopharyngeal carcinoma. Technol Cancer Res Treat, 2020, 19:1533033820909911., DOI 10.1177/1533033820909911 Ding Y, Pan Y, Liu S, Jiang F, Jiao J. Elevation of MiR-9-3p suppresses the epithelial-mesenchymal transition of nasopharyngeal carcinoma cells via downregulating FN1, ITGB1 and ITGAV. Cancer Biol Ther, 2017, 18(6):414–24., DOI 10. 1080/15384047.2017.1323585 Wang J, Deng L, Huang J, Cai R, Zhu X, Liu F, et al. High expression of Fibronectin 1 suppresses apoptosis through the NF-κB pathway and is associated with migration in nasopharyngeal carcinoma. Am J Transl Res, 2017, 9(10):4502–11. Liu Q, Zhang W, Wu Z, Liu H, Hu H, Shi H, et al. Construction of a circular RNA-microRNA-messengerRNA regulatory network in stomach adenocarcinoma. J Cel Biochem, 2020, 121(2):1317–31., DOI 10.1002/jcb.29368 Peng Y, Croce CM. The role of MicroRNAs in human cancer. Signal Transduct Target Ther, 2016, 1:15004., DOI 10.1038/sigtrans.2015.4 Lin X, Wang S, Lin K, Zong J, Zheng Q, Su Y, et al. Competitive endogenous RNA landscape in epstein-barr virus associated nasopharyngeal carcinoma. Front Cel Dev Biol, 2021, 9:782473., DOI 10.3389/fcell.2021.782473 Pamudurti NR, Bartok O, Jens M, Ashwal-Fluss R, Stottmeister C, Ruhe L, et al. Translation of CircRNAs. Mol Cel, 2017, 66(1):9–21., DOI 10.1016/j.molcel.2017.02.021 Zhang S, Li Y, Xin S, Yang L, Jiang M, Xin Y, et al. Insight into LncRNA- and CircRNA-mediated CeRNAs: Regulatory network and implications in nasopharyngeal carcinoma-A narrative literature review. Cancers, Basel)., 2022, 14(19):4564., DOI 10.3390/cancers14194564 Li S, Wang Q. Hsa_circ_0081534 increases the proliferation and invasion of nasopharyngeal carcinoma cells through regulating the miR-508-5p/FN1 axis. Aging, Albany NY,, 2020, 12(20):20645–57., DOI 10.18632/aging.103963 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610960 EP 1610971 DI 10.3389/pore.2023.1610960 PG 12 ER PT J AU Cao, X Wu, B Guo, Sh Zhong, W Zhu, Sh Zhang, Z Gu, L Li, H AF Cao, Xiying Wu, Bingqun Guo, Shaoming Zhong, Weixiang Zhu, Shenyu Zhang, Zuxiong Gu, Liang Li, Hui TI APOC1 predicts a worse prognosis for esophageal squamous cell carcinoma and is associated with tumor immune infiltration during tumorigenesis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE bioinformatics; prognosis; expression; APOC1; ESCA ID bioinformatics; prognosis; expression; APOC1; ESCA AB Background: Esophageal carcinoma (ESCA), a common malignant tumor of the digestive tract with insidious onset, is a serious threat to human health. Despite multiple treatment modalities for patients with ESCA, the overall prognosis remains poor. Apolipoprotein C1 (APOC1) is involved in tumorigenesis as an inflammation-related molecule, and its role in esophageal cancer is still unknown. Methods: We downloaded documents and clinical data using The Cancer Genome Atlas (TCGA)and Gene Expression Omnibus (GEO) databases. We also conducted bioinformatics studies on the diagnostic value, prognostic value, and correlation between APOC1 and immune infiltrating cells in ESCA through STRING (https://cn.string-db.org/), the TISIDB (http://cis.hku.hk/ TISIDB/) website, and various other analysis tools. Results: In patients with ESCA, APOC1 was significantly more highly expressed in tumor tissues than in normal tissues (p < 0.001). APOC1 could diagnose ESCA more accurately and determine the TNM stage and disease classification with high accuracy (area under the curve, AUC≥0.807). The results of the Kaplan–Meier curve analysis showed that APOC1 has prognostic value for esophageal squamous carcinoma (ESCC) (p = 0.043). Univariate analysis showed that high APOC1 expression in ESCC was significantly associated with worse overall survival (OS) (p = 0.043), and multivariate analysis shows that high APOC1 expression was an independent risk factor for the OS of patients with ESCC (p = 0.030). In addition, the GO (gene ontology)/KEGG (Kyoto encyclopedia of genes and genomes) analysis showed a concentration of gene enrichment in the regulation of T-cell activation, cornification, cytolysis, external side of the plasma membrane, MHC protein complex, MHC class II protein complex, serine-type peptidase activity, serine-type endopeptidase activity, Staphylococcus aureus infection, antigen processing and presentation, and graft-versus-host disease (all p < 0.001). GSEA (gene set enrichment analysis) showed that enrichment pathways such as immunoregulatory-interactions between a lymphoid and non-lymphoid cell (NES = 1.493, p. adj = 0.023, FDR = 0.017) and FCERI-mediated NF-KB activation (NES = 1.437, p. adj = 0.023, FDR = 0.017) were significantly enriched in APOC1-related phenotypes. In addition, APOC1 was significantly associated with tumor immune infiltrating cells and immune chemokines. Conclusion: APOC1 can be used as a prognostic biomarker for esophageal cancer. Furthermore, as a novel prognostic marker for patients with ESCC, it may have potential value for further investigation regarding the diagnosis and treatment of this group of patients. C1 [Cao, Xiying] Capital Medical University, Beijing Chao-Yang Hospital, Department of Thoracic SurgeryBeijing, China. [Wu, Bingqun] First Hospital of Tsinghua University Beijing, Huaxin Hospital, Department of Thoracic SurgeryBeijing, China. [Guo, Shaoming] The First Affiliated Hospital of Gannan Medical University, Department of Thoracic SurgeryGanzhou, Jiangxi, China. [Zhong, Weixiang] The First Affiliated Hospital of Gannan Medical University, Department of Thoracic SurgeryGanzhou, Jiangxi, China. [Zhu, Shenyu] The First Affiliated Hospital of Gannan Medical University, Department of Thoracic SurgeryGanzhou, Jiangxi, China. [Zhang, Zuxiong] The First Affiliated Hospital of Gannan Medical University, Department of Thoracic SurgeryGanzhou, Jiangxi, China. [Gu, Liang] The First Affiliated Hospital of Gannan Medical University, Department of Thoracic SurgeryGanzhou, Jiangxi, China. [Li, Hui] Capital Medical University, Beijing Chao-Yang Hospital, Department of Thoracic SurgeryBeijing, China. RP Li, H (reprint author), Capital Medical University, Beijing Chao-Yang Hospital, Department of Thoracic Surgery, Beijing, China. EM huilee@vip.sina.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Pennathur A, Gibson MK, Jobe BA, Luketich JD. Oesophageal carcinoma. Lancet, 2013, 381(9864):400–12., DOI 10.1016/S0140-6736(12)60643-6 Arnold M, Soerjomataram I, Ferlay J, Forman D. Global incidence of oesophageal cancer by histological subtype in 2012. Gut, 2015, 64(3):381–7., DOI 10.1136/gutjnl-2014-308124 Lin Y, Totsuka Y, He Y, Kikuchi S, Qiao Y, Ueda J, et al. Epidemiology of esophageal cancer in Japan and China. J Epidemiol, 2013, 23(4):233–42., DOI 10. 2188/jea.je20120162 Graham AJ, Shrive FM, Ghali WA, Manns BJ, Grondin SC, Finley RJ, et al. Defining the optimal treatment of locally advanced esophageal cancer: A systematic review and decision analysis. Ann Thorac Surg, 2007, 83(4):1257–64., DOI 10.1016/j. athoracsur.2006.11.061 Smyth EC, Lagergren J, Fitzgerald RC, Lordick F, Shah MA, Lagergren P, et al. Oesophageal cancer. Nat Rev Dis Primers, 2017, 3:17048., DOI 10.1038/nrdp.2017.48 ChenW, Zheng R, Zeng H, Zhang S. The updated incidences and mortalities of major cancers in China, 2011. Chin J Cancer, 2015, 34(11):502–7., DOI 10.1186/ s40880-015-0042-6 Chen G, Wang Z, Liu XY, Liu FY. Recurrence pattern of squamous cell carcinoma in the middle thoracic esophagus after modified Ivor-Lewis esophagectomy. World J Surg, 2007, 31(5):1107–14., DOI 10.1007/s00268-006- 0551-1 Homet Moreno B, Ribas A. Anti-programmed cell death protein-1/ligand- 1 therapy in different cancers. Br J Cancer, 2015, 112(9):1421–7., DOI 10.1038/bjc. 2015.124 Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov, 2019, 18(3):175–96., DOI 10.1038/s41573- 018-0006-z Zhang W, Yan C, Gao X, Li X, Cao F, Zhao G, et al. Safety and feasibility of radiotherapy plus camrelizumab for locally advanced esophageal squamous cell carcinoma. Oncologist, 2021, 26:e1110–e1124., DOI 10.1002/onco.13797 Chouaib S. EMT in immuno-resistance. Oncoscience, 2015, 2:841–2., DOI 10. 18632/oncoscience.226 Yamamura K, Izumi D, Kandimalla R, Sonohara F, Baba Y, Yoshida N, et al. Intratumoral fusobacteriukm nucleatum levels predict therapeutic response to neoadjuvant chemotherapy in esophageal squamous cell carcinoma. Clin Cancer Res, 2019, 25:6170–9., DOI 10.1158/1078-0432.CCR-19-0318 Jong MC, Hofker MH, Havekes LM. Role of ApoCs in lipoprotein metabolism: Functional differences between ApoC1, ApoC2, and ApoC3, Arterioscler Thromb Vasc Biol, 1999, 19(3):472–84., DOI 10.1161/01. atv.19.3.472 Yi J, Ren L,Wu J, Li W, Zheng X, Du G, et al. Apolipoprotein C1, APOC1, as a novel diagnostic and prognostic biomarker for gastric cancer, Ann Transl Med, 2019, 7(16):380., DOI 10.21037/atm.2019.07.59 Zhang J, Guo F, Wang L, Zhao W, Zhang D, Yang H, et al. Identification of apolipoprotein C-I as a potential Wilms’ tumor marker after excluding inflammatory factors. Int J Mol Sci, 2014, 15(9):16186–95., DOI 10.3390/ ijms150916186 Ko HL, Wang YS, Fong WL, Chi MS, Chi KH, Kao SJ. Apolipoprotein C1, APOC1, as a novel diagnostic and prognostic biomarker for lung cancer: A marker phase I trial. Thorac Cancer, 2014, 5:500–8., DOI 10.1111/1759-7714. 12117 Ren H, Chen Z, Yang L, Xiong W, Yang H, Xu K, et al. Apolipoprotein C1, APOC1, promotes tumor progression via MAPK signaling pathways in colorectal cancer. Cancer Manag Res, 2019, 11(1):4917–30., DOI 10.2147/CMAR.S192529 Jiang H, Tang JY, Xue D, Chen YM, Wu TC, Zhuang QF, et al. Apolipoprotein C1 stimulates the malignant process of renal cell carcinoma via the Wnt3a signaling, Cancer Cel Int, 2021, 21(1):41., DOI 10.1186/s12935-020-01713-x Moore LE, Brennan P, Karami S, Menashe I, Berndt SI, Dong LM, et al. Apolipoprotein E/C1 locus variants modify renal cell carcinoma risk. Cancer Res, 2009, 69(20):8001–8., DOI 10.1158/0008-5472.CAN-09-1734 Takano S, Yoshitomi H, Togawa A, Sogawa K, Shida T, Kimura F, et al. Apolipoprotein C-1 maintains cell survival by preventing from apoptosis in pancreatic cancer cells. Oncogene, 2008, 27(20):2810–22., DOI 10.1038/sj.onc. 1210951 Dvorak HF. Tumors: Wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med, 1986, 315(26):1650–9., DOI 10. 1056/NEJM198612253152606 Chechlinska M, Kowalewska M, Nowak R. Systemic inflammation as a confounding factor in cancer biomarker discovery and validation. Nat Rev Cancer, 2010, 10(1):2–3., DOI 10.1038/nrc2782 Dvorak HF. Tumors: Wounds that do not heal-redux. Cancer Immunol Res, 2015, 3(1):1–11., DOI 10.1158/2326-6066.CIR-14-0209 Wu CX, Rao DY, Sang CP, Zhu SY, Gu L, Wu YY, et al. Peripheral blood inflammation indices are effective predictors of anastomotic leakage in elective esophageal surgery. J Gastrointest Oncol, 2021, 12, 2675–84., DOI 10.21037/jgo- 21-812 Berbee JFP, Havekes LM, Rensen PCN. Apolipoproteins modulate the inflammatory response to lipopolysaccharide. SAGE, 2005, 112:97–103., DOI 10. 1177/09680519050110020501 Guo Q, Liu XL, Jiang N, Zhang WJ, Guo SW, Yang H, et al. Decreased APOC1 expression inhibited cancer progression and was associated with better prognosis and immune microenvironment in esophageal cancer. Am J Cancer Res, 2022, 12(11):4904–29. Clough E, Barrett T. The gene expression omnibus database. Methods Mol Biol, 2016, 1418:93–110., DOI 10.1007/978-1-4939-3578-9_5 Tomczak K, Czerwinska P, Wiznerowicz M. The cancer genome atlas, TCGA): An immeasurable source of knowledge. Contemp Oncol, Pozn,, 2015, 19:A68–77., DOI 10.5114/wo.2014.47136 Consortium GT. The genotype-tissue expression, GTEx, project. Nat Genet, 2013, 45(6):580–5., DOI 10.1038/ng.2653 Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res, 2019, 47:D607–D613., DOI 10.1093/nar/gky1131 Ru B, Wong CN, Tong Y, Zhong JY, Zhong SSW, Wu WC, et al. Tisidb: An integrated repository portal for tumor-immune system interactions. Bioinformatics, 2019, 35(20):4200–2., DOI 10.1093/bioinformatics/btz210 Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell, 2015, 27(4): 450–61., DOI 10.1016/j.ccell.2015.03.001 Hanahan D, Coussens LM. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell, 2012, 21(3):309–22., DOI 10. 1016/j.ccr.2012.02.022 Gibney GT,Weiner LM, Atkins MB, Predictive biomarkers for checkpoint inhibitorbased immunotherapy. Lancet Oncol, 2016, 17(3):e542–51., DOI 10.1016/S1470-2045(16, 30406-5 Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol, 2002, 23:549–55., DOI 10.1016/s1471-4906(02)02302-5 MiX, Xu R, Hong S, Xu T, ZhangW, Liu M. Macrophage-derived exosomal lncRNA AFAP1-AS1 and MicroRNA-26a affect cell migration and metastasis in esophageal cancer. Mol Ther Nucleic Acids, 2020, 22:779–90., DOI 10.1016/j.omtn.2020.09.035 Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res, 2013, 73:1128–41., DOI 10.1158/0008-5472.CAN-12-2731 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610976 EP 1610990 DI 10.3389/pore.2023.1610976 PG 15 ER PT J AU Zhao, N Mei, N Yi, Y Wang, H Wang, Y Yao, Y Li, Ch AF Zhao, Ni Mei, Nan Yi, Ye Wang, Hongyan Wang, Yajian Yao, Yu Li, Chunli TI Case report: Pathological and genetic features of pancreatic undifferentiated carcinoma with osteoclast-like giant cells SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE prognosis; pathological features; undifferentiated carcinoma; osteoclast-like giant cells; pancreatic carcinoma ID prognosis; pathological features; undifferentiated carcinoma; osteoclast-like giant cells; pancreatic carcinoma AB Objectives: Pancreatic undifferentiated carcinoma accounts for 2%–7% of pancreatic carcinomas. We aimed to investigate the pathological and genetic characteristics of pancreatic undifferentiated carcinoma with osteoclast-like giant cells and the key points of treatment. Methods: The clinical data and follow-up results of four patients diagnosed with pancreatic undifferentiated carcinoma with osteoclast-like giant cells between May 2015 and May 2020 at the First Affiliated Hospital of Xi’an Jiaotong University were retrospectively analyzed. Results: Chief complaints included “pain and discomfort in the upper abdomen” (2/4), “nausea and vomiting” (1/4) or no symptoms (1/4). Preoperative mildly elevated tumor markers included carcinoembryonic antigen (1/4) and CA19-9 (1/4). The tumors were located in the tail of the pancreas in three patients and the head and neck in one patient. Tumor metastasis was found in pancreatic adipose tissue in two patients and lymph node metastasis in one patient, with microscopic heterogeneous mononuclear cells and scattered osteoclast-like giant cells of various sizes. One patient (1/4) had a mucinous cystic tumor of the pancreas, and two patients (2/4) had adenocarcinoma of the pancreatic duct. Only one patient received postoperative gemcitabine combined with albuminbound paclitaxel chemotherapy. Conclusion: Currently, treatment guidelines are lacking for PUC-OGC, and prognosis varies markedly. More cases must be reported to clarify its origination. The long-term follow-up of diagnosed patients and genetic mutation testing can also contribute to improving treatment and prognosis of this disease. C1 [Zhao, Ni] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Medical OncologyXi’an, Shaanxi, China. [Mei, Nan] The First Affiliated Hospital of Xi’an Jiaotong University, Department of HematologyXi’an, Shaanxi, China. [Yi, Ye] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Medical OncologyXi’an, Shaanxi, China. [Wang, Hongyan] The First Affiliated Hospital of Xi’an Jiaotong University, Department of PathologyXi’an, Shaanxi, China. [Wang, Yajian] The First Affiliated Hospital of Xi’an Jiaotong University, Department of PathologyXi’an, Shaanxi, China. [Yao, Yu] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Medical OncologyXi’an, Shaanxi, China. [Li, Chunli] The First Affiliated Hospital of Xi’an Jiaotong University, Department of Medical OncologyXi’an, Shaanxi, China. RP Li, Ch (reprint author), The First Affiliated Hospital of Xi’an Jiaotong University, Department of Medical Oncology, Xi’an, China. EM chunli5158@163.com CR Rosai J. Carcinoma of pancreas simulating giant cell tumor of bone. Electronmicroscopic evidence of its acinar cell origin. Cancer, 1968, 22(2):333–44., DOI 10. 1002/1097-0142(196808)22:2<333::aid-cncr2820220210>3.0.co;2-a Manduch M, Dexter DF, Jalink DW, Vanner SJ, Hurlbut DJ. Undifferentiated pancreatic carcinoma with osteoclast-like giant cells: Report of a case with osteochondroid differentiation. Pathol Res Pract, 2009, 205(5):353–9., DOI 10. 1016/j.prp.2008.11.006 Freedman AN, Klabunde CN, Wiant K, Enewold L, Gray SW, Filipski KK, et al. Use of next-generation sequencing tests to guide cancer treatment: Results from a nationally representative survey of oncologists in the United States. Jco Precision Oncol, 2018, 2:1–13., DOI 10.1200/PO.18.00169 Fu LP, Cheng AP, Wang XG, Fu JL, Jin L. 18F-FDG PET/CT in the detection of undifferentiated carcinoma with osteoclast-like giant cells of the pancreas. Clin Nucl Med, 2017, 42(8):615–6., DOI 10.1097/RLU.0000000000001719 Yang KY, Choi JI, Choi MH, Park MY, Rha SE, Byun JY, et al. Magnetic resonance imaging findings of undifferentiated carcinoma with osteoclast-like giant cells of pancreas. Clin Imaging, 2016, 40(1):148–51., DOI 10.1016/j.clinimag.2015. 09.013 Putzer D, Jaschke W. Radiological evaluation of focal pancreatic lesions. Dig Dis, 2015, 33(1):91–8., DOI 10.1159/000366045 Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature, 2015, 518(7540):495–501., DOI 10.1038/nature14169 Wu C, Miao X, Huang L, Che X, Jiang G, Yu D, et al. Genome-wide association study identifies five loci associated with susceptibility to pancreatic cancer in Chinese populations. Nat Genet, 2011, 44(1):62–6., DOI 10.1038/ng.1020 Visani M, de Biase D, Baccarini P, Fabbri C, Polifemo AM, Zanini N, et al. Multiple KRAS mutations in pancreatic adenocarcinoma: molecular features of neoplastic clones indicate the selection of divergent populations of tumor cells. Int J Surg Pathol, 2013, 21(6):546–52., DOI 10.1177/ 1066896912475073 Mann KM, Ying H, Juan J, Jenkins NA, Copeland NG. KRAS-related proteins in pancreatic cancer. Pharmacol Ther, 2016, 168:29–42., DOI 10.1016/j.pharmthera. 2016.09.003 Li L, Mao Y, Zhao L, Wu J, Zhao M, Du W, et al. p53 regulation of ammonia metabolism through urea cycle controls polyamine biosynthesis. Nature, 2019, 567(7747):253–6., DOI 10.1038/s41586-019-0996-7 Grochola LF, Taubert H, Greither T, Bhanot U, Udelnow A, Wurl P. Elevated transcript levels from the MDM2 P1 promoter and low p53 transcript levels are associated with poor prognosis in human pancreatic ductal adenocarcinoma. Pancreas, 2011, 40(2):265–70., DOI 10.1097/mpa. 0b013e3181f95104 Chan TA, Wolchok JD, Snyder A. Genetic basis for clinical response to CTLA- 4 blockade in melanoma. N Engl J Med, 2015, 373:1984., DOI 10.1056/ NEJMc1508163 Zhang CM, Lv JF, Gong L, Yu LY, Chen XP, Zhou HH, et al. Role of deficient mismatch repair in the personalized management of colorectal cancer. Int J Environ Res Public Health, 2016, 13:892., DOI 10.3390/ijerph13090892 Singhi AD, George B, Greenbowe JR, Chung J, Suh J, Maitra A, et al. Realtime targeted genome profile analysis of pancreatic ductal adenocarcinomas identifies genetic alterations that might Be targeted with existing drugs or used as biomarkers. Gastroenterology, 2019, 156(8):2242–53., DOI 10.1053/j.gastro.2019. 02.037 Salem ME, Puccini A, Grothey A, Raghavan D, Goldberg RM, Xiu J, et al. Landscape of tumor mutation load, mismatch repair deficiency, and PD-L1 expression in a large patient cohort of gastrointestinal cancers. Mol Cancer Res, 2018, 16(5):805–12., DOI 10.1158/1541-7786.MCR-17-0735 Jiang Y, Li Y, Zhu B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis, 2015, 6(6):e1792., DOI 10.1038/cddis.2015.162 LoosM, Giese NA, Kleeff J, Giese T, Gaida MM, Bergmann F, et al. Clinical significance and regulation of the costimulatory molecule B7-H1 in pancreatic cancer. Cancer Lett Sep 8, 2008, 268(1):98–109., DOI 10.1016/j.canlet.2008. 03.056 Chen Y, Sun J, Zhao H, Zhu D, Zhi Q, Song S, et al. The coexpression and clinical significance of costimulatory molecules B7-H1, B7-H3, and B7-H4 in human pancreatic cancer. Onco Targets Ther, 2014, 7:1465–72., DOI 10.2147/ OTT.S66809 Wang L, Ma Q, Chen X, Guo K, Li J, Zhang M. Clinical significance of B7-H1 and B7-1 expressions in pancreatic carcinoma. World J Surg, 2010, 34(5):1059–65., DOI 10.1007/s00268-010-0448-x Geng L, Huang D, Liu J, Qian Y, Deng J, et al. B7-H1 up-regulated expression in human pancreatic carcinoma tissue associates with tumor progression. J Cancer Res Clin Oncol, 2008, 134(9):1021–7., DOI 10.1007/s00432-008-0364-8 Birnbaum DJ, Finetti P, Lopresti A, Gilabert M, Poizat F, Turrini O, et al. Prognostic value of PDL1 expression in pancreatic cancer. Oncotarget, 2016, 7(44): 71198–210., DOI 10.18632/oncotarget.11685 Luchini C, Cros J, Pea A, Pilati C, Veronese N, Rusev B, et al. PD-1, PD-L1, and CD163 in pancreatic undifferentiated carcinoma with osteoclast-like giant cells: Expression patterns and clinical implications. Hum Pathol, 2018, 81:157–65., DOI 10.1016/j.humpath.2018.07.006 Hrudka J, Lawrie K, Waldauf P, Ciprova V, Moravcova J, Matej R. Negative prognostic impact of PD-L1 expression in tumor cells of undifferentiated, anaplastic, carcinoma with osteoclast-like giant cells of the pancreas: Study of 13 cases comparing ductal pancreatic carcinoma and review of the literature. Virchows Arch, 2020, 477(5):687–96., DOI 10.1007/s00428-020-02830-8 Khorana AA, McKernin SE, Berlin J, Hong TS, Maitra A, Moravek C, et al. Potentially curable pancreatic adenocarcinoma: ASCO clinical Practice guideline update. J ClinOncol, 2019, 37(23):2082–8., DOI 10.1200/jco.19.00946 Temesgen WM, Wachtel M, Dissanaike S. Osteoclastic giant cell tumor of the pancreas. Int J Surg Case Rep, 2014, 5(4):175–9., DOI 10.1016/j.ijscr.2014. 01.002 Sohal DPS, Kennedy EB, Khorana A, Copur MS, Crane CH, Garrido- Laguna I, et al. Metastatic pancreatic cancer: ASCO clinical Practice guideline update. J Clin Oncol, 2018, 36(24):2545–56., DOI 10.1200/JCO.2018. 78.9636 Yoshioka M, Uchinami H, Watanabe G, Takahashi T, Nakagawa Y, Andoh H, et al. Effective use of gemcitabine in the treatment of undifferentiated carcinoma with osteoclast-like giant cells of the pancreas with portal vein tumor thrombus. Intern Med, 2012, 51(16):2145–50., DOI 10.2169/ internalmedicine.51.7670 Gao HQ, Yang YM, Zhuang Y, Liu P. Locally advanced undifferentiated carcinoma with osteoclast-like giant cells of the pancreas. World J Gastroenterol, 2015, 21(2):694–8., DOI 10.3748/wjg.v21.i2.694 Singhal A, Shrago SS, Li SF, Huang Y, Kohli V. Giant cell tumor of the pancreas: A pathological diagnosis with poor prognosis. Hepatobiliary Pancreat Dis Int, 2010, 9(4):433–7. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610983 EP 1610993 DI 10.3389/pore.2023.1610983 PG 11 ER PT J AU Ma, YT Li, Y Yan, L Hua, F Wang, DG Xu, GY Yang, HL Xue, YJ Qin, YJ Sha, D Ning, H Zhao, MQ Yao, ZG AF Ma, Yu-Ting Li, Yan Yan, Li Hua, Fang Wang, Dong-Guan Xu, Guo-Ying Yang, Hong-Lan Xue, Ying-Jie Qin, Ye-Jun Sha, Dan Ning, Hao Zhao, Miao-Qing Yao, Zhi-Gang TI Corrigendum: Case report: Potential predictive value of MMR/MSI status and PD-1 expression in immunotherapy for urothelial carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE immunotherapy; microsatellite instability; immune checkpoint inhibitors; urothelial carcinoma; lynch syndrome; PD-1/PD-L1 ID immunotherapy; microsatellite instability; immune checkpoint inhibitors; urothelial carcinoma; lynch syndrome; PD-1/PD-L1 AB A Corrigendum on Case report: Potential predictive value of MMR/MSI status and PD-1 expression in immunotherapy for urothelial carcinoma by Ma Y-T, Li Y, Yan L, Hua F, Wang D-G, Xu G-Y, Yang H-L, Xue Y-J, Qin Y-J, Sha D, Ning H, Zhao M-Q and Yao Z-G (2022). Pathol Oncol Res. 28:1610638. doi: 10.3389/pore.2022. 1610638 C1 [Ma, Yu-Ting] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Li, Yan] Dongying City People’s Hospital, Department of OncologyDongying, China. [Yan, Li] Dongying City People’s Hospital, Department of PathologyDongying, China. [Hua, Fang] Tulane University, Department of Microbiology and ImmunologyNew Orleans, LA, USA. [Wang, Dong-Guan] Dongying City People’s Hospital, Department of PathologyDongying, China. [Xu, Guo-Ying] Dongying Hospital of Traditional Chinese Medicine, Department of Urology SurgeryDongying, China. [Yang, Hong-Lan] Dongying City People’s Hospital, Department of OncologyDongying, China. [Xue, Ying-Jie] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Qin, Ye-Jun] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Sha, Dan] Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of OncologyJinan, China. [Ning, Hao] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of UrologyJinan, China. [Zhao, Miao-Qing] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. [Yao, Zhi-Gang] Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of PathologyJinan, China. RP Yao, ZG (reprint author), Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of Pathology, Jinan, China. EM zhaomqsd@163.com NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610989 EP 1610989 DI 10.3389/pore.2023.1610989 PG 1 ER PT J AU Sztankovics, D Moldvai, D Petovari, G Gelencser, R Krencz, I Raffay, R Danko, T Sebestyen, A AF Sztankovics, Daniel Moldvai, Dorottya Petovari, Gabor Gelencser, Rebeka Krencz, Ildiko Raffay, Regina Danko, Titanilla Sebestyen, Anna TI 3D bioprinting and the revolution in experimental cancer model systems—A review of developing new models and experiences with in vitro 3D bioprinted breast cancer tissue-mimetic structures SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cancer; breast cancer; 3D bioprinting; disease models; biofabrication ID cancer; breast cancer; 3D bioprinting; disease models; biofabrication AB Growing evidence propagates those alternative technologies (relevant human cell-based—e.g., organ-on-chips or biofabricated models—or artificial intelligence-combined technologies) that could help in vitro test and predict human response and toxicity in medical research more accurately. In vitro disease model developments have great efforts to create and serve the need of reducing and replacing animal experiments and establishing human cell-based in vitro test systems for research use, innovations, and drug tests. We need human cell-based test systems for disease models and experimental cancer research; therefore, in vitro three-dimensional (3D) models have a renaissance, and the rediscovery and development of these technologies are growing ever faster. This recent paper summarises the early history of cell biology/cellular pathology, cell-, tissue culturing, and cancer research models. In addition, we highlight the results of the increasing use of 3D model systems and the 3D bioprinted/biofabricated model developments. Moreover, we present our newly established 3D bioprinted luminal B type breast cancer model system, and the advantages of in vitro 3D models, especially the bioprinted ones. Based on our results and the reviewed developments of in vitro breast cancer models, the heterogeneity and the real in vivo situation of cancer tissues can be represented better by using 3D bioprinted, biofabricated models. However, standardising the 3D bioprinting methods is necessary for future applications in different high-throughput drug tests and patient-derived tumour models. Applying these standardised new models can lead to the point that cancer drug developments will be more successful, efficient, and consequently costeffective in the near future. C1 [Sztankovics, Daniel] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Moldvai, Dorottya] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Petovari, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Gelencser, Rebeka] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Krencz, Ildiko] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Raffay, Regina] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Danko, Titanilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Sebestyen, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM hsebanna@gmail.com CR Regulation of the European parliament and of the Council on cosmetic products, No 1223/2009, 2009). Available at: https://eur-lex.europa.eu/legal-content/EN/ ALL/?uri=celex%3A32009R1223, Accessed November 03, 2022). Swaters D, van Veen A, van Meurs W, Turner JE, Ritskes-Hoitinga M. A history of regulatory animal testing: What can we learn? Alternatives Lab Anim, 2022, 50(5):322–9., DOI 10.1177/02611929221118001 Van Norman GA. Limitations of animal studies for predicting toxicity in clinical trials: Is it time to rethink our current approach? JACC Basic Transl Sci, 2019, 4(7):845–54., DOI 10.1016/j.jacbts.2019.10.008 Wilson JG, Ritter EJ, Scott WJ, Fradkin R. Comparative distribution and embryotoxicity of acetylsalicylic acid in pregnant rats and rhesus monkeys. Toxicol Appl Pharmacol, 1977, 41(1):67–78., DOI 10.1016/0041-008x(77, 90054-0 Ritskes-Hoitinga M. Medical regulators: Look beyond animal tests. Nature, 2022, 604(7907):599., DOI 10.1038/d41586-022-01110-6 Sandeep Ganesh G, Kolusu AS, Prasad K, Samudrala PK, Nemmani KVS. Advancing health care via artificial intelligence: From concept to clinic. Eur J Pharmacol, 2022, 934:175320., DOI 10.1016/j.ejphar.2022.175320 Jahagirdar D, Bangde P, Jain R, Dandekar P. Degenerative disease-on-a-chip: Developing microfluidic models for rapid availability of newer therapies. Biotechnol J, 2021, 16(10):e2100154., DOI 10.1002/biot.202100154 Parke EC. Flies from meat and wasps from trees: Reevaluating Francesco Redi’s spontaneous generation experiments. Stud Hist Philos Biol Biomed Sci, 2014, 45: 34–42., DOI 10.1016/j.shpsc.2013.12.005 Hajdu SI. The first use of the microscope in medicine. Ann Clin Lab Sci, 2002, 32(3):309–10. Mazzarello P. A unifying concept: The history of cell theory. Nat Cel Biol, 1999, 1(1):E13–5., DOI 10.1038/8964 Louis P. On spontaneous generation. Revue de Cours Scientifics, 1864)(1, 257–64. Schwann T, Smith H, Schleiden MJ. Microscopical researches into the accordance in the structure and growth of animals and plants. London: Sydenham Society, 1847). Hall TS. Ideas of life and matter; studies in the history of general physiology, 600 B.C.-1900 A.D. Chicago: Thomas S. HallUniversity of Chicago Press, 1969). Schultz MG. Rudolf Virchow. Emerging Infect Dis, 2008, 14(9):1480–1., DOI 10.3201/eid1409.086672 Wagner RP. Anecdotal, historical and critical commentaries on genetics. Rudolph Virchow and the genetic basis of somatic ecology. Genetics, 1999, 151(3): 917–20., DOI 10.1093/genetics/151.3.917 Roux W. Gesammelte Abhandlungen uber Entwickelungsmechanik der Organismen. Leipzig: Wilhelm Engelmann, 1895). Blumenthal HT, Loeb L. Leo Loeb, experimental pathologist and humanitarian. Science, 1960, 131(3404):907–8., DOI 10.1126/science.131.3404.907 Harrison RG. Observations on the living developing nerve fiber. Proc Soc Exp Biol Med, 1906, 4(1):140–3., DOI 10.3181/00379727-4-98 Carrel A, Burrows MT. An addition to the technique of the cultivation of tissues in vitro. J Exp Med, 1911, 14(3):244–7., DOI 10.1084/jem.14.3.244 Rous P, Jones FS. A method for obtaining suspensions of living cells from the fixed tissues, and for the plating out of individual cells. J Exp Med, 1916, 23(4): 549–55., DOI 10.1084/jem.23.4.549 Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cel Res, 1961, 25:585–621., DOI 10.1016/0014-4827(61)90192-6 Hahn WC. Immortalization and transformation of human cells. Mol Cell, 2002, 13(3):351–61. Yamanaka S. Induced pluripotent stem cells: Past, present, and future. Cell Stem Cell, 2012, 10(6):678–84., DOI 10.1016/j.stem.2012.05.005 Hajdu SI. Pathfinders in oncology from ancient times to the end of theMiddle Ages. Cancer, 2016, 122(11):1638–46., DOI 10.1002/cncr.29955 Waldron HA. A brief history of scrotal cancer. Br J Ind Med, 1983, 40(4): 390–401., DOI 10.1136/oem.40.4.390 Rous P. A transmissible avian neoplasm. sarcoma of the common fowl. J Exp Med, 1910, 12(5):696–705., DOI 10.1084/jem.12.5.696 Stehelin D, Varmus HE, Bishop JM, Vogt PK. DNA related to the transforming gene(s, of avian sarcoma viruses is present in normal avian DNA. Nature, 1976, 260(5547):170–3., DOI 10.1038/260170a0 Rygaard J, Povlsen CO. Heterotransplantation of a human malignant tumour to "Nude" mice. Acta Pathol Microbiol Scand, 1969, 77(4):758–60., DOI 10.1111/j. 1699-0463.1969.tb04520.x Stanbridge EJ, Perkins FT, Hayflick L. Cell tumourigenicity detected by heterotransplantation into mice immunosuppressed with anti-lymphocytic serum. Prog Immunobiol Stand, 1971, 5:243–51. Kopper L, Steel GG. The therapeutic response of three human tumor lines maintained in immune-suppressed mice. Cancer Res, 1975, 35(10):2704–13. Justice BA, Badr NA, Felder RA. 3D cell culture opens new dimensions in cellbased assays. Drug Discov Today, 2009, 14(1-2):102–7., DOI 10.1016/j.drudis.2008.11.006 Knight A. Systematic reviews of animal experiments demonstrate poor human clinical and toxicological utility. Altern Lab Anim, 2007, 35(6):641–59., DOI 10.1177/ 026119290703500610 Hanahan D. Hallmarks of cancer: New dimensions. Cancer Discov, 2022, 12(1):31–46., DOI 10.1158/2159-8290.Cd-21-1059 Rodrigues J, Heinrich MA, Teixeira LM, Prakash J. 3D in vitro model, R, evolution: Unveiling tumor-stroma interactions. Trends Cancer, 2021, 7(3):249–64., DOI 10.1016/j.trecan.2020.10.009 Asghar W, El Assal R, Shafiee H, Pitteri S, Paulmurugan R, Demirci U. Engineering cancer microenvironments for in vitro 3-D tumor models. Mater Today, Kidlington,, 2015, 18(10):539–53., DOI 10.1016/j.mattod.2015.05.002 Sung KE, Beebe DJ. Microfluidic 3D models of cancer. Adv Drug Deliv Rev, 2014, 79-80:68–78., DOI 10.1016/j.addr.2014.07.002 Langhans SA. Three-dimensional in vitro cell culture models in drug discovery and drug repositioning. Front Pharmacol, 2018, 9:6., DOI 10.3389/ fphar.2018.00006 Duval K, Grover H, Han LH, Mou Y, Pegoraro AF, Fredberg J, et al. Modeling physiological events in 2D vs. 3D cell culture. Physiology, Bethesda,, 2017, 32(4): 266–77., DOI 10.1152/physiol.00036.2016 Jensen C, Teng Y. Is it time to start transitioning from 2D to 3D cell culture? Front Mol Biosci, 2020, 7:33., DOI 10.3389/fmolb.2020.00033 Boland T, Mironov V, Gutowska A, Roth EA, Markwald RR. Cell and organ printing 2: Fusion of cell aggregates in three-dimensional gels. Anat Rec A Discov Mol Cel Evol Biol, 2003, 272(2):497–502., DOI 10.1002/ar.a.10059 Jakab K, Damon B, Neagu A, Kachurin A, Forgacs G. Three-dimensional tissue constructs built by bioprinting. Biorheology, 2006, 43(34):509–13. Asulin M, Michael I, Shapira A, Dvir T. One-step 3D printing of heart patches with built-in electronics for performance regulation. Adv Sci, Weinh,, 2021, 8(9): 2004205., DOI 10.1002/advs.202004205 Shah AM, Han JJ. First successful porcine to human heart transplantation performed in the United States. Artif Organs, 2022, 46(4):543–5., DOI 10.1111/aor. 14203 Yang H, Sun L, Pang Y, Hu D, Xu H, Mao S, et al. Three-dimensional bioprinted hepatorganoids prolong survival of mice with liver failure. Gut, 2021, 70(3):567–74., DOI 10.1136/gutjnl-2019-319960 Ho CMB, Ng SH, Yoon Y-J. A review on 3D printed bioimplants. Int J Precision Eng Manufacturing, 2015, 16(5):1035–46., DOI 10.1007/s12541-015- 0134-x Diment LE, Thompson MS, Bergmann JHM. Clinical efficacy and effectiveness of 3D printing: A systematic review. BMJ Open, 2017, 7(12): e016891., DOI 10.1136/bmjopen-2017-016891 Huang SH, Liu P, Mokasdar A, Hou L. Additive manufacturing and its societal impact: A literature review. Int J Adv Manufacturing Technology, 2013, 67(5): 1191–203., DOI 10.1007/s00170-012-4558-5 Ramesh S, Harrysson OLA, Rao PK, Tamayol A, Cormier D, Zhang Y, et al. Extrusion bioprinting: Recent progress, challenges, and future opportunities. Bioprinting, 2021, 21:e00116., DOI 10.1016/j.bprint.2020.e00116 Yan Q, Dong H, Su J, Han J, Song B, Wei Q, et al. A review of 3D printing technology for medical applications. Engineering, 2018, 4(5):729–42., DOI 10.1016/j. eng.2018.07.021 Nuseir A, Hatamleh MM, Alnazzawi A, Al-Rabab’ah M, Kamel B, Jaradat E. Direct 3D printing of flexible nasal prosthesis: Optimized digital workflow from scan to fit. J Prosthodont, 2019, 28(1):10–4., DOI 10.1111/jopr.13001 Faglin P, GradwohlM, Depoortere C, Germain N, Drucbert A-S, Brun S, et al. Rationale for the design of 3D-printable bioresorbable tissue-engineering chambers to promote the growth of adipose tissue. Scientific Rep, 2020, 10(1):11779., DOI 10. 1038/s41598-020-68776-8 Wang X, Ao Q, Tian X, Fan J, Wei Y, Hou W, et al. 3D bioprinting technologies for hard tissue and organ engineering. Materials, Basel,, 2016, 9(10):802., DOI 10.3390/ma9100802 Groll J, Burdick JA, Cho DW, Derby B, Gelinsky M, Heilshorn SC, et al. A definition of bioinks and their distinction from biomaterial inks. Biofabrication, 2018, 11(1):013001., DOI 10.1088/1758-5090/aaec52 Panwar A, Tan LP. Current status of bioinks for micro-extrusion-based 3D bioprinting. Molecules, 2016, 21(6):685., DOI 10.3390/molecules21060685 Gopinathan J, Noh I. Recent trends in bioinks for 3D printing. Biomater Res, 2018, 22:11., DOI 10.1186/s40824-018-0122-1 Gungor-Ozkerim PS, Inci I, Zhang YS, Khademhosseini A, Dokmeci MR. Bioinks for 3D bioprinting: An overview. Biomater Sci, 2018, 6(5):915–46., DOI 10. 1039/c7bm00765e Das S, Basu B. An overview of hydrogel-based bioinks for 3D bioprinting of soft tissues. J Indian Inst Sci, 2019, 99:405–28., DOI 10.1007/s41745-019-00129-5 GhavamiNejad A, Ashammakhi N,Wu XY, Khademhosseini A. Crosslinking strategies for 3D bioprinting of polymeric hydrogels. Small, 2020, 16(35):2002931., DOI 10.1002/smll.202002931 Germain N, Dhayer M, Dekiouk S, Marchetti P. Current advances in 3D bioprinting for cancer modeling and personalized medicine. Int J Mol Sci, 2022, 23(7):3432., DOI 10.3390/ijms23073432 Augustine R, Kalva SN, Ahmad R, Zahid AA, Hasan S, Nayeem A, et al. 3D Bioprinted cancer models: Revolutionizing personalized cancer therapy. Transl Oncol, 2021, 14(4):101015., DOI 10.1016/j.tranon.2021.101015 Schueler J, Borenstein J, Buti L, Dong M, Masmoudi F, Hribar K, et al. How to build a tumor: An industry perspective. Drug Discov Today, 2022, 27(10):103329., DOI 10.1016/j.drudis.2022.07.014 Moon S, Hasan SK, Song YS, Xu F, Keles HO, Manzur F, et al. Layer by layer three-dimensional tissue epitaxy by cell-laden hydrogel droplets. Tissue Eng C Methods, 2010, 16(1):157–66., DOI 10.1089/ten.TEC.2009.0179 Leonard F, Godin B. 3D in vitro model for breast cancer research using magnetic levitation and bioprinting method. Methods Mol Biol, 2016, 1406:239–51., DOI 10.1007/978-1-4939-3444-7_21 Zhao Y, Yao R, Ouyang L, Ding H, Zhang T, Zhang K, et al. Threedimensional printing of Hela cells for cervical tumor model in vitro. Biofabrication, 2014, 6(3):035001., DOI 10.1088/1758-5082/6/3/035001 Zhu W, Wang M, Fu Y, Castro NJ, Fu SW, Zhang LG. Engineering a biomimetic three-dimensional nanostructured bone model for breast cancer bone metastasis study. Acta Biomater, 2015, 14:164–74., DOI 10.1016/j.actbio. 2014.12.008 Liu C, Hu A, Chen H, Liang J, Gu M, Xiong Y, et al. The osteogenic nichetargeted arsenic nanoparticles prevent colonization of disseminated breast tumor cells in the bone. Acta Pharm Sin B, 2022, 12(1):364–77., DOI 10.1016/j.apsb.2021. 06.012 Kolesky DB, Truby RL, Gladman AS, Busbee TA, Homan KA, Lewis JA. 3D bioprinting of vascularized, heterogeneous cell-laden tissue constructs. Adv Mater, 2014, 26(19):3124–30., DOI 10.1002/adma.201305506 Neiman JA, Raman R, Chan V, Rhoads MG, Raredon MS, Velazquez JJ, et al. Photopatterning of hydrogel scaffolds coupled to filter materials using stereolithography for perfused 3D culture of hepatocytes. Biotechnol Bioeng, 2015, 112(4):777–87., DOI 10.1002/bit.25494 Nguyen DG, Funk J, Robbins JB, Crogan-Grundy C, Presnell SC, Singer T, et al. Bioprinted 3D primary liver tissues allow assessment of organ-level response to clinical drug induced toxicity in vitro. PLoS One, 2016, 11(7):e0158674., DOI 10. 1371/journal.pone.0158674 Li R, Ting Y-H, Youssef SH, Song Y, Garg S. Three-dimensional printing for cancer applications: Research landscape and technologies. Pharmaceuticals, 2021, 14(8):787., DOI 10.3390/ph14080787 Petersen OW, Ronnov-Jessen L, Howlett AR, Bissell MJ. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proc Natl Acad Sci U S A, 1992, 89(19):9064–8., DOI 10.1073/pnas.89.19.9064 Sokol ES, Miller DH, Breggia A, Spencer KC, Arendt LM, Gupta PB. Growth of human breast tissues from patient cells in 3D hydrogel scaffolds. Breast Cancer Res, 2016, 18(1):19., DOI 10.1186/s13058-016-0677-5 Swaminathan S, Hamid Q, Sun W, Clyne AM. Bioprinting of 3D breast epithelial spheroids for human cancer models. Biofabrication, 2019, 11(2):025003., DOI 10.1088/1758-5090/aafc49 Reid JA, Palmer XL, Mollica PA, Northam N, Sachs PC, Bruno RD. A 3D bioprinter platform for mechanistic analysis of tumoroids and chimeric mammary organoids. Sci Rep, 2019, 9(1):7466., DOI 10.1038/s41598-019-43922-z Mollica PA, Booth-Creech EN, Reid JA, Zamponi M, Sullivan SM, Palmer XL, et al. 3D bioprinted mammary organoids and tumoroids in human mammary derived ECM hydrogels. Acta Biomater, 2019, 95:201–13., DOI 10.1016/j.actbio. 2019.06.017 Swaminathan S, Clyne AM. Direct bioprinting of 3D multicellular breast spheroids onto endothelial networks. J Vis Exp, 2020, 165., DOI 10.3791/61791 Chaji S, Al-Saleh J, Gomillion CT. Bioprinted three-dimensional cell-laden hydrogels to evaluate adipocyte-breast cancer cell interactions. Gels, 2020, 6(1):10., DOI 10.3390/gels6010010 Shao L, Gao Q, Xie C, Fu J, Xiang M, He Y. Directly coaxial 3D bioprinting of large-scale vascularized tissue constructs. Biofabrication, 2020, 12(3):035014., DOI 10.1088/1758-5090/ab7e76 Dey M, Ayan B, Yurieva M, Unutmaz D, Ozbolat IT. Studying tumor angiogenesis and cancer invasion in a three-dimensional vascularized breast cancer micro-environment. Adv Biol, Weinh,, 2021, 5(7):e2100090., DOI 10.1002/ adbi.202100090 Suarez-Martinez AD, Sole-Gras M, Dykes SS, Wakefield ZR, Bauer K, Majbour D, et al. Bioprinting on live tissue for investigating cancer cell dynamics. Tissue Eng A, 2021, 27(7-8):438–53., DOI 10.1089/ten.TEA.2020. 0190 Zhu W, Qu X, Zhu J, Ma X, Patel S, Liu J, et al. Direct 3D bioprinting of prevascularized tissue constructs with complex microarchitecture. Biomaterials, 2017, 124:106–15., DOI 10.1016/j.biomaterials.2017.01.042 Horder H, Guaza Lasheras M, Grummel N, Nadernezhad A, Herbig J, Ergun S, et al. Bioprinting and differentiation of adipose-derived stromal cell spheroids for a 3D breast cancer-adipose tissue model. Cells, 2021, 10(4):803., DOI 10.3390/ cells10040803 Langer EM, Allen-Petersen BL, King SM, Kendsersky ND, Turnidge MA, Kuziel GM, et al. Modeling tumor phenotypes in vitro with three-dimensional bioprinting. Cell Rep, 2019, 26(3):608–23., DOI 10.1016/j.celrep.2018.12.090 TangM, Rich JN, Chen S. Biomaterials and 3D bioprinting strategies to model glioblastoma and the blood-brain barrier. AdvMater, 2021, 33(5):e2004776., DOI 10. 1002/adma.202004776 van Pel DM, Harada K, Song D, Naus CC, Sin WC. Modelling glioma invasion using 3D bioprinting and scaffold-free 3D culture. J Cel Commun Signal, 2018, 12(4):723–30., DOI 10.1007/s12079-018-0469-z Heinrich MA, Bansal R, Lammers T, Zhang YS, Michel Schiffelers R, Prakash J. 3D-Bioprinted mini-brain: A glioblastoma model to study cellular interactions and therapeutics. Adv Mater, 2019, 31(14):e1806590., DOI 10.1002/adma.201806590 Nothdurfter D, Ploner C, Coraca-Huber DC, Wilflingseder D, Muller T, Hermann M, et al. 3D bioprinted, vascularized neuroblastoma tumor environment in fluidic chip devices for precision medicine drug testing. Biofabrication, 2022, 14(3):035002., DOI 10.1088/1758-5090/ac5fb7 Neufeld L, Yeini E, Reisman N, Shtilerman Y, Ben-Shushan D, Pozzi S, et al. Microengineered perfusable 3D-bioprinted glioblastoma model for in vivo mimicry of tumor microenvironment. Sci Adv, 2021, 7(34):eabi9119., DOI 10.1126/sciadv.abi9119 Hermida MA, Kumar JD, Schwarz D, Laverty KG, Di Bartolo A, ArdronM, et al. Three dimensional in vitro models of cancer: Bioprinting multilineage glioblastoma models. Adv Biol Regul, 2020, 75:100658., DOI 10.1016/j.jbior.2019.100658 Sivandzade F, Cucullo L. In-vitro blood-brain barrier modeling: A review of modern and fast-advancing technologies. J Cereb Blood Flow Metab, 2018, 38(10): 1667–81., DOI 10.1177/0271678x18788769 Marino A, Tricinci O, BattagliniM, Filippeschi C,Mattoli V, Sinibaldi E, et al. A 3D real-scale, biomimetic, and biohybrid model of the blood-brain barrier fabricated through two-photon lithography. Small, 2018, 14(6):1702959., DOI 10. 1002/smll.201702959 Maloney E, Clark C, Sivakumar H, Yoo K, Aleman J, Rajan SAP, et al. Immersion bioprinting of tumor organoids in multi-well plates for increasing chemotherapy screening throughput. Micromachines, Basel,, 2020, 11(2):208., DOI 10.3390/mi11020208 Gebeyehu A, Surapaneni SK, Huang J, Mondal A, Wang VZ, Haruna NF, et al. Polysaccharide hydrogel based 3D printed tumor models for chemotherapeutic drug screening. Scientific Rep, 2021, 11(1):372., DOI 10. 1038/s41598-020-79325-8 Hughes AM, Kolb AD, Shupp AB, Shine KM, Bussard KM. Printing the pathway forward in bone metastatic cancer research: Applications of 3D engineered models and bioprinted scaffolds to recapitulate the bone-tumor niche. Cancers, Basel,, 2021, 13(3):507., DOI 10.3390/cancers13030507 Grunewald L, Lam T, Andersch L, Klaus A, Schwiebert S, Winkler A, et al. A reproducible bioprinted 3D tumor model serves as a preselection tool for CAR T cell therapy optimization. Front Immunol, 2021, 12:689697., DOI 10.3389/fimmu.2021. 689697 Meng F, Meyer CM, Joung D, Vallera DA, McAlpine MC, Panoskaltsis- Mortari A. 3D bioprinted in vitro metastatic models via reconstruction of tumor microenvironments. Adv Mater, 2019, 31(10):e1806899., DOI 10.1002/adma. 201806899 Quinn CH, Beierle AM, Hutchins SC, Marayati R, Bownes LV, Stewart JE, et al. Targeting high-risk neuroblastoma patient-derived xenografts with oncolytic virotherapy. Cancers, Basel,, 2022, 14(3):762., DOI 10.3390/cancers14030762 Lopez J, Ruiz-Toranzo M, Antich C, Chocarro-Wrona C, Lopez-Ruiz E, Jimenez G, et al. Biofabrication of a tri-layered 3D-bioprinted CSC-based malignant melanoma model for personalized cancer treatment. Biofabrication, 2022)., DOI 10. 1088/1758-5090/ac8dc6 Trujillo-de Santiago G, Flores-Garza BG, Tavares-Negrete JA, Lara- Mayorga IM, Gonzalez-Gamboa I, Zhang YS, et al. The tumor-on-chip: Recent advances in the development of microfluidic systems to recapitulate the physiology of solid tumors. Materials, Basel,, 2019, 12(18):2945., DOI 10. 3390/ma12182945 Ma X, Liu J, Zhu W, Tang M, Lawrence N, Yu C, et al. 3D bioprinting of functional tissue models for personalized drug screening and in vitro disease modeling. Adv Drug Deliv Rev, 2018, 132:235–51., DOI 10.1016/j.addr.2018.06.011 Neufeld L, Yeini E, Pozzi S, Satchi-Fainaro R. 3D bioprinted cancer models: From basic biology to drug development. Nat Rev Cancer, 2022, 22(12):679–92., DOI 10.1038/s41568-022-00514-w Danko T, Petovari G, Sztankovics D, Moldvai D, Raffay R, Lorincz P, et al. Rapamycin plus doxycycline combination affects growth arrest and selective autophagy-dependent cell death in breast cancer cells. Int J Mol Sci, 2021, 22(15):8019., DOI 10.3390/ijms22158019 Danko T, Petovari G, Raffay R, Sztankovics D, Moldvai D, Vetlenyi E, et al. Characterisation of 3D bioprinted human breast cancer model for in vitro drug and metabolic targeting. Int J Mol Sci, 2022, 23(13):7444., DOI 10.3390/ijms23137444 Petovari G, Danko T, Tokes AM, Vetlenyi E, Krencz I, Raffay R, et al. In situ metabolic characterisation of breast cancer and its potential impact on therapy. Cancers, Basel,, 2020, 12(9):2492., DOI 10.3390/cancers12092492 Vasan N, Baselga J, Hyman DM. A view on drug resistance in cancer. Nature, 2019, 575(7782):299–309., DOI 10.1038/s41586-019-1730-1 Fiorillo M, Sotgia F, Lisanti MP. Energetic" cancer stem cells, e-CSCs): A new hyper-metabolic and proliferative tumor cell phenotype, driven by mitochondrial energy. Front Oncol, 2018, 8:677., DOI 10.3389/fonc.2018.00677 Witkiewicz AK, Whitaker-Menezes D, Dasgupta A, Philp NJ, Lin Z, Gandara R, et al. Using the "reverse Warburg effect" to identify high-risk breast cancer patients: Stromal MCT4 predicts poor clinical outcome in triplenegative breast cancers. Cell Cycle, 2012, 11(6):1108–17., DOI 10.4161/cc.11. 6.19530 Riedl A, Schlederer M, Pudelko K, Stadler M, Walter S, Unterleuthner D, et al. Comparison of cancer cells in 2D vs 3D culture reveals differences in AKTmTOR- S6K signaling and drug responses. J Cel Sci, 2017, 130(1):203–18., DOI 10. 1242/jcs.188102 Dey M, Kim MH, Nagamine M, Karhan E, Kozhaya L, Dogan M, et al. Biofabrication of 3D breast cancer models for dissecting the cytotoxic response of human T cells expressing engineered MAIT cell receptors. Biofabrication, 2022, 14(4):044105., DOI 10.1088/1758-5090/ac925a Simon KA, Park KM, Mosadegh B, Subramaniam AB, Mazzeo AD, Ngo PM, et al. Polymer-based mesh as supports for multi-layered 3D cell culture and assays. Biomaterials, 2014, 35(1):259–68., DOI 10.1016/j.biomaterials.2013.09.049 Peela N, Sam FS, Christenson W, Truong D, Watson AW, Mouneimne G, et al. A three dimensional micropatterned tumor model for breast cancer cell migration studies. Biomaterials, 2016, 81:72–83., DOI 10.1016/j.biomaterials.2015. 11.039 Schmid R, Schmidt SK, Hazur J, Detsch R, Maurer E, Boccaccini AR, et al. Comparison of hydrogels for the development of well-defined 3D cancer models of breast cancer and melanoma. Cancers, Basel,, 2020, 12(8):2320., DOI 10.3390/ cancers12082320 Zhu W, Castro NJ, Cui H, Zhou X, Boualam B, McGrane R, et al. A 3D printed nano bone matrix for characterization of breast cancer cell and osteoblast interactions. Nanotechnology, 2016, 27(31):315103., DOI 10.1088/0957-4484/27/31/315103 Nam KH, Jeong CB, Kim H, Ahn M, Ahn SJ, Hur H, et al. Quantitative photothermal characterization with bioprinted 3D complex tissue constructs for early-stage breast cancer therapy using gold nanorods. Adv Healthc Mater, 2021, 10(18):e2100636., DOI 10.1002/adhm.202100636 Nanou A, Lorenzo-Moldero I, Gazouleas KD, Cortese B, Moroni L. 3D culture modeling of metastatic breast cancer cells in additive manufactured scaffolds. ACS Appl Mater Inter, 2022, 14(24):28389–402., DOI 10.1021/acsami. 2c07492 Xie M, Gao Q, Fu J, Chen Z, He Y. Bioprinting of novel 3D tumor array chip for drug screening. Bio-Design andManufacturing, 2020, 3(3):175–88., DOI 10.1007/ s42242-020-00078-4 Jung M, Skhinas JN, Du EY, Tolentino MAK, Utama RH, Engel M, et al. A high-throughput 3D bioprinted cancer cell migration and invasion model with versatile and broad biological applicability. Biomater Sci, 2022, 10(20):5876–87., DOI 10.1039/D2BM00651K Engel M, Belfiore L, Aghaei B, Sutija M. Enabling high throughput drug discovery in 3D cell cultures through a novel bioprinting workflow. SLAS Technology, 2022, 27(1):32–8., DOI 10.1016/j.slast.2021.10.002 Campbell A, Mohl JE, Gutierrez DA, Varela-Ramirez A, Boland T. Thermal bioprinting causes ample alterations of expression of LUCAT1, IL6, CCL26, and NRN1L genes and massive phosphorylation of critical oncogenic drug resistance pathways in breast cancer cells. Front Bioeng Biotechnol, 2020, 8:82., DOI 10.3389/ fbioe.2020.00082 Flores-Torres S, Peza-Chavez O, Kuasne H, Munguia-Lopez JG, Kort- Mascort J, Ferri L, et al. Alginate-gelatin-Matrigel hydrogels enable the development and multigenerational passaging of patient-derived 3D bioprinted cancer spheroid models. Biofabrication, 2021, 13(2):025001., DOI 10.1088/1758- 5090/abdb87 Campbell A, Gutierrez DA, Knight C, Vines CM, Heydarian R, Philipovskiy A, et al. Novel combinatorial strategy using thermal inkjet bioprinting, chemotherapy, and radiation on human breast cancer cells; an in-vitro cell viability assessment. Materials, Basel,, 2021, 14(24):7864., DOI 10.3390/ ma14247864 Blanco-Fernandez B, Rey-Vinolas S, Bagci G, Rubi-Sans G, Otero J, Navajas D, et al. Bioprinting decellularized breast tissue for the development of threedimensional breast cancer models. ACS Appl Mater Inter, 2022, 14(26):29467–82., DOI 10.1021/acsami.2c00920 Hong S, Song JM. 3D bioprinted drug-resistant breast cancer spheroids for quantitative in situ evaluation of drug resistance. Acta Biomater, 2022, 138:228–39., DOI 10.1016/j.actbio.2021.10.031 Chen Y, Xu L, Li W, Chen W, He Q, Zhang X, et al. 3D bioprinted tumor model with extracellular matrix enhanced bioinks for nanoparticle evaluation. Biofabrication, 2022, 14(2):025002., DOI 10.1088/1758-5090/ac48e4 Axpe E, Oyen ML. Applications of alginate-based bioinks in 3D bioprinting. Int J Mol Sci, 2016, 17(12):1976., DOI 10.3390/ijms17121976 Qiao SP, Zhao YF, Li CF, Yin YB, Meng QY, Lin FH, et al. An alginate-based platform for cancer stem cell research. Acta Biomater, 2016, 37:83–92., DOI 10.1016/ j.actbio.2016.04.032 Reig-Vano B, Tylkowski B, Montane X, Giamberini M. Alginate-based hydrogels for cancer therapy and research. Int J Biol Macromol, 2021, 170:424–36., DOI 10.1016/j.ijbiomac.2020.12.161 Jiang T,Munguia-Lopez JG, Gu K, BavouxMM, Flores-Torres S, Kort-Mascort J, et al. Engineering bioprintable alginate/gelatin composite hydrogels with tunable mechanical and cell adhesive properties to modulate tumor spheroid growth kinetics. Biofabrication, 2019, 12(1):015024., DOI 10.1088/1758-5090/ab3a5c He H, Li D, Lin Z, Peng L, Yang J, Wu M, et al. Temperatureprogrammable and enzymatically solidifiable gelatin-based bioinks enable facile extrusion bioprinting. Biofabrication, 2020, 12(4):045003., DOI 10.1088/ 1758-5090/ab9906 Dai X, Ma C, Lan Q, Xu T. 3D bioprinted glioma stem cells for brain tumor model and applications of drug susceptibility. Biofabrication, 2016, 8(4):045005., DOI 10.1088/1758-5090/8/4/045005 Piras CC, Fernandez-Prieto S, De Borggraeve WM. Nanocellulosic materials as bioinks for 3D bioprinting. Biomater Sci, 2017, 5(10):1988–92., DOI 10.1039/ c7bm00510e Wang X, Wang Q, Xu C. Nanocellulose-based inks for 3D bioprinting: Key aspects in research development and challenging perspectives in applications-A mini review. Bioengineering, Basel,, 2020, 7(2):40., DOI 10.3390/ bioengineering7020040 Kim J, Jang J, Cho DW. Controlling cancer cell behavior by improving the stiffness of gastric tissue-decellularized ECM bioink with cellulose nanoparticles. Front Bioeng Biotechnol, 2021, 9:605819., DOI 10.3389/fbioe. 2021.605819 De Stefano P, Briatico-Vangosa F, Bianchi E, Pellegata AF, Hartung de Hartungen A, Corti P, et al. Bioprinting of matrigel scaffolds for cancer research. Polymers, Basel,, 2021, 13(12):2026., DOI 10.3390/polym13122026 Fan R, Piou M, Darling E, Cormier D, Sun J, Wan J. Bio-printing cell-laden Matrigel-agarose constructs. J Biomater Appl, 2016, 31(5):684–92., DOI 10.1177/ 0885328216669238 Snyder JE, Hamid Q, Wang C, Chang R, Emami K,Wu H, et al. Bioprinting cell-laden matrigel for radioprotection study of liver by pro-drug conversion in a dual-tissue microfluidic chip. Biofabrication, 2011, 3(3):034112., DOI 10.1088/1758- 5082/3/3/034112 Osidak EO, Kozhukhov VI, Osidak MS, Domogatsky SP. Collagen as bioink for bioprinting: A comprehensive review. Int J Bioprint, 2020, 6(3):270., DOI 10. 18063/ijb.v6i3.270 Marques CF, Diogo GS, Pina S, Oliveira JM, Silva TH, Reis RL. Collagenbased bioinks for hard tissue engineering applications: A comprehensive review. J Mater Sci Mater Med, 2019, 30(3):32., DOI 10.1007/s10856-019- 6234-x Duarte Campos DF, Bonnin Marquez A, O’Seanain C, Fischer H, Blaeser A, Vogt M, et al. Exploring cancer cell behavior in vitro in three-dimensional multicellular bioprintable collagen-based hydrogels. Cancers, Basel,, 2019, 11(2):180., DOI 10.3390/cancers11020180 Noh I, Kim N, Tran HN, Lee J, Lee C. 3D printable hyaluronic acid-based hydrogel for its potential application as a bioink in tissue engineering. Biomater Res, 2019, 23:3., DOI 10.1186/s40824-018-0152-8 Petta D, D’Amora U, Ambrosio L, Grijpma DW, Eglin D, D’Este M. Hyaluronic acid as a bioink for extrusion-based 3D printing. Biofabrication, 2020, 12(3):032001., DOI 10.1088/1758-5090/ab8752 Lopez-Marcial GR, Zeng AY, Osuna C, Dennis J, Garcia JM, O’Connell GD. Agarose-based hydrogels as suitable bioprinting materials for tissue engineering. ACS Biomater Sci Eng, 2018, 4(10):3610–6., DOI 10.1021/acsbiomaterials.8b00903 Abelseth E, Abelseth L, De la Vega L, Beyer ST,Wadsworth SJ, Willerth SM. 3D printing of neural tissues derived from human induced pluripotent stem cells using a fibrin-based bioink. ACS Biomater Sci Eng, 2019, 5(1):234–43., DOI 10.1021/ acsbiomaterials.8b01235 Sharma R, Smits IPM, De La Vega L, Lee C, Willerth SM. 3D bioprinting pluripotent stem cell derived neural tissues using a novel fibrin bioink containing drug releasing microspheres. Front Bioeng Biotechnol, 2020, 8:57., DOI 10.3389/ fbioe.2020.00057 Roth AD, Lama P, Dunn S, Hong S, Lee MY. Polymer coating on a micropillar chip for robust attachment of PuraMatrix peptide hydrogel for 3D hepatic cell culture. Mater Sci Eng CMater Biol Appl, 2018, 90:634–44., DOI 10.1016/ j.msec.2018.04.092 Yang Z, Xu H, Zhao X. Designer self-assembling peptide hydrogels to engineer 3D cell microenvironments for cell constructs formation and precise oncology remodeling in ovarian cancer. Adv Sci, Weinh,, 2020, 7(9):1903718., DOI 10.1002/advs.201903718 Pati F, Jang J, Ha DH, Won Kim S, Rhie JW, Shim JH, et al. Printing threedimensional tissue analogues with decellularized extracellular matrix bioink. Nat Commun, 2014, 5:3935., DOI 10.1038/ncomms4935 Ferreira LP, Gaspar VM, Mano JF. Decellularized extracellular matrix for bioengineering physiomimetic 3D in vitro tumor models. Trends Biotechnol, 2020, 38(12):1397–414., DOI 10.1016/j.tibtech.2020.04.006 Jang J, Park HJ, KimSW, KimH, Park JY,Na SJ, et al. 3Dprinted complex tissue construct using stem cell-laden decellularized extracellular matrix bioinks for cardiac repair. Biomaterials, 2017, 112:264–74., DOI 10.1016/j.biomaterials.2016.10.026 Yue K, Trujillo-de Santiago G, Alvarez MM, Tamayol A, Annabi N, Khademhosseini A. Synthesis, properties, and biomedical applications of gelatin methacryloyl, GelMA, hydrogels. Biomaterials, 2015, 73:254–71., DOI 10.1016/j. biomaterials.2015.08.045 Ying G, Jiang N, Yu C, Zhang YS. Three-dimensional bioprinting of gelatin methacryloyl, GelMA). Bio-Design and Manufacturing, 2018, 1(4):215–24., DOI 10. 1007/s42242-018-0028-8 Rajabi N, Rezaei A, Kharaziha M, Bakhsheshi-Rad HR, Luo H, RamaKrishna S, et al. Recent advances on bioprinted gelatin methacrylate-based hydrogels for tissue repair. Tissue Eng Part A, 2021, 27(11-12):679–702., DOI 10.1089/ten.TEA.2020.0350 Yang X, Wang Y, Zhou Y, Chen J, Wan Q. The application of polycaprolactone in three-dimensional printing scaffolds for bone tissue engineering. Polymers, Basel,, 2021, 13(16):2754., DOI 10.3390/polym13162754 Gori M, Giannitelli SM, Torre M, Mozetic P, Abbruzzese F, Trombetta M, et al. Biofabrication of hepatic constructs by 3D bioprinting of a cell-laden thermogel: An effective tool to assess drug-induced hepatotoxic response. Adv Healthc Mater, 2020, 9(21):e2001163., DOI 10.1002/adhm.202001163 Klak M, Gomolka M, Dobrzanski T, Tymicki G, Cywoniuk P, Kowalska P, et al. Irradiation with 365 nm and 405 nm wavelength shows differences in DNA damage of swine pancreatic islets. PLoS One, 2020, 15(6):e0235052., DOI 10.1371/ journal.pone.0235052 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1610996 EP 1611017 DI 10.3389/pore.2023.1610996 PG 22 ER PT J AU Zhang, Y Li, Y Zuo, Z Li, T An, Y Zhang, W AF Zhang, Yu Li, Yan Zuo, Zan Li, Ting An, Ying Zhang, Wenjing TI An epithelial–mesenchymal transition-related mRNA signature associated with the prognosis, immune infiltration and therapeutic response of colon adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE epithelial-mesenchymal transition; bioinformatics; prognostic signature; tumor immune microenvironment; colon adenocarcinoma ID epithelial-mesenchymal transition; bioinformatics; prognostic signature; tumor immune microenvironment; colon adenocarcinoma AB Background: Epithelial-mesenchymal transition (EMT) is closely associated with cancer cell metastasis. Colon adenocarcinoma (COAD) is one of the most common malignancies in the world, and its metastasis leading to poor prognosis remains a challenge for clinicians. The purpose of this study was to explore the prognostic value of EMT-related genes (EMTRGs) by bioinformatics analysis and to develop a new EMTRGs prognostic signature for COAD. Methods: The TCGA-COAD dataset was downloaded from the TCGA portal as the training cohort, and the GSE17538 and GSE29621 datasets were obtained from the GEO database as the validation cohort. The best EMTRGs prognostic signature was constructed by differential expression analysis, Cox, and LASSO regression analysis. Gene set enrichment analysis (GSEA) is used to reveal pathways that are enriched in high-risk and low-risk groups. Differences in tumor immune cell levels were analyzed using microenvironmental cell population counter and single sample gene set enrichment analysis. Subclass mapping analysis and Genomics of Drug Sensitivity in Cancer were applied for prediction of immunotherapy response and chemotherapy response, respectively. Results: A total of 77 differentially expressed EMTRGs were identified in the TCGA-COAD cohort, and they were significantly associated with functions and pathways related to cancer cell metastasis, proliferation, and apoptosis. We constructed EMTRGs prognostic signature with COMP, MYL9, PCOLCE2, SCG2, and TIMP1 as new COAD prognostic biomarkers. The high-risk group had a poorer prognosis with enhanced immune cell infiltration. The GSEA demonstrated that the high-risk group was involved in “ECM Receptor Interaction,” “WNT Signaling Pathway” and “Colorectal Cancer.” Furthermore, patients with high risk scores may respond to anti-CTLA4 therapy and may be more resistant to targeted therapy agents BI 2536 and ABT-888. Conclusion: Together, we developed a new EMTRGs prognostic signature that can be an independent prognostic factor for COAD. This study has guiding implications for individualized counseling and treatment of COAD patients. C1 [Zhang, Yu] First People’s Hospital of Yunnan Province, Department of GastroenterologyKunming, China. [Li, Yan] First People’s Hospital of Yunnan Province, Department of GastroenterologyKunming, China. [Zuo, Zan] First People’s Hospital of Yunnan Province, Department of GastroenterologyKunming, China. [Li, Ting] First People’s Hospital of Yunnan Province, Department of GastroenterologyKunming, China. [An, Ying] First People’s Hospital of Yunnan Province, Department of GastroenterologyKunming, China. [Zhang, Wenjing] Kunming University of Science and Technology, Faculty of MedicineKunming, China. RP Zhang, W (reprint author), Kunming University of Science and Technology, Faculty of Medicine, Kunming, China. EM wenjing_zhang1@163.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Shi JF, Wang L, Ran JC, Wang H, Liu CC, Zhang HZ, et al. Clinical characteristics, medical service utilization, and expenditure for colorectal cancer in China, 2005 to 2014: Overall design and results from a multicenter retrospective epidemiologic survey. Cancer, 2021, 127(11):1880–93., DOI 10.1002/cncr.33445 Ganesh K, Massague J. Targeting metastatic cancer. Nat Med, 2021, 27(1): 34–44., DOI 10.1038/s41591-020-01195-4 Derynck R, Weinberg RA. EMT and cancer: More than meets the eye. Dev Cel, 2019, 49(3):313–6., DOI 10.1016/j.devcel.2019.04.026 Dongre A, Weinberg RA. New insights into the mechanisms of epithelialmesenchymal transition and implications for cancer. Nat Rev Mol Cel Biol, 2019, 20(2):69–84., DOI 10.1038/s41580-018-0080-4 Bakir B, Chiarella AM, Pitarresi JR, Rustgi AK. EMT,MET, plasticity, and tumor metastasis. Trends Cel Biol, 2020, 30(10):764–76., DOI 10.1016/j.tcb.2020.07.003 Wang J, Cai H, Liu Q, Xia Y, Xing L, Zuo Q, et al. Cinobufacini inhibits colon cancer invasion and metastasis via suppressing wnt/β-catenin signaling pathway and EMT. Am J Chin Med, 2020, 48(3):703–18., DOI 10.1142/S0192415X20500354 Wen J, Min X, Shen M, Hua Q, Han Y, Zhao L, et al. ACLY facilitates colon cancer cell metastasis by CTNNB1. J Exp Clin Cancer Res, 2019, 38(1):401., DOI 10. 1186/s13046-019-1391-9 Cai L, Hu C, Yu S, Liu L, Zhao J, Zhao Y, et al. Identification of EMT-related gene signatures to predict the prognosis of patients with endometrial cancer. Front Genet, 2020, 11:582274., DOI 10.3389/fgene.2020.582274 Tong H, Li T, Gao S, Yin H, Cao H, He W. An epithelial-mesenchymal transition-related long noncoding RNA signature correlates with the prognosis and progression in patients with bladder cancer. Biosci Rep, 2021, 41(1)., DOI 10.1042/ BSR20203944 Xu F, Tang Q,Wang Y, Qian K, Ju L, Xiao Y Development and validation of a six-gene prognostic signature for bladder cancer. Front Genet, 2021, 12:758612., DOI 10.3389/fgene.2021.758612 Xu BH, Jiang JH, Luo T, Jiang ZJ, Liu XY, Li LQ. Signature of prognostic epithelial-mesenchymal transition related long noncoding RNAs, ERLs, in hepatocellular carcinoma. Medicine, Baltimore,, 2021, 100(30):e26762., DOI 10. 1097/MD.0000000000026762 Zhang Z, Gu J, Yin M, Wang D, Ma C, Du J, et al. Establishment and investigation of a multiple gene expression signature to predict long-term survival in pancreatic cancer. Biomed Res Int, 2020, 2020:1570862., DOI 10.1155/2020/ 1570862 Xue K, Zheng H, Qian X, Chen Z, Gu Y, Hu Z, et al. Identification of key mRNAs as prediction models for early metastasis of pancreatic cancer based on LASSO. Front Bioeng Biotechnol, 2021, 9:701039., DOI 10.3389/fbioe.2021.701039 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A, 2005, 102(43): 15545–50., DOI 10.1073/pnas.0506580102 Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, Kim H, Torres- Garcia W, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun, 2013, 4:2612., DOI 10.1038/ncomms3612 Becht E, Giraldo NA, Lacroix L, Buttard B, Elarouci N, Petitprez F, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol, 2016, 17(1):218., DOI 10.1186/ s13059-016-1070-5 Finotello F, Trajanoski Z. Quantifying tumor-infiltrating immune cells from transcriptomics data. Cancer Immunol Immunother, 2018, 67(7):1031–40., DOI 10. 1007/s00262-018-2150-z Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity, 2018, 48(4):812–30.e14., DOI 10.1016/ j.immuni.2018.03.023 Hoshida Y, Brunet JP, Tamayo P, Golub TR, Mesirov JP. Subclass mapping: identifying common subtypes in independent disease data sets. PLoS One, 2007, 2(11):e1195., DOI 10.1371/journal.pone.0001195 Geeleher P, Cox N, Huang RS. pRRophetic: an R package for prediction of clinical chemotherapeutic response from tumor gene expression levels. PLoS One, 2014, 9(9):e107468., DOI 10.1371/journal.pone.0107468 Zachou K, Gabeta S, Shums Z, Gatselis NK, Koukoulis GK, Norman GL, et al. COMP serum levels: A new non-invasive biomarker of liver fibrosis in patients with chronic viral hepatitis. Eur J Intern Med, 2017, 38:83–8., DOI 10.1016/j.ejim.2017. 01.007 Zhong W, Hou H, Liu T, Su S, Xi X, Liao Y, et al. Cartilage oligomeric matrix protein promotes epithelial-mesenchymal transition by interacting with transgelin in colorectal cancer. Theranostics, 2020, 10(19):8790–806., DOI 10.7150/thno.44456 Nfonsam VN, Nfonsam LE, Chen D, Omesiete PN, Cruz A, Runyan RB, et al. COMP gene coexpresses with EMT genes and is associated with poor survival in colon cancer patients. J Surg Res, 2019, 233:297–303., DOI 10.1016/j.jss.2018.08.021 Kimura MY, Koyama-Nasu R, Yagi R, Nakayama T. A new therapeutic target: the CD69-myl9 system in immune responses. Semin Immunopathol, 2019, 41(3): 349–58., DOI 10.1007/s00281-019-00734-7 Zhao B, Baloch Z, Ma Y, Wan Z, Huo Y, Li F, et al. Identification of potential key genes and pathways in early-onset colorectal cancer through bioinformatics analysis. Cancer Control, 2019, 26(1):1073274819831260., DOI 10.1177/ 1073274819831260 Qiu X, Cheng SH, Xu F, Yin JW, Wang LY, Zhang XY. Weighted gene coexpression network analysis identified MYL9 and CNN1 are associated with recurrence in colorectal cancer. J Cancer, 2020, 11(8):2348–59., DOI 10.7150/jca. 39723 Deng Y, Liu L, Feng W, Lin Z, Ning Y, Luo X. High expression of MYL9 indicates poor clinical prognosis of epithelial ovarian cancer. Recent Pat Anticancer Drug Discov, 2021, 16(4):533–9., DOI 10.2174/ 1574891X16666210706153740 Wang JH, Zhang L, Huang ST, Xu J, Zhou Y, Yu XJ, et al. Expression and prognostic significance ofMYL9 in esophageal squamous cell carcinoma. PLoS One, 2017, 12(4):e0175280., DOI 10.1371/journal.pone.0175280 Kruthika BS, Sugur H, Nandaki K, Arimappamagan A, Paturu K, Santosh V. Expression pattern and prognostic significance of myosin light chain 9, MYL9): a novel biomarker in glioblastoma. J Clin Pathol, 2019, 72(10):677–81., DOI 10.1136/ jclinpath-2019-205834 Huang YQ, Han ZD, Liang YX, Lin ZY, Ling XH, Fu X, et al. Decreased expression of myosin light chain MYL9 in stroma predicts malignant progression and poor biochemical recurrence-free survival in prostate cancer. Med Oncol, 2014, 31(1):820., DOI 10.1007/s12032-013-0820-4 Feng M, Dong N, Zhou X, Ma L, Xiang R. Myosin light chain 9 promotes the proliferation, invasion, migration and angiogenesis of colorectal cancer cells by binding to Yes-associated protein 1 and regulating Hippo signaling. Bioengineered, 2022, 13(1):96–106., DOI 10.1080/21655979.2021.2008641 Zhu K, Wang Y, Liu L, Li S, Yu W. Long non-coding RNA MBNL1-AS1 regulates proliferation, migration, and invasion of cancer stem cells in colon cancer by interacting with MYL9 via sponging microRNA-412-3p. Clin Res Hepatol Gastroenterol, 2020, 44(1):101–14., DOI 10.1016/j.clinre.2019.05.001 Shi C, Xie Y, Li X, Li G, LiuW, Pei W, et al. Identification of ferroptosis-related genes signature predicting the efficiency of invasion and metastasis ability in colon adenocarcinoma. Front Cel Dev Biol, 2021, 9:815104., DOI 10.3389/fcell.2021.815104 Liu J, Lan Y, Tian G, Yang J. A systematic framework for identifying prognostic genes in the tumor microenvironment of colon cancer. Front Oncol, 2022, 12:899156., DOI 10.3389/fonc.2022.899156 Yao H, Li C, Tan X. An age stratified analysis of the biomarkers in patients with colorectal cancer. Sci Rep, 2021, 11(1):22464., DOI 10.1038/s41598-021- 01850-x Xu H, Wan H, Zhu M, Feng L, Zhang H, Su F. Discovery and validation of an epithelial-mesenchymal transition-based signature in gastric cancer by Genomics and prognosis analysis. Biomed Res Int, 2021, 2021:9026918., DOI 10.1155/2021/ 9026918 Chen Q, Cai L, Liang J. Construction of prognosis model of bladder cancer based on transcriptome. J Zhejiang Da Xue Xue Bao Yi Xue Ban, 2021, 50(7):1–9. Tian S, Meng G, Zhang W. A six-mRNA prognostic model to predict survival in head and neck squamous cell carcinoma. Cancer Manag Res, 2019, 11:131–42., DOI 10.2147/CMAR.S185875 Luo Y, Chen R, Ning Z, Fu N, XieM. Identification of a four-gene signature for determining the prognosis of papillary thyroid carcinoma by integrated bioinformatics analysis. Int J Gen Med, 2022, 15:1147–60., DOI 10.2147/IJGM. S346058 Liu JW, Yu F, Tan YF, Huo JP, Liu Z, Wang XJ, et al. Profiling of tumor microenvironment components identifies five stroma-related genes with prognostic implications in colorectal cancer. Cancer Biother Radiopharm, 2022, 37:882–92., DOI 10.1089/cbr.2020.4118 Wang H, Yin J, Hong Y, Ren A, Li M, Zhao Q, et al. SCG2 is a prognostic biomarker associated with immune infiltration and macrophage polarization in colorectal cancer. Front Cel Dev Biol, 2021, 9:795133., DOI 10.3389/fcell.2021.795133 Wang C, Chen S, Li S, Mi H. A prognostic model for predicting tumor mutation burden and tumor-infiltrating immune cells in bladder urothelial carcinoma. Front Genet, 2022, 13:708003., DOI 10.3389/fgene.2022.708003 Luo Y, Chen L, Zhou Q, Xiong Y, Wang G, Liu X, et al. Identification of a prognostic gene signature based on an immunogenomic landscape analysis of bladder cancer. J Cel Mol Med, 2020, 24(22):13370–82., DOI 10.1111/jcmm.15960 Tao D, Wang Y, Zhang X, Wang C, Yang D, Chen J, et al. Identification of angiogenesis-related prognostic biomarkers associated with immune cell infiltration in breast cancer. Front Cel Dev Biol, 2022, 10:853324., DOI 10.3389/ fcell.2022.853324 Ma C, Li F, Wang Z, Luo H. A novel immune-related gene signature predicts prognosis of lung adenocarcinoma. Biomed Res Int, 2022, 2022:4995874., DOI 10. 1155/2022/4995874 Fang C, Dai L, Wang C, Fan C, Yu Y, Yang L, et al. Secretogranin II impairs tumor growth and angiogenesis by promoting degradation of hypoxia-inducible factor-1α in colorectal cancer. Mol Oncol, 2021, 15(12):3513–26., DOI 10.1002/1878- 0261.13044 Justo BL, Jasiulionis MG. Characteristics of TIMP1, CD63, and β1-integrin and the functional impact of their interaction in cancer. Int J Mol Sci, 2021, 22(17): 9319., DOI 10.3390/ijms22179319 Cabral-Pacheco GA, Garza-Veloz I, Castruita-De La Rosa C, Ramirez-Acuna JM, Perez-Romero BA, Guerrero-Rodriguez JF, et al. The roles of matrix metalloproteinases and their inhibitors in human diseases. Int J Mol Sci, 2020, 21(24):9739., DOI 10.3390/ijms21249739 Zeng C, Chen Y. HTR1D, TIMP1, SERPINE1, MMP3 and CNR2 affect the survival of patients with colon adenocarcinoma. Oncol Lett, 2019, 18(3):2448–54., DOI 10.3892/ol.2019.10545 Wu T, Dai Y. Tumor microenvironment and therapeutic response[J]. Cancer Lett, 2017, 387:61–8. Hui L, Chen Y. Tumor microenvironment: Sanctuary of the devil. Cancer Lett, 2015, 368(1):7–13., DOI 10.1016/j.canlet.2015.07.039 Taki M, Abiko K, Ukita M, Murakami R, Yamanoi K, Yamaguchi K, et al. Tumor immune microenvironment during epithelial-mesenchymal transition. Clin Cancer Res, 2021, 27(17):4669–79., DOI 10.1158/1078-0432.CCR-20-4459 Hanus M, Parada-Venegas D, Landskron G, Wielandt AM, Hurtado C, Alvarez K, et al. Immune system, microbiota, and microbial metabolites: The unresolved triad in colorectal cancer microenvironment. Front Immunol, 2021, 12: 612826., DOI 10.3389/fimmu.2021.612826 Rocca YS, Roberti MP, Arriaga JM, Amat M, Bruno L, Pampena MB, et al. Altered phenotype in peripheral blood and tumor-associated NK cells from colorectal cancer patients. Innate Immun, 2013, 19(1):76–85., DOI 10.1177/ 1753425912453187 Ren X, Chen X, Zhang X, Jiang S, Zhang T, Li G, et al. Immune microenvironment and response in prostate cancer using large population cohorts. Front Immunol, 2021, 12:686809., DOI 10.3389/fimmu.2021.686809 Lv D, Wu X, Chen X, Yang S, Wang M, Liu Y, et al. A novel immune-related gene-based prognostic signature to predict biochemical recurrence in patients with prostate cancer after radical prostatectomy. Cancer Immunol Immunother, 2021, 70(12):3587–602., DOI 10.1007/s00262-021-02923-6 Li F, Teng H, Liu M, Liu B, Zhang D, Xu Z, et al. Prognostic value of immunerelated genes in the tumor microenvironment of bladder cancer. Front Oncol, 2020, 10:1302., DOI 10.3389/fonc.2020.01302 Zhou Y, Bian S, Zhou X, Cui Y, Wang W, Wen L, et al. Single-cell multiomics sequencing reveals prevalent genomic alterations in tumor stromal cells of human colorectal cancer. Cancer Cell, 2020, 38(6):818–28.e5., DOI 10.1016/j.ccell.2020. 09.015 Gutteridge RE, Ndiaye MA, Liu X, Ahmad N. Plk1 inhibitors in cancer therapy: From laboratory to clinics. Mol Cancer Ther, 2016, 15(7):1427–35., DOI 10. 1158/1535-7163.MCT-15-0897 Wagner LM. Profile of veliparib and its potential in the treatment of solid tumors. Onco Targets Ther, 2015, 8:1931–9., DOI 10.2147/OTT.S69935 Solanes-Casado S, Cebrian A, Rodriguez-RemirezM,Mahillo I, Garcia-Garcia L, Rio-Vilarino A, et al. Overcoming PLK1 inhibitor resistance by targeting mevalonate pathway to impair AXL-TWIST axis in colorectal cancer. Biomed Pharmacother, 2021, 144:112347., DOI 10.1016/j.biopha.2021.112347 Lund-Andersen C, Patzke S, Nahse-Kumpf V, Syljuasen RG. PLK1-inhibition can cause radiosensitization or radioresistance dependent on the treatment schedule. Radiother Oncol, 2014, 110(2):355–61., DOI 10.1016/j.radonc.2013.12.014 Czito BG, Deming DA, JamesonGS,Mulcahy MF,Vaghefi H, Dudley MW, et al. Safety and tolerability of veliparib combined with capecitabine plus radiotherapy in patients with locally advanced rectal cancer: a phase 1b study. Lancet Gastroenterol Hepatol, 2017, 2(6):418–26., DOI 10.1016/S2468-1253(17)30012-2 Pishvaian MJ, Slack RS, JiangW, Hwang JJ, Hankin A, Dorsch-Vogel K, et al. A phase 2 study of the PARP inhibitor veliparib plus temozolomide in patients with heavily pretreated metastatic colorectal cancer. Cancer, 2018, 124(11):2337–46., DOI 10.1002/cncr.31309 Gorbunova V, Beck JT, Hofheinz RD, Garcia-Alfonso P, Nechaeva M, Cubillo Gracian A, et al. A phase 2 randomised study of veliparib plus FOLFIRI±bevacizumab versus placebo plus FOLFIRI±bevacizumab in metastatic colorectal cancer. Br J Cancer, 2019, 120(2):183–9., DOI 10.1038/s41416-018-0343-z Morano F, Raimondi A, Pagani F, Lonardi S, Salvatore L, Cremolini C, et al. Temozolomide followed by combination with low-dose ipilimumab and nivolumab in patients with microsatellite-stable, O6-methylguanine-DNA methyltransferasesilenced metastatic colorectal cancer: The MAYA trial. J Clin Oncol, 2022, 40(14): 1562–73., DOI 10.1200/JCO.21.02583 Parikh AR, Szabolcs A, Allen JN, Clark JW, Wo JY, Raabe M, et al. Radiation therapy enhances immunotherapy response in microsatellite stable colorectal and pancreatic adenocarcinoma in a phase II trial. Nat Cancer, 2021, 2(11):1124–35., DOI 10.1038/s43018-021-00269-7 Kanikarla Marie P, Haymaker C, Parra ER, Kim YU, Lazcano R, Gite S, et al. Pilot clinical trial of perioperative durvalumab and tremelimumab in the treatment of resectable colorectal cancer liver metastases. Clin Cancer Res, 2021, 27(11): 3039–49., DOI 10.1158/1078-0432.CCR-21-0163 Chung KY, Gore I, Fong L, Venook A, Beck SB, Dorazio P, et al. Phase II study of the anti-cytotoxic T-lymphocyte-associated antigen 4 monoclonal antibody, tremelimumab, in patients with refractory metastatic colorectal cancer. J Clin Oncol, 2010, 28(21):3485–90., DOI 10.1200/JCO.2010.28.3994 Huang X, Hou Y, Weng X, Pang W, Hou L, Liang Y, et al. Diethyldithiocarbamate-copper complex, CuET, inhibits colorectal cancer progression via miR-16-5p and 15b-5p/ALDH1A3/PKM2 axis-mediated aerobic glycolysis pathway. Oncogenesis, 2021, 10(1):4., DOI 10.1038/s41389-020-00295-7 Wu H, Wei M, Jiang X, Tan J, Xu W, Fan X, et al. lncRNA PVT1 promotes tumorigenesis of colorectal cancer by stabilizingmiR-16-5p and interacting with the VEGFA/VEGFR1/AKT Axis. Mol Ther Nucleic Acids, 2020, 20:438–50., DOI 10. 1016/j.omtn.2020.03.006 Yan S, Yue Y, Wang J, Li W, Sun M, Gu C, et al. LINC00668 promotes tumorigenesis and progression through sponging miR-188-5p and regulating USP47 in colorectal cancer. Eur J Pharmacol, 2019, 858:172464., DOI 10.1016/j. ejphar.2019.172464 Si M, Song Y, Wang X, Wang D, Liu X, Qu X, et al. CXCL12/CXCR7/β- arrestin1 biased signal promotes epithelial-to-mesenchymal transition of colorectal cancer by repressing miRNAs through YAP1 nuclear translocation. Cell Biosci, 2022, 12(1):171., DOI 10.1186/s13578-022-00908-1 Li N, Li J,Mi Q, Xie Y, Li P, Wang L, et al. Long non-coding RNA ADAMTS9- AS1 suppresses colorectal cancer by inhibiting the Wnt/β-catenin signalling pathway and is a potential diagnostic biomarker. J Cel Mol Med, 2020, 24(19): 11318–29., DOI 10.1111/jcmm.15713 Zhou J, Lin J, Zhang H, Zhu F, Xie R. LncRNA HAND2-AS1 sponging miR- 1275 suppresses colorectal cancer progression by upregulating KLF14. Biochem Biophys Res Commun, 2018, 503(3):1848–53., DOI 10.1016/j.bbrc.2018.07.125 Krattinger R, Bostrom A, Schioth HB, Thasler WE, Mwinyi J, Kullak-Ublick GA. microRNA-192 suppresses the expression of the farnesoid X receptor. Am J Physiol Gastrointest Liver Physiol, 2016, 310(11):G1044–51., DOI 10.1152/ajpgi. 00297.2015 Cui S, Yang X, Zhang L, Zhao Y, Yan W. LncRNA MAFG-AS1 promotes the progression of colorectal cancer by sponging miR-147b and activation of NDUFA4. Biochem Biophys Res Commun, 2018, 506(1):251–8., DOI 10.1016/j.bbrc.2018.10.112 Chen T, Huang B, Pan Y. Long non-coding RNA MAFG-AS1 promotes cell proliferation, migration, and EMT by miR-3196/STRN4 in drug-resistant cells of liver cancer. Front Cel Dev Biol, 2021, 9:688603., DOI 10.3389/fcell.2021.688603 Sun W, NieW,Wang Z, Zhang H, Li Y, Fang X. Lnc HAGLR promotes colon cancer progression through sponging miR-185-5p and activating CDK4 and CDK6 in vitro and in vivo. Onco Targets Ther, 2020, 13:5913–25., DOI 10.2147/ OTT.S246092 Yang Y, Feng M, Bai L, Liao W, Zhou K, Zhang M, et al. Comprehensive analysis of EMT-related genes and lncRNAs in the prognosis, immunity, and drug treatment of colorectal cancer. J Transl Med, 2021, 19(1):391., DOI 10.1186/s12967- 021-03065-0 Liu H, Tian Y, Li J, Zhang G, Liu Q, Yang M, et al. Identification and functional analysis of lncRNAs and mRNAs between tumorigenesis and metastasis in CRC. J Aging, Albany Ny,, 2021, 13(24):25859–85., DOI 10.18632/aging.203775 Wang K, Song K, Ma Z, Yao Y, Liu C, Yang J, et al. Identification of EMTrelated high-risk stage II colorectal cancer and characterisation ofmetastasis-related genes. Br J Cancer, 2020, 123(3):410–7., DOI 10.1038/s41416-020-0902-y Wang L, Liu D, Liu S, Liao T, Jiao Y, Jiang X, et al. Identification of IMPA2 as the hub gene associated with colorectal cancer and liver metastasis by integrated bioinformatics analysis. Transl Oncol, 2022, 21:101435., DOI 10.1016/j.tranon.2022. 101435 Zhang Z, Zheng S, Lin Y, Sun J, Ding N, Chen J, et al. Genomics and prognosis analysis of epithelial-mesenchymal transition in colorectal cancer patients. BMC Cancer, 2020, 20(1):1135., DOI 10.1186/s12885-020-07615-5 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1611016 EP 1611029 DI 10.3389/pore.2023.1611016 PG 14 ER PT J AU Zheng, Y Li, Z Wang, Y Chen, W Lin, Y Guo, J Ye, G AF Zheng, Ying Li, Zhe Wang, Yao Chen, Wanjiao Lin, Yifan Guo, Junming Ye, Guoliang TI CircRNA: A new class of targets for gastric cancer drug resistance therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE gastric cancer; drug resistance; circular RNA; therapeutic target; biogenesis ID gastric cancer; drug resistance; circular RNA; therapeutic target; biogenesis AB Gastric cancer (GC) is one of the most common malignancies worldwide. Patients with advanced GC need palliative care to ensure survival. This includes the use of chemotherapy agents, such as cisplatin, 5- fluorouracil, oxaliplatin, paclitaxel, and pemetrexed, as well as targeted agents. However, the emergence of drug resistance evidence in poor patient outcomes and poor prognosis is a motivation to determine the specific mechanism of drug resistance. Interestingly, circular RNAs (circRNAs) play an important part in the carcinogenesis and progression of GC and are involved in GC drug resistance. This review systematically summarizes the functions and mechanisms of circRNAs underlying GC drug resistance, especially chemoresistance. It also emphasizes that circRNAs can serve as promising targets for improving drug resistance and therapeutic efficacy. C1 [Zheng, Ying] Ningbo University School of Medicine, Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of PathophysiologyNingbo, China. [Li, Zhe] The Affiliated Hospital of Medical School of Ningbo University, Department of GastroenterologyNingbo, China. [Wang, Yao] Ningbo University School of Medicine, Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of PathophysiologyNingbo, China. [Chen, Wanjiao] The Affiliated Hospital of Medical School of Ningbo University, Department of GastroenterologyNingbo, China. [Lin, Yifan] The Affiliated Hospital of Medical School of Ningbo University, Department of GastroenterologyNingbo, China. [Guo, Junming] Ningbo University School of Medicine, Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of PathophysiologyNingbo, China. [Ye, Guoliang] The Affiliated Hospital of Medical School of Ningbo University, Department of GastroenterologyNingbo, China. RP Ye, G (reprint author), The Affiliated Hospital of Medical School of Ningbo University, Department of Gastroenterology, Ningbo, China. CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Lu T, Sun L, Zhu X. Yes-associated protein enhances proliferation and attenuates sensitivity to cisplatin in human gastric cancer cells. Biomed Pharmacother, 2018, 105:1269–75., DOI 10.1016/j.biopha.2018.06.031 McDonald J, Swami N, Hannon B, Lo C, Pope A, Oza A, et al. Impact of early palliative care on caregivers of patients with advanced cancer: Cluster randomised trial. Ann Oncol, 2017, 28:163–8., DOI 10.1093/annonc/mdw438 Japanese Gastric Cancer Treatment Guidelines 2021. Japanese gastric cancer treatment guidelines 2021, 6th edition). Gastric Cancer, 2023, 26:1–25., DOI 10. 1007/s10120-022-01331-8 Mokdad AA, Yopp AC, Polanco PM, Mansour JC, Reznik SI, Heitjan DF, et al. Adjuvant chemotherapy vs postoperative observation following preoperative chemoradiotherapy and resection in gastroesophageal cancer: A propensity score-matched analysis. JAMA Oncol, 2018, 4:31–8., DOI 10.1001/jamaoncol. 2017.2805 Naitoh H, Yamamoto H, Murata S, Kobayashi H, Inoue K, Tani T. Stratified phase II trial to establish the usefulness of the collagen gel droplet embedded culture-drug sensitivity test, CD-DST, for advanced gastric cancer. Gastric Cancer, 2014, 17:630–7., DOI 10.1007/s10120-013-0320-4 Lee J, Kim ST, Kim K, Lee H, Kozarewa I,Mortimer PGS, et al. Tumor genomic profiling guides patients with metastatic gastric cancer to targeted treatment: The VIKTORY umbrella trial.Cancer Discov, 2019, 9:1388–405., DOI 10.1158/2159-8290. CD-19-0442 Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J, et al. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA, 2013, 19: 141–57., DOI 10.1261/rna.035667.112 Huang X, Li Z, Zhang Q, Wang W, Li B, Wang L, et al. Circular RNA AKT3 upregulates PIK3R1 to enhance cisplatin resistance in gastric cancer viamiR- 198 suppression. Mol Cancer, 2019, 18:71., DOI 10.1186/s12943-019-0969-3 Sanger HL, Klotz G, Riesner D, Gross HJ, Kleinschmidt AK. Viroids are single-stranded covalently closed circular RNA molecules existing as highly basepaired rod-like structures. Proc Natl Acad Sci U S A, 1976, 73:3852–6., DOI 10.1073/ pnas.73.11.3852 Cocquerelle C, Mascrez B, Hetuin D, Bailleul B. Mis-splicing yields circular RNA molecules. Faseb j, 1993, 7:155–60., DOI 10.1096/fasebj.7.1.7678559 Zheng W, Su H, Lv X, Xin S, Xu T. Exon-intron circular RNA circRNF217 promotes innate immunity and antibacterial activity in teleost fish by reducing miR-130-3p function. J Immunol, 2022, 208:1099–114., DOI 10.4049/ jimmunol.2100890 Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, et al. Circular intronic long noncoding RNAs. Mol Cel, 2013, 51:792–806., DOI 10.1016/j.molcel. 2013.08.017 Kristensen LS, AndersenMS, Stagsted LVW, Ebbesen KK, Hansen TB, Kjems J. The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet, 2019, 20:675–91., DOI 10.1038/s41576-019-0158-7 Misir S, Wu N, Yang BB. Specific expression and functions of circular RNAs. Cell Death Differ, 2022, 29:481–91., DOI 10.1038/s41418-022-00948-7 Conn SJ, Pillman KA, Toubia J, Conn VM, Salmanidis M, Phillips CA, et al. The RNA binding protein quaking regulates formation of circRNAs. Cell, 2015, 160:1125–34., DOI 10.1016/j.cell.2015.02.014 Deng P, Sun M, Zhao WY, Hou B, Li K, Zhang T, et al. Circular RNA circVAPA promotes chemotherapy drug resistance in gastric cancer progression by regulating miR-125b-5p/STAT3 axis. World J Gastroenterol, 2021, 27:487–500., DOI 10.3748/wjg.v27.i6.487 Yu CY, Li TC, Wu YY, Yeh CH, Chiang W, Chuang CY, et al. The circular RNA circBIRC6 participates in the molecular circuitry controlling human pluripotency. Nat Commun, 2017, 8:1149., DOI 10.1038/s41467-017-01216-w Li J, SangM, Zheng Y, Meng L, Gu Z, Li Z, et al. HNRNPUL1 inhibits cisplatin sensitivity of esophageal squamous cell carcinoma through regulating the formation of circMAN1A2. Exp Cel Res, 2021, 409:112891., DOI 10.1016/j.yexcr.2021.112891 Li X, Yang L, Chen LL. The biogenesis, functions, and challenges of circular RNAs. Mol Cel, 2018, 71:428–42., DOI 10.1016/j.molcel.2018.06.034 Zhang W, Chen S, Du Q, Bian P, Chen Y, Liu Z, et al. CircVPS13C promotes pituitary adenoma growth by decreasing the stability of IFITM1 mRNA via interacting with RRBP1. Oncogene, 2022, 41:1550–62., DOI 10.1038/s41388-022- 02186-0 Wu N, Yuan Z, Du KY, Fang L, Lyu J, Zhang C, et al. Translation of yesassociated protein, YAP, was antagonized by its circular RNA via suppressing the assembly of the translation initiation machinery. Cel Death Differ, 2019, 26: 2758–73., DOI 10.1038/s41418-019-0337-2 Chen CK, Cheng R, Demeter J, Chen J, Weingarten-Gabbay S, Jiang L, et al. Structured elements drive extensive circular RNA translation. Mol Cel, 2021, 81: 4300–18.e13., DOI 10.1016/j.molcel.2021.07.042 Legnini I, Di Timoteo G, Rossi F, Morlando M, Briganti F, Sthandier O, et al. Circ-ZNF609 is a circular RNA that can Be translated and functions in myogenesis. Mol Cel, 2017, 66:22–37.e9., DOI 10.1016/j.molcel.2017.02.017 Meyer KD, Patil DP, Zhou J, Zinoviev A, Skabkin MA, Elemento O, et al. 5’ UTR m(6)A promotes cap-independent translation. Cell, 2015, 163:999–1010., DOI 10.1016/j.cell.2015.10.012 Wang T, Liu Z, She Y, Deng J, Zhong Y, Zhao M, et al. A novel protein encoded by circASK1 ameliorates gefitinib resistance in lung adenocarcinoma by competitively activating ASK1-dependent apoptosis. J].Cancer Lett, 2021, 520: 321–31., DOI 10.1016/j.canlet.2021.08.007 Li J, Sun D, Pu W, Wang J, Peng Y. Circular RNAs in cancer: Biogenesis, function, and clinical significance. Trends Cancer, 2020, 6:319–36., DOI 10.1016/j. trecan.2020.01.012 Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol, 2015, 22:256–64., DOI 10.1038/nsmb.2959 Ashwal-Fluss R, Meyer M, Pamudurti NR, Ivanov A, Bartok O, Hanan M, et al. circRNA biogenesis competes with pre-mRNA splicing. Mol Cel, 2014, 56: 55–66., DOI 10.1016/j.molcel.2014.08.019 Brabec V, Kasparkova J. Modifications of DNA by platinum complexes. Relation to resistance of tumors to platinum antitumor drugs. Drug Resist Updat, 2005, 8:131–46., DOI 10.1016/j.drup.2005.04.006 Fang L, Lv J, Xuan Z, Li B, Li Z, He Z, et al. Circular CPM promotes chemoresistance of gastric cancer via activating PRKAA2-mediated autophagy. Clin Transl Med, 2022, 12:e708., DOI 10.1002/ctm2.708 Wu Q, Wang H, Liu L, Zhu K, Yu W, Guo J. Hsa_circ_0001546 acts as a miRNA-421 sponge to inhibit the chemoresistance of gastric cancer cells via ATM/ Chk2/p53-dependent pathway. Biochem Biophys Res Commun, 2020, 521:303–9., DOI 10.1016/j.bbrc.2019.10.117 Cai J, Chen Z,Wang J, Wang J, Chen X, Liang L, et al. circHECTD1 facilitates glutaminolysis to promote gastric cancer progression by targeting miR-1256 and activating β-catenin/c-Myc signaling. Cell Death Dis, 2019, 10:576., DOI 10.1038/ s41419-019-1814-8 Dong X, Wang Y, Zhou Y, Wen J, Wang S, Shen L. Aquaporin 3 facilitates chemoresistance in gastric cancer cells to cisplatin via autophagy. Cell Death Discov, 2016, 2:16087., DOI 10.1038/cddiscovery.2016.87 Lin XM, Li S, Zhou C, Li RZ, Wang H, Luo W, et al. Cisplatin induces chemoresistance through the PTGS2-mediated anti-apoptosis in gastric cancer. Int J Biochem Cel Biol, 2019, 116:105610., DOI 10.1016/j.biocel.2019.105610 Peng L, Sang H, Wei S, Li Y, Jin D, Zhu X, et al. circCUL2 regulates gastric cancer malignant transformation and cisplatin resistance by modulating autophagy activation via miR-142-3p/ROCK2. Mol Cancer, 2020, 19:156., DOI 10.1186/s12943- 020-01270-x Zhang Q, Miao Y, Fu Q, Hu H, Chen H, Zeng A, et al. CircRNACCDC66 regulates cisplatin resistance in gastric cancer via the miR- 618/BCL2 axis. Biochem Biophys Res Commun, 2020, 526:713–20., DOI 10.1016/j. bbrc.2020.03.156 Sun G, Li Z, He Z, Wang W, Wang S, Zhang X, et al. Circular RNA MCTP2 inhibits cisplatin resistance in gastric cancer by miR-99a-5p-mediated induction of MTMR3 expression. J Exp Clin Cancer Res, 2020, 39:246., DOI 10.1186/ s13046-020-01758-w Huang XX, Zhang Q, Hu H, Jin Y, Zeng AL, Xia YB, et al. A novel circular RNA circFN1 enhances cisplatin resistance in gastric cancer via sponging miR-182- 5p. J Cel Biochem, 2020, 122:1009–20., DOI 10.1002/jcb.29641 Sun Y, Ma J, Lin J, Sun D, Song P, Shi L, et al. Circular RNA circ_ ASAP2 regulates drug sensitivity and functional behaviors of cisplatin-resistant gastric cancer cells by the miR-330-3p/NT5E axis. Anticancer Drugs, 2021, 32: 950–61., DOI 10.1097/CAD.0000000000001087 Zhang J, Zha W, Qian C, Ding A, Mao Z. Circular RNA circ_ 0001017 sensitizes cisplatin-resistant gastric cancer cells to chemotherapy by the miR-543/PHLPP2 Axis. Biochem Genet, 2022, 60:558–75., DOI 10.1007/s10528-021- 10110-6 Liu Y, Xu J, JiangM, Ni L, Ling Y. CircRNA DONSON contributes to cisplatin resistance in gastric cancer cells by regulating miR-802/BMI1 axis. Cancer Cel Int, 2020, 20:261., DOI 10.1186/s12935-020-01358-w Lugea A, Waldron RT. Exosome-mediated intercellular communication between stellate cells and cancer cells in pancreatic ductal adenocarcinoma. Pancreas, 2017, 46:1–4., DOI 10.1097/MPA.0000000000000686 Ashrafizaveh S, Ashrafizadeh M, Zarrabi A, Husmandi K, Zabolian A, Shahinozzaman M, et al. Long non-coding RNAs in the doxorubicin resistance of cancer cells. Cancer Lett, 2021, 508:104–14., DOI 10.1016/j.canlet.2021.03.018 Lasser C, Alikhani VS, Ekstrom K, Eldh M, Paredes PT, Bossios A, et al. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J Transl Med, 2011, 9:9., DOI 10.1186/1479-5876-9-9 Wang H, Zeng X, Zheng Y, Wang Y, Zhou Y. Exosomal circRNA in digestive system tumors: The main player or coadjuvants?[J]. Front Oncol, 2021, 11:614462., DOI 10.3389/fonc.2021.614462 Yao W, Guo P, Mu Q, Wang Y. Exosome-derived circ-PVT1 contributes to cisplatin resistance by regulating autophagy, invasion, and apoptosis via miR-30a- 5p/YAP1 Axis in gastric cancer cells. Cancer Biother Radiopharm, 2021, 36:347–59., DOI 10.1089/cbr.2020.3578 Yang X, Zhang Q, Guan B. Circ_0110805 knockdown enhances cisplatin sensitivity and inhibits gastric cancer progression by miR-299-3p/ENDOPDI Axis. Onco Targets Ther, 2020, 13:11445–57., DOI 10.2147/OTT.S279563 Xue M, Li G, Fang X, Wang L, Jin Y, Zhou Q. hsa_circ_0081143 promotes cisplatin resistance in gastric cancer by targeting miR-646/CDK6 pathway. Cancer Cel Int, 2019, 19:25., DOI 10.1186/s12935-019-0737-x Zhang Z, Yu X, Zhou B, Zhang J, Chang J. Circular RNA circ_ 0026359 enhances cisplatin resistance in gastric cancer via targeting miR-1200/ POLD4 pathway. Biomed Res Int, 2020, 2020:5103272., DOI 10.1155/2020/5103272 Liu S, Wu M, Peng M. Circ_0000260 regulates the development and deterioration of gastric adenocarcinoma with cisplatin resistance by upregulating MMP11 via targeting MiR-129-5p. Cancer Manag Res, 2020, 12:10505–19., DOI 10. 2147/CMAR.S272324 Xia Y, Lv J, Jiang T, Li B, Li Y, He Z, et al. CircFAM73A promotes the cancer stem cell-like properties of gastric cancer through the miR-490-3p/ HMGA2 positive feedback loop and HNRNPK-mediated β-catenin stabilization. J Exp Clin Cancer Res, 2021, 40:103., DOI 10.1186/s13046-021-01896-9 Xu B, Guo J, Chen M. Circ_0017274 acts on miR-637/CDX2 axis to facilitate cisplatin resistance in gastric cancer. Clin Exp Pharmacol Physiol, 2022, 49: 1105–15., DOI 10.1111/1440-1681.13692 Yang G, Tan J, Guo J, Wu Z, Zhan Q. Exosome-mediated transfer of circ_ 0063526 enhances cisplatin resistance in gastric cancer cells via regulating miR- 449a/SHMT2 axis. Anticancer Drugs, 2022, 33:1047–57., DOI 10.1097/CAD. 0000000000001386 Liang Q, Chu F, Zhang L, Jiang Y, Li L, Wu H. circ-LDLRAD3 knockdown reduces cisplatin chemoresistance and inhibits the development of gastric cancer with cisplatin resistance through miR-588 enrichment-mediated SOX5 inhibition. Gut Liver, 2022, [Epub ahead of print]., DOI 10.5009/gnl210195 Christensen S, Van der Roest B, Besselink N, Janssen R, Boymans S, Martens JWM, et al. 5-Fluorouracil treatment induces characteristic T>G mutations in human cancer. Nat Commun, 2019, 10:4571., DOI 10.1038/s41467-019-12594-8 Longley DB, Harkin DP, Johnston PG. 5-fluorouracil: Mechanisms of action and clinical strategies. Nat Rev Cancer, 2003, 3:330–8., DOI 10.1038/nrc1074 Xu G, Li M, Wu J, Qin C, Tao Y, He H. Circular RNA circNRIP1 sponges microRNA-138-5p to maintain hypoxia-induced resistance to 5-fluorouracil through HIF-1α-Dependent glucose metabolism in gastric carcinoma. Cancer Manag Res, 2020, 12:2789–802., DOI 10.2147/CMAR.S246272 Yang F, Yan Z, NieW, Cheng X, Liu Z, Wang W, et al. LACTB induced apoptosis of oxaliplatin-resistant gastric cancer through regulating autophagy-mediated mitochondrial apoptosis pathway. Am J Transl Res, 2021, 13:601–16. Gao H, Xu J, Qiao F, Xue L. Depletion of hsa_circ_0000144 suppresses oxaliplatin resistance of gastric cancer cells by regulating miR-502-5p/ ADAM9 Axis. Onco Targets Ther, 2021, 14:2773–87., DOI 10.2147/OTT.S281238 Zhong Y, Wang D, Ding Y, Tian G, Jiang B. Circular RNA circ_ 0032821 contributes to oxaliplatin, OXA, resistance of gastric cancer cells by regulating SOX9 via miR-515-5p. Biotechnol Lett, 2021, 43:339–51., DOI 10.1007/ s10529-020-03036-3 Yin S, Zeng C, Hari M, Cabral F. Paclitaxel resistance by random mutagenesis of α-tubulin. Cytoskeleton, Hoboken,, 2013, 70:849–62., DOI 10.1002/cm.21154 Sakamoto J,Matsui T, Kodera Y. Paclitaxel chemotherapy for the treatment of gastric cancer. Gastric Cancer, 2009, 12:69–78., DOI 10.1007/s10120-009-0505-z Wang M, Qiu R, Yu S, Xu X, Li G, Gu R, et al. Paclitaxel‑resistant gastric cancer MGC‑803 cells promote epithelial‑to‑mesenchymal transition and chemoresistance in paclitaxel‑sensitive cells via exosomal delivery of miR‑155‑5p. Int J Oncol, 2019, 54:326–38., DOI 10.3892/ijo.2018.4601 Liu YY, Zhang LY, Du WZ. Circular RNA circ-PVT1 contributes to paclitaxel resistance of gastric cancer cells through the regulation of ZEB1 expression by sponging miR-124-3p. Biosci Rep, 2019, 39(12):BSR20193045., DOI 10.1042/ BSR20193045 Zhou N, Wang W, Xu C, Yu W. Circular RNA PLEC acts as a sponge of microRNA-198 to promote gastric carcinoma cell resistance to paclitaxel and tumorigenesis. Pathol Res Pract, 2021, 224:153487., DOI 10.1016/j.prp.2021.153487 Celio L, Sternberg CN, Labianca R, La Torre I, Amoroso V, Barone C, et al. Pemetrexed in combination with oxaliplatin as a first-line therapy for advanced gastric cancer: A multi-institutional phase II study. Ann Oncol, 2009, 20:1062–7., DOI 10.1093/annonc/mdn766 Xu QY, Xie MJ, Huang J,Wang ZW. Effect of circ MTHFD2 on resistance to pemetrexed in gastric cancer through regulating expression of miR-124. Eur Rev Med Pharmacol Sci, 2019, 23:10290–9., DOI 10.26355/eurrev_201912_19667 Aoki M, Iwasa S, Boku N. Trastuzumab deruxtecan for the treatment of HER2-positive advanced gastric cancer: A clinical perspective. Gastric Cancer, 2021, 24:567–76., DOI 10.1007/s10120-021-01164-x Gravalos C, Jimeno A. HER2 in gastric cancer: A new prognostic factor and a novel therapeutic target. Ann Oncol, 2008, 19:1523–9., DOI 10.1093/annonc/ mdn169 Lv X, Li P,Wang J, Gao H, Hei Y, Zhang J, et al. hsa_circ_0000520 influences herceptin resistance in gastric cancer cells through PI3K-Akt signaling pathway. J Clin Lab Anal, 2020, 34:e23449., DOI 10.1002/jcla.23449 Zhao L, Peng Y, He S, Li R, Wang Z, Huang J, et al. Apatinib induced ferroptosis by lipid peroxidation in gastric cancer. Gastric Cancer, 2021, 24:642–54., DOI 10.1007/s10120-021-01159-8 Ma L, Wang Z, Xie M, Quan Y, Zhu W, Yang F, et al. Silencing of circRACGAP1 sensitizes gastric cancer cells to apatinib via modulating autophagy by targeting miR-3657 and ATG7. Cel Death Dis, 2020, 11:169., DOI 10.1038/s41419-020-2352-0 Wang JM, Ji LL, Branford-White CJ, Wang ZY, Shen KK, Liu H, et al. Antitumor activity of Dioscorea bulbifera L. rhizome in vivo. Fitoterapia, 2012, 83: 388–94., DOI 10.1016/j.fitote.2011.12.001 Lu Y, Li L, Li L, Wu G, Liu G. Circular RNA circHECTD1 prevents Diosbulbin-B-sensitivity via miR-137/PBX3 axis in gastric cancer. Cancer Cel Int, 2021, 21:264., DOI 10.1186/s12935-021-01957-1 Ma YY, Zhang Y, Mou XZ, Liu ZC, Ru GQ, Li E. High level of homeobox A9 and PBX homeobox 3 expression in gastric cancer correlates with poor prognosis. Oncol Lett, 2017, 14:5883–9., DOI 10.3892/ol.2017.6937 Cann C, Ciombor KK. Systemic therapy for gastric cancer: Perioperative strategies and beyond. J Surg Oncol, 2022, 125:1151–60., DOI 10.1002/jso.26834 Beilerli A, Gareev I, BeylerliO, Yang G, Pavlov V, Aliev G, et al. Circular RNAs as biomarkers and therapeutic targets in cancer. Semin Cancer Biol, 2022, 83: 242–52., DOI 10.1016/j.semcancer.2020.12.026 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1611033 EP 1611042 DI 10.3389/pore.2023.1611033 PG 10 ER PT J AU Wang, H Yang, L Li, Q Song, H Ji, H AF Wang, Hongyu Yang, Liqun Li, Qiuyao Song, Haiyun Ji, Hong TI Case report: Composite mantle cell lymphoma and classical Hodgkin lymphoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE next-generation sequencing; classical Hodgkin lymphoma; mantle cell lymphoma; composite lymphoma; histogenesis ID next-generation sequencing; classical Hodgkin lymphoma; mantle cell lymphoma; composite lymphoma; histogenesis AB Composite mantle cell lymphoma and classical Hodgkin lymphoma is very rare and the actual origin of it is still unclear. Here we reported a new case of composite mantle cell lymphoma and classical Hodgkin lymphoma and analyzed its molecular changes. Eight mutations were identified in its Hodgkin component through next-generation sequencing. In addition, we reviewed the published cases of composite mantle cell lymphoma and classical Hodgkin lymphoma and summarized the molecular changes of reported cases as well as the current case to explore the possible pathway of histogenesis. C1 [Wang, Hongyu] Shandong University, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Department of PathologyQingdao, China. [Yang, Liqun] Chengdu Huayin Medical Laboratory Center, Huayin Health Hematopathology Comprehensive Diagnostic CenterChengdu, China. [Li, Qiuyao] Shandong University, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Department of PathologyQingdao, China. [Song, Haiyun] Shandong University, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Department of PathologyQingdao, China. [Ji, Hong] Shandong University, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Department of PathologyQingdao, China. RP Ji, H (reprint author), Shandong University, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Department of Pathology, Qingdao, China. EM hezejihong@163.com CR Kuppers R, Duhrsen U, Hansmann ML. Pathogenesis, diagnosis and treatment of composite lymphomas. Lancet Oncol, 2014, 15:e435–46., DOI 10.1016/S1470-2045(14)70153-6 Caleo A, Sanchez-Aguilera A, Rodriguez S, Ana M, Beltran L, Larrinoa A, et al. Composite Hodgkin lymphoma and mantle cell lymphoma, two clonally unrelated tumors. Am J Surg Pathol, 2003, 27:1577–80., DOI 10.1097/00000478-200312000- 00012 Tinguely M, Rosenquist R, Sundstro¨m C, Amini R, Ku¨ppers R, Hansmann M, et al. Analysis of a clonally related mantle cell and Hodgkin lymphoma indicates Epstein–Barr virus infection of a Hodgkin/Reed–Sternberg cell precursor in a germinal center. Am J Surg Pathol, 2003, 27:1483–8., DOI 10.1097/00000478-200311000-00014 Hayes SJ, Banerjee SS, Cook Y, Houghton JB, Menasce LP. Composite mantlecell lymphoma and classical Hodgkin lymphoma. Histopathology, 2006, 48:621–3., DOI 10.1111/j.1365-2559.2005.02296.x Stefanie S, Barbara C, Markus S, Stefano A, Sylvia H, Martin-Leo H, et al. Subclonal evolution of a classical Hodgkin lymphoma from a germinal center B-cell-derived mantle cell lymphoma. Int J Cancer, 2014, 134:832–43., DOI 10.1002/ijc.28422 Giua R, Fontana D, Deda G, Bianchi A, Rabitti C, Incalzi R, et al. Composite mantle-cell lymphoma and classical Hodgkin lymphoma in a very old adult. J Am Geriatr Soc, 2015, 63:824–6., DOI 10.1111/jgs.13355 Ciara M, Fiona Q, Gyorgy I, Walker J, Castriciano G, O’Sullivan P, et al. Composite blastoid variant of mantle cell lymphoma and classical Hodgkin lymphoma. Int J Surg Pathol, 2017, 25:281–6., DOI 10.1177/1066896916672556 Sharma S, Singh V, Bisaria D, Tangri R. Composite lymphoma comprisingmantle cell lymphoma and epstein-barr virus positive classic Hodgkin lymphoma: A rare case. Indian J Pathol Microbiol, 2019, 62:488–90., DOI 10.4103/IJPM.IJPM_34_18 Liang G, Jing B, Weikai Y, Xiumei D, Zhuang T, Jingna G, et al. Composite mantle cell lymphoma and classical Hodgkin lymphoma: Report of a case. Chin J Pathol, 2019, 48:409–10., DOI 10.3760/cma.j.issn.0529-5807.2019.05.018 Hammad T, Kseniya PD, Connie LB,Maria EA,Mikhail R, Filiz S, et al. Divergent clonal evolution of a common precursor to mantle cell lymphoma and classic Hodgkin lymphoma. Cold Spring Harbor Mol Case Stud, 2019, 5:a004259., DOI 10.1101/mcs.a004259 Schmitz R, RenneC, Rosenquist R, TinguelyM,Distler V, Menestrina F, et al. Insights into themultistep transformation process of lymphomas: IgH-associated translocations and tumor suppressor gene mutations in clonally related composite hodgkin’s and nonhodgkin’s lymphomas. Leukemia, 2005, 19:1452–8., DOI 10.1038/sj.leu.2403841 Yu C, Yi L, Jiani X, Bin L, Xuefeng W, Jun L, et al. Role of PRKDC in cancer initiation, progression, and treatment. Cancer Cel Int, 2021, 21:563., DOI 10.1186/ s12935-021-02229-8 Keren X, Niquelle BW, Amie EH, David VC, Maryam S, Thomas MM, et al. Whole-exome sequencing in multiplex families to identify novel AYA classical Hodgkin lymphoma predisposition genes. Blood, 2021, 138(1):3499., DOI 10.1182/ blood-2021-153247 Priti A, Andrew BC, Srinivasan R, Nathanael JS, Ali A, Francisco CV, et al. ARID1B, a molecular suppressor of erythropoiesis, is essential for the prevention of Monge’s disease. Exp Mol Med, 2022, 54:777–87., DOI 10.1038/s12276-022-00769-1 Carmela G, Paola C, Rosa B, Pier PS, Khedoudja N, Laura JR, et al. Loss of the tumor suppressor PML in human cancers of multiple histologic origins. J Natl Cancer Inst, 2004, 96:269–79., DOI 10.1093/jnci/djh043 Xin X, Ting W, Weijie Z, Rosalind A, Ye L, Hui Z, et al. Down-regulation of cylindromatosis protein phosphorylation by BTK inhibitor promotes apoptosis of non-GCB-difuse large B-cell lymphoma. Cancer Cel Int, 2021, 21:1–14. Peter S, Rachel S, Arnold R. Inhibitor of apoptosis protein BIRC3, API2, cIAP2, AIP1, is upregulated by the non-canonical NFKB pathway. Cancer Res, 2007, 67(9):5327. Eugen T, Stephan S. BIRC3 mutations in chronic lymphocytic leukemia – uncommon and unfavorable. Haematologica, 2020, 105:255–6., DOI 10.3324/haematol.2019.238691 Qiong L, Wei Z, Jiali L, Jingkang X, Jia L, Ting C, et al. Plasma circulating tumor DNA assessment reveals KMT2D as a potential poor prognostic factor in extranodal NK/T-cell lymphoma. Biomarker Res, 2020, 8:27., DOI 10.1186/s40364- -00205-4 20. Simone F, Davide R, Andrea R, Alessio B, Valeria S, Christian WE, et al. KMT2D mutations and TP53 disruptions are poor prognostic biomarkers in mantle cell lymphoma receiving high-dose therapy: A FIL study. Haematologica, 2020, 105:1604–12., DOI 10.3324/haematol.2018.214056 Zizhen P, Yanyu G, Xiaoqiu L. Role of FAT1 in health and disease, Review). Oncol Lett, 2021, 21:398., DOI 10.3892/ol.2021.12659 Richard G, Lisa P, Bettina J, Carmen F, Lukas R, Olga S, et al. Sequential immunotherapy in a patient with primary refractory Hodgkin lymphoma and novel mutations. Oncotarget, 2018, 9:20928–40., DOI 10.18632/oncotarget.25037 Alexis T, Marie D, Juliette F, Herve G, Laurent J, Gabriel A, et al. Plasticity in classical Hodgkin composite lymphomas: A systematic review. Cancers, 2022, 14: 5695., DOI 10.3390/cancers14225695 Steven K, Guldeep U, ZiXuan W, Jerald Z. Mantle cell lymphoma with Hodgkin and reed-sternberg cells: Review with illustrative case. Appl Immunohistochem Mol Morphol, 2019, 27:8–14., DOI 10.1097/PAI. 0000000000000527 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1611051 EP 1611056 DI 10.3389/pore.2023.1611051 PG 6 ER PT J AU Chen, Y Ma, T AF Chen, Yan Ma, Tao TI LAMP5 may promote MM progression by activating p38 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apoptosis; recurrence; prognosis; multiple myeloma; LAMP5 ID apoptosis; recurrence; prognosis; multiple myeloma; LAMP5 AB Multiple myeloma (MM) is the second most common tumor of the hematologic system. MM remains incurable at this time. In this study, we used bioinformatics analysis to find key genes in the pathogenesis of MM. We first found that Lysosome associated membrane protein 5 (LAMP5) expression was sequentially increased in healthy donors (HD), monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM) and newly diagnosed MM (NDMM), relapsed MM (RMM). We collected bone marrow from patients with NDMM, HD and post-treatment MM (PTMM) and performed qPCR analysis of LAMP5, and found that the expression of LAMP5 is stronger in NDMM than in HD, and decreases after treatment. Western blotting assay also found more expression of LAMP5 in NDMM than in HD. Patients with high LAMP5 expression have a higher DS (Durie-Salmon) stage and worse prognosis. We next verified the expression of LAMP5 in four MM cell lines and silenced LAMP5 expression in RPMI-8226 and AMO-1, and explored the effects of LAMP5 silencing onMMcell apoptosis and cell cycle by flow cytometry and western blotting. Knockdown of LAMP5 promoted apoptosis in MM cells, but had no effect on the cell cycle. Mechanistically, LAMP5 may exert its pro-tumor effects in MM in part through activation of p38 protein. We screened LAMP5 for the first time as a key gene for MMprogression and recurrence, and found that LAMP5 may exert its pro-tumor effects in MM through activation of p38 protein. C1 [Chen, Yan] The Affiliated Hospital of Southwest Medical University, Department of HematologyLuzhou, China. [Ma, Tao] The Affiliated Hospital of Southwest Medical University, Department of HematologyLuzhou, China. RP Ma, T (reprint author), The Affiliated Hospital of Southwest Medical University, Department of Hematology, Luzhou, China. EM matao1990@swmu.edu.cn CR Das S, Juliana N, Yazit NAA, Azmani S, Abu IF. Multiple Myeloma: Challenges Encountered and Future Options for Better Treatment. Int J Mol Sci, 2022, 23(3): 1649., DOI 10.3390/ijms23031649 Maura F, Rustad EH, Boyle EM, Morgan GJ. Reconstructing the Evolutionary History of Multiple Myeloma. Best Pract Res Clin Haematol, 2020, 33(1):101145., DOI 10.1016/j.beha.2020.101145 Huang J, Chan SC, Lok V, Zhang L, Lucero-Prisno DE, 3rd, Xu W, et al. The Epidemiological Landscape of Multiple Myeloma: a Global Cancer Registry Estimate of Disease burden, Risk Factors, and Temporal Trends. Lancet Haematol, 2022, 9(9):e670–e677., DOI 10.1016/S2352-3026(22)00165-X van de Donk N, Pawlyn C, Yong KL. Multiple Myeloma. Lancet., 2021, 397(10272):410–27., DOI 10.1016/S0140-6736(21)00135-5 Kazandjian D. Multiple Myeloma Epidemiology and Survival: A Unique Malignancy. Semin Oncol, 2016, 43(6):676–81., DOI 10.1053/j.seminoncol.2016. 11.004 Cowan AJ, Green DJ, KwokM, Lee S, Coffey DG,Holmberg LA, et al. Diagnosis and Management of Multiple Myeloma: A Review. Jama., 2022, 327(5):464–77., DOI 10.1001/jama.2022.0003 Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC. Understanding Multiple Myeloma Pathogenesis in the Bone Marrow to Identify New Therapeutic Targets. Nat Rev Cancer, 2007, 7(8):585–98., DOI 10.1038/nrc2189 Bianchi G, Munshi NC. Pathogenesis beyond the Cancer Clone(s, in Multiple Myeloma. Blood, 2015, 125(20):3049–58., DOI 10.1182/blood-2014-11-568881 He H, Li Z, Lu J, Qiang W, Jiang S, Xu Y, et al. Single-cell RNA-Seq Reveals Clonal Diversity and Prognostic Genes of Relapsed Multiple Myeloma. Clin Transl Med, 2022, 12(3):e757., DOI 10.1002/ctm2.757 Alessandrini F, Pezze L, Ciribilli Y. LAMPs: Shedding Light on Cancer Biology. Semin Oncol, 2017, 44(4):239–53., DOI 10.1053/j.seminoncol.2017.10.013 Defays A, David A, de Gassart A, De Angelis Rigotti F, Wenger T, Camossetto V, et al. BAD-LAMP Is a Novel Biomarker of Nonactivated Human Plasmacytoid Dendritic Cells. Blood, 2011, 118(3):609–17., DOI 10.1182/blood-2010-11-319699 Deng Y, Bi M, Delerue F, Forrest SL, Chan G, van der Hoven J, et al. Loss of LAMP5 Interneurons Drives Neuronal Network Dysfunction in Alzheimer’s Disease. Acta Neuropathol, 2022, 144(4):637–50., DOI 10.1007/s00401-022-02457-w Combes A, Camosseto V, N’Guessan P, Arguello RJ, Mussard J, Caux C, et al. BAD-LAMP Controls TLR9 Trafficking and Signalling in Human Plasmacytoid Dendritic Cells. Nat Commun, 2017, 8(1):913., DOI 10.1038/s41467-017-00695-1 Vrabel D, Pour L, Sevcikova S. The Impact of NF-Κb Signaling on Pathogenesis and Current Treatment Strategies in Multiple Myeloma. Blood Rev, 2019, 34:56–66., DOI 10.1016/j.blre.2018.11.003 Umeda S, Kanda M, Shimizu D, Nakamura S, Sawaki K, Inokawa Y, et al. Lysosomal-associated Membrane Protein Family Member 5 Promotes the Metastatic Potential of Gastric Cancer Cells. Gastric Cancer, 2022, 25(3): 558–72., DOI 10.1007/s10120-022-01284-y Beird HC, Khan M,Wang F, Alfayez M, Cai T, Zhao L, et al. Features of Nonactivation Dendritic State and Immune Deficiency in Blastic Plasmacytoid Dendritic Cell Neoplasm, BPDCN). Blood Cancer J, 2019, 9(12):99., DOI 10. 1038/s41408-019-0262-0 Gracia-Maldonado G, Clark J, Burwinkel M, Greenslade B, Wunderlich M, Salomonis N, et al. LAMP-5 Is an Essential Inflammatory-Signaling Regulator and Novel Immunotherapy Target for Mixed Lineage Leukemia-Rearranged Acute Leukemia. Haematologica, 2022, 107(4):803–15., DOI 10.3324/haematol.2020.257451 Wang WT, Han C, Sun YM, Chen ZH, Fang K, Huang W, et al. Activation of the Lysosome-Associated Membrane Protein LAMP5 by DOT1L Serves as a Bodyguard for MLL Fusion Oncoproteins to Evade Degradation in Leukemia. Clin Cancer Res, 2019, 25(9):2795–808., DOI 10.1158/1078-0432.CCR-18-1474 Chng WJ, Kumar S, Vanwier S, Ahmann G, Price-Troska T, Henderson K, et al. Molecular Dissection of Hyperdiploid Multiple Myeloma by Gene Expression Profiling. Cancer Res, 2007, 67(7):2982–9., DOI 10.1158/0008-5472.CAN-06-4046 Barrett T, Troup DB, Wilhite SE, Ledoux P, Evangelista C, Kim IF, et al. NCBI GEO: Archive for Functional Genomics Data Sets--10 Years on. Nucleic Acids Res, 2011, 39:D1005–1010., DOI 10.1093/nar/gkq1184 Ma T, Chen Y, Yi ZG, Liu J, Li YH, Bai J, et al. NORAD Promotes Multiple Myeloma Cell Progression via BMP6/P-Erk1/2 axis. Cell Signal, 2022, 100:110474., DOI 10.1016/j.cellsig.2022.110474 Barwick BG, Gupta VA, Vertino PM, Boise LH. Cell of Origin and Genetic Alterations in the Pathogenesis of Multiple Myeloma. Front Immunol, 2019, 10: 1121., DOI 10.3389/fimmu.2019.01121 Kyle RA, Therneau TM, Rajkumar SV, Offord JR, Larson DR, Plevak MF, et al. A Long-Term Study of Prognosis in Monoclonal Gammopathy of Undetermined Significance. N Engl J Med, 2002, 346(8):564–9., DOI 10.1056/ NEJMoa01133202 Kyle RA, Rajkumar SV. Management of Monoclonal Gammopathy of Undetermined Significance, MGUS, and Smoldering Multiple Myeloma, SMM). Oncology, Williston Park,, 2011, 25(7):578–86. Zhou L, Yu Q, Wei G, Wang L, Huang Y, Hu K, et al. Measuring the Global, Regional, and National burden of Multiple Myeloma from 1990 to 2019. BMC Cancer, 2021, 21(1):606., DOI 10.1186/s12885-021-08280-y Landgren O, Iskander K. Modern Multiple Myeloma Therapy: Deep, Sustained Treatment Response and Good Clinical Outcomes. J Intern Med, 2017, 281(4):365–82., DOI 10.1111/joim.12590 Kumar SK, Dispenzieri A, Lacy MQ, Gertz MA, Buadi FK, Pandey S, et al. Continued Improvement in Survival in Multiple Myeloma: Changes in Early Mortality and Outcomes in Older Patients. Leukemia, 2014, 28(5):1122–8., DOI 10.1038/leu.2013.313 Jasielec JK, Kubicki T, Raje N, Vij R, Reece D, Berdeja J, et al. Carfilzomib, Lenalidomide, and Dexamethasone Plus Transplant in Newly Diagnosed Multiple Myeloma. Blood., 2020, 136(22):2513–23., DOI 10.1182/blood.2020007522 Ravi G, Costa LJ. Bispecific T-Cell Engagers for Treatment of Multiple Myeloma. Am J Hematol, 2022, 98:S13–S21., DOI 10.1002/ajh.26628 Pawlyn C, Davies FE. Toward Personalized Treatment in Multiple Myeloma Based on Molecular Characteristics. Blood., 2019, 133(7):660–75., DOI 10.1182/ blood-2018-09-825331 Mondala PK, Vora AA, Zhou T, Lazzari E, Ladel L, Luo X, et al. Selective Antisense Oligonucleotide Inhibition of Human IRF4 Prevents MalignantMyeloma Regeneration via Cell Cycle Disruption. Cell Stem Cell, 2021, 28(4):623–36.e9., DOI 10.1016/j.stem.2020.12.017 Chavan SS, He J, Tytarenko R, Deshpande S, Patel P, Bailey M, et al. Bi-allelic Inactivation Is More Prevalent at Relapse in Multiple Myeloma, Identifying RB1 as an Independent Prognostic Marker. Blood Cancer J, 2017, 7(2):e535., DOI 10.1038/ bcj.2017.12 Kefaloyianni E, Gaitanaki C, Beis I. ERK1/2 and P38-MAPK Signalling Pathways, through MSK1, Are Involved in NF-kappaB Transactivation during Oxidative Stress in Skeletal Myoblasts. Cell Signal., 2006, 18(12):2238–51., DOI 10. 1016/j.cellsig.2006.05.004 El Mchichi B, Hadji A, Vazquez A, Leca G. p38 MAPK and MSK1 Mediate Caspase-8 Activation in Manganese-Induced Mitochondria-dependent Cell Death. Cell Death Differ, 2007, 14(10):1826–36., DOI 10.1038/sj.cdd.4402187 Terazawa S, Mori S, Nakajima H, Yasuda M, Imokawa G. The UVBStimulated Expression of Transglutaminase 1 Is Mediated Predominantly via the NFκB Signaling Pathway: New Evidence of its Significant Attenuation through the Specific Interruption of the p38/MSK1/NFκBp65 Ser276 Axis. PLoS One, 2015, 10(8):e0136311., DOI 10.1371/journal.pone.0136311 Shen X, Shen P, Yang Q, Yin Q, Wang F, Cong H, et al. Knockdown of Long Non-coding RNA PCAT-1 Inhibits Myeloma Cell Growth and Drug Resistance via P38 and JNK MAPK Pathways. J Cancer, 2019, 10(26):6502–10., DOI 10.7150/jca. 35098 Xiao W, Xu Z, Chang S, Li B, Yu D, Wu H, et al. Rafoxanide, an Organohalogen Drug, Triggers Apoptosis and Cell Cycle Arrest in Multiple Myeloma by Enhancing DNA Damage Responses and Suppressing the P38 MAPK Pathway. Cancer Lett, 2019, 444:45–59., DOI 10.1016/j.canlet.2018. 12.014 He J, Liu Z, Zheng Y, Qian J, Li H, Lu Y, et al. p38 MAPK in Myeloma Cells Regulates Osteoclast and Osteoblast Activity and Induces Bone Destruction. Cancer Res, 2012, 72(24):6393–402., DOI 10.1158/0008-5472.CAN-12-2664 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1611083 EP 1611094 DI 10.3389/pore.2023.1611083 PG 12 ER PT J AU Boniface, Ch Spellman, P AF Boniface, T. Christopher Spellman, T. Paul TI Corrigendum: Blood, toil, and taxoteres: Biological determinants of treatment-induced ctDNA dynamics for interpreting tumor response SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE biomarkers; liquid biopsy; circulating tumor DNA; ctDNA; tumor growth; treatment response ID biomarkers; liquid biopsy; circulating tumor DNA; ctDNA; tumor growth; treatment response AB In the published article, there were two typographical errors in the article title. Instead of “Blood, toil, and taxoteres: Biological determinates of treatment-induce ctDNA dynamics for interpreting tumor response.” It should be “Blood, toil, and taxoteres: Biological determinants of treatment-induced ctDNA dynamics for interpreting tumor response.” The authors apologize for this error and state that this does not change the scientific conclusions of the article in any way. The original article has been updated. C1 [Boniface, T. Christopher] Oregon Health & Science University, Knight Cancer InstitutePortland, OR, USA. [Spellman, T. Paul] Oregon Health & Science University, Knight Cancer InstitutePortland, OR, USA. RP Spellman, P (reprint author), Oregon Health & Science University, Knight Cancer Institute, Portland, USA. EM spellmap@ohsu.edu NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1611133 EP 1611133 DI 10.3389/pore.2023.1611133 PG 1 ER PT J TI Editorial: Liquid biopsy—A great hope or just hype? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Editorial DE liquid biopsy; ctDNA; circulating tumor cells; exosomes; monitoring ID liquid biopsy; ctDNA; circulating tumor cells; exosomes; monitoring AB Editorial on the Special Issue Liquid biopsy—A great hope or just hype? NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2023 VL 29 IS 1 BP 1611170 EP 1611172 DI 10.3389/pore.2023.1611170 PG 3 ER PT J AU Sun, J Sun, Y Miniderima, Wang, X AF Sun, Jiayang Sun, Yushu Miniderima, Wang, Xiumei TI Cytokine-induced killer cell treatment is superior to chemotherapy alone in esophageal cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE esophageal cancer; immunotherapy; cytokine-induced killer cells; dendritic cells; network meta-analysis ID esophageal cancer; immunotherapy; cytokine-induced killer cells; dendritic cells; network meta-analysis AB Background: The therapeutic efficacy of cytokine-induced killer (CIK) cells versus dendritic cells (DC) co-cultured with CIK cells (DC-CIK) in treating esophageal cancer (EC) remains unclear due to the absence of a direct comparison of these two regimens. This study evaluated the comparative efficacy and safety of CIK cells versus DC-CIK using network meta-analysis in treating EC. Material and methods: We identified eligible studies from previous metaanalyses, then conducted an updated search to retrieve additional trials between February 2020 and July 2021. The primary outcomes included overall survival (OS), objective response rate (ORR), and disease control rate (DCR), and the secondary outcomes included quality of life improved rate (QLIR) and adverse events (AEs). A network meta-analysis of 12 studies was conducted using ADDIS software. Results: Twelve studies were identified, including six comparing CIK or DC-CIK plus chemotherapy (CT) with CT alone. Immunotherapy plus CT significantly improved overall survival (OS) (odds ratio [OR] 4.10, 95% confidence interval [CI] 1.23–13.69), objective response rate (ORR) (OR 2.72, 95% CI 1.79–4.11), disease control rate (DCR) (OR 3.45, 95% CI 2.32–5.14), and quality of life improvement rate (QLIR) (OR 3.54, 95% CI 2.31–5.41). DC-CIK+CT decreased the risk of leukopenia compared with CT alone. However, no statistical difference was detected between CIK-CT and DC-CIK+CT. Conclusion: Based on the available evidence, we concluded that CIK cell treatment is superior to CT alone, but CIK-CT and DC-CIK+CT may be comparable in treating EC. However, comparing CIK-CT and DC-CIK+CT is only based on indirect evidence, so it is undoubtedly necessary to conduct studies to compare CIK-CT with DC-CIK+CT in EC patients directly. C1 [Sun, Jiayang] Affiliated Hospital of Inner Mongolia Medical University, Department of Thoracic SurgeryHohhot, Inner Mongolia, China. [Sun, Yushu] Inner Mongolia Cancer Hospital and Affiliated People’s Hospital of Inner Mongolia Medical University, Department of OncologyHohhot, Inner Mongolia, China. [Miniderima, ] Inner Mongolia Cancer Hospital and Affiliated People’s Hospital of Inner Mongolia Medical University, Department of OncologyHohhot, Inner Mongolia, China. [Wang, Xiumei] Inner Mongolia Cancer Hospital and Affiliated People’s Hospital of Inner Mongolia Medical University, Department of OncologyHohhot, Inner Mongolia, China. RP Wang, X (reprint author), Inner Mongolia Cancer Hospital and Affiliated People’s Hospital of Inner Mongolia Medical University, Department of Oncology, Hohhot, China. EM wangxiumei75@163.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Zhang S, Sun K, Zheng R, Zeng H, Wang S, Chen R, et al. Cancer incidence and mortality in China, 2015. J Natl Cancer Cent, 2021, 1(1):2–11., DOI 10.1016/j.jncc. 2020.12.001 Goode EF, Smyth EC. Immunotherapy for gastroesophageal cancer. J Clin Med, 2016, 5(10):84., DOI 10.3390/jcm5100084 Mu Y, Zhou CH, Chen SF, Ding J, Zhang YX, Yang YP, et al. Effectiveness and safety of chemotherapy combined with cytokine-induced killer cell/dendritic cellcytokine- induced killer cell therapy for treatment of gastric cancer in China: A systematic review and meta-analysis. Cytotherapy, 2016, 18(9):1162–77., DOI 10. 1016/j.jcyt.2016.05.015 Zhou C, Liu D, Li J, Sun H, Zheng X, Wang S, et al. Chemotherapy plus dendritic cells co-cultured with cytokine-induced killer cells versus chemotherapy alone to treat advanced non-small-cell lung cancer: A meta-analysis. Oncotarget, 2016, 7(52):86500–10., DOI 10.18632/oncotarget.13394 Shi L, Zhou Q, Wu J, Ji M, Li G, Jiang J, et al. Efficacy of adjuvant immunotherapy with cytokine-induced killer cells in patients with locally advanced gastric cancer. Cancer Immunol Immunother, 2012, 61(12):2251–9., DOI 10.1007/s00262-012-1289-2 Zhao Z, Liao H, Ju Y. Effect of compound Kushen injection on T-cell subgroups and natural killer cells in patients with locally advanced non-small-cell lung cancer treated with concomitant radiochemotherapy. J Tradit ChinMed, 2016, 36(1):14–8., DOI 10.1016/s0254-6272(16)30002-4 Dudley ME, Gross CA, Somerville RP, Hong Y, Schaub NP, Rosati SF, et al. Randomized selection design trial evaluating CD8+-enriched versus unselected tumor-infiltrating lymphocytes for adoptive cell therapy for patients with melanoma. J Clin Oncol, 2013, 31(17):2152–9., DOI 10.1200/ JCO.2012.46.6441 Chia WK, Teo M, Wang WW, Lee B, Ang SF, Tai WM, et al. Adoptive T-cell transfer and chemotherapy in the first-line treatment of metastatic and/or locally recurrent nasopharyngeal carcinoma. Mol Ther, 2014, 22(1):132–9., DOI 10.1038/ mt.2013.242 Peoples GE, Gurney JM, Hueman MT, Woll MM, Ryan GB, Storrer CE, et al. Clinical trial results of a HER2/neu, E75, vaccine to prevent recurrence in high-risk breast cancer patients. J Clin Oncol, 2005, 23(30):7536–45., DOI 10.1200/JCO.2005. 03.047 Jakel CE, Vogt A, Gonzalez-Carmona MA, Schmidt-Wolf IG. Clinical studies applying cytokine-induced killer cells for the treatment of gastrointestinal tumors. J Immunol Res, 2014, 2014:897214., DOI 10.1155/2014/897214 Mesiano G, Todorovic M, Gammaitoni L, Leuci V, Giraudo Diego L, Carnevale-Schianca F, et al. Cytokine-induced killer, CIK, cells as feasible and effective adoptive immunotherapy for the treatment of solid tumors. Expert Opin Biol Ther, 2012, 12(6):673–84., DOI 10.1517/14712598.2012.675323 Schmidt-Wolf IG, Negrin RS, Kiem HP, Blume KG, Weissman IL. Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity. J Exp Med, 1991, 174(1):139–49., DOI 10.1084/ jem.174.1.139 Mehta BA, Schmidt-Wolf IG, Weissman IL, Negrin RS. Two pathways of exocytosis of cytoplasmic granule contents and target cell killing by cytokineinduced CD3+ CD56+ killer cells. Blood, 1995, 86(9):3493–9., DOI 10.1182/blood. v86.9.3493.bloodjournal8693493 Zhu Y, Zhang H, Li Y, Bai J, Liu L, Liu Y, et al. Efficacy of postoperative adjuvant transfusion of cytokine-induced killer cells combined with chemotherapy in patients with colorectal cancer. Cancer Immunol Immunother, 2013, 62(10): 1629–35., DOI 10.1007/s00262-013-1465-z Wei XC, Yang DD, Han XR, Zhao YA, Li YC, Zhang LJ, et al. Bioactivity of umbilical cord blood dendritic cells and anti-leukemia effect. Int J Clin Exp Med, 2015, 8(10):19725–30. Zhang Y, Ellinger J, Ritter M, Schmidt-Wolf IGH. Clinical studies applying cytokine-induced killer cells for the treatment of renal cell carcinoma. Cancers, Basel,, 2020, 12(9):2471., DOI 10.3390/cancers12092471 Ling JY, Wang QL, Fan JC, Han T, Yang ZQ. Cytokine-induced killers of adoptive immunotherapy combined with chemotherapy for the treatment of esophageal cancer: A meta-analysis [in Chinese]. J Mod Oncol, 2021, 29(9): 1524–30. Liu Y, Mu Y, Zhang A, Ren S, Wang W, Xie J, et al. Cytokine-induced killer cells/dendritic cells and cytokine-induced killer cells immunotherapy for the treatment of esophageal cancer in China: A meta-analysis. Onco Targets Ther, 2017, 10:1897–908., DOI 10.2147/OTT.S132507 Yuan X, Zhang AZ, Ren YL, Wang XL, Jiang CH, Yang L, et al. Cytokineinduced killer cells/dendritic cells and cytokine-induced killer cells immunotherapy for the treatment of esophageal cancer: A meta-analysis. Medicine, 2021, 100(13): e24519., DOI 10.1097/MD.0000000000024519 Higgins JPT, Green S. Cochrane handbook for systematic reviews of interventions. Version 5.1.0. London, United Kingdom: The Cochrane Collaboration, 2011). Available from www.handbookcochrane.org, updated March, 2011). Page MJ, Moher D, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. PRISMA 2020 explanation and elaboration: Updated guidance and exemplars for reporting systematic reviews. BMJ, Clin Res ed,, 2021, 372:n160., DOI 10.1136/bmj. n160 Hutton B, Salanti G, Caldwell DM, Chaimani A, Schmid CH, Cameron C, et al. The PRISMA extension statement for reporting of systematic reviews incorporating network meta-analyses of health care interventions: Checklist and explanations. Ann Intern Med, 2015, 162(11):777–84., DOI 10.7326/M14- 2385 Han RX, Liu X, Pan P, Jia YJ, Yu JC. Effectiveness and safety of chemotherapy combined with dendritic cells co-cultured with cytokine-induced killer cells in the treatment of advanced non-small-cell lung cancer: A systematic review and metaanalysis. PloS one, 2014, 9(9):e108958., DOI 10.1371/journal.pone.0108958 Higgins JP, Altman DG, Gotzsche PC, Juni P, Moher D, Oxman AD, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ, 2011, 343:d5928., DOI 10.1136/bmj.d5928 Bowden J, Tierney JF, Copas AJ, Burdett S. Quantifying, displaying and accounting for heterogeneity in the meta-analysis of RCTs using standard and generalised Q statistics. BMC Med Res Methodol, 2011, 11:41., DOI 10.1186/1471- 2288-11-41 Higgins JP, Thompson SG. Quantifying heterogeneity in a meta-analysis. Stat Med, 2002, 21(11):1539–58., DOI 10.1002/sim.1186 Cipriani A, Higgins JPT, Geddes JR, Salanti G. Conceptual and technical challenges in network meta-analysis. Ann Intern Med, 2013, 159(2):130–7., DOI 10. 7326/0003-4819-159-2-201307160-00008 Dias S, Welton NJ, Caldwell DM, Ades AE. Checking consistency in mixed treatment comparison meta-analysis. Stat Med, 2010, 29(7-8):932–44., DOI 10.1002/ sim.3767 Albert I, Makowski D. Ranking crop species using mixed treatment comparisons. Res Synth Methods, 2019, 10(3):343–59., DOI 10.1002/jrsm.1328 Brooks S, Gelman A. Generalmethods for monitoring convergence of iterative simulations. J Comput Graphi Stat, 1998, 7:434–55., DOI 10.1080/10618600.1998. 10474787 Burger DA, Schall R. A bayesian nonlinear mixed-effects regression model for the characterization of early bactericidal activity of tuberculosis drugs. J Biopharm Stat, 2015, 25(6):1247–71., DOI 10.1080/10543406.2014.971170 Salanti G, Ades AE, Ioannidis JP. Graphical methods and numerical summaries for presenting results from multiple-treatment meta-analysis: An overview and tutorial. J Clin Epidemiol, 2011, 64(2):163–71., DOI 10.1016/j. jclinepi.2010.03.016 Palma Perez S, Delgado Rodriguez M. Practical considerations on detection of publication bias. Gac Sanit, 2006, 20(3):10–6., DOI 10.1157/13101085 Chang ZG, Xu QX, Ma L, He LM. Clinical observation on the treatment of advanced esophageal cancer with cytokine induced killer cell infusion combined with chemotherapy [in Chinese]. Shandong Med J, 2013, 53(23):53–4. Gu MW, Sun ZY, Wang MF. Application of DIK combined with chemotherapy in the treatment of advanced esophagus cancer [in Chinese]. J Hunan Univ Chin Med, 2013, 33(12):8. Liu GJ, Mei JZ, Xiao P, Li RJ, Li M. Efficacy of chemotherapy combined with cytokine-induced killer cells in treatment of advanced esophageal carcinoma [in Chinese]. J Zhengzhou Univ, Med Sci,, 2011, 46(2):169–73. Qu WF, Zhang WM, Ding XY. IL-12 induced CIK cells combined with chemotherapy improve immune function and efficacy in patients with esophageal carcinoma- 12 induced CIK cells combined with chemotherapy on esophageal cancer and immune function [in Chinese]. World Chin J Digestol, 2015, 23(28): 4553–7., DOI 10.11569/wcjd.v23.i28.4553 Xi XX, Lyu BH, Ye T, Chen R, Xu X. Effect of DC-CIK cells’ biotherapy in comprehensive therapy of esophagus cancer: Arandomized controlled trial [in Chinese]. Chin J Clin Thorac Cardiovasc Surg, 2015, 22(8):765–9. Xu Y, Huang Y. Effect of endoscopic chemo-seed implantation combined with DC-ClK therapy on tumor malignancy in patients with obstructive esophageal cancer [in Chinese]. J Hainan Med Univ, 2018, 24(9):967–70. Yang B, Chen YM, Wang LH, Tian ZY. Treatment of esophageal cancer in the elderly by gastroscopic implantation of chemotherapy particles combined with DC-CIK immune cell infusion [in Chinese]. Chin J Clin Res, 2016, 29(3): 331–3. Yang B, Li W, Su XA, Chen YM. The clinical efficacy of endoscopic implantation with chemotherapeutic slow-release combined with DC-CIK immune cells infusion for patients with obstructive esophageal cancers [in Chinese]. China J Endosc, 2015, 21(5):483–6. Zhang NB, He MW. Clinical research of DCIK cell combined chemotherapy in treatment of patients with advanced esophageal squamous carcinoma [in Chinese]. Chin J Prim Med Pharm, 2016, 23(6):859–62. Zhang ZS, Yao TY, Zhang M, Wang GH. Efficacy of dendritic cells and cytokine-induced killercells for esophageal cancer [in Chinese]. Chin J Clin Oncol Rehabil, 2017, 24(7):828–30. Zhao Y, Ning S, Yang G, Zhang SG. Evaluation on clinical efficacy of DC-CIK combined with hemotherapy in treating moderate and advanced esophageal carcinoma [in Chinese]. Lab Med Clin, 2015, 12(19):2823–6. Zhu HP, Zhang QA. Analysis of the effect of injection in treatment of advanced esophageal cancer paclitaxel cisplatin regimen combined with autologous CIK cells [inn Chinese]. Chin J Clin Oncol Rehabil, 2014, 21(7): 837–9. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1610710 EP 1610719 DI 10.3389/pore.2023.1610710 PG 10 ER PT J AU Liu, Yh Meng, R Zhu, B Zhan, Qq Yang, X Ding, Gy Jia, Chl Liu, Qy Xu, Wg AF Liu, Yu-hang Meng, Rui Zhu, Bing Zhan, Qi-qi Yang, Xin Ding, Guan-yi Jia, Chun-liang Liu, Qian-yu Xu, Wei-guo TI Integrated oxidative stress score for predicting prognosis in stage III gastric cancer undergoing surgery SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gastric cancer; oxidative stress; albumin; bilirubin; blood urea nitrogen ID gastric cancer; oxidative stress; albumin; bilirubin; blood urea nitrogen AB Objective: This study aimed to develop a novel scoring system, named the integrated oxidative stress score (IOSS), based on oxidative stress indices to predict the prognosis in stage III gastric cancer. Methods: Retrospective analysis of stage III gastric cancer patients who were operated on between January 2014 and December 2016 were enrolled into this research. IOSS is a comprehensive index based on an achievable oxidative stress index, comprising albumin, blood urea nitrogen, and direct bilirubin. The patients were divided according to receiver operating characteristic curve into two groups of low IOSS (IOSS ≤ 2.00) and high IOSS (IOSS > 2.00). The grouping variable was performed by Chi-square test or Fisher’s precision probability test. The continuous variables were evaluated by t-test. The disease free survival (DFS) and overall survival (OS) were performed by Kaplan-Meier and Log-Rank tests. Univariate Cox proportional hazards regression models and stepwise multivariate Cox proportional hazards regression analysis were determined to appraise the potential prognostic factors for DFS and OS. A nomogram of the potential prognostic factors by the multivariate analysis for DFS and OS was established with R software. In order to assess the accuracy of the nomogram in forecasting prognosis, the calibration curve and decision curve analysis were produced, contrasting the observed outcomes with the predicted outcomes. Results: The IOSS was significantly correlated with the DFS and OS, and was a potential prognostic factor in patients with stage III gastric cancer. Patients with low IOSS had longer survival (DFS: χ2 = 6.632, p = 0.010; OS: χ2 = 6.519, p = 0.011), and higher survival rates. According to the univariate and multivariate analyses, the IOSS was a potential prognostic factor. The nomograms were conducted on the potential prognostic factors to improve the correctness of survival prediction and evaluate the prognosis in stage III gastric cancer patients. The calibration curve indicated a good agreement in 1-, 3-, 5-year lifetime rates. The decision curve analysis indicated that the nomogram’s predictive clinical utility for clinical decision was better than IOSS. Conclusion: IOSS is a nonspecific tumor predictor based on available oxidative stress index, and low IOSS is found to be a vigorous factor of better prognosis in stage III gastric cancer. C1 [Liu, Yu-hang] North China University of Science and Technology, School of Clinical MedicineTangshan, China. [Meng, Rui] Tongji University, Yangpu Hospital, Department of Emergency Intensive Care UnitShanghai, China. [Zhu, Bing] Tangshan Gongren HospitalTangshan, China. [Zhan, Qi-qi] North China University of Science and Technology, Affiliated Tangshan Gongren HospitalTangshan, China. [Yang, Xin] North China University of Science and Technology, Affiliated Tangshan Gongren HospitalTangshan, China. [Ding, Guan-yi] Tangshan Gongren HospitalTangshan, China. [Jia, Chun-liang] Tangshan People’s HospitalTangshan, China. [Liu, Qian-yu] North China University of Science and Technology, Affiliated Tangshan Gongren HospitalTangshan, China. [Xu, Wei-guo] North China University of Science and Technology, Affiliated Tangshan Gongren HospitalTangshan, China. RP Xu, Wg (reprint author), North China University of Science and Technology, Affiliated Tangshan Gongren Hospital, Tangshan, China. EM xwg853@163.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–249., DOI 10.3322/caac.21660 Zheng X, Wang Y, Chen Y. Gastric cancer subtypes in tumour and nontumour tissues by immunologic and hallmark gene sets. J Oncol, 2022, 2022:7887711., DOI 10.1155/2022/7887711 Cao W, Chen HD, Yu YW, Li N, Chen WQ. Changing profiles of cancer burden worldwide and in China: A secondary analysis of the global cancer statistics 2020. Chin Med J, Engl,, 2021, 134(7):783–791., DOI 10.1097/CM9. 0000000000001474 Arnold M, Ferlay J, van Berge Henegouwen MI, Soerjomataram I. Global burden of oesophageal and gastric cancer by histology and subsite in 2018. Gut, 2020, 69(9):1564–1571., DOI 10.1136/gutjnl-2020-321600 Japanese Gastric Cancer Association. Japanese gastric cancer treatment guidelines 2018, 5th edition). Gastric Cancer, 2021, 24(1):1–21., DOI 10.1007/ s10120-020-01042-y Sekiguchi M, Oda I, Matsuda T, Saito Y. Epidemiological trends and future perspectives of gastric cancer in eastern Asia. Digestion, 2022, 103(1):22–28., DOI 10. 1159/000518483 Young E, Philpott H, Singh R. Endoscopic diagnosis and treatment of gastric dysplasia and early cancer: Current evidence and what the future may hold. World J Gastroenterol, 2021, 27(31):5126–5151., DOI 10.3748/wjg.v27.i31. 5126 Komatsu Y, Hironaka S, Tanizawa Y, Cai Z, Piao Y, Boku N. Treatment pattern for advanced gastric cancer in Japan and factors associated with sequential treatment: A retrospective administrative claims database study. Adv Ther, 2022, 39(1):296–313., DOI 10.1007/s12325-021-01931-3 Kung CH, Jestin Hannan C, Linder G, Johansson J, NilssonM, Hedberg J, et al. Impact of surgical resection rate on survival in gastric cancer: Nationwide study. BJS Open, 2021, 5(2):zraa017., DOI 10.1093/bjsopen/zraa017 Lyons K, Le LC, Pham YT, Borron C, Park JY, Tran CTD, et al. Gastric cancer: Epidemiology, biology, and prevention: A mini review. Eur J Cancer Prev, 2019, 28(5):397–412., DOI 10.1097/CEJ.0000000000000480 Sexton RE, Hallak MNA, Uddin MH, Diab M, Azmi AS. Gastric cancer heterogeneity and clinical outcomes. Technol Cancer Res Treat, 2020, 19: 1533033820935477., DOI 10.1177/1533033820935477 Hirahara N, Tajima Y, Matsubara T, Fujii Y, Kaji S, Kawabata Y, et al. Systemic immune-inflammation index predicts overall survival in patients with gastric cancer: A propensity score-matched analysis. J Gastrointest Surg, 2021, 25(5):1124–1133., DOI 10.1007/s11605-020-04710-7 Li Q, Zhang X, Tang M, Song M, Zhang Q, Zhang K, et al. Different muscle mass indices of the Global Leadership Initiative on Malnutrition in diagnosing malnutrition and predicting survival of patients with gastric cancer. Nutrition, 2021, 89:111286., DOI 10.1016/j.nut.2021.111286 Sano T, Coit DG, Kim HH, Roviello F, Kassab P, Wittekind C, et al. Proposal of a new stage grouping of gastric cancer for TNM classification: International Gastric Cancer Association staging project. Gastric Cancer, 2017, 20(2):217–225., DOI 10.1007/s10120-016-0601-9 Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet, 2020, 396(10251):635–648., DOI 10.1016/S0140-6736(20)31288-5 Jelic MD, Mandic AD, Maricic SM, Srdjenovic BU. Oxidative stress and its role in cancer. J Cancer Res Ther, 2021, 17(1):22–28., DOI 10.4103/jcrt.JCRT_862_16 Yun HR, Jo YH, Kim J, Shin Y, Kim SS, Choi TG. Roles of autophagy in oxidative stress. Int J Mol Sci, 2020, 21(9):3289., DOI 10.3390/ijms21093289 Szczuko M, Kikut J, Szczuko U, Szydlowska I, Nawrocka-Rutkowska J, Zietek M, et al. Nutrition strategy and life style in polycystic ovary syndrome-narrative review. Nutrients, 2021, 13(7):2452., DOI 10.3390/nu13072452 Henriksen HB, Raeder H, Bohn SK, Paur I, Kvaerner AS, Billington SA, et al. The Norwegian dietary guidelines and colorectal cancer survival, CRC-NORDIET, study: A food-based multicentre randomized controlled trial. BMC Cancer, 2017, 17(1):83., DOI 10.1186/s12885-017-3072-4 Wu Z, Wang L, Wen Z, Yao J. Integrated analysis identifies oxidative stress genes associated with progression and prognosis in gastric cancer. Sci Rep, 2021, 11(1):3292., DOI 10.1038/s41598-021-82976-w Geng R, Chen Z, Zhao X, Qiu L, Liu X, Liu R, et al. Oxidative stress-related genetic polymorphisms are associated with the prognosis of metastatic gastric cancer patients treated with epirubicin, oxaliplatin and 5-fluorouracil combination chemotherapy. PLoS One, 2014, 9(12):e116027., DOI 10.1371/journal.pone.0116027 Cao Y, Deng S, Yan L, Gu J, Mao F, Xue Y, et al. An oxidative stress indexbased score for prognostic prediction in colorectal cancer patients undergoing surgery. Oxid Med Cell Longev, 2021, 2021:6693707., DOI 10.1155/2021/6693707 Joharatnam-Hogan N, Shiu KK, Khan K. Challenges in the treatment of gastric cancer in the older patient. Cancer Treat Rev, 2020, 85:101980., DOI 10.1016/ j.ctrv.2020.101980 Stocker G, Thieme R, Lordick F. Neoadjuvant and perioperative treatment of gastric cancer, current studies and new biomarkers. Chirurg, 2021, 92(6):499–505., DOI 10.1007/s00104-021-01355-3 Chen D, Liu Z, Liu W, Fu M, Jiang W, Xu S, et al. Predicting postoperative peritoneal metastasis in gastric cancer with serosal invasion using a collagen nomogram. Nat Commun, 2021, 12(1):179., DOI 10.1038/ s41467-020-20429-0 Sczepanik FSC, Grossi ML, Casati M, Goldberg M, Glogauer M, Fine N, et al. Periodontitis is an inflammatory disease of oxidative stress: We should treat it that way. Periodontol, 2020, 84(1):45–68., DOI 10.1111/prd.12342 O’Malley J, Kumar R, Inigo J, Yadava N, Chandra D. Mitochondrial stress response and cancer. Trends Cancer, 2020, 6(8):688–701., DOI 10.1016/j.trecan. 2020.04.009 Li T, Zhang T, Gao H, Liu R, Gu M, Yang Y, et al. Tempol ameliorates polycystic ovary syndrome through attenuating intestinal oxidative stress and modulating of gut microbiota composition-serum metabolites interaction. Redox Biol, 2021, 41:101886., DOI 10.1016/j.redox.2021.101886 Tumer AA, Rashid FA. The relationship between circulating irisin and oxidative stress in gastric and colorectal cancer patients. Asian Pac J Cancer Prev, 2022, 23(8):2649–2654., DOI 10.31557/APJCP.2022.23.8. 2649 Jakubik JA, Kolodziejczyk-Czepas J, Kedzierska M, Kaczmarek M, Nowak P, Potemski P, et al. A preliminary evaluation of oxidative stress in patients with gastric cancer before chemotherapy. Arch Med Sci, 2021, 18(2):440–447., DOI 10. 5114/aoms/102344 Danis N, Ozkan AE, Karatas F, Cakici C, Yigitbasi T, Sevencan NO, et al. The importance of antioxidant status in gastric intestinal metaplasia. J Med Biochem, 2021, 40(4):378–383., DOI 10.5937/jomb0-29349 Terhaar HM, Allbaugh RA, Mochel JP, Sebbag L. Serum albumin and total protein concentration in the tear film of horses with healthy or diseased eyes. Vet Ophthalmol, 2021, 24(1):20–27., DOI 10.1111/vop.12822 Lei J, Wang Y, Guo X, Yan S, Ma D,Wang P, et al. Low preoperative serum ALB level is independently associated with poor overall survival in endometrial cancer patients. Future Oncol, 2020, 16(8):307–316., DOI 10.2217/fon-2019- 0732 Cattaneo L, Lopreiato V, Piccioli-Cappelli F, Trevisi E, Minuti A. Plasma albumin-to-globulin ratio before dry-off as a possible index of inflammatory status and performance in the subsequent lactation in dairy cows. J Dairy Sci, 2021, 104(7):8228–8242., DOI 10.3168/jds.2020-19944 Li ZZ, Huang Q, Yang XL, Zeng J, Wang QH, Tang HM, et al. Cholesterol metabolic markers for differential evaluation of patients with hyperlipidemia and familial hypercholesterolemia. Dis Markers, 2022, 2022:2008556., DOI 10.1155/ 2022/2008556 Jia Z, Zhu Z, Wang Y, Ding J, Lin Z, Zhang Y, et al. The prognostic value of serum bilirubin in colorectal cancer patients with surgical resection. Int J Biol Markers, 2021, 36(2):17246008211036128., DOI 10.1177/ 17246008211036128 Turkmen D. Serum bilirubin and uric acid antioxidant levels in rosacea patients. J Cosmet Dermatol, 2020, 19(10):2717–2720., DOI 10.1111/jocd.13395 Zhang L, Zhang C, Meng Z, Gong L, Pang C, Liu X, et al. Serum bilirubin is negatively associated with white blood cell count. Clinics, Sao Paulo,, 2019, 74: e775., DOI 10.6061/clinics/2019/e775 Jiang D, Shi J, Yuan M, Duan X, Li L, Li Q. Levels of serum bilirubin in small cell lung cancer and non-small cell lung cancer patients. Cell Mol Biol, Noisy-legrand,, 2018, 64(6):71–76. Monroy-IglesiasMJ,Moss C, Beckmann K, Hammar N, Walldius G, Bosco C, et al. Serum total bilirubin and risk of cancer: A Swedish cohort study and metaanalysis. Cancers, Basel,, 2021, 13(21):5540., DOI 10.3390/cancers13215540 Yang L, Ge LY, Yu T, Liang Y, Yin Y, Chen H. The prognostic impact of serum bilirubin in stage IV colorectal cancer patients. J Clin Lab Anal, 2018, 32(2):e22272., DOI 10.1002/jcla.22272 Shen S, Yan X, Xu B. The blood urea nitrogen/creatinine, BUN/cre, ratio was U-shaped associated with all-cause mortality in general population. Ren Fail, 2022, 44(1):184–190., DOI 10.1080/0886022X.2022.2030359 Peng X, Huang Y, Fu H, Zhang Z, He A, Luo R. Prognostic value of blood urea nitrogen to serum albumin ratio in intensive care unit patients with lung cancer. Int J Gen Med, 2021, 14:7349–7359., DOI 10.2147/IJGM.S337822 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1610897 EP 1610909 DI 10.3389/pore.2023.1610897 PG 13 ER PT J AU Zuo, Y Leng, G Leng, P AF Zuo, Yanhua Leng, Guangyi Leng, Ping TI Identification and validation of molecular subtype and prognostic signature for lung adenocarcinoma based on neutrophil extracellular traps SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunotherapy; lung adenocarcinoma; tumor microenvironment; prognostic signature; neutrophil extracellular traps ID immunotherapy; lung adenocarcinoma; tumor microenvironment; prognostic signature; neutrophil extracellular traps AB Background: Neutrophil Extracellular Traps (NETs) are fibrous networks made of DNA-histone complexes and proteins protruded from activated neutrophils. Accumulating evidences have highlighted the vital role of NETs in tumor progression and diffusion. However, limited systematic studies regarding the role of NETs in LUAD have been performed. Methods: Differentially expressed NETs-related genes and their mutation landscape were identified with TCGA database. Consensus clustering analysis was performed to determine the NETs-related subtypes of LUAD. LASSO algorithm was employed to construct a prognostic signature. Moreover, GSE30219 and GSE31210 were used as independent validation. We also constructed a lncRNA-miRNA-mRNA regulatory axis with several miRNA and lncRNA databases. Results: Consensus clustering identified two NETs-related clusters in LUAD. High NETs score was correlated with a favorable overall survival, abundant immune cell infiltration, and high activity of immune response signal pathways. Six NET-related genes (G0S2, KCNJ15, S100A12, AKT2, CTSG, and HMGB1) with significant prognostic value were screened to develop a prognostic signature. LUAD patients with low-risk had a significantly favorable overall survival both in the training set and validation set. Moreover, NETs-related risk score and clinical stage could act as an independent prognostic factor for LUAD patients. Significant correlation was obtained between risk score and tumor immune microenvironment. We also identified lncRNA BCYRN1/miR-3664-5p/CTSG regulatory axis that may be involved in the progression of LUAD. Conclusion: We developed two molecular subtypes and a prognostic signature for LUAD based on NETs-related genes. This stratification could provide more evidences for estimating the prognosis and immunotherapy of LAUD patients. C1 [Zuo, Yanhua] The Affiliated Hospital of Qingdao University, Department of PharmacyQingdao, China. [Leng, Guangyi] Shenyang Pharmaceutical University, Laboratory of Drug Metabolism and PharmacokineticsShenyang, China. [Leng, Ping] The Affiliated Hospital of Qingdao University, Department of PharmacyQingdao, China. RP Leng, P (reprint author), The Affiliated Hospital of Qingdao University, Department of Pharmacy, Qingdao, China. EM 18661808926@163.com CR Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin, 2020, 70(1):7–30., DOI 10.3322/caac.21590 Brody H. Lung cancer. Nature, 2020, 587(7834):S7., DOI 10.1038/d41586-020- 152-0 3. Slim A, Kamoun H, Hadidene Y, Smadhi H, Meddeb A, Megdiche ML. Postoperative recurrence of primary lung cancer: Anatomo-clinical and therapeutic study. Tunis Med, 2021, 99(5):560–8. Denisenko TV, Budkevich IN, Zhivotovsky B. Cell death-based treatment of lung adenocarcinoma. Cell Death Dis, 2018, 9(2):117., DOI 10.1038/s41419-017- 0063-y Cui Y, Fang W, Li C, Tang K, Zhang J, Lei Y, et al. Development and validation of a novel signature to predict overall survival in "driver gene-negative" lung adenocarcinoma, LUAD): Results of a multicenter study. Clin Cancer Res, 2019, 25(5):1546–56., DOI 10.1158/1078-0432.CCR-18-2545 Lee KC, Koh J, Chung DH, Jeon YK. A case of concomitant EGFR/ALK alteration against amutated EGFR background in early-stage lung adenocarcinoma. J Pathol Transl Med, 2021, 55(2):139–44., DOI 10.4132/jptm.2020.12.16 Ocana A, Nieto-Jimenez C, Pandiella A, Templeton AJ. Neutrophils in cancer: Prognostic role and therapeutic strategies. Mol Cancer, 2017, 16(1):137., DOI 10. 1186/s12943-017-0707-7 Rada B. Neutrophil extracellular traps. Methods Mol Biol, 2019, 1982:517–28., DOI 10.1007/978-1-4939-9424-3_31 Zhang Y, Guo L, Dai Q, Shang B, Xiao T, Di X, et al. A signature for pan-cancer prognosis based on neutrophil extracellular traps. J Immunother Cancer, 2022, 10(6):e004210., DOI 10.1136/jitc-2021-004210 Fang C, Liu F, Wang Y, Yuan S, Chen R, Qiu X, et al. A innovative prognostic symbol based on neutrophil extracellular traps, NETs)-related lncRNA signature in non-small-cell lung cancer. Aging, Albany NY,, 2021, 13(13):17864–79., DOI 10. 18632/aging.203289 Klopf J, Brostjan C, Eilenberg W, Neumayer C. Neutrophil extracellular traps and their implications in cardiovascular and inflammatory disease. Int J Mol Sci, 2021, 22(2):559., DOI 10.3390/ijms22020559 Masucci MT, Minopoli M, Del Vecchio S, Carriero MV. The emerging role of neutrophil extracellular traps, NETs, in tumor progression and metastasis. Front Immunol, 2020, 11:1749., DOI 10.3389/fimmu.2020.01749 Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol, 2018, 18(2):134–47., DOI 10.1038/nri.2017.105 Senbabaoglu Y, Gejman RS, Winer AG, Liu M, Van Allen EM, de Velasco G, et al. Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol, 2016, 17(1):231., DOI 10.1186/s13059-016-1092-z Khan U, Chowdhury S, Billah MM, Islam KMD, Thorlacius H, Rahman M. Neutrophil extracellular traps in colorectal cancer progression and metastasis. Int J Mol Sci, 2021, 22(14):7260., DOI 10.3390/ijms22147260 Demkow U. Neutrophil extracellular traps, NETs, in cancer invasion, evasion and metastasis. Cancers, Basel,, 2021, 13(17):4495., DOI 10.3390/cancers13174495 Yang D, Liu J. Neutrophil extracellular traps: A new player in cancer metastasis and therapeutic target. J Exp Clin Cancer Res, 2021, 40(1):233., DOI 10.1186/s13046-021-02013-6 Sun S, Guo W, Wang Z, Wang X, Zhang G, Zhang H, et al. Development and validation of an immune-related prognostic signature in lung adenocarcinoma. Cancer Med, 2020, 9(16):5960–75., DOI 10.1002/cam4.3240 Liu YT, Sun ZJ. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics, 2021, 11(11):5365–86., DOI 10.7150/thno.58390 Li B, Cui Y, Nambiar DK, Sunwoo JB, Li R. The immune subtypes and landscape of squamous cell carcinoma. Clin Cancer Res, 2019, 25(12):3528–37., DOI 10.1158/1078-0432.CCR-18-4085 Wei K, Ma Z, Yang F, Zhao X, JiangW, Pan C, et al. M2 macrophage-derived exosomes promote lung adenocarcinoma progression by delivering miR-942. Cancer Lett, 2022, 526:205–16., DOI 10.1016/j.canlet.2021.10.045 Lan J, Sun L, Xu F, Liu L, Hu F, Song D, et al. M2 macrophage-derived exosomes promote cell migration and invasion in colon cancer. Cancer Res, 2019, 79(1):146–58., DOI 10.1158/0008-5472.CAN-18-0014 Chen J, Zhang K, Zhi Y, Wu Y, Chen B, Bai J, et al. Tumor-derived exosomal miR-19b-3p facilitates M2 macrophage polarization and exosomal LINC00273 secretion to promote lung adenocarcinoma metastasis via Hippo pathway. Clin Transl Med, 2021, 11(9):e478., DOI 10.1002/ctm2.478 Morandi F, Airoldi I. HLA-G and other immune checkpoint molecules as targets for novel combined immunotherapies. Int J Mol Sci, 2022, 23(6):2925., DOI 10.3390/ijms23062925 Liu T, Zhu J, Du W, Ning W, Zhang Y, Zeng Y, et al. AKT2 drives cancer progression and is negatively modulated by miR-124 in human lung adenocarcinoma. Respir Res, 2020, 21(1):227., DOI 10.1186/s12931-020-01491-0 Attoub S, Arafat K, Kamel Hammadi N, Mester J, Gaben AM. Akt2 knockdown reveals its contribution to human lung cancer cell proliferation, growth, motility, invasion and endothelial cell tube formation. Sci Rep, 2015, 5:12759., DOI 10.1038/srep12759 Wu L, Yang L. The function andmechanism of HMGB1 in lung cancer and its potential therapeutic implications. Oncol Lett, 2018, 15(5):6799–805., DOI 10.3892/ ol.2018.8215 Ghafouri-Fard S, Dashti S, Hussen BM, Farsi M, Taheri M. BCYRN1: An oncogenic lncRNA in diverse cancers. Pathol Res Pract, 2021, 220:153385., DOI 10. 1016/j.prp.2021.153385 Lang N, Wang C, Zhao J, Shi F, Wu T, Cao H. Long non-coding RNA BCYRN1 promotes glycolysis and tumor progression by regulating the miR-149/ PKM2 axis in non-small-cell lung cancer. Mol Med Rep, 2020, 21(3):1509–16., DOI 10.3892/mmr.2020.10944 Hu T, Lu YR. BCYRN1, a c-MYC-activated long non-coding RNA, regulates cell metastasis of non-small-cell lung cancer. Cancer Cel Int, 2015, 15:36., DOI 10. 1186/s12935-015-0183-3 Jiao Y, Yang H, Qian J, Gong Y, Liu H, Wu S, et al. miR-3664-5P suppresses the proliferation and metastasis of gastric cancer by attenuating the NF-κB signaling pathway through targeting MTDH. Int J Oncol, 2019, 54(3):845–58., DOI 10.3892/ ijo.2019.4680 Huang GZ, Wu QQ, Zheng ZN, Shao TR, Li F, Lu XY, et al. Bioinformatics analyses indicate that cathepsin G, CTSG, is a potential immune-related biomarker in oral squamous cell carcinoma, OSCC). Onco Targets Ther, 2021, 14:1275–89., DOI 10.2147/OTT.S293148 Li N, Li Y, Zheng P, Zhan X. Cancer stemness-based prognostic immunerelated gene signatures in lung adenocarcinoma and lung squamous cell carcinoma. Front Endocrinol, Lausanne,, 2021, 12:755805., DOI 10.3389/fendo.2021.755805 Song Y, Jin D, Chen J, Luo Z, Chen G, Yang Y, et al. Identification of an immune-related long non-coding RNA signature and nomogram as prognostic target for muscle-invasive bladder cancer. Aging, Albany NY,, 2020, 12(12): 12051–73., DOI 10.18632/aging.103369 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1610899 EP 1610912 DI 10.3389/pore.2023.1610899 PG 14 ER PT J AU Janka, M Zalatnai, A AF Janka, Maria Zalatnai, Attila TI Correlations between the histopathological alterations in minor salivary glands and the clinically suspected Sjogren’s syndrome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; clinicopathology; retrospective study; Sjogren syndrome; labial gland biopsy ID immunohistochemistry; clinicopathology; retrospective study; Sjogren syndrome; labial gland biopsy AB In sicca syndrome patients the xerostomia, xerophthalmia and the serological findings may strongly suggest the autoimmune Sjogren’s syndrome, but the histological findings in the labial salivary gland biopsies do not always justify the suspected diagnosis. The aim of this study was to compare the histomorphological changes and the clinical findings in patients with pathologically established Sjogren’s syndrome and in cases with negative histology. A total of 133 labial biopsies have been retrospectively evaluated from 2015 to May 2022, and the characteristic Sjogren’s lesions were found in 67 cases. According to the clinical data, 34 cases proved to be primary, and 33 were associated (“secondary”) forms. In 66 cases, the histology did not justify Sjogren’s syndrome; a significant acinar loss, fibrolipomatous infiltration, and mild sialadenitis had led to the clinical symptoms. In Sjogren’s histologies, the acinar loss was detected in just 31.8% of cases, which might indicate that the diminished saliva production represents immune-mediated hypofunction rather than direct damage of the acini. This is the first systemic study in Hungary investigating the correlation between pathological alterations and clinical findings. C1 [Janka, Maria] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Zalatnai, Attila] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Zalatnai, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM zalatnai.attila@med.semmelweis-univ.hu CR Pasoto SG, deOliveira Martins VA, Bonfa E. Sjogren’s syndrome and systemic lupus erythematosus: Links and risks. Open Access Rheumatol, 2019, 11:33–45., DOI 10.2147/OARRR.S167783 Maciel G, Crowson CS, Matteson L, Cornec D. Prevalence of primary sjogren’s syndrome in a US population-based cohort. Arthritis Care Res, Hoboken,, 2017, 69: 1612–6., DOI 10.1002/acr.23173 Cornec D, Chiche L. Is primary sjogren’s syndrome an orphan disease? A critical appraisal of prevalence studies in Europe. Ann Rheum Dis, 2015, 74:e25., DOI 10.1136/annrheumdis-2014-206860 Dezsi AJ, Erdei C, Demeter T, Kovacs A, Nagy G, Mensch K, et al. Prevalence of Sjogren’s syndrome in patients with dry mouth in the region of Central Hungary. Oral Dis, 2022)., DOI 10.1111/odi.14264 Yousefi H, Abdollahi M. An update on drug-induced oral reactions. J Pharm Pharm Sci, 2018, 21:171–83., DOI 10.18433/jpps29893 Parisis D, Chivasso C, Perret J, Soyfoo MS, Delporte C. Current state of knowledge on primary Sjogren’s syndrome, an autoimmune exocrinopathy. J Clin Med, 2020, 9:2299., DOI 10.3390/jcm9072299 Negrini S, Emmi G, GrecoM, BorroM, Sardanelli F, Murdaca G, et al. Sjogren’s syndrome: A systemic autoimmune disease. Clin Exp Med, 2022, 22:9–25., DOI 10. 1007/s10238-021-00728-6 Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, et al. 2016 American college of rheumatology/European league against rheumatism classification criteria for primary sjogren’s syndrome: A consensus and data-driven methodology involving three international patient cohorts. Arthritis Rheumatol, 2017, 69:35–45., DOI 10.1002/art.39859 Alunno A, Bartoloni E, Valentini V, La Paglia GMC, Valentini E, Leone MC, et al. Discrepancy between subjective symptoms, objective measures and disease activity indexes: The lesson of primary sjogren’s syndrome. Clin Exp Rheumatol, 2018, 36:210–4. Kim H, Cho SK, Kim HW, Han J, Kim Y, Hwang KG, et al. The prevalence of sjogren’s syndrome in rheumatoid arthritis patients and their clinical features. Korean Med Sci, 2020, 35:e369., DOI 10.3346/jkms.2020.35.e369 Metskula K, Salur L,Mandel M, Uibo R. Demonstration of high prevalence of SS-A antibodies in a general population: Association with HLA-DR and enterovirus antibodies. Immunol Lett, 2006, 106:14–8., DOI 10.1016/j.imlet.2006.03.005 Shen D, Ono K, Do Q, Ohyama H, Nakamura K, Obata K, et al. Clinical anatomy of the inferior labial gland: A narrative review. Gland Surg, 2021, 10: 2284–92., DOI 10.21037/gs-21-143 Soyfoo MS, Catteau X, Delporte C. Parotid gland biopsy as an additional diagnostic tool for supporting the diagnosis of sjogren’s syndrome. Int J Rheumatol, 2011, 2011:302527., DOI 10.1155/2011/302527 Spijkervet FK, Haacke E, Kroese FG, Bootsma H, Vissink A. Parotid gland biopsy, the alternative way to diagnose sjogren syndrome. Rheum Dis Clin North Am, 2016, 42:485–99., DOI 10.1016/j.rdc.2016.03.007 Chisholm DM, Mason DK. Labial salivary gland biopsy in Sjogren’s disease. J Clin Pathol, 1968, 21:656–60., DOI 10.1136/jcp.21.5.656 Witas R, Gupta S, Nguyen CQ. Contributions of major cell populations to sjogren’s syndrome. J Clin Med, 2020, 9:3057., DOI 10.3390/jcm9093057 Zhou H, Yang J, Tian J, Wang S. CD8(+, T lymphocytes: Crucial players in sjogren’s syndrome. Front Immunol, 2021, 11:602823., DOI 10.3389/fimmu.2020.602823 Hamza N, Bos NA, Kallenberg CG. B-cell populations and sub-populations in Sjogren’s syndrome. Presse Med, 2012, 41:e475–83., DOI 10.1016/j.lpm.2012.05.021 Verstappen GM, Pringle S, Bootsma H, Kroese FGM. Epithelial-immune cell interplay in primary Sjogren syndrome salivary gland pathogenesis. Nat Rev Rheumatol, 2012, 17:333–48., DOI 10.1038/s41584-021-00605-2 Venables PJ. Sjogren’s syndrome. Best Pract Res Clin Rheumatol, 2004, 18: 313–29., DOI 10.1016/j.berh.2004.02.010 Shiboski SC, Shiboski CH, Criswell L, Baer A, Challacombe S, Lanfranchi H, et al. Sjogren’s international collaborative clinical alliance, SICCA, research groups, 2012, American college of rheumatology classification criteria for sjogren’s syndrome: A data-driven, expert consensus approach in the sjogren’s international collaborative clinical alliance cohort. Arthritis Care Res, Hoboken, 64 :475–87., DOI 10.1002/acr.21591 Daniels TE, Cox D, Shiboski CH, Schiodt M, Wu A, Lanfranchi H, et al. Sjogren’s International Collaborative Clinical Alliance Research Groups, 2011, Associations between salivary gland histopathologic diagnoses and phenotypic features of Sjogren’s syndrome among 1,726 registry participants. Arthritis Rheum 63: 2021–30., DOI 10.1002/art.30381 Nordmark G, Alm GV, Ronnblom L. Mechanisms of disease: Primary sjogren’s syndrome and the type I interferon system. Nat Clin Pract Rheumatol, 2006, 2:262–9., DOI 10.1038/ncprheum0173 Cornec D, Devauchelle-Pensec V, Tobon GJ, Pers JO, Jousse-Joulin S, Saraux A. B cells in sjogren’s syndrome: From pathophysiology to diagnosis and treatment. J Autoimmun, 2012, 39:161–7., DOI 10.1016/j.jaut.2012.05.014 Chen YH, Wang XY, Jin X1, Yang Z1, Xu J. Rituximab therapy for primary sjogren’s syndrome. Front Pharmacol, 2021, 12:731122., DOI 10.3389/fphar.2021. 731122 Gandolfo S, De Vita S. Double anti-B cell and anti-BAFF targeting for the treatment of primary Sjogren’s syndrome. Clin Exp Rheumatol, 2019, 37: 199–208. Zandonella Callegher S, Giovannini I, Zenz S, Manfre V, Stradner MH, Hocevar A, et al. Sjogren syndrome: Looking forward to the future. Ther Adv Musculoskelet Dis, 2022, 14:1759720X221100295., DOI 10.1177/ 1759720X221100295 Menendez A, Gomez J, Caminal-Montero L, Diaz-Lopez JB, Cabezas- Rodriguez I, Mozo L. Common and specific associations of anti-SSA/Ro60 and anti-Ro52/TRIM21 antibodies in systemic lupus erythematosus. Sci World J, 2013, 2013:832789., DOI 10.1155/2013/832789 Sanchez-Montalva A, Fernandez-Luque A, Simeon CP, Fonollosa-Pla V, Marin A, Guillen A, et al. Anti-SSA/Ro52 autoantibodies in scleroderma: Results of an observational, cross-sectional study. Clin Exp Rheumatol, 2014, 32:177–82. Smeele HTW, Schreurs MWJ, Costedoat-Chalumeau N, Cornette JMJ, Dolhain RJEM. Low prevalence of anti-SSA, anti-Ro, and anti-SSB, anti-La, autoantibodies in female patients with rheumatoid arthritis with a wish to conceive. RMD Open, 2021, 7:e001727., DOI 10.1136/rmdopen-2021- 001727 Theander E, Jonsson R, Sjostrom B, Brokstad K, Olsson P, Henriksson G. Prediction of Sjogren’s syndrome years before diagnosis and identification of patients with early onset and severe disease course by autoantibody profiling. Arthritis Rheumatol, 2015, 67:2427–36., DOI 10. 1002/art.39214 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1610905 EP 1610911 DI 10.3389/pore.2023.1610905 PG 7 ER PT J AU Vlahovic, I Rajc, J Svagelj, I Solic, K Svagelj, D AF Vlahovic, Ivan Rajc, Jasmina Svagelj, Ivan Solic, Kresimir Svagelj, Drazen TI Potential predictors for CDX2 expression loss and mismatch repair deficiency in colorectal cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; colorectal cancer (CRC); CDX2; dMMR; regression model ID immunohistochemistry; colorectal cancer (CRC); CDX2; dMMR; regression model AB CDX2 expression loss is commonly associated with mismatch repair deficiency (dMMR) in colorectal cancer (CRC). However, there are only a few studies that have attempted to correlate CDX2 expression loss with specific MMR genes (MLH1, MSH2, MSH6, PMS2). This is a retrospective study of 327 patients who underwent surgery due to CRC. Nine patients (2.9%) had two synchronous CRCs, making the total sample 336 CRC. Histopathological data such as tumor type, tumor grade, perineural, lymphatic, and vascular invasion, pT stage, pN stage, peritumoral and intratumoral lymphocytic infiltration were collected and recorded in the database. After immunohistochemical analysis, CDX2 expression, MLH1, MSH2, MSH6, and PMS2 deficiency were also recorded. CDX2 expression loss was detected in 19 out of 336 CRCs (5.9%) and was associated with ascending colon CRC, partially mucinous adenocarcinoma, poorly differentiated carcinoma, and dMMR. Forty-four (13.1%) of CRCs were dMMR. We found a statistically significant association between CDX2 expression loss and MLH1 and PMS2 deficiency. Considering that most expression phenotypes include pairs of MMR genes, we analyzed MLH1/PMS2 and MSH2/MSH6 as heterodimers. Analysis of heterodimers showed a similar result, namely, that MLH1/PMS2 heterodimer deficiency was significantly associated with CDX2 expression loss. We also constructed a regression model for CDX2 expression loss and for dMMR. Poor tumor differentiation and MLH1/PMS2 heterodimer deficiency have been identified as potential predictors for CDX2 expression loss. CRCin the ascending colon and CDX2 expression loss have been identified as positive potential predictors of dMMR with rectal cancer as negative potential predictor of dMMR.Our study showed a significant association betweenCDX2expression loss and MLH1 and PMS2 deficiency in CRC. We also managed to produce a regression model for CDX2 expression and showed that poor tumor differentiation and MLH1/ PMS2 heterodimer deficiency are independent factors for CDX2 expression loss. We were the first to include CDX2 expression in a regression model for dMMR and showed thatCDX2 expression loss can be used as a predictive factor for dMMR,which should be confirmed by further studies. C1 [Vlahovic, Ivan] University of Osijek, Faculty of Medicine, Clinical Hospital Center Osijek, Department of Abdominal SurgeryOsijek, Croatia. [Rajc, Jasmina] University of Osijek, Faculty of Medicine, Clinical Hospital Center Osijek, Department of Pathology and Forensic MedicineOsijek, Croatia. [Svagelj, Ivan] General County Hospital Vinkovci, Department of Pathology and CytologyVinkovci, Croatia. [Solic, Kresimir] University of Osijek, Faculty of Medicine, Clinical Hospital Center Osijek, Department of Medical Statistics and Medical InformaticsOsijek, Croatia. [Svagelj, Drazen] General County Hospital Vinkovci, Department of Pathology and CytologyVinkovci, Croatia. RP Vlahovic, I (reprint author), University of Osijek, Faculty of Medicine, Clinical Hospital Center Osijek, Department of Abdominal Surgery, Osijek, Croatia. EM ivan_vlahovic@hotmail.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Saad RS, Ghorab Z, Khalifa MA, Xu M. CDX2 as a marker for intestinal differentiation: Its utility and limitations. World J Gastrointest Surg, 2011, 3(11): 159–66., DOI 10.4240/wjgs.v3.i11.159 Asgari-Karchekani S, Karimian M, Mazoochi T, Taheri MA, Khamehchian T. CDX2 protein expression in colorectal cancer and ItsCorrelation with clinical and pathological characteristics, prognosis, and survival rate of patients. J Gastrointest Cancer, 2020, 51(3):844–9., DOI 10.1007/s12029-019-00314-w Olsen J, Eiholm S, Kirkeby LT, Espersen MLM, Jess P, Gogenur I, et al. CDX2 downregulation is associated with poor differentiation and MMR deficiency in colon cancer. Exp Mol Pathol, 2016, 100(1):59–66., DOI 10.1016/j.yexmp.2015. 11.009 Dawson H, Koelzer VH, Lukesch AC, Mallaev M, Inderbitzin D, Lugli A, et al. Loss of Cdx2 expression in primary tumors and lymph node metastases is specific for mismatch repair-deficiency in colorectal cancer. Front Oncol, 2013, 3:265–6., DOI 10.3389/fonc.2013.00265 Bae JM, Lee TH, Cho NY, Kim TY, Kang GH. Loss of CDX2 expression is associated with poor prognosis in colorectal cancer patients. World J Gastroenterol, 2015, 21(5):1457–67., DOI 10.3748/wjg.v21.i5.1457 Dalerba P, Sahoo D, Paik S, Guo X, Yothers G, Song N, et al. CDX2 as a prognostic biomarker in stage II and stage III colon cancer. N Engl J Med, 2016, 374(3):211–22., DOI 10.1056/NEJMoa1506597 Alarid-Escudero F, Schrag D, Kuntz KM. CDX2 biomarker testing and adjuvant therapy for stage II colon cancer: An exploratory cost-effectiveness analysis. Value Heal, 2022, 25(3):409–18., DOI 10.1016/j.jval.2021.07.019 Brierly JD, Gospodarowicz MKWC. TNM classification of malignant tumours. 8th ed. New Jersey, United States: Wiley, 2017). Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, et al. The 2019 WHO classification of tumours of the digestive system. Histopathology, 2020, 76(2):182–8., DOI 10.1111/his.13975 Hestetun KE, Aasebo K, Rosenlund NB, Muller Y, Dahl O, Myklebust MP. Mismatch repair phenotype determines the implications of tumor grade and CDX2 expression in stage II–III colon cancer. Mod Pathol, 2021, 34(1):161–70., DOI 10.1038/s41379-020-0634-9 Hashmi AA, Ali R, Hussain ZF, Faridi N, Khan EY, Bakar SMA, et al. Mismatch repair deficiency screening in colorectal carcinoma by a four-antibody immunohistochemical panel in Pakistani population and its correlation with histopathological parameters. World J Surg Oncol, 2017, 15(1):116., DOI 10.1186/ s12957-017-1158-8 Neumann J, Heinemann V, Engel J, Kirchner T, Stintzing S. The prognostic impact of CDX2 correlates with the underlying mismatch repair status and BRAF mutational status but not with distant metastasis in colorectal cancer. Virchows Arch, 2018, 473(2):199–207., DOI 10.1007/ s00428-018-2360-y Konukiewitz B, Schmitt M, Silva M, Pohl J, Lang C, Steiger K, et al. Loss of CDX2 in colorectal cancer is associated with histopathologic subtypes and microsatellite instability but is prognostically inferior to hematoxylin–eosinbased morphologic parameters from the WHO classification. Br J Cancer, 2021, 125(12):1632–46., DOI 10.1038/s41416-021-01553-0 Kim JH, Rhee YY, Bae JM, Cho NY, Kang GH. Loss of CDX2/ CK20 expression is associated with poorly differentiated carcinoma, the CpG island methylator phenotype, and adverse prognosis in microsatellite-unstable colorectal cancer. Am J Surg Pathol, 2013, 37(10):1532–41., DOI 10.1097/PAS. 0b013e31829ab1c1 Bruun J, Sveen A, Barros R, Eide PW, Eilertsen I, Kolberg M, et al. Prognostic, predictive, and pharmacogenomic assessments of CDX2 refine stratification of colorectal cancer. Mol Oncol, 2018, 12(9):1639–55., DOI 10. 1002/1878-0261.12347 Baba Y, Nosho K, Shima K, Freed E, Irahara N, Philips J, et al. Relationship of CDX2 loss with molecular features and prognosis in colorectal cancer. Clin Cancer Res, 2009, 15(14):4665–73., DOI 10.1158/1078-0432.CCR-09-0401 Ribeirinho-Soares S, Padua D, Amaral AL, Valentini E, Azevedo D, Marques C, et al. Prognostic significance of MUC2, CDX2 and SOX2 in stage II colorectal cancer patients. BMC Cancer, 2021, 21(1):359–13., DOI 10.1186/s12885-021- 08070-6 Shigematsu Y, Inamura K, Mise Y, Saiura A, Rehnberg E, Yamamoto N, et al. CDX2 expression is concordant between primary colorectal cancer lesions and corresponding liver metastases independent of chemotherapy: A single-center retrospective study in Japan. Oncotarget, 2018, 9(24):17056–65., DOI 10.18632/ oncotarget.24842 Pilati C, Taieb J, Balogoun R, Marisa L, de Reynies A, Laurent-Puig P. CDX2 prognostic value in stage II/III resected colon cancer is related to CMS classification. Ann Oncol Off J Eur Soc Med Oncol, 2017, 28(5):1032–5., DOI 10.1093/ annonc/mdx066 Shigematsu Y, Inamura K, Yamamoto N, Mise Y, Saiura A, Ishikawa Y, et al. Impact of CDX2 expression status on the survival of patients after curative resection for colorectal cancer liver metastasis 11 Medical and Health Sciences 1112 Oncology and Carcinogenesis. BMC Cancer, 2018, 18(1):980., DOI 10.1186/s12885-018-4902-8 Slik K, Turkki R, Carpen O, Kurki S, Korkeila E, Sundstrom J, et al. CDX2 loss with microsatellite stable phenotype predicts poor clinical outcome in stage II colorectal carcinoma. Am J Surg Pathol, 2019, 43(11):1473–82., DOI 10.1097/PAS. 0000000000001356 Graule J, Uth K, Fischer E, Centeno I, Galvan JA, Eichmann M, et al. CDX2 in colorectal cancer is an independent prognostic factor and regulated by promoter methylation and histone deacetylation in tumors of the serrated pathway. Clin Epigenetics, 2018, 10(1):120., DOI 10.1186/s13148-018-0548-2 Melincovici CS, Bosca AB, Susman S, Cutas A, Marginean M, Ilea A, et al. Assessment of mismatch repair deficiency, CDX2, beta-catenin and E-cadherin expression in colon cancer: Molecular characteristics and impact on prognosis and survival - an immunohistochemical study. Rom J Morphol Embryol, 2020, 61(3): 715–27., DOI 10.47162/RJME.61.3.10 Rozek LS, Lipkin SM, Fearon ER, Hanash S, Giordano TJ, Greenson JK, et al. CDX2 polymorphisms, RNA expression, and risk of colorectal cancer. Cancer Res, 2005, 65(13):5488–92., DOI 10.1158/0008-5472.CAN-04-3645 Ohki D, Yamamichi N, Sakaguchi Y, Takahashi Y, Kageyama-Yahara N, YamamichiM, et al. Transcriptome of sessile serrated adenoma/polyps is associated with MSI- high colorectal cancer and decreased expression of CDX2. Cancer Med, 2022, 1–13:5066–5078., DOI 10.1002/cam4.4810 Ryan EJ, Creavin B, Khaw YL, Kelly ME,Mohan HM, Geraghty R, et al. Effects of CDX2 on prognosis and chemotherapy responsiveness in mismatch repairdeficient colorectal cancer. BJS Open, 2018, 2(6):456–63., DOI 10.1002/bjs5.91 Lugli A, Tzankov A, Zlobec I, Terracciano LM. Differential diagnostic and functional role of the multi-marker phenotype CDX2/CK20/CK7 in colorectal cancer stratified by mismatch repair status. Mod Pathol, 2008, 21(11):1403–12., DOI 10.1038/modpathol.2008.117 Dawson H, Galvan JA, Helbling M, Muller DE, Karamitopoulou E, Koelzer VH, et al. Possible role of Cdx2 in the serrated pathway of colorectal cancer characterized by BRAF mutation, high-level CpG Island methylator phenotype and mismatch repair-deficiency. Int J Cancer, 2014, 134(10):2342–51., DOI 10.1002/ijc. 28564 Tomasello G, Barni S, Turati L, Ghidini M, Pezzica E, Passalacqua R, et al. Association of CDX2 expression with survival in early colorectal cancer: A systematic review and meta-analysis. Clin Colorectal Cancer, 2018, 17(2): 97–103., DOI 10.1016/j.clcc.2018.02.001 De’angelis GL, Bottarelli L, Azzoni C, De’angelis N, Leandro G, Di Mario F, et al. Microsatellite instability in colorectal cancer. Acta Biomed, 2018, 89(6): 97–101., DOI 10.23750/abm.v89i9-S.7960 Boland CR, Goel A. Microsatellite instability in colorectal cancer. Gastroenterology, 2010, 138(6):2073–87., DOI 10.1053/j.gastro.2009.12.064 Sayar I, Akbas EM, Isik A, Gokce A, Peker K, Demirtas L, et al. Relationship among mismatch repair deficiency, CDX2 loss, p53 and E-cadherin in colon carcinoma and suitability of using a double panel of mismatch repair proteins by immunohistochemistry. Polish J Pathol, 2015, 66(3):246–53., DOI 10.5114/pjp.2015.54958 Toth C, Sukosd F, Valicsek E, Herpel E, Schirmacher P, Tiszlavicz L. Loss of CDX2 gene expression is associated with DNA repair proteins and is a crucial member of the Wnt signaling pathway in liver metastasis of colorectal cancer. Oncol Lett, 2018, 15(3):3586–93., DOI 10.3892/ol.2018.7756 Kim JH, Kang GH. Molecular and prognostic heterogeneity of microsatelliteunstable colorectal cancer. World J Gastroenterol, 2014, 20(15):4230–43., DOI 10. 3748/wjg.v20.i15.4230 Lee BC, Yu CS, Kim J, Lee JL, Kim CW, Yoon YS, et al. Clinicopathological features and surgical options for synchronous colorectal cancer. Med, United States,, 2017, 96(9):e6224., DOI 10.1097/MD.0000000000006224 Chikatani K, Chika N, Suzuki O, Sakimoto T, Ishibashi K, Eguchi H, et al. A model for Predicting DNA mismatch repair-deficient colorectal cancer. Anticancer Res, 2020, 40(8):4379–85., DOI 10.21873/anticanres.14441 Jenkins MA, Hayashi S, O’Shea AM, Burgart LJ, Smyrk TC, Shimizu D, et al. Pathology features in bethesda guidelines predict colorectal cancer microsatellite instability: A population-based study. Gastroenterology, 2007, 133(1):48–56., DOI 10. 1053/j.gastro.2007.04.044 Fujiyoshi K, Yamaguchi T, Kakuta M, Takahashi A, Arai Y, Yamada M, et al. Predictive model for high-frequency microsatellite instability in colorectal cancer patients over 50 years of age. Cancer Med, 2017, 6(6):1255–63., DOI 10.1002/cam4. 1088 Roman R, Verdu M, Calvo M, Vidal A, Sanjuan X, Jimeno M, et al. Microsatellite instability of the colorectal carcinoma can be predicted in the conventional pathologic examination. A prospective multicentric study and the statistical analysis of 615 cases consolidate our previously proposed logistic regression model. Virchows Arch, 2010, 456(5):533–41., DOI 10.1007/s00428-010- 0896-6 Greenson JK, Huang SC, Herron C, Moreno V, Bonner JD, Tomsho LP, et al. Pathologic predictors of microsatellite instability in colorectal cancer. Am J Surg Pathol, 2009, 33(1):126–33., DOI 10.1097/PAS.0b013e31817ec2b1 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1610908 EP 1610918 DI 10.3389/pore.2023.1610908 PG 11 ER PT J TI Characterization of the input material quality for the production of tisagenlecleucel by multiparameter flow cytometry and its relation to the clinical outcome SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunotherapy; CAR-T cells; tisagenlecleucel; B-cell lymphoma and leukemia; Kymriah ID immunotherapy; CAR-T cells; tisagenlecleucel; B-cell lymphoma and leukemia; Kymriah AB Tisagenlecleucel (tisa-cel) is a CD19-specific CAR-T cell product approved for the treatment of relapsed/refractory (r/r) DLBCL or B-ALL. We have followed a group of patients diagnosed with childhood B-ALL (n = 5), adult B-ALL (n = 2), and DLBCL (n = 25) who were treated with tisa-cel under non-clinical trial conditions. The goal was to determine how the intensive pretreatment of patients affects the produced CAR-T cells, their in vivo expansion, and the outcome of the therapy. Multiparametric flow cytometry was used to analyze the material used for manufacturing CAR-T cells (apheresis), the CAR-T cell product itself, and blood samples obtained at three timepoints after administration. We present the analysis of memory phenotype of CD4/ CD8 CAR-T lymphocytes (CD45RA, CD62L, CD27, CD28) and the expression of inhibitory receptors (PD-1, TIGIT). In addition, we show its relation to the patients’ clinical characteristics, such as tumor burden and sensitivity to prior therapies. Patients who responded to therapy had a higher percentage of CD8+CD45RA+CD27+ T cells in the apheresis, although not in the produced CAR-Ts. Patients with primary refractory aggressive B-cell lymphomas had the poorest outcomes which was characterized by undetectable CAR-T cell expansion in vivo. No clear correlation of the outcome with the immunophenotypes of CAR-Ts was observed. Our results suggest that an important parameter predicting therapy efficacy is CAR-Ts’ level of expansion in vivo but not the immunophenotype. After CAR-T cells’ administration, measurements at several timepoints accurately detect their proliferation intensity in vivo. The outcome of CAR-T cell therapy largely depends on biological characteristics of the tumors rather than on the immunophenotype of produced CAR-Ts. CR Siddiqi T, Abramson JS, Li D, Brown W, Devries T, Dave K, et al. Patient characteristics and pre-infusion biomarkers of inflammation correlate with clinical outcomes after treatment with the defined composition, CD19-targeted CAR T cell product, JCAR017. Blood, 2017, 130:193., DOI 10.1182/blood.V130.Suppl_1.193.193 Zhang X, Yang JF, Li JJ, Li WQ, Song D, Lu XA, et al. Factors associated with treatment response to CD19 CAR-T therapy among a large cohort of B cell acute lymphoblastic leukemia. Cancer Immunol Immunother, 2022, 71(3):689–703., DOI 10.1007/s00262-021-03009-z Martino M, Alati C, Canale FA, Musuraca G, Martinelli G, Cerchione C. A Review of clinical outcomes of CAR T-Cell therapies for B-Acute lymphoblastic leukemia. Int J Mol Sci, 2021, 22:2150., DOI 10.3390/ijms22042150 Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, McGuirk JP, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med, 2019, 380:45–56., DOI 10.1056/NEJMoa1804980 Vercellino L, Di Blasi R, Kanoun S, Tessoulin B, Rossi C, D’Aveni-Piney M, et al. Predictive factors of early progression after CAR T-cell therapy in relapsed/ refractory diffuse large B-cell lymphoma. Blood Adv, 2020, 4(22):5607–15., DOI 10. 1182/bloodadvances.2020003001 Riedell PA, Walling C, Nastoupil LJ, Pennisi M, Maziarz RT, McGuirk JP, et al. A multicenter retrospective analysis of clinical outcomes, toxicities, and patterns of use in institutions utilizing commercial axicabtagene ciloleucel and tisagenlecleucel for relapsed/refractory aggressive B-cell lymphomas. Blood, 2019, 134:1599., DOI 10. 1182/blood-2019-127490 Dean EA, Mhaskar RS, Lu H, Mousa MS, Krivenko GS, Lazaryan A, et al. High metabolic tumor volume is associated with decreased efficacy of axicabtagene ciloleucel in large B-cell lymphoma. Blood Adv, 2020, 4(14):3268–76., DOI 10. 1182/bloodadvances.2020001900 Monfrini C, Stella F, Aragona V, Magni M, Ljevar S, Vella C, et al. Phenotypic composition of commercial anti-CD19 CAR-T cells affects in vivo expansion and disease response in large B-cell lymphoma patients. Clin Cancer Res, 2022, 28: 3378–86., DOI 10.1158/1078-0432.ccr-22-0164 Cuffel A, Allain V, Faivre L, Di Blasi R, Morin F, Vercellino L, et al. Real-world characteristics of T-cell apheresis and clinical response to tisagenlecleucel in B-cell lymphoma. Blood Adv, 2022, 6:4657–60., DOI 10.1182/bloodadvances.2022007057 Byrne M, Oluwole OO, Savani B, Majhail NS, Hill BT, Locke FL. Understanding and managing large B cell lymphoma relapses after chimeric antigen receptor T cell therapy. Biol Blood Marrow Transplant, 2019, 25: e344–e351., DOI 10.1016/j.bbmt.2019.06.036 Blache U, Weiss R, Boldt A, Kapinsky M, Blaudszun AR, Quaiser A, et al. Advanced flow cytometry assays for immune monitoring of CAR-T cell applications. Front Immunol, 2021, 12:658314., DOI 10.3389/fimmu.2021. 658314 Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, et al. Determinants of response and resistance to CD19 chimeric antigen receptor, CAR, T cell therapy of chronic lymphocytic leukemia. Nat Med, 2018, 24(5): 563–71., DOI 10.1038/s41591-018-0010-1 Schuster SJ, Svoboda J, Chong EA, Nasta SD, Mato AR, Anak O, et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med, 2017, 377:2545–54., DOI 10.1056/NEJMoa1708566 Gattinoni L, Klebanoff CA, Palmer DC, Wrzesinski C, Kerstann K, Yu Z, et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J Clin Invest, 2005, 115(6):1616–26., DOI 10.1172/JCI24480 Sommermeyer D, Hudecek M, Kosasih PL, Gogishvili T, Maloney DG, Turtle CJ, et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia, 2016, 30(2):492–500., DOI 10.1038/leu. 2015.247 An L, Lin Y, Deng B, Yin Z, Zhao D, Ling Z, et al. Humanized CD19 CAR-T cells in relapsed/refractory B-ALL patients who relapsed after or failed murine CD19 CAR-T therapy. BMC Cancer, 2022, 22(1):393., DOI 10.1186/s12885-022- 09489-1 Park JH, Riviere I, Gonen M, Wang X, Senechal B, Curran KJ, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med, 2018, 378(5):449–59., DOI 10.1056/NEJMoa1709919 Stach M, Ptackova P, Mucha M, Musil J, Klener P, Otahal P. Inducible secretion of IL-21 augments anti-tumor activity of piggyBac-manufactured chimeric antigen receptor T cells. Cytotherapy, 2020, 22(12):744–54., DOI 10. 1016/j.jcyt.2020.08.005 Cheson BD, Fisher RI, Barrington SF, Cavalli F, Schwartz LH, Zucca E, et al. Recommendations for initial evaluation, staging, and response assessment of hodgkin and non-hodgkin lymphoma: The lugano classification. J Clin Oncol, 2014, 32(27):3059–68., DOI 10.1200/JCO.2013.54.8800 Kratochvil M, Koladiya A, Vondrasek J. Generalized EmbedSOM on quadtree-structured self-organizing maps. F1000Res, 2020, 8:2120,, DOI 10.12688/ f1000research.21642.2 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1610914 EP 1610925 DI 10.3389/pore.2023.1610914 PG 12 ER PT J AU Horesh, A Pollack, R Nechushtan, H Dresner-Pollak, R Neuman, T AF Horesh, Adi Pollack, Rena Nechushtan, Hovav Dresner-Pollak, Rivka Neuman, Tzahi TI Tumor PD-L1 expression and molecular profiling are not associated with immune checkpoint inhibitor-induced thyroid dysfunction in advanced NSCLC patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE NSCLC; Non-small cell lung cancer; PD-1; programmed cell death 1; PD-L1; programmed cell death ligand 1 ID NSCLC; Non-small cell lung cancer; PD-1; programmed cell death 1; PD-L1; programmed cell death ligand 1 AB Background: Immune-checkpoint inhibitors (ICIs) have revolutionized the treatment of advanced non-small cell lung cancer (NSCLC), however are frequently associated with thyroid immune-related adverse events (IRAEs). We investigated the association between patient characteristics, tumor PDL1 expression and molecular profile with the development of thyroid IRAEs in NSCLC patients. Methods: Single center, retrospective study including 107 NSCLC patients treated with PD-1/PD-L1 inhibitors from April 2016 to July 2020. All patients were euthyroid at baseline with at least two TSH measurements post-treatment initiation. The primary outcome was the difference in tumor PD-L1 expression in patients who developed any thyroid IRAEs versus those who remained euthyroid. Additional outcomes included development of overt thyroid dysfunction, the association of specific molecular alterations with thyroid IRAEs, and onset of thyroid IRAEs as a function of tumor PD-L1 expression. Results: Overall, 37 (34.6%) patients developed any thyroid dysfunction and 18 (16.8%) developed overt thyroid dysfunction. Tumor PD-L1 staining intensity was not associated with thyroid IRAEs. TP53 mutation was less likely to be associated with any thyroid dysfunction (p < 0.05) and no association was found between EGFR, ROS, ALK or KRAS mutations. There was no association between PD-L1 expression and time to develop thyroid IRAEs. Conclusion: PD-L1 expression is not associated with the development of thyroid dysfunction in advanced NSCLC patients treated with ICIs, suggesting that thyroid IRAEs are unrelated to tumor PD-L1 expression. C1 [Horesh, Adi] Hebrew University of Jerusalem, The Faculty of MedicineJerusalem, Israel. [Pollack, Rena] Hebrew University of Jerusalem, The Faculty of MedicineJerusalem, Israel. [Nechushtan, Hovav] Hebrew University of Jerusalem, The Faculty of MedicineJerusalem, Israel. [Dresner-Pollak, Rivka] Hebrew University of Jerusalem, The Faculty of MedicineJerusalem, Israel. [Neuman, Tzahi] Hebrew University of Jerusalem, The Faculty of MedicineJerusalem, Israel. RP Neuman, T (reprint author), Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel. EM tneuman@hadassah.org.il CR Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–small-cell Lung Cancer. N Engl J Med, 2016, 375:1823–33., DOI 10.1056/nejmoa1606774 Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab Plus Chemotherapy in Metastatic Non–small-cell Lung Cancer. N Engl J Med, 2018, 378:2078–92., DOI 10.1056/nejmoa1801005 Scott ES, Long GV, Guminski A, Clifton-Bligh RJ,Menzies AM, Tsang VH. The Spectrum, Incidence, Kinetics andManagement of Endocrinopathies with Immune Checkpoint Inhibitors for Metastatic Melanoma. Eur J Endocrinol, 2018, 178: 173–80., DOI 10.1530/EJE-17-0810 Morganstein DL, Lai Z, Spain L, Diem S, Levine D, Mace C, et al. Thyroid Abnormalities Following the Use of Cytotoxic T-Lymphocyte Antigen-4 and Programmed Death Receptor Protein-1 Inhibitors in the Treatment of Melanoma. Clin Endocrinol, Oxf,, 2017, 86:614–20., DOI 10.1111/cen.13297 D’aiello A, Lin J, Gucalp R, Tabatabaie V, Cheng H, Bloomgarden NA, et al. Thyroid Dysfunction in Lung Cancer Patients Treated with Immune Checkpoint Inhibitors, Icis): Outcomes in a Multiethnic Urban Cohort. Cancers, Basel,, 2021, 13:1464., DOI 10.3390/cancers13061464 Brody HM, Macherla S, Bulumulle A, Namireddy P, Cherry CR. The Real- World Incidence of Immunotherapy-Related Thyroid Dysfunction: A Retrospective Analysis of a Single center’s Experience over Five Years. J Clin Oncol, 2020, 38:98., DOI 10.1200/jco.2020.38.5_suppl.98 Hosoya K, Fujimoto D, Morimoto T, Kumagai T, Tamiya A, Taniguchi Y, et al. Association between Early Immune-Related Adverse Events and Clinical Outcomes in Patients with Non–small Cell Lung Cancer Treated with Immune Checkpoint Inhibitors. Clin Lung Cancer, 2020, 21:e315–28., DOI 10.1016/j.cllc. 2020.01.003 Freeman-Keller M, Kim Y, Cronin H, Richards A, Gibney G, Weber JS. Nivolumab in Resected and Unresectable Metastatic Melanoma: Characteristics of Immune-Related Adverse Events and Association with Outcomes. Clin Cancer Res, 2016, 22:886–94., DOI 10.1158/1078-0432.CCR-15-1136 Cortellini A, Friedlaender A, Banna GL, Porzio G, Bersanelli M, Cappuzzo F, et al. Immune-related Adverse Events of Pembrolizumab in a Large Real-World Cohort of Patients with NSCLC with a PD-L1 Expression ≥ 50% and Their Relationship with Clinical Outcomes. Clin Lung Cancer, 2020, 21:498–508.e2., DOI 10.1016/j.cllc.2020.06.010 Baldini E, Lunghi A, Cortesi E, Turci D, Signorelli D, Stati V, et al. Immune-related Adverse Events Correlate with Clinical Outcomes in NSCLC Patients Treated with Nivolumab: The Italian NSCLC Expanded Access Program. Lung Cancer, 2020, 140:59–64., DOI 10.1016/j.lungcan.2019. 12.014 Narita T, Oiso N, Taketomo Y, Okahashi K, Yamauchi K, Sato M, et al. Serological Aggravation of Autoimmune Thyroid Disease in Two Cases Receiving Nivolumab. J Dermatol, 2016, 43:210–4., DOI 10.1111/1346-8138.13028 Hollowell JG, Staehling NW, Dana Flanders W, Hannon WH, Gunter EW, Spencer CA, et al. Serum TSH, T4, and Thyroid Antibodies in the United States Population, 1988 to 1994): National Health and Nutrition Examination Survey, NHANES III). J Clin Endocrinol Metab, 2002, 87:489–99., DOI 10.1210/jcem.87.2. 8182 Kobayashi T, Iwama S, Yasuda Y, Okada N, Tsunekawa T, Onoue T, et al. Patients with Antithyroid Antibodies Are Prone to Develop Destructive Thyroiditis by Nivolumab: A Prospective Study. J Endocr Soc, 2018, 2:241–51., DOI 10.1210/JS. 2017-00432 Osorio JC, Ni A, Chaft JE, Pollina R, Kasler MK, Stephens D, et al. Antibodymediated Thyroid Dysfunction during T-Cell Checkpoint Blockade in Patients with Non-small-cell Lung Cancer. Ann Oncol, 2017, 28:583–9., DOI 10.1093/annonc/ mdw640 Iwama S, Kobayashi T, Yasuda Y, Okuji T, Ito M, Ando M, et al. Increased Risk of Thyroid Dysfunction by PD-1 and CTLA-4 Blockade in Patients without Thyroid Autoantibodies at Baseline. J Clin Endocrinol Metab Published Online First, 2021, 107:e1620–e1630., DOI 10.1210/CLINEM/DGAB829 Wang Z, Aguilar EG, Luna JI, Dunai C, Khuat LT, Le CT, et al. Paradoxical Effects of Obesity on T Cell Function during Tumor Progression and PD-1 Checkpoint Blockade. Nat Med, 2019, 25:141–51., DOI 10.1038/s41591-018-0221-5 Pollack RM, Kagan M, Lotem M, Dresner-Pollak R. Baseline TSH Level Is Associated with Risk of Anti–PD-1–induced Thyroid Dysfunction. Endocr Pract, 2019, 25:824–9., DOI 10.4158/EP-2018-0472 Fuchs CS, Doi T, Jang RW, Muro K, Satoh T, Machado M, et al. Safety and Efficacy of Pembrolizumab Monotherapy in Patients with Previously Treated Advanced Gastric and Gastroesophageal junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA Oncol, 2018, 4:e180013., DOI 10.1001/jamaoncol. 2018.0013 Pollack R, Ashash A, Cahn A, Rottenberg Y, Stern H, Dresner-Pollak R. Immune Checkpoint Inhibitor-Induced Thyroid Dysfunction Is Associated with Higher Body Mass Index. J Clin Endocrinol Metab, 2020, 105:dgaa458–8., DOI 10. 1210/CLINEM/DGAA458 Ettinger DS, Wood DE, Chair V. Continue NCCN Guidelines Panel Disclosures NCCN Guidelines Version 7.2021 Non-small Cell Lung Cancer, 2021). Neuman T, London M, Kania-Almog J, Litvin A, Zohar Y, Fridel L, et al. A Harmonization Study for the Use of 22C3 PD-L1 Immunohistochemical Staining on Ventana’s Platform. J Thorac Oncol, 2016, 11:1863–8., DOI 10.1016/J.JTHO.2016. 08.146 Basak EA, Van Der Meer JWM, Hurkmans DP, Schreurs MWJ, Oomen-de Hoop E, van der Veldt AAM, et al. Overt Thyroid Dysfunction and Anti-thyroid Antibodies Predict Response to Anti-PD-1 Immunotherapy in Cancer Patients. Thyroid, 2020, 30:966–73., DOI 10.1089/THY.2019.0726 Sugisaka J, Yukihoro T, Taguri M, Kawashima Y, Aiba T, Kawana S, et al. Relationship between Programmed Cell Death Protein Ligand 1 Expression and Immune-Related Adverse Events in Non-small-cell Lung Cancer Patients Treated with Pembrolizumab. JMA J, 2019, 3:58–66., DOI 10.31662/jmaj.2019-0005 Lisberg A, Andrew Tucker D, Goldman JW,Wolf B, Carroll J, Hardy A, et al. Treatment-related Adverse Events Predict Improved Clinical Outcome in Nsclc Patients on KEYNOTE-001 at a Single center. Cancer Immunol Res, 2018, 6: 288–94., DOI 10.1158/2326-6066.CIR-17-0063 Peters S, Gettinger S, Johnson ML, Janne PA, Garassino MC, Christoph D, et al. Phase II Trial of Atezolizumab as First-Line or Subsequent Therapy for Patients with Programmed Death-Ligand 1-Selected Advanced Non-small-cell Lung Cancer, BIRCH). J Clin Oncol, 2017, 35:2781–9., DOI 10.1200/JCO.2016.71. 9476 WangM, Attardi LD. A Balancing Act: P53 Activity from Tumor Suppression to Pathology and Therapeutic Implications. Review, 2021, 17., DOI 10.1146/ ANNUREV-PATHOL-042320-025840 Gudkov AV, Komarova EA. The Role of P53 in Determining Sensitivity to Radiotherapy. Nat Rev Cancer, 2003, 3:117–29., DOI 10.1038/nrc992 Blagih J, Zani F, Chakravarty P, Hennequart M, Pilley S, Hobor S, et al. Cancer-Specific Loss of P53 Leads to a Modulation of Myeloid and T Cell Responses. Cell Rep, 2020, 30:481–96.e6., DOI 10.1016/J.CELREP.2019.12.028 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1610951 EP 1610958 DI 10.3389/pore.2023.1610951 PG 8 ER PT J AU Li, Y Xiong, Ch Wu, LL Zhang, YB Wu, Sh Chen, FY Xu, HQ Liao, FH AF Li, Yue Xiong, Chao Wu, Li Li Zhang, Yuan Bo Wu, Sha Chen, Fen Yu Xu, Hua Qi Liao, Fei Hong TI Tumor subtypes and signature model construction based on chromatin regulators for better prediction of prognosis in uveal melanoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; uveal melanoma; TCGA; chromatin regulators; tumor subtypes ID prognosis; uveal melanoma; TCGA; chromatin regulators; tumor subtypes AB Background: Uveal Melanoma (UM) is the most prevalent primary intraocular malignancy in adults. This study assessed the importance of chromatin regulators (CRs) in UM and developed a model to predict UM prognosis. Methods: Gene expression data and clinical information for UM were obtained from public databases. Samples were typed according to the gene expression of CRs associated with UM prognosis. The prognostic key genes were further screened by the protein interaction network, and the risk model was to predict UMprognosis using the least absolute shrinkage and selection operator (LASSO) regression analysis and performed a test of the risk mode. In addition, we performed gene set variation analysis, tumor microenvironment, and tumor immune analysis between subtypes and risk groups to explore the mechanisms influencing the development of UM. Results: We constructed a signature model consisting of three CRs (RUVBL1, SIRT3, and SMARCD3), which was shown to be accurate, and valid for predicting prognostic outcomes in UM. Higher immune cell infiltration in poor prognostic subtypes and risk groups. The Tumor immune analysis and Tumor Immune Dysfunction and Exclusion (TIDE) score provided a basis for clinical immunotherapy in UM. Conclusion: The risk model has prognostic value for UM survival and provides new insights into the treatment of UM. C1 [Li, Yue] Nanchang University, School of Ophthalmology and OptometryNanchang, Jiangxi, China. [Xiong, Chao] Nanchang University, School of Ophthalmology and OptometryNanchang, Jiangxi, China. [Wu, Li Li] Nanchang University, School of Ophthalmology and OptometryNanchang, Jiangxi, China. [Zhang, Yuan Bo] Nanchang University, School of Ophthalmology and OptometryNanchang, Jiangxi, China. [Wu, Sha] Nanchang University, School of Ophthalmology and OptometryNanchang, Jiangxi, China. [Chen, Fen Yu] Nanchang University, School of Ophthalmology and OptometryNanchang, Jiangxi, China. [Xu, Hua Qi] Nanchang University, School of Ophthalmology and OptometryNanchang, Jiangxi, China. [Liao, Fei Hong] Nanchang University, School of Ophthalmology and OptometryNanchang, Jiangxi, China. RP Liao, FH (reprint author), Nanchang University, School of Ophthalmology and Optometry, Nanchang, China. EM lhfzf@126.com CR Kaur J, Malik MA, Gulati R, Azad SV, Goswami S. Genetic determinants of uveal melanoma. Tumour Biol, 2014, 35:11711–7., DOI 10.1007/s13277-014-2681-7 Kaliki S, Shields CL. Uveal melanoma: Relatively rare but deadly cancer. Eye, 2016, 31(2):241–57., DOI 10.1038/eye.2016.275 Hoiom V, Helgadottir H. The genetics of uveal melanoma: Current insights. Appl Clin Genet, 2016, 9:147–55., DOI 10.2147/tacg.s69210 Augsburger JJ, Correa ZM, Shaikh AH. Effectiveness of treatments for metastatic uveal melanoma. Am J Ophthalmol, 2009, 148(1):119–27., DOI 10. 1016/j.ajo.2009.01.023 Rietschel P, Panageas KS, Hanlon C, Patel A, Abramson DH, Chapman PB. Variates of survival in metastatic uveal melanoma. J Clin Oncol, 2005, 23(31): 8076–80., DOI 10.1200/jco.2005.02.6534 Patrone S, Maric I, Rutigliani M, Lanza F, Puntoni M, Banelli B, et al. Prognostic value of chromosomal imbalances, gene mutations, and BAP1 expression in uveal melanoma. Genes, Chromosomes, and Cancer, 2018, 57(8):387–400., DOI 10.1002/gcc.22541 Damato B. Ocular treatment of choroidal melanoma in relation to the prevention of metastatic death – a personal view. Prog Retin Eye Res, 2018, 66: 187–99., DOI 10.1016/j.preteyeres.2018.03.004 Smit KN, Jager MJ, de Klein A, Kiliҫ E. Uveal melanoma: Towards a molecular understanding. Prog Retin Eye Res, 2020, 75:100800., DOI 10.1016/j.preteyeres.2019. 100800 Onken MD, Worley LA, Ehlers JP, Harbour JW. Gene expression profiling in uveal melanoma reveals twomolecular classes and predictsmetastatic death. Cancer Res, 2004, 64(20):7205–9., DOI 10.1158/0008-5472.CAN-04-1750 Jager MJ, Brouwer NJ, Esmaeli B. The cancer genome Atlas Project: An integrated molecular view of uveal melanoma. Ophthalmology, 2018, 125(8): 1139–42., DOI 10.1016/j.ophtha.2018.03.011 Bol KF, van den Bosch T, Schreibelt G, Mensink HW, Keunen JEE, Kilic E, et al. Adjuvant dendritic cell vaccination in high-risk uveal melanoma. Ophthalmology, 2016, 123(10):2265–7., DOI 10.1016/j.ophtha.2016.06.027 Verdegaal EM. Adoptive cell therapy: A highly successful individualized therapy for melanoma with great potential for other malignancies. Curr Opin Immunol, 2016, 39:90–5., DOI 10.1016/j.coi.2016.01.004 Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. New Engl J Med, 2015, 373(1):23–34., DOI 10.1056/nejmoa1504030 Weber JS, D’Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment, CheckMate 037): A randomised, controlled, open-label, phase 3 trial. Lancet Oncol, 2015, 16(4):375–84., DOI 10. 1016/s1470-2045(15)70076-8 Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. New Engl J Med, 2010, 363(8):711–23., DOI 10.1056/nejmoa1003466 Xue M, Shang J, Chen B, Yang Z, Song Q, Sun X, et al. Identification of prognostic signatures for predicting the overall survival of uveal melanoma patients. J Cancer, 2019, 10(20):4921–31., DOI 10.7150/jca.30618 Wan Q, Tang J, Lu J, Jin L, Su Y, Wang S, et al. Six-gene-based prognostic model predicts overall survival in patients with uveal melanoma. Cancer Biomarkers, 2020, 27(3):343–56., DOI 10.3233/cbm-190825 Li Y, Yang X, Yang J,WangH,Wei W. An 11-gene-based prognostic signature for uveal melanoma metastasis based on gene expression and DNA methylation profile. J Cell Biochem, 2018, 120(5):8630–9., DOI 10.1002/jcb.28151 Sharma A, Stei MM, Frohlich H, Holz FG, Loeffler KU, Herwig-Carl MC. Genetic and epigenetic insights into uveal melanoma. Clin Genet, 2018, 93(5): 952–61., DOI 10.1111/cge.13136 Robertson AG, Shih J, Yau C, Gibb EA, Oba J, Mungall KL, et al. Integrative analysis identifies four molecular and clinical subsets in uveal melanoma. Cancer cell, 2017, 32(2):204–20.e15., DOI 10.1016/j.ccell.2017.07.003 Herlihy N, Dogrusoz M, van Essen TH, Harbour JW, van der Velden PA, van Eggermond MC, et al. Skewed expression of the genes encoding epigenetic modifiers in high-risk uveal melanoma. Invest Ophthalmol Vis Sci, 2015, 56(3): 1447–58., DOI 10.1167/iovs.14-15250 Field MG, Kuznetsov JN, Bussies PL, Cai LZ, Alawa KA, Decatur CL, et al. BAP1 loss is associated with DNA methylomic repatterning in highly aggressive Class 2 uveal melanomas. Clin Cancer Res, 2019, 25:5663–73., DOI 10.1158/1078- 0432.CCR-19-0366 Landreville S, Agapova OA, Matatall KA, Kneass ZT, Onken MD, Lee RS, et al. Histone deacetylase inhibitors induce growth arrest and differentiation in uveal melanoma. Clin Cancer Res, 2012, 18:408–16., DOI 10.1158/1078-0432.CCR-11- 0946 Matatall KA, Agapova OA, Onken MD, Worley LA, Bowcock AM, Harbour JW. BAP1 deficiency causes loss of melanocytic cell identity in uveal melanoma. BMC Cancer, 2013, 13:371., DOI 10.1186/1471-2407-13-371 Kuznetsov JN, Aguero TH, Owens DA, Kurtenbach S, Field MG, Durante MA, et al. BAP1 regulates epigenetic switch from pluripotency to differentiation in developmental lineages giving rise to BAP1-mutant cancers. Sci Adv, 2019, 5: eaax1738., DOI 10.1126/sciadv.aax1738 Deng S, Calin GA, Croce CM, Coukos G, Zhang L. Mechanisms ofmicroRNA deregulation in human cancer. Cell Cycle, 2008, 7:2643–6., DOI 10.4161/cc.7.17.6597 Chen X, He D, Da Dong X, Dong F,Wang J,Wang L, et al. MicroRNA-124a is epigenetically regulated and acts as a tumor suppressor by controlling multiple targets in uveal melanoma. Investig Ophthalmol Vis Sci, 2013, 54:2248–56., DOI 10. 1167/iovs.12-10977 Chen X, Wang J, Shen H, Lu J, Li C, Hu D-N, et al. Epigenetics, microRNAs, and carcinogenesis: Functional role of microRNA-137 in uveal melanoma. Investig Ophthalmol Vis Sci, 2011, 52:1193–9., DOI 10.1167/iovs.10-5272 Plass C, Pfister SM, Lindroth AM, Bogatyrova O, Claus R, Lichter P. Mutations in regulators of the epigenome and their connections to global chromatin patterns in cancer. Nat Rev Genet, 2013, 14(11):765–80., DOI 10.1038/ nrg3554 Gonzalez-Perez A, Jene-Sanz A, Lopez-Bigas N. The mutational landscape of chromatin regulatory factors across 4,623 tumor samples. Genome Biol, 2013, 14(9):r106., DOI 10.1186/gb-2013-14-9-r106 Medvedeva YA, Lennartsson A, Ehsani R, Kulakovskiy IV, Vorontsov IE, Panahandeh P, et al. EpiFactors: A comprehensive database of human epigenetic factors and complexes. Database, 2015, 2015:bav067., DOI 10.1093/database/bav067 Marazzi I, Greenbaum BD, Low DHP, Guccione E. Chromatin dependencies in cancer and inflammation. Nat Rev Mol Cel Biol, 2017, 19(4):245–61., DOI 10.1038/ nrm.2017.113 Li T, Yang J, Yang B, Zhao G, Lin H, Liu Q, et al. Ketamine inhibits ovarian cancer cell growth by regulating the lncRNA-PVT1/EZH2/p57 Axis. Front Genet, 2021, 11:597467., DOI 10.3389/fgene.2020.597467 Chu Y, Chen W, Peng W, Liu Y, Xu L, Zuo J, et al. Amnion-derived mesenchymal stem cell exosomes-mediated autophagy promotes the survival of trophoblasts under hypoxia through mTOR pathway by the downregulation of EZH2. Front Cel Develop Biol, 2020, 8:545852., DOI 10.3389/fcell.2020.545852 Chen J, Wang F, Xu H, Xu L, Chen D, Wang J, et al. Long non-coding RNA SNHG1 regulates the wnt/β-catenin and PI3K/AKT/mTOR signaling pathways via EZH2 to affect the proliferation, apoptosis, and autophagy of prostate cancer cell. Front Oncol, 2020, 10:552907., DOI 10.3389/fonc.2020.552907 Damaschke NA, Yang B, Blute ML, Lin CP, Huang W, Jarrard DF. Frequent disruption of chromodomain helicase DNA-binding protein 8, CHD8, and functionally associated chromatin regulators in prostate cancer. Neoplasia, 2014, 16(12):1018–27., DOI 10.1016/j.neo.2014.10.003 Lu J, Xu J, Li J, Pan T, Bai J, Wang L, et al. Facer: Comprehensive molecular and functional characterization of epigenetic chromatin regulators. Nucleic Acids Res, 2018, 46(19):10019–33., DOI 10.1093/nar/gky679 Laurent C, Valet F, Planque N, Silveri L, Maacha S, Anezo O, et al. High PTP4A3 phosphatase expression correlates with metastatic risk in uveal melanoma patients. Cancer Res, 2011, 71(3):666–74., DOI 10.1158/0008-5472.can-10-0605 van Essen TH, van Pelt SI, Bronkhorst IHG, Versluis M, Nemati F, Laurent C, et al. Upregulation of HLA expression in primary uveal melanoma by infiltrating leukocytes. PLOS ONE, 2016, 11(10):e0164292., DOI 10.1371/journal.pone.0164292 Dai W, Li Y, Mo S, Feng Y, Zhang L, Xu Y, et al. A robust gene signature for the prediction of early relapse in stage I-III colon cancer. Mol Oncol, 2018, 12(4): 463–75., DOI 10.1002/1878-0261.12175 Gaujoux R, Seoighe C. A flexible R package for nonnegative matrix factorization. BMC Bioinformatics, 2010, 11(1):367., DOI 10.1186/1471-2105-11-367 TangW, Hu J, Zhang H, Wu P, He H. Kappa coefficient: A popular measure of rater agreement. Shanghai Arch Psychiatry, 2015, 27(1):62–7., DOI 10.11919/j.issn. 1002-0829.215010 McHughML. Interrater reliability: The kappa statistic. Biochemia Med, 2012, 22(3):276–82., DOI 10.11613/bm.2012.031 Marseglia M, Amaro A, Solari N, Gangemi R, Croce E, Tanda ET, et al. How to make immunotherapy an effective therapeutic choice for uveal melanoma. Cancers, 2021, 13(9):2043., DOI 10.3390/cancers13092043 Zhu K, Liu X, DengW,Wang G, Fu B. Identification of a chromatin regulator signature and potential candidate drugs for bladder cancer. Hereditas, 2022, 159(1): 13., DOI 10.1186/s41065-021-00212-x Mardinian K, Adashek JJ, Botta GP, Kato S, Kurzrock R. SMARCA4: Implications of an altered chromatin-remodeling gene for cancer development and therapy. Mol Cancer Ther, 2021, 20(12):2341–51., DOI 10.1158/1535-7163.mct- 21-0433 Shu X-S, Li L, Tao Q. Chromatin regulators with tumor suppressor properties and their alterations in human cancers. Epigenomics, 2012, 4(5):537–49., DOI 10. 2217/epi.12.50 Gurard-Levin ZA, Wilson LOW, Pancaldi V, Postel-Vinay S, Sousa FG, Reyes C, et al. Chromatin regulators as a guide for cancer treatment choice. Mol Cancer Ther, 2016, 15(7):1768–77., DOI 10.1158/1535-7163.mct-15-1008 Debaugny RE, Skok JA. CTCF and CTCFL in cancer. Curr Opin Genet Develop, 2020, 61:44–52., DOI 10.1016/j.gde.2020.02.021 Jiang Y, Liu S, Chen X, Cao Y, Tao Y. Genome-wide distribution of DNA methylation and DNA demethylation and related chromatin regulators in cancer. Biochim Biophys Acta, Bba, - Rev Cancer, 2013, 1835(2):155–63., DOI 10.1016/j. bbcan.2012.12.003 Becht E, Giraldo NA, Lacroix L, Buttard B, Elarouci N, Petitprez F, et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol, 2016, 17(1):218., DOI 10.1186/ s13059-016-1070-5 Zhao H, Chen Y, Shen P, Gong L. Identification of immune cell infiltration landscape and their prognostic significance in uveal melanoma. Front Cel Dev Biol, 2021, 9:713569., DOI 10.3389/fcell.2021.713569 Hellstrom KE, Hellstrom I. From the Hellstrom paradox toward cancer cure. Prog Mol Biol translational Sci, 2019, 164:1–24., DOI 10.1016/bs.pmbts.2018.11.002 Ma QY, Huang DY, Zhang HJ, Chen J, Miller W, Chen XF. Function of follicular helper T cell is impaired and correlates with survival time in non-small cell lung cancer. Int immunopharmacology, 2016, 41:1–7., DOI 10.1016/j.intimp.2016. 10.014 Zappasodi R, Budhu S, Hellmann MD, Postow MA, Senbabaoglu Y, Manne S, et al. Non-conventional inhibitory CD4+Foxp3-PD-1hi T cells as a biomarker of immune checkpoint blockade activity. Cancer cell, 2018, 33(6):1017–32., DOI 10. 1016/j.ccell.2018.05.009 Niogret J, Berger H, Rebe C, Mary R, Ballot E, Truntzer C, et al. Follicular helper-T cells restore CD8+-dependent antitumor immunity and anti-PD-L1/PD- 1 efficacy. J Immunother Cancer, 2021, 9(6):e002157., DOI 10.1136/jitc-2020-002157 Bromwich EJ,McArdle PA, Canna K, McMillan DC, McNicol AM, BrownM, et al. The relationship between T-lymphocyte infiltration, stage, tumour grade and survival in patients undergoing curative surgery for renal cell cancer. Br J Cancer, 2003, 89(10):1906–8., DOI 10.1038/sj.bjc.6601400 Kinoshita T, Muramatsu R, Fujita T, Nagumo H, Sakurai T, Noji S, et al. Prognostic value of tumor-infiltrating lymphocytes differs depending on histological type and smoking habit in completely resected non-small-cell lung cancer. Ann Oncol : official J Eur Soc Med Oncol, 2016, 27(11):2117–23., DOI 10. 1093/annonc/mdw319 McArdle PA, Canna K, McMillan DC, McNicol AM, Campbell R, Underwood MA. The relationship between T-lymphocyte subset infiltration and survival in patients with prostate cancer. Br J Cancer, 2004, 91(3):541–3., DOI 10.1038/sj.bjc. 6601943 Droeser R, Zlobec I, Kilic E, Guth U, Heberer M, Spagnoli G, et al. Differential pattern and prognostic significance of CD4+, FOXP3+ and IL-17+ tumor infiltrating lymphocytes in ductal and lobular breast cancers. BMC cancer, 2012, 12:134., DOI 10.1186/1471-2407-12-134 Li M, Zhao J, Yang R, Cai R, Liu X, Xie J, et al. CENPF as an independent prognostic and metastasis biomarker corresponding to CD4+ memory T cells in cutaneous melanoma. Cancer Sci, 2022, 113(4):1220–34., DOI 10.1111/cas.15303 Mantovani A,Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol, 2017, 14(7): 399–416., DOI 10.1038/nrclinonc.2016.217 Cassetta L, Pollard JW. Targeting macrophages: Therapeutic approaches in cancer. Nat Rev Drug Discov, 2018, 17(12):887–904., DOI 10.1038/nrd.2018.169 Herwig MC, Bergstrom C, Wells JR, Holler T, Grossniklaus HE. M2/M1 ratio of tumor associated macrophages and PPAR-gamma expression in uveal melanomas with class 1 and class 2 molecular profiles. Exp Eye Res, 2013, 107: 52–8., DOI 10.1016/j.exer.2012.11.012 Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol, 2013, 14(10):1014–22., DOI 10.1038/ni. 2703 Joyce JA, Fearon DT. T cell exclusion, immune privilege, and the tumor microenvironment. Science, New York, N.Y.,, 2015, 348(6230):74–80., DOI 10.1126/ science.aaa6204 Spranger S, Gajewski TF. Tumor-intrinsic oncogene pathways mediating immune avoidance. Oncoimmunology, 2015, 5(3):e1086862., DOI 10.1080/ 2162402X.2015.1086862 Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med, 2018, 24(10): 1550–8., DOI 10.1038/s41591-018-0136-1 Torrens-Mas M, Oliver J, Roca P, Sastre-Serra J. SIRT3: Oncogene and tumor suppressor in cancer. Cancers, 2017, 9(7):90., DOI 10.3390/ cancers9070090 Kenny TC, Craig AJ, Villanueva A, Germain D. Mitohormesis primes tumor invasion and metastasis. Cel Rep, 2019, 27(8):2292–303., DOI 10.1016/j.celrep.2019. 04.095 Kenny TC, Hart P, Ragazzi M, Sersinghe M, Chipuk J, Sagar MAK, et al. Selected mitochondrial DNA landscapes activate the SIRT3 axis of the UPRmt to promote metastasis. Oncogene, 2017, 36(31):4393–404., DOI 10.1038/onc.2017.52 Lee JJ, van de Ven RAH, Zaganjor E, Ng MR, Barakat A, Demmers JJPG, et al. Inhibition of epithelial cell migration and Src/FAK signaling by SIRT3. Proc Natl Acad Sci United States America, 2018, 115(27):7057–62., DOI 10.1073/pnas. 1800440115 Wei Z, Song J,Wang G, Cui X, Zheng J, Tang Y, et al. Deacetylation of serine hydroxymethyl-transferase 2 by SIRT3 promotes colorectal carcinogenesis. Nat Commun, 2018, 9(1):4468., DOI 10.1038/s41467-018-06812-y Liang L, Li Q, Huang L, Li D, Li X. Sirt3 binds to and deacetylates mitochondrial pyruvate carrier 1 to enhance its activity. Biochem biophysical Res Commun, 2015, 468(4):807–12., DOI 10.1016/j.bbrc.2015.11.036 Fu W, Li H, Fu H, Zhao S, Shi W, SunM, et al. The SIRT3 and SIRT6 promote prostate cancer progression by inhibiting necroptosis-mediated innate immune response. J Immunol Res, 2020, 2020:8820355., DOI 10.1155/2020/8820355 Li R, Quan Y, Xia W. SIRT3 inhibits prostate cancer metastasis through regulation of FOXO3A by suppressing Wnt/β-catenin pathway. Exp Cel Res, 2018, 364(2):143–51., DOI 10.1016/j.yexcr.2018.01.036 Luo H, Ma C, Shao J, Cao J. Prognostic implications of novel ten-gene signature in uveal melanoma. Front Oncol, 2020, 10:567512., DOI 10.3389/fonc.2020. 567512 Ho L, Crabtree GR. Chromatin remodelling during development. Nature, 2010, 463(7280):474–84., DOI 10.1038/nature08911 Lickert H, Takeuchi JK, von Both I, Walls JR, McAuliffe F, Lee Adamson S, et al. Baf60c is essential for function of BAF chromatin remodelling complexes in heart development. Nature, 2004, 432(7013):107–12., DOI 10.1038/nature03071 Tropee R, de la Pena Avalos B, Gough M, Snell C, Duijf PHG, Dray E. The SWI/SNF subunit SMARCD3 regulates cell cycle progression and predicts survival outcome in ER+ breast cancer. Breast Cancer Res Treat, 2020, 185(3):601–14., DOI 10.1007/s10549-020-05997-5 Jordan NV, Prat A, Abell AN, Zawistowski JS, Sciaky N, Karginova OA, et al. SWI/SNF chromatin-remodeling factor smarcd3/baf60c controls epithelialmesenchymal transition by inducing Wnt5a signaling. Mol Cell Biol, 2013, 33(15):3011–25., DOI 10.1128/mcb.01443-12 Jiang M, Wang H, Chen H, Han Y. SMARCD3 is a potential prognostic marker and therapeutic target in CAFs. Aging, 2020, 12(20):20835–61., DOI 10. 18632/aging.104102 Takita J, Ishii M, Tsutsumi S, Tanaka Y, Kato K, Toyoda Y, et al. Gene expression profiling and identification of novel prognostic marker genes in neuroblastoma. Genes, Chromosomes and Cancer, 2004, 40(2):120–32., DOI 10. 1002/gcc.20021 Ajore R, Niroula A, Pertesi M, Cafaro C, Thodberg M, Went M, et al. Functional dissection of inherited non-coding variation influencing multiple myeloma risk. Nat Commun, 2022, 13(1):151., DOI 10.1038/s41467-021-27666-x Zhao D-D, Zhao X, Li WT. Identification of differentially expressed metastatic genes and their signatures to predict the overall survival of uveal melanoma patients by bioinformatics analysis. Int J Ophthalmol, 2020, 13(7):1046–53., DOI 10.18240/ ijo.2020.07.05 Matias PM, Gorynia S, Donner P, Carrondo MA. Crystal structure of the human AAA+ protein RuvBL1. J Biol Chem, 2006, 281(50):38918–29., DOI 10.1074/ jbc.m605625200 Guo H, Zhang X-Y, Peng J, Huang Y, Yang Y, Liu Y, et al. RUVBL1, a novel C-RAF-binding protein, activates the RAF/MEK/ERK pathway to promote lung cancer tumorigenesis. Biochem Biophysical Res Commun, 2018, 498(4):932–9., DOI 10.1016/j.bbrc.2018.03.084 Fan W, Xie J, Xia J, Zhang Y, Yang M, Wang H, et al. RUVBL1-ITFG1 interaction is required for collective invasion in breast cancer. Biochim Biophys Acta, Bba, - Gen Subjects, 2017, 1861(7):1788–800., DOI 10.1016/j.bbagen.2017.03.016 Mello T, Materozzi M, Zanieri F, Simeone I, Ceni E, Bereshchenko O, et al. Liver haploinsufficiency of RuvBL1 causes hepatic insulin resistance and enhances hepatocellular carcinoma progression. Int J Cancer, 2019, 146(12):3410–22., DOI 10. 1002/ijc.32787 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1610980 EP 1610996 DI 10.3389/pore.2023.1610980 PG 17 ER PT J AU Huang, H Li, Z Xia, Y Zhao, Z Wang, D Jin, H Liu, F Yang, Y Shen, L Lu, Z AF Huang, Huizhen Li, Zhiheng Xia, Yue Zhao, Zhenhua Wang, Dandan Jin, Hongyan Liu, Fang Yang, Ye Shen, Liyijing Lu, Zengxin TI Association between radiomics features of DCE-MRI and CD8+ and CD4+ TILs in advanced gastric cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE radiomics; CD4+; CD8+; advanced gastric cancer (AGC); dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) ID radiomics; CD4+; CD8+; advanced gastric cancer (AGC); dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) AB Objective: The aim of this investigation was to explore the correlation between the levels of tumor-infiltrating CD8+ and CD4+ T cells and the quantitative pharmacokinetic parameters of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) in patients with advanced gastric cancer. Methods: We retrospectively analyzed the data of 103 patients with histopathologically confirmed advanced gastric cancer (AGC). Three pharmacokinetic parameters, Kep, Ktrans, and Ve, and their radiomics characteristics were obtained by Omni Kinetics software. Immunohistochemical staining was used to determine CD4+ and CD8+ TILs. Statistical analysis was subsequently performed to assess the correlation between radiomics characteristics and CD4+ and CD8+ TIL density. Results: All patients included in this study were finally divided into either a CD8+ TILs low-density group (n = 51) (CD8+ TILs < 138) or a high-density group (n = 52) (CD8+ TILs ≥ 138), and a CD4+ TILs low-density group (n = 51) (CD4+ TILs < 87) or a high-density group (n = 52) (CD4+ TILs ≥ 87). ClusterShade and Skewness based on Kep and Skewness based on Ktrans both showed moderate negative correlation with CD8+ TIL levels (r = 0.630–0.349, p < 0.001), with ClusterShade based on Kep having the highest negative correlation (r = −0.630, p < 0.001). Inertia-based Kep showed a moderate positive correlation with the CD4+ TIL level (r = 0.549, p < 0.001), and the Correlation based on Kep showed a moderate negative correlation with the CD4+ TIL level, which also had the highest correlation coefficient (r = −0.616, p < 0.001). The diagnostic efficacy of the above features was assessed by ROC curves. For CD8+ TILs, ClusterShade of Kep had the highest mean area under the curve (AUC) (0.863). For CD4+ TILs, the Correlation of Kep had the highest mean AUC (0.856). Conclusion: The radiomics features of DCE-MRI are associated with the expression of tumor-infiltrating CD8+ and CD4+ T cells in AGC, which have the potential to noninvasively evaluate the expression of CD8+ and CD4+ TILs in AGC patients. C1 [Huang, Huizhen] Shaoxing University School of MedicineShaoxing, China. [Li, Zhiheng] Anhui Provincial Hospital, Department of RadiologyHefei, China. [Xia, Yue] Shaoxing University School of MedicineShaoxing, China. [Zhao, Zhenhua] Shaoxing People’s Hospital, Department of RadiologyShaoxing, China. [Wang, Dandan] Shaoxing People’s Hospital, Department of RadiologyShaoxing, China. [Jin, Hongyan] Shaoxing People’s Hospital, Country Department of PathologyShaoxing, China. [Liu, Fang] Shaoxing People’s Hospital, Country Department of PathologyShaoxing, China. [Yang, Ye] Shaoxing People’s Hospital, Country Department of PathologyShaoxing, China. [Shen, Liyijing] Shaoxing People’s Hospital, Department of RadiologyShaoxing, China. [Lu, Zengxin] Shaoxing People’s Hospital, Department of RadiologyShaoxing, China. RP Lu, Z (reprint author), Shaoxing People’s Hospital, Department of Radiology, Shaoxing, China. EM luzx777@163.com CR Xie J, Fu L, Jin L. Immunotherapy of gastric cancer: Past, future perspective and challenges. Pathol Res Pract, 2021, 218:218153322., DOI 10.1016/j.prp.2020. 153322 Emaan H, Abdullah E, Ibrahim M, Haneen S, Maen A. Recent trends and advancements in the diagnosis and management of gastric cancer. Cancers, 2022, 14(22):5615., DOI 10.3390/cancers14225615 Meng X, Wang L, Zhu B, Sun T, Guo S, Wang Y, et al. Totally laparoscopic gastrectomy versus laparoscopic-assisted gastrectomy for gastric cancer: A systematic review and meta-analysis. J Laparoendosc Adv Surg Tech A, 2021, 31(6):676–691., DOI 10.1089/lap.2020.0566 Cui Q, Mao Y,Wu D, Hu Y,Ma D, Zhang L, et al. Apatinib combined with PD- 1 antibody for third-line or later treatment of advanced gastric cancer. Front Oncol, 2022, 12:12952494., DOI 10.3389/fonc.2022.952494 El Helali A, Tao J, Wong CHL, Chan WW, Mok KC, Wu WF, et al. A metaanalysis with systematic review: Efficacy and safety of immune checkpoint inhibitors in patients with advanced gastric cancer. Front Oncol, 2022, 12: 908026., DOI 10.3389/fonc.2022.908026 Granhoj JS, Witness Praest Jensen A, Presti M, Met O, Svane IM, Donia M. Tumor-infiltrating lymphocytes for adoptive cell therapy: Recent advances, challenges, and future directions. Expert Opin Biol Ther, 2022, 22(5):627–641., DOI 10.1080/14712598.2022.2064711 Zhang D, He W, Wu C, Tan Y, He Y, Xu B, et al. Scoring system for tumorinfiltrating lymphocytes and its prognostic value for gastric cancer. Front Immunol, 2019, 10:1071., DOI 10.3389/fimmu.2019.00071 Escors D, Bocanegra A, Chocarro L, Blanco E, Pineiro-Hermida S, Garnica M, et al. Systemic CD4 immunity and PD-L1/PD-1 blockade immunotherapy. Int J Mol Sci, 2022, 23(21):13241., DOI 10.3390/ijms232113241 Hung K, Hayashi R, Lafond-Walker A, Lowenstein C, Pardoll D, Levitsky H. The central role of CD4(+, T cells in the antitumor immune response. J Exp Med, 1998, 188(12):2357–68., DOI 10.1084/jem.188.12.2357 Zhang N, Wang D, Hu X, Zhang G, Li Z, Zhao Y, et al. Analysis of immune status in gastric adenocarcinoma with different infiltrating patterns and origin sites. Front Immunol, 2022, 13:13978715., DOI 10.3389/fimmu.2022.978715 Zhao L, Liu Y, Zhang S, Wei L, Cheng H, Wang J, et al. Impacts and mechanisms of metabolic reprogramming of tumor microenvironment for immunotherapy in gastric cancer. Cell Death Dis, 2022, 13(4):378., DOI 10.1038/ s41419-022-04821-w He J, Xiong X, Yang H, Li D, Liu X, Li S, et al. Defined tumor antigen-specific T cells potentiate personalized TCR-T cell therapy and prediction of immunotherapy response. Cell Res, 2022, 32(6):530–542., DOI 10.1038/s41422- 022-00627-9 Liao H, Zhang Z, Chen J, Liao M, Xu L, Wu Z, et al. Preoperative radiomic approach to evaluate tumor-infiltrating CD8+ T cells in hepatocellular carcinoma patients using contrast-enhanced computed tomography. Ann Surg Oncol, 2019, 26(13):4537–47., DOI 10.1245/s10434-019-07815-9 El Naqa I, Ten Haken RK. Can radiomics personalise immunotherapy? Lancet Oncol, 2018, 19(9):1138–9., DOI 10.1016/S1470-2045(18)30429-7 Li Z, Zhao Z, Wang C, Wang D, Mao H, Liu F, et al. Association between DCE-MRI perfusion histogram parameters and EGFR and VEGF expressions in different lauren classifications of advanced gastric cancer. Pathol Oncol Res, 2021, 27:271610001., DOI 10.3389/pore.2021.1610001 Braman N, Prasanna P, Whitney J, Singh S, Beig N, Etesami M, et al. Association of peritumoral radiomics with tumor biology and pathologic response to preoperative targeted therapy for HER2, ERBB2)-Positive breast cancer. JAMA Netw Open, 2019, 2(4):e192561., DOI 10.1001/jamanetworkopen.2019.2561 Jiang Y, Wang H, Wu J, Chen C, Yuan Q, Huang W, et al. Noninvasive imaging evaluation of tumor immune microenvironment to predict outcomes in gastric cancer. Ann Oncol, 2020, 31(6):760–768., DOI 10.1016/j.annonc.2020.03.295 Lee HE, Chae SW, Lee YJ, Kim MA, Lee HS, Lee BL, et al. Prognostic implications of type and density of tumour-infiltrating lymphocytes in gastric cancer. Br J Cancer, 2008, 99(10):1704–11., DOI 10.1038/sj.bjc.6604738 Sarhadi VK, Armengol G. Molecular biomarkers in cancer. Biomolecules, 2022, 12(8):1021., DOI 10.3390/biom12081021 Mayerhoefer ME, Materka A, Langs G, Haggstrom I, Szczypinski P, Gibbs P, et al. Introduction to radiomics. J Nucl Med, 2020, 61(4):488–95., DOI 10.2967/ jnumed.118.222893 Chetan MR, Gleeson FV. Radiomics in predicting treatment response in nonsmall- cell lung cancer: Current status, challenges and future perspectives. Eur Radiol, 2021, 31(2):1049–1058., DOI 10.1007/s00330-020-07141-9 Fiz F, Vigano L, Gennaro N, Costa G, La Bella L, Boichuk A, et al. Radiomics of liver metastases: A systematic review. Cancers, Basel,, 2020, 12(10):2881., DOI 10. 3390/cancers12102881 Yu Y, He Z, Ouyang J, Tan Y, Chen Y, Gu Y, et al. Magnetic resonance imaging radiomics predicts preoperative axillary lymph node metastasis to support surgical decisions and is associated with tumor microenvironment in invasive breast cancer: A machine learning, multicenter study. EBioMedicine, 2021, 69:69103460., DOI 10.1016/j.ebiom.2021.103460 Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet, 2020, 396(10251):635–48., DOI 10.1016/S0140-6736(20)31288-5 Ruffin AT, Li H, Vujanovic L, Zandberg DP, Ferris RL, Bruno TC. Improving head and neck cancer therapies by immunomodulation of the tumourmicroenvironment. Nat Rev Cancer, 2022, 23:173–188., DOI 10.1038/s41568-022-00531-9 Park S, Cha H, Kim HS, Lee B, Kim S, Kim TM, et al. Transcriptional upregulation of CXCL13 is correlated with a favorable response to immune checkpoint inhibitors in lung adenocarcinoma. Cancer Med, 2022, 12: 7639–7650., DOI 10.1002/cam4.5460 Cho H, Kim JE, Hong YS, Kim SY, Kim J, Ryu YM, et al. Comprehensive evaluation of the tumor immune microenvironment and its dynamic changes in patients with locally advanced rectal cancer treated with preoperative chemoradiotherapy: From the phase II ADORE study. Oncoimmunology, 2022, 11(1):2148374., DOI 10.1080/2162402X.2022.2148374 Liang Y, Tan Y, Guan B, Guo B, Xia M, Li J, et al. Single-cell atlases link macrophages and CD8+ T-cell subpopulations to disease progression and immunotherapy response in urothelial carcinoma. Theranostics, 2022, 12(18): 7745–7759., DOI 10.7150/thno.77281 Mehrdad R, Elio A, Biagio R, Lynette MS, Weiwei S, Joao VA, et al. Association of machine learning-based assessment of tumor-infiltrating lymphocytes on standard histologic images with outcomes of immunotherapy in patients with NSCLC. JAMA Oncol, 2022, 9(1):51–60., DOI 10.1001/jamaoncol.2022. 4933 Smith JD, Bellile EL, Ellsperman SE, Heft-Neal ME, Mann JE, Birkeland AC, et al. Prognostic value of CD103+ tumor-infiltrating lymphocytes and programmed death ligand-1, PD-L1, combined positive score in recurrent laryngeal squamous cell carcinoma. Oral Oncol, 2022, 135:135106226., DOI 10.1016/j.oraloncology.2022. 106226 Zhang C, Sun Y, Li S, Shen L, Teng X, Xiao Y, et al. Autophagic flux restoration enhances the antitumor efficacy of tumor infiltrating lymphocytes. J Immunother Cancer, 2022, 10(10):e004868., DOI 10.1136/jitc-2022-004868 Wang XR, Zhao WP, He LH, Wang SL, Li WD, Wang J, et al. dNLR and TILS can be used as indicators for prognosis and efficacy evaluation of neo-adjuvant chemotherapy in breast cancer. Am J translational Res, 2021, 13(11):13093–13098. Available from: https://pubmed.ncbi.nlm.nih.gov/34956528/(Accessed December 4, 2022). Anais JR, Daniel NA, Carmen DA, Luis AV. Synthetic TILs: Engineered tumor-infiltrating lymphocytes with improved therapeutic potential. Front Oncol, 2021, 10:593848., DOI 10.3389/fonc.2020.593848 Okcu O, Ozturk SD, Ozturk C, Sen B, Yasin AI, Bedir R. Tumor-infiltrating lymphocytes, TILs)/volume and prognosis: The value of TILs for survival in HER2 and TN breast cancer patients treated with chemotherapy. Ann Diagn Pathol, 2022, 58:151930., DOI 10.1016/j.anndiagpath.2022.151930 Li F, Sun Y, Huang J, Xu W, Liu J, Yuan Z. CD4/CD8 + T cells, DC subsets, Foxp3, and Ido expression are predictive indictors of gastric cancer prognosis. Cancer Med, 2019, 8(17):7330–44., DOI 10.1002/cam4.2596 Ahn H, Song GJ, Jang S-H, Lee HJ, Lee M-S, Lee J-H, et al. Relationship of FDG PET/CT textural features with the tumor microenvironment and recurrence risks in patients with advanced gastric cancers. Cancers, Basel,, 2022, 14(16):3936., DOI 10.3390/cancers14163936 Li J, Shi Z, Liu F, Fang X, Cao K, Meng Y, et al. XGBoost classifier based on computed tomography radiomics for prediction of tumor-infiltrating CD8+ T-cells in patients with pancreatic ductal adenocarcinoma. Front Oncol, 2021, 11: 11671333., DOI 10.3389/fonc.2021.671333 Albano D, Bruno F, Agostini A, Angileri SA, Benenati M, Bicchierai G, et al. Dynamic contrast-enhanced, DCE, imaging: State of the art and applications in whole-body imaging. Jpn J Radiol, 2022, 40(4):341–366., DOI 10.1007/s11604-021- 01223-4 Li Z, Huang H, Wang C, Zhao Z, Ma W,Wang D, et al. DCE-MRI radiomics models predicting the expression of radioresistant-related factors of LRP-1 and survivin in locally advanced rectal cancer. Front Oncol, 2022, 12:12881341., DOI 10. 3389/fonc.2022.881341 Lambin P, Leijenaar RTH, Deist TM, Peerlings J, de Jong EEC, van Timmeren J, et al. Radiomics: The bridge between medical imaging and personalized medicine. Nat Rev Clin Oncol, 2017, 14(12):749–62., DOI 10.1038/nrclinonc.2017.141 Tang W, Kong Q, Cheng Z, Liang Y, Jin Z, Chen L, et al. Performance of radiomics models for tumour-infiltrating lymphocyte, TIL, prediction in breast cancer: The role of the dynamic contrast-enhanced, DCE, MRI phase. Eur Radiol, 2022, 32(2):864–875., DOI 10.1007/s00330-021-08173-5 Wu J, Li X, Teng X, Rubin DL, Napel S, Daniel BL, et al. Magnetic resonance imaging and molecular features associated with tumor-infiltrating lymphocytes in breast cancer. Breast Cancer Res, 2018, 20(1):101., DOI 10.1186/s13058-018-1039-2 Li X, Hu Y, Xie Y, Lu R, Li Q, Tao H, et al. Whole-tumor histogram analysis of diffusion-weighted imaging and dynamic contrast-enhanced MRI for soft tissue sarcoma: Correlation with HIF-1alpha expression. Eur Radiol, 2022, 33:3961–3973., DOI 10.1007/s00330-022-09296-z Kang SR, Kim HW, Kim HS. Evaluating the relationship between dynamic contrast-enhanced MRI, DCE-MRI, parameters and pathological characteristics in breast cancer. J Magn Reson Imaging, 2020, 52(5):1360–73., DOI 10.1002/jmri.27241 Dolina JS, Van Braeckel-Budimir N, Thomas GD, Salek-Ardakani S. CD8+ T cell exhaustion in cancer. Front Immunol, 2021, 12:715234., DOI 10.3389/fimmu. 2021.715234 Reading JL, Galvez-Cancino F, Swanton C, Lladser A, Peggs KS, Quezada SA. The function and dysfunction of memory CD8+ T cells in tumor immunity. Immunol Rev, 2018, 283(1):194–212., DOI 10.1111/imr.12657 van der Leun AM, Thommen DS, Schumacher TN. CD8+ T cell states in human cancer: Insights from single-cell analysis. Nat Rev Cancer, 2020, 20(4): 218–32., DOI 10.1038/s41568-019-0235-4 Wang S, Shi Q, Zhao Y, Song Y, Qiao G, Liu G, et al. Expansion of CD3+CD8+PD1+ T lymphocytes and TCR repertoire diversity predict clinical responses to adoptive cell therapy in advanced gastric cancer. Am J Cancer Res, 2022, 12(5):2203–2215. Ahn H, Song GJ, Jang S-H, Son MW, Lee HJ, Lee M-S, et al. Predicting the recurrence of gastric cancer using the textural features of perigastric adipose tissue on [18F] FDG PET/CT. Int J Mol Sci, 2022, 23(19):11985., DOI 10.3390/ ijms231911985 Chen D, Fu M, Chi L, Lin L, Cheng J, Xue W, et al. Prognostic and predictive value of a pathomics signature in gastric cancer. Nat Commun, 2022, 13(1):6903., DOI 10.1038/s41467-022-34703-w Thomas SN, Zhu F, Schnaar RL, Alves CS, Konstantopoulos K. Carcinoembryonic antigen and CD44 variant isoforms cooperate to mediate colon carcinoma cell adhesion to E- and L-selectin in shear flow. J Biol Chem, 2008, 283(23):15647–55., DOI 10.1074/jbc.M800543200 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611001 EP 1611011 DI 10.3389/pore.2023.1611001 PG 11 ER PT J AU Zhao, B Wang, J Ma, Z Ye, H Yang, T Meng, K AF Zhao, Bochao Wang, Jingchao Ma, Zhicheng Ye, Haikun Yang, Tao Meng, Kewei TI Development and validation of a prognostic nomogram for rectal cancer patients who underwent surgical resection SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE survival; prognostic; nomogram; SEER; rectal cancer ID survival; prognostic; nomogram; SEER; rectal cancer AB Objective: The purpose of this study was to develop and validate a nomogram model for the prediction of survival outcome in rectal cancer patients who underwent surgical resection. Methods: A total of 9,919 consecutive patients were retrospectively identified using the Surveillance, Epidemiology, and End Results (SEER) database. Significant prognostic factors were determined by the univariate and multivariate Cox analysis. The nomogram model for the prediction of cancer-specific survival (CSS) in rectal cancer patients were developed based on these prognostic variables, and its predictive power was assessed by the concordance index (C-index). Calibration curves were plotted to evaluate the associations between predicted probabilities and actual observations. The internal and external cohort were used to further validate the predictive performance of the prognostic nomogram. Results: All patients from the SEER database were randomly split into a training cohort (n = 6,944) and an internal validation cohort (n = 2,975). The baseline characteristics of two cohorts was comparable. Independent prognostic factors were identified as age, pT stage, lymph node metastasis, serum CEA level, tumor size, differentiation type, perineural invasion, circumferential resection margin involvement and inadequate lymph node yield. In the training cohort, the C-index of the nomogram was 0.719 (95% CI: 0.696–0.742), which was significantly higher than that of the TNM staging system (C-index: 0.606, 95% CI: 0.583–0.629). The nomogram had a C-index of 0.726 (95% CI: 0.691–0.761) for the internal validation cohort, indicating a good predictive power. In addition, an independent cohort composed of 202 rectal cancer patients from our institution were enrolled as the external validation. Compared with the TNM staging system (C-index: 0.573, 95% CI: 0.492–0.654), the prognostic nomogram still showed a better predictive performance, with the C-index of 0.704 (95% CI: 0.626–0.782). Calibration plots showed a good consistency between predicted probability and the actual observation in the training and two validation cohorts. Conclusion: The nomogram showed an excellent predictive ability for survival outcome of rectal cancer patients, and it might provide an accurate prognostic stratification and help clinicians determine individualized treatment strategies. C1 [Zhao, Bochao] Tianjin First Central Hospital, Department of Gastrointestinal SurgeryTianjin, China. [Wang, Jingchao] Tianjin First Central Hospital, Department of Gastrointestinal SurgeryTianjin, China. [Ma, Zhicheng] Tianjin First Central Hospital, Department of Gastrointestinal SurgeryTianjin, China. [Ye, Haikun] Tianjin First Central Hospital, Department of Gastrointestinal SurgeryTianjin, China. [Yang, Tao] Tianjin First Central Hospital, Department of Gastrointestinal SurgeryTianjin, China. [Meng, Kewei] Tianjin First Central Hospital, Department of Gastrointestinal SurgeryTianjin, China. RP Meng, K (reprint author), Tianjin First Central Hospital, Department of Gastrointestinal Surgery, Tianjin, China. EM doctormkw1969@126.com CR Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, et al. Colorectal cancer statistics, 2020. CA Cancer J Clin, 2020, 70:145–64., DOI 10. 3322/caac.21601 Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics. CA Cancer J Clin, 2022, 72:7–33., DOI 10.3322/caac.21708 Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen YJ, Ciombor KK, et al. Rectal cancer, version 2.2018, NCCN clinical practice guidelines in Oncology. J Natl Compr Canc Netw, 2018, 16:874–901., DOI 10.6004/jnccn.2018.0061 Schmoll HJ, Van Cutsem E, Stein A, Valentini V, Glimelius B, Haustermans K, et al. ESMO Consensus Guidelines for management of patients with colon and rectal cancer. a personalized approach to clinical decision making. Ann Oncol, 2012, 23:2479–516., DOI 10.1093/annonc/mds236 van Gijn W, Marijnen CA, Nagtegaal ID, Kranenbarg EM, PutterH,Wiggers T, et al. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer: 12-year follow-up of the multicentre, randomised controlled TME trial. Lancet Oncol, 2011, 12:575–82., DOI 10.1016/S1470- 2045(11)70097-3 Engelen SM, Maas M, Lahaye MJ, Leijtens JW, van Berlo CL, Jansen RL, et al. Modern multidisciplinary treatment of rectal cancer based on staging with magnetic resonance imaging leads to excellent local control, but distant control remains a challenge. Eur J Cancer, 2013, 49:2311–20., DOI 10.1016/j.ejca.2013.03.006 Nikberg M, Kindler C, Chabok A, Letocha H, Shetye J, Smedh K. Circumferential resection margin as a prognostic marker in the modern multidisciplinary management of rectal cancer. Dis Colon Rectum, 2015, 58: 275–82., DOI 10.1097/DCR.0000000000000250 Bosset JF, Calais G, Mineur L, Maingon P, Stojanovic-Rundic S, Bensadoun RJ, et al. Fluorouracil-based adjuvant chemotherapy after preoperative chemoradiotherapy in rectal cancer: Long-term results of the EORTC 22921 randomised study. Lancet Oncol, 2014, 15:184–90., DOI 10.1016/S1470- 2045(13)70599-0 Breugom AJ, Swets M, Bosset JF, Collette L, Sainato A, Cionini L, et al. Adjuvant chemotherapy after preoperative, chemo)radiotherapy and surgery for patients with rectal cancer: A systematic review and meta-analysis of individual patient data. Lancet Oncol, 2015, 16:200–7., DOI 10.1016/S1470-2045(14)71199-4 Valentini V, van Stiphout RG, Lammering G, Gambacorta MA, Barba MC, Bebenek M, et al. Nomograms for predicting local recurrence, distant metastases, and overall survival for patients with locally advanced rectal cancer on the basis of European randomized clinical trials. J Clin Oncol, 2011, 29:3163–72., DOI 10.1200/ JCO.2010.33.1595 Li ZH, Xie PY, Zhang DF, Li YJ,Wu L, Dong J, et al. Nomogramfor predicting disease-free survival among a multicenter cohort of Chinese patients with locally advanced rectal cancer. Cancer Manag Res, 2019, 11:2471–83., DOI 10.2147/CMAR. S196614 Fan S, Li T, Zhou P, Peng Q, Zhu Y. Development and validation of nomogram combining serum biomarker for predicting survival in patients with resected rectal cancer. Biosci Rep, 2019, 39:BSR20192636., DOI 10.1042/ BSR20192636 Iasonos A, Schrag D, Raj GV, Panageas KS. How to build and interpret a nomogram for cancer prognosis. J Clin Oncol, 2008, 26:1364–70., DOI 10.1200/JCO. 2007.12.9791 Kim KH, Yang SS, Yoon YS, Lim SB, Yu CS, Kim JC. Validation of the seventh edition of the American Joint committee on cancer tumor-node-metastasis, AJCC TNM, staging in patients with stage II and stage III colorectal carcinoma: Analysis of 2511 cases from a medical centre in korea. Colorectal Dis, 2011, 13:e220–6., DOI 10.1111/j.1463-1318.2011.02625.x Tarantino I, Warschkow R, Worni M, Merati-Kashani K, Koberle D, Schmied BM, et al. Elevated preoperative CEA is associated with worse survival in stage I-III rectal cancer patients. Br J Cancer, 2012, 107:266–74., DOI 10. 1038/bjc.2012.267 Tayyab M, Razack A, Sharma A, Gunn J, Hartley JE. Correlation of rectal tumor volumes with oncological outcomes for low rectal cancers: Does tumor size matter. Surg Today, 2015, 45:826–33., DOI 10.1007/s00595-014- 1068-0 Hyngstrom JR, Hu CY, Xing Y, You YN, Feig BW, Skibber JM, et al. Clinicopathology and outcomes for mucinous and signet ring colorectal adenocarcinoma: Analysis from the national cancer data base. Ann Surg Oncol, 2012, 19:2814–21., DOI 10.1245/s10434-012-2321-7 Song JH, Yu M, Kang KM, Lee JH, Kim SH, Nam TK, et al. Significance of perineural and lymphovascular invasion in locally advanced rectal cancer treated by preoperative chemoradiotherapy and radical surgery: Can perineural invasion be an indication of adjuvant chemotherapy. Radiother Oncol, 2019, 133:125–31., DOI 10. 1016/j.radonc.2019.01.002 Bernstein TE, Endreseth BH, Romundstad P, Wibe ANorwegian Colorectal Cancer Group. Circumferential resection margin as a prognostic factor in rectal cancer. Br J Surg, 2009, 96:1348–57., DOI 10.1002/bjs.6739 Lykke J, Jess P, Roikjaer ODanish Colorectal Cancer Group. Increased lymph node yield is associated with improved survival in rectal cancer irrespective of neoadjuvant treatment: Results from a national cohort study. Dis Colon Rectum, 2015, 58:823–30., DOI 10.1097/DCR.0000000000000429 Shen L, van Soest J, Wang J, Yu J, Hu W, Gong YU, et al. Validation of a rectal cancer outcome prediction model with a cohort of Chinese patients. Oncotarget, 2015, 6:38327–35., DOI 10.18632/oncotarget.5195 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611014 EP 1611023 DI 10.3389/pore.2023.1611014 PG 10 ER PT J AU Seftor, R Seftor, E Hendrix, M AF Seftor, E. B. Richard Seftor, A. Elisabeth Hendrix, J. C. Mary TI CVM-1118 (foslinanib), a 2-phenyl-4-quinolone derivative, promotes apoptosis and inhibits vasculogenic mimicry via targeting TRAP1 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE TRAP1; vasculogenic mimicry; CVM-1118; foslinanib; Hsp75; STK11; NF2 ID TRAP1; vasculogenic mimicry; CVM-1118; foslinanib; Hsp75; STK11; NF2 AB CVM-1118 (foslinanib) is a phosphoric ester compound selected from 2- phenyl-4-quinolone derivatives. The NCI 60 cancer panel screening showed CVM-1125, the major active metabolite of CVM-1118, to exhibit growth inhibitory and cytotoxic effects at nanomolar range. CVM-1118 possesses multiple bioactivities, including inducing cellular apoptosis, cell cycle arrest at G2/M, as well as inhibiting vasculogenic mimicry (VM) formation. The TNF receptor associated protein 1 (TRAP1) was identified as the binding target of CVM-1125 using nematic protein organization technique (NPOT) interactome analysis. Further studies demonstrated CVM-1125 reduced the protein level of TRAP1 and impeded its downstream signaling by reduction of cellular succinate levels and destabilization of HIF-1α. The pharmacogenomic biomarkers associated with CVM-1118 were also examined by Whole Genome CRISPR Knock-Out Screening. Two hits (STK11 and NF2) were confirmed with higher sensitivity to the drug in cell knock-down experiments. Biological assays indicate that the mechanism of action of CVM-1118 is via targeting TRAP1 to induce mitochondrial apoptosis, suppress tumor cell growth, and inhibit vasculogenic mimicry formation. Most importantly, the loss-of-function mutations of STK11 and NF2 are potential biomarkers of CVM-1118 which can be applied in the selection of cancer patients for CVM-1118 treatment. CVM-1118 is currently in its Phase 2a clinical development. C1 [Seftor, E. B. Richard] Shepherd University, Department of BiologyShepherdstown, WV, USA. [Seftor, A. Elisabeth] Shepherd University, Department of BiologyShepherdstown, WV, USA. [Hendrix, J. C. Mary] Shepherd University, Department of BiologyShepherdstown, WV, USA. CR Chou L-C, Yang J-S, Huang L-J, Wu H-C, Lu C-C, Chiang J-H, et al. The synthesized 2-(2-fluorophenyl)-6,7-methylenedioxyquinolin-4-one, CHM-1, promoted G2/M arrest through inhibition of CDK1 and induced apoptosis through the mitochondrial-dependent pathway in CT-26 murine colorectal adenocarcinoma cells. J Gastroenterol, 2009, 44:1055–63., DOI 10.1007/s00535- 009-0111-1 Chou LC, Chen CT, Lee JC, Way TD, Huang CH, Huang SM, et al. Synthesis and preclinical evaluations of 2-(2-fluorophenyl)-6,7- methylenedioxyquinolln-4- one monosodium phosphate, CHM-I-P-Na, as a potent antitumor agent. J Med Chem, 2010, 53:1616–26., DOI 10.1021/jm901292j Chou LC, Tsai MT, Hsu MH, Wang SH, Way TD, Huang CH, et al. Design, synthesis, and preclinical evaluation of new 5,6-, or 6,7-, disubstituted-2-, fluorophenyl)quinolin-4-one derivatives as potent antitumor agents. J Med Chem, 2010, 53:8047–58., DOI 10.1021/jm100780c Hour MJ, Huang LJ, Kuo SC, Xia Y, Bastow K, Nakanishi Y, et al. 6- Alkylamino- and 2,3-dihydro-3’-methoxy-2-phenyl-4-quinazolinones and related compounds: Their synthesis, cytotoxicity, and inhibition of tubulin polymerization. J Med Chem, 2000, 43:4479–87., DOI 10.1021/jm000151c Kuo S-C, Lee H-Z, Juang J, Lin Y-T, Wu T-S, Chang J-J, et al. Synthesis and cytotoxicity of 1,6,7,8-substit uted 2-(4’-S ubstit uted phenyl)-4-quinolones and related compounds: Identification as antimitotic agents interacting with tubulin. J Med Chem, 1993, 39:1146–56., DOI 10.1021/jm00061a005 Li L, Wang HK, Kuo SC, Wu TS, Mauger A, Lin CM, et al. Antitumor agents 155. Synthesis and biological evaluation of 3’,6,7-substituted 2-phenyl-4-quinolones as antimicrotubule agents. J Med Chem, 1994, 37:3400–7., DOI 10.1021/jm00046a025 Li L, Wang HK, Kuo SC, Wu TS, Lednicer D, Lin CM, et al. Antitumor agents 150. 2’,3’,4’,5’,5,6,7-substituted 2-phenyl-4-quinolones and related compounds: Their synthesis, cytotoxicity, and inhibition of tubulin polymerization. J Med Chem, 1994, 37:1126–35., DOI 10.1021/jm00034a010 Xia Y, Yang ZY, Xia P, Bastow KF, Tachibana Y, Kuo SC, et al. Antitumor agents 181. Synthesis and biological evaluation of 6,7,2’,3’,4’-substituted-1,2,3,4- tetrahydro-2-phenyl-4-quinolones as a new class of antimitotic antitumor agents. J Med Chem, 1998, 41:1155–62., DOI 10.1021/jm9707479 Xia Y, Yang ZY, Xia P, Hackl T, Hamel E, Mauger A, et al. Antitumor Agents 211. Fluorinated 2-phenyl-4-quinolone derivatives as antimitotic antitumor agents. J Med Chem, 2001, 44:3932–6., DOI 10.1021/jm0101085 Hendrix MJC, Seftor EA, Seftor REB, Chao JT, Chien DS, Chu YW. Tumor cell vascular mimicry: Novel targeting opportunity in melanoma. Pharmacol Ther, 2016, 159:83–92., DOI 10.1016/j.pharmthera.2016.01.006 Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe’er J, et al. Vascular channel formation by human melanoma cells in vivo and in vitro: Vasculogenic mimicry. Am J Pathol, 1999, 155:739–52., DOI 10.1016/S0002-9440(10)65173-5 Wagenblast E, Soto M, Gutierrez-Angel S, Hartl CA, Gable AL, Maceli AR, et al. A model of breast cancer heterogeneity reveals vascular mimicry as a driver of metastasis. Nature, 2015, 520:358–62., DOI 10.1038/nature14403 Luo Q, Wang J, Zhao W, Peng Z, Liu X, Li B, et al. Vasculogenic mimicry in carcinogenesis and clinical applications. J Hematol Oncol, 2020, 13:19., DOI 10.1186/ s13045-020-00858-6 Dey N, De P, Brian L-J. Evading anti-angiogenic therapy: Resistance to antiangiogenic therapy in solid tumors. Am J Transl Res, 2015, 7:1675–98. PintoMP, Sotomayor P, Carrasco-Avino G, Corvalan AH, Owen GI. Escaping antiangiogenic therapy: Strategies employed by cancer cells. Int J Mol Sci, 2016, 17., DOI 10.3390/ijms17091489 Altieri DC, Stein GS, Lian JB, Languino LR. TRAP-1, the mitochondrial Hsp90. Biochim Biophys Acta Mol Cel Res, 2012, 1823:767–73., DOI 10.1016/j. bbamcr.2011.08.007 Song HY, Dunbar JD, Zhang YX, Guo D, Donner DB. Identification of a protein with homology to hsp90 that binds the type 1 tumor necrosis factor receptor. J Biol Chem, 1995, 270:3574–81., DOI 10.1074/jbc.270.8.3574 Costantino E, Maddalena F, Calise S, Piscazzi A, Tirino V, Fersini A, et al. TRAP1, a novel mitochondrial chaperone responsible formulti-drug resistance and protection from apoptotis in human colorectal carcinoma cells. Cancer Lett, 2009, 279:39–46., DOI 10.1016/j.canlet.2009.01.018 Im CN. Past, present, and emerging roles of mitochondrial heat shock protein TRAP1 in the metabolism and regulation of cancer stem cells. Cell Stress Chaperones, 2016, 21:553–62., DOI 10.1007/s12192-016-0687-3 Gesualdi NM, Chirico G, Pirozzi G, Costantino E, Landriscina M, Esposito F. Tumor necrosis factor-associated protein 1, TRAP-1, protects cells from oxidative stress and apoptosis. Stress, 2007, 10:342–50., DOI 10.1080/10253890701314863 Sisinni L, Maddalena F, Lettini G, Condelli V, Matassa DS, Esposito F, et al. TRAP1 role in endoplasmic reticulum stress protection favors resistance to anthracyclins in breast carcinoma cells. Int J Oncol, 2014, 44:573–82., DOI 10.3892/ ijo.2013.2199 Yoshida S, Tsutsumi S, Muhlebach G, Sourbier C, Lee MJ, Lee S, et al. Molecular chaperone TRAP1 regulates a metabolic switch between mitochondrial respiration and aerobic glycolysis. Proc Natl Acad Sci U S A, 2013, 110:E1604–12., DOI 10.1073/pnas.1220659110 Zhang B, Wang J, Huang Z,Wei P, Liu Y, Hao J, et al. Aberrantly upregulated TRAP1 is required for tumorigenesis of breast cancer. Oncotarget, 2015, 6: 44495–508., DOI 10.18632/oncotarget.6252 Megger DA, Bracht T, Kohl M, Ahrens M, Naboulsi W, Weber F, et al. Proteomic differences between hepatocellular carcinoma and nontumorous liver tissue investigated by a combined gel-based and label-free quantitative proteomics study. Mol Cell Proteomics, 2013, 12:2006–20., DOI 10.1074/mcp.M113.028027 Li S, Lv Q, Sun H, Xue Y, Wang P, Liu L, et al. Expression of TRAP1 predicts poor survival of malignant glioma patients. J Mol Neurosci, 2015, 55:62–8., DOI 10. 1007/s12031-014-0413-5 Pak MG, Koh HJ, Roh MS. Clinicopathologic significance of TRAP1 expression in colorectal cancer: A large scale study of human colorectal adenocarcinoma tissues. Diagn Pathol, 2017, 12:6., DOI 10.1186/s13000-017-0598-3 Agorreta J,Hu J, Liu D, Delia D, TurleyH, Ferguson DJP, et al. TRAP1 regulates proliferation, mitochondrial function, and has prognostic significance in NSCLC. Mol Cancer Res, 2014, 12:660–9., DOI 10.1158/1541-7786.MCR-13-0481 Gee MB, Smith PE. Kirkwood-Buff theory of molecular and protein association, aggregation, and cellular crowding. J Chem Phys, 2009, 131:165101., DOI 10.1063/1.3253299 Shimizu S, Boon CL. The Kirkwood-Buff theory and the effect of cosolvents on biochemical reactions. J Chem Phys, 2004, 121:9147–55., DOI 10.1063/1.1806402 Beyrath J, PellegriniM, Renkema H,Houben L, Pecheritsyna S, van Zandvoort P, et al. KH176 safeguards mitochondrial diseased cells from redox stress-induced cell death by interacting with the thioredoxin system/peroxiredoxin enzyme machinery. Sci Rep, 2018, 8:6577., DOI 10.1038/s41598-018-24900-3 Walf-Vorderwulbecke V, Pearce K, Brooks T, Hubank M, van den Heuvel- Eibrink MM, Zwaan CM, et al. Targeting acute myeloid leukemia by drug-induced c-MYB degradation. Leukemia, 2018, 32:882–9., DOI 10.1038/leu.2017.317 Wang L, Eftekhari P, Schachner D, Ignatova ID, Palme V, Schilcher N, et al. Novel interactomics approach identifies ABCA1 as direct target of evodiamine, which increases macrophage cholesterol efflux. Sci Rep, 2018, 8:11061., DOI 10.1038/ s41598-018-29281-1 LiW, Xu H, Xiao T, Cong L, LoveMI, Zhang F, et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol, 2014, 15:554., DOI 10.1186/s13059-014-0554-4 Colic M, Wang G, Zimmermann M, Mascall K, McLaughlin M, Bertolet L, et al. Identifying chemogenetic interactions from CRISPR screens with drugZ. Genome Med, 2019, 11:52., DOI 10.1186/s13073-019-0665-3 Sciacovelli M, Guzzo G, Morello V, Frezza C, Zheng L, Nannini N, et al. The mitochondrial chaperone TRAP1 promotes neoplastic growth by inhibiting succinate dehydrogenase. Cell Metab, 2013, 17:988–99., DOI 10.1016/j.cmet.2013. 04.019 Zhao T, Mu X, You Q. Succinate: An initiator in tumorigenesis and progression. Oncotarget, 2017, 8:53819., DOI 10.18632/ONCOTARGET.17734 Wu JY, Huang TW, Hsieh YT, Wang YF, Yen CC, Lee GL, et al. Cancer-derived succinate promotes macrophage polarization and cancer metastasis via succinate receptor. Mol Cel, 2020, 77:213–27.e5., DOI 10.1016/J.MOLCEL.2019.10.023 Kang BH. TRAP1 regulation of mitochondrial life or death decision in cancer cells and mitochondria-targeted TRAP1 inhibitors. BMB Rep, 2012, 45:1–6., DOI 10. 5483/BMBRep.2012.45.1.1 Xie S,Wang X, Gan S, Tang X, Kang X, Zhu S. The mitochondrial chaperone TRAP1 as a candidate target of oncotherapy. Front Oncol, 2021, 10:585047., DOI 10. 3389/fonc.2020.585047 Hardy KM, Kirschmann DA, Seftor EA, Margaryan NV, Postovit LM, Strizzi L, et al. Regulation of the embryonic morphogen nodal by Notch4 facilitates manifestation of the aggressive melanoma phenotype. Cancer Res, 2010, 70: 10340–50., DOI 10.1158/0008-5472.CAN-10-0705 Schier AF. Nodal signaling in vertebrate development. Annu Rev Cel Dev Biol, 2003, 19:589–621., DOI 10.1146/annurev.cellbio.19.041603.094522 Kang BH, Plescia J, Dohi T, Rosa J, Doxsey SJ, Altieri DC. Regulation of tumor cell mitochondrial homeostasis by an organelle-specific Hsp90 chaperone network. Cell, 2007, 131:257–70., DOI 10.1016/j.cell.2007.08.028 Masgras I, Sanchez-Martin C, Colombo G, Rasola A. The chaperone TRAP1 as a modulator of the mitochondrial adaptations in cancer cells. Front Oncol, 2017, 7:58., DOI 10.3389/fonc.2017.00058 Park H-K, Yoon NG, Lee J-E, Hu S, Yoon S, Kim SY, et al. Unleashing the full potential of Hsp90 inhibitors as cancer therapeutics through simultaneous inactivation of Hsp90, Grp94, and TRAP1. Exp Mol Med, 2020, 52:79–91., DOI 10.1038/s12276-019-0360-x Hoter A, El-Sabban ME, Naim HY. The HSP90 family: Structure, regulation, function, and implications in health and disease. Int J Mol Sci, 2018, 19., DOI 10. 3390/ijms19092560 Mograbi B, Heeke S, Hofman P. The importance of stk11/lkb1 assessment in non-small cell lung carcinomas. Diagnostics, 2021, 11:196., DOI 10.3390/ diagnostics11020196 Petrilli AM, Fernandez-Valle C. Role of Merlin/NF2 inactivation in tumor biology. Oncogene, 2016, 35:537–48., DOI 10.1038/onc.2015.125 Shackelford DB, Shaw RJ. The LKB1-AMPK pathway: Metabolism and growth control in tumour suppression. Nat Rev Cancer, 2009, 9:563–75., DOI 10.1038/nrc2676 Faubert B, Vincent EE, Griss T, Samborska B, Izreig S, Svensson RU, et al. Loss of the tumor suppressor LKB1 promotes metabolic reprogramming of cancer cells via HIF-1α. Proc Natl Acad Sci U S A, 2014, 111:2554–9., DOI 10.1073/pnas. 1312570111 Shen H, Zhu M, Wang C. Precision oncology of lung cancer: Genetic and genomic differences in Chinese population. NPJ Precis Oncol, 2019, 3:14., DOI 10. 1038/s41698-019-0086-1 Wang H, Guo J, Shang X, Wang Z. Less immune cell infiltration and worse prognosis after immunotherapy for patients with lung adenocarcinoma who harbored STK11 mutation. Int Immunopharmacol, 2020, 84:106574., DOI 10. 1016/j.intimp.2020.106574 Ikediobi ON, Davies H, Bignell G, Edkins S, Stevens C, O’Meara S, et al. Mutation analysis of 24 known cancer genes in the NCI-60 cell line set. Mol Cancer Ther, 2006, 5:2606–12., DOI 10.1158/1535-7163.MCT-06-0433 Agnihotri S, Gugel I, Remke M, Bornemann A, Pantazis G, Mack SC, et al. Gene-expression profiling elucidates molecular signaling networks that can be therapeutically targeted in vestibular schwannoma. J Neurosurg, 2014, 121: 1434–45., DOI 10.3171/2014.6.JNS131433 Caino MC, Chae YC, Vaira V, Ferrero S, Nosotti M, Martin NM, et al. Metabolic stress regulates cytoskeletal dynamics and metastasis of cancer cells. J Clin Invest, 2013, 123:2907–20., DOI 10.1172/JCI67841 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611038 EP 1611054 DI 10.3389/pore.2023.1611038 PG 17 ER PT J AU He, X Zhou, S Li, H Gou, Y Jia, D AF He, Xiaohong Zhou, Sicheng Li, Hongjun Gou, Yue Jia, Dan TI Prognostic role of pretreatment skeletal muscle index in gastric cancer patients: A meta-analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE meta-analysis; prognosis; gastric cancer; pretreatment; skeletal muscle index ID meta-analysis; prognosis; gastric cancer; pretreatment; skeletal muscle index AB Background: The association between pretreatment skeletal muscle index (SMI) and long-term survival of gastric cancer patients remains unclear up to now. The aim of this meta-analysis was to identify the prognostic value of pretreatment SMI in gastric cancer. Methods: The PubMed, EMBASE and Web of Science electronic databases were searched up to 5 June 2022 for relevant studies. The primary outcome was overall survival (OS) and the second outcomes were disease-free survival (DFS) and cancer-specific survival (CSS). The hazard ratios (HRs) and 95% confidence intervals (CIs) were combined to assess the relationship between pretreatment SMI and survival of gastric cancer patients. All statistical analyses were conducted by STATA 15.0 software. Results: A total of 31 retrospective studies involving 12,434 patients were enrolled in this meta-analysis. The pooled results demonstrated that lower pretreatment was significantly associated with poorer OS (HR = 1.53, p < 0.001). Besides, lower pretreatment SMI was also related with worse DFS (HR = 1.39, p < 0.001) and CSS (HR = 1.96, p < 0.001). Conclusion: Pretreatment SMI was significantly associated with prognosis of gastric cancer patients and lower SMI predicted worse survival. However, more prospective high-quality studies are still needed to verify our findings. C1 [He, Xiaohong] Sichuan University, West China Hospital, Cancer CenterChengdu, China. [Zhou, Sicheng] Sichuan University / West China School of Nusing Sichuan University, West China Hospital, Outpatient DepartmentChengdu, China. [Li, Hongjun] Sichuan University / West China School of Nusing Sichuan University, West China Hospital, Outpatient DepartmentChengdu, China. [Gou, Yue] Sichuan University / West China School of Nusing Sichuan University, West China Hospital, Outpatient DepartmentChengdu, China. [Jia, Dan] Sichuan University / West China School of Nusing Sichuan University, West China Hospital, Outpatient DepartmentChengdu, China. RP Jia, D (reprint author), Sichuan University / West China School of Nusing Sichuan University, West China Hospital, Outpatient Department, Chengdu, China. EM 417522774@qq.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/ caac.21492 CaoW, Chen HD, Yu YW, Li N, Chen WQ. Changing profiles of cancer burden worldwide and in China: A secondary analysis of the global cancer statistics 2020. Chin Med J, 2021, 134(7):783–91., DOI 10.1097/CM9.0000000000001474 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Porro C, La Torre ME, Tartaglia N, Benameur T, Santini M, Ambrosi A, et al. The potential role of nutrition in lung cancer establishment and progression. Life, Basel, Switzerland,, 2022, 12(2):270., DOI 10.3390/life12020270 Supportive PDQ. Palliative care editorial B: Nutrition in cancer care, PDQ®): Health professional version. In: PDQ cancer information summaries. edn. Bethesda, MD): National Cancer Institute US, 2002). Takagi K, Domagala P, Polak WG, Buettner S, Wijnhoven BPL, Ijzermans JNM. Prognostic significance of the controlling nutritional status, CONUT, score in patients undergoing gastrectomy for gastric cancer: A systematic review and meta-analysis. BMC Surg, 2019, 19(1):129., DOI 10.1186/s12893-019-0593-6 Wang SH, Zhai ST, Lin H. Role of prognostic nutritional index in patients with gastric cancer: A meta-analysis. Minerva Med, 2016, 107(5):322–7. Yang Y, Gao P, Song Y, Sun J, Chen X, Zhao J, et al. The prognostic nutritional index is a predictive indicator of prognosis and postoperative complications in gastric cancer: A meta-analysis. Eur J Surg Oncol, 2016, 42(8):1176–82., DOI 10. 1016/j.ejso.2016.05.029 Lee J, Lin JB, Chen TC, Jan YT, Sun FJ, Chen YJ, et al. Progressive skeletal muscle loss after surgery and adjuvant radiotherapy impact survival outcomes in patients with early stage cervical cancer. Front Nutr, 2021, 8:773506., DOI 10.3389/ fnut.2021.773506 Nose Y, Yamashita K, Takeoka T, Momose K, Saito T, Tanaka K, et al. Perioperative ghrelin administration attenuates postoperative skeletal muscle loss in patients undergoing esophagectomy for esophageal cancer: Secondary analysis of a randomized controlled trial. Ann Surg Oncol, 2022, 29:3604–12., DOI 10.1245/ s10434-022-11436-0 Okamoto A, Aikou S, Iwata R, Oya S, Kawasaki K, Okumura Y, et al. The type of gastrectomy affects skeletal muscle loss and the long-term outcomes of elderly patients with gastric cancer: A retrospective study using computed tomography images. Surg Today, 2021, 52:812–21., DOI 10.1007/s00595-021-02414-2 Tang R, Deng JP, Zhang L, Zhang WW, Sun JY, Chi F, et al. Prognostic significance of the skeletal muscle index and systemic inflammatory index in patients with lymph node-positive breast cancer after radical mastectomy. BMC Cancer, 2022, 22(1):234., DOI 10.1186/s12885-022-09312-x Di Giorgio A, Rotolo S, Cintoni M, Rinninella E, Pulcini G, Schena CA, et al. The prognostic value of skeletal muscle index on clinical and survival outcomes after cytoreduction and HIPEC for peritoneal metastases from colorectal cancer: A systematic review and meta-analysis. Eur J Surg Oncol, 2022, 48(3):649–56., DOI 10. 1016/j.ejso.2021.10.008 Tranoulis A, Kwong FLA, Lakhiani A, Georgiou D, Yap J, Balega J. Prevalence of computed tomography-based sarcopenia and the prognostic value of skeletal muscle index and muscle attenuation amongst women with epithelial ovarian malignancy: A systematic review and meta-analysis. Eur J Surg Oncol, 2022, 48: 1441–54., DOI 10.1016/j.ejso.2022.02.024 Xu L, Jing Y, Zhao C, Zhang Q, Zhao X, Yang J, et al. Preoperative computed tomography-assessed skeletal muscle index is a novel prognostic factor in patients with hepatocellular carcinoma following hepatectomy: A meta-analysis. J Gastrointest Oncol, 2020, 11(5):1040–53., DOI 10.21037/jgo-20-122 McLeroy KR, Northridge ME, Balcazar H, Greenberg MR, Landers SJ. Reporting guidelines and the American journal of public health’s adoption of preferred reporting Items for systematic reviews and meta-analyses. Am J Public Health, 2012, 102(5):780–4., DOI 10.2105/AJPH.2011.300630 Stang A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol, 2010, 25(9):603–5., DOI 10.1007/s10654-010-9491-z Begg CB, MazumdarM. Operating characteristics of a rank correlation test for publication bias. Biometrics, 1994, 50(4):1088–101., DOI 10.2307/2533446 Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ, Clinical research ed,, 1997, 315(7109): 629–34., DOI 10.1136/bmj.315.7109.629 Wang Y, Huang D, Xu WY, Wang YW, Che GW. Prognostic value of pretreatment lymphocyte-to-monocyte ratio in non-small cell lung cancer: A metaanalysis. Oncol Res Treat, 2019, 42(10):523–31., DOI 10.1159/000501726 Hayashi N, Ando Y, Gyawali B, Shimokata T, Maeda O, Fukaya M, et al. Low skeletal muscle density is associated with poor survival in patients who receive chemotherapy for metastatic gastric cancer. Oncol Rep, 2016, 35(3):1727–31., DOI 10.3892/or.2015.4475 Zhuang CL, Huang DD, Pang WY, Zhou CJ, Wang SL, Lou N, et al. Sarcopenia is an independent predictor of severe postoperative complications and long-term survival after radical gastrectomy for gastric cancer: Analysis from a large-scale cohort. Medicine, Baltimore,, 2016, 95(13):e3164., DOI 10. 1097/MD.0000000000003164 Zheng ZF, Lu J, Zheng CH, Li P, Xie JW, Wang JB, et al. A novel prognostic scoring system based on preoperative sarcopenia predicts the long-term outcome for patients after R0 resection for gastric cancer: Experiences of a high-volume center. Ann Surg Oncol, 2017, 24(7):1795–803., DOI 10.1245/s10434-017-5813-7 Lee JS, Kim YS, Kim EY, Jin W. Prognostic significance of CT-determined sarcopenia in patients with advanced gastric cancer. Plos One, 2018, 13(8): e0202700., DOI 10.1371/journal.pone.0202700 O’Brien S, Twomey M, Moloney F, Kavanagh RG, Carey BW, Power D, et al. Sarcopenia and post-operative morbidity and mortality in patients with gastric cancer. J Gastric Cancer, 2018, 18(3):242–52., DOI 10.5230/jgc.2018.18.e25 Park HS, Kim HS, Beom SH, Rha SY, Chung HC, Kim JH, et al. Marked loss of muscle, visceral fat, or subcutaneous fat after gastrectomy predicts poor survival in advanced gastric cancer: Single-center study from the CLASSIC trial. Ann Surg Oncol, 2018, 25(11):3222–30., DOI 10.1245/s10434-018-6624-1 Sugiyama K, Narita Y, Mitani S, Honda K, Masuishi T, Taniguchi H, et al. Baseline sarcopenia and skeletal muscle loss during chemotherapy affect survival outcomes in metastatic gastric cancer. Anticancer Res, 2018, 38(10):5859–66., DOI 10.21873/anticanres.12928 Sierzega M, Chrzan R, Wiktorowicz M, Kolodziejczyk P, Richter P. Prognostic and predictive implications of sarcopenia in Western patients undergoing gastric resections for carcinoma of the stomach. J Surg Oncol, 2019, 120(3):473–82., DOI 10. 1002/jso.25509 Zhuang CL, Shen X, Zou HB, Dong QT, Cai HY, Chen XL, et al. EWGSOP2 versus EWGSOP1 for sarcopenia to predict prognosis in patients with gastric cancer after radical gastrectomy: Analysis from a large-scale prospective study. Clin Nutr, Edinburgh, Scotland,, 2019, 39(7):2301–10., DOI 10. 1016/j.clnu.2019.10.024 Dong QT, Cai HY, Zhang Z, Zou HB, Dong WX, Wang WB, et al. Influence of body composition, muscle strength, and physical performance on the postoperative complications and survival after radical gastrectomy for gastric cancer: A comprehensive analysis from a large-scale prospective study. Clin Nutr, Edinburgh, Scotland,, 2020, 40(5):3360–9., DOI 10.1016/j.clnu.2020.11.007 Kim EY, Jun KH, Kim SY, Chin HM. Body mass index and skeletal muscle index are useful prognostic factors for overall survival after gastrectomy for gastric cancer Retrospective cohort study. Medicine, 2020, 99(47):e23363., DOI 10.1097/ MD.0000000000023363 Wang JB, Xue Z, Lu J, He QL, Zheng ZF, Xu BB, et al. Effect of sarcopenia on short- and long-term outcomes in patients with gastric neuroendocrine neoplasms after radical gastrectomy: Results from a large, two-institution series. BMC Cancer, 2020, 20(1):1002., DOI 10.1186/s12885-020-07506-9 Yang W, Xia F, Wang J, Zhou M, Li G, Shen L, et al. Quantifying skeletal muscle wasting during chemoradiotherapy with Jacobian calculations for the prediction of survival and toxicity in patients with gastric cancer. Ejso, 2020, 46(7):1254–61., DOI 10.1016/j.ejso.2020.03.223 Yu JI, Choi C, Lee J, Kang WK, Park SH, Kim ST, et al. Effect of baseline sarcopenia on adjuvant treatment for D2 dissected gastric cancer: Analysis of the ARTIST phase III trial. Radiother Oncol : J Eur Soc Ther Radiol Oncol, 2020, 152: 19–25., DOI 10.1016/j.radonc.2020.07.043 Alnimri F, Sivakumar J, Sutherland T, Johnson MA, Ward S, Chong L, et al. Pre-operative low muscle mass is associated with major complications and lower recurrence-free survival after gastric cancer surgery. Anz J Surg, 2021, 91(3): 316–22., DOI 10.1111/ans.16590 An S, Eo W, Kim Y-J. Muscle-related parameters as determinants of survival in patients with stage I-III gastric cancer undergoing gastrectomy. J Cancer, 2021, 12(18):5664–73., DOI 10.7150/jca.61199 Huang DD, Yu DY, Song HN, Wang WB, Luo X, Wu GF, et al. The relationship between the GLIM-defined malnutrition, body composition and functional parameters, and clinical outcomes in elderly patients undergoing radical gastrectomy for gastric cancer. Eur J Surg Oncol, 2021, 47(9):2323–31., DOI 10.1016/j.ejso.2021.02.032 Kim J, Han SH, Kim H-I. Detection of sarcopenic obesity and prediction of long-term survival in patients with gastric cancer using preoperative computed tomography and machine learning. J Surg Oncol, 2021, 124(8):1347–55., DOI 10. 1002/jso.26668 Kim KW, Lee K, Lee JB, Park T, Khang S, Jeong H, et al. Preoperative nutritional risk index and postoperative one-year skeletal muscle loss can predict the prognosis of patients with gastric adenocarcinoma: A registry-based study. BMC Cancer, 2021, 21(1):157., DOI 10.1186/s12885-021-07885-7 Kim YY, Lee J, Jeong WK, Kim ST, Kim JH, Hong JY, et al. Prognostic significance of sarcopenia in microsatellite-stable gastric cancer patients treated with programmed death-1 inhibitors. Gastric Cancer, 2021, 24(2):457–66., DOI 10. 1007/s10120-020-01124-x Lee JK, Park YS, Lee K, Youn SI,Won Y, Min SH, et al. Prognostic significance of surgery-induced sarcopenia in the survival of gastric cancer patients: A sex-specific analysis. J Cachexia Sarcopenia Muscle, 2021, 12(6):1897–907., DOI 10.1002/jcsm.12793 Matsunaga T, Saito H, Miyauchi W, Shishido Y, Miyatani K, Morimoto M, et al. Impact of skeletal muscle mass in patients with unresectable gastric cancer who received palliative first-line chemotherapy based on 5-fluorouracil. Bmc Cancer, 2021, 21(1):1219., DOI 10.1186/s12885-021-08953-8 Rimini M, Pecchi A, Prampolini F, Bussei C, Salati M, Forni D, et al. The prognostic role of early skeletal muscle mass depletion in multimodality management of patients with advanced gastric cancer treated with first line chemotherapy: A pilot experience from modena cancer center. J Clin Med, 2021, 10(8):1705., DOI 10.3390/jcm10081705 Sakurai K, Kubo N, Tamamori Y, Aomatsu N, Nishii T, Tachimori A, et al. Depletion of skeletal muscle mass adversely affects long-term outcomes for men undergoing gastrectomy for gastric cancer. Plos One, 2021, 16(8):e0256365., DOI 10. 1371/journal.pone.0256365 Taki Y, Sato S, Nakatani E, Higashizono K, Nagai E, Nishida M, et al. Preoperative skeletal muscle index and visceral-to-subcutaneous fat area ratio are associated with long-term outcomes of elderly gastric cancer patients after gastrectomy. Langenbecks Arch Surg, 2021, 406(2):463–71., DOI 10.1007/s00423- 021-02092-1 Matsui R, Inaki N, Tsuji T. Impact of diabetes mellitus on long-term prognosis after gastrectomy for advanced gastric cancer: A propensity score matching analysis. Surg Today, 2022, 52:1382–91., DOI 10.1007/s00595-022-02482-y Ricciardolo AA, De Ruvo N, Serra F, Prampolini F, Solaini L, Battisti S, et al. Strong impact of sarcopenia as a risk factor of survival in resected gastric cancer patients: First Italian report of a bicentric study. Updates Surg, 2022, 74(1):283–93., DOI 10.1007/s13304-021-01175-4 Tan S, Zhuang Q, Zhang Z, Li S, Xu J, Wang J, et al. Postoperative loss of skeletal muscle mass predicts poor survival after gastric cancer surgery. Front Nutr, 2022, 9:794576., DOI 10.3389/fnut.2022.794576 Xiong J, Hu H, Kang W, Shao X, Li Y, Jin P, et al. Association of sarcopenia and expression of interleukin-16 in gastric cancer survival. Nutrients, 2022, 14(4): 838., DOI 10.3390/nu14040838 Chen F, Chi J, Zhao B, Mei F, Gao Q, Zhao L, et al. Impact of preoperative sarcopenia on postoperative complications and survival outcomes of patients with esophageal cancer: A meta-analysis of cohort studies. Dis Esophagus : Off J Int Soc Dis Esophagus, 2022, 35:doab100., DOI 10.1093/dote/doab100 Li S, Wang T, Lai W, Zhang M, Cheng B, Wang S, et al. Prognostic impact of sarcopenia on immune-related adverse events in malignancies received immune checkpoint inhibitors: A systematic review and meta-analysis. Transl Cancer Res, 2021, 10(12):5150–8., DOI 10.21037/tcr-21-1470 Dolin TG,Mikkelsen MK, Jakobsen HL, Vinther A, Zerahn B, Nielsen DL, et al. The prevalence of sarcopenia and cachexia in older patients with localized colorectal cancer. J Geriatr Oncol, 2023, 14(1):101402., DOI 10.1016/j.jgo.2022.11.001 Petermann-Rocha F, Balntzi V, Gray SR, Lara J, Ho FK, Pell JP, et al. Global prevalence of sarcopenia and severe sarcopenia: A systematic review and metaanalysis. J Cachexia Sarcopenia Muscle, 2022, 13(1):86–99., DOI 10.1002/jcsm.12783 Takenaka Y, Takemoto N, Oya R, Inohara H. Prognostic impact of sarcopenia in patients with head and neck cancer treated with surgery or radiation: A metaanalysis. PLoS One, 2021, 16(10):e0259288., DOI 10.1371/journal.pone.0259288 Waalboer RB, Meyer YM, Galjart B, Olthof PB, van Vugt JLA, Grunhagen DJ, et al. Sarcopenia and long-term survival outcomes after local therapy for colorectal liver metastasis: A meta-analysis. HPB : Off J Int Hepato Pancreato Biliary Assoc, 2022, 24(1):9–16., DOI 10.1016/j.hpb.2021.08.947 Watanabe J, Matsui R, Sasanuma H, Ishizaki Y, Fukunaga T, Kotani K, et al. Body composition assessment and sarcopenia in patients with biliary tract cancer: A systematic review and meta-analysis. Clin Nutr, Edinburgh, Scotland,, 2022, 41(2): 321–8., DOI 10.1016/j.clnu.2021.12.005 Schiaffino S, Dyar KA, Ciciliot S, Blaauw B, Sandri M. Mechanisms regulating skeletal muscle growth and atrophy. FEBS J, 2013, 280(17):4294–314., DOI 10.1111/ febs.12253 Webster JM, Kempen L, Hardy RS, Langen RCJ. Inflammation and skeletal muscle wasting during cachexia. Front Physiol, 2020, 11:597675., DOI 10.3389/fphys. 2020.597675 Ekramzadeh M, Santoro D, Kopple JD. The effect of nutrition and exercise on body composition, exercise capacity, and physical functioning in advanced CKD patients. Nutrients, 2022, 14(10):2129., DOI 10.3390/nu14102129 Dasarathy S, Merli M. Sarcopenia from mechanism to diagnosis and treatment in liver disease. J Hepatol, 2016, 65(6):1232–44., DOI 10.1016/j.jhep. 2016.07.040 Tian J, Chung HK, Moon JS, Nga HT, Lee HY, Kim JT, et al. Skeletal muscle mitoribosomal defects are linked to low bone mass caused by bone marrow inflammation in male mice. J Cachexia Sarcopenia Muscle, 2022, 13(3):1785–99., DOI 10.1002/jcsm.12975 Nishikawa H, Fukunishi S, Asai A, Yokohama K, Nishiguchi S, Higuchi K. Pathophysiology and mechanisms of primary sarcopenia, Review). Int J Mol Med, 2021, 48(2):156., DOI 10.3892/ijmm.2021.4989 Par A, Hegyi JP, Vancsa S, Par G. Sarcopenia - 2021: Pathophysiology, diagnosis, therapy. Orvosi Hetilap, 2021, 162(1):3–12., DOI 10.1556/650.2021.32015 Awad S, Tan BH, Cui H, Bhalla A, Fearon KC, Parsons SL, et al. Marked changes in body composition following neoadjuvant chemotherapy for oesophagogastric cancer. Clin Nutr, Edinburgh, Scotland,, 2012, 31(1):74–7., DOI 10.1016/j.clnu.2011.08.008 Palmela C, Velho S, Agostinho L, Branco F, Santos M, Santos MP, et al. Body composition as a prognostic factor of neoadjuvant chemotherapy toxicity and outcome in patients with locally advanced gastric cancer. J Gastric Cancer, 2017, 17(1):74–87., DOI 10.5230/jgc.2017.17.e8 Erkul O, Cekic AB, Cansu A, Yildirim R, Guner A. Effects of sarcopenia on postoperative outcomes in patients who underwent gastrectomy for gastric cancer. J Surg Res, 2022, 274:196–206., DOI 10.1016/j.jss.2021.12.051 Hisada H, Tamura N, Tsuji Y, Nagao S, Fukagawa K, Miura Y, et al. The impact of sarcopenia on adverse events associated with gastric endoscopic submucosal dissection. Surg Endosc, 2022, 36:6387–95., DOI 10.1007/s00464-021- 08965-2 Huang ZX, Zhang HH, Zhang WT, Shi MM, Ren JH, Xu LB, et al. Effect of short-term preoperative parenteral nutrition support for gastric cancer patients with sarcopenia: A propensity score matching analysis. J Gastrointest Surg, 2022, 26:1362–72., DOI 10.1007/s11605-021-05185-w Kim HJ, Lee ES, Kim BJ, Kim WS, Park JY, Kim JG, et al. Risk factors and clinical outcomes of postgastrectomy sarcopenia newly developed after curative resection for gastric cancer. Medicine, Baltimore,, 2022, 101(6):e28699., DOI 10. 1097/MD.0000000000028699 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611055 EP 1611064 DI 10.3389/pore.2023.1611055 PG 10 ER PT J AU Zhu, J Zhu, X Xie, F Ding, Y Lu, H Dong, Y Li, P Fu, J Liang, A Zeng, Y Xiu, B AF Zhu, Jingjing Zhu, Xinyu Xie, Fengyang Ding, Yi Lu, Huina Dong, Yan Li, Ping Fu, Jianfei Liang, Aibin Zeng, Yu Xiu, Bing TI Case report: Circulating tumor DNA technology displays temporal and spatial heterogeneity in Waldenstrom macroglobulinemia during treatment with BTK inhibitors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE resistance; Waldenstrom macroglobulinemia; circulating tumor DNA (ctDNA); BTK inhibitor; ascites ID resistance; Waldenstrom macroglobulinemia; circulating tumor DNA (ctDNA); BTK inhibitor; ascites AB Background: Waldenstrom macroglobulinemia (WM) is a rare subtype of B-cell lymphoma. Rituximab-based combination therapy and Bruton’s tyrosine kinase (BTK) inhibitors have greatly improved the prognosis of WM. Despite the high response rate and good tolerance of BTK inhibitors in treatment of WM, a proportion of patients still experience disease progression. Case presentation: We report a 55-year-old man with relapsed WM. The patient achieved partial remission after six courses of CHOP chemotherapy and multiple plasma exchanges in initial treatment. He was admitted to the hospital with abdominal distension, and was diagnosed with relapsed WM and subsequently started on zanubrutinib. Disease progression and histological transformation occurred during treatment. We performed liquid biopsies on transformed plasma, tumor tissue and ascites at the same time and found high consistency between ascites and tissues. Moreover, we detected resistance mutations of BTK inhibitors (BTK, PLCG2) in ascites that were not detected in plasma or tissue. Eventually, the patient died during the 15-month follow-up after relapse. Conclusion: We describe a rare case of WM transformation to DLCBCL treated with chemoimmunotherapy and BTK inhibition. We analyzed tumor DNA obtained at different anatomic sites and circulating tumor DNA (ctDNA) derived from plasma and ascites specimens, with apparent significant temporal and spatial heterogeneity. The case specifically highlights the clinical value of ctDNA of ascites supernatant from WM patients, which is a more convenient and relatively noninvasive method compared with traditional invasive tissue biopsy. C1 [Zhu, Jingjing] Tongji University School of Medicine, Tongji Hospital, Department of HematologyShanghai, China. [Zhu, Xinyu] Tongji University School of Medicine, Tongji Hospital, Department of HematologyShanghai, China. [Xie, Fengyang] Tongji University School of Medicine, Tongji Hospital, Department of HematologyShanghai, China. [Ding, Yi] Tongji University School of Medicine, Tongji Hospital, Department of HematologyShanghai, China. [Lu, Huina] Tongji University School of Medicine, Tongji Hospital, Department of HematologyShanghai, China. [Dong, Yan] Tongji University School of Medicine, Tongji Hospital, Department of HematologyShanghai, China. [Li, Ping] Tongji University School of Medicine, Tongji Hospital, Department of HematologyShanghai, China. [Fu, Jianfei] Tongji University School of Medicine, Tongji Hospital, Department of HematologyShanghai, China. [Liang, Aibin] Tongji University School of Medicine, Tongji Hospital, Department of HematologyShanghai, China. [Zeng, Yu] Tongji University School of Medicine, Tongji Hospital, Department of PathologyShanghai, China. [Xiu, Bing] Tongji University School of Medicine, Tongji Hospital, Department of HematologyShanghai, China. RP Xiu, B (reprint author), Tongji University School of Medicine, Tongji Hospital, Department of Hematology, Shanghai, China. EM xiubing1233@tongji.edu.cn CR Owen RG, Treon SP, Al-Katib A, Fonseca R, Greipp PR, McMaster ML, et al. Clinicopathological definition of waldenstrom’s macroglobulinemia: Consensus panel recommendations from the second international Workshop on waldenstrom’s macroglobulinemia. Semin Oncol, 2003, 30(2):110–5., DOI 10. 1053/sonc.2003.50082 Gertz MA. Waldenstrom macroglobulinemia: 2021 update on diagnosis, risk stratification, and management. Am J Hematol, 2021, 96(2):258–69., DOI 10.1002/ ajh.26082 Treon SP, Cao Y, Xu L, Yang G, Liu X, Hunter ZR. Somatic mutations in MYD88 and CXCR4 are determinants of clinical presentation and overall survival in Waldenstrom macroglobulinemia. Blood, 2014, 123(18):2791–6., DOI 10.1182/ blood-2014-01-550905 Castillo JJ, Advani RH, Branagan AR, Buske C, Dimopoulos MA, D’Sa S, et al. Consensus treatment recommendations from the tenth international Workshop for Waldenstrom macroglobulinaemia. Lancet Haematol, 2020, 7(11):e827–e837., DOI 10.1016/s2352-3026(20)30224-6 Bagratuni T, Ntanasis-Stathopoulos I, Gavriatopoulou M, Mavrianou- Koutsoukou N, Liacos C, Patseas D, et al. Detection of MYD88 and CXCR4 mutations in cell-free DNA of patients with IgM monoclonal gammopathies. Leukemia, 2018, 32(12):2617–25., DOI 10.1038/s41375-018- 0197-7 Jang H, Choi CM, Lee SH, Lee S, Jeong MK. Read count patterns and detection of cancerous copy number alterations in plasma cell-free DNA whole exome sequencing data for advanced non-small cell lung cancer. Int J Mol Sci, 2022, 23(21):12932., DOI 10.3390/ijms232112932 Li M, Chen J, Zhang B, Yu J, Wang N, Li D, et al. Dynamic monitoring of cerebrospinal fluid circulating tumor DNA to identify unique genetic profiles of brain metastatic tumors and better predict intracranial tumor responses in nonsmall cell lung cancer patients with brain metastases: A prospective cohort study, GASTO 1028). BMC Med, 2022, 20(1):398., DOI 10.1186/s12916-022-02595-8 Husain H, Nykin D, Bui N, Quan D, Gomez G, Woodward B, et al. Cell-Free DNA from ascites and pleural effusions: Molecular insights into genomic aberrations and disease biology. Mol Cancer Ther, 2017, 16(5):948–55., DOI 10. 1158/1535-7163.Mct-16-0436 Treon SP. How I treat Waldenstrom macroglobulinemia. Blood, 2009, 114(12): 2375–85., DOI 10.1182/blood-2009-05-174359 Treon SP, Xu L, Guerrera ML, Jimenez C, Hunter ZR, Liu X, et al. Genomic landscape of Waldenstrom macroglobulinemia and its impact on treatment strategies. J Clin Oncol, 2020, 38(11):1198–208., DOI 10.1200/jco.19.02314 Siravegna G,Marsoni S, Siena S, Bardelli A. Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol, 2017, 14(9):531–48., DOI 10.1038/ nrclinonc.2017.14 Kwok M,Wu SP, Mo C, Summers T, Roschewski M. Circulating tumor DNA to monitor therapy for aggressive B-cell lymphomas. Curr Treat Options Oncol, 2016, 17(9):47., DOI 10.1007/s11864-016-0425-1 Nakamura S, Yokoyama K, Shimizu E, Yusa N, Kondoh K, Ogawa M, et al. Prognostic impact of circulating tumor DNA status post-allogeneic hematopoietic stem cell transplantation in AML and MDS. Blood, 2019, 133(25):2682–95., DOI 10. 1182/blood-2018-10-880690 Yeh P, Hunter T, Sinha D, Ftouni S, Wallach E, Jiang D, et al. Circulating tumour DNA reflects treatment response and clonal evolution in chronic lymphocytic leukaemia. Nat Commun, 2017, 8:14756., DOI 10.1038/ncomms14756 Allegra A, Cancemi G, Mirabile G, Tonacci A, Musolino C, Gangemi S. Circulating tumour cells, cell free DNA and tumour-educated platelets as reliable prognostic and management biomarkers for the liquid biopsy in multiple myeloma. Cancers, Basel,, 2022, 14(17):4136., DOI 10.3390/cancers14174136 Ntanasis-Stathopoulos I, Bagratuni T, GavriatopoulouM, Patseas D, Liacos C, Kanellias N, et al. Cell-free DNA analysis for the detection of MYD88 and CXCR4 mutations in IgM monoclonal gammopathies; an update with clinicopathological correlations. Am J Hematol, 2020, 95(6):E148–E150., DOI 10. 1002/ajh.25802 Bagratuni T, Markou A, Patseas D, Mavrianou-Koutsoukou N, Aktypi F, Liacos CI, et al. Determination of MYD88L265P mutation fraction in IgM monoclonal gammopathies. Blood Adv, 2022, 6(1):189–99., DOI 10.1182/ bloodadvances.2021005354 Alegria-Landa V, Prieto-Torres L, Santonja C, Cordoba R, Manso R, Requena L, et al. MYD88 L265P mutation in cutaneous involvement by Waldenstrom macroglobulinemia. J Cutan Pathol, 2017, 44(7):625–31., DOI 10.1111/cup.12944 Gustine JN, Meid K, Hunter ZR, Xu L, Treon SP, Castillo JJ. MYD88 mutations can be used to identify malignant pleural effusions in Waldenstrom macroglobulinaemia. Br J Haematol, 2018, 180(4):578–81., DOI 10. 1111/bjh.14386 Hiemcke-Jiwa LS, Minnema MC, Radersma-van Loon JH, Jiwa NM, de Boer M, Leguit RJ, et al. The use of droplet digital pcr in liquid biopsies: A highly sensitive technique for MYD88 p.(L265P, detection in cerebrospinal fluid. Hematological Oncol, 2018, 36(2):429–35., DOI 10.1002/hon.2489 Wu H, Ji H, Yang W, Zhang M, Guo Y, Li B, et al. Liquid biopsy using ascitic fluid and pleural effusion supernatants for genomic profiling in gastrointestinal and lung cancers. BMC Cancer, 2022, 22(1):1020., DOI 10. 1186/s12885-022-09922-5 Zhou S, Xu B, Qi L, Zhu D, Liu B, Wei J. Next-generation sequencing reveals mutational accordance between cell-free DNA from plasma, malignant pleural effusion and ascites and directs targeted therapy in a gastric cancer patient. Cancer Biol Ther, 2019, 20(1):15–20., DOI 10.1080/15384047.2018. 1504720 Treon SP, Tripsas CK, Meid K, Warren D, Varma G, Green R, et al. Ibrutinib in previously treated Waldenstrom’s macroglobulinemia. New Engl J Med, 2015, 372(15):1430–40., DOI 10.1056/NEJMoa1501548 Argyropoulos KV, Palomba ML. First-Generation and second-generation Bruton tyrosine kinase inhibitors in Waldenstrom macroglobulinemia. Hematol Oncol Clin North Am, 2018, 32(5):853–64., DOI 10.1016/j.hoc.2018.05.012 Tam CS, Opat S, D’Sa S, Jurczak W, Lee HP, Cull G, et al. A randomized phase 3 trial of zanubrutinib vs ibrutinib in symptomatic Waldenstrom macroglobulinemia: The ASPEN study. Blood, 2020, 136(18):2038–50., DOI 10. 1182/blood.2020006844 Sharma S, Galanina N, Guo A, Lee J, Kadri S, Van Slambrouck C, et al. Identification of a structurally novel BTK mutation that drives ibrutinib resistance in CLL. Oncotarget, 2016, 7(42):68833–41., DOI 10.18632/oncotarget.11932 Woyach JA, Furman RR, Liu TM, Ozer HG, Zapatka M, Ruppert AS, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. New Engl J Med, 2014, 370(24):2286–94., DOI 10.1056/NEJMoa1400029 Liu TM, Woyach JA, Zhong Y, Lozanski A, Lozanski G, Dong S, et al. Hypermorphic mutation of phospholipase C, γ2 acquired in ibrutinib-resistant CLL confers BTK independency upon B-cell receptor activation. Blood, 2015, 126(1): 61–8., DOI 10.1182/blood-2015-02-626846 Xu L, Tsakmaklis N, Yang G, Chen JG, Liu X, Demos M, et al. Acquired mutations associated with ibrutinib resistance in Waldenstrom macroglobulinemia. Blood, 2017, 129(18):2519–25., DOI 10.1182/blood-2017-01-761726 Chen JG, Liu X, Munshi M, Xu L, Tsakmaklis N, Demos MG, et al. BTK(Cys481Ser, drives ibrutinib resistance via ERK1/2 and protects BTK(wildtype, MYD88-mutated cells by a paracrine mechanism. Blood, 2018, 131(18): 2047–59., DOI 10.1182/blood-2017-10-811752 Bodor C, Kotmayer L, Laszlo T, Takacs F, Barna G, Kiss R, et al. Screening and monitoring of the BTK(C481S, mutation in a real-world cohort of patients with relapsed/refractory chronic lymphocytic leukaemia during ibrutinib therapy. Br J Haematol, 2021, 194(2):355–64., DOI 10.1111/bjh.17502 Maddocks KJ, Ruppert AS, Lozanski G, Heerema NA, Zhao W, Abruzzo L, et al. Etiology of ibrutinib therapy discontinuation and outcomes in patients with chronic lymphocytic leukemia. JAMA Oncol, 2015, 1(1):80–7., DOI 10.1001/ jamaoncol.2014.218 Talaulikar D, Biscoe A, Lim JH, Gibson J, Arthur C, Mackinlay N, et al. Genetic analysis of Diffuse Large B-cell Lymphoma occurring in cases with antecedent Waldenstrom Macroglobulinaemia reveals different patterns of clonal evolution. Br J Haematol, 2019, 185(4):767–70., DOI 10.1111/bjh.15610 Poulain S, Roumier C, Bertrand E, Renneville A, Caillault-Venet A, Doye E, et al. TP53 mutation and its prognostic significance in waldenstrom’s macroglobulinemia. Clin Cancer Res, 2017, 23(20):6325–35., DOI 10.1158/1078- 0432.Ccr-17-0007 GavriatopoulouM, Terpos E, Kastritis E, Dimopoulos MA. Current treatment options and investigational drugs for Waldenstrom’s Macroglobulinemia. Expert Opin Investig Drugs, 2017, 26(2):197–205., DOI 10.1080/13543784.2017.1275561 Lin P, Mansoor A, Bueso-Ramos C, Hao S, Lai R, Medeiros LJ. Diffuse large B-cell lymphoma occurring in patients with lymphoplasmacytic lymphoma/ Waldenstrom macroglobulinemia. Clinicopathologic features of 12 cases. Am J Clin Pathol, 2003, 120(2):246–53., DOI 10.1309/r01v-xg46-mfcd-vnhl Castillo JJ, Gustine J, Meid K, Dubeau T, Hunter ZR, Treon SP. Histological transformation to diffuse large B-cell lymphoma in patients with Waldenstrom macroglobulinemia. Am J Hematol, 2016, 91(10):1032–5., DOI 10.1002/ajh.24477 Durot E, Tomowiak C, Michallet AS, Dupuis J, Hivert B, Lepretre S, et al. Transformed Waldenstrom macroglobulinaemia: Clinical presentation and outcome. A multi-institutional retrospective study of 77 cases from the French innovative leukemia organization, FILO). Br J Haematol, 2017, 179(3):439–48., DOI 10.1111/bjh.14881 Jimenez C, Alonso-Alvarez S, Alcoceba M, Ordonez GR, Garcia-Alvarez M, Prieto-Conde MI, et al. From Waldenstrom’s macroglobulinemia to aggressive diffuse large B-cell lymphoma: A whole-exome analysis of abnormalities leading to transformation. Blood Cancer J, 2017, 7(8):e591., DOI 10.1038/bcj.2017.72 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611070 EP 1611078 DI 10.3389/pore.2023.1611070 PG 9 ER PT J AU Revesz, J Posfai, B Pajor, L Papdan, T Varga, L Paczona, V Varga, Z Sukosd, F Maraz, A AF Revesz, Janos Posfai, Boglarka Pajor, Laszlo Papdan, Timea Varga, Linda Paczona, R. Viktor Varga, Zoltan Sukosd, Farkas Maraz, Aniko TI Correlation between fibroblast growth factor receptor mutation, programmed death ligand-1 expression and survival in urinary bladder cancer based on real-world data SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE urinary bladder cancer; fibroblast growth factor receptor; FGFR mutation; programmed death-ligand 1 expression; combined positive score ID urinary bladder cancer; fibroblast growth factor receptor; FGFR mutation; programmed death-ligand 1 expression; combined positive score AB Background: Programmed cell death (PD)-1/PD-ligand 1 (PD-L1) inhibitors have made a breakthrough in the therapy of advanced urothelial bladder cancer (UBC). The impact of Fibroblast Growth Factor Receptor 3 (FGFR3) mutation on the effectiveness of PD-L1 treatment remains still unclear. Objective: Our study aimed to investigate the frequency of FGFR mutations at different tumor stages, and their relation to PD-L1 status and survival. Methods: 310 patients with urothelial bladder cancer and subsequent radical cystectomy were included in a retrospective study over a 10-year study period at the University of Szeged, Hungary. FGFR3 mutations from the most infiltrative areas of the tumor were analyzed by targeted next-generation sequencing and PD-L1 (28-8 DAKO) tests (tumor positive score -TPS and combined positives score–CPS). In T0 cases FGFR3 mutations were analyzed from the earlier resection samples. Survival and oncological treatment data were collected from the National Health Insurance Fund (NHIF). Neoadjuvant, adjuvant and palliative conventional chemotherapies were allowed; immunotherapies were not. The relationship between the covariates was tested using chi-square tests, and survival analysis was performed using the Kaplan-Meier model and Cox proportional hazards regression. Results: PD-L1 and FGFR could be tested successfully in 215 of the 310 UBC samples [pT0cyst 19 (8.8%); St.0-I 43 (20%); St.II 41 (19%); St.III-IV 112 (52%)]. Significant pairwise dependency was found between tumor stage, FGFR3 mutation status and PD-L1 expression (p < 0.01). Samples with FGFR mutation were more common in less advanced stages and were also less likely to demonstrate PD-L1 expression. The effect of all investigated factors on survival was found to correlate with tumor stage. Conclusion: FGFR alteration frequency varied between the different stages of cancer. Higher positivity rates were observed at early stages, but lower levels of PD-L1 expression were detected in patients with FGFR mutations across at all stages of the disease. C1 [Revesz, Janos] Szegedi Tudomanyegyem, Doktori IskolaSzeged, Hungary. [Posfai, Boglarka] University of Szeged, Department of OncotherapySzeged, Hungary. [Pajor, Laszlo] University of Szeged, Department of UrologySzeged, Hungary. [Papdan, Timea] University of Szeged, Department of OncotherapySzeged, Hungary. [Varga, Linda] University of Szeged, Department of OncotherapySzeged, Hungary. [Paczona, R. Viktor] University of Szeged, Department of OncotherapySzeged, Hungary. [Varga, Zoltan] University of Szeged, Department of OncotherapySzeged, Hungary. [Sukosd, Farkas] University of Szeged, Department of PathologySzeged, Hungary. [Maraz, Aniko] University of Szeged, Department of OncotherapySzeged, Hungary. RP Maraz, A (reprint author), University of Szeged, Department of Oncotherapy, Szeged, Hungary. EM dr.aniko.maraz@gmail.com CR Richters A, Aben KKH, Kiemeney LALM. The global burden of urinary bladder cancer: An update. World J Urol, 2020, 38:1895–904., DOI 10.1007/s00345-019- 984-4 2. Witjes JA, Bruins HM, Cathomas R, Hernandez V, Neuzillet Y, Comperat EM, et al. The importance of hospital and surgeon volume as major determinants of morbidity and mortality after radical cystectomy for bladder cancer: A systematic review and recommendations by the European association of urology muscleinvasive and metastatic bladder cancer guideline panel. Eur Urol Oncol, 2020, 3: 131–44., DOI 10.1016/j.euo.2019.11.005 Flaig TW, Spiess PE, Abern M. NCCN Guidelines® insights: Bladder cancer, 2022). Version 2.2022., DOI 10.6004/jnccn.2022.0041 Gajewski TF, Woo SR, Zha Y, Spaapen R, Zheng Y, Corrales L, et al. Cancer immunotherapy strategies based on overcoming barriers within the tumor microenvironment. Curr Opin Immunol, 2013, 2:268–76., DOI 10.1016/j.coi.2013. 02.009 Ding X, Chen Q, Yang Z, Li J, Zhan H, Lu N, et al. Clinicopathological and prognostic value of PD-L1 in urothelial carcinoma: A meta-analysis. Cancer Manag Res, 2019, 11:4171–84., DOI 10.2147/CMAR.S176937 Liu J, Zhang C, Hu J, Tian Q, Wang X, Gu H, et al. Effectiveness of anti-PD-1/ PD-L1 antibodies in urothelial carcinoma patients with different PD-L1 expression levels: A meta-analysis. Oncotarget, 2018, 9:12400–7., DOI 10.18632/oncotarget. 24249 Balar AV, Castellano D,O’Donnell PH, Grivas P, Vuky J, Powles T, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer, KEYNOTE-052): A multicentre, single-arm, phase 2 study. Lancet Oncol, 2017, 18:1483–92., DOI 10.1016/S1470-2045(17)30616-2 O’Donnell PH, Balar AV, Vuky J, Bellmunt J, Powles T, Bajorin DF, et al. Firstline pembrolizumab, pembro, in cisplatin-ineligible patients with advanced urothelial cancer, UC): Response and survival results up to five years from the KEYNOTE-052 phase 2 study. JCO, 2021, 39:4508., DOI 10.1200/JCO.2021.39.15_ suppl.4508 Cancer Genome Atlas Research Network. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature, 2014, 507:315–22., DOI 10.1038/nature12965 Kamoun A, de Reynies A, Allory Y, Sjodahl G, Robertson AG, Seiler R, et al. A Consensus molecular classification of muscle-invasive bladder cancer. Cancer Eur Urol, 2020, 77:420–33., DOI 10.1016/j.eururo.2019.09.006 Robertson AG, Kim J, Al-Ahmadie H, Bellmunt J, Guo G, Cherniack AD, et al. Comprehensive molecular characterization of muscle-invasive bladder cancer. Cel, 2017, 171:540–56.e25., DOI 10.1016/j.cell.2017.09.007 Turner N. Fibroblast growth factor signalling: From development to cancer. Nat Rev Cancer, 2010, 10:116–29., DOI 10.1038/nrc2780 Krook MA, Reeser JW, Barker H, Wilberding M, Li G. Fibroblast growth factor receptors in cancer: Genetic alterations, diagnostics, therapeutic targets and mechanisms of resistance. Br J Cancer, 2021, 124:880–92., DOI 10.1038/s41416-020- 01157-0 di Martino E, Tomlinson DC, Williams SV, Knowles MA. A place for precision medicine in bladder cancer: Targeting the FGFRs. Future Oncol, 2016, 12:2243–63., DOI 10.2217/fon-2016-0042 Siefker-Radtke A, Necchi A, Rosenbaum E, Culine S, Burgess EF, O’Donnell PH, et al. Efficacy of programmed death 1, PD-1, and programmed death 1 ligand, PD-L1, inhibitors in patients with FGFR mutations and gene fusions: Results from a data analysis of an ongoing phase 2 study of erdafitinib, JNJ-42756493, in patients, pts, with advanced urothelial cancer, UC). J Oncol, 2018, 36:450., DOI 10.1200/jco. 2018.36.6_suppl.450 Sweis RF, Spranger S, Bao R, Paner GP, StadlerWM, Steinberg G, et al. Molecular drivers of the non-T-cell-inflamed tumormicroenvironment in urothelial bladder cancer. Cancer Immunol Res, 2016, 4:563–8., DOI 10.1158/2326-6066.CIR-15-0274 Wang L, Gong Y, Saci A, Szabo PM, Martini A, Necchi A, et al. Fibroblast growth factor receptor 3 alterations and response to PD-1/PD-L1 blockade in patients with metastatic urothelial cancer. EurUrol, 2019, 76:599–603., DOI 10.1016/j.eururo.2019.06.025 Kalebasty AR, Benjamin DJ, Loriot Y, Papantoniou D, Siefker-Radtke AO, Necchi A, et al. Outcomes of patients with advanced urothelial carcinoma after antiprogrammed death-(ligand, 1 therapy by fibroblast growth factor receptor gene alteration status: An observational study. Eur Urol Open Sci, 2023, 47:48–57., DOI 10. 1016/j.euros.2022.11.001 Palumbo C, Pecoraro A, Rosiello G, Luzzago S, Deuker M, Stolzenbach F, et al. Bladder cancer incidence rates and trends in young adults aged 20-39 years. Urol Oncol Semin Orig, 2020, 38:934.e11–934.934.e19., DOI 10.1016/j.urolonc.2020.06.009 Yin M, Joshi M, Meier RP, Glantz M, Holder S, Harvey HA, Neoadjuvant chemotherapy for muscle-invasive bladder cancer: A systematic review and two-step meta-analysis. Oncologist, 21, 2016). p. 708–15., DOI 10.1634/theoncologist.2015-0440 Maraz A, Varga L, Posfai B, Geczi L, Kuronya Z. New aspects of chemotherapy and indications for maintenance immuno-therapy in urothelial cancers. Magy Onkol, 2021, 65:329–37. Necchi A, Raggi D, Gallina A, Madison R, Colecchia M, Luciano R, et al. Updated results of PURE-01 with preliminary activity of neoadjuvant pembrolizumab in patients with muscle-invasive bladder carcinoma with variant histologies. Eur Urol, 2020, 77:439–46., DOI 10.1016/j.eururo.2019.10.026 Tilki D, Svatek RS, Novara G, Seitz M, Godoy G, Karakiewicz PI, et al. Stage pT0 at radical cystectomy confers improved survival: An international study of 4,430 patients. J Urol, 2010, 184:888–94., DOI 10.1016/j.juro.2010.04.081 Ding X, Chen Q, Yang Z, Li J, Zhan H, Lu N, et al. Clinicopathological and prognostic value of PD-L1 in urothelial carcinoma: A meta-analysis. Cancer Manag Res, 2019, 11:4171–84., DOI 10.2147/CMAR.S176937 Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature, 2013, 499:214–8., DOI 10.1038/nature12213 Billerey C, Chopin D, Aubriot-Lorton MH, Ricol D, Gil Diez de Medina S, Van Rhijn B, et al. Frequent FGFR3 mutations in papillary non-invasive bladder, pTa, tumors. Am J Pathol, 2001, 158:1955–9., DOI 10.1016/S0002-9440(10)64665-2 Kawahara T, Kojima T, Kandori S, Kurobe M, Yoshino T, Kimura T, et al. TP53 codon 72 polymorphism is associated with FGFR3 and RAS mutation in nonmuscle- invasive bladder cancer. PLoS One, 2019, 14:e0220173., DOI 10.1371/journal. pone.0220173 Fernandez S, Lacalle M, Moreno R, Barquin Garcia A, Yague Fernandez M, Navarro Alcaraz P, et al. Prognostic value and clinical significance of FGFR genomic alterations, GAs, in metastatic urothelial cancer patients. J Clin Med, 2022, 11: 4483., DOI 10.3390/jcm11154483 Hernandez S, Lopez-Knowles E, Lloreta J, KogevinasM, Amoros A, Tardon A, et al. Prospective study of FGFR3 mutations as a prognostic factor in nonmuscle invasive urothelial bladder carcinomas. J Clin Oncol, 2006, 24:3664–71., DOI 10. 1200/JCO.2005.05.1771 Casadei C, Dizman N, Schepisi G, Cursano MC, Basso U, Santini D, et al. Targeted therapies for advanced bladder cancer: New strategies with FGFR inhibitors. Ther Adv Med Oncol, 2019, 11:1758835919890285., DOI 10.1177/ 1758835919890285 Kim J, Kwiatkowski D,McConkey DJ,Meeks JJ, Freeman SS, Bellmunt J, et al. The cancer genome Atlas expression subtypes stratify response to checkpoint inhibition in advanced urothelial cancer and identify a subset of patients with high survival probability. Eur Urol, 2019, 75(6):961–4., DOI 10.1016/j.eururo.2019. 02.017 Joerger M, Cassier P, Penel N, Cathomas R, Richly H, Schostak M, et al. Rogaratinib treatment of patients with advanced urothelial carcinomas prescreened for tumor FGFR mRNA expression. J Clin Oncol, 2018, 36(6):494., DOI 10.1200/jco. 2018.36.6_suppl.494 Kim WJ, Rose TL, Roghmann F, Eckstein M, Jarczyk J, Zengerling F, et al. Predictive value of fibroblast growth factor receptor, FGFR, alterations on anti-PD-, L)1 treatment outcomes in patients, Pts, with advanced urothelial cancer, UC): Pooled analysis of real-world data. J Clin Oncol, 2020, 38(6):493., DOI 10.1200/jco. 2020.38.6_suppl.493 Loriot Y, Necchi A, Park SH, Huddart R, Burgess E, Zhong B, et al. Erdafitinib compared with vinflunine or docetaxel or pembrolizumab in patients, pts, with metastatic or surgically unresectable, M/UR, urothelial carcinoma, UC, and selected fgfr gene alterations, FGFRalt): The phase III THOR study. Ann Oncol, 2018, 29:viii327–8., DOI 10.1093/annonc/ mdy283.129 Maraz A, Takacs P, Lawson J, Santiago-Walker A, Pajor L, Sukosd F, et al. Correlation between FGFR mutation and PD-L1 expression of urinary bladder cancers: A real-world based biomarker study. J Clin Oncol Oncol, 2019, 37:e16030., DOI 10.1200/jco.2019.37.15_suppl.e16030 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611077 EP 1611088 DI 10.3389/pore.2023.1611077 PG 12 ER PT J AU Li, H Yang, Ch Chen, K Sun, M AF Li, Hui Yang, Chenbo Chen, Kuisheng Sun, Miaomiao TI Expression significance of Emi1, UBCH10 and CyclinB1 in esophageal squamous cell carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tumor proliferation; Emi1; UBCH10; CyclinB1; tumor apoptosis ID tumor proliferation; Emi1; UBCH10; CyclinB1; tumor apoptosis AB Despite significant advances in the diagnosis and treatment of esophageal squamous cell carcinoma (ESCC), esophageal cancer is still a heavy social and medical burden due to its high incidence. Uncontrolled division and proliferation is one of the characteristics of tumor cells, which will promote rapid tumor growth and metastasis. Early mitotic inhibitor 1 (Emi1), ubiquitinconjugating enzyme 10 (UBCH10) and CyclinB1 are important proteins involved in the regulation of cell cycle. In this study, the expression of Emi1, UBCH10 and CyclinB1 in ESCC tissues and adjacent normal tissues will be analyzed by immunohistochemistry and in-situ hybridization techniques, and their relationship with tumor proliferation and apoptosis will be analyzed. The results showed that Emi1, UBCH10 and CyclinB1 genes and proteins were highly expressed in tumor tissues, which were correlated with tumor grade, lymph node metastasis and pathological stage, and positively correlated with tumor proliferation. Emi1, UBCH10 and CyclinB1 are also positively correlated. It is speculated that Emi1, UBCH10 and CyclinB1 genes synergically promote tumor proliferation and inhibit apoptosis, which may be potential diagnostic and therapeutic targets for ESCC. C1 [Li, Hui] The First Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. [Yang, Chenbo] The First Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. [Chen, Kuisheng] The First Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. [Sun, Miaomiao] The First Affiliated Hospital of Zhengzhou University, Department of PathologyZhengzhou, China. RP Sun, M (reprint author), The First Affiliated Hospital of Zhengzhou University, Department of Pathology, Zhengzhou, China. EM sunmiaomiaohd@163.com CR Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA, et al. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology, 2020, 159:335–49., DOI 10.1053/j.gastro.2020.02.068 Lezaja A, Altmeyer M. Dealing with DNA lesions: When one cell cycle is not enough. Curr Opin Cel Biol, 2021, 70:27–36., DOI 10.1016/j.ceb.2020.11.001 Zhao Y, Tang Q, Ni R, Huang X, Wang Y, Lu C, et al. Early mitotic inhibitor-1, an anaphase-promoting complex/cyclosome inhibitor, can control tumor cell proliferation in hepatocellular carcinoma: Correlation with Skp2 stability and degradation of p27(kip1). Hum Pathol, 2013, 44:365–73., DOI 10.1016/j.humpath. 2012.03.030 Presta I, Novellino F, Donato A, La Torre D, Palleria C, Russo E, et al. UbcH10 a major actor in cancerogenesis and a potential tool for diagnosis and therapy. Int J Mol Sci, 2020, 21:2041., DOI 10.3390/ijms21062041 Wood DJ, Endicott JA. Structural insights into the functional diversity of the CDK-cyclin family. Open Biol, 2018, 8:180112., DOI 10.1098/rsob.180112 Lei H, Wang K, Jiang T, Lu J, Dong X, Wang F, et al. KIAA0101 and UbcH10 interact to regulate non-small cell lung cancer cell proliferation by disrupting the function of the spindle assembly checkpoint. BMC Cancer, 2020, 20:957., DOI 10.1186/s12885-020-07463-3 Liu B, Liu Y, Wang Y, Xie C, Gan M, Han T, et al. CyclinB1 deubiquitination by USP14 regulates cell cycle progression in breast cancer. Pathol Res Pract, 2019, 215: 152592., DOI 10.1016/j.prp.2019.152592 Sun Y, Liu Y, Ma X, Hu H. The influence of cell cycle regulation on chemotherapy. Int J Mol Sci, 2021, 22:6923., DOI 10.3390/ijms22136923 Peters JM. The anaphase promoting complex/cyclosome: A machine designed to destroy. Nat Rev Mol Cel Biol., 2006, 7:644–56., DOI 10.1038/nrm1988 Yamano H. EMI1, a three-in-one ubiquitylation inhibitor. Nat Struct Mol Biol, 2013, 20:773–4., DOI 10.1038/nsmb.2626 Liu X, Wang H, Ma J, Xu J, Sheng C, Yang S, et al. The expression and prognosis of Emi1 and Skp2 in breast carcinoma: Associated with PI3K/akt pathway and cell proliferation. Med Oncol, 2013, 30:735., DOI 10.1007/s12032-013-0735-0 Donato G, Iofrida G, Lavano A, Volpentesta G, Signorelli F, Pallante PL, et al. Analysis of UbcH10 expression represents a useful tool for the diagnosis and therapy of astrocytic tumors. Clin Neuropathol, 2008, 27:219–23., DOI 10.5414/ npp27219 Jiang L, Bao Y, Luo C, Hu G, Huang C, Ding X, et al. Knockdown of ubiquitinconjugating enzyme E2C/UbcH10 expression by RNA interference inhibits glioma cell proliferation and enhances cell apoptosis in vitro. J Cancer Res Clin Oncol, 2010, 136:211–7., DOI 10.1007/s00432-009-0651-z Perrotta I, Bruno L, Maltese L, Russo E, Donato A, Donato G. Immunohistochemical analysis of the ubiquitin-conjugating enzyme UbcH10 in lung cancer: A useful tool for diagnosis and therapy. J Histochem Cytochem, 2012, 60:359–65., DOI 10.1369/0022155412439717 Pallante P, Malapelle U, Berlingieri MT, Bellevicine C, Sepe R, Federico A, et al. UbcH10 overexpression in human lung carcinomas and its correlation with EGFR and p53 mutational status. Eur J Cancer, 2013, 49:1117–26., DOI 10.1016/j. ejca.2012.09.033 Wang C, Pan YH, Shan M, Xu M, Bao JL, Zhao LM. Knockdown of UbcH10 enhances the chemosensitivity of dual drug resistant breast cancer cells to epirubicin and docetaxel. Int J Mol Sci, 2015, 16:4698–712., DOI 10.3390/ ijms16034698 Zhu B, Zhang Q, Wu Y, Luo J, Zheng X, Xu L, et al. SNAP23 suppresses cervical cancer progression via modulating the cell cycle. Gene, 2018, 673:217–24., DOI 10.1016/j.gene.2018.06.028 Cheng YM, Tsai CC, Hsu YC. Sulforaphane, a dietary isothiocyanate, induces G₂/M arrest in cervical cancer cells through CyclinB1 downregulation and gadd45β/ CDC2 association. Int J Mol Sci, 2016, 17:1530., DOI 10.3390/ijms17091530 Vaidyanathan S, Cato K, Tang L, Pavey S, Haass NK, Gabrielli BG, et al. In vivo overexpression of Emi1 promotes chromosome instability and tumorigenesis. Oncogene, 2016, 35:5446–55., DOI 10.1038/onc.2016.94 van Ree JH, Jeganathan KB, Malureanu L, van Deursen JM. Overexpression of the E2 ubiquitin-conjugating enzyme UbcH10 causes chromosome missegregation and tumor formation. J Cel Biol, 2010, 188:83–100., DOI 10.1083/jcb.200906147 Gavet O, Pines J. Progressive activation of CyclinB1-Cdk1 coordinates entry to mitosis. Dev Cel, 2010, 18:533–43., DOI 10.1016/j.devcel.2010.02.013 Lee H, Lee DJ, Oh SP, Park HD, Nam HH, Kim JM, et al. Mouse emi1 has an essential function in mitotic progression during early embryogenesis. Mol Cel Biol, 2006, 26:5373–81., DOI 10.1128/mcb.00043-06 Frye JJ, Brown NG, Petzold G, Watson ER, Grace CR, Nourse A, et al. Electron microscopy structure of human APC/C(CDH1)-EMI1 reveals multimodal mechanism of E3 ligase shutdown. Nat Struct Mol Biol, 2013, 20:827–35., DOI 10.1038/nsmb.2593 Bonacci T, Emanuele MJ. Impressionist portraits of mitotic exit: APC/C, K11- linked ubiquitin chains and cezanne. Cell Cycle, Georgetown, Tex.,, 2019, 18: 652–60., DOI 10.1080/15384101.2019.1593646 Wang W, Kirschner MW. Emi1 preferentially inhibits ubiquitin chain elongation by the anaphase-promoting complex. Nat Cel Biol, 2013, 15: 797–806., DOI 10.1038/ncb2755 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611081 EP 1611092 DI 10.3389/pore.2023.1611081 PG 12 ER PT J AU Long, J Cong, F Wei, Y Liu, J Tang, W AF Long, Junxian Cong, Fengyun Wei, Yousheng Liu, Jungang Tang, Weizhong TI Increased Kremen2 predicts worse prognosis in colon cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colon cancer; prognosis; bioinformatic; Kremen2; tumour infiltrating cells ID colon cancer; prognosis; bioinformatic; Kremen2; tumour infiltrating cells AB Background: Colon cancer (CC) is the fifth most prevalent cancer around the globe and poses a major risk to human health. Even though Kremen2 serves as a prognostic indicator in individuals with malignant tumours, its role in evaluating the prognosis of individuals with colon cancer has not been confirmed. Methods: Here, we examined the protein expression of Kremen2 in CC tissues and paired adjacent normal tissues by immunohistochemistry (IHC), then analyzed the clinical and RNA-seq data presented in The Cancer Genome Atlas (TCGA) database to confirm the relationship between Kremen2 levels and CC. In addition, the associations between Kremen2 mRNA expression and infiltrating immune cells were examined. Results: The study showed that the mRNA expression and protein level of Kremen2 were increased in CC tissues compared with adjacent normal tissues. According to Kaplan–Meier analysis, high Kremen2 expression in CC was linked to poor overall survival and progression-free survival. Clinical correlation analysis highlighted that a high level of Kremen2 expression was strongly linked with tumour progression, particularly lymph node metastasis. Cox regression analysis highlighted that Kremen2 was an independent prognostic indicator for CC. Bioinformatic studies highlighted that Kremen2 might be associated with the immune status in CC. Conclusion: Increased Kremen2 could serve as a potential prognostic CC biomarker. C1 [Long, Junxian] Guangxi Medical University Cancer Hospital, Department of Gastrointestinal SurgeryNanning, Guangxi, China. [Cong, Fengyun] Guangxi Medical University Cancer Hospital, Department of Gastrointestinal SurgeryNanning, Guangxi, China. [Wei, Yousheng] Guangxi Medical University Cancer Hospital, Department of Gynecologic OncologyNanning, Guangxi, China. [Liu, Jungang] Guangxi Medical University Cancer Hospital, Department of Gastrointestinal SurgeryNanning, Guangxi, China. [Tang, Weizhong] Guangxi Medical University Cancer Hospital, Department of Gastrointestinal SurgeryNanning, Guangxi, China. RP Tang, W (reprint author), Guangxi Medical University Cancer Hospital, Department of Gastrointestinal Surgery, Nanning, China. EM tangweizhong@gxmu.edu.cn CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 ChenW, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin, 2016, 66(2):115–32., DOI 10.3322/caac.21338 Puccini A, Berger MD, Naseem M, Tokunaga R, Battaglin F, Cao S, et al. Colorectal cancer: Epigenetic alterations and their clinical implications. Biochim Biophys Acta Rev Cancer, 2017, 1868(2):439–48., DOI 10.1016/j.bbcan.2017.09.003 Uppada SB, Gowrikumar S, Ahmad R, Kumar B, Szeglin B, Chen X, et al. MASTL induces Colon Cancer progression and Chemoresistance by promoting Wnt/β-catenin signaling. Mol Cancer, 2018, 17(1):111., DOI 10.1186/s12943-018- 0848-3 Kaprio T, Lindstrom AM, Rasila T, Saavalainen O, Beilmann-Lehtonen I, Mustonen H, et al. Elevated tumor expression of Astroprincin, FAM171A1, is an independent marker of poor prognosis in colon cancer. BMC Gastroenterol, 2021, 21(1):341., DOI 10.1186/s12876-021-01918-y Mengjie L, Xiao F, Lili J, Jiequn M, Xiaoqiang Z, Shuhong W, et al. Colon cancer cells secreted CXCL11 via RBP-Jκ to facilitated tumour-associated macrophageinduced cancer metastasis. J Cell Mol Med, 2021, 25(22):10575–90., DOI 10.1111/ jcmm.16989 Nakamura T, Aoki S, Kitajima K, Takahashi T, Matsumoto K, Nakamura T. Molecular cloning and characterization of Kremen, a novel kringle-containing transmembrane protein. Biochim Biophys Acta, 2001, 1518(1-2):63–72., DOI 10. 1016/s0167-4781(01)00168-3 Sumia I, Pierani A. Kremen1-induced cell death is regulated by homo- and heterodimerization. Cell Death Discov, 2019, 5:591., DOI 10.1038/s41420-019- 0175-5 Mao B, Wu W, Davidson G, Marhold J, Li M, Mechler BM, et al. Kremen proteins are Dickkopf receptors that regulate Wnt/beta-catenin signalling. Nature, 2002, 417(6889):664–7., DOI 10.1038/nature756 Mao B, Wu W, Li Y, Hoppe D, Stannek P, Glinka A, et al. LDL-receptorrelated protein 6 is a receptor for Dickkopf proteins. Nature, 2001, 411(6835): 321–5., DOI 10.1038/35077108 Hassler C, Cruciat CM, Huang YL, Kuriyama S, Mayor R. Kremen is required for neural crest induction in Xenopus and promotes LRP6-mediated Wnt signaling. Development, 2007, 134(23):4255–63., DOI 10.1242/dev.005942 Schulze J, Seitz S, Saito H, Schneebauer M, Marshall RP, Baranowsky A, et al. Negative regulation of bone formation by the transmembrane Wnt antagonist Kremen-2. PLoS One, 2010, 5(4):e10309., DOI 10.1371/journal.pone.0010309 Wen L. Identification and validation of xenobiotic metabolism-associated prognostic signature based on five genes to evaluate immune microenvironment in colon cancer. J Gastrointest Oncol, 2021, 12(6): 2788–802., DOI 10.21037/jgo-21-655 Wang K, Feng X, Zheng L, Chai Z, Yu J, You X, et al. TRPV4 is a prognostic biomarker that correlates with the immunosuppressive microenvironment and chemoresistance of anti-cancer drugs. Front Mol Biosci, 2021, 8:8690500., DOI 10. 3389/fmolb.2021.690500 Chong W, Shang L, Liu J, Fang Z, Du F, Wu H, et al. m(6 A regulator-based methylation modification patterns characterized by distinct tumor microenvironment immune profiles in colon cancer. Theranostics, 2021, 11(5): 2201–17., DOI 10.7150/thno.52717 Lu J, Wang Z, Maimaiti M, Hui W, Abudourexiti A, Gao F. Identification of diagnostic signatures in ulcerative colitis patients via bioinformatic analysis integrated with machine learning. Hum Cel, 2022, 35(1):179–88., DOI 10.1007/ s13577-021-00641-w Mou Y, Wu J, Zhang Y, Abdihamid O, Duan C, Li B. Low expression of ferritinophagy-related NCOA4 gene in relation to unfavorable outcome and defective immune cells infiltration in clear cell renal carcinoma. BMC Cancer, 2021, 21(1):18., DOI 10.1186/s12885-020-07726-z Wong M, Huang J, Lok V, Wang J, Fung F, Ding H, et al. Differences in incidence and mortality trends of colorectal cancer worldwide based on sex, age, and anatomic location. Clin Gastroenterol Hepatol, 2021, 19(5):955–66.e61., DOI 10. 1016/j.cgh.2020.02.026 Chen EX, Jonker DJ, Loree JM, Kennecke HF, Berry SR, Couture F, et al. Effect of combined immune checkpoint inhibition vs best supportive care alone in patients with advanced colorectal cancer: The Canadian cancer trials group CO.26 study. JAMA Oncol, 2020, 6(6):831–8., DOI 10.1001/jamaoncol.2020.0910 Deng J, Chen X, Zhan T, Chen M, Yan X, Huang X. CRYAB predicts clinical prognosis and is associated with immunocyte infiltration in colorectal cancer. PeerJ, 2021, 9:e12578., DOI 10.7717/peerj.12578 Plotkin LI.Molecular signaling in bone cells: Regulation of cell differentiation and survival. Adv Protein Chem Struct Biol, 2019, 116:116237–281., DOI 10.1016/bs. apcsb.2019.01.002 Zhou Q, Wang C, Zhu Y, Wu Q, Jiang Y, Huang Y, et al. Key genes and pathways controlled by E2F1 in human castration-resistant prostate cancer cells. Onco Targets Ther, 2019, 12:128961–8976., DOI 10.2147/OTT.S217347 Cselenyi CS, Lee E. Context-dependent activation or inhibition of Wnt-betacatenin signaling by Kremen. Sci Signal, 2008, 1(8):pe10., DOI 10.1126/stke.18pe10 Clines KL. DKK1 and kremen expression predicts the osteoblastic response to bone metastasis. Transl Oncol, 2018, 11(4):873–82., DOI 10.1016/j.tranon.2018. 04.013 Chen B, Wang SQ, Huang J, Xu W, Lv H, Nie C, et al. Knockdown of Kremen2 inhibits tumor growth andmigration in gastric cancer. Front Oncol, 2020, 10:10534095., DOI 10.3389/fonc.2020.534095 Xu Y, Huang Y,Wang X, Sun Y, Ye D, Chi P. Prognostic significance of lymph node yield in patients with synchronous colorectal carcinomas. Int J Colorectal Dis, 2020, 35(12):2273–82., DOI 10.1007/s00384-020-03700-0 Imamoglu GI, Oguz A, Cimen S, Eren T, Karacin C, Colak D, et al. The impact of lymph node ratio on overall survival in patients with colorectal cancer. J Cancer Res Ther, 2021, 17(4):1069–74., DOI 10.4103/jcrt.JCRT_11_19 Kim HJ. Clinical implications of lymph node metastasis in colorectal cancer: Current status and future perspectives. Ann Coloproctol, 2019, 35(3):109–17., DOI 10.3393/ac.2019.06.12 Giannone G, Ghisoni E, Genta S, Scotto G, Tuninetti V, TurinettoM, et al. Immuno-Metabolism and microenvironment in cancer: Key players for immunotherapy. Int J Mol Sci, 2020, 21(12):4414., DOI 10.3390/ijms21124414 Freeman JW. Structural biology of the tumor microenvironment. Adv Exp Med Biol, 2021, 1350:135091–100., DOI 10.1007/978-3-030-83282-7_4 Ye L, Zhang T, Kang Z, Guo G, Sun Y, Lin K, et al. Tumor-infiltrating immune cells act as a marker for prognosis in colorectal cancer. Front Immunol, 2019, 10: 102368., DOI 10.3389/fimmu.2019.02368 Zhou R, Zhang J, Zeng D, Sun H, Rong X, Shi M, et al. Immune cell infiltration as a biomarker for the diagnosis and prognosis of stage I-III colon cancer. Cancer Immunol Immunother, 2019, 68(3):433–42., DOI 10.1007/s00262-018-2289-7 Zhang N, Ning F, Guo R, Pei J, Qiao Y, Fan J, et al. Prognostic values of preoperative inflammatory and nutritional markers for colorectal cancer. Front Oncol, 2020, 10:10585083., DOI 10.3389/fonc.2020.585083 Shitara K, Nishikawa H. Regulatory T cells: A potential target in cancer immunotherapy. Ann N Y Acad Sci, 2018, 1417(1):104–15., DOI 10.1111/nyas.13625 Myers JA, Miller JS. Exploring the NK cell platform for cancer immunotherapy. Nat Rev Clin Oncol, 2021, 18(2):85–100., DOI 10.1038/s41571-020-0426-7 Imai K, Matsuyama S,Miyake S, Suga K, Nakachi K. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: An 11-year follow-up study of a general population. Lancet, 2000, 356(9244):1795–9., DOI 10.1016/S0140- 6736(00)03231-1 Katayama C, Yokobori T, Ozawa N, Suga K, Shiraishi T, Okada T, et al. Low level of stromal lectin-like oxidized LDL receptor 1 and CD8(+, cytotoxic T-lymphocytes indicate poor prognosis of colorectal cancer. Cancer Rep, Hoboken,, 2021, 4(4):e1364., DOI 10.1002/cnr2.1364 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611082 EP 1611092 DI 10.3389/pore.2023.1611082 PG 11 ER PT J TI Potential predictors of immunotherapy in small cell lung cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE tumor microenvironment; PD-L1 expression; small cell lung cancer; mutational burden; molecular subtyping ID tumor microenvironment; PD-L1 expression; small cell lung cancer; mutational burden; molecular subtyping AB Lung cancer is one of the leading causes of cancer-related deaths worldwide, with small cell lung cancer (SCLC) having the worst prognosis. SCLC is diagnosed late in the disease’s progression, limiting treatment options. The most common treatment for SCLC is chemotherapy. As the disease progresses, immunotherapy, most commonly checkpoint inhibitor medication, becomes more important. Efforts should be made in the development of immunotherapy to map specific biomarkers, which play a role in properly assigning a type of immunotherapy to the right cohort of patients, where the benefits outweigh any risks or adverse effects. The objective of this review was to provide a thorough assessment of current knowledge about the nature of the tumor process and treatment options for small cell lung cancer, with a focus on predictive biomarkers. According to the information obtained, the greatest potential, which has already been directly demonstrated in some studies, has characteristics such as tumor microenvironment composition, tumor mutation burden, and molecular subtyping of SCLC. Several other aspects appear to be promising, but more research, particularly prospective studies on a larger number of probands, is required. However, it is clear that this field of study will continue to expand, as developing a reliable method to predict immunotherapy response is a very appealing goal of current medicine and research in the field of targeted cancer therapy. CR Petruzelka L, Votruba J. Karcinom plic. Prague: Farmakon Press, 2019). 192. Skrickova J, Babickova L, Spedlova J. Postaveni chemoterapie v lecbe karcinomu plic. In: Onkologie. Olomouc: Print, 2009). 285–91. Skrickova J. Nadory plic a prudusek: Jak lecit nadory plic a prudusek? Olomouc: Solen, 2013). 28. Thai AA, Solomon BJ, Sequist LV, Gainor JF, Heist RS. Lung cancer. Lancet, 2021, 398(10299):535–54., DOI 10.1016/s0140-6736(21)00312-3 Travis WD. Pathology of lung cancer. Clin Chest Med, 2011, 32(4):669–92., DOI 10.1016/j.ccm.2011.08.005 Zheng M. Classification and pathology of lung cancer. Surg Oncol Clin N Am, 2016, 25(3):447–68., DOI 10.1016/j.soc.2016.02.003 Nicholson AG, Tsao MS, Beasley MB, Borczuk AC, Brambilla E, Cooper WA, et al. The 2021 WHO classification of lung tumors: Impact of advances since 2015. J Thorac Oncol, 2022, 17(3):362–87., DOI 10.1016/j.jtho.2021. 11.003 Sutic M, Vukic A, Baranasic J, Forsti A, Dzubur F, Samarzija M, et al. Diagnostic, predictive, and prognostic biomarkers in non-small cell lung cancer, NSCLC, management. J Pers Med, 2021, 11(11):1102., DOI 10.3390/jpm11111102 Latimer KM, Mott TF. Lung cancer: Diagnosis, treatment principles, and screening. Am Fam Physician, 2015, 91(4):250–6. Rodriguez-Canales J, Parra-Cuentas E, Wistuba, II. Diagnosis and molecular classification of lung cancer. Cancer Treat Res, 2016, 170:25–46., DOI 10.1007/978-3- 319-40389-2_2 Ryska A. Doporuceny postup pro histologicke vysetreni karcinomu plic. In: Lekarske listy. Prague: Ambit Media, 2013). 4–6. Travis WD. Classification of lung cancer. Semin Roentgenol, 2011, 46(3): 178–86., DOI 10.1053/j.ro.2011.02.003 Petersen I. The morphological and molecular diagnosis of lung cancer. Dtsch Arztebl Int, 2011, 108(31–32):525–31., DOI 10.3238/arztebl.2011.0525 Pesek M, Muzik J. Malobunecny karcinom plic: Epidemiologie, diagnostika a lecba. In: Vnitrni lekarstvi. Prague: Solen, 2018). 876–83. van Meerbeeck JP, Fennell DA, De Ruysscher DKM. Small-cell lung cancer. Lancet, 2011, 378(9804):1741–55., DOI 10.1016/s0140-6736(11)60165-7 Carter BW, Glisson BS, Truong MT, Erasmus JJ. Small cell lung carcinoma: Staging, imaging, and treatment considerations. Radiographics, 2014, 34(6): 1707–21., DOI 10.1148/rg.346140178 Rudin CM, Brambilla E, Faivre-Finn C, Sage J. Small-cell lung cancer. Nat Rev Dis Primers, 2021, 7(1):3., DOI 10.1038/s41572-020-00235-0 Dingemans A-MC, Fruh M, Ardizzoni A, Besse B, Faivre-Finn C, Hendriks LE, et al. Small-cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-upq. Ann Oncol, 2021, 32(7):839–53., DOI 10.1016/j.annonc.2021.03.207 Poirier JT, George J, Owonikoko TK, Berns A, Brambilla E, Byers LA, et al. New approaches to SCLC therapy: From the laboratory to the clinic. J Thorac Oncol, 2020, 15(4):520–40., DOI 10.1016/j.jtho.2020.01.016 Horejsi V, Bartunkova J, Brdicka T, Spisek R. Zaklady imunologie. Prague: Triton, 2017). 304. Zatloukalova P. Uloha PD-1/PD-L1 signalizace v protinadorove imunitni odpovedi. In: Klinicka onkologie. Brno: ApS BRNO, 2016). 72–7. Solomon BL, Garrido-Laguna I. Tigit: A novel immunotherapy target moving from bench to bedside. Cancer Immunol Immunother, 2018, 67(11):1659–67., DOI 10.1007/s00262-018-2246-5 Chauvin J-M, Zarour HM. TIGIT in cancer immunotherapy. J Immunother Cancer, 2020, 8(2):e000957., DOI 10.1136/jitc-2020-000957 Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ, Jr, Wu Y-L, et al. Lung cancer: Current therapies and new targeted treatments. Lancet, 2017, 389(10066):299–311., DOI 10.1016/S0140-6736(16)30958-8 Martinkova J. Farmakologie pro studenty zdravotnickych oboru. Prague: Grada Publishing, 2018). 520. American Cancer Society. Treating small cell lung cancer, 2021). Available from: https://www.cancer.org/cancer/lung-cancer/treating-small-cell.html, Accessed July 06, 2022). Ku GY, Yuan J, Page DB, Schroeder SEA, Panageas KS, Carvajal RD, et al. Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: Lymphocyte count after 2 doses correlates with survival. Cancer, 2010, 116(7):1767–75., DOI 10.1002/cncr.24951 Mahoney KM, Atkins MB. Prognostic and predictive markers for the new immunotherapies. In: Oncology. Williston Park: CMP Media, 2014). 39–48. Bianconi F, Fravolini ML, Bello-Cerezo R, Minestrini M, Scialpi M, Palumbo B. Evaluation of shape and textural features from CT as prognostic biomarkers in non-small cell lung cancer. Anticancer Res, 2018, 38(4):2155–60., DOI 10.21873/ anticanres.12456 Taniguchi H, Sen T, Rudin CM. Targeted therapies and biomarkers in small cell lung cancer. Front Oncol, 2020, 10:741., DOI 10.3389/fonc.2020.00741 Guo H, Li L, Cui J. Advances and challenges in immunotherapy of small cell lung cancer. Chin J Cancer Res, 2020, 32(1):115–28., DOI 10.21147/j.issn.1000-9604. 2020.01.13 O’Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol, 2019, 16(3):151–67., DOI 10. 1038/s41571-018-0142-8 Kalbasi A, June CH, Haas N, Vapiwala N. Radiation and immunotherapy: A synergistic combination. J Clin Invest, 2013, 123(7):2756–63., DOI 10.1172/ JCI69219 von Locquenghien M, Rozalen C, Celia-Terrassa T. Interferons in cancer immunoediting: Sculpting metastasis and immunotherapy response. J Clin Invest, 2021, 131(1):e143296., DOI 10.1172/JCI143296 Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science, 2011, 331(6024):1565–70., DOI 10.1126/science.1203486 Swann JB, Smyth MJ. Immune surveillance of tumors. J Clin Invest, 2007, 117(5):1137–46., DOI 10.1172/JCI31405 Tavakoli F, Sartakhti JS, Manshaei MH, Basanta D. Cancer immunoediting: A game theoretical approach. Silico Biol, 2021, 14(1–2):1–12., DOI 10.3233/ISB-200475 Kim R, Emi M, Tanabe K. Cancer immunoediting from immune surveillance to immune escape. Immunology, 2007, 121(1):1–14., DOI 10.1111/j.1365-2567.2007. 02587.x Wagner M, Koyasu S. Cancer immunoediting by innate lymphoid cells. Trends Immunol, 2019, 40(5):415–30., DOI 10.1016/j.it.2019.03.004 Bremnes RM, Busund L-T, Kilvaer TL, Andersen S, Richardsen E, Paulsen EE, et al. The role of tumor-infiltrating lymphocytes in development, progression, and prognosis of non-small cell lung cancer. J Thorac Oncol, 2016, 11(6):789–800., DOI 10.1016/j.jtho.2016.01.015 Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell, 2011, 144(5):646–74., DOI 10.1016/j.cell.2011.02.013 Hendry S, Salgado R, Gevaert T, Russell PA, John T, Thapa B, et al. Assessing tumor-infiltrating lymphocytes in solid tumors: A practical review for pathologists and proposal for a standardized method from the international immunooncology biomarkers working group: Part 1: Assessing the host immune response, TILs in invasive breast carcinoma and ductal carcinoma in situ, metastatic tumor deposits and areas for further research. Adv Anat Pathol, 2017, 24(5):235–51., DOI 10.1097/PAP. 0000000000000162 Dundr P, Nemejcova K, Bartu M, Matej R, Rohan Z, Ticha I. Evaluation of inflammatory cells, tumor infiltrating lymphocytes, in solid tumors. Klin Onkol, 2017, 30:10–21., DOI 10.14735/amko20173S10 Hanahan D, Coussens LM. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell, 2012, 21(3):309–22., DOI 10. 1016/j.ccr.2012.02.022 Galli F, Aguilera JV, Palermo B, Markovic SN, Nistico P, Signore A. Relevance of immune cell and tumor microenvironment imaging in the new era of immunotherapy. J Exp Clin Cancer Res, 2020, 39(1):89., DOI 10.1186/s13046- 020-01586-y Bhardwaj N. Harnessing the immune system to treat cancer. J Clin Invest, 2007, 117(5):1130–6., DOI 10.1172/JCI32136 Zhang Q, Lou Y, Bai X-L, Liang T-B. Immunometabolism: A novel perspective of liver cancer microenvironment and its influence on tumor progression. World J Gastroenterol, 2018, 24(31):3500–12., DOI 10.3748/wjg.v24. i31.3500 Goubran HA, Kotb RR, Stakiw J, Emara ME, Burnouf T. Regulation of tumor growth and metastasis: The role of tumor microenvironment. Cancer Growth Metastasis, 2014, 7:9–18., DOI 10.4137/CGM.S11285 Regan D, Dow S.Manipulation of innate immunity for cancer therapy in dogs. Vet Sci, 2015, 2(4):423–39., DOI 10.3390/vetsci2040423 Le Q-V, Yang G, Wu Y, Jang HW, ShokouhimehrM, Oh Y-K. Nanomaterials for modulating innate immune cells in cancer immunotherapy. Asian J Pharm Sci, 2019, 14(1):16–29., DOI 10.1016/j.ajps.2018.07.003 Lim JU, Kang HS. A narrative review of current and potential prognostic biomarkers for immunotherapy in small-cell lung cancer. Ann Transl Med, 2021, 9(9):809., DOI 10.21037/atm-21-68 Longo V, Catino A, Montrone M, Pizzutilo P, Annese T, Pesola F, et al. What are the biomarkers for immunotherapy in SCLC? Int J Mol Sci, 2021, 22(20):11123., DOI 10.3390/ijms222011123 Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol, 2016, 17(12):e542–51., DOI 10.1016/ s1470-2045(16)30406-5 Uruga H, Mino-Kenudson M. Predictive biomarkers for response to immune checkpoint inhibitors in lung cancer: PD-L1 and beyond. Virchows Arch, 2021, 478(1):31–44., DOI 10.1007/s00428-021-03030-8 Zhao X, Kallakury B, Chahine JJ, Hartmann D, Zhang Y, Chen Y, et al. Surgical resection of SCLC: Prognostic factors and the tumor microenvironment. J Thorac Oncol, 2019, 14(5):914–23., DOI 10.1016/j.jtho.2019.01.019 Muppa P, Parrilha Terra SBS, Sharma A,Mansfield AS, AubryM-C, Bhinge K, et al. Immune cell infiltration may be a key determinant of long-term survival in small cell lung cancer. J Thorac Oncol, 2019, 14(7):1286–95., DOI 10.1016/j.jtho. 2019.03.028 Pavan A, Attili I, Pasello G, Guarneri V, Conte PF, Bonanno L. Immunotherapy in small-cell lung cancer: From molecular promises to clinical challenges. J Immunother Cancer, 2019, 7(1):205., DOI 10.1186/s40425- 019-0690-1 Wang W, Hodkinson P, McLaren F, MacKinnon A, Wallace W, Howie S, et al. Small cell lung cancer tumour cells induce regulatory T lymphocytes, and patient survival correlates negatively with FOXP3+ cells in tumour infiltrate. Int J Cancer, 2012, 131(6):E928–37., DOI 10.1002/ijc.27613 Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity, 2009, 30(6):899–911., DOI 10.1016/j.immuni. 2009.03.019 Spigel DR, Socinski MA. Rationale for chemotherapy, immunotherapy, and checkpoint blockade in SCLC: Beyond traditional treatment approaches. J Thorac Oncol, 2013, 8(5):587–98., DOI 10.1097/JTO.0b013e318286cf88 Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol, 2010, 10(7):490–500., DOI 10.1038/nri2785 Bonanno L, Pavan A, Dieci MV, Di Liso E, Schiavon M, Comacchio G, et al. The role of immune microenvironment in small-cell lung cancer: Distribution of PD-L1 expression and prognostic role of FOXP3-positive tumour infiltrating lymphocytes. Eur J Cancer, 2018, 101:191–200., DOI 10.1016/j.ejca.2018.06.023 Hendriks LEL, Menis J, Reck M. Prospects of targeted and immune therapies in SCLC. Expert Rev Anticancer Ther, 2019, 19(2):151–67., DOI 10.1080/14737140. 2019.1559057 Wang W, Hodkinson P, McLaren F, Mackean MJ, Williams L, Howie SEM, et al. Histologic assessment of tumor-associated CD45(+, cell numbers is an independent predictor of prognosis in small cell lung cancer. Chest, 2013, 143(1):146–51., DOI 10.1378/chest.12-0681 Gelsomino F, Lamberti G, Parisi C, Casolari L, Melotti B, Sperandi F, et al. The evolving landscape of immunotherapy in small-cell lung cancer: A focus on predictive biomarkers. Cancer Treat Rev, 2019, 79(101887):101887., DOI 10.1016/ j.ctrv.2019.08.003 Calles A, Aguado G, Sandoval C, Alvarez R. The role of immunotherapy in small cell lung cancer. Clin Transl Oncol, 2019, 21(8):961–76., DOI 10.1007/s12094- 018-02011-9 Boumber Y. Tumor mutational burden, TMB, as a biomarker of response to immunotherapy in small cell lung cancer. J Thorac Dis, 2018, 10(8):4689–93., DOI 10.21037/jtd.2018.07.120 Sen T, Gay CM, Byers LA. Targeting DNA damage repair in small cell lung cancer and the biomarker landscape. Transl Lung Cancer Res, 2018, 7(1):50–68., DOI 10.21037/tlcr.2018.02.03 Fumet J-D, Truntzer C, Yarchoan M, Ghiringhelli F. Tumour mutational burden as a biomarker for immunotherapy: Current data and emerging concepts. Eur J Cancer, 2020, 131:40–50., DOI 10.1016/j.ejca.2020.02.038 Hellmann MD, Callahan MK, Awad MM, Calvo E, Ascierto PA, Atmaca A, et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell, 2018, 33(5): 853–61., DOI 10.1016/j.ccell.2018.04.001 Tay RY, Heigener D, Reck M, Califano R. Immune checkpoint blockade in small cell lung cancer. Lung Cancer, 2019, 137:31–7., DOI 10.1016/j.lungcan.2019. 08.024 Ott PA, Bang Y-J, Piha-Paul SA, Razak ARA, Bennouna J, Soria J-C, et al. T-cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol, 2019, 37(4): 318–27., DOI 10.1200/JCO.2018.78.2276 Iams WT, Porter J, Horn L. Immunotherapeutic approaches for small-cell lung cancer. Nat Rev Clin Oncol, 2020, 17(5):300–12., DOI 10.1038/s41571-019- 0316-z Galluzzi L, Chan TA, Kroemer G, Wolchok JD, Lopez-Soto A. The hallmarks of successful anticancer immunotherapy. Sci Transl Med, 2018, 10(459):eaat7807., DOI 10.1126/scitranslmed.aat7807 Sadeghi Rad H, Bazaz SR, Monkman J, Ebrahimi Warkiani M, Rezaei N, O’Byrne K, et al. The evolving landscape of predictive biomarkers in immunooncology with a focus on spatial technologies. Clin Transl Immunol, 2020, 9(11): e1215., DOI 10.1002/cti2.1215 Lei Y, Li X, Huang Q, Zheng X, Liu M. Progress and challenges of predictive biomarkers for immune checkpoint blockade. Front Oncol, 2021, 11:617335., DOI 10.3389/fonc.2021.617335 Ragavan M, Das M. Systemic therapy of extensive stage small cell lung cancer in the era of immunotherapy. Curr Treat Options Oncol, 2020, 21(8):64., DOI 10. 1007/s11864-020-00762-8 Ott PA, Elez E, Hiret S, Kim D-W, Morosky A, Saraf S, et al. Pembrolizumab in patients with extensive-stage small-cell lung cancer: Results from the phase Ib KEYNOTE-028 study. J Clin Oncol, 2017, 35(34):3823–9., DOI 10.1200/jco.2017.72. 5069 Esposito G, Palumbo G, Carillio G, Manzo A, Montanino A, Sforza V, et al. Immunotherapy in small cell lung cancer. Cancers, Basel,, 2020, 12(9):2522., DOI 10. 3390/cancers12092522 Ulrich BC, Guibert N. Non-invasive assessment of tumor PD-L1 status with circulating tumor cells. Ann Transl Med, 2018, 6:S48., DOI 10.21037/atm.2018.10.09 Reck M, Heigener D, Reinmuth N. Immunotherapy for small-cell lung cancer: Emerging evidence. Future Oncol, 2016, 12(7):931–43., DOI 10.2217/fon-2015-0012 Chae YK, Pan A, Davis AA, Mohindra N, Matsangou M, Villaflor V, et al. Recent advances and future strategies for immune-checkpoint inhibition in smallcell lung cancer. Clin Lung Cancer, 2017, 18(2):132–40., DOI 10.1016/j.cllc.2016. 07.004 Horn L, Reck M, Spigel DR. The future of immunotherapy in the treatment of small cell lung cancer. Oncologist, 2016, 21(8):910–21., DOI 10.1634/theoncologist. 2015-0523 Schwendenwein A, Megyesfalvi Z, Barany N, Valko Z, Bugyik E, Lang C, et al. Molecular profiles of small cell lung cancer subtypes: Therapeutic implications. Mol Ther Oncolytics, 2021, 20:470–83., DOI 10.1016/j.omto.2021.02.004 Zhang B, Birer SR, Dvorkin M, Shruti J, Byers L. New therapies and biomarkers: Are we ready for personalized treatment in small cell lung cancer? Am Soc Clin Oncol Educ Book, 2021, 41:1–10., DOI 10.1200/EDBK_320673 Gazdar AF, Bunn PA, Minna JD. Small-cell lung cancer: What we know, what we need to know and the path forward. Nat Rev Cancer, 2017, 17(12):725–37., DOI 10.1038/nrc.2017.87 Rudin CM, Poirier JT, Byers LA, Dive C, Dowlati A, George J, et al. Molecular subtypes of small cell lung cancer: A synthesis of human and mouse model data. Nat Rev Cancer, 2019, 19(5):289–97., DOI 10.1038/s41568-019-0133-9 Gay CM, Stewart CA, Park EM, Diao L, Groves SM, Heeke S, et al. Patterns of transcription factor programs and immune pathway activation define four major subtypes of SCLC with distinct therapeutic vulnerabilities. Cancer Cell, 2021, 39(3): 346–60.e7., DOI 10.1016/j.ccell.2020.12.014 Ruan J. Tumor cell plasticity and intrinsic immunogenicity: Implications for immunotherapy resistance in small-cell lung cancer. Thorac Cancer, 2021, 12(19): 2523–5., DOI 10.1111/1759-7714.14117 Shigematsu Y, Inamura K. Gut microbiome: A key player in cancer immunotherapy. Hepatobiliary Surg Nutr, 2018, 7(6):479–80., DOI 10.21037/ hbsn.2018.10.02 Zitvogel L, Ma Y, Raoult D, Kroemer G, Gajewski TF. The microbiome in cancer immunotherapy: Diagnostic tools and therapeutic strategies. Science, 2018, 359(6382):1366–70., DOI 10.1126/science.aar6918 Krajsova I. Importance of the immune system and immunotherapeutic options in malignant melanoma. Klin Onkol, 2015, 28:4S56–63., DOI 10.14735/ amko20154s56 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611086 EP 1611097 DI 10.3389/pore.2023.1611086 PG 12 ER PT J AU Zuo, X Wu, W Shi, P Liu, T Yu, N Li, L AF Zuo, Xuan Wu, L. Wei Shi, Peng Liu, M. Tian Yu, Na Li, Lei TI A case report of recurrent leiomyosarcoma with chondrosarcoma differentiation in the abdominal wall and a review of the literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE recurrence; case report; leiomyosarcoma; heterologous differentiation; chondrosarcoma ID recurrence; case report; leiomyosarcoma; heterologous differentiation; chondrosarcoma AB Leiomyosarcoma with heterologous differentiation is relatively rare. To date, only 19 cases have been reported in the English literature. Heterologous components frequently show histological pleomorphism, while those exhibiting well-differentiated morphology are seldom reported. Here, we report a 34-year-old female diagnosed with leiomyosarcoma and developed abdominal wall recurrence 8 years after primary surgery. The recurrent tumor mainly comprised well-differentiated chondrosarcoma except a single focus of leiomyosarcoma. Due to the rarity and prolonged onset of such a transition, our case provides insight into the understanding of this phenomenon. C1 [Zuo, Xuan] Sichuan University, West China Second University Hospital, Department of PathologyChengdu, China. [Wu, L. Wei] Sichuan University, West China Second University Hospital, Department of PathologyChengdu, China. [Shi, Peng] Sichuan University, West China Second University Hospital, Department of PathologyChengdu, China. [Liu, M. Tian] Sichuan University, West China Second University Hospital, Department of PathologyChengdu, China. [Yu, Na] Sichuan University, West China Second University Hospital, Department of PathologyChengdu, China. [Li, Lei] Sichuan University, West China Second University Hospital, Department of PathologyChengdu, China. RP Li, L (reprint author), Sichuan University, West China Second University Hospital, Department of Pathology, Chengdu, China. EM lipath@163.com CR Thiryayi SA, Turashvili G, Latta EK, Swanson D, Zhang L, Antonescu CR, et al. PLAG1-rearrangment in a uterine leiomyosarcoma with myxoid stroma and heterologous differentiation. Genes Chromosomes Cancer, 2021, 60(10):713–7., DOI 10.1002/gcc.22980 Bapat K, Brustein S. Uterine sarcoma with liposarcomatous differentiation: Report of a case and review of the literature. Int J Gynaecol Obstet, 1989, 28(1):71–5., DOI 10.1016/0020-7292(89)90547-x Damjanov I, Fan F. Chondrosarcomatous differentiation of a metastatic leiomyosarcoma of the uterus. Virchows Arch, 2006, 449(4):493–4., DOI 10.1007/ s00428-006-0283-5 Yu S, Hornick JL. "Malignant mesenchymoma" revisited: A clinicopathologic study of leiomyosarcomas with osteosarcomatous differentiation. Am J Surg Pathol, 2022, 46(10):1430–5., DOI 10.1097/PAS.0000000000001928 Parikh P, Maheshwari A, Rekhi B. Two uncommon cases of uterine leiomyosarcomas displaying heterologous osteosarcomatous de-differentiation. J Cancer Res Ther, 2015, 11(3):654., DOI 10.4103/0973-1482.140984 Cappuccini F, Tewari K, Fuller PN, Pindur A, DiSaia PJ. Aggressive clinical behavior of a rare uterine sarcoma. Gynecol Oncol, 1998, 70(1):147–51., DOI 10. 1006/gyno.1998.5020 Boyraz B, Gogakos T, Raskin KA, Nielsen GP. Metastatic uterine leiomyosarcoma with rhabdomyosarcomatous heterologous differentiation. Int J Gynecol Pathol, 2022, 42:151–4., DOI 10.1097/PGP.0000000000000874 Anh Tran T, Holloway RW. Metastatic leiomyosarcoma of the uterus with heterologous differentiation to malignant mesenchymoma. Int J Gynecol Pathol, 2012, 31(5):453–7., DOI 10.1097/PGP.0b013e318246977d Den Bakker MA, Hegt VN, Sleddens HB, Nuijten ASM, Dinjens WNM. Malignant mesenchymoma of the uterus, arising in a leiomyoma. Histopathology, 2002, 40(1):65–70., DOI 10.1046/j.1365-2559.2002.01290.x Setia A, Kanotra S, Aggarwal R, Bhavthankar DP. Epithelioid leiomyosarcoma of uterus [J]. BMJ Case Rep, 2012, 2012:bcr1120115144., DOI 10.1136/bcr.11.2011. 5144 Thway K, Jones RL, Noujaim J, Zaidi S, Miah AB, Fisher C. Dedifferentiated liposarcoma: Updates on morphology, genetics, and therapeutic strategies. Adv Anat Pathol, 2016, 23(1):30–40., DOI 10.1097/ PAP.0000000000000101 Lee ATJ, Thway K, Huang PH, Jones RL. Clinical and molecular spectrum of liposarcoma. J Clin Oncol, 2018, 36(2):151–9., DOI 10.1200/JCO.2017.74. 9598 Stout AP. Mesenchymoma, the mixed tumor of mesenchymal derivatives. Ann Surg, 1948, 127(2):278–90., DOI 10.1097/00000658- 194802000-00007 Buonomo F, Bussolaro S, Giorda G, Romano F, Biffi S, Ricci G. Cotyledonoid leiomyoma clinical characteristics, imaging features, and review of the literature. J Ultrasound Med, 2021, 40(7):1459–69., DOI 10.1002/jum.15510 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611109 EP 1611113 DI 10.3389/pore.2023.1611109 PG 5 ER PT J AU Fan, B Zheng, X Wang, Y Hu, X AF Fan, Bohan Zheng, Xin Wang, Yicun Hu, Xiaopeng TI Predicting prognosis and clinical efficacy of immune checkpoint blockade therapy via interferon-alpha response in muscle-invasive bladder cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE muscle-invasive bladder cancer; immune checkpoint blockade therapy; interferonalpha response; tumor microenvironment; prognostic biomarkers ID muscle-invasive bladder cancer; immune checkpoint blockade therapy; interferonalpha response; tumor microenvironment; prognostic biomarkers AB Background: Immune checkpoint blockade (ICB) can prompt durable and robust responses in multiple cancers, involving muscle-invasive bladder cancer (MIBC). However, only a limited fraction of patients received clinical benefit. Clarifying the determinants of response and exploring corresponding predictive biomarkers is key to improving outcomes. Methods: Four independent formerly published cohorts consisting of 641 MIBC patients were enrolled in this study. We first analyzed the associations between various cancer hallmarks and ICB therapy response in two immunotherapeutic cohorts to identify the leading prognostic hallmark in MIBC. Furthermore, advanced machine learning methods were performed to select robust and promising predictors from genes functioning in the above leading pathway. The predictive ability of selected genes was also validated in multiple MIBC cohorts. Results: We identified and verified IFNα response as the leading cancer hallmark indicating better treatment responses, favorable overall survival, and an inflamed tumor microenvironment with higher infiltration of immune effector cells in MIBC patients treated with ICB therapy. Subsequently, two commonly selected genes, CXCL10 and LAMP3, implied better therapy response and the CXCL10highLAMP3high patients would benefit more from ICB therapy, which was comprehensively validated from the perspective of gene expression, clinical response, patient survival and immune features. Conclusion: Higher IFNα response primarily predicted better ICB therapeutic responses and reflected an inflamed microenvironment in MIBC. A composite of CXCL10 and LAMP3 expression could serve as promising predictive biomarkers for ICB therapeutic responses and be beneficial for clinical decision-making in MIBC. C1 [Fan, Bohan] Capital Medical University, Beijing Chao-Yang Hospital, Department of UrologyBeijing, China. [Zheng, Xin] Northwestern University, Feinberg School of Medicine, Comprehensive Transplant CenterChicago, IL, USA. [Wang, Yicun] Capital Medical University, Beijing Chao-Yang Hospital, Department of UrologyBeijing, China. [Hu, Xiaopeng] Capital Medical University, Beijing Chao-Yang Hospital, Department of UrologyBeijing, China. RP Hu, X (reprint author), Capital Medical University, Beijing Chao-Yang Hospital, Department of Urology, Beijing, China. EM xiaopeng_hu@sina.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3): 209–49., DOI 10.3322/caac.21660 Antoni S, Ferlay J, Soerjomataram I, Znaor A, Jemal A, Bray F. Bladder cancer incidence and mortality: A global overview and recent trends. Eur Urol, 2017, 71(1):96–108., DOI 10.1016/j.eururo.2016.06.010 Liu C, Liu Z, Jin K, Zeng H, Shao F, Chang Y, et al. Integrative tumour mutation burden with Cd39 and Pd-L1 for the prediction of response to Pd-L1 blockade and adjuvant chemotherapy in muscle-invasive bladder cancer patients. Br J Cancer, 2022, 127(9):1718–25., DOI 10.1038/s41416-022-01943-y Witjes JA, Bruins HM, Cathomas R, Comperat EM, Cowan NC, Gakis G, et al. European association of urology guidelines on muscle-invasive and metastatic bladder cancer: Summary of the 2020 guidelines. Eur Urol, 2021, 79(1):82–104., DOI 10.1016/j.eururo.2020.03.055 Rijnders M, de Wit R, Boormans JL, Lolkema MPJ, van der Veldt AAM. Systematic review of immune checkpoint inhibition in urological cancers. Eur Urol, 2017, 72(3):411–23., DOI 10.1016/j.eururo.2017.06.012 Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: A single-arm, multicentre, phase 2 trial. Lancet, 2017, 389(10064):67–76., DOI 10.1016/S0140-6736(16)32455-2 Chen X, Xu R, He D, Zhang Y, Chen H, Zhu Y, et al. CD8+ T effector and immune checkpoint signatures predict prognosis and responsiveness to immunotherapy in bladder cancer. Oncogene, 2021, 40(43):6223–34., DOI 10. 1038/s41388-021-02019-6 Litchfield K, Reading JL, Puttick C, Thakkar K, Abbosh C, Bentham R, et al. Metaanalysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell, 2021, 184(3):596–614.e14., DOI 10.1016/j.cell.2021.01.002 Davis AA, Patel VG. The role of Pd-L1 expression as a predictive biomarker: An analysis of all us food and drug administration, fda, approvals of immune checkpoint inhibitors. J Immunother Cancer, 2019, 7(1):278., DOI 10.1186/s40425- 019-0768-9 Shen X, Zhao B. Efficacy of Pd-1 or Pd-L1 inhibitors and Pd-L1 expression status in cancer: Meta-analysis. BMJ, 2018, 362:k3529., DOI 10.1136/bmj.k3529 Ott PA, Bang Y-J, Piha-Paul SA, Razak ARA, Bennouna J, Soria J-C, et al. T-Cell-Inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: Keynote-028. J Clin Oncol, 2019, 37(4): 318–27., DOI 10.1200/JCO.2018.78.2276 Ivashkiv LB, Donlin LT. Regulation of type I interferon responses. Nat Rev Immunol, 2014, 14(1):36–49., DOI 10.1038/nri3581 Herzer K, Hofmann TG, Teufel A, Schimanski CC, Moehler M, Kanzler S, et al. Ifn-alpha-induced apoptosis in hepatocellular carcinoma involves promyelocytic leukemia protein and trail independently of P53. Cancer Res, 2009, 69(3):855–62., DOI 10.1158/0008-5472.CAN-08-2831 Cheon H, Borden EC, Stark GR. Interferons and their stimulated genes in the tumor microenvironment. Semin Oncol, 2014, 41(2):156–73., DOI 10.1053/j. seminoncol.2014.02.002 Davar D, Wang H, Chauvin J-M, Pagliano O, Fourcade JJ, Ka M, et al. Phase ib/ii study of pembrolizumab and pegylated-interferon alfa-2b in advanced melanoma. J Clin Oncol, 2018, 36(35):JCO1800632., DOI 10.1200/JCO.18.00632 Zhu Y, ChenM, Xu D, Li T-E, Zhang Z, Li J-H, et al. The combination of Pd-1 blockade with interferon-A has a synergistic effect on hepatocellular carcinoma. Cell Mol Immunol, 2022, 19(6):726–37., DOI 10.1038/s41423-022-00848-3 Hu B, Yu M, Ma X, Sun J, Liu C, Wang C, et al. Ifnα potentiates anti-Pd- 1 efficacy by remodeling glucose metabolism in the hepatocellular carcinoma microenvironment. Cancer Discov, 2022, 12(7):1718–41., DOI 10.1158/2159-8290. CD-21-1022 Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. Tgfβ attenuates tumour response to Pd-L1 blockade by contributing to exclusion of T cells. Nature, 2018, 554(7693):544–8., DOI 10.1038/nature25501 Rose TL, Weir WH, Mayhew GM, Shibata Y, Eulitt P, Uronis JM, et al. Fibroblast growth factor receptor 3 alterations and response to immune checkpoint inhibition in metastatic urothelial cancer: A real world experience. Br J Cancer, 2021, 125(9):1251–60., DOI 10.1038/s41416-021-01488-6 Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, et al. Signatures of T Cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med, 2018, 24(10): 1550–8., DOI 10.1038/s41591-018-0136-1 Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The molecular signatures Database, msigdb, hallmark gene set collection. Cell Syst, 2015, 1(6):417–25., DOI 10.1016/j.cels.2015.12.004 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A, 2005, 102(43): 15545–50., DOI 10.1073/pnas.0506580102 Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, Kim H, Torres- Garcia W, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun, 2013, 4:2612., DOI 10.1038/ncomms3612 Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods, 2015, 12(5):453–7., DOI 10.1038/nmeth.3337 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers differential expression analyses for rna-sequencing and microarray studies. Nucleic Acids Res, 2015, 43(7):e47., DOI 10.1093/nar/gkv007 Taylor JMG. Random survival forests. J Thorac Oncol, 2011, 6(12):1974–5., DOI 10.1097/JTO.0b013e318233d835 Murciano-Goroff YR, Warner AB, Wolchok JD. The future of cancer immunotherapy: Microenvironment-targeting combinations. Cell Res, 2020, 30(6):507–19., DOI 10.1038/s41422-020-0337-2 Marin-Acevedo JA, Chirila RM, Dronca RS. Immune checkpoint inhibitor toxicities. Mayo Clin Proc, 2019, 94(7):1321–9., DOI 10.1016/j.mayocp.2019.03.012 Liu S, Galat V, Galat Y, Lee YKA, Wainwright D, Wu J. Nk cell-based cancer immunotherapy: From basic biology to clinical development. J Hematol Oncol, 2021, 14(1):7., DOI 10.1186/s13045-020-01014-w Rahimi Kalateh Shah Mohammad G, Ghahremanloo A, Soltani A, Fathi E, Hashemy SI. Cytokines as potential combination agents with Pd-1/Pd-L1 blockade for cancer treatment. J Cel Physiol, 2020, 235(7-8):5449–60., DOI 10.1002/jcp.29491 Terawaki S, Chikuma S, Shibayama S, Hayashi T, Yoshida T, Okazaki T, et al. Ifn-A directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity. J Immunol, 2011, 186(5):2772–9., DOI 10. 4049/jimmunol.1003208 Liang Y, Tang H, Guo J, Qiu X, Yang Z, Ren Z, et al. Targeting ifnα to tumor by anti-Pd-L1 creates feedforward antitumor responses to overcome checkpoint blockade resistance. Nat Commun, 2018, 9(1):4586., DOI 10.1038/s41467-018- 06890-y Adusumilli PS, Cherkassky L, Villena-Vargas J, Colovos C, Servais E, Plotkin J, et al. Regional delivery of mesothelin-targeted car T cell therapy generates potent and long-lasting Cd4-dependent tumor immunity. Sci Transl Med, 2014, 6(261): 261ra151., DOI 10.1126/scitranslmed.3010162 Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, Collins MA, et al. Decade-long leukaemia remissions with persistence of CD4+ CAR T cells. Nature, 2022, 602(7897):503–9., DOI 10.1038/s41586-021-04390-6 Peng P, Lou Y, Wang S, Wang J, Zhang Z, Du P, et al. Activated nk cells reprogram mdscs via nkg2d-nkg2dl and ifn-Γ to modulate antitumor T-cell response after cryo-thermal therapy. J Immunother Cancer, 2022, 10(12): e005769., DOI 10.1136/jitc-2022-005769 Ilander M, Kreutzman A, Mustjoki S. Ifnα induces prolonged remissions modeling curative immunologic responses in chronic myeloid leukemia. Oncoimmunology, 2014, 3:e28781., DOI 10.4161/onci.28781 Zhao H,Wu L, Yan G, Chen Y, ZhouM,Wu Y, et al. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduct Target Ther, 2021, 6(1):263., DOI 10.1038/s41392-021-00658-5 Reschke R, Yu J, Flood B, Higgs EF, Hatogai K, Gajewski TF. Immune cell and tumor cell-derived Cxcl10 is indicative of immunotherapy response in metastatic melanoma. J Immunother Cancer, 2021, 9(9):e003521., DOI 10.1136/jitc-2021- 003521 Chen P-L, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov, 2016, 6(8):827–37., DOI 10.1158/2159-8290.CD-15-1545 Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, et al. Chemokine expression in melanoma metastases associated with Cd8+ T-cell recruitment. Cancer Res, 2009, 69(7):3077–85., DOI 10.1158/0008-5472.CAN-08- 2281 Spranger S, Dai D, Horton B, Gajewski TF. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell, 2017, 31(5):711–23.e4., DOI 10.1016/j.ccell.2017.04.003 Chow MT, Ozga AJ, Servis RL, Frederick DT, Lo JA, Fisher DE, et al. Intratumoral activity of the Cxcr3 chemokine system is required for the efficacy of anti-Pd-1 therapy. Immunity, 2019, 50(6):1498–512.e5., DOI 10.1016/j.immuni. 2019.04.010 Saviano A, Henderson NC, Baumert TF. Single-cell genomics and spatial transcriptomics: Discovery of novel cell states and cellular interactions in liver physiology and disease biology. J Hepatol, 2020, 73(5):1219–30., DOI 10.1016/j.jhep. 2020.06.004 Hui Z, Zhang J, Ren Y, Li X, Yan C, YuW, et al. Single-cell profiling of immune cells after neoadjuvant pembrolizumab and chemotherapy in iiia non-small cell lung cancer, nsclc). Cell Death Dis, 2022, 13(7):607., DOI 10.1038/s41419-022-05057-4 Hoffmann C, Noel F, Grandclaudon M, Massenet-Regad L, Michea P, Sirven P, et al. Pd-L1 and icosl discriminate human secretory and helper dendritic cells in cancer, allergy and autoimmunity. Nat Commun, 2022, 13(1):1983., DOI 10.1038/ s41467-022-29516-w NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611117 EP 1611127 DI 10.3389/pore.2023.1611117 PG 11 ER PT J AU Revesz, M Oberna, F Slezak, A Ferenczi, Kenessey, I Takacsi-Nagy, Z AF Revesz, Monika Oberna, Ferenc Slezak, Andras Ferenczi, Ors Kenessey, Istvan Takacsi-Nagy, Zoltan TI The characteristics of head and neck squamous cell cancer in young adults: A retrospective single-center study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE survival; risk factors; prognostic factors; head and neck cancer; young adults ID survival; risk factors; prognostic factors; head and neck cancer; young adults AB We aimed to characterize clinical and prognostical factors of primary head and neck squamous cell carcinoma (HNSCC) in 85 young patients (≤39 years, median age: 37 years; between 2000–2018) in comparison with 140 institutional general HNSCC patients (median age: 61.5 years). The patient’s medical records were collected from the institutional database. The prevalence of smoking and alcohol consumption (65.8% and 48.1%) in the young group exceeded the regional population average but was below the institutional (86.4% and 55%) general HNSCC patient population. Primary tumor sites in the group of young patients were as follows: oral cavity (56.4%), oropharynx (17.6%), hypopharynx (11.7%), and larynx (14.1%). Cumulative fiveyear overall survival was 44.2% in the young group, but significantly better with early T (T1-2 vs. T3-4: 52.6% vs. 26.7%; p = 0.0058) and N0 status (N0 vs. N+: 65.2% vs. 32.3%; p = 0.0013). Young age, abstinence, earlier stage and laryngeal tumor site might predict a better prognosis. The age distribution and the high prevalence of traditional risk factors among the young patients as well as the predominance of oral cavity tumor localization suggest that the early onset of tumor development could be originated from the premature failure of the intrinsic protective mechanisms. C1 [Revesz, Monika] National Institute of Oncology, Multidisciplinary Center of Head and Neck OncologyBudapest, Hungary. [Oberna, Ferenc] National Institute of Oncology, Multidisciplinary Center of Head and Neck OncologyBudapest, Hungary. [Slezak, Andras] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Ferenczi, Ors] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. [Kenessey, Istvan] National Institute of Oncology, National Cancer RegistryBudapest, Hungary. [Takacsi-Nagy, Zoltan] National Institute of Oncology, Center of RadiotherapyBudapest, Hungary. RP Takacsi-Nagy, Z (reprint author), National Institute of Oncology, Center of Radiotherapy, Budapest, Hungary. EM takacsi.zoltan@oncol.hu CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 World Health Organization. Cancer today, 2022). Available at: http://gco.iarc. fr/today/home, Accessed December 15, 2022). Mafi N, Kadivar M, Hosseini N, Ahmadi S, Zare-Mirzaie A. Head and neck squamous cell carcinoma in Iranian patients and risk factors in young adults: A fifteenyear study. Asian Pac J Cancer Prev, 2012, 13:3373–8., DOI 10.7314/apjcp.2012.13.7.3373 Llewellyn CD, Johnson NW, Warnakulasuriya KA. Risk factors for squamous cell carcinoma of the oral cavity in young people--a comprehensive literature review. Oral Oncol, 2001, 37:401–18., DOI 10.1016/s1368-8375(00)00135-4 Gamez ME, Kraus R, Hinni ML, Moore EJ, Ma DJ, Ko SJ, et al. Treatment outcomes of squamous cell carcinoma of the oral cavity in young adults. Oral Oncol, 2018, 87:43–8., DOI 10.1016/j.oraloncology.2018.10.014 van Monsjou HS, Wreesmann VB, van den Brekel MWM, Balm AJM. Head and neck squamous cell carcinoma in young patients. Oral Oncol, 2013, 49: 1097–102., DOI 10.1016/j.oraloncology.2013.09.001 Turi K, Barabas P, Csurgay K, Lehner GY, Lorincz A, Nemeth ZS. An analysis of the epidemiological and etiological factors of oral tumors of young adults in a Central-Eastern European population. Pathol Oncol Res, 2013, 19:353–63., DOI 10. 1007/s12253-013-9628-y Majchrzak E, Szybiak B, Wegner A, Pienkowski P, Pazdrowski J, Luczewski L, et al. Oral cavity and oropharyngeal squamous cell carcinoma in young adults: A review of the literature. Radiol Oncol, 2014, 48:1–10., DOI 10.2478/raon-2013-0057 Lacy PD, Piccirillo JF, Merritt MG, Zequeira MR. Head and neck squamous cell carcinoma: Better to be young. Otolaryngol–head Neck Surg Off J Am Acad Otolaryngol-head Neck Surg, 2000, 122:253–8., DOI 10.1016/S0194-5998(00, 70249-X Nemeth Z, Turi K, Lehner G, Veres SD, Csurgay K. The prognostic role of age in oral cancer. A clinical study. Magy Onkol, 2013, 57:166–72. Coccia PF, Pappo AS, Beaupin L, Borges VF, Borinstein SC, Chugh R, et al. Adolescent and young adult oncology, version 2.2018, NCCN clinical practice guidelines in oncology. JNCCN, 2018, 16:66–97. Allen PW. ICDO—international classification of diseases for Oncology. Pathology, Phila,, 1991, 23:280., DOI 10.1016/S0031-3025(16)36112-8 World Health Organization. International guide for monitoring alcohol consumption and related harm, 2000). Available at: https://apps.who.int/iris/ handle/10665/66529, Accessed June 17, 2012). Sobin LH, Gospodarowicz MK,Wittekind C. TNM classification of malignant tumours. 7th ed. Wiley-Blackwell, 2010). Du E, Mazul AL, Farquhar D, Brennan P, Anantharaman D, Abedi-Ardekani B, et al. Long-term survival in head and neck cancer: Impact of site, stage, smoking, and human papillomavirus status. The Laryngoscope, 2019, 129:2506–13., DOI 10. 1002/lary.27807 Cadoni G, Giraldi L, Petrelli L, Pandolfini M, Giuliani M, Paludetti G, et al. Prognostic factors in head and neck cancer: A 10-year retrospective analysis in a single-institution in Italy. Acta Otorhinolaryngol Ital Organo Uff Della Soc Ital Otorinolaringol E Chir Cerv-facc, 2017, 37:458–66., DOI 10.14639/ 0392-100X-1246 Toner M, O’Regan EM. Head and neck squamous cell carcinoma in the young: A spectrum or a distinct group? Part 1. Head Neck Pathol, 2009, 3:246–8., DOI 10. 1007/s12105-009-0135-0 Pickering CR, Zhang J, Neskey DM, Zhao M, Jasser SA, Wang J, et al. Squamous cell carcinoma of the oral tongue in young non-smokers is genomically similar to tumors in older smokers. Clin Cancer Res, 2014, 20:3842–8., DOI 10.1158/ 1078-0432.CCR-14-0565 Ritchie H, Roser M. Smoking. Our World data, 2013). Available at: https:// ourworldindata.org/smoking, Accessed August 5, 2021). Ritchie H, Roser M. Alcohol consumption. Our World Data, 2018). Available at: https://ourworldindata.org/alcohol-consumption, Accessed August 5, 2021). Csurgay K, Zalatnai A, BenczikM, Csomo BK, Horvath F, Lorincz A, et al. A study of prognostic factors in young patients with non-HPV oral cancer in central Europe. Pathol Oncol Res, 2021, 27:1609991., DOI 10.3389/pore.2021. 1609991 Wang L-E, Hu Z, Sturgis EM, Spitz MR, Strom SS, Amos CI, et al. Reduced DNA repair capacity for removing tobacco carcinogen-induced DNA adducts contributes to risk of head and neck cancer but not tumor characteristics. Clin Cancer Res, 2010, 16:764–74., DOI 10.1158/1078-0432. ccr-09-2156 Kostrzewska-Poczekaj M, Gawecki W, Illmer J, Rydzanicz M, Gajecka M, Szyfter W, et al. Polymorphisms of DNA repair genes and risk of squamous cell carcinoma of the head and neck in young adults. Eur Arch Oto-Rhino-Laryngol Off J Eur Fed Oto-Rhino-Laryngol Soc EUFOS Affil Ger Soc Oto-Rhino-Laryngol - Head Neck Surg, 2013, 270:271–6., DOI 10.1007/s00405-012-1993-8 Wei Q, Cheng L, Amos CI, Wang LE, Guo Z, Hong WK, et al. Repair of tobacco carcinogen-induced DNA adducts and lung cancer risk: A molecular epidemiologic study. J Natl Cancer Inst, 2000, 92:1764–72., DOI 10.1093/jnci/92. 21.1764 Landi S, Gemignani F, Canzian F, Gaborieau V, Barale R, Landi D, et al. DNA repair and cell cycle control genes and the risk of young-onset lung cancer. Cancer Res, 2006, 66:11062–9., DOI 10.1158/0008-5472.CAN-06-1039 Gardner IH, Siddharthan R, Watson K, Dewey E, Ruhl R, Khou S, et al. A distinct innate immune signature of early onset colorectal cancer. ImmunoHorizons, 2021, 5:489–99., DOI 10.4049/immunohorizons.2000092 Lee SU, Moon SH, Choi SW, Cho KH, Park JY, Jung Y-S, et al. Prognostic significance of smoking and alcohol history in young age oral cavity cancer. Oral Dis, 2020, 26:1440–8., DOI 10.1111/odi.13432 Vankos JB, Piurko V, Suba Z, Nemeth Z, Timar J, Kenessey I. The prognostic role of expression of p16 tumor suppressor gene in Hungarian patients with oral squamous cell carcinoma. Magy Onkol, 2015, 59:352–9. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611123 EP 1611130 DI 10.3389/pore.2023.1611123 PG 8 ER PT J AU Khonrak, Th Watcharadetwittaya, S Chamgramol, Y Intarawichian, P Deenonpoe, R AF Khonrak, Thitima Watcharadetwittaya, Sasithorn Chamgramol, Yaovalux Intarawichian, Piyapharom Deenonpoe, Raksawan TI RET rearrangements are relevant to histopathologic subtypes and clinicopathological features in Thai papillary thyroid carcinoma patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE papillary thyroid carcinoma; RET rearrangements; CCDC6::RET rearrangement; NCOA4::RET rearrangement; gene rearrangements ID papillary thyroid carcinoma; RET rearrangements; CCDC6::RET rearrangement; NCOA4::RET rearrangement; gene rearrangements AB Background: Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer. The RET gene rearrangements CCDC6::RET and NCOA4::RET are the most common RET gene rearrangements in PTC patients. Different RET::PTC rearrangements are associated with different PTC phenotypes. Methods: Eighty-three formalin-fixed paraffin-embedded (FFPE) PTC samples were examined. The prevalence and expression levels of CCDC6::RET and NCOA4::RET were determined using semi-quantitative polymerase chain reaction (qRT-PCR). The association of these rearrangements with clinicopathological data was investigated. Results: The presence of CCDC6::RET rearrangement was significantly associated with the classic subtype and absence of angio/lymphatic invasion (p < 0.05). While NCOA4::RET was associated with the tall-cell subtype, and presence of angio/ lymphatic invasion and lymph node metastasis (p < 0.05). Multivariate analysis demonstrated that an absence of extrathyroidal extension and extranodal extension were independent predictive factors for CCDC6::RET, whereas the tall-cell subtype, large tumor size, angioinvasion, lymphatic invasion and perineural invasion were independent predictive factors for NCOA4::RET (p < 0.05). However, the mRNA expression level of CCDC6::RET and of NCOA4:: RET were not significantly associated with clinicopathological data. Conclusion: CCDC6::RET was correlated with an innocent PTC subtype and characteristics, but NCOA4::RET correlated with an aggressive phenotype of PTC. Therefore, these RET rearrangements strongly associated with clinicopathological phenotypes and can be used as predictive markers in PTC patients. C1 [Khonrak, Thitima] Khon Kaen University, Faculty of Medicine, Department of PathologyKhon Kaen, Thailand. [Watcharadetwittaya, Sasithorn] Khon Kaen University, Faculty of Medicine, Department of PathologyKhon Kaen, Thailand. [Chamgramol, Yaovalux] Khon Kaen University, Faculty of Medicine, Department of PathologyKhon Kaen, Thailand. [Intarawichian, Piyapharom] Khon Kaen University, Faculty of Medicine, Department of PathologyKhon Kaen, Thailand. [Deenonpoe, Raksawan] Khon Kaen University, Faculty of Medicine, Department of PathologyKhon Kaen, Thailand. RP Deenonpoe, R (reprint author), Khon Kaen University, Faculty of Medicine, Department of Pathology, Khon Kaen, Thailand. EM raksde@kku.ac.th CR Fagin JA, Wells SA, Jr. Biologic and clinical perspectives on thyroid cancer. N Engl J Med, 2016, 375(11):1054–67., DOI 10.1056/NEJMra1501993 Liu RT, Chou FF,Wang CH, Lin CL, Chao FP, Chung JC, et al. Low prevalence of RET rearrangements, RET/PTC1, RET/PTC2, RET/PTC3, and ELKS-RET, in sporadic papillary thyroid carcinomas in taiwan Chinese. Thyroid, 2005, 15(4): 326–35., DOI 10.1089/thy.2005.15.326 Fenton CL, Lukes Y, NicholsonD,DinauerCA, Francis GL, Tuttle RM. The ret/PTC mutations are common in sporadic papillary thyroid carcinoma of children and young adults. J Clin Endocrinol Metab, 2000, 85(3):1170–5., DOI 10.1210/jcem.85.3.6472 Guerra A, Rosaria Sapio M, Marotta V, Campanile E, Ilaria Moretti M, Deandrea M, et al. Prevalence of RET/PTC rearrangement in benign and malignant thyroid nodules and its clinical application. Endocr J, 2011, 58(1): 31–8., DOI 10.1507/endocrj.k10e-260 Liu S, Gao A, Zhang B, Zhang Z, Zhao Y, Chen P, et al. Assessment ofmolecular testing in fine-needle aspiration biopsy samples: An experience in a Chinese population. Exp Mol Pathol, 2014, 97(2):292–7., DOI 10.1016/j.yexmp.2014.08.005 Staubitz JI, Musholt TJ, Schad A, Springer E, Lang H, Rajalingam K, et al. ANKRD26-RET - a novel gene fusion involving RET in papillary thyroid carcinoma. Cancer Genet, 2019, 238:10–7., DOI 10.1016/j.cancergen.2019.07.002 Santoro M, Carlomagno F. Central role of RET in thyroid cancer. Cold Spring Harb Perspect Biol, 2013, 5(12):a009233., DOI 10.1101/cshperspect.a009233 Nikiforov YE, Rowland JM, Bove KE, Monforte-Munoz H, Fagin JA. Distinct pattern of ret oncogene rearrangements in morphological variants of radiationinduced and sporadic thyroid papillary carcinomas in children. Cancer Res, 1997, 57(9):1690–4. Rabes HM, Demidchik EP, Sidorow JD, Lengfelder E, Beimfohr C, Hoelzel D, et al. Pattern of radiation-induced RET and NTRK1 rearrangements in 191 postchernobyl papillary thyroid carcinomas: Biological, phenotypic, and clinical implications. Clin Cancer Res, 2000, 6(3):1093–103. Smida J, Salassidis K, Hieber L, Zitzelsberger H, Kellerer AM, Demidchik EP, et al. Distinct frequency of ret rearrangements in papillary thyroid carcinomas of children and adults from Belarus. Int J Cancer, 1999, 80(1):32–8., DOI 10.1002/(sici, 1097-0215(19990105)80:1<32:aid-ijc7>3.0.co;2-l Powell DJ, Russell J, Nibu K, Li G, Rhee E, Liao M, et al. The RET/ PTC3 oncogene: Metastatic solid-type papillary carcinomas in murine thyroids. Cancer Res, 1998, 58(23):5523–8. Sugg SL, Ezzat S, Rosen IB, Freeman JL, Asa SL. Distinct multiple RET/PTC gene rearrangements in multifocal papillary thyroid neoplasia. J Clin Endocrinol Metab, 1998, 83(11):4116–22., DOI 10.1210/jcem.83.11.5271 Thomas GA, Bunnell H, Cook HA, Williams ED, Nerovnya A, Cherstvoy ED, et al. High prevalence of RET/PTC rearrangements in Ukrainian and belarussian post-chernobyl thyroid papillary carcinomas: A strong correlation between RETPTC3 and the solid-follicular variant. J Clin Endocrinol Metab, 1999, 84(11):4232–8., DOI 10.1210/jcem.84.11.6129 Basolo F, Giannini R, Monaco C, Melillo RM, Carlomagno F, Pancrazi M, et al. Potent mitogenicity of the RET/PTC3 oncogene correlates with its prevalence in tall-cell variant of papillary thyroid carcinoma. Am J Pathol, 2002, 160(1): 247–54., DOI 10.1016/S0002-9440(10)64368-4 Abdullah MI, Junit SM, Ng KL, Jayapalan JJ, Karikalan B, Hashim OH. Papillary thyroid cancer: Genetic alterations and molecular biomarker investigations. Int J Med Sci, 2019, 16(3):450–60., DOI 10.7150/ijms.29935 Lee CH, Hsu LS, Chi CW, Chen GD, Yang AH, Chen JY. High frequency of rearrangement of the RET protooncogene, RET/PTC, in Chinese papillary thyroid carcinomas. J Clin Endocrinol Metab, 1998, 83(5):1629–32., DOI 10.1210/jcem.83.5. 4774 Chua EL, Wu WM, Tran KT, Mccarthy SW, Lauer CS, Dubourdieu D, et al. Prevalence and distribution of RET/PTC 1, 2, and 3 in papillary thyroid carcinoma in New Caledonia and Australia. J Clin Endocrinol Metab, 2000, 85(8):2733–9., DOI 10.1210/jcem.85.8.6722 Khan MS, Qadri Q, Makhdoomi MJ, Wani MA, Malik AA, Niyaz M, et al. RET/PTC gene rearrangements in thyroid carcinogenesis: Assessment and clinicopathological correlations. Pathol Oncol Res, 2020, 26(1):507–13., DOI 10.1007/ s12253-018-0540-3 Chung JH, Hahm JR, Min YK, Lee MS, Lee MK, Kim KW, et al. Detection of RET/PTC oncogene rearrangements in Korean papillary thyroid carcinomas. Thyroid, 1999, 9(12):1237–43., DOI 10.1089/thy.1999.9.1237 Zhu Z, Ciampi R, Nikiforova MN, Gandhi M, Nikiforov YE. Prevalence of RET/PTC rearrangements in thyroid papillary carcinomas: Effects of the detection methods and genetic heterogeneity. J Clin Endocrinol Metab, 2006, 91(9):3603–10., DOI 10.1210/jc.2006-1006 Rhoden KJ, Johnson C, Brandao G, Howe JG, Smith BR, Tallini G. Real-time quantitative RT-PCR identifies distinct c-RET, RET/PTC1 and RET/PTC3 expression patterns in papillary thyroid carcinoma. Lab Invest, 2004, 84(12): 1557–70., DOI 10.1038/labinvest.3700198 Musholt TJ, Staubitz JI, Antonio Camara RJ, Musholt PB, Humberg D, Springer E, et al. Detection of RET rearrangements in papillary thyroid carcinoma using RT-PCR and FISH techniques - a molecular and clinical analysis. Eur J Surg Oncol, 2019, 45(6):1018–24., DOI 10.1016/j.ejso.2018.11.009 Naito H, Pairojkul C, Kitahori Y, Yane K, Miyahara H, Konishi N, et al. Different ras gene mutational frequencies in thyroid papillary carcinomas in Japan and Thailand. Cancer Lett, 1998, 131(2):171–5., DOI 10.1016/s0304-3835(98, 00149-9 Sherman SI, Clary DO, Elisei R, Schlumberger MJ, Cohen EW, Schoffski P, et al. Correlative analyses of RET and RAS mutations in a phase 3 trial of cabozantinib in patients with progressive, metastatic medullary thyroid cancer. Cancer, 2016, 122(24):3856–64., DOI 10.1002/cncr.30252 Melillo RM, Castellone MD, Guarino V, Falco VD, Cirafici AM, Salvatore G, et al. The RET/PTC-RAS-BRAF linear signaling cascade mediates the motile and mitogenic phenotype of thyroid cancer cells. J Clin Invest, 2005, 115(4):1068–81., DOI 10.1172/JCI22758 Romei C, Elisei R. RET/PTC translocations and clinico-pathological features in human papillary thyroid carcinoma. Front Endocrinol, 2012, 3:54., DOI 10.3389/ fendo.2012.00054 Gandhi M, Dillon LW, Pramanik S, Nikiforov YE, Wang YH. DNA breaks at fragile sites generate oncogenic RET/PTC rearrangements in human thyroid cells. Oncogene, 2010, 29(15):2272–80., DOI 10.1038/onc. 2009.502 Gandhi M, Evdokimova V, Nikiforov YE. Mechanisms of chromosomal rearrangements in solid tumors: The model of papillary thyroid carcinoma. Mol Cel Endocrinol, 2010, 321(1):36–43., DOI 10.1016/j.mce.2009.09.013 Adeniran AJ, Zhu Z, Gandhi M, Steward DL, Fidler JP, Giordano TJ, et al. Correlation between genetic alterations and microscopic features, clinical manifestations, and prognostic characteristics of thyroid papillary carcinomas. Am J Surg Pathol, 2006, 30(2):216–22., DOI 10.1097/01.pas. 0000176432.73455.1b Mochizuki K, Kondo T, Nakazawa T, Iwashina M, Kawasaki T, Nakamura N, et al. RET rearrangements and BRAF mutation in undifferentiated thyroid carcinomas having papillary carcinoma components: Genetic background of composite UTC. Histopathology, 2010, 57(3):444–50., DOI 10.1111/j.1365-2559. 2010.03646.x Galuppini F, Vianello F, Censi S, Barollo S, Bertazza L, Carducci S, et al. Differentiated thyroid carcinoma in pediatric age: Genetic and clinical scenario. Front Endocrinol, 2019, 10:552., DOI 10.3389/fendo.2019.00552 Rogounovitch TI, Mankovskaya SV, Fridman MV, Leonova TA, Kondratovitch VA, Konoplya NE, et al. Major oncogenic drivers and their clinicopathological correlations in sporadic childhood papillary thyroid carcinoma in Belarus. Cancers, 2021, 13(13):3374., DOI 10.3390/cancers13133374 Tallini G, Santoro M, Helie M, Carlomagno F, Salvatore G, Chiappetta G, et al. RET/PTC oncogene activation defines a subset of papillary thyroid carcinomas lacking evidence of progression to poorly differentiated or undifferentiated tumor phenotypes. Clin Cancer Res Off J Am Assoc Cancer Res, 1998, 4(2):287–94. Martinez JRW, Vargas-Salas S, Gamboa SU, Munoz E, Dominguez JM, Leon A, et al. The combination of RET, BRAF and demographic data identifies subsets of patients with aggressive papillary thyroid cancer. Horm Cancer, 2019, 10(2–3): 97–106., DOI 10.1007/s12672-019-0359-8 Fugazzola L, Pilotti S, Pinchera A, Vorontsova TV, Mondellini P, Bongarzone I, et al. Oncogenic rearrangements of the RET proto-oncogene in papillary thyroid carcinomas from children exposed to the chernobyl nuclear accident. Cancer Res, 1995, 55(23):5617–20. Baloch ZW, Asa SL, Barletta JA, Ghossein RA, Juhlin CC, Jung CK, et al. Overview of the 2022 WHO classification of thyroid neoplasms. Endocr Pathol, 2022, 33(1):27–63., DOI 10.1007/s12022-022-09707-3 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611138 EP 1611150 DI 10.3389/pore.2023.1611138 PG 13 ER PT J AU Han, W Wang, B Yong, X Zhang, Y Shao, M Wang, Ch AF Han, Wen Wang, Bei Yong, Xiang Zhang, Yi Shao, Mingyu Wang, Chun TI Indolent T-lymphoblastic proliferation with fibrolamellar hepatocellular carcinoma developed after colorectal adenocarcinoma: a case report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE differential diagnosis; indolent T-lymphoblastic proliferation; iT-LBP; fibrolamellar hepatocellular carcinoma; T-lymphoblastic lymphoma ID differential diagnosis; indolent T-lymphoblastic proliferation; iT-LBP; fibrolamellar hepatocellular carcinoma; T-lymphoblastic lymphoma AB Objective: Indolent T-lymphoblastic proliferation (iT-LBP) is a non-neoplastic disease with an indolent clinical course, manifesting as hyperplasia of immature extrathymic T-lymphoblastic cells. Isolated iT-LBP has been observed, but the majority of iT-LBP cases has been seen in conjunction with other diseases. iTLBP is easily misdiagnosed as T-lymphoblastic lymphoma/leukemia, and understanding the disease of indolent T-lymphoblastic proliferation may prevent misdiagnosis and missed diagnosis in pathological diagnosis. Case presentation: We report a case morphology, immunophenotypic, and molecular features of iT-LBP combined with fibrolamellar hepatocellular carcinoma developed after colorectal adenocarcinoma and review relevant literature. Conclusion: iT-LBP combined with fibrolamellar hepatocellular carcinoma developed after colorectal adenocarcinoma is relatively rare and should always be considered as a differential diagnosis of T-lymphoblastic lymphoma and scirrhous hepatocellular carcinoma as the two disorders show highly similar clinical features. C1 [Han, Wen] People’s Hospital of Xinjiang Uygur Autonomous Region, Department of PathologyUrumqi, China. [Wang, Bei] People’s Hospital of Xinjiang Uygur Autonomous Region, Graduate Education DepartmentUrumqi, China. [Yong, Xiang] Anhui Wanbei Coal-Electricity Group General Hospital, Department of PathologySuzhou, China. [Zhang, Yi] People’s Hospital of Xinjiang Uygur Autonomous Region, Department of PathologyUrumqi, China. [Shao, Mingyu] People’s Hospital of Xinjiang Uygur Autonomous Region, Department of PathologyUrumqi, China. [Wang, Chun] People’s Hospital of Xinjiang Uygur Autonomous Region, Department of PathologyUrumqi, China. RP Wang, Ch (reprint author), People’s Hospital of Xinjiang Uygur Autonomous Region, Department of Pathology, Urumqi, China. EM hanwen-2@126.com CR Alaggio R, Amador C, Anagnostopoulos I, Attygalle AD, Araujo IBO, Berti E, et al. The 5th edition of the World Health organization classification of haematolymphoid tumours: Lymphoid neoplasms. Leukemia, 2022, 36(7): 1720–48., DOI 10.1038/s41375-022-01620-2 Yang F, Liu T, Zhao H, Hu Z, Xiao L, Liu Y, et al. Indolent T-lymphblastic proliferation: report of a case involving the upper aerodigestive tract. Int J Clin Exp Pathol, 2014, 7(9):6350–6. Ohgami RS, Zhao S, Ohgami JK, Leavitt MO, Zehnder JL, West RB, et al. TdT+ T-lymphoblastic populations are increased in Castleman disease, in Castleman disease in association with follicular dendritic cell tumors, and in angioimmunoblastic T-cell lymphoma. Am J Surg Pathol, 2012, 36(11):1619–28., DOI 10.1097/PAS.0b013e318264e223 Strauchen JA. Indolent T-lymphoblastic proliferation: Report of a case with an 11-year history and association with myasthenia gravis. Am J Surg Pathol, 2001, 25(3):411–5., DOI 10.1097/00000478-200103000-00018 Wang ZM, Xiao WB, Zheng SS, Sun K, Wang LJ. Hepatocellular carcinoma with indolent T-lymphoblastic proliferation. Leuk Lymphoma, 2006, 47(11): 2424–6., DOI 10.1080/10428190600822151 Woo CG, Huh J. TdT+ T-lymphoblastic proliferation in castleman disease. J Pathol Transl Med, 2015, 49(1):1–4., DOI 10.4132/jptm.2014.11.17 Karki NR, Arfa AS, Savage N, Kutlar A. Indolent T-lymphoblastic proliferation in idiopathic multicentric castleman disease. Acta Haematol, 2022, 145(2):214–20., DOI 10.1159/000520240 Yasuda H, TsutsuiM, Ota Y, Tanaka M, Komatsu N. Indolent T-lymphoblastic proliferation concomitant with acinic cell carcinoma mimicking T-lymphoblastic lymphoma: Case report and literature review. Histopathology, 2018, 72(5):862–6., DOI 10.1111/his.13433 Meyers RL. Tumors of the liver in children. Surg Oncol, 2007, 16(3):195–203., DOI 10.1016/j.suronc.2007.07.002 Samdanci ET, Akatli AN, Soylu NK. Clinicopathological features of two extremely rare hepatocellular carcinoma variants: A brief review of fibrolamellar and scirrhous hepatocellular carcinoma. J Gastrointest Cancer, 2020, 51(4): 1187–92., DOI 10.1007/s12029-020-00500-1 Kakar S, Burgart LJ, Batts KP, Garcia J, Jain D, Ferrell LD. Clinicopathologic features and survival in fibrolamellar carcinoma: Comparison with conventional hepatocellular carcinoma with and without cirrhosis. Mod Pathol, 2005, 18(11): 1417–23., DOI 10.1038/modpathol.3800449 Lalazar G, Simon SM. Fibrolamellar carcinoma: Recent advances and unresolved questions on the molecular mechanisms. Semin Liver Dis, 2018, 38(1):51–9., DOI 10.1055/s-0037-1621710 Eun S, Jeon YK, Jang JJ. Hepatocellular carcinoma with immature T-cell, T-lymphoblastic, proliferation. J Korean Med Sci, 2010, 25(2):309–12., DOI 10. 3346/jkms.2010.25.2.309 Ohgami RS, Arber DA, Zehnder JL, Natkunam Y, Warnke RA. Indolent T-lymphoblastic proliferation, iT-LBP): A review of clinical and pathologic features and distinction from malignant T-lymphoblastic lymphoma. Adv Anat Pathol, 2013, 20(3):137–40., DOI 10.1097/PAP.0b013e31828d17ec Kong M,Wang Z, Teng X, Xu L, Liu J, Jiang C, et al. Hepatic carcinoma with indolent T-lymphoblastic proliferation, iT-LBP). Int J Clin Exp Pathol, 2018, 11(3): 1674–8. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611151 EP 1611156 DI 10.3389/pore.2023.1611151 PG 6 ER PT J AU Korpas, LK Beke, L Varga, D Bidiga, L Mehes, G Molnar, S AF Korpas, Levente Kristof Beke, Livia Varga, Daniel Bidiga, Laszlo Mehes, Gabor Molnar, Sarolta TI Grade Group accuracy is improved by extensive prostate biopsy sampling, but unrelated to prostatectomy specimen sampling or use of immunohistochemistry SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prostate cancer; needle biopsy; prostatectomy; grade group; pathology ID prostate cancer; needle biopsy; prostatectomy; grade group; pathology AB Assessing the accurate Grade Group of a prostate needle biopsy specimen is essential for choosing the adequate therapeutic modality for prostate cancer patients. However, it is well-known that biopsy Grade Group tends to up- or downgrade significantly at radical prostatectomy. We aimed to investigate the correlation between accuracy and biopsy core number, performed immunohistochemical staining (IHC) or prostatectomy specimen sampling, with the latest also being correlated with higher detection rates of adverse pathological features, e.g., positive surgical margins, higher pathological stage or presence of perineural invasion (PnI status). The study cohort consisted of 315 consecutive patients diagnosed with prostate adenocarcinoma via transrectal ultrasound-guided needle biopsy who later underwent radical prostatectomy. We grouped and compared patients based on Grade Group accuracy, presence of IHC on biopsy, margin status, pathological stage, and PnI status. Inter-observer reproducibility was also calculated. Statistical analyzes included ANOVA, Tukey’s multiple comparisons post hoc test, Chi-squared test, and Fleiss kappa statistics. Undergraded cases harboured a significantly lower number of biopsy cores (p < 0.05), than accurately graded cases. Using IHC did not affect grading accuracy significantly, nor did the number of slides from prostatectomy specimens. The mean number of slides was virtually identical when margin status, pathological stage and PnI status of prostatectomy specimens were compared. Inter-observer reproducibility at our institute was calculated as fair (overall kappa = 0.29). Grade Group accuracy is significantly improved by obtaining more cores at biopsy but is unrelated to performed IHC. The extent of sampling prostatectomy specimens, however, did not affect accuracy and failed to significantly improve detection of adverse pathological features. C1 [Korpas, Levente Kristof] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Beke, Livia] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Varga, Daniel] University of Debrecen, Medical and Health Science Center, Department of UrologyDebrecen, Hungary. [Bidiga, Laszlo] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Molnar, Sarolta] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. RP Molnar, S (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, Debrecen, Hungary. EM molnar.sarolta@med.unideb.hu CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Brawley S, Mohan R, Nein CD. Localized prostate cancer: Treatment options. Am Fam Physician, 2018, 97(12):798–805. Epstein JI, Egevad L, Amin MB, Delahunt B, Srigley JR, Humphrey PA, et al. The 2014 international society of urological pathology, ISUP, consensus conference on Gleason grading of prostatic carcinoma: Definition of grading patterns and proposal for a new grading system. Am J Surg Pathol, 2016, 40(2):244–52., DOI 10. 1097/PAS.0000000000000530 Penney KL, Stampfer MJ, Jahn JL, Sinnott JA, Flavin R, Rider JR, et al. Gleason grade progression is uncommon. Cancer Res, 2013, 73(16):5163–8., DOI 10.1158/ 0008-5472.CAN-13-0427 Freedland SJ, Kane CJ, Amling CL, Aronson WJ, Presti JC, Terris MK, et al. Delay of radical prostatectomy and risk of biochemical progression inmen with low risk prostate cancer. J Urol, 2006, 175(4):1298–302., DOI 10.1016/S0022-5347(05, 00646-4 Sampurno F, Earnest A, Millar J, Frydenberg M, Murphy D, Delprado W, et al. Population-based study of grade progression in patients who harboured Gleason 3 + 3. World J Urol, 2017, 35(11):1689–99., DOI 10.1007/s00345-017- 2047-z Inoue LY, Trock BJ, Partin AW, Carter HB, Etzioni R. Modeling grade progression in an active surveillance study. Stat Med, 2014, 33(6):930–9., DOI 10. 1002/sim.6003 Epstein JI, Egevad L, Humphrey PA, Montironi R, Group MotIIiDUP. Best practices recommendations in the application of immunohistochemistry in the prostate: Report from the international society of urologic pathology consensus conference. Am J Surg Pathol, 2014, 38(8):e6–e19., DOI 10.1097/PAS. 0000000000000238 Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics, 1977, 33(1):159–74., DOI 10.2307/2529310 Cohen MS, Hanley RS, Kurteva T, Ruthazer R, Silverman ML, Sorcini A, et al. Comparing the Gleason prostate biopsy and Gleason prostatectomy grading system: The lahey clinic medical center experience and an international metaanalysis. Eur Urol, 2008, 54(2):371–81., DOI 10.1016/j.eururo.2008.03.049 Danneman D, Drevin L, Delahunt B, Samaratunga H, Robinson D, Bratt O, et al. Accuracy of prostate biopsies for predicting Gleason score in radical prostatectomy specimens: Nationwide trends 2000-2012. BJU Int, 2017, 119(1): 50–6., DOI 10.1111/bju.13458 San Francisco IF, DeWolf WC, Rosen S, Upton M, Olumi AF. Extended prostate needle biopsy improves concordance of Gleason grading between prostate needle biopsy and radical prostatectomy. J Urol, 2003, 169(1):136–40., DOI 10.1097/ 01.ju.0000042811.83736.04 Mian BM, Lehr DJ,Moore CK, Fisher HA, Kaufman RP, Ross JS, et al. Role of prostate biopsy schemes in accurate prediction of Gleason scores. Urology, 2006, 67(2):379–83., DOI 10.1016/j.urology.2005.08.018 Coogan CL, Latchamsetty KC, Greenfield J, Corman JM, Lynch B, Porter CR. Increasing the number of biopsy cores improves the concordance of biopsy Gleason score to prostatectomy Gleason score. BJU Int, 2005, 96(3):324–7., DOI 10.1111/j. 1464-410X.2005.05624.x Athanazio D, Gotto G, Shea-Budgell M, Yilmaz A, Trpkov K. Global Gleason grade groups in prostate cancer: Concordance of biopsy and radical prostatectomy grades and predictors of upgrade and downgrade. Histopathology, 2017, 70(7): 1098–106., DOI 10.1111/his.13179 Chun FK, Briganti A, Shariat SF, Graefen M, Montorsi F, Erbersdobler A, et al. Significant upgrading affects a third of men diagnosed with prostate cancer: Predictive nomogram and internal validation. BJU Int, 2006, 98(2):329–34., DOI 10.1111/j.1464-410X.2006.06262.x Egevad L, Norlen BJ, Norberg M. The value of multiple core biopsies for predicting the Gleason score of prostate cancer. BJU Int, 2001, 88(7):716–21., DOI 10.1046/j.1464-4096.2001.02419.x Miyake H, Kurahashi T, Takenaka A, Hara I, Fujisawa M. Improved accuracy for predicting the Gleason score of prostate cancer by increasing the number of transrectal biopsy cores. Urol Int, 2007, 79(4):302–6., DOI 10.1159/ 000109713 Antunes AA, Leite KR, Dall’Oglio MF, Cury J, Srougi M. The effect of the number of biopsy cores on the concordance between prostate biopsy and prostatectomy Gleason score: A prostate volume-controlled study. Arch Pathol LabMed, 2008, 132(6):989–92., DOI 10.1043/1543-2165(2008)132[989:TEOTNO]2. 0.CO;2 Jiang Z, Li C, Fischer A, Dresser K, Woda BA. Using an AMACR, P504S)/ 34betaE12/p63 cocktail for the detection of small focal prostate carcinoma in needle biopsy specimens. Am J Clin Pathol, 2005, 123(2):231–6., DOI 10.1309/ 1g1nk9dbgfnb792l Yu T, Zhu SX, Zheng S, Chen SP. Detection of AMACR, P504S), P63 and 34betaE12 cocktail in the early diagnosis of prostate cancer. Zhonghua Nan Ke Xue, 2007, 13(3):222–5. Paner GP, Luthringer DJ, Amin MB. Best practice in diagnostic immunohistochemistry: Prostate carcinoma and its mimics in needle core biopsies. Arch Pathol Lab Med, 2008, 132(9):1388–96., DOI 10.1043/1543- 2165(2008)132[1388:BPIDIP]2.0.CO;2 Collette ERP, den Bakker MA, Klaver SO, Vis AN, Kliffen M. Partial versus complete prostatectomy specimen sampling: Prospective non-inferiority study for pT3a tumours and surgical margin involvement. BMJ Open, 2019, 9(4):e024524., DOI 10.1136/bmjopen-2018-024524 Hall GS, Kramer CE, Epstein JI. Evaluation of radical prostatectomy specimens. A comparative analysis of sampling methods. Am J Surg Pathol, 1992, 16(4):315–24., DOI 10.1097/00000478-199204000-00001 Cohen MB, Soloway MS, Murphy WM. Sampling of radical prostatectomy specimens. How much is adequate? Am J Clin Pathol, 1994, 101(3):250–2., DOI 10. 1093/ajcp/101.3.250 Hollenbeck BK, Bassily N, Wei JT, Montie JE, Hayasaka S, Taylor JM, et al. Whole mounted radical prostatectomy specimens do not increase detection of adverse pathological features. J Urol, 2000, 164(5):1583–6., DOI 10.1016/s0022- 5347(05)67033-4 Sehdev AE, Pan CC, Epstein JI. Comparative analysis of samplingmethods for grossing radical prostatectomy specimens performed for nonpalpable, stage T1c, prostatic adenocarcinoma. Hum Pathol, 2001, 32(5):494–9., DOI 10.1053/hupa. 2001.24322 Vainer B, Toft BG, Olsen KE, Jacobsen GK, Marcussen N. Handling of radical prostatectomy specimens: Total or partial embedding? Histopathology, 2011, 58(2): 211–6., DOI 10.1111/j.1365-2559.2011.03741.x Llanos CA, Blieden C, Vernon SE. Processing radical prostatectomies: An alternate-slice method is comparable with total embedding. Ann Diagn Pathol, 2012, 16(4):284–7., DOI 10.1016/j.anndiagpath.2011.10.004 Kim K, Pak PJ, Ro JY, Shin D, Huh SJ, Cho YM. Limited sampling of radical prostatectomy specimens with excellent preservation of prognostic parameters of prostate cancer. Arch Pathol Lab Med, 2009, 133(8):1278–84., DOI 10.1043/1543- 2165-133.8.1278 Desai A, Wu H, Sun L, Sesterhenn IA, Mostofi FK, McLeod D, et al. Complete embedding and close step-sectioning of radical prostatectomy specimens both increase detection of extra-prostatic extension, and correlate with increased diseasefree survival by stage of prostate cancer patients. Prostate Cancer Prostatic Dis, 2002, 5(3):212–8., DOI 10.1038/sj.pcan.4500600 Fadul LC, da Silva MT, de Freitas LA, Athanazio DA. Assessment of the circumferential margins, extraprostatic extension and Gleason score in radical prostatectomy specimens: Comparison of a partial embedding method with supplemental total inclusion of peripheral tissues. Pathol Res Pract, 2016, 212(3):217–21., DOI 10.1016/j.prp.2015.12.014 Grossfeld GD, Chang JJ, Broering JM, Miller DP, Yu J, Flanders SC, et al. Does the completeness of prostate sampling predict outcome for patients undergoing radical prostatectomy?: Data from the CAPSURE database. Urology, 2000, 56(3): 430–5., DOI 10.1016/s0090-4295(00)00705-6 Iremashvili V, Lokeshwar SD, Jorda M, Pelaez L, Soloway MS. Prognostic implications of partial sampling of radical prostatectomy specimens: Comparison of 3 methods. J Urol, 2013, 190(1):84–90., DOI 10.1016/j.juro.2012.12.021 Iremashvili V, Lokeshwar SD, Soloway MS, Pelaez L, Umar SA, Manoharan M, et al. Partial sampling of radical prostatectomy specimens: Detection of positive margins and extraprostatic extension. Am J Surg Pathol, 2013, 37(2):219–25., DOI 10.1097/PAS.0b013e318268ccc1 Mottet N, Bellmunt J, Bolla M, Briers E, Cumberbatch MG, De Santis M, et al. EAU-ESTRO-SIOG guidelines on prostate cancer. Part 1: Screening, diagnosis, and local treatment with curative intent. Eur Urol, 2017, 71(4):618–29., DOI 10.1016/j. eururo.2016.08.003 Samaratunga H, Montironi R, True L, Epstein JI, Griffiths DF, Humphrey PA, et al. International society of urological pathology, ISUP, consensus conference on handling and staging of radical prostatectomy specimens. Working group 1: Specimen handling. Mod Pathol, 2011, 24(1):6–15., DOI 10.1038/modpathol. 2010.178 Ozkan TA, Eruyar AT, Cebeci OO, Memik O, Ozcan L, Kuskonmaz I. Interobserver variability in Gleason histological grading of prostate cancer. Scand J Urol, 2016, 50(6):420–4., DOI 10.1080/21681805.2016.1206619 Bori R, Salamon F, Moczar C, Cserni G. Interobserver reproducibility of Gleason grading in prostate biopsy samples. Orv Hetil, 2013, 154(31):1219–25., DOI 10.1556/OH.2013.29659 Singh RV, Agashe SR, Gosavi AV, Sulhyan KR. Interobserver reproducibility of Gleason grading of prostatic adenocarcinoma among general pathologists. Indian J Cancer, 2011, 48(4):488–95., DOI 10.4103/0019-509X.92277 Oyama T, Allsbrook WC, Kurokawa K,Matsuda H, Segawa A, Sano T, et al. A comparison of interobserver reproducibility of Gleason grading of prostatic carcinoma in Japan and the United States. Arch Pathol Lab Med, 2005, 129(8): 1004–10., DOI 10.1043/1543-2165(2005)129[1004:ACOIRO]2.0.CO;2 Mulay K, Swain M, Jaiman S, Gowrishankar S. Gleason scoring of prostatic carcinoma: Impact of a web-based tutorial on inter- and intra-observer variability. Indian J Pathol Microbiol, 2008, 51(1):22–5., DOI 10.4103/0377-4929.40385 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611157 EP 1611166 DI 10.3389/pore.2023.1611157 PG 10 ER PT J AU Leeha, M Kanokwiroon, K Laohawiriyakamol, S Thongsuksai, P AF Leeha, Marisa Kanokwiroon, Kanyanatt Laohawiriyakamol, Suphawat Thongsuksai, Paramee TI Immunohistochemistry-based molecular subtyping of triple-negative breast cancer and its prognostic significance SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; breast cancer; prognosis; triple-negative breast cancer; molecular subtype ID immunohistochemistry; breast cancer; prognosis; triple-negative breast cancer; molecular subtype AB Background: Immunohistochemistry (IHC)-based protein markers representing molecular subtypes are of great value for routine use. This study aimed to evaluate the frequency distributions of the molecular subtypes of triple-negative breast cancer (TNBC) using IHC-based surrogate markers and examined their prognostic value. Methods: Patients with TNBC treated at a university hospital in Southern Thailand were included in this study. Expression levels of androgen receptor, CD8, Forkhead box transcription factor C1, and Doublecortin-like kinase 1 were detected in tumor tissue to classify them into luminal androgen receptor (LAR), immunomodulatory (IM), basal-like immunosuppressed (BLIS), mesenchymallike (MES), and unclassifiable (UC) subtypes. The association between variables and disease-free survival (DFS) and overall survival (OS) was analyzed using Cox proportional hazards regression. Results: Among the 195 cases of TNBC, the frequency distribution of the IHCbased subtype was as follows: BLIS, 52.8%; LAR, 19.0%; IM, 17.4%; MES, 0.5%; and un-classifiable, 10.3%. BLIS subtype was significantly found in younger ages (mean: 49.6 years) than other subtypes (mean: 51–57.7 years). LAR and BLIS subtypes were significantly associated with poorer OS compared to the IM subtype in univariate analysis, however, only BLIS was significant in multivariate analysis (HR: 3.29, 95% CI: 1.01–10.72). IHC-based subtype was not found to be associated with DFS. Conclusion: This study revealed the differences in the proportion frequency of IHCbased TNBC subtypes in Thai patients compared to other populations. IHC-based molecular subtypingmay be beneficial for prognosis. However further refinement of the molecular classification of TNBC is needed for better clinical relevance. C1 [Leeha, Marisa] Prince of Songkla University, Faculty of Medicine, Department of Biomedical Sciences and Biomedical EngineeringHat Yai, Songkhla, Thailand. [Kanokwiroon, Kanyanatt] Prince of Songkla University, Faculty of Medicine, Department of Biomedical Sciences and Biomedical EngineeringHat Yai, Songkhla, Thailand. [Laohawiriyakamol, Suphawat] Prince of Songkla University, Faculty of Medicine, Department of SurgeryHat Yai, Songkhla, Thailand. [Thongsuksai, Paramee] Prince of Songkla University, Faculty of Medicine, Department of PathologyHat Yai, Songkhla, Thailand. RP Thongsuksai, P (reprint author), Prince of Songkla University, Faculty of Medicine, Department of Pathology, Hat Yai, Thailand. EM tparamee@gmail.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Morris GJ, Naidu S, Topham AK, Guiles F, Xu Y,McCue P, et al. Differences in breast carcinoma characteristics in newly diagnosed african-American and caucasian patients: A single-institution compilation compared with the national cancer institute’s surveillance, epidemiology, and end results database. Cancer, 2007, 110:876–84., DOI 10.1002/cncr.22836 Hines LM, Risendal B, Byers T, Mengshol S, Lowery J, Singh M. Ethnic disparities in breast tumor phenotypic subtypes in Hispanic and non-Hispanic white women. J Womens Health, Larchmt,, 2011, 20:1543–50., DOI 10.1089/jwh. 2010.2558 Zhang W, Bai Y, Sun C, Lv Z, Wang S. Racial and regional disparities of triple negative breast cancer incidence rates in the United States: An analysis of 2011- 2019 NPCR and SEER incidence data. Front Public Health, 2022, 10:1058722., DOI 10.3389/fpubh.2022.1058722 Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, et al. Triple-negative breast cancer: Clinical features and patterns of recurrence. Clin Cancer Res, 2007, 13:4429–34., DOI 10.1158/1078-0432.CCR-06-3045 Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. Clin Invest, 2011, 121:2750–67., DOI 10.1172/JCI45014 Lehmann BD, Jovanovic B, Chen X, Estrada MV, Johnson KN, Shyr Y, et al. Refinement of triple-negative breast cancer molecular subtypes: Implications for neoadjuvant chemotherapy selection. PLoS One, 2016, 11:e0157368., DOI 10.1371/ journal.pone.0157368 Burstein MD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SA, et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res, 2015, 21:1688–98., DOI 10.1158/1078- 0432.CCR-14-0432 Jiang YZ, Ma D, Suo C, Shi J, Xue M, Hu X, et al. Genomic and transcriptomic landscape of triple-negative breast cancers: Subtypes and treatment strategies. Cancer Cell, 2019, 35:428–40., DOI 10.1016/j.ccell.2019. 02.001 Zhao S, Ma D, Xiao Y, Li XM, Ma JL, Zhang H, et al. Molecular subtyping of triple-negative breast cancers by immunohistochemistry: Molecular basis and clinical relevance. Oncologist, 2020, 25:e1481–e1491., DOI 10.1634/theoncologist.2019-0982 Niyomnaitham S, Parinyanitikul N, Roothumnong E, Jinda W, Samarnthai N, Atikankul T, et al. Tumor mutational profile of triple negative breast cancer patients in Thailand revealed distinctive genetic alteration in chromatin remodeling gene. PeerJ, 2019, 7:e6501., DOI 10.7717/peerj.6501 Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, et al. American society of clinical oncology/college of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. Arch Pathol Lab Med, 2010, 134:907–22., DOI 10.1043/ 1543-2165-134.6.907 Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American society of clinical oncology/college of American Pathologists clinical practice guideline update. J Clin Oncol, 2013, 31: 3997–4013., DOI 10.1200/JCO.2013.50.9984 Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, et al. Human epidermal growth factor receptor 2 testing in breast cancer: American society of clinical oncology/college of American Pathologists clinical practice guideline focused update. Arch Pathol Lab Med, 2018, 142:1364–82., DOI 10.5858/ arpa.2018-0902-SA Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, et al. The evaluation of tumor-infiltrating lymphocytes, TILs, in breast cancer: Recommendations by an international TILs working group 2014. Ann Oncol, 2015, 26:259–71., DOI 10.1093/annonc/mdu450 Ding YC, Steele L, Warden C, Wilczynski S, Mortimer J, Yuan Y, et al. Molecular subtypes of tri-ple-negative breast cancer in women of different race and ethnicity. Oncotarget, 2019, 10:198–208., DOI 10.18632/oncotarget.26559 Goff SL, Danforth DN. The role of immune cells in breast tissue and immunotherapy for the treatment of breast cancer. Clin Breast Cancer, 2021, 21:e63–e73., DOI 10.1016/j.clbc.2020.06.011 Katsuta E, Yan L, OpyrchalM, Kalinski P, Takabe K. Cytotoxic T-lymphocyte infiltration and chemokine predict long-term patient survival independently of tumor mutational burden in triple-negative breast cancer. Ther Adv Med Oncol, 2021, 13:17588359211006680., DOI 10.1177/17588359211006680 Chen F, Chen N, Gao Y, Jia L, Lyu Z, Cui J. Clinical progress of PD-1/ L1 inhibitors in breast cancer immunotherapy. Front Oncol, 2022, 11:724424., DOI 10.3389/fonc.2021.724424 Lian J, Ma HX, Xu EW, Bu P, Yun KM, Xi YF. Subclassifying triple-negative breast cancers and its potential clinical utility. Virchows Arch, 2022, 481:13–21., DOI 10.1007/s00428-022-03329-0 Salisbury T, Abozina A, Zhang C,Mao E, Banyi N, Leo J, et al. Histological subtype is associated with PD-L1 expression and CD8+ T-cell infiltrates in triple-negative breast carcinoma. Ann Diagn Pathol, 2022, 57:151901., DOI 10.1016/j.anndiagpath.2022.151901 Kim S, Moon BI, Lim W, Park S, ChoMS, Sung SH. Feasibility of classification of triple negative breast cancer by immunohistochemical surrogate markers. Clin Breast Cancer, 2018, 18:e1123–e1132., DOI 10.1016/j.clbc.2018.03.012 Kumar S, Bal A, Das A, Bhattacharyya S, Laroiya I, Khare S, et al. Molecular subtyping of triple negative breast cancer by surrogate immunohistochemistry markers. Appl Immunohistochem Mol Morphol, 2021, 29:251–7., DOI 10.1097/PAI. 0000000000000897 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611162 EP 1611171 DI 10.3389/pore.2023.1611162 PG 10 ER PT J AU Gyulai, M Megyesfalvi, Zs Reiniger, L Harko, T Ferencz, B Karsko, L Agocs, L Fillinger, J Dome, B Szallasi, Z Moldvay, J AF Gyulai, Marton Megyesfalvi, Zsolt Reiniger, Lilla Harko, Tunde Ferencz, Bence Karsko, Luca Agocs, Laszlo Fillinger, Janos Dome, Balazs Szallasi, Zoltan Moldvay, Judit TI PD-1 and PD-L1 expression in rare lung tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE adenoid cystic carcinoma; mucoepidermoid carcinoma; programmed cell death ligand-1 (PD-L1); programmed cell death-1 (PD-1); rare lung tumors ID adenoid cystic carcinoma; mucoepidermoid carcinoma; programmed cell death ligand-1 (PD-L1); programmed cell death-1 (PD-1); rare lung tumors AB Background: Our knowledge is still limited about the characteristics and treatment of rare lung tumors. The aim of our study was to determine programmed cell death ligand-1 (PD-L1) and programmed cell death-1 (PD- 1) expression in rare pulmonary tumors to assess the potential role of immunotherapy. Methods: 66 pathologically confirmed rare lung tumors including 26 mucoepidermoid carcinomas (MECs), 27 adenoid cystic carcinomas (ACCs), and 13 tracheobronchial papillomas (TBPs) were collected retrospectively. Immunohistochemical (IHC) staining was performed on formalin fixed paraffin embedded (FFPE) tumor tissues, and PD-L1 expression on tumor cells (TCs) and immune cells (ICs), and PD-1 expression on ICs were determined. The cut off value for positive immunostaining was set at 1% for all markers. Results: PD-L1 expression on TCs was observed in two cases of MEC (7.7%), one case of ACC (3.7%), and was absent in TBP samples. PD-L1 expression on ICs could be demonstrated in nine cases of MEC (34.6%), four cases of ACC (14.8%), and was absent in TBPs. All PD-L1 TC positive tumors were also PDL1 IC positive. Higher expression level than 5% of PD-L1 TC and/or IC was observed only in one ACC and in two MEC patients. Among them, strong PDL1 immunopositivity of >50% on TCs and of >10% on ICs could be demonstrated in one MEC sample. PD-L1 expression of ≥1% on ICs was significantly more common in MEC, than in TBP (p < 0.001). In MEC ≥1% PDL1 TC or IC expressions were significantly more common in patients aged 55 or older, than in younger patients (p = 0.046, and p = 0.01, respectively). PD-1 expression on ICs was found in five cases of MEC (19.2%), four cases of ACC (14.8%), and in two cases of TBP (15.4%). Only one MEC case showed a higher than 5% expression level of PD-1 on ICs. Conclusion: This retrospective study comprehensively demonstrated the rare expression of PD-L1 and PD-1 in pulmonary MEC, ACC, and TBP. However, we found very strong PD-L1 immunopositivity on both TCs and ICs in one MEC sample, which warrants further investigations in a larger cohort. C1 [Gyulai, Marton] County Hospital of PulmonologyTorokbalint, Hungary. [Megyesfalvi, Zsolt] National Koranyi Institute of PulmonologyBudapest, Hungary. [Reiniger, Lilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Harko, Tunde] National Koranyi Institute of PulmonologyBudapest, Hungary. [Ferencz, Bence] National Koranyi Institute of PulmonologyBudapest, Hungary. [Karsko, Luca] National Koranyi Institute of PulmonologyBudapest, Hungary. [Agocs, Laszlo] National Koranyi Institute of PulmonologyBudapest, Hungary. [Fillinger, Janos] National Koranyi Institute of PulmonologyBudapest, Hungary. [Dome, Balazs] National Koranyi Institute of PulmonologyBudapest, Hungary. [Szallasi, Zoltan] National Koranyi Institute of PulmonologyBudapest, Hungary. [Moldvay, Judit] National Koranyi Institute of PulmonologyBudapest, Hungary. RP Moldvay, J (reprint author), National Koranyi Institute of Pulmonology, Budapest, Hungary. EM drmoldvay@hotmail.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Shao T, Zhao M, Liang L, Tang W. A systematic review and network metaanalysis of first-line immune checkpoint inhibitor combination therapies in patients with advanced non-squamous non-small cell lung cancer. Front Immunol, 2022, 13:948597., DOI 10.3389/fimmu.2022.948597 O’Byrne K, Popoff E, Badin F, Lee A, Yuan Y, Lozano-Ortega G, et al. Longterm comparative efficacy and safety of nivolumab plus ipilimumab relative to other first-line therapies for advanced non-small-cell lung cancer: A systematic literature review and network meta-analysis. Lung Cancer, 2023, 177:11–20., DOI 10.1016/j. lungcan.2023.01.006 Ancevski Hunter K, Socinski MA, Villaruz LC. PD-L1 testing in guiding patient selection for PD-1/PD-L1 inhibitor therapy in lung cancer. Mol Diagn Ther, 2018, 22(1):1–10., DOI 10.1007/s40291-017-0308-6 Maule JG, Clinton LK, Graf RP, Xiao J, Oxnard GR, Ross JS, et al. Comparison of PD-L1 tumor cell expression with 22C3, 28-8, and SP142 IHC assays across multiple tumor types. J Immunother Cancer, 2022, 10(10):e005573., DOI 10.1136/ jitc-2022-005573 Pisano C, Witel G, De Filippis M, Listi ̀A, Napoli F, Righi L, et al. Moving through rare lung cancer histologies: A narrative review on diagnosis and treatment of selected infrequent entities. Precis Cancer Med, 2022, 5:27., DOI 10.21037/pcm- 22-4 Lee RH, Wai KC, Chan JW, Ha PK, Kang H. Approaches to themanagement of metastatic adenoid cystic carcinoma. Cancers, Basel,, 2022, 14(22):5698., DOI 10. 3390/cancers14225698 Garg PK, Sharma G, Rai S, Jakhetiya A. Primary salivary gland-type tumors of the lung: A systematic review and pooled analysis. Lung India, 2019, 36(2):118–22., DOI 10.4103/lungindia.lungindia_284_18 Zheng KF, Liu YJ, Ma N, Xiong YL, Tang XY, Zhang Q, et al. PD-L1 expression and immune cells infiltration in primary tracheobronchial neoplasm. Transl Lung Cancer Res, 2021, 10(12):4617–30., DOI 10.21037/tlcr-21-958 Festino L, Botti G, Lorigan P, Masucci GV, Hipp JD, Horak CE, et al. Cancer treatment with anti-PD-1/PD-L1 agents: Is PD-L1 expression a biomarker for patient selection? Drugs, 2016, 76(9):925–45., DOI 10.1007/ s40265-016-0588-x Yang CY, Lin MW, Chang YL, Wu CT, Yang PC. Programmed cell deathligand 1 expression is associated with a favourable immune microenvironment and better overall survival in stage I pulmonary squamous cell carcinoma. Eur J Cancer, 2016, 57:91–103., DOI 10.1016/j.ejca.2015.12.033 Teglasi V, Pipek O, Lozsa R, Berta K, Szuts D, Harko T, et al. PD-L1 expression of lung cancer cells, unlike infiltrating immune cells, is stable and unaffected by therapy during brain metastasis. Clin Lung Cancer, 2019, 20(5):363–9., DOI 10.1016/ j.cllc.2019.05.008 Reiniger L, Teglasi V, Pipek O, Rojko L, Glasz T, Vagvolgyi A, et al. Tumor necrosis correlates with PD-L1 and PD-1 expression in lung adenocarcinoma. Acta Oncol, 2019, 58(8):1087–94., DOI 10.1080/0284186X. 2019.1598575 Rojko L, Reiniger L, Teglasi V, Fabian K, Pipek O, Vagvolgyi A, et al. Chemotherapy treatment is associated with altered PD-L1 expression in lung cancer patients. J Cancer Res Clin Oncol, 2018, 144(7):1219–26., DOI 10.1007/s00432-018- 2642-4 Teglasi V, Reiniger L, Fabian K, Pipek O, Csala I, Bago AG, et al. Evaluating the significance of density, localization, and PD-1/PDL1 immunopositivity of mononuclear cells in the clinical course of lung adenocarcinoma patients with brain metastasis. Neuro Oncol, 2017, 19(8): 1058–67., DOI 10.1093/neuonc/now309 Guan R, Lyu Q, Lin A, Liang J, Ding W, CaoM, et al. Influence of different age cutoff points on the prediction of prognosis of cancer patients receiving ICIs and potential mechanistic exploration. Front Oncol, 2021, 11:670927., DOI 10.3389/fonc. 2021.670927 Li D, Pang X, Zhu X, Shanzhou Q, Wen G, Ma D. Low-dose radiotherapy combined with immunotherapy for suboral adenoid cystic carcinoma with bilateral lung metastasis: A case report and literature review. Oncol Lett, 2022, 24(2):279., DOI 10.3892/ol.2022.13399 Farhat R, Asna N, Avraham Y, Khater A, Asakla M, Safia A, et al. Pembrolizumab as a first line therapy in a patient with extensive mucoepidermoid salivary gland carcinoma. A complete clinical, radiological and pathological response. A very specific case. Discov Oncol, 2022, 13(1):37., DOI 10. 1007/s12672-022-00502-4 Pharaon RR, Gernon T, Chang S, Vora N, Villaflor VM, Bell D, et al. Prolonged response to checkpoint inhibitor therapy in two metastatic mucoepidermoid salivary gland carcinoma cases: A research report. Cold Spring Harb Mol Case Stud, 2022, 8(3):a006189., DOI 10.1101/mcs.a006189 Mueller SK, Haderlein M, Lettmaier S, Agaimy A, Haller F, Hecht M, et al. Targeted therapy, chemotherapy, immunotherapy and novel treatment options for different subtypes of salivary gland cancer. J Clin Med, 2022, 11(3):720., DOI 10. 3390/jcm11030720 Hu S, Gong J, Zhu X, Lu H. Pulmonary salivary gland tumor, mucoepidermoid carcinoma: A literature review. J Oncol, 2022, 2022:9742091., DOI 10.1155/2022/9742091 Liu J, Wu L, Lin Q. Expression of programmed death receptor 1 and its ligand in primary lung mucoepidermoid carcinoma and its clinical significance. Fujian Med J, 2018, 40(6):16–9. Gerdabi S, Asadian F, Kiani R, Khademi B, Haghshenas MR, Erfani N. Simultaneous expression of PD-1 and PD-L1 in peripheral and central immune cells and tumor cells in the benign and malignant salivary gland tumors microenvironment. Head Neck Pathol, 2022, 17(1):178–92., DOI 10.1007/s12105- 022-01486-x Witte HM, Gebauer N, Lappohn D, Umathum VG, Riecke A, Arndt A, et al. Prognostic impact of PD-L1 expression in malignant salivary gland tumors as assessed by established scoring criteria: Tumor proportion score, TPS), combined positivity score, CPS), and immune cell, IC, infiltrate. Cancers, 2020, 12(4):873., DOI 10.3390/cancers12040873 Xu B, Jungbluth AA, Frosina D, Alzumaili B, Aleynick N, Slodkowska E, et al. The immune microenvironment and expression of PD-L1, PD-1, PRAME and MHC I in salivary duct carcinoma. Histopathology, 2019, 75(5):672–82., DOI 10. 1111/his.13944 Mosconi C, de Arruda JAA, de Farias ACR, Oliveira GAQ, de Paula HM, Fonseca FP, et al. Immune microenvironment and evasion mechanisms in adenoid cystic carcinomas of salivary glands. Oral Oncol, 2019, 88:95–101., DOI 10.1016/j. oraloncology.2018.11.028 Hendry S, Byrne DJ, Wright GM, Young RJ, Sturrock S, Cooper WA, et al. Comparison of four PD-L1 immunohistochemical assays in lung cancer. J Thorac Oncol, 2018, 13(3):367–76., DOI 10.1016/j.jtho.2017.11.112 Peng Y, Li Z, Fu Y, Pan Y, Zeng Y, Liu J, et al. Progress and perspectives of perioperative immunotherapy in non-small cell lung cancer. Front Oncol, 2023, 13: 1011810., DOI 10.3389/fonc.2023.1011810 Shah M, Marmarelis ME, Mamtani R, Hennessy S. Association between survival and very high versus high PD-L1 expression in patients receiving pembrolizumab as first-line treatment for advanced non-small cell lung cancer. Clin Lung Cancer, 2022, 23(8):731–6., DOI 10.1016/j.cllc.2022.07.003 Stinchcombe TE. Narrative review: Blood and tumor biomarker testing in non-small cell lung cancer without an oncogenic driver. Transl Lung Cancer Res, 2023, 12(1):158–67., DOI 10.21037/tlcr-22-530 Wankhede D, Hofman P, Grover S. PD-1/PD-L1 inhibitors in treatmentnaive, advanced non-small cell lung cancer patients with < 1% PD-L1 expression: A meta-analysis of randomized controlled trials. J Cancer Res Clin Oncol, 2022, 149: 2179–89., DOI 10.1007/s00432-022-04286-6 Chevallier M, Borgeaud M, Addeo A, Friedlaender A. Oncogenic driver mutations in non-small cell lung cancer: Past, present and future. World J Clin Oncol, 2021, 12(4):217–37., DOI 10.5306/wjco.v12.i4.217 Lang C, Lantos A, Megyesfalvi Z, Egger F, HodaMA,Mosleh B, et al. Clinical and prognostic implications of CD47 and PD-L1 expression in surgically resected small-cell lung cancer. ESMO Open, 2022, 7(6):100631., DOI 10.1016/j.esmoop.2022.100631 Qi J, Zhang J, Liu N, Zhao L, Xu B. Prognostic implications of molecular subtypes in primary small cell lung cancer and Their correlation with cancer immunity. Front Oncol, 2022, 12:779276., DOI 10.3389/fonc.2022.779276 de Sousa LG, Jovanovic K, Ferrarotto R. Metastatic adenoid cystic carcinoma: Genomic landscape and emerging treatments. Curr Treat Options Oncol, 2022, 23(8):1135–50., DOI 10.1007/s11864-022-01001-y NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611164 EP 1611172 DI 10.3389/pore.2023.1611164 PG 9 ER PT J AU Wiedemann, Szita, RV Horvath, R Szederjesi, A Sebo, A Toth, DA Masszi, T Varga, G AF Wiedemann, Adam Szita, Reka Virag Horvath, Robert Szederjesi, Attila Sebo, Attila Toth, David Andras Masszi, Tamas Varga, Gergely TI Soluble B-cell maturation antigen as a monitoring marker for multiple myeloma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE multiple myeloma; monitoring; monoclonal gammopathie; soluble B-cell maturation antigen; BCMA ID multiple myeloma; monitoring; monoclonal gammopathie; soluble B-cell maturation antigen; BCMA AB Objective: Response to treatment in multiple myeloma (MM) is routinely measured by serum and urine M-protein and free light chain (FLC), as described by the International Myeloma Working Group (IMWG) consensus statement. A non-negligible subgroup of patients however present without measurable biomarkers, others become oligo or non-secretory during recurrent relapses. The aim of our research was to evaluate soluble B-cell maturation antigen (sBCMA) as a monitoring marker measured concurrent with the standard monitoring in MM patients at diagnosis, at relapse and during follow up, in order to establish its potential usefulness in oligo and nonsecretory disease. Method: sBCMA levels were measured in 149 patients treated for plasma cell dyscrasia (3 monoclonal gammopathy of unknown significance, 5 smoldering myeloma, 7 plasmacytoma, 8 AL amyloidosis and 126 MM) and 16 control subjects using a commercial ELISA kit. In 43 newly diagnosed patients sBCMA levels were measured at multiple timepoints during treatment, and compared to conventional IMWG response and progression free survival (PFS). Results: sBCMA levels among control subjects were significantly lower than among newly diagnosed or relapsed MM patients [20.8 (14.7–38.7) ng/mL vs. 676 (89.5–1,650) and 264 (20.7–1,603) ng/mL, respectively]. Significant correlations were found between sBCMA and the degree of bone marrow plasma cell infiltration. Out of the 37 newly diagnosed patients who have reached partial response or better per IMWG criteria, 33 (89%) have had at least a 50% drop in sBCMA level by therapy week 4. Cohorts made similarly to IMWG response criteria—achieving a 50% or 90% drop in sBCMA levels compared to level at diagnosis—had statistically significant differences in PFS. Conclusion: Our results confirmed that sBCMA levels are prognostic at important decision points in myeloma, and the percentage of BCMA change is predictive for PFS. This highlights the great potential use of sBCMA in oligoand non-secretory myeloma. C1 [Wiedemann, Adam] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Szita, Reka Virag] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Horvath, Robert] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Szederjesi, Attila] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Sebo, Attila] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Toth, David Andras] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Masszi, Tamas] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Varga, Gergely] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. RP Varga, G (reprint author), Semmelweis University, Department of Internal Medicine and Haematology, Budapest, Hungary. EM vargager@gmail.com CR Durie BGM, Harousseau J-L, Miguel JS, Blade J, Barlogie B, Anderson K, et al. International uniform response criteria for multiple myeloma. Leukemia, 2006, 20: 1467–73., DOI 10.1038/sj.leu.2404284 Kyle RA, Rajkumar SV. Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma. Leukemia, 2009, 23:3–9., DOI 10.1038/ leu.2008.291 Blade J, Kyle RA. Nonsecretory myeloma, immunoglobulin D myeloma, and plasma cell leukemia. Hematol/oncol Clin North Am, 1999, 13:1259–72., DOI 10. 1016/S0889-8588(05)70125-8 Smith DB, Harris M, Gowland E, Chang J, Scarffe JH. Non-secretory multiple myeloma: A report of 13 cases with a review of the literature. Hematol Oncol, 1986, 4(4):307–13., DOI 10.1002/hon.2900040407 Ma ES, Shek TW, Ma SY. Non-secretory plasma cell myeloma of the true nonproducer type. Br J Haematol, 2007, 138(5):561., DOI 10.1111/j.1365-2141.2007. 691.x 6. Decourt C, Galea HR, Sirac C, Cogne M. Immunologic basis for the rare occurrence of true nonsecretory plasma cell dyscrasias. J Leukoc Biol, 2004, 76(3): 528–36., DOI 10.1189/jlb.0803382 Coriu D, Weaver K, Schell M, Eulitz M, Murphy CL, Weiss DT, et al. A molecular basis for nonsecretory myeloma. Blood, 2004, 104(3):829–31., DOI 10. 1182/blood-2004-02-0477 Meister S, Schubert U, Neubert K, Herrmann K, Burger R, Gramatzki M, et al. Extensive immunoglobulin production sensitizes myeloma cells for proteasome inhibition. Cancer Res, 2007, 67(4):1783–92., DOI 10.1158/0008-5472.CAN-06-2258 Trudel S, Lendvai N, Popat R, Voorhees PM, Reeves B, Libby EN, et al. Targeting B-cell maturation antigen with GSK2857916 antibody-drug conjugate in relapsed or refractory multiple myeloma, BMA117159): A dose escalation and expansion phase 1 trial. Lancet Oncol, 2018, 19(12):1641–53., DOI 10.1016/S1470- 2045(18)30576-X Berdeja JG, Madduri D, Usmani SZ, Jakubowiak A, Agha M, Cohen AD, et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma, CARTITUDE-1): A phase 1b/2 open-label study. Lancet, 2021, 398(10297): 314–24., DOI 10.1016/S0140-6736(21)00933-8 Raje N, Berdeja J, Lin Y, Siegel D, Jagannath S, Madduri D, et al. Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med, 2019, 380(18):1726–37., DOI 10.1056/NEJMoa1817226 Moreau P, Garfall AL, van de Donk NWCJ, Nahi H, San-Miguel JF, Oriol A, et al. Teclistamab in relapsed or refractory multiple myeloma. N Engl J Med, 2022, 387(6):495–505., DOI 10.1056/NEJMoa2203478 Laurent SA, Hoffmann FS, Kuhn PH, Cheng Q, Chu Y, Schmidt-Supprian M, et al. γ-Secretase directly sheds the survival receptor BCMA from plasma cells. Nat Commun, 2015, 6:7333., DOI 10.1038/ncomms8333 Samur MK, Fulciniti M, Aktas Samur A, Bazarbachi AH, Tai YT, Prabhala R, et al. Biallelic loss of BCMA as a resistance mechanism to CAR T cell therapy in a patient with multiple myeloma. Nat Commun, 2021, 12(1):868., DOI 10.1038/ s41467-021-21177-5 Seipel K, Porret N, Wiedemann G, Jeker B, Bacher VU, Pabst T. sBCMA plasma level dynamics and anti-BCMA CAR-T-cell treatment in relapsed multiple myeloma. Curr Issues Mol Biol, 2022, 44(4):1463–71., DOI 10.3390/cimb44040098 Sanchez E, Li M, Kitto A, Li J, Wang CS, Kirk DT, et al. Serum B-cell maturation antigen is elevated in multiple myeloma and correlates with disease status and survival. Br J Haematol, 2012, 158(6):727–38., DOI 10.1111/j.1365-2141. 2012.09241.x Visram A, Soof C, Rajkumar SV, Kumar SK, Bujarski S, Spektor TM, et al. Serum BCMA levels predict outcomes in MGUS and smoldering myeloma patients. Blood Cancer J, 2021, 11(6):120., DOI 10.1038/s41408-021-00505-4 Ghermezi M, Li M, Vardanyan S, Harutyunyan NM, Gottlieb J, Berenson A, et al. Serum B-cell maturation antigen: A novel biomarker to predict outcomes for multiple myeloma patients. Haematologica, 2017, 102(4):785–95., DOI 10.3324/ haematol.2016.150896 Ghermezi M, Vardanyan S, Harutyunyan NM, Gottlieb J, Berenson A, Andreu-Vieyra C, et al. Serum B-cell maturation antigen is a novel prognostic indicator for multiple myeloma patients and correlates with clinical status and survival. Blood, 2014, 124(21):3405., DOI 10.1182/blood.V124.21.3405.3405 Puig N, Contreras MT, Agullo C, Martinez-Lopez J, Oriol A, Blanchard MJ, et al. Mass spectrometry vs immunofixation for treatment monitoring in multiple myeloma. Blood Adv, 2022, 6(11):3234–9., DOI 10.1182/bloodadvances.2021006762 Dasari S, Kohlhagen MC, Dispenzieri A,Willrich MAV, Snyder MR, Kourelis TV, et al. Detection of plasma cell disorders by mass spectrometry: A comprehensive review of 19,523 cases. Mayo Clin Proc, 2022, 97(2):294–307., DOI 10.1016/j.mayocp.2021.07.024 Dispenzieri A, Arendt B, Dasari S, Kohlhagen M, Kourelis T, Kumar SK, et al. Blood mass spectrometry detects residual disease better than standard techniques in light-chain amyloidosis. Blood Cancer J, 2020, 10(2):20., DOI 10.1038/s41408-020- 0291-8 Tovar N, Fernandez de Larrea C, Elena M, Cibeira MT, Arostegui JI, Rosinol L, et al. Prognostic impact of serum immunoglobulin heavy/light chain ratio in patients with multiple myeloma in complete remission after autologous stem cell transplantation. Biol Blood Marrow Transpl, 2012, 18(7):1076–9., DOI 10.1016/j. bbmt.2012.03.004 Kumar S, Paiva B, Anderson KC, Durie B, Landgren O, Moreau P, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol, 2016, 17(8):e328–46., DOI 10.1016/s1470-2045(16)30206-6 Seidel C, Sundan A, Hjorth M, Turesson I, Dahl IM, Abildgaard N, et al. Serum syndecan-1: A new independent prognostic marker in multiple myeloma. Blood, 2000, 95(2):388–92., DOI 10.1182/blood.v95.2.388 Richard L, Janet AD, Gulnaz B, Nicola JB, Tim P, Paul AM, et al. Soluble syndecan-1 level at diagnosis is an independent prognostic factor in multiple myeloma and the extent of fall from diagnosis to plateau predicts for overall survival. B J Haem, 2005, 130:542–8., DOI 10.1111/j.1365-2141.2005.05647.x Janosi J, Sebestyen A, Mikala G, Nemeth J, Kiss Z, Valyi-Nagy I. Soluble syndecan-1 levels in different plasma cell dyscrasias and in different stages of multiple myeloma. Haematologica, 2004, 89:370–1. NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611171 EP 1611178 DI 10.3389/pore.2023.1611171 PG 8 ER PT J AU Zhang, L Zhang, X Wu, B Han, X Guo, C Li, B Jing, H Cheng, W AF Zhang, Lei Zhang, Xuefei Wu, Bolin Han, Xue Guo, Cunli Li, Bo Jing, Hui Cheng, Wen TI Prognostic value of albumin-bilirubin score in pancreatic cancer patients after pancreatoduodenectomy with liver metastasis following radiofrequency ablation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chemotherapy; pancreatic cancer; liver metastasis; radiofrequency ablation; pancreatoduodenectomy ID chemotherapy; pancreatic cancer; liver metastasis; radiofrequency ablation; pancreatoduodenectomy AB Objective: The current study aimed to investigate the prognostic value of albumin-bilirubin (ALBI) score in predicting clinical outcomes of pancreatic cancer patients after pancreatoduodenectomy with liver metastasis following radiofrequency ablation. Methods: This retrospective study included 90 pancreatic cancer patients after pancreatoduodenectomy with liver metastasis from January 2012 to December 2018. In this study, the Chi-square or Fisher’s exact tests, the receiver operating characteristic (ROC) curve, Kaplan-Meier method and Log-rank test, univariate and multivariate Cox proportional hazard regression analyses, nomogram, calibration curves and decision curve analysis were used for all statistical analysis. Results: We analyzed the optimal cut-off value of ALBI by ROC curve, and the optimal cut-off value was −2.60. According to ALBI score, these patients were divided into two groups: low ALBI group (n = 33) and high ALBI group (n = 57). Patients with low ALBI score was significantly related to longer progression free survival (PFS) (p = 0.0002, HR: 3.039, 95% CI: 1.772–5.210) and overall survival (OS) (p = 0.0005, HR: 2.697, 95% CI: 1.539–4.720). The 1-, 3-, and 5-year PFS and OS rates in low ALBI group were higher than those in high ALBI group. ALBI was a potential independent prognostic factor for pancreatic cancer patients after pancreatoduodenectomy with liver metastasis following radiofrequency ablation. Moreover, the nomogram was used to predict the 1-, 3-, and 5-year survival probabilities of PFS and OS. The calibration curve shown that the prediction line matched the reference line well for postoperative 3-year PFS and OS. The DCA shown that nomogram model was better than the only ALBI, and indicated the ability for clinical decision-making, especially in 1-year PFS, and 3-, 5-year OS. Conclusion: ALBI is a potential independent factor for PFS and OS, and can predict the prognosis of pancreatic cancer patients after pancreatoduodenectomy with liver metastasis following radiofrequency ablation. C1 [Zhang, Lei] Harbin Medical University Cancer Hospital, Department of UltrasoundHarbin, China. [Zhang, Xuefei] Harbin Medical University Cancer Hospital, Department of UltrasoundHarbin, China. [Wu, Bolin] Harbin Medical University Cancer Hospital, Department of UltrasoundHarbin, China. [Han, Xue] Harbin Medical University Cancer Hospital, Department of UltrasoundHarbin, China. [Guo, Cunli] Harbin Medical University Cancer Hospital, Department of UltrasoundHarbin, China. [Li, Bo] Harbin Medical University Cancer Hospital, Department of UltrasoundHarbin, China. [Jing, Hui] Harbin Medical University Cancer Hospital, Department of UltrasoundHarbin, China. [Cheng, Wen] Harbin Medical University Cancer Hospital, Department of UltrasoundHarbin, China. RP Cheng, W (reprint author), Harbin Medical University Cancer Hospital, Department of Ultrasound, Harbin, China. EM chengwen@hrbmu.edu.cn CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Neoptolemos JP, Kleeff J, Michl P, Costello E, Greenhalf W, Palmer DH. Therapeutic developments in pancreatic cancer: Current and future perspectives. Nat Rev Gastroenterol Hepatol, 2018, 15(6):333–48., DOI 10.1038/s41575-018- 0005-x Hackert T, Klaiber U, Pausch T, Mihaljevic AL, Buchler MW. Fifty years of surgery for pancreatic cancer. Pancreas, 2020, 49(8):1005–13., DOI 10.1097/MPA. 0000000000001634 McGuigan A, Kelly P, Turkington RC, Jones C, Coleman HG, McCain RS. Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J Gastroenterol, 2018, 24(43):4846–61., DOI 10.3748/wjg.v24.i43. 4846 McGuire S. World cancer report 2014. Geneva, Switzerland: World health organization, international agency for research on cancer, WHO press, 2015. Adv Nutr, 2016, 7(2):418–9., DOI 10.3945/an.116.012211 David M, Lepage C, Jouve JL, Jooste V, Chauvenet M, Faivre J, et al. Management and prognosis of pancreatic cancer over a 30-year period. Br J Cancer, 2009, 101(2):215–8., DOI 10.1038/sj.bjc.6605150 Hayashibe A, Kameyama M, Shinbo M, Makimoto S. Clinical results on intraarterial adjuvant chemotherapy for prevention of liver metastasis following curative resection of pancreatic cancer. Ann Surg Oncol, 2007, 14(1):190–4., DOI 10.1245/ s10434-006-9110-0 Horn SR, Stoltzfus KC, Lehrer EJ, Dawson LA, Tchelebi L, Gusani NJ, et al. Epidemiology of liver metastases. Cancer Epidemiol, 2020, 67:101760., DOI 10.1016/ j.canep.2020.101760 Paiella S, De Pastena M, Romeo F, D’onofrio M, Fontana M, Pea A, et al. Ablation treatments in unresectable pancreatic cancer. Minerva Chir, 2019, 74(3): 263–9., DOI 10.23736/S0026-4733.18.07881-1 Hua YQ, Wang P, Zhu XY, Shen YH, Wang K, Shi WD, et al. Radiofrequency ablation for hepatic oligometastatic pancreatic cancer: An analysis of safety and efficacy. Pancreatology, 2017, 17(6):967–73., DOI 10. 1016/j.pan.2017.08.072 Andreou A, Knitter S, Klein F, Malinka T, Schmelzle M, Struecker B, et al. The role of hepatectomy for synchronous liver metastases from pancreatic adenocarcinoma. Surg Oncol, 2018, 27(4):688–94., DOI 10.1016/j.suronc.2018. 09.004 Macedo FI, Ryon E, Maithel SK, Lee RM, Kooby DA, Fields RC, et al. Survival outcomes associated with clinical and pathological response following neoadjuvant FOLFIRINOX or gemcitabine/nab-paclitaxel chemotherapy in resected pancreatic cancer. Ann Surg, 2019, 270(3):400–13., DOI 10.1097/SLA. 0000000000003468 Akahori T, Sho M, Tanaka T, Nishiofuku H, Kinoshita S, Nagai M, et al. Significant efficacy of new transcatheter arterial chemoembolization technique for hepatic metastases of pancreatic neuroendocrine tumors. Anticancer Res, 2013, 33(8):3355–8., DOI 10.3109/0284186X.2013.806820 Hlavsa J, Prochazka V, Andrasina T, Pavlik T, Penka I, Kala Z. Radiofrequency ablation in pancreatic cancer. Rozhl Chir, 2019, 98(11):441–9., DOI 10.33699/PIS.2019.98.11.441-449 de Geus SWL, Eskander MF, Kasumova GG, Ng SC, Kent TS, Mancias JD, et al. Stereotactic body radiotherapy for unresected pancreatic cancer: A nationwide review. Cancer, 2017, 123(21):4158–67., DOI 10.1002/cncr.30856 Wellenstein MD, Coffelt SB, Duits DEM, van Miltenburg MH, Slagter M, de Rink I, et al. Loss of p53 triggers WNT-dependent systemic inflammation to drive breast cancer metastasis. Nature, 2019, 572(7770):538–42., DOI 10.1038/s41586- 019-1450-6 Cupp MA, Cariolou M, Tzoulaki I, Aune D, Evangelou E, Berlanga-Taylor AJ. Neutrophil to lymphocyte ratio and cancer prognosis: An umbrella review of systematic reviews and meta-analyses of observational studies. BMC Med, 2020, 18(1):360., DOI 10.1186/s12916-020-01817-1 Kasahara K, Ishizaki T, Enomoto M, Mazaki J, Okazaki N, Tago T, et al. Preoperative lymphocyte-to-monocyte ratio in the prognostication of advanced resectable colon cancer: A retrospective observational study. Indian J Surg Oncol, 2021, 12(3):498–506., DOI 10.1007/s13193-021- 01356-y Johnson PJ, Berhane S, Kagebayashi C, Satomura S, Teng M, Reeves HL, et al. Assessment of liver function in patients with hepatocellular carcinoma: A new evidence-based approach-the ALBI grade. J Clin Oncol, 2015, 33(6):550–8., DOI 10. 1200/JCO.2014.57.9151 Lee HG, Lim SB, Lee JL, Kim CW, Yoon YS, Park IJ, et al. Preoperative albumin-bilirubin score as a prognostic indicator in patients with stage III colon cancer. Sci Rep, 2022, 12(1):14910., DOI 10.1038/s41598-022-19329-8 Zhang J, Xu Q, Zhang H, Zhang Y, Yang Y, Luo H, et al. High preoperative albumin-bilirubin score predicts poor survival in patients with newly diagnosed high-grade gliomas. Transl Oncol, 2021, 14(4):101038., DOI 10.1016/j.tranon.2021. 101038 Park HJ, Seo KI, Kim SJ, Lee SU, Yun BC, Han BH, et al. Effectiveness of albumin-bilirubin score as a predictor of post-hepatectomy liver failure in patients with hepatocellular carcinoma. Korean J Gastroenterol, 2021, 77(3):115–22., DOI 10. 4166/kjg.2020.148 Gupta GP, Massague J. Cancer metastasis: Building a framework. Cell, 2006, 127(4):679–95., DOI 10.1016/j.cell.2006.11.001 Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science, 2011, 331(6024):1559–64., DOI 10.1126/science.1203543 Clark AM, Ma B, Taylor DL, Griffith L, Wells A. Liver metastases: Microenvironments and ex-vivo models. Exp Biol Med, Maywood,, 2016, 241(15):1639–52., DOI 10.1177/1535370216658144 Mielgo A, Schmid MC. Liver tropism in cancer: The hepatic metastatic niche. Cold Spring Harb Perspect Med, 2020, 10(3):a037259., DOI 10.1101/cshperspect. a037259 Ma R, Feng Y, Lin S, Chen J, Lin H, Liang X, et al. Mechanisms involved in breast cancer liver metastasis. J Transl Med, 2015, 13:64., DOI 10.1186/s12967-015- 0425-0 Engstrand J, Nilsson H, Stromberg C, Jonas E, Freedman J. Colorectal cancer livermetastases - a population-based study on incidence, management and survival. BMC Cancer, 2018, 18(1):78., DOI 10.1186/s12885-017-3925-x Riihimaki M, Hemminki A, Sundquist K, Sundquist J, Hemminki K. The epidemiology of metastases in neuroendocrine tumors. Int J Cancer, 2016, 139(12): 2679–86., DOI 10.1002/ijc.30400 van Roekel C, Jonges TGN, Lock TMTW. What are the odds? Prostate metastases to ureter and peritoneum. BMJ Case Rep, 2018, 2018:bcr2018225730., DOI 10.1136/bcr-2018-225730 Kawahara K, MakinoH, Kametaka H, Hoshino I, Fukada T, Seike K, et al. Outcomes of surgical resection for gastric cancer liver metastases: A retrospective analysis. World J Surg Oncol, 2020, 18(1):41., DOI 10.1186/ s12957-020-01816-9 Rowcroft A, Loveday BPT, Thomson BNJ, Banting S, Knowles B. Systematic review of liver directed therapy for uveal melanoma hepatic metastases. HPB, Oxford,, 2020, 22(4):497–505., DOI 10.1016/j.hpb.2019.11.002 Saade Lemus P, Anderson K, Smith M, Bullock A. Spontaneous regression of pancreatic cancer with liver metastases. BMJ Case Rep, 2019, 12(5):e229619., DOI 10. 1136/bcr-2019-229619 Gilbert BK. The 26th annual bowditch lecture: New computer technologies and their potential for expanded vistas in biomedicine. Physiologist, 1982, 25(1):2–18. Stoop TF, Ateeb Z, Ghorbani P, Scholten L, Arnelo U, Besselink MG, et al. Surgical outcomes after total pancreatectomy: A high-volume center experience. Ann Surg Oncol, 2021, 28(3):1543–51., DOI 10.1245/s10434-020-08957-x Seufferlein T, Bachet JB, Van Cutsem E, Rougier PESMO Guidelines Working Group. Pancreatic adenocarcinoma: ESMO-ESDO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol, 2012, 23(7):vii33–40., DOI 10.1093/ annonc/mds224 Alemi F, Alseidi A, Scott Helton W, Rocha FG. Multidisciplinary management of locally advanced pancreatic ductal adenocarcinoma. Curr Probl Surg, 2015, 52(9):362–98., DOI 10.1067/j.cpsurg.2015.07.003 KhushmanM, Dempsey N, Maldonado JC, Loaiza-Bonilla A, Velez M, Carcas L, et al. Full dose neoadjuvant FOLFIRINOX is associated with prolonged survival in patients with locally advanced pancreatic adenocarcinoma. Pancreatology, 2015, 15(6):667–73., DOI 10.1016/j.pan.2015.08.010 Syed AR, Carleton NM, Horne Z, Dhawan A, Bedi G, Kochhar G, et al. Survival trends for resectable pancreatic cancer using a multidisciplinary conference: The impact of post-operative chemotherapy. J Gastrointest Cancer, 2020, 51(3):836–43., DOI 10.1007/s12029-019-00303-z Wang H, Ding W, Shi H, Bao H, Lu Y, Jiang TA. Combination therapy with low-frequency ultrasound irradiation and radiofrequency ablation as a synergistic treatment for pancreatic cancer. Bioengineered, 2021, 12(2):9832–46., DOI 10.1080/ 21655979.2021.1995581 Deng Q, He M, Fu C, Feng K, Ma K, Zhang L. Radiofrequency ablation in the treatment of hepatocellular carcinoma. Int J Hyperthermia, 2022, 39(1):1052–63., DOI 10.1080/02656736.2022.2059581 Zhang TN, Yin RH, Wang LW. The prognostic and predictive value of the albumin-bilirubin score in advanced pancreatic cancer. Medicine, Baltimore,, 2020, 99(28):e20654., DOI 10.1097/MD.0000000000020654 Sakin A, Sahin S, Sakin A, Atci MM, Yasar N, Arici S, et al. Assessment of pretreatment albumin-bilirubin grade in pancreatic cancer patients with liver metastasis. J BUON, 2020, 25(4):1941–6. El Nakeeb A, El Shobary M, El Dosoky M, Nabeh A, El Sorogy M, El Eneen AA, et al. Prognostic factors affecting survival after pancreaticoduodenectomy for pancreatic adenocarcinoma, single center experience). Hepatogastroenterology, 2014, 61(133):1426–38., DOI 10.5754/hge13579 Qi Q, Zhuang L, Shen Y, Geng Y, Yu S, Chen H, et al. A novel systemic inflammation response index, SIRI, for predicting the survival of patients with pancreatic cancer after chemotherapy. Cancer, 2016, 122(14):2158–67., DOI 10. 1002/cncr.30057 Wang Y, Li C,WangW,Wang J, Li J, Qian S, et al. Serum albumin to globulin ratio is associated with the presence and severity of inflammatory bowel disease. J Inflamm Res, 2022, 15:1907–20., DOI 10.2147/JIR.S347161 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611175 EP 1611188 DI 10.3389/pore.2023.1611175 PG 14 ER PT J AU Zhang, G Hao, Y Chen, L Li, Z Gao, L Tian, J Qiao, Q Zhang, J AF Zhang, Guangwen Hao, Yongfei Chen, Ling Li, Zengshan Gao, Langlang Tian, Jian Qiao, Qing Zhang, Jinsong TI Expression of aquaporin 1, 3 and 5 in colorectal carcinoma: correlation with clinicopathological characteristics and prognosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE biomarker; prognosis; clinical features; colorectal cancer; aquaporin ID biomarker; prognosis; clinical features; colorectal cancer; aquaporin AB Background: Prognostic biomarkers in colorectal carcinoma (CRC) have an important role in therapeutic strategy. Studies have shown that high expression of Aquaporin (AQP) is associated with poor prognosis in a variety of human tumors. AQP is involved in the initiation and development of CRC. The present study aimed to investigate the correlation between the expression of AQP1, 3 and 5 and clinicopathological features or prognosis in CRC. Methods: The AQP1, 3 and 5 expressions were analyzed based on the immunohistochemical staining of tissue microarray specimens including 112 patients with CRC between June 2006 and November 2008. The expression score of AQP (Allred_score and H_score) was digitally obtained with Qupath software. Patients were divided into high or low expression subgroups based on the optimal cut-off values. The relationship between expression of AQP and clinicopathological characteristics were evaluated using chi-square test, t-test, or one-way ANOVA, when appropriate. Survival analysis of 5-year progression free survival (PFS) and overall survival (OS) was performed with timedependent ROC, Kaplan-Meier curves, univariate and multivariate COX analysis. Results: The AQP1, 3 and 5 expressions were associated with regional lymph node metastasis, histological grading, and tumor location in CRC, respectively (p < 0.05). Kaplan-Meier curves showed that patients with high AQP1 expression had worse 5-year PFS than those with low AQP1 expression (Allred_score: 47% vs. 72%, p = 0.015; H_score: 52% vs. 78% p = 0.006), as well as 5-year OS (Allred_score: 51% vs. 75%, p = 0.005; H_score: 56% vs. 80%, p = 0.002). Multivariate Cox regression analysis indicated that AQP1 expression was an independent risk prognostic factor (p = 0.033, HR = 2.274, HR95% CI: 1.069–4.836). There was no significant correlation between the expression of AQP3 and 5 and the prognosis. Conclusion: The AQP1, 3 and 5 expressions correlate with different clinicopathological characteristics and the AQP1 expression may be a potential biomarker of prognosis in CRC. C1 [Zhang, Guangwen] Fourth Military Medical University, Xijing Hospital, Department of RadiologyXi’an, Shaanxi, China. [Hao, Yongfei] Fourth Military Medical University, Xijing Hospital, Department of RadiologyXi’an, Shaanxi, China. [Chen, Ling] Fourth Military Medical University, Xijing Hospital, Department of PathologyXi’an, Shaanxi, China. [Li, Zengshan] Fourth Military Medical University, Xijing Hospital, Department of PathologyXi’an, Shaanxi, China. [Gao, Langlang] Fourth Military Medical University, Xijing Hospital, Department of RadiologyXi’an, Shaanxi, China. [Tian, Jian] Fourth Military Medical University, Xijing Hospital, Department of RadiologyXi’an, Shaanxi, China. [Qiao, Qing] Fourth Military Medical University, Tangdu Hospital, Department of general surgeryXi’an, Shaanxi, China. [Zhang, Jinsong] Fourth Military Medical University, Xijing Hospital, Department of RadiologyXi’an, Shaanxi, China. RP Zhang, J (reprint author), Fourth Military Medical University, Xijing Hospital, Department of Radiology, Xi’an, China. EM stspine@163.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Bender U, Rho YS, Barrera I, Aghajanyan S, Acoba J, Kavan P. Adjuvant therapy for stages II and III colon cancer: Risk stratification, treatment duration, and future directions. Curr Oncol, 2019, 26:S43–s52., DOI 10.3747/co.26.5605 Siegel RL, Miller KD, Sauer AG, Fedewa SA, Butterly LF, Anderson JC, et al. Colorectal cancer statistics, 2020. Ca-a Cancer J Clinicians, 2020, 70:145–64., DOI 10. 3322/caac.21601 Malki A, Abu ElRuz R, Gupta I, Allouch A, Vranic S, Al Moustafa AE. Molecular mechanisms of colon cancer progression and metastasis: Recent insights and advancements. Int J Mol Sci, 2021, 22:130., DOI 10.3390/ijms22010130 Bregni G, Telli TA, Camera S, Deleporte A, Moretti L, Bali AM, et al. Adjuvant chemotherapy for rectal cancer: Current evidence and recommendations for clinical practice. Cancer Treat Rev, 2020, 83:101948., DOI 10.1016/j.ctrv.2019.101948 Ribatti D, Ranieri G, Annese T, Nico B. Aquaporins in cancer. Biochim Biophys Acta, 2014, 1840:1550–3., DOI 10.1016/j.bbagen.2013.09.025 Zannetti A, Benga G, Brunetti A, Napolitano F, Avallone L, Pelagalli A. Role of aquaporins in the physiological functions ofmesenchymal stem cells. Cells, 2020, 9: 2678., DOI 10.3390/cells9122678 Prata C, Hrelia S, Fiorentini D. Peroxiporins in cancer. Int J Mol Sci, 2019, 20: 1371., DOI 10.3390/ijms20061371 Moon CS, Moon D, Kang SK. Aquaporins in cancer biology. Front Oncol, 2022, 12:782829., DOI 10.3389/fonc.2022.782829 Papadopoulos MC, Saadoun S. Key roles of aquaporins in tumor biology. Biochim Biophys Acta-Biomembranes, 2015, 1848:2576–83., DOI 10.1016/j.bbamem. 2014.09.001 Morrissey JJ, Mellnick VM, Luo J, Siegel MJ, Figenshau RS, Bhayani S, et al. Evaluation of urine aquaporin-1 and perilipin-2 concentrations as biomarkers to screen for renal cell carcinoma A prospective cohort study. Jama Oncol, 2015, 1: 204–12., DOI 10.1001/jamaoncol.2015.0213 Kushwaha PP, Verma S, Gupta S. Aquaporins as prognostic biomarker in prostate cancer. Cancers, Basel,, 2023, 15:331., DOI 10.3390/cancers15020331 Rodrigues C, Milkovic L, Bujak IT, Tomljanovic M, Soveral G, Cipak Gasparovic A. Lipid profile and aquaporin expression under oxidative stress in breast cancer cells of different malignancies. Oxid Med Cell Longev, 2019, 2019: 2061830., DOI 10.1155/2019/2061830 Chen G, Song H, Yang Z, Du T, Zheng Y, Lu Z, et al. AQP5 is a novel prognostic biomarker in pancreatic adenocarcinoma. Front Oncol, 2022, 12: 890193., DOI 10.3389/fonc.2022.890193 Kao YC, Jheng JR, Pan HJ, Liao WY, Lee CH, Kuo PL. Elevated hydrostatic pressure enhances the motility and enlarges the size of the lung cancer cells through aquaporin upregulation mediated by caveolin-1 and ERK1/2 signaling. Oncogene, 2017, 36:863–74., DOI 10.1038/onc.2016.255 Yoshida T, Hojo S, Sekine S, Sawada S, Okumura T,Nagata T, et al. Expression of aquaporin-1 is a poor prognostic factor for stage II and III colon cancer. Mol Clin Oncol, 2013, 1:953–8., DOI 10.3892/mco.2013.165 Kang BW, Kim JG, Lee SJ, Chae YS, Jeong JY, Yoon GS, et al. Expression of aquaporin-1, aquaporin-3, and aquaporin-5 correlates with nodal metastasis in colon cancer. Oncology, 2015, 88:369–76., DOI 10.1159/000369073 Imaizumi H, Ishibashi K, Takenoshita S, Ishida H. Aquaporin 1 expression is associated with response to adjuvant chemotherapy in stage II and III colorectal cancer. Oncol Lett, 2018, 15:6450–6., DOI 10.3892/ol.2018.8170 Zhang G, Ma W, Dong H, Shu J, Hou W, Guo Y, et al. Based on histogram analysis: ADC(aqp, derived from ultra-high b-value DWI could be a non-invasive specific biomarker for rectal cancer prognosis. Sci Rep, 2020, 10:10158., DOI 10.1038/ s41598-020-67263-4 Moosavi MS, Elham Y. Aquaporins 1, 3 and 5 in different tumors, their expression, prognosis value and role as new therapeutic targets. Pathol Oncol Res, 2020, 26:615–25., DOI 10.1007/s12253-019-00646-9 Harvey JM, Clark GM, Osborne CK, Allred DC. Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol, 1999, 17: 1474–81., DOI 10.1200/jco.1999.17.5.1474 McCarty KS, Jr, Miller LS, Cox EB, Konrath J, McCarty KS. Estrogen receptor analyses. Correlation of biochemical and immunohistochemical methods using monoclonal antireceptor antibodies. Arch Pathol Lab Med, 1985, 109:716–21. Ribatti D, Ranieri G, Annese T, Nico B. Aquaporins in cancer. Biochim Biophys Acta-General Subjects, 2014, 1840:1550–3., DOI 10.1016/j.bbagen.2013. 09.025 Tomita Y, Dorward H, Yool AJ, Smith E, Townsend AR, Price TJ, et al. Role of aquaporin 1 signalling in cancer development and progression. Int J Mol Sci, 2017, 18:299., DOI 10.3390/ijms18020299 Meng FB, Rui YF, Xu LL, Wan C, Jiang XH, Li G. Aqp1 enhances migration of bone marrow mesenchymal stem cells through regulation of FAK and beta-catenin. Stem Cell Development, 2014, 23:66–75., DOI 10.1089/scd.2013.0185 Clevers H, Nusse R. Wnt/β-catenin signaling and disease. Cell, 2012, 149: 1192–205., DOI 10.1016/j.cell.2012.05.012 Tetsu O, McCormick F. Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature, 1999, 398:422–6., DOI 10.1038/18884 Marlar S, Jensen HH, Login FH, Nejsum LN. Aquaporin-3 in cancer. Int JMol Sci, 2017, 18:2106., DOI 10.3390/ijms18102106 Li A, Lu DH, Zhang YP, Li J, Fang Y, Li F, et al. Critical role of aquaporin-3 in epidermal growth factor-induced migration of colorectal carcinoma cells and its clinical significance. Oncol Rep, 2013, 29:535–40., DOI 10.3892/or.2012.2144 Shan T, Cui X, Li W, LinW, Li Y. AQP5: A novel biomarker that predicts poor clinical outcome in colorectal cancer. Oncol Rep, 2014, 32:1564–70., DOI 10.3892/or. 2014.3377 Humphries MP, Maxwell P, Salto-Tellez M, QuPath: The global impact of an open source digital pathology system. Comput Struct Biotechnol J, 2021, 19:852–9., DOI 10.1016/j.csbj.2021.01.022 Roberti MP, Yonekura S, Duong CPM, Picard M, Ferrere G, Alou MT, et al. Chemotherapy-induced ileal crypt apoptosis and the ileal microbiome shape immunosurveillance and prognosis of proximal colon cancer. Nat Med, 2020, 26:919–31., DOI 10.1038/s41591-020-0882-8 Choudhury SR, Babes L, Rahn JJ, Ahn BY, Goring KAR, King JC, et al. Dipeptidase-1 is an adhesion receptor for neutrophil recruitment in lungs and liver. Cell, 2019, 178:1205–21., DOI 10.1016/j.cell.2019.07.017 Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science, 2020, 368:973–80., DOI 10.1126/science.aay9189 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611179 EP 1611188 DI 10.3389/pore.2023.1611179 PG 10 ER PT J AU Tan, S Nemeth, P AF Tan, Songwen Nemeth, Peter TI Editorial: In vivo and in vitro models for research in pathology SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Editorial DE analytical pathology; pathogenesis; tumor diagnosis; in vivo techniques; molecular pathology ID analytical pathology; pathogenesis; tumor diagnosis; in vivo techniques; molecular pathology C1 [Tan, Songwen] Central South University, Xiangya School of Pharmaceutical SciencesChangsha, China. [Nemeth, Peter] University of Pecs, Faculty of Medicine, Department of Immunology and BiotechnologyPecs, Hungary. RP Tan, S (reprint author), Central South University, Xiangya School of Pharmaceutical Sciences, Changsha, China. EM stan0309@uni.sydney.edu.au CR Liu Q, Shi Y, Cai J, Duan Y,Wang R, Zhang H, et al. Pathological changes in the lungs and lymphatic organs of 12 COVID-19 autopsy cases. Natl Sci Rev, 2020, 7(12):1868–78., DOI 10.1093/nsr/nwaa247 Fernandez MA, Bah F, Ma L, Lee Y, Schmidt M, Welch E, et al. Loss of endosomal exchanger NHE6 leads to pathological changes in tau in human neurons. Stem Cel Rep, 2022, 17(9):2111–26., DOI 10.1016/j.stemcr.2022. 08.001 Tavares de Sousa M, Hecher K, Yamamura J, Kording F, Ruprecht C, Fehrs K, et al. Dynamic fetal cardiac magnetic resonance imaging in four-chamber view using Doppler ultrasound gating in normal fetal heart and in congenital heart disease: Comparison with fetal echocardiography. Ultrasound Obstet Gynecol, 2019, 53(5):669–75., DOI 10.1002/uog.20167 Na S, Russin JJ, Lin L, Yuan X, Hu P, Jann KB, et al. Massively parallel functional photoacoustic computed tomography of the human brain. Nat Biomed Eng, 2022, 6(5):584–92., DOI 10.1038/s41551-021-00735-8 Noor MBT, Zenia NZ, Kaiser MS, Mamun SA, Mahmud M. Application of deep learning in detecting neurological disorders from magnetic resonance images: A survey on the detection of alzheimer’s disease, Parkinson’s disease and schizophrenia. Brain Inform, 2020, 7:11–21., DOI 10.1186/s40708-020-00112-2 Rabaan A, Mutair A, Alawi Z, Alhumaid S, Mohaini M, Aldali J, et al. Comparative pathology, molecular pathogenicity, immunological features, and genetic characterization of three highly pathogenic human coronaviruses, MERS-CoV, SARS-CoV, and SARS-CoV-2). Eur Rev Med Pharmacol Sci, 2021, 25(22):7162–84., DOI 10.26355/eurrev_202111_27270 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611196 EP 1611198 DI 10.3389/pore.2023.1611196 PG 3 ER PT J AU Zhang, L Yuan, P Cao, Q Mu, J Ying, J Guo, Ch AF Zhang, Letian Yuan, Pei Cao, Qi Mu, Jiali Ying, Jianming Guo, Changyuan TI Case report: A rare case of tumor-to-tumor metastasis: metastatic lobular breast carcinoma to clear cell renal cell carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE breast cancer; clear cell renal cell carcinoma; tumor-to-tumor metastasis; rare case; metastatic lobular breast carcinoma ID breast cancer; clear cell renal cell carcinoma; tumor-to-tumor metastasis; rare case; metastatic lobular breast carcinoma AB Tumor-to-tumor metastasis is a rare phenomenon. Although renal cell carcinoma is the most common recipient tumor, metastatic lobular breast carcinoma to clear cell renal cell carcinoma is even rarer, with only one case reported to date. We present a 66-year-old female patient with an invasive lobular carcinoma history who was admitted to the hospital with a right renal mass. The patient received partial nephrectomy. The final established diagnosis is lobular breast carcinoma metastasizing to clear cell renal cell carcinoma (ccRCC). Thus, although rare, the simultaneous or consecutive find of a renal mass in follow-up should be carefully evaluated, especially in high-risk patients, including women with an advanced breast cancer history, as in this scenario. C1 [Zhang, Letian] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Yuan, Pei] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Cao, Qi] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Mu, Jiali] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Ying, Jianming] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Guo, Changyuan] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. RP Guo, Ch (reprint author), Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of Pathology, Beijing, China. EM guocycicams@sina.com CR Richardson JF, Katayama I. Neoplasm to neoplasm metastasis. An acidophil adenoma harbouring metastatic carcinoma: A case report. Arch Pathol, 1971, 91: 135–9. Moller MG, Gribbin T, Ebrom S, Padula G, Fitzgerald TL. Breast cancer metastatic to renal cell carcinoma. Surgery, 2006, 139:577–9., DOI 10.1016/j.surg. 2005.10.016 Ricketts R, Tamboli P, Czerniak B, Guo CC. Tumor-to-Tumor metastasis: Report of 2 cases of metastatic carcinoma to angiomyolipoma of the kidney. Arch Pathol Lab Med, 2008, 132:1016–20., DOI 10.1043/1543-2165(2008)132[1016: TMROCO]2.0.CO;2 Chen TD, Lee LY. A case of renal cell carcinoma metastasizing to invasive ductal breast carcinoma. J Formos Med Assoc, 2014, 113:133–6., DOI 10.1016/j.jfma. 2012.07.022 Ashman D, Quiroga-Garza G, Lee D. Rare presentation of metastatic lobular breast carcinoma involving clear cell renal cell carcinoma. Case Rep Oncol Med, 2020, 2020:5315178., DOI 10.1155/2020/5315178 Campbell LV, Jr, Gilbert E, Chamberlain CR, Jr, Watne AL. Metastases of cancer to cancer. Cancer, 1968, 22:635–43., DOI 10.1002/1097-0142(196809)22: 3<635::aid-cncr2820220320>3.0.co;2-o Pamphlett R. Carcinoma metastasis to meningioma. J Neurol Neurosurg Psychiatry, 1984, 47:561–3., DOI 10.1136/jnnp.47.5.561 Takei H, Powell SZ. Tumor-to-tumor metastasis to the central nervous system. Neuropathology, 2009, 29:303–8., DOI 10.1111/j.1440-1789.2008.00952.x Huo Z, Gao Y, Yu Z, Zuo W, Zhang Y. Metastasis of breast cancer to renal cancer: Report of a rare case. Int J Clin Exp Pathol, 2015, 8:15417–21. eCollection 2015. Sella A, Ro JY. Renal cell cancer: Best recipient of tumor-to-tumor metastasis. Urology, 1987, 30:35–8., DOI 10.1016/0090-4295(87)90568-1 Beisland C, Talleraas O, Bakke A, Norstein J. Multiple primary malignancies in patients with renal cell carcinoma: A national population-based cohort study. BJU Int, 2006, 97:698–702., DOI 10.1111/j.1464-410X.2006.06004.x Granville LA, Ostrowski ML, Truong LD, Shen S. Pathologic quiz case: Unusual morphology in an otherwise classic renal cell carcinoma. In: Tumor-totumor metastasis: Pulmonary adenocarcinoma metastatic to clear cell renal cell carcinoma, 2005). Fidler IJ, Poste G. The "seed and soil" hypothesis revisited. Lancet Oncol, 2008, 9:808., DOI 10.1016/S1470-2045(08)70201-8 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611204 EP 1611207 DI 10.3389/pore.2023.1611204 PG 4 ER PT J TI Tumor budding as a predictor of disease-free survival in patients with cholangiocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; cholangiocarcinoma; tumor budding; prediction; disease-free survival ID prognosis; cholangiocarcinoma; tumor budding; prediction; disease-free survival AB Background: Tumor budding is considered a prognostic factor in several solid cancer types. However, we lack comprehensive information on the importance of tumor budding in cholangiocarcinoma. Therefore, we aimed to assess the prognostic value of tumor budding in intrahepatic and extrahepatic cholangiocarcinomas and to evaluate its correlations with other clinicopathological parameters. Methods: We monitored 219 patients who underwent surgery for intrahepatic or extrahepatic cholangiocarcinoma at the Pusan National University Hospital between 2012 and 2021. Tumor budding was evaluated using the International Tumor Budding Consensus Conference scoring system. Tumor budding was classified into low (0–4), intermediate (5–9), and high (≥10). For statistical analysis, tumor budding was divided into two groups based on the cut-off value of 10 (lower: 0–9 vs. higher: ≥10). The correlations between clinicopathological parameters were examined using the chi-square and Fisher’s exact test. The prognostic values of the variables were analyzed using the log-rank test and Cox regression analysis. Results: Low, intermediate, and high tumor buddings were identified in 135 (61.6%), 63 (28.8), and 21 (9.6%), patients, respectively. Higher tumor budding was related to the presence of lymphatic invasion (p = 0.017), higher tumor grade (p = 0.001), higher N category (p = 0.034). In the univariable and multivariable analyses, higher tumor budding was associated with shorter disease-free survival in 97 (44.3%) patients who underwent R0 resection (p < 0.001 and p = 0.011). Tumor budding did not significantly correlate with disease-specific survival in entire patients. Conclusion: Tumor budding may serve as a prognostic factor for intrahepatic and extrahepatic cholangiocarcinomas treated with R0 resection. CR Khan SA, Tavolari S, Brandi G. Cholangiocarcinoma: Epidemiology and risk factors. Liver Int, 2019, 39:19–31., DOI 10.1111/liv.14095 Khan AS, Dageforde LA. Cholangiocarcinoma. Surg Clin North Am, 2019, 99(2):315–35., DOI 10.1016/j.suc.2018.12.004 Ueno H, Murphy J, Jass JR, Mochizuki H, Talbot IC. Tumour ’budding’ as an index to estimate the potential of aggressiveness in rectal cancer. Histopathology, 2002, 40(2):127–32., DOI 10.1046/j.1365-2559.2002.01324.x Imai T. The growth of human carcinoma: A morphological analysis. Fukuoka Igaku Zasshi, 1954, 45:72–102. De Smedt L, Palmans S, Sagaert X. Tumour budding in colorectal cancer: What do we know and what can we do? Virchows Arch, 2016, 468(4):397–408., DOI 10. 1007/s00428-015-1886-5 Lugli A, Kirsch R, Ajioka Y, Bosman F, Cathomas G, Dawson H, et al. Recommendations for reporting tumor budding in colorectal cancer based on the International Tumor Budding Consensus Conference, ITBCC, 2016. Mod Pathol, 2017, 30(9):1299–311., DOI 10.1038/modpathol.2017.46 Karamitopoulou E, Wartenberg M, Zlobec I, Cibin S, Worni M, Gloor B, et al. Tumour budding in pancreatic cancer revisited: Validation of the ITBCC scoring system. Histopathology, 2018, 73(1):137–46., DOI 10.1111/ his.13508 Kim HN, Lee SY, Kim BH, Kim CY, Kim A, Kim H. Prognostic value of tumor budding in gallbladder cancer: Application of the international tumor budding consensus conference scoring system. Virchows Arch, 2021, 478(6):1071–8., DOI 10. 1007/s00428-020-03012-2 Lohneis P, Hieggelke L, Gebauer F, Ball M, Bruns C, Buttner R, et al. Tumor budding assessed according to the criteria of the International Tumor Budding Consensus Conference determines prognosis in resected esophageal adenocarcinoma. Virchows Arch, 2021, 478(3):393–400., DOI 10.1007/s00428- 020-02897-3 Szalai L, Jakab A, Kocsmar I, Szirtes I, Kenessey I, Szijarto A, et al. Prognostic ability of tumor budding outperforms poorly differentiated clusters in gastric cancer. Cancers, Basel,, 2022, 14(19):4731., DOI 10.3390/ cancers14194731 Agostini-Vulaj D, Cates JMM, Bratton LE, Gonzalez RS. Increasing tumor budding in cholangiocarcinoma is associated with decreased diseasespecific survival. Hum Pathol, 2021, 111:75–83., DOI 10.1016/j.humpath. 2021.03.004 Ito T, Kuriyama N, Kozuka Y, Komatsubara H, Ichikawa K, Noguchi D, et al. High tumor budding is a strong predictor of poor prognosis in the resected perihilar cholangiocarcinoma patients regardless of neoadjuvant therapy, showing survival similar to those without resection. BMC Cancer, 2020, 20(1):209., DOI 10.1186/ s12885-020-6695-9 Kosaka H, Ishida M, Ueno M, Komeda K, Hokutou D, Iida H, et al. Tumor budding may be a promising prognostic indicator in intrahepatic cholangiocarcinoma: A multicenter retrospective study. Ann Gastroenterol Surg, 2023, 7(1):138–46., DOI 10.1002/ags3.12605 Nakayama G, Hisaka T, Sakai H, Akashi M, Yuichi G, Sato T, et al. Tumour budding as an independent prognostic factor for survival in patients with distal bile duct cancer. Anticancer Res, 2022, 42(8):4079–87., DOI 10. 21873/anticanres.15905 Ogino M, Nakanishi Y, Mitsuhashi T, Hatanaka Y, Amano T, Marukawa K, et al. Impact of tumour budding grade in 310 patients who underwent surgical resection for extrahepatic cholangiocarcinoma. Histopathology, 2019, 74(6): 861–72., DOI 10.1111/his.13827 Okubo S, Mitsunaga S, Kato Y, KojimaM, SugimotoM, Gotohda N, et al. The prognostic impact of differentiation at the invasive front of biliary tract cancer. J Surg Oncol, 2018, 117(6):1278–87., DOI 10.1002/jso.24946 Regmi P, Paudyal A, Paudyal P, Hu HJ, Liu F, Ma WJ, et al. Prognostic significance of tumor budding in biliary tract cancer. Eur J Surg Oncol, 2022, 48(1): 160–8., DOI 10.1016/j.ejso.2021.08.008 Tanaka M, Yamauchi N, Ushiku T, Shibahara J, Hayashi A, Misumi K, et al. Tumor budding in intrahepatic cholangiocarcinoma: A predictor of postsurgery outcomes. Am J Surg Pathol, 2019, 43(9):1180–90., DOI 10.1097/PAS. 0000000000001332 Yoshizawa T, Hong SM, Jung D, Noe M, Kiemen A, Wu PH, et al. Threedimensional analysis of extrahepatic cholangiocarcinoma and tumor budding. J Pathol, 2020, 251(4):400–10., DOI 10.1002/path.5474 Amin MB, Edge SB, Greene FL, Byrd DR, Brookland RK, Washington MK, et al. AJCC cancer staging manual. 8th ed. New York: Springer, 2017). Cao Z, Livas T, Kyprianou N. Anoikis and EMT: Lethal "liaisons" during cancer progression. Crit Rev Oncog, 2016, 21(3-4):155–68., DOI 10.1615/ CritRevOncog.2016016955 Lugli A, Zlobec I, Berger MD, Kirsch R, Nagtegaal ID. Tumour budding in solid cancers. Nat Rev Clin Oncol, 2021, 18(2):101–15., DOI 10.1038/s41571-020- 0422-y Tanaka M, Kunita A, Yamagishi M, Katoh H, Ishikawa S, Yamamoto H, et al. KRAS mutation in intrahepatic cholangiocarcinoma: Linkage with metastasis-free survival and reduced E-cadherin expression. Liver Int, 2022, 42(10):2329–40., DOI 10.1111/liv.15366 Zlobec I, Bachli M, Galuppini F, Berger MD, Dawson HE, Nagtegaal ID, et al. Refining the ITBCC tumor budding scoring system with a "zero-budding" category in colorectal cancer. Virchows Arch, 2021, 479(6):1085–90., DOI 10.1007/s00428- 021-03090-w NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611216 EP 1611225 DI 10.3389/pore.2023.1611216 PG 10 ER PT J AU Lv, J Chen, P Wu, J Hu, C AF Lv, Jing Chen, Peirui Wu, Jianqiang Hu, Caihong TI Prognostic value of pretreatment Controlling Nutritional Status score in esophageal cancer: a meta-analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE meta-analysis; prognosis; esophageal cancer; pretreatment; Controlling Nutritional Status score ID meta-analysis; prognosis; esophageal cancer; pretreatment; Controlling Nutritional Status score AB Background and purpose: The association between the pretreatment Controlling Nutritional Status (CONUT) score and the prognosis of esophageal cancer patients remains unclear. The aim of this meta-analysis was to further elucidate the prognostic role of the pretreatment CONUT score in esophageal cancer based on current evidence. Methods: The PubMed, Embase, Web of Science and CNKI databases were searched up to 27 September 2022. The primary and secondary outcomes were overall survival (OS) and progression-free survival (PFS)/cancer-specific survival (CSS), and the hazard ratio (HR) and 95% confidence interval (CI) were pooled for analysis. Results: A total of 11 retrospective studies involving 3,783 participants were included. The pooled results demonstrated that a higher pretreatment CONUT score was significantly related to poor OS (HR = 1.82, 95% CI: 1.31–2.54, p < 0.001), and subgroup analysis stratified by pathological type showed similar results. In addition, the pretreatment CONUT score was associated with poor PFS (HR = 1.19, 95% CI: 1.10–1.28, p < 0.001) and CSS (HR = 2.67, 95% CI: 1.77–4.02, p < 0.001). Conclusion: The pretreatment CONUT score was predictive of worse prognosis in esophageal cancer, and patients with a higher CONUT score showed worse survival. C1 [Lv, Jing] People’s Hospital of Deyang City, Department of Thoracic SurgeryDeyang, China. [Chen, Peirui] People’s Hospital of Deyang City, Department of Thoracic SurgeryDeyang, China. [Wu, Jianqiang] People’s Hospital of Deyang City, Department of Thoracic SurgeryDeyang, China. [Hu, Caihong] People’s Hospital of Deyang City, Department of Thoracic SurgeryDeyang, China. RP Hu, C (reprint author), People’s Hospital of Deyang City, Department of Thoracic Surgery, Deyang, China. EM 523161885@qq.com CR Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA: a Cancer J clinicians, 2022, 72(1):7–33., DOI 10.3322/caac.21708 Xia C, Dong X, Li H, Cao M, Sun D, He S, et al. Cancer statistics in China and United States, 2022: Profiles, trends, and determinants. Chin Med J, 2022, 135(5): 584–90., DOI 10.1097/CM9.0000000000002108 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer J clinicians, 2018, 68(6):394–424., DOI 10. 3322/caac.21492 Hursting SD, Berger NA. Energy balance, host-related factors, and cancer progression. J Clin Oncol : official J Am Soc Clin Oncol, 2010, 28(26):4058–65., DOI 10.1200/JCO.2010.27.9935 Norman K, Pichard C, Lochs H, Pirlich M. Prognostic impact of disease-related malnutrition. Clin Nutr, Edinburgh, Scotland,, 2008, 27(1):5–15., DOI 10.1016/j. clnu.2007.10.007 Liao G, Zhao Z, Yang H, Chen M, Li X. Can prognostic nutritional index be a prediction factor in esophageal cancer?: A meta-analysis. Nutr Cancer, 2020, 72(2): 187–93., DOI 10.1080/01635581.2019.1631859 Ma LX, Taylor K, Espin-Garcia O, Anconina R, Suzuki C, Allen MJ, et al. Prognostic significance of nutritional markers in metastatic gastric and esophageal adenocarcinoma. Cancer Med, 2021, 10(1):199–207., DOI 10.1002/cam4.3604 Yu J, Zhang W, Wang C, Hu Y. The prognostic value of pretreatment geriatric nutritional risk index in esophageal cancer: A meta-analysis. Nutr Cancer, 2022, 74(9):3202–10., DOI 10.1080/01635581.2022.2069273 Wang Y, Chen L,Wu Y, Li P, Che G. The prognostic value ofmodified Glasgow prognostic score in patients with esophageal squamous cell cancer: A meta-analysis. Nutr Cancer, 2020, 72(7):1146–54., DOI 10.1080/01635581.2019.1677925 Lopez-Larramona G, Lucendo AJ, Tenias JM. Association between nutritional screening via the Controlling Nutritional Status index and bone mineral density in chronic liver disease of various etiologies. Hepatol Res : official J Jpn Soc Hepatol, 2015, 45(6):618–28., DOI 10.1111/hepr.12395 Iseki Y, ShibutaniM,Maeda K, Nagahara H, OhtaniH, Sugano K, et al. Impact of the preoperative controlling nutritional status, CONUT, score on the survival after curative surgery for colorectal cancer. PloS one, 2015, 10(7):e0132488., DOI 10. 1371/journal.pone.0132488 Yoshida N, Baba Y, Shigaki H, Harada K, Iwatsuki M, Kurashige J, et al. Preoperative nutritional assessment by controlling nutritional status, CONUT, is useful to estimate postoperative morbidity after esophagectomy for esophageal cancer. World J Surg, 2016, 40(8):1910–7., DOI 10.1007/s00268-016-3549-3 Takagi K, Buettner S, Ijzermans JNM. Prognostic significance of the controlling nutritional status, CONUT, score in patients with colorectal cancer: A systematic review and meta-analysis. Int J Surg, London, England,, 2020, 78: 91–6., DOI 10.1016/j.ijsu.2020.04.046 Ma X, Zou W, Sun Y. Prognostic value of pretreatment controlling nutritional status score for patients with pancreatic cancer: Ameta-analysis. Front Oncol, 2021, 11:770894., DOI 10.3389/fonc.2021.770894 Zhang C, Li XK, Cong ZZ, Zheng C, Luo C, Xie K, et al. Controlling nutritional status is a prognostic factor for patients with lung cancer: A systematic review and meta-analysis. Ann Palliat Med, 2021, 10(4): 3896–905., DOI 10.21037/apm-20-2328 Peng L,Meng C, Li J, You C, Du Y, XiongW, et al. The prognostic significance of controlling nutritional status, CONUT, score for surgically treated renal cell cancer and upper urinary tract urothelial cancer: A systematic review and metaanalysis. Eur J Clin Nutr, 2022, 76(6):801–10., DOI 10.1038/s41430-021-01014-0 Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ, Clinical research ed,, 2021, 372:790–9., DOI 10.1016/j. rec.2021.07.010 Takagi K, Buettner S, Ijzermans JNM, Wijnhoven BPL. Systematic review on the controlling nutritional status, CONUT, score in patients undergoing esophagectomy for esophageal cancer. Anticancer Res, 2020, 40(10):5343–9., DOI 10.21873/anticanres.14541 WangY, Li J, Chang S,DongY,CheG. Risk andinfluencing factors for subsequent primary lung cancer after treatment of breast cancer: A systematic review and two metaanalyses based on four million cases. J Thorac Oncol : official Publ Int Assoc Study Lung Cancer, 2021, 16(11):1893–908., DOI 10.1016/j.jtho.2021.07.001 Begg CB, MazumdarM. Operating characteristics of a rank correlation test for publication bias. Biometrics, 1994, 50(4):1088–101., DOI 10.2307/2533446 Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ, Clinical research ed,, 1997, 315(7109): 629–34., DOI 10.1136/bmj.315.7109.629 Wang Y, Huang D, Xu WY, Wang YW, Che GW. Prognostic value of pretreatment lymphocyte-to-monocyte ratio in non-small cell lung cancer: A meta-analysis. Oncol Res Treat, 2019, 42(10):523–31., DOI 10.1159/ 000501726 Toyokawa T, Kubo N, Tamura T, Sakurai K, Amano R, Tanaka H, et al. The pretreatment Controlling Nutritional Status, CONUT, score is an independent prognostic factor in patients with resectable thoracic esophageal squamous cell carcinoma: Results from a retrospective study. BMC cancer, 2016, 16(1):722., DOI 10. 1186/s12885-016-2696-0 Yoshida N, Harada K, Baba Y, Kosumi K, Iwatsuki M, Kinoshita K, et al. Preoperative controlling nutritional status, CONUT, is useful to estimate the prognosis after esophagectomy for esophageal cancer. Langenbeck’s Arch Surg, 2017, 402(2):333–41., DOI 10.1007/s00423-017-1553-1 Hirahara N, Matsubara T, Hayashi H, Takai K, Nakada S, Tajima Y. Prognostic importance of controlling nutritional status in patients undergoing curative thoracoscopic esophagectomy for esophageal cancer. Am J Ther, 2018, 25(5):E524–E532., DOI 10.1097/MJT.0000000000000414 Xu J. Analysis of preoperative nutritional data and prognostic model of esophageal squamous cell carcinoma. Doctor. Hangzhou, China: Zhejiang University, 2018). Hikage M, Taniyama Y, Sakurai T, Sato C, Takaya K, Okamoto H, et al. The influence of the perioperative nutritional status on the survival outcomes for esophageal cancer patients with neoadjuvant chemotherapy. Ann Surg Oncol, 2019, 26(13):4744–53., DOI 10.1245/s10434-019-07742-9 Sakai M, Sohda M, Saito H, Ubukata Y, Nakazawa N, Kuriyama K, et al. Comparative analysis of immunoinflammatory and nutritional measures in surgically resected esophageal cancer: A single-center retrospective study. vivo, Athens, Greece,, 2020, 34(2):881–7., DOI 10.21873/invivo.11853 Suzuki T, Furukawa K, Funasaka K, Ishikawa E, Sawada T, Maeda K, et al. Long-term prognostic predictors of esophageal squamous cell carcinoma potentially indicated for endoscopic submucosal dissection. Digestion, 2021, 102(4):563–71., DOI 10.1159/000510091 Urabe M, Ueno M, Ogawa Y, Yago A, Shimoyama H, Honda A, et al. Comparative analysis of the prognostic utility of preoperative nutritional parameters in patients with resectable esophageal carcinoma. Gen Thorac Cardiovasc Surg, 2021, 69(2):326–35., DOI 10.1007/s11748-020-01555-4 Xu D. Inflammatory and nutritional indicators and circRNA in the prognosis of esophageal cancer. Nanjing, China: Nanjing Medical University, 2021). 硕士. Yoon JP, Nam JS, Abidin M, Kim SO, Lee EH, Choi IC, et al. Comparison of preoperative nutritional indexes for outcomes after primary esophageal surgery for esophageal squamous cell carcinoma. Nutrients, 2021, 13(11):4086., DOI 10.3390/ nu13114086 Chang L, Cheng Q,Ma Y,Wu C, Zhang X, Ma Q, et al. Prognostic effect of the controlling nutritional status score in patients with esophageal cancer treated with immune checkpoint inhibitor. J Immunother, 2022, 45:415–22., DOI 10.1097/CJI. 0000000000000438 Tsunematsu M, Haruki K, Fujiwara Y, Furukawa K, Onda S, Matsumoto M, et al. Preoperative controlling nutritional status, CONUT, score predicts long-term outcomes in patients with non-B non-C hepatocellular carcinoma after curative hepatic resection. Langenbeck’s Arch Surg, 2021, 406(1):99–107., DOI 10.1007/ s00423-020-01987-9 Poulia KA, Yannakoulia M, Karageorgou D, Gamaletsou M, Panagiotakos DB, Sipsas NV, et al. Evaluation of the efficacy of six nutritional screening tools to predict malnutrition in the elderly. Clin Nutr, Edinburgh, Scotland,, 2012, 31(3): 378–85., DOI 10.1016/j.clnu.2011.11.017 Bouillanne O, Morineau G, Dupont C, Coulombel I, Vincent JP, Nicolis I, et al. Geriatric nutritional risk index: A new index for evaluating at-risk elderly medical patients. Am J Clin Nutr, 2005, 82(4):777–83., DOI 10.1093/ajcn/82. 4.777 Loosen SH, Kostev K, Luedde M, Luedde T, Roderburg C. Low blood levels of high-density lipoprotein, HDL, cholesterol are positively associated with cancer. J Cancer Res Clin Oncol, 2022, 148(11):3039–46., DOI 10.1007/ s00432-021-03867-1 Tang Q, Liang B, Zhang L, Li X, Li H, Jing W, et al. Enhanced CHOLESTEROL biosynthesis promotes breast cancer metastasis via modulating CCDC25 expression and neutrophil extracellular traps formation. Scientific Rep, 2022, 12(1):17350., DOI 10.1038/s41598-022-22410-x Tao M, Luo J, Gu T, Yu X, Song Z, Jun Y, et al. LPCAT1 reprogramming cholesterol metabolism promotes the progression of esophageal squamous cell carcinoma. Cell Death Dis, 2021, 12(9):845., DOI 10.1038/s41419-021-04132-6 Li Y, Luo H, Ye B, Zhang K, Liu C, Zu R, et al. Prognostic value of nutritional and inflammatory indicators in females with esophageal squamous cell cancer: A propensity score matching study. Front Genet, 2022, 13:1026685., DOI 10.3389/ fgene.2022.1026685 Harborg S, Zachariae R, Olsen J, Johannsen M, Cronin-Fenton D, Boggild H, et al. Overweight and prognosis in triple-negative breast cancer patients: A systematic review and meta-analysis. NPJ breast cancer, 2021, 7(1):119., DOI 10. 1038/s41523-021-00325-6 Simillis C, Taylor B, Ahmad A, Lal N, Afxentiou T, Powar MP, et al. A systematic review and meta-analysis assessing the impact of body mass index on long-term survival outcomes after surgery for colorectal cancer. Eur J Cancer, 2022, 172:237–51., DOI 10.1016/j.ejca.2022.05.020 Yao L, Wang L, Yin Y, Che G, Yang M. Prognostic value of pretreatment skeletal muscle mass index in esophageal cancer patients: A meta-analysis. Nutr Cancer, 2022, 74(10):3592–600., DOI 10.1080/01635581.2022.2088814 Nishimura E, Kawakubo H, Matsuda S, Fukuda K, Nakamura R, Kitagawa Y. Long-term variation in psoas muscle mass index is affected by short-term loss after esophagectomy in survivors of esophageal cancer. Dis esophagus : official J Int Soc Dis Esophagus, 2022, 36:doac053., DOI 10.1093/dote/doac053 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611221 EP 1611230 DI 10.3389/pore.2023.1611221 PG 10 ER PT J AU Reisz, Z Radics, LB Nemes, P Laxton, R Kaizer, L Gabor, MK Novak, T Barzo, P Al-Sarraj, S Bodi, I AF Reisz, Zita Radics, Laszlo Bence Nemes, Peter Laxton, Ross Kaizer, Laszlo Gabor, Mita Krisztina Novak, Timea Barzo, Pal Al-Sarraj, Safa Bodi, Istvan TI Case Report: Brainstem angiocentric glioma presenting in a toddler child–diagnostic and therapeutic challenges SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE paediatric brainstem glioma; angiocentric glioma; MYB:QKI fusion; DNA methylation profiling; RNA sequencing ID paediatric brainstem glioma; angiocentric glioma; MYB:QKI fusion; DNA methylation profiling; RNA sequencing AB Introduction: Angiocentric gliomas (AG) in brainstem location are exceedingly rare and might cause differential diagnostic problems and uncertainty regarding the best therapeutic approach. Hereby, we describe the clinicopathological findings in a brainstem AG presenting in a toddler child and review the literature. Case report: A 2-year-old boy presented with 5 weeks history of gait disturbances, frequent falls, left-sided torticollis and swallowing problems. MRI head showed a T2-hyperintense, partly exophytic mass lesion centred in the pontomedullary region, raising the possibility of diffuse midline glioma. The exophytic component was partially resected by suboccipital craniotomy, leaving intact the infiltrative component. Ventriculoperitoneal shunt was implanted due to postoperative hydrocephalus. Histological examination revealed a moderately cellular tumour consisted of bland glial cells infiltrating the brain parenchyma and radially arranged around the blood vessels. By immunohistochemistry, the tumour strongly expressed S100 and GFAP in addition to intense nestin positivity, while OLIG2 was negative in the perivascular tumour cells. DNA methylation array profiled the tumour as “methylation class diffuse astrocytoma, MYB or MYBL1-altered subtype B (infratentorial)” and an in-frame MYB::QKI fusion was identified by RNA sequencing, confirming the diagnosis of angiocentric glioma. The patient has been initially treated with angiogenesis inhibitor and mTOR inhibitor, and now he is receiving palliative vinblastine. He is clinically stable on 9 months follow-up. Conclusion: Brainstem AG may cause a diagnostic problem, and the surgical and oncological management is challenging due to unresectability and lack of response to conventional chemo-radiation. In the future, genetically-tailored therapies might improve the prognosis. C1 [Reisz, Zita] King’s College Hospital, Clinical NeuropathologyLondon, UK. [Radics, Laszlo Bence] University of Szeged, Department of PathologySzeged, Hungary. [Nemes, Peter] University of Szeged, Department of NeurosurgerySzeged, Hungary. [Laxton, Ross] King’s College Hospital, Clinical NeuropathologyLondon, UK. [Kaizer, Laszlo] University of Szeged, Department of PathologySzeged, Hungary. [Gabor, Mita Krisztina] University of Szeged, Department of PediatricsSzeged, Hungary. [Novak, Timea] University of Szeged, Department of RadiologySzeged, Hungary. [Barzo, Pal] University of Szeged, Department of NeurosurgerySzeged, Hungary. [Al-Sarraj, Safa] King’s College Hospital, Clinical NeuropathologyLondon, UK. [Bodi, Istvan] King’s College Hospital, Clinical NeuropathologyLondon, UK. RP Bodi, I (reprint author), King’s College Hospital, Clinical Neuropathology, London, UK. EM istvan.bodi@nhs.net CR Chen LH, Pan C, Diplas BH, Xu C, Hansen LJ, Wu Y, et al. The integrated genomic and epigenomic landscape of brainstem glioma. Nat Commun, 2020, 11(1):3077–11., DOI 10.1038/s41467-020-16682-y Ryall S, Tabori U, Hawkins C. Pediatric low-grade glioma in the era of molecular diagnostics. Acta Neuropathol Commun, 2020, 8(1):30–22., DOI 10. 1186/s40478-020-00902-z Ryall S, Zapotocky M, Fukuoka K, Nobre L, Guerreiro Stucklin A, Bennett J, et al. Integrated molecular and clinical analysis of 1,000 pediatric low-grade gliomas. Cancer Cell, 2020, 37(4):569–83., DOI 10.1016/j.ccell.2020.03.011 Wang M, Tihan T, Rojiani AM, Bodhireddy SR, Prayson RA, Iacuone JJ, et al. Monomorphous angiocentric glioma: A distinctive epileptogenic neoplasm with features of infiltrating astrocytoma and ependymoma. J Neuropathol Exp Neurol, 2005, 64(10):875–81., DOI 10.1097/01.jnen.0000182981.02355.10 Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol, 2007, 114(2):97–109., DOI 10.1007/s00401-007-0243-4 Bandopadhayay P, Ramkissoon LA, Jain P, Bergthold G, Wala J, Zeid R, et al. MYB-QKI rearrangements in angiocentric glioma drive tumorigenicity through a tripartite mechanism. Nat Genet, 2016, 48(3):273–82., DOI 10.1038/ng.3500 Qaddoumi I, Orisme W,Wen J, Santiago T, Gupta K, Dalton JD, et al. Genetic alterations in uncommon low-grade neuroepithelial tumors: BRAF, FGFR1, and MYB mutations occur at high frequency and align with morphology. Acta Neuropathol, 2016, 131(6):833–45., DOI 10.1007/s00401-016-1539-z Blumcke I, Aronica E, Becker A, Capper D, Coras R, Honavar M, et al. Lowgrade epilepsy-associated neuroepithelial tumours—The 2016WHO classification. Nat Rev Neurol, 2016, 12(12):732–40., DOI 10.1038/nrneurol.2016.173 Lellouch-Tubiana A, Boddaert N, Bourgeois M, Fohlen M, Jouvet A, Delalande O, et al. Angiocentric neuroepithelial tumor, ANET): A new epilepsy-related clinicopathological entity with distinctive MRI. Brain Pathol, 2005, 15(4):281–6., DOI 10.1111/j.1750-3639.2005.tb00112.x Preusser M, Hoischen A, Novak K, Czech T, Prayer D, Hainfellner JA, et al. Angiocentric glioma: Report of clinico-pathologic and genetic findings in 8 cases. Am J Surg Pathol, 2007, 31(11):1709–18., DOI 10.1097/PAS.0b013e31804a7ebb Chan E, Bollen AW, Sirohi D, Van Ziffle J, Grenert JP, Kline CN, et al. Angiocentric glioma with MYB-QKI fusion located in the brainstem, rather than cerebral cortex. Acta Neuropathol, 2017, 134(4):671–3., DOI 10.1007/s00401-017- 1759-x Covington DB, Rosenblum MK, Brathwaite C, Sandberg DI. Angiocentric glioma-like tumor of the midbrain. Pediatr Neurosurg, 2010, 45:429–33., DOI 10. 1159/000277616 D’Aronco L, Rouleau C, Gayden T, Crevier L, Decarie J-C, Perreault S, et al. Brainstem angiocentric gliomas with MYB-QKI rearrangements. Acta Neuropathol, 2017, 134(4):667–9., DOI 10.1007/s00401-017-1763-1 Weaver KJ, Crawford LM, Bennett J, Rivera-Zengotita ML, Pincus DW. Brainstem angiocentric glioma: Report of 2 cases. J Neurosurg Pediatr, 2017, 20(4): 347–51., DOI 10.3171/2017.5.PEDS16402 Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO classification of tumors of the central nervous system: A summary. Neuro Oncol, 2021, 23(8):1231–51., DOI 10.1093/neuonc/noab106 Han G, Zhang J, Ma Y, Gui Q, Yin S. Clinical characteristics, treatment and prognosis of angiocentric glioma. Oncol Lett, 2020, 20(2):1641–8., DOI 10.3892/ol. 2020.11723 Chiang J, Harreld JH, Tinkle CL, Moreira DC, Li X, Acharya S, et al. A singlecenter study of the clinicopathologic correlates of gliomas with a MYB or MYBL1 alteration. Acta Neuropathol, 2019, 138:1091–2., DOI 10.1007/s00401- 019-02081-1 Almubarak AO, Alahmari A, Al Hindi H, AlShail E. Angiocentric glioma of brainstem. Neurosciences, 2020, 25(5):416–20., DOI 10.17712/nsj.2020.5. 20200026 Kurokawa R, Baba A, Emile P, Kurokawa M, Ota Y, Kim J, et al. Neuroimaging features of angiocentric glioma: A case series and systematic review. J Neuroimaging Off J Am Soc Neuroimaging, 2022, 32(3):389–99., DOI 10. 1111/jon.12983 Capper D, Stichel D, Sahm F, Jones DTW, Schrimpf D, Sill M, et al. Practical implementation of DNA methylation and copy-number-based CNS tumor diagnostics: The heidelberg experience. Acta Neuropathol, 2018, 136(2): 181–210., DOI 10.1007/s00401-018-1879-y Ho AL, Vasudeva SD, Schwartz GK. Abstract 638: The impact of PI3K/Akt pathway inhibition upon the c-myb oncogene. Cancer Res, 2011, 15;638., DOI 10. 1158/1538-7445.AM2011-638 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611231 EP 1611237 DI 10.3389/pore.2023.1611231 PG 7 ER PT J AU Cegledi, A Dolgos, J Fekete, M Gopcsa, L Varkonyi, A Vilimi, B Mikala, G Bodo, I AF Cegledi, Andrea Dolgos, Janos Fekete, Monika Gopcsa, Laszlo Varkonyi, Andrea Vilimi, Beata Mikala, Gabor Bodo, Imre TI Delayed spontaneous remission of acquired factor V inhibitor refractory to immunosuppressive therapy with pregnancy-associated improvement SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE factor V inhibitor; coagulopathy; gestation; bleeding disorder; autoimmune ID factor V inhibitor; coagulopathy; gestation; bleeding disorder; autoimmune AB Introduction: Acquired factor V inhibitor (AFVI) is a rare autoimmune bleeding disorder. The treatment of AFVI is challenging, and patients often require both bleeding control and inhibitor eradication. Methods: We conducted a retrospective analysis of the medical records of a 35-year-old Caucasian woman who presented with severe AFVI-induced bleeding and subsequent immunosuppressive therapy. Results: To provide haemostasis, rFVIIa was given with good efficacy. The patient was treated with various combinations of immunosuppressive regimens over the course of 2.5 years, including plasmapheresis plus immunoglobulins, dexamethasone + rituximab, cyclophosphamide + dexamethasone + rituximab + cyclosporine, cyclosporin + sirolimus + cyclophosphamide + dexamethasone, bortezomib + sirolimus + methylprednisolone, and sirolimus + mycophenolate mofetil. Although these treatment modalities resulted in intermittent partial reversals of AFVI over 2.5 years, eventually the inhibitor became therapy-resistant. However, following the discontinuation of all immunosuppressive therapy, the patient experienced a partial spontaneous remission, which was followed by a pregnancy. During the pregnancy, the FV activity increased to 54% and the coagulation parameters returned to normal levels. The patient underwent Caesarean section without any bleeding complications and delivered a healthy child. Discussion: The use of an activated bypassing agent for bleeding control is effective in patients with severe AFVI. The presented case is unique because the treatment regimens included multiple combinations of immunosuppressive agents. This demonstrates that AFVI patients may undergo spontaneous remission even after multiple courses of ineffective immunosuppressive protocols. Additionally, pregnancy-associated improvement of AFVI is an important finding that warrants further investigation. C1 [Cegledi, Andrea] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Dolgos, Janos] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Fekete, Monika] Semmelweis University, Department of Public HealthBudapest, Hungary. [Gopcsa, Laszlo] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Varkonyi, Andrea] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Vilimi, Beata] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Mikala, Gabor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Bodo, Imre] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. RP Cegledi, A (reprint author), Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Budapest, Hungary. EM ceglediandi@freemail.hu CR Wang X, Qin X, Yu Y,Wang R, Liu X, Ji M, et al. Acquired factor V deficiency in a patient with a urinary tract infection presenting with haematuria followed by multiple haemorrhages with an extremely low level of factor V inhibitor: A case report and review of the literature. Blood Coagul Fibrinolysis, 2017, 28(4):334–41., DOI 10.1097/MBC.0000000000000581 Aliaga JL, de Gracia J, Vidal R, Pico M, Flores P, Sampol G. Acquired factor V deficiency in a patient with pulmonary tuberculosis. Eur Respir J, 1990, 3(1): 109–10., DOI 10.1183/09031936.93.03010109 De Maertelaere E, Castoldi E, VanHaute I,Deeren D, Devreese KM. The interaction of factor V and tissue factor pathway inhibitor in amyeloma patient with acquired factor V deficiency. Haemophilia, 2018, 24(3):e160–3., DOI 10.1111/hae.13492 Foster KN, Kim H, Potter K, Matthews MR, Pressman M, Caruso DM. Acquired factor V deficiency associated with exposure to bovine thrombin in a burn patient. J Burn Care Res, 2010, 31(2):353–60., DOI 10.1097/BCR. 0b013e3181d0f5b4 Quek JKS,Wong WH, Tan CW, Tha MH, Nagarajan C, Lee LH, et al. Acquired factor V deficiency in a patient with myeloma and amyloidosis. Thromb Res, 2018, 164:1–3., DOI 10.1016/j.thromres.2018.01.045 Bennett J, Cunningham MT, Howard C, Hoffmann M, Plapp FV. Acquired factor V inhibitor in the setting of coronavirus disease 2019 infection. Blood Coagul Fibrinolysis, 2021, 32(4):294–7., DOI 10.1097/MBC.0000000000001009 Brickner LA, Scannell KA, Sahud MA. Acquired factor V inhibitor in a patient with acquired human immunodeficiency syndrome. Am J Hematol, 1996, 52(4): 332–3., DOI 10.1002/(SICI)1096-8652(199608)52:4<332:AID-AJH22>3.0.CO;2-E Bruna R, Moia R, Valpreda A, Dosio E, Rolla R, Federici A, et al. An acquired factor V inhibitor induced uncontrolled bleeding in a postsurgery patient. Clin Case Rep, 2021, 9(1):98–101., DOI 10.1002/ccr3.3452 Chartier AR, Hillert CJ, Gill H, Jha P. Acquired factor V inhibitor after antibiotic therapy: A clinical case report and review of the literature. Cureus, 2020, 12(7):e9481., DOI 10.7759/cureus.9481 Chenthilmurugan R, Kodali S, Chenthilmurugan H, Steier W, Schwartz J, Kalavar M. Acquired factor V inhibitor in a patient with gingival squamous cell cancer and tuberculosis. Clin Adv Hematol Oncol, 2008, 6(12):921–7. Chiurazzi F, Tufano A, Esposito M, D’Agostino F, Casoria A, Capasso F, et al. Acquired factor V inhibitor after coronavirus disease 2019, COVID-19). Semin Thromb Hemost, 2021, 48:124–6., DOI 10.1055/s-0041-1735452 Fortuna GMG, Guerra V, Chaaya G. Acquired factor V inhibitor presenting as life threatening bleeding after initiation of apixaban. Haemophilia, 2021, 27(5): e636–e637., DOI 10.1111/hae.14374 Fujita Y, Nakazato T, Ito C, Masuda K, Osada Y, Aisa Y, et al. A rare case of an acquired factor V inhibitor in a patient with myelodysplastic syndrome during azacitidine treatment. Ann Hematol, 2018, 97(10):2009–10., DOI 10.1007/s00277- 018-3348-z Giuffrida G, Markovic U, Nicolosi D, Calafiore V. A rare case of acquired factor V inhibitor, during treatment with dabigatran for chronic atrial fibrillation, successfully treated with bypassing agents. Clin Case Rep, 2021, 9(2):623–8., DOI 10. 1002/ccr3.3586 Hirata H, Sakurai Y, Takeda T, Kasetani T, Morita T. Development of acquired factor V inhibitor after surgical procedure without the use of fibrin tissue adhesives: A case report. Cureus, 2021, 13(1):e12708., DOI 10.7759/cureus.12708 Kirkeby KM, Aronowitz P. Acquired factor V inhibitor: A common and avoidable complication of topical bovine thrombin application. Am J Med, 2005, 118(7):805., DOI 10.1016/j.amjmed.2005.01.049 Li F, Shu K, Liu J, Shen C, Wang X, Zhang Z, et al. Acquired factor V inhibitor with symptoms and titer of inhibitor differences: Report of two cases. Acta Haematol, 2019, 141(3):148–50., DOI 10.1159/000496080 Maughan EO, Wilson J, Wilde JT. Spontaneous resolution of acquired factor V inhibitor associated with ovarian carcinoma. Int J Lab Hematol, 2007, 29(4): 316–9., DOI 10.1111/j.1365-2257.2006.00873.x MiharaM, Ogawa Y, NagasakaM, Kobayashi N, Shimizu H, Shinozawa K, et al. Successful management of acquired factor V inhibitor by monitoring factor V activity, antigen, and inhibitor values during immunosuppressive therapy. Acta Haematol, 2020, 143(5):486–90., DOI 10.1159/000502730 Mima F, Minami R, Asako M, Matsunaga H, Fujita Y, Takimoto Y, et al. Acquired factor V inhibitor complicated with immune thrombocytopenia. Intern Med, 2022, 61(1):91–5., DOI 10.2169/internalmedicine.7173-21 Mitsuhashi T, Takamiya O. Acquired factor V inhibitor-related severe bleeding due to unformed prothrombinase complex. Thromb Res, 2018, 171: 81–3., DOI 10.1016/j.thromres.2018.09.052 Morris CJ, Curry N. Acquired factor V inhibitor in a critically ill patient. Anaesthesia, 2009, 64(9):1014–7., DOI 10.1111/j.1365-2044.2009.05945.x Nakata K, Ueda S, Matsunaga H, Mima F, Ueda H, Yoshioka A, et al. High titer of acquired factor V inhibitor presenting with a pseudo-deficiency of multiple coagulation factors. Intern Med, 2018, 57(3):393–7., DOI 10.2169/internalmedicine. 9150-17 Nozu T, Mita H, Okumura T. Acquired factor V inhibitor in a patient with a mechanical aortic valve under warfarin therapy. Intern Med, 2010, 49(20):2229–33., DOI 10.2169/internalmedicine.49.4042 Pineau-Vincent F, Toulon P, Lemaire P, Laribi K. Acquired factor V inhibitor: Success of steroids in a patient with primary sclerosing cholangitis, ulcero haemorragic rectocolitis, biological Biermer’s disease. Transfus Clin Biol, 2021, 28(2):191–3., DOI 10.1016/j.tracli.2021.01.004 Sakatoku K, Takakuwa T, Miura A, Araki T, Fujitani Y, Yamamura R. Acquired factor V inhibitor with hemorrhagic symptoms after prasugrel hydrochloride treatment. Acta Haematol, 2020, 143(5):478–80., DOI 10.1159/ 000502406 Siekanska-Cholewa A, Jarosz A, Goralczyk T, Iwaniec T, Wegrzyn W, Drwila R, et al. Acquired factor V inhibitor in a woman following aortic aneurysm surgery. Blood Coagul Fibrinolysis, 2014, 25(5):515–7., DOI 10.1097/MBC. 0000000000000059 Sosa IR, Ellery P, Mast A, Neff AT, Gailani D. Acquired factor V deficiency in a patient without evidence of a classical inhibitor. Haemophilia, 2014, 20(1):e81–3., DOI 10.1111/hae.12280 Taniwaki M, Katsutani S, YamasakiM, Kawamoto K, Funaishi K,Matsumoto Y, et al. Acquired factor V inhibitor after antibiotic treatment in a patient with pneumonia: A case report. Ann Hematol, 2019, 98(8):1989–90., DOI 10.1007/ s00277-019-03638-6 Wiwanitkit V. Spectrum of bleeding in acquired factor V inhibitor: A summary of 33 cases. Clin Appl Thromb Hemost, 2006, 12(4):485–8., DOI 10. 1177/1076029606293438 Yokota Y, Inatomi O, Nakagawa M, Kakuda K, Hiroe K, Sakai S, et al. Acquired coagulation factor V inhibitor that was successfully treated with oral corticosteroid therapy. Intern Med, 2021, 60(16):2663–6., DOI 10.2169/ internalmedicine.7453-21 Wada H, Ichinose A, Shiraki K, Shimpo H, Shimaoka M. Coagulation factor V inhibitors, a review of the case report literature. Thromb Update, 2021, 4:100058., DOI 10.1016/j.tru.2021.100058 Ang AL, Kuperan P, Ng CH, Ng HJ. Acquired factor V inhibitor. A problembased systematic review. Thromb Haemost, 2009, 101(5):852–9., DOI 10.1160/th08- 11-0767 Kinjo Y, Yoshimura K, Suzuki T, Shinozawa K, Nakasone K, Yoshimata T, et al. Development of asymptomatic acquired factor V inhibitor after the administration of antibiotics. Rinsho Ketsueki, 2014, 55(11):2311–5., DOI 10. 11406/rinketsu.55.2311 Ashrani AA, Tefferi A, Pruthi RK, Pardanani A. Acquired factor V deficiency in myeloproliferative neoplasms: A mayo clinic series of 33 patients. Br J Haematol, 2015, 171(5):875–9., DOI 10.1111/bjh.13438 Simon B, Cegledi A, Dolgos J, Farkas P, Gaddh M, Hanko L, et al. Combined immunosuppression for acquired hemophilia A: CyDRi is a highly effective lowtoxicity regimen. Blood, 2022, 140(18):1983–92., DOI 10.1182/blood.2022016873 Ortel TL. Clinical and laboratory manifestations of anti-factor V antibodies. J Lab Clin Med, 1999, 133(4):326–34., DOI 10.1016/s0022-2143(99)90062-8 Kato T, Hanawa T, Asou M, Asakawa T, Sakamaki H, Araki M. Autoimmune factor V deficiency that took 16 Years to diagnose due to pseudodeficiency of multiple coagulation factors. Case Rep Med, 2021, 2021:4657501., DOI 10.1155/2021/4657501 Izumi T, Kim SW, Greist A, Macedo-Ribeiro S, Fuentes-Prior P, Bode W, et al. Fine mapping of inhibitory anti-factor V antibodies using factor V C2 domain mutants. Identification of two antigenic epitopes involved in phospholipid binding. Thromb Haemost, 2001, 85(6):1048–54., DOI 10.1055/s-0037-1615962 Ortel TL, Moore KD, Quinn-Allen MA, Okamura T, Sinclair AJ, Lazarchick J, et al. Inhibitory anti-factor V antibodies bind to the factor V C2 domain and are associated with hemorrhagic manifestations. Blood, 1998, 91(11):4188–96., DOI 10. 1182/blood.v91.11.4188.411k33_4188_4196 Terrab A, Pawlak D, Spaay P, Hoppensteadt D, Fareed J. Further removal of factor v related antigen from bovine thrombin by utilizing a membrane-filtration step. Clin Appl Thromb Hemost, 2008, 14(2):135–40., DOI 10.1177/1076029607308866 Agarwal V, Sachdev A, Singh R, Lehl S, Basu S. Autoimmune hemolytic anemia associated with benign ovarian cyst: A case report and review of literature. Indian J Med Sci, 2003, 57(11):504–6. Baker LR, Brain MC, Azzopardi JG, Worlledge SM. Autoimmune haemolytic anaemia associated with ovarian dermoid cyst. J Clin Pathol, 1968, 21(5):626–30., DOI 10.1136/jcp.21.5.626 Cobo F, Pereira A, Nomdedeu B, Gallart T, Ordi J, Torne A, et al. Ovarian dermoid cyst-associated autoimmune hemolytic anemia: A case report with emphasis on pathogenic mechanisms. Am J Clin Pathol, 1996, 105(5):567–71., DOI 10.1093/ajcp/105.5.567 Murad MD,MohlerDN,Carpenter JT, Jr. Remission of immune hemolytic anemia after removal of ovarian dermoid cyst:Case report. VaMed, 1982, 109(3):187–90., DOI 10. 1093/ajcp/105.5.567 Takemoto K, Hamada O, Kitamura K, Fujiwara N, Miyakawa Y. Challenges in management of unusual acquired factor V deficiency: A case report. Medicine, Baltimore,, 2019, 98(17):e15259., DOI 10.1097/MD.0000000000015259 Hirai D, Yamashita Y, Masunaga N, Katsura T, Akao M, Okuno Y, et al. Acquired factor V inhibitor. Intern Med, 2016, 55(20):3039–42., DOI 10.2169/ internalmedicine.55.6459 Chong LL, Wong YC. A case of factor V inhibitor. Am J Hematol, 1985, 19(4): 395–9., DOI 10.1002/ajh.2830190410 Andreadis P, Kafantari K, Agapidou A, Vakalopoulou S, Vlachaki E. Successful outcome of severe intra-cerebral bleeding associated with acquired factor V inhibition: Utilization of multiple therapeutic agents. Balkan Med J, 2018, 35(1):112–5., DOI 10.4274/balkanmedj.2017.0158 Perdekamp MT, Rubenstein DA, Jesty J, Hultin MB. Platelet factor V supports hemostasis in a patient with an acquired factor V inhibitor, as shown by prothrombinase and tenase assays. Blood Coagul Fibrinolysis, 2006, 17(7):593–7., DOI 10.1097/01.mbc.0000245297.64644.ee Lebrun A, Leroy-Matheron C, Arlet JB, Bartolucci P, Michel M. Successful treatment with rituximab in a patient with an acquired factor V inhibitor. Am J Hematol, 2008, 83(2):163–4., DOI 10.1002/ajh.21056 Yanagiya R, Kanouchi K, Toubai T, Yamada A, Aizawa K, Shiono Y, et al. Plasma exchange as an initial treatment for severe bleeding induced by acquired factor V deficiency: A case report and mini literature review. Acta Haematol, 2021, 144(1):82–7., DOI 10.1159/000505770 Guptill JT, Juel VC, Massey JM, Anderson AC, ChopraM, Yi JS, et al. Effect of therapeutic plasma exchange on immunoglobulins in myasthenia gravis. Autoimmunity, 2016, 49(7):472–9., DOI 10.1080/08916934.2016.1214823 de Raucourt E, Barbier C, Sinda P, Dib M, Peltier JY, Ternisien C. High-dose intravenous immunoglobulin treatment in two patients with acquired factor V inhibitors. Am J Hematol, 2003, 74(3):187–90., DOI 10.1002/ajh.10420 Feng Y, Xiao Y, Yan H,Wang P, Zhu W, Cassady K, et al. Sirolimus as rescue therapy for refractory/relapsed immune thrombocytopenia: Results of a singlecenter, prospective, single-arm study. Front Med, Lausanne,, 2020, 7:110., DOI 10. 3389/fmed.2020.00110 Rech J, Hueber AJ, Leipe J, Manger B, Schett G, Kallert S. A case report of plasma exchange, steroids, mycophenolate mofetil and cyclophosphamide in acquired factor VIII inhibitors. Ther Apher Dial, 2008, 12(5):406–8., DOI 10. 1111/j.1744-9987.2008.00618.x Wermes C, Niekrens C, Sykora KW. Successful long-time treatment with mycophenolate-mofetil in a child with acquired factor VIII inhibitor. Hamostaseologie, 2012, 32(Suppl. 1):S75–8. Akashi N, Ogawa Y, Yanagisawa K, Osaki Y, Shimizu H, Ishizaki T, et al. Recurrence of acquired factor V inhibitor after four years of remission. Rinsho Ketsueki, 2019, 60(1):46–50., DOI 10.11406/rinketsu.60.46 Muratori P, Loffreda S, Muratori L, Ferrari R, Afandi K, Cassani F, et al. Spontaneous remission of autoimmune hepatitis during pregnancy. Dig Liver Dis, 2002, 34(8):608–9., DOI 10.1016/s1590-8658(02)80098-6 Ostensen M, Villiger PM. The remission of rheumatoid arthritis during pregnancy. Semin Immunopathol, 2007, 29(2):185–91., DOI 10.1007/s00281-007- 0072-5 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611250 EP 1611257 DI 10.3389/pore.2023.1611250 PG 8 ER PT J AU Wang, X Wei, X AF Wang, Xiaoxue Wei, Xin TI Case report: Vulval sebaceous carcinoma: a report of two cases and literature review focus on treatment and survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE extraocular sebaceous carcinoma; vulval sebaceous carcinoma; surgery; adjuvant therapy; prognosis ID extraocular sebaceous carcinoma; vulval sebaceous carcinoma; surgery; adjuvant therapy; prognosis AB Background: Extraocular sebaceous carcinoma (SC) arising in the vulva is extremely rare that no treatment consensus has been well-defined. Case presentation: We here presented two cases of vulval SC in a 31-year-old and a 62-year-old woman, respectively. Radical wide local excision was performed with free margin and they received no postoperative adjuvant therapy. No evidence of disease was detected after follow-ups for 12 months and 49 months, respectively. A comprehensive literature review of vulval SC was further conducted and other ten cases were included. The mean age was 55.9 years, nine patients were diagnosed with FIGO stage I diseases while the remaining three patients had metastatic lesions at initial diagnosis. Surgery was the mainstay treatment option that 11 (91.7%) underwent surgical resection, of which 5 patients received inguinal lymphadenectomy and 2 patients showed lymph nodes involved. Radiotherapy and chemotherapy were given in 2 and 1 patient, respectively. Two patients experienced recurrence within 1 year after initial therapy. At the final follow-up, ten patients had no evidence of disease, one patient was alive with the disease, and only one died of the disease. Conclusion: Radical wide local excision may be preferred in early-stage vulval SC and utilization of sentinel lymph node sampling should be recommended. Postoperative adjuvant therapy may be spared in patients with negative surgical margin and absence of lymph node involvement. Treatment of vulval SC referring to the guidelines of vulvar cancer should be administered in case of positive margins or metastatic disease. C1 [Wang, Xiaoxue] Peking Union Medical College Hospital, Department of Obstetrics and GynecologyBeijing, China. [Wei, Xin] University of South China, Hengyang Medical School, The Affiliated Changsha Central Hospital, Department of Obstetrics and GynecologyChangsha, China. RP Wei, X (reprint author), University of South China, Hengyang Medical School, The Affiliated Changsha Central Hospital, Department of Obstetrics and Gynecology, Changsha, China. EM weixin1017a@163.com CR Knackstedt T, Samie FH. Sebaceous carcinoma: A review of the scientific literature. Curr Treat Options Oncol, 2017, 18(8):47., DOI 10.1007/s11864-017-0490-0 Owen JL, Kibbi N, Worley B, Kelm RC, Wang JV, Barker CA, et al. Sebaceous carcinoma: Evidence-based clinical practice guidelines. Lancet Oncol, 2019, 20(12): e699–e714., DOI 10.1016/S1470-2045(19)30673-4 Tripathi R, Chen Z, Li L, Bordeaux JS. Incidence and survival of sebaceous carcinoma in the United States. J Am Acad Dermatol, 2016, 75(6):1210–5., DOI 10. 1016/j.jaad.2016.07.046 Yamamoto A, Chigusa Y, Fujimoto M, Yamanoi K, Minamiguchi S, Yasuda E, et al. Sebaceous carcinoma of the vulva treated with sentinel lymph node biopsy: A case report and literature review. Int Cancer Conf J, 2021, 10(3):239–43., DOI 10. 1007/s13691-021-00488-w Kawamoto M, Fukuda Y, Kamoi S, Sugisaki Y, Yamanaka N. Sebaceous carcinoma of the vulva. Pathol Int, 1995, 45(10):767–73., DOI 10.1111/j.1440-1827.1995.tb03395.x Carlson JW, McGlennen RC, Gomez R, Longbella C, Carter J, Carson LF. Sebaceous carcinoma of the vulva: A case report and review of the literature. Gynecol Oncol, 1996, 60(3):489–91., DOI 10.1006/gyno.1996.0079 Sullivan SA, Tran AQ, O’Connor S, Gehrig PA. Sebaceous carcinoma of the vulva: A case report and review of the literature. Gynecol Oncol Rep, 2016, 18:40–1., DOI 10.1016/j.gore.2016.10.008 Rocha AC, Sa MI, Abrantes C, Sousa R. Sebaceous carcinoma of the vulva: An unexpected diagnosis and literature review. Acta Med Port, 2022, 35(1):63–7., DOI 10.20344/amp.13551 Khan Z, Misra G, Fiander AN, Dallimore NS. Sebaceous carcinoma of the vulva. Bjog, 2003, 110(2):227–8., DOI 10.1046/j.1471-0528.2003.01157.x Alharthi H, Alnuaim H, Aljarbou O, Arabi H. Sebaceous carcinoma of the vulva: A case report and review of the literature. Avicenna JMed, 2021, 11(1):49–53., DOI 10.4103/ajm.ajm_183_20 Schmults CD, Blitzblau R, Aasi SZ, Alam M, Andersen JS, Baumann BC, et al. NCCN Guidelines® insights: Squamous cell skin cancer, version 1.2022. J Natl Compr Canc Netw, 2021, 19(12):1382–94., DOI 10.6004/jnccn.2021.0059 Thakur BK, Verma S, Khonglah Y, Jitani A. Multifocal sebaceous carcinoma of the vulva. Indian J Dermatol Venereol Leprol, 2017, 83(2):221–4., DOI 10.4103/ 0378-6323.198436 Tryggvason G, Bayon R, Pagedar NA. Epidemiology of sebaceous carcinoma of the head and neck: Implications for lymph node management. Head Neck, 2012, 34(12):1765–8., DOI 10.1002/hed.22009 Van der Zee AG, Oonk MH, De Hullu JA, Ansink AC, Vergote I, Verheijen RH, et al. Sentinel node dissection is safe in the treatment of early-stage vulvar cancer. J Clin Oncol, 2008, 26(6):884–9., DOI 10.1200/JCO.2007.14.0566 Oonk MHM, Slomovitz B, Baldwin PJW, van Doorn HC, van der Velden J, de Hullu JA, et al. Radiotherapy versus inguinofemoral lymphadenectomy as treatment for vulvar cancer patients with micrometastases in the sentinel node: Results of GROINSS-V II. J Clin Oncol, 2021, 39(32):3623–32., DOI 10.1200/JCO.21. 00006 Bhaijee F, Brown AS. Muir-Torre syndrome. Arch Pathol Lab Med, 2014, 138(12):1685–9., DOI 10.5858/arpa.2013-0301-RS Chang AY, Miller CJ, Elenitsas R, Newman JG, Sobanko JF. Management considerations in extraocular sebaceous carcinoma. Dermatol Surg, 2016, 42(1): S57–65., DOI 10.1097/DSS.0000000000000575 Bogani G, Leone Roberti Maggiore U, Raspagliesi F. Lynch syndrome - muirtorre variant: Implication in gynecologic oncology. J Gynecol Oncol, 2018, 29(5): e84., DOI 10.3802/jgo.2018.29.e84 Dores GM, Curtis RE, Toro JR, Devesa SS, Fraumeni JF, Jr. Incidence of cutaneous sebaceous carcinoma and risk of associated neoplasms: Insight into muir-torre syndrome. Cancer, 2008, 113(12):3372–81., DOI 10.1002/cncr.23963 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611259 EP 1611265 DI 10.3389/pore.2023.1611259 PG 7 ER PT J AU Varga, Zs Biro, A Torok, M Toth, D AF Varga, Zsolt Biro, Adrienn Torok, Miklos Toth, Dezso TI A combined approach for individualized lymphadenectomy in gastric cancer patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gastric cancer; surgery; lymphadenectomy; sentinel; Maruyama ID gastric cancer; surgery; lymphadenectomy; sentinel; Maruyama AB Introduction: Gastric cancer ranks as the fifth most common cancer globally. The presence of lymph node metastasis is a significant prognostic factor influencing survival. Postoperative morbidity and nodal staging accuracy are heavily affected by the extent of lymph node dissection. Our study aimed to explore the potential integration of two contemporary methods, sentinel node navigation surgery (SNNS) and the Maruyama Computer Program (MCP), to improve the accuracy of nodal staging. Materials and methods: We conducted a prospective data collection involving patients with gastric adenocarcinoma from 2008 to 2018 at the Department of Surgery, University of Debrecen, Hungary. Data from 100 consecutive patients were collected. The primary and secondary endpoints included evaluating the rate of node-negative patients and the diagnostic accuracy of our combined approach. Results: Sentinel node mapping was successful in 97 out of 100 patients. We found that using the threshold value of the Maruyama Index (MI) ≥ 28, all metastatic stations of sentinel-node-negative patients could be identified. Our method achieved 100% sensitivity and negative predictive value, with a specificity of 60.42% (95% CI = 46.31%–72.98%). Discussion: The combined application of SNNS and MCP has proven to be an effective diagnostic technique in the synergistic approach for identifying metastasis-positive lymph node stations. Despite its limitations, this combination may assist clinicians in customizing lymphadenectomy for gastric cancer patients. C1 [Varga, Zsolt] University of Debrecen, Department of Surgery and Operative TechniquesDebrecen, Hungary. [Biro, Adrienn] Somogy Megyei Kaposi Mor Oktato Korhaz, Sebeszeti OsztalyKaposvar, Hungary. [Torok, Miklos] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Toth, Dezso] University of Debrecen, Department of Surgery and Operative TechniquesDebrecen, Hungary. RP Toth, D (reprint author), University of Debrecen, Department of Surgery and Operative Techniques, Debrecen, Hungary. EM dr.toth.dezso@med.unideb.hu CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer Statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3): 209–49., DOI 10.3322/caac.21660 Eom SS, Choi W, Eom BW, Park SH, Kim SJ, Kim YI, et al. A comprehensive and comparative review of global gastric cancer treatment guidelines. J Gastric Cancer, 2022, 22(1):3–23., DOI 10.5230/jgc.2022.22.e10 Hartgrink HH, van de Velde CJ, Putter H, Bonenkamp JJ, Klein Kranenbarg E, Songun I, et al. Extended lymph node dissection for gastric cancer: Who may benefit? Final results of the randomized Dutch gastric cancer group trial. J Clin Oncol, 2004, 22(11):2069–77., DOI 10.1200/JCO. 2004.08.026 Degiuli M, Reddavid R, Tomatis M, Ponti A, Morino M, Sasako M, et al. D2 dissection improves disease-specific survival in advanced gastric cancer patients: 15-Year follow-up results of the Italian gastric cancer study group D1 versus D2 randomised controlled trial. Eur J Cancer, 2021, 150:10–22., DOI 10.1016/j.ejca. 2021.03.031 Songun I, Putter H, Kranenbarg EM, Sasako M, van de Velde CJ. Surgical treatment of gastric cancer: 15-Year follow-up results of the randomised nationwide Dutch D1d2 trial. Lancet Oncol, 2010, 11(5):439–49., DOI 10.1016/S1470-2045(10, 70070-X Habermann CR, Weiss F, Riecken R, Honarpisheh H, Bohnacker S, Staedtler C, et al. Preoperative staging of gastric adenocarcinoma: Comparison of helical ct and endoscopic us. Radiology, 2004, 230(2):465–71., DOI 10.1148/radiol. 2302020828 Kim HJ, Kim AY, Oh ST, Kim JS, Kim KW, Kim PN, et al. Gastric cancer staging at multi-detector row ct gastrography: Comparison of transverse and volumetric ct scanning. Radiology, 2005, 236(3):879–85., DOI 10.1148/radiol. 2363041101 Findlay JM, Antonowicz S, Segaran A, El Kafsi J, Zhang A, Bradley KM, et al. Routinely staging gastric cancer with, 18)F-fdg pet-ct detects additional metastases and predicts early recurrence and death after surgery. Eur Radiol, 2019, 29(5): 2490–8., DOI 10.1007/s00330-018-5904-2 Choi YY, An JY, Guner A, Kang DR, Cho I, Kwon IG, et al. Skip lymph node metastasis in gastric cancer: Is it skipping or skipped? Gastric Cancer, 2016, 19(1): 206–15., DOI 10.1007/s10120-015-0472-5 Faige DO. Eus in patients with benign and malignant lymphadenopathy. Gastrointest Endosc, 2001, 53(6):593–8., DOI 10.1067/mge.2001.114060 Hwang SW, Lee DH, Lee SH, Park YS, Hwang JH, Kim JW, et al. Preoperative staging of gastric cancer by endoscopic ultrasonography and multidetector-row computed tomography. J Gastroenterol Hepatol, 2010, 25(3):512–8., DOI 10.1111/j. 1440-1746.2009.06106.x Japanese Gastric Cancer A. Japanese gastric cancer treatment guidelines 2018, 5th edition). Gastric Cancer, 2021, 24(1):1–21., DOI 10.1007/s10120-020- 01042-y Ohdaira H, Nimura H, Mitsumori N, Takahashi N, Kashiwagi H, Yanaga K. Validity of modified gastrectomy combined with sentinel node navigation surgery for early gastric cancer. Gastric Cancer, 2007, 10(2):117–22., DOI 10.1007/s10120- 007-0419-6 Wei J, Bu Z. Sentinel lymph node detection for gastric cancer: Promise or pitfall? Surg Oncol, 2020, 33:1–6., DOI 10.1016/j.suronc.2019.12.005 Miwa K, Kinami S, Taniguchi K, Fushida S, Fujimura T, Nonomura A. Mapping sentinel nodes in patients with early-stage gastric carcinoma. Br J Surg, 2003, 90(2):178–82., DOI 10.1002/bjs.4031 Lee SE, Lee JH, Ryu KW, Cho SJ, Lee JY, Kim CG, et al. Sentinel nodemapping and skip metastases in patients with early gastric cancer. Ann Surg Oncol, 2009, 16(3):603–8., DOI 10.1245/s10434-008-0283-6 Liu JY, Deng JY, Zhang NN, Liu HF, Sun WL, He WT, et al. Clinical significance of skip lymph-node metastasis in Pn1 gastric-cancer patients after curative surgery. Gastroenterol Rep, Oxf,, 2019, 7(3):193–8., DOI 10.1093/gastro/ goz008 Jeuck TL, Wittekind C. Gastric carcinoma: Stage migration by immunohistochemically detected lymph node micrometastases. Gastric Cancer, 2015, 18(1):100–8., DOI 10.1007/s10120-014-0352-4 Ryu KW, Kim YW, Min JS, An JY, Yoon HM, Eom BW, et al. Laparoscopic sentinel node navigation surgery versus laparoscopic standard gastrectomy with lymph node dissection in early gastric cancer: Final threeyear survival results of multicenter randomized controlled phase iii trial, senorita trial). J Clin Oncol, 2020, 38(15):4510., DOI 10.1200/JCO.2020.38. 15_suppl.4510 Hur H, Lee YJ, Kim Y-W, Min J-S, Yoon HM, An JY, et al. Clinical efficacy of laparoscopic sentinel node navigation surgery for early gastric cancer: Five-year results of senorita trial. J Clin Oncol, 2022, 40(16):4050., DOI 10.1200/JCO.2022.40. 16_suppl.4050 Kampschoer GH, Maruyama K, van de Velde CJ, Sasako M, Kinoshita T, Okabayashi K. Computer analysis in making preoperative decisions: A rational approach to lymph node dissection in gastric cancer patients. Br J Surg, 1989, 76(9): 905–8., DOI 10.1002/bjs.1800760910 Maruyama K, Gunven P, Okabayashi K, Sasako M, Kinoshita T. Lymph node metastases of gastric cancer. General pattern in 1931 patients. Ann Surg, 1989, 210(5):596–602., DOI 10.1097/00000658-198911000-00005 Bollschweiler EH, Monig SP, Hensler K, Baldus SE, Maruyama K, Holscher AH. Artificial neural network for prediction of lymph node metastases in gastric cancer: A phase ii diagnostic study. Ann Surg Oncol, 2004, 11(5):506–11., DOI 10. 1245/ASO.2004.04.018 Toth D, Torok M, Kincses Z, Damjanovich L. Prospective, comparative study for the evaluation of lymph node involvement in gastric cancer: Maruyama computer program versus sentinel lymph node biopsy. Gastric Cancer, 2013, 16(2):201–7., DOI 10.1007/s10120-012-0170-5 Hundahl SA, Macdonald JS, Benedetti J, Fitzsimmons TSouthwest Oncology Group and the Gastric Intergroup. Surgical treatment variation in a prospective, randomized trial of chemoradiotherapy in gastric cancer: The effect of undertreatment. Ann Surg Oncol, 2002, 9(3):278–86., DOI 10.1007/ BF02573066 Peeters KC, Hundahl SA, Kranenbarg EK, Hartgrink H, van de Velde CJ. Low Maruyama index surgery for gastric cancer: Blinded reanalysis of the Dutch D1-D2 trial. World J Surg, 2005, 29(12):1576–84., DOI 10.1007/s00268-005- 7907-9 Athauda A, Nankivell M, Langer R, Pritchard S, Langley RE, von Loga K, et al. Pathological regression of primary tumour and metastatic lymph nodes following chemotherapy in resectable og cancer: Pooled analysis of two trials. Br J Cancer, 2023, 128(11):2036–43., DOI 10.1038/s41416-023-02217-x Zhu YL, Sun YK, Xue XM, Yue JY, Yang L, Xue LY. Unnecessity of lymph node regression evaluation for predicting gastric adenocarcinoma outcome after neoadjuvant chemotherapy. World J Gastrointest Oncol, 2019, 11(1):48–58., DOI 10. 4251/wjgo.v11.i1.48 PereiraMA, RamosM, Dias AR, Cardili L, Ribeiro RRE, Charruf AZ, et al. Lymph node regression after neoadjuvant chemotherapy: A predictor of survival in gastric cancer. J Surg Oncol, 2020, 121(5):795–803., DOI 10.1002/ jso.25785 Park JY, Kim YW, Ryu KW, Nam BH, Lee YJ, Jeong SH, et al. Assessment of laparoscopic stomach preserving surgery with sentinel basin dissection versus standard gastrectomy with lymphadenectomy in early gastric cancer-a multicenter randomized phase iii clinical trial, senorita trial, protocol. BMC Cancer, 2016, 16:340., DOI 10.1186/s12885-016-2336-8 Miyashiro I, Hiratsuka M, Sasako M, Sano T, Mizusawa J, Nakamura K, et al. High false-negative proportion of intraoperative histological examination as a serious problem for clinical application of sentinel node biopsy for early gastric cancer: Final results of the Japan clinical oncology group multicenter trial Jcog0302. Gastric Cancer, 2014, 17(2):316–23., DOI 10.1007/s10120-013-0285-3 Toth D, Kincses Z, Plosz J, Torok M, Kovacs I, Kiss C, et al. Value of sentinel lymph node mapping using a blue dye-only method in gastric cancer: A singlecenter experience from north-east Hungary. Gastric Cancer, 2011, 14(4):360–4., DOI 10.1007/s10120-011-0048-y Toth D, Kathy S, Csoban T, Kincses Z, Torok M, Plosz J, et al. Gyomortumorok orszemnyirokcsomo-jelolesenek prospektiv, osszehasonlito vizsgalata – submucosus kontra subserosus jeloles. Magy Seb, 2012, 65(1):3–8., DOI 10.1556/MaSeb.65.2012.1.1 Shimada A, Takeuchi H, Nishi T, Mayanagi S, Fukuda K, Suda K, et al. Utility of the one-step nucleic acid amplification assay in sentinel node mapping for early gastric cancer patients. Gastric Cancer, 2020, 23(3):418–25., DOI 10.1007/s10120- 019-01016-9 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611270 EP 1611279 DI 10.3389/pore.2023.1611270 PG 10 ER PT J AU Ogasawara, H Yoshizawa, T Oshima, K Ogasawara, K Kubota, Sh Goto, Sh Morohashi, S Wakiya, T Kimura, N Ishido, K Kijima, H Hakamada, K AF Ogasawara, Hirokazu Yoshizawa, Tadashi Oshima, Kiyoko Ogasawara, Kenta Kubota, Shunsuke Goto, Shintaro Morohashi, Satoko Wakiya, Taiichi Kimura, Norihisa Ishido, Keinosuke Kijima, Hiroshi Hakamada, Kenichi TI Three-dimensional analysis of perineural invasion in extrahepatic cholangiocarcinoma using tissue clearing SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tissue clearing; three-dimensional; perineural invasion; cholangiocarcinoma; iDISCO ID tissue clearing; three-dimensional; perineural invasion; cholangiocarcinoma; iDISCO AB Perineural invasion (PNI) is a characteristic invasion pattern of distal cholangiocarcinoma (DCC). Conventional histopathologic examination is a challenging approach to analyze the spatial relationship between cancer and neural tissue in full-thickness bile duct specimens. Therefore, we used a tissue clearing method to examine PNI in DCC with three-dimensional (3D) structural analysis. The immunolabeling-enabled 3D imaging of solvent-cleared organs method was performed to examine 20 DCC specimens from five patients and 8 non-neoplastic bile duct specimens from two controls. The bile duct epithelium and neural tissue were labeled with CK19 and S100 antibodies, respectively. Two-dimensional hematoxylin/eosin staining revealed only PNI around thick nerve fibers in the deep layer of the bile duct, whereas PNI was not identified in the superficial layer. 3D analysis revealed that the parts of DCC closer to the mucosa exhibited more nerves than the normal bile duct. The nerve fibers were continuously branched and connected with thick nerve fibers in the deep layer of the bile duct. DCC formed a tubular structure invading from the epithelium and extending around thin nerve fibers in the superficial layer. DCC exhibited continuous infiltration around the thick nerve fibers in the deep layer. This is the first study using a tissue clearing method to examine the PNI of DCC, providing new insights into the underlying mechanisms. C1 [Ogasawara, Hirokazu] Hirosaki University Graduate School of Medicine, Department of Pathology and BioscienceHirosaki, Aomori, Japan. [Yoshizawa, Tadashi] Hirosaki University Graduate School of Medicine, Department of Pathology and BioscienceHirosaki, Aomori, Japan. [Oshima, Kiyoko] Johns Hopkins School of Medicine, Department of PathologyBaltimore, MD, USA. [Ogasawara, Kenta] Hirosaki University Graduate School of Medicine, Department of Pathology and BioscienceHirosaki, Aomori, Japan. [Kubota, Shunsuke] Hirosaki University Graduate School of Medicine, Department of Pathology and BioscienceHirosaki, Aomori, Japan. [Goto, Shintaro] Hirosaki University Graduate School of Medicine, Department of Pathology and BioscienceHirosaki, Aomori, Japan. [Morohashi, Satoko] Hirosaki University Graduate School of Medicine, Department of Pathology and BioscienceHirosaki, Aomori, Japan. [Wakiya, Taiichi] Hirosaki University Graduate School of Medicine, Department of Gastroenterological SurgeryHirosaki, Aomori, Japan. [Kimura, Norihisa] Hirosaki University Graduate School of Medicine, Department of Gastroenterological SurgeryHirosaki, Aomori, Japan. [Ishido, Keinosuke] Hirosaki University Graduate School of Medicine, Department of Gastroenterological SurgeryHirosaki, Aomori, Japan. [Kijima, Hiroshi] Hirosaki University Graduate School of Medicine, Department of Pathology and BioscienceHirosaki, Aomori, Japan. [Hakamada, Kenichi] Hirosaki University Graduate School of Medicine, Department of Gastroenterological SurgeryHirosaki, Aomori, Japan. RP Yoshizawa, T (reprint author), Hirosaki University Graduate School of Medicine, Department of Pathology and Bioscience, Hirosaki, Japan. EM tyoshi@hirosaki-u.ac.jp CR Wu R, Zhang G, Feng J, Zhang L, Yang Z. Proposal of the optimal numbers of examined and positive lymph nodes to the 8th edition of American joint committee on cancer, AJCC, staging for 758 patients with distal cholangiocarcinoma. PLOS ONE, 2020, 15:e0234464., DOI 10.1371/journal.pone.0234464 Kijima H,Wu Y, Yosizawa T, Suzuki T, Tsugeno Y, Haga T, et al. Pathological characteristics of early to advanced gallbladder carcinoma and extrahepatic cholangiocarcinoma. J Hepato-bil Pancreat Sci, 2014, 21:453–8., DOI 10.1002/ jhbp.72 Bhuiya MR, Nimura Y, Kamiya J, Kondo S, Fukata S, Hayakawa N, et al. Clinicopathologic studies on perineural invasion of bile duct carcinoma. Ann Surg, 1992, 215:344–9., DOI 10.1097/00000658-199204000-00007 Wellner UF, Shen Y, Keck T, Jin W, Xu Z. The survival outcome and prognostic factors for distal cholangiocarcinoma following surgical resection: A meta-analysis for the 5-year survival. Surg Today, 2017, 47:271–9., DOI 10.1007/s00595-016- 1362-0 Kondo N, Murakami Y, Uemura K, Hashimoto Y, Nakagawa N, Sasaki H, et al. An increased number of perineural invasions is independently associated with poor survival of patients with resectable pancreatic ductal adenocarcinoma. Pancreas, 2015, 44:1345–51., DOI 10.1097/MPA.0000000000000413 Ma J, Jiang Y, Jiang Y, Sun Y, Zhao X. Expression of nerve growth factor and tyrosine kinase receptor A and correlation with perineural invasion in pancreatic cancer. J Gastroenterol Hepatol, 2008, 23:1852–9., DOI 10.1111/j.1440-1746.2008. 05579.x Li S, Jo YS, Lee JH, Min JK, Lee ES, Park T, et al. L1 cell adhesion molecule is a novel independent poor prognostic factor of extrahepatic cholangiocarcinoma. Clin Cancer Res, 2009, 15:7345–51., DOI 10.1158/1078-0432.CCR-09-0959 Na’ara S, Amit M, Gil Z. L1CAM induces perineural invasion of pancreas cancer cells by upregulation of metalloproteinase expression. Oncogene, 2019, 38: 596–608., DOI 10.1038/s41388-018-0458-y Feng Y, Hu X, Liu G, Lu L, ZhaoW, Shen F, et al. M3 muscarinic acetylcholine receptors regulate epithelial-mesenchymal transition, perineural invasion, and migration/metastasis in cholangiocarcinoma through the AKT pathway. Cancer Cell Int, 2018, 18:173., DOI 10.1186/s12935-018-0667-z Nan L, Qin T, Xiao Y, Qian W, Li J, Wang Z, et al. Pancreatic stellate cells facilitate perineural invasion of pancreatic cancer via HGF/c-met pathway. Cell Transpl, 2019, 28(9-10):1289–98., DOI 10.1177/0963689719851772 Maxwell P, Hamilton PW, Sloan JM. Three-dimensional reconstruction of perineural invasion in carcinoma of the extrahepatic bile ducts. J Pathol, 1996, 180: 142–5., DOI 10.1002/(SICI)1096-9896(199610)180:2<142::AID-PATH618>3.0.CO; 2-6 Miyashita Y, Ikeda T, Shinto E, Okano S, Korehisa S, Shimazaki H, et al. Three-dimensional imaging of intramural perineural invasion in colorectal cancer: Three-dimensional reconstruction approach with multiple immunohistochemically stained sections. Pathol Int, 2022, 72:293–9., DOI 10.1111/pin.13222 Renier N, Wu Z, Simon DJ, Yang J, Ariel P, Tessier-Lavigne M. iDISCO: A simple, rapid method to immunolabel large tissue samples for volume imaging. Cell, 2014, 159:896–910., DOI 10.1016/j.cell.2014.10.010 Noe M, Rezaee N, Asrani K, Skaro M, Groot VP, Wu PH, et al. Immunolabeling of cleared human pancreata provides insights into threedimensional pancreatic anatomy and pathology. Am J Pathol, 2018, 188: 1530–5., DOI 10.1016/j.ajpath.2018.04.002 Hong SM, Jung D, Kiemen A, Gaida MM, Yoshizawa T, Braxton AM, et al. Three-dimensional visualization of cleared human pancreas cancer reveals that sustained epithelial-to-mesenchymal transition is not required for venous invasion. Mod Pathol, 2020, 33:639–47., DOI 10.1038/s41379-019- 0409-3 Yoshizawa T, Hong SM, Jung D, Noe M, Kiemen A, Wu PH, et al. Threedimensional analysis of extrahepatic cholangiocarcinoma and tumor budding. J Pathol, 2020, 251:400–10., DOI 10.1002/path.5474 Hashiguchi G. Studies on perineural invasion of the extrahepatic bile duct cancer. Tokyo Jikeikai Med J, 1984, 99:687–98. Ayala GE, Wheeler TM, Shine HD, Schmelz M, Frolov A, Chakraborty S, et al. In vitro dorsal root ganglia and human prostate cell line interaction: Redefining perineural invasion in prostate cancer. Prostate, 2001, 49:213–23., DOI 10.1002/pros. 1137 Ayala GE, Dai H, Powell M, Li R, Ding Y, Wheeler TM, et al. Cancer-related axonogenesis and neurogenesis in prostate cancer. Clin Cancer Res, 2008, 14: 7593–603., DOI 10.1158/1078-0432.CCR-08-1164 Suzuki M, Takahashi T, Ouchi K, Matsuno S. Perineural tumor invasion and its relation with the lymphogenous spread in human and experimental carcinoma of bile duct. A computer-aided 3-D reconstruction study. Tohoku J Exp Med, 1994, 172:17–28., DOI 10.1620/tjem.172.17 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2023 VL 29 IS 2 BP 1611284 EP 1611290 DI 10.3389/pore.2023.1611284 PG 7 ER PT J AU Han, G Sun, Ch Cui, L Huang, Y Yu, L Liu, Sh Tao, M AF Han, Guohu Sun, Changchun Cui, Lihua Huang, Yufeng Yu, Lijiang Liu, Shenzha Tao, Min TI Efficacy of the low dose apatinib plus deep hyperthermia as third-line or later treatment in HER-2 negative advanced gastric cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apatinib; deep hyperthermia; gastric cancer; efficacy evaluation; prognosis ID apatinib; deep hyperthermia; gastric cancer; efficacy evaluation; prognosis AB Aim: To observe the efficacy of the low dose apatinib plus deep hyperthermia as third-line or later treatment for patients with human epidermal growth factor receptor 2 (HER-2) negative advanced gastric cancer. Methods: 80 eligible patients with HER-2 negative advanced gastric cancer admitted to Jingjiang People’s Hospital Affiliated with Yangzhou Universityfrom March 2021 to March 2022 were selected, and they were divided into the control group (n = 40, apatinib) and experimental group (n = 40, apatinib plus deep hyperthermia) on the basis of random number table method. The levels of serum carcinoembryonic antigen (CEA), carbohydrate antigen 199 (CA199), and vascular endothelial growth factor (VEGF) were monitored, and the efficacy of the two groups was analyzed by referring to Karnofsky performance status (KPS), overall survival (OS) and disease control rate (DCR) before and after treatment. Results: The levels of CEA, CA199, and VEGF in both groups were lower after treatment than before (p < 0.05), and lower (CEA: 8.85 ± 1.36 vs. 12.87 ± 1.23, CA199: 34.19 ± 4.68 vs. 50.11 ± 5.73, VEGF: 124.8 ± 18.03 vs. 205.9 ± 19.91) in the experimental group than in the control group (p < 0.05). The DCR and KPS of the patients in the experimental group were significantly higher (DCR: 62.50% vs. 40.00%; KPS: 83.25 ± 1.15 vs. 76.25 ± 1.17) than in the control group (p < 0.05). In survival analysis, patients with control group had shorter OS than the experimental group. (median 5.65 vs. 6.50 months; hazard ratio [HR], 1.63 [95% confidence interval (CI) 1.02–2.60], p = 0.0396). Conclusion: The application of low-dose apatinib plus deep hyperthermia for patients with HER-2 negative gastric cancer who failed second-line treatment should be a promising option. C1 [Han, Guohu] Dushu Lake Hospital Affiliated to Soochow University, Department of OncologySuzhou, Jiangsu, China. [Sun, Changchun] The Seventh Affiliated Hospital of Yangzhou University, Jingjiang People’s Hospital, Department of OncologyJingjiang, Jiangsu, China. [Cui, Lihua] The Seventh Affiliated Hospital of Yangzhou University, Jingjiang People’s Hospital, Department of OncologyJingjiang, Jiangsu, China. [Huang, Yufeng] The Seventh Affiliated Hospital of Yangzhou University, Jingjiang People’s Hospital, Department of OncologyJingjiang, Jiangsu, China. [Yu, Lijiang] The Seventh Affiliated Hospital of Yangzhou University, Jingjiang People’s Hospital, Department of OncologyJingjiang, Jiangsu, China. [Liu, Shenzha] The Seventh Affiliated Hospital of Yangzhou University, Jingjiang People’s Hospital, Department of OncologyJingjiang, Jiangsu, China. [Tao, Min] Dushu Lake Hospital Affiliated to Soochow University, Department of OncologySuzhou, Jiangsu, China. RP Tao, M (reprint author), Dushu Lake Hospital Affiliated to Soochow University, Department of Oncology, Suzhou, China. EM taomin@suda.edu.cn CR Kawazoe A, Shitara K, Boku N, Yoshikawa T, Terashima M. Current status of immunotherapy for advanced gastric cancer. Jpn J Clin Oncol, 2021, 51(1):20–7., DOI 10.1093/jjco/hyaa202 Leiting JL, Grotz TE. Advancements and challenges in treating advanced gastric cancer in the West. World J Gastrointest Oncol, 2019, 11(9):652–64., DOI 10.4251/ wjgo.v11.i9.652 van Amelsfoort RM, van der Sluis K, Schats W, Jansen E, van Sandick JW, Verheij M, et al. Health-related quality of life in locally advanced gastric cancer: A systematic review. Cancers, Basel,, 2021, 13(23):5934., DOI 10.3390/ cancers13235934 Chen ZD, Zhang PF, Xi HQ, Wei B, Chen L, Tang Y. Recent advances in the diagnosis, staging, treatment, and prognosis of advanced gastric cancer: A literature review. Front Med, Lausanne,, 2021, 8:744839., DOI 10.3389/fmed.2021.744839 Izumi D, Nunobe S. How to decide approaches and procedures for early and advanced gastric cancer? Can J Gastroenterol Hepatol, 2022, 2022:8324242., DOI 10. 1155/2022/8324242 Wagner AD, Syn NL, Moehler M, Grothe W, Yong WP, Tai BC, et al. Chemotherapy for advanced gastric cancer. Cochrane Database Syst Rev, 2017, 8(8):CD004064., DOI 10.1002/14651858.CD004064.pub4 Tian Z, Niu X, Yao W. Efficacy and response biomarkers of apatinib in the treatment of malignancies in China: A review. Front Oncol, 2021, 11:749083., DOI 10.3389/fonc.2021.749083 Lindner LH, Blay JY, Eggermont A, Issels RD. Perioperative chemotherapy and regional hyperthermia for high-risk adult-type soft tissue sarcomas. Eur J Cancer, 2021, 147:164–9., DOI 10.1016/j.ejca.2021.02.002 Gronau F, Jara M, Feldbrugge L, Wolf V, Oeff A, Rau B. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in gastric cancer. Chirurg, 2021, 92:522–7., DOI 10.1007/s00104-021-01371-3 Sun D, Li H, Cao M, He S, Lei L, Peng J, et al. Cancer burden in China: Trends, risk factors and prevention. Cancer Biol Med, 2020, 17(4):879–95., DOI 10.20892/j. issn.2095-3941.2020.0387 Song Z,Wu Y, Yang J, Yang D, Fang X. Progress in the treatment of advanced gastric cancer. Tumour Biol, 2017, 39(7):1010428317714626., DOI 10.1177/ 1010428317714626 Westermann AM, Grosen EA, Katschinski DM, Jager D, Rietbroek R, Schink JC, et al. A pilot study of whole body hyperthermia and carboplatin in platinumresistant ovarian cancer. Eur J Cancer, 2001, 37(9):1111–7., DOI 10.1016/s0959- 8049(01)00074-0 Teng F, Xu Z, Chen J, Zheng G, Zheng G, Lv H, et al. DUSP1 induces apatinib resistance by activating the MAPK pathway in gastric cancer. Oncol Rep, 2018, 40(3):1203–22., DOI 10.3892/or.2018.6520 Scott LJ. Apatinib: A review in advanced gastric cancer and other advanced cancers. Drugs, 2018, 78(7):747–58., DOI 10.1007/s40265-018-0903-9 Zhang Y, Gou M, Han C, Li J, Wang L, Qiao Q, et al. Efficacy and safety of apatinib as second-line therapy for advanced gastric cancer: A single-center observational study. Anticancer Drugs, 2018, 29(2):184–9., DOI 10.1097/CAD. 0000000000000582 Chen D, Zhong X, Lin L, Xie J, Lian Y, Xu L. Comparative efficacy and adverse reactions of apatinib-chemotherapy combinations versus chemotherapy alone for treatment of advanced colorectal cancer: A meta-analysis of randomized controlled trials. Am J Transl Res, 2022, 14(9):6703–11. Xue JM, Astere M, Zhong MX, Lin H, Shen J, Zhu YX. Efficacy and safety of apatinib treatment for gastric cancer, hepatocellular carcinoma and non-small cell lung cancer: A meta-analysis. Onco Targets Ther, 2018, 11:6119–28., DOI 10.2147/ OTT.S172717 Dong CR, Hu DX, Liu SC, Luo HL, Zhang WJ. AKT/GSK-3beta/VEGF signaling is involved in P2RY2 activation-induced the proliferation and metastasis of gastric cancer. Carcinogenesis, 2023, 44(1):65–79., DOI 10.1093/carcin/bgac095 Pang L, Wang J, Fan Y, Xu R, Bai Y, Bai L. Correlations of TNM staging and lymph node metastasis of gastric cancer with MRI features and VEGF expression. Cancer Biomark, 2018, 23(1):53–9., DOI 10.3233/CBM-181287 Ning S, WeiW, Li J,Hou B, Zhong J, Xie Y, et al. Clinical significance and diagnostic capacity of serum TK1, CEA, CA 19-9 and CA 72-4 levels in gastric and colorectal cancer patients. J Cancer, 2018, 9(3):494–501., DOI 10.7150/jca. 21562 Sun Z, Zhang N. Clinical evaluation of CEA, CA19-9, CA72-4 and CA125 in gastric cancer patients with neoadjuvant chemotherapy. World J Surg Oncol, 2014, 12:397., DOI 10.1186/1477-7819-12-397 Du Y, Cao Q, Jiang C, Liang H, Ning Z, Ji C, et al. Effectiveness and safety of low-dose apatinib in advanced gastric cancer: A real-world study. Cancer Med, 2020, 9(14):5008–14., DOI 10.1002/cam4.3105 Huang M, Li J, Yu X, Xu Q, Zhang X, Dai X, et al. Comparison of efficacy and safety of third-line treatments for advanced gastric cancer: A systematic review with bayesian network meta-analysis. Front Oncol, 2021, 11:734323., DOI 10.3389/fonc. 2021.734323 Mi DH, Li Z, Yang KH, Cao N, Lethaby A, Tian JH, et al. Surgery combined with intraoperative hyperthermic intraperitoneal chemotherapy, IHIC, for gastric cancer: A systematic review and meta-analysis of randomised controlled trials. Int J Hyperthermia, 2013, 29(2):156–67., DOI 10.3109/02656736.2013.768359 Cesna V, Sukovas A, Jasukaitiene A, Naginiene R, Barauskas G, Dambrauskas Z, et al. Narrow line between benefit and harm: Additivity of hyperthermia to cisplatin cytotoxicity in different gastrointestinal cancer cells. World J Gastroenterol, 2018, 24(10):1072–83., DOI 10.3748/wjg.v24.i10.1072 Gao S, Li X, Shi W, Huo L, Liu H. Efficacy of the low dose apatinib plus chemotherapy on advanced gastric carcinoma. J Oncol, 2022, 2022:3009494., DOI 10. 1155/2022/3009494 Sawaji Y, Sato T, Takeuchi A, Hirata M, Ito A. Anti-angiogenic action of hyperthermia by suppressing gene expression and production of tumour-derived vascular endothelial growth factor in vivo and in vitro. Br J Cancer, 2022, 86(10): 1597–603., DOI 10.1038/sj.bjc.6600268 Andresen TL, Jensen SS, Jorgensen K. Advanced strategies in liposomal cancer therapy: Problems and prospects of active and tumor specific drug release. Prog Lipid Res, 2005, 44(1):68–97., DOI 10.1016/j.plipres.2004.12.001 Wust P, Hildebrandt B, Sreenivasa G, Rau B, Gellermann J, Riess H, et al. Hyperthermia in combined treatment of cancer. Lancet Oncol, 2002, 3(8):487–97., DOI 10.1016/s1470-2045(02)00818-5 Falk MH, Issels RD. Hyperthermia in oncology. Int J Hyperthermia, 2001, 17(1):1–18., DOI 10.1080/02656730150201552 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611114 EP 1611121 DI 10.3389/pore.2023.1611114 PG 8 ER PT J AU Gao, Sh Wei, G Hao, Y AF Gao, Shan Wei, Guanjing Hao, Yanrong TI Vitiligo-like lesions induced by cyclin-dependent kinase 4/ 6 inhibitor Palbociclib: a case report and literature review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE breast cancer; CDK4/6 inhibitors; Palbociclib; adverse events; vitiligo-like lesions ID breast cancer; CDK4/6 inhibitors; Palbociclib; adverse events; vitiligo-like lesions AB Endocrine therapy has played an essential role in hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2−) breast cancer. With the continuous development of endocrine targeting drugs, especially the emergence of selective cyclin-dependent kinase (CDK4/6) inhibitors, the overall survival time in patients with HR+HER2− advanced breast cancer has been greatly improved. Their adverse reactions also need more attention in response to the climbing number of CDK4/6 inhibitors. The common side effects of CDK4/6 inhibitors were hematological toxicity, diarrhea, and liver function damage. Skin toxicity related to CDK4/ 6 inhibitors was rare. We describe herein our preliminary observation of one HR+HER2− advanced metastatic breast cancer patient diagnosed with vitiligolike lesions after 10 months of taking Palbociclib. Hoping to share our experience to increase the clinician awareness of this unusual adverse and contribute to the information in the literature. C1 [Gao, Shan] People’s Hospital of Guangxi Zhuang Autonomous Region, Clinical Oncology Center, Medical Oncology Division 1Nanning, China. [Wei, Guanjing] People’s Hospital of Guangxi Zhuang Autonomous Region, Department of Dermatology and VenereologyNanning, China. [Hao, Yanrong] People’s Hospital of Guangxi Zhuang Autonomous Region, Clinical Oncology Center, Medical Oncology Division 1Nanning, China. RP Wei, G (reprint author), People’s Hospital of Guangxi Zhuang Autonomous Region, Department of Dermatology and Venereology, Nanning, China. EM 641634540@qq.com CR Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA-Cancer J Clin, 2022, 72:7–33., DOI 10.3322/caac.21708 Hortobagyi GN, Stemmer SM, Burris HA, Yap YS, Sonke GS, Hart L, et al. Overall survival with Ribociclib plus letrozole in advanced breast cancer. New Engl J Med, 2022, 386:942–50., DOI 10.1056/NEJMoa2114663 Sledge GJ, ToiM, Neven P, Sohn J, Inoue K, Pivot X, et al. The effect of Abemaciclib plus fulvestrant on overall survival in hormone receptor-positive, ERBB2-negative breast cancer that progressed on endocrine therapy-MONARCH 2: A randomized clinical trial. Jama Oncol, 2020, 6:116–24., DOI 10.1001/jamaoncol.2019.4782 Spring LM, Wander SA,Andre F, Moy B, TurnerNC, Bardia A. Cyclin-dependent kinase 4 and 6 inhibitors for hormone receptor-positive breast cancer: Past, present, and future. Lancet, 2020, 395:817–27., DOI 10.1016/S0140-6736(20)30165-3 Giuliano M, Schettini F, Rognoni C, Milani M, Jerusalem G, Bachelot T, et al. Endocrine treatment versus chemotherapy in postmenopausal women with hormone receptor-positive, HER2-negative, metastatic breast cancer: A systematic review and network meta-analysis. Lancet Oncol, 2019, 20:1360–9., DOI 10.1016/S1470-2045(19)30420-6 Silvestri M, Cristaudo A, Morrone A, Messina C, Bennardo L, Nistico SP, et al. Emerging skin toxicities in patients with breast cancer treated with new cyclindependent kinase 4/6 inhibitors: A systematic review. Drug Safety, 2021, 44:725–32., DOI 10.1007/s40264-021-01071-1 Silvestre TN, Aguilar MA, Echarri GM, Tabbara CS, Roman SJ, Gruber VF. Ribociclib-induced vitiligo: A case report. Dermatol Pract Conce, 2022, 12: e2022045., DOI 10.5826/dpc.1202a45 Chan OB, Su JC, Yazdabadi A, Chan A. Drug induced vitiligo-like depigmentation from a CDK 4/6 inhibitor. Asia-Pac J Clin Onco, 2022, 18: e154–e156., DOI 10.1111/ajco.13585 Gagnier JJ, Kienle G, Altman DG, Moher D, Sox H, Riley D, et al. The CARE guidelines: Consensus-based clinical case reporting guideline development. BMJ Case Rep, 2013, 2:38–43., DOI 10.7453/gahmj.2013.008 Finn RS,Martin M, Rugo HS, Jones S, Im SA, Gelmon K, et al. Palbociclib and letrozole in advanced breast cancer. New Engl J Med, 2016, 375:1925–36., DOI 10. 1056/NEJMoa1607303 Anjaneyan G, Keechilat P, Duraisamy P, Eapen M. Ribociclib-induced extensive vitiligo-like lesions: Possible pathomechanisms with clinical, dermoscopic and histological correlation. BMJ Case Rep, 2022, 15:e248782., DOI 10.1136/bcr-2022-248782 Papadopoulos L, Bor R, Legg C. Coping with the disfiguring effects of vitiligo: A preliminary investigation into the effects of cognitive-behavioural therapy. Br J Med Psychol, 1999, 72(3):385–96., DOI 10.1348/ 000711299160077 Abdeen M, Vusqa UT, Asawa P, Felton K, Rinchuse D, Khan C, et al. Venetoclax-induced vitiligo in a patient with chronic lymphocytic leukemia. Anti-Cancer Drug, 2022, 33:1167–70., DOI 10.1097/CAD.0000000000001350 Zhou M, Lin F, Xu W, Jin R, Xu A. Decreased SUMOylation of the retinoblastoma protein in keratinocytes during the pathogenesis of vitiligo. Mol Med Rep, 2018, 18:3469–75., DOI 10.3892/mmr.2018.9299 Sollena P, Nikolaou V, Soupos N, Kotteas E, Voudouri D, Stratigos AJ, et al. Vitiligo-like lesions in patients with advanced breast cancer treated with cyclinedependent kinases 4 and 6 inhibitors. Breast Cancer Res Treat, 2021, 185:247–53., DOI 10.1007/s10549-020-05914-w Romagnuolo M, Alberti VS, Riva D, Barberi F, Moltrasio C. Vitiligo-like lesions induced by cyclin-dependent kinase 4/6 inhibitor palbociclib. Int J Dermatol, 2023, 62:e98–e100., DOI 10.1111/ijd.16356 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611115 EP 1611119 DI 10.3389/pore.2023.1611115 PG 5 ER PT J TI Fibroblast activation protein-α expression in fibroblasts is common in the tumor microenvironment of colorectal cancer and may serve as a therapeutic target SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal cancer; tumor microenvironment; cancer-associated fibroblasts; fibroblastactivation protein; radioligand therapy ID colorectal cancer; tumor microenvironment; cancer-associated fibroblasts; fibroblastactivation protein; radioligand therapy AB Background: Colorectal cancer (CRC) is still one of the leading causes of cancer death worldwide, emphasizing the need for further diagnostic and therapeutic approaches. Cancer invasion and metastasis are affected by the tumor microenvironment (TME), with cancer-associated fibroblasts (CAF) being the predominant cellular component. An important marker for CAF is fibroblast activation protein-α (FAP) which has been evaluated as therapeutic target for, e.g., radioligand therapy. The aim of this study was to examine CRC regarding the FAP expression as a candidate for targeted therapy. Methods: 67 CRC, 24 adenomas, 18 tissue samples of inflammation sites and 28 non-neoplastic, non-inflammatory tissue samples of colonic mucosa were evaluated for immunohistochemical FAP expression of CAF in tissue microarrays. The results were correlated with clinicopathological data, tumor biology and concurrent expression of additional immunohistochemical parameters. Results: 53/67 (79%) CRC and 6/18 (33%) inflammatory tissue specimens showed expression of FAP. However, FAP was only present in 1/24 (4%) adenomas and absent in normal mucosa (0/28). Thus, FAP expression in CRC was significantly higher than in the other investigated groups. Within the CRC cohort, expression of FAP did not correlate with tumor stage, grading or the MSI status. However, it was observed that tumors exhibiting high immunohistochemical expression of Ki-67, CD3, p53, and β-Catenin showed a significantly higher incidence of FAP expression. Conclusion: In the crosstalk between tumor cells and TME, CAF play a key role in carcinogenesis and metastatic spread. Expression of FAP was detectable in the majority of CRC but nearly absent in precursor lesions and non-neoplastic, non-inflammatory tissue. This finding indicates that FAP has the potential to emerge as a target for new diagnostic and therapeutic concepts in CRC. Additionally, the association between FAP expression and other immunohistochemical parameters displays the interaction between different components of the TME and demands further investigation. CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/ caac.21492 Siegel RL,Miller KD, Jemal A. Cancer statistics. CA Cancer J Clin, 2018, 68(1): 7–30., DOI 10.3322/caac.21442 Yoshida GJ. Regulation of heterogeneous cancer-associated fibroblasts: the molecular pathology of activated signaling pathways. J Exp Clin Cancer Res, 2020, 39(1):112., DOI 10.1186/s13046-020-01611-0 Nurmik M, Ullmann P, Rodriguez F, Haan S, Letellier E. In search of definitions: Cancer-associated fibroblasts and their markers. Int J Cancer, 2020, 146(4):895–905., DOI 10.1002/ijc.32193 Sperb N, Tsesmelis M, Wirth T. Crosstalk between tumor and stromal cells in pancreatic ductal adenocarcinoma. Int J Mol Sci, 2020, 21(15):5486., DOI 10.3390/ ijms21155486 Chan TS, Shaked Y, Tsai KK. Targeting the interplay between cancer fibroblasts, mesenchymal stem cells, and cancer stem cells in desmoplastic cancers. Front Oncol, 2019, 9:688., DOI 10.3389/fonc.2019.00688 Erdogan B, Webb DJ. Cancer-associated fibroblasts modulate growth factor signaling and extracellular matrix remodeling to regulate tumor metastasis. Biochem Soc Trans, 2017, 45(1):229–36., DOI 10.1042/ bst20160387 Coto-Llerena M, Ercan C, Kancherla V, Taha-Mehlitz S, Eppenberger-Castori S, Soysal SD, et al. High expression of FAP in colorectal cancer is associated with angiogenesis and immunoregulation processes. Front Oncol, 2020, 10:979., DOI 10. 3389/fonc.2020.00979 Hurtado P, Martinez-Pena I, Pineiro R. Dangerous liaisons: circulating tumor cells, CTCs, and cancer-associated fibroblasts, CAFs). Cancers, 2020, 12(10):2861., DOI 10.3390/cancers12102861 Gonzalez-Villarreal CA, Quiroz-Reyes AG, Islas JF, Garza-Trevino EN. Colorectal cancer stem cells in the progression to liver metastasis. Front Oncol, 2020, 10:1511., DOI 10.3389/fonc.2020.01511 Hu JL, Wang W, Lan XL, Zeng ZC, Liang YS, Yan YR, et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol Cancer, 2019, 18(1):91., DOI 10.1186/s12943-019-1019-x Kanzaki R, Pietras K. Heterogeneity of cancer-associated fibroblasts: opportunities for precision medicine. Cancer Sci, 2020, 111(8):2708–17., DOI 10. 1111/cas.14537 Lindner T, Loktev A, Giesel F, Kratochwil C, Altmann A, Haberkorn U. Targeting of activated fibroblasts for imaging and therapy. EJNMMI Radiopharmacy Chem, 2019, 4(1):16., DOI 10.1186/s41181-019-0069-0 Fitzgerald AA, Weiner LM. The role of fibroblast activation protein in health and malignancy. Cancer Metastasis Rev, 2020, 39(3):783–803., DOI 10.1007/s10555- 020-09909-3 Jacob M, Chang L, Pure E. Fibroblast activation protein in remodeling tissues. Curr Mol Med, 2012, 12(10):1220–43., DOI 10.2174/156652412803833607 Rovedatti L, Di Sabatino A, Knowles CH, Sengupta N, Biancheri P, Corazza GR, et al. Fibroblast activation protein expression in Crohn’s disease strictures. Inflamm Bowel Dis, 2011, 17(5):1251–3., DOI 10.1002/ibd.21446 Liu F, Qi L, Liu B, Liu J, Zhang H, Che D, et al. Fibroblast activation protein overexpression and clinical implications in solid tumors: a meta-analysis. PloS one, 2015, 10(3):e0116683., DOI 10.1371/journal.pone.0116683 Solano-Iturri JD, Beitia M, Errarte P, Calvete-Candenas J, Etxezarraga MC, Loizate A, et al. Altered expression of fibroblast activation protein-α, FAP, in colorectal adenoma-carcinoma sequence and in lymph node and liver metastases. Aging, 2020, 12(11):10337–58., DOI 10.18632/aging.103261 Loktev A, Lindner T, Mier W, Debus J, Altmann A, Jager D, et al. A tumorimaging method targeting cancer-associated fibroblasts. J Nucl Med, 2018, 59(9): 1423–9., DOI 10.2967/jnumed.118.210435 Paauwe M, Schoonderwoerd MJA, Helderman R, Harryvan TJ, Groenewoud A, van Pelt GW, et al. Endoglin expression on cancer-associated fibroblasts regulates invasion and stimulates colorectal cancer metastasis. Clin Cancer Res, 2018, 24(24):6331–44., DOI 10.1158/1078-0432.Ccr-18-0329 Barrett RL, Pure E. Cancer-associated fibroblasts and their influence on tumor immunity and immunotherapy. eLife, 2020, 9:e57243., DOI 10.7554/eLife.57243 Yang M, Li J, Gu P, Fan X. The application of nanoparticles in cancer immunotherapy: targeting tumor microenvironment. Bioact Mater, 2021, 6(7): 1973–87., DOI 10.1016/j.bioactmat.2020.12.010 Greimelmaier K. Fibroblast activation protein-α expressing fibroblasts are common in and may serve as a therapeutic target in colorectal cancer. Deutsche Gesellschaft fur Pathologie e.V., 2021, 2021:307–8. Henry LR, Lee HO, Lee JS, Klein-Szanto A, Watts P, Ross EA, et al. Clinical implications of fibroblast activation protein in patients with colon cancer. Clin Cancer Res, 2007, 13(6):1736–41., DOI 10.1158/1078-0432.Ccr-06-1746 Stone JG, Robertson D, Houlston RS. Immunohistochemistry for MSH2 and MHL1: a method for identifying mismatch repair deficient colorectal cancer. J Clin Pathol, 2001, 54(6):484–7., DOI 10.1136/jcp.54.6.484 Wikberg ML, Edin S, Lundberg IV, Van Guelpen B, Dahlin AM, Rutegard J, et al. High intratumoral expression of fibroblast activation protein, FAP, in colon cancer is associated with poorer patient prognosis. Tumour Biol, 2013, 34(2): 1013–20., DOI 10.1007/s13277-012-0638-2 Son GM, Kwon MS, Shin DH, Shin N, Ryu D, Kang CD. Comparisons of cancer-associated fibroblasts in the intratumoral stroma and invasive front in colorectal cancer. Medicine, 2019, 98(18):e15164., DOI 10.1097/md. 0000000000015164 Moreno-Ruiz P, Corvigno S, Te GrootenhuisNC, La Fleur L, BackmanM, Strell C, et al. Stromal FAP is an independent poor prognosis marker in nonsmall cell lung adenocarcinoma and associated with p53 mutation. Lung Cancer, Amsterdam, Netherlands,, 2021, 155:10–9., DOI 10.1016/j.lungcan. 2021.02.028 Kanapathipillai M. Treating p53 mutant aggregation-associated cancer. Cancers, 2018, 10(6):154., DOI 10.3390/cancers10060154 Taciak B, Pruszynska I, Kiraga L, Bialasek M, Krol M. Wnt signaling pathway in development and cancer. J Physiol Pharmacol., 2018, 69(2)., DOI 10.26402/jpp. 2018.2.07 Herrera M, Mezheyeuski A, Villabona L, Corvigno S, Strell C, Klein C, et al. Prognostic interactions between FAP+ fibroblasts and CD8a+ T cells in colon cancer. Cancers, 2020, 12(11):3238., DOI 10.3390/cancers12113238 Pure E, Blomberg R. Pro-tumorigenic roles of fibroblast activation protein in cancer: Back to the basics. Oncogene., 2018, 37(32):4343–57., DOI 10.1038/s41388- 018-0275-3 FDA. 2021 SNMMI highlights lecture: oncology and therapy, Part 2. J Nucl Med, 2021, 62(2):11N–16N. GuptaM, Karthikeyan G, Choudhury PS, Babu Koyyala VP, SharmaM, Jain P, et al. A walk with Lu-177 PSMA: how close we have reached from bench to bedside? Cancer Invest, 2020, 38(8-9):486–92., DOI 10.1080/07357907.2020. 1811301 Lamberts LE, Menke-van der Houven van Oordt CW, ter Weele EJ, Bensch F, Smeenk MM, Voortman J, et al. ImmunoPET with anti-mesothelin antibody in patients with pancreatic and ovarian cancer before anti-mesothelin antibody-drug conjugate treatment. Clin Cancer Res, 2016, 22(7):1642–52., DOI 10.1158/1078- 0432.CCR-15-1272 Kobayashi K, Sasaki T, Takenaka F, Yakushiji H, Fujii Y, Kishi Y, et al. A novel PET imaging using, 6,, 4)Cu-labeled monoclonal antibody against mesothelin commonly expressed on cancer cells. J Immunol Res, 2015, 2015:268172., DOI 10. 1155/2015/268172 Mayerhoefer ME, Raderer M, Lamm W, Pichler V, Pfaff S, Weber M, et al. CXCR4 PET imaging of mantle cell lymphoma using [(68)Ga]pentixafor: Comparison with [(18)F]FDG-PET. Theranostics, 2021, 11(2):567–78., DOI 10. 7150/thno.48620 Walenkamp AME, Lapa C, Herrmann K,Wester HJ. CXCR4 ligands: the next big hit? J Nucl Med, 2017, 58(2):77S–82S., DOI 10.2967/jnumed.116.186874 Hayrapetian A, Girgis MD, Yanagawa J, French SW, Schelbert HR, Auerbach MS, et al. Incidental detection of elastofibroma dorsi with 68Ga-FAPI-46 and 18Ffdg PET/CT in a patient with esophageal cancer. Clin Nucl Med, 2021, 46(2): e86–e87., DOI 10.1097/RLU.0000000000003218 Pang Y, Wei J, Shang Q, Zhao L, Chen H. 68Ga-Fibroblast activation protein inhibitor, a promising radiopharmaceutical in PET/CT to detect the primary and metastatic lesions of chromophobe renal cell carcinoma. Clin Nucl Med, 2021, 46(2):177–9., DOI 10.1097/RLU.0000000000003454 Watabe T, Liu Y, Kaneda-Nakashima K, Shirakami Y, Lindner T, Ooe K, et al. Theranostics targeting fibroblast activation protein in the tumor stroma:, 64)Cuand, 225)Ac-labeled FAPI-04 in pancreatic cancer xenograft mouse models. J Nucl Med, 2020, 61(4):563–9., DOI 10.2967/jnumed.119.233122 Kratochwil C, Flechsig P, Lindner T, Abderrahim L, Altmann A,Mier W, et al. 68)Ga-FAPI PET/CT: tracer uptake in 28 different kinds of cancer. J Nucl Med, 2019, 60(6):801–5., DOI 10.2967/jnumed.119.227967 Ballal S, Yadav MP, Kramer V, Moon ES, Roesch F, Tripathi M, et al. A theranostic approach of [(68)Ga]Ga-DOTA.SA.FAPi PET/CT-guided [(177)Lu] Lu-DOTA.SA.FAPi radionuclide therapy in an end-stage breast cancer patient: new frontier in targeted radionuclide therapy. Eur J NuclMed Mol Imaging, 2021, 48(3): 942–4., DOI 10.1007/s00259-020-04990-w Syed M, Flechsig P, Liermann J, Windisch P, Staudinger F, Akbaba S, et al. Fibroblast activation protein inhibitor, FAPI, PET for diagnostics and advanced targeted radiotherapy in head and neck cancers. Eur J NuclMed Mol Imaging, 2020, 47(12):2836–45., DOI 10.1007/s00259-020-04859-y Lindner T, Altmann A, Kramer S, Kleist C, Loktev A, Kratochwil C, et al. Design and development of, 99m)Tc-labeled FAPI tracers for SPECT imaging and, 188)Re therapy. J Nucl Med, 2020, 61(10):1507–13., DOI 10.2967/jnumed.119. 239731 Loktev A, Lindner T, Burger EM, Altmann A, Giesel F, Kratochwil C, et al. Development of fibroblast activation protein-targeted radiotracers with improved tumor retention. J Nucl Med, 2019, 60(10):1421–9., DOI 10.2967/ jnumed.118.224469 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611163 EP 1611171 DI 10.3389/pore.2023.1611163 PG 9 ER PT J AU Rabindran, B Corben, A AF Rabindran, Beryl Corben, D. Adriana TI Wide-field optical coherence tomography for microstructural analysis of key tissue types: a proof-of-concept evaluation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE histopathology; breast-conserving surgery; histology; optical coherence tomography; intraoperative margin assessment ID histopathology; breast-conserving surgery; histology; optical coherence tomography; intraoperative margin assessment AB Introduction: The presence of positive margins following tumor resection is a frequent cause of re-excision surgery. Nondestructive, real-time intraoperative histopathological imaging methods may improve margin status assessment at the time of surgery; optical coherence tomography (OCT) has been identified as a potential solution but has not been tested with the most common tissue types in surgical oncology using a single, standardized platform. Methods: This was a proof-of-concept evaluation of a novel device that employs wide-field OCT (WF-OCT; OTIS 2.0 System) to image tissue specimens. Various cadaveric tissues were obtained from a single autopsy and were imaged with WF-OCT then processed for permanent histology. The quality and resolution of the WF-OCT images were evaluated and compared to histology and with images in previous literature. Results: A total of 30 specimens were collected and tissue-specific microarchitecture consistent with previous literature were identified on both WF-OCT images and histology slides for all specimens, and corresponding sections were correlated. Application of vacuum pressure during scanning did not affect specimen integrity. On average, specimens were scanned at a speed of 10.3 s/cm2 with approximately three features observed per tissue type. Conclusion: The WF-OCT images captured in this study displayed the key features of the most common human tissue types encountered in surgical oncology with utility comparable to histology, confirming the utility of an FDAcleared imaging platform. With further study, WF-OCT may have the potential to bridge the gap between the immediate information needs of the operating room and the longer timeline inherent to histology workflow. C1 [Rabindran, Beryl] Perimeter Medical Imaging AIToronto, ON, Canada. [Corben, D. Adriana] Icahn School of Medicine at Mount SinaiNew York, NY, USA. RP Rabindran, B (reprint author), Perimeter Medical Imaging AI, Toronto, Canada. EM baugustine@perimetermed.com CR American Cancer Society. Cancer facts & Figures 2021, 2021). Available from: https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/ cancer-facts-figures-2022.html, Accessed January 4, 2023). Miller KD, Nogueira L, Mariotto AB, Rowland JH, Yabroff KR, Alfano CM, et al. Cancer treatment and survivorship statistics, 2019. CA Cancer J Clin, 2019, 69: 363–85., DOI 10.3322/caac.21565 Clarke M, Collins R, Darby S, Davies C, Elphinstone P, Evans V, et al. Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: An overview of the randomised trials. Lancet, 2005, 366:2087–106., DOI 10.1016/S0140-6736(05)67887-7 Moran MS, Schnitt SJ, Giuliano AE, Harris JR, Khan SA, Horton J, et al. Society of Surgical Oncology-American Society for Radiation Oncology consensus guideline on margins for breast-conserving surgery with whole-breast irradiation in stages I and II invasive breast cancer. Ann Surg Oncol, 2014, 21: 704–16., DOI 10.1245/s10434-014-3481-4 Morrow M, Van Zee KJ, Solin LJ, Houssami N, Chavez-MacGregor M, Harris JR, et al. Society of surgical oncology-American society for radiation oncology- American society of clinical oncology consensus guideline on margins for breastconserving surgery with whole-breast irradiation in ductal carcinoma in situ. Ann Surg Oncol, 2016, 23:3801–10., DOI 10.1245/s10434-016-5449-z Voskuil FJ, Vonk J, van der Vegt B, Kruijff S, Ntziachristos V, van der Zaag PJ, et al. Intraoperative imaging in pathology-assisted surgery. Nat Biomed Eng, 2022, 6:503–14., DOI 10.1038/s41551-021-00808-8 Schnitt SJ, Moran MS, Giuliano AE. Lumpectomy margins for invasive breast cancer and ductal carcinoma in situ: Current guideline recommendations, their implications, and impact. J Clin Oncol, 2020, 38:2240–5., DOI 10.1200/JCO.19.03213 Atkins J, Al Mushawah F, Appleton CM, Cyr AE, Gillanders WE, Aft RL, et al. Positive margin rates following breast-conserving surgery for stage I-iii breast cancer: Palpable versus nonpalpable tumors. J Surg Res, 2012, 177:109–15., DOI 10. 1016/j.jss.2012.03.045 Balch GC, Mithani SK, Simpson JF, Kelley MC. Accuracy of intraoperative gross examination of surgical margin status in women undergoing partial mastectomy for breast malignancy. Am Surg, 2005, 71:22–8., DOI 10.1177/ 000313480507100104 Brown JQ, Bydlon TM, Richards LM, Yu B, Kennedy SA, Geradts J, et al. Optical assessment of tumor resection margins in the breast. IEEE J Sel Top Quan Electron, 2010, 16:530–44., DOI 10.1109/jstqe.2009.2033257 Fleming FJ, Hill AD, Mc Dermott EW, O’Doherty A, O’Higgins NJ, Quinn CM. Intraoperative margin assessment and re-excision rate in breast conserving surgery. Eur J Surg Oncol, 2004, 30:233–7., DOI 10.1016/j.ejso.2003.11.008 McCahill LE, Single RM, Aiello Bowles EJ, Feigelson HS, James TA, Barney T, et al. Variability in reexcision following breast conservation surgery. JAMA, 2012, 307:467–75., DOI 10.1001/jama.2012.43 Bolshinsky V, Lin MJ, Serpell J, Leung M, Wolfe R, McLean C, et al. Frequency of residual melanoma in wide local excision, WLE, specimens after complete excisional biopsy. J Am Acad Dermatol, 2016, 74:102–7., DOI 10.1016/j.jaad.2015. 08.065 Drexler W, Fujimoto JG. Optical coherence tomography: Technology and applications. Switzerland AG: Springer International Publishing, 2015). Huang D, Swanson EA, Lin CP, Schuman JS, Stinson WG, Chang W, et al. Optical coherence tomography. Science, 1991, 254:1178–81., DOI 10.1126/science. 1957169 Subban V, Raffel OC. Optical coherence tomography: Fundamentals and clinical utility. Cardiovasc Diagn Ther, 2020, 10:1389–414., DOI 10.21037/cdt- 20-253 Wang J, Xu Y, Boppart SA. Review of optical coherence tomography in oncology. J Biomed Opt, 2017, 22:1–23., DOI 10.1117/1.JBO.22.12.121711 Zysk AM, Nguyen FT, Oldenburg AL, Marks DL, Boppart SA. Optical coherence tomography: A review of clinical development from bench to bedside. J Biomed Opt, 2007, 12:051403., DOI 10.1117/1.2793736 Tung ET, Yim KHC, Li CL, Cheung CY, Chan YC. Optical coherence tomography in peripheral arterial disease: A systematic review. Int J Clin Pract, 2021, 75:e14628., DOI 10.1111/ijcp.14628 Badhey AK, Schwarz JS, Laitman BM, Veremis BM, Westra WH, YaoM, et al. Intraoperative use of wide-field optical coherence tomography to evaluate tissue microstructure in the oral cavity and oropharynx. JAMA Otolaryngol Head Neck Surg, 2022, 149:71–8., DOI 10.1001/jamaoto.2022.3763 DuPree BB, PapezMJ, Pirruccello E, Hassenflug A. Potential utility of adjunct imaging with wide-field optical coherence tomography for gross and microscopic evaluation of breast specimens in real-time in the operating suite. Indian J Surg, 2021, 84:751–6., DOI 10.1007/s12262-021-03079-4 Schmidt H, Connolly C, Jaffer S, Oza T,Weltz CR, Port ER, et al. Evaluation of surgically excised breast tissue microstructure using wide-field optical coherence tomography. Breast J, 2020, 26:917–23., DOI 10.1111/tbj.13663 Ha R, Friedlander LC, Hibshoosh H, Hendon C, Feldman S, Ahn S, et al. Optical coherence tomography: A novel imaging method for post-lumpectomy breast margin assessment-a multi-reader study. Acad Radiol, 2018, 25:279–87., DOI 10.1016/j.acra.2017.09.018 Erickson-Bhatt SJ, Nolan RM, Shemonski ND, Adie SG, Putney J, Darga D, et al. Real-time imaging of the resection bed using a handheld probe to reduce incidence of microscopic positive margins in cancer surgery. Cancer Res, 2015, 75: 3706–12., DOI 10.1158/0008-5472.CAN-15-0464 Conti de Freitas LC, Phelan E, Liu L, Gardecki J, Namati E,Warger WC, et al. Optical coherence tomography imaging during thyroid and parathyroid surgery: A novel system of tissue identification and differentiation to obviate tissue resection and frozen section. Head Neck, 2014, 36:1329–34., DOI 10.1002/hed.23452 Gan Y, Tsay D, Amir SB, Marboe CC, Hendon CP. Automated classification of optical coherence tomography images of human atrial tissue. J Biomed Opt, 2016, 21:101407., DOI 10.1117/1.JBO.21.10.101407 Gan Y, Yao X, Chang E, Amir SB, Hibshoosh H, Feldman S, et al. Comparative study of texture features in OCT images at different scales for human breast tissue classification. In: 2016 38th Annual International Conference of the IEEE Engineering in Medicine and Biology Society; 16-20 August 2016; Orlando, FL, USA, 2016). p. 3926–9. Hariri LP, Mino-Kenudson M, Lanuti M, Miller AJ, Mark EJ, Suter MJ. Diagnosing lung carcinomas with optical coherence tomography. Ann Am Thorac Soc, 2015, 12:193–201., DOI 10.1513/AnnalsATS.201408-370OC Iftimia N, Cizginer S, Deshpande V, Pitman M, Tatli S, Iftimia NA, et al. Differentiation of pancreatic cysts with optical coherence tomography, OCT, imaging: An ex vivo pilot study. Biomed Opt Express, 2011, 2:2372–82., DOI 10. 1364/BOE.2.002372 Jackle S, Gladkova N, Feldchtein F, Terentieva A, Brand B, Gelikonov G, et al. In vivo endoscopic optical coherence tomography of the human gastrointestinal tract-toward optical biopsy. Endoscopy, 2000, 32:743–9., DOI 10.1055/s-2000-7711 Lee HC, Zhou C, Cohen DW, Mondelblatt AE, Wang Y, Aguirre AD, et al. Integrated optical coherence tomography and optical coherence microscopy imaging of ex vivo human renal tissues. J Urol, 2012, 187:691–9., DOI 10.1016/j. juro.2011.09.149 Poneros JM, Tearney GJ, Shiskov M, Kelsey PB, Lauwers GY, Nishioka NS, et al. Optical coherence tomography of the biliary tree during ERCP. Gastrointest Endosc, 2002, 55:84–8., DOI 10.1067/mge.2002.120098 St John ER, Al-Khudairi R, Ashrafian H, Athanasiou T, Takats Z, Hadjiminas DJ, et al. Diagnostic accuracy of intraoperative techniques for margin assessment in breast cancer surgery: A meta-analysis. Ann Surg, 2017, 265:300–10., DOI 10.1097/ SLA.0000000000001897 Gray RJ, Pockaj BA, Garvey E, Blair S. Intraoperative margin management in breast-conserving surgery: A systematic review of the literature. Ann Surg Oncol, 2018, 25:18–27., DOI 10.1245/s10434-016-5756-4 Schnabel F, Boolbol SK, Gittleman M, Karni T, Tafra L, Feldman S, et al. A randomized prospective study of lumpectomy margin assessment with use of MarginProbe in patients with nonpalpable breast malignancies. Ann Surg Oncol, 2014, 21:1589–95., DOI 10.1245/s10434-014-3602-0 Ali ZA, Karimi Galougahi K,Maehara A, Shlofmitz RA, Ben-Yehuda O, Mintz GS, et al. Intracoronary optical coherence tomography 2018: Current status and future directions. JACC Cardiovasc Interv, 2017, 10:2473–87., DOI 10.1016/j.jcin. 2017.09.042 Ang M, Tan ACS, Cheung CMG, Keane PA, Dolz-Marco R, Sng CCA, et al. Optical coherence tomography angiography: A review of current and future clinical applications. Graefes Arch Clin Exp Ophthalmol, 2018, 256:237–45., DOI 10.1007/ s00417-017-3896-2 Fujii K, Kawakami R, Hirota S. Histopathological validation of optical coherence tomography findings of the coronary arteries. J Cardiol, 2018, 72: 179–85., DOI 10.1016/j.jjcc.2018.03.003 Vakoc BJ, Fukumura D, Jain RK, Bouma BE. Cancer imaging by optical coherence tomography: Preclinical progress and clinical potential. Nat Rev Cancer, 2012, 12:363–8., DOI 10.1038/nrc3235 Nguyen FT, Zysk AM, Chaney EJ, Kotynek JG, Oliphant UJ, Bellafiore FJ, et al. Intraoperative evaluation of breast tumor margins with optical coherence tomography. Cancer Res, 2009, 69:8790–6., DOI 10.1158/0008-5472.CAN-08-4340 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611167 EP 1611176 DI 10.3389/pore.2023.1611167 PG 10 ER PT J AU Danos, K Horvath, A Halasz, J Tamas, L Polony, G AF Danos, Kornel Horvath, Angela Halasz, Judit Tamas, Laszlo Polony, Gabor TI Patient delay and its clinical significance among head and neck cancer patients in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE survival; diagnosis; head and neck cancer; patient delay; symptom ID survival; diagnosis; head and neck cancer; patient delay; symptom AB Introduction: Head and neck cancers represent a major health problem in Hungary. With their high incidence and mortality rates, Hungary is one of the world leaders in these indicators. The length of patient delay, defined as time from onset of symptoms to first medical consultation, is unknown in Hungarian patients with head and neck cancer. We aimed to use a representative sample of the Hungarian head and neck cancer patient population to determine patient delay according to disease localization and stage and to identify correlations with other clinical parameters. Methods: In our retrospective study, we reviewed patient documentation. For the inclusion, the patients had to be diagnosed with malignant tumors of the oral cavity, oropharynx, hypopharynx or larynx at the Department Head and Neck Surgery of Semmelweis University between 2012 and 2017. Results: We identified 236 patientswho met the inclusion criteria. The median delay was 9.5weeks (range0–209 weeks) and the mean delay of patientswas 17.57weeks (SD 23.67). There was a significant difference in patient delay data by location. Among glottic cancers, the mostcommondiagnosiswas an early stage (67%), comparedwith other localizations, including most commonly the oropharynx (81%) and hypopharynx (80%), where a locoregionally advanced stage was more frequent. Discussion: Compared to data from different countries, the delay of Hungarian patients with head and neck cancer is significantly longer, which may contribute to the high mortality in Hungary. Screening and patient education in high-risk groups could contribute to earlier diagnosis and thus improve prognosis. C1 [Danos, Kornel] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Horvath, Angela] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Halasz, Judit] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Tamas, Laszlo] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. [Polony, Gabor] Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck SurgeryBudapest, Hungary. RP Horvath, A (reprint author), Semmelweis University, Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Budapest, Hungary. EM horvathangela11@gmail.com CR International Agency for Research on Cancer. Globocan: Global cancer observatory, 2020). Retrieved from: https://gco.iarc.fr/today/online-analysismultibars? v=2020&mode=population&mode_population=countries&population= 900&populations=900&key=crude_rate&sex=0&cancer=39&type=0&statistic= 5&prevalence=0&population_group=0&ages_group%5B%5D=0&ages_group%5B %5D=17&nb_items=10&group_cancer=0&include_nmsc=0&include_nmsc_ other=1&type_multiple=%257B%2522inc%2522%253Afalse%252C%2522mort% 2522%253Atrue%252C%2522prev%2522%253Afalse%257D&orientation= horizontal&type_sort=1&type_nb_items=%257B%2522top%2522%253Atrue% 252C%2522bottom%2522%253Afalse%257D#collapse-others, Accessed July 5, 2023). Neal RD. Do diagnostic delays in cancer matter? Br J Cancer, 2009, 101(2): S9–S12., DOI 10.1038/sj.bjc.6605384 Goy J, Hall SF, Feldman-Stewart D, Groome PA. Diagnostic delay and disease stage in head and neck cancer: A systematic review. Laryngoscope, 2009, 119(5): 889–98., DOI 10.1002/lary.20185 Rozniatowski O, Reich M, Mallet Y, Penel N, Fournier C, Lefebvre JL. Psychosocial factors involved in delayed consultation by patients with head and neck cancer. Head Neck, 2005, 27(4):274–80., DOI 10.1002/hed.20157 Franceschi S, Barzan L, Talamini R. Screening for cancer of the head and neck: If not now, when? Oral Oncol, 1997, 33(5):313–6., DOI 10.1016/s1368-8375(97, 00034-1 Reich M, Licitra L, Vermorken JB, Bernier J, Parmar S, GolusinskiW, et al. Best practice guidelines in the psychosocial management of HPV-related head and neck cancer: Recommendations from the European head and neck cancer society’s make Sense campaign. Ann Oncol, 2016, 27(10):1848–54., DOI 10. 1093/annonc/mdw272 Edge SB, Compton CC. The American joint committee on cancer: The 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol, 2010, 17(6):1471–4., DOI 10.1245/s10434-010-0985-4 Dok R, Nuyts S. HPV positive head and neck cancers: Molecular pathogenesis and evolving treatment strategies. Cancers, Basel,, 2016, 8(4):41., DOI 10.3390/ cancers8040041 Teppo H, Alho OP. Relative importance of diagnostic delays in different head and neck cancers. Clin Otolaryngol, 2008, 33(4):325–30., DOI 10.1111/j.1749-4486. 2008.01704.x Ambrosch P, Fazel A. Functional organ preservation in laryngeal and hypopharyngeal cancer. GMS Curr Top Otorhinolaryngol Head Neck Surg, 2011, 90(1):Doc02–109. Laryngorhinootologie., DOI 10.3205/cto000075 Szentkuti G, Danos K, Brauswetter D, Kiszner G, Krenacs T, Csako L, et al. Correlations between prognosis and regional biomarker profiles in head and neck squamous cell carcinomas. Pathol Oncol Res, 2015, 21(3):643–50., DOI 10.1007/ s12253-014-9869-4 Chen RC, Khorsandi AS, Shatzkes DR, Holliday RA. The radiology of referred otalgia. AJNR Am J Neuroradiol, 2009, 30(10):1817–23., DOI 10.3174/ajnr.A1605 Goldenberg D, Begum S, Westra WH, Khan Z, Sciubba J, Pai SI, et al. Cystic lymph node metastasis in patients with head and neck cancer: An HPV-associated phenomenon. Head Neck, 2008, 30(7):898–903., DOI 10.1002/hed.20796 Kassirian S, Dzioba A, Hamel S, Patel K, Sahovaler A, Palma DA, et al. Delay in diagnosis of patients with head-and-neck cancer in Canada: Impact of patient and provider delay. Curr Oncol, 2020, 27(5):e467–e77., DOI 10.3747/co.27.6547 Felippu AW, Freire EC, Silva Rde A, Guimaraes AV, Dedivitis RA. Impact of delay in the diagnosis and treatment of head and neck cancer. Braz J Otorhinolaryngol, 2016, 82(2):140–3., DOI 10.1016/j.bjorl.2015.10.009 Amir Z, Kwan SY, Landes D, Feber T,Williams SA. Diagnostic delays in head and neck cancers. Eur J Cancer Care, Engl,, 1999, 8(4):198–203., DOI 10.1046/j. 1365-2354.1999.00165.x Ganesan S, Sivagnanganesan S, Thulasingam M, Karunanithi G, Kalaiarasi K, Ravichandran S, et al. Diagnostic delay for head and neck cancer in South India: A mixed-methods study. Asian Pac J Cancer Prev, 2020, 21(6):1673–8., DOI 10.31557/ APJCP.2020.21.6.1673 Smith MM, Abrol A, Gardner GM. Assessing delays in laryngeal cancer treatment. Laryngoscope, 2016, 126(7):1612–5., DOI 10.1002/lary.25734 Liao DZ, Schlecht NF, Rosenblatt G, Kinkhabwala CM, Leonard JA, Ference RS, et al. Association of delayed time to treatment initiation with overall survival and recurrence among patients with head and neck squamous cell carcinoma in an underserved urban population. JAMA Otolaryngol Head Neck Surg, 2019, 145(11): 1001–9., DOI 10.1001/jamaoto.2019.2414 Sargeran K, Murtomaa H, Safavi SM, Teronen O. Delayed diagnosis of oral cancer in Iran: Challenge for prevention. Oral Health Prev Dent, 2009, 7(1):69–76., DOI 10.3290/j.ohpd.a15270 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611206 EP 1611215 DI 10.3389/pore.2023.1611206 PG 10 ER PT J AU Jiang, W Hu, K Liu, X Gao, J Zhu, L AF Jiang, Wenru Hu, Kangyao Liu, Xiaofei Gao, Jili Zhu, Liping TI Single-cell transcriptome analysis reveals the clinical implications of myeloid-derived suppressor cells in head and neck squamous cell carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE head and neck squamous cell carcinoma; myeloid-derived suppressor cell; single cell RNA sequencing (scRNA-seq); prognosis; immunotherapeutic ID head and neck squamous cell carcinoma; myeloid-derived suppressor cell; single cell RNA sequencing (scRNA-seq); prognosis; immunotherapeutic AB Head and neck squamous cell carcinoma (HNSC) is the most common malignant tumor that arises in the epithelium of the head and neck regions. Myeloid-derived suppressor cells (MDSCs) are one of the tumor-infiltrating immune cell populations, which play a powerful role in inhibiting antitumor immune response. Herein, we employed a single-cell RNA sequencing (scRNA-seq) dataset to dissect the heterogeneity of myeloid cells. We found that SPP1+ tumor-associated macrophages (TAMs) and MDSCs were the most abundant myeloid cells in the microenvironment. By cell cluster deconvolution from bulk RNA-seq datasets of larger patient groups, we observed that highly-infiltrated MDSC was a poor prognostic marker for patients’ overall survival (OS) probabilities. To better apply the MDSC OS prediction values, we identified a set of six MDSC-related genes (ALDOA, CD52, FTH1, RTN4, SLC2A3, and TNFAIP6) as the prognostic signature. In both training and test cohorts, MDSC-related prognostic signature showed a promising value for predicting patients’ prognosis outcomes. Further parsing the ligand-receptor pairs of intercellular communications by CellChat, we found that MDSCs could frequently interact with cytotoxic CD8+ T cells, SPP1+ TAMs, and endothelial cells. These interactions likely contributed to the establishment of an immunosuppressive microenvironment and the promotion of tumor angiogenesis. Our findings suggest that targeting MDSCs may serve as an alternative and promising target for the immunotherapy of HNSC. C1 [Jiang, Wenru] Harbin First Hospital, Department of Implant and ProsthodonticsHarbin, China. [Hu, Kangyao] Harbin First Hospital, Department of Implant and ProsthodonticsHarbin, China. [Liu, Xiaofei] Harbin First Hospital, Department of Implant and ProsthodonticsHarbin, China. [Gao, Jili] Harbin First Hospital, Department of Implant and ProsthodonticsHarbin, China. [Zhu, Liping] Harbin First Hospital, Department of Implant and ProsthodonticsHarbin, China. RP Zhu, L (reprint author), Harbin First Hospital, Department of Implant and Prosthodontics, Harbin, China. EM 1920636476@qq.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE, Grandis JR. Head and neck squamous cell carcinoma. Nat Rev Dis Primers, 2020, 6(1):92., DOI 10. 1038/s41572-020-00224-3 Pulte D, Brenner H. Changes in survival in head and neck cancers in the late 20th and early 21st century: A period analysis. Oncologist, 2010, 15(9):994–1001., DOI 10.1634/theoncologist.2009-0289 ChenW, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA: A Cancer J Clinicians, 2016, 66(2):115–32., DOI 10.3322/caac. 21338 Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol, 2021, 21(8):485–98., DOI 10.1038/s41577-020-00490-y Al-Khami AA, Rodriguez PC, Ochoa AC. Metabolic reprogramming of myeloid-derived suppressor cells, MDSC, in cancer. Oncoimmunology, 2016, 5(8):e1200771., DOI 10.1080/2162402X.2016.1200771 Ma P, Beatty PL, McKolanis J, Brand R, Schoen RE, Finn OJ. Circulating myeloid derived suppressor cells, MDSC, that accumulate in premalignancy share phenotypic and functional characteristics with MDSC in cancer. Front Immunol, 2019, 10:1401., DOI 10.3389/fimmu.2019.01401 Albeituni SH, Ding C, Yan J. Hampering immune suppressors: Therapeutic targeting of myeloid-derived suppressor cells in cancer. Cancer J, Sudbury, Mass,, 2013, 19(6):490–501., DOI 10.1097/PPO.0000000000000006 NomanMZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSCmediated T cell activation. J Exp Med, 2014, 211(5):781–90., DOI 10.1084/jem. 20131916 Schouppe E, Mommer C, Movahedi K, Laoui D, Morias Y, Gysemans C, et al. Tumor-induced myeloid-derived suppressor cell subsets exert either inhibitory or stimulatory effects on distinct CD 8+ T-cell activation events. Eur J Immunol, 2013, 43(11):2930–42., DOI 10.1002/eji.201343349 Fultang L, Panetti S, Ng M, Collins P, Graef S, Rizkalla N, et al. MDSC targeting with Gemtuzumab ozogamicin restores T cell immunity and immunotherapy against cancers. EBioMedicine, 2019, 47:235–46., DOI 10.1016/j. ebiom.2019.08.025 Li T, Liu T, Zhu W, Xie S, Zhao Z, Feng B, et al. TargetingMDSC for immunecheckpoint blockade in cancer immunotherapy: Current progress and new prospects. Clin Med Insights: Oncol, 2021, 15:11795549211035540., DOI 10.1177/ 11795549211035540 Ghansah T. A novel strategy for modulation of MDSC to enhance cancer immunotherapy. Oncoimmunology, 2012, 1(6):984–5., DOI 10.4161/onci.20201 Califano JA, Khan Z, Noonan KA, Rudraraju L, Zhang Z, Wang H, et al. Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res, 2015, 21(1):30–8., DOI 10.1158/1078- 0432.CCR-14-1716 Weed DT, Vella JL, Reis IM, De la Fuente AC, Gomez C, Sargi Z, et al. Tadalafil reduces myeloid-derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res, 2015, 21(1):39–48., DOI 10.1158/1078-0432.CCR-14- 1711 Pang X, Fan HY, Tang YL, Wang SS, Cao MX, Wang HF, et al. Myeloid derived suppressor cells contribute to the malignant progression of oral squamous cell carcinoma. PLoS One, 2020, 15(2):e0229089., DOI 10.1371/journal.pone. 0229089 Peng Y, Xiao L, Rong H, Ou Z, Cai T, Liu N, et al. Single-cell profiling of tumor-infiltrating TCF1/TCF7+ T cells reveals a T lymphocyte subset associated with tertiary lymphoid structures/organs and a superior prognosis in oral cancer. Oral Oncol, 2021, 119:105348., DOI 10.1016/j. oraloncology.2021.105348 Wichmann G, Rosolowski M, Krohn K, Kreuz M, Boehm A, Reiche A, et al. The role of HPV RNA transcription, immune response-related gene expression and disruptive TP53 mutations in diagnostic and prognostic profiling of head and neck cancer. Int J Cancer, 2015, 137(12):2846–57., DOI 10.1002/ijc.29649 Lohavanichbutr P, Mendez E, Holsinger FC, Rue TC, Zhang Y, Houck J, et al. A 13-gene signature prognostic of HPV-negative OSCC: Discovery and external validation. Clin Cancer Res, 2013, 19(5):1197–203., DOI 10.1158/1078-0432.CCR- 12-2647 Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol, 2018, 36(5):411–20., DOI 10.1038/nbt.4096 Wolock SL, Lopez R, Klein AM. Scrublet: Computational identification of cell doublets in single-cell transcriptomic data. Cell Syst, 2019, 8(4):281–91.e9., DOI 10. 1016/j.cels.2018.11.005 Kamath T, Abdulraouf A, Burris SJ, Langlieb J, Gazestani V, Nadaf NM, et al. Single-cell genomic profiling of human dopamine neurons identifies a population that selectively degenerates in Parkinson’s disease. Nat Neurosci, 2022, 25(5): 588–95., DOI 10.1038/s41593-022-01061-1 Huang L, Wang X, Pei S, Li X, Dong L, Bian X, et al. Single-cell profiling reveals sustained immune infiltration, surveillance, and tumor heterogeneity in infiltrative BCC. J Invest Dermatol, 2023)., DOI 10.1016/j.jid.2023.04.020 Li X, Zhao S, Bian X, Zhang L, Lu L, Pei S, et al. Signatures of EMT, immunosuppression, and inflammation in primary and recurrent human cutaneous squamous cell carcinoma at single-cell resolution. Theranostics, 2022, 12(17):7532–49., DOI 10.7150/thno.77528 Chu T, Wang Z, Pe’er D, Danko CG. Cell type and gene expression deconvolution with BayesPrism enables Bayesian integrative analysis across bulk and single-cell RNA sequencing in oncology. Nat Cancer, 2022, 3(4):505–17., DOI 10.1038/s43018-022-00356-3 Gao R, Bai S, Henderson YC, Lin Y, Schalck A, Yan Y, et al. Delineating copy number and clonal substructure in human tumors from single-cell transcriptomes. Nat Biotechnol, 2021, 39(5):599–608., DOI 10.1038/s41587-020-00795-2 Gulati GS, Sikandar SS, Wesche DJ, Manjunath A, Bharadwaj A, Berger MJ, et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science, 2020, 367(6476):405–11., DOI 10.1126/science.aax0249 Aibar S, Gonzalez-Blas CB, Moerman T, Huynh-Thu VA, Imrichova H, Hulselmans G, et al. Scenic: Single-cell regulatory network inference and clustering. Nat Methods, 2017, 14(11):1083–6., DOI 10.1038/nmeth.4463 Wang J, Yin X, Zhang YQ, Ji X. Identification and validation of a novel immune-related four-lncRNA signature for lung adenocarcinoma. Front Genet, 2021, 12:639254., DOI 10.3389/fgene.2021.639254 Pan X, Zhang C, Wang J, Wang P, Gao Y, Shang S, et al. Epigenome signature as an immunophenotype indicator prompts durable clinical immunotherapy benefits in lung adenocarcinoma. Brief Bioinform, 2022, 23(1):bbab481., DOI 10. 1093/bib/bbab481 Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan C-H, et al. Inference and analysis of cell-cell communication using CellChat. Nat Commun, 2021, 12(1):1088., DOI 10.1038/s41467-021-21246-9 Chu M, Su YX, Wang L, Zhang TH, Liang YJ, Liang LZ, et al.Myeloid-derived suppressor cells contribute to oral cancer progression in 4NQO-treated mice. Oral Dis, 2012, 18(1):67–73., DOI 10.1111/j.1601-0825.2011.01846.x Jiang Y, Wang C, Wang Y, Zhang W, Liu L, Cheng J. Prognostic role of CD11b(+, myeloid-derived suppressor cells in oral squamous cell carcinoma. Arch Med Sci, 2023, 19(1):171–9., DOI 10.5114/aoms/116683 Qi J, Sun H, Zhang Y, Wang Z, Xun Z, Li Z, et al. Single-cell and spatial analysis reveal interaction of FAP+ fibroblasts and SPP1+ macrophages in colorectal cancer. Nat Commun, 2022, 13(1):1742., DOI 10.1038/s41467-022- 29366-6 Georgoudaki AM, Prokopec KE, Boura VF, Hellqvist E, Sohn S, Ostling J, et al. Reprogramming tumor-associated macrophages by antibody targeting inhibits cancer progression and metastasis. Cell Rep, 2016, 15(9):2000–11., DOI 10.1016/j. celrep.2016.04.084 Groth C, Hu X, Weber R, Fleming V, Altevogt P, Utikal J, et al. Immunosuppression mediated by myeloid-derived suppressor cells, MDSCs, during tumour progression. Br J Cancer, 2019, 120(1):16–25., DOI 10.1038/ s41416-018-0333-1 Li K, Shi H, Zhang B, Ou X, Ma Q, Chen Y, et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduction Targeted Ther, 2021, 6(1):362., DOI 10.1038/s41392-021-00670-9 Kochan G, Escors D, Breckpot K, Guerrero-Setas D. Role of non-classical MHC class I molecules in cancer immunosuppression. Oncoimmunology, 2013, 2(11):e26491., DOI 10.4161/onci.26491 Liu X, Song J, Zhang H, Liu X, Zuo F, Zhao Y, et al. Immune checkpoint HLA-E:CD94-NKG2A mediates evasion of circulating tumor cells from NK cell surveillance. Cancer Cell, 2023, 41(2):272–87.e9., DOI 10.1016/j.ccell. 2023.01.001 Ma R-Y, Black A, Qian B-Z. Macrophage diversity in cancer revisited in the era of single-cell omics. Trends Immunol, 2022, 43(7):546–63., DOI 10.1016/j.it.2022. 04.008 Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol, 2017, 17(9):559–72., DOI 10.1038/nri.2017.49 Potente M, Makinen T. Vascular heterogeneity and specialization in development and disease. Nat Rev Mol Cel Biol, 2017, 18(8):477–94., DOI 10. 1038/nrm.2017.36 Salazar N, Zabel BA. Support of tumor endothelial cells by chemokine receptors. Front Immunol, 2019, 10:147., DOI 10.3389/fimmu.2019.00147 Zhang L, Li Z, Skrzypczynska KM, FangQ, ZhangW, O’Brien SA, et al. Singlecell analyses inform mechanisms ofmyeloid-targeted therapies in colon cancer. Cell, 2020, 181(2):442–59.e29., DOI 10.1016/j.cell.2020.03.048 He Z, Chen Q, HeW, Cao J, Yao S, Huang Q, et al. Hepatocellular carcinoma subtypes based on metabolic pathways reveals potential therapeutic targets. Front Oncol, 2023, 13:1086604., DOI 10.3389/fonc.2023.1086604 Chiu C-F. Abstract 378: Ferritin regulates oxaliplatin sensitivity and immunosuppression in pancreatic ductal adenocarcinoma. Cancer Res, 2023, 83:378. 7_Supplement., DOI 10.1158/1538-7445.am2023-378 Li L, Yang L, Chen X, Chen X, Diao L, Zeng Y, et al. TNFAIP6 defines theMSC subpopulation with enhanced immune suppression activities. Stem Cel Res Ther, 2022, 13(1):479., DOI 10.1186/s13287-022-03176-5 Kuang Q, Liang Y, Zhuo Y, Cai Z, Jiang F, Xie J, et al. The ALDOAmetabolism pathway as a potential target for regulation of prostate cancer proliferation. Onco Targets Ther, 2021, 14:3353–66., DOI 10.2147/OTT.S290284 Zhang F, Lin J-D, Zuo X-Y, Zhuang Y-X,Hong C-Q, Zhang G-J, et al. Elevated transcriptional levels of aldolase A, ALDOA, associates with cell cycle-related genes in patients with NSCLC and several solid tumors. BioData Mining, 2017, 10(1):6., DOI 10.1186/s13040-016-0122-4 Yao X, He Z, Qin C, Deng X, Bai L, Li G, et al. SLC2A3 promotes macrophage infiltration by glycolysis reprogramming in gastric cancer. Cancer Cel Int, 2020, 20: 503., DOI 10.1186/s12935-020-01599-9 Xiao P, Gu J, Xu W, Niu X, Zhang J, Li J, et al. RTN4/Nogo-AS1PR2 negatively regulates angiogenesis and secondary neural repair through enhancing vascular autophagy in the thalamus after cerebral cortical infarction. Autophagy, 2022, 18(11):2711–30., DOI 10.1080/15548627.2022.2047344 Sai B, Dai Y, Fan S, Wang F, Wang L, Li Z, et al. Cancer-educated mesenchymal stem cells promote the survival of cancer cells at primary and distant metastatic sites via the expansion of bone marrow-derived-PMNMDSCs. Cell Death Dis, 2019, 10(12):941., DOI 10.1038/s41419-019-2149-1 Desbois M, Udyavar AR, Ryner L, Kozlowski C, Guan Y, Durrbaum M, et al. Integrated digital pathology and transcriptome analysis identifies molecular mediators of T-cell exclusion in ovarian cancer. Nat Commun, 2020, 11(1): 5583., DOI 10.1038/s41467-020-19408-2 Choi H, Lee RH, Bazhanov N, Oh JY, Prockop DJ. Anti-inflammatory protein TSG-6 secreted by activated MSCs attenuates zymosan-induced mouse peritonitis by decreasing TLR2/NF-κB signaling in residentmacrophages. Blood, 2011, 118(2): 330–8., DOI 10.1182/blood-2010-12-327353 Mittal M, Tiruppathi C, Nepal S, Zhao YY, Grzych D, Soni D, et al. TNFα- stimulated gene-6, TSG6, activates macrophage phenotype transition to prevent inflammatory lung injury. Proc Natl Acad Sci U S A, 2016, 113(50):E8151–e8158., DOI 10.1073/pnas.1614935113 Kuznetsova SA, Mahoney DJ, Martin-Manso G, Ali T, Nentwich HA, Sipes JM, et al. TSG-6 binds via its CUB_C domain to the cell-binding domain of fibronectin and increases fibronectin matrix assembly. Matrix Biol, 2008, 27(3): 201–10., DOI 10.1016/j.matbio.2007.10.003 Lawrance W, Banerji S, Day AJ, Bhattacharjee S, Jackson DG. Binding of hyaluronan to the native lymphatic vessel endothelial receptor LYVE-1 is critically dependent on receptor clustering and hyaluronan organization. J Biol Chem, 2016, 291(15):8014–30., DOI 10.1074/jbc.M115.708305 Azzaoui I, Uhel F, Rossille D, Pangault C, Dulong J, Le Priol J, et al. T-cell defect in diffuse large B-cell lymphomas involves expansion of myeloid-derived suppressor cells. Blood, 2016, 128(8):1081–92., DOI 10.1182/blood-2015-08-662783 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611210 EP 1611219 DI 10.3389/pore.2023.1611210 PG 10 ER PT J AU Chai, J Liu, X Hu, X Wang, Ch AF Chai, Jie Liu, Xiangli Hu, Xinju Wang, Chunfang TI Correlation analysis of circulating tumor cells and Claudin-4 in breast cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; circulating tumor cells; tight junction protein; claudin-4; molecular subtype ID breast cancer; circulating tumor cells; tight junction protein; claudin-4; molecular subtype AB Objective: We aimed to explore the relationship between peripheral blood circulating tumor cells (CTCs) and the expression of Claudin-4 in patients with breast cancer, and further explore the potential impact on clinical prognosis and risk assessment. Methods: We classified and enumerated circulating tumor cells in the blood of breast cancer patients by CTC-enriched in situ hybridization and the detection of Claudin-4 expression by immunohistochemistry. We carried out an analysis of the correlation between the two and the comparison of their impact on clinical parameters and prognosis. Results: There were 38 patients with a low expression of Claudin-4 and 27 patients with a high expression of Claudin-4. Compared with Claudin-4 low-expression patients, the number of CTCs was higher in patients with high Claudin-4 expression (11.7 vs. 7.4, p < 0.001). High Claudin-4 expression was associated with a lower count of epithelial CTCs (E-CTCs) (3.4 vs. 5.0, p = 0.033), higher counts of mesenchymal CTCs (M-CTC) (4.4 vs. 1.1, p < 0.001), and epithelial/mesenchymal CTCs (E/M-CTCs) (4.0 vs. 3.5, p = 0.021). The intensity of Claudin-4 was positively correlated with CTC (rs = 0.43, p = 0.001). Multivariate COX regression analysis showed that CTC counts (HR = 1.3, p < 0.001), Claudin-4 (HR = 4.6, p = 0.008), and Lymphatic metastasis (HR = 12.9, p = 0.001) were independent factors for poor prognosis. COX regression of CTC classification showed that epithelial/mesenchymal CTCs (E/M-CTC) (HR = 1.9, p = 0.001) and mesenchymal CTCs (M-CTC) (HR = 1.5, p = 0.001) were independent influencing factors of adverse reactions in breast cancer patients. Conclusion: The number of CTC in breast cancer is positively correlated with the expression of Claudin-4. High CTC counts and a high proportion of M-CTCs correlated with Claudin-4 expression. CTC counts and Claudin-4 expression were independent predictors of poor prognosis in breast cancer patients. C1 [Chai, Jie] The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Pathology DepartmentZhengzhou, China. [Liu, Xiangli] The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Pathology DepartmentZhengzhou, China. [Hu, Xinju] The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Pathology DepartmentZhengzhou, China. [Wang, Chunfang] The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Breast SurgeryZhengzhou, China. RP Wang, Ch (reprint author), The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Breast Surgery, Zhengzhou, China. EM asangna009@163.com CR Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. Ca: A Cancer J Clin, 2022, 72(1):7–33., DOI 10.3322/caac.21708 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Ca: A Cancer J Clin, 2021, 71(3): 209–49., DOI 10.3322/caac.21660 Harbeck N, Gnant M. Breast cancer. Lancet, 2017, 389(10074):1134–50., DOI 10.1016/s0140-6736(16)31891-8 Senkus E, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rutgers E, et al. Primary breast cancer: Esmo clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol, 2015, 26(5):v8–30., DOI 10.1093/annonc/mdv298 Cardoso F, Costa A, Senkus E, Aapro M, Andre F, Barrios CH, et al. 3rd ESOESMO international consensus guidelines for advanced breast cancer, ABC 3). Ann Oncol, 2017, 28(1):16–33., DOI 10.1093/annonc/mdw544 Nccn Clinical Practice Guidelines in Oncology, Nccn Guidelines). Breast cancer. Version 2.2022. Fort Washington: National Comprehensive Cancer Network, 2021). Nccn Clinical Practice Guidelines in Oncology, Nccn Guidelines). Breast cancer risk reduction. Version 1.2022. Fort Washington: National Comprehensive Cancer Network, 2022). Nccn Clinical Practice Guidelines in Oncology, Nccn Guidelines). Breast cancer screening and diagnosis. Version 1.2021. Fort Washington: National Comprehensive Cancer Network, 2021). Gotzsche PC, Jorgensen KJ. Screening for breast cancer with mammography. Cochrane Database Syst Rev, 2013, 2013(6):Cd001877., DOI 10.1002/14651858 Myers ER, Moorman P, Gierisch JM, Havrilesky LJ, Grimm LJ, Ghate S, et al. Benefits and harms of breast cancer screening: A systematic review. Jama, 2015, 314(15):1615–34., DOI 10.1001/jama.2015.13183 Nelson HD, Fu R, Cantor A, PappasM, Daeges M, Humphrey L. Effectiveness of breast cancer screening: Systematic review and meta-analysis to update the 2009 U.S. Preventive services task force recommendation. Ann Intern Med, 2016, 164(4):244–55., DOI 10.7326/m15-0969 Liu X, Tang JH, Wang MS. Expression of claudin-4 in human breast cancer tissue and clinical significance. J Jainan Med Coll, 2014, 20(12):1607–13., DOI 10. 13210/j.cnki.jhmu.20140926.005 Li R, Gong Z, Liu Y, Zhao X, Guo SJNF. Detection of circulating tumor cells and single cell extraction Technology: Principle, effect and application prospect. Futures, 2021, 5(3):032002., DOI 10.1088/2399-1984/ac1325 Wu F, Zhao J, Zhang X. Comparison of value of circulating tumor cells and tumor markers in lung cancer diagnosis. Int J Inspection Med, 2020, 41(5):527–31., DOI 10.3969/j.issn.1673-4130.2020.05.004 Thery L, Meddis A, Cabel L, Proudhon C, Latouche A, Pierga JY, et al. Circulating tumor cells in early breast cancer. JNCI Cancer Spectr, 2019, 3(2): pkz026., DOI 10.1093/jncics/pkz026 Zhang H, Lin X, Huang Y, Wang M, Cen C, Tang S, et al. Detection methods and clinical applications of circulating tumor cells in breast cancer. Front Oncol, 2021, 11:652253., DOI 10.3389/fonc.2021.652253 Luo L, Ma GR, Liu FG. Expression and clinical significance of tight junction proteins 1,3 and 4 in breast cancer patients. J Difficult Dis, 2017, 16(06):610., DOI 10. 3969/j.issn.1671-6450.2017.06.017 Lee KW, Lee NK, KimJH, Kang MS, Yoo HY, KimHH, et al. Twist1 causes the transcriptional repression of claudin-4 with prognostic significance in esophageal cancer. Biochem Biophys Res Commun, 2012, 423(3):454–60., DOI 10.1016/j.bbrc.2012.05.140 Xiang RL, Su YC, Pei XQ. Progress in functions of tight junction protein claudin-4. Sheng Li Ke Xue Jin Zhan [Progress Physiol], 2012, 43(4):310–4., DOI 10. 3969/j.issn.0559-7765 Abd-Elazeem MA, Abd-Elazeem MA. Claudin 4 expression in triple-negative breast cancer: Correlation with androgen receptors and ki-67 expression. Ann Diagn Pathol, 2015, 19(1):37–42., DOI 10.1016/j.anndiagpath.2014.10.003 Luo Y, Kishi S, Sasaki T, Ohmori H, Fujiwara-Tani R, Mori S, et al. Targeting claudin-4 enhances chemosensitivity in breast cancer. Cancer Sci, 2020, 111(5): 1840–50., DOI 10.1111/cas.14361 Duarte GM, Almeida NR, Tocchet F, Espinola J, Barreto CTR, Pinto GA, et al. Claudin-4 expression is associated with disease-free survival in breast carcinomain- situ: Mean follow-up of 8.2 years. Clin Breast Cancer, 2018, 18(5):e1111–6., DOI 10.1016/j.clbc.2018.06.005 Wang F, Gao Y, Tang L, Ning K, Geng N, Zhang H, et al. A novel pak4-cebpbcldn4 Axis involving in breast cancer cell migration and invasion. Biochem Biophys Res Commun, 2019, 511(2):404–8., DOI 10.1016/j.bbrc.2019.02.070 Singh AB, Dhawan P. Claudins and cancer: Fall of the soldiers entrusted to protect the gate and keep the barrier intact. Semin Cel Dev Biol, 2015, 42:58–65., DOI 10.1016/j.semcdb.2015.05.001 Tabaries S, Siegel PM. The role of claudins in cancer metastasis. Oncogene, 2017, 36:1176–90., DOI 10.1038/onc.2016.289 Webb PG, Spillman MA, Baumgartner HK. Claudins play a role in normal and tumor cell motility. BMC Cel Biol, 2013, 14:19., DOI 10.1186/1471-2121- 14-19 Escudero-Esparza A, Jiang WG, Martin TA. The claudin family and its role in cancer and metastasis. Front Biosci, Landmark Edition,, 2011, 16(3):1069–83., DOI 10.2741/3736 Fabisiewicz A, Szostakowska-Rodzos M, Zaczek AJ, Grzybowska EA. Circulating tumor cells in early and advanced breast cancer; biology and prognostic value. Int J Mol Sci, 2020, 21(5):1671., DOI 10.3390/ijms21051671 Kolokytha P, Yiannou P, Keramopoulos D, Kolokythas A, Nonni A, Patsouris E, et al. Claudin-3 and claudin-4: Distinct prognostic significance in triple-negative and luminal breast cancer. Appl Immunohistochem Mol Morphol, 2014, 22(2): 125–31., DOI 10.1097/PAI.0b013e31828d9d62 Radi DA, Abd-Elazeem MA. Prognostic significance of lymphatic vessel density detected by D2-40 and its relation to claudin-4 expression in prostatic adenocarcinoma. Int J Surg Pathol, 2016, 24(3):219–26., DOI 10.1177/ 1066896915611488 Ricardo S, Gerhard R, Cameselle-Teijeiro JF, Schmitt F, Paredes J. Claudin expression in breast cancer: High or low, what to expect? Histol Histopathol, 2012, 27(10):1283–95., DOI 10.14670/hh-27.1283 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611224 EP 1611232 DI 10.3389/pore.2023.1611224 PG 9 ER PT J AU Villanueva-Cotrina, F Velarde, J Rodriguez, R Bonilla, A Laura, M Saavedra, T Portillo-Alvarez, D Bustamante, Y Fernandez, C Galvez-Nino, M AF Villanueva-Cotrina, Freddy Velarde, Juan Rodriguez, Ricardo Bonilla, Alejandra Laura, Marco Saavedra, Tania Portillo-Alvarez, Diana Bustamante, Yovel Fernandez, Cesar Galvez-Nino, Marco TI Active cancer as the main predictor of mortality for COVID-19 in oncology patients in a specialized center SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE SARS-CoV-2; imaging study; biomarkers; death prognosis; active cancer ID SARS-CoV-2; imaging study; biomarkers; death prognosis; active cancer AB Introduction: The role of the type, stage and status of cancer in the outcome of COVID-19 remains unclear. Moreover, the characteristic pathological changes of severe COVID-19 reveled by laboratory and radiological findings are similar to those due to the development of cancer itself and antineoplastic therapies. Objective: To identify potential predictors of mortality of COVID-19 in cancer patients. Materials and methods: A retrospective and cross-sectional study was carried out in patients with clinical suspicion of COVID-19 who were confirmed for COVID-19 diagnosis by RT-PCR testing at the National Institute of Neoplastic Diseases between April and December 2020. Demographic, clinical, laboratory and radiological data were analyzed. Statistical analyses included area under the curve and univariate and multivariate logistic regression analyses. Results: A total of 226 patients had clinical suspicion ofCOVID-19, the diagnosiswas confirmed in 177 (78.3%), and 70/177 (39.5%) died. Age, active cancer, leukocyte count ≥12.8× 109/L, urea ≥7.4mmol/L, ferritin ≥1,640, lactate ≥2.0 mmol/L, and lung involvement ≥35% were found to be independent predictors of COVID-19mortality. Conclusion: Active cancer represents the main prognosis factor of death, while the role of cancer stage and type is unclear. Chest CT is a useful tool in the prognosis of death from COVID-19 in cancer patients. It is a challenge to establish the prognostic utility of laboratory markers as their altered values it could have either oncological or pandemic origins. C1 [Villanueva-Cotrina, Freddy] Instituto Nacional de Enfermedades Neoplasicas, Department of PathologyLima, Peru. [Velarde, Juan] Instituto Nacional de Enfermedades Neoplasicas, Department of Infectious DiseasesLima, Peru. [Rodriguez, Ricardo] Instituto Nacional de Enfermedades Neoplasicas, Department of PathologyLima, Peru. [Bonilla, Alejandra] Instituto Nacional de Enfermedades Neoplasicas, Department of RadiodiagnosisLima, Peru. [Laura, Marco] Instituto Nacional de Enfermedades Neoplasicas, Department of RadiodiagnosisLima, Peru. [Saavedra, Tania] Instituto Nacional de Enfermedades Neoplasicas, Department of Critical Care MedicineLima, Peru. [Portillo-Alvarez, Diana] Instituto Nacional de Enfermedades Neoplasicas, Department of Infectious DiseasesLima, Peru. [Bustamante, Yovel] Instituto Nacional de Enfermedades Neoplasicas, Department of PathologyLima, Peru. [Fernandez, Cesar] Instituto Nacional de Enfermedades Neoplasicas, Department of PathologyLima, Peru. [Galvez-Nino, Marco] Universidad Privada San Juan Bautista, Professional School of Human MedicineLima, Peru. RP Villanueva-Cotrina, F (reprint author), Instituto Nacional de Enfermedades Neoplasicas, Department of Pathology, Lima, Peru. EM freddyvillanue@gmail.com CR GuanWJ, Niyi Z, Hu Y, Liang WH, Ou CQ, He JX, et al. Clinical characteristics of coronavirus disease 2019 in China. New Engl J Med, 2020, 382(18):1708–20., DOI 10.1056/nejmoa2002032 Shi M, Chen L, Yang Y, Zhang J, Xu J, Xu G, et al. Analysis of clinical features and outcomes of 161 patients with severe and critical COVID-19: a multicenter descriptive study. J Clin Lab Anal, 2020, 34(9):e23415., DOI 10.1002/jcla.23415 Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA - J Am Med Assoc, 2020, 323(11):1061–9., DOI 10.1001/ jama.2020.1585 Yakovenko OK, Khanin OG, Lotysh VV, Gryf SL. Clinical features of severe COVID-19 with lethal outcome in volyn region residents. Ukrainian Pulmonology J, 2021, 29(2):16–24., DOI 10.31215/2306-4927-2021-29-2-16-24 Yang L, Chai P, Yu J, Fan X. Effects of cancer on patients with COVID-19: a systematic review and meta-analysis of 63,019 participants. Cancer Biol Med, 2021, 18:298–307., DOI 10.20892/j.issn.2095-3941.2020.0559 Bernard A, Cottenet J, Bonniaud P, Piroth L, Arveux P, Tubert-Bitter P, et al. Comparison of cancer patients to non-cancer patients among COVID-19 inpatients at a national level. Cancers, Basel,, 2021, 13(6):1436–15., DOI 10.3390/ cancers13061436 Rajeev Nadkarni A, Vijayakumaran SC, Gupta S, Divatia JV. Mortality in cancer patients with COVID-19 who are admitted to an ICU or who have severe COVID- 19: a systematic review and meta-analysis. JCO Glob Oncol, 2021, 7:1286–305., DOI 10.1200/go.21.00072 Wang L, Sun Y, Yuan Y, Mei Q, Yuan X. Clinical challenges in cancer patients with COVID-19: aging, immunosuppression, and comorbidities. Aging, 2020, 12: 24462–74., DOI 10.18632/aging.104205 Health Organization W. Guideline clinical management of COVID-19 patients: living guideline, 2021). Available from: http://apps.who.int/bookorders, Accessed November 23, 2021). Nader Marta G, Colombo Bonadio R, Nicole Encinas Sejas O, Watarai G, Mathias Machado MC, Teixeira Frasson L, et al. Outcomes and prognostic factors in a large cohort of hospitalized cancer patients with COVID-19. JCO Glob Oncol, 2021, 7:1084–92., DOI 10.1200/GO.21.00087 Mina A, Galvez C, Karmali R, Mulcahy M, Mi X, Kocherginsky M, et al. Outcomes of cancer patients with COVID-19 in a hospital system in the Chicago metropolitan area. Cancers, Basel,, 2022, 14(9):2209., DOI 10.3390/ cancers14092209 Benderra MA, Aparicio A, Leblanc J, Wassermann D, Kempf E, Galula G, et al. Clinical characteristics, care trajectories and mortality rate of SARS-CoV-2 infected cancer patients: a multicenter cohort study. Cancers, Basel)., 2021, 13(19):4749., DOI 10.3390/cancers13194749 Katal S, Aghaghazvini L, Gholamrezanezhad A. Chest-CT findings of COVID-19 in patients with pre-existing malignancies; a pictorial review. Clin Imaging, 2020, 67:121–9., DOI 10.1016/j.clinimag.2020.06.004 Bourdoncle S, Eche T, McGale J, Yiu K, Partouche E, Yeh R, et al. Investigating of the role of CT scan for cancer patients during the first wave of COVID-19 pandemic. Res Diagn Interv Imaging, 2022, 1:100004., DOI 10.1016/j.redii.2022. 100004 Levi M. Disseminated intravascular coagulation in cancer patients. Best Pract Res Clin Haematol, 2009, 22:129–36., DOI 10.1016/j.beha.2008.12.005 Humphreys BD, Soiffer RJ, Magee CC. Renal failure associated with cancer and its treatment: an update. J Am Soc Nephrol, 2005, 16:151–61., DOI 10.1681/ASN. 2004100843 Levi M. Disseminated intravascular coagulation in cancer: an update. Semin Thromb Hemost, 2019, 45(4):342–7., DOI 10.1055/s-0039-1687890 Lameire NH, Flombaum CD, Moreau D, Ronco C. Acute renal failure in cancer patients. Ann Med, 2005, 37:13–25., DOI 10.1080/07853890510007205 Kavanaugh DY, Carbone DP. Immunologic dysfunction in cancer. Oncology, Williston Park,, 1996, 10:927–51., DOI 10.1016/s0889-8588(05)70376-2 Cedervall J, Zhang Y, Olsson AK. Tumor-induced NETosis as a risk factor for metastasis and organ failure. Cancer Res, 2016, 76:4311–5., DOI 10.1158/0008-5472. CAN-15-3051 Nava S, Cuomo AM. Acute respiratory failure in the cancer patient: the role of non-invasive mechanical ventilation. Crit Rev Oncology/Hematology, 2004, 51: 91–103., DOI 10.1016/j.critrevonc.2004.04.004 Vial T, Descotes J. Immunosuppressive drugs and cancer. Toxicology, 2003, 185(3):229–40., DOI 10.1016/s0300-483x(02)00612-1 Ngo D, Jia JB, Green CS, Gulati AT, Lall C. Cancer therapy related complications in the liver, pancreas, and biliary system: an imaging perspective. Insight Imaging, 2015, 6:665–77., DOI 10.1007/s13244-015-0436-7 Pan F, Ye T, Sun P, Gui S, Liang B, Li L, et al. Time course of lung changes at chest CT during recovery from Coronavirus disease 2019, COVID-19). Radiology, 2020, 295(3):715–21., DOI 10.1148/radiol.2020200370 Prokop M, van Everdingen W, van Rees Vellinga T, van Ufford HQ, Stoger L, Beenen L, et al. CO-RADS: a categorical CT assessment scheme for patients suspected of having COVID-19-definition and evaluation. Radiology, 2020, 296(2):E97–104., DOI 10.1148/radiol.2020201473 World Health Organization. Clinical management of severe acute respiratory infection when novel coronavirus, nCoV, infection is suspected: interim guidance 2 1. In: Triage: early recognition of patients with SARI associated with nCoV infection, 2020). Available from: https:// apps.who.int/iris/bitstream/handle/10665/331446/WHO-2019-nCoVclinical- 2020.4-eng.pdf?sequence=1&isAllowed=y, Accessed March 13, 2020). Ferrari BL, Carlos, Ferreira G, Menezes M, de Marchi P, Canedo J, et al. Determinants of COVID-19 mortality in patients with cancer from a community oncology practice in Brazil. JCO Glob Oncol, 2021, 7:46–55., DOI 10.1200/GO.20. 00444 Meza Riquelme MJS, Condori Pereyra AR, Encalada Carbajal DA. Analisis de politicas publicas en el Peru ante la crisis derivada de la COVID-19. Semestre Economico, 2020, 23(55):113–38., DOI 10.22395/seec.v23n55a5 Zorzi M, Guzzinati S, Avossa F, Fedeli U, Calcinotto A, Rugge M. SARS-CoV- 2 infection in cancer patients: a population-based study. Front Oncol, 2021, 11: 730131., DOI 10.3389/fonc.2021.730131 Kuderer NM, Choueiri TK, Shah DP, Shyr Y, Rubinstein SM, Rivera DR, et al. Clinical impact of COVID-19 on patients with cancer, CCC19): a cohort study. The Lancet, 2020, 395(10241):1907–18., DOI 10.1016/S0140-6736(20)31187-9 Borno HT, Kim MO, Hong JC, Yousefi S, Lin A, Tolstykh I, et al. COVID-19 outcomes among patients with cancer: observations from the university of California cancer consortium COVID-19 project outcomes registry. Oncologist, 2022, 27(5):398–406., DOI 10.1093/oncolo/oyac038 Pinato DJ, Scotti L, Gennari A, Colomba-Blameble E, Dolly S, Loizidou A, et al. Determinants of enhanced vulnerability to coronavirus disease 2019 in UK patients with cancer: a European study. Eur J Cancer, 2021, 150:190–202., DOI 10. 1016/j.ejca.2021.03.035 Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science, 2011, 331(6024):1565–70., DOI 10.1126/science.1203486 Khorana AA, Mackman N, Falanga A, Pabinger I, Noble S, Ageno W, et al. Cancer-associated venous thromboembolism. Nat Rev Dis Primers, 2022, 8(1): 11–8., DOI 10.1038/s41572-022-00336-y Connors JM, Levy JH. Thromboinflammation and the hypercoagulability of COVID-19. J Thromb Haemost, 2020, 18:1559–61., DOI 10.1111/jth.14849 Mangalmurti N, Hunter CA. Cytokine storms understanding COVID-19. Immunity, 2020, 53(1):19–25., DOI 10.1016/j.immuni.2020.06.017 Owusuaa C, Dijkland SA, Nieboer D, van der Heide A, van der Rijt CCD. Predictors of mortality in patients with advanced cancer—a systematic review and meta-analysis. Cancers, 2022, 14:328., DOI 10.3390/cancers14020328 Ullgren H, Camuto A, Rosas S, Pahnke S, Ginman B, Enblad G, et al. Clinical characteristics and factors associated with COVID-19-related death and morbidity among hospitalized patients with cancer: a Swedish cohort study. Acta Oncologica, 2021, 60(11):1459–65., DOI 10.1080/0284186X.2021.1958005 Liu Y, Lu H, Wang W, Liu Q, Zhu C. Clinical risk factors for mortality in patients with cancer and COVID-19: a systematic review and meta-analysis of recent observational studies. Expert Rev Anticancer Ther, 2021, 21(1):107–19., DOI 10.1080/14737140.2021.1837628 Fattore GL, Olivos NSA, Olalla JEC, Gomez L, Marucco AF, Mena MPR. Mortality in patients with cancer and SARS-CoV-2 infection: results from the argentinean network of hospital-based cancer registries. Cancer Epidemiol, 2022, 79:102200., DOI 10.1016/j.canep.2022.102200 Jergovic M, Coplen CP, Uhrlaub JL, Nikolich-Zugich J. Immune response to COVID-19 in older adults. J Heart Lung Transplant, 2021, 40(10):1082–9., DOI 10. 1016/j.healun.2021.04.017 Kernan KF, Carcillo JA. Hyperferritinemia and inflammation. Int Immunol, 2017, 29(9):401–9., DOI 10.1093/intimm/dxx031 Matzner Y, Konijn AM, Hershko C. Serum ferritin in hematologic malignancies. Am J Hematol, 1980, 9:13–22., DOI 10.1002/ajh.2830090103 Aulbert E, Steffens O. Ferritin im serum-ein «tumormarker» bei malignen lymphomen? Oncology Research and Treatment, 1990, 13(2):102–8., DOI 10.1159/ 000216735 Qeadan F, Tingey B, Gu LY, Packard AH, Erdei E, Saeed AI. Prognostic values of serum ferritin and d-dimer trajectory in patients with covid-19. Viruses, 2021, 13(3):419., DOI 10.3390/v13030419 Depalma RG, Hayes VW, O’Leary TJ. Optimal serum ferritin level range: iron status measure and inflammatory biomarker. Metallomics, 2021, 13(6):mfab030., DOI 10.1093/mtomcs/mfab030 Bakker J, Gris P, Coffernils M, Kahn RJ, Vincent JL. Serial blood lactate levels can predict the development of multiple organ failure following septic shock. Am J Surg, 1996, 171:221–6., DOI 10.1016/S0002-9610(97)89552-9 Bakker J, Vincent JL. The oxygen supply dependency phenomenon is associated with increased blood lactate levels. J Crit Care, 1991, 6:152–9., DOI 10. 1016/0883-9441(91)90006-f Nazer LH, Rimawi D, Hawari FI. Evaluating the predictive value of lactate in patients with cancer having septic shock. J Intensive CareMed, 2020, 35(8):789–96., DOI 10.1177/0885066618788821 Bagshaw SM, Gibney RTN. Conventional markers of kidney function. Crit Care Med, 2008, 36:S152–8., DOI 10.1097/CCM.0b013e318168c613 Stevens LA, Levey AS. Measurement of kidney function. Med Clin North America, 2005, 89:457–73., DOI 10.1016/j.mcna.2004.11.009 Peleg Y, Kudose S, D’Agati V, Siddall E, Ahmad S, Nickolas T, et al. Acute kidney injury due to collapsing glomerulopathy following COVID-19 infection. Kidney Int Rep, 2020, 5(6):940–5., DOI 10.1016/j.ekir.2020.04.017 Chueh TI, Zheng CM, Hou YC, Lu KC. Novel evidence of acute kidney injury in COVID-19. J Clin Med, 2020, 9(11):3547., DOI 10.3390/jcm9113547 Nath SS, Yadav NU, Derkach A, Perez-Johnston R, Tachiki L, Maguire K, et al. Outcomes of patients with COVID-19 from a specialized cancer care emergency room. Cancer Invest, 2022, 40(1):17–25., DOI 10.1080/07357907.2021. 1985134 Williams Donaldson V. A clinical study of visualization on depressed white blood cell count in medical patient. Appl Psychophysiology Biofeedback, 2000, 25(2): 117–28., DOI 10.1023/a:1009518925859 Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol, 2013, 13:159–75., DOI 10.1038/nri3399 Balkwill F, Charles KA, Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell, 2005, 7:211–7., DOI 10.1016/j.ccr.2005.02.013 Coussens LM, Werb Z. Inflammation and cancer. Nature, 2002, 420(6917): 860–867.doi:10.1038/nature01322 Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature, 2008, 454:436–44., DOI 10.1038/nature07205 Shitara K, Matsuo K, Oze I, Mizota A, Kondo C, Nomura M, et al. Metaanalysis of neutropenia or leukopenia as a prognostic factor in patients with malignant disease undergoing chemotherapy. Cancer Chemother Pharmacol, 2011, 68(2):301–7., DOI 10.1007/s00280-010-1487-6 Lowenthal RM, Eaton K. Toxicity of chemotherapy. Hematology/Oncology Clinics, 1996, 10(4):967–90., DOI 10.1016/s0889-8588(05)70378-6 Kvolik S, Jukic M, Matijevic M, Marjanovic K, Glavas-Obrovac L. An overview of coagulation disorders in cancer patients. Surg Oncol, 2010, 19:e33–46., DOI 10. 1016/j.suronc.2009.03.008 de Cicco M. The prothrombotic state in cancer: pathogenic mechanisms. Crit Rev Oncology/Hematology, 2004, 50:187–96., DOI 10.1016/j.critrevonc.2003. 10.003 Francone M, Iafrate F, Masci GM, Coco S, Cilia F, Manganaro L, et al. Chest CT score in COVID-19 patients: correlation with disease severity and short-term prognosis. Eur Radiol, 2020, 30(12):6808–17., DOI 10.1007/s00330-020-07033-y Angeli E, Dalto S, Marchese S, Setti L, Bonacina M, Galli F, et al. Prognostic value of CT integrated with clinical and laboratory data during the first peak of the COVID-19 pandemic in northern Italy: a nomogram to predict unfavorable outcome. Eur J Radiol, 2021, 137:109612., DOI 10.1016/j. ejrad.2021.109612 Bayrak V, Senturk Durukan N, Demirer Aydemir F, Ergan B, Gezer NS, Eren Kutsoylu OO, et al. Risk factors associated with mortality in intensive care COVID- 19 patients: the importance of chest CT score and intubation timing as risk factors. Turk J Med Sci, 2021, 51(4):1665–74., DOI 10.3906/sag-2101-89 Li K, Li K, Chen D, Chen D, Chen S, Chen S, et al. Predictors of fatality including radiographic findings in adults with COVID-19. Respir Res, 2020, 21(1): 146., DOI 10.1186/s12931-020-01411-2 Hajiahmadi S, Shayganfar A, Janghorbani M, Esfahani MM, Mahnam M, Bakhtiarvand N, et al. Chest computed tomography severity score to predict adverse outcomes of patients with COVID-19. Infect Chemother, 2021, 53:308–18., DOI 10. 3947/ic.2021.0024 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611236 EP 1611247 DI 10.3389/pore.2023.1611236 PG 12 ER PT J AU Cazzaniga, M Zonzini, BG Pierro, DF Palazzi, MCh Cardinali, M Bertuccioli, A AF Cazzaniga, Massimiliano Zonzini, Bruno Giordano Pierro, Di Francesco Palazzi, Maria Chiara Cardinali, Marco Bertuccioli, Alexander TI Influence of the microbiota on the effectiveness and toxicity of oncological therapies, with a focus on chemotherapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE microbiota; chemotherapy; oncology; toxicity; microbiome ID microbiota; chemotherapy; oncology; toxicity; microbiome AB Recent studies have highlighted a possible correlation between microbiota composition and the pathogenesis of various oncological diseases. Also, many bacterial groups are now directly or indirectly associated with the capability of stimulating or inhibiting carcinogenic pathways. However, little is known about the importance and impact of microbiota patterns related to the efficacy and toxicity of cancer treatments. We have recently begun to understand how oncological therapies and the microbiota are closely interconnected and could influence each other. Chemotherapy effectiveness, for example, appears to be strongly influenced by the presence of some microorganisms capable of modulating the pharmacokinetics and pharmacodynamics of the compounds used, thus varying the real response and therefore the efficacy of the oncological treatment. Similarly, chemotherapeutic agents can modulate the microbiota with variations that could facilitate or avoid the onset of important side effects. This finding has or could have considerable relevance as it is possible that our ability to modulate and modify the microbial structure before, during, and after treatment could influence all the clinical parameters related to pharmacological treatments and, eventually, the prognosis of the disease. C1 [Cazzaniga, Massimiliano] Velleja Research, Scientific and Research DepartmentMilano, Italy. [Zonzini, Bruno Giordano] University of Urbino Carlo Bo, Department of Biomolecular SciencesUrbino, Italy. [Pierro, Di Francesco] Velleja Research, Scientific and Research DepartmentMilano, Italy. [Palazzi, Maria Chiara] Associazione Italiana Fitness e MedicinaRavenna, Italy. [Cardinali, Marco] Azienda Unita Sanitaria Locale Romagna, Infermi Hospital, Department of Internal MedicineRimini, Italy. [Bertuccioli, Alexander] University of Urbino Carlo Bo, Department of Biomolecular SciencesUrbino, Italy. RP Bertuccioli, A (reprint author), University of Urbino Carlo Bo, Department of Biomolecular Sciences, Urbino, Italy. EM alexander.bertuccioli@uniurb.it CR El-Sayed A, Aleya L, Kamel M. Microbiota’s role in health and diseases. Environ Sci Pollut Res Int, 2021, 28(28):36967–83., DOI 10.1007/s11356-021-14593-z Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Nejman Sci, 2020, 368(6494):973–80., DOI 10.1126/science.aay9189 Audirac-Chalifour A, Torres-Poveda K, Bahena-Roman M, Tellez-Sosa J, Martinez-Barnetche J, Cortina-Ceballos B, et al. Cervical microbiome and cytokine profile at various stages of cervical cancer: A pilot study. PLoS One, 2016, 11(4):e0153274., DOI 10.1371/journal.pone.0153274 Gethings-Behncke C, Coleman HG, Jordao HWT, Longley DB, Crawford N, Murray LJ, et al. Fusobacterium nucleatum in the colorectum and its association with cancer risk and survival: A systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev, 2020, 29(3):539–48., DOI 10.1158/1055-9965.EPI-18-1295 Roy S, Trinchieri G. Microbiota: A key orchestrator of cancer therapy. Nat Rev Cancer, 2017, 17(5):271–85., DOI 10.1038/nrc.2017.13 Sevcikova A, Izoldova N, Stevurkova V, Kasperova B, Chovanec M, Ciernikova S, et al. The impact of the microbiome on resistance to cancer treatment with chemotherapeutic agents and immunotherapy. Int J Mol Sci, 2022, 23:488., DOI 10. 3390/ijms23010488 Sharom FJ. ABC multidrug transporters: Structure, function and role in chemoresistance. Pharmacogenomics, 2008, 9:105–27., DOI 10.2217/14622416.9.1.105 Li LY, Guan YD, Chen XS, Yang JM, Cheng Y. DNA repair pathways in cancer therapy and resistance. Front Pharmacol, 2020, 11:629266., DOI 10.3389/fphar.2020. 629266 Lehouritis P, Cummins J, Stanton M, Murphy CT, McCarthy FO, Reid G, et al. Local bacteria affect the efficacy of chemotherapeutic drugs. Sci Rep, 2015, 5:14554., DOI 10.1038/srep14554 Mini E, Nobili S, Caciagli B, Landini I, Mazzei T. Cellular pharmacology of gemcitabine. Ann Oncol, 2006, 17:v7–12., DOI 10.1093/annonc/mdj941 Geller LT, Straussman R. Intratumoral bacteria may elicit chemoresistance by metabolizing anticancer agents. Mol Cell Oncol., 2017, 5(1):e1405139., DOI 10.1080/ 23723556.2017.1405139 Geller LT, Barzily-Rokni M, Danino T, Jonas OH, Shental N, Nejman D, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science, 2017, 357(6356):1156–60., DOI 10. 1126/science.aah5043 Shang FM, Liu HL. Fusobacterium nucleatum and colorectal cancer: A review. World J Gastrointest Oncol, 2018, 10(3):71–81., DOI 10.4251/wjgo.v10.i3.71 Li YJ, Lei YH, Yao N, Wang CR, Hu N, Ye WC, et al. Autophagy and multidrug resistance in cancer. Chin J Cancer, 2017, 36(1):52., DOI 10.1186/s40880- 017-0219-2 TaChung Y, Guo F, Yu Y, Sun T,Ma D, Han J, et al. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell, 2017, 170:548–63., DOI 10.1016/j.cell.2017.07.008 Emadi A, Jones RJ, Brodsky RA. Cyclophosphamide and cancer: Golden anniversary. Nat Rev Clin Oncol, 2009, 6:638–47., DOI 10.1038/nrclinonc.2009.146 Ahlmann M, Hempel G. The effect of cyclophosphamide on the immune system: Implications for clinical cancer therapy. Cancer Chemother Pharmacol, 2016, 78:661–71., DOI 10.1007/s00280-016-3152-1 Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere R, Hannani D, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science, 2013, 342(6161):971–6., DOI 10.1126/science.1240537 Lida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N,Weingarten RA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science, 2013, 342(6161):967–70., DOI 10.1126/science.1240527 Bailly C. Irinotecan: 25 years of cancer treatment. Pharmacol Res, 2019, 148: 104398., DOI 10.1016/j.phrs.2019.104398 Kciuk M, Marciniak B, Kontek R. Irinotecan-still an important player in cancer chemotherapy: A comprehensive overview. Int J Mol Sci, 2020, 21(14):4919., DOI 10.3390/ijms21144919 Yue B, Gao R, Wang Z, Dou W. Microbiota-host-irinotecan Axis: A new insight toward irinotecan chemotherapy. Front Cell Infect Microbiol, 2021, 11: 710945., DOI 10.3389/fcimb.2021.710945 Jasiewicz F, Qurban Z, Hughes C. Treatment-induced mucositis in oncology. Br J Hosp Med, Lond,, 2022, 83(9):1–8., DOI 10.12968/hmed.2022.0324 Keefe DM, Gibson RJ, Hauer-Jensen M. Gastrointestinal mucositis. Semin Oncol Nurs, 2004, 20:38–47., DOI 10.1053/j.soncn.2003.10.007 Sonis ST. The pathobiology of mucositis. Nat Rev Cancer, 2004, 4(4):277–84., DOI 10.1038/nrc1318 Van Vliet MJ, Harmsen HJM, de Bont ESJM, Tissing WJE. The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. Plos Pathog, 2010, 6(5):e1000879., DOI 10.1371/journal.ppat.1000879 Holscher HD. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes, 2017, 8(2):172–84., DOI 10.1080/19490976.2017.1290756 Stoeva MK, Garcia-So J, Justice N, Myers J, Tyagi S, Nemchek M, et al. Butyrate-producing human gut symbiont, Clostridium butyricum, and its role in health and disease. Gut Microbes, 2021, 13(1):1–28., DOI 10.1080/19490976.2021. 1907272 Ariyoshi T, Hagihara M, Tomono S, Eguchi S, Minemura A, Miura D, et al. Clostridium butyricum MIYAIRI 588 modifies bacterial composition under antibiotic-induced dysbiosis for the activation of interactions via lipid metabolism between the gut microbiome and the host. Biomedicines, 2021, 9(8):1065., DOI 10.3390/biomedicines9081065 Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F. From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell, 2016, 165(6):1332–45., DOI 10.1016/j.cell.2016.05.041 Liu P, Wang Y, Yang G, Zhang Q, Meng L, Xin Y, et al. The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacol Res, 2021, 165:105420., DOI 10.1016/j.phrs.2021.105420 Mirzaei R, Afaghi A, Babakhani S, Sohrabi MR, Hosseini-Fard SR, Babolhavaeji K, et al. Role of microbiota-derived short-chain fatty acids in cancer development and prevention. Biomed Pharmacother, 2021, 139:111619., DOI 10.1016/j.biopha.2021.111619 Al-Qadami GH, Secombe KR, Subramaniam CB, Wardill HR, Bowen JM. Gut microbiota-derived short-chain fatty acids: Impact on cancer treatment response and toxicities. Microorganisms, 2022, 10(10):2048., DOI 10.3390/microorganisms10102048 Danne C, Sokol H. Butyrate, a new microbiota-dependent player in CD8+ T cells immunity and cancer therapy? Cell Rep Med, 2021, 2(7):100328., DOI 10. 1016/j.xcrm.2021.100328 Russo M, Guida F, Paparo L, Trinchese G, Aitoro R, Avagliano C, et al. The novel butyrate derivative phenylalanine-butyramide protects from doxorubicininduced cardiotoxicity. Eur J Heart Fail, 2019, 21(4):519–28., DOI 10.1002/ejhf.1439 Cristiano C, Cuozzo M, Coretti L, Liguori FM, Cimmino F, Turco L, et al. Oral sodium butyrate supplementation ameliorates paclitaxel-induced behavioral and intestinal dysfunction. Biomed Pharmacother, 2022, 153:113528., DOI 10.1016/j. biopha.2022.113528 Cong J, Zhang C, Zhou S, Zhu J, Liang C. A pilot study: Favorable effects of clostridium butyricum on intestinal microbiota for adjuvant therapy of lung cancer. Cancers, Basel,, 2022, 14(15):3599., DOI 10.3390/cancers14153599 Tomita Y, Ikeda T, Sakata S, Saruwatari K, Sato R, Iyama S, et al. Association of probiotic clostridium butyricum therapy with survival and response to immune checkpoint blockade in patients with lung cancer. Cancer Immunol Res, 2020, 8(10):1236–42., DOI 10.1158/2326-6066.CIR-20-0051 Geng HW, Yin FY, Zhang ZF, Gong X, Yang Y. Butyrate suppresses glucose metabolism of colorectal cancer cells via GPR109a-AKT signaling pathway and enhances chemotherapy. Front Mol Biosci, 2021, 8:634874., DOI 10.3389/fmolb. 2021.634874 Pascale RM, Calvisi DF, Simile MM, Feo CF, Feo F. The Warburg effect 97 Years after its discovery. Cancers, Basel,, 2020, 12(10):2819., DOI 10.3390/ cancers12102819 Song D, Hao J, Fan D. Biological properties and clinical applications of berberine. Front Med, 2020, 14(5):564–82., DOI 10.1007/s11684-019-0724-6 Patel SS, Acharya A, Ray RS, Agrawal R, Raghuwanshi R, Jain P. Cellular and molecular mechanisms of curcumin in prevention and treatment of disease. Crit Rev Food Sci Nutr, 2020, 60(6):887–939., DOI 10.1080/10408398.2018.1552244 Yue B, Gao R, Lv C, Yu Z, Wang H, Geng X, et al. Berberine improves irinotecan-induced intestinal mucositis without impairing the anti-colorectal cancer efficacy of irinotecan by inhibiting bacterial β-glucuronidase. Front Pharmacol, 2021, 12:774560., DOI 10.3389/fphar.2021.774560 Mohammadinejad R, Ahmadi Z, Tavakol S, Ashrafizadeh M. Berberine as a potential autophagy modulator. J Cell Physiol, 2019, 234(9):14914–26., DOI 10.1002/ jcp.28325 Belcaro G, Hosoi M, Pellegrini L, Appendino G, Ippolito E, Ricci A, et al. A controlled study of a lecithinized delivery system of curcumin, Meriva®, to alleviate the adverse effects of cancer treatment. Phytother Res, 2014, 28(3):444–50., DOI 10. 1002/ptr.5014 Shakeri A, Cicero AFG, Panahi Y, Mohajeri M, Sahebkar A. Curcumin: A naturally occurring autophagy modulator. J Cell Physiol, 2019, 234(5):5643–54., DOI 10.1002/jcp.27404 Waghray D, Zhang Q. Inhibit or evade multidrug resistance P-glycoprotein in cancer treatment. J Med Chem, 2018, 61(12):5108–21., DOI 10.1021/acs.jmedchem. 7b01457 Lopes-Rodrigues V, Sousa E, Vasconcelos MH. Curcumin as a modulator of P-glycoprotein in cancer: Challenges and perspectives. Pharmaceuticals, Basel,, 2016, 9(4):71., DOI 10.3390/ph9040071 Guo S, Lv L, Shen Y, Hu Z, He Q, Chen X. A nanoparticulate prechemosensitizer for efficacious chemotherapy of multidrug resistant breast cancer. Sci Rep, 2016, 6:21459., DOI 10.1038/srep21459 Sreekanth CN, Bava SV, Sreekumar E, Anto RJ. Molecular evidences for the chemosensitizing efficacy of liposomal curcumin in paclitaxel chemotherapy in mouse models of cervical cancer. Oncogene, 2011, 30(28):3139–52., DOI 10.1038/ onc.2011.23 Yallapu MM, Maher DM, Sundram V, Bell MC, Jaggi M, Chauhan SC. Curcumin induces chemo/radio-sensitization in ovarian cancer cells and curcumin nanoparticles inhibit ovarian cancer cell growth. J Ovarian Res, 2010, 3:11., DOI 10. 1186/1757-2215-3-11 JiangM,Huang O, Zhang X, Xie Z, Shen A, Liu H, et al. Curcumin induces cell death and restores tamoxifen sensitivity in the antiestrogen-resistant breast cancer cell lines MCF-7/LCC2 and MCF-7/LCC9. Molecules, 2013, 18(1):701–20., DOI 10. 3390/molecules18010701 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611300 EP 1611308 DI 10.3389/pore.2023.1611300 PG 9 ER PT J AU Gyulai, M Harko, T Fabian, K Karsko, L Agocs, L Szigeti, B Fillinger, J Szallasi, Z Pipek, O Moldvay, J AF Gyulai, Marton Harko, Tunde Fabian, Katalin Karsko, Luca Agocs, Laszlo Szigeti, Balazs Fillinger, Janos Szallasi, Zoltan Pipek, Orsolya Moldvay, Judit TI Claudin expression in pulmonary adenoid cystic carcinoma and mucoepidermoid carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE adenoid cystic carcinoma; mucoepidermoid carcinoma; rare lung tumors; claudin expression; immunohistochemistry ID adenoid cystic carcinoma; mucoepidermoid carcinoma; rare lung tumors; claudin expression; immunohistochemistry AB Background: Although the expression of tight junction protein claudins (CLDNs) is well known in common histological subtypes of lung cancer, it has not been investigated in rare lung cancers. The aim of our study was to examine the expression of different CLDNs in pulmonary salivary gland tumors. Methods: 35 rare lung cancers including pathologically confirmed 12 adenoid cystic carcinomas (ACCs) and 23 mucoepidermoid carcinomas (MECs) were collected retrospectively. Immunohistochemical (IHC) staining was performed on formalin fixed paraffin embedded (FFPE) tumor tissues, and CLDN1, -2, -3, -4, -5, -7, and -18 protein expressions were analyzed. The levels of immunopositivity were determined with H-score. Certain pathological characteristics of ACC and MEC samples (tumor grade, presence of necrosis, presence of blood vessel infiltration, and degree of lymphoid infiltration) were also analyzed. Results: CLDN overexpression was observed in both tumor types, especially in CLDN2, -7, and -18 IHC. Markedly different patterns of CLDN expression were found for ACC and MEC tumors, especially for CLDN1, -2, -4, and -7, although none of these trends remained significant after correction for multiple testing. Positive correlations between expressions of CLDN2 and -5, CLDN3 and -4, and CLDN5 and -18 were also demonstrated. Tumors of never-smokers presented lower levels of CLDN18 than tumors of current smokers (p-value: 0.003). Conclusion: This is the first study to comprehensively describe the expression of different CLDNs in lung ACC and MEC. Overexpression of certain CLDNs may pave the way for targeted anti-claudin therapy in these rare histological subtypes of lung cancer. C1 [Gyulai, Marton] County Hospital of PulmonologyTorokbalint, Hungary. [Harko, Tunde] Orszagos Koranyi Pulmonologiai Intezet, Patologiai OsztalyBudapest, Hungary. [Fabian, Katalin] Municipal Hospital, Del-Pest, Department of PathologyBudapest, Hungary. [Karsko, Luca] Orszagos Koranyi Tbc es Pulmonologiai Intezet, MellkassebeszetBudapest, Hungary. [Agocs, Laszlo] Orszagos Koranyi Tbc es Pulmonologiai Intezet, MellkassebeszetBudapest, Hungary. [Szigeti, Balazs] Orszagos Koranyi Pulmonologiai Intezet, Patologiai OsztalyBudapest, Hungary. [Fillinger, Janos] Orszagos Koranyi Pulmonologiai Intezet, Patologiai OsztalyBudapest, Hungary. [Szallasi, Zoltan] National Koranyi Institute of PulmonologyBudapest, Hungary. [Pipek, Orsolya] Eotvos Lorand University, Department of Physics of Complex SystemsBudapest, Hungary. [Moldvay, Judit] National Koranyi Institute of Pulmonology, Semmelweis University, Ist Department of PulmonologyBudapest, Hungary. RP Moldvay, J (reprint author), National Koranyi Institute of Pulmonology, Semmelweis University, Ist Department of Pulmonology, Budapest, Hungary. EM drmoldvay@hotmail.com CR Pisano C, Witel G, De Filippis M, Listi A, Napoli F, Righi L, et al. Moving through rare lung cancer histologies: A narrative review on diagnosis and treatment of selected infrequent entities. Precision Cancer Med, 2022, 5:27., DOI 10.21037/ pcm-22-4 Qin BD, Jiao XD, Liu K, Wu Y, He X, Liu J, et al. Clinical, pathological and treatment factors associated with the survival of patients with primary pulmonary salivary gland-type tumors. Lung Cancer, Amsterdam, Netherlands,, 2018, 126: 174–81., DOI 10.1016/j.lungcan.2018.11.010 Lee RH, Wai KC, Chan JW, Ha PK, Kang H. Approaches to themanagement of metastatic adenoid cystic carcinoma. Cancers, 2022, 14(22):5698., DOI 10.3390/ cancers14225698 Wang DW, Zhang WH, Danil G, Yang K, Hu JK. The role and mechanism of claudins in cancer. Front Oncol, 2022, 12:1051497., DOI 10.3389/fonc.2022.1051497 Mineta K, Yamamoto Y, Yamazaki Y, Tanaka H, Tada Y, Saito K, et al. Predicted expansion of the claudin multigene family. FEBS Lett, 2011, 585(4): 606–12., DOI 10.1016/j.febslet.2011.01.028 Osanai M, Takasawa A, Murata M, Sawada N. Claudins in cancer: Bench to bedside. Pflugers Archiv Eur J Physiol, 2017, 469(1):55–67., DOI 10.1007/s00424- 016-1877-7 Popova OP, Kuznetsova AV, Bogomazova SY, Ivanov AA. Claudins as biomarkers of differential diagnosis and prognosis of tumors. J Cancer Res Clin Oncol, 2021, 147(10):2803–17., DOI 10.1007/s00432-021-03725-0 Moldvay J, Fabian K, Jackel M, Nemeth Z, Bogos K, Furak J, et al. Claudin-1 protein expression is a good prognostic factor in non-small cell lung cancer, but only in squamous cell carcinoma cases. Pathol Oncol Res, 2017, 23(1):151–6., DOI 10. 1007/s12253-016-0115-0 Moldvay J, Jackel M, Paska C, Soltesz I, Schaff Z, Kiss A. Distinct claudin expression profile in histologic subtypes of lung cancer. Lung Cancer, 2007, 57(2):159–67. Amsterdam, Netherlands., DOI 10.1016/j. lungcan.2007.02.018 Wang YB, Shi Q, Li G, Zheng JH, Lin J, Qiu W. MicroRNA-488 inhibits progression of colorectal cancer via inhibition of the mitogen-activated protein kinase pathway by targeting claudin-2. Am J Physiol Cel Physiol, 2019, 316(1): C33–C47., DOI 10.1152/ajpcell.00047.2018 Beier LS, Piontek J, Piontek A, Protze J, Kobelt D, Walther W. Claudintargeted suicide gene therapy for claudin-overexpressing tumor cells by using modified Clostridium perfringens enterotoxin, CPE). Methods Mol Biol, Clifton, NJ,, 2022, 2521:173–88., DOI 10.1007/978-1-0716-2441-8_9 Arrojo ML, Oliveira KK, Bettim BB, Kowalski LP, Carraro DM, Meira I, et al. Tight junction gene expression in salivary gland tumors. Pathol Res Pract, 2022, 238:154113., DOI 10.1016/j.prp.2022.154113 Maria OM, Kim JW, Gerstenhaber JA, Baum BJ, Tran SD. Distribution of tight junction proteins in adult human salivary glands. J Histochem Cytochem official J Histochem Soc, 2008, 56(12):1093–8., DOI 10.1369/jhc. 2008.951780 Aoyama T, Takasawa A, Murata M, Osanai M, Takano K, Hasagawa T, et al. Immunoreactivity patterns of tight junction proteins are useful for differential diagnosis of human salivary gland tumors. Med Mol Morphol, 2019, 52(1):23–35., DOI 10.1007/s00795-018-0199-6 Morris LGT, Chandramohan R, West L, Zehir A, Chakravarty D, Pfister DG, et al. The molecular landscape of recurrent and metastatic head and neck cancers: Insights from a precision oncology sequencing platform. JAMA Oncol, 2017, 3(2): 244–55., DOI 10.1001/jamaoncol.2016.1790 Aro K, Rosa LE, Bello IO, Soini Y, Makitie AA, Salo T, et al. Expression pattern of claudins 1 and 3-an auxiliary tool in predicting behavior of mucoepidermoid carcinoma of salivary gland origin. Virchows Archiv : Int J Pathol, 2011, 458(3): 341–8., DOI 10.1007/s00428-010-1026-1 Garg PK, Sharma G, Rai S, Jakhetiya A. Primary salivary gland-type tumors of the lung: A systematic review and pooled analysis. Lung India, 2019, 36(2):118–22., DOI 10.4103/lungindia.lungindia_284_18 Li D, Pang X, Zhu X, Shanzhou Q, Wen G, Ma D. Low-dose radiotherapy combined with immunotherapy for suboral adenoid cystic carcinoma with bilateral lung metastasis: A case report and literature review. Oncol Lett, 2022, 24(2):279., DOI 10.3892/ol.2022.13399 Mueller SK, Haderlein M, Lettmaier S, Agaimy A, Haller F, Hecht M, et al. Targeted therapy, chemotherapy, immunotherapy and novel treatment options for different subtypes of salivary gland cancer. J Clin Med, 2022, 11(3):720., DOI 10. 3390/jcm11030720 Hu S, Gong J, Zhu X, Lu H. Pulmonary salivary gland tumor, mucoepidermoid carcinoma: A literature review. J Oncol, 2022, 2022:9742091., DOI 10.1155/2022/9742091 de Sousa LG, Jovanovic K, Ferrarotto R. Metastatic adenoid cystic carcinoma: Genomic landscape and emerging treatments. Curr Treat Options Oncol, 2022, 23(8):1135–50., DOI 10.1007/s11864-022-01001-y de Sousa LG, Neto FL, Lin J, Ferrarotto R. Treatment of recurrent or metastatic adenoid cystic carcinoma. Curr Oncol Rep, 2022, 24(5):621–31., DOI 10.1007/s11912-022-01233-z Roehlen N, Muller M, Nehme Z, Crouchet E, Juhling F, Del Zompo F, et al. Treatment of HCC with claudin-1-specific antibodies suppresses carcinogenic signaling and reprograms the tumor microenvironment. J Hepatol, 2023, 78(2): 343–55., DOI 10.1016/j.jhep.2022.10.011 Wei M, Zhang Y, Yang X, Ma P, Li Y, Wu Y, et al. Claudin-2 promotes colorectal cancer growth and metastasis by suppressing NDRG1 transcription. Clin translational Med, 2021, 11(12):e667., DOI 10.1002/ctm2.667 Maeda T, Murata M, Chiba H, Takasawa A, Tanaka S, Kojima T, et al. Claudin-4-targeted therapy using Clostridium perfringens enterotoxin for prostate cancer. The Prostate, 2012, 72(4):351–60., DOI 10.1002/pros.21436 Luo Y, Kishi S, Sasaki T, Ohmori H, Fujiwara-Tani R, Mori S, et al. Targeting claudin-4 enhances chemosensitivity in breast cancer. Cancer Sci, 2020, 111(5): 1840–50., DOI 10.1111/cas.14361 Li Y, Gong Y, Ning X, Peng D, Liu L, He S, et al. Downregulation of CLDN7 due to promoter hypermethylation is associated with human clear cell renal cell carcinoma progression and poor prognosis. J Exp Clin Cancer Res : CR, 2018, 37(1):276., DOI 10.1186/s13046-018-0924-y Philip R, Heiler S, Mu W, Buchler MW, Zoller M, Thuma F. Claudin-7 promotes the epithelial-mesenchymal transition in human colorectal cancer. Oncotarget, 2015, 6(4):2046–63., DOI 10.18632/oncotarget.2858 Hoggard J, Fan J, Lu Z, Lu Q, Sutton L, Chen YH. Claudin-7 increases chemosensitivity to cisplatin through the upregulation of caspase pathway in human NCI-H522 lung cancer cells. Cancer Sci, 2013, 104(5):611–8., DOI 10. 1111/cas.12135 Xu G, Qian N, Liu Y, Li H, Yang C, Wang J, et al. Preclinical characterization of a Fab-like CD3/CLDN18.2 XFab® bispecific antibody against solid tumors. Immunobiology, 2022, 227(6):152283., DOI 10.1016/j.imbio.2022.152283 Wang F, Yang Y, Du X, Zhu X, Hu Y, Lu C, et al. Claudin18.2 as a potential therapeutic target for primary ovarian mucinous carcinomas andmetastatic ovarian mucinous carcinomas from upper gastrointestinal primary tumours. BMC cancer, 2023, 23(1):44., DOI 10.1186/s12885-023-10533-x Shitara K, Lordick F, Bang YJ, Enzinger P, Ilson D, Shah MA, et al. Zolbetuximab plus mFOLFOX6 in patients with cldn18.2-positive, HER2- negative, untreated, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma, SPOTLIGHT): A multicentre, randomised, double-blind, phase 3 trial. Lancet, 2023, 401(10389):1655–68. London, England., DOI 10.1016/s0140-6736(23)00620-7 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611328 EP 1611337 DI 10.3389/pore.2023.1611328 PG 10 ER PT J AU Ni, H Ding, X Wu, Sh Jin, X AF Ni, Hua Ding, Xinjia Wu, Shikai Jin, Xuan TI Case report: Clinical experience of treating pembrolizumab-induced systemic capillary leak syndrome (SCLS) in one patient with metastatic gastroesophageal junction squamous cell carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE immune checkpoint inhibitor; pembrolizumab; adverse effect; systemic capillary leak syndrome; SCLS ID immune checkpoint inhibitor; pembrolizumab; adverse effect; systemic capillary leak syndrome; SCLS AB Systemic capillary leak syndrome (SCLS) is a rare and complex adverse effect of immune checkpoint inhibitors (ICIs). The diagnosis of drug-induced SCLS is based on diffuse infusions of exudative fluid into the interstitial areas and the exclusion of other causes. The best management of ICIs-induced SCLS is not settled, though proper supportive care and corticosteroids were commonly applied as the first-line treatment. In our patient with advanced gastroesophageal junction squamous cell carcinoma, although ICIs-induced SCLS was successfully controlled with corticosteroids, the patient soon experienced cancer progress and died of pulmonary infections. Based on our experience and the reported cases by other hospitals, different stages of SCLS might respond differently to the same treatment. Therefore, a grading of ICIs-induced SCLS might help to stratify the patient for different treatment strategies. Besides, corticosteroids-sensitive patients, though waived from deadly SCLS, might be at higher risk of cancer progress and subsequent infections due to the application of corticosteroids. Considering that the inflammatory factors should be closely involved in the development of ICIsinduced SCLS, targeted therapy against the driver inflammatory cytokine might offer treatment regimens that are more effective and safer. C1 [Ni, Hua] Peking University First Hospital, Department of Medical OncologyBeijing, China. [Ding, Xinjia] Peking University First Hospital, Department of Medical OncologyBeijing, China. [Wu, Shikai] Peking University First Hospital, Department of Medical OncologyBeijing, China. [Jin, Xuan] Peking University First Hospital, Department of Medical OncologyBeijing, China. RP Jin, X (reprint author), Peking University First Hospital, Department of Medical Oncology, Beijing, China. EM jinxuanbdyy@outlook.com CR Clarkson B, Thompson D, Horwith M, Luckey EH. Cyclical edema and shock due to increased capillary permeability. Am J Med, 1960, 29:193–216., DOI 10.1016/ 0002-9343(60)90018-8 Siddall E, Khatri M, Radhakrishnan J. Capillary leak syndrome: etiologies, pathophysiology, and management. Kidney Int, 2017, 92(1):37–46., DOI 10.1016/j. kint.2016.11.029 Jeong GH, Lee KH, Lee IR, Oh JH, Kim DW, Shin JW, et al. Incidence of capillary leak syndrome as an adverse effect of drugs in cancer patients: a systematic review and Meta-Analysis. J Clin Med, 2019, 8(2):143., DOI 10.3390/jcm8020143 Izzedine H, Mathian A, Amoura Z, Ng JH, Jhaveri KD. Anticancer Drug- Induced capillary leak syndrome. Kidney Int Rep, 2022, 7(5):945–53., DOI 10.1016/j. ekir.2022.02.014 Qin H, Vlaminck B, Owoyemi I, Herrmann SM, Leung N, Markovic SN. Successful treatment of Pembrolizumab-Induced severe capillary leak syndrome and lymphatic capillary dysfunction. Mayo Clin Proc Innov Qual Outcomes, 2021, 5(3):670–4., DOI 10.1016/j.mayocpiqo.2021.01.004 Umeda Y, Hayashi H, Sugiyama S, Aoyama Y. Systemic capillary leak syndrome triggered by Anti-Programmed death 1 checkpoint inhibitor in psoriasis. J Dermatol, 2020, 47(11):1322–5., DOI 10.1111/1346-8138.15541 Lescure C, Lescoat A, Sale A, Bazin Y, Duverge L, Desrues B, et al. Systemic capillary leak syndrome, Clarkson’s Disease, as a complication of Anti- Programmed death 1 immunotherapy. J Thorac Oncol, 2019, 14(6):e131–2., DOI 10.1016/j.jtho.2019.02.003 Abdel-Wahab N, ShahM, Suarez-Almazor ME. Adverse events associated with immune checkpoint blockade in patients with cancer: a systematic review of case reports. PLoS One, 2016, 11(7):e0160221., DOI 10.1371/journal.pone.0160221 Percik R, Nethanel A, Liel Y. Capillary-Leak syndrome: an unrecognized early immune adverse effect of Checkpoint-Inhibitors treatment. Immunotherapy, 2021, 13(8):653–9., DOI 10.2217/imt-2020-0332 Itzam Marin A, Deitz GA, Mudie LI, Reddy AK, Palestine AG. Bilateral choroidal effusions and angle closure in the setting of systemic capillary leak syndrome from HLA-Directed vaccine and pembrolizumab therapy for squamous cell carcinoma. Am J Ophthalmol Case Rep, 2023, 29:101777., DOI 10.1016/j.ajoc. 2022.101777 Dhir V, Arya V, Malav IC, Suryanarayanan BS, Gupta R, Dey AB. Idiopathic systemic capillary leak syndrome, SCLS): case report and systematic review of cases reported in the last 16 years. Intern Med, 2007, 46(12):899–904., DOI 10.2169/ internalmedicine.46.6129 Yabe H, Yabe M, Koike T, Shimizu T, Morimoto T, Kato S. Rapid improvement of Life-Threatening capillary leak syndrome after stem cell transplantation by bevacizumab. Blood, 2010, 115(13):2723–4., DOI 10.1182/ blood-2009-11-247056 Jolles S, Sewell WA,Misbah SA. Clinical uses of intravenous immunoglobulin. Clin Exp Immunol, 2005, 142(1):1–11., DOI 10.1111/j.1365-2249.2005.02834.x Lambert M, Launay D, Hachulla E, Morell-Dubois S, Soland V, Queyrel V, et al. High-Dose intravenous immunoglobulins dramatically reverse systemic capillary leak syndrome. Crit Care Med, 2008, 36(7):2184–7., DOI 10.1097/CCM. 0b013e31817d7c71 Pineton de Chambrun M, Gousseff M, Mauhin W, Lega JC, Lambert M, Riviere S, et al. Intravenous immunoglobulins improve survival in monoclonal Gammopathy-Associated systemic Capillary-Leak syndrome. Am J Med, 2017, 130(10):1219 e19–1219.e27., DOI 10.1016/j.amjmed.2017.05.023 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611330 EP 1611333 DI 10.3389/pore.2023.1611330 PG 4 ER PT J AU Tie, W Ma, T Yi, Z Liu, J Li, Y Bai, J Li, L Zhang, L AF Tie, Wenting Ma, Tao Yi, Zhigang Liu, Jia Li, Yanhong Bai, Jun Li, Lijuan Zhang, Liansheng TI Obesity as a risk factor for multiple myeloma: insight on the role of adipokines SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE multiple myeloma; adipokine; obesity; adiponectin; leptin ID multiple myeloma; adipokine; obesity; adiponectin; leptin AB Multiple myeloma (MM) is a hematologic disorder characterized by the accumulation of malignant plasma cells in the bone marrow. Genetic and environmental factors are contributed to the etiology of MM. Notably, studies have shown that obesity increases the risk of MM and worsens outcomes for MM patients. Adipokines play an important role in mediating the close association between MM and metabolic derangements. In this review, we summarize the epidemiologic studies to show that the risk of MM is increased in obese. Accumulating clinical evidence suggests that adipokines could display a correlation with MM. In vitro and in vivo studies have shown that adipokines are linked to MM, including roles in the biological behavior of MM cells, cancer-associated bone loss, the progression of MM, and drug resistance. Current and potential therapeutic strategies targeted to adipokines are discussed, proposing that adipokines can guide early patient diagnosis and treatment. C1 [Tie, Wenting] Lanzhou University Second Hospital, Department of HematologyLanzhou, China. [Ma, Tao] The Affiliated Hospital of Southwest Medical University, Department of HematologyLuzhou, China. [Yi, Zhigang] Lanzhou University Second Hospital, Department of HematologyLanzhou, China. [Liu, Jia] Lanzhou University Second Hospital, Department of HematologyLanzhou, China. [Li, Yanhong] Lanzhou University Second Hospital, Department of HematologyLanzhou, China. [Bai, Jun] Lanzhou University Second Hospital, Department of HematologyLanzhou, China. [Li, Lijuan] Lanzhou University Second Hospital, Department of HematologyLanzhou, China. [Zhang, Liansheng] Lanzhou University Second Hospital, Department of HematologyLanzhou, China. RP Zhang, L (reprint author), Lanzhou University Second Hospital, Department of Hematology, Lanzhou, China. EM doctorzhanglsh@sina.com CR Cowan AJ, Green DJ, KwokM, Lee S, Coffey DG, Holmberg LA, et al. Diagnosis and management of multiple myeloma: A review. JAMA, 2022, 327:464–77., DOI 10. 1001/jama.2022.0003 van de Donk NWCJ, Pawlyn C, Yong KL. Multiple myeloma. Lancet, 2021, 397:410–27., DOI 10.1016/S0140-6736(21)00135-5 Podar K, Richardson PG, Hideshima T, Chauhan D, Anderson KC. The malignant clone and the bone-marrow environment. Best Pract Res Clin Haematol, 2007, 20:597–612., DOI 10.1016/j.beha.2007.08.002 Teras LR, Kitahara CM, Birmann BM, Hartge PA, Wang SS, Robien K, et al. Body size and multiple myeloma mortality: A pooled analysis of 20 prospective studies. Br J Haematol, 2014, 166:667–76., DOI 10.1111/bjh.12935 Kleinstern G, Larson DR, Allmer C, Norman AD, Muntifering G, Sinnwell J, et al. Body mass index associated with monoclonal gammopathy of undetermined significance, MGUS, progression in Olmsted County, Minnesota. Blood Cancer J, 2022, 12:67., DOI 10.1038/s41408-022-00659-9 Went M, Cornish AJ, Law PJ, Kinnersley B, van Duin M, Weinhold N, et al. Search for multiple myeloma risk factors using Mendelian randomization. Blood Adv, 2020, 4:2172–9., DOI 10.1182/bloodadvances.2020001502 Veldhuis-Vlug AG, Rosen CJ. Clinical implications of bone marrow adiposity. J Intern Med, 2018, 283:121–39., DOI 10.1111/joim.12718 Morris EV, Suchacki KJ, Hocking J, Cartwright R, Sowman A, Gamez B, et al. Myeloma cells down-regulate adiponectin in bone marrow adipocytes via TNFalpha. J Bone Mineral Res, 2020, 35:942–55., DOI 10.1002/jbmr.3951 Bullwinkle EM, Parker MD, Bonan NF, Falkenberg LG, Davison SP, DeCicco- Skinner KL. Adipocytes contribute to the growth and progression of multiple myeloma: Unraveling obesity related differences in adipocyte signaling. Cancer Lett, 2016, 380:114–21., DOI 10.1016/j.canlet.2016.06.010 Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F, Straif K, et al. Body fatness and cancer--viewpoint of the IARC working group. N Engl J Med, 2016, 375:794–8., DOI 10.1056/NEJMsr1606602 Avgerinos KI, Spyrou N, Mantzoros CS, Dalamaga M. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism, 2019, 92: 121–35., DOI 10.1016/j.metabol.2018.11.001 Tarasiuk A, Mosinska P, Fichna J. The mechanisms linking obesity to colon cancer: An overview. Obes Res Clin Pract, 2018, 12:251–9., DOI 10.1016/j.orcp.2018. 01.005 Divella R, De Luca R, Abbate I, Naglieri E, Daniele A. Obesity and cancer: The role of adipose tissue and adipo-cytokines-induced chronic inflammation. J Cancer, 2016, 7:2346–59., DOI 10.7150/jca.16884 Diedrich J, Gusky HC, Podgorski I. Adipose tissue dysfunction and its effects on tumor metabolism. Horm Mol Biol Clin Investig, 2015, 21:17–41., DOI 10.1515/ hmbci-2014-0045 Vegiopoulos A, Rohm M, Herzig S. Adipose tissue: Between the extremes. EMBO J, 2017, 36:1999–2017., DOI 10.15252/embj.201696206 Fasshauer M, Bluher M. Adipokines in health and disease. Trends Pharmacol Sci, 2015, 36:461–70., DOI 10.1016/j.tips.2015.04.014 Khandekar MJ, Cohen P, Spiegelman BM. Molecular mechanisms of cancer development in obesity. Nat Rev Cancer, 2011, 11:886–95., DOI 10.1038/nrc3174 Khoramipour K, Chamari K, Hekmatikar AA, Ziyaiyan A, Taherkhani S, Elguindy NM, et al. Adiponectin: Structure, physiological functions, role in diseases, and effects of nutrition. Nutrients, 2021, 13:1180., DOI 10.3390/nu13041180 Cawthorn WP, Scheller EL, Learman BS, Parlee SD, Simon BR, Mori H, et al. Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction. Cell Metab, 2014, 20:368–75., DOI 10.1016/j.cmet.2014.06.003 Yamauchi T, Iwabu M, Okada-Iwabu M, Kadowaki T. Adiponectin receptors: A review of their structure, function and how they work. Best Pract Res Clin Endocrinol Metab, 2014, 28:15–23., DOI 10.1016/j.beem.2013.09.003 Hug C, Wang J, Ahmad NS, Bogan JS, Tsao T-S, Lodish HF. T-cadherin is a receptor for hexameric and high-molecular-weight forms of Acrp30/adiponectin. Proc Natl Acad Sci U. S. A., 2004, 101:10308–13., DOI 10.1073/pnas.0403382101 Fang H, Judd RL. Adiponectin regulation and function. Compr Physiol, 2018, 8:1031–63., DOI 10.1002/cphy.c170046 Lindberg S, Jensen JS, Pedersen SH, Galatius S, Frystyk J, Flyvbjerg A, et al. Low adiponectin levels and increased risk of type 2 diabetes in patients with myocardial infarction. Diabetes Care, 2014, 37:3003–8., DOI 10.2337/dc14-0932 Liu L, Shi Z, Ji X, Zhang W, Luan J, Zahr T, et al. Adipokines, adiposity, and atherosclerosis. Cell Mol Life Sci, 2022, 79:272., DOI 10.1007/s00018-022-04286-2 Yanai H, Yoshida H. Beneficial effects of adiponectin on glucose and lipid metabolism and atherosclerotic progression: Mechanisms and perspectives. Int J Mol Sci, 2019, 20:1190., DOI 10.3390/ijms20051190 Ma W, Zhang S, Li Y, Chen T, Yang Q, Feng X. Adiponectin alleviates nonalcoholic fatty liver injury via regulating oxidative stress in liver cells. Minerva Med, 2020, 113:990–9., DOI 10.23736/S0026-4806.20.06734-8 Tilg H, Moschen AR. Adipocytokines: Mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol, 2006, 6:772–83., DOI 10.1038/ nri1937 Di Zazzo E, Polito R, Bartollino S, Nigro E, Porcile C, Bianco A, et al. Adiponectin as link factor between adipose tissue and cancer. Int J Mol Sci, 2019, 20:839., DOI 10.3390/ijms20040839 Hofmann JN, Mailankody S, Korde N, Wang Y, Tageja N, Costello R, et al. Circulating adiponectin levels differ between patients with multiple myeloma and its precursor disease. Obesity, Silver Spring,, 2017, 25:1317–20., DOI 10.1002/oby. 21894 Hofmann JN, Birmann BM, Teras LR, Pfeiffer RM, Wang Y, Albanes D, et al. Low levels of circulating adiponectin are associated with multiple myeloma risk in overweight and obese individuals. Cancer Res, 2016, 76:1935–41., DOI 10.1158/0008- 5472.CAN-15-2406 Fowler JA, Lwin ST, Drake MT, Edwards JR, Kyle RA, Mundy GR, et al. Hostderived adiponectin is tumor-suppressive and a novel therapeutic target for multiple myeloma and the associated bone disease. Blood, 2011, 118:5872–82., DOI 10.1182/blood-2011-01-330407 Medina EA, Oberheu K, Polusani SR, Ortega V, Velagaleti GVN, Oyajobi BO. PKA/AMPK signaling in relation to adiponectin’s antiproliferative effect on multiple myeloma cells. Leukemia, 2014, 28:2080–9., DOI 10.1038/leu.2014.112 Akhmetzyanova I, Aaron T, Galbo P, Tikhonova A, Dolgalev I, Tanaka M, et al. Tissue-resident macrophages promote early dissemination of multiple myeloma via IL-6 and TNFα. Blood Adv, 2021, 5:3592–608., DOI 10.1182/ bloodadvances.2021005327 Meacham CE, Jeffery EC, Burgess RJ, Sivakumar CD, Arora MA, Stanley AM, et al. Adiponectin receptors sustain haematopoietic stem cells throughout adulthood by protecting them from inflammation. Nat Cell Biol, 2022, 24: 697–707., DOI 10.1038/s41556-022-00909-9 Bernstein ZS, Kim EB, Raje N. Bone disease inmultiple myeloma: Biologic and clinical implications. Cells, 2022, 11:2308., DOI 10.3390/cells11152308 Liu Z, Liu H, Li Y, Wang Y, Xing R, Mi F, et al. Adiponectin inhibits the differentiation and maturation of osteoclasts via the mTOR pathway in multiple myeloma. Int J Mol Med, 2020, 45:1112–20., DOI 10.3892/ijmm.2020.4475 Olechnowicz SWZ, Weivoda MM, Lwin ST, Leung SK, Gooding S, Nador G, et al. Multiple myeloma increases nerve growth factor and other pain-related markers through interactions with the bone microenvironment. Scientific Rep, 2019, 9:14189., DOI 10.1038/s41598-019-50591-5 Peterson SJ, Drummond G, Kim DH, Li M, Kruger AL, Ikehara S, et al. L-4F treatment reduces adiposity, increases adiponectin levels, and improves insulin sensitivity in obese mice. J Lipid Res, 2008, 49:1658–69., DOI 10.1194/jlr.M800046- JLR200 Zhang Y, Proenca R, Maffei M, BaroneM, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature, 1994, 372: 425–32., DOI 10.1038/372425a0 Trinh T, Broxmeyer HE. Role for leptin and leptin receptors in stem cells during health and diseases. Stem Cell Rev Rep, 2021, 17:511–22., DOI 10.1007/ s12015-021-10132-y Obradovic M, Sudar-Milovanovic E, Soskic S, Essack M, Arya S, Stewart AJ, et al. Leptin and obesity: Role and clinical implication. Front Endocrinol, 2021, 12: 585887., DOI 10.3389/fendo.2021.585887 Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med, 1996, 334:292–5., DOI 10.1056/ NEJM199602013340503 Mhaidat NM, Alzoubi KH, Kubas MA, Banihani MN, Hamdan N, Al-Jaberi TM. High levels of leptin and non-high molecular weight-adiponectin in patients with colorectal cancer: Association with chemotherapy and common genetic polymorphisms. Biomed Rep, 2021, 14:13., DOI 10.3892/br.2020.1389 Garcia-Estevez L, Gonzalez-Martinez S, Moreno-Bueno G. The leptin Axis and its association with the adaptive immune system in breast cancer. Front Immunol, 2021, 12:784823., DOI 10.3389/fimmu.2021.784823 Xu CJ, Dong LL, Kang XL, Li ZM, Zhang HY. Leptin promotes proliferation and inhibits apoptosis of prostate cancer cells by regulating ERK1/2 signaling pathway. Eur Rev Med Pharmacol Sci, 2020, 24:8341–8., DOI 10.26355/eurrev_ 202008_22630 Wang J, Zhou F, Li F, Wang B, Hu Y, Li X. Autocrined leptin promotes proliferation of non-small cell lung cancer, NSCLC, via PI3K/AKT and p53 pathways. Ann Transl Med, 2021, 9:568., DOI 10.21037/atm-20-7482 Kashiwagi E, Abe T, Kinoshita F, UshijimaM,Masaoka H, Shiota M, et al. The role of adipocytokines and their receptors in bladder cancer: Expression of adiponectin or leptin is an independent prognosticator. Am J Transl Res, 2020, 12:e195–45., DOI 10.1016/s2666-1683(20)32677-x Pamuk GE, Demir M, Harmandar F, Yesil Y, Turgut B, Vural O. Leptin and resistin levels in serum of patients with hematologic malignancies: Correlation with clinical characteristics. Exp Oncol, 2006, 28:241–4. Reseland JE, Reppe S, Olstad OK, Hjorth-Hansen H, Brenne AT, Syversen U, et al. Abnormal adipokine levels and leptin-induced changes in gene expression profiles in multiple myeloma. Eur JHaematol, 2009, 83:460–70., DOI 10.1111/j.1600- 0609.2009.01311.x Liu R, Gao D, Lv Y, ZhaiM,He A. Importance of circulating adipocytokines in multiple myeloma: A systematic review and meta-analysis based on case-control studies. BMC Endocr Disord, 2022, 22:29., DOI 10.1186/s12902-022-00939-2 Alexandrakis MG, Passam FH, Sfiridaki A, Pappa CA, Moschandrea JA, Kandidakis E, et al. Serum levels of leptin in multiple myeloma patients and its relation to angiogenic and inflammatory cytokines. The Int J Biol Markers, 2004, 19:52–7., DOI 10.5301/jbm.2008.510 Hofmann JN, Liao LM, Pollak MN, Wang Y, Pfeiffer RM, Baris D, et al. A prospective study of circulating adipokine levels and risk of multiple myeloma. Blood, 2012, 120:4418–20., DOI 10.1182/blood-2012-06-438606 YuW, Cao D-D, Li Q-B, Mei H-L, Hu Y, Guo T. Adipocytes secreted leptin is a pro-tumor factor for survival of multiple myeloma under chemotherapy. Oncotarget, 2016, 7:86075–86., DOI 10.18632/oncotarget.13342 Liu Z, Xu J, He J, Liu H, Lin P, Wan X, et al. Mature adipocytes in bone marrow protect myeloma cells against chemotherapy through autophagy activation. Oncotarget, 2015, 6:34329–41., DOI 10.18632/oncotarget.6020 Chan AC, Neeson P, Leeansyah E, Tainton K, Quach H, Prince HM, et al. Natural killer T cell defects in multiple myeloma and the impact of lenalidomide therapy. Clin Exp Immunol, 2014, 175:49–58., DOI 10.1111/cei.12196 Dhodapkar MV, Richter J. Harnessing natural killer T, NKT, cells in human myeloma: Progress and challenges. Clin Immunol, Orlando, Fla, 2011, 140:160–6., DOI 10.1016/j.clim.2010.12.010 Favreau M, Menu E, Gaublomme D, Vanderkerken K, Faict S, Maes K, et al. Leptin receptor antagonism of iNKT cell function: A novel strategy to combat multiple myeloma. Leukemia, 2017, 31:2678–85., DOI 10.1038/leu.2017.146 Wang Z, Aguilar EG, Luna JI, Dunai C, Khuat LT, Le CT, et al. Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade. Nat Med, 2019, 25:141–51., DOI 10.1038/s41591-018-0221-5 Sih R, Morley JE, Kaiser FE, Perry HM, Patrick P, Ross C. Testosterone replacement in older hypogonadal men: A 12-month randomized controlled trial. J Clin Endocrinol Metab, 1997, 82:1661–7., DOI 10.1210/jcem.82.6.3988 Simon C, Gronfier C, Schlienger JL, Brandenberger G. Circadian and ultradian variations of leptin in normal man under continuous enteral nutrition: Relationship to sleep and body temperature. J Clin Endocrinol Metab, 1998, 83: 1893–9., DOI 10.1210/jcem.83.6.4864 Montague CT, Prins JB, Sanders L, Digby JE, O’Rahilly S. Depot- and sexspecific differences in human leptin mRNA expression: Implications for the control of regional fat distribution. Diabetes, 1997, 46:342–7., DOI 10.2337/diab.46.3.342 Samal B, Sun Y, Stearns G, Xie C, Suggs S, McNiece I. Cloning and characterization of the cDNA encoding a novel human pre-B-cell colonyenhancing factor. Mol Cell Biol, 1994, 14:1431–7., DOI 10.1128/mcb.14.2.1431 Zhai RG, Rizzi M, Garavaglia S. Nicotinamide/nicotinic acid mononucleotide adenylyltransferase, new insights into an ancient enzyme. Cell Mol Life Sci, 2009, 66:2805–18., DOI 10.1007/s00018-009-0047-x Fukuhara A, Matsuda M, Nishizawa M, Segawa K, Tanaka M, Kishimoto K, et al. Visfatin: A protein secreted by visceral fat that mimics the effects of insulin. Science, New York, N.Y.,, 2005, 307:426–30., DOI 10.1126/science.1097243 Chang Y-H, Chang D-M, Lin K-C, Shin S-J, Lee Y-J. Visfatin in overweight/ obesity, type 2 diabetes mellitus, insulin resistance, metabolic syndrome and cardiovascular diseases: A meta-analysis and systemic review. Diabetes Metab Res Rev, 2011, 27:515–27., DOI 10.1002/dmrr.1201 Fazeli MS, Dashti H, Akbarzadeh S, Assadi M, Aminian A, Keramati MR, et al. Circulating levels of novel adipocytokines in patients with colorectal cancer. Cytokine, 2013, 62:81–5., DOI 10.1016/j.cyto.2013.02.012 Nakajima TE, Yamada Y, Hamano T, Furuta K, Gotoda T, Katai H, et al. Adipocytokine levels in gastric cancer patients: Resistin and visfatin as biomarkers of gastric cancer. J Gastroenterol, 2009, 44:685–90., DOI 10.1007/s00535-009-0063-5 Reddy PS, Umesh S, Thota B, Tandon A, Pandey P, Hegde AS, et al. PBEF1/ NAmPRTase/Visfatin: A potential malignant astrocytoma/glioblastoma serum marker with prognostic value. Cancer Biol Ther, 2008, 7:663–8., DOI 10.4161/cbt. 7.5.5663 Lu Z, Fang Z, Guo Y, Liu X, Chen S. Cisplatin resistance of NSCLC cells involves upregulation of visfatin through activation of its transcription and stabilization of mRNA. Chem Biol Interact, 2022, 351:109705:109705., DOI 10. 1016/j.cbi.2021.109705 Bong IPN, Ng CC, Fakiruddin SK, Lim MN, Zakaria Z. Small interfering RNA-mediated silencing of nicotinamide phosphoribosyltransferase, NAMPT, and lysosomal trafficking regulator, lyst, induce growth inhibition and apoptosis in human multiple myeloma cells: A preliminary study. Bosnian J Basic Med Sci, 2016, 16:268–75., DOI 10.17305/bjbms.2016.1568 Cea M, Cagnetta A, Patrone F, Nencioni A, Gobbi M, Anderson KC. Intracellular NAD(+, depletion induces autophagic death in multiple myeloma cells. Autophagy, 2013, 9:410–2., DOI 10.4161/auto.22866 Cea M, Cagnetta A, Fulciniti M, Tai Y-T, Hideshima T, Chauhan D, et al. Targeting NAD+ salvage pathway induces autophagy in multiple myeloma cells via mTORC1 and extracellular signal-regulated kinase, ERK1/2, inhibition. Blood, 2012, 120:3519–29., DOI 10.1182/blood-2012-03-416776 Cagnetta A, Cea M, Calimeri T, Acharya C, Fulciniti M, Tai Y-T, et al. Intracellular NAD⁺ depletion enhances bortezomib-induced anti-myeloma activity. Blood, 2013, 122:1243–55., DOI 10.1182/blood-2013-02-483511 Kennedy BE, Sadek M, Elnenaei MO, Reiman A, Gujar SA. Targeting NAD+ synthesis to potentiate CD38-based immunotherapy of multiple myeloma. Trends Cancer, 2020, 6:9–12., DOI 10.1016/j.trecan.2019.11.005 Venkateshaiah SU, Khan S, Ling W, Bam R, Li X, van Rhee F, et al. NAMPT/ PBEF1 enzymatic activity is indispensable for myeloma cell growth and osteoclast activity. Exp Hematol, 2013, 41:547–57., DOI 10.1016/j.exphem.2013.02.008 Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, et al. The hormone resistin links obesity to diabetes. Nature, 2001, 409:307–12., DOI 10.1038/ 35053000 Patel L, Buckels AC, Kinghorn IJ, Murdock PR, Holbrook JD, Plumpton C, et al. Resistin is expressed in human macrophages and directly regulated by PPAR gamma activators. Biochem Biophys Res Commun, 2003, 300:472–6., DOI 10.1016/ s0006-291x(02)02841-3 Deb A, Deshmukh B, Ramteke P, Bhati FK, Bhat MK. Resistin: A journey from metabolism to cancer. Transl Oncol, 2021, 14:101178., DOI 10.1016/j.tranon.2021. 101178 Qiu L, Zhang G-F, Yu L, Wang H-Y, Jia X-J, Wang T-J. Novel oncogenic and chemoresistance-inducing functions of resistin in ovarian cancer cells require miRNAs-mediated induction of epithelial-to-mesenchymal transition. Scientific Rep, 2018, 8:12522., DOI 10.1038/s41598-018-30978-6 Lee JO, Kim N, Lee HJ, Lee YW, Kim SJ, Park SH, et al. Resistin, a fat-derived secretory factor, promotes metastasis of MDA-MB-231 human breast cancer cells through ERM activation. Scientific Rep, 2016, 6:18923., DOI 10.1038/srep18923 Pang L, Zhang Y, Yu Y, Zhang S. Resistin promotes the expression of vascular endothelial growth factor in ovary carcinoma cells. Int J Mol Sci, 2013, 14:9751–66., DOI 10.3390/ijms14059751 Rak A, Drwal E, Wrobel A, Gregoraszczuk EL. Resistin is a survival factor for porcine ovarian follicular cells. Reproduction, Cambridge, England,, 2015, 150: 343–55., DOI 10.1530/REP-15-0255 Mu H, Ohashi R, Yan S, Chai H, Yang H, Lin P, et al. Adipokine resistin promotes in vitro angiogenesis of human endothelial cells. Cardiovasc Res, 2006, 70:146–57., DOI 10.1016/j.cardiores.2006.01.015 Owecki M, Miczke A, Nikisch E, Pupek-Musialik D, Sowinski J. Serumresistin concentrations are higher in human obesity but independent from insulin resistance. Exp Clin Endocrinol Diabetes, 2011, 119:117–21., DOI 10.1055/s-0030- 1263111 Deshmukh SK, Srivastava SK, Zubair H, Bhardwaj A, Tyagi N, Al-Ghadhban A, et al. Resistin potentiates chemoresistance and stemness of breast cancer cells: Implications for racially disparate therapeutic outcomes. Cancer Lett, 2017, 396: 21–9., DOI 10.1016/j.canlet.2017.03.010 Benomar Y, Gertler A, De Lacy P, Crepin D, Ould Hamouda H, Riffault L, et al. Central resistin overexposure induces insulin resistance through Toll-like receptor 4. Diabetes, 2013, 62:102–14., DOI 10.2337/db12-0237 DalamagaM, Karmaniolas K, Panagiotou A, Hsi A, Chamberland J, Dimas C, et al. Low circulating adiponectin and resistin, but not leptin, levels are associated with multiple myeloma risk: A case-control study. Cancer Causes Control, 2009, 20: 193–9., DOI 10.1007/s10552-008-9233-7 Santo L, Teras LR, Giles GG, Weinstein SJ, Albanes D, Wang Y, et al. Circulating resistin levels and risk of multiple myeloma in three prospective cohorts. Br J Cancer, 2017, 117:1241–5., DOI 10.1038/bjc.2017.282 Pang J, Shi Q, Liu Z, He J, Liu H, Lin P, et al. Resistin induces multidrug resistance in myeloma by inhibiting cell death and upregulating ABC transporter expression. Haematologica, 2017, 102:1273–80., DOI 10.3324/haematol.2016.154062 Millar SR, Navarro P, Harrington JM, Shivappa N, Hebert JR, Perry IJ, et al. Dietary score associations with markers of chronic low-grade inflammation: A cross-sectional comparative analysis of a middle-to older-aged population. Eur J Nutr, 2022, 61:3377–90., DOI 10.1007/s00394-022-02892-1 Tatemoto K, Hosoya M, Habata Y, Fujii R, Kakegawa T, Zou MX, et al. Isolation and characterization of a novel endogenous peptide ligand for the human APJ receptor. Biochem Biophys Res Commun, 1998, 251:471–6., DOI 10.1006/bbrc. 1998.9489 Charo DN, Ho M, Fajardo G, Kawana M, Kundu RK, Sheikh AY, et al. Endogenous regulation of cardiovascular function by apelin-APJ. Am J Physiol Heart Circulatory Physiol, 2009, 297:H1904–13., DOI 10.1152/ajpheart.00686.2009 Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin Endocrinol Metab, 2004, 89:2548–56., DOI 10.1210/jc.2004-0395 Dawid M, Mlyczynska E, Jurek M, Respekta N, Pich K, Kurowska P, et al. Apelin, APJ, and ELABELA: Role in placental function, pregnancy, and foetal development-an overview. Cells, 2021, 11:99., DOI 10.3390/ cells11010099 Boucher J, Masri B, Daviaud D, Gesta S, Guigne C, Mazzucotelli A, et al. Apelin, a newly identified adipokine up-regulated by insulin and obesity. Endocrinology, 2005, 146:1764–71., DOI 10.1210/en.2004-1427 Picault F-X, Chaves-Almagro C, Projetti F, Prats H, Masri B, Audigier Y. Tumour co-expression of apelin and its receptor is the basis of an autocrine loop involved in the growth of colon adenocarcinomas. Eur J Cancer, 2014, 50:663–74., DOI 10.1016/j.ejca.2013.11.017 Sorli SC, Le Gonidec S, Knibiehler B, Audigier Y. Apelin is a potent activator of tumour neoangiogenesis. Oncogene, 2007, 26:7692–9., DOI 10.1038/sj.onc. 1210573 Kalin RE, Glass R. APLN/APLNR signaling controls key pathological parameters of glioblastoma. Cancers, 2021, 13:3899., DOI 10.3390/ cancers13153899 Maden M, Pamuk ON, Pamuk GE. High apelin levels could be used as a diagnostic marker in multiple myeloma: A comparative study. Cancer Biomark, 2016, 17:391–6., DOI 10.3233/CBM-160654 Fischer TF, Beck-Sickinger AG. Chemerin - exploring a versatile adipokine. Biol Chem, 2022, 403:625–42., DOI 10.1515/hsz-2021-0409 Helfer G, Wu Q-F. Chemerin: A multifaceted adipokine involved in metabolic disorders. J Endocrinol, 2018, 238:R79–R94., DOI 10.1530/JOE-18-0174 Bozaoglu K, Curran JE, Stocker CJ, Zaibi MS, Segal D, Konstantopoulos N, et al. Chemerin, a novel adipokine in the regulation of angiogenesis. J Clin Endocrinol Metab, 2010, 95:2476–85., DOI 10.1210/jc.2010-0042 Rourke JL, Dranse HJ, Sinal CJ. Towards an integrative approach to understanding the role of chemerin in human health and disease. Obes Rev, 2013, 14:245–62., DOI 10.1111/obr.12009 Wang C, Wu WKK, Liu X, To K-F, Chen GG, Yu J, et al. Increased serum chemerin level promotes cellular invasiveness in gastric cancer: A clinical and experimental study. Peptides, 2014, 51:131–8., DOI 10.1016/j.peptides.2013.10.009 Erdogan S, Yilmaz FM, Yazici O, Yozgat A, Sezer S, Ozdemir N, et al. Inflammation and chemerin in colorectal cancer. Tumour Biol, 2016, 37:6337–42., DOI 10.1007/s13277-015-4483-y Wang Q, Wang H, Ding Y, Wan M, Xu M. The role of adipokines in pancreatic cancer. Front Oncol, 2022, 12:926230., DOI 10.3389/fonc.2022. 926230 Gao C, Shi J, Zhang J, Li Y, Zhang Y. Chemerin promotes proliferation and migration of ovarian cancer cells by upregulating expression of PD-L1. J Zhejiang Univ Sci B, 2022, 23:164–70., DOI 10.1631/jzus.B2100392 Friebus-Kardash J, Schulz P, Reinicke S, Karthaus C, Schefer Q, Bandholtz S, et al. A chemerin peptide analog stimulates tumor growth in two xenograft mouse models of human colorectal carcinoma. Cancers, 2021, 14:125., DOI 10.3390/ cancers14010125 Westhrin M, Moen SH, Kristensen IB, Buene G, Mylin AK, Turesson I, et al. Chemerin is elevated in multiple myeloma patients and is expressed by stromal cells and pre-adipocytes. Biomark Res, 2018, 6:21., DOI 10.1186/s40364-018-0134-y Muruganandan S, Roman AA, Sinal CJ. Role of chemerin/ CMKLR1 signaling in adipogenesis and osteoblastogenesis of bone marrow stem cells. J Bone Mineral Res, 2010, 25:222–34., DOI 10.1359/jbmr.091106 Morris EV, Edwards CM. Adipokines, adiposity, and bone marrow adipocytes: Dangerous accomplices in multiple myeloma. J Cel Physiol, 2018, 233:9159–66., DOI 10.1002/jcp.26884 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611338 EP 1611347 DI 10.3389/pore.2023.1611338 PG 10 ER PT J TI A unique case of AH-dominant type nodular pulmonary amyloidosis presenting as a spontaneous pneumothorax: a case report and review of the literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE amyloidosis; nodular pulmonary amyloidosis; amyloidoma; AH amyloidosis; pneumothorax ID amyloidosis; nodular pulmonary amyloidosis; amyloidoma; AH amyloidosis; pneumothorax AB Amyloidosis is a rare metabolic disorder primarily brought on by misfolding of an autologous protein, which causes its local or systemic deposition in an aberrant fibrillar form. It is quite rare for pulmonary tissue to be impacted by amyloidosis; of the three forms it can take when involving pulmonary tissue, nodular pulmonary amyloidosis is the most uncommon. Nodular pulmonary amyloidosis rarely induces clinical symptoms, and most often, it is discovered accidentally during an autopsy or via imaging techniques. Only one case of nodular pulmonary amyloidosis, which manifested as a spontaneous pneumothorax, was found in the literature. In terms of more precise subtyping, nodular amyloidosis is typically AL or mixed AL/AH type. No publications on AH-dominant type of nodular amyloidosis were found in the literature. We present a case of an 81 years-old male with nodular pulmonary AH-dominant type amyloidosis who presented with spontaneous pneumothorax. For a deeper understanding of the subject, this study also provides a review of the literature on cases with nodular pulmonary amyloidosis in relation to precise amyloid fibril subtyping. Since it is often a difficult process, accurate amyloid type identification is rarely accomplished. However, this information is very helpful for identifying the underlying disease process (if any) and outlining the subsequent diagnostic and treatment steps. Even so, it is crucial to be aware of this unit and make sure it is taken into consideration when making a differential diagnosis of pulmonary lesions. CR Milani P, Basset M, Russo F, Foli A, Palladini G, Merlini G. The lung in amyloidosis. Eur Respir Rev, 2017, 26(145):170046., DOI 10.1183/16000617.0046- 2017 Yang MC, Blutreich A, Das K. Nodular pulmonary amyloidosis with an unusual protein composition diagnosed by fine-needle aspiration biopsy: a case report. Diagn Cytopathol, 2009, 37(4):286–9., DOI 10.1002/dc.21023 Rajagopala S, Singh N, Gupta K, Gupta D. Pulmonary amyloidosis in sjogren’s syndrome: a case report and systematic review of the literature. Respirology, 2010, 15(5):860–6., DOI 10.1111/j.1440-1843.2010.01772.x Kruczak K, Duplaga M, Sanak M, Papla B, Soja J, Nizankowska-Mogilnicka E, et al. Transthyretin amyloidosis with pulmonary involvement in a patient with monoclonal gammapathy. Pneumonol Alergol Pol, 2013, 81(6):537–41., DOI 10. 5603/arm.35995 Miyamoto T, Kobayashi T, Makiyama M, Kitada S, Fujishima M, Hagari Y, et al. Monoclonality of infiltrating plasma cells in primary pulmonary nodular amyloidosis: detection with polymerase chain reaction. J Clin Pathol, 1999, 52(6): 464–7., DOI 10.1136/jcp.52.6.464 Davis CJ, Butchart EG, Gibbs AR. Nodular pulmonary amyloidosis occurring in association with pulmonary lymphoma. Thorax, 1991, 46(3):217–8., DOI 10.1136/ thx.46.3.217 Roden AC, Aubry MC, Zhang K, Brady JO, Levin D, Dogan A, et al. Nodular senile pulmonary amyloidosis: a unique case confirmed by immunohistochemistry, mass spectrometry, and genetic study. Hum Pathol, 2010, 41(7):1040–5., DOI 10. 1016/j.humpath.2009.11.019 Kinoshita Y, Ikeda T, Miyamura T, Ueda Y, Yoshida Y, Ueda M, et al. Nodular pulmonary amyloidosis associated with sjogren’s syndrome. Intern Med, 2022, 61(6):877–81., DOI 10.2169/internalmedicine.8169-21 Grogg KL, Aubry MC, Vrana JA, Theis JD, Dogan A. Nodular pulmonary amyloidosis is characterized by localized immunoglobulin deposition and is frequently associated with an indolent B-cell lymphoproliferative disorder. Am J Surg Pathol, 2013, 37(3):406–12., DOI 10.1097/PAS.0b013e318272fe19 Gorospe Sarasua L, Munoz-Molina GM, Arrieta P. Loculated secondary spontaneous pneumothorax: A very rare complication of nodular pulmonary amyloidosis. Arch Bronconeumol, 2016, 52(10):530., DOI 10.1016/j.arbres.2015. 012 11. Ikeda S, Takabayashi Y, Maejima Y, Tachibana N, Ehara T, Nezu A, et al. Nodular lung disease with five year survival and unilateral pleural effusion in AL amyloidosis. Amyloid, 1999, 6(4):292–6., DOI 10.3109/13506129909007343 Calatayud J, Candelas G, Gomez A, Morado C, Trancho FH. Nodular pulmonary amyloidosis in a patient with rheumatoid arthritis. Clin Rheumatol, 2007, 26(10):1797–8., DOI 10.1007/s10067-007-0552-y Moscatelli B, Capoccetta GB, Anselmi P, Di Girolamo M, Campagna D. Kappa L-chain primary nodular lung amyloidosis: a case report. Amyloid, 2011, 18(1):121–3., DOI 10.3109/13506129.2011.574354045 Ishii S, Kubota K, Minamimoto R, Kouketu R, Morooka M, Kawai S, et al. Lung amyloid nodule detected by 99mTc-aprotinin scintigraphy. Ann Nucl Med, 2012, 26(6):522–6., DOI 10.1007/s12149-012-0606-3 Chung YY, Wang CC, Lai KJ, Chang CC. Waldenstrom’smacroglobulinemiaassociated renal amyloidosis presenting as a solitary lung mass. Ren Fail, 2012, 34(9):1173–6., DOI 10.3109/0886022X.2012.717488 Fukatsu H, Miyoshi H, Ishiki K. Spontaneous resolution of multiple nodular pulmonary AA amyloidosis. Intern Med, 2010, 49(21):2303–7., DOI 10.2169/ internalmedicine.49.3894 Kaplan B, Martin BM, Boykov O, Gal R, Pras M, Shechtman I, et al. Codeposition of amyloidogenic immunoglobulin light and heavy chains in localized pulmonary amyloidosis. Virchows Arch, 2005, 447(4):756–61., DOI 10.1007/s00428- 005-0009-0 Seguchi T, Kyoraku Y, Saita K, Ihi T, Nagai M, Akiyama Y, et al. Human T-cell lymphotropic virus type I, HTLV-1, associated myelopathy and sjogren’s syndrome representing pulmonary nodular amyloidosis and multiple bullae: report of an autopsy case. Virchows Arch, 2006, 448(6):874–6., DOI 10.1007/ s00428-005-0028-x Wieker K, Rocken C, Koenigsmann M, Roessner A, Franke A. Pulmonary low-grade MALT-lymphoma associated with localized pulmonary amyloidosis. A case report. Amyloid, 2002, 9(3):190–3., DOI 10.3109/13506120209114821 Beer TW, Edwards CW. Pulmonary nodules due to reactive systemic amyloidosis, AA, in Crohn’s disease. Thorax, 1993, 48(12):1287–8., DOI 10.1136/thx.48.12.1287 Kamei K, Kusumoto K, Suzuki T. Pulmonary amyloidosis with pulmonary arteriovenous fistula. Chest, 1989, 96(6):1435–6., DOI 10.1378/chest.96.6.1435 Laden SA, Cohen ML, Harley RA. Nodular pulmonary amyloidosis with extrapulmonary involvement. Hum Pathol, 1984, 15(6):594–7., DOI 10.1016/s0046- 8177(84)80017-9 Kobayashi H, Matsuoka R, Kitamura S, Tsunoda N, Saito K. Sjogren’s syndrome with multiple bullae and pulmonary nodular amyloidosis. Chest, 1988, 94(2):438–40., DOI 10.1378/chest.94.2.438 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611390 EP 1611399 DI 10.3389/pore.2023.1611390 PG 10 ER PT J AU Wang, Q Liang, Q Wei, W Niu, W Liang, Ch Wang, X Wang, X Pan, H AF Wang, Qiang Liang, Qiujin Wei, Wuting Niu, Wenhao Liang, Chong Wang, Xiaoliang Wang, Xiaoxuan Pan, Hao TI Concordance analysis of cerebrospinal fluid with the tumor tissue for integrated diagnosis in gliomas based on next-generation sequencing SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE cerebrospinal fluid; glioma; circulating tumor DNA; integrated diagnosis; IDH1 R132C ID cerebrospinal fluid; glioma; circulating tumor DNA; integrated diagnosis; IDH1 R132C AB Purpose: The driver mutations of gliomas have been identified in cerebrospinal fluid (CSF). Here we compared the concordance between CSF and tumor tissue for integrated diagnosis in gliomas using next-generation sequencing (NGS) to evaluate the feasibility of CSF detection in gliomas. Patients and methods: 27 paired CSF/tumor tissues of glioma patients were sequenced by a customized gene panel based on NGS. All CSF samples were collected through lumbar puncture before surgery. Integrated diagnosis was made by analysis of histology and tumor DNA molecular pathology according to the 2021 WHO classification of the central nervous system tumors. Results: A total of 24 patients had detectable circulating tumor DNA (ctDNA) and 22 had at least one somatic mutation or chromosome alteration in CSF. The ctDNA levels varied significantly across different ages, Ki-67 index, magnetic resonance imaging signal and glioma subtypes (p < 0.05). The concordance between integrated ctDNA diagnosis and the final diagnosis came up to 91.6% (Kappa, 0.800). We reclassified the clinical diagnosis of 3 patients based on the results of CSF ctDNA sequencing, and 4 patients were reassessed depending on tumor DNA. Interestingly, a rare IDH1 R132C was identified in CSF ctDNA, but not in the corresponding tumor sample. Conclusion: This study demonstrates a high concordance between integrated ctDNA diagnosis and the final diagnosis of gliomas, highlighting the practicability of NGS based detection of mutations of CSF in assisting integrated diagnosis of gliomas, especially glioblastoma. C1 [Wang, Qiang] Nanjing University, School of Medicine, Jinling Hospital, Department of NeurosurgeryNanjing, China. [Liang, Qiujin] Jiangsu Simcere Diagnostics Co., Ltd., State Key Laboratory of Neurology and Oncology Drug DevelopmentNanjing, China. [Wei, Wuting] Nanjing University, School of Medicine, Jinling Hospital, Department of NeurosurgeryNanjing, China. [Niu, Wenhao] Nanjing University, School of Medicine, Jinling Hospital, Department of NeurosurgeryNanjing, China. [Liang, Chong] Nanjing University, School of Medicine, Jinling Hospital, Department of NeurosurgeryNanjing, China. [Wang, Xiaoliang] Nanjing University, School of Medicine, Jinling Hospital, Department of NeurosurgeryNanjing, China. [Wang, Xiaoxuan] Jiangsu Simcere Diagnostics Co., Ltd., State Key Laboratory of Neurology and Oncology Drug DevelopmentNanjing, China. [Pan, Hao] Nanjing University, School of Medicine, Jinling Hospital, Department of NeurosurgeryNanjing, China. RP Pan, H (reprint author), Nanjing University, School of Medicine, Jinling Hospital, Department of Neurosurgery, Nanjing, China. EM Panhao_nz@163.com CR Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, Barnholtz-Sloan JS. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2013-2017. Neuro Oncol, 2020, 22:iv1–iv96., DOI 10.1093/neuonc/noaa200 Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World health organization classification of tumors of the central nervous system: a summary. Acta Neuropathol, 2016, 131:803–20., DOI 10.1007/s00401-016-1545-1 Stupp R, Brada M, van den Bent MJ, Tonn JC, Pentheroudakis G, ESMO Guidelines Working Group. High-grade glioma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol, 2014, 25(3):iii93–101., DOI 10. 1093/annonc/mdu050 Alix-Panabieres C, Pantel K. Liquid biopsy: from discovery to clinical application. Cancer Discov, 2021, 11:858–73., DOI 10.1158/2159-8290.CD-20-1311 Heitzer E, Ulz P, Geigl JB. Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem, 2015, 61:112–23., DOI 10.1373/clinchem.2014.222679 Keller L, Belloum Y, Wikman H, Pantel K. Clinical relevance of blood-based ctDNA analysis: mutation detection and beyond. Br J Cancer, 2021, 124:345–58., DOI 10.1038/s41416-020-01047-5 Daneman R, Prat A. The blood-brain barrier. Cold Spring Harb Perspect Biol, 2015, 7:a020412., DOI 10.1101/cshperspect.a020412 Rincon-Torroella J, Khela H, Bettegowda A, Bettegowda C. Biomarkers and focused ultrasound: the future of liquid biopsy for brain tumor patients. J Neurooncol, 2022, 156:33–48., DOI 10.1007/s11060-021-03837-0 De Mattos-Arruda L, Mayor R, Ng CKY, Weigelt B, Martinez-Ricarte F, Torrejon D, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun, 2015, 6:8839., DOI 10.1038/ncomms9839 Miller AM, Shah RH, Pentsova EI, PourmalekiM, Briggs S, Distefano N, et al. Tracking tumour evolution in glioma through liquid biopsies of cerebrospinal fluid. Nature, 2019, 565:654–8., DOI 10.1038/s41586-019-0882-3 Wang Y, Springer S, Zhang M, McMahon KW, Kinde I, Dobbyn L, et al. Detection of tumor-derived DNA in cerebrospinal fluid of patients with primary tumors of the brain and spinal cord. Proc Natl Acad Sci U S A, 2015, 112:9704–9., DOI 10.1073/pnas.1511694112 Wang Y, Wang L, Blumcke I, Zhang W, Fu Y, Shan Y, et al. Integrated genotype-phenotype analysis of long-term epilepsy-associated ganglioglioma. Brain Pathol, 2022, 32(1):e13011., DOI 10.1111/bpa.13011 Brat DJ, Aldape K, Colman H, Holland EC, Louis DN, Jenkins RB, et al. cIMPACT-NOW update 3: recommended diagnostic criteria for "Diffuse astrocytic glioma, IDH-wildtype, with molecular features of glioblastoma, WHO grade IV". Acta Neuropathol, 2018, 136:805–10., DOI 10.1007/s00401- 018-1913-0 Brat DJ, Aldape K, Colman H, Figrarella-Branger D, Fuller GN, Giannini C, et al. cIMPACT-NOW update 5: recommended grading criteria and terminologies for IDH-mutant astrocytomas. Acta Neuropathol, 2020, 139:603–8., DOI 10.1007/ s00401-020-02127-9 Louis DN, Wesseling P, Aldape K, Brat DJ, Capper D, Cree IA, et al. cIMPACT-NOW update 6: new entity and diagnostic principle recommendations of the cIMPACT-Utrecht meeting on future CNS tumor classification and grading. Brain Pathol, 2020, 30:844–56., DOI 10.1111/bpa.12832 Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, et al. An integrated genomic analysis of human glioblastoma multiforme. Science, 2008, 321: 1807–12., DOI 10.1126/science.1164382 Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics, 1977, 33:159–74., DOI 10.2307/2529310 Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol, 2021, 23:1231–51., DOI 10.1093/neuonc/noab106 Pentsova EI, Shah RH, Tang J, Boire A, You D, Briggs S, et al. Evaluating cancer of the central nervous system through next-generation sequencing of cerebrospinal fluid. J Clin Oncol, 2016, 34:2404–15., DOI 10.1200/JCO.2016.66. 6487 Miller AM, Szalontay L, Bouvier N, Hill K, Ahmad H, Rafailov J, et al. Nextgeneration sequencing of cerebrospinal fluid for clinical molecular diagnostics in pediatric, adolescent and young adult brain tumor patients. Neuro Oncol, 2022, 24: 1763–72., DOI 10.1093/neuonc/noac035 Saenz-Antonanzas A, Auzmendi-Iriarte J, Carrasco-Garcia E, Moreno- Cugnon L, Ruiz I, Villanua J, et al. Liquid biopsy in glioblastoma: opportunities, applications and challenges. Cancers, Basel,, 2019, 11(7):950., DOI 10.3390/ cancers11070950 Fujioka Y, Hata N, Akagi Y, Kuga D, Hatae R, Sangatsuda Y, et al. Molecular diagnosis of diffuse glioma using a chip-based digital PCR system to analyze IDH, TERT, and H3 mutations in the cerebrospinal fluid. J Neurooncol, 2021, 152:47–54., DOI 10.1007/s11060-020-03682-7 Guan YF, Li GR, Wang RJ, Yi YT, Yang L, Jiang D, et al. Application of nextgeneration sequencing in clinical oncology to advance personalized treatment of cancer. Chin J Cancer, 2012, 31(10):463–70., DOI 10.5732/cjc.012.10216 Piccioni DE, Achrol AS, Kiedrowski LA, Banks KC, Boucher N, Barkhoudarian G, et al. Analysis of cell-free circulating tumor DNA in 419 patients with glioblastoma and other primary brain tumors. CNS Oncol, 2019, 8:CNS34., DOI 10.2217/cns-2018-0015 Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med, 2014, 6:224ra24., DOI 10.1126/scitranslmed.3007094 Diaz LA, Jr, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol, 2014, 32:579–86., DOI 10.1200/JCO.2012.45.2011 Bryzgunova OE, Konoshenko MY, Laktionov PP. Concentration of cell-free DNA in different tumor types. Expert Rev Mol Diagn, 2021, 21:63–75., DOI 10.1080/ 14737159.2020.1860021 Gao Y, Zhang K, Xi H, Cai A, Wu X, Cui J, et al. Diagnostic and prognostic value of circulating tumor DNA in gastric cancer: a meta-analysis. Oncotarget, 2017, 8:6330–40., DOI 10.18632/oncotarget.14064 Scholzen T, Gerdes J. The ki-67 protein: from the known and the unknown. J Cell Physiol, 2000, 182:311–22., DOI 10.1002/(SICI)1097-4652(200003)182:3<311: AID-JCP1>3.0.CO;2-9 Xu H, Chen L, Shao Y, Zhu D, Zhi X, Zhang Q, et al. Clinical application of circulating tumor DNA in the genetic analysis of patients with advanced GIST. Mol Cancer Ther, 2018, 17:290–6., DOI 10.1158/1535-7163.MCT-17-0436 Torre M, Vasudevaraja V, Serrano J, DeLorenzo M, Malinowski S, Blandin AF, et al. Molecular and clinicopathologic features of gliomas harboring NTRK fusions. Acta Neuropathol Commun, 2020, 8:107., DOI 10.1186/s40478-020- 00980-z Pekmezci M, Rice T, Molinaro AM, Walsh KM, Decker PA, Hansen H, et al. Adult infiltrating gliomas with WHO 2016 integrated diagnosis: additional prognostic roles of ATRX and TERT. Acta Neuropathol, 2017, 133:1001–16., DOI 10.1007/s00401-017-1690-1 Sturm D, Witt H, Hovestadt V, Khuong-Quang DA, Jones DT, Konermann C, et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell, 2012, 22:425–37., DOI 10.1016/ j.ccr.2012.08.024 Kang Y, Lin X, Kang D. Diagnostic value of circulating tumor DNA in molecular characterization of glioma: a meta-analysis. Medicine, Baltimore,, 2020, 99:e21196., DOI 10.1097/MD.0000000000021196 Weber S, Spiegl B, Perakis SO, Ulz CM, Abuja PM, Kashofer K, et al. Technical evaluation of commercial mutation analysis platforms and reference materials for liquid biopsy profiling. Cancers, Basel,, 2020, 12:1588., DOI 10.3390/ cancers12061588 Gerlinger M, Rowan AJ, Horswell S, Math M, Larkin J, Endesfelder D, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med, 2012, 366:883–92., DOI 10.1056/NEJMoa1113205 McGranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell, 2015, 27:15–26., DOI 10.1016/j.ccell. 2014.12.001 Peng M, Chen C, Hulbert A, Brock MV, Yu F. Non-blood circulating tumor DNA detection in cancer. Oncotarget, 2017, 8:69162–73., DOI 10.18632/oncotarget. 19942 Molenaar RJ, Wilmink JW. IDH1/2 mutations in cancer stem cells and their implications for differentiation therapy. J Histochem Cytochem, 2022, 70:83–97., DOI 10.1369/00221554211062499 Tesileanu CMS, Vallentgoed WR, Sanson M, Taal W, Clement PM, Wick W, et al. Non-IDH1-R132H IDH1/2 mutations are associated with increased DNA methylation and improved survival in astrocytomas, compared to IDH1-R132H mutations. Acta Neuropathol, 2021, 141:945–57., DOI 10.1007/s00401-021-02291-6 Rohle D, Popovici-Muller J, Palaskas N, Turcan S, Grommes C, Campos C, et al. An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science, 2013, 340:626–30., DOI 10.1126/ science.1236062 Avellaneda Matteo D, Grunseth AJ, Gonzalez ER, Anselmo SL, Kennedy MA, Moman P, et al. Molecular mechanisms of isocitrate dehydrogenase 1, IDH1, mutations identified in tumors: the role of size and hydrophobicity at residue 132 on catalytic efficiency. J Biol Chem, 2017, 292:7971–83., DOI 10. 1074/jbc.M117.776179 Stichel D, Ebrahimi A, Reuss D, Schrimpf D, Ono T, Shirahata M, et al. Distribution of EGFR amplification, combined chromosome 7 gain and chromosome 10 loss, and TERT promoter mutation in brain tumors and their potential for the reclassification of IDHwt astrocytoma to glioblastoma. Acta Neuropathol, 2018, 136:793–803., DOI 10.1007/s00401-018-1905-0 Weller M, Weber RG, Willscher E, Riehmer V, Hentschel B, Kreuz M, et al. Molecular classification of diffuse cerebral WHO grade II/III gliomas using genome- and transcriptome-wide profiling improves stratification of prognostically distinct patient groups. Acta Neuropathol, 2015, 129:679–93., DOI 10.1007/s00401-015-1409-0 Zhao Z, Zhang C, Li M, Shen Y, Feng S, Liu J, et al. Applications of cerebrospinal fluid circulating tumor DNA in the diagnosis of gliomas. Jpn J Clin Oncol, 2020, 50(3):325–32., DOI 10.1093/jjco/hyz156 Martinez-Ricarte F, Mayor R, Martinez-Saez E, Rubio-Perez C, Pineda E, Cordero E, et al. Molecular diagnosis of diffuse gliomas through sequencing of cellfree circulating tumor DNA from cerebrospinal fluid. Clin Cancer Res, 2018, 24(12):2812–9., DOI 10.1158/1078-0432.CCR-17-3800 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611391 EP 1611401 DI 10.3389/pore.2023.1611391 PG 11 ER PT J AU Klemm, E Nowak, A AF Klemm, Eckart Nowak, Andreas TI Commentary: Percutaneous tracheostomy: comparison of three different methods with respect to tracheal cartilage injury in cadavers—randomized controlled study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Letter DE tracheostomy; tracheal ring fracture; histology; tracheal stenosis; intensive care unit ID tracheostomy; tracheal ring fracture; histology; tracheal stenosis; intensive care unit C1 [Klemm, Eckart] Stadtisches Klinikum Dresden, Department of Otorhinolaryngology, Head and Neck Surgery, Plastic SurgeryDresden, Germany. [Nowak, Andreas] Stadtisches Klinikum Dresden, Department of Anesthesiology and Intensive Care Medicine, Emergency Medicine and Pain ManagementDresden, Germany. RP Nowak, A (reprint author), Stadtisches Klinikum Dresden, Department of Anesthesiology and Intensive Care Medicine, Emergency Medicine and Pain Management, Dresden, Germany. EM andreas.nowak@klinikum-dresden.de CR Bodis F, Orosz G, Toth JT, Szabo M, Elo LG, Gal J, et al. Percutaneous tracheostomy: comparison of three different methods with respect to tracheal cartilage injury in cadavers-randomized controlled study. Pathol Oncol Res, 2023, 29:1610934., DOI 10.3389/pore.2023.1610934 Klemm E, Nowak A, Haroske G. Histomorphologische Befunde der 2. und Trachealspange von Tracheotomiepatienten der Intensivmedizin. GMS Curr Posters Otorhinolaryngol Head Neck Surg, 2011, 7:Doc43, DOI 10.3205/ cpo000632 3. Nowak A, Kern P, Koscielny S, Usichenko T, Hahnenkamp K, et al. Feasibility and safety of dilatational tracheotomy using the rigid endoscope: a multicenter study. BMC Anesthesiol, 2017, 17:7., DOI 10.1186/s12871-017- 0301-y NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2023 VL 29 IS 3 BP 1611451 EP 1611452 DI 10.3389/pore.2023.1611451 PG 2 ER PT J AU Mai, Y Feng, L Liu, Z Nie, Y Jiang, Z Qin, J AF Mai, Yanxing Feng, Lei Liu, Zhenxi Nie, Yu Jiang, Zesheng Qin, Jiasheng TI Urachus adenocarcinoma mistaken for umbilical incision implant cancer after laparoscopic cholecystectomy: a case report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE urachus adenocarcinoma; laparoscopic cholecystectomy; umbilical incision implant cancer; invasive adenocarcinoma; case report ID urachus adenocarcinoma; laparoscopic cholecystectomy; umbilical incision implant cancer; invasive adenocarcinoma; case report AB Umbilical incision implant cancer after LC is rare. Elective cholecystectomy was planned for a 49 years-old female patient with symptomatic gallstones. The patient underwent transumbilical single-port LC after admission to our hospital. Gallbladder specimens were obtained directly through the umbilical puncture hole, and histopathology suggested chronic cholecystitis. Three months after surgery, the patient experienced painful induration in the umbilicus. We initially considered incision scar hyperplasia complicated with pain, and used drugs to treat it conservatively without taking special treatment measures. Six months after LC, the umbilical induration pain affected her quality of life, and the patient requested surgical resection. Preoperative ultrasonography and abdominal computerized tomography (CT) revealed nodular changes around the umbilicus and no abdominal mass. Local resection of the periumbilical mass was performed, and the pathological confirmation was invasive adenocarcinoma. Subsequently, the patient underwent repeat periumbilical mass enlargement resection. Postoperative pathology showed no cancer at the enlarged resection margin, yet the umbilical center pathology showed invasive adenocarcinoma. The excised pathology was sent to the Sun Yat-sen University Cancer Center for consultation because of the rare nature of the findings associated with the case. After consultation, a diagnosis of umbilical urachus adenocarcinoma was confirmed based on pathological morphology, immunohistochemistry, and the specific anatomical location of the tumor. This case report shown that when there is a persistent mass induration in the navel after LC surgery, the possibility of incision tumor should be considered, rather than simply excluding the possibility of a cancer based on a non-cancer medical history. C1 [Mai, Yanxing] Southern Medical University, Zhujiang Hospital, Department of GeriatricsGuangzhou, China. [Feng, Lei] Southern Medical University, Zhujiang Hospital, Department of Hepatobiliary Surgery II, General Surgery Center, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative MedicineGuangzhou, China. [Liu, Zhenxi] Southern Medical University, Zhujiang Hospital, Department of PathologyGuangzhou, China. [Nie, Yu] Southern Medical University, Zhujiang Hospital, Department of Hepatobiliary Surgery II, General Surgery Center, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative MedicineGuangzhou, China. [Jiang, Zesheng] Southern Medical University, Zhujiang Hospital, Department of Hepatobiliary Surgery II, General Surgery Center, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative MedicineGuangzhou, China. [Qin, Jiasheng] Southern Medical University, Zhujiang Hospital, Department of Hepatobiliary Surgery II, General Surgery Center, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative MedicineGuangzhou, China. RP Qin, J (reprint author), Southern Medical University, Zhujiang Hospital, Department of Hepatobiliary Surgery II, General Surgery Center, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Guangzhou, China. EM 66707647@qq.com CR Otsuka I. Cutaneous metastasis after surgery, injury, lymphadenopathy, and peritonitis: possible mechanisms. Int J Mol Sci, 2019, 20(13):3286., DOI 10.3390/ ijms20133286 Gural Z, Yucel S, Oskeroglu S, Agaoglu F. Urachal adenocarcinoma: a case report and review of the literature. J Cancer Res Ther, 2022, 18(1):291–3., DOI 10. 4103/jcrt.JCRT_28_20 Chen YH, Tseng JS. Concurrent urachal abscess and florid cystitis glandularis masquerading as malignancy: a case report and literature review. BMC Surg, 2022, 22(1):105., DOI 10.1186/s12893-021-01430-w Das JP, Vargas HA, Ghafoor S, Goh AC, Ulaner GA. Clinical utility of 18F-fdg PET/CT for staging and treatment planning in urachal adenocarcinoma. J NuclMed, 2021, 62(5):643–7., DOI 10.2967/jnumed.120.251561 Drouard F, Delamarre J, Capron JP. Cutaneous seeding of gallbladder cancer after laparoscopic cholecystectomy. N Engl J Med, 1991, 325(18):1316., DOI 10.1056/ NEJM199110313251816 Piekarski JH, Kusinska R, Nejc D, Pluta P, Sek P, Bilski A, et al. Recurrence of cholangiogenous carcinoma in port-sites two years after laparoscopic removal of noncancerous gallbladder. Eur J Gastroenterol Hepatol, 2008, 20(5):474–7., DOI 10. 1097/MEG.0b013e3282f16421 Z’Graggen K, Birrer S, Maurer CA, Wehrli H, Klaiber C, Baer HU. Incidence of port site recurrence after laparoscopic cholecystectomy for preoperatively unsuspected gallbladder carcinoma. Surgery, 1998, 124(5):831–8. Winston CB, Chen JW, Fong Y, Schwartz LH, Panicek DM. Recurrent gallbladder carcinoma along laparoscopic cholecystectomy port tracks: CT demonstration. Radiology, 1999, 212(2):439–44., DOI 10.1148/radiology.212.2. r99au17439 Echelard P, Roy SF, Trinh VQ, Garant MP, Collin Y, Nguyen BN, et al. Age, operation time and surgical approach can be used to detect incidental gallbladder carcinoma in cholecystectomy specimens from low-incidence settings. Histopathology, 2021, 79(4):667–73., DOI 10.1111/his.14423 Adsay V, Saka B, Basturk O, Roa JC. Criteria for pathologic sampling of gallbladder specimens. Am J Clin Pathol, 2013, 140(2):278–80., DOI 10.1309/ AJCPUJPGQIZ6DC6A Bruins HM, Visser O, PloegM, Hulsbergen-van de Kaa CA, Kiemeney LALM, Witjes JA. The clinical epidemiology of urachal carcinoma: results of a large, population based study. J Urol, 2012, 188(4):1102–7., DOI 10.1016/j.juro.2012. 06.020 Zhou YW, Long YX, Song K, Liang LB, Cheng K, Chen Y, et al. Anterior wall adenocarcinoma of bladder with similar clinicopathological and prognostic characteristics as common bladder carcinomas should not be treated as or classified into urachal adenocarcinomas. Cancer Med, 2021, 10(16):5415–28., DOI 10.1002/cam4.4053 Krysiewicz S. Diagnosis of urachal carcinoma by computed tomography and magnetic resonance imaging. Clin Imaging, 1990, 14(3):251–4., DOI 10.1016/0899- 7071(90)90085-p Thali-Schwab CM, Woodward PJ, Wagner BJ. Computed tomographic appearance of urachal adenocarcinomas: review of 25 cases. Eur Radiol, 2005, 15(1):79–84., DOI 10.1007/s00330-004-2408-z Das JP,Woo S, Ghafoor S, Andrieu PIC, Ulaner GA, Donahue TF, et al. Value of MRI in evaluating urachal carcinoma: a single center retrospective study. Urol Oncol, 2022, 40(7):345.e9–345345.e17., DOI 10.1016/j.urolonc.2022.02.017 Reis H, Szarvas T. Urachal cancer-current concepts of a rare cancer. Pathologe, 2019, 40(1):31–9., DOI 10.1007/s00292-018-0516-9 Dhillon J, Liang Y, Kamat AM, Siefker-Radtke A, Dinney CP, Czerniak B, et al. Urachal carcinoma: a pathologic and clinical study of 46 cases. Hum Pathol, 2015, 46(12):1808–14., DOI 10.1016/j.humpath.2015.07.021 Behrendt MA, van Rhijn BW. Genetics and biological markers in urachal cancer. Transl Androl Urol, 2016, 5(5):655–61., DOI 10.21037/tau.2016.04.01 Reis H, Krafft U, Niedworok C, Modos O, Herold T, Behrendt M, et al. Biomarkers in urachal cancer and adenocarcinomas in the bladder: a comprehensive review supplemented by own data. Dis Markers, 2018, 2018: 7308168., DOI 10.1155/2018/7308168 Petryshyn N, Drazic T, Hogendorf P, Strzelczyk J, Strzalka A, Szwedziak K, et al. Port site metastases a year after initial laparoscopic cholecystectomy. Should the use of retrieval bags during laparoscopic cholecystectomy be the new gold standard? Pol Przegl Chir, 2021, 93(6):61–5., DOI 10.5604/01.3001.0015.3280 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611334 EP 1611341 DI 10.3389/pore.2023.1611334 PG 8 ER PT J AU Zhang, C Jia, Y Kong, Q AF Zhang, Caixin Jia, Yong Kong, Qingnuan TI Case report: Squamous cell carcinoma of the prostate-a clinicopathological and genomic sequencing-based investigation SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE squamous cell carcinoma; prostate adenocarcinoma; whole exome sequencing; gene mutation; SCC ID squamous cell carcinoma; prostate adenocarcinoma; whole exome sequencing; gene mutation; SCC AB Squamous differentiation of prostate cancer, which accounts for less than 1% of all cases, is typically associated with androgen deprivation treatment (ADT) or radiotherapy. This entity is aggressive and exhibits poor prognosis due to limited response to traditional treatment. However, the underlying molecular mechanisms and etiology are not fully understood. Previous findings suggest that squamous cell differentiation may potentially arise from prostate adenocarcinoma (AC), but further validation is required to confirm this hypothesis. This paper presents a case of advanced prostate cancer with a combined histologic pattern, including keratinizing SCC and AC. The study utilized whole-exome sequencing (WES) data to analyze both subtypes and identified a significant overlap in driver gene mutations between them. This suggests that the two components shared a common origin of clones. These findings emphasize the importance of personalized clinical management for prostate SCC, and specific molecular findings can help optimize treatment strategies. C1 [Zhang, Caixin] Qingdao Municipal Hospital, Department of PathologyQingdao, China. [Jia, Yong] Qingdao Municipal Hospital, Department of UrologyQingdao, China. [Kong, Qingnuan] Qingdao Municipal Hospital, Department of PathologyQingdao, China. RP Kong, Q (reprint author), Qingdao Municipal Hospital, Department of Pathology, Qingdao, China. EM nuanyun4621@126.com CR Dizman N, Salgia M, Ali SM, Wu H, Arvanitis L, Chung JH, et al. Squamous transformation of prostate adenocarcinoma: a report of two cases with genomic profiling. ClinGenitourin Cancer, 2020, 18(3):e289–92., DOI 10.1016/j.clgc.2019.11.020 Lau HD, Clark M. Metastatic squamous cell carcinoma transformed from prostatic adenocarcinoma following androgen deprivation therapy: a case report with clinicopathologic and molecular findings. Diagn Cytopathol, 2020, 48(12): E14–E17., DOI 10.1002/dc.24539 Lara PN, Heilmann AM, Elvin JA, Parikh M, de Vere White R, Gandour- Edwards R, et al. TMPRSS2-ERG fusions unexpectedly identified in men initially diagnosed with nonprostatic malignancies. JCO Precis Oncol, 2017, 2017:1–6., DOI 10.1200/PO.17.00065 Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, et al. Mutational landscape and significance across 12 major cancer types. Nature, 2013, 502(7471): 333–9., DOI 10.1038/nature12634 Tamborero D, Gonzalez-Perez A, Perez-Llamas C, Deu-Pons J, Kandoth C, Reimand J, et al. Comprehensive identification of mutational cancer driver genes across 12 tumor types. Sci Rep, 2013, 3:2650., DOI 10.1038/srep02650 Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA, Kinzler KW. Cancer genome landscapes. Science, 2013, 339(6127):1546–58., DOI 10.1126/science. 1235122 Vlachostergios PJ, Puca L, Beltran H. Emerging variants of castration-resistant prostate cancer. Curr Oncol Rep, 2017, 19(5):32., DOI 10.1007/s11912-017-0593-6 Mohan H, Bal A, Punia RP, Bawa AS. Squamous cell carcinoma of the prostate. Int J Urol, 2003, 10(2):114–6., DOI 10.1046/j.1442-2042.2003.00580.x Adams EJ, Karthaus WR, Hoover E, Liu D, Gruet A, Zhang Z, et al. FOXA1 mutations alter pioneering activity, differentiation and prostate cancer phenotypes. Nature, 2019, 571(7765):408–12., DOI 10.1038/s41586-019-1318-9 Teng M, Zhou S, Cai C, Lupien M, He HH. Pioneer of prostate cancer: past, present and the future of FOXA1. Protein Cell, 2021, 12(1):29–38., DOI 10.1007/ s13238-020-00786-8 Parolia A, Cieslik M, Chu SC, Xiao L, Ouchi T, Zhang Y, et al. Distinct structural classes of activating FOXA1 alterations in advanced prostate cancer. Nature, 2019, 571(7765):413–8., DOI 10.1038/s41586-019-1347-4 Bednarz N, Eltze E, Semjonow A, Rink M, Andreas A, Mulder L, et al. BRCA1 loss preexisting in small subpopulations of prostate cancer is associated with advanced disease and metastatic spread to lymph nodes and peripheral blood. Clin Cancer Res, 2010, 16(13):3340–8., DOI 10.1158/1078-0432.CCR-10-0150 Shah S, Rachmat R, Enyioma S, Ghose A, Revythis A, Boussios S. BRCA mutations in prostate cancer: assessment, implications and treatment considerations. Int J Mol Sci, 2021, 22(23):12628., DOI 10.3390/ijms222312628 Kaur E, Agrawal R, Sengupta S. Functions of BLM helicase in cells: is it acting like a double-edged sword? Front Genet, 2021, 12:634789., DOI 10.3389/fgene.2021. 634789 Guo Y, Xu H, Huang M, Ruan Y. BLM promotes malignancy in PCa by inducing KRAS expression and RhoA suppression via its interaction with HDGF and activation of MAPK/ERK pathway. J Cell Commun Signal, 2022, 17:757–72., DOI 10.1007/s12079-022-00717-8 Ledet EM, Antonarakis ES, Isaacs WB, Lotan TL, Pritchard C, Sartor AO. Germline BLM mutations and metastatic prostate cancer. Prostate, 2020, 80(2): 235–7., DOI 10.1002/pros.23924 Wang Y, Cortez D, Yazdi P, Neff N, Elledge SJ, Qin J. BASC, a super complex of BRCA1-associated proteins involved in the recognition and repair of aberrant DNA structures. Genes Dev, 2000, 14(8):927–39., DOI 10.1101/gad.14.8.927 Mackay RP, Xu Q, Weinberger PM. R-loop physiology and pathology: a brief review. DNA Cell Biol, 2020, 39(11):1914–25., DOI 10.1089/dna.2020. 5906 Hatchi E, Skourti-Stathaki K, Ventz S, Pinello L, Yen A, Kamieniarz-Gdula K, et al. BRCA1 recruitment to transcriptional pause sites is required for R-loop-driven DNA damage repair. Mol Cell, 2015, 57(4):636–47., DOI 10.1016/j.molcel.2015. 01.011 Groh M, Gromak N. Out of balance: R-loops in human disease. PLoS Genet, 2014, 10(9):e1004630., DOI 10.1371/journal.pgen.1004630 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal, 2013, 6(269):pl1., DOI 10.1126/scisignal.2004088 Crossley MP, Song C, BocekMJ, Choi JH, Kousorous J, Sathirachinda A, et al. R-loop-derived cytoplasmic RNA-DNA hybrids activate an immune response. Nature, 2023, 613(7942):187–94., DOI 10.1038/s41586-022-05545-9 Cho HJ, Lee S, Ji YG, Lee DH. Association of specific gene mutations derived from machine learning with survival in lung adenocarcinoma. PLoS One, 2018, 13(11):e0207204., DOI 10.1371/journal.pone.0207204 Biswas T, Podder T, Lepera PA, Walker P. Primary squamous cell carcinoma of the prostate: a case report of a rare clinical entity. Future Sci OA, 2015, 1(3): FSO18., DOI 10.4155/fso.15.16 Fine SW. Variants and unusual patterns of prostate cancer: clinicopathologic and differential diagnostic considerations. Adv Anat Pathol, 2012, 19(4):204–16., DOI 10.1097/PAP.0b013e31825c6b92 Wu X, Li J, Yan T, Ke X, Li X, Zhu Y, et al. HOXB7 acts as an oncogenic biomarker in head and neck squamous cell carcinoma. Cancer Cell Int, 2021, 21(1): 393., DOI 10.1186/s12935-021-02093-6 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611343 EP 1611348 DI 10.3389/pore.2023.1611343 PG 6 ER PT J AU Almasi, Sz Nagy, Krenacs, T Lantos, T Zombori, T Cserni, G AF Almasi, Szintia Nagy, Agnes Krenacs, Tibor Lantos, Tamas Zombori, Tamas Cserni, Gabor TI The prognostic value of stem cell markers in triple-negative breast cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE immunohistochemistry; prognosis; CD44; ALDH1; stem cell markers; triple-negative breast cancer ID immunohistochemistry; prognosis; CD44; ALDH1; stem cell markers; triple-negative breast cancer AB Among the many consecutive theories of cancer, the stem cell theory is currently the most accepted one. Cancer stem cells are located in small niches with specific environment, renew themselves and are believed to be responsible for many recurrences. They can be highlighted with stem cell markers, but often these markers also label tumor cells, and this may represent a phenotypical change associated with prognosis. In this study, we attempted to match tumor outcomes with the expression of the following stem cell markers: ALDH1, AnnexinA1, CD44, CD117, CD166, Nanog and oct-4. Tissue microarray blocks from triple-negative breast cancers were immunostained for the listed markers, and their expression by the majority of tumor cells (diffuse positivity) was correlated with prognosis. Of the 106 tumors investigated, diffuse positivity was seen in 7 (ALDH1), 33 (AnnexinA1), 53 (CD44), 44 (CD117 membranous only), 49 (CD117), 72 (CD166), 19 (Nanog), and 11 (oct-4) cases. With a median follow-up of 83 months, ALDH1 and CD117 expression was associated with DFS, whereas CD44, CD117 and CD166 were associated with OS estimates, based on Kaplan- Meier analyses. In the multivariate Cox proportional hazard models (including the examined markers and clinicopathological data which had a statistical impact in the univariate analysis), the pN category and the lack of ALDH1 expression were independent prognosticators for DFS, and the pN category and diffuse CD44 staining were independent prognosticators for OS. In the multivariate analysis including all of the examined clinicopathological data and markers, only CD117 showed a statistical impact on OS. Wefailed to demonstrate a prognostic impact for most stem cell markers tested in triple-negative breast cancer, but lack of ALDH1 staining and CD44 expression appears as of prognostic value, requiring further examination in independent studies. C1 [Almasi, Szintia] University of Szeged, Department of PathologySzeged, Hungary. [Nagy, Agnes] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Lantos, Tamas] University of Szeged, Department of Medical InformaticsSzeged, Hungary. [Zombori, Tamas] University of Szeged, Department of PathologySzeged, Hungary. [Cserni, Gabor] University of Szeged, Department of PathologySzeged, Hungary. RP Almasi, Sz (reprint author), University of Szeged, Department of Pathology, Szeged, Hungary. EM szinti951023@gmail.com CR Alvarez-Elizondo MB, Weihs D. Breast cancer stem cells: mechanobiology reveals highly invasive cancer cell subpopulations. Cell Mol Life Sci, 2022, 79(3): 134., DOI 10.1007/s00018-022-04181-w Ma I, Allan AL. The role of human aldehyde dehydrogenase in normal and cancer stem cells. Stem Cel Rev Rep, 2011, 7(2):292–306., DOI 10.1007/s12015-010- 9208-4 Demir H, Dulgar O, Gulle BT, Turna H, Ilvan S. Prognostic value of aldehyde dehydrogenase 1, ALDH1, in invasive breast carcinomas. Bosn J Basic Med Sci, 2018, 18(4):313–9., DOI 10.17305/bjbms.2018.3094 Shao G, Zhou H, Zhang Q, Jin Y, Fu C. Advancements of Annexin A1 in inflammation and tumorigenesis. OncoTargets Ther, 2019, 12:3245–54., DOI 10. 2147/ott.s202271 Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, et al. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res, 2004, 10(16):5367–74., DOI 10.1158/ 1078-0432.CCR-04-0220 Cheang MC, Voduc D, Bajdik C, Leung S, McKinney S, Chia SK, et al. Basal-like breast cancer defined by five biomarkers has superior prognostic value than triplenegative phenotype. Clin Cancer Res, 2008, 14(5):1368–76., DOI 10.1158/1078-0432. CCR-07-1658 Basakran NS. CD44 as a potential diagnostic tumormarker. Saudi Med J, 2015, 36(3):273–9., DOI 10.15537/smj.2015.3.9622 Xu H, Niu M, Yuan X, Wu K, Liu A. CD44 as a tumor biomarker and therapeutic target. Exp Hematol Oncol, 2020, 9(1):36., DOI 10.1186/s40164-020- 00192-0 Shan NL, Shin Y, Yang G, Furmanski P, Suh N. Breast cancer stem cells: a review of their characteristics and the agents that affect them. Mol Carcinog, 2021, 60(2):73–100., DOI 10.1002/mc.23277 Kim J, Villadsen R. Expression of luminal progenitor marker CD117 in the human breast gland. J Histochem Cytochem, 2018, 66(12):879–88., DOI 10.1369/ 0022155418788845 Foster BM, Zaidi D, Young TR, Mobley ME, Kerr BA. CD117/c-kit in cancer stem cell-mediated progression and therapeutic resistance. Biomedicines, 2018, 6(1):31., DOI 10.3390/biomedicines6010031 Huang R, Wu D, Yuan Y, Li X, Holm R, Trope CG, et al. CD117 expression in fibroblasts-like stromal cells indicates unfavorable clinical outcomes in ovarian carcinoma patients. PLoS One, 2014, 9(11):e112209., DOI 10.1371/journal.pone.0112209 Ferragut F, Vachetta VS, Troncoso MF, Rabinovich GA, Elola MT. ALCAM/ CD166: a pleiotropic mediator of cell adhesion, stemness and cancer progression. Cytokine Growth Factor Rev, 2021, 61:27–37., DOI 10.1016/j.cytogfr.2021.07.001 Jeter CR, Yang T, Wang J, Chao HP, Tang DG. Concise review: NANOG in cancer stem cells and tumor development: an update and outstanding questions. Stem Cells, 2015, 33(8):2381–90., DOI 10.1002/stem.2007 Grubelnik G, Bostjancic E, Pavlic A, Kos M, Zidar N. NANOG expression in human development and cancerogenesis. Exp Biol Med, Maywood,, 2020, 245(5): 456–64., DOI 10.1177/1535370220905560 Vasefifar P, Motafakkerazad R, Maleki LA, Najafi S, Ghrobaninezhad F, Najafzadeh B, et al. Nanog, as a key cancer stem cell marker in tumor progression. Gene, 2022, 15:146448., DOI 10.1016/j.gene.2022.146448 Mohiuddin IS, Wei SJ, Kang MH. Role of OCT4 in cancer stem-like cells and chemotherapy resistance. Biochim Biophys Acta Mol Basis Dis, 2020, 1866(4): 165432., DOI 10.1016/j.bbadis.2019.03.005 Tai MH, Chang CC, Kiupel M, Webster JD, Olson LK, Trosko JE. Oct4 expression in adult human stem cells: evidence in support of the stem cell theory of carcinogenesis. Carcinogenesis, 2005, 26(2):495–502., DOI 10.1093/carcin/ bgh321 Wang LP, Bi J, Yao C, Xu XD, Li XX,Wang SM, et al. Annexin A1 expression and its prognostic significance in human breast cancer. Neoplasma, 2010, 57(3): 253–9., DOI 10.4149/neo_2010_03_253 Nagata T, Shimada Y, Sekine S, Hori R, Matsui K, Okumura T, et al. Prognostic significance of NANOG and KLF4 for breast cancer. Breast Cancer, 2014, 21(1):96–101., DOI 10.1007/s12282-012-0357-y Wang D, Lu P, Zhang H, Luo M, Zhang X, Wei X, et al. Oct-4 and Nanog promote the epithelial-mesenchymal transition of breast cancer stem cells and are associated with poor prognosis in breast cancer patients. Oncotarget, 2014, 5(21): 10803–15., DOI 10.18632/oncotarget.2506 Luo Y, Huang W, Zhang H, Liu G. Prognostic significance of CD117 expression and TP53 missense mutations in triple-negative breast cancer. Oncol Lett, 2018, 15(5):6161–70., DOI 10.3892/ol.2018.8104 Tsutsui S, Yasuda K, Suzuki K, Takeuchi H, Nishizaki T, Higashi H, et al. A loss of c-kit expression is associated with an advanced stage and poor prognosis in breast cancer. Br J Cancer, 2006, 94(12):1874–8., DOI 10.1038/sj.bjc.6603183 Hein S, Muller V, Wikman H, Krenkel S, Streichert T, Schweizer M, et al. Biologic role of activated leukocyte cell adhesion molecule overexpression in breast cancer cell lines and clinical tumor tissue. Breast Cancer Res Treat, 2011, 129: 347–60., DOI 10.1007/s10549-010-1219-y Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, et al. ALDH1 is a marker of normal andmalignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell, 2007, 1(5):555–67., DOI 10.1016/ j.stem.2007.08.014 Zhou L, Li K, Luo Y, Tian L, Wang M, Li C, et al. Novel prognosticmarkers for patients with triple-negative breast cancer. Hum Pathol, 2013, 44(10):2180–7., DOI 10.1016/j.humpath.2013.03.021 Kida K, Ishikawa T, Yamada A, Shimada K, Narui K, Sugae S, et al. Effect of ALDH1 on prognosis and chemoresistance by breast cancer subtype. Breast Cancer Res Treat, 2016, 156(2):261–9., DOI 10.1007/s10549-016-3738-7 Ma F, Li H, Li Y, Ding X, Wang H, Fan Y, et al. Aldehyde dehydrogenase 1, ALDH1, expression is an independent prognostic factor in triple negative breast cancer, TNBC). Medicine, Baltimore,, 2017, 96(14):6561., DOI 10.1097/MD. 0000000000006561 Kim HJ, Kim MJ, Ahn SH, Son BH, Kim SB, Ahn JH, et al. Different prognostic significance of CD24 and CD44 expression in breast cancer according to hormone receptor status. Breast, 2011, 20(1):78–85., DOI 10.1016/j.breast.2010. 08.001 Collina F, Di Bonito M, Li Bergolis V, De Laurentiis M, Vitagliano C, Cerrone M, et al. Prognostic value of cancer stem cells markers in triple-negative breast cancer. Biomed Res Int, 2015, 2015:158682., DOI 10.1155/2015/158682 Abdoli Shadbad M, Hosseinkhani N, Asadzadeh Z, Derakhshani A, Karim Ahangar N, Hemmat N, et al. A systematic review to clarify the prognostic values of CD44 and CD44+CD24- phenotype in triple-negative breast cancer patients: lessons learned and the road ahead. Front Oncol, 2021, 11:689839., DOI 10.3389/ fonc.2021.689839 Louderbough JM, Schroeder JA. Understanding the dual nature of CD44 in breast cancer progression. Mol Cancer Res, 2011, 9(12):1573–86., DOI 10.1158/1541- 7786.MCR-11-0156 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611365 EP 1611377 DI 10.3389/pore.2023.1611365 PG 13 ER PT J AU Tornyi, I Arkosy, P Horvath, I Furka, A AF Tornyi, Ilona Arkosy, Peter Horvath, Ildiko Furka, Andrea TI A new perspective on the proper timing of radiotherapy during CDK4/6 inhibitor therapy in patients with “bone-only” metastatic breast cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE breast cancer; radiotherapy; radiosensitivity; CDK4/6 inhibitors; treatment timing ID breast cancer; radiotherapy; radiosensitivity; CDK4/6 inhibitors; treatment timing AB The vast majority of hormone positive and HER2 negative advanced breast cancers can be controlled well by endocrine therapy combined with the groundbreaking use of CDK4/6 inhibitors in the metastatic first-line setting. Approximately 50%–60% of these patients have “bone-only” metastatic disease. In oligometastatic cases or if a certain number of uncontrolled lesions develop during the aforementioned therapy, ablative radiotherapy can be delivered or, in symptomatic cases, urgent irradiation is needed with palliative intent. To achieve the most effective results, parallel with good quality of life, the timing of radiotherapy must be determined precisely, taking into account that different cell cycles are involved during different treatment modalities; therefore, optimization of treatment schedules ensures longer and safer post-progression overall survival. The key question is whether the two treatment modalities are safe concurrently or whether they should be administered separately, and if so, what is the optimal sequence and why? This manuscript aims to answer this important question, with a focus on quality of life. Existing publications focus on safety and toxicity profiles, and efficacy is detailed only tangentially and minimally. C1 [Tornyi, Ilona] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Arkosy, Peter] University of Debrecen, Department of OncologyDebrecen, Hungary. [Horvath, Ildiko] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Furka, Andrea] University of Debrecen, Department of OncologyDebrecen, Hungary. RP Horvath, I (reprint author), University of Debrecen Medical and Health Science Center, Department of Pulmonary Medicine, Debrecen, Hungary. EM ildiko.horvath@koranyi.hu CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Ca: Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Howlader N, Altekruse SF, Li CI, Chen VW, Clarke CA, Ries LA, et al. US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status. Jnci: J Natl Cancer Inst, 2014, 106(5):dju055–8., DOI 10.1093/jnci/ dju055 Bosacki C, BouleftourW, Sotton S, Vallard A, Daguenet E, Ouaz H, et al. CDK 4/6 inhibitors combined with radiotherapy: a review of literature. Clin Transl Radiat Oncol, 2021, 26:79–85., DOI 10.1016/j.ctro.2020.11.010 Feng Y, Spezia M, Huang S, Yuan C, Zeng Z, Zhang L, et al. Breast cancer development and progression: risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis, 2018, 5:77–106., DOI 10.1016/j. gendis.2018.05.001 Wu Q, Li J, Zhu S, Wu J, Chen C, Liu Q, et al. Breast cancer subtypes predict the preferential site of distant metastases: a SEER based study. Oncotarget, 2017, 8: 27990–6., DOI 10.18632/oncotarget.15856 Svensson E, Christiansen CF, Ulrichsen SP, RorthSorensen MRHT, Sorensen HT. Survival after bone metastasis by primary cancer type: a Danish populationbased cohort study. BMJ open, 2017, 7:e016022–7., DOI 10.1136/bmjopen-2017- 016022 Cristofanilli M, Turner NC, Bondarenko I, Ro J, Im SA, Masuda N, et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy, PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol, 2016, 17:425–39., DOI 10.1016/S1470-2045(15)00613-0 Tripathy D, ImSA, ColleoniM, Franke F, Bardia A, Harbeck N, et al. Ribociclib plus endocrine therapy for premenopausal women with hormone-receptorpositive, advanced breast cancer, MONALEESA-7): a randomised phase 3 trial. Lancet Oncol, 2018, 19:904–15., DOI 10.1016/S1470-2045(18)30292-4 Johnston S, Martin M, Di Leo A, Im SA, Awada A, Forrester T, et al. MONARCH 3 final PFS: a randomized study of abemaciclib as initial therapy for advanced breast cancer. NPJ Breast Cancer, 2019, 5:5–8., DOI 10.1038/s41523- 018-0097-z Johnston SR, Harbeck N, Hegg R, Toi M, Martin M, Shao ZM, et al. Abemaciclib combined with endocrine therapy for the adjuvant treatment of HR+, HER2−, node-positive, high-risk, early breast cancer, monarchE). J Clin Oncol, 2020, 38:3987–98., DOI 10.1200/JCO.20.02514 Xu B, Zhang Q, Zhang P, Hu X, Li W, Tong Z, et al. Dalpiciclib or placebo plus fulvestrant in hormone receptor-positive and HER2-negative advanced breast cancer: a randomized, phase 3 trial. Nat Med, 2021, 27:1904–9., DOI 10.1038/ s41591-021-01562-9 Yin L, Yao Z, Wang Y, Huang YH, Mazuranic M, Yin A. 4MO Preclinical evaluation of novel CDK4/6 inhibitor GLR2007 in breast and lung cancer models. Ann Oncol, 2021, 32:S362–362., DOI 10.1016/j.annonc.2021.08.282 Braal CL, Jongbloed EM, Wilting SM, Mathijssen RH, Koolen SL, Jager A. Inhibiting CDK4/6 in breast cancer with palbociclib, ribociclib, and abemaciclib: similarities and differences. Drugs, 2021, 81:317–31., DOI 10.1007/s40265-020- 61-2 14. Parkes A, Clifton K, Al-Awadhi A, Oke O, Warneke CL, Litton JK, et al. Characterization of bone only metastasis patients with respect to tumor subtypes. NPJ Breast Cancer, 2018, 4:2–7., DOI 10.1038/s41523-018-0054-x Petroni G, Galluzzi L. Impact of treatment schedule on the efficacy of cytostatic and immunostimulatory agents. Oncoimmunology, 2021, 10: 1889101–4., DOI 10.1080/2162402X.2021.1889101 Petroni G, Buque A, Yamazaki T, Bloy N, Liberto MD, Chen-Kiang S, et al. Radiotherapy delivered before CDK4/6 inhibitors mediates superior therapeutic effects in ER+ breast cancer. Clin Cancer Res, 2021, 27:1855–63., DOI 10.1158/1078- 0432.CCR-20-3871 Yang Y, Luo J, Chen X, Yang Z, Mei X, Ma J, et al. CDK4/6 inhibitors: a novel strategy for tumor radiosensitization. J Exp Clin Cancer Res, 2020, 39:188–11., DOI 10.1186/s13046-020-01693-w Ades F, Tryfonidis K, Zardavas D. The past and future of breast cancer treatment—from the papyrus to individualised treatment approaches. Ecancermedicalscience, 2017, 11:746–11., DOI 10.3332/ecancer.2017.746 Vozenin MC, Baumann M, Coppes RP, Bourhis J. FLASH radiotherapy international workshop. Radiother Oncol, 2019, 139:1–3., DOI 10.1016/j.radonc. 2019.07.020 Yokoya A, Shikazono N, Fujii K, Urushibara A, Akamatsu K, Watanabe R. DNA damage induced by the direct effect of radiation. Radiat Phys Chem, 2008, 77: 1280–5., DOI 10.1016/j.radphyschem.2008.05.021 O’neill P, Fielden EM. Primary free radical processes in DNA. Adv Radiat Biol, 1993, 17:53–120., DOI 10.1016/B978-0-12-035417-7.50005-2 Abraham RT. Checkpoint signalling: focusing on 53BP1. Nat Cel Biol, 2002, 4:E277–9., DOI 10.1038/ncb1202-e277 Lin Y, Raj J, Li J, Ha A, Hossain MA, Richardson C, et al. APE1 senses DNA single-strand breaks for repair and signaling. Nucleic Acids Res, 2020, 48:1925–40., DOI 10.1093/nar/gkz1175 Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018. Cell Death Differ, 2018, 25:486–541., DOI 10.1038/ s41418-017-0012-4 Kumari S, Mukherjee S, Sinha D, Abdisalaam S, Krishnan S, Asaithamby A. Immunomodulatory effects of radiotherapy. Int J Mol Sci, 2020, 21:8151–29., DOI 10. 3390/ijms21218151 Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development, 2013, 140:3079–93., DOI 10.1242/dev.091744 Pesch AM, Hirsh NH, Michmerhuizen AR, Jungles KM, Wilder-Romans K, Chandler BC, et al. RB expression confers sensitivity to CDK4/6 inhibitor–mediated radiosensitization across breast cancer subtypes. JCI Insight, 2022, 7:e154402–18., DOI 10.1172/jci.insight.154402 Filmus J, Robles AI, Shi W, Wong MJ, Colombo LL, Conti CJ. Induction of cyclin D1 overexpression by activated ras. Oncogene, 1994, 9:3627–33. Tanyi J, Tory K, Bankfalvi A, Shroder W, Rath W, Fuzesi L. Analysis of p53 mutation and cyclin D1 expression in breast tumors. Pathol Oncol Res, 1999, 5(2):90–4., DOI 10.1053/paor.1999.0201 Asghar U, Witkiewicz AK, Turner NC, Knudsen ES. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov, 2015, 14:130–46., DOI 10.1038/nrd4504 Pesch AM, Hirsh NH, Chandler BC, Michmerhuizen AR, Ritter CL, Androsiglio MP, et al. Short-term CDK4/6 inhibition radiosensitizes estrogen receptor-positive breast cancers. Clin Cancer Res, 2020, 26:6568–80., DOI 10. 1158/1078-0432.CCR-20-2269 Klapp V, Buque A, Bloy N, Sato A, Yamazaki T, Zhou XK, et al. Cellular senescence in the response of HR+ breast cancer to radiotherapy and CDK4/ 6 inhibitors. J Transl Med, 2023, 21:110–0., DOI 10.1186/s12967-023-03964-4 Alomran R, White M, Bruce M, Bressel M, Roache S, Karroum L, et al. Stereotactic radiotherapy for oligoprogressive ER-positive breast cancer, AVATAR). BMC Cancer, 2021, 21:303–9., DOI 10.1186/s12885-021-08042-w Meattini I, Desideri I, Scotti V, Simontacchi G, Livi L. Ribociclib plus letrozole and concomitant palliative radiotherapy for metastatic breast cancer. Breast, 2018, 42:1–2., DOI 10.1016/j.breast.2018.08.096 David S, Ho G, Day D, Harris M, Tan J, Goel S, et al. Enhanced toxicity with CDK 4/6 inhibitors and palliative radiotherapy: non-consecutive case series and review of the literature. Transl Oncol, 2021, 14:100939–7., DOI 10.1016/j.tranon. 2020.100939 Kubeczko M, Gabrys D, Gawkowska M, Polakiewicz-Gilowska A, Cortez AJ, Krzywon A, et al. Safety and feasibility of radiation therapy combined with CDK 4/ 6 inhibitors in the management of advanced breast cancer. Cancers, 2023, 15: 690–17., DOI 10.3390/cancers15030690 Harbeck N, Franke F, Villanueva-Vazquez R, Lu YS, Tripathy D, Chow L, et al. Health-related quality of life in premenopausal women with hormone-receptorpositive, HER2-negative advanced breast cancer treated with ribociclib plus endocrine therapy: results from a phase III randomized clinical trial, MONALEESA-7). Ther Adv Med Oncol, 2020, 12:1758835920943065–8., DOI 10. 1177/1758835920943065 Dickler MN, Tolaney SM, Rugo HS, Cortes J, Dieras V, Patt D, et al. MONARCH 1, A phase II study of abemaciclib, a CDK4 and CDK6 inhibitor, as a single agent, in patients with refractory hr+/HER2- metastatic breast cancer. Clin Cancer Res, 2017, 23:5218–24., DOI 10.1158/1078-0432.CCR-17-0754 Sledge GW, Jr, Toi M, Neven P, Sohn J, Inoue K, Pivot X, et al. Monarch 2: abemaciclib in combination with fulvestrant in women with HR+/HER2− advanced breast cancer who had progressed while receiving endocrine therapy. J Clin Oncol, 2017, 35:2875–84., DOI 10.1200/JCO.2017.73.7585 Goetz MP, Toi M, Campone M, Sohn J, Paluch-Shimon S, Huober J, et al. Monarch 3: abemaciclib as initial therapy for advanced breast cancer. J Clin Oncol, 2017, 35:3638–46., DOI 10.1200/JCO.2017.75.6155 Finn RS, Crown JP, Lang I, Boer K, Bondarenko IM, Kulyk SO, et al. The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2- negative, advanced breast cancer, PALOMA-1/TRIO-18): a randomised phase 2 study. Lancet Oncol, 2015, 16:25–35., DOI 10.1016/S1470-2045(14)71159-3 Finn RS,Martin M, Rugo HS, Jones S, Im SA, Gelmon K, et al. Palbociclib and letrozole in advanced breast cancer. N Engl J Med, 2016, 375:1925–36., DOI 10.1056/ NEJMoa1607303 Lelliott EJ, Kong IY, Zethoven M, Ramsbottom KM, Martelotto LG, Meyran D, et al. CDK4/6 inhibition promotes antitumor immunity through the induction of T-cell memory. Cancer Discov, 2021, 11:2582–601., DOI 10.1158/2159-8290.CD-20-1554 Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer, 2020, 8:e000337–21., DOI 10.1136/ jitc-2019-000337 Gottgens EL, Bussink J, Leszczynska KB, Peters H, Span PN, Hammond EM. Inhibition of CDK4/CDK6 enhances radiosensitivity of HPV negative head and neck squamous cell carcinomas. Int J Radiat Oncol Biol Phys, 2019, 105:548–58., DOI 10.1016/j.ijrobp.2019.06.2531 Xie X, Zheng W, Chen T, Lin W, Liao Z, Liu J, et al. CDK4/6 inhibitor palbociclib amplifies the radiosensitivity to nasopharyngeal carcinoma cells via mediating apoptosis and suppressing DNA damage repair. Onco Targets Ther, 2019, 12:11107–17., DOI 10.2147/OTT.S234221 Al-Rashdan A, Quirk S, Roumeliotis M, Abedin T, Amaro CP, Barbera L, et al. Radiation therapy with cyclin-dependent kinase 4/6 inhibitors: a multi-institutional safety and toxicity study. Int J Radiat Oncol Biol Phys, 2022, 114:399–408., DOI 10. 1016/j.ijrobp.2022.07.005 Rubovszky G, Kocsis J, Boer K, Chilingirova N, Dank M, Kahan Z, et al. Systemic treatment of breast cancer. 1st central-eastern European professional consensus statement on breast cancer. Pathol Oncol Res, 2022, 28:1–26., DOI 10. 3389/pore.2022.1610383 Helm A, Fournier C, Durante M. Particle radiotherapy and molecular therapies: mechanisms and strategies towards clinical applications. Expert Rev Mol Med, 2022, 24:e8–11., DOI 10.1017/erm.2022.2 Nikitaki Z, Velalopoulou A, Zanni V, Tremi I, Havaki S, Kokkoris M, et al. Key biological mechanisms involved in high-LET radiation therapies with a focus on DNA damage and repair. Expert Rev Mol Med, 2022, 24:e15–15., DOI 10.1017/erm.2022.6 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611369 EP 1611376 DI 10.3389/pore.2023.1611369 PG 8 ER PT J AU Cegledi, A Csukly, Z Fekete, M Kozma, A Szemlaky, Zs Andrikovics, H Mikala, G AF Cegledi, Andrea Csukly, Zoltan Fekete, Monika Kozma, Andras Szemlaky, Zsuzsanna Andrikovics, Hajnalka Mikala, Gabor TI Effective venetoclax-based treatment in relapsed/refractory multiple myeloma patients with translocation t(6;14) SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE multiple myeloma; venetoclax; translocation t(6; 14); IGH::CCND3; personalized therapy ID multiple myeloma; venetoclax; translocation t(6; 14); IGH::CCND3; personalized therapy AB Introduction: The selective Bcl-2 inhibitor venetoclax has shown promising therapeutic potential in multiple myeloma, particularly in cases associated with t(11;14) IGH::CCND1 translocation. However, the efficacy of venetoclax in myeloma patients with the t(6;14) IGH::CCND3 translocation remains less investigated. Methods: In this study, we conducted a retrospective analysis to investigate the efficacy of venetoclax-based therapy in relapsed/refractory myeloma patients with t(6;14) translocation. The treatment courses of three patients, that included previous therapies and responses to venetoclax, were assessed. Clinical data, laboratory results, and adverse events were analyzed to evaluate treatment outcomes. Results: Our findings demonstrated remarkable therapeutic responses in three consecutive patients with t(6;14) translocation-associated myeloma who received venetoclax-based therapy. Patient 1, a lenalidomide-bortezomibdaratumumab and alkylator treatment refractory patient, achieved sustained stringent complete remission (sCR) after combining carfilzomibdexamethasone with venetoclax, which was his best response ever. Similarly, Patient 2, refractory to frontline bortezomib-thalidomide-dexamethasone therapy, attained CR following a transition to bortezomib-dexamethasonvenetoclax treatment. Patient 3, who was immunomodulatory (IMID)- intolerant, showed a highly favorable response to venetoclaxdexamethasone therapy after his first relapse following autologous stem cell transplantation. No significant adverse effects were observed in any of the patients. Discussion: Our study provides compelling preliminary evidence for the efficacy of venetoclax in t(6;14) translocation-associated myeloma. The outcomes observed in our patients suggest that venetoclax-based therapy holds substantial promise as an effective treatment option for this specific genetic subgroup. Furthermore, the similarities in treatment response between t(11;14) and t(6;14) translocation subgroups highlight the importance of personalized approaches targeting specific genetic abnormalities to optimize therapeutic outcomes. C1 [Cegledi, Andrea] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Csukly, Zoltan] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Fekete, Monika] Semmelweis University, Department of Public HealthBudapest, Hungary. [Kozma, Andras] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Molekularis Genetikai LaboratoriumBudapest, Hungary. [Szemlaky, Zsuzsanna] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Andrikovics, Hajnalka] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Molekularis Genetikai LaboratoriumBudapest, Hungary. [Mikala, Gabor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. RP Mikala, G (reprint author), Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Budapest, Hungary. EM gmikala@dpckorhaz.hu CR Urban VS, Cegledi A, Mikala G. Multiple myeloma, a quintessential malignant disease of aging: a geroscience perspective on pathogenesis and treatment. Geroscience, 2023, 45:727–46., DOI 10.1007/s11357-022-00698-x Kumar SK, Harrison SJ, Cavo M, de la Rubia J, Popat R, Gasparetto C, et al. Venetoclax or placebo in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma, BELLINI): a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol, 2020, 21:1630–42., DOI 10. 1016/S1470-2045(20)30525-8 Bahlis NJ, Baz R, Harrison SJ, Quach H, Ho SJ, Vangsted AJ, et al. Phase I study of venetoclax plus daratumumab and dexamethasone, with or without bortezomib, in patients with relapsed or refractory multiple myeloma with and without t(11;14). J Clin Oncol, 2021, 39:3602–12., DOI 10.1200/JCO.21.00443 Kaufman JL, Gasparetto C, Schjesvold FH, Moreau P, Touzeau C, Facon T, et al. Targeting BCL-2 with venetoclax and dexamethasone in patients with relapsed/refractory t(11;14, multiple myeloma. Am J Hematol, 2021, 96:418–27., DOI 10.1002/ajh.26083 Regidor B, GoldwaterMS, Wang J, Bujarski S, Swift R, Eades B, et al. Low dose venetoclax in combination with bortezomib, daratumumab, and dexamethasone for the treatment of relapsed/refractorymultiple myeloma patients-a single-center retrospective study. Ann Hematol, 2021, 100:2061–70., DOI 10.1007/s00277-021-04555-3 Nguyen N, Chaudhry S, Totiger TM, Diaz R, Roberts E, Montoya S, et al. Combination venetoclax and selinexor effective in relapsed refractory multiple myeloma with translocation t(11;14). NPJ Precis Oncol, 2022, 6:73., DOI 10.1038/ s41698-022-00315-2 Abuelgasim KA, Alherz N, Alhejazi A, Damlaj M. Venetoclax in combination with carfilzomib and dexamethasone in relapsed/refractory multiple myeloma harboring t(11,14)(q13;q32): two case reports and a review of the literature. J Med Case Rep, 2020, 14:54., DOI 10.1186/s13256-020-02376-y Basali D, Chakraborty R, Rybicki L, Rosko N, Reed J, Karam M, et al. Realworld data on safety and efficacy of venetoclax-based regimens in relapsed/ refractory t(11;14, multiple myeloma. Br J Haematol, 2020, 189:1136–40., DOI 10.1111/bjh.16454 Boccon-Gibod C, Talbot A, Le Bras F, Frenzel L, Royer B, Harel S, et al. Carfilzomib, venetoclax and dexamethasone for relapsed/refractory multiple myeloma. Br J Haematol, 2020, 189:e73–6., DOI 10.1111/bjh.16483 Costa LJ, Davies FE, Monohan GP, Kovacsovics T, Burwick N, Jakubowiak A, et al. Phase 2 study of venetoclax plus carfilzomib and dexamethasone in patients with relapsed/refractory multiple myeloma. Blood Adv, 2021, 5:3748–59., DOI 10. 1182/bloodadvances.2020004146 Gasparetto C, Bowles KM, Abdallah AO, Morris L, Mander G, Coppola S, et al. A phase II study of venetoclax in combination with pomalidomide and dexamethasone in relapsed/refractory multiple myeloma. Clin Lymphoma Myeloma Leuk, 2021, 21:775–84., DOI 10.1016/j.clml.2021.07.029 HeW,He F,HuH. Efficacy and safety of Venetoclax-based regimens in relapsed or refractory multiple myeloma: a systematic review and meta-analysis of prospective clinical trials. Ann Med, 2023, 55:1029–36., DOI 10.1080/07853890.2023.2186480 Kovacs SB, Luan J, Dold SM,Weis A, Pantic M, Duyster J, et al. Venetoclax in combination with carfilzomib, doxorubicin and dexamethasone restores responsiveness in an otherwise treatment-refractory multiple myeloma patient. Haematologica, 2020, 105:e138–40., DOI 10.3324/haematol.2019.232330 Kumar S, Kaufman JL, Gasparetto C, Mikhael J, Vij R, Pegourie B, et al. Efficacy of venetoclax as targeted therapy for relapsed/refractory t(11;14, multiple myeloma. Blood, 2017, 130:2401–9., DOI 10.1182/blood-2017-06- 788786 Sidiqi MH, Al Saleh AS, Kumar SK, Leung N, Jevremovic D, Muchtar E, et al. Venetoclax for the treatment of multiple myeloma: outcomes outside of clinical trials. Am J Hematol, 2021, 96:1131–6., DOI 10.1002/ajh.26269 Swan D, Delaney C, Natoni A, O’Dwyer M, Krawczyk J. Successful venetoclax salvage in the setting of refractory, dialysis-dependent multiple myeloma with t(11; 14). Haematologica, 2020, 105:e141–3., DOI 10.3324/haematol.2019.228338 Szita VR, Mikala G, Kozma A, Fabian J, Hardi A, Alizadeh H, et al. Targeted venetoclax therapy in t(11;14, multiple myeloma: real world data from seven hungarian centers. Pathol Oncol Res, 2022, 28:1610276., DOI 10.3389/pore.2022. 1610276 Touzeau C, Le Gouill S, Mahe B, Boudreault JS, Gastinne T, Blin N, et al. Deep and sustained response after venetoclax therapy in a patient with very advanced refractory myeloma with translocation t(11;14). Haematologica, 2017, 102:e112–4., DOI 10.3324/haematol.2016.160408 Lee R, Cho SY, Lee DG, Choi H, Park S, Cho BS, et al. Infections of venetoclaxbased chemotherapy in acute myeloid leukemia: rationale for proper antimicrobial prophylaxis. Cancers, Basel,, 2021, 13:6285., DOI 10.3390/cancers13246285 Papayannidis C,Nanni J, CristianoG,Marconi G, Sartor C, Parisi S, et al. Impact of infectious comorbidity and overall time of hospitalization in total outpatient management of acute myeloid leukemia patients following venetoclax and hypomethylating agents. Eur J Haematol, 2022, 108:449–59., DOI 10.1111/ejh.13753 Zhu LX, Chen RR, Wang LL, Sun JN, Zhou D, Li L, et al. A real-world study of infectious complications of venetoclax combined with decitabine or azacitidine in adult acute myeloid leukemia. Support Care Cancer, 2022, 30:7031–8., DOI 10.1007/ s00520-022-07126-y Bhalla S, Melnekoff DT, Aleman A, Leshchenko V, Restrepo P, Keats J, et al. Patient similarity network of newly diagnosed multiple myeloma identifies patient subgroups with distinct genetic features and clinical implications. Sci Adv, 2021, 7: eabg9551., DOI 10.1126/sciadv.abg9551 Shaughnessy JD, Jr, Barlogie B. Using genomics to identify high-risk myeloma after autologous stem cell transplantation. Biol Blood Marrow Transpl, 2006, 12: 77–80., DOI 10.1016/j.bbmt.2005.10.002 Sarin V, Yu K, Ferguson ID, Gugliemini O, Nix MA, Hann B, et al. Evaluating the efficacy of multiple myeloma cell lines as models for patient tumors via transcriptomic correlation analysis. Leukemia, 2020, 34:2754–65., DOI 10.1038/ s41375-020-0785-1 Szalat R, Avet-Loiseau H, Munshi NC. Gene expression profiles in myeloma: ready for the real world? Clin Cancer Res, 2016, 22:5434–42., DOI 10.1158/1078- 0432.CCR-16-0867 Gupta VA, Barwick BG, Matulis SM, Shirasaki R, Jaye DL, Keats JJ, et al. Venetoclax sensitivity in multiple myeloma is associated with B-cell gene expression. Blood, 2021, 137:3604–15., DOI 10.1182/blood.2020007899 Todoerti K, Taiana E, Puccio N, Favasuli V, Lionetti M, Silvestris I, et al. Transcriptomic analysis in multiple myeloma and primary plasma cell leukemia with t(11;14, reveals different expression patterns with biological implications in venetoclax sensitivity. Cancers, Basel,, 2021, 13:4898., DOI 10.3390/ cancers13194898 Bajpai R, Sharma A, Achreja A, Edgar CL, Wei C, Siddiqa AA, et al. Electron transport chain activity is a predictor and target for venetoclax sensitivity in multiple myeloma. Nat Commun, 2020, 11:1228., DOI 10.1038/s41467-020- 15051-z Matulis SM, Gupta VA, Neri P, Bahlis NJ, Maciag P, Leverson JD, et al. Functional profiling of venetoclax sensitivity can predict clinical response in multiple myeloma. Leukemia, 2019, 33:1291–6., DOI 10.1038/s41375-018-0374-8 van Laar R, Flinchum R, Brown N, Ramsey J, Riccitelli S, Heuck C, et al. Translating a gene expression signature for multiple myeloma prognosis into a robust high-throughput assay for clinical use. BMC Med Genomics, 2014, 7:25., DOI 10.1186/1755-8794-7-25 Zhan F, Barlogie B, Mulligan G, Shaughnessy JD, Jr, Bryant B. High-risk myeloma: a gene expression based risk-stratification model for newly diagnosed multiple myeloma treated with high-dose therapy is predictive of outcome in relapsed disease treated with single-agent bortezomib or high-dose dexamethasone. Blood, 2008, 111:968–9., DOI 10.1182/blood-2007-10-119321 Shaughnessy JD, Jr, Zhan F, Burington BE, Huang Y, Colla S, Hanamura I, et al. A validated gene expression model of high-risk multiple myeloma is defined by deregulated expression of genes mapping to chromosome 1. Blood, 2007, 109: 2276–84., DOI 10.1182/blood-2006-07-038430 Shaughnessy J, Jr, Zhan F, Barlogie B, Stewart AK. Gene expression profiling and multiple myeloma. Best Pract Res Clin Haematol, 2005, 18:537–52., DOI 10. 1016/j.beha.2005.02.003 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611375 EP 1611381 DI 10.3389/pore.2023.1611375 PG 7 ER PT J AU Wang, Z Li, P Bai, J Liu, Y Jiao, G AF Wang, Zhen Li, Peng Bai, Jiayu Liu, Yujia Jiao, Guangyu TI Quantitative analysis of endobronchial elastography combined with serum tumour markers of lung cancer in the diagnosis of benign and malignant mediastinal and hilar lymph nodes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE elastography; endobronchial ultrasound-guided transbronchial needle aspiration; mediastinal and hilar lymph nodes; lung cancer; serum tumour markers ID elastography; endobronchial ultrasound-guided transbronchial needle aspiration; mediastinal and hilar lymph nodes; lung cancer; serum tumour markers AB Purpose: In malignant tumours, elastography and serum tumour markers have shown high diagnostic efficacy. Therefore, we aimed to quantitatively analyse the results of endobronchial elastography combined with serum tumour markers of lung cancer to accurately distinguish benign and malignant mediastinal and hilar lymph nodes. Methods: Data of patients who underwent endobronchial ultrasound-guided transbronchial needle aspiration for mediastinal lymph node enlargement in our hospital between January 2018 and August 2022 were retrospectively collected. The characteristics of quantitative elastography and serum tumour markers were evaluated. Results: We enrolled 197 patients (273 lymph nodes). In the differential diagnosis of benign and malignant mediastinal and hilar lymph nodes, the stiffness area ratio (SAR), strain ratio (SR), and strain rate in lymph nodes were significant, among which SAR had the highest diagnostic value (cut-off value, 0.409). The combination of the four tumour markers had a high diagnostic value (AUC, 0.886). Three types of quantitative elastography indices combined with serum tumour markers for lung cancer showed a higher diagnostic value (AUC, 0.930; sensitivity, 83.5%; specificity, 89.3%; positive predictive value, 88.1%; negative predictive value, 85%) (p < 0.05). In the differential diagnosis of pathological types of lung cancer, different quantitative elastography indicators and serum tumour markers for lung cancer have different diagnostic significance for the differential diagnosis of lung cancer pathological types. Conclusion: The quantitative analysis of endobronchial ultrasound elastography combined with tumour markers can improve the diagnosis rate of benign and malignant mediastinal and hilar lymph nodes, help guide the puncture of false negative lymph nodes, and reduce the misdiagnosis rate. C1 [Wang, Zhen] Shengjing Hospital of China Medical University, Department of Respiratory MedicineShenyang, China. [Li, Peng] Shengjing Hospital of China Medical University, Department of Respiratory MedicineShenyang, China. [Bai, Jiayu] The First Affiliated Hospital of China Medical University, Department of RheumatologyShenyang, China. [Liu, Yujia] Liaoning University of Traditional Chinese Medicine, College of Traditional Chinese MedicineShenyang, China. [Jiao, Guangyu] Shengjing Hospital of China Medical University, Department of Respiratory MedicineShenyang, China. RP Jiao, G (reprint author), Shengjing Hospital of China Medical University, Department of Respiratory Medicine, Shenyang, China. EM jiao_gy@163.com CR Mohan A, Naik S, Pandey RM, Mills J, Munavvar M. Diagnostic utility of endobronchial ultrasound guided transbronchial needle aspiration for mediastinal lesions: a prospective three year, single centre analysis. Thorac Cancer, 2011, 2(4): 183–9., DOI 10.1111/j.1759-7714.2011.00063.x Iyer H, Anand A, Sryma PB, Gupta K, Naranje P, Damle N, et al. Mediastinal lymphadenopathy: a practical approach. Expert Rev Respir Med, 2021, 15(10): 1317–34., DOI 10.1080/17476348.2021.1920404 Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin, 2015, 65(2):87–108., DOI 10.3322/ caac.21262 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin, 2017, 67(1):7–30., DOI 10.3322/caac.21387 Pirker R. Conquering lung cancer: current status and prospects for the future. Pulmonology, 2020, 26(5):283–90., DOI 10.1016/j.pulmoe.2020.02.005 Sakairi Y, Nakajima T, Yoshino I. Role of endobronchial ultrasoundguided transbronchial needle aspiration in lung cancer management. Expert Rev Respir Med, 2019, 13(9):863–70., DOI 10.1080/17476348.2019.1646642 Okasha HH, El-Meligui A, Pawlak KM, Zorniak M, Atalla H, Abou- Elmagd A, et al. Practical approach to linear EUS examination of the mediastinum. Endosc Ultrasound, 2021, 10(6):406–13., DOI 10.4103/EUS-D- 21-00019 Xu C, Qin L, Lei W, Jiang J, Ni C, Huang J. The role of endobronchial ultrasound-guided transbronchial needle aspiration liquid-based cytology in the diagnosis of mediastinal lymphadenopathy. Diagn Cytopathol, 2020, 48(4):316–21., DOI 10.1002/dc.24374 Cui XW, Li KN, Yi AJ, Wang B, Wei Q, Wu GG, et al. Ultrasound elastography. Endosc Ultrasound, 2022, 11(4):252–74., DOI 10.4103/EUS-D-21- 00151 Cui XW, Chang JM, Kan QC, Chiorean L, Ignee A, Dietrich CF. Endoscopic ultrasound elastography: current status and future perspectives. World J Gastroenterol, 2015, 21(47):13212–24., DOI 10.3748/wjg.v21.i47.13212 Chen YF, Mao XW, Zhang YJ, Zhang CY, Yu YF, Qin E, et al. Endobronchial ultrasound elastography differentiates intrathoracic lymph nodes: a meta-analysis. Ann Thorac Surg, 2018, 106(4):1251–7., DOI 10. 1016/j.athoracsur.2018.04.003 Izumo T, Sasada S, Chavez C, Matsumoto Y, Tsuchida T. Endobronchial ultrasound elastography in the diagnosis of mediastinal and hilar lymph nodes. Jpn J Clin Oncol, 2014, 44(10):956–62., DOI 10.1093/jjco/hyu105 Rozman A, Malovrh MM, Adamic K, Subic T, Kovac V, Flezar M. Endobronchial ultrasound elastography strain ratio for mediastinal lymph node diagnosis. Radiol Oncol, 2015, 49(4):334–40., DOI 10.1515/raon-2015- 0020 Huang H, Huang Z,Wang Q,Wang X, Dong Y, Zhang W, et al. Effectiveness of the benign and malignant diagnosis of mediastinal and hilar lymph nodes by endobronchial ultrasound elastography. J Cancer, 2017, 8(10):1843–8., DOI 10.7150/ jca.19819 Ma H, An Z, Xia P, Cao J, Gao Q, Ren G, et al. Semi-quantitative analysis of EBUS elastography as a feasible approach in diagnosing mediastinal and hilar lymph nodes of lung cancer patients. Sci Rep, 2018, 8(1):3571., DOI 10.1038/s41598- 018-22006-4 Lin CK, Yu KL, Chang LY, Fan HJ, Wen YF, Ho CC. Differentiating malignant and benign lymph nodes using endobronchial ultrasound elastography. J Formos Med Assoc, 2019, 118(1 Pt 3):436–43., DOI 10.1016/j. jfma.2018.06.021 Uchimura K, Yamasaki K, Sasada S, Hara S, Ikushima I, Chiba Y, et al. Quantitative analysis of endobronchial ultrasound elastography in computed tomography-negative mediastinal and hilar lymph nodes. Thorac Cancer, 2020, 11(9):2590–9., DOI 10.1111/1759-7714.13579 Zhao X, Wang M. Clinical utility of serum tumor markers in lung cancer. Zhongguo Fei Ai Za Zhi, 2011, 14(3):286–91., DOI 10.3779/j.issn.1009-3419.2011. 03.16 Liu L, Teng J, Zhang L, Cong P, Yao Y, Sun G, et al. The combination of the tumor markers suggests the histological diagnosis of lung cancer. Biomed Res Int, 2017, 2017:2013989., DOI 10.1155/2017/2013989 RuschVW,AsamuraH,WatanabeH, Giroux DJ, Rami-Porta R,Goldstraw P, et al. The IASLC lung cancer staging project: a proposal for a new international lymph node map in the forthcoming seventh edition of the TNM classification for lung cancer. J Thorac Oncol, 2009, 4(5):568–77., DOI 10.1097/JTO.0b013e3181a0d82e Giovannini M, Hookey LC, Bories E, Pesenti C, Monges G, Delpero JR. Endoscopic ultrasound elastography: the first step towards virtual biopsy? Preliminary results in 49 patients. Endoscopy, 2006, 38(4):344–8., DOI 10.1055/s- 2006-925158 Fujiwara T, Yasufuku K,Nakajima T, Chiyo M, Yoshida S, SuzukiM, et al. The utility of sonographic features during endobronchial ultrasound-guided transbronchial needle aspiration for lymph node staging in patients with lung cancer: a standard endobronchial ultrasound image classification system. Chest, 2010, 138(3):641–7., DOI 10.1378/chest.09-2006 Fujiwara T, Nakajima T, Inage T, Sata Y, Sakairi Y, Tamura H, et al. The combination of endobronchial elastography and sonographic findings during endobronchial ultrasound-guided transbronchial needle aspiration for predicting nodal metastasis. Thorac Cancer, 2019, 10(10):2000–5., DOI 10.1111/1759-7714.13186 Xin L, Gao Y, Wang TJ, Meng QQ, Jin ZD, Fu ZJ, et al. EUS development in China: results from national surveys in 2013 and 2020. Endosc Ultrasound, 2022, 12:90., DOI 10.4103/EUS-D-22-00003 Zhi X, Chen J, Xie F, Sun J, Herth FJF. Diagnostic value of endobronchial ultrasound image features: a specialized review. Endosc Ultrasound, 2021, 10(1): 3–18., DOI 10.4103/eus.eus_43_20 He HY, Chen JL, Ma H, Zhu J, Wu DD, Lv XD. Value of endobronchial ultrasound elastography in diagnosis of central lung lesions. Med Sci Monit, 2017, 23:3269–75., DOI 10.12659/msm.901808 Korrungruang P, Boonsarngsuk V. Diagnostic value of endobronchial ultrasound elastography for the differentiation of benign and malignant intrathoracic lymph nodes. Respirology, 2017, 22(5):972–7., DOI 10.1111/resp.12979 Nakajima T, Inage T, Sata Y, Morimoto J, Tagawa T, Suzuki H, et al. Elastography for predicting and localizing nodal metastases during endobronchial ultrasound. Respiration, 2015, 90(6):499–506., DOI 10.1159/ 000441798 Hao Y, Ren G, Yang W, Zheng W, Wu Y, Li W, et al. Combination diagnosis with elastography strain ratio and molecular markers effectively improves the diagnosis rate of small breast cancer and lymph node metastasis. Quant Imaging Med Surg, 2020, 10(3):678–91., DOI 10.21037/qims.2020.02.14 He HY, Huang M, Zhu J, Ma H, Lyu XD. Endobronchial ultrasound elastography for diagnosing mediastinal and hilar lymph nodes. Chin Med J, Engl,, 2015, 128(20):2720–5., DOI 10.4103/0366-6999.167296 Dietrich CF, Hirche TO, Ott M, Ignee A. Real-time tissue elastography in the diagnosis of autoimmune pancreatitis. Endoscopy, 2009, 41(8):718–20., DOI 10. 1055/s-0029-1214866 Gao S, Zhang G, Lian Y, Yan L, Gao H. Exploration and analysis of the value of tumor-marker joint detection in the pathological type of lung cancer. Cel Mol Biol, Noisy-le-grand,, 2020, 66(6):93–7., DOI 10.14715/cmb/2020.66.6.17 Wu H, Wang Q, Liu Q, Zhang Q, Huang Q, Yu Z. The serum tumor markers in combination for clinical diagnosis of lung cancer. Clin Lab, 2020, 66(3)., DOI 10. 7754/Clin.Lab.2019.190533 Gao J, Zhang L, Peng K, Sun H. Diagnostic value of serum tumor markers CEA, CYFRA21-1, SCCAg, NSE and ProGRP for lung cancers of different pathological types. Nan Fang Yi Ke Da Xue Xue Bao, 2022, 42(6):886–91., DOI 10.12122/j.issn.1673-4254.2022.06.12 Liu E, Bhutani MS, Sun S. Artificial intelligence: the new wave of innovation in EUS. Endosc Ultrasound, 2021, 10(2):79–83., DOI 10.4103/ EUS-D-21-00052 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611377 EP 1611389 DI 10.3389/pore.2023.1611377 PG 13 ER PT J AU Ferenczi, K Nagy, FZs Istenes, I Eid, H Bodor, Cs Timar, B Demeter, J AF Ferenczi, Kata Nagy, Flora Zsofia Istenes, Ildiko Eid, Hanna Bodor, Csaba Timar, Botond Demeter, Judit TI Long term follow-up of refractory/relapsed hairy cell leukaemia patients treated with low-dose vemurafenib between 2013 and 2022 at the Department of Internal Medicine and Oncology, Semmelweis University SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE relapsed/refractory; infection; hairy cell leukemia; BRAF inhibitors; vemurafenib ID relapsed/refractory; infection; hairy cell leukemia; BRAF inhibitors; vemurafenib AB Introduction: Hairy cell leukemia (HCL) is an indolent B-cell lymphoproliferative disease. BRAF V600E mutation is detected in nearly all classical HCL cases which offers the possibility of targeted therapy. Objective: The aim of our study was to assess the efficacy of low-dose vemurafenib as well as to assess the long term outcome of HCL patients treated with this drug at the Department of Internal Medicine and Oncology at Semmelweis University. Methods: We report on 10 patients with classical HCL treated with low-dose vemurafenib at our Department between 2013 and 2022. Results: As a result of fixed time low-dose vemurafenib treatment, 5 of 10 patients (5/10) achieved partial remission, 4 (4/10) had stable disease, and 1 (1/10) had MRD positivity. No patients achieved complete remission. The median progression-free survival was 28.5 months while the overall survival was 82 months. Conclusion: We confirm that low dose of vemurafenib is effective and safe in the vast majority of patients with HCL. This small-molecule oral treatment allows to gain valuable time—months or even years—before further, usually parenteral treatment options have to be given or before previous treatment has to be repeated. There are also promising data supporting the combination of vemurafenib with other drugs for the treatment of HCL patients which could provide even further possibility to bridge treatment. C1 [Ferenczi, Kata] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Nagy, Flora Zsofia] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Istenes, Ildiko] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Eid, Hanna] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Bodor, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Timar, Botond] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Demeter, Judit] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. RP Timar, B (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM timar.botond@semmelweis.hu CR Bouroncle BA, Wiseman BK, Doan CA. Leukemic reticuloendotheliosis. Blood, 1958, 13:164 609–30., DOI 10.1182/blood.v13.7.609.609 Sari E, Rajnai H, Denes K, Bodor C, Csomor J, Korosmezey G, et al. Novelties in the diagnostics and therapy of hairy cell leukemia. Uj utak a hajas sejtes leukemia es a rokon korkepek diagnosztikajaban es kezeleseben. Magy Onkol, 2016, 60:137–44. [Hungarian]. Sari E, Nagy ZG, Baghy K, Rajnai H, Bodor C, Csomor J, et al. Treatment of refractory hairy cell leukemia with a BRAF-inhibitor: lessons to be learnt. Pathol Oncol Res, 2014, 20(4):973–80., DOI 10.1007/s12253-014-9783-9 Toth-Liptak J, Piukovics K, Borbenyi Z, Demeter J, Bagdi E, Krenacs L. A comprehensive immunophenotypic marker analysis of hairy cell leukemia in paraffin-embedded bone marrow trephine biopsies-a tissue microarray study. Pathol Oncol Res, 2015, 21:203–11., DOI 10.1007/s12253-014-9807-5 Tiacci E, Trifonov V, Schiavoni G,Holmes A, Kern W, Martelli MP, et al. BRAF mutations in hairy-cell leukemia. New Engl J Med, 2011, 364(24):2305–15., DOI 10. 1056/NEJMoa1014209 Andrulis M, Penzel R, Weichert W, von Deimling A, Capper D. Application of a BRAF V600E mutation-specific antibody for the diagnosis of hairy cell leukemia. Am J Surg Pathol, 2012, 36(12):1796–800., DOI 10.1097/PAS.0b013e3182549b50 Maitre E, Cornet E, Troussard X. Hairy cell leukemia. 2020 update on diagnosis, risk stratification and treatment. Am J Hematol, 2019, 94:1413–22., DOI 10.1002/ajh.25653 Saven A, Burian C, Koziol JA, Piro LD. Long-term follow-up of patients with hairy cell leukemia after cladribine treatment. Blood, 1998, 92:1918–26., DOI 10. 1182/blood.v92.6.1918.418k33_1918_1926 Dinmohamed AG, Posthuma EFM, Visser O, Kater AP, Raymakers A, Doorduijn JK, et al. Relative survival reaches a plateau in HCL. Blood, 2018, 131:1380–8., DOI 10.1182/blood-2017-12-820381 Thomas Da O, Brian S, Bueso-Ramos C, Fader S, Michael J, Francis J, et al. Rituximab in relapsed or refractory hairy cell leukemia. Blood, 2003, 102:3906–11., DOI 10.1182/blood-2003-02-0630 Ravandi F O, Brian S, Jorgenden J, Pierce S, Faderl S, Ferrajoli A, et al. Phase 2 study of cladribine followed by rituximab in patients with hairy cell leukemia. Blood, 2011, 118:3818–23., DOI 10.1182/blood-2011-04-351502 Robak T, Matutes E, Catovsky D ESMO Guidelines Committee. Hairy cell leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and followup. Ann Oncol, 2015, 5:100–7., DOI 10.1093/annonc/mdv200 Dietrich S, Glimm H, Andrulis M, von Kalle C, Ho AD, Zenz T. BRAF inhibition in refractory hairy-cell leukemia. N Engl J Med, 2012, 366:2038–40., DOI 10.1056/NEJMc1202124 Bohn JP, Wanner D, Steurer M. Ibrutinib for relapsed/refractory hairy cell leukemia variant. Leuk Lymphoma, 2017, 58(5):1224–6., DOI 10.1080/10428194. 2016.1239262 Tiacci E, Park J, De Carolis L, Chung SS, Broccoli A, Scott S, et al. Targeting mutant BRAF in relapsed or refractory hairy-cell leukemia. N Engl J Med, 2015, 373:1733–47., DOI 10.1056/NEJMoa1506583 Dietrich S, Pircher A, Endris V, Peyrade F, Wendtner CM, Follows GA, et al. BRAF inhibition in hairy cell leukemia with low-dose vemurafenib. Blood, 2016, 127(23):2847–55., DOI 10.1182/blood-2015-11-680074 Tiacci E, De Carolis L, Simonetti E, Capponi M, Ambrosetti A, Lucia E, et al. Vemurafenib plus rituximab in refractory or relapsed hairy-cell leukemia. New Engl J Med, 2021, 384(19):1810–23., DOI 10.1056/NEJMoa2031298 Vereertbrugghen A, Colado A, Gargiulo E, Bezares RF, Fernandez Grecco H, Cordini G, et al. In vitro sensitivity to venetoclax and microenvironment protection in hairy cell leukemia. Front Oncol, 2021, 11(11):2595., DOI 10.3389/fonc.2021.598319 Bohn JP, Pircher A, Wanner D, Vill D, Foeger B, Wolf D, et al. Low-dose vemurafenib in hairy cell leukemia patients with active infection. Am J Hematol, 2019, 6:E180–2., DOI 10.1002/ajh.25474 Grever M, Andritsos L, Banerji V, Barrientos JC, Bhat S, Blachly JS, et al. Hairy cell leukemia and COVID-19 adaptation of treatment guidelines. Leukemia, 2021, 35(7):1864–72., DOI 10.1038/s41375-021-01257-7 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611378 EP 1611385 DI 10.3389/pore.2023.1611378 PG 8 ER PT J AU Zhang, F Wang, X Zhu, Y Xia, P AF Zhang, Fang Wang, XiaoShuang Zhu, YuanTing Xia, Peng TI Conjoint analysis of clinical, imaging, and pathological features of schistosomiasis and colorectal cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE schistosomiasis; colorectal cancer; imaging; t stage; pathology ID schistosomiasis; colorectal cancer; imaging; t stage; pathology AB This study aims to examine and compare clinical, radiological, and pathological data between colorectal cancer (CRC) patients with and without schistosomiasis and uncover distinctive CRC characteristics when accompanied by schistosomiasis. This retrospective study is based on data collected from 341 patients diagnosed with CRC post-surgery and pathology. Of these patients, 101 (Group A) were diagnosed with colorectal cancer cooccurring with schistosomiasis (CRC-S), while 240 patients (Group B) were diagnosed with colorectal cancer without concurrent schistosomiasis (CRCNS). Both groups were compared and analyzed based on their clinical data, imaging-based TNM staging, lymph node metastasis, nerve invasion, vascular cancer thrombus, and histopathological differentiation. A Chi-squared test revealed a significant difference in gender distribution between the patients with CRC-S (Group A) and CRC-NS (Group B), with a p -value of 0.043 and χ2 = 4.115. Specifically, a higher incidence rate was observed among males in Group A. There was a difference in the overall distribution of TNM staging between the two groups (p = 0.034, χ2 = 6.764). After pairwise comparison, a statistically significant difference was observed in the T3 stage (p <0.05). The proportion of the T3 stage in Group A was significantly higher than that in Group B, indicating certain advantages. There was a difference in postoperative histopathological grading between the two groups (p = 0.005, χ2 = 10.626). After pairwise comparison, a statistically significant difference was observed between the well-differentiated adenocarcinoma and the moderately and poorly differentiated adenocarcinoma (p <0.05), with a higher proportion of welldifferentiated patients in Group A compared to Group B. There was no significant difference in age, lymph node metastasis, nerve invasion, and vascular invasion between the two groups of patients (p > 0.05). Among the 101 patients with CRC-S, 87 (86%) showed linear calcification on CT imaging. Patients with CRC-S are mainly male, with tumor staging mostly in the middle stage, high tumor differentiation, and low malignancy. CT imaging can help identify the presence of lumps and linear calcification indicative of schistosome deposits. MRI can early clarify TNM staging and determine the presence of lymph node metastasis and nerve and vascular invasion. C1 [Zhang, Fang] Jingzhou Hospital Affiliated to Yangtze University, Department of RadiologyJingzhou, China. [Wang, XiaoShuang] Jingzhou Hospital Affiliated to Yangtze University, Department of RadiologyJingzhou, China. [Zhu, YuanTing] Jingzhou Hospital Affiliated to Yangtze University, Department of PathologyJingzhou, China. [Xia, Peng] Yangtze University, School of Basic Medicine, Health Science Center, Department of ParasitologyJingzhou, China. RP Xia, P (reprint author), Yangtze University, School of Basic Medicine, Health Science Center, Department of Parasitology, Jingzhou, China. EM 805451046@qq.com CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018, 68(6):394–424., DOI 10.3322/ caac.21492 Liu W, Zeng HZ, Wang QM, Yi H, Mou Y, Wu CC, et al. Schistosomiasis combined with colorectal carcinoma diagnosed based on endoscopic findings and clinicopathological characteristics: a report on 32 cases. Asian Pac J Cancer Prev, 2013, 14(8):4839–42., DOI 10.7314/apjcp.2013.14.8.4839 Darre T, Djiwa T, Dare S, Alassani F, Napo-Koura G. Difficult causality relationship between colorectal cancer and schistosomiasis. Pathol Oncol Res, 2020, 26(1):597–8., DOI 10.1007/s12253-018-00566-0 Canepa M, Fanta PT, Weidner N, PetersonMR. Schistosomiasis and signet ring cell carcinoma of the rectum. Ann Diagn Pathol, 2012, 16(5):385–7., DOI 10.1016/j. anndiagpath.2010.12.008 Wang M, Wu QB, He WB, Wang ZQ. Clinicopathological characteristics and prognosis of schistosomal colorectal cancer. Colorectal Dis, 2016, 18(10):1005–9., DOI 10.1111/codi.13317 Vennervald BJ, Polman K. Helminths and malignancy. Parasite Immunol, 2009, 31(11):686–96., DOI 10.1111/j.1365-3024.2009.01163.x Imai J, IchikawaH,Mizukami H, Suzuki T,Watanabe N,Mine T. Colonic highgrade tubular adenomas associated with schistosoma japonicum. Tokai J Exp Clin Med, 2016, 41(1):22–3. Abe T, Yunokizaki H, lijima H, Tamura K, Lee ZL, Higashi D, et al. Schistosoma japonicum showing flat colon polyp Appearance. Gastrointest Endosc, 2011, 73(4):8202–2., DOI 10.1016/j.gie.2010.10.038 Regula J, Rupinski M, Kraszewska E, Polkowski M, Pachlewski J, Orlowska J, et al. Colonoscopy in colorectal-cancer screening for detection of advanced neoplasia. N Engl J Med, 2006, 355(18):1863–72., DOI 10.1056/NEJMoa054967 Wang ZJ, Yang JH. Effect of schistosomiasis japonica on the development of gastric andcolorectal cancer. Zhong guo Xue Xi Chong Bing Fang Zhi Za Zhi, 2020, 32(2):148–53., DOI 10.16250/j.32.1374.2019240 Wu C, WangGao YHL, Zhang Y, Dai M, Li S. Relationship between saltinducible kinase 2, SIK2, and lymph node metastasis in colorectal cancer patients complicated with chronic schistosomiasis. Zhejiang Da Xue Xue Bao Yi Xue Ban, 2021, 50(5):607–13., DOI 10.3724/zdxbyxb-2021-0157 Wang W, Lu K, Wang LM, Jing H, Pan W, Huang S, et al. Comparison of nonschistosomal colorectal cancer and schistosomal colorectal cancer. Comp Study World J Surg Oncol, 2020, 18(1):149., DOI 10.1186/s12957-020-01925-5 Zheng N, WangYu HQ, Wang C, Bai C, Pan A Changing trends, clinicopathological characteristics, surgical treatment patterns, and prognosis of schistosomiasis-associated versus non-schistosomiasis-associated colorectal cancer: a large retrospective cohort study of 31 153 cases in Shanghai, China, 2001-2021). Int J Surg, 2023, 109(4):772–84., DOI 10.1097/js9.0000000000000293 Wang Z, Du Z, Liu Y, Wang W, Liang M, Zhang A, et al. Comparison of the clinicopathological features and prognoses of patients with schistosomal and nonschistosomal colorectal cancer. Oncol Lett, 2020, 19(3):2375–83., DOI 10. 3892/ol.2020.11331 Hamid HKS. Schistosoma japonicum-associated colorectal cancer: a review. Am J Trop Med Hyg, 2019, 100(3):501–5., DOI 10.4269/ajtmh.18-0807 Xiao R, Li F, Zou X. Correlation analysis between chronic schistosomiasis and colorectal cancer. J Clin Surg, 2019, 27(9):779–81. Barsoum RS, Esmat G, Ei-Baz T. Human schistosomiasis: clinical perspective review. J Adv Res, 2013, 4(5):433–44., DOI 10.1016/j.jare.2013.01.005 Qin X, Liu CY, Xiong YL, Bai T, Zhang L, Hou XH, et al. The clinical features of chronic intestinal schistosomiasis-related intestinal lesions. BMC Gastroenterol, 2021, 21(1):12., DOI 10.1186/s12876-020-01591-7 Yosry A. Schistosomiasis and neoplasia. Contrib Microbiol, 2006, 13:81–100., DOI 10.1159/000092967 Chen YB, Liu Z, Qian J, Feng HY, Li DC, Fan YT. Expression difference of DNA mismatch repair gene hMLH1 and hMSH2 between schistomiasis-associated colorectal cancer and sporadic colorectal cancer. Zhonghua Wei Chang Wai Ke Za Zhi, 2016, 19(1):75–9. Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen YJ, Ciombor KK, et al. Rectal cancer, version 2.2018,NCCN clinical practice guidelines in Oncology. J Natl Compr Canc Netw, 2018, 16(7):874–901., DOI 10.6004/jnccn. 2018.0061 Zhang W, Wang PJ, Shen X, Wang GL, Zhao XH, Seema SF, et al. CT presentations of colorectal cancer with chronic schistosomiasis: a comparative study with pathological findings. Eur J Radiol, 2012, 81:835–43., DOI 10.1016/j.ejrad. 2012.02.020 Feng H, Lu AG, Zhao XW, Han DP, Zhao JK, Shi L, et al. Comparison of nonschistosomal rectosigmoid cancer and schistosomal rectosigmoid cancer. World J Gastroenterol, 2015, 21(23):7225–32., DOI 10.3748/wjg.v21.i23.7225 Samdani T, Garcia-Aguilar J. Imaging in rectal cancer: magnetic resonance imaging versus endorectal ultrasonography. Surg Oncol Clin North America, 2014, 23(1):59–77., DOI 10.1016/j.soc.2013.09.011 Cheever AW. Schistosomiasis and colon cancer. Lancet, 1981, 1(8234): 1369–70., DOI 10.1016/s0140-6736(81)92547-2 Van Tong H, Brindley PJ, Meyer CG, Velavan TP. Parasite infection, carcinogenesis and human malignancy. EBioMedicine, 2017, 15:12–23., DOI 10. 1016/j.ebiom.2016.11.034 Wu CY, Du XY, Tang LL,WuJH, ZhaoW, Guo XK, et al. Schistosoma japonicum SJE16.7 protein promotes tumor development via the receptor for advanced glycation end products(RAGE). Front Immunol, 2020, 11:1767., DOI 10.3389/fimmu.2020.01767 Snider AJ, Bialkowska AB, Ghaleb AM, Yang VW, Obeid LM, Hannun YA. Murine model for colitis-associated cancer of the colon. Methods Mol Biol, 2016, 1438:245–54., DOI 10.1007/978-1-4939-3661-8_14 WangM, Zhang YC, Yang XY, Wang ZQ. Prognostic analysis of schistosomal rectal cancer. Astan Pac J Cancer Prev, 2014, 15(21):9271–5., DOI 10.7314/apjcp. 2014.15.21.9271 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611396 EP 1611403 DI 10.3389/pore.2023.1611396 PG 8 ER PT J AU Verma, P Rishi, B George, GN Kushwaha, N Dhandha, H Kaur, M Jain, A Jain, A Chaudhry, S Singh, A Siraj, F Misra, A AF Verma, Priyanka Rishi, Bhavika George, Grace Noreen Kushwaha, Neetu Dhandha, Himanshu Kaur, Manpreet Jain, Ankur Jain, Aditi Chaudhry, Sumita Singh, Amitabh Siraj, Fouzia Misra, Aroonima TI Recent advances and future directions in etiopathogenesis and mechanisms of reactive oxygen species in cancer treatment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE reactive oxygen species (ROS); recent advances; future directions; cancer therapy; ferroptosis ID reactive oxygen species (ROS); recent advances; future directions; cancer therapy; ferroptosis AB A class of exceptionally bioactive molecules known as reactive oxygen species (ROS) have been widely studied in the context of cancer. They play a significant role in the etiopathogenesis for cancer. Implication of ROS in cancer biology is an evolving area, considering the recent advances; insights into their generation, role of genomic and epigenetic regulators for ROS, earlier thought to be a chemical process, with interrelations with cell death pathways- Apoptosis, ferroptosis, necroptosis and autophagy has been explored for newer targets that shift the balance of ROS towards cancer cell death. ROS are signal transducers that induce angiogenesis, invasion, cell migration, and proliferation at low to moderate concentrations and are considered normal by-products of a range of biological activities. Although ROS is known to exist in the oncology domain since time immemorial, its excessive quantities are known to damage organelles, membranes, lipids, proteins, and nucleic acids, resulting in cell death. In the last two decades, numerous studies have demonstrated immunotherapies and other anticancer treatments that modulate ROS levels have promising in vitro and in vivo effects. This review also explores recent targets for therapeutic interventions in cancer that are based on ROS generation or inhibition to disrupt the cell oxidative stress balance. Examples include-metabolic targets, targeted therapy with biomarkers, natural extracts and nutraceuticals and targets developed in the area of nano medicine. In this review, we present the molecular pathways which can be used to create therapy plans that target cancer by regulating ROS levels, particularly current developments and potential prospects for the effective implementation of ROS-mediated therapies in clinical settings. The recent advances in complex interaction with apoptosis especially ferroptosis and its role in epigenomics and modifications are a new paradigm, to just mechanical action of ROS, as highlighted in this review. Their inhibition by nutraceuticals and natural extracts has been a scientific challenging avenue that is explored. Also, the inhibition of generation of ROS by inhibitors, immune modulators and inhibitors of apoptosis and ferroptosis is explored in this review. C1 [Verma, Priyanka] Indian Council of Medical Research-National Institute of Pathology, Department of Health ResearchNew Delhi, India. [Rishi, Bhavika] Indian Council of Medical Research-National Institute of Pathology, Department of Health ResearchNew Delhi, India. [George, Grace Noreen] Indian Council of Medical Research-National Institute of Pathology, Department of Health ResearchNew Delhi, India. [Kushwaha, Neetu] Indian Council of Medical Research-National Institute of Pathology, Department of Health ResearchNew Delhi, India. [Dhandha, Himanshu] Indian Council of Medical Research-National Institute of Pathology, Department of Health ResearchNew Delhi, India. [Kaur, Manpreet] Indian Council of Medical Research-National Institute of Pathology, Department of Health ResearchNew Delhi, India. [Jain, Ankur] Vardhman Mahavir Medical College and Safdarjung HospitalNew Delhi, India. [Jain, Aditi] Vardhman Mahavir Medical College and Safdarjung HospitalNew Delhi, India. [Chaudhry, Sumita] Vardhman Mahavir Medical College and Safdarjung HospitalNew Delhi, India. [Singh, Amitabh] Vardhman Mahavir Medical College and Safdarjung HospitalNew Delhi, India. [Siraj, Fouzia] Indian Council of Medical Research-National Institute of Pathology, Department of Health ResearchNew Delhi, India. [Misra, Aroonima] Indian Council of Medical Research-National Institute of Pathology, Department of Health ResearchNew Delhi, India. RP Misra, A (reprint author), Indian Council of Medical Research-National Institute of Pathology, Department of Health Research, New Delhi, India. EM dr.aroo.2402@gmail.com CR Chhikara Bhupender S, Keykavous P. Global cancer statistics 2022: the trends projection analysis. Chem Biol Lett, 2022, 1:451. Huang R, Chen H, Liang J, Li Y, Yang J, Luo C, et al. Dual role of reactive oxygen species and their application in cancer therapy. J Cancer, 2021, 12(18): 5543–61., DOI 10.7150/jca.54699 Sies H, Jones DP. Reactive oxygen species, ROS, as pleiotropic physiological signalling agents. Nat Rev Mol Cel Biol, 2020, 21(7):363–83., DOI 10.1038/s41580- 020-0230-3 Article R, Takada K. Reactive oxygen species in cancer: current findings and future directions. Cancer Sci, 2021, 2(10):3945–52., DOI 10.1111/cas.15068 Perillo B, Di Donato M, Pezone A, Di Zazzo E, Giovannelli P, Galasso G, et al. ROS in cancer therapy: the bright side of the moon. Exp Mol Med, 2020, 52: 192–203., DOI 10.1038/s12276-020-0384-2 Sullivan LB, Chandel NS. Mitochondrial reactive oxygen species and cancer. Cancer Metab, 2014, 2:17., DOI 10.1186/2049-3002-2-17 Kirtonia A, Sethi G, Garg M, Artesunate ART. The multifaceted role of reactive oxygen species in tumorigenesis. Cell Mol Life Sci, 2020, 77:4459–83., DOI 10.1007/ s00018-020-03536-5 Redza-dutordoir M, Averill-bates DAB Activation of apoptosis signalling pathways by reactive oxygen species. Biochim Biophys Acta, 2016, 1863(12): 2977–92., DOI 10.1016/j.bbamcr.2016.09.012 Wu Q, Ni X. ROS-mediated DNA methylation pattern alterations in carcinogenesis. Curre Drug Targets, 2015, 16(1):13–9., DOI 10.2174/ 1389450116666150113121054 Angireddy R, KazmiHR, Srinivasan S, Sun L, Iqbal J, Fuchs SY, et al. Cytochrome c oxidase dysfunction enhances phagocytic function and osteoclast formation in macrophages. FASEB J, 2019, 10:9167–81., DOI 10.1096/fj.201900010RR Reczek CR, Chandel NS. The two faces of reactive oxygen species in cancer. Annu Rev Cancer Biol, 2017, 1:79–98., DOI 10.1146/annurev-cancerbio-041916-065808 Zhou Y, Feng X, Wang L, Du J, Yu H LGH00031, a novel ortho-quinonoid inhibitor of cell division cycle 25B, inhibits human cancer cells via ROS generation. Acta Pharmacol Sin, 2009, 30(9):1359–68., DOI 10.1038/aps.2009.131 Ramalingam V, Rajaram R. A paradoxical role of reactive oxygen species in cancer signaling pathway: physiology and pathology. Process Biochem, 2021, 100: 69–81., DOI 10.1016/j.procbio.2020.09.032 RenschlerMF. The emerging role of reactive oxygen species in cancer therapy. Eur J Cancer, 2004, 40(13):1934–40., DOI 10.1016/j.ejca.2004.02.031 Prieto-Bermejo R, Romo-Gonzalez M, Perez-Fernandez A, Ijurko C, Hernandez-Hernandez A. Reactive oxygen species in haematopoiesis: leukaemic cells take a walk on the wild side. J Exp Clin Cancer Res, 2018, 37:125., DOI 10.1186/ s13046-018-0797-0 Oliveira MDS, Barbosa MIF, de Souza TB, Moreira DRM, Martins FT, Villarreal W, et al. A novel platinum complex containing a piplartine derivative exhibits enhanced cytotoxicity, causes oxidative stress and triggers apoptotic cell death by ERK/p38 pathway in human acute promyelocytic leukemia HL-60 cells. Redox Biol, 2019, 20:182–94., DOI 10.1016/j.redox.2018.10.006 Hasan A, Rizvi SF, Parveen S, Pathak N, Nazir A, Mir SS. Crosstalk between ROS and autophagy in tumorigenesis: understanding the multifaceted paradox. Front Oncol, 2022, 12:852424., DOI 10.3389/fonc.2022.852424 Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, et al. Ferroptosis: past, present and future. Cell Death Dis, 2020, 11:88., DOI 10.1038/s41419-020-2298-2 Zhang C, Liu X, Jin S, Chen Y, Guo R. Ferroptosis in cancer therapy: a novel approach to reversing drug resistance. Mol Cancer, 2022, 21:47–12., DOI 10.1186/ s12943-022-01530-y YangWS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell, 2015, 156: 317–31., DOI 10.1016/j.cell.2013.12.010 Galaris D, Skiada V, Barbouti A. Redox signaling and cancer: the role of labile iron. Cancer Lett, 2008, 266:21–9., DOI 10.1016/j.canlet.2008.02.038 Zou Y, Graham ET, Deik AA, Eaton JK, Wang W Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol, 2020, 16(3):302–9., DOI 10.1038/s41589-020-0472-6 Liang WEI, He X, Bi J, Hu T, Sun Y. Role of reactive oxygen species in tumors based on the ‘ seed and soil ’ theory: a complex interaction, Review). Oncol Rep, 2021, 46(3):208., DOI 10.3892/or.2021.8159 Didier AJ, Fang L,Watkins D, Dworkin LD, Creeden JF. Antioxidant and antitumor effects of dietary vitamins A, C, and E. Antioxidants, Basel,, 2023, 12(3):632., DOI 10.3390/antiox12030632 Grant WB. Review of recent advances in understanding the role of vitamin D in reducing cancer risk: breast, colorectal, prostate, and overall cancer. Anticancer Res, 2020, 40(1):491–9., DOI 10.21873/anticanres.13977 Yun J, Mullarky E, Lu C, Bosch KN, Kavalier A, Rivera K, et al. Vitamin C selectively kills KRAS and BRAF mutant colorectal cancer cells by targeting GAPDH. Science, 2016, 350:1391–6., DOI 10.1126/science.aaa5004 Liou G, Storz P. Reactive oxygen species in cancer. Free Radic Res, 2010, 44: 479–96., DOI 10.3109/10715761003667554 Xin J, Jiang X, Ben S, Yuan Q, Su L, Zhang Z, et al. Association between circulating vitamin E and ten common cancers: evidence from large - scale Mendelian randomization analysis and a longitudinal cohort study. BMC Med, 2022, 20:168., DOI 10.1186/s12916-022-02366-5 Mclean L, Soto U, Agama K, Francis J, Jimenez R, Pommier Y, et al. Aminoflavone induces oxidative DNA damage and reactive oxidative speciesmediated apoptosis in breast cancer cells. Int J Cancer, 2008, 122(7):1665–74., DOI 10.1002/ijc.23244 Das K, Roychoudhury A, Bhattacharjee H, Deka A, Yambem D, Tahiliani PS, et al. Parasitic diseases of zoonotic importance in humans of northeast India, with special reference to ocular involvement. Front Environ Sci, 2014, 2:1–8., DOI 10. 2147/EB.S64404 Khan N, Mukhtar H. Cancer and metastasis: prevention and treatment by green tea. Cancer Metastasis Rev, 2010, 2010:435–45., DOI 10.1007/s10555-010- 9236-1 Khan A, Chen HC, Wan XX, Tania M, Xu AH, Chen FZ, et al. Regulatory effects of resveratrol on antioxidant enzymes: a mechanism of growth inhibition and apoptosis induction in cancer cells. Mol Cell, 2013, 35:219–25., DOI 10.1007/ s10059-013-2259-z Elumalai P, Ezhilarasan D, Raghunandhakumar S. Molecular targets of nimbolide for anti-cancer therapy: an updated review. J Environ Pathol Toxicol Oncol, 2022, 41:69–88., DOI 10.1615/JEnvironPatholToxicolOncol. 2021040263 Gupta SC, Hevia D, Patchva S, Park B, Koh W, Aggarwal BB. Upsides and downsides of reactive oxygen species for cancer: the roles of reactive oxygen species in tumorigenesis, prevention, and therapy. Antioxid Redox Signal, 2012, 16(11): 1295–322., DOI 10.1089/ars.2011.44.14 Kashyap D, Tuli HS, Varol A, Thakral F, Yerer MB, Sak K, et al. Role of reactive oxygen species in cancer progression: molecular mechanisms and recent advancements. Biomolecules, 2019, 9:735., DOI 10.3390/biom9110735 Hacioglu C, Kar F. Capsaicin induces redox imbalance and ferroptosis through ACSL4/GPx4 signaling pathways in U87-MG and U251 glioblastoma cells. Metab Brain Dis, 2023, 38:393–408., DOI 10.1007/ s11011-022-00983-w Rather RA, Bhagat M. Cancer chemoprevention and piperine: molecular mechanisms and therapeutic opportunities. Front Cel Dev Biol, 2018, 6:10., DOI 10. 3389/fcell.2018.00010 Yuan Z, Liang Z, Chen X, Li R,Wu Y Protective effect of koumine, an alkaloid from Gelsemium sempervirens, on injury induced by H-O-in IPEC-J2 cells. Int J Mol Sci, 2019, 20(3):754., DOI 10.3390/ijms20030754 Rahman A, Hongsprabhas P. Genistein as antioxidant and antibrowning agents in in vivo and in vitro: a review. Biomed Pharmacother, 2016, 82:379–92., DOI 10.1016/j.biopha.2016.05.023 Osman Y, Subramaniam B, Nyamathulla S, Shamsuddin N, Arshad NM, Mun KS, et al. Natural products for cancer therapy: a review of their mechanism of actions and toxicity in the past decade. J Trop Med, 2022, 2022:5794350., DOI 10. 1155/2022/5794350 Zhang C, Wang X, Du J, Gu Z, Zhao Y. Reactive oxygen species-regulating strategies based on nanomaterials for disease treatment. Adv Sci, 2020, 8(3): 2002797., DOI 10.1002/advs.202002797 Li Y, Yang J, Sun X. Reactive oxygen species-based nanomaterials for cancer therapy. Front Chem, 2021, 9:1–12., DOI 10.3389/fchem.2021.650587 Zhang Q, Zhang J, Song J, Liu Y, Ren X, Zhao Y. Protein-based nanomedicine for therapeutic bene fi ts of cancer. ACS Nano, 2021, 5:8001–38., DOI 10.1021/ acsnano.1c00476 Zhang Y, Khalique A, Du X, Gao Z,Wu J, Zhang X, et al. Biomimetic design of mitochondria-targeted hybrid nanozymes as superoxide scavengers. Adv Mater, 2021, 33(9):2006570., DOI 10.1002/adma.202006570 Qin J, Gong N, Liao Z, Zhang S, Timashev P, Huo S, et al. Recent progress in mitochondria-targeting-based nanotechnology for cancer treatment. Nanoscale, 2021, 13:7108–18., DOI 10.1039/d1nr01068a NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611415 EP 1611426 DI 10.3389/pore.2023.1611415 PG 12 ER PT J AU Kisivan, K Farkas, A Kovacs, P Glavak, Cs Lukacs, G Mahr, K Szabo, Zs Csima, PM Gulyban, Toth, Z Kaposztas, Zs Lakosi, F AF Kisivan, Katalin Farkas, Andrea Kovacs, Peter Glavak, Csaba Lukacs, Gabor Mahr, Karoly Szabo, Zsolt Csima, Petone Melinda Gulyban, Akos Toth, Zoltan Kaposztas, Zsolt Lakosi, Ferenc TI Pancreatic SABR using peritumoral fiducials, triggered imaging and breath-hold SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE motion control; pancreatic cancer; triggered imaging; stereotactic ablative radiotherapy; deep inspiration breath hold ID motion control; pancreatic cancer; triggered imaging; stereotactic ablative radiotherapy; deep inspiration breath hold AB Background: We aim to present our linear accelerator-based workflow for pancreatic stereotactic ablative radiotherapy (SABR) in order to address the following issues: intrafractional organ motion management, Cone Beam CT (CBCT) image quality, residual errors with dosimetric consequences, treatment time, and clinical results. Methods: Between 2016 and 2021, 14 patients with locally advanced pancreatic cancer were treated with induction chemotherapy and SABR using volumetric modulated arc therapy (VMAT). Internal target volume (ITV) concept (5), phasegated (4), or breath hold (5) techniques were used. Treatment was verified by CBCT before and after irradiation, while tumor motion was monitored and controlled by kV triggered imaging and beam hold using peritumoral surgical clips. Beam interruptions and treatment time were recorded. The CBCT image quality was scored and supplemented by an agreement analysis (Krippendorff’s- α) of breath-hold CBCT images to determine the position of OARs relative to the planning risk volumes (PRV). Residual errors and their dosimetry impact were also calculated. Progression free (PFS) and overall survival (OS) were assessed by the Kaplan-Meier analysis with acute and late toxicity reporting (CTCAEv4). Results: On average, beams were interrupted once (range: 0–3) per treatment session on triggered imaging. The total median treatment time was 16.7 ± 10.8 min, significantly less for breath-hold vs. phase-gated sessions (18.8 ± 6.2 vs. 26.5 ± 13.4, p < 0.001). The best image quality was achieved by breath hold CBCT. The Krippendorff’s-α test showed a strong agreement among five radiation therapists (mean K-α value: 0.8 (97.5%). The mean residual errors were <0.2 cm in each direction resulting in an average difference of <2% in dosimetry for OAR and target volume. Two patients received offline adaptation. The median OS/PFS after induction chemotherapy and SABR was 20/12 months and 15/8 months. No Gr. ≥2 acute/late RT-related toxicity was noted. Conclusion: Linear accelerator based pancreatic SABR with the combination of CBCT and triggered imaging + beam hold is feasible. Peritumoral fiducials improve utility while breath-hold CBCT provides the best image quality at a reasonable treatment time with offline adaptation possibilities. In well-selected cases, it can be an effective alternative in clinics where CBCT/MRI-guided online adaptive workflow is not available. C1 [Kisivan, Katalin] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. [Farkas, Andrea] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. [Kovacs, Peter] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. [Glavak, Csaba] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. [Lukacs, Gabor] Somogy Megyei Kaposi Mor Oktato Korhaz, Klinikai OnkologiaKaposvar, Hungary. [Mahr, Karoly] Zala Megyei Szent Rafael Korhaz, OnkologiaZalaegerszeg, Hungary. [Szabo, Zsolt] Zala Megyei Szent Rafael Korhaz, OnkologiaZalaegerszeg, Hungary. [Csima, Petone Melinda] Hungarian University of Agricultural and Life Sciences, Institute of EducationGodollo, Hungary. [Gulyban, Akos] Institut Jules Bordet, Department of Medical PhysicsBrussels, Belgium. [Toth, Zoltan] MEDICOPUS Egeszsegugyi Szolgaltato Kozhasznu Nonprofit Kft.Kaposvar, Hungary. [Kaposztas, Zsolt] Somogy Megyei Kaposi Mor Oktato Korhaz, Sebeszeti OsztalyKaposvar, Hungary. [Lakosi, Ferenc] Kaposvar University, Department of Radiation OncologyKaposvar, Hungary. RP Lakosi, F (reprint author), Kaposvar University, Department of Radiation Oncology, Kaposvar, Hungary. EM lakosiferenc@yahoo.com CR Chiorean EG, Chiaro MD, Tempero MA, Malafa MP, Benson AB, Cardin DB, et al. Ampullary adenocarcinoma, version 1.2023, NCCN clinical practice guidelines in oncology. JNCCN J Natl Compr Cancer Netw, 2023, 21(7):753–82., DOI 10.6004/jnccn.2023.0034 Balaban EP, Mangu PB, Yee NS Locally advanced unresectable pancreatic cancer: American society of clinical oncology clinical practice guideline summary. J Oncol Pract, 2017, 13(4):265–9., DOI 10.1200/JOP.2016.017376 Ferlay J, Colombet M, Soerjomataram I, Parkin DM, Pineros M, Znaor A, et al. Cancer statistics for the year 2020: an overview. Int J Cancer, 2021, 149(4):778–89., DOI 10.1002/ijc.33588 Versteijne E, Van Dam JL, Suker M, Janssen QP, Groothuis K, Akkermans- Vogelaar JM, et al. Neoadjuvant chemoradiotherapy versus upfront surgery for resectable and borderline resectable pancreatic cancer: long-term results of the dutch randomized PREOPANC trial. J Clin Oncol, 2022, 40(11):1220–30., DOI 10. 1200/JCO.21.02233 Comito T, Cozzi L, Clerici E, Franzese C, Tozzi A, Iftode C, et al. Can stereotactic body radiation therapy be a viable and efficient therapeutic option for unresectable locally advanced pancreatic adenocarcinoma? results of a phase 2 study. Tech Cancer Res Treat, 2017, 16(3):295–301., DOI 10.1177/1533034616650778 Panje C, Andratschke N, Brunner TB, Niyazi M, Guckenberger M Stereotactic body radiotherapy for renal cell cancer and pancreatic cancer: literature review and practice recommendations of the DEGRO working group on stereotactic radiotherapy. Strahlentherapie Und Onkologie, 2016, 192(12):875–85., DOI 10. 1007/s00066-016-1053-1 Herman JM, Chang DT, Goodman KA, Dholakia AS, Raman SP, Hacker-Prietz A, et al. Phase 2 multi-institutional trial evaluating gemcitabine and stereotactic body radiotherapy for patients with locally advanced unresectable pancreatic adenocarcinoma. Cancer, 2015, 121(7):1128–37., DOI 10.1002/cncr.29161 Petrelli F, Comito T, Ghidini A, Torri V, Scorsetti M, Barni S Stereotactic body radiation therapy for locally advanced pancreatic cancer: a systematic review and pooled analysis of 19 trials. Int J Radiat Oncol Biol Phys, 2017, 97(2):313–22., DOI 10. 1016/j.ijrobp.2016.10.030 Rossi G, Simoni N, Paiella S, Rossi R, Venezia M, Micera R, et al. Risk adapted ablative radiotherapy after intensive chemotherapy for locally advanced pancreatic cancer. Front Oncol, 2021, 11(April):662205–9., DOI 10.3389/fonc.2021.662205 Manderlier M, Navez J, Hein M, EngelholmJ, Closset J, Bali MA, et al. Isotoxic high-dose stereotactic body radiotherapy, iHD-SBRT, versus conventional chemoradiotherapy for localized pancreatic cancer: a single cancer center evaluation. Cancers, 2022, 14(23):5730., DOI 10.3390/cancers14235730 Reyngold M, O’Reilly EM, Varghese AM, Fiasconaro M, Zinovoy M, Romesser PB, et al. Association of ablative radiation therapy with survival among patients with inoperable pancreatic cancer. JAMA Oncol, 2021, 7(5): 735–8., DOI 10.1001/jamaoncol.2021.0057 Krishnan S, Chadha AS, Suh Y, Chen HC, Rao A, Das P, et al. Focal radiation therapy dose escalation improves overall survival in locally advanced pancreatic cancer patients receiving induction chemotherapy and consolidative chemoradiation. Int J Radiat Oncol Biol Phys, 2016, 94(4):755–65., DOI 10.1016/j. ijrobp.2015.12.003 Teriaca MA, Loi M, Suker M, Eskens FALM, van Eijck CHJ, Nuyttens JJ A phase II study of stereotactic radiotherapy after FOLFIRINOX for locally advanced pancreatic cancer, LAPC-1 trial): long-term outcome. Radiother Oncol, 2021, 155: 232–6., DOI 10.1016/j.radonc.2020.11.006 Rudra S, JiangN, Rosenberg SA, Olsen JR, RoachMC,Wan L, et al.Using adaptive magnetic resonance image-guided radiation therapy for treatment of inoperable pancreatic cancer. Cancer Med, 2019, 8(5):2123–32., DOI 10.1002/cam4.2100 Palta M, Godfrey D, Goodman KA, Hoffe S, Dawson LA, Dessert D, et al. Radiation therapy for pancreatic cancer: executive summary of an ASTRO clinical practice guideline. Pract Radiat Oncol, 2019, 9(5):322–32., DOI 10.1016/j.prro.2019. 06.016 Loi M, Magallon-Baro A, Suker M, van Eijck C, Sharma A, Hoogeman M, et al. Pancreatic cancer treated with SBRT: effect of anatomical interfraction variations on dose to organs at risk. Radiother Oncol, 2019, 134:67–73., DOI 10.1016/j.radonc.2019.01.020 Tyran M, Jiang N, Cao M, Raldow A, Lamb JM, Low D, et al. Retrospective evaluation of decision-making for pancreatic stereotactic MR-guided adaptive radiotherapy. Radiother Oncol, 2018, 129(2):319–25., DOI 10.1016/j.radonc.2018. 08.009 Kim M, Schiff JP, Price A, Laugeman E, Samson PP, Kim H, et al. The first reported case of a patient with pancreatic cancer treated with cone beam computed tomography-guided stereotactic adaptive radiotherapy, CT-STAR). Radiat Oncol, 2022, 17(1):157–11., DOI 10.1186/s13014-022-02125-z Khan M, Sandhu N, Naeem M, Ealden R, Pearson M, Ali A, et al. Implementation of a comprehensive set of optimised CBCT protocols and validation through imaging quality and dose audit. Br J Radiol, 2022, 95(1139): 20220070., DOI 10.1259/bjr.20220070 Kawahara D, Ozawa S, Nakashima T, Suzuki T, Tsuneda M, Tanaka S, et al. Absorbed dose and image quality of varian TrueBeam CBCT compared with OBI CBCT. Physica Med, 2016, 32(12):1628–33., DOI 10.1016/j.ejmp.2016.11.118 Kaur G, Lehmann J, Greer P, Simpson J Assessment of the accuracy of truebeam intrafraction motion review, IMR, system for prostate treatment guidance. Australas Phys Eng Sci Med, 2019, 42(2):585–98., DOI 10.1007/s13246- 019-00760-7 Kisivan K, Erdelyesi D, Gutyina D, Sajti P, Gugyeras D, Farkas A, et al. Hasnyalmirigyrak linearis gyorsito alapu sztereotaxias sugarterapiaja kezeles alatti tumorkovetessel. Magyar Onkologia, 2021, 65(1):6–13. Hanna GG, Murray L, Patel R, Jain S, Aitken KL, Franks KN, et al. UK consensus on normal tissue dose constraints for stereotactic radiotherapy. Clin Oncol, 2018, 30(1):5–14., DOI 10.1016/j.clon.2017.09.007 Koay EJ, Hanania AN, Hall WA, Taniguchi CM, Rebueno N, Myrehaug S, et al. Dose-escalated radiation therapy for pancreatic cancer: a simultaneous integrated boost approach. Pract Radiat Oncol, 2020, 10(6):e495–e507., DOI 10. 1016/j.prro.2020.01.012 Krippendorff K Agreement and information in the reliability of coding. Commun Methods Measures, 2011, 5(2):93–112., DOI 10.1080/19312458.2011. 568376 Gaya A, Camilleri P, Nash A, Hughes D, Good J Implementation of stereotactic MRI-guided adaptive radiotherapy, SMART, for hepatobiliary and pancreatic cancers in the United Kingdom – fifty in five. Cureus, 2021, 13(5): e15075., DOI 10.7759/cureus.15075 Chuong MD, Bryant J, Mittauer KE, Hall M, Kotecha R, Alvarez D, et al. Ablative 5-fraction stereotactic magnetic resonance–guided radiation therapy with on-table adaptive replanning and elective nodal irradiation for inoperable pancreas cancer. Pract Radiat Oncol, 2021, 11(2):134–47., DOI 10.1016/j.prro. 2020.09.005 Reyngold M, Parikh P, Crane CH Ablative radiation therapy for locally advanced pancreatic cancer: techniques and results. Radiat Oncol, 2019, 14(1):95., DOI 10.1186/s13014-019-1309-x Colbert LE, Rebueno N, Moningi S, Beddar S, Sawakuchi GO, Herman JM, et al. Dose escalation for locally advanced pancreatic cancer: how high can we go? Adv Radiat Oncol, 2018, 3(4):693–700., DOI 10.1016/j.adro.2018.07.008 Zaorsky NG, Lehrer EJ, Handorf E, Meyer JE Dose escalation in stereotactic body radiation therapy for pancreatic cancer: a meta-analysis. Am J Clin Oncol Cancer Clin Trials, 2019, 42(1):46–55., DOI 10.1097/COC.0000000000000472 Chuong MD, Kirsch C, Herrera R, Rubens M, Gungor G, Schaff EM, et al. Long-term multi-institutional outcomes of 5-fraction ablative stereotactic MRguided adaptive radiation therapy, SMART, for inoperable pancreas cancer with median prescribed biologically effective dose of 100 Gy10. Int J Radiat Oncol. Biol. Phys., 2021, 111(3):S147–8., DOI 10.1016/j.ijrobp.2021.07.330 Parikh PJ, Lee P, Low D, Kim J, Mittauer KE, Bassetti MF, et al. Stereotactic MR-guided on-table adaptive radiation therapy, SMART, for patients with borderline or locally advanced pancreatic cancer: primary endpoint outcomes of a prospective phase II multi-center international trial. Int J Radiat Oncol. Biol. Phys., 2022, 114(5):1062–3., DOI 10.1016/j.ijrobp.2022.09.010 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611456 EP 1611467 DI 10.3389/pore.2023.1611456 PG 12 ER PT J AU Hasanova, A Asadov, Ch Karimova, N Shirinova, A Aliyeva, G Alimirzoyeva, Z AF Hasanova, Aypara Asadov, Chingiz Karimova, Nigar Shirinova, Aytan Aliyeva, Gunay Alimirzoyeva, Zohra TI Spectrum of BCR-ABL mutations in Azerbaijanian imatinib-resistant patients with chronic myeloid leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chronic myeloid leukemia; mutation; tyrosine kinase inhibitor resistance; kinase domain; BCR-ABL ID chronic myeloid leukemia; mutation; tyrosine kinase inhibitor resistance; kinase domain; BCR-ABL AB Objective: BCR-ABL1 kinase domain (KD) mutations can lead to resistance to first- and second-generation tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia (CML). Here, we present the first report of the spectrum of mutations in the BCR-ABL1 KD of CML patients from Azerbaijan. Materials and methods: Samples for mutation screening were obtained from patients experiencing resistance to first line TKIs or from patients in acceleration phase (AP) or blast crisis (BC) at the time of diagnosis. The cDNA region corresponding to BCR-ABL1 KD was sequenced by pyrosequencing method. The χ2 test was used to assess the association of categorical variables between mutation-positive and -negative groups. In addition, the Kaplan-Meier method was applied to generate survival curves. Results: Eight different pointmutations were identified in 22 (13.4%) out of 163CML patients experiencing resistance to TKIs. The types of mutations detected were as follows: Contact binding site mutations 50% (11), SH2 domain mutations 27.4% (six), P-loop mutations 18.1% (four), and SH3 domain mutations accounting for 4.5% (one). The most common mutation was T315I, accounting for 5% (n = 8) of all patients. Significant association was identified between BCR-ABL1 mutations and additional chromosomal aberrations as well as between the mutations and disease phases (p < 0.05). Twelve out of 22 patients with BCR-ABL1 mutations and seven out of eight with T315I were in BC. Overall survival (OS) of the patients with BCRABL1 mutations was significantly lower comparing to the patients with nomutation (p < 0.05) and 8 patients with T315I mutation presented OS of 0%. Conclusion: T315I was the most commonly identified BCR-ABL1 mutation in TKI-resistant CML patients of Azerbaijani origin, being associated with disease progression and poor OS. C1 [Hasanova, Aypara] Institute of Hematology and Transfusiology, Leukemogenesis LaboratoryBaku, Azerbaijan. [Asadov, Chingiz] Institute of Hematology and Transfusiology, Hematopoiesis DepartmentBaku, Azerbaijan. [Karimova, Nigar] Institute of Genetic Resources, Biotechnology Department, Human Genetics LaboratoryBaku, Azerbaijan. [Shirinova, Aytan] Institute of Hematology and Transfusiology, Hematology DepartmentBaku, Azerbaijan. [Aliyeva, Gunay] Institute of Hematology and Transfusiology, Hematopoiesis DepartmentBaku, Azerbaijan. [Alimirzoyeva, Zohra] Institute of Hematology and Transfusiology, Hematology DepartmentBaku, Azerbaijan. RP Aliyeva, G (reprint author), Institute of Hematology and Transfusiology, Hematopoiesis Department, Baku, Azerbaijan. EM galiyeva@rocketmail.com CR Baccarani W, Deininger MW, Rosti G, Hochhaus A, Soverini S, Apperley JF, et al. European leukemianet recommendations for the management of chronic myeloid leukemia. Blood, 2013, 122(6):872–84., DOI 10.1182/blood-2013-05-501569 Apperley JF. Chronic myeloid leukaemia. Lancet, 2015, 385:1447–59., DOI 10. 1016/S0140-6736(13)62120-0 O’Brien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, Cervantes F, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med, 2003, 348: 994–1004., DOI 10.1056/nejmoa022457 Gratwohl A, Brand R, Apperley J, Ruutu T, Corradini P, Carreras E, et al. Allogeneic hematopoietic stem cell transplantation for chronic myeloid leukemia in Europe 2006: transplant activity, long-term data and current results. An analysis by the chronic leukemia working party of the european group for blood and marrow transplantation, EBMT). Haematologica, 2006, 91:513e21. Hughes T, Saglio G, Branford S, Soverini S, Kim DW, Muller MC, et al. Impact of baseline BCR-ABL mutations on response to nilotinib in patients with chronic myeloid leukemia in chronic phase. J Clin Oncol, 2009, 27(25):4204–10., DOI 10. 1200/JCO.2009.21.8230 Muller MC, Cortes JE, Kim DW, Druker BJ, Erben P, Pasquini R, et al. Dasatinib treatment of chronic-phase chronic myeloid leukemia: analysis of responses according to preexisting BCR-ABL mutations. Blood, 2009, 114(24): 4944–53., DOI 10.1182/blood-2009-04-214221 Parker WT, Yeung DTO, Yeoman AL, Altamura HK, Jamison BA, Field CR, et al. The impact of multiple low-level BCR-ABL1 mutations on response to ponatinib. Blood, 2016, 127(15):1870–80., DOI 10.1182/blood-2015-09-666214 Pasic I, Lipton JH. Current approach to the treatment of chronic myeloid leukaemia. Leuk Res, 2017, 55:65–78., DOI 10.1016/j.leukres.2017.01.005 Loscocco F, Visani G, Galimberti S, Curti A, Isidori A. BCR-ABL independent mechanisms of resistance in chronic myeloid leukemia. Front Oncol, 2019, 9:939., DOI 10.3389/fonc.2019.00939 Eide CA, O’Hare T. Chronic myeloid leukemia: advances in understanding disease biology and mechanisms of resistance to tyrosine kinase inhibitors. Curr Hematol Malig Rep, 2015, 10:158–66., DOI 10.1007/s11899-015-0248-3 Hochhaus A. Chronic myelogenous leukemia, CML): resistance to tyrosine kinase inhibitors. Ann Oncol, 2006, 17(Suppl. 10):x274–9., DOI 10.1093/annonc/ mdl273 Baccarani M, Cortes J, Pane F, Niederwieser D, Saglio G, Apperley J, et al. Chronicmyeloid leukemia: an update of concepts and management recommendations of European LeukemiaNet. J Clin Oncol, 2009, 27:6041–51., DOI 10.1200/JCO.2009.25.0779 O’Brien S, Radich JP, Abboud CN, Akhtari M, Altman JK, Berman E, et al. Chronic myelogenous leukemia, version 1.2014. J Natl Compr Canc Netw, 2013, 11(11):1327–1340., DOI 10.6004/jnccn.2013.0157 Apperley JF. Part I: mechanisms of resistance to imatinib in chronic myeloid leukaemia. Lancet Oncol, 2007, 8:1018–29., DOI 10.1016/S1470-2045(07)70342-X Khorashad JS, Kelley TW, Szankasi P, Mason CC, Soverini S, Adrian LT, et al. BCR-ABL1 compound mutations in tyrosine kinase inhibitor-resistant CML: frequency and clonal relationships. Blood, 2013, 121:489–98., DOI 10.1182/blood- 2012-05-431379 Soverini S, Hochhaus A, Nicolini FE, Gruber F, Lange T, Saglio G, et al. BCRABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet. Blood, 2011, 118:1208–15., DOI 10.1182/blood-2010-12- 326405 Shiotsu Y, Kiyoi H, Ishikawa Y, Tanizaki R, Shimizu M, Umehara H, et al. KW-2449, a novel multikinase inhibitor, suppresses the growth of leukemia cells with FLT3 mutations or T315I-mutated BCR/ABL translocation. Blood, 2009, 114: 1607–17., DOI 10.1182/blood-2009-01-199307 Moore AS, Blagg J, Linardopoulos S, Pearson ADJ. Aurora kinase inhibitors: novel small molecules with promising activity in acute myeloid and Philadelphiapositive leukemias. Leukemia, 2010, 24:671–8., DOI 10.1038/leu.2010.15 Giles FJ, Cortes J, Jones D, Bergstrom D, Kantarjian H, Freedman SJ. MK- 0457, a novel kinase inhibitor, is active in patients with chronic myeloid leukemia or acute lymphocytic leukemia with the T315I BCR-ABL mutation. Blood, 2007, 109: 500–2., DOI 10.1182/blood-2006-05-025049 Modugno M, Casale E, Soncini C, Rosettani P, Colombo R, Lupi R, et al. Crystal structure of the T315I Abl mutant in complex with the aurora kinases inhibitor PHA-739358. Cancer Res, 2007, 67:7987–90., DOI 10.1158/0008-5472. CAN-07-1825 Hughes T, Saglio G, Branford S, Soverini S, Kim DW,Muller MC, et al. Impact of baseline BCR-ABL mutations on response to nilotinib in patients with chronic myeloid leukemia in chronic phase. J Clin Oncol, 2009, 27:4204–10., DOI 10.1200/ JCO.2009.21.8230 Soverini S, Gnani A, Colarossi S, Castagnetti F, Abruzzese E, Paolini S, et al. Philadelphia-positive patients who already harbor imatinib-resistant Bcr-Abl kinase domain mutations have a higher likelihood of developing additional mutations associated with resistance to second- or third-line tyrosine kinase inhibitors. Blood, 2009, 114:2168–71., DOI 10.1182/blood-2009-01-197186 Muller MC, Cortes JE, Kim DW, Druker BJ, Erben P, Pasquini R, et al. Dasatinib treatment of chronic-phase chronic myeloid leukemia: analysis of responses according to preexisting BCR-ABL mutations. Blood, 2009, 114: 4944–53., DOI 10.1182/blood-2009-04-214221 Shah NP, Skaggs BJ, Branford S, Hughes TP, Nicoll JM, Paquette RL, et al. Sequential ABL kinase inhibitor therapy selects for compound drug-resistant BCRABL mutations with altered oncogenic potency. J Clin Invest, 2007, 117:2562–9., DOI 10.1172/JCI30890 Soverini S, Colarossi S, Gnani A, Castagnetti F, Rosti G, Bosi C, et al. Resistance to dasatinib in Philadelphia-positive leukemia patients and the presence or the selection of mutations at residues 315 and 317 in the BCR-ABL kinase domain. Haematological, 2007, 92:401–4., DOI 10.3324/haematol.10822 Parker WT, Ho M, Scott HS, Hughes TP, Branford S. Poor response to second-line kinase inhibitors in chronic myeloid leukemia patients with multiple low-level mutations, irrespective of their resistance profile. Blood, 2012, 119: 2234–8., DOI 10.1182/blood-2011-08-375535 Parker WT, Lawrence RM, Ho M, Irwin DL, Scott HS, Hughes TP, et al. Sensitive detection of BCR-ABL1 mutations in patients with chronic myeloid leukemia after imatinib resistance is predictive of outcome during subsequent therapy. J Clin Oncol, 2011, 29:4250–9., DOI 10.1200/JCO.2011.35.0934 Deininger MW, Shah NP, Altman JK, Berman E, Bhatia R, Bhatnagar B, et al. Chronic myeloid leukemia, version 2.2021, NCCN clinical practice guidelines in oncology. NCCN Clinical Practice Guidelines Oncology, 2020, 18:1385–415., DOI 10. 6004/jnccn.2020.0047 Apperley JF. Part I: mechanisms of resistance to imatinib in chronic myeloid leukaemia. Lancet Oncol, 2007, 8:1018–29., DOI 10.1016/S1470-2045(07)70342-X Baccarani M, Castagnetti F, Gugliotta G, Rosti G. A review of the European LeukemiaNet recommendations for the management of CML. Ann Hematol, 2015, 94(Suppl. 2):S141–7., DOI 10.1007/s00277-015-2322-2 Soverini S, De Benedittis C, Mancini M, Martinelli G. Present and future of molecular monitoring in chronic myeloid leukaemia. Br J Haematol, 2016, 173: 337–49., DOI 10.1111/bjh.13966 Corbin AS, La Rosee P, Stoffregen EP, Druker BJ, Deininger MW. Several BCRABL kinase domain mutants associated with imatinib mesylate resistance remain sensitive to imatinib. Blood, 2003, 101:4611–4., DOI 10.1182/blood-2002-12-3659 Hochhaus A, Schenk T, Erben P, Ernst T, La Rosee P, Muller MC. Cause and management of therapy resistance. Best Pract Res Clin Haematol, 2009, 22:367–79., DOI 10.1016/j.beha.2009.05.004 Soverini S, de Benedittis C,Mancini M, Martinelli G. Mutations in the BCRABL1 kinase domain and elsewhere in chronic myeloid leukemia. Clin Lymphoma Myeloma Leuk, 2015, S1(Suppl. l):S120–S128., DOI 10.1016/j.clml. 2015.02.035 Hochhaus A, Baccarani M, Silver RT, Schiffer C, Apperley JF, Cervantes F, et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia, 2020, 34:966–84., DOI 10.1038/s41375-020- 0776-2 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611518 EP 1611528 DI 10.3389/pore.2023.1611518 PG 11 ER PT J AU Makk, E Bohonyi, N Oszter, A Eles, K Tornoczky, T Toth, A Kalman, E Kovacs, K AF Makk, Evelin Bohonyi, Noemi Oszter, Angela Eles, Klara Tornoczky, Tamas Toth, Arnold Kalman, Endre Kovacs, Krisztina TI Comparative analysis of EZH2, p16 and p53 expression in uterine carcinosarcomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE uterine cancer; carcinosarcoma; EZH2; p16; p53 ID uterine cancer; carcinosarcoma; EZH2; p16; p53 AB Introduction: The role of p16 and p53 immunohistochemistry in the diagnosis of rare and aggressive uterine carcinosarcoma (UCS) has been well established. However, enhancer of zeste homolog 2 (EZH2), a histone methyltransferase and a member of the polycomb group family is a relatively new biomarker, with limited published data on its significance in this tumor type. The goal of this study was to examine EZH2 expression in UCS and its components, in correlation with morphological features, and p16 and p53 staining patterns. Methods: Twenty-eight UCSs were included in the study. EZH2, p16 and p53 immunoreactivity were assessed independently by two pathologists in both tumor components (epithelial and mesenchymal). EZH2 and p16 immunostains were scored semiquantitatively: based on the percentage and intensity of tumor cell staining a binary staining index (“high- or lowexpressing”) was calculated. The p53 staining pattern was evaluated as wildtype or aberrant (diffuse nuclear, null, or cytoplasmic expression). Statistical tests were used to evaluate the correlation between staining patterns for all three markers and the different tumor components and histotypes. Results: High EZH2 and p16 expression and aberrant p53 patterns were present in 89.3% 78.6% and 85.7% of the epithelial component and in 78.6%, 62.5% and 82.1% of the mesenchymal component, respectively. Differences among these expression rates were not found to be significant (p > 0.05). Regarding the epithelial component, aberrant p53 pattern was found to be significantly (p = 0.0474) more frequent in the serous (100%) than in endometrioid (66.6%) histotypes. Within the mesenchymal component, p53 null expression pattern occurred significantly (p = 0.0257) more frequently in heterologous sarcoma components (71.4%) compared to the homologous histotype (18.8%). Conclusion: In conclusion, EZH2, p16 and p53 seem to play a universal role in the pathogenesis of UCS; however, a distinctive pattern of p53 expression appears to exist between the serous and endometrioid carcinoma components and also between the homologous and heterologous sarcoma components. C1 [Makk, Evelin] University of Pecs, Department of PathologyPecs, Hungary. [Bohonyi, Noemi] Pecsi Tudomanyegyetem, Altalanos Orvostudomanyi Kar, Szuleszeti es Nogyogyaszati KlinikaPecs, Hungary. [Oszter, Angela] University of Pecs, Department of PathologyPecs, Hungary. [Eles, Klara] University of Pecs, Department of PathologyPecs, Hungary. [Tornoczky, Tamas] University of Pecs, Department of PathologyPecs, Hungary. [Toth, Arnold] PTE KK, Radiologiai KlinikaPecs, Hungary. [Kalman, Endre] University of Pecs, Department of PathologyPecs, Hungary. [Kovacs, Krisztina] University of Pecs, Department of PathologyPecs, Hungary. RP Makk, E (reprint author), University of Pecs, Department of Pathology, Pecs, Hungary. EM makk.evelin@gmail.com CR Loda M, Mucci LA, Mittelstadt ML, HemelrijckMV, Cotter MB. Pathology and epidemiology of cancer. Cham: Springer International Publishing, 2016)., DOI 10. 1007/978-3-319-35153-7 Silverberg SG, Major FJ, Blessing JA, Fetter B, Askin FB, Liao SY, et al. Carcinosarcoma, malignant mixed mesodermal tumor, of the uterus: a gynecologic oncology group pathologic study of 203 cases. Int J Gynecol Pathol, 1990, 9(1):1–19., DOI 10.1097/00004347-199001000-00001 Artioli G, Wabersich J, Ludwig K, Gardiman MP, Borgato L, Garbin F. Rare uterine cancer: carcinosarcomas: review from histology to treatment. Crit Rev Oncol Hematol, 2015, 94(1):98–104., DOI 10.1016/j.critrevonc.2014.10.013 Leskela S, Perez-Mies B, Rosa-Rosa JM, Cristobal E, Biscuola M, Palacios- Berraquero ML, et al. Molecular basis of tumor heterogeneity in endometrial carcinosarcoma. Cancers, Basel,, 2019, 11:964., DOI 10.3390/cancers11070964 Kernochan LE, Garcia RL. Carcinosarcomas, malignant mixed mullerian tumor, of the uterus: advances in elucidation of biologic and clinical characteristics. J Natl Compr Cancer Netw, 2009, 7(5):550–6., DOI 10.6004/jnccn.2009.0037 Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest, 2009, 119(6):1420–8., DOI 10.1172/JCI39104 Cherniack AD, Shen H, Walter V, Stewart C, Murray BA, Bowlby R, et al. Integrated molecular characterization of uterine carcinosarcoma. Cancer Cell, 2017, 31(3):411–23., DOI 10.1016/j.ccell.2017.02.010 George E, Lillemoe TJ, Twiggs LB, Perrone T. Malignant mixed mullerian tumor versus high-grade endometrial carcinoma and aggressive variants of endometrial carcinoma: a comparative analysis of survival. Int J Gynecol Pathol, 1995, 14(1):39–44., DOI 10.1097/00004347-199501000-00007 Zhang C, Hu W, Jia N, Li Q, Hua K, Tao X, et al. Uterine carcinosarcoma and high-risk endometrial carcinomas: a clinicopathological comparison. Int J Gynecol Cancer, 2015, 25(4):629–36., DOI 10.1097/IGC.0000000000000350 Hock H. A complex Polycomb issue: the two faces of EZH2 in cancer. Genes Dev, 2012, 26(8):751–5., DOI 10.1101/gad.191163.112 Xie Z, Zhong C, Shen J, Jia Y, Duan S. LINC00963: a potential cancer diagnostic and therapeutic target. Biomed Pharmacother, 2022, 150:113019., DOI 10. 1016/J.BIOPHA.2022.113019 Sasaki M, Nakanuma Y. Cellular senescence in biliary pathology. Special emphasis on expression of a polycomb group protein EZH2 and a senescent marker p16INK4a in bile ductular tumors and lesions. Histol Histopathol, 2015, 30(3): 267–75., DOI 10.14670/HH-30.267 Said J. Biomarker discovery in urogenital cancer. Biomarkers, 2005, 10(1): 83–6., DOI 10.1080/13547500500215050 Pourakbar S, Pluard TJ, Aaccurso AD, Farassati F. Ezh2, a novel target in detection and therapy of breast cancer. Onco Targets Ther, 2017, 10:2685–7., DOI 10. 2147/OTT.S138777 Findeis-Hosey JJ, Huang J, Li F, Yang Q, McMahon LA, Xu H. High-grade neuroendocrine carcinomas of the lung highly express enhancer of zeste homolog 2, but carcinoids do not. Hum Pathol, 2011, 42(6):867–72., DOI 10.1016/j.humpath. 2010.09.019 Choi JH, Song YS, Yoon JS, Song KW, Lee YY. Enhancer of zeste homolog 2 expression is associated with tumor cell proliferation and metastasis in gastric cancer. APMIS, 2010, 118(3):196–202., DOI 10.1111/j.1600-0463.2009. 02579.x Fluge O, Gravdal K, Carlsen E, Vonen B, Kjellevold K, Refsum S, et al. Expression of EZH2 and Ki-67 in colorectal cancer and associations with treatment response and prognosis. Br J Cancer, 2009, 101(8):1282–9., DOI 10.1038/sj.bjc. 6605333 Toll AD, Dasgupta A, Potoczek M, Yeo CJ, Kleer CG, Brody JR, et al. Implications of enhancer of zeste homologue 2 expression in pancreatic ductal adenocarcinoma. Hum Pathol, 2010, 41(9):1205–9., DOI 10.1016/j.humpath.2010. 03.004 Zhai R, Tang F, Gong J, Zhang J, Lei B, Li B, et al. The relationship between the expression of USP22, BMI1, and EZH2 in hepatocellular carcinoma and their impacts on prognosis. Onco Targets Ther, 2016, 9:6987–98., DOI 10.2147/OTT. S110985 Borbone E, Troncone G, Ferraro A, Jasencakova Z, Stojic L, Esposito F, et al. Enhancer of zeste homolog 2 overexpression has a role in the development of anaplastic thyroid carcinomas. J Clin Endocrinol Metab, 2011, 96(4):1029–38., DOI 10.1210/jc.2010-1784 Aziza EA, Shimaa AA, Ragab AA. Prognostic value of twist-1, E-cadherin and EZH2 in prostate cancer: an immunohistochemical study. Turk Patoloji Derg, 2017, 1:198–210., DOI 10.5146/tjpath.2016.01392 Raman JD, Mongan NP, Tickoo SK, Boorjian SA, Scherr DS, Gudas LJ. Increased expression of the polycomb group gene, EZH2, in transitional cell carcinoma of the bladder. Clin Cancer Res, 2005, 11(24):8570–6., DOI 10.1158/ 1078-0432.CCR-05-1047 Jin M, Yang Z, Ye W, Yu X, Hua X. Prognostic significance of histone methyltransferase enhancer of zeste homolog 2 in patients with cervical squamous cell carcinoma. Oncol Lett, 2015, 10(2):857–62., DOI 10.3892/ol.2015.3319 Makk E, Balint L, Cifra J, Tornoczky T, Oszter A, Toth A, et al. Robust expression of EZH2 in endocervical neoplastic lesions. Virchows Arch, 2019, 475(1):95–104., DOI 10.1007/s00428-019-02550-8 Zhou J, Roh JW, Bandyopadhyay S, Chen Z, Munkarah AR, Hussein Y, et al. Overexpression of enhancer of zeste homolog 2, EZH2, and focal adhesion kinase, FAK, in high grade endometrial carcinoma. Gynecol Oncol, 2013, 128(2):344–8., DOI 10.1016/j.ygyno.2012.07.128 Krill L, Deng W, Eskander R, Mutch D, Zweizig S, Hoang B, et al. Overexpression of enhance of zeste homolog 2, EZH2, in endometrial carcinoma: an NRG oncology/gynecologic oncology group study. Gynecol Oncol, 2020, 156(2):423–9., DOI 10.1016/J.YGYNO.2019.12.003 Xu Y, Li X, Wang H, Xie P, Yan X, Bai Y, et al. Hypermethylation of CDH13, DKK3 and FOXL2 promoters and the expression of EZH2 in ovary granulosa cell tumors. Mol Med Rep, 2016, 14(3):2739–45., DOI 10.3892/mmr. 2016.5521 Li H, Cai Q, Godwin AK, Zhang R. Enhancer of zeste homolog 2 promotes the proliferation and invasion of epithelial ovarian cancer cells. Mol Cancer Res, 2010, 8(12):1610–8., DOI 10.1158/1541-7786.MCR-10-0398 Ramaglia M, D’Angelo V, Iannotta A, Di Pinto D, Pota E, Affinita MC, et al. High EZH2 expression is correlated to metastatic disease in pediatric soft tissue sarcomas. Cancer Cell Int, 2016, 16(1):59., DOI 10.1186/S12935-016-0338-X Adesso L, Leoncini PP, Dall’agnese A, Walters ZS, Verginelli F, Boldrini R, et al. The Polycomb group, PcG, protein EZH2 supports the survival of PAX3- FOXO1 alveolar rhabdomyosarcoma by repressing FBXO32, Atrogin1/MAFbx). Oncogene, 2014, 33:4173–84., DOI 10.1038/onc.2013.471 Zhang N, Zeng Z, Li S, Wang F, Huang P. High expression of EZH2 as a marker for the differential diagnosis of malignant and benign myogenic tumors. Sci Rep, 2018, 8:12331., DOI 10.1038/s41598-018-30648-7 Shen JK, Cote GM, Gao Y, Choy E, Mankin HJ, Hornicek FJ, et al. Targeting EZH2-mediated methylation of H3K27 inhibits proliferation and migration of Synovial Sarcoma in vitro. Sci Rep, 2016, 6:25239., DOI 10.1038/srep25239 Sun R, Shen J, Gao Y, Zhou Y, Yu Z, Hornicek F, et al. Overexpression of EZH2 is associated with the poor prognosis in osteosarcoma and function analysis indicates a therapeutic potential. Oncotarget, 2016, 7(25):38333–46., DOI 10.18632/ oncotarget.9518 Girard N, Bazille C, Lhuissier E, Benateau H, Llombart-Bosch A, Boumediene K, et al. 3-Deazaneplanocin A, DZNep), an inhibitor of the histone methyltransferase EZH2, induces apoptosis and reduces cell migration in chondrosarcoma cells. PLoS One, 2014, 9(5):e98176., DOI 10.1371/JOURNAL. PONE.0098176 Cao Q, Yu J, Dhanasekaran SM, Kim JH, Mani RS, Tomlins SA, et al. Repression of E-cadherin by the polycomb group protein EZH2 in cancer. Oncogene, 2008, 27(58):7274–84., DOI 10.1038/onc.2008.333 Ihira K, Dong P, Xiong Y, Watari H, Konno Y, Hanley SJB, et al. EZH2 inhibition suppresses endometrial cancer progression via miR-361/Twist axis. Oncotarget, 2017, 8(8):13509–20., DOI 10.18632/oncotarget.14586 Ang PP, Tan GC, Karim N, Wong YP. Diagnostic value of the EZH2 immunomarker in malignant effusion cytology. Acta Cytol, 2020, 64(3): 248–55., DOI 10.1159/000501406 Huvila J, Pors J, Thompson EF, Gilks CB. Endometrial carcinoma: molecular subtypes, precursors and the role of pathology in early diagnosis. J Pathol, 2021, 253(4):355–65., DOI 10.1002/PATH.5608 Buza N, Tavassoli FA. Comparative analysis of P16 and P53 expression in uterine malignant mixed mullerian tumors. Int J Gynecol Pathol, 2009, 28(6): 514–21., DOI 10.1097/PGP.0b013e3181a934e9 WHO Classification of Tumours Editorial Board. Female genital tumours. 5th ed. International Agency for Research on Cancer, 2020). Volume 4. Available at: https://publications.iarc.fr/Book-And-Report-Series/Who-Classification-Of- Tumours/Female-Genital-Tumours-2020. Bosari S, Lee AKC, Viale G, Heatley GJ, Coggi G. Abnormal p53 immunoreactivity and prognosis in node-negative breast carcinomas with long-term follow-up. Virchows Arch A Pathol Anat Histopathol, 1992, 421(4): 291–5., DOI 10.1007/BF01660975 Bachmann IM, Halvorsen OJ, Collett K, Stefansson IM, Straume O, Haukaas SA, et al. EZH2 expression is associated with high proliferation rate and aggressive tumor subgroups in cutaneous melanoma and cancers of the endometrium, prostate, and breast. J Clin Oncol, 2006, 24:268–73., DOI 10.1200/JCO.2005.01.5180 Wagener N, Macher-Goeppinger S, Pritsch M, Husing J, Hoppe-Seyler K, Schirmacher P, et al. Enhancer of zeste homolog 2, EZH2, expression is an independent prognostic factor in renal cell carcinoma. BMC Cancer, 2010, 10(1):524., DOI 10.1186/1471-2407-10-524 Sasaki M, Matsubara T, Kakuda Y, Sato Y, Nakanuma Y. Immunostaining for polycomb group protein EZH2 and senescent marker p16INK4a may be useful to differentiate cholangiolocellular carcinoma from ductular reaction and bile duct adenoma. Am J Surg Pathol, 2014, 38(3):364–9., DOI 10.1097/PAS. 0000000000000125 Salvesen HB, Iversen OE, Akslen LA. Prognostic significance of angiogenesis and ki-67, p53, and p21 expression: a population-based endometrial carcinoma study. J Clin Oncol, 1999, 17(5):1382–90., DOI 10.1200/JCO.1999.17.5.1382 Chen X, Arend R, Hamele-Bena D, Tergas AI, Hawver M, Tong GX, et al. Uterine carcinosarcomas: clinical, histopathologic and immunohistochemical characteristics. Int J Gynecol Pathol, 2017, 36(5):412–9., DOI 10.1097/PGP. 0000000000000346 Kobel M, Ronnett BM, Singh N, Soslow RA, Gilks CB, McCluggage WG. Interpretation of P53 immunohistochemistry in endometrial carcinomas: toward increased reproducibility. Int J Gynecol Pathol, 2019, 38(1):S123–S131., DOI 10. 1097/PGP.0000000000000488 Rabban JT, Garg K, Ladwig NR, Zaloudek CJ, Devine WP. Cytoplasmic pattern p53 immunoexpression in pelvic and endometrial carcinomas with TP53 mutation involving nuclear localization domains: an uncommon but potential diagnostic pitfall with clinical implications. Am J Surg Pathol, 2021, 45(11):1441–51., DOI 10.1097/PAS.0000000000001713 Schoonjans F, Zalata A, Depuydt CE, Comhaire FH. MedCalc: a new computer program for medical statistics. Comput Methods Programs Biomed, 1995, 48(3):257–62., DOI 10.1016/0169-2607(95)01703-8 Cohen J. A coefficient of agreement for nominal scales. Educ Psychol Meas, 1960, 20(1):37–46., DOI 10.1177/001316446002000104 Engelsen IB, Stefansson I, Akslen LA, Salvesen HB. Pathologic expression of p53 or p16 in preoperative curettage specimens identifies high-risk endometrial carcinomas. Am J Obstet Gynecol, 2006, 195(4):979–86., DOI 10.1016/j.ajog.2006. 02.045 Priya A, Chaurasia JK, P K, Panwar H, Yadav SK, Kapoor N. Evaluation of immunohistochemical expression of enhancer of zeste homolog 2, EZH2, and its association with clinicopathological variables in carcinoma cervix. Cureus, 2023, 15(3):e36946., DOI 10.7759/CUREUS.36946 Oki S, Sone K, Oda K, Hamamoto R, Ikemura M, Maeda D, et al. Oncogenic histone methyltransferase EZH2: a novel prognostic marker with therapeutic potential in endometrial cancer. Oncotarget, 2017, 8:40402–11., DOI 10.18632/ oncotarget.16316 Rao ZY, Cai MY, Yang GF, He LR, Mai SJ, Hua WF, et al. EZH2 supports ovarian carcinoma cell invasion and/or metastasis via regulation of TGF-beta1 and is a predictor of outcome in ovarian carcinoma patients. Carcinogenesis, 2010, 31(9):1576–83., DOI 10.1093/carcin/bgq150 Ding M, Zhang H, Li Z, Wang C, Chen J, Shi L, et al. The polycomb group protein enhancer of zeste 2 is a novel therapeutic target for cervical cancer. Clin Exp Pharmacol Physiol, 2015, 42(5):458–64., DOI 10.1111/1440-1681.12382 Bitler BG, Aird KM, Garipov A, Li H, Amatangelo M, Kossenkov AV, et al. Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1Amutated cancers. Nat Med, 2015, 21(3):231–8., DOI 10.1038/NM.3799 Roh JW, Eun Choi J, Dong Han H, Hu W, Matsuo K, Nishimura M, et al. Clinical and biological significance of EZH2 expression in endometrial cancer. Cancer Biol Ther, 2019, 21:147–56., DOI 10.1080/15384047.2019.1672455 Shi L, Zhang Q, Zhu S, Tang Q, Chen X, Lan R, et al. Pharmacological inhibition of EZH2 using a covalent inhibitor suppresses human ovarian cancer cell migration and invasion. Mol Cell Biochem, 2023)., DOI 10.1007/S11010-023-04767-3 Abargel A, Avinoach I, Kravtsov V, Boaz M, Glezerman M, Menczer J. Expression of p27 and p53: comparative analysis of uterine carcinosarcoma and endometrial carcinoma. Int J Gynecol Cancer, 2004, 14(2):354–9., DOI 10.1111/j. 1048-891x.2004.014221.x Menczer J, Kravtsov V, Levy T, Berger E, Glezerman M, Avinoach I. Expression of c-kit in uterine carcinosarcoma. Gynecol Oncol, 2005, 96(1): 210–5., DOI 10.1016/j.ygyno.2004.09.045 Menczer J, Schreiber L, Sukmanov O, Kravtsov V, Berger E, Golan A, et al. COX-2 expression in uterine carcinosarcoma. Acta Obstet Gynecol Scand, 2010, 89(1):120–5., DOI 10.3109/00016340903342006 Livasy CA, Reading FC, Moore DT, Boggess JF, Lininger RA. EGFR expression and HER2/neu overexpression/amplification in endometrial carcinosarcoma. Gynecol Oncol, 2006, 100(1):101–6., DOI 10.1016/j.ygyno.2005. 07.124 Saglam O, Husain S, Toruner G. AKT, EGFR, C-ErbB-2, and C-kit expression in uterine carcinosarcoma. Int J Gynecol Pathol, 2013, 32(5):493–500., DOI 10.1097/ PGP.0b013e31827fedef Rottmann D, Snir OL, Wu X, Wong S, Hui P, Santin AD, et al. HER2 testing of gynecologic carcinosarcomas: tumor stratification for potential targeted therapy. Mod Pathol, 2020, 33(1):118–27., DOI 10.1038/S41379-019-0358-x Jenkins TM, Cantrell LA, Stoler MH, Mills AM. HER2 overexpression and amplification in uterine carcinosarcomas with serousmorphology. Am J Surg Pathol, 2022, 46(4):435–42., DOI 10.1097/PAS.0000000000001870 Murali R, Davidson B, Fadare O, Carlson JA, Crum CP, Gilks CB, et al. Highgrade endometrial carcinomas: morphologic and immunohistochemical features, diagnostic challenges and recommendations. Int J Gynecol Pathol, 2019, 38(1): S40–S63., DOI 10.1097/PGP.0000000000000491 Yemelyanova A, Ji H, Shih IM, Wang TL, Wu LSF, Ronnett BM. Utility of p16 expression for distinction of uterine serous carcinomas from endometrial endometrioid and endocervical adenocarcinomas: immunohistochemical analysis of 201 cases. Am J Surg Pathol, 2009, 33(10):1504–14., DOI 10.1097/PAS. 0B013E3181AC35F5 Gilks CB, Oliva E, Soslow RA. Poor interobserver reproducibility in the diagnosis of high-grade endometrial carcinoma. Am J Surg Pathol, 2013, 37(6): 874–81., DOI 10.1097/PAS.0B013E31827F576A Garg K, Leitao MM, Wynveen CA, Sica GL, Shia J, Shi W, et al. p53 overexpression in morphologically ambiguous endometrial carcinomas correlates with adverse clinical outcomes. Mod Pathol, 2010, 23(1):80–92., DOI 10.1038/MODPATHOL.2009.153 Tashiro H, Isacson C, Levine R, Kurman RJ, Cho KR, Hedrick L. p53 gene mutations are common in uterine serous carcinoma and occur early in their pathogenesis. Am J Pathol, 1997, 150(1):177–85. Available from: https://www. ncbi.nlm.nih.gov/pmc/articles/PMC1858541/, Accessed July 27, 2023). Soslow RA, Shen PUF, Chung MH, Isacson C. Distinctive p53 and mdm2 immunohistochemical expression profiles suggest different pathogenetic pathways in poorly differentiated endometrial carcinoma. Int J Gynecol Pathol, 1998, 17(2):129–34., DOI 10.1097/00004347-199804000-00006 Gu Y, Zhang J, Guan H. Expression of EZH2 in endometrial carcinoma and its effects on proliferation and invasion of endometrial carcinoma cells. Oncol Lett, 2017, 14(6):7191–6., DOI 10.3892/ol.2017.7171 Castilla MA, Moreno-Bueno G, Romero-Perez L, Van De Vijver K, Biscuola M, Lopez-Garcia MA, et al. Micro-RNA signature of the epithelial-mesenchymal transition in endometrial carcinosarcoma. J Pathol, 2011, 223(1):72–80., DOI 10. 1002/PATH.2802 Chiyoda T, Tsuda H, Tanaka H, Kataoka F, Nomura H, Nishimura S, et al. Expression profiles of carcinosarcoma of the uterine corpus—are these similar to carcinoma or sarcoma? Genes, Chromosom Cancer, 2012, 51(3):229–39., DOI 10. 1002/GCC.20947 McCluggage WG. Uterine carcinosarcomas, malignant mixed Mullerian tumors, are metaplastic carcinomas. Int J Gynecol Cancer, 2002, 12(6):687–90., DOI 10.1046/j.1525-1438.2002.01151.x Kanthan R, Senger JLB, Diudea D. Malignant mixed Mullerian tumors of the uterus: histopathological evaluation of cell cycle and apoptotic regulatory proteins. World J Surg Oncol, 2010, 8:60., DOI 10.1186/1477-7819-8-60 Kobel M, Kang EY. The many uses of p53 immunohistochemistry in gynecological pathology: proceedings of the ISGyP companion society session at the 2020 USCAP annual9 meeting. Int J Gynecol Pathol, 2021, 40(1):32–40., DOI 10. 1097/PGP.0000000000000725 Rabban JT, Blake Gilks C, Malpica A, Matias-Guiu X, Mittal K, Mutter GL, et al. Issues in the differential diagnosis of uterine low-grade endometrioid carcinoma, including mixed endometrial carcinomas: recommendations from the international society of gynecological pathologists. Int J Gynecol Pathol, 2019, 38(1):S25–S39., DOI 10.1097/PGP.0000000000000512 Kunc M, Gabrych A, Rekawiecki B, Gorczynski A, Haybaeck J, Biernat W, et al. Immunohistochemical evaluation of mismatch repair proteins and p53 expression in extrauterine carcinosarcoma/sarcomatoid carcinoma. Wspolczesna Onkol, 2020, 24(1):1–4., DOI 10.5114/WO.2020.94718 Liu FS, Kohler MF, Marks JR, Bast RC, Jr, Boyd J, Berchuck A. Mutation and overexpression of the P53 tumor suppressor gene frequently occurs in uterine and ovarian sarcomas. Obstet Gynecol, 1994, 83(1):118–24. De Jong RA, Nijman HW, Wijbrandi TF, Reyners AK, Boezen HM, Hollema H. Molecular markers and clinical behavior of uterine carcinosarcomas: focus on the epithelial tumor component. Mod Pathol, 2011, 24(10):1368–79., DOI 10.1038/ MODPATHOL.2011.88 Kobel M, Piskorz AM, Lee S, Lui S, LePage C, Marass F, et al. Optimized p53 immunohistochemistry is an accurate predictor of TP53 mutation in ovarian carcinoma. J Pathol Clin Res, 2016, 2(4):247–58., DOI 10.1002/CJP2.53 da Costa LT, Dos Anjos LG, Kagohara LT, Torrezan GT, De Paula CAA, Baracat EC, et al. The mutational repertoire of uterine sarcomas and carcinosarcomas in a Brazilian cohort: a preliminary study. Clinics, 2021, 76: e2324–15., DOI 10.6061/CLINICS/2021/E2324 Zhao S, Bellone S, Lopez S, Thakral D, Schwab C, English DP, et al. Mutational landscape of uterine and ovarian carcinosarcomas implicates histone genes in epithelial-mesenchymal transition. Proc Natl Acad Sci U S A, 2016, 113(43): 12238–43., DOI 10.1073/pnas.1614120113 Taylor NP, Zighelboim I, Huettner PC, Powell MA, Gibb RK, Rader JS, et al. DNA mismatch repair and TP53 defects are early events in uterine carcinosarcoma tumorigenesis. Mod Pathol, 2006, 19:1333–8., DOI 10.1038/modpathol.3800654 Jones S, Stransky N, McCord CL, Cerami E, Lagowski J, Kelly D, et al. Genomic analyses of gynaecologic carcinosarcomas reveal frequent mutations in chromatin remodelling genes. Nat Commun, 2014, 5:5006., DOI 10.1038/ NCOMMS6006 McConechy MK, Ding J, CheangMCU, Wiegand K, Senz J, Tone A, et al. Use of mutation profiles to refine the classification of endometrialcarcinomas. J Pathol, 2012, 228(1):20–30., DOI 10.1002/PATH.4056 Di Fiore R, Suleiman S, Drago-Ferrante R, Subbannayya Y, Vasileva-Slaveva M, Yordanov A, et al. The role of FBXW7 in gynecologic malignancies. Cells, 2023, 12(10):1415., DOI 10.3390/cells12101415 Cuevas IC, Sahoo SS, Kumar A, Zhang H, Westcott J, Aguilar M, et al. Fbxw7 is a driver of uterine carcinosarcoma by promoting epithelial-mesenchymal transition. Proc Natl Acad Sci U S A, 2019, 116(51):25880–90., DOI 10.1073/pnas. 1911310116 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611547 EP 1611557 DI 10.3389/pore.2023.1611547 PG 11 ER PT J AU Zhang, L Yuan, P Cao, Q Mu, J Ying, J Guo, Ch AF Zhang, Letian Yuan, Pei Cao, Qi Mu, Jiali Ying, Jianming Guo, Changyuan TI Expression of Concern: Case report: A rare case of tumor-to-tumor metastasis: metastatic lobular breast carcinoma to clear cell renal cell carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report AB Following publication, the authors contacted the Editorial Office stating that the findings reported in the article are no longer supported by the analyses. With this notice, Pathology and Oncology Research states its awareness of concerns regarding “A rare case of tumor-to-tumor metastasis: metastatic lobular breast carcinoma to clear cell renal cell carcinoma” published on 12 June 2023. This expression of concern has been posted while Pathology and Oncology Research awaits further clarification from the authors. It will be updated accordingly after that time. C1 [Zhang, Letian] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Yuan, Pei] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Cao, Qi] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Mu, Jiali] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Ying, Jianming] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. [Guo, Changyuan] Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of PathologyBeijing, China. RP Guo, Ch (reprint author), Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Department of Pathology, Beijing, China. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611563 EP 1611563 DI 10.3389/pore.2023.1611563 PG 1 ER PT J AU Pathology and Oncology Research, EO AF Pathology and Oncology Research, Editorial Office TI Retraction: Frequencies of Porphyromonas gingivalis detection in oral-digestive tract tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Retraction AB The journal retracts the 1 April 2021 article cited above. Following publication, concerns were raised regarding data misrepresentation. Following provision of raw data by the authors, the Editor in Chief concluded that the article’s conclusions and assertions were not sufficiently supported by the findings from the material provided; therefore, the article has been retracted. The authors do not agree to this retraction. C1 [Pathology and Oncology Research, Editorial Office] Frontiers Media SALausanne, Switzerland. RP Pathology and Oncology Research, EO (reprint author), Frontiers Media SA, Lausanne, Switzerland. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611616 EP 1611616 DI 10.3389/pore.2023.1611616 PG 1 ER PT J AU Editorial Office, PaOR AF Editorial Office, Pathology and Oncology Research TI Retraction: Associations of porphyromonas gingivalis infection and low beclin1 expression with clinicopathological parameters and survival of esophageal squamous cell carcinoma patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Retraction AB The journal retracts the 08 December 2021 article cited above. Following publication, concerns were raised regarding data misrepresentation. In particular, multiple discrepancies were identified in the flow cytometry graphs, western blots and bar graphs in Figure 4. Following provision of raw data by the authors, the Editor in Chief concluded that the article’s conclusions and assertions were not sufficiently supported by the findings from the material provided; therefore, the article has been retracted. The authors do not agree to this retraction. C1 [Editorial Office, Pathology and Oncology Research] Frontiers Media SALausanne, Switzerland. RP Editorial Office, PaOR (reprint author), Frontiers Media SA, Lausanne, Switzerland. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD NOV PY 2023 VL 29 IS 4 BP 1611617 EP 1611617 DI 10.3389/pore.2023.1611617 PG 1 ER PT J AU Hu, L Tiesinga, J AF Hu, Lina Tiesinga, James TI Case report: Primary vulvar adenocarcinoma of mammary gland type—its genetic characteristics by focused next-generation sequencing SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE MLVA; mutations by NGS; immune markers; triple positive; immunohistochemical features ID MLVA; mutations by NGS; immune markers; triple positive; immunohistochemical features AB Mammary-like vulvar adenocarcinoma (MLVA) is an exceedingly rare subtype of vulvar adenocarcinoma that shares features with mammary gland tissue. Due to its rarity and lack of consensus, MLVA presents diagnostic challenges to pathologists. We present the case of a 59-yearold female with an ulceratedmass on the right side of the external genitalia, diagnosed as MLVA. Comprehensive immunohistochemistry (IHC) and gene sequencing studies were performed to characterize the tumor. IHC analysis revealed triple expression of hormonal receptors (estrogen receptor, progesterone receptor, and HER2), supporting the mammary gland origin of the tumor. Gene sequencing identified unique genetic mutations associated with the expression of hormonal markers. One fusion gene (ERBB2-NAGLU) has not been reported in any tumors, and other mutations with unique mutation types have not been previously reported in MLVA. Our findings shed light on the molecular characteristics of MLV and may help improve the diagnosis and treatment of this rare type of vulvar adenocarcinoma. C1 [Hu, Lina] Alaska Native Medical Center, Department of PathologyAnchorage, AK, USA. [Tiesinga, James] Alaska Native Medical Center, Department of PathologyAnchorage, AK, USA. RP Hu, L (reprint author), Alaska Native Medical Center, Department of Pathology, Anchorage, USA. EM lxhu@anthc.org CR Grewal JK, Eirew P, Jones M, Chiu K, Tessier-Cloutier B, Karnezis AN, et al. Detection and genomic characterization of a mammary-like adenocarcinoma. Cold Spring Harb Mol Case Stud, 2017, 3(6):a002170., DOI 10.1101/mcs.a002170 Stueben BL, Lara JF. Primary adnexal adenocarcinoma of the vulva: a diagnosis of exclusion based on location, immunohistochemistry, and pattern of spread. Gynecol Oncol Case Rep, 2013, 4:7–8., DOI 10.1016/j.gynor.2012.11.002 Niakan S, Love H, Cao Q, Kawar N. Primary invasive lobular carcinoma arising in mammary-like glands of the vulva managed with neoadjuvant trastuzumabbased chemotherapy, excision, and sentinel lymph node biopsy. Clin case Rep, 2021, 9(1):118–22., DOI 10.1002/ccr3.3475 Kalwiba C, Lunn J, Wu etc H. Adenocarcinoma of mammary-like glands of the vulva: a case report and literature review. South Afr J Gynaecol Oncol, 2022, 14(1). Moraisl M, Vaz Silva J, Tavares MV. Diagnosis and management of primary vulvar adenocarcinoma of mammary gland type: report of two distinct cases. BMJ case Rep, 2022, 15(6):e245580., DOI 10.1136/bcr-2021-245580 Tessier-Cloutier B, Asleh-Aburaya K, Shah V, McCluggage WG, Tinker A, Gilks CB. Molecular subtyping of mammary-like adenocarcinoma of the vulva shows molecular similarity to breast carcinomas. Histopathology, 2017, 71(3): 446–52., DOI 10.1111/his.13239 Veeraraghavan J, Tan Y, Cao X-X, Kim JA, Wang X, Chamness GC, et al. Recurrent ESR1-CCDC170 rearrangements in an aggressive subset of oestrogen receptor-positive breast cancers. Nat Commun, 2014, 5:4577., DOI 10.1038/ ncomms5577 Li L, Lin L, Veeraraghavan J, Hu Y, Wang X, Lee S, et al. Therapeutic role of recurrent ESR1-CCDC170 gene fusions in breast cancer endocrine resistance. Breast Cancer Res, 2020, 22(84):84–15., DOI 10.1186/s13058-020-01325-3 Vitale SR, Ruigrok-Ritstier K, Timmermans AM, Foekens R, Trapman-Jansen AMAC, Beaufort CM, et al. The prognostic and predictive value of ESR1 fusion gene transcripts in primary breast cancer. BMC Cancer, 2022, 165:165–16., DOI 10.1186/ s12885-022-09265-1 Zardavas D, Phillips WA, Loi S. PIK3CA mutations in breast cancer: reconciling findings from preclinical and clinical data. Breast Cancer Res, 2014, 16(1):201., DOI 10.1186/bcr3605 Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature, 2012, 490:61–70., DOI 10.1038/nature11412 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611376 EP 1611381 DI 10.3389/pore.2024.1611376 PG 6 ER PT J AU Vargova, D Kolkova, Z Dargaj, J Bris, L Luptak, J Dankova, Z Franova, S Svihra, J Slavik, P Sutovska, M AF Vargova, Daniela Kolkova, Zuzana Dargaj, Jan Bris, Lukas Luptak, Jan Dankova, Zuzana Franova, Sona Svihra, Jan Slavik, Pavol Sutovska, Martina TI Analysis of HIF-1α expression and genetic polymorphisms in human clear cell renal cell carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HIF-1α; SNP; RCC; tumor; kidney ID HIF-1α; SNP; RCC; tumor; kidney AB Introduction: Clear cell renal cell carcinoma (ccRCC) is mostly diagnosed incidentally and has relatively high recurrence rates. Alterations in VHL/HIF and mTOR pathways are commonly present in ccRCC. The present study attempted to identify potential diagnostic markers at the biochemical and molecular level. Methods: In total, 54 subjects (36 patients with ccRCC and 18 cancer-free controls) were enrolled. ELISA was used to measure the levels of HIF-1α in the tumor and healthy kidney tissue. The association between five selected SNPs (rs779805, rs11549465, rs2057482, rs2295080 and rs701848) located in genes of pathologically relevant pathways (VHL/HIF and mTOR) and the risk of ccRCC in the Slovak cohort was studied using real-time PCR. Results: Significant differences in HIF-1α tissue levels were observed between the tumor and healthy kidney tissue (p < 0.001). In the majority (69%) of cases, the levels of HIF-1α were higher in the kidney than in the tumor. Furthermore, the concentration of HIF-1α in the tumor showed a significant positive correlation with CCL3 and IL-1β (p (R2) 0.007 (0.47); p (R2) 0.011 (0.38). No relationship between intratumoral levels of HIF-1α and clinical tumor characteristics was observed. Rs11549465, rs2057482 in the HIF1A gene did not correlate with the expression of HIF-1α either in the tumor or in the normal kidney. None of the selected SNPs has influenced the susceptibility to ccRCC. Conclusion: More research is neccesary to elucidate the role of HIF-1α in the pathogenesis of ccRCC and the association between selected SNPs and susceptibility to this cancer. C1 [Vargova, Daniela] Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Department of PharmacologyMartin, Slovakia. [Kolkova, Zuzana] Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Biomedical Center MartinMartin, Slovakia. [Dargaj, Jan] Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Department of UrologyMartin, Slovakia. [Bris, Lukas] Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Department of UrologyMartin, Slovakia. [Luptak, Jan] Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Department of UrologyMartin, Slovakia. [Dankova, Zuzana] Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Biomedical Center MartinMartin, Slovakia. [Franova, Sona] Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Department of PharmacologyMartin, Slovakia. [Svihra, Jan] Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Department of UrologyMartin, Slovakia. [Slavik, Pavol] Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Department of Pathological AnatomyMartin, Slovakia. [Sutovska, Martina] Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Department of PharmacologyMartin, Slovakia. RP Vargova, D (reprint author), Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Department of Pharmacology, Martin, Slovakia. EM vargova238@uniba.sk CR Warren AY, Harrison D. WHO/ISUP classification, grading and pathological staging of renal cell carcinoma: standards and controversies. World J Urol, 2018, 36: 1913–26., DOI 10.1007/s00345-018-2447-8 Chow W-H, Dong LM, Devesa SS. Epidemiology and risk factors for kidney cancer. Nat Rev Urol, 2010, 7:245–57., DOI 10.1038/nrurol.2010.46 Vasudev NS, Wilson M, Stewart GD, Adeyoju A, Cartledge J, Kimuli M, et al. Challenges of early renal cancer detection: symptom patterns and incidental diagnosis rate in a multicentre prospective UK cohort of patients presenting with suspected renal cancer. BMJ Open, 2020, 10:e035938., DOI 10. 1136/bmjopen-2019-035938 Makhov P, Joshi S, Ghatalia P, Kutikov A, Uzzo RG, Kolenko VM. Resistance to systemic therapies in clear cell renal cell carcinoma: mechanisms and management strategies. Mol Cancer Ther, 2018, 17:1355–64., DOI 10.1158/1535-7163.MCT-17-1299 Kuthi L, Jenei A, Hajdu A, Nemeth I, Varga Z, Bajory Z, et al. Prognostic factors for renal cell carcinoma subtypes diagnosed according to the 2016 WHO renal tumor classification: a study involving 928 patients. Pathol Oncol Res, 2017, 23: 689–98., DOI 10.1007/s12253-016-0179-x Kim SH, Park B, Hwang EC, Hong S-H, Jeong CW, Kwak C, et al. Retrospective multicenter long-term follow-up analysis of prognostic risk factors for recurrencefree, metastasis-free, cancer-specific, and overall survival after curative nephrectomy in non-metastatic renal cell carcinoma. Front Oncol, 2019, 9:859., DOI 10.3389/fonc.2019.00859 Nabi S, Kessler ER, Bernard B, Flaig TW, Lam ET. Renal cell carcinoma: a review of biology and pathophysiology. F1000Res, 2018, 7:307., DOI 10.12688/ f1000research.13179.1 Sato Y, Yoshizato T, Shiraishi Y, Maekawa S, Okuno Y, Kamura T, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet, 2013, 45: 860–7., DOI 10.1038/ng.2699 Hoefflin R, Harlander S, Schafer S, Metzger P, Kuo F, Schonenberger D, et al. HIF-1α and HIF-2α differently regulate tumour development and inflammation of clear cell renal cell carcinoma in mice. Nat Commun, 2020, 11:4111., DOI 10.1038/ s41467-020-17873-3 Gudas LJ, Fu L, Minton DR, Mongan NP, Nanus DM. The role of HIF1α in renal cell carcinoma tumorigenesis. J Mol Med, 2014, 92:825–36., DOI 10.1007/ s00109-014-1180-z Shen C, Kaelin WG. The VHL/HIF axis in clear cell renal carcinoma. Semin Cancer Biol, 2013, 23:18–25., DOI 10.1016/j.semcancer.2012.06.001 Klatte T, Seligson DB, Riggs SB, Leppert JT, Berkman MK, Kleid MD, et al. Hypoxia-inducible factor 1 alpha in clear cell renal cell carcinoma. Clin Cancer Res, 2007, 13:7388–93., DOI 10.1158/1078-0432.CCR-07-0411 Fan Y, Li H, Ma X, Gao Y, Chen L, Li X, et al. Prognostic significance of hypoxia-inducible factor expression in renal cell carcinoma: a PRISMA-compliant systematic review and meta-analysis. Medicine, 2015, 94:e1646., DOI 10.1097/MD. 0000000000001646 Balamurugan K. HIF-1 at the crossroads of hypoxia, inflammation, and cancer. Int J Cancer, 2016, 138:1058–66., DOI 10.1002/ijc.29519 Alayev A, Holz MK. mTOR signaling for biological control and cancer. J Cel Physiol, 2013, 228:1658–64., DOI 10.1002/jcp.24351 Cargnello M, Tcherkezian J, Roux PP. The expanding role of mTOR in cancer cell growth and proliferation. Mutagenesis, 2015, 30:169–76., DOI 10.1093/mutage/ geu045 Pezzicoli G, Filoni E, Gernone A, Cosmai L, Rizzo M, Porta C. Playing the devil’s advocate: should we give a second chance to mTOR inhibition in renal clear cell carcinoma? – ie strategies to revert resistance to mTOR inhibitors. Cancer Manag Res, 2021, 13:7623–36., DOI 10.2147/CMAR.S267220 Tian T, Li X, Zhang J. mTOR signaling in cancer and mtor inhibitors in solid tumor targeting therapy. Int J Mol Sci, 2019, 20:755., DOI 10.3390/ijms20030755 Enedina J-C, Maria del Pilar C-V, VictorAlberto M-A, Patricia M-B, Javier F-T, Alberto L-R, et al. Hypoxia inducible factor-1α gene rs11549465 might be protective factor for the development of type 1 diabetes mellitus. Endocrinol Diabetes Res, 2018, 04., DOI 10.4172/2470-7570.1000131 Tanimoto K, Yoshiga K, Eguchi H, Kaneyasu M, Ukon K, Kumazaki T, et al. Hypoxia-inducible factor-1alpha polymorphisms associated with enhanced transactivation capacity, implying clinical significance. Carcinogenesis, 2003, 24: 1779–83., DOI 10.1093/carcin/bgg132 OllerenshawM, Page T, Hammonds J, Demaine A. Polymorphisms in the hypoxia inducible factor-1alpha gene, HIF1A, are associated with the renal cell carcinoma phenotype. Cancer Genet Cytogenet, 2004, 153:122–6., DOI 10.1016/j.cancergencyto.2004. 01.014 Qin C, Cao Q, Ju X,Wang M,Meng X, Zhu J, et al. The polymorphisms in the VHL and HIF1A genes are associated with the prognosis but not the development of renal cell carcinoma. Ann Oncol, 2012, 23:981–9., DOI 10.1093/annonc/mdr325 Cao Q, Ju X, Li P, Meng X, Shao P, Cai H, et al. A functional variant in the MTOR promoter modulates its expression and is associated with renal cell cancer risk. PLoS One, 2012, 7:e50302., DOI 10.1371/journal.pone.0050302 Qi G-H, Wang C-H, Zhang H-G, Yu J-G, Ding F, Song Z-C, et al. Comprehensive analysis of the effect of rs2295080 and rs2536 polymorphisms within the mTOR gene on cancer risk. Biosci Rep, 2020, 40., DOI 10.1042/BSR20191825 Zhang Z, Chen Q, Zhang J, Wang Y, Hu X, Yin S, et al. Associations of genetic polymorphisms in pTEN/AKT/mTOR signaling pathway genes with cancer risk: a metaanalysis in Asian population. Sci Rep, 2017, 7:17844., DOI 10.1038/s41598-017-17250-z Lv C, Bai Z, Liu Z, Luo P, Zhang J. Renal cell carcinoma risk is associated with the interactions of APOE, VHL and MTHFR gene polymorphisms. Int J Clin Exp Pathol, 2015, 8:5781–6. van de Pol JAA, van den Brandt PA, van Engeland M, Godschalk RWL, van Schooten FJ, Hogervorst JGF, et al. Germline polymorphisms in the Von Hippel-Lindau andHypoxia-inducible factor 1-alpha genes, gene-environment and gene-gene interactions and renal cell cancer. Sci Rep, 2020, 10:137., DOI 10.1038/s41598-019-56980-0 Wang W-C, Tsou M-H, Chen H-J, Hsu W-F, Lai Y-C. Two single nucleotide polymorphisms in the von Hippel-Lindau tumor suppressor gene in Taiwanese with renal cell carcinoma. BMC Res Notes, 2014, 7:638., DOI 10.1186/1756-0500-7-638 Bensouilah FZ, Chellat-Rezgoune D, Garcia-Gonzalez MA, Carrera N, Abadi N, DahdouhA, et al. Association of single nucleotide polymorphisms with renal cell carcinoma in Algerian population. Afr J Urol, 2020, 26:48., DOI 10.1186/s12301-020-00055-4 Min Z, Mi Y, Lv Z, Sun Y, Tang B, Wu H, et al. Associations of genetic polymorphisms of mTOR rs2295080 T/G and rs1883965 G/A with susceptibility of urinary systemcancers. DisMarkers, 2022, 2022:1720851–16., DOI 10.1155/2022/1720851 Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO classification of tumours of the urinary system and male genital organs—Part A: renal, penile, and testicular tumours. Eur Urol, 2016, 70: 93–105., DOI 10.1016/j.eururo.2016.02.029 Vargova D, Dargaj J, Franova S, Dohal M, Luptak J, Svihra J, et al. Immunobiochemical profile of clear cell renal cell carcinoma, ccRCC): a preliminary study. Gen Biophys Physiol, 2023, 42:387–401., DOI 10.4149/gpb_2023015 Medina Villaamil V, Aparicio Gallego G, Santamarina Cainzos I, Valladares- Ayerbes M, Anton Aparicio LM. Searching for Hif1-α interacting proteins in renal cell carcinoma. Clin Translational Oncol, 2012, 14:698–708., DOI 10.1007/s12094-012-0857-4 Monzon FA, Alvarez K, Peterson L, Truong L, Amato RJ,Hernandez-McClain J, et al. Chromosome 14q loss defines a molecular subtype of clear-cell renal cell carcinoma associated with poor prognosis. Mod Pathol, 2011, 24:1470–9., DOI 10. 1038/modpathol.2011.107 Chen B, Li L, Li M, Wang X. HIF1A expression correlates with increased tumor immune and stromal signatures and aggressive phenotypes in human cancers. Cell Oncol, 2020, 43:877–88., DOI 10.1007/s13402-020-00534-4 Lidgren A, Hedberg Y, Grankvist K, Rasmuson T, Bergh A, Ljungberg B. Hypoxia-inducible factor 1alpha expression in renal cell carcinoma analyzed by tissue microarray. Eur Urol, 2006, 50:1272–7., DOI 10.1016/j.eururo.2006.05.043 Kojima H, Gu H, Nomura S, Caldwell CC, Kobata T, Carmeliet P, et al. Abnormal B lymphocyte development and autoimmunity in hypoxia-inducible factor 1alpha -deficient chimeric mice. Proc Natl Acad Sci, 2002, 99:2170–4., DOI 10. 1073/pnas.052706699 Lukashev D, Caldwell C, Ohta A, Chen P, SitkovskyM. Differential regulation of two alternatively spliced isoforms of hypoxia-inducible factor-1 alpha in activated T lymphocytes. J Biol Chem, 2001, 276:48754–63., DOI 10.1074/jbc.M104782200 Cramer T, Yamanishi Y, Clausen BE, Forster I, Pawlinski R, Mackman N, et al. HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell, 2003, 112: 645–57., DOI 10.1016/S0092-8674(03)00154-5 Islam SMT, Won J, Khan M, Mannie MD, Singh I. Hypoxia-inducible factor- 1 drives divergent immunomodulatory functions in the pathogenesis of autoimmune diseases. Immunology, 2021, 164:31–42., DOI 10.1111/imm.13335 Greten FR, Grivennikov SI. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity, 2019, 51:27–41., DOI 10.1016/j.immuni.2019.06.025 Malkov MI, Lee CT, Taylor CT. Regulation of the hypoxia-inducible factor, HIF, by pro-inflammatory cytokines. Cells, 2021, 10:2340., DOI 10.3390/cells10092340 Singh N, Baby D, Rajguru J, Patil P, Thakkannavar S, Pujari V. Inflammation and cancer. Ann Afr Med, 2019, 18:121–6., DOI 10.4103/aam.aam_56_18 Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? The Lancet, 2001, 357:539–45., DOI 10.1016/S0140-6736(00)04046-0 Hu F, Liu H, Xu L, Li Y, Liu X, Shi L, et al. Hypoxia-inducible factor-1α perpetuates synovial fibroblast interactions with T cells and B cells in rheumatoid arthritis. Eur J Immunol, 2016, 46:742–51., DOI 10.1002/eji.201545784 Rider P, Kaplanov I, Romzova M, Braiman L, Braiman A, Voronov E, et al. The transcription of the alarmin cytokine interleukin-1 alpha is controlled by hypoxia inducible factors 1 and 2 alpha in hypoxic cells. Front Immunol, 2012, 3: 290., DOI 10.3389/fimmu.2012.00290 Bhavsar I, Miller CS, Al-Sabbagh M. Macrophage inflammatory protein-1 alpha, MIP-1 alpha)/CCL3: as a biomarker. Gen Methods Biomarker Res their Appl, 2015, 2015:223–49., DOI 10.1007/978-94-007-7696-8_27 Rebe C, Ghiringhelli F. Interleukin-1β and cancer. Cancers, Basel,, 2020, 12: 1791., DOI 10.3390/cancers12071791 Storti P, Bolzoni M, Donofrio G, Airoldi I, Guasco D, Toscani D, et al. Hypoxia-inducible factor, HIF)-1α suppression in myeloma cells blocks tumoral growth in vivo inhibiting angiogenesis and bone destruction. Leukemia, 2013, 27: 1697–706., DOI 10.1038/leu.2013.24 Jung Y-J, Isaacs JS, Lee S, Trepel J, Neckers L. IL-1β mediated upregulation of HIF-lα via an NFkB/COX-2 pathway identifies HIF-1 as a critical link between inflammation and oncogenesis. FASEB J, 2003, 17:1–22., DOI 10. 1096/fj.03-0329fje Wang X, Ren H, Zhao T, Ma W, Dong J, Zhang S, et al. Single nucleotide polymorphism in themicroRNA-199a binding site of HIF1A gene is associated with pancreatic ductal adenocarcinoma risk and worse clinical outcomes. Oncotarget, 2016, 7:13717–29., DOI 10.18632/oncotarget.7263 Yamamoto Y, Kiyohara C, Ogata-Suetsugu S, Hamada N, Nakanishi Y. Association between genetic polymorphisms involved in the hypoxia-inducible factor pathway and lung cancer risk: a case-control study in Japan. Asia Pac J Clin Oncol, 2017, 13:234–42., DOI 10.1111/ajco.12640 Peckham-Gregory EC, Thapa DR, Martinson J, Duggal P, Penugonda S, Bream JH, et al. MicroRNA-related polymorphisms and non-Hodgkin lymphoma susceptibility in the Multicenter AIDS Cohort Study. Cancer Epidemiol, 2016, 45: 47–57., DOI 10.1016/j.canep.2016.09.007 AlmasiM, Besse L, Brozova L, Jarkovsky J, Bezdekova R, Pour L, et al. Selected genetic polymorphisms associated with hypoxia and multidrug resistance in monoclonal gammopathies patients. Klinicka Onkologie, 2018, 31:213–29., DOI 10.14735/amko2018213 WuL-F, XuG-P, ZhaoQ, ZhouL-J,WangD, ChenW-X. The associationbetween hypoxia inducible factor 1 subunit alpha gene rs2057482 polymorphism and cancer risk: a meta-analysis. BMC Cancer, 2019, 19:1123., DOI 10.1186/s12885-019-6329-2 Moore LE, Nickerson ML, Brennan P, Toro JR, Jaeger E, Rinsky J, et al. Von hippel-lindau, VHL, inactivation in sporadic clear cell renal cancer: associations with germline VHL polymorphisms and etiologic risk factors. Plos Genet, 2011, 7: e1002312., DOI 10.1371/journal.pgen.1002312 Miricescu D, Balan D, Tulin A, Stiru O, Vacaroiu I, Mihai D, et al. PI3K/AKT/ mTOR signalling pathway involvement in renal cell carcinoma pathogenesis, Review). Exp Ther Med, 2021, 21:540., DOI 10.3892/etm.2021.9972 The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature, 2013, 499:43–9., DOI 10. 1038/nature12222 Pantuck AJ, Seligson DB, Klatte T, Yu H, Leppert JT, Moore L, et al. Prognostic relevance of the mTOR pathway in renal cell carcinoma: implications for molecular patient selection for targeted therapy. Cancer, 2007, 109:2257–67., DOI 10.1002/cncr.22677 Fusco N, Sajjadi E, Venetis K, Gaudioso G, Lopez G, Corti C, et al. PTEN alterations and their role in cancer management: are we making headway on precision medicine? Genes, Basel,, 2020, 11:719., DOI 10.3390/genes11070719 Fan C, Zhao C, Wang F, Li S, Wang J. Significance of PTEN mutation in cellular process, prognosis, and drug selection in clear cell renal cell carcinoma. Front Oncol, 2019, 9:357., DOI 10.3389/fonc.2019.00357 Song MS, Salmena L, Pandolfi PP. The functions and regulation of the PTEN tumour suppressor. Nat Rev Mol Cel Biol, 2012, 13:283–96., DOI 10.1038/nrm3330 Lin L, Zhang Z, Zhang W, Wang L, Wang J. Roles of genetic variants in the PI3K/PTEN pathways in susceptibility to colorectal carcinoma and clinical outcomes treated with FOLFOX regimen. Int J Clin Exp Pathol, 2015, 8:13314–22. Ding J, Gao Y, Liu R, Xu F, Liu H. Association of PTEN polymorphisms with susceptibility to hepatocellular carcinoma in a Han Chinese population. DNA Cel Biol, 2011, 30:229–34., DOI 10.1089/dna.2010.1126 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611444 EP 1611453 DI 10.3389/pore.2023.1611444 PG 10 ER PT J AU Bely, M Apathy, AF Bely, Miklos Apathy, Agnes TI Crystal induced arthropathies—a comparative study of 40 patients with apatite rheumatism, chondrocalcinosis and primary synovial chondromatosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE apatite rheumatism; chondrocalcinosis; primary synovial chondromatosis; conventional stains; non-staining technique ID apatite rheumatism; chondrocalcinosis; primary synovial chondromatosis; conventional stains; non-staining technique AB Introduction: Apatite rheumatism (AR), chondrocalcinosis (Ch-C), and primary synovial chondromatosis (prSynCh) are regarded as distinct clinical entities. The introduction of the non-staining technique by Bely and Apathy (2013) opened a new era in the microscopic diagnosis of crystal induced diseases, allowing the analysis of MSU (monosodium urate monohydrate) HA (calcium hydroxyapatite), CPPD (calcium pyrophosphate dihydrate) crystals, cholesterol, crystalline liquid lipid droplets, and other crystals in unstained sections of conventionally proceeded (aqueous formaldehyde fixed, paraffinembedded) tissue samples. The aim of this study was to describe the characteristic histology of crystal deposits in AR, Ch-C, and prSynCh with traditional stains and histochemical reactions comparing with unstained tissue sections according to Bely and Apathy (2013). Patients and methods: Tissue samples of 4 with apatite rheumatism (Milwaukee syndrome), 16 with chondrocalcinosis, and 20 with clinically diagnosed primary synovial chondromatosis were analyzed. Results and conclusion: Apatite rheumatism, chondrocalcinosis, and primary synovial chondromatosis are related metabolic disorders with HA and CPPD depositions. The authors assume that AR and Ch-C are different stages of the same metabolic disorder, which differ from prSynCh in amorphous mineral production, furthermore in the production of chondroid, osteoid and/or bone. prSynCh is a defective variant of HA and CPPD induced metabolic disorders with reduced mineralization capabilities, where the deficient mineralization is replaced by chondroid and/or bone formation. The non-staining technique of Bely and Apathy proved to be a much more effective method for the demonstration of crystals in metabolic diseases than conventional stains and histochemical reactions. C1 [Bely, Miklos] St John's Hospital, Department of PathologyBudapest, Hungary. [Apathy, Agnes] St. Margaret Clinic, Department of RheumatologyBudapest, Hungary. RP Bely, M (reprint author), St John's Hospital, Department of Pathology, Budapest, Hungary. EM dr.bely.miklos@gmail.com CR Bely M, Apathy A. Monckeberg sclerosis – kristaly indukalta angiopathia, Monckeberg‟s sclerosis: crystal-induced angiopathy). Orvosi Hetilap, 2013, 154(23):908–13., DOI 10.1556/OH.2013.29628 Bely M, Apathy A. Vascular calcification and crystal deposition diseases - a comparative study on 67 amputated legs with atherosclerosis and/or Monckeberg sclerosis. EC Cardiol, 2021, 8(10):01–19. Pearse AGE. Fat soluble colorant methods. In: Pearse AGE, editor. Histochemistry theoretical and applied. Volume one: p. 129-131, Volume two: analytical technology, Lipids, lipoproteins and proteolipids. Ch. 16, p. 786-849. Edinburgh, London, Melbourne and New York: Churchill Livingstone, 1985). Swan A, Chapman B, Heap P, Dieppe P. Submicroscopic crystals in osteoarthritic synovial fluids. Ann Rheum Dis, 1994, 53(7):467–70., DOI 10.1136/ ard.53.7.467 Pay S, Terkeltaub R. Calcium pyrophosphate dihydrate and hydroxyapatite crystal deposition in the joint: new developments relevant to the clinician. Curr Rheumatol Rep, 2003, 5(3):235–43., DOI 10.1007/s11926-003-0073-x Shidham V, Chivukula M, Basir Z, Shidham G. Evaluation of crystals in formalin-fixed, paraffin-embedded tissue sections for the differential diagnosis of pseudogout, gout, and tumoral calcinosis. Mod Pathol, 2001, 14(8):806–10., DOI 10. 1038/modpathol.3880394 Forster CJ, Oglesby RJ, Szkutnik AJ, Roberts JR. Positive alizarin red clumps in Milwaukee shoulder syndrome. J Rheumatol, 2009, 36(12):2853., DOI 10.3899/ jrheum.090163 Gatter RA, Schumacher HR. “Microscopic findings under compensated polarized light and phase light” in: gatter RA. In: Schumacher HR, editor. A practical handbook of joint synovial fluid analysis. Philadelphia, London: Lea & Febiger, 1991). p. 46. Paul H, Reginato AJ, Schumacher HR. Alizarin red S staining as a screening test to detect calcium compounds in synovial fluid. Arthritis Rheum, 1983, 26(2): 191–200. PMID: 6186260., DOI 10.1002/art.1780260211 Shoji K. Alizarin red S staining of calcium compound crystals in synovial fluid. Nihon Seikeigeka Gakkai Zasshi, 1993, 67(4):201–10. PMID: 7686572. Yang JH, Oh KJ, Pandher DS. Hydroxyapatite crystal deposition causing rapidly destructive arthropathy of the hip joint. Indian J Orthopaedics, 2011, 45: 569–72., DOI 10.4103/0019-5413.87139 Bely M, Apathy A. A simple method of diagnostic pathology for identification of crystal deposits in metabolic and crystal induced diseases, 2022). Available from: http://structural-crystallography.imedpub.com/archive.php, Accessed February 15, 2016). Bely M, Apathy A. Metabolic diseases and crystal induced arthropathies technic of non-staining histologic sections - a comparative study of standard stains and histochemical reactions. Clin Arch Bone Jt Dis, 2018, 1(2)., DOI 10.23937/cabjd- 2017/1710007 Bely M, Apathy A. Crystal deposits in tissue of patients with chondrocalcinosis and apatite rheumatism – microscopic identification of CPPD and HA with the non-staining technique of Bely and Apathy. BAOJ Clin Trials, 2018, 4(1):018. Apathy A, Bely M. AB1262 amorphous mineralization, chondroid and/or osteoid formation in crystal induced metabolic disorders – a comparative study of apatite rheumatism, chondrocalcinosis and primary synovial chondromatosis apatite rheumatism, chondrocalcinosis, primary synovial chondromatosis. Ann Rheum Dis, 2023, 82(1):1857–8., DOI 10.1136/annrheumdis-2023-eular.193 Bely M, Apathy Ά. A comparative microscopic study of apatite rheumatism, chondrocalcinosis and synovial chondromatosis – HA and CPPD induced metabolic disorders. EC Pulmonology Respir Med, 2023, 12(6):01–17. Ohira T, Ishikawa K. Preservation of calcium pyrophosphate dihydrate crystals: effect of Mayer’s haematoxylin staining period. Ann Rheum Dis, 2001, 60(1):80–2. PMID 11114290., DOI 10.1136/ard.60.1.80 Carson FL. “Mayer’s hematoxylin” In: carson FL. In: Histotechnology. Chicago: ASCP Press, 1990). p. 100–5. McManus JFA, Mowry RW. Methods of general utility for the routine study of tissues, sodium Alizarin sulfonate stain for calcium and Von Kossa’s method for phosphates and carbonates. In: McManus JFA, Mowry RW, editors. Staining methods, histologic and histochemical. New York: Hoeber PB Inc, 1960). p. 55–72. Vacca LL. Alizarin red S. In: Vacca LL, editor. Laboratory manual of histochemistry. New York: Raven Press, 1985). p. 333–4. Lillie RD. Von Kossa’smethod. In: Lillie RD, editor. Histpathologic technic and practical histochemistry. New York, Toronto, London: The Blakiston Division McGraw-Hill Book Company, 1954). p. 264–5. Lentner C. “Statistical methods” volume 2. In: Lentner C, Diem K, Seldrup J, editors. Geigy scientific tables. Basle, Switzerland: Ciba-Geigy Limited, 1982). p. 227. Bely M, Apathy A. Metabolikus megbetegedesek – a kristalyok csodalatos vilaga, Metabolic disorders – the wonderful world of crystals). Orvostovabbkepzo Szemle, 2023, 9:66–81. Bely M, Apathy A. Crystal deposits in apatite rheumatism and chondrocalcinosis – microscopic identification of hydroxyapatite and calcium pyrophosphate dihydrate crystals with standard stains and histochemical reactions and with the nonstaining technique of Bely and Apathy. EC Pulmonology, 2022, 1:03–24. Bely M, Apathy A. Apatite rheumatism and chondrocalcinosis are different stages of the same metabolic disorder – a clinicopathologic study of 21 patients with clinically diagnosed apatite rheumatism or chondrocalcinosis. J Interdiscip Histopathology, 2022, 10:1–14. Reginato AM, Yuvienco C. Hydroxyapatite crystal-induced rheumatology, 2022). Available from: https://www.rheumatologyadvisor.com/home/decisionsupport- in-medicine/rheumatology/hydroxyapatite-crystal-induced/, Accessed November 15, 2022). Mohr W. Kalziumpirophosphat-arthropathie, apatitkrankheiten, primare synoviale osteochondromatose. In: Mohr W, editor. Gelenkpathologie, historische Grundlagen, Ursachen und Entwicklungen von Gelenkleiden und ihre Pathomorphologie. Berlin, Heidelberg: Springer-Verlag, 2000). p. 193–212. Reginato AJ, Reginato AM. Diseases associated with deposition of calcium pyrophosphate or hydroxyapatite. In: Ruddy S, Harris ED, Sledge CB, editors. Crystal-associated synovitis, section XV, kelly’s textbook of rheumatology. 6th ed. Philadelphia, London, New York, St. Louis, Sydney, Toronto: WB Saunders Company: A division of Harcourt Brace & Company, 2001). p. 1377–90. Ch. 90. Zitnan D, Sitaj S. Chondrocalcinosis articularis Section L clinical and radiological study. Ann Rheum Dis, 1963, 22:142–52., DOI 10.1136/ard.22.3.142 Gardner DL, McClure J. Metabolic, nutritional and endocrine diseases of connective tissue, synovial osteochondromatosis. In: Arnold E, editor. Pathological basis of the connective tissue diseases. 1st ed. London, Melbourne, Auckland: Great Britain, 1992). p. 984–6. Ch. 24. McCarty DJ, Lehr RJ, Halverson PB. Crystal populations in human synovial fluid. Identification of apatite, octacalcium phosphate, and tricalcium phosphate. Arthritis Rheum, 1983, 26:1220–4., DOI 10.1002/art.1780261008 Fassbender HG. Crystal-associated arthropathies” in Pathology and pathobiology of rheumatic diseases. 2nd ed. Berlin, Heidelberg, New York, Germany: Springer-Verlag, 2002). p. 353–69. ch. 17. Gupta SJ. Crystal induced arthritis: an overview. INJR, Indian Journal of Rheumatology - formerly). J Indian Rheumatol Assoc, 2002, 10:5–13. Rosenthal AK, Ryan LM. Calcium pyrophosphate deposition disease. New Engl J Med, 2016, 374:2575–84., DOI 10.1056/NEJMra1511117 Dieppe PA, Crocker P, Huskisson EC, Willoughby DA. Apatite deposition disease. A new arthropathy. Lancet, 1976, 307:266–9., DOI 10.1016/S0140-6736(76)91400-8 Dieppe PA. Milwaukee shoulder. Br Med J, Clin Res Ed)., 1981, 283(6305): 1488–9. PMC1507904., DOI 10.1136/bmj.283.6305.1488 Rosenthal A, Dalbeth N, Romain PL. Clinical manifestations and diagnosis of calcium pyrophosphate crystal deposition, CPPD, disease”. Wolters Kluwer Canada, 2024). Available from: https://www.uptodate.com/contents/clinicalmanifestations- and-diagnosis-of-calcium-pyrophosphate-crystal-deposition-cppddisease, Accessed November 01, 2023). Bachmann D, Resnick D. “Calcium pyrophosphate dihydrate crystal deposition disease” and “Calcium hydroxyapatite crystal deposition disease”. In: Bachmann D, Resnick D, editors. Radiological atlas of rheumatological diseases. Basel, Switzerland: Hoffmann-La Roche Ltd., 1994). p. 108–23. mayoclinic. Pseudogout, 2022). Available from: https://www.mayoclinic.org/ diseases-conditions/pseudogout/symptoms-causes/syc-20376983, Accessed July 28, 2022). Palinkas M, Poor G. Kristalyatrthritisek. In: Szekanecz Z, Nagy G, editors. Reumatologia. Budapest: Medicina konyvkiado Zrt, 2019). p. 572. Ch. 41. Bywaters EGL. Calcium pyrophosphate deposits in synovial membrane. Ann Rheum Dis, 1972, 31(3):219–21., DOI 10.1136/ard.31.3.219-b Hayes CW, Conway WF. Calcium hydroxyapatite deposition disease. RadioGraphics, 1990, 10:1031–48., DOI 10.1148/radiographics.10.6.2175444 Resnick D. Calcium hydroxyapatite crystal deposition disease. In: Resnick D, Niawayama G, editors. Diagnosis of bone and joint disorders. 2nd ed. Saunders Philadelphia, 1988). p. 1733–64. GARD. Synovial chondromatosis – gard, 2023). Available from: https:// rarediseases.info.nih.gov/diseases/6054/synovial-chondromatosis, Accessed February, 2023). AAOS. Synovial chondromatosis - OrthoInfo – AAOS, 2022). Available from: ht tps:/ /orthoinfo.aaos.org/en/diseases–conditions/synovialchondromatosis, Accessed January 2022). Bely M, Apathy A, Pinter T, Ratko J. Generalized secondary amyloidosis in rheumatoid arthritis. Acta morphologica Hungarica, 1992, 40:49–69. Bely M. “Identification of amyloid deposits by histochemical methods of romhanyi” in: bely M, guest editor. Amyloid, 2001, 8.2:177–82. Bely M, Apathy A. Clinical pathology of rheumatoid arthritis: cause of death, lethal complications and associated diseases in rheumatoid arthritis. Budapest: Akademiai Kiado, 2012). p. 1–440. Bely M, Apathy A. Formal pathogenesis of systemic and localized amyloidosis. EC Cardiol, 2019, 6(5):444–69. Wald A. Sequential analysis. New York: Wiley Mathematical Statistics Series, Chapman & Hall, 1947). Uhthoff HK, Loehr JW. Calcific tendinopathy of the rotator cuff: pathogenesis, diagnosis, and management. J Am Acad Orthopaedic Surgeons, 1997, 5(4):183–91., DOI 10.5435/00124635-199707000-00001 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611454 EP 1611468 DI 10.3389/pore.2024.1611454 PG 15 ER PT J AU Ferreira de Araujo, GM Almondes Santana Lemos, EL Negromonte Guerra, LP dos Santos Lima Didjurgeit, MF Cezar, BA Faquini, VI Cirne de Azevedo Filho, RH AF Ferreira de Araujo, Gomes Mayle Almondes Santana Lemos, Euripedes Luiz Negromonte Guerra, Lucas Pedro dos Santos Lima Didjurgeit, Marcia Fernanda Cezar, Batista Auricelio Faquini, Vilela Igor Cirne de Azevedo Filho, Rocha Hildo TI Supratentorial meningeal melanocytoma mimicking meningioma: case report and literature review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE primary melanocytic tumor; melanocytoma; brain tumor; primary brain tumor; primary meningeal melanocytomas ID primary melanocytic tumor; melanocytoma; brain tumor; primary brain tumor; primary meningeal melanocytomas AB Introduction: Primary melanocytic tumors originating from CNS melanocytes are rare, with a low incidence of 0.7 cases per 10 million annually. This study focuses on primary leptomeningeal melanocytomas, emphasizing their epidemiology, clinical characteristics, and diagnostic challenges. Despite their infrequency, these tumors warrant attention due to their unique features and potential for local recurrence. Case Report: A 32-year-old female presented with syncope and seizures, leading to the discovery of two left-sided supratentorial lesions initially misidentified as convexity meningiomas. Detailed imaging suggested meningioma-like features, but intraoperative findings revealed unexpected hyperpigmented lesions. Histopathological examination, supported by immunohistochemistry, confirmed primary leptomeningeal melanocytoma. The surgical approach and subsequent management are discussed. Discussion: The discussion emphasizes challenges in diagnosing primary leptomeningeal melanocytomas. Treatment debates, especially regarding adjuvant radiotherapy, are explored. Recurrence risks stress the importance of vigilant follow-up, advocating for complete surgical resection as the primary approach. The rarity of supratentorial cases adds complexity to diagnosis, necessitating a multidisciplinary approach. Insights from this case contribute to understanding and managing primary leptomeningeal melanocytomas, addressing challenges in differentiation from more common tumors and prompting ongoing research for refined diagnostics and optimized treatments. Conclusion: This study contributes insights into primary leptomeningeal melanocytomas, highlighting their rarity in supratentorial regions. The case underscores the importance of a multidisciplinary approach, incorporating clinical, radiological, and histopathological expertise for accurate diagnosis and tailored management. Ongoing research is crucial to refine treatment strategies, enhance prognostic precision, and improve outcomes for individuals with this uncommon CNS neoplasm. C1 [Ferreira de Araujo, Gomes Mayle] Hospital da Restauracao, Department of NeurosurgeryRecife, Brazil. [Almondes Santana Lemos, Euripedes Luiz] Hospital da Restauracao, Department of NeurosurgeryRecife, Brazil. [Negromonte Guerra, Lucas Pedro] Universidade Federal de Pernambuco, Center for Medical SciencesRecife, Brazil. [dos Santos Lima Didjurgeit, Marcia Fernanda] Hospital da Restauracao, Department of NeurosurgeryRecife, Brazil. [Cezar, Batista Auricelio] Hospital da Restauracao, Department of NeurosurgeryRecife, Brazil. [Faquini, Vilela Igor] Hospital da Restauracao, Department of NeurosurgeryRecife, Brazil. [Cirne de Azevedo Filho, Rocha Hildo] Hospital da Restauracao, Department of NeurosurgeryRecife, Brazil. RP Negromonte Guerra, LP (reprint author), Universidade Federal de Pernambuco, Center for Medical Sciences, Recife, Brazil. EM pedronegromontee@gmail.com CR Brat DJ, Giannini C, Scheithauer BW, Burger PC. Primary melanocytic neoplasms of the central nervous systems. Am J Surg Pathol, 1999, 23(7): 745–54., DOI 10.1097/00000478-199907000-00001 Goldgeier MH, Klein LE, Klein-Angerer S, Moellmann G, Nordlund JJ. The distribution of melanocytes in the leptomeninges of the human brain. J Invest Dermatol, 1984, 82:235–8., DOI 10.1111/1523-1747.ep12260111 Chukwueke U, Batchelor T, Brastianos P. Management of brain metastases in patients with melanoma. J Oncol Pract, 2016, 12:536–42., DOI 10.1200/JOP.2016.011882 Varela-Poblete J, Vidal-Tellez A, Cruz-Quiroga JP, Montoya-Salvadores F, Medina-Escobar J. Melanocytic lesions of the central nervous system: a case series. Arq Neuropsiquiatr, 2022, 80(2):153–60., DOI 10.1590/0004-282X-ANP-2021-0082 Brat DJ, Perry A. “Melanocytic lesions. Chapter 10.” In: Louis DN, Ohgaki H, Wiestler OD, Cavenee WK WHO classification of tumors of the central nervous system. Lyon: International Agency for Research on Cancer, 2007). pp. 181. Quillo-Olvera J, Uribe-Olalde JS, Alcantara-Gomez LA, Rejon-Perez JD, Palomera- Gomez HG. Melanoma maligno primario del sistema nervioso central: un reto diagnostico. Cir Cir, 2015, 83(2):129–34., DOI 10.1016/j.circir.2015.04.008 Hsieh YY, Yang ST, Li WH, Hu CJ, Wang LS. Primary leptomeningeal melanoma mimicking meningitis: a case report and literature review. J Clin Oncol, 2015, 33(12):e57–e61., DOI 10.1200/JCO.2013.50.0264 Honigberg MC, Papavassiliou E, Cohen YZ. Primary leptomeningeal melanocytosis presenting as chronic meningitis. J Clin Neurosci, 2014, 21(6): 1056–8., DOI 10.1016/j.jocn.2013.10.014 Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol, 2021, 23(8):1231–51., DOI 10.1093/neuonc/noab106 Beseoglu K, Knobbe CB, Reifenberger G, Steiger HJ, Stummer W. Supratentorial meningeal melanocytoma mimicking a convexity meningioma. Acta Neurochir, Wien,, 2006, 148(4):485–90., DOI 10.1007/s00701-005-0705-1 Lin B, Yang H, Qu L, Li Y, Yu J. Primary meningeal melanocytoma of the anterior cranial fossa: a case report and review of the literature. World J Surg Oncol, 2012, 10:135., DOI 10.1186/1477-7819-10-135 Kusters-Vandevelde HV, Kusters B, van Engen-van Grunsven AC, Groenen PJ, Wesseling P, Blokx WA. Primary melanocytic tumors of the central nervous system: a review with focus on molecular aspects. Brain Pathol, 2015, 25(2):209–26., DOI 10.1111/bpa.12241 Liubinas SV, Maartens N, Drummond KJ. Primary melanocytic neoplasms of the central nervous system. Clin Neurosci, 2010, 17:1227–32., DOI 10.1016/j.jocn. 2010.01.017 Winston KR, Sortel A, Schnitt SJ. Meningeal melanocytoma. Case report and review of the clinical and histological features. J Neurosurg, 1987, 66:50–7., DOI 10. 3171/jns.1987.66.1.0050 Kurita H, Segawa H, Shin M, Ueki K, Ichi S, Sasaki T, et al. Radiosurgery of meningeal melanocytoma. J Neurooncol, 2000, 46:57–61., DOI 10.1023/a: 1006335616839 Limas C, Tio FO. Meningeal melanocytoma, "melanotic meningioma"). Its melanocytic origin as revealed by electron microscopy. Cancer, 1972, 30: 1286–94., DOI 10.1002/1097-0142(197211)30:5<1286::aid-cncr2820300522>3. 0.co;2-v Hamasaki O, Nakahara T, Sakamoto S, Kutsuna M, Sakoda K. Intracranial meningeal melanocytoma. Neurol Med Chir, Tokyo,, 2002, 42(11):504–9., DOI 10. 2176/nmc.42.504 Kinnen F, Fleck SK, Baldauf J, Hans V, Daeschlein G, Rathmann E, et al. Primary leptomeningeal melanocytic tumors of the spine: report of two cases and review of the literature. World Neurosurg, 2019, 124(19):228–36. S1878-8750., DOI 10.1016/j.wneu.2019.01.015 Urtatiz O, Van Raamsdonk CD. Gnaq and Gna11 in the endothelin signaling pathway and melanoma. Front Genet, 2016, 7:59., DOI 10.3389/fgene. 2016.00059 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611482 EP 1611488 DI 10.3389/pore.2023.1611482 PG 7 ER PT J AU Batar, P Alizadeh, H Rokszin, Gy Abonyi-Toth, Zs Demeter, J AF Batar, Peter Alizadeh, Hussain Rokszin, Gyorgy Abonyi-Toth, Zsolt Demeter, Judit TI Comorbidities and outcomes of patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors: a real-world, nationwide, retrospective study from Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chronic myeloid leukemia; real-word evidence; tyrosine kinase inhibitors; treatment outcome; overall survival ID chronic myeloid leukemia; real-word evidence; tyrosine kinase inhibitors; treatment outcome; overall survival AB Purpose: This study aimed to provide real-world evidence on the characteristics, treatment patterns, and outcomes of patients with chronic myeloid leukemia (CML) receiving tyrosine kinase inhibitor (TKI) treatment in Hungary between 2011 and 2019. Patients and methods: This nationwide, retrospective study included patients who were newly diagnosed with CML in Hungarian clinical practice between January 2011 and December 2019. The analysis was based on the reimbursed prescription claims for imatinib, bosutinib, dasatinib, nilotinib, or ponatinib with the ICD-10 code C9210 in a public pharmacy between January 2009 and December 2019 using data from the National Health Insurance Fund (NHIF) of Hungary. CML incidence and prevalence, TKI treatment patterns, comorbidities, and overall survival (OS) were examined. Results: Between 2011 and 2019, altogether 1,407 patients were diagnosed with CML, with an annual average of 156 patients. The number of patients newly initiating first-line TKI therapy for CML significantly increased between 2011 and 2019 (2011: n = 136 vs. 2019: n = 191; p = 0.0043). Nilotinib was typically prescribed for younger patients (≤64 years), while older patients (≥65 years) mostly received imatinib. The most common comorbidity of CML patients was hypertension, and the proportion of patients with other malignancies was relatively high in all treatment groups. 5-year OS was 77.1% during the whole study period. Patients initiating first-line TKI treatment for CML in 2015 had significantly better 4-year OS compared to those starting treatment in 2011 (82.4% vs. 73.5%, respectively, (HR 0.53 (95%CI 0.32–0.87) p = 0.0118). Conclusion: This study is the first to provide insights into the characteristics, treatment patterns, and outcomes of CML patients treated with TKIs in Hungarian clinical practice between 2011 and 2019. We found slightly lower OS rates compared to other European countries, however, there was a statistically significant improvement in 4-year OS during the study period. The management of CML was in line with international guidelines and recommendations. C1 [Batar, Peter] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Alizadeh, Hussain] University of Pecs, Medical School, Clinical Center, 1st Department of Medicine, Division of HaematologyPecs, Hungary. [Rokszin, Gyorgy] RxTarget LtdSzolnok, Hungary. [Abonyi-Toth, Zsolt] RxTarget LtdSzolnok, Hungary. [Demeter, Judit] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. RP Batar, P (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM pbatar@med.unideb.hu CR Thielen N, Visser O, Ossenkoppele G, Janssen J. Chronic myeloid leukemia in The Netherlands: a population-based study on incidence, treatment, and survival in 3585 patients from 1989 to 2012. Eur J Haematol, 2016, 97(2):145–54., DOI 10.1111/ ejh.12695 Jabbour E, Kantarjian H. Chronic myeloid leukemia: 2018 update on diagnosis, therapy and monitoring. Am J Hematol, 2018, 93(3):442–59., DOI 10. 1002/ajh.25011 Iqbal N, Iqbal N. Imatinib: a breakthrough of targeted therapy in cancer. Chemother Res Pract, 2014, 2014:357027., DOI 10.1155/2014/357027 Pan P, Wang L, Wang Y, Shen L, Zheng P, Bi C, et al. Systematic review and meta-analysis of -New-Generation tyrosine kinase inhibitors versus imatinib for newly diagnosed chronic myeloid leukemia. Acta Haematol, 2020, 143(3):204–16., DOI 10.1159/000501537 Hochhaus A, BaccaraniM, Silver RT, Schiffer C, Apperley JF, Cervantes F, et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia, 2020, 34(4):966–84., DOI 10.1038/s41375-020-0776-2 Hehlmann R, Muller MC, Lauseker M, Hanfstein B, Fabarius A, Schreiber A, et al. Deep molecular response is reached by the majority of patients treated with imatinib, predicts survival, and is achieved more quickly by optimized high-dose imatinib: results from the randomized CML-study IV. J Clin Oncol, 2014, 32(5): 415–23., DOI 10.1200/JCO.2013.49.9020 Sasaki K, Strom SS, O’Brien S, Jabbour E, Ravandi F, Konopleva M, et al. Relative survival in patients with chronic-phase chronic myeloid leukaemia in the tyrosine-kinase inhibitor era: analysis of patient data from six prospective clinical trials. Lancet Haematol, 2015, 2(5):e186–93., DOI 10.1016/S2352- 3026(15)00048-4 Webster J, Smith BD. The case for real-world evidence in the future of clinical research on chronic myeloid leukemia. Clin Ther, 2019, 41(2):336–49., DOI 10.1016/ j.clinthera.2018.12.013 Goldberg SL, Cortes JE, Gambacorti-Passerini C, Hehlmann R, Khoury HJ, Michallet M, et al. First-line treatment selection and early monitoring patterns in chronic phase-chronic myeloid leukemia in routine clinical practice: SIMPLICITY. Am J Hematol, 2017, 92(11):1214–23., DOI 10.1002/ajh.24887 Hoffmann VS, Baccarani M, Lindoerfer D, Castagnetti F, Turkina A, Zaritsky A, et al. The EUTOS prognostic score: review and validation in 1288 patients with CML treated frontline with imatinib. Leukemia, 2013, 27(10):2016–22., DOI 10. 1038/leu.2013.171 Hoffmann VS, Baccarani M, Hasford J, Lindoerfer D, Burgstaller S, Sertic D, et al. The EUTOS population-based registry: incidence and clinical characteristics of 2904 CML patients in 20 European Countries. Leukemia, 2015, 29(6):1336–43., DOI 10.1038/leu.2015.73 Hasford J, Baccarani M, Hoffmann V, Guilhot J, Saussele S, Rosti G, et al. Predicting complete cytogenetic response and subsequent progression-free survival in 2060 patients with CML on imatinib treatment: the EUTOS score. Blood, 2011, 118(3):686–92., DOI 10.1182/blood-2010-12-319038 Dahlen T, Edgren G, Lambe M, Hoglund M, Bjorkholm M, Sandin F, et al. Cardiovascular events associated with use of tyrosine kinase inhibitors in chronic myeloid leukemia: a population-based cohort study. Ann Intern Med, 2016, 165(3): 161–6., DOI 10.7326/M15-2306 Smith BD, Liu J, Latremouille-Viau D, Guerin A, Fernandez D, Chen L. Treatment patterns, overall survival, healthcare resource use and costs in elderly Medicare beneficiaries with chronic myeloid leukemia using second-generation tyrosine kinase inhibitors as second-line therapy. Curr Med Res Opin, 2016, 32(5): 817–27., DOI 10.1185/03007995.2016.1140030 Geelen IGP, Thielen N, Janssen JJWM, Hoogendoorn M, Roosma TJA, Willemsen SP, et al. Treatment outcome in a population-based, ’real-world’ cohort of patients with chronic myeloid leukemia. Haematologica, 2017, 102(11):1842–9., DOI 10.3324/haematol.2017.174953 Hoglund M, Sandin F, Hellstrom K, Bjoreman M, Bjorkholm M, Brune M, et al. Tyrosine kinase inhibitor usage, treatment outcome, and prognostic scores in CML: report from the population-based Swedish CML registry. Blood, 2013, 122(7):1284–92., DOI 10.1182/blood-2013-04-495598 Gunnarsson N, Sandin F, HoglundM, Stenke L, BjorkholmM, LambeM, et al. Population-based assessment of chronic myeloid leukemia in Sweden: striking increase in survival and prevalence. Eur J Haematol, 2016, 97(4):387–92., DOI 10. 1111/ejh.12743 Beinortas T, Tavorienė I, Zvirblis T, Gerbutavicius R, Jurgutis M, Griskevicius L. Chronic myeloid leukemia incidence, survival and accessibility of tyrosine kinase inhibitors: a report from population-based Lithuanian haematological disease registry 2000-2013. BMC Cancer, 2016, 16:198., DOI 10.1186/s12885-016-2238-9 Castagnetti F, Di Raimondo F, De Vivo A, Spitaleri A, Gugliotta G, Fabbiano F, et al. A population-based study of chronic myeloid leukemia patients treated with imatinib in first line. Am J Hematol, 2017, 92(1):82–7., DOI 10.1002/ajh.24591 Saussele S, Krauss MP, Hehlmann R, LausekerM, Proetel U, Kalmanti L, et al. Impact of comorbidities on overall survival in patients with chronic myeloid leukemia: results of the randomized CML study IV. Blood, 2015, 126(1):42–9., DOI 10.1182/blood-2015-01-617993 Apperley JF. Chronic myeloid leukaemia. Lancet, 2015, 385(9976):1447–59., DOI 10.1016/S0140-6736(13)62120-0 Goldman JM. Chronic myeloid leukemia: a historical perspective. Semin Hematol, 2010, 47(4):302–11., DOI 10.1053/j.seminhematol.2010.07.001 O’Brien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, Cervantes F, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med, 2003, 348(11):994–1004., DOI 10.1056/NEJMoa022457 Druker BJ, Guilhot F, O’Brien SG, Gathmann I, Kantarjian H, Gattermann N, et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl JMed, 2006, 355(23):2408–17., DOI 10.1056/ NEJMoa062867 Hochhaus A, Larson RA, Guilhot F, Radich JP, Branford S, Hughes TP, et al. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med, 2017, 376(10):917–27., DOI 10.1056/NEJMoa1609324 Hehlmann R, Lauseker M, Saußele S, Pfirrmann M, Krause S, Kolb HJ, et al. Assessment of imatinib as first-line treatment of chronic myeloid leukemia: 10-year survival results of the randomized CML study IV and impact of non-CML determinants. Leukemia, 2017, 31(11):2398–406., DOI 10.1038/leu.2017.253 Bixby D, Talpaz M. Mechanisms of resistance to tyrosine kinase inhibitors in chronic myeloid leukemia and recent therapeutic strategies to overcome resistance. Hematol Am Soc Hematol Educ Program, 2009, 2009:461–76., DOI 10.1182/ asheducation-2009.1.461 Molica M, Scalzulli E, Colafigli G, Foa R, Breccia M. Insights into the optimal use of ponatinib in patients with chronic phase chronic myeloid leukaemia. Ther Adv Hematol, 2019, 10:2040620719826444., DOI 10.1177/ 2040620719826444 Cortes JE, Saglio G, Kantarjian HM, Baccarani M, Mayer J, Boque C, et al. Final 5-year study results of DASISION: the dasatinib versus imatinib study in treatment-naive chronicmyeloid leukemia patients trial. J Clin Oncol, 2016, 34(20): 2333–40., DOI 10.1200/JCO.2015.64.8899 Hochhaus A, Saglio G, Hughes TP, Larson RA, Kim DW, Issaragrisil S, et al. Long-term benefits and risks of frontline nilotinib vs imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia, 2016, 30(5):1044–54., DOI 10.1038/leu.2016.5 Cortes JE, Gambacorti-Passerini C, Deininger MW, MauroMJ, Chuah C, Kim DW, et al. Bosutinib versus imatinib for newly diagnosed chronicmyeloid leukemia: results from the randomized BFORE trial. J Clin Oncol, 2018, 36(3):231–7., DOI 10. 1200/JCO.2017.74.7162 Cortes JE, Kim DW, Pinilla-Ibarz J, le Coutre PD, Paquette R, Chuah C, et al. Ponatinib efficacy and safety in Philadelphia chromosome-positive leukemia: final 5-year results of the phase 2 PACE trial. Blood, 2018, 132(4):393–404., DOI 10.1182/ blood-2016-09-739086 Brummendorf TH, Cortes JE, Milojkovic D, Gambacorti-Passerini C, Clark RE, le Coutre PD, et al. Bosutinib, BOS, versus imatinib for newly diagnosed chronic phase, CP, chronic myeloid leukemia, CML): final 5-year results from the bfore trial. In: Presented at the 62nd ASH Annual Meeting and Exposition; December 5, 2020. Washington, DC: American Society of Hematology, 2020). Available from: https://ash.confex.com/ash/2020/webprogram/Paper137393.html, Accessed November 24, 2021). Ono T, Takahashi N, Kizaki M, Kawaguchi T, Suzuki R, Yamamoto K, et al. Prognostic effect of comorbidities in patients with chronicmyeloid leukemia treated with a tyrosine kinase inhibitor. Cancer Sci, 2020, 111(10):3714–25., DOI 10.1111/ cas.14580 Jabbour E, Makenbaeva D, Lingohr-Smith M, Lin J. Use of real-world claim databases to assess prevalence of comorbid conditions relevant to the treatment of chronic myelogenous leukemia based on national comprehensive network treatment guidelines. Clin Lymphoma Myeloma Leuk, 2015, 15(12):797–802., DOI 10.1016/j.clml.2015.09.008 Cortes J. How to manage CML patients with comorbidities. Hematol Am Soc Hematol Educ Program, 2020, 2020(1):237–42., DOI 10.1182/hematology. 2020006911 Breccia M, Latagliata R, Stagno F, Luciano L, Gozzini A, Castagnetti F, et al. Charlson comorbidity index and adult comorbidity evaluation-27 scores might predict treatment compliance and development of pleural effusions in elderly patients with chronic myeloid leukemia treated with second-line dasatinib. Haematologica, 2011, 96(10):1457–61., DOI 10.3324/haematol.2011.041251 Jain P, Kantarjian H, Boddu PC, Nogueras-Gonzalez GM, Verstovsek S, Garcia-Manero G, et al. Analysis of cardiovascular and arteriothrombotic adverse events in chronic-phase CML patients after frontline TKIs. Blood Adv, 2019, 3(6): 851–61., DOI 10.1182/bloodadvances.2018025874 Douxfils J, Haguet H, Mullier F, Chatelain C, Graux C, Dogne JM. Association between BCR-ABL tyrosine kinase inhibitors for chronic myeloid leukemia and cardiovascular events, major molecular response, and overall survival: a systematic review and meta-analysis. JAMA Oncol, 2016, 2(5):625–32., DOI 10.1001/jamaoncol. 2015.5932 Assuncao PM, Lana TP, Delamain MT, Duarte GO, Zulli R, Lorand-Metze I, et al. Cardiovascular risk and cardiovascular events in patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors. Clin Lymphoma Myeloma Leuk, 2019, 19(3):162–6., DOI 10.1016/j.clml.2018.12.004 ICON Clinical Research. Studying first line treatment of chronic myeloid leukemia, CML, in a real-world setting, SIMPLICITY,, 2022). Available from: https://clinicaltrials.gov/ct2/show/NCT01244750, Accessed January 10, 2024). World Health Organization. Global report on hypertension: the race against a silent killer, 2023). p. 152. Available from: https://www.who.int/publications/i/item/ 9789240081062, Accessed January 10, 2024). Quintas-Cardama A, Kantarjian H, Cortes J. Nilotinib-associated vascular events. Clin Lymphoma Myeloma Leuk, 2012, 12(5):337–40., DOI 10.1016/j.clml. 2012.04.005 Specchia G, Pregno P, Breccia M, Castagnetti F, Monagheddu C, Bonifacio M, et al. Prognostic factors for overall survival in chronic myeloid leukemia patients: a multicentric cohort study by the Italian CML GIMEMA network. Front Oncol, 2021, 11:739171., DOI 10.3389/fonc.2021.739171 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611497 EP 1611507 DI 10.3389/pore.2024.1611497 PG 11 ER PT J AU Dai, J Jiang, Y Hu, H Zhang, Sh Chen, Y AF Dai, Jie Jiang, Yong Hu, Haoyue Zhang, Shuang Chen, Yue TI Extracellular vesicles as modulators of glioblastoma progression and tumor microenvironment SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE extracellular vesicles; tumor microenvironment; glioblastoma; exosomes; microvesicles ID extracellular vesicles; tumor microenvironment; glioblastoma; exosomes; microvesicles AB Glioblastoma is the most aggressive brain tumor with extremely poor prognosis in adults. Routine treatments include surgery, chemotherapy, and radiotherapy; however, these may lead to rapid relapse and development of therapy-resistant tumor. Glioblastoma cells are known to communicate with macrophages, microglia, endothelial cells, astrocytes, and immune cells in the tumor microenvironment (TME) to promote tumor preservation. It was recently demonstrated that Glioblastoma-derived extracellular vesicles (EVs) participate in bidirectional intercellular communication in the TME. Apart from promoting glioblastoma cell proliferation, migration, and angiogenesis, EVs and their cargos (primarily proteins and miRNAs) can act as biomarkers for tumor diagnosis and prognosis. Furthermore, they can be used as therapeutic tools. In this review, the mechanisms of Glioblastoma-EVs biogenesis and intercellular communication with TME have been summarized. Moreover, there is discussion surrounding EVs as novel diagnostic structures and therapeutic tools for glioblastoma. Finally, unclear questions that require future investigation have been reviewed. C1 [Dai, Jie] University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Department of PathologyChengdu, China. [Jiang, Yong] Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Department of NeurosurgeryChengdu, China. [Hu, Haoyue] University of Electronic Science and Technology of China, School of Medicine, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Medical OncologyChengdu, China. [Zhang, Shuang] Capital Medical University, Beijing Shijitan Hospital, Department of PathologyBeijing, China. [Chen, Yue] Capital Medical University, Beijing Shijitan Hospital, Department of PathologyBeijing, China. RP Chen, Y (reprint author), Capital Medical University, Beijing Shijitan Hospital, Department of Pathology, Beijing, China. EM chenyue8249@163.com CR Nabors LB, Portnow J, Ahluwalia M, Baehring J, Brem H, Brem S, et al. Central nervous system cancers, version 3.2020, nccn clinical practice guidelines in oncology. J Natl Compr Canc Netw, 2020, 18(11):1537–70., DOI 10.6004/jnccn. 2020.0052 Molinaro AM, Taylor JW, Wiencke JK, Wrensch MR. Genetic and molecular epidemiology of adult diffuse glioma. Nat Rev Neurol, 2019, 15(7):405–17., DOI 10. 1038/s41582-019-0220-2 Whitfield BT, Huse JT. Classification of adult-type diffuse gliomas: impact of the world Health organization 2021 update. Brain Pathol, 2022, 32(4):e13062., DOI 10.1111/bpa.13062 Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 who classification of tumors of the central nervous system: a summary. Neuro Oncol, 2021, 23(8):1231–51., DOI 10.1093/neuonc/noab106 Stupp R, Mason WP, van den BentMJ, Weller M, Fisher B, TaphoornMJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med, 2005, 352(10):987–96., DOI 10.1056/NEJMoa043330 Burster T, Traut R, Yermekkyzy Z, Mayer K, Westhoff MA, Bischof J, et al. Critical view of novel treatment strategies for glioblastoma: failure and success of resistance mechanisms by glioblastoma cells. Front Cell Dev Biol, 2021, 9:695325., DOI 10.3389/fcell.2021.695325 Kopper TJ, Yu X, Graner MW. Immunopathology of extracellular vesicles in macrophage and glioma cross-talk. J Clin Med, 2023, 12(10):3430., DOI 10.3390/ jcm12103430 Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, et al. Single-cell rna-seq highlights intratumoral heterogeneity in primary glioblastoma. Science, 2014, 344(6190):1396–401., DOI 10.1126/science.1254257 Burko P, D’Amico G, Miltykh I, Scalia F, Conway de Macario E, Macario AJL, et al. Molecular pathways implicated in radioresistance of glioblastoma multiforme: what is the role of extracellular vesicles? Int J Mol Sci, 2023, 24(5):4883., DOI 10. 3390/ijms24054883 Perus LJM, Walsh LA. Microenvironmental heterogeneity in brain malignancies. Front Immunol, 2019, 10:2294., DOI 10.3389/fimmu.2019.02294 Broekman ML, Maas SLN, Abels ER, Mempel TR, Krichevsky AM, Breakefield XO. Multidimensional communication in the microenvirons of glioblastoma. Nat Rev Neurol, 2018, 14(8):482–95., DOI 10.1038/s41582-018- 0025-8 Matarredona ER, Pastor AM. Extracellular vesicle-mediated communication between the glioblastoma and its microenvironment. Cells, 2019, 9(1):96., DOI 10. 3390/cells9010096 van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol, 2018, 19(4):213–28., DOI 10.1038/nrm. 2017.125 Cano A, Ettcheto M, Bernuz M, Puerta R, Esteban de Antonio E, Sanchez- Lopez E, et al. Extracellular vesicles, the emerging mirrors of brain physiopathology. Int J Biol Sci, 2023, 19(3):721–43., DOI 10.7150/ijbs.79063 Colombo M, Raposo G, Thery C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol, 2014, 30:255–89., DOI 10.1146/annurev-cellbio-101512-122326 Spelat R, Jihua N, Sanchez Trivino CA, Pifferi S, Pozzi D, Manzati M, et al. The dual action of glioma-derived exosomes on neuronal activity: synchronization and disruption of synchrony. Cell Death Dis, 2022, 13(8):705., DOI 10.1038/s41419-022- 05144-6 Busatto S, Yang Y, Walker SA, Davidovich I, Lin WH, Lewis-Tuffin L, et al. Brain metastases-derived extracellular vesicles induce binding and aggregation of low-density lipoprotein. J Nanobiotechnology, 2020, 18(1):162., DOI 10.1186/ s12951-020-00722-2 Barile L, Vassalli G. Exosomes: therapy delivery tools and biomarkers of diseases. Pharmacol Ther, 2017, 174:63–78., DOI 10.1016/j.pharmthera.2017.02.020 Virgintino D, Rizzi M, Errede M, Strippoli M, Girolamo F, Bertossi M, et al. Plasma membrane-derived microvesicles released from tip endothelial cells during vascular sprouting. Angiogenesis, 2012, 15(4):761–9., DOI 10.1007/s10456-012- 9292-y Ban LA, Shackel NA, McLennan SV. Extracellular vesicles: a new frontier in biomarker discovery for non-alcoholic fatty liver disease. Int J Mol Sci, 2016, 17(3): 376., DOI 10.3390/ijms17030376 Vlassov AV, Magdaleno S, Setterquist R, Conrad R. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim Biophys Acta, 2012, 1820(7):940–8., DOI 10.1016/ j.bbagen.2012.03.017 Azam Z, Quillien V, Wang G, To ST. The potential diagnostic and prognostic role of extracellular vesicles in glioma: current status and future perspectives. Acta Oncol, 2019, 58(3):353–62., DOI 10.1080/0284186X.2018.1551621 Grant BD, Donaldson JG. Pathways and mechanisms of endocytic recycling. Nat Rev Mol Cell Biol, 2009, 10(9):597–608., DOI 10.1038/nrm2755 Xu M, Ji J, Jin D, Wu Y, Wu T, Lin R, et al. The biogenesis and secretion of exosomes and multivesicular bodies, mvbs): intercellular shuttles and implications in human diseases. Genes Dis, 2022, 10(5):1894–907., DOI 10.1016/j.gendis.2022. 03.021 Tschuschke M, Kocherova I, Bryja A, Mozdziak P, Angelova Volponi A, Janowicz K, et al. Inclusion biogenesis, methods of isolation and clinical application of human cellular exosomes. J Clin Med, 2020, 9(2):436., DOI 10.3390/jcm9020436 Juan T, FurthauerM. Biogenesis and function of escrt-dependent extracellular vesicles. Semin Cell Dev Biol, 2018, 74:66–77., DOI 10.1016/j.semcdb.2017.08.022 Babst M. Mvb vesicle formation: escrt-dependent, escrt-independent and everything in between. Curr Opin Cell Biol, 2011, 23(4):452–7., DOI 10.1016/j.ceb. 2011.04.008 Xie S, Zhang Q, Jiang L. Current knowledge on exosome biogenesis, cargosorting mechanism and therapeutic implications. Membranes, Basel,, 2022, 12(5): 498., DOI 10.3390/membranes12050498 Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci, 2018, 75(2):193–208., DOI 10.1007/s00018-017- 2595-9 Jin Y, Ma L, Zhang W, Yang W, Feng Q, Wang H. Extracellular signals regulate the biogenesis of extracellular vesicles. Biol Res, 2022, 55(1):35., DOI 10. 1186/s40659-022-00405-2 Pavlic A, Poelman H, Wasilewski G, Wichapong K, Lux P, Maassen C, et al. Inhibition of neutral sphingomyelinase 2 by novel small molecule inhibitors results in decreased release of extracellular vesicles by vascular smooth muscle cells and attenuated calcification. Int J Mol Sci, 2023, 24(3): 2027., DOI 10.3390/ijms24032027 Hurwitz SN, Cheerathodi MR, Nkosi D, York SB, Meckes DG. Tetraspanin Cd63 bridges autophagic and endosomal processes to regulate exosomal secretion and intracellular signaling of epstein-barr virus Lmp1. J Virol, 2018, 92(5):e01969., DOI 10.1128/JVI.01969-17 Al-Nedawi K, Meehan B, Rak J. Microvesicles: messengers and mediators of tumor progression. Cell Cycle, 2009, 8(13):2014–8., DOI 10.4161/cc.8.13.8988 Stahl PD, Raposo G. Extracellular vesicles: exosomes and microvesicles, integrators of homeostasis. Physiology, Bethesda,, 2019, 34(3):169–77., DOI 10. 1152/physiol.00045.2018 Nabhan JF, Hu R, Oh RS, Cohen SN, Lu Q. Formation and release of arrestin domain-containing protein 1-mediated microvesicles, armms, at plasma membrane by recruitment of Tsg101 protein. Proc Natl Acad Sci U S A, 2012, 109(11):4146–51., DOI 10.1073/pnas.1200448109 Russo MN, Whaley LA, Norton ES, Zarco N, Guerrero-Cazares H. Extracellular vesicles in the glioblastoma microenvironment: a diagnostic and therapeutic perspective. Mol Aspects Med, 2023, 91:101167., DOI 10.1016/j.mam. 2022.101167 Minciacchi VR, You S, Spinelli C, Morley S, Zandian M, Aspuria PJ, et al. Large oncosomes contain distinct protein cargo and represent a separate functional class of tumor-derived extracellular vesicles. Oncotarget, 2015, 6(13):11327–41., DOI 10.18632/oncotarget.3598 Chen J, Li P, Zhang T, Xu Z, Huang X,Wang R, et al. Review on strategies and technologies for exosome isolation and purification. Front Bioeng Biotechnol, 2021, 9:811971., DOI 10.3389/fbioe.2021.811971 Lin S, Yu Z, Chen D, Wang Z, Miao J, Li Q, et al. Progress in microfluidicsbased exosome separation and detection technologies for diagnostic applications. Small, 2020, 16(9):e1903916., DOI 10.1002/smll.201903916 Ding L, Yang X, Gao Z, Effah CY, Zhang X,Wu Y, et al. A holistic review of the state-of-the-art microfluidics for exosome separation: an overview of the current status, existing obstacles, and future outlook. Small, 2021, 17(29):e2007174., DOI 10. 1002/smll.202007174 Mohammadi M, Zargartalebi H, Salahandish R, Aburashed R, Wey Yong K, Sanati-Nezhad A. Emerging technologies and commercial products in exosomebased cancer diagnosis and prognosis. Biosens Bioelectron, 2021, 183:113176., DOI 10.1016/j.bios.2021.113176 Coumans FAW, Brisson AR, Buzas EI, Dignat-George F, Drees EEE, El- Andaloussi S, et al. Methodological guidelines to study extracellular vesicles. Circ Res, 2017, 120(10):1632–48., DOI 10.1161/CIRCRESAHA.117.309417 Jayachandran M, Miller VM, Heit JA, Owen WG. Methodology for isolation, identification and characterization of microvesicles in peripheral blood. J Immunol Methods, 2012, 375(1-2):207–14., DOI 10.1016/j.jim.2011.10.012 Thery C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018, Misev2018): a position statement of the international society for extracellular vesicles and update of the Misev2014 guidelines. J Extracell Vesicles, 2018, 7(1):1535750., DOI 10.1080/ 20013078.2018.1535750 Iraci N, Leonardi T, Gessler F, Vega B, Pluchino S. Focus on extracellular vesicles: physiological role and signalling properties of extracellular membrane vesicles. Int J Mol Sci, 2016, 17(2):171., DOI 10.3390/ijms17020171 Pancholi S, Tripathi A, Bhan A, AcharyaMM, Pillai P. Emerging concepts on the role of extracellular vesicles and its cargo contents in glioblastomamicroglial crosstalk. Mol Neurobiol, 2022, 59(5):2822–37., DOI 10.1007/s12035- 022-02752-3 Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport rna and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol, 2008, 10(12):1470–6., DOI 10.1038/ncb1800 Zottel A, Samec N, Kump A, Raspor Dall’Olio LR, Puzar Dominkus P, Romih R, et al. Analysis of mir-9-5p, mir-124-3p, mir-21-5p, mir-138-5p, and mir-1-3p in glioblastoma cell lines and extracellular vesicles. Int J Mol Sci, 2020, 21(22):8491., DOI 10.3390/ijms21228491 Wang J, Li T, Wang B. Exosomal transfer of mir-25-3p promotes the proliferation and temozolomide resistance of glioblastoma cells by targeting Fbxw7. Int J Oncol, 2021, 59(2):64., DOI 10.3892/ijo.2021.5244 Zhao G, Yu H, Ding L, Wang W, Wang H, Hu Y, et al. Microrna-27a-3p delivered by extracellular vesicles from glioblastoma cells induces M2 macrophage polarization via the ezh1/kdm3a/ctgf Axis. Cell Death Discov, 2022, 8(1):260., DOI 10.1038/s41420-022-01035-z Yin J, Ge X, Shi Z, Yu C, Lu C,Wei Y, et al. Extracellular vesicles derived from hypoxic glioma stem-like cells confer temozolomide resistance on glioblastoma by delivering mir-30b-3p. Theranostics, 2021, 11(4):1763–79., DOI 10.7150/thno.47057 Li J, Yuan H, Xu H, Zhao H, Xiong N. Hypoxic cancer-secreted exosomal mir- 182-5p promotes glioblastoma angiogenesis by targeting kruppel-like factor 2 and 4. Mol Cancer Res, 2020, 18(8):1218–31., DOI 10.1158/1541-7786.MCR-19-0725 Yin J, Zeng A, Zhang Z, Shi Z, Yan W, You Y. Exosomal transfer of mir-1238 contributes to temozolomide-resistance in glioblastoma. EBioMedicine, 2019, 42: 238–51., DOI 10.1016/j.ebiom.2019.03.016 Qian M, Wang S, Guo X, Wang J, Zhang Z, Qiu W, et al. Hypoxic gliomaderived exosomes deliver microRNA-1246 to induce M2 macrophage polarization by targeting TERF2IP via the STAT3 and NF-κB pathways. Oncogene, 2020, 39(2): 428–42., DOI 10.1038/s41388-019-0996-y Hong S, You JY, Paek K, Park J, Kang SJ, Han EH, et al. Inhibition of tumor progression and M2 microglial polarization by extracellular vesicle-mediated microrna-124 in a 3d microfluidic glioblastoma microenvironment. Theranostics, 2021, 11(19):9687–704., DOI 10.7150/thno.60851 Bier A, Hong X, Cazacu S, Goldstein H, Rand D, Xiang C, et al. Mir-504 modulates the stemness and mesenchymal transition of glioma stem cells and their interaction with microglia via delivery by extracellular vesicles. Cell Death Dis, 2020, 11(10):899., DOI 10.1038/s41419-020-03088-3 Yan T, Wu M, Lv S, Hu Q, Xu W, Zeng A, et al. Exosomes derived from microrna-512-5p-transfected bone mesenchymal stem cells inhibit glioblastoma progression by targeting Jag1. Aging, Albany NY,, 2021, 13(7):9911–26., DOI 10. 18632/aging.202747 Ma X, Li Z, Li T, Zhu L, Li Z, Tian N. Long non-coding rna hotair enhances angiogenesis by induction of vegfa expression in glioma cells and transmission to endothelial cells via glioma cell derived-extracellular vesicles. Am J Transl Res, 2017, 9(11):5012–21. Zhang Z, Yin J, Lu C, Wei Y, Zeng A, You Y. Exosomal transfer of long noncoding rna sbf2-as1 enhances chemoresistance to temozolomide in glioblastoma. J Exp Clin Cancer Res, 2019, 38(1):166., DOI 10.1186/s13046-019-1139-6 Chai Y,Wu HT, Liang CD, You CY, Xie MX, Xiao SW. Exosomal lncrna ror1- as1 derived from tumor cells promotes glioma progression via regulating mir-4686. Int J Nanomedicine, 2020, 15:8863–72., DOI 10.2147/IJN.S271795 Jiang Y, Zhao J, Xu J, Zhang H, Zhou J, Li H, et al. Glioblastoma-associated microglia-derived exosomal Circkif18a promotes angiogenesis by targeting Foxc2. Oncogene, 2022, 41(26):3461–73., DOI 10.1038/s41388-022-02360-4 Stella M, Falzone L, Caponnetto A, Gattuso G, Barbagallo C, Battaglia R, et al. Serum extracellular vesicle-derived Circhipk3 and Circsmarca5 are two novel diagnostic biomarkers for glioblastoma multiforme. Pharmaceuticals, Basel,, 2021, 14(7):618., DOI 10.3390/ph14070618 Scholl JN, de Fraga Dias A, Pizzato PR, Lopes DV, Moritz CEJ, Jandrey EHF, et al. Characterization and antiproliferative activity of glioma-derived extracellular vesicles. Nanomedicine, Lond,, 2020, 15(10):1001–18., DOI 10.2217/nnm-2019-0431 Ngo MT, Harley BAC. Perivascular signals alter global gene expression profile of glioblastoma and response to temozolomide in a gelatin hydrogel. Biomaterials, 2019, 198:122–34., DOI 10.1016/j.biomaterials.2018.06.013 Szatanek R, Baj-Krzyworzeka M. Cd44 and tumor-derived extracellular vesicles, tevs). Possible gateway to cancer metastasis. Int J Mol Sci, 2021, 22(3): 1463., DOI 10.3390/ijms22031463 Lucero R, Zappulli V, Sammarco A, Murillo OD, Cheah PS, Srinivasan S, et al. Glioma-derived mirna-containing extracellular vesicles induce angiogenesis by reprogramming brain endothelial cells. Cell Rep, 2020, 30(7):2065–74., DOI 10. 1016/j.celrep.2020.01.073 Musatova OE, Rubtsov YP. Effects of glioblastoma-derived extracellular vesicles on the functions of immune cells. Front Cell Dev Biol, 2023, 11: 1060000., DOI 10.3389/fcell.2023.1060000 de Mooij T, Peterson TE, Evans J, McCutcheon B, Parney IF. Short noncoding rna sequencing of glioblastoma extracellular vesicles. J Neurooncol, 2020, 146(2):253–63., DOI 10.1007/s11060-019-03384-9 Li Q, Barres BA. Microglia and macrophages in brain homeostasis and disease. Nat Rev Immunol, 2018, 18(4):225–42., DOI 10.1038/nri.2017.125 Hambardzumyan D, Gutmann DH, Kettenmann H. The role ofmicroglia and macrophages in glioma maintenance and progression. Nat Neurosci, 2016, 19(1): 20–7., DOI 10.1038/nn.4185 Hou L, Zhao Y, Song GQ, Ma YH, Jin XH, Jin SL, et al. Interfering cellular lactate homeostasis overcomes taxol resistance of breast cancer cells through the microrna-124-mediated lactate transporter, Mct1, inhibition. Cancer Cell Int, 2019, 19:193., DOI 10.1186/s12935-019-0904-0 Serpe C, Monaco L, Relucenti M, Iovino L, Familiari P, Scavizzi F, et al. Microglia-derived small extracellular vesicles reduce glioma growth by modifying tumor cell metabolism and enhancing glutamate clearance through mir-124. Cells, 2021, 10(8):2066., DOI 10.3390/cells10082066 Kiyokawa J, Kawamura Y, Ghouse SM, Acar S, Barcin E, Martinez-Quintanilla J, et al. Modification of extracellular matrix enhances oncolytic adenovirus immunotherapy in glioblastoma. Clin Cancer Res, 2021, 27(3):889–902., DOI 10. 1158/1078-0432.CCR-20-2400 Kim K, Kim IK, Yang JM, Lee E, Koh BI, Song S, et al. Soxf transcription factors are positive feedback regulators of vegf signaling. Circ Res, 2016, 119(7): 839–52., DOI 10.1161/CIRCRESAHA.116.308483 Svensson A, Ozen I, Genove G, Paul G, Bengzon J. Endogenous brain pericytes are widely activated and contribute to mouse glioma microvasculature. PLoS One, 2015, 10(4):e0123553., DOI 10.1371/journal.pone.0123553 de Gooijer MC, de Vries NA, Buckle T, Buil LCM, Beijnen JH, Boogerd W, et al. Improved brain penetration and antitumor efficacy of temozolomide by inhibition of Abcb1 and Abcg2. Neoplasia, 2018, 20(7):710–20., DOI 10.1016/j.neo. 2018.05.001 Wang Z, Yuan Y, Ji X, Xiao X, Li Z, Yi X, et al. The hippo-taz Axis mediates vascular endothelial growth factor C in glioblastoma-derived exosomes to promote angiogenesis. Cancer Lett, 2021, 513:1–13., DOI 10.1016/j.canlet.2021.05.002 Choi D, Montermini L, Kim DK, Meehan B, Roth FP, Rak J. The impact of oncogenic egfrviii on the proteome of extracellular vesicles released from glioblastoma cells. Mol Cell Proteomics, 2018, 17(10):1948–64., DOI 10.1074/mcp. RA118.000644 Yang ZJ, Bi QC, Gan LJ, Zhang LL,Wei MJ, Hong T, et al. Exosomes derived from glioma cells under hypoxia promote angiogenesis through up-regulated exosomal connexin 43. Int J Med Sci, 2022, 19(7):1205–15., DOI 10.7150/ijms.71912 Wang M, Pan W, Xu Y, Zhang J, Wan J, Jiang H. Microglia-mediated neuroinflammation: a potential target for the treatment of cardiovascular diseases. J Inflamm Res, 2022, 15:3083–94., DOI 10.2147/JIR.S350109 Himes BT, Peterson TE, de Mooij T, Garcia LMC, Jung MY, Uhm S, et al. The role of extracellular vesicles and Pd-L1 in glioblastoma-mediated immunosuppressive monocyte induction. Neuro Oncol, 2020, 22(7):967–78., DOI 10.1093/neuonc/noaa029 Thompson LF, Eltzschig HK, Ibla JC, Van De Wiele CJ, Resta R, Morote- Garcia JC, et al. Crucial role for ecto-5’-nucleotidase, Cd73, in vascular leakage during hypoxia. J Exp Med, 2004, 200(11):1395–405., DOI 10.1084/jem.20040915 Wang M, Jia J, Cui Y, Peng Y, Jiang Y. Cd73-Positive extracellular vesicles promote glioblastoma immunosuppression by inhibiting T-cell clonal expansion. Cell Death Dis, 2021, 12(11):1065., DOI 10.1038/s41419-021-04359-3 Liang T,Wang X,Wang F, Feng E, You G. Galectin-9: a predictive biomarker negatively regulating immune response in glioma patients. World Neurosurg, 2019, 132:e455–e62., DOI 10.1016/j.wneu.2019.08.117 Wang M, Cai Y, Peng Y, Xu B, Hui W, Jiang Y. Exosomal Lgals9 in the cerebrospinal fluid of glioblastoma patients suppressed dendritic cell antigen presentation and cytotoxic T-cell immunity. Cell Death Dis, 2020, 11(10):896., DOI 10.1038/s41419-020-03042-3 Vargova L, Sykova E. Astrocytes and extracellular matrix in extrasynaptic volume transmission. Philos Trans R Soc Lond B Biol Sci, 2014, 369(1654): 20130608., DOI 10.1098/rstb.2013.0608 BrandaoM, Simon T, Critchley G, Giamas G. Astrocytes, the rising stars of the glioblastoma microenvironment. Glia, 2019, 67(5):779–90., DOI 10.1002/glia.23520 Oushy S, Hellwinkel JE, Wang M, Nguyen GJ, Gunaydin D, Harland TA, et al. Glioblastoma multiforme-derived extracellular vesicles drive normal astrocytes towards a tumour-enhancing phenotype. Philos Trans R Soc Lond B Biol Sci, 2018, 373(1737):20160477., DOI 10.1098/rstb.2016.0477 Zeng A, Wei Z, Rabinovsky R, Jun HJ, El Fatimy R, Deforzh E, et al. Glioblastoma-derived extracellular vesicles facilitate transformation of astrocytes via reprogramming oncogenic metabolism. iScience, 2020, 23(8):101420., DOI 10. 1016/j.isci.2020.101420 Biasoli D, Sobrinho MF, da Fonseca AC, de Matos DG, Romao L, de Moraes Maciel R, et al. Glioblastoma cells inhibit astrocytic P53-expression favoring cancer malignancy. Oncogenesis, 2014, 3(10):e123., DOI 10.1038/oncsis.2014.36 Laurenti E, Wilson A, Trumpp A.Myc’s other life: stem cells and beyond. Curr Opin Cell Biol, 2009, 21(6):844–54., DOI 10.1016/j.ceb.2009.09.006 Hallal S, Mallawaaratchy DM, Wei H, Ebrahimkhani S, Stringer BW, Day BW, et al. Extracellular vesicles released by glioblastoma cells stimulate normal astrocytes to acquire a tumor-supportive phenotype via P53 and myc signaling pathways. Mol Neurobiol, 2019, 56(6):4566–81., DOI 10.1007/s12035-018-1385-1 Neagu MR, Huang RY, Reardon DA, Wen PY. How treatment monitoring is influencing treatment decisions in glioblastomas. Curr Treat Options Neurol, 2015, 17(4):343., DOI 10.1007/s11940-015-0343-8 Marei HE, Althani A, Afifi N, Hasan A, Caceci T, Cifola I, et al. Glioma extracellular vesicles for precision medicine: prognostic and theragnostic application. Discov Oncol, 2022, 13(1):49., DOI 10.1007/s12672-022-00514-0 Zhou B, Xu K, Zheng X, Chen T, Wang J, Song Y, et al. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct Target Ther, 2020, 5(1):144., DOI 10.1038/s41392-020-00258-9 Macedo-Pereira A, Martins C, Lima J, Sarmento B. Digging the intercellular crosstalk via extracellular vesicles: may exosomes Be the drug delivery solution for target glioblastoma? J Control Release, 2023, 358:98–115., DOI 10.1016/j.jconrel. 2023.04.038 Osti D, Del Bene M, Rappa G, Santos M, Matafora V, Richichi C, et al. Clinical significance of extracellular vesicles in plasma from glioblastoma patients. Clin Cancer Res, 2019, 25(1):266–76., DOI 10.1158/1078-0432.CCR- 18-1941 Akers JC, Ramakrishnan V, Kim R, Skog J, Nakano I, Pingle S, et al. Mir-21 in the extracellular vesicles, evs, of cerebrospinal fluid, csf): a platform for glioblastoma biomarker development. PLoS One, 2013, 8(10):e78115., DOI 10. 1371/journal.pone.0078115 Sabbagh Q, Andre-Gregoire G, Guevel L, Vesiclemia GJ. Vesiclemia: counting on extracellular vesicles for glioblastoma patients. Oncogene, 2020, 39(38):6043–52., DOI 10.1038/s41388-020-01420-x Catalano M, O’Driscoll L. Inhibiting extracellular vesicles formation and release: a review of ev inhibitors. J Extracell Vesicles, 2020, 9(1):1703244., DOI 10. 1080/20013078.2019.1703244 Fox JE, Austin CD, Reynolds CC, Steffen PK. Evidence that agonist-induced activation of calpain causes the shedding of procoagulant-containing microvesicles from the membrane of aggregating platelets. J Biol Chem, 1991, 266(20):13289–95., DOI 10.1016/s0021-9258(18)98837-x Atanassoff AP, Wolfmeier H, Schoenauer R, Hostettler A, Ring A, Draeger A, et al. Microvesicle shedding and lysosomal repair fulfill divergent cellular needs during the repair of streptolysin O-induced plasmalemmal damage. PLoS One, 2014, 9(2):e89743., DOI 10.1371/journal.pone.0089743 Jorfi S, Ansa-Addo EA, Kholia S, Stratton D, Valley S, Lange S, et al. Inhibition of microvesiculation sensitizes prostate cancer cells to chemotherapy and reduces docetaxel dose required to limit tumor growth in vivo. Sci Rep, 2015, 5: 13006., DOI 10.1038/srep13006 Cai L, Li Q, LiW,Wang C, TuM, Zhu Z, et al. Calpain suppresses cell growth and invasion of glioblastomamultiforme by producing the cleavage of filamin A. Int J Clin Oncol, 2020, 25(6):1055–66., DOI 10.1007/s10147-020-01636-7 Stillger MN, Chen CY, Lai ZW, Li M, Schafer A, Pagenstecher A, et al. Changes in calpain-2 expression during glioblastoma progression predisposes tumor cells to temozolomide resistance by minimizing DNA damage and P53- dependent apoptosis. Cancer Cell Int, 2023, 23(1):49., DOI 10.1186/s12935-023- 02889-8 Datta A, Kim H, LalM,McGee L, Johnson A,Moustafa AA, et al.Manumycin a suppresses exosome biogenesis and secretion via targeted inhibition of ras/raf/erk1/ 2 signaling and hnrnp H1 in castration-resistant prostate cancer cells. Cancer Lett, 2017, 408:73–81., DOI 10.1016/j.canlet.2017.08.020 Munson JM, Fried L, Rowson SA, Bonner MY, Karumbaiah L, Diaz B, et al. Anti-invasive adjuvant therapy with imipramine blue enhances chemotherapeutic efficacy against glioma. Sci Transl Med, 2012, 4(127):127ra36., DOI 10.1126/ scitranslmed.3003016 Xu X, Liu Y, Li Y, Chen H, Zhang Y, Liu J, et al. Selective exosome exclusion of mir-375 by glioma cells promotes glioma progression by activating the ctgf-egfr pathway. J Exp Clin Cancer Res, 2021, 40(1):16., DOI 10.1186/s13046-020-01810-9 Munoz JL, Bliss SA, Greco SJ, Ramkissoon SH, Ligon KL, Rameshwar P. Delivery of functional anti-mir-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol Ther Nucleic Acids, 2013, 2(10):e126., DOI 10.1038/mtna.2013.60 Geng L, Xu J, Zhu Y, Hu X, Liu Y, Yang K, et al. Targeting mir-9 in glioma stem cell-derived extracellular vesicles: a novel diagnostic and therapeutic biomarker. Transl Oncol, 2022, 22:101451., DOI 10.1016/j.tranon.2022.101451 Gabizon AA, de Rosales RTM, La-Beck NM. Translational considerations in nanomedicine: the oncology perspective. Adv Drug Deliv Rev, 2020, 158:140–57., DOI 10.1016/j.addr.2020.05.012 Benecke L, Coray M, Umbricht S, Chiang D, Figueiro F, Muller L. Exosomes: small evs with large immunomodulatory effect in glioblastoma. Int J Mol Sci, 2021, 22(7):3600., DOI 10.3390/ijms22073600 Simon T, Kumaran A, Veselu DF, Giamas G. Three method-combination protocol for improving purity of extracellular vesicles. Int J Mol Sci, 2020, 21(9): 3071., DOI 10.3390/ijms21093071 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611549 EP 1611562 DI 10.3389/pore.2024.1611549 PG 14 ER PT J AU Hatchett, JW Brunetti, M Andersen, K Tandsaether, RM Lobmaier, I Lund-Iversen, M Lien-Dahl, Th Micci, F Panagopoulos, I AF Hatchett, John William Brunetti, Marta Andersen, Kristin Tandsaether, Randi Maren Lobmaier, Ingvild Lund-Iversen, Marius Lien-Dahl, Thomas Micci, Francesca Panagopoulos, Ioannis TI Genetic characterization of intramuscular myxomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE intramuscular myxoma; chromosomal aberrations; GNAS; codon 201; codon 227; Sanger sequencing; Ion torrent ID intramuscular myxoma; chromosomal aberrations; GNAS; codon 201; codon 227; Sanger sequencing; Ion torrent AB Introduction: Intramuscular myxomas are benign tumors that are challenging to diagnose, especially on core needle biopsies. Acquired chromosomal aberrations and pathogenic variants in codon 201 or codon 227 in GNAS complex locus gene (GNAS) have been reported in these tumors. Here we present our genetic findings in a series of 22 intramuscular myxomas. Materials and methods: The tumors were investigated for the presence of acquired chromosomal aberrations using G-banding and karyotyping. Pathogenic variants in codon 201 or codon 227 of GNAS were assessed using direct cycle Sanger sequencing and Ion AmpliSeq Cancer Hotspot Panel v2 methodologies. Results: Eleven tumors carried chromosomal abnormalities. Six tumors had numerical, four had structural, and one had both numerical and structural chromosomal aberrations. Gains of chromosomes 7 and 8 were the most common abnormalities being found in five and four tumors respectively. Pathogenic variants in GNAS were detected in 19 myxomas (86%) with both methodologies. The detected pathogenic variants were p.R201H in nine cases (seven with abnormal and two with normal karyotypes), p.R201C in five cases, all with normal karyotypes, p.R201S in three cases (two with abnormal and one with normal karyotype), p.R201G in one case with a normal karyotype, and p.Q227E in one case with a normal karyotype. Conclusion: Firstly, our data indicate a possible association between chromosomal abnormalities and GNAS pathogenic variants in intramuscular myxomas. Secondly, the presence of the rare pathogenic variants R201S, p.R201G and p.Q227E in 26% (5 out of 19) of myxomas with GNAS pathogenic variants shows that methodologies designed to detect only the common “hotspot” of p.R201C and p.R201H will give false negative results. Finally, a comparison between Ion AmpliSeq Cancer Hotspot Panel v2 and direct cycle Sanger sequencing showed that direct cycle Sanger sequencing provides a quick, reliable, and relatively cheap method to detect GNAS pathogenic variants, matching even the most cutting-edge sequencing methods. C1 [Hatchett, John William] The Norwegian Radium Hospital, Oslo University Hospital, Institute for Cancer Genetics and Informatics, Section for Cancer CytogeneticsOslo, Norway. [Brunetti, Marta] The Norwegian Radium Hospital, Oslo University Hospital, Institute for Cancer Genetics and Informatics, Section for Cancer CytogeneticsOslo, Norway. [Andersen, Kristin] The Norwegian Radium Hospital, Oslo University Hospital, Institute for Cancer Genetics and Informatics, Section for Cancer CytogeneticsOslo, Norway. [Tandsaether, Randi Maren] The Norwegian Radium Hospital, Oslo University Hospital, Institute for Cancer Genetics and Informatics, Section for Cancer CytogeneticsOslo, Norway. [Lobmaier, Ingvild] The Norwegian Radium Hospital, University of Oslo, Department of PathologyOslo, Norway. [Lund-Iversen, Marius] The Norwegian Radium Hospital, University of Oslo, Department of PathologyOslo, Norway. [Lien-Dahl, Thomas] The Norwegian Radium Hospital, University of Oslo, Department of PathologyOslo, Norway. [Micci, Francesca] The Norwegian Radium Hospital, Oslo University Hospital, Institute for Cancer Genetics and Informatics, Section for Cancer CytogeneticsOslo, Norway. [Panagopoulos, Ioannis] The Norwegian Radium Hospital, Oslo University Hospital, Institute for Cancer Genetics and Informatics, Section for Cancer CytogeneticsOslo, Norway. RP Panagopoulos, I (reprint author), The Norwegian Radium Hospital, Oslo University Hospital, Institute for Cancer Genetics and Informatics, Section for Cancer Cytogenetics, Oslo, Norway. EM ioannis.panagopoulos@rrresearch.no CR Enzinger FM. Intramuscular myxoma: a review and follow-up study of 34 cases. Am J Clin Pathol, 1965, 43:104–13., DOI 10.1093/ajcp/43.2.104 Miettinen M, Markku M. Modern soft tissue pathology: tumors and non-neoplastic conditions. Cambridge University Press, 2016). p. 1062. Available from: https://www. cambridge.org/core/product/716C3345EDB53691CEE30DB640C84E7E, Accessed June 13, 2023). Nielsen GP, O’Connell JX, Rosenberg AE. Intramuscular myxoma: a clinicopathologic study of 51 cases with emphasis on hypercellular and hypervascular variants. Am J Surg Pathol, 1998, 22:1222–7., DOI 10.1097/ 00000478-199810000-00007 Kindblom L-G, Stener B, Angervall L. Intramuscular myxoma. Cancer, 1974, 34:1737–44., DOI 10.1002/1097-0142(197411)34:5<1737::AIDCNCR2820340523> 3.0.CO;2-U Miettinen M, Hockerstedt K, Reitamo J, Totterman S. Intramuscular myxoma—a clinicopathological study of twenty-three cases. Am J Clin Pathol, 1985, 84:265–72., DOI 10.1093/ajcp/84.3.265 Hashimoto H, Tsuneyoshi M, Daimaru Y, Enjoji M, Shinohara N. Intramuscular myxoma: a clinicopathologic, immunohistochemical, and electron microscopic study. Cancer, 1986, 58:740–7., DOI 10.1002/1097-0142(19860801)58: 3<740::AID-CNCR2820580322>3.0.CO;2-K Heymans O, Gebhart M, Alexiou J, de Saint Aubain N, Larsimont D. Intramuscular myxoma. Acta Chir Belg, 1998, 98:120–2., DOI 10.1080/00015458. 1998.12098394 Allen PW. Myxoma is not a single entity: a review of the concept of myxoma. Ann Diagn Pathol, 2000, 4:99–123., DOI 10.1016/S1092-9134(00)90019-4 Murphey MD, McRae GA, Fanburg-Smith JC, Temple HT, Levine AM, Aboulafia AJ. Imaging of soft-tissue myxoma with emphasis on CT and MR and comparison of radiologic and pathologic findings. Radiology, 2002, 225: 215–24., DOI 10.1148/radiol.2251011627 Okamoto S, Hisaoka M, Ushijima M, Nakahara S, Toyoshima S, Hashimoto H. Activating Gs(alpha, mutation in intramuscular myxomas with and without fibrous dysplasia of bone. Virchows Archiv, 2000, 437:133–7., DOI 10.1007/ s004280000217 Schwindinger WF, Francomano CA, Levine MA. Identification of a mutation in the gene encoding the alpha subunit of the stimulatory G protein of adenylyl cyclase in McCune-Albright syndrome. Proc Natl Acad Sci, 1992, 89:5152–6., DOI 10.1073/pnas.89.11.5152 Shenker A, Weinstein LS, Moran A, Pescovitz OH, Charest NJ, Boney CM, et al. Severe endocrine and nonendocrine manifestations of the McCune-Albright syndrome associated with activating mutations of stimulatory G protein Gs. J Pediatr, 1993, 123:509–18., DOI 10.1016/S0022-3476(05)80943-6 Shenker A, Weinstein LS, Sweet DE, Spiegel AM. An activating Gs alpha mutation is present in fibrous dysplasia of bone in the McCune-Albright syndrome. J Clin Endocrinol Metab, 1994, 79:750–5., DOI 10.1210/jcem.79.3.8077356 Weinstein LS, Shenker A, Gejman PV, Merino MJ, Friedman E, Spiegel AM. Activating mutations of the stimulatory G protein in the McCune–albright syndrome. New Engl J Med, 1991, 325:1688–95., DOI 10.1056/ NEJM199112123252403 More A, Ito I, Haridas V, Chowdhury S, Gu Y, Dickson P, et al. Oncogene addiction to GNAS in GNASR201 mutant tumors. Oncogene, 2022, 41:4159–68., DOI 10.1038/s41388-022-02388-6 O’Hayre M, Vazquez-Prado J, Kufareva I, Stawiski EW, Handel TM, Seshagiri S, et al. The emerging mutational landscape of G proteins and G-protein-coupled receptors in cancer. Nat Rev Cancer, 2013, 13:412–24., DOI 10.1038/nrc3521 Weinstein LS, Chen M, Xie T, Liu J. Genetic diseases associated with heterotrimeric G proteins. Trends Pharmacol Sci, 2006, 27:260–6., DOI 10.1016/j. tips.2006.03.005 Delaney D, Diss TC, Presneau N, Hing S, Berisha F, Idowu BD, et al. GNAS1mutations occurmore commonly than previously thought in intramuscular myxoma. Mod Pathol, 2009, 22:718–24., DOI 10.1038/modpathol.2009.32 Walther I, Walther BM, Chen Y, Petersen I. Analysis of GNAS1 mutations in myxoid soft tissue and bone tumors. Pathol Res Pract, 2014, 210:1–4., DOI 10.1016/j. prp.2013.09.003 Bekers EM, Eijkelenboom A, Rombout P, van Zwam P, Mol S, Ruijter E, et al. Identification of novel GNAS mutations in intramuscular myxoma using nextgeneration sequencing with single-molecule tagged molecular inversion probes. Diagn Pathol, 2019, 14:15., DOI 10.1186/s13000-019-0787-3 Sunitsch S, Gilg MM, Kashofer K, Gollowitsch F, Leithner A, Liegl-Atzwanger B. Detection of GNAS mutations in intramuscular/cellular myxomas as diagnostic tool in the classification of myxoid soft tissue tumors. Diagn Pathol, 2018, 13:52., DOI 10.1186/s13000-018-0734-8 Libbrecht L, Bempt IV, Schubert T, Sciot R, Galant C. Next generation sequencing for GNAS uncovers CD34 as a sensitive marker for intramuscular myxoma. Ann Diagn Pathol, 2019, 43:151409., DOI 10.1016/j.anndiagpath.2019. 151409 Bartuma H, Moller E, Collin A, Domanski HA, Von Steyern FV, Mandahl N, et al. Fusion of the FUS and CREB3L2 genes in a supernumerary ring chromosome in low-grade fibromyxoid sarcoma. Cancer Genet Cytogenet, 2010, 199:143–6., DOI 10.1016/j.cancergencyto.2010.02.011 Mertens F, Fletcher CDM, Antonescu CR, Coindre J-M, Colecchia M, Domanski HA, et al. Clinicopathologic and molecular genetic characterization of low-grade fibromyxoid sarcoma, and cloning of a novel FUS/CREB3L1 fusion gene. Lab Invest, 2005, 85:408–15., DOI 10.1038/labinvest.3700230 Mentzel T, Calonje E, Wadden C, Camplejohn RS, Beham A, Smith MA, et al. Myxofibrosarcoma. Clinicopathologic analysis of 75 cases with emphasis on the low-grade variant. Am J Surg Pathol, 1996, 20:391–405., DOI 10.1097/00000478- 199604000-00001 Ogilvie CM, Torbert JT, Finstein JL, Fox EJ, Lackman RD. Clinical utility of percutaneous biopsies of musculoskeletal tumors. Clin Orthop Relat Res, 2006, 450: 95–100., DOI 10.1097/01.blo.0000229302.52147.c7 Panagopoulos I, Gorunova L, Lobmaier I, Bjerkehagen B, Heim S. Karyotyping and analysis of GNAS locus in intramuscular myxomas. Oncotarget, 2017, 8:22086–94., DOI 10.18632/oncotarget.14986 Fecteau RE, Lutterbaugh J, Markowitz SD,Willis J, Guda K. GNAS mutations identify a set of right-sided, RAS mutant, villous colon cancers. PLoS One, 2014, 9: e87966., DOI 10.1371/journal.pone.0087966 Dal Molin M, Matthaei H, Wu J, Blackford A, Debeljak M, Rezaee N, et al. Clinicopathological correlates of activating GNAS mutations in intraductal papillary mucinous neoplasm, IPMN, of the pancreas. Ann Surg Oncol, 2013, 20:3802–8., DOI 10.1245/s10434-013-3096-1 Gal S. Sequencing of double-stranded PCR products. In: Graham CA, Hill AJM, editors. DNA sequencing protocols. New Jersey: Humana Press, 1993). p. 183–90., DOI 10.1385/0-89603-248-5:183 Dorit RL, Ohara O, Hwang CB-C, Kim JB, Blackshaw S. Direct DNA sequencing of PCR products. In: Ausubel FM, Brent R, Kingston RE, Moore DD, Seidman JG, Struhl K, editors. Current protocols in molecular biology. Hoboken, NJ, USA: John Wiley and Sons, Inc., 2001)., DOI 10.1002/ 0471142727.mb1502s56 Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci U S A, 1977, 74:5463–7., DOI 10.1073/pnas.74.12.5463 Bevan IS, Rapley R, Walker MR. Sequencing of PCR-amplified DNA. PCR Methods Appl, 1992, 1:222–8., DOI 10.1101/gr.1.4.222 Kretz K, Callen W, Hedden V. Cycle sequencing. PCR Methods Appl, 1994, 3: S107–12., DOI 10.1101/gr.3.5.s107 Mardis ER. The impact of next-generation sequencing on cancer genomics: from discovery to clinic. Cold Spring Harb Perspect Med, 2019, 9:a036269., DOI 10. 1101/cshperspect.a036269 Baudhuin LM, Lagerstedt SA, Klee EW, Fadra N, Oglesbee D, Ferber MJ. Confirming variants in next-generation sequencing panel testing by sanger sequencing. J Mol Diagn, 2015, 17:456–61., DOI 10.1016/j.jmoldx.2015.03.004 Mu W, Lu H-M, Chen J, Li S, Elliott AM. Sanger confirmation is required to achieve optimal sensitivity and specificity in next-generation sequencing panel testing. J Mol Diagn, 2016, 18:923–32., DOI 10.1016/j.jmoldx.2016.07.006 De Cario R, Kura A, Suraci S, Magi A, Volta A, Marcucci R, et al. Sanger validation of high-throughput sequencing in genetic diagnosis: still the best practice? Front Genet, 2020, 11:592588., DOI 10.3389/fgene.2020.592588 Nagahashi M, Shimada Y, Ichikawa H, Kameyama H, Takabe K, Okuda S, et al. Next generation sequencing-based gene panel tests for the management of solid tumors. Cancer Sci, 2019, 110:6–15., DOI 10.1111/cas.13837 Colomer R, Miranda J, Romero-Laorden N, Hornedo J, Gonzalez-Cortijo L, Mouron S, et al. Usefulness and real-world outcomes of next generation sequencing testing in patients with cancer: an observational study on the impact of selection based on clinical judgement. EClinicalMedicine, 2023, 60:102029., DOI 10.1016/j. eclinm.2023.102029 Mauer CB, Pirzadeh-Miller SM, Robinson LD, Euhus DM. The integration of next-generation sequencing panels in the clinical cancer genetics practice: an institutional experience. Genet Med, 2014, 16:407–12., DOI 10.1038/gim.2013.160 Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, et al. COSMIC: the Catalogue of somatic mutations in cancer. Nucleic Acids Res, 2019, 47: D941–D947., DOI 10.1093/nar/gky1015 Panagopoulos I, Gorunova L, Lund-Iversen M, Andersen K, Andersen HK, Lobmaier I, et al. Cytogenetics of spindle cell/pleomorphic lipomas: karyotyping and FISH analysis of 31 tumors. Cancer Genomics Proteomics, 2018, 15:193–200., DOI 10.21873/cgp.20077 McGowan-Jordan J, Hastings R, Moore S, editors. ISCN 2020: an international System for human cytogenomic nomenclature. Karger, 2020). p. 164., DOI 10.1159/ isbn.978-3-318-06867-2 Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Basic local alignment search tool. J Mol Biol, 1990, 215:403–10., DOI 10.1016/S0022-2836(05)80360-2 Smith SC, Poznanski AA, Fullen DR, Ma L, McHugh JB, Lucas DR, et al. CD34-positive superficial myxofibrosarcoma: a potential diagnostic pitfall. J Cutan Pathol, 2013, 40:639–45., DOI 10.1111/cup.12158 Sugiura Y, Machinami R, Matsumoto S, Kanda H, Ae K, Takazawa Y, et al. Prognostic value of CD34 expression status in patients with myxofibrosarcomas and undifferentiated pleomorphic sarcomas. Sci Rep, 2021, 11:15494., DOI 10.1038/ s41598-021-94834-w Smits AJJ, Kummer JA, de Bruin PC, Bol M, van den Tweel JG, Seldenrijk KA, et al. The estimation of tumor cell percentage for molecular testing by pathologists is not accurate. Mod Pathol, 2014, 27:168–74., DOI 10.1038/ modpathol.2013.134 Lhermitte B, Egele C, Weingertner N, Ambrosetti D, Dadone B, Kubiniek V, et al. Adequately defining tumor cell proportion in tissue samples for molecular testing improves interobserver reproducibility of its assessment. Virchows Archiv, 2017, 470:21–7., DOI 10.1007/s00428-016-2042-6 Sreekantaiah C, Karakousis CP, Leong SP, Sandberg AA. Trisomy 8 as a nonrandom secondary change in myxoid liposarcoma. Cancer Genet Cytogenet, 1991, 51:195–205., DOI 10.1016/0165-4608(91)90132-e Bridge JA, Swarts SJ, Buresh C, Nelson M, Degenhardt JM, Spanier S, et al. Trisomies 8 and 20 characterize a subgroup of benign fibrous lesions arising in both soft tissue and bone. Am J Pathol, 1999, 154:729–33., DOI 10.1016/S0002-9440(10, 65319-9 Paulsson K, Johansson B. Trisomy 8 as the sole chromosomal aberration in acute myeloid leukemia and myelodysplastic syndromes. Pathol Biol, Paris,, 2007, 55:37–48., DOI 10.1016/j.patbio.2006.04.007 Hemsing AL, Hovland R, Tsykunova G, Reikvam H. Trisomy 8 in acute myeloid leukemia. Expert Rev Hematol, 2019, 12:947–58., DOI 10.1080/17474086. 2019.1657400 Broberg K, Toksvig-Larsen S, Lindstrand A, Mertens F. Trisomy 7 accumulates with age in solid tumors and non-neoplastic synovia. Genes Chromosomes Cancer, 2001, 30:310–5., DOI 10.1002/1098-2264(2000)9999:9999<:: aid-gcc1096>3.0.co;2-c Swansbury J. Cytogenetic and genetic studies in solid tumors: background. Methods Mol Biol, 2003, 220:125–33., DOI 10.1385/1-59259-363-1:125 Johansson B, Heim S, Mandahl N, Mertens F, Mitelman F. Trisomy 7 in nonneoplastic cells. Genes Chromosomes Cancer, 1993, 6:199–205., DOI 10.1002/gcc. 2870060402 Hayashi K. PCR-SSCP: a simple and sensitive method for detection of mutations in the genomic DNA. PCR Methods Appl, 1991, 1:34–8., DOI 10.1101/ gr.1.1.34 Riminucci M, Fisher LW, Majolagbe A, Corsi A, Lala R, De Sanctis C, et al. A novel GNAS1 mutation, R201G, inMcCune-albright syndrome. J BoneMineral Res, 1999, 14:1987–9., DOI 10.1359/jbmr.1999.14.11.1987 Liang Q, Wei M, Hodge L, Fanburg-Smith JC, Nelson A, Miettinen M, et al. Quantitative analysis of activating alpha subunit of the G protein, Gsα, mutation by pyrosequencing in fibrous dysplasia and other bone lesions. J Mol Diagn, 2011, 13: 137–42., DOI 10.1016/j.jmoldx.2010.10.003 Romanet P, Philibert P, Fina F, Cuny T, Roche C, Ouafik L, et al. Using digital droplet polymerase chain reaction to detect the mosaic GNAS mutations in whole blood DNA or circulating cell-free DNA in fibrous dysplasia and McCune-albright syndrome. J Pediatr, 2019, 205:281–5., DOI 10.1016/j.jpeds.2018.09.070 Lin Y-L, Ma R, Li Y. The biological basis and function of GNAS mutation in pseudomyxoma peritonei: a review. J Cancer Res Clin Oncol, 2020, 146:2179–88., DOI 10.1007/s00432-020-03321-8 Weinstein LS, Liu J, Sakamoto A, Xie T, Chen M. Minireview: GNAS: normal and abnormal functions. Endocrinology, 2004, 145:5459–64., DOI 10.1210/en.2004- 0865 Weinstein LS, Yu S, Warner DR, Liu J. Endocrine manifestations of stimulatory G protein alpha-subunit mutations and the role of genomic imprinting. Endocr Rev, 2001, 22:675–705., DOI 10.1210/edrv.22.5.0439 Khan SK, Yadav PS, Elliott G, Hu DZ, Xu R, Yang Y. Induced GnasR201H expression from the endogenous Gnas locus causes fibrous dysplasia by upregulating Wnt/β-catenin signaling. Proc Natl Acad Sci U S A, 2018, 115: E418–E427., DOI 10.1073/pnas.1714313114 Nomura R, Saito T, Mitomi H, Hidaka Y, Lee S, Watanabe S, et al. GNAS mutation as an alternative mechanism of activation of the Wnt/β-catenin signaling pathway in gastric adenocarcinoma of the fundic gland type. Hum Pathol, 2014, 45: 2488–96., DOI 10.1016/j.humpath.2014.08.016 Wilson CH, McIntyre RE, Arends MJ, Adams DJ. The activating mutation R201C in GNAS promotes intestinal tumourigenesis in Apc(Min/+, mice through activation of Wnt and ERK1/2 MAPK pathways. Oncogene, 2010, 29:4567–75., DOI 10.1038/onc.2010.202 Tirosh A, Jin DX, De Marco L, Laitman Y, Friedman E. Activating genomic alterations in the Gs alpha gene, GNAS, in 274 694 tumors. Genes Chromosomes Cancer, 2020, 59:503–16., DOI 10.1002/gcc.22854 Zhao X, Deng P, Iglesias-Bartolome R, Amornphimoltham P, Steffen DJ, Jin Y, et al. Expression of an active Gαs mutant in skeletal stem cells is sufficient and necessary for fibrous dysplasia initiation and maintenance. Proc Natl Acad Sci U S A, 2018, 115:E428–E437., DOI 10.1073/pnas.1713710115 Zhadina M, Roszko KL, Geels RES, de Castro LF, Collins MT, Boyce AM. Genotype-phenotype correlation in fibrous dysplasia/McCune-albright syndrome. J Clin Endocrinol Metab, 2021, 106:1482–90., DOI 10.1210/clinem/dgab053 Lumbroso S, Paris F, Sultan CEuropean Collaborative Study. Activating Gsalpha mutations: analysis of 113 patients with signs of McCune-Albright syndrome--a European Collaborative Study. J Clin Endocrinol Metab, 2004, 89: 2107–13., DOI 10.1210/jc.2003-031225 Jour G, Oultache A, Sadowska J, Mitchell T, Healey J, Nafa K, et al. GNAS mutations in fibrous dysplasia: a comparative study of standard sequencing and locked nucleic acid PCR sequencing on decalcified and nondecalcified formalinfixed paraffin-embedded tissues. Appl Immunohistochem Mol Morphol, 2016, 24: 660–7., DOI 10.1097/PAI.0000000000000242 Wheeler EC, Vora S, Mayer D, Kotini AG, Olszewska M, Park SS, et al. Integrative RNA-omics discovers GNAS alternative splicing as a phenotypic driver of splicing factor–mutant neoplasms. Cancer Discov, 2022, 12:836–55., DOI 10.1158/ 2159-8290.CD-21-0508 Meyer AP, Forrest ME, Nicolau S, Wiszniewski W, Bland MP, Tsao C, et al. Pathogenic missense variants altering codon 336 of GARS1 lead to divergent dominant phenotypes. Hum Mutat, 2022, 43:869–76., DOI 10.1002/humu.24372 Roeber S, Grasbon-Frodl E-M, Windl O, Krebs B, XiangW, Vollmert C, et al. Evidence for a pathogenic role of different mutations at codon 188 of PRNP. PLoS One, 2008, 3:e2147., DOI 10.1371/journal.pone.0002147 White KA, Ruiz DG, Szpiech ZA, Strauli NB, Hernandez RD, Jacobson MP, et al. Cancer-associated arginine-to-histidine mutations confer a gain in pH sensing to mutant proteins. Sci Signal, 2017, 10:eaam9931., DOI 10.1126/scisignal.aam9931 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611553 EP 1611562 DI 10.3389/pore.2024.1611553 PG 10 ER PT J AU Sakurai, Sh Ishida, Y Shintani, T Yamasaki, S Matsui, K Hamana, T Nobumoto, T Yanamoto, S Hayashido, Y AF Sakurai, Shigeru Ishida, Yasutaka Shintani, Tomoaki Yamasaki, Sachiko Matsui, Kensaku Hamana, Tomoaki Nobumoto, Tadayoshi Yanamoto, Souichi Hayashido, Yasutaka TI Clinical significance of integrin αV and β superfamily members and focal adhesion kinase activity in oral squamous cell carcinoma: a retrospective observational study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE prognosis; oral squamous cell carcinoma; integrin β superfamily members; FAK signaling pathway; integrin αv ID prognosis; oral squamous cell carcinoma; integrin β superfamily members; FAK signaling pathway; integrin αv AB Objectives: Integrins are heterodimeric transmembrane plasma membrane proteins composed of α- and β-chains. They bind to extracellular matrix (ECM) and cytoskeletal proteins as ECM protein receptors. Upon ECM protein binding, integrins activate focal adhesion kinase (FAK) and transduce various signals. Despite their importance, integrin and FAK expression in oral squamous cell carcinoma (OSCC) tissue and the prognosis of patients with OSCC remains elusive. Methods: In a retrospective observational study, we immunohistochemically evaluated integrin αV, β1, β3, β5, β6, FAK, and phosphorylated-FAK (pFAK) expressions as prognostic predictors in 96 patients with OSCC. Patients were classified as positive or negative based on staining intensity, and clinicopathologic characteristics and survival rates of the two groups were compared. The association between above integrin-related proteins and PD-1 or PD-L1 in OSCC tissues was investigated. Results: We observed immunohistochemical integrin αV, β1, β6, β8, and FAK expressions in the cell membrane and cytoplasm but not integrin β3 and β5 in the OSCC tissues. pFAK was expressed in the cytoplasm of OSCC cells. The overall survival rate significantly decreased in pFAK-positive OSCC patients compared to the negative group, and cervical lymph node metastasis significantly increased in integrin β8-positive patients with OSCC (p < 0.05). No association between integrin-related proteins and PD-1 or PD-L1 in OSCC tissues was observed. Conclusion: Our results indicate that pFAK and integrin β8 are prognostic factors for OSCC. Therefore, pFAK- and integrin β8-targeting new oral cancer diagnostic and therapeutic methods hold a promising potential. C1 [Sakurai, Shigeru] Hiroshima University, Graduate School of Biomedical and Health Science, Department of Oral OncologyHiroshima, Japan. [Ishida, Yasutaka] Hiroshima University, Graduate School of Biomedical and Health Science, Department of Oral OncologyHiroshima, Japan. [Shintani, Tomoaki] Hiroshima University Hospital, Center of Oral Clinical ExaminationHiroshima, Japan. [Yamasaki, Sachiko] Hiroshima University, Graduate School of Biomedical and Health Science, Department of Oral OncologyHiroshima, Japan. [Matsui, Kensaku] Hiroshima University, Graduate School of Biomedical and Health Science, Department of Oral OncologyHiroshima, Japan. [Hamana, Tomoaki] Hiroshima University, Graduate School of Biomedical and Health Science, Department of Oral OncologyHiroshima, Japan. [Nobumoto, Tadayoshi] Hiroshima University, Graduate School of Biomedical and Health Science, Department of Oral OncologyHiroshima, Japan. [Yanamoto, Souichi] Hiroshima University, Graduate School of Biomedical and Health Science, Department of Oral OncologyHiroshima, Japan. [Hayashido, Yasutaka] Hiroshima University, Graduate School of Biomedical and Health Science, Department of Oral OncologyHiroshima, Japan. RP Hayashido, Y (reprint author), Hiroshima University, Graduate School of Biomedical and Health Science, Department of Oral Oncology, Hiroshima, Japan. EM ishidayas@hiroshima-u.ac.jp CR Chrenek MA, Wong P, Weaver VM. Tumour-stromal interactions. Integrins and cell adhesions as modulators of mammary cell survival and transformation. Breast Cancer Res, 2001, 3:224–9., DOI 10.1186/bcr300 He Y, Liu XD, Chen ZY, Zhu J, Xiong Y, Li K, et al. Interaction between cancer cells and stromal fibroblasts is required for activation of the uPAR-uPA-MMP- 2 cascade in pancreatic cancer metastasis. Clin Cancer Res, 2007, 13:3115–24., DOI 10.1158/1078-0432.Ccr-06-2088 Ingber DE. Cancer as a disease of epithelial-mesenchymal interactions and extracellular matrix regulation. Differentiation, 2002, 70:547–60., DOI 10.1046/j.1432- 36.2002.700908.x 4. Mahadevan D, Von Hoff DD. Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol Cancer Ther, 2007, 6:1186–97., DOI 10.1158/1535-7163.Mct- 06-0686 Shekhar MP, Pauley R, Heppner G. Host microenvironment in breast cancer development: extracellular matrix-stromal cell contribution to neoplastic phenotype of epithelial cells in the breast. Breast Cancer Res, 2003, 5:130–5., DOI 10.1186/bcr580 Smillie CS, Biton M, Ordovas-Montanes J, Sullivan KM, Burgin G, Graham DB, et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell, 2019, 178:714–30., DOI 10.1016/j.cell.2019.06.029 Ahmad K, Lee EJ, Shaikh S, Kumar A, Rao KM, Park SY, et al. Targeting integrins for cancer management using nanotherapeutic approaches: recent advances and challenges. Semin Cancer Biol, 2021, 69:325–36., DOI 10.1016/j. semcancer.2019.08.030 Kechagia JZ, Ivaska J, Roca-Cusachs P. Integrins as biomechanical sensors of the microenvironment. Nat Rev Mol Cel Biol, 2019, 20:457–73., DOI 10.1038/ s41580-019-0134-2 Lewczuk L, Pryczynicz A, Guzinska-Ustymowicz K. Cell adhesion molecules in endometrial cancer - a systematic review. Adv Med Sci, 2019, 64:423–9., DOI 10. 1016/j.advms.2019.08.003 Bair EL, Massey CP, Tran NL, Borchers AH, Heimark RL, Cress AE, et al. Integrin- and cadherin-mediated induction of the matrix metalloprotease matrilysin in cocultures of malignant oral squamous cell carcinoma cells and dermal fibroblasts. Exp Cel Res, 2001, 270:259–67., DOI 10.1006/excr.2001.5347 Berry MG, Gui GP, Wells CA, Carpenter R. Integrin expression and survival in human breast cancer. Eur J Surg Oncol, 2004, 30:484–9., DOI 10.1016/j.ejso.2004. 01.016 Takada Y, Ye X, Simon S. The integrins. Genome Biol, 2007, 8:215., DOI 10. 1186/gb-2007-8-5-215 Barczyk M, Carracedo S, Gullberg D. Integrins. Cell Tissue Res., 2010, 339: 269–80., DOI 10.1007/s00441-009-0834-6 Fu G, Wang W, Luo BH. Overview: structural biology of integrins. Methods Mol Biol, 2012, 757:81–99., DOI 10.1007/978-1-61779-166-6_7 Zhang P, Azizi L, Kukkurainen S, Gao T, Baikoghli M, Jacquier MC, et al. Crystal structure of the FERM-folded talin head reveals the determinants for integrin binding. Proc Natl Acad Sci U S A, 2020, 117:32402–12., DOI 10.1073/ pnas.2014583117 Ziegler WH, Gingras AR, Critchley DR, Emsley J. Integrin connections to the cytoskeleton through talin and vinculin. Biochem Soc Trans, 2008, 36:235–9., DOI 10. 1042/bst0360235 Svitkina T. The actin cytoskeleton and actin-based motility. Cold Spring Harb Perspect Biol, 2018, 10:a018267., DOI 10.1101/cshperspect.a018267 Huang Y, Liao J, Vlashi R, Chen G. Focal adhesion kinase, FAK): its structure, characteristics, and signaling in skeletal system. Cell Signal, 2023, 111:110852., DOI 10.1016/j.cellsig.2023.110852 Peterman E, Gibieza P, Schafer J, Skeberdis VA, Kaupinis A, Valius M, et al. The post-abscission midbody is an intracellular signaling organelle that regulates cell proliferation. Nat Commun, 2019, 10:3181., DOI 10.1038/s41467-019-10871-0 Mitra SK, Schlaepfer DD. Integrin-regulated FAK-Src signaling in normal and cancer cells. Curr Opin Cel Biol, 2006, 18:516–23., DOI 10.1016/j.ceb.2006.08.011 Hehlgans S, Haase M, Cordes N. Signalling via integrins: implications for cell survival and anticancer strategies. Biochim Biophys Acta, 2007, 1775:163–80., DOI 10.1016/j.bbcan.2006.09.001 Wang S, Basson MD. Integrin-linked kinase: a multi-functional regulator modulating extracellular pressure-stimulated cancer cell adhesion through focal adhesion kinase and AKT. Cell Oncol, 2009, 31:273–89., DOI 10.3233/clo-2009-0469 Cavallaro U, Dejana E. Adhesion molecule signalling: not always a sticky business. Nat Rev Mol Cel Biol, 2011, 12:189–97., DOI 10.1038/nrm3068 Mobley AK, McCarty JH. Use of cre-lox technology to analyze integrin functions in astrocytes. Methods Mol Biol, 2012, 814:555–70., DOI 10.1007/978-1- 61779-452-0_37 Bokel C, Brown NH. Integrins in development:moving on, responding to, and sticking to the extracellular matrix. Dev Cel, 2002, 3:311–21., DOI 10.1016/s1534- 5807(02)00265-4 Juliano RL, Reddig P, Alahari S, EdinM, Howe A, Aplin A. Integrin regulation of cell signalling and motility. Biochem Soc Trans, 2004, 32:443–6., DOI 10.1042/ bst0320443 Raymond K, Faraldo MM, Deugnier MA, Glukhova MA. Integrins in mammary development. Semin Cel Dev Biol, 2012, 23:599–605., DOI 10.1016/j. semcdb.2012.03.008 Kelly AJ, Long A. Targeting T-cell integrins in autoimmune and inflammatory diseases. Clin Exp Immunol, 2023, 215:15–26., DOI 10.1093/cei/uxad093 Vay C, Hosch SB, Stoecklein NH, Klein CA, Vallbohmer D, Link BC, et al. Integrin expression in esophageal squamous cell carcinoma: loss of the physiological integrin expression pattern correlates with disease progression. PLoS One, 2014, 9:e109026., DOI 10.1371/journal.pone.0109026 Zhang PF, Zeng GQ, Yi LZ, Liu JP, Wan XX, Qu JQ, et al. Identification of integrin β1 as a prognostic biomarker for human lung adenocarcinoma using 2DLC- MS/MS combined with iTRAQ technology. Oncol Rep, 2013, 30:341–9., DOI 10. 3892/or.2013.2477 Khan S, Qadir M, Khalid A, Ashraf S, Ahmad I. Characterization of cervical tissue using mueller matrix polarimetry. Lasers Med Sci, 2023, 38:46., DOI 10.1007/ s10103-023-03712-6 Fukumoto T, Sakaguchi M, Oka M, Nishimura M, Mukohara T, Nishigori C. Malignant melanoma with bone marrow involvement diagnosed from hypercalcemia: development of a neural cell adhesion molecule stain. J Dermatol, 2017, 44:e105–6., DOI 10.1111/1346-8138.13718 Koistinen P, Ahonen M, Kahari VM, Heino J. AlphaV integrin promotes in vitro and in vivo survival of cells in metastatic melanoma. Int J Cancer, 2004, 112: 61–70., DOI 10.1002/ijc.20377 Lu JG, Sun YN, Wang C, Jin DJ, Liu M. Role of the alpha v-integrin subunit in cell proliferation, apoptosis and tumor metastasis of laryngeal and hypopharyngeal squamous cell carcinomas: a clinical and in vitro investigation. Eur Arch Otorhinolaryngol, 2009, 266:89–96., DOI 10.1007/s00405-008-0675-z Dingemans AM, van den Boogaart V, Vosse BA, van Suylen RJ, Griffioen AW, Thijssen VL. Integrin expression profiling identifies integrin alpha5 and beta1 as prognostic factors in early stage non-small cell lung cancer. Mol Cancer, 2010, 9: 152., DOI 10.1186/1476-4598-9-152 Hayashido Y, Kitano H, Sakaue T, Fujii T, Suematsu M, Sakurai S, et al. Overexpression of integrin αv facilitates proliferation and invasion of oral squamous cell carcinoma cells via MEK/ERK signaling pathway that is activated by interaction of integrin αvβ8 with type Ⅰ collagen. Int J Oncol, 2014, 45:1875–82., DOI 10.3892/ijo.2014.2642 Ramos DM, But M, Regezi J, Schmidt BL, Atakilit A, Dang D, et al. Expression of integrin beta 6 enhances invasive behavior in oral squamous cell carcinoma. Matrix Biol, 2002, 21:297–307., DOI 10.1016/s0945-053x(02)00002-1 Ramos DM, Dang D, Sadler S. The role of the integrin alpha v beta6 in regulating the epithelial to mesenchymal transition in oral cancer. Anticancer Res, 2009, 29:125–30. Li HX, Zheng JH, Fan HX, Li HP, Gao ZX, Chen D. Expression of αvβ6 integrin and collagen fibre in oral squamous cell carcinoma: association with clinical outcomes and prognostic implications. J Oral Pathol Med, 2013, 42: 547–56., DOI 10.1111/jop.12044 Eke I, Dickreuter E, Cordes N. Enhanced radiosensitivity of head and neck squamous cell carcinoma cells by β1 integrin inhibition. Radiother Oncol, 2012, 104:235–42., DOI 10.1016/j.radonc.2012.05.009 Eriksen JG, Steiniche T, Sogaard H, Overgaard J. Expression of integrins and E-cadherin in squamous cell carcinomas of the head and neck. Apmis, 2004, 112: 560–8., DOI 10.1111/j.1600-0463.2004.apm1120902.x Lydiatt WM, Patel SG, O’Sullivan B, Brandwein MS, Ridge JA, Migliacci JC, et al. Head and neck cancers-major changes in the American Joint Committee on cancer eighth edition cancer staging manual. CA Cancer J Clin, 2017, 67:122–37., DOI 10.3322/caac.21389 Ito N, Yamasaki S, Shintani T, Matsui K, Obayashi F, Koizumi K, et al. Tumorinfiltrating CD45RO(+, memory cells are associated with favorable prognosis in oral squamous cell carcinoma patients. Cancers, Basel,, 2023, 15:2221., DOI 10.3390/ cancers15082221 Shintani T, Higaki M, Okamoto T. Heparin-binding protein 17/fibroblast growth factor-binding protein-1 knockout inhibits proliferation and induces differentiation of squamous cell carcinoma cells. Cancers, Basel,, 2021, 13:2684., DOI 10.3390/cancers13112684 Higaki M, Shintani T, Hamada A, Rosli SNZ, Okamoto T. Eldecalcitol, ED-71)- induced exosomal miR-6887-5p suppresses squamous cell carcinoma cell growth by targeting heparin-binding protein 17/fibroblast growth factor-binding protein-1, HBp17/ FGFBP-1). Vitro Cel Dev Biol Anim, 2020, 56:222–33., DOI 10.1007/s11626-020-00440-x Liu J, Cheng S, Zhang Y, Li H, Huang J, Zhang P. Association between polymorphisms in the integrin gene predicted microRNA binding sites and bladder cancer risk. Int J Clin Exp Med, 2014, 7:4398–405. Samanna V, Wei H, Ego-Osuala D, Chellaiah MA. Alpha-V-dependent outside-in signaling is required for the regulation of CD44 surface expression, MMP-2 secretion, and cell migration by osteopontin in humanmelanoma cells. Exp Cel Res, 2006, 312:2214–30., DOI 10.1016/j.yexcr.2006.03.022 Pavlakis E, Neumann M, Merle N, Wieboldt R, Wanzel M, Ponath V, et al. Mutant p53-ENTPD5 control of the calnexin/calreticulin cycle: a druggable target for inhibiting integrin-α5-driven metastasis. J Exp Clin Cancer Res, 2023, 42:203., DOI 10.1186/s13046-023-02785-z Sabeh F, Ota I, Holmbeck K, Birkedal-Hansen H, Soloway P, Balbin M, et al. Tumor cell traffic through the extracellular matrix is controlled by the membrane-anchored collagenase MT1-MMP. J Cel Biol, 2004, 167:769–81., DOI 10.1083/jcb.200408028 Hayashido Y, Urabe K, Yoshioka Y, Kitano H, Okamoto T, Matsuya T. Participation of fibroblasts in MMP-2 binding and activation on the surface of oral squamous cell carcinoma cells. Int J Oncol, 2003, 22:657–62. Kurokawa A, Nagata M, Kitamura N, Noman AA, Ohnishi M, Ohyama T, et al. Diagnostic value of integrin alpha3, beta4, and beta5 gene expression levels for the clinical outcome of tongue squamous cell carcinoma. Cancer, 2008, 112: 1272–81., DOI 10.1002/cncr.23295 Fan QC, Tian H, Wang Y, Liu XB. Integrin-α5 promoted the progression of oral squamous cell carcinoma and modulated PI3K/AKT signaling pathway. Arch Oral Biol, 2019, 101:85–91., DOI 10.1016/j.archoralbio.2019.03.007 Scharinger K, Maxeiner S, Schalla C, Rutten S, Zenke M, Sechi A. LSP1- myosin1e bimolecular complex regulates focal adhesion dynamics and cell migration. Faseb J, 2021, 35:e21268., DOI 10.1096/fj.202000740RR Liu D, Liu S, Fang Y, Liu L, Hu K. Comprehensive analysis of the expression and prognosis for ITGBs: identification of ITGB5 as a biomarker of poor prognosis and correlated with immune infiltrates in gastric cancer. Front Cel Dev Biol, 2021, 9: 816230., DOI 10.3389/fcell.2021.816230 Zhang J, Wang H, Yuan C, Wu J, Xu J, Chen S, et al. ITGAL as a prognostic biomarker correlated with immune infiltrates in gastric cancer. Front Cel Dev Biol, 2022, 10:808212., DOI 10.3389/fcell.2022.808212 Ma SR, Liu JF, Jia R, Deng WW, Jia J. Identification of a favorable prognostic subgroup in oral squamous cell carcinoma: characterization of ITGB4/PD-L1(high, with CD8/PD-1(high). Biomolecules, 2023, 13:1014., DOI 10.3390/biom13061014 Xu C, Xie XL, Kang N, Jiang HQ. Evaluation of ITGB1 expression as a predictor of the therapeutic effects of immune checkpoint inhibitors in gastric cancer. BMC Gastroenterol, 2023, 23:298., DOI 10.1186/s12876-023-02930-0 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611571 EP 1611581 DI 10.3389/pore.2024.1611571 PG 11 ER PT J AU Deng, M Zhang, X Xu, Ch Luo, R Chen, L Zhou, Y Hou, Y AF Deng, Minying Zhang, Xinyi Xu, Chen Luo, Rongkui Chen, Lingli Zhou, Yuhong Hou, Yingyong TI Clinical and pathological observation of conversion therapy for malignant peritoneal mesothelioma: a case report and literature review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE peritoneal malignant mesothelioma; conversion therapy; clinical pathology; delayed interstitial pneumonitis; PD-L1 expression ID peritoneal malignant mesothelioma; conversion therapy; clinical pathology; delayed interstitial pneumonitis; PD-L1 expression AB Background: Malignant mesothelioma (MM) is a tumor originating from the pleura, peritoneum, or pericardial cavity. It is divided into diffuse and localized malignant mesothelioma, with four subtypes in diffuse MM: epithelioid, sarcomatoid, desmoplastic, and biphasic, with biphasic being less common. The onset of this tumor is insidious, and the prognosis is extremely poor in some cases, with a median survival of 6–18 months and no standard treatment options in the past. Aims: We report a case of peritoneal malignant mesothelioma that was successfully treated with transformative therapy. We also review the literature in the hope of providing reference for the treatment and pathological diagnosis of such patients. Methods: The case of the peritoneal malignant mesothelioma was processed and reported in the routine manner for biopsy specimens at different stages. Results and conclusion: We report a case of a malignant tumor originating in the hepatorenal recess, which was diagnosed as biphasic malignant mesothelioma through a biopsy. Immunohistochemical testing showed PDL1 expression. After multidisciplinary discussion, the patient received transformative treatment, including a trial of combined immunotherapy. The tumor significantly shrank, and the patient obtained a chance for curative surgical resection. Microscopic examination showed significant collagenization in the lesion area, with almost no residual tumor. After 19 months of comprehensive treatment, the patient developed multiple fluffy opacities under the pleura of both lungs. Transthoracic core needle biopsy under CT guidance, the pathology showed organizing pneumonia, considering it as delayed interstitial pneumonitis due to immunotherapy based on previous treatment history. Successful comprehensive treatment was achieved for this case of peritoneal malignant mesothelioma, and the patient has been alive without evidence of disease for 33 months, with long-term follow-up. In this process, the pathologist had three opportunities for pathological diagnosis, which required understanding the patient’s medical history, being attentive to the clinical purpose of the specimen, and providing accurate responses to morphological changes at different stages, along with corresponding descriptions and diagnoses to provide effective information for clinical treatment. C1 [Deng, Minying] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Zhang, Xinyi] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Xu, Chen] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Luo, Rongkui] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Chen, Lingli] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. [Zhou, Yuhong] Fudan University, Zhongshan Hospital, Department of Medical OncologyShanghai, China. [Hou, Yingyong] Fudan University, Zhongshan Hospital, Department of PathologyShanghai, China. RP Hou, Y (reprint author), Fudan University, Zhongshan Hospital, Department of Pathology, Shanghai, China. EM hou.yingyong@zs-hospital.sh.cn CR Fernandez-Cuesta L, Mangiante L, Alcala N, Foll M. Challenges in lung and thoracic pathology: molecular advances in the classification of pleural mesotheliomas. Virchows Arch, 2021, 478(1):73–80., DOI 10.1007/s00428-020- 980-9 2. Miller J, Wynn H. A malignant tumour arising from the endothelium of the peritoneum, and producing a mucoid ascitic fluid. J Pathol Bacteriol, 1908, 12: 267–78., DOI 10.1002/path.1700120212 Teta MJ, Mink PJ, Lau E, Sceurman BK, Foster ED. US mesothelioma patterns 1973-2002: indicators of change and insights into background rates. Eur J Cancer Prev, 2008, 17(6):525–34., DOI 10.1097/CEJ. 0b013e3282f0c0a2 Mirarabshahii P, Pillai K, Chua TC, Pourgholami MH, Morris DL. Diffuse malignant peritoneal mesothelioma--an update on treatment. Cancer Treat Rev, 2012, 38(6):605–12., DOI 10.1016/j.ctrv.2011.10.006 Naka H, Naka A. [Clinicopathological study on 100 Japanese patients with peritoneal mesothelioma in Japan]. Gan No Rinsho, 1984, 30:1–10. Metintas M, Hillerdal G, Metintas S. Malignant mesothelioma due to environmental exposure to erionite: follow-up of a Turkish emigrant cohort. Eur Respir J, 1999, 13(3):523–6., DOI 10.1183/09031936.99.13352399 Stey C, Landolt-Weber U, Vetter W, Sauter C, Marincek B. Malignant peritoneal mesothelioma after Thorotrast exposure. Am J Clin Oncol, 1995, 18(4):313–7., DOI 10.1097/00000421-199508000-00009 Weissmann LB, Corson JM, Neugut AI, Antman KH. Malignant mesothelioma following treatment for Hodgkin’s disease. J Clin Oncol, 1996, 14(7):2098–100., DOI 10.1200/JCO.1996.14.7.2098 Shivapurkar N, Wiethege T, Wistuba II, Salomon E, Milchgrub S, Muller KM, et al. Presence of simian virus 40 sequences in malignant mesotheliomas and mesothelial cell proliferations. J Cell Biochem, 1999, 76(2):181–8., DOI 10.1002/(sici, 1097-4644(20000201)76:2<181::aid-jcb2>3.3.co;2-0 Marinaccio A, Montanaro F, Mastrantonio M, Uccelli R, Altavista P, Nesti M, et al. Predictions of mortality from pleural mesothelioma in Italy: a model based on asbestos consumption figures supports results from age-period-cohort models. Int J Cancer, 2005, 115(1):142–7., DOI 10.1002/ijc.20820 Price B, Ware A. Special article. J Am Med Assoc, 1946, 131(13):1053., DOI 10. 1001/jama.1946.02870300021006 Wagner JC, Sleggs CA, Marchand P. Diffuse pleural mesothelioma and asbestos exposure in the north western cape province. Br J Ind Med, 1960, 17(4):260–71., DOI 10.1136/oem.17.4.260 Acherman YI, Welch LS, Bromley CM, Sugarbaker PH. Clinical presentation of peritonealmesothelioma. Tumori, 2003, 89(3):269–73., DOI 10.1177/030089160308900307 Manzini VP, Recchia L, Cafferata M, Porta C, Siena S, Giannetta L, et al. Malignant peritoneal mesothelioma: a multicenter study on 81 cases. Ann Oncol, 2010, 21(2):348–53., DOI 10.1093/annonc/mdp307 Chinese Society of Clinical Oncology Abdominal Tumor Professional Committee, Chinese Society of Clinical Oncology Tumor Hyperthermia Professional Committee, Beijing Cancer Prevention Society Tumor Hyperthermia Professional Committee. Chinese expert consensus on the diagnosis and treatment of diffuse malignant peritoneal mesothelioma. Chin Med J, 2021, 101(36):2839–49., DOI 10.3760/cma.j.cn112137-20210210-00405 Offin M, Yang SR, Egger J, Jayakumaran G, Spencer RS, Lopardo J, et al. Molecular characterization of peritoneal mesotheliomas. J Thorac Oncol, 2022, 17(3):455–60., DOI 10.1016/j.jtho.2021.09.012 Joseph NM, Chen YY, Nasr A, Yeh I, Talevich E, Onodera C, et al. Genomic profiling of malignant peritoneal mesothelioma reveals recurrent alterations in epigenetic regulatory genes BAP1, SETD2, and DDX3X. Mod Pathol, 2017, 30(2): 246–54., DOI 10.1038/modpathol.2016.188 Panagopoulos I, Thorsen J, Gorunova L, Micci F, Haugom L, Davidson B, et al. RNA sequencing identifies fusion of the EWSR1 and YY1 genes in mesothelioma with t(14;22)(q32;q12). Genes Chromosomes Cancer, 2013, 52(8):733–40., DOI 10. 1002/gcc.22068 Rodriguez D, Cheung MC, Housri N, Koniaris LG. Malignant abdominal mesothelioma: defining the role of surgery. J Surg Oncol, 2009, 99(1):51–7., DOI 10. 1002/jso.21167 DeSantis CE, Kramer JL, Jemal A. The burden of rare cancers in the United States. CA Cancer J Clin, 2017, 67(4):261–72., DOI 10.3322/caac.21400 Feldman AL, Libutti SK, Pingpank JF, Bartlett DL, Beresnev TH, Mavroukakis SM, et al. Analysis of factors associated with outcome in patients with malignant peritoneal mesothelioma undergoing surgical debulking and intraperitoneal chemotherapy. J Clin Oncol, 2003, 21(24):4560–7., DOI 10.1200/JCO.2003.04.150 Cao S, Jin S, Cao J, Shen J, Hu J, Che D, et al. Advances in malignant peritoneal mesothelioma. Int J Colorectal Dis, 2015, 30(1):1–10., DOI 10.1007/s00384-014-2029-1 Turner K, Varghese S, Alexander HR, Jr. Current concepts in the evaluation and treatment of patients with diffuse malignant peritoneal mesothelioma. J Natl Compr Canc Netw, 2012, 10(1):49–57., DOI 10.6004/jnccn.2012.0008 Janne PA,Wozniak AJ, Belani CP, Keohan ML, Ross HJ, Polikoff JA, et al. Open-label study of pemetrexed alone or in combination with cisplatin for the treatment of patients with peritoneal mesothelioma: outcomes of an expanded access program. Clin Lung Cancer, 2005, 7(1):40–6., DOI 10.3816/CLC.2005. n.020 Raghav K, Liu S, Overman MJ, Willett AF, Knafl M, Fu SC, et al. Efficacy, safety, and biomarker analysis of combined PD-L1, atezolizumab, and VEGF, bevacizumab, blockade in advanced malignant peritoneal mesothelioma. Cancer Discov, 2021, 11(11):2738–47., DOI 10.1158/2159-8290.CD-21-0331 Hassan R, Thomas A, Nemunaitis JJ, Patel MR, Bennouna J, Chen FL, et al. Efficacy and safety of avelumab treatment in patients with advanced unresectable mesothelioma: phase 1b results from the JAVELIN solid tumor trial. JAMA Oncol, 2019, 5(3):351–7., DOI 10.1001/jamaoncol.2018.5428 Fennell DA, Ewings S, Ottensmeier C, Califano R, Hanna GG, Hill K, et al. Nivolumab versus placebo in patients with relapsed malignant mesothelioma, CONFIRM): a multicentre, double-blind, randomised, phase 3 trial. Lancet Oncol, 2021, 22(11):1530–40., DOI 10.1016/S1470- 2045(21)00471-X White MG, Schulte JJ, Xue L, Berger Y, Schuitevoerder D, Vining CC, et al. Heterogeneity in PD-L1 expression in malignant peritoneal mesothelioma with systemic or intraperitoneal chemotherapy. Br J Cancer, 2021, 124(3):564–6., DOI 10. 1038/s41416-020-01130-x de Boer NL, van Kooten JP, Burger JWA, Verhoef C, Aerts JGJV, Madsen EVE. Adjuvant dendritic cell based immunotherapy, DCBI, after cytoreductive surgery, CRS, and hyperthermic intraperitoneal chemotherapy, HIPEC, for peritoneal mesothelioma, a phase II single centre open-label clinical trial: rationale and design of the MESOPEC trial. BMJ Open, 2019, 9(5):e026779., DOI 10.1136/bmjopen-2018-026779 Valmary-Degano S, Colpart P, Villeneuve L, Monnien F, M’Hamdi L, Lang Averous G, et al. Immunohistochemical evaluation of two antibodies against PD-L1 and prognostic significance of PD-L1 expression in epithelioid peritoneal malignant mesothelioma: a RENAPE study. Eur J Surg Oncol, 2017, 43(10):1915–23., DOI 10. 1016/j.ejso.2017.05.009 Si J,Wang A, Li X, Fang F, Liu J, Xia Y, et al. A case of cardiac toxicity caused by immune checkpoint inhibitors in the treatment of malignant mesothelioma. Chin J Heart Fail Cardiomyopathy, 2019, 3(4):228–30. English and Chinese Edition., DOI 10.3760/cma.j.issn.2096-3076.2019.04.008 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611577 EP 1611584 DI 10.3389/pore.2023.1611577 PG 8 ER PT J AU Moldvay, J Timar, J AF Moldvay, Judit Timar, Jozsef TI KRASG12C mutant lung adenocarcinoma: unique biology, novel therapies and new challenges SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE KRAS; lung adenocarcinoma; G12C mutation; sotorasib; adagrasib ID KRAS; lung adenocarcinoma; G12C mutation; sotorasib; adagrasib AB KRAS mutant lung cancer is the most prevalent molecular subclass of adenocarcinoma (LUAD), which is a heterogenous group depending on the mutation-type which affects not only the function of the oncogene but affects the biological behavior of the cancer as well. Furthermore, KRAS mutation affects radiation sensitivity but leads also to bevacizumab and bisphosphonate resistance as well. It was highly significant that allele specific irreversible inhibitors have been developed for the smoking associated G12C mutant KRAS (sotorasib and adagrasib). Based on trial data both sotorasib and adagrasib obtained conditional approval by FDA for the treatment of previously treated advanced LUAD. Similar to other target therapies, clinical administration of KRASG12C inhibitors (sotorasib and adagrasib) resulted in acquired resistance due to various genetic changes not only in KRAS but in other oncogenes as well. Recent clinical studies are aiming to increase the efficacy of G12C inhibitors by novel combination strategies. C1 [Moldvay, Judit] National Koranyi Institute of PulmonologyBudapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM jtimar@gmail.com CR Timar J, Kashofer K. Molecular epidemiology and diagnostics of KRAS mutations in human cancer. Cancer Metast Rev, 2020, 39:1029–38., DOI 10.1007/ s10555-020-09915-5 Ceddia S, Landi L, Cappuzzo F. KRAS-mutant non-small cell lung cancer: from past efforts to future challenges. Int J Mol Sci, 2022, 23:9391., DOI 10.3390/ ijms23169391 Nicholson AG, Tsao MS, Beasley MB, Borczuk AC, Brambilla E, Cooper WA, et al. The 2021 WHO classification of lung tumors: impact of advances since 2015. J Thorac Oncol, 2021, 17:362–87., DOI 10.1016/j.jtho.2021.11.003 Hathaway F, Martins R, Sorcher S, Bzura A, Dudbridge F, Fenell DA. Family matters: germline testing in thoracic cancer. ASCO EDBK, 2023, 389956:2023. Moore AR, Rosenberg SC, McCormick F, Malek S. RAS-targeted therapies: is the undruggable drugged? Nat Rev Drug Discov, 2020, 19:533–52., DOI 10.1038/ s41573-020-0068-6 Timar J. The clinical relevance of KRAS gene mutation in non-small-cell lung cancer. Curr Opin Oncol, 2014, 26:138–44., DOI 10.1097/CCO.0000000000000051 Skoulidis F, Byers LA, Diao L, Papadimitrakopoulou VA, Tong P, Izzo J, et al. Co-occurring genomic alterations define major subsets of KRAS mutant lung adenocarcinoma with distinct biology, immune profiles and therapeutic vulnerabilities. Cancer Discov, 2015, 5:860–77., DOI 10.1158/2159-8290.CD-14-1236 Lim TLH, Skoulidis F, Kerr KM, Ahn M-I, Kapp JR, Soares FA, et al. KRAS G12C in advanced NSCLC: prevalence, co-mutations and testing. Lung Cancer, 2023, 184:107293., DOI 10.1016/j.lungcan.2023.107293 Estcott PM, Hallwill KD, To MD, Rashid M, Rust AG, Keane TM, et al. The mutational landscapes of genetic and chemical models of Kras-driven lung cancer. Nature, 2015, 517:489–92., DOI 10.1038/nature13898 Cserepes M, Ostoros G, Lohinai Z, Raso E, Barbai T, Timar J, et al. Subtypespecific KRAS mutations in advanced lung adenocarcinoma: a retrospective study of patients treated with platinum-based chemotherapy. Eur J Cancer, 2014, 50: 1819–28., DOI 10.1016/j.ejca.2014.04.001 Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. Biochemical and structural analysis of common cancer-associated KRAS mutations. Mol Cancer Res, 2015, 13:1325–35., DOI 10.1158/1541-7786.MCR-15-0203 Burgess MR, Hwang E, mrou R, Bielski CM, Wandler A, Huang BJ, et al. KRAS allelic imbalance enhances fitness and modulates MAP kinase dependence in cancer. Cell, 2017, 168:817–29.e15., DOI 10.1016/j.cell.2017.01.020 Temko D, Tomlison JPM, Severini S, Schuster-Bockler B, Graham TA. The effects of mutational processes and selection on driver mutations across cancer types. Nat Comm, 2018, 9:1857., DOI 10.1038/s41467-018-04208-6 Lohinai Z, Klikovits T, Moldvay J, Ostoros G, Raso E, Timar J, et al. KRASmutation incidence and prognostic value are metastatic site-specific in lung adenocarcinoma: poor prognosis in patients with KRAS mutation and bone metastasis. Sci Rep, 2017, 7:39721., DOI 10.1038/srep39721 Ghimessy AK, Gellert A, Schleghl E, Hegedus B, Raso E, Barbai T, et al. KRAS mutations predict response and outcome in advanced lung adenocarcinoma patients receiving first-line bevacizumab and platinum-based chemotherapy. Cancers, 2019, 11:1514., DOI 10.3390/cancers11101514 Radeczky P, Medgyesfalvi Z, Laszlo V, Fillinger J, Moldvay J, Raso E, et al. The effects of bisphosphonate and radiation therapy in bone-metastatic lung adenocarcinoma: the impact of KRAS mutation. Transl Lung Cancer Res, 2021, 10:675–84., DOI 10.21037/tlcr-20-754 Kenessey I, Koi K, Horvath O, Cserepes M, Molnar D, Izsak V, et al. KRASmutation status dependent effect of zoledronic acid in human non-small cell cancer preclinical models. Oncotarget, 2016, 7:79503–14., DOI 10.18632/oncotarget.12806 Ricciuti B, Alessi JV, Elkrief A, Wang X, cortellini A, Li YY, et al. Dissecting the clinicopathologic, genomic and immunophenotypic correlates of KRASG12Dmutated non-small-cell lung cancer. Ann Oncol, 2022, 33:1029–40., DOI 10.1016/j. annonc.2022.07.005 Rathod LS, Dabhade PS, Mokale SN. Recent progress in targeting KRAS mutant cancers with covalent G12C-specific inhibitors. Drug Discov Today, 2023, 28:103557., DOI 10.1016/j.drudis.2023.103557 Osrem JM, Peters U, sos ML, Wells JA, Shokat KM. K-Ras(G12C, inhibitors allosterically control GTP affinity and effector interactions. Nature, 2013, 503: 548–51. Canon J, Rex K, Saiki A, Mohr C, Cooke K, Bagal D, et al. The clinical KRAS(G12C, inhibitorAMG 510 drives anti-tumour immunity. Nature, 2019, 575: 217–23., DOI 10.1038/s41586-019-1694-1 Fell JB, Fischer JP, Baer BR, Blake JF, Bouhana K, Briere DM, et al. Identification of the clinical development candidate MRTX849, a covalent KRASG12C inhibitor for the treatment of cancer. J Med Chem, 2020, 63: 6679–93., DOI 10.1021/acs.jmedchem.9b02052 Kwan AK, Piazza GA, Keeton AB, leite CA. The path to the clinic: a comprehensive review on direct KRASG12C inhibitors. J Exp Clin Cancer Res, 2022, 41:27., DOI 10.1186/s13046-021-02225-w Sacher A, LoRusso P, Patel MR, Miller W, Garralda E, Forster MD, et al. Single-agent divarasib, GDC-6036, in solid tumors with a KRAS G12C mutation. N Engl J Med, 2023, 389:710–21., DOI 10.1056/NEJMoa2303810 Wang X, Allen S, Blake JF, Browcut V, Briene DM, Calinisan A, et al. Identification of MRTX1133, a noncovalent, potent, and selective KRAS(G12D, inhibitor. J Med Chem, 2022, 65:3123–33., DOI 10.1021/acs.jmedchem.1c01688 Mullard A. The KRAS crowd targets its next cancer mutations. Nat Drug Discov, 2023, 22:167–71., DOI 10.1038/d41573-023-00015-x Tran TH, Alexander P, Dharmaiah S, Agamasu C, Nissley DV, McCormick F, et al. The small molecule BI-2852 induces a nonfunctional dimer of KRAS. PNAS, 2020, 117:3363–4., DOI 10.1073/pnas.1918164117 Poh A. Drugging RAS: moving beyond KRASG12C. Cancer Discov, 2023)., DOI 10.1158/2159-8290.CD.ND2023-0010 Hillig RC, Sautier B, Schroeder J,Moosmayer D, Hilpmann A, Stegmann CM, et al. Discovery of potent SOS1 inhibitors that block RAS activation via disruption of the RAS-SOS1 interaction. PNAS, 2019, 116:2551–60., DOI 10.1073/pnas. 1812963116 Drilon A, Sharma MR, Johnson ML, Yap TA, Gadgeel S, Nepert D, et al. SHP2 inhibition sensitizes diverse oncogene-addicted solid tumors to Re-treatment with targeted therapy. Cancer Discov, 2023, 13:1789–801., DOI 10.1158/2159-8290. CD-23-0361 Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, et al. Sotorasib for lung cancers with KRAS p.G12C mutation. N Engl J Med, 2021, 384:2371–81., DOI 10.1056/NEJMoa2103695 Janne PA, Riely GJ, Gadgeel SM, Heist RS, Ou SHI, Pacheco JM, et al. Adagrasib in non-small-cell lung cancer harboring a KRASG12C mutation. N Engl J Med, 2022, 387:120–31., DOI 10.1056/NEJMoa2204619 Oya Y, Mitsudomi T. Is adagrasib just another sotorasib?or, should we differentiate their usage according to patients’ clinical presentation? Transl Lung Cancer Res, 2023, 12:940–3., DOI 10.21037/tlcr-23-97 de Langen AJ, Johnson ML, Mazieres J, Dingemans A-M, Mountzios G, Pless M, et al. Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRASG12C mutation: a randomised, open-label, phase 3 trial. Lancet, 2023, 401:733–46., DOI 10.1016/S0140-6736(23)00221-0 Mok TSK, Lawler WE, Shum MK, Dakhil SR, Spira AI, Barlesi F, et al. KRYSTAL-12: a randomized phase 3 study of adagrasib, MRTX849, versus docetaxel in patients, pts, with previously treated non-small-cell lung cancer, NSCLC, with KRASG12C mutation. J Clin Oncol, 2021, 39:TPS9129., DOI 10. 1200/jco.2021.39.15_suppl.tps9129 Sabari JK, Velcheti V, Shimizu K, Strickland MR, Heist RS, Singh M, et al. Activity of adagrasib, MRTX849, in brain metastases: preclinical models and clinical data from patients with KRASG12C-mutant non-small cell lung cancer. Clin Cancer Res Í, 2022, 28:3318–28., DOI 10.1158/1078-0432.CCR- 22-0383 Patil T, Smith DE, Bunn PA, Aisner DL, Le AT, Hancock M, et al. The incidence of brain metastases in stage IV ROS1-rearranged non-small cell lung cancer and rate of central nervous system progression on crizotinib. J Thorac Oncol, 2018, 13:1717–26., DOI 10.1016/j.jtho.2018.07.001 Arrieta O, Ramirez-Tirado LA, Caballe-Perez E, Mejia-Perez A, Zatarain- Barron ZL, Cardona AF, et al. Response rate of patients with baseline brain metastases from recently diagnosed non-small cell lung cancer receiving radiotherapy according to EGFR, ALK and KRAS mutation status. Thorac Cancer, 2020, 11:1026–37., DOI 10.1111/1759-7714.13359 Ramalingam S, Skoulidis F, Govindan R, Velcheti V, Li B, Besse B, et al. P52.03 efficacy of sotorasib in KRAS p.G12C-mutated NSCLC with stable brain metastases: a post-hoc analysis of CodeBreaK 100. J Thorac Oncol, 2021, 16:S1123., DOI 10.1016/j.jtho.2021.08.547 Koster KL, Appenzeller C, Lauber A, Fruh M, Schmid S. Sotorasib shows intracranial activity in patients with KRAS G12C-mutated adenocarcinoma of the lung and untreated active brain metastases. Case Rep Oncol, 2022, 15:720–5., DOI 10. 1159/000525341 Yeh J, Marks JA, Alzeer AH, Sloan EA, Varghese R, Paudel N, et al. Remarkable intracranial response to sotorasib in a patient with KRAS, G12C)- Mutated lung adenocarcinoma and untreated brain metastases: a case report. JTO Clin Res Rep, 2022, 3:100428., DOI 10.1016/j.jtocrr.2022.100428 Negrao MV, Spira AI, Heist RS, Janne PA, Pacheco JM, Weiss J, et al. Intracranial efficacy of adagrasib in patients from the KRYSTAL-1 trial with KRASG12C-Mutated non-small-cell lung cancer who have untreated CNS metastases. J Clin Oncol, 2023, 41:4472–7., DOI 10.1200/JCO.23.00046 Arbour KC, Jordan E, Kim HR, Dienstag J, Yu HA, Sanchez-Vega F, et al. Effects of Co-occurring genomic alterations on outcomes in patients with KRASmutant non-small cell lung cancer. Clin Cancer Res, 2018, 24:334–40., DOI 10.1158/ 1078-0432.CCR-17-1841 Luo J, Osrem J, Pellini B, Imbody D, Stern Y, Solanki HS, et al. Overcoming KRAS-mutant lung cancer. Am Soc Clin Oncol Educ Book, 2022, 10:700–10., DOI 10. 1200/edbk_360354 Negrao MV, Araujo HA, Lamberti G, Cooper AJ, Akhave NS, Zhou T, et al. Comutations and KRASG12C inhibitor efficacy in advanced NSCLC. Cancer Discov, 2023, 13:1556–71., DOI 10.1158/2159-8290.CD-22-1420 Zhao Y, Murciano-Goroff YR, Xue JY, Ang A, Lucas J, Mai TT, et al. Diverse alterations associated with resistance to KRASG12C inhibition. Nature, 2021, 599: 679–83., DOI 10.1038/s41586-021-04065-2 Tanaka N, Lin JJ, Li C, Ryan MB, Zhang J, Kiedrowski LA, et al. Clinical acquired resistance to KRASG12C inhibition through a novel KRAS switch-II pocket mutation and polyclonal alterations converging on RAS-MAPK reactivation. Cancer Discov, 2021, 11:1913–22., DOI 10.1158/2159-8290.CD-21-0365 Ho CSL, Tuns AI, Schildhaus H-U, Wiesweg M, Gruner BM, Hegedus B, et al. HER2 mediates clinical resistance to the KRASG12C inhibitor sotorasib, which is overcome by co-targeting SHP2. Eur J Cancer, 2021, 159:16–23., DOI 10.1016/j.ejca. 2021.10.003 Awad MM, Liu S, Rybkin II, Arbour KC, Dilly J, Zhu VW, et al. Acquired resistance to KRASG12C inhibition in cancer. N Engl J Med, 2021, 384:2382–93., DOI 10.1056/NEJMoa2105281 Koga T, Suda K, Fujino T, Ohara S, Hamada A, Nishino M, et al. KRAS secondary mutations that confer acquired resistance to KRAS G12C inhibitors, sotorasib and adagrasib, and overcoming strategies: insights from in vitro experiments. J Thorac Oncol, 2021, 16:1321–32., DOI 10.1016/j.jtho.2021.04.015 Lietman CD, Johnson ML, McCormick F, Lindsay CR. More to the RAS story: KRASG12C inhibition, resistance mechanisms and moving beyond KRASG12C. Am Soc Clin Oncol Educ Book, 2022, 42:1–13., DOI 10.1200/EDBK_351333 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611580 EP 1611588 DI 10.3389/pore.2023.1611580 PG 9 ER PT J AU Kovacs, Trasi, K Barabas, M Gal, K Csiki, E Sipos, D Papp, J Simon, M AF Kovacs, Arpad Trasi, Krisztina Barabas, Marton Gal, Kristof Csiki, Emese Sipos, David Papp, Judit Simon, Mihaly TI LINAC-based SBRT in treating early-stage NSCLC patients— single institution experience and survival data analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE VATS; NSCLC; SBRT; LINAC-based SBRT; early stage ID VATS; NSCLC; SBRT; LINAC-based SBRT; early stage AB Aim: This single institute prospective study aimed to evaluate the feasibility of LINAC-based stereotactic body radiotherapy (SBRT) in treating patients with early-stage non-small cell lung cancer (NSLSC). We focused on the survival data with the local and distant control profiles and the cancer- and non-cancerspecific survival. Treatment-related side effects were also collected and analyzed. Methods: Patients with early-stage NSCLC between January 2018 and October 2021 were included in our prospective study; a total of 77 patients receiving LINAC-based SBRT were analyzed. All patients had pretreatment multidisciplinary tumor board decisions on SBRT. The average patient age was 68.8 years (median: 70 years, range: 52–82); 70 patients were in ECOG 0 status (91%), while seven patients were in ECOG 1-2 status (9%). 52% of the patients (40) had histologically verified NSCLC, and the other 48% were verified based on PETCT results. We applied the SBRT scheme 8 x 7.5 Gy for central tumors (74%) or 4 x 12 Gy for peripheral tumors (26%). Results: The mean follow-up time was 25.4 months (median 23, range 18–50). The Kaplan-Meier estimation for overall survival in patients receiving LINAC-based SBRTwas 41.67months.Of the 77 patients treated by SBRT, deathwas reported for 17 patients (9 cases cancer-specific, 8 cases non-cancer specific reason). The mean local tumor control was 34.25months (range 8.4–41), and the mean systemic control was 24.24months (range 7–25). During the treatments, no Grade I-II were reported; in 30 cases, Grade I non-symptomatic treatmentrelated lung fibrosis and two asymptomatic rib fractures were reported. Conclusion: In the treatment of early-stage NSCLC, LINAC-based SBRT can be a feasible alternative to surgery. Although we reported worse OS data in our patient cohort compared to the literature, the higher older average age and the initial worse general condition (ECOG1-2) in our patient cohort appear to be the reason for this difference. With the comparable local control and survival data and the favorable side effect profile, SBRT might be preferable over surgery in selected cases. C1 [Kovacs, Arpad] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Trasi, Krisztina] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Barabas, Marton] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Gal, Kristof] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Csiki, Emese] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Sipos, David] Pecsi Tudomanyegyetem, Egeszsegtudomanyi Kar, Doktori IskolaPecs, Hungary. [Papp, Judit] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Simon, Mihaly] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. RP Kovacs, (reprint author), Debreceni Egyetem, Onkologiai Klinika, Sugarterapia Osztaly, Debrecen, Hungary. EM kovacsarpad@med.unideb.hu CR LuCE Europe. Report on lung cancer: challenges in lung cancer in Europe. 6th Edn, 2021). Available from: https://content/uploads/2017/10/LuCE-Report-final. pdf, Accessed November, 2021). Dyba T, Randi G, Bray F, Martos C, Giusti F, Nicholson N, et al. The European cancer burden in 2020: incidence and mortality estimates for 40 countries and 25 major cancers. Eur J Cancer, 2021, 157:308–47., DOI 10.1016/j.ejca.2021.07.039 Molina JR, Yang P, Cassivi SD, Schild SE, Adjei AA. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc, 2008, 83(5):584–94., DOI 10.4065/83.5.584 Bareschino MA, Schettino C, Rossi A, Maione P, Sacco PC, Zeppa R, et al. Treatment of advanced non small cell lung cancer. J Thorac Dis, 2011, 3(2):122–33., DOI 10.3978/j.issn.2072-1439.2010.12.08 Dong B, Zhu X, Shu Z, Ji Y, Lu F, Wang J, et al. Video-assisted thoracoscopic lobectomy versus stereotactic body radiotherapy treatment for early-stage nonsmall cell lung cancer: a propensity score-matching analysis. Front Oncol, 2020, 10: 585709., DOI 10.3389/fonc.2020.585709 Cai Y, Fu X, Xu Q, Sun W, Zhang N. Thoracoscopic lobectomy versus open lobectomy in stage I non-small cell lung cancer: a meta-analysis. PLoS One, 2013, 8(12):e82366., DOI 10.1371/journal.pone.0082366 Milano MT, Kong FMS, Movsas B. Stereotactic body radiotherapy as salvage treatment for recurrence of non-small cell lung cancer after prior surgery or radiotherapy. Transl Lung Cancer Res, 2018, 8(1):78–87., DOI 10.21037/tlcr.2018.08.15 Ball D, Mai GT, Vinod S, Babington S, Ruben J, Kron T, et al. Stereotactic ablative radiotherapy versus standard radiotherapy in stage 1 non-small-cell lung cancer, TROG 09.02 CHISEL): a phase 3, open-label, randomised controlled trial. Lancet Oncol, 2019, 20(4):494–503., DOI 10.1016/S1470-2045(18)30896-9 Boada M, Guzman R, Montesinos M, Libreros A, Guirao A, Sanchez-Lorente D, et al. Upstaging, centrality and survival in early stage non-small cell lung cancer video-assisted surgery. Lung Cancer, 2019, 134:254–8., DOI 10.1016/j.lungcan.2019. 06.030 Chi A, Fang W, Sun Y, Wen S. Comparison of long-term survival of patients with early-stage non-small cell lung cancer after surgery vs stereotactic body radiotherapy. JAMA Netw Open, 2019, 2(11):e1915724., DOI 10.1001/ jamanetworkopen.2019.15724 Hamaji M, Chen F, Matsuo Y, Kawaguchi A, Morita S, Ueki N, et al. Video-assisted thoracoscopic lobectomy versus stereotactic radiotherapy for stage I lung cancer. Ann Thorac Surg, 2015, 99(4):1122–9., DOI 10.1016/j. athoracsur.2014.11.009 Tandberg DJ, Tong BC, Ackerson BG, Kelsey CR. Surgery versus stereotactic body radiation therapy for stage I non-small cell lung cancer: a comprehensive review. Cancer, 2018, 124(4):667–78., DOI 10.1002/cncr.31196 Davidson-Pilon C. Lifelines: survival analysis in Python. J Open Source Softw, 2019, 4(40):1317., DOI 10.21105/joss.01317 Zarogoulidis K, Zarogoulidis P, Darwiche K, Boutsikou E, Machairiotis N, Tsakiridis K, et al. Treatment of non-small cell lung cancer, NSCLC). J Thorac Dis, 2013, 5(4):2–9., DOI 10.3978/j.issn.2072-1439.2013.07.10 Darling GE, Decker PA, Ballman K, Malthaner RA, Inculet RI, Jones DR, et al. Number of lymph nodes harvested from a mediastinal lymphadenectomy: results of the randomized, prospective American College of Surgeons Oncology Group Z0030 trial. Chest, 2011, 139(5):1124–9., DOI 10.1378/chest.10-0859 Howington JA, Blum MG, Chang AC, Balekian AA, Murthy SC. Treatment of stage I and II non-small cell lung cancer: diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest, 2013, 143(5 Suppl): e278S–e313S., DOI 10.1378/chest.12-2359 Detillon DDEMA, Veen EJ. Postoperative outcome after pulmonary surgery for non-small cell lung cancer in elderly patients. Ann Thorac Surg, 2018, 105(1): 287–93., DOI 10.1016/j.athoracsur.2017.07.032 Burt BM, Kosinski AS, Shrager JB, Onaitis MW, Weigel T. Thoracoscopic lobectomy is associated with acceptable morbidity and mortality in patients with predicted postoperative forced expiratory volume in 1 second or diffusing capacity for carbon monoxide less than 40% of normal. J Thorac Cardiovasc Surg, 2014, 148(1):19–28. discussion 28- 29.e1., DOI 10.1016/j.jtcvs.2014.03.007 Nwogu CE, Pang H, Gu L, Wang X, Richards WG, Veit LJ, et al. VATS lobectomy has better perioperative outcomes than open lobectomy: CALGB 31001, an ancillary analysis of CALGB 140202, Alliance). Ann Thorac Surg, 2015, 99(2): 399–405., DOI 10.1016/j.athoracsur.2014.09.018 Prezzano KM, Ma SJ, Hermann GM, Rivers CI, Gomez-Suescun JA, Singh AK. Stereotactic body radiation therapy for non-small cell lung cancer: a review. World J Clin Oncol, 2019, 10(1):14–27., DOI 10.5306/wjco.v10.i1.14 Vansteenkiste J, Crino L, Dooms C, Douillard JY, Faivre-Finn C, Lim E, et al. 2nd ESMO Consensus Conference on Lung Cancer: early-stage non-small-cell lung cancer consensus on diagnosis, treatment and follow-up. Ann Oncol Off J Eur Soc Med Oncol, 2014, 25(8):1462–74., DOI 10.1093/annonc/mdu089 Grills IS, Mangona VS, Welsh R, Chmielewski G, McInerney E, Martin S, et al. Outcomes after stereotactic lung radiotherapy or wedge resection for stage I non-small-cell lung cancer. J Clin Oncol, 2010, 28(6):928–35., DOI 10. 1200/JCO.2009.25.0928 Chua GWY, Chua KLM. Which patients benefit most from stereotactic body radiotherapy or surgery in medically operable non-small cell lung cancer? An indepth look at patient characteristics on both sides of the debate. Thorac Cancer, 2019, 10(10):1857–67., DOI 10.1111/1759-7714.13160 Stokes WA, Bronsert MR, Meguid RA, Blum MG, Jones BL, Koshy M, et al. Post-treatment mortality after surgery and stereotactic body radiotherapy for earlystage non-small-cell lung cancer. J Clin Oncol, 2018, 36(7):642–51., DOI 10.1200/ JCO.2017.75.6536 Mayne NR, Darling AJ, Raman V, Patel DC, Liou DZ, D’Amico TA, et al. Stereotactic body radiotherapy versus delayed surgery for early-stage non-small-cell lung cancer. Ann Surg, 2020, 272(6):925–9., DOI 10.1097/SLA.0000000000004363 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611589 EP 1611594 DI 10.3389/pore.2024.1611589 PG 6 ER PT J AU Kumbrink, J Demes, MCh Jeroch, J Brauninger, A Hartung, K Gerstenmaier, U Marienfeld, R Hillmer, A Bohn, N Lehning, Ch Ferch, F Wild, P Gattenlohner, S Moller, P Klauschen, F Jung, A AF Kumbrink, Jorg Demes, Melanie-Christin Jeroch, Jan Brauninger, Andreas Hartung, Kristin Gerstenmaier, Uwe Marienfeld, Ralf Hillmer, Axel Bohn, Nadine Lehning, Christina Ferch, Ferdinand Wild, Peter Gattenlohner, Stefan Moller, Peter Klauschen, Frederick Jung, Andreas TI Development, testing and validation of a targeted NGS-panel for the detection of actionable mutations in lung cancer (NSCLC) using anchored multiplex PCR technology in a multicentric setting SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lung cancer; targeted therapy; next-generation sequencing panel; anchored multiplex PCR; actionable mutations ID lung cancer; targeted therapy; next-generation sequencing panel; anchored multiplex PCR; actionable mutations AB Lung cancer is a paradigm for a genetically driven tumor. A variety of drugs were developed targeting specific biomarkers requiring testing for tumor genetic alterations in relevant biomarkers. Different next-generation sequencing technologies are available for library generation: 1) anchored multiplex-, 2) amplicon based- and 3) hybrid capture-based-PCR. Anchored multiplex PCRbased sequencing was investigated for routine molecular testing within the national Network Genomic Medicine Lung Cancer (nNGM). Four centers applied the anchored multiplex ArcherDX-Variantplex nNGMv2 panel to reanalyze samples pre-tested during routine diagnostics. Data analyses were performed by each center and compiled centrally according to study design. Pre-defined standards were utilized, and panel sensitivity was determined by dilution experiments. nNGMv2 panel sequencing was successful in 98.9% of the samples (N = 90). With default filter settings, all but two potential MET exon 14 skipping variants were identified at similar allele frequencies. Both MET variants were found with an adapted calling filter. Three additional variants (KEAP1, STK11, TP53) were called that were not identified in pre-testing analyses. Only total DNA amount but not a qPCR-based DNA quality score correlated with average coverage. Analysis was successful with a DNA input as low as 6.25 ng. Anchored multiplex PCR-based sequencing (nNGMv2) and a sophisticated user-friendly Archer-Analysis pipeline is a robust and specific technology to detect tumor genetic mutations for precision medicine of lung cancer patients. C1 [Kumbrink, Jorg] Ludwig Maximilian University of Munich (LMU), Faculty of Medicine, Institute of PathologyMunich, Germany. [Demes, Melanie-Christin] University Hospital Frankfurt, Dr. Senckenberg Institute of PathologyFrankfurt, Germany. [Jeroch, Jan] University Hospital Frankfurt, Dr. Senckenberg Institute of PathologyFrankfurt, Germany. [Brauninger, Andreas] Justus-Liebig-University, Institute for PathologyGiessen, Germany. [Hartung, Kristin] Justus-Liebig-University, Institute for PathologyGiessen, Germany. [Gerstenmaier, Uwe] Ulm University, Department of PathologyUlm, Germany. [Marienfeld, Ralf] Ulm University, Department of PathologyUlm, Germany. [Hillmer, Axel] University of Cologne Medical School, Institute of PathologyCologne, Germany. [Bohn, Nadine] ArcherBoulder, CO, USA. [Lehning, Christina] ArcherBoulder, CO, USA. [Ferch, Ferdinand] ArcherBoulder, CO, USA. [Wild, Peter] University Hospital Frankfurt, Dr. Senckenberg Institute of PathologyFrankfurt, Germany. [Gattenlohner, Stefan] Justus-Liebig-University, Institute for PathologyGiessen, Germany. [Moller, Peter] Ulm University, Department of PathologyUlm, Germany. [Klauschen, Frederick] Ludwig Maximilian University of Munich (LMU), Faculty of Medicine, Institute of PathologyMunich, Germany. [Jung, Andreas] Ludwig Maximilian University of Munich (LMU), Faculty of Medicine, Institute of PathologyMunich, Germany. RP Kumbrink, J (reprint author), Ludwig Maximilian University of Munich (LMU), Faculty of Medicine, Institute of Pathology, Munich, Germany. EM joerg.kumbrink@med.uni-muenchen.de CR Falcone R, Lombardi P, Filetti M, Duranti S, Pietragalla A, Fabi A, et al. Oncologic drugs approval in europe for solid tumors: overview of the last 6 years. Cancers, Basel,, 2022, 14:889., DOI 10.3390/cancers14040889 Oda Y, Narukawa M. Response rate of anticancer drugs approved by the Food and Drug Administration based on a single-arm trial. BMC Cancer, 2022, 22:277., DOI 10.1186/s12885-022-09383-w Turner JH. An introduction to the clinical practice of theranostics in oncology. Br J Radiol, 2018, 91:20180440., DOI 10.1259/bjr.20180440 Herrera-Juarez M, Serrano-Gomez C, Bote-de-Cabo H, Paz-Ares L. Targeted therapy for lung cancer: beyond EGFR and ALK. Cancer, 2023, 129:1803–20., DOI 10.1002/cncr.34757 Khadela A, Postwala H, Rana D, Dave H, Ranch K, Boddu SHS. A review of recent advances in the novel therapeutic targets and immunotherapy for lung cancer. Immunotherapy Lung Cancer Med Oncol, 2023, 40:152., DOI 10.1007/ s12032-023-02005-w Morash M, Mitchell H, Beltran H, Elemento O, Pathak J. The role of nextgeneration sequencing in precision medicine: a review of outcomes in oncology. J Pers Med, 2018, 8:30., DOI 10.3390/jpm8030030 Buttner R, Wolf J, Kron A, Nationales Netzwerk Genomische M. The national Network Genomic Medicine, nNGM): model for innovative diagnostics and therapy of lung cancer within a public healthcare system. Pathologe, 2019, 40: 276–80., DOI 10.1007/s00292-019-0605-4 Singh RR. Target enrichment approaches for next-generation sequencing applications in oncology. Diagnostics, Basel,, 2022, 12:1539., DOI 10.3390/ diagnostics12071539 Zheng Z, Liebers M, Zhelyazkova B, Cao Y, Panditi D, Lynch KD, et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat Med, 2014, 20:1479–84., DOI 10.1038/nm.3729 Robinson JT, Thorvaldsdottir H, Wenger AM, Zehir A, Mesirov JP. Variant review with the integrative genomics viewer. Cancer Res, 2017, 77:e31–4., DOI 10. 1158/0008-5472.CAN-17-0337 Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, et al. COSMIC: the catalogue of somatic mutations. Cancer Nucleic Acids Res, 2019, 47: D941–D947., DOI 10.1093/nar/gky1015 Landrum MJ, Lee JM, Benson M, Brown GR, Chao C, Chitipiralla S, et al. ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res, 2018, 46:D1062–D1067., DOI 10.1093/nar/gkx1153 Chakravarty D, Gao J, Phillips SM, Kundra R, Zhang H, Wang J, et al. OncoKB: a precision oncology knowledge base. JCO Precis Oncol, 2017, 2017:1–16., DOI 10.1200/PO.17.00011 Kopanos C, Tsiolkas V, Kouris A, Chapple CE, Albarca Aguilera M, Meyer R, et al. VarSome: the human genomic variant search engine. Bioinformatics, 2019, 35: 1978–80., DOI 10.1093/bioinformatics/bty897 den Dunnen JT, Dalgleish R, Maglott DR, Hart RK, Greenblatt MS, McGowan-Jordan J, et al. HGVS recommendations for the description of sequence variants: 2016 update. Hum Mutat, 2016, 37:564–9., DOI 10.1002/ humu.22981 Awad MM, Oxnard GR, Jackman DM, Savukoski DO, Hall D, Shivdasani P, et al. MET exon 14 mutations in non-small-cell lung cancer are associated with advanced age and stage-dependent MET genomic amplification and c-met overexpression. J Clin Oncol, 2016, 34:721–30., DOI 10.1200/JCO.2015.63.4600 West HJ,McClelandM, Cappuzzo F, Reck M,Mok TS, Jotte RM, et al. Clinical efficacy of atezolizumab plus bevacizumab and chemotherapy in KRAS-mutated non-small cell lung cancer with STK11, KEAP1, or TP53 comutations: subgroup results from the phase III IMpower150 trial. J Immunother Cancer, 2022, 10: e003027., DOI 10.1136/jitc-2021-003027 Marinelli D, Mazzotta M, Scalera S, Terrenato I, Sperati F, D’Ambrosio L, et al. KEAP1-driven co-mutations in lung adenocarcinoma unresponsive to immunotherapy despite high tumor mutational burden. Ann Oncol, 2020, 31: 1746–54., DOI 10.1016/j.annonc.2020.08.2105 Ricciuti B, Arbour KC, Lin JJ, Vajdi A, Vokes N, Hong L, et al. Diminished efficacy of programmed death-(ligand)1 inhibition in STK11- and KEAP1-mutant lung adenocarcinoma is affected by KRAS mutation status. J Thorac Oncol, 2022, 17:399–410., DOI 10.1016/j.jtho.2021.10.013 Dang J, Mendez P, Lee S, Kim JW, Yoon JH, Kim TW, et al. Development of a robust DNA quality and quantity assessment qPCR assay for targeted nextgeneration sequencing library preparation. Int J Oncol, 2016, 49:1755–65., DOI 10.3892/ijo.2016.3654 Jennings LJ, Arcila ME, Corless C, Kamel-Reid S, Lubin IM, Pfeifer J, et al. Guidelines for validation of next-generation sequencing-based oncology panels: a joint consensus recommendation of the association for molecular pathology and college of American pathologists. J Mol Diagn, 2017, 19:341–65., DOI 10.1016/j. jmoldx.2017.01.011 Desmeules P, Boudreau DK, Bastien N, Boulanger MC, Bosse Y, Joubert P, et al. Performance of an RNA-based next-generation sequencing assay for combined detection of clinically actionable fusions and hotspot mutations in NSCLC. JTO Clin Res Rep, 2022, 3:100276., DOI 10.1016/j.jtocrr.2022.100276 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611590 EP 1611604 DI 10.3389/pore.2024.1611590 PG 15 ER PT J AU Wu, J Wang, Y Cheng, Y Cheng, L Zhang, L AF Wu, Jiaxin Wang, Yuanying Cheng, Yi Cheng, Li Zhang, LuShun TI Comprehensive landscape and future perspectives of non-coding RNAs in esophageal squamous cell carcinoma, a bibliometric analysis from 2008 to 2023 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE novel biomarker; non-coding RNAs; digestive system cancer; bibliometric analysis; VOSviewer ID novel biomarker; non-coding RNAs; digestive system cancer; bibliometric analysis; VOSviewer AB Objectives: Summarize the progress and hot topic evolution of non-coding RNAs (ncRNAs) research in esophageal squamous cell carcinoma (ESCC) in recent years and predict future research directions. Methods: Relevant articles from the Web of Science until 31 October 2023 were obtained. Bibliometric analysis of included articles was performed using software (VOSviewer, CiteSpace, and Bibliometrix). The volume and citation of publications, as well as the country, institution, author, journal, keywords of the articles were used as variables to analyze the research trends and hot spot evolution. Results: 1,118 literature from 2008 to 2023 were retrieved from database, with 25 countries/regions, 793 institutions, 5,426 authors, 261 journals involved. Global cooperation was centered on China, Japan, and the United States. Zhengzhou University, an institution from China, had the highest publication. The most prolific author was Guo Wei, and the most prolific journal was Oncology Letters. Analysis of keywords revealed that the research in this field revolved around the role of ncRNAs in the occurrence, development, diagnosis, treatment, and prognosis of ESCC, mainly including micro RNAs, long non-coding RNAs, and then circular RNAs. Conclusion: Overall, research on ncRNAs in ESCC remains strong. Previous research has mainly focused on the basic research, with a focus on the mechanism of ncRNAs in the occurrence, development, diagnosis, treatment, and prognosis of ESCC. Combining current research with emerging disciplines to further explore its mechanisms of action or shifting the focus of research from preclinical research to clinical research based on diagnosis, treatment, and prognosis, will be the main breakthrough in this field in the future. C1 [Wu, Jiaxin] Chengdu Medical College, Graduate SchoolChengdu, China. [Wang, Yuanying] Chengdu Medical College, Graduate SchoolChengdu, China. [Cheng, Yi] People’s Hospital of Lushan County, Department of RadiologyYa’an, China. [Cheng, Li] Cheng Du Medical College, Department of Pathology and PathophysiologyChengdu, China. [Zhang, LuShun] Cheng Du Medical College, Department of Pathology and PathophysiologyChengdu, China. RP Zhang, L (reprint author), Cheng Du Medical College, Department of Pathology and Pathophysiology, Chengdu, China. EM zhangls2012@cmc.edu.cn CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer J clinicians, 2021, 71(3): 209–49., DOI 10.3322/caac.21660 Huang J, Koulaouzidis A, Marlicz W, Lok V, Chu C, Ngai CH, et al. Global burden, risk factors, and trends of esophageal cancer: an analysis of cancer registries from 48 countries. Cancers, 2021, 13(1):141., DOI 10.3390/cancers13010141 Moore JE, Purcaro MJ, Pratt HE, Epstein CB, Shoresh N, Adrian J, et al. Expanded encyclopaedias of DNA elements in the human and mouse genomes. Nature, 2020, 583(7818):699–710., DOI 10.1038/s41586-020-2493-4 Wang C, Zhou M, Zhu P, Ju C, Sheng J, Du D, et al. IGF2BP2-induced circRUNX1 facilitates the growth and metastasis of esophageal squamous cell carcinoma through miR-449b-5p/FOXP3 axis. J Exp Clin Cancer Res : CR, 2022, 41(1):347., DOI 10.1186/s13046-022-02550-8 Wu Q, Zhang H, Yang D, Min Q, Wang Y, Zhang W, et al. The m6A-induced lncRNA CASC8 promotes proliferation and chemoresistance via upregulation of hnRNPL in esophageal squamous cell carcinoma. Int J Biol Sci, 2022, 18(13): 4824–36., DOI 10.7150/ijbs.71234 Song B, Liu X, Dong H, Roy R.miR-140-3P induces chemotherapy resistance in esophageal carcinoma by targeting the NFYA-MDR1 Axis. Appl Biochem Biotechnol, 2023, 195(2):973–91., DOI 10.1007/s12010-022-04139-5 Ninkov A, Frank JR, Maggio LA. Bibliometrics: methods for studying academic publishing. Perspect Med Educ, 2022, 11(3):173–6., DOI 10.1007/s40037-021-00695-4 Okubo Y. “Bibliometric indicators and analysis of research systems: methods and examples,” in OECD science, technology and industry working papers, No. 1997/ 01. Paris: OECD Publishing, 1997)., DOI 10.1787/208277770603 Ge Y, Chao T, Sun J, LiuW, Chen Y,Wang C. Frontiers and hotspots evolution in psycho-cardiology: a bibliometric analysis from 2004 to 2022. Curr Probl Cardiol, 2022, 47(12):101361., DOI 10.1016/j.cpcardiol.2022.101361 Ahmad P, Slots J. A bibliometric analysis of periodontology. Periodontol 2000, 2021, 85(1):237–40., DOI 10.1111/prd.12376 Hassan W, Zafar M, Duarte AE, Kamdem JP, Teixeira da Rocha JB. Pharmacological Research: a bibliometric analysis from 1989 to 2019. Pharmacol Res, 2021, 169:105645., DOI 10.1016/j.phrs.2021.105645 Aria M, Cuccurullo C. bibliometrix: an R-tool for comprehensive science mapping analysis. J Informetrics, 2017, 11(4):959–75., DOI 10.1016/j.joi.2017.08.007 Guo Y, Chen Z, Zhang L, Zhou F, Shi S, Feng X, et al. Distinctive microRNA profiles relating to patient survival in esophageal squamous cell carcinoma. Cancer Res, 2008, 68(1):26–33., DOI 10.1158/0008-5472.CAN-06-4418 Li CQ, Huang GW, Wu ZY, Xu YJ, Li XC, Xue YJ, et al. Integrative analyses of transcriptome sequencing identify novel functional lncRNAs in esophageal squamous cell carcinoma. Oncogenesis, 2017, 6(2):e297., DOI 10. 1038/oncsis.2017.1 Fan L, Cao Q, Liu J, Zhang J, Li B. Circular RNA profiling and its potential for esophageal squamous cell cancer diagnosis and prognosis. Mol Cancer, 2019, 18(1): doi:10.1186/s12943-018-0936-4 16. Su H, Lin F, Deng X, Shen L, Fang Y, Fei Z, et al. Profiling and bioinformatics analyses reveal differential circular RNA expression in radioresistant esophageal cancer cells. J translational Med, 2016, 14(1):225., DOI 10.1186/s12967-016-0977-7 Liu H, Zhang Q, Lou Q, Zhang X, Cui Y,Wang P, et al. Differential analysis of lncRNA, miRNA and mRNA expression profiles and the prognostic value of lncRNA in esophageal cancer. Pathol Oncol Res : POR, 2020, 26(2):1029–39., DOI 10.1007/s12253-019-00655-8 Wagner CS, Jonkers K. Open countries have strong science. Nature, 2017, 550(7674):32–3., DOI 10.1038/550032a Bol T, de Vaan M, van de Rijt A. The Matthew effect in science funding. Proc Natl Acad Sci USA, 2018, 115(19):4887–90., DOI 10.1073/pnas.1719557115 Zhou H, Hao X, Zhang P, He S. Noncoding RNA mutations in cancer. Wiley Interdiscip Rev RNA, 2023, 14(6):e1812., DOI 10.1002/wrna.1812 Yan H, Bu P. Non-coding RNA in cancer. Essays Biochem, 2021, 65(4): 625–39., DOI 10.1042/EBC20200032 Takeshita N, Hoshino I, Mori M, Akutsu Y, Hanari N, Yoneyama Y, et al. Serum microRNA expression profile: miR-1246 as a novel diagnostic and prognostic biomarker for oesophageal squamous cell carcinoma. Br J Cancer, 2013, 108(3):644–52., DOI 10.1038/bjc.2013.8 Liu S, Lin Z, Zheng Z, Rao W, Lin Y, Chen H, et al. Serum exosomal microRNA-766-3p expression is associated with poor prognosis of esophageal squamous cell carcinoma. Cancer Sci, 2020, 111(10):3881–92., DOI 10.1111/cas. 14550 Huang Z, Zhang L, Zhu D, Shan X, Zhou X, Qi LW, et al. A novel serum microRNA signature to screen esophageal squamous cell carcinoma. Cancer Med, 2017, 6(1):109–19., DOI 10.1002/cam4.973 Sun H, Wang L, Zhao Q, Dai J. Diagnostic and prognostic value of serum miRNA-1290 in human esophageal squamous cell carcinoma. Cancer biomarkers : section A Dis markers, 2019, 25(4):381–7., DOI 10.3233/CBM-190007 Chen FJ, Sun M, Li SQ, Wu QQ, Ji L, Liu ZL, et al. Upregulation of the long non-coding RNA HOTAIR promotes esophageal squamous cell carcinoma metastasis and poor prognosis. Mol carcinogenesis, 2013, 52(11):908–15., DOI 10. 1002/mc.21944 Cao X, Zhao R, Chen Q, Zhao Y, Zhang B, Zhang Y, et al. MALAT1 might be a predictive marker of poor prognosis in patients who underwent radical resection of middle thoracic esophageal squamous cell carcinoma. Cancer biomarkers : section A Dis markers, 2015, 15(6):717–23., DOI 10.3233/CBM-150513 Zhang X, Xu Y, He C, Guo X, Zhang J, He C, et al. Elevated expression of CCAT2 is associated with poor prognosis in esophageal squamous cell carcinoma. J Surg Oncol, 2015, 111(7):834–9., DOI 10.1002/jso.23888 Shi WH, Wu QQ, Li SQ, Yang TX, Liu ZH, Tong YS, et al. Upregulation of the long noncoding RNA PCAT-1 correlates with advanced clinical stage and poor prognosis in esophageal squamous carcinoma. Tumour Biol : J Int Soc Oncodevelopmental Biol Med, 2015, 36(4):2501–7., DOI 10.1007/s13277-014- 2863-3 Huang E, Fu J, Yu Q, Xie P, Yang Z, Ji H, et al. CircRNA hsa_circ_ 0004771 promotes esophageal squamous cell cancer progression via miR-339- 5p/CDC25A axis. Epigenomics, 2020, 12(7):587–603., DOI 10.2217/epi-2019-0404 Zhang Y, Wang Q, Zhu D, Rong J, Shi W, Cao X. Up-regulation of circ- SMAD7 inhibits tumor proliferation and migration in esophageal squamous cell carcinoma. Biomed Pharmacother, 2019, 111:596–601., DOI 10.1016/j.biopha.2018. 12.116 Hu X,Wu D, He X, Zhao H, He Z, Lin J, et al. circGSK3β promotes metastasis in esophageal squamous cell carcinoma by augmenting β-catenin signaling. Mol Cancer, 2019, 18(1):160., DOI 10.1186/s12943-019-1095-y Miyoshi J, Zhu Z, Luo A, Toden S, Zhou X, Izumi D, et al. A microRNA-based liquid biopsy signature for the early detection of esophageal squamous cell carcinoma: a retrospective, prospective and multicenter study. Mol Cancer, 2022, 21(1):44., DOI 10.1186/s12943-022-01507-x Qiao G, Dai C, He Y, Shi J, Xu C. Effects of miR-106b-3p on cell proliferation and epithelial-mesenchymal transition, and targeting of ZNRF3 in esophageal squamous cell carcinoma. Int J Mol Med, 2019, 43(4):1817–29., DOI 10.3892/ ijmm.2019.4107 Guo Y, Sun P, Guo W, Yin Q, Han J, Sheng S, et al. LncRNA DDX11 antisense RNA 1 promotes EMT process of esophageal squamous cell carcinoma by sponging miR-30d-5p to regulate SNAI1/ZEB2 expression and Wnt/β-catenin pathway. Bioengineered, 2021, 12(2):11425–40., DOI 10.1080/ 21655979.2021.2008759 Gu L, Sang Y, Nan X, Zheng Y, Liu F, Meng L, et al. circCYP24A1 facilitates esophageal squamous cell carcinoma progression through binding PKM2 to regulate NF-κB-induced CCL5 secretion. Mol Cancer, 2022, 21(1):217., DOI 10. 1186/s12943-022-01686-7 Shi X, Liu X, Huang S, Hao Y, Pan S, Ke Y, et al. miR-4443 promotes radiation resistance of esophageal squamous cell carcinoma via targeting PTPRJ. J translational Med, 2022, 20(1):626., DOI 10.1186/s12967-022-03818-5 Lu HB. MicroRNA-556-3p promotes the progression of esophageal cancer via targeting DAB2IP. Eur Rev Med Pharmacol Sci, 2018, 22(20):6816–23., DOI 10. 26355/eurrev_201810_16149 Tan X, Ren S, Fu MZ, Ren S, Yang C,Wu X, et al. microRNA-196b promotes esophageal squamous cell carcinogenesis and chemoradioresistance by inhibiting EPHA7, thereby restoring EPHA2 activity. Am J Cancer Res, 2021, 11(7):3594–610. Xu C, Li B, Zhao S, Jin B, Jia R, Ge J, et al. MicroRNA-186-5p inhibits proliferation and metastasis of esophageal cancer by mediating HOXA9. OncoTargets Ther, 2019, 12:8905–14., DOI 10.2147/OTT.S227920 Wang N, Yu Y, Xu B, Zhang M, Li Q, Miao L. Pivotal prognostic and diagnostic role of the long non-coding RNA colon cancer-associated transcript 1 expression in human cancer, Review). Mol Med Rep, 2019, 19(2):771–82., DOI 10. 3892/mmr.2018.9721 Jia J, Li H, Chu J, Sheng J, Wang C, Jia Z, et al. LncRNA FAM83A-AS1 promotes ESCC progression by regulating miR-214/CDC25B axis. J Cancer, 2021, 12(4):1200–11., DOI 10.7150/jca.54007 Sang M, Meng L, Sang Y, Liu S, Ding P, Ju Y, et al. Circular RNA ciRS-7 accelerates ESCC progression through acting as a miR-876-5p sponge to enhance MAGE-A family expression. Cancer Lett, 2018, 426:37–46., DOI 10.1016/j.canlet. 2018.03.049 Shi Y, Fang N, Li Y, Guo Z, Jiang W, He Y, et al. Circular RNA LPAR3 sponges microRNA-198 to facilitate esophageal cancer migration, invasion, and metastasis. Cancer Sci, 2020, 111(8):2824–36., DOI 10.1111/cas.14511 Toshimitsu H, Hashimoto K, Tangoku A, Iizuka N, Yamamoto K, Kawauchi S, et al. Molecular signature linked to acquired resistance to cisplatin in esophageal cancer cells. Cancer Lett, 2004, 211(1):69–78., DOI 10.1016/j.canlet.2004.01.038 Jin YY, Chen QJ, Xu K, Ren HT, Bao X, Ma YN, et al. Involvement of microRNA-141-3p in 5-fluorouracil and oxaliplatin chemo-resistance in esophageal cancer cells via regulation of PTEN. Mol Cell Biochem, 2016, 422(1- 2):161–70., DOI 10.1007/s11010-016-2816-9 Zhu H, Du F, Cao C. Restoration of circPSMC3 sensitizes gefitinib-resistant esophageal squamous cell carcinoma cells to gefitinib by regulating miR-10a-5p/ PTEN axis. Cel Biol Int, 2021, 45(1):107–16., DOI 10.1002/cbin.11473 Jia Y, Tian C, Wang H, Yu F, Lv W, Duan Y, et al. Long non-coding RNA NORAD/miR-224-3p/MTDH axis contributes to CDDP resistance of esophageal squamous cell carcinoma by promoting nuclear accumulation of β-catenin. Mol Cancer, 2021, 20(1):162., DOI 10.1186/s12943-021-01455-y Yan J, Shi L, Lin S, Li Y. MicroRNA-624-mediated ARRDC3/YAP/HIF1α axis enhances esophageal squamous cell carcinoma cell resistance to cisplatin and paclitaxel. Bioengineered, 2021, 12(1):5334–47., DOI 10.1080/21655979.2021. 1938497 Cheng J, Zhang R, Yan M, Li Y. Circular RNA hsa_circ_0000277 promotes tumor progression and DDP resistance in esophageal squamous cell carcinoma. BMC cancer, 2022, 22(1):238., DOI 10.1186/s12885-022-09241-9 Liu Z, Gu S, Wu K, Li L, Dong C, Wang W, et al. CircRNA-DOPEY2 enhances the chemosensitivity of esophageal cancer cells by inhibiting CPEB4-mediated Mcl- 1 translation. J Exp Clin Cancer Res : CR, 2021, 40(1):361., DOI 10.1186/s13046-021- 02149-5 Sharma U, Murmu M, Barwal TS, Tuli HS, Jain M, Prakash H, et al. A pleiotropic role of long non-coding RNAs in the modulation of wnt/β-catenin and PI3K/Akt/mTOR signaling pathways in esophageal squamous cell carcinoma: implication in chemotherapeutic drug response. Curr Oncol, Toronto, Ont,, 2022, 29(4):2326–49., DOI 10.3390/curroncol29040189 Lin J, Liu Z, Liao S, Li E, Wu X, Zeng W. Elevation of long non-coding RNA GAS5 and knockdown of microRNA-21 up-regulate RECK expression to enhance esophageal squamous cell carcinoma cell radio-sensitivity after radiotherapy. Genomics, 2020, 112(3):2173–85., DOI 10.1016/j.ygeno.2019.12.013 He Y, Mingyan E, Wang C, Liu G, Shi M, Liu S. CircVRK1 regulates tumor progression and radioresistance in esophageal squamous cell carcinoma by regulating miR-624-3p/PTEN/PI3K/AKT signaling pathway. Int J Biol macromolecules, 2019, 125:116–23., DOI 10.1016/j.ijbiomac.2018.11.273 Yao W, Jia X, Zhu L, Xu L, Zhang Q, Xia T, et al. Exosomal circ_ 0026611 contributes to lymphangiogenesis by reducing PROX1 acetylation and ubiquitination in human lymphatic endothelial cells, HLECs). Cell Mol Biol Lett, 2023, 28(1):13., DOI 10.1186/s11658-022-00410-z Zhao L, Yu L, Wang X, He J, Zhu X, Zhang R, et al. Mechanisms of function and clinical potential of exosomes in esophageal squamous cell carcinoma. Cancer Lett, 2023, 553:215993., DOI 10.1016/j.canlet.2022.215993 Zhao F, Li Z, Dong Z,Wang Z, Guo P, Zhang D, et al. Exploring the potential of exosome-related LncRNA pairs as predictors for immune microenvironment, survival outcome, and microbiotain landscape in esophageal squamous cell carcinoma. Front Immunol, 2022, 13:918154., DOI 10.3389/fimmu.2022.918154 Zheng Y, Chen Z, Han Y, Han L, Zou X, Zhou B, et al. Immune suppressive landscape in the human esophageal squamous cell carcinoma microenvironment. Nat Commun, 2020, 11(1):6268., DOI 10.1038/s41467- 020-20019-0 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611595 EP 1611606 DI 10.3389/pore.2024.1611595 PG 12 ER PT J AU Fekete, M Major, D Feher, A Fazekas-Pongor, V Lehoczki, A AF Fekete, Monika Major, David Feher, Agnes Fazekas-Pongor, Vince Lehoczki, Andrea TI Geroscience and pathology: a new frontier in understanding age-related diseases SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE geroscience; senescence; cancer; cardiovascular disease; ageing ID geroscience; senescence; cancer; cardiovascular disease; ageing AB Geroscience, a burgeoning discipline at the intersection of aging and disease, aims to unravel the intricate relationship between the aging process and pathogenesis of age-related diseases. This paper explores the pivotal role played by geroscience in reshaping our understanding of pathology, with a particular focus on age-related diseases. These diseases, spanning cardiovascular and cerebrovascular disorders, malignancies, and neurodegenerative conditions, significantly contribute to the morbidity and mortality of older individuals. We delve into the fundamental cellular and molecular mechanisms underpinning aging, including mitochondrial dysfunction and cellular senescence, and elucidate their profound implications for the pathogenesis of various age-related diseases. Emphasis is placed on the importance of assessing key biomarkers of aging and biological age within the realm of pathology. We also scrutinize the interplay between cellular senescence and cancer biology as a central area of focus, underscoring its paramount significance in contemporary pathological research. Moreover, we shed light on the integration of anti-aging interventions that target fundamental aging processes, such as senolytics, mitochondria-targeted treatments, and interventions that influence epigenetic regulation within the domain of pathology research. In conclusion, the integration of geroscience concepts into pathological research heralds a transformative paradigm shift in our understanding of disease pathogenesis and promises breakthroughs in disease prevention and treatment. C1 [Fekete, Monika] Semmelweis University, Department of Public HealthBudapest, Hungary. [Major, David] Semmelweis University, Department of Public HealthBudapest, Hungary. [Feher, Agnes] Semmelweis University, Department of Public HealthBudapest, Hungary. [Fazekas-Pongor, Vince] Semmelweis University, Department of Public HealthBudapest, Hungary. [Lehoczki, Andrea] Semmelweis University, Department of Public HealthBudapest, Hungary. RP Fazekas-Pongor, V (reprint author), Semmelweis University, Department of Public Health, Budapest, Hungary. EM pongor.vince@semmelweis.hu CR Hernandez CM, Hernandez AR, Hoffman JM, King PH, McMahon LL, Buford TW, et al. A neuroscience primer for integrating geroscience with the neurobiology of aging. J Gerontol A Biol Sci Med Sci, 2022, 77:e19–e33., DOI 10.1093/gerona/ glab301 Llarena N, Hine C. Reproductive longevity and aging: geroscience approaches to maintain long-term ovarian fitness. J Gerontol A Biol Sci Med Sci, 2021, 76: 1551–60., DOI 10.1093/gerona/glaa204 Byers JM, 3rd. Rudolf Virchow--father of cellular pathology. Am J Clin Pathol, 1989, 92:S2–8. Duque G, Lipsitz LA, Ferrucci L, Addie S, Carrington-Lawrence S, Kohanski R. Geroscience for the next chapter of medicine. J Gerontol A Biol Sci Med Sci, 2023, 78:791–2., DOI 10.1093/gerona/glad083 Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell, 2013, 153:1194–217., DOI 10.1016/j.cell.2013.05.039 Urban VS, Cegledi A, Mikala G. Multiple myeloma, a quintessential malignant disease of aging: a geroscience perspective on pathogenesis and treatment. Geroscience, 2023, 45:727–46., DOI 10.1007/s11357-022-00698-x Lamanuzzi A, Saltarella I, Frassanito MA, Ribatti D, Melaccio A, Desantis V, et al. Thrombopoietin promotes angiogenesis and disease progression in patients with multiple myeloma. Am J Pathol, 2021, 191:748–58., DOI 10.1016/j.ajpath.2020. 12.016 Dorigatti AO, Riordan R, Yu Z, Ross G, Wang R, Reynolds-Lallement N, et al. Brain cellular senescence in mouse models of Alzheimer’s disease. Geroscience, 2022, 44:1157–68., DOI 10.1007/s11357-022-00531-5 Pomilio C, Gorojod RM, Riudavets M, Vinuesa A, Presa J, Gregosa A, et al. Microglial autophagy is impaired by prolonged exposure to β-amyloid peptides: evidence from experimental models and Alzheimer’s disease patients. Geroscience, 2020, 42:613–32., DOI 10.1007/s11357-020-00161-9 DiBattista AM, Sierra F, Masliah E. NIA workshop on senescence in brain aging and Alzheimer’s disease and its related dementias. Geroscience, 2020, 42: 389–96., DOI 10.1007/s11357-020-00153-9 Liu H, Lutz M, Luo S, Alzheimer’s Disease Neuroimaging Initiative. Genetic association between epigenetic aging-acceleration and the progression of mild cognitive impairment to Alzheimer’s disease. J Gerontol A Biol Sci Med Sci, 2022, 77:1734–42., DOI 10.1093/gerona/glac138 Wang H, Fu J, Xu X, Yang Z, Zhang T. Rapamycin activates mitophagy and alleviates cognitive and synaptic plasticity deficits in a mouse model of Alzheimer’s disease. J Gerontol A Biol Sci Med Sci, 2021, 76:1707–13., DOI 10.1093/gerona/ glab142 Toth L, Czigler A, Hegedus E, Komaromy H, Amrein K, Czeiter E, et al. Agerelated decline in circulating IGF-1 associates with impaired neurovascular coupling responses in older adults. Geroscience, 2022, 44:2771–83., DOI 10.1007/ s11357-022-00623-2 Sakamuri S, Sure VN, Kolli L, Liu N, Evans WR, Sperling JA, et al. Glycolytic and oxidative phosphorylation defects precede the development of senescence in primary human brain microvascular endothelial cells. Geroscience, 2022, 44: 1975–94., DOI 10.1007/s11357-022-00550-2 Towner RA, Gulej R, Zalles M, Saunders D, Smith N, Lerner M, et al. Rapamycin restores brain vasculature, metabolism, and blood-brain barrier in an inflammaging model. Geroscience, 2021, 43:563–78., DOI 10.1007/s11357-021- 00363-9 Islam MT, Tuday E, Allen S, Kim J, Trott DW, Holland WL, et al. Senolytic drugs, dasatinib and quercetin, attenuate adipose tissue inflammation, and ameliorate metabolic function in old age. Aging Cell, 2023, 22:e13767., DOI 10. 1111/acel.13767 Bloom SI, Tucker JR, Machin DR, Abdeahad H, Adeyemo AO, Thomas TG, et al. Reduction of double-strand DNA break repair exacerbates vascular aging. Aging, Albany NY,, 2023, 15:9913–47., DOI 10.18632/aging.205066 Bloom SI, Tucker JR, Lim J, Thomas TG, Stoddard GJ, Lesniewski LA, et al. Aging results in DNA damage and telomere dysfunction that is greater in endothelial versus vascular smooth muscle cells and is exacerbated in atheroprone regions. Geroscience, 2022, 44:2741–55., DOI 10.1007/s11357-022- 00681-6 Ungvari Z, Tarantini S, Donato AJ, Galvan V, Csiszar A. Mechanisms of vascular aging. Circ Res, 2018, 123:849–67., DOI 10.1161/CIRCRESAHA.118.311378 Liu Y, Bloom SI, Donato AJ. The role of senescence, telomere dysfunction and shelterin in vascular aging. Microcirculation, 2018, 26:e12487., DOI 10.1111/micc. 12487 Rossman MJ, Kaplon RE, Hill SD, McNamara MN, Santos-Parker JR, Pierce GL, et al. Endothelial cell senescence with aging in healthy humans: prevention by habitual exercise and relation to vascular endothelial function. Am J Physiol Heart Circ Physiol, 2017, 313:H890–H895., DOI 10.1152/ajpheart.00416.2017 Kennedy BK, Berger SL, Brunet A, Campisi J, Cuervo AM, Epel ES, et al. Geroscience: linking aging to chronic disease. Cell, 2014, 159:709–13., DOI 10.1016/j. cell.2014.10.039 Rolland Y, Sierra F, Ferrucci L, Barzilai N, De Cabo R, Mannick J, et al. Challenges in developing Geroscience trials. Nat Commun, 2023, 14:5038., DOI 10. 1038/s41467-023-39786-7 Kaushik S, Cuervo AM. Proteostasis and aging. Nat Med, 2015, 21:1406–15., DOI 10.1038/nm.4001 Korovila I, HugoM, Castro JP,Weber D, Hohn A, Grune T, et al. Proteostasis, oxidative stress and aging. Redox Biol, 2017, 13:550–67., DOI 10.1016/j.redox.2017. 07.008 Santra M, Dill KA, de Graff AMR. Proteostasis collapse is a driver of cell aging and death. Proc Natl Acad Sci U S A, 2019, 116:22173–8., DOI 10.1073/pnas. 1906592116 Taylor RC, Hetz C. Mastering organismal aging through the endoplasmic reticulum proteostasis network. Aging Cell, 2020, 19:e13265., DOI 10.1111/acel.13265 Ferreira JV, da Rosa Soares A, Pereira P. Cell non-autonomous proteostasis regulation in aging and disease. Front Neurosci, 2022, 16:878296., DOI 10.3389/fnins. 2022.878296 Pearson KJ, Lewis KN, Price NL, Chang JW, Perez E, Cascajo MV, et al. Nrf2 mediates cancer protection but not prolongevity induced by caloric restriction. Proc Natl Acad Sci U S A, 2008, 105:2325–30., DOI 10.1073/pnas.0712162105 Ungvari Z, Bailey-Downs L, Gautam T, Sosnowska D, Wang M, Monticone RE, et al. Age-associated vascular oxidative stress, Nrf2 dysfunction, and NF- {kappa}B activation in the nonhuman primate Macaca mulatta. J Gerontol A Biol Sci Med Sci, 2011, 66:866–75., DOI 10.1093/gerona/glr092 Ungvari Z, Bailey-Downs L, Sosnowska D, Gautam T, Koncz P, Losonczy G, et al. Vascular oxidative stress in aging: a homeostatic failure due to dysregulation of Nrf2-mediated antioxidant response. Am J Physiol Heart Circ Physiol, 2011, 301: H363–72., DOI 10.1152/ajpheart.01134.2010 Ungvari ZI, Bailey-Downs L, Gautam T, Jimenez R, Losonczy G, Zhang C, et al. Adaptive induction of NF-E2-Related Factor-2-driven antioxidant genes in endothelial cells in response to hyperglycemia. Am J Physiol Heart Circ Physiol, 2011, 300:H1133–40., DOI 10.1152/ajpheart.00402.2010 Ahn B, Pharaoh G, Premkumar P, Huseman K, Ranjit R, Kinter M, et al. Nrf2 deficiency exacerbates age-related contractile dysfunction and loss of skeletal muscle mass. Redox Biol, 2018, 17:47–58., DOI 10.1016/j.redox.2018.04.004 Fulop GA, Kiss T, Tarantini S, Balasubramanian P, Yabluchanskiy A, Farkas E, et al. Nrf2 deficiency in aged mice exacerbates cellular senescence promoting cerebrovascular inflammation. Geroscience, 2018, 40:513–21., DOI 10.1007/s11357- 018-0047-6 Tarantini S, Valcarcel-Ares MN, Yabluchanskiy A, Tucsek Z, Hertelendy P, Kiss T, et al. Nrf2 deficiency exacerbates obesity-induced oxidative stress, neurovascular dysfunction, blood brain barrier disruption, neuroinflammation, amyloidogenic gene expression and cognitive decline in mice, mimicking the aging phenotype. J Gerontol A Biol Sci Med Sci, 2018, 73:853–63., DOI 10.1093/gerona/ glx177 Ungvari Z, Tarantini S, Nyul-Toth A, Kiss T, Yabluchanskiy A, Csipo T, et al. Nrf2 dysfunction and impaired cellular resilience to oxidative stressors in the aged vasculature: from increased cellular senescence to the pathogenesis of age-related vascular diseases. Geroscience, 2019, 41:727–38., DOI 10.1007/s11357-019-00107-w Maynard S, Fang EF, Scheibye-Knudsen M, Croteau DL, Bohr VA. DNA damage, DNA repair, aging, and neurodegeneration. Cold Spring Harb Perspect Med, 2015, 5:a025130., DOI 10.1101/cshperspect.a025130 Yousefzadeh MJ,Wilkinson JE, Hughes B, Gadela N, Ladiges WC, Vo N, et al. Heterochronic parabiosis regulates the extent of cellular senescence in multiple tissues. Geroscience, 2020, 42:951–61., DOI 10.1007/s11357-020-00185-1 Gonzalez-Armenta JL, Li N, Lee RL, Lu B, Molina AJA. Heterochronic parabiosis: old blood induces changes in mitochondrial structure and function of young mice. J Gerontol A Biol Sci Med Sci, 2021, 76:434–9., DOI 10.1093/gerona/ glaa299 Miller HA, Dean ES, Pletcher SD, Leiser SF. Cell non-autonomous regulation of health and longevity. Elife, 2020, 9:e62659., DOI 10.7554/eLife.62659 Lecot P, Alimirah F, Desprez PY, Campisi J, Wiley C. Context-dependent effects of cellular senescence in cancer development. Br J Cancer, 2016, 114:1180–4., DOI 10.1038/bjc.2016.115 Chinta SJ, Woods G, Rane A, Demaria M, Campisi J, Andersen JK. Cellular senescence and the aging brain. Exp Gerontol, 2014, 68:3–7., DOI 10.1016/j.exger. 2014.09.018 Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest, 2013, 123:966–72., DOI 10.1172/JCI64098 Henderson YO, Bithi N, Yang J, Link C, Zhang A, Baron B, et al. A long-term obesogenic high-fat diet in mice partially dampens the anti-frailty benefits of latelife intermittent fasting. Geroscience, 2023, 45:1247–62., DOI 10.1007/s11357-022- 00678-1 Nunan E, Wright CL, Semola OA, Subramanian M, Balasubramanian P, Lovern PC, et al. Obesity as a premature aging phenotype - implications for sarcopenic obesity. Geroscience, 2022, 44:1393–405., DOI 10.1007/s11357-022- 00567-7 Seligman BJ, Berry SD, Lipsitz LA, Travison TG, Kiel DP. Epigenetic age acceleration and change in frailty in MOBILIZE boston. J Gerontol A Biol Sci Med Sci, 2022, 77:1760–5., DOI 10.1093/gerona/glac019 O’Shea DM, Maynard T, Tremont G. DNA methylation "GrimAge" acceleration mediates sex/gender differences in verbal memory and processing speed: findings from the health and retirement study. J Gerontol A Biol Sci Med Sci, 2022, 77:2402–12., DOI 10.1093/gerona/glac133 Lin WY. Lifestyle factors and genetic variants on 2 biological age measures: evidence from 94 443 taiwan biobank participants. J Gerontol A Biol Sci Med Sci, 2022, 77:1189–98., DOI 10.1093/gerona/glab251 Zhou YF, Song XY, Pan XF, Feng L, Luo N, Yuan JM, et al. Association between combined lifestyle factors and healthy ageing in Chinese adults: the Singapore Chinese health study. J Gerontol A Biol Sci Med Sci, 2021, 76: 1796–805., DOI 10.1093/gerona/glab033 Wu Q, Ailshire JA, Kim JK, Crimmins EM. Cardiometabolic risk trajectory among older Americans: findings from the health and retirement study. J Gerontol A Biol Sci Med Sci, 2021, 76:2265–74., DOI 10.1093/gerona/glab205 Pandics T, Major D, Fazekas-Pongor V, Szarvas Z, Peterfi A, Mukli P, et al. Exposome and unhealthy aging: environmental drivers from air pollution to occupational exposures. Geroscience, 2023, 45:3381–408., DOI 10.1007/s11357- 023-00913-3 Chen GC, Hukportie DN, Zhongxiao W, Li FR, Wu XB. The association between exposure to air pollution and dementia incidence: the modifying effect of smoking. J Gerontol A Biol Sci Med Sci, 2022, 78:2309–17., DOI 10.1093/gerona/ glac228 Harman D. Aging: a theory based on free radical and radiation chemistry. J Gerontol, 1956, 11:298–300., DOI 10.1093/geronj/11.3.298 Harman D. The biologic clock: the mitochondria? J Am Geriatr Soc, 1972, 20: 145–7., DOI 10.1111/j.1532-5415.1972.tb00787.x Oudbier SJ, Goh J, Looijaard S, Reijnierse EM, Meskers CGM, Maier AB. Pathophysiological mechanisms explaining the association between low skeletal muscle mass and cognitive function. J Gerontol A Biol Sci Med Sci, 2022, 77: 1959–68., DOI 10.1093/gerona/glac121 Liao FF, Lin G, Chen X, Chen L, Zheng W, Raghow R, et al. Endothelial nitric oxide synthase-deficientmice: amodel of spontaneous cerebral small-vessel disease. Am J Pathol, 2021, 191:1932–45., DOI 10.1016/j.ajpath.2021.02.022 Serikbaeva A, Li Y, Ganesh B, Zelkha R, Kazlauskas A. Hyperglycemia promotes mitophagy and thereby mitigates hyperglycemia-induced damage. Am J Pathol, 2022, 192:1779–94., DOI 10.1016/j.ajpath.2022.08.004 Ionescu-Tucker A, Cotman CW. Emerging roles of oxidative stress in brain aging and Alzheimer’s disease. Neurobiol Aging, 2021, 107:86–95., DOI 10.1016/j. neurobiolaging.2021.07.014 Logan S, Royce GH, Owen D, Farley J, Ranjo-Bishop M, Sonntag WE, et al. Accelerated decline in cognition in a mouse model of increased oxidative stress. Geroscience, 2019, 41:591–607., DOI 10.1007/s11357-019-00105-y Dai DF, Chiao YA, Marcinek DJ, Szeto HH, Rabinovitch PS. Mitochondrial oxidative stress in aging and healthspan. Longev Healthspan, 2014, 3:6., DOI 10.1186/ 2046-2395-3-6 Csiszar A, Ungvari Z, Edwards JG, Kaminski PM, Wolin MS, Koller A, et al. Aging-induced phenotypic changes and oxidative stress impair coronary arteriolar function. Circ Res, 2002, 90:1159–66., DOI 10.1161/01.res.0000020401.61826.ea Sun D, Huang A, Yan EH, Wu Z, Yan C, Kaminski PM, et al. Reduced release of nitric oxide to shear stress in mesenteric arteries of aged rats. Am J Physiol Heart Circ Physiol, 2004, 286:H2249–56., DOI 10.1152/ajpheart.00854.2003 Hamilton CA, Brosnan MJ, McIntyre M, Graham D, Dominiczak AF. Superoxide excess in hypertension and aging: a common cause of endothelial dysfunction. Hypertension, 2001, 37:529–34., DOI 10.1161/01.hyp.37.2.529 van der Loo B, Labugger R, Skepper JN, Bachschmid M, Kilo J, Powell JM, et al. Enhanced peroxynitrite formation is associated with vascular aging. J Exp Med, 2000, 192:1731–44., DOI 10.1084/jem.192.12.1731 Francia P, delli Gatti C, BachschmidM,Martin-Padura I, Savoia C,Migliaccio E, et al. Deletion of p66shc gene protects against age-related endothelial dysfunction. Circulation, 2004, 110:2889–95., DOI 10.1161/01.CIR.0000147731. 24444.4D Csiszar A, Labinskyy N, Orosz Z, Xiangmin Z, Buffenstein R, Ungvari Z. Vascular aging in the longest-living rodent, the naked mole-rat. Am J Physiol, 2007, 293:H919–27., DOI 10.1152/ajpheart.01287.2006 Ungvari ZI, Orosz Z, Labinskyy N, Rivera A, Xiangmin Z, Smith KE, et al. Increased mitochondrial H2O2 production promotes endothelial NF-kappaB activation in aged rat arteries. Am J Physiol Heart Circ Physiol, 2007, 293: H37–47., DOI 10.1152/ajpheart.01346.2006 Jablonski KL, Seals DR, Eskurza I, Monahan KD, Donato AJ. High-dose ascorbic acid infusion abolishes chronic vasoconstriction and restores resting leg blood flow in healthy older men. J Appl Physiol, 2007, 103:1715–21., DOI 10.1152/ japplphysiol.00533.2007 Donato AJ, Eskurza I, Silver AE, Levy AS, Pierce GL, Gates PE, et al. Direct evidence of endothelial oxidative stress with aging in humans: relation to impaired endothelium-dependent dilation and upregulation of nuclear factor-kappaB. Circ Res, 2007, 100:1659–66., DOI 10.1161/01.RES.0000269183.13937.e8 Fleenor BS, Eng JS, Sindler AL, Pham BT, Kloor JD, Seals DR. Superoxide signaling in perivascular adipose tissue promotes age-related artery stiffness. Aging Cell, 2014, 13:576–8., DOI 10.1111/acel.12196 Toth P, Tarantini S, Springo Z, Tucsek Z, Gautam T, Giles CB, et al. Aging exacerbates hypertension-induced cerebralmicrohemorrhages inmice: role of resveratrol treatment in vasoprotection. Aging Cell, 2015, 14:400–8., DOI 10.1111/acel.12315 Ashton KJ, Kiessling CJ, Thompson JM, Aziz AY, Thomas WG, Headrick JP, et al. Early cardiac aging linked to impaired stress-resistance and transcriptional control of stress response, quality control and mitochondrial pathways. Exp Gerontol, 2023, 171:112011., DOI 10.1016/j.exger.2022.112011 de Souza RG, Yu Z, Hernandez H, Trujillo-Vargas CM, Lee A, Mauk KE, et al. Modulation of oxidative stress and inflammation in the aged lacrimal gland. Am J Pathol, 2021, 191:294–308., DOI 10.1016/j.ajpath.2020.10.013 Suh JH, Shenvi SV, Dixon BM, Liu H, Jaiswal AK, Liu RM, et al. Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid. Proc Natl Acad Sci U S A, 2004, 101:3381–6., DOI 10.1073/pnas.0400282101 Csiszar A, Gautam T, Sosnowska D, Tarantini S, Banki E, Tucsek Z, et al. Caloric restriction confers persistent anti-oxidative, pro-angiogenic, and antiinflammatory effects and promotes anti-aging miRNA expression profile in cerebromicrovascular endothelial cells of aged rats. Am J Physiol Heart Circ Physiol, 2014, 307:H292–306., DOI 10.1152/ajpheart.00307.2014 Martin GM, Hisama FM, Oshima J. Review of how genetic research on segmental progeroid syndromes has documented genomic instability as a hallmark of aging but let us now pursue antigeroid syndromes. J Gerontol A Biol Sci Med Sci, 2021, 76:253–9., DOI 10.1093/gerona/glaa273 Durik M, Kavousi M, van der Pluijm I, Isaacs A, Cheng C, Verdonk K, et al. Nucleotide excision DNA repair is associated with age-related vascular dysfunction. Circulation, 2012, 126:468–78., DOI 10.1161/CIRCULATIONAHA.112.104380 Gray K, Kumar S, Figg N, Harrison J, Baker L, Mercer J, et al. Effects of DNA damage in smooth muscle cells in atherosclerosis. Circ Res, 2015, 116:816–26., DOI 10.1161/CIRCRESAHA.116.304921 Diez-Diez M, Amoros-Perez M, de la Barrera J, Vazquez E, Quintas A, Pascual-Figal DA, et al. Clonal hematopoiesis is not prevalent in Hutchinson- Gilford progeria syndrome. Geroscience, 2023, 45:1231–6., DOI 10.1007/s11357-022- 00607-2 Del Pozo-Valero M, Corton M, Lopez-Rodriguez R, Mahillo-Fernandez I, Ruiz-Hornillos J, Minguez P, et al. Age-dependent association of clonal hematopoiesis with COVID-19 mortality in patients over 60 years. Geroscience, 2023, 45:543–53., DOI 10.1007/s11357-022-00666-5 Steensma DP, Bejar R, Jaiswal S, Lindsley RC, Sekeres MA, Hasserjian RP, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood, 2015, 126:9–16., DOI 10.1182/blood-2015-03- 631747 Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N Engl J Med, 2017, 377:111–21., DOI 10.1056/NEJMoa1701719 Fuster JJ, Walsh K. Somatic mutations and clonal hematopoiesis: unexpected potential new drivers of age-related cardiovascular disease. Circ Res, 2018, 122: 523–32., DOI 10.1161/CIRCRESAHA.117.312115 Libby P, Ebert BL. CHIP, clonal hematopoiesis of indeterminate potential): potent and newly recognized contributor to cardiovascular risk. Circulation, 2018, 138:666–8., DOI 10.1161/CIRCULATIONAHA.118.034392 Khetarpal SA, Qamar A, Bick AG, Fuster JJ, Kathiresan S, Jaiswal S, et al. Clonal hematopoiesis of indeterminate potential reshapes age-related CVD: JACC review topic of the week. J Am Coll Cardiol, 2019, 74:578–86., DOI 10.1016/j.jacc. 2019.05.045 Nachun D, Lu AT, Bick AG, Natarajan P, Weinstock J, Szeto MD, et al. Clonal hematopoiesis associated with epigenetic aging and clinical outcomes. Aging Cell, 2021, 20:e13366., DOI 10.1111/acel.13366 Bhattacharya R, Zekavat SM, Haessler J, Fornage M, Raffield L, Uddin MM, et al. Clonal hematopoiesis is associated with higher risk of stroke. Stroke, 2022, 53: 788–97., DOI 10.1161/STROKEAHA.121.037388 Uddin MDM, Nguyen NQH, Yu B, Brody JA, Pampana A, Nakao T, et al. Clonal hematopoiesis of indeterminate potential, DNA methylation, and risk for coronary artery disease. Nat Commun, 2022, 13:5350., DOI 10.1038/s41467-022- 33093-3 Huang Z, Liu C, Ruan Y, Guo Y, Sun S, Shi Y, et al. Dynamics of leukocyte telomere length in adults aged 50 and older: a longitudinal population-based cohort study. Geroscience, 2021, 43:645–54., DOI 10.1007/s11357-020-00320-y Sindi S, Solomon A, Kareholt I, Hovatta I, Antikainen R, Hanninen T, et al. Telomere length change in a multidomain lifestyle intervention to prevent cognitive decline: a randomized clinical trial. J Gerontol A Biol Sci Med Sci, 2021, 76:491–8., DOI 10.1093/gerona/glaa279 Pudas S, Josefsson M, Nordin Adolfsson A, Landfors M, Kauppi K, Veng- Taasti LM, et al. Short leukocyte telomeres, but not telomere attrition rates, predict memory decline in the 20-year longitudinal betula study. J Gerontol A Biol Sci Med Sci, 2021, 76:955–63., DOI 10.1093/gerona/glaa322 Kemp BR, Ferraro KF. Are biological consequences of childhood exposures detectable in telomere length decades later? J Gerontol A Biol Sci Med Sci, 2021, 76: 7–14., DOI 10.1093/gerona/glaa019 Demanelis K, Tong L, Pierce BL. Genetically increased telomere length and aging-related traits in the U.K. Biobank. J Gerontol A Biol Sci Med Sci, 2021, 76: 15–22., DOI 10.1093/gerona/glz240 Pearce EE, Alsaggaf R, Katta S, Dagnall C, Aubert G, Hicks BD, et al. Telomere length and epigenetic clocks as markers of cellular aging: a comparative study. Geroscience, 2022, 44:1861–9., DOI 10.1007/s11357-022-00586-4 Zhan Y, Hagg S. Association between genetically predicted telomere length and facial skin aging in the UK Biobank: a Mendelian randomization study. Geroscience, 2021, 43:1519–25., DOI 10.1007/s11357-020-00283-0 El Assar M, Angulo J, Carnicero JA, Walter S, Garcia-Garcia FJ, Rodriguez-Artalejo F, et al. Association between telomere length, frailty and death in older adults. Geroscience, 2021, 43:1015–27., DOI 10.1007/s11357-020- 00291-0 Katz R, Gay EL, Kuipers AL, Lee JH, Honig LS, Christensen K, et al. Association of leukocyte telomere length with perceived physical fatigability. Exp Gerontol, 2022, 170:111988., DOI 10.1016/j.exger.2022.111988 Yusipov I, Kondakova E, Kalyakulina A, Krivonosov M, Lobanova N, Bacalini MG, et al. Accelerated epigenetic aging and inflammatory/immunological profile, ipAGE, in patients with chronic kidney disease. Geroscience, 2022, 44:817–34., DOI 10.1007/s11357-022-00540-4 Vetter VM, Sommerer Y, Kalies CH, Spira D, Bertram L, Demuth I. Vitamin D supplementation is associated with slower epigenetic aging. Geroscience, 2022, 44: 1847–59., DOI 10.1007/s11357-022-00581-9 Fraszczyk E, Thio CHL, Wackers P, Dolle MET, Bloks VW, Hodemaekers H, et al. DNA methylation trajectories and accelerated epigenetic aging in incident type 2 diabetes. Geroscience, 2022, 44:2671–84., DOI 10.1007/s11357-022-00626-z Schachtschneider KM, Schook LB, Meudt JJ, Shanmuganayagam D, Zoller JA, Haghani A, et al. Epigenetic clock and DNA methylation analysis of porcine models of aging and obesity. Geroscience, 2021, 43:2467–83., DOI 10.1007/s11357- 021-00439-6 Horvath S, Zoller JA, Haghani A, Lu AT, Raj K, Jasinska AJ, et al. DNA methylation age analysis of rapamycin in common marmosets. Geroscience, 2021, 43:2413–25., DOI 10.1007/s11357-021-00438-7 Horvath S, Zoller JA, Haghani A, Jasinska AJ, Raj K, Breeze CE, et al. Epigenetic clock and methylation studies in the rhesus macaque. Geroscience, 2021, 43:2441–53., DOI 10.1007/s11357-021-00429-8 Tolkach Y, Zarbl R, Bauer S, Ritter M, Ellinger J, Hauser S, et al. DNA promoter methylation and ERG regulate the expression of CD24 in prostate cancer. Am J Pathol, 2021, 191:618–30., DOI 10.1016/j.ajpath.2020.12.014 Toghill BJ, Saratzis A, Harrison SC, Verissimo AR, Mallon EB, Bown MJ. The potential role of DNA methylation in the pathogenesis of abdominal aortic aneurysm. Atherosclerosis, 2015, 241:121–9., DOI 10.1016/j.atherosclerosis.2015. 05.001 Li P, Newhardt MF, Matsuzaki S, Eyster C, Pranay A, Peelor FF, 3rd, et al. The loss of cardiac SIRT3 decreases metabolic flexibility and proteostasis in an agedependent manner. Geroscience, 2023, 45:983–99., DOI 10.1007/s11357-022- 00695-0 Jang YC, Rodriguez K, Lustgarten MS, Muller FL, Bhattacharya A, Pierce A, et al. Superoxide-mediated oxidative stress accelerates skeletal muscle atrophy by synchronous activation of proteolytic systems. Geroscience, 2020, 42:1579–91., DOI 10.1007/s11357-020-00200-5 Tomaru U, Ito T, Ohmura Y, Higashikawa K, Miyajima S, Tomatsu R, et al. Decreased proteasomal function induces neuronal loss and memory impairment. Am J Pathol, 2021, 191:144–56., DOI 10.1016/j.ajpath.2020.10.004 Gal J, Katsumata Y, Zhu H, Srinivasan S, Chen J, Johnson LA, et al. Apolipoprotein E proteinopathy is a major dementia-associated pathologic biomarker in individuals with or without the APOE epsilon 4 allele. Am J Pathol, 2022, 192:564–78., DOI 10.1016/j.ajpath.2021.11.013 Sabatini DM. Twenty-five years of mTOR: uncovering the link from nutrients to growth. Proc Natl Acad Sci U S A, 2017, 114:11818–25., DOI 10. 1073/pnas.1716173114 Teuliere J, Bernard C, Corel E, Lapointe FJ, Martens J, Lopez P, et al. Network analyses unveil ageing-associated pathways evolutionarily conserved from fungi to animals. Geroscience, 2023, 45:1059–80., DOI 10.1007/s11357-022-00704-2 Mota-Martorell N, Jove M, Pradas I, Berdun R, Sanchez I, Naudi A, et al. Gene expression and regulatory factors of the mechanistic target of rapamycin, mTOR, complex 1 predict mammalian longevity. Geroscience, 2020, 42:1157–73., DOI 10.1007/s11357-020-00210-3 Selvarani R, Mohammed S, Richardson A. Effect of rapamycin on aging and age-related diseases-past and future. Geroscience, 2021, 43:1135–58., DOI 10.1007/ s11357-020-00274-1 Castro-Martinez F, Candelario-Martinez A, Encarnacion-Garcia MR, Piedra-Quintero Z, Bonilla-Moreno R, Betanzos A, et al. Rictor/Mammalian target of rapamycin complex 2 signaling protects colonocytes from apoptosis and prevents epithelial barrier breakdown. Am J Pathol, 2021, 191:1537–49., DOI 10.1016/j.ajpath.2021.06.004 Lee HJ, Shin DH, Song JS, Park JY, Kim SY, Hwang CS, et al. mTOR inhibition increases transcription factor E3, TFE3, activity and modulates programmed death-ligand 1, PD-L1, expression in translocation renal cell carcinoma. Am J Pathol, 2021, 191:1999–2008., DOI 10.1016/j.ajpath.2021.07.007 Lu X, Peng B, Chen G, Pes MG, Ribback S, Ament C, et al. YAP accelerates notch-driven cholangiocarcinogenesis via mTORC1 in mice. Am J Pathol, 2021, 191:1651–67., DOI 10.1016/j.ajpath.2021.05.017 Sahu D, Huan J, Wang H, Sahoo D, Casteel DE, Klemke RL, et al. Bladder cancer invasion is mediated by mammalian target of rapamycin complex 2-driven regulation of nitric oxide and invadopodia formation. Am J Pathol, 2021, 191: 2203–18., DOI 10.1016/j.ajpath.2021.08.002 Wang M, Miller RA. Fibroblasts from long-lived mutant mice exhibit increased autophagy and lower TOR activity after nutrient deprivation or oxidative stress. Aging Cell, 2012, 11:668–74., DOI 10.1111/j.1474-9726.2012. 00833.x LaRocca TJ, Gioscia-Ryan RA, Hearon CM, Seals DR. The autophagy enhancer spermidine reverses arterial aging. Mech Ageing Dev, 2013, 134: 314–20., DOI 10.1016/j.mad.2013.04.004 Kang C, Xu Q, Martin TD, Li MZ, Demaria M, Aron L, et al. The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4. Science, 2015, 349:aaa5612., DOI 10.1126/science.aaa5612 Cuervo AM, Dice JF. Age-related decline in chaperone-mediated autophagy. J Biol Chem, 2000, 275:31505–13., DOI 10.1074/jbc.M002102200 Tabibzadeh S. Role of autophagy in aging: the good, the bad, and the ugly. Aging Cell, 2023, 22:e13753., DOI 10.1111/acel.13753 Martinez RAS, Pinky PD, Harlan BA, Brewer GJ. GTP energy dependence of endocytosis and autophagy in the aging brain and Alzheimer’s disease. Geroscience, 2023, 45:757–80., DOI 10.1007/s11357-022-00717-x Kaushik S, Tasset I, Arias E, Pampliega O, Wong E, Martinez-Vicente M, et al. Autophagy and the hallmarks of aging. Ageing Res Rev, 2021, 72:101468., DOI 10.1016/j.arr.2021.101468 Aman Y, Schmauck-Medina T, Hansen M, Morimoto RI, Simon AK, Bjedov I, et al. Autophagy in healthy aging and disease. Nat Aging, 2021, 1:634–50., DOI 10. 1038/s43587-021-00098-4 Zhang K, Huang Q, Peng L, Lin S, Liu J, Zhang J, et al. The multifunctional roles of autophagy in the innate immune response: implications for regulation of transplantation rejection. Front Cel Dev Biol, 2022, 10:1007559., DOI 10.3389/fcell. 2022.1007559 Wu YX, Jin SH, Cui J. Autophagy and immune tolerance. Adv Exp Med Biol, 2019, 1206:635–65., DOI 10.1007/978-981-15-0602-4_28 Hashemi M, Paskeh MDA, Orouei S, Abbasi P, Khorrami R, Dehghanpour A, et al. Towards dual function of autophagy in breast cancer: a potent regulator of tumor progression and therapy response. Biomed Pharmacother, 2023, 161:114546., DOI 10.1016/j.biopha.2023.114546 Ballesteros-Alvarez J, Andersen JKmTORC2. mTORC2: the other mTOR in autophagy regulation. Aging Cell, 2021, 20:e13431., DOI 10.1111/acel.13431 Al-Bari MAA, Xu P. Molecular regulation of autophagy machinery by mTOR-dependent and -independent pathways. Ann N Y Acad Sci, 2020, 1467: 3–20., DOI 10.1111/nyas.14305 Keller CW, Adamopoulos IE, Lunemann JD. Autophagy pathways in autoimmune diseases. J Autoimmun, 2023, 136:103030., DOI 10.1016/j.jaut.2023. 103030 Zhou L, Pinho R, Gu Y, Radak Z. The role of SIRT3 in exercise and aging. Cells, 2022, 11:2596., DOI 10.3390/cells11162596 Lautrup S, Sinclair DA, Mattson MP, Fang EF. NAD(+, in brain aging and neurodegenerative disorders. Cell Metab, 2019, 30:630–55., DOI 10.1016/j.cmet. 2019.09.001 Yoshino J, Baur JA, Imai SI. NAD(+, intermediates: the biology and therapeutic potential of NMN and NR. Cel Metab, 2018, 27:513–28., DOI 10. 1016/j.cmet.2017.11.002 Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cel Metab, 2018, 27:529–47., DOI 10.1016/j.cmet. 2018.02.011 Ansari A, Rahman MS, Saha SK, Saikot FK, Deep A, Kim KH. Function of the SIRT3 mitochondrial deacetylase in cellular physiology, cancer, and neurodegenerative disease. Aging Cell, 2017, 16:4–16., DOI 10.1111/acel.12538 KitadaM, Ogura Y, Koya D. The protective role of Sirt1 in vascular tissue: its relationship to vascular aging and atherosclerosis. Aging, Albany NY,, 2016, 8: 2290–307., DOI 10.18632/aging.101068 Imai SI, Guarente L. It takes two to tango: NAD(+, and sirtuins in aging/ longevity control. NPJ AgingMech Dis, 2016, 2:16017., DOI 10.1038/npjamd.2016.17 Houtkooper RH, Pirinen E, Auwerx J. Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Cel Biol, 2012, 13:225–38., DOI 10.1038/nrm3293 Chalkiadaki A, Guarente L. Sirtuins mediate mammalian metabolic responses to nutrient availability. Nat Rev Endocrinol, 2012, 8:287–96., DOI 10. 1038/nrendo.2011.225 Minor RK, Baur JA, Gomes AP, Ward TM, Csiszar A, Mercken EM, et al. SRT1720 improves survival and healthspan of obese mice. Sci Rep, 2011, 1:70., DOI 10.1038/srep00070 Csiszar A, Labinskyy N, Jimenez R, Pinto JT, Ballabh P, Losonczy G, et al. Anti-oxidative and anti-inflammatory vasoprotective effects of caloric restriction in aging: role of circulating factors and SIRT1. Mech Ageing Dev, 2009, 130:518–27., DOI 10.1016/j.mad.2009.06.004 Ferrara N, Rinaldi B, Corbi G, Conti V, Stiuso P, Boccuti S, et al. Exercise training promotes SIRT1 activity in aged rats. Rejuvenation Res, 2008, 11:139–50., DOI 10.1089/rej.2007.0576 Csiszar A, Labinskyy N, Podlutsky A, Kaminski PM, Wolin MS, Zhang C, et al. Vasoprotective effects of resveratrol and SIRT1: attenuation of cigarette smoke-induced oxidative stress and proinflammatory phenotypic alterations. Am J Physiol Heart Circ Physiol, 2008, 294:H2721–35., DOI 10.1152/ajpheart.00235.2008 Dasgupta B, Milbrandt J. Resveratrol stimulates AMP kinase activity in neurons. Proc Natl Acad Sci USA, 2007, 104:7217–22., DOI 10.1073/pnas. 0610068104 Alcendor RR, Gao S, Zhai P, Zablocki D, Holle E, Yu X, et al. Sirt1 regulates aging and resistance to oxidative stress in the heart. Circ Res, 2007, 100:1512–21., DOI 10.1161/01.RES.0000267723.65696.4a Guarente L. Calorie restriction and SIR2 genes--towards a mechanism. Mech Ageing Dev, 2005, 126:923–8., DOI 10.1016/j.mad.2005.03.013 Nemoto S, Fergusson MM, Finkel T. Nutrient availability regulates SIRT1 through a forkhead-dependent pathway. Science, 2004, 306:2105–8., DOI 10.1126/science.1101731 Cohen HY, Miller C, Bitterman KJ, Wall NR, Hekking B, Kessler B, et al. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science, 2004, 305:390–2., DOI 10.1126/science.1099196 Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature, 2003, 425:191–6., DOI 10.1038/nature01960 You Y, Liang W. SIRT1 and SIRT6: the role in aging-related diseases. Biochim Biophys Acta Mol Basis Dis, 2023, 1869:166815., DOI 10.1016/j.bbadis.2023. 166815 Chen C, Zhou M, Ge Y, Wang X. SIRT1 and aging related signaling pathways. Mech Ageing Dev, 2020, 187:111215., DOI 10.1016/j.mad.2020.111215 Raza U, Tang X, Liu Z, Liu B. SIRT7: the seventh key to unlocking the mystery of aging. Physiol Rev, 2024, 104:253–80., DOI 10.1152/physrev.00044.2022 Li H, Cai Z. SIRT3 regulates mitochondrial biogenesis in aging-related diseases. J Biomed Res, 2022, 37:77–88., DOI 10.7555/JBR.36.20220078 McDonnell E, Peterson BS, Bomze HM, Hirschey MD. SIRT3 regulates progression and development of diseases of aging. Trends Endocrinol Metab, 2015, 26:486–92., DOI 10.1016/j.tem.2015.06.001 He L, Liu Q, Cheng J, Cao M, Zhang S, Wan X, et al. SIRT4 in ageing. Biogerontology, 2023, 24:347–62., DOI 10.1007/s10522-023-10022-5 Fabbrizi E, Fiorentino F, Carafa V, Altucci L, Mai A, Rotili D. Emerging roles of SIRT5 in metabolism, cancer, and SARS-CoV-2 infection. Cells, 2023, 12:852., DOI 10.3390/cells12060852 Bringman-Rodenbarger LR, Guo AH, Lyssiotis CA, Lombard DB. Emerging roles for SIRT5 in metabolism and cancer. Antioxid Redox Signal, 2018, 28:677–90., DOI 10.1089/ars.2017.7264 Wallace DC. A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet, 2005, 39:359–407., DOI 10.1146/annurev.genet.39.110304.095751 Schriner SE, Linford NJ, Martin GM, Treuting P, Ogburn CE, Emond M, et al. Extension of murine life span by overexpression of catalase targeted to mitochondria. Science, 2005, 308:1909–11., DOI 10.1126/science.1106653 Herbst A, Prior SJ, Lee CC, Aiken JM, McKenzie D, Hoang A, et al. Skeletal muscle mitochondrial DNA copy number and mitochondrial DNA deletion mutation frequency as predictors of physical performance in older men and women. Geroscience, 2021, 43:1253–64., DOI 10.1007/s11357-021-00351-z Ahmed NS, Foote JB, Singh KK. Impaired mitochondria promote agingassociated sebaceous gland dysfunction and pathology. Am J Pathol, 2022, 192: 1546–58., DOI 10.1016/j.ajpath.2022.07.006 Reiss AB, Ahmed S, Dayaramani C, Glass AD, Gomolin IH, Pinkhasov A, et al. The role of mitochondrial dysfunction in Alzheimer’s disease: a potential pathway to treatment. Exp Gerontol, 2022, 164:111828., DOI 10.1016/j.exger.2022. 111828 Springo Z, Tarantini S, Toth P, Tucsek Z, Koller A, Sonntag WE, et al. Aging exacerbates pressure-induced mitochondrial oxidative stress in mouse cerebral arteries. J Gerontol A Biol Sci Med Sci, 2015, 70:1355–9., DOI 10.1093/gerona/glu244 Tarantini S, Valcarcel-Ares NM, Yabluchanskiy A, Fulop GA, Hertelendy P, Gautam T, et al. Treatment with themitochondrial-targeted antioxidant peptide SS- 31 rescues neurovascular coupling responses and cerebrovascular endothelial function and improves cognition in aged mice. Aging Cell, 2018, 17:e12731., DOI 10. 1111/acel.12731 Zampino M, Spencer RG, Fishbein KW, Simonsick EM, Ferrucci L. Cardiovascular health and mitochondrial function: testing an association. J Gerontol A Biol Sci Med Sci, 2021, 76:361–7., DOI 10.1093/gerona/glaa297 Saberi M, Zhang X, Mobasheri A. Targeting mitochondrial dysfunction with small molecules in intervertebral disc aging and degeneration. Geroscience, 2021, 43:517–37., DOI 10.1007/s11357-021-00341-1 Bua E, Johnson J, Herbst A, Delong B, McKenzie D, Salamat S, et al. Mitochondrial DNA-deletion mutations accumulate intracellularly to detrimental levels in aged human skeletal muscle fibers. Am J Hum Genet, 2006, 79:469–80., DOI 10.1086/507132 Swift ML, Sell C, Azizkhan-Clifford J. DNA damage-induced degradation of Sp1 promotes cellular senescence. Geroscience, 2022, 44:683–98., DOI 10.1007/ s11357-021-00456-5 Han X, Lei Q, Xie J, Liu H, Li J, Zhang X, et al. Potential regulators of the senescence-associated secretory phenotype during senescence and aging. J Gerontol A Biol Sci Med Sci, 2022, 77:2207–18., DOI 10.1093/gerona/glac097 Matacchione G, Perugini J, Di Mercurio E, Sabbatinelli J, Prattichizzo F, Senzacqua M, et al. Senescent macrophages in the human adipose tissue as a source of inflammaging. Geroscience, 2022, 44:1941–60., DOI 10.1007/s11357-022-00536-0 Fielding RA, Atkinson EJ, Aversa Z, White TA, Heeren AA, Achenbach SJ, et al. Associations between biomarkers of cellular senescence and physical function in humans: observations from the lifestyle interventions for elders, LIFE, study. Geroscience, 2022, 44:2757–70., DOI 10.1007/s11357-022-00685-2 Kavanagh K, Sherrill C, Ruggiero A, Block M, Vemuri R, Davis M, et al. Biomarkers of senescence in non-human primate adipose depots relate to aging. Geroscience, 2021, 43:343–52., DOI 10.1007/s11357-020-00230-z Karin O, Alon U. Senescent cell accumulation mechanisms inferred from parabiosis. Geroscience, 2021, 43:329–41., DOI 10.1007/s11357-020-00286-x Hense JD, Garcia DN, Isola JV, Alvarado-Rincon JA, Zanini BM, Prosczek JB, et al. Senolytic treatment reverses obesity-mediated senescent cell accumulation in the ovary. Geroscience, 2022, 44:1747–59., DOI 10.1007/s11357-022-00573-9 Dungan CM, Figueiredo VC, Wen Y, VonLehmden GL, Zdunek CJ, Thomas NT, et al. Senolytic treatment rescues blunted muscle hypertrophy in old mice. Geroscience, 2022, 44:1925–40., DOI 10.1007/s11357-022-00542-2 Iijima H, Gilmer G, Wang K, Sivakumar S, Evans C, Matsui Y, et al. Metaanalysis integrated with multi-omics data analysis to elucidate pathogenic mechanisms of age-related knee osteoarthritis in mice. J Gerontol A Biol Sci Med Sci, 2022, 77:1321–34., DOI 10.1093/gerona/glab386 Csipo T, Lipecz A, Ashpole NM, Balasubramanian P, Tarantini S. Astrocyte senescence contributes to cognitive decline. Geroscience, 2020, 42:51–5., DOI 10. 1007/s11357-019-00140-9 Kiss T, Nyul-Toth A, Balasubramanian P, Tarantini S, Ahire C, DelFavero J, et al. Single-cell RNA sequencing identifies senescent cerebromicrovascular endothelial cells in the aged mouse brain. Geroscience, 2020, 42:429–44., DOI 10. 1007/s11357-020-00177-1 Tarantini S, Balasubramanian P, Delfavero J, Csipo T, Yabluchanskiy A, Kiss T, et al. Treatment with the BCL-2/BCL-xL inhibitor senolytic drug ABT263/ Navitoclax improves functional hyperemia in aged mice. Geroscience, 2021, 43: 2427–40., DOI 10.1007/s11357-021-00440-z Chae JB, Jang H, Son C, Park CW, Choi H, Jin S, et al. Targeting senescent retinal pigment epithelial cells facilitates retinal regeneration in mouse models of age-related macular degeneration. Geroscience, 2021, 43:2809–33., DOI 10.1007/ s11357-021-00457-4 Park JW, Park JE, Kim SR, Sim MK, Kang CM, Kim KS. Metformin alleviates ionizing radiation-induced senescence by restoring BARD1-mediated DNA repair in human aortic endothelial cells. Exp Gerontol, 2022, 160:111706., DOI 10.1016/j. exger.2022.111706 Yabluchanskiy A, Tarantini S, Balasubramanian P, Kiss T, Csipo T, Fulop GA, et al. Pharmacological or genetic depletion of senescent astrocytes prevents whole brain irradiation-induced impairment of neurovascular coupling responses protecting cognitive function in mice. Geroscience, 2020, 42:409–28., DOI 10.1007/ s11357-020-00154-8 Sun T, Zhang L, Feng J, Bao L, Wang J, Song Z, et al. Characterization of cellular senescence in doxorubicin-induced aging mice. Exp Gerontol, 2022, 163: 111800., DOI 10.1016/j.exger.2022.111800 Khadirnaikar S, Chatterjee A, Shukla S. Identification and characterization of senescence phenotype in lung adenocarcinoma with high drug sensitivity. Am J Pathol, 2021, 191:1966–73., DOI 10.1016/j.ajpath.2021.07.005 Aires R, Porto ML, de Assis LM, Pereira PAN, Carvalho GR, Coco LZ, et al. DNA damage and aging on hematopoietic stem cells: impact of oxidative stress in ApoE(-)/(-, mice. Exp Gerontol, 2021, 156:111607., DOI 10.1016/j.exger.2021. 111607 Bencivenga L, StrumiaM, Rolland Y,Martinez L, Cestac P, Guyonnet S, et al. Biomarkers of mitochondrial dysfunction and inflammaging in older adults and blood pressure variability. Geroscience, 2023, 45:797–809., DOI 10.1007/s11357-022- 00697-y Dugue PA, Hodge AM, Ulvik A, Ueland PM, Midttun O, Rinaldi S, et al. Association of markers of inflammation, the kynurenine pathway and B vitamins with age and mortality, and a signature of inflammaging. J Gerontol A Biol Sci Med Sci, 2022, 77:826–36., DOI 10.1093/gerona/glab163 Cribb L, Hodge AM, Yu C, Li SX, English DR, Makalic E, et al. Inflammation and epigenetic aging are largely independent markers of biological aging and mortality. J Gerontol A Biol Sci Med Sci, 2022, 77:2378–86., DOI 10.1093/gerona/ glac147 ZamboniM, Nori N, Brunelli A, Zoico E. How does adipose tissue contribute to inflammageing? Exp Gerontol, 2021, 143:111162., DOI 10.1016/j.exger.2020. 111162 Sanfilippo C, Castrogiovanni P, Vinciguerra M, Imbesi R, Ulivieri M, Fazio F, et al. A sex-stratified analysis of neuroimmune gene expression signatures in Alzheimer’s disease brains. Geroscience, 2023, 45:523–41., DOI 10.1007/s11357- 022-00664-7 Kiss T, Nyul-Toth A, DelFavero J, Balasubramanian P, Tarantini S, Faakye J, et al. Spatial transcriptomic analysis reveals inflammatory foci defined by senescent cells in the white matter, hippocampi and cortical grey matter in the aged mouse brain. Geroscience, 2022, 44:661–81., DOI 10.1007/s11357-022-00521-7 Thadathil N, Nicklas EH, Mohammed S, Lewis TL, Richardson A, Deepa SS. Necroptosis increases with age in the brain and contributes to age-related neuroinflammation. Geroscience, 2021, 43:2345–61., DOI 10.1007/s11357-021- 00448-5 Mehdipour M, Mehdipour T, Skinner CM, Wong N, Liu C, Chen CC, et al. Plasma dilution improves cognition and attenuates neuroinflammation in old mice. Geroscience, 2021, 43:1–18., DOI 10.1007/s11357-020-00297-8 Liang Z, Zhang T, Liu H, Li Z, Peng L, Wang C, et al. Inflammaging: the ground for sarcopenia? Exp Gerontol, 2022, 168:111931., DOI 10.1016/j.exger.2022. 111931 Luo J, le Cessie S, Blauw GJ, Franceschi C, Noordam R, van Heemst D. Systemic inflammatory markers in relation to cognitive function and measures of brain atrophy: a Mendelian randomization study. Geroscience, 2022, 44:2259–70., DOI 10.1007/s11357-022-00602-7 Lu WH, Giudici KV, Morley JE, Guyonnet S, Parini A, Aggarwal G, et al. Investigating the combination of plasma amyloid-beta and geroscience biomarkers on the incidence of clinically meaningful cognitive decline in older adults. Geroscience, 2022, 44:1489–503., DOI 10.1007/s11357-022-00554-y Jimenez L, Silva A, Calissi G, Grenho I, Monteiro R, Mayoral-Varo V, et al. Screening health-promoting compounds for their capacity to induce the activity of FOXO3. J Gerontol A Biol Sci Med Sci, 2022, 77:1485–93., DOI 10.1093/gerona/ glab265 Li S, Hou Y, Liu K, Zhu H, Qiao M, Sun X, et al. Metformin protects against inflammation, oxidative stress to delay poly I:C-induced aging-like phenomena in the gut of an annual fish. J Gerontol A Biol Sci Med Sci, 2022, 77:276–82., DOI 10. 1093/gerona/glab298 Brazao V, Colato RP, Santello FH, Duarte A, Goulart A, Sampaio PA, et al. Melatonin regulates antioxidant defense and inflammatory response by activating Nrf2-dependent mechanisms and inhibiting NFkappaB expression in middle-aged T. cruzi infected rats. Exp Gerontol, 2022, 167:111895., DOI 10.1016/j.exger.2022.111895 Caldeira CA, Santos MA, Araujo GR, Lara RC, Franco FN, Chaves MM. Resveratrol: change of SIRT 1 and AMPK signaling pattern during the aging process. Exp Gerontol, 2021, 146:111226., DOI 10.1016/j.exger.2021.111226 Gregory S, Pullen H, Ritchie CW, Shannon OM, Stevenson EJ, Muniz- Terrera G. Mediterranean diet and structural neuroimaging biomarkers of Alzheimer’s and cerebrovascular disease: a systematic review. Exp Gerontol, 2023, 172:112065., DOI 10.1016/j.exger.2022.112065 Chenhuichen C, Cabello-Olmo M, Barajas M, Izquierdo M, Ramirez-Velez R, Zambom-Ferraresi F, et al. Impact of probiotics and prebiotics in the modulation of the major events of the aging process: a systematic review of randomized controlled trials. Exp Gerontol, 2022, 164:111809., DOI 10.1016/j.exger.2022.111809 Ezzati A, Pak VM. The effects of time-restricted eating on sleep, cognitive decline, and Alzheimer’s disease. Exp Gerontol, 2023, 171:112033., DOI 10.1016/j. exger.2022.112033 Ramaker ME, Corcoran DL, Apsley AT, Kobor MS, Kraus VB, Kraus WE, et al. Epigenome-wide association study analysis of calorie restriction in humans, CALERIETM trial analysis. J Gerontol A Biol Sci Med Sci, 2022, 77:2395–401., DOI 10.1093/gerona/glac168 Fassier P, Kang JH, Lee IM, Grodstein F, Vercambre MN. Vigorous Physical Activity and Cognitive Trajectory Later in Life: prospective Association and Interaction by Apolipoprotein E e4 in the Nurses’ Health Study. J Gerontol A Biol Sci Med Sci, 2022, 77:817–25., DOI 10.1093/gerona/glab169 Gudiksen A, Qoqaj A, Ringholm S, Wojtaszewski J, Plomgaard P, Pilegaard H. Ameliorating effects of lifelong physical activity on healthy aging and mitochondrial function in human white adipose tissue. J Gerontol A Biol Sci Med Sci, 2022, 77:1101–11., DOI 10.1093/gerona/glab356 Romashkan S, Chang H, Hadley EC. National institute on aging workshop: repurposing drugs or dietary supplements for their senolytic or senomorphic effects: considerations for clinical trials. J Gerontol A Biol Sci Med Sci, 2021, 76: 1144–52., DOI 10.1093/gerona/glab028 Saccon TD, Nagpal R, Yadav H, Cavalcante MB, Nunes ADC, Schneider A, et al. Senolytic combination of dasatinib and quercetin alleviates intestinal senescence and inflammation and modulates the gut microbiome in aged mice. J Gerontol A Biol Sci Med Sci, 2021, 76:1895–905., DOI 10.1093/gerona/glab002 Kowald A, Kirkwood TBL. Senolytics and the compression of late-life mortality. Exp Gerontol, 2021, 155:111588., DOI 10.1016/j.exger.2021.111588 Whitson JA, Johnson R, Wang L, Bammler TK, Imai SI, Zhang H, et al. Agerelated disruption of the proteome and acetylome inmouse hearts is associated with loss of function and attenuated by elamipretide, SS-31, and nicotinamide mononucleotide, NMN, treatment. Geroscience, 2022, 44:1621–39., DOI 10.1007/ s11357-022-00564-w Whitson JA,Martin-Perez M, Zhang T, Gaffrey MJ,Merrihew GE, Huang E, et al. Elamipretide, SS-31, treatment attenuates age-associated post-translational modifications of heart proteins. Geroscience, 2021, 43:2395–412., DOI 10.1007/ s11357-021-00447-6 Alfaras I, Di Germanio C, Bernier M, Csiszar A, Ungvari Z, Lakatta EG, et al. Pharmacological strategies to retard cardiovascular aging. Circ Res, 2016, 118: 1626–42., DOI 10.1161/CIRCRESAHA.116.307475 Mitchell SJ, Martin-Montalvo A, Mercken EM, Palacios HH, Ward TM, Abulwerdi G, et al. The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet. Cell Rep, 2014, 6:836–43., DOI 10.1016/j.celrep. 2014.01.031 Hubbard BP, Sinclair DA. Small molecule SIRT1 activators for the treatment of aging and age-related diseases. Trends Pharmacol Sci, 2014, 35:146–54., DOI 10. 1016/j.tips.2013.12.004 Kiss T, Balasubramanian P, Valcarcel-Ares MN, Tarantini S, Yabluchanskiy A, Csipo T, et al. Nicotinamide mononucleotide, NMN, treatment attenuates oxidative stress and rescues angiogenic capacity in aged cerebromicrovascular endothelial cells: a potential mechanism for the prevention of vascular cognitive impairment. Gero Sci, 2019, 41:619–30., DOI 10.1007/s11357-019-00074-2 Kiss T, Giles CB, Tarantini S, Yabluchanskiy A, Balasubramanian P, Gautam T, et al. Nicotinamide mononucleotide, NMN, supplementation promotes antiaging miRNA expression profile in the aorta of aged mice, predicting epigenetic rejuvenation and anti-atherogenic effects. Geroscience, 2019, 41:419–39., DOI 10. 1007/s11357-019-00095-x Tarantini S, Valcarcel-Ares MN, Toth P, Yabluchanskiy A, Tucsek Z, Kiss T, et al. Nicotinamide mononucleotide, NMN, supplementation rescues cerebromicrovascular endothelial function and neurovascular coupling responses and improves cognitive function in aged mice. Redox Biol, 2019, 24:101192., DOI 10. 1016/j.redox.2019.101192 Csiszar A, Tarantini S, Yabluchanskiy A, Balasubramanian P, Kiss T, Farkas E, et al. Role of endothelial NAD+ deficiency in age-related vascular dysfunction. Am J Physiol Heart Circ Physiol, 2019, 316:H1253–H1266., DOI 10.1152/ajpheart. 00039.2019 Kiss T, Nyul-Toth A, Balasubramanian P, Tarantini S, Ahire C, Yabluchanskiy A, et al. Nicotinamide mononucleotide, NMN, supplementation promotes neurovascular rejuvenation in aged mice: transcriptional footprint of SIRT1 activation, mitochondrial protection, anti-inflammatory, and anti-apoptotic effects. Geroscience, 2020, 42:527–46., DOI 10.1007/s11357-020-00165-5 Tarantini S, Yabluchanskiy A, Csipo T, Fulop G, Kiss T, Balasubramanian P, et al. Treatment with the poly(ADP-ribose, polymerase inhibitor PJ-34 improves cerebromicrovascular endothelial function, neurovascular coupling responses and cognitive performance in aged mice, supporting the NAD+ depletion hypothesis of neurovascular aging. Geroscience, 2019, 41:533–42., DOI 10.1007/s11357-019- 00101-2 Das A, Huang GX, Bonkowski MS, Longchamp A, Li C, Schultz MB, et al. Impairment of an endothelial NAD(+)-H2S signaling network is a reversible cause of vascular aging. Cell, 2018, 173:74–89., DOI 10.1016/j.cell.2018.02.008 Clement J, Wong M, Poljak A, Sachdev P, Braidy N. The plasma NAD(+, metabolome is dysregulated in "normal" aging. Rejuvenation Res, 2018, 22:121–30., DOI 10.1089/rej.2018.2077 Mills KF, Yoshida S, Stein LR, Grozio A, Kubota S, Sasaki Y, et al. Long-Term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cel Metab, 2016, 24:795–806., DOI 10.1016/j.cmet. 2016.09.013 de Picciotto NE, Gano LB, Johnson LC, Martens CR, Sindler AL, Mills KF, et al. Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice. Aging Cell, 2016, 15:522–30., DOI 10.1111/acel.12461 Araki T, Sasaki Y, Milbrandt J. Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration. Science, 2004, 305:1010–3., DOI 10. 1126/science.1098014 Blair RV, Vaccari M, Doyle-Meyers LA, Roy CJ, Russell-Lodrigue K, Fahlberg M, et al. Acute respiratory distress in aged, SARS-CoV-2-infected african green monkeys but not rhesus macaques. Am J Pathol, 2021, 191: 274–82., DOI 10.1016/j.ajpath.2020.10.016 Mulka KR, Beck SE, Solis CV, Johanson AL, Queen SE, McCarron ME, et al. Progression and resolution of severe acute respiratory syndrome coronavirus 2, SARS-CoV-2, infection in golden Syrian hamsters. Am J Pathol, 2022, 192: 195–207., DOI 10.1016/j.ajpath.2021.10.009 Gustine JN, Jones D. Immunopathology of hyperinflammation in COVID- 19. Am J Pathol, 2021, 191:4–17., DOI 10.1016/j.ajpath.2020.08.009 Johnson JE, McGuone D, Xu ML, Jane-Wit D, Mitchell RN, Libby P, et al. Coronavirus disease 2019, COVID-19, coronary vascular thrombosis: correlation with neutrophil but not endothelial activation. Am J Pathol, 2022, 192:112–20., DOI 10.1016/j.ajpath.2021.09.004 Martin TR. Lung injury and repair in coronavirus disease 2019-related acute lung injury. Am J Pathol, 2022, 192:406–9., DOI 10.1016/j.ajpath.2022.01.001 Nicosia RF, Ligresti G, Caporarello N, Akilesh S, Ribatti D. COVID-19 vasculopathy: mounting evidence for an indirect mechanism of endothelial injury. Am J Pathol, 2021, 191:1374–84., DOI 10.1016/j.ajpath.2021.05.007 Pujadas E, Beaumont M, Shah H, Schrode N, Francoeur N, Shroff S, et al. Molecular profiling of coronavirus disease 2019, COVID-19, autopsies uncovers novel disease mechanisms. Am J Pathol, 2021, 191:2064–71., DOI 10.1016/j.ajpath. 2021.08.009 Salazar E, Christensen PA, Graviss EA, Nguyen DT, Castillo B, Chen J, et al. Significantly decreased mortality in a large cohort of coronavirus disease 2019, COVID-19, patients transfused early with convalescent plasma containing hightiter anti-severe acute respiratory syndrome coronavirus 2, SARS-CoV-2, spike protein IgG. Am J Pathol, 2021, 191:90–107., DOI 10.1016/j.ajpath.2020.10.008 Ting C, AspalM, Vaishampayan N, Huang SK, Riemondy KA, Wang F, et al. Fatal COVID-19 and non-COVID-19 acute respiratory distress syndrome is associated with incomplete alveolar type 1 epithelial cell differentiation from the transitional state without fibrosis. Am J Pathol, 2022, 192:454–67., DOI 10.1016/j. ajpath.2021.11.014 Whitmore HAB, Kim LA. Understanding the role of blood vessels in the neurologic manifestations of coronavirus disease 2019, COVID-19). Am J Pathol, 2021, 191:1946–54., DOI 10.1016/j.ajpath.2021.04.017 Fagyas M, Nagy B, Raduly AP, Manyine IS, Martha L, Erdosi G, et al. The majority of severe COVID-19 patients develop anti-cardiac autoantibodies. Geroscience, 2022, 44:2347–60., DOI 10.1007/s11357-022-00649-6 Moccia F, Gerbino A, Lionetti V,MiragoliM, Munaron LM, Pagliaro P, et al. COVID-19-associated cardiovascular morbidity in older adults: a position paper from the Italian Society of Cardiovascular Researches. Geroscience, 2020, 42: 1021–49., DOI 10.1007/s11357-020-00198-w Danics K, Pesti A, Toro K, Kiss-Dala N, Szlavik J, Lakatos B, et al. A COVID- 19-association-dependent categorization of death causes in 100 autopsy cases. Geroscience, 2021, 43:2265–87., DOI 10.1007/s11357-021-00451-w Peterfi A, Meszaros A, Szarvas Z, Penzes M, Fekete M, Feher A, et al. Comorbidities and increased mortality of COVID-19 among the elderly: a systematic review. Physiol Int, 2022, 109:163–76., DOI 10.1556/2060.2022.00206 Gado K, Kovacs AK, Domjan G, Nagy ZZ, Bednarik GD. COVID-19 and the elderly. Physiol Int, 2022, 109:177–85., DOI 10.1556/2060.2022.00203 Bello-Chavolla OY, Gonzalez-Diaz A, Antonio-Villa NE, Fermin-Martinez CA, Marquez-Salinas A, Vargas-Vazquez A, et al. Unequal impact of structural health determinants and comorbidity on COVID-19 severity and lethality in older Mexican adults: considerations beyond chronological aging. J Gerontol A Biol Sci Med Sci, 2021, 76:e52–e59., DOI 10.1093/gerona/glaa163 Esme M, Koca M, Dikmeer A, Balci C, Ata N, Dogu BB, et al. Older adults with coronavirus disease 2019: a nationwide study in Turkey. J Gerontol A Biol Sci Med Sci, 2021, 76:e68–e75., DOI 10.1093/gerona/glaa219 Farrelly C. The COVID-19 pandemic, biogerontology, and the ageing of humanity. J Gerontol A Biol Sci Med Sci, 2021, 76:e92–e96., DOI 10.1093/gerona/ glab027 Kuo CL, Pilling LC, Atkins JL, Masoli JAH, Delgado J, Tignanelli C, et al. Biological aging predicts vulnerability to COVID-19 severity in UK biobank participants. J Gerontol A Biol Sci Med Sci, 2021, 76:e133–e141., DOI 10.1093/ gerona/glab060 Marengoni A, Zucchelli A, Vetrano DL, Armellini A, Botteri E, Nicosia F, et al. Beyond chronological age: frailty and multimorbidity predict in-hospital mortality in patients with coronavirus disease 2019. J Gerontol A Biol Sci Med Sci, 2021, 76:e38–e45., DOI 10.1093/gerona/glaa291 Marquez-Salinas A, Fermin-Martinez CA, Antonio-Villa NE, Vargas- Vazquez A, Guerra EC, Campos-Munoz A, et al. Adaptive metabolic and inflammatory responses identified using accelerated aging metrics are linked to adverse outcomes in severe SARS-CoV-2 infection. J Gerontol A Biol Sci Med Sci, 2021, 76:e117–e126., DOI 10.1093/gerona/glab078 Mendes A, Herrmann FR, Perivier S, Gold G, Graf CE, Zekry D. Delirium in older patients with COVID-19: prevalence, risk factors, and clinical relevance. J Gerontol A Biol Sci Med Sci, 2021, 76:e142–e146., DOI 10.1093/gerona/glab039 Poco PCE, Aliberti MJR, Dias MB, Takahashi SF, Leonel FC, Altona M, et al. Divergent: age, frailty, and atypical presentations of COVID-19 in hospitalized patients. J Gerontol A Biol Sci Med Sci, 2021, 76:e46–e51., DOI 10.1093/gerona/ glaa280 Zerah L, Baudouin E, Pepin M, Mary M, Krypciak S, Bianco C, et al. Clinical characteristics and outcomes of 821 older patients with SARS-cov-2 infection admitted to acute care geriatric wards. J Gerontol A Biol Sci Med Sci, 2021, 76: e4–e12., DOI 10.1093/gerona/glaa210 Penfold RS, Zazzara MB, Osterdahl MF, Welch C, Ni Lochlainn M, Freidin MB, et al. Individual factors including age, BMI, and heritable factors underlie temperature variation in sickness and in health: an observational, multi-cohort study. J Gerontol A Biol Sci Med Sci, 2022, 77:1890–7., DOI 10.1093/gerona/glab295 Tisminetzky M, Delude C, Hebert T, Carr C, Goldberg RJ, Gurwitz JH. Age, multiple chronic conditions, and COVID-19: a literature review. J Gerontol A Biol Sci Med Sci, 2022, 77:872–8., DOI 10.1093/gerona/glaa320 VoPham T, Harris HR, Tinker LF, Manson JE, Meliker JR, Wassertheil- Smoller S, et al. The impact of the COVID-19 pandemic on older women in the women’s health initiative. J Gerontol A Biol Sci Med Sci, 2022, 77:S3–S12., DOI 10. 1093/gerona/glac056 Farshbafnadi M, Kamali Zonouzi S, Sabahi M, Dolatshahi M, Aarabi MH. Aging and COVID-19 susceptibility, disease severity, and clinical outcomes: the role of entangled risk factors. Exp Gerontol, 2021, 154:111507., DOI 10.1016/j.exger.2021. 111507 Nidadavolu LS,Walston JD. Underlying vulnerabilities to the cytokine storm and adverse COVID-19 outcomes in the aging immune system. J Gerontol A Biol Sci Med Sci, 2021, 76:e13–e18., DOI 10.1093/gerona/glaa209 Zhou J, Sun Y, Huang W, Ye K. Altered blood cell traits underlie a major genetic locus of severe COVID-19. J Gerontol A Biol Sci Med Sci, 2021, 76: e147–e154., DOI 10.1093/gerona/glab035 Garcia-Torre A, Bueno-Garcia E, Lopez-Martinez R, Rioseras B, Moro- Garcia MA, Alonso-Alvarez S, et al. Surviving older patients show preserved cellular and humoral immunological memory several months after SARS-CoV-2 infection. J Gerontol A Biol Sci Med Sci, 2022, 77:33–40., DOI 10.1093/gerona/glab206 Justice JN, Gubbi S, Kulkarni AS, Bartley JM, Kuchel GA, Barzilai N. A geroscience perspective on immune resilience and infectious diseases: a potential case for metformin. Geroscience, 2021, 43:1093–112., DOI 10.1007/s11357-020- 00261-6 Pence BD. Severe COVID-19 and aging: are monocytes the key? Geroscience, 2020, 42:1051–61., DOI 10.1007/s11357-020-00213-0 Xu K, Wei Y, Giunta S, Zhou M, Xia S. Do inflammaging and coagul-aging play a role as conditions contributing to the co-occurrence of the severe hyperinflammatory state and deadly coagulopathy during COVID-19 in older people? Exp Gerontol, 2021, 151:111423., DOI 10.1016/j.exger.2021.111423 Fekete M, Szarvas Z, Fazekas-Pongor V, Feher A, Dosa N, Lehoczki A, et al. COVID-19 infection in patients with chronic obstructive pulmonary disease: from pathophysiology to therapy. Mini-review. Physiol Int, 2022, 109:9–19., DOI 10.1556/ 2060.2022.00172 Feher A, Szarvas Z, Lehoczki A, Fekete M, Fazekas-Pongor V. Co-infections in COVID-19 patients and correlation with mortality rate. Minireview. Physiol Int, 2022, 109:1–8., DOI 10.1556/2060.2022.00015 Kohli J, Campisi J, Demaria M. A novel suicide gene therapy for the treatment of p16(Ink4a)-overexpressing tumors. Oncotarget, 2018, 9:7274–81., DOI 10.18632/oncotarget.23752 Abdul-Aziz AM, Sun Y, Hellmich C, Marlein CR, Mistry J, Forde E, et al. Acute myeloid leukemia induces protumoral p16INK4a-driven senescence in the bone marrow microenvironment. Blood, 2019, 133:446–56., DOI 10.1182/blood- 2018-04-845420 Patil P, Dong Q, Wang D, Chang J, Wiley C, Demaria M, et al. Systemic clearance of p16(INK4a, -positive senescent cells mitigates age-associated intervertebral disc degeneration. Aging Cell, 2019, 18:e12927., DOI 10.1111/acel. 12927 Jeon OH, Kim C, Laberge RM, Demaria M, Rathod S, Vasserot AP, et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat Med, 2017, 23: 775–81., DOI 10.1038/nm.4324 Kim HN, Chang J, Iyer S, Han L, Campisi J, Manolagas SC, et al. Elimination of senescent osteoclast progenitors has no effect on the age-associated loss of bone mass in mice. Aging Cell, 2019, 18:e12923., DOI 10.1111/acel.12923 Istvan L, Czako C, Elo A,Mihaly Z, Sotonyi P, Varga A, et al. Imaging retinal microvascular manifestations of carotid artery disease in older adults: from diagnosis of ocular complications to understanding microvascular contributions to cognitive impairment. Geroscience, 2021, 43:1703–23., DOI 10.1007/s11357-021- 00392-4 Czako C, Kovacs T, Ungvari Z, Csiszar A, Yabluchanskiy A, Conley S, et al. Retinal biomarkers for Alzheimer’s disease and vascular cognitive impairment and dementia, VCID): implication for early diagnosis and prognosis. Geroscience, 2020, 42:1499–525., DOI 10.1007/s11357-020-00252-7 Drewelies J, Hueluer G, Duezel S, Vetter VM, Pawelec G, Steinhagen- Thiessen E, et al. Using blood test parameters to define biological age among older adults: association with morbidity and mortality independent of chronological age validated in two separate birth cohorts. Geroscience, 2022, 44:2685–99., DOI 10.1007/ s11357-022-00662-9 Cummings SR, Kritchevsky SB. Endpoints for geroscience clinical trials: health outcomes, biomarkers, and biologic age. Geroscience, 2022, 44:2925–31., DOI 10.1007/s11357-022-00671-8 Amgalan A, Maher AS, Ghosh S, Chui HC, Bogdan P, Irimia A. Brain age estimation reveals older adults’ accelerated senescence after traumatic brain injury. Geroscience, 2022, 44:2509–25., DOI 10.1007/s11357-022-00597-1 Kwon D, Belsky DW. A toolkit for quantification of biological age from blood chemistry and organ function test data: BioAge. Geroscience, 2021, 43: 2795–808., DOI 10.1007/s11357-021-00480-5 Crimmins EM, Thyagarajan B, Kim JK, Weir D, Faul J. Quest for a summary measure of biological age: the health and retirement study. Geroscience, 2021, 43: 395–408., DOI 10.1007/s11357-021-00325-1 Chan MS, Arnold M, Offer A, Hammami I, Mafham M, Armitage J, et al. A biomarker-based biological age in UK biobank: composition and prediction of mortality and hospital admissions. J Gerontol A Biol Sci Med Sci, 2021, 76: 1295–302., DOI 10.1093/gerona/glab069 Horvath S, Lin DTS, Kobor MS, Zoller JA, Said JW, Morgello S, et al. HIV, pathology and epigenetic age acceleration in different human tissues. Geroscience, 2022, 44:1609–20., DOI 10.1007/s11357-022-00560-0 Nwanaji-Enwerem JC, Colicino E, Gao X, Wang C, Vokonas P, Boyer EW, et al. Associations of plasma folate and vitamin B6 with blood DNA methylation age: an analysis of one-carbon metabolites in the VA normative aging study. J Gerontol A Biol Sci Med Sci, 2021, 76:760–9., DOI 10.1093/gerona/glaa257 Kresovich JK, Martinez Lopez AM, Garval EL, Xu Z, White AJ, Sandler DP, et al. Alcohol consumption and methylation-based measures of biological age. J Gerontol A Biol Sci Med Sci, 2021, 76:2107–11., DOI 10.1093/gerona/glab149 Crimmins EM, Thyagarajan B, Levine ME, Weir DR, Faul J. Associations of age, sex, race/ethnicity, and education with 13 epigenetic clocks in a nationally representative U.S. Sample: the health and retirement study. J Gerontol A Biol Sci Med Sci, 2021, 76:1117–23., DOI 10.1093/gerona/glab016 Vetter VM, Kalies CH, Sommerer Y, Spira D, Drewelies J, Regitz-Zagrosek V, et al. Relationship between 5 epigenetic clocks, telomere length, and functional capacity assessed in older adults: cross-sectional and longitudinal analyses. J Gerontol A Biol Sci Med Sci, 2022, 77:1724–33., DOI 10.1093/gerona/glab381 McCrory C, Fiorito G, Hernandez B, Polidoro S, O’Halloran AM, Hever A, et al. GrimAge outperforms other epigenetic clocks in the prediction of age-related clinical phenotypes and all-cause mortality. J Gerontol A Biol Sci Med Sci, 2021, 76: 741–9., DOI 10.1093/gerona/glaa286 Van Remmen H, Freeman WM, Miller BF, Kinter M, Wren JD, Chiao A, et al. Oklahoma Nathan Shock Aging Center - assessing the basic biology of aging from genetics to protein and function. Geroscience, 2021, 43:2183–203., DOI 10. 1007/s11357-021-00454-7 Shadel GS, Adams PD, Berggren WT, Diedrich JK, Diffenderfer KE, Gage FH, et al. The San Diego Nathan Shock Center: tackling the heterogeneity of aging. Geroscience, 2021, 43:2139–48., DOI 10.1007/s11357-021-00426-x Salmon AB, Nelson JF, Gelfond JAL, Javors M, Ginsburg B, Lopez-Cruzan M, et al. San Antonio Nathan Shock Center: your one-stop shop for aging research. Geroscience, 2021, 43:2105–18., DOI 10.1007/s11357-021-00417-y Korstanje R, Peters LL, Robinson LL, Krasinski SD, Churchill GA. The jackson laboratory nathan shock center: impact of genetic diversity on aging. Geroscience, 2021, 43:2129–37., DOI 10.1007/s11357-021-00421-2 Kaeberlein M, Bitto A, Dunham MJ, Ladiges W, Lee SI, MacCoss MJ, et al. University of Washington Nathan Shock Center: innovation to advance aging research. Geroscience, 2021, 43:2161–5., DOI 10.1007/s11357-021- 00413-2 Curran SP, Lithgow GJ, Verdin E, Cohen P. University of Southern California and buck institute nathan shock center: multidimensional models of aging. Geroscience, 2021, 43:2119–27., DOI 10.1007/s11357-021-00416-z Cuervo AM, Huffman DM, Vijg J, Milman S, Singh R, Barzilai N. Einstein-Nathan shock center: translating the hallmarks of aging to extend human health span. Geroscience, 2021, 43:2167–82., DOI 10.1007/s11357-021- 00428-9 Austad SN, van derWillik O, Lederman S, Kerr C. The nathan shock centers. Geroscience, 2021, 43:2103–4., DOI 10.1007/s11357-021-00425-y Austad SN, Buford TW, Allison DB, Ballinger SW, Brown AW, Carter CS, et al. University of Alabama at birmingham nathan shock center: comparative energetics of aging. Geroscience, 2021, 43:2149–60., DOI 10.1007/s11357-021- 00414-1 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611623 EP 1611636 DI 10.3389/pore.2024.1611623 PG 14 ER PT J AU Sztankovics, D Moldvai, D Petovari, G Danko, T Szalai, F Miyaura, R Varga, V Nagy, N Papp, G Papay, J Krencz, I Sebestyen, A AF Sztankovics, Daniel Moldvai, Dorottya Petovari, Gabor Danko, Titanilla Szalai, Fatime Miyaura, Risa Varga, Viktoria Nagy, Noemi Papp, Gergo Papay, Judit Krencz, Ildiko Sebestyen, Anna TI mTOR hyperactivity and RICTOR amplification as targets for personalized treatments in malignancies SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE mTOR; mTORC2 hyperactivity; RICTOR amplification; Rictor overexpression; malignancies ID mTOR; mTORC2 hyperactivity; RICTOR amplification; Rictor overexpression; malignancies AB The increasing knowledge of molecular alterations in malignancies, including mutations and regulatory failures in the mTOR (mechanistic target of rapamycin) signaling pathway, highlights the importance of mTOR hyperactivity as a validated target in common and rare malignancies. This review summarises recent findings on the characterization and prognostic role of mTOR kinase complexes (mTORC1 and mTORC2) activity regarding differences in their function, structure, regulatory mechanisms, and inhibitor sensitivity. We have recently identified new tumor types with RICTOR (rapamycin-insensitive companion of mTOR) amplification and associated mTORC2 hyperactivity as useful potential targets for developing targeted therapies in lung cancer and other newly described malignancies. The activity of mTOR complexes is recommended to be assessed and considered in cancers before mTOR inhibitor therapy, as current firstgeneration mTOR inhibitors (rapamycin and analogs) can be ineffective in the presence of mTORC2 hyperactivity. We have introduced and proposed a marker panel to determine tissue characteristics of mTOR activity in biopsy specimens, patient materials, and cell lines. Ongoing phase trials of new inhibitors and combination therapies are promising in advanced-stage patients selected by genetic alterations, molecular markers, and/or protein expression changes in the mTOR signaling pathway. Hopefully, the summarized results, our findings, and the suggested characterization of mTOR activity will support therapeutic decisions. C1 [Sztankovics, Daniel] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Moldvai, Dorottya] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Petovari, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Danko, Titanilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Szalai, Fatime] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Miyaura, Risa] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Varga, Viktoria] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Nagy, Noemi] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Papp, Gergo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Papay, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Krencz, Ildiko] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Sebestyen, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM hsebanna@gmail.com CR Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell, 2017, 168:960–76., DOI 10.1016/j.cell.2017.02.004 Wullschleger S, Loewith R, Hall MN. TOR signaling in growth andmetabolism. Cell, 2006, 124:471–84., DOI 10.1016/j.cell.2006.01.016 Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cel Biol, 2020, 4:183–203., DOI 10.1038/s41580-019-0199-y Fu W, Hall MN. Regulation of mTORC2 signaling. Genes, Basel,, 2020, 11: 1045., DOI 10.3390/genes11091045 Sebestyen A, Danko T, Sztankovics D, Moldvai D, Raffay R, Cervi C, et al. The role of metabolic ecosystem in cancer progression - metabolic plasticity and mTOR hyperactivity in tumor tissues. Cancer Metastasis Rev, 2021, 40:989–1033., DOI 10. 1007/s10555-021-10006-2 Yamaguchi H, Kawazu M, Yasuda T, Soda M, Ueno T, Kojima S, et al. Transforming somatic mutations of mammalian target of rapamycin kinase in human cancer. Cancer Sci, 2015, 106:1687–92., DOI 10.1111/cas.12828 Grabiner BC, Nardi V, Birsoy K, Possemato R, Shen K, Sinha S, et al. A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity. Cancer Discov, 2014, 4:554–63., DOI 10.1158/2159-8290.CD-13-0929 Wagle N, Grabiner BC, Van Allen EM, Amin-Mansour A, Taylor-Weiner A, Rosenberg M, et al. Response and acquired resistance to everolimus in anaplastic thyroid cancer. N Engl J Med, 2014, 371:1426–33., DOI 10.1056/NEJMoa1403352 Menon S, Manning BD. Common corruption of the mTOR signaling network in human tumors. Oncogene, 2008, 27(Suppl. 2(0 2)):S43–51., DOI 10.1038/onc. 2009.352 Eng CP, Sehgal SN, Vezina C. Activity of rapamycin, AY-22,989, against transplanted tumors. J Antibiot, 1984, 37:1231–7., DOI 10.7164/antibiotics.37.1231 Choe G, Horvath S, Cloughesy TF, Crosby K, Seligson D, Palotie A, et al. Analysis of the phosphatidylinositol 3’-kinase signaling pathway in glioblastoma patients in vivo. Cancer Res, 2003, 63:2742–6. Zhou X, Tan M, Stone Hawthorne V, Klos KS, Lan KH, Yang Y, et al. Activation of the Akt/mammalian target of rapamycin/4E-BP1 pathway by ErbB2 overexpression predicts tumor progression in breast cancers. Clin Cancer Res, 2004, 10:6779–88., DOI 10.1158/1078-0432.CCR-04-0112 Nathan CO, Amirghahari N, Abreo F, Rong X, Caldito G, Jones ML, et al. Overexpressed eIF4E is functionally active in surgical margins of head and neck cancer patients via activation of the Akt/mammalian target of rapamycin pathway. Clin Cancer Res, 2004, 10:5820–7., DOI 10.1158/1078-0432.CCR-03- 0483 Sahin F, Kannangai R, Adegbola O, Wang J, Su G, Torbenson M. mTOR and P70 S6 kinase expression in primary liver neoplasms. Clin Cancer Res, 2004, 10: 8421–5., DOI 10.1158/1078-0432.CCR-04-0941 Balsara BR, Pei J, Mitsuuchi Y, Page R, Klein-Szanto A, Wang H, et al. Frequent activation of AKT in non-small cell lung carcinomas and preneoplastic bronchial lesions. Carcinogenesis, 2004, 25:2053–9., DOI 10.1093/carcin/bgh226 Xu G, Zhang W, Bertram P, Zheng XF, McLeod H. Pharmacogenomic profiling of the PI3K/PTEN-AKT-mTOR pathway in common human tumors. Int J Oncol, 2004, 24:893–900., DOI 10.3892/ijo.24.4.893 Nozawa H, Watanabe T, Nagawa H. Phosphorylation of ribosomal p70 S6 kinase and rapamycin sensitivity in human colorectal cancer. Cancer Lett, 2007, 251:105–13., DOI 10.1016/j.canlet.2006.11.008 Lin F, Zhang PL, Yang XJ, Prichard JW, Lun M, Brown RE. Morphoproteomic and molecular concomitants of an overexpressed and activated mTOR pathway in renal cell carcinomas. Ann Clin Lab Sci, 2006, 36:283–93. Kremer CL, Klein RR, Mendelson J, Browne W, Samadzedeh LK, Vanpatten K, et al. Expression of mTOR signaling pathway markers in prostate cancer progression. Prostate, 2006, 66:1203–12., DOI 10.1002/pros.20410 Liu Y, Hidayat S, Su WH, Deng X, Yu DH, Yu BZ. Expression and activity of mTOR and its substrates in different cell cycle phases and in oral squamous cell carcinomas of different malignant grade. Cell Biochem Funct, 2007, 25:45–53., DOI 10.1002/cbf.1332 Boone J, Ten Kate FJ, Offerhaus GJ, van Diest PJ, Rinkes IH, van Hillegersberg R. mTOR in squamous cell carcinoma of the oesophagus: a potential target for molecular therapy? J Clin Pathol, 2008, 61:909–13., DOI 10.1136/jcp.2008.055772 Karbowniczek M, Spittle CS, Morrison T, Wu H, Henske EP. mTOR is activated in the majority of malignant melanomas. J Invest Dermatol, 2008, 128: 980–7., DOI 10.1038/sj.jid.5701074 Pham NA, Schwock J, Iakovlev V, Pond G, Hedley DW, Tsao MS. Immunohistochemical analysis of changes in signaling pathway activation downstream of growth factor receptors in pancreatic duct cell carcinogenesis. BMC Cancer, 2008, 8:43., DOI 10.1186/1471-2407-8-43 Feng W, Duan X, Liu J, Xiao J, Brown RE. Morphoproteomic evidence of constitutively activated and overexpressed mTOR pathway in cervical squamous carcinoma and high grade squamous intraepithelial lesions. Int J Clin Exp Pathol, 2009, 2:249–60. Yu G, Wang J, Chen Y, Wang X, Pan J, Li G, et al. Overexpression of phosphorylated mammalian target of rapamycin predicts lymph node metastasis and prognosis of Chinese patients with gastric cancer. Clin Cancer Res, 2009, 15: 1821–9., DOI 10.1158/1078-0432.CCR-08-2138 Leseux L, Hamdi SM, Al Saati T, Capilla F, Recher C, Laurent G, et al. Sykdependent mTOR activation in follicular lymphoma cells. Blood, 2006, 108: 4156–62., DOI 10.1182/blood-2006-05-026203 Peponi E, Drakos E, Reyes G, Leventaki V, Rassidakis GZ, Medeiros LJ. Activation of mammalian target of rapamycin signaling promotes cell cycle progression and protects cells from apoptosis in mantle cell lymphoma. Am J Pathol, 2006, 169:2171–80., DOI 10.2353/ajpath.2006.051078 Follo MY, Mongiorgi S, Bosi C, Cappellini A, Finelli C, Chiarini F, et al. The Akt/mammalian target of rapamycin signal transduction pathway is activated in high-risk myelodysplastic syndromes and influences cell survival and proliferation. Cancer Res, 2007, 67:4287–94., DOI 10.1158/0008-5472.CAN-06-4409 Marzec M, Liu X, Kasprzycka M, Witkiewicz A, Raghunath PN, El-Salem M, et al. IL-2- and IL-15-induced activation of the rapamycin-sensitive mTORC1 pathway in malignant CD4+ T lymphocytes. Blood, 2008, 111: 2181–9., DOI 10.1182/blood-2007-06-095182 Zhao MY, Auerbach A, D’Costa AM, Rapoport AP, Burger AM, Sausville EA, et al. Phospho-p70S6K/p85S6K and cdc2/cdk1 are novel targets for diffuse large B-cell lymphoma combination therapy. Clin Cancer Res, 2009, 15:1708–20., DOI 10. 1158/1078-0432.CCR-08-1543 Mora J, Rodriguez E, de Torres C, Cardesa T, Rios J, Hernandez T, et al. Activated growth signaling pathway expression in Ewing sarcoma and clinical outcome. Pediatr Blood Cancer, 2012, 58:532–8., DOI 10.1002/pbc.23348 Izycka-Swieszewska E, Drozynska E, Rzepko R, Kobierska-Gulida G, Grajkowska W, Perek D, et al. Analysis of PI3K/AKT/mTOR signalling pathway in high risk neuroblastic tumours. Pol J Pathol, 2010, 61:192–8. Schmid K, Bago-Horvath Z, Berger W, Haitel A, Cejka D, Werzowa J, et al. Dual inhibition of EGFR and mTOR pathways in small cell lung cancer. Br J Cancer, 2010, 103:622–8., DOI 10.1038/sj.bjc.6605761 Liu J, Brown RE. Morphoproteomics demonstrates activation of mTOR pathway in anaplastic thyroid carcinoma: a preliminary observation. Ann Clin Lab Sci, 2010, 40:211–7. McCampbell AS, Harris HA, Crabtree JS, Winneker RC, Walker CL, Broaddus RR. Loss of inhibitory insulin receptor substrate-1 phosphorylation is an early event in mammalian target of rapamycin-dependent endometrial hyperplasia and carcinoma. Cancer Prev Res, Phila,, 2010, 3:290–300., DOI 10. 1158/1940-6207.CAPR-09-0199 Tickoo SK, Milowsky MI, Dhar N, Dudas ME, Gallagher DJ, Al-Ahmadie H, et al. Hypoxia-inducible factor and mammalian target of rapamycin pathway markers in urothelial carcinoma of the bladder: possible therapeutic implications. BJU Int, 2011, 107:844–9., DOI 10.1111/j.1464-410X.2010.09517.x Sapi Z, Fule T, Hajdu M, Matolcsy A, Moskovszky L, Mark A, et al. The activated targets of mTOR signaling pathway are characteristic for PDGFRA mutant and wild-type rather than KIT mutant GISTs. Diagn Mol Pathol, 2011, 20:22–33., DOI 10.1097/PDM.0b013e3181eb931b Sebestyen A, Sticz TB, Mark A, Hajdu M, Timar B, Nemes K, et al. Activity and complexes of mTOR in diffuse large B-cell lymphomas--a tissue microarray study. Mod Pathol, 2012, 25:1623–8., DOI 10.1038/modpathol.2012.141 Suzuki S, Dobashi Y, Minato H, Tajiri R, Yoshizaki T, Ooi A. EGFR and HER2-Akt-mTOR signaling pathways are activated in subgroups of salivary gland carcinomas. Virchows Arch, 2012, 461:271–82., DOI 10.1007/s00428-012-1282-3 Nemes K, Sebestyen A, Mark A, Hajdu M, Kenessey I, Sticz T, et al. Mammalian target of rapamycin, mTOR, activity dependent phospho-protein expression in childhood acute lymphoblastic leukemia, ALL). PLoS One, 2013, 8:e59335., DOI 10.1371/journal.pone.0059335 Leal P, Garcia P, Sandoval A, Letelier P, Brebi P, Ili C, et al. Immunohistochemical expression of phospho-mTOR is associated with poor prognosis in patients with gallbladder adenocarcinoma. Arch Pathol Lab Med, 2013, 137:552–7., DOI 10.5858/arpa.2012-0032-OA Mark A, Hajdu M, Varadi Z, Sticz TB, Nagy N, Csomor J, et al. Characteristic mTOR activity in Hodgkin-lymphomas offers a potential therapeutic target in high risk disease--a combined tissue microarray, in vitro and in vivo study. BMC Cancer, 2013, 13:250., DOI 10.1186/1471-2407-13-250 Pocza T, Sebestyen A, Turanyi E, Krenacs T, Mark A, Sticz TB, et al. mTOR pathway as a potential target in a subset of human medulloblastoma. Pathol Oncol Res, 2014, 20:893–900., DOI 10.1007/s12253-014-9771-0 Sticz T, MolnarA, MarkA, HajduM,NagyN,Vegso G, et al.mTOR activity and its prognostic significance in human colorectal carcinoma depending on C1 and C2 complexrelated protein expression.J ClinPathol, 2017, 70:410–6., DOI 10.1136/jclinpath-2016-203913 Krencz I, Sebestyen A, Fabian K, Mark A, Moldvay J, Khoor A, et al. Expression of mTORC1/2-related proteins in primary and brain metastatic lung adenocarcinoma. Hum Pathol, 2017, 62:66–73., DOI 10.1016/j.humpath.2016.12.012 Krencz I, Sebestyen A, Papay J, Lou Y, Lutz GF, Majewicz TL, et al. Correlation between immunohistochemistry and RICTOR fluorescence in situ hybridization amplification in small cell lung carcinoma. Hum Pathol, 2019, 93:74–80., DOI 10. 1016/j.humpath.2019.08.018 Krencz I, Sebestyen A, Papay J, Jeney A, Hujber Z, Burger CD, et al. In situ analysis of mTORC1/2 and cellular metabolism-related proteins in humanLymphangioleiomyomatosis. Hum Pathol, 2018, 79:199–207., DOI 10.1016/j.humpath.2018.05.018 Petovari G, Danko T, Tokes AM, Vetlenyi E, Krencz I, Raffay R, et al. In situ metabolic characterisation of breast cancer and its potential impact on therapy. Cancers, Basel,, 2020, 12:2492., DOI 10.3390/cancers12092492 Petovari G, Danko T, Krencz I, Hujber Z, Rajnai H, Vetlenyi E, et al. Inhibition of metabolic shift can decrease therapy resistance in human high-grade glioma cells. Pathol Oncol Res, 2020, 26:23–33., DOI 10.1007/s12253-019-00677-2 Felkai L, Krencz I, Kiss DJ, Nagy N, Petovari G, Danko T, et al. Characterization of mTOR activity and metabolic profile in pediatric rhabdomyosarcoma. Cancers, Basel,, 2020, 12:1947., DOI 10.3390/cancers12071947 Krencz I, Vetlenyi E, Danko T, Petovari G, Moldvai D, Sztankovics D, et al. Metabolic adaptation as potential target in papillary renal cell carcinomas based on their in situ metabolic characteristics. Int J Mol Sci, 2022, 23:10587., DOI 10.3390/ ijms231810587 Mohas A, Krencz I, Varadi Z, Arato G, Felkai L, Kiss DJ, et al. In situ analysis of mTORC1/C2 and metabolism-related proteins in pediatric osteosarcoma. Pathol Oncol Res, 2022, 28:1610231., DOI 10.3389/pore.2022.1610231 Hujber Z, Petovari G, Szoboszlai N, Danko T, Nagy N, Kriston C, et al. Rapamycin, mTORC1 inhibitor, reduces the production of lactate and 2- hydroxyglutarate oncometabolites in IDH1 mutant fibrosarcoma cells. J Exp Clin Cancer Res, 2017, 36:74., DOI 10.1186/s13046-017-0544-y Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature, 2006, 441:424–30., DOI 10.1038/nature04869 Gkountakos A, Pilotto S, Mafficini A, Vicentini C, Simbolo M, Milella M, et al. Unmasking the impact of Rictor in cancer: novel insights of mTORC2 complex. Carcinogenesis, 2018, 39:971–80., DOI 10.1093/carcin/bgy086 Krencz I, Sebestyen A, Khoor A. mTOR in lung neoplasms. Pathol Oncol Res, 2020, 26:35–48., DOI 10.1007/s12253-020-00796-1 Jiang WJ, Feng RX, Liu JT, Fan LL, Wang H, Sun GP. RICTOR expression in esophageal squamous cell carcinoma and its clinical significance. Med Oncol, 2017, 34:32., DOI 10.1007/s12032-017-0894-5 Wazir U, Newbold RF, Jiang WG, Sharma AK, Mokbel K. Prognostic and therapeutic implications of mTORC1 and Rictor expression in human breast cancer. Oncol Rep, 2013, 29:1969–74., DOI 10.3892/or.2013.2346 Driscoll DR, Karim SA, Sano M, Gay DM, Jacob W, Yu J, et al. mTORC2 signaling drives the development and progression of pancreatic cancer. Cancer Res, 2016, 76:6911–23., DOI 10.1158/0008-5472.CAN-16-0810 Joechle K, Guenzle J, Hellerbrand C, Strnad P, Cramer T, Neumann UP, et al. Role of mammalian target of rapamycin complex 2 in primary and secondary liver cancer. World J Gastrointest Oncol, 2021, 13:1632–47., DOI 10.4251/wjgo.v13.i11. 1632 Jebali A, Battistella M, Lebbe C, Dumaz N. RICTOR affects melanoma tumorigenesis and its resistance to targeted therapy. Biomedicines, 2021, 9:1498., DOI 10.3390/biomedicines9101498 Morrison Joly M, Hicks DJ, Jones B, Sanchez V, Estrada MV, Young C, et al. Rictor/mTORC2 drives progression and therapeutic resistance of HER2-amplified breast cancers. Cancer Res, 2016, 76:4752–64., DOI 10.1158/0008-5472.CAN-15- 3393 Morrison Joly M, Williams MM, Hicks DJ, Jones B, Sanchez V, Young CD, et al. Two distinct mTORC2-dependent pathways converge on Rac1 to drive breast cancer metastasis. Breast Cancer Res, 2017, 19:74., DOI 10.1186/s13058-017-0868-8 Werfel TA, Wang S, Jackson MA, Kavanaugh TE, Joly MM, Lee LH, et al. Selective mTORC2 inhibitor therapeutically blocks breast cancer cell growth and survival. Cancer Res, 2018, 78:1845–58., DOI 10.1158/0008-5472.CAN-17-2388 Zhang F, Zhang X, Li M, Chen P, Zhang B, Guo H, et al. mTOR complex component Rictor interacts with PKCzeta and regulates cancer cell metastasis. Cancer Res, 2010, 70:9360–70., DOI 10.1158/0008-5472.CAN-10-0207 Liu Y, Lu Y, Li A, Celiku O, Han S, Qian M, et al. mTORC2/Rac1 pathway predisposes cancer aggressiveness in IDH1-mutated glioma. Cancers, Basel,, 2020, 12:787., DOI 10.3390/cancers12040787 Alvarenga AW, Machado LE, Rodrigues BR, Lupinacci FC, Sanemastu P, Matta E, et al. Evaluation of Akt and RICTOR expression levels in astrocytomas of all grades. J Histochem Cytochem, 2017, 65:93–103., DOI 10.1369/ 0022155416675850 Petovari G, Hujber Z, Krencz I, Danko T, Nagy N, Toth F, et al. Targeting cellular metabolism using rapamycin and/or doxycycline enhances anti-tumour effects in human glioma cells. Cancer Cel Int, 2018, 18:211., DOI 10.1186/s12935- 018-0710-0 Akgul S, Li Y, Zheng S, Kool M, Treisman DM, Li C, et al. Opposing tumorpromoting and -suppressive functions of rictor/mTORC2 signaling in adult glioma and pediatric SHH medulloblastoma. Cell Rep, 2018, 24:463–78., DOI 10.1016/j. celrep.2018.06.050 Gulhati P, Bowen KA, Liu J, Stevens PD, Rychahou PG, Chen M, et al. mTORC1 and mTORC2 regulate EMT, motility, andmetastasis of colorectal cancer via RhoA and Rac1 signaling pathways. Cancer Res, 2011, 71:3246–56., DOI 10.1158/ 0008-5472.CAN-10-4058 Wen FF, Li XY, Li YY, He S, Xu XY, Liu YH, et al. Expression of Raptor and Rictor and their relationships with angiogenesis in colorectal cancer. Neoplasma, 2020, 67:501–8., DOI 10.4149/neo_2020_190705N597 Wang L, Qi J, Yu J, Chen H, Zou Z, Lin X, et al. Overexpression of Rictor protein in colorectal cancer is correlated with tumor progression and prognosis. Oncol Lett, 2017, 14:6198–202., DOI 10.3892/ol.2017.6936 Bang H, Ahn S, Ji Kim E, Kim ST, Park HY, Lee J, et al. Correlation between RICTOR overexpression and amplification in advanced solid tumors. Pathol Res Pract, 2020, 216:152734., DOI 10.1016/j.prp.2019.152734 Kim ST, Kim SY, Klempner SJ, Yoon J, Kim N, Ahn S, et al. Rapamycininsensitive companion of mTOR, RICTOR, amplification defines a subset of advanced gastric cancer and is sensitive to AZD2014-mediated mTORC1/ 2 inhibition. Ann Oncol, 2017, 28:547–54., DOI 10.1093/annonc/mdw669 Cao RZ, Min L, Liu S, Tian RY, Jiang HY, Liu J, et al. Rictor activates cav 1 through the Akt signaling pathway to inhibit the apoptosis of gastric cancer cells. Front Oncol, 2021, 11:641453., DOI 10.3389/fonc.2021.641453 Kaibori M, Shikata N, Sakaguchi T, Ishizaki M, Matsui K, Iida H, et al. Influence of rictor and raptor expression of mTOR signaling on long-term outcomes of patients with hepatocellular carcinoma. Dig Dis Sci, 2015, 60: 919–28., DOI 10.1007/s10620-014-3417-7 Villanueva A, Chiang DY, Newell P, Peix J, Thung S, Alsinet C, et al. Pivotal role of mTOR signaling in hepatocellular carcinoma. Gastroenterology, 2008, 135: 1972–83., DOI 10.1053/j.gastro.2008.08.008 Schmidt KM, Hellerbrand C, Ruemmele P, Michalski CW, Kong B, Kroemer A, et al. Inhibition of mTORC2 component RICTOR impairs tumor growth in pancreatic cancer models. Oncotarget, 2017, 8:24491–505., DOI 10.18632/ oncotarget.15524 Wen SY, Li CH, Zhang YL, Bian YH, Ma L, Ge QL, et al. Rictor is an independent prognostic factor for endometrial carcinoma. Int J Clin Exp Pathol, 2014, 7:2068–78. Naruse T, Yanamoto S, Okuyama K, Yamashita K, Omori K, Nakao Y, et al. Therapeutic implication of mTORC2 in oral squamous cell carcinoma. Oral Oncol, 2017, 65:23–32., DOI 10.1016/j.oraloncology.2016.12.012 Ruicci KM, Plantinga P, Pinto N, Khan MI, Stecho W, Dhaliwal SS, et al. Disruption of the RICTOR/mTORC2 complex enhances the response of head and neck squamous cell carcinoma cells to PI3K inhibition. Mol Oncol, 2019, 13: 2160–77., DOI 10.1002/1878-0261.12558 Kawasaki G, Naruse T, Furukawa K, Umeda M. mTORC1 and mTORC2 expression levels in oral squamous cell carcinoma: an immunohistochemical and clinicopathological study. Anticancer Res, 2018, 38: 1623–8., DOI 10.21873/anticanres.12393 Kondo S, Hirakawa H, Ikegami T, Uehara T, Agena S, Uezato J, et al. Raptor and rictor expression in patients with human papillomavirus-related oropharyngeal squamous cell carcinoma. BMC Cancer, 2021, 21:87., DOI 10.1186/s12885-021- 07794-9 Zhang X, Wang X, Xu T, Zhong S, Shen Z. Targeting of mTORC2 may have advantages over selective targeting of mTORC1 in the treatment of malignant pheochromocytoma. Tumour Biol, 2015, 36:5273–81., DOI 10.1007/s13277-015- 3187-7 Schmidt KM, Dietrich P, Hackl C, Guenzle J, Bronsert P, Wagner C, et al. Inhibition of mTORC2/RICTOR impairs melanoma hepatic metastasis. Neoplasia, 2018, 20:1198–208., DOI 10.1016/j.neo.2018.10.001 Gibault L, Ferreira C, Perot G, Audebourg A, Chibon F, Bonnin S, et al. From PTEN loss of expression to RICTOR role in smooth muscle differentiation: complex involvement of the mTOR pathway in leiomyosarcomas and pleomorphic sarcomas. Mod Pathol, 2012, 25:197–211., DOI 10.1038/modpathol.2011.163 Okada T, Lee AY, Qin LX, Agaram N, Mimae T, Shen Y, et al. Integrin-α10 dependency identifies RAC and RICTOR as therapeutic targets in high-grade myxofibrosarcoma. Cancer Discov, 2016, 6:1148–65., DOI 10.1158/2159-8290.CD- 15-1481 Ross JS, Wang K, Elkadi OR, Tarasen A, Foulke L, Sheehan CE, et al. Nextgeneration sequencing reveals frequent consistent genomic alterations in small cell undifferentiated lung cancer. J Clin Pathol, 2014, 67:772–6., DOI 10.1136/jclinpath- 2014-202447 Umemura S, Mimaki S, Makinoshima H, Tada S, Ishii G, Ohmatsu H, et al. Therapeutic priority of the PI3K/AKT/mTOR pathway in small cell lung cancers as revealed by a comprehensive genomic analysis. J Thorac Oncol, 2014, 9:1324–31., DOI 10.1097/JTO.0000000000000250 Sakre N, Wildey G, Behtaj M, Kresak A, Yang M, Fu P, et al. RICTOR amplification identifies a subgroup in small cell lung cancer and predicts response to drugs targeting mTOR. Oncotarget, 2017, 8:5992–6002., DOI 10.18632/oncotarget. 13362 Zeng Z, Sarbassov dos D, Samudio IJ, Yee KW, Munsell MF, Ellen Jackson C, et al. Rapamycin derivatives reduce mTORC2 signaling and inhibit AKT activation in AML. Blood, 2007, 109:3509–12., DOI 10.1182/blood-2006-06-030833 Zhuang J, Hawkins SF, Glenn MA, Lin K, Johnson GG, Carter A, et al. Akt is activated in chronic lymphocytic leukemia cells and delivers a pro-survival signal: the therapeutic potential of Akt inhibition. Haematologica, 2010, 95:110–8., DOI 10. 3324/haematol.2009.010272 Simioni C, Martelli AM, Zauli G, Melloni E, Neri LM. Targeting mTOR in acute lymphoblastic leukemia. Cells, 2019, 8:190., DOI 10.3390/cells8020190 Muller A, Zang C, Chumduri C, Dorken B, Daniel PT, Scholz CW. Concurrent inhibition of PI3K and mTORC1/mTORC2 overcomes resistance to rapamycin induced apoptosis by down-regulation of Mcl-1 in mantle cell lymphoma. Int J Cancer, 2013, 133:1813–24., DOI 10.1002/ijc.28206 Sahu D, Huan J, Wang H, Sahoo D, Casteel DE, Klemke RL, et al. Bladder cancer invasion is mediated by mammalian target of rapamycin complex 2-driven regulation of nitric oxide and invadopodia formation. Am J Pathol, 2021, 191: 2203–18., DOI 10.1016/j.ajpath.2021.08.002 Juengel E, Kim D, Makarevic J, Reiter M, Tsaur I, Bartsch G, et al. Molecular analysis of sunitinib resistant renal cell carcinoma cells after sequential treatment with RAD001, everolimus, or sorafenib. J Cel Mol Med, 2015, 19:430–41., DOI 10. 1111/jcmm.12471 Sztankovics D, Krencz I, Moldvai D, Danko T, Nagy A, Nagy N, et al. Novel RICTOR amplification harbouring entities: FISH validation of RICTOR amplification in tumour tissue after next-generation sequencing. Sci Rep, 2023, 13:19610., DOI 10.1038/s41598-023-46927-x Tian T, Li X, Zhang J. mTOR signaling in cancer andmTOR inhibitors in solid tumor targeting therapy. Int J Mol Sci, 2019, 20:755., DOI 10.3390/ijms20030755 Dobashi Y, Watanabe Y, Miwa C, Suzuki S, Koyama S. Mammalian target of rapamycin: a central node of complex signaling cascades. Int J Clin Exp Pathol, 2011, 4:476–95. Chen B, Tan Z, Gao J, Wu W, Liu L, Jin W, et al. Hyperphosphorylation of ribosomal protein S6 predicts unfavorable clinical survival in non-small cell lung cancer. J Exp Clin Cancer Res, 2015, 34:126., DOI 10.1186/s13046-015-0239-1 Ekman S, Wynes MW, Hirsch FR. The mTOR pathway in lung cancer and implications for therapy and biomarker analysis. J Thorac Oncol, 2012, 7:947–53., DOI 10.1097/JTO.0b013e31825581bd Spoerke JM, O’Brien C, Huw L, Koeppen H, Fridlyand J, Brachmann RK, et al. Phosphoinositide 3-kinase, PI3K, pathway alterations are associated with histologic subtypes and are predictive of sensitivity to PI3K inhibitors in lung cancer preclinical models. Clin Cancer Res, 2012, 18:6771–83., DOI 10.1158/1078-0432.CCR-12-2347 Szalai F, Sztankovics D, Krencz I, Moldvai D, Papay J, Sebestyen A, et al. Rictor—amediator of progression and metastasis in lung cancer. Cancers, 2024, 16: 543., DOI 10.3390/cancers16030543 Conde E, Angulo B, TangM,Morente M, Torres-Lanzas J, Lopez-Encuentra A, et al. Molecular context of the EGFR mutations: evidence for the activation of mTOR/S6K signaling. Clin Cancer Res, 2006, 12:710–7., DOI 10.1158/1078-0432. CCR-05-1362 Hendifar AE, Marchevsky AM, Tuli R. Neuroendocrine tumors of the lung: current challenges and advances in the diagnosis and management of welldifferentiated disease. J Thorac Oncol, 2017, 12:425–36., DOI 10.1016/j.jtho.2016. 11.2222 Krencz I, Sztankovics D, Danko T, Sebestyen A, Khoor A. Progression and metastasis of small cell lung carcinoma: the role of the PI3K/Akt/mTOR pathway and metabolic alterations. Cancer Metastasis Rev, 2021, 40:1141–57., DOI 10.1007/ s10555-021-10012-4 Glasgow CG, Steagall WK, Taveira-Dasilva A, Pacheco-Rodriguez G, Cai X, El-Chemaly S, et al. Lymphangioleiomyomatosis, LAM): molecular insights lead to targeted therapies. Respir Med, 2010, 104:S45–S58., DOI 10.1016/j.rmed.2010.03.017 Henske EP, McCormack FX. Lymphangioleiomyomatosis - a wolf in sheep’s clothing. J Clin Invest, 2012, 122:3807–16., DOI 10.1172/JCI58709 Adachi K, Miki Y, Saito R, Hata S, Yamauchi M, Mikami Y, et al. Intracrine steroid production and mammalian target of rapamycin pathways in pulmonary lymphangioleiomyomatosis. Hum Pathol, 2015, 46:1685–93., DOI 10.1016/j. humpath.2015.02.019 Hayashi T, Kumasaka T, Mitani K, Okada Y, Kondo T, Date H, et al. Bronchial involvement in advanced stage lymphangioleiomyomatosis: histopathologic and molecular analyses. Hum Pathol, 2016, 50:34–42., DOI 10. 1016/j.humpath.2015.11.002 Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science, 2005, 307:1098–101., DOI 10.1126/science.1106148 Janku F, Yap TA, Meric-Bernstam F. Targeting the PI3K pathway in cancer: are we making headway? Nat Rev Clin Oncol, 2018, 15:273–91., DOI 10.1038/ nrclinonc.2018.28 Owonikoko TK, Khuri FR. Targeting the PI3K/AKT/mTOR pathway: biomarkers of success and tribulation. Am Soc Clin Oncol Educ Book, 2013, 10: e395–401., DOI 10.14694/EdBook_AM.2013.33.e395 Hobby G, Clark R, Woywodt A. A treasure from a barren island: the discovery of rapamycin. Clin Kidney J, 2022, 15:1971–2., DOI 10.1093/ckj/sfac116 Sehgal SN, Baker H, Vezina C. Rapamycin, AY-22,989), a new antifungal antibiotic. II. Fermentation, isolation and characterization. J Antibiot, 1975, 28: 727–32., DOI 10.7164/antibiotics.28.727 Mao B, Zhang Q, Ma L, Zhao DS, Zhao P, Yan P. Overview of research into mTOR inhibitors. Molecules, 2022, 27:5295., DOI 10.3390/molecules27165295 Guenzle J, Akasaka H, Joechle K, Reichardt W, Venkatasamy A, Hoeppner J, et al. Pharmacological inhibition of mTORC2 reduces migration and metastasis in melanoma. Int J Mol Sci, 2020, 22:30., DOI 10. 3390/ijms22010030 Conciatori F, Ciuffreda L, Bazzichetto C, Falcone I, Pilotto S, Bria E, et al. mTOR cross-talk in cancer and potential for combination therapy. Cancers, Basel,, 2018, 10:23., DOI 10.3390/cancers10010023 Ali ES, Mitra K, Akter S, Ramproshad S, Mondal B, Khan IN, et al. Recent advances and limitations of mTOR inhibitors in the treatment of cancer. Cancer Cel Int, 2022, 22:284., DOI 10.1186/s12935-022-02706-8 Park S, Shim J, Mortimer PGS, Smith SA, Godin RE, Hollingsworth SJ, et al. Biomarker-driven phase 2 umbrella trial study for patients with recurrent small cell lung cancer failing platinum-based chemotherapy. Cancer, 2020, 126:4002–12., DOI 10.1002/cncr.33048 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611643 EP 1611661 DI 10.3389/pore.2024.1611643 PG 19 ER PT J AU Molnar, A Palfi, E Belak, B Blasszauer, C Reibl, D Lovey, J AF Molnar, Andrea Palfi, Erzsebet Belak, Barbara Blasszauer, Celia Reibl, Daniel Lovey, Jozsef TI Positive correlation between persistence of medical nutrition therapy and overall survival in patients with head and neck cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE medical nutrition therapy; survival; real -word evidence; persistence of nutrition and survival; head and neck cancer ID medical nutrition therapy; survival; real -word evidence; persistence of nutrition and survival; head and neck cancer AB Background: Several factors can affect overall survival of head and neck cancer (HNC) patients, including characteristics of the cancer disease and response to treatments. However, patients’ nutritional status and the effectiveness of medical nutrition therapy (MNT) can also impact overall survival. The primary goal of our research was to collect real-life data on the use of MNT in HNC patients and to specifically investigate the correlation between survival and the duration of uninterrupted (persistent) nutrition. Method: The data of this retrospective, analytical, cohort study was collected from electronic healthcare records from the Hungarian National Health Insurance Fund Management. Overall, 38,675 HNC patients’ data of the period between 2012 and 2021 was used. We applied multi-step exclusions to identify patient groups accurately and to avoid biasing factors. Statistical analysis was done by the Kaplan-Meier method, log-rank test, and Cox regression analysis. Results: Throughout the investigated period 16,871 (64%) patients received MNT therapy out of 26,253 newly diagnosed patients (≥18 years). In terms of the persistence of MNT, we divided the patients into three groups (1–3; 4–6; ≥7- month duration of MNT). When comparing these groups, we found that patients receiving long-term (≥7 months) MNT had a significantly longer overall survival (p < 0.0001) than those who received MNT for a shorter duration, both in locally advanced and recurrent/metastatic cases. Conclusion: The main outcome of the study is that there is a positive correlation between the persistence of MNT and the overall survival in HNC patients when nutritional intervention lasts several months. It highlights the responsibility of the specialists during the patient journey to use MNT early and to continue its use for as long as it is beneficial to the patients. C1 [Molnar, Andrea] Semmelweis University, Doctoral School, Health Sciences DivisionBudapest, Hungary. [Palfi, Erzsebet] Semmelweis University, Doctoral School, Health Sciences DivisionBudapest, Hungary. [Belak, Barbara] Semmelweis University, Doctoral School, Health Sciences DivisionBudapest, Hungary. [Blasszauer, Celia] MedicalScan Kft.Budapest, Hungary. [Reibl, Daniel] MedicalScan Kft.Budapest, Hungary. [Lovey, Jozsef] Orszagos Onkologiai Intezet, Nemzeti Tumorbiologiai LaboratoriumBudapest, Hungary. RP Molnar, A (reprint author), Semmelweis University, Doctoral School, Health Sciences Division, Budapest, Hungary. EM dr.molnarandrea.rd@gmail.com CR Arends J, Strasser F, Gonella S, Solheim TS, Madeddu C, Ravasco P, et al. Cancer cachexia in adult patients: ESMO clinical practice guidelines. ESMO Open, 2021, 6(3):100092., DOI 10.1016/j.esmoop.2021.100092 MuscaritoliM, Arends J, Bachmann P, Baracos V, Barthelemy N, Bertz H, et al. ESPEN practical guideline: clinical Nutrition in cancer. Clin Nutr, 2021, 40(5): 2898–913., DOI 10.1016/j.clnu.2021.02.005 Seo Y, Eo W, Kim S, Shim B, Lee S. Can nutritional status predict overall survival in patients with advanced non-small cell lung cancer?. Nutr Cancer, 2019, 71(7):1108–17., DOI 10.1080/01635581.2019.1598564 Yu Y, Wu H, Qiu J, Ke D,Wu Y, Lin M, et al. A nutrition-related factor-based risk stratification for exploring the clinical benefits in the treatment of patients with locally advanced esophageal squamous cell carcinoma receiving definitive chemoradiotherapy: a retrospective cohort study. Front Nutr, 2022, 9:896847., DOI 10.3389/fnut.2022.896847 Migita K, Matsumoto S,Wakatsuki K, Ito M, Kunishige T, Nakade H, et al. A decrease in the prognostic nutritional index is associated with a worse longterm outcome in gastric cancer patients undergoing neoadjuvant chemotherapy. Surg Today, 2017, 47(8):1018–26., DOI 10.1007/s00595-017- 1469-y Mardas M, Madry R, Stelmach-Mardas M. Dietary intake variability in the cycle of cytotoxic chemotherapy. Support Care Cancer, 2016, 24(6):2619–25., DOI 10.1007/s00520-015-3072-3 Castillo-Martinez L, Castro-Eguiluz D, Copca-Mendoza ET, Perez-Camargo DA, Reyes-Torres CA, Avila EA, et al. Nutritional assessment tools for the identification of malnutrition and nutritional risk associated with cancer treatment. Rev Invest Clin, 2018, 70(3):121–5., DOI 10.24875/RIC.18002524 Koyama S, Tabuchi T, Okawa S, Taniyama Y, Nakata K, Morishima T, et al. Hospital volume and 5-year survival in head and neck cancer patients in Osaka, Japan. Jpn J Clin Oncol, 2021, 51(10):1515–22., DOI 10.1093/jjco/hyab132 Du E, Mazul AL, Farquhar D, Brennan P, Anantharaman D, Abedi-Ardekani B, et al. Long-term survival in head and neck cancer: impact of site, stage, smoking, and human papillomavirus status. Laryngoscope, 2019, 129(11):2506–13., DOI 10. 1002/lary.27807 Guo K, Xiao W, Chen X, Zhao Z, Lin Y, Chen G. Epidemiological trends of head and neck cancer: a population-based study. Biomed Res Int, 2021, 2021: 1738932., DOI 10.1155/2021/1738932 Golusinski P, Corry J, Poorten VV, Simo R, Sjogren E, Makitie A, et al. Deescalation studies in HPV-positive oropharyngeal cancer: how should we proceed? Oral Oncol, 2021, 123:105620., DOI 10.1016/j.oraloncology.2021.105620 Migliaro M, Massuh D, Infante MF, Brahm AM, San Martin MT, Ortuno D. Role of epstein-barr virus and human papilloma virus in the development of oropharyngeal cancer: a literature review. Int J Dent, 2022, 2022:3191569., DOI 10. 1155/2022/3191569 Alterio D, Marvaso G, Ferrari A, Volpe S, Orecchia R, Jereczek-Fossa BA. Modern radiotherapy for head and neck cancer. Semin Oncol, 2019, 46(3):233–45., DOI 10.1053/j.seminoncol.2019.07.002 ESMO pocket gudeline. Head and neck cancer, 2021). Available from: https:// interactiveguidelines.esmo.org/esmo-web-app/toc/index.php?subjectAreaId= 11&loadPdf=1, Accessed December 26, 2023). Bossi P, Delrio P, Mascheroni A, Zanetti M. The spectrum of malnutrition/ cachexia/sarcopenia in oncology according to different cancer types and settings: a narrative review. Nutrients, 2021, 13(6):1980., DOI 10.3390/nu13061980 Cawood AL, Burden ST, Smith T, Stratton RJ. A systematic review and metaanalysis of the effects of community use of oral nutritional supplements on clinical outcomes. Ageing Res Rev, 2023, 88:101953., DOI 10.1016/j.arr.2023.101953 Molnar A, Csontos AA, Dako S, Hencz R, Anton DA, Palfi E, et al. Investigating the efficacy of nutrition therapy for outpatients with inflammatory bowel disease. Orv Hetil, 2017, 158(19):731–9., DOI 10.1556/650.2017.30719 Lovey J, Molnar A, Banky B. Long-term nutrition in patients candidate to neoadjuvant and adjuvant treatments. Eur J Surg Oncol, 2023):106850., DOI 10.1016/ j.ejso.2023.02.007 Saesen R, Kantidakis G, Marinus A, Lacombe D, Huys I. How do cancer clinicians perceive real-world data and the evidence derived therefrom? Findings from an international survey of the European Organisation for Research and Treatment of Cancer. Front Pharmacol, 2022, 13:969778., DOI 10.3389/fphar. 2022.969778 Caccialanza R, Goldwasser F, Marschal O, Ottery F, Schiefke I, Tilleul P, et al. Unmet needs in clinical nutrition in oncology: amultinational analysis of real-world evidence. Ther Adv Med Oncol, 2020, 12:1758835919899852., DOI 10.1177/ 1758835919899852 Martin L, Senesse P, Gioulbasanis I, Antoun S, Bozzetti F, Deans C, et al. Diagnostic criteria for the classification of cancer-associated weight loss. J Clin Oncol, 2015, 33(1):90–9., DOI 10.1200/JCO.2014.56.1894 Bischoff SC, Austin P, Boeykens K, Chourdakis M, Cuerda C, Jonkers- Schuitema C, et al. ESPEN practical guideline: home enteral nutrition. Clin Nutr, 2022, 41(2):468–88., DOI 10.1016/j.clnu.2021.10.018 Pironi L, Boeykens K, Bozzetti F, Joly F, Klek S, Lal S, et al. ESPEN practical guideline: home parenteral nutrition. Clin Nutr, 2023, 42(3):411–30., DOI 10.1016/j. clnu.2022.12.003 Galffy G, Molnar A, Blasszauer C, Komka I, Reibl D, Lovey J. Az alacsony testtomegindex es testtomeg csokkenes vizsgalata magyar tudodaganatos betegek adatainak felhasznalasava. Magy Onkol, 2022, 66(1):55–63. de Oliveira Faria S, Alvim Moravia R, Howell D, Eluf Neto J. Adherence to nutritional interventions in head and neck cancer patients: a systematic scoping review of the literature. J Hum Nutr Diet, 2021, 34(3):562–71., DOI 10.1111/jhn. 12848 Muller-Richter U, Betz C, Hartmann S, Brands RC. Nutritionmanagement for head and neck cancer patients improves clinical outcome and survival. Nutr Res, 2017, 48:1–8., DOI 10.1016/j.nutres.2017.08.007 Wunderle C, Gomes F, Schuetz P, Stumpf F, Austin P, Ballesteros-Pomar MD, et al. ESPEN practical guideline: nutritional support for polymorbid medical inpatients. Clin Nutr, 2024, 43(3):674–91., DOI 10.1016/j.clnu.2024.01.008 Burgos R, Breton I, Cereda E, Desport JC, Dziewas R, Genton L, et al. ESPEN guideline clinical nutrition in neurology. Clin Nutr, 2018, 37(1):354–96., DOI 10. 1016/j.clnu.2017.09.003 Cantwell LA, Fahy E, Walters ER, Patterson JM. Nutritional prehabilitation in head and neck cancer: a systematic review. Support Care Cancer, 2022, 30(11): 8831–43., DOI 10.1007/s00520-022-07239-4 Loewen I, Jeffery CC, Rieger J, Constantinescu G. Prehabilitation in head and neck cancer patients: a literature review. J Otolaryngol Head Neck Surg, 2021, 50(1): 2., DOI 10.1186/s40463-020-00486-7 Brady R, McSharry L, Lawson S, Regan J. The impact of dysphagia prehabilitation on swallowing outcomes post-chemoradiation therapy in head and neck cancer: a systematic review. Eur J Cancer Care, Engl,, 2022, 31(3): e13549., DOI 10.1111/ecc.13549 Furka A. Oncological prehabilitation. Orv Hetil, 2022, 163(50):1975–81., DOI 10.1556/650.2022.32646 Almada-Correia I, Neves PM, Makitie A, Ravasco P. Body composition evaluation in head and neck cancer patients: a review. Front Oncol, 2019, 9:1112., DOI 10.3389/fonc.2019.01112 Silver HJ, Dietrich MS, Murphy BA. Changes in body mass, energy balance, physical function, and inflammatory state in patients with locally advanced head and neck cancer treated with concurrent chemoradiation after low-dose induction chemotherapy. Head Neck, 2007, 29(10):893–900., DOI 10.1002/hed.20607 Belak B, Molnar A, Palfi E, Blasszauer C, Reibl D, Lovey J. Effect of long-term medical nutrition therapy on the survival of head and neck cancer patients - based on real-world data. Magy Onkol, 2023, 67(4):341–4. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611664 EP 1611673 DI 10.3389/pore.2024.1611664 PG 10 ER PT J AU Timar, J Ladanyi, A Sebestyen, A Kopper, L AF Timar, Jozsef Ladanyi, Andrea Sebestyen, Anna Kopper, Laszlo TI Editorial: Pathology and Oncology Research: addressing publication ethics issues SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Editorial DE peer review; publication ethics; scientometry; quality control; transparency ID peer review; publication ethics; scientometry; quality control; transparency C1 [Timar, Jozsef] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Ladanyi, Andrea] National Institute of OncologyBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kopper, Laszlo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM jtimar@gmail.com CR Kopper L, Timar J. 25th anniversary of Pathology Oncology Research. Pathol Oncol Res, 2020, 26:1., DOI 10.1007/s12253-020-00795-2 SCImago. SJR — SCImago journal and country rank, 2023). Available from: http://www.scimagojr.com, Accessed May 1, 2023). Journal Impact Factor. Journal citation reports. United Kingdom: Clarivate, 2023). COPE Council. COPE discussion document. Americas: Predatory Publishing, 2019)., DOI 10.24318/cope.2019.3.6 Kulka J, Cserni G. Editorial: guidelines from the central-eastern European professional consensus statement on breast cancer. Pathol Oncol Res, 2022, 28: 1610587., DOI 10.3389/pore.2022.1610587 Guidelines from the Central-Eastern European Professional Consensus Statement on Breast Cancer. Special issue eBook. Lausanne: Frontiers, 2022). ISBN 978-2-88976-944-5., DOI 10.3389/978-2-88976-944-5 Ryska A. Editorial: liquid biopsy—a great hope or just hype? Pathol Oncol Res, 2023, 29:1611170., DOI 10.3389/pore.2023.1611170 Liquid Biopsy - A Great Hope or Just Hype?. Special issue eBook. Lausanne: Frontiers, 2023). ISBN 978-2-83252-182-3., DOI 10.3389/978-2-83252-182-3 Pathology and Oncology Research – Editors’ Picks from2021. Special issue eBook. Lausanne: Frontiers, 2022). ISBN978-2-88974-770-2., DOI 10.3389/978-2-88974-770-2 Pathology and Oncology Research – Editors’ Picks from 2022. Special issue eBook. Lausanne: Frontiers, 2023). ISBN 978-2-83251-901-1., DOI 10.3389/978-2-83251-901-1 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611691 EP 1611692 DI 10.3389/pore.2024.1611691 PG 2 ER PT J AU Liu, Y Chen, Z Wang, L Li, B AF Liu, Yuqing Chen, Zhenwei Wang, Lu Li, Baizhou TI Intestinal Langerhans cell histiocytosis presenting with symptoms similar to inflammatory bowel disease: a case report SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Langerhans cell histiocytosis; histiocytosis; intestine; infant; case report ID Langerhans cell histiocytosis; histiocytosis; intestine; infant; case report AB Background: Langerhans cell histiocytosis is a rare disease characterized by the abnormal proliferation of Langerhans cells within a single organ or multiple organs. This case report aims to improve the knowledge of the presentation of gastrointestinal Langerhans cell histiocytosis to facilitate the diagnosis and management of this rare disorder. Case presentation: A 19-month-old female presented with repeatedly mucinous bloody stools. The abdominal ultrasound revealed a slightly enlarged spleen. The initial colonoscopy revealed chronic enteritis with a very early onset inflammatory bowel disease. After anti-inflammatory treatment without improvement, an intestinal biopsy was performed at The Forth Affiliated Hospital of Zhejiang University. The final intestinal biopsy and histopathology examination confirmed the presence of Langerhans cell histiocytosis. After diagnosis, additional lung and head imaging examinations revealed no abnormalities. Her condition improved gradually after being treated with chemotherapy (vincristine and prednisone) and molecular-targeted drug(dalafinil) treatment. Conclusion: The clinical symptoms of Langerhans cell histiocytosis involving the gastrointestinal tract are not specific and may resemble symptoms observed in inflammatory bowel disease and other primary gastrointestinal tumors. Therefore, in cases of infants presenting with inflammatory gastrointestinal symptoms that do not resolve after treatment, a biopsy is essential to obtain a differential diagnosis. C1 [Liu, Yuqing] Zhejiang University, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Department of PathologyYiwu, Zhejiang, China. [Chen, Zhenwei] Zhejiang University, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Department of PathologyYiwu, Zhejiang, China. [Wang, Lu] Zhejiang University, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Department of PathologyYiwu, Zhejiang, China. [Li, Baizhou] Zhejiang University, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Department of PathologyYiwu, Zhejiang, China. RP Li, B (reprint author), Zhejiang University, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Department of Pathology, Yiwu, China. EM libaizhou@zju.edu.cn CR Allen CE, Merad M, McClain KL. Langerhans-cell histiocytosis. N Engl J Med, 2018, 379(9):856–68., DOI 10.1056/NEJMra1607548 Matsubara Y, Kobayashi M, Hijikata Y, Ota Y, Hirata Y, Lim LA, et al. Gastrointestinal lesion in adult-onset Langerhans cell histiocytosis. Int J Clin Oncol, 2020, 25(11):1945–50., DOI 10.1007/s10147-020-01739-1 Wang L, Yang F, Ding Y, Lu L, Li H, Cui Y, et al. Gastrointestinal Langerhans cell histiocytosis with unifocal, single-system involvement in adults: cases report and literature review. J Clin Lab Anal, 2022, 36(12):e24765., DOI 10.1002/jcla.24765 Wang H, Wang Y, Wang R, Li X. Case report: two infant cases of langerhans cell histiocytosis involving the digestive tract. Front Pediatr, 2021, 24(9):545771., DOI 10.3389/fped.2021.545771 Zei M, Meyers AB, Boyd KP, Larson-Nath C, Suchi M. Langerhans cell histiocytosis of the digestive tract identified on an upper gastrointestinal examination. Pediatr Radiol, 2016, 46(9):1341–4., DOI 10.1007/s00247-016-3558-2 Abdullgaffar B, Al-Murbati B, Al-Falasi M, Al-Otaibi L. Unsuspected Langerhans cell histiocytosis can be easily missed in a colonic biopsy. Fetal Pediatr Pathol, 2014, 33(2):98–103., DOI 10.3109/15513815.2013.864349 Hait E, Liang M, Degar B, Glickman J, Fox VL. Gastrointestinal tract involvement in Langerhans cell histiocytosis: case report and literature review. Pediatrics, 2006, 118(5):e1593–9., DOI 10.1542/peds.2006-0708 Mayumi A, Imamura T, Sakamoto K, Ota T, Osone S, Usami I, et al. Successful treatment with 2-chlorodeoxyadenosine of refractory pediatric Langerhans cell histiocytosis with initial involvement of the gastrointestinal tract. Int J Hematol, 2019, 110(6):756–62., DOI 10.1007/s12185-019-02711-0 Shima H, Takahashi T, Shimada H. Protein-losing enteropathy caused by gastrointestinal tract-involved Langerhans cell histiocytosis. Pediatrics, 2010, 125(2):e426–32., DOI 10.1542/peds.2009-1742 Levy J, Khaskelberg A, Garvin J. Langerhans cell histiocytosis, LCH, involving the gastrointestinal tract: a clinical-electrogastrographic correlation. J Pediatr Gastroenterol Nutr, 2001, 33(4):511–4., DOI 10.1097/00005176-200110000-00020 Godoy MM, Fernandes M, Halenda GM, Javid G, Ngo PD. Gastrointestinal Langerhans cell histiocytosis. J Pediatr Gastroenterol Nutr, 2014, 59(2):e17., DOI 10. 1097/MPG.0000000000000027 Fang J, Chen J, Deng C, Li J, Huang T, Li Y, et al. Langerhans cell histiocytosis in a pediatric patient with simultaneous gastrointestinal and skin involvement: a case report and literature review. Fetal Pediatr Pathol, 2023, 42(3):483–7., DOI 10. 1080/15513815.2022.2142488 Miller M, Iyer R, Bishop P, Nowicki M. Protein-losing enteropathy due to intestinal and colonic involvement with langerhans cell histiocytosis and review of the literature. Clin Pediatr, Phila,, 2014, 53(1):89–94., DOI 10.1177/ 0009922813492884 Choi SW, Bangaru BS, Wu CD, Finlay JL. Gastrointestinal involvement in disseminated Langerhans cell histiocytosis, LCH, with durable complete response to 2-chlorodeoxyadenosine and high-dose cytarabine. J Pediatr Hematol Oncol, 2003, 25(6):503–6., DOI 10.1097/00043426-200306000-00016 Lv X, Wang L, Pi Z, Zhang C. Langerhans cell histiocytosis misdiagnosed as cow protein allergy: a case report. BMC Pediatr, 2021, 21(1):469., DOI 10.1186/ s12887-021-02921-8 Boccon-Gibod LA, Krichen H, Carlier-Mercier L, Salaun JF, Fontaine JL, Leverger GR. Digestive tract involvement with exudative enteropathy in Langerhans cell histiocytosis. Pediatr Pathol, 1992, 12:515–24., DOI 10.3109/ 15513819209024201 Gilmore BS, Cohen M. Barium enema findings in a case of Langerhans cell histiocytosis involving the colon. Pediatr Radiol, 1993, 23(8):589–90., DOI 10.1007/ BF02014971 Santos-Machado TM, Cristofani LM, Almeida MT, Maluf PT, Costa PA, Pereira MA, et al. Disseminated Langerhans’ cell histiocytosis and massive proteinlosing enteropathy. Braz J Med Biol Res, 1999, 32(9):1095–9., DOI 10.1590/s0100- 879x1999000900007 Carlier-Mercier L, Fontaine JL, Boccon-Gibod L, Girardet JP, Josset P, Gouraud F, et al. A rare cause of neonatal exudative enteropathy: congenital Langerhans cell histiocytosis, histiocytosis X). Arch Fr Pediatr, 1992, 49(3): 199–201. Patel BJ, Chippindale AJ, Gupta SC. Case report: small bowel histiocytosis-X. Clin Radiol, 1991, 44(1):62–3., DOI 10.1016/s0009-9260(05)80231-4 Geissmann F, Thomas C, Emile JF,Micheau M, Canioni D, Cerf-Bensussan N, et al. Digestive tract involvement in langerhans cell histiocytosis. The French langerhans cell histiocytosis study group. J Pediatr, 1996, 129(6):836–45., DOI 10. 1016/s0022-3476(96)70027-6 Lee RG, Braziel RM, Stenzel P. Gastrointestinal involvement in Langerhans cell histiocytosis, histiocytosis X): diagnosis by rectal biopsy. Mod Pathol, 1990, 3(2):154–7. Yadav SP, Kharya G, Mohan N, Sehgal A, Bhat S, Jain S, et al. Langerhans cell histiocytosis with digestive tract involvement. Pediatr Blood Cancer, 2010, 55(4): 748–53., DOI 10.1002/pbc.22663 Vetter-Laracy S, Salinas JA, Martin-Santiago A, Guibelalde M, Balliu PR. Digestive tract symptoms in congenital Langerhans cell histiocytosis: a fatal condition in an illness usually considered benign. J Pediatr Hematol Oncol, 2014, 36(6):426–9., DOI 10.1097/MPH.0b013e31829f35ad Yan F, Zhou Q, Gao Y, Chang H, Li X, Li Y, et al. Isolated Langerhans cell histiocytosis of the stomach: a case report and literature review. Int J Clin Exp Pathol, 2018, 11(12):5962–8. Singhi AD, Montgomery EA. Gastrointestinal tract Langerhans cell histiocytosis: a clinicopathologic study of 12 patients. Am J Surg Pathol, 2011, 35(2):305–10., DOI 10.1097/PAS.0b013e31820654e4 Durham BH. Molecular characterization of the histiocytoses: neoplasia of dendritic cells and macrophages. Semin Cell Dev Biol, 2019, 86:62–76., DOI 10.1016/ j.semcdb.2018.03.002 Kobayashi M, Tojo A. Langerhans cell histiocytosis in adults: advances in pathophysiology and treatment. Cancer Sci, 2018, 109(12):3707–13., DOI 10.1111/ cas.13817 Behdad A, Owens SR. Langerhans cell histiocytosis involving the gastrointestinal tract. Arch Pathol Lab Med, 2014, 138(10):1350–2., DOI 10.5858/ arpa.2014-0290-CC McClain KL, Bigenwald C, Collin M, Haroche J, Marsh RA, Merad M, et al. Histiocytic disorders. Nat Rev Dis Primers, 2021, 7(1):73., DOI 10.1038/s41572-021- 00307-9 Yoon HS, Lee JH, Michlitsch J, Garcia-Carega M, Jeng M. Langerhans cell histiocytosis of the gastrointestinal tract: evidence for risk organ status. J Pediatr, 2019, 212:66–72., DOI 10.1016/j.jpeds.2019.05.003 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611705 EP 1611711 DI 10.3389/pore.2024.1611705 PG 7 ER PT J AU Csiki, E Simon, M Papp, J Barabas, M Mikaczo, J Gal, K Sipos, D Kovacs, AF Csiki, Emese Simon, Mihaly Papp, Judit Barabas, Marton Mikaczo, Johanna Gal, Kristof Sipos, David Kovacs, Arpad TI Stereotactic body radiotherapy in lung cancer: a contemporary review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE SBRT; NSCLC; lung cancer; review; radiotherapy ID SBRT; NSCLC; lung cancer; review; radiotherapy AB The treatment of early stage non-small cell lung cancer (NSCLC) has improved enormously in the last two decades. Although surgery is not the only choice, lobectomy is still the gold standard treatment type for operable patients. For inoperable patients stereotactic body radiotherapy (SBRT) should be offered, reaching very high local control and overall survival rates. With SBRT we can precisely irradiate small, well-defined lesions with high doses. To select the appropriate fractionation schedule it is important to determine the size, localization and extent of the lung tumor. The introduction of novel and further developed planning (contouring guidelines, diagnostic image application, planning systems) and delivery techniques (motion management, image guided radiotherapy) led to lower rates of side effects and more conformal target volume coverage. The purpose of this study is to summarize the current developments, randomised studies, guidelines about lung SBRT, with emphasis on the possibility of increasing local control and overall rates in “fit,” operable patients as well, so SBRT would be eligible in place of surgery. C1 [Csiki, Emese] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Simon, Mihaly] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Papp, Judit] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Barabas, Marton] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Mikaczo, Johanna] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Gal, Kristof] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. [Sipos, David] University of Pecs, Faculty of Health SciencesPecs, Hungary. [Kovacs, Arpad] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. RP Simon, M (reprint author), Debreceni Egyetem, Onkologiai Klinika, Sugarterapia Osztaly, Debrecen, Hungary. EM simonm@med.unideb.hu CR American Cancer Society. Facts & Figures 2019. Am Cancer Soc [Internet], 2019):1–76. Available at: https://www.cancer.org/content/dam/cancer-org/ research/cancerfacts-and-statistics/annual-cancer-facts-andfigures/2019/cancerfacts- andfigures. Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, et al. The 2015 world health organization classification of lung tumors: impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol, 2015, 10:1243–60., DOI 10.1097/JTO.0000000000000630 Howlader N, Forjaz G, Mooradian MJ,Meza R, Kong CY, Cronin KA, et al. The effect of advances in lung-cancer treatment on population mortality. N Engl J Med, 2020, 383(7):640–9., DOI 10.1056/NEJMoa1916623 Ganti M, Alyssa BK, Ion C, Seal B, Chou E. Update of incidence, prevalence, survival, and initial treatment in patients with non-small cell lung cancer in the US. JAMA Oncol, 2021, 7(12):1824–32., DOI 10.1001/jamaoncol.2021.4932 Goldstraw P, Chansky K, Crowley J, Rami-Porta R, Asamura H, Eberhardt WEE, et al. The IASLC lung cancer staging project: proposals for revision of the TNM stage groupings in the forthcoming, eighth, edition of the TNM Classification for lung cancer. J Thorac Oncol, 2016, 11(1):39–51., DOI 10. 1016/j.jtho.2015.09.009 NCCN. NCCN Guidelines Version 3. 2023 Non-small cell lung cancer, 2023). Howington JA, Blum MG, Chang AC, Balekian AA, Murthy SC. Treatment of stage I and II non-small cell lung cancer: diagnosis and management of lung cancer, 3rd ed: american College of Chest Physicians evidence-based clinical practice guidelines. Chest, 2013, 143(5):e278S–e313S., DOI 10.1378/chest.12-2359 Chang JY, Senan S, Paul MA, Mehran RJ, Louie AV, Balter P, et al. Stereotactic ablative radiotherapy versus lobectomy for operable stage I non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Oncol, 2015, 16(6):630–7., DOI 10.1016/S1470-2045(15)70168-3 Timmerman RD, Herman J, Cho LC. Emergence of stereotactic body radiation therapy and its impact on current and future clinical practice. J Clin Oncol, 2014, 32:2847–54., DOI 10.1200/JCO.2014.55.4675 Prezzano KM, Ma SJ, Hermann GM, Rivers CI, Gomez-Suescun JA, Singh AK. Stereotactic body radiation therapy for non-small cell lung cancer: a review. World J Clin Oncol, 2019, 10(1):14–27., DOI 10.5306/wjco.v10.i1.14 Rowell NP, Williams CJ. Radical radiotherapy for stage I/II non-small cell lung cancer in patients not sufficiently fit for or declining surgery, medically inoperable). Cochrane Database Syst Rev, 2001, 2:CD002935., DOI 10.1002/ 14651858.CD002935 Nyman J, Hallqvist A, Lund JA, Brustugun OT, Bergman B, Bergstrom P, et al. SPACE – a randomized study of SBRT vs conventional fractionated radiotherapy in medically inoperable stage I NSCLC. Radiother Oncol, 2016, 121(1):1–8., DOI 10. 1016/j.radonc.2016.08.015 Ball D, Mai GT, Vinod S, Babington S, Ruben J, Kron T, et al. Stereotactic ablative radiotherapy versus standard radiotherapy in stage 1 non-small-cell lung cancer, TROG 09.02 CHISEL): a phase 3, open-label, randomised controlled trial. Lancet Oncol, 2019, 20(4):494–503., DOI 10.1016/S1470-2045(18)30896-9 von Reibnitz D, Shaikh F, Wu AJ, Treharne GC, Dick-Godfrey R, Foster A, et al. Stereotactic body radiation therapy, SBRT, improves local control and overall survival compared to conventionally fractionated radiation for stage I non-small cell lung cancer, NSCLC). Acta Oncol, Madr,, 2018, 57(11):1567–73., DOI 10.1080/ 0284186X.2018.1481292 Videtic GMM, Hu C, Singh AK, Chang JY, Parker W, Olivier KR, et al. A randomized phase 2 study comparing 2 stereotactic body radiation therapy schedules for medically inoperable patients with stage i peripheral non-small cell lung cancer: NRG Oncology RTOG 0915, NCCTG N0927). Int J Radiat Oncol Biol Phys, 2015, 93:757–64., DOI 10.1016/j.ijrobp.2015.07.2260 Rodriguez De Dios N, Navarro-Martin A, Cigarral C, Chicas-Sett R, Garcia R, Garcia V, et al. GOECP/SEOR radiotheraphy guidelines for non-small-cell lung cancer. World J Clin Oncol, 2022, 13(4):237–66., DOI 10.5306/wjco.v13.i4.237 Crabtree T, Puri V, Timmerman R, Fernando H, Bradley J, Decker PA, et al. Treatment of stage i lung cancer in high-risk and inoperable patients: comparison of prospective clinical trials using stereotactic body radiotherapy, RTOG 0236), sublobar resection, ACOSOG Z4032), and radiofrequency ablation, ACOSOG Z4033). J Thorac Cardiovasc Surg, 2013, 145(3):692–9., DOI 10.1016/j.jtcvs.2012. 10.038 Sun B, Brooks ED, Komaki RU, Liao Z, Jeter MD, McAleer MF, et al. 7-year follow-up after stereotactic ablative radiotherapy for patients with stage I non–small cell lung cancer: results of a phase 2 clinical trial. Cancer, 2017, 123(16):3031–9., DOI 10.1002/cncr.30693 Nagata Y, Hiraoka M, Shibata T, Onishi H, Kokubo M, Karasawa K, et al. Prospective trial of stereotactic body radiation therapy for both operable and inoperable T1N0M0 non-small cell lung cancer: japan Clinical Oncology Group Study JCOG0403. Int J Radiat Oncol Biol Phys, 2015, 93(5):989–96., DOI 10.1016/j. ijrobp.2015.07.2278 Timmerman RD, Hu C, Michalski JM, Bradley JC, Galvin J, Johnstone DW, et al. Long-term results of stereotactic body radiation therapy in medically inoperable stage I non-small cell lung cancer. JAMA Oncol, 2018, 4(9):1287–8., DOI 10.1001/jamaoncol.2018.1258 Verstegen NE, Oosterhuis JWA, Palma DA, Rodrigues G, Lagerwaard FJ, van der Elst A, et al. Stage I-II non-small-cell lung cancer treated usingeither stereotactic ablative radiotherapy, SABR, orlobectomy by video-assisted thoracoscopic surgery(VATS): outcomes of a propensity score-matchedanalysis. Ann Oncol, 2013, 24(6):1543–8., DOI 10.1093/annonc/mdt026 Franks KN, McParland L, Webster J, Baldwin DR, Sebag-Montefiore D, Evison M, et al. SABRTooth: a randomised controlled feasibility study of stereotactic ablative radiotherapy, SABR, with surgery in patients with peripheral stage I nonsmall cell lung cancer considered to be at higher risk of complications from surgical resection. Eur Respir J, 2020, 56(5):2000118., DOI 10. 1183/13993003.00118-2020 Yamamoto K, Ohsumi A, Kojima F, Imanishi N, Matsuoka K, Ueda M, et al. Long-term survival after video-assisted thoracic surgery lobectomy for primary lung cancer. Ann Thorac Surg, 2010, 89(2):353–9., DOI 10.1016/j.athoracsur.2009.10.034 Ilonen IK, Rasanen JV, Knuuttila A, Salo JA, Sihvo EI. Anatomic thoracoscopic lung resection for non-small cell lung cancer in stage i is associated with less morbidity and shorter hospitalization than thoracotomy. Acta Oncol, Madr,, 2011, 50(7):1126–32., DOI 10.3109/0284186X.2011.555780 Onkologie A, Daten D, Breyer-kohansal R, Hartl S, Burghuber OC, Urban M, et al. National Lung Cancer Audit annual report, for the audit period 2018). Eur Respir J, 2021, 14:2103201. Chang JY, Mehran RJ, Feng L, Verma V, Liao Z, Welsh JW, et al. Stereotactic ablative radiotherapy for operable stage I non-small-cell lung cancer, revised STARS): long-term results of a single-arm, prospective trial with prespecified comparison to surgery. Lancet Oncol, 2021, 22(10):1448–57., DOI 10.1016/S1470- 2045(21)00401-0 Timmerman RD, Paulus R, Pass HI, Gore EM, Edelman MJ, Galvin J, et al. Stereotactic body radiation therapy for operable early-stage lung cancer: findings from the NRG oncology RTOG 0618 trial. JAMA Oncol, 2018, 4(9):1263–6., DOI 10. 1001/jamaoncol.2018.1251 Moghanaki D. Strategic initiatives for veterans with lung cancer. Fed Pract, 2020, 37:S76–S80., DOI 10.12788/fp.0019 Chang JY, Bezjak A, Mornex F, IASLC Advanced Radiation Technology Committee. Stereotactic ablative radiotherapy for centrally located early stage non–small-cell lung cancer: what we have learned. J Thorac Oncol, 2015, 10: 577–85., DOI 10.1097/JTO.0000000000000453 Adebahr S, Collette S, Shash E, Lambrecht M, Le Pechoux C, Faivre-Finn C, et al. LungTech, an EORTC Phase II trial of stereotactic body radiotherapy for centrally located lung tumours: a clinical perspective. Br J Radiol, 2015, 88(1051): 20150036., DOI 10.1259/bjr.20150036 Stephans KL, Djemil T, Tendulkar RD, Robinson CG, Reddy CA, Videtic GMM. Prediction of chest wall toxicity from lung stereotactic body radiotherapy, SBRT). Int J Radiat Oncol Biol Phys, 2012, 82(2):974–80., DOI 10.1016/j.ijrobp.2010. 12.002 Roach MC, Robinson CG, DeWees TA, Ganachaud J, Przybysz D, Drzymala R, et al. Stereotactic body radiation therapy for central early-stage NSCLC: results of a prospective phase I/II trial. J Thorac Oncol, 2018, 13(11):1727–32., DOI 10.1016/j. jtho.2018.07.017 Timmerman R, McGarry R, Yiannoutsos C, Papiez L, Tudor K, DeLuca J, et al. Excessive toxicity when treating central tumors in a phase II study of stereotactic body radiation therapy for medically inoperable early-stage lung cancer. J Clin Oncol, 2006, 24(30):4833–9., DOI 10.1200/JCO.2006.07.5937 Fakiris AJ, McGarry RC, Yiannoutsos CT, Papiez L, Williams M, Henderson MA, et al. Stereotactic body radiation therapy for early-stage non-small-cell lung carcinoma: four-year results of a prospective phase II study. Int J Radiat Oncol Biol Phys, 2009, 75(3):677–82., DOI 10.1016/j.ijrobp.2008.11.042 Bezjak A, Paulus R, Gaspar LE, Timmerman RD, Straube WL, Ryan WF, et al. Safety and efficacy of a five-fraction stereotactic body radiotherapy schedule for centrally located non-small-cell lung cancer: NRG oncology/RTOG 0813 trial. J Clin Oncol, 2019, 37:1316–25., DOI 10.1200/JCO.18.00622 Haasbeek CJA, Lagerwaard FJ, Slotman BJ, Senan S. Outcomes of stereotactic ablative radiotherapy for centrally located early-stage lung cancer. J Thorac Oncol, 2011, 6., DOI 10.1097/JTO.0b013e31822e71d8 Chaudhuri AA, Chen K, DiehnM, Loo BW. Stereotactic ablative radiotherapy for central and ultra-central lung tumors. Ther Radiol Oncol, 2019, 3:18., DOI 10. 21037/tro.2019.05.01 Chang JY, Li QQ, Xu QY, Allen PK, Rebueno N, Gomez DR, et al. Stereotactic ablative radiation therapy for centrally located early stage or isolated parenchymal recurrences of non-small cell lung cancer: how to fly in a “no fly zone.”. Int J Radiat Oncol Biol Phys, 2014, 88(5):1120–8., DOI 10.1016/j.ijrobp.2014.01.022 Giuliani M, Mathew AS, Bahig H, Bratman SV, Filion E, Glick D, et al. SUNSET: stereotactic radiation for ultracentral non–small-cell lung cancer—a safety and efficacy trial. Clin Lung Cancer, 2018, 19(4):e529–32., DOI 10.1016/j. cllc.2018.04.001 Rosenberg SA, Mak R, Kotecha R, Loo BW, Senan S. The nordic-HILUS trial: ultracentral lung stereotactic ablative radiotherapy and a narrow therapeutic window. J Thorac Oncol, 2021, 16:e79–80., DOI 10.1016/j.jtho.2021.06.030 Lindberg K, Grozman V, Karlsson K, Lindberg S, Lax I, Wersall P, et al. The HILUS-trial—a prospective nordic multicenter phase 2 study of ultracentral lung tumors treated with stereotactic body radiotherapy. J Thorac Oncol, 2021, 16(7): 1200–10., DOI 10.1016/j.jtho.2021.03.019 Chen H, Laba JM, Zayed S, Boldt RG, Palma DA, Louie AV. Safety and effectiveness of stereotactic ablative radiotherapy for ultra-central lung lesions: a systematic review. J Thorac Oncol, 2019, 14:1332–42., DOI 10.1016/j.jtho.2019. 04.018 Yan M, Louie AV, Kotecha R, Ashfaq Ahmed M, Zhang Z, Guckenberger M, et al. Stereotactic body radiotherapy for ultra-central lung tumors: a systematic review and meta-analysis and international stereotactic radiosurgery society practice guidelines. Lung Cancer, 2023, 182:107281., DOI 10.1016/j.lungcan.2023. 107281 Regnery S, Ristau J, Weykamp F, Hoegen P, Sprengel SD, Paul KM, et al. Magnetic resonance guided adaptive stereotactic body radiotherapy for lung tumors in ultracentral location: the MAGELLAN trial, ARO 2021-3). Radiat Oncol, 2022, 17(1):102., DOI 10.1186/s13014-022-02070-x Guckenberger M. Dose and fractionation in stereotactic body radiation therapy for stage i non-small cell lung cancer: lessons learned and where do we go next? Int J Radiat Oncol Biol Phys, 2015, 93:765–8., DOI 10.1016/j.ijrobp.2015. 08.025 Onishi H, Araki T, Shirato H, Nagata Y, Hiraoka M, Gomi K, et al. Stereotactic hypofractionated high-dose irradiation for stage I nonsmall cell lung carcinoma: clinical outcomes in 245 subjects in a Japanese multiinstitutional study. Cancer, 2004, 101(7):1623–31., DOI 10.1002/cncr.20539 Menten MJ, Wetscherek A, Fast MF. MRI-guided lung SBRT: present and future developments. Physica Med, 2017, 44:139–49., DOI 10.1016/j.ejmp.2017. 02.003 Onishi H, Shirato H, Nagata Y, Hiraoka M, Fujino M, Gomi K, et al. Hypofractionated stereotactic radiotherapy, HypoFXSRT, for stage I non-small cell lung cancer: updated results of 257 patients in a Japanese multi-institutional study. J Thorac Oncol, 2007, 2., DOI 10.1097/JTO.0b013e318074de34 Moreno AC, Fellman B, Hobbs BP, Liao Z, Gomez DR, Chen A, et al. Biologically effective dose in stereotactic body radiotherapy and survival for patients with early-stage NSCLC. J Thorac Oncol, 2020, 15(1):101–9., DOI 10.1016/j.jtho. 2019.08.2505 Koshy M, Malik R, Weichselbaum RR, Sher DJ. Increasing radiation therapy dose is associated with improved survival in patients undergoing stereotactic body radiation therapy for stage I non-small-cell lung cancer. Int J Radiat Oncol Biol Phys, 2015, 91(2):344–50., DOI 10.1016/j.ijrobp.2014.10.002 Videtic GM, Paulus R, Singh AK, Chang JY, Parker W, Olivier KR, et al. Longterm follow-up on NRG oncology RTOG 0915, NCCTG N0927): a randomized phase 2 study comparing 2 stereotactic body radiation therapy schedules for medically inoperable patients with stage I peripheral non-small cell lung cancer. Int J Radiat Oncol Biol Phys, 2019, 103(5):1077–84., DOI 10.1016/j.ijrobp.2018. 11.051 Singh AK, Gomez-Suescun JA, Stephans KL, Bogart JA, Hermann GM, Tian L, et al. One versus three fractions of stereotactic body radiation therapy for peripheral stage I to II non-small cell lung cancer: a randomized, multi-institution, phase 2 trial. Int J Radiat Oncol Biol Phys, 2019, 105(4):752–9., DOI 10.1016/j.ijrobp. 2019.08.019 Tjong MC, Louie AV, Singh AK, Videtic G, Stephans K, Plumridge N, et al. Single-fraction stereotactic ablative body radiotherapy to the lung – the knockout punch. Clin Oncol, 2022, 34(5):e183–94., DOI 10.1016/j.clon.2022.02.004 GuckenbergerM, Belka C, Bezjak A, Bradley J, Daly ME, DeRuysscher D, et al. Practice recommendations for lung cancer radiotherapy during the COVID-19 pandemic: an ESTRO-ASTRO consensus statement. Radiother Oncol, 2020, 146: 223–9., DOI 10.1016/j.radonc.2020.04.001 Park C, Papiez L, Zhang S, Story M, Timmerman RD. Universal survival curve and single fraction equivalent dose: useful tools in understanding potency of ablative radiotherapy. Int J Radiat Oncol Biol Phys, 2008, 70(3):847–52., DOI 10. 1016/j.ijrobp.2007.10.059 Guckenberger M, Klement RJ, Allgauer M, Appold S, Dieckmann K, Ernst I, et al. Applicability of the linear-quadratic formalism for modeling local tumor control probability in high dose per fraction stereotactic body radiotherapy for early stage non-small cell lung cancer. Radiother Oncol, 2013, 109(1):13–20., DOI 10.1016/ j.radonc.2013.09.005 Wang B, Dong Y, Yu X, Li F, Wang J, Chen H, et al. A review on the applications of Traditional Chinese medicine polysaccharides in drug delivery systems. Front Oncol, 2022, 17:12., DOI 10.1186/s13020-021-00567-3 Verma V, Shostrom VK, Kumar SS, Zhen W, Hallemeier CL, Braunstein SE, et al. Multi-institutional experience of stereotactic body radiotherapy for large, ≥5 centimeters, non–small cell lung tumors. Cancer, 2017, 123(4):688–96., DOI 10. 1002/cncr.30375 Benedict SH, Yenice KM, Followill D, Galvin JM, Hinson W, Kavanagh B, et al. Stereotactic body radiation therapy: the report of AAPM Task Group 101. Med Phys, 2010, 37:4078–101., DOI 10.1118/1.3438081 Keall PJ, Mageras GS, Balter JM, Emery RS, Forster KM, Jiang SB, et al. The management of respiratory motion in radiation oncology report of AAPM Task Group 76. Med Phys, 2006, 33:3874–900., DOI 10.1118/1.2349696 Caillet V, Booth JT, Keall P. IGRT andmotionmanagement during lung SBRT delivery. Physica Med, 2017, 44:113–22., DOI 10.1016/j.ejmp.2017.06.006 Jin JY, Ajlouni M, Chen Q, Kong FM, Ryu S, Movsas B. Quantification of incidental dose to potential clinical target volume, CTV, under different stereotactic body radiation therapy, SBRT, techniques for non-small cell lung cancer - tumor motion and using internal target volume, ITV, could improve dose distribution in CTV. Radiother Oncol, 2007, 85(2):267–76., DOI 10.1016/j.radonc. 2007.09.004 Marks LB, Yorke ED, Jackson A, Ten Haken RK, Constine LS, Eisbruch A, et al. Use of normal tissue complication probability models in the clinic. Int J Radiat Oncol Biol Phys, 2010, 76(3):S10–S19., DOI 10.1016/j.ijrobp.2009.07. 1754 Diez P, Hanna GG, Aitken KL, van As N, Carver A, Colaco RJ, et al. UK 2022 consensus on normal tissue dose-volume constraints for oligometastatic, primary lung and hepatocellular carcinoma stereotactic ablative radiotherapy. Clin Oncol, 2022, 34(5):288–300., DOI 10.1016/j.clon.2022.02.010 Grimm J, Marks LB, Jackson A, Kavanagh BD, Xue J, Yorke E. High dose per fraction, hypofractionated treatment effects in the clinic, HyTEC): an overview. Int J Radiat Oncol Biol Phys, 2021, 110(1):1–10., DOI 10.1016/j.ijrobp.2020.10.039 Verbakel WFAR, Senan S, Cuijpers JP, Slotman BJ, Lagerwaard FJ. Rapid delivery of stereotactic radiotherapy for peripheral lung tumors using volumetric intensity-modulated arcs. Radiother Oncol, 2009, 93(1):122–4., DOI 10.1016/j. radonc.2009.05.020 Navarria P, Ascolese AM, Mancosu P, Alongi F, Clerici E, Tozzi A, et al. Volumetric modulated arc therapy with flattening filter free, FFF, beams for stereotactic body radiation therapy, SBRT, in patients with medically inoperable early stage non small cell lung cancer, NSCLC). Radiother Oncol, 2013, 107(3): 414–8., DOI 10.1016/j.radonc.2013.04.016 De Ruysscher D, Belderbos J, Reymen B, Van Elmpt W, Van Baardwijk A, Wanders R, et al. State of the art radiation therapy for lung cancer 2012: a glimpse of the future. Clin Lung Cancer, 2013, 14:89–95., DOI 10.1016/j.cllc.2012.06.006 Devaraj A, Cook GJR, Hansell DM. PET/CT in non-small cell lung cancer staging-promises and problems. Clin Radiol, 2007, 62:97–108., DOI 10.1016/j.crad. 2006.09.015 Cheebsumon P, Boellaard R, de Ruysscher D, van ElmptW, van Baardwijk A, Yaqub M, et al. Assessment of tumour size in PET/CT lung cancer studies: PET- and CT-based methods compared to pathology. EJNMMI Res, 2012, 2(1):56–9., DOI 10. 1186/2191-219X-2-56 Geiger GA, Kim MB, Xanthopoulos EP, Pryma DA, Grover S, Plastaras JP, et al. Stage migration in planning PET/CT scans in patients due to receive radiotherapy for non-small-cell lung cancer. Clin Lung Cancer, 2014, 15(1): 79–85., DOI 10.1016/j.cllc.2013.08.004 Loganadane G, Martinetti F, Mercier O, Krhili S, Riet FG, Mbagui R, et al. Stereotactic ablative radiotherapy for early stage non-small cell lung cancer: a critical literature review of predictive factors of relapse. Cancer Treat Rev, 2016, 50: 240–6., DOI 10.1016/j.ctrv.2016.10.002 Chang JY, Liu H, Balter P, Komaki R, Liao Z, Welsh J, et al. Clinical outcome and predictors of survival and pneumonitis after stereotactic ablative radiotherapy for stage I non-small cell lung cancer. Radiat Oncol, 2012, 7(1):152., DOI 10.1186/ 1748-717X-7-152 Vansteenkiste J, Crino L, Dooms C, Douillard JY, Faivre-Finn C, Lim E, et al. 2nd ESMO consensus conference on lung cancer: early-stage non-small-cell lung cancer consensus on diagnosis, treatment and follow-up. Ann Oncol, 2014, 25(8): 1462–74., DOI 10.1093/annonc/mdu089 Videtic GMM, Donington J, Giuliani M, Heinzerling J, Karas TZ, Kelsey CR, et al. Stereotactic body radiation therapy for early-stage non-small cell lung cancer: executive Summary of an ASTRO Evidence-Based Guideline. Pract Radiat Oncol, 2017, 7(5):295–301., DOI 10.1016/j.prro.2017.04.014 Zeng J, Harris TJ, Lim M, Drake CG, Tran PT. Immune modulation and stereotactic radiation: improving local and abscopal responses. Biomed Res Int, 2013, 2013:658126., DOI 10.1155/2013/658126 Chen Y, Gao M, Huang Z, Yu J, Meng X. SBRT combined with PD-1/PDL1 inhibitors in NSCLC treatment: a focus on the mechanisms, advances, and future challenges. J Hematol Oncol, 2020, 13:105., DOI 10.1186/s13045-020-00940-z Shaverdian N, Lisberg AE, Bornazyan K, Veruttipong D, Goldman JW, Formenti SC, et al. Previous radiotherapy and the clinical activity and toxicity of pembrolizumab in the treatment of non-small-cell lung cancer: a secondary analysis of the KEYNOTE-001 phase 1 trial. Lancet Oncol, 2017, 18(7):895–903., DOI 10. 1016/S1470-2045(17)30380-7 Wang YY, Wang YS, Liu T, Yang K, Yang GQ, Liu HC, et al. Efficacy study of CyberKnife stereotactic radiosurgery combined with CIK cell immunotherapy for advanced refractory lung cancer. Exp TherMed, 2013, 5(2):453–6., DOI 10.3892/etm. 2012.818 Seung SK, Curti BD, CrittendenM, Walker E, Coffey T, Siebert JC, et al. Phase 1 study of stereotactic body radiotherapy and interleukin-2--tumor and immunological responses. Sci Transl Med, 2012, 4(137):137ra74., DOI 10.1126/ scitranslmed.3003649 Monjazeb AM, Daly ME, Luxardi G, Maverakis E, Merleev AA, Marusina AI, et al. Atezolizumab plus stereotactic ablative radiotherapy for medically inoperable patients with early-stage non-small cell lung cancer: a multi-institutional phase I trial. Nat Commun, 2023, 14(1):5332., DOI 10.1038/s41467-023-40813-w Huang K, Dahele M, Senan S, Guckenberger M, Rodrigues GB, Ward A, et al. Radiographic changes after lung stereotactic ablative radiotherapy, SABR, - can we distinguish recurrence from fibrosis? A systematic review of the literature. Radiother Oncol, 2012, 102:335–42., DOI 10.1016/j.radonc.2011.12.018 Bucknell NW, Kron T, Herschtal A, Hardcastle N, Irving L, MacManus M, et al. Comparison of changes in pulmonary function after stereotactic body radiation therapy versus conventional 3-dimensional conformal radiation therapy for stage I and IIA non-small cell lung cancer: an analysis of the TROG 09.02, CHISEL, phase 3 trial. Int J Radiat Oncol Biol Phys, 2023, 117(2):378–86., DOI 10.1016/j.ijrobp.2023.04.009 Yu V, Kishan AU, CaoM, Low D, Lee P, Ruan D. Dose impact in radiographic lung injury following lung SBRT: statistical analysis and geometric interpretation. Med Phys, 2014, 41(3):031701., DOI 10.1118/1.4863483 Stanic S, Paulus R, Timmerman RD, Michalski JM, Barriger RB, Bezjak A, et al. No clinically significant changes in pulmonary function following stereotactic body radiation therapy for early-stage peripheral non-small cell lung cancer: an analysis of RTOG 0236. Int J Radiat Oncol Biol Phys, 2014, 88(5):1092–9., DOI 10. 1016/j.ijrobp.2013.12.050 Finazzi T, Palacios MA, Spoelstra FOB, Haasbeek CJA, Bruynzeel AME, Slotman BJ, et al. Role of on-table plan adaptation in MR-guided ablative radiation therapy for central lung tumors. Int J Radiat Oncol Biol Phys, 2019, 104(4):933–41., DOI 10.1016/j.ijrobp.2019.03.035 Finazzi T, Palacios MA, Haasbeek CJA, Admiraal MA, Spoelstra FOB, Bruynzeel AME, et al. Stereotactic MR-guided adaptive radiation therapy for peripheral lung tumors. Radiother Oncol, 2020, 144:46–52., DOI 10.1016/j.radonc. 2019.10.013 Henke LE, Olsen JR, Contreras JA, Curcuru A, DeWees TA, Green OL, et al. Stereotactic MR-guided online adaptive radiation therapy, SMART, for ultracentral thorax malignancies: results of a phase 1 trial. Adv Radiat Oncol, 2019, 4(1):201–9., DOI 10.1016/j.adro.2018.10.003 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611709 EP 1611721 DI 10.3389/pore.2024.1611709 PG 13 ER PT J AU Galffy, G Morocz, Korompay, R Hecz, R Bujdoso, R Puskas, R Lovas, T Gaspar, E Yahya, K Kiraly, P Lohinai, Z AF Galffy, Gabriella Morocz, Eva Korompay, Reka Hecz, Reka Bujdoso, Reka Puskas, Rita Lovas, Timea Gaspar, Eszter Yahya, Kamel Kiraly, Peter Lohinai, Zoltan TI Targeted therapeutic options in early and metastatic NSCLC-overview SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE NSCLC; driver oncogenes; targeted therapy; central nervous system efficacy; molecular testing ID NSCLC; driver oncogenes; targeted therapy; central nervous system efficacy; molecular testing AB The complex therapeutic strategy of non-small cell lung cancer (NSCLC) has changed significantly in recent years. Disease-free survival increased significantly with immunotherapy and chemotherapy registered in perioperative treatments, as well as adjuvant registered immunotherapy and targeted therapy (osimertinib) in case of EGFR mutation. In oncogenic-addictive metastatic NSCLC, primarily in adenocarcinoma, the range of targeted therapies is expanding, with which the expected overall survival increases significantly, measured in years. By 2021, the FDA and EMA have approved targeted agents to inhibit EGFR activating mutations, T790 M resistance mutation, BRAF V600E mutation, ALK, ROS1, NTRK and RET fusion. In 2022, the range of authorized target therapies was expanded. With therapies that inhibit KRASG12C, EGFR exon 20, HER2 and MET. Until now, there was no registered targeted therapy for the KRAS mutations, which affect 30% of adenocarcinomas. Thus, the greatest expectation surrounded the inhibition of the KRAS G12C mutation, which occurs in ~15% of NSCLC, mainly in smokers and is characterized by a poor prognosis. Sotorasib and adagrasib are approved as second-line agents after at least one prior course of chemotherapy and/or immunotherapy. Adagrasib in first-line combination with pembrolizumab immunotherapy proved more beneficial, especially in patients with high expression of PD-L1. In EGFR exon 20 insertion mutation of lung adenocarcinoma, amivantanab was registered for progression after platinum-based chemotherapy. Lung adenocarcinoma carries an EGFR exon 20, HER2 insertion mutation in 2%, for which the first targeted therapy is trastuzumab deruxtecan, in patients already treated with platinum-based chemotherapy. Two orally administered selective c-MET inhibitors, capmatinib and tepotinib, were also approved after chemotherapy in adenocarcinoma carrying MET exon 14 skipping mutations of about 3%. Incorporating reflex testing with next-generation sequencing (NGS) expands personalized therapies by identifying guideline-recommended molecular alterations. C1 [Galffy, Gabriella] County Hospital of PulmonologyTorokbalint, Hungary. [Morocz, Eva] County Hospital of PulmonologyTorokbalint, Hungary. [Korompay, Reka] County Hospital of PulmonologyTorokbalint, Hungary. [Hecz, Reka] County Hospital of PulmonologyTorokbalint, Hungary. [Bujdoso, Reka] County Hospital of PulmonologyTorokbalint, Hungary. [Puskas, Rita] County Hospital of PulmonologyTorokbalint, Hungary. [Lovas, Timea] County Hospital of PulmonologyTorokbalint, Hungary. [Gaspar, Eszter] County Hospital of PulmonologyTorokbalint, Hungary. [Yahya, Kamel] County Hospital of PulmonologyTorokbalint, Hungary. [Kiraly, Peter] County Hospital of PulmonologyTorokbalint, Hungary. [Lohinai, Zoltan] County Hospital of PulmonologyTorokbalint, Hungary. RP Lohinai, Z (reprint author), County Hospital of Pulmonology, Torokbalint, Hungary. EM lohinai.zoltan@semmelweis.hu CR Hendriks LE, Kerr KM,Menis J, Mok TS, Nestle U, Passaro A, et al. Oncogeneaddicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol, 2023, 34:339–57., DOI 10.1016/j. annonc.2022.12.009 NCCN. NCCN guidelines non-small cell lung cancer version 1, 2024). Available from: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf, Accessed January 30, 2024). Ahn M, Arcila M, Bazhenova L, Beasley M, Berezowska S, Bubendorf L, et al. IASLC atlas of molecular testing for targeted therapy in lung cancer. Denver: International Association for the Study of Lung Cancer, 2023). Fois SS, Paliogiannis P, Zinellu A, Fois AG, Cossu A, Palmieri G Molecular epidemiology of the main druggable genetic alterations in non-small cell lung cancer. Int J Mol Sci, 2021, 22:612., DOI 10.3390/ijms22020612 Westcott PMK, Halliwill KD, To MD, Rashid M, Rust AG, Keane TM, et al. The mutational landscapes of genetic and chemical models of Kras-driven lung cancer. Nature, 2015, 517:489–92., DOI 10.1038/nature13898 Cserepes M, Ostoros G, Lohinai Z, Raso E, Barbai T, Timar J, et al. Subtypespecific KRAS mutations in advanced lung adenocarcinoma: a retrospective study of patients treated with platinum-based chemotherapy. Eur J Cancer Oxf Engl, 2014, 50:1819–28., DOI 10.1016/j.ejca.2014.04.001 Wu YL, John T, Grohe C, Majem M, Goldman JW, Kim SW, et al. Postoperative chemotherapy use and outcomes from ADAURA: osimertinib as adjuvant therapy for resected EGFR-mutated NSCLC. J Thorac Oncol, 2022, 17: 423–33., DOI 10.1016/j.jtho.2021.10.014 Herbst RS, Wu Y-L, John T, Grohe C, Majem M, Wang J, et al. Adjuvant osimertinib for resected EGFR-mutated stage IB-IIIA non-small-cell lung cancer: updated results from the phase III randomized ADAURA trial. J Clin Oncol, 2023, 41:1830–40., DOI 10.1200/JCO.22.02186 Kohno T, Matsui T, Enatsu S Differences between EGFR exon 19 deletion and exon 21 L858R point mutation, frequently detected EGFR mutations in patients with non-small cell lung cancer, from a molecular biology viewpoint. Gan To Kagaku Ryoho, 2021, 48:1463–7. Karachaliou N, Fernandez-Bruno M, Bracht JWP, Rosell R EGFR first- and second-generation TKIs—there is still place for them in EGFR-mutant NSCLC patients. Transl Cancer Res, 2019, 8:S23–47., DOI 10.21037/tcr.2018.10.06 Ko HW, Shie SS, Wang CW, Chiu CT, Wang CL, Yang TY, et al. Association of smoking status with non-small cell lung cancer patients harboring uncommon epidermal growth factor receptor mutation. Front Immunol, 2022, 13:1011092., DOI 10.3389/fimmu.2022.1011092 Malapelle U, Pilotto S, Passiglia F, Pepe F, Pisapia P, Righi L, et al. Dealing with NSCLC EGFR mutation testing and treatment: a comprehensive review with an Italian real-world perspective. Crit Rev Oncol Hematol, 2021, 160:103300., DOI 10.1016/j.critrevonc.2021.103300 Fukuoka M, Wu Y-L, Thongprasert S, Sunpaweravong P, Leong S-S, Sriuranpong V, et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/ paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia, IPASS). J Clin Oncol, 2011, 29:2866–74., DOI 10.1200/JCO.2010.33.4235 Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutationpositive non-small-cell lung cancer, OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol, 2011, 12:735–42., DOI 10. 1016/S1470-2045(11)70184-X Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer, EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol, 2012, 13:239–46., DOI 10.1016/S1470-2045(11)70393-X Zhou C, Wu Y-L, Chen G, Feng J, Liu X, Wang C, et al. Efficacy results from the randomised phase III OPTIMAL, CTONG 0802, study comparing first-line erlotinib versus carboplatin, CBDCA, plus gemcitabine, GEM, in Chinese advanced non small cell lung cancer, NSCLC, patients, PTS, with EGFR activating mutations. Ann Oncol, 2010, 21(supp.8):LBA13., DOI 10.1093/annonc/ mdq601 Chen KL, Lin CC, Cho YT, Yang CW, Sheen YS, Tsai HE, et al. Comparison of skin toxic effects associated with gefitinib, erlotinib, or afatinib treatment for nonsmall cell lung cancer. JAMA Dermatol, 2016, 152:340–2., DOI 10.1001/ jamadermatol.2015.4448 Cardenas-Fernandez D, Soberanis Pina P, Turcott JG, Chavez-Tapia N, Conde-Flores E, Cardona AF, et al. Management of diarrhea induced by EGFRTKIs in advanced lung adenocarcinoma. Ther Adv Med Oncol, 2023, 15: 17588359231192396., DOI 10.1177/17588359231192396 Nakagawa K, Garon EB, Seto T, Nishio M, Ponce Aix S, Paz-Ares L, et al. Ramucirumab plus erlotinib in patients with untreated, EGFR-mutated, advanced non-small-cell lung cancer, RELAY): a randomised, double-blind, placebocontrolled, phase 3 trial. Lancet Oncol, 2019, 20:1655–69., DOI 10.1016/S1470- 2045(19)30634-5 Sequist LV, Yang JC-H, Yamamoto N, O’Byrne K, Hirsh V, Mok T, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol, 2013, 31:3327–34., DOI 10.1200/JCO.2012.44.2806 Wu Y-L, Zhou C, Hu C-P, Feng J, Lu S, Huang Y, et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations, LUX-Lung 6): an openlabel, randomised phase 3 trial. Lancet Oncol, 2014, 15:213–22., DOI 10.1016/S1470- 2045(13)70604-1 Yang JC-H, Wu Y-L, Schuler M, Sebastian M, Popat S, Yamamoto N, et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma, LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol, 2015, 16:141–51., DOI 10.1016/ S1470-2045(14)71173-8 Park K, Tan E-H, O’Byrne K, Zhang L, Boyer M, Mok T, et al. Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-smallcell lung cancer, LUX-Lung 7): a phase 2B, open-label, randomised controlled trial. Lancet Oncol, 2016, 17:577–89., DOI 10.1016/S1470-2045(16)30033-X Wu Y-L, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer, ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol, 2017, 18:1454–66., DOI 10.1016/S1470-2045(17)30608-3 Mok TS, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, et al. Improvement in overall survival in a randomized study that compared dacomitinib with gefitinib in patients with advanced non-small-cell lung cancer and EGFR-activating mutations. J Clin Oncol, 2018, 36:2244–50., DOI 10.1200/JCO.2018.78.7994 He J, Huang Z, Han L, Gong Y, Xie C Mechanisms and management of 3rdgeneration EGFR-TKI resistance in advanced non-small cell lung cancer, Review). Int J Oncol, 2021, 59:90., DOI 10.3892/ijo.2021.5270 Papadimitrakopoulou VA, Mok TS, Han J-Y, Ahn M-J, Delmonte A, Ramalingam SS, et al. Osimertinib versus platinum-pemetrexed for patients with EGFR T790M advanced NSCLC and progression on a prior EGFR-tyrosine kinase inhibitor: AURA3 overall survival analysis. Ann Oncol, 2020, 31:1536–44., DOI 10.1016/j.annonc.2020.08.2100 Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, Ohe Y, et al. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med, 2020, 382:41–50., DOI 10.1056/NEJMoa1913662 Belani N, Liang K, Fradley M, Judd J, Borghaei H How to treat EGFR-mutated non–small cell lung cancer. JACC CardioOncology, 2023, 5:542–5., DOI 10.1016/j. jaccao.2023.04.005 Vaid AK, Gupta A, Momi G Overall survival in stage IV EGFR mutationpositive NSCLC: comparing first-second- and third-generation EGFR-TKIs, Review). Int J Oncol, 2021, 58:171–84., DOI 10.3892/ijo.2021.5168 Tatineni V, O’Shea PJ, Ozair A, Khosla AA, Saxena S, Rauf Y, et al. Firstversus third-generation EGFR tyrosine kinase inhibitors in EGFR-mutated nonsmall cell lung cancer patients with brain metastases. Cancers, 2023, 15:2382., DOI 10.3390/cancers15082382 Nogami N, Barlesi F, Socinski MA, Reck M, Thomas CA, Cappuzzo F, et al. IMpower150 final exploratory analyses for atezolizumab plus bevacizumab and chemotherapy in key NSCLC patient subgroups with EGFR mutations or metastases in the liver or brain. J Thorac Oncol, 2022, 17:309–23., DOI 10.1016/j. jtho.2021.09.014 Bai Q, Wang J, Zhou X EGFR exon20 insertion mutations in non-small cell lung cancer: clinical implications and recent advances in targeted therapies. Cancer Treat Rev, 2023, 120:102605., DOI 10.1016/j.ctrv.2023.102605 Passaro A, Mok T, Peters S, Popat S, Ahn M-J, de Marinis F Recent advances on the role of EGFR tyrosine kinase inhibitors in the management of NSCLC with uncommon, non exon 20 insertions, EGFR mutations. J Thorac Oncol, 2021, 16: 764–73., DOI 10.1016/j.jtho.2020.12.002 Zugazagoitia J, Ramos I, Trigo JM, Palka M, Gomez-Rueda A, Jantus-Lewintre E, et al. Clinical utility of plasma-based digital next-generation sequencing in patients with advance-stage lung adenocarcinomas with insufficient tumor samples for tissue genotyping. Ann Oncol, 2019, 30:290–6., DOI 10.1093/annonc/mdy512 Weickhardt AJ,Aisner DL, FranklinWA, Varella-Garcia M, Doebele RC, Camidge DR Diagnostic assays for identification of anaplastic lymphoma kinase-positive nonsmall cell lung cancer. Cancer, 2013, 119:1467–77., DOI 10.1002/cncr.27913 LiW, Zhang J, Guo L, Chuai S, Shan L, Ying J Combinational analysis of FISH and immunohistochemistry reveals rare genomic events in ALK fusion patterns in NSCLC that responds to crizotinib treatment. J Thorac Oncol, 2017, 12:94–101., DOI 10.1016/j.jtho.2016.08.145 Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, Costa DB, Heist RS, et al. Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol, 2009, 27:4247–53., DOI 10.1200/JCO.2009.22.6993 National Institutes of Health. Cancer of the lung and bronchus - cancer stat facts, 2024). Available from: https://seer.cancer.gov/statfacts/html/lungb.html, Accessed January 24, 2024). Solomon BJ, Mok T, Kim D-W,Wu Y-L, Nakagawa K, Mekhail T, et al. Firstline crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med, 2014, 371:2167–77., DOI 10.1056/NEJMoa1408440 Soria J-C, Tan DSW, Chiari R, Wu Y-L, Paz-Ares L, Wolf J, et al. First-line ceritinib versus platinum-based chemotherapy in advanced ALK-rearranged non-small-cell lung cancer, ASCEND-4): a randomised, open-label, phase 3 study. Lancet Lond Engl, 2017, 389:917–29., DOI 10.1016/S0140-6736(17, 30123-X Camidge DR, Dziadziuszko R, Peters S, Mok T, Noe J, Nowicka M, et al. Updated efficacy and safety data and impact of the EML4-ALK fusion variant on the efficacy of alectinib in untreated ALK-positive advanced non-small cell lung cancer in the global phase III ALEX study. J Thorac Oncol, 2019, 14:1233–43., DOI 10.1016/ j.jtho.2019.03.007 Camidge DR, Kim HR, Ahn M-J, Yang JCH, Han J-Y, Hochmair MJ, et al. Brigatinib versus crizotinib in ALK inhibitor-naive advanced ALK-positive NSCLC: final results of phase 3 ALTA-1L trial. J Thorac Oncol, 2021, 16:2091–108., DOI 10. 1016/j.jtho.2021.07.035 Horn L, Wang Z, Wu G, Poddubskaya E, Mok T, Reck M, et al. Ensartinib vs crizotinib for patients with anaplastic lymphoma kinase-positive non-small cell lung cancer: a randomized clinical trial. JAMA Oncol, 2021, 7:1617–25., DOI 10. 1001/jamaoncol.2021.3523 Shaw AT, Bauer TM, de Marinis F, Felip E, Goto Y, Liu G, et al. First-line lorlatinib or crizotinib in advanced ALK-positive lung cancer. N Engl J Med, 2020, 383:2018–29., DOI 10.1056/NEJMoa2027187 Mok T, Camidge DR, Gadgeel SM, Rosell R, Dziadziuszko R, Kim D-W, et al. Updated overall survival and final progression-free survival data for patients with treatment-naive advanced ALK-positive non-small-cell lung cancer in the ALEX study. Ann Oncol, 2020, 31:1056–64., DOI 10.1016/j.annonc.2020.04.478 Solomon BJ, Bauer TM, Ignatius Ou S-H, Liu G, HayashiH, Bearz A, et al. Post hoc analysis of lorlatinib intracranial efficacy and safety in patients with ALKpositive advanced non-small-cell lung cancer from the phase III CROWN study. J Clin Oncol, 2022, 40:3593–602., DOI 10.1200/JCO.21.02278 EMA. Summary of the risk management plan for Lorviqua, lorlatinib,, 2024). Available from: https://www.ema.europa.eu/en/documents/rmp-summary/lorviquaepar- risk-management-plan-summary_en.pdf, Accessed January 31, 2024). Novello S, Mazieres J, Oh I-J, de Castro J, Migliorino MR, Helland A, et al. Alectinib versus chemotherapy in crizotinib-pretreated anaplastic lymphoma kinase, ALK)-positive non-small-cell lung cancer: results from the phase III ALUR study. Ann Oncol, 2018, 29:1409–16., DOI 10.1093/annonc/mdy121 Huber RM, Hansen KH, Paz-Ares Rodriguez L, West HL, Reckamp KL, Leighl NB, et al. Brigatinib in crizotinib-refractory ALK+ NSCLC: 2-year follow-up on systemic and intracranial outcomes in the phase 2 ALTA trial. J Thorac Oncol, 2020, 15:404–15., DOI 10.1016/j.jtho.2019.11.004 Shaw AT, Kim TM, Crino L, Gridelli C, Kiura K, Liu G, et al. Ceritinib versus chemotherapy in patients with ALK-rearranged non-small-cell lung cancer previously given chemotherapy and crizotinib, ASCEND-5): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol, 2017, 18:874–86., DOI 10. 1016/S1470-2045(17)30339-X Solomon BJ, Besse B, Bauer TM, Felip E, Soo RA, Camidge DR, et al. Lorlatinib in patients with ALK-positive non-small-cell lung cancer: results from a global phase 2 study. Lancet Oncol, 2018, 19:1654–67., DOI 10.1016/S1470-2045(18, 30649-1 Shaw AT, Solomon BJ, Besse B, Bauer TM, Lin CC, Soo RA, et al. ALK resistance mutations and efficacy of lorlatinib in advanced anaplastic lymphoma kinase-positive non-small-cell lung cancer. J Clin Oncol, 2019, 37:1370–9., DOI 10. 1200/JCO.18.02236 Lin JJ, Zhu VW, Schoenfeld AJ, Yeap BY, Saxena A, Ferris LA, et al. Brigatinib in patients with alectinib-refractory ALK-positive NSCLC. J Thorac Oncol, 2018, 13:1530–8., DOI 10.1016/j.jtho.2018.06.005 Yoshida H, Kim YH, Ozasa H, Sakamori Y, Tsuji T, Nomizo T, et al. Efficacy of ceritinib after alectinib for ALK-positive non-small cell lung cancer. Vivo, 2018, 32:1587–90., DOI 10.21873/invivo.11418 Lin JJ, Schoenfeld AJ, Zhu VW, Yeap BY, Chin E, Rooney M, et al. Efficacy of platinum/pemetrexed combination chemotherapy in ALK-positive NSCLC refractory to second-generation ALK inhibitors. J Thorac Oncol, 2020, 15: 258–65., DOI 10.1016/j.jtho.2019.10.014 Socinski MA, Jotte RM, Cappuzzo F, Orlandi FJ, Stroyakovskiy D, Nogami N, et al. Overall survival, OS, analysis of IMpower150, a randomized Ph 3 study of atezolizumab, atezo, + chemotherapy, chemo, ± bevacizumab, bev, vs chemo + bev in 1L nonsquamous, NSQ, NSCLC. J Clin Oncol, 2018, 36:9002., DOI 10.1200/ JCO.2018.36.15_suppl.9002 Singh K, Saxena S, Khosla AA, McDermott MW, Kotecha RR, Ahluwalia MS Update on the management of brain metastasis. Neurotherapeutics, 2022, 19: 1772–81., DOI 10.1007/s13311-022-01312-w Moldvay J, Timar J KRASG12C mutant lung adenocarcinoma: unique biology, novel therapies and new challenges. Pathol Oncol Res, 2024, 29: 1611580., DOI 10.3389/pore.2023.1611580 Shepherd FA, Domerg C, Hainaut P, Janne PA, Pignon J-P, Graziano S, et al. Pooled analysis of the prognostic and predictive effects of KRAS mutation status and KRAS mutation subtype in early-stage resected non-small-cell lung cancer in four trials of adjuvant chemotherapy. J Clin Oncol, 2013, 31:2173–81., DOI 10.1200/ JCO.2012.48.1390 Nakajima EC, Drezner N, Li X, Mishra-Kalyani PS, Liu Y, Zhao H, et al. FDA approval summary: sotorasib for KRAS G12C-Mutated metastatic NSCLC. Clin Cancer Res, 2022, 28(8):1482–1486., DOI 10.1158/1078-0432.CCR-21-3074 de Langen AJ, Johnson ML, Mazieres J, Dingemans A-MC, Mountzios G, Pless M, et al. Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRASG12C mutation: a randomised, open-label, phase 3 trial. Lancet Lond Engl, 2023, 401:733–46., DOI 10.1016/S0140-6736(23)00221-0 Dy GK, Govindan R, Velcheti V, Falchook GS, Italiano A, Wolf J, et al. Longterm outcomes and molecular correlates of sotorasib efficacy in patients with pretreated KRAS G12C-mutated non-small-cell lung cancer: 2-year analysis of CodeBreaK 100. J Clin Oncol, 2023, 41:3311–7., DOI 10.1200/JCO.22.02524 ESMO. EMA recommends granting a conditional marketing authorisation for adagrasib following a Re-examination procedure, 2023). Available from: https:// www.esmo.org/oncology-news/ema-recommends-granting-a-conditionalmarketing- authorisation-for-adagrasib-following-a-re-examination-procedure, Accessed January 24, 2024). Janne PA, Riely GJ, Gadgeel SM, Heist RS, Ou S-HI, Pacheco JM, et al. Adagrasib in non-small-cell lung cancer harboring a KRASG12C mutation. N Engl J Med, 2022, 387:120–31., DOI 10.1056/NEJMoa2204619 Birchmeier C, Sharma S, Wigler M Expression and rearrangement of the ROS1 gene in human glioblastoma cells. Proc Natl Acad Sci U S A, 1987, 84:9270–4., DOI 10.1073/pnas.84.24.9270 Drilon A, Jenkins C, Iyer S, Schoenfeld A, Keddy C, Davare MA ROS1- dependent cancers - biology, diagnostics and therapeutics. Nat Rev Clin Oncol, 2021, 18:35–55., DOI 10.1038/s41571-020-0408-9 Lin JJ, Ritterhouse LL, Ali SM, Bailey M, Schrock AB, Gainor JF, et al. ROS1 fusions rarely overlap with other oncogenic drivers in non-small cell lung cancer. J Thorac Oncol, 2017, 12:872–7., DOI 10.1016/j.jtho.2017.01.004 Gendarme S, Bylicki O, Chouaid C, Guisier F ROS-1 fusions in non-small-cell lung cancer: evidence to date. Curr Oncol Tor Ont, 2022, 29:641–58., DOI 10.3390/ curroncol29020057 Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, et al. Updated molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors: guideline from the college of American pathologists, the international association for the study of lung cancer, and the association for molecular pathology. Arch Pathol Lab Med, 2018, 142: 321–46., DOI 10.5858/arpa.2017-0388-CP Shaw AT, Riely GJ, Bang YJ, Kim DW, Camidge DR, Solomon BJ, et al. Crizotinib in ROS1-rearranged advanced non-small-cell lung cancer, NSCLC): updated results, including overall survival, from PROFILE 1001. Ann Oncol, 2019, 30:1121–6., DOI 10.1093/annonc/mdz131 Fischer H, Ullah M, de la Cruz CC, Hunsaker T, Senn C, Wirz T, et al. Entrectinib, a TRK/ROS1 inhibitor with anti-CNS tumor activity: differentiation from other inhibitors in its class due to weak interaction with P-glycoprotein. Neuro-Oncol, 2020, 22:819–29., DOI 10.1093/neuonc/noaa052 Drilon A, Siena S, Dziadziuszko R, Barlesi F, Krebs MG, Shaw AT, et al. Entrectinib in ROS1 fusion-positive non-small-cell lung cancer: integrated analysis of three phase 1-2 trials. Lancet Oncol, 2020, 21:261–70., DOI 10.1016/S1470- 2045(19)30690-4 Dziadziuszko R, Krebs MG, De Braud F, Siena S, Drilon A, Doebele RC, et al. Updated integrated analysis of the efficacy and safety of entrectinib in locally advanced or metastatic ROS1 fusion-positive non-small-cell lung cancer. J Clin Oncol, 2021, 39:1253–63., DOI 10.1200/JCO.20.03025 Lim SM, Kim HR, Lee J-S, Lee KH, Lee Y-G, Min YJ, et al. Open-label, multicenter, phase II study of ceritinib in patients with non-small-cell lung cancer harboring ROS1 rearrangement. J Clin Oncol, 2017, 35:2613–8., DOI 10.1200/JCO. 2016.71.3701 Shaw AT, Solomon BJ, Chiari R, Riely GJ, Besse B, Soo RA, et al. Lorlatinib in advanced ROS1-positive non-small-cell lung cancer: a multicentre, open-label, single-arm, phase 1-2 trial. Lancet Oncol, 2019, 20:1691–701., DOI 10.1016/ S1470-2045(19)30655-2 Lin JJ, Drilon AE, Cho BC, Felip E, De Langen A, Yang N, et al. Intracranial and systemic efficacy of repotrectinib in advanced ROS1 fusion-positive, ROS1+, non-small cell lung cancer, NSCLC, and central nervous system metastases, CNS mets, in the phase 1/2 TRIDENT-1. J Clin Oncol, 2023, 41:9017., DOI 10.1200/JCO. 2023.41.16_suppl.9017 Yun MR, Kim DH, Kim SY, Joo HS, Lee YW, Choi HM, et al. Repotrectinib exhibits potent antitumor activity in treatment-naive and solvent-front-mutant ROS1-rearranged non-small cell lung cancer. Clin Cancer Res, 2020, 26:3287–95., DOI 10.1158/1078-0432.CCR-19-2777 FDA. FDA approves repotrectinib for ROS1-positive non-small cell lung cancer, 2023). Available from: https://www.fda.gov/drugs/resources-informationapproved- drugs/fda-approves-repotrectinib-ros1-positive-non-small-cell-lungcancer, Accessed January 24, 2024). Dudnik E, Agbarya A, Grinberg R, Cyjon A, Bar J,Moskovitz M, et al. Clinical activity of brigatinib in ROS1-rearranged non-small cell lung cancer. Clin Transl Oncol, 2020, 22:2303–11., DOI 10.1007/s12094-020-02376-w Katayama R, Gong B, Togashi N, Miyamoto M, Kiga M, Iwasaki S, et al. The new-generation selective ROS1/NTRK inhibitor DS-6051b overcomes crizotinib resistant ROS1-G2032R mutation in preclinical models. Nat Commun, 2019, 10: 3604., DOI 10.1038/s41467-019-11496-z Papadopoulos KP, Borazanci E, Shaw AT, Katayama R, Shimizu Y, Zhu VW, et al. U.S. Phase I first-in-human study of taletrectinib, DS-6051b/AB-106), a ROS1/TRK inhibitor, in patients with advanced solid tumors. Clin Cancer Res, 2020, 26:4785–94., DOI 10.1158/1078-0432.CCR-20-1630 Ai X,Wang Q, Cheng Y, Liu X, Cao L, Chen J, et al. Safety but limited efficacy of ensartinib in ROS1-positive NSCLC: a single-arm, multicenter phase 2 study. J Thorac Oncol, 2021, 16:1959–63., DOI 10.1016/j.jtho.2021.06.023 Mulligan LM RET revisited: expanding the oncogenic portfolio. Nat Rev Cancer, 2014, 14:173–86., DOI 10.1038/nrc3680 Ferrara R, Auger N, Auclin E, Besse B Clinical and translational implications of RET rearrangements in non-small cell lung cancer. J Thorac Oncol, 2018, 13: 27–45., DOI 10.1016/j.jtho.2017.10.021 Mulligan LM, Kwok JB, Healey CS, Elsdon MJ, Eng C, Gardner E, et al. Germline mutations of the RET proto-oncogene in multiple endocrine neoplasia type 2A. Nature, 1993, 363:458–60., DOI 10.1038/363458a0 Romeo G, Ronchetto P, Luo Y, Barone V, Seri M, Ceccherini I, et al. Point mutations affecting the tyrosine kinase domain of the RET proto-oncogene in Hirschsprung’s disease. Nature, 1994, 367:377–8., DOI 10.1038/367377a0 Drilon A, Lin JJ, Filleron T, Ni A, Milia J, Bergagnini I, et al. Frequency of brain metastases and multikinase inhibitor outcomes in patients with RETrearranged lung cancers. J Thorac Oncol, 2018, 13:1595–601., DOI 10.1016/j.jtho. 2018.07.004 Feng J, Li Y,Wei B, Guo L, Li W, Xia Q, et al. Clinicopathologic characteristics and diagnostic methods of RET rearrangement in Chinese non-small cell lung cancer patients. Transl Lung Cancer Res, 2022, 11:617–31., DOI 10.21037/tlcr-22-202 Andrini E, Mosca M, Galvani L, Sperandi F, Ricciuti B, Metro G, et al. Nonsmall- cell lung cancer: how to manage RET-positive disease. Drugs Context, 2022, 11:1–12., DOI 10.7573/dic.2022-1-5 Drilon A, Oxnard GR, Tan DSW, Loong HHF, Johnson M, Gainor J, et al. Efficacy of selpercatinib in RET fusion-positive non-small-cell lung cancer. N Engl J Med, 2020, 383:813–24., DOI 10.1056/NEJMoa2005653 Duke ES, Bradford D, Marcovitz M, Amatya AK, Mishra-Kalyani PS, Nguyen E, et al. FDA approval summary: selpercatinib for the treatment of advanced RET fusion-positive solid tumors. Clin Cancer Res, 2023, 29:3573–8., DOI 10.1158/1078- 0432.CCR-23-0459 Solomon BJ, Zhou CC, Drilon A, Park K, Wolf J, Elamin Y, et al. Phase III study of selpercatinib versus chemotherapy ± pembrolizumab in untreated RET positive non-small-cell lung cancer. Future Oncol Lond Engl, 2021, 17:763–73., DOI 10.2217/fon-2020-0935 Loong HHF, Goto K, Solomon BJ, Park K, Perol M, Arriola E, et al. LBA4 Randomized phase III study of first-line selpercatinib versus chemotherapy and pembrolizumab in RET fusion-positive NSCLC. Ann Oncol, 2023, 34:S1303., DOI 10.1016/j.annonc.2023.10.059 Zhou C, Solomon B, Loong HH, Park K, Perol M, Arriola E, et al. First-line selpercatinib or chemotherapy and pembrolizumab in RET fusion-positive NSCLC. N Engl J Med, 2023, 389:1839–50., DOI 10.1056/NEJMoa2309457 FDA. FDA approves selpercatinib for lung and thyroid cancers with RET gene mutations or fusions, 2021). Available from: https://www.fda.gov/drugs/resourcesinformation- approved-drugs/fda-approves-selpercatinib-lung-and-thyroidcancers- ret-gene-mutations-or-fusions, Accessed January 24, 2024). FDA. FDA approves pralsetinib for lung cancer with RET gene fusions, 2020). Available from: https://www.fda.gov/drugs/resources-information-approveddrugs/ fda-approves-pralsetinib-lung-cancer-ret-gene-fusions, Accessed January 24, 2024). EMA. Selpercatinib - summary of opinion, post authorisation,, 2022). Available from: https://www.ema.europa.eu/en/documents/smop/chmp-postauthorisation- summary-positive-opinion-retsevmo-ii-0014-g_en.pdf, Accessed January 31, 2024). EMA. Pralsetinib - summary of product characteristics, 2024). Available from: https://www.ema.europa.eu/en/documents/product-information/gavreto-eparproduct- information_en.pdf, Accessed January 31, 2024). Griesinger F, Curigliano G, ThomasM, Subbiah V, Baik CS, Tan DSW, et al. Safety and efficacy of pralsetinib in RET fusion-positive non-small-cell lung cancer including as first-line therapy: update from the ARROW trial. Ann Oncol, 2022, 33: 1168–78., DOI 10.1016/j.annonc.2022.08.002 Besse B, Felip E, Kim ES, Clifford C, Louie-Gao M, Yagui-Beltran A, et al. P87.02 AcceleRET lung: a phase 3 study of first-line pralsetinib in patients with RET-fusion+ advanced/metastatic NSCLC. J Thorac Oncol, 2021, 16:S684., DOI 10. 1016/j.jtho.2021.01.1257 Chevallier M, Borgeaud M, Addeo A, Friedlaender A Oncogenic driver mutations in non-small cell lung cancer: past, present and future. World J Clin Oncol, 2021, 12:217–37., DOI 10.5306/wjco.v12.i4.217 Abdayem P, Planchard D Ongoing progress in BRAF-mutated non-small cell lung cancer. Clin Adv Hematol Oncol HO, 2022, 20(1):662–672. O’Leary CG, Andelkovic V, Ladwa R, Pavlakis N, Zhou C, Hirsch F, et al. Targeting BRAF mutations in non-small cell lung cancer. Transl Lung Cancer Res, 2019, 8:1119–24., DOI 10.21037/tlcr.2019.10.22 Harada G, Yang S-R, Cocco E, Drilon A Rare molecular subtypes of lung cancer. Nat Rev Clin Oncol, 2023, 20:229–49., DOI 10.1038/s41571-023-00733-6 Hendriks LE, Kerr KM, Menis J, Mok TS, Nestle U, Passaro A, et al. Nononcogene- addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol Off J Eur Soc Med Oncol, 2023, 34:358–76., DOI 10.1016/j.annonc.2022.12.013 Davies KD, Lomboy A, Lawrence CA, Yourshaw M, Bocsi GT, Camidge DR, et al. DNA-based versus RNA-based detection of MET exon 14 skipping events in lung cancer. J Thorac Oncol, 2019, 14:737–41., DOI 10.1016/j.jtho.2018.12.020 Socinski MA, Pennell NA, Davies KD MET exon 14 skipping mutations in non-small-cell lung cancer: an overview of biology, clinical outcomes, and testing considerations. JCO Precis Oncol, 2021, 5:653–63., DOI 10.1200/PO.20.00516 FDA. List of cleared or approved companion diagnostic devices, in vitro and imaging tools,, 2024). Available from: https://www.fda.gov/medical-devices/invitro- diagnostics/list-cleared-or-approved-companion-diagnostic-devices-invitro- and-imaging-tools, Accessed January 31, 2024). Wolf J, Seto T, Han J-Y, Reguart N, Garon EB, Groen HJM, et al. Capmatinib in MET exon 14-mutated or MET-amplified non-small-cell lung cancer. N Engl J Med, 2020, 383:944–57., DOI 10.1056/NEJMoa2002787 ClinicalTrials.gov. Clinical study of oral cMET inhibitor INC280 in adult patients with EGFR wild-type advanced non-small cell lung cancer, geometry mono-1,, 2023). Available from: https://clinicaltrials.gov/study/NCT02414139, Accessed January 31, 2024). Le X, Sakai H, Felip E, Veillon R, Garassino MC, Raskin J, et al. Tepotinib efficacy and safety in patients with MET exon 14 skipping NSCLC: outcomes in patient subgroups from the VISION study with relevance for clinical practice. Clin Cancer Res, 2022, 28:1117–26., DOI 10.1158/1078-0432.CCR-21-2733 ClinicalTrials.gov. Tepotinib phase II in NSCLC harboring MET alterations, VISION,, 2023). Available from: https://clinicaltrials.gov/study/NCT02864992, Accessed January 31, 2024). Paik PK, Felip E, Veillon R, Sakai H, Cortot AB, Garassino MC, et al. Tepotinib in non-small-cell lung cancer with MET exon 14 skipping mutations. N Engl J Med, 2020, 383:931–43., DOI 10.1056/NEJMoa2004407 Xiong W, Papasouliotis O, Jonsson EN, Strotmann R, Girard P Population pharmacokinetic analysis of tepotinib, an oral MET kinase inhibitor, including data from the VISION study. Cancer Chemother Pharmacol, 2022, 89:655–69., DOI 10. 1007/s00280-022-04423-5 Jebbink M, de Langen AJ, Boelens MC, Monkhorst K, Smit EF The force of HER2 - a druggable target in NSCLC? Cancer Treat Rev, 2020, 86:101996., DOI 10. 1016/j.ctrv.2020.101996 Li BT, Smit EF, Goto Y, Nakagawa K, Udagawa H, Mazieres J, et al. Trastuzumab deruxtecan in HER2-mutant non-small-cell lung cancer. N Engl J Med, 2022, 386:241–51., DOI 10.1056/NEJMoa2112431 Goto K, Goto Y, Kubo T, Ninomiya K, Kim S-W, Planchard D, et al. Trastuzumab deruxtecan in patients with HER2-mutant metastatic non-small-cell lung cancer: primary results from the randomized, phase II DESTINY-lung02 trial. J Clin Oncol, 2023, 41:4852–63., DOI 10.1200/JCO.23.01361 ClinicalTrials.gov. A study to investigate the efficacy and safety of trastuzumab deruxtecan as the first treatment option for unresectable, locally advanced/metastatic non-small cell lung cancer with HER2 mutations, 2023). Available from: https://clinicaltrials.gov/study/NCT05048797, Accessed January 31, 2024). Demetri GD, De Braud F, Drilon A, Siena S, Patel MR, Cho BC, et al. Updated integrated analysis of the efficacy and safety of entrectinib in patients with NTRK fusion-positive solid tumors. Clin Cancer Res, 2022, 28:1302–12., DOI 10. 1158/1078-0432.CCR-21-3597 Drilon A, Tan DSW, Lassen UN, Leyvraz S, Liu Y, Patel JD, et al. Efficacy and safety of larotrectinib in patients with tropomyosin receptor kinase fusion-positive lung cancers. JCO Precis Oncol, 2022, 6:e2100418., DOI 10.1200/PO.21.00418 Aggarwal C, Marmarelis ME, Hwang W-T, Scholes DG, McWilliams TL, Singh AP, et al. Association between availability of molecular genotyping results and overall survival in patients with advanced nonsquamous non-small-cell lung cancer. JCO Precis Oncol, 2023, 7:e2300191., DOI 10.1200/PO.23.00191 Aggarwal C, Marmarelis ME, Hwang W-T, Scholes DG, McWilliams T, Singh AP, et al. Association of comprehensive molecular genotyping and overall survival in patients with advanced non-squamous non-small cell lung cancer. J Clin Oncol, 2022, 40:9022., DOI 10.1200/JCO.2022.40.16_suppl.9022 Sequist LV, Neal JV, Jett JR, Ross ME Personalized, genotype-directed therapy for advanced non-small cell lung cancer. UpToDate, 2016). Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N Engl J Med, 2022, 386:1973–85., DOI 10.1056/NEJMoa2202170 Heymach JV, Harpole D, Mitsudomi T, Taube JM, Galffy G, Hochmair M, et al. Perioperative durvalumab for resectable non-small-cell lung cancer. N Engl J Med, 2023, 389:1672–84., DOI 10.1056/NEJMoa2304875 Wu Y-L, Tsuboi M, He J, John T, Grohe C, Majem M, et al. Osimertinib in resected EGFR-mutated non-small-cell lung cancer. N Engl J Med, 2020, 383: 1711–23., DOI 10.1056/NEJMoa2027071 Tsuboi M, Herbst RS, John T, Kato T, Majem M, Grohe C, et al. Overall survival with osimertinib in resected EGFR-mutated NSCLC. N Engl J Med, 2023, 389:137–47., DOI 10.1056/NEJMoa2304594 Solomon BJ, Ahn JS, Dziadziuszko R, Barlesi F, Nishio M, Lee DH, et al. LBA2 ALINA: efficacy and safety of adjuvant alectinib versus chemotherapy in patients with early-stage ALK+ non-small cell lung cancer, NSCLC). Ann Oncol, 2023, 34:S1295–6., DOI 10.1016/j.annonc.2023.10.051 Solomon BJ, Ahn JS, Barlesi F, Dziadziuszko R, Nishio M, Shaw AT, et al. ALINA: a phase III study of alectinib versus chemotherapy as adjuvant therapy in patients with stage IB–IIIA anaplastic lymphoma kinase-positive, ALK+, nonsmall cell lung cancer, NSCLC). J Clin Oncol, 2019, 37:TPS8569., DOI 10.1200/JCO. 2019.37.15_suppl.TPS8569 Gillespie CS, Mustafa MA, Richardson GE, Alam AM, Lee KS, Hughes DM, et al. Genomic alterations and the incidence of brain metastases in advanced and metastatic NSCLC: a systematic review and meta-analysis. J Thorac Oncol, 2023, 18: 1703–13., DOI 10.1016/j.jtho.2023.06.017 Nishino M, Soejima K, Mitsudomi T Brain metastases in oncogene-driven non-small cell lung cancer. Transl Lung Cancer Res, 2019, 8:S298–S307., DOI 10. 21037/tlcr.2019.05.15 Rangachari D, Yamaguchi N, VanderLaan PA, Folch E, Mahadevan A, Floyd SR, et al. Brain metastases in patients with EGFR-mutated or ALK-rearranged nonsmall- cell lung cancers. Lung Cancer Amst Neth, 2015, 88:108–11., DOI 10.1016/j. lungcan.2015.01.020 Tan AC, Tan DSW Targeted therapies for lung cancer patients with oncogenic driver molecular alterations. JCO, 2022, 40:611–625., DOI 10.1200/ JCO.21.01626 De Carlo E, Bertoli E, Del Conte A, Stanzione B, Berto E, Revelant A, et al. Brain metastases management in oncogene-addicted non-small cell lung cancer in the targeted therapies era. Int J Mol Sci, 2022, 23:6477., DOI 10.3390/ ijms23126477 Le Rhun E, Guckenberger M, Smits M, Dummer R, Bachelot T, Sahm F, et al. EANO-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up of patients with brain metastasis from solid tumours. Ann Oncol, 2021, 32:1332–47., DOI 10.1016/j.annonc.2021.07.016 Thomas NJ, Myall NJ, Sun F, Patil T, Mushtaq R, Yu C, et al. Brain metastases in EGFR- and ALK-positive NSCLC: outcomes of central nervous system-penetrant tyrosine kinase inhibitors alone versus in combination with radiation. J Thorac Oncol, 2022, 17:116–29., DOI 10.1016/j.jtho.2021.08.009 NCCN. NCCN guidelines brain metastases version 1, 2023). Available from: https://www.nccn.org/professionals/physician_gls/pdf/cns.pdf, Accessed January 30, 2024). Soria J-C, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, et al. Osimertinib in untreated EGFR-mutated advanced non-smallcell lung cancer. N Engl J Med, 2018, 378:113–25., DOI 10.1056/ NEJMoa1713137 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611715 EP 1611735 DI 10.3389/pore.2024.1611715 PG 21 ER PT J AU Editorial Office, AF Editorial Office, TI Retraction: High expression of long noncoding RNA HOTAIRM1 is associated with the proliferation and migration in pancreatic ductal adenocarcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Retraction AB Following publication, the authors contacted the Editorial Office to request the retraction of the cited article, stating that concerns were raised on the PubPeer platform regarding the reuse of certain images. The authors have stated that some of the experiments were conducted by collaboration laboratories and that certain images were mistakenly used. C1 [Editorial Office, ] Pathology & Oncology ResearchBudapest, Hungary. RP Editorial Office, (reprint author), Pathology & Oncology Research, Budapest, Hungary. NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611728 EP 1611729 DI 10.3389/pore.2024.1611728 PG 2 ER PT J AU Batar, P Alizadeh, H Rokszin, Gy Abonyi-Toth, Zs Demeter, J AF Batar, Peter Alizadeh, Hussain Rokszin, Gyorgy Abonyi-Toth, Zsolt Demeter, Judit TI Corrigendum: Comorbidities and outcomes of patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors: a real-world, nationwide, retrospective study from Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE chronic myeloid leukemia; real-word evidence; tyrosine kinase inhibitors; treatment outcome; overall survival ID chronic myeloid leukemia; real-word evidence; tyrosine kinase inhibitors; treatment outcome; overall survival C1 [Batar, Peter] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Alizadeh, Hussain] University of Pecs, I. Department of Internal MedicinePecs, Hungary. [Rokszin, Gyorgy] RxTarget LtdSzolnok, Hungary. [Abonyi-Toth, Zsolt] RxTarget LtdSzolnok, Hungary. [Demeter, Judit] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. RP Batar, P (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM pbatar@med.unideb.hu NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611758 EP 1611759 DI 10.3389/pore.2024.1611758 PG 2 ER PT J AU Editorial Office, AF Editorial Office, TI Retraction: MiR-184 retarded the proliferation, invasiveness and migration of glioblastoma cells by repressing stanniocalcin-2 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Retraction C1 [Editorial Office, ] Pathology & Oncology ResearchBudapest, Hungary. RP Editorial Office, (reprint author), Pathology & Oncology Research, Budapest, Hungary. EM editorialoffice@por-journal.com NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2024 VL 30 IS 1 BP 1611767 EP 1611767 DI 10.3389/pore.2024.1611767 PG 1 ER PT J AU Jakab, A Patai, Darvas, M Tormassi-Bely, K Micsik, T AF Jakab, Anna Patai, V. Arpad Darvas, Monika Tormassi-Bely, Karolina Micsik, Tamas TI Microenvironment, systemic inflammatory response and tumor markers considering consensus molecular subtypes of colorectal cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal cancer; consensus molecular subtypes; tumor microenvironment (TME); tumor-stroma ratio; systemic inflammation ID colorectal cancer; consensus molecular subtypes; tumor microenvironment (TME); tumor-stroma ratio; systemic inflammation AB Introduction: Colorectal carcinomas (CRC) are one of the most frequent malignancies worldwide. Based on gene expression profile analysis, CRCs can be classified into four distinct subtypes also known as the consensus molecular subtypes (CMS), which predict biological behaviour. Besides CMS, several other aspects of tumor microenvironment (TME) and systemic inflammatory response (SIR) influence the outcome of CRC patients. TME and inflammation have important role in the immune (CMS1) and mesenchymal (CMS4) subtypes, however, the relationship between these and systemic inflammation has not been assessed yet. Our objective was to evaluate the connection between CMS, TME and SIR, and to analyze the correlation between these markers and routinely used tumor markers, such as CEA (Carcinoembryonic Antigen) and CA19-9 (Carbohydrate Antigen 19-9). Methods: FFPE (Formalin Fixed Paraffin Embedded) samples of 185 CRC patients were collected. TME was described using tumor-stroma ratio (TSR), Klintrup-Makinen (KM) grade, and Glasgow Microenvironment Score (GMS). CMS classification was performed on tissue microarray using MLH1, PMS2, MSH2 and MSH6, and pan-cytokeratin, CDX2, FRMD6, HTR2B and ZEB1 immunohistochemical stains. Pre-operative tumor marker levels and inflammatory markers [C-reactive protein - CRP, albumin, absolute neutrophil count (ANC), absolute lymphocyte count (ALC), absolute platelet count (APC)] and patient history were retrieved using MedSolution database. Results: Amongst TME-markers, TSR correlated most consistently with adverse clinicopathological features (p < 0.001) and overall survival (p < 0.001). Elevated CRP and modified Glasgow Prognostic Score (mGPS) were associated with worse outcome and aggressive phenotype, similarly to tumor markers CEA and CA19-9. Stroma–Tumor Marker score (STM score), a new combined score of CA19-9 and TSR delivered the second best prognostication after mGPS. Furthermore, CMS4 showed association with TSR and several laboratory markers (albumin and platelet derived factors), but not with other SIR descriptors. CMS did not show any association with CEA and CA19-9 tumor markers. Conclusion: More routinely available TME, SIR and tumor markers alone and in combination deliver reliable prognostic data for choosing the patients with higher risk for propagation. CMS4 is linked with high TSR and poor prognosis, but in overall, CMS-classification showed only limited effect on SIR- and tumor-markers. C1 [Jakab, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Patai, V. Arpad] Semmelweis University, Interdisciplinary Gastroenterology Working GroupBudapest, Hungary. [Darvas, Monika] Semmelweis University, Interdisciplinary Gastroenterology Working GroupBudapest, Hungary. [Tormassi-Bely, Karolina] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Micsik, Tamas] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Jakab, A (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM annajakab272@gmail.com CR WHO. Colorectal cancer, 2023). Available at: https://www.who.int/news-room/ fact-sheets/detail/colorectal-cancer?gad_source=1&gclid=CjwKCAjwzNvBhAkEiwAYiO7oDbqYfWJo7SRc 4nb rjuz_2OyLP 0 z 0 d 5 Z 9 b u - ez8ShQKny8ZkUyhi4xoCBucQAvD_BwE. Di Nicolantonio F, Vitiello PP, Marsoni S, Siena S, Tabernero J, Trusolino L, et al. Precision oncology in metastatic colorectal cancer — from biology to medicine. Nat Rev Clin Oncol, 2021, 18(8):506–25., DOI 10.1038/s41571-021- 00495-z Kobayashi H, Enomoto A, Woods SL, Burt AD, Takahashi M, Worthley DL. Cancer-associated fibroblasts in gastrointestinal cancer. Nat Rev Gastroenterol Hepatol, 2019, 16(5):282–95., DOI 10.1038/s41575-019-0115-0 Schurch CM, Bhate SS, Barlow GL, Phillips DJ, Noti L, Zlobec I, et al. Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front. Cell, 2020, 182(5):1341–59., DOI 10.1016/j.cell. 2020.07.005 Wilkinson K, Ng W, Roberts TL, Becker TM, Lim SHS, ChuaW, et al. Tumour immune microenvironment biomarkers predicting cytotoxic chemotherapy efficacy in colorectal cancer. J Clin Pathol, 2021, 74(10):625–34., DOI 10.1136/jclinpath- 2020-207309 Klintrup K, Makinen JM, Kauppila S, Vare PO, Melkko J, Tuominen H, et al. Inflammation and prognosis in colorectal cancer. Eur J Cancer, 2005, 41(17): 2645–54., DOI 10.1016/j.ejca.2005.07.017 Jakubowska K, Kisielewski W, Kanczuga-Koda L, Koda M, Famulski W. Diagnostic value of inflammatory cell infiltrates, tumor stroma percentage and disease-free survival in patients with colorectal cancer. Oncol Lett, 2017, 14(3): 3869–77., DOI 10.3892/ol.2017.6639 van Pelt GW, Kjaer-Frifeldt S, van Krieken JHJM, Al Dieri R, Morreau H, Tollenaar RAEM, et al. Scoring the tumor-stroma ratio in colon cancer: procedure and recommendations. Virchows Arch, 2018, 473(4):405–12., DOI 10.1007/s00428- 018-2408-z Mesker WE, Junggeburt JMC, Szuhai K, de Heer P, Morreau H, Tanke HJ, et al. The carcinoma-stromal ratio of colon carcinoma is an independent factor for survival compared to lymph node status and tumor stage. Cell Oncol, 2007, 29(5): 387–98., DOI 10.1155/2007/175276 Huijbers A, Tollenaar RAEM, v Pelt GW, Zeestraten ECM, Dutton S, McConkey CC, et al. The proportion of tumor-stroma as a strong prognosticator for stage II and III colon cancer patients: validation in the VICTOR trial. Ann Oncol, 2013, 24(1):179–85., DOI 10.1093/annonc/mds246 Hansen TF, Kjaer-Frifeldt S, Lindebjerg J, Rafaelsen SR, Jensen LH, Jakobsen A, et al. Tumor-stroma ratio predicts recurrence in patients with colon cancer treated with neoadjuvant chemotherapy. Acta Oncol, 2018, 57(4):528–33., DOI 10. 1080/0284186x.2017.1385841 Park JH, McMillan DC, Edwards J, Horgan PG, Roxburgh CSD. Comparison of the prognostic value of measures of the tumor inflammatory cell infiltrate and tumor-associated stroma in patients with primary operable colorectal cancer. Oncoimmunology, 2016, 5(3):e1098801., DOI 10.1080/2162402x.2015.1098801 Alexander PG, Roseweir AK, Pennel KAF, van Wyk HC, Powell AGMT, McMillan DC, et al. The Glasgow Microenvironment Score associates with prognosis and adjuvant chemotherapy response in colorectal cancer. Br J Cancer, 2021, 124(4):786–96., DOI 10.1038/s41416-020-01168-x Guinney J, Dienstmann R, Wang X, de Reynies A, Schlicker A, Soneson C, et al. The consensus molecular subtypes of colorectal cancer. Nat Med, 2015, 21(11):1350–6., DOI 10.1038/nm.3967 Becht E, de Reynies A, Giraldo NA, Pilati C, Buttard B, Lacroix L, et al. Immune and stromal classification of colorectal cancer is associated with molecular subtypes and relevant for precision immunotherapy. Clin Cancer Res, 2016, 22(16): 4057–66., DOI 10.1158/1078-0432.ccr-15-2879 Dienstmann R, Vermeulen L, Guinney J, Kopetz S, Tejpar S, Tabernero J. Consensusmolecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer, 2017, 17(2):79–92., DOI 10.1038/nrc.2016.126 Roepman P, Schlicker A, Tabernero J, Majewski I, Tian S, Moreno V, et al. Colorectal cancer intrinsic subtypes predict chemotherapy benefit, deficient mismatch repair and epithelial-to-mesenchymal transition. Int J Cancer, 2014, 134(3):552–62., DOI 10.1002/ijc.28387 De Sousa E Melo F, Wang X, Jansen M, Fessler E, Trinh A, de Rooij LPMH, et al. Poor-prognosis colon cancer is defined by a molecularly distinct subtype and develops from serrated precursor lesions. NatMed, 2013, 19(5):614–8., DOI 10.1038/ nm.3174 Golder AM, McMillan DC, Park JH, Mansouri D, Horgan PG, Roxburgh CS. The prognostic value of combined measures of the systemic inflammatory response in patients with colon cancer: an analysis of 1700 patients. Br J Cancer, 2021, 124(11):1828–35., DOI 10.1038/s41416-021-01308-x Park JH, Fuglestad AJ, Kostner AH, Oliwa A, Graham J, Horgan PG, et al. Systemic inflammation and outcome in 2295 patients with stage I–III colorectal cancer from scotland and Norway: first results from the ScotScan colorectal cancer group. Ann Surg Oncol, 2020, 27(8):2784–94., DOI 10.1245/s10434-020- 08268-1 ClimentM, Ryan EJ, Stakelum A, Khaw YL, Creavin B, Lloyd A, et al. Systemic inflammatory response predicts oncological outcomes in patients undergoing elective surgery for mismatch repair-deficient colorectal cancer. Int J Colorectal Dis, 2019, 34(6):1069–78., DOI 10.1007/s00384-019-03274-6 Park JH, Watt G, Roxburgh CSD, Horgan PG, McMillan DC Colorectal cancer, systemic inflammation, and outcome. Ann Surg, 2016, 263(2):326–36., DOI 10.1097/sla.0000000000001122 Yamamoto T, Kawada K, Obama K. Inflammation-related biomarkers for the prediction of prognosis in colorectal cancer patients. Int J Mol Sci, 2021, 22(15): 8002., DOI 10.3390/ijms22158002 Watt DG, Proctor MJ, Park JH, Horgan PG, McMillan DC. The neutrophilplatelet score, NPS, predicts survival in primary operable colorectal cancer and a variety of common cancers. PLOS ONE, 2015, 10(11):e0142159., DOI 10.1371/ journal.pone.0142159 Yakabe T, Nakafusa Y, Sumi K, Miyoshi A, Kitajima Y, Sato S, et al. Clinical significance of CEA and CA19-9 in postoperative follow-up of colorectal cancer. Ann Surg Oncol, 2010, 17(9):2349–56., DOI 10.1245/s10434-010-1004-5 Liu J-M, Wang YY, Liu W, Xu D, Wang K, Xing BC. Preoperative CA19-9: a competitive predictor of recurrence in patients with colorectal cancer liver metastases after hepatectomy. Int J Colorectal Dis, 2021, 36(4):767–78., DOI 10. 1007/s00384-020-03828-z Yang SH, Jiang JK, Chang SC, Juang CJ, Lin JK. Clinical significance of CA19-9 in the follow-up of colorectal cancer patients with elevated preoperative serum CA19-9. Hepatogastroenterology, 2013, 60(125):1021–7., DOI 10.5754/ hge121151 Zhang SY, Lin M, Zhang HB. Diagnostic value of carcinoembryonic antigen and carcinoma antigen 19-9 for colorectal carcinoma. Int J Clin Exp Pathol, 2015, 8(8):9404–9. Baqar AR, Wilkins S, Staples M, Angus Lee CH, Oliva K, McMurrick P. The role of preoperative CEA in the management of colorectal cancer: a cohort study from two cancer centres. Int J Surg, 2019, 64:10–5., DOI 10.1016/j.ijsu.2019.02.014 Jia J, Zhang P, Gou M, Yang F, Qian N, Dai G. The role of serum CEA and CA19-9 in efficacy evaluations and progression-free survival predictions for patients treated with cetuximab combined with FOLFOX4 or FOLFIRI as a first-line treatment for advanced colorectal cancer. Dis markers, 2019, 2019: 6812045., DOI 10.1155/2019/6812045 Nowak JA, Hornick JL. Molecular evaluation of colorectal adenocarcinoma: current practice and emerging concepts. Surg Pathol Clin, 2016, 9(3):427–39., DOI 10.1016/j.path.2016.04.007 Ten Hoorn S, Trinh A, de Jong J, Koens L, Vermeulen L. Classification of colorectal cancer in molecular subtypes by immunohistochemistry. Methods Mol Biol, 2018, 1765:179–91., DOI 10.1007/978-1-4939-7765-9_11 Trinh A, Trumpi K, De Sousa E Melo F, Wang X, de Jong JH, Fessler E, et al. Practical and robust identification of molecular subtypes in colorectal cancer by immunohistochemistry. Clin Cancer Res, 2017, 23(2):387–98., DOI 10.1158/1078- 0432.Ccr-16-0680 Park JH, Richards CH, McMillan DC, Horgan PG, Roxburgh CSD. The relationship between tumour stroma percentage, the tumour microenvironment and survival in patients with primary operable colorectal cancer. Ann Oncol, 2014, 25(3):644–51., DOI 10.1093/annonc/mdt593 Park JH, Powell AG, Roxburgh CSD, Horgan PG, McMillan DC, Edwards J. Mismatch repair status in patients with primary operable colorectal cancer: associations with the local and systemic tumour environment. Br J Cancer, 2016, 114(5):562–70., DOI 10.1038/bjc.2016.17 Park JH,McMillan DC, Powell AG, Richards CH, Horgan PG, Edwards J, et al. Evaluation of a tumor microenvironment–based prognostic score in primary operable colorectal cancer. Clin Cancer Res, 2015, 21(4):882–8., DOI 10.1158/ 1078-0432.ccr-14-1686 Park JH, Ishizuka M, McSorley ST, Kubota K, Roxburgh CSD, Nagata H, et al. Staging the tumor and staging the host: a two centre, two country comparison of systemic inflammatory responses of patients undergoing resection of primary operable colorectal cancer. Am J Surg, 2018, 216(3):458–64., DOI 10.1016/j. amjsurg.2017.08.044 Black S, Kushner I, Samols D. C-Reactive protein. J Biol Chem, 2004, 279(47): 48487–90., DOI 10.1074/jbc.R400025200 Butkiewicz AM, Kemona H, Dymicka-Piekarska V, Matowicka-Karna J, Radziwon P, Lipska A. Platelet count, mean platelet volume and thrombocytopoietic indices in healthy women and men. Thromb Res, 2006, 118(2):199–204., DOI 10.1016/j.thromres.2005.06.021 Long Y, Wang T, Gao Q, Zhou C. Prognostic significance of pretreatment elevated platelet count in patients with colorectal cancer: a meta-analysis. Oncotarget, 2016, 7(49):81849–61., DOI 10.18632/oncotarget.13248 Wu T,Mo Y,Wu C. Prognostic values of CEA, CA19-9, and CA72-4 in patients with stages I-III colorectal cancer. Int J Clin Exp Pathol, 2020, 13(7):1608–14. Rao H, Wu H, Huang Q, Yu Z, Zhong Z. Clinical value of serum CEA, CA24-2 and CA19-9 in patients with colorectal cancer. Clin Lab, 2021, 67(4)., DOI 10.7754/ Clin.Lab.2020.200828 Lam M, Roszik J, Kanikarla-Marie P, Davis JS, Morris J, Kopetz S, et al. The potential role of platelets in the consensus molecular subtypes of colorectal cancer. Cancer Metastasis Rev, 2017, 36(2):273–88., DOI 10.1007/s10555-017-9678-9 Soldevilla B, Carretero-Puche C, Gomez-Lopez G, Al-Shahrour F, Riesco MC, Gil-Calderon B, et al. The correlation between immune subtypes and consensus molecular subtypes in colorectal cancer identifies novel tumour microenvironment profiles, with prognostic and therapeutic implications. Eur J Cancer, 2019, 123: 118–29., DOI 10.1016/j.ejca.2019.09.008 Fontana E, Eason K, Cervantes A, Salazar R, Sadanandam A. Context mattersconsensus molecular subtypes of colorectal cancer as biomarkers for clinical trials. Ann Oncol, 2019, 30(4):520–7., DOI 10.1093/annonc/mdz052 Dunne PD, McArt DG, Bradley CA, O’Reilly PG, Barrett HL, Cummins R, et al. Challenging the cancer molecular stratification dogma: intratumoral heterogeneity undermines consensus molecular subtypes and potential diagnostic value in colorectal cancer. Clin Cancer Res, 2016, 22(16):4095–104., DOI 10.1158/1078-0432.ccr-16-0032 Kagawa H, Hatakeyama K, Shiomi A, Hino H, Manabe S, Yamaoka Y, et al. Consensus molecular subtyping improves the clinical usefulness of canonical tumor markers for colorectal cancer. Biomed Res, 2022, 43(6):201–9., DOI 10.2220/ biomedres.43.201 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611574 EP 1611593 DI 10.3389/pore.2024.1611574 PG 20 ER PT J AU Grayson, K Greenlee, J Himmel, L Hapach, L Reinhart-King, C King, M AF Grayson, A. Korie Greenlee, D. Joshua Himmel, E. Lauren Hapach, A. Lauren Reinhart-King, A. Cynthia King, R. Michael TI Spatial distribution of tumor-associated macrophages in an orthotopic prostate cancer mouse model SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tumor-associated macrophages; TRAIL; liposomes; prostate cancer; pathologic analysis ID tumor-associated macrophages; TRAIL; liposomes; prostate cancer; pathologic analysis AB Mounting evidence suggests that the immune landscape within prostate tumors influences progression, metastasis, treatment response, and patient outcomes. In this study, we investigated the spatial density of innate immune cell populations within NOD.SCID orthotopic prostate cancer xenografts following microinjection of human DU145 prostate cancer cells. Our laboratory has previously developed nanoscale liposomes that attach to leukocytes via conjugated E-selectin (ES) and kill cancer cells via TNF-related apoptosis inducing ligand (TRAIL). Immunohistochemistry (IHC) staining was performed on tumor samples to identify and quantify leukocyte infiltration for different periods of tumor growth and E-selectin/TRAIL (EST) liposome treatments. We examined the spatialtemporal dynamics of three different immune cell types infiltrating tumors using QuPath image analysis software. IHC staining revealed that F4/80+ tumor-associated macrophages (TAMs) were the most abundant immune cells in all groups, irrespective of time or treatment. The density of TAMs decreased over the course of tumor growth and decreased in response to EST liposome treatments. Intratumoral versus marginal analysis showed a greater presence of TAMs in the marginal regions at 3 weeks of tumor growth which became more evenly distributed over time and in tumors treated with EST liposomes. TUNEL staining indicated that EST liposomes significantly increased cell apoptosis in treated tumors. Additionally, confocal microscopy identified liposome-coated TAMs in both the core and periphery of tumors, highlighting the ability of liposomes to infiltrate tumors by “piggybacking” on macrophages. The results of this study indicate that TAMs represent the majority of innate immune cells within NOD.SCID orthotopic prostate tumors, and spatial density varieswidely as a function of tumor size, duration of tumor growth, and treatment of EST liposomes. C1 [Grayson, A. Korie] Vanderbilt University, Department of Biomedical EngineeringNashville, TN, USA. [Greenlee, D. Joshua] Vanderbilt University, Department of Biomedical EngineeringNashville, TN, USA. [Himmel, E. Lauren] Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology, Translational Pathology Shared ResourceNashville, TN, USA. [Hapach, A. Lauren] Vanderbilt University, Department of Biomedical EngineeringNashville, TN, USA. [Reinhart-King, A. Cynthia] Vanderbilt University, Department of Biomedical EngineeringNashville, TN, USA. [King, R. Michael] Vanderbilt University, Department of Biomedical EngineeringNashville, TN, USA. RP King, M (reprint author), Vanderbilt University, Department of Biomedical Engineering, Nashville, USA. EM mike.king@rice.edu CR Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA A Cancer J Clin, 2020, 70:7–30., DOI 10.3322/caac.21590 Denmeade SR, Isaacs JT. A history of prostate cancer treatment. Nat Rev Cancer, 2002, 2:389–96., DOI 10.1038/nrc801 Nuhn P, De Bono JS, Fizazi K, Freedland SJ, Grilli M, Kantoff PW, et al. Update on systemic prostate cancer therapies: management of metastatic castrationresistant prostate cancer in the era of precision Oncology. Eur Urol, 2019, 75: 88–99., DOI 10.1016/j.eururo.2018.03.028 Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature, 2008, 454:436–44., DOI 10.1038/nature07205 Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? The Lancet, 2001, 357:539–45., DOI 10.1016/S0140-6736(00)04046-0 Jiang J, Li J, Zhang Y, Zhu H, Liu J, Pumill C. The role of prostatitis in prostate cancer: meta-analysis. PLoS One, 2013, 8:e85179., DOI 10.1371/journal.pone. 0085179 Nakai Y, Nonomura N. Inflammation and prostate carcinogenesis. Int J Urol, 2013, 20:150–60., DOI 10.1111/j.1442-2042.2012.03101.x Bardan R, Dumache R, Dema A, Cumpanas A, Bucuras V. The role of prostatic inflammation biomarkers in the diagnosis of prostate diseases. Clin Biochem, 2014, 47:909–15., DOI 10.1016/j.clinbiochem.2014.02.008 Lanca T, Silva-Santos B. The split nature of tumor-infiltrating leukocytes: implications for cancer surveillance and immunotherapy. OncoImmunology, 2012, 1:717–25., DOI 10.4161/onci.20068 Gajewski TF, Schreiber H, Fu Y-X. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol, 2013, 14:1014–22., DOI 10.1038/ni.2703 Sutcliffe S, Platz EA. Inflammation in the etiology of prostate cancer: an epidemiologic perspective. Urol Oncol Semin Original Invest, 2007, 25:242–9., DOI 10.1016/j.urolonc.2006.09.014 Dennis LK, Lynch CF, Torner JC. Epidemiologic association between prostatitis and prostate cancer. Urology, 2002, 60:78–83., DOI 10.1016/s0090-4295(02)01637-0 Solinas C, Chanza NM, Awada A, Scartozzi M. The immune infiltrate in prostate, bladder and testicular tumors: an old friend for new challenges. Cancer Treat Rev, 2017, 53:138–45., DOI 10.1016/j.ctrv.2016.12.004 Strasner A, Karin M. Immune infiltration and prostate cancer. Front Oncol, 2015, 5:128., DOI 10.3389/fonc.2015.00128 Flammiger A, Bayer F, Cirugeda-Kuhnert A, Huland H, Tennstedt P, Simon R, et al. Intratumoral T but not B lymphocytes are related to clinical outcome in prostate cancer. APMIS, 2012, 120:901–8., DOI 10.1111/j.1600-0463.2012.02924.x Gannon PO, Poisson AO, Delvoye N, Lapointe R, Mes-Masson A-M, Saad F. Characterization of the intra-prostatic immune cell infiltration in androgendeprived prostate cancer patients. J Immunological Methods, 2009, 348:9–17., DOI 10.1016/j.jim.2009.06.004 Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa K, Hoimes CJ, et al. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study. J Clin Oncol, 2020, 38:395–405., DOI 10.1200/JCO.19.01638 Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. New Engl J Med, 2010, 363:411–22., DOI 10.1056/NEJMoa1001294 Mantovani A, Allavena P, Marchesi F, Garlanda C. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov, 2022, 21:799–820., DOI 10.1038/ s41573-022-00520-5 Lo CH, Lynch CC. Multifaceted roles for macrophages in prostate cancer skeletal metastasis. Front Endocrinol, Lausanne,, 2018, 9:247., DOI 10.3389/fendo.2018.00247 Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer, 2004, 4:71–8., DOI 10.1038/nrc1256 Lanciotti M, Masieri L, Raspollini MR, Minervini A, Mari A, Comito G, et al. The role of M1 and M2 macrophages in prostate cancer in relation to extracapsular tumor extension and biochemical recurrence after radical prostatectomy. Biomed Res Int, 2014, 2014:e486798., DOI 10.1155/2014/486798 Laviron M, Boissonnas A. Ontogeny of tumor-associated macrophages. Front Immunol, 2019, 10:1799., DOI 10.3389/fimmu.2019.01799 Duan Z, Luo Y. Targeting macrophages in cancer immunotherapy. Sig Transduct Target Ther, 2021, 6:127–1., DOI 10.1038/s41392-021-00506-6 Pyonteck SM, Akkari L, Schuhmacher AJ, Bowman RL, Sevenich L, Quail DF, et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med, 2013, 19:1264–72., DOI 10.1038/nm.3337 Pfirschke C, Zilionis R, Engblom C, Messemaker M, Zou AE, Rickelt S, et al. Macrophage-targeted therapy unlocks antitumoral cross-talk between ifnγ- secreting lymphocytes and IL12-producing dendritic cells. Cancer Immunol Res, 2022, 10:40–55., DOI 10.1158/2326-6066.CIR-21-0326 Wang S, El-Deiry WS. TRAIL and apoptosis induction by TNF-family death receptors. Oncogene, 2003, 22:8628–33., DOI 10.1038/sj.onc.1207232 Ashkenazi A, Pai RC, Fong S, Leung S, Lawrence DA, Marsters SA, et al. Safety and antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest, 1999, 104: 155–62., DOI 10.1172/JCI6926 von Karstedt S, Montinaro A, Walczak H. Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy. Nat Rev Cancer, 2017, 17:352–66., DOI 10. 1038/nrc.2017.28 Snajdauf M, Havlova K, Vachtenheim J, Ozaniak A, Lischke R, Bartunkova J, et al. The TRAIL in the treatment of human cancer: an update on clinical trials. Front Mol Biosciences, 2021, 8:628332., DOI 10.3389/fmolb.2021.628332 Stuckey DW, Shah K. TRAIL on trial: preclinical advances in cancer therapy. Trends Molecular Medicine, 2013, 19:685–94., DOI 10.1016/j.molmed.2013.08.007 Mitchell MJ, Wayne E, Rana K, Schaffer CB, King MR. TRAIL-coated leukocytes that kill cancer cells in the circulation. Proc Natl Acad Sci, 2014, 111:930–5., DOI 10.1073/pnas.1316312111 Greenlee JD, Lopez-Cavestany M, Ortiz-Otero N, Liu K, Subramanian T, Cagir B, et al. Oxaliplatin resistance in colorectal cancer enhances TRAIL sensitivity via death receptor 4 upregulation and lipid raft localization. eLife, 2021, 10:e67750., DOI 10.7554/eLife.67750 Ortiz-Otero N, Marshall JR, Lash B, King MR. Chemotherapy-induced release of circulating-tumor cells into the bloodstream in collective migration units with cancer-associated fibroblasts in metastatic cancer patients. BMC Cancer, 2020, 20: 873., DOI 10.1186/s12885-020-07376-1 Wayne EC, Chandrasekaran S, Mitchell MJ, Chan MF, Lee RE, Schaffer CB, et al. TRAIL-coated leukocytes that prevent the bloodborne metastasis of prostate cancer. J Control Release, 2016, 223:215–23., DOI 10.1016/j.jconrel. 2015.12.048 Jyotsana N, Zhang Z, Himmel LE, Yu F, King MR. Minimal dosing of leukocyte targeting TRAIL decreases triple-negative breast cancer metastasis following tumor resection. Sci Adv, 2019, 5:eaaw4197., DOI 10.1126/sciadv.aaw4197 Pavese J, Ogden IM, Bergan RC. An orthotopic murine model of human prostate cancer metastasis. J Vis Exp, 2013, 2013:50873., DOI 10.3791/50873 Somers KD, Brown RR, Holterman DA, Yousefieh N, Glass WF, Wright GL, et al. Orthotopic treatment model of prostate cancer and metastasis in the immunocompetent mouse: efficacy of flt3 ligand immunotherapy. Int J Cancer, 2003, 107:773–80., DOI 10.1002/ijc.11464 Shahryari V, Nip H, Saini S, Dar AA, Yamamura S, Mitsui Y, et al. Pre-clinical orthotopic murine model of human prostate cancer. J Vis Exp, 2016):54125., DOI 10. 3791/54125 Copeland BT, Shallal H, Shen C, Pienta KJ, Foss CA, Pomper MG. Imaging and characterization of macrophage distribution in mouse models of human prostate cancer. Mol Imaging Biol, 2019, 21:1054–63., DOI 10.1007/s11307-019- 01318-5 Halin Bergstrom S, Nilsson M, Adamo H, Thysell E, Jernberg E, Stattin P, et al. Extratumoral heme oxygenase-1, HO-1, expressing macrophages likely promote primary and metastatic prostate tumor growth. PLoS One, 2016, 11:e0157280., DOI 10.1371/journal.pone.0157280 Saltz J, Gupta R, Hou L, Kurc T, Singh P, Nguyen V, et al. Spatial organization and molecular correlation of tumor-infiltrating lymphocytes using deep learning on pathology images. Cell Rep, 2018, 23:181–93.e7., DOI 10.1016/j.celrep.2018.03.086 Bankhead P, Loughrey MB, Fernandez JA, Dombrowski Y, McArt DG, Dunne PD, et al. QuPath: open source software for digital pathology image analysis. Sci Rep, 2017, 7:16878., DOI 10.1038/s41598-017-17204-5 Bankhead P. Creating annotations around the tumor. Pete’s blog, 2018). Available from: https://petebankhead.github.io/qupath/scripts/2018/08/08/threeregions. html, Accessed March 17, 2024). Waddell LA, Lefevre L, Bush SJ, Raper A, Young R, Lisowski ZM, et al. ADGRE1, EMR1, F4/80, is a rapidly-evolving gene expressed in mammalian monocyte-macrophages. Front Immunol, 2018, 9:2246., DOI 10.3389/fimmu.2018. 02246 Schmid MC, Khan SQ, Kaneda MM, Pathria P, Shepard R, Louis TL, et al. Integrin CD11b activation drives anti-tumor innate immunity. Nat Commun, 2018, 9:5379., DOI 10.1038/s41467-018-07387-4 Odobasic D, Kitching AR, Holdsworth SR. Neutrophil-mediated regulation of innate and adaptive immunity: the role of myeloperoxidase. J Immunol Res, 2016, 2016:2349817., DOI 10.1155/2016/2349817 Chandrasekaran S, Chan MF, Li J, King MR. Super natural killer cells that target metastases in the tumor draining lymph nodes. Biomaterials, 2016, 77:66–76., DOI 10.1016/j.biomaterials.2015.11.001 Jackson Laboratory. Physiological data summary – NOD.CB17-prkdcscid/ J, 001303,, 2007). Available from: http://jackson.jax.org/rs/444-BUH-304/images/ physiological_data_001303.pdf, Accessed May 13, 2020). O’Connell KE, Mikkola AM, Stepanek AM, Vernet A, Hall CD, Sun CC, et al. Practical murine hematopathology: a comparative review and implications for research. Comp Med, 2015, 65:96–113. Gabelloni ML, Trevani AS, Sabatte J, Geffner J. Mechanisms regulating neutrophil survival and cell death. Semin Immunopathol, 2013, 35:423–37., DOI 10.1007/s00281-013-0364-x Wu L, Zhang XH-F. Tumor-associated neutrophils and macrophages—heterogenous but not chaotic. Front Immunol, 2020, 11:553967., DOI 10.3389/fimmu.2020.553967 Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, et al. Tissue resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity, 2013, 38:792–804., DOI 10. 1016/j.immuni.2013.04.004 Shimura S, Yang G, Ebara S, Wheeler TM, Frolov A, Thompson TC. Reduced infiltration of tumor-associated macrophages in human prostate cancer: association with cancer progression. Cancer Res, 2000, 60:5857–61. Sag D, Ayyildiz ZO, Gunalp S, Wingender G. The role of TRAIL/DRs in the modulation of immune cells and responses. Cancers, Basel,, 2019, 11:1469., DOI 10. 3390/cancers11101469 Liguori M, Buracchi C, Pasqualini F, Bergomas F, Pesce S, Sironi M, et al. Functional TRAIL receptors in monocytes and tumor-associated macrophages: a possible targeting pathway in the tumor microenvironment. Oncotarget, 2016, 7: 41662–76., DOI 10.18632/oncotarget.9340 Grayson KA, Jyotsana N, Ortiz-Otero N, King MR. Overcoming TRAILresistance by sensitizing prostate cancer 3D spheroids with taxanes. PLoS One, 2021, 16:e0246733., DOI 10.1371/journal.pone.0246733 GraysonKA,Hope JM, WangW, Reinhart-KingCA, KingMR.Taxanes sensitize prostate cancer cells to TRAIL-induced apoptotic synergy via endoplasmic reticulum stress. Mol Cancer Ther, 2021, 20:833–45., DOI 10.1158/1535-7163.MCT-20-0495 Park S, Cho DH, Andera L, Suh N, Kim I. Curcumin enhances TRAILinduced apoptosis of breast cancer cells by regulating apoptosis-related proteins. Mol Cell Biochem, 2013, 383:39–48., DOI 10.1007/s11010-013-1752-1 Sharkey CC, Li J, Roy S,Wu Q, King MR. Two-stage nanoparticle delivery of piperlongumine and tumor necrosis factor-related apoptosis-inducing ligand, TRAIL, anti-cancer therapy. Technology, Singap World Sci)., 2016, 4:60–9., DOI 10.1142/S2339547816500011 Hope JM, Lopez-Cavestany M, Wang W, Reinhart-King CA, King MR. Activation of Piezo1 sensitizes cells to TRAIL-mediated apoptosis through mitochondrial outer membrane permeability. Cell Death Dis, 2019, 10:837–15., DOI 10.1038/s41419-019-2063-6 Greenlee JD, Liu K, Lopez-Cavestany M, King MR. Piezo1 mechanoactivation is augmented by resveratrol and differs between colorectal cancer cells of primary and metastatic origin. Molecules, 2022, 27:5430., DOI 10.3390/ molecules27175430 Simons BW, Kothari V, Benzon B, Ghabili K, Hughes R, Zarif JC, et al. A mouse model of prostate cancer bone metastasis in a syngeneic immunocompetent host. Oncotarget, 2019, 10:6845–54., DOI 10.18632/oncotarget.27317 Allen TM, Brehm MA, Bridges S, Ferguson S, Kumar P, Mirochnitchenko O, et al. Humanized immune system mouse models: progress, challenges and opportunities. Nat Immunol, 2019, 20:770–4., DOI 10.1038/s41590-019-0416-z NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611586 EP 1611598 DI 10.3389/pore.2024.1611586 PG 13 ER PT J AU Krencz, I Sztankovics, D Sebestyen, A Papay, J Danko, T Moldvai, D Lutz, E Khoor, A AF Krencz, Ildiko Sztankovics, Daniel Sebestyen, Anna Papay, Judit Danko, Titanilla Moldvai, Dorottya Lutz, Elmar Khoor, Andras TI RICTOR amplification is associated with Rictor membrane staining and does not correlate with PD-L1 expression in lung squamous cell carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE RICTOR amplification; Rictor expression; mTORC2; PD-L1; lung squamous cell carcinoma ID RICTOR amplification; Rictor expression; mTORC2; PD-L1; lung squamous cell carcinoma AB RICTOR gene, which encodes the scaffold protein of mTORC2, can be amplified in various tumor types, including squamous cell carcinoma (SCC) of the lung. RICTOR amplification can lead to hyperactivation of mTORC2 and may serve as a targetable genetic alteration, including in lung SCC patients with no PD-L1 expression who are not expected to benefit from immune checkpoint inhibitor therapy. This study aimed to compare RICTOR amplification detected by fluorescence in situ hybridization (FISH) with Rictor and PD-L1 protein expression detected by immunohistochemistry (IHC) in SCC of the lung. The study was complemented by analysis of the publicly available Lung Squamous Cell Carcinoma (TCGA, Firehose legacy) dataset. RICTOR amplification was observed in 20% of our cases and 16% of the lung SCC cases of the TCGA dataset. Rictor and PD-L1 expression was seen in 74% and 44% of the cases, respectively. Rictor IHC showed two staining patterns: membrane staining (16% of the cases) and cytoplasmic staining (58% of the cases). Rictor membrane staining predicted RICTOR amplification as detected by FISH with high specificity (95%) and sensitivity (70%). We did not find any correlation between RICTOR amplification and PD-L1 expression; RICTOR amplification was detected in 18% and 26% of PD-L1 positive and negative cases, respectively. The TCGA dataset analysis showed similar results; RICTOR copy number correlated with Rictor mRNA and protein expression but showed no association with PD-L1 mRNA and protein expression. In conclusion, the correlation between RICTOR amplification and Rictor membrane staining suggests that the latter can potentially be used as a surrogate marker to identify lung SCC cases with RICTOR amplification. Since a significant proportion of PD-L1 negative SCC cases harbor RICTOR amplification, analyzing PD-L1 negative tumors by RICTOR FISH or Rictor IHC can help select patients who may benefit from mTORC2 inhibitor therapy. C1 [Krencz, Ildiko] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sztankovics, Daniel] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sebestyen, Anna] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Papay, Judit] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Danko, Titanilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Moldvai, Dorottya] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Lutz, Elmar] Mayo Clinic, Department of Laboratory Medicine and PathologyJacksonville, FL, USA. [Khoor, Andras] Mayo Clinic, Department of Laboratory Medicine and PathologyJacksonville, FL, USA. RP Khoor, A (reprint author), Mayo Clinic, Department of Laboratory Medicine and Pathology, Jacksonville, USA. EM khoor.andras@mayo.edu CR Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018, 68:394–424., DOI 10.3322/caac. 21492 Herbst RS, Morgensztern D, Boshoff C. The biology and management of nonsmall cell lung cancer. Nature, 2018, 553:446–54., DOI 10.1038/nature25183 Anusewicz D, Orzechowska M, Bednarek AK. Lung squamous cell carcinoma and lung adenocarcinoma differential gene expression regulation through pathways of Notch, Hedgehog, Wnt, and ErbB signalling. Sci Rep, 2020, 10:21128., DOI 10.1038/s41598-020- 77284-8 Sharma A, Merritt E, Hu X, Cruz A, Jiang C, Sarkodie H, et al. Non-genetic intra-tumor heterogeneity is a major predictor of phenotypic heterogeneity and ongoing evolutionary dynamics in lung tumors. Cell Rep, 2019, 29:2164–74., DOI 10. 1016/j.celrep.2019.10.045 Santos ES, Rodriguez E. Treatment considerations for patients with advanced squamous cell carcinoma of the lung. Clin Lung Cancer, 2022, 23:457–66., DOI 10. 1016/j.cllc.2022.06.002 National Comprehensive Cancer Network. Non-small cell lung cancer, 2022). Version: 5. Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell, 2017, 168:960–76., DOI 10.1016/j.cell.2017.02.004 Krencz I, Sebestyen A, Papay J, Lou Y, Lutz GF,Majewicz TL, et al. Correlation between immunohistochemistry and RICTOR fluorescence in situ hybridization amplification in small cell lung carcinoma. Hum Pathol, 2019, 93:74–80., DOI 10. 1016/j.humpath.2019.08.018 Krencz I, Sztankovics D, Danko T, Sebestyen A, Khoor A. Progression and metastasis of small cell lung carcinoma: the role of the PI3K/Akt/mTOR pathway and metabolic alterations. Cancer Metastasis Rev, 2021, 40:1141–57., DOI 10.1007/ s10555-021-10012-4 Sztankovics D, Krencz I, Moldvai D, Danko T, Nagy A, Nagy N, et al. Novel RICTOR amplification harbouring entities: FISH validation of RICTOR amplification in tumour tissue after next-generation sequencing. Sci Rep, 2023, 13:19610., DOI 10.1038/s41598-023-46927-x Krencz I, Sebestyen A, Khoor A. mTOR in lung neoplasms. Pathol Oncol Res, 2020, 26:35–48., DOI 10.1007/s12253-020-00796-1 Sholl LM, Sun H, Butaney M, Zhang C, Lee C, Janne PA, et al. ROS1 immunohistochemistry for detection of ROS1-rearranged lung adenocarcinomas. Am J Surg Pathol, 2013, 37:1441–9., DOI 10.1097/PAS. 0b013e3182960fa7 Wynes MW, Sholl LM, Dietel M, Schuuring E, Tsao MS, Yatabe Y, et al. An international interpretation study using the ALK IHC antibody D5F3 and a sensitive detection kit demonstrates high concordance between ALK IHC and ALK FISH and between evaluators. J Thorac Oncol, 2014, 9:631–8., DOI 10.1097/ JTO.0000000000000115 Nicholson AG, Tsao MS, Beasley MB, Borczuk AC, Brambilla E, Cooper WA, et al. The 2021 WHO classification of lung tumors: impact of advances since 2015. J Thorac Oncol, 2022, 17:362–87., DOI 10.1016/j.jtho.2021.11.003 Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med, 2015, 372:2018–28., DOI 10.1056/NEJMoa1501824 Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer, KEYNOTE-010): a randomised controlled trial. Lancet, 2016, 387:1540–50., DOI 10.1016/S0140-6736(15)01281-7 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal, 2013, 6:pl1., DOI 10.1126/scisignal.2004088 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov, 2012, 2:401–4., DOI 10.1158/2159-8290.CD-12-0095 Cheng H, Zou Y, Ross JS, Wang K, Liu X, Halmos B, et al. RICTOR amplification defines a novel subset of patients with lung cancer who may benefit from treatment with mTORC1/2 inhibitors. Cancer Discov, 2015, 5: 1262–70., DOI 10.1158/2159-8290.CD-14-0971 Kim LC, Rhee CH, Chen J. RICTOR amplification promotes NSCLC cell proliferation through formation and activation of mTORC2 at the expense of mTORC1. Mol Cancer Res, 2020, 18:1675–84., DOI 10.1158/1541-7786.MCR-20- 0262 Bang H, Ahn S, Ji Kim E, Kim ST, Park HY, Lee J, et al. Correlation between RICTOR overexpression and amplification in advanced solid tumors. Pathol Res Pract, 2020, 216:152734., DOI 10.1016/j.prp.2019.152734 Munari E, Zamboni G, Lunardi G, Marchionni L, MarconiM, Sommaggio M, et al. PD-L1 expression heterogeneity in non-small cell lung cancer: defining criteria for harmonization between biopsy specimens and whole sections. J Thorac Oncol, 2018, 13:1113–20., DOI 10.1016/j.jtho.2018.04.017 Ben Dori S, Aizic A, Sabo E, Hershkovitz D. Spatial heterogeneity of PD-L1 expression and the risk for misclassification of PD-L1 immunohistochemistry in non-small cell lung cancer. Lung Cancer, 2020, 147:91–8., DOI 10.1016/j.lungcan. 2020.07.012 OhWJ, Jacinto E. mTOR complex 2 signaling and functions. Cell Cycle, 2011, 10:2305–16., DOI 10.4161/cc.10.14.16586 Boulbes DR, Shaiken T, Sarbassov dos D. Endoplasmic reticulum is a main localization site of mTORC2. Biochem Biophys Res Commun, 2011, 413:46–52., DOI 10.1016/j.bbrc.2011.08.034 Gleason CE, Oses-Prieto JA, Li KH, Saha B, Situ G, Burlingame AL, et al. Phosphorylation at distinct subcellular locations underlies specificity in mTORC2- mediated activation of SGK1 and Akt. J Cel Sci, 2019, 132:jcs224931., DOI 10.1242/ jcs.224931 McDonald PC, Oloumi A, Mills J, Dobreva I, Maidan M, Gray V, et al. Rictor and integrin-linked kinase interact and regulate Akt phosphorylation and cancer cell survival. Cancer Res, 2008, 68:1618–24., DOI 10.1158/0008-5472.CAN-07-5869 Tsirtsaki K, Gkretsi V. The focal adhesion protein Integrin-Linked Kinase, ILK, as an important player in breast cancer pathogenesis. Cell AdhMigr, 2020, 14: 204–13., DOI 10.1080/19336918.2020.1829263 Luciano AK, Korobkina ED, Lyons SP, Haley JA, Fluharty SM, Jung SM, et al. Proximity labeling of endogenous RICTOR identifies mTOR complex 2 regulation by ADP ribosylation factor ARF1. J Biol Chem, 2022, 298:102379., DOI 10.1016/j.jbc. 2022.102379 Lau SCM, Pan Y, Velcheti V, Wong KK. Squamous cell lung cancer: current landscape and future therapeutic options. Cancer Cell, 2022, 40:1279–93., DOI 10. 1016/j.ccell.2022.09.018 Yang CY, Lin MW, Chang YL, Wu CT, Yang PC. Programmed cell deathligand 1 expression is associated with a favourable immune microenvironment and better overall survival in stage I pulmonary squamous cell carcinoma. Eur J Cancer, 2016, 57:91–103., DOI 10.1016/j.ejca.2015.12.033 Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, Abraham RT. The PI3K pathway in human disease. Cell, 2017, 170:605–35., DOI 10.1016/j.cell.2017. 07.029 Guenzle J, Akasaka H, Joechle K, Reichardt W, Venkatasamy A, Hoeppner J, et al. Pharmacological inhibition of mTORC2 reduces migration and metastasis in melanoma. Int J Mol Sci, 2020, 22:30., DOI 10.3390/ijms22010030 Werfel TA, Wang S, Jackson MA, Kavanaugh TE, Joly MM, Lee LH, et al. Selective mTORC2 inhibitor therapeutically blocks breast cancer cell growth and survival. Cancer Res, 2018, 78:1845–58., DOI 10.1158/0008-5472.CAN-17-2388 Xu Y, Li H, Huang Z, Chen K, Yu X, Sheng J, et al. Predictive values of genomic variation, tumor mutational burden, and PD-L1 expression in advanced lung squamous cell carcinoma treated with immunotherapy. Transl Lung Cancer Res, 2020, 9:2367–79., DOI 10.21037/tlcr-20-1130 Jebali A, Battistella M, Lebbe C, Dumaz N. RICTOR affects melanoma tumorigenesis and its resistance to targeted therapy. Biomedicines, 2021, 9:1498., DOI 10.3390/biomedicines9101498 Sun D, Yan W, Zhu H, Liu Q, Hou H. Case report: primary and acquired resistance mechanisms of nimotuzumab in advanced esophageal squamous cell carcinoma revealed by targeted sequencing. Front Oncol, 2020, 10:574523., DOI 10. 3389/fonc.2020.574523 Zhao D, Jiang M, Zhang X, Hou H. The role of RICTOR amplification in targeted therapy and drug resistance. Mol Med, 2020, 26:20., DOI 10.1186/s10020- 020-0146-6 Krencz I, Sebestyen A, Fabian K, Mark A, Moldvay J, Khoor A, et al. Expression of mTORC1/2-related proteins in primary and brain metastatic lung adenocarcinoma. Hum Pathol, 2017, 62:66–73., DOI 10.1016/j.humpath.2016.12.012 Schmidt KM, Dietrich P, Hackl C, Guenzle J, Bronsert P, Wagner C, et al. Inhibition of mTORC2/RICTOR impairs melanoma hepatic metastasis. Neoplasia, 2018, 20:1198–208., DOI 10.1016/j.neo.2018.10.001 Sakre N, Wildey G, Behtaj M, Kresak A, Yang M, Fu P, et al. RICTOR amplification identifies a subgroup in small cell lung cancer and predicts response to drugs targeting mTOR. Oncotarget, 2017, 8:5992–6002., DOI 10.18632/ oncotarget.13362 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611593 EP 1611602 DI 10.3389/pore.2024.1611593 PG 10 ER PT J AU Kerpel-Fronius, A Bogos, K AF Kerpel-Fronius, Anna Bogos, Krisztina TI HUNCHEST projects—advancing low-dose CT lung cancer screening in Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE lung cancer; screening; LDCT; pulmonary nodules; implementation ID lung cancer; screening; LDCT; pulmonary nodules; implementation AB Lung cancer, the leading cause of malignancy-related deaths worldwide, demands proactive measures to mitigate its impact. Low-dose computer tomography (LDCT) has emerged as a promising tool for secondary prevention through lung cancer screening (LCS). The HUNCHEST study, inspired by the success of international trials, including the National Lung Cancer Screening Trial and the Dutch NELSON study, embarked on the first LDCT-based LCS program in Hungary. The initiative assessed the screening efficiency, incorporating lung function tests and exploring the interplay between lung cancer and chronic obstructive pulmonary disease (COPD). Building upon this foundation, an implementation trial involving 18 Hungarian centers supported by the Ministry of Human Capacities demonstrated the feasibility of LCS within a multicentric framework. These centers, equipped with radiology capabilities, collaborated with multidisciplinary oncology teams, ensuring optimal patient pathways. However, a critical challenge remained the patient recruitment. To address this, the HUNCHEST 3 project, initiated in 2023, seeks to engage general practitioners (GPs) to reach out to eligible patients within a municipality collective of 60 thousand inhabitants. The project’s ultimate success is contingent upon the willingness of eligible individuals to undergo LDCT scans. In conclusion, the HUNCHEST program represents a crucial step in advancing lung cancer screening in Hungary. With a focus on efficiency, multidisciplinary collaboration, and innovative patient recruitment strategies, it endeavors to contribute to the reduction of lung cancer mortality and serve as a blueprint for potential nationwide LCS programs. C1 [Kerpel-Fronius, Anna] Orszagos Koranyi Tbc es Pulmonologiai Intezet, Radiologiai OsztalyBudapest, Hungary. [Bogos, Krisztina] National Koranyi Institute for Pulmonology, DirectorBudapest, Hungary. RP Kerpel-Fronius, A (reprint author), Orszagos Koranyi Tbc es Pulmonologiai Intezet, Radiologiai Osztaly, Budapest, Hungary. EM kerpel.anna@koranyi.hu CR Timmermann C. Lung cancer and consumption in the nineteenth century: bodies, tissues, cells and the making of a rare disease. In: A history of lung cancer. Science, technology and medicine in modern history. London: Palgrave Macmillan, 2014). Kozponti Statisztikai Hivatal 4.1.1.25. Reported new malignant neoplasms*, 2023). Available from: https://www.ksh.hu/stadat_files/ege/hu/ege0025.html, Accessed December 1, 2023). Blandin Knight S, Crosbie PA, Balata H, Chudziak J, Hussell T, Dive C. Progress and prospects of early detection in lung cancer. Open Biol, 2017, 7:170070., DOI 10.1098/rsob.170070 Kubik A, Parkin DM, Khlat M, Erban J, Polak J, Adamec M. Lack of benefit from semi-annual screening for cancer of the lung: follow-up report of a randomized controlled trial on a population of high-risk males in Czechoslovakia. Int J Cancer, 1990, 45(1):26–33., DOI 10.1002/ijc.2910450107 Tockman MS. Survival and mortality from lung cancer in a screened population. CHEST, 1986, 89(4):324S–5S., DOI 10.1378/chest.89.4_Supplement. 324S-a Fontana RS, Sanderson DR, Woolner LB, Taylor WF, Miller WE, Muhm JR. Lung cancer screening: the Mayo program. J Occup Med, 1986, 28(8):746–50., DOI 10.1097/00043764-198608000-00038 National Cancer Institute Prostate, lung, colorectal, and ovarian cancer screening trial, PLCO,, 2023). Available from: https://prevention.cancer.gov/ major-programs/prostate-lung-colorectal-and-ovarian-cancer-screening-trial-plco, Accessed December 1, 2023). Ostoros G, Ajkay Z, Boszormenyi NG, Kovacs G. Gondolatok a tudorak szurovizsgalatarol. Orvosi Hetilap, 1998, 139(42):2491–4. Koranyi Bulletin Koranyi bulletin, 2023). Available from: https://szakmai. koranyi.hu/bulletin/, Accessed December 1, 2023). Henschke CI, McCauley DI, Yankelevitz DF, Naidich DP, McGuinness G, Miettinen OS, et al. Early Lung Cancer Action Project: overall design and findings from baseline screening. Lancet, 1999, 354:99–105., DOI 10.1016/S0140-6736(99, 06093-6 The National Lung Screening Trial Research TeamAberle DR, Adams AM, Berg CD, Black WC, Clapp JD, et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med, 2011, 365:395–409., DOI 10.1056/ NEJMoa1102873 U.S. Preventive Services Task Force, USPSTF, Lung cancer: scrrening, 2021). Available from: https://www.uspreventiveservicestaskforce.org/uspstf/ recommendation/lung-cancer-screening, Accessed December 1, 2023). Yong PC, Sigel K, Rehmani S, Wisnivesky J, Kale MS. Lung cancer screening uptake in the United States. Chest, 2020, 157(1):236–8., DOI 10.1016/j.chest.2019.08. 2176 de Koning HJ, van der Aalst CM, de Jong PA, Scholten ET, Nackaerts K, Heuvelmans MA, et al. Reduced lung-cancermortality with volume CT screening in a randomized trial. N Engl J Med, 2020, 382(6):503–13., DOI 10.1056/ NEJMoa1911793 Kauczor HU, Bonomo L, Gaga M, Nackaerts K, Peled N, Prokop M, et al. ESR/ ERS white paper on lung cancer screening. Eur Radiol, 2015, 25(9):2519–31., DOI 10. 1007/s00330-015-3697-0 Oudkerk M, Devaraj A, Vliegenthart R, Henzler T, Prosch H, Heussel CP, et al. European position statement on lung cancer screening. Lancet Oncol, 2017, 18(12):e754–66., DOI 10.1016/S1470-2045(17)30861-6 Kerpel-Fronius A,Monostori Z, Kovacs G, Ostoros G, Horvath I, Solymosi D, et al. Nationwide lung cancer screening with low-dose computed tomography: implementation and first results of the HUNCHEST screening program. Eur Radiol, 2022, 32:4457–67., DOI 10.1007/s00330-022-08589-7 Nagy B, Szilberhorn L, Gyorbiro DM, Moizs M, Bajzik G, Kerpel-Fronius A, et al. Shall we screen lung cancer with low-dose computed tomography? Costeffectiveness in Hungary. Value Health Reg Issues, 2023, 34:55–64., DOI 10.1016/j. vhri.2022.10.002 Kerpel-Fronius A, Megyesfalvi Z, Markoczy Z, Solymosi D, Csanyi P, Tisza J, et al. HUNCHEST-II contributes to a shift to earlier-stage lung cancer detection: final results of a nationwide screening program. Eur Radiol, 2023)., DOI 10.1007/ s00330-023-10379-8 Forte GC, Altmayer S, Silva RF, Stefani MT, Libermann LL, Cavion CC, et al. Deep learning algorithms for diagnosis of lung cancer: a systematic review and meta-analysis. Cancers, 2022, 14:3856., DOI 10.3390/cancers14163856 Wait S, Alvarez-Rosete A, Osama T, Bancroft D, Cornelissen R, Marusic A, et al. Implementing lung cancer screening in Europe: taking a systems approach. JTO Clin Res Rep, 2022, 3(5):100329., DOI 10.1016/j.jtocrr.2022.100329 NHS Blog, 2024). Available from: https://www.england.nhs.uk/blog/rollingout- targeted-lung-health-checks/, Accessed April 1, 2024). Schmidt S Sebastian Schmidt: lung cancer screening – results from Croatia, 2024). Available from: https://oncodaily.com/blog/38363.html, Accessed April 1, 2024). Lung Cancer Policy Network LDCT screening pilots in Poland, 2022). Available from: https://www.lungcancerpolicynetwork.com/lung-cancerscreening- in-poland/, Accessed April 1, 2024). Prevenceproplice Prevenceproplice, 2024). Available from: https://www. prevenceproplice.cz/en/about/, Accessed April 1, 2024). Pirker R, Prosch H, Popper H, Klepetko W, Dieckmann K, Burghuber OC, et al. Lung cancer in Austria. J Thorac Oncol, 2021, 16(5):725–33., DOI 10.1016/j. jtho.2020.10.158 Juskanic D, Sandor F, Denkova L, Juhos P, Janik M, Hontzas J, et al. Lung cancer screening initiative in Slovakia: guidelines of screening implementation. Bratisl Lek Listy, 2023, 124(2):109–15., DOI 10.4149/BLL_2023_016 Stupnik T. Thoracic surgery in Slovenia. J Thorac Dis, 2022, 14(6):2326–34., DOI 10.21037/jtd-21-1172 Cavic M, Kovacevic T, Zaric B, Stojiljkovic D, Korda NJ, Rancic M, et al. Lung cancer in Serbia. J Thorac Oncol, 2022, 17(7):867–72., DOI 10.1016/j.jtho.2022. 04.010 Kinsinger LS, Anderson C, Kim J, Larson M, Chan SH, King HA, et al. Implementation of lung cancer screening in the Veterans health administration. JAMA Intern Med, 2017, 177(3):399–406., DOI 10.1001/jamainternmed.2016.9022 SOLACE Strengthening the screening of lung cancer in Europe, 2023). Available from: https://europeanlung.org/solace/, Accessed December 1, 2023). NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611635 EP 1611642 DI 10.3389/pore.2024.1611635 PG 8 ER PT J AU Szakallas, N Bartak, B Valcz, G Nagy, Zs Takacs, I Molnar, B AF Szakallas, Nikolett Bartak, K. Barbara Valcz, Gabor Nagy, B. Zsofia Takacs, Istvan Molnar, Bela TI Can long-read sequencing tackle the barriers, which the next-generation could not? A review SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE sequencing; short-read; long-read; bioinformatics; DNA ID sequencing; short-read; long-read; bioinformatics; DNA AB The large-scale heterogeneity of genetic diseases necessitated the deeper examination of nucleotide sequence alterations enhancing the discovery of new targeted drug attack points. The appearance of new sequencing techniques was essential to get more interpretable genomic data. In contrast to the previous short-reads, longer lengths can provide a better insight into the potential health threatening genetic abnormalities. Longreads offer more accurate variant identification and genome assembly methods, indicating advances in nucleotide deflect-related studies. In this review, we introduce the historical background of sequencing technologies and show their benefits and limits, as well. Furthermore, we highlight the differences between short- and long-read approaches, including their unique advances and difficulties in methodologies and evaluation. Additionally, we provide a detailed description of the corresponding bioinformatics and the current applications. C1 [Szakallas, Nikolett] Eotvos Lorand University, Faculty of Science, Department of Biological PhysicsBudapest, Hungary. [Bartak, K. Barbara] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Valcz, Gabor] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Nagy, B. Zsofia] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Takacs, Istvan] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Molnar, Bela] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. RP Szakallas, N (reprint author), Eotvos Lorand University, Faculty of Science, Department of Biological Physics, Budapest, Hungary. EM szakallasn3@gmail.com CR Alberts B. 4th chapter: DNA, chromosomes and genomes. In: Molecular biology of the cell. 6th ed. W.W. Norton & Company, 2015). Adewale BA. Will long-read sequencing technologies replace short-read sequencing technologies in the next 10 years? Afr J Lab Med, 2020, 9(1):1340., DOI 10.4102/ajlm.v9i1.1340 Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci USA, 1977, 74(12):5463–7., DOI 10.1073/pnas.74.12. 5463 Maxam AM, Gilbert W. A new method for sequencing DNA. Proc Natl Acad Sci USA, 1977, 74(2):560–4., DOI 10.1073/pnas.74.2.560 Marulies M, Egholm M, Altman WE, Attiya S, Bader JS, Bemben LA, et al. Genome sequencing in microfabricated high-density picolitre reactors. Nature, 2005, 437:376–80., DOI 10.1038/nature03959 Guo J, Yu L, Turro NJ, Ju J. An integrated system for DNA sequencing by synthesis using novel nucleotide analogues. Acc Chem Res, 2010, 43(4):551–63., DOI 10.1021/ar900255c Liu L, Li Y, Li S, Hu N, He Y, Pong R, et al. Comparison of next-generation sequencing systems. J Biomed Biotechnol, 2012, 2012:251364., DOI 10.1155/2012/ 251364 Harrington CT, Lin EI, Olson MT, Eshleman JR. Fundamentals of pyrosequencing. Arch Pathol Lab Med, 2013, 137(9):1296–303., DOI 10.5858/ arpa.2012-0463-RA Grigorev K, Foox J, Bezdan D, Butler D, Luxton JJ, Reed J, et al. Haplotype diversity and sequence heterogeneity of human telomeres. Genome Res, 2021, 31(7):1269–79., DOI 10.1101/gr.274639.120 Kumar KR, Cowley MJ, Davis RL. Next-generation sequencing and emerging technologies. Semin Thromb Hemost, 2019, 45(7):661–73., DOI 10.1055/s-0039- 1688446 Chen X, Xu H, Shu X, Song CX. Mapping epigenetic modifications by sequencing technologies. Cell Death Differ, 2023)., DOI 10.1038/s41418-023-01213-1 Xiao T, ZhouW. The third generation sequencing: the advanced approach to genetic diseases. Transl Pediatr, 2020, 9(2):163–73., DOI 10.21037/tp.2020.03.06 Athanasopoulou K, Boti MA, Adamopoulos PG, Skourou PC, Scorilas A. Third-generation sequencing: the spearhead towards the radical transformation of modern genomics. Life, Basel,, 2021, 12(1):30., DOI 10.3390/ life12010030 Kaplun L, Krautz-Peterson G, Neerman N, Stanley C, Hussey S, Folwick M, et al. ONT long-read WGS for variant discovery and orthogonal confirmation of short read WGS derived genetic variants in clinical genetic testing. Front Genet, 2023, 14:1145285., DOI 10.3389/fgene.2023.1145285 Roberts RJ, Carneiro MO, Schatz MC. The advantages of SMRT sequencing. Genome Biol, 2013, 14(7):405., DOI 10.1186/gb-2013-14-6-405 ONT Nanopore Technologies. Continuous development and improvement, 2023). Available from: https://nanoporetech.com/about-us/continuousdevelopment- and-improvement, Accessed 2024). Pollard MO, Gurdasani D, Mentzer AJ, Porter T, Sandhu MS. Long reads: their purpose and place. Hum Mol Genet, 2018, 27(R2):R234–R241., DOI 10.1093/ hmg/ddy177 Quick J, Loman NJ. Nanopore sequencing: an introduction. World Scientific Press, 2019). Deamer D, Akeson M, Branton D. Three decades of nanopore sequencing. Nat Biotechnol, 2016, 34:518–24., DOI 10.1038/nbt.3423 ONT Nanopore Technologies. Flow cells, 2023). Available from: https:// nanoporetech.com/how-it-works/flow-cells-and-nanopores, Accessed 2023). Nicholls SM, Quick JC, Tang S, Loman NJ. Ultra-deep, long-read nanopore sequencing of mock microbial community standards. GigaScience, 2019, 8(5): giz043., DOI 10.1093/gigascience/giz043 Ni Y, Liu X, Simeneh ZM, Yang M, Li R. Benchmarking of Nanopore R10.4 and R9.4.1 flow cells in single-cell whole-genome amplification and wholegenome shotgun sequencing. Comput Struct Biotechnol J, 2023, 21:2352–64., DOI 10. 1016/j.csbj.2023.03.038 Jennings W. Illumina sequencing, 2016)., DOI 10.1201/9781315181431-7 Stefan CP, Hall AT, Graham AS, Minogue TD. Comparison of illumina and Oxford nanopore sequencing technologies for pathogen detection from clinical matrices using molecular inversion probes. J Mol Diagn, 2022, 24(4):395–405., DOI 10.1016/j.jmoldx.2021.12.005 Harris TD, Buzby PR, Babcock H, Beer E, Bowers J, Braslavsky I, et al. Singlemolecule DNA sequencing of a viral genome. Science, 2008, 320(5872):106–9., DOI 10.1126/science.1150427 Eid J, Fehr A, Gray J, Luong K, Lyle J, Otto G, et al. Real-time DNA sequencing from single polymerase molecules. Science, 2009, 323(5910):133–8., DOI 10.1126/ science.1162986 Levene MJ, Korlach J, Turner SW, Foquet M, Craighead HG, Webb WW. Zero-mode waveguides for single-molecule analysis at high concentrations. Science, 2003, 299(5607):682–6., DOI 10.1126/science.1079700 Garrido-Cardenas JA, Garcia-Maroto F, Alvarez-Bermejo JA, Manzano- Agugliaro F. DNA sequencing sensors: an overview. Sensors, Basel,, 2017, 17(3):588., DOI 10.3390/s17030588 Vasudevan K, Devanga Ragupathi NK, Jacob JJ, Veeraraghavan B. Highly accurate-single chromosomal complete genomes using IonTorrent and MinION sequencing of clinical pathogens. Genomics, 2020, 112(1):545–51., DOI 10.1016/j. ygeno.2019.04.006 Warburton PE, Sebra RP. Long-read DNA sequencing: recent advances and remaining challenges. Annu Rev Genomics Hum Genet, 2023, 24:109–32., DOI 10.1146/annurev-genom-101722-103045 Ebler J, Haukness M, Pesout T, Marschall T, Paten B. Haplotype-aware diplotyping from noisy long reads. Genome Biol, 2019, 20:116., DOI 10.1186/s13059- 019-1709-0 Delahaye C, Nicolas J. Sequencing DNA with nanopores: troubles and biases. PLoS One, 2021, 16(10):e0257521., DOI 10.1371/journal.pone.0257521 Amarasinghe SL, Su S, Dong X, Zappia L, Ritchie ME, Gouil Q. Opportunities and challenges in long-read sequencing data analysis. Genome Biol, 2020, 21:30., DOI 10.1186/s13059-020-1935-5 Liu Q, Fang L, Yu G, Wang D, Xiao CL, Wang K. Detection of DNA base modifications by deep recurrent neural network on Oxford Nanopore sequencing data. Nat Commun, 2019, 10(1):2449., DOI 10.1038/s41467-019- 10168-2 Baid G, Cook DE, Shafin K, Yun T, Llinares-Lopez F, Berthet Q, et al. DeepConsensus: gap-aware sequence transformers for sequence correction. Nat Biotechnol, 2023, 41(2):232–8., DOI 10.1038/s41587-022-01435-7 Zhang H, Jain C, Aluru S. A comprehensive evaluation of long read error correction methods. BMC Genomics, 2020, 21(6):889., DOI 10.1186/s12864-020- 07227-0 Ardui S, Ameur A, Vermeesch JR, Hestand MS. Single molecule real-time, SMRT, sequencing comes of age: applications and utilities for medical diagnostics. Nucleic Acids Res, 2018, 46(5):2159–68., DOI 10.1093/nar/gky066 ONT Nanopore Technologies. Clive Brown’s keynote at nanopore community meeting, 2018). Available from: https://nanoporetech.com/resource-centre/clivebrown- ncm-2018, Accessed 2018). Pacific BioSciences HiFi sequencing, 2023). Available from: https://www.pacb. com/technology/hifi-sequencing/, Accessed 2024). Completing Human Genomes. Completing human genomes. Nat Methods, 2022, 19:629., DOI 10.1038/s41592-022-01537-9 Goodwin S, Gurtowski J, Ethe-Sayers S, Deshpande P, Schatz MC, McCombie WR. Oxford Nanopore sequencing, hybrid error correction, and de novo assembly of a eukaryotic genome. Genome Res, 2015, 25(11):1750–6., DOI 10.1101/gr. 191395.115 Hood L, Rowen L. The Human Genome Project: big science transforms biology and medicine. Genome Med, 2013, 5:79., DOI 10.1186/gm483 Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, et al. Initial sequencing and analysis of the human genome. Nature, 2001, 409:860–921., DOI 10.1038/35057062 Nurk S, Koren S, Rhie A, Rautiainen M, Bzikadze AV, Mikheenko A, et al. The complete sequence of a human genome. Science, 2022, 376:44–53., DOI 10.1126/ science.abj6987 Suzuki Y. Informatics for PacBio long-reads. Single molecule and single cell sequencing. In: Suzuki Y, editor. Advances in experimental medicine and biology. Springer, 2019). Oxford Nanopore Technologies. Oxford nanopore community, 2023). Available from: https://nanoporetech.com/community, Accessed 2024). Bioinformatics. Babraham bioinformatics, 2023). Available from: https:// www.bioinformatics.babraham.ac.uk/projects/fastqc/, Accessed 2023). Ewels P, Magnusson M, Lundin S, Kaller M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics, 2016, 32(19):3047–8., DOI 10.1093/bioinformatics/btw354 Fukasawa Y, Ermini L, Wang H, Carty K, Cheung MS. LongQC: a quality control tool for third generation sequencing long read data. G3 Genes, Genomes, Genet, 2020, 10(4):1193–6., DOI 10.1534/g3.119.400864 Coster WD, D’Hert S, Schultz DT, Cruts M, Van Broeckhoven C. NanoPack: visualizing and processing long-read sequencing data. Bioinformatics, 2018, 34(15): 2666–9., DOI 10.1093/bioinformatics/bty149 Lanfear R, Schalamun M, Kainer D, Wang W, Schwessinger B. MinIONQC: fast and simple quality control for MinION sequencing data. Bioinformatics, 2019, 35(3):523–5., DOI 10.1093/bioinformatics/bty654 Bolognini D, Bartalucci N, Mingrino A, Vannucchi AM, Magi A. NanoR: a user-friendly R package to analyze and compare nanopore sequencing data. PLoS One, 2019, 14(5):e0216471., DOI 10.1371/journal.pone.0216471 Graubert A, Aguet F, Ravi A, Ardlie KG, Getz G. RNA-SeQC 2: efficient RNAseq quality control and quantification for large cohorts. Bioinformatics, 2021, 37(18):3048–50., DOI 10.1093/bioinformatics/btab135 PacBio SMRT®. Tools reference guide, v11.0,, 2022). Available from: https:// www.pacb.com/wp-content/uploads/SMRT_Tools_Reference_Guide_v11.0.pdf, Accessed 2022). Oxford Nanopore Technologies. Oxford nanopore technologies, 2023). Available from: https://github.com/nanoporetech/dorado, Accessed 2023). Oxford Nanopore Technologies. Oxford nanopore technologies, 2024). Available from: https://community.nanoporetech.com/docs/prepare/library_ prep_protocols/Guppy-protocol/v/gpb_2003_v1_revax_14dec2018/guppysoftware- overview, Accessed 2024). Zheng Z, Li S, Su J, Leung AWS, Lam TW, Luo R. Symphonizing pileup and full-alignment for deep learning-based long-read variant calling. Nat Comput Sci, 2022, 2(12):797–803., DOI 10.1038/s43588-022-00387-x Sedlazeck FJ, Rescheneder P, SmolkaM, Fang H, NattestadM, von Haeseler A, et al. Accurate detection of complex structural variations using single-molecule sequencing. Nat Methods, 2018, 15:461–8., DOI 10.1038/s41592-018-0001-7 Romagnoli S, Bartalucci N, Vannucchi AM. Resolving complex structural variants via nanopore sequencing. Front Genet, 2023, 14:1213917., DOI 10.3389/ fgene.2023.1213917 Oxford Nanopore Technologies. Oxford nanopore technologies, 2018). Available from: https://github.com/nanoporetech/medaka, Accessed 2018). Ni P, Huang N, Zhang Z, Wang DP, Liang F, Miao Y, et al. DeepSignal: detecting DNA methylation state from Nanopore sequencing reads using deeplearning. Bioinformatics, 2019, 35(22):4586–95., DOI 10.1093/bioinformatics/btz276 Kolmogorov M, Yuan J, Lin Y, Pevzner P. Assembly of long error-prone reads using repeat graphs. Nat Biotechnol, 2019, 37(5):540–6., DOI 10.1038/s41587-019- 0072-8 Koren S, Walenz BP, Berlin K, Miller JR, Bergman NH, Phillippy AM. Canu: scalable and accurate long-read assembly via adaptive k-mer weighting and repeat separation. Genome Res, 2017, 27(5):722–36., DOI 10.1101/gr. 215087.116 Nurk S, Walenz BP, Rhie A, Vollger MR, Logsdon GA, Grothe R, et al. HiCanu: accurate assembly of segmental duplications, satellites, and allelic variants from high-fidelity long reads. Genome Res, 2020, 30(9):1291–305., DOI 10.1101/gr. 263566.120 Chaisson MJ, Tesler G. Mapping single molecule sequencing reads using basic local alignment with successive refinement, BLASR): application and theory. BMC Bioinformatics, 2012, 13:238., DOI 10.1186/1471-2105- 13-238 Chin CS, Peluso P, Sedlazeck FJ, Nattestad M, Concepcion GT, Clum A, et al. Phased diploid genome assembly with single-molecule real-time sequencing. Nat Methods, 2016, 13(12):1050–4., DOI 10.1038/nmeth.4035 Mayakonda A, Lin DC, Assenov Y, Plass C, Koeffler HP. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res, 2018, 28(11):1747–56., DOI 10.1101/gr.239244.118 Wickham H ggplot2: elegant graphics for data analysis. New York: Springer- Verlag, 2016). Available from: https://ggplot2.tidyverse.org, Accessed 2009). Hunter JD. Matplotlib: a 2D graphics environment. Comput Sci Eng, 2007, 9(3):90–5., DOI 10.1109/MCSE.2007.55 Bruce J, Abeel T, Shea T, PriestM, Abouelliel A, Sakthikumar S, et al. Pilon: an integrated tool for comprehensive microbial variant detection and genome assembly improvement. PLoS ONE, 2014, 9(11):e112963., DOI 10.1371/journal. pone.0112963 Vaser R, Sovic I, Nagarajan N, Sikic M. Fast and accurate de novo genome assembly from long uncorrected reads. Genome Res, 2017, 27(5):737–46., DOI 10.1101/gr.214270.116 Yepez VA, Gusic M, Kopajtich R, Mertes C, Smith NH, Alston CL, et al. Clinical implementation of RNA sequencing for Mendelian disease diagnostics. Genome Med, 2022, 14(1):38., DOI 10.1186/s13073-022-01019-9 Peresini P, Boza V, Brejova B, Vinar T. Nanopore base calling on the edge. Bioinformatics, 2021, 37(24):4661–7., DOI 10.1093/bioinformatics/btab528 Ledergerber C, Dessimoz C. Base-calling for next-generation sequencing platforms. Brief Bioinform, 2011, 12(5):489–97., DOI 10.1093/bib/bbq077 Koboldt DC. Best practices for variant calling in clinical sequencing. Genome Med, 2020, 12(91):91., DOI 10.1186/s13073-020-00791-w Schmieder R, Edwards R. Quality control and preprocessing of metagenomic datasets. Bioinformatics, 2011, 27(6):863–4., DOI 10.1093/ bioinformatics/btr026 Bolognini D, Semeraro R, Magi A. Versatile quality control methods for nanopore sequencing. Evol Bioinform Online, 2019, 15:1176934319863068., DOI 10.1177/1176934319863068 Frye M, Harada BT, Behm M, He C. RNA modifications modulate gene expression during development. Science, 2018, 361(6409):1346–9., DOI 10.1126/ science.aau1646 Field AE, Robertson NA, Wang T, Havas A, Ideker T, Adams PD. DNA methylation clocks in aging: categories, causes, and consequences. Mol Cel, 2018, 71(6):882–95., DOI 10.1016/j.molcel.2018.08.008 Esteller M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet, 2007, 8:286–98., DOI 10.1038/nrg2005 Kumar S, Chinnusamy V, Mohapatra T. Epigenetics of modified DNA bases: 5-methylcytosine and beyond. Front Genet, 2018, 9(18):640., DOI 10.3389/fgene. 2018.00640 Duffy K, Arangundy-Franklin S, Holliger P. Modified nucleic acids: replication, evolution, and next-generation therapeutics. BMC Biol, 2020, 18(112):112., DOI 10.1186/s12915-020-00803-6 Kumar S, Mohapatra T. Deciphering epitranscriptome: modification of mRNA bases provides a new perspective for post-transcriptional regulation of gene expression. Front Cel Dev. Biol., 2021, 9(16):628415., DOI 10.3389/fcell.2021. 628415 Soto J, Rodriguez-Antolin C, Vallespin E, de Castro Carpeno J, Ibanez de Caceres I. The impact of next-generation sequencing on the DNA methylation–based translational cancer research. Translational Res, 2016, 169: 1–18., DOI 10.1016/j.trsl.2015.11.003 Hirst M, Marra MA. Next generation sequencing based approaches to epigenomics. Brief Funct Genomics, 2010, 9(5-6):455–65., DOI 10.1093/bfgp/elq035 Flusberg BA, Webster DR, Lee JH, Travers KJ, Olivares EC, Clark TA, et al. Direct detection of DNA methylation during single-molecule, real-time sequencing. Nat Methods, 2010, 7:461–5., DOI 10.1038/nmeth.1459 Xu L, Seki M. Recent advances in the detection of base modifications using the Nanopore sequencer. J Hum Genet, 2020, 65:25–33., DOI 10.1038/s10038-019- 0679-0 Smolka M, Paulin LF, Grochowski CM, Horner DW, Mahmoud M, Behera S, et al. Detection of mosaic and population-level structural variants with Sniffles2. Nat Biotechnol, 2024)., DOI 10.1038/s41587-023-02024-y Stoiber M, Quick J, Egan R, Lee JE, Celniker S, Neely RK, et al. De novo identification of DNA modifications enabled by genome-guided nanopore signal processing. bioRxiv 094672, 2017)., DOI 10.1101/094672 Mandelker D, Ceyhan-Birsoy O. Evolving significance of tumor-normal sequencing in cancer care. Trends Cancer, 2020, 6(1):31–9., DOI 10.1016/j.trecan. 2019.11.006 Alkan C, Coe B, Eichler E. Genome structural variation discovery and genotyping. Nat Rev Genet, 2011, 12:363–76., DOI 10.1038/nrg2958 Zverinova S, Guryev V. Variant calling: considerations, practices, and developments. Hum Mutat, 2022, 43(8):976–85., DOI 10.1002/humu.24311 Mahmoud M, Gobet N, Cruz-Davalos DI, Mounier N, Dessimoz C, Sedlazeck FJ. Structural variant calling: the long and the short of it. Genome Biol, 2019, 20(246):246., DOI 10.1186/s13059-019-1828-7 Mitsuhashi S, Matsumoto N. Long-read sequencing for rare human genetic diseases. J Hum Genet, 2020, 65:11–9., DOI 10.1038/s10038-019-0671-8 Schule B, McFarland KN, Lee K, Tsai YC, Nguyen KD, Sun C, et al. Parkinson’s disease associated with pure ATXN10 repeat expansion. Parkinson’s Dis, 2017, 3(27):27., DOI 10.1038/s41531-017-0029-x McColgan P, Tabrizi SJ. Huntington’s disease: a clinical review. Eur J Neurol, 2018, 25(1):24–34., DOI 10.1111/ene.13413 Sakamoto Y, Zaha S, Suzuki Y, Seki M, Suzuki A. Application of long-read sequencing to the detection of structural variants in human cancer genomes. Comput Struct Biotechnol J, 2021, 19:4207–16., DOI 10.1016/j.csbj.2021.07.030 Phillippy A. New advances in sequence assembly. Genome Res, 2017, 27(5): xi–xiii., DOI 10.1101/gr.223057.117 van Dijk EL, Jaszczyszyn Y, Naquin D, Thermes C. The third revolution in sequencing technology. Trends Genet, 2018, 34(9):666–81., DOI 10.1016/j.tig.2018. 05.008 Nagarajan N, Pop M. Sequence assembly demystified. Nat Rev Genet, 2013, 14:157–67., DOI 10.1038/nrg3367 Chen Z, Erickson DL, Meng J. Benchmarking hybrid assembly approaches for genomic analyses of bacterial pathogens using Illumina and Oxford Nanopore sequencing. BMC Genomics, 2020, 21:631., DOI 10.1186/s12864-020-07041-8 Zhong Y, Xu F, Wu J, Schubert J, Li MM. Application of next generation sequencing in laboratory medicine. Ann Lab Med, 2021, 41(1):25–43., DOI 10.3343/ alm.2021.41.1.25 Bartalucci N, Romagnoli S, Vannucchi AM. A blood drop through the pore: nanopore sequencing in hematology. Trends Genet, 2022, 38(6):572–86., DOI 10.1016/j.tig.2021.11.003 Erlich RL, Jia X, Anderson S, Banks E, Gao X, Carrington M, et al. Nextgeneration sequencing for HLA typing of class I loci. BMC Genomics, 2011, 12:42., DOI 10.1186/1471-2164-12-42 Klasberg S, Surendranath V, Lange V, Schofl G. Bioinformatics strategies, challenges, and opportunities for next generation sequencingbased HLA genotyping. Transfus Med Hemother, 2019, 46(5):312–25., DOI 10.1159/000502487 Garg S. Computational methods for chromosome-scale haplotype reconstruction. Genome Biol, 2021, 22(1):101., DOI 10.1186/s13059-021-02328-9 Cilibrasi R, van Iersel L, Kelk S, Tromp J. The complexity of the single individual SNP haplotyping problem. Algorithmica, 2007, 49:13–36., DOI 10.1007/ s00453-007-0029-z Sakamoto Y, Sereewattanawoot S, Suzuki A. A new era of long-read sequencing for cancer genomics. J Hum Genet, 2020, 65:3–10., DOI 10.1038/ s10038-019-0658-5 Tarabichi M, Demetter P, Craciun L, Maenhaut C, Detours V. Thyroid cancer under the scope of emerging technologies. Mol Cel Endocrinol, 2022, 541: 111491., DOI 10.1016/j.mce.2021.111491 Munoz-Barrera A, Rubio-Rodriguez LA, Diaz-de Usera A, Jaspez D, Lorenzo-Salazar JM, Gonzalez-Montelongo R, et al. From samples to germline and somatic sequence variation: a focus on next-generation sequencing in melanoma research. Life, Basel,, 2022, 12(11):1939., DOI 10.3390/life12111939 Vollmers AC. Long noncoding RNA. Introduction and overview. In: Crusio WE, Dong H, Radeke HH, Rezaei N, Steinlein O, Xiao J, editors. Advances in experimental medicine and biology. Springer, 2022). Park EG, Pyo SJ, Cui Y, Yoon SH, Nam JW. Tumor immune microenvironment lncRNAs. Brief Bioinform, 2022, 23(1):bbab504., DOI 10.1093/ bib/bbab504 Li X, Wang C-Y. From bulk, single-cell to spatial RNA sequencing. Int J Oral Sci, 2021, 13(36):36., DOI 10.1038/s41368-021-00146-0 Depledge DP, Srinivas KP, Sadaoka T, Bready D,Mori Y, Placantonakis DG, et al. Direct RNA sequencing on nanopore arrays redefines the transcriptional complexity of a viral pathogen. Nat Commun, 2019, 10:754., DOI 10.1038/s41467- 019-08734-9 Jovic D, Liang X, Zeng H, Lin L, Xu F, Luo Y. Single-cell RNA sequencing technologies and applications: a brief overview. Clin Trans Med, 2022, 12(3):e694., DOI 10.1002/ctm2.694 Razavi P, Li BT, Brown DN, Jung B, Hubbell E, Shen R, et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat Med, 2019, 25:1928–37., DOI 10.1038/s41591-019-0652-7 Song P,Wu LR, Yan YH, Zhang JX, Chu T, Kwong LN, et al. Limitations and opportunities of technologies for the analysis of cell-free DNA in cancer diagnostics. Nat Biomed Eng, 2022, 6(3):232–45., DOI 10.1038/s41551-021- 00837-3 Shieh JTC. Genomic technologies to improve variation identification in undiagnosed diseases. Ped Neonatal, 2023, 64(S1):S18–S21., DOI 10.1016/J.pedneo. 2022.10.002 Macken WL, Vandrovcova J, Hanna MG, Pitceathly RDS. Applying genomic and transcriptomic advances to mitochondrial medicine. Nat Rev Neurol, 2021, 17: 215–30., DOI 10.1038/s41582-021-00455-2 Esteller M. Epigenetic gene silencing in cancer: the DNA hypermethylome. Hum Mol Genet, 2007, 16(Spec No 1):R50–9., DOI 10.1093/hmg/ddm018 Lakshminarasimhan R, Liang G. The role of DNA methylation in cancer. Adv Exp Med Biol, 2016, 945:151–72., DOI 10.1007/978-3-319-43624-1_7 Abante J, Kambhampati S, Feinberg AP, Goutsias J. Estimating DNA methylation potential energy landscapes from nanopore sequencing data. Sci Rep, 2021, 11(1):21619., DOI 10.1038/s41598-021-00781-x Jhanwar S, Deogade A. 5-Methylcytosine and 5-hydroxymethylcytosine signatures underlying pediatric cancers. Epigenomes, 2019, 3(2):9., DOI 10.3390/ epigenomes3020009 Zhou D, Guo S, Wang Y, Zhao J, Liu H, Zhou F, et al. Functional characteristics of DNA N6-methyladenine modification based on long-read sequencing in pancreatic cancer. Brief Funct Genomics, 2023, 23:150–62., DOI 10.1093/bfgp/elad021 Brockley LJ, Souza VGP, Forder A, Pewarchuk ME, Erkan M, Telkar N, et al. Sequence-based platforms for discovering biomarkers in liquid biopsy of nonsmall- cell lung cancer. Cancers, Basel,, 2023, 15(8):2275., DOI 10.3390/ cancers15082275 Ibrahim J, Peeters M, Van Camp G, Op de Beeck K. Methylation biomarkers for early cancer detection and diagnosis: current and future perspectives. Eur J Cancer, 2023, 178:91–113., DOI 10.1016/j.ejca.2022.10.015 Sahm F, Schrimpf D, Jones DTW, Meyer J, Kratz A, Reuss D, et al. Nextgeneration sequencing in routine brain tumor diagnostics enables an integrated diagnosis and identifies actionable targets. Acta Neuropathol, 2016, 131(6):903–10., DOI 10.1007/s00401-015-1519-8 Arts P, Simons A, AlZahrani MS, Yilmaz E, AlIdrissi E, van Aerde KJ, et al. Exome sequencing in routine diagnostics: a generic test for 254 patients with primary immunodeficiencies. Genome Med, 2019, 11:38., DOI 10.1186/s13073-019- 0649-3 Breinholt MF, Nielsen K, Schejbel L, Fassi DE, Schollkopf C, Novotny GW, et al. The value of next-generation sequencing in routine diagnostics and management of patients with cytopenia. Int J Lab Hematol, 2022, 44(3):531–7., DOI 10.1111/ijlh.13802 Fogel BL, Lee H, Strom SP, Deignan JL, Nelson SF. Clinical exome sequencing in neurogenetic and neuropsychiatric disorders. Ann N Y Acad Sci, 2016, 1366(1):49–60., DOI 10.1111/nyas.12850 Schmidt J, Blessing F, Fimpler L, Wenzel F. Nanopore sequencing in a clinical routine laboratory: challenges and opportunities. Clin Lab, 2020, 66(6)., DOI 10.7754/Clin.Lab.2019.191114 Olivucci G, Iovino E, Innella G, Turchetti D, Pippucci T, Magini P. Long read sequencing on its way to the routine diagnostics of genetic diseases. Front Genet, 2024, 15:1374860., DOI 10.3389/fgene.2024.1374860 Eagle SHC, Robertson J, Bastedo DP, Liu K, Nash JHE. Evaluation of five commercial DNA extraction kits using Salmonella as amodel for implementation of rapid Nanopore sequencing in routine diagnostic laboratories. Access Microbiol, 2023, 5(2):000468v3., DOI 10.1099/acmi.0.000468.v3 Erdmann H, Schoberl F, Giurgiu M, Leal Silva RM, Scholz V, Scharf F, et al. Parallel in-depth analysis of repeat expansions in ataxia patients by long-read sequencing. Brain, 2023, 146(5):1831–43., DOI 10.1093/brain/awac377 Matern BM, Olieslagers TI, Groeneweg M, Duygu B, Wieten L, Tilanus MGJ, et al. Long-read nanopore sequencing validated for human leukocyte antigen class I typing in routine diagnostics. JMol Diagn, 2020, 22(7):912–9., DOI 10.1016/j.jmoldx. 2020.04.001 Buenestado-Serrano S, Herranz M, Otero-Sobrino A, Molero-Salinas A, Rodriguez-Grande C, Sanz-Perez A, et al. Accelerating SARS-CoV-2 genomic surveillance in a routine clinical setting with nanopore sequencing. Int J Med Microbiol, 2024, 314:151599., DOI 10.1016/j.ijmm.2024.151599 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611676 EP 1611687 DI 10.3389/pore.2024.1611676 PG 12 ER PT J AU Han, Z Yang, F Wang, F Zheng, H Chen, X Meng, H Li, F AF Han, Zesen Yang, Fujun Wang, Fang Zheng, Huayu Chen, Xiujian Meng, Hongyu Li, Fenglei TI Advances in combined neuroendocrine carcinoma of lung cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE small cell neuroendocrine carcinoma; large cell neuroendocrine carcinoma; adenocarcinoma; squamous cell carcinoma; lung cancer ID small cell neuroendocrine carcinoma; large cell neuroendocrine carcinoma; adenocarcinoma; squamous cell carcinoma; lung cancer AB Lung cancer incidence and mortality rates are increasing worldwide, posing a significant public health challenge and an immense burden to affected families. Lung cancer encompasses distinct subtypes, namely, non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC). In clinical investigations, researchers have observed that neuroendocrine tumors can be classified into four types: typical carcinoid, atypical carcinoid, small-cell carcinoma, and largecell neuroendocrine carcinoma based on their unique features. However, there exist combined forms of neuroendocrine cancer. This study focuses specifically on combined pulmonary carcinomas with a neuroendocrine component. In this comprehensive review article, the authors provide an overview of combined lung cancers and present two pathological images to visually depict these distinctive subtypes. C1 [Han, Zesen] Hua Country People’s HospitalAnyang, Henan, China. [Yang, Fujun] Henan University of Science and Technology, Sanmenxia Central Hospital, Department of Medical OncologySanmenxia, China. [Wang, Fang] Hua Country People’s HospitalAnyang, Henan, China. [Zheng, Huayu] Hua Country People’s HospitalAnyang, Henan, China. [Chen, Xiujian] Hua Country People’s HospitalAnyang, Henan, China. [Meng, Hongyu] Hua Country People’s HospitalAnyang, Henan, China. [Li, Fenglei] Hua Country People’s HospitalAnyang, Henan, China. RP Han, Z (reprint author), Hua Country People’s Hospital, Anyang, China. EM hanzesen908@163.com CR Bade BC, Dela Cruz CS. Lung cancer 2020: epidemiology, etiology, and prevention. Clin Chest Med, 2020, 41(1):1–24., DOI 10.1016/j.ccm.2019.10.001 Wu F, Wang L, Zhou C. Lung cancer in China: current and prospect. Curr Opin Oncol, 2021, 33(1):40–6., DOI 10.1097/CCO.0000000000000703 Thai AA, Solomon BJ, Sequist LV, Gainor JF, Heist RS. Lung cancer. Lancet, 2021, 398(10299):535–54., DOI 10.1016/S0140-6736(21)00312-3 Svoboda E. Artificial intelligence is improving the detection of lung cancer. Nature, 2020, 587(7834):S20–S22., DOI 10.1038/d41586-020-03157-9 Li Y, Wu X, Yang P, Jiang G, Luo Y. Machine learning for lung cancer diagnosis, treatment, and prognosis. Genomics Proteomics Bioinformatics, 2022, 20(5): 850–66., DOI 10.1016/j.gpb.2022.11.003 Howlader N, Forjaz G, Mooradian MJ,Meza R, Kong CY, Cronin KA, et al. The effect of advances in lung-cancer treatment on population mortality. N Engl J Med, 2020, 383(7):640–9., DOI 10.1056/NEJMoa1916623 Mithoowani H, Febbraro M. Non-small-cell lung cancer in 2022: a review for general practitioners in oncology. Curr Oncol, 2022, 29(3):1828–39., DOI 10.3390/ curroncol29030150 Wang Y, Zou S, Zhao Z, Liu P, Ke C, Xu S. New insights into small-cell lung cancer development and therapy. Cell Biol Int, 2020, 44(8):1564–76., DOI 10.1002/ cbin.11359 Meijer JJ, Leonetti A, Airo G, Tiseo M, Rolfo C, Giovannetti E, et al. Small cell lung cancer: novel treatments beyond immunotherapy. Semin Cancer Biol, 2022, 86(Pt 2):376–85., DOI 10.1016/j.semcancer.2022.05.004 Nicholson AG, Tsao MS, Beasley MB, Borczuk AC, Brambilla E, Cooper WA, et al. The 2021 WHO classification of lung tumors: impact of advances since 2015. J Thorac Oncol, 2022, 17(3):362–87., DOI 10.1016/j.jtho.2021.11.003 Zheng M. Classification and pathology of lung cancer. Surg Oncol Clin N Am, 2016, 25(3):447–68., DOI 10.1016/j.soc.2016.02.003 Wang W, Xu C, Chen H, Jia J, Wang L, Feng H, et al. Genomic alterations and clinical outcomes in patients with lung adenocarcinoma with transformation to small cell lung cancer after treatment with EGFR tyrosine kinase inhibitors: a multicenter retrospective study. Lung Cancer, 2021, 155:20–7., DOI 10.1016/j. lungcan.2021.03.006 Morinaga R, Okamoto I, Furuta K, Kawano Y, Sekijima M, Dote K, et al. Sequential occurrence of non-small cell and small cell lung cancer with the same EGFR mutation. Lung Cancer, 2007, 58(3):411–3., DOI 10.1016/j.lungcan.2007. 05.014 Liu Y. Small cell lung cancer transformation from EGFR-mutated lung adenocarcinoma: a case report and literature review. Cancer Biol Ther, 2018, 19(6):445–9., DOI 10.1080/15384047.2018.1435222 Yin X, Li Y, Wang H, Jia T, Wang E, Luo Y, et al. Small cell lung cancer transformation: from pathogenesis to treatment. Semin Cancer Biol, 2022, 86(Pt 2): 595–606., DOI 10.1016/j.semcancer.2022.03.006 Norkowski E, Ghigna MR, Lacroix L, Le Chevalier T, Fadel E, Dartevelle P, et al. Small-cell carcinoma in the setting of pulmonary adenocarcinoma: new insights in the era of molecular pathology. J Thorac Oncol, 2013, 8(10):1265–71., DOI 10.1097/JTO.0b013e3182a407fa Giaccone G, He Y. Current knowledge of small cell lung cancer transformation from non-small cell lung cancer. Semin Cancer Biol, 2023, 94: 1–10., DOI 10.1016/j.semcancer.2023.05.006 Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med, 2011, 3(75):75ra26., DOI 10.1126/scitranslmed.3002003 Oxnard GR, Arcila ME, Sima CS, Riely GJ, Chmielecki J, Kris MG, et al. Acquired resistance to EGFR tyrosine kinase inhibitors in EGFR-mutant lung cancer: distinct natural history of patients with tumors harboring the T790M mutation. Clin Cancer Res, 2011, 17(6):1616–22., DOI 10.1158/1078-0432.CCR-10-2692 Hao L, Chen H, Wang L, Zhou H, Zhang Z, Han J, et al. Transformation or tumor heterogeneity: mutations in EGFR, SOX2, TP53, and RB1 persist in the histological rapid conversion from lung adenocarcinoma to small-cell lung cancer. Thorac Cancer, 2023, 14(11):1036–41., DOI 10.1111/1759-7714.14832 Li R, Jiang L, Zhou X, Lu Y, Zhang Y. Pseudo-small cell transformation in EGFR-mutant adenocarcinoma. Lung Cancer, 2021, 153:120–5., DOI 10.1016/j. lungcan.2020.12.036 Yamada K, Maeshima AM, Tsuta K, Tsuda H. Combined high-grade neuroendocrine carcinoma of the lung: clinicopathological and immunohistochemical study of 34 surgically resected cases. Pathol Int, 2014, 64(1):28–33., DOI 10.1111/pin.12127 Ebisu Y, Ishida M, Saito T,Murakawa T, Uemura Y, Tsuta K. Combined small cell carcinoma with giant cell carcinoma component of the lung: a case successfully diagnosed by computed tomography-guided fine-needle aspiration cytology. Oncol Lett, 2018, 15(2):1907–11., DOI 10.3892/ol.2017.7448 Fellegara G, D’adda T, Pilato FP, Froio E, Ampollini L, Rusca M, et al. Genetics of a combined lung small cell carcinoma and large cell neuroendocrine carcinoma with adenocarcinoma. Virchows Arch, 2008, 453(1):107–15., DOI 10.1007/s00428- 008-0620-y Zhuo M, Zhuang X, TangW, Xu J, Zhang C, Qian X, et al. The impact of IL-16 3’UTR polymorphism rs859 on lung carcinoma susceptibility among Chinese han individuals. Biomed Res Int, 2018, 2018:8305745., DOI 10.1155/2018/8305745 Kanan AD, Corey E, Vencio RZN, Ishwar A, Liu AY. Lineage relationship between prostate adenocarcinoma and small cell carcinoma. BMC Cancer, 2019, 19(1):518., DOI 10.1186/s12885-019-5680-7 Quintanal-Villalonga A, Durani V, Sabet A, Redin E, Kawasaki K, Shafer M, et al. Exportin 1 inhibition prevents neuroendocrine transformation through SOX2 down-regulation in lung and prostate cancers. Sci Transl Med, 2023, 15(707):eadf7006., DOI 10.1126/scitranslmed.adf7006 Antonia SJ, Lopez-Martin JA, Bendell J, Ott PA, Taylor M, Eder JP, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer, CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol, 2016, 17(7):883–95., DOI 10.1016/S1470-2045(16)30098-5 Rudin CM, Pietanza MC, Bauer TM, Ready N, Morgensztern D, Glisson BS, et al. Rovalpituzumab tesirine, a DLL3-targeted antibody-drug conjugate, in recurrent small-cell lung cancer: a first-in-human, first-in-class, open-label, phase 1 study. Lancet Oncol, 2017, 18(1):42–51., DOI 10.1016/S1470-2045(16, 30565-4 Farago AF, Piotrowska Z, Sequist LV. Unlocking the mystery of small-cell lung cancer transformations in EGFR mutant adenocarcinoma. J Clin Oncol, 2017, 35(26):2987–8., DOI 10.1200/JCO.2017.73.5696 Mao X, Liu J, Hu F, Niu Y, Pan F, Fu X, et al. Serum NSE is early marker of transformed neuroendocrine tumor after EGFR-TKI treatment of lung adenocarcinoma. Cancer Manag Res, 2022, 14:1293–302., DOI 10.2147/CMAR. S349082 Hendifar AE, Marchevsky AM, Tuli R. Neuroendocrine tumors of the lung: current challenges and advances in the diagnosis and management of welldifferentiated disease. J Thorac Oncol, 2017, 12(3):425–36., DOI 10.1016/j.jtho. 2016.11.2222 Lo RG, Pusceddu S, Proto C, Macerelli M, Signorelli D, Vitali M, et al. Treatment of lung large cell neuroendocrine carcinoma. Tumour Biol, 2016, 37(6): 7047–57., DOI 10.1007/s13277-016-5003-4 Cakir E, Demirag E, Aydin M, Unsal E. Clinicopathologic features and prognostic significance of lung tumours with mixed histologic patterns. Acta Chir Belg, 2009, 109(4):489–93., DOI 10.1080/00015458.2009.11680466 Filosso PL. Large-cell neuroendocrine carcinoma, LCNC, of the lung: a dilemma. Eur J Cardio-Thoracic Surg, 2003, 24(4):671–2., DOI 10.1016/s1010- 7940(03)00467-6 Ortega MA, Navarro F, Pekarek L, Fraile-Martinez O, Garcia-Montero C, Saez MA, et al. Exploring histopathological and serum biomarkers in lung adenocarcinoma: clinical applications and translational opportunities, Review). Int J Oncol, 2022, 61(6):154., DOI 10.3892/ijo.2022.5444 Affandi KA, Tizen NMS, Mustangin M, Zin RRMRM. p40 immunohistochemistry is an excellent marker in primary lung squamous cell carcinoma. J Pathol Transl Med, 2018, 52(5):283–9., DOI 10.4132/jptm.2018. 08.14 Wang Y, Qian F, Chen Y, Yang Z, Hu M, Lu J, et al. Comparative study of pulmonary combined large-cell neuroendocrine carcinoma and combined smallcell carcinoma in surgically resected high-grade neuroendocrine tumors of the lung. Front Oncol, 2021, 11:714549., DOI 10.3389/fonc.2021.714549 Yang Z, Wang Y, Chen Y, Qian F, Zhang Y, Hu M, et al. Combined large cell neuroendocrine carcinoma: clinical characteristics, prognosis and postoperative management. Eur J Cardiothorac Surg, 2022, 62(2):ezac069., DOI 10.1093/ejcts/ ezac069 Filosso PL, Fontana EC, Ruffini E. Large-cell neuroendocrine carcinoma and combined large-cell neuroendocrine carcinoma: 2 characters in search of an author. Eur J Cardiothorac Surg, 2022, 62(2):ezac176., DOI 10.1093/ejcts/ezac176 Li M, Yang L, Lu H. Pulmonary combined large cell neuroendocrine carcinoma. Pathol Oncol Res, 2022, 28:1610747., DOI 10.3389/pore.2022.1610747 YamamotoM, Ito R, Tsujibata A, Morita H, Yoneda T, Matano Y, et al. Largecell neuroendocrine carcinoma of the lung with carcinoid syndrome. Intern Med, 2023, 62(10):1557–63., DOI 10.2169/internalmedicine.0099-22 Bessho T, Yokochi K, Sakurai T. Combined large cell neuroendocrine carcinoma. Jpn J Thorac Cardiovasc Surg, 2004, 52(9):426–8., DOI 10.1007/ s11748-004-0037-8 Masuya D, Gotoh M, Nakashima T, Liu D, Ishikawa S, Yamamoto Y, et al. Combined large cell neuroendocrine carcinoma and squamous cell carcinoma of the lung; report of a case. Kyobu Geka, 2006, 59(6):491–5. Pelosi G, BarbareschiM, Cavazza A, Graziano P, Rossi G, Papotti M. Large cell carcinoma of the lung: a tumor in search of an author. A clinically oriented critical reappraisal. Lung Cancer, 2015, 87(3):226–31., DOI 10.1016/j.lungcan.2015.01.008 Davenport ML, Echols JB, Silva AD, Anderson JC, Owens P, Yates C, et al. miR-31 displays subtype specificity in lung cancer. Cancer Res, 2021, 81(8): 1942–53., DOI 10.1158/0008-5472.CAN-20-2769 Milione M, Maisonneuve P, Grillo F, Mangogna A, Centonze G, Prinzi N, et al. Ki-67 index of 55% distinguishes two groups of bronchopulmonary pure and composite large cell neuroendocrine carcinomas with distinct prognosis. Neuroendocrinology, 2021, 111(5):475–89., DOI 10.1159/000508376 Zhu Z, Liu Y, Xu H, Ning H, Xia Y, Shen L. Combined large cell neuroendocrine carcinoma, lung adenocarcinoma, and squamous cell carcinoma: a case report and review of the literature. J Cardiothorac Surg, 2023, 18(1):254., DOI 10.1186/s13019-023-02349-4 Oda R, Okuda K, Yamashita Y, Sakane T, Tatematsu T, Yokota K, et al. Longterm survivor of pulmonary combined large cell neuroendocrine carcinoma treated with nivolumab. Thorac Cancer, 2020, 11(7):2036–9., DOI 10.1111/1759-7714.13471 Archibald H, Keel S, Becker E. Large cell neuroendocrine carcinoma of the floor of mouth and retromolar trigone in association with basaloid squamous cell carcinoma. Int J Oral Maxillofac Surg, 2022, 51(12):1520–4., DOI 10.1016/j.ijom. 2022.01.020 Le DT, Do KH, Do TA, Bui HTT, Nguyen CV. Combined large-cell neuroendocrine and squamous cell carcinoma of the uterine cervix with a personal history of the primary breast duct carcinoma in situ: a clinicopathological characteristic and outcome. Case Rep Oncol, 2022, 15(2): 770–5., DOI 10.1159/000526337 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611693 EP 1611698 DI 10.3389/pore.2024.1611693 PG 6 ER PT J AU Orosz, Zs Kovacs, AF Orosz, Zsuzsanna Kovacs, Arpad TI The role of chemoradiotherapy and immunotherapy in stage III NSCLC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE locally advanced; NSCLC; chemoradiotherapy; immunotherapy; durvalumab ID locally advanced; NSCLC; chemoradiotherapy; immunotherapy; durvalumab AB Locally advanced non-small lung cancer encompasses a diverse range of tumors. In the last few years, the treatment of stage III unresectable nonsmall lung cancer has evolved significantly. The PACIFIC trial opened a new therapeutic era in the treatment of locally advanced NSCLC, establishing durvalumab consolidation therapy as the new standard of care worldwide. A careful evaluation of this type of lung cancer and a discussion of the management of these patients within a multidisciplinary team represents a crucial step in defining the best treatment strategy for each patient. For unresectable stage III NSCLC, definitive concurrent chemoradiotherapy (CCRT) was historically recommended as a treatment with a 5-year survival rate ranging from 20% to 30%. The PACIFIC study conducted in 2017 compared the use of chemoradiotherapy and maintenance therapy with the anti-PDL1 monoclonal antibody durvalumab to a placebo in patients with locally advanced NSCLC who had not experienced disease progression. The study was prospective, randomized, and phase III. The administration of this medication in patients with locally advanced non-small cell lung cancer (NSCLC) has demonstrated a notable improvement in overall survival. Multiple clinical trials are currently exploring various immune checkpoint inhibition regimens to enhance the treatment efficacy in patients with stage III cancer. Our goal is to offer an up-to-date summary of the planned clinical trials for treatment options, focusing on the significant obstacles and prospects in the post-PACIFIC era. C1 [Orosz, Zsuzsanna] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Kovacs, Arpad] Debreceni Egyetem, Onkologiai Klinika, Sugarterapia OsztalyDebrecen, Hungary. RP Orosz, Zs (reprint author), University of Debrecen Medical and Health Science Center, Department of Pulmonary Medicine, Debrecen, Hungary. EM zsuzsa.orosz@gmail.com CR Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin, 2023, 73(1):17–48., DOI 10.3322/caac.21763 Detterbeck FC, Boffa DJ, Kim AW, Tanoue LT. The eighth edition lung cancer stage classification. Chest, 2017, 151(1):193–203., DOI 10.1016/j.chest. 2016.10.010 Aredo JV, Mambetsariev I, Hellyer JA, Amini A, Neal JW, Padda SK, et al. Durvalumab for stage III EGFR-mutated NSCLC after definitive chemoradiotherapy. J Thorac Oncol, 2021, 16(6):1030–41., DOI 10.1016/j.jtho.2021.01.1628 Passiglia F, Leone G, Olmetto E, Delcuratolo MD, Tabbo F, Reale ML, et al. Immune-checkpoint inhibition in stage III unresectable NSCLC: challenges and opportunities in the post-PACIFIC era. Lung Cancer, 2021, 157:85–91., DOI 10. 1016/j.lungcan.2021.05.009 Remon J, Soria JC, Peters S, ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org. Early and locally advanced non-small-cell lung cancer: an update of the ESMO clinical practice guidelines focusing on diagnosis, staging, systemic and local therapy. Ann Oncol, 2021, 32(12): 1637–42., DOI 10.1016/j.annonc.2021.08.1994 Curran WJ, Paulus R, Langer CJ, Komaki R, Lee JS, Hauser S, et al. Sequential vs. concurrent chemoradiation for stage III non-small cell lung cancer: randomized phase III trial RTOG 9410. J Natl Cancer Inst, 2011, 103(19):1452–60., DOI 10.1093/jnci/djr325 Auperin A, Le Pechoux C, Pignon JP, Koning C, Jeremic B, Clamon G, et al. Concomitant radio-chemotherapy based on platin compounds in patients with locally advanced non-small cell lung cancer, NSCLC): a meta-analysis of individual data from 1764 patients. Ann Oncol, 2006, 17(3):473–83., DOI 10.1093/annonc/mdj117 Duma N, Santana-Davila R, Molina JR Non-small cell lung cancer: epidemiology, screening, diagnosis, and treatment. Mayo Clin Proc, 2019, 94(8): 1623–40., DOI 10.1016/j.mayocp.2019.01.013 Cheema PK, Rothenstein J, Melosky B, Brade A, Hirsh V. Perspectives on treatment advances for stage III locally advanced unresectable non-small-cell lung cancer. Curr Oncol, 2019, 26(1):37–42., DOI 10.3747/co.25.4096 Albain KS, Crowley JJ, Turrisi AT, Gandara DR, Farrar WB, Clark JI, et al. Concurrent cisplatin, etoposide, and chest radiotherapy in pathologic stage IIIB non-small-cell lung cancer: a Southwest Oncology Group phase II study, SWOG 9019. J Clin Oncol, 2002, 20(16):3454–60., DOI 10.1200/JCO.2002.03.055 Govindan R, Bogart J, Stinchcombe T, Wang X, Hodgson L, Kratzke R, et al. Randomized phase II study of pemetrexed, carboplatin, and thoracic radiation with or without cetuximab in patients with locally advanced unresectable non-small-cell lung cancer: cancer and Leukemia Group B trial 30407. J Clin Oncol, 2011, 29(23): 3120–5., DOI 10.1200/JCO.2010.33.4979 Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, et al. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med, 2017, 377(20):1919–29., DOI 10.1056/NEJMoa1709937 Spigel DR, Faivre-Finn C, Gray JE, Vicente D, Planchard D, Paz-Ares L, et al. Five-year survival outcomes from the PACIFIC trial: durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. J Clin Oncol, 2022, 40(12):1301–11., DOI 10.1200/JCO.21.01308 Antonia SJ, Ozguroglu M, Durvalumab in stage III non-small-cell lung cancer. N Engl J Med, 2018, 378(9):869–70., DOI 10.1056/NEJMc1716426 Naidoo J, Antonia S, Wu YL, Cho BC, Thiyagarajah P, Mann H, et al. Brief report: durvalumab after chemoradiotherapy in unresectable stage III EGFRmutant NSCLC: a post hoc subgroup analysis from PACIFIC. J Thorac Oncol, 2023, 18(5):657–63., DOI 10.1016/j.jtho.2023.02.009 Faivre-Finn C, Vicente D, Kurata T, Planchard D, Paz-Ares L, Vansteenkiste JF, et al. Four-year survival with durvalumab after chemoradiotherapy in stage III NSCLC-an update from the PACIFIC trial. J Thorac Oncol, 2021, 16(5):860–7., DOI 10.1016/j.jtho.2020.12.015 Girard N, Bar J, Garrido P, Garassino MC, McDonald F, Mornex F, et al. Treatment characteristics and real-world progression-free survival in patients with unresectable stage III NSCLC who received durvalumab after chemoradiotherapy: findings from the PACIFIC-R study. J Thorac Oncol, 2023, 18(2):181–93., DOI 10. 1016/j.jtho.2022.10.003 Hendriks LE, Kerr KM, Menis J, Mok TS, Nestle U, Passaro A, et al. Nononcogene- addicted metastatic non-small-cell lung cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol, 2023, 34(4):358–76., DOI 10.1016/j.annonc.2022.12.013 O’Rourke N, Roque I Figuls M, Farre Bernado N, Macbeth F. Concurrent chemoradiotherapy in non-small cell lung cancer. Cochrane Database Syst Rev, 2010, 16(6):CD002140., DOI 10.1002/14651858.CD002140.pub3 Wu L, Zhang Z, Bai M, Yan Y, Yu J, Xu Y. Radiation combined with immune checkpoint inhibitors for unresectable locally advanced non-small cell lung cancer: synergistic mechanisms, current state, challenges, and orientations. Cell Commun Signal, 2023, 21(1):119., DOI 10.1186/s12964-023-01139-8 DurmGA, Jabbour SK, Althouse SK, Liu Z, Sadiq AA, Zon RT, et al. A phase 2 trial of consolidation pembrolizumab following concurrent chemoradiation for patients with unresectable stage III non-small cell lung cancer: hoosier Cancer Research Network LUN 14-179. Cancer, 2020, 126(19):4353–61., DOI 10.1002/cncr.33083 Zhou Q, Chen M, Jiang O, Pan Y, Hu D, Lin Q, et al. Sugemalimab versus placebo after concurrent or sequential chemoradiotherapy in patients with locally advanced, unresectable, stage III non-small-cell lung cancer in China, GEMSTONE- 301): interim results of a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol, 2022, 23(2):209–19., DOI 10.1016/S1470-2045(21)00630-6 Wu YL Sequential chemoradiotherapy followed by sugemalimab for locally advanced NSCLC - author’s reply. Lancet Oncol, 2022, 23(4):e159., DOI 10.1016/ S1470-2045(22)00144-9 Garassino MC, Mazieres J, ReckM, Chouaid C, Bischoff H, Reinmuth N, et al. Durvalumab after sequential chemoradiotherapy in stage III, unresectable NSCLC: the phase 2 PACIFIC-6 trial. J Thorac Oncol, 2022, 17(12):1415–27., DOI 10.1016/j. jtho.2022.07.1148 Liu Y, Yao L, Kalhor N, Carter BW, AltanM, Blumenschein G, et al. Final efficacy outcomes of atezolizumab with chemoradiation for unresectable NSCLC: the phase II DETERRED trial. Lung Cancer, 2022, 174:112–7., DOI 10.1016/j.lungcan.2022.10.006 Manapov F, Kenndoff S, Kasmann L NICOLAS, DETERRED and KEYNOTE 799: focus on escalation of conventionally fractionated chemoradiotherapy by immune checkpoint inhibition in unresectable stage III non-small cell lung cancer. Transl Lung Cancer Res, 2022, 11(4):702–5., DOI 10.21037/tlcr-21-950 Jabbour SK, Lee KH, Frost N, Breder V, Kowalski DM, Pollock T, et al. Pembrolizumab plus concurrent chemoradiation therapy in patients with unresectable, locally advanced, stage III non-small cell lung cancer: the phase 2 KEYNOTE-799 nonrandomized trial. JAMA Oncol, 2021, 7(9):1–9., DOI 10.1001/jamaoncol.2021.2301 Carter L, Apte V, Shukla A, Ghose A, Mamidi R, Petohazi A, et al. Stage 3 N2 lung cancer: a multidisciplinary therapeutic conundrum. Curr Oncol Rep, 2024, 26(1):65–79., DOI 10.1007/s11912-023-01486-2 Wu YL, Zhou Q, ChenM, Pan Y, Jian O, Hu D, et al. OA02.05 sugemalimab vs placebo after cCRT or sCRT in pts with unresectable stage III NSCLC: final PFS analysis of a phase 3 study. J Thorac Oncol, 2022, 17(9):S7–S8., DOI 10.1016/j.jtho. 2022.07.021 Peters S, Felip E, Dafni U, Tufman A, Guckenberger M, Alvarez R, et al. Progression-free and overall survival for concurrent nivolumab with standard concurrent chemoradiotherapy in locally advanced stage IIIA-B NSCLC: results from the European thoracic oncology platform NICOLAS phase II trial, European thoracic oncology platform 6-14). J Thorac Oncol, 2021, 16(2):278–88., DOI 10.1016/ j.jtho.2020.10.129 Filippi AR, Dziadziuszko R, Garcia Campelo MR, Paoli JB, Sawyer W, Diaz Perez IE. DUART: durvalumab after radiotherapy in patients with unresectable, stage III NSCLC who are ineligible for chemotherapy. Future Oncol, 2021, 17(34): 4657–63., DOI 10.2217/fon-2021-0952 Ma L, Deng L, Peng J, Yu J, Meng X. Chemotherapy-free radiotherapy combined with immune checkpoint inhibitors: a new regimen for locally advanced non-small cell lung cancer? Cancer Biol Med, 2024, 20(12):1035–46., DOI 10.20892/j. issn.2095-3941.2023.0402 Lin SH, Pugh SL, Tsao AS, Edelman MJ, Doemer A, Simone CB, et al. Safety results of NRG-LU004: phase I trial of accelerated or conventionally fractionated radiotherapy combined with durvalumab in PD-L1–high locally advanced nonsmall cell lung cancer. J Clin Oncol, 2022, 40(16_Suppl. l):8513., DOI 10.1200/jco. 2022.40.16_suppl.8513 De Ruysscher D, Ramalingam S, Urbanic J, Gerber DE, Tan DSW, Cai J, et al. CheckMate 73L: a phase 3 study comparing nivolumab plus concurrent chemoradiotherapy followed by nivolumab with or without ipilimumab versus concurrent chemoradiotherapy followed by durvalumab for previously untreated, locally advanced stage III non-small-cell lung cancer. Clin Lung Cancer, 2022, 23(3):e264–e268., DOI 10.1016/j.cllc.2021.07.005 Mazieres J, Drilon A, Lusque A, Mhanna L, Cortot AB, Mezquita L, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol, 2019, 30(8):1321–8., DOI 10.1093/annonc/mdz167 Passiglia F, Bironzo P, Bertaglia V, Listi A, Garbo E, Scagliotti GV. Optimizing the clinical management of EGFR-mutant advanced non-small cell lung cancer: a literature review. Transl Lung Cancer Res, 2022, 11(5):935–49., DOI 10.21037/tlcr- 22-1 Loh J, Low JL, Sachdeva M, Low PQ, Wong RSJ, Huang Y, et al. Management of oncogene driven locally advanced unresectable non-small cell lung cancer. Expert Rev Anticancer Ther, 2023, 23(9):913–26., DOI 10.1080/14737140.2023.2245140 Hellyer JA, Aredo JV, DasM, Ramchandran K, Padda SK, Neal JW, et al. Role of consolidation durvalumab in patients with EGFR- and HER2-mutant unresectable stage III NSCLC. J Thorac Oncol, 2021, 16(5):868–72., DOI 10.1016/ j.jtho.2020.12.020 Lu S, Casarini I, Kato T, Cobo M, Ozguroglu M, Hodge R, et al. Osimertinib maintenance after definitive chemoradiation in patients with unresectable EGFR mutation positive stage III non-small-cell lung cancer: LAURA trial in progress. Clin Lung Cancer, 2021, 22(4):371–5., DOI 10.1016/j.cllc.2020.11.004 Passaro A, Leighl N, Blackhall F, Popat S, Kerr K, AhnMJ, et al. ESMO expert consensus statements on the management of EGFR mutant non-small-cell lung cancer. Ann Oncol, 2022, 33(5):466–87., DOI 10.1016/j.annonc.2022.02.003 Addeo A, Passaro A, Malapelle U, Banna GL, Subbiah V, Friedlaender A. Immunotherapy in non-small cell lung cancer harbouring driver mutations. Cancer Treat Rev, 2021, 96:102179., DOI 10.1016/j.ctrv.2021.102179 Attili I, Passaro A, Corvaja C, Trillo Aliaga P, Del Signore E, Spitaleri G, et al. Immune checkpoint inhibitors in EGFR-mutant non-small cell lung cancer: a systematic review. Cancer Treat Rev, 2023, 119:102602., DOI 10.1016/j.ctrv.2023.102602 Senan S, Bahce I Pulmonary toxicity in patients treated with immune checkpoint inhibitors and radiation. Ann Oncol, 2020, 31(12):1597–8., DOI 10. 1016/j.annonc.2020.10.476 Suresh K, Naidoo J Lower survival in patients who develop pneumonitis following immunotherapy for lung cancer. Clin Lung Cancer, 2020, 21(3):e169–70., DOI 10.1016/j.cllc.2019.10.009 Berner F, Bomze D, Diem S, Ali OH, Fassler M, Ring S, et al. Association of checkpoint inhibitor-induced toxic effects with shared cancer and tissue antigens in non-small cell lung cancer. JAMA Oncol, 2019, 5(7):1043–7., DOI 10.1001/ jamaoncol.2019.0402 Fukihara J, Sakamoto K, Koyama J, Ito T, Iwano S,Morise M, et al. Prognostic impact and risk factors of immune-related pneumonitis in patients with non-smallcell lung cancer who received programmed death 1 inhibitors. Clin Lung Cancer, 2019, 20(6):442–50., DOI 10.1016/j.cllc.2019.07.006 Fukihara J, Sakamoto K, Koyama J, Ito T, Iwano S, Morise M, et al. Reply to "prognostic impact and risk factors of immune-related pneumonitis in patients with non-small-cell lung cancer who received programmed death-1 inhibitors. Clin Lung Cancer, 2020, 21(3):e205., DOI 10.1016/j.cllc.2019.11.013 Akkad N, Thomas TS, Luo S, Knoche E, Sanfilippo KM, Keller JW. A realworld study of pneumonitis in non-small cell lung cancer patients receiving durvalumab following concurrent chemoradiation. J Thorac Dis, 2023, 15(12): 6427–35., DOI 10.21037/jtd-22-1604 DelaunayM, Cadranel J, Lusque A,Meyer N, Gounant V,Moro-Sibilot D, et al. Immune-checkpoint inhibitors associated with interstitial lung disease in cancer patients. Eur Respir J, 2017, 50(2):1700050., DOI 10.1183/13993003.00050-2017 Naidoo J, Vansteenkiste JF, Faivre-Finn C, Ozguroglu M, Murakami S, Hui R, et al. Characterizing immune-mediated adverse events with durvalumab in patients with unresectable stage III NSCLC: a post-hoc analysis of the PACIFIC trial. Lung Cancer, 2022, 166:84–93., DOI 10.1016/j.lungcan.2022.02.003 Haanen J, Obeid M, Spain L, Carbonnel F, Wang Y, Robert C, et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol, 2022, 33(12): 1217–38., DOI 10.1016/j.annonc.2022.10.001 Massaccesi M, Calcagni ML, Spitilli MG, Cocciolillo F, Pelligro F, Bonomo L, et al. 18F-FDG PET-CT during chemo-radiotherapy in patients with non-small cell lung cancer: the early metabolic response correlates with the delivered radiation dose. Radiat Oncol, 2012, 7:106., DOI 10.1186/1748-717X-7-106 Delasos L, Wei W, Hassan KA, Pennell NA, Patil P, Stevenson J. Clinical outcomes with pembrolizumab-based therapies in recurrent/refractory NSCLC after chemoradiation and consolidative durvalumab. Clin Lung Cancer, 2023, 24(6):e205–e213., DOI 10.1016/j.cllc.2023.04.008 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611716 EP 1611721 DI 10.3389/pore.2024.1611716 PG 6 ER PT J AU Nadorvari, M Lotz, G Kulka, J Kiss, A Timar, J AF Nadorvari, L. Maja Lotz, Gabor Kulka, Janina Kiss, Andras Timar, Jozsef TI Microsatellite instability and mismatch repair protein deficiency: equal predictive markers? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE mismatch repair deficiency; microsatellite instability; immunohistochemistry; molecular testing; cancer ID mismatch repair deficiency; microsatellite instability; immunohistochemistry; molecular testing; cancer AB Current clinical guidelines recommend mismatch repair (MMR) protein immunohistochemistry (IHC) or molecular microsatellite instability (MSI) tests as predictive markers of immunotherapies. Most of the pathological guidelines consider MMR protein IHC as the gold standard test to identify cancers with MMR deficiency and recommend molecular MSI tests only in special circumstances or to screen for Lynch syndrome. However, there are data in the literature which suggest that the two test types may not be equal. For example, molecular epidemiology studies reported different rates of deficient MMR (dMMR) and MSI in various cancer types. Additionally, direct comparisons of the two tests revealed relatively frequent discrepancies between MMR IHC and MSI tests, especially in non-colorectal and non-endometrial cancers and in cases with unusual dMMR phenotypes. There are also scattered clinical data showing that the efficacy of immune checkpoint inhibitors is different if the patient selection was based on dMMR versus MSI status of the cancers. All these observations question the current dogma that dMMR phenotype and genetic MSI status are equal predictive markers of the immunotherapies. C1 [Nadorvari, L. Maja] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Lotz, Gabor] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Timar, J (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM jtimar@gmail.com CR Sepulveda AR, Hamilton SR, Allegra CJ, Grody W, Cushman-Vokoun AM, Funkhouser WK, et al. Molecular biomarkers for the evaluation of colorectal cancer: guideline from the American society for clinical pathology, College of American pathologists, association for molecular pathology, and the American society of clinical oncology. J Clin Oncol, 2017, 35:1453–86., DOI 10.1200/JCO.2016.71.9807 Sarode VR, Robinson L. Screening for Lynch syndrome by immunohistochemistry of mismatch repair proteins: significance of indeterminate result and correlation with mutational studies. Arch Pathol Lab Med, 2019, 143:1225–33., DOI 10.5858/arpa.2018-0201-OA Le DT, Uram JN, Wang H, Barlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med, 2015, 372: 2509–20., DOI 10.1056/NEJMoa1500596 Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta AD, Delord JP, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol, 2019, 38:1–10., DOI 10.1200/JCO.19.02105 Le DT, Kim WT, Van Cutsem E, Geva R, Jager D, Hara H, et al. Phase-II open label study of pembrolizumab in treatment refractory, microsatellite instabilityhigh/ mismatch repair deficient metastatic colorectal cancer. KEYNOTE-164. J Clin Oncol, 2020, 38:11–9., DOI 10.1200/JCO.19.02107 Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz H-J, Morse M. A., et al. Nivolumab in patients with metastatic DNA mismatch repair deficient/ microsatellite instability-high colorectal cancer, CheckMate 142). Lancet Oncol, 2017, 18:1182–91., DOI 10.1016/S1470-2045(17)30422-9 Lenz H-J, van Cutsem E, Limon M, Wong KYM, Hendlis A, Aglietta M, et al. First-line nivolumab plus low-dose ipilimumab for microsatellite instability-high/ mismatch repair-deficient metastatic colorectal cancer: the phase II Checkmate 142 study. J Clin Oncol, 2021, 40:161–70., DOI 10.1200/JCO.21.01015 Palmeri M, Mehnert J, Silk AW, Jabbour SK, Ganesan S, Popli P, et al. Realworld application of tumor mutational burden-high, TMB-high, and microsatellite instability, MSI, confirms their utility as immunotherapy biomarkers. ESMO Open, 2022, 7:100336., DOI 10.1016/j.esmoop.2021.100336 Hopkins JL, Lan L, Zou L. DNA repair defects in cancer and therapeutic opportunities. Genes Dev, 2022, 36:278–93., DOI 10.1101/gad.349431.122 Bellizzi AM, Frankel WL. Colorectal cancer due to deficiency in DNA mismatch repair function: a review. Adv Anat Pathol, 2009, 16:405–17., DOI 10. 1097/PAP.0b013e3181bb6bdc Hause RJ, Pritchard CC, Shendure J, Salipante SJ. Classification and characterization of microsatellite instability across 18 cancer types. Nat Med, 2016, 22:1342–50., DOI 10.1038/nm.4191 Loewer SS, McGurk MP, Clark AG, Barbash DA. Satellite DNA evolution: old ideas, new approaches. Curr Opin Genet Dev, 2018, 49:70–8., DOI 10.1016/j.gde. 2018.03.003 Evrard C, Tachon G, Randrian V, Karayan-Tapon L, Tougeron D. Microsatellite instability: diagnosis, heterogeneity, discordance and clinical impact in colorectal cancer. Cancers, 2019, 11:1567., DOI 10.3390/cancers11101567 Campbell BB, light N, Fabrizio D, Zatzman M, Fuligni F, de Borja R, et al. Comprehensive analysis of hypermutation in human cancer. Cell, 2017, 171: 1042–56., DOI 10.1016/j.cell.2017.09.048 Chen W, Swanson BJ, Frankel WL. Molecular genetics of microsatelliteunstable colorectal cancer for pathologists. Diagn Pathol, 2017, 12:24., DOI 10.1186/ s13000-017-0613-8 Modrich P, Lahue R. Mismatch repair in replication fidelity, genetic recombination and cancer biology. Ann Rev Biochem, 1996, 65:101–33., DOI 10. 1146/annurev.bi.65.070196.000533 Abildgaard AB, Nielsen SV, Bernstein I, Stein A, Lindorff-Larsen K, Hartmann-Petersen R. Lynch syndrome, molecular mechanisms and variant classification. Br J Cancer, 2022, 128:726–34., DOI 10.1038/s41416-022-02059-z Peltomaki P. Update on Lynch syndrome genomics. Fam Cancer, 2016, 15: 385–93., DOI 10.1007/s10689-016-9882-8 Li K, Luo H, Huang L, Luo H, Zhu X. Microsatellite instability: a review of what the oncologist should know. Cancer Cel Int, 2020, 20:16., DOI 10.1186/s12935- 019-1091-8 Gutierrez C, Ogino S, Meyerhardt JA, Iorgulescu B. The prevalence and prognosis of microsatellite instability-high/mismatch repair deficient colorectal adenocarcinomas in the United States. JCO Precis Oncol, 2023, 7:e2200179., DOI 10.1200/PO.22.00179 Lorenzi M, Amonkar M, Zhang J, Mehta S, Liaw K-L. Epidemiology of microsatellite instability high, MSI-H, and deficient mismatch repair, dMMR, in solid tumors: a structured literature review. J Oncol, 2020, 2020:1–17., DOI 10.1155/ 2020/1807929 Evrard C, Messina S, Sefrioui D, Frouin E, Auriault M-L, Chautard R, et al. Heterogeneity of mismatch repair status and microsatellite instability between primary tumour and metastasis and its implications for immunotherapy in colorectal cancers. Int J Mol Sci, 2022, 23:4427., DOI 10.3390/ijms23084427 Bonneville R, Paruchuri A,Wing MR, Krook MA, Reeser JW, Chen HZ, et al. Characterization of clonal evolution in microsatellite unstable metastatic cancers through multiregional tumor sequencing. Mol Cancer Res, 2021, 19:465–74., DOI 10. 1158/1541-7786.MCR-19-0955 Toieb J, Shi O, Pederson L, Alberts S, Wolmark N, Van Cutsem E, de Gramont A, et al. Prognosis of microsatellite instability and/or mismatch repair deficiency stage III colon cancer patients after disease recurrence following adjuvant treatments: results of an ACCENT pooled analysis of seven studies. Ann Oncol, 2019, 30:1466–71., DOI 10.1093/annonc/mdz208 Cohen SA, Pritchard CC, Jarvik GP. Lynch syndrome: from screening to diagnosis to treatment in the era of modern molecular oncology. Ann Rev Genomics Hum Genet, 2019, 20:293–307., DOI 10.1146/annurev-genom-083118-015406 JaffrelotM, Fares N, Brunac AC, Laurenty AP, DanjouxM, Grand D, et al. An unusual phenotype occurs in 15% of mismatch repair-deficient tumors and is associated with non-colorectal cancers and genetic syndromes. Mod Pathol, 2022, 35:427–37., DOI 10.1038/s41379-021-00918-3 Cerretelli G, Ager A, Arends MJ, Frayling IM. Molecular pathology of Lynch syndrome. J Pathol, 2020, 250:518–31., DOI 10.1002/path.5422 Williams MH, Hadjinicolaou AV, Norton BC, Kader R, Lovat LB. Lynch syndrome: from detection to treatment. Front Oncol, 2023, 13:1166238., DOI 10. 3389/fonc.2023.1166238 Yoshida H, Takigawa W, Kobayashi-Kato M, Nishikawa T, Shiraishi K, Ishikawa M. Mismatch repair protein expression in endometrial cancer: assessing concordance and unveiling pitfalls in two different immunohistochemistry assays. J Pers Med, 2023, 13:1260., DOI 10.3390/jpm13081260 Sari A, Pollett A, Eriksson LR, Lumsden-Johanson B, van de Laar E, Kazerouni H, et al. Interobserver agreement for mismatch repair protein immunohistochemistry in endometrial and nonserous, nonmucinous ovarian carcinomas. Am J Surg Path, 2019, 43:591–600., DOI 10.1097/PAS. 0000000000001220 Malapelle U, Parente P, Pepe F, de Luca C, Cerino P, Covelli C, et al. Impact of pre-analytical factors on MSI test accuracy inmucinous colorectal adenocarcinoma: a multi-assay concordance study. Cells, 2020, 9:9092019., DOI 10.3390/cells9092019 Wang C, Zhang L, Vakiani E, Shia J. Detecting mismatch repair deficiency in solid neoplasms: immunohistochemistry, microsatellite instability or both? Mod Pathol, 2022, 35:1515–28., DOI 10.1038/s41379-022-01109-4 Reidinger CJ, Esnakula A, Haight PJ, Suarez AA, Chen W, Gillespie J, et al. Characterization of mismatch-repair, MMR)/microsatellite instability, MSI)- discordant endometrial cancers. Cancer, 2023, 2023:35030., DOI 10.1002/cncr.35030 Bartley AN, Mills AM, Konnick E, Overman M, Ventura CB, Souter L, et al. Mismatch repair and microsatellite instability testing for immune checkpoint inhibitor therapy: guideline from the College of American pathologists in collaboration with the association for molecular pathology and fight colorectal cancer. Arch Pathol LabMed, 2022, 146:1194–210., DOI 10.5858/arpa.2021-0632-CP McCarthy AJ, Capo-Chichi J-M, Spence T, Grenier S, Stockley T, Kamel-Reid S, et al. Heterogenous loss of mismatch repair, MMR, protein expression: a challenge for immunohistochemical interpretation and microsatellite instability, MSI, evaluation. J Pathol Clin Res, 2019, 5:115–29., DOI 10.1002/cjp2.120 Amato M, Franco R, Facchini G, Addeo R, Ciardiello F, Berretta M, et al. Microsatellite instability: from the implementation of the detection to a prognostic and predictive role in cancers. Int J Mol Sci, 2022, 23:8726., DOI 10.3390/ ijms23158726 Hechtman JF, Rana S, Middha S, Stadler Z, Latham A, Benayed R, et al. Retained mismatch repair protein expression occurs in approximately 6% of microsatellite instability-high cancers and is associated with missense mutations in mismatch repair genes. Mod Pathol, 2020, 33:871–9., DOI 10.1038/s41379-019- 0414-6 Watkins JC, Nucci MR, Ritterhause LL, Howitz BE, Sholl LM. Unusual mismatch repair immunohistochemical patterns in endometrial carcinoma. Am J Surg Pathol, 2016, 40:909–16., DOI 10.1097/PAS.0000000000000663 Quaas A, Rehkaemper J, Rueschoff J, Pamuk A, Zander T, Hillmer A, et al. Occurrence of high microsatellite-instability/mismatch repair deficiency in nearly 2,000 human adenocarcinomas of the gastrointestinal tract, pancreas, and bile ducts: a study from a large German comprehensive cancer center. Front Oncol, 2021, 11:569–475., DOI 10.3389/fonc.2021.569475 Aiyer KTS, Doeleman T, Ryan NA, Nielsen M, Crosbie EJ, Smit VTHBM, et al. Validity of a two-antibody resting algorithm for mismatch repair deficiency testing in cancer: a systematic literature review and meta-analysis. Mod Pathol, 2022, 35: 1775–83., DOI 10.1038/s41379-022-01149-w Dedeurwaerdere F, Claes KBM, Van Dorpe J, Rottiers I, Van der Meulen J, Breyne J, et al. Comparison of microsatellite instability detection by immunohistochemistry and molecular techniques in colorectal and endometrial cancer. Sci Rep, 2021, 11:12880., DOI 10.1038/s41598-021-91974-x Cortez-Ciriano I, Lee S, Park W-Y, Kim T-M, Park PJ. A molecular portrait of microsatellite instability across multiple cancers. Nat Commun, 2017, 8:15180., DOI 10.1038/ncomms15180 Murphy KM, Zhang S, Geiger T, Hafez MJ, Bacher J, Berg KD, et al. Comparison of the microsatellite instability analysis system and the Bethesda panel for determination of microsatellite instability in colorectal cancers. J Mol Diagn, 2006, 8:305–11., DOI 10.2353/jmoldx.2006.050092 Libera L, Sahnane N, Pepe F, Pisapia P, De Luca C, Russo G, et al. Critical aspects of microsatellite instability testing in endometrial cancer: a comparison study. Hum Pathol, 2022, 128:134–40., DOI 10.1016/j.humpath.2022.07.014 Malapelle U, Parente P, Pepe P, Pepe F, De Luca C, Sgariglia R, et al. Evaluation of microsatellite instability and mismatch repair status in different solid tumors: a multicenter analysis in a real world setting. Cells, 2021, 10:1878., DOI 10. 3390/cells10081878 Siemanowski J, Schoming-Markiefka B, Buhl T, Haak A, Siebolts U, Dietmaier W, et al. Managing difficulties of microsatellite instability testing in endometrial cancer. Limitations and advantages of four different PCR-based approaches. Cancers, 2021, 13:1268., DOI 10.3390/cancers13061268 Zwaenepoel K, Holmgaard Duelund J, deWinne K, Maes V, Weyn C, Lambin S, et al. Clinical performance of the Idylla MSI test for a rapid assessment of the DNA microsatellite status in human colorectal cancer. J Mol Diagn, 2020, 22: 386–95., DOI 10.1016/j.jmoldx.2019.12.002 Trabucco SE, Gowen K, Maund SL, Sanford E, Fabrizio DA, Hall MJ, et al. A novel next generation sequencing approach to detecting microsatellite instability and pan-tumor characterization of 1000 microsatellite instability-high cases in 67,000 patient samples. J Mol Diagn, 2019, 21:1053–66., DOI 10.1016/j.jmoldx.2019. 06.011 Middha S, Zhang L, Nafa K, Jayakumaran G, Wong D, Kim HR, et al. Reliable pan-cancer microsatellite instability assessment by using targeted next generation sequencing data. JCO Precis Oncol, 2017, 2017:1–17., DOI 10.1200/PO.17.00084 VanderWalde A, Spetzler D, Xiao N, Gatalica Z, Marshall J. Microsatellite instability status determined by next generation sequencing and compared with PD-L1 and tumor mutational burden in 11,348 patients. Cancer Med, 2018, 7: 746–56., DOI 10.1002/cam4.1372 Bartels S, Grote I, Wagner M, Boog J, Schipper E, Reineke-Plaass T, et al. Concordance in detection of microsatellite instability by PCR and NGS in routinely processed tumor specimens of several cancer types. Cancer Med, 2023, 12: 16707–15., DOI 10.1002/cam4.6293 Luchini C, Bibeau F, Ligtenberg MJL, Singh N, Nottegar A, Bosse T, et al. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer and its relationship with PD-1/PD-L1 expression and tumor mutational burden. Ann Oncol, 2019, 30:1232–43., DOI 10.1093/annonc/mdz116 Vikas P, Messersmith H, Compton C, Sholl L, Bradduss RR, Davis A, et al. Mismatch repair and microsatellite instability testing for immune checkpoint inhibitor therapy. ASCO endorsement of College of American Pathologists guideline. J Clin Oncol, 2023, 41:1943–8., DOI 10.1200/JCO.22.02462 Ruschoff J, Schildhaus H-U, Ruschoff JH, Johrens K, Edmonston TB, Dietmaier W, et al. Testing for deficient mismatch repair and microsatellite instability: A focused update. Die Pathologie, 2023, 44:61–70., DOI 10.1007/ s00292-023-01208-2 Bonneville R, Krook MA, Kautto EA, Miya J, Wing MR, Chen HZ, et al. Landscape of microsatellite instability across 39 cancer types. JCO Precis Oncol, 2017, 1:1–15., DOI 10.1200/PO.17.00073 Long DR, Waalkes A, Panicker VP, Hause RJ, Salipante SJ. Identifying optimal loci for the molecular diagnosis of microsatellite instability. Clin Chem, 2020, 66: 1310–8., DOI 10.1093/clinchem/hvaa177 Salipante SJ, Scroggins SM, Hampel HL, Turner EH, Pritchard CC. Microsatellite instability testing in endometrial cancer. Limitations and advantages of four different PCR based approaches. Clin Chem, 2014, 60: 1192–9., DOI 10.1373/clinchem.2014.223677 Papke D, Yurgelun MB, Noffsinger AE, Redston M, Genta RM. Prevalence of mismatch repair deficiency in rectal adenocarcinomas. N Engl J Med, 2022, 387: 1714–6., DOI 10.1056/NEJMc2210175 Kang Y-J, O’Hare S, Franchini F, Uzerman M, Zalcberg J, Macrae F, et al. A scoping review and meta-analysis on the prevalence of pan-tumour biomarkers, dMMR, MSI, high TMB, in different solid tumours. Sci Rep, 2022, 12:20495., DOI 10.1038/s41598-022-23319-1 Mitric C, Salman L, Abrahamyan L, Kim SR, Pechlivanoglou P, Chan KKW, et al. Mismatch repair deficiency, microsatellite instability and Lynch syndrome in ovarian cancer: a systematic review and meta-analysis. Gynecol Oncol, 2023, 170: 133–42., DOI 10.1016/j.ygyno.2022.12.008 De Salins AGD, Tachon G, Cohen R, Karayan-Tapon L, Junca A, Frouin E, et al. Discordance between immunohistochemistry ofmismatch repair proteins and molecular testing of microsatellite instability in colorectal cancer. ESMO Open, 2021, 3:100120., DOI 10.1016/jesmoop.2021.100120 Loughrey MB, McGrath J, Coleman HG, Bankhead P, Maxwell P, McGready C, et al. Identifying mismatch repair-deficient colon cancer: near-perfect concordance between immunohistochemistry and microsatellite instability testing in a large, population-based series. Histopathology, 2021, 78:401–13., DOI 10.1111/his.14233 Chen M-L, Chen J-Y, Hu J, Chen Q, Liu B-R, Qian X-P, et al. Comparison of microsatellite status detection methods in colorectal carcinoma. Int J Exp Pathol, 2018, 11:1431–8. Nadorvari ML, Kenessey I, Kiss A, Barbai T, Kulka J, Raso E, et al. Comparison of standard mismatch repair deficiency and microsatellite instability tests in a large cancer series. J Transl Med, 2024, 22:150., DOI 10.1186/s12967-024-04960-y Yamamoto G, Ito T, Suzuki O, Kamae N, Kakuta M, Takahashi A, et al. Concordance between microsatellite instability testing and immunohistochemistry for mismatch repair proteins and efficient screening of mismatch repair deficient gastric cancer. Oncol Lett, 2023, 26:494., DOI 10.3892/ol.2023.14081 Stelloo E, Jansen AML, Osse EM, nout RA, Creutzberg CL, Ruano D, et al. Practical guidance for mismatch repair-deficiency testing in endometrial cancer. Ann Oncol, 2017, 28:96–102., DOI 10.1093/annonc/mdw542 Latham A, Srinivasan P, Kemel Y, Shia J, Bandlamudi C, Mandelker D, et al. Microsatellite instability is associated with the presence of Lynch syndrome pancancer. J Clin Oncol, 2018, 37:286–95., DOI 10.1200/JCO.18.00283 Mandal R, Samstein RM, Lee K-W, Havel JJ, Wang H, Krishna C, et al. Genetic diversity of tumors with mismatch repair deficiency influences anti-PD1 immunotherapy response. Science, 2019, 364:485–91., DOI 10.1126/science.aau0447 Wang Q-X, Qu C-H, Gao Y-H, Ding P-R, Yun J-P, Xie D, et al. The degree of microsatellite instability predicts response to PD-1 blockade immunotherapy in mismatch repair deficient/microsatellite instability-high colorectal cancers. Exp Hematol Oncol, 2021, 10:2., DOI 10.1186/s40164-020-00193-z Quintanilha JCF, Graf RP, Fisher VA, Oxnard GR, Ellis H, Panarelli N, et al. Comparative effectiveness of immune checkpoint inhibitors vs chemotherapy in patients with metastatic colorectal cancer with measures of microsatellite instability, mismatch repair or tumor mutational burden. JAMA Netw Open, 2023, 6: e2252244., DOI 10.1001/jamanetworkopen.2022.52244 Hill BL, Graf RP, Shah K, Danziger N, Lin D, Quintanilha J, et al. Mismatch repair deficiency, next generation sequencing-based microsatellite instability and tumor mutational burden as predictive biomarkers for immune checkpoint inhibitor effectiveness in frontline treatment of advanced stage endometrial cancer. Int J Gynecol Cancer, 2023, 33:504–13., DOI 10.1136/ijgc- 2022-004026 Schrock AB, Ouyang C, Sandhu J, Sokol E, Jin D, Ross JS, et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-highmetastatic colorectal cancer. Ann Oncol, 2019, 30:1096–103., DOI 10.1093/ annonc/mdz134 Graf RP, Fisher V, Creeden J, Schrock AB, Ross JS, Nimeiri H, et al. Real-world validation of TMB and microsatellite instability as predictive biomarkers of immune checkpoint inhibitor effectiveness in advanced gastroesophageal cancer. Cancer Res Commun, 2022, 2:1037–48., DOI 10.1158/2767-9764.CRC-22-0161 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611719 EP 1611730 DI 10.3389/pore.2024.1611719 PG 12 ER PT J AU Cegledi, A Batai, Dolgos, J Fekete, M Gopcsa, L Kiraly, V Lakatos, G Nagy, Gy Szemlaky, Zs Varkonyi, A Vilimi, B Mikala, G Bodo, I AF Cegledi, Andrea Batai, Arpad Dolgos, Janos Fekete, Monika Gopcsa, Laszlo Kiraly, Viktoria Lakatos, Gergely Nagy, Gyorgy Szemlaky, Zsuzsanna Varkonyi, Andrea Vilimi, Beata Mikala, Gabor Bodo, Imre TI Case Report: Effective management of adalimumab-induced acquired hemophilia A with the CyDRI protocol SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE adalimumab; acquired hemophilia A; bleeding disorder; immunosuppression; rheumatoid arthritis ID adalimumab; acquired hemophilia A; bleeding disorder; immunosuppression; rheumatoid arthritis AB Introduction: Acquired Hemophilia A (AHA) is a rare autoimmune disorder characterized by the emergence of inhibitors that specifically target coagulation Factor VIII, frequently resulting in severe bleeding episodes. Methods: We conducted a retrospective analysis of the medical records of a 68-year-old male patient who presented with adalimumabinduced AHA. Results: The patient received adalimumab, a tumor necrosis factor inhibitor antibody, as part of his treatment for rheumatoid arthritis. The patient’s clinical journey, characterized by intense bleeding and coagulopathy, was effectively managed with the application of recombinant Factor VIIa (rFVIIa) and the CyDRi protocol. Discussion: The case emphasizes the importance of prompt coagulation assessment in patients with bleeding symptoms receiving disease-modifying therapy for rheumatoid arthritis that includes adalimumab therapy, considering the rare yet life-threatening nature of AHA. Additionally, this report provides an extensive review of the existing literature on druginduced AHA, with a special emphasis on cases linked to immunomodulatory medications. Through this two-pronged approach, our report aims to enhance understanding and awareness of this severe complication among healthcare providers, promoting timely diagnosis and intervention. C1 [Cegledi, Andrea] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Batai, Arpad] Fejer County Szent Gyorgy HospitalSzekesfehervar, Hungary. [Dolgos, Janos] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Fekete, Monika] Semmelweis University, Department of Public HealthBudapest, Hungary. [Gopcsa, Laszlo] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Kiraly, Viktoria] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Lakatos, Gergely] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Nagy, Gyorgy] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Szemlaky, Zsuzsanna] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Varkonyi, Andrea] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Vilimi, Beata] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Mikala, Gabor] Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios OsztalyBudapest, Hungary. [Bodo, Imre] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. RP Cegledi, A (reprint author), Del-pesti Centrumkorhaz – Orszagos Hematologiai es Infektologiai Intezet, Hematologiai es Ossejt-transzplantacios Osztaly, Budapest, Hungary. EM ceglediandi@freemail.hu CR Knoebl P, Marco P, Baudo F, Collins P, Huth-Kuhne A, Nemes L, et al. Demographic and clinical data in acquired hemophilia A: results from the European Acquired Haemophilia Registry, EACH2). J Thromb Haemost, 2012, 10:622–31., DOI 10.1111/j.1538-7836.2012.04654.x Collins PW, Hirsch S, Baglin TP, Dolan G, Hanley J, Makris M, et al. Acquired hemophilia A in the United Kingdom: a 2-year national surveillance study by the United Kingdom haemophilia centre doctors’ organisation. Blood, 2007, 109: 1870–7., DOI 10.1182/blood-2006-06-029850 Tian C, Perija B, Kotb R, Houston BL, Israels SJ, Houston DS, et al. Acquired haemophilia A: a 15-year population-based review of incidence rate, patient demographics and treatment outcomes. Haemophilia, 2023, 29:1269–75., DOI 10. 1111/hae.14845 Jayakar JP, O’Neill N, Yan M, Nisenbaum R, Garvey MB, Teitel J, et al. Retrospective review of Acquired Haemophilia A from the largest Canadian Haemophilia treatment centre. Haemophilia, 2018, 24:e383–e387., DOI 10.1111/ hae.13598 Tiede A, Wahler S. The rising incidence of acquired haemophilia A in Germany. Haemophilia, 2021, 27:e466–e468., DOI 10.1111/hae.14149 Yamaguchi T, Itoh M, Umezawa Y, Asahina A, Hanabusa H, Nakagawa H. Acquired hemophilia A and fulminant diabetes mellitus possibly caused by adalimumab in a patient with psoriatic arthritis. J Dermatol, 2017, 44:e3–e4., DOI 10.1111/1346-8138.13468 Liberman P, Burkholder BM. Adalimumab-associated acquired hemophilia in a patient with scleritis. Ocul Immunol Inflamm, 2022, 30:294–6., DOI 10.1080/ 09273948.2020.1808227 Arthanari S, Ahmad H, Nisar M. Fatal acquired hemophilia A in a patient with rheumatoid arthritis treated with adalimumab. J Clin Rheumatol, 2012, 18:50–1., DOI 10.1097/RHU.0b013e31823ee3cd Franchini M, Capra F, Nicolini N, Veneri D, Manzato F, Baudo F, et al. Druginduced anti-factor VIII antibodies: a systematic review. Med Sci Monit, 2007, 13: RA55–61. Konstantinov K, Dolladille C, Gillet B, Alexandre J, Aouba A, Deshayes S, et al. Drug-associated acquired hemophilia A: an analysis based on 185 cases from the WHO pharmacovigilance database. Haemophilia, 2023, 29:186–92., DOI 10.1111/ hae.14692 Haj M, Dasani H, Kundu S, Mohite U, Collins PW. Acquired haemophilia A may be associated with clopidogrel. BMJ, 2004, 329:323., DOI 10.1136/bmj.329. 7461.323 Hwang HW, Kong JH, Yu DW, Kim WT, Kim HS, Lee CI. A patient with acquired hemophilia A induced by clopidogrel. Korean J Hematol, 2012, 47:80–2., DOI 10.5045/kjh.2012.47.1.80 Madeley J, Hodges G, Birchley A. Development of acquired haemophilia A in a patient treated with alemtuzumab for multiple sclerosis. BMJ Case Rep, 2018, 2018:bcr2018226588., DOI 10.1136/bcr-2018-226588 Pisa M, Della Valle P, Coluccia A, Martinelli V, Comi G, D’Angelo A, et al. Acquired haemophilia A as a secondary autoimmune disease after alemtuzumab treatment in multiple sclerosis: a case report. Mult Scler Relat Disord, 2019, 27: 403–5., DOI 10.1016/j.msard.2018.11.029 Brink HS, Moll W, Sandberg Y. Acquired haemophilia A after alemtuzumab treatment of multiple sclerosis. Br J Haematol, 2020, 190:13., DOI 10.1111/bjh.16644 van der Zwan M, Leebeek FWG, Sandberg Y, Kruip M, Hesselink DA. Acquired haemophilia A after alemtuzumab therapy. Haemophilia, 2020, 26: e337–e339., DOI 10.1111/hae.14107 Gounder K, Batt T, Dreyer M. Two case reports of acquired haemophilia A as complications of alemtuzumab treatment for multiple sclerosis. BMJ Neurol Open, 2021, 3:e000095., DOI 10.1136/bmjno-2020-000095 Fawaz H, Hodroj MH, Tarhini H, Trad CO, Taher A. Alemtuzumab induced acquired hemophilia A inmultiple sclerosis: a case report. Ann Hematol, 2023, 102: 3271–3., DOI 10.1007/s00277-023-05370-8 Mangin MA, Lienhart A, Gouraud A, Roux S, Hodique F, Jouen F, et al. Onset of acquired haemophilia A after omalizumab treatment in severe bullous pemphigoid - a report on two cases successfully treated with mycophenolate mofetil. Ann Dermatol Venereol, 2021, 148:57–9., DOI 10.1016/j.annder.2020.09.577 El-Osta H, Reddy P, Deutsch JM. Acquired factor VIII inhibitors: case reports of paclitaxel and penicillin-induced entities. Blood Coagul Fibrinolysis, 2009, 20: 719–21., DOI 10.1097/MBC.0b013e32832fb1b9 Takeyama M, Nogami K, Kajimoto T, Ogiwara K, Matsumoto T, Shima M. First report of real-time monitoring of coagulation function potential and IgG subtype of anti-FVIII autoantibodies in a child with acquired hemophilia A associated with streptococcal infection and amoxicillin. Int J Hematol, 2018, 107:112–6., DOI 10.1007/s12185-017-2273-6 Moraca RJ, Ragni MV. Acquired anti-FVIII inhibitors in children. Haemophilia, 2002, 8:28–32., DOI 10.1046/j.1365-2516.2002.00574.x Campos-de-Magalhaes M, Eduardo Brandao-Mello C, Lucia Elias Pires M, Cecilia da Fonseca Salgado M, Barcelo de Brito S, Jose de Almeida A. Factor VIII and IX deficiencies related to acquired inhibitors in a patient with chronic hepatitis C virus infection receiving treatment with pegylated interferon plus ribavirin. Hematology, 2011, 16:80–5., DOI 10.1179/102453311X12902908412156 Zheng WL, Zhang GS, Shen JK, PeiMF, Peng HL, Xu M. Acquired factor VIII inhibitor as presentation of chronic myelogenous leukemia during interferon-alpha therapy. Thromb Res, 2011, 128:202–3., DOI 10.1016/j.thromres.2011.04.014 Goyal J, Tyagi P, Kumar N. Acquired haemophilia in a patient treated with interferon-α for hepatitis C infection. Haemophilia, 2012, 18:e73–74., DOI 10.1111/j. 1365-2516.2012.02775.x Nayar S, Parvathi KBE, Kaliannan M, Sivasailam P. Acquired inhibitor of factor VIII presenting as delayed wound healing. J Clin Diagn Res, 2017, 11: ED21–ED23., DOI 10.7860/JCDR/2017/21598.10109 Famularo G, De Maria S, Minisola G, Nicotra GC. Severe acquired hemophilia with factor VIII inhibition associated with acetaminophen and chlorpheniramine. Ann Pharmacother, 2004, 38:1432–4., DOI 10.1345/aph.1E100 Comini-Frota ER, Campos APF, Neto APG, Christo PP. Acquired hemophilia A and other autoimmune diseases after alemtuzumab therapy for multiple sclerosis: a report of two cases. Mult Scler Relat Disord, 2020, 44:102181., DOI 10.1016/j.msard. 2020.102181 McCaughan G, Massey J, Sutton I, Curnow J. Acquired haemophilia A complicating alemtuzumab therapy for multiple sclerosis. BMJ Case Rep, 2017, 2017:bcr2017223016., DOI 10.1136/bcr-2017-223016 Massey J, Barnett Y, Curnow J, Sutton I. B cell depletion therapy resulting in sustained remission of severe autoimmune complications following Alemtuzumab treatment of Multiple Sclerosis. Mult Scler Relat Disord, 2019, 35:100–3., DOI 10. 1016/j.msard.2019.07.016 Kato R, Hayashi H, Sano K, Handa K, Kumode T, Ueda H, et al. Nivolumabinduced hemophilia A presenting as gastric ulcer bleeding in a patient with nsclc. J Thorac Oncol, 2018, 13:e239–41., DOI 10.1016/j.jtho.2018.06.024 Gokozan HN, Friedman JD, Schmaier AH, Downes KA, Farah LA, Reeves HM. Acquired hemophilia A after nivolumab therapy in a patient with metastatic squamous cell carcinoma of the lung successfully managed with rituximab. Clin Lung Cancer, 2019, 20:e560–3., DOI 10.1016/j.cllc.2019.06.022 Gidaro A, Palmieri G, Donadoni M, Mameli LA, La Cava L, Sanna G, et al. A diagnostic of acquired hemophilia following PD1/PDL1 inhibitors in advanced melanoma: the experience of two patients and a literature review. Diagnostics, Basel,, 2022, 12:2559., DOI 10.3390/diagnostics12102559 Banse C, Benhamou Y, Lequerre T, Le Cam-Duchez V, Levesque H, Vittecoq O. Acquired hemophilia possibly induced by etanercept in a patient with rheumatoid arthritis. Jt Bone Spine, 2015, 82:200–2., DOI 10.1016/j.jbspin.2014. 12.003 Singh P, Szaraz-Szeles M, Mezei Z, Barath S, Hevessy Z. Age-dependent frequency of unconventional T cells in a healthy adult Caucasian population: a combinational study of invariant natural killer T cells, γδ T cells, and mucosaassociated invariant T cells. Geroscience, 2022, 44:2047–60., DOI 10.1007/s11357- 022-00515-5 Matacchione G, Perugini J, Di Mercurio E, Sabbatinelli J, Prattichizzo F, Senzacqua M, et al. Senescent macrophages in the human adipose tissue as a source of inflammaging. Geroscience, 2022, 44:1941–60., DOI 10.1007/s11357-022-00536-0 ZampinoM, Brennan NA, Kuo PL, Spencer RG, Fishbein KW, Simonsick EM, et al. Poor mitochondrial health and systemic inflammation? Test of a classic hypothesis in the Baltimore Longitudinal Study of Aging. Geroscience, 2020, 42: 1175–82., DOI 10.1007/s11357-020-00208-x Pandics T, Major D, Fazekas-Pongor V, Szarvas Z, Peterfi A, Mukli P, et al. Exposome and unhealthy aging: environmental drivers from air pollution to occupational exposures. Geroscience, 2023, 45:3381–408., DOI 10.1007/s11357- 023-00913-3 Urban VS, Cegledi A,Mikala G. Multiple myeloma, a quintessential malignant disease of aging: a geroscience perspective on pathogenesis and treatment. Geroscience, 2023, 45:727–46., DOI 10.1007/s11357-022-00698-x Pardos-Gea J, Martin-Fernandez L, Closa L, Ferrero A, Marzo C, Rubio-Rivas M, et al. Key genes of the immune system and predisposition to acquired hemophilia A: evidence from a Spanish cohort of 49 patients using nextgeneration sequencing. Int J Mol Sci, 2023, 24:16372., DOI 10.3390/ijms242216372 Pavlova A, Zeitler H, Scharrer I, Brackmann HH, Oldenburg J. HLA genotype in patients with acquired haemophilia A. Haemophilia, 2010, 16:107–12., DOI 10. 1111/j.1365-2516.2008.01976.x Pavlova A, Diaz-Lacava A, Zeitler H, Satoguina J, Niemann B, Krause M, et al. Increased frequency of the CTLA-4 49 A/G polymorphism in patients with acquired haemophilia A compared to healthy controls. Haemophilia, 2008, 14:355–60., DOI 10.1111/j.1365-2516.2007.01618.x Oldenburg J, Zeitler H, Pavlova A. Genetic markers in acquired haemophilia. Haemophilia, 2010, 16(3):41–5., DOI 10.1111/j.1365-2516.2010.02259.x Scheinfeld N. Adalimumab: a review of side effects. Expert Opin Drug Saf, 2005, 4:637–41., DOI 10.1517/14740338.4.4.637 Lie MR, Kreijne JE, van derWoude CJ. Sex is associated with adalimumab side effects and drug survival in patients with Crohn’s disease. Inflamm Bowel Dis, 2017, 23:75–81., DOI 10.1097/MIB.0000000000000981 Goel K, BunkerM, Balog A, Silverman JF. Fulminant herpes simplex hepatitis secondary to adalimumab in Crohn’s disease: a case report. Clin Med Insights Case Rep, 2019, 12:1179547619858979., DOI 10.1177/1179547619858979 Madani A, Almuhaideb Q. Adalimumab therapy in a patient with psoriasis, down syndrome, and concomitant hepatitis B virus infection. Biologics, 2021, 15: 375–8., DOI 10.2147/BTT.S317888 Bensouda-Grimaldi L, Mulleman D, Valat JP, Autret-Leca E. Adalimumabassociated multiple sclerosis. J Rheumatol, 2007, 34:239–40. Matsumoto T, Nakamura I, Miura A, Momoyama G, Ito K. New-onset multiple sclerosis associated with adalimumab treatment in rheumatoid arthritis: a case report and literature review. Clin Rheumatol, 2013, 32:271–5., DOI 10.1007/ s10067-012-2113-2 Uygunoglu U, Uluduz D, Tascilar K, Saip S. Multiple sclerosis during adalimumab treatment in a case with ankylosing spondylitis. Rheumatol Int, 2014, 34:141–3., DOI 10.1007/s00296-012-2625-8 Baresic M, Reihl CrnogajM, Zadro I, Anic B. Demyelinating disease, multiple sclerosis, in a patient with psoriatic arthritis treated with adalimumab: a case-based review. Rheumatol Int, 2021, 41:2233–9., DOI 10.1007/s00296-021-04995-0 Sokmen O, Gocmen R, Tuncer A. Multiple sclerosis - like demyelinating lesions during adalimumab treatment in a case with Crohn’s disease. Noro Psikiyatr Ars, 2022, 59:342–4., DOI 10.29399/npa.27973 Rojas-Carabali W, Boada-Robayo L, Chacon-Zambrano D, Criollo Porras E, Kerguelen Dumar V, de-la-Torre A. Multiple sclerosis in a patient with intermediate uveitis and juvenile idiopathic arthritis treated with adalimumab: a case report. Ocul Immunol Inflamm, 2023, 31:1873–6., DOI 10.1080/09273948.2022. 2113800 Mansito Lopez C, Torres Laboy P, Ortiz BM, Quintero Noriega A, Cintron Rivera V. Fatal new-onset congestive heart failure related to adalimumab use in a patient with relapsing hidradenitis suppurativa: a case report. Am J Case Rep, 2021, 22:e929148., DOI 10.12659/AJCR.929148 Hagel S, Bruns T, Theis B, Herrmann A, Stallmach A. Subacute liver failure induced by adalimumab. Int J Clin Pharmacol Ther, 2011, 49:38–40., DOI 10.5414/ cpp49038 Harada Y, Yamamoto H, Sato M, Kodaira M, Kono T. Autoimmune hemolytic anemia during adalimumab treatment for plaque psoriasis. Intern Med, 2015, 54:1103–4., DOI 10.2169/internalmedicine.54.3433 Nagashima T, Minota S. Autoimmune hemolytic anemia induced by adalimumab. Intern Med, 2016, 55:715., DOI 10.2169/internalmedicine.55.5773 Sanchez-Pujol MJ, Docampo-Simon A, Moscardo C, Betlloch-Mas I. Adalimumab-induced hemolytic anemia in a girl with psoriasis. Dermatol Ther, 2020, 33:e13711., DOI 10.1111/dth.13711 Schep SJ, van Dijk WEM, Beckers EAM, Meijer K, Coppens M, Eikenboom J, et al. Treatment of acquired hemophilia A, a balancing act: results from a 27-year Dutch cohort study. Am J Hematol, 2021, 96:51–9., DOI 10.1002/ajh.26009 Miatech JL, Kantamani D, Stagg MP. Management of acquired factor VIII inhibitors with NovoSeven and obizur. Cureus, 2021, 13:e19145., DOI 10.7759/ cureus.19145 Kruse-Jarres R, Kempton CL, Baudo F, Collins PW, Knoebl P, Leissinger CA, et al. Acquired hemophilia A: updated review of evidence and treatment guidance. Am J Hematol, 2017, 92:695–705., DOI 10.1002/ajh.24777 Tiede A, Klamroth R, Scharf RE, Trappe RU, Holstein K, Huth-Kuhne A, et al. Prognostic factors for remission of and survival in acquired hemophilia A, AHA): results from the GTH-AH 01/2010 study. Blood, 2015, 125:1091–7., DOI 10.1182/ blood-2014-07-587089 Simon B, Cegledi A, Dolgos J, Farkas P, Gaddh M, Hanko L, et al. Combined immunosuppression for acquired hemophilia A: CyDRi is a highly effective low-toxicity regimen. Blood, 2022, 140:1983–92., DOI 10.1182/blood. 2022016873 Collins P, Baudo F, Knoebl P, Levesque H, Nemes L, Pellegrini F, et al. Immunosuppression for acquired hemophilia A: results from the European acquired haemophilia registry, EACH2). Blood, 2012, 120:47–55., DOI 10.1182/ blood-2012-02-409185 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611720 EP 1611727 DI 10.3389/pore.2024.1611720 PG 8 ER PT J AU Toth, JL Mokanszki, A Mehes, G AF Toth, Jozsef Laszlo Mokanszki, Attila Mehes, Gabor TI The rapidly changing field of predictive biomarkers of non-small cell lung cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE NSCLC; driver oncogenes; immune checkpoint inhibitor; gene fusion; biomarker ID NSCLC; driver oncogenes; immune checkpoint inhibitor; gene fusion; biomarker AB Lung cancer is a leading cause of cancer-related death worldwide in both men and women, however mortality in the US and EU are recently declining in parallel with the gradual cut of smoking prevalence. Consequently, the relative frequency of adenocarcinoma increased while that of squamous and small cell carcinomas declined. During the last two decades a plethora of targeted drug therapies have appeared for the treatment of metastasizing non-small cell lung carcinomas (NSCLC). Personalized oncology aims to precisely match patients to treatments with the highest potential of success. Extensive research is done to introduce biomarkers which can predict the effectiveness of a specific targeted therapeutic approach. The EGFR signaling pathway includes several sufficient targets for the treatment of human cancers including NSCLC. Lung adenocarcinoma may harbor both activating and resistance mutations of the EGFR gene, and further, mutations of KRAS and BRAF oncogenes. Less frequent but targetable genetic alterations include ALK, ROS1, RET gene rearrangements, and various alterations of MET proto-oncogene. In addition, the importance of anti-tumor immunity and of tumor microenvironment has become evident recently. Accumulation of mutations generally trigger tumor specific immune defense, but immune protection may be upregulated as an aggressive feature. The blockade of immune checkpoints results in potential reactivation of tumor cell killing and induces significant tumor regression in various tumor types, such as lung carcinoma. Therapeutic responses to anti PD1-PD-L1 treatment may correlate with the expression of PD-L1 by tumor cells. Due to the wide range of diagnostic and predictive features in lung cancer a plenty of tests are required from a single small biopsy or cytology specimen, which is challenged by major issues of sample quantity and quality. Thus, the efficacy of biomarker testing should be warranted by standardized policy and optimal material usage. In this review we aim to discuss major targeted therapy-related biomarkers in NSCLC and testing possibilities comprehensively. C1 [Toth, Jozsef Laszlo] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Mokanszki, Attila] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. RP Toth, JL (reprint author), University of Debrecen, Faculty of Medicine, Department of Pathology, Debrecen, Hungary. EM tothlasz@med.unideb.hu CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71:209–49., DOI 10.3322/caac.21660 Islami F, Torre LA, Jemal A. Global trends of lung cancer mortality and smoking prevalence. Transl Lung Cancer Res, 2015, 4:327–38., DOI 10.3978/j.issn. 2218-6751.2015.08.04 Bogos K, Kiss Z, Galffy G, Tamasi L, Ostoros G, Muller V, et al. Revising incidence and mortality of lung cancer in central Europe: an epidemiology review from Hungary. Front Oncol, 2019, 9:1051., DOI 10.3389/fonc.2019.01051 Tsao MS, Asamura H, Borczuk AC, Dacic S, Devesa SS, Kerr KM, et al. Tumours of the lung. Introduction. In WHO classification of tumours editorial board. In: Thoracic tumours. In: WHO classification of tumours series. 5th ed., 5. Lyon: IASCL Press, 2021). p. 20–8. Zhang Y, Vaccarella S, Morgan E, Li M, Etxeberria J, Chokunonga E, et al. Global variations in lung cancer incidence by histological subtype in 2020: a population-based study. Lancet Oncol, 2023, 24:1206–18., DOI 10.1016/S1470- 2045(23)00444-8 Brooks DR, Austin JH, Heelan RT, Ginsberg MS, Shin V, Olson SH, et al. Influence of type of cigarette on peripheral versus central lung cancer. Cancer Epidemiol Biomarkers Prev, 2005, 14:576–81., DOI 10.1158/1055-9965.EPI-04-0468 Ito H, Matsuo K, Tanaka H, Koestler DC, Ombao H, Fulton J, et al. Nonfilter and filter cigarette consumption and the incidence of lung cancer by histological type in Japan and the United States: analysis of 30-year data from population-based cancer registries. Int J Cancer, 2011, 128:1918–28., DOI 10. 1002/ijc.25531 Stellman SD, Muscat JE, Hoffmann D, Wynder EL. Impact of filter cigarette smoking on lung cancer histology. Prev Med, 1997, 26:451–6., DOI 10.1006/pmed. 1997.0212 Hoffmann D, Hoffmann I. The changing cigarette, 1950-1995. J Toxicol Environ Health, 1997, 50:307–64., DOI 10.1080/009841097160393 Everaert S, Schoeters G, Lardon F, Janssens A, Van Larebeke N, Raquez JM, et al. Protecting public health and the environment: towards a general ban on cellulose acetate cigarette filters in the European Union. Front Public Health, 2023, 11:1282655., DOI 10.3389/fpubh.2023.1282655 Rudin CM, Poirier JT, Byers LA, Dive C, Dowlati A, George J, et al. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer, 2019, 19:289–97., DOI 10.1038/s41568-019-0133-9 Yatabe Y. Molecular pathology of non-small cell carcinoma. Histopathology., 2024, 84:50–66., DOI 10.1111/his.15080 Lu T, Yang X, Huang Y, Zhao M, Li M, Ma K, et al. Trends in the incidence, treatment, and survival of patients with lung cancer in the last four decades. Cancer Manag Res, 2019, 11:943–53., DOI 10.2147/CMAR.S187317 Lofling L, Bahmanyar S, Kieler H, LambeM,Wagenius G. Temporal trends in lung cancer survival: a population-based study. Acta Oncologica, 2022, 61:625–31., DOI 10.1080/0284186X.2021.2013529 Kiss Z, Galffy G, Muller V, Moldvay J, Sarosi V, Papai-Szekely Z, et al. Significant changes in advanced lung cancer survival during the past decade in Hungary: impact of modern immunotherapy and the COVID-19 pandemic. Front Oncol, 2023, 13:1207295., DOI 10.3389/fonc.2023.1207295 Mendelsohn J. The epidermal growth factor receptor as a target for cancer therapy. Endocr Relat Cancer, 2001, 8:3–9., DOI 10.1677/erc.0.0080003 Baselga J, Albanell J. Targeting epidermal growth factor receptor in lung cancer. Curr Oncol Rep, 2002, 4:317–24., DOI 10.1007/s11912-002-0007-1 Rusch V, Klimstra D, Venkatraman E, Pisters PW, Langenfeld J, Dmitrovsky E. Overexpression of the epidermal growth factor receptor and its ligand transforming growth factor alpha is frequent in resectable non-small cell lung cancer but does not predict tumor progression. Clin Cancer Res, 1997, 3:515–22. PMID: 9815714. Ciardiello F, De Vita F, Orditura M, Tortora G. The role of EGFR inhibitors in nonsmall cell lung cancer. Curr Opin Oncol, 2004, 16:130–5., DOI 10.1097/ 00001622-200403000-00008 Herbst RS,Maddox AM, Rothenberg ML, Small EJ, Rubin EH, Baselga J, et al. Selective oral epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 is generally well-tolerated and has activity in non-small-cell lung cancer and other solid tumors: results of a phase I trial. J Clin Oncol, 2002, 20:3815–25., DOI 10.1200/ JCO.2002.03.038 Fukuoka M, Yano S, Giaccone G, Tamura T, Nakagawa K, Douillard JY, et al. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non–small-cell lung cancer. J Clin Oncol, 2023, 41:1162–71., DOI 10.1200/JCO.22.02499 Janne PA, Gurubhagavatula S, Yeap BY, Lucca J, Ostler P, Skarin AT, et al. Outcomes of patients with advanced non-small cell lung cancer treated with gefitinib, ZD1839, "Iressa", on an expanded access study. Lung Cancer, 2004, 44:221–30., DOI 10.1016/j.lungcan.2003.12.014 Bailey R, Kris M, Wolf M, Kay A, Averbuch S, Askaa J, et al. Gefitinib, ‘Iressa’, ZD1839, monotherapy for pretreated advanced non-small-cell lung cancer in IDEAL 1 and 2: tumor response is not clinically relevantly predictable from tumor EGFR membrane staining alone. Lung Cancer, 2003, 41:S2–S71., DOI 10. 1016/S0169-5002(03)91900-3 Parra HS, Cavina R, Latteri F, Zucali PA, Campagnoli E, Morenghi E, et al. Analysis of epidermal growth factor receptor expression as a predictive factor for response to gefitinib, ’Iressa’, ZD1839, in non-small-cell lung cancer. Br J Cancer, 2004, 91:208–12., DOI 10.1038/sj.bjc.6601923 Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science, 2004, 304:1497–500., DOI 10.1126/science.1099314 Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med, 2004, 350: 2129–39., DOI 10.1056/NEJMoa040938 Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I, et al. EGF receptor gene mutations are common in lung cancers from "never smokers" and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci USA, 2004, 101:13306–11., DOI 10.1073/pnas.0405220101 Tang X, Shigematsu H, Bekele BN, Roth JA,Minna JD, Hong WK, et al. EGFR tyrosine kinase domain mutations are detected in histologically normal respiratory epithelium in lung cancer patients. Cancer Res, 2005, 65(17):7568–72., DOI 10.1158/ 0008-5472.CAN-05-1705 Graham RP, Treece AL, Lindeman NI, Vasalos P, Shan M, Jennings LJ, et al. Worldwide frequency of commonly detected EGFR mutations. Arch Pathol Lab Med, 2018, 142:163–7., DOI 10.5858/arpa.2016-0579-CP Sharma M, Basu DA, Nathany S, Amrith BP, Batra U. A narrative review of the role of common EGFR mutations in pathogenesis and treatment of non-smallcell lung carcinoma. Cancer Res Stat Treat, 2022, 5:507–18., DOI 10.4103/crst.crst_ 222_22 Yun CH, Boggon TJ, Li Y, Woo MS, Greulich H, Meyerson M, et al. Structures of lung cancer-derived EGFR mutants and inhibitor complexes: mechanism of activation and insights into differential inhibitor sensitivity. Cancer Cell, 2007, 11: 217–27., DOI 10.1016/j.ccr.2006.12.017 Shan Y, Eastwood MP, Zhang X, Kim ET, Arkhipov A, Dror RO, et al. Oncogenic mutations counteract intrinsic disorder in the EGFR kinase and promote receptor dimerization. Cell, 2012, 149(4):860–70., DOI 10.1016/j.cell. 2012.02.063 Harrison PT, Vyse S, Huang PH. Rare epidermal growth factor receptor, EGFR, mutations in non-small cell lung cancer. Semin Cancer Biol, 2020, 61: 167–79., DOI 10.1016/j.semcancer.2019.09.015 Kobayashi Y, Mitsudomi T. Not all epidermal growth factor receptor mutations in lung cancer are created equal: perspectives for individualized treatment strategy. Cancer Sci, 2016, 107:1179–86., DOI 10.1111/cas.12996 Yasuda H, Park E, Yun CH, Sng NJ, Lucena-Araujo AR, Yeo WL, et al. Structural, biochemical, and clinical characterization of epidermal growth factor receptor, EGFR, exon 20 insertion mutations in lung cancer. Sci Transl Med, 2013, 5(216):216ra177., DOI 10.1126/scitranslmed.3007205 Voon PJ, Tsui DW, Rosenfeld N, Chin TM. EGFR exon 20 insertion A763- Y764insFQEA and response to erlotinib--Letter. Mol Cancer Ther, 2013, 12: 2614–5., DOI 10.1158/1535-7163.MCT-13-0192 Bai Q, Wang J, Zhou X. EGFR exon20 insertion mutations in non-small cell lung cancer: clinical implications and recent advances in targeted therapies. Cancer Treat Rev, 2023, 120:102605., DOI 10.1016/j.ctrv.2023.102605 Yang G, Yang Y, Hu J, Xu H, Zhang S, Wang Y. EGFR exon 20 insertion variants A763_Y764insFQEA and D770delinsGY confer favorable sensitivity to currently approved EGFR-specific tyrosine kinase inhibitors. Front Pharmacol, 2022, 13:984503., DOI 10.3389/fphar.2022.984503 Liu M, Niu X, Liu H, Chen J. Germline EGFR mutations in lung cancer, Review). Oncol Lett, 2023, 26:282., DOI 10.3892/ol.2023.13868 Ji H, Zhao X, Yuza Y, Shimamura T, Li D, Protopopov A, et al. Epidermal growth factor receptor variant III mutations in lung tumorigenesis and sensitivity to tyrosine kinase inhibitors. Proc Natl Acad Sci USA, 2006, 103:7817–22., DOI 10.1073/ pnas.0510284103 Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non–small-cell lung cancer with mutated EGFR. N Engl J Med, 2010, 362:2380–8., DOI 10.1056/NEJMoa0909530 Laface C, Maselli FM, Santoro AN, Iaia ML, Ambrogio F, Laterza M, et al. The resistance to EGFR-TKIs in non-small cell lung cancer: from molecular mechanisms to clinical application of new therapeutic strategies. Pharmaceutics, 2023, 15:1604., DOI 10.3390/pharmaceutics15061604 Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med, 2011, 3:75ra26., DOI 10.1126/scitranslmed. 3002003 Yu L, Bazhenova L, Gold K, Tran L, Hilburn V, Vu P, et al. Clinicopathologic and molecular characteristics of EGFR-mutant lung adenocarcinomas that transform to small cell lung cancer after TKI therapy. Transl Lung Cancer Res, 2022, 11:452–61., DOI 10.21037/tlcr-21-665 Hill W, Lim EL, Weeden CE, Lee CL, Augustine M, Chen K, et al. Lung adenocarcinoma promotion by air pollutants. Nature, 2023, 616:159–67., DOI 10. 1038/s41586-023-05874-3 Ruano-Ravina A, Torres-Duran M, Kelsey KT, Parente-Lamelas I, Leiro- Fernandez V, Abdulkader I, et al. Residential radon, EGFR mutations and ALK alterations in never-smoking lung cancer cases. Eur Respir J, 2016, 48:1462–70., DOI 10.1183/13993003.00407-2016 Sholl LM, Xiao Y, Joshi V, Yeap BY, Cioffredi LA, Jackman DM, et al. EGFR mutation is a better predictor of response to tyrosine kinase inhibitors in non-small cell lung carcinoma than FISH, CISH, and immunohistochemistry. Am J Clin Pathol, 2010, 133:922–34., DOI 10.1309/AJCPST1CTHZS3PSZ Yu J, Kane S, Wu J, Benedettini E, Li D, Reeves C, et al. Mutation-specific antibodies for the detection of EGFR mutations in non-small-cell lung cancer. Clin Cancer Res, 2009, 15:3023–8., DOI 10.1158/1078-0432.CCR-08-2739 Bondgaard AL, Hogdall E, Mellemgaard A, Skov BG. High specificity but low sensitivity of mutation-specific antibodies against EGFR mutations in nonsmall- cell lung cancer. Mod Pathol, 2014, 27:1590–8., DOI 10.1038/modpathol. 2014.67 Kato Y, Peled N, Wynes MW, Yoshida K, Pardo M, Mascaux C, et al. Novel epidermal growth factor receptor mutation-specific antibodies for non-small cell lung cancer: immunohistochemistry as a possible screening method for epidermal growth factor receptor mutations. J Thorac Oncol, 2010, 5:1551–8., DOI 10.1097/ JTO.0b013e3181e9da60 Brevet M, Arcila M, Ladanyi M. Assessment of EGFR mutation status in lung adenocarcinoma by immunohistochemistry using antibodies specific to the two major forms of mutant EGFR. J Mol Diagn, 2010, 12:169–76., DOI 10.2353/jmoldx. 2010.090140 Seo AN, Park TI, Jin Y, Sun PL, Kim H, Chang H, et al. Novel EGFRmutationspecific antibodies for lung adenocarcinoma: highly specific but not sensitive detection of an E746_A750 deletion in exon 19 and an L858R mutation in exon 21 by immunohistochemistry. Lung Cancer, 2014, 83:316–23., DOI 10.1016/j. lungcan.2013.12.008 Ragazzi M, Tamagnini I, Bisagni A, Cavazza A, Pagano M, Baldi L, et al. Diamond: immunohistochemistry versus sequencing in EGFR analysis of lung adenocarcinomas. J Clin Pathol, 2016, 69:440–7., DOI 10.1136/jclinpath-2015- 203348 Warth A, Penzel R, Brandt R, Sers C, Fischer JR, Thomas M, et al. Optimized algorithm for Sanger sequencing-based EGFR mutation analyses in NSCLC biopsies. Virchows Arch, 2012, 460:407–14., DOI 10.1007/s00428-012-1219-x Bos JL. Ras oncogenes in human cancer: a review. Cancer Res, 1989, 49: 4682–9. Erratum in: Cancer Res, 1990, 50: 1352. PMID: 2547513. Chang EH, Gonda MA, Ellis RW, Scolnick EM, Lowy DR. Human genome contains four genes homologous to transforming genes of Harvey and Kirsten murine sarcoma viruses. Proc Natl Acad Sci U S A., 1982, 79:4848–52., DOI 10.1073/ pnas.79.16.4848 McBride OW, Swan DC, Tronick SR, Gol R, Klimanis D, Moore DE, et al. Regional chromosomal localization of N-ras, K-ras-1, K-ras-2 and myb oncogenes in human cells. Nucleic Acids Res, 1983, 11:8221–36., DOI 10.1093/nar/11.23.8221 Barbacid M. Ras genes. Annu Rev Biochem, 1987, 56:779–827., DOI 10.1146/ annurev.bi.56.070187.004023 Karnoub AE, Weinberg RA. Ras oncogenes: split personalities. Nat Rev Mol Cell Biol, 2008, 9:517–31., DOI 10.1038/nrm2438 Pantsar T. The current understanding of KRAS protein structure and dynamics. Comput Struct Biotechnol J, 2019, 18:189–98., DOI 10.1016/j.csbj.2019. 12.004 Simanshu DK, Nissley DV, McCormick F. RAS proteins and their regulators in human disease. Cell, 2017, 170:17–33., DOI 10.1016/j.cell.2017.06.009 Bahar ME, Kim HJ, Kim DR. Targeting the RAS/RAF/MAPK pathway for cancer therapy: from mechanism to clinical studies. Signal Transduct Target Ther, 2023, 8:455., DOI 10.1038/s41392-023-01705-z Molina JR, Adjei AA. The Ras/Raf/MAPK pathway. J Thorac Oncol, 2006, 1: 7–9. PMID: 17409820., DOI 10.1016/s1556-0864(15)31506-9 Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, AbrahamRT. The PI3K pathway in human disease. Cell, 2017, 170:605–35., DOI 10.1016/j.cell.2017. 07.029 Mo SP, Coulson JM, Prior IA. RAS variant signalling. Biochem Soc Trans., 2018, 46:1325–32., DOI 10.1042/BST20180173 Prior IA, Hood FE, Hartley JL. The frequency of ras mutations in cancer. Cancer Res, 2020, 80:2969–74., DOI 10.1158/0008-5472.CAN-19-3682 Ihle NT, Byers LA, Kim ES, Saintigny P, Lee JJ, Blumenschein GR, et al. Effect of KRAS oncogene substitutions on protein behavior: implications for signaling and clinical outcome. J Natl Cancer Inst, 2012, 104:228–39., DOI 10.1093/jnci/djr523 Rodenhuis S, Slebos RJ, Boot AJ, Evers SG, Mooi WJ, Wagenaar SS, et al. Incidence and possible clinical significance of K-ras oncogene activation in adenocarcinoma of the human lung. Cancer Res, 1988, 48:5738–41. PMID: 3048648. Suzuki Y, Orita M, Shiraishi M, Hayashi K, Sekiya T. Detection of ras gene mutations in human lung cancers by single-strand conformation polymorphism analysis of polymerase chain reaction products. Oncogene, 1990, 5:1037–43. PMID: 2197591. Dearden S, Stevens J, Wu YL, Blowers D. Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology, mutMap). Ann Oncol, 2013, 24:2371–6., DOI 10.1093/annonc/mdt205 Dogan S, Shen R, Ang DC, Johnson ML, D’Angelo SP, Paik PK, et al. Molecular epidemiology of EGFR and KRAS mutations in 3,026 lung adenocarcinomas: higher susceptibility of women to smoking-related KRASmutant cancers. Clin Cancer Res, 2012, 18:6169–77., DOI 10.1158/1078-0432. CCR-11-3265 Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. Biochemical and structural analysis of common cancer-associated KRAS mutations. Mol Cancer Res, 2015, 13:1325–35., DOI 10.1158/1541-7786.MCR-15-0203 Zafra MP, Parsons MJ, Kim J, Alonso-Curbelo D, Goswami S, Schatoff EM, et al. An in vivo kras allelic series reveals distinct phenotypes of common oncogenic variants. Cancer Discov., 2020, 10:1654–71., DOI 10.1158/2159-8290.CD-20-0442 Timar J, Kashofer K. Molecular epidemiology and diagnostics of KRAS mutations in human cancer. Cancer Metastasis Rev., 2020, 39:1029–38., DOI 10. 1007/s10555-020-09915-5 Wang WZ, Shulman A, Amann JM, Carbone DP, Tsichlis PN. Small cell lung cancer: subtypes and therapeutic implications. Semin Cancer Biol, 2022, 86(Pt 2): 543–54., DOI 10.1016/j.semcancer.2022.04.001 Lee B, Lee T, Lee SH, Choi YL, Han J. Clinicopathologic characteristics of EGFR, KRAS, and ALK alterations in 6,595 lung cancers. Oncotarget, 2016, 7: 23874–84., DOI 10.18632/oncotarget.8074 Kadota K, Yeh YC, D’Angelo SP, Moreira AL, Kuk D, Sima CS, et al. Associations between mutations and histologic patterns of mucin in lung adenocarcinoma: invasive mucinous pattern and extracellular mucin are associated with KRAS mutation. Am J Surg Pathol, 2014, 38:1118–27., DOI 10. 1097/PAS.0000000000000246 Judd J, Abdel KN, Khan H,Naqash AR, Baca Y, Xiu J, et al. Characterization of KRAS mutation subtypes in non-small cell lung cancer. Mol Cancer Ther, 2021, 20: 2577–84., DOI 10.1158/1535-7163.MCT-21-0201 Wu MY, Zhang EW, Strickland MR, Mendoza DP, Lipkin L, Lennerz JK, et al. Clinical and imaging features of non-small cell lung cancer with G12C KRAS mutation. Cancers, Basel,, 2021, 13:3572., DOI 10.3390/cancers13143572 Cui W, Franchini F, Alexander M, Officer A, Wong HL, Ijzerman M, et al. Real world outcomes in KRAS G12C mutation positive non-small cell lung cancer. Lung Cancer, 2020, 146:310–7., DOI 10.1016/j.lungcan.2020.06.030 Gainor JF, Varghese AM, Ou SH, Kabraji S, Awad MM, Katayama R, et al. ALK rearrangements are mutually exclusive with mutations in EGFR or KRAS: an analysis of 1,683 patients with non-small cell lung cancer. Clin Cancer Res, 2013, 19: 4273–81., DOI 10.1158/1078-0432.CCR-13-0318 Zhuang X, Zhao C, Li J, Su C, Chen X, Ren S, et al. Clinical features and therapeutic options in non-small cell lung cancer patients with concomitant mutations of EGFR, ALK, ROS1, KRAS or BRAF. Cancer Med, 2019, 8: 2858–66., DOI 10.1002/cam4.2183 Lantuejoul S, Nicholson AG, Sartori G, Piolat C, Danel C, Brabencova E, et al. Mucinous cells in type 1 pulmonary congenital cystic adenomatoid malformation as mucinous bronchioloalveolar carcinoma precursors. Am J Surg Pathol, 2007, 31: 961–9., DOI 10.1097/01.pas.0000249444.90594.27 Hermelijn SM, Wolf JL, Dorine den Toom T, Wijnen RMH, Rottier RJ, Schnater JM, et al. Early KRAS oncogenic driver mutations in nonmucinous tissue of congenital pulmonary airway malformations as an indicator of potential malignant behavior. Hum Pathol, 2020, 103:95–106., DOI 10.1016/j.humpath. 2020.07.015 Garinet S, Rahshenas M, Galmiche-Rolland L, Abbo O, Bonnard A, Hameury F, et al. Cancer-associated mutations in congenital pulmonary malformations: a prospective cohort. Am J Respir Crit Care Med, 2023, 207:615–9., DOI 10.1164/rccm. 202208-1573LE Ghimessy A, Radeczky P, Laszlo V, Hegedus B, Renyi-Vamos F, Fillinger J, et al. Current therapy of KRAS-mutant lung cancer. Cancer Metastasis Rev., 2020, 39:1159–77., DOI 10.1007/s10555-020-09903-9 Mascaux C, Iannino N, Martin B, PaesmansM, Berghmans T, Dusart M, et al. The role of RAS oncogene in survival of patients with lung cancer: a systematic review of the literature with meta-analysis. Br J Cancer, 2005, 92:131–9., DOI 10. 1038/sj.bjc.6602258 Guan JL, ZhongWZ,An SJ, Yang JJ, Su J, Chen ZH, et al. KRASmutation in patients with lung cancer: a predictor for poor prognosis but not for EGFR-TKIs or chemotherapy. Ann Surg Oncol, 2013, 20:1381–8., DOI 10.1245/s10434-012- 2754-z Cao H, Ma Z, Li Y, Zhang Y, Chen H. Prognostic value of KRAS G12C mutation in lung adenocarcinoma stratified by stages and radiological features. J Thorac Cardiovasc Surg, 2023, 166:e479–e499., DOI 10.1016/j.jtcvs.2023.04.037 Finn SP, Addeo A, Dafni U, Thunnissen E, Bubendorf L, Madsen LB, et al. Prognostic impact of KRAS G12C mutation in patients with NSCLC: results from the European thoracic oncology platform lungscape project. J Thorac Oncol, 2021, 16:990–1002., DOI 10.1016/j.jtho.2021.02.016 Wahl SGF, Dai HY, Emdal EF, Ottestad AL, Dale VG, Richardsen E, et al. Prognostic value of absolute quantification of mutated KRAS in circulating tumour DNA in lung adenocarcinoma patients prior to therapy. J Pathol Clin Res, 2021, 7: 209–19., DOI 10.1002/cjp2.200 Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst RS, et al.Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol, 2005, 23: 5900–9., DOI 10.1200/JCO.2005.02.857 Tsao MS, Aviel-Ronen S, Ding K, Lau D, Liu N, Sakurada A, et al. Prognostic and predictive importance of p53 and RAS for adjuvant chemotherapy in non small-cell lung cancer. J Clin Oncol, 2007, 25:5240–7., DOI 10.1200/JCO.2007. 12.6953 Zalcman G, Beau-Faller M, Creveuil C, de Fraipont F, Mounawar M, Richard N, et al. Use of Ras effector RASSF1A promoter gene methylation and chromosome 9p loss of heterozygosity, LOH, to predict progression-free survival, PFS, in perioperative chemotherapy, CT, phase III trial IFCT- 0002 in resectable nonsmall cell lung cancer. J Clin Oncol, 2008, 26(15_Suppl. l):7500., DOI 10.1200/jco. 2008.26.15_suppl.7500 Hames ML, Chen H, Iams W, Aston J, Lovly CM, Horn L. Correlation between KRAS mutation status and response to chemotherapy in patients with advanced non-small cell lung cancer. Lung Cancer, 2016, 92:29–34., DOI 10.1016/j. lungcan.2015.11.004 Mao C, Qiu LX, Liao RY, Du FB, Ding H, Yang WC, et al. KRAS mutations and resistance to EGFR-TKIs treatment in patients with non-small cell lung cancer: a meta-analysis of 22 studies. Lung Cancer, 2010, 69:272–8., DOI 10.1016/j.lungcan. 2009.11.020 Zer A, Ding K, Lee SM, Goss GD, Seymour L, Ellis PM, et al. Pooled analysis of the prognostic and predictive value of KRAS mutation status and mutation subtype in patients with non-small cell lung cancer treated with epidermal growth factor receptor tyrosine kinase inhibitors. J Thorac Oncol, 2016, 11:312–23., DOI 10.1016/j. jtho.2015.11.010 Roberts PJ, Stinchcombe TE. KRAS mutation: should we test for it, and does it matter? J Clin Oncol, 2013, 31:1112–21., DOI 10.1200/JCO.2012.43.0454 Palmieri M, Zulato E, Wahl SGF, Guibert N, Frullanti E. Diagnostic accuracy of circulating free DNA testing for the detection of KRAS mutations in non-small cell lung cancer: a systematic review and meta-analysis. Front. Genet., 2022, 13: 1015161., DOI 10.3389/fgene.2022.1015161 Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer, 2003, 3:459–65., DOI 10.1038/nrc1097 Dankner M, Rose AAN, Rajkumar S, Siegel PM, Watson IR. Classifying BRAF alterations in cancer: new rational therapeutic strategies for actionable mutations. Oncogene, 2018, 37:3183–99., DOI 10.1038/s41388-018-0171-x Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature, 2002, 417:949–54., DOI 10.1038/ nature00766 Owsley J, Stein MK, Porter J, In GK, Salem M, O’Day S, et al. Prevalence of class I-III BRAF mutations among 114,662 cancer patients in a large genomic database. Exp Biol Med, Maywood,, 2021, 246:31–9., DOI 10.1177/ 1535370220959657 Namba H, Nakashima M, Hayashi T, Hayashida N, Maeda S, Rogounovitch TI, et al. Clinical implication of hot spot BRAF mutation, V599E, in papillary thyroid cancers. J Clin Endocrinol Metab, 2003, 88:4393–7., DOI 10.1210/jc.2003- 030305 Jones JC, Renfro LA, Al-Shamsi HO, Schrock AB, Rankin A, Zhang BY, et al. Non-V600 BRAF mutations define a clinically distinct molecular subtype of metastatic colorectal cancer. J Clin Oncol, 2017, 35:2624–30., DOI 10.1200/JCO. 2016.71.4394 Ambrosini-Spaltro A, Rengucci C, Capelli L, Chiadini E, Calistri D, Bennati C, et al. Clinicopathological features of non-small cell lung carcinoma with BRAF mutation. Curr Oncol, 2023, 30:10019–32., DOI 10.3390/curroncol30110728 Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell, 2004, 116:855–67., DOI 10.1016/s0092-8674(04, 00215-6 Dankort D, Filenova E, Collado M, Serrano M, Jones K, McMahon M. A new mouse model to explore the initiation, progression, and therapy of BRAFV600E-induced lung tumors. Genes Dev, 2007, 21:379–84., DOI 10.1101/ gad.1516407 Villaruz LC, Socinski MA, Abberbock S, Berry LD, Johnson BE, Kwiatkowski DJ, et al. Clinicopathologic features and outcomes of patients with lung adenocarcinomas harboring BRAF mutations in the Lung Cancer Mutation Consortium. Cancer, 2015, 121:448–56., DOI 10.1002/cncr.29042 Paik PK, Arcila ME, Fara M, Sima CS, Miller VA, Kris MG, et al. Clinical characteristics of patients with lung adenocarcinomas harboring BRAF mutations. J Clin Oncol, 2011, 29:2046–51., DOI 10.1200/JCO.2010.33.1280 Marchetti A, Felicioni L, Malatesta S, Grazia Sciarrotta M, Guetti L, Chella A, et al. Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations. J Clin Oncol, 2011, 29:3574–9., DOI 10.1200/JCO.2011. 35.9638 Cardarella S, Ogino A, Nishino M, Butaney M, Shen J, Lydon C, et al. Clinical, pathologic, and biologic features associated with BRAF mutations in nonsmall cell lung cancer. Clin Cancer Res, 2013, 19:4532–40., DOI 10.1158/1078-0432. CCR-13-0657 Capper D, Preusser M, Habel A, Sahm F, Ackermann U, Schindler G, et al. Assessment of BRAF V600E mutation status by immunohistochemistry with a mutation-specific monoclonal antibody. Acta Neuropathol, 2011, 122:11–9., DOI 10. 1007/s00401-011-0841-z Dvorak K, Higgins A, Palting J, Cohen M, Brunhoeber P. Immunohistochemistry with anti-BRAF V600E, VE1, mouse monoclonal antibody is a sensitive method for detection of the BRAF V600E mutation in colon cancer: evaluation of 120 cases with and without KRAS mutation and literature review. Pathol Oncol Res, 2019, 25:349–59., DOI 10.1007/s12253-017- 0344-x Ritterhouse LL, Barletta JA. BRAF V600E mutation-specific antibody: a review. Semin Diagn Pathol, 2015, 32:400–8., DOI 10.1053/j.semdp.2015.02.010 Rossle M, Sigg M, Ruschoff JH,Wild PJ, Moch H, Weber A, et al. Ultra-deep sequencing confirms immunohistochemistry as a highly sensitive and specific method for detecting BRAF V600E mutations in colorectal carcinoma. Virchows Arch, 2013, 463:623–31., DOI 10.1007/s00428-013-1492-3 Routhier CA, Mochel MC, Lynch K, Dias-Santagata D, Louis DN, Hoang MP. Comparison of 2 monoclonal antibodies for immunohistochemical detection of BRAF V600E mutation in malignant melanoma, pulmonary carcinoma, gastrointestinal carcinoma, thyroid carcinoma, and gliomas. Hum Pathol., 2013, 44:2563–70., DOI 10.1016/j.humpath.2013.06.018 Qiu T, Lu H, Guo L, Huang W, Ling Y, Shan L, et al. Detection of BRAF mutation in Chinese tumor patients using a highly sensitive antibody immunohistochemistry assay. Sci Rep, 2015, 5:9211., DOI 10.1038/srep09211 Hwang I, Choi YL, Lee H, Hwang S, Lee B, Yang H, et al. Selection strategies and practical application of BRAF V600e-mutated non-small cell lung carcinoma. Cancer Res Treat, 2022, 54:782–92., DOI 10.4143/crt.2021.843 Ettinger DS, Wood DE, Riely GJ, Aisner DL, Akerley W, Bauman JR, et al. NCCN guidelines insights: non-small cell lung cancer, version 2.2021. J Natl Compr Canc Netw, 2021, 19:254–66., DOI 10.6004/jnccn.2021.0013 Bartram CR, de Klein A, Hagemeijer A, van Agthoven T, Geurts van Kessel A, Bootsma D, et al. Translocation of c-ab1 oncogene correlates with the presence of a Philadelphia chromosome in chronic myelocytic leukaemia. Nature, 1983, 306: 277–80., DOI 10.1038/306277a0 Iwahara T, Fujimoto J, Wen D, Cupples R, Bucay N, Arakawa T, et al. Molecular characterization of ALK, a receptor tyrosine kinase expressed specifically in the nervous system. Oncogene, 1997, 14:439–49., DOI 10.1038/sj.onc.1200849 Stoica GE, Kuo A, Aigner A, Sunitha I, Souttou B, Malerczyk C, et al. Identification of anaplastic lymphoma kinase as a receptor for the growth factor pleiotrophin. J Biol Chem, 2001, 276:16772–9., DOI 10.1074/jbc.M010660200 De Munck S, Provost M, Kurikawa M, Omori I, Mukohyama J, Felix J, et al. Structural basis of cytokine-mediated activation of ALK family receptors. Nature, 2021, 600:143–7., DOI 10.1038/s41586-021-03959-5 Barreca A, Lasorsa E, Riera L, Machiorlatti R, Piva R, Ponzoni M, et al. Anaplastic lymphoma kinase in human cancer. JMol Endocrinol, 2011, 47:R11–23., DOI 10.1530/JME-11-0004 Caren H, Abel F, Kogner P, Martinsson T. High incidence of DNA mutations and gene amplifications of the ALK gene in advanced sporadic neuroblastoma tumours. Biochem J, 2008, 416:153–9., DOI 10.1042/bj20081834 Zito Marino F, Rocco G, Morabito A, Mignogna C, Intartaglia M, Liguori G, et al. A new look at the ALK gene in cancer: copy number gain and amplification. Expert Rev Anticancer Ther, 2016, 16:493–502., DOI 10.1586/14737140.2016. 1162098 Smolle E, Taucher V, Lindenmann J, Jost PJ, Pichler M. Current knowledge about mechanisms of drug resistance against ALK inhibitors in non-small cell lung cancer. Cancers, Basel,, 2021, 13:699., DOI 10.3390/cancers13040699 Elshatlawy M, Sampson J, Clarke K, Bayliss R. EML4-ALK biology and drug resistance in non-small cell lung cancer: a new phase of discoveries. Mol Oncol, 2023, 17:950–63., DOI 10.1002/1878-0261.13446 Morris SW, Kirstein MN, Valentine MB, Dittmer KG, Shapiro DN, Saltman DL, et al. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non- Hodgkin’s lymphoma. Science, 1994, 263:1281–4., DOI 10.1126/science.8122112 Griffin CA, Hawkins AL, Dvorak C, Henkle C, Ellingham T, Perlman EJ. Recurrent involvement of 2p23 in inflammatory myofibroblastic tumors. Cancer Res, 1999, 59:2776–80. PMID: 10383129. Lin E, Li L, Guan Y, Soriano R, Rivers CS, Mohan S, et al. Exon array profiling detects EML4-ALK fusion in breast, colorectal, and non-small cell lung cancers. Mol Cancer Res, 2009, 7:1466–76., DOI 10.1158/1541-7786.MCR-08-0522 Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature, 2007, 448:561–6., DOI 10.1038/nature05945 Inamura K, Takeuchi K, Togashi Y, Nomura K, Ninomiya H, Okui M, et al. EML4-ALK fusion is linked to histological characteristics in a subset of lung cancers. J Thorac Oncol, 2008, 3:13–7., DOI 10.1097/JTO.0b013e31815e8b60 Takahashi T, Sonobe M, Kobayashi M, Yoshizawa A, Menju T, Nakayama E, et al. Clinicopathologic features of non-small-cell lung cancer with EML4-ALK fusion gene. Ann Surg Oncol, 2010, 17:889–97., DOI 10.1245/s10434-009-0808-7 Mino-Kenudson M, Chirieac LR, Law K, Hornick JL, Lindeman N,Mark EJ, et al. A novel, highly sensitive antibody allows for the routine detection of ALKrearranged lung adenocarcinomas by standard immunohistochemistry. Clin Cancer Res, 2010, 16:1561–71., DOI 10.1158/1078-0432.CCR-09-2845 Paik JH, Choi CM, Kim H, Jang SJ, Choe G, Kim DK, et al. Clinicopathologic implication of ALK rearrangement in surgically resected lung cancer: a proposal of diagnostic algorithm for ALK-rearranged adenocarcinoma. Lung Cancer, 2012, 76: 403–9., DOI 10.1016/j.lungcan.2011.11.008 Conde E, Rojo F, Gomez J, Enguita AB, Abdulkader I, Gonzalez A, et al. Molecular diagnosis in non-small-cell lung cancer: expert opinion on ALK and ROS1 testing. J Clin Pathol, 2022, 75:145–53., DOI 10.1136/jclinpath-2021-207490 Zhou J, Yao H, Zhao J, Zhang S, You Q, Sun K, et al. Cell block samples from malignant pleural effusion might be valid alternative samples for anaplastic lymphoma kinase detection in patients with advanced non-small-cell lung cancer. Histopathology., 2015, 66:949–54., DOI 10.1111/his.12560 Matsushime H, Wang LH, Shibuya M. Human c-ros-1 gene homologous to the v-ros sequence of UR2 sarcoma virus encodes for a transmembrane receptorlike molecule. Mol Cell Biol, 1986, 6:3000–4., DOI 10.1128/mcb.6.8.3000 Balduzzi PC, Notter MF, Morgan HR, Shibuya M. Some biological properties of two new avian sarcoma viruses. J Virol, 1981, 40:268–75., DOI 10.1128/JVI.40.1. 268-275.1981 Acquaviva J, Wong R, Charest A. The multifaceted roles of the receptor tyrosine kinase ROS in development and cancer. Biochim Biophys Acta, 2009, 1795: 37–52., DOI 10.1016/j.bbcan.2008.07.006 Rimkunas VM, Crosby KE, Li D, Hu Y, Kelly ME, Gu TL, et al. Analysis of receptor tyrosine kinase ROS1-positive tumors in non-small cell lung cancer: identification of a FIG-ROS1 fusion. Clin Cancer Res, 2012, 18:4449–57., DOI 10. 1158/1078-0432.CCR-11-3351 Nguyen KT, Zong CS, Uttamsingh S, Sachdev P, BhanotM, Le MT, et al. The role of phosphatidylinositol 3-kinase, rho family GTPases, and STAT3 in Rosinduced cell transformation. J Biol Chem, 2002, 277:11107–15., DOI 10.1074/jbc. M108166200 Charest A, Lane K, McMahon K, Park J, Preisinger E, Conroy H, et al. Fusion of FIG to the receptor tyrosine kinase ROS in a glioblastoma with an interstitial del(6)(q21q21). Genes Chromosomes Cancer, 2003, 37:58–71., DOI 10.1002/gcc. 10207 Rikova K, Guo A, Zeng Q, Possemato A, Yu J, Haack H, et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell, 2007, 131:1190–203., DOI 10.1016/j.cell.2007.11.025 Zhang Q, Wu C, Ding W, Zhang Z, Qiu X, Mu D, et al. Prevalence of ROS1 fusion in Chinese patients with non-small cell lung cancer. Thorac Cancer, 2019, 10:47–53., DOI 10.1111/1759-7714.12899 Cui M, Han Y, Li P, Zhang J, Ou Q, Tong X, et al. Molecular and clinicopathological characteristics of ROS1-rearranged non-small-cell lung cancers identified by next-generation sequencing. Mol Oncol, 2020, 14:2787–95., DOI 10.1002/1878-0261.12789 Qiu Z, Ye B, Wang K, Zhou P, Zhao S, Li W, et al. Unique genetic characteristics and clinical prognosis of female patients with lung cancer harboring RET fusion gene. Sci Rep, 2020, 10:10387., DOI 10.1038/s41598-020- 66883-0 Bergethon K, Shaw AT, Ou SH, Katayama R, Lovly CM, McDonald NT, et al. ROS1 rearrangements define a unique molecular class of lung cancers. J Clin Oncol, 2012, 30:863–70., DOI 10.1200/JCO.2011.35.6345 Zhu Q, Zhan P, Zhang X, Lv T, Song Y. Clinicopathologic characteristics of patients with ROS1 fusion gene in non-small cell lung cancer: a metaanalysis. Transl Lung Cancer Res, 2015, 4:300–9., DOI 10.3978/j.issn.2218-6751. 2015.05.01 Zhu VW, Zhao JJ, Gao Y, Syn NL, Zhang SS, Ou SI, et al. Thromboembolism in ALK+ and ROS1+ NSCLC patients: a systematic review and meta-analysis. Lung Cancer, 2021, 157:147–55., DOI 10.1016/j.lungcan.2021.05.019 Yoshida A, Tsuta K, Wakai S, Arai Y, Asamura H, Shibata T, et al. Immunohistochemical detection of ROS1 is useful for identifying ROS1 rearrangements in lung cancers. Mod Pathol., 2014, 27:711–20., DOI 10. 1038/modpathol.2013.192 Selinger CI, Li BT, Pavlakis N, Links M, Gill AJ, Lee A, et al. Screening for ROS1 gene rearrangements in non-small-cell lung cancers using immunohistochemistry with FISH confirmation is an effective method to identify this rare target. Histopathology, 2017, 70:402–11., DOI 10.1111/his.13076 Su Y, Goncalves T, Dias-Santagata D, Hoang MP. Immunohistochemical detection of ROS1 fusion. Am J Clin Pathol, 2017, 147:77–82., DOI 10.1093/ajcp/ aqw201 Zhao J, Chen X, Zheng J, Kong M, Wang B, Ding W. A genomic and clinicopathological study of non-small-cell lung cancers with discordant ROS1 gene status by fluorescence in-situ hybridisation and immunohistochemical analysis. Histopathology., 2018, 73:19–28., DOI 10.1111/his.13492 Sharma S, Mishra SK, Bhardwaj M, Jha S, Geller M, Dewan A, et al. Correlation of ROS1, D4D6, immunohistochemistry with ROS1 fluorescence in situ hybridization assay in a contemporary cohort of pulmonary adenocarcinomas. South Asian J Cancer, 2022, 11:249–55., DOI 10.1055/s-0042-1750187 Hung YP, Sholl LM. Diagnostic and predictive immunohistochemistry for non-small cell lung carcinomas. Adv Anat Pathol, 2018, 25:374–86., DOI 10.1097/ PAP.0000000000000206 Bubendorf L, Buttner R, Al-Dayel F, Dietel M, Elmberger G, Kerr K, et al. Testing for ROS1 in non-small cell lung cancer: a review with recommendations. Virchows Arch, 2016, 469:489–503., DOI 10.1007/s00428-016-2000-3 Hofman V, Goffinet S, Bontoux C, Long-Mira E, Lassalle S, Ilie M, et al. A real-world experience from a single center, LPCE, nice, France, highlights the urgent need to abandon immunohistochemistry for ROS1 rearrangement screening of advanced non-squamous non-small cell lung cancer. J Pers Med, 2023, 13:810., DOI 10.3390/jpm13050810 Takahashi M, Ritz J, Cooper GM. Activation of a novel human transforming gene, ret, by DNA rearrangement. Cell, 1985, 42:581–8., DOI 10.1016/0092-8674(85, 90115-1 Ishizaka Y, Itoh F, Tahira T, Ikeda I, Sugimura T, Tucker J, et al. Human ret proto-oncogene mapped to chromosome 10q11.2. Oncogene, 1989, 4:1519–21. PMID: 2687772. Kjaer S, Ibanez CF. Identification of a surface for binding to the GDNF-GFR alpha 1 complex in the first cadherin-like domain of RET. J Biol Chem, 2003, 278: 47898–904., DOI 10.1074/jbc.M309772200 Regua AT, Najjar M, Lo HW. RET signaling pathway and RET inhibitors in human cancer. Front Oncol, 2022, 12:932353., DOI 10.3389/fonc.2022. 932353 Ibanez CF. Structure and physiology of the ret receptor tyrosine kinase. Cold Spring Harb Perspect Biol, 2013, 5:a009134–10., DOI 10.1101/cshperspect.a009134 Tomuschat C, Puri P. Ret gene is a major risk factor for Hirschsprung’s disease: a meta-analysis. Pediatr Surg Int, 2015, 31:701–10., DOI 10.1007/s00383- 015-3731-y Grieco M, Santoro M, Berlingieri MT, Melillo RM, Donghi R, Bongarzone I, et al. PTC is a novel rearranged form of the ret proto-oncogene and is frequently detected in vivo in human thyroid papillary carcinomas. Cell, 1990, 60:557–63., DOI 10.1016/0092-8674(90)90659-3 Donis-Keller H, Dou S, Chi D, Carlson KM, Toshima K, Lairmore TC, et al. Mutations in the RET proto-oncogene are associated with MEN 2A and FMTC. Hum Mol Genet, 1993, 2:851–6., DOI 10.1093/hmg/2.7.851 Mulligan LM, Kwok JB, Healey CS, Elsdon MJ, Eng C, Gardner E, et al. Germ-line mutations of the RET proto-oncogene in multiple endocrine neoplasia type 2A. Nature, 1993, 363:458–60., DOI 10.1038/363458a0 Ju YS, Lee WC, Shin JY, Lee S, Bleazard T, Won JK, et al. A transforming KIF5B and RET gene fusion in lung adenocarcinoma revealed from whole-genome and transcriptome sequencing. Genome Res, 2012, 22:436–45., DOI 10.1101/gr. 133645.111 Lipson D, Capelletti M, Yelensky R, Otto G, Parker A, Jarosz M, et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat Med, 2012, 18:382–4., DOI 10.1038/nm.2673 Kohno T, Ichikawa H, Totoki Y, Yasuda K, Hiramoto M, Nammo T, et al. KIF5B-RET fusions in lung adenocarcinoma. Nat. Med., 2012, 18:375–7., DOI 10. 1038/nm.2644 Takeuchi K, Soda M, Togashi Y, Suzuki R, Sakata S, Hatano S, et al. RET, ROS1 and ALK fusions in lung cancer. Nat. Med., 2012, 18:378–81., DOI 10.1038/ nm.2658 Kohno T, Tsuta K, Tsuchihara K, Nakaoku T, Yoh K, Goto K. RET fusion gene: translation to personalized lung cancer therapy. Cancer Sci, 2013, 104: 1396–400., DOI 10.1111/cas.12275 Kato S, Subbiah V, Marchlik E, Elkin SK, Carter JL, Kurzrock R. Ret aberrations in diverse cancers: next-generation sequencing of 4,871 patients. Clin Cancer Res, 2017, 23:1988–97., DOI 10.1158/1078-0432.CCR-16-1679 Piotrowska Z, Isozaki H, Lennerz JK, Gainor JF, Lennes IT, Zhu VW, et al. Landscape of acquired resistance to osimertinib in EGFR-mutant NSCLC and clinical validation of combined EGFR and RET inhibition with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov., 2018, 8:1529–39., DOI 10.1158/ 2159-8290.CD-18-1022 Wang R, Hu H, Pan Y, Li Y, Ye T, Li C, et al. RET fusions define a unique molecular and clinicopathologic subtype of non-small-cell lung cancer. J Clin Oncol, 2012, 30:4352–9., DOI 10.1200/JCO.2012.44.1477 Drilon A, Lin JJ, Filleron T, Ni A, Milia J, Bergagnini I, et al. Frequency of brain metastases and multikinase inhibitor outcomes in patients with RETrearranged lung cancers. J Thorac Oncol, 2018, 13:1595–601., DOI 10.1016/j.jtho. 2018.07.004 Drilon A, Wang L, Hasanovic A, Suehara Y, Lipson D, Stephens P, et al. Response to Cabozantinib in patients with RET fusion-positive lung adenocarcinomas. Cancer Discov, 2013, 3:630–5., DOI 10.1158/2159-8290.CD- 13-0035 Subbiah V, Velcheti V, Tuch BB, Ebata K, Busaidy NL, Cabanillas ME, et al. Selective RET kinase inhibition for patients with RET-altered cancers. Ann Oncol, 2018, 29:1869–76., DOI 10.1093/annonc/mdy137 Mulligan L. Selective RET kinase inhibitors and lung cancer. N Engl J Med, 2023, 389:1913–6., DOI 10.1056/NEJMe2311295 Zhou C, Solomon B, Loong HH, Park K, Perol M, Arriola E, et al. Firstline selpercatinib or chemotherapy and pembrolizumab in RET fusionpositive NSCLC. N Engl J Med, 2023, 389:1839–50., DOI 10.1056/ NEJMoa2309457 Go H, Jung YJ, Kang HW, Park IK, Kang CH, Lee JW, et al. Diagnostic method for the detection of KIF5B-RET transformation in lung adenocarcinoma. Lung Cancer, 2013, 82:44–50., DOI 10.1016/j.lungcan.2013.07.009 Feng J, Li Y, Wei B, Guo L, Li W, Xia Q, et al. Clinicopathologic characteristics and diagnostic methods of RET rearrangement in Chinese nonsmall cell lung cancer patients. Transl Lung Cancer Res, 2022, 11:617–31., DOI 10. 21037/tlcr-22-202 Yang SR, Aypar U, Rosen EY, Mata DA, Benayed R, Mullaney K, et al. A performance comparison of commonly used assays to detect RET fusions. Clin Cancer Res, 2021, 27:1316–28., DOI 10.1158/1078-0432.CCR-20-3208 Pu X, Xu C, Wang Q, Wang W, Wu F, Cai X, et al. Expert consensus on the diagnosis and treatment of RET gene fusion non-small cell lung cancer in China. Thorac Cancer, 2023, 14:3166–77., DOI 10.1111/1759-7714.15105 Huang EJ, Reichardt LF. Trk receptors: roles in neuronal signal transduction. Annu Rev Biochem, 2003, 72:609–42., DOI 10.1146/annurev.biochem.72.121801. 161629 Donovan MJ, Miranda RC, Kraemer R, McCaffrey TA, Tessarollo L, Mahadeo D, et al. Neurotrophin and neurotrophin receptors in vascular smooth muscle cells. Regulation of expression in response to injury. Am J Pathol, 1995, 147: 309–24. PMID: 7639328. Arevalo JC, Wu SH. Neurotrophin signaling: many exciting surprises. Cell Mol Life Sci, 2006, 63:1523–37., DOI 10.1007/s00018-006-6010-1 Martin-Zanca D, Hughes SH, Barbacid M. A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Nature, 1986, 319(6056):743–8., DOI 10.1038/319743a0 Knezevich SR, McFadden DE, TaoW, Lim JF, Sorensen PH. A novel ETV6- NTRK3 gene fusion in congenital fibrosarcoma. Nat Genet, 1998, 18:184–7., DOI 10. 1038/ng0298-184 Tognon C, Knezevich SR, Huntsman D, Roskelley CD, Melnyk N, Mathers JA, et al. Expression of the ETV6-NTRK3 gene fusion as a primary event in human secretory breast carcinoma. Cancer Cell, 2002, 2:367–76., DOI 10.1016/s1535- 6108(02)00180-0 Westphalen CB, Krebs MG, Le Tourneau C, Sokol ES, Maund SL, Wilson TR, et al. Genomic context of NTRK1/2/3 fusion-positive tumours from a large realworld population. NPJ Precis Oncol, 2021, 5:69., DOI 10.1038/s41698-021-00206-y Vaishnavi A, Capelletti M, Le AT, Kako S, Butaney M, Ercan D, et al. Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer. Nat Med, 2013, 19:1469–72., DOI 10.1038/nm.3352 Gatalica Z, Xiu J, Swensen J, Vranic S. Molecular characterization of cancers with NTRK gene fusions. Mod Pathol., 2019, 32:147–53., DOI 10.1038/s41379-018- 0118-3 Solomon JP, Linkov I, Rosado A, Mullaney K, Rosen EY, Frosina D, et al. NTRK fusion detection across multiple assays and 33,997 cases: diagnostic implications and pitfalls. Mod Pathol, 2020, 33:38–46., DOI 10.1038/s41379-019- 0324-7 Farago AF, Taylor MS, Doebele RC, Zhu VW, Kummar S, Spira AI, et al. Clinicopathologic features of non-small-cell lung cancer harboring an NTRK gene fusion. JCO Precis Oncol, 2018, 2018:1–12. PO.18.00037., DOI 10.1200/PO.18.00037 Harada G, Santini FC, Wilhelm C, Drilon A. NTRK fusions in lung cancer: from biology to therapy. Lung Cancer, 2021, 161:108–13., DOI 10.1016/j.lungcan. 2021.09.005 Delgado J, Pean E, Melchiorri D, Migali C, Josephson F, Enzmann H, et al. The European Medicines Agency review of entrectinib for the treatment of adult or paediatric patients with solid tumours who have a neurotrophic tyrosine receptor kinase gene fusions and adult patients with non-small-cell lung cancer harbouring ROS1 rearrangements. ESMO Open, 2021, 6:100087., DOI 10.1016/j.esmoop.2021. 100087 Cho BC, Chiu CH, Massarelli E, Buchschacher GL, Goto K, Overbeck TR, et al. Updated efficacy and safety of entrectinib in NTRK fusion-positive non-small cell lung cancer. Lung Cancer, 2023, 188:107442., DOI 10.1016/j.lungcan.2023. 107442 Liu F,Wei Y, Zhang H, Jiang J, Zhang P, Chu Q. NTRK fusion in non-small cell lung cancer: diagnosis, therapy, and TRK inhibitor resistance. Front Oncol, 2022, 12:864666., DOI 10.3389/fonc.2022.864666 Strohmeier S, Brcic I, Popper H, Liegl-Atzwanger B, Lindenmann J, Brcic L. Applicability of pan-TRK immunohistochemistry for identification of NTRK fusions in lung carcinoma. Sci Rep, 2021, 11:9785., DOI 10.1038/s41598-021- 89373-3 Vingiani A, Lorenzini D, Conca E, Volpi CC, Trupia DV, Gloghini A, et al. Pan-TRK immunohistochemistry as screening tool for NTRK fusions: a diagnostic workflow for the identification of positive patients in clinical practice. Cancer Biomark, 2023, 38:301–9., DOI 10.3233/CBM-220357 Hechtman JF, Benayed R, Hyman DM, Drilon A, Zehir A, Frosina D, et al. Pan-trk immunohistochemistry is an efficient and reliable screen for the detection of NTRK fusions. Am J Surg Pathol., 2017, 41:1547–51., DOI 10.1097/PAS. 0000000000000911 Overbeck TR, Reiffert A, Schmitz K, Rittmeyer A, Korber W, Hugo S, et al. NTRK gene fusions in non-small-cell lung cancer: real-world screening data of 1068 unselected patients. Cancers, Basel,, 2023, 15:2966., DOI 10.3390/ cancers15112966 Cooper CS, Park M, Blair DG, Tainsky MA, Huebner K, Croce CM, et al. Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature, 1984, 311:29–33., DOI 10.1038/311029a0 Dean M, Park M, Le Beau MM, Robins TS, Diaz MO, Rowley JD, et al. The human met oncogene is related to the tyrosine kinase oncogenes. Nature, 1985, 318:385–8., DOI 10.1038/318385a0 Naldini L, Weidner KM, Vigna E, Gaudino G, Bardelli A, Ponzetto C, et al. Scatter factor and hepatocyte growth factor are indistinguishable ligands for the MET receptor. EMBO J., 1991, 10:2867–78., DOI 10.1002/j.1460-2075.1991. tb07836.x Giordano S, Di Renzo MF, Narsimhan RP, Cooper CS, Rosa C, Comoglio PM. Biosynthesis of the protein encoded by the c-met proto-oncogene. Oncogene, 1989, 4:1383–8. PMID: 2554238. Gherardi E, Youles ME, Miguel RN, Blundell TL, Iamele L, Gough J, et al. Functional map and domain structure of MET, the product of the c-met protooncogene and receptor for hepatocyte growth factor/scatter factor. Proc Natl Acad Sci U S A, 2003, 100:12039–44., DOI 10.1073/pnas.2034936100 Lamorte L, Royal I, Naujokas M, Park M. Crk adapter proteins promote an epithelial-mesenchymal-like transition and are required for HGF-mediated cell spreading and breakdown of epithelial adherens junctions. Mol Biol Cell, 2002, 13: 1449–61., DOI 10.1091/mbc.01-10-0477 Organ SL, Tsao MS. An overview of the c-MET signaling pathway. Ther Adv Med Oncol, 2011, 3:S7–S19., DOI 10.1177/1758834011422556 Gherardi E, Birchmeier W, Birchmeier C, Vande Woude G. Targeting MET in cancer: rationale and progress. Nat Rev Cancer, 2012, 12:89–103., DOI 10.1038/ nrc3205 Petrelli A, Gilestro GF, Lanzardo S, Comoglio PM, Migone N, Giordano S. The endophilin-CIN85-Cbl complex mediates ligand-dependent downregulation of c-Met. Nature, 2002, 416:187–90., DOI 10.1038/416187a Joffre C, Barrow R, Menard L, Calleja V, Hart IR, Kermorgant S. A direct role for Met endocytosis in tumorigenesis. Nat Cell Biol, 2011, 13:827–37., DOI 10.1038/ ncb2257 Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science, 2007, 316:1039–43., DOI 10.1126/science.1141478 Bean J, Brennan C, Shih JY, Riely G, Viale A, Wang L, et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci U S A, 2007, 104:20932–7., DOI 10.1073/pnas.0710370104 Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P, et al. Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat Genet, 1997, 16:68–73., DOI 10.1038/ng0597-68 Gow CH, Liu YN, Li HY, Hsieh MS, Chang SH, Luo SC, et al. Oncogenic function of a KIF5B-met fusion variant in non-small cell lung cancer. Neoplasia, 2018, 20:838–47., DOI 10.1016/j.neo.2018.06.007 Kong-Beltran M, Seshagiri S, Zha J, Zhu W, Bhawe K, Mendoza N, et al. Somatic mutations lead to an oncogenic deletion of met in lung cancer. Cancer Res, 2006, 66:283–9., DOI 10.1158/0008-5472.CAN-05-2749 Lee JH, Gao CF, Lee CC, Kim MD, Vande Woude GF. An alternatively spliced form ofMet receptor is tumorigenic. ExpMol Med, 2006, 38:565–73., DOI 10. 1038/emm.2006.66 Frampton GM, Ali SM, Rosenzweig M, Chmielecki J, Lu X, Bauer TM, et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov, 2015, 5: 850–9., DOI 10.1158/2159-8290.CD-15-0285 Mazieres J, Vioix H, Pfeiffer BM, Campden RI, Chen Z, Heeg B, et al. MET exon 14 skipping in NSCLC: a systematic literature review of epidemiology, clinical characteristics, and outcomes. Clin Lung Cancer, 2023, 24:483–97., DOI 10.1016/j. cllc.2023.06.008 Markham A. Savolitinib: first approval. Drugs, 2021, 81:1665–70., DOI 10. 1007/s40265-021-01584-0 Cortot AB, Kherrouche Z, Descarpentries C,Wislez M, Baldacci S, Furlan A, et al. Exon 14 deleted MET receptor as a new biomarker and target in cancers. J Natl Cancer Inst, 2017, 109., DOI 10.1093/jnci/djw262 Ma PC, Tretiakova MS,MacKinnon AC, Ramnath N, Johnson C, Dietrich S, et al. Expression and mutational analysis of MET in human solid cancers. Genes Chromosomes Cancer, 2008, 47:1025–37., DOI 10.1002/gcc.20604 Salgia R. MET in lung cancer: biomarker selection based on scientific rationale. Mol Cancer Ther, 2017, 16:555–65., DOI 10.1158/1535-7163.MCT- 16-0472 Guo R, Berry LD, Aisner DL, Sheren J, Boyle T, Bunn PA, Jr, et al. MET IHC is a poor screen for MET amplification or MET exon 14 mutations in lung adenocarcinomas: data from a tri-institutional cohort of the lung cancer mutation consortium. J Thorac Oncol, 2019, 14:1666–71., DOI 10.1016/j.jtho. 2019.06.009 Davies KD, Lomboy A, Lawrence CA, Yourshaw M, Bocsi GT, Camidge DR, et al. DNA-based versus RNA-based detection of MET exon 14 skipping events in lung cancer. J Thorac Oncol, 2019, 14:737–41., DOI 10.1016/j.jtho.2018. 12.020 Poirot B, Doucet L, Benhenda S, Champ J, Meignin V, Lehmann-Che J. MET exon 14 alterations and new resistance mutations to tyrosine kinase inhibitors: risk of inadequate detection with current amplicon-based NGS panels. J Thorac Oncol, 2017, 12:1582–7., DOI 10.1016/j.jtho.2017.07.026 Heydt C, Ihle MA, Merkelbach-Bruse S. Overview of molecular detection technologies for MET in lung cancer. Cancers, Basel,, 2023, 15:2932., DOI 10.3390/ cancers15112932 Lee JK, Madison R, Classon A, Gjoerup O, Rosenzweig M, Frampton GM, et al. Characterization of non-small-cell lung cancers with MET exon 14 skipping alterations detected in tissue or liquid: clinicogenomics and real-world treatment patterns. JCO Precis Oncol, 2021, 5:1354–76. PO.21.00122., DOI 10.1200/PO.21. 00122 Bargmann CI, Hung MC, Weinberg RA. The neu oncogene encodes an epidermal growth factor receptor-related protein. Nature, 1986, 319:226–30., DOI 10.1038/319226a0 Gutierrez C, Schiff R. HER2: biology, detection, and clinical implications. Arch Pathol Lab Med, 2011, 135:55–62., DOI 10.1043/2010-0454-RAR.1 Goldenberg MM. Trastuzumab, a recombinant DNA-derived humanized monoclonal antibody, a novel agent for the treatment of metastatic breast cancer. Clin Ther, 1999, 21:309–18., DOI 10.1016/S0149-2918(00)88288-0 Hechtman JF, Polydorides AD. HER2/neu gene amplification and protein overexpression in gastric and gastroesophageal junction adenocarcinoma: a review of histopathology, diagnostic testing, and clinical implications. Arch Pathol Lab Med., 2012, 136:691–7., DOI 10.5858/arpa.2011-0168-RS Hirsch FR, Varella-Garcia M, Franklin WA, Veve R, Chen L, Helfrich B, et al. Evaluation of HER-2/neu gene amplification and protein expression in nonsmall cell lung carcinomas. Br J Cancer, 2002, 86:1449–56., DOI 10.1038/sj.bjc. 6600286 Stephens P, Hunter C, Bignell G, Edkins S, Davies H, Teague J, et al. Lung cancer: intragenic ERBB2 kinase mutations in tumours. Nature, 2004, 431:525–6., DOI 10.1038/431525b Shigematsu H, Takahashi T, Nomura M, Majmudar K, Suzuki M, Lee H, et al. Somatic mutations of the HER2 kinase domain in lung adenocarcinomas. Cancer Res, 2005, 65:1642–6., DOI 10.1158/0008-5472.CAN-04-4235 Wang SE, Narasanna A, Perez-Torres M, Xiang B, Wu FY, Yang S, et al. HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer Cell, 2006, 10:25–38., DOI 10.1016/j.ccr.2006.05.023 Sentana-Lledo D, Academia E, Viray H, Rangachari D, Kobayashi SS, VanderLaan PA, et al. EGFR exon 20 insertion mutations and ERBB2 mutations in lung cancer: a narrative review on approved targeted therapies from oral kinase inhibitors to antibody-drug conjugates. Transl Lung Cancer Res, 2023, 12:1590–610., DOI 10.21037/tlcr-23-98 Yang S, Wang Y, Zhao C, Li X, Liu Q, Mao S, et al. Exon 20 YVMA insertion is associated with high incidence of brain metastasis and inferior outcome of chemotherapy in advanced non-small cell lung cancer patients with HER2 kinase domain mutations. Transl Lung Cancer Res, 2021, 10:753–65., DOI 10.21037/tlcr- 20-559 Offin M, Feldman D, Ni A, Myers ML, Lai WV, Pentsova E, et al. Frequency and outcomes of brain metastases in patients with HER2-mutant lung cancers. Cancer, 2019, 125:4380–7., DOI 10.1002/cncr.32461 Pillai RN, Behera M, Berry LD, Rossi MR, Kris MG, Johnson BE, et al. HER2 mutations in lung adenocarcinomas: a report from the Lung Cancer Mutation Consortium. Cancer, 2017, 123:4099–105., DOI 10.1002/cncr.30869 Arcila ME, Chaft JE, Nafa K, Roy-Chowdhuri S, Lau C, Zaidinski M, et al. Prevalence, clinicopathologic associations, and molecular spectrum of ERBB2, HER2, tyrosine kinase mutations in lung adenocarcinomas. Clin Cancer Res, 2012, 18:4910–8., DOI 10.1158/1078-0432.CCR-12-0912 Ramalingam SS, Yang JC, Lee CK, Kurata T, Kim DW, John T, et al. Osimertinib as first-line treatment of EGFRmutation-positive advanced non-smallcell lung cancer. J Clin Oncol, 2018, 36:841–9., DOI 10.1200/JCO.2017.74.7576 Ren S, Wang J, Ying J, Mitsudomi T, Lee DH, Wang Z, et al. Consensus for HER2 alterations testing in non-small-cell lung cancer. ESMO Open, 2022, 7: 100395., DOI 10.1016/j.esmoop.2022.100395 Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science, 2015, 348:124–8., DOI 10.1126/science. aaa1348 Sun C, Mezzadra R, Schumacher TN. Regulation and function of the PD-L1 checkpoint. Immunity, 2018, 48:434–52., DOI 10.1016/j.immuni.2018.03.014 Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J., 1992, 11:3887–95., DOI 10.1002/j.1460-2075.1992.tb05481.x Zak KM, Kitel R, Przetocka S, Golik P, Guzik K, Musielak B, et al. Structure of the complex of human programmed death 1, PD-1, and its ligand PD-L1. Structure, 2015, 23:2341–8., DOI 10.1016/j.str.2015.09.010 Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupuslike autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motifcarrying immunoreceptor. Immunity, 1999, 11:141–51., DOI 10.1016/s1074- 7613(00)80089-8 Pan ZK, Ye F, Wu X, An HX, Wu JX. Clinicopathological and prognostic significance of programmed cell death ligand1, PD-L1, expression in patients with non-small cell lung cancer: a meta-analysis. J Thorac Dis, 2015, 7:462–70., DOI 10. 3978/j.issn.2072-1439.2015.02.13 Skov BG, Rorvig SB, Jensen THL, Skov T. The prevalence of programmed death ligand-1, PD-L1, expression in non-small cell lung cancer in an unselected, consecutive population. Mod Pathol, 2020, 33:109–17., DOI 10.1038/s41379-019- 0339-0 Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, et al. Phase I study of single-agent anti-programmed death-1, MDX-1106, in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol, 2010, 28:3167–75., DOI 10.1200/JCO.2009.26.7609 Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med, 2015, 372:2018–28., DOI 10.1056/NEJMoa1501824 Jorgensen JT. Companion and complementary diagnostics: clinical and regulatory perspectives. Trends Cancer, 2016, 2:706–12., DOI 10.1016/j.trecan.2016. 10.013 Dolled-Filhart M, Roach C, Toland G, Stanforth D, Jansson M, Lubiniecki GM, et al. Development of a companion diagnostic for pembrolizumab in nonsmall cell lung cancer using immunohistochemistry for programmed death ligand-1. Arch Pathol Lab Med, 2016, 140:1243–9., DOI 10.5858/arpa.2015- 0542-OA Kazandjian D, Suzman DL, Blumenthal G, Mushti S, He K, Libeg M, et al. FDA approval summary: nivolumab for the treatment of metastatic non-small cell lung cancer with progression on or after platinum-based chemotherapy. Oncologist, 2016, 21:634–42., DOI 10.1634/theoncologist.2015-0507 Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer, POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet, 2016, 387:1837–46., DOI 10.1016/S0140- 6736(16)00587-0 Sezer A, Kilickap S, Gumus M, Bondarenko I, Ozguroglu M, Gogishvili M, et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: a multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet, 2021, 397:592–604., DOI 10.1016/S0140- 6736(21)00228-2 Kim SY, Kim TE, Park CK, Yoon HK, Sa YJ, Kim HR, et al. Comprehensive comparison of 22C3 and SP263 PD-L1 expression in non-small-cell lung cancer using routine clinical and conditioned archives. Cancers, Basel,, 2022, 14:3138., DOI 10.3390/cancers14133138 Hendriks LE, Kerr KM, Menis J, Mok TS, Nestle U, Passaro A, et al. Nononcogene- addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol, 2023, 34:358–76., DOI 10.1016/j.annonc.2022.12.013 Rimm DL, Han G, Taube JM, Yi ES, Bridge JA, Flieder DB, et al. A prospective, multi-institutional, pathologist-based assessment of 4 immunohistochemistry assays for PD-L1 expression in non-small cell lung cancer. JAMA Oncol, 2017, 3:1051–8., DOI 10.1001/jamaoncol.2017.0013 Hirsch FR, McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, et al. PD-L1 immunohistochemistry assays for lung cancer: results from phase 1 of the Blueprint PD-L1 IHC assay comparison project. J Thorac Oncol, 2017, 12:208–22., DOI 10.1016/j.jtho.2016.11.2228 Tsao MS, Kerr KM, Kockx M, Beasley MB, Borczuk AC, Botling J, et al. PDL1 immunohistochemistry comparability study in real-life clinical samples: results of Blueprint phase 2 project. J Thorac Oncol, 2018, 13:1302–11., DOI 10.1016/j.jtho. 2018.05.013 LozanoMD, Abengozar-MuelaM, Echeveste JI, Subtil JC, Berto J, Gurpide A, et al. Programmed death-ligand 1 expression on direct Pap-stained cytology smears from non-small cell lung cancer: comparison with cell blocks and surgical resection specimens. Cancer Cytopathol, 2019, 127:470–80., DOI 10.1002/cncy. 22155 Tejerina E, Garca Tobar L, Echeveste JI, de Andrea CE, Vigliar E, Lozano MD. PD-L1 in cytological samples: a review and a practical approach. Front Med, Lausanne,, 2021, 8:668612., DOI 10.3389/fmed.2021.668612 Capizzi E, Ricci C, Giunchi F, Zagnoni S, Ceccarelli C, Gomez BUA, et al. Validation of the immunohistochemical expression of programmed death ligand 1, PD-L1, on cytological smears in advanced non small cell lung cancer. Lung Cancer, 2018, 126:9–14., DOI 10.1016/j.lungcan.2018.10.017 Heymann JJ, Bulman WA, Swinarski D, Pagan CA, Crapanzano JP, Haghighi M, et al. PD-L1 expression in non-small cell lung carcinoma: comparison among cytology, small biopsy, and surgical resection specimens. Cancer Cytopathol, 2017, 125:896–907., DOI 10.1002/cncy.21937 Jain D, Nambirajan A, Borczuk A, Chen G, Minami Y, Moreira AL, et al. Immunocytochemistry for predictive biomarker testing in lung cancer cytology. Cancer Cytopathol, 2019, 127:325–39., DOI 10.1002/cncy.22137 Zou Y, Xu L, Tang Q, You Q, Wang X, Ding W, et al. Cytology cell blocks from malignant pleural effusion are good candidates for PD-L1 detection in advanced NSCLC compared with matched histology samples. BMC Cancer, 2020, 20:344., DOI 10.1186/s12885-020-06851-z Layfield LJ, Zhang T, Esebua M. PD-L1 immunohistochemical testing: a review with reference to cytology specimens. Diagn Cytopathol, 2023, 51:51–8., DOI 10.1002/dc.25043 Pak MG, Roh MS. Cell-blocks are suitable material for programmed cell death ligand-1 immunohistochemistry: comparison of cell-blocks and matched surgical resection specimens in lung cancer. Cytopathology, 2019, 30:578–85., DOI 10.1111/cyt.12743 Huang Z, Chen L, Lv L, Fu CC, Jin Y, Zheng Q, et al. A new AI-assisted scoring system for PD-L1 expression in NSCLC. Comput Methods Programs Biomed, 2022, 221:106829., DOI 10.1016/j.cmpb.2022.106829 Cui Q, Li W, Wang D, Wang S, Yu J. Prognostic significance of blood-based PD-L1 analysis in patients with non-small cell lung cancer undergoing immune checkpoint inhibitor therapy: a systematic review and meta-analysis. World J Surg Oncol, 2023, 21:318., DOI 10.1186/s12957-023-03215-2 Negrao MV, Skoulidis F, Montesion M, Schulze K, Bara I, Shen V, et al. Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer. J Immunother Cancer, 2021, 9:e002891., DOI 10.1136/jitc-2021-002891 Lan B, Ma C, Zhang C, Chai S, Wang P, Ding L, et al. Association between PD-L1 expression and driver gene status in non-small-cell lung cancer: a meta-analysis. Oncotarget, 2018, 9:7684–99., DOI 10.18632/ oncotarget.23969 Liu J, Liu Y. Molecular diagnostic characteristics based on the next generation sequencing in lung cancer and its relationship with the expression of PD-L1. Pathol Res Pract, 2020, 216:152797., DOI 10.1016/j.prp.2019.152797 Otano I, Ucero AC, Zugazagoitia J, Paz-Ares L. At the crossroads of immunotherapy for oncogene-addicted subsets of NSCLC. Nat Rev Clin Oncol, 2023, 20:143–59., DOI 10.1038/s41571-022-00718-x Lee CK, Man J, Lord S, Links M, Gebski V, Mok T, et al. Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer-A meta-analysis. J Thorac Oncol, 2017, 12(2):403–7., DOI 10.1016/j.jtho.2016.10.007 Mazieres J, Drilon A, Lusque A, Mhanna L, Cortot AB, Mezquita L, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol, 2019, 30:1321–8., DOI 10.1093/annonc/mdz167 Gainor JF, Shaw AT, Sequist LV, Fu X, Azzoli CG, Piotrowska Z, et al. EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin Cancer Res, 2016, 22:4585–93., DOI 10.1158/1078-0432.CCR-15- 3101 Guisier F, Dubos-Arvis C, Vinas F, Doubre H, Ricordel C, Ropert S, et al. Efficacy and safety of anti-PD-1 immunotherapy in patients with advanced NSCLC with BRAF, HER2, or MET mutations or RET translocation: GFPC 01-2018. J Thorac Oncol, 2020, 15:628–36., DOI 10.1016/j.jtho.2019.12.129 Offin M, Guo R, Wu SL, Sabari J, Land JD, Ni A, et al. Immunophenotype and response to immunotherapy of RET-rearranged lung cancers. JCO Precis Oncol, 2019, 3:1–8. PO.18.00386., DOI 10.1200/PO.18.00386 Calles A, Liao X, Sholl LM, Rodig SJ, Freeman GJ, Butaney M, et al. Expression of PD-1 and its ligands, PD-L1 and PD-L2, in smokers and never smokers with KRAS-mutant lung cancer. J Thorac Oncol, 2015, 10:1726–35., DOI 10. 1097/JTO.0000000000000687 Jeanson A, Tomasini P, Souquet-Bressand M, Brandone N, Boucekine M, Grangeon M, et al. Efficacy of immune checkpoint inhibitors in KRAS-mutant nonsmall cell lung cancer, NSCLC). J Thorac Oncol, 2019, 14:1095–101., DOI 10.1016/j. jtho.2019.01.011 Skoulidis F, Byers LA, Diao L, Papadimitrakopoulou VA, Tong P, Izzo J, et al. Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities. Cancer Discov., 2015, 5:860–77., DOI 10.1158/2159-8290.CD- 14-1236 Ricciuti B, Arbour KC, Lin JJ, Vajdi A, Vokes N, Hong L, et al. Diminished efficacy of programmed death-(ligand)1 inhibition in STK11- and KEAP1-mutant lung adenocarcinoma is affected by KRAS mutation status. J Thorac Oncol, 2022, 17:399–410., DOI 10.1016/j.jtho.2021.10.013 Murciano-Goroff YR, Pak T,Mondaca S, Flynn JR, Montecalvo J, Rekhtman N, et al. Immune biomarkers and response to checkpoint inhibition of BRAFV600 and BRAF non-V600 altered lung cancers. Br J Cancer, 2022, 126: 889–98., DOI 10.1038/s41416-021-01679-1 Dudnik E, Peled N, Nechushtan H, Wollner M, Onn A, Agbarya A, et al. BRAF mutant lung cancer: programmed death ligand 1 expression, tumor mutational burden, microsatellite instability status, and response to immune check-point inhibitors. J Thorac Oncol, 2018, 13:1128–37., DOI 10.1016/j.jtho. 2018.04.024 Sabari JK, Leonardi GC, Shu CA, Umeton R,Montecalvo J, Ni A, et al. PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients withMET exon 14 altered lung cancers. Ann Oncol, 2018, 29:2085–91., DOI 10.1093/ annonc/mdy334 Travis WD, Rekhtman N, Riley GJ, Geisinger KR, Asamura H, Brambilla E, et al. Pathologic diagnosis of advanced lung cancer based on small biopsies and cytology: a paradigm shift. J Thorac Oncol, 2010, 5:411–4., DOI 10.1097/JTO. 0b013e3181d57f6e Kini SR Color atlas of pulmonary cytopathology. New York: Springer Verlag, 2002). p. 3–5. Bubendorf L, Lantuejoul S, de Langen AJ, Thunnissen E. Nonsmall cell lung carcinoma: diagnostic difficulties in small biopsies and cytological specimens: number 2 in the Series "Pathology for the clinician" Edited by Peter Dorfmuller and Alberto Cavazza. Eur Respir Rev, 2017, 26:170007., DOI 10.1183/16000617. 0007-2017 Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, et al. Updated molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors: guideline from the college of American pathologists, the international association for the study of lung cancer, and the association for molecular pathology. Arch Pathol Lab Med., 2018, 142: 321–46., DOI 10.5858/arpa.2017-0388-CP Hendriks LE, Kerr KM, Menis J, Mok TS, Nestle U, Passaro A, et al. Oncogene-addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol, 2023, 34:339–57., DOI 10.1016/j.annonc.2022.12.009 Ryska A, Buiga R, Fakirova A, Kern I, OlszewskiW, Plank L, et al. Non-small cell lung cancer in countries of central and southeastern Europe: diagnostic procedures and treatment reimbursement surveyed by the central European cooperative oncology group. Oncologist, 2018, 23:e152–8. Epub 2018 Aug 3. PMID: 30076278., DOI 10.1634/theoncologist.2018-0008 de Jager VD, Timens W, Bayle A, Botling J, Brcic L, Buttner R, et al. Developments in predictive biomarker testing and targeted therapy in advanced stage non-small cell lung cancer and their application across European countries. Lancet Reg Health Eur, 2024, 38:100838., DOI 10.1016/j. lanepe.2024.100838 Liam CK, Mallawathantri S, Fong KM. Is tissue still the issue in detecting molecular alterations in lung cancer? Respirology, 2020, 25:933–43., DOI 10.1111/ resp.13823 Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, et al. Recommendations for the use of next-generation sequencing, NGS, for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann Oncol, 2020, 31:1491–505., DOI 10.1016/j.annonc.2020. 07.014 Lemmon CA, Zhou J, Hobbs B, Pennell NA. Modeling costs and life-years gained by population-wide next-generation sequencing or single-gene testing in nonsquamous non-small-cell lung cancer in the United States. JCO Precis Oncol, 2023, 7:e2200294., DOI 10.1200/PO.22.00294 Matsuda H, Ogawa T, Sadatsuki Y, Tsujino T,Wada S, Kim SW, et al. Budget impact analysis of next-generation sequencing versus sequential single-gene testing in Japanese patients with advanced non-small-cell lung cancer. Respir Investig, 2023, 61:61–73., DOI 10.1016/j.resinv.2022.10.002 Kerr KM, Bibeau F, Thunnissen E, Botling J, Ryska A, Wolf J, et al. The evolving landscape of biomarker testing for non-small cell lung cancer in Europe. Lung Cancer, 2021, 154:161–75., DOI 10.1016/j.lungcan.2021. 02.026 Roberts ME, Neville E, Berrisford RG, Antunes G, Ali NJ, BTS Pleural Disease Guideline Group. Management of a malignant pleural effusion: British thoracic society pleural disease guideline 2010. Thorax, 2010, 65(Suppl. 2):ii32–40., DOI 10.1136/thx.2010.136994 Porcel JM. Malignant pleural effusions because of lung cancer. Curr Opin Pulm Med, 2016, 22:356–61., DOI 10.1097/MCP. 0000000000000264 Shojaee S, Roy-Chowdhuri S, Safi J, Grosu HB. Cytologic investigations for the diagnosis of malignant pleural effusion in non-small cell lung cancer: state-ofthe- art review for pulmonologists. J Bronchology Interv Pulmonol, 2021, 28:310–21., DOI 10.1097/LBR.0000000000000789 Veale N, Succony L, Rassl DM, Rintoul RC. Respiratory cytology in malignant lung disease - the thoracic oncologist’s perspective. Cytopathology, 2022, 33:39–43., DOI 10.1111/cyt.13021 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611733 EP 1611759 DI 10.3389/pore.2024.1611733 PG 27 ER PT J AU Li, H Zheng, L Zhong, G Yu, X Zhang, X Chen, L Chen, X AF Li, Houqiang Zheng, Lanqing Zhong, Guodong Yu, Xunbin Zhang, Xia Chen, Linying Chen, Xin TI Gastric epithelial neoplasm of fundic-gland mucosa lineage: representative of the low atypia differentiated gastric tumor and Ki67 may help in their identification SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gastric cancer; gastric adenocarcinoma of fundic-gland type; gastric adenocarcinoma of fundic-gland mucosa type; oxyntic gland adenoma; low-grade differentiated gastric tumors ID gastric cancer; gastric adenocarcinoma of fundic-gland type; gastric adenocarcinoma of fundic-gland mucosa type; oxyntic gland adenoma; low-grade differentiated gastric tumors AB Background: Gastric epithelial neoplasm of the fundic-gland mucosa lineages (GEN-FGMLs) are rare forms of gastric tumors that encompass oxyntic gland adenoma (OGA), gastric adenocarcinoma of the fundic-gland type (GA-FG), and gastric adenocarcinoma of the fundic-gland mucosa type (GA-FGM). There is no consensus on the cause, classification, and clinicopathological features of GEN-FGMLs, and misdiagnosis is common because of similarities in symptoms. Methods: 37 cases diagnosed with GEN-FGMLs were included in this study. H&E-stained slides were reviewed and clinicopathological parameters were recorded. Immunohistochemical staining was conducted for MUC2, MUC5AC, MUC6, CD10, CD56, synaptophysin, chromograninA, p53, Ki67, pepsinogen-I, H+/K+-ATPase and Desmin. Results: The patients’ ages ranged from 42 to 79 years, with a median age of 60. 17 were male and 20 were female. Morphologically, 19 OGAs, 16 GA-FGs, and two GA-FGMs were identified. Histopathological similarities exist between OGA, GA-FG, and GA-FGM. The tumors demonstrated well-formed glands, expanding with dense growth patterns comprising pale, blue-grey columnar cells with mild nuclear atypia. These cells resembled fundic gland cells. None of the OGA invaded the submucosal layer. The normal gastric pit epithelium covered the entire surface of the OGA and GA-FG, but the dysplasia pit epithelium covered the GA-FGM. Non-atrophic gastritis was observed in more than half of the background mucosa. All cases were diffusely positive for MUC6 and pepsinogen-I on immunohistochemistry. H+/K+-ATPase staining was negative or showed a scattered pattern in most cases. MUC5AC was expressed on the surface of GA-FGMs. p53 was focally expressed and the Ki67 index was low (1%–20%). Compared with OGA, GA-FG and GA-FGM were more prominent in the macroscopic view (p < 0.05) and had larger sizes (p < 0.0001). Additionally, GA-FG and GA-FGM exhibited higher Ki67 indices than OGA (p < 0.0001). Specimens with Ki-67 proliferation indices >2.5% and size >4.5mm are more likely to be diagnosed with GA-FG and GAFGM than OGA. Conclusion: GEN-FGMLs are group of well-differentiated gastric tumors with favourable biological behaviours, low cellular atypia, and low proliferation. Immunohistochemistry is critical for confirming diagnosis. Compared with OGA, GA-FG and GA-FGM have larger sizes and higher Ki67 proliferation indices, indicating that they play a critical role in the identification of GENFGML. Pathologists and endoscopists should be cautious to prevent misdiagnosis and overtreatment, especially in biopsy specimens. C1 [Li, Houqiang] Fujian Medical University, Shengli Clinical Medical CollegeFuzhou, Fujian, China. [Zheng, Lanqing] Fujian Medical University, Shengli Clinical Medical CollegeFuzhou, Fujian, China. [Zhong, Guodong] Fujian Traditional Chinese Medical University, The Second Affiliated HospitalFuzhou, Fujian, China. [Yu, Xunbin] Fujian Medical University, Shengli Clinical Medical CollegeFuzhou, Fujian, China. [Zhang, Xia] Fujian Medical University, Shengli Clinical Medical CollegeFuzhou, Fujian, China. [Chen, Linying] Fujian Medical University, The First Affiliated HospitalFuzhou, Fujian, China. [Chen, Xin] Fujian Medical University, Shengli Clinical Medical CollegeFuzhou, Fujian, China. RP Li, H (reprint author), Fujian Medical University, Shengli Clinical Medical College, Fuzhou, China. EM docli254@126.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Ueyama H, Yao T, Nakashima Y, Hirakawa K, Oshiro Y, Hirahashi M, et al. Gastric adenocarcinoma of fundic gland type, chief cell predominant type): proposal for a new entity of gastric adenocarcinoma. Am J Surg Pathol, 2010, 34:609–19., DOI 10.1097/PAS.0b013e3181d94d53 Japanese Gastric Cancer Association. Japanese gastric cancer treatment guidelines 2021, 6th edition). Gastric Cancer, 2023, 26(1):1–25., DOI 10.1007/ s10120-022-01331-8 Ueyama H, Yao T, Akazawa Y, Hayashi T, Kurahara K, Oshiro Y, et al. Gastric epithelial neoplasm of fundic-gland mucosa lineage: proposal for a new classification in association with gastric adenocarcinoma of fundic-gland type. J Gastroenterol, 2021, 56(9):814–28., DOI 10.1007/s00535-021-01813-z Tanabe H, Iwashita A, Ikeda K. Histopathological characteristics of gastric adenocarcinoma of fundic gland type. StomachIntest, 2015, 50:1521–31. Ushiku T, Kunita A, Kuroda R, Shinozaki-Ushiku A, Yamazawa S, Tsuji Y, et al. Oxyntic gland neoplasm of the stomach: expanding the spectrum and proposal of terminology. Mod Pathol, 2020, 33:206–16., DOI 10.1038/s41379-019-0338-1 Singhi AD, Lazenby AJ, Montgomery EA. Gastric adenocarcinoma with chief cell differentiation: a proposal for reclassification as oxyntic gland polyp/adenoma. Am J Surg Pathol, 2012, 36(7):1030–5., DOI 10.1097/PAS.0b013e31825033e7 Okumura Y, Takamatsu M, Ohashi M, Yamamoto Y, Yamamoto N, Kawachi H, et al. Gastric adenocarcinoma of fundic gland type with aggressive transformation and lymph node metastasis: a case report. J Gastric Cancer, 2018, 18:409–16., DOI 10.5230/jgc.2018.18.e22 IARC. WHO classifification of tumours editorial board: WHO classification of tumours, 5th edn, vol 1 digestive system tumours. Lyon: IARC, 2019). Kanesaka T, Uedo N, Yao K, Tanabe H, Yamasaki Y, Takeuchi Y, et al. New subtype of gastric adenocarcinoma: mixed fundic and pyloric mucosa-type adenocarcinoma. Clin J Gastroenterol, 2017, 10:224–8., DOI 10.1007/s12328-017- 0727-2 Kai K, Satake M, Tokunaga O. Gastric adenocarcinoma of fundic gland type with signet-ring cell carcinoma component: a case report and review of the literature. World J Gastroenterol, 2018, 24:2915–20., DOI 10.3748/wjg.v24.i26.2915 Li C, Wu X, Yang S, Yao J, Zheng H. Gastric adenocarcinoma of the fundic gland type: clinicopathological features of eight patients treated with endoscopic submucosal dissection. Diagn Pathol, 2020, 15(1):131., DOI 10.1186/s13000-020- 01047-2 Imamura K, Yao K, Nimura S, Tanabe H, Kanemitsu T, Miyaoka M, et al. Characteristic endoscopic findings of gastric adenocarcinoma of fundic-gland mucosa type. Gastric Cancer, 2021, 24(6):1307–19., DOI 10.1007/s10120-021- 01208-2 Sun WW, Zhang L, Gu MM, Zhang YQ, Qiu CM, Da Q. Gastric adenocarcinoma of the fundic gland type: clinicopathological analysis of six cases. Chin J Pathol, 2020, 49(4):343–7., DOI 10.3760/cma.j.cn112151-20190720- 00404 Shen FY, Zhuang HJ, Bai DY, Li NN, Zhang HF. Clinicopathological characteristics of 18 cases of oxyntic gland neoplasm from gastric fundic gland. Chin J Pathol, 2023, 52(7):718–20., DOI 10.3760/cma.j.cn112151-20221027-00891 Iwamuro M, Kusumoto C, Nakagawa M, Kobayashi S, Yoshioka M, Inaba T, et al. Endoscopic resection is a suitable initial treatment strategy for oxyntic gland adenoma or gastric adenocarcinoma of the fundic gland type. Sci Rep, 2021, 11(1): 7375., DOI 10.1038/s41598-021-86893-w Iwamuro M, Kusumoto C, Nakagawa M, Matsueda K, Kobayashi S, Yoshioka M, et al. Endoscopic features of oxyntic gland adenoma and gastric adenocarcinoma of the fundic gland type differ between patients with and without Helicobacter pylori infection: a retrospective observational study. BMC Gastroenterol, 2022, 22(1):294., DOI 10.1186/s12876-022-02368-w Zhou WX, Chen J. Consideration of diagnosing extremely well differentiated adenocarcinoma of gastrointestinal tract. Chin J Pathol, 2022, 51(8):693–5., DOI 10. 3760/cma.j.cn112151-20220517-00425 Hou WH, Duan XK, Hou WD, Liu YX, Wang JH, Wang XZ, et al. Clinicopathological features of very well-differentiated adenocarcinoma of the stomach. Chin J Pathol, 2022, 51(2):96–102., DOI 10.3760/cma.j.cn112151- 20210723-00525 Karam SM, Leblond CP. Identifying and counting epithelial cell types in the "corpus" of the mouse stomach. Anat Rec, 1992, 232(2):231–46., DOI 10.1002/ar. 1092320208 Lee SY, Saito T, Mitomi H, Hidaka Y, Murakami T, Nomura R, et al. Mutation spectrum in the Wnt/β-catenin signaling pathway in gastric fundic gland-associated neoplasms/polyps. Virchows Arch, 2015, 467(1):27–38., DOI 10.1007/s00428-015- 1753-4 Murakami T, Mitomi H, Yao T, Saito T, Shibuya T, Watanabe S. Epigenetic regulation of Wnt/β-catenin signal-associated genes in gastric neoplasia of the fundic gland, chief cell-predominant, type. Pathol Int, 2017, 67:147–55., DOI 10. 1111/pin.12509 Tajima Y, Murakami T, Saito T, Hiromoto T, Akazawa Y, Sasahara N, et al. Distinct involvement of the sonic Hedgehog signaling pathway in gastric adenocarcinoma of fundic gland type and conventional gastric adenocarcinoma. Digestion, 2017, 96:81–91., DOI 10.1159/000478999 Matsubara A, Sekine S, Kushima R, Ogawa R, Taniguchi H, Tsuda H, et al. Frequent GNAS and Kras mutations in pyloric gland adenoma of the stomach and duodenum. J Pathol, 2013, 229:579–87., DOI 10.1002/path.4153 Kushima R, Sekine S, Matsubara A, Taniguchi H, Ikegami M, Tsuda H. Gastric adenocarcinoma of the fundic gland type shares common genetic and phenotypic features with pyloric gland adenoma. Pathol Int, 2013, 63:318–25., DOI 10.1111/pin.12070 Nam KT, Lee HJ, Sousa JF, Weis VG, O’Neal RL, Finke PE, et al. Mature chief cells are cryptic progenitors for metaplasia in the stomach. Gastroenterology, 2010, 139(6):2028–37., DOI 10.1053/j.gastro.2010.09.005 Leushacke M, Tan SH, Wong A, Swathi Y, Hajamohideen A, Tan LT, et al. Lgr5-expressing chief cells drive epithelial regeneration and cancer in the oxyntic stomach. Nat Cell Biol, 2017, 19(7):774–86., DOI 10.1038/ncb3541 Kumagai K, Shimizu T, Nikaido M, Hirano T, Kakiuchi N, Takeuchi Y, et al. On the origin of gastric tumours: analysis of a case with intramucosal gastric carcinoma and oxyntic gland adenoma. J Pathol, 2023, 259(4):362–8., DOI 10.1002/ path.6050 Fahrenkamp AG, Wibbeke C, Winde G, Ofner D, Bocker W, Fischer-Colbrie R, et al. Immunohistochemical distribution of chromogranins A and B and secretogranin II in neuroendocrine tumours of the gastrointestinal tract. Virchows Arch, 1995, 426(4):361–7., DOI 10.1007/BF00191345 Sano W, Inoue F, Hirata D, Iwatate D, Hattori S, Fujita M, et al. Sporadic fundic gland polyps with dysplasia or carcinoma: clinical and endoscopic characteristics. World J Gastrointest Oncol, 2021, 13(7):662–72., DOI 10.4251/ wjgo.v13.i7.662 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611734 EP 1611743 DI 10.3389/pore.2024.1611734 PG 10 ER PT J AU Deme, D Tamaskovics, FB Jammoul, N Kovacs, S Kayode, OE Grice, J Telekes, A AF Deme, Daniel Tamaskovics, Ferenc Balint Jammoul, Nizar Kovacs, Sandor Kayode, Oladunjoye Emmanuel Grice, W. James Telekes, Andras TI Association between pathological characteristics and recurrence score by OncotypeDX in resected T1-3 and N0-1 breast cancer: a real-life experience of a North Hungarian regional center SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE breast cancer; pathological characteristics; OncotypeDX; recurrence score; observation oriented modeling ID breast cancer; pathological characteristics; OncotypeDX; recurrence score; observation oriented modeling AB Introduction: The 21-gene analysis (OncotypeDX) is validated test for pT1-3, pN0-1 with hormone receptor (HR) positive and normal expression of human epidermal growth factor receptor-2 (HER2) breast cancer (BC) to determine the aggressiveness of the disease based on the calculation of Recurrence Score (RS). Methods: In this retrospective study the authors correlated pathological characteristics and Recurrence Score (RS) by traditional statistical methods and Observed Oriented Modeling (OOM) in a realistic cohort of BC patients. Results: OncotypeDX tests were performed in 94 tumour specimens of 90 BC patients. >83% of node-negative (pN0) and >72% of node-positive (pN1) cases could avoid chemotherapy. For pN0 cases, non-parametric correlation and tests demonstrated significant association in eight types of characteristics [progesterone receptor (PR) expression, Ki-67 value, Ki-67 group, PR group, grade, estrogen receptor (ER) expression, Nottingham Prognostic Index (NPI) and Clinical Risk]. For pN1 cases, parametric correlation and tests showed significant association in six characteristic types (number of positive nodes, ER and PR expression, PR group, Ki-67 group and NPI). Based on OOM for pN0 cases, significant associations were established in three characteristics (Ki- 67 group, grade and NPI group). For pN1 cases OOM found significant associations in seven characteristics (PR group, PNI, LVI, Ki-67 group, grade, NPI group and number of positive nodes). Conclusion: First in oncology, OOM was applied, which found some other significant characteristics associated with RS than traditional statistical methods. There were few patients, where no clinical associations were found between characteristics and RS contrary to statistically significant differences. Therefore, the results of these statistical analyses can be neither applied for individual cases nor able to provide the bases for screening patients, i.e., whether they need for OncotypeDX testing or not. OncotypeDX still provides a personalised approach in BC. C1 [Deme, Daniel] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. [Tamaskovics, Ferenc Balint] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. [Jammoul, Nizar] Szent Lazar Megyei Korhaz, Patologiai OsztalySalgotarjan, Hungary. [Kovacs, Sandor] University of Debrecen, Department of Economical and Financial MathematicsDebrecen, Hungary. [Kayode, Oladunjoye Emmanuel] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. [Grice, W. James] Oklahoma State University, Department of PsychologyStillwater, OK, USA. [Telekes, Andras] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. RP Deme, D (reprint author), Szent Lazar Megyei Korhaz, Onkologiai Osztaly, Salgotarjan, Hungary. EM danieldeme_md@ymail.com CR WHO. International agency for research on cancer. Hungary: World Health Organization, 2023). Available from: https://gco.iarc.fr/today/data/factsheets/ populations/348-hungary-fact-sheets.pdf, Accessed June 01, 2023). Hungarian National Cancer Registry. Hungarian national cancer Registry, 2023). Available from: https://stat.nrr.hu/nrrstat/regi/0, Accessed June 01, 2023). Dyba T, Randi G, Bray F, Martos C, Giusti F, Nicholson N, et al. The European cancer burden in 2020: incidence and mortality estimates for 40 countries and 25 major cancers. Eur J Cancer, 2021, 157:308–47., DOI 10.1016/j.ejca.2021.07.039 ECIS. European cancer information system, ECIS,, 2020). Available from: https://stat.link/p4cg6v, Accessed June 01, 2023). Kenessey I, Szoke G, Dobozi M, Szatmari I, Weber A, Fogarassy G, et al. Comparison of cancer survival trends in Hungary in the periods 2001-2005 and 2011-2015 according to a population-based cancer Registry. Pathol Oncol Res, 2022, 28:1610668., DOI 10.3389/pore.2022.1610668 Fisher B, Dignam J, Bryant J, Wolmark N. Five versus more than five years of tamoxifen for lymph node-negative breast cancer: updated findings from the National Surgical Adjuvant Breast and Bowel Project B-14 randomized trial. J Natl Cancer Inst, 2001, 93:684–90., DOI 10.1093/jnci/93.9.684 Fisher B, Dignam J, Wolmark N, DeCillis A, Emir B, Wickerham DL, et al. Tamoxifen and chemotherapy for lymph node-negative, estrogen receptorpositive breast cancer. J Natl Cancer Inst, 1997, 89:1673–82., DOI 10.1093/jnci/ 89.22.1673 Sparano JA, Gray RJ, Makower DF, Pritchard KI, Albain KS, Hayes DF, et al. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med, 2018, 379:111–21., DOI 10.1056/NEJMoa1804710 Kalinsky K, Barlow WE, Gralow JR, Meric-Bernstam F, Albain KS, Hayes DF, et al. 21-Gene assay to inform chemotherapy benefit in node-positive breast cancer. N Engl J Med, 2021, 385:2336–47., DOI 10.1056/NEJMoa2108873 Breast Cancer. National comprehensive cancer Network, NCCN, clinical practice guidelines in oncology, version 1, 2024). Available from: https://www.nccn.org/ professionals/physician_gls/pdf/breast.pdf, Accessed January 31, 2024). National Comprehensive Cancer Network. Clinical practice guidelines in oncology, breast cancer version 1.2018, may 20), 3.2018, october 25), 3.2019, september 6), 5.2020, july 15), 8.2021, september 13), 4.2022, June 21, and 4.2023, march 23,, 2023). Available from: https://www.nccn.org/, Accessed July 19, 2018 for 1.2018; November 23, 2018 for 3.2018; November 11, 2019 for 3.2019; September 4, 2020 for 5.2020; October 7, 2021 for 8.2021; July 4, 2022 for 4.2022; June 12, 2023 for 4.2023). Grice JW. Observation oriented modeling: analysis of cause in the behavioral Sciences. Academic Press, Elsevier, 2011). Grice JW, Cota LD, Barrett PT, Wuensch KL, Potetat GM. A simple and transparent alternative to logistic regression. Adv Soc Sci Res J, 2016, 3:147–65., DOI 10.14738/assrj.37.2125 Elston CW, Ellis O. Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term followup. Histopathology, 1991, 19:403–10., DOI 10.1111/j.1365-2559.1991.tb00229.x Dowsett M, Nielsen TO, A’Hern R, Bartlett J, Coombes RC, Cuzick J, et al. Assessment of Ki67 in breast cancer: recommendations from the international Ki67 in breast cancer working group. J Natl Cancer Inst, 2011, 103:1656–64., DOI 10. 1093/jnci/djr393 Nielsen TO, Leung SCY, Rimm DL, Dodson A, Acs B, Badve S, et al. Assessment of Ki67 in breast cancer: updated recommendations from the international Ki67 in breast cancer working group. J Natl Cancer Inst, 2021, 113:808–19., DOI 10.1093/jnci/djaa201 Haybittle JL, Blamey RW, Elston CW, Johnson J, Doyle PJ, Campbell FC, et al. A prognostic index in primary breast cancer. Br J Cancer, 1982, 45:361–6., DOI 10. 1038/bjc.1982.62 Allison KH, Hammond ME, Dowsett M, McKernin SE, Carey LA, Fitzgibbons PL, et al. Estrogen and progesterone receptor testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists guideline update. Arch Pathol Lab Med, 2020, 144:545–63., DOI 10.5858/arpa.2019-0904-SA Dash SS, Sahu A, Toppo A. p53 expression in breast carcinoma and its association with tumor aggressiveness. J Datta Meghe Inst Med Sci Univ, 2021, 16: 266–72., DOI 10.4103/jdmimsu.jdmimsu_10_21 Galea MH, Blamey RW, Elston CE, Ellis IO. The Nottingham prognostic index in primary breast cancer. Breast Cancer Res Treat, 1992, 22:207–19., DOI 10. 1007/BF01840834 Blamey RW, Ellis IO, Pinder SE, Lee AH, Macmillan RD, Morgan DA, et al. Survival of invasive breast cancer according to the Nottingham Prognostic Index in cases diagnosed in 1990-1999. Eur J Cancer, 2007, 43:1548–55., DOI 10.1016/j.ejca. 2007.01.016 Cardoso F, van’t Veer LJ, Bogaerts J, Slaets L, Viale G, Delaloge S, et al. 70- Gene signature as an aid to treatment decisions in early-stage breast cancer. N Engl J Med, 2016, 375:717–29., DOI 10.1056/NEJMoa1602253 Grice JW. Drawing inferences from randomization tests. Personal Individual Differences, 2021, 179:110931., DOI 10.1016/j.paid.2021.110931 Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med, 2004, 351:2817–26., DOI 10.1056/NEJMoa041588 Paik S, Tang G, Shak S, Kim C, Baker J, KimW, et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptorpositive breast cancer. J Clin Oncol, 2006, 24:3726–34., DOI 10.1200/JCO.2005. 04.7985 Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst, 2009, 101:1446–52., DOI 10.1093/jnci/djp335 Albain KS, Barlow WE, Shak S, Hortobagyi GN, Livingston RB, Yeh IT, et al. Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptorpositive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol, 2010, 11:55–65., DOI 10.1016/S1470- 2045(09)70314-6 Nitz U, Gluz O, Christgen M, Kates RE, Clemens M, Malter W, et al. Reducing chemotherapy use in clinically high-risk, genomically low-risk pN0 and pN1 early breast cancer patients: five-year data from the prospective, randomised phase 3 West German Study Group, WSG, PlanB trial. Breast Cancer Res Treat, 2017, 165:573–83., DOI 10.1007/s10549-017- 4358-6 Valentine KD, Buchanan EM. JAM-boree: an application of observation oriented modeling to judgements of associative memory. J Cogn Psychol, 2013, 25: 400–22., DOI 10.1080/20445911.2013.775120 Abramson CI. Why the study of comparative psychology is important to neuroscientists. Front Behav Neurosci, 2023, 16:1095033., DOI 10.3389/fnbeh.2022.1095033 Patel R, Hovstadius M, Kier MW, Moshier E, Zimmerman BS, Cascetta KP, et al. Correlation of Ki67 working group prognostic risk categories with oncotype DX recurrence score, RS, in early breast cancer, EBC). Wiley Online Library, 2022). Available at: https://onlinelibrary.wiley.com/. downloaded from ascopubs.org on June 15, 2023. Diana A, Carlino F, Buono G, Antoniol G, Famiglietti V, De Angelis C, et al. Prognostic relevance of progesterone receptor levels in early luminal-like HER2 negative breast cancer subtypes: a retrospective analysis. Front Oncol, 2022, 12:813462., DOI 10.3389/fonc.2022.813462 Sibia US, Mylander C, Martin T, Rosman M, Sanders TJ, Lee Y, et al. OncotypeDX testing does not benefit patients with estrogen and progesterone receptor positive grade 1 breast cancers: a TAILORx validated study. Hematol Oncol Stem Cel Ther, 2023, 16:412–9., DOI 10.56875/2589-0646.1089 LambertiniM, Pinto AC, Ameye L, Jongen L, DelMastro L, Puglisi F, et al. The prognostic performance ofAdjuvant!Online andNottinghamPrognostic Index in young breast cancer patients. Br J Cancer, 2016, 115:1471–8., DOI 10.1038/bjc.2016.359 NICE Guidance. NICE Guidance [DG34]: tumour profiling tests to guide adjuvant chemotherapy decisions in early breast cancer, 2018). Available from: https://www.nice.org.uk/guidance/dg34, Accessed June 01, 2023). Hillyar C, Rizki H, Abbassi O, Miles-Dua S, Clayton G, Gandamihardja T, et al. Correlation between oncotype DX, PREDICT and the Nottingham prognostic index: implications for the management of early breast cancer. Cureus, 2020, 12: e7552., DOI 10.7759/cureus.7552 Kerin EP, Davey MG, McLaughlin RP, Sweeney KJ, Barry MK, Malone CM, et al. Comparison of the Nottingham Prognostic Index and OncotypeDX© recurrence score in predicting outcome in estrogen receptor positive breast cancer. Breast, 2022, 66:227–35., DOI 10.1016/j.breast.2022.11.001 Goldstein DA, Mayer C, Shochat T, Reinhorn D, Moore A, Sarfaty M, et al. The concordance of treatment decision guided by OncotypeDX and the PREDICT tool in real-world early-stage breast cancer. Cancer Med, 2020, 9:4603–12., DOI 10. 1002/cam4.3088 Predict breast cancer. What is Predict?, 2023). Available from: https://breast. predict.nhs.uk/predi%E2%80%8Bct_v2.1/tool, Accessed August 10, 2023). Hosoya K, Wakahara M, Ikeda K, Umekita Y. Perineural invasion predicts unfavorable prognosis in patients with invasive breast cancer. Cancer Diagn Progn, 2023, 3:208–14., DOI 10.21873/cdp.10203 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611735 EP 1611761 DI 10.3389/pore.2024.1611735 PG 27 ER PT J AU Mihalekne Fur, G Nemes, K Mago, Beno, A Topolcsanyi, P Moldvay, J Pongor, L AF Mihalekne Fur, Gabriella Nemes, Kolos Mago, Eva Beno, A. Alexandra Topolcsanyi, Petronella Moldvay, Judit Pongor, S. Lorinc TI Applied models and molecular characteristics of small cell lung cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE SCLC; drug response; ctDNA; liquid biopsy; databases ID SCLC; drug response; ctDNA; liquid biopsy; databases AB Small cell lung cancer (SCLC) is a highly aggressive type of cancer frequently diagnosed with metastatic spread, rendering it surgically unresectable for the majority of patients. Although initial responses to platinum-based therapies are often observed, SCLC invariably relapses within months, frequently developing drug-resistance ultimately contributing to short overall survival rates. Recently, SCLC research aimed to elucidate the dynamic changes in the genetic and epigenetic landscape. These have revealed distinct subtypes of SCLC, each characterized by unique molecular signatures. The recent understanding of the molecular heterogeneity of SCLC has opened up potential avenues for precision medicine, enabling the development of targeted therapeutic strategies. In this review, we delve into the applied models and computational approaches that have been instrumental in the identification of promising drug candidates. We also explore the emerging molecular diagnostic tools that hold the potential to transform clinical practice and patient care. C1 [Mihalekne Fur, Gabriella] Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Cancer Genomics and Epigenetics Core GroupSzeged, Hungary. [Nemes, Kolos] Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Cancer Genomics and Epigenetics Core GroupSzeged, Hungary. [Mago, Eva] Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Cancer Genomics and Epigenetics Core GroupSzeged, Hungary. [Beno, A. Alexandra] Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Cancer Genomics and Epigenetics Core GroupSzeged, Hungary. [Topolcsanyi, Petronella] Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Cancer Genomics and Epigenetics Core GroupSzeged, Hungary. [Moldvay, Judit] Szentgyorgyi A. University, Pulmonology ClinicSzeged, Hungary. [Pongor, S. Lorinc] Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Cancer Genomics and Epigenetics Core GroupSzeged, Hungary. RP Pongor, L (reprint author), Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Cancer Genomics and Epigenetics Core Group, Szeged, Hungary. EM lorinc.pongor@hcemm.eu CR Thomas A, Pattanayak P, Szabo E, Pinsky P. Characteristics and outcomes of small cell lung cancer detected by CT screening. Chest, 2018, 154(6):1284–90., DOI 10.1016/j.chest.2018.07.029 Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, et al. The 2015 world health organization classification of lung tumors: impact of genetic, clinical and radiologic advances since the 2004 classification. J Thorac Oncol, 2015, 10(9):1243–60., DOI 10.1097/JTO.0000000000000630 Carney DN, Gazdar AF, Bepler G, Guccion JG, Marangos PJ,Moody TW, et al. Establishment and identification of small cell lung cancer cell lines having classic and variant features. Cancer Res, 1985, 45(6):2913–23. Takahashi T, Nau MM, Chiba I, Birrer MJ, Rosenberg RK, Vinocour M, et al. p53: a frequent target for genetic abnormalities in lung cancer. Science, 1989, 246(4929):491–4., DOI 10.1126/science.2554494 Rudin CM, Durinck S, Stawiski EW, Poirier JT, Modrusan Z, Shames DS, et al. Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat Genet, 2012, 44(10):1111–6., DOI 10.1038/ng.2405 Peifer M, Fernandez-Cuesta L, Sos ML, George J, Seidel D, Kasper LH, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet, 2012, 44(10):1104–10., DOI 10.1038/ng.2396 George J, Lim JS, Jang SJ, Cun Y, Ozretic L, Kong G, et al. Comprehensive genomic profiles of small cell lung cancer. Nature, 2015, 524(7563):47–53., DOI 10. 1038/nature14664 Guinee DG, Fishback NF, Koss MN, Abbondanzo SL, Travis WD. The spectrum of immunohistochemical staining of small-cell lung carcinoma in specimens from transbronchial and open-lung biopsies. Am J Clin Pathol, 1994, 102(4):406–14., DOI 10.1093/ajcp/102.4.406 Gay CM, Stewart CA, Park EM, Diao L, Groves SM, Heeke S, et al. Patterns of transcription factor programs and immune pathway activation define four major subtypes of SCLC with distinct therapeutic vulnerabilities. Cancer Cell, 2021, 39(3): 346–60.e7., DOI 10.1016/j.ccell.2020.12.014 Rudin CM, Poirier JT, Byers LA, Dive C, Dowlati A, George J, et al. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer, 2019, 19(5):289–97., DOI 10.1038/s41568-019- 0133-9 Qu S, Fetsch P, Thomas A, Pommier Y, Schrump DS, Miettinen MM, et al. Molecular subtypes of primary SCLC tumors and their associations with neuroendocrine and therapeutic markers. J Thorac Oncol, 2022, 17(1):141–53., DOI 10.1016/j.jtho.2021.08.763 Zhang W, Girard L, Zhang YA, Haruki T, Papari-Zareei M, Stastny V, et al. Small cell lung cancer tumors and preclinical models display heterogeneity of neuroendocrine phenotypes. Transl Lung Cancer Res, 2018, 7(1):32–49., DOI 10. 21037/tlcr.2018.02.02 Borges M, Linnoila RI, van de Velde HJ, Chen H, Nelkin BD, Mabry M, et al. An achaete-scute homologue essential for neuroendocrine differentiation in the lung. Nature, 1997, 386(6627):852–5., DOI 10.1038/386852a0 Borromeo MD, Savage TK, Kollipara RK, He M, Augustyn A, Osborne JK, et al. ASCL1 and NEUROD1 reveal heterogeneity in pulmonary neuroendocrine tumors and regulate distinct genetic programs. Cell Rep, 2016, 16(5):1259–72., DOI 10.1016/j.celrep.2016.06.081 Huang YH, Klingbeil O, He XY, Wu XS, Arun G, Lu B, et al. POU2F3 is a master regulator of a tuft cell-like variant of small cell lung cancer. Genes Dev, 2018, 32(13–14):915–28., DOI 10.1101/gad.314815.118 Werneburg N, Gores GJ, Smoot RL. The Hippo pathway and YAP signaling: emerging concepts in regulation, signaling, and experimental targeting strategies with implications for hepatobiliary malignancies. Gene Expr, 2020, 20(1):67–74., DOI 10.3727/105221619X15617324583639 Tlemsani C, Pongor L, Elloumi F, Girard L, Huffman KE, Roper N, et al. SCLC-CellMiner: a resource for small cell lung cancer cell line genomics and pharmacology based on genomic signatures. Cel Rep, 2020, 33(3):108296., DOI 10. 1016/j.celrep.2020.108296 Wooten DJ, Groves SM, Tyson DR, Liu Q, Lim JS, Albert R, et al. Systemslevel network modeling of small cell lung cancer subtypes identifies master regulators and destabilizers. Plos Comput Biol, 2019, 15(10):e1007343., DOI 10. 1371/journal.pcbi.1007343 Lissa D, Takahashi N, Desai P, Manukyan I, Schultz CW, Rajapakse V, et al. Heterogeneity of neuroendocrine transcriptional states in metastatic small cell lung cancers and patient-derived models. Nat Commun, 2022, 13(1):2023., DOI 10.1038/ s41467-022-29517-9 Lim JS, Ibaseta A, Fischer MM, Cancilla B, O’Young G, Cristea S, et al. Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature, 2017, 545(7654):360–4., DOI 10.1038/nature22323 Fujino K, Motooka Y, Hassan WA, Ali Abdalla MO, Sato Y, Kudoh S, et al. Insulinoma-associated protein 1 is a crucial regulator of neuroendocrine differentiation in lung cancer. Am J Pathol, 2015, 185(12):3164–77., DOI 10.1016/ j.ajpath.2015.08.018 Pedersen N, Pedersen MW, Lan MS, Breslin MB, Poulsen HS. The insulinoma-associated 1: a novel promoter for targeted cancer gene therapy for small-cell lung cancer. Cancer Gene Ther, 2006, 13(4):375–84., DOI 10.1038/sj.cgt. 7700887 Christensen CL, Gjetting T, Poulsen TT, Cramer F, Roth JA, Poulsen HS. Targeted cytosine deaminase-uracil phosphoribosyl transferase suicide gene therapy induces small cell lung cancer-specific cytotoxicity and tumor growth delay. Clin Cancer Res, 2010, 16(8):2308–19., DOI 10.1158/1078-0432.CCR-09-3057 Takagi H, Zhao S, Muto S, Yokouchi H, Nishihara H, Harada T, et al. Deltalike 1 homolog, DLK1, as a possible therapeutic target and its application to radioimmunotherapy using 125I-labelled anti-DLK1 antibody in lung cancer models, HOT1801 and FIGHT004). Lung Cancer, 2021, 153:134–42., DOI 10. 1016/j.lungcan.2021.01.014 Hofsli E, Wheeler TE, LangaasM, Laegreid A, Thommesen L. Identification of novel neuroendocrine-specific tumour genes. Br J Cancer, 2008, 99(8):1330–9., DOI 10.1038/sj.bjc.6604565 Cardnell RJ, Li L, Sen T, Bara R, Tong P, Fujimoto J, et al. Protein expression of TTF1 and cMYC define distinct molecular subgroups of small cell lung cancer with unique vulnerabilities to aurora kinase inhibition, DLL3 targeting, and other targeted therapies. Oncotarget, 2017, 8(43):73419–32., DOI 10.18632/oncotarget. 20621 Shue YT, Drainas AP, Li NY, Pearsall SM, Morgan D, Sinnott-Armstrong N, et al. A conserved YAP/Notch/REST network controls the neuroendocrine cell fate in the lungs. Nat Commun, 2022, 13(1):2690., DOI 10.1038/s41467-022- 30416-2 Ito T, Kudoh S, Ichimura T, Fujino K, Hassan WAMA, Udaka N. Small cell lung cancer, an epithelial to mesenchymal transition, EMT)-like cancer: significance of inactive Notch signaling and expression of achaete-scute complex homologue 1. Hum Cel, 2017, 30(1):1–10., DOI 10.1007/s13577-016-0149-3 Gratton MO, Torban E, Jasmin SB, Theriault FM, German MS, Stifani S. Hes6 promotes cortical neurogenesis and inhibits Hes1 transcription repression activity by multiple mechanisms. Mol Cel Biol, 2003, 23(19):6922–35., DOI 10.1128/ mcb.23.19.6922-6935.2003 Owen DH, Giffin MJ, Bailis JM, Smit MAD, Carbone DP, He K. DLL3: an emerging target in small cell lung cancer. J Hematol Oncol, 2019, 12(1):61., DOI 10. 1186/s13045-019-0745-2 Totaro A, Castellan M, Di Biagio D, Piccolo S. Crosstalk between YAP/TAZ and Notch signaling. Trends Cel Biol, 2018, 28(7):560–73., DOI 10.1016/j.tcb.2018. 03.001 Sutherland KD, Ireland AS, Oliver TG. Killing SCLC: insights into how to target a shapeshifting tumor. Genes Dev, 2022, 36(5–6):241–58., DOI 10.1101/gad. 349359.122 Thiery JP, Acloque H, Huang RYJ, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell, 2009, 139(5):871–90., DOI 10.1016/j. cell.2009.11.007 Ireland AS, Micinski AM, Kastner DW, Guo B, Wait SJ, Spainhower KB, et al. MYC drives temporal evolution of small cell lung cancer subtypes by reprogramming neuroendocrine fate. Cancer Cell, 2020, 38(1):60–78., DOI 10. 1016/j.ccell.2020.05.001 Mollaoglu G, Guthrie MR, Bohm S, Bragelmann J, Can I, Ballieu PM, et al. MYC drives progression of small cell lung cancer to a variant neuroendocrine subtype with vulnerability to aurora kinase inhibition. Cancer Cell, 2017, 31(2): 270–85., DOI 10.1016/j.ccell.2016.12.005 Thomas A, Pommier Y. Small cell lung cancer: time to revisit DNA-damaging chemotherapy. Sci Transl Med, 2016, 8(346):346fs12., DOI 10.1126/scitranslmed. aaf6282 Macheret M, Halazonetis TD. Intragenic origins due to short G1 phases underlie oncogene-induced DNA replication stress. Nature, 2018, 555(7694): 112–6., DOI 10.1038/nature25507 Dominguez-Sola D, Gautier J. MYC and the control of DNA replication. Cold Spring Harb Perspect Med, 2014, 4(6):a014423., DOI 10.1101/cshperspect.a014423 Bian X, Lin W. Targeting DNA replication stress and DNA double-strand break repair for optimizing SCLC treatment. Cancers, Basel,, 2019, 11(9):1289., DOI 10.3390/cancers11091289 Takahashi N, Kim S, Schultz CW, Rajapakse VN, Zhang Y, Redon CE, et al. Replication stress defines distinct molecular subtypes across cancers. Cancer Res Commun, 2022, 2(6):503–17., DOI 10.1158/2767-9764.crc-22-0168 Horn L, Mansfield AS, Szczesna A, Havel L, Krzakowski M, Hochmair MJ, et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med, 2018, 379(23):2220–9., DOI 10.1056/NEJMoa1809064 Oboshi S, Tsugawa S, Seido T, Shimosato Y, Koide T. A new floating cell line derived from human pulmonary carcinoma of oat cell type. Gan, 1971, 62(6): 505–14. Carney DN, Gazdar AF, Minna JD. Positive correlation between histological tumor involvement and generation of tumor cell colonies in agarose in specimens taken directly from patients with small-cell carcinoma of the lung. Cancer Res, 1980, 40(6):1820–3. Pettengill OS, Sorenson GD, Wurster-Hill DH, Curphey TJ, Noll WW, Cate CC, et al. Isolation and growth characteristics of continuous cell lines from smallcell carcinoma of the lung. Cancer, 1980, 45(5):906–18., DOI 10.1002/1097- 0142(19800301)45:5<906::aid-cncr2820450513>3.0.co;2-h Drapkin BJ, Rudin CM. Advances in small-cell lung cancer, SCLC, translational research. Cold Spring Harb Perspect Med, 2021, 11(4):a038240., DOI 10.1101/cshperspect.a038240 Monks A, Scudiero D, Skehan P, Shoemaker R, Paull K, Vistica D, et al. Feasibility of a high-flux anticancer drug screen using a diverse panel of cultured human tumor cell lines. J Natl Cancer Inst, 1991, 83(11):757–66., DOI 10.1093/jnci/ 83.11.757 Wilding JL, Bodmer WF. Cancer cell lines for drug discovery and development. Cancer Res, 2014, 74(9):2377–84., DOI 10.1158/0008-5472.CAN- 13-2971 Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature, 2012, 483(7391):603–7., DOI 10.1038/nature11003 Yang W, Soares J, Greninger P, Edelman EJ, Lightfoot H, Forbes S, et al. Genomics of drug sensitivity in cancer, GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res, 2013, 41(Database issue):D955–61., DOI 10.1093/nar/gks1111 Polley E, Kunkel M, Evans D, Silvers T, Delosh R, Laudeman J, et al. Small cell lung cancer screen of oncology drugs, investigational agents, and gene and microRNA expression. J Natl Cancer Inst, 2016, 108(10):djw122., DOI 10.1093/jnci/ djw122 Sen C, Freund D, Gomperts BN. Three-dimensional models of the lung: past, present and future: a mini review. Biochem Soc Trans, 2022, 50(2):1045–56., DOI 10. 1042/BST20190569 Martinez-Pacheco S, O’Driscoll L. Pre-clinical in vitro models used in cancer research: results of a worldwide survey. Cancers, Basel,, 2021, 13(23):6033., DOI 10. 3390/cancers13236033 Sato T, Vries RG, Snippert HJ, van de WeteringM, Barker N, Stange DE, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature, 2009, 459(7244):262–5., DOI 10.1038/nature07935 Hou X, Du C, Lu L, Yuan S, Zhan M, You P, et al. Opportunities and challenges of patient-derived models in cancer research: patient-derived xenografts, patient-derived organoid and patient-derived cells. World J Surg Oncol, 2022, 20(1):37., DOI 10.1186/s12957-022-02510-8 Endo H, Okami J, Okuyama H, Kumagai T, Uchida J, Kondo J, et al. Spheroid culture of primary lung cancer cells with neuregulin 1/HER3 pathway activation. J Thorac Oncol, 2013, 8(2):131–9., DOI 10.1097/JTO.0b013e3182779ccf Yokota E, Iwai M, Yukawa T, Yoshida M, Naomoto Y, Haisa M, et al. Clinical application of a lung cancer organoid, tumoroid, culture system. NPJ Precis Oncol, 2021, 5(1):29., DOI 10.1038/s41698-021-00166-3 Li Y, Gao X, Ni C, Zhao B, Cheng X. The application of patient-derived organoid in the research of lung cancer. Cel Oncol, Dordr,, 2023, 46(3):503–19., DOI 10.1007/s13402-023-00771-3 Kunz-Schughart LA, Groebe K, Mueller-Klieser W. Three-dimensional cell culture induces novel proliferative and metabolic alterations associated with oncogenic transformation. Int J Cancer, 1996, 66(4):578–86., DOI 10.1002/(SICI, 1097-0215(19960516)66:4<578::AID-IJC25>3.0.CO;2-2 Phan N, Hong JJ, Tofig B, Mapua M, Elashoff D, Moatamed NA, et al. A simple high-throughput approach identifies actionable drug sensitivities in patientderived tumor organoids. Commun Biol, 2019, 2:78., DOI 10.1038/s42003-019- 0305-x Song Y, Wang Y, Guan A, Xue J, Li B, Huang Z, et al. Footprints: stamping hallmarks of lung cancer with patient-derived models, from molecular mechanisms to clinical translation. Front Bioeng Biotechnol, 2023, 11:1132940., DOI 10.3389/ fbioe.2023.1132940 Hutmacher DW. Biomaterials offer cancer research the third dimension. Nat Mater, 2010, 9(2):90–3., DOI 10.1038/nmat2619 Shi R, Radulovich N, Ng C, Liu N, Notsuda H, Cabanero M, et al. Organoid cultures as preclinical models of non-small cell lung cancer. Clin Cancer Res, 2020, 26(5):1162–74., DOI 10.1158/1078-0432.CCR-19-1376 Kim SY, Kim SM, Lim S, Lee JY, Choi SJ, Yang SD, et al. Modeling clinical responses to targeted therapies by patient-derived organoids of advanced lung adenocarcinoma. Clin Cancer Res, 2021, 27(15):4397–409., DOI 10.1158/1078-0432. CCR-20-5026 Kim M, Mun H, Sung CO, Cho EJ, Jeon HJ, Chun SM, et al. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun, 2019, 10(1):3991., DOI 10.1038/s41467-019-11867-6 Choi SY, Cho YH, Kim DS, Ji W, Choi CM, Lee JC, et al. Establishment and long-term expansion of small cell lung cancer patient-derived tumor organoids. Int J Mol Sci, 2021, 22(3):1349., DOI 10.3390/ijms22031349 Zhang Z, Shiratsuchi H, Lin J, Chen G, Reddy RM, Azizi E, et al. Expansion of CTCs from early stage lung cancer patients using a microfluidic co-culture model. Oncotarget, 2014, 5(23):12383–97., DOI 10.18632/oncotarget.2592 Zhu Z, Hu E, Shen H, Tan J, Zeng S. The functional and clinical roles of liquid biopsy in patient-derived models. J Hematol Oncol, 2023, 16(1):36., DOI 10.1186/ s13045-023-01433-5 Na F, Pan X, Chen J, Chen X, Wang M, Chi P, et al. KMT2C deficiency promotes small cell lung cancer metastasis through DNMT3A-mediated epigenetic reprogramming. Nat Cancer, 2022, 3(6):753–67., DOI 10.1038/s43018-022-00361-6 Rossi R, De Angelis ML, Xhelili E, Sette G, Eramo A, De Maria R, et al. Lung cancer organoids: the rough path to personalized medicine. Cancers, Basel)., 2022, 14(15):3703., DOI 10.3390/cancers14153703 Quaranta V, Linkous A. Organoids as a systems platform for SCLC brain metastasis. Front Oncol, 2022, 12:881989., DOI 10.3389/fonc.2022.881989 Hidalgo M, Amant F, Biankin AV, Budinska E, Byrne AT, Caldas C, et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov, 2014, 4(9):998–1013., DOI 10.1158/2159-8290.CD-14-0001 Shoemaker AR, Mitten MJ, Adickes J, Ackler S, Refici M, Ferguson D, et al. Activity of the Bcl-2 family inhibitor ABT-263 in a panel of small cell lung cancer xenograft models. Clin Cancer Res, 2008, 14(11):3268–77., DOI 10.1158/1078-0432. CCR-07-4622 Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli BA, et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature, 2005, 435(7042):677–81., DOI 10.1038/nature03579 Hann CL, Daniel VC, Sugar EA, Dobromilskaya I, Murphy SC, Cope L, et al. Therapeutic efficacy of ABT-737, a selective inhibitor of BCL-2, in small cell lung cancer. Cancer Res, 2008, 68(7):2321–8., DOI 10.1158/0008-5472. CAN-07-5031 Daniel VC, Marchionni L, Hierman JS, Rhodes JT, Devereux WL, Rudin CM, et al. A primary xenograft model of small-cell lung cancer reveals irreversible changes in gene expression imposed by culture in vitro. Cancer Res, 2009, 69(8): 3364–73., DOI 10.1158/0008-5472.CAN-08-4210 Drapkin BJ, George J, Christensen CL, Mino-Kenudson M, Dries R, Sundaresan T, et al. Genomic and functional fidelity of small cell lung cancer patient-derived xenografts. Cancer Discov, 2018, 8(5):600–15., DOI 10.1158/2159- 8290.CD-17-0935 Hodgkinson CL, Morrow CJ, Li Y, Metcalf RL, Rothwell DG, Trapani F, et al. Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer. Nat Med, 2014, 20(8):897–903., DOI 10.1038/nm.3600 Lallo A, Schenk MW, Frese KK, Blackhall F, Dive C. Circulating tumor cells and CDX models as a tool for preclinical drug development. Transl Lung Cancer Res, 2017, 6(4):397–408., DOI 10.21037/tlcr.2017.08.01 Guo S, Qian W, Cai J, Zhang L, Wery JP, Li QX. Molecular pathology of patient tumors, patient-derived xenografts, and cancer cell lines. Cancer Res, 2016, 76(16):4619–26., DOI 10.1158/0008-5472.CAN-15-3245 Jung J, Seol HS, Chang S. The generation and application of patient-derived xenograft model for cancer research. Cancer Res Treat, 2018, 50(1):1–10., DOI 10. 4143/crt.2017.307 Kim KB. The new way to define key oncogenic drivers of small cell lung cancer. Dev Reprod, 2023, 27(1):1–7., DOI 10.12717/DR.2023.27.1.1 Bragelmann J, Bohm S, Guthrie MR, Mollaoglu G, Oliver TG, Sos ML. Family matters: how MYC family oncogenes impact small cell lung cancer. Cell Cycle, 2017, 16(16):1489–98., DOI 10.1080/15384101.2017.1339849 Semenova EA, Nagel R, Berns A. Origins, genetic landscape, and emerging therapies of small cell lung cancer. Genes Dev, 2015, 29(14):1447–62., DOI 10.1101/ gad.263145.115 Schaffer BE, Park KS, Yiu G, Conklin JF, Lin C, Burkhart DL, et al. Loss of p130 accelerates tumor development in a mouse model for human small-cell lung carcinoma. Cancer Res, 2010, 70(10):3877–83., DOI 10.1158/0008-5472.CAN-09-4228 Sault C, Auger C, Roux B, Corvee N, Monier JC. Monoclonal antibodies against an epitope common to desmin and some keratins. Immunol Lett, 1987, 16(2):151–5., DOI 10.1016/0165-2478(87)90123-4 Nishida K, Arazoe T, Yachie N, Banno S, Kakimoto M, Tabata M, et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science, 2016, 353(6305):aaf8729., DOI 10.1126/science.aaf8729 Komor AC, Badran AH, Liu DR. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell, 2017, 169(3):559., DOI 10.1016/j.cell. 2017.04.005 Katti A, Vega-Perez A, Foronda M, Zimmerman J, Zafra MP, Granowsky E, et al. Generation of precision preclinical cancer models using regulated in vivo base editing. Nat Biotechnol, 2023, 42:437–47., DOI 10.1038/s41587-023-01900-x Ely ZA, Mathey-Andrews N, Naranjo S, Gould SI, Mercer KL, Newby GA, et al. A prime editormouse tomodel a broad spectrum of somaticmutations in vivo. Nat Biotechnol, 2023, 42:424–36., DOI 10.1038/s41587-023-01783-y Oser MG, MacPherson D, Oliver TG, Sage J, Park KS. Genetically-engineered mouse models of small cell lung cancer: the next generation. Oncogene, 2024, 43: 457–69., DOI 10.1038/s41388-023-02929-7 Leinonen R, Sugawara H, Shumway M, International Nucleotide Sequence Database Collaboration. The sequence read archive. Nucleic Acids Res, 2011, 39(Database issue):D19–21., DOI 10.1093/nar/gkq1019 Burgin J, Ahamed A, Cummins C, Devraj R, Gueye K, Gupta D, et al. The European nucleotide archive in 2022. Nucleic Acids Res, 2023, 51(D1):D121–5., DOI 10.1093/nar/gkac1051 Lappalainen I, Almeida-King J, Kumanduri V, Senf A, Spalding JD, Ur-Rehman S, et al. The European Genome-phenome Archive of human data consented for biomedical research. Nat Genet, 2015, 47(7):692–5., DOI 10.1038/ng.3312 Mailman MD, Feolo M, Jin Y, Kimura M, Tryka K, Bagoutdinov R, et al. The NCBI dbGaP database of genotypes and phenotypes. Nat Genet, 2007, 39(10): 1181–6., DOI 10.1038/ng1007-1181 Feng S, Zhang X, Gu X, Zhou M, Chen Y,Wang C. Identification of six novel prognostic gene signatures as potential biomarkers in small cell lung cancer. Comb Chem High Throughput Screen, 2023, 26(5):938–49., DOI 10.2174/ 1386207325666220427121619 Pongor LS, Schultz CW, Rinaldi L, Wangsa D, Redon CE, Takahashi N, et al. Extrachromosomal DNA amplification contributes to small cell lung cancer heterogeneity and is associated with worse outcomes. Cancer Discov, 2023, 13(4):928–49., DOI 10.1158/2159-8290.CD-22-0796 Pongor LS, Tlemsani C, Elloumi F, Arakawa Y, Jo U, Gross JM, et al. Integrative epigenomic analyses of small cell lung cancer cells demonstrates the clinical translational relevance of gene body methylation. iScience, 2022, 25(11): 105338., DOI 10.1016/j.isci.2022.105338 Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, et al. COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res, 2019, 47(D1): D941–7., DOI 10.1093/nar/gky1015 Jin X, Demere Z, Nair K, Ali A, Ferraro GB,Natoli T, et al. Ametastasis map of human cancer cell lines. Nature, 2020, 588(7837):331–6., DOI 10.1038/s41586-020- 2969-2 Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A, 2005, 102(43): 15545–50., DOI 10.1073/pnas.0506580102 Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The molecular signatures database, MSigDB, hallmark gene set collection. Cell Syst., 2015, 1(6):417–25., DOI 10.1016/j.cels.2015.12.004 Dressler L, Bortolomeazzi M, Keddar MR, Misetic H, Sartini G, Acha- Sagredo A, et al. Comparative assessment of genes driving cancer and somatic evolution in non-cancer tissues: an update of the network of cancer genes, NCG, resource. Genome Biol, 2022, 23(1):35., DOI 10.1186/s13059-022-02607-z Nemes K, Beno A, Topolcsanyi P, Mago E, Fur GM, Pongor LS. Predicting drug response of small cell lung cancer cell lines based on enrichment analysis of complex gene signatures. J Biotechnol, 2024, 383:86–93., DOI 10.1016/j.jbiotec.2024. 01.010 Sampson ER, McMurray HR, Hassane DC, Newman L, Salzman P, Jordan CT, et al. Gene signature critical to cancer phenotype as a paradigm for anticancer drug discovery. Oncogene, 2013, 32(33):3809–18., DOI 10.1038/onc.2012.389 Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau KW, et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature, 2012, 483(7391):570–5., DOI 10.1038/nature11005 Iorio F, Knijnenburg TA, Vis DJ, Bignell GR, Menden MP, Schubert M, et al. A landscape of pharmacogenomic interactions in cancer. Cell, 2016, 166(3):740–54., DOI 10.1016/j.cell.2016.06.017 Tsherniak A, Vazquez F,Montgomery PG, Weir BA, Kryukov G, Cowley GS, et al. Defining a cancer dependency map. Cell, 2017, 170(3):564–76., DOI 10.1016/j. cell.2017.06.010 McMillan EA, Ryu MJ, Diep CH, Mendiratta S, Clemenceau JR, Vaden RM, et al. Chemistry-first approach for nomination of personalized treatment in lung cancer. Cell, 2018, 173(4):864–78., DOI 10.1016/j.cell.2018.03.028 Rajapakse VN, Luna A, Yamade M, Loman L, Varma S, Sunshine M, et al. CellMinerCDB for integrative cross-database genomics and pharmacogenomics analyses of cancer cell lines. iScience, 2018, 10:247–64., DOI 10.1016/j.isci.2018. 11.029 Krushkal J, Silvers T, Reinhold WC, Sonkin D, Vural S, Connelly J, et al. Epigenome-wide DNA methylation analysis of small cell lung cancer cell lines suggests potential chemotherapy targets. Clin Epigenetics, 2020, 12(1):93., DOI 10. 1186/s13148-020-00876-8 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov, 2012, 2(5):401–4., DOI 10.1158/2159-8290.CD- 12-0095 Fruh M, De Ruysscher D, Popat S, Crino L, Peters S, Felip E, et al. Smallcell lung cancer, SCLC): ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol, 2013, 24(Suppl. 6):vi99–105., DOI 10. 1093/annonc/mdt178 Micke P, Faldum A, Metz T, Beeh KM, Bittinger F, Hengstler JG, et al. Staging small cell lung cancer: veterans administration lung study group versus international association for the study of lung cancer--what limits limited disease? Lung Cancer, 2002, 37(3):271–6., DOI 10.1016/s0169-5002(02)00072-7 Krpina K, Vranic S, Tomic K, Samarzija M, Baticic L. Small cell lung carcinoma: current diagnosis, biomarkers, and treatment options with future perspectives. Biomedicines, 2023, 11(7):1982., DOI 10.3390/biomedicines11071982 Megyesfalvi Z, Gay CM, Popper H, Pirker R, Ostoros G, Heeke S, et al. Clinical insights into small cell lung cancer: tumor heterogeneity, diagnosis, therapy, and future directions. CA Cancer J Clin, 2023, 73(6):620–52., DOI 10. 3322/caac.21785 Siravegna G, Mussolin B, Venesio T, Marsoni S, Seoane J, Dive C, et al. How liquid biopsies can change clinical practice in oncology. Ann Oncol, 2019, 30(10): 1580–90., DOI 10.1093/annonc/mdz227 Su Z, Wang Z, Ni X, Duan J, Gao Y, Zhuo M, et al. Inferring the evolution and progression of small-cell lung cancer by single-cell sequencing of circulating tumor cells. Clin Cancer Res, 2019, 25(16):5049–60., DOI 10.1158/1078-0432.CCR- 18-3571 Aggarwal C, Wang X, Ranganathan A, Torigian D, Troxel A, Evans T, et al. Circulating tumor cells as a predictive biomarker in patients with small cell lung cancer undergoing chemotherapy. Lung Cancer, 2017, 112:118–25., DOI 10.1016/j. lungcan.2017.08.008 Iams WT, Kopparapu PR, Yan Y, Muterspaugh A, Zhao Z, Chen H, et al. Blood-based surveillance monitoring of circulating tumor DNA from patients with SCLC detects disease relapse and predicts death in patients with limited-stage disease. JTO Clin Res Rep, 2020, 1(2):100024., DOI 10.1016/j.jtocrr.2020.100024 Devarakonda S, Sankararaman S, Herzog BH, Gold KA, Waqar SN, Ward JP, et al. Circulating tumor DNA profiling in small-cell lung cancer identifies potentially targetable alterations. Clin Cancer Res, 2019, 25(20):6119–26., DOI 10. 1158/1078-0432.CCR-19-0879 Mohan S, Foy V, Ayub M, Leong HS, Schofield P, Sahoo S, et al. Profiling of circulating free DNA using targeted and genome-wide sequencing in patients with SCLC. J Thorac Oncol, 2020, 15(2):216–30., DOI 10.1016/j.jtho.2019.10.007 Sivapalan L, IamsWT, Belcaid Z, Scott SC,NiknafsN, Balan A, et al.Dynamics of sequence and structural cell-free DNA landscapes in small-cell lung cancer. Clin Cancer Res, 2023, 29(12):2310–23., DOI 10.1158/1078-0432.CCR-22-2242 Fernandez-Garcia D, Hills A, Page K, Hastings RK, Toghill B, Goddard KS, et al. Plasma cell-free DNA, cfDNA, as a predictive and prognostic marker in patients with metastatic breast cancer. Breast Cancer Res, 2019, 21(1):149., DOI 10. 1186/s13058-019-1235-8 Gristina V, Barraco N, La Mantia M, Castellana L, Insalaco L, Bono M, et al. Clinical potential of circulating cell-free DNA, cfDNA, for longitudinally monitoring clinical outcomes in the first-line setting of non-small-cell lung cancer, NSCLC): a real-world prospective study. Cancers, Basel,, 2022, 14(23): 6013., DOI 10.3390/cancers14236013 Nong J, Gong Y, Guan Y, Yi X, Yi Y, Chang L, et al. Circulating tumor DNA analysis depicts subclonal architecture and genomic evolution of small cell lung cancer. Nat Commun, 2018, 9(1):3114., DOI 10.1038/s41467-018-05327-w Herbreteau G, Langlais A, Greillier L, Audigier-Valette C, Uwer L, Hureaux J, et al. Circulating tumor DNA as a prognostic determinant in small cell lung cancer patients receiving atezolizumab. J Clin Med, 2020, 9(12):3861., DOI 10.3390/ jcm9123861 Almodovar K, Iams WT, Meador CB, Zhao Z, York S, Horn L, et al. Longitudinal cell-free DNA analysis in patients with small cell lung cancer reveals dynamic insights into treatment efficacy and disease relapse. J Thorac Oncol, 2018, 13(1):112–23., DOI 10.1016/j.jtho.2017.09.1951 Fernandez-Cuesta L, Perdomo S, Avogbe PH, Leblay N, Delhomme TM, Gaborieau V, et al. Identification of circulating tumor DNA for the early detection of small-cell lung cancer. EBioMedicine, 2016, 10:117–23., DOI 10.1016/j.ebiom. 2016.06.032 Mondelo-Macia P, Garcia-Gonzalez J, Abalo A, Mosquera-Presedo M, Aguin S, Mateos M, et al. Plasma cell-free DNA and circulating tumor cells as prognostic biomarkers in small cell lung cancer patients. Transl Lung Cancer Res, 2022, 11(10):1995–2009., DOI 10.21037/tlcr-22-273 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611743 EP 1611755 DI 10.3389/pore.2024.1611743 PG 13 ER PT J AU Kaval, G Kartal, GDM Azamat, S Cingoz, E Ertas, G Karaman, S Kurtuldu, B Keskin, M Berker, N Karabulut, S Oral, NE Sakin, DN AF Kaval, Gizem Kartal, Gulbiz Dagoglu Merve Azamat, Sena Cingoz, Eda Ertas, Gokhan Karaman, Sule Kurtuldu, Basak Keskin, Metin Berker, Neslihan Karabulut, Senem Oral, Nezih Ethem Sakin, Dagoglu Nergiz TI Evaluating complete response prediction rates in locally advanced rectal cancer with different radiomics segmentation approaches SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE rectal cancer; watch and wait; radiomics; radiotherapy; pathological complete response ratio ID rectal cancer; watch and wait; radiomics; radiotherapy; pathological complete response ratio AB Purpose: Studies examining prediction of complete response (CR) in locally advanced rectum cancer (LARC) from pre/post chemoradiotherapy (CRT) magnetic resonance imaging (MRI) are performed mostly with segmentations of the tumor, whereas only in two studies segmentation included tumor and mesorectum. Additionally, pelvic extramesorectal region, which is included in the clinical target volume (CTV) of radiotherapy, may contain information. Therefore, we aimed to compare predictive rates of radiomics analysis with features extracted from segmentations of tumor, tumor+mesorectum, and CTV. Methods and materials: Ninety-three LARC patients who underwent CRT in our institution between 2012 and 2019 were retrospectively scanned. Patients were divided into CR and non-CR groups. Tumor, tumor+mesorectum and CTV were segmented on T2 preCRT MRI images. Extracted features were compared for best area under the curve (AUC) of CR prediction with 15 machinelearning models. Results: CR was observed in 25 patients (26.8%), of whom 13 had pathological, and 12 had clinical complete response. For tumor, tumor+mesorectum and CTV segmentations, the best AUC were 0.84, 0.81, 0.77 in the training set and 0.85, 0.83 and 0.72 in the test set, respectively; sensitivity and specificity for the test set were 76%, 90%, 76% and 71%, 67% and 62%, respectively. Conclusion: Although the highest AUC result is obtained from the tumor segmentation, the highest accuracy and sensitivity are detected with tumor+mesorectum segmentation and these findings align with previous studies, suggesting that the mesorectum contains valuable insights for CR. The lowest result is obtained with CTV segmentation. More studies with mesorectum and pelvic nodal regions included in segmentation are needed. C1 [Kaval, Gizem] Istanbul University, Istanbul Medicine Faculty, Institute of Oncology, Department of Radiation OncologyIstanbul, Turkey. [Kartal, Gulbiz Dagoglu Merve] Cerrahpasa Medical School, Istanbul University, Department of RadiologyIstanbul, Turkey. [Azamat, Sena] Cam and Sakura City Hospital, Department of RadiologyIstanbul, Turkey. [Cingoz, Eda] Bagcilar Training and Research Hospital, Department of RadiologyIstanbul, Turkey. [Ertas, Gokhan] Yeditepe University, Department of Biomedical EngineeringIstanbul, Turkey. [Karaman, Sule] Istanbul University, Istanbul Medicine Faculty, Institute of Oncology, Department of Radiation OncologyIstanbul, Turkey. [Kurtuldu, Basak] Hackalibaba Hospital, Department of EmergencyTrabzon, Turkey. [Keskin, Metin] Istanbul Faculty of Medicine, Istanbul University, Department of SurgeryIstanbul, Turkey. [Berker, Neslihan] Istanbul Medical Faculty, Department of PathologyIstanbul, Turkey. [Karabulut, Senem] Istanbul Medical Faculty, Istanbul University, Oncology InstituteIstanbul, Turkey. [Oral, Nezih Ethem] Istanbul University, Istanbul Medicine Faculty, Institute of Oncology, Department of Radiation OncologyIstanbul, Turkey. [Sakin, Dagoglu Nergiz] Istanbul University, Istanbul Medicine Faculty, Institute of Oncology, Department of Radiation OncologyIstanbul, Turkey. RP Kaval, G (reprint author), Istanbul University, Istanbul Medicine Faculty, Institute of Oncology, Department of Radiation Oncology, Istanbul, Turkey. EM gizemkaval1@istanbul.edu.tr CR GLOBOCAN. Estimated age-standardized incidence rates, World, in 2020, rectum. France: GLOBOCAN, 2020). Available from: https://gco.iarc.fr/, Accessed February 2023). Bahadoer RR, Dijkstra EA, van Etten B, Marijnen CAM, Putter H, Kranenbarg EM, et al. Short-course radiotherapy followed by chemotherapy before total mesorectal excision, TME, versus preoperative chemoradiotherapy, TME, and optional adjuvant chemotherapy in locally advanced rectal cancer, RAPIDO): a randomised, open-label, phase 3 trial. Lancet Oncol, 2021, 22:29–42., DOI 10.1016/ S1470-2045(20)30555-6 Cisel B, Pietrzak L, Michalski W, Wyrwicz L, Rutkowski A, Kosakowska E, et al. Long-course preoperative chemoradiation versus 5 × 5 Gy and consolidation chemotherapy for clinical T4 and fixed clinical T3 rectal cancer: long-term results of the randomized Polish II study. Ann Oncol, 2019, 30:1298–303., DOI 10.1093/annonc/mdz186 Jin J, Tang Y, Hu C, Jiang LM, Jiang J, Li N, et al. Multicenter, randomized, phase III trial of short-term radiotherapy plus chemotherapy versus long-term chemoradiotherapy in locally advanced rectal cancer, STELLAR). J Clin Oncol, 2022, 40:1681–92., DOI 10.1200/JCO.21.01667 Maas M, Nelemans PJ, Valentini V, Das P, Rodel C, Kuo L-J, et al. Long-term outcome in patients with a pathological complete response after chemoradiation for rectal cancer: a pooled analysis of individual patient data. Lancet Oncol, 2010, 11: 835–44., DOI 10.1016/S1470-2045(10)70172-8 Maas M, Lambregts DM, Nelemans PJ, Heijnen LA, Martens MH, Leijtens JW, et al. Assessment of clinical complete response after chemoradiation for rectal cancer with digital rectal examination, endoscopy, and MRI: selection for organsaving treatment. Ann Surg Oncol, 2015, 22:3873–80., DOI 10.1245/s10434-015- 4687-9 Lambregts DM, Beets GL, Maas M, Curvo-Semedo L, Kessels AG, Thywissen T, et al. Tumour ADC measurements in rectal cancer: effect of ROI methods on ADC values and interobserver variability. Eur Radiol, 2011, 21:2567–74., DOI 10.1007/ s00330-011-2220-5 Sassen S, de BooijM, Sosef M, Berendsen R, Lammering G, Clarijs R, et al. Locally advanced rectal cancer: is diffusion weighted MRI helpful for the identification of complete responders, ypT0N0, after neoadjuvant chemoradiation therapy? Eur Radiol, 2013, 23:3440–9., DOI 10.1007/s00330-013-2956-1 Dattani M, Marijnen C, Moran B, Tait D, Cunningham C, Rodriguez-Bigas M, et al. Session 4: shaping radiotherapy for rectal cancer: should this be personalized? Colorectal Dis, 2018, 20(Suppl. 1):92–6., DOI 10.1111/codi.14087 Hardiman KM, Ulintz PJ, Kuick RD, Hovelson DH, Gates CM, Bhasi A, et al. Intra-tumor genetic heterogeneity in rectal cancer. Lab Invest, 2016, 96:4–15., DOI 10.1038/labinvest.2015.131 Chu K, Bos SA, Gill CM, Torriani M, Bredella MA. Brown adipose tissue and cancer progression. Skeletal Radiol, 2020, 49:635–9., DOI 10.1007/s00256-019- 03322-w Conti G, Calderan L, Quintero Sierra LA, Conti A, Ossanna R, Boschi F, et al. Tumor and peritumoral adipose tissue crosstalk: de-differentiated adipocytes influence spread of colon carcinoma cells. Tissue Cell, 2023, 80:101990., DOI 10. 1016/j.tice.2022.101990 Gillies RJ, Kinahan PE, Hricak H. Radiomics: images are more than pictures, they are data. Radiology, 2016, 278:563–77., DOI 10.1148/radiol.2015151169 Miranda J, Horvat N, Araujo-Filho JAB, Albuquerque KS, Charbel C, Trindade BMC, et al. The role of radiomics in rectal cancer. J Gastrointest Cancer, 2023, 54:1158–80., DOI 10.1007/s12029-022-00909-w Jayaprakasam VS, Paroder V, Gibbs P, Bajwa R, Gangai N, Sosa RE, et al. MRI radiomics features of mesorectal fat can predict response to neoadjuvant chemoradiation therapy and tumor recurrence in patients with locally advanced rectal cancer. Eur Radiol, 2022, 32:971–80., DOI 10.1007/s00330-021-08144-w Shaish H, Aukerman A, Vanguri R, Spinelli A, Armenta P, Jambawalikar S, et al. Radiomics of MRI for pretreatment prediction of pathologic complete response, tumor regression grade, and neoadjuvant rectal score in patients with locally advanced rectal cancer undergoing neoadjuvant chemoradiation: an international multicenter study. Eur Radiol, 2020, 30:6263–73., DOI 10.1007/ s00330-020-06968-6 Myerson RJ, Garofalo MC, El Naqa I, Abrams RA, Apte A, Bosch WR, et al. Elective clinical target volumes for conformal therapy in anorectal cancer: a radiation therapy oncology group consensus panel contouring atlas. Int J Radiat Oncol Biol Phys, 2009, 74:824–30., DOI 10.1016/j.ijrobp.2008.08.070 Dworak Lk O, Hoffmann A. Pathological features of rectal cancer after preoperative radiochemotherapy. Int J Colorectal Dis, 1997, 12:19–23., DOI 10.1007/ s003840050072 Maas M, Beets-Tan RG, Lambregts DM, Lammering G, Nelemans PJ, Engelen SM, et al. Wait-and-see policy for clinical complete responders after chemoradiation for rectal cancer. J Clin Oncol, 2011, 29:4633–40., DOI 10.1200/ JCO.2011.37.7176 Saito T, Rehmsmeier M. The precision-recall plot is more informative than the ROC plot when evaluating binary classifiers on imbalanced datasets. PLoS One, 2015, 10:e0118432., DOI 10.1371/journal.pone.0118432 Kim SH, Chang HJ, Kim DY, Park JW, Baek JY, Kim SY, et al. What is the ideal tumor regression grading system in rectal cancer patients after preoperative chemoradiotherapy? Cancer Res Treat, 2016, 48:998–1009., DOI 10.4143/crt.2015.254 Zhang S, Tang B, Yu M, He L, Zheng P, Yan C, et al. Development and validation of a radiomics model based on lymph-node regression grading after neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Int J Radiat Oncol Biol Phys, 2023, 117:821–33., DOI 10.1016/j.ijrobp.2023.05.027 Otero de Pablos J, Mayol J. Controversies in the management of lateral pelvic lymph nodes in patients with advanced rectal cancer: east or west? Front Surg, 2019, 6:79., DOI 10.3389/fsurg.2019.00079 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611744 EP 1611752 DI 10.3389/pore.2024.1611744 PG 9 ER PT J AU Galffy, G Szabo, TG Tamasi, L Muller, V Moldvay, J Sarosi, V Kerpel-Fronius, A Kardos, T Csada, E Papai-Szekely, Zs Szasz, Z Kiraly, Zs Hodi, G Kovacs, Zs Balogh, Kovacs, AK Darida, M Buga, V Rokszin, Gy Abonyi-Toth, Zs Kiss, Z Voko, Z Bogos, K AF Galffy, Gabriella Szabo, Tamas Geza Tamasi, Lilla Muller, Veronika Moldvay, Judit Sarosi, Veronika Kerpel-Fronius, Anna Kardos, Tamas Csada, Edit Papai-Szekely, Zsolt Szasz, Zoltan Kiraly, Zsolt Hodi, Gabor Kovacs, Zsuzsanna Balogh, Eva Kovacs, Andrea Krisztina Darida, Miklos Buga, Viktoria Rokszin, Gyorgy Abonyi-Toth, Zsolt Kiss, Zoltan Voko, Zoltan Bogos, Krisztina TI Decreasing incidence and mortality of lung cancer in Hungary between 2011 and 2021 revealed by robust estimates reconciling multiple data sources SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lung cancer; incidence; mortality; COVID-19; Hungary ID lung cancer; incidence; mortality; COVID-19; Hungary AB Objective: Hungary has repeatedly been shown to have the highest cancerrelated mortality and incidence in Europe. Despite lung cancer being the most abundant malignant diagnosis in Hungary, numerous concerns have been raised recently regarding the bias inherent to reported incidence estimates. Re-analysis of reimbursement claims has been suggested previously by our group as an alternative approach, offering revised figures of lung cancer incidence between 2011 and 2016. Leveraging on this methodology, we aimed at updating Hungarian lung cancer incidence estimates with an additional 5 years (2017–2021), including years affected by the COVID-19 pandemic. Additionally, we also attempted to improve the robustness of estimates by taking additional characteristics of the patient pathway into account. Methods: Lung cancer patients between 2011 and 2021 were identified based on reimbursement-associated ICD-10 codes, histology codes and time patterns. Multiple query architectures were tested for sensitivity and compared to official estimates of the Hungarian National Cancer Registry (HNCR). Epidemiological trends were estimated by Poisson-regression, corrected for age and sex. Results: A total of 89,948 lung cancer patients diagnosed in Hungary between 2011 and 2021 have been identified by our study. In 2019 alone, 7,887 patientswere diagnosed according to our optimized query. ESP2013 standardized rate was estimated between 92.5/100,000 (2011) and 78.4/100,000 (2019). In 2019, standardized incidence was 106.8/100,000 for men and 59.7/100,000 for women. Up until the COVID-19 pandemic, lung cancer incidence was decreasing by 3.18% (2.1%–4.3%) yearly in men, while there was no significant decrease in women. Young age groups (40–49 and 50–59) featured the largest improvement, butwomen aged 60–79 are at an increasing risk for developing lung cancer. The COVID-19 pandemic resulted in a statistically significant decrease in lung cancer incidence, especially in the 50–59 age group (both sexes). Conclusion: Our results show that using an optimized approach, re-analysis of reimbursement claims yields robust estimates of lung cancer incidence. According to this approach, the incidence rate of male lung cancer is declining in Hungary, in concordance with the trend observed for lung cancer mortality. Among women aged 60–79, the incidence of lung cancer has risen, requiring more attention in the near future. C1 [Galffy, Gabriella] County Hospital of PulmonologyTorokbalint, Hungary. [Szabo, Tamas Geza] MSD Pharma Hungary Kft.Budapest, Hungary. [Tamasi, Lilla] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Muller, Veronika] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Moldvay, Judit] National Koranyi Institute for TB and Pulmonology, Department of PulmonologyBudapest, Hungary. [Sarosi, Veronika] University of Pecs, Faculty of MedicinePecs, Hungary. [Kerpel-Fronius, Anna] Orszagos Koranyi Tbc es Pulmonologiai Intezet, Radiologiai OsztalyBudapest, Hungary. [Kardos, Tamas] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Csada, Edit] Szentgyorgyi A. University, Pulmonology ClinicSzeged, Hungary. [Papai-Szekely, Zsolt] Fejer County Szent Gyorgy HospitalSzekesfehervar, Hungary. [Szasz, Zoltan] Petz Aladar Megyei Oktato Korhaz, PulmonologiaGyor, Hungary. [Kiraly, Zsolt] Veszprem Megyei Onkormanyzat TudogyogyintezeteFarkasgyepu, Hungary. [Hodi, Gabor] MSD Pharma Hungary Kft.Budapest, Hungary. [Kovacs, Zsuzsanna] MSD Pharma Hungary Kft.Budapest, Hungary. [Balogh, Eva] MSD Pharma Hungary Kft.Budapest, Hungary. [Kovacs, Andrea Krisztina] MSD Pharma Hungary Kft.Budapest, Hungary. [Darida, Miklos] MSD Pharma Hungary Kft.Budapest, Hungary. [Buga, Viktoria] MSD Pharma Hungary Kft.Budapest, Hungary. [Rokszin, Gyorgy] RxTarget LtdSzolnok, Hungary. [Abonyi-Toth, Zsolt] RxTarget LtdSzolnok, Hungary. [Kiss, Zoltan] MSD Pharma Hungary Kft.Budapest, Hungary. [Voko, Zoltan] Semmelweis University, Center for Health Technology AssessmentBudapest, Hungary. [Bogos, Krisztina] National Koranyi Institute of PulmonologyBudapest, Hungary. RP Kiss, Z (reprint author), MSD Pharma Hungary Kft., Budapest, Hungary. EM zoltan_kiss2@merck.com CR Dyba T, Randi G, Bray F, Martos C, Giusti F, Nicholson N, et al. The European cancer burden in 2020: incidence and mortality estimates for 40 countries and 25 major cancers. Eur J Cancer, 2021, 157:308–47., DOI 10.1016/j.ejca.2021.07.039 Ferlay J, Colombet M, Soerjomataram I, Dyba T, Randi G, Bettio M, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries and 25 major cancers in 2018. Eur J Cancer, 2018, 103:356–87., DOI 10.1016/j.ejca.2018.07.005 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer, 2013, 49(6):1374–403., DOI 10.1016/j.ejca.2012.12.027 Bogos K, Kiss Z, Galffy G, Tamasi L, Ostoros G, Muller V, et al. Revising incidence and mortality of lung cancer in central Europe: an epidemiology review from Hungary. Front Oncol, 2019, 9:1051., DOI 10.3389/fonc.2019.01051 Weber A, Szatmari I, DoboziM, Hilbert L, Branyiczkine Geczy G, Nagy P, et al. Comparison of Hungarian central statistical office’s causes of death data with the database of the Hungarian national cancer registry. Orv Hetil, 2022, 163(37): 1481–9., DOI 10.1556/650.2022.32573 Weber A, Mery L, Nagy P, Polgar C, Bray F, Kenessey I. Evaluation of data quality at the Hungarian national cancer registry, 2000-2019. Cancer Epidemiol, 2023, 82:102306., DOI 10.1016/j.canep.2022.102306 Kiss ZN, Bogos K, Tamasi L, Ostoros G, Muller V, Bittner N, et al. Underlying reasons for post-mortem diagnosed lung cancer cases - a robust retrospective comparative study from Hungary, HULC study). Front Oncol, 2022, 12:1032366., DOI 10.3389/fonc.2022.1032366 GalffyG, Vastag A, Bogos K, Kiss Z, Ostoros G,Muller V, et al. Significant regional differences in lung cancer incidence in Hungary: epidemiological study between 2011 and 2016. Pathol Oncol Res, 2021, 27:1609916., DOI 10.3389/pore.2021.1609916 Kiss Z, Galffy G, Muller V, Moldvay J, Sarosi V, Papai-Szekely Z, et al. Significant changes in advanced lung cancer survival during the past decade in Hungary: impact of modern immunotherapy and the COVID-19 pandemic. Front Oncol, 2023, 13:1207295., DOI 10.3389/fonc.2023.1207295 Zeileis A. Object-oriented computation of sandwich Estimators. J Stat Softw, 2006, 16:9., DOI 10.18637/jss.v016.i09 Zentrum fur Krebsregisterdaten. Krebs in Deutschland fur 2019/2020, 2023). Available from: https://www.krebsdaten.de/Krebs/DE/Content/Publikationen/Krebs_ in_Deutschland/kid_2023/kid_2023_c33_c34_lunge.pdf, Accessed January 12, 2024). MenyhartO, Fekete JT,Gyorffy B.Demographic shift disproportionately increases cancer burden in an aging nation: current and expected incidence and mortality in Hungary up to 2030. Clin Epidemiol, 2018, 10:1093–108., DOI 10.2147/CLEP.S155063 Li X,Hemminki K. Inherited predisposition to early onset lung cancer according to histological type. Int J Cancer, 2004, 112(3):451–7., DOI 10.1002/ijc.20436 Gallus S, Lugo A, Liu X, Behrakis P, Boffi R, Bosetti C, et al. Who smokes in Europe? Data from 12 European countries in the TackSHS survey, 2017-2018). J Epidemiol, 2021, 31(2):145–51., DOI 10.2188/jea.JE20190344 Raaschou-Nielsen O, Andersen ZJ, Beelen R, Samoli E, Stafoggia M,Weinmayr G, et al. Air pollution and lung cancer incidence in 17 European cohorts: prospective analyses from the European Study of Cohorts for Air Pollution Effects, ESCAPE). Lancet Oncol, 2013, 14(9):813–22., DOI 10.1016/S1470-2045(13)70279-1 Hill W, Lim EL, Weeden CE, Lee C, Augustine M, Chen K, et al. Lung adenocarcinoma promotion by air pollutants. Nature, 2023, 616(7955):159–67., DOI 10.1038/s41586-023-05874-3 European Environment Agency. European air quality index, 2023). Available from: https://airindex.eea.europa.eu/Map/AQI/Viewer/, Accessed January 12, 2024). Carel R, Olsson AC, Zaridze D, Szeszenia-Dabrowska N, Rudnai P, Lissowska J, et al. Occupational exposure to asbestos and man-made vitreous fibres and risk of lung cancer: a multicentre case-control study in Europe. Occup Environ Med, 2007, 64(8):502–8., DOI 10.1136/oem.2006.027748 Lissowska J, Foretova L, Dabek J, Zaridze D, Szeszenia-Dabrowska N, Rudnai P, et al. Family history and lung cancer risk: international multicentre case-control study in Eastern and Central Europe and meta-analyses. Cancer Causes Control, 2010, 21(7):1091–104., DOI 10.1007/s10552-010-9537-2 Guarga L, Ameijide A, Marcos-Gragera R, Carulla M, Delgadillo J, Borras JM, et al. Trends in lung cancer incidence by age, sex and histology from 2012 to 2025 in Catalonia, Spain). Sci Rep, 2021, 11:23274., DOI 10.1038/ s41598-021-02582-8 Gysling S, Morgan H, Ifesemen O, West D, Conibear J, Navani N, et al. The impact of COVID-19 on lung cancer incidence in england: analysis of the national lung cancer audit 2019 and 2020 rapid cancer registration datasets. Chest, 2023, 163:1599–607., DOI 10.1016/j.chest.2023.01.008 Elek P, Csanadi M, Fadgyas-Freyler P, Gervai N, Oross-Becsi R, Szecsenyi-Nagy B, et al. Heterogeneous impact of the COVID-19 pandemic on lung, colorectal and breast cancer incidence in Hungary: results from time series and panel data models. BMJ Open, 2022, 12(8):e061941., DOI 10.1136/ bmjopen-2022-061941 Kiss Z, Kocsis J, Nikolenyi A, Horvath Z, Knollmajer K, Benedek A, et al. Opposite trends in incidence of breast cancer in young and old female cohorts in Hungary and the impact of the Covid-19 pandemic: a nationwide study between 2011-2020. Front Oncol, 2023, 13:1182170., DOI 10.3389/fonc.2023. 1182170 Laczo A, Bodis J, Bogner P, Molnar K, Vajda R, Ponusz-Kovacs D, et al. Participation indicators of organizedmammography screening in Hungary between 2012-2021. Magy Onkol, 2022, 66(3):195–200. Maringe C, Spicer J, Morris M, Purushotham A, Nolte E, Sullivan R, et al. The impact of the COVID-19 pandemic on cancer deaths due to delays in diagnosis in England, UK: a national, population-based, modelling study. Lancet Oncol, 2020, 21(8):1023–34., DOI 10.1016/S1470-2045(20)30388-0 Luo Q, O’Connell DL, Yu XQ, Kahn C, Caruana M, Pesola F, et al. Cancer incidence and mortality in Australia from 2020 to 2044 and an exploratory analysis of the potential effect of treatment delays during the COVID-19 pandemic: a statistical modelling study. Lancet Public Health, 2022, 7(6):e537–e548., DOI 10. 1016/S2468-2667(22)00090-1 Valluri S, Lakshmi HN, Sunkavalli C. Incidental findings in CT scans on screening for COVID-19. Indian J Surg Oncol, 2023, 14(2):318–23., DOI 10.1007/ s13193-022-01669-6 Kenessey I, Nagy P, Polgar C. The Hungarian situation of cancer epidemiology in the second decade of the 21st century. Magy Onkol, 2022, 66(3):175–84. Global Burden of Disease Cancer Collaboration. Fitzmaurice C, Akinyemiju TF, Al Lami FH, Alam T, Alizadeh-Navaei R, et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 29 cancer groups, 1990 to 2016: a systematic analysis for the global burden of disease study. JAMA Oncol, 2018, 4(11): 1553–68., DOI 10.1001/jamaoncol.2018.2706 Global Burden of Disease 2019 Cancer Collaboration. Kocarnik JM, Compton K, Dean FE, FuW, Gaw BL, et al. Cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life years for 29 cancer groups from 2010 to 2019: a systematic analysis for the global burden of disease study 2019. JAMA Oncol, 2022, 8(3):420–44., DOI 10.1001/jamaoncol.2021.6987 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611754 EP 1611764 DI 10.3389/pore.2024.1611754 PG 11 ER PT J AU Antal, G Zsigmond, A Till, Orsi, E Szanto, I Buki, G Kereskai, L Herbert, Zs Hadzsiev, K Bene, J AF Antal, Greta Zsigmond, Anna Till, Agnes Orsi, Eniko Szanto, Ildiko Buki, Gergely Kereskai, Laszlo Herbert, Zsuzsanna Hadzsiev, Kinga Bene, Judit TI Case report: Initial atypical skeletal symptoms and dental anomalies as first signs of Gardner syndrome: the importance of genetic analysis in the early diagnosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Gardner syndrome; APC gene; osteoma; dental abnormalities; WES ID Gardner syndrome; APC gene; osteoma; dental abnormalities; WES AB Background: Gardner syndrome is a rare genetic cancer predisposition disorder characterized by intestinal polyposis, multiple osteomas, and soft and hard tissue tumors. Dental anomalies are present in approximately 30%– 70% of patients with Gardner syndrome and can be discovered during routine dental examinations. However, sometimes the diagnosis is challenging due to the high clinical variability and incomplete clinical picture. Herein, we report a family with various dental and bone anomalies, in which the definitive diagnosis was established with the help of a comprehensive genetic analysis based on state-of-the-art next-generation sequencing technology. Case presentation: A 17-year-old female index patient presented with dental (caries, impacted, retained and anteriorly located teeth) and atypical bone anomalies not resembling Gardner syndrome. She was first referred to our Genetic Counselling Unit at the age of 11 due to an atypical bone abnormality identified by a panoramic X-ray. Tooth 3.6 was surgically removed and the histopathology report revealed a Paget’s disease-like bone metabolic disorder with mixed osteoblastic and osteoclastic activity of the mandible. A small lumbar subcutaneous tumor was discovered by physical examination. Ultrasound examination of the tumor raised the possibility of a soft tissue propagation of chondromatosis. Her sister, 2 years younger at the age of 14, had some benign tumors (multiple exostoses, odontomas, epidermoid cysts) and impacted teeth. Their mother had also skeletal symptoms. Her lower teeth did not develop, the 9th-10th ribs were fused, and she complained of intermittent jaw pain. A cranial CT scan showed fibrous dysplasia on the cranial bones. Whole exome sequencing identified a heterozygous pathogenic nonsense mutation (c.4700C>G; p.Ser1567*) in the APC gene in the index patient’s DNA. Targeted sequencing revealed the same variant in the DNA of the other affected family members (the sister and the mother). Conclusion: Early diagnosis of this rare, genetically determined syndrome is very important, because of the potentially high malignant transformation of intestinal polyps. Dentists should be familiar with the typical maxillofacial features of this disorder, to be able to refer patients to genetic counseling. Dental anomalies often precede the intestinal polyposis and facilitate the early diagnosis, thereby increasing the patients’ chances of survival. Genetic analysis may be necessary in patients with atypical phenotypic signs. C1 [Antal, Greta] University of Pecs, Department of Oral and Maxillofacial SurgeryPecs, Hungary. [Zsigmond, Anna] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Till, Agnes] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Orsi, Eniko] University of Pecs, Department of Oral and Maxillofacial SurgeryPecs, Hungary. [Szanto, Ildiko] University of Pecs, Department of Oral and Maxillofacial SurgeryPecs, Hungary. [Buki, Gergely] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Kereskai, Laszlo] University of Pecs, Department of PathologyPecs, Hungary. [Herbert, Zsuzsanna] PTE KK, Radiologiai KlinikaPecs, Hungary. [Hadzsiev, Kinga] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Bene, Judit] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. RP Bene, J (reprint author), University of Pecs, Department of Medical Genetics and Child Development, Pecs, Hungary. EM bene.judit@pte.hu CR Oliveira MR, Rodrigues WC, Gabrielli MF, Gabrielli MA, Onofre MA, Filho VA. Gardner syndrome with unusual maxillofacial manifestation. J Craniofac Surg, 2016, 27(5):1253–5., DOI 10.1097/SCS. 0000000000002741 Salti L, Rasse M, Al-Ouf K. Maxillofacial Radiographic study of Gardner’s syndrome presenting with odontogenic myxoma: a rare case report. Stomatologija, 2018, 20(2):59–64. Bisgaard ML, Fenger K, Bulow S, Niebuhr E, Mohr J. Familial adenomatous polyposis, FAP): frequency, penetrance, and mutation rate. Hum Mutat, 1994, 3(2):121–5., DOI 10.1002/humu.1380030206 Yu D, Ng CB, Zhu H, Liu J, Lin Y. Bone and dental abnormalities as first signs of familial Gardner’s syndrome in a Chinese family: a literature review and a case report. Med Sci, Paris,, 2018, 34(Focus issue F1):20–5., DOI 10.1051/medsci/ 201834f104 Gardner EJ, Richards RC. Multiple cutaneous and subcutaneous lesions occurring simultaneously with hereditary polyposis and osteomatosis. Am J Hum Genet, 1953, 5(2):139–47. Cruz-Correa M, Giardiello FM. Familial adenomatous polyposis. Gastrointest Endosc, 2003, 58(6):885–94., DOI 10.1016/s0016-5107(03)02336-8 Vasen HF, Moslein G, Alonso A, Aretz S, Bernstein I, Bertario L, et al. Guidelines for the clinical management of familial adenomatous polyposis, FAP). Gut, 2008, 57(5):704–13., DOI 10.1136/gut.2007.136127 Davies AS. Gardner’s syndrome--a case report. Br J Oral Surg, 1970, 8(1):51–7., DOI 10.1016/s0007-117x(70)80068-3 D’Agostino S, Dell’Olio F, Tempesta A, Cervinara F, D’Amati A, Dolci M, et al. Osteoma of the jaw as first clinical sign of gardner’s syndrome: the experience of two Italian centers and review. J Clin Med, 2023, 12(4):1496., DOI 10.3390/jcm12041496 Wijn MA, Keller JJ, Giardiello FM, Brand HS. Oral and maxillofacial manifestations of familial adenomatous polyposis. Oral Dis, 2007, 13(4):360–5., DOI 10.1111/j.1601-0825.2006.01293.x Hanson CA, Miller JR. Non-traditional roles for the Adenomatous Polyposis Coli, APC, tumor suppressor protein. Gene, 2005, 361:1–12., DOI 10.1016/j.gene. 2005.07.024 Seehra J, Patel S, Bryant C. Gardner’s Syndrome revisited: a clinical case and overview of the literature. J Orthod, 2016, 43(1):59–64., DOI 10.1179/1465313315Y. 0000000008 Jarvinen HJ, Peltokallio P, LandtmanM,Wolf J. Gardner’s stigmas in patients with familial adenomatosis coli. Br J Surg, 1982, 69(12):718–21., DOI 10.1002/bjs. 1800691211 Lyons LA, Lewis RA, Strong LC, Zuckerbrod S, Ferrell RE. A genetic study of Gardner syndrome and congenital hypertrophy of the retinal pigment epithelium. Am J Hum Genet, 1988, 42(2):290–6. Gurbuz AK, Giardiello FM, Petersen GM, Krush AJ, Offerhaus GJ, Booker SV, et al. Desmoid tumours in familial adenomatous polyposis. Gut, 1994, 35(3): 377–81., DOI 10.1136/gut.35.3.377 Ben Lagha N, Galeazzi JM, Chapireau D, Oxeda P, Bouhnik Y, Maman L. Surgical management of osteoma associated with a familial Gardner’s syndrome. J Oral Maxillofac Surg, 2007, 65(6):1234–40., DOI 10.1016/j.joms.2005.10.047 Carl W, Herrera L. Dental and bone abnormalities in patients with familial polyposis coli. Semin Surg Oncol, 1987, 3(2):77–83., DOI 10.1002/ssu.2980030206 Panjwani S, Bagewadi A, Keluskar V, Arora S. Gardner’s syndrome. J Clin Imaging Sci, 2011, 1:65., DOI 10.4103/2156-7514.92187 Adisen MZ, Okkesim A, Misirlioglu M. The importance of early diagnosis of gardner’s syndrome in dental examination. Niger J Clin Pract, 2018, 21(1):114–6., DOI 10.4103/njcp.njcp_381_16 Fonseca LC, Kodama NK, Nunes FC, Maciel PH, Fonseca FA, Roitberg M, et al. Radiographic assessment of Gardner’s syndrome. Dentomaxillofac Radiol, 2007, 36(2):121–4., DOI 10.1259/dmfr/18554322 Joob-Fancsaly A, Pataky L, Martonffy K, Ujpal M, Hrabak K. Gardner syndrome--on the basis of three cases. Orv Hetil, 2000, 141(21):1139–41. Gorlin RJ, Cohen JMM, Hennekam RC Syndromes of the head and neck. 2nd ed., 1976). p. 324–8. Provenzale D, Gupta S, Ahnen DJ, Bray T, Cannon JA, Cooper G, et al. Genetic/familial high-risk assessment: colorectal version 1.2016, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw, 2016, 14(8):1010–30., DOI 10.6004/jnccn.2016.0108 Bailleul-Forestier I, Berdal A, Vinckier F, de Ravel T, Fryns JP, Verloes A. The genetic basis of inherited anomalies of the teeth. Part 2: syndromes with significant dental involvement. Eur J Med Genet, 2008, 51(5):383–408., DOI 10.1016/j.ejmg. 2008.05.003 Salerno C, D’Avola V, Oberti L, Almonte E, Bazzini EM, Tartaglia GM, et al. Rare genetic syndromes and oral anomalies: a review of the literature and case series with a new classification proposal. Children, Basel,, 2021, 9(1):12., DOI 10.3390/ children9010012 Furedi G, Varga I, Maj C, Szilagyi A, Madacsy L, Paal Z, et al. Gardner’s syndrome, a rare disease. Magyar sebeszet, 2019, 72(3):107–11., DOI 10.1556/1046. 72.2019.3.4 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611768 EP 1611773 DI 10.3389/pore.2024.1611768 PG 6 ER PT J AU Wang, Y Lei, Y Zheng, D Yang, Y Luo, L Li, J Xie, X AF Wang, Yi Lei, Yu Zheng, Delai Yang, Yanhui Luo, Lei Li, Ji Xie, Xiaoyang TI Prognostic value of lung immune prognostic index in non-small cell lung cancer patients receiving immune checkpoint inhibitors: a meta-analysis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE lung immune prognostic index; non-small cell lung cancer; immune checkpoint inhibitor; prognosis; meta-analysis ID lung immune prognostic index; non-small cell lung cancer; immune checkpoint inhibitor; prognosis; meta-analysis AB Background and Purpose: Until now, it has been difficult to accurately predict the efficacy of immunotherapy in patients with non-small cell lung cancer (NSCLC). A novel indicator, the lung immune prognostic index (LIPI), has shown relatively high prognostic value in patients with solid cancer. Therefore, this study aimed to further identify the association between LIPI and the survival of patients with NSCLC who receive immune checkpoint inhibitors (ICIs). Methods: Several electronic databases were searched for available publications up to April 23, 2023. Immunotherapy outcomes included overall survival (OS), progression-free survival (PFS), and hazard ratios (HRs) with 95% confidence intervals (CIs). Subgroup analysis based on the study design and comparison of the LIPI was conducted. Results: In thismeta-analysis, 21 studies with 9,010 patients were included in this meta-analysis. The pooled results demonstrated that elevated LIPI was significantly associated with poor OS (HR = 2.50, 95% CI:2.09–2.99, p < 0.001) and PFS (HR = 1.77, 95% CI:1.64–1.91, p < 0.001). Subgroup analyses stratified by study design (retrospective vs. prospective) and comparison of LIPI (1 vs. 0, 2 vs. 0, 1–2 vs. 0, 2 vs. 1 vs. 0, 2 vs. 0–1 and 2 vs. 1) showed similar results. Conclusion: LIPI could serve as a novel and reliable prognostic factor in NSCLC treated with ICIs, and elevated LIPI predicts worse prognosis. C1 [Wang, Yi] Neijiang Affiliated Hospital of Chongqing Medical University, The First People’s Hospital of Neijiang, Department of Thoracic SurgeryNeijiang, Sichuan, China. [Lei, Yu] Neijiang Affiliated Hospital of Chongqing Medical University, The First People’s Hospital of Neijiang, Department of Thoracic SurgeryNeijiang, Sichuan, China. [Zheng, Delai] Neijiang Affiliated Hospital of Chongqing Medical University, The First People’s Hospital of Neijiang, Department of Thoracic SurgeryNeijiang, Sichuan, China. [Yang, Yanhui] Neijiang Affiliated Hospital of Chongqing Medical University, The First People’s Hospital of Neijiang, Department of Thoracic SurgeryNeijiang, Sichuan, China. [Luo, Lei] Neijiang Affiliated Hospital of Chongqing Medical University, The First People’s Hospital of Neijiang, Department of Thoracic SurgeryNeijiang, Sichuan, China. [Li, Ji] Neijiang Affiliated Hospital of Chongqing Medical University, The First People’s Hospital of Neijiang, Department of Thoracic SurgeryNeijiang, Sichuan, China. [Xie, Xiaoyang] Neijiang Affiliated Hospital of Chongqing Medical University, The First People’s Hospital of Neijiang, Department of Thoracic SurgeryNeijiang, Sichuan, China. RP Xie, X (reprint author), Neijiang Affiliated Hospital of Chongqing Medical University, The First People’s Hospital of Neijiang, Department of Thoracic Surgery, Neijiang, China. EM 13990559066@163.com CR Torre LA, Siegel RL, Jemal A. Lung cancer statistics. Adv Exp Med Biol, 2016, 893:1–19., DOI 10.1007/978-3-319-24223-1_1 Jenkins R, Walker J. 2022 cancer statistics: focus on lung cancer. Future Oncol, London, England,, 2023, 1–11., DOI 10.2217/fon-2022-1214 Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA: a Cancer J clinicians, 2021, 71(1):7–33., DOI 10.3322/caac.21654 Ju W, Zheng R, Zhang S, Zeng H, Sun K, Wang S, et al. Cancer statistics in Chinese older people, 2022: current burden, time trends, and comparisons with the US, Japan, and the Republic of Korea. Sci China Life Sci, 2022, 66:1079–91., DOI 10. 1007/s11427-022-2218-x Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics. CA: a Cancer J clinicians, 2023, 73(1):17–30., DOI 10.3322/caac.21332 Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. New Engl J Med, 2015, 372(21):2018–28., DOI 10.1056/NEJMoa1501824 Adam T, Becker TM, Chua W, Bray V, Roberts TL. The multiple potential biomarkers for predicting immunotherapy response-finding the needle in the haystack. Cancers, 2021, 13(2):277., DOI 10.3390/cancers13020277 Dong A, Zhao Y, Li Z, Hu H. PD-L1 versus tumor mutation burden: which is the better immunotherapy biomarker in advanced non-small cell lung cancer? J Gene Med, 2021, 23(2):e3294., DOI 10.1002/jgm.3294 Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer, KEYNOTE-010): a randomised controlled trial. Lancet, London, England,, 2016, 387(10027):1540–50., DOI 10.1016/S0140- 6736(15)01281-7 Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. New Engl J Med, 2016, 375(19):1823–33., DOI 10.1056/NEJMoa1606774 Wang Y, Hu X, XuW,Wang H, Huang Y, Che G. Prognostic value of a novel scoring system using inflammatory response biomarkers in non-small cell lung cancer: a retrospective study. Thorac Cancer, 2019, 10(6):1402–11., DOI 10.1111/ 1759-7714.13085 Tian L, Wang Y, Tian J, Song W, Li L, Che G. Prognostic value and genome signature of m6A/m5C regulated genes in early-stage lung adenocarcinoma. Int J Mol Sci, 2023, 24(7):6520., DOI 10.3390/ijms24076520 Yang T, Hao L, Yang X, Luo C, Wang G, Lin CC, et al. Prognostic value of derived neutrophil-to-lymphocyte ratio, dNLR, in patients with non-small cell lung cancer receiving immune checkpoint inhibitors: a meta-analysis. BMJ Open, 2021, 11(9):e049123., DOI 10.1136/bmjopen-2021-049123 Platini H, Ferdinand E, Kohar K, Prayogo SA, Amirah S, Komariah M, et al. Neutrophil-to-Lymphocyte ratio and platelet-to-lymphocyte ratio as prognostic markers for advanced non-small-cell lung cancer treated with immunotherapy: a systematic review and meta-analysis. Medicina, Kaunas, Lithuania,, 2022, 58(8): 1069., DOI 10.3390/medicina58081069 Mezquita L, Auclin E, Ferrara R, Charrier M, Remon J, Planchard D, et al. Association of the lung immune prognostic index with immune checkpoint inhibitor outcomes in patients with advanced non-small cell lung cancer. JAMA Oncol, 2018, 4(3):351–7., DOI 10.1001/jamaoncol.2017.4771 Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ, Clinical research ed,, 2021, 372:n71., DOI 10.1136/bmj.n71 Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias. Biometrics, 1994, 50(4):1088–101., DOI 10.2307/2533446 Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ, Clinical research ed,, 1997, 315(7109): 629–34., DOI 10.1136/bmj.315.7109.629 Zeng X, Ye L, Luo M, Zeng D, Chen Y. Prognostic value of pretreatment systemic immune-inflammation index in Chinese esophageal squamous cell carcinoma patients receiving radical radiotherapy: a meta-analysis. Medicine, 2023, 102(25):e34117., DOI 10.1097/MD.0000000000034117 Kazandjian D, Gong Y, Keegan P, Pazdur R, Blumenthal GM. Prognostic value of the lung immune prognostic index for patients treated for metastatic nonsmall cell lung cancer. JAMA Oncol, 2019, 5(10):1481–5., DOI 10.1001/jamaoncol. 2019.1747 Ruiz-Banobre J, Areses-Manrique MC, Mosquera-Martinez J, Cortego A, Afonso-Afonso FJ, de Dios-Alvarez N, et al. Evaluation of the lung immune prognostic index in advanced non-small cell lung cancer patients under nivolumab monotherapy. Translational Lung Cancer Res, 2019, 8(6):1078–85., DOI 10.21037/tlcr.2019.11.07 Sorich MJ, Rowland A, Karapetis CS, Hopkins AM. Evaluation of the lung immune prognostic index for prediction of survival and response in patients treated with atezolizumab for non-small cell lung cancer: pooled analysis of clinical trials. J Thorac Oncol : official Publ Int Assoc Study Lung Cancer, 2019, 15., DOI 10.1016/j. jtho.2019.04.006 Mazzaschi G, Minari R, Zecca A, Cavazzoni A, Ferri V, Mori C, et al. Soluble PD-L1 and circulating cd8+pd-1+ and NK cells enclose a prognostic and predictive immune effector score in immunotherapy treated NSCLC patients. Lung Cancer, Amsterdam, Netherlands,, 2020, 148:1–11., DOI 10.1016/j.lungcan.2020.07.028 Wang W, Huang Z, Yu Z, Zhuang W, Zheng W, Cai Z, et al. Prognostic value of the lung immune prognostic index may differ in patients treated with immune checkpoint inhibitor monotherapy or combined with chemotherapy for non-small cell lung cancer. Front Oncol, 2020, 10:572853., DOI 10.3389/ fonc.2020.572853 Ali WAS, Hui P,Ma Y,Wu Y, Zhang Y, Chen Y, et al. Determinants of survival in advanced non-small cell lung cancer patients treatedwith anti-PD-1/PD-L1 therapy. Ann translational Med, 2021, 9(22):1639., DOI 10.21037/atm-21-1702 Chen P. Clinical study on prognostic factors of immunotherapy for non-small cell lung cancer. Taiyuan, China: Shanxi Medical University, 2021). Master. Galland L, Le Page AL, Lecuelle J, Bibeau F, Oulkhouir Y, Derangere V, et al. Prognostic value of Thyroid Transcription Factor-1 expression in lung adenocarcinoma in patients treated with anti PD-1/PD-L1. Oncoimmunology, 2021, 10(1):1957603., DOI 10.1080/2162402X.2021.1957603 Grosjean HAI, Dolter S, Meyers DE, Ding PQ, Stukalin I, Goutam S, et al. Effectiveness and safety of first-line pembrolizumab in older adults with PD-L1 positive non-small cell lung cancer: a retrospective cohort study of the Alberta immunotherapy database. Curr Oncol, 2021, 28(5):4213–22., DOI 10.3390/ curroncol28050357 Hopkins AM, Kichenadasse G, Abuhelwa AY, McKinnon RA, Rowland A, Sorich MJ. Value of the lung immune prognostic index in patients with non-small cell lung cancer initiating first-line atezolizumab combination therapy: subgroup analysis of the impower150 trial. Cancers, 2021, 13(5):1176–9., DOI 10.3390/ cancers13051176 Mountzios G, Samantas E, Senghas K, Zervas E, Krisam J, Samitas K, et al. Association of the advanced lung cancer inflammation index, ALI, with immune checkpoint inhibitor efficacy in patients with advanced non-small-cell lung cancer. ESMO open, 2021, 6(5):100254., DOI 10.1016/j.esmoop.2021.100254 Chen J, Wei S, Zhao T, Wang Y, Zhang X. Clinical significance of serum biomarkers in stage IV non-small-cell lung cancer treated with PD-1 inhibitors: LIPI score, NLR, dNLR, LMR, and PAB. Dis markers, 2022, 2022:7137357., DOI 10. 1155/2022/7137357 De Giglio A, Tassinari E, Zappi A, Di Federico A, Lenzi B, Sperandi F, et al. The palliative prognostic, PaP, score without clinical evaluation predicts early mortality among advanced NSCLC patients treated with immunotherapy. Cancers, 2022, 14(5845):5845., DOI 10.3390/cancers14235845 Holtzman L, Moskovitz M, Urban D, Nechushtan H, Keren S, Reinhorn D, et al. dNLR-based score predicting overall survival benefit for the addition of platinum-based chemotherapy to pembrolizumab in advanced NSCLC with PD-L1 tumor proportion score ≥50. Clin Lung Cancer, 2022, 23(2):122–34., DOI 10.1016/j. cllc.2021.12.006 Ortega-Franco A, Hodgson C, Raja H, Carter M, Lindsay C, Hughes S, et al. Real-world data on pembrolizumab for pretreated non-small-cell lung cancer: clinical outcome and relevance of the lung immune prognostic index. Targeted Oncol, 2022, 17(4):453–65., DOI 10.1007/s11523-022-00889-8 Tanaka S, Uchino J, Yokoi T, Kijima T, Goto Y, Suga Y, et al. Prognostic nutritional index and lung immune prognostic index as prognostic predictors for combination therapies of immune checkpoint inhibitors and cytotoxic anticancer chemotherapy for patients with advanced non-small cell lung cancer. Diagnostics, 2022, 12(2):423., DOI 10.3390/diagnostics12020423 Zhou J, Bao M, Gao G, Cai Y, Wu L, Lei L, et al. Increased blood-based intratumor heterogeneity, bITH, is associated with unfavorable outcomes of immune checkpoint inhibitors plus chemotherapy in non-small cell lung cancer. BMC Med, 2022, 20(256):256., DOI 10.1186/s12916-022-02444-8 Zhou S, Ren F, Meng X. Efficacy of immune checkpoint inhibitor therapy in EGFR mutation-positive patients with NSCLC and brain metastases who have failed EGFR-TKI therapy. Front Immunol, 2022, 13:955944., DOI 10.3389/fimmu.2022. 955944 Zhou Y, Wu L, Xu T, Ma C, Song L, Zhang J. Predictive value of lung immune prognostic index scoring system for patients treated with immunotherapy in nonsmall cell lung cancer. Clin J Med Offic, 2022, 50(03):237–41., DOI 10.16680/j.1671- 3826.2022.03.05 Huang J, Pu H, He J, Tang X. Prognostic value of the lung immune prognostic index for metastatic non-small cell lung cancer patients: a Chinese cohort study. Int J Gen Med, 2023, 16:881–93., DOI 10.2147/IJGM.S393263 Shah SC, Itzkowitz SH. Colorectal cancer in inflammatory bowel disease: mechanisms and management. Gastroenterology, 2022, 162(3):715–30.e3., DOI 10. 1053/j.gastro.2021.10.035 KolbM. Inflammation and dysregulated fibroblast proliferation--newmechanisms? Sarcoidosis, vasculitis, diffuse Lung Dis : official J WASOG, 2013, 30(Suppl. 1):21–6. Yu K, LiD, Xu F, GuoH, Feng F,Ding Y, et al. Ido1 as a newimmune biomarker for diabetic nephropathy and its correlation with immune cell infiltration. Int immunopharmacology, 2021, 94:107446., DOI 10.1016/j.intimp.2021.107446 Ai L, Xu A, Xu J. Roles of PD-1/PD-L1 pathway: signaling, cancer, and beyond. Adv Exp Med Biol, 2020, 1248:33–59., DOI 10.1007/978-981-15-3266-5_3 Mizuno R, Kawada K, Itatani Y, Ogawa R, Kiyasu Y, Sakai Y. The role of tumor-associated neutrophils in colorectal cancer. Int J Mol Sci, 2019, 20(3):529., DOI 10.3390/ijms20030529 Yang R, Zhong L, Yang XQ, Jiang KL, Li L, Song H, et al. Neutrophil elastase enhances the proliferation and decreases apoptosis of leukemia cells via activation of PI3K/Akt signaling. Mol Med Rep, 2016, 13(5):4175–82., DOI 10.3892/mmr.2016.5051 Liu C, Liu R, Wang B, Lian J, Yao Y, Sun H, et al. Blocking IL-17A enhances tumor response to anti-PD-1 immunotherapy in microsatellite stable colorectal cancer. J Immunother Cancer, 2021, 9(1):e001895., DOI 10.1136/jitc-2020-001895 Huang H, Liu Q, Zhu L, Zhang Y, Lu X, Wu Y, et al. Prognostic value of preoperative systemic immune-inflammation index in patients with cervical cancer. Scientific Rep, 2019, 9(1):3284., DOI 10.1038/s41598-019-39150-0 Takada K, Kashiwagi S, Asano Y, Goto W, Morisaki T, Shibutani M, et al. Differences in tumor-infiltrating lymphocyte density and prognostic factors for breast cancer by patient age. World J Surg Oncol, 2022, 20(1):38., DOI 10.1186/ s12957-022-02513-5 Comandatore A, Franczak M, Smolenski RT,Morelli L, Peters GJ, Giovannetti E. Lactate Dehydrogenase and its clinical significance in pancreatic and thoracic cancers. Semin Cancer Biol, 2022, 86(Pt 2):93–100., DOI 10.1016/j.semcancer.2022.09.001 Dang C, Deng YH, Qin RY. Prognostic value of ALP and LDH in periampullary carcinoma patients undergoing surgery. Curr Med Sci, 2022, 42(1):150–8., DOI 10.1007/s11596-021-2452-9 Ye Y, Chen M, Chen X, Xiao J, Liao L, Lin F. Clinical significance and prognostic value of lactate dehydrogenase expression in cervical cancer. Genet Test Mol biomarkers, 2022, 26(3):107–17., DOI 10.1089/gtmb.2021.0006 Paredes F, Williams HC, San Martin A. Metabolic adaptation in hypoxia and cancer. Cancer Lett, 2021, 502:133–42., DOI 10.1016/j.canlet.2020.12.020 Shao X, Hua S, Feng T, Ocansey DKW, Yin L. Hypoxia-regulated tumorderived exosomes and tumor progression: a focus on immune evasion. Int J Mol Sci, 2022, 23(19):11789., DOI 10.3390/ijms231911789 Korbecki J, Siminska D, Gassowska-Dobrowolska M, Listos J, Gutowska I, Chlubek D, et al. Chronic and cycling hypoxia: drivers of cancer chronic inflammation through HIF-1 and NF-κB activation: a review of the molecular mechanisms. Int J Mol Sci, 2021, 22(19):10701., DOI 10.3390/ijms221910701 Melincovici CS, Bosca AB, Susman S, Marginean M, Mihu C, Istrate M, et al. Vascular endothelial growth factor, VEGF, - key factor in normal and pathological angiogenesis. Rom J Morphol Embryol = Revue roumaine de morphologie embryologie, 2018, 59(2):455–67. Liu H, Yang XL, Yang XY, Dong ZR, Chen ZQ, Hong JG, et al. The prediction potential of the pretreatment lung immune prognostic index for the therapeutic outcomes of immune checkpoint inhibitors in patients with solid cancer: a systematic review and meta-analysis. Front Oncol, 2021, 11:691002., DOI 10.3389/fonc.2021.691002 Xie J, Zang Y, Liu M, Peng L, Zhang H. The lung immune prognostic index may predict the efficacy of different treatments in patients with advanced NSCLC: a meta-analysis. Oncol Res Treat, 2021, 44(4):164–75., DOI 10. 1159/000514443 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611773 EP 1611785 DI 10.3389/pore.2024.1611773 PG 13 ER PT J AU Papai-Szekely, Zs Grmela, G Sarosi, V AF Papai-Szekely, Zsolt Grmela, Gabor Sarosi, Veronika TI Novel diagnostic processes and challenges in bronchoscopy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE diagnostic bronchoscopy; respiratory medicine; endobronchial ultrasound; virtual bronchoscopy; autofluorescence bronchoscopy ID diagnostic bronchoscopy; respiratory medicine; endobronchial ultrasound; virtual bronchoscopy; autofluorescence bronchoscopy AB Diagnostic bronchoscopy is a minimally invasive procedure that plays a crucial role in the diagnosis and management of various respiratory conditions. This paper explores the advancements in technology that have revolutionized the field and focuses on the new diagnostic procedures in bronchoscopy that have emerged in recent years. These innovative techniques have expanded the diagnostic capabilities of bronchoscopy, allowing for more accurate and comprehensive evaluation of respiratory conditions. This paper will also discuss the challenges in the diagnostic process with bronchoscope. C1 [Papai-Szekely, Zsolt] Szent Gyorgy University Teaching Hospital of Fejer County, Department of PulmonologySzekesfehervar, Hungary. [Grmela, Gabor] Szent Gyorgy University Teaching Hospital of Fejer County, Department of PulmonologySzekesfehervar, Hungary. [Sarosi, Veronika] University of Pecs, Faculty of Medicine, Department of PulmonologyPecs, Hungary. RP Papai-Szekely, Zs (reprint author), Szent Gyorgy University Teaching Hospital of Fejer County, Department of Pulmonology, Szekesfehervar, Hungary. EM zsoltpapai@yahoo.com CR Silvestri GA, Gonzalez AV, Jantz MA, Margolis ML, Gould MK, Tanoue LT, et al. Methods for staging non-small cell lung cancer: diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest, 2013, 143(Suppl. 5):e211S–e250S., DOI 10.1378/chest.12- 2355 Lam B, Wong MP, Fung SL, Lam DC, Wong PC, Mok TY, et al. The clinical value of autofluorescence bronchoscopy for the diagnosis of lung cancer. Eur Respir J, 2006, 28(5):915–9., DOI 10.1183/09031936.06.00131405 Goorsenberg A, Kalverda KA, Annema J, Bonta P. Advances in optical coherence tomography and confocal laser endomicroscopy in pulmonary diseases. Respiration, 2020, 99(3):190–205., DOI 10.1159/000503261 Zheng Y, Zhang L, Lou Y, Fan B, Cui Y, Wu X, et al. The cryobiopsy in interstitial lung diseases guided by probe-based confocal laser endomicroscopy is feasible. Clin Respir J, 2023, 17(10):998–1005., DOI 10. 1111/crj.13669 Burks AC, Akulian J. Bronchoscopic diagnostic procedures available to the pulmonologist. Clin Chest Med, 2020, 41:129–44., DOI 10.1016/j.ccm.2019. 11.002 Kalanjeri S, Holladay RC, Gildea TR. State-of-the-Art modalities for peripheral lung nodule biopsy. Clin Chest Med, 2018, 39:125–38., DOI 10.1016/j.ccm.2017. 11.007 Oki M, Saka H, Ando M, Asano F, Kurimoto N, Morita K, et al. Ultrathin bronchoscopy with multimodal devices for peripheral pulmonary lesions. A randomized trial. Am J Respir Crit Care Med, 2015, 192:468–76., DOI 10.1164/ rccm.201502-0205OC Jiang S, Xie F, Mao X,Ma H, Sun J. The value of navigation bronchoscopy in the diagnosis of peripheral pulmonary lesions: a meta-analysis. Thorac Cancer, 2020, 11:1191–201., DOI 10.1111/1759-7714.13373 Cherian SV, Kaur S, Karanth S, Xian JZ, Estrada-Y-Martin RM. Diagnostic yield of electromagnetic navigational bronchoscopy: a safety net community-based hospital experience in the United States. Ann Thorac Med, 2021, 16:102–9., DOI 10. 4103/atm.ATM_388_20 Kato A, Yasuo M, Tokoro Y, Kobayashi T, Ichiyama T, Tateishi K, et al. Virtual bronchoscopic navigation as an aid to CT-guided transbronchial biopsy improves the diagnostic yield for small peripheral pulmonary lesions. Respirology, 2018, 23:1049–54., DOI 10.1111/resp.13377 Giri M, Puri A, Wang T, Huang G, Guo S. Virtual bronchoscopic navigation versus non-virtual bronchoscopic navigation assisted bronchoscopy for the diagnosis of peripheral pulmonary lesions: a systematic review and metaanalysis. Ther Adv Respir Dis, 2021, 15:17534666211017048., DOI 10.1177/ 17534666211017048 Casal RF. Cone beam CT-guided bronchoscopy: here to stay? J Bronchol Interv Pulmonol, 2018, 25:255–6., DOI 10.1097/LBR.0000000000000546 Murgu SD. Robotic assisted-bronchoscopy: technical tips and lessons learned from the initial experience with sampling peripheral lung lesions. BMC Pulm Med, 2019, 19:89–8., DOI 10.1186/s12890-019-0857-z Fielding DI, Bashirzadeh F, Son JH, Todman M, Chin A, Tan L, et al. First human use of a new robotic-assisted fiber optic sensing navigation system for small peripheral pulmonary nodules. Respiration, 2019, 98:142–50., DOI 10.1159/ 000498951 Rojas-Solano JR, Ugalde-Gamboa L, Machuzak M. Robotic bronchoscopy for diagnosis of suspected lung cancer: a feasibility study. J Bronchol Interv Pulmonol, 2018, 25:168–75., DOI 10.1097/LBR.0000000000000499 Chaddha U, Hogarth DK, Murgu S Bronchoscopic ablative therapies for malignant central airway obstruction and peripheral lung tumors. Ann Am Thorac Soc, 2019, 16:1220–9., DOI 10.1513/AnnalsATS.201812-892CME NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611774 EP 1611782 DI 10.3389/pore.2024.1611774 PG 9 ER PT J AU Editorial Office, AF Editorial Office, TI Retraction: Case report: A rare case of tumor-to-tumor metastasis: metastatic lobular breast carcinoma to clear cell renal cell carcinoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Retraction AB A Retraction of the Case Report Article Case report: A rare case of tumor-to-tumor metastasis: metastatic lobular breast carcinoma to clear cell renal cell carcinoma by Zhang L, Yuan P, Cao Q, Mu J, Ying J and Guo C (2023). Pathol. Oncol. Res. 29:1611204. doi: 10.3389/pore.2023.1611204 C1 [Editorial Office, ] Pathology & Oncology ResearchBudapest, Hungary. RP Editorial Office, (reprint author), Pathology & Oncology Research, Budapest, Hungary. EM editorialoffice@por-journal.com NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611784 EP 1611784 DI 10.3389/pore.2024.1611784 PG 1 ER PT J AU Imanbayev, N Iztleuov, Y Kamyshanskiy, Y Zhumasheva, A AF Imanbayev, M. Nauryzbay Iztleuov, M. Yerbolat Kamyshanskiy, K. Yevgeniy Zhumasheva, V. Aigul TI Diagnostic and prognostic significance of keloid-like collagen remodeling patterns in the extracellular matrix of colorectal cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE colorectal cancer; intermediate desmoplastic reaction; keloid-like collagen; desmoplastic reaction; extracellular matrix of colorectal cancer ID colorectal cancer; intermediate desmoplastic reaction; keloid-like collagen; desmoplastic reaction; extracellular matrix of colorectal cancer AB Background: The desmoplastic reaction is considered a promising prognostic parameter for colorectal cancer. However, intermediate desmoplastic reaction is characterized by sizeable stromal heterogeneity, including both small amounts of keloid-like collagen (KC) in the fibrotic stroma and thick tufts of KC circumferentially surrounding cancer nests and occupying most of the fields of view. The present study aimed to evaluate the diagnostic and prognostic significance of KC histophenotyping with a quantitative visual assessment of its presence in the stroma of the invasive margin of TNM (The “tumor-node-metastasis” classification) stage II/III colorectal cancer (CRC). Methods and results: 175 resected tumors from patients with TNM stage II/III CRC were examined. Keloid-like collagen was assessed according to Ueno H. criteria. KC was assessed at the primary tumor invasive margin using Hematoxylin & Eosin and Masson’s trichrome staining. The cut-off point for KC was examined using “the best cutoff approach by log-rank test.” Using a cutoff point of 30%, we histologically divided fibrous stroma in the invasive area into two groups: “type A”—KC ≤ 0.3 and “type B”—KC>0.3. Type A stroma was observed in 48% of patients, type B—in 52%. The association between collagen amount and 5-year recurrence-free survival (5-RFS) was assessed using Cox regression analysis. Kaplan-Meier analysis and logrank tests were used to assess the significance of survival analysis. Analysis of categorical variables showed that increased KC in CRC stroma predicted adverse outcomes for 5-RFS (hazard ratio [HR] = 3.143, 95%, confidence interval [CI] = 1.643–6.012, p = 0.001). Moreover, in Kaplan-Meier analysis, the log-rank test showed that type B exhibited worse 5- RFS than type A (p = 0.000). Conclusion: KC is an independent predictor of 5-year overall and RFS in patients with TNM stage II/III CRC treated with surgery, with worse survival rates when the amount of KC increases by >30%. C1 [Imanbayev, M. Nauryzbay] West Kazakhstan Marat Ospanov Medical University, Department of OncologyAktobe, Kazakhstan. [Iztleuov, M. Yerbolat] West Kazakhstan Marat Ospanov Medical University, Department of RadiologyAktobe, Kazakhstan. [Kamyshanskiy, K. Yevgeniy] Karaganda Medical University, Department of PathologyKaraganda, Kazakhstan. [Zhumasheva, V. Aigul] Medical Centre of West Kazakhstan Marat Ospanov Medical University, Department of PathomorphologyAktobe, Kazakhstan. RP Imanbayev, N (reprint author), West Kazakhstan Marat Ospanov Medical University, Department of Oncology, Aktobe, Kazakhstan. EM dr.imanbayev@gmail.com CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Li J, Yi CH, Hu YT, Yuan Y, Zhang SZ, Zheng S, et al. TNM staging of colorectal cancer should be reconsidered according to weighting of the T stage: verification based on a 25-year follow-up. Medicine, Baltimore,, 2016, 95(6):e2711., DOI 10. 1097/MD.0000000000002711 Son GM, Kwon MS, Shin DH, Shin N, Ryu D, Kang CD. Comparisons of cancer-associated fibroblasts in the intratumoral stroma and invasive front in colorectal cancer. Medicine, Baltimore,, 2019, 98(18):e15164., DOI 10.1097/MD. 0000000000015164 Nearchou IP, Kajiwara Y, Mochizuki S, Harrison DJ, Caie PD, Ueno H. Novel internationally verified method reports desmoplastic reaction as the most significant prognostic feature for disease-specific survival in stage II colorectal cancer. Am J Surg Pathol, 2019, 43(9):1239–48., DOI 10.1097/PAS. 0000000000001304 Ueno H, Kajiwara Y, Ajioka Y, Sugai T, Sekine S, Ishiguro M, et al. Histopathological atlas of desmoplastic reaction characterization in colorectal cancer. Jpn J Clin Oncol, 2021, 51(6):1004–12., DOI 10.1093/jjco/hyab040 Masson P Human tumors: histology, diagnosis, and technique. 2d ed., rev. And enl. Translated by sidney D. Kobernick. Wayne State University Press, 1970). Ueno H, Jones AM, Wilkinson KH, Jass JR, Talbot IC. Histological categorisation of fibrotic cancer stroma in advanced rectal cancer. Gut, 2004, 53(4):581–6., DOI 10.1136/gut.2003.028365 Shin N, Son GM, Shin DH, Kwon MS, Park BS, Kim HS, et al. Cancerassociated fibroblasts and desmoplastic reactions related to cancer invasiveness in patients with colorectal cancer. Ann Coloproctol, 2019, 35(1):36–46., DOI 10.3393/ac. 2018.09.10 Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun, 2020, 11(1):5120. Published 2020 Oct 9., DOI 10.1038/s41467-020- 18794-x Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. Extracellular matrix structure. Adv Drug Deliv Rev, 2016, 97:4–27., DOI 10.1016/j.addr.2015.11.001 Penet MF, Kakkad S, Pathak AP, Krishnamachary B, Mironchik Y, Raman V, et al. Structure and function of a prostate cancer dissemination-permissive extracellular matrix. Clin Cancer Res, 2017, 23(9):2245–54., DOI 10.1158/1078- 0432.CCR-16-1516 Chen D, Liu Z, Liu W, Fu M, Jiang W, Xu S, et al. Predicting postoperative peritoneal metastasis in gastric cancer with serosal invasion using a collagen nomogram. Nat Commun, 2021, 12(1):179. Published 2021 Jan 8., DOI 10.1038/ s41467-020-20429-0 Desa DE, Bhanote M, Hill RL, Majeski JB, Buscaglia B, D’Aguiar M, et al. Second-harmonic generation directionality is associated with neoadjuvant chemotherapy response in breast cancer core needle biopsies. J Biomed Opt, 2019, 24(8):1–9., DOI 10.1117/1.JBO.24.8.086503 Jiang W, LiM, Tan J, Feng M, Zheng J, Chen D, et al. A nomogram based on a collagen feature support vector machine for predicting the treatment response to neoadjuvant chemoradiotherapy in rectal cancer patients. Ann Surg Oncol, 2021, 28(11):6408–21., DOI 10.1245/s10434-021-10218-4 Jiang W, Wang S, Wan J, Zheng J, Dong X, Liu Z, et al. Association of the collagen signature with pathological complete response in rectal cancer patients. Cancer Sci, 2022, 113(7):2409–24., DOI 10.1111/cas.15385 Kakkad SM, Penet MF, Akhbardeh A, Pathak AP, Solaiyappan M, Raman V, et al. Hypoxic tumor environments exhibit disrupted collagen I fibers and low macromolecular transport. PLoS One, 2013, 8(12):e81869. Published 2013 Dec 12., DOI 10.1371/journal.pone.0081869 Wolf K, Alexander S, Schacht V, Coussens LM, von Andrian UH, van Rheenen J, et al. Collagen-based cell migration models in vitro and in vivo. Semin Cel Dev Biol, 2009, 20(8):931–41., DOI 10.1016/j.semcdb.2009. 08.005 Egeblad M, Ewald AJ, Askautrud HA, Truitt ML, Welm BE, Bainbridge E, et al. Visualizing stromal cell dynamics in different tumor microenvironments by spinning disk confocal microscopy. Dis Model Mech, 2008, 1(2- 3):155–67., DOI 10. 1242/dmm.000596 Han W, Chen S, Yuan W, Fan Q, Tian J, Wang X, et al. Oriented collagen fibers direct tumor cell intravasation. Proc Natl Acad Sci U S A, 2016, 113(40): 11208–13., DOI 10.1073/pnas.1610347113 Conklin MW, Gangnon RE, Sprague BL, Van Gemert L, Hampton JM, Eliceiri KW, et al. Collagen alignment as a predictor of recurrence after ductal carcinoma in situ. Cancer Epidemiol Biomarkers Prev, 2018, 27(2):138–45., DOI 10.1158/1055- 9965.EPI-17-0720 Conklin MW, Eickhoff JC, Riching KM, Pehlke CA, Eliceiri KW, Provenzano PP, et al. Aligned collagen is a prognostic signature for survival in human breast carcinoma. Am J Pathol, 2011, 178(3):1221–32., DOI 10.1016/j. ajpath.2010.11.076 Najafi M, Farhood B, Mortezaee K. Extracellular matrix, ECM, stiffness and degradation as cancer drivers. J Cel Biochem, 2019, 120(3):2782–90., DOI 10.1002/ jcb.27681 Brauchle E, Kasper J, Daum R, Schierbaum N, Falch C, Kirschniak A, et al. Biomechanical and biomolecular characterization of extracellular matrix structures in human colon carcinomas. Matrix Biol, 2018, 68-69:180–93., DOI 10.1016/j. matbio.2018.03.016 Paul CD, Hruska A, Staunton JR, Burr HA, Daly KM, Kim J, et al. Probing cellular response to topography in three dimensions. Biomaterials, 2019, 197: 101–18., DOI 10.1016/j.biomaterials.2019.01.009 Joint Commission on the Quality of Medical Services of theMinistry of Health of the Republic of Kazakhstan. Clinical protocol for diagnosis and treatment of malignant tumors of the colon was approved by the joint commission on the quality of medical services of the ministry of health of the republic of Kazakhstan dated july 1, 2022 protocol No. 164, 2022). Imai T. Histological comparison of cancer of the stomach in autopsy and operation cases. Jpn J Cancer Res, 1949, 40:199–201. Lugli A, Kirsch R, Ajioka Y, Bosman F, Cathomas G, Dawson H, et al. Recommendations for reporting tumor budding in colorectal cancer based on the International Tumor Budding Consensus Conference, ITBCC, 2016. Mod Pathol, 2017, 30:1299–311., DOI 10.1038/modpathol.2017.46 Lim SB, Yu CS, Jang SJ, Kim TW, Kim JH, Kim JC. Prognostic significance of lymphovascular invasion in sporadic colorectal cancer. Dis Colon Rectum, 2010, 53(4):377–84., DOI 10.1007/DCR.0b013e3181cf8ae5 Huh JW, Kim HR, Kim YJ. Prognostic value of perineural invasion in patients with stage II colorectal cancer. Ann Surg Oncol, 2010, 17(8):2066–72., DOI 10.1245/ s10434-010-0982-7 Fagan JJ, Collins B, Barnes L, D’Amico F, Myers EN, Johnson JT. Perineural invasion in squamous cell carcinoma of the head and neck. Arch Otolaryngol Head Neck Surg, 1998, 124(6):637–40., DOI 10.1001/archotol.124.6.637 Yilmaz T, Hosal AS, Gedikoglu G, Onerci M, Gursel B. Prognostic significance of vascular and perineural invasion in cancer of the larynx. Am J Otolaryngol, 1998, 19(2):83–8., DOI 10.1016/s0196-0709(98)90100-4 Akimoto N, Vayrynen JP, Zhao M, Ugai T, Fujiyoshi K, Borowsky J, et al. Desmoplastic reaction, immune cell response, and prognosis in colorectal cancer. Front Immunol, 2022, 13:840198. Published 2022 Mar 22., DOI 10.3389/fimmu.2022.840198 Sandberg TP, Stuart MPME, Oosting J, Tollenaar RAEM, Sier CFM, MeskerWE. Increased expression of cancer-associated fibroblast markers at the invasive front and its association with tumor-stroma ratio in colorectal cancer. BMC Cancer, 2019, 19(1):284. Published 2019 Mar 29., DOI 10.1186/s12885-019-5462-2 Brown KM, Xue A, Smith RC, Samra JS, Gill AJ, Hugh TJ. Cancer-associated stroma reveals prognostic biomarkers and novel insights into the tumour microenvironment of colorectal cancer and colorectal liver metastases. Cancer Med, 2022, 11(2):492–506., DOI 10.1002/cam4.4452 Arun Gopinathan P, Kokila G, Jyothi M, Ananjan C, Pradeep L, Humaira Nazir S. Study of collagen birefringence in different grades of oral squamous cell carcinoma using picrosirius red and polarized light microscopy. Scientifica, Cairo)., 2015, 2015:802980., DOI 10.1155/2015/802980 Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer, 2009, 9(4):239–52., DOI 10.1038/nrc2618 Bissell MJ, Radisky D. Putting tumours in context. Nat Rev Cancer, 2001, 1(1):46–54., DOI 10.1038/35094059 Beacham DA, Cukierman E. Stromagenesis: the changing face of fibroblastic microenvironments during tumor progression. Semin Cancer Biol, 2005, 15(5): 329–41., DOI 10.1016/j.semcancer.2005.05.003 Guinney J, Dienstmann R, Wang X, de Reynies A, Schlicker A, Soneson C, et al. The consensus molecular subtypes of colorectal cancer. Nat Med, 2015, 21(11):1350–6., DOI 10.1038/nm.3967 Quail DF, Joyce JA.Microenvironmental regulation of tumor progression and metastasis. Nat Med, 2013, 19(11):1423–37., DOI 10.1038/nm.3394 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611789 EP 1611798 DI 10.3389/pore.2024.1611789 PG 10 ER PT J AU Della Corte, L Ascione, M Bifulco, G AF Della Corte, Luigi Ascione, Mario Bifulco, Giuseppe TI Modified clock mapping biopsy sec. Della Corte-Bifulco in the preoperative assessment of excisional surgery for vulvar Paget’s disease SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE gynecological cancer; extramammary Paget’s disease of the vulva; vulvar malignant lesion; preoperative biopsy; new technique ID gynecological cancer; extramammary Paget’s disease of the vulva; vulvar malignant lesion; preoperative biopsy; new technique AB We have developed a biopsy technique aimed at preoperative evaluating the extent of Paget’s vulvar disease in order to plan subsequent radical vulvar surgery. The aim is to find all possible lesion sites that are not visible macroscopically, to obtain a clear evaluation of the disease spread and to tailor the radical surgical procedure to remove even microscopic lesions, avoiding recurrences and excessively destructive surgery, adopting as conservative an approach as possible. We used this procedure for the first time to establish the radicality of the surgical intervention in a 68-year-old patient initially suffering from a single invasive vulvar Paget’s lesion. C1 [Della Corte, Luigi] University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and DentistryNaples, Italy. [Ascione, Mario] University of Naples Federico II, Department of Public HealthNaples, Italy. [Bifulco, Giuseppe] University of Naples Federico II, Department of Public HealthNaples, Italy. RP Della Corte, L (reprint author), University of Naples Federico II, School of Medicine, Department of Neuroscience, Reproductive Sciences and Dentistry, Naples, Italy. EM dellacorte.luigi25@gmail.com CR Lloyd J, Flanagan A. Mammary and extramammary Paget’s disease. J Clin Pathol, 2000, 53:742–9., DOI 10.1136/jcp.53.10.742 Lam C, Funaro D. Extramammary Paget’s disease: summary of current knowledge. Dermatol Clin, 2010, 28:807–26., DOI 10.1016/j.det.2010.08.002 Noel A, Bhat RM, Rao SV, Fernandes S. An unusual secondary extramammary Paget’s disease of vulva. Indian J Sex Transm Dis AIDS, 2020, 41:210–2., DOI 10. 4103/ijstd.IJSTD_69_19 McDaniel B, Brown F, Crane JS. Extramammary Paget disease. Treasure Island, FL, USA: StatPearls Publishing, 2022). Della Corte L, Cafasso V, Conte C, Cuomo L, Giampaolinio P, Lavitola G, et al. Medical and surgical strategies in vulvar Paget disease: let’s throw some light!. J Pers Med, 2023, 13(1):100., DOI 10.3390/jpm13010100 Zollo JD, Zeitouni NC. The roswell park cancer institute experience with extramammary Paget’s disease. Br J Dermatol, 2000, 142:59–65., DOI 10.1046/j. 1365-2133.2000.03242.x Hatta N, Yamada M, Hirano T, Fujimoto A, Morita R. Extramammary Paget’s disease: treatment, prognostic factors and outcome in 76 patients. Br J Dermatol, 2008, 158(2):313–8., DOI 10.1111/j.1365-2133.2007.08314.x Borghi C, Bogani G, Ditto A, Martinelli F, Signorelli M, Chiappa V, et al. Invasive Paget disease of the vulva. Int J Gynecol Cancer, 2018, 28(1):176–82., DOI 10.1097/IGC.0000000000001131 Garganese G, Inzani F, Mantovani G, Santoro A, Valente M, Babini G, et al. The vulvar immunohistochemical panel, VIP, project: molecular profiles of vulvar Paget’s disease. J Cancer Res Clin Oncol, 2019, 145(9):2211–25., DOI 10.1007/s00432- 019-02975-3 Kato T, Fujimoto N, Fujii N, Tanaka T. Mapping biopsy with punch biopsies to determine surgical margin in extramammary Paget’s disease. J Dermatol, 2013, 40:968–72., DOI 10.1111/1346-8138.12347 GarganeseG, Anchora LP, Fragomeni SM, MantovaniG, Santoro A, Gentileschi S, et al. “Clock mapping” prior to excisional surgery in vulvar Paget’s disease: tailoring the surgical plan. Arch Gynecol Obstet, 2022, 306:473–80., DOI 10.1007/s00404-022-06408-4 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611803 EP 1611806 DI 10.3389/pore.2024.1611803 PG 4 ER PT J AU Jeon, H Gor, R D’Aiello, A Stiles, B Illei, P Halmos, B AF Jeon, Hyein Gor, Rajvi D’Aiello, Angelica Stiles, Brendon Illei, B. Peter Halmos, Balazs TI Advancing neoadjuvant therapies in resectable non-small cell lung cancer: implications for novel treatment strategies and biomarker discovery SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE NSCLC; perioperative; neoadjuvant; adjuvant; lung cancer ID NSCLC; perioperative; neoadjuvant; adjuvant; lung cancer AB The delivery of neoadjuvant and perioperative therapies for non-small cell lung cancer has been radically altered by significant advances and by the incorporation of targeted therapies as well as immune checkpoint inhibitors alone or alongside conventional chemotherapy. This evolution has been particularly notable in the incorporation of immunotherapy and targeted therapy into the treatment of resectable NSCLC, where recent FDA approvals of drugs such as nivolumab and pembrolizumab, in combination with platinum doublet chemotherapy, have led to considerable improvements in pathological complete response rates and the potential for enhanced longterm survival outcomes. This review emphasizes the growing importance of biomarkers in optimizing treatment selection and explores the impact of emerging studies that challenge existing treatment paradigms and investigate novel therapeutic combinations poised to redefine standard of care practices. Furthermore, the discussion extends to the unmet needs within perioperative treatment assessment and prognostication, highlighting the prospective value of biomarkers in evaluating treatment responses and prognosis. C1 [Jeon, Hyein] Montefiore Medical Center/Albert Einstein College of Medicine, Department of OncologyBronx, NY, USA. [Gor, Rajvi] Jacobi Medical Center, Department of MedicineBronx, NY, USA. [D’Aiello, Angelica] Montefiore Medical Center/Albert Einstein College of Medicine, Department of OncologyBronx, NY, USA. [Stiles, Brendon] Albert Einstein College of Medicine, Montefiore Medical Center, Department of Cardiothoracic and Vascular SurgeryBronx, NY, USA. [Illei, B. Peter] Johns Hopkins School of Medicine, Department of PathologyBaltimore, MD, USA. [Halmos, Balazs] Montefiore Medical Center/Albert Einstein College of Medicine, Department of OncologyBronx, NY, USA. RP Halmos, B (reprint author), Montefiore Medical Center/Albert Einstein College of Medicine, Department of Oncology, Bronx, USA. EM bahalmos@montefiore.org CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J Clinicians, 2021, 71(3): 209–49., DOI 10.3322/caac.21660 Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, et al. Paclitaxel–carboplatin alone or with bevacizumab for non–small-cell lung cancer. New Engl J Med, 2006, 355(24):2542–50., DOI 10.1056/ NEJMoa061884 Vansteenkiste JF, Cho BC, Vanakesa T, De Pas T, Zielinski M, Kim MS, et al. Efficacy of the MAGE-A3 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive non-small-cell lung cancer, MAGRIT): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol, 2016, 17(6):822–35., DOI 10.1016/S1470-2045(16)00099-1 Felip E, Rosell R, Maestre JA, Rodriguez-Paniagua JM, Moran T, Astudillo J, et al. Preoperative chemotherapy plus surgery versus surgery plus adjuvant chemotherapy versus surgery alone in early-stage non–small-cell lung cancer. J Clin Oncol, 2010, 28(19):3138–45., DOI 10.1200/JCO.2009.27.6204 LimE, Harris G, PatelA,Adachi I, Edmonds L, Song F. Preoperative versus postoperative chemotherapy in patients with resectable non-small cell lung cancer: systematic review and indirect comparison meta-analysis of randomized trials. J Thorac Oncol, 2009, 4(11):1380–8., DOI 10.1097/JTO. 0b013e3181b9ecca NSCLC Meta-analysis Collaborative Group. Preoperative chemotherapy for non-small-cell lung cancer: a systematic review and meta-analysis of individual participant data. The Lancet, 2014, 383(9928):1561–71., DOI 10. 1016/S0140-6736(13)62159-5 Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hellmann MD, et al. Neoadjuvant PD-1 blockade in resectable lung cancer. New Engl J Med, 2018, 378(21):1976–86., DOI 10.1056/NEJMoa1716078 Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. New Engl J Med, 2022, 386(21):1973–85., DOI 10.1056/NEJMoa2202170 Wakelee H, Liberman M, Kato T, Tsuboi M, Lee SH, Gao S, et al. Perioperative pembrolizumab for early-stage non-small-cell lung cancer. N Engl J Med, 2023, 389(6):491–503., DOI 10.1056/NEJMoa2302983 Provencio M, Serna-Blasco R, Nadal E, Insa A, Garcia-Campelo MR, Casal RJ, et al. Overall survival and biomarker analysis of neoadjuvant nivolumab plus chemotherapy in operable stage IIIA non–small-cell lung cancer, NADIM phase II trial). J Clin Oncol, 2022, 40(25):2924–33., DOI 10. 1200/JCO.21.02660 Provencio M, Nadal E, Insa A, Garcia-Campelo MR, Casal-Rubio J, Domine M, et al. Neoadjuvant chemotherapy and nivolumab in resectable nonsmall- cell lung cancer, NADIM): an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol, 2020, 21(11):1413–22., DOI 10.1016/S1470-2045(20, 30453-8 Provencio M, Nadal E, Gonzalez-Larriba JL, Martinez-Marti A, Bernabe R, Bosch-Barrera J, et al. Perioperative nivolumab and chemotherapy in stage III non-small-cell lung cancer. N Engl J Med, 2023, 389(6):504–13., DOI 10. 1056/NEJMoa2215530 Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer, KEYNOTE-010): a randomised controlled trial. Lancet, 2016, 387(10027):1540–50., DOI 10.1016/S0140- 6736(15)01281-7 Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, Von Pawel J, et al. Atezolizumab versus docetaxel in patients with previously treated nonsmall- cell lung cancer, OAK): a phase 3, open-label, multicentre randomised controlled trial. The Lancet, 2017, 389(10066):255–65., DOI 10.1016/S0140- 6736(16)32517-X Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non–small-cell lung cancer. New Engl J Med, 2015, 373(17):1627–39., DOI 10.1056/NEJMoa1507643 Hong L, Lewis WE, Nilsson M, Patel S, Varghese S, Rivera MJ, et al. Limited benefit from the addition of immunotherapy to chemotherapy in TKI-refractory EGFR-mutant lung adenocarcinoma. Cancers, 2022, 14(14):3473., DOI 10.3390/ cancers14143473 Heymach JV, Harpole D, Mitsudomi T, Taube JM, Galffy G, Hochmair M, et al. Perioperative durvalumab for resectable non-small-cell lung cancer. N Engl J Med, 2023, 389(18):1672–84., DOI 10.1056/NEJMoa2304875 Lu S, Zhang W, Wu L, Wang W, Zhang P, Fang W, et al. Perioperative toripalimab plus chemotherapy for patients with resectable non-small cell lung cancer: the neotorch randomized clinical trial. JAMA, 2024, 331(3):201–11., DOI 10. 1001/jama.2023.24735 Wu Y-L, Tsuboi M, He J, John T, Grohe C, Majem M, et al. Osimertinib in resected EGFR-mutated non–small-cell lung cancer. New Engl J Med, 2020, 383(18):1711–23., DOI 10.1056/NEJMoa2027071 Herbst RS, Wu Y-L, John T, Grohe C, Majem M, Wang J, et al. Adjuvant osimertinib for resected EGFR-mutated stage IB-IIIA non–small-cell lung cancer: updated results from the phase III randomized ADAURA trial. J Clin Oncol, 2023, 41(10):1830–40., DOI 10.1200/JCO.22.02186 Akinboro O, Drezner N, Amatya A, Runyan J, Fourie-Zirkelbach J, Zhao M, et al. US food and drug administration approval summary: nivolumab plus platinum-doublet chemotherapy for the neoadjuvant treatment of patients with resectable non–small-cell lung cancer. J Clin Oncol, 2023, 41(17):3249–59., DOI 10. 1200/JCO.22.02509 Cascone T, Awad MM, Spicer JD, He J, Lu S, Sepesi B, et al. LBA1 CheckMate 77T: phase III study comparing neoadjuvant nivolumab, NIVO, plus chemotherapy, chemo, vs neoadjuvant placebo plus chemo followed by surgery and adjuvant NIVO or placebo for previously untreated, resectable stage II–IIIb NSCLC. Ann Oncol, 2023, 34:S1295., DOI 10.1016/j.annonc.2023.10.050 Wu YL, Dziadziuszko R, Ahn JS, Barlesi F, Nishio M, Lee DH, et al. Alectinib in resected ALK-positive non-small-cell lung cancer. N Engl J Med, 2024, 390(14): 1265–76., DOI 10.1056/NEJMoa2310532 Aredo JV, Urisman A, Gubens MA, Mulvey C, Allen GM, Rotow JK, et al. Phase II trial of neoadjuvant osimertinib for surgically resectable EGFR-mutated non-small cell lung cancer. J Clin Oncol, 2023, 41(16):8508., DOI 10.1200/jco.2023. 41.16_suppl.8508 Bian D, Sun L, Hu J, Duan L, Xia H, Zhu X, et al. Neoadjuvant Afatinib for stage III EGFR-mutant non-small cell lung cancer: a phase II study. Nat Commun, 2023, 14(1):4655., DOI 10.1038/s41467-023-40349-z Tsuboi M, Weder W, Escriu C, Blakely C, He J, Dacic S, et al. P03.02 neoadjuvant osimertinib with/without chemotherapy vs chemotherapy for EGFR mutated resectable NSCLC: NeoADAURA. J Thorac Oncol, 2021, 16(3):S258., DOI 10.1016/j.jtho.2021.01.375 Lee JM, Sepesi B, Toloza EM, Lin J, Pass HI, Johnson BE, et al. EP02.04- 005 phase II NAUTIKA1 study of targeted therapies in stage II-III NSCLC: preliminary data of neoadjuvant alectinib for ALK+ NSCLC. J Thorac Oncol, 2022, 17(9):S233–4., DOI 10.1016/j.jtho.2022.07.390 Leonetti A, Minari R, Boni L, Gnetti L, Bordi P, Verze M, et al. EP02.04- 001 alectinib as neoadjuvant treatment in surgically resectable stage III ALKpositive NSCLC: ALNEO phase II trial, GOIRC-01-2020). J Thorac Oncol, 2022, 17(9):S231., DOI 10.1016/j.jtho.2022.07.386 Pataer A, Kalhor N, Correa AM, Raso MG, Erasmus JJ, Kim ES, et al. Histopathologic response criteria predict survival of patients with resected lung cancer after neoadjuvant chemotherapy. J Thorac Oncol, 2012, 7(5):825–32., DOI 10. 1097/JTO.0b013e318247504a Hellmann MD, Chaft JE, William WN, Rusch V, Pisters KMW, Kalhor N, et al. Pathological response after neoadjuvant chemotherapy in resectable nonsmall- cell lung cancers: proposal for the use of major pathological response as a surrogate endpoint. Lancet Oncol, 2014, 15(1):e42–e50., DOI 10.1016/S1470- 2045(13)70334-6 Hines JB, Cameron RB, Esposito A, Kim L, Porcu L, Nuccio A, et al. Evaluation of major pathologic response and pathologic complete response as surrogate end points for survival in randomized controlled trials of neoadjuvant immune checkpoint blockade in resectable in NSCLC. J Thorac Oncol, 2024)., DOI 10.1016/j.jtho.2024.03.010 Travis WD, Dacic S, Wistuba I, Sholl L, Adusumilli P, Bubendorf L, et al. IASLC multidisciplinary recommendations for pathologic assessment of lung cancer resection specimens after neoadjuvant therapy. J Thorac Oncol, 2020, 15(5):709–40., DOI 10.1016/j.jtho.2020.01.005 Blakely CM, Weder W, Bubendorf L, He J, Majem M, Shyr Y, et al. Primary endpoints to assess the efficacy of novel therapeutic approaches in epidermal growth factor receptor-mutated, surgically resectable non-small cell lung cancer: a review. Lung Cancer, 2023, 177:59–72., DOI 10.1016/j.lungcan.2023.01.002 Rudqvist N-P, Charpentier M, Lhuillier C, Wennerberg E, Spada S, Sheridan C, et al. Immunotherapy targeting different immune compartments in combination with radiation therapy induces regression of resistant tumors. Nat Commun, 2023, 14(1):5146., DOI 10.1038/s41467-023-40844-3 Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun, 2017, 8(1):15618., DOI 10.1038/ncomms15618 Theelen WSME, Peulen HMU, Lalezari F, Van Der Noort V, De Vries JF, Aerts JGJV, et al. Effect of pembrolizumab after stereotactic body radiotherapy vs pembrolizumab alone on tumor response in patients with advanced non-small cell lung cancer: results of the PEMBRO-RT phase 2 randomized clinical trial. JAMA Oncol, 2019, 5(9):1276–82., DOI 10.1001/jamaoncol.2019.1478 Formenti SC, Rudqvist N-P, Golden E, Cooper B, Wennerberg E, Lhuillier C, et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat Med, 2018, 24(12):1845–51., DOI 10.1038/s41591-018-0232-2 Altorki NK, Walsh ZH, Melms JC, Port JL, Lee BE, Nasar A, et al. Neoadjuvant durvalumab plus radiation versus durvalumab alone in stages I–III non-small cell lung cancer: survival outcomes and molecular correlates of a randomized phase II trial. Nat Commun, 2023, 14(1):8435., DOI 10.1038/ s41467-023-44195-x Bradley JD, Nishio M, Okamoto I, Newton MD, Trani L, Shire NJ, et al. PACIFIC-2: phase 3 study of concurrent durvalumab and platinum-based chemoradiotherapy in patients with unresectable, stage III NSCLC. J Clin Oncol, 2019, 37(15):TPS8573., DOI 10.1200/jco.2019.37.15_suppl.tps8573 Lee NY, Ferris RL, Psyrri A, Haddad RI, TaharaM, Bourhis J, et al. Avelumab plus standard-of-care chemoradiotherapy versus chemoradiotherapy alone in patients with locally advanced squamous cell carcinoma of the head and neck: a randomised, double-blind, placebo-controlled, multicentre, phase 3 trial. Lancet Oncol, 2021, 22(4):450–62., DOI 10.1016/S1470-2045(20)30737-3 Cascone T, Kar G, Spicer JD, Garcia-Campelo R, Weder W, Daniel DB, et al. Neoadjuvant durvalumab alone or combined with novel immuno-oncology agents in resectable lung cancer: the phase II NeoCOAST platform trial. Cancer Discov, 2023, 13(11):2394–411., DOI 10.1158/2159-8290.CD-23-0436 Schuler M, Cuppens K, Plones T, Wiesweg M, Du Pont B, Hegedus B, et al. Neoadjuvant nivolumab with or without relatlimab in resectable non-small-cell lung cancer: a randomized phase 2 trial. Nat Med, 2024)., DOI 10.1038/s41591-024- 02965-0 Inamura K, Yokouchi Y, Kobayashi M, Ninomiya H, Sakakibara R, Subat S, et al. Association of tumor TROP2 expression with prognosis varies among lung cancer subtypes. Oncotarget, 2017, 8(17):28725–35., DOI 10.18632/oncotarget.15647 Tagawa ST, Balar AV, Petrylak DP, Kalebasty AR, Loriot Y, Flechon A, et al. TROPHY-U-01: a phase II open-label study of sacituzumab govitecan in patients with metastatic urothelial carcinoma progressing after platinum-based chemotherapy and checkpoint inhibitors. J Clin Oncol, 2021, 39(22):2474–85., DOI 10.1200/JCO.20.03489 Bardia A, Hurvitz SA, Tolaney SM, Loirat D, Punie K, Oliveira M, et al. Sacituzumab govitecan in metastatic triple-negative breast cancer. New Engl J Med, 2021, 384(16):1529–41., DOI 10.1056/NEJMoa2028485 Rugo HS, Bardia A, Marme F, Cortes J, Schmid P, Loirat D, et al. Overall survival with sacituzumab govitecan in hormone receptor-positive and human epidermal growth factor receptor 2-negative metastatic breast cancer, TROPiCS- 02): a randomised, open-label, multicentre, phase 3 trial. The Lancet, 2023, 402(10411):1423–33., DOI 10.1016/S0140-6736(23)01245-X Goto Y, Su W-C, Levy BP, Rixe O, Yang T-Y, Tolcher AW, et al. TROPION-Lung02: datopotamab deruxtecan, Dato-DXd, plus pembrolizumab, pembro, with or without platinum chemotherapy, Pt-CT, in advanced non-small cell lung cancer, aNSCLC). J Clin Oncol, 2023, 41(16): 9004., DOI 10.1200/jco.2023.41.16_suppl.9004 Ahn MJ, Lisberg A, Paz-Ares L, Cornelissen R, Girard N, Pons-Tostivint E, et al. LBA12 Datopotamab deruxtecan, Dato-DXd, vs docetaxel in previously treated advanced/metastatic, adv/met, non-small cell lung cancer, NSCLC): results of the randomized phase III study TROPION-Lung01. Ann Oncol, 2023, 34: S1305–6., DOI 10.1016/j.annonc.2023.10.061 Okamoto I, Kuyama S, Girard N, Lu S, Franke FA, Pan E, et al. 1505TiP TROPION-Lung07: a phase III trial of datopotamab deruxtecan, Dato-DXd, plus pembrolizumab, pembro, with or without platinum chemotherapy, Pt-CT, as firstline, 1L, therapy in advanced/metastatic, adv/met, non-small cell lung cancer, NSCLC, with PD. Ann Oncol, 2023, 34:S847–S8., DOI 10.1016/j.annonc.2023.09. 2536 Levy BP, Felip E, Reck M, Yang JC, Cappuzzo F, Yoneshima Y, et al. TROPION-Lung08: phase III study of datopotamab deruxtecan plus pembrolizumab as first-line therapy for advanced NSCLC. Future Oncol, 2023, 19(21):1461–72., DOI 10.2217/fon-2023-0230 Weber JS, Carlino MS, Khattak A, Meniawy T, Ansstas G, Taylor MH, et al. Individualised neoantigen therapy mRNA-4157, V940, plus pembrolizumab versus pembrolizumab monotherapy in resected melanoma, KEYNOTE-942): a randomised, phase 2b study. Lancet, 2024, 403(10427):632–44., DOI 10.1016/ S0140-6736(23)02268-7 Xia L, Mei J, Kang R, Deng S, Chen Y, Yang Y, et al. Perioperative ctDNAbased molecular residual disease detection for non–small cell lung cancer: a prospective multicenter cohort study, LUNGCA-1). Clin Cancer Res, 2022, 28(15):3308–17., DOI 10.1158/1078-0432.CCR-21-3044 Zhou C, Das Thakur M, Srivastava MK, ZouW, Xu H, Ballinger M, et al. 2O IMpower010: biomarkers of disease-free survival, DFS, in a phase III study of atezolizumab, atezo, vs best supportive care, BSC, after adjuvant chemotherapy in stage IB-IIIA NSCLC. Ann Oncol, 2021, 32:S1374., DOI 10.1016/j.annonc.2021.10.018 Assaf ZJF, Zou W, Fine AD, Socinski MA, Young A, Lipson D, et al. A longitudinal circulating tumor DNA-based model associated with survival in metastatic non-small-cell lung cancer. Nat Med, 2023, 29(4):859–68., DOI 10. 1038/s41591-023-02226-6 Yaung SJ, Fuhlbruck F, Peterson M, Zou W, Palma JF, Patil NS, et al. Clonal hematopoiesis in late-stage non-small-cell lung cancer and its impact on targeted panel next-generation sequencing. JCO Precis Oncol, 2020, 4:1271–9., DOI 10.1200/ PO.20.00046 Pascual J, Attard G, Bidard FC, Curigliano G, De Mattos-Arruda L, Diehn M, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO Precision Medicine Working Group. Ann Oncol, 2022, 33(8):750–68., DOI 10.1016/j.annonc.2022.05.520 Marcus L, Fashoyin-Aje LA, Donoghue M, Yuan M, Rodriguez L, Gallagher PS, et al. FDA approval summary: pembrolizumab for the treatment of tumor mutational burden–high solid tumors. Clin Cancer Res, 2021, 27(17):4685–9., DOI 10.1158/1078-0432.CCR-21-0327 Cascone T, Leung CH, Weissferdt A, Pataer A, Carter BW, Godoy MCB, et al. Neoadjuvant chemotherapy plus nivolumab with or without ipilimumab in operable non-small cell lung cancer: the phase 2 platform NEOSTAR trial. Nat Med, 2023, 29(3):593–604., DOI 10.1038/s41591-022-02189-0 Chaft JE, Oezkan F, Kris MG, Bunn PA, Wistuba II, Kwiatkowski DJ, et al. Neoadjuvant atezolizumab for resectable non-small cell lung cancer: an openlabel, single-arm phase II trial. Nat Med, 2022, 28(10):2155–61., DOI 10.1038/ s41591-022-01962-5 O’Brien M, Paz-Ares L, Marreaud S, Dafni U, Oselin K, Havel L, et al. Pembrolizumab versus placebo as adjuvant therapy for completely resected stage IB-IIIA non-small-cell lung cancer, PEARLS/KEYNOTE-091): an interim analysis of a randomised, triple-blind, phase 3 trial. Lancet Oncol, 2022, 23(10):1274–86., DOI 10.1016/S1470-2045(22)00518-6 Casarrubios M, Provencio M, Nadal E, Insa A, Del Rosario Garcia-Campelo M, Lazaro-Quintela M, et al. Tumor microenvironment gene expression profiles associated to complete pathological response and disease progression in resectable NSCLC patients treated with neoadjuvant chemoimmunotherapy. J ImmunoTherapy Cancer, 2022, 10(9):e005320., DOI 10.1136/jitc-2022-005320 Laza-Briviesca R, Cruz-Bermudez A, Nadal E, Insa A, Garcia-Campelo MDR, Huidobro G, et al. Blood biomarkers associated to complete pathological response on NSCLC patients treated with neoadjuvant chemoimmunotherapy included in NADIM clinical trial. Clin Translational Med, 2021, 11(7):e491., DOI 10.1002/ ctm2.491 Peters S, Cho BC, Luft A, Alatorre-Alexander J, Geater SL, Kim SW, et al. OA15.04 association between KRAS/STK11/KEAP1 mutations and outcomes in POSEIDON: durvalumab ± tremelimumab + chemotherapy in mNSCLC. J Thorac Oncol, 2022, 17(9):S39–S41., DOI 10.1016/j.jtho.2022.07.073 Liu J, Dong M, Sun X, Li W, Xing L, Yu J. Prognostic value of 18F-fdg PET/CT in surgical non-small cell lung cancer: a meta-analysis. PLOS ONE, 2016, 11(1): e0146195., DOI 10.1371/journal.pone.0146195 Sun R, Limkin EJ, Vakalopoulou M, Dercle L, Champiat S, Han SR, et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti- PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol, 2018, 19(9):1180–91., DOI 10.1016/S1470- 2045(18)30413-3 Zerunian M, Caruso D, Zucchelli A, Polici M, Capalbo C, Filetti M, et al. CT based radiomic approach on first line pembrolizumab in lung cancer. Scientific Rep, 2021, 11(1):6633., DOI 10.1038/s41598-021-86113-5 NR 2 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD APR PY 2024 VL 30 IS 2 BP 1611817 EP 1611830 DI 10.3389/pore.2024.1611817 PG 14 ER PT J AU Lieber, A Makai, A Orosz, Zs Kardos, T Isaac, JS Tornyi, I Bittner, N AF Lieber, Attila Makai, Attila Orosz, Zsuzsanna Kardos, Tamas Isaac, Joe Susil Tornyi, Ilona Bittner, Nora TI The role of immunotherapy in early-stage and metastatic NSCLC SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE NSCLC; PD-(L)1; ICI; perioperative; metastatic ID NSCLC; PD-(L)1; ICI; perioperative; metastatic AB In the past decade we have seen new advances and thus remarkable progress in the therapeutic options for non-small cell lung cancer (NSCLC). Among cytostatic therapies with new approaches in molecularly targeted therapies, we see new developments in a wide range of applications for immunotherapies. In this review we discuss the new potential modalities for the use of immune checkpoint inhibitors (ICIs) in the frontlines, including in early-stage (perioperative) and metastatic settings. The perioperative use of ICIs in both neoadjuvant and adjuvant settings may show benefits for patients. In earlystage NSCLC (from stage IIB and above) a multimodality approach is recommended as the gold standard for the treatment. After surgical resection platinum-based adjuvant chemotherapy has been the standard of care for many years. Based on the benefit of disease-free survival, the approval of adjuvant atezolizumab and adjuvant pembrolizumab was a significant breakthrough. In the metastatic setting, the use of immune checkpoint inhibitors with chemotherapy, regardless of PD-L1 expression or ICI alone (PD-L1 expression equal to or greater than 50%) also improves overall survival and progression-free survival. C1 [Lieber, Attila] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Makai, Attila] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Orosz, Zsuzsanna] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Kardos, Tamas] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Isaac, Joe Susil] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Tornyi, Ilona] University of Debrecen Medical and Health Science Center, Department of Pulmonary MedicineDebrecen, Hungary. [Bittner, Nora] National Koranyi Institute of PulmonologyBudapest, Hungary. RP Lieber, A (reprint author), University of Debrecen Medical and Health Science Center, Department of Pulmonary Medicine, Debrecen, Hungary. EM lieber.attila@med.unideb.hu CR Siegel RL, Miller KD, Wagle NS. Cancer statistics, 2023. Ca Cancer J Clin, 2023, 73:17–48., DOI 10.3322/caac.21763 Yang H, Shen K, Zhu C, Li Q, Zhao Y,Ma X. Safety and efficacy of durvalumab, MEDI4736, in various solid tumors. Drug Des Devel Ther, 2018, 12:2085–96., DOI 10.2147/DDDT.S162214 Naidoo J, Page DB, Wolchok JD. Immune checkpoint blockade. Hematol Oncol Clin North Am, 2014, 28:585–600., DOI 10.1016/j.hoc.2014.02.002 Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med, 2016, 375:1823–33., DOI 10.1056/NEJMoa1606774 Mok TS, Wu YL, Kudaba I, Kowalski DM, Cho BC, Turna HZ, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1- expressing, locally advanced or metastatic non-small-cell lung cancer, KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet, 2019, 393:1819–30., DOI 10.1016/S0140-6736(18)32409-7 Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med, 2018, 378:2078–92., DOI 10.1056/NEJMoa1801005 Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gumus M, Mazieres J, et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med, 2018, 379:2040–51., DOI 10.1056/NEJMoa1810865 Herbst RS, Giaccone G, de Marinis F, Reinmuth N, Vergnenegre A, Barrios CH, et al. Atezolizumab for first-line treatment of PD-L1-selected patients with NSCLC. N Engl J Med, 2020, 383:1328–39., DOI 10.1056/NEJMoa1917346 West H, McCleod M, Hussein M, Morabito A, Rittmeyer A, Conter HJ, et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic nonsquamous non-small-cell lung cancer, IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol, 2019, 20:924–37., DOI 10.1016/S1470- 2045(19)30167-6 Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med, 2018, 378:2288–301., DOI 10.1056/NEJMoa1716948 Sezer A, Kilickap S, Gumus M, Bondarenko I, Ozguroglu M, Gogishvili M, et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: a multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet, 2021, 397:592–604., DOI 10.1016/S0140- 6736(21)00228-2 Gogishvili M, Melkadze T, Makharadze T, Giorgadze D, Dvorkin M, Penkov K, et al. Cemiplimab plus chemotherapy versus chemotherapy alone in non-small cell lung cancer: a randomized, controlled, double-blind phase 3 trial. Nat Med, 2022, 28:2374–80., DOI 10.1038/s41591-022-01977-y Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa E, et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N Engl J Med, 2019, 381:2020–31., DOI 10.1056/NEJMoa1910231 Paz-Ares L, Ciuleanu TE, Cobo M, Schenker M, Zurawski B, Menezes J, et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer, CheckMate 9LA): an international, randomised, open-label, phase 3 trial. Lancet Oncol, 2021, 22:198–211., DOI 10.1016/ S1470-2045(20)30641-0 Johnson ML, Cho BC, Luft A, Alatorre-Alexander J, Geater SL, Laktionov K, et al. Durvalumab with or without tremelimumab in combination with chemotherapy as first-line therapy for metastatic non-small-cell lung cancer: the phase III POSEIDON study. J Clin Oncol, 2023, 41:1213–27., DOI 10.1200/JCO.22. 00975 Akinboro O, Larkins E, Pai-Scherf LH, Mathieu LN, Ren Y, Cheng J, et al. FDA approval summary: pembrolizumab, atezolizumab, and cemiplimab-rwlc as single agents for first-line treatment of advanced/metastatic PD-L1-high NSCLC. Clin Cancer Res, 2022, 28:2221–8., DOI 10.1158/1078-0432.CCR-21-3844 Reuss JE, Anagnostou V, Cottrell TR, Smith KN, Verde F, Zahurak M, et al. Neoadjuvant nivolumab plus ipilimumab in resectable non-small cell lung cancer. J Immunother Cancer, 2020, 8:e001282., DOI 10.1136/jitc-2020-001282 National Comprehensive Cancer Network. NCCN clinical practice guideline in oncology: non small cell lung cancer, version 1, 2024). Available from: https:// www.nccn.org/guidelines/guidelines-detail?category=1&id=1450, Accessed December 21, 2023). Cao W, Tang Q, Zeng J, Jin X, Zu L, Xu S. A review of biomarkers and their clinical impact in resected early-stage non-small-cell lung cancer. Cancers, 2023, 15:4561., DOI 10.3390/cancers15184561 Tang WF, Ye HY, Tang X, Su JW, Xu KM, Zhong WZM, et al. Adjuvant immunotherapy in early-stage resectable non–small cell lung cancer: a new milestone. Front Oncol, 2023, 13:1063183., DOI 10.3389/fonc.2023.1063183 Song WA, Zhou NK, Wang W, Chu XY, Liang CY, Tian XD, et al. Survival benefit of neoadjuvant chemotherapy in nonsmall cell lung cancer: an updated meta-analysis of 13 randomized control trials. J Thorac Oncol, 2010, 5:510–6., DOI 10.1097/JTO.0b013e3181cd3345 Pisters KM, Kris MG, Gralla RJ, Zaman MB, Heelan RT, Martini N. Pathologic complete response in advanced non-small-cell lung cancer following preoperative chemotherapy: implications for the design of future non-small-cell lung cancer combined modality trials. J Clin Oncol, 1993, 11:1757–62., DOI 10.1200/ JCO.1993.11.9.1757 Travis WD, Dacic S, Wistuba I, Sholl L, Adusumilli P, Bubendor L, et al. IASLC multidisciplinary recommendations for pathologic assessment of lung cancer resection specimens after neoadjuvant therapy. J Thorac Oncol, 2020, 15: 709–40., DOI 10.1016/j.jtho.2020.01.005 Pataer A, Kalhor N, Correa AM, Raso MG, Erasmus JJ, Kim ES, et al. Histopathologic response criteria predict survival of patients with resected lung cancer after neoadjuvant chemotherapy. J Thorac Oncol, 2012, 7:825–32., DOI 10. 1097/JTO.0b013e318247504a Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N Engl J Med, 2022, 386:1973–85., DOI 10.1056/NEJMoa2202170 Passaro A, Attili I, de Marinis F. Neoadjuvant chemotherapy plus immunotherapy in early-stage resectable non–small-cell lung cancer. J Clin Oncol, 2022, 40:2871–7., DOI 10.1200/JCO.22.00873 Provencio M, Serna-Blasco R, Nadal E, Insa A, Garcia-Campelo MR, Casal Rubio J, et al. Overall survival and biomarker analysis of neoadjuvant nivolumab plus chemotherapy in operable stage IIIA non–small-cell lung cancer, NADIM phase II trial). J Clin Oncol, 2022, 40:2924–33., DOI 10.1200/JCO.21.02660 Provencio Pulla M, Nadal E, Gonzalez Larriba JL, Martinez Marti A, Bernabe R, Bosch Barrera J, et al. Perioperative nivolumab and chemotherapy in stage III non–small-cell lung cancer. N Engl J Med, 2023, 389:504–13., DOI 10.1056/ NEJMoa2215530 Cascone T, Awad MM, Spicer JD, He J, Lu S, Sepesi B, et al. LBA1 CheckMate 77T: phase III study comparing neoadjuvant nivolumab, NIVO, plus chemotherapy, chemo, vs neoadjuvant placebo plus chemo followed by surgery and adjuvant NIVO or placebo for previously untreated, resectable stage II–IIIb NSCLC. Ann Oncol, 2023, 34:S1295., DOI 10.1016/j.annonc.2023.10.050 Peters S, Kim AW, Solomon B, Gandara DR, Dziadziuszko R, Brunelli A, et al. IMpower030: phase III study evaluating neoadjuvant treatment of resectable stage II-IIIB non-small cell lung cancer, NSCLC, with atezolizumab, atezo, + chemotherapy. Ann Oncol, 2019, 30:ii30., DOI 10.1093/annonc/mdz064.014 Lu S, Wu L, Zhang W, Zhang P, Wang W, Fang W, et al. Perioperative toripalimab+ platinum-doublet chemotherapy vs chemotherapy in resectable stage II/III non-small cell lung cancer, NSCLC): interim event-free survival, EFS, analysis of the phase III Neotorch study. J Clin Oncol, 2023, 41:425126., DOI 10. 1200/JCO.2023.41.36_suppl.425126 Wakelee H, Liberman M, Kato T, Tsuboi M, Lee SH, Gao S, et al. Perioperative pembrolizumab for early-stage non–small-cell lung cancer. N Engl J Med, 2023, 389:491–503., DOI 10.1056/NEJMoa2302983 Heymach JV, Mitsudomi T, Harpole D, Aperghis M, Jones S, Mann H, et al. Design and rationale for a phase III, double-blind, placebo-controlled study of neoadjuvant durvalumab + chemotherapy followed by adjuvant durvalumab for the treatment of patients with resectable stages II and III non-small-cell lung cancer: the AEGEAN trial. Clin Lung Cancer, 2022, 23:e247–e251., DOI 10.1016/j.cllc.2021. 09.010 Isaacs J, Stinchcombe TE. Neoadjuvant and adjuvant systemic therapy for early-stage non-small-cell lung cancer. Drugs, 2022, 82:855–63., DOI 10.1007/ s40265-022-01721-3 Pignon JP, Tribodet H, Scagliotti GV,Douillard JY, Shepherd FA, Stephens RJ, et al. Lung adjuvant cisplatin evaluation: a pooled analysis by the LACE Collaborative Group. J Clin Oncol, 2008, 26:3552–9., DOI 10.1200/JCO.2007.13.9030 Bakos O, Lawson C, Rouleau S, Tai LH. Combining surgery and immunotherapy: turning an immunosuppressive effect into a therapeutic opportunity. J Immunother Cancer, 2018, 6:86–11., DOI 10.1186/s40425-018- 0398-7 Stoecklein VM, Osuka A, Lederer JA. Trauma equals danger–damage control by the immune system. J Leukocyte Biol, 2012, 92:539–51., DOI 10.1189/jlb.0212072 Ogawa K, Hirai M, Katsube T, Murayama M, Hamaguchi K, Shimakawa T, et al. Suppression of cellular immunity by surgical stress. Surgery, 2000, 127: 329–36., DOI 10.1067/msy.2000.103498 Xu P, Zhang P, Sun Z, Wang Y, Chen J, Miao C. Surgical trauma induces postoperative T-cell dysfunction in lung cancer patients through the programmed death-1 pathway. Cancer Immunol Immunother CII, 2015, 64:1383–92., DOI 10. 1007/s00262-015-1740-2 Vansteenkiste J, Wauters E, Reymen B, Ackermann CJ, Peters S, De Ruysscher D. Current status of immune checkpoint inhibition in early-stage NSCLC. Ann Oncol, 2019, 30:1244–53., DOI 10.1093/annonc/mdz175 Kuchuk M, Kuchuk I, Sabri E, Hutton B, Clemons M, Wheatley-Price P. The incidence and clinical impact of bonemetastases in non-small cell lung cancer. Lung Cancer, 2015, 89:197–202., DOI 10.1016/j.lungcan.2015.04.007 Felip E, Altorki N, Zhou C, Csoszi T, Vynnychenko I, Goloborodko O, et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB–IIIA non-small-cell lung cancer, IMpower010): a randomised, multicentre, open-label, phase 3 trial. Lancet, 2021, 398:1344–57., DOI 10. 1016/S0140-6736(21)02098-5 O’Brien M, Paz-Ares L, Marreaud S, Dafni U, Oselin K, Havel L, et al. Pembrolizumab versus placebo as adjuvant therapy for completely resected stage IB–IIIA non-small-cell lung cancer, PEARLS/KEYNOTE-091): an interim analysis of a randomised, triple-blind, phase 3 trial. Lancet Oncol, 2022, 23:1274–86., DOI 10. 1016/S1470-2045(22)00518-6 Chaft JE, Dahlberg SE, Khullar OV, Edelman MJ, Simone CB, Heymach J, et al. EA5142 adjuvant nivolumab in resected lung cancers, ANVIL). J Clin Oncol, 2018, 36:TPS8581., DOI 10.1200/jco.2018.36.15_suppl.tps8581 Sands J, Mandrekar SJ, Oxnard GR, Kozono DE, Hillman SL, Dahlberg SE, et al. ALCHEMIST: adjuvant targeted therapy or immunotherapy for high-risk resected NSCLC. J Clin Oncol, 2020, 38:TPS9077., DOI 10.1200/JCO.2020.38.15_ suppl.TPS9077 Sands JM,Mandrekar SJ, Kozono D, Oxnard GR, Hillman SL,Wigle DA, et al. Integration of immunotherapy into adjuvant therapy for resected non-small-cell lung cancer: ALCHEMIST chemo-IO, ACCIO). Immunotherapy, 2021, 13:727–34., DOI 10.2217/imt-2021-0019 Peters S, Spigel D, Ahn M, Tsuboi M, Chaft J, Harpole D, et al. P03.03 MERMAID- 1: a phase III study of adjuvant durvalumab plus chemotherapy in resected NSCLC patients with MRD+ post-surgery. J Thorac Oncol, 2021, 16:S258–9., DOI 10.1016/j.jtho.2021.01.376 Spigel DR, Peters S, Ahn MJ, Tsuboi M, Chaft J, Harpole D, et al. 93TiP MERMAID-2: phase III study of durvalumab in patients with resected, stage II-III NSCLC who become MRD+ after curative-intent therapy. J Thorac Oncol, 2021, 16: S745–6., DOI 10.1016/S1556-0864(21)01935-3 Hendriks LE, Kerr KM, Menis J, Mok TS, Nestle U, Passaro A, et al. Nononcogene- addicted metastatic non-small-cell lung cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol, 2023, 34:358–76., DOI 10.1016/j.annonc.2022.12.013 Singh N, Temin S, Baker S, Blanchard E, Brahmer JR, Celano P, et al. Therapy for stage IV non–small-cell lung cancer without driver alterations: ASCO living guideline. J Clin Oncol, 2022, 40:3323–43., DOI 10.1200/JCO.22.00825 Garassino M, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, Speranza G, et al. Evaluation of TMB in KEYNOTE-189: pembrolizumab plus chemotherapy vs placebo plus chemotherapy for nonsquamous NSCLC. J Thorac Oncol, 2019, 14: S216–7., DOI 10.1016/j.jtho.2019.08.427 Mantia CM, Buchbinder EI. Immunotherapy toxicity. Hematol Oncol Clin North Am, 2019, 33:275–90., DOI 10.1016/j.hoc.2018.12.008 Hanna NH, Schneider BJ, Temin S, Baker S, Jr, Brahmer J, Ellis PM, et al. Therapy for stage IV non-small-cell lung cancer without driver alterations: ASCO and OH, CCO, joint guideline update. J Clin Oncol, 2020, 38:1608–32., DOI 10.1200/ JCO.19.03022 Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Five-year outcomes with pembrolizumab versus chemotherapy for metastatic nonsmall- cell lung cancer with PD-L1 tumor proportion score ≥ 50. J Clin Oncol, 2021, 39:2339–49., DOI 10.1200/JCO.21.00174 US National Library of Medicine. Pembrolizumab injection. United States prescribing information, 2024). Available from: https://www.accessdata.fda.gov/ drugsatfda_docs/label/2020/125514s059s064s076s083lbl.pdf, Accessed on January 29, 2024). Boyer M, Sendur MA, Rodriguez-Abreu D, Park K, Lee DH, Cicin I, et al. Pembrolizumab plus ipilimumab or placebo for metastatic non-small-cell lung cancer with PD-L1 tumor proportion score ≥ 50%: randomized, double-blind phase III KEYNOTE-598 study. J Clin Oncol, 2021, 39:2327–38., DOI 10.1200/JCO.20. 03579 Ozguroglu M, Kilickap S, Sezer A, Gumus M, Bondarenko I, Gogishvili M, et al. First-line cemiplimab monotherapy and continued cemiplimab beyond progression plus chemotherapy for advanced non-small-cell lung cancer with PD-L1 50% or more, EMPOWER-Lung 1): 35-month follow-up from a mutlicentre, open-label, randomised, phase 3 trial. Lancet Oncol., 2023, 24: 989–1001., DOI 10.1016/S1470-2045(23)00329-7 Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, SteinsM, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med, 2017, 376:2415–26., DOI 10.1056/NEJMoa1613493 Rizvi NA, Cho BC, Reinmuth N, Lee KH, Luft A, Ahn MJ, et al. Durvalumab with or without tremelimumab vs standard chemotherapy in first-line treatment of metastatic non-small cell lung cancer: the MYSTIC phase 3 randomized clinical trial. JAMA Oncol, 2020, 6:661–74., DOI 10.1001/jamaoncol.2020.0237 Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, et al. Metastatic non-small cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol, 2018, 29:v192–237., DOI 10.1093/ annonc/mdy275 Garassino MC, Gadgeel S, Speranza G, Felip E, Esteban E, Domine M, et al. Pembrolizumab plus pemetrexed and platinum in nonsquamous non-small-cell lung cancer: 5-year outcomes from the phase 3 KEYNOTE-189 study. J Clin Oncol, 2023, 41:1992–8., DOI 10.1200/JCO.22.01989 Novello S, Kowalski DM, Luft A, Gumus M, Vicente D, Mazieres J, et al. Pembrolizumab plus chemotherapy in squamous non-small-cell lung cancer: 5-year update of the phase III KEYNOTE-407 study. J Clin Oncol, 2023, 41:1999–2006., DOI 10.1200/JCO.22.01990 Makharadze T, Gogishvili M, Melkadze T, Baramidze A, Giorgadze D, Penkov K, et al. Cemiplimab plus chemotherapy versus chemotherapy alone in advanced NSCLC: 2-year follow-up from the phase 3 EMPOWER-lung 3 part 2 trial. J Thorac Oncol, 2023, 18:755–68., DOI 10.1016/j.jtho.2023.03.008 Robinson AG, Vicente D, Tafreshi A, Parra HS, Mazieres J, Cicin I, et al. 97O First-line pembrolizumab plus chemotherapy for patients with advanced squamous NSCLC: 3-year follow-up from KEYNOTE-407. J Thorac Oncol, 2021, 16:S748–9., DOI 10.1016/S1556-0864(21)01939-0 Paz-Ares L, Vicente D, Tafreshi A, Robinson A, Parra HS, Mazieres J, et al. A randomized, placebo-controlled trial of pembrolizumab plus chemotherapy in patients with metastatic squamous NSCLC: protocol-specified final analysis of KEYNOTE-407. J Thorac Oncol, 2020, 15:1657–69., DOI 10.1016/j.jtho.2020.06.015 Socinski MA, Nishio M, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, et al. IMpower150 final overall survival analyses for atezolizumab plus bevacizumab and chemotherapy in first-line metastatic nonsquamous NSCLC. J Thorac Oncol, 2021, 16:1909–24., DOI 10.1016/j.jtho.2021.07.009 Reck M, Mok TS, Nishio M, Jotte RM, Cappuzzo F, Orlandi F, et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer, IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir Med, 2019, 7:387–401., DOI 10.1016/S2213-2600(19)30084-0 Reck M, Wehler T, Orlandi F, Nogami N, Barone C, Moro-Sibilot D, et al. Safety and patient-reported outcomes of atezolizumab plus chemotherapy with or without bevacizumab versus bevacizumab plus chemotherapy in non-small-cell lung cancer. J Clin Oncol, 2020, 38:2530–42., DOI 10.1200/JCO.19.03158 Kvistborg P, Philips D, Kelderman S, Hageman L, Ottensmeier C, Joseph- Pietras D, et al. Anti-CTLA-4 therapy broadens themelanoma-reactive CD8+ T cell response. Sci transl Med, 2014, 6:254ra128., DOI 10.1126/scitranslmed.3008918 Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med, 2015, 373:23–34., DOI 10.1056/NEJMoa1504030 Brahmer JR, Lee JS, Ciuleanu TE, Caro RB, Nishio M, Urban L, et al. Five-year survival outcomes with nivolumab plus ipilimumab versus chemotherapy as firstline treatment for metastatic non-small-cell lung cancer in CheckMate 227. J Clin Oncol, 2023, 41:1200–12., DOI 10.1200/JCO.22.01503 Paz-Ares LG, Ciuleanu TE, Cobo M, Bennouna J, Schenker M, Cheng Y, et al. First-line nivolumab plus ipilimumab with chemotherapy versus chemotherapy alone for metastatic NSCLC in CheckMate 9LA: 3-year clinical update and outcomes in patients with brain metastases or select somatic mutations. J Thorac Oncol, 2023, 18:204–22., DOI 10.1016/j.jtho.2022.10.014 Carbone DP, Ciuleanu TE, Schenker M, Cobo-Dols M, Bordenave S, Juan- Vidal O, et al. First-line, 1L, nivolumab, N, + ipilimumab, I, + chemotherapy, C, vs C alone in patients, pts, with metastatic NSCLC, mNSCLC, from CheckMate 9LA: 4-y clinical update and outcomes by tumor histologic subtype, THS). J Clin Oncol, 2023, 41:LBA9023., DOI 10.1200/JCO.2023.41.17_suppl.LBA9023 Jotte R, Cappuzzo F, Vynnychenko I, Stroyakovskiy D, Rodriguez-Abreu D, Hussein M, et al. Atezolizumab in combination with carboplatin and nab-paclitaxel in advanced squamous NSCLC, IMpower131): results from a randomized phase III trial. J Thorac Oncol, 2020, 15:1351–60., DOI 10.1016/j.jtho.2020.03.028 de Castro G, Jr, Rizvi NA, Schmid P, Syrigos K, Martin C, Yamamoto N, et al. NEPTUNE: phase 3 study of first-line durvalumab plus tremelimumab in patients with metastatic NSCLC. J Thorac Oncol, 2023, 18:106–19., DOI 10.1016/j.jtho.2022. 09.223 Addeo A, Passaro A, Malapelle U, Banna GL, Subbiah V, Friedlaender A. Immunotherapy in non-small cell lung cancer harbouring driver mutations. Cancer Treat Rev, 2021, 96:102179., DOI 10.1016/j.ctrv.2021.102179 Attili I, Passaro A, Corvaja C, Aliaga PT, Del Signore E, Spitaleri G, et al. Immune checkpoint inhibitors in EGFR-mutant non-small cell lung cancer: a systematic review. Cancer Treat Rev, 2023, 119:102602., DOI 10.1016/j.ctrv.2023. 102602 Addeo A, Tabbo F, Robinson T, Buffoni L, Novello S. Precision medicine in ALK rearranged NSCLC: a rapidly evolving scenario. Crit Rev Oncol Hemat, 2018, 122:150–6., DOI 10.1016/j.critrevonc.2017.12.015 Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, et al. KRASG12C inhibition with sotorasib in advanced solid tumors. N Engl J Med, 2020, 383:1207–17., DOI 10.1056/NEJMoa1917239 Torralvo J, Friedlaender A, Achard V, Addeo A. The activity of immune checkpoint inhibition in KRAS mutated non-small cell lung cancer: a single centre experience. Cancer Genom Proteom, 2019, 16:577–82., DOI 10.21873/cgp.20160 Mazieres J, Drilon A, Lusque A, Mhanna L, Cortot AB, Mezquita L, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol, 2019, 30:1321–8., DOI 10.1093/annonc/mdz167 Frisone D, Friedlaender A, Malapelle U, Banna G, Addeo A. A BRAF new world. Crit Rev Oncol Hemat, 2020, 152:103008., DOI 10.1016/j.critrevonc.2020. 103008 Dudnik E, Peled N, Nechushtan H, Wollne,r M, Onn A, Agbarya A, et al. BRAF mutant lung cancer: programmed death ligand 1 expression, tumor mutational burden, microsatellite instability status, and response to immune check-point inhibitors. J Thoracic Oncol, 2018, 13:1128–37., DOI 10.1016/j.jtho. 2018.04.024 Negrao MV, Skoulidis F, Montesion M, Schulze K, Bara I, Shen V, et al. Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer. J Immunother Cancer, 2021, 9:e002891., DOI 10.1136/jitc-2021-002891 Cai R, Zhu H, Liu Y, Sha H, Peng W, Yin R, et al. To be, or not to be: the dilemma of immunotherapy for non-small cell lung cancer harboring various driver mutations. J Cancer Res Clin Oncol, 2023, 149:10027–40., DOI 10.1007/s00432-023- 04919-4 Sabari JK, Leonardi GC, Shu CA, Umeton R, Montecalvo J, Ni A, et al. PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients withMET exon 14 altered lung cancers. Ann Oncol, 2018, 29:2085–91., DOI 10.1093/ annonc/mdy334 Guisier F, Dubos-Arvis C, Vinas F, Doubre H, Ricordel C, Ropert S, et al. Efficacy and safety of anti–PD-1 immunotherapy in patients with advanced NSCLC with BRAF, HER2, or MET mutations or RET translocation: GFPC 01-2018. J Thorac Oncol, 2020, 15:628–36., DOI 10.1016/j.jtho.2019.12.129 Drilon A, Clark JW, Weis J, Ou SH, Camidge DR, Solomon BJ, et al. Antitumor activity of crizotinib in lung cancers harboring a MET exon 14 alteration. Nat Med, 2020, 26:47–51., DOI 10.1038/s41591-019-0716-8 Lai WC, Feldman DL, Buonocore DJ, Brzostowski EB, Rizvi H, Plodkowski AJ, et al. PD-L1 expression, tumor mutation burden and response to immune checkpoint blockade in patients with HER2-mutant lung cancers. J Clin Oncol, 2018, 36:9060., DOI 10.1200/JCO.2018.36.15_suppl.9060 Assoun S, Theou-Anton N, Nguenang M, Cazes A, Danel C, Abbar B, et al. Association of TP53 mutations with response and longer survival under immune checkpoint inhibitors in advanced non-small-cell lung cancer. Lung Cancer, 2019, 132:65–71., DOI 10.1016/j.lungcan.2019.04.005 Wang S, Jiang M, Yang Z, Huang X, Li N. The role of distinct co-mutation patterns with TP53 mutation in immunotherapy for NSCLC. Genes Dis, 2020, 9: 245–51., DOI 10.1016/j.gendis.2020.04.001 Kohno T, Ichikawa H, Totoki Y, Yasuda K, Hiramoto M, Nammo T, et al. KIF5B-RET fusions in lung adenocarcinoma. Nat Med, 2012, 18:375–7., DOI 10. 1038/nm.2644 Krzyzanowska N, Krawczyk P, Wojas-Krawczyk K, Kucharczyk T, Milanowski J. Immunotherapy in non-small-cell lung cancer patients with driver alterations: a new strategy? Cells, 2022, 11:3280., DOI 10.3390/cells11203280 Meira DD, de Castro E, Caetano MC, Casotti MC, Zetum ASS, Goncalves AFM, et al. Prognostic factors and markers in non-small cell lung cancer: recent progress and future challenges. Genes, Basel,, 2023, 14:1906., DOI 10.3390/ genes14101906 Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, et al. Nivolumab versus docetaxel in advanced squamous-cell non-smallcell lung cancer. N Engl J Med, 2015, 373:123–35., DOI 10.1056/ NEJMoa1504627 Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med, 2015, 373:1627–39., DOI 10.1056/NEJMoa1507643 Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia LJ, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced NonSmall-cell lung cancer, KEYNOTE-010): a randomised controlled trial. Lancet, 2016, 387:1540–50., DOI 10.1016/S0140-6736(15)01281-7 Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, et al. Atezolizumab versus docetaxel in patients with previously treated nonsmall- cell lung cancer, OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet, 2017, 389:255–65., DOI 10.1016/S0140-6736(16, 32517-X Herbst RS, Garon EB, Kim DW, Cho BC, Gervais R, Perez-Gracia JL, et al. Five year survival update from KEYNOTE-010: pembrolizumab versus docetaxel for previously treated, programmed death-ligand 1-positive advanced NSCLC. J Thorac Oncol, 2021, 16:1718–32., DOI 10.1016/j.jtho.2021.05.001 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611713 EP 1611729 DI 10.3389/pore.2024.1611713 PG 17 ER PT J AU Lalic, N Bojovic, M Bursac, D Bokan, D Krstic, V Kuhajda, I Parapid, B Tomic, S Sipka, A AF Lalic, Nensi Bojovic, Marko Bursac, Daliborka Bokan, Darijo Krstic, Ceriman Vesna Kuhajda, Ivan Parapid, Biljana Tomic, Sanja Sipka, Aleksandar TI The efficacy outcomes in non-small cell lung cancer patients treated with PD axis inhibitor agents - a population-based study of the Vojvodina region SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE lung carcinoma; immune checkpoint inhibitors; immunotherapy; PD-L1 receptor; pembrolizumab ID lung carcinoma; immune checkpoint inhibitors; immunotherapy; PD-L1 receptor; pembrolizumab AB Background: By 2021, the FDA approved the use of the drugs pembrolizumab and atezolizumab in the first-line treatment of patients with high positivity of programmed death ligand-1 (PD-L1) in locally advanced and metastatic nonsmall- cell-lung cancer (NSCLC). This approval was the result of statistically significant evidence from international, multicentric clinical studies that all reported increasing progression-free survival (PFS) and overall survival (OS) in these patients. Methods: In our study, we reported the demographic and clinical characteristics of 79 patients diagnosed with NSCLC with expression of PDL1 ≥50% from January 2019 to December 2022 at the Institute for Pulmonary Diseases of Vojvodina, who received pembrolizumab therapy as the first-line treatment. Patients were divided according to the histological type of lung cancer as adenocarcinoma (ADC) or squamous cell carcinoma (SCC) of the lung. In 52 of the 79 patients, PFS and in 32 of them overall survival (censored OS) was shown according to the histological type of tumor, the tumor proportion score (TPS) of PDL-1 expression, and the metastatic status within the Tumor Nodes Metastasis (TNM) disease classification. Independent factors of death outcome were shown by multivariable proportional hazard regression analysis. Results: The study included 79 patients diagnosed with NSCLC with an expression of PD-L1 ≥50%, 50 (63.3%) patients with ADC, and 29 (36.7%) patients with SCC, whose 55 (69.6%) PDL-1 expression was obtained from broncho biopsy (BB). The majority of patients, 49 (62%), had a TPS PD-L1 score of 51%–79%. Median, PFS for adenocarcinoma was 22 months and censored OS was 27 months, while for squamous cell carcinoma, median PFS was 12 months, and censored OS was 21 months. M1b disease stage, which was the most common in patients, had a PFS of 16 months and a censored OS of 18 months. Conclusion: Pembrolizumab monotherapy in patients with NSCLC in the fourth stage of the disease and with the positivity of the immune checkpoint protein TPS PD-L1 above 50% represents a safe therapy that allows a satisfactory period without disease progression and overall survival with acceptable treatment complications. C1 [Lalic, Nensi] University of Novi Sad, Medical Faculty Novi SadNovi Sad, Serbia. [Bojovic, Marko] University of Novi Sad, Medical Faculty Novi SadNovi Sad, Serbia. [Bursac, Daliborka] University of Novi Sad, Medical Faculty Novi SadNovi Sad, Serbia. [Bokan, Darijo] University of Novi Sad, Medical Faculty Novi SadNovi Sad, Serbia. [Krstic, Ceriman Vesna] University of Belgrade, Faculty of MedicineBelgrade, Serbia. [Kuhajda, Ivan] University of Novi Sad, Medical Faculty Novi SadNovi Sad, Serbia. [Parapid, Biljana] University of Belgrade, Faculty of MedicineBelgrade, Serbia. [Tomic, Sanja] University of Novi Sad, Medical Faculty Novi SadNovi Sad, Serbia. [Sipka, Aleksandar] University of Novi Sad, Medical Faculty Novi SadNovi Sad, Serbia. RP Bojovic, M (reprint author), University of Novi Sad, Medical Faculty Novi Sad, Novi Sad, Serbia. EM marko.bojovic@mf.uns.ac.rs CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Lee JB, Kim HR, Ha SJ. Immune checkpoint inhibitors in 10 Years: contribution of basic research and clinical application in cancer immunotherapy. Immune Netw, 2022, 22(1):e2., DOI 10.4110/in.2022.22.e2 Vinay DS, Ryan EP, Pawelec G, Talib WH, Stagg J, Elkord E, et al. Immune evasion in cancer: mechanistic basis and therapeutic strategies. Semin Cancer Biol, 2015, 35:S185–98., DOI 10.1016/j.semcancer.2015.03.004 Vaddepally RK, Kharel P, Pandey R, Garje R, Chandra AB. Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers, Basel,, 2020, 12(3):738., DOI 10. 3390/cancers12030738 Zam W, Ali L. Immune checkpoint inhibitors in the treatment of cancer. Curr Rev Clin Exp Pharmacol, 2022, 17(2):103–13., DOI 10.2174/ 1574884716666210325095022 Chen J, Jiang CC, Jin L, Zhang XD. Regulation of PD-L1: a novel role of pro-survival signaling in cancer. Ann Oncol, 2016, 27(3):409–16., DOI 10.1093/annonc/mdv615 Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med, 2016, 375(19):1823–33., DOI 10.1056/NEJMoa1606774 Apetoh L, Ladoire S, Coukos G, Ghiringhelli F. Combining immunotherapy and anticancer agents: the right path to achieve cancer cure? Ann Oncol, 2015, 26(9):1813–23., DOI 10.1093/annonc/mdv209 Rodriguez-Abreu D, Powell SF, Hochmair MJ, Gadgeel S, Esteban E, Felip E, et al. Pemetrexed plus platinum with or without pembrolizumab in patients with previously untreated metastatic nonsquamous NSCLC: protocol-specified final analysis from KEYNOTE-189. Ann Oncol, 2021, 32(7):881–95., DOI 10.1016/j. annonc.2021.04.008 Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gumus M, Mazieres J, et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med, 2018, 379(21):2040–51., DOI 10.1056/NEJMoa1810865 Paz-Ares L, Vicente D, Tafreshi A, Robinson A, Soto Parra H, Mazieres J, et al. A randomized, placebo-controlled trial of pembrolizumab plus chemotherapy in patients with metastatic squamous NSCLC: protocol-specified final analysis of KEYNOTE-407. J Thorac Oncol, 2020, 15(10):1657–69., DOI 10.1016/j.jtho.2020.06.015 Soldera SV, Leighl NB. Update on the treatment of metastatic squamous nonsmall cell lung cancer in new era of personalized medicine. Front Oncol, 2017, 7:50., DOI 10.3389/fonc.2017.00050 Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science, 2011, 331(6024):1565–70., DOI 10.1126/science.1203486 Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer, 2012, 12(4):252–64., DOI 10.1038/nrc3239 Hamid O, Puzanov I, Dummer R, Schachter J, Daud A, Schadendorf D, et al. Final analysis of a randomised trial comparing pembrolizumab versus investigatorchoice chemotherapy for ipilimumab-refractory advanced melanoma. Eur J Cancer, 2017, 86:37–45., DOI 10.1016/j.ejca.2017.07.022 Herbst RS, Garon EB, Kim DW, Cho BC, Perez-Gracia JL, Han JY, et al. Longterm outcomes and retreatment among patients with previously treated, programmed death-ligand 1‒positive, advanced non‒small-cell lung cancer in the KEYNOTE-010 study. J Clin Oncol, 2020, 38(14):1580–90., DOI 10.1200/JCO. 19.02446 Gadgeel S, Rodriguez-Abreu D, Speranza G, Esteban E, Felip E, Domine M, et al. Updated analysis from KEYNOTE-189: pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous nonsmall- cell lung cancer. J Clin Oncol, 2020, 38(14):1505–17., DOI 10.1200/JCO.19. 03136 Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Five-year outcomes with pembrolizumab versus chemotherapy for metastatic non–small–cell lung cancer with PD-L1 tumor proportion score ≥ 50. J Clin Oncol, 2021, 39(21):2339–49., DOI 10.1200/JCO.21.00174 de Castro G, Kudaba I, Wu YL, Lopes G, Kowalski DM, Turna HZ, et al. Fiveyear outcomes with pembrolizumab versus chemotherapy as first-line therapy in patients with non-small-cell lung cancer and programmed death ligand-1 tumor proportion score ≥ 1% in the KEYNOTE-042 study. J Clin Oncol, 2023, 41(11): 1986–91., DOI 10.1200/JCO.21.02885 Skov BG, Skov T. Paired comparison of PD-L1 expression on cytologic and histologic specimens from malignancies in the lung assessed with PD-L1 IHC 28- 8pharmDx and PD-L1 IHC 22C3pharmDx. Appl Immunohistochem Mol Morphol, 2017, 25(7):453–9., DOI 10.1097/PAI.0000000000000540 Kitazono S, Fujiwara Y, Tsuta K, Utsumi H, Kanda S, Horinouchi H, et al. Reliability of small biopsy samples compared with resected specimens for the determination of programmed death-ligand 1 expression in non-small-cell lung cancer. Clin Lung Cancer, 2015, 16(5):385–90., DOI 10.1016/j.cllc.2015.03.008 Sakata KK, Midthun DE, Mullon JJ, Kern RM, Nelson DR, Edell ES, et al. Comparison of programmed death ligand-1 immunohistochemical staining between endobronchial ultrasound transbronchial needle aspiration and resected lung cancer specimens. Chest, 2018, 154(4):827–37., DOI 10.1016/j.chest.2018.07.017 Pons-Tostivint E, Latouche A, Vaflard P, Ricci F, Loirat D, Hescot S, et al. Comparative analysis of durable responses on immune checkpoint inhibitors versus other systemic therapies: a pooled analysis of phase III trials. JCO Precis Oncol, 2019, 3(3):1–10., DOI 10.1200/PO.18.00114 Frelaut M, du Rusquec P, de Moura A, Le Tourneau C, Borcoman E. Pseudoprogression and hyperprogression as new forms of response to immunotherapy. BioDrugs, 2020, 34(4):463–76., DOI 10.1007/s40259-020-00425-y Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, et al. Non–small cell lung cancer, version 3.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Cancer Netw, 2022, 20(5):497–530., DOI 10.6004/jnccn.2022. 0025 Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, et al. NCCN Guidelines® insights: non-small cell lung cancer, version 2.2023. J Natl Compr Canc Netw, 2023, 21(4):340–50., DOI 10.6004/jnccn.2023.0020 Chiou VL, Burotto M. Pseudoprogression and immune-related response in solid tumors. J Clin Oncol, 2015, 33(31):3541–3., DOI 10.1200/JCO.2015.61.6870 Manitz J, D’Angelo SP, Apolo AB, Eggleton SP, Bajars M, Bohnsack O, et al. Comparison of tumor assessments using RECIST 1.1 and irRECIST, and association with overall survival. J Immunother Cancer, 2022, 10(2):e003302., DOI 10.1136/jitc-2021-003302 Wang Q, Gao J, Wu X. Pseudoprogression and hyperprogression after checkpoint blockade. Int Immunopharmacol, 2018, 58:125–35., DOI 10.1016/j. intimp.2018.03.018 Borcoman E, Nandikolla A, Long G, Goel S, Le Tourneau C. Patterns of response and progression to immunotherapy. Am Soc Clin Oncol Educ B, 2018, 38(38):169–78., DOI 10.1200/EDBK_200643 Tanizaki J, Hayashi H, Kimura M, Tanaka K, Takeda M, Shimizu S, et al. Report of two cases of pseudoprogression in patients with non–small cell lung cancer treated with nivolumab—including histological analysis of one case after tumor regression. Lung Cancer, 2016, 102:44–8., DOI 10.1016/j.lungcan.2016.10.014 Thompson JA, Schneider BJ, Brahmer J, Andrews S, Armand P, Bhatia S, et al. NCCN guidelines insights: management of immunotherapy-related toxicities, version 1.2020. J Natl Compr Cancer Netw, 2020, 18(3):230–41., DOI 10.6004/jnccn.2020.0012 Chen TW, Razak AR, Bedard PL, Siu LL, Hansen AR. A systematic review of immune-related adverse event reporting in clinical trials of immune checkpoint inhibitors. Ann Oncol, 2015, 26(9):1824–9., DOI 10.1093/annonc/mdv182 Zhou N, Velez MA, Bachrach B, Gukasyan J, Fares CM, Cummings AL, et al. Immune checkpoint inhibitor-induced thyroid dysfunction is a frequent event posttreatment in NSCLC. Lung Cancer, 2021, 161:34–41., DOI 10.1016/j.lungcan.2021. 08.009 Awad MM, Gadgeel SM, BorghaeiH, Patnaik A, Yang JCH, Powell SF, et al. Longterm overall survival from KEYNOTE-021 cohort G: pemetrexed and carboplatin with or without pembrolizumab as first-line therapy for advanced nonsquamous NSCLC. J Thorac Oncol, 2021, 16(1):162–8., DOI 10.1016/j.jtho.2020.09.015 Li F, Zhai S, Lv Z, Yuan L, Wang S, Jin D, et al. Effect of histology on the efficacy of immune checkpoint inhibitors in advanced non-small cell lung cancer: a systematic review and meta-analysis. Front Oncol, 2022, 12:968517., DOI 10.3389/ fonc.2022.968517 Reck M, Rodriguez–Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Updated analysis of KEYNOTE-024: pembrolizumab versus platinum-based chemotherapy for advanced non-–non-small-cell lung cancer with PD-L1 tumor proportion score of 50% or greater. J Clin Oncol, 2019, 37(7):537–46., DOI 10.1200/ JCO.18.00149 Liu LU, Xie B, Zhu W, He Q, Zhou J, Liu S, et al. High expression of PD-L1 mainly occurs in non-small cell lung cancer patients with squamous cell carcinoma or poor differentiation. Oncol Res, 2023, 31(3):275–86., DOI 10.32604/or.2023.028227 Xia H, Zhang W, Zhang Y, Shang X, Liu Y, Wang X. Liver metastases and the efficacy of immune checkpoint inhibitors in advanced lung cancer: a systematic review and meta-analysis. Front Oncol, 2022, 12:978069., DOI 10.3389/fonc. 2022.978069 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611717 EP 1611728 DI 10.3389/pore.2024.1611717 PG 12 ER PT J AU Kishikawa, S Kondo, A Yao, T Saito, T AF Kishikawa, Satsuki Kondo, Akihide Yao, Takashi Saito, Tsuyoshi TI Case report: A mesenchymal chondrosarcoma with alternative HEY1::NCOA2 fusions in the sella turcica SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE mesenchymal chondrosarcoma; intracranial mesenchymal chondrosarcoma; CNS sarcoma; HEY1::NCOA2; sella turcica ID mesenchymal chondrosarcoma; intracranial mesenchymal chondrosarcoma; CNS sarcoma; HEY1::NCOA2; sella turcica AB Introduction: Mesenchymal chondrosarcoma (MCS) is a rare subtype of chondrosarcoma that occurs at widespread anatomical locations, such as bone, soft tissue, and intracranial sites. The central nervous system (CNS) is one of the most common origins of extraosseous MCS. However, alternative HEY1::NCOA2 fusions have not been reported in this tumor. Case report: We report a case of intracranial MCS with HEY1::NCOA2 rearrangement. A 52-year-old woman presented with a 15-mm calcified mass around the sella turcica. She initially underwent transsphenoidal surgery for tumor resection and then additional resections for five local recurrences over 5 years. Histologically, the tumor was composed of small round to spindle-shaped cells admixed with well-differentiated hyaline cartilaginous islands. A hemangiopericytoma-like vascular pattern and small sinusoid-like vessels were also observed. RNA sequencing using RNA extracted from formalin-fixed paraffin-embedded samples from the last operation revealed two alternative variants of the HEY1::NCOA2 fusion: HEY1(ex4):: NCOA2 (ex13) and HEY1(ex4)::NCOA2(ex14). Both variants were confirmed as in-frame fusions using reverse transcription-polymerase chain reaction. Discussion: Cartilaginous components were often not apparent during the recurrences. In addition to the non-typical pathological finding, the correct diagnosis was hampered by the poor RNA quality of the surgical specimens and non-specific STAT6 nuclear staining. Conclusion: This is the first reported case of intracranial MCS with an alternative HEY1::NCOA2 fusion. C1 [Kishikawa, Satsuki] Juntendo University, Graduate School of Medicine, Department of Human PathologyTokyo, Japan. [Kondo, Akihide] Juntendo University School of Medicine, Department of PathologyTokyo, Japan. [Yao, Takashi] Juntendo University, Graduate School of Medicine, Department of Human PathologyTokyo, Japan. [Saito, Tsuyoshi] Juntendo University, Graduate School of Medicine, Department of Human PathologyTokyo, Japan. RP Saito, T (reprint author), Juntendo University, Graduate School of Medicine, Department of Human Pathology, Tokyo, Japan. EM tysaitou@juntendo.ac.jp CR WHO Classification of Tumours Editorial Board. Soft tissue and bone tumours WHO classification of tumours series, 5th edition, volume 3. Lyon, France): International Agency for Research on Cancer, 2020). WHO Classification of Tumours Editorial Board. Central nervous system tumours WHO classification of tumours series, 5th edition, volume 6. Lyon, France): International Agency for Research on Cancer, 2021). Bloch OG, Jian BJ, Yang I, Han SJ, Aranda D, Ahn BJ, et al. A systematic review of intracranial chondrosarcoma and survival. J Clin Neurosci, 2009, 16(12): 1547–51., DOI 10.1016/j.jocn.2009.05.003 Haider AS, Palmisciano P, Sagoo NS, Bin Alamer O, El Ahmadieh TY, Pan E, et al. Primary central nervous system sarcomas in adults: a systematic review. Clin Neurol Neurosurg, 2022, 214:107127., DOI 10.1016/j.clineuro.2022.107127 Frezza AM, Cesari M, Baumhoer D, Biau D, Bielack S, Campanacci DA, et al. Mesenchymal chondrosarcoma: prognostic factors and outcome in 113 patients. A European Musculoskeletal Oncology Society study. Eur J Cancer, 2015, 51(3): 374–81., DOI 10.1016/j.ejca.2014.11.007 Wang L, Motoi T, Khanin R, Olshen A, Mertens F, Bridge J, et al. Identification of a novel, recurrent HEY1-NCOA2 fusion inmesenchymal chondrosarcoma based on a genome-wide screen of exon-level expression data. Genes Chromosomes Cancer, 2012, 51(2):127–39., DOI 10.1002/gcc.20937 Nyquist KB, Panagopoulos I, Thorsen J, Haugom L, Gorunova L, Bjerkehagen B, et al. Whole-transcriptome sequencing identifies novel IRF2BP2-CDX1 fusion gene brought about by translocation t(1;5)(q42;q32, in mesenchymal chondrosarcoma. PLoS One, 2012, 7(11):e49705., DOI 10.1371/journal.pone.0049705 Yoshida KI, Machado I, Motoi T, Parafioriti A, Lacambra M, Ichikawa H, et al. NKX3-1 Is a useful immunohistochemical marker of EWSR1-NFATC2 sarcoma and mesenchymal chondrosarcoma. Am J Surg Pathol, 2020, 44(6):719–28., DOI 10. 1097/PAS.0000000000001441 Yoshida A,Hashimoto T, Ryo E, Yoshida KI,Motoi T, Yatabe Y, et al. Confirmation of NKX3-1 expression in EWSR1-NFATC2 sarcoma and mesenchymal chondrosarcoma using monoclonal antibody immunohistochemistry, RT-PCR, and RNA in situ hybridization. Am J Surg Pathol, 2021, 45(4):578–82., DOI 10.1097/PAS. 0000000000001627 Tlemsani C, Larousserie F, De Percin S, Audard V, Hadjadj D, Chen J, et al. Biology and management of high-grade chondrosarcoma: an update on targets and treatment options. Int J Mol Sci, 2023, 24(2):1361., DOI 10.3390/ ijms24021361 Xu B, Rooper LM, Dermawan JK, Zhang Y, Suurmeijer AJH, Dickson BC, et al. Mesenchymal chondrosarcoma of the head and neck with HEY1:: NCOA2 fusion: a clinicopathologic and molecular study of 13 cases with emphasis on diagnostic pitfalls. Genes Chromosomes Cancer, 2022, 61(11): 670–7., DOI 10.1002/gcc.23075 Huvos AG, Rosen G, Dabska M, Marcove RC. Mesenchymal chondrosarcoma. A clinicopathologic analysis of 35 patients with emphasis on treatment. Cancer, 1983, 51(7):1230–7., DOI 10.1002/1097-0142(19830401)51: 7<1230::aid-cncr2820510710>3.0.co;2-q Tsuda Y, Ogura K, Hakozaki M, Kikuta K, Ae K, Tsuchiya H, et al. Mesenchymal chondrosarcoma: a Japanese musculoskeletal oncology group, JMOG, study on 57 patients. J Surg Oncol, 2017, 115(6):760–7., DOI 10.1002/ jso.24567 Wehrli BM, Huang W, De Crombrugghe B, Ayala AG, Czerniak B. Sox9, a master regulator of chondrogenesis, distinguishes mesenchymal chondrosarcoma from other small blue round cell tumors. Hum Pathol, 2003, 34(3):263–9., DOI 10. 1053/hupa.2003.41 Wang WL,Mayordomo E, Zhang W, Hernandez VS, Tuvin D, Garcia L, et al. Detection and characterization of EWSR1/ATF1 and EWSR1/CREB1 chimeric transcripts in clear cell sarcoma, melanoma of soft parts). Mod Pathol, 2009, 22(9): 1201–9., DOI 10.1038/modpathol.2009.85 Qi W, Rosikiewicz W, Yin Z, Xu B, Jiang H, Wan S, et al. Genomic profiling identifies genes and pathways dysregulated by HEY1-NCOA2 fusion and shines a light on mesenchymal chondrosarcoma tumorigenesis. J Pathol, 2022, 257(5): 579–92., DOI 10.1002/path.5899 Safaric TP, Segovia D, Jevtic S, Ramirez D, Lyons SK, Sordella R. Patientderived xenografts and in vitro model show rationale for imatinib mesylate repurposing in HEY1-NCoA2-driven mesenchymal chondrosarcoma. Lab Invest, 2021, 102(9):1038–49., DOI 10.1038/s41374-021-00704-4 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611730 EP 1611735 DI 10.3389/pore.2024.1611730 PG 6 ER PT J AU Marton, A Veres, BK Erdodi, F Udvardy, M Illes, Rejto, L AF Marton, Adrienn Veres, Beata Katalin Erdodi, Ferenc Udvardy, Miklos Illes, Arpad Rejto, Laszlo TI The roles of phosphorylation of signaling proteins in the prognosis of acute myeloid leukemia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE acute myeloid leukemia; retinoblastoma; AKT; ERK; protein phosphorylation ID acute myeloid leukemia; retinoblastoma; AKT; ERK; protein phosphorylation AB Signaling pathways of Retinoblastoma (Rb) protein, Akt-kinase, and Erk-kinase (extracellular signal-regulated kinase) have an important role in the pathogenesis of acute myeloid leukemia. Constitutive activation of these proteins by phosphorylation contributes to cell survival by regulation of cell cycle, proliferation and proapoptotic signaling processes. According to previous data phosphorylated forms of these proteins represent a worse outcome for cancer patients. We investigated the presence of phosphorylated Rb (P-Rb), Akt (P-Akt) and Erk (P-Erk) proteins by Western blot technique using phospho-specific antibodies in bone marrow or peripheral blood samples of 69 AML patients, 36 patients with myelodysplastic syndrome (MDS) and 10 healthy volunteers. Expression level of PTEN (Phosphatase and tensin homolog) and PHLPP (PH domain and leucine-rich repeat Protein Phosphatase) phosphatases, the negative regulators of Akt kinase pathway were also examined. We tested the effect of these proteins on survival and on the correlation with known prognostic features in AML. We found 46.3% of AML patients had detectable P-Rb, 34.7% had P-Akt and 28.9% had P-Erk protein. 66.1% of patients expressing PTEN, 38.9% PHLPP, 37.2% both PTEN and PHLPP and 32.2% neither PTEN nor PHLPP phosphatases. Compared to nucleophosmin mutation (NPMc) negative samples P-Erk was significantly less in nucleophosmin mutated patients, P-Rb was significantly less in patients’ group with more than 30 G/L peripheral leukocyte count by diagnosis. PHLPP was significantly present in FAB type M5. The expression of P-Rb represented significant better overall survival (OS), while P-Akt represented significantly worse event-free survival (EFS) in unfavorable cytogenetics patients. The presence of both PHLPP and PTEN phosphatases contributes to better OS and EFS, although the differences were not statistically significant. We confirmed significant positive correlation between P-Akt and PHLPP. Assessing the phosphorylation of Rb, Akt and Erk may define a subgroup of AML patients who would benefit especially from new targeted treatment options complemented the standard chemotherapy, and it may contribute to monitoring remission, relapse or progression of AML. C1 [Marton, Adrienn] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Veres, Beata Katalin] MEDYAG Kft, Laboratory of Cellular TherapyDebrecen, Hungary. [Erdodi, Ferenc] University of Debrecen, Faculty of Medicine, Department of Medical ChemistryDebrecen, Hungary. [Udvardy, Miklos] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Rejto, Laszlo] Szabolcs-Szatmar-Bereg County Josa Andras Hospital, Department of HaematologyNyiregyhaza, Hungary. RP Marton, A (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM martonadika@gmail.com CR Steelman LS, Franklin RA, Abrams SL, Chappell W, Kempf CR, Basecke J, et al. Roles of the Ras/Raf/MEK/ERK pathway in leukemia therapy. Leukemia, 2011, 25: 1080–94., DOI 10.1038/leu.2011.66 Martelli AM, Evangelisti C, Chappell W, Abrams SL, Basecke J, Stivala F, et al. Targeting the translational apparatus to improve leukemia therapy: roles of the PI3K/PTEN/Akt/mTOR pathway. Leukemia, 2011, 25:1064–79., DOI 10.1038/leu. 2011.46 Kornblau SM, Xu HJ, Zhang W, Hu SX, Beran M, Smith TL, et al. Levels of retinoblastoma protein expression in newly diagnosed acute myelogenous leukemia. Blood, 1994, 84:256–61., DOI 10.1182/blood.v84.1.256. bloodjournal841256 Cheng X, Bennett RL, Liu X, Byrne M, Stratford May W. PKR negatively regulates leukemia progression in association with PP2A activation, Bcl-2 inhibition and increased apoptosis. Blood Cancer J, 2013, 3:e144., DOI 10.1038/bcj.2013.42 Grethe S, Porn-Ares MI. p38 MAPK regulates phosphorylation of Bad via PP2A-dependent suppression of the MEK1/2-ERK1/2 survival pathway in TNFalpha induced endothelial apoptosis. Cell. Signal., 2006, 18:531–40., DOI 10.1016/j. cellsig.2005.05.023 Nakao M, Yokota S, Iwai T, Kaneko H, Horiike S, Kashima K, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leuk Off J Leuk Soc Am Leuk Res Fund, U.K, 1996, 10:1911–8. Stirewalt DL, Radich JP. The role of FLT3 in haematopoietic malignancies. Nat Rev Cancer, 2003, 3:650–65., DOI 10.1038/nrc1169 Takahashi S. Downstream molecular pathways of FLT3 in the pathogenesis of acute myeloid leukemia: biology and therapeutic implications. J Hematol Oncol, 2011, 4:13., DOI 10.1186/1756-8722-4-13 Gary Gilliland D, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood, 2002, 100:1532–42., DOI 10.1182/blood-2002-02-0492 Mizuki M, Fenski R, Halfter H, Matsumura I, Schmidt R, Muller C, et al. Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways. Blood, 2000, 96:3907–14., DOI 10.1182/blood.v96.12.3907.h8003907_3907_3914 Zheng R, Friedman AD, Levis M, Li L, Weir EG, Small D, et al. Internal tandem duplication mutation of FLT3 blocks myeloid differentiation through suppression of C/EBPα expression. Blood, 2004, 103:1883–90., DOI 10.1182/ blood-2003-06-1978 Pearson G, Robinson F, Beers Gibson T, Xu BE, Karandikar M, Berman K, et al. Mitogen-activated protein, MAP, kinase pathways: regulation and physiological functions. Endocr Rev, 2001, 22:153–83., DOI 10.1210/edrv.22. 2.0428 Roux PP, Blenis J. ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev, 2004, 68: 320–44., DOI 10.1128/MMBR.68.2.320-344.2004 Ramos JW. The regulation of extracellular signal-regulated kinase, ERK, in mammalian cells. Int J Biochem Cel Biol., 2008, 40:2707–19., DOI 10.1016/j.biocel. 2008.04.009 Cargnello M, Roux PP. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev, 2011, 75: 50–83., DOI 10.1128/MMBR.00031-10 Omerovic J, Laude AJ, Prior IA. Ras proteins: paradigms for compartmentalised and isoform-specific signalling. Cell. Mol. Life Sci., 2007, 64: 2575–89., DOI 10.1007/s00018-007-7133-8 Fukuda M, Gotoh Y, Nishida E. Interaction of MAP kinase with MAP kinase kinase: its possible role in the control of nucleocytoplasmic transport of MAP kinase. EMBO J, 1997, 16:1901–8., DOI 10.1093/emboj/16.8.1901 Marais R, Wynne J, Treisman R. The SRF accessory protein Elk-1 contains a growth factor-regulated transcriptional activation domain. Cell, 1993, 73:381–93., DOI 10.1016/0092-8674(93)90237-k Kortenjann M, Thomae O, Shaw PE. Inhibition of v-raf-dependent c-fos expression and transformation by a kinase-defective mutant of the mitogenactivated protein kinase Erk2. Mol Cel Biol., 1994, 14:4815–24., DOI 10.1128/ mcb.14.7.4815 Sears R, Nuckolls F, Haura E, Taya Y, Tamai K, Nevins JR. Multiple Rasdependent phosphorylation pathways regulate Myc protein stability. Genes Dev, 2000, 14:2501–14., DOI 10.1101/gad.836800 Owens DM, Keyse SM. Differential regulation of MAP kinase signalling by dual-specificity protein phosphatases. Oncogene, 2007, 26:3203–13., DOI 10.1038/sj. onc.1210412 Van Kanegan MJ, Adams DG, Wadzinski BE, Strack S. Distinct protein phosphatase 2A heterotrimers modulate growth factor signaling to extracellular signal-regulated kinases and Akt. J Biol Chem, 2005, 280:36029–36., DOI 10.1074/ jbc.M506986200 Dedinszki D, Kiss A, Markasz L, Marton A, Toth E, Szekely L, et al. Inhibition of protein phosphatase-1 and -2A decreases the chemosensitivity of leukemic cells to chemotherapeutic drugs. Cel Signal., 2015, 27:363–72., DOI 10.1016/j.cellsig.2014. 11.021 Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, et al. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell, 1997, 91:231–41., DOI 10.1016/s0092-8674(00)80405-5 Cardone MH, Roy N, Stennicke HR, Salvesen GS, Franke TF, Stanbridge E, et al. Regulation of cell death protease caspase-9 by phosphorylation. Science, 1998, 282:1318–21., DOI 10.1126/science.282.5392.1318 Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell, 1999, 96:857–68., DOI 10.1016/s0092-8674(00)80595-4 Mayo LD, Donner DB. A phosphatidylinositol 3-kinase/Akt pathway promotes translocation of Mdm2 from the cytoplasm to the nucleus. Proc Natl Acad Sci U S A, 2001, 98:11598–603., DOI 10.1073/pnas.181181198 Kane LP, Shapiro VS, Stokoe D, Weiss A. Induction of NF-kappaB by the Akt/ PKB kinase. Curr Biol, 1999, 9:601–4., DOI 10.1016/s0960-9822(99)80265-6 Du K, Montminy M. CREB is a regulatory target for the protein kinase Akt/ PKB. J Biol Chem, 1998, 273:32377–9., DOI 10.1074/jbc.273.49.32377 Pugazhenthit S, Nesterova A, Sable C, Heidenreich KA, Boxer LM, Heasley LE, et al. Akt/protein kinase B up-regulates Bcl-2 expression through cAMPresponse element-binding protein. J Biol Chem, 2000, 275:10761–6., DOI 10.1074/ jbc.275.15.10761 Diehl JA, Cheng M, Roussel MF, Sherr CJ. Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization. Genes Dev, 1998, 12: 3499–511., DOI 10.1101/gad.12.22.3499 Alessi DR, Andjelkovic M, Caudwell B, Cron P, Morrice N, Cohen P, et al. Mechanism of activation of protein kinase B by insulin and IGF-1. EMBO J, 1996, 15:6541–51., DOI 10.1002/j.1460-2075.1996.tb01045.x Sarbassov DD, Guertin D. a, Ali, S. M. and Sabatini, D. M. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science, 2005, 307: 1098–101., DOI 10.1126/science.1106148 Wu X, Senechal K, Neshat MS, Whang YE, Sawyers CL. The PTEN/ MMAC1 tumor suppressor phosphatase functions as a negative regulator of the phosphoinositide 3-kinase/Akt pathway. Proc Natl Acad Sci U S A, 1998, 95: 15587–91., DOI 10.1073/pnas.95.26.15587 Stambolic V, Suzuki A, de la Pompa JL, Brothers GM, Mirtsos C, Sasaki T, et al. Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN. Cell, 1998, 95:29–39., DOI 10.1016/s0092-8674(00)81780-8 Gao T, Furnari F, Newton AC. PHLPP: a phosphatase that directly dephosphorylates Akt, promotes apoptosis, and suppresses tumor growth. Mol Cel, 2005, 18:13–24., DOI 10.1016/j.molcel.2005.03.008 Weinberg RA. The retinoblastoma protein and cell cycle control. Cell, 1995, 81:323–30., DOI 10.1016/0092-8674(95)90385-2 Pearson BE, Nasheuer HP, Wang TS. Human DNA polymerase alpha gene: sequences controlling expression in cycling and serum-stimulated cells. Mol Cel Biol, 1991, 11:2081–95., DOI 10.1128/mcb.11.4.2081 Ohtani K, DeGregori J, Nevins JR. Regulation of the cyclin E gene by transcription factor E2F1. Proc Natl Acad Sci U S A, 1995, 92:12146–50., DOI 10. 1073/pnas.92.26.12146 Morris EJ, Dyson NJ. Retinoblastoma protein partners. Adv Cancer Res, 2001, 82:1–54., DOI 10.1016/S0065–230X(01)82001-7 Brehms A, Miska EA, McCance DJ, Reid JL, Bannister AJ, Kouzarides T. Retinoblastoma protein recruits histone deacetylase to repress transcription. Nature, 1998, 391:597–601., DOI 10.1038/35404 Lukas J, Bartkova J, Bartek J. Convergence of mitogenic signalling cascades from diverse classes of receptors at the cyclin D-cyclin-dependent kinase-pRbcontrolled G1 checkpoint. Mol Cel Biol., 1996, 16:6917–25., DOI 10.1128/mcb.16. 12.6917 Chellappan SP, Hiebert S, Mudryj M, Horowitz JM, Nevins JR. The E2F transcription factor is a cellular target for the RB protein. Cell, 1991, 65:1053–61., DOI 10.1016/0092-8674(91)90557-f La Thangue NB. DRTF1/E2F: an expanding family of heterodimeric transcription factors implicated in cell-cycle control. Trends Biochem Sci, 1994, 19:108–14., DOI 10.1016/0968-0004(94)90202-x Knudsen ES, Wang JYJ. Differential regulation of retinoblastoma protein function by specific Cdk phosphorylation sites. J Biol Chem, 1996, 271:8313–20., DOI 10.1074/jbc.271.14.8313 Kornblau SM, Andreeff M, Hu SX, Xu HJ, Patel S, Theriault A, et al. Low and maximally phosphorylated levels of the retinoblastoma protein confer poor prognosis in newly diagnosed acute myelogenous leukemia: a prospective study. Clin Cancer Res, 1998, 4:1955–63. Kolupaeva V, Janssens V. PP1 and PP2A phosphatases - cooperating partners in modulating retinoblastoma protein activation. FEBS J, 2013, 280:627–43., DOI 10. 1111/j.1742-4658.2012.08511.x Venney D,Mohd-Sarip A, Mills KI. The impact of epigenetic modifications in myeloid malignancies. Int J Mol Sci, 2021, 22(9):5013., DOI 10.3390/ijms22095013 Fong CY, Morison J, Dawson MA. Epigenetics in the hematologic malignancies. Haematologica, 2014, 99:1772–83., DOI 10.3324/haematol.2013. 092007 Yang Q, Jiang W, Hou P. Emerging role of PI3K/AKT in tumor-related epigenetic regulation. Semin Cancer Biol, 2019, 59:112–24., DOI 10.1016/j. semcancer.2019.04.001 Spangle JM, Roberts TM, Zhao JJ. The emerging role of PI3K/AKT-mediated epigenetic regulation in cancer. Biochim Biophys Acta - Rev Cancer, 2017, 1868: 123–31., DOI 10.1016/j.bbcan.2017.03.002 Li J,Wang R, Hu X, Gao Y, Wang Z, Li J, et al. Activated MEK/ERK pathway drives widespread and coordinated overexpression of UHRF1 and DNMT1 in cancer cells. Sci Rep, 2019, 9:907., DOI 10.1038/s41598-018-37258-3 Suganuma T, Workman JL. MAP kinases and histone modification. J Mol Cel Biol., 2012, 4:348–50., DOI 10.1093/jmcb/mjs043 Guzman F, Fazeli Y, Khuu M, Salcido K, Singh S, Benavente CA. Retinoblastoma tumor suppressor protein roles in epigenetic regulation. Cancers, Basel)., 2020, 12:2807., DOI 10.3390/cancers12102807 Lamba JK, Cao X, Raimondi S, Downing J, Ribeiro R, Gruber TA, et al. Dna methylation clusters and their relation to cytogenetic features in pediatric aml. Cancers, Basel)., 2020, 12:3024., DOI 10.3390/cancers12103024 Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood, 2016, 127:2391–405., DOI 10.1182/blood- 2016-03-643544 Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Buchner T, et al. Diagnosis andmanagement of AML in adults: 2017 ELN recommendations from an international expert panel. Blood, 2017, 129:424–47., DOI 10.1182/blood-2016-08- 733196 Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G, Sole F, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood, 2012, 120:2454–65., DOI 10.1182/blood-2012-03-420489 Gandevia B, Tovell A. Australia’s first medical library. Med J Aust, 1964, 2: 91–2., DOI 10.5694/j.1326-5377.1973.tb128668.x Cheson BD, Bennett JM, Kopecky KJ, Buchner T,Willman CL, Estey EH, et al. Revised Recommendations of the International Working Group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia. J Clin Oncol, 2003, 21:4642–9., DOI 10.1200/JCO.2003.04.036 Selmeczi A, Udvardy M, Illes A, Telek B, Kiss A, Batar P, et al. Treatment of acute myeloid leukemia -- a single center experience, 2007-2013). Orv Hetil, 2014, 155:653–8., DOI 10.1556/oh.2014.29884 Schiller G, Lee M. Long-term outcome of high-dose cytarabine-based consolidation chemotherapy for older patients with acute myelogenous leukemia. Lymphoma, 1997, 25:111–9., DOI 10.3109/10428199709042501 Fernandez HF, Sun Z, Yao X, Litzow MR, Luger SM, Paietta EM, et al. Anthracycline dose intensification in acute myeloid leukemia. N Engl J Med, 2009, 361:1249–59., DOI 10.1056/nejmoa0904544 Sanz MA, Martin G, Rayon C, Esteve J, Gonzalez M, Diaz-Mediavilla J, et al. A modified AIDA protocol with anthracycline-based consolidation results in high antileukemic efficacy and reduced toxicity in newly diagnosed PML/RARalphapositive acute promyelocytic leukemia. PETHEMA group. Blood, 1999, 94: 3015–21. Parker JE, Pagliuca A,Mijovic A, Cullis JO, Czepulkowski B, Rassam SM, et al. Fludarabine, cytarabine, G-CSF and idarubicin, FLAG-IDA, for the treatment of poor-risk myelodysplastic syndromes and acute myeloid leukaemia. Br J Haematol, 1997, 99:939–44., DOI 10.1046/j.1365–2141.1997.4763281.x Hiddemann W, Kreutzmann H, Straif K, Ludwig WD, Mertelsmann R, Donhuijsen-Ant R, et al. High-dose cytosine arabinoside and mitoxantrone: a highly effective regimen in refractory acute myeloid leukemia. Blood, 1987, 69: 744–9., DOI 10.1182/blood.v69.3.744.bloodjournal693744 Amadori S, Arcese W, Isacchi G, Meloni G, Petti MC, Monarca B, et al. Mitoxantrone, etoposide, and intermediate-dose cytarabine: an effective and tolerable regimen for the treatment of refractory acute myeloid leukemia. J Clin Oncol, 1991, 9:1210–4., DOI 10.1200/JCO.1991.9.7.1210 Schmid C, Schleuning M, Ledderose G, Tischer J, Kolb HJ. Sequential regimen of chemotherapy, reduced-intensity conditioning for allogeneic stem-cell transplantation, and prophylactic donor lymphocyte transfusion in high-risk acute myeloid leukemia and myelodysplastic syndrome. J Clin Oncol, 2005, 23: 5675–87., DOI 10.1200/JCO.2005.07.061 Wierzbowska A, Robak T, Pluta A, Wawrzyniak E, Cebula B, Holowiecki J, et al. Cladribine combined with high doses of arabinoside cytosine, mitoxantrone, and G-CSF, CLAG-M, is a highly effective salvage regimen in patients with refractory and relapsed acute myeloid leukemia of the poor risk: a final report of the Polish Adult Leukemia Group. Eur J Haematol, 2008, 80:115–26., DOI 10. 1111/j.1600–0609.2007.00988.x ScienceDirect. Event free survival - an overview, 2022). Available from: https:// www.sciencedirect.com/topics/medicine-and-dentistry/event-free-survival Accessed 2019. Min YH, Eom JI, Cheong JW, Maeng HO, Kim JY, Jeung HK, et al. Constitutive phosphorylation of Akt/PKB protein in acute myeloid leukemia: its significance as a prognostic variable. Leukemia, 2003, 17:995–7., DOI 10.1038/sj.leu. 2402874 Ricciardi MR, McQueen T, Chism D, Milella M, Estey E, Kaldjian E, et al. Quantitative single cell determination of ERK phosphorylation and regulation in relapsed and refractory primary acute myeloid leukemia. Leukemia, 2005, 19: 1543–9., DOI 10.1038/sj.leu.2403859 Lv D, Yang H, Wang W, Xie Y, HuW, Ye M, et al. High PHLPP expression is associated with better prognosis in patients with resected lung adenocarcinoma. BMC Cancer, 2015, 15:687., DOI 10.1186/s12885-015-1711-1 Wang Z, Shu H, Wang Z, Li G, Cui J, Wu H, et al. Loss expression of PHLPP1 correlates with lymph node metastasis and exhibits a poor prognosis in patients with gastric cancer. J Surg Oncol, 2013, 108:427–32., DOI 10.1002/ jso.23419 Ono T, Nakamura S, Kazuyuki S, Takeshita K, Keiji O, Hirano I, et al. Expression analysis and prognostic significance of the PHLPP gene in acute leukemias and myelodysplastic syndromes. Blood, 2007, 110:4234., DOI 10.1182/ blood.v110.11.4234.4234 Zou XL, Liu T, Meng WT, Huang XO. Expression of tumor suppressor gene pten in patients with myelodysplastic syndrome and acute myeloid leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi, 2008, 16:1086–90. Shen Q, Chen Z, Liu XP, Xing HY, Wang M, Wang JX. Expression of PTEN mRNA in acute leukemia and its clinical significance. Zhonghua Xue Ye Xue Za Zhi, 2005, 26:493–6. Huang X, Li D, Li T, Zhao B, Chen X. Prognostic value of the expression of phosphatase and tensin homolog and CD44 in elderly patients with refractory acute myeloid leukemia. Oncol Lett, 2015, 10:103–10., DOI 10.3892/ol.2015.3189 Xie J, Qian J, Yang J, Wang S, Freeman ME, Yi Q. Critical roles of Raf/MEK/ ERK and PI3K/AKT signaling and inactivation of p38 MAP kinase in the differentiation and survival of monocyte-derived immature dendritic cells. Exp Hematol, 2005, 33:564–72., DOI 10.1016/j.exphem.2005.03.001 Miranda MB, Johnson DE. Signal transduction pathways that contribute to myeloid differentiation. Leukemia, 2007, 21:1363–77., DOI 10.1038/sj.leu.2404690 Pei S, Pollyea DA, Gustafson A, Stevens BM, Minhajuddin M, Fu R, et al. Monocytic subclones confer resistance to venetoclax-based therapy in patients with acute myeloid leukemia. Cancer Discov, 2020, 10:536–51., DOI 10.1158/2159-8290. CD-19-0710 Sancho M, Leiva D, Lucendo E, Orzaez M. Understanding MCL1: from cellular function and regulation to pharmacological inhibition. FEBS J, 2022, 289: 6209–34., DOI 10.1111/febs.16136 Liu J, Chen Y, Yu L, Yang L. Mechanisms of venetoclax resistance and solutions. Front Oncol, 2022, 12:1005659., DOI 10.3389/fonc.2022.1005659 Huang S, Jiang C, Guo H,Wang J, Liu Y, Li C, et al. Resistance mechanisms underlying venetoclax resistance in mantle cell lymphoma. Blood, 2017, 130: 2749. Alkhatabi HA, Zohny SF, Shait Mohammed MR, Choudhry H, Rehan M, Ahmad A, et al. Venetoclax-resistant MV4-11 leukemic cells activate PI3K/AKT pathway for metabolic reprogramming and redox adaptation for survival. Antioxidants, 2022, 11:461., DOI 10.3390/antiox11030461 Li X, Su Y, Hege K, Madlambayan G, Edwards H, Knight T, et al. The HDAC and PI3K dual inhibitor CUDC-907 synergistically enhances the antileukemic activity of venetoclax in preclinical models of acute myeloid leukemia. Haematologica, 2020, 106:1262–77., DOI 10.3324/haematol.2019.233445 Zagorski WA, Knudsen ES, Reed MF. Retinoblastoma deficiency increases chemosensitivity in lung cancer. Cancer Res, 2007, 67:8264–73., DOI 10.1158/ 0008–5472.CAN-06-4753 Derenzini M, Donati G, Mazzini G, Montanaro L, Vici M, Ceccarelli C, et al. Loss of retinoblastoma tumor suppressor protein makes human breast cancer cells more sensitive to antimetabolite exposure. Clin Cancer Res, 2008, 14:2199–209., DOI 10.1158/1078–0432.CCR-07-2065 Trere D, Brighenti E, Donati G, Ceccarelli C, Santini D, Taffurelli M, et al. High prevalence of retinoblastoma protein loss in triple-negative breast cancers and its association with a good prognosis in patients treated with adjuvant chemotherapy. Ann Oncol, 2009, 20:1818–23., DOI 10.1093/annonc/mdp209 Zheng Z, Chen X, Zhang Y, Ren F, Ma Y. MEK/ERK and PI3K/AKT pathway inhibitors affect the transformation of myelodysplastic syndrome into acute myeloid leukemia via H3K27me3 methylases and de-methylases. Int J Oncol, 2023, 63:140., DOI 10.3892/ijo.2023.5588 Zhang T, Xu ZJ, Gu Y, Wen XM, Ma JC, Zhang W, et al. Identification and validation of prognosis-related DLX5 methylation as an epigenetic driver inmyeloid neoplasms. Clin Transl Med, 2020, 10:e29., DOI 10.1002/ctm2.29 Zheng Z, Li L, Li G, Zhang Y, Dong C, Ren F, et al. EZH2/EHMT2 histone methyltransferases inhibit the transcription of DLX5 and promote the transformation of myelodysplastic syndrome to acute myeloid leukemia. Front Cel Dev. Biol., 2021, 9:619795., DOI 10.3389/fcell.2021.619795 Nyakern M, Tazzari PL, Finelli C, Bosi C, Follo MY, Grafone T, et al. Frequent elevation of Akt kinase phosphorylation in blood marrow and peripheral blood mononuclear cells from high-risk myelodysplastic syndrome patients. Leukemia, 2006, 20:230–8., DOI 10.1038/sj.leu.2404057 Zhang W, Konopleva M, Shi Y, McQueen T, Harris D, Ling X, et al. Mutant FLT3: a direct target of sorafenib in acute myelogenous leukemia. J Natl Cancer Inst, 2008, 100:184–98., DOI 10.1093/jnci/djm328 Konopleva MY, Dail M, Daver NG, Garcia JS, Jonas BA, Yee KWL, et al. Venetoclax and cobimetinib in relapsed/refractory AML: a phase 1b trial. Clin Lymphoma Myeloma Leuk, 2024, 24:364–74., DOI 10.1016/j.clml.2024.01.007 Weisberg E,Meng C, Case A, Sattler M, Tiv HL, Gokhale PC, et al. Evaluation of ERK as a therapeutic target in acute myelogenous leukemia. Leukemia, 2020, 34: 625–9., DOI 10.1038/s41375-019-0552-3 Navada SC, Garcia-Manero G, OdchimarReissig R, Pemmaraju N, Alvarado Y, Ohanian MN, et al. Rigosertib in combination with azacitidine in patients with myelodysplastic syndromes or acute myeloid leukemia: results of a phase 1 study. Leuk Res, 2020, 94:106369., DOI 10.1016/j.leukres.2020.106369 Konopleva MY, Walter RB, Faderl SH, Jabbour EJ, Zeng Z, Borthakur G, et al. Preclinical and early clinical evaluation of the oral AKT inhibitor, MK-2206, for the treatment of acute myelogenous leukemia. Clin Cancer Res, 2014, 20:2226–35., DOI 10.1158/1078–0432.CCR-13-1978 Bhatt VR, Wichman C, Bouska AC, Maness L, Al-Kadhimi ZS, Iqbal J, et al. Novel first-in-class drug ONC201 as a post-transplant maintenance for AML and MDS: a phase I trial in progress. Blood, 2022, 140:12918–9., DOI 10.1182/blood- 2022-156316 DiNardo CD, Pratz K, Pullarkat V, Jonas BA, Arellano M, Becker PS, et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood, 2019, 133:7–17., DOI 10.1182/blood- 2018-08-868752 DiNardo CD, Pratz KW, Letai A, Jonas BA, Wei AH, Thirman M, et al. Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study. Lancet Oncol, 2018, 19:216–28., DOI 10.1016/S1470- 2045(18)30010-X Dohner H,Wei AH, Appelbaum FR, Craddock C, DiNardo CD, Dombret H, et al. Diagnosis andmanagement of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood, 2022, 140:1345–77., DOI 10. 1182/blood.2022016867 Kantarjian H, Kadia T, DiNardo C, Daver N, Borthakur G, Jabbour E, et al. Acute myeloid leukemia: current progress and future directions. Blood Cancer J, 2021, 11:41., DOI 10.1038/s41408-021-00425-3 Premnath N, Madanat YF. Paradigm shift in the management of acute myeloid leukemia—approved options in 2023. Cancers, Basel)., 2023, 15:3002., DOI 10.3390/cancers15113002 Chatzikalil E, Roka K, Diamantopoulos PT, Rigatou E, Avgerinou G, Kattamis A, et al. Venetoclax combination treatment of acute myeloid leukemia in adolescents and young adult patients. J Clin Med, 2024, 13:2046., DOI 10.3390/ jcm13072046 Raj K, John A, Ho A, Chronis C, Khan S, Samuel J, et al. CDKN2B methylation status and isolated chromosome 7 abnormalities predict responses to treatment with 5-azacytidine. Leukemia, 2007, 21:1937–44., DOI 10.1038/sj.leu. 2404796 Greve G, Schuler J, Gruning BA, Berberich B, Stomper J, Zimmer D, et al. Decitabine induces gene derepression on monosomic chromosomes: in vitro and in vivo effects in adverse-risk cytogenetics AML. Cancer Res, 2021, 81:834–46., DOI 10. 1158/0008-5472.CAN-20-1430 Welch JS, Petti AA, Miller CA, Fronick CC, O’Laughlin M, Fulton RS, et al. TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N Engl J Med, 2016, 375:2023–36., DOI 10.1056/NEJMoa1605949 Pollyea DA, Pratz KW, Wei AH, Pullarkat V, Jonas BA, Recher C, et al. Outcomes in patients with poor-risk cytogenetics with or without TP53 mutations treated with venetoclax and azacitidine. Clin Cancer Res, 2022, 28:5272–9., DOI 10. 1158/1078-0432.CCR-22-1183 Pollyea DA, Amaya M, Strati P, Konopleva MY. Venetoclax for AML: changing the treatment paradigm. Blood Adv, 2019, 3:4326–35., DOI 10.1182/ bloodadvances.2019000937 Konopleva M, Tari AM, Estrov Z, Harris D, Xie Z, Zhao S, et al. Liposomal Bcl-2 antisense oligonucleotides enhance proliferation, sensitize acute myeloid leukemia to cytosine-arabinoside, and induce apoptosis independent of other antiapoptotic proteins. Blood, 2000, 95:3929–38., DOI 10.1182/blood.v95.12.3929. 012k01_3929_3938 Erba HP, Becker PS, Shami PJ, Grunwald MR, Flesher DL, Zhu M, et al. Phase 1b study of the MDM2 inhibitor AMG 232 with or without trametinib in relapsed/refractory acute myeloid leukemia. Blood Adv, 2019, 3:1939–49., DOI 10. 1182/bloodadvances.2019030916 Kantharidis P, El-Osta A, deSilvaM, Wall DM, Hu XF, Slater A, et al. Altered methylation of the human MDR1 promoter is associated with acquired multidrug resistance. Clin Cancer Res, 1997, 3:2025–32. Baker EK, El-Osta A. The rise of DNA methylation and the importance of chromatin on multidrug resistance in cancer. Exp Cel Res., 2003, 290:177–94., DOI 10.1016/s0014-4827(03)00342-2 Lee S, Rauch J, Kolch W. Targeting MAPK signaling in cancer: mechanisms of drug resistance and sensitivity. Int J Mol Sci, 2020, 21:1102., DOI 10.3390/ ijms21031102 Zhang J, Gu Y, Chen B. Mechanisms of drug resistance in acute myeloid leukemia. Onco Targets Ther, 2019, 12:1937–45., DOI 10.2147/OTT.S191621 Veres KB. A retinoblasztoma feherje, Rb), az Akt-kinaz, Erk-kinaz es a Bcl- 2 feherje foszforilaltsagi allapotanak vizsgalata akut myeloid leukemias es mielodiszplazias szindromas betegekben. Dissertation. Debrecen: University of Debrecen, 2014). NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611747 EP 1611765 DI 10.3389/pore.2024.1611747 PG 19 ER PT J AU Brugos, B Simon, Zs Mehes, G Illes, Pfliegler, Gy AF Brugos, Boglarka Simon, Zsofia Mehes, Gabor Illes, Arpad Pfliegler, Gyorgy TI Diagnostic challenges in patients with Castleman disease, a single center experience from Hungary SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE unicentric Castleman disease; multicentric Castleman disease; interleukin-6; siltuximab; rituximab ID unicentric Castleman disease; multicentric Castleman disease; interleukin-6; siltuximab; rituximab AB Castleman disease is a rare and atypical lymphoproliferative disorder characterized by diverse clinicalmanifestations. It has both unicentric andmulticentric forms, the latter with further subdivisions, i.e., human herpesvirus 8-associated and idiopathic forms. The diagnosis ofCastleman disease is often delayed, as it is rare, and because it shares clinical features with different autoimmune, inflammatory, and malignant lymphoproliferative disorders. The first-line treatment in unicentric form is mainly surgical, while in idiopathic Castleman disease, anti-interleukin-6 treatment is the therapy of choice. In virus-associated diseases, antiretroviral therapy and rituximab are recommended. In Hungary, only a few cases of Castleman disease have been published. This report presents our two decades of experience in the challenging diagnosis and management of this rare disorder, most properly underdiagnosed in Hungary. We provide insights into seven unicentric and five idiopathic multicentric Castleman disease cases, the latter ones especially highlighting the diagnostic and therapeutic challenges due to the variable and unique clinical features both of patients and diseases, e.g., bronchiolitis obliterans, stage IV diabetic renal failure, anti-HBc positivity, siltuximab treatment period, respectively. C1 [Brugos, Boglarka] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Simon, Zsofia] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Mehes, Gabor] University of Debrecen, Faculty of Medicine, Department of PathologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Pfliegler, Gyorgy] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. RP Brugos, B (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM brugosb@med.unideb.hu CR Fajgenbaum DC, Uldrick TS, Bagg A, Frank D, Wu D, Srkalovic G, et al. International, evidence-based consensus diagnostic criteria for HHV-8-negative/ idiopathic multicentric Castleman disease. Blood, 2017, 129(12):1646–57., DOI 10. 1182/blood-2016-10-746933 Dispenzieri A, Fajgenbaum DC. Overview of castleman disease. Blood, 2020, 135(16):1353–64., DOI 10.1182/blood.2019000931 Mukherjee S, Martin R, Sande B, Paige JS, Fajgenbaum DC. Epidemiology and treatment patterns of idiopathic multicentric Castleman disease in the era of IL- 6–directed therapy. Blood Adv, 2022, 6(2):359–67., DOI 10.1182/bloodadvances. 2021004441 Fajgenbaum DC, Shilling D. Castleman disease pathogenesis. Hematol Oncol Clin N Am, 2018, 32(1):11–21., DOI 10.1016/j.hoc.2017.09.002 Amy YL, Nabel CS, Finkelman BS, Ruth JR, Kurzrock R, vanRhee F, et al. Idiopathic multicentric Castleman’s disease: a systemic literature review. Lancet Hematol, 2016, 3(4):e163–75., DOI 10.1016/S2352-3026(16)00006-5 Uldrick TS, Polizzotto MN, Yarchoan R. Recent advances in Kaposi sarcoma herpesvirus-associated multicentric Castleman disease. Curr Opin Oncol, 2012, 24(5):495–505., DOI 10.1097/CCO.0b013e328355e0f3 Alaggio R, Amador C, Anagnostopoulos I, Attygalle AD, Araujo IBO, Berti E, et al. The 5th edition of the world health organization classification of haematolymphoid tumours: lymphoid neoplasms. Leukemia 2022; 36: 1720–48., DOI 10.1038/s41375-022-01620-2 Fajgenbaum DC, van Rhee F, Nabel CS. HHV-8 negative, idiopathic multicentric Castleman disease: novel insights into biology, pathogenesis, and therapy. Blood, 2014, 123(19):2924–33., DOI 10.1182/blood-2013-12-545087 Kojima M, Motoori T, Asano S, Nakamura S. Histological diversity of reactive and atypical proliferative lymph node lesions in systemic lupus erythematosus patients. Pathol Res Pract, 2007, 203(6):423–31., DOI 10.1016/ j.prp.2007.03.002 Kojima M, Motoori T, Nakamura S. Benign, atypical and malignant lymphoproliferative disorders in rheumatoid arthritis patients. Biomed Pharmacother, 2006, 60(10):663–72., DOI 10.1016/j.biopha.2006.09.004 Kone-Paut I, Hentgen V, Guillaume-Czitrom S, Trau T-A, Touitou I. The clinical spectrum of 94 patients carrying a single mutated MEFV gene allele. Rheumatology, Oxford,, 2009, 48(7):840–2., DOI 10.1093/rheumatology/ kep121 Stone K, Woods E, Szmania SM, Stephens OW, Garg TK, Barlogie B, et al. Interleukin-6 receptor polymorphism is prevalent in HIV-negative Castleman disease and is associated with increased soluble interleukin-6 receptor levels. PLos One, 2013, 8(1):E54610., DOI 10.1371/journal.pone.0054610 Al-Maghrabi J, Kamel-Reid S, Bailey D. Immunglobulin and T-cell receptor gene rearrangement in Castleman’s disease: molecular genetic analysis. Histopathology, 2006, 48(3):233–8., DOI 10.1111/j.1365-2559.2005.02319.x Pierson SK, Kanhai K, Bagg A, Alapat D, Lim MS, Lechowicz MJ, et al. Characterizing mortality associated with idiopathic multicentric Castleman disease. Blood, 2021, 138(Suppl. 1):1623., DOI 10.1182/blood-2021-153397 Wu D, LimMS, Jaffe ES. Pathology of Castleman disease. Hematol Oncol Clin N Am, 2018, 32:37–52., DOI 10.1016/j.hoc.2017.09.004 van Rhee F, Oksenhendler E, Srkalovic G, Voorhees P, Lim M, Dispenzieri A, et al. International, evidence-based consensus diagnostic and treatment guidelines for unicentric Castleman disease. Blood Adv, 2020, 4(23):6039–50., DOI 10.1182/ bloodadvances.2020003334 van Rhee F, Casper C, Voorhees PM, Fayad LE, Gibson D, Kanhai K, et al. Long-term safety of siltuximab in patients with idiopathic Castleman disease: a prespecified, open-label, extension analysis of two trials. Lancet Haematol, 2020, 7(3):e209–e217., DOI 10.1016/S2352-3026(19)30257-1 Sarosiek S, Shah R, Munshi NC. Review of siltuximab in the treatment of multicentric Castleman’s disease. Ther Adv Hematol, 2016, 7(6):360–6., DOI 10. 1177/2040620716653745 van Rhee F, Voorhees P, Dispenzieri A, Fossa A, Srkalovic G, Ide M, et al. International, evidence-based consensus treatment guidelines for idiopathic multicentric Castleman disease. Blood, 2018, 132(20):2115–24., DOI 10.1182/ blood-2018-07-862334 Guayboon T, Chinthammitr Y, Sukpanichnant HN, Angkasekwinai N. Human herpesvirus 8-associated multicentric Castleman disease in a patient with advanced HIV infection. Medicine, 2021, 100:49., DOI 10.1097/MD. 0000000000028077 Payer E, Miltenyi Z, Simon Z, Magyari F, Barna S, Mehes G, et al. Idiopathic multicentric Castleman disease with positive diffuse large B-cell lymphoma arising from HHV-8 positive Castleman disease. J Hematol, 2012, 1(2-3):65–9., DOI 10. 4021/jh27w Brandstadter JD, Fajgenbaum DC. How we manage idiopathic multicentric Castleman Disease. Clin Adv Hematol Oncol, 2022, 20(9):564–71. Beckham TH, Yang JC, Chau KW, Noy A, Yahalom J. Excellent outcome with surgery or radiotherapy in the management of Castleman disease including a case of oligocentric disease. Clin Lymph Myeloma Leuk, 2020, 20(10):685–9., DOI 10.1016/j. clml.2020.05.002 Gunduz E, Kirkizlar HO, Umit EG, Gulsaran SK, Ozkocaman V, Ozkalemkas F, et al. Castleman disease: a multicenter case series from Turkey. Turk J Hematol, 2022, 39:130–5., DOI 10.4274/tjh.galenos.2022.2021.0670 Kondo M, Matsuda N, Chiyotani A, Kawakami M, Konno K, Takizawa T, et al. A case of bronchobronchiolitis obliterans with Castleman’s lymphoma. Nihon Kyobu Shikkan Gakkai Zasshi, 1989, 27(6):735–41. Radzikowska E, Pawlowski J, Chabowski M, Langfort R. Constrictive bronchiolitis obliterans in patients with Castleman’s disease. Monaldi Arch Chest Di, 2005, 63(4):226–9., DOI 10.4081/monaldi.2005.624 Bustamante MS, Pierson SK, Ren Y, Bagg A, Brandstadter JD, Skralovic G, et al. Longitudinal, natural history study reveals the disease burden of idiopathic multicentric Castleman disease. Haematologica, 2024, 109(7): 2196–206., DOI 10. 3324/haematol.2023.283603 Kim HJ, Han JH, Bang CH, Park KS, Cho S-G, Yoo DS, et al. Cutaneous disorders associated with Castleman’s disease. Acta Derm Venereol, 2019, 99:984–9., DOI 10.2340/00015555-3253 Fujimoto W, Kanehiro A, Kuwamoto-Hara K, Saitoh M, Nakakita T, Amagai M, et al. Paraneoplastic pemphigus associated with Castleman’s disease and asymptomatic bronchiolitis obliterans. Eur J Dermatol, 2002, 12(4):355–9. Miltenyi Z, Toth J, Gonda A, Tar I, Remenyik E, Illes A. Successful immunomodulatory therapy in Castleman disease with paraneoplastic pemphigus vulgaris. Pathol Oncol Res, 2009, 15(3):375–81., DOI 10.1007/s12253- 008-9133-x Nishimura Y, Nishikori A, Sawada H, Czech T, Otsuka Y, Nishimura MF, et al. Idiopathic multicentric Castleman disease with positive antiphospholipid antibody: atypical or undiagnosed autoimmune disease. J Clin Exp Hematop, 2022, 62(2):99–105., DOI 10.3960/jslrt.21038 Chrispal A, Vasuki Z, Thomas EM, Boorugu HK. Mixed connective tissue disorder and Castleman’s disease. J Assoc Physicians India, 2010, 58:515–7. Fujimoto S, Kawabata H, Kurose N, Kawanami-Iwao H, Sakai T, Kawanami T, et al. Sjogren’s syndrome manifesting as clinicopathological features of TAFRO syndrome: a case report. Medicine, Baltimore,, 2017, 96(5):e9220., DOI 10.1097/MD. 0000000000009220 Demirkan FG, Dogan S, Ucar AK, Sonmez HE, Ayaz NA. Systemic lupus erythematosus complicated with Castleman disease: a case-based review. Rheumatol Int, 2021, 41(2):475–9., DOI 10.1007/s00296-020-04684-4 Sitenga J, Aird G, Ahmed A, Silberstein PT. Impact of siltuximab on patientrelated outcomes in multicentric Castleman’s disease. Patients Relat Outcome Meas, 2018, 9:35–41., DOI 10.2147/PROM.S140011 Carbone A, Borok M, Damania B, Gloghini A, Polizzotto MN, Jayanthan RK, et al. Castleman disease. Nat Rev Dis Primers, 2021, 7(1):84., DOI 10.1038/s41572- 021-00317-7 Karoui AE, Vuiblet V, Dion D, Izzedine H, Guitard J, Frimat L, et al. Renal involvement in Castleman disease. Nephrol Dial Transpl, 2011, 26:599–609., DOI 10. 1093/ndt/gfq427 Leurs A, Gnemmi V, Lionet A, Renaud L, Gibier J-B, Copin M-C, et al. Renal pathologic findings in TAFRO syndrome: is there a continuum between thrombotic microangiopathy and membranoproliferative glomerulonephritis? A case report and literature review. Front Immunol, 2019, 10:1489., DOI 10.3389/fimmu.2019. 01489 Lang E, Sande B, Brodkin S, vanRhee F. Idiopathic multicentric Castleman disease treated with siltuximab for 15 years: a case report. Ther Adv Hematol, 2022, 13:20406207221082552–6., DOI 10.1177/20406207221082552 van Rhee F,Wong RS,Munshi N, Rossi J-F, Ke X-Y, Fossa A, et al. Siltuximab for multicentric Castleman’s disease: a randomised, double-blind, placebo-controlled trial. Lancet Oncol, 2014, 15:966–74., DOI 10.1016/S1470-2045(14)70319-5 Muzes G, Sipos F, Csomor J, Sreter L. Multicentric Castleman’s disease: a challenging diagnosis. Pathol Oncol Res, 2013, 19:345–51., DOI 10.1007/s12253-013- 9619-z NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611785 EP 1611794 DI 10.3389/pore.2024.1611785 PG 10 ER PT J AU Cakir, U Balogh, P Ferenczik, A Brodszky, V Krenacs, T Karpati, S Sardy, M Hollo, P Fabian, M AF Cakir, Ugur Balogh, Petra Ferenczik, Aniko Brodszky, Valentin Krenacs, Tibor Karpati, Sarolta Sardy, Miklos Hollo, Peter Fabian, Melinda TI G protein-coupled estrogen receptor 1 and collagen XVII endodomain expression in human cutaneous melanomas: can they serve as prognostic factors? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE estrogen receptor; collagen xvii; prognostic factors; immunohistochemistry; prognosis ID estrogen receptor; collagen xvii; prognostic factors; immunohistochemistry; prognosis AB Melanoma incidence is increasing globally. Although novel therapies have improved the survival of primary melanoma patients over the past decade, the overall survival rate for metastatic melanoma remains low. In addition to traditional prognostic factors such as Breslow thickness, ulceration, and mitotic rate, novel genetic and molecular markers have been investigated. In our study, we analyzed the expression of G-protein coupled estrogen receptor 1 (GPER1) and the endodomain of collagen XVII (COL17) in relation to clinicopathological factors in primary cutaneous melanomas with known lymph node status in both sexes, using immunohistochemistry. We found, that GPER1 expression correlated with favorable clinicopathological factors, including lower Breslow thickness, lower mitotic rate and absence of ulceration. In contrast, COL17 expression was associated with poor prognostic features, such as higher tumor thickness, higher mitotic rate, presence of ulceration and presence of regression. Melanomas positive for both GPER1 and COL17 had significantly lower mean Breslow thickness and mitotic rate compared to cases positive for COL17 only. Our data indicate that GPER1 and COL17 proteins may be of potential prognostic value in primary cutaneous melanomas. C1 [Cakir, Ugur] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Balogh, Petra] University Hospitals Birmingham, Queen Elizabeth Hospital, Cellular Pathology DepartmentBirmingham, UK. [Ferenczik, Aniko] Corvinus University of Budapest, Doctoral School of Economics, Business and InformaticsBudapest, Hungary. [Brodszky, Valentin] Corvinus University of Budapest, Institute of Social and Political Sciences, Department of Health PolicyBudapest, Hungary. [Krenacs, Tibor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Karpati, Sarolta] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Sardy, Miklos] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Hollo, Peter] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. [Fabian, Melinda] Semmelweis University, Department of Dermatology, Venereology and DermatooncologyBudapest, Hungary. RP Cakir, U (reprint author), Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Budapest, Hungary. EM cakir.ugur@semmelweis.hu CR Erdmann F, Lortet-Tieulent J, Schuz J, Zeeb H, Greinert R, Breitbart EW, et al. International trends in the incidence of malignant melanoma 1953–2008--are recent generations at higher or lower risk? Int J Cancer, 2013, 132:385–400., DOI 10.1002/ijc.27616 Sacchetto L, Zanetti R, Comber H, Bouchardy C, Brewster DH, Broganelli P, et al. Trends in incidence of thick, thin and in situ melanoma in Europe. Eur J Cancer Oxf Engl, 2018, 92:108–18., DOI 10.1016/j.ejca.2017.12.024 Current Staging. Prognostic factors in melanoma- ClinicalKey. Available from: https://www.clinicalkey.com/#!/content/playContent/1-s2.0-S1055320714001240? returnurl=https:%2F%2Flinkinghub.elsevier.com%2Fretrieve%2Fpii% 2FS1055320714001240%3Fshowall%3Dtrue&referrer=https:%2F%2Fpubmed. ncbi.nlm.nih.gov%2F, Accessed March 9, 2021). Bellenghi M, Puglisi R, Pontecorvi G, De Feo A, Care A, Mattia G. Sex and gender disparities in melanoma. Cancers, 2020, 12:1819., DOI 10.3390/cancers12071819 Smalley KS. Why do women with melanoma do better than men? eLife, 2018, 7:e33511., DOI 10.7554/eLife.33511 Fabian M, Rencz F, Krenacs T, Brodszky V, Harsing J, Nemeth K, et al. Expression of G protein-coupled oestrogen receptor in melanoma and in pregnancy-associated melanoma. J Eur Acad Dermatol Venereol JEADV, 2017, 31:1453–61., DOI 10.1111/jdv.14304 Dika E, Patrizi A, Lambertini M, Manuelpillai N, Fiorentino M, Altimari A, et al. Estrogen receptors and melanoma: a Review. Cells, 2019, 8:1463., DOI 10.3390/ cells8111463 Zhou JH, Kim KB, Myers JN, Fox PS, Ning J, Bassett RL, et al. Immunohistochemical expression of hormone receptors in melanoma of pregnant women, nonpregnant women, and men. Am J Dermatopathol, 2014, 36:74–9., DOI 10.1097/DAD.0b013e3182914c64 Dika E, Fanti PA, Vaccari S, Capizzi E, Degiovanni A, Gobbi A, et al. Oestrogen and progesterone receptors in melanoma and nevi: an immunohistochemical study. Eur J Dermatol EJD, 2017, 27:254–9., DOI 10.1684/ejd.2017.3019 de Giorgi V, Mavilia C, Massi D, Sestini S, Grazzini M, Brandi ML, et al. The role of estrogens in melanoma and skin cancer. Carcinogenesis, 2009, 30:720., DOI 10.1093/carcin/bgp025 Lee HJ,Wall B, Chen S. G-protein-coupled receptors and melanoma. Pigment Cell Melanoma Res, 2008, 21:415–28., DOI 10.1111/j.1755-148X.2008.00478.x Grundker C, Emons G. The role of gonadotropin-releasing hormone in cancer cell proliferation and metastasis. Front Endocrinol, 2017, 8:187., DOI 10.3389/fendo. 2017.00187 Jala VR, Radde BN, Haribabu B, Klinge CM. Enhanced expression of G-protein coupled estrogen receptor, GPER/GPR30, in lung cancer. BMC Cancer, 2012, 12:624., DOI 10.1186/1471-2407-12-624 Meng R, Qin Q, Xiong Y, Wang Y, Zheng J, Zhao Y, et al. NHERF1, a novel GPER associated protein, increases stability and activation of GPER in ER-positive breast cancer. Oncotarget, 2016, 7:54983–97., DOI 10.18632/oncotarget.10713 Jacenik D, Cygankiewicz AI, Krajewska WM. The G protein-coupled estrogen receptor as a modulator of neoplastic transformation. Mol Cell Endocrinol, 2016, 429:10–8., DOI 10.1016/j.mce.2016.04.011 Liu Q, Chen Z, Jiang G, Zhou Y, Yang X, Huang H, et al. Epigenetic down regulation of G protein-coupled estrogen receptor, GPER, functions as a tumor suppressor in colorectal cancer. Mol Cancer, 2017, 16:87., DOI 10.1186/s12943-017- 0654-3 Lau K-M, Ma FM-T, Xia JT, Chan QKY, Ng CF, To KF. Activation of GPR30 stimulates GTP-binding of Gαi1 protein to sustain activation of Erk1/2 in inhibition of prostate cancer cell growth and modulates metastatic properties. Exp Cell Res, 2017, 350:199–209., DOI 10.1016/j.yexcr.2016.11.022 Chimento A, Sirianni R, Casaburi I, Zolea F, Rizza P, Avena P, et al. GPER agonist G-1 decreases adrenocortical carcinoma, ACC, cell growth in vitro and in vivo. Oncotarget, 2015, 6:19190–203., DOI 10.18632/oncotarget.4241 Talia M, De Francesco EM, Rigiracciolo DC, Muoio MG, Muglia L, Belfiore A, et al. The G protein-coupled estrogen receptor, GPER, expression correlates with pro-metastatic pathways in ER-negative breast cancer: a bioinformatics analysis. Cells, 2020, 9:622., DOI 10.3390/cells9030622 Zhang D, Wang J, Chen H, Yan S. Cytoplasmic G protein-coupled estrogen receptor 1 as a prognostic indicator of breast cancer: a meta-analysis. Technol Cancer Res Treat, 2022, 21:15330338221131664., DOI 10.1177/15330338221131664 Martin SG, Lebot MN, Sukkarn B, Ball G, Green AR, Rakha EA, et al. Low expression of G protein-coupled oestrogen receptor 1, GPER, is associated with adverse survival of breast cancer patients. Oncotarget, 2018, 9:25946–56., DOI 10. 18632/oncotarget.25408 Lv X, Hua G, He C, Davis JS, Wang C. Abstract B82: G-protein coupled estrogen receptor, GPER, agonist G-1 inhibits growth of human granulosa cell tumor cells via blocking microtubule assembly. Clin Cancer Res, 2014, 19:B82., DOI 10.1158/1078-0432.OVCA13-B82 Glatthaar H, Katto J, Vogt T, Mahlknecht U. Estrogen receptor alpha, ESR1, single-nucleotide polymorphisms, SNPs, affect malignant melanoma susceptibility and disease course. Genet Epigenetics, 2016, 8:1–6., DOI 10.4137/GEG.S31264 Fuentes N, Silveyra P. Estrogen receptor signaling mechanisms. Adv Protein Chem Struct Biol, 2019, 116:135–70., DOI 10.1016/bs.apcsb.2019.01.001 de Giorgi V, Mavilia C, Massi D, Gozzini A, Aragona P, Tanini A, et al. Estrogen receptor expression in cutaneous melanoma: a real-time reverse transcriptase-polymerase chain reaction and immunohistochemical study. Arch Dermatol, 2009, 145:30–6., DOI 10.1001/archdermatol.2008.537 Marzagalli M, Montagnani Marelli M, Casati L, Fontana F, Moretti RM, Limonta P. Estrogen receptor β in melanoma: from molecular insights to potential clinical utility. Front Endocrinol, 2016, 7:140., DOI 10.3389/fendo.2016.00140 Xu S, Yu S, Dong D, Lee LTO. G protein-coupled estrogen receptor: a potential therapeutic target in cancer. Front Endocrinol, 2019, 10:725., DOI 10.3389/ fendo.2019.00725 Kvingedal AM, Smeland EB. A novel putative G-protein-coupled receptor expressed in lung, heart and lymphoid tissue. FEBS Lett, 1997, 407:59–62., DOI 10. 1016/s0014-5793(97)00278-0 Feng Y, Gregor P. Cloning of a novel member of the G protein-coupled receptor family related to peptide receptors. Biochem Biophys Res Commun, 1997, 231:651–4., DOI 10.1006/bbrc.1997.6161 Pepermans RA, Sharma G, Prossnitz ER. G protein-coupled estrogen receptor in cancer and stromal cells: functions and novel therapeutic perspectives. Cells, 2021, 10:672., DOI 10.3390/cells10030672 Sjostrom M, Hartman L, Fornander T, Grabau D, Malmstrom P, Nordenskjold B, et al. Abstract P1-08-12: G protein-coupled estrogen receptor in the plasma membrane is prognostic in early breast cancer. Cancer Res, 2013, 73: P1–08-12-P1-08-12., DOI 10.1158/0008-5472.SABCS13-P1-08-12 Yang F, Shao Z-M. Double-edged role of G protein-coupled estrogen receptor 1 in breast cancer prognosis: an analysis of 167 breast cancer samples and online data sets. Oncotargets Ther, 2016, 9:6407–15., DOI 10.2147/OTT.S111846 Natale CA, Duperret EK, Zhang J, Sadeghi R, Dahal A, O’Brien KT, et al. Sex steroids regulate skin pigmentation through nonclassical membrane-bound receptors. eLife, 2016, 5:e15104., DOI 10.7554/eLife.15104 Nicolaidou E, Katsambas AD. Pigmentation disorders: hyperpigmentation and hypopigmentation. Clin Dermatol, 2014, 32:66–72., DOI 10.1016/j.clindermatol. 2013.05.026 SunM, Xie H-F, Tang Y, Lin SQ, Li JM, Sun SN, et al. G protein-coupled estrogen receptor enhances melanogenesis via cAMP-protein kinase, PKA, by upregulating microphthalmia-related transcription factor-tyrosinase in melanoma. J Steroid Biochem Mol Biol, 2017, 165:236–46., DOI 10.1016/j. jsbmb.2016.06.012 Ribeiro MPC, Santos AE, Custodio JBA. The activation of the G proteincoupled estrogen receptor, GPER, inhibits the proliferation of mouse melanoma K1735-M2 cells. Chem Biol Interact, 2017, 277:176–84., DOI 10.1016/j.cbi.2017. 09.017 Natale CA, Li J, Zhang J, Dahal A, Dentchev T, Stanger BZ, et al. Activation of G protein-coupled estrogen receptor signaling inhibits melanoma and improves response to immune checkpoint blockade. eLife, 2018, 7:e31770., DOI 10.7554/eLife. 31770 Mallipeddi R, Keane FM, McGrath JA, Mayou BJ, Eady RAJ. Increased risk of squamous cell carcinoma in junctional epidermolysis bullosa. J Eur Acad Dermatol Venereol JEADV, 2004, 18:521–6., DOI 10.1111/j.1468-3083.2004.00968.x Jones VA, Patel PM, Gibson FT, Cordova A, Amber KT. The role of collagen XVII in cancer: squamous cell carcinoma and beyond. Front Oncol, 2020, 10:352., DOI 10.3389/fonc.2020.00352 Hammers CM, Stanley JR. Mechanisms of disease: pemphigus and bullous pemphigoid. Annu Rev Pathol, 2016, 11:175–97., DOI 10.1146/annurev-pathol- 012615-044313 Koster J, Geerts D, Favre B, Borradori L, Sonnenberg A. Analysis of the interactions between BP180, BP230, plectin and the integrin alpha6beta4 important for hemidesmosome assembly. J Cell Sci, 2003, 116:387–99., DOI 10.1242/jcs.00241 Stelkovics E, Korom I, Marczinovits I, Molnar J, Rasky K, Raso E, et al. Collagen XVII/BP180 protein expression in squamous cell carcinoma of the skin detected with novel monoclonal antibodies in archived tissues using tissue microarrays and digital microscopy. Appl Immunohistochem Mol Morphol AIMM, 2008, 16:433–41., DOI 10.1097/PAI.0b013e318162f8aa Moilanen JM, Loffek S, Kokkonen N, Salo S, Vayrynen JP, Hurskainen T, et al. Significant role of collagen XVII and integrin β4 in migration and invasion of the less aggressive squamous cell carcinoma cells. Sci Rep, 2017, 7:45057., DOI 10.1038/ srep45057 Meng X, Matsumoto F, Mori T, Miura N, Ino Y, Onidani K, et al. BP180 is a prognostic factor in head and neck squamous cell carcinoma. Anticancer Res, 2021, 41:1089–99., DOI 10.21873/anticanres.14867 Moilanen JM, Kokkonen N, Loffek S, Vayrynen JP, Syvaniemi E, Hurskainen T, et al. Collagen XVII expression correlates with the invasion and metastasis of colorectal cancer. Hum Pathol, 2015, 46:434–42., DOI 10.1016/j.humpath.2014. 11.020 Laval S, Laklai H, Fanjul M, Pucelle M, Laurell H, Billon-Gales A, et al. Dual roles of hemidesmosomal proteins in the pancreatic epithelium: the phosphoinositide 3-kinase decides. Oncogene, 2014, 33:1934–44., DOI 10.1038/ onc.2013.146 Papay J, Krenacs T, Moldvay J, Stelkovics E, Furak J, Molnar B, et al. Immunophenotypic profiling of nonsmall cell lung cancer progression using the tissue microarray approach. Appl Immunohistochem Mol Morphol AIMM, 2007, 15:19–30., DOI 10.1097/01.pai.0000213143.32030.f5 Bergstraesser LM, Srinivasan G, Jones JC, Stahl S,Weitzman SA. Expression of hemidesmosomes and component proteins is lost by invasive breast cancer cells. Am J Pathol, 1995, 147:1823–39. Krenacs T, Kiszner G, Stelkovics E, Balla P, Teleki I, Nemeth I, et al. Collagen XVII is expressed in malignant but not in benign melanocytic tumors and it can mediate antibody induced melanoma apoptosis. Histochem Cell Biol, 2012, 138: 653–67., DOI 10.1007/s00418-012-0981-9 Blondeaux E, Massarotti C, Fontana V, Poggio F, Arecco L, Fregatti P, et al. The PREgnancy and FERtility, PREFER, study investigating the need for ovarian function and/or fertility preservation strategies in premenopausal women with early breast cancer. Front Oncol, 2021, 11:690320., DOI 10.3389/fonc.2021.690320 Keung EZ, Gershenwald JE. The eighth edition American Joint Committee on Cancer, AJCC, melanoma staging system: implications for melanoma treatment and care. Expert Rev Anticancer Ther, 2018, 18:775–84., DOI 10.1080/14737140. 2018.1489246 Hirsch FR, Varella-GarciaM, Bunn PA, Di Maria MV, Veve R, Bremmes RM, et al. Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol Off J Am Soc Clin Oncol, 2003, 21:3798–807., DOI 10.1200/ JCO.2003.11.069 McCarty KS, Miller LS, Cox EB, Konrath J. Estrogen receptor analyses. Correlation of biochemical and immunohistochemical methods using monoclonal antireceptor antibodies. Arch Pathol Lab Med, 1985, 109:716–21. McCarty KS, Szabo E, Flowers JL, Cox EB, Leight GS, Miller L, et al. Use of a monoclonal anti-estrogen receptor antibody in the immunohistochemical evaluation of human tumors. Cancer Res, 1986, 46:4244s–8s. Ruengwanichayakun P. Histochemical scoring assessment, H-score). Asian Arch Pathol, 2021). Available from: https://www.asianarchpath.com/view/152, Accessed July 20, 2024). DennisMK, Burai R, Ramesh C, Petrie WK, Alcon SN, Nayak TK, et al. In vivo effects of a GPR30 antagonist. Nat Chem Biol, 2009, 5:421–7., DOI 10.1038/ nchembio.168 Wang C, Prossnitz ER, Roy SK. Expression of G protein-coupled receptor 30 in the hamster ovary: differential regulation by gonadotropins and steroid hormones. Endocrinology, 2007, 148:4853–64., DOI 10.1210/en.2007-0727 Peyton C, Thomas P. Involvement of epidermal growth factor receptor signaling in estrogen inhibition of oocyte maturation mediated through the G protein-coupled estrogen receptor, Gper, in zebrafish, Danio rerio). Biol Reprod, 2011, 85:42–50., DOI 10.1095/biolreprod.110.088765 Gao F, Ma X, Ostmann AB, Das SK. GPR30 activation opposes estrogendependent uterine growth via inhibition of stromal ERK1/2 and estrogen receptor alpha, ERα, phosphorylation signals. Endocrinology, 2011, 152:1434–47., DOI 10. 1210/en.2010-1368 Patel VH, Chen J, Ramanjaneya M, Karteris E, Zachariades E, Thomas P, et al. G-protein coupled estrogen receptor 1 expression in rat and human heart: protective role during ischaemic stress. Int J Mol Med, 2010, 26:193–9., DOI 10. 3892/ijmm_00000452 Uea M, Salehi SA, Windahl S, Gomez MF, Sward K, Daszkiewicz-Nilsson J, et al. Deletion of the G protein-coupled receptor 30 impairs glucose tolerance, reduces bone growth, increases blood pressure, and eliminates estradiol-stimulated insulin release in female mice. Endocrinology, 2009, 150:687–98., DOI 10.1210/en. 2008-0623 Haas E, Bhattacharya I, Brailoiu E, Damjanovic M, Brailoiu GC, Gao X, et al. Regulatory role of G protein-coupled estrogen receptor for vascular function and obesity. Circ Res, 2009, 104:288–91., DOI 10.1161/CIRCRESAHA.108.190892 Wang C, Dehghani B, Magrisso IJ, Rick EA, Bonhomme E, Cody DB, et al. GPR30 contributes to estrogen-induced thymic atrophy. Mol Endocrinol Baltim Md, 2008, 22:636–48., DOI 10.1210/me.2007-0359 Brunsing RL, Prossnitz ER. Induction of interleukin-10 in the T helper type 17 effector population by the G protein coupled estrogen receptor, GPER, agonist G-1. Immunology, 2011, 134:93–106., DOI 10.1111/j.1365-2567.2011.03471.x Yates MA, Li Y, Chlebeck PJ, Offner H. GPR30, but not estrogen receptoralpha, is crucial in the treatment of experimental autoimmune encephalomyelitis by oral ethinyl estradiol. BMC Immunol, 2010, 11:20., DOI 10.1186/1471-2172-11-20 Jenei-Lanzl Z, Straub RH, Dienstknecht T, Huber M, Hager M, Grassel S, et al. Estradiol inhibits chondrogenic differentiation of mesenchymal stem cells via nonclassic signaling. Arthritis Rheum, 2010, 62:1088–96., DOI 10.1002/art.27328 Filardo EJ, Graeber CT, Quinn JA, Resnick MB, Giri D, DeLellis RA, et al. Distribution of GPR30, a seven membrane–spanning estrogen receptor, in primary breast cancer and its association with clinicopathologic determinants of tumor progression. Clin Cancer Res, 2006, 12:6359–66., DOI 10.1158/1078- 0432.CCR-06-0860 Smith HO, Arias-Pulido H, Kuo DY, Howard T, Qualls CR, Lee SJ, et al. GPR30 predicts poor survival for ovarian cancer. Gynecol Oncol, 2009, 114:465–71., DOI 10.1016/j.ygyno.2009.05.015 Jung J. Role of G Protein-Coupled estrogen receptor in cancer progression. Toxicol Res, 2019, 35:209–14., DOI 10.5487/TR.2019.35.3.209 Liu H, Yan Y, Wen H, Jiang X, Cao X, Zhang G, et al. A novel estrogen receptor GPER mediates proliferation induced by 17β-estradiol and selective GPER agonist G-1 in estrogen receptor α, ERα)-negative ovarian cancer cells. Cell Biol Int, 2014, 38:631–8., DOI 10.1002/cbin.10243 Yan Y, Liu H, Wen H, Jiang X, Cao X, Zhang G, et al. The novel estrogen receptor GPER regulates the migration and invasion of ovarian cancer cells. Mol Cell Biochem, 2013, 378:1–7., DOI 10.1007/s11010-013-1579-9 Smith HO, Leslie KK, Singh M, Qualls CR, Revankar CM, Joste NE, et al. GPR30: a novel indicator of poor survival for endometrial carcinoma. Am J Obstet Gynecol, 2007, 196:386.e1–9. discussion 386.e9-11., DOI 10.1016/j.ajog.2007.01.004 Petrie WK, Dennis MK, Hu C, Dai D, Arterburn JB, Smith HO, et al. G protein-coupled estrogen receptor-selective ligands modulate endometrial tumor growth. Obstet Gynecol Int, 2013, 2013:472720., DOI 10.1155/2013/472720 Avino S, De Marco P, Cirillo F, Santolla MF, De Francesco EM, Perri MG, et al. Stimulatory actions of IGF-I are mediated by IGF-IR cross-talk with GPER and DDR1 in mesothelioma and lung cancer cells. Oncotarget, 2016, 7:52710–28., DOI 10.18632/oncotarget.10348 Liu C, Liao Y, Fan S, Fu X, Xiong J, Zhou S, et al. G-Protein-Coupled estrogen receptor antagonist G15 decreases estrogen-induced development of non-small cell lung cancer. Oncol Res, 2019, 27:283–92., DOI 10.3727/096504017X15035795904677 Torres-Lopez L, Olivas-Aguirre M, Villatoro-Gomez K, Dobrovinskaya O. The G-protein-coupled estrogen receptor agonist G-1 inhibits proliferation and causes apoptosis in leukemia cell lines of T lineage. Front Cell Dev Biol, 2022, 10: 811479., DOI 10.3389/fcell.2022.811479 Zhou L, Yu T, Yang F, Han J, Zuo B, Huang L, et al. G protein-coupled estrogen receptor agonist G-1 inhibits mantle cell lymphoma growth in preclinical models. Front Oncol, 2021, 11:668617., DOI 10.3389/fonc.2021.668617 Natale CA, Li J, Pitarresi JR, Norgard RJ, Dentchev T, Capell BC, et al. Pharmacologic activation of the G protein-coupled estrogen receptor inhibits pancreatic ductal adenocarcinoma. Cell Mol Gastroenterol Hepatol, 2020, 10: 868–80., DOI 10.1016/j.jcmgh.2020.04.016 Hirtz A, Lebourdais N, Rech F, Bailly Y, Vaginay A, Smail-Tabbone M, et al. GPER agonist G-1 disrupts tubulin dynamics and potentiates temozolomide to impair glioblastoma cell proliferation. Cells, 2021, 10:3438., DOI 10.3390/ cells10123438 Schuler-Toprak S, Skrzypczak M, Ignatov T, Ortmann O, Treeck O. G protein-coupled estrogen receptor 1, GPER-1, and agonist G-1 inhibit growth of ovarian cancer cells by activation of anti-tumoral transcriptome responses: impact of GPER-1 mRNA on survival. J Cancer Res Clin Oncol, 2020, 146: 3175–88., DOI 10.1007/s00432-020-03333-4 Lee S-J, KimTW, Park GL, Hwang YS, Cho HJ, Kim J-T, et al. G proteincoupled estrogen receptor-1 agonist induces chemotherapeutic effect via ER stress signaling in gastric cancer. BMB Rep, 2019, 52:647–52., DOI 10.5483/ BMBRep.2019.52.11.007 Ambrosini G, Natale CA, Musi E, Garyantes T, Schwartz GK. The GPER agonist LNS8801 induces mitotic arrest and apoptosis in uveal melanoma cells. Cancer Res Commun, 2023, 3:540–7., DOI 10.1158/2767-9764.CRC-22-0399 Linnaeus Therapeutics, Inc. Study assessing MTD, safety, tolerability, PK and anti-tumor effects of LNS8801alone and with pembrolizumab. clinicaltrials.gov, 2022). Available from: https://clinicaltrials.gov/study/NCT04130516?intr= LNS8801&rank=1, Accessed August 18, 2022). Liu C-C, Lin J-H, Hsu T-W, Hsu JW, Chang JW, Su K, et al. Collagen XVII/ laminin-5 activates epithelial-to-mesenchymal transition and is associated with poor prognosis in lung cancer. Oncotarget, 2018, 9:1656–72., DOI 10.18632/oncotarget.11208 Yamada T, Endo R, Tsukagoshi K, Fujita S, Honda K, Kinoshita M, et al. Aberrant expression of a hemidesmosomal protein, bullous pemphigoid antigen 2, in human squamous cell carcinoma. Lab Investig J Tech Methods Pathol, 1996, 75: 589–600. Thangavelu PU, Krenacs T, Dray E, Duijf PHG. In epithelial cancers, aberrant COL17A1 promoter methylation predicts its misexpression and increased invasion. Clin Epigenetics, 2016, 8:120., DOI 10.1186/s13148-016-0290-6 Tamas L, Szentkuti G, Eros M, Danos K, Brauswetter D, Szende B, et al. Differential biomarker expression in head and neck cancer correlates with anatomical localization. Pathol Oncol Res POR, 2011, 17:721–7., DOI 10.1007/ s12253-011-9376-9 Parikka M, Kainulainen T, Tasanen K, Vaananen A, Bruckner-Tuderman L, Salo T. Alterations of collagen XVII expression during transformation of oral epithelium to dysplasia and carcinoma. J Histochem Cytochem Off J Histochem Soc, 2003, 51:921–9., DOI 10.1177/002215540305100707 ParikkaM, Nissinen L, Kainulainen T, Bruckner-Tuderman L, Salo T, Heino J, et al. Collagen XVII promotes integrin-mediated squamous cell carcinoma transmigration--a novel role for alphaIIb integrin and tirofiban. Exp Cell Res, 2006, 312:1431–8., DOI 10.1016/j.yexcr.2006.01.015 Hwang B-J, Zhang Y, Brozowski JM, Liu Z, Burette S, Lough K, et al. The dysfunction of BP180/collagen XVII in keratinocytes promotes melanoma progression. Oncogene, 2019, 38:7491–503., DOI 10.1038/s41388- 019-0961-9 Tong D, Tanaka M, Eguchi H, Okazaki Y, Muramatsu M, Arai T. COL17A1 germline variant p.Ser1029Ala and mucosal malignant melanoma: an autopsy study. Mol Clin Oncol, 2022, 16:32., DOI 10.3892/ mco.2021.2465 LothongM, Sakares W, Rojsitthisak P, Tanikawa C,Matsuda K, Yodsurang V. Collagen XVII inhibits breast cancer cell proliferation and growth through deactivation of the AKT/mTOR signaling pathway. PLoS One, 2021, 16: e0255179., DOI 10.1371/journal.pone.0255179 Fujiwara S, Terai Y, Kawaguchi H, Takai M, Yoo S, Tanaka Y, et al. GPR30 regulates the EGFR-Akt cascade and predicts lower survival in patients with ovarian cancer. J Ovarian Res, 2012, 5:35., DOI 10.1186/1757-2215-5-35 Chen J, Zhao R, Wang Y, Xiao H, Lin W, Diao M, et al. G protein-coupled estrogen receptor activates PI3K/AKT/mTOR signaling to suppress ferroptosis via SREBP1/SCD1-mediated lipogenesis. Mol Med, 2024, 30:28., DOI 10.1186/s10020- 023-00763-x Wang Y, Peng Z, Meng R, Tao T, Wang Q, Zhao C, et al. NHERF1 inhibits proliferation of triple-negative breast cancer cells by suppressing GPER signaling. Oncol Rep, 2017, 38:221–8., DOI 10.3892/or.2017.5649 Hou X, Zhao M, Wang T, Zhang G. Upregulation of estrogen receptor mediates migration, invasion and proliferation of endometrial carcinoma cells by regulating the PI3K/AKT/mTOR pathway. Oncol Rep, 2014, 31:1175–82., DOI 10. 3892/or.2013.2944 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611809 EP 1611820 DI 10.3389/pore.2024.1611809 PG 12 ER PT J AU Plander, M Kanyasi, M Szendrei, T Skrapits, J Timar, B AF Plander, Mark Kanyasi, Maria Szendrei, Tamas Skrapits, Judit Timar, Botond TI Flow cytometry in the differential diagnosis of myelodysplastic neoplasm with low blasts and cytopenia of other causes SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE flow cytometry; myelodysplastic neoplasms; low blasts; differential diagnosis; accuracy ID flow cytometry; myelodysplastic neoplasms; low blasts; differential diagnosis; accuracy AB Background: Myelodysplastic neoplasms (MDS) are characterized by cytopenia, morphologic dysplasia, and genetic abnormalities. Multiparameter flow cytometry (FCM) is recommended in the diagnostic work-up of suspected MDS, but alone is not sufficient to establish the diagnosis. Our aim was to investigate the diagnostic power of FCM in a heterogeneous population of patients with cytopenia, excluding cases with increased blast count. Methods: We analyzed bone marrow samples from 179 patients with cytopenia (58 MDS, 121 non-MDS) using a standardized 8-color FCM method. We evaluated the sensitivity, specificity, and accuracy of several simple diagnostic approaches, including Ogata score, extended Ogata score, the WHO and ELN iMDSFlow recommended “3 aberrations in two cell compartments method,” and the combination of the Ogata score and “3 aberrations in two cell compartments method.” The patients were followed until the diagnosis was confirmed, with a median follow-up of 2 months (range 0.2–27). Results: The combination of Ogata score and “3 aberrations in two cell compartments method” achieved the highest diagnostic accuracy (78%) with sensitivity and specificity 61% and 86%, respectively. When using only the “3 aberrations in two cell compartments method,” the accuracy was 77% with a sensitivity of 72% and a specificity of 79%. The most frequently observed etiologies among the false positive cases were substrate deficiencies, inflammation/infection, or toxic effects. MDS can be excluded in all these cases after a thorough clinical evaluation and a relatively short follow-up. Conclusion: FCM remains an important but supplementary part in an integrated diagnostic process of MDS with low blasts. The combination of the Ogata score and the “3 aberrations in two cell compartments method” slightly improves accuracy compared to the detection of “3 aberrations in twocell compartments method” alone. C1 [Plander, Mark] Vas County Markusovszky Hospital, Department of HematologySzombathely, Hungary. [Kanyasi, Maria] Markusovszky County Hospital, Central LaboratorySzombathely, Hungary. [Szendrei, Tamas] Vas County Markusovszky Hospital, Department of HematologySzombathely, Hungary. [Skrapits, Judit] Markusovszky County Hospital, Central LaboratorySzombathely, Hungary. [Timar, Botond] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Plander, M (reprint author), Vas County Markusovszky Hospital, Department of Hematology, Szombathely, Hungary. EM planderm@yahoo.com CR Khoury JD, Solary E, Abla O, Akkari Y, Alaggio R, Apperley JF, et al. The 5th edition of the world Health organization classification of haematolymphoid tumours: myeloid and histiocytic/Dendritic Neoplasms. Leukemia, 2022, 36: 1703–19., DOI 10.1038/s41375-022-01613-1 Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le BeauMM, et al. The 2016 revision to the World Health Organization classification ofmyeloid neoplasms and acute leukemia. Blood, 2016, 127(20):2391–405., DOI 10.1182/blood-2016-03- 643544 Senent L, Arenillas L, Luno E, Ruiz JC, Sanz G, Florensa L. Reproducibility of the world Health organization 2008 criteria for myelodysplastic syndromes. Haematologica, 2013, 984:568–75., DOI 10.3324/haematol.2012.071449 Font P, Loscertales J, Benavente C, Bermejo A, Callejas M, Garcia-Alonso L, et al. Inter-observer variance with the diagnosis of myelodysplastic syndromes, MDS, following the 2008 WHO classification. Ann Hematol, 2013, 921:19–24., DOI 10.1007/s00277-012-1565-4 Font P, Loscertales J, Soto C, Ricard P, Novas CM, Martin-Clavero E, et al. Interobserver variance in myelodysplastic syndromes with less than 5 % bone marrow blasts: unilineage vs. multilineage dysplasia and reproducibility of the threshold of 2 % blasts. Ann Hematol, 2015, 94(4):565–73., DOI 10.1007/s00277-014-2252-4 Della Porta MG, Travaglino E, Boveri E, Ponzoni M, Malcovati L, Papaemmanuil E, et al. Rete Ematologica Lombarda, REL, Clinical Network Minimal morphological criteria for defining bone marrow dysplasia: basis for clinical implementation of WHO classification of myelodysplastic syndromes. Leukemia, 2015, 291:66–75., DOI 10.1038/leu.2014.161 Haase D, Germing U, Schanz J, Pfeilstocker M, Nosslinger T, Hildebrandt B, et al. New insights into the prognostic impact of the karyotype in MDS and correlation with subtypes: evidence from a core dataset of 2124 patients. Blood, 2007, 110(13):4385–95., DOI 10.1182/blood-2007-03-082404 Kern W, Westers TM, Bellos F, Bene MC, Bettelheim P, Brodersen LE, et al. Multicenter prospective evaluation of diagnostic potential of flow cytometric aberrancies in myelodysplastic syndromes by the ELN iMDS flow working group. Cytometry B Clin Cytom, 2023, 104(1):51–65., DOI 10.1002/cyto.b.22105 Malcovati L, Hellstrom-Lindberg E, Bowen D, Ades L, Cermak J, del Canizo C, et al. Diagnosis and treatment of primary myelodysplastic syndromes in adults: recommendations from the European LeukemiaNet. Blood, 2013, 122(17): 2943–64., DOI 10.1182/blood-2013-03-492884 Porwit A, van de Loosdrecht AA, Bettelheim P, Eidenschink Brodersen L, Burbury K, Cremers E, et al. Revisiting guidelines for integration of flow cytometry results in the WHO classification of myelodysplastic syndromes—proposal from the International/European LeukemiaNet Working Group for Flow Cytometry in MDS. Leukemia, 2014, 28:1793–8., DOI 10.1038/leu.2014.191 Della Porta MG, Picone C, Pascutto C, Malcovati L, Tamura H, Handa H, et al. Multicenter validation of a reproducible flow cytometric score for the diagnosis of low-grade myelodysplastic syndromes: results of a European LeukemiaNET study. Haematologica, 2012, 97(8):1209–17., DOI 10.3324/haematol.2011.048421 Bardet V, Wagner-Ballon O, Guy J, Morvan C, Debord C, Trimoreau F, et al. Multicentric study underlining the interest of adding CD5, CD7 and CD56 expression assessment to the flow cytometric Ogata score in myelodysplastic syndromes and myelodysplastic/myeloproliferative neoplasms. Haematologica, 2015, 100(4):472–8., DOI 10.3324/haematol.2014.112755 Mathis S, Chapuis N, Debord C, Rouquette A, Radford-Weiss I, Park S, et al. Flow cytometric detection of dyserythropoiesis: a sensitive and powerful diagnostic tool for myelodysplastic syndromes. Leukemia, 2013, 27:1981–7., DOI 10.1038/leu.2013.178 Westers TM, Cremers EMP, Oelschlaegel U, Johansson U, Bettelheim P, Matarraz S, et al. Immunophenotypic analysis of erythroid dysplasia in myelodysplastic syndromes. A report from the IMDSFlow working group. Haematologica, 2017, 102:308–19., DOI 10.3324/haematol.2016.147835 Wells DA, Benesch M, Loken MR, Vallejo C, Myerson D, Leisenring WM, et al. Myeloid andmonocytic dyspoiesis as determined by flow cytometric scoring in myelodysplastic syndrome correlates with the IPSS and with outcome after hematopoietic stem cell transplantation. Blood, 2003, 102:394–403., DOI 10.1182/ blood-2002-09-2768 Cremers EMP, Westers TM, Alhan C, Cali C, Visser-Wisselaar HA, Chitu DA, et al. Implementation of erythroid lineage analysis by flow cytometry in diagnostic models for myelodysplastic syndromes. Haematologica, 2017, 102:320–6., DOI 10. 3324/haematol.2016.147843 Westers TM, Ireland R, Kern W, Alhan C, Balleisen JS, Bettelheim P, et al. Standardization of flow cytometry in myelodysplastic syndromes: a report from an international consortium and the European LeukemiaNet Working Group. Leukemia, 2012, 26(7):1730–41., DOI 10.1038/leu.2012.30 Valent P, Orazi A, Steensma DP, Ebert BL, Haase D, Malcovati L, et al. Proposed minimal diagnostic criteria for myelodysplastic syndromes, MDS, and potential pre-MDS conditions. Oncotarget, 2017, 8(43):73483–500., DOI 10.18632/oncotarget.19008 Karai B, Bedekovics J, Miltenyi Z, Gergely L, Szerafin L, Ujfalusi A, et al. A single-tube flow cytometric procedure for enhancing the diagnosis and prognostic classification of patients with myelodysplastic syndromes. Int J Lab Hematol, 2017, 39(6):577–84., DOI 10.1111/ijlh.12700 Davydova YO, Parovichnikova EN, Galtseva IV, Kokhno AV, Dvirnyk VN, Kovrigina AM, et al. Diagnostic significance of flow cytometry scales in diagnostics of myelodysplastic syndromes. Cytometry B Clin Cytom, 2021, 100(3):312–21., DOI 10.1002/cyto.b.21965 Oelschlaegel U, Oelschlaeger L, von Bonin M, Kramer M, Sockel K, Mohr B, et al. Comparison of five diagnostic flow cytometry scores in patients with myelodysplastic syndromes: diagnostic power and prognostic impact. Cytometry B Clin Cytom, 2023, 104:141–50., DOI 10.1002/cyto.b.22030 Grille E, Iastrebner M, Lorand-Metze I, van der Veldene VHJ, Ikoma MRV, Vidal-Senmache G, et al. Flow cytometry diagnosis in myelodysplastic syndrome: current practice in Latin America and comparison with other regions of the world. Leuk Res, 2019, 79:69–74., DOI 10.1016/j.leukres.2019.01.009 Matarraz S, Lopez A, Barrena S, Fernandez C, Jensen E, Flores-Montero J, et al. Bone marrow cells from myelodysplastic syndromes show altered immunophenotypic profiles that may contribute to the diagnosis and prognostic stratification of the disease: a pilot study on a series of 56 patients. Cytometry, 2010, 78B:154–68., DOI 10.1002/cyto.b.20513 Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G, Sole F, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood, 2012, 120(12):2454–65., DOI 10.1182/blood-2012-03-420489 Rajab A, Porwit A Screening bone marrow samples for abnormal lymphoid populations and myelodysplasia-related features with one 10-color 14-antibody screening tube. Cytometry, 2015, 88(4):253–60., DOI 10.1002/cyto.b.21233 Valiathan R, AshmanM, Asthana D. Effects of ageing on the immune system: infants to elderly. Scand J Immunol, 2016, 83(4):255–66., DOI 10.1111/sji.12413 Cain D, Kondo M, Chen H, Kelsoe G. Effects of acute and chronic inflammation on B-cell development and differentiation. J Invest Dermatol, 2009, 129(2):266–77., DOI 10.1038/jid.2008.286 Wascher TC, Hermann J, Brezinschek HP, Brezinschek R, Wilders-Truschnig M, Rainer F, et al. Cell-type specific response of peripheral blood lymphocytes to methotrexate in the treatment of rheumatoid arthritis. Clin Investig, 1994, 72(7): 535–40., DOI 10.1007/BF00207484 Stetler-Stevenson M, Arthur DC, Jabbour N, Xie XY, Molldrem J, Barrett AJ, et al. Diagnostic utility of flow cytometric immunophenotyping in myelodysplastic syndrome. Blood, 2001, 98(4):979–87., DOI 10.1182/blood.v98.4.979 Ogata K, Della Porta MG, Malcovati L, Picone C, Yokose N, Matsuda A, et al. Diagnostic utility of flow cytometry in low-grade myelodysplastic syndromes: a prospective validation study. Haematologica, 2009, 94(8):1066–74., DOI 10.3324/ haematol.2009.008532 Kalina T, Flores-Montero J, van der Velden VHJ, Martin-Ayuso M, Bottcher S, Ritgen M, et al. EuroFlow standardization of flow cytometer instrument settings and immunophenotyping protocols. Leukemia, 2012, 26(9):1986–2010., DOI 10. 1038/leu.2012.122 Matzen S, Raaschou-Jensen KK, Kallenbach K Implementation of the Ogata flow cytometric scoring system in routine diagnostics of myelodysplastic syndrome. Health Sci Rep, 2018, 1(11):e90., DOI 10.1002/hsr2.90 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611811 EP 1611818 DI 10.3389/pore.2024.1611811 PG 8 ER PT J AU Barati, L Maasz, A Miko, A Bercesi, Al Kalbani, S Bene, J Kovacs, S Mangel, L Hadzsiev, K AF Barati, Laszlo Maasz, Anita Miko, Alexandra Bercesi, Eva Al Kalbani, Sultan Bene, Judit Kovacs, Sebestyen Mangel, Laszlo Hadzsiev, Kinga TI Molecular genetic investigation of hereditary breast and ovarian cancer patients in the Southern Transdanubian region: widening the mutation spectrum and searching for new pathogenic variants using next-generation methods SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HBOC; new generation sequencing; multigene panel testing; BRCA1; BRCA2 ID HBOC; new generation sequencing; multigene panel testing; BRCA1; BRCA2 AB Hereditary breast and ovarian cancer is a well-known genetic condition, inherited mainly in an autosomal dominant way, which elevates the risk of developing malignancies at a young age in heterozygous carriers. Advances in new generation sequencing have enabled medical professionals to determine whether a patient is harbouring mutations in moderate- or high penetrance susceptibility genes. We conducted a retrospective analysis among 275 patients who underwent genetic counselling and multigene panel testing for hereditary breast and ovarian cancer syndrome in our department. From these patients 74.5% (205/275) were affected by some type of malignancy, while the remaining 25.5% (70/275) had a positive family history of different cancers, suggesting a genetic predisposition. These tests confirmed a genetic variant in 29.8% and 28.6% of these patient groups respectively. The results also mirrored our general knowledge concerning the genetic background of hereditary breast and ovarian cancer, as variants in either one of the BRCA1 and BRCA2 genes proved to be the most common cause among our patients with 41.5%. Our test also detected a novel mutation in the CDH1 gene and three patients with double heterozygosity in two different susceptibility genes. This study demonstrates the relevance of genetic counselling and non-BRCA gene sequencing among cancer patients and patients who fulfil the criteria for genetic testing, while also providing important details about the genetic profile of Hungarian patients. C1 [Barati, Laszlo] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Maasz, Anita] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Miko, Alexandra] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Bercesi, Eva] University of Pecs, Oncotherapy InstitutePecs, Hungary. [Al Kalbani, Sultan] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Bene, Judit] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Kovacs, Sebestyen] University of Pecs, Department of UrologyPecs, Hungary. [Mangel, Laszlo] University of Pecs, Oncotherapy InstitutePecs, Hungary. [Hadzsiev, Kinga] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. RP Barati, L (reprint author), University of Pecs, Department of Medical Genetics and Child Development, Pecs, Hungary. EM barati.laszlo@pte.hu CR Mattiuzzi C, Lippi G. Current cancer epidemiology. J Epidemiol Glob Health, 2019, 9(4):217–22., DOI 10.2991/jegh.k.191008.001 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer J clinicians, 2021, 71(3): 209–49., DOI 10.3322/caac.21660 Garner WB, Smith BD, Ludmir EB, Wakefield DV, Shabason J, Williams GR, et al. Predicting future cancer incidence by age, race, ethnicity, and sex. J Geriatr Oncol, 2023, 14(1):101393., DOI 10.1016/j.jgo.2022.10.008 Nemzeti Rakregiszter. Nemzeti rakregiszter, 2022). Available from: http://stat. nrr.hu/, Accessed May 26, 2022). Howell A, Anderson AS, Clarke RB, Duffy SW, Evans DG, Garcia-Closas M, et al. Risk determination and prevention of breast cancer. Breast Cancer Res : BCR, 2014, 16(5):446., DOI 10.1186/s13058-014-0446-2 Kruk J, Aboul-Enein HY. Environmental exposure, and other behavioral risk factors in breast cancer. Curr Cancer Ther Rev, 2006, 2:3–21., DOI 10.2174/ 157339406775471795 Momenimovahed Z, Salehiniya H. Epidemiological characteristics of and risk factors for breast cancer in the world. Breast Cancer, Dove Med Press,, 2019, 11: 151–64., DOI 10.2147/BCTT.S176070 Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, et al. Patterns of somatic mutation in human cancer genomes. Nature, 2007, 446(7132): 153–8., DOI 10.1038/nature05610 Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, et al. COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res, 2019, 47(D1): D941–947., DOI 10.1093/nar/gky1015 Fan X, Wynn J, Shang N, Liu C, Fedotov A, Hallquist MLG, et al. Penetrance of breast cancer susceptibility genes from the eMERGE III Network. JNCI Cancer Spectr, 2021, 5(4):pkab044., DOI 10.1093/jncics/pkab044 Garber JE, Offit K. Hereditary cancer predisposition syndromes. J Clin Oncol, 2005, 23(2):276–92., DOI 10.1200/JCO.2005.10.042 Rebbeck TR, Friebel TM, Mitra N, Wan F, Chen S, Andrulis IL, et al. Inheritance of deleteriousmutations at both BRCA1 and BRCA2 in an international sample of 32,295 women. Breast Cancer Res : BCR, 2016, 18(1):112., DOI 10.1186/ s13058-016-0768-3 Gohil D, SarkerAH, Roy R. Base excision repair:mechanisms and impact in biology, disease, and medicine. Int J Mol Sci, 2023, 24(18):14186., DOI 10.3390/ijms241814186 Hirai Y, Banno K, Suzuki M, Ichikawa Y, Udagawa Y, Sugano K, et al. Molecular epidemiological and mutational analysis of DNA mismatch repair, MMR, genes in endometrial cancer patients with HNPCC-associated familial predisposition to cancer. Cancer Sci, 2008, 99(9):1715–9., DOI 10.1111/j.1349-7006. 2008.00886.x Shao C,Wan J, Lam FC, Tang H, Marley AR, Song Y, et al. A comprehensive literature review and meta-analysis of the prevalence of pan-cancer BRCA mutations, homologous recombination repair gene mutations, and homologous recombination deficiencies. Environ Mol mutagenesis, 2022, 63(6):308–16., DOI 10. 1002/em.22505 Butz H, Nagy P, Papp J, Bozsik A, Grolmusz VK, Pocza T, et al. PALB2 variants extend the mutational profile of Hungarian patients with breast and ovarian cancer. Cancers, 2023, 15(17):4350., DOI 10.3390/cancers15174350 Slavin TP, Maxwell KN, Lilyquist J, Vijai J, Neuhausen SL, Hart SN, et al. The contribution of pathogenic variants in breast cancer susceptibility genes to familial breast cancer risk. NPJ breast cancer, 2017, 3:22., DOI 10.1038/s41523- 017-0024-8 Tischkowitz M, Balmana J, FoulkesWD, James P, Ngeow J, Schmutzler R, et al. Management of individuals with germline variants in PALB2: a clinical practice resource of the American College of Medical Genetics and Genomics, ACMG). Genet Med, 2021, 23(8):1416–23., DOI 10.1038/s41436- 021-01151-8 Graffeo R, Rana HQ, Conforti F, Bonanni B, Cardoso MJ, Paluch-Shimon S, et al. Moderate penetrance genes complicate genetic testing for breast cancer diagnosis: ATM, CHEK2, BARD1 and RAD51D. Breast, Edinburgh, Scotland,, 2022, 65:32–40., DOI 10.1016/j.breast.2022.06.003 Grafodatskaya D, O’Rielly DD, Bedard K, Butcher DT, Howlett CJ, Lytwyn A, et al. Practice guidelines for BRCA1/2 tumour testing in ovarian cancer. JMed Genet, 2022, 59(8):727–36., DOI 10.1136/jmedgenet-2021-108238 Bouwman P, van der Gulden H, van der Heijden I, Drost R, Klijn CN, Prasetyanti P, et al. A high-throughput functional complementation assay for classification of BRCA1 missense variants. Cancer Discov, 2013, 3(10):1142–55., DOI 10.1158/2159-8290.CD-13-0094 Antoniou AC, Pharoah PD, Narod S, Risch HA, Eyfjord JE, Hopper JL, et al. Breast and ovarian cancer risks to carriers of the BRCA1 5382insC and 185delAG and BRCA2 6174delT mutations: a combined analysis of 22 population based studies. J Med Genet, 2005, 42(7):602–3., DOI 10.1136/jmg.2004.024133 Finkelman BS, Rubinstein WS, Friedman S, Friebel TM, Dubitsky S, Schonberger NS, et al. Breast and ovarian cancer risk and risk reduction in Jewish BRCA1/2 mutation carriers. J Clin Oncol, 2012, 30(12):1321–8., DOI 10. 1200/jco.2011.37.8133 Fostira F, Saloustros E, Apostolou P, Vagena A, Kalfakakou D, Mauri D, et al. Germline deleterious mutations in genes other than BRCA2 are infrequent in male breast cancer. Breast Cancer Res Treat, 2018, 169(1):105–13., DOI 10.1007/s10549- 018-4661-x Kluz T, Jasiewicz A, Marczyk E, Jach R, Jakubowska A, Lubinski J, et al. Frequency of BRCA1 and BRCA2 causative founder variants in ovarian cancer patients in South-East Poland. Hereditary Cancer Clin Pract, 2018, 16:6., DOI 10. 1186/s13053-018-0089-x Pritchard CC, Mateo J, Walsh MF, De Sarkar N, Abida W, Beltran H, et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. New Engl J Med, 2016, 375(5):443–53., DOI 10.1056/NEJMoa1603144 Yurgelun MB, Chittenden AB, Morales-Oyarvide V, Rubinson DA, Dunne RF, Kozak MM, et al. Germline cancer susceptibility gene variants, somatic second hits, and survival outcomes in patients with resected pancreatic cancer. Genet Med, 2019, 21(1):213–23., DOI 10.1038/s41436-018-0009-5 Ladopoulou A, Kroupis C, Konstantopoulou I, Ioannidou-Mouzaka L, Schofield AC, Pantazidis A, et al. Germ line BRCA1 & BRCA2 mutations in Greek breast/ovarian cancer families: 5382insC is the most frequent mutation observed. Cancer Lett, 2002, 185(1):61–70., DOI 10.1016/s0304-3835(01, 00845-x Chen S, Francioli LC, Goodrich JK, Collins RL, Kanai M, Wang Q, et al. A genomic mutational constraint map using variation in 76,156 human genomes. Nature, 2024, 625(7993):92–100., DOI 10.1038/s41586-023-06045-0 Campeau PM, Foulkes WD, Tischkowitz MD. Hereditary breast cancer: new genetic developments, new therapeutic avenues. Hum Genet, 2008, 124(1):31–42., DOI 10.1007/s00439-008-0529-1 Marmolejo DH, WongMYZ, Bajalica-Lagercrantz S, Tischkowitz M, Balmana J, extended ERN-GENTURIS Thematic Group 3. Overview of hereditary breast and ovarian cancer, HBOC, guidelines across Europe. Eur J Med Genet, 2021, 64(12): 104350., DOI 10.1016/j.ejmg.2021.104350 Owens DK, Davidson KW, Krist AH, Barry MJ, Cabana M, Caughey AB, et al. Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer: US preventive services task force recommendation statement. Jama, 2019, 322(7): 652–65., DOI 10.1001/jama.2019.10987 Pashayan N, Antoniou AC, Ivanus U, Esserman LJ, Easton DF, French D, et al. Personalized early detection and prevention of breast cancer: ENVISION consensus statement. Nat Rev Clin Oncol, 2020, 17(11):687–705., DOI 10.1038/s41571-020-0388-9 Miller DT, Lee K, Abul-Husn NS, Amendola LM, Brothers K, Chung WK, et al. ACMG SF v3.1 list for reporting of secondary findings in clinical exome and genome sequencing: a policy statement of the American College of Medical Genetics and Genomics, ACMG). Genet Med, 2022, 24(7):1407–14., DOI 10. 1016/j.gim.2022.04.006 Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of medical genetics and genomics and the association for molecular Pathology. Genet Med, 2015, 17(5):405–24., DOI 10.1038/gim.2015.30 Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, et al. A method and server for predicting damaging missense mutations. Nat Methods, 2010, 7(4):248–9., DOI 10.1038/nmeth0410-248 Laish I, Friedman E, Levi-Reznick G, Kedar I, Katz L, Levi Z, et al. Double heterozygotes of BRCA1/BRCA2 and mismatch repair gene pathogenic variants: case series and clinical implications. Breast Cancer Res Treat, 2021, 188(3):685–94., DOI 10.1007/s10549-021-06258-9 Megid TBC, Barros-Filho MC, Pisani JP, Achatz MI. Double heterozygous pathogenic variants prevalence in a cohort of patients with hereditary breast cancer. Front Oncol, 2022, 12:873395., DOI 10.3389/fonc.2022.873395 Vos JR, Giepmans L, Rohl C, Geverink N, Hoogerbrugge N, ERN GENTURIS. Boosting care and knowledge about hereditary cancer: European reference Network on genetic tumour risk syndromes. Fam Cancer, 2019, 18(2):281–4., DOI 10.1007/ s10689-018-0110-6 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611813 EP 1611824 DI 10.3389/pore.2024.1611813 PG 12 ER PT J AU Barati, L Maasz, A Miko, A Bercesi, Al Kalbani, S Bene, J Kovacs, S Mangel, L Hadzsiev, K AF Barati, Laszlo Maasz, Anita Miko, Alexandra Bercesi, Eva Al Kalbani, Sultan Bene, Judit Kovacs, Sebestyen Mangel, Laszlo Hadzsiev, Kinga TI Molecular genetic investigation of hereditary breast and ovarian cancer patients in the Southern Transdanubian region: widening the mutation spectrum and searching for new pathogenic variants using next-generation methods SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HBOC; new generation sequencing; multigene panel testing; BRCA1; BRCA2 ID HBOC; new generation sequencing; multigene panel testing; BRCA1; BRCA2 AB Hereditary breast and ovarian cancer is a well-known genetic condition, inherited mainly in an autosomal dominant way, which elevates the risk of developing malignancies at a young age in heterozygous carriers. Advances in new generation sequencing have enabled medical professionals to determine whether a patient is harbouring mutations in moderate- or high penetrance susceptibility genes. We conducted a retrospective analysis among 275 patients who underwent genetic counselling and multigene panel testing for hereditary breast and ovarian cancer syndrome in our department. From these patients 74.5% (205/275) were affected by some type of malignancy, while the remaining 25.5% (70/275) had a positive family history of different cancers, suggesting a genetic predisposition. These tests confirmed a genetic variant in 29.8% and 28.6% of these patient groups respectively. The results also mirrored our general knowledge concerning the genetic background of hereditary breast and ovarian cancer, as variants in either one of the BRCA1 and BRCA2 genes proved to be the most common cause among our patients with 41.5%. Our test also detected a novel mutation in the CDH1 gene and three patients with double heterozygosity in two different susceptibility genes. This study demonstrates the relevance of genetic counselling and non-BRCA gene sequencing among cancer patients and patients who fulfil the criteria for genetic testing, while also providing important details about the genetic profile of Hungarian patients. C1 [Barati, Laszlo] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Maasz, Anita] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Miko, Alexandra] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Bercesi, Eva] University of Pecs, Oncotherapy InstitutePecs, Hungary. [Al Kalbani, Sultan] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Bene, Judit] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. [Kovacs, Sebestyen] University of Pecs, Department of UrologyPecs, Hungary. [Mangel, Laszlo] University of Pecs, Oncotherapy InstitutePecs, Hungary. [Hadzsiev, Kinga] University of Pecs, Department of Medical Genetics and Child DevelopmentPecs, Hungary. RP Barati, L (reprint author), University of Pecs, Department of Medical Genetics and Child Development, Pecs, Hungary. EM barati.laszlo@pte.hu CR Mattiuzzi C, Lippi G. Current cancer epidemiology. J Epidemiol Glob Health, 2019, 9(4):217–22., DOI 10.2991/jegh.k.191008.001 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer J clinicians, 2021, 71(3): 209–49., DOI 10.3322/caac.21660 Garner WB, Smith BD, Ludmir EB, Wakefield DV, Shabason J, Williams GR, et al. Predicting future cancer incidence by age, race, ethnicity, and sex. J Geriatr Oncol, 2023, 14(1):101393., DOI 10.1016/j.jgo.2022.10.008 Nemzeti Rakregiszter. Nemzeti rakregiszter, 2022). Available from: http://stat. nrr.hu/, Accessed May 26, 2022). Howell A, Anderson AS, Clarke RB, Duffy SW, Evans DG, Garcia-Closas M, et al. Risk determination and prevention of breast cancer. Breast Cancer Res : BCR, 2014, 16(5):446., DOI 10.1186/s13058-014-0446-2 Kruk J, Aboul-Enein HY. Environmental exposure, and other behavioral risk factors in breast cancer. Curr Cancer Ther Rev, 2006, 2:3–21., DOI 10.2174/ 157339406775471795 Momenimovahed Z, Salehiniya H. Epidemiological characteristics of and risk factors for breast cancer in the world. Breast Cancer, Dove Med Press,, 2019, 11: 151–64., DOI 10.2147/BCTT.S176070 Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, et al. Patterns of somatic mutation in human cancer genomes. Nature, 2007, 446(7132): 153–8., DOI 10.1038/nature05610 Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, et al. COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res, 2019, 47(D1): D941–947., DOI 10.1093/nar/gky1015 Fan X, Wynn J, Shang N, Liu C, Fedotov A, Hallquist MLG, et al. Penetrance of breast cancer susceptibility genes from the eMERGE III Network. JNCI Cancer Spectr, 2021, 5(4):pkab044., DOI 10.1093/jncics/pkab044 Garber JE, Offit K. Hereditary cancer predisposition syndromes. J Clin Oncol, 2005, 23(2):276–92., DOI 10.1200/JCO.2005.10.042 Rebbeck TR, Friebel TM, Mitra N, Wan F, Chen S, Andrulis IL, et al. Inheritance of deleteriousmutations at both BRCA1 and BRCA2 in an international sample of 32,295 women. Breast Cancer Res : BCR, 2016, 18(1):112., DOI 10.1186/ s13058-016-0768-3 Gohil D, SarkerAH, Roy R. Base excision repair:mechanisms and impact in biology, disease, and medicine. Int J Mol Sci, 2023, 24(18):14186., DOI 10.3390/ijms241814186 Hirai Y, Banno K, Suzuki M, Ichikawa Y, Udagawa Y, Sugano K, et al. Molecular epidemiological and mutational analysis of DNA mismatch repair, MMR, genes in endometrial cancer patients with HNPCC-associated familial predisposition to cancer. Cancer Sci, 2008, 99(9):1715–9., DOI 10.1111/j.1349-7006. 2008.00886.x Shao C,Wan J, Lam FC, Tang H, Marley AR, Song Y, et al. A comprehensive literature review and meta-analysis of the prevalence of pan-cancer BRCA mutations, homologous recombination repair gene mutations, and homologous recombination deficiencies. Environ Mol mutagenesis, 2022, 63(6):308–16., DOI 10. 1002/em.22505 Butz H, Nagy P, Papp J, Bozsik A, Grolmusz VK, Pocza T, et al. PALB2 variants extend the mutational profile of Hungarian patients with breast and ovarian cancer. Cancers, 2023, 15(17):4350., DOI 10.3390/cancers15174350 Slavin TP, Maxwell KN, Lilyquist J, Vijai J, Neuhausen SL, Hart SN, et al. The contribution of pathogenic variants in breast cancer susceptibility genes to familial breast cancer risk. NPJ breast cancer, 2017, 3:22., DOI 10.1038/s41523- 017-0024-8 Tischkowitz M, Balmana J, FoulkesWD, James P, Ngeow J, Schmutzler R, et al. Management of individuals with germline variants in PALB2: a clinical practice resource of the American College of Medical Genetics and Genomics, ACMG). Genet Med, 2021, 23(8):1416–23., DOI 10.1038/s41436- 021-01151-8 Graffeo R, Rana HQ, Conforti F, Bonanni B, Cardoso MJ, Paluch-Shimon S, et al. Moderate penetrance genes complicate genetic testing for breast cancer diagnosis: ATM, CHEK2, BARD1 and RAD51D. Breast, Edinburgh, Scotland,, 2022, 65:32–40., DOI 10.1016/j.breast.2022.06.003 Grafodatskaya D, O’Rielly DD, Bedard K, Butcher DT, Howlett CJ, Lytwyn A, et al. Practice guidelines for BRCA1/2 tumour testing in ovarian cancer. JMed Genet, 2022, 59(8):727–36., DOI 10.1136/jmedgenet-2021-108238 Bouwman P, van der Gulden H, van der Heijden I, Drost R, Klijn CN, Prasetyanti P, et al. A high-throughput functional complementation assay for classification of BRCA1 missense variants. Cancer Discov, 2013, 3(10):1142–55., DOI 10.1158/2159-8290.CD-13-0094 Antoniou AC, Pharoah PD, Narod S, Risch HA, Eyfjord JE, Hopper JL, et al. Breast and ovarian cancer risks to carriers of the BRCA1 5382insC and 185delAG and BRCA2 6174delT mutations: a combined analysis of 22 population based studies. J Med Genet, 2005, 42(7):602–3., DOI 10.1136/jmg.2004.024133 Finkelman BS, Rubinstein WS, Friedman S, Friebel TM, Dubitsky S, Schonberger NS, et al. Breast and ovarian cancer risk and risk reduction in Jewish BRCA1/2 mutation carriers. J Clin Oncol, 2012, 30(12):1321–8., DOI 10. 1200/jco.2011.37.8133 Fostira F, Saloustros E, Apostolou P, Vagena A, Kalfakakou D, Mauri D, et al. Germline deleterious mutations in genes other than BRCA2 are infrequent in male breast cancer. Breast Cancer Res Treat, 2018, 169(1):105–13., DOI 10.1007/s10549- 018-4661-x Kluz T, Jasiewicz A, Marczyk E, Jach R, Jakubowska A, Lubinski J, et al. Frequency of BRCA1 and BRCA2 causative founder variants in ovarian cancer patients in South-East Poland. Hereditary Cancer Clin Pract, 2018, 16:6., DOI 10. 1186/s13053-018-0089-x Pritchard CC, Mateo J, Walsh MF, De Sarkar N, Abida W, Beltran H, et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. New Engl J Med, 2016, 375(5):443–53., DOI 10.1056/NEJMoa1603144 Yurgelun MB, Chittenden AB, Morales-Oyarvide V, Rubinson DA, Dunne RF, Kozak MM, et al. Germline cancer susceptibility gene variants, somatic second hits, and survival outcomes in patients with resected pancreatic cancer. Genet Med, 2019, 21(1):213–23., DOI 10.1038/s41436-018-0009-5 Ladopoulou A, Kroupis C, Konstantopoulou I, Ioannidou-Mouzaka L, Schofield AC, Pantazidis A, et al. Germ line BRCA1 & BRCA2 mutations in Greek breast/ovarian cancer families: 5382insC is the most frequent mutation observed. Cancer Lett, 2002, 185(1):61–70., DOI 10.1016/s0304-3835(01, 00845-x Chen S, Francioli LC, Goodrich JK, Collins RL, Kanai M, Wang Q, et al. A genomic mutational constraint map using variation in 76,156 human genomes. Nature, 2024, 625(7993):92–100., DOI 10.1038/s41586-023-06045-0 Campeau PM, Foulkes WD, Tischkowitz MD. Hereditary breast cancer: new genetic developments, new therapeutic avenues. Hum Genet, 2008, 124(1):31–42., DOI 10.1007/s00439-008-0529-1 Marmolejo DH, WongMYZ, Bajalica-Lagercrantz S, Tischkowitz M, Balmana J, extended ERN-GENTURIS Thematic Group 3. Overview of hereditary breast and ovarian cancer, HBOC, guidelines across Europe. Eur J Med Genet, 2021, 64(12): 104350., DOI 10.1016/j.ejmg.2021.104350 Owens DK, Davidson KW, Krist AH, Barry MJ, Cabana M, Caughey AB, et al. Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer: US preventive services task force recommendation statement. Jama, 2019, 322(7): 652–65., DOI 10.1001/jama.2019.10987 Pashayan N, Antoniou AC, Ivanus U, Esserman LJ, Easton DF, French D, et al. Personalized early detection and prevention of breast cancer: ENVISION consensus statement. Nat Rev Clin Oncol, 2020, 17(11):687–705., DOI 10.1038/s41571-020-0388-9 Miller DT, Lee K, Abul-Husn NS, Amendola LM, Brothers K, Chung WK, et al. ACMG SF v3.1 list for reporting of secondary findings in clinical exome and genome sequencing: a policy statement of the American College of Medical Genetics and Genomics, ACMG). Genet Med, 2022, 24(7):1407–14., DOI 10. 1016/j.gim.2022.04.006 Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of medical genetics and genomics and the association for molecular Pathology. Genet Med, 2015, 17(5):405–24., DOI 10.1038/gim.2015.30 Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, et al. A method and server for predicting damaging missense mutations. Nat Methods, 2010, 7(4):248–9., DOI 10.1038/nmeth0410-248 Laish I, Friedman E, Levi-Reznick G, Kedar I, Katz L, Levi Z, et al. Double heterozygotes of BRCA1/BRCA2 and mismatch repair gene pathogenic variants: case series and clinical implications. Breast Cancer Res Treat, 2021, 188(3):685–94., DOI 10.1007/s10549-021-06258-9 Megid TBC, Barros-Filho MC, Pisani JP, Achatz MI. Double heterozygous pathogenic variants prevalence in a cohort of patients with hereditary breast cancer. Front Oncol, 2022, 12:873395., DOI 10.3389/fonc.2022.873395 Vos JR, Giepmans L, Rohl C, Geverink N, Hoogerbrugge N, ERN GENTURIS. Boosting care and knowledge about hereditary cancer: European reference Network on genetic tumour risk syndromes. Fam Cancer, 2019, 18(2):281–4., DOI 10.1007/ s10689-018-0110-6 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611814 EP 1611825 DI 10.3389/pore.2024.1611813 PG 12 ER PT J AU Csontos, A Nemeth, D Szako, L Berke, G Sindler, LD Berki, D Papp, Cs Hegyi, P Vereczkei, A Papp, A AF Csontos, Armand Nemeth, David Szako, Lajos Berke, Gergo Sindler, Lili Dora Berki, David Papp, Csenge Hegyi, Peter Vereczkei, Andras Papp, Andras TI Intraoperative pyloric drainage is unnecessary during esophagectomies: a meta-analysis and systematic review of randomized controlled trials SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE esophageal cancer; esophagectomy; esophageal surgery; pyloroplasty; minimal invasive ID esophageal cancer; esophagectomy; esophageal surgery; pyloroplasty; minimal invasive AB Objective: The topic of this meta-analysis is the comparison of gastric conduit esophageal reconstructions with or without pyloroplasty. Background: Surgical procedures, especially minimal invasive esophagectomy (MIE) can be a curative treatment in the early stages of esophageal cancer. Previously, intraoperative pyloroplasty was routinely performed, but nowadays it became debated again in the light of minimally invasive esophagectomy. Methods: A comprehensive search was performed in multiple databases to identify randomized controlled trials investigating the topic. Two independent authors performed the selection based on predefined criteria. Statistical analysis was performed to assess any significant difference, then the bias and quality of the data were estimated. Results: Nine relevant RCTs consisting of 529 patients with esophageal cancer were identified. No significance was found in mortality [odds ratio (OR): 0.85; p = 0.642], anastomosis leakage (OR: 0.57; p = 0.254), respiratory morbidity (OR: 0.51; p = 0.214) and vomiting (OR: 0.74; p = 0.520), however the results about gastric emptying time (GET) were controversial (weighted mean difference (WMD): −67.71; p = 0.009, OR: 2.75; p = 0.072). Significant heterogeneity was not detected except for GET. Trial sequential analyses (TSA) show that a certain conclusion would require more data except in the binary variables of GET. Conclusion: We conclude that the pyloric drainage procedure is not routinely necessary, but further well-designed studies would be needed, especially in Europe. C1 [Csontos, Armand] University of Pecs, Department of SurgeryPecs, Hungary. [Nemeth, David] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Szako, Lajos] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Berke, Gergo] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Sindler, Lili Dora] University of Pecs, Department of SurgeryPecs, Hungary. [Berki, David] Military Hospital Medical Centre, Hungarian Defense Forces, First Department of SurgeryBudapest, Hungary. [Papp, Csenge] University of Pecs, Department of SurgeryPecs, Hungary. [Hegyi, Peter] University of Pecs, Institute for Translational MedicinePecs, Hungary. [Vereczkei, Andras] University of Pecs, Department of SurgeryPecs, Hungary. [Papp, Andras] University of Pecs, Department of SurgeryPecs, Hungary. RP Csontos, A (reprint author), University of Pecs, Department of Surgery, Pecs, Hungary. EM csontos.armand@pte.hu CR GLOBOCAN. Cancer today - IARC, 150 cours Albert Thomas, 69372 Lyon CEDEX 08, France - Tel: +33, 0)4 72 73 84 85 - powered by GLOBOCAN, 2020). Tinusz B, Szapary LB, Paladi B, Papp A, Bogner B, Hegedus I, et al. The esophageal adenocarcinoma epidemic has reached Hungary: a multicenter, cross-sectional study. Front Oncol, 2020, 10:541794., DOI 10.3389/fonc.2020. 541794 Arnold M, Soerjomataram I, Ferlay J, Forman D. Global incidence of oesophageal cancer by histological subtype in 2012. Gut, 2015, 64(3):381–7., DOI 10.1136/gutjnl-2014-308124 Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin, 2014, 64(1):9–29. Epub 2014 Jan 7. Erratum in: CA Cancer J Clin. 2014 Sep-Oct; 64(5): 364., DOI 10.3322/caac.21208 De Angelis R, Sant M, Coleman MP, Francisci S, Baili P, Pierannunzio D, et al. Cancer survival in Europe 1999–2007 by country and age: results of EUROCARE-- 5-a population-based study. Lancet Oncol, 2014, 15(1):23–34., DOI 10.1016/S1470- 2045(13)70546-1 Yendamuri S, Swisher SG, Correa AM, Hofstetter W, Ajani JA, Francis A, et al. Esophageal tumor length is independently associated with long-term survival. Cancer, 2009, 115(3):508–16., DOI 10.1002/cncr.24062 Kim SH, Lee KS, Shim YM, Kim K, Yang PS, Kim TS. Esophageal resection: indications, techniques, and radiologic assessment. Radiographics, 2001, 21(5): 1119–37. discussion 1138-40., DOI 10.1148/radiographics.21.5.g01se031119 Rice TW, Patil DT, Blackstone EH. 8th edition AJCC/UICC staging of cancers of the esophagus and esophagogastric junction: application to clinical practice. Ann Cardiothorac Surg, 2017, 6(2):119–30., DOI 10.21037/acs.2017.03.14 Noordman BJ, Gisbertz SS. Minimally invasive oesophagectomy as standard of care. Br J Surg, 2023, 13:1118–9., DOI 10.1093/bjs/znad209 Siaw-Acheampong K, Kamarajah SK, Gujjuri R, Bundred JR, Singh P, Griffiths EA. Minimally invasive techniques for transthoracic oesophagectomy for oesophageal cancer: systematic review and network meta-analysis. BJS Open, 2020, 4(5):787–803., DOI 10.1002/bjs5.50330 Nevo Y, Calderone A, Kammili A, Boulila C, Renaud S, Cools-Lartigue J, et al. Endoscopic pyloromyotomy in minimally invasive esophagectomy: a novel approach. Surg Endosc, 2022, 36(4):2341–8., DOI 10.1007/s00464-021- 08511-0 Nobel T, Tan KS, Barbetta A, Adusumilli P, Bains M, Bott M, et al. Does pyloric drainage have a role in the era of minimally invasive esophagectomy? Surg Endosc, 2019, 33(10):3218–27., DOI 10.1007/s00464-018-06607-8 Kim D. The optimal pyloric procedure: a collective review. Korean J Thorac Cardiovasc Surg, 2020, 53(4):233–41., DOI 10.5090/kjtcs.2020.53.4.233 Hagen JA, Peyre CG. Gastric emptying procedures after esophagectomy. In: Ferguson MK, editor. Difficult decisions in thoracic surgery. London: Springer, 2007)., DOI 10.1007/978-1-84628-474-8_30 Gaur P, Swanson SJ. Should we continue to drain the pylorus in patients undergoing an esophagectomy? Dis Esophagus, 2014, 27(6):568–73., DOI 10.1111/ dote.12035 Arya S, Markar SR, Karthikesalingam A, Hanna GB. The impact of pyloric drainage on clinical outcome following esophagectomy: a systematic review. Dis Esophagus, 2015, 28(4):326–35., DOI 10.1111/dote.12191 Palmes D, Weilinghoff M, Colombo-Benkmann M, Senninger N, Bruewer M. Effect of pyloric drainage procedures on gastric passage and bile reflux after esophagectomy with gastric conduit reconstruction. Langenbecks Arch Surg, 2007, 392(2):135–41., DOI 10.1007/s00423-006-0119-4 Mohajeri G, Tabatabaei SA, Hashemi SM, Hemmati H. Comparison of pyloromyotomy, pyloric buginage, and intact pylorus on gastric drainage in gastric pull-up surgery after esophagectomy. J Res Med Sci, 2016, 21:33., DOI 10. 4103/1735-1995.181993 Urschel JD, Blewett CJ, Young JE, Miller JD, Bennett WF. Pyloric drainage, pyloroplasty, or no drainage in gastric reconstruction after esophagectomy: a meta-analysis of randomized controlled trials. Dig Surg, 2002, 19(3):160–4., DOI 10. 1159/000064206 Csontos A, Nemeth D, Szako L, Berke G, Sindler DL, Hegyi P, et al., 2022). The necessity of pyloric drainage in esophagectomies: protocol of a meta-analysis and a systematic review of randomized controlled trials. Available from: https:// www.medrxiv.org/., DOI 10.1101/2022.08.24.22279164 DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials, 1986, 7(3):177–88., DOI 10.1016/0197-2456(86)90046-2 Higgins JP, Altman DG, Gotzsche PC, Juni P, Moher D, Oxman AD, et al. The Cochrane collaboration’s tool for assessing risk of bias in randomised trials. BMJ, 2011, 343:d5928., DOI 10.1136/bmj.d5928 Brozek JL, Akl EA, Alonso-Coello P, Lang D, Jaeschke R, Williams JW, et al. Grading quality of evidence and strength of recommendations in clinical practice guidelines. Part 1 of 3. An overview of the GRADE approach and grading quality of evidence about interventions. Allergy, 2009, 64(5):669–77., DOI 10.1111/j.1398- 9995.2009.01973.x Fok M, Cheng SW, Wong J. Pyloroplasty versus no drainage in gastric replacement of the esophagus. Am J Surg, 1991, 162(5):447–52., DOI 10.1016/0002- 9610(91)90258-f Mannell A, McKnight A, Esser JD. Role of pyloroplasty in the retrosternal stomach: results of a prospective, randomized, controlled trial. Br J Surg, 1990, 77(1):57–9., DOI 10.1002/bjs.1800770120 Chattopadhyay TK, Gupta S, Padhy AK, Kapoor VK. Is pyloroplasty necessary following intrathoracic transposition of stomach? Results of a prospective clinical study. Aust N Z J Surg, 1991, 61(5):366–9., DOI 10.1111/j. 1445-2197.1991.tb00236.x Chattopadhyay TK, Shad SK, Kumar A. Intragastric bile acid and symptoms in patients with an intrathoracic stomach after oesophagectomy. Br J Surg, 1993, 80(3):371–3., DOI 10.1002/bjs.1800800336 Zieren HU, Muller JM, Jacobi CA, Pichlmaier H. Soll beim magenhochzug nach subtotaler oesophagektomie mit oesophago-gastraler anastomose am hals eine pyloroplastik durchgefuhrt werden? Eine prospektiv randomisierte studie should a pyloroplasty be carried out in stomach transposition after subtotal esophagectomy with esophago-gastric anastomosis at the neck? A prospective randomized study. Chirurg, 1995, 66(4):319–25. Kao CH, Chen CY, Chen CL, Wang SJ, Yeh SH. Gastric emptying of the intrathoracic stomach as oesophageal replacement for oesophageal carcinomas. Nucl Med Commun, 1994, 15(3):152–5., DOI 10.1097/00006231-199403000-00006 Gupta S, Chattopadhyay TK, Gopinath PG, Kapoor VK, Sharma LK. Emptying of the intrathoracic stomach with and without pyloroplasty. Am J Gastroenterol, 1989, 84(8):921–3. Huang GJ, Zhang DC, Zhang DW. A Comparative study of resection of carcinoma of the esophagus with and without pyloroplasty. In: Demeester TR, Skinner DB, editors. Esophageal disorder: pathophysiology and therapy. New York, NY: Raven Press, 1985). p. 383. Manjari R, Padhy AK, Chattopadhyay TK. Emptying of the intrathoracic stomach using three different pylorus drainage procedures--results of a comparative study. Surg Today, 1996, 26(8):581–5., DOI 10.1007/BF00311660 Bagheri R, Fattahi SH, Haghi SZ, Aryana K, Aryanniya A, Akhlaghi S, et al. Botulinum toxin for prevention of delayed gastric emptying after esophagectomy. Asian Cardiovasc Thorac Ann, 2013, 21(6):689–92., DOI 10.1177/ 0218492312468438 Law S, Cheung MC, Fok M, Chu KM, Wong J. Pyloroplasty and pyloromyotomy in gastric replacement of the esophagus after esophagectomy: a randomized controlled trial. J Am Coll Surg, 1997, 184(6):630–6. Nikbakhsh N, Hashemi R, Noorbaran A. Esophagectomy without pyloroplasty or pyloromyotomy. J Isfahan Med Sch, 2014, 32(277). Nakamura Y, Nakayama T, Mitomi T, Ueda M, Kaitsu N. Clinical study on pyloroplasty and myotomy of the pylorus in surgery of esophageal cancer. Shujutsu, 1967, 21(9):899–910. Cheung HC, Siu KF, Wong J. Is pyloroplasty necessary in esophageal replacement by stomach? A prospective, randomized controlled trial. Surgery, 1987, 102(1):19–24. Kobayashi A, Ide H, Eguchi R, Nakamura T, Hayashi K, Hanyu F. The efficacy of pyloroplasty affecting to oral-intake quality of life using reconstruction with gastric tube post esophagectomy. Nihon Kyobu Geka Gakkai Zasshi, 1996, 44(6): 770–8. Khan OA, Manners J, Rengarajan A, Dunning J. Does pyloroplasty following esophagectomy improve early clinical outcomes? Interact Cardiovasc Thorac Surg, 2007, 6(2):247–50., DOI 10.1510/icvts.2006.149500 Oezcelik A, DeMeester SR, Hindoyan K, Leers JM, Ayazi S, Abate E, et al. Circular stapled pyloroplasty: a fast and effective technique for pyloric disruption during esophagectomy with gastric pull-up. Dis Esophagus, 2011, 24(6):423–9., DOI 10.1111/j.1442-2050.2010.01169.x Szako L, Nemeth D, Farkas N, Kiss S, Domotor RZ, Engh MA, et al. Network meta-analysis of randomized controlled trials on esophagectomies in esophageal cancer: the superiority of minimally invasive surgery. World J Gastroenterol, 2022, 28(30):4201–10., DOI 10.3748/wjg.v28.i30.4201 Fuchs HF, Broderick RC, Harnsberger CR, Divo FA, Coker AM, Jacobsen GR, et al. Intraoperative endoscopic botox injection during total esophagectomy prevents the need for pyloromyotomy or dilatation. J Laparoendosc Adv Surg Tech A, 2016, 26(6):433–8., DOI 10.1089/lap.2015.0575 Anderson MJ, Sippey M, Marks J. Gastric per oral pyloromyotomy for postvagotomy- induced gastroparesis following esophagectomy. J Gastrointest Surg, 2020, 24(3):715–9., DOI 10.1007/s11605-019-04418-3 Giugliano DN, Berger AC, Meidl H, Pucci MJ, Rosato EL, Keith SW, et al. Do intraoperative pyloric interventions predict the need for postoperative endoscopic interventions after minimally invasive esophagectomy? Dis Esophagus, 2017, 30(4): 1–8., DOI 10.1093/dote/dow034 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611823 EP 1611833 DI 10.3389/pore.2024.1611823 PG 11 ER PT J AU Liao, CChH Bakoglu, N Cesmecioglu, E Hanna, M Pareja, F Wen, H D’Alfonso, T Brogi, E Yagi, Y Ross, D AF Liao, Connie Chiu-Hsiang Bakoglu, Nilay Cesmecioglu, Emine Hanna, Matthew Pareja, Fresia Wen, Y. Hannah D’Alfonso, M. Timothy Brogi, Edi Yagi, Yukako Ross, S. Dara TI Semi-automated analysis of HER2 immunohistochemistry in invasive breast carcinoma using whole slide images: utility for interpretation in clinical practice SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE HER2; immunohistochemistry; breast carcinoma; whole slide imaging; semiautomated analysis ID HER2; immunohistochemistry; breast carcinoma; whole slide imaging; semiautomated analysis AB Human epidermal growth factor receptor 2 (HER2) gene amplification and subsequent protein overexpression is a strong prognostic and predictive biomarker in invasive breast carcinoma (IBC). ASCO/CAP recommended tests for HER2 assessment include immunohistochemistry (IHC) and/or in situ hybridization (ISH). Accurate HER2 IHC scoring (0, 1+, 2+, 3+) is key for appropriate classification and treatment of IBC. HER2-targeted therapies, including anti-HER2 monoclonal antibodies and antibody drug conjugates (ADC), have revolutionized the treatment of HER2-positive IBC. Recently, ADC have also been approved for treatment of HER2-low (IHC 1+, IHC 2+/ ISH-) advanced breast carcinoma, making a distinction between IHC 0 and 1+ crucial. In this focused study, 32 IBC with HER2 IHC scores from 0 to 3+ and HER2 FISH results formed a calibration dataset, and 77 IBC with HER2 IHC score 2+ and paired FISH results (27 amplified, 50 nonamplified) formed a validation dataset. H&E and HER2 IHC whole slide images (WSI) were scanned. Regions of interest were manually annotated and IHC scores generated by the software QuantCenter (MembraneQuant application) by 3DHISTECH Ltd. (Budapest, Hungary) and compared to the microscopic IHC score. H-scores [(3×%IHC3+) +(2×%IHC2+) +(1×% IHC1+)] were calculated for semi-automated (MembraneQuant) analysis. Concordance between microscopic IHC scoring and 3DHISTECH MembraneQuant semi-automated scoring in the calibration dataset showed a Kappa value of 0.77 (standard error 0.09). Microscopic IHC and MembraneQuant image analysis for the detection of HER2 amplification yielded a sensitivity of 100% for both and a specificity of 56% and 61%, respectively. In the validation set of IHC 2+ cases, only 13 of 77 cases (17%) had discordant results between microscopic and MembraneQuant images, and various artifacts limiting the interpretation of HER2 IHC, including cytoplasmic/granular staining and crush artifact were noted. Semi-automated analysis using WSI and microscopic evaluation yielded similar HER2 IHC scores, demonstrating the potential utility of this tool for interpretation in clinical practice and subsequent accurate treatment. In this study, it was shown that semi-automatic HER2 IHC interpretation provides an objective approach to a test known to be quite subjective. C1 [Liao, Connie Chiu-Hsiang] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [Bakoglu, Nilay] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [Cesmecioglu, Emine] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [Hanna, Matthew] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [Pareja, Fresia] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [Wen, Y. Hannah] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [D’Alfonso, M. Timothy] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [Brogi, Edi] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [Yagi, Yukako] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [Ross, S. Dara] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. RP Ross, D (reprint author), Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory Medicine, New York, USA. EM rossd@mskcc.org CR Wolff AC, Somerfield MR, Dowsett M, Hammond ME, Hayes DF, McShane LM, et al. Human epidermal growth factor receptor 2 testing in breast cancer: ASCO–College of American Pathologists Guideline Update. J Clin Oncol, 2023, 41(22):3867–72., DOI 10.1200/JCO.22.02864 Hechtman JF, Ross DS. The past, present, and future of HER2, ERBB2, in cancer: approaches to molecular testing and an evolving role in targeted therapy. Cancer Cytopathology, 2019, 127(7):428–31., DOI 10.1002/cncy.22124 Modi S, Park H, Murthy RK, Iwata H, Tamura K, Tsurutani J, et al. Antitumor activity and safety of trastuzumab deruxtecan in patients with HER2- low–expressing advanced breast cancer: results from a phase Ib study. J Clin Oncol, 2020, 38(17):1887–96., DOI 10.1200/JCO.19.02318 Modi S, Jacot W, Yamashita T, Sohn J, Vidal M, Tokunaga E, et al. Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer. New Engl J Med, 2022, 387(1):9–20., DOI 10.1056/NEJMoa2203690 Tarantino P, Hamilton E, Tolaney SM, Cortes J, Morganti S, Ferraro E, et al. HER2-low breast cancer: pathological and clinical landscape. J Clin Oncol, 2020, 38(17):1951–62., DOI 10.1200/JCO.19.02488 Fernandez AI, Liu M, Bellizzi A, Brock J, Fadare O, Hanley K, et al. Examination of low ERBB2 protein expression in breast cancer tissue. JAMA Oncol, 2022, 8(4):1–4., DOI 10.1001/jamaoncol.2021.7239 Zhang H, Katerji H, Turner BM, Hicks DG. HER2-low breast cancers: new opportunities and challenges. Am J Clin Pathol, 2022, 157(3):328–36., DOI 10.1093/ ajcp/aqab117 Wu S, Li X, Miao J, Xian D, Yue M, Liu H, et al. Artificial intelligence for assisted HER2 immunohistochemistry evaluation of breast cancer: a systematic review and meta-analysis. Pathology-Research Pract, 2024, 16:155472., DOI 10.1016/ j.prp.2024.155472 Wolff AC, Hammond ME, Allison KH, Harvey BE, McShane LM, Dowsett M. HER2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline focused update summary. J Oncol Pract, 2018, 14(7):437–41., DOI 10.1200/JOP.18.00206 Allison KH, Hammond ME, Dowsett M, McKernin SE, Carey LA, Fitzgibbons PL, et al. Estrogen and progesterone receptor testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists guideline update. Arch Pathol and Lab Med, 2020, 144(5):545–63., DOI 10.5858/arpa.2019-0904-SA Smith B, Hermsen M, Lesser E, Ravichandar D, Kremers W. Developing image analysis pipelines of whole-slide images: pre-and post-processing. J Clin Translational Sci, 2021, 5(1):e38., DOI 10.1017/cts.2020.531 Manuel C, Zehnder P, Kaya S, Sullivan R, Hu F. Impact of color augmentation and tissue type in deep learning for hematoxylin and eosin image super resolution. J Pathol Inform, 2022, 13:100148., DOI 10.1016/j.jpi.2022.100148 Palm C, Connolly CE, Masser R, Padberg Sgier B, Karamitopoulou E, Simon Q, et al. Determining HER2 status by artificial intelligence: an investigation of primary, metastatic, and HER2 low breast tumors. Diagnostics, 2023, 13(1):168., DOI 10.3390/diagnostics13010168 Guarneri V, Dieci MV, Barbieri E, Piacentini F, Omarini C, Ficarra G, et al. Loss of HER2 positivity and prognosis after neoadjuvant therapy in HER2-positive breast cancer patients. Ann Oncol, 2013, 24(12):2990–4., DOI 10.1093/annonc/ mdt364 Holten-Rossing H, Moller Talman ML, Kristensson M, Vainer B. Optimizing HER2 assessment in breast cancer: application of automated image analysis. Breast Cancer Res Treat, 2015, 152:367–75., DOI 10.1007/ s10549-015-3475-3 Jung M, Song SG, Cho SI, Shin S, Lee T, Jung W, et al. Augmented interpretation of HER2, ER, and PR in breast cancer by artificial intelligence analyzer: enhancing interobserver agreement through a reader study of 201 cases. Breast Cancer Res, 2024, 26:31., DOI 10.1186/s13058-024-01784-y Gokhale S, Rosen D, Sneige N, Diaz LK, Resetkova E, Sahin A, et al. Assessment of two automated imaging systems in evaluating estrogen receptor status in breast carcinoma. Appl Immunohistochem and Mol Morphol, 2007, 15(4): 451–5., DOI 10.1097/PAI.0b013e31802ee998 Gandomkar Z, Brennan PC, Mello-Thoms C, 2016). Computer-based image analysis in breast pathology. J Pathol Inform, 7(1), 43., DOI 10.4103/2153-3539. 192814 Faratian D, Kay C, Robson T, Campbell FM, Grant M, Rea D, et al. Automated image analysis for high-throughput quantitative detection of ER and PR expression levels in large-scale clinical studies: the TEAM Trial Experience. Histopathology, 2009, 55(5):587–93., DOI 10.1111/j.1365-2559. 2009.03419.x Selcuk SY, Yang X, Bai B, Zhang Y, Li Y, Aydin M, et al. Automated HER2 scoring in breast cancer images using deep learning and pyramid sampling. BMEF, BME Frontiers,, 2024, 5:0048., DOI 10.34133/bmef.0048 Ohnishi C, Ohnishi T, Ntiamoah P, Ross DS, Yamaguchi M, Yagi Y. Standardizing HER2 immunohistochemistry assessment: calibration of color and intensity variation in whole slide imaging caused by staining and scanning. Appl Microsc, 2023, 53(1):8., DOI 10.1186/s42649-023-00091-8 Kondo Y, Iijima T, Noguchi M. Evaluation of immunohistochemical staining using whole-slide imaging for HER2 scoring of breast cancer in comparison with real glass slides. Pathol Int, 2012, 62(9):592–9., DOI 10.1111/j.1440-1827.2012. 02847.x Hanna MG, Parwani A, Sirintrapun SJ. Whole slide imaging: technology and applications. Adv Anat Pathol, 2020, 27(4):251–9., DOI 10.1097/PAP. 0000000000000273 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611826 EP 1611835 DI 10.3389/pore.2024.1611826 PG 10 ER PT J AU Han, Y Qiu, W Zhang, Y Hua, M Liu, Sh Dong, Z AF Han, Yibing Qiu, Wen Zhang, Yu Hua, Mengmeng Liu, Shaohua Dong, Zuoqing TI From prenatal diagnosis to surgical treatment: two case reports of congenital granular cell epulis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE case report; surgical treatment; congenital granular cell epulis; prenatal diagnosis; oral and maxillofacial regions ID case report; surgical treatment; congenital granular cell epulis; prenatal diagnosis; oral and maxillofacial regions AB Herein, we detail a multidisciplinary approach and sequential treatment for two infants with congenital granular cell epulis (CGCE). Ultrasonic examinations at 34 weeks of gestation revealed prominent oral masses in both fetuses. To devise a carefully considered treatment strategy, a comprehensive multidisciplinary consultation including oral and maxillofacial surgeons, pediatricians, obstetricians, and anesthesiologists was convened. Following cesarean sections, the lesions were successfully removed, measuring approximately 30 × 15 mm and 30 × 20 mm in size, respectively. Immunohistochemical analysis showed that vimentin was positive, S-100 protein was negative, and NSE protein and CD68 protein were negative. These findings underscore the importance of prenatal diagnosis of congenital granular cell epulis for the effective management of these rare benign conditions. C1 [Han, Yibing] Qi Lu Hospital of Shandong University, Department of Oral and Maxillofacial SurgeryJinan, Shandong, China. [Qiu, Wen] Shandong Academy of Medical SciencesJinan, Shandong, China. [Zhang, Yu] Shandong University, Qilu Hospital, Department of PathologyJinan, Shandong, China. [Hua, Mengmeng] Qi Lu Hospital of Shandong University, Department of Oral and Maxillofacial SurgeryJinan, Shandong, China. [Liu, Shaohua] Qi Lu Hospital of Shandong University, Department of Oral and Maxillofacial SurgeryJinan, Shandong, China. [Dong, Zuoqing] Qi Lu Hospital of Shandong University, Department of Oral and Maxillofacial SurgeryJinan, Shandong, China. RP Dong, Z (reprint author), Qi Lu Hospital of Shandong University, Department of Oral and Maxillofacial Surgery, Jinan, China. EM dongzq@126.com CR Williams RW, Grave B, Stewart M, Heggie AA. Prenatal and postnatal management of congenital granular cell tumours: a case report. Br J Oral Maxillofac Surg, 2009, 47(1):56–8., DOI 10.1016/j.bjoms.2008.04.007 Merglova V, Mukensnabl P, Andrle P. Congenital epulis. BMJ Case Rep, 2012, 2012:bcr0120125483., DOI 10.1136/bcr.01.2012.5483 Neumann E. Ein von congenital epulis. Arch Heilkd, 1871, 12:189–90. Klingler PJ, Seelig MH, DeVault KR, Wetscher GJ, Floch NR, Branton SA, et al. Ingested foreign bodies within the appendix: a 100-year review of the literature. Dig Dis, 1998, 16(5):308–14., DOI 10.1159/000016880 Bosanquet D, Roblin G. Congenital epulis: a case report and estimation of incidence. Int J Otolaryngol, 2009, 2009:508780., DOI 10.1155/2009/508780 Damm DD, Cibull ML, Geissler RH, Neville BW, Bowden CM, Lehmann JE. Investigation into the histogenesis of congenital epulis of the newborn. Oral Surg Oral Med Oral Pathol, 1993, 76:205–12., DOI 10.1016/0030-4220(93)90206-j Koch BL, Myeriii C, Egelhoff JC. Congenital epulis. AJNR, 1997, 18:739–41. Yuwanati M, Mhaske S, Mhaske A. Congenital granular cell tumor - a rare entity. J Neonatal Surg, 2015, 4(2):17., DOI 10.47338/jns.v4.170 Kameyama Y, Mizohata M, Takehana S, Murata H, Manabe H, Mukai Y. Ultrastructure of the congenital epulis. Virchows Arch A Pathol Anat Histopathol, 1983, 401:251–60., DOI 10.1007/BF00734843 Lifshitz MS, Flotte TJ, Greco MA. Congenital granular cell epulis. Immunohistochemical and ultrastructural observations. Cancer, 1984, 53: 1845–8., DOI 10.1002/1097-0142(19840501)53:9<1845::aid-cncr2820530908>3.0. co;2-l Adeyemi BF, Oluwasola AO, Adisa AO. Congenital epulis. Indian J Dent Res, 2010, 21(2):292–4., DOI 10.4103/0970-9290.66638 Kokubun K, Matsuzaka K, Akashi Y, SumiM, Nakajima K, Murakami S, et al. Congenital epulis: a case and review of the literature. Bull Tokyo Dent Coll, 2018, 59(2):127–32., DOI 10.2209/tdcpublication.2017-0028 Dzieniecka M, Komorowska A, Grzelak-Krzymianowska A, Kulig A. Multiple congenital epuli, congenital granular cell tumours, in the newborn: a case report and review of literature. Pol J Pathol, 2011, 62(1): 69–71. Review. Pellicano M, Zullo F, Catizone C, Guida F, Catizone F, Nappi C. Prenatal diagnosis of congenital granular cell epulis. Ultrasound Obstet Gynecol, 1998, 11(2): 144–6., DOI 10.1046/j.1469-0705.1998.11020144.x Sasaki R, Okamoto T, Watanabe Y, Kagawa C, Ando T. Congenital granular cell epulis. Plast Reconstr Surg Glob Open, 2018, 6(11):e1989., DOI 10.1097/GOX. 0000000000001989 Kadivar M, Sangsari R, Alavi A. Prenatal diagnosis of granular cell tumor. Iran J Med Sci, 2014, 39(2):144–7. Cussen LJ, MacMahon RA. Congenital granular-cell myoblastoma. J Pediatr Surg, 1975, 10:249–53., DOI 10.1016/0022-3468(75)90287-0 Godra A, D’Cruz CA, Labat MF, Isaacson G. Pathologic quiz case: a newborn with a midline buccal mucosa mass. Arch Pathol Lab Med, 2004, 128(5):585–6., DOI 10.5858/2004-128-585-PQCANW Sweeney K, Spurway J, Mein B, Magotti R, Benzie R, Challis D, et al. Congenital Epulis: a clinical case presentation. Australas J Ultrasound Med, 2014, 17(2):85–8., DOI 10.1002/j.2205-0140.2014.tb00112.x Bhoil R, Bhoil R, Chopra R, Sharma M, Mistry K. Congenital epulis: a rare benign jaw tumour in a 2-day-old male baby. Pol J Radiol, 2015, 80:395–7., DOI 10. 12659/PJR.894698 Wittebole A, Bayet B, Veyckemans F, Gosseye S, Vanwijck R. Congenital epulis of the newborn. Acta Chir Belg, 2003, 103:235–7., DOI 10.1080/00015458. 2003.11679415 Thoma V, Idrissi B, Kohler M, Becmeur F, Viville B, Favre R. Prenatal diagnosis of congenital epulis. A case study. Fetal Diagn Ther, 2006, 21(4):321–5., DOI 10.1159/000092458 Kumar P, Kim HH, Zahtz GD, Valderrama E, Steele AM. Obstructive congenital epulis: prenatal diagnosis and perinatal management. Laryngoscope, 2002, 112:1935–9., DOI 10.1097/00005537-200211000-00005 Bang KO, Bodhade AS, Dive AM. Congenital granular cell epulis of a newborn. Dent Res J, Isfahan,, 2012, 9(Suppl. 1):S136–8. Tucker MC, Rusnock EJ, Azumi N, Hoy GR, Lack FE. Gingival granular cell tumor of the newborn: an ultrastructural and immunohistochemical study. Arch Pathol Lab Med, 1990, 114:895–8. Zarbo RJ, Lloyd RV, Beals TF, McClaehey KD. Congenital gingival cell tumor with smooth muscle cytodifferentiation. Oral Surg Oral Med Oral Pathol, 1983, 56: 512–20., DOI 10.1016/0030-4220(83)90099-3 Takahashi H, Fujita S, Satoh H, Okabe H. Immunohistochemical study of congenital granular cell tumor, congenital epulis). J Oral Pathol, 1990, 19:492–6. Ugras S, Demmirtas I, Beckerecioglu M, Kutluhan A, Karakok M, Peker O. Immunohistochemical study on histogenesis of congenital epulis and review of the literature. Pathol Int, 1997, 47:627–32., DOI 10.1111/j.1440-1827.1997.tb04553.x Hiradfar M, Zabolinejad N, Gharavi M, Sebt S. Multiple congenital epulis of the mandibular ridge: a case report. Iran J Otorhinolaryngol, 2012, 24(69):193–6. Leocata P, Bifaretti G, Saltarelli S, Corbacelli A, Ventura L. Congenital, granular cell, epulis of the newborn: a case report with immunohistochemical study on the histogenesis. Ann Saudi Med, 1999, 19(6):527–9., DOI 10.5144/0256- 4947.1999.527 Liang Y, Yang YS1, Zhang Y. Multiple congenital granular cell epulis in a female newborn: a case report. J Med Case Rep, 2014, 8:413., DOI 10.1186/1752- 1947-8-413 Kato H, Nomura J, Matsumura Y, Yanase S, Nakanishi K, Tagawa T. A case of congenital granular cell epulis in the maxillary anterior ridge: a study of cell proliferation using immunohistological staining. Maxillofac Oral Surg, 2013, 12(3): 333–7., DOI 10.1007/s12663-011-0248-3 Ritwik P, Brannon RB, Musselman RJ. Spontaneous regression of congenital epulis: a case report and review of the literature. JMed Case Rep, 2010, 4:331., DOI 10. 1186/1752-1947-4-331 Dhareula A, Jaiswal M, Goyal A, Gauba K. Congenital granular cell tumor of the newborn - spontaneous regression or early surgical intervention. J Indian Soc Pedod Prev Dent, 2018, 36(3):319–23., DOI 10.4103/JISPPD.JISPPD_ 1187_17 Diniz MB, Giro Elisa MA, Zuanon Angela CC, Costa CA, Hebling J. Congenital epulis: a rare benign tumor in the newborn. J Indian Soc Pedod Prev Dent, 2010, 28(3):230–3., DOI 10.4103/0970-4388.73787 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611834 EP 1611841 DI 10.3389/pore.2024.1611834 PG 8 ER PT J AU Casanova, J da Costa, A Lopes, PA Catarino, A Nave, M Sousa, CA Lima, J AF Casanova, Joao da Costa, G. Ana Lopes, Pestana Ana Catarino, Ana Nave, Monica Sousa, Carla Ana Lima, Jorge TI Molecular classification of endometrial cancer: preliminary experience from a single Portuguese academic center SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE endometrial cancer; molecular subtypes; molecular profiling; POLE mutation; DNA sequencing ID endometrial cancer; molecular subtypes; molecular profiling; POLE mutation; DNA sequencing AB Background: Since the seminal publication of the TCGA consortium in 2013, the molecular classification of endometrial cancer has been widely accepted as a new and powerful tool to better understand the natural history of this malignancy. Adoption of routine molecular classification around the world has been limited. We sought to demonstrate our initial experience in incorporating the four molecular subtypes for endometrioid carcinomas. Methods: This was a retrospective analysis at a single center in Portugal. Molecular classification was determined using immunohistochemical staining for MMR and p53 and Sanger Sequencing to determine POLE mutation status as per published PROMISE method. Descriptive statistics were reported. Results: 20 patients with endometrioid histology were included. Median age of the cohort was 64 years (range 45–76). Median Body Mass Index (kg/m2) was 29.81 (range 21.3–43.1). In terms of tumor grading, 16 (80%) of the endometrial carcinomas of the cohort were low-grade (either grade 1 or grade 2). 16 (80%) of the cases were FIGO stage I. Regarding the molecular classification the tumors were classified as: MMRd [n = 6 (30%)]; p53 abn [n = 2 (10%)]; NSMP (n = 10 (50%)), POLE ultramut [n = 2 (10%)]. Conclusion: Despite the small sample size, we were able to show that molecular classification is feasible. To our knowledge this is the first cohort of endometroid endometrial carcinomas fully characterized according to the TCGA classification in Portugal, from one single center. C1 [Casanova, Joao] Hospital da Luz Lisboa, Department of Surgery, Obstetrics and Gynecology ServiceLisbon, Portugal. [da Costa, G. Ana] Hospital da Luz Lisboa, Department of Surgery, Obstetrics and Gynecology ServiceLisbon, Portugal. [Lopes, Pestana Ana] Hospital da Luz Lisboa, Department of Pathology, Gynecologic Oncology UnitLisbon, Portugal. [Catarino, Ana] Hospital da Luz Lisboa, Department of Pathology, Gynecologic Oncology UnitLisbon, Portugal. [Nave, Monica] Hospital da Luz Lisboa, Department of Oncology, Gynecologic Oncology UnitLisbon, Portugal. [Sousa, Carla Ana] GenoMed - Diagnosticos de Medicina MolecularLisbon, Portugal. [Lima, Jorge] Hospital da Luz Lisboa, Department of Surgery, Obstetrics and Gynecology ServiceLisbon, Portugal. RP Lima, J (reprint author), Hospital da Luz Lisboa, Department of Surgery, Obstetrics and Gynecology Service, Lisbon, Portugal. EM jorgeramoslima@sapo.pt CR Lu KH, Broaddus RR. Endometrial cancer. N Engl J Med, 2020, 383(21): 2053–64. From NLM Medline., DOI 10.1056/NEJMra1514010 Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecol Oncol, 1983, 15(1):10–7. From NLM., DOI 10.1016/0090-8258(83)90111-7 Hanby AM, walker C, Tavassoli FA, Devilee P. Tavassoli FA, devilee P: Pathology and genetics: tumours of the breast and female genital organs. WHO classification of tumours series - volume IV. Lyon, France: IARC press: 2003. 250pp. ISBN 92 832 2412 4. Breast Cancer Res, 2004, 6(3):133., DOI 10.1186/bcr788 Cancer Genome Atlas Research N, Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated genomic characterization of endometrial carcinoma. Nature, 2013, 497(7447):67–73. From NLM Medline., DOI 10.1038/ nature12113 Henry CE, Phan K, Orsman EJ, Kenwright D, Thunders MC, Filoche SK. Molecular profiling of endometrial cancer: an exploratory study in aotearoa, New Zealand. Cancers, Basel,, 2021, 13(22):5641. From NLM PubMed-not- MEDLINE., DOI 10.3390/cancers13225641 Kommoss S, McConechy MK, Kommoss F, Leung S, Bunz A, Magrill J, et al. Final validation of the ProMisE molecular classifier for endometrial carcinoma in a large population-based case series. Ann Oncol, 2018, 29(5):1180–8. From NLM Medline., DOI 10.1093/annonc/mdy058 Leon-Castillo A, Britton H, McConechy MK, McAlpine JN, Nout R, Kommoss S, et al. Interpretation of somatic POLE mutations in endometrial carcinoma. J Pathol, 2020, 250(3):323–35. From NLM., DOI 10.1002/path.5372 Leon-Castillo A, de Boer SM, Powell ME, Mileshkin LR, Mackay HJ, Leary A, et al. Molecular classification of the PORTEC-3 trial for high-risk endometrial cancer: impact on prognosis and benefit from adjuvant therapy. J Clin Oncol, 2020, 38(29):3388–97. From NLM Medline., DOI 10.1200/JCO.20.00549 Berek JS, Matias-Guiu X, Creutzberg C, Fotopoulou C, Gaffney D, Kehoe S, et al. FIGO staging of endometrial cancer: 2023. Int J Gynaecol Obstet, 2023, 162(2): 383–94. From NLM Medline., DOI 10.1002/ijgo.14923 Casanova J, Duarte GS, da Costa AG, Catarino A, Nave M, Antunes T, et al. Prognosis of polymerase epsilon, POLE, mutation in high-grade endometrioid endometrial cancer: systematic review andmeta-analysis. Gynecol Oncol, 2024, 182: 99–107. From NLM Publisher., DOI 10.1016/j.ygyno.2024.01.018 Imboden S, Nastic D, Ghaderi M, Rydberg F, Rau TT, Mueller MD, et al. Phenotype of POLE-mutated endometrial cancer. PLoS One, 2019, 14(3):e0214318. From NLM Medline., DOI 10.1371/journal.pone.0214318 Church DN, Stelloo E, Nout RA, Valtcheva N, Depreeuw J, ter Haar N, et al. Prognostic significance of POLE proofreading mutations in endometrial cancer. J Natl Cancer Inst, 2015, 107(1):402. From NLM Medline., DOI 10.1093/ jnci/dju402 Stelloo E, Bosse T, Nout RA, MacKay HJ, Church DN, Nijman HW, et al. Refining prognosis and identifying targetable pathways for high-risk endometrial cancer; a TransPORTEC initiative. Mod Pathol, 2015, 28(6):836–44. From NLM Medline., DOI 10.1038/modpathol.2015.43 Stelloo E, Nout RA, Osse EM, Jurgenliemk-Schulz IJ, Jobsen JJ, Lutgens LC, et al. Improved risk assessment by integrating molecular and clinicopathological factors in early-stage endometrial cancer-combined analysis of the PORTEC cohorts. Clin Cancer Res, 2016, 22(16):4215–24. From NLM Medline., DOI 10. 1158/1078-0432.CCR-15-2878 McMeekin DS, Tritchler DL, Cohn DE, Mutch DG, Lankes HA, Geller MA, et al. Clinicopathologic significance of mismatch repair defects in endometrial cancer: an NRG oncology/gynecologic oncology group study. J Clin Oncol, 2016, 34(25):3062–8. From NLM Medline., DOI 10.1200/JCO.2016.67.8722 Black D, Soslow RA, Levine DA, Tornos C, Chen SC, Hummer AJ, et al. Clinicopathologic significance of defective DNA mismatch repair in endometrial carcinoma. J Clin Oncol, 2006, 24(11):1745–53. From NLM Medline., DOI 10.1200/ JCO.2005.04.1574 Zighelboim I, Goodfellow PJ, Gao F, Gibb RK, Powell MA, Rader JS, et al. Microsatellite instability and epigenetic inactivation of MLH1 and outcome of patients with endometrial carcinomas of the endometrioid type. J Clin Oncol, 2007, 25(15):2042–8. From NLM Medline., DOI 10.1200/JCO.2006.08.2107 Kato M, Takano M, Miyamoto M, Sasaki N, Goto T, Tsuda H, et al. DNA mismatch repair-related protein loss as a prognostic factor in endometrial cancers. J Gynecol Oncol, 2015, 26(1):40–5. From NLM Medline., DOI 10.3802/jgo.2015.26.1.40 Vermij L, Leon-Castillo A, Singh N, Powell ME, Edmondson RJ, Genestie C, et al. p53 immunohistochemistry in endometrial cancer: clinical and molecular correlates in the PORTEC-3 trial. Mod Pathol, 2022, 35(10):1475–83. From NLM Medline., DOI 10.1038/s41379-022-01102-x Kasius JC, Pijnenborg JMA, Lindemann K, Forsse D, van Zwol J, Kristensen GB, et al. Risk stratification of endometrial cancer patients: FIGO stage, biomarkers and molecular classification. Cancers, Basel,, 2021, 13(22):5848. From NLM PubMed-not-MEDLINE., DOI 10.3390/cancers13225848 van den Heerik A, Horeweg N, Nout RA, Lutgens L, van der Steen-Banasik EM, Westerveld GH, et al. PORTEC-4a: international randomized trial of molecular profilebased adjuvant treatment for women with high-intermediate risk endometrial cancer. Int J Gynecol Cancer, 2020, 30(12):2002–7. From NLM., DOI 10.1136/ijgc-2020-001929 Concin N,Matias-Guiu X, Vergote I, Cibula D,Mirza MR,Marnitz S, et al. ESGO/ ESTRO/ESP guidelines for the management of patients with endometrial carcinoma. Int JGynecol Cancer, 2021, 31(1):12–39. FromNLMMedline., DOI 10.1136/ijgc-2020-002230 Billingsley CC, Cohn DE, Mutch DG, Hade EM, Goodfellow PJ. Prognostic significance of POLE exonuclease domain mutations in high-grade endometrioid endometrial cancer on survival and recurrence: a subanalysis. Int J Gynecol Cancer, 2016, 26(5):933–8. From NLM Medline., DOI 10.1097/IGC.0000000000000681 Bosse T, Nout RA, McAlpine JN, McConechy MK, Britton H, Hussein YR, et al. Molecular classification of grade 3 endometrioid endometrial cancers identifies distinct prognostic subgroups. Am J Surg Pathol, 2018, 42(5):561–8. From NLM Medline., DOI 10.1097/PAS.0000000000001020 Meng B, Hoang LN, McIntyre JB, Duggan MA, Nelson GS, Lee CH, et al. POLE exonuclease domain mutation predicts long progression-free survival in grade 3 endometrioid carcinoma of the endometrium. Gynecol Oncol, 2014, 134(1): 15–9. From NLM Medline., DOI 10.1016/j.ygyno.2014.05.006 Yu S, Sun Z, Zong L, Yan J, Yu M, Chen J, et al. Clinicopathological and molecular characterization of high-grade endometrial carcinoma with POLE mutation: a single center study. J Gynecol Oncol, 2022, 33(3):e38. From NLM Medline., DOI 10.3802/jgo.2022.33.e38 Zong L, Mo S, Sun Z, Lu Z, Chen J, Yu S, et al. Incorporating molecular classification when stratifying the survival risk of patients with high-grade endometrial carcinomas. J Clin Med, 2023, 12(2):530. From NLM PubMed-not- MEDLINE., DOI 10.3390/jcm12020530 Talhouk A, McConechy MK, Leung S, Yang W, Lum A, Senz J, et al. Confirmation of ProMisE: a simple, genomics-based clinical classifier for endometrial cancer. Cancer, 2017, 123(5):802–13. From NLM Medline., DOI 10. 1002/cncr.30496 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611835 EP 1611842 DI 10.3389/pore.2024.1611835 PG 8 ER PT J AU Bicsko, RR Nyilas, R Szasz, R Varoczy, L Kiss, A Udvardy, M Illes, Gergely, L AF Bicsko, Rahel Reka Nyilas, Renata Szasz, Robert Varoczy, Laszlo Kiss, Attila Udvardy, Miklos Illes, Arpad Gergely, Lajos TI The efficacy and safety of second salvage autologous transplantation in myeloma patients SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE multiple myeloma; salvage therapy; salvage transplantation; second autologous stem cell transplantation; survival ID multiple myeloma; salvage therapy; salvage transplantation; second autologous stem cell transplantation; survival AB Despite the availability of many novel therapies for multiple myeloma, it remains an incurable disease with relapse fated in almost all patients. In the era of modern agents, second autologous stem cell transplantation still holds its role in patients relapsing after first-line autologous transplant. The authors reviewed a single-center experience with a second auto-SCT for relapsed multiple myeloma. Thirty patients had received a salvage auto-SCT at the institution. The median follow-up after diagnosis was 86 months, and the median time between transplants was 59.1 months. Response before second ASCT was the following: CR – 11 cases, VGPR – 9 cases, PR – 10 cases. Most patients received reduced dose (140 mg/m2) of melphalan as a conditioning regimen for the second auto-SCT. Treatment-related mortality was 3%. With a median followup time of 34 months after the second transplant, median progression-free survival was 24 months. The median PFS in the patients achieving CR or VGPR at day 100 after the second transplantation was 32 months. By 15 months, all patients achieved only partial remission progressed, with a median PFS of 8.5 months. During the follow-up period, no MDS or AML developed, and the frequency of second malignancy was also low, 3%. In conclusion, second autologous stem cell transplantation is a well-tolerated and effective treatment option for relapsed multiple myeloma in selected patients, though with a shorter PFS than in first remission. C1 [Bicsko, Rahel Reka] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Nyilas, Renata] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Szasz, Robert] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Varoczy, Laszlo] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Kiss, Attila] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Udvardy, Miklos] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Illes, Arpad] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. [Gergely, Lajos] University of Debrecen, Faculty of Medicine, Department of HematologyDebrecen, Hungary. RP Bicsko, RR (reprint author), University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen, Hungary. EM bicskorekaa@gmail.com CR Barlogie B, Gahrton G. Bone marrow transplantation in multiple myeloma. Bone Marrow Transpl, 1991, 7:71–9. Moreau P, Misbahi R, Milpied N, Morineau N, Mahe B, Vigier M, et al. Longterm results, 12 years, of high-dose therapy in 127 patients with de novo multiple myeloma. Leukemia, 2002, 16:1838–43., DOI 10.1038/sj.leu.2402613 Lahuerta JJ, Grande C, Martinez-Lopez J, De La Serna J, Toscano R, Ortiz MC, et al. Tandem transplants with different high-dose regimens improve the complete remission rates in multiple myeloma. Results of a Grupo Espanol de Sindromes Linfoproliferativos/Trasplante Autologo de Medula Osea phase II trial. Br J Haematol, 2003, 120:296–303., DOI 10.1046/j.1365-2141.2003.04067.x Krivanova A, Hajek R, Krejci M, Scudla V, Indrak K, Bacovsky J, et al. Second autologous transplantation for multiple myeloma patients relapsing after the first autograft - a pilot study for the evaluation of experimental maintenance therapies. Report of the prospective non-randomized pilot study of the Czech Myeloma Group. Onkologie, 2004, 27:275–9., DOI 10.1159/000077977 Al Hadidi S, Ababneh O, Schinke C, Thanendrarajan S, Bailey C, Smith RT, et al. Three yearsmaintenance with VRD in multiplemyeloma: results of total therapy IIIB with a 15-year follow up. Blood Adv, 2023, 8:703–7., DOI 10.1182/bloodadvances.2023011601 Qazilbash MH, Saliba R, De Lima M, Hosing C, Couriel D, Aleman A, et al. Second autologous or allogeneic transplantation after the failure of first autograft in patients with multiple myeloma. Cancer, 2006, 106:1084–9., DOI 10.1002/cncr.21700 Olin RL, Vogl DT, Porter DL, Luger SM, Schuster SJ, Tsai DE, et al. Second auto-SCT is safe and effective salvage therapy for relapsed multiple myeloma. Bone Marrow Transpl, 2009, 43:417–22., DOI 10.1038/bmt.2008.334 Jimenez-Zepeda VH, Mikhael J, Winter A, Franke N, Masih-Khan E, Trudel S, et al. Second autologous stem cell transplantation as salvage therapy for multiple myeloma: impact on progression-free and overall survival. Biol Blood Marrow Transpl, 2012, 18:773–9., DOI 10.1016/j.bbmt.2011.10.044 Auner HW, Szydlo R, Rone A, Chaidos A, Giles C, Kanfer E, et al. Salvage autologous stem cell transplantation for multiple myeloma relapsing or progressing after up-front autologous transplantation. Leuk Lymphoma, 2013, 54:2200–4., DOI 10.3109/10428194.2013.773998 Grovdal M, Nahi H, Gahrton G, Liwing J, Waage A, Abildgaard N, et al. Autologous stem cell transplantation versus novel drugs or conventional chemotherapy for patients with relapsed multiple myeloma after previous ASCT. Bone Marrow Transpl, 2015, 50:808–12., DOI 10.1038/bmt.2015.39 Lemieux C, Muffly LS, Iberri DJ, Craig JK, Johnston LJ, Lowsky R, et al. Outcomes after delayed and second autologous stem cell transplant in patients with relapsed multiple myeloma. Bone Marrow Transpl, 2021, 56:2664–71., DOI 10.1038/ s41409-021-01371-1 Kumar S, Paiva B, Anderson KC, Durie B, Landgren O, Moreau P, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol, 2016, 17:e328–46., DOI 10.1016/S1470-2045(16)30206-6 Cook G, Liakopoulou E, Pearce R, Cavet J, Morgan GJ, Kirkland K, et al. Factors influencing the outcome of a second autologous stem cell transplant, ASCT, in relapsed multiple myeloma: a study from the British Society of Blood and Marrow Transplantation Registry. Biol Blood Marrow Transpl, 2011, 17:1638–45., DOI 10.1016/j.bbmt.2011.04.005 Hagen PA, Stiff P. The role of salvage second autologous hematopoietic cell transplantation in relapsed multiple myeloma. Biol Blood Marrow Transpl, 2019, 25:e98–e107., DOI 10.1016/j.bbmt.2018.12.002 Dhakal B, D’Souza A, Kleman A, Chhabra S, Mohan M, Hari P. Salvage second transplantation in relapsed multiple myeloma. Leukemia, 2021, 35(4): 1214–7., DOI 10.1038/s41375-020-1005-8 Galligan D, Williamson S,Myers J, Silbermann R,Medvedova E, Nagle S, et al. Second autologous stem cell transplant as salvage in multiple myeloma - the Oregon health and science university experience. Clin Lymphoma Myeloma Leuk, 2022, 22: 105–12., DOI 10.1016/j.clml.2021.08.008 Pasvolsky O, Yeshurun M, Fraser R, Estrada-Merly N, Rozovski U, Shargian- Alon L, et al. Maintenance therapy after second autologous hematopoietic cell transplantation for multiple myeloma. A CIBMTR analysis. Bone Marrow Transpl, 2022, 57:31–7., DOI 10.1038/s41409-021-01455-y Fermand JP, Katsahian S, Divine M, Leblond V, Dreyfus F, Macro M, et al. High-dose therapy and autologous blood stem-cell transplantation compared with conventional treatment in myeloma patients aged 55 to 65 years: long-term results of a randomized control trial from the Group Myelome-Autogreffe. J Clin Oncol, 2005, 23:9227–33., DOI 10.1200/JCO.2005. 03.0551 Cavo M, Tosi P, Zamagni E, Cellini C, Tacchetti P, Patriarca F, et al. Prospective, randomized study of single compared with double autologous stem-cell transplantation for multiple myeloma: bologna 96 clinical study. J Clin Oncol, 2007, 25:2434–41., DOI 10.1200/JCO.2006.10.2509 Mehta J. One or two autografts for myeloma. Blood, 2008, 111(7):3899–900., DOI 10.1182/blood-2007-12-127704 Ferment B, Lambert J, Caillot D, Lafon I, Karlin L, Lazareth A, et al. French early nationwide idecabtagene vicleucel chimeric antigen receptor T-cell therapy experience in patients with relapsed/refractory multiple myeloma, FENIX): a realworld IFM study from the DESCAR-T registry. Br J Haematol, 2024)., DOI 10.1111/ bjh.19505 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611851 EP 1611857 DI 10.3389/pore.2024.1611851 PG 7 ER PT J AU Lakatos, L Illyes, I Budai, A Bencze, V Szijarto, A Kiss, A Banky, B AF Lakatos, Lorand Illyes, Ildiko Budai, Andras Bencze, Viktoria Szijarto, Attila Kiss, Andras Banky, Balazs TI Feasibility of indocyanine green (ICG) fluorescence in ex vivo pathological dissection of colorectal lymph nodes—a pilot study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE indocyanine green; colorectal cancer; lymph node; ex vivo; colorectal resection ID indocyanine green; colorectal cancer; lymph node; ex vivo; colorectal resection AB Accurate lymph node (LN) retrieval during colorectal carcinoma resection is pivotal for precise N-staging and the determination of adjuvant therapy. Current guidelines recommend the examination of at least 12 mesocolic or mesorectal lymph nodes for accurate staging. Traditional histological processing techniques, reliant on visual inspection and palpation, are time-consuming and heavily dependent on the examiner’s expertise and availability. Various methods have been documented to enhance LN retrieval from colorectal specimens, including intra-arterial ex vivo methylene blue injection. Recent studies have explored the utility of indocyanine green (ICG) fluorescence imaging for visualizing pericolic lymph nodes and identifying sentinel lymph nodes in colorectal malignancies. This study included 10 patients who underwent colon resection for malignant tumors. During surgery, intravenous ICGdye and an endoscopic camerawere employed to assess intestinal perfusion. Post-resection, ex vivo intra-arterial administration of ICG dye was performed on the specimens, followed by routine histological processing and an ICG-assisted lymph node dissection. The objective was to evaluatewhether ICG imaging could identify additional lymph nodes compared to routine manual dissection and to assess the clinical relevance of these findings. For each patient, a minimum of 12 lymph nodes (median = 25.5, interquartile range = 12.25, maximum = 33) were examined. ICG imaging facilitated the detection of a median of three additional lymph nodes not identified during routine processing. Metastatic lymph nodes were found in four patients however no additional metastatic nodes were detected with ICG assistance. Our findings suggest that ex vivo intra-arterial administration of indocyanine green dye can augment lymph node dissection, particularly in caseswhere the number of lymph nodes retrieved is below the recommended threshold of 12. C1 [Lakatos, Lorand] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Illyes, Ildiko] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Budai, Andras] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Bencze, Viktoria] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Szijarto, Attila] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Kiss, Andras] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Banky, Balazs] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. RP Lakatos, L (reprint author), Semmelweis University, Department of Transplantation and Surgery, Budapest, Hungary. EM lakatoslorandl@gmail.com CR Kim J, Huynh R, Abraham I, Kim E, Kumar RR. Number of lymph nodes examined and its impact on colorectal cancer staging. The Am surgeon, 2006, 72(10):902–5., DOI 10.1177/000313480607201013 Markl B, Roessle J, Arnholdt HM, Schaller T, Krammer I, Cacchi C, et al. The clinical significance of lymph node size in colon cancer. Mod Pathol, 2012, 25: 1413–22., DOI 10.1038/modpathol.2012.92 Destri GL, Di Carlo I, Scilletta R, Scilletta B, Puleo S. Colorectal cancer and lymph nodes: the obsession with the number 12. World J Gastroenterol WJG, 2014, 20(8):1951–60., DOI 10.3748/wjg.v20.i8.1951 Borowski DW, Banky B, Banerjee AK, Agarwal AK, Tabaqchali MA, Garg DK, et al. Intra-arterial methylene blue injection into ex vivo colorectal cancer specimens improves lymph node staging accuracy: a randomized controlled trial. Colorectal Dis, 2014, 16(9):681–9. PMID: 24911342., DOI 10.1111/codi.12681 Gregurek SF, Wu HH. Can GEWF solution improve the retrieval of lymph nodes from colorectal cancer resections? Arch Pathol Lab Med, 2009, 133(1):83–6. PMID: 19123742., DOI 10.1043/1543-2165-133.1.83 Schofield JB,Mounter NA, Mallett R, Haboubi NY. The importance of accurate pathological assessment of lymph node involvement in colorectal cancer. Colorectal Dis, 2006, 8(6):460–70., DOI 10.1111/j.1463-1318.2006.01044.x Chand M, Keller DS, Joshi HM, Devoto L, Rodriguez-Justo M, Cohen R. Feasibility of fluorescence lymph node imaging in colon cancer: FLICC. Tech Coloproctol, 2018, 22(4):271–7. Epub 2018 Mar 17. PMID: 29551004., DOI 10.1007/ s10151-018-1773-6 Sposito C, Maspero M, Belotti P, Simonotti N, Altomare M, Ciana P, et al. Indocyanine green fluorescence-guided surgery for gastrointestinal tumors: a systematic review. Ann Surg Open, 2022, 3(3):e190. PMID: 37601143; PMCID: PMC10431291., DOI 10.1097/AS9.0000000000000190 Watanabe J, OtaM, Suwa Y, Suzuki S, Suwa H,MomiyamaM, et al. Evaluation of the intestinal blood flow near the rectosigmoid junction using the indocyanine green fluorescence method in a colorectal cancer surgery. Int J colorectal Dis, 2015, 30:329–35., DOI 10.1007/s00384-015-2129-6 Markl B, Arnholdt HM, Jahnig H, Spatz H, Anthuber M, Oruzio DV, et al. A new concept for the role of ex vivo sentinel lymph nodes in node-negative colorectal cancer. Ann Surg Oncol, 2010, 17:2647–55., DOI 10.1245/s10434-010-1030-3 Leiloglou M, Kedrzycki MS, Chalau V, Chiarini N, Thiruchelvam PT, Hadjiminas DJ, et al. Indocyanine green fluorescence image processing techniques for breast cancer macroscopic demarcation. Scientific Rep, 2022, 12(1):8607., DOI 10.1038/s41598-022-12504-x Fry DE. The Hawthorne effect revisited. Dis Colon Rectum, 2018, 61(1):6–7. PMID: 29219914., DOI 10.1097/DCR.0000000000000928 Edler D, Ohrling K, Hallstrom M, Karlberg M, Ragnhammar P. The number of analyzed lymph nodes–a prognostic factor in colorectal cancer. Acta Oncologica, 2007, 46(7):975–81., DOI 10.1080/02841860701203537 Keller DS, Berho M, Perez RO, Wexner SD, Chand M. The multidisciplinary management of rectal cancer. Nat Rev Gastroenterol Hepatol, 2020, 17(7):414–29., DOI 10.1038/s41575-020-0275-y Brouwer NP, Stijns RC, Lemmens VE, Nagtegaal ID, Beets-Tan RG, Futterer JJ, et al. Clinical lymph node staging in colorectal cancer; a flip of the coin? Eur J Surg Oncol, 2018, 44(8):1241–6., DOI 10.1016/j.ejso.2018.04.008 Rodriguez-Bigas MA, Maamoun S,Weber TK, Penetrante RB, Blumenson LE, Petrelli NJ. Clinical significance of colorectal cancer: metastases in lymph nodes< 5 mm in size. Ann Surg Oncol, 1996, 3:124–30., DOI 10.1007/BF02305790 Susztak N, Besznyak I, Almasi K, Bursics A, Kelemen D, Borowski DW, et al. Improved accuracy of lymph node staging and long-term survival benefit in colorectal cancer with ex vivo arterial methylene blue infiltration. Pathol Oncol Res, 2022, 28: 1610742. PMID: 36330051; PMCID: PMC9624224., DOI 10.3389/pore.2022.1610742 Tan L, Liu ZL, Ma Z, He Z, Tang LH, Liu YL, et al. Prognostic impact of at least 12 lymph nodes after neoadjuvant therapy in rectal cancer: a meta-analysis. World J Gastrointest Oncol, 2020, 12(12):1443–55. PMID: 33362914; PMCID: PMC7739152., DOI 10.4251/wjgo.v12.i12.1443 Lykke J, Jess P, Roikjaer O, Danish Colorectal Cancer Group. Increased lymph node yield is associated with improved survival in rectal cancer irrespective of neoadjuvant treatment: results from a national cohort study. Dis Colon Rectum, 2015, 58:823–30., DOI 10.1097/DCR.0000000000000429 Parnaby CN, Scott NW, Ramsay G, MacKay C, Samuel L, Murray GI, et al. Prognostic value of lymph node ratio and extramural vascular invasion on survival for patients undergoing curative colon cancer resection. Br J Cancer, 2015, 113(2):212–9. Epub 2015 Jun 16. PMID: 26079302; PMCID: PMC4506392., DOI 10.1038/bjc.2015.211 Pathology & NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611853 EP 1611859 DI 10.3389/pore.2024.1611853 PG 7 ER PT J AU Deme, D Tamaskovics, FB Jammoul, N Kovacs, S Kayode, OK Grice, J Telekes, A AF Deme, Daniel Tamaskovics, Ferenc Balint Jammoul, Nizar Kovacs, Sandor Kayode, Oladunjoye Kmmanuel Grice, W. James Telekes, Andras TI Corrigendum: Association between pathological characteristics and recurrence score by OncotypeDX in resected T1-3 and N0-1 breast cancer: a real-life experience of a North Hungarian regional center SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Technical Report DE breast cancer; pathological characteristics; OncotypeDX; recurrence score; observation oriented modeling ID breast cancer; pathological characteristics; OncotypeDX; recurrence score; observation oriented modeling AB A Corrigendum on Association between pathological characteristics and recurrence score by OncotypeDX in resected T1-3 and N0-1 breast cancer: a real-life experience of a North Hungarian regional center by Deme D, Tamaskovics BF, Jammoul N, Kovacs S, Kayode EO, Grice JWand Telekes A (2024). Pathol. Oncol. Res. 30:1611735. doi: 10.3389/pore.2024.1611735 C1 [Deme, Daniel] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. [Tamaskovics, Ferenc Balint] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. [Jammoul, Nizar] Szent Lazar Megyei Korhaz, Patologiai OsztalySalgotarjan, Hungary. [Kovacs, Sandor] University of Debrecen, Department of Economical and Financial MathematicsDebrecen, Hungary. [Kayode, Oladunjoye Kmmanuel] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. [Grice, W. James] Oklahoma State University, Department of PsychologyStillwater, OK, USA. [Telekes, Andras] Szent Lazar Megyei Korhaz, Onkologiai OsztalySalgotarjan, Hungary. RP Deme, D (reprint author), Szent Lazar Megyei Korhaz, Onkologiai Osztaly, Salgotarjan, Hungary. EM danieldeme_md@ymail.com NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611862 EP 1611865 DI 10.3389/pore.2024.1611862 PG 4 ER PT J AU Gascard, P Wang, X Nosrati, M Kim, K Kashani-Sabet, M Tlsty, Th Leong, S Hendrix, M AF Gascard, D. Philippe Wang, Xianhong Nosrati, Mehdi Kim, B. Kevin Kashani-Sabet, Mohammed Tlsty, D. Thea Leong, P. Stanley Hendrix, J. C. Mary TI Higher Nodal expression is often associated with poorer survival in patients diagnosed with melanoma and treated with anti-PD1 therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE melanoma; nodal; anti-PD1 therapy; immune checkpoint inhibitors; multiplex immunohistochemistry ID melanoma; nodal; anti-PD1 therapy; immune checkpoint inhibitors; multiplex immunohistochemistry AB Advanced melanoma is considered the most aggressive and deadly form of skin cancer whose incidence has been rising over the past three decades. In the absence of treatment, the median overall survival for advanced-stage metastatic disease is less than 6 months. Although most melanomas detected at an early stage can be cured with surgery, a subset of these eventually metastasize. Therefore, a critical need exists to identify unique molecular features that would be predictive of long-term outcome and response to specific therapies. Recent promising therapeutic regimens have included the use of immune checkpoint inhibitors, such as anti-PD1 antibodies. However, the ability to identify responders and non-responders to this therapy remains elusive. To address this challenge at the molecular level, previously our laboratory identified the emergence of a stem cell phenotype associated with advanced melanoma and other aggressive forms of cancer. Underlying this phenotype is the aberrant re-expression of the embryonic morphogen “Nodal”. Particularly noteworthy, we have observed Nodal to remain in advanced tumors of non-responders to standard-of-care therapies (i.e., BRAFi). This pilot study is the first proof-of-principle attempt to predict treatment response survival outcome in a small cohort of melanoma patients receiving anti-PD1 immune checkpoint inhibitor therapy – based on their Nodal expression profile. Using advanced multiplex immunohistochemistry-based digital pathology, the major finding of this preliminary study indicates that higher Nodal expression is often associated with poorer overall survival after anti-PD1 therapy, reaching nearly statistical relevance. C1 [Gascard, D. Philippe] University of California San Francisco, Department of PathologySan Francisco, CA, USA. [Wang, Xianhong] University of California San Francisco, Department of PathologySan Francisco, CA, USA. [Nosrati, Mehdi] Sutter Health, Center for Melanoma Research and Treatment, California Pacific Medical CenterSan Francisco, CA, USA. [Kim, B. Kevin] Sutter Health, Center for Melanoma Research and Treatment, California Pacific Medical CenterSan Francisco, CA, USA. [Kashani-Sabet, Mohammed] Sutter Health, Center for Melanoma Research and Treatment, California Pacific Medical CenterSan Francisco, CA, USA. [Tlsty, D. Thea] University of California San Francisco, Department of PathologySan Francisco, CA, USA. [Leong, P. Stanley] Sutter Health, Center for Melanoma Research and Treatment, California Pacific Medical CenterSan Francisco, CA, USA. [Hendrix, J. C. Mary] Shepherd University, Department of BiologyShepherdstown, WV, USA. RP Hendrix, M (reprint author), Shepherd University, Department of Biology, Shepherdstown, USA. EM mhendrix@shepherd.edu CR American Cancer Society. Key statistics for melanoma skin cancer, 2023). Garg M, Couturier DL, Nsengimana J, Fonseca NA, Wongchenko M, Yan Y, et al. Tumour gene expression signature in primary melanoma predicts long-term outcomes. Nat Commun, 2021, 12(1):1137., DOI 10.1038/s41467-021-21207-2 Netanely D, Leibou S, Parikh R, Stern N, Vaknine H, Brenner R, et al. Classification of node-positive melanomas into prognostic subgroups using keratin, immune, and melanogenesis expression patterns. Oncogene, 2021, 40(10):1792–805., DOI 10.1038/s41388-021-01665-0 Whitman ED, Koshenkov VP, Gastman BR, Lewis D, Hsueh EC, Pak H, et al. Integrating 31-gene expression profiling with clinicopathologic features to optimize cutaneous melanoma sentinel lymph node metastasis prediction. JCO Precis Oncol, 2021, 5:1466–79., DOI 10.1200/PO.21.00162 Karapetyan L, Gooding W, Li A, Yang X, Knight A, Abushukair HM, et al. Sentinel lymph node gene expression signature predicts recurrence-free survival in cutaneous melanoma. Cancers, Basel,, 2022, 14(20):4973., DOI 10.3390/ cancers14204973 Shui IM, Scherrer E, Frederickson A, Li JW, Mynzhassarova A, Druyts E, et al. Resistance to anti-PD1 therapies in patients with advanced melanoma: systematic literature review and application of the Society for Immunotherapy of Cancer Immunotherapy Resistance Taskforce anti-PD1 resistance definitions. Melanoma Res, 2022, 32(6):393–404., DOI 10.1097/CMR.0000000000000850 Bittner M, Meltzer P, Chen Y, Jiang Y, Seftor E, Hendrix M, et al. Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature, 2000, 406(6795):536–40., DOI 10.1038/35020115 Topczewska JM, Postovit LM, Margaryan NV, Sam A, Hess AR, Wheaton WW, et al. Embryonic and tumorigenic pathways converge via Nodal signaling: role in melanoma aggressiveness. Nat Med, 2006, 12(8):925–32., DOI 10.1038/nm1448 Postovit LM, Margaryan NV, Seftor EA, Hendrix MJ. Role of nodal signaling and the microenvironment underlyingmelanoma plasticity. Pigment Cel Melanoma Res, 2008, 21(3):348–57., DOI 10.1111/j.1755-148X.2008.00463.x Strizzi L, Postovit LM, Margaryan NV, Lipavsky A, Gadiot J, Blank C, et al. Nodal as a biomarker for melanoma progression and a new therapeutic target for clinical intervention. Expert Rev Dermatol, 2009, 4(1):67–78., DOI 10.1586/ 17469872.4.1.67 Yu L, Harms PW, Pouryazdanparast P, Kim DS,Ma L, Fullen DR. Expression of the embryonic morphogen Nodal in cutaneous melanocytic lesions. Mod Pathol, 2010, 23(9):1209–14., DOI 10.1038/modpathol.2010.101 Hendrix MJC, Seftor EA, Margaryan NV, Seftor REB. Heterogeneity and plasticity of melanoma: challenges of current therapies. Cutaneous melanoma: etiology and therapy. Brisbane, Australia: Codon Publications, 2017). Margaryan NV, Hazard-Jenkins H, Salkeni MA, Smolkin MB, Coad JA, Wen S, et al. The stem cell phenotype of aggressive breast cancer cells. Cancers, Basel,, 2019, 11(3):340., DOI 10.3390/cancers11030340 Strizzi L, Sandomenico A, Margaryan NV, Foca A, Sanguigno L, Bodenstine TM, et al. Effects of a novel Nodal-targeting monoclonal antibody in melanoma. Oncotarget, 2015, 6(33):34071–86., DOI 10.18632/oncotarget.6049 Hendrix MJ, Kandela I, Mazar AP, Seftor EA, Seftor RE, Margaryan NV, et al. Targeting melanoma with front-line therapy does not abrogate Nodal-expressing tumor cells. Lab Invest, 2017, 97(2):176–86., DOI 10.1038/labinvest.2016.107 Harms PW, Frankel TL, Moutafi M, Rao A, Rimm DL, Taube JM, et al. Multiplex immunohistochemistry and immunofluorescence: a practical update for pathologists. Mod Pathol, 2023, 36(7):100197., DOI 10.1016/j.modpat.2023.100197 Stack EC, Wang C, Roman KA, Hoyt CC. Multiplexed immunohistochemistry, imaging, and quantitation: a review, with an assessment of Tyramide signal amplification, multispectral imaging and multiplex analysis. Methods, 2014, 70(1):46–58., DOI 10.1016/j.ymeth.2014.08.016 Bankhead P, Loughrey MB, Fernandez JA, Dombrowski Y,McArt DG, Dunne PD, et al. QuPath: open source software for digital pathology image analysis. Sci Rep, 2017, 7(1):16878., DOI 10.1038/s41598-017-17204-5 Strizzi L, Hardy KM, Kirschmann DA, Ahrlund-Richter L, Hendrix MJ. Nodal expression and detection in cancer: experience and challenges. Cancer Res, 2012, 72(8):1915–20., DOI 10.1158/0008-5472.CAN-11-3419 Ning F,Wang HF, Guo Q, Liu ZC, Li ZQ, Du J. Expression and significance of Nodal in human cancers: a meta-analysis. Int J Clin Exp Med, 2015, 8(11): 20227–35. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann Oncol, 2019, 30(4):582–8., DOI 10.1093/ annonc/mdz011 Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med, 2019, 381(16):1535–46., DOI 10.1056/NEJMoa1910836 Robert C, RibasA, Schachter J, AranceA, Grob JJ,Mortier L, et al.Pembrolizumab versus ipilimumab in advanced melanoma, KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol, 2019, 20(9):1239–51., DOI 10.1016/S1470-2045(19)30388-2 Wolchok JD, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, et al. Long-term outcomes with nivolumab Plus ipilimumab or nivolumab alone versus ipilimumab in patients with advanced melanoma. J Clin Oncol, 2022, 40(2): 127–37., DOI 10.1200/JCO.21.02229 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611889 EP 1611896 DI 10.3389/pore.2024.1611889 PG 8 ER PT J AU Nagy, FZs Arvai, K Lakatos, P Beke Debreceni, I Szili, B Istenes, I Bodor, Cs Demeter, J AF Nagy, Flora Zsofia Arvai, Kristof Lakatos, Peter Beke Debreceni, Ildiko Szili, Balazs Istenes, Ildiko Bodor, Csaba Demeter, Judit TI Case report: Comprehensive clinical, pathological and genetic investigations to decipher the background of cyclic thrombocytopenia SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE case report; WES; cyclic thrombocytopaenia; thrombocytopaenia; cyclic ID case report; WES; cyclic thrombocytopaenia; thrombocytopaenia; cyclic AB Cyclic thrombocytopenia (CTP) is a rare disease characterized by the oscillations seen in the platelet count of the patients. The pathomechanism of the disease is poorly understood, several pathological processes have been implied in the background of CTP. In our current study, we aimed to thoroughly investigate the case of a 41-year-old female patient with a 22-year history of CTP. Wide-ranging laboratory testing, histological analyses and genetic investigations were carried out to investigate all the defects and alterations of physiological pathways described in the background of CTP to date. Bone marrow biopsy showed normal hemopoiesis with the abundance of megakaryocytes, some of which displayed hypolobulated nuclei. T-cell receptor rearrangement studies showed a polyclonal pattern with no indication of a monoclonal cell population. Flow cytometric assessment of the platelets revealed large number of immature platelets and decreased expression of glycoprotein IIb and IIIa at platelet zenith. Increased expression of glycoprotein IIb, IIIa and glycoprotein Ib-IX complex was observed at the nadir of the cycle. Whole exome sequencing revealed a heterozygous missense variant of uncertain significance in the SERPINC1 gene, which has been associated with hereditary antithrombin deficiency. The screening of autoantibodies did not reveal signs of autoreactive processes, and no thyroid dysfunction was found. Furthermore, synchronization with the menstrual cycle could not be concluded based on our patient’s case. With our results we contribute to the very limited data known about the long-term course of the disease and provide valuable insights into the genetic architecture of CTP. C1 [Nagy, Flora Zsofia] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Arvai, Kristof] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Lakatos, Peter] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Beke Debreceni, Ildiko] University of Debrecen, Faculty of Medicine, Department of Laboratory MedicineDebrecen, Hungary. [Szili, Balazs] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Istenes, Ildiko] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Bodor, Csaba] Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, HCEMM-SE Molecular Oncohematology Research GroupBudapest, Hungary. [Demeter, Judit] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. RP Nagy, FZs (reprint author), Semmelweis University, Department of Internal Medicine and Oncology, Budapest, Hungary. EM nagy.zsofia.flora@semmelweis.hu CR Oyenuga M, Onyechi A, Sartaj S, Patel R, Sinha J. Cyclic thrombocytopenia: a rare cause of recurrent thrombocytopenia case presentation. Cureus, 2022, 14:10–2., DOI 10.7759/cureus.22525 Arnold DM, Nazy I, Clare R, Jaffer AM, Aubie B, Li N, et al. Misdiagnosis of primary immune thrombocytopenia and frequency of bleeding: lessons from the McMaster ITP Registry. Blood Adv, 2017, 1:2414–20., DOI 10.1182/bloodadvances. 2017010942 Kyrle PA, Eichinger S. How I manage cyclic thrombocytopenia. Blood, 2021, 137:178–84., DOI 10.1182/blood.2020008218 Helleberg C, Taaning E, Hansen PB. Cyclic thrombocytopenia successfully treated with low dose hormonal contraception. Am J Hematol, 1995, 48:62–3., DOI 10.1002/ajh.2830480117 Dan K, Inokuchi K, An E, Nomura T. Cell-mediated cyclic thrombocytopenia treated with azathioprine. Br J Haematol, 1991, 77:365–70., DOI 10.1111/j.1365- 2141.1991.tb08585.x Neunert C, Terrell DR, Arnold DM, Buchanan G, Cines DB, Cooper N, et al. American Society of Hematology 2019 guidelines for immune thrombocytopenia. Blood Adv, 2019, 3:3829–66., DOI 10.1182/bloodadvances.2019000966 Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of medical genetics and genomics and the association for molecular Pathology. Genet Med, 2015, 17:405–24., DOI 10.1038/gim.2015.30 Steinbrecher O, Mitrovic M, Eischer L, Sinkovec H, Eichinger S, Kyrle PA. Clinical and laboratory characteristics of cyclic thrombocytopenia: an observational study. Haematologica, 2020, 105:198–201., DOI 10.3324/haematol.2019.237909 Chen G, Chen L, Qin X, Xie X, Li G, Xu B. Cyclic thrombocytopenia related to menstrual cycle: a case report and literature review. Int J Clin Exp Med, 2014, 7: 3595–8. Kuyama J, Take H, Matsumoto S, Nakajima T, Sugase T, Hata A, et al. Synchronous fluctuation of interleukin-6 and platelet count in cyclic thrombocytopenia and thrombocytosis. Intern Med, 1995, 34:636–9., DOI 10. 2169/internalmedicine.34.636 Nagasawa T, Hasegawa Y, Kamositha M, Ohtani K, Komeno T, Itoh T, et al. Megakaryopoiesis in patients with cyclic thrombocytopenia. Br J Haematol, 1995, 91:185–90., DOI 10.1111/j.1365-2141.1995.tb05267.x Yanabu M, Nomura S, Fukuroi T, Kawakatsu T, Kido H, Yamaguchi K, et al. Periodic production of antiplatelet autoantibody directed against gpiiia in cyclic thrombocytopenia. Acta Haematol, 1993, 89:155–9., DOI 10.1159/ 000204510 Fogarty PF, Stetler-Stevenson M, Pereira A, Dunbar CE. Large granular lymphocytic proliferation-associated cyclic thrombocytopenia. Am J Hematol, 2005, 79:334–6., DOI 10.1002/ajh.20375 Fureder W, Mitterbauer G, Thalhammer R, Geissler K, Panzer S, Krebs M, et al. Clonal T cell-mediated cyclic thrombocytopenia. Br J Haematol, 2002, 119: 1059–61., DOI 10.1046/j.1365-2141.2002.03951.x Kojima K, Fujii N, Omoto E, Nose S, Yoneyama M, Sugii Y, et al. Cyclic thrombocytopenia and polycythemia vera. Ann Hematol, 2003, 82:61–3., DOI 10. 1007/s00277-002-0580-2 Steensma D, Harrison C, Tefferi A. Hydroxyurea-associated platelet count oscillations in polycythemia vera: a report of four new cases and a review. Leuk Lymphoma, 2001, 42:1243–53., DOI 10.3109/10428190109097749 Lu Q, Hu Y, Sun J, Cheng Y, Cheung KH, Zhao H. A statistical framework to predict functional non-coding regions in the human genome through integrated analysis of annotation data. Sci Rep, 2015, 5:10576–13., DOI 10.1038/srep10576 Mourey L, Samama JP, Delarue M, Choay J, Lormeau JC, Petitou M, et al. Antithrombin III: structural and functional aspects. Biochimie, 1990, 72:599–608., DOI 10.1016/0300-9084(90)90123-X Szabo R, Netzel-Arnett S, Hobson JP, Antalis TM, Bugge TH. Malriptase-3 is a novel phylogenetically preserved membrane-anchored serine protease with broad serpin reactivity. Biochem J, 2005, 390:231–42., DOI 10.1042/ BJ20050299 NR 3 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JUL PY 2024 VL 30 IS 3 BP 1611914 EP 1611921 DI 10.3389/pore.2024.1611914 PG 8 ER PT J AU Zhao, H Kou, Ch Zhao, H Liu, Q He, M Wang, C Zhu, S Ma, L Wang, Y AF Zhao, Han Kou, Changhua Zhao, Hao Liu, Qing He, Maosheng Wang, Cong Zhu, Saisai Ma, Li Wang, Yun TI Impact of limb ischemic preconditioning on the incidence of vein thrombosis in patients with peripherally inserted central catheter SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tumor; limb ischemic preconditioning; peripherally inserted central catheter; vein thrombosis; PICC-related complications ID tumor; limb ischemic preconditioning; peripherally inserted central catheter; vein thrombosis; PICC-related complications AB Background: Peripherally inserted central catheters (PICC) are increasingly used in clinical practice, which also leads to an increased incidence of PICC-related thrombosis. Local thrombus formation could be prevented by limb ischemic preconditioning (IPC). This study aimed to determine whether IPC can prevent deep vein thrombosis in patients with PICC. Methods: A total of 600 breast cancer patients receiving PICC were randomized into two groups between July 2016 and July 2018 at the Department of Radiation Oncology. In the preconditioning group, 5 min of ischemic preconditioning was performed three times before PICC, whereas no preconditioning was performed in the sham group. The coagulation function levels, the PICC-related complications, the length of hospital stay, the cost of hospitalization, and the satisfaction of patients were compared. Results: The coagulation function levels of patients in the preconditioning group were more normal than in patients from the sham group. In total, 56/ 300 patients in the sham group had presence of PICC-related thrombosis, with only 23/300 in the IPC group, with no significant difference in other complications between the two groups. However, a longer hospital stay was observed in the sham group compared to the IPC group. Moreover, the cost of hospitalization was also reduced in the IPC group, which also improved the satisfaction of patients. Conclusion: Limb ischemic preconditioning may attenuate the severity of vein thrombosis in patients with PICC, which contributes to reducing the incidence of PICC-related thrombosis in clinical practice. C1 [Zhao, Han] Hannover Medical School, Clinic for Kidney and Hypertensive DiseasesHannover, Germany. [Kou, Changhua] The Affiliated Hospital of the Southeast University Medical School, Xuzhou City Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Department of Hepatobiliary Pancreatic CenterXuzhou, Jiangsu, China. [Zhao, Hao] The Affiliated Hospital of the Southeast University Medical School, Xuzhou City Central Hospital, Department of Vascular SurgeryXuzhou, Jiangsu, China. [Liu, Qing] The Affiliated Hospital of the Southeast University Medical School, Xuzhou City Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Department of Gynecology DepartmentXuzhou, Jiangsu, China. [He, Maosheng] The Affiliated Hospital of the Southeast University Medical School, Xuzhou City Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Department of Color Ultrasound DepartmentXuzhou, Jiangsu, China. [Wang, Cong] The Affiliated Hospital of the Southeast University Medical School, Xuzhou City Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Department of Hepatobiliary Pancreatic CenterXuzhou, Jiangsu, China. [Zhu, Saisai] The Affiliated Hospital of the Southeast University Medical School, Xuzhou City Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Department of Hepatobiliary Pancreatic CenterXuzhou, Jiangsu, China. [Ma, Li] The Affiliated Hospital of the Southeast University Medical School, Xuzhou City Central Hospital, Department of Thyroid and Breast SurgeryXuzhou, Jiangsu, China. [Wang, Yun] The Affiliated Hospital of the Southeast University Medical School, Xuzhou City Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Department of Hepatobiliary Pancreatic CenterXuzhou, Jiangsu, China. RP Wang, Y (reprint author), The Affiliated Hospital of the Southeast University Medical School, Xuzhou City Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Department of Hepatobiliary Pancreatic Center, Xuzhou, China. EM wyhh666@gmail.com CR Galen B, Baron S, Young S, Hall A, Berger-Spivack L, Southern W. Reducing peripherally inserted central catheters and midline catheters by training nurses in ultrasound-guided peripheral intravenous catheter placement. BMJ Qual Saf, 2020, 29(3):245–9. Epub 2019 Oct 3. PMID: 31582569., DOI 10.1136/bmjqs-2019-009923 Santos FKY, Flumignan RLG, Areias LL, Sarpe AKP, Amaral FCF, Avila RB, et al. Peripherally inserted central catheter versus central venous catheter for intravenous access: a protocol for systematic review and meta-analysis. Medicine, Baltimore,, 2020, 99(30):e20352. PMID: 32791657; PMCID: PMC7386962., DOI 10. 1097/MD.0000000000020352 Chen X, LiangM. A meta-analysis of incidence of catheter-related bloodstream infection with midline catheters and peripherally inserted central catheters. J Healthc Eng, 2022, 12:6383777. PMID: 35313516; PMCID: PMC8934223., DOI 10.1155/2022/6383777 Swaminathan L, Flanders S, Horowitz J, Zhang Q, O’Malley M, Chopra V. Safety and outcomes of midline catheters vs peripherally inserted central catheters for patients with short-term indications: a multicenter study. JAMA Intern Med, 2022, 182(1):50–8. PMID: 34842905; PMCID: PMC8630646., DOI 10.1001/ jamainternmed.2021.6844 Mielke D, Wittig A, Teichgraber U. Peripherally inserted central venous catheter, PICC, in outpatient and inpatient oncological treatment. Support Care Cancer, 2020, 28(10):4753–60. Epub 2020 Jan 22. PMID: 31970514; PMCID: PMC7447660., DOI 10.1007/s00520-019-05276-0 Lamidi S, Baker DM, Wilson MJ, Lee MJ. Remote ischemic preconditioning in non-cardiac surgery: a systematic review and meta-analysis. J Surg Res, 2021, 261: 261–73. Epub 2021 Jan 15. PMID: 33460972., DOI 10.1016/j.jss.2020.12.037 Leurcharusmee P, Sawaddiruk P, Punjasawadwong Y, Sugandhavesa N, Klunklin K, Tongprasert S, et al. Ischemic preconditioning upregulates Mitofusin2 and preserves muscle strength in tourniquet-induced ischemia/ reperfusion. J Orthop Translat, 2022, 35:113–21. PMID: 36312592; PMCID: PMC9582561., DOI 10.1016/j.jot.2022.09.012 Wu J, Yu C, Zeng X, Sun C. The hepatoprotective effect from ischemiareperfusion injury of remote ischemic preconditioning in the liver related surgery: a meta-analysis. ANZ J Surg, 2022, 92(6):1332–7. Epub 2021 Dec 2. PMID: 34854193., DOI 10.1111/ans.17236 Gorog DA, Farag M, Spinthakis N, Yellon DM, Botker HE, Kharbanda RK, et al. Effect of remote ischaemic conditioning on platelet reactivity and endogenous fibrinolysis in ST-elevation myocardial infarction: a substudy of the CONDI-2/ ERIC-PPCI randomized controlled trial. Cardiovasc Res, 2021, 117(2):623–34. PMID: 32163139; PMCID: PMC7820881., DOI 10.1093/cvr/cvaa061 Kleidon TM, Horowitz J, Rickard CM, Ullman AJ, Marsh N, Schults J, et al. Peripherally inserted central catheter thrombosis after placement via electrocardiography vs traditional methods. Am J Med, 2021, 134(2):e79–e88. Epub 2020 Jul 14. PMID: 32673624., DOI 10.1016/j.amjmed.2020.06.010 Yuen HLA, Tran H, Chunilal S. Upper extremity deep vein thrombosis: current knowledge and future directions. Semin Thromb Hemost, 2021, 47(6): 677–91. Epub 2021 May 10. PMID: 33971684., DOI 10.1055/s-0041-1725116 Krag AE, Kiil BJ, Hvas CL, Hvas AM. Effect of remote ischemic preconditioning on hemostasis and fibrinolysis in head and neck cancer surgery: a randomized controlled trial. PLoS One, 2019, 14(7):e0219496. PMID: 31283796; PMCID: PMC6613699., DOI 10.1371/journal.pone.0219496 UrtechoM, Torres Roldan VD, Nayfeh T, Espinoza Suarez NR, Ranganath N, Sampathkumar P, et al. Comparing complication rates of midline catheter vs peripherally inserted central catheter. A systematic review and meta-analysis. Open Forum Infect Dis, 2023, 10(2):ofad024. PMID: 36751645; PMCID: PMC9898877., DOI 10.1093/ofid/ofad024 Estrada-Orozco K, Cantor-Cruz F, Larrotta-Castillo D, Diaz-Rios S, Ruiz- Cardozo MA. Central venous catheter insertion and maintenance: evidence-based clinical recommendations. Rev Colomb Obstet Ginecol, 2020, 71(2):115–62. PMID: 32770871., DOI 10.18597/rcog.3413 Balsorano P, Virgili G, Villa G, Pittiruti M, Romagnoli S, De Gaudio AR, et al. Peripherally inserted central catheter-related thrombosis rate in modern vascular access era-when insertion techniquematters: a systematic review andmeta-analysis. J Vasc Access, 2020, 21(1):45–54. Epub 2019 Jun 10. PMID: 31177939., DOI 10.1177/ 1129729819852203 Marsh N, Webster J, Ullman AJ, Mihala G, Cooke M, Chopra V, et al. Peripheral intravenous catheter non-infectious complications in adults: a systematic review and meta-analysis. J Adv Nurs, 2020, 76(12):3346–62. Epub 2020 Oct 5. PMID: 33016412., DOI 10.1111/jan.14565 Hu Q, Su Y, Yan L. Effects of peripherally inserted central catheter, PICC, catheterization nursing on bloodstream infection in peripheral central venous catheters in lung cancer: a single-center, retrospective study. Comput Math Methods Med, 2022, 2022:2791464. PMID: 36158127; PMCID: PMC9499753., DOI 10.1155/2022/2791464 Liang J, Han R, Zhou B. Metabolic reprogramming: strategy for ischemic stroke treatment by ischemic preconditioning. Biology, Basel,, 2021, 10(5):424. PMID: 34064579; PMCID: PMC8151271., DOI 10.3390/biology10050424 Donato M, Bin EP, D Annunzio V, Gelpi RJ. Myocardial remote ischemic preconditioning: from cell biology to clinical application. Mol Cel Biochem, 2021, 476(10):3857–67. Epub 2021 Jun 14. PMID: 34125317., DOI 10.1007/s11010-021-04192-4 Litrico L, Aid R, Youkharibache A, Letourneur D, Cristofari S. Effect of ischemic preconditioning on skeletal tissue tolerance after warm venous ischemia. Ann Chir Plast Esthet, 2023, 68(4):315–25. Epub 2023 Mar 23. PMID: 36966096., DOI 10.1016/j.anplas.2023.02.003 Reddel CJ, Pennings GJ, Lau JK, Chen VM, Kritharides L. Circulating platelet-derived extracellular vesicles are decreased after remote ischemic preconditioning in patients with coronary disease: a randomized controlled trial. J Thromb Haemost, 2021, 19(10):2605–11. Epub 2021 Aug 10. PMID: 34196106., DOI 10.1111/jth.15441 Wang W, Wang JN, Yu W, Zhu SN, Gao Y, Zhang JQ. Comparison of coagulation function between adrenocorticotropic hormone independent Cushing syndrome and nonfunctional adrenal adenoma and its influence factors. Beijing Da Xue Xue Bao Yi Xue Ban, 2023, 55(6):1062–7. Chinese. PMID: 38101790; PMCID: PMC10723997., DOI 10.19723/j.issn.1671-167X.2023.06.017 Li X, Li T, Fan Y. Efficacy of intravascular mechanical thrombectomy combined with thrombolysis and anticoagulant therapy in the treatment of cerebral venous sinus thrombosis and its effect on neurological function and coagulation indices. Am J Transl Res, 2021, 13(6):6921–8. PMID: 34306444; PMCID: PMC8290642. MSM, Samal DB, Amirtraj JV, Subramanian S, Venkatasubbu GD. Enhanced coagulation cascade activation and styptic effects of Zn@SiO2nanocomposite. Colloids Surf B Biointerfaces, 2024, 239:113927. Epub 2024 Apr 25. PMID: 38714078., DOI 10.1016/j.colsurfb.2024.113927 Sun Y, Mao P, Lu J, Li L, Lu W, Jiang Q, et al. Localized lower extremity ischemic preconditioning prevents against local thrombus formation. Vasa, 2015, 44(4):285–8. PMID: 26314360., DOI 10.1024/0301-1526/a000443 Yusof M, Kamada K, Kalogeris T, Gaskin FS, Korthuis RJ. Hydrogen sulfide triggers late-phase preconditioning in postischemic small intestine by an NO- and p38 MAPK-dependent mechanism. Am J Physiol Heart Circ Physiol, 2009, 296(3): H868–76. Epub 2009 Jan 23. PMID: 19168723; PMCID: PMC2660238., DOI 10.1152/ ajpheart.01111.2007 Kram L, Grambow E, Mueller-Graf F, Sorg H, Vollmar B. The anti-thrombotic effect of hydrogen sulfide is partly mediated by an upregulation of nitric oxide synthases. Thromb Res, 2013, 132(2):e112–7. Epub 2013 Aug 2. PMID: 23916820., DOI 10.1016/j.thromres.2013.07.010 Ekeloef S, Koyuncu S, Holst-Knudsen J,GundelO,Meyhoff CS,HomiliusM, et al. Cardiovascular events in patients undergoing hip fracture surgery treated with remote ischaemic preconditioning: 1-year follow-up of a randomised clinical trial. Anaesthesia, 2021, 76(8):1042–50. Epub 2021 Jan 13. PMID: 33440017., DOI 10.1111/anae.15357 Fu J, CaiW, Zeng B, He L, Bao L, Lin Z, et al. Development and validation of a predictive model for peripherally inserted central catheter-related thrombosis in breast cancer patients based on artificial neural network: a prospective cohort study. Int J Nurs Stud, 2022, 135:104341. Epub 2022 Aug 8. PMID: 36084529., DOI 10.1016/ j.ijnurstu.2022.104341 Matysiak K, Szewczuk M, Sobocki J, Zdziarska M, Siatkowski I. Complications of tunneled peripherally inserted and tunneled-cuffed central catheters in home parenteral nutrition. Nutrition, 2021, 91-92:111354–92. Epub 2021 May 26. PMID: 34246088., DOI 10.1016/j.nut.2021.111354 Chen P, Wan G, Zhu B. Incidence and risk factors of symptomatic thrombosis related to peripherally inserted central catheter in patients with lung cancer. J AdvNurs, 2021, 77(3):1284–92. Epub 2020 Nov 29. PMID: 33249623., DOI 10.1111/jan.14666 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1611596 EP 1611604 DI 10.3389/pore.2024.1611596 PG 9 ER PT J AU Bakoglu, N Sakamoto, H Yoshida, M Ohnishi, T Lee, SY Smith, L Yagi, Y AF Bakoglu, Nilay Sakamoto, Hirotsugu Yoshida, Masao Ohnishi, Takashi Lee, Seung-Yi Smith, Lindsey Yagi, Yukako TI Artificial intelligence-based automated determination in breast and colon cancer and distinction between atypical and typical mitosis using a cloud-based platform SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE mitosis; colon adenecarcinoma; breast; artificial intelligence; invasive carcinoma ID mitosis; colon adenecarcinoma; breast; artificial intelligence; invasive carcinoma AB Artificial intelligence (AI) technology in pathology has been utilized in many areas and requires supervised machine learning. Notably, the annotations that define the ground truth for the identification of different confusing process pathologies, vary from study to study. In this study, we present our findings in the detection of invasive breast cancer for the IHC/ISH assessment system, along with the automated analysis of each tissue layer, cancer type, etc. in colorectal specimens. Additionally, models for the detection of atypical and typical mitosis in several organs were developed using existing whole-slide image (WSI) sets from other AI projects. All H&E slides were scanned by different scanners with a resolution of 0.12–0.50 μm/pixel, and then uploaded to a cloud-based AI platform. Convolutional neural networks (CNN) training sets consisted of invasive carcinoma, atypical and typical mitosis, and colonic tissue elements (mucosa-epithelium, lamina propria, muscularis mucosa, submucosa, muscularis propria, subserosa, vessels, and lymph nodes). In total, 59 WSIs from 59 breast cases, 217 WSIs from 54 colon cases, and 28 WSIs from 23 different types of tumor cases with relatively higher amounts of mitosis were annotated for the training. The harmonic average of precision and sensitivity was scored as F1 by AI. The final AI models of the Breast Project showed an F1 score of 94.49% for Invasive carcinoma. The mitosis project showed F1 scores of 80.18%, 97.40%, and 97.68% for mitosis, atypical, and typical mitosis layers, respectively. Overall F1 scores for the current results of the colon project were 90.02% for invasive carcinoma, 94.81% for the submucosa layer, and 98.02% for vessels and lymph nodes. After the training and optimization of the AI models and validation of each model, external validators evaluated the results of the AI models via blind-reader tasks. The AI models developed in this study were able to identify tumor foci, distinguish in situ areas, define colonic layers, detect vessels and lymph nodes, and catch the difference between atypical and typical mitosis. All results were exported for integration into our in-house applications for breast cancer and AI model development for both wholeblock and whole-slide image-based 3D imaging assessment. C1 [Bakoglu, Nilay] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [Sakamoto, Hirotsugu] Jichi Medical University, Department of Medicine, Division of GastroenterologyTochigi, Japan. [Yoshida, Masao] Shizuoka Cancer Center, Division of EndoscopyShizuoka, Japan. [Ohnishi, Takashi] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. [Lee, Seung-Yi] Aiforia TechnologiesCambridge, MA, USA. [Smith, Lindsey] Flywheel.ioMinneapolis, MN, USA. [Yagi, Yukako] Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory MedicineNew York, NY, USA. RP Yagi, Y (reprint author), Memorial Sloan Kettering Cancer Center, Department of Pathology and Laboratory Medicine, New York, USA. EM yagiy@mskcc.org CR Laury AR, Blom S, Ropponen T, Virtanen A, Carpen OM. Artificial intelligence-based image analysis can predict outcome in high-grade serous carcinoma via histology alone. Scientific Rep, 2021, 11(1):19165., DOI 10.1038/ s41598-021-98480-0 Coudray N, Ocampo PS, Sakellaropoulos T, Narula N, Snuderl M, Fenyo D, et al. Classification, and mutation prediction from non–small cell lung cancer histopathology images using deep learning. NatMed, 2018, 24(10):1559–67., DOI 10. 1038/s41591-018-0177-5 Arvaniti E, Fricker KS, Moret M, Rupp N, Hermanns T, Fankhauser C, et al. Automated Gleason grading of prostate cancer tissue microarrays via deep learning. Scientific Rep, 2018, 8(1):12054., DOI 10.1038/s41598-018-30535-1 Baxi V, Edwards R, Montalto M, Saha S. Digital pathology and artificial intelligence in translational medicine and clinical practice. Mod Pathol, 2022, 35(1): 23–32., DOI 10.1038/s41379-021-00919-2 Tizhoosh HR, Pantanowitz L. Artificial intelligence and digital pathology: challenges and opportunities. J Pathol Inform, 2018, 9(1):38., DOI 10.4103/jpi.jpi_ 53_18 Pantanowitz L, Sharma A, Carter AB, Kurc T, Sussman A, Saltz J. Twenty years of digital pathology: an overview of the road travelled, what is on the horizon, and the emergence of vendor-neutral archives. J Pathol Inform, 2018, 9(1):40., DOI 10. 4103/jpi.jpi_69_18 Colling R, Pitman H, Oien K, Rajpoot N, Macklin P, Snead D, et al. Artificial intelligence in digital pathology: a roadmap to routine use in clinical practice. The J Pathol, 2019, 249(2):143–50., DOI 10.1002/path.5310 Ehteshami BB, Veta M, Diest PJvan, Ginneken B, van KN, Litjens G, et al. Diagnostic assessment of deep learning algorithms for detection of lymph node metastases in women with breast cancer. JAMA, 2017, 318:2199–0., DOI 10.1001/ jama.2017.14585 Thakur N, Yoon H, Chong Y. Current trends of artificial intelligence for colorectal cancer pathology image analysis: a systematic review. Cancers, 2020, 12(7):1884., DOI 10.3390/cancers12071884 Van der Laak J, Litjens G, Ciompi F. Deep learning in histopathology: the path to the clinic. Nat Med, 2021, 27(5):775–84., DOI 10.1038/s41591-021- 01343-4 Litjens G, Sanchez CI, Timofeeva N, Hermsen M, Nagtegaal I, Kovacs I, et al. Deep learning as a tool for increased accuracy and efficiency of histopathological diagnosis. Sci Rep, 2016, 6:26286., DOI 10.1038/srep26286 Deo SV, Sharma J, Kumar S. GLOBOCAN 2020 report on global cancer burden: challenges and opportunities for surgical oncologists. Ann Surg Oncol, 2022, 29(11):6497–500., DOI 10.1245/s10434-022-12151-6 Giordano SH, Franzoi MA, Temin S, Anders CK, Chandarlapaty S, Crews JR, et al. Systemic therapy for advanced human epidermal growth factor receptor 2–positive breast cancer: ASCO guideline update. J Clin Oncol, 2022, 40(23): 2612–35., DOI 10.1200/JCO.22.00519 Lim TH, Lim AS, Tien SL, Tan PH. Impact of the updated 2018 American Society of Clinical Oncology/College of American Pathologists, ASCO/CAP, guidelines on Human Epidermal Growth Factor Receptor 2, HER2, gene testing in invasive breast cancers: a single center study. Ann Diagn Pathol, 2022, 58: 151935., DOI 10.1016/j.anndiagpath.2022.151935 Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, et al. Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment, and follow-up. Ann Oncol, 2019, 30(8):1194–220., DOI 10.1093/ annonc/mdz173 Hossain MS, Hanna MG, Uraoka N, Nakamura T, Edelweiss M, Brogi E, et al. Automatic quantification of HER2 gene amplification in invasive breast cancer from chromogenic in situ hybridization whole slide images. J Med Imaging, 2019, 6(4):047501., DOI 10.1117/1.JMI.6.4.047501 Yu Z, Jiang D, Huang W, Luo R, Wang H, Su J, et al. Comparison of two pathological processing methods for large endoscopic submucosal dissection, ESD, specimens. J Clin Pathol, 2023, 76(11):757–62., DOI 10.1136/jcp-2022-208491 Ahmed Y, Othman M. EMR/ESD: techniques, complications, and evidence. Curr Gastroenterol Rep, 2020, 22:39–2., DOI 10.1007/s11894-020-00777-z Ishihara R, ArimaM, Iizuka T, Oyama T, Katada C, Kato M, et al. Endoscopic submucosal dissection/endoscopic mucosal resection guidelines for esophageal cancer. Dig Endosc, 2020, 32(4):452–93., DOI 10.1111/den.13654 Mathew T, Kini JR, Rajan J. Computational methods for automated mitosis detection in histopathology images: a review. Biocybernetics Biomed Eng, 2021, 41(1):64–82., DOI 10.1016/j.bbe.2020.11.005 Li C, Wang X, Liu W, Latecki LJ. DeepMitosis: mitosis detection via deep detection, verification and segmentation networks. Med image Anal, 2018, 45: 121–33., DOI 10.1016/j.media.2017.12.002 Zehra T, Anjum S, Mahmood T, ShamsM, Sultan BA, Ahmad Z, et al. A novel deep learning-based mitosis recognition approach and dataset for uterine leiomyosarcoma histopathology. Cancers, 2022, 14(15):3785., DOI 10.3390/ cancers14153785 Ibrahim A, Lashen AG, Katayama A,Mihai R, Ball G, Toss MS, et al. Defining the area of mitoses counting in invasive breast cancer using whole slide image. Mod Pathol, 2022, 35(6):739–48., DOI 10.1038/s41379-021-00981-w Lakshmanan B, Priyadharsini S, Selvakumar B. Computer assisted mitotic figure detection in histopathology images based on DenseNetPCA framework. Mater Today Proc, 2022, 62:4936–9., DOI 10.1016/j.matpr.2022.03.683 Ibrahim A, Lashen A, Toss M, Mihai R, Rakha E. Assessment of mitotic activity in breast cancer: revisited in the digital pathology era. J Clin Pathol, 2021, 23:365–72., DOI 10.1136/jclinpath-2021-207742 Anttila TT, Aspinen S, Pierides G, Haapamaki V, Laitinen MK, Ryhanen J. Enchondroma detection from hand radiographs with an interactive deep learning segmentation tool—a feasibility study. J Clin Med, 2023, 12(22):7129., DOI 10.3390/ jcm12227129 Kather JN, Heij LR, Grabsch HI, Loeffler C, Echle A, Muti HS, et al. Pancancer image-based detection of clinically actionable genetic alterations. Nat Cancer, 2020, 1(8):789–99., DOI 10.1038/s43018-020-0087-6 Satturwar S, Parwani AV. Artificial intelligence-enabled prostate cancer diagnosis and prognosis: current state and future implications. Adv Anat Pathol, 2024, 31(2):136–44., DOI 10.1097/PAP.0000000000000425 BakogluN, Cesmecioglu E,Ohnishi T, OhnishiC, RossDR, YagiY Evaluation of AIintegrated HER-2 dual ISH detection systems in invasive breast carcinoma, 104. Modern Pathology: Laboratory Investigation, 2024). p. S115–117. Issue3, Suppl 1, 100428. Ozkan TA, Eruyar AT, Cebeci OO, Memik O, Ozcan L, Kuskonmaz I. Interobserver variability in Gleason histological grading of prostate cancer. Scand J Urol, 2016, 50(6):420–4., DOI 10.1080/21681805.2016.1206619 Aeffner F, Wilson K, Bolon B, Kanaly S, Mahrt CR, Rudmann D, et al. Commentary: roles for pathologists in a high-throughput image analysis team. Toxicologic Pathol, 2016, 44(6):825–34., DOI 10.1177/0192623316653492 Cayir S, Solmaz G, Kusetogullari H, Tokat F, Bozaba E, Karakaya S, et al. MITNET: a novel dataset and a two-stage deep learning approach for mitosis recognition in whole slide images of breast cancer tissue. Neural Comput Appl, 2022, 34(20):17837–51., DOI 10.1007/s00521-022-07441-9 Cree IA, Tan PH, Travis WD, Wesseling P, Yagi Y, White VA, et al. Counting mitoses: SI, ze, matters! Mod Pathol. 2021;34(9):1651–7., DOI 10.1038/s41379-021- 00825-7 Li J, Adachi T, Takeyama S, Yamaguchi M, Yagi Y. U-Net based mitosis detection from H&E-stained images with the semi-automatic annotation using pHH3 IHC-stained images. In: Medical imaging 2022: image processing. SPIE, 2022), 12032:669–681. Matsuda Y, Yoshimura H, Ishiwata T, Sumiyoshi H,Matsushita A, Nakamura Y, et al. Mitotic index and multipolar mitosis in routine histologic sections as prognostic markers of pancreatic cancers: a clinicopathological study. Pancreatology, 2016, 16(1):127–32., DOI 10.1016/j.pan.2015.10.005 Xu B, Teplov A, Ibrahim K, Inoue T, Stueben B, Katabi N, et al. Detection and assessment of capsular invasion, vascular invasion and lymph node metastasis volume in thyroid carcinoma using microCT scanning of paraffin tissue blocks, 3D whole block imaging): a proof of concept. Mod Pathol, 2020, 33(12):2449–57., DOI 10.1038/s41379-020-0605-1 Ohnishi T, Teplov A, Kawata N, Ibrahim K, Ntiamoah P, Firat C, et al. Threedimensional vessel segmentation in whole-tissue and whole-block imaging using a deep neural network: proof-of-concept study. The Am J Pathol, 2021, 191(3): 463–74., DOI 10.1016/j.ajpath.2020.12.008 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1611815 EP 1611829 DI 10.3389/pore.2024.1611815 PG 15 ER PT J AU Li, Y Wang, Y He, W AF Li, Yanxi Wang, Yujiao He, Weimin TI Case report: Orbital myeloid sarcoma: a report of two rare cases and review of the literature SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE myeloid sarcoma (MS); orbit; histopathology; immunohistochemistry; diagnosis; treatment ID myeloid sarcoma (MS); orbit; histopathology; immunohistochemistry; diagnosis; treatment AB Myeloid sarcoma (MS) occurs when primitive or naive myeloid cells form outside the bone marrow. It occurs mainly in soft/connective tissue and skin; orbital involvement is rare. We report the cases of two female adults, analyze the clinicopathologic characteristics, and review the literature. The average age of both patients was 28 years and they presented unilateral proptosis combined with varying degrees of impaired visual acuity and restricted ocular motility in the affected eye. Despite this, they maintained good overall health and no notable family history. However, the patients had no systemic clinical manifestations of acute myeloid leukemia (AML). Both patients underwent surgical resection of the orbital tumor. Immunohistochemistry showed positive staining for CD43, Leukocyte Common Antigen (LCA), and myeloperoxidase (MPO) and a high level of positive staining for Ki67, which were diagnostic for MS. Bone marrow cytology examination showed no apparent abnormalities. Postoperative chemotherapy, local radiotherapy, and allogeneic hematopoietic stem cell transplantation (allo-HSCT) were performed in Case 1, while the second patient underwent adjuvant chemotherapy and radiotherapy. No recurrence or metastasis was found in either patient during follow-up (one more than 5 years, the other more than 10 years). The occurrence of orbital MS is infrequent, with atypical clinical and imaging findings. The diagnosis depends on pathomorphology and immunohistochemical staining, and the prognosis is good with postoperative adjuvant chemotherapy, local radiotherapy, and allo- HSCT. C1 [Li, Yanxi] West China Hospital of Sichuan University, Department of OphthalmologyChengdu, Sichuan, China. [Wang, Yujiao] West China Hospital of Sichuan University, Department of OphthalmologyChengdu, Sichuan, China. [He, Weimin] West China Hospital of Sichuan University, Department of OphthalmologyChengdu, Sichuan, China. RP He, W (reprint author), West China Hospital of Sichuan University, Department of Ophthalmology, Chengdu, China. EM hewm888@hotmail.com CR Khoury JD, Solary E, Abla O, Akkari Y, Alaggio R, Apperley JF, et al. The 5th edition of the World health organization classification of haematolymphoid tumours: myeloid and histiocytic/dendritic neoplasms. Leukemia, 2022, 36(7): 1703–19., DOI 10.1038/s41375-022-01613-1 Arber DA, Orazi A, Hasserjian RP, Borowitz MJ, Calvo KR, Kvasnicka HM, et al. International consensus classification of myeloid neoplasms and acute leukemias: integrating morphologic, clinical, and genomic data. Blood, 2022, 140(11):1200–28., DOI 10.1182/blood.2022015850 Shallis RM, Gale RP, Lazarus HM, Roberts KB, Xu ML, Seropian SE, et al. Myeloid sarcoma, chloroma, or extramedullary acute myeloid leukemia tumor: a tale of misnomers, controversy and the unresolved. Blood Rev, 2021, 47:100773., DOI 10.1016/j.blre.2020.100773 Abbas HA, Reville PK, Geppner A, Rausch CR, Pemmaraju N, Ohanian M, et al. Clinical andmolecular characterization of myeloid sarcoma without medullary leukemia. Leuk Lymphoma, 2021, 62(14):3402–10., DOI 10.1080/10428194.2021. 1961235 Purohit BS, Vargas MI, Ailianou A, Merlini L, Poletti PA, Platon A, et al. Orbital tumours and tumour-like lesions: exploring the armamentarium of multiparametric imaging. Insights into imaging, 2016, 7(1):43–68., DOI 10.1007/ s13244-015-0443-8 AlSemari MA, Perrotta M, Russo C, Alkatan HM, Maktabi A, Elkhamary S, et al. Orbital myeloid sarcoma, chloroma): report of 2 cases and literature review. Am J Ophthalmol Case Rep, 2020, 19:100806., DOI 10.1016/j.ajoc.2020.100806 Pileri SA, Ascani S, Cox MC, Campidelli C, Bacci F, Piccioli M, et al. Myeloid sarcoma: clinico-pathologic, phenotypic and cytogenetic analysis of 92 adult patients. Leukemia, 2007, 21(2):340–50., DOI 10.1038/sj.leu.2404491 Alexiev BA, Wang W, Ning Y, Chumsri S, Gojo I, Rodgers WH, et al. Myeloid sarcomas: a histologic, immunohistochemical, and cytogenetic study. Diagn Pathol, 2007, 2:42., DOI 10.1186/1746-1596-2-42 Xiao AW, Jia Y, Baughn LB, Pearce KE, Pitel BA, Aster JC, et al. IGH rearrangement in myeloid neoplasms. Haematologica, 2020, 105(6):e315–7., DOI 10. 3324/haematol.2020.246744 Bakst R, Powers A, Yahalom J. Diagnostic and therapeutic considerations for extramedullary leukemia. Curr Oncol Rep, 2020, 22(7):75., DOI 10.1007/s11912-020- 00919-6 Liu H. Emerging agents and regimens for AML. J Hematol Oncol, 2021, 14(1): 49., DOI 10.1186/s13045-021-01062-w Chevallier P, Labopin M, Cornelissen J, Socie G, Rocha V, Mohty M. Allogeneic hematopoietic stem cell transplantation for isolated and leukemic myeloid sarcoma in adults: a report from the Acute Leukemia Working Party of the European group for blood and marrow transplantation. Haematologica, 2011, 96(9):1391–4., DOI 10.3324/haematol.2011.041418 Dohner H, Wei AH, Appelbaum FR, Craddock C, DiNardo CD, Dombret H, et al. Diagnosis andmanagement of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood, 2022, 140(12):1345–77., DOI 10.1182/blood.2022016867 Bakst R, Wolden S, Yahalom J. Radiation therapy for chloroma, granulocytic sarcoma). Int JRadiatOncol Biol Phys, 2012, 82(5):1816–22., DOI 10.1016/j.ijrobp.2011.02.057 Zimmerman LE, Font RL. Ophthalmologic manifestations of granulocytic sarcoma, myeloid sarcoma or chloroma). The third Pan American Association of Ophthalmology and American Journal of Ophthalmology Lecture. Am J Ophthalmol, 1975, 80(6):975–90., DOI 10.1016/0002-9394(75)90326-8 Cavdar AO, Babacan E, Gozdasoglu S, Kilicturgay K, Arcasoy A, Cin S, et al. High risk subgroup of acute myelomonocytic leukemia, AMML, with orbito-ocular granulocytic sarcoma, OOGS, in Turkish children. Retrospective analysis of clinical, hematological, ultrastructural and therapeutical findings of thirty-three OOGS. Acta Haematologica, 1989, 81(2):80–5., DOI 10.1159/000205531 Bidar M, Wilson MW, Laquis SJ, Wilson TD, Fleming JC, Wesley RE, et al. Clinical and imaging characteristics of orbital leukemic tumors. Ophthalmic Plast Reconstr Surg, 2007, 23(2):87–93., DOI 10.1097/IOP.0b013e3180333a85 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1611818 EP 1611824 DI 10.3389/pore.2024.1611818 PG 7 ER PT J AU Chen, P Chen, M Bu, Y Che, G Cheng, Ch Wang, Y AF Chen, Pingrun Chen, Maojia Bu, Yijie Che, Guowei Cheng, Chao Wang, Yan TI Prognostic role of lymph node regression in patients with esophageal cancer undergoing neoadjuvant therapy SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Review DE lymph node regression; esophageal cancer; neoadjuvant therapy; survival; meta-analysis ID lymph node regression; esophageal cancer; neoadjuvant therapy; survival; meta-analysis AB Purpose: To clarify the prognostic value of lymph node regression (LNR) status including the lymph node regression grade (LNRG) and N downstaging in patients with esophageal cancer receiving neoadjuvant therapy based on available evidence. Methods: Several databases were searched up to 25 March 2024. The main outcomes included overall survival (OS), disease-free survival (DFS) and cancerspecific survival (CSS). Hazard ratios (HRs) and 95% confidence intervals (CIs) were combined. Subgroup analyses based on the neoadjuvant therapy and pathological type were also conducted. Results: In total, 14 retrospective studies with 3,212 participants were included. Nine and five studies explored the relationship between LNRG and N downstaging and survival, respectively. Pooled results indicated that complete LNR predicted significantly improved OS (HR = 0.47, 95% CI: 0.41–0.55, P < 0.001) and DFS (HR = 0.42, 95% CI: 0.32–0.55, P < 0.001) and subgroup analysis based on neoadjuvant therapy and pathological type manifested similar results. Besides, N downstaging was also significantly related to improved OS (HR = 0.40, 95% CI: 0.21–0.77, P = 0.006) and CSS (HR = 0.27, 95% CI: 0.12–0.60, P < 0.001). Conclusion: LNR could serve as a novel and reliable prognostic factor in patients with esophageal cancer receiving neoadjuvant therapy and complete LNR and N downstaging predict better survival. C1 [Chen, Pingrun] Sichuan University, West China Hospital, Department of GastroenterologyChengdu, China. [Chen, Maojia] Sichuan University, West China Hospital, Animal Experiment CenterChengdu, China. [Bu, Yijie] Sichuan University, West China Hospital, Department of Thoracic SurgeryChengdu, China. [Che, Guowei] Sichuan University, West China Hospital, Division of Thoracic OncologyChengdu, China. [Cheng, Chao] Sichuan University, West China Hospital, Department of Thoracic SurgeryChengdu, China. [Wang, Yan] Sichuan University, West China Hospital, Department of Thoracic SurgeryChengdu, China. RP Wang, Y (reprint author), Sichuan University, West China Hospital, Department of Thoracic Surgery, Chengdu, China. EM wangyanxw@wchscu.cn CR Ju W, Zheng R, Zhang S, Zeng H, Sun K, Wang S, et al. Cancer statistics in Chinese older people, 2022: current burden, time trends, and comparisons with the US, Japan, and the Republic of Korea. Sci China Life Sci, 2023, 66(5):1079–91., DOI 10.1007/s11427-022-2218-x Xia C, Dong X, Li H, Cao M, Sun D, He S, et al. Cancer statistics in China and United States, 2022: profiles, trends, and determinants. Chin Med J, Engl,, 2022, 135(5):584–90., DOI 10.1097/CM9.0000000000002108 Yang W, Niu Y, Sun Y. Current neoadjuvant therapy for operable locally advanced esophageal cancer. Med Oncol, 2023, 40(9):252., DOI 10.1007/s12032-023- 02097-4 Liang Z, Chen T, Li W, Lai H, Li L, Wu J, et al. Efficacy and safety of neoadjuvant chemoradiotherapy versus neoadjuvant chemotherapy in locally advanced esophageal cancer: an updated meta-analysis. Medicine, Baltimore,, 2024, 103(3):e36785., DOI 10.1097/MD.0000000000036785 Tang H, Wang H, Fang Y, Zhu JY, Yin J, Shen YX, et al. Neoadjuvant chemoradiotherapy versus neoadjuvant chemotherapy followed by minimally invasive esophagectomy for locally advanced esophageal squamous cell carcinoma: a prospective multicenter randomized clinical trial. Ann Oncol, 2023, 34(2):163–72., DOI 10.1016/j.annonc.2022.10.508 Eyck BM, van Lanschot JJB, Hulshof M, van der Wilk BJ, Shapiro J, van Hagen P, et al. Ten-year outcome of neoadjuvant chemoradiotherapy plus surgery for esophageal cancer: the randomized controlled CROSS trial. J Clin Oncol, 2021, 39(18):1995–2004., DOI 10.1200/JCO.20.03614 Li Q, Liu T, Ding Z. Neoadjuvant immunotherapy for resectable esophageal cancer: a review. Front Immunol, 2022, 13:1051841., DOI 10.3389/fimmu.2022.1051841 Baksh K, Prithviraj G, Kim Y, Hoffe S, Shridhar R, Coppola D, et al. Correlation between standardized uptake value in preneoadjuvant and postneoadjuvant chemoradiotherapy and tumor regression grade in patients with locally advanced esophageal cancer. Am J Clin Oncol, 2018, 41(3):254–8., DOI 10.1097/ COC.0000000000000258 Yun JK, Kim Y, Lee GD, Choi S, Kim YH, Kim DK, et al. Tumor regression grade combined with lymph node status in esophageal squamous cell carcinoma after neoadjuvant chemoradiotherapy. Cancer Med, 2022, 11(19):3623–32., DOI 10. 1002/cam4.4748 Gu YM, Lyu SM, Shang QX, Zhang HL, Yang YS, Wang WP, et al. Is tumor regression grade sufficient to predict survival in esophageal cancer with trimodal therapy? J Invest Surg, 2022, 35(11-12):1818–23., DOI 10.1080/08941939.2022. 2127036 Yasuda T, Yano M, Miyata H, Yamasaki M, Takiguchi S, Fujiwara Y, et al. Prognostic significance of, 18)F-fluorodeoxyglucose positron emission tomography, FDG-PET)-Positive lymph nodes following neoadjuvant chemotherapy and surgery for resectable thoracic esophageal squamous cell carcinoma. Ann Surg Oncol, 2015, 22(8):2599–607., DOI 10.1245/s10434-014-4299-9 Zhang X, Tan R, Lam WC, Yao L, Wang X, Cheng CW, et al. PRISMA, preferred reporting Items for systematic reviews and meta-analyses, extension for Chinese herbal medicines 2020, PRISMA-CHM 2020). Am J Chin Med, 2020, 48(6):1279–313., DOI 10.1142/S0192415X20500639 Wang Y, Li J, Chang S, Dong Y, Che G. Risk and influencing factors for subsequent primary lung cancer after treatment of breast cancer: a systematic review and two meta-analyses based on four million cases. J Thorac Oncol : official Publ Int Assoc Study Lung Cancer, 2021, 16(11):1893–908., DOI 10.1016/j.jtho.2021. 07.001 Barili F, Parolari A, Kappetein PA, Freemantle N. Statistical Primer: heterogeneity, random- or fixed-effects model analyses? Interact Cardiovasc Thorac Surg, 2018, 27(3):317–21., DOI 10.1093/icvts/ivy163 Begg CB, MazumdarM. Operating characteristics of a rank correlation test for publication bias. Biometrics, 1994, 50(4):1088–101., DOI 10.2307/2533446 Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ, Clinical research ed,, 1997, 315(7109): 629–34., DOI 10.1136/bmj.315.7109.629 Rice TW, Blackstone EH, Adelstein DJ, Zuccaro G, Jr, Vargo JJ, Goldblum JR, et al. N1 esophageal carcinoma: the importance of staging and downstaging. J Thorac Cardiovasc Surg, 2001, 121(3):454–64., DOI 10.1067/mtc.2001.112470 Bollschweiler E, Hoelscher AH, Metzger R, Besch S, Moenig SP, Baldus SE, et al. Prognostic significance of a new grading system of lymph node morphology after neoadjuvant radiochemotherapy for esophageal. Ann Thorac Surg, 2011, 92(6):2020–7., DOI 10.1016/j.athoracsur.2011.06.091 Donohoe CL, O’Farrell NJ, Grant T, King S, Clarke L, Muldoon C, et al. Classification of pathologic response to neoadjuvant therapy in esophageal and junctional cancer: assessment of existing measures and proposal of a novel 3-point standard. Ann Surg, 2013, 258(5):784–92. discussion 792., DOI 10.1097/SLA. 0b013e3182a66588 Nieman DR, Peyre CG, Watson TJ, Cao W, Lunt MD, Lada MJ, et al. Neoadjuvant treatment response in negative nodes is an important prognosticator after esophagectomy. Ann Thorac Surg, 2015, 99(1):277–83., DOI 10.1016/j. athoracsur.2014.07.037 Philippron A, Bollschweiler E, Kunikata A, Plum P, Schmidt C, Favi F, et al. Prognostic relevance of lymph node regression after neoadjuvant chemoradiation for esophageal cancer. Semin Thorac Cardiovasc Surg, 2016, 28(2):549–58., DOI 10. 1053/j.semtcvs.2016.04.003 Zanoni A, Verlato G, Giacopuzzi S, Motton M, Casella F, Weindelmayer J, et al. ypN0: does it matter how you get there? Nodal downstaging in esophageal cancer. Ann Surg Oncol, 2016, 23(Suppl. 5):998–1004., DOI 10.1245/s10434-016- 5440-8 Noble F, Lloyd MA, Turkington R, Griffiths E, O’DonovanM, O’Neill JR, et al. Multicentre cohort study to define and validate pathological assessment of response to neoadjuvant therapy in oesophagogastric adenocarcinoma. Br J Surg, 2017, 104(13):1816–28., DOI 10.1002/bjs.10627 Shapiro J, Biermann K, van Klaveren D, Offerhaus GJ, Ten Kate FJ,Meijer SL, et al. Prognostic value of pretreatment pathological tumor extent in patients treated with neoadjuvant chemoradiotherapy plus surgery for esophageal or junctional cancer. Ann Surg, 2017, 265(2):356–62., DOI 10.1097/SLA.0000000000001630 Davies AR, Myoteri D, Zylstra J, Baker CR, Wulaningsih W, Van Hemelrijck M, et al. Lymph node regression and survival following neoadjuvant chemotherapy in oesophageal adenocarcinoma. Br J Surg, 2018, 105(12):1639–49., DOI 10.1002/bjs. 10900 Hsu P-K, Yeh Y-C, Chien L-I, Huang C-S, Hsu H-S. Clinicopathological significance of pathologic complete lymph node regression after neoadjuvant chemoradiotherapy in esophageal squamous cell carcinoma. Ann Surg Oncol, 2021, 28(4):2048–58., DOI 10.1245/s10434-020-09363-z KoemansWJ, Larue R, Kloft M, Ruisch JE, Compter I, Riedl RG, et al. Lymph node response to chemoradiotherapy in oesophageal cancer patients: relationship with radiotherapy fields. Esophagus, 2021, 18(1):100–10., DOI 10.1007/s10388-020- 00777-y Evans RP, Kamarajah SK, Kunene V, Zardo D, Elshafie M, Griffiths EA. Impact of neoadjuvant chemotherapy on nodal regression and survival in oesophageal adenocarcinoma. Eur J Surg Oncol, 2022, 48(5):1001–10., DOI 10. 1016/j.ejso.2021.12.021 Moore JL, Green M, Santaolalla A, Deere H, Evans RPT, Elshafie M, et al. Pathologic lymph node regression after neoadjuvant chemotherapy predicts recurrence and survival in esophageal adenocarcinoma: a multicenter study in the United Kingdom. J Clin Oncol, 2023, 41(28):4522–34., DOI 10.1200/JCO.23. 00139 Yehan Z, Ying L, Peng G, Zongyao H, Chengmin Z, Hong Y, et al. Prognostic significance of positive lymph node regression grade to neoadjuvant chemoradiation for esophageal squamous cell carcinoma. J Surg Oncol, 2024, 129(4):708–17., DOI 10.1002/jso.27555 Hagens E, Tukanova K, Jamel S, van Berge Henegouwen M, Hanna GB, Gisbertz S, et al. Prognostic relevance of lymph node regression on survival in esophageal cancer: a systematic review and meta-analysis. Dis Esophagus, 2022, 35(1):doab021., DOI 10.1093/dote/doab021 Walter D, Harter PN, Battke F, Winkelmann R, Schneider M, Holzer K, et al. Genetic heterogeneity of primary lesion and metastasis in small intestine neuroendocrine tumors. Sci Rep, 2018, 8(1):3811., DOI 10.1038/s41598-018- 22115-0 Choi JY, Lee YS, Shim DM, Lee YK, Seo SW. GNAQ knockdown promotes bone metastasis through epithelial-mesenchymal transition in lung cancer cells. Bone Joint Res, 2021, 10(5):310–20., DOI 10.1302/2046-3758.105.BJR-2020-0262.R3 Fischer HP. Regression and therapy-resistance of primary liver tumors and liver metastases after regional chemotherapy and local tumor ablation. Pathologe, 2005, 26(3):191–200., DOI 10.1007/s00292-005-0749-2 Popper H. Primary tumor and metastasis-sectioning the different steps of the metastatic cascade. Transl Lung Cancer Res, 2020, 9(5):2277–300., DOI 10.21037/ tlcr-20-175 Cacho-Diaz B, Garcia-Botello DR, Wegman-Ostrosky T, Reyes-Soto G, Ortiz- Sanchez E, Herrera-Montalvo LA. Tumor microenvironment differences between primary tumor and brain metastases. J Transl Med, 2020, 18(1):1., DOI 10.1186/ s12967-019-02189-8 Wood SL, Pernemalm M, Crosbie PA, Whetton AD. The role of the tumormicroenvironment in lung cancer-metastasis and its relationship to potential therapeutic targets. Cancer Treat Rev, 2014, 40(4):558–66., DOI 10.1016/j.ctrv. 2013.10.001 Xu CW,Wang WX, Wu MJ, Zhu YC, Zhuang W, Lin G, et al. Comparison of the c-MET gene amplification between primary tumor and metastatic lymph nodes in non-small cell lung cancer. Thorac Cancer, 2017, 8(5):417–22., DOI 10.1111/1759- 7714.12455 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1611844 EP 1611854 DI 10.3389/pore.2024.1611844 PG 11 ER PT J TI Diagnostic challenges in complicated case of glioblastoma SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE diagnostics; cytogenomics; MLPA; I-FISH; aCGH/SNP; gene panel; WGS ID diagnostics; cytogenomics; MLPA; I-FISH; aCGH/SNP; gene panel; WGS AB Glioblastoma is the commonest primary malignant brain tumor, with a very poor prognosis and short overall survival. It is characterized by its high intra- and intertumoral heterogeneity, in terms of both the level of single-nucleotide variants, copy number alterations, and aneuploidy. Therefore, routine diagnosis can be challenging in some cases. We present a complicated case of glioblastoma, which was characterized with five cytogenomic methods: interphase fluorescence in situ hybridization, multiplex ligation-dependent probe amplification, comparative genomic hybridization array and singlenucleotide polymorphism, targeted gene panel, and whole-genome sequencing. These cytogenomic methods revealed classical findings associated with glioblastoma, such as a lack of IDH and TERT mutations, gain of chromosome 7, and loss of chromosome 10. At least three pathological clones were identified, including one with whole-genome duplication, and one with loss of 1p and suspected loss of 19q. Deletion and mutation of the TP53 gene were detected with numerous breakends on 17p and 20q. Based on these findings, we recommend a combined approach to the diagnosis of glioblastoma involving the detection of copy number alterations, mutations, and aneuploidy. The choice of the best combination of methods is based on cost, time required, staff expertise, and laboratory equipment. This integrated strategy could contribute directly to tangible improvements in the diagnosis, prognosis, and prediction of the therapeutic responses of patients with brain tumors. CR Rasras S, Zibara K, Vosughi T, Zayeri Z. Genetics and epigenetics of glioblastoma: therapeutic challenges. Clin Cancer Invest J, 2018, 7:43–9., DOI 10. 4103/ccij.ccij_82_17 Stupp R, Mason WP, Van Den Bent MJ,Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med, 2005, 352:987–96., DOI 10.1056/nejmoa043330 Park SH, Won J, Kim SI, Lee Y, Park CK, Kim SK, et al. Molecular testing of brain tumor. J Pathol Transl Med, 2017, 51:205–23., DOI 10.4132/jptm.2017.03.08 Perry A, Wesseling P. Histologic classification of gliomas. Handbook Clin Neurol, 2016, 134:71–95., DOI 10.1016/B978-0-12-802997-8.00005-0 Pessoa IA, Amorim CK, Ferreira WAS, Sagica F, Brito JR, Othman M, et al. Detection and correlation of single and concomitant TP53, PTEN, and CDKN2A alterations in gliomas. Int J Mol Sci, 2019, 20:2658–20., DOI 10.3390/ ijms20112658 Horbinski C, Ligon KL, Brastianos P, Huse JT, Venere M, Chang S, et al. The medical necessity of advanced molecular testing in the diagnosis and treatment of brain tumor patients. Neuro-Oncol, 2019, 21:1498–508., DOI 10.1093/neuonc/ noz119 Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro-Oncol, 2021, 23:1231–51., DOI 10.1093/neuonc/noab106 Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, et al. EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol, 2021, 18:170–86., DOI 10.1038/s41571-020- 00447-z DeCordova S, Shastri A, Tsolaki AG, Yasmin H, Klein L, Singh SK, et al. Molecular heterogeneity and immunosuppressive microenvironment in glioblastoma. Front Immunol, 2020, 11:1402–18., DOI 10.3389/fimmu.2020.01402 Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM,Wakimoto H, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science, 2014, 344:1396–401., DOI 10.1126/science.1254257 Almendro V, Marusyk A, Polyak K. Cellular heterogeneity and molecular evolution in cancer. Ann Rev Pathol, 2013, 8:277–302., DOI 10.1146/annurevpathol- 020712-163923 Maley CC, Galipeau PC, Finley JC, Wongsurawat VJ, Li X, Sanchez CA, et al. Genetic clonal diversity predicts progression to esophageal adenocarcinoma. Nat Genet, 2006, 38:468–73., DOI 10.1038/ng1768 Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer, 2012, 12:323–34., DOI 10.1038/nrc3261 Park SY, Gonen M, Kim HJ, Michor F, Polyak K. Cellular and genetic diversity in the progression of in situ human breast carcinomas to an invasive phenotype. J Clin Invest, 2010, 120:636–44., DOI 10.1172/JCI40724 Albertson DG, Pinkel D. Genomic microarrays in human genetic disease and cancer. Hum Mol Genet, 2003, 12:145–52., DOI 10.1093/hmg/ddg261 Zemanova Z,Michalova K, Babicka L, Pavlistova L, Jarosova M, Holzerova M, et al. Clinical relevance of complex chromosomal aberrations in bone marrow cells of 107 children with ETV6/RUNX1 positive acute lymphoblastic leukemia, ALL). Blood, 2006, 108:2278., DOI 10.1182/blood. v108.11.2278.2278 Andrews S. FastQC: a quality control tool for high throughput sequence data, 2010). Available from: http://www.bioinformatics.babraham.ac.uk/projects/fastqc, Accessed February 2, 2024). Garcia M, Juhos S, Larsson M, Olason PI, Martin M, Eisfeldt J, et al. Sarek: a portable workflow for whole-genome sequencing analysis of germline and somatic variants. F1000Res, 2020, 9:63., DOI 10.12688/f1000research.16665.1 Ewels P, Magnusson M, Lundin S, Kaller M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics, 2016, 32: 3047–8., DOI 10.1093/bioinformatics/btw354 Schneider VA, Graves-Lindsay T, Howe K, Bouk N, Chen HC, Kitts PA, et al. Evaluation of GRCh38 and de novo haploid genome assemblies demonstrates the enduring quality of the reference assembly. Genome Res, 2017, 27:849–64., DOI 10. 1101/gr.213611.116 Kim S, Scheffler K, Halpern AL, Bekritsky MA, Noh E, Kallberg M, et al. Strelka2: fast and accurate calling of germline and somatic variants. Nat Methods, 2018, 15:591–4., DOI 10.1038/s41592-018-0051-x Chen X, Schulz-Trieglaff O, Shaw R, Barnes B, Schlesinger F, Kallberg M, et al. Manta: rapid detection of structural variants and indels for germline and cancer sequencing applications. Bioinformatics, 2016, 32:1220–2., DOI 10.1093/ bioinformatics/btv710 Boeva V, Popova T, Bleakley K, Chiche P, Cappo J, Schleiermacher G, et al. Control-FREEC: a tool for assessing copy number and allelic content using nextgeneration sequencing data. Bioinformatics, 2012, 28:423–5., DOI 10.1093/ bioinformatics/btr670 Van Loo P, Nordgard SH, Lingjaerde OC, Russnes HG, Rye IH, Sun W, et al. Allele-specific copy number analysis of tumors. Proc Natl Acad Sci USA, 2010, 107: 16910–5., DOI 10.1073/pnas.1009843107 Cingolani P, Platts A, Wang LL, Coon M, Nguyen T, Wang L, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the Genome of Drosophila melanogaster Strain w1118; iso-2; iso-3. Fly, Austin,, 2012, 6:80–92., DOI 10.4161/fly.19695 McLaren W, Gil L, Hunt SE, Riat HS, Ritchie GR, Thormann A, et al. The Ensembl variant Effect predictor. Genome Biol, 2016, 17:122., DOI 10.1186/s13059- 016-0974-4 The R Development Core Team. R: a language and environment for statistical computing. 2022. Krzywinski M, Schein J, Birol I, Connors J, Gascoyne R, Horsman D, et al. Circos: an information aesthetic for comparative genomics. Genome Res, 2009, 19: 1639–45., DOI 10.1101/gr.092759.109 Xiao J, Jin X, Zhang C, Zou H, Chang Z, Han N, et al. Systematic analysis of enhancer regulatory circuit perturbation driven by copy number variations in malignant glioma. Theranostics, 2021, 11:3060–73., DOI 10.7150/THNO. 54150 Eckel-Passow JE, Decker PA, Kosel ML, Kollmeyer TM, Molinaro AM, Rice T, et al. Using germline variants to estimate glioma and subtype risks. Neuro-Oncol, 2019, 21:451–61., DOI 10.1093/neuonc/noz009 Clarke M, Mackay A, Ismer B, Pickles JC, Tatevossian RG, Newman S, et al. Infant high-grade gliomas comprise multiple subgroups characterized by novel targetable gene fusions and favorable outcomes. Cancer Discov, 2020, 10:942–63., DOI 10.1158/2159-8290.CD-19-1030 Zheng C, Miao X, Li Y, Huang Y, Ruan J, Ma X, et al. Determination of genomic copy number alteration emphasizing a restriction site-based strategy of genome re-sequencing. Bioinformatics, 2013, 29:2813–21., DOI 10.1093/ bioinformatics/btt481 Jenkins RB, Blair H, Ballman KV, Giannini C, Arusell RM, LawM, et al. A t(1; 19)(q10; p10, mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer Res, 2006, 66:9852–61., DOI 10.1158/0008-5472.CAN-06-1796 Felsberg J, Erkwoh A, Sabel MC, Kirsch L, Fimmers R, Blaschke B, et al. Oligodendroglial tumors: refinement of candidate regions on chromosome arm 1p and correlation of 1p/19q status with survival. Brain Pathol, 2004, 14:121–30., DOI 10.1111/j.1750-3639.2004.tb00044.x Smith J, Alderete B, Minn Y, Borell TJ, Perry A, Mohapatra G, et al. Localization of common deletion regions on 1p and 19q in human gliomas and their association with histological subtype. Oncogene, 1999, 18:4144–52., DOI 10. 1038/sj.onc.1202759 Henrich KO, Schwab M, Westermann F. 1p36 tumor suppression—a matter of dosage? Cancer Res, 2012, 72:6079–88., DOI 10.1158/0008-5472.CAN-12-2230 Ichimura K, Vogazianou AP, Liu L, Pearson DM, Backlund LM, Plant K, et al. 1p36 is a preferential target of chromosome 1 deletions in astrocytic tumours and homozygously deleted in a subset of glioblastomas. Oncogene, 2008, 27:2097–108., DOI 10.1038/sj.onc.1210848 Ball MK, Kollmeyer TM, Praska CE, McKenna ML, Giannini C, Raghunathan A, et al. Frequency of false-positive FISH 1p/19q codeletion in adult diffuse astrocytic gliomas. Neuro-oncol Adv, 2020, 2:vdaa109., DOI 10. 1093/noajnl/vdaa109 Brandner S. Molecular diagnostics of adult gliomas in neuropathological practice. Acta Med Acad, 2021, 50:29–46., DOI 10.5644/ama2006-124.324 Brennan CW, Verhaak RGW, McKenna A, Campos B, Noushmehr H, Salama SR, et al. The somatic genomic landscape of glioblastoma. Cell, 2013, 155:462–77., DOI 10.1016/j.cell.2013.09.034 Zacher A, Kaulich K, Stepanow S, Wolter M, Kohrer K, Felsberg J, et al. Molecular diagnostics of gliomas using next generation sequencing of a glioma-tailored gene panel. Brain Pathol, 2017, 27:146–59., DOI 10.1111/bpa. 12367 AlbertoniM, Daub DM, Arden KC, Viars CS, Powell C, VanMeir EG. Genetic instability leads to loss of both p53 alleles in a human glioblastoma. Oncogene, 1998, 16:321–6., DOI 10.1038/sj.onc.1201544 Shiraishi S, Tada K, Nakamura H,Makino K, KochiM, Saya H, et al. Influence of p53 mutations on prognosis of patients with glioblastoma. Cancer, 2002, 95: 249–57., DOI 10.1002/cncr.10677 Bielski CM, Zehir A, Penson AV, Donoghue MTA, Chatila W, Armenia J, et al. Genome doubling shapes the evolution and prognosis of advanced cancers. Nat Genet, 2018, 50:1189–95., DOI 10.1038/s41588-018-0165-1 Zack TI, Schumacher SE, Carter SL, Cherniack AD, Saksena G, Tabak B, et al. Pan-cancer patterns of somatic copy number alteration. Nat Genet, 2013, 45: 1134–40., DOI 10.1038/ng.2760 Boisselier B, Dugay F, Belaud-Rotureau M-A, Coutolleau A, Garcion E, Menei P, et al. Whole genome duplication is an early event leading to aneuploidy in IDHwild type glioblastoma. Oncotarget, 2018, 9:36017–28., DOI 10.18632/oncotarget. 26330 Carter SL, Cibulskis K, Helman E, McKenna A, Shen H, Zack T, et al. Absolute quantification of somatic DNA alterations in human cancer. Nat Biotechnol, 2012, 30:413–21., DOI 10.1038/nbt.2203 Quinton RJ, DiDomizio A, Vittoria MA, Kotynkova K, Ticas CJ, Patel S, et al. Whole-genome doubling confers unique genetic vulnerabilities on tumour cells. Nature, 2021, 590:492–7., DOI 10.1038/s41586-020-03133-3 Neftel C, Laffy J, FilbinMG,Hara T, Shore ME, Rahme GJ, et al. An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell, 2019, 178: 835–49., DOI 10.1016/j.cell.2019.06.024 Wang T, Li B, Nelson CE, Nabavi S. Comparative analysis of differential gene expression analysis tools for single-cell RNA sequencing data. BMC Bioinformatics, 2019, 20:40–16., DOI 10.1186/s12859-019-2599-6 Wrzeszczynski KO, Frank MO, Koyama T, Rhrissorrakrai K, Robine N, Utro F, et al. Comparing sequencing assays and human-machine analyses in actionable genomics for glioblastoma. Neurol Genet, 2017, 3:e164–8., DOI 10.1212/NXG. 0000000000000164 Gray PN, Dunlop CL, Elliott AM. Not all next generation sequencing diagnostics are created equal: understanding the nuances of solid tumor assay design for somatic mutation detection. Cancers, 2015, 7:1313–32., DOI 10.3390/ cancers7030837 Gilly A, Southam L, Suveges D, Kuchenbaecker K, Moore R, Melloni GEM, et al. Very low-depth whole-genome sequencing in complex trait association studies. Bioinformatics, 2019, 35:2555–61., DOI 10.1093/bioinformatics/bty1032 NCCN. The NCCN clinical practice guidelines in oncology, NCCN Guidelines®, for guideline central nervous system cancers version 3.2024 © national comprehensive cancer network, inc. 2024. All rights reserved. Available from: https://www.nccn.org/, Accessed October 3, 2024). Diplas BH, He X, Brosnan-Cashman JA, Liu H, Chen LH, Wang Z, et al. The genomic landscape of TERT promoter wildtype-IDH wildtype glioblastoma. Nat Commun, 2018, 9:2087., DOI 10.1038/s41467-018-04448-6 Yeo AT, Shah R, Aliazis K, Pal R, Xu T, Zhang P, et al. Driver mutations dictate the immunologic landscape and response to checkpoint immunotherapy of glioblastoma. Cancer Immunol Res, 2023, 11:629–45., DOI 10.1158/2326-6066.cir- 22-0655 Klein AM, De Queiroz RM, Venkatesh D, Prives C. The roles and regulation of MDM2 and MDMX: it is not just about P53. Genes Dev, 2021, 35:575–601., DOI 10. 1101/gad.347872.120 Pellot Ortiz KI, Rechberger JS, Nonnenbroich LF, Daniels DJ, Sarkaria JN. MDM2 inhibition in the treatment of glioblastoma: from concept to clinical investigation. Biomedicines, 2023, 11:1879., DOI 10.3390/biomedicines11071879 An Z, Aksoy O, Zheng T, Fan Q-W, Weiss WA. Epidermal growth factor receptor, EGFR, and EGFRvIII in glioblastoma, GBM): signaling pathways and targeted therapies. Oncogene, 2018, 37:1561–75., DOI 10.1038/s41388-017-0045-7 Liu X, Chen X, Shi L, Shan Q, Cao Q, Yue C, et al. The third-generation EGFR inhibitor AZD9291 overcomes primary resistance by continuously blocking ERK signaling in glioblastoma. J Exp Clin Cancer Res, 2019, 38:219., DOI 10.1186/s13046- 019-1235-7 Choi SW, Jung HA, Cho H-J, Kim TM, Park C-K, Nam D-H, et al. A multicenter, phase II trial of GC1118, a novel anti-EGFR antibody, for recurrent glioblastoma patients with EGFR amplification. Cancer Med, 2023, 12:15788–96., DOI 10.1002/cam4.6213 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1611875 EP 1611884 DI 10.3389/pore.2024.1611875 PG 10 ER PT J AU Gyimesi, Gy Keczer, B Rein, P Horvath, M Szucs, Marjai, T Szijarto, A Hritz, I AF Gyimesi, Gyorgy Keczer, Bank Rein, Peter Horvath, Miklos Szucs, Akos Marjai, Tamas Szijarto, Attila Hritz, Istvan TI Diagnostic performance of intracystic carcinoembryonic antigen (CEA) versus glucose in differentiation of mucinous and non-mucinous pancreatic cysts SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE pancreatic cyst; endosonography; FNA; intracystic CEA; intracystic glucose ID pancreatic cyst; endosonography; FNA; intracystic CEA; intracystic glucose AB Background and Objectives: Pancreatic cysts have various potential for malignant transformation. Differentiating mucinous from non-mucinous cysts is crucial to make the right decision about further management, since mucinous cysts carry the risk of malignancy. Using endoscopic ultrasound (EUS) guided fine needle aspiration to determine intracystic carcinoembryonic antigen (CEA) levels is the recommended method for identifying mucinous cysts, although intracystic glucose assessment has also proved to be an effective tool. This study aims to compare the diagnostic performance of intracystic glucose and CEA in distinguishing between mucinous and nonmucinous pancreatic cystic lesions. Methods: In this single center study, we prospectively collected and analyzed the data of 91 consecutive patients who underwent endoscopic ultrasound (EUS)-guided fine-needle aspiration (FNA) with cytological analysis and measurement of intracystic CEA and glucose levels. The cyst type was classified based on radiological and EUS morphology, string sign, CEA, cytological and histological findings in resected cases. The diagnosis was established retrospectively by three experienced gastroenterologists blinded for glucose level in cases without definitive cytology or histology. We calculated the sensitivity, specificity, the positive- and negative predictive value of glucose and CEA respectively, and compared the two methods. Results: The sensitivity of intracystic glucose versus CEA proved to be 96.2% vs. 69.2% in identifying mucinous cysts, while the specificity of glucose was shown to be 79.5%, compared to 100% for CEA. Conclusion: Intracystic glucose is a sensitive, easily accessible biomarker in identifying mucinous pancreatic cysts, however, the specificity is lower compared to CEA. The measurement of intracystic glucose level could help in decision-making in daily clinical practice, however the diagnostic performance of the method remains inferior to “through-the-needle” techniques, such as confocal laser endomicroscopy and Moray forceps biopsy. C1 [Gyimesi, Gyorgy] Semmelweis University, School of PhD StudiesBudapest, Hungary. [Keczer, Bank] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Rein, Peter] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Horvath, Miklos] Semmelweis University, Department of Surgery, Transplantation and Gastroenterology, Division of Interventional GastroenterologyBudapest, Hungary. [Szucs, Akos] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Marjai, Tamas] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Szijarto, Attila] Semmelweis University, Department of Transplantation and SurgeryBudapest, Hungary. [Hritz, Istvan] Semmelweis University, Department of Surgery, Transplantation and Gastroenterology, Division of Interventional GastroenterologyBudapest, Hungary. RP Gyimesi, Gy (reprint author), Semmelweis University, School of PhD Studies, Budapest, Hungary. EM drgyimesigy@gmail.com CR Becker WF, Welsh RA, Pratt HS. Cystadenoma and cystadenocarcinoma of the pancreas. Ann Surg, 1965, 161(6):845–63., DOI 10.1097/00000658-196506000- 00005 Zhang XM, Mitchell DG, Dohke M, Holland GA, Parker L. Pancreatic cysts: depiction on single-shot fast spin-echo MR images. Radiology, 2002, 223(2): 547–53., DOI 10.1148/radiol.22320108153 Diagnosis and management of cystic pancreatic lesions. European evidencebased guidelines on pancreatic cystic neoplasms. Gut, 2018, 67:789–804., DOI 10. 1136/gutjnl-2018-316027 Tanaka M, Fernandez-Del Castillo C, Kamisawa T, Jang JY, Levy P, Ohtsuka T, et al. Revisions of international consensus Fukuoka guidelines for the management of IPMNof the pancreas. Pancreatology, 2017, 17:738–53., DOI 10.1016/j.pan.2017.07.007 Ohtsuka T, Del Castillo CF, Furukawa T, Hijioka S, Jang J-Y, Lennon AM, et al. International evidence-basedKyoto guidelines for the management of intraductal papillary mucinous neoplasm of the pancreas 2024;24(2):255–70., DOI 10.1016/j.pan.2023.12.009 Dushyant VS, Kambadakone A, Macari M, Takahashi N, Chari S, Fernandez-del Castillo C. Diagnosis and management of cystic pancreatic lesions Am. J Roentgenol, 2013, 200(2):343–54., DOI 10.2214/AJR.12.8862 Brendan CV1, Yeh BM, Qayyum A, Way LW, McCulloch CE, Coakley FV. Characterization of cystic pancreatic masses: relative accuracy of CT and MRI. Am J Roentgenol, 2007, 189(3):648–56., DOI 10.2214/AJR.07.2365 McCarty TR, Garg R, Rustagi T. Pancreatic cyst fluid glucose in differentiating mucinous from nonmucinous pancreatic cysts: a systematic review and meta-analysis. Gastrointest Endosc, 2021, 94(698–712):698–712.e6., DOI 10.1016/j.gie.2021.04.025 Faias S, Cravo M, Chaves PL. Comparative analysis of glucose and carcinoembryonic antigen in the diagnosis of pancreatic mucinous cysts: a systematic review and metaanalysis. Gastrointest Endosc, 2021, 94(2):235–47., DOI 10.1016/j.gie.2021.03.935 Guzman CE, Martinez MB, Casellas JA, Sparicio JR. Intracystic glucose levels appear useful for diagnosis of pancreatic cystic lesions: a systematic review and meta-analysis. Dig Dis Sci, 2022, 67(6):2562–70., DOI 10.1007/s10620-021-07035-w BruggeWR, Lewandrowski K, Lee-Lewandrowski E, CentenoBA, SzydloT,Regan S, et al. Diagnosis of pancreatic cystic neoplasms: a report of the cooperative pancreatic cyst study. Gastroenterology, 2004, 126:1330–6., DOI 10.1053/j.gastro.2004.02.013 Zikos T, Pham K, Bowen R, Chen AM, Banerjee S, Friedland S, et al. Cyst fluid glucose is rapidly feasible and accurate in diagnosing mucinous pancreatic cysts. Am J Gastroenterol, 2015, 110:909–14., DOI 10.1038/ajg.2015.148 Lekkerkerker SJ, Besselink MG, Busch OR, Verheij J, Engelbrecht MR, Rauws EA, et al. Comparing 3 guidelines on the management of surgically removed pancreatic cysts with regard to pathological outcome. Gastrointest Endosc, 2017, 85(5):1025–31., DOI 10.1016/j.gie.2016.09.027 van Huijgevoort NCM, Hoogenboom SA, Lekkerkerker SJ, Topazian M, Chandrasekhara V, Morris-Stif G, et al. The diagnostic accuracy of carcinoembryonic antigen in differentiating mucinous and non-mucinous pancreatic cystic neoplasms—a systematic review and individual patient data meta-analysis. In: ESGE days 2018 accepted abstracts. Budapest, Hungary, New York City, NY: Endoscopy, 2018). Park WG, Wu M, Bowen R, Zheng M, Fitch WL, Pai RK, et al. Metabolomic-derived novel cyst fluid biomarkers for pancreatic cysts: glucose and kynurenine. Gastrointest Endosc, 2013, 78(2):295–302., DOI 10. 1016/j.gie.2013.02.037 Boot C. A review of pancreatic cyst fluid analysis in the differential diagnosis of pancreatic cyst lesions. Ann Clin Biochem, 2014, 51:151–66., DOI 10.1177/ 0004563213503819 Singhi AD, McGrath K, Brand RE, Khalid A, Zeh HJ, Chennat JS, et al. Preoperative next-generation sequencing of pancreatic cyst fluid is highly accurate in cyst classification and detection of advanced neoplasia. Gut, 2018, 67:2131–41., DOI 10.1136/gutjnl-2016-313586 Rockacy MJ, Zahid M, McGrath KM, Fasanella KE, Khalid A. Association between KRASmutation, detected in pancreatic cyst fluid, and long-term outcomes of patients. Clin Gastroenterol Hepatol, 2013, 11:425–9., DOI 10.1016/j.cgh.2012. 12.008 Nikiforova MN, Khalid A, Fasanella KE, McGrath KM, Brand RE, Chennat JS, et al. Integration of KRAS testing in the diagnosis of pancreatic cystic lesions: a clinical experience of 618 pancreatic cysts. Mod Pathol, 2013, 26:1478–87., DOI 10. 1038/modpathol.2013.91 Li S-Y, Wang Z-J, Pan C-Y, Wu C, Li Z-S, Jin Z-D, et al. Comparative performance of endoscopic ultrasound-based techniques in patients with pancreatic cystic lesions: a network meta-analysis. Am J Gastroenterol, 2023, 118(2):243–55., DOI 10.14309/ajg.0000000000002088 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1611881 EP 1611887 DI 10.3389/pore.2024.1611881 PG 7 ER PT J AU Czeti, Sashalmi, S Takacs, F Szaloki, G Kriston, Cs Varga, G Farkas, P Hamed, A Mark, Barna, G AF Czeti, Agnes Sashalmi, Soma Takacs, Ferenc Szaloki, Gabor Kriston, Csilla Varga, Gergely Farkas, Peter Hamed, Aryan Mark, Agnes Barna, Gabor TI Investigating the effect of immunomagnetic separation on the immunophenotype and viability of plasma cells in plasma cell disorders SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE multiple myeloma; flow cytometry; immunomagnetic separation; immunophenotype; positive selection ID multiple myeloma; flow cytometry; immunomagnetic separation; immunophenotype; positive selection AB Plasma cell enrichment plays a pivotal role in the accurate prognosis and molecular characterization of multiple myeloma. The separation is commonly carried out by positive cell selection using CD138 monoclonal antibody conjugated to magnetic beads. Optimally, during the separation procedure, the cells should neither be damaged, nor should their phenotype be significantly altered, as these changes would falsify the results if the isolated cells were subsequently used. For this reason, we investigated the expression patterns of different surface markers by flow cytometry before and after magnetic isolation using bone marrow or peripheral blood samples from 12 patients with plasma cell disorders. The selected markers are not only used as backbone markers in routine diagnostics (CD19, CD38, CD45, CD117, and CD138), but they also play an important role in cell adhesion and connection with microenvironment (CD44, CD49d, CD56, and CD81) or possibly drug resistance (CD69, CD86, and CD184), making them promising targets for myeloma research. Moreover, we examined the effects of separation on cell viability in 8 cases. The intensities of 8 out of the 12 investigated markers were slightly influenced, while CD138, CD38, CD56, and CD184 were changed significantly, however the immunophenotype of the cells was not changed. Positive markers remained positive and negative ones remained negative after the separation procedure. In addition, the number of apoptotic plasma cells was significantly reduced during separation, facilitating further examination of the cells. Our results showed that magnetic isolation can be considered as a reliable option but the immunophenotype of plasma cells should be validated after the separation if the intensities of the markers are important for further experiments. C1 [Czeti, Agnes] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sashalmi, Soma] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Takacs, Ferenc] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Szaloki, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Kriston, Csilla] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Varga, Gergely] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Farkas, Peter] Semmelweis University, Department of Internal Medicine and HaematologyBudapest, Hungary. [Hamed, Aryan] Petz Aladar County Hospital, II. Department of Internal Medicine and HematologyGyor, Hungary. [Mark, Agnes] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Barna, Gabor] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Barna, G (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM barna.gabor@semmelweis.hu CR Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin, 2019, 69:7–34., DOI 10.3322/caac.21551 Tai Y, Teoh G, Shima Y, Chauhan D, Treon SP, Raje N, et al. Isolation and characterization of human multiple myeloma cell enriched populations. J Immunol Methods, 2000, 235:11–9., DOI 10.1016/s0022-1759(99)00199-4 Shin SY, Jang S, Park C, Chi H, Lee J, Lee JH, et al. Application of an immunemagnetic cell sorting method for CD138-positive plasma cells in FISH analysis of multiple myeloma. Int J Lab Hematol, 2012, 34:541–6., DOI 10.1111/j.1751-553X. 2012.01433.x Kishimoto RK, de Freitas SLVV, Ratis CA, Borri D, Sitnik R, Edrp V. Validation of interphase fluorescence in situ hybridization, iFISH, for multiple myeloma using CD138 positive cells. Rev Bras Hematol Hemoter, 2016, 38:113–20., DOI 10.1016/j. bjhh.2016.01.005 Zhan F, Huang Y, Colla S, Stewart JP, Hanamura I, Gupta S, et al. The molecular classification of multiple myeloma. Blood, 2006, 108:2020–8., DOI 10. 1182/blood-2005-11-013458 Mattioli M, Agnelli L, Fabris S, Baldini L, Morabito F, Bicciato S, et al. Gene expression profiling of plasma cell dyscrasias reveals molecular patterns associated with distinct IGH translocations in multiple myeloma. Oncogene, 2005, 24: 2461–73., DOI 10.1038/sj.onc.1208447 Cumova J, Kovarova L, Potacova A, Buresova I, Kryukov F, Penka M, et al. Optimization of immunomagnetic selection of myeloma cells from bone marrow using magnetic activated cell sorting. Int J Hematol, 2010, 92:314–9., DOI 10.1007/ s12185-010-0651-4 Wijdenes J, Vooijs WC, Clement C, Post J, Morard F, Vita N, et al. A plasmocyte selective monoclonal antibody, B-B4, recognizes syndecan-1. Br J Haematol, 1996, 94:318–23., DOI 10.1046/j.1365-2141.1996.d01-1811.x Potecova A, Stossova J, Buresova I, Kovarova L, Almasi M, Penka M, et al. Sample processing and methodological pitfalls in multiple myeloma research. Klin Onkol, 2011, 24:18–23. Bansal R, Kimlinger T, Gyotoku KA, Smith M, Rajkumar V, Kumar S. Impact of CD138 magnetic bead-based positive selection on bone marrow plasma cell surface markers. Clin Lymphoma Myeloma Leuk, 2021, 21:e48–e51., DOI 10.1016/j.clml.2020.08.003 Kimlinger TK, Rajkumar SV, Kline MP, Haug JL, Timm MM, Witzig TE, et al. Changes in bone marrow plasma cell surface markers following cd138-magnetic bead based positive selection. Blood, 2006, 108:3516., DOI 10.1182/blood.v108.11. 3516.3516 Flores-Montero J, de Tute R, Paiva B, Juan Perez J, Wind H, Sanoja L, et al. Immunophenotype of normal vs. myeloma plasma cells: toward antibody panel specifications for MRD detection in multiple myeloma. Euroflow Cytom B Clin Cytom, 2016, 90:61–72., DOI 10.1002/cyto.b.21265 Tatsumi T, Shimazaki C, Goto H, Araki SI, Sudo Y, Yamagata N, et al. Expression of adhesion molecules on myeloma cells. Jpn J. Cancer Res, 1996, 87: 837–42., DOI 10.1111/j.1349-7006.1996.tb02108.x Bjorklund CC, Baladandayuthapani V, Lin HY, Jones RJ, Kuiatse I, Wang H, et al. Evidence of a role for CD44 and cell adhesion in mediating resistance to lenalidomide in multiple myeloma: therapeutic implications. Leukemia, 2014, 28: 373–83., DOI 10.1038/leu.2013.174 Mark A, Varga G, Timar B, Kriston C, Szabo O, Deak L, et al. The effect of microenvironmental factors on the development of myeloma cells. Hematol Oncol, 2017, 35:741–5., DOI 10.1002/hon.2354 Buda G, Carulli G, Orciuolo E, Sammuri P, Galimberti S, Rousseau M, et al. CD69 expression predicts favorable outcome in multiple myeloma patients treated with VTD. Blood, 2015, 126:1768., DOI 10.1182/blood.v126. 23.1768.1768 Stessman HAF, Mansoor A, Zhan F, Linden MA, Van Ness B, Baughn LB. Bortezomib resistance can be reversed by induced expression of plasma cell maturation markers in a mouse in vitro model of multiple myeloma. PLoS One, 2013, 8:e77608., DOI 10.1371/journal.pone.0077608 Moser-Katz T, Gavile CM, Barwick BG, Lee KP, Boise LH. PDZ proteins SCRIB and DLG1 regulate myeloma cell surface CD86 expression, growth, and survival. Mol Cancer Res, 2022, 20:1122–36., DOI 10.1158/1541-7786.MCR-21- 0681 Ullah TR. The role of CXCR4 in multiple myeloma: cells’ journey from bone marrow to beyond. J Bone Oncol, 2019, 17:100253., DOI 10.1016/j.jbo.2019.100253 McKenna RW, Kyle RA, Kuehl WM, Harris NL, Coupland RWFF. Plasm cell neoplasms. In: Swerdlow SH, Campo NLH E, editors. WHO Classif. tumors Haematop. lymphoid tissues. 4th ed. Lyon, 2017). p. 241–58. Rajkumar SV, DimopoulosMA, Palumbo A, Blade J,Merlini G,MateosMV, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol, 2014, 15:e538–48., DOI 10.1016/S1470-2045(14)70442-5 Anon. EasySepTM human WB and BM CD138 positive selection kit II | STEMCELL technologies, 2022). Available from: https://www.stemcell.com/ easysep-human-whole-blood-and-bone-marrow-cd138-positive-selection-kit-ii. html#section-data-and-publications, Accessed August 21, 2022). Hayashida K, Bartlett AH, Chen Y, Park PW. Molecular and cellular mechanisms of ectodomain shedding. Anat Rec, 2010, 293:925–37., DOI 10.1002/ ar.20757 Fitzgerald ML, Wang Z, Park PW, Murphy G, Bernfield M. Shedding of syndecan-1 and-4 ectodomains is regulated by multiple signaling pathways and mediated by a TIMP-3-sensitive metalloproteinase, 2000). 811–24. Jourdan M, Ferlin M, Legouffe E, Horvathova M, Liautard J, Rossi JF, et al. The myeloma cell antigen syndecan-1 is lost by apoptotic myeloma cells. Br J Haematol, 1998, 100:637–46., DOI 10.1046/j.1365-2141.1998. 00623.x Kimlinger TK, Rajkumar SV, Kline M, Timm MM, Haug JL,Witzig TE, et al. Apoptosis rates in myeloma cells can Be significantly affected by CD138 sorting and by the specific method used to detect apoptotic cells. Blood, 2006, 108:3442., DOI 10. 1182/blood.v108.11.3442.3442 Piedra-Quintero ZL, Wilson Z, Nava P, Guerau-de-Arellano M. CD38: an immunomodulatory molecule in inflammation and autoimmunity. Front Immunol, 2020, 11:3–6., DOI 10.3389/fimmu.2020.597959 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1611882 EP 1611891 DI 10.3389/pore.2024.1611882 PG 10 ER PT J AU Molnar, E Baranyi, M Szigeti, K Hegedus, L Bordas, F Gabriel, Zs Petenyi, G Tovari, J Hegedus, B Timar, J AF Molnar, Eszter Baranyi, Marcell Szigeti, Krisztina Hegedus, Luca Bordas, Fanni Gabriel, Zsofia Petenyi, Greta Tovari, Jozsef Hegedus, Balazs Timar, Jozsef TI Combination of farnesyl-transferase inhibition with KRAS G12D targeting breaks down therapeutic resistance in pancreatic cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE PDAC; G12D mutant KRAS; KRAS inhibitor resistance; FTI; combination therapy ID PDAC; G12D mutant KRAS; KRAS inhibitor resistance; FTI; combination therapy AB Pancreatic adenocarcinoma is one of the deadliest forms of cancer with no effective therapeutic options. A KRAS mutation can be found in up to 90% of all pancreatic tumors, making it a promising therapeutic target. The introduction of new KRAS inhibitors has been a milestone in the history of KRAS mutant tumors; however, therapeutic resistance limits their efficacy. Thus, new therapeutic options, including combination therapies, are urgently needed. Recently, we have shown that KRAS G12C inhibitors in combination with farnesyl-transferase inhibitors exert synergistic antitumor effects. Here, we provide evidence for the feasibility of this combinational approach to break down resistance in KRAS G12D mutant pancreatic cancer. Although we have shown that the 3D environment dramatically sensitizes cells to MRTX1133 treatment, the synergistic effect of this drug combination is present in both 2D and 3D in the PANC1 pancreatic adenocarcinoma model, which showed high resistance to MRTX1133 in 2D. The effects of the combination treatment show an association with the inhibition of farnesylated regulatory proteins, including HRAS and RHEB, along with the expression level of KRAS. Our study warrants further investigation for the potential applicability of KRAS G12D inhibitors in combination with farnesyl-transferase inhibitors for the treatment of KRAS mutant pancreatic adenocarcinoma. C1 [Molnar, Eszter] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Baranyi, Marcell] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Szigeti, Krisztina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Hegedus, Luca] University Duisburg-Essen, Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Bordas, Fanni] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Gabriel, Zsofia] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Petenyi, Greta] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Tovari, Jozsef] Orszagos Onkologiai Intezet, Nemzeti Tumorbiologiai LaboratoriumBudapest, Hungary. [Hegedus, Balazs] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Timar, Jozsef] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Baranyi, M (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM baranyi2marci@gmail.com CR Wood LD, CantoMI, Jaffee EM, Simeone DM. Pancreatic cancer: pathogenesis, screening, diagnosis, and treatment. Gastroenterology, 2022, 163(2):386–402 e1., DOI 10.1053/j.gastro.2022.03.056 HuHF, Ye Z, Qin Y, Xu XW, Yu XJ, Zhuo QF, et al.Mutations in key driver genes of pancreatic cancer: molecularly targeted therapies and other clinical implications. Acta Pharmacol Sin, 2021, 42(11):1725–41., DOI 10.1038/s41401- 020-00584-2 Golan T, Hammel P. Management of BRCA mutation carriers with pancreatic adenocarcinoma. J Natl Compr Canc Netw, 2021, 19(4):469–73., DOI 10.6004/jnccn. 2021.7031 Tran LC, Ozdemir BC, Berger MD. The role of immune checkpoint inhibitors in metastatic pancreatic cancer: current state and outlook. Pharmaceuticals, Basel,, 2023, 16(10):1411., DOI 10.3390/ph16101411 Wong W, Alouani E, Wei A, Ryu YK, Chabot JA, Manji GA. Future of immunotherapy in pancreas cancer and the trials, tribulations and successes thus far. Semin Oncol, 2021, 48(1):57–68., DOI 10.1053/j.seminoncol.2021. 02.007 Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, et al. Pancreatic cancer. Nat Rev Dis Primers, 2016, 2:16022., DOI 10.1038/nrdp.2016.22 Timar J, Kashofer K. Molecular epidemiology and diagnostics of KRAS mutations in human cancer. Cancer Metastasis Rev, 2020, 39(4):1029–38., DOI 10.1007/s10555-020-09915-5 Temko D, Tomlinson IPM, Severini S, Schuster-Bockler B, Graham TA. The effects of mutational processes and selection on driver mutations across cancer types. Nat Commun, 2018, 9(1):1857., DOI 10.1038/s41467-018-04208-6 Liu P, Wang Y, Li X. Targeting the untargetable KRAS in cancer therapy. Acta Pharm Sin B, 2019, 9(5):871–9., DOI 10.1016/j.apsb.2019.03.002 Rathod LS, Dabhade PS, Mokale SN. Recent progress in targeting KRAS mutant cancers with covalent G12C-specific inhibitors. Drug Discov Today, 2023, 28(5):103557., DOI 10.1016/j.drudis.2023.103557 Zheng Q, Zhang Z, Guiley KZ, Shokat KM. Strain-release alkylation of Asp12 enables mutant selective targeting of K-Ras-G12D. Nat Chem Biol, 2024, 20: 1114–22., DOI 10.1038/s41589-024-01565-w Wei D, Wang L, Zuo X, Maitra A, Bresalier RS. A small molecule with big impact: MRTX1133 targets the KRASG12D mutation in pancreatic cancer. Clin Cancer Res, 2024, 30(4):655–62., DOI 10.1158/1078-0432.CCR-23-2098 Hallin J, Bowcut V, Calinisan A, Briere DM, Hargis L, Engstrom LD, et al. Anti-tumor efficacy of a potent and selective non-covalent KRAS(G12D, inhibitor. Nat Med, 2022, 28(10):2171–82., DOI 10.1038/s41591-022-02007-7 Ye W, Lu X, Qiao Y, Ou WB. Activity and resistance to KRAS(G12C, inhibitors in non-small cell lung cancer and colorectal cancer. Biochim Biophys Acta Rev Cancer, 2024, 1879(3):189108., DOI 10.1016/j.bbcan. 2024.189108 Ryan MB, Coker O, Sorokin A, Fella K, Barnes H,Wong E, et al. KRAS(G12C)- independent feedback activation of wild-type RAS constrains KRAS(G12C, inhibitor efficacy. Cell Rep, 2022, 39(12):110993., DOI 10.1016/j.celrep.2022.110993 Gulay KCM, Zhang X, Pantazopoulou V, Patel J, Esparza E, Pran Babu DS, et al. Dual inhibition of KRASG12D and pan-ERBB is synergistic in pancreatic ductal adenocarcinoma. Cancer Res, 2023, 83(18):3001–12., DOI 10.1158/0008-5472. CAN-23-1313 Kataoka M, Kitazawa M, Nakamura S, Koyama M, Yamamoto Y, Miyazaki S, et al. Cetuximab enhances the efficacy of MRTX1133, a novel KRAS(G12D, inhibitor, in colorectal cancer treatment. Anticancer Res, 2023, 43(10):4341–8., DOI 10.21873/anticanres.16629 Feng J, Hu Z, Xia X, Liu X, Lian Z, Wang H, et al. Feedback activation of EGFR/wild-type RAS signaling axis limits KRAS(G12D, inhibitor efficacy in KRAS(G12D)-mutated colorectal cancer. Oncogene, 2023, 42(20):1620–33., DOI 10.1038/s41388-023-02676-9 Dilly J, Hoffman MT, Abbassi L, Li Z, Paradiso F, Parent BD, et al. Mechanisms of resistance to oncogenic KRAS inhibition in pancreatic cancer. Cancer Discov, 2024, 14:2135–61., DOI 10.1158/2159-8290.CD-24-0177 Punekar SR, Velcheti V, Neel BG, Wong KK. The current state of the art and future trends in RAS-targeted cancer therapies. Nat Rev Clin Oncol, 2022, 19(10): 637–55., DOI 10.1038/s41571-022-00671-9 Baranyi M, Molnar E, Hegedus L, Gabriel Z, Petenyi FG, Bordas F, et al. Farnesyl-transferase inhibitors show synergistic anticancer effects in combination with novel KRAS-G12C inhibitors. Br J Cancer, 2024, 130(6):1059–72., DOI 10.1038/ s41416-024-02586-x Zheng S, Wang W, Aldahdooh J, Malyutina A, Shadbahr T, Tanoli Z, et al. SynergyFinder plus: toward better interpretation and annotation of drug combination screening datasets. Genomics Proteomics Bioinformatics, 2022, 20(3):587–96., DOI 10.1016/j.gpb.2022.01.004 Almacellas E, Pelletier J, Day C, Ambrosio S, Tauler A, Mauvezin C. Lysosomal degradation ensures accurate chromosomal segregation to prevent chromosomal instability. Autophagy, 2021, 17(3):796–813., DOI 10.1080/ 15548627.2020.1764727 Janes MR, Zhang J, Li LS, Hansen R, Peters U, Guo X, et al. Targeting KRAS mutant cancers with a covalent G12C-specific inhibitor. Cell, 2018, 172(3): 578–89.e17., DOI 10.1016/j.cell.2018.01.006 Brancato V, Oliveira JM, Correlo VM, Reis RL, Kundu SC. Could 3D models of cancer enhance drug screening? Biomaterials, 2020, 232:119744., DOI 10.1016/j. biomaterials.2019.119744 Han K, Pierce SE, Li A, Spees K, Anderson GR, Seoane JA, et al. CRISPR screens in cancer spheroids identify 3D growth-specific vulnerabilities. Nature, 2020, 580(7801):136–41., DOI 10.1038/s41586-020-2099-x Tidwell TR, Rosland G, Tronstad KJ, Soreide K, Hagland HR. Comparing in vitro cytotoxic drug sensitivity in colon and pancreatic cancer using 2D and 3D cell models: contrasting viability and growth inhibition in clinically relevant dose and repeated drug cycles. Cancer Med, 2024, 13(11):e7318., DOI 10.1002/cam4.7318 Kumarasamy V, Wang J, Frangou C, Wan Y, Dynka A, Rosenheck H, et al. The extracellular niche and tumor microenvironment enhance KRAS inhibitor efficacy in pancreatic cancer. Cancer Res, 2024, 84(7):1115–32., DOI 10.1158/0008- 5472.CAN-23-2504 Cao Z, Liao Q, Su M, Huang K, Jin J, Cao D. AKT and ERK dual inhibitors: the way forward? Cancer Lett, 2019, 459:30–40., DOI 10.1016/j.canlet.2019. 05.025 Baranyi M, Buday L, Hegedus B. K-Ras prenylation as a potential anticancer target. Cancer Metastasis Rev, 2020, 39(4):1127–41., DOI 10.1007/s10555-020- 09902-w NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1611948 EP 1611959 DI 10.3389/pore.2024.1611948 PG 12 ER PT J AU Katib, Y Essatari, M AF Katib, Yousef Essatari, Murad TI Case report: A rare case of oral sebaceous carcinoma in the upper lip SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE sebaceous carcinoma; oral cavity; lip; immunohistochemistry; treatment; oral cavity ID sebaceous carcinoma; oral cavity; lip; immunohistochemistry; treatment; oral cavity AB Sebaceous carcinoma (SC) is a rare aggressive malignant tumor that originates in the adnexal epithelium of the sebaceous gland. While occurrences on the lips are extremely uncommon, there have been a few reported cases in the literature. Our case involves a 47-year-old smoker male who presented with a painless, non-mobile lesion on his upper lip that had been present for 12 months. Upon clinical examination, an ulcerated, exophytic, and irregularly shaped mass was observed on the upper lip. No other intraoral lesions were found. An incisional biopsy was performed, revealing a malignant tumor with a nodular pattern consisting of basaloid cells with obvious sebaceous differentiations and frequent mitoses. The neoplastic cells tested positive for broad-spectrum cytokeratin (AE1-AE3), epithelial membrane antigen (EMA), and P53, while testing negative for S-100 and carcinoembryonic antigen (CEA). Based on these results, a diagnosis of SC of the upper lip was made. This case report and review aimed to describe the histogenesis, unique clinicopathological features, and current treatment options for SC. C1 [Katib, Yousef] Taibah University, College of MedicineMadinah, Saudi Arabia. [Essatari, Murad] Prince Noorah Oncology Center, King Abdulaziz Medical CityJeddah, Saudi Arabia. RP Katib, Y (reprint author), Taibah University, College of Medicine, Madinah, Saudi Arabia. EM ykatib@taibahu.edu.sa CR Dubovy S, Eberhart CG, Vemuganti G. Sebaceous carcinoma of the conjunctiva and caruncle. In: WHO classification of tumours of the eye. 4th ed. Lyon: IARC, 2018). p. pp25–26. Bailet JW, Zimmerman MC, Arnstein DP, Wollman JS, Mickel RA. Sebaceous carcinoma of the head and neck. Case report and literature review. Arch Otolaryngol Head Neck Surg, 1992, 118(11):1245–9., DOI 10.1001/archotol.1992. 01880110113020 Alawi F, Siddiqui A. Sebaceous carcinoma of the oral mucosa: case report and review of the literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod, 2005, 99(1):79–84., DOI 10.1016/j.tripleo.2004.05.007 Handschel J, Herbst H, Brand B, Meyer U, Piffko J. Intraoral sebaceous carcinoma. Br J Oral Maxillofac Surg, 2003, 41(2):84–7., DOI 10.1016/s0266- 4356(03)00036-6 Cicinelli MV, Kaliki S. Ocular sebaceous gland carcinoma: an update of the literature. Int Ophthalmol, 2019, 39(5):1187–97., DOI 10.1007/s10792-018-0925-z Papadimitriou I, Vakirlis E, Sotiriou E, Bakirtzi K, Lallas A, Ioannides D. Sebaceous neoplasms. Diagnostics, Basel,, 2023, 13(10):1676., DOI 10.3390/ diagnostics13101676 Esnal Leal F, Garcia-Rostan y Perez GM, Garatea Crelgo J, Gorriaran Terreros M, Arzoz Sainz de Murieta E. Sebaceous carcinoma of salivary gland. Report of two cases of infrequent location. An Otorrinolaringol Ibero Am, 1997, 24(4):401–13. Halperin V, Kolas S, Jefferis KR, Huddleston SO, Robinson HB. The occurrence of Fordyce spots, benign migratory glossitis, median rhomboid glossitis, and fissured tongue in 2,478 dental patients. Oral Surg Oral Med Oral Pathol, 1953, 6(9):1072–7., DOI 10.1016/0030-4220(53)90220-5 Lu Q, Fu XY, Huang Y. Sebaceous carcinoma of the right palate: case report and literature review. Gland Surg, 2021, 10(5):1819–25., DOI 10.21037/gs-21-218 Damm DD, O’Connor WN, White DK, Drummond JF, Morrow LW, Kenady DE. Intraoral sebaceous carcinoma. Oral Surg Oral Med Oral Pathol, 1991, 72(6): 709–11., DOI 10.1016/0030-4220(91)90016-6 Innocenzi D, Balzani A, Lupi F, Panetta C, Skroza N, Cantoresi F, et al. Morpheaform extraocular sebaceous carcinoma. J Surg Oncol, 2005, 92(4):344–6., DOI 10.1002/jso.20383 Greenall CJ, Drage NA. Sebaceous carcinoma of the lip: comparing normal lip and cheek anatomy with the imaging features of a rare cutaneous malignancy. Ultrasound, 2015, 23(2):126–9., DOI 10.1177/1742271X15569295 Arai N, Tomihara K, Atakei R, Noto Z, Wada S, NoGuchi M. A case of sebaceous carcinoma of the lower lip. Jpn J Oral Maxillofacial Surg, 2015, 61(Issue 4):217–21. Released on J-STAGE October 06, 2015, Online ISSN 2186-1579, Print ISSN 0021 5163., DOI 10.5794/jjoms.61.217 Di Cosola M, Spirito F, AmbrosinoM, Somma P, Santarelli A, Staibano S, et al. Sebaceous carcinoma of the lip: a case report and review of the literature. JMed Case Rep, 2022, 16(1):241., DOI 10.1186/s13256-022-03435-2 Benedict K, Al Hmada Y, Gordon C, Hoppe I. Squamous cell carcinoma admixed with sebaceous carcinoma of upper lip in a 7-year-old female. Ped Hem Onco J, 2022, 7(4):p126–9., DOI 10.1016/j.phoj.2022.08.002 Everett JN, Raymond VM, Dandapani M, Marvin M, Kohlmann W, Chittenden A, et al. Screening for germline mismatch repair mutations following diagnosis of sebaceous neoplasm. JAMA Dermatol, 2014, 150(12): 1315–21., DOI 10.1001/jamadermatol.2014.1217 Dasgupta T, Wilson LD, Yu JB. A retrospective review of 1349 cases of sebaceous carcinoma. Cancer, 2009, 115:158–65., DOI 10.1002/cncr.23952 Bassetto F, Baraziol R, Sottosanti MV, Scarpa C, Montesco M. Biological behavior of the sebaceous carcinoma of the head. Dermatol Surg, 2004, 30(3): 472–6., DOI 10.1111/j.1524-4725.2004.30025.x Duman DG, Ceyhan BB, Celikel T, Ahiskali R. Extraorbital sebaceous carcinoma with rapidly developing visceral metastases. Dermatol Surg, 2003, 29(9):987–9., DOI 10.1046/j.1524-4725.2003.29268.x DogruM,Matsuo H, Inoue M, Okubo K, Yamamoto M.Management of eyelid sebaceous carcinomas. Ophthalmologica, 1997, 211(1):40–3., DOI 10.1159/000310872 Abuzeid M, Gangopadhyay K, Rayappa CS, Antonios JI. Intraoral sebaceous carcinoma. J Laryngol Otol, 1996, 110(5):500–2., DOI 10.1017/s0022215100134103 Belaid A, Nasr C, BennaM, CherifA, JmourO, Bouguila H, et al. Radiation therapy for primary eyelid cancers in Tunisia. Asian Pac J Cancer Prev, 2016, 17(7):3643–6., DOI 10.14456/apjcp.2016.148/APJCP.2016.17.7.3643 Takeuchi D, Ishida M, Yasuda E, Ueda K, Hirose Y. Ocular and extraocular sebaceous carcinomas: a retrospective study with emphasis on the presence of in situ lesion and discussion and review of the histogenesis of extraocular sebaceous carcinoma. Oncol Lett, 2023, 26(2):337. PMID: 37427342; PMCID: PMC10326651., DOI 10.3892/ol.2023.13923 Lee MJ, Kim YC, Lew W. A case of superficial epithelioma with sebaceous differentiation. Yonsei Med J, 2003, 44(2):347–50., DOI 10.3349/ymj.2003.44.2.347 Friedman KJ, Boudreau S, Farmer ER. Superficial epithelioma with sebaceous differentiation. J Cutan Pathol, 1987, 14(4):193–7., DOI 10.1111/j.1600-0560.1987.tb01331.x Ohara N, Taguchi K, YamamotoM, Nagano T, Akagi T. Sebaceous carcinoma of the submandibular gland with high-grade malignancy: report of a case. Pathol Int, 1998, 48(4):287–91., DOI 10.1111/j.1440-1827.1998.tb03907.x Siriwardena BS, Tilakaratne WM, Rajapakshe RM. A case of sebaceous carcinoma of the parotid gland. J Oral Pathol Med, 2003, 32(2):121–3., DOI 10. 1034/j.1600-0714.2003.00037.x NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1611968 EP 1611972 DI 10.3389/pore.2024.1611968 PG 5 ER PT J AU Kramer, Zs Budai, A Pesti, A Kulka, J Tokes, AM AF Kramer, Zsofia Budai, Andras Pesti, Adrian Kulka, Janina Tokes, Anna-Maria TI Invasive micropapillary carcinoma of the breast and invasive breast carcinoma of no special type: a comparison of claudin proteins’ expression and its impact on survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE invasive micropapillary carcinoma of the breast; invasive breast carcinoma of no special type; claudin expression; tight junction; prognosis ID invasive micropapillary carcinoma of the breast; invasive breast carcinoma of no special type; claudin expression; tight junction; prognosis AB Invasive micropapillary carcinoma of the breast is characterized by clusters of cells presenting with inverted polarity. Although the apico–basal polarity is a fundamental property of the epithelium, the biological alterations leading to the inside-out pattern observed in invasive micropapillary carcinoma (IMPC) remain mostly unknown. The regulation of tight junctions in polarity formation and maintenance is acknowledged. By using immunohistochemistry, we have analysed claudin-1, -3, -4, and -7 tight junction proteins expression and their prognostic value on IMPCs and compared them to invasive breast carcinomas of no special type (IBC-NST) tumors. Our cohort consisted of 37 IMPCs, 36 IBC-NST and 9 mixed IMPC/IBC-NST tumors. Two scoring systems were used to quantify protein expression: a 4-tier scoring system and the H-score method. Distant metastasis free survival (DMFS) intervals and overal survival (OS) data were used for prognosis evaluation. The analysed samples were characterized mainly by low or no claudin-1 expression whereas claudins-3, -4 and -7 showed variable positivity. We have found no significant differences in claudin-3 and -4 protein expression between IMPC and IBC-NST groups with either scoring methods, however high claudin-7 expression was found in significantly more IMPCs than IBC-NST tumors according to the H-score system (p = 0.02). The 4-tier scoring method revealed association of claudin-7 expression with molecular tumor subtypes (p = 0.001). IMPC and IBC-NST tumors did not show difference in DMFS (p = 0.70). In the analysis of pure IMPC and IBC-NST tumors, positive/high claudin-4 protein expression was significantly associated with shorter DMFS (p = 0.02/p = 0.008, respectively according to the two scoring methods). Claudin-3 and claudin-7 expression showed no association with DMFS or OS. Changes in epithelial polarity seem not to be related to claudin-1, -3, and -4 expression. Increased claudin-4 expression may have a role in breast cancer progression. C1 [Kramer, Zsofia] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Budai, Andras] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Pesti, Adrian] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Kulka, Janina] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Tokes, Anna-Maria] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. RP Kramer, Zs (reprint author), Semmelweis University, 2nd Department of Pathology, Budapest, Hungary. EM kramer.zsofia@semmelweis.hu CR Cakir E, Yilmaz A, Demirag F, Oguztuzun S, Sahin S, Yazici UE, et al. Prognostic significance of micropapillary pattern in lung adenocarcinoma and expression of apoptosis-related markers: caspase-3, bcl-2, and p53. APMIS, 2011, 119(9):574–80., DOI 10.1111/j.1600-0463.2011.02778.x Comperat E, Roupret M, Yaxley J, Reynolds J, Varinot J, Ouzaid I, et al. Micropapillary urothelial carcinoma of the urinary bladder: a clinicopathological analysis of 72 cases. Pathology, 2010, 42(7):650–4., DOI 10.3109/00313025.2010. 522173 Fisher ER, Palekar AS, Redmond C, Barton B, Fisher B. Pathologic findings from the national surgical adjuvant breast project, protocol no. 4). VI. Invasive papillary cancer. Am J Clin Pathol, 1980, 73(3):313–22., DOI 10.1093/ajcp/73.3.313 Guzinska-Ustymowicz K, Niewiarowska K, Pryczynicz A. Invasive micropapillary carcinoma: a distinct type of adenocarcinomas in the gastrointestinal tract. World J Gastroenterol, 2014, 20(16):4597–606., DOI 10. 3748/wjg.v20.i16.4597 Chen H,Wu K,Wang M,Wang F, Zhang M, Zhang P. Invasive micropapillary carcinoma of the breast has a better long-term survival than invasive ductal carcinoma of the breast in spite of its aggressive clinical presentations: a comparison based on large population database and case-control analysis. Cancer Med, 2017, 6(12):2775–86., DOI 10.1002/cam4.1227 Wu SG, Zhang WW, Sun JY, Li FY, Chen YX, He ZY. Postoperative radiotherapy for invasive micropapillary carcinoma of the breast: an analysis of Surveillance, Epidemiology, and End Results database. Cancer Manag Res, 2017, 9: 453–9., DOI 10.2147/CMAR.S141338 Hao S, Zhao YY, Peng JJ, Ren F, Yang WT, Yu KD, et al. Invasive micropapillary carcinoma of the breast had no difference in prognosis compared with invasive ductal carcinoma: a propensity-matched analysis. Sci Rep, 2019, 9(1):286., DOI 10.1038/s41598-018-36362-8 Li G, Yang S, Yao J, Wang Z, Yao G, Liu M, et al. Invasive micropapillary carcinoma of the breast had poor clinical characteristics but showed no difference in prognosis compared with invasive ductal carcinoma. World J Surg Oncol, 2016, 14(1):207., DOI 10.1186/s12957-016-0960-z Ye F, Yu P, Li N, Yang A, Xie X, Tang H, et al. Prognosis of invasive micropapillary carcinoma compared with invasive ductal carcinoma in breast: a meta-analysis of PSM studies. Breast, 2020, 51:11–20., DOI 10.1016/j.breast.2020. 01.041 Kramer Z, Kenessey I, Gango A, Lendvai G, Kulka J, Tokes AM. Cell polarity and cell adhesion associated gene expression differences between invasive micropapillary and no special type breast carcinomas and their prognostic significance. Sci Rep, 2021, 11(1):18484., DOI 10.1038/s41598- 021-97347-8 Gruel N, Benhamo V, Bhalshankar J, Popova T, Freneaux P, Arnould L, et al. Polarity gene alterations in pure invasive micropapillary carcinomas of the breast. Breast Cancer Res, 2014, 16(3):R46., DOI 10.1186/bcr3653 Marchio C, Iravani M, Natrajan R, Lambros MB, Savage K, Tamber N, et al. Genomic and immunophenotypical characterization of pure micropapillary carcinomas of the breast. J Pathol, 2008, 215(4):398–410., DOI 10.1002/path.2368 Yu JI, Choi DH, Huh SJ, Cho EY, Kim K, Chie EK, et al. Differences in prognostic factors and failure patterns between invasive micropapillary carcinoma and carcinoma with micropapillary component versus invasive ductal carcinoma of the breast: retrospective multicenter case-control study, KROG 13-06). Clin Breast Cancer, 2015, 15(5):353–61.e612. e1-2., DOI 10.1016/j.clbc.2015.01.008 Shi Q, Shao K, Jia H, Cao B, Li W, Dong S, et al. Genomic alterations and evolution of cell clusters in metastatic invasive micropapillary carcinoma of the breast. Nat Commun, 2022, 13(1):111., DOI 10.1038/s41467-021-27794-4 Badyal RK, Bal A, Das A, Singh G. Invasive micropapillary carcinoma of the breast: immunophenotypic analysis and role of cell adhesion molecules, CD44 and E-cadherin, in nodal metastasis. Appl Immunohistochem Mol Morphol, 2016, 24(3):151–8., DOI 10.1097/PAI.0000000000000167 Furuse M, Fujita K, Hiiragi T, Fujimoto K, Tsukita S. claudin-1 and -2: novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cel Biol, 1998, 141(7):1539–50., DOI 10.1083/jcb.141.7.1539 Van Itallie CM, Lidman KF, Tietgens AJ, Anderson JM. Newly synthesized claudins but not occludin are added to the basal side of the tight junction. Mol Biol Cel, 2019, 30(12):1406–24., DOI 10.1091/mbc.E19-01-0008 Lal-Nag M, Morin PJ. The claudins. Genome Biol, 2009, 10(8):235., DOI 10. 1186/gb-2009-10-8-235 Otani T, Nguyen TP, Tokuda S, Sugihara K, Sugawara T, Furuse K, et al. Claudins and JAM-A coordinately regulate tight junction formation and epithelial polarity. J Cel Biol, 2019, 218(10):3372–96., DOI 10.1083/jcb.201812157 Singh AB, Sharma A, Dhawan P. Claudin family of proteins and cancer: an overview. J Oncol, 2010, 2010:541957., DOI 10.1155/2010/541957 Morohashi S, Kusumi T, Sato F, Odagiri H, Chiba H, Yoshihara S, et al. Decreased expression of claudin-1 correlates with recurrence status in breast cancer. Int J Mol Med, 2007, 20(2):139–43., DOI 10.3892/ijmm.20.2.139 Blanchard AA, Skliris GP, Watson PH, Murphy LC, Penner C, Tomes L, et al. Claudins 1, 3, and 4 protein expression in ER negative breast cancer correlates with markers of the basal phenotype. Virchows Arch, 2009, 454(6):647–56., DOI 10.1007/ s00428-009-0770-6 Lu S, Singh K, Mangray S, Tavares R, Noble L, Resnick MB, et al. Claudin expression in high-grade invasive ductal carcinoma of the breast: correlation with the molecular subtype. Mod Pathol, 2013, 26(4):485–95., DOI 10.1038/modpathol. 2012.187 Kulka J, Szasz AM, Nemeth Z, Madaras L, Schaff Z, Molnar IA, et al. Expression of tight junction protein claudin-4 in basal-like breast carcinomas. Pathol Oncol Res, 2009, 15(1):59–64., DOI 10.1007/s12253-008-9089-x Lanigan F, McKiernan E, Brennan DJ, Hegarty S, Millikan RC, McBryan J, et al. Increased claudin-4 expression is associated with poor prognosis and high tumour grade in breast cancer. Int J Cancer, 2009, 124(9):2088–97., DOI 10.1002/ijc. 24159 Kolokytha P, Yiannou P, Keramopoulos D, Kolokythas A, Nonni A, Patsouris E, et al. claudin-3 and claudin-4: distinct prognostic significance in triple-negative and luminal breast cancer. Appl Immunohistochem Mol Morphol, 2014, 22(2): 125–31., DOI 10.1097/PAI.0b013e31828d9d62 Dias K, Dvorkin-Gheva A, Hallett RM, Wu Y, Hassell J, Pond GR, et al. Claudin-low breast cancer; clinical and pathological characteristics. PLoS One, 2017, 12(1):e0168669., DOI 10.1371/journal.pone.0168669 Jaaskelainen A, Soini Y, Jukkola-Vuorinen A, Auvinen P, Haapasaari KM, Karihtala P. High-level cytoplasmic claudin 3 expression is an independent predictor of poor survival in triple-negative breast cancer. BMC Cancer, 2018, 18(1):223., DOI 10.1186/s12885-018-4141-z Kominsky SL, Argani P, Korz D, Evron E, Raman V, Garrett E, et al. Loss of the tight junction protein claudin-7 correlates with histological grade in both ductal carcinoma in situ and invasive ductal carcinoma of the breast. Oncogene, 2003, 22(13):2021–33., DOI 10.1038/sj.onc.1206199 Prat A, Parker JS, Karginova O, Fan C, Livasy C, Herschkowitz JI, et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res, 2010, 12(5):R68., DOI 10.1186/bcr2635 Sauer T, Pedersen MK, Ebeltoft K, Naess O. Reduced expression of claudin-7 in fine needle aspirates from breast carcinomas correlate with grading and metastatic disease. Cytopathology, 2005, 16(4):193–8., DOI 10.1111/j.1365-2303. 2005.00257.x Tokes AM, Kulka J, Paku S, Szik A, Paska C, Novak PK, et al. claudin-1, -3 and -4 proteins and mRNA expression in benign and malignant breast lesions: a research study. Breast Cancer Res, 2005, 7(2):R296–305., DOI 10.1186/bcr983 Lakhani IafRoC SR. WHO classification of breast tumours. 5th ed., 2019). Luna-More S, Gonzalez B, Acedo C, Rodrigo I, Luna C. Invasive micropapillary carcinoma of the breast. A new special type of invasive mammary carcinoma. Pathol Res Pract, 1994, 190(7):668–74., DOI 10.1016/ S0344-0338(11)80745-4 Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, et al. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen international expert Consensus on the primary therapy of early breast cancer 2013. Ann Oncol, 2013, 24(9):2206–23., DOI 10.1093/ annonc/mdt303 Nakazawa T, Hasegawa A, Nagasaka T, Yoshida K, Guo F, Wu D, et al. Differential expression of claudin 1 and 4 in basal cell carcinoma of the skin. Dermatol Res Pract, 2023, 2023:9936551., DOI 10.1155/2023/9936551 Naimi A, Zare N, Amjadi E, Soltan M. High claudin-4 antigen expression in triple-negative breast cancer by the immunohistochemistry method. J Res Med Sci, 2022, 27:20., DOI 10.4103/jrms.jrms_1389_20 Gyulai M, Harko T, Fabian K, Karsko L, Agocs L, Szigeti B, et al. Claudin expression in pulmonary adenoid cystic carcinoma and mucoepidermoid carcinoma. Pathol Oncol Res, 2023, 29:1611328., DOI 10.3389/pore.2023. 1611328 Dottermusch M, Kruger S, Behrens HM, Halske C, Rocken C. Expression of the potential therapeutic target claudin-18.2 is frequently decreased in gastric cancer: results from a large Caucasian cohort study. Virchows Arch, 2019, 475(5):563–71., DOI 10.1007/s00428-019-02624-7 Szade J, Kunc M, Pieczynska-Uzieblo B, Swierblewski M, Biernat W, Jassem J, et al. Comparison of claudin-3 and claudin-4 expression in bilateral and unilateral breast cancer. Neoplasma, 2021, 68(2):283–9., DOI 10.4149/neo_ 2020_200816N867 Murakami-Nishimagi Y, Sugimoto K, Kobayashi M, Tachibana K, Kojima M, Okano M, et al. Claudin-4-adhesion signaling drives breast cancer metabolism and progression via liver X receptor β. Breast Cancer Res, 2023, 25(1):41., DOI 10.1186/ s13058-023-01646-z Graudenz MS, Cadore E, Ferreira BP, Schulz R, Borba C, Uchoa D, et al. Claudins 1, 3, 4, 7 and 10-year survival in triple-negative breast tumors. Mastology, 2019, 29(2):71–8., DOI 10.29289/2594539420190000419 Li J. Dysregulated expression of claudins in cancer. Oncol Lett, 2021, 22(3): 641., DOI 10.3892/ol.2021.12902 Goehring NW. PAR polarity: from complexity to design principles. Exp Cel Res, 2014, 328(2):258–66., DOI 10.1016/j.yexcr.2014.08.009 Bonello TT, Peifer M. Scribble: a master scaffold in polarity, adhesion, synaptogenesis, and proliferation. J Cel Biol, 2019, 218(3):742–56., DOI 10.1083/ jcb.201810103 Tan B, YatimS, Peng S, Gunaratne J, Hunziker W, Ludwig A. Themammalian Crumbs complex defines a distinct polarity domain apical of epithelial tight junctions. Curr Biol, 2020, 30(14):2791–804.e6., DOI 10.1016/j.cub.2020.05.032 Onuma K, Inoue M. Abnormality of apico-basal polarity in adenocarcinoma. Cancer Sci, 2022, 113(11):3657–63., DOI 10.1111/cas.15549 Nehme Z, Roehlen N, Dhawan P, Baumert TF. Tight junction protein signaling and cancer biology. Cells, 2023, 12(2):243., DOI 10.3390/cells12020243 Ma X, Miao H, Jing B, Pan Q, Zhang H, Chen Y, et al. claudin-4 controls the proliferation, apoptosis, migration and in vivo growth of MCF-7 breast cancer cells. Oncol Rep, 2015, 34(2):681–90., DOI 10.3892/or.2015.4037 Park D, Karesen R, Axcrona U, Noren T, Sauer T. Expression pattern of adhesion molecules, E-cadherin, alpha-beta-gamma-catenin and claudin-7), their influence on survival in primary breast carcinoma, and their corresponding axillary lymph node metastasis. APMIS, 2007, 115(1):52–65., DOI 10.1111/j.1600-0463.2007. apm_524.x Fougner C, Bergholtz H, Norum JH, Sorlie T. Re-definition of claudin-low as a breast cancer phenotype. Nat Commun, 2020, 11(1):1787., DOI 10.1038/s41467-020- 15574-5 Pan C, Xu A, Ma X, Yao Y, Zhao Y, Wang C, et al. Research progress of claudin-low breast cancer. Front Oncol, 2023, 13:1226118., DOI 10.3389/fonc.2023. 1226118 Luo Y, Kishi S, Sasaki T, Ohmori H, Fujiwara-Tani R, Mori S, et al. Targeting claudin-4 enhances chemosensitivity in breast cancer. Cancer Sci, 2020, 111(5): 1840–50., DOI 10.1111/cas.14361 NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1611987 EP 1611998 DI 10.3389/pore.2024.1611987 PG 12 ER PT J AU Pathology and Oncology Research, EO AF Pathology and Oncology Research, Editorial Office TI Retraction: The Plasmodium circumsporozoite protein, a novel NF-κB inhibitor, suppresses the growth of SW480 SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Retraction AB A Retraction of the Original Research Article The Plasmodium circumsporozoite protein, a novel NF-κB inhibitor, suppresses the growth of SW480 by Ding Y, Huang X, Liu T, Fu Y, Tan Z, Zheng H, Zhou T, Dai J and Xu W (2012). Pathol. Oncol. Res. 19:11–18. doi: 10.1007/s12253-012-9519-7 C1 [Pathology and Oncology Research, Editorial Office] Frontiers Media SA, Switzerland. RP Pathology and Oncology Research, EO (reprint author), Frontiers Media SA, , Switzerland. NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1612018 EP 1612019 DI 10.3389/pore.2024.1612018 PG 2 ER PT J AU Editorial Office, PaOR AF Editorial Office, Pathology and Oncology Research TI Retraction: Effects of gallotannin-enriched extract of Galla Rhois on the activation of apoptosis, cell cycle arrest, and inhibition of migration ability in LLC1 cells and LLC1 tumors SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Retraction AB A Retraction of the Original Research Article Effects of gallotannin-enriched extract of Galla Rhois on the activation of apoptosis, cell cycle arrest, and inhibition of migration ability in LLC1 cells and LLC1 tumors C1 [Editorial Office, Pathology and Oncology Research] Frontiers Media SALausanne, Switzerland. RP Editorial Office, PaOR (reprint author), Frontiers Media SA, Lausanne, Switzerland. EM editorialoffice@por-journal.com NR 4 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD OCT PY 2024 VL 30 IS 4 BP 1612042 EP 1612043 DI 10.3389/pore.2024.1612042 PG 2 ER PT J AU Jia, G Li, X AF Jia, Guohua Li, Xiangpan TI Survival trends of gastrointestinal stromal tumor in real-world settings: a population-based retrospective study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE GIST; survival; tumor-specific mortality; trend; outcome ID GIST; survival; tumor-specific mortality; trend; outcome AB Purpose: This study aims to evaluate whether survival outcomes for GIST patients have improved over the past decades and to identify the specific patient subgroups that have benefited from advances in treatment. Patients and methods: A total of 4,127 GIST patients diagnosed between January 1980, and December 2019, were included in this study using data from the Surveillance, Epidemiology, and End Results (SEER)-9 Registries. Survival differences among GIST patients were analyzed across five time periods (1980–1999, 2000–2004, 2005–2009, 2010–2014, and 2015–2019) and within demographic, neoplastic, temporal, economic, and geographic categories using the log-rank test. Multivariable Cox regression models were employed to identify risk factors associated with GIST-specific survival. Associations between time periods and GIST-specific mortality (TSM) were examined using a multivariable Cox regression model. Results: Survival outcomes for GIST patients significantly improved in the 2000–2009 period but showed no substantial improvement in the 2010–2019 period. After adjusting for age, gender, tumor location, ethnicity, tumor stage, median household income, and geographic area, the multivariable Cox regression models revealed that older age (≥65 years) (HR = 1.977, 95% CI = 1.470–2.657), tumors located outside the gastrointestinal tract (HR = 1.505, 95% CI = 1.267–1.786), regional lesions (HR = 2.225, 95% CI = 1.828–2.708), and distant lesions (HR = 5.177, 95% CI = 4.417–6.069) were independent risk factors for TSM (p < 0.05). After adjusting for time periods and age, gender, tumor location, tumor stage, median household income, patients in 2000–2004 (HR = 0.662, 95% CI = 0.523–0.839), 2005–2009 (HR = 0.431, 95% CI = 0.339–0.549), 2010–2014 (HR = 0.437, 95% CI = 0.341–0.561), and 2015–2019 (HR = 0.365, 95% CI = 0.273–0.489) had a significantly lower risk of TSM than patients in 1980–1999 (p < 0.05). Similarly, patients in 2005–2009 (HR = 0.661, 95% CI = 0.555–0.788), 2010–2014 (HR = 0.696, 95% CI = 0.578–0.838), and 2015–2019 (HR = 0.607, 95% CI = 0.476–0.773) also had a significantly lower risk of TSM than patients in 2000–2004 (p < 0.05). However, patients in 2010–2014 (HR = 1.042, 5% CI = 0.863–1.258) and 2015–2019 (HR = 0.945, 95% CI = 0.734–1.216) did not have a significantly lower risk of TSM compared to patients in 2005–2009 (p > 0.05). Conclusion: GIST survival has significantly improved during the period 2000–2009 but showed no substantial improvement in 2010–2019, with the turning point for lower risk of TSM being 2005. Innovative strategies are needed to further improve survival outcomes for GIST patients, particularly for older patients and those with tumors originating outside the gastrointestinal tract. C1 [Jia, Guohua] Wuhan University, Renmin Hospital, Cancer CenterWuhan, Hubei, China. [Li, Xiangpan] Wuhan University, Renmin Hospital, Department of OncologyWuhan, Hubei, China. RP Li, X (reprint author), Wuhan University, Renmin Hospital, Department of Oncology, Wuhan, China. EM rm001227@whu.edu.cn CR Blay JY, Kang YK, Nishida T, von Mehren M. Gastrointestinal stromal tumours. Nat Rev Dis Primers, 2021, 7(1):22., DOI 10.1038/s41572-021-00254-5 Wada R, Arai H, Kure S, PengWX, Naito Z. Wild type GIST: clinicopathological features and clinical practice. Pathol Int, 2016, 66(8):431–7., DOI 10.1111/pin.12431 Kelly CM, Gutierrez Sainz L, Chi P. The management of metastatic GIST: current standard and investigational therapeutics. J Hematol Oncol, 2021, 14(1):2., DOI 10.1186/s13045-020-01026-6 Lu T, Yang X, Huang Y, Zhao M, Li M, Ma K, et al. Trends in the incidence, treatment, and survival of patients with lung cancer in the last four decades. Cancer Manag Res, 2019, 11:943–53., DOI 10.2147/CMAR.S187317 Han J, Wang B, Liu W, Wang S, Chen R, Chen M, et al. Declining disease burden of HCC in the United States, 1992-2017: a population-based analysis. Hepatology, 2022, 76(3):576–88., DOI 10.1002/hep.32355 Everatt R, Gudaviciene D. An analysis of time trends in breast and prostate cancer mortality rates in Lithuania, 1986-2020. BMC Public Health, 2022, 22(1): 1812., DOI 10.1186/s12889-022-14207-4 Gold JS, GonenM, Gutierrez A, Broto JM, Garcia-del-Muro X, Smyrk TC, et al. Development and validation of a prognostic nomogram for recurrence-free survival after complete surgical resection of localised primary gastrointestinal stromal tumour: a retrospective analysis. Lancet Oncol, 2009, 10(11):1045–52., DOI 10. 1016/S1470-2045(09)70242-6 Guller U, Tarantino I, Cerny T, Schmied BM, Warschkow R. Population-based SEER trend analysis of overall and cancer-specific survival in 5138 patients with gastrointestinal stromal tumor. BMC Cancer, 2015, 15:557., DOI 10.1186/s12885- 015-1554-9 Khan J, Ullah A, Waheed A, Karki N, Adhikari N, Vemavarapu L, et al. Gastrointestinal stromal tumors, GIST): a population-based study using the SEER database, including management and recent advances in targeted therapy. Cancers, Basel,, 2022, 14(15):3689., DOI 10.3390/cancers14153689 Patel N, Benipal B. Incidence of gastrointestinal stromal tumors in the United States from 2001-2015: a United States cancer statistics analysis of 50 states. Cureus, 2019, 11(2):e4120., DOI 10.7759/cureus.4120 Mantese G. Gastrointestinal stromal tumor: epidemiology, diagnosis, and treatment. Curr Opin Gastroenterol, 2019, 35(6):555–9., DOI 10.1097/MOG. 0000000000000584 Cho MY, Sohn JH, Kim JM, Kim KM, Park YS, Kim WH, et al. Current trends in the epidemiological and pathological characteristics of gastrointestinal stromal tumors in Korea, 2003-2004. J Korean Med Sci, 2010, 25(6):853–62., DOI 10.3346/ jkms.2010.25.6.853 Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Niksic M, et al. Global surveillance of trends in cancer survival 2000-14, CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet, 2018, 391(10125):1023–75., DOI 10.1016/S0140-6736(17)33326-3 Allemani C,Weir HK, Carreira H, Harewood R, Spika D, Wang X, et al. Global surveillance of cancer survival 1995-2009: analysis of individual data for 25,676,887 patients from 279 population-based registries in 67 countries, CONCORD- 2). Lancet, 2015, 385(9972):977–1010., DOI 10.1016/S0140-6736(14)62038-9 Coleman MP, QuaresmaM, Berrino F, Lutz J, Angelis RD, Capocaccia R, et al. Cancer survival in five continents: a worldwide population-based study, CONCORD). Lancet Oncol, 2008, 9(8):730–56., DOI 10.1016/S1470-2045(08, 70179-7 Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Ishiguro S, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science, 1998, 279(5350):577–80., DOI 10.1126/science.279.5350.577 Mohammadi M, Gelderblom H. Systemic therapy of advanced/metastatic gastrointestinal stromal tumors: an update on progress beyond imatinib, sunitinib, and regorafenib. Expert Opin Investig Drugs, 2021, 30(2):143–52., DOI 10.1080/ 13543784.2021.1857363 Wang MX, Devine C, Segaran N, Ganeshan D. Current update on molecular cytogenetics, diagnosis and management of gastrointestinal stromal tumors. World J Gastroenterol, 2021, 27(41):7125–33., DOI 10.3748/wjg.v27.i41.7125 Klug LR, Khosroyani HM, Kent JD, Heinrich MC. New treatment strategies for advanced-stage gastrointestinal stromal tumours. Nat Rev Clin Oncol, 2022, 19(5):328–41., DOI 10.1038/s41571-022-00606-4 Martin-Broto J, Moura DS. New drugs in gastrointestinal stromal tumors. Curr Opin Oncol, 2020, 32(4):314–20., DOI 10.1097/CCO.0000000000000642 Serrano C. New treatments in advanced gastrointestinal stromal tumor. Curr Opin Oncol, 2021, 33(4):323–8., DOI 10.1097/CCO.0000000000000745 Blay JY, Serrano C, Heinrich MC, Zalcberg J, Bauer S, Gelderblom H, et al. Ripretinib in patients with advanced gastrointestinal stromal tumours, INVICTUS): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol, 2020, 21(7):923–34., DOI 10.1016/S1470-2045(20)30168-6 Fernandez JA, Alconchel F, Gomez B, Martinez J, Ramirez P. Unresectable GIST liver metastases and liver transplantation: a review and theoretical basis for a new indication. Int J Surg, 2021, 94:106126., DOI 10.1016/j.ijsu.2021.106126 Keung EZ, Fairweather M, Raut CP. The role of surgery in metastatic gastrointestinal stromal tumors. Curr Treat Options Oncol, 2016, 17(2):8., DOI 10.1007/s11864-015-0384-y Yonkus JA, Alva-Ruiz R, Grotz TE. Surgical management of metastatic gastrointestinal stromal tumors. Curr Treat Options Oncol, 2021, 22(5):37., DOI 10.1007/s11864-021-00837-0 Fairweather M, Balachandran VP, Li GZ, Bertagnolli MM, Antonescu C, Tap W, et al. Cytoreductive surgery for metastatic gastrointestinal stromal tumors treated with tyrosine kinase inhibitors: a 2-institutional analysis. Ann Surg, 2018, 268(2):296–302., DOI 10.1097/SLA.0000000000002281 Fairweather M, Cavnar MJ, Li GZ, Bertagnolli MM, DeMatteo RP, Raut CP. Prediction of morbidity following cytoreductive surgery for metastatic gastrointestinal stromal tumour in patients on tyrosine kinase inhibitor therapy. Br J Surg, 2018, 105(6):743–50., DOI 10.1002/bjs.10774 Fero KE, Coe TM, Fanta PT, Tang CM, Murphy JD, Sicklick JK. Surgical management of adolescents and young adults with gastrointestinal stromal tumors: a US population-based analysis. JAMA Surg, 2017, 152(5):443–51., DOI 10.1001/ jamasurg.2016.5047 Miettinen M, Lasota J. Gastrointestinal stromal tumors. Gastroenterol Clin North Am, 2013, 42(2):399–415., DOI 10.1016/j.gtc.2013.01.001 Liu L, Feng Y, Ye Y, Wang Z, Xu X. Survival analysis of extragastrointestinal stromal tumors based on the SEER database: a population-based study. Surg Endosc, 2023, 37(11):8498–510., DOI 10.1007/s00464-023-10433-y Joensuu H, Vehtari A, Riihimaki J, Nishida T, Steigen SE, Brabec P, et al. Risk of recurrence of gastrointestinal stromal tumour after surgery: an analysis of pooled population-based cohorts. Lancet Oncol, 2012, 13(3):265–74., DOI 10.1016/S1470- 2045(11)70299-6 Zhang H, Liu Q. Prognostic indicators for gastrointestinal stromal tumors: a review. Transl Oncol, 2020, 13(10):100812., DOI 10.1016/j.tranon.2020.100812 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1611896 EP 1611906 DI 10.3389/pore.2025.1611896 PG 11 ER PT J AU Fellows, D Kotowska, J Stevenson, Th Brown, J Orosz, Zs Siddiqi, A Whitwell, D Cosker, Th GIbbons, Ch AF Fellows, David Kotowska, Julia Stevenson, Thomas Brown, Jennifer Orosz, Zsolt Siddiqi, Ather Whitwell, Duncan Cosker, Thomas GIbbons, L. M. H. Christopher TI Management and surveillance of metastatic giant cell tumour of bone SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE giant cell tumor of bone; pulmonary metastasis; sarcoma; orthopaedic oncology; surveillance ID giant cell tumor of bone; pulmonary metastasis; sarcoma; orthopaedic oncology; surveillance AB Giant cell tumour of bone (GCTB) is viewed as a benign, locally aggressive primary bone tumour with metastatic potential. Current management is surgery with bone curettage or resection and systemic therapy with denosumab. Diagnosis is confirmed histologically prior to surgery, with staging for pulmonary disease, as pulmonary metastases (PM) reportedly occur in <8%. This study aimed to assess incidence, surveillance and management of PM in patients with GCTB, with histopathological review. A retrospective audit of the Oxford bone tumour registry was performed from January 2014 – October 2023. Inclusion criterion was histological confirmation of GCTB. Exclusion criteria were incomplete medical, imaging or histology records, or referral for secondary MDT opinion for diagnosis. From an initial group of 126 GCTB patients, 83 patients met the full selection criteria. Pulmonary metastases were identified in 11 patients. Three with PM were excluded on histopathological review as being giant cell rich osteosarcoma rather than metastatic GCTB. This left 8 (9.6%) patients, one had PM at presentation and seven at follow-up between 2 and 42 months. Two were histologically confirmed after cardiothoracic surgery and biopsy, six radiologically diagnosed. Three (37.5%) patients with PM have died (between 1 and 12 months after confirmed PM), five are alive with stable disease. Seven (87.5%) of patients with pulmonary disease were treated with denosumab/chemotherapy (three before, four after pulmonary diagnosis). Five (62.5%) with pulmonary disease had recurrence of local disease requiring further surgery. Local recurrence was an independent risk factor for PM on statistical analysis. GCTB may present with PM, but more commonly, metastasis occurs after surgery, presenting on surveillance and can progress. There were no distinct differences in histopathological appearance between patients with GCTB that developed PM and those that did not, therefore morphological features of the tumour cannot be currently used to predict tumour behaviour. PM can behave aggressively, necessitating identifying histological markers to recognise patients at risk of metastatic GCTB, for example, through mRNA single cell analysis. We propose GCTB patients with PM receive regular chest surveillance with PET scan and/or CT to monitor disease progression, and a multi-centre audit of GCTB outcome undertaken to further define optimal clinical management. C1 [Fellows, David] Nuffield Orthopaedic Centre, Oxford Sarcoma ServiceOxford, England, UK. [Kotowska, Julia] Hampshire Hospitals NHS Foundation Trust, Trauma and OrthopaedicsBasingstoke, UK. [Stevenson, Thomas] Institute of Naval Medicine, Trauma and OrthopaedicsAlverstoke, UK. [Brown, Jennifer] Nuffield Orthopaedic Centre, Oxford Sarcoma ServiceOxford, England, UK. [Orosz, Zsolt] Nuffield Orthopaedic Centre, Oxford Sarcoma ServiceOxford, England, UK. [Siddiqi, Ather] Nuffield Orthopaedic Centre, Oxford Sarcoma ServiceOxford, England, UK. [Whitwell, Duncan] Nuffield Orthopaedic Centre, Oxford Sarcoma ServiceOxford, England, UK. [Cosker, Thomas] Nuffield Orthopaedic Centre, Oxford Sarcoma ServiceOxford, England, UK. [GIbbons, L. M. H. Christopher] Nuffield Orthopaedic Centre, Oxford Sarcoma ServiceOxford, England, UK. RP Fellows, D (reprint author), Nuffield Orthopaedic Centre, Oxford Sarcoma Service, Oxford, UK. EM david.fellows2@nhs.net CR Siegal GPHM, Bloem JL, Cates JMM. WHO classification of tumors 5th edition soft tissue and bone tumors. 5th ed. Geneva, Switzerland: World Health Organization, 2020). 440–7. Turcotte RE. Giant cell tumor of bone. Orthop Clin North Am, 2006, 37(1): 35–51., DOI 10.1016/j.ocl.2005.08.005 Unni KK, Inwards CY. Dahlin’s bone tumors: general aspects and data on 10,165 cases. 6th ed. Philadelphia, United States: Lippincott Williams and Wilkins, 2012). Yang Y, Huang Z, Niu X, Xu H, Li Y, Liu W. Clinical characteristics and risk factors analysis of lung metastasis from benign giant cell tumor of bone. J Bone Oncol, 2017)(7, 23–8., DOI 10.1016/j.jbo.2017.04.001 Barik S, Jain A, Ahmad S, Singh V. Functional outcome in giant cell tumor of distal radius treated with excision and fibular arthroplasty: a case series. Eur J Orthop Surg Traumatol, 2020, 30(6):1109–17., DOI 10.1007/s00590-020-02679-2 Sarcoma. Giant cell tumour of the bone. Available from: https://sarcoma.org. uk/about-sarcoma/what-is-sarcoma/types-of-sarcoma/giant-cell-tumour-of-thebone/, Accessed November 30, 2023). Hosseinzadeh S, De Jesus O. Giant cell tumor. StatPearls. Treasure Island, FL): StatPearls Publishing, 2023). Available from: https://www.ncbi.nlm.nih.gov/books/ NBK559229/, Accessed January 8, 2024). Kundu ZS, Sen R, Dhiman A, Sharma P, Siwach R, Rana P. Effect of intravenous zoledronic acid on histopathology and recurrence after extended curettage in giant cell tumors of bone: a comparative prospective study. Indian J Orthop, 2018, 52(1):45–50., DOI 10.4103/ortho.IJOrtho_216_17 Akiyama T, Yoshimatsu Y, Noguchi R, Sin Y, Tsuchiya R, Ono T, et al. Establishment and characterization of NCC-GCTB5-C1: a novel cell line of giant cell tumor of bone. Hum Cel, 2022, 35(5):1621–9., DOI 10.1007/s13577-022-00724-2 Brodowicz T, Hemetsberger M, Windhager R. Denosumab for the treatment of giant cell tumour of the bone. Future Oncol, 2015, 11(13):1881–94., DOI 10.2217/ fon.15.94 Imre A, Zoltan S, Miklos S. Current indications for denosumab in benign bone tumours. EFFORT Open Rev, 2023, 8(12):895–905., DOI 10.1530/EOR- 23-0138 van der Heijden L, Dijkstra PDS, van de Sande MAJ, Kroep JR, Nout RA, van Rijswijk CSP, et al. The clinical approach toward giant cell tumor of bone. The Oncologist, 2014, 19(5):550–61., DOI 10.1634/theoncologist.2013-0432 Caudell JJ, Ballo MT, Zagars GK, Lewis VO, Weber KL, Lin PP, et al. Radiotherapy in themanagement of giant cell tumor of bone. Int J Radiat Oncol Biol Phys, 2003, 57(1):158–65., DOI 10.1016/s0360-3016(03)00416-4 Errani C, Tsukamoto S, Ciani G, Donati DM. Present day controversies and consensus in curettage for giant cell tumor of bone. J Clin Orthop Trauma, 2019, 10(6):1015–20., DOI 10.1016/j.jcot.2019.09.017 Balke M, Schremper L, Gebert C, Ahrens H, Streitbuerger A, Koehler G, et al. Giant cell tumor of bone: treatment and outcome of 214 cases. J Cancer Res Clin Oncol, 2008, 134(9):969–78., DOI 10.1007/s00432-008-0370-x Arbeitsgemeinschaft K, Becker WT, Dohle J, Bernd L, Braun A, Cserhati M, et al. Local recurrence of giant cell tumor of bone after intralesional treatment with and without adjuvant therapy. J Bone Joint Surg Am, 2008, 90(5):1060–7., DOI 10. 2106/JBJS.D.02771 van der Heijden L, Dijkstra PDS, Blay JY, Gelderblom H. Giant cell tumour of bone in the denosumab era. Eur J Cancer, 2017, 77:75–83., DOI 10.1016/j.ejca.2017. 02.021 Atkins GJ, Haynes DR, Graves SE, Evdokiou A, Hay S, Bouralexis S, et al. Expression of osteoclast differentiation signals by stromal elements of giant cell tumors. J Bone Miner Res Off J Am Soc Bone Miner Res, 2000, 15(4):640–9., DOI 10. 1359/jbmr.2000.15.4.640 Wulling M, Delling G, Kaiser E. The origin of the neoplastic stromal cell in giant cell tumor of bone. Hum Pathol, 2003, 34(10):983–93., DOI 10.1053/s0046- 8177(03)00413-1 Behjati S, Tarpey PS, Presneau N, Scheipl S, Pillay N, Van Loo P, et al. Distinct H3F3A and H3F3B driver mutations define chondroblastoma and giant cell tumor of bone. Nat Genet, 2013, 45(12):1479–82., DOI 10.1038/ng.2814 Yamamoto H, Ishihara S, Toda Y, Oda Y. Histone H3.3 mutation in giant cell tumor of bone: an update in pathology. Med Mol Morphol, 2020, 53(1):1–6., DOI 10. 1007/s00795-019-00238-1 Lutsik P, Baude A,Mancarella D, Oz S, Kuhn A, Toth R, et al. Globally altered epigenetic landscape and delayed osteogenic differentiation in H3.3-G34W-mutant giant cell tumor of bone. Nat Commun, 2020, 11(1):5414., DOI 10.1038/s41467-020- 18955-y Chawla S, Blay JY, Rutkowski P, Le Cesne A, Reichardt P, Gelderblom H, et al. Denosumab in patients with giant-cell tumour of bone: a multicentre, open-label, phase 2 study. Lancet Oncol, 2019, 20(12):1719–29., DOI 10.1016/S1470-2045(19, 30663-1 Zoccali C, Formica VM, Sperduti I, Checcucci E, Scotto di Uccio A, Pagnotta A, et al.Wide resection for giant-cell tumor of the distal radius: which reconstruction? A systematic review of the literature and pooled analysis of 176 cases. Hand Surg Rehabil, 2022, 41(5):552–60., DOI 10.1016/j.hansur.2022.07.002 Kremen TJ, Jr, Bernthal NM, Eckardt MA, Eckardt JJ. Giant cell tumor of bone: are we stratifying results appropriately? Clin Orthop Relat Res, 2012, 470(3): 677–83., DOI 10.1007/s11999-011-2172-8 Xing R, Yang J, Kong Q, Tu C, Zhou Y, Duan H. Giant cell tumour of bone in the appendicular skeleton: an analysis of 276 cases. Acta Orthop Belg, 2013, 79(6): 731–7., DOI 10.1302/2058-5241.6.200154 Saikia KC, Bhuyan SK, Borgohain M, Saikia SP, Bora A, Ahmed F. Giant cell tumour of bone: an analysis of 139 Indian patients. J Orthop Sci, 2011, 16(5):581–8., DOI 10.1007/s00776-011-0033-7 Luengo-Alonso G, Mellado-Romero M, Shemesh S, Ramos-Pascua L, Pretell- Mazzini J. Denosumab treatment for giant-cell tumor of bone: a systematic review of the literature. Arch Orthop Trauma Surg, 2019, 139(10):1339–49., DOI 10.1007/ s00402-019-03167-x Gaston CL, Bhumbra R, Watanuki M, Abudu AT, Carter SR, Jeys LM, et al. Does the addition of cement improve the rate of local recurrence after curettage of giant cell tumours in bone? J Bone Joint Surg Br, 2011, 93(12):1665–9., DOI 10.1302/ 1-620X.93B12.27663 30. Chanchairujira K, Jiranantanakorn T, Phimolsarnti R, Asavamongkolkul A, Waikakul S. Factors of local recurrence of giant cell tumor of long bone after treatment: plain radiographs, pathology and surgical procedures. J Med Assoc Thai, 2011, 94(10):1230–7. Aoude A, Nikomarov D, Perera JR, Ibe IK, Griffin AM, Tsoi KM, et al. Giant cell tumour of bone. Bone Joint J, 2023, 105-B(5):559–67., DOI 10.1302/0301-620X. 105B5.BJJ-2022-1231.R1 Errani C, Tsukamoto S, Leone G, Akahane M, Cevolani L, Tanzi P, et al. Higher local recurrence rates after intralesional surgery for giant cell tumor of the proximal femur compared to other sites. Eur J Orthop Surg Traumatol, 2017, 27(6): 813–9., DOI 10.1007/s00590-017-1983-z Turcotte RE, Wunder JS, Isler MH, Bell RS, Schachar N, Masri BA, et al. Giant cell tumor of long bone: a Canadian Sarcoma Group study. Clin Orthop Relat Res, 2002, 397:248–58., DOI 10.1097/00003086-200204000-00029 Becker RG, Galia CR, Pestilho JFCS, Antunes BP, Baptista AM, Guedes A. Giant cell tumor of bone: a multicenter epidemiological study in Brazil. Acta Ortop Bras, 2024, 32(1):e273066., DOI 10.1590/1413- 785220243201e273066 Abuhejleh H, Wunder JS, Ferguson PC, Isler MH, Mottard S, Werier JA, et al. Extended intralesional curettage preferred over resection-arthrodesis for giant cell tumour of the distal radius. Eur J Orthop Surg Traumatol Orthop Traumatol, 2020, 30(1):11–7., DOI 10.1007/s00590-019-02496-2 Kito M, Matusmoto S, Ae K, Tanizawa T, Gokita T, Kobayashi H, et al. Pulmonary metastasis from giant cell tumor of bone: clinical outcome prior to the introduction of molecular target therapy. Jpn J Clin Oncol, 2017, 47(6):529–34., DOI 10.1093/jjco/hyx033 Jiang N, Qin CH, Tan CX, Wen SF, Ma YF, Dong F, et al. A retrospective analysis of 140 patients with giant cell tumor in the extremity: a multicenter study based on four hospitals in South China. Cancer Epidemiol, 2013, 37(3):294–9., DOI 10.1016/j.canep.2013.01.009 Al-Ibraheemi A, Inwards CY, Zreik RT, Wenger DE, Jenkins SM, Carter JM, et al. Histologic spectrum of giant cell tumor, GCT, of bone in patients 18 Years of age and below: a study of 63 patients. Am J Surg Pathol, 2016, 40(12):1702–12., DOI 10.1097/PAS.0000000000000715 Machak GN, Snetkov AI. The impact of curettage technique on local control in giant cell tumour of bone. Int Orthop, 2021, 45(3):779–89., DOI 10.1007/s00264- 020-04860-y Niu X, Zhang Q, Hao L, Ding Y, Li Y, Xu H, et al. Giant cell tumor of the extremity: retrospective analysis of 621 Chinese patients from one institution. JBJS, 2012, 94(5):461–7., DOI 10.2106/JBJS.J.01922 Vari S, Riva F, Onesti CE, Cosimati A, Renna D, Biagini R, et al. Malignant transformation of giant cell tumour of bone: a review of literature and the experience of a referral centre. Int J Mol Sci, 2022, 23(18):10721., DOI 10.3390/ ijms231810721 Leland CR, Pratilas CA, Gross JM, Levin AS. Diffuse pulmonary metastases at presentation of giant cell tumor of bone: a case report and synthesis of literature. JBJS Case Connect, 2023, 13(1)., DOI 10.2106/JBJS.CC. 22.00496 Tsukamoto S, Mavrogenis AF, Tanaka Y, Kido A, Honoki K, Tanaka Y, et al. Metastasectomy versus non-metastasectomy for giant cell tumor of bone lung metastases. Orthopedics, 2021, 44(6):e707–e712., DOI 10.3928/01477447- 20211001-01 Dominkus M, Ruggieri P, Bertoni F, Briccoli A, Picci P, Rocca M, et al. Histologically verified lung metastases in benign giant cell tumours--14 cases from a single institution. Int Orthop, 2006, 30(6):499–504., DOI 10.1007/s00264- 006-0204-x Lans J, Oflazoglu K, Lee H, Harness NG, Castelein RM, Chen NC, et al. Giant cell tumors of the upper extremity: predictors of recurrence. J Hand Surg, 2020, 45(8):738–45., DOI 10.1016/j.jhsa.2020.04.020 Viswanathan S, Jambhekar NA. Metastatic giant cell tumor of bone: are there associated factors and best treatment modalities? Clin Orthop Relat Res, 2010, 468(3):827–33., DOI 10.1007/s11999-009-0966-8 Kamal AF, Simbolon EL, Prabowo Y, Hutagalung EU. Wide resection versus curettage with adjuvant therapy for giant cell tumour of bone. J Orthop Surg Hong Kong, 2016, 24(2):228–31., DOI 10.1177/1602400221 Tsukamoto S, Mavrogenis AF, Leone G, Righi A, Akahane M, Tanzi P, et al. Denosumab does not decrease the risk of lung metastases from bone giant cell tumour. Int Orthop, 2019, 43(2):483–9., DOI 10.1007/s00264-018-4085-6 Wang J, Liu X, Yang Y, Yang R, Tang X, Yan T, et al. Pulmonary metastasis of giant cell tumour: a retrospective study of three hundred and ten cases. Int Orthop, 2021, 45(3):769–78., DOI 10.1007/s00264-020-04907-0 Yayan J. Increased risk of lung metastases in patients with giant cell bone tumors: a systematic review. Adv Exp Med Biol, 2019, 1176:1–17., DOI 10.1007/ 5584_2019_372 Chan CM, Adler Z, Reith JD, Gibbs CP, Jr. Risk factors for pulmonary metastases from giant cell tumor of bone. J Bone Joint Surg Am, 2015, 97(5):420–8., DOI 10.2106/JBJS.N.00678 Rosario M, KimHS, Yun JY, Han I. Surveillance for lung metastasis from giant cell tumor of bone. J Surg Oncol, 2017, 116(7):907–13., DOI 10.1002/jso.24739 Fittall MW, Lyskjaer I, Ellery P, Lombard P, Ijaz J, Strobl AC, et al. Drivers underpinning the malignant transformation of giant cell tumour of bone. J Pathol, 2020, 252(4):433–40., DOI 10.1002/path.5537 Tsukamoto S, Righi A, Vanel D, Honoki K, Donati DM, Errani C. Development of high-grade osteosarcoma in a patient with recurrent giant cell tumor of the ischium while receiving treatment with denosumab. Jpn J Clin Oncol, 2017, 47(11):1090–6., DOI 10.1093/jjco/hyx112 Park A, Cipriano CA, Hill K, Kyriakos M, McDonald DJ. Malignant transformation of a giant cell tumor of bone treated with denosumab: a case report. Jbjs Case Connect, 2016, 6(3):e78., DOI 10.2106/JBJS.CC.16.00024 Bertoni F, Bacchini P, Staals EL. Malignancy in giant cell tumor of bone. Cancer, 2003, 97(10):2520–9., DOI 10.1002/cncr.11359 Liu W, Chan CM, Gong L, Bui MM, Han G, Letson GD, et al. Malignancy in giant cell tumor of bone in the extremities. J Bone Oncol, 2021, 26:100334., DOI 10. 1016/j.jbo.2020.100334 Chakarun CJ, Forrester DM, Gottsegen CJ, Patel DB, White EA, Matcuk GR, Jr. Giant cell tumor of bone: review, mimics, and new developments in treatment. Radiographics, 2013, 33(1):197–211., DOI 10.1148/rg.331125089 Yeo CD, Roh SY, Shin OR, Bahk WJ, Kim KH, Kim JW. A case of pulmonary metastasis of giant cell tumor of bone presenting as pulmonary arteriovenous malformation. J Formos Med Assoc, 2015, 114(4):369–72., DOI 10.1016/j.jfma.2012. 03.014 Gong L, Liu W, Sun X, Sajdik C, Tian X, Niu X, et al. Histological and clinical characteristics of malignant giant cell tumor of bone. Virchows Arch, 2012, 460(3): 327–34., DOI 10.1007/s00428-012-1198-y Muheremu A, Niu X. Pulmonary metastasis of giant cell tumor of bones. World J Surg Oncol, 2014, 12:261., DOI 10.1186/1477-7819-12-261 Xu R, Choong PFM. Metastatic giant cell tumour of bone: a narrative review of management options and approaches. ANZ J Surg, 2022, 92(4):691–6., DOI 10.1111/ ans.17520 van Langevelde K, McCarthy CL. Radiological findings of denosumab treatment for giant cell tumours of bone. Skeletal Radiol, 2020, 49(9):1345–58., DOI 10.1007/s00256-020-03449-1 Lawless ME, Jour G, Hoch BL, Rendi MH. Beta-human chorionic gonadotropin expression in recurrent and metastatic giant cell tumors of bone: a potential mimicker of germ cell tumor. Int J Surg Pathol, 2014, 22(7):617–22., DOI 10.1177/1066896914534466 Yoshida K, Nakano Y, Honda-Kitahara M, Wake S, Motoi OK, et al. Absence of H3F3A mutation in a subset of malignant giant cell tumour of bone. Mod Pathol, 2019, 32(12):1751–61., DOI 10.1038/s41379-019-0318-5 Amary F, Berisha F, Ye H, Gupta M, Gutteridge A, Baumhoer D, et al. H3F3A, histone 3.3, G34W immunohistochemistry: a reliable marker defining benign and malignant giant cell tumor of bone. Am J Surg Pathol, 2017, 41(8):1059–68., DOI 10. 1097/PAS.0000000000000859 Antal I, Sapi Z, Szendroi M. The prognostic significance of DNA cytophotometry and proliferation index, Ki-67, in giant cell tumors of bone. Int Orthop, 1999, 23(6):315–9., DOI 10.1007/s002640050381 Rousseau MA, Handra-Luca A, Lazennec JY, Catonne Y, Saillant G. Metachronous multicentric giant-cell tumor of the bone in the lower limb. Case report and Ki-67 immunohistochemistry study. Virchows Arch, 2004, 445(1): 79–82., DOI 10.1007/s00428-004-1011-7 Alberghini M, Kliskey K, Krenacs T, Picci P, Kindblom L, Forsyth R, et al. Morphological and immunophenotypic features of primary and metastatic giant cell tumour of bone. Virchows Arch, 2010, 456:97–103., DOI 10.1007/s00428-009-0863-2 Ismail FW, Shamsudin AM, Wan Z, Daud SM, Samarendra MS. Ki-67 immuno-histochemistry index in stage III giant cell tumor of the bone. J Exp Clin Cancer Res CR, 2010, 29(1):25., DOI 10.1186/1756-9966- 29-25 Parmeggiani A, Micelli M, Errani C, Facchini G. State of the art and new concepts in giant cell tumour of bone: imaging features and tumour characteristics. Cancers, 2021, 13(24):6298., DOI 10.3390/cancers13246298 Chen S, Du Z, Wu B, Shen H, Liu C, Qiu X, et al. STAT1, IGF1, RAC1, and MDM2 are associated with recurrence of giant cell tumor of bone. J Immunol Res, 2018, 2018:4564328., DOI 10.1155/2018/4564328 Metovic J, Anaratone L, Linari A, Osella-Abate S,Musuraca C, Veneziano F, et al. Prognostic role of PD-L1 and immune-related gene expression profiles in giant cell tumours of bone. Cancer Immunol Immunother, 2020, 69(9): 1905–16., DOI 10.1007/s00262-020-02594-9 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1611916 EP 1611926 DI 10.3389/pore.2025.1611916 PG 11 ER PT J AU Budi, L Hammer, D Varga, R Muller, V Tarnoki, DA Tarnoki, LD Meszaros, M Bikov, A Horvath, P AF Budi, Lilla Hammer, Daniel Varga, Rita Muller, Veronika Tarnoki, Domonkos Adam Tarnoki, Laszlo David Meszaros, Martina Bikov, Andras Horvath, Peter TI Anti-ceramide antibody and sphingosine-1-phosphate as potential biomarkers of unresectable non-small cell lung cancer SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE S1P; anti-ceramide antibody; NSCLC; biomarkers; lung cancer ID S1P; anti-ceramide antibody; NSCLC; biomarkers; lung cancer AB Objectives: Spingosine-1-phosphate (S1P) and ceramides are bioactive sphingolipids that influence cancer cell fate. Anti-ceramide antibodies might inhibit the effects of ceramide. The aim of this study was to assess the potential role of circulating S1P and anti-ceramide antibody as biomarkers in non-small cell lung cancer (NSCLC). Methods: We recruited 66 subjects (34 controls and 32 patients with NSCLC). Patient history and clinical variables were taken from all participants. Venous blood samples were collected to evaluate plasma biomarkers. If bronchoscopy was performed, bronchial washing fluid (BWF) was also analyzed. We measured the levels of S1P and anti-ceramide antibody with ELISA. Results: S1P levels were significantly higher in the NSCLC group (3770.99 ± 762.29 ng/mL vs. 366.53 ± 249.38 ng/mL, patients with NSCLC vs. controls, respectively, p < 0.001). Anti-ceramide antibody levels were significantly elevated in the NSCLC group (278.70 ± 19.26 ng/mL vs. 178.60 ± 18 ng/mL, patients with NSCLC vs. controls, respectively, p = 0.007). Age or BMI had no significant effect on anti-ceramide antibody or S1P levels. BWF samples had higher levels of anti-ceramide antibody (155.29 ± 27.58 ng/mL vs. 105.87 ± 9.99 ng/mL, patients with NSCLC vs. controls, respectively, p < 0.001). Overall survival (OS) was 13.36 months. OS was not affected by anti-ceramide antibody or S1P levels. Conclusion: Higher levels of S1P and anti-ceramide antibody were associated with active cancer. These results suggest that sphingolipid alterations might be important features of NSCLC. C1 [Budi, Lilla] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Hammer, Daniel] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Varga, Rita] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Muller, Veronika] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Tarnoki, Domonkos Adam] Semmelweis University, Medical Imaging CenterBudapest, Hungary. [Tarnoki, Laszlo David] Semmelweis University, Medical Imaging CenterBudapest, Hungary. [Meszaros, Martina] Semmelweis University, Department of PulmonologyBudapest, Hungary. [Bikov, Andras] Manchester Academic Health Science Centre, Manchester University National Health Service Foundation Trust, Wythenshawe HospitalManchester, UK. [Horvath, Peter] Semmelweis University, Department of PulmonologyBudapest, Hungary. RP Budi, L (reprint author), Semmelweis University, Department of Pulmonology, Budapest, Hungary. EM budi.lilla@semmelweis.hu CR Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021, 71(3):209–49., DOI 10.3322/caac.21660 Falzone L, Salomone S, Libra M. Evolution of cancer pharmacological treatments at the turn of the third millennium. Front Pharmacol, 2018, 9:1300., DOI 10.3389/fphar.2018.01300 Tsimberidou AM, Fountzilas E, Nikanjam M, Kurzrock R. Review of precision cancer medicine: evolution of the treatment paradigm. Cancer Treat Rev, 2020, 86: 102019., DOI 10.1016/j.ctrv.2020.102019 Shah DR, Masters GA. Precisionmedicine in lung cancer treatment. Surg Oncol Clin N Am, 2020, 29(1):15–21., DOI 10.1016/j.soc.2019.08.002 Fang H, Declerck YA. Targeting the tumor microenvironment: from understanding pathways to effective clinical trials. Cancer Res, 2013, 73(16): 4965–77., DOI 10.1158/0008-5472.CAN-13-0661 Ogretmen B, Hannun YA. Biologically active sphingolipids in cancer pathogenesis and treatment. Nat Rev Cancer, 2004, 4(8):604–16., DOI 10.1038/ nrc1411 Nagahashi M, Hait NC, Maceyka M, Avni D, Takabe K, Milstien S, et al. Sphingosine-1-phosphate in chronic intestinal inflammation and cancer. Adv Biol Regul, 2014, 54:112–20., DOI 10.1016/j.jbior.2013.10.001 Kunkel GT, Maceyka M, Milstien S, Spiegel S. Targeting the sphingosine-1- phosphate axis in cancer, inflammation and beyond. Nat Rev Drug Discov, 2013, 12(9):688–702., DOI 10.1038/nrd4099 Sedic M, Grbcic P, Pavelic SK. Bioactive sphingolipids as biomarkers predictive of disease severity and treatment response in cancer: current status and translational challenges. Anticancer Res, 2019, 39(1):41–56., DOI 10.21873/anticanres.13078 Nagahashi M, Abe M, Sakimura K, Takabe K, Wakai T. The role of sphingosine-1-phosphate in inflammation and cancer progression. Cancer Sci, 2018, 109(12):3671–8., DOI 10.1111/cas.13802 Gault CR, Obeid LM, Hannun YA. An overview of sphingolipid metabolism: from synthesis to breakdown. In: Chalfant C, Poeta MD, editors. Sphingolipids as signaling and regulatory molecules. New York, NY: Springer New York, 2010). p. 1–23. Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol, 2008, 9(2):139–50., DOI 10.1038/nrm2329 Ueda N. Ceramide-induced apoptosis in renal tubular cells: a role of mitochondria and sphingosine-1-phoshate. Int J Mol Sci, 2015, 16(3):5076–124., DOI 10.3390/ijms16035076 Sukocheva O, Wadham C. Role of sphingolipids in oestrogen signalling in breast cancer cells: an update. J Endocrinol, 2014, 220(3):R25–35., DOI 10.1530/JOE- 13-0388 Garcia-Barros M, Coant N, Truman JP, Snider AJ, Hannun YA. Sphingolipids in colon cancer. Biochim Biophys Acta, 2014, 1841(5):773–82., DOI 10.1016/j.bbalip. 2013.09.007 Furuya H, Shimizu Y, Kawamori T. Sphingolipids in cancer. Cancer Metastasis Rev, 2011, 30(3-4):567–76., DOI 10.1007/s10555-011-9304-1 Zaky M, Obaid Z, Khedr M, El-Sokkary H, Elsaie M. Implications of serum anti-ceramide antibodies and interleukin 4 on nerve damage and physical impairments among leprotic patients: a case-controlled study. J Drugs Dermatol, 2022, 21(3):284–91., DOI 10.36849/JDD.5543 Rotolo J, Stancevic B, Zhang J, Hua G, Fuller J, Yin X, et al. Anti-ceramide antibody prevents the radiation gastrointestinal syndrome in mice. J Clin Invest, 2012, 122(5):1786–90., DOI 10.1172/JCI59920 Zhang L, Wang X, Bullock AJ, Callea M, Shah H, Song J, et al. Anti-S1P antibody as a novel therapeutic strategy for VEGFR TKI-resistant renal cancer. Clin Cancer Res, 2015, 21(8):1925–34., DOI 10.1158/1078-0432.CCR-14-2031 Brierley J, Gospodarowicz MK, Wittekind C. TNM classification of malignant tumours. 8th ed. ed. Chichester, West Sussex, UK; Hoboken, NJ: Wiley Blackwell; 2017. xviii, 253 pages p. Faul F, Erdfelder E, Buchner A, Lang A-G. Statistical power analyses using G*Power 3.1: tests for correlation and regression analyses. Behav Res Methods, 2009, 41(4):1149–60., DOI 10.3758/BRM.41.4.1149 National Center for Chronic Disease P, Health Promotion Office on S, Health. Reports of the surgeon general. The Health consequences of smoking—50 Years of progress: a report of the surgeon general. Atlanta, GA): Centers for Disease Control and Prevention, US, 2014). Shrotriya S, Walsh D, Bennani-Baiti N, Thomas S, Lorton C. C-reactive protein is an important biomarker for prognosis tumor recurrence and treatment response in adult solid tumors: a systematic review. PLoS One, 2015, 10(12): e0143080., DOI 10.1371/journal.pone.0143080 Hait NC, Maiti A. The role of sphingosine-1-phosphate and ceramide-1- phosphate in inflammation and cancer. Mediators Inflamm, 2017, 2017:4806541., DOI 10.1155/2017/4806541 Maceyka M, Harikumar KB, Milstien S, Spiegel S. Sphingosine-1-phosphate signaling and its role in disease. Trends Cell Biol, 2012, 22(1):50–60., DOI 10.1016/j. tcb.2011.09.003 Lima S, Milstien S, Spiegel S. Sphingosine and sphingosine kinase 1 involvement in endocytic membrane trafficking. J Biol Chem, 2017, 292(8): 3074–88., DOI 10.1074/jbc.M116.762377 Ogretmen B. Sphingolipid metabolism in cancer signalling and therapy. Nat Rev Cancer, 2018, 18(1):33–50., DOI 10.1038/nrc.2017.96 Sutphen R, Xu Y, Wilbanks GD, Fiorica J, Grendys EC, Jr, LaPolla JP, et al. Lysophospholipids are potential biomarkers of ovarian cancer. Cancer Epidemiol Biomarkers Prev, 2004, 13(7):1185–91., DOI 10.1158/1055-9965.1185.13.7 Grammatikos G, Schoell N, Ferreiros N, Bon D, Herrmann E, Farnik H, et al. Serum sphingolipidomic analyses reveal an upregulation of C16-ceramide and sphingosine-1-phosphate in hepatocellular carcinoma. Oncotarget, 2016, 7(14): 18095–105., DOI 10.18632/oncotarget.7741 Alberg AJ, Armeson K, Pierce JS, Bielawski J, Bielawska A, Visvanathan K, et al. Plasma sphingolipids and lung cancer: a population-based, nested case-control study. Cancer Epidemiol Biomarkers Prev, 2013, 22(8):1374–82., DOI 10.1158/1055- 9965.EPI-12-1424 Meng Q, Hu X, Zhao X, Kong X, Meng YM, Chen Y, et al. A circular network of coregulated sphingolipids dictates lung cancer growth and progression. EBioMedicine, 2021, 66:103301., DOI 10.1016/j.ebiom.2021.103301 Chen Y, Ma Z, Min L, Li H, Wang B, Zhong J, et al. Biomarker identification and pathway analysis by serum metabolomics of lung cancer. Biomed Res Int, 2015, 2015(1):183624., DOI 10.1155/2015/183624 McGinley MP, Cohen JA. Sphingosine 1-phosphate receptor modulators in multiple sclerosis and other conditions. Lancet, 2021, 398(10306):1184–94., DOI 10. 1016/S0140-6736(21)00244-0 Sahu SK, Hannun YA, Yao N. Emergence of membrane sphingolipids as a potential therapeutic target. Biochimie, 2019, 158:257–64., DOI 10.1016/j.biochi. 2019.01.018 Jiang W, Ogretmen B. Autophagy paradox and ceramide. Biochim Biophys Acta, 2014, 1841(5):783–92., DOI 10.1016/j.bbalip.2013.09.005 Giussani P, Prinetti A, Tringali C. The role of sphingolipids in cancer immunotherapy. Int J Mol Sci, 2021, 22(12):6492., DOI 10.3390/ijms22126492 Ogretmen B, Hannun YA. Updates on functions of ceramide in chemotherapy-induced cell death and in multidrug resistance. Drug Resist Updat, 2001, 4(6):368–77., DOI 10.1054/drup.2001.0225 Senkal CE, Ponnusamy S, Rossi MJ, Sundararaj K, Szulc Z, Bielawski J, et al. Potent antitumor activity of a novel cationic pyridinium-ceramide alone or in combination with gemcitabine against human head and neck squamous cell carcinomas in vitro and in vivo. J Pharmacol Exp Ther, 2006, 317(3):1188–99., DOI 10.1124/jpet.106.101949 Beckham TH, Lu P, Jones EE, Marrison T, Lewis CS, Cheng JC, et al. LCL124, a cationic analog of ceramide, selectively induces pancreatic cancer cell death by accumulating in mitochondria. J Pharmacol Exp Ther, 2013, 344(1):167–78., DOI 10. 1124/jpet.112.199216 Liu X, Ryland L, Yang J, Liao A, Aliaga C, Watts R, et al. Targeting of survivin by nanoliposomal ceramide induces complete remission in a rat model of NK-LGL leukemia. Blood, 2010, 116(20):4192–201., DOI 10.1182/blood-2010-02-271080 Nadler E, Espirito JL, Pavilack M, Boyd M, Vergara-Silva A, Fernandes A. Treatment patterns and clinical outcomes among metastatic non–small-cell lung cancer patients treated in the community practice setting. Clin Lung Cancer, 2018, 19(4):360–70., DOI 10.1016/j.cllc.2018.02.002 Simeone JC, NordstromBL, Patel K, Klein AB. Treatment patterns and overall survival in metastatic non-small-cell lung cancer in a real-world, US setting. Future Oncol, 2019, 15(30):3491–502., DOI 10.2217/fon-2019-0348 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1611929 EP 1611938 DI 10.3389/pore.2024.1611929 PG 10 ER PT J AU Hou, Ch Song, X Chen, H Chang, Ch Lu, J Li, Ch Qu, H Guo, R Xu, J Xu, L AF Hou, Chunxiao Song, Xueru Chen, Hongwei Chang, Chengdong Lu, Jinfeng Li, Cheng Qu, Haiyan Guo, Rui Xu, Jingyi Xu, Liming TI A novel automated IHC staining system for quality control application in ALK immunohistochemistry testing SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE automated; quality control; anaplastic lymphoma kinase gene ALK; rabbit monoclonal antibody; controls in liquid form (CLFs); lung adenocarcinoma ID automated; quality control; anaplastic lymphoma kinase gene ALK; rabbit monoclonal antibody; controls in liquid form (CLFs); lung adenocarcinoma AB The establishment of positive and negative controls in immunohistochemistry (IHC) screening for anaplastic lymphoma kinase (ALK) rearrangements is essential in the treatment of lung adenocarcinoma. However, positive control of patient tissue is rare and comes with ethical issues. A novel automated solution for ALK IHC quality control management was investigated by comparison with the established D5F3 antibody on the VENTANA system in 87 lung adenocarcinoma specimens with known ALK status re-analyzed by fluorescence in situ hybridization. The BP6165 concentrated antibody on the LYNX480 PLUS platform demonstrated excellent sensitivity and specificity (98.30% and 100%, respectively) in 87 biopsy specimens. The ALK controls in liquid form (CLFs) applied in an automated way showed a more regular circular shape and better cell distribution than those applied manually. In addition, the novel controls can show changes in the same pattern as tissue controls under different antibody concentrations and antigen retrieval conditions. The automated solution for ALK IHC quality control management provides a convenient solution without the consumption of scarce tissue for IHC testing in day-to-day pathology practice. The availability of standardized protocols for the detection of ALK rearrangements using the BP6165 concentrated antibody on the LYNX480 PLUS platform will expand the number of laboratories that can reliably and consistently determine the eligibility of patients with lung adenocarcinoma for treatment with ALK tyrosine kinase inhibitors. C1 [Hou, Chunxiao] Haining People’s Hospital, Department of PathologyHaining, Zhejiang, China. [Song, Xueru] Zhejiang University School of Medicine, Affiliated Hangzhou First People’s Hospital, Department of Surgical PathologyHangzhou, China. [Chen, Hongwei] Zhejiang University School of Medicine, Affiliated Hangzhou First People’s Hospital, Department of Surgical PathologyHangzhou, China. [Chang, Chengdong] Zhejiang University School of Medicine, Affiliated Hangzhou First People’s Hospital, Department of Surgical PathologyHangzhou, China. [Lu, Jinfeng] Hangzhou Bailing Biotechnology Co., Ltd.Hangzhou, China. [Li, Cheng] Hangzhou Bailing Biotechnology Co., Ltd.Hangzhou, China. [Qu, Haiyan] Hangzhou Bailing Biotechnology Co., Ltd.Hangzhou, China. [Guo, Rui] Hangzhou Bailing Biotechnology Co., Ltd.Hangzhou, China. [Xu, Jingyi] Hangzhou Bailing Biotechnology Co., Ltd.Hangzhou, China. [Xu, Liming] Zhejiang University School of Medicine, Affiliated Hangzhou First People’s Hospital, Department of Surgical PathologyHangzhou, China. RP Hou, Ch (reprint author), Haining People’s Hospital, Department of Pathology, Haining, China. EM 110080389@qq.com CR Thunnissen E, Bubendorf L, Dietel M, Elmberger G, Kerr K, Lopez-Rios F, et al. Eml4-ALK testing in non-small cell carcinomas of the lung: a review with recommendations. Virchows Arch, 2012, 461:245–57., DOI 10.1007/s00428-012-1281-4 Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, et al. Identification of the transforming Eml4-ALK fusion gene in non-small-cell lung cancer. Nature, 2007, 448:561–6., DOI 10.1038/nature05945 Sullivan I, Planchard D. ALK inhibitors in non-small cell lung cancer: the latest evidence and developments. Ther Adv Med Oncol, 2016, 8:32–47., DOI 10.1177/ 1758834015617355 De Mello RA, Liu DJ, Aguiar PN, Tadokoro H. Egfr and Eml4-ALK updated therapies in non-small cell lung cancer. Recent Pat Anticancer Drug Discov, 2016, 11:393–400., DOI 10.2174/1574892811666160803090944 Imyanitov EN, Iyevleva AG, Levchenko EV. Molecular testing and targeted therapy for non-small cell lung cancer: current status and perspectives. Crit Rev Oncol Hematol, 2021, 157:103194., DOI 10.1016/j.critrevonc.2020.103194 Shaw AT, Engelman JA. ALK in lung cancer: past, present, and future. J Clin Oncol, 2013, 31:1105–11., DOI 10.1200/jco.2012.44.5353 Niu X, Chuang JC, Berry GJ, Wakelee HA. Anaplastic lymphoma kinase testing: IHC vs. FISH vs. NGS. Curr Treat Options Oncol, 2017, 18:71., DOI 10.1007/ s11864-017-0513-x Larkins E, Blumenthal GM, Chen H, He K, Agarwal R, Gieser G, et al. FDA approval: alectinib for the treatment ofmetastatic, ALK-positive non-small cell lung cancer following crizotinib. Clin Cancer Res, 2016, 22:5171–6., DOI 10.1158/1078- 0432.Ccr-16-1293 Conde E, Hernandez S, Prieto M, Martinez R, Lopez-Rios F. Profile of ventana ALK, D5f3, companion diagnostic assay for non-small-cell lung carcinomas. Expert Rev Mol Diagn, 2016, 16:707–13., DOI 10.1586/14737159.2016.1172963 Duma N, Santana-Davila R, Molina JR. Non-small cell lung cancer: epidemiology, screening, diagnosis, and treatment. Mayo Clin Proc, 2019, 94: 1623–40., DOI 10.1016/j.mayocp.2019.01.013 Wynes MW, Sholl LM, Dietel M, Schuuring E, Tsao MS, Yatabe Y, et al. An international interpretation study using the ALK IHC antibody D5F3 and a sensitive detection kit demonstrates high concordance between ALK IHC and ALK FISH and between evaluators. J Thorac Oncol, 2014, 9:631–8., DOI 10.1097/jto. 0000000000000115 Mino-Kenudson M, Chirieac LR, Law K, Hornick JL, Lindeman N, Mark EJ, et al. A novel, highly sensitive antibody allows for the routine detection of ALKrearranged lung adenocarcinomas by standard immunohistochemistry. Clin Cancer Res, 2010, 16:1561–71., DOI 10.1158/1078-0432.Ccr-09-2845 Wu YC, Chang IC,Wang CL, Chen TD, Chen YT, Liu HP, et al. Comparison of IHC, FISH and RT-PCR methods for detection of ALK rearrangements in 312 non-small cell lung cancer patients in Taiwan. PLoS One, 2013, 8:e70839., DOI 10.1371/journal.pone.0070839 Boland JM, Erdogan S, Vasmatzis G, Yang P, Tillmans LS, Johnson MR, et al. Anaplastic lymphoma kinase immunoreactivity correlates with ALK gene rearrangement and transcriptional up-regulation in non-small cell lung carcinomas. Hum Pathol, 2009, 40:1152–8., DOI 10.1016/j.humpath.2009.01.012 Rimm DL. Next-gen immunohistochemistry. Nat Methods, 2014, 11:381–3., DOI 10.1038/nmeth.2896 Taylor C, Shi S-R, Barr N, Wu N. Techniques of immunohistochemistry: principles, pitfalls and standardization. In: Dabbs DJ, editor. Diagnostic immunohistochemistry. 2nd ed., Churchill Livingstone,, 2006). p. 1–42. Hewitt SM, Robinowitz M. Quality assurance for design control and implementation of immunohistochemistry assays; approved guideline. 2nd ed. Clinical and Laboratory Standards Institute, 2011). p. 152. Torlakovic EE, Francis G, Garratt J, Gilks B, Hyjek E, Ibrahim M, et al. Standardization of negative controls in diagnostic immunohistochemistry: recommendations from the international ad hoc expert panel. Appl Immunohistochem Mol Morphol, 2014, 22:241–52., DOI 10.1097/pai. 0000000000000069 Reis-Filho JS, Pusztai L. Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet, 2011, 378:1812–23., DOI 10.1016/s0140-6736(11)61539-0 McLeer-Florin A, Moro-Sibilot D, Melis A, Salameire D, Lefebvre C, Ceccaldi F, et al. Dual IHC and FISH testing for ALK gene rearrangement in lung adenocarcinomas in a routine practice: a French study. J Thorac Oncol, 2012, 7: 348–54., DOI 10.1097/JTO.0b013e3182381535 Selinger CI, Rogers TM, Russell PA, O’Toole S, Yip P, Wright GM, et al. Testing for ALK rearrangement in lung adenocarcinoma: a multicenter comparison of immunohistochemistry and fluorescent in situ hybridization. Mod Pathol, 2013, 26:1545–53., DOI 10.1038/modpathol.2013.87 Le Quesne J,Maurya M, Yancheva SG, O’Brien M, Popat S,Wotherspoon AC, et al. A comparison of immunohistochemical assays and FISH in detecting the ALK translocation in diagnostic histological and cytological lung tumor material. J Thorac Oncol, 2014, 9:769–74., DOI 10.1097/jto.0000000000000157 Hutarew G, Hauser-Kronberger C, Strasser F, Llenos IC, Dietze O. Immunohistochemistry as a screening tool for ALK rearrangement in NSCLC: evaluation of five different ALK antibody clones and ALK FISH. Histopathology, 2014, 65:398–407., DOI 10.1111/his.12399 Shan L, Lian F, Guo L, Yang X, Ying J, Lin D. Combination of conventional immunohistochemistry and qRT-PCR to detect ALK rearrangement. Diagn Pathol, 2014, 9:3., DOI 10.1186/1746-1596-9-3 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1611964 EP 1611974 DI 10.3389/pore.2025.1611964 PG 11 ER PT J AU Almasi, Sz Balajthy, Zs Barath, B Torok, KZs Szaszak, P Lantos, T Kovari, B Sejben, A AF Almasi, Szintia Balajthy, Zsofia Barath, Bence Torok, Krisztina Zsofia Szaszak, Panna Lantos, Tamas Kovari, Bence Sejben, Anita TI Examination of non-conventional dysplasias adjacent to colorectal adenocarcinoma in patients with IBD SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE non-conventional dysplasia; colorectal adenocarcinoma; inflammatory bowel disease; ulcerative colitis; Crohn’s disease ID non-conventional dysplasia; colorectal adenocarcinoma; inflammatory bowel disease; ulcerative colitis; Crohn’s disease AB Objective: Recently, several non-conventional variants of IBD-associated dysplasia have been described; however, their prevalence in Central-Eastern Europe is unknown. We aimed to perform a retrospective pilot study by reevaluating several IBD-associated adenocarcinoma cases to survey the incidence of adjacent non-conventional dysplasia and validate that recent North American findings may apply to a European population. Methods: Retrospectively, 28 randomly chosen cases of IBD-associated adenocarcinomas diagnosed between 2010 and 2022 were re-evaluated. The patient’s sex, age (at the diagnosis of IBD and neoplasia), type of IBD, type of specimen [biopsy (n = 8)/surgical specimen (n = 20)], histological type, grade, localisation, stage, disease-free (DFS) and overall survival (OS) were obtained. Statistical analyses were carried out by using Mann-Whitney (continuous variables), Fisher’s exact (categorical variables), Kaplan-Meier (DFS/OS curves), and logrank test (survival curves). Results: Exclusively, conventional dysplasia was observed in 11, and nonconventional dysplasia in 8 patients. Combined conventional and nonconventional dysplasia was detected in 9 patients. Non-conventional dysplasia showing a combination of multiple subtypes was noted in 10 cases. Altogether, 25 non-conventional dysplastic foci were identified, which were diagnosed as hypermucinous (n = 9), goblet cell-deficient (n = 6), serrated not otherwise specified (NOS) (n = 6), and traditional serrated adenoma-like (n = 4). The majority of non-conventional dysplasias were associated with ulcerative colitis (n = 12). Mucinous adenocarcinoma was exclusively associated with non-conventional dysplasia, while medullary carcinoma was only with conventional dysplasias (p = 0.014 and 0.041). Conclusion: Based on our results, non-conventional dysplasia is common (60%) adjacent to IBD-associated adenocarcinomas in a Central-Eastern European population and may be detected in biopsies. As multiple recent publications reported evidence of a worse prognosis and more common flat morphology compared to conventional dysplasias, their recognition is of great importance, and stricter follow-up with random biopsy samples may be considered. C1 [Almasi, Szintia] University of Szeged, Albert Szent-Gyorgyi Medical SchoolSzeged, Hungary. [Balajthy, Zsofia] University of Szeged, Albert Szent-Gyorgyi Medical SchoolSzeged, Hungary. [Barath, Bence] University of Szeged, Albert Szent-Gyorgyi Medical SchoolSzeged, Hungary. [Torok, Krisztina Zsofia] University of Szeged, Albert Szent-Gyorgyi Medical SchoolSzeged, Hungary. [Szaszak, Panna] University of Szeged, Albert Szent-Gyorgyi Medical SchoolSzeged, Hungary. [Lantos, Tamas] University of Szeged, Albert Szent-Gyorgyi Medical SchoolSzeged, Hungary. [Kovari, Bence] Harvard Medical School, Mass General BrighamBoston, MA, USA. [Sejben, Anita] University of Szeged, Albert Szent-Gyorgyi Medical SchoolSzeged, Hungary. RP Sejben, A (reprint author), University of Szeged, Albert Szent-Gyorgyi Medical School, Szeged, Hungary. EM sejben.anita@gmail.com CR Nagao-Kitamoto H, Kitamoto S, Kamada N. Inflammatory bowel disease and carcinogenesis. Cancer Metastasis Rev, 2022, 41:301–16., DOI 10.1007/s10555-022- 10028-4 Katsanos KH, Vermeire S, Christodoulou DK, Riis L, Wolters F, Odes S, et al. Dysplasia and cancer in inflammatory bowel disease 10 years after diagnosis: results of a population-based European collaborative follow-up study. Digestion, 2007, 75: 113–21., DOI 10.1159/000104731 Soderlund S, Brandt L, Lapidus A, Karlen P, Brostrom O, Lofberg R, et al. Decreasing time-trends of colorectal cancer in a large cohort of patients with inflammatory bowel disease. Gastroenterology, 2009, 136:1561–819., DOI 10.1053/j. gastro.2009.01.064 Choi WT, Yozu M, Miller GC, Shih AR, Kumarasinghe P, Misdraji J, et al. Nonconventional dysplasia in patients with inflammatory bowel disease and colorectal carcinoma: a multicenter clinicopathologic study. Mod Pathol, 2020, 33:933–43., DOI 10.1038/s41379-019-0419-1 Choi WT, Salomao M, Zhao L, Alpert L, Setia N, Liao X, et al. Hypermucinous, goblet cell-deficient and crypt cell dysplasias in inflammatory bowel disease are often associated with flat/invisible endoscopic appearance and advanced neoplasia on follow-up. J Crohns Colitis, 2022, 16:98–108., DOI 10.1093/ecco-jcc/jjab120 Choi WT, Kovari BP, Lauwers GY. The significance of flat/invisible dysplasia and nonconventional dysplastic subtypes in inflammatory bowel disease: a review of their morphologic, clinicopathologic, and molecular characteristics. Adv Anat Pathol, 2022, 29:15–24., DOI 10.1097/pap.0000000000000316 Riddell RH, Goldman H, Ransohoff DF, Appelman HD, Fenoglio CM, Haggitt RC, et al. Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Mod Pathol, 1983, 14:931–68., DOI 10.1016/s0046-8177(83)80175-0 Andersen SN, Lovig T, Clausen OP, Bakka A, Fausa O, Rognum TO. Villous, hypermucinous mucosa in long standing ulcerative colitis shows high frequency of K-ras mutations. Gut, 1999, 45:686–92., DOI 10.1136/gut.45.5.686 Patil DT, Goldblum JR, Odze RD. Immunohistochemical and molecular characterisation of dysplasia subtypes in ulcerative colitis. Mod Pathol, 2017, 30:194A. Lee H, Rabinovitch PS, Mattis AN, Lauwers GY, Choi WT. Non-conventional dysplasia in inflammatory bowel disease is more frequently associated with advanced neoplasia and aneuploidy than conventional dysplasia. Histopathology, 2021, 78:814–30., DOI 10.1111/his.14298 Zhang R, Lauwers GY, Choi WT. Increased risk of non-conventional and invisible dysplasias in patients with primary sclerosing cholangitis and inflammatory bowel disease. J Crohns Colitis, 2022, 16:1825–34., DOI 10.1093/ecco-jcc/jjac090 Nguyen ED, Wang D, Lauwers GY, Choi WT. Increased histologic inflammation is an independent risk factor for nonconventional dysplasia in ulcerative colitis. Histopathology, 2022, 81:644–52., DOI 10.1111/his.14765 Bahceci D, Lauwers GY, Choi WT. Clinicopathologic features of undetected dysplasia found in total colectomy or proctocolectomy specimens of patients with inflammatory bowel disease. Histopathology, 2022, 81:183–91., DOI 10.1111/his. 14673 Akarca FG, Yozu M, Alpert L, Kovari BP, Zhao L, Salomao M, et al. Nonconventional dysplasia is frequently associated with low-grade tubuloglandular and mucinous adenocarcinomas in inflammatory bowel disease. Histopathology, 2023, 83:276–85., DOI 10.1111/his.14922 Zhang R, Rabinovitch PS, Mattis AN, Lauwers GY, Choi WT. DNA content abnormality frequently develops in the right/proximal colon in patients with primary sclerosing cholangitis and inflammatory bowel disease and is highly predictive of subsequent detection of dysplasia. Histopathology, 2023, 83: 116–25., DOI 10.1111/his.14913 Lang-Schwarz C, Buttner-Herold M, Burian S, Erber R, Hartmann A, Jesinghaus M, et al. Morphological subtypes of colorectal low-grade intraepithelial neoplasia: diagnostic reproducibility, frequency and clinical impact. J Clin Pathol, 2023, 209206–2023-209206., DOI 10.1136/jcp-2023-209206 Nasreddin N, Jansen M, Loughrey MB,Wang LM, Koelzer VH, Rodriguez-Justo M, et al. Poor diagnostic reproducibility in the identification of nonconventional dysplasia in colitis impacts the application of histologic stratification tools. Mod Pathol, 2023, 37:100419., DOI 10.1016/j.modpat.2023.100419 Musulen E, Gene M, Cuatrecasas M, Amat I, Veiga JA, Fernandez-Acenero MJ, et al. Gastric metaplasia as a precursor of nonconventional dysplasia in inflammatory bowel disease. Hum Pathol, 2023, 143:50–61., DOI 10.1016/j. humpath.2023.11.011 Harpaz N, Goldblum JR, Shepherd NA, Riddell RH, Rubio CA, Vieth M, et al. Colorectal dysplasia in chronic inflammatory bowel disease: a contemporary consensus classification and interobserver study. Hum Pathol, 2023, 138:49–61., DOI 10.1016/j.humpath.2023.05.008 Wen KW, Umetsu SE, Goldblum JR, Gill RM, Kim GE, JosephNM, et al. DNA flow cytometric and interobserver study of crypt cell atypia in inflammatory bowel disease. Histopathology, 2019, 75:578–88., DOI 10.1111/his.13923 Miller GC, Liu C, Bettington ML, Leggett B, Whitehall VL, Rosty C. Traditional serrated adenoma-like lesions in patients with inflammatory bowel disease. Hum Pathol, 2020, 97:19–28., DOI 10.1016/j.humpath.2019.12.005 Kovari B, Clauditz T, Kamaradova K, Bathori A, Hegedus F, Miller G, et al. Hypermucinous dysplasia in inflammatory bowel disease: a heterogeneous condition with frequent foveolar differentiation. Mod Pathol, 2022, 35:481–2., DOI 10.1038/s41379-022-01036-4 Sz A, Barath B, Szaszak P, Kovari B, Sejben A. Hypermucinous and goblet cell deficient, IBD-associated, non-conventional dysplasia besides colorectal adenocarcinoma – case presentation. Orv Hetil, 2023, 164:2039–44., DOI 10.1556/ 650.2023.32946 Guerini C, Furlan D, Ferrario G, Grillo F, Libera L, Arpa G, et al. IDH1- mutated Crohn’s disease-associated small bowel adenocarcinomas: distinctive pathological features and association with MGMT methylation and serratedtype dysplasia. Histopathology, 2024, 84:515–24., DOI 10.1111/his.15095 Arpa G, Vanoli A, Grillo F, Fiocca R, Klersy C, Furlan D, et al. Prognostic relevance and putative histogenetic role of cytokeratin 7 and MUC5AC expression in Crohn’s disease-associated small bowel carcinoma. Virchows Arch, 2021, 479: 667–78., DOI 10.1007/s00428-021-03109-2 Pereira D, Kovari B, Brown I, Chaves P, Choi WT, Clauditz T, et al. Nonconventional dysplasias of the tubular gut: a review and illustration of their histomorphological spectrum. Histopathology, 2021, 78:658–75., DOI 10.1111/his.14294 Choi WT. Non-conventional dysplastic subtypes in inflammatory bowel disease: a review of their diagnostic characteristics and potential clinical implications. J Pathol Transl Med, 2021, 55:83–93., DOI 10.4132/jptm.2021.02.17 Kamaradova K. Non-conventional types of dysplastic changes in gastrointestinal tract mucosa - review of morphological features of individual subtypes. Cesk Patol, 2022, 58:38–51. Choi WT. Characteristics, reporting, and potential clinical significance of nonconventional dysplasia in inflammatory bowel disease. Surg Pathol Clin, 2023, 16:687–702., DOI 10.1016/j.path.2023.05.006 Waters KM, Singhi AD, Montgomery EA. Exploring the spectrum of serrated epithelium encountered in inflammatory bowel disease. Hum Pathol, 2023, 132: 126–34., DOI 10.1016/j.humpath.2022.06.018 Enea D, Lauwers G, Svrcek M. Dysplasies conventionnelles et non conventionnelles compliquant les maladies inflammatoires chroniques de l’intestin [Conventional and non-conventional dysplasia in patients with inflammatory bowel disease]. Ann Pathol, 2023, 43:180–91., DOI 10.1016/j.annpat.2023.02.006 Alipour Z, Stashek K. Recently described types of dysplasia associated with IBD: tips and clues for the practising pathologist. J Clin Pathol, 2024, 77:77–81., DOI 10.1136/jcp-2023-209141 Kamaradova K, Vosmikova H, Rozkosova K, Ryska A, Tacheci I, Laco J. Morphological, immunohistochemical and molecular features of inflammatory bowel disease associated colorectal carcinoma and associated mucosal lesions - single institution experience. Pathol Res Pract, 2019, 215:730–7., DOI 10.1016/j.prp. 2019.01.010 Kamaradova K, Vosmikova H, Rozkosova K, Ryska A, Tacheci I, Laco J. Nonconventional mucosal lesions, serrated epithelial change, villous hypermucinous change, are frequent in patients with inflammatory bowel disease-results of molecular and immunohistochemical single institutional study. Virchows Arch, 2020, 476:231–41., DOI 10.1007/s00428-019-02627-4 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1611978 EP 1611989 DI 10.3389/pore.2024.1611978 PG 12 ER PT J AU Francis, SS Sharma, S AF Francis, Sebastian Sachin Sharma, Swati TI Tumour budding in invasive ductal breast carcinomas: correlation with clinicopathological prognostic parameters and hormone receptor status SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE invasive breast carcinoma; intra tumoral budding; peripheral tumour budding; prognostic parameters; Ki-67 ID invasive breast carcinoma; intra tumoral budding; peripheral tumour budding; prognostic parameters; Ki-67 AB Introduction: Breast cancer is a leading cause of morbidity and mortality among women. Advances in molecular biology have improved detection and treatment, but conventional histopathological factors remain crucial for prognosis. Tumour budding, defined as clusters of less than 5 tumour cells detached from the main tumour, has been linked to poor prognosis in several cancers. This study explores the association between intra-tumoral budding (ITB) and peripheral tumour budding (PTB) with known prognostic factors in Invasive Breast Carcinoma of no special type (IBC NST). Materials and methods: This retrospective study analysed 70 cases of IBC NST diagnosed at Kasturba Medical College, Manipal, between January 2020 and December 2021. Tumour budding was classified as high-grade or low-grade based on density, which denotes the number of buds per x20 field. Clinicopathological data, including hormone receptor status, Ki-67 index, lymphovascular invasion (LVI), perineural invasion (PNI), and axillary lymph node involvement, were obtained. Statistical analyses were performed to identify a significant association between tumour budding and these factors. Univariate and multivariate logistic regression analyses were also done to demonstrate the significance of association. Results: High-grade PTB showed significant associations with LVI (p = 0.046), PNI (p = 0.017), and axillary lymph node involvement (p = 0.021). In contrast, high-grade ITB was only significantly correlated with axillary lymph node involvement (p = 0.044). LVI (p-value = 0.240) and axillary lymph node involvement (p-value = 0.142) did not show any association with PTB on multivariate analysis and PNI (p-value = 0.074) near significant association with PTB). A significant inverse association was observed between PTB and Ki-67 (p = 0.012), which remained significant in univariate and multivariate analysis (p-value = 0.017). No significant associations were found between tumour budding and hormone receptor status or menopausal status. Conclusion: Peripheral tumour budding (PTB) is significantly associated with several poor prognostic factors in IBC NST, while intra-tumoral budding (ITB) correlates primarily with axillary lymph node involvement. Tumor budding, particularly PTB, could serve as an important prognostic marker in breast cancer. Further research is needed to standardize tumour budding assessment in clinical practice. C1 [Francis, Sebastian Sachin] Manipal Academy of Higher Education, Kasturba Medical College, Center of Basic Sciences, Department of PathologyManipal, Karnataka, India. [Sharma, Swati] Manipal Academy of Higher Education, Kasturba Medical College, Center of Basic Sciences, Department of PathologyManipal, Karnataka, India. RP Sharma, S (reprint author), Manipal Academy of Higher Education, Kasturba Medical College, Center of Basic Sciences, Department of Pathology, Manipal, India. EM swati.sharma@manipal.edu CR Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin, 2020, 70(1):7–30., DOI 10.3322/caac.21590 Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2024, 74(3):229–63., DOI 10.3322/caac.21834 Liang F, Cao W, Wang Y, Li L, Zhang G, Wang Z. The prognostic value of tumor budding in invasive breast cancer. Pathol - Res Pract, 2013, 209(5):269–75., DOI 10.1016/j.prp.2013.01.009 Mozarowski P, Rasaiah B, Reed M, Lewis A, Walde N, Voutsadakis IA. Prognostic role of tumor budding in breast cancer patients receiving neoadjuvant therapy. J Clin Med, 2021, 10(4):827., DOI 10.3390/jcm10040827 Voutsadakis IA. Prognostic role of tumor budding in breast cancer. World J Exp Med, 2018, 8(2):12–7., DOI 10.5493/wjem.v8.i2.12 Grigore A, Jolly M, Jia D, Farach-Carson M, Levine H. Tumor budding: the name is EMT. Partial EMT. J Clin Med, 2016, 5(5):51., DOI 10.3390/jcm5050051 Dawson H, Lugli A. Molecular and pathogenetic aspects of tumor budding in colorectal cancer. Front Med, 2015, 2:11., DOI 10.3389/fmed.2015.00011 Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest, 2009, 119:1420–8., DOI 10.1172/JCI39104 Micalizzi DS, Farabaugh SM, Ford HL. Epithelial-mesenchymal transition in cancer: parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia, 2010, 15:117–34., DOI 10.1007/s10911-010-9178-9 Nieto MA. Epithelial plasticity: a common theme in embryonic and cancer cells. Science, 2013, 342:1234850., DOI 10.1126/science.1234850 Imai T. Growth patterns in human carcinoma. Their classification and relation to prognosis. Obstet Gynecol, 1960, 16:296–308. Agarwal R, Khurana N, Singh T, Agarwal P. Tumor budding in infiltrating breast carcinoma: correlation with known clinicopathological parameters and hormone receptor status. Indian J Pathol Microbiol, 2019, 62(2):222–5., DOI 10. 4103/IJPM.IJPM_120_18 Li W, Lu N, Chen C, Lu X, Li W, Lu N, et al. Identifying the optimal cutoff point of Ki-67 in breast cancer: a single-center experience. J Int Med Res, 2023, 51(8):3000605231195468., DOI 10.1177/03000605231195468 Bustreo S, Osella-Abate S, Cassoni P, Donadio M, Airoldi M, Pedani F, et al. Optimal Ki67 cut-off for luminal breast cancer prognostic evaluation: a large case series study with a long-term follow-up. Breast Cancer Res Treat, 2016, 157:363–71., DOI 10.1007/s10549-016-3817-9 Salhia B, Trippel M, Pfaltz K, Cihoric N, Grogg A, Ladrach C, et al. High tumor budding stratifies breast cancer with metastatic properties. Breast Cancer Res Treat, 2015, 150(2):363–71., DOI 10.1007/s10549-015-3333-3 Silva DJ, Miranda G, Amaro T, Salgado M, Mesquita A. Prognostic value of tumor budding for early breast cancer. Biomedicines, 2023, 11(11):2906., DOI 10. 3390/biomedicines11112906 Gonzalez LO, Eiro N, Fraile M, Sanchez R, Andicoechea A, Fernandez- Francos S, et al. Joint tumor bud–MMP/TIMP count at the invasive front improves the prognostic evaluation of invasive breast carcinoma. Biomedicines, 2021, 9(2): 196., DOI 10.3390/biomedicines9020196 K A. The role of tumour budding as a prognostic factor in primary invasive ductal carcinoma of breast. Online J Health Allied Sci, 2022, 21(3), 9. Available from: https://www.ojhas.org/issue83/2022-3-9.html, Accessed January 22, 2024). Singh T, Chandra K, Kumar N, Mishra A, Singh S, Singh A, et al. A retrospective study of association of tumor budding, tumor microenvironment, and clinicopathological characteristics of invasive breast carcinoma. J Lab Physicians, 2022, 14(04):485–90., DOI 10.1055/s-0042-1747676 Gujam FJA, McMillan DC, Mohammed ZMA, Edwards J, Going JJ. The relationship between tumour budding, the tumour microenvironment and survival in patients with invasive ductal breast cancer. Br J Cancer, 2015, 113(7):1066–74., DOI 10.1038/bjc.2015.287 Muda NM, Deshmukh AV, Shivkumar VB, Shirbhate PN. Role of tumour budding in breast cancer and its correlation with other prognostic factors: a cross sectional study in rural Central India. Indian J Gynecol Oncol, 2024, 22(1):6., DOI 10. 1007/s40944-023-00769-7 Rathod GB, Desai KN, Shrivastava A, Maru AM. Correlation of tumor budding with known clinicopathological, histomorphological and hormonal receptor status in patients with invasive breast carcinoma. Cureus, 2022, 14(9): e29637., DOI 10.7759/cureus.29637 Gupta V, Patel M. Evaluation of prognostic role of tumour budding in breast carcinoma and its correlation with known clinicopathological parameters. Indian J Pathol Oncol, 2022, 9(3):238–42., DOI 10.18231/j.ijpo.2022.056 Ozturk C, Askan G, Ozturk SD, Okcu O, Sen B, Bedir R. Does the number of cell forming tumor budding alter the prognostic value in invasive ductal carcinoma of breast? Pathol - Res Pract, 2022, 240:154157., DOI 10.1016/j.prp. 2022.154157 Kumarguru B, Ramaswamy A, Shaik S, Karri A, Srinivas V, Prashant B. Tumor budding in invasive breast cancer - an indispensable budding touchstone. Indian J Pathol Microbiol, 2020, 63(5):117–S122., DOI 10.4103/ IJPM.IJPM_731_18 Masilamani DS, P DK. Evaluation of clinicopathologic significance of tumor budding in breast carcinoma. Int J Clin Diagn Pathol, 2019, 2(1):171–3., DOI 10. 33545/pathol.2019.v2.i1c.25 Svensson S, Nilsson K, Ringberg A, Landberg G. Invade or proliferate? Two contrasting events in malignant behavior governed by p16(INK4a, and an intact Rb pathway illustrated by a model system of basal cell carcinoma. Cancer Res, 2003, 63(8):1737–42. Jie G, Zhixiang S, Lei S, Hesheng L, Xiaojun T. Relationship between expression and methylation status of p16INK4a and the proliferative activity of different areas’ tumour cells in human colorectal cancer: p16INK4a gene and the proliferative activity of different areas’ tumour cells in colorectal cancer. Int J Clin Pract, 2007, 61(9):1523–9., DOI 10.1111/j.1742-1241.2006. 01033.x Kin R, Hoshi D, Fujita H, Kosaka T, Takamura H, Kiyokawa E. Prognostic significance of p16, p21, and Ki67 expression at the invasive front of colorectal cancers. Pathol Int, 2023, 73(2):81–90., DOI 10.1111/pin.13295 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1611983 EP 1611992 DI 10.3389/pore.2025.1611983 PG 10 ER PT J AU Qiu, Z Wu, J Pang, G Xu, X Lin, J Wang, P AF Qiu, Zijian Wu, Jiaji Pang, Guanchao Xu, Xia Lin, Jun Wang, Pingli TI CD34 evaluation of microvasculature in lung adenocarcinoma and its microvascular density predicts postoperative tumor recurrence SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE CD34; microvascular density; lung adenocarcinoma; prognosis; tumor recurrence ID CD34; microvascular density; lung adenocarcinoma; prognosis; tumor recurrence AB Background: Angiogenesis is closely associated with tumor growth and metastasis, and microvascular density (MVD) is currently the clinical standard for evaluating tumor angiogenesis. Thus, the detection of intratumoral MVD is of great significance for understanding disease progression and predicting patient prognosis. Methods: Tumor tissue sections of 238 patients with lung adenocarcinoma (LUAD) who underwent radical surgery were retrospectively analyzed. Immunohistochemical (IHC) staining was carried out using a CD34 polyclonal antibody to determine intratumoral MVD, and the relationship of CD34-MVD with the clinicopathological characteristics and survival time of LUAD patients was analyzed. Results: CD34-MVD was associated with tumor size, lymph node metastasis, tumor recurrence, and patient survival status; patients with tumor size ≤3 cm (P = 0.015), negative for lymph node metastasis (P = 0.049), no tumor recurrence (P = 0.021), and survival (P = 0.042) had higher MVD. Survival analysis suggested that patients with high MVD had higher disease-free survival (log-rank P = 0.005) and overall survival (log-rank P = 0.004) compared to patients with low MVD. The Cox proportional hazards model showed that a high MVD (P = 0.022) reduced the risk of postoperative tumor recurrence in patients with LUAD. Conclusion: Decreased intratumoral CD34 positive microvessels were associated with tumor development in patients with LUAD. CD34-MVD is an independent risk factor affecting postoperative tumor recurrence in patients with LUAD and can be used as a prognostic indicator for this group of patients. C1 [Qiu, Zijian] The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Department of Radiation OncologyQuzhou, Zhejiang, China. [Wu, Jiaji] The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Department of Respiratory MedicineYiwu, Zhejiang, China. [Pang, Guanchao] The Second Affiliated Hospital of Zhejiang University School of Medicine, Department of Respiratory and Critical Care MedicineHangzhou, Zhejiang, China. [Xu, Xia] The Second Affiliated Hospital of Zhejiang University School of Medicine, Department of PathologyHangzhou, Zhejiang, China. [Lin, Jun] The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Department of PathologyQuzhou, Zhejiang, China. [Wang, Pingli] The Second Affiliated Hospital of Zhejiang University School of Medicine, Department of Respiratory and Critical Care MedicineHangzhou, Zhejiang, China. RP Wang, P (reprint author), The Second Affiliated Hospital of Zhejiang University School of Medicine, Department of Respiratory and Critical Care Medicine, Hangzhou, China. EM pingliwang@zju.edu.cn CR Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2024, 68:394–424., DOI 10.3322/caac.21492 Qi J, Li M, Wang L, Hu Y, LiuW, Long Z, et al. National and subnational trends in cancer burden in China, 2005-20: an analysis of national mortality surveillance data. Lancet Public Health, 2023, 8(12):e943–e955., DOI 10.1016/S2468-2667(23, 00211-6 Zhang Y, Vaccarella S, Morgan E, Li M, Etxeberria J, Chokunonga E, et al. Global variations in lung cancer incidence by histological subtype in 2020: a population-based study. Lancet Oncol, 2023, 24(11):1206–18., DOI 10.1016/ S1470-2045(23)00444-8 Muraoka Y, Yotsukura M, Yoshida Y, Nakagawa K, Shiraishi K, Kohno T, et al. Dynamics of recurrence after curative resection of nonsmall cell lung cancer. J Surg Oncol, 2023, 128(7):1205–12., DOI 10.1002/jso.27395 Meira DD, de Castro ECMC, Casotti MC, Zetum ASS, Goncalves AFM, Moreira AR, et al. Prognostic factors and markers in non-small cell lung cancer: recent progress and future challenges. Genes, Basel,, 2023, 14(10):1906., DOI 10. 3390/genes14101906 Schegoleva AA, Khozyainova AA, Fedorov AA, Gerashchenko TS, Rodionov EO, Topolnitsky EB, et al. Prognosis of different types of non-small cell lung cancer progression: current state and perspectives. Cell Physiol Biochem, 2021, 55(S2): 29–48., DOI 10.33594/000000340 Weis SM, Cheresh DA. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med, 2011, 17(11):1359–70., DOI 10.1038/nm.2537 Nico B, Benagiano V, Mangieri D, Maruotti N, Vacca A, Ribatti D. Evaluation of microvascular density in tumors: pro and contra. Histol Histopathol, 2008, 23(5): 601–7., DOI 10.14670/HH-23.601 Marion-Audibert AM, Barel C, Gouysse G, Dumortier J, Pilleul F, Pourreyron C, et al. Low microvessel density is an unfavorable histoprognostic factor in pancreatic endocrine tumors. Gastroenterology, 2003, 125(4):1094–104., DOI 10. 1016/s0016-5085(03)01198-3 Meert AP, Paesmans M, Martin B, Delmotte P, Berghmans T, Verdebout JM, et al. The role of microvessel density on the survival of patients with lung cancer: a systematic review of the literature with meta-analysis. Br J Cancer, 2002, 87(7): 694–701., DOI 10.1038/sj.bjc.6600551 Yildiz E, Ayan S, Goze F, Gokce G, Gultekin EY. Relation of microvessel density with microvascular invasion, metastasis and prognosis in renal cell carcinoma. BJU Int, 2008, 101(6):758–64., DOI 10.1111/j.1464-410X.2007.07318.x Tanaka F, Otake Y, Yanagihara K, Kawano Y, Miyahara R, Li M, et al. Evaluation of angiogenesis in non-small cell lung cancer: comparison between anti- CD34 antibody and anti-CD105 antibody. Clin Cancer Res, 2001, 7(11):3410–5. Donnem T, Al-Saad S, Al-Shibli K, Delghandi MP, Persson M, Nilsen MN, et al. Inverse prognostic impact of angiogenic marker expression in tumor cells versus stromal cells in non small cell lung cancer. Clin Cancer Res, 2007, 13(22 Pt 1):6649–57., DOI 10.1158/1078-0432.CCR-07-0414 Kadota K, Huang CL, Liu D, Ueno M, Kushida Y, Haba R, et al. The clinical significance of lymphangiogenesis and angiogenesis in non-small cell lung cancer patients. Eur J Cancer, 2008, 44(7):1057–67., DOI 10.1016/j.ejca.2008.03.012 Bing Z, Jian-ru Y, Yao-quan J, Shi-feng C. Evaluation of angiogenesis in nonsmall cell lung carcinoma by CD34 immunohistochemistry. Cell Biochem Biophys, 2014, 70(1):327–31., DOI 10.1007/s12013-014-9916-5 Pomme G, Augustin F, FieglM, Droeser RA, Sterlacci W, Tzankov A. Detailed assessment of microvasculature markers in non-small cell lung cancer reveals potentially clinically relevant characteristics. Virchows Arch, 2015, 467(1):55–66., DOI 10.1007/s00428-015-1767-y Weidner N, Folkman J, Pozza F, Bevilacqua P, Allred EN, Moore DH, et al. Tumor angiogenesis: a new significant and independent prognostic indicator in early-stage breast carcinoma. J Natl Cancer Inst, 1992, 84(24):1875–87., DOI 10. 1093/jnci/84.24.1875 Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci, 2020, 77(9): 1745–70., DOI 10.1007/s00018-019-03351-7 MashreghiM, Azarpara H, Bazaz MR, Jafari A, Masoudifar A,Mirzaei H, et al. Angiogenesis biomarkers and their targeting ligands as potential targets for tumor angiogenesis. J Cell Physiol, 2018, 233(4):2949–65., DOI 10.1002/jcp.26049 Rakocevic J, Orlic D, Mitrovic-Ajtic O, Tomasevic M, Dobric M, Zlatic N, et al. Endothelial cell markers from clinician’s perspective. Exp Mol Pathol, 2017, 102(2):303–13., DOI 10.1016/j.yexmp.2017.02.005 Carlini MJ, Dalurzo MC, Lastiri JM, Smith DE, Vasallo BC, Puricelli LI, et al. Mast cell phenotypes andmicrovessels in non-small cell lung cancer and its prognostic significance. Hum Pathol, 2010, 41(5):697–705., DOI 10.1016/j.humpath.2009.04.029 Zhao YY, Xue C, Jiang W, Zhao HY, Huang Y, Feenstra K, et al. Predictive value of intratumoral microvascular density in patients with advanced non-small cell lung cancer receiving chemotherapy plus bevacizumab. J Thorac Oncol, 2012, 7(1):71–5., DOI 10.1097/JTO.0b013e31823085f4 YaoX,QianCN, ZhangZF, TanMH, Kort EJ,YangXJ, et al.Two distinct types of blood vessels in clear cell renal cell carcinoma have contrasting prognostic implications. Clin Cancer Res, 2007, 13(1):161–9., DOI 10.1158/1078-0432.CCR-06-0774 Wang HB, Qin Y, Yang JY. Research on the prognosis of different types of microvessels in bladder transitional cell carcinoma. World J Clin Cases, 2021, 9(25): 7381–90., DOI 10.12998/wjcc.v9.i25.7381 Sopo M, Anttila M, Muukkonen OT, YlA-Herttuala S, Kosma VM, Keski- Nisula L, et al. Microvessels in epithelial ovarian tumors: high microvessel density is a significant feature of malignant ovarian tumors. Anticancer Res, 2020, 40(12): 6923–31., DOI 10.21873/anticanres.14716 Yugawa K, Itoh S, Yoshizumi T, Iseda N, Tomiyama T, Toshima T, et al. Prognostic impact of tumor microvessels in intrahepatic cholangiocarcinoma: association with tumor-infiltrating lymphocytes. Mod Pathol, 2021, 34(4): 798–807., DOI 10.1038/s41379-020-00702-9 Murakami K, Kasajima A, Kawagishi N, Ohuchi N, Sasano H. Microvessel density in hepatocellular carcinoma: prognostic significance and review of the previous published work. Hepatol Res, 2015, 45(12):1185–94., DOI 10.1111/hepr. 12487 Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R, et al. Bevacizumab, Avastin®, in cancer treatment: a review of 15 years of clinical experience and future outlook. Cancer Treat Rev, 2020, 86:102017., DOI 10.1016/ j.ctrv.2020.102017 Liu D, Ding G. Predictive value of microvascular density for response to anlotinib in advanced NSCLC. Medicine, Baltimore,, 2022, 101(3):e28647., DOI 10. 1097/MD.0000000000028647 Zhang Q,Wu J, Bai X, Liang T. Evaluation of intra-tumoral vascularization in hepatocellular carcinomas. Front Med, Lausanne,, 2020, 7:584250., DOI 10.3389/ fmed.2020.584250 Adamo A, Bruno A,Menallo G, FrancipaneMG, FazzariM, Pirrone R, et al. Blood vessel detection algorithm for tissue engineering and quantitative histology. Ann Biomed Eng, 2022, 50(4):387–400., DOI 10.1007/s10439-022- 02923-2 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1611985 EP 1611994 DI 10.3389/pore.2025.1611985 PG 10 ER PT J AU Schwab, H Kerkhoff, M Plaumann, P Collaud, S Dirksen, U Theegarten, D Herold, Th Kalbourtzis, S Bolukbas, S Hegedus, B Hegedus, L AF Schwab, Hannah Kerkhoff, Maximilian Plaumann, Pauline Collaud, Stephane Dirksen, Uta Theegarten, Dirk Herold, Thomas Kalbourtzis, Stavros Bolukbas, Servet Hegedus, Balazs Hegedus, Luca TI Characterization of a novel sarcoma cell line with an EWSR1:: POU2AF3 fusion SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE sarcoma; chemotherapy; EWSR1; POU2AF3; patient derived cell line ID sarcoma; chemotherapy; EWSR1; POU2AF3; patient derived cell line AB Sarcomas with an EWSR1::POU2AF3(COLCA2) fusion are a very recently described entity of preferentially sinonasal origin and with undifferentiated round/spindle cell morphology. We established a novel cell line (PF1095) carrying a EWSR1::POU2AF3 fusion from the malignant pleural effusion of a 25-year-old sarcoma patient. The patient was first diagnosed with poorly differentiated neuroendocrine carcinoma based on tumor cell morphology and positivity to markers such as EMA, synaptophysin, and CD56. Later, the EWSR1 translocation was identified in the tumor cells with unknown partners and the patient received chemotherapy according to the Ewing 2008 protocol in combination with surgery and proton beam radiotherapy. At the time of cell line establishment, the disease progressed to pleural sarcomatosis with pleural effusion. In the cell line, we identified POU2AF3 as a fusion partner of EWSR1 and a TP53 frameshift deletion. Next, we determined the sensitivity of PF1095 cells to the currently approved chemotherapies in comparison to two conventional Ewing sarcoma lines (EW-7 and MHH-ES1) with the two most frequent EWSR::FLI1 fusions. Finally, we tested potential new combination therapies. We performed cell viability, proliferation, and cell cycle assays. We found that the proliferation rate of PF1095 cells was much slower than the EWSR1::FLI1 fusion lines and they also had a lower sensitivity to both irinotecan and doxorubicin treatment. Expression level of SLFN11, a predictor of sensitivity to DNA damaging agents, was also lower in PF1095 cells. Combination treatment with the PARP inhibitors olaparib and irinotecan or doxorubicin synergistically reduced cell viability and induced cell death and cell cycle arrest. This unique cell model provides an opportunity to test therapeutic approaches preclinically for this novel and aggressive sarcoma entity. C1 [Schwab, Hannah] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Kerkhoff, Maximilian] University Medicine Essen, West German Cancer Center, Pediatrics IIIEssen, Germany. [Plaumann, Pauline] University Medicine Essen, West German Cancer Center, Pediatrics IIIEssen, Germany. [Collaud, Stephane] University of Witten/Herdecke, Cologne Merheim Hospital, Department of Thoracic SurgeryCologne, Germany. [Dirksen, Uta] University Medicine Essen, West German Cancer Center, Pediatrics IIIEssen, Germany. [Theegarten, Dirk] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Herold, Thomas] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Kalbourtzis, Stavros] University of Duisburg-Essen, University Hospital of Essen, Institute of PathologyEssen, Germany. [Bolukbas, Servet] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Hegedus, Balazs] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. [Hegedus, Luca] University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic SurgeryEssen, Germany. RP Hegedus, B (reprint author), University Duisburg-Essen, University Medicine Essen–Ruhrlandklinik, Department of Thoracic Surgery, Essen, Germany. EM balazs.hegedues@rlk.uk-essen.de CR Cidre-Aranaz F, Watson S, Amatruda JF, Nakamura T, Delattre O, de Alava E, et al. Small round cell sarcomas. Nat Rev Dis Primers, 2022, 8(1):66., DOI 10.1038/ s41572-022-00393-3 Kallen ME, Hornick JL. From the ashes of “Ewing-like” sarcoma: a contemporary update of the classification, immunohistochemistry, and molecular genetics of round cell sarcomas. Semin Diagn Pathol, 2022, 39(1): 29–37., DOI 10.1053/j.semdp.2021.10.002 Grunewald TGP, Cidre-Aranaz F, Surdez D, Tomazou EM, de Alava E, Kovar H, et al. Ewing sarcoma. Nat Rev Dis Primers, 2018, 4(1):5., DOI 10.1038/s41572- 018-0003-x Sbaraglia M, Righi A, Gambarotti M, Dei Tos AP. Ewing sarcoma and Ewinglike tumors. Virchows Arch, 2020, 476(1):109–19., DOI 10.1007/s00428-019-02720-8 Sankar S, Lessnick SL. Promiscuous partnerships in Ewing’s sarcoma. Cancer Genet, 2011, 204(7):351–65., DOI 10.1016/j.cancergen.2011.07.008 Tu J, Huo Z, Gingold J, Zhao R, Shen J, Lee DF. The histogenesis of ewing sarcoma. Cancer Rep Rev, 2017, 1(2)., DOI 10.15761/CRR.1000111 Sand LG, Szuhai K, Hogendoorn PC. Sequencing overview of ewing sarcoma: a journey across genomic, epigenomic and transcriptomic landscapes. Int J Mol Sci, 2015, 16(7):16176–215., DOI 10. 3390/ijms160716176 Orth MF, Surdez D, Faehling T, Ehlers AC, Marchetto A, Grossetete S, et al. Systematic multi-omics cell line profiling uncovers principles of Ewing sarcoma fusion oncogene-mediated gene regulation. Cell Rep, 2022, 41(10):111761., DOI 10. 1016/j.celrep.2022.111761 Wang WL, Lazar AJ. Undifferentiated “round” cell, “Ewing-like”, sarcoma: not always so-round nor Ewing-like. Virchows Arch, 2017, 470(4):371–2., DOI 10.1007/ s00428-017-2080-8 Kailayangiri S, Altvater B, Meltzer J, Pscherer S, Luecke A, Dierkes C, et al. The ganglioside antigen G(D2, is surface-expressed in Ewing sarcoma and allows for MHC-independent immune targeting. Br J Cancer, 2012, 106(6):1123–33., DOI 10. 1038/bjc.2012.57 Zollner SK, Amatruda JF, Bauer S, Collaud S, de Alava E, DuBois SG, et al. Ewing sarcoma-diagnosis, treatment, clinical challenges and future perspectives. J Clin Med, 2021, 10(8):1685., DOI 10.3390/jcm10081685 Tirode F, Surdez D,Ma X, Parker M, Le Deley MC, Bahrami A, et al. Genomic landscape of Ewing sarcoma defines an aggressive subtype with co-association of STAG2 and TP53 mutations. Cancer Discov, 2014, 4(11):1342–53., DOI 10.1158/ 2159-8290.CD-14-0622 Davicioni E, Wai DH, Anderson MJ. Diagnostic and prognostic sarcoma signatures. Mol Diagn Ther, 2008, 12(6):359–74., DOI 10.1007/BF03256302 Yoshida A. Ewing and Ewing-like sarcomas: a morphological guide through genetically-defined entities. Pathol Int, 2023, 73(1):12–26., DOI 10.1111/pin.13293 Sbaraglia M, Bellan E, Dei Tos AP. The 2020 WHO classification of soft tissue tumours: news and perspectives. Pathologica, 2021, 113(2):70–84., DOI 10.32074/ 1591-951X-213 Hiemenz MC, Kaur J, Kuang Z, Huang RSP, Harries L, Metzger D, et al. POU2AF3-rearranged sarcomas: a novel tumor defined by fusions of EWSR1 or FUS to a gene formerly designated COLCA2. Genes Chromosomes Cancer, 2023, 62(8):460–70., DOI 10.1002/gcc.23136 Peltekova VD, Lemire M, Qazi AM, Zaidi SH, Trinh QM, Bielecki R, et al. Identification of genes expressed by immune cells of the colon that are regulated by colorectal cancer-associated variants. Int J Cancer, 2014, 134(10):2330–41., DOI 10. 1002/ijc.28557 Hitomi Y, Aiba Y, Kawai Y, Kojima K, Ueno K, Nishida N, et al. rs1944919 on chromosome 11q23.1 and its effector genes COLCA1/COLCA2 confer susceptibility to primary biliary cholangitis. Sci Rep, 2021, 11(1):4557., DOI 10. 1038/s41598-021-84042-x Wu XS, He XY, Ipsaro JJ, Huang YH, Preall JB, Ng D, et al. OCA-T1 and OCA-T2 are coactivators of POU2F3 in the tuft cell lineage. Nature, 2022, 607(7917):169–75., DOI 10.1038/s41586-022-04842-7 Yamaguchi S, Yamazaki Y, Ishikawa Y, Kawaguchi N, Mukai H, Nakamura T. EWSR1 is fused to POU5F1 in a bone tumor with translocation t(6;22)(p21;q12). Genes Chromosomes Cancer, 2005, 43(2):217–22., DOI 10.1002/gcc.20171 Koshyk O, Dehner CA, van den Hout M, Bempt IV, Sciot R, Huang HY, et al. EWSR1::POU2AF3(COLCA2, sarcoma: an aggressive, polyphenotypic sarcoma with a head and neck predilection. Mod Pathol, 2023, 36(12):100337., DOI 10. 1016/j.modpat.2023.100337 Agaimy A, Baneckova M, De Almeida J, Dickson BC, Dimmler A, Hartmann W, et al. Recurrent EWSR1::COLCA2 fusions define a novel sarcoma with spindle/ round cell morphology and strong predilection for the sinonasal tract. Am J Surg Pathol, 2023, 47(3):361–9., DOI 10.1097/PAS.0000000000002000 Stewart E, Goshorn R, Bradley C, Griffiths LM, Benavente C, Twarog NR, et al. Targeting the DNA repair pathway in Ewing sarcoma. Cell Rep, 2014, 9(3):829–41., DOI 10.1016/j.celrep.2014.09.028 Flores G, Grohar PJ. One oncogene, several vulnerabilities: EWS/FLI targeted therapies for Ewing sarcoma. J Bone Oncol, 2021, 31:100404., DOI 10.1016/j.jbo. 2021.100404 Renzi S, Anderson ND, Light N, Gupta A. Ewing-like sarcoma: an emerging family of round cell sarcomas. J Cell Physiol, 2019, 234(6):7999–8007., DOI 10.1002/jcp.27558 Mangoni M, Sottili M, Salvatore G, Campanacci D, Scoccianti G, Beltrami G, et al. Soft tissue sarcomas: new opportunity of treatment with PARP inhibitors? Radiol Med, 2019, 124(4):282–9., DOI 10.1007/s11547-018-0877-4 Brenner JC, Feng FY, Han S, Patel S, Goyal SV, Bou-Maroun LM, et al. PARP- 1 inhibition as a targeted strategy to treat Ewing’s sarcoma. Cancer Res, 2012, 72(7): 1608–13., DOI 10.1158/0008-5472.CAN-11-3648 Zoppoli G, Regairaz M, Leo E, Reinhold WC, Varma S, Ballestrero A, et al. Putative DNA/RNA helicase Schlafen-11, SLFN11, sensitizes cancer cells to DNAdamaging agents. Proc Natl Acad Sci U S A., 2012, 109(37):15030–5., DOI 10.1073/ pnas.1205943109 Tang SW, Bilke S, Cao L, Murai J, Sousa FG, Yamade M, et al. SLFN11 is a transcriptional target of EWS-FLI1 and a determinant of drug response in ewing sarcoma. Clin Cancer Res, 2015, 21(18):4184–93., DOI 10.1158/1078-0432.CCR-14- 2112 Gartrell J, Mellado-Largarde M, Clay MR, Bahrami A, Sahr NA, Sykes A, et al. SLFN11 is widely expressed in pediatric sarcoma and induces variable sensitization to replicative stress caused by DNA-damaging agents. Mol Cancer Ther, 2021, 20(11):2151–65., DOI 10.1158/1535-7163.MCT-21-0089 Choy E, Butrynski JE, Harmon DC, Morgan JA, George S, Wagner AJ, et al. Phase II study of olaparib in patients with refractory Ewing sarcoma following failure of standard chemotherapy. BMC Cancer, 2014, 14:813., DOI 10.1186/1471- 2407-14-813 Morland B, Platt K, Whelan JS. A phase II window study of irinotecan, CPT- 11, in high risk Ewing sarcoma: a Euro-E.W.I.N.G. study. Pediatr Blood Cancer, 2014, 61(3):442–5., DOI 10.1002/pbc.24767 Federico SM, Pappo AS, Sahr N, Sykes A, Campagne O, Stewart CF, et al. A phase I trial of talazoparib and irinotecan with and without temozolomide in children and young adults with recurrent or refractory solid malignancies. Eur J Cancer, 2020, 137:204–13., DOI 10.1016/j.ejca.2020. 06.014 Schiavo R, Tullio C, La Grotteria M, Andreotti IC, Scarpati B, Romiti L, et al. Establishment and characterization of a new Ewing’s sarcoma cell line from a malignant pleural effusion. Anticancer Res, 2007, 27(5A):3273–8. Hegedus L, Okumus O, Mairinger F, Ploenes T, Reuter S, Schuler M, et al. TROP2 expression and SN38 antitumor activity inmalignant pleural mesothelioma cells provide a rationale for antibody-drug conjugate therapy. Lung Cancer, 2023, 178:237–46., DOI 10.1016/j.lungcan.2023.03.003 Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res, 2010, 70(2):440–6., DOI 10.1158/0008-5472. CAN-09-1947 Li JY, Jing R, Wei H,Wang M, Xiaowei Q, Liu H, et al. Germline mutations in 40 cancer susceptibility genes among Chinese patients with high hereditary risk breast cancer. Int J Cancer, 2019, 144(2):281–9., DOI 10.1002/ijc.31601 Nazha B, Inal C, Owonikoko TK. Disialoganglioside GD2 expression in solid tumors and role as a target for cancer therapy. Front Oncol, 2020, 10:1000., DOI 10. 3389/fonc.2020.01000 Chan GC, Chan CM. Anti-GD2 directed immunotherapy for high-risk and metastatic neuroblastoma. Biomolecules, 2022, 12(3):358., DOI 10.3390/ biom12030358 Evdokimova V, Gassmann H, Radvanyi L, Burdach SEG. Current state of immunotherapy and mechanisms of immune evasion in ewing sarcoma and osteosarcoma. Cancers, Basel,, 2022, 15(1):272., DOI 10.3390/cancers15010272 Chen AP, Sharon E, O’Sullivan-Coyne G, Moore N, Foster JC, Hu JS, et al. Atezolizumab for advanced alveolar soft Part Sarcoma. N Engl J Med, 2023, 389(10):911–21., DOI 10.1056/NEJMoa2303383 Chandhanayingyong C, Kim Y, Staples JR, Hahn C, Lee FY. MAPK/ERK signaling in osteosarcomas, ewing sarcomas and chondrosarcomas: therapeutic implications and future directions. Sarcoma, 2012, 2012:404810., DOI 10.1155/2012/ 404810 McAloney CA, Makkawi R, Budhathoki Y, Cannon MV, Franz EM, Gross AC, et al. Host-derived growth factors drive ERK phosphorylation and MCL1 expression to promote osteosarcoma cell survival during metastatic lung colonization. Cell Oncol, Dordr,, 2024, 47(1):259–82., DOI 10.1007/s13402-023- 00867-w Gardner EE, Lok BH, Schneeberger VE, Desmeules P, Miles LA, Arnold PK, et al. Chemosensitive relapse in small cell lung cancer proceeds through an EZH2-SLFN11 Axis. Cancer Cell, 2017, 31(2):286–99., DOI 10.1016/j.ccell.2017. 01.006 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1611986 EP 1611997 DI 10.3389/pore.2025.1611986 PG 12 ER PT J AU Lippai, Z Papp, G Sapi, J Dezso, K Sapi, Z AF Lippai, Zoltan Papp, Gergo Sapi, Johanna Dezso, Katalin Sapi, Zoltan TI NTRK amplification occurs frequently in pan-TRK immunopositive dedifferentiated liposarcomas SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE soft tissue sarcoma; dedifferentiated liposarcoma; gene rearrangement; gene amplification; NTRK ID soft tissue sarcoma; dedifferentiated liposarcoma; gene rearrangement; gene amplification; NTRK AB The neurotrophic tyrosine kinase receptor (NTRK) gene family is of rising importance as their fusions are oncogenic, and specific target drugs are available to inhibit the chimera proteins. Pan-TRK antibody, which shows the overexpression of the NTRK1-2-3 genes, is a useful tool to detect tumors with or without NTRK gene alterations, due to high negative predictive value. Though it is well known that pan-TRK immunopositivity is usually not connected to NTRK fusion, the role of other possible genetic alterations is under-researched. In our previous work, we found 3 NTRK1 amplified cases out of 6 cases with recurrent NTRK1 tyrosine kinase domain mutation pair, so we extended our investigation to a larger series to estimate amplification frequency. Pan-TRK immunopositivity was seen in 76 of the 132 dedifferentiated liposarcomas cases, followed by NTRK1-2-3 break-apart FISH tests in 76 pan-TRK positive cases to detect oncogenic fusions or other copy number alterations of these genes. None of the pan-TRK immunopositive dedifferentiated liposarcomas showed absolutely certain sign of fusion, however, 18 (28%) cases showed amplification of one of the genes, 13 had polysomy, 34 were normal, 11 were not evaluable. The extent of pan-TRK immunoreaction showed a positive correlation (p = 0.002) with the NTRK status found by FISH. Analyzing publicly available data from large series of 265 liposarcoma samples consisting of both welldifferentiated and dedifferentiated liposarcoma case, 23 (8.6%) cases showed a mutual exclusive amplification of the NTRK genomic loci in a nonpreselected, independent patient population indicating that our findings are presented in other cohorts. Our results underline the so far not revealed frequent occurrence of NTRK amplifications which might be important in the TRK inhibition therapy. C1 [Lippai, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Papp, Gergo] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sapi, Johanna] Obuda University, Physiological Controls Research Center, John von Neumann Faculty of InformaticsBudapest, Hungary. [Dezso, Katalin] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Sapi, Zoltan] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. RP Sapi, Z (reprint author), Semmelweis University, 1st Department of Pathology and Experimental Cancer Research, Budapest, Hungary. EM sapi.zoltan.dr@gmail.com CR Doebele RC, Davis LE, Vaishnavi A, Le AT, Estrada-Bernal A, Keysar S, et al. An oncogenic NTRK fusion in a patient with soft-tissue sarcoma with response to the tropomyosin-related kinase inhibitor LOXO-101. Cancer Discov, 2015, 5(10): 1049–57., DOI 10.1158/2159-8290.CD-15-0443 Farago AF, Le LP, Zheng Z, Muzikansky A, Drilon A, Patel M, et al. Durable clinical response to entrectinib in NTRK1-rearranged non-small cell lung cancer. J Thorac Oncol, 2015, 10(12):1670–4., DOI 10.1097/01.JTO. 0000473485.38553.f0 Drilon A, Ou SHI, Cho BC, Kim DW, Lee J, Lin JJ, et al. Repotrectinib, TPX- 0005, is a next-generation ROS1/TRK/ALK inhibitor that potently inhibits ROS1/ TRK/ALK solvent- front mutations. Cancer Discov, 2018, 8(10):1227–36., DOI 10. 1158/2159-8290.CD-18-0484 Drilon A, Nagasubramanian R, Blake JF, Ku N, Tuch BB, Ebata K, et al. A nextgeneration TRK kinase inhibitor overcomes acquired resistance to prior TRK kinase inhibition in patients with TRK fusion-positive solid tumors. Cancer Discov, 2017, 7(9):963–72., DOI 10.1158/2159-8290.CD-17-0507 Lippai Z, Sapi Z. Diagnostic approach of tumors with neurotrophic tropomyosin receptor tyrosine kinase gene fusions. Orv Hetil, 2020, 161(41): 1753–63., DOI 10.1556/650.2020.31846 Vaishnavi A, Le AT, Doebele RC. TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov, 2015, 5(1):25–34., DOI 10.1158/2159-8290.CD- 14-0765 Solomon JP, Benayed R, Hechtman JF, Ladanyi M. Identifying patients with NTRK fusion cancer. Ann Oncol, 2019, 30(Suppl. 8):viii16–viii22., DOI 10.1093/ annonc/mdz384 Davis JL, Lockwood CM, Albert CM, Tsuchiya K, Hawkins DS, Rudzinski ER. Infantile NTRK-associated mesenchymal tumors. Pediatr Dev Pathol, 2018, 21(1): 68–78., DOI 10.1177/1093526617712639 Zhao M, Yin M, Kuick CH, Chen H, Aw SJ, Merchant K, et al. Congenital mesoblastic nephroma is characterised by kinase mutations including EGFR internal tandem duplications, the ETV6-NTRK3 fusion, and the rare KLHL7- BRAF fusion. Histopathology, 2020, 77(4):611–21., DOI 10.1111/his.14194 Harada G, Santini FC, Wilhelm C, Drilon A. NTRK fusions in lung cancer: from biology to therapy. Lung Cancer, 2021, 161:108–13., DOI 10.1016/j.lungcan. 2021.09.005 Ratti M, Grizzi G, Passalacqua R, Lampis A, Cereatti F, Grassia R, et al. NTRK fusions in colorectal cancer: clinical meaning and future perspective. Expert Opin Ther Targets, 2021, 25(8):677–83., DOI 10.1080/14728222.2021.1978070 Forschner A, Forchhammer S, Bonzheim I. NTRK gene fusions in melanoma: detection, prevalence and potential therapeutic implications. J Dtsch Dermatol Ges, 2020, 18(12):1387–92., DOI 10.1111/ddg.14160 Joshi SK, Qian K, Bisson WH, Watanabe-Smith K, Huang A, Bottomly D, et al. Discovery and characterization of targetable NTRK point mutations in hematologic neoplasms. Blood, 2020, 135(24):2159–70., DOI 10.1182/blood. 2019003691 Iacobucci I, Wen J, Meggendorfer M, Choi JK, Shi L, Pounds SB, et al. Genomic subtyping and therapeutic targeting of acute erythroleukemia. Nat Genet, 2019, 51(4):694–704., DOI 10.1038/s41588-019-0375-1 Reuther GW, Lambert QT, Caligiuri MA, Der CJ. Identification and characterization of an activating TrkA deletion mutation in acute myeloid leukemia. Mol Cell Biol, 2000, 20(23):8655–66., DOI 10.1128/mcb.20.23.8655- 8666.2000 Tacconelli A, Farina AR, Cappabianca L, Desantis G, Tessitore A, Vetuschi A, et al. TrkA alternative splicing: a regulated tumor-promoting switch in human neuroblastoma. Cancer Cell, 2004, 6(4):347–60., DOI 10.1016/j.ccr.2004.09.011 Stravodimou A, Voutsadakis IA. Neurotrophic receptor tyrosine kinase family members in secretory and non-secretory breast carcinomas. World J Clin Oncol, 2022, 13(2):135–46., DOI 10.5306/wjco.v13.i2.135 Okamura R, Boichard A, Kato S, Sicklick JK, Bazhenova L, Kurzrock R. Analysis of NTRK alterations in pan-cancer adult and pediatric malignancies: implications for NTRK-targeted therapeutics. JCO Precis Oncol, 2018, 2018:1–20., DOI 10.1200/PO.18.00183 Hsiao SJ, Zehir A, Sireci AN, Aisner DL. Detection of tumor NTRK gene fusions to identify patients who may benefit from tyrosine kinase, TRK, inhibitor therapy. J Mol Diagn, 2019, 21(4):553–71., DOI 10.1016/j.jmoldx.2019.03.008 Kiss E, Papai Z. Novel targeted therapeutic option in oncology: tropomyosin receptor tyrosine kinase inhibitors. Orv Hetil, 2021, 162(34):1362–9., DOI 10.1556/ 650.2021.32183 Rudzinski ER, Lockwood CM, Stohr BA, Vargas SO, Sheridan R, Black JO, et al. Pan-trk immunohistochemistry identifies NTRK rearrangements in pediatric mesenchymal tumors. Am J Surg Pathol, 2018, 42(7):927–35., DOI 10.1097/PAS. 0000000000001062 Chiang S, Cotzia P, Hyman DM, Drilon A, Tap WD, Zhang L, et al. NTRK fusions define a novel uterine sarcoma subtype with features of fibrosarcoma. Am J Surg Pathol, 2018, 42(6):791–8., DOI 10.1097/PAS.0000000000001055 Hung YP, Fletcher CDM, Hornick JL. Evaluation of pan-TRK immunohistochemistry in infantile fibrosarcoma, lipofibromatosis-like neural tumour and histological mimics. Histopathology, 2018, 73(4):634–44., DOI 10. 1111/his.13666 Lippai Z, Peterfia B, Papp G, Dezso K, Bedics G, Papai Z, et al. A recurrent NTRK1 tyrosine kinase domain mutation pair is characteristic in a subset of dedifferentiated liposarcomas. Eur J Cancer, 2024, 202:114005., DOI 10.1016/j.ejca. 2024.114005 Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data, vol 2, pg 401, 2012). Cancer Discov, 2012, 2(10):960. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal, 2013, 6(269):pl1., DOI 10.1126/scisignal.2004088 Lazar AJ, Cancer Genome Atlas Research Network. Comprehensive and integrated genomic characterization of adult soft tissue sarcomas. Cell, 2017, 171(4):950–65.e28., DOI 10.1016/j.cell.2017.10.014 Nacev BA, Sanchez-Vega F, Smith SA, Antonescu CR, Rosenbaum E, Shi H, et al. Clinical sequencing of soft tissue and bone sarcomas delineates diverse genomic landscapes and potential therapeutic targets. Nat Commun, 2022, 13(1):3405., DOI 10.1038/s41467-022-30453-x Barretina J, Taylor BS, Banerji S, Ramos AH, Lagos-Quintana M, Decarolis PL, et al. Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nat Genet, 2010, 42(8):715–21., DOI 10.1038/ng.619 Harwell MR. Summarizing monte-carlo results in methodological research. J Educ Stat, 1992, 17(4):297–313., DOI 10.2307/1165126 Lix LM, Keselman JC, Keselman HJ. Consequences of assumption violations revisited: a quantitative review of alternatives to the one-way analysis of variance F test. Rev Educ Res, 1996, 66(4):579–619., DOI 10.2307/1170654 Szuhai K, Ijszenga M, de Jong D, Karseladze A, Tanke HJ, Hogendoorn PCW. The NFATc2 gene is involved in a novel cloned translocation in a Ewing sarcoma variant that couples its function in immunology to oncology. Clin Cancer Res, 2009, 15(7):2259–68., DOI 10.1158/1078-0432.CCR-08-2184 Brahmi M, Dufresne A, Verret B, Tirode F, Blay JY. NTRK fusion in soft tissue sarcomas harboring MDM2/CDK4 amplification: three case reports. Ann Oncol, 2021, 32(6):813–4., DOI 10.1016/j.annonc.2021.02.019 Xu Y, Shi X, Wang W, Zhang L, Cheung S, Rudolph M, et al. Prevalence and clinico-genomic characteristics of patients with TRK fusion cancer in China. NPJ Precis Oncol, 2023, 7(1):75., DOI 10.1038/s41698-023-00427-3 Suurmeijer AJ, Dickson BC, Swanson D, Zhang L, Sung YS, Huang HY, et al. The histologic spectrum of soft tissue spindle cell tumors with NTRK3 gene rearrangements. Genes Chromosomes Cancer, 2019, 58(11):739–46., DOI 10.1002/ gcc.22767 Vargas AC, Joy C, Maclean FM, Bonar F, Wong DD, Gill AJ, et al. Kinase expression in angiomatoid fibrous histiocytoma: panTRK is commonly expressed in the absence of NTRK rearrangement. J Clin Pathol, 2024, 77(4):251–4., DOI 10.1136/ jcp-2023-209225 Rizzato M, Brignola S, Munari G, Gatti M, Dadduzio V, Borga C, et al. Prognostic impact of FGFR2/3 alterations in patients with biliary tract cancers receiving systemic chemotherapy: the BITCOIN study. Eur J Cancer, 2022, 166: 165–75., DOI 10.1016/j.ejca.2022.02.013 Li D, Liu J, Zhang X, Han J, Jin H, Wang L, et al. Combined lorlatinib, dabrafenib, and trametinib treatment for ROS1-rearranged advanced non-smallcell lung cancer with a lorlatinib-induced braf V600E mutation: a case report. Cancer Manag Res, 2022, 14:3175–9., DOI 10.2147/CMAR.S387211 Lee SJ, Kim NK, Lee SH, Kim ST, Park SH, Park JO, et al. Gene amplification in patients with metastatic cancer. Precision Future Med, 2017, 1(3):129–37. Xu AD, Fu Y, Pu XH, Wu HY, Sun Q, Fan XS. Clinicopathological features of gastric carcinomas with NTRK-rearrangement/amplification: report of four cases. Zhonghua Bing Li Xue Za Zhi, 2023, 52(5):454–9., DOI 10.3760/cma.j.cn112151- 20221008-00840 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1611993 EP 1612001 DI 10.3389/pore.2024.1611993 PG 9 ER PT J TI Evaluating the correlation between pretreatment 18F-FDG PET/CT metabolic parameters and tumor-infiltrating lymphocyte levels in nonluminal breast cancer and impact on survival SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE tumor-infiltrating lymphocytes; nonluminal breast cancer; tumor heterogeneity indices; FDG; metabolic parameter ID tumor-infiltrating lymphocytes; nonluminal breast cancer; tumor heterogeneity indices; FDG; metabolic parameter AB Background and Objectives: This study aims to evaluate the correlation between Tumor-Infiltrating Lymphocyte (TIL) levels and Fluorine-18 fluorodeoxyglucose (18F-FDG) metabolic parameters, including spleen and bone marrow FDG uptake and tumor heterogeneity in non-luminal breast cancers (NLBC), and to elucidate their association with survival outcomes. Methods: We retrospectively analyzed data from 100 females with stage 2–4 NLBC who underwent pretreatment 18F-FDG Positron emission tomography-computed tomography (PET/CT). TIL was scored based on Hematoxylin-Eosin-stained specimens and 18F-FDG PET metabolic parameters, including maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), metabolic tumor volume (MTV), total lesion glycolysis (TLG), liver, spleen, and bone marrow FDG uptake were calculated. Heterogeneity Index (HI)1, HI2, and HI3 indices were analyzed with FDG metabolic parameters. The association between these factors and overall survival was analyzed using multivariate Cox regression models. Results: TIL showed weak negative correlations with tumor size, tumor (T), and metastasis (M) stages. No significant correlation was found between TIL levels and overall SUV values. However, in stage 4, TIL correlated positively with liver, spleen, and bone marrow SUV values and negatively with heterogeneity indices (HI2, HI3). Higher tumor size, HI values, and Bone marrow-to-liver ratio (BLR) SUVmean were associated with increased mortality. A TIL cut-off value of <5 was linked to significantly worse survival. Conclusion: Our study demonstrates a strong connection between TIL, FDG metabolic parameters, and tumor heterogeneity, particularly in advanced NLBC. Although TIL is not generally associated with SUV values, its association with certain metabolic and heterogeneity indices suggests that it is important in influencing survival. Further research involving larger cohorts and diverse breast cancer subtypes is needed to validate these results. CR Denkert C, Minckwitz G, Darb-Esfahani S, Lederer B, Heppner B, Weber K, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol, 2018, 19:40–50., DOI 10.1016/S1470-2045(17)30904-X Goldhirsch A, Wood WC, Coates AS, Thurlimann B, Senn H-J, Senn HJ, et al. Strategies for subtypes – dealing with the diversity of breast cancer: highlights of the St. Gallen international expert consensus on the primary therapy of early breast cancer. Ann Oncol, 2011, 22:1736–47., DOI 10.1093/annonc/mdr304 Salgado R, Denkert C, Demaria NS, Sirtaine N, Klauschen F, Pruneri G, et al. The evaluation of tumor-infiltrating lymphocytes, TILS, in breast cancer: recommendations by an International TILS Working Group 2014. Ann Oncol, 2015, 26:259–71., DOI 10.1093/annonc/mdu450 Ciarka A, Piatek M, Peksa R, Kunc M, Senkus E. Tumor-infiltrating lymphocytes, TILs, in breast cancer: prognostic and predictive significance across molecular subtypes. Biomedicines, 2024, 12(4):763., DOI 10.3390/ biomedicines12040763 Yu X, Zhang Z, Wang Z, Wu P, Qiu F, Huang J. Prognostic and predictive value of tumor-infiltrating lymphocytes in breast cancer: a systematic review and metaanalysis. Clin Transl Oncol, 2016, 18(5):497–506., DOI 10.1007/s12094-015-1391-y Althobiti M, Aleskandarany MA, Joseph C, Toss M, Mongan N, Diez- Rodriguez M, et al. Heterogeneity of tumor-infiltrating lymphocytes in breast cancer and its prognostic significance. Histopathology, 2018, 73(6):887–96., DOI 10.1111/his.13695 Saber MM, El Zawahry HM, Hilal AM, Abou-Bakr A, Namour AE, Sabere MM, et al. Prognostic value of tumor infiltrating lymphocytes in locally advanced HER2 enriched breast cancer. Asian Pac J Cancer Prev, 2022, 23(2):553–60., DOI 10.31557/APJCP.2022.23.2.553 MurakamiW, Tozaki M, Sasaki M, Hida A, Ohi Y, Kubota K, et al. Correlation between 18F-FDG uptake on PET/MRI and the level of tumor-infiltrating lymphocytes, TILs, in triple-negative and HER2-positive breast cancer. Eur J Radiol, 2020, 123:108773., DOI 10.1016/j.ejrad.2019.108773 de Moraes FCA, Souza MEC, Sano VKT, Moraes RA, Melo AC. Association of tumor-infiltrating lymphocytes with clinical outcomes in patients with triplenegative breast cancer receiving neoadjuvant chemotherapy: a systematic review and meta-analysis. Clin Transl Oncol, 2024))., DOI 10.1007/s12094-024-03661-8 Kitajima K, Higuchi T, Fujimoto Y, Ishikawa E, Yokoyama H, Komoto H, et al. Relationship between FDG-PET and the immune microenvironment in breast cancer. Eur J Radiol, 2023, 158:110661., DOI 10.1016/j.ejrad.2022.110661 Seban RD, Rouzier R, Latouche A, Deleval N, Guinebretiere JM, Buvatet I, et al. Total metabolic tumor volume and spleen metabolism on baseline [18F]-FDG PET/CT as independent prognostic biomarkers of recurrence in resected breast cancer. Eur J Nucl Med Mol Imaging, 2021, 48(11):3560–70., DOI 10.1007/s00259- 021-05322-2 Park S, Min EK, Bae SJ, Cha C, Kim D, Lee J, et al. Relationship of the standard uptake value of, 18)F-FDG-PET-CT with tumor-infiltrating lymphocytes in breast tumors measuring 1 cm. Sci Rep, 2021, 11(1):12046., DOI 10.1038/s41598-021- 91404-y Lee JW, Kim SY, Han SW, Lee JE, Lee HJ, Heo NH, et al. [18F]FDG uptake of bone marrow on PET/CT for predicting distant recurrence in breast cancer patients after surgical resection. EJNMMI Res, 2020, 10(1):72., DOI 10.1186/s13550-020- 00660-y Wang Y, Li Y, Jiang H, Zuo C, Xu W. Elevated splenic 18F-fluorodeoxyglucose positron emission tomography/computed tomography activity is associated with 5- year risk of recurrence in non-metastatic invasive ductal carcinoma of the breast. Br J Radiol, 2024, 97(1153):237–48., DOI 10.1093/bjr/tqad015 Bang JI, Yoon HJ, Kim BS. Clinical utility of FDG uptake within reticuloendothelial system on F-18 FDG PET/CT for prediction of tumor recurrence in breast cancer. PLoS One, 2018, 13(12):e0208861., DOI 10.1371/ journal.pone.0208861 Liu G, Yin H, Cheng X, Wang Y, Hu Y, Liu T, et al. Intra-tumor metabolic heterogeneity of gastric cancer on 18F-FDG PETCT indicates patient survival outcomes. Clin Exp Med, 2021, 21:129–38., DOI 10.1007/s10238-020-00659-8 Huang B, Chan T, Kwong DL-W, Chan WKS, Khong PL. Nasopharyngeal carcinoma: investigation of intratumoral heterogeneity with FDG PET/CT. Am J Roentgenol, 2012, 199:169–74., DOI 10.2214/AJR.11.7336 Babacan GB, Tamam MO, Saracoglu S, Acar Tayyar MN, Sahin MC, Kulduk G, et al. Can tumor heterogeneity in TNBC obtained with FDG PET/CT predict survival? Novel method: heterogeneity index 3. Eur J Nucl Med Mol Imaging, 2023, 50(Suppl. 1):S478–S898., DOI 10.1007/s00259-023-06333-x Sasada S, Kimura Y, Emi A, Masumoto N, Kadoya T, Arihiro K, et al. Tumorinfiltrating lymphocyte score based on FDG PET/CT for predicting the effect of neoadjuvant chemotherapy in breast cancer. Anticancer Res, 2020, 40(6):3395–400., DOI 10.21873/anticanres.14323 Giuliano AE, Connolly JL, Edge SB, Mittendorf EA, Rugo HS, Solin LJ, et al. Breast Cancer-Major changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J Clin, 2017, 67:290–303., DOI 10.3322/ caac.21393 Wolff AC, Hammond MEH, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol, 2007, 25(1):118–45., DOI 10.1200/JCO.2006.09.2775 Frankowska K, Zarobkiewicz M, Dabrowska I, Bojarska-Junak A. Tumor infiltrating lymphocytes and radiological picture of the tumor. Med Oncol, 2023, 40(6):176., DOI 10.1007/s12032-023-02036-3 Sasada S, Shiroma N, Goda N, Kajitani K, Emi A, Masumoto N, et al. The relationship between ring-type dedicated breast PET and immune microenvironment in early breast cancer. Breast Cancer Res Treat, 2019, 177: 651–7., DOI 10.1007/s10549-019-05339-0 Hirakata T, Fujii T, Kurozumi S, Katayama A, Honda C, Yanai K, et al. FDG uptake reflects breast cancer immunological features:the PD-L1 expression and degree of TILs in primary breast cancer. Breast Cancer Res Treat, 2020, 181:331–8., DOI 10.1007/s10549-020-05619-0 Kajary K, Lengyel Z, Tokes A-M, Kulka J, Dank M, Tokes T. Dynamic FDGPET/ CT in the initial staging of primary breast cancer: clinicopathological correlations. Pathol Oncol Res, 2020, 26:997–1006., DOI 10.1007/s12253-019- 00641-0 Kim Y, Kim YJ, Paeng JC, Cheon GJ, Lee DS, Chung JK, et al. Heterogeneity index evaluated by slope of linear regression on 18F-FDG PET/CT as a prognostic marker for predicting tumor recurrence in pancreatic ductal adenocarcinoma. Eur J Nucl Med Mol Imaging, 2017, 44: 1995–2003., DOI 10.1007/s00259-017-3755-8 Li R, Lin J, Wang L, Zheng X, Tang K. The association between 18Ffluorodeoxyglucose PET intratumoral metabolic heterogeneity and pathological parameters in non-small cell lung cancer. Nucl Med Commun, 2019, 40:1022–8., DOI 10.1097/MNM.0000000000001058 Park HS, Heo I, Kim JY, Kim S, Nam S, Park S, et al. No effect of tumorinfiltrating lymphocytes, TILs, on prognosis in patients with early triplenegative breast cancer: validation of recommendations by the International TILs Working Group 2014. J Surg Oncol, 2016, 114(1):17–21., DOI 10.1002/ jso.24275 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1612014 EP 1612027 DI 10.3389/pore.2024.1612014 PG 14 ER PT J AU Zecevic, A Blanka-Protic, A Jandric, A Adzic-Vukicevic, T AF Zecevic, Andrej Blanka-Protic, Ana Jandric, Aleksandar Adzic-Vukicevic, Tatjana TI Are patients with chronic obstructive pulmonary disease at a greater risk for the development of autoimmune thyroiditis as an adverse event of immunotherapy in non-small cell lung cancer treatment? SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE chronic obstructive lung disease; non-small cell lung cancer; immunotherapy; thyroiditis; adverse event ID chronic obstructive lung disease; non-small cell lung cancer; immunotherapy; thyroiditis; adverse event AB Introduction: Immunotherapy has made a significant improvement in the treatment of patients with non-small cell lung cancer (NSCLC). It has a role in boosting the immune system, so it can fight cancer cells. Sometimes, this mechanism can lead to an overstimulation or misdirection of immune response, so it can act against the body itself. One of the organs most affected by this reaction is the thyroid gland, and there is no definitive explanation of the causes of this adverse event. Material and methods: In this retrospective observational study, we enrolled 103 patients with NSCLC and high PD-L1 expression (>= 50%) who were treated in our Clinic for pulmonology, University Clinical Center of Serbia, using Pembrolizumab as the first-line therapy. Results: Data analysis showed that 41 (39.81%) of 103 patients in our study had an adverse event of immunotherapy, and 21 of them had autoimmune thyroiditis (20.39%). Of all the patients, 19 of them were treated for chronic obstructive pulmonary disease (COPD) before the onset of Pembrolizumab. During treatment, eight of these patients developed thyroid dysfunction. Patients with COPD were at increased risk of developing autoimmune thyroiditis compared to non-COPD patients (OR 3.9 95% CI 1.135–13.260, p = 0.0227). Conclusion: Our study showed that patients dealing with COPD have a 3.9 times greater risk of developing autoimmune thyroiditis as an adverse event during Pembrolizumab treatment compared with patients without COPD. C1 [Zecevic, Andrej] University Clinical Center of Serbia, Clinic for PulmonologyBelgrade, Serbia. [Blanka-Protic, Ana] University Clinical Center of Serbia, Clinic for PulmonologyBelgrade, Serbia. [Jandric, Aleksandar] University Clinical Center of Serbia, Clinic for PulmonologyBelgrade, Serbia. [Adzic-Vukicevic, Tatjana] University Clinical Center of Serbia, Clinic for PulmonologyBelgrade, Serbia. RP Zecevic, A (reprint author), University Clinical Center of Serbia, Clinic for Pulmonology, Belgrade, Serbia. EM zecevic92andrej@gmail.com CR Durham AL, Adcock IM. The relationship between COPD and lung cancer. Lung Cancer, 2015, 90(2):121–7., DOI 10.1016/j.lungcan.2015.08.017 Qi C, Sun SW, Xiong XZ. From COPD to lung cancer: mechanisms linking, diagnosis, treatment, and prognosis. Int J Chron Obstruct Pulmon Dis, 2022, 17: 2603–21., DOI 10.2147/COPD.S380732 Forder A, Zhuang R, Souza VGP, Brockley LJ, Pewarchuk ME, Telkar N, et al. Mechanisms contributing to the comorbidity of COPD and lung cancer. Int J Mol Sci, 2023, 24(3):2859., DOI 10.3390/ijms24032859 Lahiri A, Maji A, Potdar PD, Singh N, Parikh P, Bisht B, et al. Lung cancer immunotherapy: progress, pitfalls, and promises. Mol Cancer, 2023, 22(1):40., DOI 10.1186/s12943-023-01740-y Steven A, Fisher SA, Robinson BW. Immunotherapy for lung cancer. Respirology, 2016, 21(5):821–33., DOI 10.1111/resp.12789 Borst J, Busselaar J, Bosma DMT, Ossendorp F. Mechanism of action of PD-1 receptor/ligand targeted cancer immunotherapy. Eur J Immunol, 2021, 51(8): 1911–20., DOI 10.1002/eji.202048994 Cavic M, Kovacevic T, Zaric B, Stojiljkovic D, Korda NJ, Rancic M, et al. Lung cancer in Serbia. J Thorac Oncol, 2022, 17(7):867–72., DOI 10.1016/j.jtho.2022. 04.010 Li Z, Xia Y, Xia M, Liu C, Wang T, Liu Y, et al. Immune-related thyroid dysfunction is associated with improved long-term prognosis in patients with nonsmall cell lung cancer treated with immunotherapy: a systematic review and metaanalysis. J Thorac Dis, 2023, 15(2):690–700., DOI 10.21037/jtd-23-254 Young RP, Hopkins RJ, Christmas T, Black PN, Metcalf P, Gamble GD. COPD prevalence is increased in lung cancer, independent of age, sex and smoking history. Eur Respir J, 2009, 34(2):380–6., DOI 10.1183/09031936. 00144208 Denault MH, Melosky B. Immunotherapy in the first-line setting in wild-type NSCLC. Curr Oncol, 2021, 28(6):4457–70., DOI 10.3390/curroncol28060378 Champiat S, Lambotte O, Barreau E, Belkhir R, Berdelou A, Carbonnel F, et al. Management of immune checkpoint blockade dysimmune toxicities: a collaborative position paper. Ann Oncol, 2016, 15:559–74., DOI 10.1093/ annonc/mdv623 Luo WX, Yang L, Li WM. Immunotherapy-related toxicity in lung cancer: clinical characteristics and managing strategy. Chin Med J, Engl,, 2021, 134(7): 780–2., DOI 10.1097/CM9.0000000000001287 Ferrari SM, Fallahi P, Elia G, Ragusa F, Ruffilli I, Patrizio A, et al. Autoimmune endocrine dysfunctions associated with cancer immunotherapies. Int J Mol Sci, 2019, 20(10):2560., DOI 10.3390/ijms20102560 D’Aiello A, Lin J, Gucalp R, Tabatabaie V, Cheng H, Bloomgarden NA, et al. Thyroid dysfunction in lung cancer patients treated with immune checkpoint inhibitors, ICIs): outcomes in a multiethnic urban cohort. Cancers, Basel,, 2021, 13(6):1464., DOI 10.3390/cancers13061464 Yuen KCJ, Samson SL, Bancos I, Gosmanov AR, Jasim S, Fecher LA, et al. American association of clinical endocrinology disease state clinical review: evaluation and management of immune checkpoint inhibitor-mediated endocrinopathies: a practical case-based clinical approach. Endocr Pract, 2022, 28(7):719–31., DOI 10.1016/j.eprac.2022.04.010 Yamauchi I, Sakane Y, Fukuda Y, Fujii T, Taura D, Hirata M, et al. Clinical features of nivolumab-induced thyroiditis: a case series study. Thyroid, 2017, 27(7): 894–901., DOI 10.1089/thy.2016.0562 Delivanis DA, Gustafson MP, Bornschlegl S, Merten MM, Kottschade L, Withers S, et al. Pembrolizumab-Induced thyroiditis: comprehensive clinical review and insights into underlying involved mechanisms. J Clin Endocrinol Metab, 2017, 102(8):2770–80., DOI 10.1210/jc.2017-00448 Iyer PC, Cabanillas ME, Waguespack SG, Hu MI, Thosani S, Lavis VR, et al. Immune-related thyroiditis with immune checkpoint inhibitors. Thyroid, 2018, 28(10):1243–51., DOI 10.1089/thy.2018.0116 Baraka B, Abosheaishaa H, Nassar M. Immunotherapy-induced thyroid dysfunction: an updated review. Egypt J Intern Med, 2023, 35:48., DOI 10.1186/ s43162-023-00210-7 Hemminki K, Liu X, Ji J, Sundquist K, Sundquist J. Subsequent COPD and lung cancer in patients with autoimmune disease. Eur Respir J, 2011, 37(2):463–5., DOI 10.1183/09031936.0007041009031936.00070410 Ciobanu L, Maciuc V, Costan R, Gorga A, Bobirca PA, Corduneanu S. Is COPD a risk factor for thyroid disorders? Eur Respir J, 2011, 38(Suppl. 55):p3593. Doehner W, Haeusler KG, Endres M, Anker SD, MacNee W, Lainscak M. Neurological and endocrinological disorders: orphans in chronic obstructive pulmonary disease. Respir Med, 2011, 105(Suppl. 1):S12–9., DOI 10.1016/S0954-6111(11)70005-1 Papaioannou O, Karampitsakos T, Barbayianni I, Chrysikos S, Xylourgidis N, Tzilas V, et al. Metabolic disorders in chronic lung diseases. Front Med, Lausanne,, 2018, 18(4):246., DOI 10.3389/fmed.2017.00246 Chaudhary SC, Ahmad T, Usman K, Sawlani KK, Gupta KK, Verma AK, et al. Prevalence of thyroid dysfunction in chronic obstructive pulmonary disease patients in a tertiary care center in North India. J Fam Med Prim Care, 2018, 7(3):584–8., DOI 10.4103/jfmpc.jfmpc_84_17 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1612022 EP 1612026 DI 10.3389/pore.2025.1612022 PG 5 ER PT J AU Nagy, FZs Pfliegler, Gy Kosa, J Arvai, K Istenes, I Doros, A Timar, B Lakatos, P Demeter, J AF Nagy, Flora Zsofia Pfliegler, Gyorgy Kosa, Janos Arvai, Kristof Istenes, Ildiko Doros, Attila Timar, Botond Lakatos, Peter Demeter, Judit TI Case Report: Importance of high-throughput genetic investigations in the differential diagnosis of unexplained erythrocytosis SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE polyglobulia; WES; diagnostic; chuvash polycythaemia; pyruvate kinase hyperactivity ID polyglobulia; WES; diagnostic; chuvash polycythaemia; pyruvate kinase hyperactivity AB Introduction: Polycythemia indicates the pathological increase in the number of red blood cells and the rise of hematocrit values. Polyglobulia can be of primary or secondary origin, with the most common primary polycythemia being a myeloproliferative neoplasm, polycythemia vera. Polyglobulia patients may develop cardiovascular complications and thromboembolic events. The gold standard of first-line treatment in polycythemia vera is phlebotomy, which is indicated to keep the hematocrit value below 0.45. Until now the goal to be achieved in secondary polyglobulia has been similar. In secondary polyglobulia this rule of thumb needs to be re-evaluated as shown by the example of two patients suffering from different rare, genetically determined polyglobulias. In our article we present the case of these two patients and discuss the diagnostic and therapeutic principles to be applied in patients with rare, genetically determined polyglobulias. Patients and methods: After completing the usual diagnostic algorithm for polyglobulia no cause could be identified in two of our male patients. Therefore, we set out to perform whole exome sequencing in both patients. Our analysis did not include copy number analysis. Results: In Patient 1 the p.Ser179Pro variant in the VHL gene was detected in the homozygous state, which is classified as likely pathogenic according to the ACMG guidelines. Homozygous VHL mutations are implicated in Chuvash polycythemia. Segregation analysis was declined by the family. In Patient 2 the PKLR gene p.His306Gln variant was detected in the heterozygous form. The gene plays a role in pyruvate metabolism. Family screening did not detect this variant in healthy family members. Discussion: We identified rare, possibly pathogenic genetic variants in two patients with polyglobulia and as a consequence of the genetic diagnosis we implemented individualized patient monitoring. We recommend the utilization of high-throughput genomic testing in cases with unexplained polyglobulia. C1 [Nagy, Flora Zsofia] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Pfliegler, Gyorgy] Debreceni Egyetem Klinikai Kozpont, Ritka Betegsegek Szakertoi Kozpont es Belgyogyaszati IntezetDebrecen, Hungary. [Kosa, Janos] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Arvai, Kristof] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Istenes, Ildiko] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Doros, Attila] Semmelweis University, Department of Radiology and OncotherapyBudapest, Hungary. [Timar, Botond] Semmelweis University, 1st Department of Pathology and Experimental Cancer ResearchBudapest, Hungary. [Lakatos, Peter] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. [Demeter, Judit] Semmelweis University, Department of Internal Medicine and OncologyBudapest, Hungary. RP Nagy, FZs (reprint author), Semmelweis University, Department of Internal Medicine and Oncology, Budapest, Hungary. EM nagy.zsofia.flora@semmelweis.hu CR Arber DA, Orazi A, Hasserjian RP, Borowitz MJ, Calvo KR, Kvasnicka HM, et al. International consensus classification of myeloid neoplasms and acute leukemias: integrating morphologic, clinical, and genomic data. Blood, 2022, 140:1200–28., DOI 10.1182/blood.2022015850 Prchal JT. Polycythemia vera and other primary polycythemias. Curr Opin Hematol, 2005, 12:112–6., DOI 10.1097/01.moh.0000154029.05396.d2 Babakhanlou R, Verstovsek S, Pemmaraju N, Rojas-Hernandez CM. Secondary erythrocytosis. Expert Rev Hematol, 2023, 16:245–51., DOI 10.1080/17474086.2023.2192475 Jako J, Takacs P, Szerafin L. Incidence of haematological malignancies in adults in Szabolcs-Szatmar-Bereg county, Hungary Analysis of data of a 36-year period. Orv Hetil, 2020, 161:1400–13., DOI 10.1556/650.2020.31750 Patnaik MM, Tefferi A. The complete evaluation of erythrocytosis: congenital and acquired. Leukemia, 2009, 23:834–44., DOI 10.1038/leu.2009.54 Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of medical genetics and genomics and the association for molecular pathology. Genet Med, 2015, 17:405–24., DOI 10.1038/gim.2015.30 Kopanos C, Tsiolkas V, Kouris A, Chapple CE, Albarca Aguilera M, Meyer R, et al. VarSome: the human genomic variant search engine. Bioinformatics, 2019, 35: 1978–80., DOI 10.1093/bioinformatics/bty897 Randi ML, Murgia A, Putti MC, Martella M, Casarin A, Opocher G, et al. Low frequency of VHL gene mutations in young individuals with polycythemia and high serum erythropoietin. Haematologica, 2005, 90:689–91. Cario H, Schwarz K, Jorch N, Kyank U, Petrides PE, Schneider DT, et al.Mutations in the von Hippel-Lindau, VHL, tumor suppressor gene and VHL-haplotype analysis in patients with presumable congenital erythrocytosis. Haematologica, 2005, 90:19–24. Pastore Y, Jedlickova K, Guan Y, Liu E, Fahner J, Hasle H, et al. Mutations of von Hippel-Lindau tumor-suppressor gene and congenital polycythemia. Am J Hum Genet, 2003, 73:412–9., DOI 10.1086/377108 Sergeyeva A, Gordeuk VR, Tokarev YN, Sokol L, Prchal JF, Prchal JT. Congenital polycythemia in Chuvashia. Blood, 1997, 89:2148–54., DOI 10.1182/ blood.v89.6.2148 Remenyi G, Bereczky Z, Gindele R, Ujfalusi A, Illes A, Udvardy M. rs779805 von hippel-lindau gene polymorphism induced/related polycythemia entity, clinical features, cancer association, and familiar characteristics. Pathol Oncol Res, 2021, 27:1609987–6., DOI 10.3389/pore. 2021.1609987 Tomasic NL, Piterkova L, Huff C, Bilic E, Yoon D, Miasnikova GY, et al. The phenotype of polycythemia due to Croatian homozygous VHL, 571C>G: H191D, mutation is different from that of Chuvash polycythemia, VHL 598C>T:R200W). Haematologica, 2013, 98:560–7., DOI 10.3324/haematol. 2012.070508 Ang SO, ChenH,Hirota K, Gordeuk VR, Jelinek J, Guan Y, et al. Disruption of oxygen homeostasis underlies congenital Chuvash polycythemia. Nat Genet, 2002, 32:614–21., DOI 10.1038/ng1019 Hickey MM, Lam JC, Bezman NA, Rathmell WK, Simon MC. Von Hippel- Lindau mutation in mice recapitulates Chuvash polycythemia via hypoxiainducible factor-2α signaling and splenic erythropoiesis. J Clin Invest, 2007, 117:3879–89., DOI 10.1172/JCI32614 Lanikova L, Lorenzo F, Yang C, Vankayalapati H, Drachtman R, Divoky V, et al. Novel homozygous VHL mutation in exon 2 is associated with congenital polycythemia but not with cancer. Blood, 2013, 121:3918–24., DOI 10.1182/blood- 2012-11-469296 Russell RC, Sufan RI, Zhou B, Pardeep H, Severa B, Stephanie SS, et al. Loss of Jak2 regulation via vhl-socs1 E3 ubiquitin heterocomplex underlies Chuvash polycythemia. Nature, 2011, 17:845–53., DOI 10.1038/nm.2370 Maher ER, Iselius L, Yates JR, Littler M, Benjamin C, Harris R, et al. Von Hippel-Lindau disease: a genetic study. J Med Genet, 1991, 28:443–7., DOI 10.1136/ jmg.28.7.443 Gordeuk VR, Sergueeva AI, Miasnikova GY, Okhotin D, Voloshin Y, Choyke PL, et al. Congenital disorder of oxygen sensing: association of the homozygous Chuvash polycythemia VHL mutation with thrombosis and vascular abnormalities but not tumors. Blood, 2004, 103:3924–32., DOI 10.1182/blood-2003-07-2535 Sarangi S, Lanikova L, Kapralova K, Acharya S, Swierczek S, Lipton JM, et al. The homozygous VHL(D126N, missense mutation is associated with dramatically elevated erythropoietin levels, consequent polycythemia, and early onset severe pulmonary hypertension. Pediatr Blood Cancer, 2014, 61:2104–6., DOI 10.1002/pbc. 25056 Brewer GJ. A new inherited abnormality of human erythrocytes - elevated erythrocytic adenosine triphosphate. Biochem Biophys Res Commun, 1965, 18: 430–4., DOI 10.1016/0006-291X(65)90726-6 Max-Audit I, Rosa R, Marie J. Pyruvate kinase hyperactivity genetically determined: metabolic consequences and molecular characterization. Blood, 1980, 56:902–9., DOI 10.1182/blood.v56.5.902.902 Beutler E, Westwood B, Van Zwieten R, Roos D. G→T transition at cdna nt 110, k37q, in the pklr, pyruvate kinase, gene is the molecular basis of a case of hereditary increase of red blood cell atp. Hum Mutat, 1997, 9:282–5., DOI 10.1002/, SICI)1098-1004(1997)9:3<282::AID-HUMU13>3.0.CO;2-Z van Oirschot BA, Francois JJJM, Van Solinge WW, van Wesel ACW, Rijksen G, van Amstel HKP, et al. Novel type of red blood cell pyruvate kinase hyperactivity predicts a remote regulatory locus involved in PKLR gene expression. Am J Hematol, 2014, 89:380–4., DOI 10.1002/ajh.23647 Zanella A, Rebulla P, Vullo C, Izzo C, Tedesco F, Sirchia G. Hereditary pyruvate kinase deficiency: role of the abnormal enzyme in red cell pathophysiology. Br J Haematol, 1978, 40:551–62., DOI 10.1111/j.1365-2141.1978. tb05831.x Zanella A, Fermo E, Bianchi P, Valentini G. Red cell pyruvate kinase deficiency: molecular and clinical aspects. Br J Haematol, 2005, 130:11–25., DOI 10.1111/j.1365-2141.2005.05527.x NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1612037 EP 1612043 DI 10.3389/pore.2025.1612037 PG 7 ER PT J AU Qiu, Z Ke, F Zhu, X AF Qiu, Zijian Ke, Fei Zhu, Xiaoping TI Case report: Alectinib-associated intestinal ulceration and colitis in a patient with non-small cell lung cancer and effective treatment with Mesalazine SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Case Reports DE Alectinib; intestinal ulcer; colitis; anaplastic lymphoma kinase inhibitor; non-small cell lung cancer ID Alectinib; intestinal ulcer; colitis; anaplastic lymphoma kinase inhibitor; non-small cell lung cancer AB Background: Alectinib is effective in extending the survival of patients with anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (NSCLC) and generally has manageable side effects. However, intestinal ulcers and colitis are rare but serious adverse reactions linked to Alectinib, meriting further investigation into their causes. Case presentation: We report the case of a 62-year-old woman with NSCLC and brain metastases, who tested positive for ALK. She had been treated with Alectinib for nearly 4 years. The patient experienced diarrhea for 4 days, and a subsequent colonoscopy revealed pancolitis along with multiple ulcers in the terminal ileum and ileocecal valve. Given the severity of these symptoms, classified as a grade 3 adverse event by the Common Terminology Criteria for Adverse Events (CTCAE), Alectinib was discontinued. Treatment with oral enteric-coated Mesalazine tablets led to a resolution of the diarrhea and a significant improvement in the pancolitis and ulcers upon follow-up. The patient’s anticancer therapy was subsequently switched to Ceritinib capsules. At follow-up, she demonstrated a stable tumor condition with no recurrence of intestinal ulcers or colitis. Conclusion: To our knowledge, this is the first reported case of intestinal ulceration and colitis induced by Alectinib. Although such adverse events are exceedingly rare, they require vigilant monitoring in clinical practice. Decisions on continuing with Alectinib should consider the severity of side effects, classified by CTCAE grade. For managing these specific adverse events, oral Mesalazine enteric-coated tablets appear to be an effective treatment option. C1 [Qiu, Zijian] The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Department of Radiation OncologyQuzhou, Zhejiang, China. [Ke, Fei] The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Department of PathologyQuzhou, Zhejiang, China. [Zhu, Xiaoping] The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Department of Radiation OncologyQuzhou, Zhejiang, China. RP Zhu, X (reprint author), The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Department of Radiation Oncology, Quzhou, China. EM 25229661@qq.com CR Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature, 2007, 448(7153):561–6., DOI 10.1038/nature05945 Solomon B, Varella-Garcia M, Camidge DR. ALK gene rearrangements: a new therapeutic target in a molecularly defined subset of non-small cell lung cancer. J Thorac Oncol, 2009, 4(12):1450–4., DOI 10.1097/JTO.0b013e3181c4dedb Peng Y, Zhao Q, Liao Z, Ma Y, Ma D. Efficacy and safety of first-line treatments for patients with advanced anaplastic lymphoma kinase mutated, non-small cell cancer: a systematic review and network meta-analysis. Cancer., 2023, 129(8): 1261–75., DOI 10.1002/cncr.34664 Lin JJ, Horan JC, Tangpeerachaikul A, Swalduz A, Valdivia A, Johnson ML, et al. NVL-655 is a selective and brain-penetrant inhibitor of diverse ALK-mutant oncoproteins, including lorlatinib-resistant compound mutations. Cancer Discov, 2024, 14:2367–86., DOI 10.1158/2159-8290.CD-24-0231 Mok T, Camidge DR, Gadgeel SM, Rosell R, Dziadziuszko R, Kim DW, et al. Updated overall survival and final progression-free survival data for patients with treatment-naive advanced ALK-positive non-small-cell lung cancer in the ALEX study. Ann Oncol, 2020, 31(8):1056–64., DOI 10.1016/j.annonc.2020.04.478 Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim DW, et al. Alectinib versus Crizotinib in untreated ALK-positive non-small-cell lung cancer. N Engl J Med, 2017, 377(9):829–38., DOI 10.1056/NEJMoa1704795 Dziadziuszko R, Peters S, Ruf T, Cardona A, Guerini E, Kurtsikidze N, et al. Clinical experience and management of adverse events in patients with advanced ALK-positive non-small-cell lung cancer receiving alectinib. ESMO Open, 2022, 7(6):100612., DOI 10.1016/j.esmoop.2022.100612 Gursoy P. Avascular necrosis during alectinib treatment: case report. J Oncol Pharm Pract, 2023, 29(1):242–5., DOI 10.1177/10781552221102370 Bajaj S, Chow A, Drilon A, Kalchiem-Dekel O. Spontaneous bilateral chylothorax development during alectinib therapy for ALK-rearranged NSCLCA case report. JTO Clin Res Rep, 2023, 4(12):100606., DOI 10.1016/j.jtocrr.2023. 100606 Ramachandran P, Morcus R, Tahir M, Onukogu I, Spinowitz B, Wang JC. Alectinib, Alecensa)-induced reversible grade IV nephrotoxicity: a case report and review of the literature. J Med Case Rep, 2018, 12(1):303., DOI 10.1186/s13256-018- 1849-y Misawa K, Nakamichi S, Iida H, Nagano A, Mikami E, Tozuka T, et al. Alectinib-induced severe hemolytic anemia in a patient with ALK-positive nonsmall cell lung cancer: a case report. Onco Targets Ther, 2023, 16:65–9., DOI 10.2147/ OTT.S398375 Chen X, Xia D, Jiang X, Cao L, Ryu JH, Hu X. Case report: indolent drugrelated pneumonitis with alectinib therapy in the treatment of non-small cell lung cancer. Front Pharmacol, 2022, 13:944685., DOI 10.3389/fphar.2022.944685 Maina RM, Rader CA, Muenyi CS, Battini R, Zalamea NN, Foretia DA. Alectinib-associated pneumoperitoneum in stage IV non-small cell lung cancer - a case report. Ann Med Surg, Lond,, 2022, 82:104601., DOI 10.1016/j.amsu.2022.104601 Patel M, Khalid B, Ghani S. Alectinib-associated perforated duodenal ulcer. Am J Ther, 2020, Publish Ahead of Print., DOI 10.1097/MJT.0000000000001301 Wu YL, Dziadziuszko R, Ahn JS, Barlesi F, Nishio M, Lee DH, et al. Alectinib in resected ALK-positive non-small-cell lung cancer. N Engl J Med, 2024, 390(14): 1265–76., DOI 10.1056/NEJMoa2310532 Luo Y, Zhang Z, Guo X, Tang X, Li S, Gong G, et al. Comparative safety of anaplastic lymphoma kinase tyrosine kinase inhibitors in advanced anaplastic lymphoma kinase-mutated non-small cell lung cancer: systematic review and network meta-analysis. Lung Cancer, 2023, 184:107319., DOI 10.1016/j.lungcan. 2023.107319 Morris SW, Kirstein MN, Valentine MB, Dittmer KG, Shapiro DN, Saltman DL, et al. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non- Hodgkin’s lymphoma. Science, 1994, 263(5151):1281–4., DOI 10.1126/science. 8122112 Englund C, Loren CE, Grabbe C, Varshney GK, Deleuil F, Hallberg B, et al. Jeb signals through the Alk receptor tyrosine kinase to drive visceral muscle fusion. Nature, 2003, 425(6957):512–6., DOI 10.1038/nature01950 Lasota J, Chlopek M, Wasag B, Kowalik A, Christiansen J, Lamoureux J, et al. Colorectal adenocarcinomas harboring ALK fusion genes: a clinicopathologic and molecular genetic study of 12 cases and review of the literature. Am J Surg Pathol, 2020, 44(9):1224–34., DOI 10.1097/PAS.0000000000001512 Van Sebille YZA, Gibson RJ, Wardill HR, Secombe KR, Ball IA, Keefe DMK, et al. Dacomitinib-induced diarrhoea is associated with altered gastrointestinal permeability and disruption in ileal histology in rats. Int J Cancer, 2017, 140(12): 2820–9., DOI 10.1002/ijc.30699 Xu X, Lai Y, Hua ZC. Apoptosis and apoptotic body: disease message and therapeutic target potentials. Biosci Rep, 2019, 39(1)., DOI 10.1042/BSR20180992 Hong S, Chen N, FangW, Zhan J, Liu Q, Kang S, et al. Upregulation of PD-L1 by EML4-ALK fusion protein mediates the immune escape in ALK positive NSCLC: implication for optional anti-PD-1/PD-L1 immune therapy for ALK-TKIs sensitive and resistant NSCLC patients. Oncoimmunology, 2016, 5(3):e1094598., DOI 10. 1080/2162402X.2015.1094598 Graff LA, Geist R, Kuenzig ME, Benchimol EI, Kaplan GG, Windsor JW, et al. The 2023 impact of inflammatory bowel disease in Canada: mental health and inflammatory bowel disease. J Can Assoc Gastroenterol, 2023, 6(Suppl. 2):S64–s75., DOI 10.1093/jcag/gwad012 Zhou F, Yang Y, Zhang L, Cheng Y, Han B, Lu Y, et al. Expert consensus of management of adverse drug reactions with anaplastic lymphoma kinase tyrosine kinase inhibitors. ESMO Open, 2023, 8(3):101560., DOI 10.1016/j.esmoop.2023.101560 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1612040 EP 1612045 DI 10.3389/pore.2025.1612040 PG 6 ER PT J AU Csepregi, BA Papp, E Adamik, I Csernak, E Engi, H Kuronya, Zs Soos, E Melegh, Zs Toth, E AF Csepregi, Beata Anna Papp, Eszter Adamik, Imola Csernak, Erzsebet Engi, Helga Kuronya, Zsofia Soos, Edina Melegh, Zsombor Toth, Erika TI Impact of comprehensive genomic profiling on the diagnosis and clinical management of malignant mesenchymal tumours SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE next generation sequencing; comprehensive genomic profiling; targeted therapy; sarcoma; mesenchymal tumours ID next generation sequencing; comprehensive genomic profiling; targeted therapy; sarcoma; mesenchymal tumours AB Comprehensive genomic profiling (CGP) is becoming an increasingly important tool in the clinical management of different tumours, but there is still very limited data available on its usefulness from a therapeutic point of view in mesenchymal tumours. Between January 2022 and September 2024, we performed CGP analysis with means of Oncomine Comprehensive Assay Plus (OCAplus) on 94 malignant mesenchymal tumours. The analysis covered more than 500 unique genes for single-gene and multigene biomarker insights, including tumour mutational burden (TMB) and homologous recombination deficiency (HRD). Genomic DNA and total RNA were extracted from formalin-fixed paraffin-embedded tissue blocks. Twentyfour out of 94 patients (25.5%) had potentially actionable alterations: 17 (18%) had specific genetic alterations suitable for targeted therapies, 4 (4.2%) had a high TMB (>10 mut/Mb), and 5 (5.3%) had a high HRD score >15). One additional patient had BRCA1 mutation, but the HRD score was low. Three patients received targeted therapy: one patient with a CDK4-amplified tumour (dedifferentiated liposarcoma) received CDK4 inhibitor therapy, two patients with angiosarcoma showing high TMB received immune checkpoint inhibitor therapy, and one patient with a uterine leiomyosarcoma and high HRD score received PARP inhibitor therapy. In addition, two patients with malignant phyllodes tumours received multi-thyrosine kinase inhibitor therapy. In three cases, there was refinement or reassignment of the diagnosis, based on the CGP findings. Our results demonstrate that CGP can provide useful additional information and can be beneficial in the clinical management of patients with mesenchymal tumours. C1 [Csepregi, Beata Anna] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Papp, Eszter] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Adamik, Imola] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Csernak, Erzsebet] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Engi, Helga] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Kuronya, Zsofia] National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology DepartmentBudapest, Hungary. [Soos, Edina] National Institute of Oncology, Urogenital Tumors and Clinical Pharmacology DepartmentBudapest, Hungary. [Melegh, Zsombor] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. [Toth, Erika] National Institute of Oncology, Center of Surgical and Molecular Tumor PathologyBudapest, Hungary. RP Toth, E (reprint author), National Institute of Oncology, Center of Surgical and Molecular Tumor Pathology, Budapest, Hungary. EM dr.toth.erika@oncol.hu CR WHOClassification of Tumours Editorial Board. Soft tissue and bone tumours. WHO classification of tumours series, international Agency for Research on cancer. 5th ed, Vol. 3. Lyon, 2020). Available online at: https://publications.iarc.fr/588, Accessed December 1, 2024) Fontebasso AM, Rytlewski JD, Blay JY, Gladdy RA, Wilky BA. Precision Oncology in soft tissue sarcomas and gastrointestinal stromal tumors. Surg Oncol Clin N Am, 2024, 33(2):387–408., DOI 10.1016/j.soc.2023.12.018 Cote GM, He J, Choy E. Next-generation sequencing for patients with sarcoma: a single center experience. Oncologist, 2018, 23(2):234–42., DOI 10.1634/ theoncologist.2017-0290 Kokkali S, Georgaki E, Mandrakis G, Valverde C, Theocharis S. Genomic profiling and clinical outcomes of targeted therapies in adult patients with soft tissue sarcomas. Cells, 2023, 12(22):2632., DOI 10.3390/cells12222632 Toth E, Kuronya Z, Soos E, Pinter T, Butz H, Horvath Z, et al. Application of comprehensive molecular genetic profiling in precision cancer medicine, Hungarian experiences. Acta Oncol, 2024, 16(63):433–40., DOI 10.2340/1651- 226X.2024.39918 Nassif EF, Auclin E, Bahleda R, Honore C, Mir O, Dumont S, et al. TP53 mutation as a prognostic and predictive marker in sarcoma: pooled analysis of MOSCATO and ProfiLER precision medicine trials. Cancers, Basel,, 2021, 13(13): 3362., DOI 10.3390/cancers13133362 Xu L, Xie X, Shi X, Zhang P, Liu A, Wang J, et al. Potential application of genomic profiling for the diagnosis and treatment of patients with sarcoma. Oncol Lett, 2021, 21(5):353., DOI 10.3892/ol.2021.12614 Gounder MM, Agaram NP, Trabucco SE, Robinson V, Ferraro RA, Millis SZ, et al. Clinical genomic profiling in the management of patients with soft tissue and bone sarcoma. Nat Commun, 2022, 13(1):3406., DOI 10.1038/s41467-022-30496-0 Zhou MY, Bui NQ, Charville GW, Ganjoo KN, Pan M. Treatment of dedifferentiated liposarcoma in the era of immunotherapy. Int J Mol Sci, 2023, 24(11): 9571., DOI 10.3390/ijms24119571 O’Leary B, Finn RS, Turner NC. Treating cancer with selective CDK4/6 inhibitors. Nat Rev Clin Oncol, 2016, 13(7):417–30., DOI 10.1038/nrclinonc.2016.26 Anastasiou M, Kyriazoglou A, Kotsantis I, Economopoulou P, Kyrkasiadou M, Giannopoulou A, et al. Immune checkpoint inhibitors in sarcomas: a systematic review. Immunooncol Technol, 2023, 20:100407., DOI 10. 1016/j.iotech.2023.100407 Yiong CS, Lin TP, Lim VY, Toh TB, Yang VS. Biomarkers for immune checkpoint inhibition in sarcomas - are we close to clinical implementation? Biomark Res, 2023, 11(1):75., DOI 10.1186/s40364-023-00513-5 Ravi V, Subramaniam A, Zheng J, Amini B, Trinh VA, Joseph J, et al. Clinical activity of checkpoint inhibitors in angiosarcoma: a retrospective cohort study. Cancer, 2022, 128(18):3383–91., DOI 10.1002/cncr.34370 Fernandes I, Dias E, Silva D, Segatelli V, Filippi RZ, Carolina de Rezende A, et al. Microsatellite instability and clinical use in sarcomas: systematic review and illustrative case report. JCO Precis Oncol, 2024, 8:e2400047., DOI 10.1200/PO.24.00047 Lucchesi C, Khalifa E, Laizet Y, Soubeyran I, Mathoulin-Pelissier S, Chomienne C, et al. Targetable alterations in adult patients with soft-tissue sarcomas: insights for personalized therapy. JAMA Oncol, 2018, 4(10): 1398–404., DOI 10.1001/jamaoncol.2018.0723 Gusho CA, Weiss MC, Lee L, Gitelis S, Blank AT, Wang D, et al. The clinical utility of next-generation sequencing for bone and soft tissue sarcoma. Acta oncologica, Stockholm, Sweden,, 2022, 61(1):38–44., DOI 10.1080/0284186X.2021.1992009 Watkins JA, Trotman J, Tadross JA, Harrington J, Hatcher H, Horan G, et al. Introduction and impact of routine whole genome sequencing in the diagnosis and management of sarcoma. Br J Cancer, 2024, 131:860–9., DOI 10.1038/s41416-024- 02721-8 Schipper LJ, Monkhorst K, Samsom KG, Bosch LJW, Snaebjornsson P, van Boven H, et al. Clinical impact of prospective whole genome sequencing in sarcoma patients. Cancers, Basel,, 2022, 14(2):436. PMID: 35053600; PMCID: PMC8773512., DOI 10.3390/cancers14020436 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1612065 EP 1612071 DI 10.3389/pore.2025.1612065 PG 7 ER PT J AU Talor, E Timar, J Lavin, P Cipriano, J Markovic, D Ladanyi, A Bondarenko, I Stosic, S Sobat, H Zhukavets, A Imamovic, N Bankowska-Wozniak, M Kisely, M Jovic, R Young, EMJ AF Talor, Eyal Timar, Jozsef Lavin, Philip Cipriano, John Markovic, Dusan Ladanyi, Andrea Bondarenko, Igor Stosic, Srboljub Sobat, Hrvoje Zhukavets, Aliaksandr Imamovic, Nazim Bankowska-Wozniak, Magdalena Kisely, Mihaly Jovic, Rajko Young, Edward Massey James TI Neoadjuvant leukocyte interleukin injection immunotherapy improves overall survival in low-risk locally advanced head and neck squamous cell carcinoma –the IT-MATTERS study SO PATHOLOGY & ONCOLOGY RESEARCH LA English DT Journal Article DE Leukocyte interleukin injection (LI); immunotherapy; neoadjuvant; SCCHN; locally advanced disease; low-risk for recurrence; lower disease burden ID Leukocyte interleukin injection (LI); immunotherapy; neoadjuvant; SCCHN; locally advanced disease; low-risk for recurrence; lower disease burden AB The randomized controlled pivotal phase 3 study evaluated efficacy and safety of neoadjuvant complex biologic, Leukocyte Interleukin Injection (LI), administered for 3 consecutive weeks pre-surgery, in treatment naive resectable locally advanced primary squamous cell carcinoma of oral cavity and soft palate. Randomization 3:1:3 to LI+/-CIZ (cyclophosphamide, indomethacin, and zinc)+SOC, or SOC (standard of care) alone. LI-treated patients received 400 IU (as interleukin-2 equivalent; 200 IU peritumorally, 200 IU perilymphatically) sequentially, daily 5 days/week for 3 weeks before surgery. All subjects were to receive SOC. Post-surgery, patients with low risk for recurrence were to receive radiotherapy, while those with high risk received concurrent chemoradiotherapy. Median follow-up was 56 months. There were 923 ITT (Intent-to-Treat) subjects (380 ITT low-risk and 467 ITT high-risk). Presurgery objective early response (45 objective early responders; 5 complete responses [CRs], 40 partial responses [PRs], confirmed by pathology at surgery. LI (+/− CIZ) had 8.5% objective early responders (45/529 ITT) and 16% objective early responders (34/212 ITT low-risk) vs. no reported SOC objective early responders (0/394 ITT). Objective early responders significantly lowered death rate to 22.2% (ITT LI-treated), 12.5% (ITT low-risk LI + CIZ + SOC), while the ITT low-risk SOC death rate was 48.7%. Thus, objective early response impacted overall survival (OS); proportional hazard ratios were 0.348 (95% CI: 0.152–0.801) for ITT low-risk LI-treated, 0.246 (95% CI: 0.077–0.787) for ITT low-risk LI + CIZ + SOC. ITT low-risk LI + CIZ + SOC demonstrated significant OS advantage vs. ITT low-risk SOC (unstratified log-rank p = 0.048; Cox hazard ratio = 0.68; 95% CI: 0.48–0.95, Wald p = 0.024 [controlling for tumor stage, tumor location, and geographic region]). Absolute OS advantage increased over time for ITT low-risk (LI + CIZ + SOC)-treated vs. ITT low-risk SOC: reaching 14.1% (62.7% vs. 48.6%) at 60 months, with 46.5 months median OS advantage (101.7 months vs. 55.2 months), respectively. Quality of life benefit for complete responders sustained for >3 years post LI treatment. Percent treatmentemergent adverse events were comparable among all treated groups. No excess safety issues were reported for LI over SOC alone post-surgery. NCT01265849, EUDRA:2010-019952-35. C1 [Talor, Eyal] CEL-SCI CorporationVienna, VA, USA. [Timar, Jozsef] Semmelweis University, 2nd Department of PathologyBudapest, Hungary. [Lavin, Philip] Boston Biostatistics Research FoundationFramingham, MA, USA. [Cipriano, John] CEL-SCI CorporationVienna, VA, USA. [Markovic, Dusan] Ergomed GroupGuildford, UK. [Ladanyi, Andrea] Orszagos Onkologiai Intezet, Nemzeti Tumorbiologiai LaboratoriumBudapest, Hungary. [Bondarenko, Igor] State Institution “Dnipropetrovsk Medical Academy Under theMinistry of Health of Ukraine”, Oncology and Medical Radiology, Municipal Non-Commercial Enterprise “City Clinical Hospital No. 4” of Dnipro City Council, Head and Neck Pathology DepartmentDnipro, Ukraine. [Stosic, Srboljub] Military Medical Academy, Clinic for Maxillofacial SurgeryBelgrade, Serbia. [Sobat, Hrvoje] University Hospital for Tumors, University Clinical Hospital Center “Sestre Milosrdnice”Zagreb, Croatia. [Zhukavets, Aliaksandr] N.N.Alexandrov National Cancer Centre of Belarus, Department of SurgeryMinsk, Belarus. [Imamovic, Nazim] University Clinical Center Tuzla, Clinic for Plastic and Maxillofacial SurgeryTuzla, Bosnia and Herzegovina. [Bankowska-Wozniak, Magdalena] Centrum Onkologii im Prof. F. Lukaszcsyka, Bydgoszcz, Department of SurgeryBydgoszcz, Poland. [Kisely, Mihaly] Markusovszky Egyetemi Oktatokorhaz, Altalanos Sebeszeti OsztalySzombathely, Hungary. [Jovic, Rajko] University Clinical Centre of Vojvodina, Clinic for Otorhinolaryngology and Head and Neck Surgery, Department of Laryngology, Oncology and PhoniatricsNovi Sad, Serbia. [Young, Edward Massey James] McMaster University, St Joseph Healthcare HamiltonHamilton, ON, Canada. RP Talor, E (reprint author), CEL-SCI Corporation, Vienna, USA. EM etalor@cel-sci.com CR Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2024, 74:229–63., DOI 10.3322/caac.21834 Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin, 2024, 74:12–49., DOI 10.3322/caac.21820 National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology, NCCN Guidelines). Head and Neck Cancers, 2023). Available online at: https://www.nccn.org/login?ReturnURL=https://www.nccn.org/professionals/ physician_gls/pdf/head-and-neck.pdf, Accessed April 20, 2023). Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, et al. SEER cancer statistics review, 1975-2017. Bethesda, MD: National Cancer Institute, 2020). Available online at: https://seer.cancer.gov/csr/1975_2017/,basedonNovember 2019SEERdatasubmission,postedtotheSEERwebsite April, 2020). Hanna EY, Mendoza TR, Rosenthal DI, Gunn GB, Sehra P, Yucel E, et al. The symptom burden of treatment-naive patients with head and neck cancer. Cancer, 2015, 121:766–73., DOI 10.1002/cncr.29097 Wissinger E, Griebsch I, Lungershausen J, Byrnes M, Travers K, Pashos CL. The humanistic burden of head and neck cancer: a systematic literature review. PharmacoEconomics, 2014, 32:1213–29., DOI 10.1007/s40273-014-0199-x Orland MD, Ullah F, Yilmaz E, Geiger JL. Immunotherapy for head and neck squamous cell carcinoma: present and future approaches and challenges. JCO Oncol Pract, 2024, 20:1588–95., DOI 10.1200/OP.24.00041 Boydell E, Sandoval JL, Michielin O, Obeid M, Addeo A, Friedlaender A. Neoadjuvant immunotherapy: a promising new standard of care. Int J Mol Sci, 2023, 24:11849., DOI 10.3390/ijms241411849 Vaidya P, Cohen EEW. Facts and hopes in neoadjuvant immunotherapy: current approvals and emerging evidence. Clin Cancer Re, 2024, 30:1232–9., DOI 10. 1158/1078-0432.CCR-23-0583 Zhao Y, Wucherpfennig KW. Neoadjuvant immune checkpoint blockade enhances local and systemic tumor immunity in head and neck cancer. Curr Opin Oncol, 2024, 36:136–42., DOI 10.1097/CCO.0000000000001023 Masarwy R, Kampel L, Horowitz G, GutfeldO,Muhanna N. Neoadjuvant PD-1/PDL1 inhibitors for resectable head and neck cancer: a systematic review and meta-analysis. JAMA Otolaryngol Head Neck Surg, 2021, 147:871–8., DOI 10.1001/jamaoto.2021.2191 NindraU,Hurwitz J, Forstner D, Chin V, Gallagher R, Lia J. A systematic review of neoadjuvant and definitive immunotherapy in locally advanced head and neck squamous cell carcinoma. Cancer Med, 2023, 12:11234–47., DOI 10.1002/cam4.5815 Cortesina G, De Stefani A, Galeazzi E, Cavallo GP, Jemma C, Giovarelli M, et al. Interleukin-2 injected around tumor-draining lymph nodes in head and neck cancer. Head Neck, 1991, 13:125–31., DOI 10.1002/hed.2880130208 Rivoltini L, Gambacorti-Passerini C, Squadrelli-Saraceno M, Grosso MI, Cantu G, Molinari R, et al. In vivo interleukin 2-induced activation of lymphokineactivated killer cells and tumor cytotoxic T-cells in cervical lymph nodes of patients with head and neck tumors. Cancer Res, 1990, 50:5551–7. Saito T, Kakiuti H, Kuki K, Yokota M, Jinnin T, Kimura T, et al. Clinical evaluation of local administration of rIL-2 in head and neck cancer. Nihon Jibiinkoka Gakkai Kaiho, 1989, 92:1265–70., DOI 10.3950/jibiinkoka.92.1265 Hadden JW, Endicott J, Baekey P, Skipper P, Hadden EM. Interleukins and contrasuppression induce immune regression of head and neck cancer. Arch Otolaryngol Head Neck Surg, 1994, 120(4):395–403., DOI 10.1001/archotol.1994. 01880280023004 Pulley M, Nagendran V, Edwards J, Dumonde D. Intravenous, intralesional and endolymphatic administration of lymphokines in human cancer. Lymphokine Res, 1986, 5:S157–63. Wolf GT, Liu S, Bellile E, Sartor M, Rozek L, Thomas D, et al. Tumor infiltrating lymphocytes after neoadjuvant IRX-2 immunotherapy in oral squamous cell carcinoma: interim findings from the INSPIRE trial. Oral Oncol, 2020, 111: 104928., DOI 10.1016/j.oraloncology.2020.104928 Park R, Li J, Slebos RJ, Chaudhary R, Poole MI, Ferraris C, et al. Phase Ib trial of IRX-2 plus durvalumab in patients with recurrent and/or metastatic head and neck squamous cell carcinoma. Oral Oncol, 2024, 154:106866., DOI 10.1016/j. oraloncology.2024.106866 Talor E. A method of pre-sensitizing cancer prior to treatment with radiation and/or chemotherapy and a novel cytokine mixture. In: US6896879B2, US patent, 2003, EP 1 773 368 B1, European patent,, 2016). Timar J, Forster-Horvath C, Lukits J, Dome B, Ladanyi A, Remenar E, et al. The effect of leukocyte interleukin injection, Multikine®, treatment on the peritumoral and intratumoral subpopulation of mononuclear cells and on tumor epithelia: a possible new approach to augmenting sensitivity to radiation therapy and chemotherapy in oral cancer—a multicenter phase I/II clinical trial. Laryngoscope, 2003, 113:2206–17., DOI 10.1097/00005537-200312000-00031 Timar J, Ladanyi A, Forster-Horvath C, Lukits J, Dome B, Remenar E, et al. Neoadjuvant immunotherapy of oral squamous cell carcinoma modulates intratumoral CD4/CD8 ratio and tumor microenvironment: a multicenter phase II clinical trial. J Clin Oncol, 2005, 23:3421–32., DOI 10.1200/JCO.2005.06.005 Feinmesser R, Hardy B, Sadov R, Shwartz A, Chretien P, Feinmesser M. Report of a clinical trial in 12 patients with head and neck cancer treated intratumorally and peritumorally with multikine. Arch Otolaryngol Head Neck Surg, 2003, 129:874–81., DOI 10.1001/archotol.129.8.874 Feinmesser M, Okon E, Schwartz A, Kaganovsky E, Hardy B, Aminov E, et al. Histologic and immunohistochemical characterization of tumor and inflammatory infiltrates in oral squamous cell carcinomas treated with local multikine immunotherapy: the macrophage at the front line. Eur Arch Otorhinolaryngol, 2004, 261:359–68., DOI 10.1007/s00405-003-0615-x Talor E,Remenar E, Zemplen B, Bencsik B,Huslar T, Balatoni Z, et al. O160Multikine therapy in phase II clinical trial: impact on survival and local regional control in OSCC patients. Oral Oncol Suppl, 2007, 1:115–6., DOI 10.1016/s1744-7895(07)70232-8 Waston L, Geczy A, Farrell C. A convenient and reliable LI-2 bioassya using frozen CTLL-2 to improve the detection of helper T lymphocytes precursors. Immunol Cel Biol, 1998, 76:190–2. WadhwaM, Bird C, HeathAB,Dilger P, Thorpe R, Participants of the collaborative study. The 2nd International standard for Interleukin-2, IL-2, Report of a collaborative study. J Immunological Methods, 2013, 397:1–7., DOI 10.1016/j.jim.2013.07.012 Mascarella MA, Olonisakin TF, Rumde P, Vendra V, Nance MA, Kim S, et al. Response to neoadjuvant targeted therapy in operable head and neck cancer confers survival benefit. Clin Cancer Res, 2023, 29:723–30., DOI 10.1158/1078-0432.CCR-22-1768 European Organisation for Research and Treatment of Cancer Quality of Life EORTC QLQ-C30, version 3,, 1995). Available online at: https://www.eortc.org/app/ uploads/sites/2/2018/08/Specimen-QLQ-C30-English.pdf, Accessed April 20, 2023). European organisation for research and treatment of cancer quality of life. EORTC QLQ - H&N35, 1994). Available online at: https://www.eortc.org/app/ uploads/sites/2/2018/08/Specimen-HN35-English.pdf, Accessed April 20, 2023). Murphy CT, Galloway TJ, Handorf EA, Egleston BL, Wang LS, Mehra R, et al. Survival impact of increasing time to treatment initiation for patients with head and neck cancer in the United States. J Clin Oncol, 2016, 34:169–78., DOI 10.1200/JCO.2015.61.5906 Liao DZ, Schlecht NF, Rosenblatt G, Kinkhabwala CM, Leonard JA, Ference RS, et al. Association of delayed time to treatment initiation with overall survival and recurrence among patients with head and neck squamous cell carcinoma in an underserved urban population. JAMA Otolaryngol Head Neck Surg, 2019, 145: 1001–9., DOI 10.1001/jamaoto.2019.2414 Graboyes EM, Kompelli AR, Neskey DM, Brennan E, Nguyen S, Sterba KR, et al. Association of treatment delays with survival for patients with head and neck cancer: a systematic review. JAMA Otolaryngol Head Neck Surg, 2019, 145:166–77., DOI 10.1001/jamaoto.2018.2716 Tsai WC, Kung PT, Wang YH, Huang KH, Liu SA. Influence of time interval from diagnosis to treatment on survival for oral cavity cancer: a nationwide cohort study. PLoS One, 2017, 12:e0175148., DOI 10.1371/journal.pone.0175148 Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, et al. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst, 2000, 92:205–16., DOI 10.1093/jnci/92.3.205 Hitt R, Paz-Ares L, Brandariz A, Castellano D, Pena C, Millan JM, et al. Induction chemotherapy with paclitaxel, cisplatin and 5-fluorouracil for squamous cell carcinoma of the head and neck: long-term results of a phase II trial. Ann Oncol, 2002, 13:1665–73., DOI 10.1093/annonc/mdf268 Monnerat C, Faivre S, Temam S, Bourhis J, Raymond E. End points for new agents in induction chemotherapy for locally advanced head and neck cancers. Ann Oncol, 2002, 13:995–1006., DOI 10.1093/annonc/mdf172 Liao CT,ChenHN,Wen YW, Lee SR,Ng SH, Liu TW, et al.Association between the diagnosis-to-treatment interval and overall survival in Taiwanese patients with oral cavity squamous cell carcinoma. Eur J Cancer, 2017, 72:226–34., DOI 10.1016/j.ejca.2016.11.010 Cooper JS, Pajak TF, Forastiere AA, Jacobs J, Campbell BH, Saxman SB, et al. Postoperative concurrent radiotherapy and chemotherapy for high-risk squamouscell carcinoma of the head and neck. N Engl J Med, 2004, 350:1937–44., DOI 10.1056/ NEJMoa032646 Bernier J, Domenge C, Ozsahin M, Matuszewska K, Lefebvre JL, Greiner RH, et al. Postoperative irradiation with or without concomitant chemotherapy for locally advanced head and neck cancer. N Engl J Med, 2004, 350:1945–52., DOI 10. 1056/NEJMoa032641 NR 1 PU Arányi Lajos Foundation PI Budapest PA Ulloi ut 26., Budapest H-1085, Hungary SN 1219-4956 J9 PAOR JI Pathol Oncol Res PD JAN PY 2025 VL 31 IS 1 BP 1612084 EP 1612100 DI 10.3389/pore.2025.1612084 PG 17 ER EF